Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
Blood ; 141(5): 490-502, 2023 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-36322928

RESUMEN

Primary myelofibrosis (PMF) is a myeloproliferative neoplasm characterized by the clonal expansion of myeloid cells, notably megakaryocytes (MKs), and an aberrant cytokine production leading to bone marrow (BM) fibrosis and insufficiency. Current treatment options are limited. TGF-ß1, a profibrotic and immunosuppressive cytokine, is involved in PMF pathogenesis. While all cell types secrete inactive, latent TGF-ß1, only a few activate the cytokine via cell type-specific mechanisms. The cellular source of the active TGF-ß1 implicated in PMF is not known. Transmembrane protein GARP binds and activates latent TGF-ß1 on the surface of regulatory T lymphocytes (Tregs) and MKs or platelets. Here, we found an increased expression of GARP in the BM and spleen of mice with PMF and tested the therapeutic potential of a monoclonal antibody (mAb) that blocks TGF-ß1 activation by GARP-expressing cells. GARP:TGF-ß1 blockade reduced not only fibrosis but also the clonal expansion of transformed cells. Using mice carrying a genetic deletion of Garp in either Tregs or MKs, we found that the therapeutic effects of GARP:TGF-ß1 blockade in PMF imply targeting GARP on Tregs. These therapeutic effects, accompanied by increased IFN-γ signals in the spleen, were lost upon CD8 T-cell depletion. Our results suggest that the selective blockade of TGF-ß1 activation by GARP-expressing Tregs increases a CD8 T-cell-mediated immune reaction that limits transformed cell expansion, providing a novel approach that could be tested to treat patients with myeloproliferative neoplasms.


Asunto(s)
Mielofibrosis Primaria , Factor de Crecimiento Transformador beta1 , Ratones , Animales , Mielofibrosis Primaria/tratamiento farmacológico , Mielofibrosis Primaria/genética , Mielofibrosis Primaria/metabolismo , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales/metabolismo , Citocinas/metabolismo , Fibrosis , Linfocitos T Reguladores
2.
Clin Immunol ; 237: 108984, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35338000

RESUMEN

The exact etiopathology of chilblains observed during the Coronavirus Disease 2019 (COVID-19) pandemic is still unclear. Initially, SARS-CoV-2 appeared as the obvious causing agent, but two years of various investigations have failed to convincingly support its direct implication. Most affected individuals have no detectable virus, no anti-SARS-CoV-2 antibodies and no symptoms of COVID-19. Analyses of skin biopsies similarly failed to unambiguously demonstrate presence of the virus or its genome. In a recent hypothesis, SARS-CoV-2 would cause the lesions before being promptly eliminated by unusually strong type I interferon responses. With others, we feel that environmental factors have not been sufficiently considered, in particular cold exposure related to unprecedented containment measures. The cause of pandemic chilblains remains a stimulating puzzle which warrants further investigation.


Asunto(s)
COVID-19 , Eritema Pernio , Eritema Pernio/diagnóstico , Eritema Pernio/epidemiología , Eritema Pernio/patología , Humanos , Pandemias , SARS-CoV-2 , Piel/patología
3.
Cancer Immunol Immunother ; 71(8): 1851-1862, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34973084

RESUMEN

Transmembrane protein GARP binds latent TGF-ß1 to form GARP:(latent)TGF-ß1 complexes on the surface of several cell types including Tregs, B-cells, and platelets. Upon stimulation, these cells release active TGF-ß1. Blocking TGF-ß1 activation by Tregs with anti-GARP:TGF-ß1 mAbs overcomes resistance to PD1/PD-L1 blockade and induces immune-mediated regressions of murine tumors, indicating that Treg-derived TGF-ß1 inhibits anti-tumor immunity. TGF-ß1 exerts a vast array of effects on immune responses. For example, it favors differentiation of TH17 cells and B-cell switch to IgA production, two important processes for mucosal immunity. Here, we sought to determine whether treatment with anti-GARP:TGF-ß1 mAbs would perturb immune responses to intestinal bacterial infection. We observed no aggravation of intestinal disease, no systemic dissemination, and no alteration of innate or adaptative immune responses upon oral gavage of C. rodentium in highly susceptible Il22r-/- mice treated with anti-GARP:TGF-ß1 mAbs. To examine the effects of GARP:TGF-ß1 blockade on Ig production, we compared B cell- and TH cell- responses to OVA or CTB protein immunization in mice carrying deletions of Garp in Tregs, B cells, or platelets. No alteration of adaptive immune responses to protein immunization was observed in the absence of GARP on any of these cells. Altogether, we show that antibody-mediated blockade of GARP:TGF-ß1 or genetic deletion of Garp in Tregs, B cells or platelets, do not alter innate or adaptive immune responses to intestinal bacterial infection or protein immunization in mice. Anti-GARP:TGF-ß1 mAbs, currently tested for cancer immunotherapy, may thus restore anti-tumor immunity without severely impairing other immune defenses. PRéCIS: Immunotherapy with GARP:TGF-ß1 mAbs may restore anti-tumor immunity without impairing immune or inflammatory responses required to maintain homeostasis or host defense against infection, notably at mucosal barriers.


Asunto(s)
Inmunidad Adaptativa , Infecciones Bacterianas , Proteínas de la Membrana , Factor de Crecimiento Transformador beta1 , Animales , Anticuerpos Monoclonales/metabolismo , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/metabolismo , Inmunidad , Inmunización , Proteínas de la Membrana/metabolismo , Ratones , Linfocitos T Reguladores , Factor de Crecimiento Transformador beta1/metabolismo
4.
Trends Immunol ; 38(8): 577-593, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28610825

RESUMEN

Cancer immunotherapy is experiencing a renaissance spearheaded by immune checkpoint inhibitors (ICIs). This has spurred interest in 'upgrading' existing immunotherapies that previously experienced only sporadic success, such as dendritic cells (DCs) vaccines. In this review, we discuss the major molecular, immunological, and clinical determinants of existing first- and second-generation DC vaccines. We also outline the future trends for next-generation DC vaccines and describe their major hallmarks and prerequisites necessary for high anticancer efficacy. In addition, using existing data we compare DC vaccines with ICIs targeting CTLA4, PD1, and PD-L1, and argue that in various contexts next-generation DC vaccines are ready to meet some challenges currently confronting ICIs, thereby raising the need to integrate DC vaccines in future combinatorial immunotherapy regimens.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Células Dendríticas/inmunología , Inmunoterapia/tendencias , Neoplasias/terapia , Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Quimioterapia Adyuvante , Humanos , Terapia Molecular Dirigida , Neoplasias/inmunología
5.
Clin Exp Rheumatol ; 38(5): 881-890, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31969218

RESUMEN

OBJECTIVES: TLR3 mediates skin solar injury by binding nuclear material released from apoptotic keratinocytes, resulting in the production of pro-inflammatory cytokines. Because the TLR3 gene is located in 4q35, a known systemic lupus erythematosus (SLE) susceptibility locus, we wondered whether TLR3 single nucleotide polymorphisms (SNPs) were associated with inflammatory mechanisms relevant to the development of SLE, and disease susceptibility. METHODS: Functional assays were carried out in TLR3-transfected HEK293 cells and in monocyte-derived dendritic cells (moDCs). TLR3 and IFNß immunofluorescence studies were performed in skin samples from 7 SLE patients and 3 controls. We performed a SNP association study in a discovery cohort of 153 patients and 105 controls, followed by a confirmation study in an independent cohort of 1,380 patients and 2,104 controls. RESULTS: TLR3 and IFNß are overexpressed in SLE skin lesions. TLR3 overexpression in HEK293 cells amplifies their sensitivity to a pro-apoptotic stimulus. Taking advantage of a naturally occurring polymorphic TLR3 variant (rs3775291) that weakly versus strongly responds to poly I:C stimulation, we found that TLR3 is associated with amplified apoptotic responses, production of the Ro/SSA autoantigen and increased maturation of myeloid-derived dendritic cells (moDC) after exposure to UV irradiation. However, TLR3 SNPs are not associated with susceptibility to SLE in a large population of patients and controls. CONCLUSIONS: TLR3 is overexpressed in SLE skin lesions and amplifies apoptotic and inflammatory responses to UV-irradiation in antigen-presenting cells in vitro. However, TLR3 SNPs do not impact susceptibility to the development of the disease.


Asunto(s)
Lupus Eritematoso Sistémico , Receptor Toll-Like 3 , Células Presentadoras de Antígenos , Apoptosis , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Lupus Eritematoso Sistémico/genética , Polimorfismo de Nucleótido Simple , Receptor Toll-Like 3/genética
6.
Proc Natl Acad Sci U S A ; 114(47): E10161-E10168, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29109269

RESUMEN

Human regulatory T cells (Tregs) suppress other T cells by converting the latent, inactive form of TGF-ß1 into active TGF-ß1. In Tregs, TGF-ß1 activation requires GARP, a transmembrane protein that binds and presents latent TGF-ß1 on the surface of Tregs stimulated through their T cell receptor. However, GARP is not sufficient because transduction of GARP in non-Treg T cells does not induce active TGF-ß1 production. RGD-binding integrins were shown to activate TGF-ß1 in several non-T cell types. Here we show that αVß8 dimers are present on stimulated human Tregs but not in other T cells, and that antibodies against αV or ß8 subunits block TGF-ß1 activation in vitro. We also show that αV and ß8 interact with GARP/latent TGF-ß1 complexes in human Tregs. Finally, a blocking antibody against ß8 inhibited immunosuppression by human Tregs in a model of xenogeneic graft-vs.-host disease induced by the transfer of human T cells in immunodeficient mice. These results show that TGF-ß1 activation on the surface of human Tregs implies an interaction between the integrin αVß8 and GARP/latent TGF-ß1 complexes. Immunosuppression by human Tregs can be inhibited by antibodies against GARP or against the integrin ß8 subunit. Such antibodies may prove beneficial against cancer or chronic infections.


Asunto(s)
Enfermedad Injerto contra Huésped/inmunología , Tolerancia Inmunológica/efectos de los fármacos , Integrinas/inmunología , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta1/inmunología , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Integrinas/antagonistas & inhibidores , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Ratones SCID , Neoplasias/inmunología , Neoplasias/terapia , Linfocitos T Reguladores/trasplante , Factor de Crecimiento Transformador beta1/metabolismo , Trasplante Heterólogo
7.
J Immunol ; 199(2): 391-396, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28607112

RESUMEN

Production of active TGF-ß is regulated at a posttranslational level and implies release of the mature cytokine dimer from the inactive, latent TGF-ß precursor. There are several cell-type specific mechanisms of TGF-ß activation. We identified a new mechanism operating on the surface of human regulatory T cells and involving membrane protein GARP, which binds latent TGF-ß1. The paracrine activity of regulatory T cell-derived TGF-ß1 contributes to immunosuppression and can be inhibited with anti-GARP Abs. Whether other immune cell types use surface GARP to activate latent TGF-ß1 was not known. We show in this study that stimulated, human B lymphocytes produce active TGF-ß1 from surface GARP/latent TGF-ß1 complexes with isotype switching to IgA production.


Asunto(s)
Linfocitos B/inmunología , Inmunoglobulina A/biosíntesis , Cambio de Clase de Inmunoglobulina , Proteínas de la Membrana/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Linfocitos B/efectos de los fármacos , Citocinas/inmunología , Humanos , Inmunoglobulina A/inmunología , Activación de Linfocitos , Oligodesoxirribonucleótidos/inmunología , Unión Proteica , Recombinación Genética , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta1/inmunología
8.
J Immunol ; 197(6): 2541-52, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27489285

RESUMEN

Adoptive T cell therapy has shown significant clinical success for patients with advanced melanoma and other tumors. Further development of T cell therapy requires improved strategies to select effective, yet nonself-reactive, TCRs. In this study, we isolated 10 TCR sequences against four MAGE-C2 (MC2) epitopes from melanoma patients who showed clinical responses following vaccination that were accompanied by significant frequencies of anti-MC2 CD8 T cells in blood and tumor without apparent side effects. We introduced these TCRs into T cells, pretreated tumor cells of different histological origins with the epigenetic drugs azacytidine and valproate, and tested tumor and self-reactivities of these TCRs. Pretreatment of tumor cells upregulated MC2 gene expression and enhanced recognition by T cells. In contrast, a panel of normal cell types did not express MC2 mRNA, and similar pretreatment did not result in recognition by MC2-directed T cells. Interestingly, the expression levels of MC2, but not those of CD80, CD86, or programmed death-ligand 1 or 2, correlated with T cell responsiveness. One of the tested TCRs consistently recognized pretreated MC2(+) cell lines from melanoma, head and neck, bladder, and triple-negative breast cancers but showed no response to MHC-eluted peptides or peptides highly similar to MC2. We conclude that targeting MC2 Ag, combined with epigenetic drug-enhanced antigenicity, allows for significant and tumor-selective T cell responses.


Asunto(s)
Antígenos de Neoplasias/inmunología , Melanoma/inmunología , Proteínas de Neoplasias/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología , Antígenos de Neoplasias/genética , Azacitidina/farmacología , Antígeno B7-1/genética , Antígeno B7-1/inmunología , Antígeno B7-2/genética , Antígeno B7-2/inmunología , Antígeno B7-H1/genética , Antígeno B7-H1/inmunología , Línea Celular Tumoral , Epítopos de Linfocito T/inmunología , Humanos , Inmunoterapia Adoptiva , Activación de Linfocitos , Melanoma/patología , Melanoma/terapia , Proteínas de Neoplasias/genética , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/efectos de los fármacos , Ácido Valproico/farmacología
9.
Eur J Immunol ; 46(6): 1449-59, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27000947

RESUMEN

IL-22 has a detrimental role in skin inflammatory processes, for example in psoriasis. As transcription factor, AhR controls the IL-22 production by several cell types (i.e. Th17 cells). Here, we analyzed the role of Ahr in IL-22 production by immune cells in the inflamed skin, using an imiquimod-induced psoriasis mouse model. Our results indicate that IL-22 is expressed in the ear of imiquimod-treated Ahr(-/-) mice but less than in wild-type mice. We then studied the role of AhR on three cell populations known to produce IL-22 in the skin: γδ T cells, Th17 cells, and ILC3, and a novel IL-22-producing cell type identified in this setting: CD4(-) CD8(-) TCRß(+) T cells. We showed that AhR is required for IL-22 production by Th17, but not by the three other cell types, in the imiquimod-treated ears. Moreover, AhR has a role in the recruitment of γδ T cells, ILC3, and CD4(-) CD8(-) TCRß(+) T cells into the inflamed skin or in their local proliferation. Taken together, AhR has a direct role in IL-22 production by Th17 cells in the mouse ear skin, but not by γδ T cells, CD4(-) CD8(-) TCRß(+) T cells and ILCs.


Asunto(s)
Aminoquinolinas/efectos adversos , Quimiotaxis/inmunología , Interleucinas/biosíntesis , Psoriasis/etiología , Psoriasis/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Animales , Proliferación Celular , Quimiotaxis/genética , Modelos Animales de Enfermedad , Imiquimod , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Interleucinas/genética , Ratones , Ratones Noqueados , Psoriasis/patología , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Receptores de Hidrocarburo de Aril/deficiencia , Receptores de Hidrocarburo de Aril/genética , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Interleucina-22
10.
J Biol Chem ; 290(33): 20105-16, 2015 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-26126825

RESUMEN

Production of active TGF-ß1 is one mechanism by which human regulatory T cells (Tregs) suppress immune responses. This production is regulated by glycoprotein A repetitions predominant (GARP), a transmembrane protein present on stimulated Tregs but not on other T lymphocytes (Th and CTLs). GARP forms disulfide bonds with proTGF-ß1, favors its cleavage into latent inactive TGF-ß1, induces the secretion and surface presentation of GARP·latent TGF-ß1 complexes, and is required for activation of the cytokine in Tregs. We explored whether additional Treg-specific protein(s) associated with GARP·TGF-ß1 complexes regulate TGF-ß1 production in Tregs. We searched for such proteins by yeast two-hybrid assay, using GARP as a bait to screen a human Treg cDNA library. We identified lysosomal-associated transmembrane protein 4B (LAPTM4B), which interacts with GARP in mammalian cells and is expressed at higher levels in Tregs than in Th cells. LAPTM4B decreases cleavage of proTGF-ß1, secretion of soluble latent TGF-ß1, and surface presentation of GARP·TGF-ß1 complexes by Tregs but does not contribute to TGF-ß1 activation. Therefore, LAPTM4B binds to GARP and is a negative regulator of TGF-ß1 production in human Tregs. It may play a role in the control of immune responses by decreasing Treg immunosuppression.


Asunto(s)
Proteínas de la Membrana/fisiología , Proteínas Oncogénicas/fisiología , Linfocitos T Reguladores/metabolismo , Factor de Crecimiento Transformador beta1/biosíntesis , Células HEK293 , Humanos , Proteínas de la Membrana/metabolismo , Técnicas del Sistema de Dos Híbridos
11.
Cancer Immunol Immunother ; 65(10): 1177-88, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27514672

RESUMEN

Nectin-4 is a tumor antigen present on the surface of breast, ovarian and lung carcinoma cells. It is rarely present in normal adult tissues and is therefore a candidate target for cancer immunotherapy. Here, we identified a Nectin-4 antigenic peptide that is naturally presented to T cells by HLA-A2 molecules. We first screened the 502 nonamer peptides of Nectin-4 (510 amino acids) for binding to and off-rate from eight different HLA class I molecules. We then combined biochemical, cellular and algorithmic assays to select 5 Nectin-4 peptides that bound to HLA-A*02:01 molecules. Cytolytic T lymphocytes were obtained from healthy donors, that specifically lyzed HLA-A2(+) cells pulsed with 2 out of the 5 peptides, indicating the presence of anti-Nectin-4 CD8(+) T lymphocytes in the human T cell repertoire. Finally, an HLA-A2-restricted cytolytic T cell clone derived from a breast cancer patient recognized peptide Nectin-4145-153 (VLVPPLPSL) and lyzed HLA-A2(+) Nectin-4(+) breast carcinoma cells. These results indicate that peptide Nectin-4145-153 is naturally processed for recognition by T cells on HLA-A2 molecules. It could be used to monitor antitumor T cell responses or to immunize breast cancer patients.


Asunto(s)
Adenocarcinoma/inmunología , Neoplasias de la Mama/inmunología , Vacunas contra el Cáncer/inmunología , Moléculas de Adhesión Celular/metabolismo , Epítopos de Linfocito T/metabolismo , Inmunoglobulinas/metabolismo , Péptidos/metabolismo , Linfocitos T Citotóxicos/inmunología , Antígenos de Neoplasias/inmunología , Línea Celular Tumoral , Células Clonales , Mapeo Epitopo , Femenino , Antígeno HLA-A2/metabolismo , Humanos , Unión Proteica
12.
J Transl Med ; 14(1): 232, 2016 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-27484791

RESUMEN

BACKGROUND: Ipilimumab (Ipi) improves the survival of advanced melanoma patients with an incremental long-term benefit in 10-15 % of patients. A tumor signature that correlates with this survival benefit could help optimizing individualized treatment strategies. METHODS: Freshly frozen melanoma metastases were collected from patients treated with either Ipi alone (n: 7) or Ipi combined with a dendritic cell vaccine (TriMixDC-MEL) (n: 11). Samples were profiled by immunohistochemistry (IHC), whole transcriptome (RNA-seq) and methyl-DNA sequencing (MBD-seq). RESULTS: Patients were divided in two groups according to clinical evolution: durable benefit (DB; 5 patients) and no clinical benefit (NB; 13 patients). 20 metastases were profiled by IHC and 12 were profiled by RNA- and MBD-seq. 325 genes were identified as differentially expressed between DB and NB. Many of these genes reflected a humoral and cellular immune response. MBD-seq revealed differences between DB and NB patients in the methylation of genes linked to nervous system development and neuron differentiation. DB tumors were more infiltrated by CD8(+) and PD-L1(+) cells than NB tumors. B cells (CD20(+)) and macrophages (CD163(+)) co-localized with T cells. Focal loss of HLA class I and TAP-1 expression was observed in several NB samples. CONCLUSION: Combined analyses of melanoma metastases with IHC, gene expression and methylation profiling can potentially identify durable responders to Ipi-based immunotherapy.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Epigénesis Genética , Inmunoterapia , Melanoma/genética , Melanoma/terapia , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/genética , Microambiente Tumoral/inmunología , Adulto , Anciano , Anticuerpos Monoclonales/farmacología , Metilación de ADN/efectos de los fármacos , Metilación de ADN/genética , Demografía , Epigénesis Genética/efectos de los fármacos , Femenino , Secciones por Congelación , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Ipilimumab , Masculino , Melanoma/patología , Persona de Mediana Edad , Metástasis de la Neoplasia , Adhesión en Parafina , Inducción de Remisión , Neoplasias Cutáneas/patología , Resultado del Tratamiento , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética
13.
Proc Natl Acad Sci U S A ; 110(21): E1933-42, 2013 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-23630278

RESUMEN

Wildebeests carry asymptomatically alcelaphine herpesvirus 1 (AlHV-1), a γ-herpesvirus inducing malignant catarrhal fever (MCF) to several ruminant species (including cattle). This acute and lethal lymphoproliferative disease occurs after a prolonged asymptomatic incubation period after transmission. Our recent findings with the rabbit model indicated that AlHV-1 infection is not productive during MCF. Here, we investigated whether latency establishment could explain this apparent absence of productive infection and sought to determine its role in MCF pathogenesis. First, whole-genome cellular and viral gene expression analyses were performed in lymph nodes of MCF-developing calves. Whereas a severe disruption in cellular genes was observed, only 10% of the entire AlHV-1 genome was expressed, contrasting with the 45% observed during productive infection in vitro. In vivo, the expressed viral genes included the latency-associated nuclear antigen homolog ORF73 but none of the regions known to be essential for productive infection. Next, genomic conformation analyses revealed that AlHV-1 was essentially episomal, further suggesting that MCF might be the consequence of a latent infection rather than abortive lytic infection. This hypothesis was further supported by the high frequencies of infected CD8(+) T cells during MCF using immunodetection of ORF73 protein and single-cell RT-PCR approaches. Finally, the role of latency-associated ORF73 was addressed. A lack of ORF73 did not impair initial virus replication in vivo, but it rendered AlHV-1 unable to induce MCF and persist in vivo and conferred protection against a lethal challenge with a WT virus. Together, these findings suggest that a latent infection is essential for MCF induction.


Asunto(s)
Antígenos Nucleares/biosíntesis , Antígenos Virales/biosíntesis , Gammaherpesvirinae/fisiología , Regulación Viral de la Expresión Génica/fisiología , Trastornos Linfoproliferativos/metabolismo , Fiebre Catarral Maligna/metabolismo , Latencia del Virus/fisiología , Enfermedad Aguda , Animales , Antígenos Nucleares/genética , Antígenos Virales/genética , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Linfocitos T CD8-positivos/virología , Bovinos , Genoma Viral/fisiología , Trastornos Linfoproliferativos/genética , Trastornos Linfoproliferativos/patología , Trastornos Linfoproliferativos/virología , Fiebre Catarral Maligna/patología , Fiebre Catarral Maligna/virología , Plásmidos/genética , Plásmidos/metabolismo , Conejos , Replicación Viral/fisiología
14.
Mol Vis ; 20: 956-69, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24991188

RESUMEN

PURPOSE: Vogt-Koyanagi-Harada (VKH) syndrome is an autoimmune disease characterized by inaugural uveomeningitidis and hearing loss and at late stages a depigmentation in eyes and skin. Melanocytes are the cells common to the four affected tissues, namely eye, brain, inner ear, and skin. Melanocytes are therefore considered as the source of self-antigens. The melanocytic proteins tyrosinase-related protein-1 (TRP1), TRP2, tyrosinase, and gp100 have been proposed as the proteins targeted by autoreactive T cells from VKH patients bearing human leukocyte antigen (HLA)-DRB1*04:05, the HLA allele classically associated with VKH disease. The objective of this work was to determine the antigens recognized by a large number of potentially autoreactive CD4 T lymphocytes obtained from the cerebrospinal fluid of one VKH patient who did not express HLA-DRB1*04:05. METHODS: T cells were isolated from the cerebrospinal fluid of a newly diagnosed HLA-DRB1*14:01,*15:03;-DPB1*01:01,*04:02 patient in the acute phase of the VKH disease and cloned by limiting dilution. Each of the 107 T cell clones, of which 90% were CD4(+), was tested for its ability to secrete cytokines upon contact with autologous antigen-presenting cells loaded with either of the melanocytic proteins TRP1, TRP2, tyrosinase, gp100, Melan-A and KU-MEL-1. The sensitivity of our recombinant bacteria-based approach was validated with a CD4 T cell clone with known antigen specificity. The ability of each of the 107 clones to secrete cytokines upon nonspecific stimulation was verified. RESULTS: None of the 107 T cell clones was able to secrete tumor necrosis factor-α, interferon-γ, interleukin (IL)-5, or IL-17 upon contact with autologous B cells loaded with any of the six common melanocytic proteins. Nine clones secreted high-level IL-17 upon stimulation with beads coated with antibodies. CONCLUSIONS: The self-antigens that triggered the VKH disease in this patient probably derive from proteins other than the six melanocytic proteins mentioned above. Further study of antigens that are recognized by potential autoreactive T cells from VKH patients is likely to benefit from testing a broader set of melanocytic proteins.


Asunto(s)
Autoantígenos/inmunología , Melanocitos/inmunología , Linfocitos T/inmunología , Síndrome Uveomeningoencefálico/líquido cefalorraquídeo , Síndrome Uveomeningoencefálico/inmunología , Adulto , Antígenos de Neoplasias/metabolismo , Linfocitos B/virología , Bacterias/metabolismo , Western Blotting , Linfocitos T CD4-Positivos/inmunología , Separación Celular , Células Clonales , Citocinas/metabolismo , Epítopos/inmunología , Herpesvirus Humano 4/inmunología , Humanos , Interleucina-17/metabolismo , Masculino , Proteínas de Neoplasias/metabolismo , Proteínas Recombinantes/metabolismo , Síndrome Uveomeningoencefálico/patología
15.
Int J Cancer ; 132(11): 2557-66, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23151995

RESUMEN

Cancer vaccines have recently been shown to induce some clinical benefits. The relationship between clinical activity and anti-vaccine T cell responses is somewhat controversial. Indeed, in many trials it has been documented that the induction of vaccine-specific T cells exceeds the clinical responses observed. Here, we evaluate immunological and clinical responses in 23 MAGE-A3(+) melanoma patients treated with autologous lymphocytes genetically engineered to express the tumor antigen MAGE-A3 and the viral gene product thymidine kinase of the herpes simplex virus (HSV-TK). HSV-TK was used as safety system in case of adverse events and as tracer antigen to monitor the immune competence of treated patients. The increase of anti-TK and anti-MAGE-A3 T-cells after vaccination was observed in 90 and 27% of patients, respectively. Among 19 patients with measurable disease, we observed a disease control rate of 26.3%, with one objective clinical response, and four durable, stable diseases. Three patients out of five with no evidence of disease (NED) at the time of vaccination remained NED after 73+, 70+ and 50+ months. Notably, we report that only patients experiencing MAGE-A3-specific immune responses showed a clinical benefit. Additionally, we report that responder and non-responder patients activate and expand T cells against the tracer antigen TK in a similar way, suggesting that local rather than systemic immune suppression might be involved in limiting clinically relevant antitumor immune responses.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/uso terapéutico , Terapia Genética , Melanoma/inmunología , Proteínas de Neoplasias/inmunología , Linfocitos T/inmunología , Adulto , Anciano , Neoplasias Óseas/inmunología , Neoplasias Óseas/mortalidad , Neoplasias Óseas/terapia , Ensayos Clínicos Fase II como Asunto , Femenino , Estudios de Seguimiento , Humanos , Hipersensibilidad Tardía , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/mortalidad , Neoplasias Hepáticas/terapia , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/terapia , Masculino , Melanoma/mortalidad , Melanoma/terapia , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/mortalidad , Neoplasias Cutáneas/terapia , Linfocitos T/metabolismo , Timidina Quinasa/inmunología , Timidina Quinasa/metabolismo
16.
Ann Surg Oncol ; 20 Suppl 3: S650-60, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23884751

RESUMEN

BACKGROUND: Inflammation is associated with a worse outcome in cancer and neutrophil:lymphocyte ratio (NLR) is a strong prognostic value. In cancer, nonsteroidal anti-inflammatory drugs (NSAIDs) could be of interest. We investigated the prognostic significance of NLR and the impact of intraoperative NSAIDs in cancer surgeries. METHODS: We performed an observational study in early breast, kidney, and lung cancers (357, 227, and 255 patients) with uni- and multivariate analyses (Cox model). RESULTS: In breast cancer (Centre 1), NLR ≥ 4 is associated with a higher risk of relapse (hazards ratio (HR) = 2.41; 95 % confidence interval (CI) 1.01-5.76; P = 0.048). In breast cancer (Centre 2), NLR ≥ 3 is associated with a higher risk of relapse (HR = 4.6; 95 % CI 1.09-19.1; P = 0.04) and higher mortality (HR = 4.0; 95 % CI 1.12-14.3; P = 0.03). In kidney cancer, NLR ≥ 5 is associated with a higher risk of relapse (HR = 1.63; 95 % CI 1.00-2.66; P = 0.05) and higher mortality (HR = 1.67; 95 % CI 1.0-2.81; P = 0.05). In lung cancer, NLR ≥ 5 is associated with higher mortality (HR = 1.45; 95 % CI 1.02-2.06; P = 0.04). The intraoperative use of NSAIDs in breast cancer patients (Centre 1) is associated with a reduced recurrence rate (HR = 0.17; 95 % CI 0.04-0.43; P = 0.0002) and a lower mortality (HR = 0.25; 95 % CI 1.08-0.75; P = 0.01). NSAIDs use at the beginning of the surgery is independently associated with a lower metastases risk after lung cancer surgery (HR = 0.16; 95 % CI 0.04-0.63; P = 0.009). Ketorolac use is independently associated with longer survival (HR = 0.55; 95 % CI 0.31-0.95; P = 0.03). CONCLUSIONS: In these cohorts, these analyses show that NLR is a strong perioperative prognosis factor for breast, lung, and kidney cancers. In this context, intraoperative NSAIDs administration could be associated with a better outcome.


Asunto(s)
Neoplasias de la Mama/patología , Diclofenaco/administración & dosificación , Ketorolaco/administración & dosificación , Neoplasias Renales/patología , Neoplasias Pulmonares/patología , Linfocitos/patología , Neutrófilos/patología , Adenocarcinoma/mortalidad , Adenocarcinoma/secundario , Adenocarcinoma/terapia , Antiinflamatorios no Esteroideos/administración & dosificación , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/terapia , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Carcinoma de Pulmón de Células no Pequeñas/secundario , Carcinoma de Pulmón de Células no Pequeñas/terapia , Carcinoma de Células Escamosas/mortalidad , Carcinoma de Células Escamosas/secundario , Carcinoma de Células Escamosas/terapia , Terapia Combinada , Femenino , Estudios de Seguimiento , Humanos , Cuidados Intraoperatorios , Neoplasias Renales/mortalidad , Neoplasias Renales/terapia , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/terapia , Masculino , Metástasis de la Neoplasia , Recurrencia Local de Neoplasia/mortalidad , Recurrencia Local de Neoplasia/patología , Recurrencia Local de Neoplasia/terapia , Estadificación de Neoplasias , Pronóstico , Estudios Prospectivos , Tasa de Supervivencia
17.
Mol Ther ; 20(5): 1063-74, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22371843

RESUMEN

It is generally thought that dendritic cells (DCs) loaded with full-length tumor antigen could improve immunotherapy by stimulating broad T-cell responses and by allowing treatment irrespective of the patient's human leukocyte antigen (HLA) type. To investigate this, we determined the specificity of T cells from melanoma patients treated with DCs loaded with mRNA encoding a full-length tumor antigen fused to a signal peptide and an HLA class II sorting signal, allowing presentation in HLA class I and II. In delayed-type hypersensitive (DTH)-biopsies and blood, we found functional CD8(+) and CD4(+) T cells recognizing novel treatment-antigen-derived epitopes, presented by several HLA types. Additionally, we identified a CD8(+) response specific for the signal peptide incorporated to elicit presentation by HLA class II and a CD4(+) response specific for the fusion region of the signal peptide and one of the antigens. This demonstrates that the fusion proteins contain newly created immunogenic sequences and provides evidence that ex vivo-generated mRNA-modified DCs can induce effector CD8(+) and CD4(+) T cells from the naive T-cell repertoire of melanoma patients. Thus, this work provides definitive proof that DCs presenting the full antigenic spectrum of tumor antigens can induce T cells specific for novel epitopes and can be administered to patients irrespective of their HLA type.


Asunto(s)
Antígenos de Neoplasias/inmunología , Células Dendríticas/trasplante , Antígenos HLA-D/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Inmunoterapia Adoptiva/métodos , Melanoma/terapia , ARN Mensajero/inmunología , Neoplasias Cutáneas/terapia , Secuencia de Aminoácidos , Presentación de Antígeno , Antígenos de Neoplasias/genética , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Citotoxicidad Inmunológica , Células Dendríticas/inmunología , Electroporación , Antígenos HLA-D/genética , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Activación de Linfocitos , Melanoma/inmunología , Melanoma/patología , Datos de Secuencia Molecular , Señales de Clasificación de Proteína , ARN Mensajero/genética , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Transfección
18.
Clin Cancer Res ; 29(2): 309-315, 2023 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-36044396

RESUMEN

T cells are key effectors of our immune response against tumors and exert their antitumor effects upon recognizing a variety of tumor-specific peptides presented by HLA molecules on the surface of tumor cells. The identification of the tumor-specific antigens of a given tumor is not required for immune checkpoint therapy (ICT), which mainly reactivates existing tumor-specific T cells together with T cells of unknown specificities. To decrease the activation of non-tumor-specific T cells, active or passive immunizations against tumor-specific antigens are considered. These immunizations require the identification of at least some of the tumor-specific antigens displayed on the tumor cells of a patient. While this has become an easy task for tumors with a large number of mutations generating neoantigens, it remains difficult for the remainder. Here, we review some facts about human tumor-specific or tumor-associated antigens, as well as some hopes for their future use in cancer immunotherapy.


Asunto(s)
Neoplasias , Linfocitos T , Humanos , Neoplasias/genética , Antígenos de Neoplasias , Inmunoterapia
19.
Int J Cancer ; 130(8): 1960-6, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21618521

RESUMEN

The human suppressive T cells that stably express transcription factor FOXP3, or regulatory T cells (Tregs), are thought to suppress antitumor immune responses. The most specific marker for human Tregs is the demethylation of CpG dinucleotides located in the first intron of FOXP3 (FOXP3i1). FOXP3i1 is completely methylated in other hematopoietic cells, including nonsuppressive T cells that transiently express FOXP3 after activation. Previously, we and others reported estimations of the frequency of Tregs in the blood of melanoma patients using a FOXP3i1 methylation-specific qPCR assay. Here, we attempted to quantify Tregs inside tumor samples using this assay. However, we found demethylated FOXP3i1 sequences in the melanoma cells themselves. This demethylation was not associated with substantial FOXP3 mRNA or protein expression, even though the demethylation extended to the promoter and terminal regions of the gene in some melanoma cells. Our results imply that analyzing Treg frequencies by quantification of demethylated FOXP3i1 will require that tumor-infiltrating T cells be separated from melanoma cells.


Asunto(s)
Metilación de ADN , Factores de Transcripción Forkhead/genética , Melanoma/genética , Linfocitos T Reguladores/metabolismo , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Islas de CpG/genética , Epigénesis Genética , Femenino , Factores de Transcripción Forkhead/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Intrones/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Masculino , Melanoma/metabolismo , Melanoma/patología , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína Smad2/genética , Proteína Smad2/metabolismo , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Linfocitos T Reguladores/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología
20.
Clin Dev Immunol ; 2012: 586314, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22400038

RESUMEN

Adoptive therapy with TCR gene-engineered T cells provides an attractive and feasible treatment option for cancer patients. Further development of TCR gene therapy requires the implementation of T-cell target epitopes that prevent "on-target" reactivity towards healthy tissues and at the same time direct a clinically effective response towards tumor tissues. Candidate epitopes that meet these criteria are MAGE-C2(336-344)/HLA-A2 (MC2/A2) and MAGE-A3(243-258)/HLA-DP4 (MA3/DP4). We molecularly characterized TCRαß genes of an MC2/A2-specific CD8 and MA3/DP4-specific CD4 T-cell clone derived from melanoma patients who responded clinically to MAGE vaccination. We identified MC2/A2 and MA3/DP4-specific TCR-Vα3/Vß28 and TCR-Vα38/Vß2 chains and validated these TCRs in vitro upon gene transfer into primary human T cells. The MC2 and MA3 TCR were surface-expressed and mediated CD8 T-cell functions towards melanoma cell lines and CD4 T-cell functions towards dendritic cells, respectively. We intend to start testing these MAGE-specific TCRs in phase I clinical trial.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Inmunoterapia Adoptiva , Melanoma/terapia , Neoplasias Cutáneas/terapia , Piel/inmunología , 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 , Ingeniería Celular , Línea Celular Tumoral , Ensayos Clínicos Fase I como Asunto , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/inmunología , Técnicas de Transferencia de Gen , Antígeno HLA-A2/genética , Antígeno HLA-A2/inmunología , Cadenas beta de HLA-DP/genética , Cadenas beta de HLA-DP/inmunología , Humanos , Melanoma/inmunología , Melanoma/patología , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Piel/efectos de los fármacos , Piel/patología , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA