Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 132
Filtrar
1.
Ann Oncol ; 29(8): 1853-1860, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29982279

RESUMEN

Background: Neoadjuvant anti-PD-1 may improve outcomes for patients with resectable NSCLC and provides a critical window for examining pathologic features associated with response. Resections showing major pathologic response to neoadjuvant therapy, defined as ≤10% residual viable tumor (RVT), may predict improved long-term patient outcome. However, %RVT calculations were developed in the context of chemotherapy (%cRVT). An immune-related %RVT (%irRVT) has yet to be developed. Patients and methods: The first trial of neoadjuvant anti-PD-1 (nivolumab, NCT02259621) was just reported. We analyzed hematoxylin and eosin-stained slides from the post-treatment resection specimens of the 20 patients with non-small-cell lung carcinoma who underwent definitive surgery. Pretreatment tumor biopsies and preresection radiographic 'tumor' measurements were also assessed. Results: We found that the regression bed (the area of immune-mediated tumor clearance) accounts for the previously noted discrepancy between CT imaging and pathologic assessment of residual tumor. The regression bed is characterized by (i) immune activation-dense tumor infiltrating lymphocytes with macrophages and tertiary lymphoid structures; (ii) massive tumor cell death-cholesterol clefts; and (iii) tissue repair-neovascularization and proliferative fibrosis (each feature enriched in major pathologic responders versus nonresponders, P < 0.05). This distinct constellation of histologic findings was not identified in any pretreatment specimens. Histopathologic features of the regression bed were used to develop 'Immune-Related Pathologic Response Criteria' (irPRC), and these criteria were shown to be reproducible amongst pathologists. Specifically, %irRVT had improved interobserver consistency compared with %cRVT [median per-case %RVT variability 5% (0%-29%) versus 10% (0%-58%), P = 0.007] and a twofold decrease in median standard deviation across pathologists within a sample (4.6 versus 2.2, P = 0.002). Conclusions: irPRC may be used to standardize pathologic assessment of immunotherapeutic efficacy. Long-term follow-up is needed to determine irPRC reliability as a surrogate for recurrence-free and overall survival.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/terapia , Neoplasias Pulmonares/terapia , Pulmón/patología , Adulto , Antineoplásicos Inmunológicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Carcinoma de Pulmón de Células no Pequeñas/patología , Estudios de Factibilidad , Humanos , Ipilimumab/farmacología , Ipilimumab/uso terapéutico , Pulmón/inmunología , Pulmón/cirugía , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Terapia Neoadyuvante/métodos , Neoplasia Residual , Nivolumab/farmacología , Nivolumab/uso terapéutico , Neumonectomía , Pronóstico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Reproducibilidad de los Resultados , Resultado del Tratamiento
2.
Mucosal Immunol ; 10(2): 421-433, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27301879

RESUMEN

Enterotoxigenic Bacteroides fragilis (ETBF), a human commensal and candidate pathogen in colorectal cancer (CRC), is a potent initiator of interleukin-17 (IL-17)-dependent colon tumorigenesis in MinApc+/- mice. We examined the role of IL-17 and ETBF on the differentiation of myeloid cells into myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages, which are known to promote tumorigenesis. The myeloid compartment associated with ETBF-induced colon tumorigenesis in Min mice was defined using flow cytometry and gene expression profiling. Cell-sorted immature myeloid cells were functionally assayed for inhibition of T-cell proliferation and inducible nitric oxide synthase expression to delineate MDSC populations. A comparison of ETBF infection with that of other oncogenic bacteria (Fusobacterium nucleatum or pks+Escherichia coli) revealed a specific, ETBF-associated colonic immune infiltrate. ETBF-triggered colon tumorigenesis is associated with an IL-17-driven myeloid signature characterized by subversion of steady-state myelopoiesis in favor of the generation of protumoral monocytic-MDSCs (MO-MDSCs). Combined action of the B. fragilis enterotoxin BFT and IL-17 on colonic epithelial cells promoted the differentiation of MO-MDSCs, which selectively upregulated Arg1 and Nos2, produced NO, and suppressed T-cell proliferation. Evidence of a pathogenic inflammatory signature in humans colonized with ETBF may allow for the identification of populations at risk for developing colon cancer.


Asunto(s)
Infecciones por Bacteroides/inmunología , Bacteroides fragilis/inmunología , Colon/microbiología , Neoplasias Colorrectales/inmunología , Células Epiteliales/inmunología , Células Supresoras de Origen Mieloide/inmunología , Linfocitos T/inmunología , Animales , Arginasa/genética , Arginasa/metabolismo , Toxinas Bacterianas/inmunología , Carcinogénesis , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Colon/inmunología , Colon/patología , Neoplasias Colorrectales/genética , Modelos Animales de Enfermedad , Genes APC , Humanos , Tolerancia Inmunológica , Interleucina-17/metabolismo , Metaloendopeptidasas/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Transcriptoma
3.
J Viral Hepat ; 15(3): 219-28, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18194173

RESUMEN

Studies assessing the function of monocyte derived dendritic cells (MD-DC) in individuals with hepatitis C virus (HCV) infection have shown conflicting results. Impaired MD-DC function in chronic HCV infection would have important implications both for understanding the pathogenesis of HCV infection and in the use of autologous MD-DC in vaccination strategies. We determined the allostimulatory capacity of MD-DC in the same patient before and after HCV infection. Next, the phenotype, cytokine production and allostimulatory function of immature and mature MD-DC in individuals with persistent HCV infection were compared directly with MD-DC from healthy individuals. Finally, we assessed the ability of MD-DC to prime autologous naïve peptide specific CD8+ T cells using HLA-A2 class-I tetramers. DCs retained the same allostimulatory capacity before and following the establishment of persistent HCV infection. The surface phenotype and the amount of interleukin (IL)-10 and IL-12(p70) produced during DC maturation did not differ between HCV-infected individuals and healthy controls. Mature DCs from HCV-infected individuals performed comparably in an allogeneic MLR compared with healthy individuals. Mature MD-DC from HCV-infected individuals stimulated the expansion of peptide specific naïve CD8+ T cells. MD-DC from HCV-infected and healthy individuals are phenotypically indistinguishable and perform comparably in functional assays.


Asunto(s)
Células Dendríticas/inmunología , Hepatitis C Crónica/inmunología , Adulto , Antígenos de Superficie/análisis , Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Células Cultivadas , Células Dendríticas/química , Femenino , Humanos , Interleucina-10/biosíntesis , Interleucina-12/biosíntesis , Activación de Linfocitos/inmunología , Masculino , Persona de Mediana Edad
4.
Gene Ther ; 13(1): 67-77, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16107858

RESUMEN

We have recently demonstrated that linkage of DNA-encoding calreticulin to DNA-encoding human papillomavirus-16 E7 antigen strongly enhances the efficacy of DNA vaccines against E7-expressing tumors in animal models. In this study, as a prelude to clinical translation, we characterized the ability of DNA-encoding calreticulin linked to DNA-encoding E7 antigen to generate HLA-A2-restricted E7-specific CD8(+) T-cell responses in HLA-A2 (AAD) transgenic mice, as well as antitumor effects against an E7(+) HLA-A2(+) tumor cell line, TC-1/A2. Our results show that while vaccination with CRT/E7 DNA generates strong H-2D(b)-restricted E7 (amino acid (aa)49-57)-specific CD8(+) T-cell immune responses in both C57BL/6 and HLA-A2 (AAD) transgenic mice, no such responses were generated to HLA-A2-restricted epitopes in either type of mouse. In contrast, vaccination with DNA-encoding calreticulin linked to DNA encoding a mutant version of E7 with a deleted aa49-57 epitope leads to the generation of an HLA-A2-restricted E7 (aa11-20)-specific CTL response in HLA-A2 (AAD) transgenic mice. More importantly, vaccination with CRT/mtE7 (del aa49-57) DNA protects against a lethal challenge with TC-1/A2 tumor cells in HLA-A2 (AAD) transgenic mice. Furthermore, our in vitro studies demonstrate that the presence of the E7 (aa49-57) epitope does not suppress presentation of the HLA-A2-restricted E7 (aa11-20) epitope through MHC class I molecules. Thus, the predominant E7 aa49-57-specific CD8+ T-cell immune response in HLA-A2 transgenic mice vaccinated with CRT/E7 is likely due to preferred expansion of E7 aa49-57-specific CD8(+) T cells in vaccinated mice. These results highlight the importance of epitope immunodominance in the evaluation of immune responses in HLA-A2 (AAD) transgenic mice.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/administración & dosificación , Terapia Genética/métodos , Antígeno HLA-A2/inmunología , Epítopos Inmunodominantes/análisis , Neoplasias/terapia , Vacunas contra Papillomavirus , Animales , Reacciones Antígeno-Anticuerpo , Calreticulina/genética , Vacunas contra el Cáncer/inmunología , Línea Celular , Epítopos de Linfocito T/análisis , Femenino , Ingeniería Genética , Antígeno HLA-A2/genética , Humanos , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias/inmunología , Proteínas Oncogénicas Virales , Proteínas E7 de Papillomavirus , Transfección , Vacunas de ADN/administración & dosificación , Vacunas de ADN/inmunología
6.
Pharm Res ; 18(7): 899-906, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11496947

RESUMEN

PURPOSE: The purpose of our study was to develop an injectable polymeric system for the long-term localized delivery of bioactive interleukin-2 for antitumor immunotherapy. METHODS: IL-2 was encapsulated into gelatin and chondroitin-6-sulfate using an aqueous-based complex coacervation. CTLL-2 cells were used to measure the bioactivity of released IL-2 and radiolabeled IL-2 was used for release studies in the rat brain and mouse liver. Antitumor efficacy studies were carried out in primary (9L gliosarcoma) and metastatic (B16-F10 melanoma) brain tumor models in rats and mice, respectively, as well as a murine liver tumor model (CT26 carcinoma). Survivors of the metastatic brain tumor challenge were rechallenged with tumor in the opposite lobe of the brain to confirm that antitumor immunologic memory had developed. RESULTS: Bioactive IL-2 was released for over 2 weeks in vitro and in vivo IL-2 release showed significant IL-2 levels for up to 21 days. Polymeric IL-2 microspheres injected intratumorally were statistically more effective in protecting animals challenged with fatal tumor doses in the brain and the liver than placebo or autologous tumor cells genetically engineered to secrete IL-2. Immunologic memory was induced following IL-2 microsphere therapy in the B16-F10 brain tumor model that was capable of protecting 42% of animals from a subsequent intracranial tumor challenge, suggesting that tumor destruction was mediated by the immune system. CONCLUSIONS: Local IL-2 therapy using novel polymeric carriers. aimed at stimulating long-lasting antitumor immunity, may provide an improved method of treating a variety of cancers.


Asunto(s)
Antineoplásicos/administración & dosificación , Materiales Biocompatibles/administración & dosificación , Neoplasias Encefálicas/tratamiento farmacológico , Sistemas de Liberación de Medicamentos/métodos , Interleucina-2/administración & dosificación , Neoplasias Hepáticas Experimentales/tratamiento farmacológico , Animales , Materiales Biocompatibles/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/patología , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/prevención & control , Neoplasias Hepáticas Experimentales/prevención & control , Melanoma Experimental , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microesferas , Polímeros/administración & dosificación , Polímeros/metabolismo , Ratas , Ratas Endogámicas F344 , Células Tumorales Cultivadas
7.
J Immunol ; 167(5): 2555-60, 2001 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-11509595

RESUMEN

Ag-specific immune tolerance in clinical organ transplantation is currently an unrealized but critical goal of transplant biology. The specificity and avidity of multimerized MHC-peptide complexes suggests their potential ability to modulate T cell sensitization and effector functions. In this study, we examined the ability of MHC-peptide dimers to modulate T cell function both in vitro and in vivo. Soluble MHC dimers induced modulation of surface TCR expression and inhibited T cell cytolytic activity at nanomolar concentrations in vitro. Furthermore, engagement of TCR by soluble dimers resulted in phosphorylation of the TCR zeta-chain and recruitment and phosphorylation of zeta-associated protein-70 to the signaling complex, the latter of which increased upon dimer cross-linking. Significantly, Ag-specific inhibition of an alloreactive TCR-transgenic T cell population in vivo resulted in consequent outgrowth of an allogeneic tumor. The prolonged Ag-specific suppression of expansion and/or effector function of cognate T cells in vivo suggests that soluble MHC dimers may be a means of inducing sustained Ag-specific T cell unresponsiveness in vivo.


Asunto(s)
Antígenos H-2/metabolismo , Linfocitos T Citotóxicos/inmunología , Animales , Citotoxicidad Inmunológica , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dimerización , Antígenos H-2/química , Humanos , Tolerancia Inmunológica , Técnicas In Vitro , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Transgénicos , Proteínas Nucleares , Proteínas Tirosina Quinasas/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Linfocitos T Citotóxicos/metabolismo , Inmunología del Trasplante , Proteína Tirosina Quinasa ZAP-70
9.
J Immunol ; 167(1): 147-55, 2001 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-11418643

RESUMEN

There is accumulating evidence that CD4(+) T cell responses are important in antitumor immunity. Accordingly, we generated CD4(+) T cells against the murine CT26 colon cancer. Three of three independent CT26-specific CD4(+) hybridomas were found to recognize the high m.w. precursor of the env gene product gp90. The CD4(+) response was completely tumor specific in that the same glycoprotein expressed by other tumors was not recognized by the CT26-specific hybridomas. The recognition of gp90 by the hybridomas was strictly dependent on the conformation of gp90. Different procedures that disrupted the conformation of the glycoprotein, such as disulfide bond reduction and thermal denaturation, completely abrogated recognition of gp90 by all three hybridomas. In CT26 cells, but not in other tumor cells tested, a large proportion of gp90 was retained in the endoplasmic reticulum, mostly bound to the endoplasmic reticulum chaperone, calreticulin. Although calreticulin was not essential for the stimulation of the gp90-specific hybridomas, most of the antigenic form of gp90 was bound to it. The antigenicity of gp90 correlated well with calreticulin binding, reflecting the fact that specificity of binding of calreticulin to its substrate required posttranslational modifications that were also necessary for the generation of this tumor-specific CD4(+) epitope.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Proteínas de Unión al Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Epítopos Inmunodominantes/metabolismo , Ribonucleoproteínas/metabolismo , Animales , Antígenos de Neoplasias/química , Linfocitos T CD4-Positivos/inmunología , Calreticulina , Retículo Endoplásmico/inmunología , Epítopos de Linfocito T/inmunología , Femenino , Productos del Gen env/metabolismo , Calor , Hibridomas/metabolismo , Virus de la Leucemia Murina/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Ratones Transgénicos , Unión Proteica/inmunología , Conformación Proteica , Desnaturalización Proteica , Células Tumorales Cultivadas
10.
J Exp Med ; 193(10): 1113-21, 2001 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-11369783

RESUMEN

Lymphoproliferative diseases are characterized by massive accumulation of CD4(-)CD8(-)B220(+) (double-negative [DN]) T cells in peripheral organs. Although evidence indicates these cells are derived from mature autoreactive alpha/beta T cells, the significance of coreceptor downregulation is not known. In this study, we examined the role CD4 coreceptor plays in the survival of repeatedly stimulated T cells. CD4(+/+) and CD4(-/-) T cells from AND T cell receptor (TCR) transgenic mice exhibited similar phenotypes after antigenic stimulation, but the CD4(-/-) T cells survived in much larger numbers than the CD4(+/+) cells upon primary and secondary major histocompatibility complex (MHC)/peptide stimulation. Enhanced survival of CD4(-/-) T cells was due to decreased apoptosis rather than enhanced proliferation. Similarly, circumvention of the CD4/MHC interaction by using a surrogate TCR ligand that does not engage CD4 led to significant enhancement of CD4(+/+) cells than when stimulated with MHC/peptide. Finally, we generated DN B220(+) T cells using an in vitro model system and showed they were more tolerant to chronic stimulation than CD4(+/+) cells. Together, these results indicate that coreceptor engagement controls expansion of normal T cells. In the absence of coreceptor, T cells survive chronic stimulation and express B220 as seen in autoimmune lymphoproliferative diseases.


Asunto(s)
Autoinmunidad/inmunología , Activación de Linfocitos/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta , Receptores Inmunológicos/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Apoptosis , Complejo CD3/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Supervivencia Celular , Antígenos Comunes de Leucocito/inmunología , Trastornos Linfoproliferativos/etiología , Ratones , Ratones Transgénicos , Subgrupos de Linfocitos T/citología
11.
J Exp Med ; 193(7): 839-46, 2001 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-11283156

RESUMEN

Dendritic cells (DCs), unique antigen-presenting cells (APCs) with potent T cell stimulatory capacity, direct the activation and differentiation of T cells by providing costimulatory signals. As such, they are critical regulators of both natural and vaccine-induced immune responses. A new B7 family member, B7-DC, whose expression is highly restricted to DCs, was identified among a library of genes differentially expressed between DCs and activated macrophages. B7-DC fails to bind the B7.1/2 receptors CD28 and cytotoxic T lymphocyte-associated antigen (CTLA)-4, but does bind PD-1, a receptor for B7-H1/PD-L1. B7-DC costimulates T cell proliferation more efficiently than B7.1 and induces a distinct pattern of lymphokine secretion. In particular, B7-DC strongly costimulates interferon gamma but not interleukin (IL)-4 or IL-10 production from isolated naive T cells. These properties of B7-DC may account for some of the unique activity of DCs, such as their ability to initiate potent T helper cell type 1 responses.


Asunto(s)
Presentación de Antígeno , Antígenos de Superficie , Antígeno B7-1/inmunología , Células Dendríticas/inmunología , Linfocitos T/inmunología , Secuencia de Aminoácidos , Animales , Antígenos CD , Proteínas Reguladoras de la Apoptosis , Antígenos CD28/metabolismo , Biblioteca de Genes , Humanos , Interferón gamma/metabolismo , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Proteína 2 Ligando de Muerte Celular Programada 1 , Receptor de Muerte Celular Programada 1 , Unión Proteica , Proteínas/metabolismo , Homología de Secuencia de Aminoácido
12.
Gene Ther ; 8(2): 128-38, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11313782

RESUMEN

Antigen-specific cancer immunotherapy involves the delivery of tumor-associated antigen to the host for the generation of tumor-specific immune responses and antitumor effects. We hypothesized that different delivery systems may influence the pattern of antigen-specific immune response and the outcome of antitumor effect. We therefore evaluated recombinant vaccinia virus and naked DNA for the generation of antigen-specific immune responses and antitumor effects. We previously found that recombinant vaccinia and naked DNA vaccines containing the chimeric Sig/E7/LAMP-1 gene were capable of controlling the growth of HPV-16 E7-expressing tumor cells (TC-1). In this study, we performed a head-to-head comparison of optimized delivery of Sig/E7/LAMP-1 vaccinia and DNA vaccines using dose-escalating tumor challenge. At a dose of 1 x 10(6) TC-1 cells per mouse, Sig/E7/LAMP-1 DNA provided 100% protection against subcutaneous growth of tumors, while Vac-Sig/E7/LAMP-1 protected only 40% of the mice. Furthermore, Sig/E7/LAMP-1 DNA vaccines are capable of protecting against challenge with a more stringent subclone of TC-1 (TC-1 P2) established from TC-1 tumors that survived initial Sig/E7/LAMP-1 vaccinia vaccination. Immunological assays revealed that both vaccines induced comparable levels of CD8(+) T cell precursors and anti-E7 antibody titers. Interestingly, Sig/E7/LAMP-1 vaccinia induced both E7-specific IFN-gamma- and IL4-secreting CD4(+) T cell precursors while Sig/E7/LAMP-1 DNA induced only E7-specific IFN-gamma-secreting CD4(+) T cell precursors. We also found that IL-4 knockout C57BL/6 mice vaccinated with Sig/E7/LAMP-1 vaccinia exhibited a more potent antitumor effect than vaccinated wild-type C57BL/6 mice in our tumor protection experiments. These results suggest that IL-4 may play a detrimental role in the antitumor effect mediated by vaccinia vaccines. Our findings suggested that DNA vaccines may provide better tumor protection than vaccinia vaccines employing the same gene, which may have implications in the future design of antigen-specific cancer immunotherapy.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias Pulmonares/prevención & control , Vacunas de ADN , Virus Vaccinia/inmunología , Vacunas Virales , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Femenino , Vectores Genéticos , Humanos , Interleucina-4/inmunología , Neoplasias Pulmonares/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Trasplante de Neoplasias , Células Tumorales Cultivadas
13.
Clin Cancer Res ; 7(3 Suppl): 838s-847s, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11300481

RESUMEN

The early genes E6 and E7 of human papillomavirus type 16 (HPV16) are consistently and exclusively expressed in HPV16-induced cancer lesions and play major roles in the development and maintenance of the malignant phenotype. Because this protein is a good example of a tumor-associated antigen, we have used E7 as a model antigen to test the potential of an experimental vaccine as an immunotherapeutic approach. In this study, we used a murine E7-expressing tumor model (TC1 cells) to assess effects of an E7-based vaccine on tumor growth. We show that vaccination with the E7 protein, formulated in the SmithKline Beecham Biologicals proprietary adjuvants (SBAS 1 and SBAS 2), leads to the rejection of pre-established tumors. Tumor rejection was associated with the induction of a strong systemic T helper 1 response, including CTLs, and the presence of an inflammatory infiltrate within the regressing tumor. Because most identified tumor-associated antigens are self antigens rather viral antigens, we used E7 transgenic mice to evaluate the E7-based vaccine in conditions where E7 is a self antigen. Transgenic mice, which constitutively and specifically express the E7 HPV16 gene in the thyroid epithelium, rapidly develop thyroid goiters and, after several months, thyroid carcinomas. We show that E7-specific antibodies and CD4 T helper responses can be obtained by vaccinating E7 transgenic mice, although a CTL response was not detected. Despite the absence of measurable CTL responses, vaccination still reduced the growth of pre-established TC1 tumors, although less efficiently than in nontransgenic animals, but was unable to suppress or delay the development of the spontaneous thyroid pathology.


Asunto(s)
Vacunas contra el Cáncer , Proteínas Oncogénicas Virales/biosíntesis , Vacunas contra Papillomavirus , Proteínas Recombinantes/metabolismo , Animales , Ensayo de Inmunoadsorción Enzimática , Epitelio/metabolismo , Inmunoglobulina G/metabolismo , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas E7 de Papillomavirus , Plásmidos/metabolismo , Linfocitos T Citotóxicos/metabolismo , Glándula Tiroides/metabolismo , Factores de Tiempo , Transgenes
14.
Cancer Res ; 61(9): 3682-8, 2001 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-11325839

RESUMEN

Activated macrophages play a central role in antitumor immunity. However, the stimuli that activate macrophages to kill tumor cells are not completely understood. Because the center of solid tumors can be hypoxic, we hypothesized that hypoxia may be an important signal in activating macrophages to kill tumor cells. Hypoxia stimulates IFN-primed macrophages to express the inducible nitric oxide synthase (NOS2) and to synthesize nitric oxide (NO). We show that this synergy between IFN and hypoxia is mediated by the direct interaction of the hypoxia inducible factor-1 (HIF-1) and IFN regulatory factor-1 (IRF-1), which are both required for the hypoxic transcription of NOS2. This interaction between HIF-1 and IRF-1 may explain the mechanism by which macrophages infiltrating into tumors are activated to express NOS2 and to produce NO, a mediator of tumor apoptosis.


Asunto(s)
Apoptosis/fisiología , Proteínas de Unión al ADN/fisiología , Interferón gamma/fisiología , Macrófagos/inmunología , Óxido Nítrico/fisiología , Proteínas Nucleares/fisiología , Fosfoproteínas/fisiología , Transducción de Señal/fisiología , Animales , Sitios de Unión , Hipoxia de la Célula/fisiología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Inducción Enzimática , Femenino , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Factor 1 Regulador del Interferón , Interferón gamma/farmacología , Activación de Macrófagos/fisiología , Macrófagos/enzimología , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa/biosíntesis , Óxido Nítrico Sintasa/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Saccharomyces cerevisiae/genética , Factores de Transcripción/fisiología , Activación Transcripcional , Transfección
15.
Vaccine ; 19(17-19): 2583-9, 2001 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-11257396

RESUMEN

Over 90% of cervical cancers are associated with HPV infection, the commonest being the HPV-16 subtype. Two early viral genes, E6 and 7, play major roles in the development and maintenance of the malignant phenotype. The vaccine potential of a recombinant HPV16 E7 protein was examined in two murine models of E7-expressing tumours. Formulations including the immunostimulants MPL and QS21 induced therapeutically active immune responses leading to regression of pre-established TC1 tumour lesions, associated with induction of IgG antibodies, lymphoproliferation and CTL. Our data provide a clear incentive to investigate the clinical application of this approach in cancer immunotherapy.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Papillomaviridae , Infecciones por Papillomavirus/terapia , Infecciones Tumorales por Virus/terapia , Neoplasias del Cuello Uterino/terapia , Vacunas Virales/uso terapéutico , Adyuvantes Inmunológicos/uso terapéutico , Animales , Anticuerpos Antineoplásicos/biosíntesis , Anticuerpos Antivirales/biosíntesis , Vacunas contra el Cáncer/genética , Femenino , Inmunoglobulina G/biosíntesis , Técnicas In Vitro , Neoplasias Pulmonares/secundario , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/inmunología , Proteínas E7 de Papillomavirus , Infecciones por Papillomavirus/inmunología , Infecciones por Papillomavirus/patología , Linfocitos T Citotóxicos/inmunología , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/patología , Neoplasias del Cuello Uterino/inmunología , Neoplasias del Cuello Uterino/patología , Vacunas Sintéticas/genética , Vacunas Sintéticas/uso terapéutico , Vacunas Virales/genética
16.
J Clin Oncol ; 19(1): 145-56, 2001 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-11134207

RESUMEN

PURPOSE: Allogeneic granulocyte-macrophage colony-stimulating factor (GM-CSF)-secreting tumor vaccines can cure established tumors in the mouse, but their efficacy against human tumors is uncertain. We have developed a novel GM-CSF-secreting pancreatic tumor vaccine. To determine its safety and ability to induce antitumor immune responses, we conducted a phase I trial in patients with surgically resected adenocarcinoma of the pancreas. PATIENTS AND METHODS: Fourteen patients with stage 1, 2, or 3 pancreatic adenocarcinoma were enrolled. Eight weeks after pancreaticoduodenectomy, three patients received 1 x 10(7) vaccine cells, three patients received 5 x 10(7) vaccine cells, three patients received 10 x 10(7) vaccine cells, and five patients received 50 x 10(7) vaccine cells. Twelve of 14 patients then went on to receive a 6-month course of adjuvant radiation and chemotherapy. One month after completing adjuvant treatment, six patients still in remission received up to three additional monthly vaccinations with the same vaccine dose that they had received originally. RESULTS: No dose-limiting toxicities were encountered. Vaccination induced increased delayed-type hypersensitivity (DTH) responses to autologous tumor cells in three patients who had received >or= 10 x 10(7) vaccine cells. These three patients also seemed to have had an increased disease-free survival time, remaining disease-free at least 25 months after diagnosis. CONCLUSION: Allogeneic GM-CSF-secreting tumor vaccines are safe in patients with pancreatic adenocarcinoma. This vaccine approach seems to induce dose-dependent systemic antitumor immunity as measured by increased postvaccination DTH responses against autologous tumors. Further clinical evaluation of this approach in patients with pancreatic cancer is warranted.


Asunto(s)
Adenocarcinoma/terapia , Vacunas contra el Cáncer/uso terapéutico , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Neoplasias Pancreáticas/terapia , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Anciano , Vacunas contra el Cáncer/efectos adversos , Vacunas contra el Cáncer/farmacocinética , Terapia Combinada , Seguridad de Productos para el Consumidor , Supervivencia sin Enfermedad , Relación Dosis-Respuesta Inmunológica , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/efectos adversos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacocinética , Humanos , Hipersensibilidad Tardía/patología , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología
17.
Immunity ; 13(4): 529-38, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11070171

RESUMEN

T cell responsiveness to an epitope is affected both by its affinity for the presenting MHC molecule and the affinity of the MHC-peptide complex for TCR. One limitation of cancer immunotherapy is that natural tumor antigens elicit relatively weak T cell responses, in part because high-affinity T cells are rendered tolerant to these antigens. We report here that amino acid substitutions in a natural MHC class I-restricted tumor antigen that increase the stability of the MHC-peptide-TCR complex are significantly more potent as tumor vaccines. The improved immunity results from enhanced in vivo expansion of T cells specific for the natural tumor epitope. These results indicate peptides that stabilize the MHC-peptide-TCR complex may provide superior antitumor immunity through enhanced stimulation of specific T cells.


Asunto(s)
Antígenos de Neoplasias/inmunología , Neoplasias Colorrectales/inmunología , Epítopos de Linfocito T/inmunología , Antígenos H-2/metabolismo , Oligopéptidos/inmunología , Oligopéptidos/metabolismo , Complejo Receptor-CD3 del Antígeno de Linfocito T/metabolismo , Alanina/metabolismo , Sustitución de Aminoácidos , Animales , Antígenos de Neoplasias/metabolismo , Células Clonales , Citotoxicidad Inmunológica/inmunología , Epítopos de Linfocito T/metabolismo , Femenino , Antígeno de Histocompatibilidad H-2D , Ligandos , Ratones , Ratones Endogámicos BALB C , Oligopéptidos/síntesis química , Resonancia por Plasmón de Superficie , Subgrupos de Linfocitos T/inmunología
18.
J Immunother ; 23(4): 438-48, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10916753

RESUMEN

To explore the potential efficacy of local cytokine delivery against tumors in the central nervous system (CNS), C57BL6 mice were simultaneously given intracranial injections of tumor challenge and of irradiated B16F10 melanoma cells transduced to secrete interleukin-2 (IL-2). Intracranial IL-2 therapy generated antitumor responses capable of extending the survival of animals that received simultaneous intracranial tumor challenge either locally or at distant sites in the brain. Nontransduced melanoma cells had little effect. Animals that survived intracranial IL-2 therapy and tumor challenge showed prolonged survival compared with controls when challenged with a second tumor dose 70 days after initial treatment. In addition, animals that rejected intracranial tumors were also protected from tumor growth upon rechallenge at sites outside the CNS (i.e., subcutaneous tumor challenge). Conversely, identical or 10-fold larger doses of IL-2-transduced cells administered by subcutaneous injection failed to generate protection against intracranial tumor challenges. Elimination of T-cell and natural killer (NK) subsets using gene knockout mice and antibody-depletion techniques demonstrated that NK cells were most important for the initial antitumor response, whereas CD4+ T-cells were not necessary. These studies demonstrate that local IL-2 therapy in the brain not only generates an immediate local antitumor immune response, but also establishes long-term immunologic memory capable of eliminating subsequent tumor challenges within and outside of the CNS. Furthermore, the antitumor response to paracrine IL-2 in the brain differed significantly from that in the flank, suggesting that the intrinsic CNS cells involved in initiating immunity within the brain have different cytokine requirements from their peripheral counterparts.


Asunto(s)
Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/terapia , Interleucina-2/administración & dosificación , Interleucina-2/inmunología , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/genética , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Antígenos CD4/genética , Antígenos CD4/inmunología , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/uso terapéutico , Femenino , Memoria Inmunológica , Inyecciones Intralesiones , Inyecciones Subcutáneas , Interleucina-2/genética , Células Asesinas Naturales/inmunología , Depleción Linfocítica , Melanoma Experimental/genética , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Comunicación Paracrina , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/terapia , Subgrupos de Linfocitos T/inmunología , Transducción Genética
19.
J Hepatol ; 33(1): 91-8, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10905591

RESUMEN

BACKGROUND/AIMS: We have previously reported a recombinant vaccinia-based vaccine (vac-Sig/E7/LAMP-1) that demonstrated a significant anti-tumor effect in a subcutaneous tumor challenge model. Since the liver is one of the most common sites for tumor metastasis and organ microenvironments may modulate tumor cell responses to therapies, the aim of the present study was to evaluate the potency of vac-Sig/E7/LAMP-1 in treating E7-expressing tumors grown in the liver. METHODS: For in vivo tumor prevention experiments, mice were vaccinated intraperitoneally with vac-Sig/E7/LAMP-1 followed by intrahepatic tumor challenge. For in vivo tumor regression experiments, mice were first challenged with tumor cells and then vaccinated with vac-Sig/E7/LAMP-1 intraperitoneally. In addition, enzyme-linked immunospot assays were used to determine the frequency of E7-specific T cell precursors. RESULTS: For in vivo tumor protection experiments, tumor growth was observed in all of the mice vaccinated with wild-type vaccinia and 60% of the mice vaccinated with wild-type E7 vaccinia. All of the mice vaccinated with vac-Sig/E7/LAMP-1 remained tumor-free 30 days after tumor challenge. For the tumor regression assays, all of the mice vaccinated with vac-Sig/E7/LAMP-1 remained tumor-free 30 days after vaccination. In contrast, all of those mice receiving culture medium, wild-type vaccinia, or wild-type E7 vaccinia developed tumors in the liver. In addition, mice vaccinated with vac-Sig/E7/LAMP-1 had the highest E7-specific CD8+ T cell precursors. CONCLUSIONS: Our data suggest that vac-Sig/E7/LAMP-1 is an effective vaccine for controlling E7-expressing tumors grown in the liver and our model suggests that antigen-specific immunotherapy may represent a powerful tool for treating liver tumors with characterized tumor-specific antigens. In addition, our data indicate that the number of E7-specific CD8+ T cell precursors directly correlated with the anti-tumor effect generated by Sig/E7/LAMP-1 vaccinia.


Asunto(s)
Inmunoterapia , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/terapia , Proteínas Oncogénicas Virales/análisis , Proteínas Oncogénicas Virales/inmunología , Animales , Formación de Anticuerpos , Antígenos CD/inmunología , División Celular , Epítopos , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/prevención & control , Proteínas de Membrana de los Lisosomas , Glicoproteínas de Membrana/inmunología , Ratones , Trasplante de Neoplasias , Proteínas E7 de Papillomavirus , Células Tumorales Cultivadas , Vacunación , Vacunas Sintéticas/uso terapéutico
20.
Int J Cancer ; 86(5): 725-30, 2000 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-10797297

RESUMEN

Granulocyte-macrophage colony-stimulating factor (GM-CSF)-transduced autologous tumor cell-based vaccines are currently one of the major forms of cancer vaccines. However, the preparation of GM-CSF-transduced autologous tumor vaccines is time-consuming and technically challenging. In addition, the host antigen presenting cells, rather than the tumor vaccine cells themselves, present tumor-specific antigens and prime the host T cells. Therefore, we tested the efficacy of antigen-specific allogeneic tumor vaccines. We used human papillomavirus 16 (HPV-16) E7 protein as a model tumor antigen, which is associated with the development of most cervical carcinoma. B16, a C57BL/6 (H-2(b)) derived melanoma cell line, was genetically engineered to produce GM-CSF alone (B16GM), HPV-16 E7 alone (B16E7), or both (B16GME7). These vaccine cells were injected into BALB/c (H-2(d)) mice (10(6) cells/mouse). Two weeks later, mice were challenged with 10(5) live HPV-16 E7(+) BL-1 (H-2(d)) tumor cells and monitored for tumor progression twice weekly. To determine the effective cell population in the antitumor immunity elicited by B16GME7, we carried out in vivo antibody depletion experiments using CD4 and CD8 specific antibodies. In addition, as a measure of the immune responses produced by B16GME7, we performed an in vitro cytotoxic T lymphocyte assay using a standard chromium release method. We found that all of the mice vaccinated with B16GME7 remained tumor free 49 days post-BL-1 challenge. In contrast, mice vaccinated with B16GM and B16E7 did not show any tumor protection against a similar dose of BL-1 cells. Furthermore, the antitumor immunity produced by B16GME7 was dependent on both CD4 and CD8 T cells. In addition, E7-specific cytotoxic T lymphocyte activity could be readily demonstrated in mice immunized with B16GME7. These results suggest that allogeneic tumor cells transduced with GM-CSF and the tumor antigen, HPV-16 E7, cannot only generate an E7-specific cytotoxic T lymphocytes response in vitro, but can also elicit a potent antitumor immune response against an E7 expressing tumor in vivo.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Neoplasias Experimentales/terapia , Proteínas Oncogénicas Virales/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/administración & dosificación , Modelos Animales de Enfermedad , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Inmunoterapia , Masculino , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/mortalidad , Proteínas Oncogénicas Virales/biosíntesis , Proteínas Oncogénicas Virales/genética , Proteínas E7 de Papillomavirus , Linfocitos T Citotóxicos/inmunología , Transducción Genética , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA