Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Int J Cancer ; 151(1): 107-119, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35179790

RESUMEN

Lynch Syndrome (LS) is an autosomal dominant genetic condition that causes a high risk of colorectal cancer. The hallmark of LS is genetic instability as a result of mismatch repair (MMR) deficiency, particularly in repetitive low complexity regions called microsatellites (MS). MLH1-/- mice deficient in MMR are prone to developing tumors in the colon, upon oral administration of dextran sodium sulfate (DSS), at a rate of more than 70%. Using this LS mouse model, we found a novel tumor neo-antigen from a deletion mutation of the coding MS in the SENP6 gene that prevented tumorigenesis or hindered tumor growth rate in immunized mice. This was accomplished via high throughput exome sequencing of DSS-induced colorectal tumors in the MLH1-/- mice and predicting the most highly immunogenic mutant gene products processed and presented as antigens in C57BL/6 MHC-I molecules. Throughout our study, we were able to prove the validity of the vaccine by analyzing the colorectal tumors in immunized DSS-treated mice using either our epitope, called Sp6D1, or an unrelated peptide as a negative control. Tumors developed in this context were found to be antigenic and Sp6D1-specific CD8+ tumor infiltrating lymphocytes were detected by flow cytometry and cytotoxic T lymphocytes (CTL) killing assays. Additionally, immunohistochemistry showed that tumor-adjacent tertiary lymphoid organs were a potentially significant source of CD8+ lymphocytes. Altogether, our results indicate that there may be a protective effect to patients carrying LS mutations through the induction of a peptide-specific CTL response from the use of neoepitope vaccination.


Asunto(s)
Neoplasias Colorrectales Hereditarias sin Poliposis , Neoplasias Colorrectales , Vacunas , Animales , Antígenos de Neoplasias/genética , Neoplasias Encefálicas , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/prevención & control , Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Neoplasias Colorrectales Hereditarias sin Poliposis/prevención & control , Cisteína Endopeptidasas/genética , Reparación de la Incompatibilidad de ADN/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Inestabilidad de Microsatélites , Homólogo 1 de la Proteína MutL/genética , Síndromes Neoplásicos Hereditarios
2.
Eur J Immunol ; 51(6): 1505-1518, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33835499

RESUMEN

A T-cell receptor (TCR) with optimal avidity to a tumor antigen can be used to redirect T cells to eradicate cancer cells via adoptive cell transfer. Cancer testis antigens (CTAs) are attractive targets because they are expressed in the testis, which is immune-privileged, and in the tumor. However, CTAs are self-antigens and natural TCRs to CTAs have low affinity/avidity due to central tolerance. We previously described a method of directed evolution of TCR avidity using somatic hypermutation. In this study, we made several improvements to this method and enhanced the avidity of the hT27 TCR, which is specific for the cancer testis antigen HLA-A2-MAGE-A1278-286 . We identified eight point mutations with varying degrees of improved avidity. Human T cells transduced with TCRs containing these mutations displayed enhanced tetramer binding, IFN-γ and IL2 production, and cytotoxicity. Most of the mutations have retained specificity, except for one mutant with extremely high avidity. We demonstrate that somatic hypermutation is capable of optimizing avidity of clinically relevant TCRs for immunotherapy.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Inmunoterapia Adoptiva/métodos , Proteínas de Neoplasias/inmunología , Fragmentos de Péptidos/inmunología , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/inmunología , Células Cultivadas , Tolerancia Central , Citotoxicidad Inmunológica , Antígeno HLA-A2/metabolismo , Humanos , Interferón gamma/metabolismo , Activación de Linfocitos , Mutación Puntual/genética , Unión Proteica , Receptores de Antígenos de Linfocitos T/metabolismo , Hipermutación Somática de Inmunoglobulina , Linfocitos T/trasplante
3.
Int J Cancer ; 145(10): 2816-2826, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31381134

RESUMEN

Adoptive transfer of T cells that have been genetically modified to express an antitumor T-cell receptor (TCR) is a potent immunotherapy, but only if TCR avidity is sufficiently high. Endogenous TCRs specific to shared (self) tumor-associated antigens (TAAs) have low affinity due to central tolerance. Therefore, for effective therapy, anti-TAA TCRs with higher and optimal avidity must be generated. Here, we describe a new in vitro system for directed evolution of TCR avidity using somatic hypermutation (SHM), a mechanism used in nature by B cells for antibody optimization. We identified 44 point mutations to the Pmel-1 TCR, specific for the H-2Db -gp10025-33 melanoma antigen. Primary T cells transduced with TCRs containing two or three of these mutations had enhanced activity in vitro. Furthermore, the triple-mutant TCR improved in vivo therapy of tumor-bearing mice, which exhibited improved survival, smaller tumors and delayed or no relapse. TCR avidity maturation by SHM may be an effective strategy to improve cancer immunotherapy.


Asunto(s)
Evolución Molecular Dirigida/métodos , Melanoma Experimental/terapia , Receptores de Antígenos de Linfocitos T/genética , Neoplasias Cutáneas/terapia , Antígeno gp100 del Melanoma/inmunología , Animales , Línea Celular Tumoral , Células HEK293 , Antígeno de Histocompatibilidad H-2D , Humanos , Inmunoterapia Adoptiva/métodos , Melanoma Experimental/inmunología , Ratones , Ratones Transgénicos , Mutagénesis Sitio-Dirigida/métodos , Péptidos/inmunología , Mutación Puntual , Prueba de Estudio Conceptual , Receptores de Antígenos de Linfocitos T/inmunología , Neoplasias Cutáneas/inmunología
4.
Int J Cancer ; 144(4): 909-921, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30106470

RESUMEN

For many years, clinicians and scientists attempt to develop methods to stimulate the immune system to target malignant cells. Recent data suggest that effective cancer vaccination requires combination immunotherapies to overcome tumor immune evasion. Through presentation of both MHC-I and II molecules, DCs-based vaccine platforms are effective in generating detectable CD4 and CD8 T cell responses against tumor-associated antigens. Several platforms include DC transfection with mRNA of the desired tumor antigen. These DCs are then delivered to the host and elicit an immune response against the antigen of interest. We have recently established an mRNA genetic platform which induced specific CD8+ cytotoxic T cell response by DC vaccination against melanoma. In our study, an MHC-II mRNA DCs vaccine platform was developed to activate CD4+ T cells and to enhance the anti-tumor response. The invariant chain (Ii) was modified and the semi-peptide CLIP was replaced with an MHC-II binding peptide sequences of melanoma antigens. These chimeric MHC-II constructs are presented by DCs and induce proliferation of tumor specific CD4+ T cells. When administered in combination with the MHC-I platform into tumor bearing mice, these constructs were able to inhibit tumor growth, and improve mouse survival. Deciphering the immunological mechanism of action, we observed an efficient CTLs killing in addition to higher levels of Th1 and Th2 subsets in the groups immunized with a combination of the MHC-I and MHC-II constructs. These universal constructs can be applied in multiple combinations and offer an attractive opportunity to improve cancer treatment.


Asunto(s)
Antígenos de Diferenciación de Linfocitos B/inmunología , Vacunas contra el Cáncer/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Melanoma Experimental/inmunología , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Vacunas contra el Cáncer/administración & dosificación , Línea Celular Tumoral , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Progresión de la Enfermedad , Inmunoterapia/métodos , Melanoma Experimental/genética , Melanoma Experimental/terapia , Ratones Endogámicos C57BL , Ratones Transgénicos , Análisis de Supervivencia , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo
5.
Immunol Cell Biol ; 96(3): 284-297, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29356071

RESUMEN

Inflammation plays pivotal roles in different stages of tumor development. Screening for predisposing genetic abnormalities and understanding the roles these genes play in the crosstalk between immune and cancer cells will provide new targets for cancer therapy and prevention. The interferon inducible transmembrane (IFITM) genes are involved in pathogenesis of the gastro-intestinal tract. We aimed at delineating the role of IFITM3 in colonic epithelial homeostasis, inflammation and colitis-associated tumorigenesis using IFITM3-deficient mice. Chemical induction of colitis in IFITM3-deficient mice results in significantly increased clinical signs of inflammation and induction of invasive tumorigenesis. Bone marrow transplantation showed that cells of the hematopoietic system are responsible for colitis deterioration. In these mice, impaired cytokine expression skewed inflammatory response toward pathogenic Th17 with reduced expression of the anti-inflammatory cytokine IL10 during the recovery phase. Intriguingly, mice lacking the entire IFITM locus developed spontaneous chronic colitis from the age of 14 weeks. Sequencing the 16S rRNA of naïve mice lacking IFITM3 gene, or the entire locus containing five IFITM genes, revealed these mice had significant bacterial differences from their wild-type littermates. Our novel results provide strong evidence for the essential role of IFITM genes in ameliorating colitis and colitis-associated tumorigenesis.


Asunto(s)
Carcinogénesis/genética , Carcinogénesis/patología , Colitis/inmunología , Colitis/microbiología , Inmunidad , Inflamación/genética , Proteínas de la Membrana/genética , Microbiota , Animales , Colitis/genética , Colitis/patología , Colon/patología , Sulfato de Dextran , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Susceptibilidad a Enfermedades , Hematopoyesis , Inmunidad/genética , Proteínas de la Membrana/deficiencia , Ratones Endogámicos C57BL , Ratones Noqueados , Microbiota/genética , Células Mieloides/patología
6.
Mol Ther ; 23(8): 1391-1400, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25997427

RESUMEN

Recently, we have developed a novel genetic platform for improving dendritic cell (DC) induction of peptide-specific CD8 T cells, based on membrane-anchored ß2-microglobulin (ß2m) linked to a selected antigenic peptide at its N-terminus and to the cytosolic domain of toll-like receptor (TLR)4 C-terminally. In vitro transcribed mRNA transfection of antigen presenting cells resulted in polypeptides that efficiently coupled peptide presentation to cellular activation. In the present study, we evaluated the immunogenicity of such constructs in mRNA-transfected immature murine bone marrow-derived DCs. We show that the encoded peptide ß2m-TLR4 products were expressed at the cell surface up to 72 hours and stimulated the maturation of DCs. In vivo, these DCs prompted efficient peptide-specific T-cell activation and target cell killing which were superior to those induced by peptide-loaded, LPS-stimulated DCs. This superiority was also evident in the ability to protect mice from tumor progression following the administration of B16F10.9 melanoma cells and to inhibit the development of pre-established B16F10.9 tumors. Our results provide evidence that the products of two recombinant genes encoding for peptide-hß2m-TLR4 and peptide-hß2m-K(b) expressed from exogenous mRNA can cooperate to couple Major Histocompatibility Complex (MHC-I) peptide presentation to TLR-mediated signaling, offering a safe, economical and highly versatile genetic platform for a novel category of CTL-inducing vaccines.


Asunto(s)
Células Dendríticas/citología , Antígenos de Histocompatibilidad Clase I/metabolismo , Péptidos/química , ARN Mensajero/metabolismo , Receptor Toll-Like 4/metabolismo , Microglobulina beta-2/metabolismo , Animales , Presentación de Antígeno/inmunología , Células Presentadoras de Antígenos/citología , Antígenos de Neoplasias/química , Antineoplásicos/química , Células de la Médula Ósea/citología , Vacunas contra el Cáncer/inmunología , Membrana Celular/metabolismo , Clonación Molecular , Medios de Cultivo , Citosol/metabolismo , Femenino , Humanos , Lipopolisacáridos/química , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Estructura Terciaria de Proteína , Transducción de Señal
7.
Cancer Immunol Immunother ; 63(4): 369-80, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24452202

RESUMEN

Cryoablation is a low-invasive surgical procedure for management of malignant tumors. Tissue destruction is obtained by repeated deep freezing and thawing and results in coagulative necrosis and in apoptosis. This procedure induces the release of tumor-associated antigens and proinflammatory factors into the microenvironment. Local administration of immature dendritic cells (DCs) potentiates the immune response induced by cryoablation. To further augment the induction of long-lasting antitumor immunity, we investigated the clinical value of combining cryoimmunotherapy consisting of cryoablation and inoculation of immature DCs with administration of the immune adjuvant, CpG oligodeoxynucleotides. Injection of the murine Lewis lung carcinoma, D122-luc-5.5 that expresses the luciferase gene, results in spontaneous metastases, which can be easily monitored in vivo. The local tumor was treated by the combined treatment. The clinical outcome was assessed by monitoring tumor growth, metastasis in distant organs, overall survival, and protection from tumor recurrence. The nature of the induced T cell responses was analyzed. Combined cryoimmunotherapy results in reduced tumor growth, low metastasis and significantly prolonged survival. Moreover, this treatment induces antitumor memory that protected mice from rechallenge. The underlying suggested mechanisms are the generation of tumor-specific type 1 T cell responses, subsequent induction of cytotoxic T lymphocytes, and generation of systemic memory. Our data highlight the combined cryoimmunotherapy as a novel antitumor vaccine with promising preclinical results. Adjustment of this technique into practice will provide the therapeutic benefits of both, ablation of the primary tumor and induction of robust antitumor and antimetastatic immunity.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Carcinoma Pulmonar de Lewis/terapia , Islas de CpG , Criocirugía/métodos , Células Dendríticas/inmunología , Inmunoterapia/métodos , Neoplasias Pulmonares/terapia , Oligodesoxirribonucleótidos/uso terapéutico , Animales , Linfocitos T CD8-positivos/inmunología , Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/secundario , Carcinoma Pulmonar de Lewis/cirugía , Células Cultivadas , Terapia Combinada , Células Dendríticas/trasplante , Pie , Memoria Inmunológica , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/cirugía , Masculino , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia , Oligodesoxirribonucleótidos/administración & dosificación , Recurrencia , Organismos Libres de Patógenos Específicos , Subgrupos de Linfocitos T/inmunología , Linfocitos T Citotóxicos/inmunología
8.
Int Immunol ; 23(7): 453-61, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21652516

RESUMEN

Priming of naive CD8 T cells by dendritic cells (DCs) entails both effective antigen presentation on MHC class I products and co-stimulatory signaling. Their optimal coupling is a major goal in the development of CTL-inducing vaccines. We recently reported that a membranal derivative of the invariant MHC-I light chain, ß(2)-microglobulin (ß(2)m), markedly stabilizes MHC-I molecules and can serve as a universal platform for exceptional presentation of genetically linked peptides. To test whether it is possible to equip the resulting MHC-I complexes with an inherent ability to activate antigen-presenting cells, we engrafted the intracellular Toll/IL-1 receptor domain of mouse Toll-like receptor (TLR) 4 or TLR2 onto the peptide-ß(2)m scaffold. We evaluated the level of peptide presentation and status of cell activation conferred by such constructs in stably transfected mouse RAW264.7 macrophages and mRNA-transfected mouse DC2.4 DCs. We show that the encoded peptide-ß(2)m-TLR polypeptides are expressed at the cell surface, pair with endogenous heavy chains, stabilize MHC-I products, prompt efficient peptide-specific T-cell recognition and confer a constitutively activated phenotype on the transfected cells, as judged by the up-regulation of pro-inflammatory genes and surface co-stimulatory molecules. Our results provide evidence that the product of a single recombinant gene can couple MHC peptide presentation to TLR-mediated signaling and offer a safe, economical and highly versatile modality for a novel category of genetic CTL-inducing vaccines.


Asunto(s)
Presentación de Antígeno/inmunología , Células Presentadoras de Antígenos/inmunología , Antígenos H-2/inmunología , Péptidos/inmunología , Animales , Vectores Genéticos/genética , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Péptidos/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología , Transfección , Microglobulina beta-2/genética , Microglobulina beta-2/inmunología
9.
Int J Cancer ; 125(12): 2810-9, 2009 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19544527

RESUMEN

The human 1-8 interferon inducible gene family consists of at least 3 functional genes; 9-27, 1-8D and 1-8U, which are all linked on an 18-kb fragment of chromosome 11 and are highly homologous. It has recently been shown by us and others that the 1-8D gene is overexpressed in colon carcinoma. Here, we show, by sequence comparison of the 1-8D in pairs of tumor/normal colon tissues, the existence of 6 different alleles, containing single-nucleotide polymorphisms with no mutations. Transformation assays revealed a possible role for the 1-8D gene as a transformation inhibitor. Further, transient expression of the human 1-8D gene in multiple mammalian cell lines showed accumulation of cells in the G1 phase followed by elevation in the subG1 phase. SubG1 elevation was confirmed as apoptosis by Annexin-V binding assays and transferase-mediated dUTP nick end labeling assays. Moreover, knock-down of 1-8D provided partial protection from Etoposide and UV-induced apoptosis. The induction of apoptosis by 1-8D is dependent on caspase activities but not on p53 expression. Although 1-8D induces apoptosis independently of p53, p53 expression downregulates 1-8D protein expression. Our data suggest a role for the 1-8D gene as a novel pro-apoptotic gene that will provide new insights into the regulated cellular pathways to death.


Asunto(s)
Apoptosis/genética , Transformación Celular Neoplásica , Proteínas de la Membrana/genética , Polimorfismo de Nucleótido Simple/genética , Proteína p53 Supresora de Tumor/metabolismo , Secuencia de Aminoácidos , Animales , Anexina A5/metabolismo , Western Blotting , Estudios de Casos y Controles , Caspasas/metabolismo , Línea Celular Tumoral , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Fibroblastos/metabolismo , Fase G1 , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , Ratones , Datos de Secuencia Molecular , Mutación/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas F344 , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Proteína p53 Supresora de Tumor/genética
10.
Oncoimmunology ; 7(7): e1445457, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29900058

RESUMEN

Despite melanoma immunogenicity and remarkable therapeutic effects of negative immune checkpoint inhibitors, a significant fraction of patients does not respond to current treatments. This could be due to limitations in tumor immunogenicity and profound immunosuppression in the melanoma microenvironment. Moreover, insufficient tumor antigen processing and presentation by dendritic cells (DC) may hamper the development of tumor-specific T cells. Using two genetically engineered mouse melanoma models (RET and BRAFV600E transgenic mice), in which checkpoint inhibitor therapy alone is not efficacious, we performed proof-of-concept studies with an improved, multivalent DC vaccination strategy based on our recently developed genetic mRNA cancer vaccines. The in vivo expression of multiple chimeric MHC class I receptors allows a simultaneous presentation of several melanoma-associated shared antigens tyrosinase related protein (TRP)-1, tyrosinase, human glycoprotein 100 and TRP-2. The DC vaccine induced a significantly improved survival in both transgenic mouse models. Vaccinated melanoma-bearing mice displayed an increased CD8 T cell reactivity indicated by a higher IFN-γ production and an upregulation of activation marker expression along with an attenuated immunosuppressive pattern of myeloid-derived suppressor cells (MDSC) and regulatory T cells (Treg). The combination of DC vaccination with ultra-low doses of paclitaxel or anti-PD-1 antibodies resulted in further prolongation of mouse survival associated with a stronger reduction of MDSC and Treg immunosuppressive phenotype. Our data suggest that an improved multivalent DC vaccine based on shared tumor antigens induces potent anti-tumor effects and could be combined with checkpoint inhibitors or targeting immunosuppressive cells to further improve their therapeutic efficiency.

11.
J Clin Invest ; 110(4): 453-62, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12189239

RESUMEN

The human milk fat globule membrane protein BA46 (lactadherin) is highly overexpressed in human breast tumors, making it a potential target for tumor immunotherapy. We have identified BA46-derived peptides that contain the motif recognized by the MHC class I molecule HLA-A2.1 and that are processed and presented by human breast carcinoma cells. In mice lacking normal class I molecules but expressing an HLA-A2.1/D(b)-beta2 microglobulin single chain (HHD mice), three peptides elicited specific CTL activity. Two of these peptides also stimulated cytotoxic activity in peripheral blood lymphocytes from HLA-A2.1-positive breast carcinoma patients. Adoptive transfer of HHD-derived bulk CTLs to nude mice bearing human breast carcinoma transplants reduced tumor growth. These peptides therefore represent naturally processed BA46-derived CTL epitopes that can be used in peptide-based antitumor vaccines.


Asunto(s)
Antígenos de Superficie/inmunología , Neoplasias de la Mama/inmunología , Carcinoma/inmunología , Antígeno HLA-A2/genética , Proteínas de la Leche/inmunología , Proteínas de Neoplasias/inmunología , Microglobulina beta-2/genética , Traslado Adoptivo , Animales , Antígenos de Superficie/química , Antígenos de Superficie/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Carcinoma/genética , Carcinoma/metabolismo , Extractos Celulares/inmunología , Epítopos/inmunología , Femenino , Antígeno HLA-A2/metabolismo , Humanos , Ratones , Ratones Noqueados , Ratones Desnudos , Ratones Transgénicos , Proteínas de la Leche/química , Proteínas de la Leche/metabolismo , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Péptidos/inmunología , ARN Neoplásico/biosíntesis , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/trasplante , Células Tumorales Cultivadas
12.
Cancer Res ; 65(14): 6435-42, 2005 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-16024648

RESUMEN

Specific immunotherapy of prostate cancer may be an alternative or be complementary to other approaches for treatment of recurrent or metastasized disease. This study aims at identifying and characterizing prostate cancer-associated peptides capable of eliciting specific CTL responses in vivo. Evaluation of peptide-induced CTL activity in vitro was done following immunization of HLA-A2 transgenic (HHD) mice. An in vivo tumor rejection was tested by adoptive transfer of HHD immune lymphocytes to nude mice bearing human tumors. To confirm the existence of peptide-specific CTL precursors in human, lymphocytes from healthy and prostate cancer individuals were stimulated in vitro in the presence of these peptides and CTL activities were assayed. Two novel immunogenic peptides derived from overexpressed prostate antigens, prostatic acid phosphatase (PAP) and six-transmembrane epithelial antigen of prostate (STEAP), were identified; these peptides were designated PAP-3 and STEAP-3. Peptide-specific CTLs lysed HLA-A2.1+ LNCaP cells and inhibited tumor growth on adoptive immunotherapy. Furthermore, peptide-primed human lymphocytes derived from healthy and prostate cancer individuals lysed peptide-pulsed T2 cells and HLA-A2.1+ LNCaP cells. Based on the results presented herein, PAP-3 and STEAP-3 are naturally processed CTL epitopes possessing anti-prostate cancer reactivity in vivo and therefore may constitute vaccine candidates to be investigated in clinical trials.


Asunto(s)
Antígenos de Neoplasias/inmunología , Epítopos de Linfocito T/inmunología , Inmunoterapia Adoptiva/métodos , Fragmentos de Péptidos/inmunología , Neoplasias de la Próstata/inmunología , Proteínas Tirosina Fosfatasas/inmunología , Linfocitos T Citotóxicos/inmunología , Fosfatasa Ácida , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Antígeno HLA-A2/genética , Antígeno HLA-A2/inmunología , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Desnudos , Ratones Transgénicos , Oxidorreductasas , Neoplasias de la Próstata/terapia , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Clin Cancer Res ; 11(13): 4955-61, 2005 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-16000595

RESUMEN

PURPOSE: Cryotherapy of localized prostate, renal, and hepatic primary tumors and metastases is considered a minimally invasive treatment demonstrating a low complication rate in comparison with conventional surgery. The main drawback of cryotherapy is that it has no systemic effect on distant metastases. We investigated whether intratumoral injections of dendritic cells following cryotherapy of local tumors (cryoimmunotherapy) provides an improved approach to cancer treatment, combining local tumor destruction and systemic anticancer immunity. EXPERIMENTAL DESIGNS: The 3LL murine Lewis lung carcinoma clone D122 and the ovalbumin-transfected B16 melanoma clone MO5 served as models for spontaneous metastasis. The antimetastatic effect of cryoimmunotherapy was assessed in the lung carcinoma model by monitoring mouse survival, lung weight, and induction of tumor-specific CTLs. The mechanism of cryoimmunotherapy was elucidated in the melanoma model using adoptive transfer of T cell receptor transgenic OT-I CTLs into the tumor-bearing mice, and analysis of Th1/Th2 responses by intracellular cytokine staining in CD4 and CD8 cells. RESULTS: Cryoimmunotherapy caused robust and tumor-specific CTL responses, increased Th1 responses, significantly prolonged survival and dramatically reduced lung metastasis. Although intratumor administration of dendritic cells alone increased the proliferation rate of CD8 cells, only cryoimmunotherapy resulted in the generation of effector memory cells. Furthermore, cryoimmunotherapyprotected mice that had survived primary MO5 tumors from rechallenge with parental tumors. CONCLUSIONS: These results present cryoimmunotherapy as a novel approach for systemic treatment of cancer. We envisage that cryotherapy of tumors combined with subsequent in situ immunotherapy by autologous unmodified immature dendritic cells can be applied in practice.


Asunto(s)
Carcinoma Pulmonar de Lewis/terapia , Crioterapia/métodos , Células Dendríticas/inmunología , Inmunoterapia Adoptiva/métodos , Melanoma Experimental/terapia , Metástasis de la Neoplasia/prevención & control , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/patología , Proliferación Celular/efectos de los fármacos , Terapia Combinada , Femenino , Citometría de Flujo , Receptores de Hialuranos/inmunología , Interferón gamma/inmunología , Interleucina-4/inmunología , Selectina L/inmunología , Masculino , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Metástasis de la Neoplasia/inmunología , Receptores de Antígenos de Linfocitos T/genética , Análisis de Supervivencia , Resultado del Tratamiento
14.
Oncoimmunology ; 5(6): e1160183, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27471629

RESUMEN

Malignant melanoma is characterized by a rapid progression, metastasis to distant organs and resistance to chemo and radiotherapy. Although melanoma is capable of eliciting an immune response, the disease progresses and the overall results of immunotherapeutic clinical studies are not satisfactory. Recently, we have developed a novel genetic platform for improving an induction of peptide-specific CD8(+) T cells by dendritic cell (DC) based on membrane-anchored ß2-microglobulin (ß2m) linked to a selected antigenic peptide at the N-terminus and to the cytosolic domain of TLR4 at the C-terminus. In vitro transcribed mRNA transfection of antigen-presenting cells (APCs) resulted in an efficient coupling of peptide presentation and cell activation. In this research, we utilize the chimeric platform to induce an immune response in ret transgenic mice that spontaneously develop malignant skin melanoma and to examine its effect on the overall survival of tumor-bearing mice. Following immunization with chimeric construct system, we observe a significantly prolonged survival of tumor-bearing mice as compared to the control group. Moreover, we see elevations in the frequency of CD62L(hi)CD44(hi) central and CD62L(lo)CD44(hi) effector memory CD8(+) T-cell subsets. Importantly, we do not observe any changes in frequencies of regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) in the vaccinated groups. Our data suggest that this novel vaccination approach could be efficiently applied for the immunotherapy of malignant melanoma.

15.
Cancer Gene Ther ; 11(3): 237-48, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14739939

RESUMEN

Perforin/granzyme B- and Fas/FasL-mediated killing pathways are the main effector mechanisms of CTL and NK cells in antitumor immune responses. In this study, we investigated the relative role of these two lytic mechanisms in protection of the host from tumor progression, as well as spontaneous metastasis, using the D122 Lewis lung carcinoma and its gene-modified cells. Utilizing perforin knockout mice (B6-PKO) and Fas and FasL mutant (B6-MRL and B6-Smn) mice, we found that perforin expression in the host plays a crucial function in the prevention of metastasis. However, local tumor rejection of an H-2K(b) and B7-1 transfectant, 39.5-B7 cells, was not dependent either on perforin or Fas/FasL expression in vivo. In addition, CTL lysis of 39.5-B7 cells was independent of perforin and Fas/FasL interactions in 18-hour in vitro assays. We also confirmed that CD8 T-cells were responsible for rejecting 39.5-B7 local tumors, yet cytokines, TNF-alpha and gammaIFN were not involved in tumor rejection in vivo. Furthermore, blocking assays using caspase inhibitors (zVAD-fmk, zLETD-fmk and zLEHD-fmk) showed that, whereas caspase activation was partially required to induce 39.5-B7 lysis mediated by the perforin-dependent pathway, 39.5-B7 lysis by CTLs through the perforin-independent mechanism required caspase activation. Thus, these results suggested that perforin, Fas/FasL, gammaIFN and TNF-alpha independent lytic mechanisms, mediated by CD8 T cells, have a crucial role in rejection of 39.5-B7 cells in vivo. Caspase activation is a pre requisite for apoptosis of targets by CTLs.


Asunto(s)
Carcinoma Pulmonar de Lewis/inmunología , Glicoproteínas de Membrana/metabolismo , Linfocitos T Citotóxicos/inmunología , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis , Antígeno B7-1/metabolismo , Inhibidores de Caspasas , Caspasas/metabolismo , Línea Celular Tumoral , Inhibidores de Cisteína Proteinasa/farmacología , Proteína Ligando Fas , Antígenos H-2/metabolismo , Antígeno de Histocompatibilidad H-2D , Interferón gamma/antagonistas & inhibidores , Células Asesinas Naturales/inmunología , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Trasplante de Neoplasias , Perforina , Proteínas Citotóxicas Formadoras de Poros , Ligando Inductor de Apoptosis Relacionado con TNF , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo
16.
Immunol Lett ; 91(2-3): 119-26, 2004 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-15019279

RESUMEN

The effects of Fas-ligand (FasL) expression by tumor cells on their tumorigenicity and immunogenicity have been reported as opposite, contradictory results. In some systems the killing of Fas positive cytotoxic T-cells (CTL) by FasL expressing tumors resulted in increased tumorigenicity while in other systems tumors expressing FasL were eliminated by neutrophil mediated inflammation. In the present study, we investigated how FasL expression influences the low immunogenic Lewis lung carcinoma clone D122 and its highly immunogenic MHC I (H-2Kb) and B7-1 (CD80) transfectant 39.5-B7, by transfecting the human FasL (FasL) gene into these cells. Despite the fact that FasL-expressing cells kill effectively appropriate target cells (L1210-fas) compared to parental cells (D122) and low expressors (DFasL-33), these tumor cells were completely rejected in syngeneic mice (C57BL/6), but not in Fas mutant B6-MRL mice, suggesting that functional Fas receptor expression in the host was required to induce an anti-tumor mechanism. In addition, although FasL-expressing immunogenic tumor cells (39.5-B7-FasL 7) kill effectively target cells in vitro, both the transfectant and the mock transfectant (39.5-B7-pBabe) were rejected in syngenic mice. The sensitivity of FasL expressing tumor cells to lysis by CTLs was similar to that of FasL non-expressors. Therefore, these results indicate that FasL expression on immunogenic tumor cells does not affect their immunogenicity in vivo, as well as CTL functions in vitro.


Asunto(s)
Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/metabolismo , Regulación Neoplásica de la Expresión Génica , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Linfocitos T Citotóxicos/inmunología , Animales , Carcinoma Pulmonar de Lewis/genética , Proteína Ligando Fas , Humanos , Ratones , Mutación/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Linfocitos T Citotóxicos/citología , Transfección , Células Tumorales Cultivadas
17.
Ann N Y Acad Sci ; 1283: 87-90, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23387369

RESUMEN

Cytotoxic T lymphocytes (CTLs) are the major effector arm of the immune system against tumors. Many tumor-associated antigens (TAAs), known today as potential rejection antigens, were identified by their ability to induce CTL responses. CTLs utilize their clonotypic T cell receptor (TCR) to recognize short antigenic peptides presented on major histocompatibility complex (MHC)-I proteins. These consist of a membrane-attached α heavy chain, which forms the peptide binding pocket, and a noncovalently associated ß2m light chain, not anchored to the cell membrane. CTL activation requires that antigenic peptides be presented initially on professional antigen presenting cells (APCs), primarily dendritic cells (DCs). Autologous DCs are a powerful tool for the induction of antitumor responses and are thus widely explored as vehicles for cancer vaccines. Although encouraging evidence for the induction of tumor-specific CTLs by ex vivo-manipulated DCs came from numerous animal studies, reproducible objective clinical response in human trials is yet to be demonstrated.


Asunto(s)
Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Microglobulina beta-2/genética , Microglobulina beta-2/metabolismo , Animales , Vacunas contra el Cáncer/uso terapéutico , Diseño de Fármacos , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/uso terapéutico , Ratones , Ingeniería de Proteínas , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/uso terapéutico
18.
PLoS One ; 8(2): e55583, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23383339

RESUMEN

Identification and quantification of immunogenic peptides and tumor-derived epitopes presented on MHC-I molecules are essential for basic studies and vaccines generation. Although lymphocytes derived from transgenic mice can serve as sensitive detectors of processes of antigen presentation and recognition, they are not always available. The use of cell lines might be extremely useful. In this study, we generated a lacZ inducible CD8⁺ hybridoma (BUSA14) capable of recognizing both human and mouse gp10025₋33 melanoma antigens presented on dendritic and tumor cell lines. This hybridoma expresses a variety of membranal T cell markers and secretes IL-2 and TNFα. Thus, BUSA14 offers a quantifiable, cheap and straightforward tool for studying peptide presentation by MHC-I molecules on the cell surface.


Asunto(s)
Linfocitos T CD8-positivos/citología , Antígenos de Histocompatibilidad Clase I/metabolismo , Hibridomas/metabolismo , Antígeno gp100 del Melanoma/metabolismo , Animales , Línea Celular Tumoral , Células Dendríticas/metabolismo , Citometría de Flujo , Humanos , Hibridomas/citología , Interleucina-2/metabolismo , Operón Lac , Ratones , Factor de Necrosis Tumoral alfa/metabolismo
19.
Cancer Res ; 68(9): 3450-7, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18451173

RESUMEN

Cytotoxic T cells (CTL) play a major role in tumor rejection. Expansion of CTLs, either by immunization or adoptive transfer, is a prominent goal in current immunotherapy. The antigen-specific nature of these expansion processes inevitably initiates a clonotypic attack on the tumor. By injecting an Ovalbumin-expressing melanoma into OT-I mice, in which >90% of CTLs recognize an Ovalbumin peptide, we show that an increased number of tumor-specific CTLs causes emergence of escape variants. We show that these escape variants are a result of antigen silencing via a yet undetermined epigenetic mechanism, which occurs frequently and is spontaneously reversible. We further show that an increase in the time of tumor onset in OT-I compared with C57BL/6J is a result of immune selection.


Asunto(s)
Linfocitos Infiltrantes de Tumor/patología , Melanoma Experimental/inmunología , Linfocitos T/patología , Escape del Tumor/inmunología , Traslado Adoptivo , Animales , Antígenos de Neoplasias/genética , Moléculas de Adhesión Celular/genética , Línea Celular Tumoral , Proliferación Celular , Silenciador del Gen/fisiología , Recuento de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Neoplasias/genética , Ovalbúmina/genética , Edición de ARN/inmunología , Edición de ARN/fisiología
20.
Cancer Immunol Immunother ; 56(2): 217-26, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16738849

RESUMEN

Conventional treatment of recurrent and metastasized prostate cancer (CaP) remains inadequate; this fact mandates development of alternative therapeutic modalities, such as specific active or passive immunotherapy. Previously, we reported the identification of a novel highly immunogenic HLA-A*0201-restricted Prostatic Acid Phosphatase-derived peptide (PAP-3) by a two-step in vivo screening in an HLA-transgenic (HHD) mouse system. In the present study we aimed at elucidating the efficiency of PAP-3-based vaccine upon active antitumor immunization. To this end we established preventive and therapeutic carcinoma models in HHD mice. The 3LL murine Lewis lung carcinoma clone D122 transduced to express HLA-A*0201 and PAP served as a platform for these models. The HLA-A*0201-PAP-3 complex specific recombinant single chain scFV-PAP-3 antibodies were generated and used to confirm an endogenous PAP processing resulting in PAP-3 presentation by HLA-A*0201. PAP-3 based vaccines significantly decreased tumor incidence in a preventive immunization setting. Therapeutic vaccination of HHD mice with PAP-3 led to rejection of early established tumors and to increase of mouse survival. These results strongly support a therapeutic relevance of the identified CTL epitope upon active antitumor immunization. The newly established carcinoma model presented herein might be a useful tool for cancer vaccine design and optimization.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Inmunoterapia/métodos , Linfocinas/inmunología , Neoplasias de la Próstata/inmunología , Proteínas Tirosina Fosfatasas/inmunología , Sialoglicoproteínas/inmunología , Fosfatasa Ácida , Animales , Vacunas contra el Cáncer/uso terapéutico , Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/terapia , Epítopos de Linfocito T/inmunología , Antígenos HLA-A/inmunología , Antígeno HLA-A2 , Humanos , Linfocinas/uso terapéutico , Masculino , Ratones , Ratones Transgénicos , Microscopía Confocal , Reacción en Cadena de la Polimerasa , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Tirosina Fosfatasas/uso terapéutico , Sialoglicoproteínas/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA