Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Pediatr Transplant ; 23(4): e13408, 2019 06.
Article in English | MEDLINE | ID: mdl-30955249

ABSTRACT

BACKGROUND: For pediatric ALL patients that relapse or respond poorly to conventional chemotherapy treatment, HSCT is one treatment option. Still, relapse occurs in 30% of the children after HSCT. Mutations in the tumor suppressor gene TP53 which can lead to an altered p53 protein expression are rare at time of diagnosis of ALL, yet occur more frequent at relapse indicating a more aggressive disease. Our aim was to evaluate if alterations in the expression of the tumor suppressor protein p53 signaled a relapse in pediatric ALL patients post-HSCT and could guide for preemptive immunotherapy. PROCEDURE: Paraffin-embedded bone marrow samples from 46 children diagnosed with ALL between 1997 and 2010, and transplanted at Karolinska University Hospital, were analyzed for p53 by IHC. Samples were analyzed independently at diagnosis, before HSCT, and after HSCT 0-3 months, 3-6 months, and 6-12 months. RESULT: Strong expression of p53 in the bone marrow at 0-3-months after HSCT was associated with increased risk of relapse, odds ratio 2.63 (confidence interval 1.08-6.40) P = 0.033. CONCLUSION: Evaluation of p53 protein expression in bone marrow from pediatric ALL patients that undergo HSCT may be a potential, additional prognostic marker for predicting relapse after HSCT.


Subject(s)
Bone Marrow/metabolism , Hematopoietic Stem Cell Transplantation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Tumor Suppressor Protein p53/metabolism , Adolescent , Child , Child, Preschool , Female , Gene Expression Regulation, Leukemic , Humans , Immunohistochemistry , Immunotherapy , Infant , Infant, Newborn , Male , Mutation , Odds Ratio , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Prognosis , Recurrence , Risk , Sweden
2.
Pediatr Blood Cancer ; 65(11): e27310, 2018 11.
Article in English | MEDLINE | ID: mdl-29968961

ABSTRACT

SCL/TAL1 interrupting locus (STIL)-T-cell acute leukaemia (TAL1) fusion genes are present in approximately 11-27% of children with paediatric T-cell acute lymphoblastic leukaemia (T-ALL), but the developmental timing of the rearrangement is still unknown. To investigate whether the fusion gene can be detected in neonatal blood spots (NBSs) from paediatric patients diagnosed with T-cell ALL, we analysed DNA from 38 paediatric patients with T-ALL by nested polymerase chain reaction and electrophoresis. The STIL-TAL1 fusion gene was not detected in NBSs from any of the 38 patients with T-ALL, suggesting that STIL-TAL1 fusion genes are most probably postnatal events in paediatric T-ALL.


Subject(s)
Intracellular Signaling Peptides and Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Adolescent , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male
3.
Pediatr Transplant ; 19(7): 767-75, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26432780

ABSTRACT

Dysregulation of genes involved in the cell cycle such as TP53, P21, P16, and PTEN plays a key role in oncogenesis. We have earlier reported increased expression of the TP53 encoded protein p53, in bone marrow samples from pediatric patients with more aggressive, rare chronic myeloid malignancies. The aim of this study was to investigate protein expression of p53, p21, p16, and PTEN before and after HSCT in pediatric patients with AML and evaluate whether any potential alterations could predict relapse after HSCT. Paraffin-embedded bone marrow samples from 34 pediatric patients with AML were collected retrospectively from time of diagnosis as well as pre- and post-HSCT. IHC was performed on tissue microarrays with antibodies against p53, p21, p16, and PTEN. Study material was analyzed by independent t-tests and nonlinear regression. t-Tests showed a statistical significant difference in p53 (p = 0.010) with overexpression in the group of patients who relapsed compared to the relapse-free patients at >3-6 months post-HSCT. Analysis of p53 protein expression by IHC may be a potential predictor for relapse after HSCT in children with AML.


Subject(s)
Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid, Acute/therapy , Tumor Suppressor Protein p53/metabolism , Adolescent , Biomarkers/metabolism , Bone Marrow/metabolism , Child , Child, Preschool , Female , Humans , Infant , Leukemia, Myeloid, Acute/metabolism , Logistic Models , Male , Recurrence , Retrospective Studies , Tissue Array Analysis , Transplantation, Homologous , Treatment Outcome
4.
Pediatr Hematol Oncol ; 31(4): 327-39, 2014 May.
Article in English | MEDLINE | ID: mdl-24761810

ABSTRACT

BACKGROUND: Alterations in the tumor suppressor gene TP53 have been associated with poor outcome in adult hematological malignancies. We have earlier reported an increased expression of the TP53 encoded protein p53, in bone marrow samples from pediatric patients with aggressive leukemia. Our aim was now to evaluate p53 protein expression at different time points before and after hematopoietic stem cell transplantation (HSCT) as a predictor of relapse in a group of children diagnosed with MDS, JMML and CML, and also investigate if potential alterations in expression could be correlated to mutations in TP53. PROCEDURE: Paraffin embedded bone marrow samples from 33 pediatric patients diagnosed with MDS, JMML and CML between 1997 and 2010 were collected retrospectively from time of diagnosis and pre and post HSCT. Immunohistochemistry (IHC) was performed on tissue microarrays (TMA) with antibodies to p53 and p21. DNA sequencing of exon 2-11 of TP53 was performed in 7 patients with JMML and 5 patients with MDS. RESULTS: Elevated p53 protein expression at diagnosis predicted for relapse, odds ratio (OR) 1.19 (95% CI: 1.02-1.40, p = .028). Sequencing of TP53 did not reveal any mutations in the 12 patients analyzed and p53 expression correlated positively to p21 expression indicating a functional p53/p21 protein pathway. CONCLUSION: Elevated p53 protein expression at diagnosis may be an indicator of relapse in children with MDS, JMML and CML.


Subject(s)
Gene Expression Regulation , Hematologic Neoplasms/metabolism , Tumor Suppressor Protein p53/biosynthesis , Adolescent , Adult , Child , Child, Preschool , Chronic Disease , Female , Hematologic Neoplasms/genetics , Hematologic Neoplasms/pathology , Hematologic Neoplasms/therapy , Humans , Infant , Infant, Newborn , Male , Mutation , Predictive Value of Tests , Recurrence , Retrospective Studies , Tumor Suppressor Protein p53/genetics
5.
J Pediatr Hematol Oncol ; 34(5): 364-7, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22713707

ABSTRACT

BACKGROUND: Neonatal dried blood spots (Guthrie cards) have been used to demonstrate a prenatal origin of clonal leukemia-specific genetic aberrations in several subgroups of childhood acute lymphoblastic leukemia (ALL). One hypothesis suggests that an infectious agent could initiate genetic transformation already in utero. In search for a possible viral agent, Guthrie cards were analyzed for the presence of 3 newly discovered polyomavirus Karolinska Institutet polymavirus (KIPyV), Washington University polyomavirus (WUPyV), and Merkel cell polyomavirus (MCPyV). METHODS: Guthrie cards from 50 children who later developed ALL and 100 matched controls were collected and analyzed by standard or real-time polymerase chain reaction for the presence of the VP1 region of KIPyV, WUPyV, and MCPyV, and the LT region for MCPyV. RESULTS AND CONCLUSIONS: DNA from KIPyV, WUPyV, and MCPyV was not detected in neonatal blood samples from children with ALL or controls. Prenatal infections with these viruses are not likely to be etiological drivers for childhood leukemogenesis.


Subject(s)
DNA, Viral/blood , Merkel cell polyomavirus/isolation & purification , Polyomavirus/isolation & purification , Precursor Cell Lymphoblastic Leukemia-Lymphoma/virology , Adolescent , Child , Child, Preschool , Core Binding Factor Alpha 2 Subunit , Female , Humans , Infant , Infant, Newborn , Male , Oncogene Proteins, Fusion/genetics , Polymerase Chain Reaction , Precursor Cell Lymphoblastic Leukemia-Lymphoma/etiology
6.
Acta Paediatr ; 96(11): 1660-4, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17888058

ABSTRACT

AIM: Aplastic anaemia following hepatitis may develop in as many as 1 of 3 patients with non-A, non-B and non-C hepatitis. Several causative factors have been discussed, such as viral infections and autoimmunity. Here we describe the natural history of this condition in 7 children and investigate possible hepatitis-causing agents. METHODS: We reviewed the medical records, bone marrow and liver biopsies of 7 children with severe hepatitis, with or without liver failure, who subsequently had developed aplastic anaemia. RESULTS: The median time from onset of hepatic symptoms until diagnosed onset of aplasia was 54 days. No associated viral infections could be identified. On liver biopsy, a majority had lobular inflammation but lacked signs of autoimmune hepatitis, findings compatible with a viral aetiology. Three of 6 children had low reticulocyte counts already at onset of hepatitis. All, but one patient is alive at median follow-up of 8 years. CONCLUSION: The unknown pathogenetic mechanism appears to target liver and bone marrow simultaneously, because half of the children concomitantly had low reticulocyte counts and severe liver failure.


Subject(s)
Anemia, Aplastic/etiology , Hepatitis/complications , Liver Failure/etiology , Adolescent , Anemia, Aplastic/therapy , Biopsy , Bone Marrow Cells/pathology , Bone Marrow Transplantation , Child , Child, Preschool , Female , Hepatitis/pathology , Hepatitis/physiopathology , Humans , Liver/pathology , Male , Medical Records , Parvovirus/pathogenicity , Retrospective Studies , Serologic Tests , Sweden , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL