Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Rinsho Ketsueki ; 64(8): 782-790, 2023.
Article in Japanese | MEDLINE | ID: mdl-37673631

ABSTRACT

Immune checkpoints suppress inappropriate immune responses to self-molecules or cells. Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1) expressed in T cells are representative molecules involved in the immune checkpoint system. The recent advent of immune checkpoint inhibitors (ICIs) has drastically changed cancer immunotherapy because a substantial proportion of patients with advanced cancers have responded to ICIs and some of them have been cured. This benefit is due to T-cell rescue from immune suppression in their tumor microenvironment by blocking cluster of differentiation 80/CTLA-4 and PD-L1/PD-1 interactions. However, blocking these interactions also liberates T cells that are reactive to self-antigens from tolerance, resulting in the occurrence of autoimmune diseases, that is, immune-related adverse events. Although the primary target organs are the lungs, gastrointestinal tract, and endocrine glands, hematopoietic cells are also affected in 0.5-3% of patients, potentially resulting in anemia or thrombocytopenia. Because hematopoietic system homeostasis is critical to maintaining life support, the occurrence of grade 3-4 irAEs in the hematopoietic system is directly life-threatening. Herein, we review the relationship between ICIs and toxicities in patients with cancer and describe the characteristics and management strategies for hematological immune-related adverse events.


Subject(s)
Autoimmune Diseases , Thrombocytopenia , Humans , Immune Checkpoint Inhibitors , CTLA-4 Antigen , Programmed Cell Death 1 Receptor
2.
Nature ; 534(7607): 402-6, 2016 06 16.
Article in English | MEDLINE | ID: mdl-27281199

ABSTRACT

Successful treatment of many patients with advanced cancer using antibodies against programmed cell death 1 (PD-1; also known as PDCD1) and its ligand (PD-L1; also known as CD274) has highlighted the critical importance of PD-1/PD-L1-mediated immune escape in cancer development. However, the genetic basis for the immune escape has not been fully elucidated, with the exception of elevated PD-L1 expression by gene amplification and utilization of an ectopic promoter by translocation, as reported in Hodgkin and other B-cell lymphomas, as well as stomach adenocarcinoma. Here we show a unique genetic mechanism of immune escape caused by structural variations (SVs) commonly disrupting the 3' region of the PD-L1 gene. Widely affecting multiple common human cancer types, including adult T-cell leukaemia/lymphoma (27%), diffuse large B-cell lymphoma (8%), and stomach adenocarcinoma (2%), these SVs invariably lead to a marked elevation of aberrant PD-L1 transcripts that are stabilized by truncation of the 3'-untranslated region (UTR). Disruption of the Pd-l1 3'-UTR in mice enables immune evasion of EG7-OVA tumour cells with elevated Pd-l1 expression in vivo, which is effectively inhibited by Pd-1/Pd-l1 blockade, supporting the role of relevant SVs in clonal selection through immune evasion. Our findings not only unmask a novel regulatory mechanism of PD-L1 expression, but also suggest that PD-L1 3'-UTR disruption could serve as a genetic marker to identify cancers that actively evade anti-tumour immunity through PD-L1 overexpression.


Subject(s)
3' Untranslated Regions/genetics , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Programmed Cell Death 1 Receptor/genetics , Tumor Escape/genetics , Up-Regulation , Adenocarcinoma/genetics , Animals , Antibodies/pharmacology , Antibodies/therapeutic use , CRISPR-Cas Systems , Cell Line, Tumor , Clonal Selection, Antigen-Mediated , Female , Genetic Markers/genetics , Humans , Leukemia-Lymphoma, Adult T-Cell/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Mice , Neoplasms/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/biosynthesis , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stomach Neoplasms/genetics
3.
Blood ; 132(11): 1134-1145, 2018 09 13.
Article in English | MEDLINE | ID: mdl-30045840

ABSTRACT

The recent success of chimeric antigen receptor (CAR)-T cell therapy for treatment of hematologic malignancies supports further development of treatments for both liquid and solid tumors. However, expansion of CAR-T cell therapy is limited by the availability of surface antigens specific for the tumor while sparing normal cells. There is a rich diversity of tumor antigens from intracellularly expressed proteins that current and conventional CAR-T cells are unable to target. Furthermore, adoptively transferred T cells often suffer from exhaustion and insufficient expansion, in part, because of the immunosuppressive mechanisms operating in tumor-bearing hosts. Therefore, it is necessary to develop means to further activate and expand those CAR-T cells in vivo. The Wilms tumor 1 (WT1) is an intracellular oncogenic transcription factor that is an attractive target for cancer immunotherapy because of its overexpression in a wide range of leukemias and solid tumors, and a low level of expression in normal adult tissues. In the present study, we developed CAR-T cells consisting of a single chain variable fragment (scFv) specific to the WT1235-243/HLA-A*2402 complex. The therapeutic efficacy of our CAR-T cells was demonstrated in a xenograft model, which was further enhanced by vaccination with dendritic cells (DCs) loaded with the corresponding antigen. This enhanced efficacy was mediated, at least partly, by the expansion and activation of CAR-T cells. CAR-T cells shown in the present study not only demonstrate the potential to expand the range of targets available to CAR-T cells, but also provide a proof of concept that efficacy of CAR-T cells targeting peptide/major histocompatibility complex can be boosted by vaccination.


Subject(s)
Immunity, Cellular , Immunotherapy, Adoptive , Neoplasms/therapy , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Vaccination , WT1 Proteins/immunology , Animals , Cell Line, Tumor , Humans , Neoplasms/immunology , Neoplasms/pathology , T-Lymphocytes/pathology , T-Lymphocytes/transplantation , Xenograft Model Antitumor Assays
4.
Blood ; 130(18): 1985-1994, 2017 11 02.
Article in English | MEDLINE | ID: mdl-28860210

ABSTRACT

Wilms' tumor 1 (WT1) is constantly expressed in leukemic cells of acute leukemia and myelodysplastic syndrome (MDS). A T-cell receptor (TCR) that specifically reacts with WT1 peptide in the context of HLA-A*24:02 has been identified. We conducted a first-in-human trial of TCR-gene transduced T-cell (TCR-T-cell) transfer in patients with refractory acute myeloblastic leukemia (AML) and high-risk MDS to investigate the safety and cell kinetics of the T cells. The WT1-specific TCR-gene was transduced to T cells using a retroviral vector encoding small interfering RNAs for endogenous TCR genes. The T cells were transferred twice with a 4-week interval in a dose-escalating design. After the second transfer, sequential WT1 peptide vaccines were given. Eight patients, divided into 2 dose cohorts, received cell transfer. No adverse events of normal tissue were seen. The TCR-T cells were detected in peripheral blood for 8 weeks at levels proportional to the dose administered, and in 5 patients, they persisted throughout the study period. The persisting cells maintained ex vivo peptide-specific immune reactivity. Two patients showed transient decreases in blast counts in bone marrow, which was associated with recovery of hematopoiesis. Four of 5 patients who had persistent T cells at the end of the study survived more than 12 months. These results suggest WT1-specific TCR-T cells manipulated by ex vivo culture of polyclonal peripheral lymphocytes survived in vivo and retained the capacity to mount an immune reaction to WT1. This trial was registered at www.umin.ac.jp as #UMIN000011519.


Subject(s)
Genes, T-Cell Receptor , Leukemia, Myeloid, Acute/therapy , Myelodysplastic Syndromes/therapy , T-Lymphocytes/metabolism , Transduction, Genetic , WT1 Proteins/genetics , Adoptive Transfer , Aged , Bone Marrow/pathology , Female , Humans , Kinetics , Leukemia, Myeloid, Acute/genetics , Male , Middle Aged , Myelodysplastic Syndromes/genetics , Peptides/pharmacology
5.
Rinsho Ketsueki ; 60(9): 1341-1350, 2019.
Article in Japanese | MEDLINE | ID: mdl-31597862

ABSTRACT

It has been eight years since the first immune checkpoint-blocking antibody, ipilimumab, was approved for metastatic malignant melanoma treatment by FDA in 2011. During this period, several other immune checkpoint blockers have been newly developed and approved for certain cancers, including malignant melanoma. However, there have been several concerns with some of these. The overall response rate did not exceed 30% in many cancers; although combination therapy with ipilimumab and nivolumab increased efficacy, immune-related adverse events also increased. This observation facilitated the reverse translational research (rTR) approach, using clinical specimens from treated patients to gradually elucidate the mechanism of resistance and biomarkers to select patients who can potentially benefit from immunotherapy. This has also promoted the development of novel combination therapies. In this review, immunological findings that highlight the resistance mechanisms of cancers against immune checkpoint blockers and the novel attempts to achieve a break-through will be discussed.


Subject(s)
Immunotherapy , Ipilimumab/therapeutic use , Melanoma/therapy , Nivolumab/therapeutic use , Skin Neoplasms/therapy , Drug Resistance, Neoplasm , Humans , Translational Research, Biomedical
6.
Vox Sang ; 113(8): 787-794, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30251432

ABSTRACT

BACKGROUND AND OBJECTIVES: Determination of the anti-A/-B titre pre- and post-transplantation is beneficial for treatment selection. Currently, the recommended method for antibody titration is the tube test (TT) assay. Dithiothreitol (DTT) is used for IgM antibody inactivation. Recently, a fully automated antibody titration assay using the column agglutination technique (CAT) was developed (auto-CAT). Our aim was to compare the auto-CAT and TT techniques for ABO antibody titration, to evaluate the effectiveness of DTT-treated plasma for use with auto-CAT and to define the cut-off value for antibody titration by auto-CAT. MATERIALS AND METHODS: We enrolled 30 healthy individuals, including 10 each for blood types A, B and O. We performed antibody titre measurement using the TT technique and auto-CAT simultaneously. Auto-CAT uses the bead column agglutination technology. RESULTS: With the auto-CAT cut-off value set to weak (w)+ with DTT treatment plasma, the concordance rate was 45%, and the weighted kappa value between TT and auto-CAT results was 0·994 in all subjects. Furthermore, there was a significant positive correlation between the anti-A/-B titre results obtained using the TT technique and auto-CAT in all blood types. Moreover, a positive bias (falsely elevated end-points due to agglomeration of A/B cells) was not observed in auto-CAT testing using DTT-treated plasma. CONCLUSION: Our results show that 1+ agglutination using the TT technique is equivalent to w+ agglutination obtained using auto-CAT. We recommend that DTT may be used with auto-CAT to measure antibody titres. Thus, we suggest that auto-CAT is useful for antibody titration in routine examination.


Subject(s)
Agglutination Tests/methods , Blood Group Antigens/immunology , Adult , Agglutination Tests/standards , Blood Group Antigens/blood , Female , Humans , Male , Middle Aged , Sensitivity and Specificity
7.
Blood ; 126(25): 2752-63, 2015 Dec 17.
Article in English | MEDLINE | ID: mdl-26432889

ABSTRACT

Acute graft-versus-host disease (aGVHD) represents one of the major complications in allogeneic stem cell transplantation and is primarily caused by genetic disparity between the donor and recipient. In HLA-matched transplants, the disparity is thought to be determined by loci encoding minor histocompatibility antigens (minor H antigens), which are presented by specific HLA molecules. We performed a genome-wide association study (GWAS) to identify minor H antigen loci associated with aGVHD. A total of 500 568 single nucleotide polymorphisms (SNPs) were genotyped for donors and recipients from 1589 unrelated bone marrow transplants matched for HLA-A, -B, -C, -DRB1, and -DQB1, followed by the imputation of unobserved SNPs. We interrogated SNPs whose disparity between the donor and recipient was significantly associated with aGVHD development. Without assuming HLA unrestriction, we successfully captured a known association between HLA-DPB1 disparity (P = 4.50 × 10(-9)) and grade II-IV aGVHD development, providing proof of concept for the GWAS design aimed at discovering genetic disparity associated with aGVHD. In HLA-restricted analyses, whereby association tests were confined to major subgroups sharing common HLA alleles to identify putative minor H antigen loci, we identified 3 novel loci significantly associated with grade III-IV aGVHD. Among these, rs17473423 (P = 1.20 × 10(-11)) at 12p12.1 within the KRAS locus showed the most significant association in the subgroup, sharing HLA-DQB1*06:01. Our result suggested that a GWAS can be successfully applied to identify allele mismatch associated with aGVHD development, contributing to the understanding of the genetic basis of aGVHD.


Subject(s)
Graft vs Host Disease/genetics , Hematopoietic Stem Cell Transplantation , Minor Histocompatibility Antigens/genetics , Adolescent , Adult , Aged , Alleles , Child , Child, Preschool , Female , Genome-Wide Association Study , Genotype , HLA-DQ beta-Chains/genetics , Histocompatibility Testing , Humans , Infant , Infant, Newborn , Male , Middle Aged , Polymorphism, Single Nucleotide , Young Adult
8.
Hematol Oncol ; 35(1): 87-93, 2017 Mar.
Article in English | MEDLINE | ID: mdl-26177728

ABSTRACT

Epstein-Barr virus (EBV)-encoded small RNA in situ hybridization (EBER-ISH) is a widely accepted method to evaluate EBV involvement in diffuse large B-cell lymphoma (DLBCL), although little is known regarding associations between EBV DNA load and the EBER status and whether EBV DNA load data provide additional clinical information. In this study, we quantified EBV DNA load in diagnostic specimens from DLBCL patients diagnosed at our hospital to evaluate clinical implications of EBV DNA load in diagnostic specimens as contrasted with EBER-ISH. Among 140 DLBCL patients without underlying immunodeficiency, 51 were evaluable for both EBER and EBV DNA load, 83 for EBER only and one for EBV DNA load only. The median EBV DNA load was 708 copies/µg. Although EBV DNA load was significantly higher for EBER-positive patients than for EBER-negative patients (p < 0.001), EBV DNA was detected in up to 72% of EBER-negative patients. Progression-free survival and overall survival were significantly worse for patients with EBV DNA load above 700 copies/µg than for those with EBV DNA load below 700 copies/µg (p = 0.009 and p = 0.003); they were also significantly worse for EBER-positive patients than for EBER-negative patients (p < 0.001 and p = 0.001). Even among EBER-negative patients, higher EBV DNA load conferred worse progression-free survival and overall survival (p = 0.041 and p = 0.013). These findings indicate that EBV DNA load in diagnostic specimens is not a simple surrogate for the EBER status and may be a potential biomarker associated with EBV involvement and prognosis in DLBCL. Copyright © 2015 John Wiley & Sons, Ltd.


Subject(s)
DNA, Viral/blood , Epstein-Barr Virus Infections/diagnosis , Herpesvirus 4, Human , Lymphoma, Large B-Cell, Diffuse/virology , Viral Load , Adult , Aged , Aged, 80 and over , Disease-Free Survival , Epstein-Barr Virus Infections/complications , Female , Humans , In Situ Hybridization , Kaplan-Meier Estimate , Lymphoma, Large B-Cell, Diffuse/complications , Lymphoma, Large B-Cell, Diffuse/diagnosis , Male , Middle Aged , Prognosis , RNA, Viral , Retrospective Studies , Treatment Outcome
9.
Genes Chromosomes Cancer ; 55(3): 242-50, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26542893

ABSTRACT

ETV6, which encodes an ETS family transcription factor, is frequently rearranged in human leukemias. We show here that a patient with acute myeloid leukemia with t(7;11)(p15;p15) gained, at the time of relapse, t(11;12)(q12.1;p13) with a split ETV6 FISH signal. Using 3'-RACE PCR analysis, we found that ETV6 was fused to LPXN at 11q12.1, which encodes leupaxin. ETV6-LPXN, an in-frame fusion between exon 4 of ETV6 and exon 2 of LPXN, did not transform the interleukin-3-dependent 32D myeloid cell line to cytokine independence; however, an enhanced proliferative response was observed when these cells were treated with G-CSF without inhibition of granulocytic differentiation. The 32D and human leukemia cell lines each transduced with ETV6-LPXN showed enhanced migration towards the chemokine CXCL12. We show here for the first time that LPXN is a fusion partner of ETV6 and present evidence indicating that ETV6-LPXN plays a crucial role in leukemia progression through enhancing the response to G-CSF and CXCL12.


Subject(s)
Cell Adhesion Molecules/genetics , Homeodomain Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Nuclear Pore Complex Proteins/genetics , Oncogene Proteins, Fusion/genetics , Phosphoproteins/genetics , Proto-Oncogene Proteins c-ets/genetics , Repressor Proteins/genetics , Translocation, Genetic , Aged , Amino Acid Sequence , Base Sequence , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 12 , Gene Fusion , Humans , Male , ETS Translocation Variant 6 Protein
10.
Rinsho Ketsueki ; 57(10): 2241-2249, 2016.
Article in Japanese | MEDLINE | ID: mdl-27795536

ABSTRACT

Adoptive immunotherapy using genetically modified T-cells is an emerging and promising treatment modality for various malignant diseases. The technology involves engineering of T-cells armed with well-characterized receptors such as T-cell receptors or chimeric antigen receptors. The latter is comprised of antibody/ligand and intracellular signaling domains. These molecules can be further modified to enhance their affinity, specificity, and several other functions. The success of adoptive immunotherapy is rooted in the application of extensive insights derived from allogeneic hematopoietic stem cell transplantations (HSCT). Herein, the historical perspectives of gene-modified T-cell therapy are discussed by comparison with the evolution of allogeneic HSCT. Furthermore, the prospects for the development and improvement of these powerful therapeutic methods are also highlighted.


Subject(s)
Cell- and Tissue-Based Therapy , Genetic Therapy , Leukemia/therapy , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Animals , Humans , Immunotherapy, Adoptive , Receptors, Antigen, T-Cell/genetics
11.
Cancer Sci ; 106(11): 1576-81, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26353084

ABSTRACT

It is still a matter of debate whether detection of Epstein-Barr virus (EBV) DNA in pretreatment serum has clinical implications for diffuse large B-cell lymphoma. For this study, we measured EBV DNA load in pretreatment serum from 127 diffuse large B-cell lymphoma patients without any underlying immunodeficiency to evaluate its effects on clinical manifestations and prognosis. Anthracycline-based chemotherapy in combination with rituximab was given as initial therapy for 119 patients (94%). Epstein-Barr virus DNA was detected in 15 patients (12%), who were older (P = 0.005) and tended to be at a more advanced disease stage (P = 0.053). They showed significantly worse progression-free survival (PFS) and overall survival (OS) than other patients (P < 0.001 each). This effect remained significant (P = 0.004 and P = 0.027, respectively) after adjustment for age, lactate dehydrogenase, performance status, stage, and extranodal sites. The status of EBV-encoded small RNA in situ hybridization was known for 123 patients; 6 of 8 positive patients (75%) and 9 of 115 negative patients (8%) had detectable EBV DNA in pretreatment serum. While patients positive for EBV-encoded small RNA had significantly worse PFS and OS than negative patients (P = 0.001 and P = 0.029, respectively), EBV DNA detection in pretreatment serum was associated with poorer PFS and OS even for the 115 patients negative for EBV-encoded small RNA (P < 0.001 each). These findings suggest that EBV DNA detection in pretreatment serum may have an adverse prognostic impact for patients with diffuse large B-cell lymphoma.


Subject(s)
Biomarkers, Tumor/blood , DNA, Viral/blood , Epstein-Barr Virus Infections/complications , Lymphoma, Large B-Cell, Diffuse/virology , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Disease-Free Survival , Female , Herpesvirus 4, Human , Humans , In Situ Hybridization , Kaplan-Meier Estimate , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/mortality , Male , Middle Aged , Prognosis , Proportional Hazards Models , Reverse Transcriptase Polymerase Chain Reaction , Treatment Outcome
12.
Cytogenet Genome Res ; 146(4): 279-84, 2015.
Article in English | MEDLINE | ID: mdl-26517539

ABSTRACT

DEK-NUP214 gene fusion in acute myeloid leukemia (AML) is associated with poor prognosis. It is most often a sole translocation and more rarely observed as complex chromosomal forms. We describe an AML case with complex karyotype abnormalities involving chromosome bands 6p23, 6q13, 7p22, and 9q34. RNA sequencing analysis revealed that exon 17 of NUP214 (9q34) was fused to exon 2 of RAC1 (7p22). We also detected that the 5'-end of intron 1 of RAC1 was fused with the antisense strand of intron 5 of COL12A1 (6q13). RT-PCR analysis confirmed the expression of DEK-NUP214, NUP214-RAC1, RAC1-COL12A1, NUP214, and RAC1. These results suggest that the 5'- and 3'-ends of NUP214 from the breakpoint in the same locus were fused to RAC1 and DEK, respectively, and the 5'-end of RAC1 was fused to COL12A1. The reading frame of NUP214 was not matched with RAC1; however, high expression of the RAC1 protein was detected by Western blotting. This study identifies the variant complex fusion genesNUP214-RAC1 and RAC1- COL12A1 in a case of AML.


Subject(s)
Chromosomes, Human , Collagen Type XII/genetics , Leukemia, Myeloid, Acute/genetics , Nuclear Pore Complex Proteins/genetics , Translocation, Genetic , rac1 GTP-Binding Protein/genetics , Adult , Humans , Male , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, RNA , Spectral Karyotyping
13.
Nature ; 459(7247): 712-6, 2009 Jun 04.
Article in English | MEDLINE | ID: mdl-19412163

ABSTRACT

A20 is a negative regulator of the NF-kappaB pathway and was initially identified as being rapidly induced after tumour-necrosis factor-alpha stimulation. It has a pivotal role in regulation of the immune response and prevents excessive activation of NF-kappaB in response to a variety of external stimuli; recent genetic studies have disclosed putative associations of polymorphic A20 (also called TNFAIP3) alleles with autoimmune disease risk. However, the involvement of A20 in the development of human cancers is unknown. Here we show, using a genome-wide analysis of genetic lesions in 238 B-cell lymphomas, that A20 is a common genetic target in B-lineage lymphomas. A20 is frequently inactivated by somatic mutations and/or deletions in mucosa-associated tissue lymphoma (18 out of 87; 21.8%) and Hodgkin's lymphoma of nodular sclerosis histology (5 out of 15; 33.3%), and, to a lesser extent, in other B-lineage lymphomas. When re-expressed in a lymphoma-derived cell line with no functional A20 alleles, wild-type A20, but not mutant A20, resulted in suppression of cell growth and induction of apoptosis, accompanied by downregulation of NF-kappaB activation. The A20-deficient cells stably generated tumours in immunodeficient mice, whereas the tumorigenicity was effectively suppressed by re-expression of A20. In A20-deficient cells, suppression of both cell growth and NF-kappaB activity due to re-expression of A20 depended, at least partly, on cell-surface-receptor signalling, including the tumour-necrosis factor receptor. Considering the physiological function of A20 in the negative modulation of NF-kappaB activation induced by multiple upstream stimuli, our findings indicate that uncontrolled signalling of NF-kappaB caused by loss of A20 function is involved in the pathogenesis of subsets of B-lineage lymphomas.


Subject(s)
Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Gene Silencing , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/physiopathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Animals , Apoptosis/physiology , Cell Line , DNA-Binding Proteins , Gene Expression , Genome/genetics , Humans , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3
14.
Blood ; 119(23): 5492-501, 2012 Jun 07.
Article in English | MEDLINE | ID: mdl-22529286

ABSTRACT

Targeted T-cell therapy is a potentially less toxic strategy than allogeneic stem cell transplantation for providing a cytotoxic antileukemic response to eliminate leukemic stem cells (LSCs) in acute myeloid leukemia (AML). However, this strategy requires identification of leukemia-associated antigens that are immunogenic and exhibit selective high expression in AML LSCs. Using microarray expression analysis of LSCs, hematopoietic cell subpopulations, and peripheral tissues to screen for candidate antigens, cyclin-A1 was identified as a candidate gene. Cyclin-A1 promotes cell proliferation and survival, has been shown to be leukemogenic in mice, is detected in LSCs of more than 50% of AML patients, and is minimally expressed in normal tissues with exception of testis. Using dendritic cells pulsed with a cyclin-A1 peptide library, we generated T cells against several cyclin-A1 oligopeptides. Two HLA A*0201-restricted epitopes were further characterized, and specific CD8 T-cell clones recognized both peptide-pulsed target cells and the HLA A*0201-positive AML line THP-1, which expresses cyclin-A1. Furthermore, cyclin-A1-specific CD8 T cells lysed primary AML cells. Thus, cyclin-A1 is the first prototypic leukemia-testis-antigen to be expressed in AML LSCs. The pro-oncogenic activity, high expression levels, and multitude of immunogenic epitopes make it a viable target for pursuing T cell-based therapy approaches.


Subject(s)
Cyclin A1/genetics , Cyclin A1/immunology , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/immunology , Neoplastic Stem Cells/immunology , Antigens, Neoplasm/analysis , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line , Cell Line, Tumor , Cyclin A1/analysis , Epitope Mapping , HLA-A2 Antigen/immunology , Humans , Male , Neoplastic Stem Cells/metabolism , Testis/immunology
15.
J Infect Chemother ; 20(12): 774-7, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25179391

ABSTRACT

Varicella, characterized by a vesicular rash, occurs primarily in young children. Although older individuals can also be affected or vaccinated, outbreaks among adults are rare. We investigated a small outbreak of varicella in B-cell lymphoma patients for elucidation of risk factor of the disease. We experienced four cases of varicella after an index herpes zoster case. All varicella cases were confirmed varicella zoster virus (VZV) infection by PCR. All varicella cases occurred in diffuse large B-cell lymphoma patients receiving rituximab-containing chemotherapy. On the other hand, only three of the 18 non-varicella patients in the same room were receiving rituximab-containing chemotherapy (P = 0.005). All varicella patients had detectable serum anti-varicella zoster virus IgG antibodies before chemotherapy. Even in the presence of neutralizing antibodies to the virus, lymphoma patients treated with rituximab-containing chemotherapy can possibly become re-infected with varicella. These findings suggest that zoster patients should be strictly isolated in hematology and oncology ward, and prophylactic acyclovir should be considered for such patients when exposed to zoster/varicella.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/adverse effects , Antineoplastic Agents/adverse effects , Chickenpox/etiology , Cross Infection/virology , Disease Outbreaks , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/virology , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Antineoplastic Agents/therapeutic use , Chickenpox/virology , Cross Infection/etiology , Female , Humans , Male , Middle Aged , Rituximab
16.
Int J Hematol ; 118(2): 252-266, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37310580

ABSTRACT

Relapsed leukemia after allogeneic hematopoietic stem cell transplantation (allo-HSCT) remains a significant challenge, with the re-emergence of the primary disease being the most frequent cause of death. Human leukocyte antigen (HLA)-DPB1 mismatch occurs in approximately 70% of unrelated allo-HSCT cases, and targeting mismatched HLA-DPB1 is considered reasonable for treating relapsed leukemia following allo-HSCT if performed under proper conditions. In this study, we established several clones restricted to HLA-DPB1*02:01, -DPB1*04:02, and -DPB1*09:01 from three patients who underwent HLA-DPB1 mismatched allo-HSCT using donor-derived alloreactive T cells primed to mismatched HLA-DPB1 in the recipient's body after transplantation. A detailed analysis of the DPB1*09:01-restricted clone 2A9 showed reactivity against various leukemia cell lines and primary myeloid leukemia blasts, even with low HLA-DP expression. T cell receptor (TCR)-T cells derived from clone 2A9 retained the ability to trigger HLA-DPB1*09:01-restricted recognition and lysis of various leukemia cell lines in vitro. Our study demonstrated that the induction of mismatched HLA-DPB1 specific T cell clones from physiologically primed post-allo-HSCT alloreactive CD4+ T cells and the redirection of T cells with cloned TCR cDNA by gene transfer are feasible as techniques for future adoptive immunotherapy.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Leukemia , Humans , CD4-Positive T-Lymphocytes , HLA-DP beta-Chains/genetics , HLA-DP beta-Chains/metabolism , Transplantation, Homologous , Hematopoietic Stem Cell Transplantation/methods , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism
17.
Nagoya J Med Sci ; 85(4): 779-796, 2023 Nov.
Article in English | MEDLINE | ID: mdl-38155626

ABSTRACT

Human leukocyte antigen (HLA)-DPB1 antigens are mismatched in approximately 70% of allogeneic hematopoietic stem cell transplantations (allo-HSCT) from HLA 10/10 matched unrelated donors. HLA-DP-mismatched transplantation was shown to be associated with an increase in acute graft-versus-host disease (GVHD) and a decreased risk of leukemia relapse due to the graft-versus-leukemia (GVL) effect. Immunotherapy targeting mismatched HLA-DP is considered reasonable to treat leukemia following allo-HCT if performed under non-inflammatory conditions. Therefore, we isolated CD4+ T cell clones that recognize mismatched HLA-DPB1 from healthy volunteer donors and generated T cell receptor (TCR)-gene-modified T cells for future clinical applications. Detailed analysis of TCR-T cells expressing TCR from candidate clone #17 demonstrated specificity to myeloid and monocytic leukemia cell lines that even expressed low levels of targeted HLA-DP. However, they did not react to non-hematopoietic cell lines with a substantial level of targeted HLA-DP expression, suggesting that the TCR recognized antigenic peptide is only present in some hematopoietic cells. This study demonstrated that induction of T cells specific for HLA-DP, consisting of hematopoietic cell lineage-derived peptide and redirection of T cells with cloned TCR cDNA by gene transfer, is feasible when using careful specificity analysis.


Subject(s)
Hematopoietic Stem Cell Transplantation , Leukemia , Humans , T-Lymphocytes , Transplantation, Homologous , Leukemia/therapy , HLA-DP beta-Chains/genetics , Chronic Disease , Recurrence , Peptides , Receptors, Antigen, T-Cell/genetics
18.
Cancer Sci ; 103(2): 350-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22049994

ABSTRACT

FMS-related tyrosine kinase 3 (FLT3) is a class III receptor tyrosine kinase that plays important roles in hematopoiesis, including early progenitors and dendritic cell development. FLT3 is expressed at high levels in 70-100% of cases of AML and in virtually all cases of B-lineage acute lymphoblastic leukemia. FLT3 is regarded as a molecular target in the development of novel therapies for acute leukemia patients. Currently, many small-molecule FLT3 inhibitors have been developed, but clinical trials have resulted in limited antileukemia effects because of off-target toxicities and drug resistance. The development of anti-FLT3 Abs might overcome these difficulties and enhance the antileukemia efficacy of FLT3 inhibitors. In the present study, we demonstrate the isolation of novel human mAbs against FLT3 with antagonistic or agonistic activities. An antagonistic Ab, designated A2, continuously inhibits FLT3 ligand (FL)-induced phosphorylation of FLT3 and MAPK. A2 cooperatively induces apoptosis with daunorubicin, even in the presence of FL. An agonistic Ab, designated 3E6, surprisingly induces the phosphorylation of FLT3 and MAPK, and supports the growth of a factor-dependent cell line independently of FL addition. In addition, A2 showed complement-dependent cytotoxicity activity, but was devoid of Ab-dependent cell mediated cytotoxicity. Finally, we evaluated Ab internalization in a cell line. Immunofluorescence and flow cytometry analyses revealed that A2 is efficiently internalized. Collectively, these data demonstrate that A2 is a potent human Ab that might be capable of delivering cytotoxic reagents and that has antagonistic effects on FLT3 signaling. In addition, 3E6 might be a potential scaffold for novel dendritic cell-based immunotherapies.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Leukemia, Myeloid, Acute/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , fms-Like Tyrosine Kinase 3/immunology , fms-Like Tyrosine Kinase 3/metabolism , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Daunorubicin/pharmacology , Humans , Leukemia, Myeloid, Acute/genetics , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Signal Transduction/drug effects , fms-Like Tyrosine Kinase 3/agonists , fms-Like Tyrosine Kinase 3/antagonists & inhibitors
19.
Blood ; 115(15): 3158-61, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20124217

ABSTRACT

We investigated human leukocyte antigen (HLA) expression on leukemic cells derived from patients at diagnosis and relapse after hematopoietic stem cell transplantation (HSCT) using flow cytometry with locus-specific antibodies. Two of 3 patients who relapsed after HLA-haploidentical HSCT demonstrated loss of HLA alleles in leukemic cells at relapse; on the other hand, no loss of HLA alleles was seen in 6 patients who relapsed after HLA-identical HSCT. Single-nucleotide polymorphism array analyses of sorted leukemic cells further revealed the copy number-neutral loss of heterozygosity, namely, acquired uniparental disomy on the short arm of chromosome 6, resulting in the total loss of the mismatched HLA haplotype. These results suggest that the escape from immunosurveillance by the loss of mismatched HLA alleles may be a crucial mechanism of relapse after HLA-haploidentical HSCT. Accordingly, the status of mismatched HLA on relapsed leukemic cells should be checked before donor lymphocyte infusion.


Subject(s)
HLA Antigens/immunology , Haploidy , Hematopoietic Stem Cell Transplantation , Histocompatibility Testing , Leukemia/immunology , Uniparental Disomy/genetics , Blast Crisis/genetics , Blast Crisis/immunology , Child , Chromosomes, Human, Pair 6/genetics , Cytotoxicity, Immunologic , Humans , Isoantigens/immunology , Recurrence , T-Lymphocytes, Cytotoxic/immunology , Tissue Donors
20.
Blood ; 115(19): 3869-78, 2010 May 13.
Article in English | MEDLINE | ID: mdl-20071660

ABSTRACT

The adoptive transfer of donor T cells that recognize recipient minor histocompatibility antigens (mHAgs) is a potential strategy for preventing or treating leukemic relapse after allogeneic hematopoietic cell transplantation (HCT). A total of 7 patients with recurrent leukemia after major histocompatibility complex (MHC)-matched allogeneic HCT were treated with infusions of donor-derived, ex vivo-expanded CD8(+) cytotoxic T lymphocyte (CTL) clones specific for tissue-restricted recipient mHAgs. The safety of T-cell therapy, in vivo persistence of transferred CTLs, and disease response were assessed. Molecular characterization of the mHAgs recognized by CTL clones administered to 3 patients was performed to provide insight into the antileukemic activity and safety of T-cell therapy. Pulmonary toxicity of CTL infusion was seen in 3 patients, was severe in 1 patient, and correlated with the level of expression of the mHAg-encoding genes in lung tissue. Adoptively transferred CTLs persisted in the blood up to 21 days after infusion, and 5 patients achieved complete but transient remissions after therapy. The results of these studies illustrate the potential to selectively enhance graft-versus-leukemia activity by the adoptive transfer of mHAg-specific T-cell clones and the challenges for the broad application of this approach in allogeneic HCT. This study has been registered at http://clinicaltrials.gov as NCT00107354.


Subject(s)
Graft vs Leukemia Effect/immunology , Hematopoietic Stem Cell Transplantation , Leukemia/therapy , Minor Histocompatibility Antigens/immunology , Neoplasm Recurrence, Local , T-Lymphocytes/transplantation , Adoptive Transfer , Adult , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cytotoxicity, Immunologic , Dermis/cytology , Dermis/immunology , Fibroblasts/immunology , Flow Cytometry , Graft vs Host Disease , Humans , Immunoenzyme Techniques , Leukemia/immunology , Middle Aged , Myelodysplastic Syndromes/immunology , Myelodysplastic Syndromes/therapy , T-Lymphocytes, Cytotoxic , Treatment Outcome , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL