Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
J Immunother Cancer ; 12(10)2024 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-39384197

RESUMEN

BACKGROUND: We aimed to develop a chemoimmunotherapy regimen consisting of a novel Wilms' tumor 1 (WT1) peptide-pulsed dendritic cell (WT1-DC) vaccine and multiagent chemotherapy and to investigate the safety, clinical outcomes, and WT1-specific immune responses of patients with unresectable advanced pancreatic ductal adenocarcinoma (UR-PDAC) who received this treatment. METHODS: Patients with UR-PDAC with stage III disease (locally advanced (LA-PDAC; n=6)), stage IV disease (metastatic (M-PDAC; n=3)), or recurrent disease after surgery (n=1) were enrolled in this phase I study. The patients received one cycle of nab-paclitaxel plus gemcitabine alone followed by 15 doses of the WT1-DC vaccine independent of chemotherapy. The novel WT1 peptide cocktail was composed of a multifunctional helper peptide specific for major histocompatibility complex class II, human leukocyte antigen (HLA)-A*02:01, or HLA-A*02:06 and a killer peptide specific for HLA-A*24:02. RESULTS: The chemoimmunotherapy regimen was well tolerated. In the nine patients for whom a prognostic analysis was feasible, the clinical outcomes of long-term WT1 peptide-specific delayed-type hypersensitivity (WT1-DTH)-positive patients (n=4) were significantly superior to those of short-term WT1-DTH-positive patients (n=5). During chemoimmunotherapy, eight patients were deemed eligible for conversion surgery and underwent R0 resection (four patients with LA-PDAC, one patient with M-PDAC, and one recurrence) or R1 resection (one patient with M-PDAC), and one patient with LA-PDAC was determined to be unresectable. Long-term WT1-DTH positivity was observed in three of the four patients with R0-resected LA-PDAC. These three patients exhibited notable infiltration of T cells and programmed cell death protein-1+ cells within the pancreatic tumor microenvironment (TME). All patients with long-term WT1-DTH positivity were alive for at least 4.5 years after starting therapy. In patients with long-term WT1-DTH positivity, the percentage of WT1-specific circulating CD4+ or CD8+ T cells that produced IFN-γ or TNF-α was significantly greater than that in patients with short-term WT1-DTH positivity after two vaccinations. Moreover, after 12 vaccinations, the percentages of both circulating regulatory T cells and myeloid-derived suppressor cells were significantly lower in patients with long-term WT1-DTH-positive PDAC than in short-term WT1-DTH-positive patients. CONCLUSIONS: Potent activation of WT1-specific immune responses through a combination chemoimmunotherapy regimen including the WT1-DC vaccine in patients with UR-PDAC may modulate the TME and enable conversion surgery, resulting in clinical benefits (Online supplemental file 1). TRIAL REGISTRATION NUMBER: jRCTc030190195.


Asunto(s)
Vacunas contra el Cáncer , Células Dendríticas , Neoplasias Pancreáticas , Microambiente Tumoral , Proteínas WT1 , Humanos , Masculino , Proteínas WT1/inmunología , Proteínas WT1/uso terapéutico , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/terapia , Neoplasias Pancreáticas/inmunología , Células Dendríticas/inmunología , Femenino , Persona de Mediana Edad , Anciano , Vacunas contra el Cáncer/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapéutico , Desoxicitidina/farmacología , Paclitaxel/uso terapéutico , Paclitaxel/farmacología , Gemcitabina , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/terapia , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/cirugía , Albúminas/uso terapéutico , Inmunoterapia/métodos , Adulto
2.
Sci Rep ; 12(1): 10698, 2022 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-35739324

RESUMEN

Fusobacteria have been suspected to be pathobionts of colon cancer and inflammatory bowel disease. However, the immunomodulatory properties that affect these inflammatory reactions in dendritic cells (DCs) under anaerobic and aerobic conditions have not yet been characterized. We directly assessed the stimulatory effects of anaerobic commensal bacteria, including fusobacteria, on a human DC line through coculture under aerobic or anaerobic conditions. Under aerobic or anaerobic conditions, stimulation of the DC line with all live commensal bacteria examined, except the probiotic Lactobacillus delbrueckii subsp. bulgaricus (L. bulgaricus), significantly increased the geometric mean fluorescent intensity (MFI) of marker proteins (HLA-ABC, HLA-DR, CD80, CD86, CD83, or CCR7) on the DC surface. In particular, both Fusobacterium nucleatum (F. nucleatum) and Escherichia coli (E. coli) significantly increased the expression of DC-associated molecules, except for CD83 under both aerobic and anaerobic conditions. The DC line stimulated with Fusobacterium varium (F. varium) significantly increased only CD80, HLA-ABC, and HLA-DR expression under anaerobic conditions. Moreover, differences in the levels of proinflammatory cytokines, such as IL-6, IL-8, and TNF-α, were detected in the DC line stimulated by all live commensal bacteria under either aerobic or anaerobic conditions. Under aerobic conditions, the DC line stimulated with E. coli produced significantly more IL-6, IL-8, and TNF-α than did the cells stimulated with any of the bacteria examined. When E. coli were used to stimulate the DC line under anaerobic conditions, TNF-α was predominantly produced compared to stimulation with any other bacteria. Compared to the DC line stimulated with any other bacteria, the cells stimulated with F. nucleatum showed significantly increased production of IL-6, IL-8 and TNF-α only under anaerobic conditions. In particular, E. coli, F. nucleatum, and F. varium strongly stimulated the DC line, resulting in significantly increased expression of surface molecules associated with DCs and production of inflammatory cytokines.


Asunto(s)
Células Dendríticas , Factor de Necrosis Tumoral alfa , Anaerobiosis , Antígeno B7-1/metabolismo , Células Cultivadas , Citocinas/metabolismo , Escherichia coli/metabolismo , Fusobacterias , Antígenos HLA-DR/metabolismo , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
3.
BMC Cancer ; 21(1): 1197, 2021 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-34758773

RESUMEN

BACKGROUND: Both activated tumor-infiltrating lymphocytes (TILs) and immune-suppressive cells, such as regulatory T cells (Tregs), in the tumor microenvironment (TME) play an important role in the prognosis of patients with pancreatic ductal adenocarcinoma (PDAC). METHODS: The densities of TILs, programmed death receptor 1 (PD-1) + T cells, and forkhead box P3 (Foxp3) + T cells were analyzed by immunohistochemical staining. The associations of the immunological status of the PDAC microenvironment with overall survival (OS) time and disease-free survival (DFS) time were evaluated. RESULTS: PDAC patients with a high density of TILs in the TME or PD-1-positive T cells in tertiary lymphoid aggregates (TLAs) demonstrated a significantly better prognosis than those with a low density of TILs or PD-1-negativity, respectively. Moreover, PDAC patients with high levels of Foxp3-expressing T cells showed a worse prognosis than those with low levels of Foxp3-expressing T cells. Importantly, even with a high density of the TILs in TME or PD-1-positive T cells in TLAs, PDAC patients with high levels of Foxp3-expressing T cells showed a worse prognosis than patients with low levels of Foxp3-expressing T cells. A PDAC TME with a high density of TILs/high PD-1 positivity/low Foxp3 expression was an independent predictive marker associated with superior prognosis. CONCLUSION: Combined assessment of TILs, PD-1+ cells, and Foxp3+ T cells in the TME may predict the prognosis of PDAC patients following surgical resection.


Asunto(s)
Carcinoma Ductal Pancreático/mortalidad , Linfocitos Infiltrantes de Tumor/inmunología , Recurrencia Local de Neoplasia/epidemiología , Neoplasias Pancreáticas/inmunología , Microambiente Tumoral/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/cirugía , Supervivencia sin Enfermedad , Femenino , Estudios de Seguimiento , Factores de Transcripción Forkhead/metabolismo , Humanos , Linfocitos Infiltrantes de Tumor/metabolismo , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/prevención & control , Páncreas/inmunología , Páncreas/patología , Páncreas/cirugía , Pancreatectomía , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/cirugía , Pronóstico , Receptor de Muerte Celular Programada 1/metabolismo , Estudios Retrospectivos
4.
Int J Oncol ; 57(4): 1047-1056, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32945369

RESUMEN

The proliferation and activation of CD4+ T helper 1 (Th1) cells and CD8+ cytotoxic T lymphocytes (CTLs) that produce interferon­Î³ (IFN­Î³) is an essential action of effective cancer vaccines. Recently, a novel Wilms' tumor 1 (WT1) helper peptide (WT1 HP34­51; amino acid sequence, WAPVLDFAPPGASAYGSL) applicable for various human leukocyte antigen (HLA) subtypes (HLA­DR, HLA­DP and HLA­DQ) was reported to increase peptide immunogenicity; however, the function of WT1 HP34­51 remains unclear. In the present study, mature dendritic cells (mDCs) pulsed with WT1 HP34­51 (mDC/WT1 HP34­51) activated not only WT1­specific CD4+ T cells but also CD8+ T cells that produced IFN­Î³ following stimulation with immature dendritic cells (imDCs) pulsed with WT1 killer peptide (imDC/WT1 KP37­45) in an HLA­A*02:01­ or HLA­A*02:06­restricted manner. Furthermore, the activated WT1­reactive CD4+ Th1 cells were predominantly effector memory (EM) T cells. In 5 of 12 (41.7%) patients with cancer carrying the HLA­A*02:01 or HLA­A*02:06 allele, WT1­reactive CD8+ T cells stimulated with mDC/WT1 HP34­51 enhanced their levels of WT1 KP37­45­specific IFN­Î³ production, with an increase >10%. Simultaneous activation of CD4+ and CD8+ T cells occurred more often when stimulation with mDC/WT1 HP34­51 was combined with imDC/WT1 KP37­45 restimulation. These results indicated that the novel mDC/WT1 HP34­51 combination induced responses by WT1­specific EM CD4+ Th1 cells and HLA­A*02:01­ or HLA­A*02:06­restricted CD8+ CTLs, suggesting its potential as a WT1­targeting cancer vaccine.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Antígeno HLA-A2/inmunología , Neoplasias Renales/terapia , Fragmentos de Péptidos/farmacología , Proteínas WT1/inmunología , Tumor de Wilms/terapia , Adulto , Anciano , Anciano de 80 o más Años , Linfocitos T CD4-Positivos/inmunología , Células Cultivadas , Células Dendríticas/efectos de los fármacos , Células Dendríticas/trasplante , Femenino , Humanos , Inmunoterapia Adoptiva/métodos , Neoplasias Renales/sangre , Neoplasias Renales/inmunología , Leucocitos Mononucleares , Masculino , Persona de Mediana Edad , Células TH1/inmunología , Tumor de Wilms/sangre , Tumor de Wilms/inmunología
5.
Cancer Invest ; 37(9): 463-477, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31490702

RESUMEN

The associations of the immunological status of the pancreatic ductal adenocarcinoma (PDA) microenvironment with prognosis were assessed. A high tumor-infiltrating lymphocyte (TIL) density was associated with a better prognosis. Importantly, even with a high density of TILs, the PDA cells with programed cell death-ligand 1 (PD-L1) expression showed a worse prognosis than the patients with negative PD-L1 expression. A significant association between a better prognosis and a tumor microenvironment with a high TIL density/negative PD-L1 expression was observed. Assessments of a combined immunological status in the tumor microenvironment may predict the prognosis of PDA patients following surgical resection.


Asunto(s)
Antígeno B7-H1/metabolismo , Carcinoma Ductal Pancreático/cirugía , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias Pancreáticas/cirugía , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma Ductal Pancreático/inmunología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/inmunología , Pronóstico , Análisis de Supervivencia , Linfocitos T/inmunología , Microambiente Tumoral
6.
Oncology ; 97(3): 135-148, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31216557

RESUMEN

BACKGROUND: We have developed a Wilms' tumor 1 (WT1)-targeting dendritic cell (DC)-based cancer vaccine combined with standard chemotherapy for patients with advanced pancreatic ductal adenocarcinoma (PDA). METHODS: We evaluated predictive markers of overall survival (OS) in PDA patients treated with multiple major histocompatibility complex class I/II-restricted, WT1 peptide-pulsed DC vaccinations (DC/WT1-I/II) in combination with chemotherapy. Throughout the entire period of immunochemotherapy, the plasma levels of soluble factors derived from granulocytes of 7 eligible PDA patients were examined. Moreover, systemic inflammatory response markers (neutrophil-to-lymphocyte ratio [NLR], monocyte-to-lymphocyte ratio [MLR], and granulocyte-to-lymphocyte ratio [GLR]) were assessed. In addition, cytoplasmic WT1 expression in PDA cells was examined. RESULTS: Compared to the 4 non-super-responders (OS <1 year), the remaining 3 super-responders (OS ≥1 year) showed significantly decreased low plasma matrix metalloproteinase-9 levels throughout long-term therapy. The NLR, MLR, and GLR after 5 DC/WT1-I/II vaccinations and 3 cycles of gemcitabine were significantly lower in the super-responders than in the non-super-responders. Furthermore, the cytoplasmic WT1 expression in the PDA cells of super-responders was relatively weak compared to that in the PDA cells of non-super-responders. CONCLUSIONS: Prolonged low levels of a granulocyte-related systemic inflammatory response after the early period of therapy and low cytoplasmic WT1 expression in PDA cells may be markers predictive of OS in PDA patients receiving WT1-targeting immunochemotherapy.


Asunto(s)
Biomarcadores de Tumor , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Inmunoterapia , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/mortalidad , Proteínas WT1/inmunología , Biomarcadores , Vacunas contra el Cáncer/administración & dosificación , Terapia Combinada , Células Dendríticas/metabolismo , Epítopos/inmunología , Femenino , Humanos , Inmunofenotipificación , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/terapia , Péptidos/inmunología , Peroxidasa/metabolismo , Pronóstico , Factor de Crecimiento Transformador beta1/metabolismo , Resultado del Tratamiento , Vacunación , Proteínas WT1/genética
7.
J Biol Chem ; 292(7): 2815-2829, 2017 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-28053092

RESUMEN

Differentiation therapy with all-trans-retinoic acid (ATRA) improves the treatment outcome of acute promyelocytic leukemia (APL); however, the molecular mechanism by which ATRA induces granulocytic differentiation remains unclear. We previously reported that the inhibition of the NAD-dependent histone deacetylase (HDAC) SIRT2 induces granulocytic differentiation in leukemia cells, suggesting the involvement of protein acetylation in ATRA-induced leukemia cell differentiation. Herein, we show that p300/CREB-binding protein-associated factor (PCAF), a histone acetyltransferase (HAT), is a prerequisite for ATRA-induced granulocytic differentiation in leukemia cells. We found that PCAF expression was markedly increased in leukemia cell lines (NB4 and HL-60) and primary APL cells during ATRA-induced granulocytic differentiation. Consistent with these results, the expression of PCAF was markedly up-regulated in the bone marrow cells of APL patients who received ATRA-containing chemotherapy. The knockdown of PCAF inhibited ATRA-induced granulocytic differentiation in leukemia cell lines and primary APL cells. Conversely, the overexpression of PCAF induced the expression of the granulocytic differentiation marker CD11b at the mRNA level. Acetylome analysis identified the acetylated proteins after ATRA treatment, and we found that histone H3, a known PCAF acetylation substrate, was preferentially acetylated by the ATRA treatment. Furthermore, we have demonstrated that PCAF is required for the acetylation of histone H3 on the promoter of ATRA target genes, such as CCL2 and FGR, and for the expression of these genes in ATRA-treated leukemia cells. These results strongly support our hypothesis that PCAF is induced and activated by ATRA, and the subsequent acetylation of PCAF substrates promotes granulocytic differentiation in leukemia cells. Targeting PCAF and its downstream acetylation targets could serve as a novel therapeutic strategy to overcome all subtypes of AML.


Asunto(s)
Diferenciación Celular/fisiología , Granulocitos/efectos de los fármacos , Leucemia Mieloide Aguda/patología , Tretinoina/farmacología , Factores de Transcripción p300-CBP/fisiología , Acetilación , Antígeno CD11b/genética , Diferenciación Celular/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Granulocitos/patología , Células HL-60 , Histonas/metabolismo , Humanos , Factores de Transcripción p300-CBP/genética
8.
Cancer Sci ; 107(5): 682-9, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26920337

RESUMEN

Although rituximab, a chimeric monoclonal antibody that specifically binds to CD20, has significantly improved the prognosis for diffuse large B cell lymphoma (DLBCL), one-third of DLBCL patients demonstrate resistance to rituximab or relapse after rituximab treatment. Thus, a novel approach to rituximab-based treatment is likely to be required to improve the efficacy of DLBCL treatment. As complement dependent cytotoxicity (CDC) is a key mechanism mediating rituximab's tumoricidal activity, rituximab binding to CD20 on tumor cells is a critical factor for effective rituximab-based treatments against DLBCL. We found that gemcitabine (GEM), but not lenalidomide (LEN) or azacitidine (AZA), can upregulate CD20 expression in TK and KML-1 cells, two human DLBCL cell lines. Treatment of TK and KML-1 cells with GEM enhanced CD20 expression at both the mRNA and protein levels. CD20 upregulation by GEM treatment was accompanied by increased rituximab binding to CD20. In TK cells, GEM treatment synergistically increased rituximab-mediated CDC activity in a dose-dependent manner. In KML cells, GEM treatment also induced upregulation of complement regulatory proteins, possibly leading to resistance to CDC. Treatment with LEN, a drug that did not upregulate CD20, did not enhance rituximab-mediated CDC activity. GEM treatment activated nuclear factor-kappa B (NF-kB) signaling in these cells. Furthermore, a specific inhibitor to NF-kB suppressed GEM-induced CD20 upregulation, indicating that GEM-induced NF-kB activation is closely associated with CD20 upregulation. These results suggest that when used in combination, GEM might enhance the antitumor efficacy of rituximab against DLBCL due to its unique ability to upregulate CD20.


Asunto(s)
Antígenos CD20/metabolismo , Proteínas del Sistema Complemento/efectos de los fármacos , Citotoxicidad Inmunológica/efectos de los fármacos , Desoxicitidina/análogos & derivados , Linfoma de Células B/inmunología , Linfoma de Células B/metabolismo , Rituximab/farmacología , Regulación hacia Arriba/efectos de los fármacos , Azacitidina/farmacología , Línea Celular Tumoral , Proteínas del Sistema Complemento/inmunología , Desoxicitidina/farmacología , Humanos , Lenalidomida , Linfoma de Células B/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/metabolismo , FN-kappa B/metabolismo , Talidomida/análogos & derivados , Talidomida/farmacología , Gemcitabina
9.
Blood ; 127(10): 1307-16, 2016 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-26817954

RESUMEN

Recurrent somatic mutations of calreticulin (CALR) have been identified in patients harboring myeloproliferative neoplasms; however, their role in tumorigenesis remains elusive. Here, we found that the expression of mutant but not wild-type CALR induces the thrombopoietin (TPO)-independent growth of UT-7/TPO cells. We demonstrated that c-MPL, the TPO receptor, is required for this cytokine-independent growth of UT-7/TPO cells. Mutant CALR preferentially associates with c-MPL that is bound to Janus kinase 2 (JAK2) over the wild-type protein. Furthermore, we demonstrated that the mutant-specific carboxyl terminus portion of CALR interferes with the P-domain of CALR to allow the N-domain to interact with c-MPL, providing an explanation for the gain-of-function property of mutant CALR. We showed that mutant CALR induces the phosphorylation of JAK2 and its downstream signaling molecules in UT-7/TPO cells and that this induction was blocked by JAK2 inhibitor treatment. Finally, we demonstrated that c-MPL is required for TPO-independent megakaryopoiesis in induced pluripotent stem cell-derived hematopoietic stem cells harboring the CALR mutation. These findings imply that mutant CALR activates the JAK2 downstream pathway via its association with c-MPL. Considering these results, we propose that mutant CALR promotes myeloproliferative neoplasm development by activating c-MPL and its downstream pathway.


Asunto(s)
Calreticulina/metabolismo , Neoplasias Hematológicas/metabolismo , Trastornos Mieloproliferativos/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores de Trombopoyetina/metabolismo , Calreticulina/genética , Línea Celular Tumoral , Células HEK293 , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/mortalidad , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Janus Quinasa 2/metabolismo , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/patología , Proteínas de Neoplasias/genética , Fosforilación , Estructura Terciaria de Proteína , Receptores de Trombopoyetina/genética , Trombopoyesis/genética , Trombopoyetina/metabolismo
10.
BMC Cancer ; 15: 726, 2015 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-26475267

RESUMEN

BACKGROUND: Although pancreatic ductal adenocarcinomas (PDAs) widely express HER2, the expression level is generally low. If HER2 expression in PDA cells could be enhanced by treatment with a given agent, then combination therapy with that agent and trastuzumab emtansine (T-DM1), a chemotherapeutic agent that is a conjugate of trastuzumab, might lead to significant antitumor effects against PDA. METHODS: Cell proliferation was examined by spectrophotometry. HER2 expression was examined by flow cytometry, immunoblot and quantitative reverse transcription polymerase chain reaction. T-DM1 binding to cells was examined by flow cytometry and enzyme-linked immunosorbent assay. RESULTS: Out of 5 tested human PDA cell lines, including MIA PaCa-2, three showed increases in HER2 expression after gemcitabine (GEM) treatment. The binding of T-DM1 to GEM-treated MIA PaCa-2 cells was higher than to untreated MIA PaCa-2 cells. Treatment with GEM and T-DM1 showed synergic cytotoxic effects on MIA PaCa-2 cells in vitro. Cells in the G2M phase of the cell cycle were retained after GEM treatment and showed higher levels of HER2 expression, possibly contributing to the synergic effect of GEM and T-DM1. CONCLUSIONS: Combined treatment with GEM and T-DM1 might confer a potent therapeutic modality against PDA as a result of GEM-mediated HER2 up-regulation.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Neoplasias Pancreáticas/tratamiento farmacológico , Receptor ErbB-2/biosíntesis , Adenocarcinoma/genética , Adenocarcinoma/patología , Ado-Trastuzumab Emtansina , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Línea Celular Tumoral , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Maitansina/administración & dosificación , Maitansina/análogos & derivados , Ratones , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Receptor ErbB-2/genética , Trastuzumab , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
11.
Oncol Rep ; 34(4): 2099-105, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26239720

RESUMEN

Monoclonal antibody therapy for immune checkpoint blockade has achieved promising results for several types of malignant tumors. For the future treatment of gastrointestinal stromal tumors (GISTs) by immune checkpoint blockade, expression of immune checkpoint-related molecules that suppress antitumor immunity in GISTs was examined. Infiltration of immune cell types into 19 GIST tissues was analyzed by immunohistochemistry, and expression of T cell immunoglobulin and mucin protein 3 (Tim-3) and programmed cell death-1 (PD-1) in the infiltrated immune cells was examined by immunofluorescence microscopy. The expression status of galectin-9 in the GIST tumor cells was also determined by immunohistochemistry. All the GIST tissues showed CD8+ T cell infiltration and 8 showed CD56+ natural killer (NK) cell infiltration, and the numbers of infiltrated CD8+ T and NK cells were strongly correlated. However, these CD8+ T and NK cells were CD69-negative inactivated cells. Tim-3 was expressed in the infiltrated NK cells in 6/8 (75%) of the GIST tissues. Expression of galectin-9, a ligand of Tim-3, was observed in 13/19 (68.4%) GIST tissues and all of the GIST tissues with Tim-3+ NK cell infiltration showed positive galectin-9 expression. No PD-1 expression in the infiltrated NK cells and neither Tim-3 nor PD-1 expression was observed in the infiltrated CD8+ T cells. Interaction between Tim-3 in infiltrated NK cells and galectin-9 in tumor cells may be involved in an immune checkpoint mechanism for suppression of antitumor immunity in GISTs. Blockade of the Tim-3/galectin-9 pathway may become a new strategy for GIST treatment.


Asunto(s)
Galectinas/metabolismo , Tumores del Estroma Gastrointestinal/inmunología , Células Asesinas Naturales/inmunología , Proteínas de la Membrana/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Antígeno CD56/metabolismo , Linfocitos T CD8-positivos/inmunología , Femenino , Tumores del Estroma Gastrointestinal/metabolismo , Receptor 2 Celular del Virus de la Hepatitis A , Humanos , Lectinas Tipo C/metabolismo , Masculino , Persona de Mediana Edad
12.
Oncol Rep ; 34(1): 504-10, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25976081

RESUMEN

Trastuzumab emtansine (T-DM1), trastuzumab-conjugated with a cytotoxic agent, has shown promising antitumor effects in breast cancer. Since a good therapeutic response using T-DM1 treatment requires high human epidermal growth factor receptor 2 (HER2) expression, breast cancers with low or no HER2 expression have not been used for T-DM1 treatment. The aim of the present study was to show that treatment of low HER2-expressing breast cancer cells with gemcitabine (GEM) enhanced HER2 expression using RT-qPCR, immunoblot and flow cytometric analysis. The results showed that GEM treatment significantly enhanced HER2 expression in MDA-MB-231, MCF7 and BT-20 breast cancer cells, while paclitaxel (PTX) treatment induced lower or no enhancement in HER2 expression. The expression of HER2 mRNA was also enhanced in GEM-treated MCF7 cells. Treatment with an inhibitor for nuclear factor-(NF)-κB suppressed GEM-induced HER2 upregulation, indicating that NF-κB activation by GEM may be associated with HER2 upregulation. T-DM1 binding to HER2 on MCF-7 cells was enhanced by GEM pretreatment and the combined treatment of GEM and T-DM1 synergistically inhibited the proliferation of MCF7 cells. Thus, the combined treatment with GEM and T-DM1 may be a promising therapeutic modality for low HER2-expressing breast cancers, which was facilitated by the unique HER2-upregulating effect of GEM.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Desoxicitidina/análogos & derivados , Maitansina/análogos & derivados , Receptor ErbB-2/genética , Regulación hacia Arriba , Ado-Trastuzumab Emtansina , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Desoxicitidina/farmacología , Sinergismo Farmacológico , Femenino , Humanos , Células MCF-7 , Maitansina/farmacología , Paclitaxel/farmacología , Receptor ErbB-2/metabolismo , Trastuzumab , Gemcitabina
13.
Int J Oncol ; 46(6): 2679, 2015 06.
Artículo en Inglés | MEDLINE | ID: mdl-25846893

RESUMEN

Some errors in Fig. 3B and Fig. 6C have been identified. The errors do not change the conclusion of the paper. The conclusion is supported by other figures in the paper, as well as results described in the text. The corrected Fig. 3B and Fig. 6C are shown below. [the original article was published in the International Journal of Oncology 45: 470-478, 2014 DOI: 10.3892/ijo.2014.2433]

14.
Anticancer Res ; 35(1): 555-62, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25550602

RESUMEN

BACKGROUND/AIM: Treatment combining dendritic cells (DCs) pulsed with three types of major histocompatibility complex (MHC) class I and II (DC/WT1-I/II)-restricted Wilms' tumor 1 (WT1) peptides with chemotherapy may stabilize disease in pancreatic cancer patients. MATERIALS AND METHODS: Laboratory data from seven patients with pancreatic cancer who underwent combined DC/WT1-I/II vaccination and chemotherapy were analyzed. The DC phenotypes and plasma cytokine profiles were analyzed via flow cytometry. RESULTS: The post-treatment neutrophil to lymphocyte (N/L) ratio was a treatment-related prognostic factor for better survival. Moreover, the mean fluorescence intensities (MFIs) of human leukocyte antigen (HLA)-DR and cluster of differentiation (CD)83 on DCs were significantly increased after chemoimmunotherapy. Interestingly, interleukin (IL)-6 level in plasma was significantly increased after chemoimmunotherapy in non-super-responders. CONCLUSION: An increased N/L ratio, as well as HLA-DR and CD83 MFI levels may be prognostic markers of longer survival in patients with advanced pancreatic cancer who undergo chemoimmunotherapy.


Asunto(s)
Adenocarcinoma/terapia , Vacunas contra el Cáncer/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Neoplasias Pancreáticas/terapia , Proteínas WT1/inmunología , Adenocarcinoma/inmunología , Adenocarcinoma/mortalidad , Adenocarcinoma/secundario , Adulto , Anciano , Antimetabolitos Antineoplásicos/uso terapéutico , Biomarcadores de Tumor , Vacunas contra el Cáncer/administración & dosificación , Células Cultivadas , Terapia Combinada , Citocinas/sangre , Células Dendríticas/inmunología , Células Dendríticas/trasplante , Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapéutico , Femenino , Humanos , Recuento de Linfocitos , Masculino , Persona de Mediana Edad , Neutrófilos/inmunología , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Fragmentos de Péptidos/inmunología , Pronóstico , Resultado del Tratamiento , Vacunación , Gemcitabina
15.
Anticancer Res ; 34(11): 6353-61, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25368235

RESUMEN

BACKGROUND/AIM: Chemoimmunotherapy has been used to treat intrahepatic cholangiocarcinoma (ICC). However, little is known about the phenomena underlying the immunomodulation of ICC cells elicited by chemoimmunotherapy. MATERIALS AND METHODS: Primary ICC cells from a patient with ICC who received gemcitabine followed by 5-fluorouracil (5-FU), both combined with dendritic cells pulsed with Wilms' tumor 1 (WT1) peptides were cultured. ICC cells were treated with gemcitabine, 5-FU or interferon (IFN)-γ in vitro. The phenotype of the ICC cells was examined by flow cytometry and quantitative reverse transcription polymerase chain reaction. RESULTS: Stimulation of the ICC cells with gemcitabine resulted in up-regulation of WT1 mRNA, programmed death receptor ligand-1 (PDL1) and calreticulin. Gemcitabine, 5-FU and IFN-γ induced up-regulation of mucin-1. Moreover, human leukocyte antigen (HLA)-ABC, HLA-DR and PDL1 were extremely up-regulated by IFN-γ. CONCLUSION: Chemoimmunomodulating agents alter the immunogenicity of ICC cells, resulting in complex clinical efficacy results.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias de los Conductos Biliares/terapia , Conductos Biliares Intrahepáticos/inmunología , Colangiocarcinoma/terapia , Células Dendríticas/inmunología , Inmunoterapia , Antígenos de Neoplasias/inmunología , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Neoplasias de los Conductos Biliares/inmunología , Neoplasias de los Conductos Biliares/patología , Conductos Biliares Intrahepáticos/efectos de los fármacos , Conductos Biliares Intrahepáticos/patología , Colangiocarcinoma/inmunología , Colangiocarcinoma/patología , Terapia Combinada , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Femenino , Fluorouracilo/administración & dosificación , Antígenos HLA-DR/genética , Antígenos HLA-DR/metabolismo , Humanos , Interferón gamma/administración & dosificación , Metástasis Linfática , Persona de Mediana Edad , Mucina-1/genética , Mucina-1/metabolismo , Neoplasias Peritoneales/inmunología , Neoplasias Peritoneales/secundario , Neoplasias Peritoneales/terapia , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Proteínas WT1/genética , Proteínas WT1/metabolismo , Gemcitabina
16.
Anticancer Res ; 34(9): 4869-76, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25202068

RESUMEN

BACKGROUND: In murine studies, cryotherapy has induced antitumor immune responses associated with the rejection of tumors. However, the effects of freezing-induced immunomodulation in breast cancer (BC) patients remain unclear. MATERIALS AND METHODS: Ten BC patients were prospectively divided into two groups: 1) cryotherapy followed by surgical excision and 2) surgical excision-alone. The cytokine profiles of plasma and peripheral blood mononuclear cells (PBMCs) were analyzed using flow cytometry following in vitro stimulation with the 30-mer MUC1 peptide. RESULTS: No differences in the percentages of interferon-γ (IFN-γ)-producing cluster of differentiation (CD)4(+) or CD8(+) T cells and the plasma levels of IFN-γ, interleukin-1ß (IL-1ß), IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, IL-12p70, tumor necrosis factor-α (TNF-α) and TNF-ß were observed between these 2 groups, and PBMCs were not significantly altered. CONCLUSION: Alternations to the type 1 and 2 helper cytokine profiles were not detected in vitro in BC patients treated with cryotherapy-alone.


Asunto(s)
Neoplasias de la Mama/inmunología , Neoplasias de la Mama/terapia , Crioterapia , Inmunomodulación , Adulto , Anciano , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/cirugía , Citocinas/biosíntesis , Femenino , Humanos , Persona de Mediana Edad , Mucina-1/inmunología , Mucina-1/metabolismo , Estudios Prospectivos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
17.
Clin Cancer Res ; 20(16): 4228-39, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25056373

RESUMEN

PURPOSE: We performed a phase I trial to investigate the safety, clinical responses, and Wilms' tumor 1 (WT1)-specific immune responses following treatment with dendritic cells (DC) pulsed with a mixture of three types of WT1 peptides, including both MHC class I and II-restricted epitopes, in combination with chemotherapy. EXPERIMENTAL DESIGN: Ten stage IV patients with pancreatic ductal adenocarcinoma (PDA) and 1 patient with intrahepatic cholangiocarcinoma (ICC) who were HLA-positive for A*02:01, A*02:06, A*24:02, DRB1*04:05, DRB1*08:03, DRB1*15:01, DRB1*15:02, DPB1*05:01, or DPB1*09:01 were enrolled. The patients received one course of gemcitabine followed by biweekly intradermal vaccinations with mature DCs pulsed with MHC class I (DC/WT1-I; 2 PDA and 1 ICC), II (DC/WT1-II; 1 PDA), or I/II-restricted WT1 peptides (DC/WT1-I/II; 7 PDA), and gemcitabine. RESULTS: The combination therapy was well tolerated. WT1-specific IFNγ-producing CD4(+) T cells were significantly increased following treatment with DC/WT1-I/II. WT1 peptide-specific delayed-type hypersensitivity (DTH) was detected in 4 of the 7 patients with PDA vaccinated with DC/WT1-I/II and in 0 of the 3 patients with PDA vaccinated with DC/WT1-I or DC/WT1-II. The WT1-specific DTH-positive patients showed significantly improved overall survival (OS) and progression-free survival (PFS) compared with the negative control patients. In particular, all 3 patients with PDA with strong DTH reactions had a median OS of 717 days. CONCLUSIONS: The activation of WT1-specific immune responses by DC/WT1-I/II combined with chemotherapy may be associated with disease stability in advanced pancreatic cancer.


Asunto(s)
Células Dendríticas/inmunología , Desoxicitidina/análogos & derivados , Epítopos/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Neoplasias Pancreáticas/terapia , Proteínas WT1/inmunología , Adenocarcinoma/inmunología , Adenocarcinoma/mortalidad , Adenocarcinoma/secundario , Adenocarcinoma/terapia , Adulto , Anciano , Antimetabolitos Antineoplásicos/uso terapéutico , Neoplasias de los Conductos Biliares/inmunología , Neoplasias de los Conductos Biliares/mortalidad , Neoplasias de los Conductos Biliares/secundario , Neoplasias de los Conductos Biliares/terapia , Conductos Biliares Intrahepáticos/inmunología , Biomarcadores de Tumor/análisis , Linfocitos T CD8-positivos/inmunología , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/mortalidad , Carcinoma Ductal Pancreático/secundario , Carcinoma Ductal Pancreático/terapia , Colangiocarcinoma/inmunología , Colangiocarcinoma/mortalidad , Colangiocarcinoma/secundario , Colangiocarcinoma/terapia , Terapia Combinada , Desoxicitidina/uso terapéutico , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Fragmentos de Péptidos/inmunología , Pronóstico , Tasa de Supervivencia , Linfocitos T Citotóxicos/inmunología , Vacunación , Gemcitabina
18.
Int J Oncol ; 45(1): 470-8, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24819411

RESUMEN

Previous work has demonstrated that fusion cells generated from autologous monocyte-derived dendritic cells (MoDCs) and whole tumor cells induce efficient antigen-specific cytotoxic T lymphocytes. A major limitation to the use of this strategy is the availability of adequate amounts of autologous tumor cells. Moreover, MoDCs from cancer patients are often defective in their antigen-processing and presentation machinery. In this study, two types of allogeneic cells, a leukemia plasmacytoid dendritic cell (pDC) line (PMDC05) and pancreatic cancer cell lines (PANC-1 or MIA PaCa-2), were fused instead of autologous MoDCs and tumor cells. We created four types of pDC/tumor fusion cells by alternating fusion partners and treating with lipopolysaccharide (LPS): i) PMDC05 fused with PANC-1 (pDC/PANC-1), ii) PMDC05 fused with MIA PaCa-2 (pDC/MIA PaCa-2), iii) LPS-stimulated pDC/PANC-1 (LPS-pDC/PANC-1) and iv) LPS-stimulated pDC/MIA PaCa-2 (LPS-pDC/MIA PaCa-2) and examined their antitumor immune responses. The LPS-pDC/tumor cell fusions were the most active, as demonstrated by their: i) upregulated expression of HLA-DR and CD86 on a per-fusion-cell basis, ii) increased production of IL-12p70, iii) generation of a higher percentage of IFN-γ-producing CD4⁺ and CD8⁺ T cells and iv) augmented induction of MUC1-specific CD8⁺ T cells that lyse target tumor cells. This study provides the first evidence for an in vitro induction of antigen-specific cytotoxic T lymphocytes by LPS-stimulated fusion cells generated from leukemia plasmacytoid DCs and tumor cells and suggests that this strategy has potential applicability to the field of adoptive immunotherapy.


Asunto(s)
Células Dendríticas/metabolismo , Células Híbridas/metabolismo , Leucemia/inmunología , Mucina-1/metabolismo , Neoplasias Pancreáticas/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Fusión Celular , Línea Celular Tumoral , Citotoxicidad Inmunológica , Células Dendríticas/inmunología , Regulación de la Expresión Génica , Humanos , Células Híbridas/inmunología , Células Híbridas/patología , Leucemia/metabolismo , Lipopolisacáridos/farmacología , Neoplasias Pancreáticas/metabolismo
19.
Cancer Immunol Immunother ; 63(5): 459-68, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24627093

RESUMEN

Vaccination of BALB/c mice with dendritic cells (DCs) loaded with the lysate of induced vascular progenitor (iVP) cells derived from murine-induced pluripotent stem (iPS) cells significantly suppressed the tumor of CMS-4 fibrosarcomas and prolonged the survival of CMS-4-inoculated mice. This prophylactic antitumor activity was more potent than that of immunization with DCs loaded with iPS cells or CMS-4 tumor cells. Tumors developed slowly in mice vaccinated with DCs loaded with iVP cells (DC/iVP) and exhibited a limited vascular bed. Immunohistochemistry and a tomato-lectin perfusion study demonstrated that the tumors that developed in the iVP-immunized mice showed a marked decrease in tumor vasculature. Immunization with DC/iVP induced a potent suppressive effect on vascular-rich CMS-4 tumors, a weaker effect on BNL tumors with moderate vasculature, and nearly no effect on C26 tumors with poor vasculature. Treatment of DC/iVP-immunized mice with a monoclonal antibody against CD4 or CD8, but not anti-asialo GM1, inhibited the antitumor activity. CD8(+) T cells from DC/iVP-vaccinated mice showed significant cytotoxic activity against murine endothelial cells and CMS-4 cells, whereas CD8(+) T cells from DC/iPS-vaccinated mice did not. DNA microarray analysis showed that the products of 29 vasculature-associated genes shared between genes upregulated by differentiation from iPS cells into iVP cells and genes shared by iVP cells and isolated Flk-1(+) vascular cells in CMS-4 tumor tissue might be possible targets in the immune response. These results suggest that iVP cells from iPS cells could be used as a cancer vaccine targeting tumor vascular cells and tumor cells.


Asunto(s)
Vacunas contra el Cáncer/farmacología , Células Dendríticas/inmunología , Células Madre Pluripotentes Inducidas/inmunología , Neoplasias Experimentales/terapia , Vacunación/métodos , Animales , Fibrosarcoma/terapia , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Linfocitos T Citotóxicos/inmunología
20.
Oncoimmunology ; 2(9): e25994, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24228229

RESUMEN

The rationale for fusing dendritic cells (DCs) with whole tumor cells to generate anticancer vaccines resides in the fact that the former operate as potent antigen-presenting cells, whereas the latter express a constellation of tumor-associated antigens (TAAs). Although the administration of DC/malignant cell fusions to cancer patients is safe and this immunotherapeutic intervention triggers efficient tumor-specific T-cell responses in vitro, a limited number of objective clinical responses to DC/cancer cell fusions has been reported thus far. This review discusses novel approaches to improve the immunogenicity of DC/malignant cell fusions as anticancer vaccines.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA