Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters











Publication year range
1.
Photosynth Res ; 149(1-2): 69-82, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33817762

ABSTRACT

When intact green leaves are exposed to the fluctuating light, in which high light (HL) and low light (LL) alternate, photosystem I (PSI) is readily damaged. This PSI inhibition is mostly alleviated by the addition of far-red (FR) light. Here, we grew Alocasia odora, a shade-tolerant species, at several light levels and examined their photosynthetic traits in relation to the fluctuating light-induced PSI inhibition. We found that, even in the absence of FR, PSI in LL-grown leaves was resistant to the fluctuating light. LL leaves showed higher chlorophyll (Chl) contents on leaf area basis, lower Chl a/b ratios, lower cytochrome f/P700 ratios, and lower PSII/PSI excitation ratios assessed by the 77 K fluorescence. Also, P700 in the HL phase of the fluctuating light was more oxidized. The results of the regression analyses of the PSI photoinhibition to these traits indicate that the lower electron flow rate to P700 and more excitation energy transfer to PSI protect PSI in LL-grown leaves. Both of these contribute oxidization of P700 to the efficient quencher form P700+. These features may be common in LL-grown shade-tolerant species, which are often exposed to strong sunflecks in their natural habitats.


Subject(s)
Adaptation, Ocular/physiology , Alocasia/metabolism , Arabidopsis/metabolism , Cytochromes f/metabolism , Photosystem I Protein Complex/metabolism , Plant Leaves/metabolism , Sunlight/adverse effects
2.
Kurume Med J ; 66(1): 43-47, 2020 Jul 01.
Article in English | MEDLINE | ID: mdl-32378536

ABSTRACT

This multicenter phase II N-DOCC-F-C-1701 trial is being planned in order to investigate the efficacy and safety of CPT-11+S-1 +Ramucirumab (IRIS+Rmab), which is anticipated to have a stronger anti-tumor effect than IRIS+Bmab in patients with metastatic colorectal cancer (mCRC) previously treated with oxaliplatin (L-OHP) containing regimen, in consideration of the result of RAISE, FIRIS and some phase II trials of IRIS+Bevacicizumab (Bmab). The number of patients is set at 38 for the statistical analysis, assuming an expected median PFS of 5.0 months (threshold: 3.0 months). The primary endpoint of the study is the progression free survival (PFS), and the secondary endpoints are the overall response rate (ORR), overall survival (OS), adverse events (AE), quality of life (QOL) and review of nausea and vomiting. This trial is registered in the UMIN Clinical Trials Registry as UMIN000028170. We intend to start conducting the trial in September 1, 2017. If this trial meets the endpoint, IRIS+Rmab might be supported as a new optional standard regimen for mCRC.


Subject(s)
Antibodies, Monoclonal, Humanized , Colorectal Neoplasms , Oxaliplatin , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm , Humans , Irinotecan/therapeutic use , Oxaliplatin/pharmacology , Oxaliplatin/therapeutic use , Quality of Life , Thiazoles , Ramucirumab
3.
Anticancer Res ; 39(9): 4699-4709, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31519569

ABSTRACT

BACKGROUND/AIM: Metformin, a drug for type 2 diabetes, also exerts anticancer effects. This study addressed the immunological effects of metformin on peritoneal dissemination. MATERIALS AND METHODS: We developed a mouse model of peritoneal dissemination via intraperitoneal injection of RLmale1, an X-ray-induced leukemia cell line, into BALB/c mice. Cell-surface markers, cytokine production, and myeloid-derived suppressor cells (MDSCs) were examined in cells from spleen and peritoneal lavage fluid. RESULTS: Metformin-treated mice exhibited suppressed intraperitoneal tumor growth and extended survival, and these effects were lost in mice with severe combined immunodeficiency. MDSCs induction was inhibited in metformin-treated mice. Although MDSC mobilization into the peritoneal cavity was correlated with suppression of interferon-γ production by tumor-infiltrating lymphocytes, the T-helper 1 ability of these lymphocytes was preserved in metformin-treated mice. CONCLUSION: Our findings demonstrate the action of metformin on both intraperitoneal tumors and immune-suppressive cells and might contribute to the development of immunotherapy against peritoneal dissemination.


Subject(s)
Immunomodulation/drug effects , Metformin/pharmacology , Peritoneal Neoplasms/immunology , Peritoneal Neoplasms/pathology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Animals , Biomarkers , Cell Line, Tumor , Disease Models, Animal , Humans , Immunophenotyping , Male , Mice , Myeloid-Derived Suppressor Cells/drug effects , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Neoplasm Metastasis , Peritoneal Neoplasms/drug therapy , Peritoneal Neoplasms/mortality , Proportional Hazards Models , Xenograft Model Antitumor Assays
4.
Case Rep Gastroenterol ; 8(2): 175-81, 2014 May.
Article in English | MEDLINE | ID: mdl-24932165

ABSTRACT

Ehlers-Danlos syndrome, vascular type (vEDS) (MIM #130050) is an autosomal dominant disorder caused by mutation in the type III collagen gene, COL3A1, leading to fragility of blood vessels, bowel and uterus that leads to spontaneous rupture. We report a previously undiagnosed vEDS patient with bowel complications. A 20-year-old female patient was referred to our hospital with abdominal pain. Computed tomography showed notable dilatation of the sigmoid colon with intraperitoneal fluid. Laparotomy revealed dilatation of the sigmoid colon, breakdown of serosa and muscularis propria of the sigmoid colon with impending perforation, and intra-abdominal hemorrhage caused by breakdown of the mesenterium. Resection of the sigmoid colon with Hartmann's pouch and an end colostomy were performed. Physical examination showed joint hypermobility, translucent skin with venous prominence and facial structure abnormalities. Genetic analysis using cDNA extracted from the patient's fibroblasts by reverse transcriptase polymerase chain reaction direct sequencing showed a missense mutation within the triple helix region of COL3A1 (c.2150 G>A; Gly717Asp).

5.
Nutrition ; 29(10): 1224-30, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23867209

ABSTRACT

OBJECTIVE: Enteral administration of synbiotics has been reported to be beneficial during various types of surgery, but its clinical value in elderly surgical patients remains unclear. The aim of this study was to quantitatively evaluate the changes in gut microbiota and environment induced by perioperative synbiotic therapy, and to investigate whether it is possible to reduce infectious complications in elderly patients undergoing gastroenterological surgery. METHODS: Forty-eight patients over the age of 70 y were randomized into a group receiving 7 d of preoperative and 10 d of postoperative synbiotic therapy (S group) and a control group without synbiotic therapy (C group). A fecal sample collected before and after surgery in each group was used for a quantitative evaluation of the microbiota. RESULTS: Forty-eight patients completed the trial (25 in the S group and 23 in the C group). Synbiotic therapy significantly maintained the status of Bifidobacterium and Lactobacillus, whereas the number of Enterobacteriaceae, Staphylococcus, and Pseudomonas was significantly decreased. The total organic acid and short-chain fatty acid concentrations were increased, and the pH was markedly decreased, in the S group compared with the C group. The incidence of postoperative infectious complications was 12% in the S group and 36% in the C group, however, the difference did not reach statistical significance (P = 0.06). A multivariate analysis revealed that only the use of perioperative blood transfusion was an independent risk factor for infectious complications. CONCLUSIONS: Synbiotic therapy improved the intestinal microbial environment, and might decrease the incidence of infectious complications in elderly surgical patients.


Subject(s)
Endoscopy, Gastrointestinal/methods , Gastrointestinal Tract/microbiology , Perioperative Care/methods , Postoperative Complications/epidemiology , Synbiotics , Aged , Aged, 80 and over , Bifidobacterium , Enterobacteriaceae , Feces/microbiology , Female , Humans , Hydrogen-Ion Concentration , Incidence , Lactobacillus , Logistic Models , Male , Multivariate Analysis , Postoperative Complications/prevention & control , Prospective Studies , Pseudomonas , Staphylococcus , Treatment Outcome
6.
Anticancer Res ; 32(11): 4897-904, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23155258

ABSTRACT

BACKGROUND/AIM: The cross-presentation system of tumor antigen by monocyte-derived dendritic cells (mo-DCs) has been observed under appropriate conditions. Both CD14-negative and CD1a-positive phenotypes were critical in our previous study. This study compared the phenotype of mo-DCs and identified the conditions that favored T helper-1 (Th1) cytokine production after stimulation with the hsc70 and NY-ESO-1 p157-165 epitope fusion protein (hsc70/ESO p157-165). MATERIALS AND METHODS: The mo-DCs were induced from healthy donors. Their surface markers and cytokine production were examined after stimulation with hsc70/ESO p157-165. RESULTS: CD1a(+) and CD1a(-) mo-DCs were generated in half of the healthy donors. The concentration of fetal calf serum in the culture medium was critical for the induction of CD1a(+) DCs, which were able to produce interleukin-12 (IL-12), but not IL-10. Neutralizing IL-6 and IL-6R antibodies affected the expression of CD1a. CONCLUSION: Anti IL-6 analogs may be effective adjuvants for the development of mo-DC-based cancer vaccine.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Cross-Priming/immunology , Dendritic Cells/immunology , HSC70 Heat-Shock Proteins/immunology , Adjuvants, Immunologic/pharmacology , Dendritic Cells/cytology , Dendritic Cells/metabolism , Flow Cytometry , Humans , Interleukin-6/immunology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lymphocyte Activation/immunology , Neoplasm Proteins/immunology , Peptide Fragments/immunology , Phenotype
7.
World J Gastroenterol ; 18(13): 1470-8, 2012 Apr 07.
Article in English | MEDLINE | ID: mdl-22509078

ABSTRACT

AIM: To investigate the immunological repertoire in the peritoneal cavity of gastric cancer patients. METHODS: The peritoneal cavity is a compartment in which immunological host-tumor interactions can occur. However, the role of lymphocytes in the peritoneal cavity of gastric cancer patients is unclear. We observed 64 patients who underwent gastrectomy for gastric cancer and 11 patients who underwent laparoscopic cholecystectomy for gallstones and acted as controls. Lymphocytes isolated from both peripheral blood and peritoneal lavage were analyzed for surface markers of lymphocytes and their cytokine production by flow cytometry. CD4(+)CD25(high) T cells isolated from the patient's peripheral blood were co-cultivated for 4 d with the intra-peritoneal lymphocytes, and a cytokine assay was performed. RESULTS: At gastrectomy, CCR7(-) CD45RA(-) CD8(+) effector memory T cells were observed in the peritoneal cavity. The frequency of CD4(+) CD25 (high) T cells in both the peripheral blood and peritoneal cavity was elevated in patients at advanced stage [control vs stage IV in the peripheral blood: 6.89 (3.39-10.4) vs 15.34 (11.37-19.31), P < 0.05, control vs stage IV in the peritoneal cavity: 8.65 (5.28-12.0) vs 19.56 (14.81-24.32), P < 0.05]. On the other hand, the suppression was restored with CD4(+) CD25(high) T cells from their own peripheral blood. This study is the first to analyze lymphocyte and cytokine production in the peritoneal cavity in patients with gastric cancer. Immune regulation at advanced stage is reversible at the point of gastrectomy. CONCLUSION: The immunological milieu in the peritoneal cavity of patients with advanced gastric cancer elicited a Th2 response even at gastrectomy, but this response was reversible.


Subject(s)
Laparotomy , Peritoneal Cavity/surgery , Stomach Neoplasms/immunology , Stomach Neoplasms/surgery , Aged , Animals , Cells, Cultured , Coculture Techniques , Cytokines/immunology , Female , Flow Cytometry/methods , Gastrectomy , Humans , Leukocytes, Mononuclear/cytology , Lymphocytes/cytology , Lymphocytes/immunology , Male , Middle Aged , Peritoneal Cavity/pathology , Peritoneal Lavage , Stomach Neoplasms/pathology , T-Lymphocytes/immunology , Th2 Cells/immunology
8.
Gan To Kagaku Ryoho ; 38(10): 1699-703, 2011 Oct.
Article in Japanese | MEDLINE | ID: mdl-21996971

ABSTRACT

We describe four cases of locally advanced colorectal cancer resected successfully after preoperative chemotherapy conducted between April of 2007 and April of 2009. The average age of the patients was 66.3 years (range, 40-77 years). Because of tumor invasion into the surrounding organs, preoperative chemotherapy with FOLFOX4 was performed. The average number of courses of chemotherapy was 5.2 (range, 4-7). After chemotherapy, we were able to perform radical operations for all four cases. Histopathological examination of the tumor revealed Grade 3 in one case. There were no postoperative complications and no recurrences in any of the cases. We performed curative surgery after chemotherapy, and good results were obtained. Preoperative chemotherapy may be effective for avoiding excessive intervention surgeries such as total pelvic exenteration, preserving bladder and rectal functions, and for maintening QOL.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Adult , Aged , Colorectal Neoplasms/pathology , Colorectal Neoplasms/surgery , Combined Modality Therapy , Fluorouracil/therapeutic use , Humans , Leucovorin/therapeutic use , Male , Neoplasm Staging , Organoplatinum Compounds/therapeutic use , Tomography, X-Ray Computed
9.
Surg Laparosc Endosc Percutan Tech ; 20(1): e38-41, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20173609

ABSTRACT

INTRODUCTION: Many techniques have been described for the surgical repair of lumbar hernias, including primary repair, local tissue flaps, and conventional mesh repair. All these open techniques require a large incision plus extensive dissection to expose the hernia ring. This report presents a case of a recurrent lumbar hernia, which was successfully repaired using a laparoscopic approach. CASE REPORT: A 75-year-old female presented with a symptomatic right lumbar hernia, 1-year after an iliac bone harvest for knee surgery. Under general anesthesia, the patient was placed in a lateral decubitus position. A 3 trocar technique was used to do adhesiolysis of the surrounding tissues, to provide an ample working space to identify the hernia. A composix dual mesh (bard) was tailored so that it would overlap the defect with intermittent fixation by a spiral tacker (protac). No hernia recurrence occurred over 2 years after surgery. CONCLUSION: The laparoscopic approach has significant advantages for the repair a lumbar hernia: it enables the exact localization of the anatomic defect, and the mesh can be placed deep into the defect, thus allowing the intraabdominal pressure to hold it in position.


Subject(s)
Hernia, Abdominal/surgery , Laparoscopy/methods , Lumbosacral Region/surgery , Aged , Female , Humans , Recurrence
10.
Cancer Sci ; 99(1): 107-12, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17991294

ABSTRACT

The cancer-testis antigen NY-ESO-1 has been implicated as one of the most attractive candidates for a cancer vaccine. However, a protein vaccine generally meets inefficient antigen presentation to CD8(+) T cells, which could be overcome by combination with an appropriate adjuvant. Heat shock protein is a natural adjuvant and activates the antigen-presenting cells to channel exogenous antigens into the classical major histocompatibility complex class I antigen-processing pathway (cross-presentation). Therefore, we genetically fused a minigene encompassing the NY-ESO-1 cytotoxic T lymphocyte (CTL) epitope 157-165 (ESO p157-165) to the human heat shock cognate protein 70 (hsc70) and expressed the resulting fusion proteins in Escherichia coli. By using a human leukocyte antigen-A*0201-restricted NY-ESO-1-specific CTL clone, the cross-presentation of ESO p157-165 by monocyte-derived dendritic cells (mo-DC) pulsed with the fusion protein was evaluated. The fusion protein-pulsed mo-DC activates the CTL clone much more efficiently than the free NY-ESO-1 protein-pulsed mo-DC. Moreover, the magnitude of the CTL activity was comparable between ESO p157-165 and the fusion protein of hsc70 and ESO p157-165 (hsc70-ESO p157-165 fusion protein). In addition, the CTL activation induced by the fusion protein, but not by the epitope, was inhibited by paraformaldehyde fixation of the mo-DC and by treatment with lactacystin, a specific inhibitor for the proteasome. Finally, the hsc70-ESO p157-165 fusion protein-pulsed DC was able to induce an antigen-specific T-cell response. These results suggest that the hsc70-ESO p157-165 fusion protein is therefore considered to be a promising candidate as a cancer vaccine.


Subject(s)
Antigens, Neoplasm/immunology , Dendritic Cells/immunology , Epitopes, T-Lymphocyte/immunology , HSP70 Heat-Shock Proteins/immunology , Membrane Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Antigen Presentation , Antigens, Neoplasm/genetics , Cross Reactions , Epitopes, T-Lymphocyte/genetics , HLA-A2 Antigen/immunology , HSP70 Heat-Shock Proteins/genetics , Humans , Lymphocyte Activation , Membrane Proteins/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology
11.
J Gastroenterol ; 42(1): 91-5, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17323000

ABSTRACT

Although stomal varices are a rare complication, bleeding stomal varices often need to be treated owing to symptoms of hypovolemic shock, recurrence of stomal bleeding, or deterioration in the quality of life. Various treatment strategies for the management of bleeding stomal varices have thus far been reported. We report the case of a 60-year-old woman with refractory recurrent bleeding from varices in a sigmoid stoma, along with nonalcoholic steatohepatitis and marked splenomegaly. A physical examination revealed that the skin was discolored and bluish around the circumference of the sigmoid stoma. The venous phase of a celiac arteriogram revealed an afferent vein from the splenic vein and another from the inferior mesenteric vein, and veins draining into the left superficial epigastric vein. A balloon-occluded retrograde transvenous obliteration (BRTO) procedure was performed. The skin around the stoma, initially discolored bluish, improved markedly. After 10 months of follow-up, the patient has remained well without further episodes of stomal bleeding. To our knowledge, this is the first case of recurrent hemorrhage from stomal varices that was successfully treated by BRTO in a patient with portal hypertension.


Subject(s)
Balloon Occlusion , Colostomy , Esophageal and Gastric Varices/therapy , Gastrointestinal Hemorrhage/therapy , Postoperative Complications/therapy , Balloon Occlusion/methods , Colon, Sigmoid/surgery , Esophageal and Gastric Varices/diagnostic imaging , Female , Humans , Middle Aged , Recurrence , Tomography, X-Ray Computed
12.
J Clin Invest ; 116(7): 1946-54, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16794737

ABSTRACT

Bacterial vectors may offer many advantages over other antigen delivery systems for cancer vaccines. We engineered a Salmonella typhimurium vaccine strain to deliver the NY-ESO-1 tumor antigen (S. typhimurium-NY-ESO-1) through a type III protein secretion system. The S. typhimurium-NY-ESO-1 construct elicited NY-ESO-1-specific CD8+ and CD4+ T cells from peripheral blood lymphocytes of cancer patients in vitro. Oral administration of S. typhimurium-NY-ESO-1 to mice resulted in the regression of established NY-ESO-1-expressing tumors. Intratumoral inoculation of S. typhimurium-NY-ESO-1 to NY-ESO-1-negative tumors resulted in delivery of antigen in vivo and led to tumor regression in the presence of preexisting NY-ESO-1-specific CD8+ T cells. Specific T cell responses against at least 2 unrelated tumor antigens not contained in the vaccine were observed, demonstrating epitope spreading. We propose that antigen delivery through the S. typhimurium type III secretion system is a promising novel strategy for cancer vaccine development.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines , Membrane Proteins/immunology , Salmonella typhimurium/metabolism , Animals , Antibodies, Neoplasm/immunology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/therapeutic use , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Epitopes , Female , Humans , Membrane Proteins/genetics , Membrane Proteins/therapeutic use , Mice , Mice, Inbred BALB C , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use , Salmonella typhimurium/genetics
14.
Eur J Immunol ; 34(10): 2919-29, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15368308

ABSTRACT

NY-ESO-1 is a germ cell antigen aberrantly expressed by different tumor types that elicits strong humoral and cellular immune responses, representing one of the most promising candidates for vaccination of cancer patients. A detailed analysis of CD8(+) T cells generated in vaccine trials using NY-ESO-1-derived peptides (157-165 and 157-167) revealed that the dominant immune response was directed against a cryptic epitope (159-167) diverting the immune response from tumor recognition. Only CTL reactivity to the NY-ESO-1(157-165) peptide appeared to be capable of lysing NY-ESO-1/HLA-A0201-expressing tumor cells. To study the process of NY-ESO-1 peptide presentation by tumor cells in more detail we generated a high-affinity (K(D)=60 nM) antibody fragment that specifically recognizes the NY-ESO-1(157-165) peptide/HLA-A0201 complex. Peptide variants such as the NY-ESO-1(157-167) peptide or the cryptic NY-ESO-1(159-167) peptide were not recognized. The antibody fragment blocked in a dose-dependent fashion the recognition of NY-ESO-1/HLA-A2-positive tumor cells by NY-ESO-1(157-165) peptide-specific CD8(+) T cells. This antibody fragment is a novel reagent that binds with TCR-like specificity to the NY-ESO-1(157-165)/HLA-A2 complex thus distinguishing between CTL responses against immunological meaningful or cryptic NY-ESO-1-derived peptides. It may therefore become a useful monitoring tool for the development of NY-ESO-1-based cancer vaccines.


Subject(s)
Antibody Specificity/immunology , Antigens, Neoplasm/immunology , Immunoglobulin Fab Fragments/immunology , Major Histocompatibility Complex/immunology , Membrane Proteins/immunology , Peptides/immunology , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Sequence , Antigen Presentation/immunology , Cancer Vaccines/immunology , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunodominant Epitopes/immunology , Protein Conformation , Transfection
15.
Proc Natl Acad Sci U S A ; 101(40): 14467-72, 2004 Oct 05.
Article in English | MEDLINE | ID: mdl-15383663

ABSTRACT

We compared monocyte-derived dendritic cells and transforming growth factor-beta1-induced Langerhans-like cells (LCs) for their capacity to cross-present exogenous NY-ESO-1 protein/antibody immune complexes to an NY-ESO-1-specific CD8+ T cell clone. In contrast to dendritic cells, LCs were not able to cross-present NY-ESO-1 to the T cell clone constitutively but did so after treatment with IFN-gamma. Remarkably, this IFN-gamma-inducible characteristic was due neither to enhanced antigen uptake nor to facilitated antigen processing in LCs. Rather, IFN-gamma acted at least in part by potentiating the maturation of otherwise refractory LCs, enabling in turn exogenous antigen to reach the processing machinery. This model of conditional cross-presentation establishes an original level of action for IFN-gamma as an effective immune modulator and supports the use of IFN-gamma in protein vaccination strategies targeting LCs.


Subject(s)
Interferon-gamma/pharmacology , Langerhans Cells/drug effects , Langerhans Cells/immunology , Amino Acid Sequence , Antigen Presentation/drug effects , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cell Differentiation/drug effects , Cell Line , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/immunology , HLA-A Antigens/metabolism , HLA-A2 Antigen , Humans , Langerhans Cells/cytology , Membrane Proteins/genetics , Membrane Proteins/immunology , Molecular Sequence Data , Phenotype , Recombinant Proteins , Signal Transduction , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta1
16.
J Immunol ; 172(5): 3289-96, 2004 Mar 01.
Article in English | MEDLINE | ID: mdl-14978137

ABSTRACT

MAGE-3 is the most commonly expressed cancer testis Ag and thus represents a prime target for cancer vaccines, despite infrequent natural occurrence of MAGE-3-specific immune responses in vivo. We report in this study the successful induction of Ab, CD8(+), and CD4(+) T cells in nonsmall cell lung cancer patients vaccinated with MAGE-3 recombinant protein. Two cohorts were analyzed: one receiving MAGE-3 protein alone, and one receiving MAGE-3 protein with adjuvant AS02B. Of nine patients in the first cohort, three developed marginal Ab titers and another one had a CD8(+) T cell response to HLA-A2-restricted peptide MAGE-3 271-279. In contrast, of eight patients from the second cohort vaccinated with MAGE-3 protein and adjuvant, seven developed high-titered Abs to MAGE-3, and four had a strong concomitant CD4(+) T cell response to HLA-DP4-restricted peptide 243-258. One patient simultaneously developed CD8(+) T cells to HLA-A1-restricted peptide 168-176. The novel monitoring methodology used in this MAGE-3 study establishes that protein vaccination induces clear CD4(+) T cell responses that correlate with Ab production. This development provides the framework for further evaluating integrated immune responses in vaccine settings and for optimizing these responses for clinical benefit.


Subject(s)
Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cancer Vaccines/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Lipid A/analogs & derivatives , Lung Neoplasms/immunology , Neoplasm Proteins/immunology , Vaccines, DNA/immunology , Adjuvants, Immunologic/administration & dosage , Amino Acid Sequence , Antibodies, Neoplasm/biosynthesis , Antibodies, Neoplasm/blood , CD8-Positive T-Lymphocytes/immunology , Cytokines/biosynthesis , Epitopes, T-Lymphocyte/immunology , Humans , Lipid A/administration & dosage , Lipid A/immunology , Lymphocyte Activation/immunology , Melanoma/immunology , Molecular Sequence Data , Saponins/administration & dosage , Saponins/immunology , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Vaccines, Combined/administration & dosage , Vaccines, Combined/immunology , Vaccines, DNA/administration & dosage
17.
J Immunol Methods ; 278(1-2): 57-66, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12957396

ABSTRACT

CD4+ T cells play an important role in the induction and maintenance of an effective antiviral and antitumor immune response. However, standardized monitoring of antigen-specific CD4+ T cells has not been established at the single-cell level. We now present a sensitive, specific, and simple methodology in which purified memory CD4+ T cells are expanded from PBMC in a single cycle of antigen-driven stimulation and quantitatively assayed by interferon-gamma ELISPOT. Issues of nonspecific background in assays were resolved with the use of innovative target cells, autologous PHA-expanded CD4+ T cells (T-APC). Remarkably, T-APC could not only present peptide epitopes from model antigens NY-ESO-1 and influenza nucleoprotein, but could also process full-length antigen endogenously expressed from recombinant fowlpox vector. This approach makes it possible to monitor CD4+ T cells in large series of patients, regardless of HLA haplotype, against the full peptide repertoire of a given antigen.


Subject(s)
Antigen-Presenting Cells/immunology , CD4-Positive T-Lymphocytes/immunology , Immunoassay/methods , Lymphocyte Activation/immunology , T-Lymphocyte Subsets/immunology , Animals , Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Humans , Immunomagnetic Separation , Lung Neoplasms , Melanoma/immunology
18.
Proc Natl Acad Sci U S A ; 100(15): 8862-7, 2003 Jul 22.
Article in English | MEDLINE | ID: mdl-12853579

ABSTRACT

NY-ESO-1 is one of the most immunogenic proteins described in human cancers, based on its capacity to elicit simultaneous antibody and CD8+ T cell responses in vivo. Although HLA class II restricted epitopes from NY-ESO-1 have been identified, no broad survey has yet established the status of natural CD4+ T cell responses in cancer patients in relation to CD8+ and antibody responses. We used a recently developed general strategy for monitoring CD4+ responses that overcomes the need for prior knowledge of epitope or HLA restriction to analyze a series of 31 cancer patients and healthy donors for the presence of CD4+ T cells to NY-ESO-1, and related this response to NY-ESO-1 expression in tumor cells and serum antibodies to NY-ESO-1. None of the 18 patients that tested seronegative for NY-ESO-1 had detectable CD4+ T cell responses. On the contrary, 11 of 13 cancer patients with serum antibodies to NY-ESO-1 had polyclonal CD4+ T cell responses directed against various known and previously undescribed NY-ESO-1 epitopes. NY-ESO-1 peptide 80-109 was the most immunogenic, with 10 of 11 patients responding to this peptide. We show here that 12-mer determinants from NY-ESO-1 eliciting a CD4+ T cell response were peptide 87-98 with promiscuous HLA class II presentation, peptide 108-119 restricted by HLA-DP4, and peptides 121-132 and 145-156, both shorter epitopes from previously described HLA-DR4 peptides, also presented by HLA-DR7. This study represents the next step in compiling a comprehensive picture of the adaptive immune response to NY-ESO-1, and provides a general strategy for analyzing the CD4+ T cell response to other tumor antigens eliciting a humoral immune response.


Subject(s)
Antibodies, Neoplasm/blood , Antigens, Neoplasm , CD4-Positive T-Lymphocytes/immunology , Membrane Proteins , Neoplasms/immunology , Proteins/immunology , Amino Acid Sequence , Antigen Presentation , Antigens, Neoplasm/genetics , Epitopes/genetics , Female , Histocompatibility Antigens Class II/metabolism , Humans , Immunization , In Vitro Techniques , Lymphocyte Activation , Melanoma/immunology , Molecular Sequence Data , Ovarian Neoplasms/immunology , Proteins/genetics , Recombinant Proteins/genetics , Recombinant Proteins/immunology
19.
J Immunol ; 170(3): 1191-6, 2003 Feb 01.
Article in English | MEDLINE | ID: mdl-12538675

ABSTRACT

NY-ESO-1, a germ cell Ag often detected in tumor tissues, frequently elicits Ab and CD8(+) T cell responses in cancer patients. Overlapping long peptides spanning the NY-ESO-1 sequence have been used to map HLA class I-restricted epitopes recognized by NY-ESO-1-specific CD8(+) T lymphocytes. To address the antigenicity of long peptides, we analyzed two synthetic 30-mer peptides from NY-ESO-1, polypeptides 80-109 and 145-174, for their capacity to be processed by APCs and to stimulate CD8(+) T cells. By incubating APCs with polypeptides at different temperatures or in the presence of protease inhibitors, we found that NY-ESO-1 polypeptides were rapidly internalized by B cells, T2 cells, or PBLs and submitted to cellular proteolytic action to yield nonamer epitopes presented by HLA class I. Polypeptides were also immunogenic in vitro and stimulated the expansion of CD8(+) T cells against naturally processed NY-ESO-1 epitopes in the context of three different HLA class I alleles. Polypeptides can thus serve as exogenous Ags that are cross-presented on HLA class I without requiring the action of professional APCs. These findings support innovative vaccination strategies using NY-ESO-1 polypeptides that would circumvent current limitations of HLA class I peptide vaccination, i.e., HLA eligibility criteria and knowledge of epitope, while allowing for facilitated immunogenicity in the presence of helper epitopes.


Subject(s)
Antigen Presentation , Antigen-Presenting Cells/immunology , HLA Antigens/metabolism , Histocompatibility Antigens Class I/metabolism , Membrane Proteins , Peptide Fragments/immunology , Peptide Fragments/metabolism , Proteins/immunology , Proteins/metabolism , Amino Acid Sequence , Antigen-Presenting Cells/metabolism , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , HLA Antigens/immunology , Histocompatibility Antigens Class I/immunology , Humans , Molecular Sequence Data
20.
Proc Natl Acad Sci U S A ; 99(18): 11813-8, 2002 Sep 03.
Article in English | MEDLINE | ID: mdl-12186971

ABSTRACT

NY-ESO-1 is a germ cell antigen aberrantly expressed in different tumor types that elicits strong humoral and cellular immune responses in cancer patients. Monitoring spontaneous CD8(+) T cell responses against NY-ESO-1 peptides 157-165 (S9C) and 157-167 (S11L) in a series of HLA-A2(+) cancer patients showed that these two peptides had overlapping antigenic profiles and were equally immunogenic. However, discrepancies between S9C and S11L reactivities were observed upon vaccination with both peptides to generate or boost T cell responses to NY-ESO-1 in cancer patients. We here analyze the fine specificity of these responses and describe an HLA-A2-restricted epitope, NY-ESO-1 peptide 159-167 (L9L), which is strongly recognized by CD8(+) T cells as a result of peptide vaccination of cancer patients. Responses to L9L were stimulated by S11L and appeared early in the course of vaccination, independently of S9C responses. However, L9L-specific CD8(+) T cells failed to recognize tumor cells naturally expressing NY-ESO-1 or B lymphoblastoid cells transduced with NY-ESO-1. Processing of L9L could be rescued after IFN-gamma treatment of tumor cells or by dendritic cells pulsed with NY-ESO-1 protein/antibody immune complexes. The present results demonstrate a dual specificity within peptide S11L, with S9C as the natural antigenic tumor epitope, and L9L as a cryptic epitope with dominant immunogenicity upon vaccination that diverts the immune response from tumor recognition. These unanticipated findings raise questions about the use of S11L in the clinic and emphasize the importance of analyzing the fine specificity of vaccine-induced T cell responses in patients as a basis for constructing effective cancer vaccines.


Subject(s)
Antigens, Neoplasm/administration & dosage , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , HLA-A2 Antigen/immunology , Immunodominant Epitopes/immunology , Membrane Proteins , Neoplasms/therapy , Proteins/administration & dosage , Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Cross Reactions , HLA-A2 Antigen/chemistry , Humans , Neoplasms/immunology , Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL