Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Childs Nerv Syst ; 40(4): 1053-1064, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38376530

ABSTRACT

PURPOSE: Brain stem tumors in children < 3 months at diagnosis are extremely rare. Our aim is to study a retrospective cohort to improve the understanding of the disease course and guide patient management. METHODS: This is a multicenter retrospective analysis across the European Society for Pediatric Oncology SIOP-E HGG/DIPG Working Group linked centers, including patients with a brainstem tumor diagnosed between 2009 and 2020 and aged < 3 months at diagnosis. Clinical data were collected, and imaging characteristics were analyzed blindly and independently by two neuroradiologists. RESULTS: Five cases were identified. No patient received any therapy. The epicenter of two tumors was in the medulla oblongata alone and in the medulla oblongata and the pons in three. For patients with tumor in equal parts in the medulla oblongata and the pons (n = 3), the extension at diagnosis involved the spinal cord; for the two patients with the tumor epicenter in the medulla oblongata alone (n = 2), the extension at diagnosis included the pons (n = 2) and the spinal cord (n = 1). Biopsy was performed in one patient identifying a pilocytic astrocytoma. Two patients died. In one patient, autopsy revealed a high-grade glioma (case 3). Three survivors showed either spontaneous tumor regression (n = 2) or stable disease (n = 1). Survivors were followed up for 10, 7, and 0.6 years, respectively. One case had the typical imaging characteristics of a dorsal exophytic low-grade glioma. CONCLUSIONS: No patient fulfilled the radiologic criteria defining a high-grade glioma. Central neuroradiological review and biopsy may provide useful information regarding the patient management.


Subject(s)
Astrocytoma , Brain Neoplasms , Brain Stem Neoplasms , Glioma , Child , Humans , Retrospective Studies , Rare Diseases , Brain Stem Neoplasms/therapy , Glioma/pathology , Astrocytoma/pathology , Brain Neoplasms/pathology
2.
Br J Cancer ; 129(11): 1780-1786, 2023 11.
Article in English | MEDLINE | ID: mdl-37813959

ABSTRACT

BACKGROUND: Short-term infusions of dinutuximab beta plus isotretinoin and cytokines administered in previous immunotherapy studies in neuroblastoma were associated with severe pain. Here, long-term, continuous infusion of single-agent dinutuximab beta was evaluated in patients with relapsed/refractory neuroblastoma. METHODS: In this open-label, single-arm, Phase 2 study, patients with either refractory or relapsed high-risk neuroblastoma received dinutuximab beta by continuous infusion over 10 days of each cycle, for up to five cycles. The primary endpoint was objective response rate 24 weeks after the end of cycle 5. Secondary endpoints included adverse events, intravenous morphine use, best response, duration of response, and three-year progression-free and overall survival. RESULTS: Of the 40 patients included, 38 had evaluable response. Objective response rate was 26% and best response rate 37%. Median duration of response was 238 days (IQR 108-290). Three-year progression-free and overall survival rates were 31% (95% CI 17-47) and 66% (95% CI 47-79), respectively. Prophylactic intravenous morphine use and duration of use decreased with increasing cycles. The most common grade 3 treatment-related adverse events were pain, diarrhea, and hypokalemia. CONCLUSION: Long-term continuous infusion of single-agent dinutuximab beta is tolerable and associated with clinically meaningful responses in patients with relapsed/refractory high-risk neuroblastoma. CLINICAL TRIAL REGISTRATION: The study is registered with ClinicalTrials.gov (NCT02743429) and EudraCT (2014-000588-42).


Subject(s)
Neuroblastoma , Humans , Morphine Derivatives/therapeutic use , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/etiology , Neuroblastoma/drug therapy , Pain/drug therapy , Pain/etiology
3.
J Allergy Clin Immunol ; 149(5): 1744-1754.e8, 2022 05.
Article in English | MEDLINE | ID: mdl-34718043

ABSTRACT

BACKGROUND: Hematopoietic stem cell transplantation (HSCT) represents a curative treatment for patients with severe combined immunodeficiency (SCID), a group of monogenic immune disorders with an otherwise fatal outcome. OBJECTIVE: We performed a comprehensive multicenter analysis of genotype-specific HSCT outcome, including detailed analysis of immune reconstitution (IR) and the predictive value for clinical outcome. METHODS: HSCT outcome was studied in 338 patients with genetically confirmed SCID who underwent transplantation in 2006-2014 and who were registered in the SCETIDE registry. In a representative subgroup of 152 patients, data on IR and long-term clinical outcome were analyzed. RESULTS: Two-year OS was similar with matched family and unrelated donors and better than mismatched donor HSCT (P < .001). The 2-year event-free survival (EFS) was similar in matched and mismatched unrelated donor and less favorable in mismatched related donor (MMRD) HSCT (P < .001). Genetic subgroups did not differ in 2-year OS (P = .1) and EFS (P = .073). In multivariate analysis, pretransplantation infections and use of MMRDs were associated with less favorable OS and EFS. With a median follow-up of 6.2 years (range, 2.0-11.8 years), 73 of 152 patients in the IR cohort were alive and well without Ig dependency. IL-2 receptor gamma chain/Janus kinase 3/IL-7 receptor-deficient SCID, myeloablative conditioning, matched donor HSCT, and naive CD4 T lymphocytes >0.5 × 10e3/µL at +1 year were identified as independent predictors of favorable clinical and immunologic outcome. CONCLUSION: Recent advances in HSCT in SCID patients have resulted in improved OS and EFS in all genotypes and donor types. To achieve a favorable long-term outcome, treatment strategies should aim for optimal naive CD4 T lymphocyte regeneration.


Subject(s)
Hematopoietic Stem Cell Transplantation , Severe Combined Immunodeficiency , Cohort Studies , Hematopoietic Stem Cell Transplantation/methods , Humans , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/therapy , Transplantation Conditioning/methods , Unrelated Donors
4.
Hum Mutat ; 41(9): 1469-1487, 2020 09.
Article in English | MEDLINE | ID: mdl-32449975

ABSTRACT

Farber disease and spinal muscular atrophy with progressive myoclonic epilepsy are a spectrum of rare lysosomal storage disorders characterized by acid ceramidase deficiency (ACD), resulting from pathogenic variants in N-acylsphingosine amidohydrolase 1 (ASAH1). Other than simple listings provided in literature reviews, a curated, comprehensive list of ASAH1 mutations associated with ACD clinical phenotypes has not yet been published. This publication includes mutations in ASAH1 collected through the Observational and Cross-Sectional Cohort Study of the Natural History and Phenotypic Spectrum of Farber Disease (NHS), ClinicalTrials.gov identifier NCT03233841, in combination with an up-to-date curated list of published mutations. The NHS is the first to collect retrospective and prospective data on living and deceased patients with ACD presenting as Farber disease, who had or had not undergone hematopoietic stem cell transplantation. Forty-five patients representing the known clinical spectrum of Farber disease (living patients aged 1-28 years) were enrolled. The curation of known ASAH1 pathogenic variants using a single reference transcript includes 10 previously unpublished from the NHS and 63 that were previously reported. The publication of ASAH1 variants will be greatly beneficial to patients undergoing genetic testing in the future by providing a significantly expanded reference list of disease-causing variants.


Subject(s)
Acid Ceramidase/genetics , Farber Lipogranulomatosis/genetics , Muscular Atrophy, Spinal/genetics , Myoclonic Epilepsies, Progressive/genetics , Adolescent , Adult , Animals , Child , Child, Preschool , Humans , Infant , Mice, Knockout , Mutation , Young Adult
5.
Acta Neuropathol ; 140(4): 569-581, 2020 10.
Article in English | MEDLINE | ID: mdl-32776277

ABSTRACT

Diffuse IDH-mutant astrocytic tumors are rarely diagnosed in the cerebellum or brainstem. In this multi-institutional study, we characterized a series of primary infratentorial IDH-mutant astrocytic tumors with respect to clinical and molecular parameters. We report that about 80% of IDH mutations in these tumors are of non-IDH1-R132H variants which are rare in supratentorial astrocytomas. Most frequently, IDH1-R132C/G and IDH2-R172S/G mutations were present. Moreover, the frequencies of ATRX-loss and MGMT promoter methylation, which are typically associated with IDH mutations in supratentorial astrocytic tumors, were significantly lower in the infratentorial compartment. Gene panel sequencing revealed two samples with IDH1-R132C/H3F3A-K27M co-mutations. Genome-wide DNA methylation as well as chromosomal copy number profiling provided further evidence for a molecular distinctiveness of infratentorial IDH-mutant astrocytomas. Clinical outcome of patients with infratentorial IDH-mutant astrocytomas is significantly better than that of patients with diffuse midline gliomas, H3K27M-mutant (p < 0.005) and significantly worse than that of patients with supratentorial IDH-mutant astrocytomas (p = 0.028). The presented data highlight the very existence and distinctiveness of infratentorial IDH-mutant astrocytomas that have important implications for diagnostics and prognostication. They imply that molecular testing is critical for detection of these tumors, since many of these tumors cannot be identified by immunohistochemistry applied for the mutated IDH1-R132H protein or loss of ATRX.


Subject(s)
Astrocytoma/genetics , Astrocytoma/pathology , Infratentorial Neoplasms/genetics , Infratentorial Neoplasms/pathology , Isocitrate Dehydrogenase/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Humans , Infant , Male , Middle Aged , Mutation , Young Adult
6.
J Pediatr Hematol Oncol ; 42(7): e583-e588, 2020 10.
Article in English | MEDLINE | ID: mdl-31985550

ABSTRACT

Pediatric cancer treatment and hematopoietic stem cell transplantation (HSCT) carry considerable risks of morbidity. We conducted a single-center retrospective analysis of intensive care unit (ICU) admissions in unselected children and adolescents treated for cancer or undergoing HSCT. In a 10-year time period, 140 patients had 188 ICU admissions for a life-threatening condition. Main reasons for ICU admission were respiratory or cardiovascular insufficiency and sepsis. Mortality in the ICU was 19.1% and related to organ failure or acute complications in 77.8% and progress of the underlying malignancy in 22.2%. Mortality rates at 30, 100, and 365 days after discharge from the ICU were 24.5%, 30.9%, and 39.9%. Kaplan-Meier survival probabilities at 5 and 10 years were 46.4% and 39.8%, respectively. Multivariable analysis revealed the number of failed organ systems, the number of prior ICU stays, and days spent in the ICU as parameters independently associated with death. Taken together, the outcome of pediatric cancer and/or HSCT patients admitted to the ICU for life-threatening conditions was not as dismal as reported elsewhere. Most patients benefitted from ICU care, and survival was predominantly compromised by the evolution of complications.


Subject(s)
Hematopoietic Stem Cell Transplantation/adverse effects , Neoplasms/complications , Neoplasms/therapy , Adolescent , Child , Child, Preschool , Europe , Female , Hematopoietic Stem Cell Transplantation/mortality , Humans , Intensive Care Units/statistics & numerical data , Male , Neoplasms/mortality , Retrospective Studies
7.
Mycoses ; 63(2): 172-180, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31661569

ABSTRACT

BACKGROUND: Epidemiology and management practices of invasive fungal diseases (IFD) after allogeneic haematopoietic stem cell transplantation (HSCT) are a subject of constant change. We investigated the contemporary incidence, diagnostics, antifungal management and outcome at a major paediatric transplant centre in Germany. METHODS: The single-centre retrospective observational study included all paediatric allogeneic HSCT patients (pts) transplanted between 2005 and 2015. Patient-related data were assessed up to 365 days post-transplant. The primary endpoint was the incidence of possible, probable and proven IFDs. Secondary endpoints included diagnostics and antifungal treatment; analysis of risk factors; and overall survival with the last follow-up in January 2017. RESULTS: A total of 221 first (196), second (21) or third (4) procedures were performed in 200 pts (median age: 9 years, range, 0.5-22) for leukaemia/lymphoma (149) and non-malignant disorders (72). Prophylaxis was administered in 208 HSCT procedures (94%; fluconazole, 116, mould-active agents, 92). At least one computed tomography scan of the chest was performed in 146, and at least one galactomannan antigen assay in 60 procedures. There were 15 cases of proven (candidemia, 4; aspergillosis, 4) or probable (aspergillosis, 7) IFDs, accounting for an incidence rate of 6.8%. Overall mortality at last follow-up was 30%; the occurrence of proven/probable IFDs was associated with a reduced survival probability (P < .001). CONCLUSION: Morbidity and mortality from IFDs at our institution were consistent with data reported from other centres. Utilisation of healthcare resources for prevention, diagnosis and management of IFDs was considerable.


Subject(s)
Delivery of Health Care/statistics & numerical data , Hematopoietic Stem Cell Transplantation/adverse effects , Invasive Fungal Infections/epidemiology , Invasive Fungal Infections/therapy , Adolescent , Antifungal Agents/therapeutic use , Child , Child, Preschool , Cohort Studies , Female , Graft vs Host Disease/diagnosis , Graft vs Host Disease/epidemiology , Humans , Incidence , Infant , Invasive Fungal Infections/etiology , Invasive Fungal Infections/prevention & control , Male , Prevalence , Retrospective Studies , Risk Factors , Transplantation, Homologous/adverse effects , Treatment Outcome , Young Adult
8.
J Inherit Metab Dis ; 42(2): 286-294, 2019 03.
Article in English | MEDLINE | ID: mdl-30815900

ABSTRACT

BACKGROUND: Farber disease (FD) is a rare, lysosomal storage disorder caused by deficient acid ceramidase activity. FD has long been considered a fatal disorder with death in the first three decades of life resulting either from respiratory insufficiency as a consequence of airway involvement or from progressive neurodegeneration because of nervous system involvement. Peripheral symptoms associated with FD, including inflammatory joint disease, have been described to improve relatively rapidly after hematopoietic cell transplantation (HCT). AIMS: To evaluate the disease-specific status and limitations in the long-term follow-up after HCT, investigate genotype/phenotype correlations and the benefit of allogeneic HCT in FD patients with nervous system involvement. PATIENTS AND METHODS: Transplant- and disease-related information of ten FD patients was obtained by using a questionnaire, physicians' letters and additional telephone surveys. ASAH1 gene mutations were identified to search for genotype/phenotype correlations. RESULTS: After mainly busulfan-based preparative regimens, all patients engrafted with one late graft loss. The inflammatory symptoms resolved completely in all patients. Abnormal neurologic findings were present pre-transplant in 4/10 patients, post-transplant in 6/10 patients. Mutational analyses revealed new mutations in the ASAH1 gene and a broad diversity of phenotypes without a genotype/phenotype correlation. With a median follow-up of 10.4 years, overall survival was 80% with two transplant-related deaths. CONCLUSION: Allogeneic HCT leads to complete and persistent resolution of the inflammatory aspects in FD patients. It appears to have no beneficial effect on progression of nervous system involvement. New mutations in the acid ceramidase gene were identified. A genotype/phenotype correlation could not be established.


Subject(s)
Acid Ceramidase/genetics , Farber Lipogranulomatosis/genetics , Farber Lipogranulomatosis/therapy , Hematopoietic Stem Cell Transplantation , Adolescent , Child , Child, Preschool , DNA Mutational Analysis , Female , Humans , Internationality , Male , Mutation , Survival Rate , Transplantation Conditioning
9.
Am J Hematol ; 94(8): 880-890, 2019 08.
Article in English | MEDLINE | ID: mdl-31095771

ABSTRACT

Severe infections (SI) significantly impact on non-relapse mortality after hematopoietic stem cell transplantation (HSCT). We assessed 432 children and adolescents with acute lymphoblastic leukemia (ALL) after total body irradiation based myeloablative HSCT within the multicenter ALL-BFM-SCT 2003 trial for SI grade 3 or higher according to common terminology criteria for adverse events. A total 172 patients experienced at least one SI. Transplantation from matched unrelated donors (MUD) was associated with any type of SI in the pre-engraftment period (hazard ratio [HR]: 2.57; P < .001), and with any SI between day +30 and + 100 (HR: 2.91; P = .011). Bacterial (HR: 2.24; P = .041) and fungal infections (HR: 4.06; P = .057) occurred more often in the pre-engraftment phase and viral infections more often before day +30 (HR: 2.66; P = .007) or between day +30 and + 100 (HR: 3.89; P = .002) after HSCT from MUD as compared to matched sibling donors. Chronic GvHD was an independent risk factor for any type of SI after day +100 (HR: 2.57; P < .002). We conclude that allogeneic HSCT from MUD in children and adolescents with pediatric ALL is associated with higher infection rates, which seems attributable to an intensified GvHD prophylaxis including serotherapy and methotrexate.


Subject(s)
Bacterial Infections/epidemiology , Hematopoietic Stem Cell Transplantation/methods , Mycoses/epidemiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Unrelated Donors , Virus Diseases/epidemiology , Adolescent , Bacterial Infections/etiology , Bacterial Infections/prevention & control , Child , Child, Preschool , Female , Humans , Incidence , Male , Multivariate Analysis , Mycoses/etiology , Mycoses/prevention & control , Prospective Studies , Severity of Illness Index , Transplantation, Homologous , Unrelated Donors/statistics & numerical data , Virus Diseases/etiology , Virus Diseases/prevention & control , Whole-Body Irradiation
10.
Pediatr Blood Cancer ; 65(6): e26967, 2018 06.
Article in English | MEDLINE | ID: mdl-29350486

ABSTRACT

BACKGROUND: The monoclonal anti-GD2 antibody ch14.18/CHO in combination with IL-2 is active and effective in high-risk neuroblastoma (NB) patients. Here, we investigated the inflammatory response and treatment tolerance of long-term infusion (LTI) of ch14.18/CHO (10 × 10 mg/m2 ; 24 hr) in combination with subcutaneous (s.c.) IL-2 in a single center program. METHODS: Fifty-three NB patients received up to six cycles of 100 mg/m2 ch14.18/CHO (d8-18, where d represents day(s)) as LTI combined with 6 × 106 IU/m2 s.c. IL-2 (d1-5; 8-12) and 160 mg/m2 oral 13-cis retinoic acid (RA) (d19-32). Side effects of ch14.18/CHO and IL-2 treatment require hospitalization of patients on d8. Treatment tolerance was evaluated daily with clinical parameters (body temperature, vital signs, Lansky performance status, requirement of i.v. concomitant medication) to define an outpatient candidate status. sIL-2-R and C-reactive protein values were determined to assess the inflammatory response. RESULTS: LTI of ch14.18/CHO (d8-18) in combination with s.c.IL-2 (d8-12) showed an acceptable treatment tolerance that allowed all patients to receive part of the treatment as an outpatient (median time point of discharge: d15 for all cycles). The treatment tolerance improved from cycle to cycle and the time to become an outpatient candidate decreased from d15 to d13 in subsequent cycles. Clinical and laboratory parameters indicate a maximum inflammatory response at d11 of each cycle. Interestingly, the soluble IL-2 receptor remained increased at baseline of the next cycle indicating immune activation over the entire treatment period of 6 months. CONCLUSIONS: LTI of ch14.18/CHO combined with s.c.IL-2 shows an improved tolerance in subsequent cycles allowing outpatient treatment.


Subject(s)
Antibodies, Monoclonal/adverse effects , Gangliosides/antagonists & inhibitors , Immunotherapy/adverse effects , Inflammation/pathology , Interleukin-2/adverse effects , Neuroblastoma/therapy , Adolescent , Adult , Antibodies, Monoclonal/administration & dosage , Child , Child, Preschool , Drug Therapy, Combination , Female , Follow-Up Studies , Gangliosides/immunology , Humans , Immune Tolerance , Infant , Inflammation/chemically induced , Infusions, Intravenous , Interleukin-2/administration & dosage , Male , Neuroblastoma/immunology , Neuroblastoma/pathology , Prognosis , Young Adult
11.
Biochim Biophys Acta Mol Basis Dis ; 1863(2): 386-394, 2017 02.
Article in English | MEDLINE | ID: mdl-27915031

ABSTRACT

Acid Ceramidase Deficiency (Farber disease, FD) is an ultra-rare Lysosomal Storage Disorder that is poorly understood and often misdiagnosed as Juvenile Idiopathic Arthritis (JIA). Hallmarks of FD are accumulation of ceramides, widespread macrophage infiltration, splenomegaly, and lymphocytosis. The cytokines involved in this abnormal hematopoietic state are unknown. There are dozens of ceramide species and derivatives, but the specific ones that accumulate in FD have not been investigated. We used a multiplex assay to analyze cytokines and mass spectrometry to analyze ceramides in plasma from patients and mice with FD, controls, Farber patients treated by hematopoietic stem cell transplantation (HSCT), JIA patients, and patients with Gaucher disease. KC, MIP-1α, and MCP-1 were sequentially upregulated in plasma from FD mice. MCP-1, IL-10, IL-6, IL-12, and VEGF levels were elevated in plasma from Farber patients but not in control or JIA patients. C16-Ceramide (C16-Cer) and dhC16-Cer were upregulated in plasma from FD mice. a-OH-C18-Cer, dhC12-Cer, dhC24:1-Cer, and C22:1-Cer-1P accumulated in plasma from patients with FD. Most cytokines and only a-OH-C18-Cer returned to baseline levels in HSCT-treated Farber patients. Sphingosines were not altered. Chitotriosidase activity was also relatively low. A unique cytokine and ceramide profile was seen in the plasma of Farber patients that was not observed in plasma from HSCT-treated Farber patients, JIA patients, or Gaucher patients. The cytokine profile can potentially be used to prevent misdiagnosis of Farber as JIA and to monitor the response to treatment. Further understanding of why these signaling molecules and lipids are elevated can lead to better understanding of the etiology and pathophysiology of FD and inform development of future treatments.


Subject(s)
Ceramides/blood , Cytokines/blood , Farber Lipogranulomatosis/blood , Animals , Arthritis, Juvenile/blood , Bone Marrow Transplantation , Farber Lipogranulomatosis/therapy , Female , Hexosaminidases/blood , Humans , Male , Mice
12.
Haematologica ; 101(6): 741-6, 2016 06.
Article in English | MEDLINE | ID: mdl-26869631

ABSTRACT

UNLABELLED: Allogeneic hematopoietic stem cell transplantation is required as rescue therapy in about 20% of pediatric patients with acute lymphoblastic leukemia. However, the relapse rates are considerable, and relapse confers a poor outcome. Early assessment of the risk of relapse is therefore of paramount importance for the development of appropriate measures. We used the EuroChimerism approach to investigate the potential impact of lineage-specific chimerism testing for relapse-risk analysis in 162 pediatric patients with acute lymphoblastic leukemia after allogeneic stem cell transplantation in a multicenter study based on standardized transplantation protocols. Within a median observation time of 4.5 years, relapses have occurred in 41/162 patients at a median of 0.6 years after transplantation (range, 0.13-5.7 years). Prospective screening at defined consecutive time points revealed that reappearance of recipient-derived cells within the CD34(+) and CD8(+) cell subsets display the most significant association with the occurrence of relapses with hazard ratios of 5.2 (P=0.003) and 2.8 (P=0.008), respectively. The appearance of recipient cells after a period of pure donor chimerism in the CD34(+) and CD8(+) leukocyte subsets revealed dynamics indicative of a significantly elevated risk of relapse or imminent disease recurrence. Assessment of chimerism within these lineages can therefore provide complementary information for further diagnostic and, potentially, therapeutic purposes aiming at the prevention of overt relapse. This study was registered at clinical. TRIALS: gov with the number NC01423747.


Subject(s)
Cell Lineage , Hematopoietic Stem Cell Transplantation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Transplantation Chimera , Adolescent , Biomarkers , Child , Child, Preschool , Female , Humans , Immunophenotyping , Infant , Leukocytes/metabolism , Leukocytes/pathology , Male , Recurrence , Risk Assessment , Risk Factors , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , Transplantation, Homologous , Treatment Outcome , Young Adult
13.
J Pediatr Hematol Oncol ; 37(1): e41-4, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25000468

ABSTRACT

Dyskeratosis congenita (DC) is a rare inherited disorder characterized by reticular skin pigmentation, oral cavity leukoplakia, and nail dystrophy. Bone marrow failure in DC can only be cured by allogeneic hematopoietic stem cell transplantation (HSCT). After a nonmyeloablative, matched unrelated donor transplant, the 21-year-old patient experienced severe lower gastrointestinal tract hemorrhage caused by diffuse colitis. The etiology remained unclear. Diffuse colitis with life-threatening hemorrhage has now been reported in 3 DC patients after unrelated allogeneic HSCT. To identify the underlying causes and the disease-specific risks, and to allow for prevention and/or optimal management, data should be prospectively collected.


Subject(s)
Colitis/etiology , Dyskeratosis Congenita/complications , Gastrointestinal Hemorrhage/etiology , Hematopoietic Stem Cell Transplantation/adverse effects , Adult , Dyskeratosis Congenita/therapy , Humans , Male , Transplantation, Homologous
14.
Blood ; 119(25): 6016-24, 2012 Jun 21.
Article in English | MEDLINE | ID: mdl-22451424

ABSTRACT

Familial hemophagocytic lymphohistiocytosis (FHL) is a genetically determined hyperinflammatory syndrome caused by uncontrolled immune response mediated by T-lymphocytes, natural killer (NK) cells, and macrophages. STXBP2 mutations have recently been associated with FHL5. To better characterize the genetic and clinical spectrum of FHL5, we analyzed a cohort of 185 patients with suspected FHL for mutations in STXBP2. We detected biallelic mutations in 37 patients from 28 families of various ethnic origins. Missense mutations and mutations affecting 1 of the exon 15 splice sites were the predominant changes detectable in this cohort. Patients with exon 15 splice-site mutations (n = 13) developed clinical manifestations significantly later than patients with other mutations (median age, 4.1 year vs 2 months) and showed less severe impairment of degranulation and cytotoxic function of NK cells and CTLs. Patients with FHL5 showed several atypical features, including sensorineural hearing deficit, abnormal bleeding, and, most frequently, severe diarrhea that was only present in early-onset disease. In conclusion, we report the largest cohort of patients with FHL5 so far, describe an extended disease spectrum, and demonstrate for the first time a clear genotype-phenotype correlation.


Subject(s)
Lymphohistiocytosis, Hemophagocytic/genetics , Munc18 Proteins/genetics , Mutation , Adolescent , Adult , Basophil Degranulation Test , Child , Child, Preschool , Cohort Studies , DNA Mutational Analysis , Epistasis, Genetic , Female , Genetic Association Studies , Humans , Infant , Infant, Newborn , Lymphohistiocytosis, Hemophagocytic/classification , Lymphohistiocytosis, Hemophagocytic/ethnology , Male , Models, Biological , Munc18 Proteins/physiology , Mutation/physiology , Qa-SNARE Proteins/genetics , Young Adult
15.
Blood ; 119(12): 2935-42, 2012 Mar 22.
Article in English | MEDLINE | ID: mdl-22160619

ABSTRACT

We investigated prospectively factors influencing the safety of hematopoietic stem cell (HSC) collection in 453 pediatric donors. The children in the study donated either BM or peripheral blood stem cells (PBSCs) according to center policy. A large variability in approach to donor issues was observed between the participating centers. Significant differences were observed between BM and PBSC donors regarding pain, blood allotransfusion, duration of hospital stay, and iron supplementation; however, differences between the groups undergoing BM vs PBSC donation preclude direct risk comparisons between the 2 procedures. The most common adverse event was pain, reported mainly by older children after BM harvest, but also observed after central venous catheter (CVC) placement for PBSC collection. With regard to severe adverse events, one patient (0.7%) developed a pneumothorax with hydrothorax after CVC placement for PBSC collection. The risk of allotransfusion after BM harvest was associated with a donor age of < 4 years and a BM harvest volume of > 20 mL/kg. Children < 4 years were at higher risk than older children for allotransfusion after BM harvest and there was a higher risk of complications from CVC placement before apheresis. We conclude that PBSC and BM collection are safe procedures in children.


Subject(s)
Blood Specimen Collection/adverse effects , Blood Specimen Collection/methods , Hematopoietic Stem Cell Transplantation , Tissue Donors , Adolescent , Bone Marrow Transplantation , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Peripheral Blood Stem Cell Transplantation , Siblings , Tissue Donors/psychology
17.
Lancet ; 379(9823): 1301-9, 2012 Apr 07.
Article in English | MEDLINE | ID: mdl-22364685

ABSTRACT

BACKGROUND: Hepatic veno-occlusive disease is a leading cause of morbidity and mortality after haemopoietic stem-cell transplantation (HSCT). We aimed to assess whether defibrotide can reduce the incidence of veno-occlusive disease in this setting. METHODS: In our phase 3 open-label, randomised controlled trial, we enrolled patients at 28 European university hospitals or academic medical centres. Eligible patients were younger than 18 years, had undergone myeloablative conditioning before allogeneic or autologous HSCT, and had one or more risk factor for veno-occlusive disease based on modified Seattle criteria. We centrally assigned eligible participants on the basis of a computer-generated randomisation sequence (1:1), stratified by centre and presence of osteopetrosis, to receive intravenous defibrotide prophylaxis (treatment group) or not (control group). The primary endpoint was incidence of veno-occlusive disease by 30 days after HSCT, adjudicated by a masked, independent review committee, in eligible patients who consented to randomisation (intention-to-treat population), and was assessed with a competing risk approach. Patients in either group who developed veno-occlusive disease received defibrotide for treatment. We assessed adverse events to 180 days after HSCT in all patients who received allocated prophylaxis. This trial is registered with ClinicalTrials.gov, number NCT00272948. FINDINGS: Between Jan 25, 2006, and Jan 29, 2009, we enrolled 356 eligible patients to the intention-to-treat population. 22 (12%) of 180 patients randomly allocated to the defibrotide group had veno-occlusive disease by 30 days after HSCT compared with 35 (20%) of 176 controls (risk difference -7·7%, 95% CI -15·3 to -0·1; Z test for competing risk analysis p=0·0488; log-rank test p=0·0507). 154 (87%) of 177 patients in the defibrotide group had adverse events by day 180 compared with 155 (88%) of 176 controls. INTERPRETATION: Defibrotide prophylaxis seems to reduce incidence of veno-occlusive disease and is well tolerated. Thus, such prophylaxis could present a useful clinical option for this serious complication of HSCT. FUNDING: Gentium SpA, European Group for Blood and Marrow Transplantation.


Subject(s)
Fibrinolytic Agents/therapeutic use , Hematopoietic Stem Cell Transplantation/adverse effects , Hepatic Veno-Occlusive Disease/prevention & control , Polydeoxyribonucleotides/therapeutic use , Adolescent , Bilirubin/blood , Child , Child, Preschool , Female , Graft vs Host Disease/epidemiology , Hepatic Veno-Occlusive Disease/drug therapy , Hepatic Veno-Occlusive Disease/epidemiology , Humans , Incidence , Infant , Infusions, Intravenous , Male , Multiple Organ Failure/epidemiology , Renal Insufficiency/epidemiology
18.
BMC Infect Dis ; 13: 330, 2013 Jul 19.
Article in English | MEDLINE | ID: mdl-23870704

ABSTRACT

BACKGROUND: Human cytomegalovirus infections are still significant causes of morbidity and mortality in transplant recipients. The use of antiviral agents is limited by toxicity and evolving resistance in immunocompromised patients with ongoing viral replication during therapy. Here, we present the first documented case of genotypic resistance against maribavir in a bone marrow transplant (BMT) recipient. CASE PRESENTATION: The female 13-year-old patient was suffering from a refractory cytopenia. Ganciclovir, foscarnet, cidofovir, leflunomide and maribavir, an inhibitor of the cytomegalovirus UL97 protein, were administered to treat a therapy-resistant cytomegalovirus infection. Viral mutations conferring resistance against nucleotide and pyrophosphate analogs as well as maribavir (MBV) have evolved sequentially. Particularly, impressive was the fast emergence of multiple mutations T409M, H411Y and H411N conferring maribavir resistance after less than 6 weeks. CONCLUSION: We describe the fast emergence of cytomegalovirus variants with different maribavir resistance associated mutations in a bone marrow transplant recipient treated with MBV 400 mg p.o. twice per day. The results suggest that a high virus load permitted a selection of several but distinct therapy-resistant HCMV mutants. Since a phase II study with MBV is intended for the treatment of resistant or refractory HCMV infections in transplant recipients this has to be kept in mind in patients with high viral loads during therapy (NCT01611974).


Subject(s)
Antiviral Agents/pharmacology , Benzimidazoles/pharmacology , Bone Marrow Transplantation , Cytomegalovirus Infections/microbiology , Cytomegalovirus/drug effects , Cytomegalovirus/isolation & purification , Ribonucleosides/pharmacology , Adolescent , Antiviral Agents/therapeutic use , Benzimidazoles/therapeutic use , Drug Resistance, Viral , Female , Humans , Ribonucleosides/therapeutic use
19.
Front Oncol ; 13: 1082771, 2023.
Article in English | MEDLINE | ID: mdl-36816982

ABSTRACT

Prognosis in children with refractory and relapsed high-risk neuroblastoma is poor. Only a minority of patients obtain remission when treated with second-line chemotherapy regimens. Chemotherapy combined with anti-GD2 antibodies has previously been shown to increase response and survival rates. We retrospectively analyzed a cohort of 25 patients with relapsed or refractory high-risk neuroblastoma who were treated with irinotecan/temozolomide chemotherapy in combination with the anti-GD2 antibody dinutuximab beta. The therapy resulted in an objective response rate of 64%, with 32% of patients achieving a complete response. Response to treatment was observed in patients with refractory disease (n=5) and those with first (n=12) or consecutive (n=8) relapses, including patients with progressing disease. In four patients, best response was achieved after more than 5 cycles, suggesting that some patients may benefit from prolonged chemotherapy and dinutuximab beta treatment. Fourteen of our 25 patients had previously received dinutuximab beta, four of whom achieved complete response and six partial response (objective response rate 71%). The therapy was well tolerated, even in heavily pre-treated patients and those who had previously received dinutuximab beta treatment. Toxicities were comparable to those previously reported for the individual therapies, and no discontinuations due to toxicities occurred. Combination of chemotherapy with dinutuximab beta is a promising treatment option for patients with relapsed or refractory high-risk neuroblastoma and should be further explored in clinical studies.

20.
J Neurosurg Case Lessons ; 5(25)2023 Jun 19.
Article in English | MEDLINE | ID: mdl-37354435

ABSTRACT

BACKGROUND: Eosinophilic granuloma (EG) belongs to the family of Langerhans cell histiocytosis (LCH) and is considered to be a benign disease typically found in children younger than 15 years of age. Here, the authors describe an EG of unusual localization and clinical presentation. OBSERVATIONS: The authors report a 9-year-old girl with an EG presenting as an osteolytic lesion of the clivus. After transsphenoidal resection and histological confirmation, adjuvant chemotherapy was initiated. Presenting signs and symptoms were weight loss, episodic grimacing, and moderate ballism-like movements. After a follow-up-period of 32 months, the patient presented with a total resolution of initial symptoms and no further tumor growth. LESSONS: Although these lesions are rare, one should consider EG as a differential diagnosis when confronted with osteolytic lesions of the clivus.

SELECTION OF CITATIONS
SEARCH DETAIL