Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Immunology ; 166(4): 429-443, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35470422

RESUMEN

The coronavirus disease 2019 (COVID-19) pandemic has created unprecedented challenges worldwide. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes COVID-19 and has a complex interaction with the immune system, including growing evidence of sex-specific differences in the immune response. Sex-disaggregated analyses of epidemiological data indicate that males experience more severe symptoms and suffer higher mortality from COVID-19 than females. Many behavioural risk factors and biological factors may contribute to the different immune response. This review examines the immune response to SARS-CoV-2 infection in the context of sex, with emphasis on potential biological mechanisms explaining differences in clinical outcomes. Understanding sex differences in the pathophysiology of SARS-CoV-2 infection will help promote the development of specific strategies to manage the disease.


Asunto(s)
COVID-19 , Femenino , Humanos , Inmunidad , Masculino , Pandemias , Factores de Riesgo , SARS-CoV-2 , Factores Sexuales
2.
Mutagenesis ; 30(2): 205-11, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25688113

RESUMEN

The clinical benefits obtained with rituximab in the treatment of CD20(+) B-cell malignancies and of imatinib in the treatment of Phi(+) leukaemias have opened a new era in oncology, transforming the concepts of tumour-targeted therapies and personalised medicine into reality. Since then, many tumour-targeted monoclonal antibodies and tyrosine kinase inhibitors have been approved for the treatment of cancers. Compared to conventional chemotherapies, these new drugs have more specificity against cancer cells and less systemic toxicities. However, like conventional chemotherapies, they often provide limited therapeutic benefits with short-lasting tumour responses as the vast majority of cancers become resistant to these drugs over time. Therefore, tumour-targeted therapies are an incremental innovation as compared to historical chemotherapies. Recently, a paradigm shift has been brought to the clinic with drugs targeting immune cells rather than cancer cells with the aim of stimulating the anti-tumour immune response of patients against their own cancer. Immunomodulatory drugs such as anti-CTLA4 and anti-PD-1 have generated long-lasting tumour responses when used as single agent in patients with refractory/relapsing cancers such as metastatic melanomas, renal cell carcinoma or non-small-cell lung carcinoma. These new immune-targeted therapies are therefore a disruptive innovation in cancer treatment: they demonstrate that long-lasting clinical benefits could be obtained by targeting molecules involved in the immune tolerance of cancer cells rather than by targeting oncogenic drivers or antigens expressed by cancer cells.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Factores Inmunológicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Anticuerpos Monoclonales/inmunología , Antineoplásicos/inmunología , Humanos , Sistema Inmunológico/efectos de los fármacos , Neoplasias/inmunología
3.
Elife ; 122023 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-38127790

RESUMEN

Glioblastoma (GBM) harbors a highly immunosuppressive tumor microenvironment (TME) which influences glioma growth. Major efforts have been undertaken to describe the TME on a single-cell level. However, human data on regional differences within the TME remain scarce. Here, we performed high-depth single-cell RNA sequencing (scRNAseq) on paired biopsies from the tumor center, peripheral infiltration zone and blood of five primary GBM patients. Through analysis of >45,000 cells, we revealed a regionally distinct transcription profile of microglia (MG) and monocyte-derived macrophages (MdMs) and an impaired activation signature in the tumor-peripheral cytotoxic-cell compartment. Comparing tumor-infiltrating CD8+ T cells with circulating cells identified CX3CR1high and CX3CR1int CD8+ T cells with effector and memory phenotype, respectively, enriched in blood but absent in the TME. Tumor CD8+ T cells displayed a tissue-resident memory phenotype with dysfunctional features. Our analysis provides a regionally resolved mapping of transcriptional states in GBM-associated leukocytes, serving as an additional asset in the effort towards novel therapeutic strategies to combat this fatal disease.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Humanos , Glioblastoma/genética , Glioblastoma/patología , Linfocitos T CD8-positivos , Macrófagos/patología , Glioma/genética , Leucocitos/patología , Microambiente Tumoral/genética , Neoplasias Encefálicas/patología
4.
Sci Transl Med ; 15(705): eadf5302, 2023 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-37467314

RESUMEN

Glioblastoma (GBM) is the most aggressive form of primary brain tumor, for which effective therapies are urgently needed. Cancer cells are capable of evading clearance by phagocytes such as microglia- and monocyte-derived cells through engaging tolerogenic programs. Here, we found that high expression of sialic acid-binding immunoglobulin-like lectin 9 (Siglec-9) correlates with reduced survival in patients with GBM. Using microglia- and monocyte-derived cell-specific knockouts of Siglec-E, the murine functional homolog of Siglec-9, together with single-cell RNA sequencing, we demonstrated that Siglec-E inhibits phagocytosis by these cells, thereby promoting immune evasion. Loss of Siglec-E on monocyte-derived cells further enhanced antigen cross-presentation and production of pro-inflammatory cytokines, which resulted in more efficient T cell priming. This bridging of innate and adaptive responses delayed tumor growth and resulted in prolonged survival in murine models of GBM. Furthermore, we showed the combinatorial activity of Siglec-E blockade and other immunotherapies demonstrating the potential for targeting Siglec-9 as a treatment for patients with GBM.


Asunto(s)
Glioblastoma , Ácido N-Acetilneuramínico , Humanos , Ratones , Animales , Ácido N-Acetilneuramínico/metabolismo , Glioblastoma/metabolismo , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/metabolismo , Fagocitosis/fisiología , Microglía/metabolismo
5.
Front Immunol ; 13: 890517, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35711466

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its associated symptoms, named coronavirus disease 2019 (COVID-19), have rapidly spread worldwide, resulting in the declaration of a pandemic. When several countries began enacting quarantine and lockdown policies, the pandemic as it is now known truly began. While most patients have minimal symptoms, approximately 20% of verified subjects are suffering from serious medical consequences. Co-existing diseases, such as cardiovascular disease, cancer, diabetes, and others, have been shown to make patients more vulnerable to severe outcomes from COVID-19 by modulating host-viral interactions and immune responses, causing severe infection and mortality. In this review, we outline the putative signaling pathways at the interface of COVID-19 and several diseases, emphasizing the clinical and molecular implications of concurring diseases in COVID-19 clinical outcomes. As evidence is limited on co-existing diseases and COVID-19, most findings are preliminary, and further research is required for optimal management of patients with comorbidities.


Asunto(s)
COVID-19 , COVID-19/epidemiología , Control de Enfermedades Transmisibles , Humanos , Pandemias , Cuarentena , SARS-CoV-2
6.
Sci Adv ; 8(26): eabn9440, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35776791

RESUMEN

A patient-tailored, ex vivo drug response platform for glioblastoma (GBM) would facilitate therapy planning, provide insights into treatment-induced mechanisms in the immune tumor microenvironment (iTME), and enable the discovery of biomarkers of response. We cultured regionally annotated GBM explants in perfusion bioreactors to assess iTME responses to immunotherapy. Explants were treated with anti-CD47, anti-PD-1, or their combination, and analyzed by multiplexed microscopy [CO-Detection by indEXing (CODEX)], enabling the spatially resolved identification of >850,000 single cells, accompanied by explant secretome interrogation. Center and periphery explants differed in their cell type and soluble factor composition, and responses to immunotherapy. A subset of explants displayed increased interferon-γ levels, which correlated with shifts in immune cell composition within specified tissue compartments. Our study demonstrates that ex vivo immunotherapy of GBM explants enables an active antitumoral immune response within the tumor center and provides a framework for multidimensional personalized assessment of tumor response to immunotherapy.

7.
J Immunother Cancer ; 8(1)2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32102830

RESUMEN

Intratumoral delivery of viruses and virus-associated molecular patterns can achieve antitumor effects that are largely mediated by the elicitation or potentiation of immune responses against the malignancy. Attenuated vaccines are approved and marketed as good manufactiring practice (GMP)-manufactured agents whose administration might be able to induce such effects. Recent reports in mouse transplantable tumor models indicate that the rotavirus, influenza and yellow fever vaccines can be especially suitable to elicit powerful antitumor immunity against cancer following intratumoral administration. These results highlight that intratumoral anti-infectious vaccines can turn cold tumors into hot, and underscore the key role played by virus-induced type I interferon pathways to overcome resistance to immune checkpoint-targeted antibodies.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Inhibidores de Puntos de Control Inmunológico/farmacología , Inmunoterapia/métodos , Neoplasias/terapia , Vacunas Virales/administración & dosificación , Animales , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Reposicionamiento de Medicamentos , Resistencia a Antineoplásicos/inmunología , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunogenicidad Vacunal , Inyecciones Intralesiones , Interferón Tipo I/inmunología , Ratones , Neoplasias/inmunología , Resultado del Tratamiento , Microambiente Tumoral/inmunología , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Vacunas Virales/inmunología
8.
Front Immunol ; 11: 571951, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33117364

RESUMEN

Tumor-associated microglia (MG) and macrophages (MΦ) are important components of the glioblastoma (GBM) immune tumor microenvironment (iTME). From the recent advances in understanding how MG and GBM cells evolve and interact during tumorigenesis, we emphasize the cooperation of MG with other immune cell types of the GBM-iTME, mainly MΦ and T cells. We provide a comprehensive overview of current immunotherapeutic clinical trials and approaches for the treatment of GBM, which in general, underestimate the counteracting contribution of immunosuppressive MG as a main factor for treatment failure. Furthermore, we summarize new developments and strategies in MG reprogramming/re-education in the GBM context, with a focus on ways to boost MG-mediated tumor cell phagocytosis and associated experimental models and methods. This ultimately converges in our proposal of novel combinatorial regimens that locally modulate MG as a central paradigm, and therefore may lead to additional, long-lasting, and effective tumoricidal responses.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Inmunoterapia/métodos , Microglía/fisiología , Animales , Neoplasias Encefálicas/inmunología , Reprogramación Celular , Ensayos Clínicos como Asunto , Glioblastoma/inmunología , Humanos , Fagocitosis , Microambiente Tumoral
9.
Front Immunol ; 10: 168, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30800125

RESUMEN

Therapeutic monoclonal antibodies targeting immune checkpoints (ICPs) have changed the treatment landscape of many tumors. However, response rate remains relatively low in most cases. A major factor involved in initial resistance to ICP inhibitors is the lack or paucity of tumor T cell infiltration, characterizing the so-called "cold tumors." In this review, we describe the main mechanisms involved in the absence of T cell infiltration, including lack of tumor antigens, defect in antigen presentation, absence of T cell activation and deficit of homing into the tumor bed. We discuss then the different therapeutic approaches that could turn cold into hot tumors. In this way, specific therapies are proposed according to their mechanism of action. In addition, ''supra-physiological'' therapies, such as T cell recruiting bispecific antibodies and Chimeric Antigen Receptor (CAR) T cells, may be active regardless of the mechanism involved, especially in MHC class I negative tumors. The determination of the main factors implicated in the lack of preexisting tumor T cell infiltration is crucial for the development of adapted algorithms of treatments for cold tumors.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Vacunas contra el Cáncer/uso terapéutico , Inmunoterapia Adoptiva , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias/terapia , Viroterapia Oncolítica , Células Presentadoras de Antígenos/inmunología , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Citocinas/metabolismo , Citocinas/uso terapéutico , Humanos , Activación de Linfocitos , Neoplasias/inmunología , Microambiente Tumoral/inmunología
10.
Sci Transl Med ; 11(515)2019 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-31645452

RESUMEN

Although immune checkpoint-targeted therapies are currently revolutionizing cancer care, only a minority of patients develop durable objective responses to anti-PD-1, PD-L1, and CTLA-4 therapy. Therefore, new therapeutic interventions are needed to increase the immunogenicity of tumors and overcome the resistance to these immunotherapies. Oncolytic properties of common viruses can be exploited for the priming of antitumor immunity, and such oncolytic viruses are currently in active clinical development in combination with immune checkpoint-targeted therapies. However, the routine implementation of these therapies is limited by their manufacturing constraints, the risk of exposure of clinical staff, and the ongoing regulations on genetically modified organisms. We sought to determine whether anti-infectious disease vaccines could be used as a commercially available source of immunostimulatory agents for cancer immunotherapy. We found that rotavirus vaccines have both immunostimulatory and oncolytic properties. In vitro, they can directly kill cancer cells with features of immunogenic cell death. In vivo, intratumoral rotavirus therapy has antitumor effects that are dependent on the immune system. In several immunocompetent murine tumor models, intratumoral rotavirus overcomes resistance to and synergizes with immune checkpoint-targeted therapy. Heat- and UV-inactivated rotavirus lost their oncolytic activity but kept their synergy with immune checkpoint-targeted antibodies through the up-regulation of the double-stranded RNA receptor retinoic acid-induced gene 1 (RIG-I). Rotavirus vaccines are clinical-grade products used in pediatric and adult populations. Therefore, in situ immunization strategies with intratumoral-attenuated rotavirus could be implemented quickly in the clinic.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Inmunoterapia/métodos , Vacunas contra Rotavirus/uso terapéutico , Adenosina Trifosfato/metabolismo , Animales , Línea Celular , Proteína 58 DEAD Box/metabolismo , Femenino , Citometría de Flujo , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , FN-kappa B/metabolismo , Receptores Inmunológicos
11.
Eur J Cancer ; 58: 8-16, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26922168

RESUMEN

PURPOSE: Well-documented stability data of monoclonal antibodies are generally missing. That is why we studied the physicochemical and biological stability of undiluted ipilimumab (IPI) in glass vial (5 mg/ml) over 28 d after opening, stored at 4 °C and 25 °C. METHOD: A stressed study (60 °C) was performed to validate our analytical methods as 'stability indicating'. The different methods used were turbidimetry, dynamic light scattering (DLS), second-derivative ultraviolet and chromatographic methods as size-exclusion chromatography (SEC) and cation-exchange (CEX). Biological characterisation was performed by an in vitro functional binding inhibition bioassay. RESULTS: We demonstrated that ipilimumab in opened vials stored at 4 °C and 25 °C remained stable for at least 28 d. No physical, chemical or structural instability was found. No aggregation was observed by turbidimetry, SEC and DLS. Hydrodynamic diameters remained unchanged, as chromatographic profiles in CEX and thermal aggregation curves. Functionally, the ability of IPI to antagonise CTLA-4/B7.2 binding remained stable over 1 month at 4 °C. CONCLUSION: These results indicate that unused residues of IPI in their original vials can be safely kept up to 28 d, following good manufacturing procedures, allowing re-use for another patient or in case of cold-chain rupture.


Asunto(s)
Anticuerpos Monoclonales/química , Antineoplásicos/química , Embalaje de Medicamentos , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Antígeno B7-2/metabolismo , Unión Competitiva , Antígeno CTLA-4/antagonistas & inhibidores , Antígeno CTLA-4/metabolismo , Química Farmacéutica , Cromatografía en Gel , Cromatografía por Intercambio Iónico , Estabilidad de Medicamentos , Vidrio , Hibridomas/metabolismo , Ipilimumab , Luz , Nefelometría y Turbidimetría , Unión Proteica , Desnaturalización Proteica , Estabilidad Proteica , Dispersión de Radiación , Espectrofotometría Ultravioleta , Tecnología Farmacéutica/métodos , Temperatura , Factores de Tiempo
12.
Sci Transl Med ; 7(283): 283ra55, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25877893

RESUMEN

The immunosurveillance mechanisms governing high-risk neuroblastoma (HR-NB), a major pediatric malignancy, have been elusive. We identify a potential role for natural killer (NK) cells, in particular the interaction between the NK receptor NKp30 and its ligand, B7-H6, in the metastatic progression and survival of HR-NB after myeloablative multimodal chemotherapy and stem cell transplantation. NB cells expressing the NKp30 ligand B7-H6 stimulated NK cells in an NKp30-dependent manner. Serum concentration of soluble B7-H6 correlated with the down-regulation of NKp30, bone marrow metastases, and chemoresistance, and soluble B7-H6 contained in the serum of HR-NB patients inhibited NK cell functions in vitro. The expression of distinct NKp30 isoforms affecting the polarization of NK cell functions correlated with 10-year event-free survival in three independent cohorts of HR-NB in remission from metastases after induction chemotherapy (n = 196, P < 0.001), adding prognostic value to known risk factors such as N-Myc amplification and age >18 months. We conclude that the interaction between NKp30 and B7-H6 may contribute to the fate of NB patients and that both the expression of NKp30 isoforms on circulating NK cells and the concentration of soluble B7-H6 in the serum may be clinically useful as biomarkers for risk stratification.


Asunto(s)
Antígenos B7/metabolismo , Neoplasias Encefálicas/metabolismo , Regulación Neoplásica de la Expresión Génica , Receptor 3 Gatillante de la Citotoxidad Natural/metabolismo , Neuroblastoma/metabolismo , Adolescente , Adulto , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor , Neoplasias Encefálicas/mortalidad , Línea Celular Tumoral , Niño , Preescolar , Supervivencia sin Enfermedad , Humanos , Lactante , Células Jurkat , Ligandos , Metástasis de la Neoplasia , Neuroblastoma/mortalidad , Fenotipo , Pronóstico , Estudios Prospectivos , Unión Proteica , Factores de Riesgo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA