Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
JCI Insight ; 7(19)2022 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-35998045

RESUMEN

New strategies that augment T cell responses are required to broaden the therapeutic arsenal against cancer. CD96, TIGIT, and CD226 are receptors that bind to a communal ligand, CD155, and transduce either inhibitory or activating signals. The function of TIGIT and CD226 is established, whereas the role of CD96 remains ambiguous. Using a panel of engineered antibodies, we discovered that the T cell stimulatory activity of anti-CD96 antibodies requires antibody cross-linking and is potentiated by Fcγ receptors. Thus, soluble "Fc silent" anti-CD96 antibodies failed to stimulate human T cells, whereas the same antibodies were stimulatory after coating onto plastic surfaces. Remarkably, the activity of soluble anti-CD96 antibodies was reinstated by engineering the Fc domain to a human IgG1 isotype, and it was dependent on antibody trans-cross-linking by FcγRI. In contrast, neither human IgG2 nor variants with increased Fcγ receptor IIB binding possessed stimulatory activity. Anti-CD96 antibodies acted directly on T cells and augmented gene expression networks associated with T cell activation, leading to proliferation, cytokine secretion, and resistance to Treg suppression. Furthermore, CD96 expression correlated with survival in HPV+ head and neck squamous cell carcinoma, and its cross-linking activated tumor-infiltrating T cells, thus highlighting the potential of anti-CD96 antibodies in cancer immunotherapy.


Asunto(s)
Células Asesinas Naturales , Neoplasias , Antígenos CD/metabolismo , Citocinas/metabolismo , Humanos , Inmunoglobulina G/metabolismo , Ligandos , Plásticos/metabolismo , Receptores de IgG/metabolismo , Receptores Inmunológicos/metabolismo
2.
J Extracell Vesicles ; 11(5): e12226, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35595718

RESUMEN

Colorectal cancer (CRC) with a mesenchymal gene expression signature has the greatest propensity for distant metastasis and is characterised by the accumulation of cancer-associated fibroblasts in the stroma. We investigated whether the epithelial to mesenchymal transition status of CRC cells influences fibroblast phenotype, with a focus on the transfer of extracellular vesicles (EVs), as a controlled means of cell-cell communication. Epithelial CRC EVs suppressed TGF-ß-driven myofibroblast differentiation, whereas mesenchymal CRC EVs did not. This was driven by miR-200 (miR-200a/b/c, -141), which was enriched in epithelial CRC EVs and transferred to recipient fibroblasts. Ectopic miR-200 expression or ZEB1 knockdown, in fibroblasts, similarly suppressed myofibroblast differentiation. Supporting these findings, there was a strong negative correlation between miR-200 and myofibroblastic markers in a cohort of CRC patients in the TCGA dataset. This was replicated in mice, by co-injecting epithelial or mesenchymal CRC cells with fibroblasts and analysing stromal markers of myofibroblastic phenotype. Fibroblasts from epithelial tumours contained more miR-200 and expressed less ACTA2 and FN1 than those from mesenchymal tumours. As such, these data provide a new mechanism for the development of fibroblast heterogeneity in CRC, through EV-mediated transfer of miRNAs, and provide an explanation as to why CRC tumours with greater metastatic potential are CAF rich.


Asunto(s)
Neoplasias Colorrectales , Transición Epitelial-Mesenquimal , Vesículas Extracelulares , MicroARNs , Animales , Neoplasias Colorrectales/genética , Transición Epitelial-Mesenquimal/genética , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo , Fibroblastos/metabolismo , Humanos , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Fenotipo
3.
J Exp Clin Cancer Res ; 41(1): 131, 2022 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-35392965

RESUMEN

BACKGROUND: Hypoxia is a hallmark of the tumor microenvironment (TME) and in addition to altering metabolism in cancer cells, it transforms tumor-associated stromal cells. Within the tumor stromal cell compartment, tumor-associated macrophages (TAMs) provide potent pro-tumoral support. However, TAMs can also be harnessed to destroy tumor cells by monoclonal antibody (mAb) immunotherapy, through antibody dependent cellular phagocytosis (ADCP). This is mediated via antibody-binding activating Fc gamma receptors (FcγR) and impaired by the single inhibitory FcγR, FcγRIIb. METHODS: We applied a multi-OMIC approach coupled with in vitro functional assays and murine tumor models to assess the effects of hypoxia inducible factor (HIF) activation on mAb mediated depletion of human and murine cancer cells. For mechanistic assessments, siRNA-mediated gene silencing, Western blotting and chromatin immune precipitation were utilized to assess the impact of identified regulators on FCGR2B gene transcription. RESULTS: We report that TAMs are FcγRIIbbright relative to healthy tissue counterparts and under hypoxic conditions, mononuclear phagocytes markedly upregulate FcγRIIb. This enhanced FcγRIIb expression is transcriptionally driven through HIFs and Activator protein 1 (AP-1). Importantly, this phenotype reduces the ability of macrophages to eliminate anti-CD20 monoclonal antibody (mAb) opsonized human chronic lymphocytic leukemia cells in vitro and EL4 lymphoma cells in vivo in human FcγRIIb+/+ transgenic mice. Furthermore, post-HIF activation, mAb mediated blockade of FcγRIIb can partially restore phagocytic function in human monocytes. CONCLUSION: Our findings provide a detailed molecular and cellular basis for hypoxia driven resistance to antitumor mAb immunotherapy, unveiling a hitherto unexplored aspect of the TME. These findings provide a mechanistic rationale for the modulation of FcγRIIb expression or its blockade as a promising strategy to enhance approved and novel mAb immunotherapies.


Asunto(s)
Leucemia Linfocítica Crónica de Células B , Receptores de IgG , Animales , Anticuerpos Monoclonales/farmacología , Humanos , Hipoxia/metabolismo , Inmunoterapia , Leucemia Linfocítica Crónica de Células B/metabolismo , Macrófagos/metabolismo , Ratones , Receptores de IgG/genética , Receptores de IgG/metabolismo , Microambiente Tumoral
4.
JCI Insight ; 5(18)2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32870822

RESUMEN

Despite advances in identifying the key immunoregulatory roles of many of the human leukocyte immunoglobulin-like receptor (LILR) family members, the function of the inhibitory molecule LILRB3 (ILT5, CD85a, LIR3) remains unclear. Studies indicate a predominant myeloid expression; however, high homology within the LILR family and a relative paucity of reagents have hindered progress toward identifying the function of this receptor. To investigate its function and potential immunomodulatory capacity, a panel of LILRB3-specific monoclonal antibodies (mAbs) was generated. LILRB3-specific mAbs bound to discrete epitopes in Ig-like domain 2 or 4. LILRB3 ligation on primary human monocytes by an agonistic mAb resulted in phenotypic and functional changes, leading to potent inhibition of immune responses in vitro, including significant reduction in T cell proliferation. Importantly, agonizing LILRB3 in humanized mice induced tolerance and permitted efficient engraftment of allogeneic cells. Our findings reveal powerful immunosuppressive functions of LILRB3 and identify it as an important myeloid checkpoint receptor.


Asunto(s)
Antígenos CD/genética , Epítopos/inmunología , Proteínas de Punto de Control Inmunitario/genética , Tolerancia Inmunológica , Linfoma/genética , Monocitos/inmunología , Receptores Inmunológicos/genética , Animales , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/metabolismo , Antígenos CD/inmunología , Línea Celular Tumoral , Proliferación Celular , Mapeo Epitopo , Epítopos/química , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Xenoinjertos , Humanos , Proteínas de Punto de Control Inmunitario/inmunología , Linfoma/inmunología , Linfoma/mortalidad , Linfoma/patología , Ratones , Monocitos/citología , Biblioteca de Péptidos , Cultivo Primario de Células , Receptores Inmunológicos/agonistas , Receptores Inmunológicos/inmunología , Análisis de Supervivencia , Linfocitos T/citología , Linfocitos T/inmunología , Trasplante Homólogo
5.
Cell Death Dis ; 10(10): 703, 2019 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-31543517

RESUMEN

Epithelial-mesenchymal transition (EMT) is a process by which tumour cells lose epithelial characteristics, become mesenchymal and highly motile. EMT pathways also induce stem cell features and resistance to apoptosis. Identifying and targeting this pool of tumour cells is a major challenge. Protein kinase C (PKC) inhibition has been shown to eliminate breast cancer stem cells but has never been assessed in hepatocellular cancer (HCC). We investigated ZEB family of EMT inducer expression as a biomarker for metastatic HCC and evaluated the efficacy of PKC inhibitors for HCC treatment. We showed that ZEB1 positivity predicted patient survival in multiple cohorts and also validated as an independent biomarker of HCC metastasis. ZEB1-expressing HCC cell lines became resistant to conventional chemotherapeutic agents and were enriched in CD44high/CD24low cell population. ZEB1- or TGFß-induced EMT increased PKCα abundance. Probing public databases ascertained a positive association of ZEB1 and PKCα expression in human HCC tumours. Inhibition of PKCα activity by small molecule inhibitors or by PKCA knockdown reduced viability of mesenchymal HCC cells in vitro and in vivo. Our results suggest that ZEB1 expression predicts survival and metastatic potential of HCC. Chemoresistant/mesenchymal HCC cells become addicted to PKC pathway and display sensitivity to PKC inhibitors such as UCN-01. Stratifying patients according to ZEB1 and combining UCN-01 with conventional chemotherapy may be an advantageous chemotherapeutic strategy.


Asunto(s)
Neoplasias Hepáticas/tratamiento farmacológico , Proteína Quinasa C/antagonistas & inhibidores , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética , Animales , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Femenino , Humanos , Neoplasias Hepáticas/patología , Ratones , Ratones SCID , Metástasis de la Neoplasia , Transfección
6.
Cancer Immunol Res ; 7(11): 1876-1890, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31451483

RESUMEN

Therapy-resistant microenvironments represent a major barrier toward effective elimination of disseminated cancer. Many hematologic and solid tumors are resistant to therapeutic antibodies in the bone marrow (BM), but not in the periphery (e.g., spleen). We previously showed that cyclophosphamide (CTX) sensitizes the BM niche to antibody therapeutics. Here, we show that (i) BM resistance was induced not only by the tumor but also by the intrinsic BM microenvironment; (ii) CTX treatment overcame both intrinsic and extrinsic resistance mechanisms by augmenting macrophage activation and phagocytosis, including significant upregulation of activating Fcγ receptors (FcγRIII and FcγRIV) and downregulation of the inhibitory receptor, FcγRIIB; and (iii) CTX synergized with cetuximab (anti-EGFR) and trastuzumab (anti-Her2) in eliminating metastatic breast cancer in the BM of humanized mice. These findings provide insights into the mechanisms by which CTX synergizes with antibody therapeutics in resistant niche-specific organs and its applicability in treating BM-resident tumors.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Médula Ósea/inmunología , Ciclofosfamida/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Receptores de IgG/genética , Animales , Antineoplásicos Inmunológicos/inmunología , Antineoplásicos Inmunológicos/uso terapéutico , Médula Ósea/efectos de los fármacos , Médula Ósea/patología , Ciclofosfamida/inmunología , Ciclofosfamida/uso terapéutico , Resistencia a Antineoplásicos/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inmunoterapia , Activación de Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/terapia , Receptores de IgG/metabolismo , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
7.
Clin Cancer Res ; 24(10): 2383-2394, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29514845

RESUMEN

Purpose: PD-1 checkpoint blockade has revolutionized the field of cancer immunotherapy, yet the frequency of responding patients is limited by inadequate T-cell priming secondary to a paucity of activatory dendritic cells (DC). DC signals can be bypassed by CD27 agonists, and we therefore investigated if the effectiveness of anti-PD-1/L1 could be improved by combining with agonist anti-CD27 monoclonal antibodies (mAb).Experimental Design: The efficacy of PD-1/L1 blockade or agonist anti-CD27 mAb was compared with a dual-therapy approach in multiple tumor models. Global transcriptional profiling and flow cytometry analysis were used to delineate mechanisms underpinning the observed synergy.Results: PD-1/PD-L1 blockade and agonist anti-CD27 mAb synergize for increased CD8+ T-cell expansion and effector function, exemplified by enhanced IFNγ, TNFα, granzyme B, and T-bet. Transcriptome analysis of CD8+ T cells revealed that combination therapy triggered a convergent program largely driven by IL2 and Myc. However, division of labor was also apparent such that anti-PD-1/L1 activates a cytotoxicity-gene expression program whereas anti-CD27 preferentially augments proliferation. In tumor models, either dependent on endogenous CD8+ T cells or adoptive transfer of transgenic T cells, anti-CD27 mAb synergized with PD-1/L1 blockade for antitumor immunity. Finally, we show that a clinically relevant anti-human CD27 mAb, varlilumab, similarly synergizes with PD-L1 blockade for protection against lymphoma in human-CD27 transgenic mice.Conclusions: Our findings suggest that suboptimal T-cell invigoration in cancer patients undergoing treatment with PD-1 checkpoint blockers will be improved by dual PD-1 blockade and CD27 agonism and provide mechanistic insight into how these approaches cooperate for CD8+ T-cell activation. Clin Cancer Res; 24(10); 2383-94. ©2018 AACR.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Neoplasias/genética , Neoplasias/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Transcriptoma , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/agonistas , Traslado Adoptivo , Animales , Antineoplásicos Inmunológicos/farmacología , Antineoplásicos Inmunológicos/uso terapéutico , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Inmunomodulación/efectos de los fármacos , Inmunomodulación/genética , Inmunomodulación/inmunología , Activación de Linfocitos/inmunología , Melanoma Experimental , Ratones , Ratones Transgénicos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo
8.
Cancer Cell ; 32(6): 777-791.e6, 2017 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-29198913

RESUMEN

Monoclonal antibodies (mAbs) can destroy tumors by recruiting effectors such as myeloid cells, or targeting immunomodulatory receptors to promote cytotoxic T cell responses. Here, we examined the therapeutic potential of combining a direct tumor-targeting mAb, anti-CD20, with an extended panel of immunomodulatory mAbs. Only the anti-CD27/CD20 combination provided cures. This was apparent in multiple lymphoma models, including huCD27 transgenic mice using the anti-huCD27, varlilumab. Detailed mechanistic analysis using single-cell RNA sequencing demonstrated that anti-CD27 stimulated CD8+ T and natural killer cells to release myeloid chemo-attractants and interferon gamma, to elicit myeloid infiltration and macrophage activation. This study demonstrates the therapeutic advantage of using an immunomodulatory mAb to regulate lymphoid cells, which then recruit and activate myeloid cells for enhanced killing of mAb-opsonized tumors.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Linfoma/inmunología , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/agonistas , Animales , Anticuerpos Monoclonales Humanizados , Humanos , Inmunoterapia/métodos , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Activación de Macrófagos/efectos de los fármacos , Activación de Macrófagos/inmunología , Ratones , Ratones Transgénicos
9.
Aging (Albany NY) ; 9(12): 2666-2694, 2017 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-29283887

RESUMEN

Colorectal cancer is a global disease with increasing incidence. Mortality is largely attributed to metastatic spread and therefore, a mechanistic dissection of the signals which influence tumor progression is needed. Cancer stroma plays a critical role in tumor proliferation, invasion and chemoresistance. Here, we sought to identify and characterize exosomal microRNAs as mediators of stromal-tumor signaling. In vitro, we demonstrated that fibroblast exosomes are transferred to colorectal cancer cells, with a resultant increase in cellular microRNA levels, impacting proliferation and chemoresistance. To probe this further, exosomal microRNAs were profiled from paired patient-derived normal and cancer-associated fibroblasts, from an ongoing prospective biomarker study. An exosomal cancer-associated fibroblast signature consisting of microRNAs 329, 181a, 199b, 382, 215 and 21 was identified. Of these, miR-21 had highest abundance and was enriched in exosomes. Orthotopic xenografts established with miR-21-overexpressing fibroblasts and CRC cells led to increased liver metastases compared to those established with control fibroblasts. Our data provide a novel stromal exosome signature in colorectal cancer, which has potential for biomarker validation. Furthermore, we confirmed the importance of stromal miR-21 in colorectal cancer progression using an orthotopic model, and propose that exosomes are a vehicle for miR-21 transfer between stromal fibroblasts and cancer cells.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Neoplasias Colorrectales/patología , Exosomas/metabolismo , MicroARNs/genética , Anciano , Animales , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Progresión de la Enfermedad , Exosomas/genética , Femenino , Xenoinjertos , Humanos , Masculino , Ratones , MicroARNs/metabolismo
10.
Proc Natl Acad Sci U S A ; 114(7): E1178-E1187, 2017 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-28137869

RESUMEN

Memory CD8+ T cells confer long-term immunity against tumors, and anticancer vaccines therefore should maximize their generation. Multiple memory CD8+ T-cell subsets with distinct functional and homing characteristics exist, but the signaling pathways that regulate their development are ill defined. Here we examined the role of the serine/threonine kinase Akt in the generation of protective immunity by CD8+ T cells. Akt is known to be activated by the T-cell antigen receptor and the cytokine IL-2, but its role in T-cell immunity in vivo has not been explored. Using CD8+ T cells from pdk1K465E/K465E knockin mice, we found that decreased Akt activity inhibited the survival of T cells during the effector-to-memory cell transition and abolished their differentiation into C-X-C chemokine receptor 3 (CXCR3)loCD43lo effector-like memory cells. Consequently, antitumor immunity by CD8+ T cells that display defective Akt signaling was substantially diminished during the memory phase. Reduced memory T-cell survival and altered memory cell differentiation were associated with up-regulation of the proapoptotic protein Bim and the T-box transcription factor eomesodermin, respectively. These findings suggest an important role for effector-like memory CD8+ T cells in tumor immune surveillance and identify Akt as a key signaling node in the development of protective memory CD8+ T-cell responses.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica/inmunología , Vigilancia Inmunológica/inmunología , Neoplasias Experimentales/inmunología , Proteínas Proto-Oncogénicas c-akt/inmunología , Animales , Sitios de Unión/genética , Linfocitos T CD8-positivos/metabolismo , Receptor 1 de Quimiocinas CX3C/inmunología , Receptor 1 de Quimiocinas CX3C/metabolismo , Línea Celular Tumoral , Vigilancia Inmunológica/genética , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Neoplasias Experimentales/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores CXCR3/inmunología , Receptores CXCR3/metabolismo , Transducción de Señal/inmunología
11.
J Clin Oncol ; 34(34): 4132-4141, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27863190

RESUMEN

Purpose In squamous cell carcinomas of the head and neck (HNSCC), the increasing incidence of oropharyngeal squamous cell carcinomas (OPSCCs) is attributable to human papillomavirus (HPV) infection. Despite commonly presenting at late stage, HPV-driven OPSCCs are associated with improved prognosis compared with HPV-negative disease. HPV DNA is also detectable in nonoropharyngeal (non-OPSCC), but its pathogenic role and clinical significance are unclear. The objectives of this study were to determine whether HPV plays a causal role in non-OPSCC and to investigate whether HPV confers a survival benefit in these tumors. Methods Meta-analysis was used to build a cross-tissue gene-expression signature for HPV-driven cancer. Classifiers trained by machine-learning approaches were used to predict the HPV status of 520 HNSCCs profiled by The Cancer Genome Atlas project. DNA methylation data were similarly used to classify 464 HNSCCs and these analyses were integrated with genomic, histopathology, and survival data to permit a comprehensive comparison of HPV transcript-positive OPSCC and non-OPSCC. Results HPV-driven tumors accounted for 4.1% of non-OPSCCs. Regardless of anatomic site, HPV+ HNSCCs shared highly similar gene expression and DNA methylation profiles; nonkeratinizing, basaloid histopathological features; and lack of TP53 or CDKN2A alterations. Improved overall survival, however, was largely restricted to HPV-driven OPSCCs, which were associated with increased levels of tumor-infiltrating lymphocytes compared with HPV-driven non-OPSCCs. Conclusion Our analysis identified a causal role for HPV in transcript-positive non-OPSCCs throughout the head and neck. Notably, however, HPV-driven non-OPSCCs display a distinct immune microenvironment and clinical behavior compared with HPV-driven OPSCCs.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma de Células Escamosas/virología , Neoplasias de Cabeza y Cuello/virología , Papillomaviridae/genética , Infecciones por Papillomavirus/virología , Metilación de ADN , Pruebas de ADN del Papillomavirus Humano , Humanos , Neoplasias Orofaríngeas/virología , Pronóstico , Análisis de Supervivencia
12.
Cancer Res ; 76(14): 4136-48, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27206847

RESUMEN

Antibodies that block T-cell-regulatory checkpoints have recently emerged as a transformative approach to cancer treatment. However, the clinical efficacy of checkpoint blockade depends upon inherent tumor immunogenicity, with variation in infiltrating T cells contributing to differences in objective response rates. Here, we sought to understand the molecular correlates of tumor-infiltrating T lymphocytes (TIL) in squamous cell carcinoma (SCC), using a systems biologic approach to integrate publicly available omics datasets with histopathologic features. We provide evidence that links TIL abundance and therapeutic outcome to the regulation of tumor glycolysis by EGFR and HIF, both of which are attractive molecular targets for use in combination with immunotherapeutics. Cancer Res; 76(14); 4136-48. ©2016 AACR.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Carcinoma de Células Escamosas/inmunología , Glucosa/metabolismo , Neoplasias de Cabeza y Cuello/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Animales , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/mortalidad , Receptores ErbB/genética , Receptores ErbB/fisiología , Femenino , Transportador de Glucosa de Tipo 1/análisis , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/mortalidad , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Ratones , Ratones Endogámicos C57BL , Carcinoma de Células Escamosas de Cabeza y Cuello , Transcriptoma , Regulación hacia Arriba
13.
Clin Cancer Res ; 22(19): 4827-4836, 2016 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-27091407

RESUMEN

PURPOSE: We have clinically evaluated a DNA fusion vaccine to target the HLA-A*0201-binding peptide CAP-1 from carcinoembryonic antigen (CEA605-613) linked to an immunostimulatory domain (DOM) from fragment C of tetanus toxin. EXPERIMENTAL DESIGN: Twenty-seven patients with CEA-expressing carcinomas were recruited: 15 patients with measurable disease (arm-I) and 12 patients without radiological evidence of disease (arm-II). Six intramuscular vaccinations of naked DNA (1 mg/dose) were administered up to week 12. Clinical and immunologic follow-up was up to week 64 or clinical/radiological disease. RESULTS: DOM-specific immune responses demonstrated successful vaccine delivery. All patients without measurable disease compared with 60% with advanced disease responded immunologically, while 58% and 20% expanded anti-CAP-1 CD8+ T cells, respectively. CAP-1-specific T cells were only detectable in the blood postvaccination but could also be identified in previously resected cancer tissue. The gastrointestinal adverse event diarrhea was reported by 48% of patients and linked to more frequent decreases in CEA (P < 0.001) and improved global immunologic responses [anti-DOM responses of greater magnitude (P < 0.001), frequency (P = 0.004), and duration] compared with patients without diarrhea. In advanced disease patients, decreases in CEA were associated with better overall survival (HR = 0.14, P = 0.017). CAP-1 peptide was detectable on MHC class I of normal bowel mucosa and primary colorectal cancer tissue by mass spectrometry, offering a mechanistic explanation for diarrhea through CD8+ T-cell attack. CONCLUSIONS: Our data suggest that DNA vaccination is able to overcome peripheral tolerance in normal and tumor tissue and warrants testing in combination studies, for example, by vaccinating in parallel to treatment with an anti-PD1 antibody. Clin Cancer Res; 22(19); 4827-36. ©2016 AACR.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Antígeno Carcinoembrionario/inmunología , Carcinoma/tratamiento farmacológico , Citotoxicidad Inmunológica/efectos de los fármacos , Vacunas de ADN/uso terapéutico , Linfocitos T CD8-positivos/efectos de los fármacos , Femenino , Humanos , Activación de Linfocitos/efectos de los fármacos , Masculino , Oligopéptidos/inmunología , Oligopéptidos/uso terapéutico
14.
Blood ; 127(24): 3015-25, 2016 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-27002119

RESUMEN

Kinase inhibitors targeting the B-cell receptor (BCR) are now prominent in the treatment of chronic lymphocytic leukemia (CLL). We have focused here on interleukin 4 (IL-4), a cytokine that protects normal and malignant B cells from apoptosis and increases surface immunoglobulin M (sIgM) expression on murine splenic B cells. First, we have demonstrated that IL-4 treatment increased sIgM expression in vitro on peripheral blood B cells obtained from healthy individuals. In CLL, IL-4 target genes are overexpressed in cells purified from the lymph nodes of patients compared with cells derived from matched blood and bone marrow samples. As for normal B cells, IL-4 increased sIgM expression on CLL cells in vitro, especially in samples expressing unmutated V-genes. IL-4-induced sIgM expression was associated with increased receptor signalling activity, measured by anti-IgM-induced calcium mobilization, and with increased expression of CD79B messenger RNA and protein, and the "mature" glycoform of sIgM. Importantly, the ability of the BCR-associated kinase inhibitors idelalisib and ibrutinib, approved for treatment of CLL and other B-cell malignancies, to inhibit anti-IgM-induced signalling was reduced following IL-4 pretreatment in samples from the majority of patients. In contrast to stimulatory effects on sIgM, IL-4 decreased CXCR4 and CXCR5 expression; therefore, CLL cells, particularly within the progressive unmutated V-gene subset, may harness the ability of IL-4 to promote BCR signalling and B-cell retention within lymph nodes. Effects of IL-4 were mediated via JAK3/STAT6 and we propose a potential role for JAK inhibitors in combination with BCR kinase inhibitors for the treatment of CLL.


Asunto(s)
Membrana Celular/metabolismo , Inmunoglobulina M/genética , Inmunoglobulina M/metabolismo , Interleucina-4/farmacología , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/metabolismo , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Células Cultivadas , Interacciones Farmacológicas , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Humanos , Janus Quinasa 3/metabolismo , Leucemia Linfocítica Crónica de Células B/inmunología , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Factor de Transcripción STAT6/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
15.
Blood ; 127(4): 449-57, 2016 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-26491071

RESUMEN

Antigenic stimulation via the B-cell receptor (BCR) is a major driver of the proliferation and survival of chronic lymphocytic leukemia (CLL) cells. However, the precise mechanisms by which BCR stimulation leads to accumulation of malignant cells remain incompletely understood. Here, we investigated the ability of BCR stimulation to increase messenger RNA (mRNA) translation, which can promote carcinogenesis by effects on both global mRNA translation and upregulated expression of specific oncoproteins. Re-analysis of gene expression profiles revealed striking upregulation of pathways linked to mRNA translation both in CLL cells derived from lymph nodes, the major site of antigen stimulation in vivo, and after BCR stimulation in vitro. Anti-IgM significantly increased mRNA translation in primary CLL cells, measured using bulk metabolic labeling and a novel flow cytometry assay to quantify responses at a single-cell level. These translational responses were suppressed by inhibitors of BTK (ibrutinib) and SYK (tamatinib). Anti-IgM-induced mRNA translation was associated with increased expression of translation initiation factors eIF4A and eIF4GI, and reduced expression of the eIF4A inhibitor, PDCD4. Anti-IgM also increased mRNA translation in normal blood B cells, but without clear modulatory effects on these factors. In addition, anti-IgM increased translation of mRNA-encoding MYC, a major driver of disease progression. mRNA translation is likely to be an important mediator of the growth-promoting effects of antigen stimulation acting, at least in part, via translational induction of MYC. Differences in mechanisms of translational regulation in CLL and normal B cells may provide opportunities for selective therapeutic attack.


Asunto(s)
Regulación Leucémica de la Expresión Génica , Leucemia Linfocítica Crónica de Células B/genética , Proteínas Proto-Oncogénicas c-myc/genética , ARN Mensajero/genética , Receptores de Antígenos de Linfocitos B/inmunología , Adenina/análogos & derivados , Agammaglobulinemia Tirosina Quinasa , Anticuerpos Antiidiotipos/inmunología , Linfocitos B/metabolismo , Linfocitos B/patología , Línea Celular Tumoral , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/patología , Piperidinas , Biosíntesis de Proteínas/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirazoles/farmacología , Pirimidinas/farmacología , Receptores de Antígenos de Linfocitos B/genética , Quinasa Syk , Células Tumorales Cultivadas
16.
J Invest Dermatol ; 134(3): 695-703, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24005050

RESUMEN

Langerhans cells (LCs) are professional antigen-presenting cells (APCs) residing in the epidermis. Despite their high potential to activate T lymphocytes, current understanding of human LC biology is limited. Genome-wide comparison of the transcriptional profiles of human skin migratory CD1a+ LCs and CD11c+ dermal dendritic cells (DDCs) demonstrated significant differences between these "dendritic cell (DC)" types, including preferential expression of 625 genes (P<0.05) in LC and 914 genes (P<0.05) in DDC. Analysis of the temporal regulation of molecular networks activated after stimulation with tumor necrosis factor-α (TNF-α) confirmed the unique molecular signature of LCs. Although LCs conformed to the phenotype of professional APC, inflammatory signaling activated primarily genes associated with cellular metabolism and mitochondrial activation (e.g., CYB561 and MRPS35), cell membrane re-organization, and antigen acquisition and degradation (CAV1 and PSMD14; P<0.05-P<0.0001). Conversely, TNF-α induced classical activation in DDCs with early downregulation of surface receptors (mannose receptor-1 (MRC1) and C-type lectins), and subsequent upregulation of cytokines, chemokines (IL1a, IL1b, and CCL18), and matrix metalloproteinases (MMP1, MMP3, and MMP9; P<0.05-P<0.0001). Functional interference of caveolin abrogated LCs superior ability to cross-present antigens to CD8+ T lymphocytes, highlighting the importance of these networks to biological function. Taken together, these observations support the idea of distinct biological roles of cutaneous DC types.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Células Dendríticas/inmunología , Regulación de la Expresión Génica/inmunología , Células de Langerhans/inmunología , Piel/inmunología , Linfocitos T/inmunología , Antígenos CD1/inmunología , Antígenos CD1/metabolismo , Antígeno CD11c/metabolismo , Caveolina 1/genética , Caveolina 1/inmunología , Caveolina 1/metabolismo , Movimiento Celular/inmunología , Células Dendríticas/citología , Células Dendríticas/metabolismo , Dermis/citología , Dermis/inmunología , Células Epidérmicas , Epidermis/inmunología , Humanos , Células de Langerhans/citología , Células de Langerhans/metabolismo , Piel/citología , Transcriptoma
17.
Eur J Immunol ; 42(1): 256-63, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22002320

RESUMEN

A major contributing factor to the final magnitude and breadth of CD8(+) T-cell responses to complex antigens is immunodomination, where CD8(+) T cells recognizing their cognate ligand inhibit the proliferation of other CD8(+) T cells engaged with the same APC. In this study, we examined how the half-life of cell surface peptide-MHC class I complexes influences this phenomenon. We found that primary CD8(+) T-cell responses to DNA vaccines in mice are shaped by competition among responding CD8(+) T cells for nonspecific stimuli early after activation and prior to cell division. The susceptibility of CD8(+) T cells to 'domination' was a direct correlate of higher kinetic stability of the competing CD8(+) T-cell cognate ligand. When high affinity competitive CD8(+) T cells were deleted by self-antigen expression, competition was abrogated. These findings show, for the first time to our knowledge, the existence of regulatory mechanisms that direct the responding CD8(+) T-cell repertoire toward epitopes with high-stability interactions with MHC class I molecules. They also provide an insight into factors that facilitate CD8(+) T-cell coexistence, with important implications for vaccine design and delivery.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Vacunas de ADN/inmunología , Animales , Células Presentadoras de Antígenos/citología , Linfocitos T CD8-positivos/citología , Epítopos de Linfocito T/inmunología , Semivida , Inmunización , Epítopos Inmunodominantes/inmunología , Cinética , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
18.
Eur J Immunol ; 40(4): 976-85, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20104490

RESUMEN

Antigen-specific CD4(+) T cells are central to natural and vaccine-induced immunity. An ongoing antigen-specific T-cell response can, however, influence surrounding T cells with unrelated antigen specificities. We previously observed this bystander effect in healthy human subjects following recall vaccination with tetanus toxoid (TT). Since this interplay could be important for maintenance of memory, we have moved to a mouse model for further analysis. We investigated whether boosting memory CD4(+) T cells against TT in vivo would influence injected CD4(+) TCR transgenic T cells (OT-II) specific for an unrelated OVA peptide. If OT-II cells were pre-activated with OVA peptide in vitro, these cells showed a bystander proliferative response during the ongoing parallel TT-specific response. Bystander proliferation was dependent on boosting of the TT-specific memory response in the recipients, with no effect in naive mice. Bystander stimulation was also proportional to the strength of the TT-specific memory T-cell response. T cells activated in vitro displayed functional receptors for IL-2 and IL-7, suggesting these as potential mediators. This crosstalk between a stimulated CD4(+) memory T-cell response and CD4(+) T cells activated by an unrelated antigen could be important in human subjects continually buffeted by environmental antigens.


Asunto(s)
Efecto Espectador/inmunología , Linfocitos T CD4-Positivos/inmunología , Inmunización Secundaria , Memoria Inmunológica/inmunología , Activación de Linfocitos , Subgrupos Linfocitarios/inmunología , Traslado Adoptivo , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Citocinas/farmacología , Humanos , Subunidad gamma Común de Receptores de Interleucina/inmunología , Subunidad alfa del Receptor de Interleucina-2/inmunología , Subunidad beta del Receptor de Interleucina-2/inmunología , Interleucinas/biosíntesis , Interleucinas/genética , Interleucinas/farmacología , Interleucinas/fisiología , Subgrupos Linfocitarios/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ovalbúmina/inmunología , Fragmentos de Péptidos/inmunología , Receptores de Interleucina/inmunología , Proteínas Recombinantes/farmacología , Especificidad del Receptor de Antígeno de Linfocitos T , Toxoide Tetánico/inmunología
19.
Eur J Immunol ; 38(2): 364-9, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18196518

RESUMEN

Peptide loading of MHC class I molecules involves multiple cofactors including tapasin. We showed previously in vitro that tapasin edits the peptide repertoire by favoring the binding of peptides with slow dissociation rates. Here, using tapasin-deficient mice and a DNA vaccine that primes directly, we confirm that tapasin establishes hierarchical responses in vivo according to peptide-MHC stability. In contrast, this hierarchy is lost when the peptides are cross-presented via an alternative DNA vaccine. By regulating transgene expression, we found that the dominant response modifier was antigen persistence. Our findings reveal strategies for activating T cells against low-affinity peptides, of potential importance for patients with repertoires narrowed by deletional tolerance.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Proteínas del Huevo/farmacocinética , Antígenos de Histocompatibilidad Clase I/metabolismo , Epítopos Inmunodominantes/metabolismo , Proteínas de Transporte de Membrana/fisiología , Ovalbúmina/farmacocinética , Animales , Reactividad Cruzada/inmunología , Proteínas del Huevo/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Epítopos Inmunodominantes/inmunología , Proteínas de Transporte de Membrana/deficiencia , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/farmacocinética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ovalbúmina/inmunología , Fragmentos de Péptidos , Transducción de Señal/inmunología , Vacunas de ADN/administración & dosificación , Vacunas de ADN/inmunología , Vacunas de ADN/farmacocinética
20.
J Immunol ; 179(12): 8313-21, 2007 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-18056376

RESUMEN

After priming, naive T cells undergo a program of expansion, contraction, and memory formation. Numerous studies have indicated that only a brief period of antigenic stimulation is required to fully commit CD8+ T cells to this program. Nonetheless, the persistence of Ag may modulate the eventual fate of CD8+ T cells. Using DNA delivery, we showed previously that direct presentation primes high levels of effector CD8+ T cells as compared with cross-presentation. One explanation now revealed is that prolonged cross-presentation limits effector cell expansion and function. To analyze this, we used a drug-responsive system to regulate Ag expression after DNA injection. Reducing expression to a single burst expanded greater numbers of peptide-specific effector CD8+ T cells than sustained Ag. Consequences for memory development were assessed after boosting and showed that, although persistent Ag maintained higher numbers of tetramer-positive CD8+ T cells, these expanded less (approximately 4-fold) than those induced by transient Ag expression (approximately 35-fold). Transient expression at priming therefore led to a net higher secondary response. In terms of vaccine design, we propose that the most effective DNA-based CD8+ T cell vaccines will be those that deliver a short burst of Ag.


Asunto(s)
Presentación de Antígeno , Linfocitos T CD8-positivos/inmunología , Reactividad Cruzada , Memoria Inmunológica , Vacunas de ADN/inmunología , Animales , Ratones , Ratones Endogámicos C57BL , Vacunación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...