Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Más filtros

Medicinas Complementárias
Tipo del documento
Intervalo de año de publicación
1.
Medicine (Baltimore) ; 99(45): e22859, 2020 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-33157928

RESUMEN

BACKGROUND: This article will evaluate the effects of traditional Chinese medicine (TCM) combined with chemotherapy on the immune function and quality of life of patients with non-small cell lung cancer (NSCLC), and evaluate the published side effects. METHODS: The systematic review and meta-analysis will be conducted in accordance with the Preferred Reporting Items for Systematic Review and Meta-Analysis guidelines. The databases we will search include: PubMed, EMBASE, Cochrane Library, Web of Science, China National Knowledge Infrastructure, China Biomedicine, Wan fang Data, and Technology Periodical Database. The search date is from inception to June 30, 2020. There are no restrictions on the document language. The literatures included in this study are randomized controlled trials. The main results include ratio of CD3, CD4, CD8, CD4/CD8, NK cells, the level of IgA, IgG, IgM, and Karnofsky performance status score. The secondary result is to evaluate various side effects during treatment. We will use the Cochrane Collaboration tool to evaluate each study and use Review Manager software (RevMan, version 5.3) to merge and analyze the data. The 2 researchers will independently cross-screen the literature, extract data, and evaluate the quality. If there are differences, we will resolve them through discussion or consultation with a third reviewer. RESULTS: The results of this study will provide high-quality evidence for the effect of TCM combined with chemotherapy on the immune function and quality of life of patients with NSCLC. CONCLUSION: This article will comprehensively evaluate the effects of TCM combined with chemotherapy on the immune function and quality of life of patients with NSCLC, and provide evidence-based evidence for clinical practice. ETHICS: Since the data used in this study is based on previous trials and does not involve patient privacy, ethical approval is not required. STUDY REGISTRATION NUMBER: INPLASY202070071.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/terapia , Neoplasias Pulmonares/terapia , Medicina Tradicional China , Calidad de Vida , Antineoplásicos/uso terapéutico , Complejo CD3/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/sangre , Humanos , Isotipos de Inmunoglobulinas/sangre , Células Asesinas Naturales/metabolismo , Neoplasias Pulmonares/sangre , Metaanálisis como Asunto , Proyectos de Investigación , Revisiones Sistemáticas como Asunto
2.
Food Funct ; 11(7): 5992-6006, 2020 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-32697211

RESUMEN

Ziziphus Jujuba cv. Pozao has been consumed as a traditional fruit with regional characteristics in China for a long time; however, fewer studies on polysaccharides from Ziziphus Jujuba cv. Pozao (JP) have been documented. This study aimed to evaluate the effect of oral administration of JP on cyclophosphamide-induced ICR mice for 28 days. The results showed that oral administration of JP could significantly improve the lymphocyte proliferation in the spleen and decrease the proportion of CD3+ and CD4+ and the ratio of CD4+/CD8+ in cyclophosphamide-induced mice in a dose-dependent manner. JP treatment also increased the levels of IL-2, IL-4, IL-10, IFN-γ, and TNF-α in serum and the intestine, and the improvement effects were proportional to the dose of JP. Similarly, JP significantly increased the levels of IgA and SIgA, as well as the expressions of Claudin-1 and Occludin in the intestine. Particularly, the expressions of Claudin-1 and Occludin were the best in the M-JP group. Furthermore, JP positively regulated the gut microbiota as indicated by the enriched microbiota diversity. At the phylum level, the relative abundance of Firmicutes was significantly decreased by JP, while that of Bacteroidetes was increased by JP treatment. More importantly, the ratio of Firmicutes/Bacteroidetes was significantly increased. And a high dose of JP is the most effective. At the genus level, the abundances of the Bacteroidales-S24-7-group, Lachnospiraceae, Alloprevotella, Alistipes and Bacteroides were increased by JP treatment. These results provided evidence for the regulating effect of JP on the peripheral immunity and intestinal barrier function in cyclophosphamide-induced hypoimmune mice.


Asunto(s)
Suplementos Dietéticos , Microbioma Gastrointestinal/efectos de los fármacos , Inmunidad/efectos de los fármacos , Intestinos/efectos de los fármacos , Linfocitos/metabolismo , Polisacáridos/farmacología , Ziziphus/química , Animales , Bacterias/efectos de los fármacos , Complejo CD3/metabolismo , Antígenos CD4/metabolismo , Relación CD4-CD8 , Claudina-1/metabolismo , Ciclofosfamida , Citocinas/metabolismo , Frutas/química , Inmunoglobulina A/metabolismo , Inmunoglobulina A Secretora/metabolismo , Intestinos/microbiología , Intestinos/fisiología , Masculino , Ratones , Ocludina/metabolismo , Extractos Vegetales/farmacología , Bazo/efectos de los fármacos , Bazo/metabolismo
3.
World J Urol ; 37(3): 539-544, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30027406

RESUMEN

PURPOSE: To evaluate the effect of hexanic extract of Serenoa repens (HESr) on prostatic inflammation in patients with diagnosed prostatic inflammation. METHODS: Patients with prostatic inflammation histologically confirmed by TRUS prostatic biopsy were randomized either to receive HESr (320 mg/day) or no treatment. A second biopsy was performed 6 months later according to standard clinical practice. Inflammation was assessed by the Irani's score and immunohistochemical staining using the CD3, CD4 and CD8 (for T-leucocytes), CD20 (for B-leucocytes) and CD163 (for macrophages) antibodies. RESULTS: Overall 97 patients were eligible for analysis. In the HESr group the mean inflammation grading and aggressiveness grading score significantly decreased from 1.55 and 1.55 at baseline to 0.79 (p = 0.001) and 0.87 (p = 0.001) at the second biopsy, respectively. In the control group the mean inflammation grading score was 1.44 at first biopsy and 1.23 at the second biopsy. The mean aggressiveness gradings core was 1.09 and 0.89, respectively. No statistical significance was found (p = 0.09 and p = 0.74).The mean decrease in all inflammation scores was statistically higher in the HESr patients compared to controls. The immunohistochemical staining showed a significant change in the expression of the analyzed antibodies for the HESr patients compared to the first biopsy. In the nontreatment group, no significant difference was found at the second biopsy. The change in expression of each antibody in the HESr group was statistical significant compared to control. CONCLUSIONS: HESr seems to reduce prostatic inflammation in terms of histological and immunohistochemical parameters in this specific patients population.


Asunto(s)
Linfocitos B/patología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/patología , Macrófagos/patología , Fitoterapia , Extractos Vegetales/uso terapéutico , Próstata/patología , Prostatitis/tratamiento farmacológico , Serenoa , Anciano , Antígenos CD/metabolismo , Antígenos CD20/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Linfocitos B/inmunología , Linfocitos B/metabolismo , Biopsia , Complejo CD3/metabolismo , Antígenos CD4/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Antígenos CD8/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Hexanos , Humanos , Inflamación , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Persona de Mediana Edad , Próstata/inmunología , Próstata/metabolismo , Prostatitis/inmunología , Prostatitis/metabolismo , Prostatitis/patología , Receptores de Superficie Celular/metabolismo
4.
Sci Transl Med ; 10(459)2018 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-30232226

RESUMEN

Severe fever with thrombocytopenia syndrome (SFTS) caused by a recently identified bunyavirus, SFTSV, is an emerging infectious disease with extensive geographical distribution and high mortality. Progressive viral replication and severe thrombocytopenia are key features of SFTSV infection and fatal outcome, whereas the underlying mechanisms are unknown. We revealed arginine deficiency in SFTS cases by performing metabolomics analysis on two independent patient cohorts, suggesting that arginine metabolism by nitric oxide synthase and arginase is a key pathway in SFTSV infection and consequential death. Arginine deficiency was associated with decreased intraplatelet nitric oxide (Plt-NO) concentration, platelet activation, and thrombocytopenia. An expansion of arginase-expressing granulocytic myeloid-derived suppressor cells was observed, which was related to T cell CD3-ζ chain down-regulation and virus clearance disturbance, implicating a role of arginase activity and arginine depletion in the impaired anti-SFTSV T cell function. Moreover, a comprehensive measurement of arginine bioavailability, global arginine bioavailability ratio, was shown to be a good prognostic marker for fatal prediction in early infection. A randomized controlled trial demonstrated that arginine administration was correlated with enhanced Plt-NO concentration, suppressed platelet activation, and elevated CD3-ζ chain expression and eventually associated with an accelerated virus clearance and thrombocytopenia recovery. Together, our findings revealed the arginine catabolism pathway-associated regulation of platelet homeostasis and T cell dysregulation after SFTSV infection, which not only provided a functional mechanism underlying SFTS pathogenesis but also offered an alternative therapy choice for SFTS.


Asunto(s)
Arginina/deficiencia , Infecciones por Bunyaviridae/complicaciones , Infecciones por Bunyaviridae/inmunología , Terapia de Inmunosupresión , Phlebovirus/fisiología , Trombocitopenia/complicaciones , Trombocitopenia/virología , Arginina/uso terapéutico , Plaquetas/metabolismo , Infecciones por Bunyaviridae/sangre , Infecciones por Bunyaviridae/tratamiento farmacológico , Complejo CD3/metabolismo , Suplementos Dietéticos , Humanos , Inmunidad , Metaboloma , Metabolómica , Células Supresoras de Origen Mieloide/metabolismo , Óxido Nítrico/metabolismo , Linfocitos T/inmunología , Trombocitopenia/sangre , Trombocitopenia/tratamiento farmacológico
5.
Exp Dermatol ; 27(10): 1092-1097, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29928760

RESUMEN

Psoriasis is a chronic skin disease caused by immune disorder. The chronic skin inflammation involves inflammatory molecules that are released from T lymphocytes and keratinocytes. Therefore, developing an anti-inflammatory therapy that is suitable for long-term treatment is needed. Electrical stimulation induces biological responses by modulating intracellular signaling pathways. Our previous studies showed that the optimized combination treatment of mild electrical stimulation (MES, 0.1-millisecond; ms, 55-pulses per second; pps) and heat shock (HS, 42°C) modulates inflammatory symptoms of metabolic disorders and chronic kidney disease in mice models and clinical trials. Here, we investigated the effect of MES+HS treatment on imiquimod-induced psoriasis mouse model. Topical application of imiquimod cream (15 mg) to mice ear induced keratinocyte hyperproliferation and psoriasis-like inflammation. In MES+HS-treated mice, imiquimod-induced skin hyperplasia was significantly decreased. MES+HS treatment reduced the protein expression of IL-17A and the infiltration of CD3-positive cells in lesioned skin. In addition, MES+HS-treated mice had decreased mRNA expression level of antimicrobial molecules (S100A8 and Reg3γ) which aggravate psoriasis. In IL-17A-stimulated HaCaT cells, MES+HS treatment significantly lowered the mRNA expression of aggravation markers (S100A8, S100A9 and ß-defensin2). Taken together, our study suggested that MES+HS treatment improves the pathology of psoriasis via decreasing the expression of inflammatory molecules.


Asunto(s)
Terapia por Estimulación Eléctrica , Hipertermia Inducida , Psoriasis/patología , Psoriasis/terapia , Piel/patología , Animales , Complejo CD3/metabolismo , Calgranulina A/genética , Calgranulina B/genética , Línea Celular , Movimiento Celular , Proliferación Celular , Terapia Combinada , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Humanos , Hiperplasia/inducido químicamente , Hiperplasia/terapia , Imiquimod , Interleucina-17/metabolismo , Queratinocitos/fisiología , Ratones , Proteínas Asociadas a Pancreatitis/genética , Psoriasis/inducido químicamente , Psoriasis/metabolismo , ARN Mensajero/metabolismo , Linfocitos T/fisiología , beta-Defensinas/genética
6.
SLAS Technol ; 23(4): 326-337, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29281498

RESUMEN

T-cell-based immunotherapies represent a growing medical paradigm that has the potential to revolutionize contemporary cancer treatments. However, manufacturing bottlenecks related to the enrichment of therapeutically optimal T-cell subpopulations from leukopak samples impede scale-up and scale-out efforts. This is mainly attributed to the challenges that current cell purification platforms face in balancing the quantitative sorting capacity needed to isolate specific T-cell subsets with the scalability to meet manufacturing throughputs. In this work, we report a continuous-flow, quantitative cell enrichment platform based on a technique known as ratcheting cytometry that can perform complex, multicomponent purification targeting various subpopulations of magnetically labeled T cells directly from apheresis or peripheral blood mononuclear cell (PBMC) samples. The integrated ratcheting cytometry instrument and cartridge demonstrated enrichment of T cells directly from concentrated apheresis samples with a 97% purity and an 85% recovery of magnetically tagged cells. Magnetic sorting of different T-cell subpopulations was also accomplished on chip by multiplexing cell surface targets onto particles with differing magnetic strengths. We believe that ratcheting cytometry's quantitative capacity and throughput scalability represents an excellent technology candidate to alleviate cell therapy manufacturing bottlenecks.


Asunto(s)
Separación Celular/métodos , Tratamiento Basado en Trasplante de Células y Tejidos , Citometría de Flujo/métodos , Fenómenos Magnéticos , Subgrupos de Linfocitos T/citología , Automatización , Complejo CD3/metabolismo , Células HL-60 , Humanos , Células Jurkat
7.
Cell Physiol Biochem ; 43(3): 891-904, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28957810

RESUMEN

BACKGROUND/AIMS: Stem cell-based therapy is attractive in many clinical studies, but current data on the safety of stem cell applications remains inadequate. This study observed the safety, immunological effect of cynomolgus monkey umbilical cord mesenchymal stem cells (mUC-MSCs) injected into cynomolgus monkeys, in order to evaluate the safety of human umbilical cord mesenchymal stem cells (hUC-MSCs) prepared for human clinical application. METHODS: Eighteen cynomolgus monkeys were divided into three groups. Group 1 is control group, Group 2 is low-dose group, Group 3 is high-dose group. After repeated administrations of mUC-MSCs, cynomolgus monkeys were observed for possible toxic reactions. RESULTS: During the experiment, no animal died. There were no toxicological abnormalities in body weight, body temperature, electrocardiogram, coagulation and pathology. In the groups 2 and 3, AST and CK transiently increased, and serum inorganic P slightly decreased. All animals were able to recover at 28 days after the infusion was stopped. In the groups 2 and 3, CD3+ and IL-6 levels significantly increased, and recovery was after 28 days of infusion. There were no obvious pathological changes associated with the infusion of cells in the general and microscopic examinations. CONCLUSIONS: The safe dosage of repeated intravenous infusion of mUC-MSCs in cynomolgus monkeys is 1.0 × 107/kg, which is 10 times of that in clinical human use.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas/efectos adversos , Células Madre Mesenquimatosas/citología , Cordón Umbilical/citología , Adipogénesis , Animales , Aspartato Aminotransferasas/metabolismo , Recuento de Células Sanguíneas , Peso Corporal , Complejo CD3/metabolismo , Diferenciación Celular , Células Cultivadas , Creatina Quinasa/metabolismo , Femenino , Infusiones Intravenosas , Interleucina-6/metabolismo , Macaca fascicularis , Masculino , Células Madre Mesenquimatosas/metabolismo , Fósforo/sangre , Linfocitos T/citología , Linfocitos T/metabolismo , Pruebas de Toxicidad Crónica , Trasplante Homólogo
8.
Mol Imaging Biol ; 19(2): 233-244, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27572293

RESUMEN

PURPOSE: The purpose of the study is to develop a targeted nanoparticle platform for T cell labeling and tracking in vivo. PROCEDURES: Through carboxylation of the polyethylene glycol (PEG) surface of SPION, carboxylated-PEG-SPION (IOPC) was generated as a precursor for further conjugation with the targeting probe. The IOPC could readily cross-link with a variety of amide-containing molecules by exploiting the reaction between 1-ethyl-3-(3-(dimethylamino)propyl)carbodiimide and N-hydroxysuccinimide. The subsequent conjugation of monoclonal anti-CD3 antibody with IOPC made it possible to construct a magnetic resonance imaging (MRI) contrast agente (CA) that targets T cells, named IOPC-CD3. RESULTS: IOPC-CD3 was found to have high transverse relaxivity, good targeting selectivity, and good safety profile in vitro. The utility of this newly synthesized CA was explored in an in vivo rodent collagen-induced arthritis (CIA) model of rheumatoid arthritis. Serial MRI experiments revealed a selective decrease in the signal-to-noise ratio of the femoral growth plates of CIA rats infused with IOPC-CD3, with this finding being consistent with immunohistochemical results showing the accumulation of T cells and iron oxide nanoparticles in the corresponding region. CONCLUSIONS: Together with the abovementioned desirable features, these results indicate that IOPC-CD3 offers a promising prospect for a wide range of cellular and molecular MRI applications.


Asunto(s)
Artritis Reumatoide/diagnóstico por imagen , Artritis Reumatoide/inmunología , Dextranos/química , Imagen por Resonancia Magnética/métodos , Nanopartículas de Magnetita/química , Linfocitos T/inmunología , Animales , Artritis Experimental/diagnóstico por imagen , Artritis Experimental/patología , Complejo CD3/metabolismo , Forma de la Célula , Supervivencia Celular , Femenino , Citometría de Flujo , Inmunohistoquímica , Polietilenglicoles/química , Ratas Endogámicas Lew , Relación Señal-Ruido , Coloración y Etiquetado
9.
J Gastroenterol ; 52(8): 904-919, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27900483

RESUMEN

BACKGROUND: Indigo Naturalis (IN) is used as a traditional herbal medicine for ulcerative colitis (UC). However, the mechanisms of action of IN have not been clarified. We aimed to evaluate the efficacy of IN for ameliorating colonic inflammation. We further investigated the mechanisms of action of IN. METHODS: Colitis severity was assessed in dextran sodium sulfate-induced colitis and trinitrobenzene sulfonic acid-induced colitis models with or without the oral administration of IN or indigo, which is a known major component of IN. Colonic lamina propria (LP) mononuclear cells isolated from IN-treated mice were analyzed with quantitative reverse transcription polymerase chain reaction (qRT-PCR) and flow cytometry. LP and splenic mononuclear cells cultured in vitro with IN or indigo were also analyzed. The role of the candidate receptor for indigo, the aryl hydrocarbon receptor (AhR), was analyzed using Ahr-deficient mice. RESULTS: Colitis severity was significantly ameliorated in the IN and indigo treatment groups compared with the control group. The mRNA expression levels of interleukin (Il)-10 and Il-22 in the LP lymphocytes were increased by IN treatment. The treatment of splenocytes with IN or indigo increased the expression of anti-inflammatory cytokines and resulted in the expansion of IL-10-producing CD4+ T cells and IL-22-producing CD3-RORγt+ cells, but not CD4+Foxp3+ regulatory T cells. The amelioration of colitis by IN or indigo was abrogated in Ahr-deficient mice, in association with diminished regulatory cytokine production. CONCLUSIONS: IN and indigo ameliorated murine colitis through AhR signaling activation, suggesting that AhR could be a promising therapeutic target for UC.


Asunto(s)
Colitis/tratamiento farmacológico , Medicamentos Herbarios Chinos/farmacología , Carmin de Índigo/farmacología , Receptores de Hidrocarburo de Aril/efectos de los fármacos , Receptores de Hidrocarburo de Aril/metabolismo , Linfocitos T/metabolismo , Animales , Complejo CD3/metabolismo , Recuento de Linfocito CD4 , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Colitis/inducido químicamente , Colitis/patología , Sulfato de Dextran , Medicamentos Herbarios Chinos/uso terapéutico , Femenino , Factores de Transcripción Forkhead/metabolismo , Expresión Génica/efectos de los fármacos , Carmin de Índigo/uso terapéutico , Interleucina-10/genética , Interleucina-10/metabolismo , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Interleucinas/genética , Interleucinas/metabolismo , Mucosa Intestinal/citología , Leucocitos Mononucleares/metabolismo , Ratones Noqueados , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , ARN Mensajero/metabolismo , Receptores de Hidrocarburo de Aril/deficiencia , Receptores de Hidrocarburo de Aril/genética , Índice de Severidad de la Enfermedad , Bazo/citología , Linfocitos T Reguladores/metabolismo , Ácido Trinitrobencenosulfónico , Interleucina-22
10.
Mediators Inflamm ; 2016: 8781740, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27956763

RESUMEN

Object. To test whether preoperative immunonutrition is efficacious in reducing postoperative complications in patients of thymoma with myasthenia gravis (MG). Material and Methods. A total of 244 patients operated on for thymoma with myasthenia gravis were prospectively assigned to two groups, each receiving seven-day preoperative and seven-day postoperative nutrition. The patients in immunonutrition group were given oral immunonutrition (IN). The patients in control group received oral standard nutrition. Immunonutritional and inflammatory biomarkers (IgA, IgG, IgM, CD3t, CD4t, CD8t, CD4t/CD8t ratio, NK-cell, prealbumin, albumin, white blood cells counts, and C-reactive protein) and clinical variables (age, gender, BMI, performance status, type of thymoma, type of MG, operative time, pathology, operative approach, postoperative complications, quantity of drainage, hospital stays) were examined. Results. A significant reduction in the length of hospital stay, quantity of drainage, and postoperative complications was observed in the IN group (p < 0.05). An increase in the level of IgA, IgG, IgM, CD3+T, CD4+T, CD4+T/CD8+T, WBC, CRP, and NK-cell in the IN group was observed after thymectomy, while a decrease was seen with regard to prealbumin and albumin (p < 0.05). Conclusion. Preoperative immunonutrition support is effective in reducing postoperative complications in patients of thymoma with MG. It helps to lower the risk of postoperative infectious complications and hospital stays.


Asunto(s)
Suplementos Dietéticos , Miastenia Gravis/dietoterapia , Miastenia Gravis/cirugía , Timoma/dietoterapia , Timoma/cirugía , Adulto , Proteína C-Reactiva/metabolismo , Complejo CD3/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Femenino , Humanos , Inmunoglobulina A/metabolismo , Inmunoglobulina G/metabolismo , Inmunoglobulina M/metabolismo , Células Asesinas Naturales/metabolismo , Tiempo de Internación , Masculino , Persona de Mediana Edad , Complicaciones Posoperatorias , Periodo Posoperatorio , Cuidados Preoperatorios , Estudios Prospectivos , Timectomía , Resultado del Tratamiento
11.
Leuk Lymphoma ; 57(5): 1021-32, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27050240

RESUMEN

Blinatumomab is a member of a novel class of T cell-engaging bispecific antibodies, so-called Bispecific T cell Engager (BiTEs). It is directed against the B cell differentiation antigen CD19 and intended for treatment of B cell malignancies. In clinical phase I/II trials, blinatumomab showed remarkable single-agent activity in patients with relapsed and/or refractory (R/R) non-Hodgkin lymphoma and R/R B cell precursor acute lymphoblastic leukemia (B-precursor ALL). Cytokine release syndrome and neurological side effects were dose-limiting. Adverse effects were well manageable and transient in nature. Based on results of an international phase II trial, blinatumomab received FDA approval for the treatment of R/R B-precursor ALL in December 2014. Ongoing and future trials will contribute to further optimization of blinatumomab-based T cell therapy and have to show that integration of blinatumomab in current and innovative treatment protocols improves overall survival and quality of life of patients with B cell malignancies.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Animales , Anticuerpos Biespecíficos/economía , Anticuerpos Biespecíficos/farmacología , Antígenos CD19/metabolismo , Antineoplásicos/economía , Antineoplásicos/farmacología , Complejo CD3/metabolismo , Ensayos Clínicos como Asunto , Análisis Costo-Beneficio , Evaluación Preclínica de Medicamentos , Resistencia a Antineoplásicos , Humanos , Linfoma no Hodgkin/tratamiento farmacológico , Linfoma no Hodgkin/inmunología , Linfoma no Hodgkin/metabolismo , Linfoma no Hodgkin/patología , Terapia Molecular Dirigida , Neoplasias/inmunología , Neoplasias/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Recurrencia , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Resultado del Tratamiento
12.
J Neurosci ; 36(4): 1410-5, 2016 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-26818526

RESUMEN

Brain-intrinsic degenerative cascades have been proposed to be an initial factor driving lesion formation in multiple sclerosis (MS). Here, we identify neurodegeneration as a potent trigger for peripheral immune cell recruitment into the mouse forebrain. Female C57BL/6 mice were fed cuprizone for 3 weeks, followed by a period of 2 weeks on normal chow to induce the formation of lesion foci in the forebrain. Subsequent immunization with myelin oligodendrocyte glycoprotein 35-55 peptide, which induces myelin autoreactive T cells in the periphery, resulted in massive immune cell recruitment into the affected forebrain. Additional adoptive transfer experiments together with flow cytometry analysis underline the importance of brain-derived signals for immune cell recruitment. This study clearly illustrates the significance of brain-intrinsic degenerative cascades for immune cell recruitment and MS lesion formation. Additional studies have to address the signaling cascades and mechanistic processes that form the top-down communication between the affected brain area, neurovascular unit, and peripheral immune cells. SIGNIFICANCE STATEMENT: We identify neurodegeneration as a potent trigger for peripheral immune cell recruitment into the forebrain. Thus, immune cell recruitment might be a second step during the formation of new inflammatory lesions in multiple sclerosis. A better understanding of factors regulating neurodegeneration-induced immune cell recruitment will pave the way for the development of novel therapeutic treatment strategies.


Asunto(s)
Linfocitos/fisiología , Monocitos/fisiología , Enfermedades Neurodegenerativas/patología , Prosencéfalo/patología , Traslado Adoptivo , Animales , Complejo CD3/metabolismo , Proteínas de Unión al Calcio/metabolismo , Quelantes/toxicidad , Cuprizona/toxicidad , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/patología , Femenino , Adyuvante de Freund/toxicidad , Ganglios Linfáticos/patología , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/metabolismo , Glicoproteína Mielina-Oligodendrócito/inmunología , Enfermedades Neurodegenerativas/inducido químicamente , Fragmentos de Péptidos/inmunología , Toxina del Pertussis/toxicidad
13.
PLoS One ; 11(1): e0146869, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26752299

RESUMEN

BACKGROUND: Acute myelogenous leukemia (AML) is a cancer of the blood that most commonly affects human adults. The specific cause of AML is unclear, but it induces abnormality of white blood cells that grow rapidly and accumulate in bone marrow interfering with the production and functions of the normal blood cells. AML patients face poor prognosis and low quality of life during chemotherapy or transplantation of hematopoietic stem cells due to the progressive impairment of their immune system. The goal of this study is to find natural products that have the potential to delay growth or eliminate the abnormal leukemic cells but cause less harmful effect to the body's immune system. METHODS AND FINDINGS: The unsaponified fraction of Riceberry rice bran (RBDS) and the main pure compound, gramisterol, were studied for cytotoxicity and biological activities in WEHI-3 cells and in the leukemic mouse model induced by transplantation of WEHI-3 cells intraperitoneally. In the in vitro assay, RBDS and gramisterol exerted sub-G1 phase cell cycle arrest with a potent induction of apoptosis. Both of them effectively decreased cell cycle controlling proteins (cyclin D1 and cyclin E), suppressed cellular DNA synthesis and mitotic division, and reduced anti-apoptosis Bcl-2 protein, but increased apoptotic proteins (p53 and Bax) and activated caspase-3 enzyme in the intrinsic cell death stimulation pathway. In leukemic mice, daily feeding of RBDS significantly increased the amount of immune function-related cells including CD3+, CD19+, and CD11b+, and elevated the serum levels of IFN-γ, TNF-α, IL-2, and IL-12ß cytokines, but suppressed IL-10 level. At the tumor sites, CD11b+ cells were polarized and became active phagocytotic cells. Treatment of mice normal immune cells with gramisterol alone or a combination of gramisterol with cytokines released from RBDS-treated leukemic mice splenocytes culture synergistically increased pSTAT1 transcriptional factor that up-regulated the genes controlling cell survival and function. Phosphorylation of STAT1 was absent in WEHI-3. Instead, similar treatments significantly decreased pSTAT3 signaling that regulates transcription of genes controlling tumor growth and proliferation. CONCLUSIONS: Rice bran gramisterol possesses a promising anti-cancer effect against a tumor of white blood cells and induces the production of anti-cancer immune-related cytokines. Gramisterol induces cell cycle arrest and apoptosis via suppression of pSTAT3 signaling control of tumor cells' growth and progression. Gramisterol increased IFN-γ production and prevented the dysfunctional immune system of leukemic mice by enhancing pSTAT1 transcription signal controlling proliferation and functions of hematopoietic cells in the spleen. Together with IFN-γ, gramisterol efficiently facilitates leukemic mice immune system modulation leading to improvement of the AML condition. Administration of RBDS containing gramisterol potentiates immune recovery of leukemic mice and extends their survival. This finding encourages the medicinal application of rice bran gramisterol as a palliative treatment or an alternative agent for future drug development against AML.


Asunto(s)
Antineoplásicos/uso terapéutico , Colestadienoles/uso terapéutico , Regulación Leucémica de la Expresión Génica , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/inmunología , Oryza/química , Esteroles/uso terapéutico , Animales , Antígenos CD19/metabolismo , Antineoplásicos/química , Apoptosis , Antígeno CD11b/metabolismo , Complejo CD3/metabolismo , Caspasa 3/metabolismo , Proliferación Celular , Colestadienoles/química , Ciclina D1/metabolismo , Ciclina E/metabolismo , Modelos Animales de Enfermedad , Ensayos de Selección de Medicamentos Antitumorales , Fase G1/efectos de los fármacos , Sistema Inmunológico , Ratones , Ratones Endogámicos BALB C , Extractos Vegetales/química , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal , Esteroles/química , Proteína p53 Supresora de Tumor/metabolismo , Proteína X Asociada a bcl-2/metabolismo
14.
Neuroimmunomodulation ; 23(1): 41-57, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26468875

RESUMEN

OBJECTIVES: We investigated whether Ginkgo biloba extract (EGb761) can provide neuroprotective effects and enhance the efficacy of bone marrow-derived mesenchymal stem cells (BMSCs) in a rat model of experimental autoimmune encephalomyelitis (EAE). METHODS: We examined the synergistic action of BMSCs combined with EGb761 treatment in EAE rats. The immunized rats received an intravenous injection of BMSCs or intraperitoneal administration of EGb761 or both on the day of the onset of clinical symptoms and for the following 21 days. Clinical severity scores were recorded daily and histopathological examination of the spinal cord and cytokine concentrations in the serum were studied on days 14 and 31 postimmunization. RESULTS: Our results showed that combined treatment with BMSCs and EGb761 further decreased the disease severity, maximal clinical score and number of infiltrated mononuclear cells, especially CD3-positive T cells. We observed that the demyelination score and the density of axonal loss in the spinal cord were significantly reduced in mice receiving the combination therapy. The serum concentrations of the phosphorylated neurofilament heavy chain, tumor necrosis factor-α and interferon-γ were reduced in the combination-treatment group. CONCLUSION: Our results suggest that combined treatment with BMSCs and EGb761 have a synergistic effect in rats with EAE by inhibiting the secretion of proinflammatory cytokines, demyelination and protecting axons and neurons.


Asunto(s)
Encefalomielitis Autoinmune Experimental/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/fisiología , Fármacos Neuroprotectores/uso terapéutico , Extractos Vegetales/uso terapéutico , Animales , Complejo CD3/metabolismo , Movimiento Celular/efectos de los fármacos , Citocinas/antagonistas & inhibidores , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/patología , Femenino , Ginkgo biloba , Células Madre Mesenquimatosas/efectos de los fármacos , Ratas , Ratas Wistar , Índice de Severidad de la Enfermedad , Tinción con Nitrato de Plata , Médula Espinal/efectos de los fármacos , Médula Espinal/patología , Médula Espinal/ultraestructura , Estadísticas no Paramétricas
15.
Eur J Cancer ; 51(17): 2708-17, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26342674

RESUMEN

AIM: To investigate whether the immune response in colorectal liver metastases is related to progression free survival (PFS) and if this may be influenced by systemic therapy. METHODS: A retrospective central collection of tumour tissue was organised for the European Organisation for Research and Treatment of Cancer (EORTC) study 40983, where patients with colorectal liver metastases were treated by either resection alone or resection with perioperative FOLFOX. Immunostaining on whole slides was performed to recognise T-lymphocytes (CD3+, CD4+, CD8+), B-lymphocytes (CD20+), macrophages (CD68+) and mast cells (CD117+) inside the tumour, at the tumour border (TNI) and in normal liver tissue surrounding the tumour (0.5-2mm from the TNI). Immunological response was compared between treatment arms and correlated to PFS. RESULTS: Tumour tissue and immune response profiles were available for 82 resected patients, 38 in the perioperative chemotherapy arm and 44 in the surgery alone arm. Baseline patient and disease characteristics were similar between the treatment arms. In response to chemotherapy, we observed increased CD3+ lymphocyte and mast cell counts inside the tumour (p<0.01), lower CD4+ lymphocytes in the normal liver tissue (p=0.02) and lower macrophage counts in normal tissue (p<0.01) and at the TNI (p=0.02). High number of CD3+ lymphocyte and mast cells, and high T-cell score were correlated with tumour regression grade (TRG). Prolonged PFS correlated with the presence of mast cells in the tumour (9.8 versus 16.5 months, Hazard ratio (HR) 0.54 p=0.03), higher CD3+ lymphocyte count at the TNI (10.8 versus 22.8 months, HR 0.57, p=0.03) and T-cell score >2 (10.8 versus 38.6 months, HR 0.51, p=0.04). CONCLUSION: Our analyses in the context of a randomised study suggest that chemotherapy influences immune cell profiles, independent of patient characteristics. Immune responses of lymphocytes and mast cells were associated with pathological response to chemotherapy and to increased PFS. High CD3+ lymphocytes at the tumour front and intratumoural mast cells appear to be prognostic for patients with colorectal liver metastases.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/cirugía , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/cirugía , Adulto , Anciano , Antígenos CD/metabolismo , Antígenos CD20/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Complejo CD3/metabolismo , Quimioterapia Adyuvante/métodos , Neoplasias Colorrectales/patología , Terapia Combinada , Supervivencia sin Enfermedad , Femenino , Fluorouracilo/administración & dosificación , Humanos , Sistema Inmunológico/efectos de los fármacos , Sistema Inmunológico/metabolismo , Sistema Inmunológico/patología , Leucovorina/administración & dosificación , Neoplasias Hepáticas/secundario , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Persona de Mediana Edad , Compuestos Organoplatinos/administración & dosificación , Oxaliplatino , Pronóstico , Estudios Retrospectivos , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Resultado del Tratamiento
16.
J Immunol Methods ; 424: 14-9, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25956037

RESUMEN

Activation of the inducible costimulator (ICOS) signaling pathway in T cells is difficult to assess with bioassays, because most T cell lines do not constitutively express ICOS. Additionally, engagement of ICOS by its natural ligand B7 related protein 1 (B7RP1) is insufficient to elicit ICOS signaling, but requires simultaneous costimulation of the T cell receptor (TCR) to be effective. Here we describe a genetically engineered human T cell line that expresses a chimeric receptor (ICOS-CD3) consisting of full-length human ICOS fused at its C-terminal end to the cytoplasmic domain of human CD3 zeta. When engaged by B7RP1, ICOS-CD3 initiated signaling independently of TCR costimulation and induced substantially more IL-2 secretion in Jurkat T cells compared to wildtype ICOS. We demonstrate that this signaling-enhanced chimeric receptor can be used in simple and sensitive bioassays to detect bioactive B7RP1, anti-B7RP1 drugs, and the presence of corresponding neutralizing anti-drug antibodies.


Asunto(s)
Proteína Coestimuladora de Linfocitos T Inducibles/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Linfocitos T/metabolismo , Bioensayo/métodos , Complejo CD3/química , Complejo CD3/genética , Complejo CD3/metabolismo , Línea Celular , Membrana Celular/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Expresión Génica , Humanos , Ligando Coestimulador de Linfocitos T Inducibles/antagonistas & inhibidores , Ligando Coestimulador de Linfocitos T Inducibles/metabolismo , Proteína Coestimuladora de Linfocitos T Inducibles/química , Proteína Coestimuladora de Linfocitos T Inducibles/genética , Interleucina-2/biosíntesis , Unión Proteica , Dominios y Motivos de Interacción de Proteínas/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas Recombinantes de Fusión/genética , Transducción de Señal/efectos de los fármacos , Linfocitos T/inmunología
17.
Chin J Integr Med ; 21(5): 339-45, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25776840

RESUMEN

OBJECTIVE: To investigate the therapeutic effects of Qingre Quyu Granule (QQG) on the patients with severe carotid stenosis, and to explore the mechanism of it. METHODS: Ninety-six patients with severe carotid stenosis were enrolled in the study and were classified into a QQG group (n=48) and a control group (n=48) randomly using consecutively numbered envelopes. The patients in the QQG group were given QQG and Western medicine, those in the control group were given Western medicine merely, the course of treatment was 16 weeks. All patients went through endarterectomy after treatment. Plaques were subjected to the analysis of CD3, CD68, soluble intercellular adhesion molecule 1 (ICAM-1), matrix metalloprotease-9 (MMP-9), CD40L, tenascin-C, and collagen content lipid content by immunohistochemistry or polarized light analysis. RESULTS: By the end of experiment, the expressions of CD3, CD68, ICAM-1, MMP9, CD40L and tenascin-C on the plaques were statistically significant lower in the QQG group compared with the control group(P<0.01). The lipid content of the plaque was also significantly lower in the QQG group compared with the control group (P<0.01). The interstitial collagen in the tissue sections of the plaques was also significantly higher in the QQG group in comparison with the control group (P<0.01). CONCLUSION: QQG could stabilize carotid artery plaques through inhibiting pro-inflammation factors and restraining the tenascin-C and MMP9 pathway.


Asunto(s)
Estenosis Carotídea/complicaciones , Estenosis Carotídea/tratamiento farmacológico , Medicamentos Herbarios Chinos/uso terapéutico , Placa Aterosclerótica/complicaciones , Placa Aterosclerótica/tratamiento farmacológico , Tenascina/metabolismo , Anciano , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Complejo CD3/metabolismo , Ligando de CD40/metabolismo , Arterias Carótidas/metabolismo , Arterias Carótidas/patología , Estenosis Carotídea/sangre , Colágeno/metabolismo , Medicamentos Herbarios Chinos/farmacología , Femenino , Humanos , Inmunohistoquímica , Inflamación/complicaciones , Inflamación/patología , Molécula 1 de Adhesión Intercelular/metabolismo , Lípidos/sangre , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Placa Aterosclerótica/sangre
18.
Mol Immunol ; 67(2 Pt A): 95-106, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25637431

RESUMEN

Bispecific antibodies are on the cusp of coming of age as therapeutics more than half a century after they were first described. Two bispecific antibodies, catumaxomab (Removab(®), anti-EpCAM×anti-CD3) and blinatumomab (Blincyto(®), anti-CD19×anti-CD3) are approved for therapy, and >30 additional bispecific antibodies are currently in clinical development. Many of these investigational bispecific antibody drugs are designed to retarget T cells to kill tumor cells, whereas most others are intended to interact with two different disease mediators such as cell surface receptors, soluble ligands and other proteins. The modular architecture of antibodies has been exploited to create more than 60 different bispecific antibody formats. These formats vary in many ways including their molecular weight, number of antigen-binding sites, spatial relationship between different binding sites, valency for each antigen, ability to support secondary immune functions and pharmacokinetic half-life. These diverse formats provide great opportunity to tailor the design of bispecific antibodies to match the proposed mechanisms of action and the intended clinical application.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Inmunoterapia/métodos , Neoplasias/terapia , Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/metabolismo , Antígenos CD19/inmunología , Antígenos CD19/metabolismo , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Antineoplásicos/inmunología , Antineoplásicos/metabolismo , Antineoplásicos/uso terapéutico , Complejo CD3/inmunología , Complejo CD3/metabolismo , Moléculas de Adhesión Celular/inmunología , Moléculas de Adhesión Celular/metabolismo , Molécula de Adhesión Celular Epitelial , Humanos , Modelos Inmunológicos , Neoplasias/inmunología , Neoplasias/metabolismo
19.
Eur J Pharmacol ; 742: 102-12, 2014 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-25218987

RESUMEN

Multiple sclerosis (MS) is an autoimmune inflammatory demyelinating disease of brain and spinal cord that has an increasing incidence worldwide and classically presents in a relapsing-remitting form. This study was designed to induce a relapsing-remitting model of experimental autoimmune encephalomyelitis (EAE) to investigate the possible modulatory effect of celastrol on Th1/Th2 cytokines profile, immunohistochemical expression of TLR2, and CD3+T-lymphocytic count. Eighteen female Sprague Dawley rats were divided into 3 groups; where group I served as normal control, group II as EAE+vehicle, and group III as EAE treated by celastrol (1mg/kg/day, i.p.) started at 10th day till 42nd day post-immunization. The clinical score of rats in group II (EAE+vehicle) was relapsed after the re-challenge at the 35th day post-immunization and exhibited significant positive association with serum TNF-α, NF-κB expression and nitrites levels in brain and spinal cord, and CD3+ T-lymphocytic count in brain tissues while serum IL-10 showed significant negative association. Treatment of EAE by celastrol caused amelioration of the clinical score and inhibited the relapse. It caused significant shift in cytokines profile from Th1 by decrease in TNF-α towards Th2 pattern by increase in IL-10. Moreover, celastrol treatment resulted in significant reduction in NF-κB expression, nitrites levels, as well as immunohistochemical expression of TLR2 and CD3+ T-lymphocytic count. The beneficial effect of celastrol was further confirmed histopathologically by reduction in H&E score. Collectively, these results provide a promising pre-clinical evidence and conclusion about use of celastrol in treatment of multiple sclerosis that must be accessed in further clinical studies.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/inmunología , Factores Inmunológicos/farmacología , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Esclerosis Múltiple Recurrente-Remitente/inmunología , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Triterpenos/farmacología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Encéfalo/patología , Complejo CD3/metabolismo , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Femenino , Interleucina-10/sangre , Recuento de Linfocitos , Esclerosis Múltiple Recurrente-Remitente/patología , FN-kappa B/metabolismo , Nitritos/metabolismo , Triterpenos Pentacíclicos , Fitoterapia , Plantas Medicinales , Ratas , Ratas Sprague-Dawley , Médula Espinal/efectos de los fármacos , Médula Espinal/inmunología , Médula Espinal/patología , Células TH1/efectos de los fármacos , Células TH1/inmunología , Células Th2/efectos de los fármacos , Células Th2/inmunología , Receptor Toll-Like 2/metabolismo , Tripterygium , Factor de Necrosis Tumoral alfa/sangre
20.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 30(7): 748-53, 758, 2014 Jul.
Artículo en Chino | MEDLINE | ID: mdl-25001943

RESUMEN

OBJECTIVE: To investigate the effects of the autologous cytokine-induced killer (CIK) cell infusion on the subpopulation distribution and activity of the CIK cells from the patients with malignant tumors when prepared in the same liquid culture system again. METHODS: A total of 201 patients who gave a written consent and received 2 courses of therapeutic infusion of CIK cells were divided into ≤ 90-day group in which the secondary preparation of CIK cells was performed in less than 90 days after the primary infusion and >90-day group in which the secondary preparation of CIK cells was performed in more than 90 days after the primary infusion. The proliferation and subtypes, including CD3⁺ cells, CD3⁺ CD4⁺ cells, CD3⁺ CD8⁺ cells, and CD3⁺ CD56⁺ cells, of CIK cells were analyzed by hemocytometer with trypan blue exclusion and flow cytometry, respectively. The expression of NKG2D receptor was also detected using flow cytometry. The cytotoxicity against K562 cells was analyzed using lactate dehydrogenase (LDH) release. RESULTS: The percentage of CD3⁺ CD56⁺ cell subpopulation in the secondary preparation of CIK cells in ≤ 90-day group [(16.7 ± 9.1)%] was higher than that in the primary preparation of CIK cells [(13.5 ± 8.6)%] (P<0.01). Furthermore, the percentage of NKG2D in the secondary CIK cell preparation [(84.1 ± 10.8)%] was significantly higher than that in the primary CIK cell preparation [(81.1 ± 14.8)%] in ≤ 90-day group (P<0.05). In contrast, the percentage of CD3⁺ CD4⁺ cells in the secondary CIK cell preparation [(15.2 ± 9.7)%] was significantly lower than that in the primary CIK cell preparation [(17.6 ± 12.5)%] (P<0.01). However, no significant differences in CD3⁺ CD56⁺ cell subpopulation and expression of NKG2D was detected between the primary and secondary CIK cell preparation in >90 d group, although the percentage of CD3⁺ CD4⁺ cells in the secondary CIK cell preparation [(14.5 ± 9.4)%] was significantly lower than that in the primary CIK cell preparation [(18.2 ± 12.9)%] (P<0.01). In addition, no significant differences in total cell number and cytotoxic activity against K562 cells between the primary and secondary CIK cell preparation was detected either in >90-day group or in ≤ 90-day group. CONCLUSION: CIK cell infusion can facilitate and enhance the proliferation and differentiation of the precursor cells of the CD3⁺ CD56⁺ subpopulation in the same CIK cell culture system and this effect does not last more than 90 days, suggesting that the secondary CIK cell infusion should be performed within 90 days in order to obtain the better therapeutic efficacy.


Asunto(s)
Transfusión de Sangre Autóloga/métodos , Complejo CD3/inmunología , Antígeno CD56/inmunología , Células Asesinas Inducidas por Citocinas/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Adulto , Anciano , Anciano de 80 o más Años , Complejo CD3/metabolismo , Antígeno CD56/metabolismo , Diferenciación Celular/inmunología , Proliferación Celular , Células Asesinas Inducidas por Citocinas/metabolismo , Citotoxicidad Inmunológica/inmunología , Femenino , Citometría de Flujo , Humanos , Células K562 , Masculino , Persona de Mediana Edad , Subfamilia K de Receptores Similares a Lectina de Células NK/inmunología , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Neoplasias/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Factores de Tiempo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA