Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Cell Rep Med ; 4(1): 100878, 2023 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-36599350

RESUMEN

Although immune checkpoint inhibitors (ICIs) are established as effective cancer therapies, overcoming therapeutic resistance remains a critical challenge. Here we identify interleukin 6 (IL-6) as a correlate of poor response to atezolizumab (anti-PD-L1) in large clinical trials of advanced kidney, breast, and bladder cancers. In pre-clinical models, combined blockade of PD-L1 and the IL-6 receptor (IL6R) causes synergistic regression of large established tumors and substantially improves anti-tumor CD8+ cytotoxic T lymphocyte (CTL) responses compared with anti-PD-L1 alone. Circulating CTLs from cancer patients with high plasma IL-6 display a repressed functional profile based on single-cell RNA sequencing, and IL-6-STAT3 signaling inhibits classical cytotoxic differentiation of CTLs in vitro. In tumor-bearing mice, CTL-specific IL6R deficiency is sufficient to improve anti-PD-L1 activity. Thus, based on both clinical and experimental evidence, agents targeting IL-6 signaling are plausible partners for combination with ICIs in cancer patients.


Asunto(s)
Antineoplásicos , Interleucina-6 , Neoplasias , Animales , Ratones , Antineoplásicos/uso terapéutico , Antígeno B7-H1/inmunología , Antígeno B7-H1/uso terapéutico , Linfocitos T CD8-positivos/metabolismo , Inmunoterapia , Interleucina-6/metabolismo , Neoplasias/inmunología , Neoplasias/terapia
2.
PLoS One ; 16(12): e0262198, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34972191

RESUMEN

BACKGROUND: Colorectal cancer (CRC) is a leading cause of cancer-related deaths, with a 5% 5-year survival rate for metastatic disease, yet with limited therapeutic advancements due to insufficient understanding of and inability to accurately capture high-risk CRC patients who are most likely to recur. We aimed to improve high-risk classification by identifying biological pathways associated with outcome in adjuvant stage II/III CRC. METHODS AND FINDINGS: We included 1062 patients with stage III or high-risk stage II colon carcinoma from the prospective three-arm randomized phase 3 AVANT trial, and performed expression profiling to identify a prognostic signature. Data from validation cohort GSE39582, The Cancer Genome Atlas, and cell lines were used to further validate the prognostic biology. Our retrospective analysis of the adjuvant AVANT trial uncovered a prognostic signature capturing three biological functions-stromal, proliferative and immune-that outperformed the Consensus Molecular Subtypes (CMS) and recurrence prediction signatures like Oncotype Dx in an independent cohort. Importantly, within the immune component, high granzyme B (GZMB) expression had a significant prognostic impact while other individual T-effector genes were less or not prognostic. In addition, we found GZMB to be endogenously expressed in CMS2 tumor cells and to be prognostic in a T cell independent fashion. A limitation of our study is that these results, although robust and derived from a large dataset, still need to be clinically validated in a prospective study. CONCLUSIONS: This work furthers our understanding of the underlying biology that propagates stage II/III CRC disease progression and provides scientific rationale for future high-risk stratification and targeted treatment evaluation in biomarker defined subpopulations of resectable high-risk CRC. Our results also shed light on an alternative GZMB source with context-specific implications on the disease's unique biology.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Granzimas/fisiología , Transcriptoma , Adulto , Anciano , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Análisis por Conglomerados , Neoplasias Colorrectales/mortalidad , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genoma Humano , Granzimas/química , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Pronóstico , Modelos de Riesgos Proporcionales , Estudios Prospectivos , Estudios Retrospectivos , Riesgo , Linfocitos T/metabolismo , Resultado del Tratamiento
4.
Nat Med ; 26(5): 693-698, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32405063

RESUMEN

Although elevated plasma interleukin-8 (pIL-8) has been associated with poor outcome to immune checkpoint blockade 1, this has not been comprehensively evaluated in large randomized studies. Here we analyzed circulating pIL-8 and IL8 gene expression in peripheral blood mononuclear cells and tumors of patients treated with atezolizumab (anti-PD-L1 monoclonal antibody) from multiple randomized trials representing 1,445 patients with metastatic urothelial carcinoma (mUC) and metastatic renal cell carcinoma. High levels of IL-8 in plasma, peripheral blood mononuclear cells and tumors were associated with decreased efficacy of atezolizumab in patients with mUC and metastatic renal cell carcinoma, even in tumors that were classically CD8+ T cell inflamed. Low baseline pIL-8 in patients with mUC was associated with increased response to atezolizumab and chemotherapy. Patients with mUC who experienced on-treatment decreases in pIL-8 exhibited improved overall survival when treated with atezolizumab but not with chemotherapy. Single-cell RNA sequencing of the immune compartment showed that IL8 is primarily expressed in circulating and intratumoral myeloid cells and that high IL8 expression is associated with downregulation of the antigen-presentation machinery. Therapies that can reverse the impacts of IL-8-mediated myeloid inflammation will be essential for improving outcomes of patients treated with immune checkpoint inhibitors.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Antígeno B7-H1/antagonistas & inhibidores , Interleucina-8/metabolismo , Neoplasias/diagnóstico , Neoplasias/tratamiento farmacológico , Adulto , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antígeno B7-H1/inmunología , Biomarcadores Farmacológicos/sangre , Biomarcadores Farmacológicos/metabolismo , Biomarcadores de Tumor/sangre , Biomarcadores de Tumor/metabolismo , Carcinoma de Células Renales/diagnóstico , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/mortalidad , Carcinoma de Células Transicionales/diagnóstico , Carcinoma de Células Transicionales/tratamiento farmacológico , Carcinoma de Células Transicionales/metabolismo , Carcinoma de Células Transicionales/mortalidad , Resistencia a Antineoplásicos , Femenino , Humanos , Interleucina-8/sangre , Neoplasias Renales/diagnóstico , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/metabolismo , Neoplasias Renales/mortalidad , Masculino , Neoplasias/metabolismo , Neoplasias/mortalidad , Pronóstico , Análisis de Supervivencia , Insuficiencia del Tratamiento , Neoplasias Urológicas/diagnóstico , Neoplasias Urológicas/tratamiento farmacológico , Neoplasias Urológicas/metabolismo , Neoplasias Urológicas/mortalidad
5.
J Clin Oncol ; 33(25): 2735-44, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26124478

RESUMEN

PURPOSE: The AVAglio (Avastin in Glioblastoma) and RTOG-0825 randomized, placebo-controlled phase III trials in newly diagnosed glioblastoma reported prolonged progression-free survival (PFS), but not overall survival (OS), with the addition of bevacizumab to radiotherapy plus temozolomide. To establish whether certain patient subgroups derived an OS benefit from the addition of bevacizumab to first-line standard-of-care therapy, AVAglio patients were retrospectively evaluated for molecular subtype, and bevacizumab efficacy was assessed for each patient subgroup. PATIENTS AND METHODS: A total of 349 pretreatment specimens (bevacizumab arm, n = 171; placebo arm, n = 178) from AVAglio patients (total, N = 921) were available for biomarker analysis. Samples were profiled for gene expression and isocitrate dehydrogenase 1 (IDH1) mutation status and classified into previously identified molecular subtypes. PFS and OS were assessed within each subtype. RESULTS: A multivariable analysis accounting for prognostic covariates revealed that bevacizumab conferred a significant OS advantage versus placebo for patients with proneural IDH1 wild-type tumors (17.1 v 12.8 months, respectively; hazard ratio, 0.43; 95% CI, 0.26 to 0.73; P = .002). This analysis also revealed an interaction between the proneural subtype biomarker and treatment arm (P = .023). The group of patients with mesenchymal and proneural tumors derived a PFS benefit from bevacizumab compared with placebo; however, this translated to an OS benefit in the proneural subset only. CONCLUSION: Retrospective analysis of AVAglio data suggests that patients with IDH1 wild-type proneural glioblastoma may derive an OS benefit from first-line bevacizumab treatment. The predictive value of the proneural subtype observed in AVAglio should be validated in an independent data set.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Antineoplásicos Alquilantes/uso terapéutico , Bevacizumab/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/mortalidad , Dacarbazina/análogos & derivados , Glioblastoma/tratamiento farmacológico , Glioblastoma/mortalidad , Adulto , Anciano , Neoplasias Encefálicas/radioterapia , Ensayos Clínicos Fase III como Asunto , Dacarbazina/uso terapéutico , Supervivencia sin Enfermedad , Femenino , Perfilación de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Glioblastoma/radioterapia , Humanos , Isocitrato Deshidrogenasa/genética , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Mutación , Estadificación de Neoplasias , Pronóstico , Radioterapia Adyuvante , Ensayos Clínicos Controlados Aleatorios como Asunto , Estudios Retrospectivos , Temozolomida , Resultado del Tratamiento
6.
J Transl Med ; 11: 76, 2013 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-23522020

RESUMEN

BACKGROUND: The phosphatidylinositol 3-kinase (PI3K) pathway plays an important role in multiple myeloma (MM), a blood cancer associated with uncontrolled proliferation of bone marrow plasma cells. This study aimed to develop a robust clinical pharmacodynamic (PD) assay to measure the on-target PD effects of the selective PI3K inhibitor GDC-0941 in MM patients. METHODS: We conducted an in vitro drug wash-out study to evaluate the feasibility of biochemical approaches in measuring the phosphorylation of S6 ribosomal protein (S6), one of the commonly used PD markers for PI3K pathway inhibition. We then developed a 7-color phospho-specific flow cytometry assay, or phospho flow assay, to measure the phosphorylation state of intracellular S6 in bone marrow aspirate (BMA) and peripheral blood (PB). Integrated mean fluorescence intensity (iMFI) was used to calculate fold changes of phosphorylation. Assay sensitivity was evaluated by comparing phospho flow with Meso Scale Discovery (MSD) and immunohistochemistry (IHC) assays. Finally, a sample handling method was developed to maintain the integrity of phospho signal during sample shipping and storage to ensure clinical application. RESULTS: The phospho flow assay provided single-cell PD monitoring of S6 phosphorylation in tumor and surrogate cells using fixed BMA and PB, assessing pathway modulation in response to GDC-0941 with sensitivity similar to that of MSD assay. The one-shot sample fixation and handling protocol herein demonstrated exceptional preservation of protein phosphorylation. In contrast, the IHC assay was less sensitive in terms of signal quantification while the biochemical approach (MSD) was less suitable to assess PD activities due to the undesirable impact associated with cell isolation on the protein phosphorylation in tumor cells. CONCLUSIONS: We developed a robust PD biomarker assay for the clinical evaluation of PI3K inhibitors in MM, allowing one to decipher the PD response in a relevant cell population. To our knowledge, this is the first report of an easily implemented clinical PD assay that incorporates an unbiased one-shot sample handling protocol, all (staining)-in-one (tube) phospho flow staining protocol, and an integrated modified data analysis for PD monitoring of kinase inhibitors in relevant cell populations in BMA and PB. The methods described here ensure a real-time, reliable and reproducible PD readout, which can provide information for dose selection as well as help to identify optimal combinations of targeted agents in early clinical trials.


Asunto(s)
Antineoplásicos/farmacología , Ensayos de Selección de Medicamentos Antitumorales/métodos , Inhibidores Enzimáticos/farmacología , Citometría de Flujo/métodos , Mieloma Múltiple/tratamiento farmacológico , Inhibidores de las Quinasa Fosfoinosítidos-3 , Biomarcadores de Tumor/metabolismo , Médula Ósea/metabolismo , Células de la Médula Ósea/efectos de los fármacos , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Indazoles/farmacología , Fosfoproteínas/metabolismo , Fosforilación , Sulfonamidas/farmacología
7.
Mol Cancer Ther ; 12(6): 1122-30, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23536720

RESUMEN

The objective of this study was to evaluate circulating hepatocyte growth factor (cHGF) as a pharmacodynamic biomarker of Met inhibition for onartuzumab (MetMAb, OA5D5v2) in a phase I trial in patients with advanced cancers and a phase II trial in non-small cell lung cancer (NSCLC). The phase I study was a dose escalation trial with onartuzumab administered i.v. once every three weeks. The phase II study was a randomized two-arm trial in which onartuzumab or placebo was administered in combination with erlotinib in 137 patients with second and third line (2/3L) NSCLC. cHGF levels were evaluated by ELISA at multiple time points over the treatment period. Onartuzumab administration resulted in an acute and sustained rise in cHGF in both the phase I and phase II studies. Elevation in cHGF was independent of dose or drug exposure and was restricted to onartuzumab treatment. Neither higher baseline nor elevated change in cHGF levels upon treatment could simply be attributed to tumor burden or number of liver metastasis. We have shown that elevated cHGF can consistently and reproducibly be measured as a pharmacodynamic biomarker of onartuzumab activity. The elevation in cHGF is independent of tumor type, dose administered, or dose duration. Although these studies were not powered to directly address the contribution of cHGF as a predictive, on-treatment, circulating biomarker, these data suggest that measurement of cHGF in future expanded studies is warranted.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Factor de Crecimiento de Hepatocito/sangre , Neoplasias Pulmonares/tratamiento farmacológico , Células Neoplásicas Circulantes/metabolismo , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Biomarcadores de Tumor/sangre , Carcinoma de Pulmón de Células no Pequeñas/sangre , Carcinoma de Pulmón de Células no Pequeñas/patología , Clorhidrato de Erlotinib , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Factor de Crecimiento de Hepatocito/biosíntesis , Humanos , Neoplasias Pulmonares/sangre , Neoplasias Pulmonares/patología , Masculino , Estadificación de Neoplasias , Proteínas Proto-Oncogénicas c-met/biosíntesis , Proteínas Proto-Oncogénicas c-met/genética , Quinazolinas/administración & dosificación
8.
Acta Crystallogr Sect E Struct Rep Online ; 68(Pt 7): m923, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22807757

RESUMEN

In the title complex, [Mn(C6H2N3O7)2(C30H26N4O)], the Mn(II) atom is coordinated by the tridentate bis-benzimidazole ligand and two atoms of the picrate anions, in a distorted square-pyramidal geometry (τ = 0.038). One nitro O atom of one picrate ion is disordered over two sites with occupancies of 0.54 (5) and 0.46 (5).

9.
Clin Cancer Res ; 18(11): 3090-9, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22496205

RESUMEN

PURPOSE: GDC-0973 is a potent and selective mitogen-activated protein (MAP)/extracellular signal-regulated kinase (ERK) kinase (MEK) inhibitor. Pharmacokinetic-pharmacodynamic (PK-PD) modeling was used to relate GDC-0973 plasma and tumor concentrations, tumor pharmacodynamics and antitumor efficacy to establish pharmacokinetic endpoints and predict active doses in the clinic. EXPERIMENTAL DESIGN: A PK-PD model was used to characterize GDC-0973 tumor disposition and in vivo potency in WM-266-4 xenograft mice. Simulations were conducted using the PK-PD model along with human pharmacokinetics to identify a target plasma concentration and predict active doses. In vivo potency and antitumor efficacy were characterized in A375 melanoma xenograft mice, and a population-based integrated PK-PD-efficacy model was used to relate tumor pharmacodynamics (%pERK decrease) to antitumor activity. RESULTS: GDC-0973 showed a sustained tumor pharmacodynamic response due to longer residence in tumor than in plasma. Following single doses of GDC-0973, estimated in vivo IC(50) values of %pERK decrease based on tumor concentrations in xenograft mice were 0.78 (WM-266-4) and 0.52 µmol/L (A375). Following multiple doses of GDC-0973, the estimated in vivo IC(50) value in WM-266-4 increased (3.89 µmol/L). Human simulations predicted a minimum target plasma concentration of 83 nmol/L and an active dose range of 28 to 112 mg. The steep relationship between tumor pharmacodynamics (%pERK decrease) and antitumor efficacy suggests a pathway modulation threshold beyond which antitumor efficacy switches on. CONCLUSIONS: Clinical observations of %pERK decrease and antitumor activity were consistent with model predictions. This article illustrates how PK-PD modeling can improve the translation of preclinical data to humans by providing a means to integrate preclinical and early clinical data.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/farmacocinética , Azetidinas/farmacología , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Piperidinas/farmacología , Animales , Antineoplásicos/uso terapéutico , Azetidinas/farmacocinética , Línea Celular Tumoral , Humanos , Ratones , Ratones Desnudos , Quinasas de Proteína Quinasa Activadas por Mitógenos/farmacocinética , Quinasas de Proteína Quinasa Activadas por Mitógenos/farmacología , Modelos Biológicos , Piperidinas/farmacocinética , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Mol Cancer Ther ; 7(9): 2599-608, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18790743

RESUMEN

The insulin-like growth factor (IGF) system consists of two ligands (IGF-I and IGF-II), which both signal through IGF-I receptor (IGF-IR) to stimulate proliferation and inhibit apoptosis, with activity contributing to malignant growth of many types of human cancers. We have developed a humanized, affinity-matured anti-human IGF-IR monoclonal antibody (h10H5), which binds with high affinity and specificity to the extracellular domain. h10H5 inhibits IGF-IR-mediated signaling by blocking IGF-I and IGF-II binding and by inducing cell surface receptor down-regulation via internalization and degradation, with the extracellular and intracellular domains of IGF-IR being differentially affected by the proteasomal and lysosomal inhibitors. In vitro, h10H5 exhibits antiproliferative effects on cancer cell lines. In vivo, h10H5 shows single-agent antitumor efficacy in human SK-N-AS neuroblastoma and SW527 breast cancer xenograft models and even greater efficacy in combination with the chemotherapeutic agent docetaxel or an anti-vascular endothelial growth factor antibody. Antitumor activity of h10H5 is associated with decreased AKT activation and glucose uptake and a 316-gene transcription profile with significant changes involving DNA metabolic and cell cycle machineries. These data support the clinical testing of h10H5 as a biotherapeutic for IGF-IR-dependent human tumors and furthermore illustrate a new method of monitoring its activity noninvasively in vivo via 2-fluoro-2-deoxy-d-glucose-positron emission tomography imaging.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Glucosa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor IGF Tipo 1/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Biomarcadores/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Sinergismo Farmacológico , Endocitosis/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Femenino , Perfilación de la Expresión Génica , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Neoplasias/enzimología , Neoplasias/genética , Neoplasias/patología , Inhibidores de Proteasoma , Subunidades de Proteína/metabolismo , Transducción de Señal/efectos de los fármacos
11.
Spectrochim Acta A Mol Biomol Spectrosc ; 71(2): 650-4, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18296107

RESUMEN

A tri-nuclear cobalt(II) cluster, [(CoL)2(OAc)2Co].2C2H5OH, has been synthesized by the reaction of cobalt(II) acetate tetrahydrate with a novel Salen-type bisoxime chelating ligand, 3,3'-dimethoxy-2,2'-[ethylenedioxybis(nitrilomethylidyne)]diphenol (H2L), and characterized by elemental analyses, IR spectra, TG-DTA and molar conductances. UV-vis spectroscopic titration in methanol solution clearly indicated the exclusive formation of the 3:2 [Co3L2]2+ cluster. The single-crystal X-ray diffraction determination of the Co(II) cluster shows that there are two acetate ions coordinate to three cobalt(II) ions through Co-O-C-O-Co bridges, and quadruple mu-phenoxo oxygen atoms from two [CoL] chelates also coordinate to cobalt(II) ions. Interestingly, different conformational central ions: five- and six-coordinated geometries were found in the cluster.


Asunto(s)
Acetona/química , Cobalto/química , Etanol/química , Cristalografía por Rayos X , Modelos Moleculares , Estructura Molecular , Espectrofotometría , Temperatura , Volumetría
12.
J Immunol ; 172(12): 7324-34, 2004 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15187108

RESUMEN

To identify novel components of the TCR signaling pathway, a large-scale retroviral-based functional screen was performed using CD69 expression as a marker for T cell activation. In addition to known regulators, two truncated forms of p21-activated kinase 2 (PAK2), PAK2DeltaL(1-224) and PAK2DeltaS(1-113), both lacking the kinase domain, were isolated in the T cell screen. The PAK2 truncation, PAK2DeltaL, blocked Ag receptor-induced NFAT activation and TCR-mediated calcium flux in Jurkat T cells. However, it had minimal effect on PMA/ionomycin-induced CD69 up-regulation in Jurkat cells, on anti-IgM-mediated CD69 up-regulation in B cells, or on the migratory responses of resting T cells to chemoattractants. We show that PAK2 kinase activity is increased in response to TCR stimulation. Furthermore, a full-length kinase-inactive form of PAK2 blocked both TCR-induced CD69 up-regulation and NFAT activity in Jurkat cells, demonstrating that kinase activity is required for PAK2 function downstream of the TCR. We also generated a GFP-fused PAK2 truncation lacking the Cdc42/Rac interactive binding region domain, GFP-PAK2(83-149). We show that this construct binds directly to the kinase domain of PAK2 and inhibits anti-TCR-stimulated T cell activation. Finally, we demonstrate that, in primary T cells, dominant-negative PAK2 prevented anti-CD3/CD28-induced IL-2 production, and TCR-induced CD40 ligand expression, both key functions of activated T cells. Taken together, these results suggest a novel role for PAK2 as a positive regulator of T cell activation.


Asunto(s)
Activación de Linfocitos , Proteínas Nucleares , Proteínas Serina-Treonina Quinasas/inmunología , Linfocitos T/inmunología , Antígenos CD/análisis , Antígenos CD/biosíntesis , Antígenos de Diferenciación de Linfocitos T/análisis , Antígenos de Diferenciación de Linfocitos T/biosíntesis , Linfocitos B/metabolismo , Biomarcadores/análisis , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Humanos , Lectinas Tipo C , Mutación , Factores de Transcripción NFATC , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , Estructura Terciaria de Proteína , Receptores de Antígenos de Linfocitos T/fisiología , Transducción de Señal , Linfocitos T/metabolismo , Factores de Transcripción/metabolismo , Quinasas p21 Activadas
13.
Inflammation ; 24(3): 195-205, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23255840

RESUMEN

Interferon gamma (IFNγ)-mediated activation of the myelomonocytic cell line HL-60 and peripheral blood monocytes will induce expression of an epitope for the monoclonal antibody which recognizes human αEß7 integrin, HML-1. Here, we found that the anti-human αEß7 monoclonal antibody Ber-ACT8 did not recognize IFNγ-stimulated, HML-1 positive HL-60 cells. Culture of blood monocytes with IFNγ also induced expression of HML-1 but not Ber-ACT8 epitopes. Moreover, migration of monocytes across a monolayer of human airway epithelial cells rapidly induced expression of HML-1, with no detectable Ber-ACT8 staining. Using two different sets of primers specific for the human αE integrin gene, we were unable to detect αE mRNA within HL-60 cells or isolated monocytes by reverse transcriptase polymerase chain reaction methods. We conclude that reactivity of the HML-1 antibody for HL-60 cells and monocytes does not correspond with the expression of the αE integrin subunit, and instead detects a marker for cellular activation.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA