Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Haematologica ; 108(8): 2080-2090, 2023 08 01.
Article in English | MEDLINE | ID: mdl-36794500

ABSTRACT

Therapy-resistant viral reactivations contribute significantly to mortality after hematopoietic stem cell transplantation. Adoptive cellular therapy with virus-specific T cells (VST) has shown efficacy in various single-center trials. However, the scalability of this therapy is hampered by laborious production methods. In this study we describe the in-house production of VST in a closed system (CliniMACS Prodigy® system, Miltenyi Biotec). In addition, we report the efficacy in 26 patients with viral disease following hematopoietic stem cell transplantation in a retrospective analysis (adenovirus, n=7; cytomegalovirus, n=8; Epstein-Barr virus, n=4; multi-viral, n=7). The production of VST was successful in 100% of cases. The safety profile of VST therapy was favorable (n=2 grade 3 and n=1 grade 4 adverse events; all three were reversible). A response was seen in 20 of 26 patients (77%). Responding patients had a significantly better overall survival than patients who did not respond (P<0.001). Virus-specific symptoms were reduced or resolved in 47% of patients. The overall survival of the whole cohort was 28% after 6 months. This study shows the feasibility of automated VST production and safety of application. The scalability of the CliniMACS Prodigy® device increases the accessibility of VST treatment.


Subject(s)
Epstein-Barr Virus Infections , Hematopoietic Stem Cell Transplantation , Virus Diseases , Humans , T-Lymphocytes , Epstein-Barr Virus Infections/therapy , Retrospective Studies , Herpesvirus 4, Human , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Virus Diseases/etiology , Virus Diseases/therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Stem Cell Transplantation
2.
Clin Infect Dis ; 68(8): 1406-1409, 2019 04 08.
Article in English | MEDLINE | ID: mdl-30357314

ABSTRACT

Here, we report the case of severe adenoviremia in a 7-year-old boy with highly-resistant, acute leukemia. A combined approach of αßTCR-CD19-depleted stem cell transplantation, enabling immunosuppression-free post-transplant care, and early transfer of adenovirus-specific donor T cells during aplasia resulted in rapid and complete clearance of the treatment-refractory adenoviremia.


Subject(s)
Adenovirus Infections, Human/therapy , Hematopoietic Stem Cell Transplantation , T-Lymphocytes/transplantation , T-Lymphocytes/virology , Viremia/therapy , Adenovirus Infections, Human/etiology , Antigens, CD19 , Child , Combined Modality Therapy , Fatal Outcome , Humans , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/virology
3.
Blood ; 125(12): 1986-94, 2015 Mar 19.
Article in English | MEDLINE | ID: mdl-25617426

ABSTRACT

Hematopoietic stem cell transplantation (HSCT) has improved over the last few decades. However, viral infections are often refractory to pharmacologic treatment and require alternative treatment strategies such as immunotherapy. Adenovirus (AdV) is th predominant disease-causing pathogen in pediatric HSCT. In a clinical trial, we analyzed safety and efficacy of ex vivo adoptive T-cell transfer (ACT) with hexon-specific T cells, predominantly of the T-helper cell 1 (Th1) phenotype, in 30 patients with AdV disease or viremia. ACT was feasible with no acute toxicities or significant onset of graft-versus-host disease. ACT led to in vivo antiviral immunity for up to 6 months with viral control, resulting in complete clearance of viremia in 86% of patients with antigen-specific T-cell responses. After ACT and a follow-up of 6 months, overall survival was markedly increased in responders (mean, 122 days; 15 survivors) compared with nonresponders who all died shortly after ACT (mean, 24 days; no survivors). AdV-related mortality was 100% in nonresponders compared with 9.5% in responders (≥1 log reduction of DNA copies per milliliter after ACT). In summary, ex vivo ACT of AdV-specific Th1 cells was well tolerated and led to successful and sustained restoration of T-cell immunity correlated with virologic response and protection from virus-related mortality. This cellular immunotherapy is a short-term available and broadly applicable treatment. The study is registered at European Union Clinical Trials Register as 2005-001092-35.


Subject(s)
Adenoviridae Infections/complications , Capsid Proteins/metabolism , Hematopoietic Stem Cell Transplantation/adverse effects , Immunotherapy, Adoptive/methods , T-Lymphocytes/cytology , Th1 Cells/cytology , Adenoviridae Infections/etiology , Adolescent , Adoptive Transfer , Adult , Child , Child, Preschool , Humans , Infant , Male , Middle Aged , Phenotype , Probability , T-Lymphocytes/immunology , Treatment Outcome , Young Adult
4.
Mol Ther ; 24(9): 1634-43, 2016 09.
Article in English | MEDLINE | ID: mdl-27380762

ABSTRACT

Prognosis of primary refractory and relapsed pediatric B-lineage acute lymphoblastic leukemia (ALL) is very poor. Relapse rates significantly correlate with persistent minimal residual disease (MRD). In MRD, favorable effector-target ratios prevail and thus this situation might be optimally suited for immunotherapy with antibodies recruiting immunological effector cells. We here report on the generation, preclinical characterization and first clinical application in B-lineage ALL of an Fc-optimized CD19 antibody. This third-generation antibody (4G7SDIE) mediated enhanced antibody-dependent cellular cytotoxicity (ADCC) against leukemic blasts with effector cells from healthy volunteers and B-lineage ALL patients. The antibody was produced in a university-owned production unit and was applied on a compassionate use basis to 14 pediatric patients with refractory and relapsed B-lineage ALL at the stage of MRD. In 10/14 patients, MRD was reduced by ≥ 1 log or below the patient-individual detection limit, and 5/14 patients have achieved ongoing complete molecular remission with a median leukemia-free survival of 428 days. Two additional patients died in complete molecular remission due to complications not related to antibody therapy. Besides profound in vivo B-cell depletion, side effects were negligible. A clinical phase 1/2 study to further assess the therapeutic activity of 4G7SDIE is in preparation.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, CD19 , Immunoglobulin Fc Fragments , Neoplasm, Residual/drug therapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adolescent , Antibodies, Monoclonal/pharmacology , Antibody-Dependent Cell Cytotoxicity , Antigens, CD19/immunology , Antigens, CD19/metabolism , Cell Line, Tumor , Child , Child, Preschool , Combined Modality Therapy , Drug Monitoring , Drug Resistance, Neoplasm , Female , Gene Expression , Humans , Immunoglobulin Fc Fragments/immunology , Kaplan-Meier Estimate , Male , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Retreatment , Treatment Outcome , Young Adult
5.
Blood ; 124(4): 628-37, 2014 Jul 24.
Article in English | MEDLINE | ID: mdl-24855206

ABSTRACT

Patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT) are threatened by potentially lethal viral manifestations like cytomegalovirus (CMV) reactivation. Because the success of today's virostatic treatment is limited by side effects and resistance development, adoptive transfer of virus-specific memory T cells derived from the stem cell donor has been proposed as an alternative therapeutic strategy. In this context, dose minimization of adoptively transferred T cells might be warranted for the avoidance of graft-versus-host disease (GVHD), in particular in prophylactic settings after T-cell-depleting allo-HSCT protocols. To establish a lower limit for successful adoptive T-cell therapy, we conducted low-dose CD8(+) T-cell transfers in the well-established murine Listeria monocytogenes (L.m.) infection model. Major histocompatibility complex-Streptamer-enriched antigen-specific CD62L(hi) but not CD62L(lo) CD8(+) memory T cells proliferated, differentiated, and protected against L.m. infections after prophylactic application. Even progenies derived from a single CD62L(hi) L.m.-specific CD8(+) T cell could be protective against bacterial challenge. In analogy, low-dose transfers of Streptamer-enriched human CMV-specific CD8(+) T cells into allo-HSCT recipients led to strong pathogen-specific T-cell expansion in a compassionate-use setting. In summary, low-dose adoptive T-cell transfer (ACT) could be a promising strategy, particularly for prophylactic treatment of infectious complications after allo-HSCT.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/immunology , Graft vs Host Disease/immunology , Immunotherapy, Adoptive , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Severe Combined Immunodeficiency/immunology , Adolescent , Animals , Cell Differentiation , Cell Proliferation , Child , Cytomegalovirus/isolation & purification , Cytomegalovirus Infections/metabolism , Cytomegalovirus Infections/therapy , Graft vs Host Disease/metabolism , Graft vs Host Disease/therapy , Hematopoietic Stem Cell Transplantation , Homeodomain Proteins/physiology , Humans , Immunization , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Ovalbumin/physiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Severe Combined Immunodeficiency/metabolism , Severe Combined Immunodeficiency/therapy , Transplantation, Homologous , Virus Activation
6.
Br J Haematol ; 169(1): 90-102, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25495919

ABSTRACT

Children with B cell malignancies refractory to standard therapy are known to have a poor prognosis and very limited treatment options. Here, we report on the treatment and follow-up of ten patients diagnosed with relapsed or refractory mature B-cell Non Hodgkin Lymphoma (B-NHL), Burkitt leukaemia (B-AL) or pre B-acute lymphoblastic leukaemia (pre B-ALL). All children were treated with FBTA05 (now designated Lymphomun), an anti-CD3 x anti-CD20 trifunctional bispecific antibody (trAb) in compassionate use. Within individual treatment schedules, Lymphomun was applied (a) after allogeneic stem cell transplantation (allo-SCT, n = 6) to induce sustained long-term remission, or (b) stand alone prior to subsequent chemotherapy to eradicate residual disease before allo-SCT (n = 4). Nine of ten children displayed a clinical response: three stable diseases (SD), one partial remission (PR) and five induced or sustained complete remissions (CR). Five of these nine responders died during follow-up. The other patients still maintain CR with a current overall survival of 874-1424 days (median: 1150 days). In conclusion, despite the dismal clinical prognosis of children refractory to standard therapy, immunotherapy with Lymphomun resulted in a favourable clinical outcome in this cohort of refractory paediatric patients.


Subject(s)
Antibodies, Bispecific/administration & dosage , Burkitt Lymphoma/therapy , Immunotherapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Stem Cell Transplantation , Adolescent , Allografts , Antibodies, Bispecific/adverse effects , Burkitt Lymphoma/mortality , Burkitt Lymphoma/pathology , Child , Child, Preschool , Disease-Free Survival , Female , Follow-Up Studies , Humans , Male , Neoplasm, Residual , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Survival Rate
7.
Br J Haematol ; 162(6): 802-7, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23829632

ABSTRACT

The treatment outcome of children with refractory acute leukaemia or relapse post-stem cell transplantation is dismal. We report 10 children (non-remission n = 7) who underwent a new haploidentical transplant approach utilizing unmanipulated bone marrow followed by CD6-depleted peripheral blood stem cells. Nine patients had successful engraftment and no evidence of leukaemia. Acute and chronic graft-versus-host-disease was observed in five and three patients, respectively; two patients died of treatment-related toxicity. Seven patients relapsed after 7 (range 3-34) months, however two patients are alive at 6·5 and 7·0 years. This approach provides anti-leukaemic activity even in heavily pre-treated children but long-term disease control requires further intervention.


Subject(s)
Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , Bone Marrow Transplantation/methods , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/immunology , Leukemia/surgery , Adolescent , Child , Child, Preschool , Graft vs Host Disease/immunology , Haploidy , Humans , Infant , Leukemia/immunology , Lymphocyte Depletion/methods , Neoplasm Recurrence, Local/surgery , Treatment Outcome
8.
Haematologica ; 98(9): 1388-96, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23716544

ABSTRACT

Current diagnostic approaches that characterize T-cell deficiency by analyzing diversity of T-cell receptor sequences effectuate limited informational gain about the actual restrictiveness. For deeper insight into T-cell receptor repertoires we developed next-generation-sequencing-spectratyping, which employs high coverage Roche/454 sequencing of T-cell receptor (ß)-chain amplicons. For automated analysis of high-throughput-sequencing data, we developed a freely available software, the TCR profiler. Gene usage, length, encoded amino acid sequence and sequence diversity of the complementarity determining region 3 were determined and comprehensively integrated into a novel complexity score. Repertoires of CD8(+) T cells from children with idiopathic or hepatitis-induced very severe aplastic anemia (n=7), children two months after bone marrow transplantation (n=7) and healthy controls (children n=5, adults n=5) were analyzed. Complexity scores clearly distinguished between healthy and diseased, and even between different immune deficiency states. The repertoire of aplastic anemia patients was dominated by public (i.e. present in more than one person) T-cell receptor clonotypes, whereas only 0.2% or 1.9% were public in normal children and adults, respectively. The CDR3 sequence ASSGVGFSGANVLT was highly prevalent in 3 cases of hepatitis-induced anemia (15-32% of all sequences), but was only low expressed in idiopathic aplastic anemia (2-5%, n=4) or healthy controls (<1%). Fifteen high frequent sequences were present exclusively in aplastic anemia patients. Next-generation-sequencing-spectratyping allows in-depth analysis of T-cell receptor repertoires and their restriction in clinical samples. A dominating clonotype was identified in hepatitis-induced anemia that may be associated with disease pathogenesis and several aplastic-anemia-associated, putatively autoreactive clonotypes were sequenced.


Subject(s)
Anemia, Aplastic/genetics , Complementarity Determining Regions/genetics , Hepatitis/genetics , High-Throughput Nucleotide Sequencing/methods , Receptors, Antigen, T-Cell/genetics , Severity of Illness Index , Adolescent , Adult , Amino Acid Sequence , Anemia, Aplastic/diagnosis , Child , Child, Preschool , Cohort Studies , Complementarity Determining Regions/chemistry , Hepatitis/diagnosis , Humans , Infant , Molecular Sequence Data , Prospective Studies
9.
J Clin Oncol ; 41(17): 3135-3148, 2023 06 10.
Article in English | MEDLINE | ID: mdl-36854071

ABSTRACT

PURPOSE: Patients with relapsed high-risk neuroblastoma (rHR-NB) have a poor prognosis. We hypothesized that graft-versus-neuroblastoma effects could be elicited by transplantation of haploidentical stem cells (haplo-SCT) exploiting cytotoxic functions of natural killer cells and their activation by the anti-GD2 antibody dinutuximab beta (DB). This phase I/II trial assessed safety, feasibility, and outcomes of immunotherapy with DB plus subcutaneous interleukin-2 (scIL2) after haplo-SCT in patients with rHR-NB. METHODS: Patients age 1-21 years underwent T-/B-cell-depleted haplo-SCT followed by DB and scIL2. The primary end point 'success of treatment' encompassed patients receiving six cycles, being alive 180 days after end of trial treatment without progressive disease, unacceptable toxicity, acute graft-versus-host-disease (GvHD) ≥grade 3, or extensive chronic GvHD. RESULTS: Seventy patients were screened, and 68 were eligible for immunotherapy. Median number of DB cycles was 6 (range, 1-9). Median number of scIL2 cycles was 3 (1-6). The primary end point was met by 37 patients (54.4%). Median observation time was 7.8 years. Five-year event-free survival (EFS) and overall survival from start of trial treatment were 43% (95% CI, 31 to 55) and 53% (95% CI, 41 to 65), respectively. Five-year EFS among patients in complete remission (CR; 52%; 95% CI, 31 to 69) or partial remission (44%; 95% CI, 27 to 60) before immunotherapy were significantly better compared with patients with nonresponse/mixed response/progressive disease (13%; 95% CI, 1 to 42; P = .026). Overall response rate in 43 patients with evidence of disease after haplo-SCT was 51% (22 patients), with 15 achieving CR (35%). Two patients developed GvHD grade 2 and 3 each. No unexpected adverse events occurred. CONCLUSION: DB therapy after haplo-SCT in patients with rHR-NB is feasible, with low risk of inducing GvHD, and results in long-term remissions likely attributable to increased antineuroblastoma activity by donor-derived effector cells.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Neuroblastoma , Humans , Infant , Child, Preschool , Child , Adolescent , Young Adult , Adult , Interleukin-2/therapeutic use , Neoplasm Recurrence, Local/therapy , Neoplasm Recurrence, Local/etiology , Neuroblastoma/drug therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Graft vs Host Disease/etiology
10.
J Clin Immunol ; 32(3): 438-40, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22354567

ABSTRACT

Allogeneic hematopoietic stem cell transplantation (HSCT) has not been a therapeutic option in ataxia telangiectasia (AT) due to overwhelming toxicity of conditioning in the context of the global DNA repair deficiency. Furthermore HSCT is unable to cure neurological involvement of AT. We report on a Turkish child with a Hyper IgM phenotype disorder, in which clinical aspects of AT were absent and thus, AT not diagnosed. He was transplanted with a reduced toxicity, but full intensity conditioning regimen comprising treosulfan, fludarabine and ATG. The peritransplant period was uneventful and the patient was discharged at day +57. 8 months after HSCT, the patient developed hepatopathy with monoclonal gammopathy of unclear significance and died due to hepatic failure and encephalopathy at the age of 32 months. Post mortem high throughput sequencing revealed a mutation in the ATM gene.


Subject(s)
Ataxia Telangiectasia/complications , Hematopoietic Stem Cell Transplantation , Hepatic Encephalopathy/complications , Transplantation, Homologous , Ataxia Telangiectasia/blood , Ataxia Telangiectasia/pathology , Child, Preschool , Fatal Outcome , Hepatic Encephalopathy/blood , Hepatic Encephalopathy/pathology , Humans , Immunoglobulins/blood , Male
11.
Blood ; 116(20): 4360-7, 2010 Nov 18.
Article in English | MEDLINE | ID: mdl-20625005

ABSTRACT

Cytomegalovirus (CMV) disease and infection refractory to antiviral treatment after allogeneic stem cell transplantation (allo-SCT) is associated with a high mortality. Adoptive transfer of CMV-specific T cells could reconstitute viral immunity after SCT and could protect from CMV-related complications. However, logistics of producing virus-specific T-cell grafts limited the clinical application. We treated 18 patients after allo-SCT from human leukocyte antigen-mismatched/haploidentical or human leukocyte antigen-matched unrelated donors with polyclonal CMV-specific T cells generated by ex vivo stimulation with pp65, followed by isolation of interferon-γ-producing cells. Patients with CMV disease or viremia refractory to antiviral chemotherapy or both were eligible for adoptive T-cell transfer and received a mean of 21 × 10³/kg pp65-specific T cells. In 83% of cases CMV infection was cleared or viral burden was significantly reduced, even in cases of CMV encephalitis (n = 2). Viral control was associated with in vivo expansion of CMV-specific T lymphocytes in 12 of 16 evaluable cases, resulting in reconstitution of antiviral T-cell responses, without graft-versus-host disease induction or acute side effects. Our findings indicate that the infusion of low numbers of CMV-specific T cells is safe, feasible, and effective as a treatment on demand for refractory CMV infection and CMV disease after allo-SCT.


Subject(s)
Adoptive Transfer/methods , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/therapy , Haploidy , Phosphoproteins/immunology , Stem Cell Transplantation , T-Lymphocytes/transplantation , Viral Matrix Proteins/immunology , Adolescent , Adult , Child , Child, Preschool , Cytomegalovirus Infections/virology , Feasibility Studies , Follow-Up Studies , Histocompatibility Testing , Humans , Middle Aged , Recurrence , T-Lymphocytes/immunology , Time Factors , Treatment Outcome
12.
Haematologica ; 97(9): 1304-11, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22315490

ABSTRACT

UNLABELLED: BACKGROUND Very severe aplastic anemia is characterized by a hypoplastic bone marrow due to destruction of CD34(+) stem cells by autoreactive T cells. Investigation of the pathomechanism by patient-specific gene expression analysis of the attacked stem cells has previously been impractical because of the scarcity of these cells at diagnosis. DESIGN AND METHODS: Employing unbiased RNA amplification, patient-specific gene expression profiling was carried out for CD34(+) cells from patients newly diagnosed with very severe aplastic anemia (n=13), refractory anemia (n=8) and healthy controls (n=10). These data were compared to profiles of myelodysplastic disease (n=55), including refractory anemia (n=18). To identify possible targets of autoimmune attack, presence of autoreactive antibodies was tested in pre-therapeutic sera of patients with very severe aplastic anemia (n=19). RESULTS: CD34(+) gene expression profiling distinguished between healthy controls, children with aplastic or refractory anemia and clonal disease. Interferon stimulated genes such as the apoptosis inducing death ligand TRAIL were strongly up-regulated in CD34(+) cells of patients with aplastic anemia, in particular in patients responding to immunosuppressive treatment. In contrast, mRNA expression of integrin GPVI and the integrin complexes GPIa/IIa, GPIIb/IIIa, GPIB/GPIX/GPV was significantly down-regulated and corresponding antibodies were detected in 7 of 11 profiled patients and in 11 of 19 aplastic anemia patients. CONCLUSIONS As a potential diagnostic tool, patient-specific gene expression profiling of CD34(+) stem cells made it possible to make the difficult differential diagnosis of most patients with aplastic and refractory anemia. Profiling indicated a prognostic correlation of TRAIL expression and patient benefit from immunosuppressive therapy. Downregulation of integrin expression and concurrent presence of autoreactive anti-integrin-antibodies suggested a previously unrecognized pathological role of integrins in aplastic anemia.


Subject(s)
Anemia, Aplastic/genetics , Anemia, Refractory/genetics , Antigens, CD34/genetics , Biomarkers, Tumor/genetics , Gene Expression Profiling , Integrins/genetics , TNF-Related Apoptosis-Inducing Ligand/genetics , Adolescent , Anemia, Aplastic/drug therapy , Anemia, Aplastic/pathology , Anemia, Refractory/drug therapy , Anemia, Refractory/pathology , Apoptosis , Case-Control Studies , Child , Child, Preschool , Female , Humans , Immunosuppressive Agents/therapeutic use , Male , Microarray Analysis , Prognosis
14.
Med Mycol ; 49 Suppl 1: S164-9, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20586680

ABSTRACT

The morbidity and mortality rates of invasive fungal infection in allogeneic stem cell recipients are still unacceptably high and have not been significantly improved by alternative antifungal strategies to date. Over the last few years, rapid methods for the clinical-scale generation of functionally active and well characterized antifungal T(H)1 cells have become available. In addition, current data on the use of donor-derived virus-specific T cells in allogeneic stem cell transplantation suggest that the risk of severe adverse events, in particular the risk of graft-versus-host disease, is negligible. Therefore, adoptive antifungal immunotherapeutic strategies should be evaluated in clinical trials. However, one has to recognize that these trials are only meaningful with sufficiently large and homogenous cohorts of patients and if the settings of adoptive antifungal immunotherapy are comparable. Ultimately, the strategy of adoptively transferring antifungal immune responses might improve the outcome in hematopoietic stem cell recipients suffering from invasive fungal infection.


Subject(s)
Hematopoietic Stem Cell Transplantation/adverse effects , Immunotherapy, Adoptive/methods , Mycoses/therapy , Postoperative Complications/therapy , T-Lymphocytes/immunology , Animals , Fungi/pathogenicity , Humans , Mycoses/microbiology , Postoperative Complications/microbiology , Transplantation, Homologous/adverse effects
16.
Bone Marrow Transplant ; 54(11): 1847-1858, 2019 11.
Article in English | MEDLINE | ID: mdl-31089287

ABSTRACT

Although allogeneic hematopoietic stem-cell transplantation (HSCT) provides high cure rates for children with high-risk acute lymphoblastic leukaemia (ALL), relapses remain the main cause of treatment failure. Whereas donor killer cell immunoglobulin-like receptor (KIR) genotype was shown to impact on relapse incidence in adult myeloid leukaemia similar studies in paediatric ALL are largely missing. Effect of donor KIR genotype on transplant outcome was evaluated in 317 children receiving a first myeloablative HSCT from an HLA-matched unrelated donor or sibling within the prospective ALL-SCT-BFM-2003 trial. Analysis of donor KIR gene polymorphism revealed that centromeric presence and telomeric absence of KIR B haplotypes was associated with reduced relapse risk. A centromeric/telomeric KIR score (ct-KIR score) integrating these observations correlated with relapse risk (hazard ratio (HR) 0.58; P = 0.002) while it had no impact on graft-versus-host disease or non-relapse mortality. In multivariable analyses ct-KIR score was associated with reduced relapse risk (HR 0.58; P = 0.003) and a trend towards improved event-free survival (HR 0.76; P = 0.059). This effect proved independent of MRD level prior to HSCT. Our data suggest that in children with ALL undergoing HSCT after myeloablative conditioning, donor selection based on KIR genotyping holds promise to improve clinical outcome by decreasing relapse risk and prolonged event-free survival.


Subject(s)
Donor Selection , Haplotypes , Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid, Acute , Polymorphism, Genetic , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Receptors, KIR/genetics , Telomere/genetics , Transplantation Conditioning , Adolescent , Adult , Child , Child, Preschool , Disease-Free Survival , Female , Humans , Infant , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/therapy , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Survival Rate
17.
Pediatr Blood Cancer ; 50(4): 921-3, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17960638

ABSTRACT

Recently published data show an extremely poor survival of infants with AML and HLXB9/ETV6 rearrangement which is the fusion, resulting from the translocation t(7;12)(q36;p13). None of the patients reported survived a period of 3 years, including four patients who have received allogeneic hematopoietic stem cell transplantation (HSCT). Herein, we report the clinical course of an 8-month-old patient with acute myeloid leukemia, M2 subtype and with a HLXB9/TEL rearrangement. The patient received a haploidentical HSCT in relapse situation without any prior re-induction. The patient became MRD-negative over a period of 53 days after HSCT. This case reinforces the potential benefit of a graft-versus-leukemia effect in the haploidentical setting even in chemoresistant myeloid leukemias with poor-prognosis molecular features.


Subject(s)
Graft vs Leukemia Effect , Hematopoietic Stem Cell Transplantation , Homeodomain Proteins/genetics , Leukemia, Myeloid, Acute/therapy , Proto-Oncogene Proteins c-ets/genetics , Repressor Proteins/genetics , Transcription Factors/genetics , Translocation, Genetic , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chromosomes, Human, Pair 12/genetics , Chromosomes, Human, Pair 7/genetics , Female , Haplotypes , Humans , In Situ Hybridization, Fluorescence , Infant , Leukemia, Myeloid, Acute/genetics , Neoplasm Recurrence, Local/therapy , Oncogene Proteins, Fusion/genetics , Reverse Transcriptase Polymerase Chain Reaction , ETS Translocation Variant 6 Protein
18.
Cancer Genomics Proteomics ; 5(5): 275-86, 2008.
Article in English | MEDLINE | ID: mdl-19129558

ABSTRACT

Recently it was shown that myeloid leukemic cells can be induced to differentiate into leukemia-derived dendritic cells (DCleu), regaining the stimulatory capacity of professional DCs while presenting the leukemic antigen repertoire. But so far, the induced antileukemic T-cell responses have varied in specificity and efficacy, or have even mediated opposite effects. In an attempt to further characterize the DC/DCleu induced T-cell response pattern, immunoscope spectratyping, a novel and powerful tool to detect T-cell receptor (TCR) rearrangements was used in combination with functional flow cytometry and non-radioactive fluorolysis assays. Human leucocyte antigen (HLA) matched donor T-cells were repeatedly stimulated, either with leukemic blasts (French-American-British, FAB M4eo) or the corresponding blast-derived DCs. Functional comparison revealed no significant difference in their T-cell stimulatory capacity, while the DC/DCleu fraction favored T-cells with a higher lytic activity, comprising a higher proportion of T-memory CD45R0+ cells. Stimulation with blasts and DC/DCleu induced a similar TCR restriction pattern, while stimulation with DC/DCleu favored the CD4 T-cell subset and seemed to cause a higher grade of restriction. In conclusion, a combined strategy using spectratyping with functional tests might not only provide useful information about the specificity and efficacy of the induced T-cell response, but also pave the way to gain effective T-cell clones for therapeutic use.


Subject(s)
Blast Crisis , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/pathology , T-Lymphocytes/immunology , Antigens, CD/immunology , Antigens, CD/metabolism , CD4-Positive T-Lymphocytes/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Flow Cytometry , Gene Rearrangement , Humans , Immunophenotyping , Leukemia, Myeloid, Acute/metabolism , Lymphocyte Activation , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
20.
Front Immunol ; 8: 773, 2017.
Article in English | MEDLINE | ID: mdl-28713390

ABSTRACT

Immunodeficiency, centromeric instability, and facial anomaly (ICF) syndrome is a rare autosomal recessive genetic condition with severe immunodeficiency, which leads to lethal infections if not recognized and treated in early childhood. Up-to-date treatment regimens consist of prophylactic and supportive treatment of the recurrent infections. Here, we report the case of a 1-year-old boy of Moroccan consanguineous parents, who was diagnosed at 4 months of age with ICF syndrome with a homozygous missense mutation in the DNMT3B gene. He was initially admitted to the hospital with recurrent pulmonary infections from the opportunistic pathogen Pneumocystis jirovecii (PJ). Further immunological workup revealed agammaglobulinemia in the presence of B cells. After successful recovery from the PJ pneumonia, he underwent hematopoietic stem cell transplantation (HSCT) from the HLA-matched healthy sister using a chemotherapeutic conditioning regimen consisting of treosulfan, fludarabine, and thiotepa. Other than acute chemotherapy-associated side effects, no serious adverse events occurred. Six months after HSCT immune-reconstitution, he had a stable chimerism with 2.9% autologous portion in the peripheral blood and a normal differential blood cell count, including all immunoglobulin subtypes. This is one of the first cases of successful HSCT in ICF syndrome. Early diagnosis and subsequent HSCT can prevent severe opportunistic infections and cure the immunodeficiency. Centromeric instability and facial anomaly remain unaffected. Although the long-term patient outcome and the neurological development remain to be seen, this curative therapy for immunodeficiency improves life expectancy and quality of life. This case is meant to raise physicians awareness for ICF syndrome and highlight the consideration for HSCT in ICF syndrome early on.

SELECTION OF CITATIONS
SEARCH DETAIL