Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Lifetime Data Anal ; 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38427151

RESUMEN

Linear mixed models are traditionally used for jointly modeling (multivariate) longitudinal outcomes and event-time(s). However, when the outcomes are non-Gaussian a quantile regression model is more appropriate. In addition, in the presence of some time-varying covariates, it might be of interest to see how the effects of different covariates vary from one quantile level (of outcomes) to the other, and consequently how the event-time changes across different quantiles. For such analyses linear quantile mixed models can be used, and an efficient computational algorithm can be developed. We analyze a dataset from the Acute Lymphocytic Leukemia (ALL) maintenance study conducted by Tata Medical Center, Kolkata. In this study, the patients suffering from ALL were treated with two standard drugs (6MP and MTx) for the first two years, and three biomarkers (e.g. lymphocyte count, neutrophil count and platelet count) were longitudinally measured. After treatment the patients were followed nearly for the next three years, and the relapse-time (if any) for each patient was recorded. For this dataset we develop a Bayesian quantile joint model for the three longitudinal biomarkers and time-to-relapse. We consider an Asymmetric Laplace Distribution (ALD) for each outcome, and exploit the mixture representation of the ALD for developing a Gibbs sampler algorithm to estimate the regression coefficients. Our proposed model allows different quantile levels for different biomarkers, but still simultaneously estimates the regression coefficients corresponding to a particular quantile combination. We infer that a higher lymphocyte count accelerates the chance of a relapse while a higher neutrophil count and a higher platelet count (jointly) reduce it. Also, we infer that across (almost) all quantiles 6MP reduces the lymphocyte count, while MTx increases the neutrophil count. Simulation studies are performed to assess the effectiveness of the proposed approach.

2.
Indian J Pediatr ; 91(1): 47-58, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37493925

RESUMEN

Cure rates in pediatric acute lymphoblastic leukemia (ALL) currently approach 90% in the developed world. Treatment involves 6-8 mo of intensive multi-drug chemotherapy followed by 24 mo of maintenance treatment (ALL-MT). The cornerstone of ALL-MT is the daily administration of oral 6-mercaptopurine (6MP), a purine analogue. 6MP is combined with weekly oral methotrexate (MTX), an antifolate drug, to augment therapeutic activity. Some protocols include additional chemotherapy drugs (such as vincristine and corticosteroids) during MT. The objective of ALL-MT is to ensure uninterrupted treatment at the highest tolerated doses of 6MP and MTX. This requires periodic adjustments of 6MP and MTX doses throughout treatment. Tolerance is determined through regular clinical assessments and careful monitoring of blood counts. Tolerated drug doses vary widely among patients, influenced by genetic and non-genetic factors, and require individualized dosing. Suboptimal treatment intensity in ALL-MT is associated with inferior outcomes and results from failure to treat at highest tolerated drug doses and/or interruptions in treatment due to non-adherence or toxicity. Management of MT thus requires close supervision to ensure treatment adherence, periodic drug dose modifications, and treatment to tolerance, while minimizing treatment interruptions due to toxicity. The review highlights these challenges and discusses approaches and strategies for the management of MT, focusing on the Indian context.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica , Leucemia-Linfoma Linfoblástico de Células Precursoras , Niño , Humanos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Mercaptopurina/uso terapéutico , Metotrexato/uso terapéutico
3.
J Biopharm Stat ; 34(1): 37-54, 2024 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-36882959

RESUMEN

The most common type of cancer diagnosed among children is the Acute Lymphocytic Leukemia (ALL). A study was conducted by Tata Translational Cancer Research Center (TTCRC) Kolkata, in which 236 children (diagnosed as ALL patients) were treated for the first two years (approximately) with two standard drugs (6MP and MTx) and were then followed nearly for the next 3 years. The goal is to identify the longitudinal biomarkers that are associated with time-to-relapse, and also to assess the effectiveness of the drugs. We develop a Bayesian joint model in which a linear mixed model is used to jointly model three biomarkers (i.e. white blood cell count, neutrophil count, and platelet count) and a semi-parametric proportional hazards model is used to model the time-to-relapse. Our proposed joint model can assess the effects of different covariates on the progression of the biomarkers, and the effects of the biomarkers (and the covariates) on time-to-relapse. In addition, the proposed joint model can impute the missing longitudinal biomarkers efficiently. Our analysis shows that the white blood cell (WBC) count is not associated with time-to-relapse, but the neutrophil count and the platelet count are significantly associated with it. We also infer that a lower dose of 6MP and a higher dose of MTx jointly result in a lower relapse probability in the follow-up period. Interestingly, we find that relapse probability is the lowest for the patients classified into the "high-risk" group at presentation. The effectiveness of the proposed joint model is assessed through the extensive simulation studies.


Asunto(s)
Mercaptopurina , Leucemia-Linfoma Linfoblástico de Células Precursoras , Niño , Humanos , Mercaptopurina/efectos adversos , Teorema de Bayes , Metotrexato/uso terapéutico , Leucemia-Linfoma Linfoblástico de Células Precursoras/inducido químicamente , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Recurrencia , Biomarcadores , Estudios Longitudinales
4.
Indian J Pediatr ; 91(2): 158-167, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37341952

RESUMEN

Outcomes for children with acute lymphoblastic leukemia (ALL) have improved worldwide to >85%. For those who relapse, outcomes have remained static at ~50% making relapsed acute lymphoblastic leukemia one of the leading causes of death in childhood cancers. Those relapsing within 18 mo in the bone marrow have a particularly dismal outcome. The mainstay of treatment is chemotherapy, local radiotherapy with or without hematopoietic stem cell transplantation (HSCT). Improved biological understanding of mechanisms of relapse and drug resistance, use of innovative strategies to identify the most effective and least toxic treatment regimens and global partnerships are needed to improve outcomes in these patients. Over the last decade, new therapeutic options and strategies have been developed for relapsed ALL including immunotherapies and cellular therapies. It is imperative to understand how and when to use these newer approaches in relapsed ALL. Increasingly, integrated precision oncology strategies are being used to individualize treatment of patients with relapsed ALL, especially in patients with poor response disease.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Leucemia-Linfoma Linfoblástico de Células Precursoras , Niño , Humanos , Medicina de Precisión , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Médula Ósea , Recurrencia
5.
Cancer Med ; 12(3): 3499-3508, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36812120

RESUMEN

BACKGROUND: To evaluate the treatment cost and cost effectiveness of a risk-stratified therapy to treat pediatric acute lymphoblastic leukemia (ALL) in India. METHODS: The cost of total treatment duration was calculated for a retrospective cohort of ALL children treated at a tertiary care facility. Children were risk stratified into standard (SR), intermediate (IR) and high (HR) for B-cell precursor ALL, and T-ALL. Cost of therapy was obtained from the hospital electronic billing systems and details of outpatient (OP) and inpatient (IP) from electronic medical records. Cost effectiveness was calculated in disability-adjusted life years. RESULTS: One hundred and forty five patients, SR (50), IR (36), HR (39), and T-ALL (20) were analyzed. Median cost of the entire treatment for SR, IR, HR, and T-ALL was found to be $3900, $5500, $7400, and $8700, respectively, with chemotherapy contributing to 25%-35% of total cost. Out-patient costs were significantly lower for SR (p < 0.0001). OP costs were higher than in-patient costs for SR and IR, while in-patient costs were higher in T-ALL. Costs for non-therapy admissions were significantly higher in HR and T-ALL (p < 0.0001), representing over 50% of costs of in-patient therapy. HR and T-ALL also had longer durations of non-therapy admissions. Based on WHO-CHOICE guidelines, the risk-stratified approach was very cost effective for all categories of patients. CONCLUSIONS: Risk-stratified approach to treat childhood ALL is very cost-effective for all categories in our setting. The cost for SR and IR patients is significantly reduced through decreased IP admissions for both, chemotherapy and non-chemotherapy reasons.


Asunto(s)
Leucemia-Linfoma Linfoblástico de Células Precursoras , Leucemia-Linfoma Linfoblástico de Células T Precursoras , Niño , Humanos , Estudios Retrospectivos , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Costos de la Atención en Salud
6.
Br J Haematol ; 198(1): 142-150, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35348200

RESUMEN

In successive UK clinical trials (UKALL 2003, UKALL 2011) for paediatric acute lymphoblastic leukaemia (ALL), polyethylene glycol-conjugated E. coli L-asparaginase (PEG-EcASNase) 1000 iu/m2 was administered intramuscularly with risk-stratified treatment. In induction, patients received two PEG-EcASNase doses, 14 days apart. Post-induction, non-high-risk patients (Regimens A, B) received 1-2 doses in delayed intensification (DI) while high-risk Regimen C patients received 6-10 PEG-EcASNase doses, including two in DI. Trial substudies monitored asparaginase (ASNase) activity, ASNase-related toxicity and ASNase-associated antibodies (total, 1112 patients). Median (interquartile range) trough plasma ASNase activity (14 ± 2 days post dose) following first and second induction doses and first DI dose was respectively 217 iu/l (144-307 iu/l), 265 iu/l (165-401 iu/l) and 292 iu/l (194-386 iu/l); 15% (138/910) samples showed subthreshold ASNase activity (<100 iu/l) at any trough time point. Older age was associated with lower (regression coefficient -9.5; p < 0.0001) and DI time point with higher ASNase activity (regression coefficient 29.9; p < 0.0001). Clinical hypersensitivity was observed in 3.8% (UKALL 2003) and 6% (UKALL 2011) of patients, and in 90% or more in Regimen C. A 7% (10/149) silent inactivation rate was observed in UKALL 2003. PEG-EcASNase schedule in UKALL paediatric trials is associated with low toxicity but wide interpatient variability. Therapeutic drug monitoring potentially permits optimisation through individualised asparaginase dosing.


Asunto(s)
Antineoplásicos , Leucemia-Linfoma Linfoblástico de Células Precursoras , Anticuerpos/uso terapéutico , Antineoplásicos/uso terapéutico , Asparaginasa , Niño , Monitoreo de Drogas , Escherichia coli , Humanos , Polietilenglicoles , Leucemia-Linfoma Linfoblástico de Células Precursoras/inducido químicamente , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico
7.
Trials ; 23(1): 102, 2022 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-35101099

RESUMEN

BACKGROUND: In the west, survival following treatment of childhood acute lymphoblastic leukaemia (ALL) approaches 90%. Outcomes in India do not exceed 70%. To address this disparity, the Indian Collaborative Childhood Leukaemia group (ICiCLe) developed in 2013 a contemporary treatment protocol for uniform risk-stratified management of first presentation ALL based on cytogenetics and minimal residual disease levels (MRD). A multicentre randomised clinical trial opened in 2016 (ICiCLe-ALL-14) and examines the benefit of randomised interventions to decrease toxicity and improve outcomes. METHODS: Patients 1-18 years with newly diagnosed ALL are categorised into four risk groups based on presentation features, tumour genetics and treatment response. Standard risk includes young (< 10 years) B cell precursor ALL (BCP-ALL) patients with low presentation leucocyte count (< 50 × 109/L) and no high-risk features. Intermediate risk includes BCP-ALL patients with no high-risk features but are older and have high presentation leucocyte counts and/or bulky disease. High risk includes BCP-ALL patients with any high-risk feature, including high-risk genetics, central nervous system leukaemia, poor prednisolone response at treatment day 8 and high MRD (≥ 0·01%) at the end of induction. Patients with T-lineage ALL constitute the fourth risk group. All patients receive four intensive treatment blocks (induction, consolidation, interim maintenance, delayed intensification) followed by 96 weeks of maintenance. Treatment intensity varies by risk group. Clinical data management is based on a web-based remote data capture system. The first randomisation examines the toxicity impact of a shorter induction schedule of prednisolone (3 vs 5 weeks) in young non-high-risk BCP-ALL. The second randomisation examines the survival benefit of substituting doxorubicin with mitoxantrone in delayed intensification for all patients. Primary outcome measures include event-free survival (overall, by risk groups), sepsis rates in induction (first randomisation) and event-free survival rates following second randomisation. DISCUSSION: ICiCLe-ALL-14 is the first multicentre randomised childhood cancer clinical trial in India. The pre-trial phase allowed standardisation of risk-stratification diagnostics and established the feasibility of collaborative practice, uniform treatment, patient enrolment and data capture. Pre-trial observations confirm the impact of risk-stratified therapy in reducing treatment-related deaths and costs. Uniform practice across centres allows patients to access care locally, potentially decreasing financial hardship and dislocation. TRIAL REGISTRATION: Clinical Trials Registry-India (CTRI) CTRI/2015/12/006434 . Registered on 11 December 2015.


Asunto(s)
Recurrencia Local de Neoplasia , Leucemia-Linfoma Linfoblástico de Células Precursoras , Humanos , Estudios Multicéntricos como Asunto , Neoplasia Residual , Leucemia-Linfoma Linfoblástico de Células Precursoras/diagnóstico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Estudios Prospectivos , Ensayos Clínicos Controlados Aleatorios como Asunto , Resultado del Tratamiento
8.
Pediatr Blood Cancer ; 68(11): e29046, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33939263

RESUMEN

BACKGROUND: The biotherapeutic asparaginase is a cornerstone of therapy in acute lymphoblastic leukaemia (ALL). With limited access to the original native Escherichia coli-derived asparaginase (EcASNase), a variety of EcASNase biogenerics are used in low-middle-income countries (LMICs). The variable quality of these biogenerics potentially influences clinical outcomes. PROCEDURE: Seven biogeneric EcASNases (P1-P7) marketed widely in India were evaluated, with P2 as an exemplar for in vivo monitoring. Therapeutic activity of P2 (10,000 IU/m2 /dose, intramuscular, every 72 hours) was monitored during induction therapy, and drug-related toxicities recorded. Molecular identity, purity and in vitro drug activity of seven biogenerics were characterised using multimodal analyses, and findings compared with reference EcASNase (R). RESULTS: In patients (N = 62) receiving P2, subtherapeutic asparaginase activity (<100 U/L) was observed in 66% (46/70) of trough timepoints (72 hours postdose) during induction. Twelve patients (19%), 11 with high-risk ALL, developed hypersensitivity. Isoforms of EcASNase were identified in all seven biogenerics. All generic products contained impurities with batch-to-batch variability. These included high levels of protein aggregates and host cell protein contamination. In vitro assays of EcASNase activity and leukaemia cell line cytotoxicity were not discriminatory. CONCLUSIONS: Our findings confirm widespread concerns over the unsatisfactory quality and therapeutic activity of native EcASNase biogenerics marketed in LMICs. Appropriate use of these products requires monitored studies to identify clinical suitability and determine appropriate dosing and schedule. For large parts of the world, assured access to high-quality asparaginases remains an unmet therapeutic need.


Asunto(s)
Antineoplásicos , Asparaginasa , Productos Biológicos/uso terapéutico , Medicamentos Genéricos/uso terapéutico , Leucemia-Linfoma Linfoblástico de Células Precursoras , Antineoplásicos/uso terapéutico , Asparaginasa/uso terapéutico , Escherichia coli/enzimología , Humanos , India , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Equivalencia Terapéutica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...