Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Natl Cancer Inst ; 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38964343

ABSTRACT

PURPOSE: The National Cancer Institute (NCI) issued a 2021 memorandum adopting the American Society of Clinical Oncology (ASCO) and Friends of Cancer Research (Friends) task force recommendations to broaden clinical study eligibility criteria. They recommended that washout periods be eliminated for most prior cancer therapy and when required, to utilize evidence/rationale-based criteria. The Therapeutic Advances in Childhood Leukemia and Lymphoma (TACL) consortium responded to this guidance. PROCESS: A TACL task force reviewed the consortium's research portfolio, the relevant literature and guidance documents from ASCO-Friends, NCI, and US Food and Drug Administration (FDA) to make expert consensus and evidence-based recommendations for modernizing, broadening and codifying TACL-study washout periods while ensuring consistency with pediatric ethics and federal regulations. TACL's screening log was reviewed to estimate the impact that updated washout periods would have on patient inclusivity and recruitment. RESULTS: Over a 19-year period, 42 patients (14.6% of all screened ineligible (n = 287) patients), were identified as excluded from TACL early-phase studies exclusively due to not meeting washout criteria. An additional six (2.1%) did not meet washout and at least one other exclusion criterion. A new TACL washout guidance document was developed/adopted for use. Where washout criteria were not eliminated, rationale/evidenced-based criteria were established with citation. CONCLUSION: In an effort to reduce unnecessary exclusion from clinical trials, TACL created rationale/evidenced-based washout period standards largely following guidance from the NCI/ASCO-Friends recommendations. These new, expanded eligibility criteria are expected to increase access to TACL clinical trials while maintaining safety and scientific excellence.

2.
Clin Cancer Res ; 29(13): 2410-2418, 2023 07 05.
Article in English | MEDLINE | ID: mdl-37134194

ABSTRACT

PURPOSE: Cisplatin-induced hearing loss (CIHL) is common and permanent. As compared with earlier otoprotectants, we hypothesized N-acetylcysteine (NAC) offers potential for stronger otoprotection through stimulation of glutathione (GSH) production. This study tested the optimal dose, safety, and efficacy of NAC to prevent CIHL. PATIENTS AND METHODS: In this nonrandomized, controlled phase Ia/Ib trial, children and adolescents newly diagnosed with nonmetastatic, cisplatin-treated tumors received NAC intravenously 4 hours post-cisplatin. The trial performed dose-escalation across three dose levels to establish a safe dose that exceeded the targeted peak serum NAC concentration of 1.5 mmol/L (as identified from preclinical models). Patients with metastatic disease or who were otherwise ineligible were enrolled in an observation-only/control arm. To evaluate efficacy, serial age-appropriate audiology assessments were performed. Integrated biology examined genes involved in GSH metabolism and post-NAC GSH concentrations. RESULTS: Of 52 patients enrolled, 24 received NAC and 28 were in the control arm. The maximum tolerated dose was not reached; analysis of peak NAC concentration identified 450 mg/kg as the recommended phase II dose (RP2D). Infusion-related reactions were common. No severe adverse events occurred. Compared with the control arm, NAC decreased likelihood of CIHL at the end of cisplatin therapy [OR, 0.13; 95% confidence interval (CI), 0.021-0.847; P = 0.033] and recommendations for hearing intervention at end of study (OR, 0.082; 95% CI, 0.011-0.60; P = 0.014). NAC increased GSH; GSTP1 influenced risk for CIHL and NAC otoprotection. CONCLUSIONS: NAC was safe at the RP2D, with strong evidence for efficacy to prevent CIHL, warranting further development as a next-generation otoprotectant.


Subject(s)
Hearing Loss , Neoplasms , Adolescent , Humans , Child , Cisplatin/adverse effects , Acetylcysteine/therapeutic use , Acetylcysteine/adverse effects , Hearing Loss/chemically induced , Hearing Loss/prevention & control , Neoplasms/drug therapy , Neoplasms/chemically induced , Administration, Intravenous
4.
Int J Cancer ; 151(11): 1913-1924, 2022 Dec 01.
Article in English | MEDLINE | ID: mdl-35793239

ABSTRACT

Pediatric oncology patients are at risk for poor outcomes with respiratory viral infections. Outcome data for COVID-19 in children and young adults with cancer are needed; data are sparse for obese/overweight and adolescent and young adult subgroups. We conducted a single center cohort study of COVID-19 outcomes in patients younger than 25 years with cancer. Candidate hospitalization risk factors were analyzed via univariable and multivariable analyses. Eighty-seven patients with cancer and COVID-19 were identified. Most were Hispanic/Latinx (n = 63, 72%). Forty-two (48%) were overweight/obese. Anticancer therapy included chemotherapy only (n = 64, 74%), chimeric antigen receptor T-cells (CAR-T, n = 7), hematopoietic stem cell transplantation (HSCT, n = 12), or CAR-T and HSCT (n = 4). There was no COVID-19 related mortality. Twenty-six patients (30%) required COVID-19 related hospitalization; 4 required multiple hospitalizations. Nine (10%) had severe/critical infection; 6 needed intensive care. COVID-19 resulted in anticancer therapy delays in 22 (34%) of 64 patients on active therapy (median delay = 14 days). Factors associated with hospitalization included steroids within 2 weeks prior to infection, lymphopenia, previous significant non-COVID infection, and low COVID-19 PCR cycle threshold value. CAR-T recipients with B-cell aplasia tended to have severe/critical infection (3 of 7 patients). A COVID-19 antibody response was detected in 14 of 32 patients (44%). A substantial proportion of COVID-19 infected children and young adults with cancer require inpatient management; morbidity may be high in B-cell immunodeficiency. However, a majority of patients can be taken through chemotherapy without prolonged therapy delays. Viral load is a potential outcome predictor in COVID-19 in pediatric cancer.


Subject(s)
COVID-19 , Hematopoietic Stem Cell Transplantation , Neoplasms , Receptors, Chimeric Antigen , Adolescent , Child , Cohort Studies , Humans , Neoplasms/complications , Neoplasms/therapy , Obesity , Overweight , Young Adult
5.
Lancet Child Adolesc Health ; 5(4): 274-283, 2021 04.
Article in English | MEDLINE | ID: mdl-33581749

ABSTRACT

BACKGROUND: Cisplatin is used to treat a wide range of childhood cancers and cisplatin-induced hearing loss (CIHL) is a common and debilitating toxicity. We aimed to address persistent knowledge gaps in CIHL by establishing benchmarks for the prevalence of and risk factors for CIHL. METHODS: In this multi-institutional cohort study, children (age 0-14 years), adolescents, and young adults (age 15-39 years) diagnosed with a cisplatin-treated tumour from paediatric cancer centres, who had available cisplatin dosing information, and primary audiology data for central review from consortia located in Canada and the USA were eligible for inclusion. Audiology was centrally reviewed and CIHL graded using the consensus International Society of Pediatric Oncology (SIOP) Boston Ototoxicity Scale. We assessed the prevalence of moderate or severe CIHL (SIOP grade ≥2) at latest follow-up and end of therapy, in each demographic, diagnosis, and treatment group and their relative contributions to risk for CIHL. Secondary endpoints explored associations of cisplatin dose reductions and CIHL with survival. We also examined whether cisplatin dose reductions and CIHL were associated with survival outcomes. FINDINGS: We included 1481 patients who received cisplatin. Of the 1414 (95·5%) participants who had audiometry at latest follow-up (mean 3·9 years [SD 4·2] since diagnosis), 620 (43·8%) patients developed moderate or severe CIHL. The highest prevalence of CIHL was seen in the youngest patients (aged <5 years; 360 [59·4%] of 606 patients) and those with a CNS tumour (221 [50·9%] of 434 patients), hepatoblastoma (110 [65·9%] of 167 patients), or neuroblastoma (154 [62·1%] of 248 patients). After accounting for cumulative cisplatin dose, higher fractionated doses were associated with risk for CIHL (for each 10mg/m2 increase per day, adjusted odds ratio [aOR] 1·15 [95% CI 1·07-1·25]; for each 50 mg/m2 increase per cycle aOR 2·16 [1·37-3·51]). Vincristine exposure was newly identified as a risk factor for CIHL (aOR 3·55 [2·19-5·84]). Dose reductions and moderate or severe CIHL were not significantly associated with survival differences. INTERPRETATION: Using this large, multicentre cohort, benchmarks were established for the prevalence of CIHL in patients treated with cisplatin. Variations in cisplatin dosing confer additive risk for developing CIHL and warrant investigation as a potential approach to decrease the burden of therapy. FUNDING: US National Institutes of Health and National Institute on Deafness and Other Communication Disorders, US National Institutes of Health and National Cancer institute, St Baldrick's Foundation, Genome Canada, Genome British Columbia, Canadian Institutes of Health Research, the Canada Foundation for Innovation, University of British Columbia, British Columbia Children's Hospital Research Institute, British Columbia Provincial Health Services Authority, Health Canada, and C17 Research Network.


Subject(s)
Antineoplastic Agents/adverse effects , Cisplatin/adverse effects , Hearing Loss, Sensorineural/epidemiology , Neoplasms/drug therapy , Ototoxicity/epidemiology , Vincristine/therapeutic use , Adolescent , Adult , Age Distribution , Bone Neoplasms/drug therapy , Bone Neoplasms/epidemiology , Canada/epidemiology , Central Nervous System Neoplasms/drug therapy , Central Nervous System Neoplasms/epidemiology , Child , Child, Preschool , Cisplatin/therapeutic use , Dose-Response Relationship, Drug , Female , Hearing Loss, Sensorineural/chemically induced , Hepatoblastoma/drug therapy , Hepatoblastoma/epidemiology , Humans , Infant , Infant, Newborn , Liver Neoplasms/drug therapy , Liver Neoplasms/epidemiology , Male , Neoplasms, Germ Cell and Embryonal/drug therapy , Neoplasms, Germ Cell and Embryonal/epidemiology , Neuroblastoma/drug therapy , Neuroblastoma/epidemiology , Odds Ratio , Osteosarcoma/drug therapy , Osteosarcoma/epidemiology , Ototoxicity/etiology , Prevalence , Risk Factors , Severity of Illness Index , United States/epidemiology , Young Adult
6.
Pediatr Hematol Oncol ; 38(3): 239-250, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33170064

ABSTRACT

Malnutrition is associated with treatment-related toxicities (TRT) in adults with solid tumors and in children with leukemia. Few studies have assessed whether malnutrition in pediatric patients treated for solid tumors impacts risk for TRT, relapse, and/or survival. To address this knowledge gap, this retrospective study evaluated the association between body mass index (BMI) at diagnosis, and imputed BMI during therapy, on the prevalence of TRT, specific toxicities, relapse, and survival in pediatric patients with solid tumors treated with cisplatin-containing regimens. Kaplan-Meier curves and regression models evaluated the association between patient-specific characteristics (including BMI) and TRT, relapse, and survival. The cohort included 221 patients, of whom 22% were malnourished at diagnosis (10% were underweight and 12% were obese). Most patients (60%) experienced at least one severe TRT, and 30% developed more than one severe TRT. Most patients with obesity at diagnosis remained obese during therapy (62%). In multivariable analysis, obesity at diagnosis was significantly associated with a more than threefold greater risk for developing severe TRT (p = 0.037), specifically for acute or chronic kidney injury (p = 0.014). Obesity at diagnosis and adolescent and young adult age (≥15 years at diagnosis) were associated with worse event-free survival (hazard ratio [HR] 2.32, p = 0.024 and HR 2.28, p = 0.010, respectively) and overall survival (HR 3.69, p = 0.006 and HR 2.6, p = 0.012, respectively). Obese and older patients therefore constitute populations at risk for poorer outcomes. Prospective studies are warranted to gain further insight into the mechanism and role of obesity and adolescence in developing TRT and/or treatment failure.


Subject(s)
Antineoplastic Agents/adverse effects , Cisplatin/adverse effects , Malnutrition/complications , Neoplasms/complications , Neoplasms/drug therapy , Obesity/complications , Adolescent , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Body Mass Index , Child , Cisplatin/therapeutic use , Cisplatin/toxicity , Female , Humans , Male , Progression-Free Survival , Proportional Hazards Models , Retrospective Studies , Survival Analysis
7.
Clin Pharmacokinet ; 59(8): 1049-1061, 2020 08.
Article in English | MEDLINE | ID: mdl-32157629

ABSTRACT

BACKGROUND: Busulfan therapeutic drug monitoring (TDM) is necessary to better achieve the target exposure in children before hematopoietic stem cell transplantation (HSCT). However, TDM-based dosing may be challenging if intra-individual pharmacokinetic variability (also denoted inter-occasion variability [IOV]) occurs during therapy. OBJECTIVES: The objectives of this study were to describe and quantify busulfan IOV in children, and to investigate its potential determinants. METHODS: We performed a new analysis of published data from children who received intravenous busulfan over 4 days before HSCT. We calculated individual pharmacokinetic parameters on each day of therapy using a published population pharmacokinetic model of busulfan and analyzed their changes. Population estimation of IOV was also performed with non-linear mixed effects (NLME) modeling. Potential predictors of significant decrease in busulfan clearance (CL) were assessed by using machine learning approaches. RESULTS: IOV could be assessed in 136 children. Between day (D) 1 and D2, most patients (80%) experienced a decrease in busulfan CL, with a median change of - 7.9%. However, both large decreases (minimum, - 48.5%) and increases in CL (maximum, + 44%) were observed. Over D1-D3 of therapy, mean CL significantly decreased (- 15%), with a decrease of ≥ 20% in 22% of patients. Some patients also showed unstable CL from day to day. NLME modeling of IOV provided a coefficient of variation of 10.6% and 13.1% for volume of distribution (Vd) and CL, respectively. Some determinants of significant decreases in busulfan CL were identified, but predictive performance of the models was limited. CONCLUSIONS: Significant busulfan intra-individual variability may occur in children who receive a HSCT and is hardly predictable. The main risk is busulfan overexposure. Performing TDM repeatedly over therapy appears to be the best way to accurately estimate busulfan exposure and perform precision dosing.


Subject(s)
Busulfan , Hematopoietic Stem Cell Transplantation , Administration, Intravenous , Busulfan/pharmacokinetics , Child , Hematopoietic Stem Cells , Humans , Transplantation Conditioning
8.
Biol Blood Marrow Transplant ; 26(1): 150-156, 2020 01.
Article in English | MEDLINE | ID: mdl-31562961

ABSTRACT

Patients undergoing hematopoietic stem cell transplantation (HSCT) are often admitted to the hospital the day they are due to begin their conditioning regimen. Timely initiation of chemotherapy during regular work hours is important for patient safety, because during the night shift fewer physicians and pharmacists are available for urgent or unexpected matters. A review of the data at our institution from October 2017 to August 2018 showed that approximately one-third of our chemotherapy was started during the night shift (after 19:00), and the average time from admission to start of chemotherapy was over 8 hours. There are currently no well-defined benchmarks for timeliness of chemotherapy initiation. The aim of this quality improvement initiative was to increase the percentage of patients who start chemotherapy in the bone marrow transplant unit before 19:00 from 65% to >80% by March 31, 2019. We identified barriers to timely initiation of chemotherapy through process mapping and analysis of failures. The primary barriers were late admissions (after 12:00 pm) and time from admission to preparation of chemotherapy. We addressed mechanisms to mitigate these barriers through Plan-Do-Study-Act testing. Interventions included providing families specific admission times and their rationales and process for notifying pharmacy of admissions immediately on arrival. We used standardized control charts to measure the impact of the interventions on change. We also monitored medication errors before and during the intervention. From September 2018 to March 2019 the percentage of patients who started preparative chemotherapy before 19:00 increased from 65% to 85%, the percentage of patients who were admitted after 12:00 remained similar before (31%) and after the interventions (33%), and the average time from admission to start of chemotherapy decreased from 8.6 hours (513 minutes) to 6.4 hours (382 minutes). Medication errors were similar before (n = 50) and after the interventions (n = 43). Using standardized processes, we demonstrated a substantial decrease in the percentage of HSCT patients starting their preparative regimen after 19:00 without a concurrent increase in errors. We believe these interventions and measurements can be used in all transplant centers and have the potential to influence patient safety and outcomes.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Chronotherapy , Hematopoietic Stem Cell Transplantation , Hospitalization , Quality Improvement , Transplantation Conditioning , Adult , Female , Humans , Male , Time Factors
9.
Bone Marrow Transplant ; 54(3): 448-457, 2019 03.
Article in English | MEDLINE | ID: mdl-30108322

ABSTRACT

Veno-occlusive disease (VOD) is a severe adverse reaction to busulfan-containing regimens used in the preparation of children for hematopoietic stem cell transplantation (HSCT). We conducted a retrospective analysis of data to examine determinants of VOD in children who received IV busulfan for HSCT conditioning. Busulfan PK parameters as well as various indices (maximal concentration-Cmax, area under the concentration-time curve-AUC) were estimated using a validated Bayesian approach. The influence of available PK, demographic, and clinical variables on the incidence of VOD was evaluated by using logistic regression and classification and regression tree (CART) analyses. Among the 293 patients included, the mean age was 6.5 years and the mean actual body weight was 26.3 kg. The incidence of VOD was 25.6%. Busulfan Cmax as well as weight <9 kg or age <3 years were identified as independent predictors of VOD in logistic regression analysis. CART analysis identified busulfan Cmax over the entire regimen as the strongest predictor of VOD. This study suggests that busulfan-associated VOD is in part a concentration-dependent reaction. In addition, the youngest children showed the highest risk of VOD. These findings may have important implications for busulfan dosing and therapeutic drug monitoring practice in HSCT children.


Subject(s)
Busulfan/therapeutic use , Hematopoietic Stem Cell Transplantation/methods , Hepatic Veno-Occlusive Disease/drug therapy , Immunosuppressive Agents/therapeutic use , Transplantation Conditioning/methods , Administration, Intravenous , Adolescent , Adult , Busulfan/pharmacology , Child , Child, Preschool , Female , Humans , Immunosuppressive Agents/pharmacology , Infant , Infant, Newborn , Male , Retrospective Studies , Young Adult
10.
mBio ; 9(3)2018 05 08.
Article in English | MEDLINE | ID: mdl-29739902

ABSTRACT

Oncogenic Kaposi's sarcoma-associated herpesvirus (KSHV) is etiologically linked to primary effusion lymphoma (PEL), an aggressive and nontreatable malignancy commonly found in AIDS patients. In this study, we performed a high-throughput screening of 3,731 characterized compounds and identified cytarabine, approved by the FDA for treating numerous types of cancer, as a potent inhibitor of KSHV-induced PEL. We showed the high efficacy of cytarabine in the growth inhibition of various PEL cells by inducing cell cycle arrest and apoptosis. Cytarabine inhibited host DNA and RNA syntheses and therefore induced cellular cytotoxicity. Furthermore, cytarabine inhibited viral DNA and RNA syntheses and induced the rapid degradation of KSHV major latent protein LANA (latency-associated nuclear antigen), leading to the suppression of KSHV latent replication. Importantly, cytarabine effectively inhibited active KSHV replication and virion production in PEL cells. Finally, cytarabine treatments not only effectively inhibited the initiation and progression of PEL tumors but also induced regression of grown PEL tumors in a xenograft mouse model. Altogether, our study has identified cytarabine as a novel therapeutic agent for treating PEL as well as eliminating KSHV persistent infection.IMPORTANCE Primary effusion lymphoma is an aggressive malignancy caused by Kaposi's sarcoma-associated herpesvirus. The outcome of primary effusion lymphoma is dismal without specific treatment. Through a high-throughput screening of characterized compounds, we identified an FDA-approved compound, cytarabine, as a potent inhibitor of primary effusion lymphoma. We showed that cytarabine induced regression of PEL tumors in a xenograft mouse model. Cytarabine inhibited host and viral DNA and RNA syntheses, resulting in the induction of cytotoxicity. Of interest, cytarabine induced the degradation of KSHV major latent protein LANA, hence suppressing KSHV latent replication, which is required for PEL cell survival. Furthermore, cytarabine inhibited KSHV lytic replication program, preventing virion production. Our findings identified cytarabine as a novel therapeutic agent for treating PEL as well as for eliminating KSHV persistent infection. Since cytarabine is already approved by the FDA, it might be an ideal candidate for repurposing for PEL therapy and for further evaluation in advanced clinical trials.


Subject(s)
Antiviral Agents/administration & dosage , Cytarabine/administration & dosage , Herpesvirus 8, Human/physiology , Lymphoma, Primary Effusion/virology , Sarcoma, Kaposi/virology , Virus Latency/drug effects , Virus Replication/drug effects , Animals , Antigens, Viral/genetics , Antigens, Viral/metabolism , Apoptosis/drug effects , Cell Line, Tumor , DNA Replication/drug effects , Female , Herpesvirus 8, Human/drug effects , Herpesvirus 8, Human/genetics , Humans , Lymphoma, Primary Effusion/drug therapy , Lymphoma, Primary Effusion/physiopathology , Mice, Inbred NOD , Mice, SCID , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Sarcoma, Kaposi/drug therapy , Sarcoma, Kaposi/physiopathology
11.
J Neurooncol ; 138(1): 199-207, 2018 May.
Article in English | MEDLINE | ID: mdl-29427149

ABSTRACT

Single agent studies targeting the tumor microenvironment in central nervous system (CNS) tumors have largely been disappointing. Combination therapies targeting various pathways and cell types may be a more effective strategy. In this phase I study, we evaluated the combination of dasatinib, lenalidomide, and temozolomide in children with relapsed or refractory primary CNS tumors. Patients 1-21 years old with relapsed or refractory CNS tumors were eligible. Starting doses of dasatinib and lenalidomide were 65 mg/m2/dose twice daily and 55 mg/m2 once daily, respectively, while temozolomide was constant at 75 mg/m2 daily. The study followed a 3 + 3 phase I design, with a 4-week dose-limiting toxicity (DLT) evaluation period. Serial peripheral blood lymphocyte subsets were evaluated in consenting patients. Fifteen patients were enrolled and thirteen were DLT-evaluable. DLTs occurred in 5 patients, including somnolence and confusion (1 patient), hypokalemia (1 patient) and thrombocytopenia (3 patients). The maximum tolerated dose for the combination was dasatinib 65 mg/m2 twice daily, lenalidomide 40 mg/m2 daily, and temozolomide 75 mg/m2 daily, for 21 days followed by 7 days rest in repeating 28-day cycles. Transient increases in natural killer effector cells and cytotoxic T-cells were seen after 1 week of treatment. One out of six response-evaluable patients showed a partial response. The combination was feasible and relatively well tolerated in this heavily pre-treated population. The most common toxicities were hematologic. Preliminary evidence of clinical benefit was seen.


Subject(s)
Antineoplastic Agents/therapeutic use , Central Nervous System Neoplasms/drug therapy , Neoplasm Recurrence, Local/drug therapy , Adolescent , Antigens, CD/metabolism , Central Nervous System Neoplasms/pathology , Child , Child, Preschool , Combined Modality Therapy , Dasatinib/therapeutic use , Disease Progression , Dose-Response Relationship, Drug , Female , Humans , Infant , Lenalidomide/therapeutic use , Leukocytes/drug effects , Leukocytes/metabolism , Male , Temozolomide/therapeutic use , Young Adult
12.
Ther Drug Monit ; 38(3): 332-42, 2016 06.
Article in English | MEDLINE | ID: mdl-26829600

ABSTRACT

BACKGROUND: Busulfan dose adjustment is routinely guided by plasma concentration monitoring using 4-9 blood samples per dose adjustment, but a pharmacometric Bayesian approach could reduce this sample burden. METHODS: The authors developed a nonparametric population model with Pmetrics. They used it to simulate optimal initial busulfan dosages, and in a blinded manner, they compared dosage adjustments using the model in the BestDose software to dosage adjustments calculated by noncompartmental estimation of area under the time-concentration curve at a national reference laboratory in a cohort of patients not included in model building. RESULTS: Mean (range) age of the 53 model-building subjects was 7.8 years (0.2-19.0 years) and weight was 26.5 kg (5.6-78.0 kg), similar to nearly 120 validation subjects. There were 16.7 samples (6-26 samples) per subject to build the model. The BestDose cohort was also diverse: 10.2 years (0.25-18 years) and 46.4 kg (5.2-110.9 kg). Mean bias and imprecision of the 1-compartment model-predicted busulfan concentrations were 0.42% and 9.2%, and were similar in the validation cohorts. Initial dosages to achieve average concentrations of 600-900 ng/mL were 1.1 mg/kg (≤12 kg, 67% in the target range) and 1.0 mg/kg (>12 kg, 76% in the target range). Using all 9 concentrations after dose 1 in the Bayesian estimation of dose requirements, the mean (95% confidence interval) bias of BestDose calculations for the third dose was 0.2% (-2.4% to 2.9%, P = 0.85), compared with the standard noncompartmental method based on 9 concentrations. With 1 optimally timed concentration 15 minutes after the infusion (calculated with the authors' novel MMopt algorithm) bias was -9.2% (-16.7% to -1.5%, P = 0.02). With 2 concentrations at 15 minutes and 4 hours bias was only 1.9% (-0.3% to 4.2%, P = 0.08). CONCLUSIONS: BestDose accurately calculates busulfan intravenous dosage requirements to achieve target plasma exposures in children up to 18 years of age and 110 kg using only 2 blood samples per adjustment compared with 6-9 samples for standard noncompartmental dose calculations.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , Busulfan/administration & dosage , Models, Biological , Administration, Intravenous , Adolescent , Algorithms , Antineoplastic Agents, Alkylating/pharmacokinetics , Area Under Curve , Bayes Theorem , Bias , Busulfan/pharmacokinetics , Child , Child, Preschool , Dose-Response Relationship, Drug , Humans , Infant , Software , Young Adult
13.
Pediatr Infect Dis J ; 34(4): e78-84, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25764103

ABSTRACT

BACKGROUND: Intensifying treatment for pediatric acute myeloid leukemia (AML) has improved survival, with infections now being a leading cause of morbidity. Because quinolone prophylaxis is effective in adults with AML and in transplant populations, ciprofloxacin prophylaxis (CPx) was introduced as the standard for pediatric AML. We report here the impact of CPx in this population. METHODS: Prevalence of fever and neutropenia, frequency and pathogen spectrum of infections, antibiotic use, supportive care and mortality before and after implementation of CPx were retrospectively compared in children with AML. RESULTS: The cohort included 35 patients with de novo and 10 with relapsed AML, who together underwent 153 chemotherapy courses. Fever and neutropenia resulting in the use of empiric antibiotics occurred in 90% of chemotherapy courses (137/153); this was associated with proven bacteremia in 26%. The use of CPx did not change the incidence of febrile or infectious episodes, number of days of fever or antibiotic treatment or mortality. CPx was associated with a significant decrease in infections caused by Gram-negative rods (13.4% vs 4.7%) but a concomitant significant increase in bacteremia caused by viridans streptococci (12% vs 28%), resulting in no significant overall difference in the incidence of bacteremia between the 2 groups (35.9% vs 31.5%). CONCLUSIONS: CPx neither alter the incidence of overall bacteremia nor change the pattern of fever or use of supportive care. Our experience supports further investigation into the use of extended-spectrum quinolone prophylaxis during therapy for pediatric AML.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Antibiotic Prophylaxis/methods , Bacteremia/epidemiology , Bacteremia/prevention & control , Ciprofloxacin/administration & dosage , Leukemia, Myeloid, Acute/complications , Adolescent , Bacteremia/microbiology , Bacteria/classification , Bacteria/isolation & purification , Child , Child, Preschool , Cohort Studies , Female , Humans , Incidence , Male , Retrospective Studies , Treatment Outcome
14.
Pediatr Infect Dis J ; 33(3): 319-21, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24030353

ABSTRACT

Cancer therapy routinely requires Pneumocystis jiroveci prophylaxis. In those intolerant of trimethoprim/sulfamethoxazole, aerosolized pentamidine is convenient and effective. Intravenous pentamidine is often substituted in young children but its efficacy remains controversial. In this retrospective study of a large pediatric oncology cohort, we confirm intravenous pentamidine to be effective and well-tolerated as second-line prophylaxis across all ages.


Subject(s)
Antibiotic Prophylaxis/methods , Antifungal Agents/therapeutic use , Neoplasms/complications , Pentamidine/therapeutic use , Pneumocystis carinii , Pneumonia, Pneumocystis/drug therapy , Pneumonia, Pneumocystis/prevention & control , Administration, Intravenous , Adolescent , Adult , Antifungal Agents/administration & dosage , Antifungal Agents/adverse effects , Child , Child, Preschool , Female , Humans , Infant , Male , Neoplasms/epidemiology , Pediatrics , Pentamidine/administration & dosage , Pentamidine/adverse effects , Pneumonia, Pneumocystis/complications , Pneumonia, Pneumocystis/epidemiology , Retrospective Studies , Young Adult
15.
J Pathog ; 2011: 826169, 2011.
Article in English | MEDLINE | ID: mdl-22567342

ABSTRACT

A retrospective study was conducted, including 61 patients with long-term intravascular devices (IVDs) admitted to the Childrens Hospital Los Angeles with diverse underlying diseases, different types of catheters, and culture-proven catheter-related bloodstream infections (BSIs). Within these patients, 125 catheter-related BSIs occurred, and the incidence of monomicrobial and polymicrobial BSIs was evaluated. Risk factors for polymicrobial BSIs were determined. Forty-two BSIs contained more than one pathogen. These polymicrobial BSIs were observed more often in younger patients (<4.1 years versus ≥4.1 years) and less in patients using venous implanted ports. No other associations were found between the occurrences of polymicrobial BSIs and underlying diseases, other types of catheters, host defense status, parenteral nutrition, recurrences, or catheter removal. Patients with long-term IVDs at a younger age have a higher risk of developing a polymicrobial BSI. Future prospective studies should address the issue of polymicrobial infection in IVDs in more detail.

16.
Clin Infect Dis ; 50(1): 27-36, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19951112

ABSTRACT

BACKGROUND: Voriconazole pharmacokinetic and pharmacodynamic data are lacking in children. METHODS: Records at the Childrens Hospital Los Angeles were reviewed for children with > or =1 serum voriconazole concentration measured from 1 May 2006 through 1 June 2007. Information on demographic characteristics, dosing histories, serum concentrations, toxicity and survival, and outcomes was obtained. RESULTS: A total of 207 voriconazole measurements were obtained from 46 patients (age, 0.8-20.5 years). A 2-compartment Michaelis-Menten pharmacokinetic model fit the data best but explained only 80% of the observed variability. The crude mortality rate was 28%, and each trough serum voriconazole concentration <1000 ng/mL was associated with a 2.6-fold increased odds of death (95% confidence interval, 1.6-4.8; P=.002). Serum voriconazole concentrations were not associated with hepatotoxicity. Simulations predicted an intravenous dose of 7 mg/kg or an oral dose of 200 mg twice daily would achieve a trough >1000 ng/mL in most patients, but with a wide range of possible concentrations. CONCLUSIONS: We found a pharmacodynamic association between a voriconazole trough >1000 ng/mL and survival and marked pharmacokinetic variability, particularly after enteral dosing, justifying the measurement of serum concentrations.


Subject(s)
Antifungal Agents/pharmacokinetics , Mycoses/metabolism , Pyrimidines/pharmacokinetics , Triazoles/pharmacokinetics , Adolescent , Antifungal Agents/administration & dosage , Antifungal Agents/adverse effects , Antifungal Agents/blood , Child , Child, Preschool , Computer Simulation , Female , Humans , Infant , Linear Models , Male , Models, Biological , Mycoses/blood , Mycoses/drug therapy , Proportional Hazards Models , Pyrimidines/administration & dosage , Pyrimidines/adverse effects , Pyrimidines/blood , Retrospective Studies , Statistics, Nonparametric , Triazoles/administration & dosage , Triazoles/adverse effects , Triazoles/blood , Voriconazole , Young Adult
17.
Biol Blood Marrow Transplant ; 13(5): 584-93, 2007 May.
Article in English | MEDLINE | ID: mdl-17448918

ABSTRACT

Graft-versus-host disease (GVHD) is a cause of serious morbidity and mortality in >50% of recipients of unrelated hematopoietic stem cell transplantation (HSCT). We performed a trial using Campath 1 H pre- and post-HSCT in an attempt to decrease the incidence of GVHD without increasing the risk of infection or relapse. Patients were retrospectively compared to a population of patients who received antithymocyte globulin (ATG) pre- and post-HSCT. Twenty-seven patients were evaluated for this study. Fourteen patients received Campath 1H and 13 patients received ATG. Demographics of patients who received Campath 1H consisted of 9 males and 5 females, with a median age of 13 years (3-17.8 years). Thirteen patients received unrelated bone marrow and 1 patient received unrelated PBSC. Demographics of patients receiving ATG consisted of 9 males, 4 females with a median age of 7.4 years (21 months-19 years). Twelve patients received unrelated bone marrow and 1 patient received unrelated PBSC. Diagnoses were similar between the 2 groups. Patients who received Campath1H received a total dose of 52 mg/m(2) pre-HSCT and 20 mg/m(2) post-HSCT. Patients who received ATG received a total dose of 60 mg/kg pre-HSCT and 100 mg/kg post-HSCT. GVHD prophylaxis and supportive care measures were similar in both groups, including aggressive antimicrobial therapy. There was a significant difference in the incidence of severe (grade III and grade IV) GVHD between the 2 groups (Campath [0 of 14] versus ATG [6 of 13], P = .006). Among the patients who were transplanted for leukemia, there was no significant difference between the 2 groups in terms of relapse (Campath [2 of 14] versus ATG [4 of 9], P = 0.16). The 100-day survival between the 2 groups was not significantly different. Patients receiving Campath 1H had the presence of CD3(+) T cells (>30 cells/mL) in their peripheral blood later than in those who received ATG (64.5 days [Campath 1H] versus 27days [ATG], P = .001). The median time to the development of a normal PHA response occurred later in the Campath 1H arm (283 days[(Campath 1H] versus 88 days [ATG], P = .0001). The median time to an antigen specific response also occurred later in those receiving Campath 1H (365 days [Campath 1H] versus 150 days [ATG], P = .004). There was no significant difference between the 2 groups in terms of fungal or viral infections. Campath 1H is effective in decreasing the incidence of GVHD without increasing the risk of relapse. Although there is a significant delay in immune reconstitution, there was no increase in infectious complications or relapse in recipients of Campath 1H. Further studies are warranted to assess if a lack of difference in infection rates are still demonstrated in larger cohorts.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/therapeutic use , CD3 Complex/drug effects , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation/methods , T-Lymphocytes/drug effects , Adolescent , Alemtuzumab , Antibodies, Monoclonal, Humanized , Antilymphocyte Serum/therapeutic use , CD3 Complex/immunology , Child , Child, Preschool , Female , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Kaplan-Meier Estimate , Male , T-Lymphocytes/classification , Transplantation, Homologous/adverse effects
18.
Arch Pediatr Adolesc Med ; 160(10): 1049-53, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17018464

ABSTRACT

OBJECTIVES: To use the ethanol-lock technique (in conjunction with systemic antibiotics) to salvage central lines from removal and to prevent persistence of catheter-related infections among pediatric patients with long-term intravascular devices. DESIGN: Medical records of patients treated with ethanol locks were retrospectively reviewed from June 1, 2004, through June 22, 2005. SETTING: Childrens Hospital Los Angeles, Los Angeles, Calif, a tertiary care pediatric hospital. Patients Forty children with diverse underlying disorders were treated for 51 catheter-related infections using the Childrens Hospital Los Angeles ethanol-lock technique. INTERVENTIONS: Eligible infected central lines were instilled with a dose volume of 0.8 to 1.4 mL of 70% ethanol into the catheter lumen during 12 to 24 hours and then withdrawn. The volume of ethanol used was based on the type of intravascular device. MAIN OUTCOME MEASURES: Clearance of infection and incidence of recurrence. RESULTS: Of the 51 ethanol-lock treatments in 40 children, no catheters were removed because of persistent infection. Eighty-eight percent (45/51) of the treated episodes cleared without recurrence (defined as a relapse within 30 days with the same pathogen). Twelve (75%) of 16 polymicrobial isolates and 33 (94%) of 35 monomicrobial isolates were successfully treated. There were no adverse reactions or adverse effects reported. CONCLUSION: This retrospective study supports the use of the ethanol-lock technique in conjunction with systemic antibiotics as an effective and safe method to retain the use of a previously infected central venous catheter, decrease the need for line removal, and eradicate persistent pathogens in catheter-related infections.


Subject(s)
Anti-Infective Agents, Local/administration & dosage , Bacteremia/drug therapy , Catheterization, Central Venous/adverse effects , Catheters, Indwelling/microbiology , Ethanol/administration & dosage , Adolescent , Bacteremia/etiology , Bacteremia/microbiology , Child , Child, Preschool , Equipment Design , Female , Humans , Infant , Male , Retrospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL
...