Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
1.
Immunol Cell Biol ; 102(6): 513-525, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38726587

ABSTRACT

We studied the associations between inflammation-related proteins in circulation and complications after pediatric allogenic hematopoietic stem cell transplantation (HSCT), to reveal proteomic signatures or individual soluble proteins associated with specific complications after HSCT. We used a proteomics method called Proximity Extension Assay to repeatedly measure 180 different proteins together with clinical variables, cellular immune reconstitution and blood viral copy numbers in 27 children (1-18 years of age) during a 2-year follow-up after allogenic HSCT. Protein profile analysis was performed using unsupervised hierarchical clustering and a regression-based method, while the Bonferroni-corrected Mann-Whitney U-test was used for time point-specific comparison of individual proteins against outcome. At 6 months after allogenic HSCT, we could identify a protein profile pattern associated with occurrence of the complications such as chronic graft-versus-host disease, viral infections, relapse and death. When protein markers were analyzed separately, the plasma concentration of the inhibitory and cytotoxic T-cell surface protein FCRL6 (Fc receptor-like 6) was higher in patients with cytomegalovirus (CMV) viremia [log2-fold change 1.5 (P = 0.00099), 2.5 (P = 0.00035) and 2.2 (P = 0.045) at time points 6, 12 and 24 months]. Flow cytometry confirmed that FCRL6 expression was higher in innate-like γδ T cells, indicating that these cells are involved in controlling CMV reactivation in HSCT recipients. In conclusion, the potentially druggable FCRL6 receptor on cytotoxic T cells appears to have a role in controlling CMV viremia after HSCT. Furthermore, our results suggest that system-level analysis is a useful addition to the studying of single biomarkers in allogenic HSCT.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , Hematopoietic Stem Cell Transplantation , Proteomics , Transplantation, Homologous , Virus Activation , Humans , Hematopoietic Stem Cell Transplantation/adverse effects , Child , Child, Preschool , Proteomics/methods , Cytomegalovirus/immunology , Cytomegalovirus/physiology , Infant , Adolescent , Female , Male , Cytomegalovirus Infections/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Graft vs Host Disease/etiology , Graft vs Host Disease/immunology , Receptors, Fc/metabolism , Biomarkers
2.
J Allergy Clin Immunol ; 151(4): 1081-1095, 2023 04.
Article in English | MEDLINE | ID: mdl-36228738

ABSTRACT

BACKGROUND: In 2014, germline signal transducer and activator of transcription (STAT) 3 gain-of-function (GOF) mutations were first described to cause a novel multisystem disease of early-onset lymphoproliferation and autoimmunity. OBJECTIVE: This pivotal cohort study defines the scope, natural history, treatment, and overall survival of a large global cohort of patients with pathogenic STAT3 GOF variants. METHODS: We identified 191 patients from 33 countries with 72 unique mutations. Inclusion criteria included symptoms of immune dysregulation and a biochemically confirmed germline heterozygous GOF variant in STAT3. RESULTS: Overall survival was 88%, median age at onset of symptoms was 2.3 years, and median age at diagnosis was 12 years. Immune dysregulatory features were present in all patients: lymphoproliferation was the most common manifestation (73%); increased frequencies of double-negative (CD4-CD8-) T cells were found in 83% of patients tested. Autoimmune cytopenias were the second most common clinical manifestation (67%), followed by growth delay, enteropathy, skin disease, pulmonary disease, endocrinopathy, arthritis, autoimmune hepatitis, neurologic disease, vasculopathy, renal disease, and malignancy. Infections were reported in 72% of the cohort. A cellular and humoral immunodeficiency was observed in 37% and 51% of patients, respectively. Clinical symptoms dramatically improved in patients treated with JAK inhibitors, while a variety of other immunomodulatory treatment modalities were less efficacious. Thus far, 23 patients have undergone bone marrow transplantation, with a 62% survival rate. CONCLUSION: STAT3 GOF patients present with a wide array of immune-mediated disease including lymphoproliferation, autoimmune cytopenias, and multisystem autoimmunity. Patient care tends to be siloed, without a clear treatment strategy. Thus, early identification and prompt treatment implementation are lifesaving for STAT3 GOF syndrome.


Subject(s)
Immune System Diseases , Immunologic Deficiency Syndromes , Child , Humans , Autoimmunity/genetics , Cohort Studies , Gain of Function Mutation , Immunologic Deficiency Syndromes/genetics , Mutation , STAT3 Transcription Factor/genetics , Cell Proliferation , Lymphocytes
3.
Int J Cancer ; 152(11): 2292-2302, 2023 06 01.
Article in English | MEDLINE | ID: mdl-36760204

ABSTRACT

Allogeneic hematopoietic stem cell transplantation (aHSCT) represents a therapeutic choice for high-risk and relapsed leukemia at a young age. In this retrospective population-based study, we evaluated cardiovascular complications after aHSCT (N = 272) vs conventional therapy (N = 1098) among patients diagnosed with acute lymphoblastic or acute myeloid leukemia below 35 years between 1985 and 2004. Additionally, siblings from a prior comparison group served as population controls (N = 39 217). Childhood leukemia and aHSCT was associated with a 16-fold HR for developing arterial hypertension (HR 16.8, 95%CI 1.5-185.5) compared with conventional therapy. A 2-fold HR for any cardiovascular complication was observed after AYA leukemia and aHSCT vs conventional treatment (HR 2.7, 95% CI 1.4-5.1). After AYA leukemia and aHSCT, the HR of cardiac arrhythmia was significantly elevated vs conventional therapy (HR 14.4, 95% CI 1.5-125.2). Moreover, after aHSCT in childhood, elevated hazard ratios (HRs) were found for cardiomyopathy/ cardiac insufficiency (HR 105.0, 95% CI 10.0-1100.0), cardiac arrhythmia, and arterial hypertension (HR 20.1, 95%CI 2.5-159.7 and HR 20.0, 95%CI 4.1-97.4) compared with healthy controls. After adolescent and young adult (AYA) leukemia and aHSCT, markedly increased HRs were observed for cardiac arrhythmia (HR 29.2, 95%CI 6.6-129.2), brain vascular thrombosis/ atherosclerosis and cardiomyopathy/cardiac insufficiency (HR 23.4, 95%CI 7.1-77.4 and HR 19.2, 95%CI 1.5-245.2) compared with healthy controls. As the cumulative incidence for cardiovascular complications rose during the follow-up of childhood and AYA leukemia patients, long-term cardiovascular surveillance is warranted to optimize the quality of life after childhood and AYA leukemia following both conventional treatment and aHSCT.


Subject(s)
Cardiovascular Diseases , Hematopoietic Stem Cell Transplantation , Hypertension , Leukemia, Myeloid, Acute , Humans , Adolescent , Young Adult , Retrospective Studies , Finland/epidemiology , Quality of Life , Leukemia, Myeloid, Acute/epidemiology , Leukemia, Myeloid, Acute/therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Cardiovascular Diseases/etiology , Cardiovascular Diseases/complications , Hypertension/etiology , Hypertension/complications
4.
Blood ; 137(15): 2033-2045, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33513601

ABSTRACT

Exocytosis of cytotoxic granules (CG) by lymphocytes is required for the elimination of infected and malignant cells. Impairments in this process underly a group of diseases with dramatic hyperferritinemic inflammation termed hemophagocytic lymphohistiocytosis (HLH). Although genetic and functional studies of HLH have identified proteins controlling distinct steps of CG exocytosis, the molecular mechanisms that spatiotemporally coordinate CG release remain partially elusive. We studied a patient exhibiting characteristic clinical features of HLH associated with markedly impaired cytotoxic T lymphocyte (CTL) and natural killer (NK) cell exocytosis functions, who beared biallelic deleterious mutations in the gene encoding the small GTPase RhoG. Experimental ablation of RHOG in a model cell line and primary CTLs from healthy individuals uncovered a hitherto unappreciated role of RhoG in retaining CGs in the vicinity of the plasma membrane (PM), a fundamental prerequisite for CG exocytotic release. We discovered that RhoG engages in a protein-protein interaction with Munc13-4, an exocytosis protein essential for CG fusion with the PM. We show that this interaction is critical for docking of Munc13-4+ CGs to the PM and subsequent membrane fusion and release of CG content. Thus, our study illuminates RhoG as a novel essential regulator of human lymphocyte cytotoxicity and provides the molecular pathomechanism behind the identified here and previously unreported genetically determined form of HLH.


Subject(s)
Killer Cells, Natural/pathology , Lymphohistiocytosis, Hemophagocytic/genetics , T-Lymphocytes, Cytotoxic/pathology , rho GTP-Binding Proteins/genetics , Cell Line , Cells, Cultured , Gene Deletion , Germ-Line Mutation , Humans , Infant , Killer Cells, Natural/metabolism , Lymphohistiocytosis, Hemophagocytic/pathology , Male , Models, Molecular , T-Lymphocytes, Cytotoxic/metabolism , rho GTP-Binding Proteins/chemistry
5.
Pediatr Res ; 93(7): 1983-1989, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36284141

ABSTRACT

BACKGROUND: Chronic lung problems are a rare but serious complication of allogeneic hematopoietic stem cell transplantation (HSCT). We studied clinical phenotypes and polysomnography appearance of breathing abnormality in late onset non-infectious pulmonary complications (NIPS). METHODS: We reviewed Finnish national reference database between the years 1999 and 2016. We identified 12 children with most severely decreased pulmonary function and performed polysomnography and 24 aged-matched controls out of 325 performed pediatric allogeneic HSCTs. RESULTS: All patients with NIPS had severely decreased pulmonary function already at 6 months post HSCT with median FEV1 value 42% (interquartile range (IQR) 30-52%) of predicted normal values. Seven children had obstructive and five children more restrictive lung function. Children with obstructive lung function showed laborious breathing (7/7), decreased oxygenation and ventilation-to-perfusion mismatch (6/7), or REM-sleep-related hypoventilation (4/7) on polysomnography. Children with restrictive lung function (5/12) did not show sleep-related breathing disorder. CONCLUSIONS: Children going through allogeneic HSCT who develop severe chronic obstructive lung function are more likely to present with sleep-related hypoxia and hypoventilation than children with restrictive lung function. IMPACT: Children with severe obstructive lung function and chronic lung graft-versus-host disease following hematopoietic stem cell transplantation are more likely to present with sleep-related mild hypoxia and hypoventilation than children with restrictive lung disease. To our knowledge there are no reports on sleep-related breathing disorders and ventilatory function measured by polysomnography in children with pulmonary complications after allogeneic HSCT. Polysomnography may add to the differential diagnostics between patients with BOS and other non-infectious pulmonary complications.


Subject(s)
Hematopoietic Stem Cell Transplantation , Respiration Disorders , Sleep Wake Disorders , Humans , Hypoventilation , Sleep , Hematopoietic Stem Cell Transplantation/adverse effects , Stem Cell Transplantation , Hypoxia
6.
Blood ; 136(10): 1201-1211, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32614953

ABSTRACT

Chronic granulomatous disease (CGD) is a primary immunodeficiency resulting in life-threatening infections and inflammatory complications. Allogeneic hematopoietic cell transplantation (allo-HCT) can cure the disease, but the indication to transplant remains controversial. We performed a retrospective multicenter study of 712 patients with CGD who underwent allo-HCT transplantation from March 1993 through December 2018. We studied 635 children (aged <18 years) and 77 adults. Median follow-up was 45 months. Median age at transplantation was 7 years (range, 0.1-48.6). Kaplan-Meier estimates of overall survival (OS) and event-free survival (EFS) at 3 years were 85.7% and 75.8%, respectively. In multivariate analysis, older age was associated with reduced survival and increased chronic graft-versus-host disease. Nevertheless, OS and EFS at 3 years for patients ≥18 years were 76% and 69%, respectively. Use of 1-antigen-mismatched donors was associated with reduced OS and EFS . No significant difference was found in OS, but a significantly reduced EFS was noted in the small group of patients who received a transplant from a donor with a >1 antigen mismatch. Choice of conditioning regimen did not influence OS or EFS. In summary, we report an excellent outcome after allo-HCT in CGD, with low incidence of graft failure and mortality in all ages. Older patients and recipients of 1-antigen-mismatched grafts had a less favorable outcome. Transplantation should be strongly considered at a younger age and particularly in the presence of a well-matched donor.


Subject(s)
Granulomatous Disease, Chronic/therapy , Hematopoietic Stem Cell Transplantation/mortality , Adolescent , Adult , Child , Child, Preschool , Female , Follow-Up Studies , Granulomatous Disease, Chronic/pathology , Hematopoietic Stem Cell Transplantation/methods , Humans , Infant , Male , Middle Aged , Prognosis , Retrospective Studies , Survival Rate , Transplantation Conditioning , Transplantation, Homologous , Young Adult
7.
Haematologica ; 107(10): 2318-2328, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35354251

ABSTRACT

Central nervous system (CNS) toxicity is common at diagnosis and during treatment of pediatric acute lymphoblastic leukemia (ALL). We studied CNS toxicity in 1,464 children aged 1.0-17.9 years, diagnosed with ALL and treated according to the Nordic Society of Pediatric Hematology and Oncology ALL2008 protocol. Genome-wide association studies, and a candidate single-nucleotide polymorphism (SNP; n=19) study were performed in 1,166 patients. Findings were validated in an independent Australian cohort of children with ALL (n=797) in whom two phenotypes were evaluated: diverse CNS toxicities (n=103) and methotrexate-related CNS toxicity (n=48). In total, 135/1,464 (9.2%) patients experienced CNS toxicity for a cumulative incidence of 8.7% (95% confidence interval: 7.31-10.20) at 12 months from diagnosis. Patients aged ≥10 years had a higher risk of CNS toxicity than had younger patients (16.3% vs. 7.4%; P<0.001). The most common CNS toxicities were posterior reversible encephalopathy syndrome (n=52, 43 with seizures), sinus venous thrombosis (n=28, 9 with seizures), and isolated seizures (n=16). The most significant SNP identified by the genome-wide association studies did not reach genomic significance (lowest P-value: 1.11x10-6), but several were annotated in genes regulating neuronal functions. In candidate SNP analysis, ATXN1 rs68082256, related to epilepsy, was associated with seizures in patients <10 years (P=0.01). ATXN1 rs68082256 was validated in the Australian cohort with diverse CNS toxicities (P=0.04). The role of ATXN1 as well as the novel SNP in neurotoxicity in pediatric ALL should be further explored.


Subject(s)
Posterior Leukoencephalopathy Syndrome , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Humans , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Australia , Central Nervous System , Genome-Wide Association Study , Genotype , Methotrexate/adverse effects , Phenotype , Posterior Leukoencephalopathy Syndrome/chemically induced , Posterior Leukoencephalopathy Syndrome/complications , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Risk Factors , Seizures/chemically induced , Seizures/complications
8.
Acta Oncol ; 61(11): 1309-1316, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36437562

ABSTRACT

BACKGROUND: Adolescents with chronic diseases are shown to be vulnerable for risky sexual behavior. Childhood cancer patients seem to engage in risky health behaviors as frequently as general population, but little is known about sexual issues in this group of patients. MATERIAL AND METHODS: We characterized the risk for sexually transmitted diseases (STD) in a Finnish population-based cohort of over 6,000 childhood cancer patients diagnosed with cancer under the age of 20 years between 1971 and 2009, compared with over 30,000 age- and sex -matched population comparisons. The data were constructed through linkage between national cancer, population, infectious diseases, and hospital discharge registries. We estimated hazard ratios (HRs) with 95% confidence intervals (CIs) using Cox regression modeling with attained age as the underlying time scale. RESULTS: Childhood cancer patients had a decreased risk for having an infection with chlamydia, the most common STD in our cohort, when comparing with population comparisons (HR 0.77, 95% CI 0.69-0.86). The risk was lowest among male patients (HR 0.64, 95% CI 0.53-0.79) and patients with central nervous system (CNS) tumors (HR 0.46, 95% CI 0.33-0.63). The overall risk for cervical dysplasia was slightly increased among female cancer patients when compared with their population comparisons (HR 1.28, 95% CI 1.02-1.60). Greatest risk elevation was found among patients diagnosed with cancer in ages 10-14 years (HR 2.31, 95% CI 1.46-3.65) and patients with lymphoma (HR 1.95, 95% CI 1.20-3.16). The risk for all explored outcomes seemed to be decreased among patients with CNS tumors. CONCLUSIONS: Our findings highlight the importance of integrating sexual issues as a part of psychosocial support and having a systematic transition program in the follow-up care of childhood cancer patients.


Subject(s)
Neoplasms , Sexually Transmitted Diseases , Adolescent , Humans , Male , Female , Child , Young Adult , Adult , Cohort Studies , Finland/epidemiology , Risk Factors , Sexually Transmitted Diseases/epidemiology , Neoplasms/psychology , Registries
9.
Br J Haematol ; 195(1): 119-122, 2021 10.
Article in English | MEDLINE | ID: mdl-34396501

ABSTRACT

Infants with acute lymphoblastic leukaemia (ALL) have a high frequency of central nervous system (CNS) involvement. Flow cytometric analysis of cerebrospinal fluid (CSF) was recently demonstrated to be a sensitive method for detecting CNS involvement in childhood ALL. In the present study, CSF from 14 infants was collected at routine lumbar punctures and analysed by multicolour flow cytometry. At initial diagnosis, leukaemic blasts were detected in CSF by flow cytometry in 11 patients (78·6%) compared to seven patients (50%) by cytospin. Larger studies are needed to determine if CSF flow cytometry has prognostic value in infant ALL.


Subject(s)
Cerebrospinal Fluid/cytology , Flow Cytometry/methods , Leukemic Infiltration/diagnosis , Neoplastic Stem Cells/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/cerebrospinal fluid , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Female , Follow-Up Studies , Humans , Infant , Infant, Newborn , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Recurrence
10.
J Clin Immunol ; 41(7): 1633-1647, 2021 10.
Article in English | MEDLINE | ID: mdl-34324127

ABSTRACT

PURPOSE: Deficiency of adenosine deaminase 2 (DADA2) is an inherited inborn error of immunity, characterized by autoinflammation (recurrent fever), vasculopathy (livedo racemosa, polyarteritis nodosa, lacunar ischemic strokes, and intracranial hemorrhages), immunodeficiency, lymphoproliferation, immune cytopenias, and bone marrow failure (BMF). Tumor necrosis factor (TNF-α) blockade is the treatment of choice for the vasculopathy, but often fails to reverse refractory cytopenia. We aimed to study the outcome of hematopoietic cell transplantation (HCT) in patients with DADA2. METHODS: We conducted a retrospective study on the outcome of HCT in patients with DADA2. The primary outcome was overall survival (OS). RESULTS: Thirty DADA2 patients from 12 countries received a total of 38 HCTs. The indications for HCT were BMF, immune cytopenia, malignancy, or immunodeficiency. Median age at HCT was 9 years (range: 2-28 years). The conditioning regimens for the final transplants were myeloablative (n = 20), reduced intensity (n = 8), or non-myeloablative (n = 2). Donors were HLA-matched related (n = 4), HLA-matched unrelated (n = 16), HLA-haploidentical (n = 2), or HLA-mismatched unrelated (n = 8). After a median follow-up of 2 years (range: 0.5-16 years), 2-year OS was 97%, and 2-year GvHD-free relapse-free survival was 73%. The hematological and immunological phenotypes resolved, and there were no new vascular events. Plasma ADA2 enzyme activity normalized in 16/17 patients tested. Six patients required more than one HCT. CONCLUSION: HCT was an effective treatment for DADA2, successfully reversing the refractory cytopenia, as well as the vasculopathy and immunodeficiency. CLINICAL IMPLICATIONS: HCT is a definitive cure for DADA2 with > 95% survival.


Subject(s)
Agammaglobulinemia/therapy , Bone Marrow Failure Disorders/therapy , Hematopoietic Stem Cell Transplantation , Severe Combined Immunodeficiency/therapy , Adenosine Deaminase/deficiency , Adolescent , Adult , Agammaglobulinemia/enzymology , Agammaglobulinemia/genetics , Agammaglobulinemia/mortality , Bone Marrow Failure Disorders/enzymology , Bone Marrow Failure Disorders/genetics , Bone Marrow Failure Disorders/mortality , Child , Child, Preschool , Female , Graft vs Host Disease/etiology , Graft vs Host Disease/mortality , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Intercellular Signaling Peptides and Proteins/deficiency , Kaplan-Meier Estimate , Male , Retrospective Studies , Severe Combined Immunodeficiency/enzymology , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/mortality , Treatment Outcome , Young Adult
11.
J Med Genet ; 57(1): 18-22, 2020 01.
Article in English | MEDLINE | ID: mdl-31413121

ABSTRACT

BACKGROUND: Metaphyseal dysplasia without hypotrichosis (MDWH) is a rare form of chondrodysplasia with no extraskeletal manifestations. MDWH is caused by RMRP mutations, but it is differentiated from the allelic condition cartilage-hair hypoplasia (CHH), which in addition to chondrodysplasia is characterised by thin hair, immunodeficiency and increased risk of malignancy. The long-term outcome of MDWH remains unknown. OBJECTIVE: We diagnosed severe agranulocytosis in a subject with RMRP mutations and normal hair. Based on this observation, we hypothesised that MDWH may, similar to CHH, associate with immune deficiency and malignancy. METHODS: We collected clinical and laboratory data for a cohort of 80 patients with RMRP mutations followed for over 30 years and analysed outcome data for those with features consistent with MDWH. RESULTS: In our cohort, we identified 10 patients with skeletal but no extraskeletal features during preschool age. Eight of these patients developed malignancy or clinically significant immunodeficiency during follow-up. Two of them died during chemotherapy for malignancy. At the time of the first extraskeletal manifestation, patients were school aged, 20, 43 and 50 years old. Laboratory signs of immunodeficiency (impaired lymphocyte proliferative responses) were demonstrated in four patients before the onset of symptoms. The patient outside this cohort, who had RMRP mutations, skeletal dysplasia, normal hair and severe agranulocytosis at 18 years of age, underwent haematopoietic stem cell transplantation. CONCLUSIONS: MDWH can present with severe late-onset extraskeletal manifestations and thus should be reclassified and managed as CHH.


Subject(s)
Agranulocytosis/etiology , Immunologic Deficiency Syndromes/etiology , Mutation , Neoplasms/etiology , Osteochondrodysplasias/pathology , RNA, Long Noncoding/genetics , Adult , Aged , Female , Hair/abnormalities , Hirschsprung Disease , Humans , Middle Aged , Osteochondrodysplasias/complications , Osteochondrodysplasias/congenital , Osteochondrodysplasias/genetics , Osteochondrodysplasias/metabolism , Primary Immunodeficiency Diseases , Young Adult
12.
Pediatr Hematol Oncol ; 38(3): 227-238, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33205673

ABSTRACT

Bone marrow minimal residual disease (MRD) is the strongest predictor of relapse in children with acute lymphoblastic leukemia (ALL). 6-mercaptopurine (6MP) in ALL therapy has wide inter-individual variation in disposition and is strongly influenced by polymorphisms in the thiopurine methyltransferase (TPMT) gene. In 952 patients treated according to the NOPHO ALL2008 protocol, we explored the association between thiopurine disposition, TPMT genotypes and MRD levels after consolidation therapy with 6MP, high-dose methotrexate (HD-MTX), asparaginase, and vincristine. The levels of the cytotoxic DNA-incorporated thioguanine were significantly higher on day 70-79 in G460A/A719G TPMT heterozygous (TPMTHZ) compared to TPMT wild type (TPMTWT) patients (mean: 230.7 vs. 149.7 fmol/µg DNA, p = 0.002). In contrast, TPMT genotype did not associate with the end of consolidation MRD levels irrespective of randomization of the patients to fixed dose (25 mg/m2/day) or 6MP escalation (up to 50 or 75 mg/m2/day) during consolidation therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Mercaptopurine/therapeutic use , Methyltransferases/genetics , Polymorphism, Genetic , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Asparaginase/therapeutic use , Child , Consolidation Chemotherapy , Female , Humans , Male , Methotrexate/therapeutic use , Neoplasm, Residual , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Vincristine/therapeutic use
13.
Int J Cancer ; 146(10): 2746-2755, 2020 05 15.
Article in English | MEDLINE | ID: mdl-31407318

ABSTRACT

Immigration in Europe has increased considerably over the past decades with the immigrant population similarly expanding in Finland. Our aim was to study childhood cancer mortality and survival in immigrants. In all, 4,437 patients diagnosed with cancer under the age of 20 years between 1990 and 2009 were identified from the Finnish Cancer Registry and their parents from the Population Register Center. Information on demographic factors was obtained from Statistics Finland. Poisson regression modeling was used to estimate hazard ratios (HRs) for cancer deaths. The life table method and the log rank test were used in survival analysis. Patients or parents of foreign background and born abroad had higher 5-year mortality (patient HR 2.03, 95% CI 1.18-3.49; maternal HR 2.11, 95% CI 1.46-3.04; paternal HR 1.85, 95% CI 1.29-2.66) compared to those of Finnish background and born in Finland. Childhood cancer survival in 5-year follow-up was higher if the mother (83% vs. 68%) or the father (83% vs. 70%) were of Finnish background and born in Finland. Despite equal access to public health care, we observed significant differences in childhood cancer mortality and survival by background. Cultural differences, linguistic obstacles and difficulties in navigating the health care system may contribute, along with genetic and biologic factors. Offering tailored information and taking cultural and linguistic aspects into account is necessary when diagnosing and treating patients from different ethnic backgrounds who have not yet integrated into the local culture and health care system.


Subject(s)
Emigrants and Immigrants/statistics & numerical data , Neoplasms/ethnology , Neoplasms/mortality , Adolescent , Cancer Survivors , Child , Child, Preschool , Female , Finland/epidemiology , Humans , Incidence , Infant , Infant, Newborn , Male , Registries , Young Adult
14.
Scand J Immunol ; 92(4): e12913, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32506568

ABSTRACT

Cartilage-hair hypoplasia (CHH) is an autosomal recessive syndromic immunodeficiency with skeletal dysplasia, short stature, hypotrichosis, variable degree of immune dysfunction and increased incidence of anaemia, Hirschsprung disease and malignancy. CHH is caused by variants in the RMRP gene, encoding the untranslated RNA molecule of the mitochondrial RNA-processing endoribonuclease, which participates in for example cell cycle regulation and telomere maintenance. Recent studies have expanded our understanding of the complex pathogenesis of CHH. Immune dysfunction has a major impact on clinical course and prognosis. Clinical features of immune dysfunction are highly variable, progressive and include infections, lung disease, immune dysregulation and malignancy. Mortality is increased compared with the general population, due to infections, malignancy and pulmonary disease. Several risk factors for early mortality have been reported in the Finnish CHH cohort and can be used to guide management. Newborn screening for severe combined immunodeficiency can possibly be of prognostic value in CHH. Regular follow-up by a multidisciplinary team should be implemented to address immune dysfunction in all patients with CHH, also in asymptomatic cases. Haematopoietic stem cell transplantation can cure immune dysfunction, but its benefits in mildly symptomatic patients with CHH remain debatable. Further research is needed to understand the mechanisms behind the variability of clinical features, to search for potential molecular treatment targets, to examine and validate risk factors for early mortality outside the Finnish CHH cohort and to develop management guidelines. This review focuses on the pathogenesis, clinical course and management of CHH.


Subject(s)
Hair/abnormalities , Hirschsprung Disease/immunology , Osteochondrodysplasias/congenital , Primary Immunodeficiency Diseases/immunology , Animals , Hair/immunology , Hair/pathology , Hair/physiopathology , Hirschsprung Disease/pathology , Hirschsprung Disease/physiopathology , Humans , Osteochondrodysplasias/immunology , Osteochondrodysplasias/pathology , Osteochondrodysplasias/physiopathology , Primary Immunodeficiency Diseases/pathology , Primary Immunodeficiency Diseases/physiopathology
15.
Pediatr Blood Cancer ; 67(2): e27999, 2020 02.
Article in English | MEDLINE | ID: mdl-31674724

ABSTRACT

BACKGROUND: Children with central nervous system (CNS) toxicity during therapy for acute lymphoblastic leukaemia (ALL) are at risk for treatment modifications, long-term sequelae and even higher mortality. A better understanding of CNS symptoms and their complications improves the potential to prevent and treat them. METHODS: Patient files from 649 children treated with Nordic Society of Pediatric Hematology and Oncology ALL92 and ALL2000 protocols in Finland were reviewed retrospectively for any acute CNS symptom. Detailed data on symptoms, examinations and treatment of the underlying CNS complications were collected from the medical records. Disease-related and outcome data were retrieved from the Nordic leukaemia registry. RESULTS: Altogether, 13% (86) of patients with ALL had acute CNS symptoms. Most symptoms (64%) occurred during the first 2 months of therapy. Posterior reversible encephalopathy syndrome was the most frequent complication (4.5%). Cerebrovascular events were diagnosed in 10 cases (1.6%), while methotrexate-related stroke-like syndrome (SLS) was observed in only one patient (0.2%). CNS symptoms due to systemic or unclear conditions, especially sepsis, were important for differential diagnosis. CNS leukaemia was associated with CNS symptoms (hazard ratio [HR] = 4.03; P = .003), and epilepsy was a common sequel of CNS complications (19%). CONCLUSIONS: Acute CNS symptoms are common during ALL therapy, occurring mainly during the first 2 months of treatment. Patients with CNS leukaemia at diagnosis are at a higher risk for CNS toxicity. Despite intensive CNS-directed methotrexate treatment, SLS was diagnosed extremely rarely in our series.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Central Nervous System Diseases/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Central Nervous System Diseases/chemically induced , Child , Female , Follow-Up Studies , Humans , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Prognosis , Retrospective Studies , Survival Rate
16.
Pediatr Transplant ; 24(8): e13846, 2020 12.
Article in English | MEDLINE | ID: mdl-33111465

ABSTRACT

Red blood cell transfusions are an essential part of supporting care in leukemia treatment. We examined the prevalence of iron overload and its effects on organ function and childhood growth in pediatric patients after allogeneic HSCT for acute lymphoblastic leukemia. Twenty-three patients were included (median age 12.6, range 7.5-21.4 years). Body iron load was determined using laboratory tests, hepatic and cardiac MRI, and by calculating iron received from transfusions. We performed multivariate analysis to determine association of body iron load with liver enzymes, cardiac function, insulin resistance, and growth. Median plasma ferritin was 344 (range 40-3235) ng/mL and exceeded 1000 ng/mL in three patients (13%). In MRI, 11 patients (48%) had hepatic iron overload and 1 patient (4%) myocardial iron overload. In cardiac MRI, 8 patients (35%) had significant but subclinical decrease in ejection fraction (median z-score -1.7, range -3.1-0.14), but cardiac function did not associate with iron status. Alanine transaminase associated with transfused iron per time unit (P = .001) after the median follow-up of 4.5 years. No correlation was found between iron load and growth or insulin resistance. Iron overload is common in children transplanted for ALL, but iron overload associated organ dysfunction is not present at early age. We recommend evaluation of iron load for all patients at least once during follow-up after transplantation.


Subject(s)
Hematopoietic Stem Cell Transplantation/adverse effects , Iron Overload/etiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Child , Female , Finland , Humans , Male
17.
Eur J Pediatr ; 179(9): 1453-1459, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32193656

ABSTRACT

Prednisolone used in the induction phase of the treatment of acute lymphoblastic leukemia (ALL) may suppress hypothalamic-pituitary-adrenal axis and require hydrocortisone substitution. In this retrospective analysis, we reviewed altogether 371 ACTH stimulation tests of 352 children after a uniform NOPHO (Nordic Society of Pediatric Hematology and Oncology) ALL induction. Both low- and standard-dose ACTH tests were used. Full recovery of adrenal function was defined by both normal basal and stimulated cortisol levels. Sixty-two percent of patients were detected with normal adrenal function in median of 15 days after tapering of prednisolone. Both low basal and stimulated cortisol levels were detected in 13% of patients. The median time to normal adrenal function was 31 days (95% CI 28-34), 24 days (95% CI 18-30), and 12 days (95% CI 10-14) for those with basal cortisol <107, 107-183, and >183 nmol/L at first ACTH testing, respectively. Patients with fluconazole prophylaxis had higher median baseline cortisol levels compared to patients without prophylaxis (207 nmol/L, range 21-839 nmol/L vs. 153 nmol/L, range 22-832 nmol/L, P = 0.003).Conclusion: These data can be used to reduce unnecessary substitution or testing, but also to guarantee hydrocortisone substitution for those at risk. What is Known: •These data can be used to reduce unnecessary hydrocortisone substitution or ACTH testing. •Our data helps to guarantee hydrocortisone substitution for those at risk of adrenal insufficiency. What is New: •Full recovery of adrenal function after ALL induction is detected in 62% of patients already at 15 days after tapering of prednisolone. •Both basal and stimulated cortisol testing are required for detection of full adrenal recovery. •Recovery time of adrenal function is extended over 3-4 weeks after tapering of prednisolone in patients with low basal cortisol levels (<107 nmol/L) at first testing.


Subject(s)
Hypothalamo-Hypophyseal System , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Child , Humans , Induction Chemotherapy , Pituitary-Adrenal System , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Retrospective Studies
18.
Acta Obstet Gynecol Scand ; 99(1): 105-111, 2020 01.
Article in English | MEDLINE | ID: mdl-31449329

ABSTRACT

INTRODUCTION: The purpose of this study was to investigate the epidemiology and characteristics of surgically treated ovarian lesions in preadolescent girls. MATERIAL AND METHODS: This was a retrospective cohort study including all 0- to 11-year-old girls operated at a single center from 1999 to 2016 for ovarian cysts, neoplasms or torsions. Patient charts were reviewed for symptoms, preoperative radiological imaging, operative details and histopathology. RESULTS: We identified 78 girls, resulting in a population-based incidence of 4.2/100 000. Infants (n = 44) presented with benign cysts (42/44, 95%, one bilateral), a benign neoplasm (1/44, 2%) and a torsion without other pathology (1/44, 2%). Torsion was found in 25/29 (86%) ovaries with complex cysts and in 3/15 (21%) ovaries with simple cysts in preoperative imaging (P < 0.001). Most infants were symptomless. Lesions in 1- to 11-year-old girls (n = 34) included benign neoplasms (n = 21/34, 62%), malignant neoplasms (n = 5/34, 15%), a cyst with torsion (n = 1/34, 3%) and torsions without other pathology (n = 7/34, 21%). Torsion was more common in benign (17/21, 81%) than in malignant neoplasms (1/5, 20%) (P < 0.020). Ovarian diameter did not differ between ovaries with or without torsion (P = 0.238) or between benign and malignant neoplasms (P = 0.293). The duration of symptoms in lesions with or without torsion was similar. CONCLUSIONS: The majority of surgically treated ovarian lesions in preadolescent are benign lesions with torsion. Surgery should be ovary-preserving and performed without delay.


Subject(s)
Ovarian Diseases/surgery , Torsion Abnormality/surgery , Child , Child, Preschool , Female , Finland/epidemiology , Humans , Incidence , Infant , Infant, Newborn , Ovarian Cysts/epidemiology , Ovarian Cysts/surgery , Ovarian Diseases/epidemiology , Ovarian Neoplasms/epidemiology , Ovarian Neoplasms/surgery , Retrospective Studies , Torsion Abnormality/epidemiology
19.
Pediatr Hematol Oncol ; 37(5): 355-364, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32166994

ABSTRACT

Treosulfan-based regimens constitute a feasible and increasingly used, but still myeloablative, conditioning in pediatric allogeneic hematopoietic stem cell transplantation (HSCT). We retrospectively analyzed the acute toxicity and outcome of all consecutive (2004-2015) pediatric HSCT patients prepared for HSCT with treosulfan in a single-center setting. We included HSCTs performed for both nonmalignant (n = 23) and malignant diseases (n = 11). The controls were patients with nonmalignant diseases or hematological malignancies conditioned with cyclophosphamide (Cy)-total body irradiation (TBI)-based (39 patients) or busulfan-based regimens (11 patients). The major toxicities of the treosulfan-based regimens were limited to oral mucosa and skin. 50% of the patients needed IV morphine for severe mucositis compared to 31% in patients conditioned with Cy-TBI (P = 0.02). Other toxicities were rare. The disease-free survival (DFS) of patients transplanted for nonmalignant disorders was 88.9 ± 7.5% at 2 years. The event-free survival (EFS) at 2 years in this small cohort for those with a malignant disease and a treosulfan-based conditioning was 54.5 ± 1.5%. We conclude that a treosulfan-based conditioning regimen gives excellent DFS in pediatric HSCT performed for a nonmalignant disorder but with substantial mucosal toxicity. In a malignant disorder a treosulfan-based regimen looks promising but larger, preferably randomized, studies are needed to prove efficacy.


Subject(s)
Antineoplastic Agents/adverse effects , Busulfan/analogs & derivatives , Hematologic Neoplasms/therapy , Hematopoietic Stem Cell Transplantation/methods , Transplantation Conditioning/adverse effects , Adolescent , Antineoplastic Agents/therapeutic use , Busulfan/adverse effects , Busulfan/therapeutic use , Child , Child, Preschool , Cyclophosphamide/therapeutic use , Disease-Free Survival , Female , Humans , Infant , Male , Myeloablative Agonists/adverse effects , Myeloablative Agonists/therapeutic use , Retrospective Studies , Transplantation Conditioning/methods , Transplantation, Homologous , Treatment Outcome , Whole-Body Irradiation/methods
20.
Cancer ; 125(20): 3631-3638, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31287570

ABSTRACT

BACKGROUND: Childhood cancer survivors have been reported to be vulnerable to psychiatric morbidities and risky health behavior. Suicides, substance abuse, accidents, and violence as causes of death can be regarded as an extreme manifestation of risky health behavior. In the current study, the authors studied the risk of suicide and other risky health behavior-related deaths among childhood cancer patients in Denmark, Finland, and Sweden. METHODS: Using linkage between national cancer, population, and cause-of-death registries, the authors investigated the causes of death in 29,285 patients diagnosed with cancer before age 20 years between 1971 and 2009 compared with a cohort of 146,282 age-matched, sex-matched, and country-matched population comparisons. Rate ratios (RRs) with 95% CIs were estimated using Poisson regression models, adjusting for demographic factors. RESULTS: The overall risk of dying of a risky health behavior was found to be increased among childhood cancer patients (RR, 1.25; 95% CI, 1.06-1.47) when compared with population comparisons. The elevated risk was statistically significant among patients with central nervous system tumors (RR, 1.49; 95% CI, 1.08-2.05) and patients diagnosed at ages 5 to 9 years and 15 to 19 years (RR, 1.50 [95% CI, 1.01-2.24] and RR, 1.31 [95% CI, 1.03-1.67], respectively). The overall risk of suicide was found to be increased (RR, 1.37; 95% CI, 1.02-1.83), and statistically significantly so when patients were diagnosed between ages 15 and 19 years (RR, 1.61; 95% CI, 1.09-2.39). CONCLUSIONS: Childhood cancer patients appear to have an increased risk of risky health behavior-related causes of death compared with the general population. The results of the current study suggest the importance of integrating psychosocial support into the follow-up care of these individuals.


Subject(s)
Cancer Survivors/psychology , Health Risk Behaviors , Neoplasms/mortality , Suicide/psychology , Adolescent , Adult , Child , Child, Preschool , Death , Female , Hospitalization , Humans , Male , Neoplasms/pathology , Neoplasms/psychology , Risk Assessment , Risk Factors , Young Adult , Suicide Prevention
SELECTION OF CITATIONS
SEARCH DETAIL