Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Cancers (Basel) ; 16(3)2024 Jan 24.
Article in English | MEDLINE | ID: mdl-38339248

ABSTRACT

Improvements in survival have been made over the past two decades for childhood acute myeloid leukemia (AML), but the approximately 40% of patients who relapse continue to have poor outcomes. A combination of checkpoint-inhibitor nivolumab and azacitidine has demonstrated improvements in median survival in adults with AML. This phase I/II study with nivolumab and azacitidine in children with relapsed/refractory AML (NCT03825367) was conducted through the Therapeutic Advances in Childhood Leukemia & Lymphoma consortium. Thirteen patients, median age 13.7 years, were enrolled. Patients had refractory disease with multiple reinduction attempts. Twelve evaluable patients were treated at the recommended phase II dose (established at dose level 1, 3 mg/kg/dose). Four patients (33%) maintained stable disease. This combination was well tolerated, with no dose-limiting toxicities observed. Grade 3-4 adverse events (AEs) were primarily hematological. Febrile neutropenia was the most common AE ≥ grade 3. A trend to improved quality of life was noted. Increases in CD8+ T cells and reductions in CD4+/CD8+ T cells and demethylation were observed. The combination was well tolerated and had an acceptable safety profile in pediatric patients with relapsed/refractory AML. Future studies might explore this combination for the maintenance of remission in children with AML at high risk of relapse.

2.
Pediatr Blood Cancer ; 69(10): e29812, 2022 10.
Article in English | MEDLINE | ID: mdl-35726868

ABSTRACT

Current therapies for relapsed/refractory (R/R) pediatric myeloid neoplasms are inadequately effective. Real-world data (RWD) can improve care by augmenting traditional studies and include individuals not eligible for clinical trials. The Therapeutic Advances in Childhood Leukemia and Lymphoma (TACL) consortium recently completed T2016-003, a phase 1 study of decitabine, vorinostat, fludarabine, cytarabine, and granulocyte colony-stimulating factor (G-CSF) in R/R acute myeloid leukemia (AML), which added epigenetic drugs to a cytotoxic backbone. We report results of RWD from six centers that treated 28 pediatric patients (26 with AML, two with other myeloid neoplasms) identically to the TACL study but who were not enrolled. This allowed unique analyses and the ability to compare data with the 35 TACL study patients. The overall response rate (ORR) (complete response [CR] plus CR with incomplete count recovery) among 26 RWD evaluable patients was 65%. The ORR of 13 patients with relapsed AML with epigenetic alterations was 69% (T2016-003 + RWD: 68%, n = 25), of eight patients with refractory AML was 38% (T2016-003 + RWD: 41%, n = 17) and of five patients with therapy-related AML (t-AML) was 80% (T2016-003 + RWD: 75%, n = 8). The mean number of Grade 3/4 toxicities experienced by the T2016-003-eligible RWD population (n = 22) (one per patient-cycle) was not meaningfully different than those (n = 6) who would have been TACL study-ineligible secondary to comorbidities (two per patient-cycle). Overall, this therapy was well tolerated and effective in pediatric patients with R/R myeloid neoplasms, particularly those with epigenetic alterations, t-AML, and refractory disease.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Leukemia, Myeloid, Acute , Neoplasm Recurrence, Local , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Child , Cytarabine , Decitabine/therapeutic use , Granulocyte Colony-Stimulating Factor , Humans , Leukemia, Myeloid, Acute/drug therapy , Neoplasm Recurrence, Local/drug therapy , Vidarabine , Vorinostat/therapeutic use
3.
Am J Hematol ; 97(5): 613-622, 2022 05.
Article in English | MEDLINE | ID: mdl-35180323

ABSTRACT

Survival outcomes for relapsed/refractory pediatric acute myeloid leukemia (R/R AML) remain dismal. Epigenetic changes can result in gene expression alterations which are thought to contribute to both leukemogenesis and chemotherapy resistance. We report results from a phase I trial with a dose expansion cohort investigating decitabine and vorinostat in combination with fludarabine, cytarabine, and G-CSF (FLAG) in pediatric patients with R/R AML [NCT02412475]. Thirty-seven patients enrolled with a median age at enrollment of 8.4 (range, 1-20) years. There were no dose limiting toxicities among the enrolled patients, including two patients with Down syndrome. The recommended phase 2 dose of decitabine in combination with vorinostat and FLAG was 10 mg/m2 . The expanded cohort design allowed for an efficacy evaluation and the overall response rate among 35 evaluable patients was 54% (16 complete response (CR) and 3 complete response with incomplete hematologic recovery (CRi)). Ninety percent of responders achieved minimal residual disease (MRD) negativity (<0.1%) by centralized flow cytometry and 84% (n = 16) successfully proceeded to hematopoietic stem cell transplant. Two-year overall survival was 75.6% [95%CI: 47.3%, 90.1%] for MRD-negative patients vs. 17.9% [95%CI: 4.4%, 38.8%] for those with residual disease (p < .001). Twelve subjects (34%) had known epigenetic alterations with 8 (67%) achieving a CR, 7 (88%) of whom were MRD negative. Correlative pharmacodynamics demonstrated the biologic activity of decitabine and vorinostat and identified specific gene enrichment signatures in nonresponding patients. Overall, this therapy was well-tolerated, biologically active, and effective in pediatric patients with R/R AML, particularly those with epigenetic alterations.


Subject(s)
Leukemia, Myeloid, Acute , Lymphoma , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Child , Cytarabine , Decitabine/therapeutic use , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Lymphoma/drug therapy , Vorinostat
4.
Lancet Haematol ; 9(1): e49-e57, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34971582

ABSTRACT

BACKGROUND: Adolescents with Hodgkin lymphoma have worse disease outcomes than children. Whether these differences persist within clinical trials is unknown. We examined survival, by age, in patients receiving response-adapted therapy for Hodgkin lymphoma on Children's Oncology Group (COG) trials. METHODS: Patients (aged 1-21 years) diagnosed with classical Hodgkin lymphoma and enrolled between Sept 23, 2002, and Jan 19, 2012, on one of three phase 3 COG trials in the USA and Canada were eligible for inclusion. The three COG trials were defined by risk group according to Ann Arbor stage, B-symptoms, and bulk (AHOD0431 [low risk; NCT00302003], AHOD0031 [intermediate risk; NCT00025259], or AHOD0831 [high risk; NCT01026220]). The outcomes of this study were event-free survival (death, relapse, or subsequent neoplasm) and overall survival. Cox proportional hazards models estimated survival, adjusting for disease and treatment factors both overall and in patients with mixed cellularity or non-mixed cellularity (nodular sclerosing and not-otherwise-specified) disease. FINDINGS: Of 2155 patients enrolled on the three trials, 1907 (88·4%; 968 [50·8%] male and 939 [49·2%] female; 1227 [64·3%] non-Hispanic White) were included in this analysis. After a median follow-up of 7·4 years (IQR 4·3-10·2), older patients (aged ≥15 years) had worse unadjusted 5-year event-free survival (80% [95% CI 78-83]) than did younger patients (aged <15 years; 86% [83-88]; HR 1·38 [1·11-1·71]; p=0·0038). Older patients also had worse unadjusted 5-year overall survival than did younger patients (96% [95% CI 95-97] vs 99% [98-99]; HR 2·50 [1·41-4·45]; p=0·0012). In patients with non-mixed cellularity histology, older patients had a significantly increased risk of having an event than did younger patients with the same histology (HR 1·32 [1·03-1·68]; p=0·027). Older patients with mixed cellularity had significantly worse 5-year event-free survival than did younger patients in unadjusted (77% [95% CI 65-86] for older patients vs 94% [88-97] for younger patients; HR 2·93 [1·37-6·29]; p=0·0039) and multivariable models (HR 3·72 [1·56-8·91]; p=0·0032). Overall, older patients were more likely to die than younger patients (HR 3·08 [1·49-6·39]; p=0·0025). INTERPRETATION: Adolescents (≥15 years) treated on COG Hodgkin lymphoma trials had worse event-free survival and increased risk of death compared with children (<15 years). Our findings highlight the need for prospective studies to examine tumour and host biology, and to test novel therapies across the age spectrum. FUNDING: National Institutes of Health, St Baldrick's Foundation, and Lymphoma Research Foundation.


Subject(s)
Hodgkin Disease , Adolescent , Antineoplastic Combined Chemotherapy Protocols , Child , Child, Preschool , Female , Hodgkin Disease/drug therapy , Humans , Infant , Male , Neoplasm Recurrence, Local , Progression-Free Survival , Prospective Studies , Retrospective Studies , Young Adult
5.
J Cardiothorac Vasc Anesth ; 35(3): 689-690, 2021 03.
Article in English | MEDLINE | ID: mdl-33436282
6.
J Pediatr Hematol Oncol ; 43(1): 24-27, 2021 01.
Article in English | MEDLINE | ID: mdl-31725543

ABSTRACT

A toddler undergoing treatment for refractory Langerhans cell histiocytosis (LCH) developed concurrent hemophagocytic lymphohistiocytosis (HLH). These are thought to be distinct histiocytic disorders, with different pathophysiologies, diagnostic criteria, and treatments. HLH in a patient with LCH is thought to be quite rare. In this report, we review the presentation of our patient, as well as review the existing literature of other pediatric patients who have been diagnosed with both LCH and HLH.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Histiocytosis, Langerhans-Cell/pathology , Lymphohistiocytosis, Hemophagocytic/pathology , Histiocytosis, Langerhans-Cell/complications , Histiocytosis, Langerhans-Cell/drug therapy , Humans , Infant , Lymphohistiocytosis, Hemophagocytic/complications , Lymphohistiocytosis, Hemophagocytic/drug therapy , Male , Prognosis
7.
Clin Cancer Res ; 26(10): 2297-2307, 2020 05 15.
Article in English | MEDLINE | ID: mdl-31969338

ABSTRACT

PURPOSE: Treatment failure from drug resistance is the primary reason for relapse in acute lymphoblastic leukemia (ALL). Improving outcomes by targeting mechanisms of drug resistance is a potential solution. PATIENTS AND METHODS: We report results investigating the epigenetic modulators decitabine and vorinostat with vincristine, dexamethasone, mitoxantrone, and PEG-asparaginase for pediatric patients with relapsed or refractory B-cell ALL (B-ALL). Twenty-three patients, median age 12 years (range, 1-21) were treated in this trial. RESULTS: The most common grade 3-4 toxicities included hypokalemia (65%), anemia (78%), febrile neutropenia (57%), hypophosphatemia (43%), leukopenia (61%), hyperbilirubinemia (39%), thrombocytopenia (87%), neutropenia (91%), and hypocalcemia (39%). Three subjects experienced dose-limiting toxicities, which included cholestasis, steatosis, and hyperbilirubinemia (n = 1); seizure, somnolence, and delirium (n = 1); and pneumonitis, hypoxia, and hyperbilirubinemia (n = 1). Infectious complications were common with 17 of 23 (74%) subjects experiencing grade ≥3 infections including invasive fungal infections in 35% (8/23). Nine subjects (39%) achieved a complete response (CR + CR without platelet recovery + CR without neutrophil recovery) and five had stable disease (22%). Nine (39%) subjects were not evaluable for response, primarily due to treatment-related toxicities. Correlative pharmacodynamics demonstrated potent in vivo modulation of epigenetic marks, and modulation of biologic pathways associated with functional antileukemic effects. CONCLUSIONS: Despite encouraging response rates and pharmacodynamics, the combination of decitabine and vorinostat on this intensive chemotherapy backbone was determined not feasible in B-ALL due to the high incidence of significant infectious toxicities. This study is registered at http://www.clinicaltrials.gov as NCT01483690.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasm Recurrence, Local/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adolescent , Adult , Asparaginase/administration & dosage , Bortezomib/administration & dosage , Child , Child, Preschool , Decitabine/administration & dosage , Dexamethasone/administration & dosage , Doxorubicin/administration & dosage , Female , Follow-Up Studies , Humans , Infant , Male , Mitoxantrone/administration & dosage , Neoplasm Recurrence, Local/pathology , Pilot Projects , Polyethylene Glycols/administration & dosage , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Prognosis , Salvage Therapy/methods , Survival Rate , Vincristine/administration & dosage , Vorinostat/administration & dosage , Young Adult
9.
J Cardiothorac Vasc Anesth ; 33(1): 1, 2019 01.
Article in English | MEDLINE | ID: mdl-30591177

ABSTRACT

This article has been removed at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.

10.
Haematologica ; 103(12): 2008-2015, 2018 12.
Article in English | MEDLINE | ID: mdl-30026338

ABSTRACT

YARS2 variants have previously been described in patients with myopathy, lactic acidosis and sideroblastic anemia 2 (MLASA2). YARS2 encodes the mitochondrial tyrosyl-tRNA synthetase, which is responsible for conjugating tyrosine to its cognate mt-tRNA for mitochondrial protein synthesis. Here we describe 14 individuals from 11 families presenting with sideroblastic anemia and YARS2 variants that we identified using a sideroblastic anemia gene panel or exome sequencing. The phenotype of these patients ranged from MLASA to isolated congenital sideroblastic anemia. As in previous cases, inter- and intra-familial phenotypic variability was observed, however, this report includes the first cases with isolated sideroblastic anemia and patients with biallelic YARS2 variants that have no clinically ascertainable phenotype. We identified ten novel YARS2 variants and three previously reported variants. In vitro amino-acylation assays of five novel missense variants showed that three had less effect on the catalytic activity of YARS2 than the most commonly reported variant, p.(Phe52Leu), associated with MLASA2, which may explain the milder phenotypes in patients with these variants. However, the other two missense variants had a more severe effect on YARS2 catalytic efficiency. Several patients carried the common YARS2 c.572 G>T, p.(Gly191Val) variant (minor allele frequency =0.1259) in trans with a rare deleterious YARS2 variant. We have previously shown that the p.(Gly191Val) variant reduces YARS2 catalytic activity. Consequently, we suggest that biallelic YARS2 variants, including severe loss-of-function alleles in trans of the common p.(Gly191Val) variant, should be considered as a cause of isolated congenital sideroblastic anemia, as well as the MLASA syndromic phenotype.


Subject(s)
Acidosis, Lactic/genetics , Anemia, Sideroblastic/genetics , Genetic Diseases, X-Linked/genetics , Germ-Line Mutation , MELAS Syndrome/genetics , Mitochondrial Proteins/genetics , Tyrosine-tRNA Ligase/genetics , Acidosis, Lactic/enzymology , Adolescent , Anemia, Sideroblastic/enzymology , Female , Genetic Association Studies , Genetic Diseases, X-Linked/enzymology , Humans , Infant , MELAS Syndrome/enzymology , Male , Middle Aged , Mutation, Missense , Young Adult
12.
Pediatr Blood Cancer ; 63(1): 39-46, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26235333

ABSTRACT

BACKGROUND: The primary aim of this Phase I study was to determine the maximum tolerated dose (MTD) of TPI 287 and the safety and tolerability of TPI 287 alone and in combination with temozolomide (TMZ) in pediatric patients with refractory or recurrent neuroblastoma or medulloblastoma. The secondary aims were to evaluate the pharmacokinetics of TPI 287 and the treatment responses. PROCEDURE: Eighteen patients were enrolled to a phase I dose escalation trial of weekly intravenous infusion of TPI 287 for two 28-day cycles with toxicity monitoring to determine the MTD, followed by two cycles of TPI 287 in combination with TMZ. Samples were collected to determine the pharmacokinetic parameters C(max), AUC(0-24), t(1/2), CL, and Vd on day 1 of cycles 1 (TPI 287 alone) and 3 (TPI 287 + TMZ) following TPI 287 infusion. Treatment response was evaluated by radiographic (CT or MRI) and radionuclide (MIBG) imaging for neuroblastoma. RESULTS: We determined the MTD of TPI 287 alone and in combination with temozolomide to be 125 mg/m(2). The non-dose-limiting toxicities at this dose were mainly anorexia and pain. The dose-limiting toxicities (DLTs) of two patients at 135 mg/m(2) were grade 3 hemorrhagic cystitis and grade 3 sensory neuropathy. CONCLUSIONS: Overall, TPI 287 was well tolerated by pediatric patients with refractory and relapsed neuroblastoma and medulloblastoma at a dose of 125 mg/m(2) IV on days 1, 8, and 15 of a 28 day cycle.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Dacarbazine/analogs & derivatives , Medulloblastoma/drug therapy , Neuroblastoma/drug therapy , Taxoids/administration & dosage , Taxoids/therapeutic use , Adolescent , Adult , Child , Child, Preschool , Dacarbazine/administration & dosage , Dacarbazine/pharmacokinetics , Dacarbazine/toxicity , Female , Humans , Infusions, Intravenous , Male , Maximum Tolerated Dose , Neoplasm Recurrence, Local , Taxoids/pharmacokinetics , Taxoids/toxicity , Temozolomide
13.
PLoS One ; 10(5): e0127246, 2015.
Article in English | MEDLINE | ID: mdl-26018967

ABSTRACT

BACKGROUND: Neuroblastoma (NB) is the most common cancer in infancy and most frequent cause of death from extracranial solid tumors in children. Ornithine decarboxylase (ODC) expression is an independent indicator of poor prognosis in NB patients. This study investigated safety, response, pharmacokinetics, genetic and metabolic factors associated with ODC in a clinical trial of the ODC inhibitor difluoromethylornithine (DFMO) ± etoposide for patients with relapsed or refractory NB. METHODS AND FINDINGS: Twenty-one patients participated in a phase I study of daily oral DFMO alone for three weeks, followed by additional three-week cycles of DFMO plus daily oral etoposide. No dose limiting toxicities (DLTs) were identified in patients taking doses of DFMO between 500-1500 mg/m2 orally twice a day. DFMO pharmacokinetics, single nucleotide polymorphisms (SNPs) in the ODC gene and urinary levels of substrates for the tissue polyamine exporter were measured. Urinary polyamine levels varied among patients at baseline. Patients with the minor T-allele at rs2302616 of the ODC gene had higher baseline levels (p=0.02) of, and larger decreases in, total urinary polyamines during the first cycle of DFMO therapy (p=0.003) and had median progression free survival (PFS) that was over three times longer, compared to patients with the major G allele at this locus although this last result was not statistically significant (p=0.07). Six of 18 evaluable patients were progression free during the trial period with three patients continuing progression free at 663, 1559 and 1573 days after initiating treatment. Median progression-free survival was less among patients having increased urinary polyamines, especially diacetylspermine, although this result was not statistically significant (p=0.056). CONCLUSIONS: DFMO doses of 500-1500 mg/m2/day are safe and well tolerated in children with relapsed NB. Children with the minor T allele at rs2302616 of the ODC gene with relapsed or refractory NB had higher levels of urinary polyamine markers and responded better to therapy containing DFMO, compared to those with the major G allele at this locus. These findings suggest that this patient subset may display dependence on polyamines and be uniquely susceptible to therapies targeting this pathway. TRIAL REGISTRATION: Clinicaltrials.gov NCT#01059071.


Subject(s)
Eflornithine/pharmacology , Neuroblastoma/drug therapy , Ornithine Decarboxylase Inhibitors/pharmacology , Phenotype , Polyamines/metabolism , Adolescent , Child , Child, Preschool , Eflornithine/adverse effects , Eflornithine/pharmacokinetics , Eflornithine/therapeutic use , Etoposide/adverse effects , Etoposide/pharmacology , Etoposide/therapeutic use , Female , Humans , Infant , Male , Neuroblastoma/enzymology , Neuroblastoma/genetics , Neuroblastoma/urine , Ornithine Decarboxylase/metabolism , Ornithine Decarboxylase Inhibitors/adverse effects , Ornithine Decarboxylase Inhibitors/pharmacokinetics , Ornithine Decarboxylase Inhibitors/therapeutic use , Polyamines/urine , Recurrence , Safety , Treatment Outcome
15.
Cancer Med ; 4(6): 871-86, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25720842

ABSTRACT

The primary objective of the study was to evaluate the feasibility and safety of a process which would utilize genome-wide expression data from tumor biopsies to support individualized treatment decisions. Current treatment options for recurrent neuroblastoma are limited and ineffective, with a survival rate of <10%. Molecular profiling may provide data which will enable the practitioner to select the most appropriate therapeutic option for individual patients, thus improving outcomes. Sixteen patients with neuroblastoma were enrolled of which fourteen were eligible for this study. Feasibility was defined as completion of tumor biopsy, pathological evaluation, RNA quality control, gene expression profiling, bioinformatics analysis, generation of a drug prediction report, molecular tumor board yielding a treatment plan, independent medical monitor review, and treatment initiation within a 21 day period. All eligible biopsies passed histopathology and RNA quality control. Expression profiling by microarray and RNA sequencing were mutually validated. The average time from biopsy to report generation was 5.9 days and from biopsy to initiation of treatment was 12.4 days. No serious adverse events were observed and all adverse events were expected. Clinical benefit was seen in 64% of patients as stabilization of disease for at least one cycle of therapy or partial response. The overall response rate was 7% and the progression free survival was 59 days. This study demonstrates the feasibility and safety of performing real-time genomic profiling to guide treatment decision making for pediatric neuroblastoma patients.


Subject(s)
Molecular Targeted Therapy/methods , Neoplasm Recurrence, Local/therapy , Neuroblastoma/therapy , Adolescent , Antineoplastic Agents/therapeutic use , Child , Child, Preschool , Chronic Disease , Feasibility Studies , Female , Gene Expression Profiling/methods , Genome-Wide Association Study/methods , Humans , Male , Molecular Targeted Therapy/adverse effects , Patient Safety , Prospective Studies , RNA, Neoplasm/genetics , Sequence Analysis, RNA/methods , Time-to-Treatment , Treatment Outcome , Young Adult
16.
Pediatr Dent ; 34(1): 51-3, 2012.
Article in English | MEDLINE | ID: mdl-22353458

ABSTRACT

In patients with deficient immune defenses, a localized oral infection can progress to a systemic infection. The purpose of this paper was to describe the case of a child with acute lymphoblastic leukemia who presented with fever, trismus, and submandibular swelling in the absence of the typical dental causes of infection such as deep dental caries or clinically significant periodontal disease. Treatment included an aggressive intravenous antibiotic regimen and extraction of the offending tooth after the recovery from bone marrow suppression.


Subject(s)
Focal Infection, Dental/etiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Tooth Exfoliation , Anti-Bacterial Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cefepime , Cephalosporins/administration & dosage , Child , Clindamycin/administration & dosage , Drug Combinations , Female , Focal Infection, Dental/complications , Focal Infection, Dental/drug therapy , Humans , Injections, Intravenous , Mercaptopurine/administration & dosage , Methotrexate/administration & dosage , Neutropenia/complications , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Tooth Extraction , Tooth, Deciduous/microbiology
18.
J Obstet Gynecol Neonatal Nurs ; 39(5): 519-24, 2010.
Article in English | MEDLINE | ID: mdl-20919998

ABSTRACT

OBJECTIVE: To provide a comprehensive analysis of the temporal structure of sucking in full-term neonates. DESIGN: Descriptive study. SETTING: Newborn nursery in a city teaching institution. PATIENTS/PARTICIPANTS: Fifty-six full-term infants with a mean birth weight of 3,128±370 g completed sucking assessments on the first and second day of life. METHODS: A 5-minute sucking assessment was completed on the first and second day of life. Instruments included an Infant Nutritive Sucking Apparatus and the Anderson Behavioral Assessment Scale. RESULTS: The number of sucks (p<.001), intersuck width (p=.008) and interburst width (p<.05) were significantly different between the first and second day of life. On the second day of life the infants generated significantly more sucks, a decrease in interburst width and a decrease in intersuck width. There was a significant increase in the presence of an alert behavioral state from the first to second sucking assessment (p<.01). In addition, with a more alert infant state there was an increased time spent bursting (p<.001). CONCLUSION: Our results show that sucking analysis is sensitive to infant status and suggest that the development of sucking methodology can be considered as a useful clinical tool to assess the normal developmental course of sucking patterns.


Subject(s)
Infant, Newborn/physiology , Sucking Behavior , Child Development , Female , Humans , Longitudinal Studies , Male , Philadelphia , Regression Analysis
20.
J Dev Behav Pediatr ; 30(1): 16-22, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19194323

ABSTRACT

OBJECTIVES: The relationship between the pattern of sucking behavior of preterm infants during the early weeks of life and neurodevelopmental outcomes during the first year of life was evaluated. METHODS: The study sample consisted of 105 preterm infants (postmenstrual age [PMA] at birth = 30.5 +/- 2.8 weeks [mean +/- SD]; birth weight = 1476 +/- 460 g; mean length of hospital stay = 41.6 +/- 31.4 days). All infants received a 5-minute sucking test at 34 and at 40 weeks PMA, with outcomes evaluated at 6 and/or 12 months corrected gestational age via the Bayley Scales of Infant Development. RESULTS: As expected, 6- and 12-month values for the Psychomotor Developmental Index (PDI) and Mental Developmental Index (MDI) of the Bayley Scales of Infant Development were significantly below the normative levels established for infants delivered at term. A significant association between neonatal sucking pattern at 40 weeks PMA and developmental outcome at 12 months corrected gestational age was obtained. Each of the 3 simple sucking parameters evaluated (number of sucks, mean number of sucks per bursts and mean sucking pressure peaks), as well as a composite parameter (average of the respective parameter z-scores), was significantly related to both PDI and MDI at 12 months. CONCLUSIONS: Multivariable models, adjusting for PMA at birth, length of hospital stay, and other predictors, affirmed that sucking performance at 40 weeks PMA was a significant, independent predictor of developmental status 1 year later. Standardization of an instrument for neonatal sucking assessment may offer a cost-effective early screening strategy for preterm infants at greatest risk for developmental delay.


Subject(s)
Child Development/physiology , Infant Behavior/physiology , Infant, Premature/growth & development , Sucking Behavior/physiology , Birth Weight/physiology , Female , Gestational Age , Humans , Infant , Infant Behavior/psychology , Infant, Newborn , Infant, Premature/physiology , Infant, Premature/psychology , Male , Neuropsychological Tests , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...