Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Crit Rev Immunol ; 40(3): 237-246, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33389887

RESUMEN

The autoimmune disease multiple sclerosis (MS) is driven by T cells that are reactive to self-antigens of the brain and spinal cord. Many drugs have been developed to treat MS, but we believe that immune-specific targeting of pathogenic T cells may be a better approach for treatment. This type of therapy identifies specific components of the self-reactive T-cell repertoire that would undergo similar natural selection criteria as those found in driver genes in cancer genesis. In the context of autoimmunity, we propose that a focused subpopulation of T cells "drive" disease and could be found in higher frequency and become over-represented during disease induction and subsequent MS relapses. In addition, identification of other key signatures of driver T cells is important. One such marker could be interleukin (IL)-17- producing T cells. Here, we discuss the use of experimental autoimmune encephalomyelitis (EAE) animal models (that mimic many pathologic mechanisms involved in MS) to identify possible driver clones of this autoimmunity within the set of T cells expressing the IL-17 cytokine. EAE can be induced by myelin injection-associated proteins in adjuvants. The disease model in the Swiss/Jackson laboratory mouse strain represents the most common form of MS in humans: relapsing remitting MS. Finally, we discuss the concept of using IL-17 as a marker for pathogenic T cells, combined with identifying their T-cell receptor V repertoire, which could provide targeted approaches designed to neutralize driver T cells for MS immunotherapy.


Asunto(s)
Células Clonales/inmunología , Encefalomielitis Autoinmune Experimental/terapia , Inmunoterapia Adoptiva/métodos , Esclerosis Múltiple Recurrente-Remitente/terapia , Células Th17/inmunología , Animales , Autoantígenos/inmunología , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Interleucina-17/genética , Interleucina-17/metabolismo , Ratones , Esclerosis Múltiple Recurrente-Remitente/genética , Esclerosis Múltiple Recurrente-Remitente/inmunología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/inmunología , Células Th17/metabolismo
2.
Jpn J Clin Oncol ; 48(7): 619-624, 2018 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-29529220

RESUMEN

BACKGROUND: The poor clinical prognosis of Stage IIIB colon cancer patients is due in part to the current lack of an effective diagnostic method being available and highlights a need for the identification of novel biomarkers like microRNA (miRNA). PATIENTS AND METHODS: We used microarray analysis to compare the miRNA expression profiles of eight Stage IIIB colon cancer patients with worse clinical outcome (those who developed liver metastases between 8 and 18 months after surgery) against eight 'cured' Stage IIIB colon cancer patients (those who remained disease free following surgery during the same monitoring period). In addition, quantitative real-time polymerase chain reaction (qRT-PCR) analysis was performed examining miRNAs in tumor tissue of 98 patients with Stage IIIB colon cancer. RESULTS: We found, miRNA-192-3p and miRNA-192-5p were down regulated in the patients with worsening disease compared to the control 'cured' Stage IIIB colon cancer patients (P = 0.026 and P = 0.042, respectively). Patients with higher expression of miRNA-192-5p had higher 5-year disease-free survival (DFS) (84.21%) and overall survival (OS) (89.47%) than those with lower targeted miRNA expression DFS (38.8%; hazard ratio (HR): 3.74, 95% confidence interval (CI): 1.52-9.23, P = 0.042) and OS (48.57%; HR: 5.01, 95% CI: 1.75-14.38, P = 0.033). In contrast, patients with higher expression of miRNA-192-3p did not appear to statistically impact the survival of patients in this setting (DFS 73.33% vs 64.7%, HR: 0.68, 95% CI: 0.31-1.52, P = 0.35; OS 76.67% vs 66.17%, HR: 0.62, 95% CI: 0.27-1.45, P = 0.27). CONCLUSIONS: The decreased expression of miRNA-192-5p found for patients with relapsing disease might represent a highly predictive marker to use for the prognosis of Stage IIIB colon cancer patients.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias del Colon/genética , Neoplasias del Colon/patología , MicroARNs/genética , Adulto , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , MicroARNs/metabolismo , Persona de Mediana Edad , Análisis Multivariante , Estadificación de Neoplasias , Modelos de Riesgos Proporcionales , Análisis de Supervivencia
3.
Analyst ; 141(7): 2284-95, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-26958659

RESUMEN

Cancer therapy via redirected lysis mediated by antibodies and antibody-derived agents relies on the availability of substantial numbers of sufficiently active immune effector cells. To monitor antitumor responses before and during therapy, sensitive methods are needed, capable of quantitating specific lysis of target cells. Here we present a chip-based single-cell cytometric assay, which uses adherent human target cells arrayed in structured micro-fields. Using a fluorescent indicator of cell death and time-lapse microscopy in an automated high-throughput mode, we measured specific target cell lysis by activated human NK cells, mediated by the therapeutic single chain triplebody SPM-2 (33-16-123). This antibody-derived tri-specific fusion protein carries binding sites for the myeloid antigens CD33 and CD123 and recruits NK cells via a binding site for the Fc-receptor CD16. Specific lysis increased with increasing triplebody concentration, and the single-cell assay was validated by direct comparison with a standard calcein-release assay. The chip-based approach allowed measurement of lysis events over 16 hours (compared to 4 hours for the calcein assay) and required far smaller numbers of primary cells. In addition, dynamic properties inaccessible to conventional methods provide new details about the activation of cytolytic effector cells by antibody-derived agents. Thus, the killing rate exhibited a dose-dependent maximum during the reaction interval. In clinical applications ex vivo monitoring of NK activity of patient's endogenous cells will likely help to choose appropriate therapy, to detect impaired or recovered NK function, and possibly to identify rare subsets of cancer cells with particular sensitivity to effector-cell mediated lysis.


Asunto(s)
Células Asesinas Naturales/citología , Procedimientos Analíticos en Microchip/métodos , Análisis de la Célula Individual/métodos , Anticuerpos de Cadena Única/metabolismo , Muerte Celular , Línea Celular Tumoral , Células HEK293 , Humanos , Factores de Tiempo
4.
J Transl Med ; 11: 289, 2013 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-24237598

RESUMEN

BACKGROUND: The capacity of patient's Natural Killer cells (NKs) to be activated for cytolysis is an important prerequisite for the success of antibody-derived agents such as single-chain triplebodies (triplebodies) in cancer therapy. NKs recovered from AML patients at diagnosis are often found to be reduced in peripheral blood titers and cytolytic activity. Here, we had the unique opportunity to compare blood titers and cytolytic function of NKs from an AML patient with those of a healthy monozygotic twin. The sibling's NKs were compared with the patient's drawn either at diagnosis or in remission after chemotherapy. The cytolytic activities of NKs from these different sources for the patient's autologous AML blasts and other leukemic target cells in conjunction with triplebody SPM-2, targeting the surface antigens CD33 and CD123 on the AML cells, were compared. METHODS: Patient NKs drawn at diagnosis were compared to NKs drawn in remission after chemotherapy and a sibling's NKs, all prepared from PBMCs by immunomagnetic beads (MACS). Redirected lysis (RDL) assays using SPM-2 and antibody-dependent cellular cytotoxicity (ADCC) assays using the therapeutic antibody RituximabTM were performed with the enriched NKs. In addition, MACS-sorted NKs were analyzed for NK cell activating receptors (NCRs) by flow cytometry, and the release of TNF-alpha and IFN-gamma from blood samples of both siblings after the addition of the triplebody were measured in ELISA-assays. RESULTS: Patient NKs isolated from peripheral blood drawn in remission produced comparable lysis as NKs from the healthy twin against the patient's autologous bone marrow (BM) blasts, mediated by SPM-2. The NCR receptor expression profiles on NKs from patient and twin were similar, but NK cell titers in peripheral blood were lower for samples drawn at diagnosis than in remission. CONCLUSIONS: Peripheral blood NK titers and ex vivo cytolytic activities mediated by triplebody SPM-2 were comparable for cells drawn from an AML patient in remission and a healthy twin. If these results can be generalized, then NKs from AML patients in remission are sufficient in numbers and cytolytic activity to make triplebodies promising new agents for the treatment of AML.


Asunto(s)
Citotoxicidad Inmunológica , Células Asesinas Naturales/inmunología , Leucemia Mieloide Aguda/inmunología , Inducción de Remisión , Gemelos Monocigóticos , Adulto , Citotoxicidad Celular Dependiente de Anticuerpos , Femenino , Citometría de Flujo , Humanos , Adulto Joven
5.
J Transl Med ; 9: 101, 2011 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-21722394

RESUMEN

BACKGROUND: Non Obese Diabetic mice lacking B cells (NOD.Igµ(null) mice) do not develop diabetes despite their susceptible background. Upon reconstitution of B cells using a chimera approach, animals start developing diabetes at 20 weeks of age. METHODS: We have used the spectratyping technique to follow the T cell receptor (TCR) V beta repertoire of NOD.Igµ(null) mice following B cell reconstitution. This technique provides an unbiased approach to understand the kinetics of TCR expansion. We have also analyzed the TCR repertoire of reconstituted animals receiving cyclophosphamide treatment and following tissue transplants to identify common aggressive clonotypes. RESULTS: We found that B cell reconstitution of NOD.Igµ(null) mice induces a polyclonal TCR repertoire in the pancreas 10 weeks later, gradually diversifying to encompass most BV families. Interestingly, these clonotypic BV expansions are mainly confined to the pancreas and are absent from pancreatic lymph nodes or spleens. Cyclophosphamide-induced diabetes at 10 weeks post-B cell reconstitution reorganized the predominant TCR repertoires by removing potential regulatory clonotypes (BV1, BV8 and BV11) and increasing the frequency of others (BV4, BV5S2, BV9, BV16-20). These same clonotypes are more frequently present in neonatal pancreatic transplants under the kidney capsule of B-cell reconstituted diabetic NOD.Igµ(null) mice, suggesting their higher invasiveness. Phenotypic analysis of the pancreas-infiltrating lymphocytes during diabetes onset in B cell reconstituted animals show a predominance of CD19+ B cells with a B:T lymphocyte ratio of 4:1. In contrast, in other lymphoid organs (pancreatic lymph nodes and spleens) analyzed by FACS, the B:T ratio was 1:1. Lymphocytes infiltrating the pancreas secrete large amounts of IL-6 and are of Th1 phenotype after CD3-CD28 stimulation in vitro. CONCLUSIONS: Diabetes in NOD.Igµ(null) mice appears to be caused by a polyclonal repertoire of T cell accumulation in pancreas without much lymphoid organ involvement and is dependent on the help by B cells.


Asunto(s)
Linfocitos B/inmunología , Diabetes Mellitus Experimental/inmunología , Cadenas mu de Inmunoglobulina/inmunología , Inmunofenotipificación/métodos , Islotes Pancreáticos/inmunología , Linfocitos T/inmunología , Animales , Animales Recién Nacidos , Linfocitos B/citología , Proliferación Celular , Células Clonales , Ciclofosfamida , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Rechazo de Injerto/complicaciones , Rechazo de Injerto/inmunología , Rechazo de Injerto/patología , Memoria Inmunológica/inmunología , Islotes Pancreáticos/patología , Trasplante de Islotes Pancreáticos , Ratones , Ratones Endogámicos NOD , Fenotipo , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Bazo/patología , Linfocitos T/citología
6.
J Transl Med ; 8: 98, 2010 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-20946663

RESUMEN

BACKGROUND: Tumor immune responses are first generated and metastases often begin in tumor sentinel lymph nodes (TSLN). Therefore, it is important to promote tumor immunity within this microenvironment. Mifepristone (RU486) treatment can interfere with cortisol signaling that can lead to suppression of tumor immunity. Here, we assessed whether treatment with RU486 in conjunction with an intratumor injection of Ad5IL-12 vector (a recombinant adenovirus expressing IL-12) could impact the TSLN microenvironment and prostate cancer progression. METHODS: The human PC3, LNCaP or murine TRAMP-C1 prostate cancer cell lines were used to generate subcutaneous tumors in NOD.scid and C57BL/6 mice, respectively. Adjuvant effects of RU486 were looked for in combination therapy with intratumor injections (IT) of Ad5IL-12 vector in comparison to PBS, DL70-3 vector, DL70-3 + RU486, RU486 and Ad5IL-12 vector treatment controls. Changes in tumor growth, cell cytotoxic activity and populations of CD4+/FoxP3+ T regulatory cells (Treg) in the TSLN were evaluated. RESULTS: Treatment of human PC3 prostate xenograft or TRAMP-C1 tumors with combination Ad5IL-12 vector and RU486 produced significantly better therapeutic efficacy in comparison to controls. In addition, we found that combination therapy increased the capacity of TSLN lymphocytes to produce Granzyme B in response to tumor cell targets. Finally, combination therapy tended towards decreases of CD4+/FoxP3+ T regulatory cell populations to be found in the TSLN. CONCLUSION: Inclusion of RU486 may serve as a useful adjuvant when combined with proinflammatory tumor killing agents by enhancement of the immune response and alteration of the TSLN microenvironment.


Asunto(s)
Adenoviridae/genética , Vectores Genéticos , Interleucina-12/administración & dosificación , Metástasis Linfática , Mifepristona/uso terapéutico , Neoplasias de la Próstata/terapia , Animales , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Interleucina-12/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología
7.
Cancer Res ; 67(5): 2290-7, 2007 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-17332360

RESUMEN

Tumor cells can evolve to evade immune responses by down-modulating surface MHC class I expression and become refractory to T cell-directed immunotherapy. We employed a strategy to bypass this escape mechanism using a recombinant adenovirus vector expressing interleukin-12 (Ad5IL-12) to target natural killer (NK) cell-mediated killing of human prostate tumors in NOD.scid mice. Fluorescence-activated cell sorting analysis revealed that LNCaP tumor cells bear negligible levels of MHC class I molecules; yet, they express MICA/B molecules, ligands for the NKG2D receptors found on NK cells. Transduction of LNCaP cells with the Ad5IL-12 vector prevented tumor formation in NOD.scid mice, indicating that NK cells alone can conduct tumor immunosurveillance and mediate protection. Intratumor injection of the Ad5IL-12 vector to established LNCaP tumors in NOD.scid mice resulted in a significant delay of tumor growth mediated by NK cell killing activity. The dependency of NK cells in this protective response was shown by the complete loss of Ad5IL-12 therapeutic efficacy on LNCaP tumors established in NOD.Cg-Rag1(tm1Mom)Prf1(tm1Sdz) congenic mice, which are devoid of NK cell activity. More pronounced attenuation of tumor growth and enhanced NK killing activity was observed when pharmacologic adrenalectomy with mitotane was done in combination with Ad5IL-12 vector treatment. The Ad5IL-12 vector treatment also induced killing of MICA/B-negative MHC class I-positive PC3 tumors formed in NOD.scid mice. Together, these results indicate that a targeted NK cell response could provide a generic approach for cancer immunotherapy, and that enhancing the NK cell response via control of cortisol levels may provide an additional therapeutic avenue in cancer.


Asunto(s)
Adenoviridae , Genes MHC Clase I , Terapia Genética/métodos , Hidrocortisona/metabolismo , Interleucina-12/uso terapéutico , Células Asesinas Naturales/fisiología , Neoplasias de la Próstata/terapia , Animales , Antineoplásicos Hormonales/uso terapéutico , Terapia Combinada , Vectores Genéticos , Humanos , Inmunidad Celular , Inmunoterapia/métodos , Interleucina-12/genética , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mitotano/uso terapéutico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Oncoimmunology ; 7(9): e1472195, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30228941

RESUMEN

A number of agents designed for immunotherapy of Acute Myeloid Leukemia (AML) are in preclinical and early clinical development. Most of them target a single antigen on the surface of AML cells. Here we describe the development and key biological properties of a tri-specific agent, the dual-targeting triplebody SPM-2, with binding sites for target antigens CD33 and CD123, and for CD16 to engage NK cells as cytolytic effectors. Primary blasts of nearly all AML patients carry at least one of these target antigens and the pair is particularly promising for the elimination of blasts and leukemia stem cells (LSCs) from a majority of AML patients by dual-targeting agents. The cytolytic activity of NK cells mediated by SPM-2 was analyzed in vitro for primary leukemic cells from 29 patients with a broad range of AML-subtypes. Blasts from all 29 patients, including patients with genomic alterations associated with an unfavorable genetic subtype, were lysed at nanomolar concentrations of SPM-2. Maximum susceptibility was observed for cells with a combined density of CD33 and CD123 above 10,000 copies/cell. Cell populations enriched for AML-LSCs (CD34pos and CD34pos CD38neg cells) from 2 AML patients carried an increased combined antigen density and were lysed at correspondingly lower concentrations of SPM-2 than unsorted blasts. These initial findings raise the expectation that SPM-2 may also be capable of eliminating AML-LSCs and thus of prolonging survival. In the future, patients with a broad range of AML subtypes may benefit from treatment with SPM-2.

9.
PLoS One ; 12(8): e0181615, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28767665

RESUMEN

BACKGROUND: We performed a systematic screening of colorectal cancer (CRC) tissues to investigate whether mismatch repair (MMR) status and ERCC1 protein expression could be predictive of clinical outcomes for these patients following the recommendation of The Evaluation of Genomic Applications in Practice of Prevention (EGAPP). METHODS: The expression of four MMR genes and ERCC1 were assessed by immunohistochemistry (IHC) from cancer tissue samples of 2233 consecutive CRC patients. RESULTS: We observed that most CRC patients with a proficient MMR (pMMR) status tended to have simultaneous ERCC1 protein expression (P< 0.001). Stage III CRC patients with deficient MMR (dMMR) had higher prognoses than the same stage patients with pMMR (DFS: 74% vs 65%, P = 0.04; OS: 79% vs 69%, P = 0.04). Here, dMMR is also associated with poorer survival for stage II patients after chemotherapy (DFS: 66% vs 78%, P = 0.04). Stage II and III patients that were shown to express ERCC1 protein had higher DFS and OS than those that were deficient in expression (stage II, DFS: 83% vs 70%, P = 0.006; OS 85% vs 73%, P = 0.02. Stage III, DFS: 67% vs56%, P = 0.03; OS: 71% vs 57%, P = 0.04). CONCLUSIONS: Our results indicate that dMMR appeared to predictive of a survival benefit for stage III CRC patients. We also found the determination of ERCC1 expression to be useful for predicting DFS or OS for stage II and III CRC patients. In addition, the expression of MMR genes and ERCC1 showed a significant relationship.


Asunto(s)
Neoplasias Colorrectales/patología , Proteínas de Unión al ADN/metabolismo , Endonucleasas/metabolismo , Expresión Génica , Endonucleasa PMS2 de Reparación del Emparejamiento Incorrecto/metabolismo , Homólogo 1 de la Proteína MutL/metabolismo , Proteína 2 Homóloga a MutS/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , China , Neoplasias Colorrectales/metabolismo , Reparación de la Incompatibilidad de ADN , Femenino , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Análisis de Supervivencia , Adulto Joven
10.
PLoS One ; 12(3): e0172799, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28253296

RESUMEN

BACKGROUND: The progression of colorectal cancer (CRC) may differ depending on the location of the tumor and the age of onset of the disease. Previous studies also suggested that the molecular basis of CRC varies with tumor location, which could affect the clinical management of patients. Therefore, we performed survival analysis looking at different age groups and mismatch repair status (MMR) of CRC patients according to primary tumor location in an attempt to identify subgroups of CRC that might help in the prognosis of disease. METHODS: A group of 2233 patients operated on to remove their CRC tumors were analyzed (521 with right colon cancer, 740 with left colon cancer and 972 with rectal cancer). The expression of four MMR genes was assessed by immunohistochemistry (IHC), independent of clinical criteria. From the data collected, a predictive model for overall survival (OS) could be constructed for some associations of tumor location and age of onset using Kaplan-Meier, logistic and Cox regression analysis. RESULTS: When tumor location was considered as the lone factor, we found no statistical difference in overall survival (OS) between right cancer (68%), left cancer (67%) or rectal cancer tumor locations (71%) (HR: 1.17, 95%CI (confidence interval): 0.97-1.43, P = 0.057). When age of onset was considered, middle age (40-59 years) and older (60-85 years) patients were found to have higher OS than younger onset cancer (20-39 years) patients (69% vs 71% vs 59%, HR: 1.07, 95% confidence interval (CI): 0.91-1.25, P = 0.008). When both age of onset and tumor location were considered in combination as disease factors, we found that the subgroup of patients with left colon cancer from middle age (69%) and older (67%) aged patients had higher OS than younger (54%) patients (HR: 0.89, 95%CI: 0.68-1.16, P = 0.048). However in patients with right colon cancers, we found no statistical difference is OS between younger, middle age or older grouped patients (60% vs 71% vs 67%, HR: 0.84, 95% CI: 0.61-1.16, P = 0.194). With regard to rectal located cancers, we found that younger (62%) and middle age (68) patients had lower OS than older (77%) patients (HR:1.46, 95%CI: 1.13-1.88, P = 0.004). The rates of deficient MMR (dMMR) was 10.4%. We found no statistical difference in OS stratified by tumor locations. However, right colon cancer patients with dMMR (86%) had higher OS than those with proficient MMR (pMMR) (63%) (HR: 3.01, 95% CI: 1.82-4.97, P<0.001). Left colon cancer patients with dMMR (76%) also had higher OS than those with pMMR (66%) (HR: 1.67, 95% CI: 0.95-2.92, P = 0.01). Oppositely, rectal cancer patients with dMMR (60%) had lower OS than those pMMR (68%) (HR: 0.77, 95% CI: 0.51-1.17, P = 0.04). CONCLUSIONS: These data demonstrate that primary tumor location can be an important factor when considered along with age of onset for the prognosis of CRC. Primary tumor location is also an important factor to evaluate the predictive effect of MMR status for the prognosis of CRC.


Asunto(s)
Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/genética , Reparación de la Incompatibilidad de ADN , Adulto , Edad de Inicio , Anciano , Anciano de 80 o más Años , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Pronóstico , Análisis de Supervivencia , Adulto Joven
11.
Oncol Res Treat ; 40(11): 702-706, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29065415

RESUMEN

BACKGROUND: The aim in this study was to determine if an association of excision repair cross-complementing group 1 (ERCC1) gene and mismatch repair (MMR) status with overall survival (OS) could be found from our analysis of a large cohort of Chinese colorectal cancer patients (CRC). METHODS: In total, 2,233 tissue samples isolated from individual CRC tumors were assessed by immunohistochemistry for the expression of ERCC1 and 4 MMR genes. RESULTS: The rates of proficient MMR (pMMR) and ERCC1 expression were 89.6 and 90.7%, respectively. We found that patients with positive ERCC1 expression and deficient (d)MMR status had higher overall survival (OS) than those with either positive ERCC1 and pMMR, negative ERCC1 and dMMR, or negative ERCC1 expression and pMMR status (OS 79 vs. 69 vs. 66 vs. 61%, hazard ratio (HR) 0.90, 95% confidence interval (CI) 0.80-1.00; p = 0.043). Despite this finding, we found no statistical difference in OS between ERCC1-positive and -negative CRC patients when ERCC1 expression was considered alone (OS 70 vs. 62%, HR 0.82, 95% CI 0.65-1.04; p = 0.11). CONCLUSION: Our results indicate that the combined examination of ERCC1 expression and dMMR status can be used to aid OS assessment in CRC patients.


Asunto(s)
Neoplasias Colorrectales/genética , Neoplasias Colorrectales/mortalidad , Reparación de la Incompatibilidad de ADN/genética , Proteínas de Unión al ADN/genética , Endonucleasas/genética , Adulto , Anciano , Anciano de 80 o más Años , China , Estudios de Cohortes , Neoplasias Colorrectales/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Estadística como Asunto , Análisis de Supervivencia
12.
Cancer Med ; 6(5): 975-981, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28345223

RESUMEN

Previous studies have suggested that deficiencies in mismatch repair genes (dMMR) often occur in patients with colorectal cancer (CRC) and contribute to disease etiology. Here, we looked for a correlation of MMR status to disease outcomes from a large number of Chinese CRC patients stratified by the age of onset of disease. A total of 2233 CRC patients were analyzed and tissue biopsies of surgically removed tumors scored for MMR gene status. The patient distribution after classification consisted of 188 younger aged patients (20-39 years of age), 1024 middle aged patients (40-59 years of age), and 1020 older aged patients (60-85 years of age). In this analysis, the expression of four MMR genes was assessed by immunohistochemistry (IHC). We found that the young group of CRC patients with dMMR had higher overall survival (OS) than the young group of patients with proficient MMR (pMMR) (77% vs. 56%, P = 0.03). Middle-aged patients with dMMR also had higher OS than middle-aged group patients with pMMR (78% vs. 68%, P = 0.012). However, we found no statistical difference in OS between dMMR and pMMR status in the older group of patients (75% vs. 71%, P = 0.224). Finally, the middle- and older-aged group set of patients had higher OS than the young group of patients (69% vs. 71% vs. 59%, P = 0.008). These data demonstrated that the age of disease onset can be an important factor to help evaluate the prognosis of CRC when combined with the analysis of MMR status within tumor biopsied tissue.


Asunto(s)
Neoplasias Colorrectales/cirugía , Proteínas de Unión al ADN/metabolismo , Endonucleasa PMS2 de Reparación del Emparejamiento Incorrecto/metabolismo , Homólogo 1 de la Proteína MutL/metabolismo , Proteína 2 Homóloga a MutS/metabolismo , Adulto , Distribución por Edad , Edad de Inicio , Anciano , Anciano de 80 o más Años , China , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Enzimas Reparadoras del ADN/metabolismo , Supervivencia sin Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Pronóstico , Análisis de Supervivencia , Adulto Joven
13.
Oncol Res Treat ; 39(11): 696-702, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27855388

RESUMEN

BACKGROUND: Conflicting results have been reported about the association between the Ki67 labeling index (Ki67-Li) and clinical outcome in patients with colorectal cancer (CRC). PATIENTS AND METHODS: Ki67 expression was assessed by immunohistochemistry (IHC) in 2,233 consecutive CRC cases. RESULTS: We determined 992 cases to have a low and 1,241 cases to have a high Ki67-Li (representing an approximately 44-56% breakdown in distribution between low versus high patients designated by phenotype). Stage III patients with a high Ki67-Li had higher 3-year disease-free survival (DFS) and overall survival (OS) than those with a low Ki67-Li (DFS 70 vs. 61%; p = 0.02 and OS 75 vs. 64%; p = 0.008). We also found significantly improved 3-year progression-free survival (PFS) for stage IV patients in the high versus the low Ki67-Li group (PFS 14 vs. 10%; p = 0.02). Yet, we found no statistical differences in prognosis for stage I and II patients and in OS for stage IV patients between high versus low Ki67-Li (p > 0.05). CONCLUSION: Our results suggest that high Ki67-Li can be an independent prognostic biomarker to aid the assessment of patient outcomes in both stage III and IV CRC.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/mortalidad , Antígeno Ki-67/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , China/epidemiología , Neoplasias Colorrectales/diagnóstico , Supervivencia sin Enfermedad , Femenino , Alemania/epidemiología , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Prevalencia , Pronóstico , Reproducibilidad de los Resultados , Medición de Riesgo/métodos , Sensibilidad y Especificidad , Tasa de Supervivencia , Adulto Joven
14.
Oncotarget ; 7(16): 22579-89, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-26981773

RESUMEN

Simultaneous targeting of multiple tumor-associated antigens (TAAs) in cancer immunotherapy is presumed to enhance tumor cell selectivity and to reduce immune escape.The combination of B lymphoid marker CD19 and myeloid marker CD33 is exclusively present on biphenotypic B/myeloid leukemia cells. Triplebody 33-3-19 binds specifically to both of these TAAs and activates T cells as immune effectors. Thereby it induces specific lysis of established myeloid (MOLM13, THP-1) and B-lymphoid cell lines (BV173, SEM, Raji, ARH77) as well as of primary patient cells. EC50 values range from 3 pM to 2.4 nM. In accordance with our hypothesis, 33-3-19 is able to induce preferential lysis of double- rather than single-positive leukemia cells in a target cell mixture: CD19/CD33 double-positive BV173 cells were eliminated to a significantly greater extent than CD19 single-positive SEM cells (36.6% vs. 20.9% in 3 hours, p = 0.0048) in the presence of both cell lines. In contrast, equivalent elimination efficiencies were observed for both cell lines, when control triplebody 19-3-19 or a mixture of the bispecific single chain variable fragments 19-3 and 33-3 were used. This result highlights the potential of dual-targeting agents for efficient and selective immune-intervention in leukemia patients.


Asunto(s)
Antígenos de Neoplasias/efectos de los fármacos , Antineoplásicos/farmacología , Inmunoterapia/métodos , Leucemia Bifenotípica Aguda , Anticuerpos de Cadena Única/farmacología , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Antígenos CD19/inmunología , Antígenos de Diferenciación Mielomonocítica/efectos de los fármacos , Antígenos de Diferenciación Mielomonocítica/inmunología , Humanos , Lectina 3 Similar a Ig de Unión al Ácido Siálico/inmunología
15.
Oncotarget ; 7(50): 83392-83408, 2016 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-27825135

RESUMEN

Triplebodies are antibody-derived recombinant proteins carrying 3 antigen-binding domains in a single polypeptide chain. Triplebody SPM-1 was designed for lysis of CD19-bearing malignant B-lymphoid cells through the engagement of CD16-expressing cytolytic effectors, including NK and γδ T cells.SPM-1 is an optimized version of triplebody ds(19-16-19) and includes humanization, disulfide stabilization and the removal of potentially immunogenic sequences. A three-step chromatographic procedure yielded 1.7 - 5.5 mg of purified, monomeric protein per liter of culture medium. In cytolysis assays with NK cell effectors, SPM-1 mediated potent lysis of cancer-derived B cell lines and primary cells from patients with various B-lymphoid malignancies, which surpassed the ADCC activity of the therapeutic antibody Rituximab. EC50-values ranged from 3 to 86 pM. Finally, in an impedance-based assay, SPM-1 mediated a particularly rapid lysis of CD19-bearing target cells by engaging and activating both primary and expanded human γδ T cells from healthy donors as effectors.These data establish SPM-1 as a useful tool for a kinetic analysis of the cytolytic reactions mediated by γδ T and NK cells and as an agent deserving further development towards clinical use for the treatment of B-lymphoid malignancies.


Asunto(s)
Antígenos CD19/inmunología , Antineoplásicos Inmunológicos/farmacología , Citotoxicidad Inmunológica/efectos de los fármacos , Linfocitos Intraepiteliales/efectos de los fármacos , Células Asesinas Naturales/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfoma de Células B/tratamiento farmacológico , Antineoplásicos Inmunológicos/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Linfocitos Intraepiteliales/inmunología , Células Asesinas Naturales/inmunología , Cinética , Activación de Linfocitos/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Linfoma de Células B/inmunología , Linfoma de Células B/patología , Rituximab/farmacología , Células Tumorales Cultivadas
16.
Oncotarget ; 5(15): 6466-83, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25115385

RESUMEN

Triplebody 19-3-19, an antibody-derived protein, carries three single chain fragment variable domains in tandem in a single polypeptide chain. 19-3-19 binds CD19-bearing lymphoid cells via its two distal domains and primary T cells via its CD3-targeting central domain in an antigen-specific manner. Here, malignant B-lymphoid cell lines and primary cells from patients with B cell malignancies were used as targets in cytotoxicity tests with pre-stimulated allogeneic T cells as effectors. 19-3-19 mediated up to 95 % specific lysis of CD19-positive tumor cells and, at picomolar EC50 doses, had similar cytolytic potency as the clinically successful agent Blinatumomab. 19-3-19 activated resting T cells from healthy unrelated donors and mediated specific lysis of both autologous and allogeneic CD19-positive cells. 19-3-19 led to the elimination of 70 % of CD19-positive target cells even with resting T cells as effectors at an effector-to-target cell ratio of 1 : 10. The molecule is therefore capable of mediating serial lysis of target cells by a single T cell. These results highlight that central domains capable of engaging different immune effectors can be incorporated into the triplebody format to provide more individualized therapy tailored to a patient's specific immune status.


Asunto(s)
Linfocitos/inmunología , Linfoma de Células B/terapia , Anticuerpos de Cadena Única/farmacología , Linfocitos T/inmunología , Anciano , Anciano de 80 o más Años , Citotoxicidad Celular Dependiente de Anticuerpos , Femenino , Células HEK293 , Humanos , Inmunización Pasiva/métodos , Activación de Linfocitos , Linfoma de Células B/inmunología , Masculino , Persona de Mediana Edad , Anticuerpos de Cadena Única/inmunología , Adulto Joven
17.
Cancers (Basel) ; 3(3): 2870-85, 2011 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-24212937

RESUMEN

Vimentin was originally identified as an intermediate filament protein present only as an intracellular component in many cell types. However, this protein has now been detected on the surface of a number of different cancer cell types in a punctate distribution pattern. Increased vimentin expression has been indicated as an important step in epithelial-mesenchymal transition (EMT) required for the metastasis of prostate cancer. Here, using two vimentin-specific monoclonal antibodies (SC5 and V9 directed against the coil one rod domain and the C-terminus of the vimentin protein, respectively), we examined whether either of these domains would be displayed on the surface of three commonly studied prostate cancer cell lines isolated from different sites of metastases. Confocal analysis of LNCaP, PC3 and DU145 prostate cancer cell lines (derived from lymph node, bone or brain prostate metastases, respectively) demonstrated that both domains of vimentin are present on the surface of these metastatic cancer cell types. In addition, flow cytometric analysis revealed that vimentin expression was readily detected along with CD44 expression but only a small subpopulation of prostate cancer cells expressed vimentin and the putative stem cell marker CD133 along with CD44. Finally, Cowpea mosaic virus (CPMV) nanoparticles that target vimentin could bind and internalize into tested prostate cancer cell lines. These results demonstrate that at least two domains of vimentin are present on the surface of metastatic prostate cancer cells and suggest that vimentin could provide a useful target for nanoparticle- or antibody- cancer therapeutic agents directed against highly invasive cancer and/or stem cells.

18.
Vaccine ; 23(2): 247-57, 2004 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-15531044

RESUMEN

Immunization with soluble leishmanial antigen (SLA) in IFA plus Ad5IL-12 vector induced protection confined to the immunized footpad in BALB/c mice. However, animals that controlled a primary infection with a Leishmania major challenge in the same immunized footpad, became resistant to subsequent contralateral rechallenges due to expansion of IFN-gamma secreting cells. This systemic immunity could be disrupted either by macrophage depletion during immunization or by lymphadenectomy after challenge. We show that this procedure does not interfere with tissue-compartmentalized protection, since lymphadenectomized and splenectomized animals were resistant to rechallenges performed in the immunized footpads. Our results indicate that SLA-Ad5IL-12 vector priming requires macrophages to generate systemic protection. Furthermore, a previously undescribed lymphoid organ-independent, protective immune response is contained within the tissue microenvironment of the immunized/challenged footpad. These results have important implications for vaccine design against leishmanial and mycobacterial infections and diseases caused by intracellular pathogens.


Asunto(s)
Interleucina-12/uso terapéutico , Leishmania major/inmunología , Leishmaniasis Cutánea/prevención & control , Ganglios Linfáticos/inmunología , Macrófagos/inmunología , Adenoviridae/genética , Animales , Antígenos de Protozoos/inmunología , Línea Celular , Vectores Genéticos/administración & dosificación , Interleucina-12/genética , Leishmania major/genética , Leishmaniasis Cutánea/inmunología , Leishmaniasis Cutánea/parasitología , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Vacunas Sintéticas/inmunología
19.
J Immunol ; 170(2): 765-74, 2003 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-12517939

RESUMEN

Adenovirus vectors are increasingly being used for genetic vaccination and may prove highly suitable for intervention in different pathological conditions due to their capacity to generate high level, transient gene expression. In this study, we report the use of a recombinant adenovirus vector to induce regulatory responses for the prevention of autoimmune diseases through transient expression of a TCR beta-chain. Immunization of B10.PL mice with a recombinant adenovirus expressing the TCR Vbeta8.2 chain (Ad5E1 mVbeta8.2), resulted in induction of regulatory type 1 CD4 T cells, directed against the framework region 3 determinant within the B5 peptide (aa 76-101) of the Vbeta8.2 chain. This determinant is readily processed and displayed in an I-A(u) context, on ambient APC. Transient genetic delivery of the TCR Vbeta8.2 chain protected mice from Ag-induced experimental autoimmune encephalomyelitis. However, when the Ad5E1 mVbeta8.2 vector was coadministered with either an IL-4- or IL-10-expressing vector, regulation was disrupted and disease was exacerbated. These results highlight the importance of the Th1-like cytokine requirement necessary for the generation and activity of effective regulatory T cells in this model of experimental autoimmune encephalomyelitis.


Asunto(s)
Adenovirus Humanos/genética , Adenovirus Humanos/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/prevención & control , Vectores Genéticos/inmunología , Interleucina-10/biosíntesis , Interleucina-4/biosíntesis , Receptores de Antígenos de Linfocitos T alfa-beta/biosíntesis , Secuencia de Aminoácidos , Animales , División Celular/genética , División Celular/inmunología , Citocinas/biosíntesis , Relación Dosis-Respuesta Inmunológica , Femenino , Vectores Genéticos/administración & dosificación , Vectores Genéticos/síntesis química , Humanos , Inmunización , Epítopos Inmunodominantes/toxicidad , Región Variable de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/inmunología , Inmunofenotipificación , Inyecciones Intramusculares , Inyecciones Intraperitoneales , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Proteína Básica de Mielina/toxicidad , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/toxicidad , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Recombinación Genética/inmunología , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Células TH1/inmunología , Células TH1/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda