Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Blood Adv ; 4(19): 4945-4954, 2020 10 13.
Article in English | MEDLINE | ID: mdl-33049054

ABSTRACT

The results from the RATIFY trial (ClinicalTrials.gov: NCT00651261; CALGB 10603) showed that midostaurin combined with standard chemotherapy significantly improved outcomes in patients with FMS-like tyrosine kinase 3 (FLT3)-mutated acute myeloid leukemia (AML), compared with placebo. In this post hoc subgroup analysis from the trial, we evaluated the impact of midostaurin in 163 patients with FLT3-tyrosine kinase domain (TKD) mutations. At a median follow-up of 60.7 months (95% CI, 55.0-70.8), the 5-year event-free survival (EFS) rate was significantly higher in patients treated with midostaurin than in those treated with placebo (45.2% vs 30.1%; P = .044). A trend toward improved disease-free survival was also observed with midostaurin (67.3% vs 53.4%; P = .089), whereas overall survival (OS) was similar in the 2 groups. Patients with AML and NPM1mut/FLT3-TKDmut or core binding factor (CBF)-rearranged/FLT3-TKDmut genotypes had significantly prolonged OS with or without censoring at hematopoietic cell transplantation (HCT), compared with NPM1WT/CBF-negative AMLs. The multivariable model for OS and EFS adjusted for allogeneic HCT in first complete remission as a time-dependent covariable, revealed NPM1 mutations and CBF rearrangements as significant favorable factors. These data show that NPM1 mutations or CBF rearrangements identify favorable prognostic groups in patients with FLT3-TKD AMLs, independent of other factors, also in the context of midostaurin treatment.


Subject(s)
Leukemia, Myeloid, Acute , fms-Like Tyrosine Kinase 3 , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Mutation , Nucleophosmin , Staurosporine/analogs & derivatives , Staurosporine/therapeutic use , fms-Like Tyrosine Kinase 3/genetics
3.
Biol Blood Marrow Transplant ; 21(3): 489-95, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25529382

ABSTRACT

Hematopoietic stem cell transplantation (HSCT) remains the only curative treatment in patients with higher risk myelodysplastic syndrome (MDS), but the choice of the optimal alternative stem cell source is still a subject of debate in patients lacking an HLA-matched sibling donor. Here, we report on a large series of patients with MDS (N = 631) transplanted either with mobilized peripheral stem cells (PBs) from unrelated donors (n = 502) or with umbilical cord blood transplant (UCB, n = 129) as alternative grafts after reduced-intensity conditioning. Neutrophil engraftment was higher after PB (98% versus 78%, P < .0001). Acute graft-versus-host disease (GVHD) was similar after PB (31%) and UCB (29%), and chronic GVHD incidence was higher after PB (41% versus 23%). Two-year nonrelapse mortality was lower after PB (31% versus 42% P = .03). There was a better overall survival (OS) and disease-free survival (DFS) after PB (49% ± 2% versus 30% ± 4%, P < .0001 and 44% ± 2% versus 28% ± 4%, P < .0001). Multivariate analysis confirmed the advantage of PB for treatment-related mortality, OS, and DFS, whereas relative risk of chronic GVHD was similar. A multivariate analysis comparing PB from a 10/10 HLA-matched donor, PB from a 9/10 HLA-matched donor, and UCB showed an advantage on treatment-related mortality, DFS, and OS only in 10/10 PB. We conclude that in MDS patients lacking an HLA-matched sibling donor, PB from a 10/10 HLA-matched unrelated donor is the preferred source of hematopoietic stem cells. HLA-mismatched unrelated donor or cord blood seem to give similar inferior results except for neutrophil engraftment, which is delayed after UCB.


Subject(s)
Cord Blood Stem Cell Transplantation , Graft vs Host Disease , Myelodysplastic Syndromes , Peripheral Blood Stem Cell Transplantation , Transplantation Conditioning , Unrelated Donors , Acute Disease , Adult , Aged , Allografts , Disease-Free Survival , Europe , Female , Graft vs Host Disease/mortality , Graft vs Host Disease/prevention & control , Humans , Male , Middle Aged , Myelodysplastic Syndromes/mortality , Myelodysplastic Syndromes/therapy , Survival Rate
4.
Clin Lymphoma Myeloma Leuk ; 14 Suppl: S42-5, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25486954

ABSTRACT

The selection of a treatment strategy before allogeneic hematopoietic stem cell transplant (HSCT) for myelodysplastic syndrome is a delicate process. The expected relapse risk and nonrelapse mortality after HSCT and the response rates to the pretransplant strategies all play a role in this process. Fit patients younger than 60 to 65 years with > 10% marrow blasts and without high-risk cytogenetic abnormalities should be seriously considered for intensive chemotherapy (ICT) to reduce tumor load before HSCT. Other patients up to the age of 75 years may be considered for hypomethylating agent therapy before transplant. Patients with high-risk cytogenetic abnormalities should be treated in investigational protocols if they are not candidates for ICT.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Hematopoietic Stem Cell Transplantation , Myelodysplastic Syndromes/therapy , DNA Methylation , Decision Making , Humans , Myelodysplastic Syndromes/genetics , Remission Induction , Risk Factors , Transplantation Conditioning , Transplantation, Homologous
5.
Am J Hematol ; 89(2): 174-80, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24123244

ABSTRACT

In chronic lymphocytic leukemia (CLL) medical progress is driven by clinical studies with relapse-free survival (RFS) as the primary endpoint. The randomized EBMT-Intergroup trial compared high-dose therapy and autologous stem cell transplantation (ASCT) to observation and demonstrated a substantial improvement of RFS without showing improved overall survival for the transplant arm. Here we report quality of life (QoL) information of the first 3 years following randomization from that study. The main objective was to assess the impact of treatment on QoL over time. Two secondary analyses were performed to further investigate the impact of ASCT and relapse on QoL. In the primary analysis, we demonstrate an adverse impact of ASCT on QoL which was largest at 4 months and continued throughout the first year after randomization. Further, we demonstrated a sustained adverse impact of relapse on QoL which worsened over time. Despite better disease control by ASCT the side effects thus turned the net effect towards inferior QoL in the first year and comparable QoL in the following 2 years after randomization. This study emphasizes the importance of information concerning QoL impacts when patients are counseled about treatments aimed at improving RFS in the absence of a survival benefit.


Subject(s)
Hematopoietic Stem Cell Transplantation , Leukemia, Lymphocytic, Chronic, B-Cell/psychology , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Quality of Life , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Female , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/mortality , Male , Middle Aged , Recurrence , Surveys and Questionnaires , Transplantation Conditioning , Transplantation, Autologous
6.
Leuk Lymphoma ; 54(3): 472-5, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22916713

ABSTRACT

An international working group within the European LeukemiaNet gathered, aiming to determine the role of flow cytometry (FC) in myelodysplastic syndromes (MDS). It was agreed that FC has a substantial application in disease characterization, diagnosis and prognosis. FC may also be useful in predicting treatment responses and monitoring novel and standard therapeutic regimens. In this article the rationale is discussed that flow cytometry should be integrated as a part of diagnostic and prognostic scoring systems in MDS.


Subject(s)
Flow Cytometry/methods , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/therapy , Outcome Assessment, Health Care/methods , Humans , International Agencies , Myelodysplastic Syndromes/classification , Prognosis , Reproducibility of Results , Sensitivity and Specificity , Societies, Scientific
7.
Cancer Res ; 71(15): 5111-22, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21659460

ABSTRACT

Tumor relapses remain a serious problem after allogeneic stem cell transplantation (alloSCT), despite the long-term persistence of minor histocompatibility antigen (MiHA)-specific memory CD8(+) T cells specific for the tumor. We hypothesized that these memory T cells may lose their function over time in transplanted patients. Here, we offer functional and mechanistic support for this hypothesis, based on immune inhibition by programmed death-1 (PD-1) expressed on MiHA-specific CD8(+) T cells and the associated role of the PD-1 ligand PD-L1 on myeloid leukemia cells, especially under inflammatory conditions. PD-L1 was highly upregulated on immature human leukemic progenitor cells, whereas costimulatory molecules such as CD80 and CD86 were not expressed. Thus, immature leukemic progenitor cells seemed to evade the immune system by inhibiting T-cell function via the PD-1/PD-L1 pathway. Blocking PD-1 signaling using human antibodies led to elevated proliferation and IFN-γ production of MiHA-specific T cells cocultured with PD-L1-expressing leukemia cells. Moreover, patients with relapsed leukemia after initial MiHA-specific T-cell responses displayed high PD-L1 expression on CD34(+) leukemia cells and increased PD-1 levels on MiHA-specific CD8(+) T cells. Importantly, blocking PD-1/PD-L1 interactions augment proliferation of MiHA-specific CD8(+) memory T cells from relapsed patients. Taken together, our findings indicate that the PD-1/PD-L pathway can be hijacked as an immune escape mechanism in hematological malignancies. Furthermore, they suggest that blocking the PD-1 immune checkpoint offers an appealing immunotherapeutic strategy following alloSCT in patients with recurrent or relapsed disease.


Subject(s)
Antigens, CD/physiology , Apoptosis Regulatory Proteins/physiology , Hematopoietic Stem Cell Transplantation , Immunologic Memory , Leukemia, Myeloid/immunology , Neoplasm Proteins/physiology , T-Lymphocyte Subsets/immunology , Tumor Escape/physiology , Apoptosis Regulatory Proteins/antagonists & inhibitors , B7-1 Antigen/biosynthesis , B7-2 Antigen/biosynthesis , B7-H1 Antigen , Coculture Techniques , Gene Expression Profiling , Gene Expression Regulation, Leukemic/drug effects , Humans , Inflammation , Interferon-gamma/pharmacology , Leukemia, Myeloid/pathology , Leukemia, Myeloid/surgery , Minor Histocompatibility Antigens/immunology , Neoplasm Proteins/antagonists & inhibitors , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , Programmed Cell Death 1 Receptor , Receptors, Purinergic P2X5/immunology , Recurrence , T-Cell Antigen Receptor Specificity , Transplantation, Homologous , Tumor Escape/immunology , Tumor Necrosis Factor-alpha/pharmacology
8.
PLoS One ; 5(2): e9221, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20169160

ABSTRACT

Immunotherapy based on natural killer (NK) cell infusions is a potential adjuvant treatment for many cancers. Such therapeutic application in humans requires large numbers of functional NK cells that have been selected and expanded using clinical grade protocols. We established an extremely efficient cytokine-based culture system for ex vivo expansion of NK cells from hematopoietic stem and progenitor cells from umbilical cord blood (UCB). Systematic refinement of this two-step system using a novel clinical grade medium resulted in a therapeutically applicable cell culture protocol. CD56(+)CD3(-) NK cell products could be routinely generated from freshly selected CD34(+) UCB cells with a mean expansion of >15,000 fold and a nearly 100% purity. Moreover, our protocol has the capacity to produce more than 3-log NK cell expansion from frozen CD34(+) UCB cells. These ex vivo-generated cell products contain NK cell subsets differentially expressing NKG2A and killer immunoglobulin-like receptors. Furthermore, UCB-derived CD56(+) NK cells generated by our protocol uniformly express high levels of activating NKG2D and natural cytotoxicity receptors. Functional analysis showed that these ex vivo-generated NK cells efficiently target myeloid leukemia and melanoma tumor cell lines, and mediate cytolysis of primary leukemia cells at low NK-target ratios. Our culture system exemplifies a major breakthrough in producing pure NK cell products from limited numbers of CD34(+) cells for cancer immunotherapy.


Subject(s)
Antigens, CD34/immunology , Cell Proliferation , Cytotoxicity, Immunologic/immunology , Killer Cells, Natural/immunology , CD56 Antigen/immunology , Cell Differentiation/immunology , Cell Line, Tumor , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Fetal Blood/cytology , Flow Cytometry , Humans , Immunophenotyping , Immunotherapy, Adoptive/methods , Interferon-gamma/metabolism , K562 Cells , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Leukemia/immunology , Leukemia/pathology , Melanoma/immunology , Melanoma/pathology , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Stem Cells/cytology , Stem Cells/immunology , Time Factors
9.
J Leukoc Biol ; 87(6): 1029-39, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20181724

ABSTRACT

DC are professional APCs that initiate and regulate adaptive immune responses by interacting with naïve and memory T cells. Chemokines released by DC play an essential role in T cell recruitment and in the maintenance of antigen-specific T cell-DC conjugates. Here, we characterized the expression of the T cell-attracting chemokine CXCL16 by murine DC. We demonstrate that through alternative RNA splicing, DC not only express the previously characterized transmembrane CXCL16 isoform, which can be cleaved from the cell surface, but also a novel isoform lacking the transmembrane and cytoplasmic domains. Transfection of HEK293 cells shows that this novel isoform, termed CXCL16v, is not expressed on the cell membrane but is secreted as a protein of approximately 10 kDa. Quantitative PCR demonstrates that CXCL16v is broadly expressed in lymphoid and nonlymphoid tissues resembling the tissue distribution of DC. Indeed, CXCL16v mRNA is expressed significantly by spleen DC and BM-DC. Moreover, we show that mature DC have increased CXCL16v mRNA levels and express transmembrane and soluble CXCL16 proteins. Finally, we show that CXCL16v specifically attracts cells expressing the chemokine receptor CXCR6. Our data demonstrate that mature DC express secreted, transmembrane, and cleaved CXCL16 isoforms to recruit and communicate efficiently with CXCR6(+) lymphoid cells.


Subject(s)
Alternative Splicing , Chemokine CXCL6/genetics , Chemotactic Factors/metabolism , Dendritic Cells/metabolism , Gene Expression Regulation/physiology , Receptors, CXCR/genetics , Animals , Base Sequence , Cells, Cultured , Chemokine CXCL16 , Chromatography, Liquid , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunoblotting , Immunoenzyme Techniques , Kidney/cytology , Kidney/metabolism , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Protein Isoforms , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, CXCR6 , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , Tandem Mass Spectrometry
10.
J Clin Oncol ; 28(3): 405-11, 2010 Jan 20.
Article in English | MEDLINE | ID: mdl-20008642

ABSTRACT

PURPOSE: This study was performed to examine the characteristics of transplant activity for patients with myelodysplastic syndromes (MDS) older than 50 years within the European Group for Blood and Marrow Transplantation, and to evaluate the factors predicting outcome within this group of patients. PATIENTS AND METHODS: We performed a retrospective multicenter analysis of 1,333 MDS patients age 50 years or older who received transplantation within the EBMT since 1998. The median recipient age was 56 years, with 884 patients (66%) age 50 to 60 years and 449 (34%) patients older than 60 years. There were 811 HLA-matched sibling (61%) and 522 (39%) unrelated donor transplants. Five hundred patients (38%) received standard myeloablative conditioning (SMC), and 833 (62%) received reduced intensity conditioning (RIC). RESULTS: The 4-year estimate for overall survival of the whole cohort was 31%. On multivariate analysis, use of RIC (hazard ratio [HR], 1.44; 95% CI, 1.13 to 1.84; P < .01) and advanced disease stage at transplantation (HR, 1.51; 95% CI, 1.18 to 1.93; P < .01) were associated with an increased relapse rate. In contrast, advanced disease stage at transplantation (HR, 1.43; 95% CI, 1.13 to 1.79; P = .01), use of an unrelated donor (P = .03), and RIC (HR, 0.79; 95% CI, 0.65 to 0.97; P = .03) were independent variables associated with nonrelapse mortality. Advanced disease stage at transplantation (HR, 1.55; 95% CI, 1.32 to 1.83; P < .01) was the major independent variable associated with an inferior 4-year overall survival. CONCLUSION: Allogeneic hematopoietic stem-cell transplantation remains a potential curative therapeutic option for many older patients with MDS. In this analysis, disease stage at time of transplantation, but not recipient age or the intensity of the conditioning regimens, was the most important factor influencing outcomes.


Subject(s)
Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid, Acute/therapy , Myelodysplastic Syndromes/therapy , Age Factors , Aged , Humans , Middle Aged , Retrospective Studies , Transplantation, Homologous
11.
Blood ; 114(26): 5264-70, 2009 Dec 17.
Article in English | MEDLINE | ID: mdl-19812383

ABSTRACT

From 2002 to 2007, 103 patients with primary myelofibrosis or postessential thrombocythemia and polycythemia vera myelofibrosis and a median age of 55 years (range, 32-68 years) were included in a prospective multicenter phase 2 trial to determine efficacy of a busulfan (10 mg/kg)/fludarabine (180 mg/m(2))-based reduced-intensity conditioning regimen followed by allogeneic stem cell transplantation from related (n = 33) or unrelated donors (n = 70). All but 2 patients (2%) showed leukocyte and platelet engraftment after a median of 18 and 22 days, respectively. Acute graft-versus-host disease grade 2 to 4 occurred in 27% and chronic graft-versus-host disease in 43% of the patients. Cumulative incidence of nonrelapse mortality at 1 year was 16% (95% confidence interval, 9%-23%) and significantly lower for patients with a completely matched donor (12% vs 38%; P = .003). The cumulative incidence of relapse at 3 years was 22% (95% confidence interval, 13%-31%) and was influenced by Lille risk profile (low, 14%; intermediate, 22%; and high, 34%; P = .02). The estimated 5-year event-free and overall survival was 51% and 67%, respectively. In a multivariate analysis, age older than 55 years (hazard ratio = 2.70; P = .02) and human leukocyte antigen-mismatched donor (hazard ratio = 3.04; P = .006) remained significant factors for survival. The study was registered at www.clinicaltrials.gov as #NCT 00599547.


Subject(s)
Primary Myelofibrosis/surgery , Stem Cell Transplantation/methods , Transplantation Conditioning/methods , Adult , Aged , Busulfan/therapeutic use , Disease-Free Survival , Female , Graft vs Host Disease/epidemiology , Humans , Male , Middle Aged , Myeloablative Agonists/therapeutic use , Primary Myelofibrosis/mortality , Recurrence , Stem Cell Transplantation/adverse effects , Transplantation, Homologous , Vidarabine/analogs & derivatives , Vidarabine/therapeutic use
12.
J Immunother ; 32(6): 539-51, 2009.
Article in English | MEDLINE | ID: mdl-19483655

ABSTRACT

Alloreactive CD8+ T cells targeting minor histocompatibility antigens (MiHA) on malignant cells of the recipient play a pivotal role in graft-versus-tumor responses observed after allogeneic stem cell transplantation and donor lymphocyte infusion (DLI). However, these MiHA-specific CD8+ T-cell responses do not result in complete eradication of tumor cells in all patients. Furthermore, CD8+ memory T cells persisting after DLI do not always efficiently expand with recurrence of the disease. Adjuvant immunotherapy using dendritic cells (DC) loaded with hematopoietic-restricted MiHA may boost antitumor CD8+ T-cell immunity without inducing graft-versus-host disease. Here, we explored the use of mRNA-electroporated DC to stimulate MiHA-specific CD8+ T-cell responses. We demonstrate that electroporation of mature DC with P2X5 mRNA encoding for hematopoietic-restricted MiHA LRH-1 results in high expression of both mRNA and protein, and has no negative effect on the mature phenotype and migratory capacity of the DC. Furthermore, these DC can efficiently stimulate LRH-1-specific CD8+ effector T cells to proliferate and produce interferon-gamma. In addition, LRH-1-specific CD8+ memory T cells that are present in patient-derived peripheral blood mononuclear cells at long periods post-DLI can be effectively activated by stimulation with P2X5 mRNA-electroporated DC to proliferate and degranulate upon target cell recognition. These results indicate that adjuvant immunotherapy using DC electroporated with mRNA encoding hematopoietic-restricted MiHA mismatched between patients and donors may enhance the graft versus tumor response induced by stem cell transplantation and DLI.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/transplantation , Graft vs Leukemia Effect , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Leukemia, Myeloid, Acute/therapy , Receptors, Purinergic P2/immunology , Cell Degranulation/immunology , Dendritic Cells/immunology , Electroporation , Humans , Immunotherapy , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Minor Histocompatibility Antigens/immunology , RNA, Messenger/genetics , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2X5 , Stem Cell Transplantation
13.
Haematologica ; 94(8): 1124-34, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19546437

ABSTRACT

The myelodysplastic syndromes are a group of clonal hematopoietic stem cell diseases characterized by cytopenia(s), dysplasia in one or more cell lineages and increased risk of evolution to acute myeloid leukemia (AML). Recent advances in immunophenotyping of hematopoietic progenitor and maturing cells in dysplastic bone marrow point to a useful role for multiparameter flow cytometry (FCM) in the diagnosis and prognostication of myelodysplastic syndromes. In March 2008, representatives from 18 European institutes participated in a European LeukemiaNet (ELN) workshop held in Amsterdam as a first step towards standardization of FCM in myelodysplastic syndromes. Consensus was reached regarding standard methods for cell sampling, handling and processing. The group also defined minimal combinations of antibodies to analyze aberrant immunophenotypes and thus dysplasia. Examples are altered numbers of CD34(+) precursors, aberrant expression of markers on myeloblasts, maturing myeloid cells, monocytes or erythroid precursors and the expression of lineage infidelity markers. When applied in practice, aberrant FCM patterns correlate well with morphology, the subclassification of myelodysplastic syndromes, and prognostic scoring systems. However, the group also concluded that despite strong evidence for an impact of FCM in myelodysplastic syndromes, further (prospective) validation of markers and immunophenotypic patterns are required against control patient groups as well as further standardization in multi-center studies. Standardization of FCM in myelodysplastic syndromes may thus contribute to improved diagnosis and prognostication of myelodysplastic syndromes in the future.


Subject(s)
Flow Cytometry/methods , Myelodysplastic Syndromes/diagnosis , Antigens, CD/immunology , Flow Cytometry/standards , Humans , Immunophenotyping/methods , Myelodysplastic Syndromes/immunology , Reference Standards
14.
Cancer Immunol Immunother ; 58(3): 429-39, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18719914

ABSTRACT

CD8(+) T cells recognizing minor histocompatibility antigens (MiHA) on solid tumor cells may mediate effective graft-versus-tumor (GVT) reactivity after allogeneic stem cell transplantation (SCT). Previously, we identified LRH-1 as a hematopoietic-restricted MiHA encoded by the P2X5 gene. Here, we report that LRH-1 is aberrantly expressed on solid tumor cells. P2X5 mRNA expression is demonstrated in a significant portion of solid tumor cell lines, including renal cell carcinoma (RCC), melanoma, colorectal carcinoma, brain cancer and breast cancer. Importantly, P2X5 gene expression was also detected in a subset of primary solid tumor specimens derived from RCC, brain cancer and breast cancer patients. Furthermore, P2X5 expressing solid tumor cells can be effectively targeted by LRH-1-specific cytotoxic T lymphocytes under inflammatory conditions. The expression of HLA-B7 and CD54 on tumor cells increases upon cytokine stimulation resulting in improved T cell activation as observed by higher levels of degranulation and enhanced tumor cell lysis. Overall, hematopoietic-restricted MiHA LRH-1 is aberrantly expressed on solid tumor cells and may be used as target in GVT-specific immunotherapy after SCT.


Subject(s)
DNA-Binding Proteins/metabolism , Hematopoietic System/metabolism , Minor Histocompatibility Antigens/metabolism , Neoplasms/immunology , Receptors, Purinergic P2/metabolism , T-Lymphocytes/metabolism , Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/immunology , DNA-Binding Proteins/physiology , Genotype , Humans , Immunotherapy/methods , Intercellular Adhesion Molecule-1/biosynthesis , Microscopy, Fluorescence/methods , Neoplasms/metabolism , RNA, Messenger/metabolism , Receptors, Purinergic P2X5 , Stem Cell Transplantation , Transcription Factors/physiology , Transplantation, Homologous
15.
Blood ; 113(10): 2312-23, 2009 Mar 05.
Article in English | MEDLINE | ID: mdl-19074734

ABSTRACT

CD8(+) T cells recognizing minor histocompatibility antigens (MiHAs) on leukemic stem and progenitor cells play a pivotal role in effective graft-versus-leukemia reactivity after allogeneic stem cell transplantation (SCT). Previously, we identified a hematopoiesis-restricted MiHA, designated LRH-1, which is presented by HLA-B7 and encoded by the P2X5 purinergic receptor gene. We found that P2X5 is significantly expressed in CD34(+) leukemic subpopulations from chronic myeloid leukemia (CML) and acute myeloid leukemia (AML) patients. Here, we demonstrate that LRH-1-specific CD8(+) T-cell responses are frequently induced in myeloid leukemia patients following donor lymphocyte infusions. Patients with high percentages of circulating LRH-1-specific CD8(+) T cells had no or only mild graft-versus-host disease. Functional analysis showed that LRH-1-specific cytotoxic T lymphocytes (CTLs) isolated from 2 different patients efficiently target LRH-1-positive leukemic CD34(+) progenitor cells from both CML and AML patients, whereas mature CML cells are only marginally lysed due to down-regulation of P2X5. Furthermore, we observed that relative resistance to LRH-1 CTL-mediated cell death due to elevated levels of antiapoptotic XIAP could be overcome by IFN-gamma prestimulation and increased CTL-target ratios. These findings provide a rationale for use of LRH-1 as immunotherapeutic target antigen to treat residual or persisting myeloid malignancies after allogeneic SCT.


Subject(s)
DNA-Binding Proteins/immunology , Leukemia, Myeloid/immunology , Neoplastic Stem Cells/immunology , T-Lymphocytes, Cytotoxic/immunology , Transcription Factors/immunology , Adult , Antigens, CD34/immunology , Antigens, CD34/metabolism , Female , Flow Cytometry , Gene Expression , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation , Humans , Leukemia, Myeloid/genetics , Male , Middle Aged , RNA, Messenger/analysis , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2X5 , Reverse Transcriptase Polymerase Chain Reaction , X-Linked Inhibitor of Apoptosis Protein/biosynthesis , X-Linked Inhibitor of Apoptosis Protein/genetics
16.
Br J Haematol ; 141(6): 799-807, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18410452

ABSTRACT

Minor histocompatibility antigens (MiHA) selectively expressed by haematopoietic cells are attractive targets for specific immunotherapy after allogeneic stem cell transplantation (SCT). Previously, we described LRH-1 as a haematopoietic-lineage restricted MiHA that is capable of eliciting an allogeneic cytotoxic T-lymphocyte (CTL) response after SCT and donor lymphocyte infusion. Importantly, the gene encoding LRH-1, P2X5, is not expressed in prominent graft-versus-host-disease target tissues such as skin, liver and gut. Here, we investigate whether LRH-1-specific immunotherapy may be exploited for the treatment of lymphoid malignancies. We examined P2X5 mRNA expression in a large panel of patient samples and cell lines from different types of lymphoid malignancies by real-time quantitative reverse transcription polymerase chain reaction. P2X5 mRNA was highly expressed in malignant cells from all stages of lymphoid development. Furthermore, all LRH-1-positive lymphoid tumour cell lines were susceptible to LRH-1 CTL-mediated lysis in flow cytometry-based cytotoxicity assays. However, interferon-gamma production was low or absent after stimulation with some cell lines, possibly due to differences in activation thresholds for CTL effector functions. Importantly, primary cells from patients with lymphoid malignancies were effectively lysed by LRH-1-specific CTL. These findings indicate that MiHA LRH-1 is a potential therapeutic target for cellular immunotherapy of lymphoid malignancies.


Subject(s)
DNA-Binding Proteins/immunology , Leukemia, Lymphoid/immunology , Lymphoma, Non-Hodgkin/immunology , Receptors, Purinergic P2/biosynthesis , T-Lymphocytes, Cytotoxic/immunology , Transcription Factors/immunology , Cytotoxicity, Immunologic/immunology , Humans , Leukemia, Lymphoid/pathology , Lymphoma, Non-Hodgkin/pathology , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2X5 , Reverse Transcriptase Polymerase Chain Reaction/methods , Tumor Cells, Cultured
17.
Blood ; 108(2): 419-25, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16609072

ABSTRACT

The myelodysplastic syndromes (MDSs) are heterogeneous with respect to clinical characteristics, pathologic features, and cytogenetic abnormalities. This heterogeneity is a challenge for evaluating response to treatment. Therapeutic trials in MDS have used various criteria to assess results, making cross-study comparisons problematic. In 2000, an International Working Group (IWG) proposed standardized response criteria for evaluating clinically significant responses in MDS. These criteria included measures of alteration in the natural history of disease, hematologic improvement, cytogenetic response, and improvement in health-related quality of life. The relevance of the response criteria has now been validated prospectively in MDS clinical trials, and they have gained acceptance in research studies and in clinical practice. Because limitations of the IWG criteria have surfaced, based on practical and reported experience, some modifications were warranted. In this report, we present recommendations for revisions of some of the initial criteria.


Subject(s)
Myelodysplastic Syndromes/diagnosis , Practice Guidelines as Topic , Humans , Remission Induction , Treatment Outcome
18.
Eur J Immunol ; 32(10): 2748-58, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12355426

ABSTRACT

Human minor histocompatibility antigens (mHag) are target antigens of the graft-versus-leukemia response observed after allogeneic HLA-identical stem cell transplantation. We previously defined the molecular nature of the B cell lineage-specific mHag HB-1. The CTL epitope was identified as the decamer peptide EEKRGSLHVW presented in the context of HLA-B44. The HB-1 antigen is encoded by a locus of yet unknown function on chromosome 5q32. A single nucleotide polymorphism within this locus results in an amino acid change from histidine (H) to tyrosine (Y) at position P8 within the CTL epitope. Based on genomic information, we have developed a PCR-RFLP assay to perform HB-1 typing at the DNA level. We determined that the allelic frequency for the H and Y variant is 0.79 and 0.21, respectively. From these data, we calculated that the expected recipient disparity between HLA-B44-matched sibling pairs for HB-1H is 2.8%, whereas recipient disparity for HB-1Y is expected to be 12.4%. Therefore, we addressed whether the HB-1Y peptide is reciprocally immunogenic. We revealed that both peptide variants bind equally efficient to HLA-B44 molecules and that the H/Y substitution has no influence on formation of epitope precursor peptides by 20 S proteasome-mediated degradation. More directly, CTL recognizing the naturally presented HB-1Y peptide could be generated from a HB-1H homozygous donor using peptide-pulsed dendritic cells. Using a set of synthetic structurally related peptide variants, we found that the H/Y substitution has a major impact on TCR recognition by CTL specific for either of the HB-1 allelic homologues. HB-1 is the first human mHag described that induces bi-directional allogeneic CTL responses that may contribute to a specific graft-versus-leukemia response following allogeneic stem cell transplantation.


Subject(s)
Alleles , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/immunology , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Sequence , Base Sequence , Cell Line , Cysteine Endopeptidases/physiology , Epitopes, T-Lymphocyte , Genetic Structures , Humans , Minor Histocompatibility Antigens/chemistry , Molecular Sequence Data , Multienzyme Complexes/physiology , Polymorphism, Genetic , Proteasome Endopeptidase Complex , Receptors, Antigen, T-Cell, alpha-beta/immunology
19.
Blood ; 100(6): 1997-2004, 2002 Sep 15.
Article in English | MEDLINE | ID: mdl-12200358

ABSTRACT

Allogeneic hematopoietic stem cell transplantation is the only curative therapy for myelodysplasia (MDS). To identify factors influencing transplantation outcome, we studied 452 recipients of HLA-identical sibling transplants for MDS from 1989 to 1997, reported to the International Bone Marrow Transplant Registry. Patients with treatment-related MDS or unclassified MDS were excluded. Median age was 38 years (range, 2-64 years). Sixty percent had refractory anemia with excess blasts (n = 136) or with excess blasts in transformation (n = 136). Conditioning regimens included total body irradiation in 199 (44%) cases. Marrow was T-cell depleted for 58 (13%) transplants. Cumulative incidences of neutrophil engraftment, grades II-IV acute graft-versus-host disease (GVHD), and chronic GVHD were 91% (95% confidence interval [CI], 88%-93%), 36% (95% CI, 31%-40%), and 39% (95% CI, 33%-44%), respectively. Three-year transplantation-related mortality (TRM), relapse, disease-free survival, and overall survival rates were 37% (95% CI, 32%-42%), 23% (95% CI, 19%-27%), 40% (95% CI, 36%-45%), and 42% (95% CI, 37%-47%), respectively. Multivariate analyses showed that young age and platelet counts higher than 100 x 10(9)/L at transplantation were associated with lower TRM and higher disease-free and overall survival rates. Relapse incidence was higher in patients with high percentages of blasts in the marrow at transplantation or presentation, with high International Prognostic Scoring System scores at diagnosis, and with T-cell-depleted transplants. These findings indicate that transplantation from an HLA-identical sibling offers the possibility of long-term, disease-free survival to patients with MDS. Best candidates are younger patients with a low percentage of blasts and preserved platelet counts.


Subject(s)
Bone Marrow Transplantation/immunology , Myelodysplastic Syndromes/therapy , Adolescent , Adult , Bone Marrow Transplantation/adverse effects , Bone Marrow Transplantation/mortality , Child , Child, Preschool , Disease-Free Survival , Graft vs Host Disease , Hematologic Diseases/complications , Hematologic Diseases/mortality , Hematologic Diseases/therapy , Histocompatibility , Histocompatibility Testing , Humans , Middle Aged , Myelodysplastic Syndromes/complications , Myelodysplastic Syndromes/mortality , Nuclear Family , Prospective Studies , Retrospective Studies , Risk , Survival Analysis , Transplantation, Isogeneic/adverse effects , Transplantation, Isogeneic/immunology , Transplantation, Isogeneic/mortality
20.
Blood ; 99(12): 4370-8, 2002 Jun 15.
Article in English | MEDLINE | ID: mdl-12036864

ABSTRACT

In this multicenter retrospective study, the outcomes of 234 patients with myelodysplastic syndrome (MDS) who underwent transplantation between 1995 and 1999 from HLA-identical siblings were analyzed according to the hematopoietic stem cell source used, that is, bone marrow (BM, n = 132) or granulocyte colony-stimulating factor-mobilized peripheral blood progenitor cells (PBPCs, n = 102). There were 69 cases of refractory anemia (RA), 86 RA with excess blasts (RAEB), 75 RAEB in transformation (RAEB-t), and 4 unclassified MDS at diagnosis. The International Prognostic Scoring System was intermediate-2 or high in 104 of the 158 available scores. Multivariate analyses focused on transplantation-related mortality (TRM), 2-year treatment failure incidence, and survival. Use of PBPCs reduced the median duration of neutropenia and thrombocytopenia by 4 and 12 days, respectively. The incidence of acute GVHD was similar whatever the graft type used. Chronic GVHD was more likely to have occurred with PBPCs (odds ratio [OR], 1.62; 95% confidence interval [CI], 0.87-3.02). Two-year TRM was significantly reduced with PBPCs (relative risk [RR], 0.33; 95% CI, 0.15-0.73; P <.007), except for patients who had either RA or high-risk cytogenetics. The 2-year treatment failure incidence was significantly decreased with PBPCs, from 38% to 13% (RR, 0.22; 95% CI, 0.10-0.48; P <.001). Estimate of the 2-year event-free survival was 50% with PBPCs versus 39% with BM. In multivariate analysis, the outcome was significantly improved with PBPCs (RR, 0.27; 95% CI, 0.13-0.52; P <.001), except for patients with either RA or high-risk cytogenetics. In conclusion, PBPCs might be preferred for allogeneic transplantation in MDS patients at high risk for relapse on the basis of morphologic criteria because the use of this hematopoietic stem cell was associated with lower treatment failure incidence and improved survival.


Subject(s)
Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/drug effects , Myelodysplastic Syndromes/therapy , Transplantation Immunology , Adolescent , Adult , Bone Marrow Cells/cytology , Child , Child, Preschool , Female , Granulocyte Colony-Stimulating Factor/administration & dosage , Hematopoietic Stem Cell Transplantation/mortality , Histocompatibility Testing , Humans , Male , Middle Aged , Myelodysplastic Syndromes/mortality , Nuclear Family , Retrospective Studies , Survival Analysis , Transplantation, Isogeneic/immunology , Transplantation, Isogeneic/methods , Transplantation, Isogeneic/mortality , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...