Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Crohns Colitis ; 2024 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-38243565

RESUMEN

BACKGROUND AND AIMS: The G protein coupled receptor GPR15 is expressed on and functionally important for T cells homing to the large intestine. However, the precise mechanisms by which GPR15 controls gut homing have been unclear. Thus, we aimed to elucidate these mechanisms as well as to explore the potential of targeting GPR15 for interfering with T cell recruitment to the colon in IBD. METHODS: We used dynamic adhesion and transmigration assays as well as a humanized in vivo model of intestinal cell trafficking to study GPR15-dependent effects on gut homing. Moreover, we analysed GPR15 and integrin expression in patients with and without IBD cross-sectionally and longitudinally. RESULTS: GPR15 controlled T cell adhesion to MAdCAM-1 and VCAM-1 upstream of α4ß7 and α4ß1 integrin, respectively. Consistently, high co-expression of these integrins with GPR15 was found on T cells from patients with IBD and GPR15 also promoted T cell recruitment to the colon in humanized mice. Anti-GPR15 antibodies effectively blocked T cell gut homing in vitro and in vivo. In vitro data as well as observations in a cohort of patients treated with vedolizumab suggest that this might be more effective than inhibiting α4ß7. CONCLUSIONS: GPR15 seems to have a broad, but organ-selective impact on T cell trafficking and is therefore a promising target for future therapy of IBD. Further studies are needed.

3.
BMJ Open ; 11(12): e049208, 2021 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-34880013

RESUMEN

INTRODUCTION: Accumulating evidence suggests that the adoptive transfer of ex vivo expanded regulatory T cells (Treg) may overcome colitogenic immune responses in patients with inflammatory bowel diseases. The objective of the ER-TREG 01 trial is to assess safety and tolerability of a single infusion of autologous ex vivo expanded Treg in adults with ulcerative colitis. METHODS AND ANALYSIS: The study is designed as a single-arm, fast-track dose-escalation trial. The study will include 10 patients with ulcerative colitis. The study intervention consists of (1) a baseline visit; (2) a second visit that includes a leukapheresis to generate the investigational medicinal product, (3) a third visit to infuse the investigational medicinal product and (4) five subsequent follow-up visits within the next 26 weeks to assess safety and tolerability. Patients will intravenously receive a single dose of 0.5×106, 1×106, 2×106, 5×106 or 10×106 autologous Treg/kg body weight. The primary objective is to define the maximum tolerable dose of a single infusion of autologous ex vivo expanded Treg. Secondary objectives include the evaluation of safety of one single infusion of autologous ex vivo expanded Treg, efficacy assessment and accompanying immunomonitoring to measure Treg function in the peripheral blood and intestinal mucosa. ETHICS AND DISSEMINATION: The study protocol was approved by the Ethics Committee of the Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany (number 417_19 Az). In addition, the study was approved by the Paul-Ehrlich Institute, Federal Institute for Vaccines and Biomedicines, Langen, Germany (number 3652/01). The study is funded by the German Research Foundation (DFG, KFO 257 project 08 and SFB/TransRegio 241 project C04). The trial will be conducted in compliance with this study protocol, the Declaration of Helsinki, Good Clinical Practice and Good Manufacturing Practice. The results will be published in peer-reviewed scientific journals and disseminated in scientific conferences and media. TRIAL REGISTRATION NUMBER: NCT04691232.


Asunto(s)
Colitis Ulcerosa , Trasplante de Células Madre Hematopoyéticas , Ensayos Clínicos Fase I como Asunto , Colitis Ulcerosa/terapia , Alemania , Humanos , Inmunidad , Linfocitos T Reguladores
4.
Int J Mol Sci ; 22(19)2021 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-34638566

RESUMEN

Natural killer (NK) cells, members of the innate immune system, play an important role in the rejection of HLA class I negative tumor cells. Hence, a therapeutic vaccine, which can activate NK cells in addition to cells of the adaptive immune system might induce a more comprehensive cellular response, which could lead to increased tumor elimination. Dendritic cells (DCs) are capable of activating and expanding NK cells, especially when the NFκB pathway is activated in the DCs thereby leading to the secretion of the cytokine IL-12. Another prominent NK cell activator is IL-15, which can be bound by the IL-15 receptor alpha-chain (IL-15Rα) to be transpresented to the NK cells. However, monocyte-derived DCs do neither secrete IL-15, nor express the IL-15Rα. Hence, we designed a chimeric protein consisting of IL-15 and the IL-15Rα. Upon mRNA electroporation, the fusion protein was detectable on the surface of the DCs, and increased the potential of NFκB-activated, IL-12-producing DC to activate NK cells in an autologous cell culture system with ex vivo-generated cells from healthy donors. These data show that a chimeric IL-15/IL-15Rα molecule can be expressed by monocyte-derived DCs, is trafficked to the cell surface, and is functional regarding the activation of NK cells. These data represent an initial proof-of-concept for an additional possibility of further improving cellular DC-based immunotherapies of cancer.


Asunto(s)
Células Dendríticas/inmunología , Interleucina-15/biosíntesis , Células Asesinas Naturales/inmunología , Receptores de Interleucina-15/biosíntesis , Proteínas Recombinantes de Fusión/biosíntesis , Células Dendríticas/efectos de los fármacos , Electroporación , Humanos , Quinasa I-kappa B/biosíntesis , Quinasa I-kappa B/genética , Inmunoterapia , Interleucina-15/química , Interleucina-15/genética , Células Asesinas Naturales/efectos de los fármacos , Leucocitos Mononucleares , FN-kappa B/farmacología , Cultivo Primario de Células , Receptores de Interleucina-15/química , Receptores de Interleucina-15/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética
5.
Front Immunol ; 12: 656452, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34017333

RESUMEN

Inflammatory bowel diseases (IBDs), including Crohn's disease (CD) and ulcerative colitis (UC) are multifactorial diseases with still unknown aetiology and an increasing prevalence and incidence worldwide. Despite plentiful therapeutic options for IBDs, the lack or loss of response in certain patients demands the development of further treatments to tackle this unmet medical need. In recent years, the success of the anti-α4ß7 antibody vedolizumab highlighted the potential of targeting the homing of immune cells, which is now an important pillar of IBD therapy. Due to its complexity, leukocyte trafficking and the involved molecules offer a largely untapped resource for a plethora of potential therapeutic interventions. In this review, we aim to summarise current and future directions of specifically interfering with immune cell trafficking. We will comment on concepts of homing, retention and recirculation and particularly focus on the role of tissue-derived chemokines. Moreover, we will give an overview of the mode of action of drugs currently in use or still in the pipeline, highlighting their mechanisms and potential to reduce disease burden.


Asunto(s)
Movimiento Celular/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Animales , Moléculas de Adhesión Celular/antagonistas & inhibidores , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Quimiocinas/antagonistas & inhibidores , Quimiocinas/metabolismo , Susceptibilidad a Enfermedades/inmunología , Desarrollo de Medicamentos , Humanos , Enfermedades Inflamatorias del Intestino/metabolismo , Integrinas/antagonistas & inhibidores , Integrinas/metabolismo , Leucocitos/efectos de los fármacos , Leucocitos/inmunología , Leucocitos/metabolismo , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Terapia Molecular Dirigida , Receptores de Esfingosina-1-Fosfato/metabolismo , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo
6.
Nat Commun ; 11(1): 5224, 2020 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-33067467

RESUMEN

Natural killer (NK) cells are important effector cells in the immune response to cancer. Clinical trials on adoptively transferred NK cells in patients with solid tumors, however, have thus far been unsuccessful. As NK cells need to pass stringent safety evaluation tests before clinical use, the cells are cryopreserved to bridge the necessary evaluation time. Standard degranulation and chromium release cytotoxicity assays confirm the ability of cryopreserved NK cells to kill target cells. Here, we report that tumor cells embedded in a 3-dimensional collagen gel, however, are killed by cryopreserved NK cells at a 5.6-fold lower rate compared to fresh NK cells. This difference is mainly caused by a 6-fold decrease in the fraction of motile NK cells after cryopreservation. These findings may explain the persistent failure of NK cell therapy in patients with solid tumors and highlight the crucial role of a 3-D environment for testing NK cell function.


Asunto(s)
Movimiento Celular , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Técnicas de Cultivo de Célula , Supervivencia Celular , Células Cultivadas , Criopreservación , Citotoxicidad Inmunológica , Humanos , Células Asesinas Naturales/química
7.
Ther Adv Med Oncol ; 11: 1758835919891622, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31853267

RESUMEN

BACKGROUND: In therapeutic cancer vaccination, monocyte-derived dendritic cells (moDCs) efficiently activate specific T-cell responses; however, optimizing the activation of innate immune cells could support and improve the antitumor effects. A major disadvantage of moDCs matured with the standard cytokine cocktail (consisting of IL-1ß, IL-6, TNFα, and PGE2) is their inability to secrete IL-12p70. IL-12 prominently activates natural killer (NK) cells, which are crucial in innate antitumor immunity, as they act as helper cells for the induction of a cytotoxic T lymphocyte (CTL) response and are also able to directly kill the tumor. METHODS: Previously we have shown that triggering the NF-κB pathway in moDCs by transfection of mRNA encoding constitutively active IKKß (caIKKß) led to IL-12p70 secretion and improved the dendritic cells' capability to activate and expand CTLs with a memory-like phenotype. In this study, we examined whether such dendritic cells could activate autologous NK cells. RESULTS: moDCs matured with the standard cytokine cocktail followed by transfection with the caIKKß-RNA were able to activate autologous NK cells, detected by the upregulation of CD54, CD69, and CD25 on the NK cells, their ability to secrete IFNγ, and their high lytic activity. Moreover, the ability of NK-cell activation was not diminished by simultaneous T-cell activation. CONCLUSION: The capacity of caIKKß-DCs to activate both the adaptive and innate immune response indicates an enhanced potential for clinical efficacy.

8.
Front Immunol ; 8: 1371, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29123521

RESUMEN

In recent years, the exploration of regulatory T cell (Treg)-based cellular therapy has become an attractive strategy to ameliorate inflammation and autoimmunity in various clinical settings. The main obstacle to the clinical application of Treg in human is their low number circulating in peripheral blood. Therefore, ex vivo expansion is inevitable. Moreover, isolation of Treg bears the risk of concurrent isolation of unwanted effector cells, which may trigger or deteriorate inflammation upon adoptive Treg transfer. Here, we present a protocol for the GMP-compliant production, lot-release and validation of ex vivo expanded Tregs for treatment of patients with autoimmune and inflammatory disorders. In the presented production protocol, large numbers of Treg, previously enriched from a leukapheresis product by using the CliniMACS® system, are ex vivo expanded in the presence of anti-CD3/anti-CD28 expander beads, exogenous IL-2 and rapamycin during 21 days. The expanded Treg drug product passed predefined lot-release criteria. These criteria include (i) sterility testing, (ii) assessment of Treg phenotype, (iii) assessment of non-Treg cellular impurities, (iv) confirmation of successful anti-CD3/anti-CD28 expander bead removal after expansion, and (v) confirmation of the biological function of the Treg product. Furthermore, the Treg drug product was shown to retain its stability and suppressive function for at least 1 year after freezing and thawing. Also, dilution of the Treg drug product in 0.9% physiological saline did not affect Treg phenotype and Treg function for up to 90 min. These data indicate that these cells are ready to use in a clinical setting in which a cell infusion time of up to 90 min can be expected. The presented production process has recently undergone on site GMP-conform evaluation and received GMP certification from the Bavarian authorities in Germany. This protocol can now be used for Treg-based therapy of various inflammatory and autoimmune disorders.

9.
Inflamm Bowel Dis ; 23(8): 1348-1359, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28708802

RESUMEN

BACKGROUND: A local imbalance between regulatory (Treg) and effector T cells is believed to play a major role in gut-specific inflammation, including ulcerative colitis (UC). Restoration of this balance through an adoptive Treg transfer is an attractive new treatment approach in patients who are refractory to current standard therapies. It was our goal to develop a Good Manufacturing Practices (GMP)-conform protocol for expansion of UC Treg cells as a rational backbone for future studies on Treg therapy in UC. METHODS: CD25 blood T cells derived from patients with UC were ex vivo expanded in the presence of IL-2, rapamycin, and anti-CD3/anti-CD28 expander beads using a GMP-conform protocol. Cells were subsequently assessed for stability and function. RESULTS: Patient-derived ex vivo rapamycin-expanded GMP-ready CD25 cells were polyclonal, hypomethylated at intron 1 of the FoxP3 locus, and suppressive in carboxyfluorescein succinimidyl ester-dilution assays against autologous peripheral blood-derived and allogeneic colon-derived responder cells. Function was mediated by soluble factors, including toxic granules. In addition to CD4 T cells, suppressive hypermethylated CD8 T-cell subsets were also induced during the expansion process. CONCLUSIONS: Patient-derived rapamycin-expanded CD25 cells are stable and functional, and as such, ready to serve in a phase I dose-escalation safety study in UC.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Colitis Ulcerosa/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Adulto , Anciano , Células Cultivadas , Colitis Ulcerosa/terapia , Femenino , Humanos , Masculino , Persona de Mediana Edad
10.
Inflamm Bowel Dis ; 23(3): 379-391, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28221249

RESUMEN

BACKGROUND: The precise mechanisms controlling homing of T effector (Teff) cells to the inflamed gut in Crohn's disease (CD) are still unclear, and clinical outcome data from patients with inflammatory bowel disease treated with the anti-α4ß7 integrin antibody vedolizumab suggest differences between ulcerative colitis and CD. METHODS: Expression of homing molecules was studied with flow cytometry and immunohistochemistry. Their functional role was investigated in in vitro adhesion assays and in a humanized mouse model of T cell homing to the inflamed gut in vivo. RESULTS: Despite in vitro blockade of CD Teff adhesion to mucosal vascular addressin cell adhesion molecule-1 (MadCAM-1) and in contrast to previous observations in ulcerative colitis, anti-α4ß7 treatment did not result in reduced Teff cell homing to the colon in vivo. However, the integrin α4ß1 was expressed in higher levels on Teffs from patients with CD compared with controls, while its expression in the peripheral blood declined, and its expression in the intestine increased during the course of clinical vedolizumab treatment. Consistently, adhesion of CD Teffs to vascular cell adhesion molecule-1 (VCAM-1) was blocked by inhibition of α4 and α4ß1 in vitro. Moreover, in vivo homing of CD Teffs to the ileum was reduced by inhibition of α4 and α4ß1 integrins, but not α4ß7 integrins. CONCLUSIONS: Our findings suggest that Teff cell homing to the ileum through the axis α4ß1-VCAM-1 is an essential and nonredundant pathway in CD in vivo, possibly affecting efficacy of clinical treatment with antiadhesion compounds.


Asunto(s)
Enfermedad de Crohn/inmunología , Íleon/inmunología , Integrina alfa4beta1/inmunología , Receptores Mensajeros de Linfocitos/inmunología , Linfocitos T/inmunología , Adulto , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Moléculas de Adhesión Celular , Movimiento Celular , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/patología , Enfermedad de Crohn/tratamiento farmacológico , Enfermedad de Crohn/patología , Femenino , Citometría de Flujo , Fármacos Gastrointestinales/farmacología , Humanos , Íleon/patología , Inmunoglobulinas/efectos de los fármacos , Inmunoglobulinas/inmunología , Inmunohistoquímica , Integrina alfa4beta1/efectos de los fármacos , Masculino , Ratones , Mucoproteínas/efectos de los fármacos , Mucoproteínas/inmunología , Receptores Mensajeros de Linfocitos/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Molécula 1 de Adhesión Celular Vascular/efectos de los fármacos , Molécula 1 de Adhesión Celular Vascular/inmunología
11.
Atherosclerosis ; 230(1): 92-9, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23958259

RESUMEN

PURPOSE: Mature dendritic cells (DCs) play a crucial role in the inflammatory process within atherosclerotic lesions by stimulation of effector T cells, which can contribute to plaque instability. Interactions between DCs and regulatory T cells (Treg), which regulate immune response by attenuating acute inflammation, are postulated to be involved in the pathogenesis of autoimmune diseases. We investigated a possible correlation between infiltrated DCs and Treg in human atherosclerotic plaques. METHODS: Cross-sections of 40 human carotid endarterectomy specimens were classified into groups of stable and vulnerable plaques using Trichrome staining. Immunohistochemical staining of plaques was used to detect infiltrated total (S100) and mature DCs (fascin, DC-LAMP, CD83), Treg (CD3, Foxp3), and to analyze the inflammatory state of the plaques (CD3, COX-2, CD68). In addition, RNA was isolated from plaque specimens and quantitative real-time PCR was performed to analyze transcription rates of DC markers (CD11c, CD209, HLA-DR), maturation markers (CD80, CD83, CD86), Treg-associated genes (CTLA-4, Foxp3) and of pro- and anti-inflammatory cytokines (TGFß-family, IL-10, IFN-γ, IL-17α, IL-6). Migration assays and adhesion experiments were performed, to investigate the effects of Treg on mature DCs in vitro. RESULTS: As compared with stable plaques, vulnerable lesions were characterized by increased numbers of COX-2-expressing cells and T lymphocytes, indicating an enhanced inflammatory process. In vulnerable plaques, numbers of total and mature DCs were significantly higher in the inflammatory plaque shoulder, whereas the numbers of Treg were decreased compared to stable plaques. This inverse correlation and the association of the observed infiltration rates with plaque stability, were confirmed by PCR analyses, showing increased transcription levels of DC-specific markers, decreased mRNA expression of Treg-associated genes and decreased anti-inflammatory cytokines in vulnerable atherosclerotic plaques. In vitro, pre-incubation of mature DCs with Treg resulted in decreased DC migration and inhibited the adhesion of DCs to endothelial cells under non-uniform shear stress. CONCLUSIONS: The results of our study provide novel insights in the direct interaction of mature DCs and Treg in plaque inflammation and stability.


Asunto(s)
Aterosclerosis/metabolismo , Estenosis Carotídea/metabolismo , Células Dendríticas/citología , Linfocitos T Reguladores/citología , Anciano , Arterias Carótidas/patología , Estenosis Carotídea/cirugía , Adhesión Celular , Quimiotaxis , Citocinas/metabolismo , Células Dendríticas/metabolismo , Endarterectomía Carotidea , Femenino , Regulación de la Expresión Génica , Humanos , Inmunohistoquímica , Inflamación , Masculino , Persona de Mediana Edad , Linfocitos T Reguladores/metabolismo , Transcripción Genética
12.
PLoS One ; 8(1): e53745, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23341990

RESUMEN

BACKGROUND: Ipilimumab, a cytotoxic T-lymphocyte antigen-4 (CTLA-4) blocking antibody, has been approved for the treatment of metastatic melanoma and induces adverse events (AE) in up to 64% of patients. Treatment algorithms for the management of common ipilimumab-induced AEs have lead to a reduction of morbidity, e.g. due to bowel perforations. However, the spectrum of less common AEs is expanding as ipilimumab is increasingly applied. Stringent recognition and management of AEs will reduce drug-induced morbidity and costs, and thus, positively impact the cost-benefit ratio of the drug. To facilitate timely identification and adequate management data on rare AEs were analyzed at 19 skin cancer centers. METHODS AND FINDINGS: Patient files (n = 752) were screened for rare ipilimumab-associated AEs. A total of 120 AEs, some of which were life-threatening or even fatal, were reported and summarized by organ system describing the most instructive cases in detail. Previously unreported AEs like drug rash with eosinophilia and systemic symptoms (DRESS), granulomatous inflammation of the central nervous system, and aseptic meningitis, were documented. Obstacles included patients delay in reporting symptoms and the differentiation of steroid-induced from ipilimumab-induced AEs under steroid treatment. Importantly, response rate was high in this patient population with tumor regression in 30.9% and a tumor control rate of 61.8% in stage IV melanoma patients despite the fact that some patients received only two of four recommended ipilimumab infusions. This suggests that ipilimumab-induced antitumor responses can have an early onset and that severe autoimmune reactions may reflect overtreatment. CONCLUSION: The wide spectrum of ipilimumab-induced AEs demands doctor and patient awareness to reduce morbidity and treatment costs and true ipilimumab success is dictated by both objective tumor responses and controlling severe side effects.


Asunto(s)
Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/inmunología , Antígeno CTLA-4/inmunología , Melanoma/tratamiento farmacológico , Melanoma/patología , Neoplasias Cutáneas/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/efectos adversos , Antineoplásicos/inmunología , Antineoplásicos/uso terapéutico , Sistema Endocrino/efectos de los fármacos , Femenino , Tracto Gastrointestinal/efectos de los fármacos , Humanos , Ipilimumab , Riñón/efectos de los fármacos , Hígado/efectos de los fármacos , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Sistema Nervioso/efectos de los fármacos , Páncreas/efectos de los fármacos , Sistema Respiratorio/efectos de los fármacos , Estudios Retrospectivos , Piel/efectos de los fármacos
13.
Head Neck ; 34(12): 1734-46, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22287423

RESUMEN

BACKGROUND: We performed a pilot study using Trojan vaccines in patients with advanced squamous cell carcinoma of the head and neck (SCCHN). These vaccines are composed of HLA-I and HLA-II restricted melanoma antigen E (MAGE)-A3 or human papillomavirus (HPV)-16 derived peptides, joined by furin-cleavable linkers, and linked to a "penetrin" peptide sequence derived from HIV-TAT. Thirty-one patients with SCCHN were screened for the trial and 5 were enrolled. METHODS: Enrolled patients were treated with 300 µg of Trojan peptide supplemented with Montanide and granulocyte-macrophage colony-stimulating factor (GM-CSF) at 4-week intervals for up to 4 injections. RESULTS: Following vaccination, peripheral blood mononuclear cells (PBMCs) from 4 of 5 patients recognized both the full Trojan constructs and constituent HLA-II peptides, whereas responses to HLA-I restricted peptides were less pronounced. CONCLUSION: This treatment regimen seems to have acceptable toxicity and elicits measurable systemic immune responses against HLA-II restricted epitopes in a subset of patients with advanced SCCHN.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/uso terapéutico , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/terapia , Epítopos/inmunología , Neoplasias de Cabeza y Cuello/inmunología , Neoplasias de Cabeza y Cuello/terapia , Papillomavirus Humano 16/inmunología , Proteínas de Neoplasias/inmunología , Adulto , Anciano , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Carcinoma de Células Escamosas/virología , Epítopos de Linfocito T , Femenino , Antígeno HLA-A2/inmunología , Neoplasias de Cabeza y Cuello/virología , Humanos , Inmunoterapia Adoptiva/métodos , Interferón gamma/inmunología , Masculino , Persona de Mediana Edad , Infecciones por Papillomavirus/complicaciones , Infecciones por Papillomavirus/inmunología , Proyectos Piloto , Reacción en Cadena de la Polimerasa , Carcinoma de Células Escamosas de Cabeza y Cuello , Linfocitos T Citotóxicos/inmunología , Activación Viral
14.
J Exp Clin Cancer Res ; 29: 134, 2010 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-20937115

RESUMEN

BACKGROUND: The possibility that autologous NK cells could serve as an effective treatment modality for solid tumors has long been considered. However, implementation is hampered by (i) the small number of NK cells in peripheral blood, (ii) the difficulties associated with large-scale production of GMP compliant cytolytic NK cells, (iii) the need to activate the NK cells in order to induce NK cell mediated killing and (iv) the constraints imposed by autologous inhibitory receptor-ligand interactions. To address these issues, we determined (i) if large numbers of NK cells could be expanded from PBMC and GMP compliant cell fractions derived by elutriation, (ii) their ability to kill allogeneic and autologous tumor targets by direct cytotoxicity and by antibody-mediated cellular cytotoxicity and (iii) defined NK cell specific receptor-ligand interactions that mediate tumor target cell killing. METHODS: Human NK cells were expanded during 14 days. Expansion efficiency, NK receptor repertoire before and after expansion, expression of NK specific ligands, cytolytic activity against allogeneic and autologous tumor targets, with and without the addition of chimeric EGFR monoclonal antibody, were investigated. RESULTS: Cell expansion shifted the NK cell receptor repertoire towards activation and resulted in cytotoxicity against various allogeneic tumor cell lines and autologous gastric cancer cells, while sparing normal PBMC. Blocking studies confirmed that autologous cytotoxicity is established through multiple activating receptor-ligand interactions. Importantly, expanded NK cells also mediated ADCC in an autologous and allogeneic setting by antibodies that are currently being used to treat patients with select solid tumors. CONCLUSION: These data demonstrate that large numbers of cytolytic NK cells can be generated from PBMC and lymphocyte-enriched fractions obtained by GMP compliant counter current elutriation from PBMC, establishing the preclinical evidence necessary to support clinical trials utilizing autologous expanded NK cells, both directly and in combination with monoclonal antibodies in future cell-based immunotherapy in select solid tumors.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos , Proliferación Celular , Células Asesinas Naturales/inmunología , Activación de Linfocitos , Receptores Inmunológicos/inmunología , Neoplasias Gástricas/inmunología , Anticuerpos Monoclonales , Línea Celular Tumoral , Separación Celular , Técnicas de Cocultivo , Receptores ErbB/inmunología , Humanos , Inmunofenotipificación , Ligandos , Fenotipo , Factores de Tiempo
15.
J Immunol ; 184(2): 787-95, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20008291

RESUMEN

CD137 (4-1BB)-mediated costimulation plays an important role in directing the fate of Ag-stimulated T cells and NK cells, yet the role of CD137 in mediating B cell function is unknown. We found that CD137 is expressed in vitro on anti-Ig-stimulated peripheral blood B cells and in vivo on tonsillar B cells with an activated phenotype. In vitro CD137 expression is enhanced by CD40 stimulation and IFN-gamma and is inhibited by IL-4, -10, and -21. The expression of CD137 on activated human B cells is functionally relevant because engagement with its ligand at the time of activation stimulates B cell proliferation, enhances B cell survival, and induces secretion of TNF-alpha and -beta. Our study suggests that CD137 costimulation may play a role in defining the fate of Ag-stimulated human B cells.


Asunto(s)
Linfocitos B/citología , Proliferación Celular , Supervivencia Celular , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/fisiología , Antígenos/inmunología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Células Sanguíneas , Antígenos CD40 , Humanos , Interleucinas , Activación de Linfocitos , Linfotoxina-alfa/metabolismo , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/agonistas , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/antagonistas & inhibidores , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
16.
Curr Mol Med ; 9(6): 683-93, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19689295

RESUMEN

In the vast majority of studies conducted to date, activation of cancer-specific T cell immunity through peptide-based immunization has failed to induce objective tumor regression. This failure is particularly troublesome given that these vaccines often stimulate T cell responses. In this review, we attempt to understand the relative failure of peptide cancer vaccines to achieve clinically meaningful responses. In the first part of the review, we discuss specific hurdles to successful application of synthetic peptide-based vaccines including patient variability and epitope selection. In the second part of this review, we summarize the importance of CD4+ T cell help in peptide-based vaccine strategies and offer a potential strategy to improve peptide-based vaccines through the generation of both HLA class I and class II vaccine specific-immune responses.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Vacunas de Subunidad/inmunología , Linfocitos T CD4-Positivos/inmunología , Ensayos Clínicos como Asunto , Epítopos/inmunología , Humanos , Inmunoterapia , Vacunas Sintéticas/inmunología
17.
J Mol Recognit ; 22(3): 242-9, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19177494

RESUMEN

Antibody based manipulation of the CD137 (4-1BB) co-signaling pathway is an attractive option for the treatment of cancer and autoimmune disease. We developed a chimeric anti-human CD137 monoclonal antibody (GG) and characterized its function. As a component of planned preclinical studies, we evaluated the binding of GG to activated peripheral blood mononuclear cells (PBMCs) from cynomolgus macaque and baboon against human. Interestingly, GG only recognized human CD137, while a commercial anti-CD137 mAb (4B4-1), recognized activated PBMCs from both human and non-human primates (NHP). Subsequent analysis revealed that the amino acid sequence of CD137 is largely conserved between primate species ( approximately 95% identical), with the extracellular domain differing by only 9-10 amino acids. Based on these data, we generated mutant constructs in the extracellular domain, replacing NHP with human CD137 sequences, and identified 3 amino acids critical for GG binding. These residues are likely part of a conformational epitope, as a peptide spanning this region is unable to block mAb binding. These data demonstrate that subtle sequence variations of defined co-stimulatory molecules amongst primate species can be employed as a strategy for mapping residues necessary for antibody binding to conformational epitopes.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Mapeo Epitopo , Primates/inmunología , Proteínas Recombinantes/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Secuencia de Aminoácidos , Animales , Células CHO , Cricetinae , Cricetulus , Epítopos/química , Glicosilación , Humanos , Leucocitos Mononucleares/inmunología , Modelos Animales , Modelos Moleculares , Datos de Secuencia Molecular , Péptidos/química , Unión Proteica , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/química
18.
Cancer Immunol Immunother ; 58(7): 997-1006, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18979096

RESUMEN

PURPOSE: The interaction of Fc fragments of antibodies with the Fcgamma receptors is an essential checkpoint in antibody-dependent cellular cytotoxicity (ADCC). Specific polymorphisms at position 158 enhance FcgammaRIIIa affinity for IgG1 and are associated with improved clinical outcome in lymphoma patients treated with IgG1 anti-CD20 antibody. The role of ADCC in the therapeutic effects of the alpha-epidermal growth factor receptor (EGFR) mAb, cetuximab, in patients with squamous cell carcinoma of the head and neck (SCCHN) is poorly defined. We employed three SCCHN cell lines to test two hypotheses: (1) SCCHN is susceptible to cetuximab-mediated ADCC, (2) efficacy of ADCC is associated with polymorphisms at position 158 of FcgammaRIIIa. EXPERIMENTAL DESIGN: FcgammaRIIIa-158 polymorphisms were determined for healthy donors, and their purified NK cells were used as effector cells against three SCCHN cell lines in ADCC assays. Cytotoxicity levels were compared for each polymorphism class. Proliferation and cell cycle assays were done to examine the direct effects of cetuximab. RESULTS: Our results indicate that SCCHN is susceptible to cetuximab-mediated ADCC in vitro. NK cytotoxic efficiency correlates with donor 158-polymorphisms in FcgammaRIIIa. Overall cytotoxicity was greatest for individuals having a single V allele when compared to homozygous F/F individuals; the cumulative percent cytotoxicity for each polymorphism among the cell lines was 58.2% V/V, 50.6% V/F, and 26.1% F/F (P < 0.001). Additionally, the presence of a V allele correlated with superior natural cytotoxicity against NK sensitive targets. CONCLUSION: These data have both prognostic and therapeutic relevance and support the design of a prospective trial to determine the influence of FcgammaRIIIa polymorphisms on the clinical outcome of patients with SCCHN treated with alpha-EGFR mAbs.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos/genética , Antineoplásicos/inmunología , Carcinoma de Células Escamosas/inmunología , Neoplasias de Cabeza y Cuello/inmunología , Células Asesinas Naturales/metabolismo , Receptores de IgG/genética , Alelos , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Antineoplásicos/uso terapéutico , Carcinoma de Células Escamosas/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cetuximab , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Humanos , Células Asesinas Naturales/inmunología , Polimorfismo Genético , Receptores de IgG/inmunología
19.
Blood ; 112(3): 699-707, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18519814

RESUMEN

CD137 (4-1BB) is a costimulatory molecule that can be manipulated for the treatment of cancer and autoimmune disease. Although it is known that agonistic antibodies (mAbs) against CD137 enhance the rejection of murine tumors in a natural killer (NK) cell- and T cell-dependent fashion, the mechanism for NK dependence is poorly understood. In this study, we evaluated the ability of 2 different glycoforms of a chimerized antihuman CD137 mAb, an aglycosylated (GA) and a low fucose form (GG), to react with human NK cells. Both mAbs bound similarly to CD137 and partially blocked the interaction between CD137 and CD137 ligand. However, unlike GA mAb, immobilized GG mAb activated NK cells and enhanced CD137 expression. These effects were seemingly dependent on Fc interaction with putative Fc receptors on the NK-cell surface, as only the immobilized Fc-fragment of GG was required for CD137 expression. Furthermore, CD137 expression could be enhanced with antibodies directed against non-CD137 epitopes, and the expression levels directly correlated with patterns of Fc-glycosylation recognized to improve Fc interaction with Fc gamma receptors. Our data suggest that CD137 can be enhanced on NK cells in an Fc-dependent fashion and that expression correlates with phenotypic and functional parameters of activation.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Fragmentos Fc de Inmunoglobulinas/fisiología , Células Asesinas Naturales/metabolismo , Receptores Fc/metabolismo , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Células Cultivadas , Glicosilación , Humanos , Proteínas Recombinantes de Fusión
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...