Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Lancet ; 403(10435): 1460-1471, 2024 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-38554725

RESUMEN

BACKGROUND: Afamitresgene autoleucel (afami-cel) showed acceptable safety and promising efficacy in a phase 1 trial (NCT03132922). The aim of this study was to further evaluate the efficacy of afami-cel for the treatment of patients with HLA-A*02 and MAGE-A4-expressing advanced synovial sarcoma or myxoid round cell liposarcoma. METHODS: SPEARHEAD-1 was an open-label, non-randomised, phase 2 trial done across 23 sites in Canada, the USA, and Europe. The trial included three cohorts, of which the main investigational cohort (cohort 1) is reported here. Cohort 1 included patients with HLA-A*02, aged 16-75 years, with metastatic or unresectable synovial sarcoma or myxoid round cell liposarcoma (confirmed by cytogenetics) expressing MAGE-A4, and who had received at least one previous line of anthracycline-containing or ifosfamide-containing chemotherapy. Patients received a single intravenous dose of afami-cel (transduced dose range 1·0 × 109-10·0 × 109 T cells) after lymphodepletion. The primary endpoint was overall response rate in cohort 1, assessed by a masked independent review committee using Response Evaluation Criteria in Solid Tumours (version 1.1) in the modified intention-to-treat population (all patients who received afami-cel). Adverse events, including those of special interest (cytokine release syndrome, prolonged cytopenia, and neurotoxicity), were monitored and are reported for the modified intention-to-treat population. This trial is registered at ClinicalTrials.gov, NCT04044768; recruitment is closed and follow-up is ongoing for cohorts 1 and 2, and recruitment is open for cohort 3. FINDINGS: Between Dec 17, 2019, and July 27, 2021, 52 patients with cytogenetically confirmed synovial sarcoma (n=44) and myxoid round cell liposarcoma (n=8) were enrolled and received afami-cel in cohort 1. Patients were heavily pre-treated (median three [IQR two to four] previous lines of systemic therapy). Median follow-up time was 32·6 months (IQR 29·4-36·1). Overall response rate was 37% (19 of 52; 95% CI 24-51) overall, 39% (17 of 44; 24-55) for patients with synovial sarcoma, and 25% (two of eight; 3-65) for patients with myxoid round cell liposarcoma. Cytokine release syndrome occurred in 37 (71%) of 52 of patients (one grade 3 event). Cytopenias were the most common grade 3 or worse adverse events (lymphopenia in 50 [96%], neutropenia 44 [85%], leukopenia 42 [81%] of 52 patients). No treatment-related deaths occurred. INTERPRETATION: Afami-cel treatment resulted in durable responses in heavily pre-treated patients with HLA-A*02 and MAGE-A4-expressing synovial sarcoma. This study shows that T-cell receptor therapy can be used to effectively target solid tumours and provides rationale to expand this approach to other solid malignancies. FUNDING: Adaptimmune.


Asunto(s)
Anemia , Liposarcoma Mixoide , Sarcoma Sinovial , Trombocitopenia , Adulto , Humanos , Sarcoma Sinovial/tratamiento farmacológico , Sarcoma Sinovial/genética , Liposarcoma Mixoide/etiología , Síndrome de Liberación de Citoquinas/etiología , Ifosfamida , Trombocitopenia/etiología , Anemia/etiología , Antígenos HLA-A , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico
2.
Nat Med ; 29(1): 104-114, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36624315

RESUMEN

Affinity-optimized T cell receptors can enhance the potency of adoptive T cell therapy. Afamitresgene autoleucel (afami-cel) is a human leukocyte antigen-restricted autologous T cell therapy targeting melanoma-associated antigen A4 (MAGE-A4), a cancer/testis antigen expressed at varying levels in multiple solid tumors. We conducted a multicenter, dose-escalation, phase 1 trial in patients with relapsed/refractory metastatic solid tumors expressing MAGE-A4, including synovial sarcoma (SS), ovarian cancer and head and neck cancer ( NCT03132922 ). The primary endpoint was safety, and the secondary efficacy endpoints included overall response rate (ORR) and duration of response. All patients (N = 38, nine tumor types) experienced Grade ≥3 hematologic toxicities; 55% of patients (90% Grade ≤2) experienced cytokine release syndrome. ORR (all partial response) was 24% (9/38), 7/16 (44%) for SS and 2/22 (9%) for all other cancers. Median duration of response was 25.6 weeks (95% confidence interval (CI): 12.286, not reached) and 28.1 weeks (95% CI: 12.286, not reached) overall and for SS, respectively. Exploratory analyses showed that afami-cel infiltrates tumors, has an interferon-γ-driven mechanism of action and triggers adaptive immune responses. In addition, afami-cel has an acceptable benefit-risk profile, with early and durable responses, especially in patients with metastatic SS. Although the small trial size limits conclusions that can be drawn, the results warrant further testing in larger studies.


Asunto(s)
Antígenos de Neoplasias , Neoplasias de Cabeza y Cuello , Masculino , Humanos , Proteínas de Neoplasias , Antígenos HLA-A , Tratamiento Basado en Trasplante de Células y Tejidos , Inmunoterapia Adoptiva/efectos adversos , Inmunoterapia Adoptiva/métodos
3.
J Immunother Cancer ; 9(2)2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33563772

RESUMEN

BACKGROUND: Tumor endothelial marker 1 (TEM1) is a protein expressed in the tumor-associated endothelium and/or stroma of various types of cancer. We previously demonstrated that immunization with a plasmid-DNA vaccine targeting TEM1 reduced tumor progression in three murine cancer models. Radiation therapy (RT) is an established cancer modality used in more than 50% of patients with solid tumors. RT can induce tumor-associated vasculature injury, triggering immunogenic cell death and inhibition of the irradiated tumor and distant non-irradiated tumor growth (abscopal effect). Combination treatment of RT with TEM1 immunotherapy may complement and augment established immune checkpoint blockade. METHODS: Mice bearing bilateral subcutaneous CT26 colorectal or TC1 lung tumors were treated with a novel heterologous TEM1-based vaccine, in combination with RT, and anti-programmed death-ligand 1 (PD-L1) antibody or combinations of these therapies, tumor growth of irradiated and abscopal tumors was subsequently assessed. Analysis of tumor blood perfusion was evaluated by CD31 staining and Doppler ultrasound imaging. Immunophenotyping of peripheral and tumor-infiltrating immune cells as well as functional analysis was analyzed by flow cytometry, ELISpot assay and adoptive cell transfer (ACT) experiments. RESULTS: We demonstrate that addition of RT to heterologous TEM1 vaccination reduces progression of CT26 and TC1 irradiated and abscopal distant tumors as compared with either single treatment. Mechanistically, RT increased major histocompatibility complex class I molecule (MHCI) expression on endothelial cells and improved immune recognition of the endothelium by anti-TEM1 T cells with subsequent severe vascular damage as measured by reduced microvascular density and tumor blood perfusion. Heterologous TEM1 vaccine and RT combination therapy boosted tumor-associated antigen (TAA) cross-priming (ie, anti-gp70) and augmented programmed cell death protein 1 (PD-1)/PD-L1 signaling within CT26 tumor. Blocking the PD-1/PD-L1 axis in combination with dual therapy further increased the antitumor effect and gp70-specific immune responses. ACT experiments show that anti-gp70 T cells are required for the antitumor effects of the combination therapy. CONCLUSION: Our findings describe novel cooperative mechanisms between heterologous TEM1 vaccination and RT, highlighting the pivotal role that TAA cross-priming plays for an effective antitumor strategy. Furthermore, we provide rationale for using heterologous TEM1 vaccination and RT as an add-on to immune checkpoint blockade as triple combination therapy into early-phase clinical trials.


Asunto(s)
Antígenos CD/metabolismo , Neoplasias Colorrectales/terapia , Inhibidores de Puntos de Control Inmunológico/administración & dosificación , Neoplasias Pulmonares/terapia , Proteínas de Neoplasias/metabolismo , Vacunas de ADN/administración & dosificación , Adenoviridae/genética , Animales , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/farmacología , Línea Celular Tumoral , Neoplasias Colorrectales/diagnóstico por imagen , Terapia Combinada , Inhibidores de Puntos de Control Inmunológico/farmacología , Neoplasias Pulmonares/diagnóstico por imagen , Ratones , Hipofraccionamiento de la Dosis de Radiación , Resultado del Tratamiento , Ultrasonografía Doppler , Vacunas de ADN/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Gynecol Oncol ; 156(1): 222-232, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31818495

RESUMEN

OBJECTIVE: The aim of this study was to "humanize" ovarian cancer patient-derived xenograft (PDX) models by autologous transfer of patient-matched tumor infiltrating lymphocytes (TILs) to evaluate immunotherapies. METHODS: Orthotopic high-grade serous ovarian cancer (HGSOC) PDX models were established from three patient donors. Models were molecularly and histologically validated by immunohistochemistry. TILs were expanded from donor tumors using a rapid expansion protocol. Ex vivo TIL and tumor co-cultures were performed to validate TIL reactivity against patient-matched autologous tumor cells. Expression of TIL activation markers and cytokine secretion was quantitated by flow cytometry and ELISA. As proof of concept, the efficacy of anti-PD-1 monotherapy was tested in autologous TIL/tumor HGSOC PDX models. RESULTS: Evaluation of T-cell activation in autologous TIL/tumor co-cultures resulted in an increase in HLA-dependent IFNγ production and T-cell activation. In response to increased IFNγ production, tumor cell expression of PD-L1 was increased. Addition of anti-PD-1 antibody to TIL/tumor co-cultures increased autologous tumor lysis in a CCNE1 amplified model. Orthotopic HGSOC PDX models from parallel patient-matched tumors maintained their original morphology and molecular marker profile. Autologous tumor-reactive TIL administration in patient-matched PDX models resulted in reduced tumor burden and increased survival, in groups that also received anti-PD-1 therapy. CONCLUSIONS: This study validates a novel, clinically relevant model system for in vivo testing of immunomodulating therapeutic strategies for ovarian cancer, and provides a unique platform for assessing patient-specific T-cell response to immunotherapy.


Asunto(s)
Inmunoterapia/métodos , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/terapia , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Animales , Femenino , Humanos , Activación de Linfocitos , Linfocitos Infiltrantes de Tumor/trasplante , Ratones , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias/métodos , Neoplasias Ováricas/patología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Linfocitos T/inmunología
5.
J Clin Invest ; 130(1): 466-479, 2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31815742

RESUMEN

Alterations in gut microbiota impact the pathophysiology of several diseases, including cancer. Radiotherapy (RT), an established curative and palliative cancer treatment, exerts potent immune modulatory effects, inducing tumor-associated antigen (TAA) cross-priming with antitumor CD8+ T cell elicitation and abscopal effects. We tested whether the gut microbiota modulates antitumor immune response following RT distal to the gut. Vancomycin, an antibiotic that acts mainly on gram-positive bacteria and is restricted to the gut, potentiated the RT-induced antitumor immune response and tumor growth inhibition. This synergy was dependent on TAA cross presentation to cytolytic CD8+ T cells and on IFN-γ. Notably, butyrate, a metabolite produced by the vancomycin-depleted gut bacteria, abrogated the vancomycin effect. In conclusion, depletion of vancomycin-sensitive bacteria enhances the antitumor activity of RT, which has important clinical ramifications.


Asunto(s)
Presentación de Antígeno/efectos de la radiación , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Microbioma Gastrointestinal , Neoplasias Experimentales , Animales , Presentación de Antígeno/genética , Antígenos de Neoplasias/genética , Butiratos/inmunología , Linfocitos T CD8-positivos/patología , Células Dendríticas/patología , Femenino , Microbioma Gastrointestinal/inmunología , Microbioma Gastrointestinal/efectos de la radiación , Ratones , Ratones Noqueados , Neoplasias Experimentales/genética , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Neoplasias Experimentales/radioterapia
6.
JCI Insight ; 3(4)2018 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-29467322

RESUMEN

Adoptive T cell therapy (ACT) is a promising new modality for malignancies. Here, we report that adoptive T cell efficacy in tumor-bearing mice is significantly affected by differences in the native composition of the gut microbiome or treatment with antibiotics, or by heterologous fecal transfer. Depletion of bacteria with vancomycin decreased the rate of tumor growth in mice from The Jackson Laboratory receiving ACT, whereas treatment with neomycin and metronidazole had no effect, indicating the role of specific bacteria in host response. Vancomycin treatment induced an increase in systemic CD8α+ DCs, which sustained systemic adoptively transferred antitumor T cells in an IL-12-dependent manner. In subjects undergoing allogeneic hematopoietic cell transplantation, we found that oral vancomycin also increased IL-12 levels. Collectively, our findings demonstrate an important role played by the gut microbiota in the antitumor effectiveness of ACT and suggest potentially new avenues to improve response to ACT by altering the gut microbiota.


Asunto(s)
Células Dendríticas/inmunología , Microbioma Gastrointestinal/inmunología , Trasplante de Células Madre Hematopoyéticas , Interacciones Microbiota-Huesped/inmunología , Inmunoterapia Adoptiva/métodos , Interleucina-12/inmunología , Neoplasias/terapia , Adulto , Anciano , Animales , Bacterias/efectos de los fármacos , Bacterias/genética , Bacterias/inmunología , Bacterias/aislamiento & purificación , Antígenos CD8/inmunología , Antígenos CD8/metabolismo , Línea Celular Tumoral/trasplante , Estudios de Cohortes , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Trasplante de Microbiota Fecal , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/genética , Interacciones Microbiota-Huesped/efectos de los fármacos , Humanos , Interleucina-12/antagonistas & inhibidores , Interleucina-12/genética , Interleucina-12/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Neomicina/administración & dosificación , Neoplasias/inmunología , Neoplasias/microbiología , Linfocitos T/inmunología , Linfocitos T/trasplante , Resultado del Tratamiento , Vancomicina/administración & dosificación
7.
Clin Cancer Res ; 23(12): 3097-3108, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27993965

RESUMEN

Purpose: PARP inhibition (PARPi) has modest clinical activity in recurrent BRCA-mutant (BRCAMUT) high-grade serous ovarian cancers (HGSOC). We hypothesized that PARPi increases dependence on ATR/CHK1 such that combination PARPi with ATR/CHK1 blockade results in increased cell death and tumor regression.Experimental Design: Effects of PARPi (olaparib), CHK1 inhibition (CHK1i;MK8776), or ATR inhibition (ATRi;AZD6738) alone or in combination on survival, colony formation, cell cycle, genome instability, and apoptosis were evaluated in BRCA1/2MUT HGSOC cells. Tumor growth in vivo was evaluated using a BRCA2MUT patient-derived xenograft (PDX) model.Results: PARPi monotherapy resulted in a decrease in BRCAMUT cell survival, colony formation and suppressed but did not eliminate tumor growth at the maximum tolerated dose (MTD) in a BRCA2MUT PDX. PARPi treatment increased pATR and pCHK1, indicating activation of the ATR-CHK1 fork protection pathway is relied upon for genome stability under PARPi. Indeed, combination of ATRi or CHK1i with PARPi synergistically decreased survival and colony formation compared with single-agent treatments in BRCAMUT cells. Notably, PARPi led to G2 phase accumulation, and the addition of ATRi or CHK1i released cells from G2 causing premature mitotic entry with increased chromosomal aberrations and apoptosis. Moreover, the combinations of PARPi with ATRi or CHK1i were synergistic in causing tumor suppression in a BRCA2MUT PDX with the PARPi-ATRi combination inducing tumor regression and in most cases, complete remission.Conclusions: PARPi causes increased reliance on ATR/CHK1 for genome stability, and combination PARPi with ATR/CHK1i is more effective than PARPi alone in reducing tumor burden in BRCAMUT models. Clin Cancer Res; 23(12); 3097-108. ©2016 AACR.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/antagonistas & inhibidores , Neoplasias Ováricas/tratamiento farmacológico , Animales , Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Proteína BRCA1/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Sinergismo Farmacológico , Femenino , Inestabilidad Genómica/efectos de los fármacos , Humanos , Indoles , Ratones , Terapia Molecular Dirigida , Morfolinas , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Ftalazinas/administración & dosificación , Piperazinas/administración & dosificación , Inhibidores de Poli(ADP-Ribosa) Polimerasas/administración & dosificación , Pirazoles/administración & dosificación , Pirimidinas/administración & dosificación , Sulfonamidas , Sulfóxidos/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Ann Transl Med ; 4(14): 265, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27563652

RESUMEN

Chronic inflammation has been linked to the initiation of carcinogenesis, as well as the advancement of established tumors. The polarization of the tumor inflammatory microenvironment can contribute to either the control, or the progression of the disease. The emerging participation of members of the complement cascade in several hallmarks of cancer, renders it a potential target for anti-tumor treatment. Moreover, the presence of complement regulatory proteins (CRPs) in most types of tumor cells is known to impede anti-tumor therapies. This review focuses on our current knowledge of complement's potential involvement in shaping the inflammatory tumor microenvironment and its role on the regulation of angiogenesis and hypoxia. Furthermore, we discuss approaches using complement-based therapies as an adjuvant in tumor immunotherapy.

9.
J Immunol ; 195(8): 4020-7, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26378078

RESUMEN

Mitochondria provide energy for cells via oxidative phosphorylation. Reactive oxygen species, a byproduct of this mitochondrial respiration, can damage mitochondrial DNA (mtDNA), and somatic mtDNA mutations have been found in all colorectal, ovarian, breast, urinary bladder, kidney, lung, and pancreatic tumors studied. The resulting altered mitochondrial proteins or tumor-associated mitochondrial Ags (TAMAs) are potentially immunogenic, suggesting that they may be targetable Ags for cancer immunotherapy. In this article, we show that the RENCA tumor cell line harbors TAMAs that can drive an antitumor immune response. We generated a cellular tumor vaccine by pulsing dendritic cells with enriched mitochondrial proteins from RENCA cells. Our dendritic cell-based RENCA mitochondrial lysate vaccine elicited a cytotoxic T cell response in vivo and conferred durable protection against challenge with RENCA cells when used in a prophylactic or therapeutic setting. By sequencing mtDNA from RENCA cells, we identified two mutated molecules: COX1 and ND5. Peptide vaccines generated from mitochondrial-encoded COX1 but not from ND5 had therapeutic properties similar to RENCA mitochondrial protein preparation. Thus, TAMAs can elicit effective antitumor immune responses, potentially providing a new immunotherapeutic strategy to treat cancer.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Carcinoma de Células Renales/prevención & control , Ciclooxigenasa 1/inmunología , Neoplasias Renales/prevención & control , Proteínas de la Membrana/inmunología , Proteínas Mitocondriales/inmunología , NADH Deshidrogenasa/inmunología , Neoplasias Experimentales/prevención & control , Animales , Antígenos de Neoplasias/farmacología , Vacunas contra el Cáncer/farmacología , Carcinoma de Células Renales/inmunología , Ciclooxigenasa 1/farmacología , Neoplasias Renales/inmunología , Proteínas de la Membrana/farmacología , Ratones , Ratones Endogámicos BALB C , Proteínas Mitocondriales/farmacología , NADH Deshidrogenasa/farmacología , Neoplasias Experimentales/inmunología
10.
J Immunol ; 194(3): 1285-91, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25548229

RESUMEN

Wound healing is a complex homeostatic response to injury that engages numerous cellular activities, processes, and cell-to-cell interactions. The complement system, an intricate network of proteins with important roles in immune surveillance and homeostasis, has been implicated in many physiological processes; however, its role in wound healing remains largely unexplored. In this study, we employ a murine model of excisional cutaneous wound healing and show that C3(-/-) mice exhibit accelerated early stages of wound healing. Reconstitution of C3(-/-) mice with serum from C3(+/+) mice or purified human C3 abrogated the accelerated wound-healing phenotype. Wound histology of C3(-/-) mice revealed a reduction in inflammatory infiltrate compared with C3(+/+) mice. C3 deficiency also resulted in increased accumulation of mast cells and advanced angiogenesis. We further show that mice deficient in the downstream complement effector C5 exhibit a similar wound-healing phenotype, which is recapitulated in C5aR1(-/-) mice, but not C3aR(-/-) or C5aR2(-/-) mice. Taken together, these data suggest that C5a signaling through C5aR may in part play a pivotal role in recruitment and activation of inflammatory cells to the wound environment, which in turn could delay the early stages of cutaneous wound healing. These findings also suggest a previously underappreciated role for complement in wound healing, and may have therapeutic implications for conditions of delayed wound healing.


Asunto(s)
Proteínas del Sistema Complemento/deficiencia , Piel/inmunología , Piel/lesiones , Cicatrización de Heridas/inmunología , Animales , Complemento C3/deficiencia , Complemento C3/genética , Complemento C3/inmunología , Complemento C5a/genética , Complemento C5a/inmunología , Proteínas del Sistema Complemento/genética , Proteínas del Sistema Complemento/inmunología , Modelos Animales de Enfermedad , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Masculino , Ratones , Ratones Noqueados , Modelos Inmunológicos , Neovascularización Fisiológica/genética , Neovascularización Fisiológica/inmunología , Receptores de Complemento/genética , Receptores de Complemento/metabolismo , Piel/metabolismo , Piel/patología , Cicatrización de Heridas/genética
11.
Proc Natl Acad Sci U S A ; 110(37): 15061-6, 2013 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-23980152

RESUMEN

The skin is colonized by a plethora of microbes that include commensals and potential pathogens, but it is currently unknown how cutaneous host immune mechanisms influence the composition, diversity, and quantity of the skin microbiota. Here we reveal an interactive role for complement in cutaneous host-microbiome interactions. Inhibiting signaling of the complement component C5a receptor (C5aR) altered the composition and diversity of the skin microbiota as revealed by deep sequencing of the bacterial 16S rRNA gene. In parallel, we demonstrate that C5aR inhibition results in down-regulation of genes encoding cutaneous antimicrobial peptides, pattern recognition receptors, and proinflammatory mediators. Immunohistochemistry of inflammatory cell infiltrates in the skin showed reduced numbers of macrophages and lymphocytes with C5aR inhibition. Further, comparing cutaneous gene expression in germ-free mice vs. conventionally raised mice suggests that the commensal microbiota regulates expression of complement genes in the skin. These findings demonstrate a component of host immunity that impacts colonization of the skin by the commensal microbiota and vice versa, a critical step toward understanding host-microbe immune mutualism of the skin and its implications for health and disease. Additionally, we reveal a role for complement in homeostatic host-microbiome interactions of the skin.


Asunto(s)
Proteínas del Sistema Complemento/metabolismo , Microbiota/inmunología , Piel/inmunología , Piel/microbiología , Animales , Péptidos Catiónicos Antimicrobianos/metabolismo , Proteínas del Sistema Complemento/genética , Regulación hacia Abajo , Regulación de la Expresión Génica , Inmunidad Innata/genética , Inflamación/inmunología , Inflamación/microbiología , Mediadores de Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , ARN Bacteriano/genética , ARN Ribosómico 16S/genética , Receptor de Anafilatoxina C5a/antagonistas & inhibidores , Receptor de Anafilatoxina C5a/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo , Transducción de Señal
12.
FASEB J ; 27(7): 2768-76, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23558338

RESUMEN

Although complement is a known contributor to biomaterial-induced complications, pathological implications and therapeutic options remain to be explored. Here we investigated the involvement of complement in the inflammatory response to polypropylene meshes commonly used for hernia repair. In vitro assays revealed deposition of complement activation fragments on the mesh after incubation in plasma. Moreover, significant mesh-induced complement and granulocyte activation was observed in plasma and leukocyte preparations, respectively. Pretreatment of plasma with the complement inhibitor compstatin reduced opsonization >2-fold, and compstatin and a C5a receptor antagonist (C5aRa) impaired granulocyte activation by 50 and 67%, respectively. We established a clinically relevant mouse model of implantation and could confirm deposition of C3 activation fragments on mesh implants in vivo using immunofluorescence. In meshes extracted after subcutaneous or peritoneal implantation, the amount of immune cell infiltrate in mice deficient in key complement components (C3, C5aR), or treated with C5aRa, was approximately half of that observed in wild-type littermates or mice treated with inactive C5aRa, respectively. Our data suggest that implantation of a widely used surgical mesh triggers the formation of an inflammatory cell microenvironment at the implant site through complement activation, and indicates a path for the therapeutic modulation of implant-related complications.


Asunto(s)
Materiales Biocompatibles/farmacología , Activación de Complemento/efectos de los fármacos , Inflamación/prevención & control , Polipropilenos/farmacología , Animales , Antígeno CD11b/metabolismo , Complemento C3/genética , Complemento C3/metabolismo , Citometría de Flujo , Granulocitos/efectos de los fármacos , Granulocitos/metabolismo , Humanos , Implantes Experimentales/efectos adversos , Inflamación/etiología , Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Proteínas Opsoninas/metabolismo , Péptidos Cíclicos/farmacología , Receptor de Anafilatoxina C5a/genética , Receptor de Anafilatoxina C5a/metabolismo , Receptores de Quimiocina/metabolismo
13.
J Immunol ; 189(9): 4674-83, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23028051

RESUMEN

The complement system contributes to various immune and inflammatory diseases, including cancer. In this study, we investigated the capacity of lung cancer cells to activate complement and characterized the consequences of complement activation on tumor progression. We focused our study on the production and role of the anaphylatoxin C5a, a potent immune mediator generated after complement activation. We first measured the capacity of lung cancer cell lines to deposit C5 and release C5a. C5 deposition, after incubation with normal human serum, was higher in lung cancer cell lines than in nonmalignant bronchial epithelial cells. Notably, lung malignant cells produced complement C5a even in the absence of serum. We also found a significant increase of C5a in plasma from patients with non-small cell lung cancer, suggesting that the local production of C5a is followed by its systemic diffusion. The contribution of C5a to lung cancer growth in vivo was evaluated in the Lewis lung cancer model. Syngeneic tumors of 3LL cells grew slower in mice treated with an antagonist of the C5a receptor. C5a did not modify 3LL cell proliferation in vitro but induced endothelial cell chemotaxis and blood-vessels formation. C5a also contributed to the immunosuppressive microenvironment required for tumor growth. In particular, blockade of C5a receptor significantly reduced myeloid-derived suppressor cells and immunomodulators ARG1, CTLA-4, IL-6, IL-10, LAG3, and PDL1 (B7H1). In conclusion, lung cancer cells have the capacity to generate C5a, a molecule that creates a favorable tumor microenvironment for lung cancer progression.


Asunto(s)
Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/patología , Complemento C5a/fisiología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Microambiente Tumoral/inmunología , Adenocarcinoma/sangre , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Animales , Carcinoma Pulmonar de Lewis/prevención & control , Carcinoma de Pulmón de Células no Pequeñas/sangre , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Células Escamosas/sangre , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/patología , Línea Celular Transformada , Línea Celular Tumoral , Activación de Complemento/genética , Activación de Complemento/inmunología , Complemento C5a/biosíntesis , Complemento C5a/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Tolerancia Inmunológica/genética , Neoplasias Pulmonares/prevención & control , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/inmunología , Neovascularización Patológica/patología , Neovascularización Patológica/terapia , Receptor de Anafilatoxina C5a/antagonistas & inhibidores , Receptor de Anafilatoxina C5a/fisiología , Mucosa Respiratoria/citología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Microambiente Tumoral/genética
14.
J Immunol ; 188(2): 641-8, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22184721

RESUMEN

The involvement of IL-4 in liver regeneration has not yet been recognized. In this article, we show that IL-4, produced by NKT cells that accumulate in regenerating livers after partial hepatectomy, contributes to this process by regulating the activation of complement after liver resection in mice. The mechanism of this regulation was associated with the maintenance of an appropriate level of IgM in mouse blood, because IgM deposited in liver parenchyma most likely initiated complement activation during liver regeneration. By controlling complement activation, IL-4 regulated the induction of IL-6, thereby influencing a key pathway involved in regenerating liver cell proliferation and survival. Furthermore, the secretion of IL-4 was controlled by complement through the recruitment of NKT cells to regenerating livers. Our study thus reveals the existence of a regulatory feedback mechanism involving complement and IL-4 that controls liver regeneration.


Asunto(s)
Complemento C3/fisiología , Interleucina-4/fisiología , Regeneración Hepática/inmunología , Animales , Movimiento Celular/genética , Movimiento Celular/inmunología , Proliferación Celular , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Activación de Complemento/genética , Activación de Complemento/inmunología , Complemento C3/deficiencia , Citocinas/biosíntesis , Hepatectomía , Hepatocitos/citología , Hepatocitos/inmunología , Hepatocitos/metabolismo , Inmunoglobulina M/sangre , Interleucina-4/biosíntesis , Interleucina-4/deficiencia , Regeneración Hepática/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/metabolismo
15.
J Immunol ; 186(11): 6568-75, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21531894

RESUMEN

Neonatal respiratory distress syndrome can progress to bronchopulmonary dysplasia (BPD), a serious pulmonary fibrotic disorder. Given the involvement of the extrinsic coagulation cascade in animal models of lung fibrosis, we examined its role in BPD. We observed a higher number of neutrophils expressing tissue factor (TF) in bronchoalveolar lavage fluid (BALF) from infants with BPD than from those with uncomplicated respiratory distress syndrome together with a parallel decrease in TF and connective tissue growth factor (CTGF) in BALF supernatants during the disease course. The involvement of coagulation in the fibrotic process associated with BPD was further evaluated by treating primary human colonic myofibroblasts with BALF supernatants from infants with BPD. These human colonic myofibroblasts demonstrated an enhanced C5a- and thrombin-dependent migration. Moreover, they expressed TF in an endothelin-1-dependent manner, with subsequent activation of the extrinsic coagulation cascade and CTGF production mediated by protease-activator receptor-1 signaling. These data provide a novel mechanism for the development of BPD and indicate that endothelin-1 signaling contributes to fibrosis by upregulating a TF/thrombin amplification loop responsible for CTGF production, and offer novel and specific therapeutic targets for pulmonary fibrotic disease.


Asunto(s)
Displasia Broncopulmonar/metabolismo , Endotelina-1/metabolismo , Síndrome de Dificultad Respiratoria del Recién Nacido/metabolismo , Transducción de Señal , Western Blotting , Líquido del Lavado Bronquioalveolar/química , Displasia Broncopulmonar/genética , Displasia Broncopulmonar/patología , Células Cultivadas , Colon/metabolismo , Colon/patología , Complemento C5a/genética , Complemento C5a/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Endotelina-1/genética , Femenino , Fibrosis , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunohistoquímica , Recién Nacido , Pulmón/metabolismo , Pulmón/patología , Masculino , Microscopía Fluorescente , Miofibroblastos/metabolismo , Miofibroblastos/patología , Receptor de Anafilatoxina C5a , Receptor PAR-1/genética , Receptor PAR-1/metabolismo , Receptores de Complemento/genética , Receptores de Complemento/metabolismo , Síndrome de Dificultad Respiratoria del Recién Nacido/genética , Síndrome de Dificultad Respiratoria del Recién Nacido/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trombina/genética , Trombina/metabolismo , Tromboplastina/genética , Tromboplastina/metabolismo
16.
Blood ; 116(22): 4395-403, 2010 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-20625009

RESUMEN

Beyond its role in immunity, complement mediates a wide range of functions in the context of morphogenetic or tissue remodeling processes. Angiogenesis is crucial during tissue remodeling in multiple pathologies; however, the knowledge about the regulation of neovascularization by the complement components is scarce. Here we studied the involvement of complement in pathological angiogenesis. Strikingly, we found that mice deficient in the central complement component C3 displayed increased neovascularization in the model of retinopathy of prematurity (ROP) and in the in vivo Matrigel plug assay. In addition, antibody-mediated blockade of C5, treatment with C5aR antagonist, or C5aR deficiency in mice resulted in enhanced pathological retina angiogenesis. While complement did not directly affect angiogenesis-related endothelial cell functions, we found that macrophages mediated the antiangiogenic activity of complement. In particular, C5a-stimulated macrophages were polarized toward an angiogenesis-inhibitory phenotype, including the up-regulated secretion of the antiangiogenic soluble vascular endothelial growth factor receptor-1. Consistently, macrophage depletion in vivo reversed the increased neovascularization associated with C3- or C5aR deficiency. Taken together, complement and in particular the C5a-C5aR axes are potent inhibitors of angiogenesis.


Asunto(s)
Complemento C3/inmunología , Complemento C5/inmunología , Inmunidad Innata , Neovascularización Patológica/inmunología , Retina/patología , Retinopatía de la Prematuridad/inmunología , Animales , Técnicas de Cultivo de Célula , Línea Celular , Complemento C3/genética , Complemento C5a/inmunología , Eliminación de Gen , Humanos , Recién Nacido , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/patología , Receptor de Anafilatoxina C5a/genética , Receptor de Anafilatoxina C5a/inmunología , Retina/inmunología , Retinopatía de la Prematuridad/patología , Factores de Crecimiento Endotelial Vascular/inmunología
17.
Blood ; 116(4): 631-9, 2010 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-20424189

RESUMEN

Thrombosis is a common complication of end-stage renal disease, particularly in patients on hemodialysis. Although substantial progress has been made in preventing thrombotic complications in various other groups of patients, the mechanisms of thrombosis during hemodialysis require clarification. In this report, we demonstrate that complement activation triggered by hemodialysis biomaterials, and the subsequent generation of the complement anaphylatoxin C5a, results in the expression of functionally active tissue factor (TF) in peripheral blood neutrophils. Because TF is a key initiator of coagulation in vivo, we postulate that the recurring complement activation that occurs during long-term hemodialysis contributes to thrombosis in dialyzed end-stage renal disease patients. Furthermore, we found that complement contributed to the induction of granulocyte colony-stimulating factor, which has been implicated in the pathogenesis of thrombosis in patients treated with the recombinant form of this molecule. Importantly, the inhibition of complement activation attenuated the TF expression and granulocyte colony-stimulating factor induction in blood passing through a hemodialysis circuit, suggesting that the complement system could become a new therapeutic target for preventing thrombosis in patients with chronic renal failure who are maintained on hemodialysis.


Asunto(s)
Complemento C5a/fisiología , Diálisis Renal/efectos adversos , Trombosis/etiología , Anciano , Anafilatoxinas/farmacología , Anafilatoxinas/fisiología , Coagulación Sanguínea/efectos de los fármacos , Células Cultivadas , Activación de Complemento/efectos de los fármacos , Complemento C5a/metabolismo , Complemento C5a/farmacología , Femenino , Factor Estimulante de Colonias de Granulocitos/metabolismo , Humanos , Fallo Renal Crónico/sangre , Fallo Renal Crónico/complicaciones , Fallo Renal Crónico/metabolismo , Fallo Renal Crónico/terapia , Leucocitos/efectos de los fármacos , Leucocitos/metabolismo , Leucocitos/patología , Masculino , Persona de Mediana Edad , Receptor de Anafilatoxina C5a/metabolismo , Tromboplastina/metabolismo , Factores de Tiempo
18.
Eur J Immunol ; 40(5): 1461-72, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20162553

RESUMEN

The induction of the autophagy machinery, a process for the catabolism of cytosolic proteins and organelles, constitutes a crucial mechanism in innate immunity. However, the involvement of autophagy in human neutrophils and the possible inducers of this process have not been completely elucidated. In this study, the induction of autophagy was examined in human neutrophils treated with various activators and detected by the formation of acidified autophagosomes through monodansylcadaverine staining and via LC-3B conversion screened by immunoblotting and immunofluorescence confocal microscopy. In addition, the expression of the ATG genes was assessed by real-time RT-PCR. We provide evidence that autophagy is implicated in human neutrophils in both a phagocytosis-independent (rapamycin, TLR agonists, PMA) and phagocytosis (Escherichia coli)-dependent initiation manner. ROS activation is a positive mechanism for autophagy induction in the case of PMA, TLR activation and phagocytosis. Furthermore, LC3B gene expression was uniformly upregulated, indicating a transcriptional level of regulation for the autophagic machinery. This study provides a stepping stone toward further investigation of autophagy in neutrophil-driven inflammatory disorders.


Asunto(s)
Autofagia/fisiología , Neutrófilos/fisiología , Adenina/análogos & derivados , Adenina/farmacología , Autofagia/efectos de los fármacos , Autofagia/genética , Cadaverina/análogos & derivados , Cadaverina/análisis , Cromonas/farmacología , Colorantes/análisis , Escherichia coli , Guanosina/análogos & derivados , Guanosina/farmacología , Humanos , Concentración de Iones de Hidrógeno , Inflamación/inmunología , Microscopía Confocal , Morfolinas/farmacología , Neutrófilos/citología , Neutrófilos/efectos de los fármacos , Fagosomas/fisiología , Fagosomas/ultraestructura , Poli I-C/farmacología , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/biosíntesis , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética , Acetato de Tetradecanoilforbol/farmacología , Receptores Toll-Like/efectos de los fármacos , Receptores Toll-Like/fisiología , Transcripción Genética , Vacuolas/fisiología
19.
Circ Res ; 103(5): 536-44, 2008 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-18658052

RESUMEN

The adipocytokine leptin modulates vascular remodeling and neointima formation. Because endothelial progenitor cells (EPCs) participate in vascular repair, we analyzed the effects of leptin on human EPC function in vitro and in vivo. After 7 days in culture, EPCs expressed the leptin receptor and responded to leptin stimulation with increased STAT3 phosphorylation. Incubation of EPCs with leptin (at concentrations between 1 and 100 ng/mL) increased the number of EPCs adhering to vitronectin and fibronectin in a receptor-specific manner. It also enhanced the capacity of EPCs to incorporate into a monolayer of human endothelial cells and the adherence of these cells to activated platelets. Leptin upregulated alphavbeta5 and alpha4 integrin expression in EPCs, and the effects of leptin on EPC function could be prevented, at least in part, by RGD peptides and function-blocking antibodies. Intravenous injection of fluorescently labeled human EPCs into athymic nude mice shortly after vascular injury revealed that preincubation of EPCs with leptin augmented their accumulation within intimal lesions, accelerating reendothelialization and decreasing neointima formation in an alphavbeta5 and alpha4 integrin-dependent manner. Our findings suggest that leptin specifically modulates the adhesive properties and the homing potential of EPCs and may thus enhance their capacity to promote vascular regeneration in vivo.


Asunto(s)
Adhesión Celular/fisiología , Células Endoteliales/citología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Leptina/metabolismo , Movimiento Celular/fisiología , Células Cultivadas , Células Endoteliales/metabolismo , Humanos , Integrina alfa4/metabolismo , Oligopéptidos/metabolismo , Fosforilación , ARN Mensajero/metabolismo , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Receptores de Vitronectina/metabolismo , Factor de Transcripción STAT3/metabolismo , Túnica Íntima/citología , Túnica Íntima/metabolismo
20.
J Immunol ; 180(11): 7368-75, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18490736

RESUMEN

Acute respiratory distress syndrome (ARDS) is characterized by the presence of fibrin-rich inflammatory exudates in the intra-alveolar spaces and the extensive migration of neutrophils into alveoli of the lungs. Tissue factor (TF)-dependent procoagulant properties of bronchoalveaolar lavage fluid (BALF) obtained from ARDS patients favor fibrin deposition, and are likely the result of cross-talk between inflammatory mediators and hemostatic mechanisms. However, the regulation of these interactions remains elusive. Prompted by previous findings suggesting that neutrophils, under certain inflammatory conditions, can express functional TF, we investigated the contribution of intra-alveolar neutrophils to the procoagulant properties of BALF from patients with ARDS. Our results confirm that the procoagulant properties of BALF from ARDS patients are the result of TF induction, and further indicate that BALF neutrophils are a main source of TF in intra-alveolar fluid. We also found that BALF neutrophils in these patients express significantly higher levels of TF than peripheral blood neutrophils. These results suggest that the alveolar microenvironment contributes to TF induction in ARDS. Additional experiments indicated that the ability of BALF to induce TF expression in neutrophils from healthy donors can be abolished by inhibiting C5a or TNF-alpha signaling, suggesting a primary role for these inflammatory mediators in the up-regulation of TF in alveolar neutrophils in ARDS. This cross-talk between inflammatory mediators and the induction of TF expression in intra-alveolar neutrophils may be a potential target for novel therapeutic strategies to limit ARDS-associated disturbances of coagulation.


Asunto(s)
Complemento C5a/metabolismo , Citocinas/metabolismo , Neutrófilos/metabolismo , Síndrome de Dificultad Respiratoria/metabolismo , Tromboplastina/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Adulto , Anciano , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Complemento C5a/inmunología , Citocinas/inmunología , Fibrina/inmunología , Fibrina/metabolismo , Humanos , Persona de Mediana Edad , Neutrófilos/inmunología , Síndrome de Dificultad Respiratoria/inmunología , Tromboplastina/inmunología , Factor de Necrosis Tumoral alfa/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA