Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Article in English | MEDLINE | ID: mdl-39271290

ABSTRACT

BACKGROUND AND PURPOSE: CNS embryonal tumor with PLAGL1/PLAGL2 amplification (ET, PLAGL) is a newly identified, highly malignant pediatric tumor. Systematic MRI descriptions of ET, PLAGL are currently lacking. MATERIALS AND METHODS: MRI data from 19 treatment-naïve patients with confirmed ET, PLAGL were analyzed. Evaluation focused on anatomical involvement, tumor localization, MRI signal characteristics, DWI behavior, and the presence of necrosis and hemorrhage. Descriptive statistics (median, interquartile range, percentage) were assessed. RESULTS: Ten patients had PLAGL1 and nine PLAGL2 amplifications. The solid components of the tumors were often multinodular with heterogeneous enhancement (mild to intermediate in 47% and intermediate to strong in 47% of cases). Non-solid components included cysts in 47% and necrosis in 84% of the cases. The tumors showed heterogeneous T2WI hyper-and isointensity (74%), relatively little diffusion restriction (ADC values < contralateral normal-appearing WM in 36% of cases with available DWI), and tendencies towards hemorrhage/calcification (42%). No reliable distinction was found between PLAGL1-and PLAGL2-amplified tumors or compared to other embryonal CNS tumors. CONCLUSIONS: The study contributes to understanding the imaging characteristics of ET, PLAGL. It underscores the need for collaboration in studying rare pediatric tumors and advocates for the use of harmonized imaging protocols for better characterization. ABBREVIATIONS: ATRT= atypical teratoid/rhabdoid tumor; ETMR= embryonal tumor with multilayered rosettes; ET, PLAGL= CNS embryonal tumor with PLAGL amplification; EVD= external ventricular drain; IQR: interquartile range; PLAGL1= pleomorphic adenoma gene-like 1; PLAGL2= pleomorphic adenoma gene-like 2; WHO= World Health Organization.

2.
ESMO Open ; 9(5): 103006, 2024 May.
Article in English | MEDLINE | ID: mdl-38657345

ABSTRACT

BACKGROUND: Patients with infantile fibrosarcoma (IFS) have shown strong and long-lasting responses to larotrectinib, a tropomyosin receptor kinase inhibitor (TRKi), in single-arm clinical trials. Conventional chemotherapy has also shown important efficacy. But, until now, no comparative data exist. This study aims to assess the therapeutic benefit of larotrectinib over the current standard of care (SOC) of chemotherapy in paediatric patients with locally advanced or metastatic IFS. PATIENTS AND METHODS: EPI VITRAKVI is a retrospective, observational, externally controlled study (NCT05236257). Data of patients aged ≤21 years with locally advanced or metastatic IFS treated with larotrectinib in the phase I/II SCOUT trial (NCT02637687) were compared with those of an external historical control group (data of Institut Curie and Cooperative Weichteilsarkom Studiengruppe) treated with a chemotherapy-based regimen. Between-group differences were assessed after balancing groups using inverse probability of treatment weighting (IPTW). RESULTS: In total, 93 patients were compared, 51 in the larotrectinib arm and 42 in the external control arm. After therapy, 4 patients (7.8%) in the larotrectinib group had a medical treatment failure event [start of new systemic treatment (2 cases), mutilating surgery (2 cases)] versus 15 (35.7%) in the external control group [start of new systemic treatment (6 cases), mutilating surgery (5 cases), radiation therapy (2 cases), and death (2 cases)]. Larotrectinib was associated with an 80% reduced likelihood of encountering a medical treatment failure event, when compared to the external control group (weighted and stratified hazard ratio 0.20, 95% confidence interval 0.06-0.63, P = 0.0060). These results were confirmed by sensitivity analyses, including exact matching, and subgroup analyses for number of lines of treatment. CONCLUSIONS: Treatment with larotrectinib reduced the need of subsequent therapies compared to SOC with chemotherapy in children with locally advanced or metastatic IFS, regardless of the line of treatment.


Subject(s)
Fibrosarcoma , Pyrazoles , Pyrimidines , Standard of Care , Humans , Fibrosarcoma/drug therapy , Female , Retrospective Studies , Male , Infant , Pyrazoles/therapeutic use , Pyrazoles/pharmacology , Pyrimidines/therapeutic use , Pyrimidines/pharmacology , Child, Preschool , Child , Adolescent , Neoplasm Metastasis , Treatment Outcome
4.
Bone Marrow Transplant ; 56(7): 1550-1557, 2021 07.
Article in English | MEDLINE | ID: mdl-33514918

ABSTRACT

Patients with advanced Ewing sarcoma (AES) carry a poor prognosis. Retrospectively, we analyzed 66 AES patients treated with allogeneic stem cell transplantation (allo-SCT) receiving HLA-mismatched (group A, n = 39) versus HLA-matched grafts (group B, n = 27). Median age at diagnosis was 13 years, and 15 years (range 3-49 years) at allo-SCT. The two groups did not differ statistically in distribution of gender, age, remission status/number of relapses at allo-SCT, or risk stratum. 9/39 (23%) group A versus 2/27 (7%) group B patients developed severe acute graft versus host disease (GvHD). Of patients alive at day 100, 7/34 (21%) group A versus 9/19 (47%) group B patients had developed chronic GvHD. In group A, 33/39 (85%) versus 20/27 (74%) group B patients died of disease and 1/39 (3%) versus 1/27 (4%) patients died of complications, respectively. Altogether 12/66 (18%) patients survived in CR. Median EFS 24 months after allo-SCT was 20% in both groups, median OS was 27% (group A) versus 17% (group B), respectively. There was no difference in EFS and OS in AES patients transplanted with HLA-mismatched versus HLA-matched graft in univariate and multivariate analyses. In this analysis, CR at allo-SCT is a condition for survival (p < 0.02).


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Sarcoma, Ewing , Adolescent , Adult , Child , Child, Preschool , Humans , Middle Aged , Neoplasm Recurrence, Local , Retrospective Studies , Sarcoma, Ewing/therapy , Transplantation Conditioning , Young Adult
5.
Eur J Pediatr ; 179(5): 749-756, 2020 May.
Article in English | MEDLINE | ID: mdl-31901982

ABSTRACT

Very rare tumors (VRTs) account for up to 11% of childhood cancers. Dedicated national groups and registries only exist in some European countries. Pleuropulmonary blastoma (PPB) is a very rare intrathoracic pediatric tumor with a potentially severe prognosis. Due to its rarity, it sometimes goes unrecognized. We investigated PPB diagnostic capability and possible correlations between diagnostic performance and VRT-dedicated activities. The number of cases of PPB registered between 2000 and 2014 at pediatric oncology centers in Europe was compared with the number of expected cases. Data sources included VRT registries, population-based cancer registries, and hospital registries. Data were obtained for 25 countries, grouped into 4 geographical regions. The expected cases were 111, and the observed cases were 129. The observed-to-expected ratio was 1.86 for Northern Europe, 1.33 for Southern Europe, 1.22 for Central Europe, and 0.65 for Eastern Europe. More cases than expected were registered in all countries with an official VRT registry.Conclusion: The number of cases observed is consistent with expectations, but disparities exist across Europe. Difficulties in diagnosing PPB emerged in most Eastern countries. The incidence rate of PPB may be underestimated. The creation of VRT-dedicated groups and a European Registry for VRTs could help to reduce inequalities.What is Known:• Very rare pediatric tumors are often not recognized, despite representing almost 11% of childhood cancers .• Pleuropulmonary blastoma is a rare pediatric tumor with a poor prognosis.What is New:• The ability to diagnose and register pleuropulmonary blastoma varies in Europe.Registries dedicated to very rare pediatric tumors improve the diagnostic rates.• The incidence rate of pleuropulmonary blastoma may currently be underestimated.


Subject(s)
Lung Neoplasms/epidemiology , Pulmonary Blastoma/epidemiology , Adolescent , Child , Child, Preschool , Cross-Sectional Studies , Europe/epidemiology , Female , Humans , Incidence , Infant , Infant, Newborn , Lung Neoplasms/diagnosis , Male , Pulmonary Blastoma/diagnosis , Registries/statistics & numerical data , Retrospective Studies
6.
Oncogene ; 35(16): 2020-30, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-26189795

ABSTRACT

Current treatment regimens for rhabdomyosarcoma (RMS), the most common pediatric soft tissue cancer, rely on conventional chemotherapy, and although they show clinical benefit, there is a significant risk of adverse side effects and secondary tumors later in life. Therefore, identifying and targeting sub-populations with higher tumorigenic potential and self-renewing capacity would offer improved patient management strategies. Hedgehog signaling has been linked to the development of embryonal RMS (ERMS) through mouse genetics and rare human syndromes. However, activating mutations in this pathway in sporadic RMS are rare and therefore the contribution of hedgehog signaling to oncogenesis remains unclear. Here, we show by genetic loss- and gain-of-function experiments and the use of clinically relevant small molecule modulators that hedgehog signaling is important for controlling self-renewal of a subpopulation of RMS cells in vitro and tumor initiation in vivo. In addition, hedgehog activity altered chemoresistance, motility and differentiation status. The core stem cell gene NANOG was determined to be important for ERMS self-renewal, possibly acting downstream of hedgehog signaling. Crucially, evaluating the presence of a subpopulation of tumor-propagating cells in patient biopsies identified by GLI1 and NANOG expression had prognostic significance. Hence, this work identifies novel functional aspects of hedgehog signaling in ERMS, redefines the rationale for its targeting as means to control ERMS self-renewal and underscores the importance of studying functional tumor heterogeneity in pediatric cancers.


Subject(s)
Carcinogenesis , Hedgehog Proteins/metabolism , Rhabdomyosarcoma, Embryonal/pathology , Signal Transduction , Humans , Prognosis , Rhabdomyosarcoma, Embryonal/metabolism , Tumor Cells, Cultured
7.
Bone Marrow Transplant ; 50 Suppl 2: S72-6, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26039213

ABSTRACT

Natural killer (NK) cell activity has been shown to have potential activity against Ewing's sarcoma (EWS) especially in tumors with low HLA I expression and high NKG2D expression. Two patients with metastatic relapsed and primary metastatic stage IV EWS who had received two courses of high dose chemotherapy with autologous stem cell rescue were transplanted from a haploidentical parental stem cell donor. Patients are alive in ongoing CR for 10.2 and 3.4 years now. Post transplant local second and first relapses were treated successfully in both patients. In vivo IL-2 stimulation not only increased the number and activity of effector cells in one patient but was also associated with severe GvHD. In vitro studies demonstrated high NK cell activity against K562 and relevant activity against EWS cell line A673 post transplant. NK activity was enhanced by cytokine prestimulation as well as by EWS targeting anti-GD2 Ab. Haploidentical hematopoietic stem cell transplantation (HSCT) might contribute to long-term survival by NK cell-mediated effect exerted by donor-derived NK cells. Local tumor recurrence was manageable in both high-risk patients indicating systemic immune control preventing subsequent metastasizing. The efficacy of haploidentical HSCT, cytokine application and tumor targeting antibodies for the use of Ab-dependent cellular cytotoxicity needs evaluation in clinical trials.


Subject(s)
Hematopoietic Stem Cell Transplantation , Killer Cells, Natural/immunology , Neoplasm Recurrence, Local , Sarcoma, Ewing , Adolescent , Female , Follow-Up Studies , Humans , Male , NK Cell Lectin-Like Receptor Subfamily K/immunology , Neoplasm Metastasis , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/prevention & control , Neoplasm Staging , Sarcoma, Ewing/immunology , Sarcoma, Ewing/pathology , Sarcoma, Ewing/prevention & control
8.
Klin Padiatr ; 226(3): 143-8, 2014 May.
Article in English | MEDLINE | ID: mdl-24633978

ABSTRACT

Rhabdoid tumors mainly affect infants and other very young children with a marked vulnerability towards intensive therapy such as invasive surgery, high dose chemotherapy (HDCT) and dose intense radiotherapy. Radiotherapy (RT) is a promising option in rhabdoid tumors but its application in infants remains controversial. Neurocognitive and vascular side effects occur even long after completion of therapy. Therapeutic recommendations suggested by the European Rhabdoid Registry including RT, high dose chemotherapy (HDCT) and methotrexate (MTX) were developed by a consensus committee. Unique to our EU-RHAB database is the ability to analyze data of 64 of 81 registered infants (under one year of age) separate from older children. 20 (age at diagnoses 2-12 months) of these had received radiotherapy. To our knowledge, this is the first report specifically analyzing treatment data of infants suffering from malignant rhabdoid tumors. Our results suggest that radiotherapy significantly increases the mean survival time as well as the 3 year overall survival in infants. We detected a doubling of survival times in infants who received RT. Overall, our results suggest that infants benefit from RT with tolerable acute side effects. Severe long term sequelae likely due to intraventricular MTX and/or RT were reported in 4 patients (leukoencephalopathy). No differences in chemotherapy-related toxicity were observed between infants and children. We suggest that a nihilistic therapeutic approach towards young infants is not warranted and that RT may not be a priori rejected as a therapeutic option in infants.


Subject(s)
Registries , Rhabdoid Tumor/therapy , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chemoradiotherapy/adverse effects , Combined Modality Therapy/adverse effects , Combined Modality Therapy/methods , Dactinomycin/administration & dosage , Dactinomycin/adverse effects , Feasibility Studies , Germany , Humans , Infant , Infant, Newborn , Infusions, Intraventricular , Interdisciplinary Communication , Methotrexate/administration & dosage , Methotrexate/adverse effects , Radiotherapy Dosage , Rhabdoid Tumor/diagnosis , Rhabdoid Tumor/mortality , Survival Rate
9.
Br J Cancer ; 109(10): 2523-32, 2013 Nov 12.
Article in English | MEDLINE | ID: mdl-24149176

ABSTRACT

BACKGROUND: Allogeneic haematopoietic stem cell transplantation (allo-SCT) may provide donor cytotoxic T cell-/NK cell-mediated disease control in patients with rhabdomyosarcoma (RMS). However, little is known about the prevalence of graft-vs-RMS effects and only a few case experiences have been reported. METHODS: We evaluated allo-SCT outcomes of 30 European Group for Blood and Marrow Transplantation (EBMT)-registered patients with advanced RMS regarding toxicity, progression-free survival (PFS) and overall survival (OS) after allo-SCT. Twenty patients were conditioned with reduced intensity and ten with high-dose chemotherapy. Twenty-three patients were transplanted with HLA-matched and seven with HLA-mismatched grafts. Three patients additionally received donor lymphocyte infusions (DLIs). Median follow-up was 9 months. RESULTS: Three-year OS was 20% (s.e.±8%) with a median survival time of 12 months. Cumulative risk of progression was 67% (s.e.±10%) and 11% (s.e.±6%) for death of complications. Thirteen patients developed acute graft-vs-host disease (GvHD) and five developed chronic GvHD. Eighteen patients died of disease and four of complications. Eight patients survived in complete remission (CR) (median: 44 months). No patients with residual disease before allo-SCT were converted to CR. CONCLUSION: The use of allo-SCT in patients with advanced RMS is currently experimental. In a subset of patients, it may constitute a valuable approach for consolidating CR, but this needs to be validated in prospective trials.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Rhabdomyosarcoma/surgery , Adolescent , Adult , Child , Child, Preschool , Disease Progression , Female , Graft vs Host Disease/etiology , Graft vs Host Disease/mortality , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Male , Neoplasm Recurrence, Local/epidemiology , Neoplasm Recurrence, Local/mortality , Retrospective Studies , Rhabdomyosarcoma/mortality , Transplantation, Homologous , Young Adult
10.
Klin Padiatr ; 225(6): 309-14, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23946091

ABSTRACT

To investigate antitumor activity and toxicity associated with combined topotecan and carboplatin treatment in children and adolescents with metastasized, untreated soft tissue sarcoma (STS).Patients (n=34) less than 21 years old and untreated, stage IV STS. Patients were treated with topotecan (1 mg/m²/d for 4 days) and carboplatin (150 mg/m²/d for 4 days) (TC course) during week 1 and 4 of a chemotherapy window trial, which was followed by chemotherapy and local therapy from week 6 on. We evaluated the side effects, toxicity and tumor response (using RECIST criteria) 6 weeks after starting the 2 TC chemotherapy courses.The objective response rate (ORR) was 38% (n=13 patients with a partial response (PR)), and a stable disease (SD) was reached in 11 cases. No patient showed a complete response (CR) of all metastatic lesions, although 1 patient showed a CR of the target lesion. 2 patients died of progress of disease (PD). Toxicity was mainly hematological (grade III/IV toxicity 79%), and nonhematological toxicities mainly included infection, fever, nausea,and vomiting. Regarding adverse events, 4 probable and 8 possible events related to study medication occurred among the 66 courses of TC.In conclusion, TC was potent against high-risk STS, but results and toxicity data were not superior to former published monotherapeutic topotecan therapies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carboplatin/administration & dosage , Sarcoma/drug therapy , Soft Tissue Neoplasms/drug therapy , Topotecan/administration & dosage , Adolescent , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carboplatin/adverse effects , Child , Child, Preschool , Cohort Studies , Combined Modality Therapy , Disease Progression , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Follow-Up Studies , Humans , Male , Neoadjuvant Therapy , Neoplasm Staging , Rhabdomyosarcoma/diagnosis , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/mortality , Rhabdomyosarcoma/pathology , Sarcoma/diagnosis , Sarcoma/mortality , Sarcoma/pathology , Sarcoma, Ewing/diagnosis , Sarcoma, Ewing/drug therapy , Sarcoma, Ewing/mortality , Sarcoma, Ewing/pathology , Sarcoma, Synovial/diagnosis , Sarcoma, Synovial/drug therapy , Sarcoma, Synovial/mortality , Sarcoma, Synovial/pathology , Soft Tissue Neoplasms/diagnosis , Soft Tissue Neoplasms/mortality , Soft Tissue Neoplasms/pathology , Survival Rate , Topotecan/adverse effects , Treatment Outcome
11.
Klin Padiatr ; 225(3): 177-8, 2013 May.
Article in English | MEDLINE | ID: mdl-23625684

ABSTRACT

Recently, germline mutations of DICER1 have been identified in patients with rare neoplasms suggesting the existence of a newly discovered cancer prone syndrome. Initially, DICER1 mutations were identified in patients with familial pleuropulmonary blastoma. Subsequently, additional manifestations of the syndrome have been identified including cystic nephroma, medulloepithelioma, Sertoli-Leydig cell tumor and others. The DICER1 gene encodes an enzyme that is involved in the biogenesis of microRNAs. The entire tumor spectrum and the respective tumor risks are unknown. We are in the process of launching a natural history study aimed at identifying more information on this new cancer syndrome.


Subject(s)
DEAD-box RNA Helicases/genetics , Lung Neoplasms/genetics , Neoplastic Syndromes, Hereditary/genetics , Pulmonary Blastoma/genetics , Ribonuclease III/genetics , Child , Chromosomes, Human, Pair 14 , Genetic Carrier Screening , Germ-Line Mutation , Humans , MicroRNAs/genetics , Registries
12.
Pediatr Blood Cancer ; 60(8): 1267-73, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23418028

ABSTRACT

BACKGROUND: Recurrences in primary localized alveolar rhabdomyosarcoma (RMA) are common. Post-relapse survival is poor. We evaluated prognostic factors including relapse treatment in patients with recurrent RMA. METHODS: Relapses occurred in 115/235 patients with nonmetastatic RMA treated in four consecutive CWS-trials after achievement of a complete remission. Sufficient information about post-relapse treatment and outcome could be obtained in 99 patients and was retrospectively analyzed. RESULTS: Nine of 99 patients received no salvage therapy and died after a median of 2 months. The remaining 90 patients received multimodal relapse treatment including mandatory chemotherapy. Recurrences were grossly resected in 39 patients; 57 patients received radiation. At a median follow-up from relapse of 8 years, 20 patients were alive and disease-free (5-year post-relapse survival [PROS] 21.3 ± 8). All surviving patients apart from a single individual had an isolated, circumscribed recurrence. Sixteen of 20 survivors were treated with adequate local relapse therapy (ALRT, i.e., either complete resection or gross resection + radiation). Survival in the subgroup of 27 individuals with circumscribed recurrences and ALRT was significantly better (PROS 53.7 ± 19) compared with disseminated recurrences and/or tumors treated without ALRT. Absence of primary lymph node involvement, circumscribed relapses, ALRT, and achievement of a second CR were identified as independent favorable risk factors. CONCLUSION: Post-relapse survival for primary localized RMA is generally poor. However, certain patient groups differed significantly in their likelihood of survival and 50% of patients with circumscribed relapses treated with ALRT survived. These findings may form the basis for an evidence-based risk-stratification for recurrent disease including relapse treatment.


Subject(s)
Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/therapy , Rhabdomyosarcoma, Alveolar/mortality , Rhabdomyosarcoma, Alveolar/therapy , Adolescent , Adult , Child , Child, Preschool , Disease-Free Survival , Female , Follow-Up Studies , Humans , Infant , Infant, Newborn , Male , Retrospective Studies , Survival Rate , Time Factors
13.
Klin Padiatr ; 224(6): 353-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22821288

ABSTRACT

BACKGROUND: Risk stratification criteria for patients with Ewing's sarcoma family of tumors (ESFT) are still limited. We hypothesized divergent human leukocyte antigen (HLA) patterns in ESFT patients and compared HLA-A, -B and -DR phenotype frequencies of patients with advanced ESFT with those of healthy controls. PATIENTS: HLA types of all German Caucasian patients with advanced ESFT and available HLA-A, -B and -DR data registered in the European Group for Blood and Marrow Transplantation, Paediatric Registry for Stem Cell Transplantation and the MetaEICESS data bases (study group, n=30) were retrospectively compared with HLA types of healthy German stem cell donors (control group, n=8 862 for single HLA frequencies and n=8 839 for allele combinations). Study group patients had been immuno-typed due to eligibility for allogeneic stem cell transplantation for high risk of treatment failure, and thus constituted a selected subgroup of ESFT patients. RESULTS: After Bonferroni correction for multiple testing (PC), phenotype frequencies of HLA-A24 remained significantly higher in the study group compared to controls (PC<0.05). Furthermore, several HLA combinations were significantly more frequent in the study group compared to controls (all PC<0.05). CONCLUSION: We report an increased incidence of circumscribed HLA patterns in German Caucasians with advanced ESFT. The possible clinical significance of this observation has to be re-assessed in prospective trials comprising larger ESFT patient numbers of all risk groups.


Subject(s)
Blood Donors , Bone Marrow Transplantation , Bone Neoplasms/genetics , Bone Neoplasms/therapy , HLA-A Antigens/genetics , HLA-B Antigens/genetics , HLA-DR Antigens/genetics , Hematopoietic Stem Cell Transplantation , Sarcoma, Ewing/genetics , Sarcoma, Ewing/therapy , Tissue Donors , Adolescent , Adult , Bone Neoplasms/pathology , Child , Disease Progression , Female , Gene Frequency , Genetics, Population , Germany , Humans , Male , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/therapy , Neoplasm Staging , Retrospective Studies , Sarcoma, Ewing/pathology , Young Adult
14.
Ann Oncol ; 22(7): 1614-1621, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21245159

ABSTRACT

BACKGROUND: Outcomes of Ewing tumor (ET) patients treated with allogeneic stem cell transplantation (allo-SCT) were compared regarding the use of reduced-intensity conditioning (RIC) and high-intensity conditioning (HIC) regimens as well as human leukocyte antigen (HLA)-matched and HLA-mismatched grafts. PATIENTS AND METHODS: We retrospectively analyzed data of 87 ET patients from the European Group for Blood and Marrow Transplantation, Pediatric Registry for Stem Cell Transplantations, Asia Pacific Blood and Marrow Transplantation and MetaEICESS registries treated with allo-SCT. Fifty patients received RIC (group A) and 37 patients received HIC (group B). Twenty-four patients received HLA-mismatched grafts and 63 received HLA-matched grafts. RESULTS: Median overall survival was 7.9 months [±1.24, 95% confidence interval (CI) 5.44-10.31] for group A and 4.4 months (±1.06, 95% CI 2.29-6.43) for group B patients (P = 1.3). Death of complications (DOC) occurred in 4 of 50 (0.08) and death of disease (DOD) in 33 of 50 (0.66) group A and in 16 of 37 (0.43) and 17 of 37 (0.46) group B patients, respectively. DOC incidence was decreased (P < 0.01) and DOD/relapse increased (P < 0.01) in group A compared with group B. HLA mismatch was not generally associated with graft-versus-Ewing tumor effect (GvETE). CONCLUSIONS: There was no improvement of survival with RIC compared with HIC due to increased DOD/relapse incidence after RIC despite less DOC incidence. This implicates general absence of a clinically relevant GvETE with current protocols.


Subject(s)
Bone Neoplasms/mortality , Bone Neoplasms/therapy , Graft vs Host Disease/therapy , Sarcoma, Ewing/mortality , Sarcoma, Ewing/therapy , Stem Cell Transplantation , Adolescent , Adult , Child , Child, Preschool , Female , Follow-Up Studies , Humans , Male , Middle Aged , Retrospective Studies , Survival Rate , Transplantation Conditioning , Transplantation, Homologous , Treatment Outcome , Young Adult
15.
Pathologe ; 31 Suppl 2: 215-20, 2010 Oct.
Article in German | MEDLINE | ID: mdl-20730458

ABSTRACT

AIMS: To improve survival of patients with advanced rhabdomyosarcomas (RMS), we aimed to adoptively transfer T-cells with redirected specificity for the fetal acetylcholine receptor (AChR), an RMS-specific cell surface antigen. METHODS: A "second generation" chimeric antigen receptor (CAR) with a combined CD28-CD3ζ signaling domain was derived from our previously described chimeric antigen receptor composed of an extracellular human anti-fAChR antibody fragment, an Fc hinge region, and the intracellular T-cell receptor zeta chain. Lymphocytes from the peripheral blood were modified by retroviral transduction and monitored by FACS analysis. Cytotoxicity of modified T-cells towards RMS cells was recorded by MTT-based viability tests; expression of co-stimulatory molecules and anti-apoptotic genes was studied by FACS and qRT-PCR analysis. RESULTS: Co-stimulatory molecules were expressed in low levels on RMS cells giving the rationale to generate a CD28-CD3ζ signalling CAR (chimeric antigen receptor) for redirecting T-cells. T-cells were successfully engineered with the "second generation" AChR-specific chimeric antigen receptor. Despite of high CAR expression engineered T-cells showed low killing efficiency towards RMS compared to redirected killing of CD20+ lymphoma or CEA-expressing adenocarcinoma cell lines when redirected by CD20- and/or CEA-specific CAR. CONCLUSIONS: Data suggest that RMS cells exhibit resistance to a T-cell attack redirected by a fAChR-specific CAR. Inhibition of anti-apoptotic pathways in those cells may improve sensitivity to conventional as well as T-cell-based therapeutics.


Subject(s)
Immunotherapy, Adoptive/methods , Rhabdomyosarcoma/therapy , T-Lymphocytes/immunology , Cell Line, Tumor , Chimerism , Cytotoxicity Tests, Immunologic , Humans , Immunoglobulin Fc Fragments/immunology , Immunoglobulin Fragments/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Cholinergic/immunology , Rhabdomyosarcoma/immunology , Rhabdomyosarcoma/pathology
17.
J Biomed Biotechnol ; 2010: 187621, 2010.
Article in English | MEDLINE | ID: mdl-20204062

ABSTRACT

Rhabdomyosarcoma (RMS) is the most common malignant soft tissue tumor in children and is highly resistant to all forms of treatment currently available once metastasis or relapse has commenced. As it has recently been determined that the acetylcholine receptor (AChR) gamma-subunit, which defines the fetal AChR (fAChR) isoform, is almost exclusively expressed in RMS post partum, we recombinantly fused a single chain variable fragment (scFv) derived from a fully human anti-fAChR Fab-fragment to Pseudomonas exotoxin A to generate an anti-fAChR immunotoxin (scFv35-ETA). While scFv35-ETA had no damaging effect on fAChR-negative control cell lines, it killed human embryonic and alveolar RMS cell lines in vitro and delayed RMS development in a murine transplantation model. These results indicate that scFv35-ETA may be a valuable new therapeutic tool as well as a relevant step towards the development of a fully human immunotoxin directed against RMS. Moreover, as approximately 20% of metastatic malignant melanomas (MMs) display rhabdoid features including the expression of fAChR, the immunotoxin we developed may also prove to be of significant use in the treatment of these more common and most often fatal neoplasms.


Subject(s)
ADP Ribose Transferases/administration & dosage , Autoantibodies/immunology , Bacterial Toxins/administration & dosage , Exotoxins/administration & dosage , Immunotoxins/administration & dosage , Receptors, Nicotinic/immunology , Recombinant Fusion Proteins/administration & dosage , Rhabdomyosarcoma/drug therapy , Single-Chain Antibodies/immunology , Virulence Factors/administration & dosage , ADP Ribose Transferases/genetics , Animals , Autoantibodies/administration & dosage , Autoantibodies/genetics , Bacterial Toxins/genetics , Cell Growth Processes/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Drug Delivery Systems/methods , Exotoxins/genetics , Female , Flow Cytometry , Humans , Immunotoxins/genetics , Immunotoxins/immunology , Mice , Mice, SCID , Receptors, Nicotinic/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Rhabdomyosarcoma/immunology , Rhabdomyosarcoma/pathology , Single-Chain Antibodies/administration & dosage , Single-Chain Antibodies/genetics , Virulence Factors/genetics , Xenograft Model Antitumor Assays , Pseudomonas aeruginosa Exotoxin A
18.
Leukemia ; 23(9): 1634-42, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19357702

ABSTRACT

The Wilms tumor antigen, WT1, is expressed at high levels in various types of leukemia and solid tumors, including lung, breast, colon cancer and soft tissue sarcomas. The WT1 protein has been found to be highly immunogenic, and spontaneous humoral and cytotoxic T-cell responses have been detected in patients suffering from leukemia. Furthermore, major histocompatibility complexes class I- and II-restricted WT1 peptide epitopes have been shown to elicit immune responses in patients with WT1-expressing tumors. As a consequence, WT1 has become an attractive target for anticancer immunotherapy. In this study, we investigated the feasibility of generating WT1-specific T cells for adoptive immunotherapy after allogeneic stem cell transplantation. We analyzed the incidence of T cells specific for WT1 peptide epitopes in cancer patients and healthy volunteers. It is noted that we could generate WT1-specific responses in nine of ten healthy volunteer donors and established T-cell clones specific for two WT1-derived peptide epitopes. These in vitro expanded WT1-specific T cells effectively lysed WT1-expressing tumor cell lines, indicating the potential clinical impact of ex vivo expanded donor-derived WT1-specific T cells for adoptive immunotherapy after allogeneic stem cell transplantation.


Subject(s)
Hematopoietic Stem Cell Transplantation , Immunotherapy, Adoptive , Leukemia/therapy , Sarcoma/therapy , T-Lymphocytes, Cytotoxic/immunology , WT1 Proteins/immunology , Cell Line, Tumor , Epitopes , HLA-A2 Antigen/analysis , Humans , Immunophenotyping , Orthomyxoviridae/immunology , Transplantation, Homologous , WT1 Proteins/genetics
19.
Klin Padiatr ; 219(6): 318-22, 2007.
Article in English | MEDLINE | ID: mdl-18050041

ABSTRACT

In nine patients (pts) with soft tissue sarcoma refractory to conventional therapy (incomplete response or relapse) intensified chemotherapy was administered combining 0.75 mg/m (2) topotecan, 100 mg/m (2) etoposide, 100 mg/m (2) carboplatin and 200 mg/m (2) cyclophosphamide on day 1-5 (TECC). To avoid prolonged intervals between the serial TECC courses autologous hematopoietic stem cell supports (median 1.0 x 10(6) CD34+ cells per kg body weight (bw), range 0.5-2.8 x 10(6) CD34+ cells/kg bw, SD 0.6 x 10(6) CD34+ cells/kg bw) were given on day 7. All pts received granulocyte colony stimulating factor (GCSF) from day 8 in addition. All together 39 TECC courses (minimum 2 courses, maximum 6 courses per pt) were administered, with a median interval of 32 (range 21-52) days until recovery. Leukopenia (<1000/microl) occurred 9 days (range 3-13 days; SD 2.4 days) after end of chemotherapy and persisted for 9 (range 3-15 days; SD 3 days) days. In 31/39 TECC courses readmission to hospital was required for supportive therapy mainly due to neutropenic fever. In this period pts received 0.83 (range 0-1) red blood cell units and 2.35 (range 1-4) platelet units. C-reactive protein in neutropenic pts as an indicator for infection after TECC chemotherapy was detectable after 36 of 39 chemotherapy courses leading to further supportive therapy (median 10.4 mg/dl, range 1.1-28.3 mg/dl; SD 6.67 mg/dl). Duration of total inpatient treatment per TECC course including supportive therapy was in median 13.5 days (range 7-53 days; SD 4.3 days). Only two children had a prolonged infection (77 and 100 days). Clinical and objective tumor responses, defined as complete remission, very good partial response and partial response were observed in 9/9 pts at eight weeks after the last TECC course and were maintained at six months in 7/9 pts. Median time to progression and median overall survival time after TECC chemotherapy were 20.3 months and 25.2 months, respectively. These data provide evidence that in very high risk pts refractory to standard high risk therapy, a combination of TECC chemotherapy and stem cell support is feasible in pts with incomplete remission respectively relapsed STS pts and demonstrates promising antineoplastic activity. Therefore, this regimen warrants further investigation in prospective trials.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Hematopoietic Stem Cell Transplantation , Sarcoma/therapy , Adolescent , Adult , Carboplatin/administration & dosage , Child , Child, Preschool , Cyclophosphamide/administration & dosage , Etoposide/administration & dosage , Feasibility Studies , Female , Follow-Up Studies , Humans , Infant , Magnetic Resonance Imaging , Male , Neoplasm, Residual/surgery , Remission Induction , Sarcoma/diagnosis , Sarcoma/drug therapy , Time Factors , Topotecan/administration & dosage , Transplantation, Autologous , Treatment Outcome
20.
Clin Endocrinol (Oxf) ; 66(5): 727-31, 2007 May.
Article in English | MEDLINE | ID: mdl-17381483

ABSTRACT

OBJECTIVE: The role of chemotherapy in thyroid sequelae after cancer treatment has not been studied systematically, especially in sarcoma patients. The aim of this study was to determine the incidence of post-therapeutic thyroid disorders and their contributing factors in a cohort of paediatric sarcoma patients. DESIGN: Late effects of sarcoma treatment have been collected prospectively within the Late Effects Surveillance System (LESS) in Germany, Austria and Switzerland since 1998. PATIENTS: We studied 340 relapse-free paediatric patients (median age at diagnosis 12.2 [interquartile range (IQR) = 7.3-15.6 years] treated for osteosarcoma, soft tissue sarcoma or Ewing's sarcoma within the COSS-96, CWS-96/CWS-2002P or EICESS-92/EURO-E.W.I.N.G.-99 therapy trials. In addition to polychemotherapy, 127 patients were irradiated (mean cumulative dose 47 +/- 9.7 Gy), including 51 patients with irradiation to the head/neck region. Median follow-up was 24.6 (IQR = 11.9-44.9) months. MEASUREMENTS: We reviewed the results of yearly examinations of serum TSH and fT4 levels and thyroid ultrasound examinations. RESULTS: The incidence of thyroid disorders was 37% (19/51, 95% CI 24-52%) in patients with head/neck irradiation, and 11% (32/289, 95% CI 8-15%) in patients without irradiation to the head/neck. Thyroid disorders were more frequent in patients treated with idarubicin (P = 0.027) and trofosfamide (P = 0.016). We also found a significant association between raised TSH levels and treatment with trofosfamide (P = 0.008) or idarubicin (P = 0.037) (n = 250). CONCLUSIONS: The incidence of thyroid disorders in the head/neck-irradiated group was high. Even without head/neck irradiation, we found an increased proportion of patients with thyroid disorders, possibly as a result of chemotherapy.


Subject(s)
Sarcoma/therapy , Thyroid Diseases/physiopathology , Thyroid Gland/physiopathology , Adolescent , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Child , Cyclophosphamide/adverse effects , Cyclophosphamide/analogs & derivatives , Dactinomycin/adverse effects , Female , Follow-Up Studies , Humans , Ifosfamide/adverse effects , Ifosfamide/therapeutic use , Incidence , Male , Multivariate Analysis , Sarcoma/complications , Thyroid Diseases/drug therapy , Thyroid Diseases/etiology , Thyroid Function Tests , Thyroid Gland/drug effects , Thyroid Gland/radiation effects , Thyrotropin/blood , Thyroxine/blood , Thyroxine/therapeutic use , Vincristine/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL