Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 195
Filter
1.
Cancer Immunol Immunother ; 72(7): 2347-2356, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36939853

ABSTRACT

CD4+ T cells that recognize antigenic peptides presented on HLA class II are essential for inducing an optimal anti-tumor immune response, and adoptive transfer of tumor antigen-specific TCR-transduced CD4+ T cells with high responsiveness against tumor is a promising strategy for cancer treatment. Whereas a precise evaluation method of functional avidity, an indicator of T cell responsiveness against tumors, has been established for HLA class I-restricted TCRs, it remains unestablished for HLA class II-restricted TCRs. In this study, we generated a novel platform cell line, CD4-2D3, in which GFP reporter was expressed by NFAT activation via TCR signaling, for correctly evaluating functional avidity of HLA class II-restricted TCRs. Furthermore, using this platform cell line, we succeeded in maturating functional avidity of an HLA class II-restricted TCR specific for a WT1-derived helper peptide by substituting amino acids in complementarity determining region 3 (CDR3) of the TCR. Importantly, we demonstrated that transduction of an avidity-maturated TCR conferred strong cytotoxicity against WT1-expressing leukemia cells on CD4+ T cells, compared to that of its original TCR. Thus, CD4-2D3 cell line should be useful not only to evaluate TCR functional avidity in HLA class II-restricted TCRs but also to screen appropriate TCRs for clinical applications such as cancer immunotherapy.


Subject(s)
Immunotherapy, Adoptive , Neoplasms , Humans , Immunotherapy, Adoptive/methods , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , CD4-Positive T-Lymphocytes , Antigens, Neoplasm
2.
Stat Med ; 42(27): 4990-5006, 2023 11 30.
Article in English | MEDLINE | ID: mdl-37705361

ABSTRACT

In immuno-oncology clinical trials, multiple immunological biomarkers are usually examined over time to comprehensively and appropriately evaluate the efficacy of treatments. Because predicting patients' future survival statuses on the basis of such recorded longitudinal information might be of great interest, joint modeling of longitudinal and time-to-event data has been intensively discussed as a toolkit to implement such a prediction. To achieve a desirable predictive performance, averaging over multiple candidate predictive models to account for the model uncertainty might be a more suitable statistical approach than selecting the single best model. Although Bayesian model averaging can be one of the approaches, several problems related to model weights with marginal likelihoods have been discussed. To address these problems, we here propose a Bayesian predictive model averaging (BPMA) method that uses Bayesian leave-one-out cross-validation predictive densities to account for the subject-specific and time-dependent nature of the prediction. We examine the operating characteristics of the proposed BPMA method in terms of the predictive accuracy (ie, the calibration and discrimination abilities) in extensive simulation studies. In addition, we discuss the strengths and limitations of the proposed method by applying it to an immuno-oncology clinical trial in patients with advanced ovarian cancer.


Subject(s)
Neoplasms , Humans , Bayes Theorem , Computer Simulation , Models, Statistical , Neoplasms/therapy , Probability , Uncertainty , Clinical Trials as Topic
3.
Cancer Immunol Immunother ; 71(1): 189-201, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34089373

ABSTRACT

We have previously revealed the overexpression of Wilms' tumor gene 1 (WT1) in malignant glioma and developed WT1 peptide vaccine cancer immunotherapy. A phase II clinical trial indicated the clinical efficacy of the WT1 peptide vaccine for recurrent malignant glioma. Here, we aimed to investigate the immunological microenvironment in glioma tissues before and after WT1 peptide vaccine treatment. Paired tissue samples were obtained from 20 malignant glioma patients who had received the WT1 peptide vaccine for > 3 months and experienced tumor progression, confirmed radiographically and/or clinically, during vaccination. We discovered that the expression of WT1 and HLA class I antigens in the tumor cells significantly decreased after vaccination. Maintenance of WT1 expression, which is the target molecule of immunotherapy, in tumor cells during the vaccination period was significantly associated with a longer progression-free and overall survival. A high expression of HLA class I antigens and low CD4+/CD8+ tumor-infiltrating lymphocytes (TIL) ratio in pre-vaccination specimens, were also associated with a good prognosis. No statistically significant difference existed in the number of infiltrating CD3+ or CD8+ T cells between the pre- and post-vaccination specimens, whereas the number of infiltrating CD4+ T cells significantly decreased in the post-vaccination specimens. This study provides insight into the mechanisms of intra-tumoral immune reaction/escape during WT1 peptide vaccine treatment and suggests potential clinical strategies for cancer immunotherapy.


Subject(s)
Brain Neoplasms/diagnosis , Brain Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Glioma/diagnosis , Glioma/metabolism , Immunotherapy/methods , WT1 Proteins/biosynthesis , Adult , Biomarkers, Tumor/biosynthesis , CD3 Complex/biosynthesis , CD4-Positive T-Lymphocytes/cytology , Cancer Vaccines , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Female , Gene Expression Profiling , Histocompatibility Antigens Class I/immunology , Humans , Male , Middle Aged , Peptides/chemistry , Prognosis , Proportional Hazards Models
4.
Int J Mol Sci ; 23(20)2022 Oct 12.
Article in English | MEDLINE | ID: mdl-36293034

ABSTRACT

Malignant pleural effusion (MPE) provides a liquid tumor microenvironment model that includes cancer cells and immune cells. However, the characteristics of tumor antigen-specific CD8+ T cells have not been investigated in detail. Here, we analyzed MPE samples taken from a patient with pancreatic cancer who received a dendritic cell vaccine targeting Wilms' Tumor 1 (WT1) antigen over the disease course (two points at MPE1st and 2nd, two months after MPE1st). Epithelial cell adhesion molecule (EpCAM)+ cancer cells (PD-L1- or T cell immunoglobulin mucin-3, TIM-3-), both PD-1 or TIM-3 positive CD8+ T cells, and CD14+CD68+CD163+TIM-3+ macrophages increased from the MPE1st to MPE2nd. The ratio of WT1-specific cytotoxic lymphocytes (WT1-CTLs) to MPE CD8+ T cells and IFN-γ secretion of WT1-CTLs were reduced with disease progression. Coincidentally, the fraction of central memory T (TCM) of WT1-CTLs was decreased. On the other hand, CD8+ T cells in response to SMAD4P130L, which is homogeneously expressed in EpCAM+ cancer cells, were detected using in vitro expansion with the HLA-A*11:01 restrictive SVCVNLYH neoantigen. Furthermore, the CD8+ T cell response to SMAD4P130L was diminished following remarkably decreased numbers of CD8+ TCM in MPE samples. In conclusion, CD8+ T cells responding to WT1 or SMAD4P130L neoantigen expressed in EpCAM+ pancreatic cancer cells were detected in MPE. A tumor antigen-specific immune response would provide novel insight into the MPE microenvironment.


Subject(s)
Pancreatic Neoplasms , Pleural Effusion, Malignant , Vaccines , Humans , Epithelial Cell Adhesion Molecule/metabolism , CD8-Positive T-Lymphocytes , B7-H1 Antigen/metabolism , Hepatitis A Virus Cellular Receptor 2/metabolism , WT1 Proteins , Programmed Cell Death 1 Receptor/metabolism , Mucin-3/metabolism , Pancreatic Neoplasms/pathology , Immunoglobulins/metabolism , Vaccines/metabolism , HLA-A Antigens , Tumor Microenvironment , Smad4 Protein/metabolism
5.
Cancer Immunol Immunother ; 70(1): 253-263, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32696072

ABSTRACT

Simultaneous induction of tumor antigen-specific cytotoxic T lymphocytes (CTLs) and helper T lymphocytes (HTLs) is required for an optimal anti-tumor immune response. WT1332, a 16-mer WT1-derived helper peptide, induce HTLs in an HLA class II-restricted manner and enhance the induction of WT1-specific CTLs in vitro. However, in vivo immune reaction to WT1332 vaccination in tumor-bearing patients remained unclear. Here, a striking difference in WT1-specific T cell responses was shown between WT1 CTL + WT1 helper peptide and WT1 CTL peptide vaccines in patients with recurrent glioma. WT1-specific CTLs were more strongly induced in the patients who were immunized with WT1 CTL + WT1 helper peptide vaccine, compared to those who were immunized with WT1 CTL vaccine alone. Importantly, a clear correlation was demonstrated between WT1-specific CTL and WT1332-specific HTL responses. Interestingly, two novel distinct populations of WT1-tetramerlow WT1-TCRlow CD5low and WT1-tetramerhigh WT1-TCRhigh CD5high CTLs were dominantly detected in WT1 CTL + WT1 helper peptide vaccine. Although natural WT1 peptide-reactive CTLs in the latter population were evidently less than those in the former population, the latter population showed natural WT1 peptide-specific proliferation capacity comparable to the former population, suggesting that the latter population highly expressing CD5, a marker of resistance to activation-induced cell death, should strongly expand and persist for a long time in patients. These results demonstrated the advantage of WT1 helper peptide vaccine for the enhancement of WT1-specific CTL induction by WT1 CTL peptide vaccine.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Peptides/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Subunit/immunology , WT1 Proteins/immunology , CD5 Antigens/immunology , Cell Death/immunology , Cell Proliferation/physiology , Cells, Cultured , Humans , Leukocytes, Mononuclear/immunology , Neoplasm Recurrence, Local/immunology , T-Lymphocytes, Helper-Inducer/immunology , Vaccination/methods
6.
Cancer Immunol Immunother ; 70(11): 3323-3335, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34272593

ABSTRACT

Helper T lymphocytes (HTLs) play a central role in cancer immunity because they can not only help the induction and proliferation of cytotoxic T lymphocytes (CTLs) but also their differentiation into cytotoxic CD4+ T cells and directly kill the target cells.This study describes the identification of three novel mouse Th epitope peptides, WT135-52, WT186-102 and WT1294-312, derived from WT1 protein, which is the most potent tumor-associated antigen. Compared to immunization with WT1 CTL peptide alone, immunization with the addition of these WT1-specific Th peptides strongly induced WT1-specific CTLs, continued to maintain them, and efficiently rejected the challenge of WT1-expressing tumor cells. Importantly, the majority of WT1-specific CTLs induced by the co-immunization with WT1 CTL and the WT1-specific Th peptides were CD44+CD62L- effector memory CD8+ T cells, which played a central role in tumor rejection. Establishment of mouse models suitable for the analysis of the detailed mechanism of these functions of HTLs is very important. These results clearly showed that WT1-specific HTLs perform an essential function in WT1-specific tumor immunity. Therefore, the WT1-specific Th peptides identified here should make a major contribution to elucidation of the mutual roles of WT1-specific CTLs and HTLs in cancer immunity in in vivo mouse models.


Subject(s)
Antigens, Neoplasm/immunology , Epitopes, T-Lymphocyte/immunology , T-Lymphocytes, Helper-Inducer/immunology , WT1 Proteins/immunology , Animals , Male , Mice , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/immunology
7.
BMC Cancer ; 21(1): 1197, 2021 Nov 10.
Article in English | MEDLINE | ID: mdl-34758773

ABSTRACT

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.


Subject(s)
Carcinoma, Pancreatic Ductal/mortality , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasm Recurrence, Local/epidemiology , Pancreatic Neoplasms/immunology , Tumor Microenvironment/immunology , Adult , Aged , Aged, 80 and over , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/surgery , Disease-Free Survival , Female , Follow-Up Studies , Forkhead Transcription Factors/metabolism , Humans , Lymphocytes, Tumor-Infiltrating/metabolism , Male , Middle Aged , Neoplasm Recurrence, Local/prevention & control , Pancreas/immunology , Pancreas/pathology , Pancreas/surgery , Pancreatectomy , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/surgery , Prognosis , Programmed Cell Death 1 Receptor/metabolism , Retrospective Studies
8.
Cancer Immunol Immunother ; 68(2): 331-340, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30430205

ABSTRACT

PURPOSE: The safety and clinical efficacy of WT1 human leukocyte antigen (HLA) class I peptide vaccine have been established, but the safety of a cocktail vaccine of WT1 HLA class I and II peptides has not. To verify its safety, we performed a phase I clinical trial for patients with recurrent malignant gliomas and assessed the immunological responses and survival data. PATIENTS AND METHODS: Fourteen HLA-A*24:02-positive patients with recurrent malignant glioma (2 with grade 3, 12 with grade 4) were enrolled. Every week, the patients received alternately a vaccine containing 3 mg of WT1 HLA-A*24:02-restricted (HLA class I) peptide and a cocktail vaccine of the HLA class I peptide and one of 0.75, 1.5 or 3 mg of the WT1 HLA class II peptide. For patients who showed no significant adverse effects within 6 weeks, the WT1 vaccine was continued at 2-4-week intervals. RESULTS: Eleven of the 14 patients completed WT1 vaccination for 6 weeks, while 3 patients dropped out earlier due to disease progression. All patients showed grade I level of skin disorders at the injection sites. No grade III/IV toxicity or dose-limiting toxicity was observed for any dose of WT1 HLA class II peptide. Six of the 14 patients had stable disease at 6 weeks. Median OS and 1-year OS rates were 24.7 weeks and 36%, respectively. CONCLUSION: The safety of a cocktail vaccine of WT1 HLA class I and II peptides for malignant gliomas was verified. This vaccine is, therefore, considered promising for patients with recurrent malignant glioma.


Subject(s)
Brain Neoplasms/drug therapy , Cancer Vaccines/therapeutic use , Glioma/drug therapy , Vaccines, Subunit/therapeutic use , Adult , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Cancer Vaccines/immunology , Female , Glioma/immunology , Glioma/pathology , HLA-A24 Antigen/immunology , Histocompatibility Antigens Class II/immunology , Humans , Male , Middle Aged , Neoplasm Recurrence, Local , Survival Analysis , Treatment Outcome , Vaccination/methods , Vaccines, Subunit/immunology , WT1 Proteins/immunology
9.
Blood ; 130(15): 1713-1721, 2017 10 12.
Article in English | MEDLINE | ID: mdl-28830889

ABSTRACT

Relapse is a major problem in acute myeloid leukemia (AML) and adversely affects survival. In this phase 2 study, we investigated the effect of vaccination with dendritic cells (DCs) electroporated with Wilms' tumor 1 (WT1) messenger RNA (mRNA) as postremission treatment in 30 patients with AML at very high risk of relapse. There was a demonstrable antileukemic response in 13 patients. Nine patients achieved molecular remission as demonstrated by normalization of WT1 transcript levels, 5 of which were sustained after a median follow-up of 109.4 months. Disease stabilization was achieved in 4 other patients. Five-year overall survival (OS) was higher in responders than in nonresponders (53.8% vs 25.0%; P = .01). In patients receiving DCs in first complete remission (CR1), there was a vaccine-induced relapse reduction rate of 25%, and 5-year relapse-free survival was higher in responders than in nonresponders (50% vs 7.7%; P < .0001). In patients age ≤65 and >65 years who received DCs in CR1, 5-year OS was 69.2% and 30.8% respectively, as compared with 51.7% and 18% in the Swedish Acute Leukemia Registry. Long-term clinical response was correlated with increased circulating frequencies of polyepitope WT1-specific CD8+ T cells. Long-term OS was correlated with interferon-γ+ and tumor necrosis factor-α+ WT1-specific responses in delayed-type hypersensitivity-infiltrating CD8+ T lymphocytes. In conclusion, vaccination of patients with AML with WT1 mRNA-electroporated DCs can be an effective strategy to prevent or delay relapse after standard chemotherapy, translating into improved OS rates, which are correlated with the induction of WT1-specific CD8+ T-cell response. This trial was registered at www.clinicaltrials.gov as #NCT00965224.


Subject(s)
Cancer Vaccines/immunology , Dendritic Cells/immunology , Leukemia, Myeloid, Acute/prevention & control , Leukemia, Myeloid, Acute/therapy , Vaccination , Aged , Biomarkers, Tumor/metabolism , Cytokines/metabolism , Disease-Free Survival , Electroporation , Female , Humans , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/immunology , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recurrence , Remission Induction , Treatment Outcome , WT1 Proteins/genetics , WT1 Proteins/metabolism
10.
Cancer Invest ; 37(9): 463-477, 2019.
Article in English | MEDLINE | ID: mdl-31490702

ABSTRACT

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.


Subject(s)
B7-H1 Antigen/metabolism , Carcinoma, Pancreatic Ductal/surgery , Lymphocytes, Tumor-Infiltrating/immunology , Pancreatic Neoplasms/surgery , Adult , Aged , Aged, 80 and over , Carcinoma, Pancreatic Ductal/immunology , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Pancreatic Neoplasms/immunology , Prognosis , Survival Analysis , T-Lymphocytes/immunology , Tumor Microenvironment
11.
Oncology ; 97(3): 135-148, 2019.
Article in English | MEDLINE | ID: mdl-31216557

ABSTRACT

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.


Subject(s)
Biomarkers, Tumor , Cancer Vaccines/immunology , Dendritic Cells/immunology , Immunotherapy , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/mortality , WT1 Proteins/immunology , Biomarkers , Cancer Vaccines/administration & dosage , Combined Modality Therapy , Dendritic Cells/metabolism , Epitopes/immunology , Female , Humans , Immunophenotyping , Male , Matrix Metalloproteinase 9/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/therapy , Peptides/immunology , Peroxidase/metabolism , Prognosis , Transforming Growth Factor beta1/metabolism , Treatment Outcome , Vaccination , WT1 Proteins/genetics
12.
Int J Cancer ; 142(11): 2375-2382, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29322496

ABSTRACT

Thymic epithelial tumors are rare malignancies, and no optimal therapeutic regimen has been defined for patients with advanced disease. Patients with advanced thymic epithelial tumors, which were resistant or intolerable to prior therapies, were eligible for this study. Patients received 9 mer-WT1-derived peptide emulsified with Montanide ISA51 adjuvant via intradermal administration once a week as a monotherapy. After the 3-month-protocol treatment, the treatment was continued mostly at intervals of 2-4 weeks until disease progression or intolerable adverse events occurred. Of the 15 patients enrolled, 11 had thymic carcinoma (TC) and 4 had invasive thymoma (IT). Median period from diagnosis to the start of treatment was 13.3 and 65.5 months for TC and IT, respectively. No patients achieved a complete or partial response. Of the 8 evaluable TC patients, 6 (75.0%) had stable disease (SD) and 2 had progressive disease (PD). Of the 4 evaluable IT patients, 3 (75.0%) had SD and 1 (25.0%) had PD. Median period of monotherapy treatment was 133 and 683 days in TC and IT patients, respectively. No severe adverse events occurred during the 3-month-protocol treatment. As adverse events in long responders, thymoma-related autoimmune complications, pure red cell aplasia and myasthenia gravis occurred in two IT patients. Cerebellar hemorrhage developed in a TC patient complicated with Von Willebrand disease. Induction of WT1-specific immune responses was observed in the majority of the patients. WT1 peptide vaccine immunotherapy may have antitumor potential against thymic malignancies.


Subject(s)
Immunotherapy , Neoplasms, Glandular and Epithelial/immunology , Neoplasms, Glandular and Epithelial/pathology , Peptides/immunology , Thymus Neoplasms/immunology , Thymus Neoplasms/pathology , WT1 Proteins/immunology , Adult , Aged , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Combined Modality Therapy , Female , Gene Expression , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasm Staging , Neoplasms, Glandular and Epithelial/diagnostic imaging , Neoplasms, Glandular and Epithelial/drug therapy , Thymus Neoplasms/diagnostic imaging , Thymus Neoplasms/drug therapy , Tomography, X-Ray Computed , Treatment Outcome , WT1 Proteins/chemistry , WT1 Proteins/metabolism
13.
Nihon Rinsho ; 75(2): 263-269, 2017 02.
Article in Japanese | MEDLINE | ID: mdl-30562862

ABSTRACT

Wilms'tumor gene 1(WT1) is a leukemia cell-specific marker, and WT1 protein is pan- tumor-associated antigen (TAA) that was ranked as a top among 75 TAAs. WT1 peptide vaccine has been performing for over 800 patients with 58 kinds of malignancies including leukemia and various types of solid tumors. Clinical results are promising, and adverse effects are limited to skin eruption on the vaccine injection sites. Two types of WT1 peptide vaccines are under clinical studies by pharmaceutical companies for the development of medicine.


Subject(s)
Cancer Vaccines/therapeutic use , Neoplasms/therapy , WT1 Proteins/immunology , Cancer Vaccines/immunology , Humans , Immunotherapy , Neoplasms/immunology , Vaccines, Subunit/immunology , Vaccines, Subunit/therapeutic use
14.
Int J Cancer ; 139(6): 1391-401, 2016 Sep 15.
Article in English | MEDLINE | ID: mdl-27170523

ABSTRACT

We previously evaluated Wilms' tumor gene 1 (WT1) peptide vaccination in a large number of patients with leukemia or solid tumors and have reported that HLA-A*24:02 restricted, 9-mer WT1-235 peptide (CYTWNQMNL) vaccine induces cellular immune responses and elicits WT1-235-specific cytotoxic T lymphocytes (CTLs). However, whether this vaccine induces humoral immune responses to produce WT1 antibody remains unknown. Thus, we measured IgG antibody levels against the WT1-235 peptide (WT1-235 IgG antibody) in patients with glioblastoma multiforme (GBM) receiving the WT1 peptide vaccine. The WT1-235 IgG antibody, which was undetectable before vaccination, became detectable in 30 (50.8%) of a total of 59 patients during 3 months of WT1 peptide vaccination. The dominant WT1-235 IgG antibody subclass was Th1-type, IgG1 and IgG3 . WT1-235 IgG antibody production was significantly and positively correlated with both progression-free survival (PFS) and overall survival (OS). Importantly, the combination of WT1-235 IgG antibody production and positive delayed type-hypersensitivity (DTH) to the WT1-235 peptide was a better prognostic marker for long-term OS than either parameter alone. These results suggested that WT1-235 peptide vaccination induces not only WT1-235-specific CTLs as previously described but also WT1-235-specific humoral immune responses associated with antitumor cellular immune response. Our results indicate that the WT1 IgG antibody against the WT1 peptide may be a useful predictive marker, with better predictive performance in combination with DTH to WT1 peptide, and provide a new insight into the antitumor immune response induction in WT1 peptide vaccine-treated patients.


Subject(s)
Cancer Vaccines/immunology , Glioblastoma/immunology , Glioblastoma/mortality , Immunoglobulin G/immunology , Peptides/immunology , WT1 Proteins/immunology , Adult , Aged , Biomarkers , Cancer Vaccines/administration & dosage , Cell Line, Tumor , Combined Modality Therapy , Enzyme-Linked Immunosorbent Assay , Female , Glioblastoma/therapy , HLA-A24 Antigen/immunology , Humans , Immunoglobulin G/blood , Immunotherapy , Male , Middle Aged , Peptides/administration & dosage , Prognosis , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Treatment Outcome , Vaccination , WT1 Proteins/chemistry , Young Adult
15.
Clin Immunol ; 168: 6-15, 2016 07.
Article in English | MEDLINE | ID: mdl-27132023

ABSTRACT

In the present study, the combined adjuvant effect of 7DW8-5, a potent α-GalCer-analog, and monophosphoryl lipid A (MPLA), a TLR4 agonist, on the induction of vaccine-induced CD8(+) T-cell responses and protective immunity was evaluated. Mice were immunized with peptides corresponding to the CD8(+) T-cell epitopes of a malaria antigen, a circumsporozoite protein of Plasmodium yoelii, and a tumor antigen, a Wilms Tumor antigen-1 (WT-1), together with 7DW8-5 and MPLA, as an adjuvant. These immunization regimens were able to induce higher levels of CD8(+) T-cell responses and, ultimately, enhanced levels of protection against malaria and tumor challenges compared to the levels induced by immunization with peptides mixed with 7DW8-5 or MPLA alone. Co-administration of 7DW8-5 and MPLA induces activation of memory-like effector natural killer T (NKT) cells, i.e. CD44(+)CD62L(-)NKT cells. Our study indicates that 7DW8-5 greatly enhances important synergistic pathways associated to memory immune responses when co-administered with MPLA, thus rendering this combination of adjuvants a novel vaccine adjuvant formulation.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , Galactosylceramides/pharmacology , Killer Cells, Natural/drug effects , Lipid A/analogs & derivatives , Toll-Like Receptor 4/agonists , Amino Acid Sequence , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Drug Synergism , Epitopes, T-Lymphocyte/immunology , Galactosylceramides/administration & dosage , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , Humans , Immunization/methods , Immunologic Memory/immunology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lipid A/administration & dosage , Lipid A/pharmacology , Malaria/immunology , Malaria/parasitology , Malaria/prevention & control , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Models, Immunological , Peptides/immunology , Plasmodium yoelii/immunology , Plasmodium yoelii/physiology , Protective Agents/administration & dosage , Protective Agents/pharmacology , Protozoan Proteins/chemistry , Protozoan Proteins/immunology , Toll-Like Receptor 4/metabolism , WT1 Proteins/genetics , WT1 Proteins/immunology
16.
Mol Carcinog ; 55(12): 2001-2009, 2016 12.
Article in English | MEDLINE | ID: mdl-26713860

ABSTRACT

In our previous study, we showed that miR-125a directly targeted a WT1 oncogene, which was overexpressed in leukemia and various kinds of solid tumors including lung, breast, gastric, and colon cancers, and brain tumors and was deeply involved in leukemogenesis and tumorigenesis and that miR-125a knockout mice overexpressed WT1 and developed myeloproliferative disease. It had been also reported that miR-125a is downregulated in leukemia and various types of solid tumors such as lung cancers, suggesting its tumor suppressor function. Therefore, it is important to elucidate what is target(s) of miR-125a for understandings of such functions although few target genes for it are known. In the present study, Zbtb7a oncogene was identified as a potential target for miR-125a by gene expression profiling in miR-125a knockout mice combined with bioinformatics target prediction. EGFP-3'UTR reporter assay showed that miR-125a suppressed Zbtb7a expression through its direct binding to the Zbtb7a-3'UTR. Zbtb7a knockdown by siRNA suppressed cell proliferation and induced G1 cell cycle arrest and apoptosis in lung cancer cells. Furthermore, miR-125a expression showed a negative correlation with Zbtb7a expression in non-small cell lung cancer tissues. The present study showed for the first time that Zbtb7a was a direct target for miR-125a and was involved in cell cycle progression and apoptosis of lung cancer cells. These results also demonstrated that deregulation of miR-125a-Zbtb7a signaling was associated with the development and progression of lung cancer. © 2015 Wiley Periodicals, Inc.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , MicroRNAs/genetics , Transcription Factors/genetics , Animals , Apoptosis , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation , Humans , Lung/metabolism , Lung/pathology , Lung Neoplasms/pathology , Mice, Knockout , Proto-Oncogene Mas
17.
Pediatr Blood Cancer ; 63(2): 234-41, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26469989

ABSTRACT

BACKGROUND: Advances in cancer immunotherapy in the pediatric field are needed in order to improve the prognosis of children with malignancies. We conducted a prospective phase I/II study of WT1 peptide vaccination for children with relapsed or refractory malignancies. METHODS: The main eligibility criteria were affected tissues or leukemic cells expressing the WT1 gene, and patients (and donors for allogeneic hematopoietic stem cell transplantation) having HLA-A*24:02. Vaccination using the WT1 peptide (CYTWNQMNL), which was modified for higher affinity to this HLA-type molecule with the adjuvant Montanide ISA51, was performed weekly 12 times. RESULTS: Twenty-six patients were enrolled and 13 (50.0%) completed the vaccination 12 times. Evidence for the induction of WT1-specific cytotoxic T-lymphocyte (CTL) responses without severe systemic side effects was obtained. Two out of 12 patients with bulky disease exhibited a transient clinical effect (one mixed response and one stable disease), three out of six patients with minimal residual disease achieved transient molecular remission, and five out of eight patients without a detectable level of the molecular marker, but with a high risk of relapse, had the best outcome of long-term continuous complete remission. CONCLUSIONS: WT1 vaccination is a safe immunotherapy and induced WT1-specific CTL responses in children; however, as a single agent, vaccination only provided patients in remission, but with a high risk of relapse, with "long-term benefits" in the context of its use for relapse prevention. WT1 peptide-based treatments in combination with other modalities, such as anti-tumor drugs or immunomodulating agents, need to be planned.


Subject(s)
Cancer Vaccines/therapeutic use , Neoplasms/therapy , Peptides/therapeutic use , WT1 Proteins/immunology , WT1 Proteins/therapeutic use , Adjuvants, Immunologic , Adolescent , Antineoplastic Agents/therapeutic use , Child , Child, Preschool , Female , Flow Cytometry , Humans , Immunohistochemistry , Infant , Infant, Newborn , Male , Mannitol/administration & dosage , Mannitol/analogs & derivatives , Neoplasms/genetics , Oleic Acids/administration & dosage , Peptides/immunology , Polymerase Chain Reaction , T-Lymphocytes, Cytotoxic/immunology
18.
Cell Physiol Biochem ; 35(2): 647-62, 2015.
Article in English | MEDLINE | ID: mdl-25613309

ABSTRACT

BACKGROUND: A novel link between oncogenic KRAS signalling and WT1 was recently identified. We sought to investigate the role of WT1 and KRAS in proliferation and apoptosis. METHODS: KRAS mutations and WT1 (cMyc) expression were detected using Sanger sequencing and real-time PCR in 77 patients with non-small cell lung cancer (NSCLC). Overexpression and knockdown of WT1 were generated with plasmid and siRNA via transient transfection technology in H1299 and H1568 cells. MTT assay for detection of cell proliferation, and TUNEL assay and proteomic profiler assay for apoptosis evaluation were carried out. Dual luciferase reporter assay and ChIP-PCR were performed to validate the effect of WT1 on the cMyc promoter. RESULTS: KRAS mutations showed a negative impact on overall survival (OS). High expressions of WT1 and cMyc were associated with poor OS in KRAS mutant subgroup. The potential mechanisms that WT1 promotes proliferation and impedes apoptosis through affecting multiple apoptosis-related regulators in KRAS mutant NSCLC cells were identified. WT1 could activate cMyc promoter directly in KRAS mutant cells. CONCLUSION: The results suggest that WT1 and c-MYC expression is important for survival in KRAS mutant tumors as opposed to KRAS wild-type tumors. For treatment of KRAS mutant NSCLC, targeting WT1 and cMyc may provide alternative therapeutic strategies.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins/genetics , WT1 Proteins/genetics , ras Proteins/genetics , Aged , Aged, 80 and over , Apoptosis , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Middle Aged , Mutation , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins p21(ras) , Survival Analysis , WT1 Proteins/metabolism
19.
Cancer Immunol Immunother ; 64(7): 791-804, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25835542

ABSTRACT

Wilms' tumor gene 1 (WT1) protein is a promising tumor-associated antigen for cancer immunotherapy. We have been performing WT1 peptide vaccination with good clinical responses in over 750 patients with leukemia or solid cancers. In this study, we generated single-cell gene-expression profiles of the effector memory (EM) subset of WT1-specific cytotoxic T lymphocytes (CTLs) in peripheral blood of nine acute myeloid leukemia patients treated with WT1 peptide vaccine, in order to discriminate responders (WT1 mRNA levels in peripheral blood decreased to undetectable levels, decreased but stayed at abnormal levels, were stable at undetectable levels, or remained unchanged from the initial abnormal levels more than 6 months after WT1 vaccination) from non-responders (leukemic blast cells and/or WT1 mRNA levels increased relative to the initial state within 6 months of WT1 vaccination) prior to WT1 vaccination. Cluster and principal component analyses performed using 83 genes did not discriminate between responders and non-responders prior to WT1 vaccination. However, these analyses revealed that EM subset of WT1-specific CTLs could be divided into two groups: the "activated" and "quiescent" states; in responders, EM subset of the CTLs shifted to the "quiescent" state, whereas in non-responders, those shifted to the "activated" state following WT1 vaccination. These results demonstrate for the first time the existence of two distinct EM states, each of which was characteristic of responders or non-responders, of WT1-specific CTLs in AML patients, and raises the possibility of using advanced gene-expression profile analysis to clearly discriminate between responders and non-responders prior to WT1 vaccination.


Subject(s)
Antigens, Neoplasm/immunology , Immunologic Memory/immunology , Leukemia, Myeloid, Acute/immunology , T-Lymphocytes, Cytotoxic/immunology , WT1 Proteins/immunology , Adult , Aged , Antigens, Neoplasm/genetics , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Female , Gene Expression Profiling , Humans , Immunotherapy/methods , Leukemia, Myeloid, Acute/therapy , Male , Middle Aged , Principal Component Analysis , RNA, Messenger/blood , RNA, Messenger/genetics , T-Lymphocytes, Cytotoxic/cytology , Vaccines, Subunit/immunology , Vaccines, Subunit/therapeutic use , WT1 Proteins/genetics
20.
Cancer Immunol Immunother ; 64(6): 707-16, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25772149

ABSTRACT

To investigate the safety of combined Wilms tumor 1 peptide vaccination and temozolomide treatment of glioblastoma, a phase I clinical trial was designed. Seven patients with histological diagnosis of glioblastoma underwent concurrent radiotherapy and temozolomide therapy. Patients first received Wilms tumor 1 peptide vaccination 1 week after the end of combined concurrent radio/temozolomide therapy, and administration was continued once per week for 7 weeks. Temozolomide maintenance was started and performed for up to 24 cycles, and the observation period for safety encompassed 6 weeks from the first administration of maintenance temozolomide. All patients showed good tolerability during the observation period. Skin disorders, such as grade 1/2 injection-site reactions, were observed in all seven patients. Although grade 3 lymphocytopenia potentially due to concurrent radio/temozolomide therapy was observed in five patients (71.4 %), no other grade 3/4 hematological or neurological toxicities were observed. No autoimmune reactions were observed. All patients are still alive, and six are on Wilms tumor 1 peptide vaccination without progression, yielding a progression-free survival from histological diagnosis of 5.2-49.1 months. Wilms tumor 1 peptide vaccination was stopped in one patient after 12 injections by the patient's request. The safety profile of the combined Wilms tumor 1 peptide vaccination and temozolomide therapy approach for treating glioblastoma was confirmed.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Brain Neoplasms/therapy , Cancer Vaccines/administration & dosage , Dacarbazine/analogs & derivatives , Glioblastoma/therapy , WT1 Proteins/administration & dosage , WT1 Proteins/immunology , Adult , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Brain Neoplasms/drug therapy , Brain Neoplasms/immunology , Cohort Studies , Combined Modality Therapy , Dacarbazine/administration & dosage , Dacarbazine/adverse effects , Disease Progression , Female , Glioblastoma/drug therapy , Glioblastoma/immunology , Humans , Male , Middle Aged , Temozolomide , WT1 Proteins/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL