Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Immunity ; 45(4): 931-943, 2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27717798

RESUMEN

The efficacy of the anti-cancer immunomodulatory agent cyclophosphamide (CTX) relies on intestinal bacteria. How and which relevant bacterial species are involved in tumor immunosurveillance, and their mechanism of action are unclear. Here, we identified two bacterial species, Enterococcus hirae and Barnesiella intestinihominis that are involved during CTX therapy. Whereas E. hirae translocated from the small intestine to secondary lymphoid organs and increased the intratumoral CD8/Treg ratio, B. intestinihominis accumulated in the colon and promoted the infiltration of IFN-γ-producing γδT cells in cancer lesions. The immune sensor, NOD2, limited CTX-induced cancer immunosurveillance and the bioactivity of these microbes. Finally, E. hirae and B. intestinihominis specific-memory Th1 cell immune responses selectively predicted longer progression-free survival in advanced lung and ovarian cancer patients treated with chemo-immunotherapy. Altogether, E. hirae and B. intestinihominis represent valuable "oncomicrobiotics" ameliorating the efficacy of the most common alkylating immunomodulatory compound.


Asunto(s)
Ciclofosfamida/farmacología , Enterococcus hirae/inmunología , Factores Inmunológicos/inmunología , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Animales , Colon/inmunología , Colon/microbiología , Memoria Inmunológica/inmunología , Inmunoterapia/métodos , Interferón gamma/inmunología , Intestino Delgado/inmunología , Intestino Delgado/microbiología , Ratones , Ratones Endogámicos C57BL , Monitorización Inmunológica , Proteína Adaptadora de Señalización NOD2/inmunología , Células TH1/inmunología
2.
BMC Cancer ; 14: 148, 2014 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-24589307

RESUMEN

BACKGROUND: The peritoneum is one of the most frequent sites of recurrent gastric carcinoma after curative treatment, despite the administration of pre- and/or postoperative systemic chemotherapy. Indeed, the prognosis of peritoneal carcinomatosis from gastric carcinoma continues to be poor, with a median survival of less than one year with systemic chemotherapy. Whereas the prognosis of peritoneal carcinomatosis from colorectal cancer has changed with the development of locally administered hyperthermic intraperitoneal chemotherapy (HIPEC), survival results following carcinomatosis from gastric cancer remain disappointing, yielding a 5-year survival rate of less than 20%. Innovative surgical therapies such as intraperitoneal immunotherapy therefore need to be developed for the immediate postoperative period after complete cytoreductive surgery. In a recent randomised study, a clinical effect was obtained after intraperitoneal infusion of catumaxomab in patients with malignant ascites, notably from gastric carcinoma. Catumaxomab, a nonhumanized chimeric antibody, is characterized by its unique ability to bind to three different types of cells: tumour cells expressing the epithelial cell adhesion molecule (EpCAM), T lymphocytes (CD3) and also accessory cells (Fcγ receptor). Because the peritoneum is an immunocompetent organ and up to 90% of gastric carcinomas express EpCAM, intraperitoneal infusion of catumaxomab after complete resection of all macroscopic disease (as defined in the treatment of carcinomatosis from colorectal cancer) could therefore efficiently treat microscopic residual disease. METHODS/DESIGN: The aim of this randomized phase II study is to assess 2-year overall survival after complete resection of limited carcinomatosis synchronous with gastric carcinoma, followed by an intraperitoneal infusion of catumaxomab with different total doses administered in each of the 2 arms. Close monitoring of peri-opertive mortality, morbidity and early surgical re-intervention will be done with stopping rules. Besides this analysis, translational research will be conducted to determine immunological markers of catumaxomab efficacy and to correlate these markers with clinical efficacy.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Carcinoma/tratamiento farmacológico , Carcinoma/cirugía , Protocolos Clínicos , Neoplasias Peritoneales/tratamiento farmacológico , Neoplasias Peritoneales/cirugía , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/cirugía , Anticuerpos Biespecíficos/administración & dosificación , Carcinoma/mortalidad , Humanos , Inmunoterapia , Infusiones Parenterales , Neoplasias Peritoneales/mortalidad
3.
Nat Med ; 12(2): 214-9, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16444265

RESUMEN

The interferon (IFN)-gamma-induced TRAIL effector mechanism is a vital component of cancer immunosurveillance by natural killer (NK) cells in mice. Here we show that the main source of IFN-gamma is not the conventional NK cell but a subset of B220(+)Ly6C(-) dendritic cells, which are atypical insofar as they express NK cell-surface molecules. Upon contact with a variety of tumor cells that are poorly recognized by NK cells, B220(+)NK1.1(+) dendritic cells secrete high levels of IFN-gamma and mediate TRAIL-dependent lysis of tumor cells. Adoptive transfer of these IFN-producing killer dendritic cells (IKDCs) into tumor-bearing Rag2(-/-)Il2rg(-/-) mice prevented tumor outgrowth, whereas transfer of conventional NK cells did not. In conclusion, we identified IKDCs as pivotal sensors and effectors of the innate antitumor immune response.


Asunto(s)
Células Dendríticas/clasificación , Células Dendríticas/inmunología , Neoplasias Experimentales/inmunología , Traslado Adoptivo , Animales , Presentación de Antígeno , Antígenos Ly , Antígenos de Superficie/metabolismo , Proteínas Reguladoras de la Apoptosis/inmunología , Antígeno CD11c/metabolismo , Línea Celular Tumoral , Citotoxicidad Inmunológica , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Células Dendríticas/ultraestructura , Femenino , Interferón gamma/biosíntesis , Subunidad gamma Común de Receptores de Interleucina , Células Asesinas Naturales/inmunología , Lectinas Tipo C/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Microscopía Electrónica , Subfamilia B de Receptores Similares a Lectina de Células NK , Receptores de Interleucina/deficiencia , Receptores de Interleucina/genética , Ligando Inductor de Apoptosis Relacionado con TNF , Factor de Necrosis Tumoral alfa/inmunología
4.
Oncoimmunology ; 12(1): 2163785, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36632566

RESUMEN

The SARS-CoV-2 pandemic still represents a threat for immunosuppressed and hematological malignancy (HM) bearing patients, causing increased morbidity and mortality. Given the low anti-SARSCoV-2 IgG titers post-vaccination, the COVID-19 threat prompted the prophylactic use of engineered anti-SARS-CoV-2 monoclonal antibodies. In addition, potential clinical significance of T cell responses has been overlooked during the first waves of the pandemic, calling for additional in-depth studies. We reported that the polarity and the repertoire of T cell immune responses govern the susceptibility to SARS-CoV-2 infection in health care workers and solid cancer patients. Here, we longitudinally analyzed humoral and cellular immune responses at each BNT162b2 mRNA vaccine injection in 47 HM patients under therapy. Only one-third of HM, mostly multiple myeloma (MM) bearing patients, could mount S1-RBD-specific IgG responses following BNT162b2 mRNA vaccines. This vaccine elicited a S1-RBD-specific Th1 immune response in about 20% patients, mostly in MM and Hodgkin lymphoma, while exacerbating Th2 responses in the 10% cases that presented this recognition pattern at baseline (mostly rituximab-treated patients). Performing a third booster barely improved the percentage of patients developing an S1-RBD-specific Th1 immunity and failed to seroconvert additional HM patients. Finally, 16 patients were infected with SARS-CoV-2, of whom 6 developed a severe infection. Only S1-RBD-specific Th1 responses were associated with protection against SARS-CoV2 infection, while Th2 responses or anti-S1-RBD IgG titers failed to correlate with protection. These findings herald the paramount relevance of vaccine-induced Th1 immune responses in hematological malignancies.


Asunto(s)
COVID-19 , Neoplasias Hematológicas , Mieloma Múltiple , Vacunas , Humanos , COVID-19/prevención & control , Vacuna BNT162 , SARS-CoV-2 , ARN Viral , Neoplasias Hematológicas/complicaciones , Anticuerpos Antivirales , Inmunoglobulina G
5.
Cancer Discov ; 12(4): 958-983, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35179201

RESUMEN

Vaccination against coronavirus disease 2019 (COVID-19) relies on the in-depth understanding of protective immune responses to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). We characterized the polarity and specificity of memory T cells directed against SARS-CoV-2 viral lysates and peptides to determine correlates with spontaneous, virus-elicited, or vaccine-induced protection against COVID-19 in disease-free and cancer-bearing individuals. A disbalance between type 1 and 2 cytokine release was associated with high susceptibility to COVID-19. Individuals susceptible to infection exhibited a specific deficit in the T helper 1/T cytotoxic 1 (Th1/Tc1) peptide repertoire affecting the receptor binding domain of the spike protein (S1-RBD), a hotspot of viral mutations. Current vaccines triggered Th1/Tc1 responses in only a fraction of all subject categories, more effectively against the original sequence of S1-RBD than that from viral variants. We speculate that the next generation of vaccines should elicit Th1/Tc1 T-cell responses against the S1-RBD domain of emerging viral variants. SIGNIFICANCE: This study prospectively analyzed virus-specific T-cell correlates of protection against COVID-19 in healthy and cancer-bearing individuals. A disbalance between Th1/Th2 recall responses conferred susceptibility to COVID-19 in both populations, coinciding with selective defects in Th1 recognition of the receptor binding domain of spike. See related commentary by McGary and Vardhana, p. 892. This article is highlighted in the In This Issue feature, p. 873.


Asunto(s)
Factores de Restricción Antivirales , COVID-19 , Neoplasias , Linfocitos T , Anticuerpos Neutralizantes , Factores de Restricción Antivirales/inmunología , COVID-19/inmunología , Humanos , Neoplasias/complicaciones , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/genética , Linfocitos T/inmunología
6.
Cancer Discov ; 12(10): 2280-2307, 2022 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-35929803

RESUMEN

Biomarkers guiding the neoadjuvant use of immune-checkpoint blockers (ICB) are needed for patients with localized muscle-invasive bladder cancers (MIBC). Profiling tumor and blood samples, we found that follicular helper CD4+ T cells (TFH) are among the best therapeutic targets of pembrolizumab correlating with progression-free survival. TFH were associated with tumoral CD8 and PD-L1 expression at baseline and the induction of tertiary lymphoid structures after pembrolizumab. Blood central memory TFH accumulated in tumors where they produce CXCL13, a chemokine found in the plasma of responders only. IgG4+CD38+ TFH residing in bladder tissues correlated with clinical benefit. Finally, TFH and IgG directed against urothelium-invasive Escherichia coli dictated clinical responses to pembrolizumab in three independent cohorts. The links between tumor infection and success of ICB immunomodulation should be prospectively assessed at a larger scale. SIGNIFICANCE: In patients with bladder cancer treated with neoadjuvant pembrolizumab, E. coli-specific CXCL13 producing TFH and IgG constitute biomarkers that predict clinical benefit. Beyond its role as a biomarker, such immune responses against E. coli might be harnessed for future therapeutic strategies. This article is highlighted in the In This Issue feature, p. 2221.


Asunto(s)
Neoplasias de la Vejiga Urinaria , Antígeno B7-H1 , Quimiocina CXCL13 , Escherichia coli , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoglobulina G , Músculos , Terapia Neoadyuvante , Receptor de Muerte Celular Programada 1 , Linfocitos T Colaboradores-Inductores , Resultado del Tratamiento , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico
7.
J Exp Med ; 202(8): 1075-85, 2005 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-16230475

RESUMEN

Tumor growth promotes the expansion of CD4+CD25+ regulatory T (T reg) cells that counteract T cell-mediated immune responses. An inverse correlation between natural killer (NK) cell activation and T reg cell expansion in tumor-bearing patients, shown here, prompted us to address the role of T reg cells in controlling innate antitumor immunity. Our experiments indicate that human T reg cells expressed membrane-bound transforming growth factor (TGF)-beta, which directly inhibited NK cell effector functions and down-regulated NKG2D receptors on the NK cell surface. Adoptive transfer of wild-type T reg cells but not TGF-beta-/- T reg cells into nude mice suppressed NK cell-mediated cytotoxicity, reduced NKG2D receptor expression, and accelerated the growth of tumors that are normally controlled by NK cells. Conversely, the depletion of mouse T reg cells exacerbated NK cell proliferation and cytotoxicity in vivo. Human NK cell-mediated tumor recognition could also be restored by depletion of T reg cells from tumor-infiltrating lymphocytes. These findings support a role for T reg cells in blunting the NK cell arm of the innate immune system.


Asunto(s)
Inmunidad Innata/inmunología , Células Asesinas Naturales/inmunología , Activación de Linfocitos/inmunología , Neoplasias/metabolismo , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Citocinas/metabolismo , Pruebas Inmunológicas de Citotoxicidad , Citometría de Flujo , Francia , Humanos , Ratones , Ratones Endogámicos C57BL , Subfamilia K de Receptores Similares a Lectina de Células NK , Receptores Inmunológicos/metabolismo , Receptores de Células Asesinas Naturales , Linfocitos T Reguladores/metabolismo , Factor de Crecimiento Transformador beta/inmunología
8.
Cytokine Growth Factor Rev ; 19(1): 79-92, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18155952

RESUMEN

Tumor growth results from a delicate balance between intrinsic dysregulation of oncogenes, tumor suppressor and stability genes counteracted by extrinsic defenses composed of immune cells shaping tumor immunogenicity. Although immune subversion might be the ultimate outcome of this process, a complex network of cellular interactions take place eventually leading to tumor specific cognate immune responses. The links between innate and cognate antitumor immunity eliciting protective T cell responses are instigated by cytokines, chemokines and damage associated molecular patterns. The intricate differentiation pathway whereby dendritic cells could undergo an efficient maturation program in the tumor microenvironment appears crucial. We will discuss the role of innate effectors and cancer therapies in the process of defense against tumor cells.


Asunto(s)
Células Dendríticas/inmunología , Inmunidad Innata/inmunología , Neoplasias/inmunología , Apoptosis/efectos de los fármacos , Benzamidas , Biomarcadores de Tumor/metabolismo , Vacunas contra el Cáncer/uso terapéutico , Islas de CpG , Citocinas/fisiología , Proteína Ligando Fas/fisiología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Proteína HMGB1/metabolismo , Humanos , Mesilato de Imatinib , Interferón Tipo I/fisiología , Interferón gamma/fisiología , Interferones/biosíntesis , Interleucina-15/fisiología , Células Asesinas Naturales/inmunología , Oligodesoxirribonucleótidos/uso terapéutico , Piperazinas/uso terapéutico , Pirimidinas/uso terapéutico , Receptor Cross-Talk/fisiología , Linfocitos T Colaboradores-Inductores/inmunología , Ácido Úrico/metabolismo
9.
Cell Death Differ ; 28(12): 3297-3315, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34230615

RESUMEN

Patients with cancer are at higher risk of severe coronavirus infectious disease 2019 (COVID-19), but the mechanisms underlying virus-host interactions during cancer therapies remain elusive. When comparing nasopharyngeal swabs from cancer and noncancer patients for RT-qPCR cycle thresholds measuring acute respiratory syndrome coronavirus-2 (SARS-CoV-2) in 1063 patients (58% with cancer), we found that malignant disease favors the magnitude and duration of viral RNA shedding concomitant with prolonged serum elevations of type 1 IFN that anticorrelated with anti-RBD IgG antibodies. Cancer patients with a prolonged SARS-CoV-2 RNA detection exhibited the typical immunopathology of severe COVID-19 at the early phase of infection including circulation of immature neutrophils, depletion of nonconventional monocytes, and a general lymphopenia that, however, was accompanied by a rise in plasmablasts, activated follicular T-helper cells, and non-naive Granzyme B+FasL+, EomeshighTCF-1high, PD-1+CD8+ Tc1 cells. Virus-induced lymphopenia worsened cancer-associated lymphocyte loss, and low lymphocyte counts correlated with chronic SARS-CoV-2 RNA shedding, COVID-19 severity, and a higher risk of cancer-related death in the first and second surge of the pandemic. Lymphocyte loss correlated with significant changes in metabolites from the polyamine and biliary salt pathways as well as increased blood DNA from Enterobacteriaceae and Micrococcaceae gut family members in long-term viral carriers. We surmise that cancer therapies may exacerbate the paradoxical association between lymphopenia and COVID-19-related immunopathology, and that the prevention of COVID-19-induced lymphocyte loss may reduce cancer-associated death.


Asunto(s)
COVID-19/complicaciones , COVID-19/virología , Linfopenia/complicaciones , Neoplasias/complicaciones , ARN Viral/análisis , SARS-CoV-2/genética , Esparcimiento de Virus , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Cohortes , ADN Bacteriano/sangre , Enterobacteriaceae/genética , Femenino , Humanos , Interferón Tipo I/sangre , Linfopenia/virología , Masculino , Micrococcaceae/genética , Persona de Mediana Edad , Nasofaringe/virología , Neoplasias/diagnóstico , Neoplasias/mortalidad , Pandemias , Pronóstico , Factores de Tiempo , Adulto Joven
10.
Nat Med ; 26(6): 919-931, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32451498

RESUMEN

The prognosis of colon cancer (CC) is dictated by tumor-infiltrating lymphocytes, including follicular helper T (TFH) cells and the efficacy of chemotherapy-induced immune responses. It remains unclear whether gut microbes contribute to the elicitation of TFH cell-driven responses. Here, we show that the ileal microbiota dictates tolerogenic versus immunogenic cell death of ileal intestinal epithelial cells (IECs) and the accumulation of TFH cells in patients with CC and mice. Suppression of IEC apoptosis led to compromised chemotherapy-induced immunosurveillance against CC in mice. Protective immune responses against CC were associated with residence of Bacteroides fragilis and Erysipelotrichaceae in the ileum. In the presence of these commensals, apoptotic ileal IECs elicited PD-1+ TFH cells in an interleukin-1R1- and interleukin-12-dependent manner. The ileal microbiome governed the efficacy of chemotherapy and PD-1 blockade in CC independently of microsatellite instability. These findings demonstrate that immunogenic ileal apoptosis contributes to the prognosis of chemotherapy-treated CC.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Microbioma Gastrointestinal/inmunología , Íleon/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Oxaliplatino/farmacología , Adenocarcinoma/inmunología , Adenocarcinoma/microbiología , Adenocarcinoma/patología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Antineoplásicos/uso terapéutico , Apoptosis/inmunología , Bacteroides fragilis , Línea Celular Tumoral , Neoplasias del Colon/inmunología , Neoplasias del Colon/microbiología , Neoplasias del Colon/patología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Células Epiteliales/patología , Femenino , Firmicutes , Microbioma Gastrointestinal/fisiología , Humanos , Íleon/inmunología , Íleon/microbiología , Íleon/patología , Muerte Celular Inmunogénica/efectos de los fármacos , Muerte Celular Inmunogénica/inmunología , Vigilancia Inmunológica/efectos de los fármacos , Vigilancia Inmunológica/inmunología , Interleucina-12/inmunología , Mucosa Intestinal , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Ratones , Persona de Mediana Edad , Oxaliplatino/uso terapéutico , Pronóstico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptores Tipo I de Interleucina-1/inmunología , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Linfocitos T Colaboradores-Inductores/inmunología
11.
J Mol Med (Berl) ; 85(10): 1069-76, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17891368

RESUMEN

The treatment of cancer by chemotherapy causes tumour cell death, mostly by apoptosis. This tumour cell death may or may not elicit an immune response. At least in some cases, the efficacy of chemotherapy critically depends on the induction of immunogenic cell death that is a type of cell demise that stimulates the activation of an adaptative anti-tumour immune response, which in turn helps to eradicate residual cancer (stem) cells. Indeed, anthracyclins care more efficient in curing tumours in immunocompetent than in T cell-deficient mice. The molecular mechanism implicated in this anti-tumour T cell activation was recently discovered. Anthracyclins cause immunogenic cell death due to their specific capacity to stimulate the translocation of calreticulin to the cell surface. Calreticulin then acts as an "eat me" signal for dendritic cells, allowing them to phagocytose tumour cells and to prime tumour antigen-specific cytotoxic T cells. Importantly, non-immunogenic chemotherapy can be rendered immunogenic by adsorbing recombinant calreticulin to tumour cells or by enforcing the translocation of endogenous calreticulin to the cell surface by means of PP1/GADD34 inhibitors. This strategy could have major implications for the treatment of human cancer. Indeed, in vivo treatments with anthracyclins can cause the translocation of calreticulin to the surface of circulating tumour cells, in patients with acute myeloid leukaemia (AML). The challenge will be to determine whether the exposure of calreticulin translocation on the tumour cell surface is linked to chemotherapy-induced anti-tumour immune responses and therapeutic efficacy in human cancer.


Asunto(s)
Antígenos de Neoplasias/inmunología , Antígenos de Superficie/inmunología , Apoptosis/inmunología , Calreticulina/inmunología , Animales , Antraciclinas/farmacología , Antraciclinas/uso terapéutico , Antígenos de Neoplasias/efectos de los fármacos , Antígenos de Neoplasias/metabolismo , Antígenos de Superficie/efectos de los fármacos , Antígenos de Superficie/metabolismo , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Calreticulina/metabolismo , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/inmunología , Membrana Celular/metabolismo , Células Dendríticas/inmunología , Humanos , Ratones , Modelos Biológicos , Neoplasias Experimentales/inmunología , Linfocitos T Citotóxicos/inmunología
12.
Science ; 359(6371): 91-97, 2018 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-29097494

RESUMEN

Immune checkpoint inhibitors (ICIs) targeting the PD-1/PD-L1 axis induce sustained clinical responses in a sizable minority of cancer patients. We found that primary resistance to ICIs can be attributed to abnormal gut microbiome composition. Antibiotics inhibited the clinical benefit of ICIs in patients with advanced cancer. Fecal microbiota transplantation (FMT) from cancer patients who responded to ICIs into germ-free or antibiotic-treated mice ameliorated the antitumor effects of PD-1 blockade, whereas FMT from nonresponding patients failed to do so. Metagenomics of patient stool samples at diagnosis revealed correlations between clinical responses to ICIs and the relative abundance of Akkermansia muciniphila Oral supplementation with A. muciniphila after FMT with nonresponder feces restored the efficacy of PD-1 blockade in an interleukin-12-dependent manner by increasing the recruitment of CCR9+CXCR3+CD4+ T lymphocytes into mouse tumor beds.


Asunto(s)
Trasplante de Microbiota Fecal , Microbioma Gastrointestinal/inmunología , Inmunoterapia/métodos , Neoplasias/terapia , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Animales , Antibacterianos/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Antígenos CD4/inmunología , Heces/microbiología , Microbioma Gastrointestinal/genética , Humanos , Interleucina-12/inmunología , Metagenoma/genética , Ratones , Receptores CCR/inmunología , Receptores CXCR3/inmunología , Linfocitos T/inmunología , Verrucomicrobia/genética , Verrucomicrobia/inmunología
13.
J Clin Invest ; 114(3): 379-88, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15286804

RESUMEN

Mutant isoforms of the KIT or PDGF receptors expressed by gastrointestinal stromal tumors (GISTs) are considered the therapeutic targets for STI571 (imatinib mesylate; Gleevec), a specific inhibitor of these tyrosine kinase receptors. Case reports of clinical efficacy of Gleevec in GISTs lacking the typical receptor mutations prompted a search for an alternate mode of action. Here we show that Gleevec can act on host DCs to promote NK cell activation. DC-mediated NK cell activation was triggered in vitro and in vivo by treatment of DCs with Gleevec as well as by a loss-of-function mutation of KIT. Therefore, tumors that are refractory to the antiproliferative effects of Gleevec in vitro responded to Gleevec in vivo in an NK cell-dependent manner. Longitudinal studies of Gleevec-treated GIST patients revealed a therapy-induced increase in IFN-gamma production by NK cells, correlating with an enhanced antitumor response. These data point to a novel mode of antitumor action for Gleevec.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Células Asesinas Naturales/metabolismo , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Animales , Antineoplásicos/farmacología , Benzamidas , Estudios de Casos y Controles , Técnicas de Cocultivo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Femenino , Neoplasias Gastrointestinales/tratamiento farmacológico , Neoplasias Gastrointestinales/genética , Neoplasias Gastrointestinales/metabolismo , Neoplasias Gastrointestinales/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Mesilato de Imatinib , Interferón gamma/efectos de los fármacos , Interferón gamma/metabolismo , Leucocitos Mononucleares/metabolismo , Estudios Longitudinales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID , Mutación , Activación Neutrófila/efectos de los fármacos , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-kit/efectos de los fármacos , Pirimidinas/farmacología , Proteínas Tirosina Quinasas Receptoras/efectos de los fármacos , Proteínas Tirosina Quinasas Receptoras/genética , Receptores del Factor de Crecimiento Derivado de Plaquetas/efectos de los fármacos , Receptores del Factor de Crecimiento Derivado de Plaquetas/genética , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Células del Estroma/efectos de los fármacos
14.
J Leukoc Biol ; 80(3): 471-8, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16809645

RESUMEN

Exosomes are nanometer-sized membrane vesicles invaginating from multivesicular bodies and secreted from different cell types. They represent an "in vitro" discovery, but vesicles with the hallmarks of exosomes are present in vivo in germinal centers and biological fluids. Their protein and lipid composition is unique and could account for their expanding functions such as eradication of obsolete proteins, antigen presentation, or "Trojan horses" for viruses or prions. The potential of dendritic cell-derived exosomes (Dex) as cell-free cancer vaccines is addressed in this review. Lessons learned from the pioneering clinical trials allowed reassessment of the priming capacities of Dex in preclinical models, optimizing clinical protocols, and delineating novel, biological features of Dex in cancer patients.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Endosomas/inmunología , Neoplasias/tratamiento farmacológico , Animales , Vacunas contra el Cáncer/uso terapéutico , Ensayos Clínicos Fase I como Asunto , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Endosomas/química , Endosomas/metabolismo , Humanos , Inmunoterapia/métodos , Neoplasias/inmunología , Neoplasias/patología
15.
Cancer Res ; 65(14): 6409-17, 2005 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-16024645

RESUMEN

BCR/ABL fusion gene, encoding a paradigmatic tyrosine kinase involved in chronic myelogenous leukemia (CML), can modulate the expression of genes involved in natural killer (NK) cell target recognition. Recent reports outline the role of allogeneic antileukemic NK effectors in the graft-versus-leukemia effect but the regulation of NK cell activation in the setting of graft-versus-leukemia effect remains unknown. Here we show that dendritic cells derived from monocytes of CML patients are selectively endowed with NK cell stimulatory capacity in vitro. We further show, using a gene transfer approach in mouse bone marrow progenitors, that ABL/ABL is necessary to promote dendritic cell-mediated NK cell activation. The dendritic cell/NK cell cross-talk in ABL/ABL-induced CML seems unique because JunB or IFN consensus sequence binding protein loss of functions, associated with other myeloproliferative disorders, do not promote dendritic cell-mediated NK cell activation. NK cell activation by leukemic dendritic cells involves NKG2D activating receptors and is blocked by imatinib mesylate. Indeed, ABL/ABL translocation enhances the expression levels of the NKG2D ligands on dendritic cells, which is counteracted by imatinib mesylate. Altogether, the clonal ABL/ABL dendritic cells display the unique and selective ability to activate NK cells and may participate in the NK cell control of CML. This study also highlights the deleterious role of imatinib mesylate at the dendritic cell level for NK cell activation.


Asunto(s)
Células Dendríticas/inmunología , Proteínas de Fusión bcr-abl/inmunología , Células Asesinas Naturales/inmunología , Animales , Células de la Médula Ósea/inmunología , Femenino , Proteínas de Fusión bcr-abl/genética , Técnicas de Transferencia de Gen , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/inmunología , Ligandos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID , Subfamilia K de Receptores Similares a Lectina de Células NK , Receptores Inmunológicos/inmunología , Receptores de Células Asesinas Naturales , Translocación Genética
16.
J Clin Invest ; 126(3): 921-37, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26854930

RESUMEN

Melanoma prognosis is dictated by tumor-infiltrating lymphocytes, the migratory and functional behavior of which is guided by chemokine or cytokine gradients. Here, we retrospectively analyzed the expression patterns of 9 homing receptors (CCR/CXCR) in naive and memory CD4+ and CD8+ T lymphocytes in 57 patients with metastatic melanoma (MMel) with various sites of metastases to evaluate whether T cell CCR/CXCR expression correlates with intratumoral accumulation, metastatic progression, and/or overall survival (OS). Homing receptor expression on lymphocytes strongly correlated with MMel dissemination. Loss of CCR6 or CXCR3, but not cutaneous lymphocyte antigen (CLA), on circulating T cell subsets was associated with skin or lymph node metastases, loss of CXCR4, CXCR5, and CCR9 corresponded with lung involvement, and a rise in CCR10 or CD103 was associated with widespread dissemination. High frequencies of CD8+CCR9+ naive T cells correlated with prolonged OS, while neutralizing the CCR9/CCL25 axis in mice stimulated tumor progression. The expansion of CLA-expressing effector memory CD8+ T cells in response to a single administration of CTLA4 blockade predicted disease control at 3 months in 47 patients with MMel. Thus, specific CCR/CXCR expression patterns on circulating T lymphocytes may guide potential diagnostic and therapeutic approaches.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Pulmonares/metabolismo , Melanoma/metabolismo , Receptores de Quimiocina/metabolismo , Neoplasias Cutáneas/metabolismo , Linfocitos T/metabolismo , Adulto , Animales , Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Estudios de Casos y Controles , Línea Celular Tumoral , Femenino , Humanos , Ipilimumab , Estimación de Kaplan-Meier , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/secundario , Metástasis Linfática , Masculino , Melanoma/inmunología , Melanoma/mortalidad , Melanoma/secundario , Ratones Endogámicos C57BL , Persona de Mediana Edad , Trasplante de Neoplasias , Modelos de Riesgos Proporcionales , Curva ROC , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/mortalidad , Neoplasias Cutáneas/patología
17.
J Transl Med ; 3(1): 10, 2005 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-15740633

RESUMEN

BACKGROUND: DC derived-exosomes are nanomeric vesicles harboring functional MHC/peptide complexes capable of promoting T cell immune responses and tumor rejection. Here we report the feasability and safety of the first Phase I clinical trial using autologous exosomes pulsed with MAGE 3 peptides for the immunization of stage III/IV melanoma patients. Secondary endpoints were the monitoring of T cell responses and the clinical outcome. PATIENTS AND METHODS: Exosomes were purified from day 7 autologous monocyte derived-DC cultures. Fifteen patients fullfilling the inclusion criteria (stage IIIB and IV, HLA-A1+, or -B35+ and HLA-DPO4+ leukocyte phenotype, tumor expressing MAGE3 antigen) were enrolled from 2000 to 2002 and received four exosome vaccinations. Two dose levels of either MHC class II molecules (0.13 versus 0.40 x 1014 molecules) or peptides (10 versus 100 mug/ml) were tested. Evaluations were performed before and 2 weeks after immunization. A continuation treatment was performed in 4 cases of non progression. RESULTS: The GMP process allowed to harvest about 5 x 1014 exosomal MHC class II molecules allowing inclusion of all 15 patients. There was no grade II toxicity and the maximal tolerated dose was not achieved. One patient exhibited a partial response according to the RECIST criteria. This HLA-B35+/A2+ patient vaccinated with A1/B35 defined CTL epitopes developed halo of depigmentation around naevi, a MART1-specific HLA-A2 restricted T cell response in the tumor bed associated with progressive loss of HLA-A2 and HLA-BC molecules on tumor cells during therapy with exosomes. In addition, one minor, two stable and one mixed responses were observed in skin and lymph node sites. MAGE3 specific CD4+ and CD8+ T cell responses could not be detected in peripheral blood. CONCLUSION: The first exosome Phase I trial highlighted the feasibility of large scale exosome production and the safety of exosome administration.

18.
Science ; 350(6264): 1079-84, 2015 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-26541610

RESUMEN

Antibodies targeting CTLA-4 have been successfully used as cancer immunotherapy. We find that the antitumor effects of CTLA-4 blockade depend on distinct Bacteroides species. In mice and patients, T cell responses specific for B. thetaiotaomicron or B. fragilis were associated with the efficacy of CTLA-4 blockade. Tumors in antibiotic-treated or germ-free mice did not respond to CTLA blockade. This defect was overcome by gavage with B. fragilis, by immunization with B. fragilis polysaccharides, or by adoptive transfer of B. fragilis-specific T cells. Fecal microbial transplantation from humans to mice confirmed that treatment of melanoma patients with antibodies against CTLA-4 favored the outgrowth of B. fragilis with anticancer properties. This study reveals a key role for Bacteroidales in the immunostimulatory effects of CTLA-4 blockade.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Bacteroides/inmunología , Antígeno CTLA-4/antagonistas & inhibidores , Microbioma Gastrointestinal/inmunología , Melanoma/terapia , Neoplasias Cutáneas/terapia , Adulto , Anciano , Anciano de 80 o más Años , Animales , Antibacterianos/farmacología , Anticuerpos Monoclonales/efectos adversos , Antígeno CTLA-4/inmunología , Disbiosis/inmunología , Trasplante de Microbiota Fecal , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Vida Libre de Gérmenes/inmunología , Humanos , Memoria Inmunológica , Inmunoterapia , Intestinos/inmunología , Intestinos/microbiología , Ipilimumab , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Linfocitos T/inmunología
19.
Eur Cytokine Netw ; 13(1): 17-27, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-11956017

RESUMEN

Dendritic cells (DC) were originally found critical in the setting of cognate immune responses. We first demonstrated that DC can also induce mouse NK cell activation and NK cell dependent-antitumor effects in mice. Here we analyzed the dynamics between DC and NK cells in human in vitro model systems. In the absence of LPS, DC do not trigger resting NK cells. Conversely, in the presence of LPS, resting bulk NK cells interacting with DC acquire CD25 and CD69 surface expression, produce high levels of IFN-gamma and lyse DAUDI cells. On activated IL-2 dependent NK cell lines, regardless of their differentiation stage, DC maintain or enhance NK cell proliferation and effector functions in the absence of exogenous cytokines. While IL-12, IL-15 and IL-18 are not critical, a direct cell-to-cell contact is mandatory for NK activation by DC and required for optimal proliferation. These data imply that DC also modulate human NK cell innate effector functions.


Asunto(s)
Comunicación Celular/fisiología , Células Dendríticas/fisiología , Células Asesinas Naturales/fisiología , Monocitos/fisiología , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Diferenciación Celular/fisiología , División Celular/fisiología , Línea Celular , Células Dendríticas/efectos de los fármacos , Humanos , Interferón gamma/metabolismo , Células Asesinas Naturales/metabolismo , Lectinas Tipo C , Lipopolisacáridos/farmacología , Receptores de Interleucina-2/metabolismo , Células Tumorales Cultivadas
20.
Bull Cancer ; 90(8-9): 695-8, 2003.
Artículo en Francés | MEDLINE | ID: mdl-14609758

RESUMEN

Exosomes are 60 to 90 nm membrane vesicles originating from late endosomes and secreted from most hematopoietic and epithelial cells in vitro. B cell derived-exosome antigenicity was first reported in 1996 in MHC class II restricted CD4+ T lymphocytes. In 1998, we reported that dendritic cell derived-exosomes are immunogenic in mice leading to tumor rejection. These findings have renewed the interest in exosomes. The current challenge consists in understanding the mechanisms and the physiological relevance of exosomes that could contribute to the design of the optimal exosome based-vaccination. Here, we will focus on the biological features pertaining to dendritic cell- and tumor cell derived-exosomes and will discuss their potential clinical implementation.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Vesículas Citoplasmáticas/inmunología , Inmunoterapia/métodos , Melanoma/terapia , Neoplasias Cutáneas/terapia , Animales , Carcinoma de Pulmón de Células no Pequeñas/terapia , Vesículas Citoplasmáticas/química , Estudios de Factibilidad , Humanos , Neoplasias Pulmonares/terapia , Melanoma/inmunología , Ratones , Neoplasias Cutáneas/inmunología , Linfocitos T Citotóxicos/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA