1
|
AlHarbi S, Taha A, Ashi A, Elimam N, Althubaiti S. Value of routine blood count surveillance in detecting relapse in acute lymphoblastic leukemia. Front Pediatr 2025; 12:1488686. [PMID: 39834493 PMCID: PMC11743496 DOI: 10.3389/fped.2024.1488686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 12/02/2024] [Indexed: 01/22/2025] Open
Abstract
Background Children with acute lymphoblastic leukemia (ALL) have excellent outcomes, with >85% survival without relapse following contemporary therapies. Clinical and complete blood count (CBC) assessments are commonly used surveillance methods to detect relapses. We aimed to evaluate the efficacy of routine blood testing for detecting relapse using a systematic method of assessing normal and abnormal results. Methods This a retrospective, single center study included children aged 1-14 years diagnosed with ALL who completed therapy and were in complete remission. Demographic data, leukemia subtypes, risk stratification, treatment responses, and outcomes were also reviewed. CBC tests were evaluated, and abnormal results were categorized. The relapse groups were classified as asymptomatic and symptomatic relapses. The clinical outcomes of relapse and complications were analyzed. The sensitivity, specificity, positive predictive value, and negative predictive value of surveillance laboratory tests for predicting relapse after the end of treatment were evaluated. Results In total, 187 patients underwent 2074 CBC tests. Ten patients underwent full surveillance, whereas the remaining patients underwent partial surveillance. The median number of surveillance blood draws per patient was 12. Relapse was observed in nine patients. Only three patients had asymptomatic relapses. Neutropenia, leukopenia, pancytopenia, thrombocytopenia, and anemia were observed in 98, 89, 10, 6, and 3 patients respectively. The sensitivity and specificity of neutropenia, leukopenia, thrombocytopenia, anemia, and pancytopenia were 11.11% and 47.9%, 0% and 50%, 33.3% and 98.31%, 0% and 99.4%, and 33.3% and 96.07%, respectively. No differences were observed between patients who had asymptomatic relapses and those whose clinical outcomes or consequences had symptomatic relapses. Conclusion Relapse after completion of therapy in ALL is rare. Regular blood count surveillance does not predict clinical outcomes or relapse. Prospective studies are required to assess appropriate risk-based surveillance and its effects on patient outcomes and quality of life.
Collapse
Affiliation(s)
- Sarah AlHarbi
- Department of Pediatric Oncology, King Abdullah Specialized Children’s Hospital, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Jeddah, Saudi Arabia
- King Abdullah International Medical Research Center, King Abdul Aziz Medical City, Jeddah, Saudi Arabia
| | - Areej Taha
- Department of Pediatric Oncology, King Abdullah Specialized Children’s Hospital, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Jeddah, Saudi Arabia
- King Abdullah International Medical Research Center, King Abdul Aziz Medical City, Jeddah, Saudi Arabia
| | - Ahmed Ashi
- King Abdullah International Medical Research Center, King Abdul Aziz Medical City, Jeddah, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Makkah—Jeddah Hwy, King Abdul Aziz Medical City, Jeddah, Saudi Arabia
| | - Naglla Elimam
- Department of Pediatric Oncology, King Abdullah Specialized Children’s Hospital, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Jeddah, Saudi Arabia
- King Abdullah International Medical Research Center, King Abdul Aziz Medical City, Jeddah, Saudi Arabia
| | - Sami Althubaiti
- Department of Pediatric Oncology, King Abdullah Specialized Children’s Hospital, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Jeddah, Saudi Arabia
- King Abdullah International Medical Research Center, King Abdul Aziz Medical City, Jeddah, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Makkah—Jeddah Hwy, King Abdul Aziz Medical City, Jeddah, Saudi Arabia
| |
Collapse
|
2
|
Bansal R, Bhojwani D, Sun BF, Sawardekar S, Wayne AS, Ouassil H, Gupte C, Marcelino C, Gonzalez Anaya MJ, Luna N, Peterson BS. Progression of brain injuries associated with methotrexate chemotherapy in childhood acute lymphoblastic leukemia. Pediatr Res 2025; 97:348-359. [PMID: 38951657 PMCID: PMC11798858 DOI: 10.1038/s41390-024-03351-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/18/2024] [Accepted: 06/07/2024] [Indexed: 07/03/2024]
Abstract
BACKGROUND Brain bases and progression of methotrexate-associated neurotoxicity and cognitive disturbances remain unknown. We tested whether brain abnormalities worsen in proportion to intrathecal methotrexate(IT-MTX) doses. METHODS In this prospective, longitudinal study, we recruited 19 patients with newly diagnosed acute lymphoblastic leukemia 4-to-20 years of age and 20 matched controls. We collected MRI and neuropsychological assessments at a pre-methotrexate baseline and at week 9, week 22, and year 1 during treatment. RESULTS Patients had baseline abnormalities in cortical and subcortical gray matter(GM), white matter(WM) volumes and microstructure, regional cerebral blood flow, and neuronal density. Abnormalities of GM, blood flow, and metabolites worsened in direct proportions to IT-MTX doses. WM abnormalities persisted until week 22 but normalized by year 1. Brain injuries were localized to dorsal and ventral attentional and frontoparietal cognitive networks. Patients had cognitive deficits at baseline that persisted at 1-year follow-up. CONCLUSIONS Baseline abnormalities are likely a consequence of neuroinflammation and oxidative stress. Baseline abnormalities in WM microstructure and volumes, and blood flow persisted until week 22 but normalized by year 1, likely due to treatment and its effects on reducing inflammation. The cytotoxic effects of IT-MTX, however, likely contributed to continued, progressive cortical thinning and reductions in neuronal density, thereby contributing to enduring cognitive deficits. IMPACT Brain abnormalities at a pre-methotrexate baseline likely are due to acute illness. The cytotoxic effects of intrathecal MTX contribute to progressive cortical thinning, reductions in neuronal density, and enduring cognitive deficits. Baseline white matter abnormalities may have normalized via methotrexate treatment and decreasing neuroinflammation. Corticosteroid and leucovorin conferred neuroprotective effects. Our findings suggest that the administration of neuroprotective and anti-inflammatory agents should be considered even earlier than they are currently administered. The neuroprotective effects of leucovorin suggest that strategies may be developed that extend the duration of this intervention or adapt it for use in standard risk patients.
Collapse
Affiliation(s)
- Ravi Bansal
- Department of Psychiatry, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
- Institute for the Developing Mind, Children's Hospital Los Angeles, Los Angeles, CA, USA.
| | - Deepa Bhojwani
- Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Norris Comprehensive Cancer Center and Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Bernice F Sun
- Institute for the Developing Mind, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Siddhant Sawardekar
- Institute for the Developing Mind, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Alan S Wayne
- Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Norris Comprehensive Cancer Center and Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Hannah Ouassil
- College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, USA
| | - Chaitanya Gupte
- Institute for the Developing Mind, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Courtney Marcelino
- Institute for the Developing Mind, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Maria J Gonzalez Anaya
- Institute for the Developing Mind, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Natalia Luna
- Institute for the Developing Mind, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Bradley S Peterson
- Department of Psychiatry, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Institute for the Developing Mind, Children's Hospital Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
3
|
Salceda-Rivera V, Ortiz-Lazareno PC, Hernández-Flores G, Vazquez-Urrutia JR, Meza-Arroyo J, Pardo-Zepeda M, Romo-Rubio H, Barba-Barba C, Sánchez-Zubieta F, Barrón-Gallardo CA, Gonzalez-Ramella O, Bravo-Cuellar A. Very early remission and increased apoptosis with the use of Pentoxifylline in children with acute lymphoblastic leukemia. Front Oncol 2024; 14:1401262. [PMID: 39421449 PMCID: PMC11484046 DOI: 10.3389/fonc.2024.1401262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 09/09/2024] [Indexed: 10/19/2024] Open
Abstract
Introduction Despite the improvement in survival in acute lymphoblastic leukemia (ALL), there are still cases with evasion of chemotherapy-induced apoptosis. The IKK/NF-κB signaling pathway contributes to antiapoptotic gene expression. Pentoxifylline (PTX) inhibits IkB phosphorylation, blocking NF-κB and antiapoptotic activity. Methods We conducted a randomized, double-blind clinical trial on pediatric ALL patients undergoing induction therapy, assigning them to PTX or placebo group. Bone marrow aspirates were obtained on days 1, 8, 15, and 22. Apoptosis was assessed using Annexin-V/propidium iodide. Results Results indicated that the PTX group exhibited higher apoptosis on day-8 (41.3% vs. 19.4%, p =0.029) and day-15 (35.0% vs. 14.2%, p <0.01). On day-8, the PTX group displayed an MRD of 0.25% vs. 18.2% (p <0.01) in placebo group; on day-15, the PTX group demonstrated an MRD of 0.09% vs. 1.4% (p =0.02). Patients achieving an MRD <0.01% on day-8 demonstrated a 3-year Overall Survival (OS) of 81.6% vs. 58.3% (p =0.03); on day-15, patients with MRD <0.01% had a 3-year OS of 77.9% vs. 54.5% (p =0.03). The PTX group achieved an MRD of <0.01% earlier on days-8 and 15, along with a higher apoptosis rate, indicating a more favorable therapeutic response. In the entire cohort, patients achieving MRD <0.01% on day-8 or 15 displayed superior OS. Conclusion Our study demonstrates that PTX enhances apoptosis and reduces MRD in pediatric acute lymphoblastic leukemia patients. Clinical trial registration https://clinicaltrials.gov/, identifier NCT02451774.
Collapse
Affiliation(s)
- Violeta Salceda-Rivera
- Immunology Division, Western Biomedical Research Center, Mexican Social Security Institute, Guadalajara, JAL, Mexico
- Doctoral Program in Biomedical Sciences, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, JAL, Mexico
- Department of Pediatric Hemato-Oncology, Hospital Civil de Guadalajara “Dr. Juan I. Menchaca”, Guadalajara, JAL, Mexico
| | - Pablo C. Ortiz-Lazareno
- Immunology Division, Western Biomedical Research Center, Mexican Social Security Institute, Guadalajara, JAL, Mexico
- Doctoral Program in Biomedical Sciences, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, JAL, Mexico
| | - Georgina Hernández-Flores
- Immunology Division, Western Biomedical Research Center, Mexican Social Security Institute, Guadalajara, JAL, Mexico
| | - Jorge R. Vazquez-Urrutia
- Immunology Division, Western Biomedical Research Center, Mexican Social Security Institute, Guadalajara, JAL, Mexico
- Centro Universitario de Ciencias de la Salud, School of Medicine, Universidad de Guadalajara, Guadalajara, JAL, Mexico
| | - Jesus Meza-Arroyo
- Doctoral Program in Biomedical Sciences, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, JAL, Mexico
| | - Monzerrat Pardo-Zepeda
- Doctoral Program in Biomedical Sciences, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, JAL, Mexico
- Department of Pediatric Hemato-Oncology, Hospital Civil de Guadalajara “Dr. Juan I. Menchaca”, Guadalajara, JAL, Mexico
| | - Hugo Romo-Rubio
- Department of Pediatric Hemato-Oncology, Hospital Civil de Guadalajara “Dr. Juan I. Menchaca”, Guadalajara, JAL, Mexico
| | - Cesar Barba-Barba
- Department of Pediatric Hemato-Oncology, Hospital Civil de Guadalajara “Dr. Juan I. Menchaca”, Guadalajara, JAL, Mexico
| | - Fernando Sánchez-Zubieta
- Department of Pediatric Hemato-Oncology, Hospital Civil de Guadalajara “Dr. Juan I. Menchaca”, Guadalajara, JAL, Mexico
- Departamento de Clinicas de Reproduccion Humana, Crecimiento y Desarrollo Infantil, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, JAL, Mexico
| | - Carlos Alfredo Barrón-Gallardo
- Departamento Académico de Disciplinas Especializantes de Ciencias de la Salud, Universidad Autonoma de Guadalajara, Zapopan, JAL, Mexico
| | - Oscar Gonzalez-Ramella
- Doctoral Program in Biomedical Sciences, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, JAL, Mexico
- Department of Pediatric Hemato-Oncology, Hospital Civil de Guadalajara “Dr. Juan I. Menchaca”, Guadalajara, JAL, Mexico
- Centro Universitario de Ciencias de la Salud, School of Medicine, Universidad de Guadalajara, Guadalajara, JAL, Mexico
| | - Alejandro Bravo-Cuellar
- Immunology Division, Western Biomedical Research Center, Mexican Social Security Institute, Guadalajara, JAL, Mexico
- Doctoral Program in Biomedical Sciences, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, JAL, Mexico
- Centro Universitario de los Altos, Universidad de Guadalajara, Tepatitlán de Morelos, JAL, Mexico
| |
Collapse
|
4
|
Khawaji ZY, Khawaji NY, Alahmadi MA, Elmoneim AA. Prediction of Response to FDA-Approved Targeted Therapy and Immunotherapy in Acute Lymphoblastic Leukemia (ALL). Curr Treat Options Oncol 2024; 25:1163-1183. [PMID: 39102166 DOI: 10.1007/s11864-024-01237-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2024] [Indexed: 08/06/2024]
Abstract
OPINION STATEMENT Acute lymphoblastic leukemia (ALL) represents the predominant cancer in pediatric populations, though its occurrence in adults is relatively rare. Pre-treatment risk stratification is crucial for predicting prognosis. Important factors for assessment include patient age, white blood cell (WBC) count at diagnosis, extramedullary involvement, immunophenotype, and cytogenetic aberrations. Minimal residual disease (MRD), primarily assessed by flow cytometry following remission, plays a substantial role in guiding management plans. Over the past decade, significant advancements in ALL outcomes have been witnessed. Conventional chemotherapy has remarkably reduced mortality rates; however, its intensive nature raises safety concerns and has led to the emergence of treatment-resistant cases with recurrence of relapses. Consequently, The U.S. Food and Drug Administration (FDA) has approved several novel treatments for relapsed/refractory ALL due to their demonstrated efficacy, as indicated by improved complete remission and survival rates. These treatments include tyrosine kinase inhibitors (TKIs), the anti-CD19 monoclonal antibody blinatumomab, anti-CD22 inotuzumab ozogamicin, anti-CD20 rituximab, and chimeric antigen receptor (CAR) T-cell therapy. Identifying the variables that influence treatment decisions is a pressing necessity for tailoring therapy based on heterogeneous patient characteristics. Key predictive factors identified in various observational studies and clinical trials include prelymphodepletion disease burden, complex genetic abnormalities, and MRD. Furthermore, the development of serious adverse events following treatment could be anticipated through predictive models, allowing for appropriate prophylactic measures to be considered. The ultimate aim is to incorporate the concept of precision medicine in the field of ALL through valid prediction platform to facilitate the selection of the most suitable treatment approach.
Collapse
Affiliation(s)
| | | | | | - Abeer Abd Elmoneim
- Women and Child Health Department, Taibah University, Madinah, Kingdom of Saudi Arabia
- 2nd Affiliation: Pediatric Department, Faculty of Medicine, Sohag University, Sohag, Egypt
| |
Collapse
|
5
|
İnce T, Gürocak ÖT, Totur G, Yılmaz Ş, Ören H, Aydın A. Waning of Humoral Immunity to Vaccine-Preventable Diseases in Children Treated for Acute Lymphoblastic Leukemia: A Single-Center Retrospective Cross-Sectional Analysis. Turk J Haematol 2024; 41:160-166. [PMID: 38801016 PMCID: PMC11589364 DOI: 10.4274/tjh.galenos.2024.2024.0150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 05/25/2024] [Indexed: 05/29/2024] Open
Abstract
Objective The survival rates of children with acute lymphoblastic leukemia (ALL) have improved over the years, but infections remain a significant cause of morbidity and mortality. Chemotherapy has a range of harmful side effects including the loss of protective antibodies against vaccine-preventable diseases. The objective of this study was to evaluate the serological status of pediatric ALL cases before and after intensive chemotherapy. Materials and Methods Children treated and followed for ALL at Dokuz Eylül University were included in this retrospective cross-sectional study. Antibody levels against hepatitis A, hepatitis B, and rubella were routinely assessed at both the time of diagnosis and 6 months after completion of chemotherapy. Measles, mumps, and varicella antibody levels were evaluated at only 6 months after treatment. Results Seventy-eight children who completed chemotherapy for ALL were enrolled in the study. All participants had non-protective antibody levels for at least one of the diseases. The highest seropositivity rate was found for hepatitis A (55.1%) and the lowest for measles (17.9%) after chemotherapy. Overall, 50.7%, 30.6%, and 45.7% of the patients significantly lost their humoral immunity against hepatitis B, hepatitis A, and rubella, respectively. Patients in the higher-risk group for ALL had lower seropositivity rates than patients of the other risk groups. There were statistically significant relationships between the protective antibody rates for hepatitis A and varicella and the ages of the patients. Except for hepatitis A vaccination, pre-chemotherapy vaccination did not affect post-chemotherapy serology. On the other hand, all children with a history of varicella before diagnosis showed immunity after chemotherapy. Conclusion Patients with ALL, including those previously fully vaccinated, are at great risk of infection due to the decrease in protective antibody levels after chemotherapy. There is a need for routine post-chemotherapy serological testing and re-vaccination based on the results obtained.
Collapse
Affiliation(s)
- Tolga İnce
- Dokuz Eylül University Faculty of Medicine, Department of Pediatrics, Division of Social Pediatrics, İzmir, Türkiye
| | - Özlem Tüfekçi Gürocak
- Dokuz Eylül University Faculty of Medicine, Department of Pediatrics, Division of Pediatric Hematology, İzmir, Türkiye
| | - Gülberat Totur
- University of Health Sciences Türkiye, İzmir Tepecik Training and Research Hospital, Clinic of Pediatrics, İzmir, Türkiye
| | - Şebnem Yılmaz
- Dokuz Eylül University Faculty of Medicine, Department of Pediatrics, Division of Pediatric Hematology, İzmir, Türkiye
| | - Hale Ören
- Dokuz Eylül University Faculty of Medicine, Department of Pediatrics, Division of Pediatric Hematology, İzmir, Türkiye
| | - Adem Aydın
- Dokuz Eylül University Faculty of Medicine, Department of Pediatrics, Division of Social Pediatrics, İzmir, Türkiye
| |
Collapse
|
6
|
Muhammad M, Saifo M, Aljamali M, Alali M, Ghanem KM. The frequency of NUDT15 rs116855232 and its impact on mercaptopurine-induced toxicity in Syrian children with acute lymphoblastic leukemia. Front Oncol 2024; 14:1334846. [PMID: 38562167 PMCID: PMC10982510 DOI: 10.3389/fonc.2024.1334846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 02/27/2024] [Indexed: 04/04/2024] Open
Abstract
Introduction Polymorphisms in NUDT15 may result in differences in mercaptopurine-induced toxicity. This study aimed to identify the frequency of the NUDT15 (c.415C>T; rs116855232) polymorphism and investigate the effect of this polymorphism on mercaptopurine-induced toxicity in a population of Syrian patients with childhood acute lymphoblastic leukemia (ALL). Methods This is a retrospective study that included children with ALL reaching at least 6 months of maintenance therapy. The NUDT15 genotyping was determined using standard targeted sequencing of polymerase chain reaction products. The odds ratio (OR) for the association between toxicity and genotype was evaluated. Results A total of 92 patients were enrolled. The majority of the patients in the study population were low-risk (63.04%), followed by intermediate-risk (25%), and high-risk (11.96%). There were 5 patients (5.4%) with NUDT15 (c.415C>T; rs116855232) CT genotype, and 1 patient (1.08%) with NUDT15 TT genotype, with allele frequencies of C=0.962 and T=0.038. The mercaptopurine median dose intensity was 100%, 54.69%, and 5% for the genotypes CC, CT, and TT, respectively (P=0.009). Early onset leukopenia was significantly associated with the NUDT15 polymorphism (OR: 6.16, 95% CI: 1.11-34.18, P=0.037). There was no association between the NUDT15 genotype and hepatotoxicity. Conclusion Approximately 6.5% of the study population exhibited reduced NUDT15 activity. The mercaptopurine dose intensity was considerably low in NUDT15 rs116855232 TT genotype compared with CT and CC. The dosage of mercaptopurine should be adjusted according to the NUDT15 genotype in pediatric patients with ALL.
Collapse
Affiliation(s)
- Muhammad Muhammad
- BASMA Pediatric Oncology Unit, Damascus, Syria
- Department of Oncology, Albairouni University Hospital, Faculty of Medicine, Damascus University, Damascus, Syria
| | - Maher Saifo
- Department of Oncology, Albairouni University Hospital, Faculty of Medicine, Damascus University, Damascus, Syria
- Faculty of Medicine, Damascus University, Damascus, Syria
| | - Majd Aljamali
- Department of Biochemistry and Microbiology, Faculty of Pharmacy, Damascus University, Damascus, Syria
- National Commission for Biotechnology (NCBT), Damascus, Syria
| | - Mousa Alali
- Department of Oncology, Albairouni University Hospital, Faculty of Medicine, Damascus University, Damascus, Syria
| | | |
Collapse
|
7
|
Kim H, Ikuse T, Matsui T, Sakaguchi H, Ishiguro A, Shoji K. Factors distinguishing leukemoid reaction from hematological malignancy in children. Pediatr Int 2024; 66:e15837. [PMID: 39692201 DOI: 10.1111/ped.15837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 07/02/2024] [Accepted: 07/31/2024] [Indexed: 12/19/2024]
Abstract
BACKGROUND Information on the etiology and prognosis for leukemoid reaction (LR) in children is still limited and little is known about the factors that distinguish LR from hematological malignancy (HM). METHODS This was a single-center, case-control study. Pediatric patients (<18 years) with a white blood cell (WBC) count of 50,000/μL or more were included in the study. Clinical information and laboratory test results were extracted from the electronic medical records. Patients were divided into the LR and HM groups. Logistic regression analysis was performed to investigate the factors that discriminated LR from HM. RESULTS We found 214 cases (115 cases in the LR group and 99 cases in the HM group) eligible for analysis. Approximately half of the LR cases were due to infectious diseases (n = 58, 50%); bacteremia and respiratory infections were the most common infections. Age younger than 2 years (odds ratio [95% confidence interval]) (2.154 [0.690-6.727]), presence of known underlying diseases (10.006 [3.119-32.102]), WBC count <60,555/μL (20.676 [6.357-67.251]), platelet count 118,000/μL or higher (15.059 [3.876-58.504]), lactate dehydrogenase (LDH) below 781 U/L (4.219 [1.378-12.915]), and C-reactive protein (CRP) ≥0.91 mg/dL (10.568 [2.736-40.825]) were identified as the predictive factors for LR by logistic regression analysis. Thirty-day mortality was higher in the LR group than in the HM group but the difference was not statistically significant (13% vs. 6%, p = 0.087). CONCLUSIONS Approximately half of the etiology of LR was infectious diseases and prognosis was poorer for LR than for HM. Age, presence of known underlying conditions, and laboratory tests, including WBC count, platelet count, LDH, and CRP, may be useful in distinguishing LR from HM.
Collapse
Affiliation(s)
- Heeyung Kim
- Center for Postgraduate Education and Training, National Center for Child Health and Development, Tokyo, Japan
| | - Tatsuki Ikuse
- Division of Infectious Diseases, Department of Medical Subspecialties, National Center for Child Health and Development, Tokyo, Japan
| | - Toshihiro Matsui
- Division of Infectious Diseases, Department of Medical Subspecialties, National Center for Child Health and Development, Tokyo, Japan
| | - Hirotoshi Sakaguchi
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Akira Ishiguro
- Center for Postgraduate Education and Training, National Center for Child Health and Development, Tokyo, Japan
| | - Kensuke Shoji
- Division of Infectious Diseases, Department of Medical Subspecialties, National Center for Child Health and Development, Tokyo, Japan
| |
Collapse
|
8
|
Jodłowska A, Postek-Stefańska L. Tooth Abnormalities and Their Age-Dependent Occurrence in Leukemia Survivors. Cancers (Basel) 2023; 15:5420. [PMID: 38001680 PMCID: PMC10670488 DOI: 10.3390/cancers15225420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 11/12/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
The multidrug nature of anticancer treatment and different treatment protocols used in the studies are likely to be a major limitation in establishing real risk factors determining the occurrence of dental abnormalities. The authors aimed to establish a relationship between the duration and the dose of chemotherapy and the number of tooth adverse effects in the group receiving the same treatment. Of the 40 anticancer therapy recipients who attended the outpatient dental clinic, 7 leukemia survivors receiving the treatment according to the ALL IC-BFM 2002 protocol were selected. The study group consisted of four females and three males aged 92 to 207 months at the time of dental examination and 29 to 91 months at leukemia diagnosis. As a result of the clinical and radiological examination, dental abnormalities such as agenesis, tooth size reduction, root abnormalities, and taurodontia were identified, and the medical records of all survivors were reviewed in terms of drugs administered, their doses, and treatment schedules. No correlation was observed between the treatment duration of an intensive therapy, the entire therapy, and the number of tooth abnormalities. No relationship was also found between the number of dental abnormalities and the cumulative dose of vincristine, L-asparaginase, methotrexate, cyclophosphamide, cytarabine, and 6-mercaptopurine. The age at the onset of antineoplastic therapy is likely to be the strongest risk factor for toxic injury during tooth development.
Collapse
Affiliation(s)
- Anna Jodłowska
- Department of Pediatric Dentistry, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland;
| | | |
Collapse
|
9
|
Zhou T, Karrs J, Ho T, Doverte A, Kochenderfer JN, Shah NN, Yuan CM, Wang HW. Circulating CD22+/CD19-/CD24- progenitors and CD22+/CD19+/CD24- mature B cells: Diagnostic pitfalls for minimal residual disease detection in B-lymphoblastic leukemia. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2023; 104:294-303. [PMID: 36433814 PMCID: PMC10735170 DOI: 10.1002/cyto.b.22104] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 11/09/2022] [Accepted: 11/14/2022] [Indexed: 08/20/2023]
Abstract
BACKGROUND Multiparametric flow cytometry (MFC) has become a powerful tool in minimal residual disease (MRD) detection in B-lymphoblastic leukemia/lymphoma (B-ALL). In the setting of targeted immunotherapy, B-ALL MRD detection often relies on alterative gating strategies, such as the utilization of CD22 and CD24. It is important to depict the full diversity of normal cell populations included in the alternative B-cell gating methods to avoid false-positive results. We describe two CD22-positive non-neoplastic cell populations in the peripheral blood (PB), including one progenitor population of uncertain lineage and one mature B-cell population, which are immunophenotypic mimics of B-ALL. METHODS Using MFC, we investigated the prevalence and phenotypic profiles of both CD22-positive populations in 278 blood samples from 52 patients with B-ALL; these were obtained pre- and post-treatment with CD19 and/or CD22 CAR-T therapies. We further assessed whether these two populations in the blood were exclusively associated with B-ALL or recent anticancer therapies, by performing the same analysis on patients diagnosed with other hematological malignancies but in long-term MRD remission. RESULTS The progenitor population and mature B-cell population were detected at low levels in PB of 61.5% and 44.2% of B-ALL patients, respectively. Both cell types showed distinctive and highly consistent antigen expression patterns that are reliably distinguishable from B-ALL. Furthermore, their presence is not restricted solely to B-ALL or recent therapy. CONCLUSIONS Our findings aid in building a complete immunophenotypic profile of normal cell populations in PB, thereby preventing misdiagnosis of B-ALL MRD and inappropriate management.
Collapse
Affiliation(s)
- Ting Zhou
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jeremiah Karrs
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Truc Ho
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Alyssa Doverte
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - James N. Kochenderfer
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Nirali N. Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Constance M. Yuan
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Hao-Wei Wang
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
10
|
Sharma Poudyal B, Paudel B, Tuladhar S, Neupane S, Bhattarai K, Joshi U. Outcome of ALL With ALL-BFM-95 Protocol in Nepal. JCO Glob Oncol 2023; 9:e2200408. [PMID: 37428991 PMCID: PMC10581644 DOI: 10.1200/go.22.00408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 04/13/2023] [Accepted: 05/24/2023] [Indexed: 07/12/2023] Open
Abstract
PURPOSE Data on survival outcomes in patients with acute lymphoblastic leukemia (ALL) originating from Nepal are limited. We aim to present the real-world data on treatment outcomes of patients with de novo ALL treated with pediatric ALL-Berlin-Frankfurt-Muenster (BFM)-95 protocol in Nepal. PATIENTS AND METHODS We used the medical records of 103 consecutive patients with ALL treated in our center from 2013 to 2016 to evaluate the overall survival (OS) and relapse-free survival (RFS) and analyzed the effects of clinicopathologic factors on survival outcomes in patients with ALL. RESULTS The 3-year OS and RFS in the entire cohort was 89.4% (95% CI, 82.1 to 96.7) and 87.3% (95% CI, 79.8 to 94.7), with a mean OS and RFS of 79.4 months (95% CI, 74.2 to 84.5) and 76.6 months (95% CI, 70.8 to 82.4), respectively. Patients with prednisone good response (PGR) showed better mean OS and RFS, whereas complete marrow response on day 33 was associated with better mean OS alone. Patients with Philadelphia (Ph)-positive ALL showed worse mean RFS compared to those with Ph-negative status. On multivariate analysis, PGR (hazard ratio [HR], 0.11; 95% CI, 0.03 to 0.49; P = .004) and sagittal vein thrombosis (SVT; HR, 5.95; 95% CI, 1.30 to 27.18; P = .02) were the only independent predictors of OS and RFS, respectively. Adverse events on BFM-95 protocol included SVT (4.9%), peripheral neuropathy (7.8%), myopathy (20.4%), hyperglycemia (24.3%), intestinal obstruction (7.8%), avascular necrosis of femur (6.8%), and mucositis (46%). CONCLUSION BFM-95 protocol appears to be a safe and effective strategy in adolescent and young adults and adult Nepalese population with ALL with a low toxicity profile.
Collapse
Affiliation(s)
- Bishesh Sharma Poudyal
- Clinical Hematology and Bone Marrow Transplant Unit, Civil Service Hospital, Kathmandu, Nepal
| | - Bishal Paudel
- Clinical Hematology and Bone Marrow Transplant Unit, Civil Service Hospital, Kathmandu, Nepal
| | | | - Samir Neupane
- Department of Pathology, Civil Service Hospital, Kathmandu, Nepal
| | - Kushal Bhattarai
- Department of Biochemistry, Karnali Academy of Health Sciences, Jumla, Nepal
| | - Utsav Joshi
- Department of Internal Medicine, Rochester General Hospital, New York, NY
| |
Collapse
|
11
|
Bordbar M, Jam N, Karimi M, Shahriari M, Zareifar S, Zekavat OR, Haghpanah S, Mottaghipisheh H. The survival of childhood leukemia: An 8-year single-center experience. Cancer Rep (Hoboken) 2023; 6:e1784. [PMID: 36700480 PMCID: PMC10075287 DOI: 10.1002/cnr2.1784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 01/04/2023] [Accepted: 01/11/2023] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND The survival of childhood leukemia has improved. We aimed to report the survival rate and the associated factors in children with acute leukemia during an 8-year follow-up. AIMS This study investigates the 8-year survival rates of children with acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) in Shiraz, the largest oncology center in Southern Iran. We also aimed to assess the independent factors associated with higher mortality in childhood leukemia. METHODS Children 0-18 years with acute leukemia were followed from 2013 to 2021 in Shiraz, Iran. The 8-year overall survival (OS) and event-free survival (EFS) rates were estimated by the Kaplan-Meier method. Independent factors associated with survival were assessed by the Cox regression hazard modeling. RESULTS We included 786 children, with 43.5% female, and a mean age of 6.32 ± 4.62 years. Patients with AML compared to ALL experienced more relapse (34.6% vs. 22.5%, p = .01) and death (31.7% vs. 11.3%, p < .001). The cumulative 8-year OS and EFS were 81% (95% confidence interval (CI), 74.3% to 86.1%) and 68.3% (95% CI, 63.5% to 72.7%) in ALL patients and 63.5% (95% CI, 52.1% to 72.9%) and 43% (95% CI, 33.1% to 52.6%) in AML patients. Multivariable analysis revealed that hepatomegaly (hazard ratio = 4, 95% CI, 1.0 to 22.3, p = .05) was the main independent risk factor of death in ALL patients. No definite risk factor was defined for AML patients. CONCLUSION The survival of childhood leukemia has recently increased dramatically in low-middle income countries. Hepatomegaly was introduced as a potential risk factor for lower survival in ALL patients. Further multicenter studies are needed to confirm the validity of this association.
Collapse
Affiliation(s)
| | - Nazila Jam
- Pediatrics Department, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehran Karimi
- Hematology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahdi Shahriari
- Hematology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soheila Zareifar
- Hematology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Omid Reza Zekavat
- Hematology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sezaneh Haghpanah
- Hematology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hadi Mottaghipisheh
- Hematology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
12
|
Bovine Colostrum Treatment of Specific Cancer Types: Current Evidence and Future Opportunities. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27248641. [PMID: 36557775 PMCID: PMC9785718 DOI: 10.3390/molecules27248641] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 11/29/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022]
Abstract
Worldwide, the incidence of cancer is on the rise. Current cancer treatments include chemotherapy, radiation therapy, and surgery. Chemotherapy and radiation treatment are typically associated with severe adverse effects and a decline in patients' quality of life. Anti-cancer substances derived from plants and animals need to be evaluated therapeutically as it is cost-effective, have fewer side effects, and can improve cancer patients' quality of life. Recently, bovine colostrum (BC) has attracted the interest of numerous researchers investigating its anti-cancer potential in humans. Dressings loaded with BC are beneficial in treating chronic wounds and diabetic foot ulcers. Lactoferrin, a glycoprotein with potent anti-oxidant, anti-inflammatory, anti-cancer, and anti-microbial effects, is abundant in BC. The BC pills successfully promote the regression of low-grade cervical intraepithelial neoplasia when administered intravaginally. The biological, genetic, and molecular mechanisms driving BC remain to be determined. Oral BC supplements are generally well-tolerated, but some flatulence and nausea may happen. To evaluate the therapeutic effects, long-term safety, and appropriate dosages of BC drugs, well-designed clinical trials are necessary. The purpose of this article is to emphasize the anti-cancer potential of BC and its constituents.
Collapse
|
13
|
Tibout P, Ledjiar O, Athale U, Rayar M, Kulkarni K, Truong T, Cellot S, Bittencourt H, Pelland-Marcotte MC, Tran TH. Prognostic factors and outcomes of infant acute lymphoblastic leukemia (ALL), hypodiploid ALL, and mixed-phenotype acute leukemia (MPAL) in Canada: a report from CYP-C. Leuk Lymphoma 2022; 63:3208-3216. [PMID: 36067507 DOI: 10.1080/10428194.2022.2118536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The epidemiology of infant acute lymphoblastic leukemia (ALL), hypodiploid ALL, and mixed-phenotype acute leukemia (MPAL) in Canada is unknown. The main objective was to describe the prevalence, prognostic factors, and outcomes of three rare and high-risk ALL subtypes in Canada. This is a retrospective study using the Cancer in Young People-Canada (CYP-C) database. Event-free survival (EFS) and overall survival (OS) were described by the Kaplan-Meier method and compared using the log-rank test. Among 2626 children aged 0-14 years diagnosed with B-cell acute lymphoblastic leukemia (B-ALL) between 2001 and 2018, 227 (8.6%) patients were identified to be infant ALL (n = 139), hypodiploid ALL (n = 43), or MPAL (n = 45). The 5-year EFS/OS was significantly worse in the infant ALL subgroup compared to that of hypodiploid ALL and MPAL. For the entire cohort, presenting White blood cells (WBCs) ≥50 × 109/L was independently associated with worse EFS/OS.
Collapse
Affiliation(s)
- Pauline Tibout
- Division of Pediatric Hematology-Oncology, Charles-Bruneau Cancer Center, CHU Sainte-Justine, University of Montreal, Montreal, Canada
| | - Omar Ledjiar
- Unité de recherche clinique appliquée, Research Centre, CHU Sainte-Justine, Montreal, Canada
| | - Uma Athale
- Division of Pediatric Hematology-Oncology, McMaster Children's Hospital, Hamilton, Canada
| | - Meera Rayar
- Division of Pediatric Hematology-Oncology, B.C. Children's Hospital, Vancouver, Canada
| | - Ketan Kulkarni
- Division of Pediatric Hematology-Oncology, IWK Health Centre and Dalhousie University, Halifax, Canada
| | - Tony Truong
- Division of Pediatric Hematology-Oncology, Alberta Children's Hospital, Calgary, Canada
| | - Sonia Cellot
- Division of Pediatric Hematology-Oncology, Charles-Bruneau Cancer Center, CHU Sainte-Justine, University of Montreal, Montreal, Canada
| | - Henrique Bittencourt
- Division of Pediatric Hematology-Oncology, Charles-Bruneau Cancer Center, CHU Sainte-Justine, University of Montreal, Montreal, Canada
| | | | - Thai Hoa Tran
- Division of Pediatric Hematology-Oncology, Charles-Bruneau Cancer Center, CHU Sainte-Justine, University of Montreal, Montreal, Canada
| |
Collapse
|
14
|
Flores-Lujano J, Duarte-Rodríguez DA, Jiménez-Hernández E, Martín-Trejo JA, Allende-López A, Peñaloza-González JG, Pérez-Saldivar ML, Medina-Sanson A, Torres-Nava JR, Solís-Labastida KA, Flores-Villegas LV, Espinosa-Elizondo RM, Amador-Sánchez R, Velázquez-Aviña MM, Merino-Pasaye LE, Núñez-Villegas NN, González-Ávila AI, del Campo-Martínez MDLÁ, Alvarado-Ibarra M, Bekker-Méndez VC, Cárdenas-Cardos R, Jiménez-Morales S, Rivera-Luna R, Rosas-Vargas H, López-Santiago NC, Rangel-López A, Hidalgo-Miranda A, Vega E, Mata-Rocha M, Sepúlveda-Robles OA, Arellano-Galindo J, Núñez-Enríquez JC, Mejía-Aranguré JM. Persistently high incidence rates of childhood acute leukemias from 2010 to 2017 in Mexico City: A population study from the MIGICCL. Front Public Health 2022; 10:918921. [PMID: 36187646 PMCID: PMC9518605 DOI: 10.3389/fpubh.2022.918921] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 08/09/2022] [Indexed: 01/22/2023] Open
Abstract
Introduction Over the years, the Hispanic population living in the United States has consistently shown high incidence rates of childhood acute leukemias (AL). Similarly, high AL incidence was previously observed in Mexico City (MC). Here, we estimated the AL incidence rates among children under 15 years of age in MC during the period 2010-2017. Methods The Mexican Interinstitutional Group for the Identification of the Causes of Childhood Leukemia conducted a study gathering clinical and epidemiological information regarding children newly diagnosed with AL at public health institutions of MC. Crude age incidence rates (cAIR) were obtained. Age-standardized incidence rates worldwide (ASIRw) and by municipalities (ASIRm) were calculated by the direct and indirect methods, respectively. These were reported per million population <15 years of age; stratified by age group, sex, AL subtypes, immunophenotype and gene rearrangements. Results A total of 903 AL cases were registered. The ASIRw was 63.3 (cases per million) for AL, 53.1 for acute lymphoblastic leukemia (ALL), and 9.4 for acute myeloblastic leukemia. The highest cAIR for AL was observed in the age group between 1 and 4 years (male: 102.34 and female: 82.73). By immunophenotype, the ASIRw was 47.3 for B-cell and 3.7 for T-cell. The incidence did not show any significant trends during the study period. The ASIRm for ALL were 68.6, 66.6 and 62.8 at Iztacalco, Venustiano Carranza and Benito Juárez, respectively, whereas, other municipalities exhibited null values mainly for AML. Conclusion The ASIRw for childhood AL in MC is among the highest reported worldwide. We observed spatial heterogeneity of rates by municipalities. The elevated AL incidence observed in Mexican children may be explained by a combination of genetic background and exposure to environmental risk factors.
Collapse
Affiliation(s)
- Janet Flores-Lujano
- Unidad de Investigación Médica en Epidemiología Clínica, Unidad Médica de Alta Especialidad, Hospital de Pediatría “Dr. Silvestre Frenk Freund, ” Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - David Aldebarán Duarte-Rodríguez
- Unidad de Investigación Médica en Epidemiología Clínica, Unidad Médica de Alta Especialidad, Hospital de Pediatría “Dr. Silvestre Frenk Freund, ” Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Elva Jiménez-Hernández
- Servicio de Hematología Pediátrica, Centro Médico Nacional “La Raza, ” Hospital General “Gaudencio González Garza, ” Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico,Servicio de Oncología, Hospital Pediátrico de Moctezuma, Secretaría de Salud de la Ciudad de México (SSCDMX), Mexico City, Mexico
| | - Jorge Alfonso Martín-Trejo
- Servicio de Hematología Pediátrica, Unidad Médica de Alta Especialidad, Hospital de Pediatría “Dr. Silvestre Frenk Freund, ” Centro Médico Nacional “Siglo XXI, ” Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Aldo Allende-López
- Unidad de Investigación Médica en Epidemiología Clínica, Unidad Médica de Alta Especialidad, Hospital de Pediatría “Dr. Silvestre Frenk Freund, ” Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | | | - María Luisa Pérez-Saldivar
- Unidad de Investigación Médica en Epidemiología Clínica, Unidad Médica de Alta Especialidad, Hospital de Pediatría “Dr. Silvestre Frenk Freund, ” Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Aurora Medina-Sanson
- Departamento de HematoOncología, Hospital Infantil de México Federico Gómez, Secretaría de Salud (SS), Mexico City, Mexico
| | - José Refugio Torres-Nava
- Servicio de Oncología, Hospital Pediátrico de Moctezuma, Secretaría de Salud de la Ciudad de México (SSCDMX), Mexico City, Mexico
| | - Karina Anastacia Solís-Labastida
- Servicio de Hematología Pediátrica, Unidad Médica de Alta Especialidad, Hospital de Pediatría “Dr. Silvestre Frenk Freund, ” Centro Médico Nacional “Siglo XXI, ” Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Luz Victoria Flores-Villegas
- Servicio de Hematología Pediátrica, Centro Médico Nacional “20 de Noviembre, ” Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado (ISSSTE), Mexico City, Mexico
| | | | - Raquel Amador-Sánchez
- Servicio de Hematología Pediátrica, Hospital General Regional 1 “Dr. Carlos McGregor Sánchez Navarro, ” Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | | | - Laura Elizabeth Merino-Pasaye
- Servicio de Hematología Pediátrica, Centro Médico Nacional “20 de Noviembre, ” Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado (ISSSTE), Mexico City, Mexico
| | - Nora Nancy Núñez-Villegas
- Servicio de Hematología Pediátrica, Centro Médico Nacional “La Raza, ” Hospital General “Gaudencio González Garza, ” Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Ana Itamar González-Ávila
- Servicio de Hematología Pediátrica, Hospital General Regional 1 “Dr. Carlos McGregor Sánchez Navarro, ” Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - María de los Ángeles del Campo-Martínez
- Servicio de Hematología Pediátrica, Centro Médico Nacional “La Raza, ” Hospital General “Gaudencio González Garza, ” Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Martha Alvarado-Ibarra
- Servicio de Hematología Pediátrica, Centro Médico Nacional “20 de Noviembre, ” Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado (ISSSTE), Mexico City, Mexico
| | - Vilma Carolina Bekker-Méndez
- Hospital de Infectología “Dr. Daniel Méndez Hernández, ” “La Raza, ” Instituto Mexicano del Seguro Social (IMSS), Unidad de Investigación Médica en Inmunología e Infectología, Mexico City, Mexico
| | - Rocío Cárdenas-Cardos
- Servicio de Oncología Pediátrica, Instituto Nacional de Pediatría, Secretaría de Salud (SS), Mexico City, Mexico
| | - Silvia Jiménez-Morales
- Laboratorio de Genómica del Cáncer, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Roberto Rivera-Luna
- Servicio de Oncología Pediátrica, Instituto Nacional de Pediatría, Secretaría de Salud (SS), Mexico City, Mexico
| | - Haydee Rosas-Vargas
- Unidad de Investigación Médica en Genética Humana, Unidad Médica de Alta Especialidad, Hospital de Pediatría “Dr. Silvestre Frenk Freund, ” Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Norma C. López-Santiago
- Servicio de Hematología Pediátrica, Instituto Nacional de Pediatría, Secretaría de Salud (SS), Mexico City, Mexico
| | - Angélica Rangel-López
- Coordinación de Investigación en Salud, Unidad Habilitada de Apoyo al Predictamen, Centro Médico Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Alfredo Hidalgo-Miranda
- Laboratorio de Genómica del Cáncer, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Elizabeth Vega
- Instituto de Ciencias de la Atmósfera y Cambio Climático, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Minerva Mata-Rocha
- Unidad de Investigación Médica en Genética Humana, Unidad Médica de Alta Especialidad, Hospital de Pediatría “Dr. Silvestre Frenk Freund, ” Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Omar Alejandro Sepúlveda-Robles
- Unidad de Investigación Médica en Genética Humana, Unidad Médica de Alta Especialidad, Hospital de Pediatría “Dr. Silvestre Frenk Freund, ” Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - José Arellano-Galindo
- Unidad de Investigación en Enfermedades Infecciosas, Laboratorio de Virología Clínica y Experimental, Hospital Infantil de México Federico Gómez, Secretaría de Salud (SS), Mexico City, Mexico
| | - Juan Carlos Núñez-Enríquez
- Unidad de Investigación Médica en Epidemiología Clínica, Unidad Médica de Alta Especialidad, Hospital de Pediatría “Dr. Silvestre Frenk Freund, ” Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico,Juan Carlos Núñez-Enríquez
| | - Juan Manuel Mejía-Aranguré
- Laboratorio de Genómica del Cáncer, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico,Unidad de Investigación Médica en Genética Humana, Unidad Médica de Alta Especialidad, Hospital de Pediatría “Dr. Silvestre Frenk Freund, ” Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico,Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico,*Correspondence: Juan Manuel Mejía-Aranguré
| |
Collapse
|
15
|
Impact of testicular boost in children with leukemia receiving total body irradiation and stem cell transplantation: a single-institution experience. Adv Radiat Oncol 2022; 8:101071. [DOI: 10.1016/j.adro.2022.101071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 09/04/2022] [Indexed: 11/21/2022] Open
|
16
|
Rahim MQ, Jones SR, Patel RB, Rupenthal J, Althouse SK, Vik T, Batra S. Early Discharge of Adolescent and Young Adult Patients During Induction Chemotherapy for Newly Diagnosed Acute Lymphoblastic Leukemia: Is It Safe? J Adolesc Young Adult Oncol 2022; 12:271-274. [PMID: 35852828 DOI: 10.1089/jayao.2022.0028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
There is a lack of consensus for safely discharging adolescent and young adults (AYA) with newly diagnosed acute lymphoblastic leukemia. From 2017 to 2019 we evaluated predefined early discharge criteria for 41 AYA patients during induction chemotherapy. Only 17% (7/41) of patients met criteria for early discharge. Two (29%) patients who were discharged early were readmitted, but not to the pediatric intensive care unit (PICU). This outcome was compared to a historic cohort at our institution of 73 patients who were discharged without predefined discharge criteria. Twenty-seven (37%, p = 0.7) patients were readmitted, but 13 (48%) were readmitted to the PICU (p = 0.004).
Collapse
Affiliation(s)
- Mahvish Q. Rahim
- Department of Pediatrics, Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Section of Pediatric Hematology-Oncology, Department of Pediatrics, Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sandra R. Jones
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Roshni B. Patel
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Joy Rupenthal
- Section of Pediatric Hematology-Oncology, Department of Pediatrics, Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sandra K. Althouse
- Department of Biostatistics and Data Health Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Terry Vik
- Department of Pediatrics, Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Section of Pediatric Hematology-Oncology, Department of Pediatrics, Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sandeep Batra
- Department of Pediatrics, Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Section of Pediatric Hematology-Oncology, Department of Pediatrics, Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Indiana University Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
17
|
St. Jude Total Therapy studies from I to XVII for childhood acute lymphoblastic leukemia: a brief review. J Egypt Natl Canc Inst 2022; 34:25. [PMID: 35696003 DOI: 10.1186/s43046-022-00126-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 05/13/2022] [Indexed: 11/10/2022] Open
Abstract
The therapy design of childhood acute lymphoblastic leukemia (ALL) has evolved over the past 60 years. The St. Jude Children's Research Hospital has developed 17 treatment protocols from 1962 to 2017, aiming to have the most effective and least toxic treatment form. This review summarizes each protocol's objectives, inclusion criteria, treatment phases, pharmacological agents, irradiation therapy, response criteria, risk stratification, type of relapse, and overall survival. The enhancement and successful application of preventive therapy for ALL and following a risk-stratified approach have progressively improved the cure rate of childhood ALL, with relatively few adverse sequelae. Moreover, St. Jude's scientific theme serves as a reminder of the principal factor of research directed to a catastrophic disease such as ALL.
Collapse
|
18
|
Li R, Tang JH, Zhang BB, Shi XY, Dai YY, Qu R. Clinical Analysis of Childhood Acute Lymphoblastic Leukemia With Epilepsy Seizures. Front Neurol 2022; 13:824268. [PMID: 35620787 PMCID: PMC9127044 DOI: 10.3389/fneur.2022.824268] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
Objective In order to analyze the clinical characteristics of epileptic seizures in children with acute lymphoblastic leukemia (ALL) during treatment. Methods The clinical and imaging data of children diagnosed as ALL with epilepsy seizures from January 2013 to December 2020 were retrospectively analyzed. Results A total of 2217 children with ALL were admitted during the study, of whom 229 (10.33%) had epileptic seizures after ALL treatment. Among them, 45 (19.65%) were in the high-risk group and 184 (80.35%) were in the low-risk group. Epileptic seizures mainly occurred in the induction remission period (24.02%), maintenance treatment period (25.33%) and after bone marrow transplantation (21.40%). The common causes were MTX-related demyelinating encephalopathy (34.06%) and reversible posterior encephalopathy syndrome (PRES) (25.3%). The first symptom was mainly convulsion (34.50%). The first attack had a comprehensive attack and partial attack. Most patients stop themselves. 30 cases (13.10%) had acute recurrence of epilepsy (recurrence within 3 months after the first attack), and 49 cases (25.76%) had neurological dysfunction after follow-up. 36 cases developed symptomatic epilepsy. Among the 130 children who completed the follow-up, 78 (60.00%) had no obvious neurological sequelae, and 52 (40.0%) had neurological sequelae. Among the 52 cases, there were 34 cases of mild sequelae and 18 cases of severe sequelae, including 8 cases of epilepsy combined with cognitive impairment. Conclusion Epileptic seizure is a common neurological complication during ALL treatment. The etiology and associated manifestations of the first epileptic seizure are diverse. Early neuroimaging and EEG examination are helpful for early diagnosis and treatment.
Collapse
Affiliation(s)
- Rui Li
- Department of Neurology, Children's Hospital of Soochow University, Suzhou, China
- Department of Pediatric, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Ji-Hong Tang
- Department of Neurology, Children's Hospital of Soochow University, Suzhou, China
| | - Bing-Bing Zhang
- Department of Neurology, Children's Hospital of Soochow University, Suzhou, China
| | - Xiao-Yan Shi
- Department of Neurology, Children's Hospital of Soochow University, Suzhou, China
| | - Yuan-Yuan Dai
- Department of Pediatric, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Rui Qu
- Department of Pediatric, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
19
|
Zheng S, Gillespie E, Naqvi AS, Hayer KE, Ang Z, Torres-Diz M, Quesnel-Vallières M, Hottman DA, Bagashev A, Chukinas J, Schmidt C, Asnani M, Shraim R, Taylor DM, Rheingold SR, O'Brien MM, Singh N, Lynch KW, Ruella M, Barash Y, Tasian SK, Thomas-Tikhonenko A. Modulation of CD22 Protein Expression in Childhood Leukemia by Pervasive Splicing Aberrations: Implications for CD22-Directed Immunotherapies. Blood Cancer Discov 2022; 3:103-115. [PMID: 35015683 PMCID: PMC9780083 DOI: 10.1158/2643-3230.bcd-21-0087] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/30/2021] [Accepted: 11/10/2021] [Indexed: 11/16/2022] Open
Abstract
Downregulation of surface epitopes causes postimmunotherapy relapses in B-lymphoblastic leukemia (B-ALL). Here we demonstrate that mRNA encoding CD22 undergoes aberrant splicing in B-ALL. We describe the plasma membrane-bound CD22 Δex5-6 splice isoform, which is resistant to chimeric antigen receptor (CAR) T cells targeting the third immunoglobulin-like domain of CD22. We also describe splice variants skipping the AUG-containing exon 2 and failing to produce any identifiable protein, thereby defining an event that is rate limiting for epitope presentation. Indeed, forcing exon 2 skipping with morpholino oligonucleotides reduced CD22 protein expression and conferred resistance to the CD22-directed antibody-drug conjugate inotuzumab ozogamicin in vitro. Furthermore, among inotuzumab-treated pediatric patients with B-ALL, we identified one nonresponder in whose leukemic blasts Δex2 isoforms comprised the majority of CD22 transcripts. In a second patient, a sharp reduction in CD22 protein levels during relapse was driven entirely by increased CD22 exon 2 skipping. Thus, dysregulated CD22 splicing is a major mechanism of epitope downregulation and ensuing resistance to immunotherapy. SIGNIFICANCE The mechanism(s) underlying downregulation of surface CD22 following CD22-directed immunotherapy remains underexplored. Our biochemical and correlative studies demonstrate that in B-ALL, CD22 expression levels are controlled by inclusion/skipping of CD22 exon 2. Thus, aberrant splicing of CD22 is an important driver/biomarker of de novo and acquired resistance to CD22-directed immunotherapies. See related commentary by Bourcier and Abdel-Wahab, p. 87. This article is highlighted in the In This Issue feature, p. 85.
Collapse
Affiliation(s)
- Sisi Zheng
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia,
Philadelphia, Pennsylvania
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia,
Pennsylvania
| | - Elisabeth Gillespie
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia,
Philadelphia, Pennsylvania
| | - Ammar S. Naqvi
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia,
Philadelphia, Pennsylvania
- Department of Biomedical and Health Informatics, Children's Hospital of
Philadelphia, Philadelphia, Pennsylvania
| | - Katharina E. Hayer
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia,
Philadelphia, Pennsylvania
- Department of Biomedical and Health Informatics, Children's Hospital of
Philadelphia, Philadelphia, Pennsylvania
| | - Zhiwei Ang
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia,
Philadelphia, Pennsylvania
| | - Manuel Torres-Diz
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia,
Philadelphia, Pennsylvania
| | - Mathieu Quesnel-Vallières
- Department of Genetics, Perelman School of Medicine at the University of
Pennsylvania, Philadelphia, Pennsylvania
- Department of Biochemistry and Biophysics, Perelman School of Medicine at
the University of Pennsylvania, Philadelphia, Pennsylvania
| | - David A. Hottman
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia,
Pennsylvania
| | - Asen Bagashev
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia,
Philadelphia, Pennsylvania
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia,
Pennsylvania
| | - John Chukinas
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia,
Pennsylvania
| | - Carolin Schmidt
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia,
Philadelphia, Pennsylvania
| | - Mukta Asnani
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia,
Philadelphia, Pennsylvania
| | - Rawan Shraim
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia,
Philadelphia, Pennsylvania
- Department of Biomedical and Health Informatics, Children's Hospital of
Philadelphia, Philadelphia, Pennsylvania
| | - Deanne M. Taylor
- Department of Biomedical and Health Informatics, Children's Hospital of
Philadelphia, Philadelphia, Pennsylvania
| | - Susan R. Rheingold
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia,
Pennsylvania
- Department of Pediatrics, Perelman School of Medicine at the University of
Pennsylvania, Philadelphia, Pennsylvania
| | - Maureen M. O'Brien
- Cincinnati Children's Hospital Medical Center, University of Cincinnati
College of Medicine, Cincinnati, Ohio
| | - Nathan Singh
- Department of Medicine, Perelman School of Medicine at the University of
Pennsylvania, Philadelphia, Pennsylvania
| | - Kristen W. Lynch
- Department of Biochemistry and Biophysics, Perelman School of Medicine at
the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Marco Ruella
- Department of Medicine, Perelman School of Medicine at the University of
Pennsylvania, Philadelphia, Pennsylvania
| | - Yoseph Barash
- Department of Genetics, Perelman School of Medicine at the University of
Pennsylvania, Philadelphia, Pennsylvania
| | - Sarah K. Tasian
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia,
Pennsylvania
- Department of Pediatrics, Perelman School of Medicine at the University of
Pennsylvania, Philadelphia, Pennsylvania
| | - Andrei Thomas-Tikhonenko
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia,
Philadelphia, Pennsylvania
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia,
Pennsylvania
- Department of Pediatrics, Perelman School of Medicine at the University of
Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine
at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
20
|
HLA-haploidentical peripheral blood stem cell transplantation following reduced-intensity conditioning with very low-dose antithymocyte globulin for relapsed/refractory acute leukemia in pediatric patients: a single-institution retrospective analysis. Int J Hematol 2022; 115:406-413. [PMID: 35028882 DOI: 10.1007/s12185-021-03270-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 10/19/2022]
Abstract
The prognosis of relapsed/refractory (R/R) pediatric acute leukemia is extremely poor. We retrospectively reviewed 20 consecutive pediatric patients with R/R acute leukemia who underwent a first HLA-haploidentical peripheral blood stem cell transplantation following reduced-intensity conditioning (haplo-RIC-PBSCT) with very low-dose antithymocyte globulin (ATG) between 2012 and 2019. Of these 20 patients, 7 patients had acute lymphoblastic leukemia, and 13 had acute myeloid leukemia. At the time of haplo-RIC-PBSCT, 15 patients had active disease. The median follow-up duration for survivors was 56 months (range 22-108 months). Graft-versus-host disease (GVHD) prophylaxis consisted of tacrolimus, short-term methotrexate, methylprednisolone, and ATG 1.25 mg/kg on day-2. The 2-year cumulative incidence of transplant-related mortality and relapse were 5.0% [95% confidence interval (CI) 0.7-30.5%)] and 57.8% (95% CI 37.4-79.6%), respectively. Among the 20 patients, 16 (80.0%) developed grade III-IV acute GVHD, and 2 developed severe chronic GVHD. The 2-year event-free survival and overall survival rates were 40.0% (95% CI 19.3-60.0%) and 50.0% (95% CI 27.1-69.2%), respectively. Although the sample size is small, the survival outcomes of the present study are encouraging.
Collapse
|
21
|
Burke MJ, Devidas M, Chen Z, Salzer WL, Raetz EA, Rabin KR, Heerema NA, Carroll AJ, Gastier-Foster JM, Borowitz MJ, Wood BL, Winick NJ, Carroll WL, Hunger SP, Loh ML, Larsen EC. Outcomes in adolescent and young adult patients (16 to 30 years) compared to younger patients treated for high-risk B-lymphoblastic leukemia: report from Children's Oncology Group Study AALL0232. Leukemia 2021; 36:648-655. [PMID: 34725453 DOI: 10.1038/s41375-021-01460-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 10/14/2021] [Accepted: 10/20/2021] [Indexed: 12/31/2022]
Abstract
Adolescent and young adult (AYA) patients 16-30 years old with high-risk acute lymphoblastic leukemia (HR-ALL) have inferior outcomes compared to younger HR-ALL patients. AALL0232 was a Phase 3 randomized Children's Oncology Group trial for newly diagnosed HR B-ALL (1-30 years). Between 2004 and 2011, 3154 patients enrolled with 3040 eligible and evaluable for induction. AYA patients comprised 20% of patients (16-21 years, n = 551; 22-30 years, n = 46). 5-year event-free survival and overall survival was 65.4 ± 2.2% and 77.4 ± 2.0% for AYA patients compared to 78.1 ± 0.9% and 87.3 ± 0.7% for younger patients (p < 0.0001). Five-year cumulative incidence of relapse was 18.5 ± 1.7% for AYA patients and 13.5 ± 0.7% for younger patients (p = 0.006), largely due to increased marrow relapses (14.0 ± 1.5% versus 9.1 ± 0.6%; p < 0.0001). Additionally, induction failure rate was higher in AYA (7.2 ± 1.1% versus 3.5 ± 0.4%; p < 0.001) and post-induction remission deaths were significantly higher in AYA (5.7 ± 1.0% versus 2.4 ± 0.3%; p < 0.0001). AALL0232 enrolled the largest number of AYA B-ALL patients to date, demonstrating significantly inferior survival and greater rates of treatment-related toxicities compared to younger patients. Although treatment intensification has improved outcomes in younger patients, they have not been associated with the same degree of improvement for older patients.
Collapse
Affiliation(s)
- Michael J Burke
- Department of Pediatrics, Children's Hospital of Wisconsin, Milwaukee, WI, USA.
| | - Meenakshi Devidas
- Department of Global Pediatric Medicine, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Zhiguo Chen
- Biostatistics, Colleges of Medicine and Public Health & Health Professions, University of Florida, Gainesville, FL, USA
| | - Wanda L Salzer
- U.S. Army Medical Research and Materiel Command, Fort Detrick, MD, USA
| | - Elizabeth A Raetz
- Department of Pediatrics, Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY, USA
| | - Karen R Rabin
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Nyla A Heerema
- Department of Pathology, The Ohio State University School of Medicine, Columbus, OH, USA
| | - Andrew J Carroll
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Michael J Borowitz
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Brent L Wood
- Department of Laboratory Medicine, University of Washington, Seattle, WA, USA
| | - Naomi J Winick
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - William L Carroll
- Department of Pediatrics, Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY, USA
| | - Stephen P Hunger
- Department of Pediatrics, Children's Hospital of Philadelphia and the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Mignon L Loh
- Department of Pediatrics, Benioff Children's Hospital and the Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, CA, USA
| | - Eric C Larsen
- Department of Pediatrics, Maine Children's Cancer Program, Scarborough, ME, USA
| |
Collapse
|
22
|
Dixon SB, Chen Y, Yasui Y, Pui CH, Hunger SP, Silverman LB, Ness KK, Green DM, Howell RM, Leisenring WM, Kadan-Lottick NS, Krull KR, Oeffinger KC, Neglia JP, Hudson MM, Robison LL, Mertens AC, Armstrong GT, Nathan PC. Impact of Risk-Stratified Therapy on Health Status in Survivors of Childhood Acute Lymphoblastic Leukemia: A Report from the Childhood Cancer Survivor Study. Cancer Epidemiol Biomarkers Prev 2021; 31:150-160. [PMID: 34697055 DOI: 10.1158/1055-9965.epi-21-0667] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/05/2021] [Accepted: 10/20/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Prior studies have identified that survivors of childhood acute lymphoblastic leukemia (ALL) report poor health status. It is unknown how risk-stratified therapy impacts the health status of ALL survivors. METHODS We estimated and compared the prevalence of self-reported poor health status among adult (≥18 years) survivors of childhood ALL diagnosed at age <21 years from 1970 to 1999 and sibling controls, excluding proxy reports. Therapy combinations defined treatment groups representative of 1970s therapy (70s), standard- and high-risk 1980s and 1990s therapy (80sSR, 80sHR, 90sSR, 90sHR), and relapse/bone marrow transplant (R/BMT). Log-binomial models, adjusted for clinical and demographic factors, compared outcomes between groups using prevalence ratios (PR) with 95% confidence intervals (CI). RESULTS Among 5,119 survivors and 4,693 siblings, survivors were more likely to report poor health status in each domain including poor general health (13.5% vs. 7.4%; PR = 1.92; 95% CI, 1.69-2.19). Compared with 70s, 90sSR and 90sHR were less likely to report poor general health (90sSR: PR = 0.75; 95% CI, 0.57-0.98; 90sHR: PR = 0.58; 95% CI, 0.39-0.87), functional impairment (90sSR: PR = 0.56; 95% CI, 0.42-0.76; 90sHR: PR = 0.63; 95% CI, 0.42-0.95), and activity limitations (90sSR: 0.61; 95% CI, 0.45-0.83; 90sHR: PR = 0.59; 95% CI, 0.38-0.91). An added adjustment for chronic conditions in multivariable models partially attenuated 90sSR risk estimates. CONCLUSIONS Risk-stratified ALL therapy has succeeded in reducing risk for poor general health, functional impairment, and activity limitations among more recent survivors of standard- and high-risk therapy. IMPACT Future research into the relationship between risk-stratified therapy, health status, and late health outcomes may provide new opportunities to further improve late morbidity among survivors.
Collapse
Affiliation(s)
- Stephanie B Dixon
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee.
| | - Yan Chen
- School of Public Health, University of Alberta, Edmonton, Alberta, Canada
| | - Yutaka Yasui
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Ching-Hon Pui
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Stephen P Hunger
- Division of Oncology and the Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Lewis B Silverman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kirsten K Ness
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Daniel M Green
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee.,Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Rebecca M Howell
- Radiation Physics Department, The University of Texas at MD Anderson Cancer Center, Houston, Texas
| | - Wendy M Leisenring
- Cancer Prevention and Clinical Statistics Programs, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Nina S Kadan-Lottick
- Section of Pediatric Hematology/Oncology at Yale School of Medicine and Yale Cancer Center, New Haven, Connecticut
| | - Kevin R Krull
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee.,Department of Psychology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | | | - Joseph P Neglia
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Melissa M Hudson
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee.,Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Leslie L Robison
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Ann C Mertens
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Gregory T Armstrong
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee.,Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Paul C Nathan
- Division of Hematology/Oncology, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
23
|
Mangum DS, Meyer JA, Mason CC, Shams S, Maese LD, Gardiner JD, Downie JM, Pei D, Cheng C, Gleason A, Luo M, Pui CH, Aplenc R, Hunger SP, Loh M, Greaves M, Trede N, Raetz E, Frazer JK, Mullighan CG, Engel ME, Miles RR, Rabin KR, Schiffman JD. Association of Combined Focal 22q11.22 Deletion and IKZF1 Alterations With Outcomes in Childhood Acute Lymphoblastic Leukemia. JAMA Oncol 2021; 7:1521-1528. [PMID: 34410295 DOI: 10.1001/jamaoncol.2021.2723] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Importance Alterations in the IKZF1 gene drive B-cell acute lymphoblastic leukemia (B-ALL) but are not routinely used to stratify patients by risk because of inconsistent associations with outcomes. We describe a novel deletion in 22q11.22 that was consistently associated with very poor outcomes in patients with B-ALL with IKZF1 alterations. Objective To determine whether focal deletions within the λ variable chain region in chromosome 22q11.22 were associated with patients with B-ALL with IKZF1 alterations with the highest risk of relapse and/or death. Design, Setting, and Participants This cohort study included 1310 primarily high-risk pediatric patients with B-ALL who were taken from 6 independent clinical cohorts, consisting of 3 multicenter cohorts (AALL0232 [2004-2011], P9906 [2000-2003], and patients with Down syndrome who were pooled from national and international studies) and 3 single-institution cohorts (University of Utah [Salt Lake City], Children's Hospital of Philadelphia [Philadelphia, Pennsylvania], and St. Jude Children's Hospital [Memphis, Tennessee]). Data analysis began in 2011 using patients from the older studies first, and data analysis concluded in 2021. Exposures Focal 22q11.22 deletions. Main Outcomes and Measures Event-free and overall survival was investigated. The hypothesis that 22q11.22 deletions stratified the prognostic effect of IKZF1 alterations was formulated while investigating nearby deletions in VPREB1 in 2 initial cohorts (n = 270). Four additional cohorts were then obtained to further study this association (n = 1040). Results This study of 1310 patients with B-ALL (717 male [56.1%] and 562 female patients [43.9%]) found that focal 22q11.22 deletions are frequent (518 of 1310 [39.5%]) in B-ALL and inconsistent with physiologic V(D)J recombination. A total of 299 of 1310 patients with B-ALL had IKZF1 alterations. Among patients with IKZF1 alterations, more than half shared concomitant focal 22q11.22 deletions (159 of 299 [53.0%]). Patients with combined IKZF1 alterations and 22q11.22 deletions had worse outcomes compared with patients with IKZF1 alterations and wild-type 22q11.22 alleles in every cohort examined (combined cohorts: 5-year event-free survival rates, 43.3% vs 68.5%; hazard ratio [HR], 2.18; 95% CI, 1.54-3.07; P < .001; 5-year overall survival rates, 66.9% vs 83.9%; HR, 2.05; 95% CI, 1.32-3.21; P = .001). While 22q11.22 deletions were not prognostic in patients with wild-type IKZF1 , concomitant 22q11.22 deletions in patients with IKZF1 alterations stratified outcomes across additional risk groups, including patients who met the IKZF1plus criteria, and maintained independent significance in multivariate analysis for event-free survival (HR, 2.05; 95% CI, 1.27-3.29; P = .003) and overall survival (HR, 1.83; 95% CI, 1.01-3.34; P = .05). Conclusions and Relevance This cohort study suggests that 22q11.22 deletions identify patients with B-ALL and IKZF1 alterations who have very poor outcomes and may offer a new genetic biomarker to further refine B-ALL risk stratification and treatment strategies.
Collapse
Affiliation(s)
- David Spencer Mangum
- Nemours/Alfred I. DuPont Hospital for Children, Division of Pediatric Hematology/Oncology, Wilmington, Delaware
| | - Julia A Meyer
- Division of Pediatric Hematology & Oncology, Department of Pediatrics, University of Utah, Salt Lake City.,Division of Pediatric Hematology and Oncology, University of California, San Francisco
| | - Clinton C Mason
- Division of Pediatric Hematology & Oncology, Department of Pediatrics, University of Utah, Salt Lake City
| | | | - Luke D Maese
- Division of Pediatric Hematology & Oncology, Department of Pediatrics, University of Utah, Salt Lake City
| | - Jamie D Gardiner
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City
| | | | - Deqing Pei
- Department of Biostatistics, St Jude Children's Research Hospital, Memphis, Tennessee
| | - Cheng Cheng
- Department of Biostatistics, St Jude Children's Research Hospital, Memphis, Tennessee
| | - Adam Gleason
- Department of Pathology & Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Minjie Luo
- Department of Pathology & Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Ching-Hon Pui
- Department of Oncology, St Jude Children's Research Hospital, Memphis, Tennessee
| | - Richard Aplenc
- Division of Oncology and the Center for Childhood Cancer Research, The Children's Hospital of Philadelphia and The Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Stephen P Hunger
- Division of Oncology and the Center for Childhood Cancer Research, The Children's Hospital of Philadelphia and The Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Mignon Loh
- Division of Pediatric Hematology and Oncology, University of California, San Francisco
| | - Mel Greaves
- Institute of Cancer Research, London, England
| | | | - Elizabeth Raetz
- Department of Pediatrics, NYU Langone Health, New York, New York
| | - J Kimble Frazer
- Jimmy Everest Section of Pediatric Hematology-Oncology, Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City
| | - Charles G Mullighan
- Department of Pathology, St Jude Children's Research Hospital, Memphis, Tennessee
| | - Michael E Engel
- Division of Pediatric Hematology Oncology, Department of Pediatrics, University of Virginia, Charlottesville
| | - Rodney R Miles
- Department of Pathology, University of Utah Health Sciences Center, Salt Lake City
| | - Karen R Rabin
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Joshua D Schiffman
- Division of Pediatric Hematology & Oncology, Department of Pediatrics, University of Utah, Salt Lake City.,Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City.,PEEL Therapeutics, Inc, Salt Lake City, Utah
| |
Collapse
|
24
|
Brown PA, Shah B, Advani A, Aoun P, Boyer MW, Burke PW, DeAngelo DJ, Dinner S, Fathi AT, Gauthier J, Jain N, Kirby S, Liedtke M, Litzow M, Logan A, Luger S, Maness LJ, Massaro S, Mattison RJ, May W, Oluwole O, Park J, Przespolewski A, Rangaraju S, Rubnitz JE, Uy GL, Vusirikala M, Wieduwilt M, Lynn B, Berardi RA, Freedman-Cass DA, Campbell M. Acute Lymphoblastic Leukemia, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2021; 19:1079-1109. [PMID: 34551384 DOI: 10.6004/jnccn.2021.0042] [Citation(s) in RCA: 139] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The NCCN Guidelines for Acute Lymphoblastic Leukemia (ALL) focus on the classification of ALL subtypes based on immunophenotype and cytogenetic/molecular markers; risk assessment and stratification for risk-adapted therapy; treatment strategies for Philadelphia chromosome (Ph)-positive and Ph-negative ALL for both adolescent and young adult and adult patients; and supportive care considerations. Given the complexity of ALL treatment regimens and the required supportive care measures, the NCCN ALL Panel recommends that patients be treated at a specialized cancer center with expertise in the management of ALL This portion of the Guidelines focuses on the management of Ph-positive and Ph-negative ALL in adolescents and young adults, and management in relapsed settings.
Collapse
Affiliation(s)
- Patrick A Brown
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | | | - Anjali Advani
- Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | | | | | | | | | - Shira Dinner
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | | | - Jordan Gauthier
- Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | - Nitin Jain
- The University of Texas MD Anderson Cancer Center
| | | | | | | | - Aaron Logan
- UCSF Helen Diller Family Comprehensive Cancer Center
| | - Selina Luger
- Abramson Cancer Center at the University of Pennsylvania
| | | | | | | | | | | | - Jae Park
- Memorial Sloan Kettering Cancer Center
| | | | | | - Jeffrey E Rubnitz
- St. Jude Children's Research Hospital/The University of Tennessee Health Science Center
| | - Geoffrey L Uy
- Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | | | | | - Beth Lynn
- National Comprehensive Cancer Network
| | | | | | | |
Collapse
|
25
|
Bernasconi DP, Antolini L, Rossi E, Blanco-Lopez JG, Galimberti S, Andersen PK, Valsecchi MG. A causal inference approach to compare leukaemia treatment outcome in the absence of randomization and with dependent censoring. Int J Epidemiol 2021; 51:314-323. [PMID: 34368848 DOI: 10.1093/ije/dyab150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 07/06/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND One cause of poor outcomes in children of low-income countries affected by acute lymphoblastic leukaemia (ALL) is loss to follow-up due to abandonment of treatment. Assuming this type of loss to follow-up as independent censoring, as in standard Kaplan-Meier estimates, ignores the likely association of abandonment with biologic and socio-economic factors related to outcome. Moreover, when comparing treatment protocols adopted in different time periods, possible imbalances in patients' characteristics must be considered. We aim to compare the outcome of children enrolled in two subsequent protocols for ALL treatment (2000-2007 and 2008-2015) in Honduras, taking both dependent censoring due to abandonment of treatment and imbalances between patient characteristics into account. METHODS Marginal structural models based on inverse probability of treatment and censoring (IPTC) weighting allow the estimation of potential event-free survival (EFS) as if no abandonment of treatment occurred and the whole cohort was exposed, or not, to both protocols. An Aalen additive model and a logistic-regression model were used to build abandonment and treatment weights, respectively. RESULTS The two protocols recruited 514 and 717 patients. Measured baseline covariates in both protocols were gender, age, white blood cell count, central nervous system involvement, tumour histology and socio-economic status. The potential EFS is slightly higher under the more recent protocol in the first 3 years but no difference is estimated in the long period [survival difference at 5 years (95% confidence interval) = 0.1% (-0.97%; 1.13%)]. Both protocols would allow reducing the event rate by 12-13% if there was no abandonment of treatment. CONCLUSIONS Using IPTC weighting, we found a similar potential effect of the two treatment protocols if the imbalance due to the different distribution of potential confounders and to abandonment of therapy was removed.
Collapse
Affiliation(s)
- Davide Paolo Bernasconi
- Bicocca Bioinformatics Biostatistics and Bioimaging Centre-B4, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Laura Antolini
- Bicocca Bioinformatics Biostatistics and Bioimaging Centre-B4, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Emanuela Rossi
- Bicocca Bioinformatics Biostatistics and Bioimaging Centre-B4, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | | | - Stefania Galimberti
- Bicocca Bioinformatics Biostatistics and Bioimaging Centre-B4, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Per Kragh Andersen
- Section of Biostatistics, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Maria Grazia Valsecchi
- Bicocca Bioinformatics Biostatistics and Bioimaging Centre-B4, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
26
|
Liu C, Huang B, Wu R, Chen J, Tang Y, Hu W, Li J, Chen X, Cai J, Zhou M, Chen C, Shen S. Adequate asparaginase is important to prevent central nervous system and testicular relapse of pediatric Philadelphia chromosome-negative B-cell acute lymphoblastic leukemia. Int J Cancer 2021; 149:158-168. [PMID: 33634856 DOI: 10.1002/ijc.33529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/13/2021] [Accepted: 02/16/2021] [Indexed: 11/11/2022]
Abstract
Asparaginase (Asp) is one of the most important drugs for treating acute lymphoblastic leukemia (ALL). However, off-protocol Asp administration (OPAA) or hypersensitivity may disturb its pharmacokinetic profile. In this retrospective study, we sought to determine whether OPAA and hypersensitivity to Escherichia coli asparaginase (E coli Asp) impaired extramedullary relapse prevention in a pediatric ALL cohort treated according to SCMC-ALL-2005 protocol from 2005 to 2014 at the Shanghai Children's Medical Center (SCMC). In total, 676 patients were enrolled in this study, including 369 with OPAA and 60 exhibiting hypersensitivity to E coli Asp. At the end of the most recent follow-up, 58 patients had extramedullary relapse. The 5-year cumulative extramedullary relapse incidence in patients with OPAA was 11.01%, whereas that in patients without OPAA was 5.28% (P = .0036). Moreover, the 5-year cumulative extramedullary relapse incidence in patients that exhibited hypersensitivity to E coli Asp was 16.48%, whereas that in patients without hypersensitivity was 7.59% (P = .0195). Concerning the relapse site, OPAA not only increased central nervous system (CNS) relapse but testicular relapse as well. Based on Fine and Gray multivariate analysis, OPAA and hypersensitivity to Asp were independent risk factors for extramedullary relapse. In conclusion, to prevent extramedullary relapse of ALL, adequate duration to administrate Asp was more important than the total dosage, and more attention should be paid to Asp inadequate due to hypersensitivity.
Collapse
Affiliation(s)
- Chenxi Liu
- Department of Hematology/Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Key Laboratory of Pediatric Hematology & Oncology of China Ministry of Health, and National Children's Medical Center, Shanghai, China
| | - Binxiao Huang
- Department of Pediatric, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruichi Wu
- Department of Hematology/Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Key Laboratory of Pediatric Hematology & Oncology of China Ministry of Health, and National Children's Medical Center, Shanghai, China
| | - Jing Chen
- Department of Hematology/Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Key Laboratory of Pediatric Hematology & Oncology of China Ministry of Health, and National Children's Medical Center, Shanghai, China
| | - Yanjing Tang
- Department of Hematology/Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Key Laboratory of Pediatric Hematology & Oncology of China Ministry of Health, and National Children's Medical Center, Shanghai, China
| | - Wenting Hu
- Department of Hematology/Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Key Laboratory of Pediatric Hematology & Oncology of China Ministry of Health, and National Children's Medical Center, Shanghai, China
| | - Jing Li
- Department of Hematology/Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Key Laboratory of Pediatric Hematology & Oncology of China Ministry of Health, and National Children's Medical Center, Shanghai, China
| | - Xiaoxiao Chen
- Department of Hematology/Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Key Laboratory of Pediatric Hematology & Oncology of China Ministry of Health, and National Children's Medical Center, Shanghai, China
| | - Jiaoyang Cai
- Department of Hematology/Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Key Laboratory of Pediatric Hematology & Oncology of China Ministry of Health, and National Children's Medical Center, Shanghai, China
| | - Min Zhou
- Department of Hematology/Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Key Laboratory of Pediatric Hematology & Oncology of China Ministry of Health, and National Children's Medical Center, Shanghai, China
| | - Changcheng Chen
- Department of Hematology/Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Key Laboratory of Pediatric Hematology & Oncology of China Ministry of Health, and National Children's Medical Center, Shanghai, China
| | - Shuhong Shen
- Department of Hematology/Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Key Laboratory of Pediatric Hematology & Oncology of China Ministry of Health, and National Children's Medical Center, Shanghai, China
| |
Collapse
|
27
|
Jasek-Gajda E, Jurkowska H, JasiŃska M, Litwin JA, Lis GJ. Combination of ERK2 and STAT3 Inhibitors Promotes Anticancer Effects on Acute Lymphoblastic Leukemia Cells. Cancer Genomics Proteomics 2021; 17:517-527. [PMID: 32859630 DOI: 10.21873/cgp.20208] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/01/2020] [Accepted: 06/10/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND/AIM Deregulated activation of signaling through the RAS/RAF/mitogen-activated protein kinase/extracellular signal-regulated kinase (RAS/RAF/MEK/ERK) and signal transducer and activator of transcription (STAT) pathways is involved in numerous hematological malignancies, making it an attractive therapeutic target. This study aimed to assess the effect of the combination of ERK2 inhibitor VX-11e and STAT3 inhibitor STA-21 on acute lymphoblastic leukemia cell lines REH and MOLT-4. MATERIALS AND METHODS REH and MOLT-4 cell lines were cultured with each drug alone and in combination. Cell viability, ERK activity, cell cycle distribution, apoptosis and oxidative stress induction were assessed by flow cytometry. Protein levels of STAT3, phospho-STAT3, protein tyrosine phosphatase 4A3 (PTP4A3), survivin, p53 and p21 were determined by western blotting. RESULTS VX-11e in combination with STA-21 significantly inhibited cell viability, induced G0/G1 cell-cycle arrest, enhanced production of reactive oxygen species, and induced apoptosis. These effects were associated with an increased level of p21 protein in REH cells and with reduced levels of phopho-STAT3, survivin and PTP4A3 proteins in MOLT-4 cells. CONCLUSION Our findings provide a rationale for combined inhibition of RAS/RAF/MEK/ERK and STAT3 pathways in order to enhance anticancer effects against acute lymphoblastic leukemia cells.
Collapse
Affiliation(s)
- Ewa Jasek-Gajda
- Department of Histology, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Halina Jurkowska
- Chair of Medical Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - MaŁgorzata JasiŃska
- Department of Histology, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Jan A Litwin
- Department of Histology, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Grzegorz J Lis
- Department of Histology, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| |
Collapse
|
28
|
Walters M, Mowbray C, Jubelirer T, Jacobs S, Kelly KM, Smith K, Yao Y, Jin Z, Ladas EJ. A bilingual dietary intervention early in treatment is feasible and prevents weight gain in childhood acute lymphoblastic leukemia. Pediatr Blood Cancer 2021; 68:e28910. [PMID: 33590674 DOI: 10.1002/pbc.28910] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 01/04/2021] [Accepted: 01/05/2021] [Indexed: 02/02/2023]
Abstract
BACKGROUND Childhood acute lymphoblastic leukemia (ALL) is the most common pediatric malignancy. The onset of obesity during childhood ALL has been well established and is associated with inferior survival rates and increased treatment-related toxicities. This pilot study sought to determine if a dietary intervention is feasible and minimizes weight gain during the initial phases of treatment for ALL. METHODS Participants were recruited from four institutions, fluent in English or Spanish, between 5 and 21 years old, and enrolled within 3 days of starting induction therapy. Participants were counseled for 6 months to follow a low glycemic diet. Dietary and anthropometric data were collected at diagnosis, end of induction, and end of month 6 (NCT03157323). RESULTS Twenty-three of 28 participants (82.1%) were evaluable and included in the analysis. Dietary changes targeted by the nutrition intervention were successful; sugar intake declined (P = .003), whereas vegetable intake increased (P = .033). The majority of participants were able to adhere to the dietary principles prescribed: ≥70.0% reduced glycemic load and ≥60.0% increased fiber intake and decreased sugar intake. Importantly, we did not observe an increase in body mass index z-score during induction or over the 6-month intervention period. Most families found the nutrition intervention easy to follow (60%) and affordable (95%) despite simultaneous initiation of treatment for ALL. CONCLUSIONS A 6-month nutrition intervention initiated during the initial phase of treatment for childhood ALL is feasible and may prevent weight gain. Our preliminary findings need to be confirmed in a larger clinical trial.
Collapse
Affiliation(s)
- Michelle Walters
- Division of Pediatric Hematology/Oncology/Stem Cell Transplant, Columbia University Irving Medical Center, New York, New York
| | - Catriona Mowbray
- Division of Oncology, Children's National Hospital, District of Columbia, Washington
| | - Tracey Jubelirer
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.,Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Shana Jacobs
- Division of Oncology, Children's National Hospital, District of Columbia, Washington
| | - Kara M Kelly
- Department of Pediatrics, Roswell Park Comprehensive Cancer Center and University at Buffalo Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York
| | - Karen Smith
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Yujing Yao
- Department of Biostatistics, Columbia University Irving Medical Center, New York, New York
| | - Zhezhen Jin
- Department of Biostatistics, Columbia University Irving Medical Center, New York, New York
| | - Elena J Ladas
- Division of Pediatric Hematology/Oncology/Stem Cell Transplant, Columbia University Irving Medical Center, New York, New York.,Institute of Human Nutrition, Columbia University, New York, New York.,Department of Epidemiology, Mailman School of Public Health, Columbia University Irving Medical Center, New York, New York
| |
Collapse
|
29
|
Orgel E, Framson C, Buxton R, Kim J, Li G, Tucci J, Freyer DR, Sun W, Oberley MJ, Dieli-Conwright C, Mittelman SD. Caloric and nutrient restriction to augment chemotherapy efficacy for acute lymphoblastic leukemia: the IDEAL trial. Blood Adv 2021; 5:1853-1861. [PMID: 33792627 PMCID: PMC8045487 DOI: 10.1182/bloodadvances.2020004018] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 01/18/2021] [Indexed: 02/07/2023] Open
Abstract
Being overweight or obese (OW/OB) during B-cell acute lymphoblastic leukemia (B-ALL) induction is associated with chemoresistance as quantified by minimal residual disease (MRD). We hypothesized that caloric and nutrient restriction from diet/exercise could lessen gains in fat mass (FM) and reduce postinduction MRD. The Improving Diet and Exercise in ALL (IDEAL) trial enrolled patients 10 to 21 years old, newly diagnosed with B-ALL (n = 40), in comparison with a recent historical control (n = 80). Designed to achieve caloric deficits ≥20% during induction, reduce fat intake/glycemic load, and increase activity, IDEAL's end points were FM gain (primary), MRD ≥0.01%, and adherence/feasibility. Integrated biology explored biomarkers of OW/OB physiology. IDEAL intervention did not significantly reduce median FM change from baseline overall (+5.1% [interquartile range [IQR], 15.8] vs +10.7% [IQR, 16.0]; P = .13), but stratified analysis showed benefit in those OW/OB (+1.5% [IQR, 6.6] vs +9.7% [IQR, 11.1]; P = .02). After accounting for prognostic factors, IDEAL intervention significantly reduced MRD risk (odds ratio, 0.30; 95% confidence interval, 0.09-0.92; P = .02). The trial exceeded its adherence (≥75% of overall diet) and feasibility (≥80% completed visits) thresholds. Integrated biology found the IDEAL intervention increased circulating adiponectin and reduced insulin resistance. The IDEAL intervention was feasible, decreased fat gain in those OW/OB, and reduced MRD. This is the first study in any hematologic malignancy to demonstrate potential benefit from caloric restriction via diet/exercise to augment chemotherapy efficacy and improve disease response. A prospective, randomized trial is warranted for validation. These trials were registered at www.clinicaltrials.gov as #NCT02708108 (IDEAL trial) and #NCT01317940 (historical control).
Collapse
Affiliation(s)
| | | | - Rubi Buxton
- Division of Pediatric Rehabilitation Medicine, Children's Hospital Los Angeles, Los Angeles, CA
| | - Jiyoon Kim
- Department of Biostatistics and Computational Medicine, Jonathan and Karin Fielding School of Public Health, and
| | - Gang Li
- Department of Biostatistics and Computational Medicine, Jonathan and Karin Fielding School of Public Health, and
| | - Jonathan Tucci
- Division of Pediatric Endocrinology, UCLA Children's Discovery and Innovation Institute, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA
| | | | - Weili Sun
- Pediatric Hematology Oncology, Department of Pediatrics, City of Hope National Medical Center, Duarte, CA
| | - Matthew J Oberley
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA; and
| | - Christina Dieli-Conwright
- Division of Population Sciences, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Steven D Mittelman
- Division of Pediatric Endocrinology, UCLA Children's Discovery and Innovation Institute, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA
| |
Collapse
|
30
|
An evaluation of participation restrictions and associated factors via the ICF-CY framework in children with acute lymphoblastic leukemia receiving maintenance chemotherapy. Eur J Pediatr 2021; 180:1081-1088. [PMID: 33063136 DOI: 10.1007/s00431-020-03833-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/14/2020] [Accepted: 10/09/2020] [Indexed: 10/23/2022]
Abstract
Our aim was to determine impairments in physical functions, activity limitations, and participation restrictions with the International Classification of Functioning, Disability and Health version for Children and Youth (ICF-CY) framework in children with acute lymphoblastic leukemia (ALL) receiving treatment. Physical functions were assessed in terms of pain level, fatigue level, handgrip strength, and motor proficiency. Fine motor activities and lower extremity performance were assessed to determine activity limitations. Participation was assessed with a patient-reported questionnaire. Thirty children with ALL (mean age: 8.45 ± 3.33 years) were included. Pain and fatigue level were mild. Poor handgrip strength was found; their mean handgrip strength was 60% of the normative. Fifty-six percent of the children had below-average motor performance. Participation scores were considerably high, except for sport and physical functioning sub-score. Participation level was positively associated with bilateral coordination and duration after diagnosis, while negatively correlated with pain and fatigue level (p ˂ 0.05).Conclusion: The ICF-CY-based evaluation was useful to understand children's limitations in everyday life. Children with ALL need supportive interventions during treatments in terms of physical functioning and participation in activities. Children with ALL with higher pain and fatigue, poor bilateral coordination, and who were in earlier period after diagnosis had higher risk for participation restriction. What is Known: • Children with ALL had physical functioning limitations on treatments. • Participation restrictions were described in children with ALL off treatment. What is New: • The ICY-CY-based health and functioning evaluation allows health care professionals to globally determine limitations of everyday life in children with ALL on treatment. • Impairments in physical functions, pain severity, fatigue severity, and duration after diagnosis are associated with participation to everyday life in children with ALL on treatment.
Collapse
|
31
|
Total Hip Arthroplasty in Adolescents and Young Adults for Management of Advanced Corticosteroid-Induced Osteonecrosis Secondary to Treatment for Hematologic Malignancies. J Arthroplasty 2021; 36:1352-1360. [PMID: 33281023 DOI: 10.1016/j.arth.2020.10.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 10/08/2020] [Accepted: 10/13/2020] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Osteonecrosis of the femoral head (ONFH) is a potentially severe toxicity associated with glucocorticoid treatment for pediatric hematologic malignancy. We examined clinical outcomes of THA in adolescents and young adults treated for hematologic malignancies who developed advanced ONFH. METHODS In a single-institution cohort, we retrospectively reviewed medical records and imaging for perioperative complications, reoperations, functional assessment at last follow-up, and radiological outcomes. Twenty-seven patients (41 hips) underwent THA (bilateral in 14 patients). There were 11 males. Median (interquartile range [IQR]) age at primary diagnosis was 14.9 years [1.8-18.9]. The median (IQR) age at THA was 19.8 years [14.6-30.3]. Mean (range) post-THA follow-up was 111.5 months (65.4-165.8). RESULTS Perioperative complications included one intraoperative calcar fracture that was secured with a cerclage wire and one posterior hip dislocation that occurred 6 days postoperatively, requiring closed reduction. One hip required a revision 21.1 months post-THA due to a fractured ceramic liner. The radiographic review was available for 38 of 41 hips and demonstrated none with loosening, subsidence, or osteolysis; nine developed periacetabular stress shielding. Incidence of stress shielding was associated with increased postoperative pain (P = .0130). There was a significant functional improvement in range of motion (ROM), pain, use of supports, participation in school, work, and sports, and use of pain medication from preoperative to postoperative clinical visits (P < .001). DISCUSSION Total hip arthroplasty in adolescents and young adults offers symptomatic and functional improvement in patients with ONFH. We found it to be safe with low perioperative complication rates even in patients undergoing active treatment for malignancy. LEVEL OF EVIDENCE Level IV, case series study. See Instructions for authors for a complete description of levels of evidence.
Collapse
|
32
|
Wood NM, Davis S, Lewing K, Noel-MacDonnell J, Glynn EF, Caragea D, Hoffman MA. Aligning EHR Data for Pediatric Leukemia With Standard Protocol Therapy. JCO Clin Cancer Inform 2021; 5:239-251. [PMID: 33656914 PMCID: PMC8140784 DOI: 10.1200/cci.20.00144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Children with acute lymphoblastic leukemia (ALL) are treated according to risk-based protocols defined by the Children's Oncology Group (COG). Alignment between real-world clinical practice and protocol milestones is not widely understood. Aggregate deidentified electronic health record (EHR) data offer a useful resource to evaluate real-world clinical practice.
Collapse
Affiliation(s)
- Nicole M Wood
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO.,Children's Mercy Research Institute, Kansas City, MO.,Department of Pediatrics, University of Missouri, Kansas City, MO
| | - Sierra Davis
- Children's Mercy Research Institute, Kansas City, MO
| | - Karen Lewing
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO.,Department of Pediatrics, University of Missouri, Kansas City, MO
| | - Janelle Noel-MacDonnell
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO.,Children's Mercy Research Institute, Kansas City, MO.,Department of Pediatrics, University of Missouri, Kansas City, MO
| | - Earl F Glynn
- Children's Mercy Research Institute, Kansas City, MO
| | | | - Mark A Hoffman
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO.,Children's Mercy Research Institute, Kansas City, MO.,Department of Pediatrics, University of Missouri, Kansas City, MO.,Department of Biomedical and Health Informatics, University of Missouri, Kansas City, MO
| |
Collapse
|
33
|
Heneghan MB, Hussain T, Barrera L, Cai SW, Haugen M, Morgan E, Rossoff J, Weinstein J, Hijiya N, Cella D, Badawy SM. Access to Technology and Preferences for an mHealth Intervention to Promote Medication Adherence in Pediatric Acute Lymphoblastic Leukemia: Approach Leveraging Behavior Change Techniques. J Med Internet Res 2021; 23:e24893. [PMID: 33599621 PMCID: PMC7932843 DOI: 10.2196/24893] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/04/2020] [Accepted: 01/07/2021] [Indexed: 12/16/2022] Open
Abstract
Background Suboptimal adherence to 6-mercaptopurine (6-MP) is prevalent in pediatric acute lymphoblastic leukemia (ALL) and associated with increased risk of relapse. Rapid uptake of personal technology makes mobile health (mHealth) an attractive platform to promote adherence. Objective Study objectives were to examine access to mobile technology and preferences for an mHealth intervention to improve medication adherence in pediatric ALL. Methods A cross-sectional survey was administered in oncology clinic to parents of children with ALL as well as adolescents and young adults (AYAs) with ALL receiving maintenance chemotherapy. Results A total of 49 parents (median age [IQR] 39 [33-42] years; female 76% [37/49]) and 15 patients (median age [IQR] 17 [16-19]; male 80% [12/15]) participated. All parents and AYAs owned electronic tablets, smartphones, or both. Parents’ most endorsed mHealth app features included a list of medications (71%, 35/49), information about 6-MP (71%, 35/49), refill reminders (71%, 35/49), and reminders to take 6-MP (71%, 35/49). AYAs' most endorsed features included refill reminders (73%, 11/15), reminders to take 6-MP (73%, 11/15), and tracking 6-MP (73%, 11/15). Conclusions Parents and AYAs reported ubiquitous access to mobile technology and strong interest in multiple adherence-specific mHealth app features. Parents and AYAs provided valuable insight into preferred features for a multifunctional behavioral intervention (mHealth app) to promote medication adherence in pediatric ALL.
Collapse
Affiliation(s)
- Mallorie B Heneghan
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, University of Utah, Salt Lake City, UT, United States
| | - Tasmeen Hussain
- Division of Internal Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Leonardo Barrera
- Mary Ann & J. Milburn Smith Child Health Research, Outreach, and Advocacy Center, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, United States
| | - Stephanie W Cai
- Department of Obstetrics & Gynecology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Maureen Haugen
- Division of Hematology, Oncology and Stem Cell Transplantation, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, United States
| | - Elaine Morgan
- Division of Hematology, Oncology and Stem Cell Transplantation, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, United States.,Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Jenna Rossoff
- Division of Hematology, Oncology and Stem Cell Transplantation, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, United States.,Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Joanna Weinstein
- Division of Hematology, Oncology and Stem Cell Transplantation, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, United States.,Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Nobuko Hijiya
- Department of Pediatrics, Columbia University Medical Center, New York, NY, United States
| | - David Cella
- Department of Medical Social Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Sherif M Badawy
- Division of Hematology, Oncology and Stem Cell Transplantation, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, United States.,Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
34
|
Mateos MK, Marshall GM, Barbaro PM, Quinn MC, George C, Mayoh C, Sutton R, Revesz T, Giles JE, Barbaric D, Alvaro F, Mechinaud F, Catchpoole D, Lawson JA, Chenevix-Trench G, MacGregor S, Kotecha RS, Dalla-Pozza L, Trahair TN. Methotrexate-related central neurotoxicity: clinical characteristics, risk factors and genome-wide association study in children treated for acute lymphoblastic leukemia. Haematologica 2021; 107:635-643. [PMID: 33567813 PMCID: PMC8883571 DOI: 10.3324/haematol.2020.268565] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Indexed: 11/09/2022] Open
Abstract
Symptomatic methotrexate-related central neurotoxicity, 'MTX neurotoxicity', is a severe toxicity experienced during acute lymphoblastic leukemia (ALL) therapy with potential long-term neurologic complications. Risk factors and long-term outcomes require further study. We conducted a systematic, retrospective review of 1251 consecutive Australian children enrolled on BFM or COG-based protocols between 1998-2013. Clinical risk predictors for MTX neurotoxicity were analyzed using regression. A genome-wide association study (GWAS) was performed on 48 cases and 537 controls. The incidence of MTX neurotoxicity was 7.6% (n=95/1251), at a median of 4 months from ALL diagnosis and 8 days after intravenous or intrathecal MTX. Grade 3 elevation of serum aspartate aminotransferase (P=0.005, OR 2.31 (1.28-4.16)) in induction/consolidation was associated with MTX neurotoxicity, after accounting for the only established risk factor, age a10 years. Cumulative incidence of CNS relapse was increased in children where intrathecal MTX was omitted following symptomatic MTX neurotoxicity (n=48) compared to where intrathecal MTX was continued throughout therapy (n=1174) (P=0.047). Five-year CNS relapsefree survival was 89.2%±4.6% when intrathecal MTX was ceased compared to 95.4%±0.6% when intrathecal MTX was continued. Recurrence of MTX neurotoxicity was low (12.9%) for patients whose intrathecal MTX was continued after their first episode. The GWAS identified SNPs associated with MTX neurotoxicity near genes regulating neuronal growth, neuronal differentiation and cytoskeletal organization (P>1E-06). In conclusion, increased serum aspartate aminotransferase and age a10 years at diagnosis were independent risk factors for MTX neurotoxicity. Our data do not support cessation of intrathecal MTX after a first MTX neurotoxicity event.
Collapse
Affiliation(s)
- Marion K Mateos
- Kids Cancer Centre, Sydney Children's Hospital Randwick, Sydney, Australia; School of Women and Children's Health, University of New South Wales (UNSW), Sydney, Australia; Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, Australia; Northern Institute for Cancer Research, Wolfson Childhood Cancer Research Centre, Newcastle-Upon-Tyne
| | - Glenn M Marshall
- Kids Cancer Centre, Sydney Children's Hospital Randwick, Sydney, Australia; School of Women and Children's Health, University of New South Wales (UNSW), Sydney, Australia; Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney
| | - Pasquale M Barbaro
- Children's Medical Research Institute, University of Sydney, Sydney, Australia; Department of Haematology, Queensland Children's Hospital, Brisbane
| | | | - Carly George
- Perth Children's Hospital, Perth, Australia; Division of Paediatrics, School of Medicine, University of Western Australia, Perth
| | - Chelsea Mayoh
- School of Women and Children's Health, University of New South Wales (UNSW), Sydney, Australia; Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney
| | - Rosemary Sutton
- School of Women and Children's Health, University of New South Wales (UNSW), Sydney, Australia; Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney
| | | | - Jodie E Giles
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney
| | - Draga Barbaric
- Kids Cancer Centre, Sydney Children's Hospital Randwick, Sydney
| | - Frank Alvaro
- John Hunter Children's Hospital, Newcastle, Australia; University of Newcastle, Newcastle
| | - Françoise Mechinaud
- The Royal Children's Hospital, Melbourne, Australia; Service d'Immuno-hématologie pédiatrique Hôpital Robert-Debré, Paris
| | - Daniel Catchpoole
- The Tumour Bank, Children's Cancer Research Unit, The Children's Hospital at Westmead, Sydney
| | - John A Lawson
- School of Women and Children's Health, University of New South Wales (UNSW), Sydney, Australia; Department of Neurology, Sydney Children's Hospital Randwick, Sydney
| | | | | | - Rishi S Kotecha
- Perth Children's Hospital, Perth, Australia; Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia, Perth, Australia; School of Pharmacy and Biomedical Sciences, Curtin University, Perth
| | - Luciano Dalla-Pozza
- Children's Medical Research Institute, University of Sydney, Sydney, Australia; Cancer Centre for Children, The Children's Hospital at Westmead, Sydney, Australia; Children's Cancer Research Unit, The Children's Hospital at Westmead, Sydney
| | - Toby N Trahair
- Kids Cancer Centre, Sydney Children's Hospital Randwick, Sydney, Australia; School of Women and Children's Health, University of New South Wales (UNSW), Sydney, Australia; Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney.
| |
Collapse
|
35
|
Brown P, Inaba H, Annesley C, Beck J, Colace S, Dallas M, DeSantes K, Kelly K, Kitko C, Lacayo N, Larrier N, Maese L, Mahadeo K, Nanda R, Nardi V, Rodriguez V, Rossoff J, Schuettpelz L, Silverman L, Sun J, Sun W, Teachey D, Wong V, Yanik G, Johnson-Chilla A, Ogba N. Pediatric Acute Lymphoblastic Leukemia, Version 2.2020, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2021; 18:81-112. [PMID: 31910389 DOI: 10.6004/jnccn.2020.0001] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Acute lymphoblastic leukemia (ALL) is the most common pediatric malignancy. Advancements in technology that enhance our understanding of the biology of the disease, risk-adapted therapy, and enhanced supportive care have contributed to improved survival rates. However, additional clinical management is needed to improve outcomes for patients classified as high risk at presentation (eg, T-ALL, infant ALL) and who experience relapse. The NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for pediatric ALL provide recommendations on the workup, diagnostic evaluation, and treatment of the disease, including guidance on supportive care, hematopoietic stem cell transplantation, and pharmacogenomics. This portion of the NCCN Guidelines focuses on the frontline and relapsed/refractory management of pediatric ALL.
Collapse
Affiliation(s)
- Patrick Brown
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | - Hiroto Inaba
- St. Jude Children's Research Hospital/The University of Tennessee Health Science Center
| | - Colleen Annesley
- Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | | | - Susan Colace
- The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute
| | - Mari Dallas
- Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | | | - Kara Kelly
- Roswell Park Comprehensive Cancer Center
| | | | | | | | - Luke Maese
- Huntsman Cancer Institute at the University of Utah
| | - Kris Mahadeo
- The University of Texas MD Anderson Cancer Center
| | | | | | | | - Jenna Rossoff
- Ann & Robert H. Lurie Children's Hospital of Chicago
| | - Laura Schuettpelz
- Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | | | | | - Weili Sun
- City of Hope National Medical Center
| | - David Teachey
- Abramson Cancer Center at the University of Pennsylvania
| | | | | | | | | |
Collapse
|
36
|
Rosdiana DS, Setiabudy R, Andalusia R, Gatot D, Louisa M, Bardosono S, Instiaty I. TPMT Genetic Variability and Its Association with Hematotoxicity in Indonesian Children with Acute Lymphoblastic Leukemia in Maintenance Therapy. Pharmgenomics Pers Med 2021; 14:199-210. [PMID: 33568932 PMCID: PMC7868246 DOI: 10.2147/pgpm.s288988] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 12/29/2020] [Indexed: 11/23/2022] Open
Abstract
PURPOSE Hematotoxicity monitoring in children with acute lymphoblastic leukemia (ALL) is critical to preventing life-threatening infections and drug discontinuation. The primary drug that causes hematotoxicity in ALL children is 6-mercaptopurine (6-MP). Genetic variability of the drug-metabolizing enzymes of 6-MP, thiopurine S-methyltransferase (TPMT), is one factor that might increase the susceptibility of children to hematotoxicity. The present study aimed to determine the variability in TPMT genotypes and phenotypes and its association with the occurrence of hematotoxicity in ALL children in maintenance therapy. PATIENTS AND METHODS A cross-sectional study was conducted at Cipto Mangunkusumo and Dharmais National Cancer Hospital, Jakarta, Indonesia, from June 2017 to October 2018. We included ALL patients, 1-18 years, who were receiving at least one month of 6-MP during maintenance therapy according to the Indonesian protocol for ALL 2013. Direct sequencing was used to determine TPMT*3A, *3B, and *3C genotypes, and LC-MS/MS analysis was performed to measure the plasma concentrations of 6-MP and its metabolites. Association analysis between the TPMT genotype and hematotoxicity was evaluated using the unpaired t-test or Mann-Whitney's test. RESULTS The prevalence of neutropenia, anemia, and thrombocytopenia in ALL children during maintenance therapy was 51.9%, 44.3%, and 6.6%, respectively. We found a low frequency of TPMT*3C, which is 0.95%. No association was found between hematotoxicity and TPMT genotypes or age, nutritional status, serum albumin levels, risk stratification, the daily dose of 6-MP, and cotrimoxazole co-administration. However, hematotoxicity was associated with 6-methylmercaptopurine (6-MeMP) plasma concentrations and the ratio 6-MeMP/6-thioguanine (6-TGN). We also found no association between TPMT genotypes and TPMT phenotypes. CONCLUSION The 6-MeMP/6-TGN ratio is associated with hematotoxicity in ALL children during maintenance therapy but is not strong enough to predict hematotoxicity.
Collapse
Affiliation(s)
- Dewi Selvina Rosdiana
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Rianto Setiabudy
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Rizka Andalusia
- Dharmais National Cancer Hospital, Jakarta, Indonesia
- Drug Registration Directorate, National Agency for Drug and Food Control, Jakarta, Indonesia
| | - Djajadiman Gatot
- Division of Hematology-Oncology, Department of Pediatrics, Faculty of Medicine, Universitas Indonesia/Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Melva Louisa
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Saptawati Bardosono
- Department of Nutrition, Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
| | - Instiaty Instiaty
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| |
Collapse
|
37
|
Orgel E, Nabais T, Douglas C, Mittelman SD, Neely M. Effect of Body Fat on Population Pharmacokinetics of High-Dose Methotrexate in Pediatric Patients With Acute Lymphoblastic Leukemia. J Clin Pharmacol 2021; 61:755-762. [PMID: 33314168 DOI: 10.1002/jcph.1799] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 12/07/2020] [Indexed: 11/10/2022]
Abstract
Nearly all international regimens for pediatric acute lymphoblastic leukemia (ALL) incorporate intravenous "high-dose" methotrexate (HDMTX, ≥1 g/m2 ) to penetrate the central nervous system. Dosing is routinely adjusted for body surface area (BSA), but limited data describe the pharmacokinetics of HDMTX, particularly in obese and/or large patients. To understand the impact of body size (BSA) and body fat percentage (BFP) on HDMTX pharmacokinetics, we performed a secondary analysis of 36 children and adolescents 10-21 years old treated for newly diagnosed ALL and who were enrolled in a prospective study examining body composition. All patients received 5 g/m2 of HDMTX infused over 24 hours. Plasma methotrexate concentrations were measured at 24, 42, and 48 hours. At 48 hours, ≥0.4 μmol/L was defined as "delayed elimination," necessitating prolonged supportive care. BFP was measured using dual-energy x-ray absorptiometry. A nonparametric population pharmacokinetic model was constructed with subsequent simulations to explore effects of BSA and BFP extremes. Despite standard BSA-adjusted dosing, we found significant intrapatient variability in mean MTX concentration (38%; range, 1.2%-86%). BSA and BFP were not linearly associated with increased area under the curve (AUC, P = 0.74 and P = 0.12), but both larger size (BSA) and greater obesity (BFP) were associated with an approximately 2-fold higher risk for delayed elimination at 48 hours. HDMTX AUC was not associated with toxicity. MTX pharmacokinetics vary among and even within patients despite BSA-adjusted dosing. Obesity and large size are identified as new risk factors for delayed elimination, requiring further investigation.
Collapse
Affiliation(s)
- Etan Orgel
- Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Los Angeles, California, USA.,Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Teresa Nabais
- Laboratory of Applied Pharmacokinetics and Bioinformatics, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Christopher Douglas
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Steven D Mittelman
- Children's Discovery and Innovation Institute, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Michael Neely
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Laboratory of Applied Pharmacokinetics and Bioinformatics, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California, USA
| |
Collapse
|
38
|
Bhansali RS, Rammohan M, Lee P, Laurent AP, Wen Q, Suraneni P, Yip BH, Tsai YC, Jenni S, Bornhauser B, Siret A, Fruit C, Pacheco-Benichou A, Harris E, Besson T, Thompson BJ, Goo YA, Hijiya N, Vilenchik M, Izraeli S, Bourquin JP, Malinge S, Crispino JD. DYRK1A regulates B cell acute lymphoblastic leukemia through phosphorylation of FOXO1 and STAT3. J Clin Invest 2021; 131:135937. [PMID: 33393494 PMCID: PMC7773384 DOI: 10.1172/jci135937] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 08/11/2020] [Indexed: 01/17/2023] Open
Abstract
DYRK1A is a serine/threonine kinase encoded on human chromosome 21 (HSA21) that has been implicated in several pathologies of Down syndrome (DS), including cognitive deficits and Alzheimer's disease. Although children with DS are predisposed to developing leukemia, especially B cell acute lymphoblastic leukemia (B-ALL), the HSA21 genes that contribute to malignancies remain largely undefined. Here, we report that DYRK1A is overexpressed and required for B-ALL. Genetic and pharmacologic inhibition of DYRK1A decreased leukemic cell expansion and suppressed B-ALL development in vitro and in vivo. Furthermore, we found that FOXO1 and STAT3, transcription factors that are indispensable for B cell development, are critical substrates of DYRK1A. Loss of DYRK1A-mediated FOXO1 and STAT3 signaling disrupted DNA damage and ROS regulation, respectively, leading to preferential cell death in leukemic B cells. Thus, we reveal a DYRK1A/FOXO1/STAT3 axis that facilitates the development and maintenance of B-ALL.
Collapse
Affiliation(s)
- Rahul S. Bhansali
- Department of Medicine, Division of Hematology/Oncology, Northwestern University, Chicago, Illinois, USA
| | - Malini Rammohan
- Department of Medicine, Division of Hematology/Oncology, Northwestern University, Chicago, Illinois, USA
| | - Paul Lee
- Abbvie, North Chicago, Illinois, USA
| | | | - Qiang Wen
- Department of Medicine, Division of Hematology/Oncology, Northwestern University, Chicago, Illinois, USA
| | - Praveen Suraneni
- Department of Medicine, Division of Hematology/Oncology, Northwestern University, Chicago, Illinois, USA
| | - Bon Ham Yip
- Division of Experimental Hematology, Department of Hematology, St. Jude Children’s Hospital, Memphis, Tennessee, USA
| | - Yi-Chien Tsai
- Department of Pediatric Oncology, Children’s Research Centre, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Silvia Jenni
- Department of Pediatric Oncology, Children’s Research Centre, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Beat Bornhauser
- Department of Pediatric Oncology, Children’s Research Centre, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Aurélie Siret
- INSERM U1170, Gustave Roussy Institute, Villejuif, France
| | - Corinne Fruit
- Normandie University, UNIROUEN, Institut National des Sciences Appliquées (INSA) Rouen, CNRS, Chimie Organique et Bioorganique — Réactivité et Analyse (COBRA) UMR 6014, Rouen, France
| | - Alexandra Pacheco-Benichou
- Normandie University, UNIROUEN, Institut National des Sciences Appliquées (INSA) Rouen, CNRS, Chimie Organique et Bioorganique — Réactivité et Analyse (COBRA) UMR 6014, Rouen, France
| | - Ethan Harris
- College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Thierry Besson
- Normandie University, UNIROUEN, Institut National des Sciences Appliquées (INSA) Rouen, CNRS, Chimie Organique et Bioorganique — Réactivité et Analyse (COBRA) UMR 6014, Rouen, France
| | | | - Young Ah Goo
- Proteomics Center of Excellence, Northwestern University, Evanston, Illinois, USA
| | - Nobuko Hijiya
- Division of Pediatric Hematology/Oncology, Columbia University, New York, New York, USA
| | | | - Shai Izraeli
- Pediatric Hematology Oncology, Schneider Children’s Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Petah Tikva, Israel
| | - Jean-Pierre Bourquin
- Department of Pediatric Oncology, Children’s Research Centre, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Sébastien Malinge
- INSERM U1170, Gustave Roussy Institute, Villejuif, France
- Telethon Kids Institute, Telethon Kids Cancer Centre (TKCC), Nedlands, Western Australia, Australia
| | - John D. Crispino
- Department of Medicine, Division of Hematology/Oncology, Northwestern University, Chicago, Illinois, USA
- Division of Experimental Hematology, Department of Hematology, St. Jude Children’s Hospital, Memphis, Tennessee, USA
| |
Collapse
|
39
|
Kim JH, Li L, Resar LM. Doubling up on function: dual-specificity tyrosine-regulated kinase 1A (DYRK1A) in B cell acute lymphoblastic leukemia. J Clin Invest 2021; 131:142627. [PMID: 33393492 PMCID: PMC7773367 DOI: 10.1172/jci142627] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
DYRK1A, the dual-specificity kinase, is again doubling up on function, as reported by Bhansali, Rammohan, and colleagues in this issue of the JCI. DYRK1A is an evolutionarily conserved protein kinase with dual specificity; it adds phosphates to serine/threonine residues of diverse regulatory proteins and activates its own function by autophosphorylating a critical tyrosine at position 321 in the activation loop. Bhansali, Rammohan, and colleagues investigated B cell acute lymphoblastic leukemia (B-ALL) in individuals with Down syndrome (DS) and in children with leukemia characterized by aneuploidy. The study revealed a DYRK1A/FOXO1 and STAT3 signaling pathway in B-ALL that could be targeted pharmacologically, thus opening the door to therapeutic strategies for patients with leukemia with or without DS.
Collapse
Affiliation(s)
| | - Liping Li
- Department of Medicine, Division of Hematology
| | - Linda M.S. Resar
- Department of Medicine, Division of Hematology
- Departments of Oncology and Pathology, and Institute of Cellular Engineering, and
- Pathobiology, Human Genetics, and Cellular and Molecular Medicine Graduate Programs, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
40
|
Bárcenas-López DA, Mendiola-Soto DK, Núñez-Enríquez JC, Mejía-Aranguré JM, Hidalgo-Miranda A, Jiménez-Morales S. Promising genes and variants to reduce chemotherapy adverse effects in acute lymphoblastic leukemia. Transl Oncol 2021; 14:100978. [PMID: 33290991 PMCID: PMC7720095 DOI: 10.1016/j.tranon.2020.100978] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/17/2020] [Accepted: 11/25/2020] [Indexed: 12/12/2022] Open
Abstract
Almost two decades ago, the sequencing of the human genome and high throughput technologies came to revolutionize the clinical and therapeutic approaches of patients with complex human diseases. In acute lymphoblastic leukemia (ALL), the most frequent childhood malignancy, these technologies have enabled to characterize the genomic landscape of the disease and have significantly improved the survival rates of ALL patients. Despite this, adverse reactions from treatment such as toxicity, drug resistance and secondary tumors formation are still serious consequences of chemotherapy, and the main obstacles to reduce ALL-related mortality. It is well known that germline variants and somatic mutations in genes involved in drug metabolism impact the efficacy of drugs used in oncohematological diseases therapy. So far, a broader spectrum of clinically actionable alterations that seems to be crucial for the progression and treatment response have been identified. Although these results are promising, it is necessary to put this knowledge into the clinics to help physician make medical decisions and generate an impact in patients' health. This review summarizes the gene variants and clinically actionable mutations that modify the efficacy of antileukemic drugs. Therefore, knowing their genetic status before treatment is critical to reduce severe adverse effects, toxicities and life-threatening consequences in ALL patients.
Collapse
Affiliation(s)
- Diego Alberto Bárcenas-López
- Laboratorio de Genómica del Cáncer, Instituto Nacional de Medicina Genómica, Periferico Sur 4809, Arenal Tepepan, Del. Tlalpan, Mexico City 14610, Mexico; Programa de Doctorado, Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Diana Karen Mendiola-Soto
- Laboratorio de Genómica del Cáncer, Instituto Nacional de Medicina Genómica, Periferico Sur 4809, Arenal Tepepan, Del. Tlalpan, Mexico City 14610, Mexico; Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Juan Carlos Núñez-Enríquez
- Unidad de Investigación Médica en Epidemiología Clínica, Hospital de Pediatría, CMNSXXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Juan Manuel Mejía-Aranguré
- Unidad de Investigación Médica en Epidemiología Clínica, Hospital de Pediatría, CMNSXXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico; Coordinación de Investigación en Salud, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Alfredo Hidalgo-Miranda
- Laboratorio de Genómica del Cáncer, Instituto Nacional de Medicina Genómica, Periferico Sur 4809, Arenal Tepepan, Del. Tlalpan, Mexico City 14610, Mexico
| | - Silvia Jiménez-Morales
- Laboratorio de Genómica del Cáncer, Instituto Nacional de Medicina Genómica, Periferico Sur 4809, Arenal Tepepan, Del. Tlalpan, Mexico City 14610, Mexico.
| |
Collapse
|
41
|
Lauer M, Kernen E, Schwabe D, Lehrnbecher T, Porto L. The role of magnetic resonance imaging in the diagnosis of central nervous system involvement in children with acute lymphoblastic leukemia. Pediatr Blood Cancer 2020; 67:e28294. [PMID: 32743972 DOI: 10.1002/pbc.28294] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 02/18/2020] [Accepted: 03/08/2020] [Indexed: 01/13/2023]
Abstract
PURPOSE Acute lymphoblastic leukemia (ALL) is the most frequent malignancy in childhood. As central nervous system (CNS) involvement requires an intensified CNS-targeted therapy, timely diagnosis is essential. The aim of this retrospective analysis was to evaluate whether cranial magnetic resonance imaging (MRI) examinations findings correlate with cerebrospinal fluid (CSF) analysis on CNS involvement and whether MRI examinations reveal incidental findings with a clinical consequence. METHODS All pediatric patients with ALL at our institution between 1998 and 2016 were identified. Patients were divided into two groups: de novo and relapsed ALL. Both groups were analyzed separately for the presence of CNS involvement. Incidental findings were also evaluated. RESULTS Two hundred fifteen patients with de novo ALL and 31 with relapsed ALL were identified. In the de novo group, no patient was diagnosed CNS positive based on MRI results alone. In relapsed patients, only one patient had a positive MRI with negative CSF results and no neurological symptoms, thus was classified CNS positive solely on the basis of the MRI. In both groups, no patient showed an incidental finding that required therapy. CONCLUSION In our study, MRI examinations do not improve the detection of CNS involvement compared with CSF analysis alone. In addition, the analysis of incidental findings does not add value to the performance of an MRI examination performed prior to treatment. Overall, MRI prior to treatment in pediatric patients with ALL is not necessary.
Collapse
Affiliation(s)
- Monika Lauer
- Institute of Neuroradiology, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany
| | - Elisabeth Kernen
- Institute of Neuroradiology, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany
| | - Dirk Schwabe
- Paediatric Haematology and Oncology, Hospital for Children and Adolescents, University Hospital Frankfurt, Goethe-University, Frankfurt, Germany
| | - Thomas Lehrnbecher
- Paediatric Haematology and Oncology, Hospital for Children and Adolescents, University Hospital Frankfurt, Goethe-University, Frankfurt, Germany
| | - Luciana Porto
- Institute of Neuroradiology, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany
| |
Collapse
|
42
|
Cardoso de Carvalho D, Pereira Colares Leitão L, Mello Junior FAR, Vieira Wanderley A, de Souza TP, Borges Andrade de Sá R, Cohen-Paes A, Rodrigues Fernandes M, Santos S, Salim Khayat A, Pimentel de Assumpção P, Pereira Carneiro dos Santos N. Association between the TPMT*3C (rs1142345) Polymorphism and the Risk of Death in the Treatment of Acute Lymphoblastic Leukemia in Children from the Brazilian Amazon Region. Genes (Basel) 2020; 11:genes11101132. [PMID: 32992962 PMCID: PMC7601477 DOI: 10.3390/genes11101132] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/11/2020] [Accepted: 09/18/2020] [Indexed: 12/21/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the leading cause of death from pediatric cancer worldwide. However, marked ethnic disparities are found in the treatment of childhood ALL with less effective results and higher mortality rates being obtained in populations with a high level of Native American ancestry. Genetic variations of the patient can affect resistance to ALL chemotherapy and potentially play an important role in this disparity. In the present study, we investigated the association of 16 genetic polymorphisms with the cell and metabolic pathways of the chemotherapeutic agents used in the treatment of ALL with the risk of death in treating childhood ALL in patients with a high contribution of Amerindian ancestry, coming from the Brazilian Amazon. The study included 121 patients with B-cell ALL treated with the BFM-2002 protocol. We are the first to identify the association between the TPMT gene rs1142345 polymorphism and the high risk of death in treating childhood ALL. Patients with the CC genotype had an approximately 25.5 times higher risk of dying during treatment of the disease than patients with other genotypes (p = 0.019). These results may help elucidate how the patient's genetic characteristics contribute to the mortality disparity in populations with a high contribution of Native American ancestry. The rs1142345 variant of the TPMT gene could be used as a potential marker to early stratify patients at high risk of death in treating childhood ALL in the investigated population.
Collapse
Affiliation(s)
- Darlen Cardoso de Carvalho
- Oncology Research Nucleus, Universidade Federal do Pará, Belém 66063-023, Brazil; (D.C.d.C.); (L.P.C.L.); (F.A.R.M.J.); (A.V.W.); (R.B.A.d.S.); (A.C.-P.); (M.R.F.); (S.S.); (A.S.K.); (P.P.d.A.)
| | - Luciana Pereira Colares Leitão
- Oncology Research Nucleus, Universidade Federal do Pará, Belém 66063-023, Brazil; (D.C.d.C.); (L.P.C.L.); (F.A.R.M.J.); (A.V.W.); (R.B.A.d.S.); (A.C.-P.); (M.R.F.); (S.S.); (A.S.K.); (P.P.d.A.)
| | - Fernando Augusto Rodrigues Mello Junior
- Oncology Research Nucleus, Universidade Federal do Pará, Belém 66063-023, Brazil; (D.C.d.C.); (L.P.C.L.); (F.A.R.M.J.); (A.V.W.); (R.B.A.d.S.); (A.C.-P.); (M.R.F.); (S.S.); (A.S.K.); (P.P.d.A.)
| | - Alayde Vieira Wanderley
- Oncology Research Nucleus, Universidade Federal do Pará, Belém 66063-023, Brazil; (D.C.d.C.); (L.P.C.L.); (F.A.R.M.J.); (A.V.W.); (R.B.A.d.S.); (A.C.-P.); (M.R.F.); (S.S.); (A.S.K.); (P.P.d.A.)
- Departamento de Pediatria, Ophir Loyola Hospital, Belém 66063-240, Brazil
| | - Tatiane Piedade de Souza
- Human and Medical Genetics Laboratory, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66075-110, Brazil;
| | - Roberta Borges Andrade de Sá
- Oncology Research Nucleus, Universidade Federal do Pará, Belém 66063-023, Brazil; (D.C.d.C.); (L.P.C.L.); (F.A.R.M.J.); (A.V.W.); (R.B.A.d.S.); (A.C.-P.); (M.R.F.); (S.S.); (A.S.K.); (P.P.d.A.)
| | - Amanda Cohen-Paes
- Oncology Research Nucleus, Universidade Federal do Pará, Belém 66063-023, Brazil; (D.C.d.C.); (L.P.C.L.); (F.A.R.M.J.); (A.V.W.); (R.B.A.d.S.); (A.C.-P.); (M.R.F.); (S.S.); (A.S.K.); (P.P.d.A.)
| | - Marianne Rodrigues Fernandes
- Oncology Research Nucleus, Universidade Federal do Pará, Belém 66063-023, Brazil; (D.C.d.C.); (L.P.C.L.); (F.A.R.M.J.); (A.V.W.); (R.B.A.d.S.); (A.C.-P.); (M.R.F.); (S.S.); (A.S.K.); (P.P.d.A.)
| | - Sidney Santos
- Oncology Research Nucleus, Universidade Federal do Pará, Belém 66063-023, Brazil; (D.C.d.C.); (L.P.C.L.); (F.A.R.M.J.); (A.V.W.); (R.B.A.d.S.); (A.C.-P.); (M.R.F.); (S.S.); (A.S.K.); (P.P.d.A.)
- Human and Medical Genetics Laboratory, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66075-110, Brazil;
| | - André Salim Khayat
- Oncology Research Nucleus, Universidade Federal do Pará, Belém 66063-023, Brazil; (D.C.d.C.); (L.P.C.L.); (F.A.R.M.J.); (A.V.W.); (R.B.A.d.S.); (A.C.-P.); (M.R.F.); (S.S.); (A.S.K.); (P.P.d.A.)
| | - Paulo Pimentel de Assumpção
- Oncology Research Nucleus, Universidade Federal do Pará, Belém 66063-023, Brazil; (D.C.d.C.); (L.P.C.L.); (F.A.R.M.J.); (A.V.W.); (R.B.A.d.S.); (A.C.-P.); (M.R.F.); (S.S.); (A.S.K.); (P.P.d.A.)
- João de Barros Barreto University Hospital, Universidade Federal do Pará, Belém 66063-023, Brazil
| | - Ney Pereira Carneiro dos Santos
- Oncology Research Nucleus, Universidade Federal do Pará, Belém 66063-023, Brazil; (D.C.d.C.); (L.P.C.L.); (F.A.R.M.J.); (A.V.W.); (R.B.A.d.S.); (A.C.-P.); (M.R.F.); (S.S.); (A.S.K.); (P.P.d.A.)
- Human and Medical Genetics Laboratory, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66075-110, Brazil;
- Correspondence:
| |
Collapse
|
43
|
Maamari D, El-Khoury H, Saifi O, Muwakkit SA, Zgheib NK. Implementation of Pharmacogenetics to Individualize Treatment Regimens for Children with Acute Lymphoblastic Leukemia. Pharmgenomics Pers Med 2020; 13:295-317. [PMID: 32848445 PMCID: PMC7429230 DOI: 10.2147/pgpm.s239602] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/20/2020] [Indexed: 12/28/2022] Open
Abstract
Despite major advances in the management and high cure rates of childhood acute lymphoblastic leukemia (ALL), patients still suffer from many drug-induced toxicities, sometimes necessitating dose reduction, or halting of cytotoxic drugs with a secondary risk of disease relapse. In addition, investigators have noted significant inter-individual variability in drug toxicities and disease outcomes, hence the role of pharmacogenetics (PGx) in elucidating genetic polymorphisms in candidate genes for the optimization of disease management. In this review, we present the PGx data in association with main toxicities seen in children treated for ALL in addition to efficacy, with a focus on the most plausible germline PGx variants. We then follow with a summary of the highest evidence drug-gene annotations with suggestions to move forward in implementing preemptive PGx for the individualization of treatment regimens for children with ALL.
Collapse
Affiliation(s)
- Dimitri Maamari
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Habib El-Khoury
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Omran Saifi
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Samar A Muwakkit
- Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Nathalie K Zgheib
- Department of Pharmacology and Toxicology, American University of Beirut, Faculty of Medicine, Beirut, Lebanon
| |
Collapse
|
44
|
Dixon SB, Chen Y, Yasui Y, Pui CH, Hunger SP, Silverman LB, Ness KK, Green DM, Howell RM, Leisenring WM, Kadan-Lottick NS, Krull KR, Oeffinger KC, Neglia JP, Mertens AC, Hudson MM, Robison LL, Armstrong GT, Nathan PC. Reduced Morbidity and Mortality in Survivors of Childhood Acute Lymphoblastic Leukemia: A Report From the Childhood Cancer Survivor Study. J Clin Oncol 2020; 38:3418-3429. [PMID: 32706634 DOI: 10.1200/jco.20.00493] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
PURPOSE Risk-stratified therapy, which modifies treatment on the basis of clinical and biologic features, has improved 5-year overall survival of childhood acute lymphoblastic leukemia (ALL) to 90%, but its impact on long-term toxicity remains unknown. METHODS We assessed all-cause and health-related late mortality (including late effects of cancer therapy), subsequent malignant neoplasms (SMNs), chronic health conditions, and neurocognitive outcomes among 6,148 survivors of childhood ALL (median age, 27.9 years; range, 5.9-61.9 years) diagnosed between 1970 and 1999. Therapy combinations and treatment intensity defined 6 groups: 1970s-like (70s), standard- or high-risk 1980s-like (80sSR, 80sHR) and 1990s-like (90sSR, 90sHR), and relapse/transplantation (R/BMT). Cumulative incidence, standardized mortality ratios, and standardized incidence ratios were compared between treatment groups and with the US population. RESULTS Overall, 20-year all-cause late mortality was 6.6% (95% CI, 6.0 to 7.1). Compared with 70s, 90sSR and 90sHR experienced lower health-related late mortality (rate ratio [95% CI]: 90sSR, 0.2 [0.1 to 0.4]; 90sHR, 0.3 [0.1 to 0.7]), comparable to the US population (standardized mortality ratio [95% CI]: 90sSR, 1.3 [0.8 to 2.0]; 90sHR, 1.7 [0.7 to 3.5]). Compared with 70s, 90sSR had a lower rate of SMN (rate ratio [95% CI], 0.3 [0.1 to 0.6]) that was not different from that of the US population (standardized incidence ratio [95% CI], 1.0 [0.6 to 1.6]). The 90sSR group had fewer severe chronic health conditions than the 70s (20-year cumulative incidence [95% CI], 11.0% [9.7% to 12.3%] v 22.5% [19.4% to 25.5%]) and a lower prevalence of impaired memory (prevalence ratio [95% CI], 0.7 [0.6 to 0.9]) and task efficiency (0.5 [0.4 to 0.7]). CONCLUSION Risk-stratified therapy has reduced late morbidity and mortality among contemporary survivors of standard-risk ALL, represented by 90sSR. Health-related late mortality and SMN risks among 5-year survivors of contemporary, standard-risk childhood ALL are comparable to the general population.
Collapse
Affiliation(s)
- Stephanie B Dixon
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN
| | - Yan Chen
- School of Public Health, University of Alberta, Edmonton, Alberta, Canada
| | - Yutaka Yasui
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, TN
| | - Ching-Hon Pui
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN
| | - Stephen P Hunger
- Division of Oncology and the Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Lewis B Silverman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Kirsten K Ness
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, TN
| | - Daniel M Green
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN
| | - Rebecca M Howell
- Radiation Physics Department, The University of Texas at MD Anderson Cancer Center, Houston, TX
| | - Wendy M Leisenring
- Cancer Prevention and Clinical Statistics Programs, Fred Hutchinson Cancer Research Center, Seattle, WA
| | | | - Kevin R Krull
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, TN.,Department of Psychology, St Jude Children's Research Hospital, Memphis, TN
| | | | - Joseph P Neglia
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN
| | - Ann C Mertens
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Melissa M Hudson
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN.,Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, TN
| | - Leslie L Robison
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, TN
| | - Gregory T Armstrong
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, TN
| | - Paul C Nathan
- Division of Hematology/Oncology, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
45
|
Zhang R, Zhu H, Yuan Y, Zhao J, Yang X, Tian Z. Risk Factors for Relapse of Childhood B Cell Acute Lymphoblastic Leukemia. Med Sci Monit 2020; 26:e923271. [PMID: 32619211 PMCID: PMC7353297 DOI: 10.12659/msm.923271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background B cell acute lymphoblastic leukemia (B-ALL) is the most common type of ALL. This study aimed to explore risk factors for relapse of childhood B-ALL. Material/Methods Total of 102 pediatric B-ALL patients were included in this study. B-ALL patients were divided into a relapse group and a non-relapse group. Chemotherapy-induced agranulocytosis time, fusion gene, and minimal residual disease (MRD) were assessed. White blood cell (WBC) count in peripheral blood and risk stratification were evaluated in newly-diagnosed patients. Kaplan-Meier plots were used to evaluate the correlation between risk factors and relapse rates. Multivariate analysis was performed with Cox proportional hazard model to estimate relative risk (RR), 95% confidence interval (95% CI), and hazard ratio (HR). Finally, 99 cases of B-ALL were included in this study. Results There were significant differences between the relapse group and the non-relapse group in age (p=0.004), chemotherapy-induced agranulocytopenia (p=0.001), WBC count in peripheral blood of newly diagnosed patients (p=0.016), risk stratification (p=0.000), and MRD at 12th week (p=0.007). Age over 10 years, high-risk stratification, long period of agranulocytopenia, higher WBC counts, and MRD more than 10−4 were correlated with higher B-ALL relapse rate (p<0.05). Multivariate analysis showed significantly higher relapse rates for age ≥10 years, high-risk stratification, and MRD at 12th week >10−4, with RR (95% CI) of 4.001 (1.005–15.930), 4.964 (1.050–23.456), and 4.646 (1.383–15.614), respectively. Conclusions Agranulocytopenia ≤7 days, peripheral blood WBC >100×109/L, and MRD at 33rd day >10−4 were associated with B-ALL relapse. Age ≥10 years, high-risk stratification, and MRD at 12th week >10−4 were independent risk factors for relapse.
Collapse
Affiliation(s)
- Rongrong Zhang
- Department of Pediatrics, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China (mainland)
| | - Haiyan Zhu
- Department of Pediatrics, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China (mainland)
| | - Yufang Yuan
- Department of Pediatrics, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China (mainland)
| | - Jiou Zhao
- Jiangsu Food and Pharmaceutical Science College, Huaian, Jiangsu, China (mainland)
| | - Xiaochun Yang
- Department of Pediatrics, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China (mainland)
| | - Zhaofang Tian
- Department of Pediatrics, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China (mainland)
| |
Collapse
|
46
|
Minervini A, Coccaro N, Anelli L, Zagaria A, Specchia G, Albano F. HMGA Proteins in Hematological Malignancies. Cancers (Basel) 2020; 12:E1456. [PMID: 32503270 PMCID: PMC7353061 DOI: 10.3390/cancers12061456] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/25/2020] [Accepted: 06/01/2020] [Indexed: 02/07/2023] Open
Abstract
The high mobility group AT-Hook (HMGA) proteins are a family of nonhistone chromatin remodeling proteins known as "architectural transcriptional factors". By binding the minor groove of AT-rich DNA sequences, they interact with the transcription apparatus, altering the chromatin modeling and regulating gene expression by either enhancing or suppressing the binding of the more usual transcriptional activators and repressors, although they do not themselves have any transcriptional activity. Their involvement in both benign and malignant neoplasias is well-known and supported by a large volume of studies. In this review, we focus on the role of the HMGA proteins in hematological malignancies, exploring the mechanisms through which they enhance neoplastic transformation and how this knowledge could be exploited to devise tailored therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | | | - Francesco Albano
- Department of Emergency and Organ Transplantation (D.E.T.O.), Hematology Section, University of Bari, 70124 Bari, Italy; (A.M.); (N.C.); (L.A.); (A.Z.); (G.S.)
| |
Collapse
|
47
|
Grimes AC, Chen Y, Bansal H, Aguilar C, Perez Prado L, Quezada G, Estrada J, Tomlinson GE. Genetic markers for treatment-related pancreatitis in a cohort of Hispanic children with acute lymphoblastic leukemia. Support Care Cancer 2020; 29:725-731. [PMID: 32447501 DOI: 10.1007/s00520-020-05530-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 05/14/2020] [Indexed: 12/15/2022]
Abstract
PURPOSE Treatment-related pancreatitis (TRP) is a serious complication occurring in children with acute lymphoblastic leukemia (ALL). Those affected are at high risk for severe organ toxicity and treatment delays that can impact outcomes. TRP is associated with asparaginase, a standard therapeutic agent in childhood ALL. Native American ancestry, older age, high-risk leukemia, and increased use of asparaginase are linked to pancreatitis risk. However, dedicated genetic studies evaluating pancreatitis in childhood ALL include few Hispanics. Thus, the genetic basis for higher risk of pancreatitis among Hispanic children with ALL remains unknown. METHODS Cases of children with ALL treated in from 1994 through 2013 were reviewed and identified 14, all Hispanic, who developed pancreatitis related to asparaginase therapy. Forty-six controls consisting of Hispanic children treated on the same regimens without pancreatitis were selected for comparison. Total DNA isolated from whole blood was used for targeted DNA sequencing of 23 selected genes, including genes associated with pancreatitis without ALL and genes involved in asparagine metabolism. RESULTS Non-synonymous polymorphisms and frameshift deletions were detected in 15 genes. Most children with TRP had variants in ABAT, ASNS, and CFTR. Notably, children with TRP harbored many more CFTR variants (71.4%) compared with controls (39.1%). Among these, V470M (rs213950) was most frequent (OR 4.27, p = 0.025). CONCLUSIONS This is the first study of genetic factors in treatment-related pancreatitis in Hispanic children with ALL. Identifying correlative variants in ethnically vulnerable populations may improve screening to identify which patients with ALL are at greatest risk for pancreatitis.
Collapse
Affiliation(s)
- Allison C Grimes
- Department of Pediatrics, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Yidong Chen
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
- Department of Population Health Sciences, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Hima Bansal
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Christine Aguilar
- Department of Pediatrics, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Luz Perez Prado
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Gerardo Quezada
- Methodist Children's Hospital, San Antonio, TX, USA
- Children's Hospital of San Antonio, San Antonio, TX, USA
| | | | - Gail E Tomlinson
- Department of Pediatrics, University of Texas Health Science Center San Antonio, San Antonio, TX, USA.
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center San Antonio, San Antonio, TX, USA.
| |
Collapse
|
48
|
Burke MJ, Kostadinov R, Sposto R, Gore L, Kelley SM, Rabik C, Trepel JB, Lee MJ, Yuno A, Lee S, Bhojwani D, Jeha S, Chang BH, Sulis ML, Hermiston ML, Gaynon P, Huynh V, Verma A, Gardner R, Heym KM, Dennis RM, Ziegler DS, Laetsch TW, Oesterheld JE, Dubois SG, Pollard JA, Glade-Bender J, Cooper TM, Kaplan JA, Farooqi MS, Yoo B, Guest E, Wayne AS, Brown PA. Decitabine and Vorinostat with Chemotherapy in Relapsed Pediatric Acute Lymphoblastic Leukemia: A TACL Pilot Study. Clin Cancer Res 2020; 26:2297-2307. [PMID: 31969338 PMCID: PMC7477726 DOI: 10.1158/1078-0432.ccr-19-1251] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 09/20/2019] [Accepted: 01/17/2020] [Indexed: 11/16/2022]
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.
Collapse
Affiliation(s)
- Michael J Burke
- Division of Pediatric Hematology-Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin.
| | - Rumen Kostadinov
- Division of Pediatric Oncology, Johns Hopkins University, Baltimore, Maryland
| | - Richard Sposto
- Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Lia Gore
- Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, Colorado
| | - Shannon M Kelley
- Division of Pediatric Oncology, Johns Hopkins University, Baltimore, Maryland
| | - Cara Rabik
- Division of Pediatric Oncology, Johns Hopkins University, Baltimore, Maryland
| | | | | | | | | | - Deepa Bhojwani
- Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Sima Jeha
- St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Bill H Chang
- Department of Pediatrics, Oregon Health and Science University, Portland, Oregon
| | - Maria Luisa Sulis
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michelle L Hermiston
- Department of Pediatrics, UCSF Medical Center-Mission Bay, San Francisco, California
| | - Paul Gaynon
- Division of Pediatric Oncology, Johns Hopkins University, Baltimore, Maryland
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Van Huynh
- Department of Pediatrics, Children's Hospital of Orange County, Orange, California
| | - Anupam Verma
- Department of Pediatrics, Primary Children's Hospital, University of Utah, Salt Lake City, Utah
| | - Rebecca Gardner
- Department of Pediatrics, Seattle Children's Hospital, Seattle, Washington
| | - Kenneth M Heym
- Department of Pediatrics, Cook Children's Medical Center, Fort Worth, Texas
| | - Robyn M Dennis
- Department of Pediatrics, Nationwide Children's Hospital, Columbus, Ohio
| | - David S Ziegler
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, Australia
| | - Theodore W Laetsch
- Department of Pediatrics, UT Southwestern/Harold C. Simmons Comprehensive Cancer Center, Dallas, Texas
- Pauline Allen Gill Center for Cancer and Blood Disorders, Children's Health, Dallas, Texas
| | - Javier E Oesterheld
- Department of Pediatrics, Carolinas Medical Center/Levine Cancer Institute, Charlotte, North Carolina
| | - Steven G Dubois
- Department of Pediatrics, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
| | - Jessica A Pollard
- Department of Pediatrics, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
| | - Julia Glade-Bender
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Todd M Cooper
- Department of Pediatrics, Seattle Children's Hospital, Seattle, Washington
| | - Joel A Kaplan
- Department of Pediatrics, Carolinas Medical Center/Levine Cancer Institute, Charlotte, North Carolina
| | - Midhat S Farooqi
- Center for Pediatric Genomic Medicine, Children's Mercy Hospital, Kansas City, Missouri
| | - Byunggil Yoo
- Center for Pediatric Genomic Medicine, Children's Mercy Hospital, Kansas City, Missouri
| | - Erin Guest
- Center for Pediatric Genomic Medicine, Children's Mercy Hospital, Kansas City, Missouri
| | - Alan S Wayne
- Division of Pediatric Oncology, Johns Hopkins University, Baltimore, Maryland
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Patrick A Brown
- Division of Pediatric Oncology, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
49
|
Ou JY, Hanson HA, Ramsay JM, Kaddas HK, Pope CA, Leiser CL, VanDerslice J, Kirchhoff AC. Fine Particulate Matter Air Pollution and Mortality among Pediatric, Adolescent, and Young Adult Cancer Patients. Cancer Epidemiol Biomarkers Prev 2020; 29:1929-1939. [PMID: 32404444 DOI: 10.1158/1055-9965.epi-19-1363] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/28/2020] [Accepted: 03/03/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Air pollution is a carcinogen and causes pulmonary and cardiac complications. We examined the association of fine particulate matter pollution (PM2.5) and mortality from cancer and all causes among pediatric, adolescent, and young adult (AYA) patients with cancer in Utah, a state with considerable variation in PM2.5. METHODS We followed 2,444 pediatric (diagnosed ages 0-14) and 13,459 AYA (diagnosed ages 15-39) patients diagnosed in 1986-2015 from diagnosis to 5 and 10 years postdiagnosis, death, or emigration. We measured average monthly PM2.5 by ZIP code during follow-up. Separate pediatric and AYA multivariable Cox models estimated the association of PM2.5 and mortality. Among AYAs, we examined effect modification of PM2.5 and mortality by stage while controlling for cancer type. RESULTS Increases in PM2.5 per 5 μg/m3 were associated with cancer mortality in pediatric lymphomas and central nervous system (CNS) tumors at both time points, and all cause mortality in lymphoid leukemias [HR5-year = 1.32 (1.02-1.71)]. Among AYAs, PM2.5 per 5 μg/m3 was associated with cancer mortality in CNS tumors and carcinomas at both time points, and all cause mortality for all AYA cancer types [HR5-year = 1.06 (1.01-1.13)]. PM2.5 ≥12 μg/m3 was associated with cancer mortality among breast [HR5-year = 1.50 (1.29-1.74); HR10-year = 1.30 (1.13-1.50)] and colorectal cancers [HR5-year = 1.74 (1.29-2.35); HR10-year = 1.67 (1.20-2.31)] at both time points. Effect modification by stage was significant, with local tumors at highest risk. CONCLUSIONS PM2.5 was associated with mortality in pediatric and AYA patients with specific cancers. IMPACT Limiting PM2.5 exposure may be important for young cancer patients with certain cancers.See all articles in this CEBP Focus section, "Environmental Carcinogenesis: Pathways to Prevention."
Collapse
Affiliation(s)
- Judy Y Ou
- Huntsman Cancer Institute, Cancer Control and Population Sciences, University of Utah, Salt Lake City, Utah.
| | - Heidi A Hanson
- Huntsman Cancer Institute, Cancer Control and Population Sciences, University of Utah, Salt Lake City, Utah
- Department of Surgery, University of Utah School of Medicine, Salt Lake City, Utah
| | - Joemy M Ramsay
- Huntsman Cancer Institute, Cancer Control and Population Sciences, University of Utah, Salt Lake City, Utah
| | - Heydon K Kaddas
- Huntsman Cancer Institute, Cancer Control and Population Sciences, University of Utah, Salt Lake City, Utah
| | | | - Claire L Leiser
- Huntsman Cancer Institute, Cancer Control and Population Sciences, University of Utah, Salt Lake City, Utah
- Department of Population Health Sciences, University of Utah School of Medicine, Salt Lake City, Utah
| | - James VanDerslice
- Department of Family and Preventive Medicine, University of Utah School of Medicine, Salt Lake City, Utah
| | - Anne C Kirchhoff
- Huntsman Cancer Institute, Cancer Control and Population Sciences, University of Utah, Salt Lake City, Utah
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah
| |
Collapse
|
50
|
Heneghan MB, Hussain T, Barrera L, Cai SW, Haugen M, Duff A, Shoop J, Morgan E, Rossoff J, Weinstein J, Hijiya N, Cella D, Badawy SM. Applying the COM-B model to patient-reported barriers to medication adherence in pediatric acute lymphoblastic leukemia. Pediatr Blood Cancer 2020; 67:e28216. [PMID: 32068338 DOI: 10.1002/pbc.28216] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/10/2020] [Accepted: 01/28/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND Adherence to oral chemotherapy, including 6-mercaptopurine (6-MP), is suboptimal in pediatric acute lymphoblastic leukemia (ALL), which is associated with increased risk of relapse. Study objectives were to examine self-reported adherence to 6-MP and related barriers to adherence, mapped to the capability, opportunity, motivation, and behavior (COM-B) model for behavior change. PROCEDURE Forty-nine parents (median, 39 years old; 76% females) and 15 patients (median, 17 years old, 20% females) completed the study survey. RESULTS Suboptimal adherence was reported in 43% of parents and 73% of patients. Most parents and patients (80% and 90%, respectively) reported ≥1 adherence barrier. Parents reported difficulty helping their child meet others with ALL (43%), contacting community organizations (39%), and meeting other parents (37%). Patients reported difficulty finding out what their medications are (40%), finding out what 6-MP does (47%), and meeting other patients (40%). Using the COM-B, we found that parents and patients endorsed barriers in multiple components, especially physical (55%, 67%) and social opportunity (56%, 47%), highlighting that barriers to adherence may be multifaceted. CONCLUSIONS Our results suggest that parents and patients with ALL face various prevalent barriers to medication adherence and provide insight into the development of behavioral interventions focused on promoting adherence, which is essential to prevent relapse and optimize health outcomes in ALL.
Collapse
Affiliation(s)
- Mallorie B Heneghan
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Division of Hematology, Oncology and Stem Cell Transplantation, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Tasmeen Hussain
- Division of Internal Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Leonardo Barrera
- Mary Ann and J. Milburn Smith Child Health Research, Outreach, and Advocacy Center, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Stephanie W Cai
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Maureen Haugen
- Division of Hematology, Oncology and Stem Cell Transplantation, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Ashley Duff
- Division of Hematology, Oncology and Stem Cell Transplantation, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Jenny Shoop
- Division of Hematology, Oncology and Stem Cell Transplantation, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Elaine Morgan
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Division of Hematology, Oncology and Stem Cell Transplantation, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Jenna Rossoff
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Division of Hematology, Oncology and Stem Cell Transplantation, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Joanna Weinstein
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Division of Hematology, Oncology and Stem Cell Transplantation, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Nobuko Hijiya
- Department of Pediatrics, Columbia University Medical Center, New York, New York
| | - David Cella
- Northwestern University, Department of Medical Social Sciences, Feinberg School of Medicine, Chicago, Illinois
| | - Sherif M Badawy
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Division of Hematology, Oncology and Stem Cell Transplantation, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| |
Collapse
|