1
|
Maillard M, Nguyen JQ, Yang W, Hoshitsuki K, Relling MV, Caudle KE, Crews KR, Jeha S, Inaba H, Pui CH, Bhatia S, Karol SE, Antillon-Klussmann FG, Haidar CE, Bhojwani D, Yang JJ. Clinical Actionability of the NUDT15 *4 (p.R139H) Allele and Its Association With Hispanic Ethnicity. Clin Pharmacol Ther 2025; 117:724-731. [PMID: 39688234 DOI: 10.1002/cpt.3501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 11/10/2024] [Indexed: 12/18/2024]
Abstract
Nudix hydrolase 15 (NUDT15) deficiency is strongly associated with thiopurine-induced myelosuppression. Currently, testing for NUDT15 deficiency is based on the genotyping of the most frequent and clinically characterized no-function variants, that is, *2, *3 and *9. The Hispanic/Latino-predominant variant NUDT15 *4 (p.R139H) is classified as "uncertain function" by the Clinical Pharmacogenetics Implementation Consortium, because of insufficient data to ascertain its clinical actionability. In this study, we evaluated the association of NUDT15 *4 with mercaptopurine (6-MP) tolerance in a retrospective cohort of 1,399 patients with acute lymphoblastic leukemia (ALL) of diverse ancestries. All patients were wildtype for thiopurine methyltransferase gene. Patients were treated with 6-MP in the context of ALL frontline clinical trials. The tolerated dose of 6-MP was used to assess drug toxicity during the maintenance phase of ALL therapy. Patients with NUDT15 *1/*4 (n = 16, all of whom self-identified as Hispanic/Latino) tolerated a significantly lower dose of 6-MP than did those with NUDT15 *1/*1: median [interquartile range] of 39.0 [21.2-52.8] mg/m2, vs. 62.2 [47.9-71.6] mg/m2, P value < 0.001. No patient homozygous for *4 was detected. In a separate retrospective validation study, six patients were identified as having NUDT15 *1/*4 by routine clinical pharmacogenetics testing and tolerated a 6-MP median dose of 38.7 mg/m2 (IQR, 33.7-54.0), confirming the need for dose reduction attributed to the NUDT15 *4 variant. These results point to the need to include NUDT15 *4 in pharmacogenetics-guided thiopurine dosing algorithms.
Collapse
Affiliation(s)
- Maud Maillard
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Jenny Q Nguyen
- Personalized Care Program, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Wenjian Yang
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Keito Hoshitsuki
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Mary V Relling
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Kelly E Caudle
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Kristine R Crews
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Sima Jeha
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Department of Global Pediatric Medicine, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Hiroto Inaba
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Ching-Hon Pui
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Department of Global Pediatric Medicine, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Smita Bhatia
- Division of Pediatric Hematology-Oncology, Institute for Cancer Outcomes and Survivorship, University of Alabama, Birmingham, Alabama, USA
| | - Seth E Karol
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | - Cyrine E Haidar
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Deepa Bhojwani
- Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Jun J Yang
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
2
|
Yang MM, Singh R, Haugen M, Duff A, Shoop J, Morgan ER, Rossoff JE, Weinstein JL, Heneghan MB, Badawy SM. Adherence to 6-Mercaptopurine (6-MP) and Habit Strength in Pediatric Acute Lymphoblastic Leukemia (ALL). Eur J Haematol 2025. [PMID: 39832489 DOI: 10.1111/ejh.14386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 12/31/2024] [Accepted: 01/05/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Low 6-mercaptopurine (6-MP) adherence (< 95%) is associated with increased relapse in pediatric acute lymphoblastic leukemia (ALL). Stronger habit has been associated with higher adherence. We examined the relationship of 6-MP adherence to habit strength and health-related quality of life in pediatric ALL. METHODS A single-center, cross-sectional study of 52 participants: 11 patients (mean age 16 ± 4) and 41 parents/caregivers (age 37 ± 5). Participants completed Visual Analogue Scale (VASdose), Patient Reported Outcomes Measurement Information System Medication Adherence Scale, and the Self-Regulated Habit Index (SRHI). Twelve semi-structured participant interviews were analyzed using thematic analysis. RESULTS 81% (42/52) of participants reported high 6-MP adherence (VASdose ≥ 95%): patients 91% (10/11), parents 78% (32/41). No significant correlation was found between adherence and habit strength. Reported adherence facilitators included reminders, care team communications, personalized tools, administration experience, self-efficacy, and social support. Conversely, financial burden, scheduling conflicts, and medication access were cited as barriers. CONCLUSIONS One-fifth of participants reported low 6-MP adherence, with habit strength not associated with adherence. Variability of 6-MP routines may prohibit automaticity. While 6-MP adherence may not correlate with habit strength, interventions promoting and strengthening habit formation may overcome barriers to 6-MP adherence and improve outcomes.
Collapse
Affiliation(s)
- Mira Muxi Yang
- Department of Medical Education, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Revika Singh
- Department of Medical Education, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Maureen Haugen
- Division of Hematology, Oncology, and Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, USA
| | - Ashley Duff
- Division of Hematology, Oncology, and Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, USA
| | - Jenny Shoop
- Division of Hematology, Oncology, and Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, USA
| | - Elaine R Morgan
- Division of Hematology, Oncology, and Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, USA
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Jenna E Rossoff
- Division of Hematology, Oncology, and Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, USA
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Joanna L Weinstein
- Division of Hematology, Oncology, and Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, USA
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Mallorie B Heneghan
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Utah, Salt Lake City, USA
| | - Sherif M Badawy
- Division of Hematology, Oncology, and Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, USA
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, USA
| |
Collapse
|
3
|
Wadhwa A, Chen Y, Hageman L, Angiolillo A, Dickens DS, Neglia JP, Ravindranath Y, Termuhlen A, Wong FL, Landier W, Bhatia S. Antimetabolite dose intensity and adverse outcomes in children with acute lymphoblastic leukemia: a COG-AALL03N1 report. Blood 2024; 144:2327-2335. [PMID: 39190431 PMCID: PMC11619787 DOI: 10.1182/blood.2024024455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 07/24/2024] [Accepted: 08/13/2024] [Indexed: 08/28/2024] Open
Abstract
ABSTRACT The association between antimetabolite dose intensity (DI) and adverse events among children receiving maintenance therapy for acute lymphoblastic leukemia (ALL) remains unclear, especially in the context of antimetabolite adherence. Using Children's Oncology Group AALL03N1 data, we examined the association between high DI during the first 4 study months and (i) treatment-related toxicities during the subsequent 2 study months; and (ii) relapse risk. Patients were classified into a high DI phenotype (either 6-mercaptopurine [6-MP] or methotrexate [MTX] DI ≥110% during the first 4 study months, or 6-MPDI or MTXDI 100%-110% at study enrollment and ≥25% increase over the 4 study months) and normal DI phenotype (all others). Only patients with wild-type TPMT and NUDT15 were included. 6-MP adherence data were available for 63.7% of study participants and used to stratify as adherent (median adherence ≥85%) and nonadherent (median adherence <85%) participants. Multivariable analyses were adjusted for sociodemographic and clinical prognosticators. Of the 644 patients, 29.3% were exposed to high DI. High DI was associated with a 2.1-fold greater odds of hematologic toxicity (95% confidence interval [CI] = 1.4-3.1; reference: normal DI) in the entire cohort and 2.9-fold higher among adherers (95% CI = 1.6-5.1); odds were comparable among nonadherers (2.1-fold; 95% CI = 0.4-10.1). Although high DI was not associated with relapse in the entire cohort (adjusted hazard ratio [aHR] = 1.4; 95% CI = 0.8-2.4), it was associated with a greater hazard of relapse among adherent participants (aHR = 2.4; 95% CI = 1.0-5.5) but not among nonadherent participants (aHR = 0.9; 95% CI = 0.2-3.8). Dose escalation above protocol doses during maintenance therapy for ALL should be done cautiously after assessing adherence to prescribed therapy.
Collapse
Affiliation(s)
- Aman Wadhwa
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
- Division of Pediatric Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL
| | - Yanjun Chen
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Lindsey Hageman
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | | | - David S. Dickens
- Division of Pediatric Hematology/Oncology, University of Iowa, Iowa City, IA
| | - Joseph P. Neglia
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN
| | | | - Amanda Termuhlen
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN
| | - F. Lennie Wong
- Department of Population Sciences, City of Hope, Duarte, CA
| | - Wendy Landier
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
- Division of Pediatric Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL
| | - Smita Bhatia
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
- Division of Pediatric Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
4
|
Källström J, Niinimäki R, Fredlund J, Vogt H, Korhonen L, Castor A, Palle J, Harila A, Borssén M, Abrahamsson J, Ek T. Effects of allopurinol on 6-mercaptopurine metabolism in unselected patients with pediatric acute lymphoblastic leukemia: a prospective phase II study. Haematologica 2024; 109:2846-2853. [PMID: 38356449 PMCID: PMC11367227 DOI: 10.3324/haematol.2023.284390] [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: 09/30/2023] [Accepted: 02/02/2024] [Indexed: 02/16/2024] Open
Abstract
Allopurinol can be used in maintenance therapy (MT) for pediatric acute lymphoblastic leukemia (ALL) to mitigate hepatic toxicity in patients with skewed 6-mercaptopurine metabolism. Allopurinol increases the erythrocyte levels of thioguanine nucleotides (e-TGN), which is the proposed main mediator of the antileukemic effect and decreases methyl mercaptopurine (e-MeMP) levels, associated with hepatotoxicity. We investigated the effects of allopurinol in thiopurine methyltransferase (TPMT) wild-type patients without previous clinical signs of skewed 6-mercaptopurine metabolism. Fifty-one patients from Sweden and Finland were enrolled in this prospective before-after trial during ALL MT. Mean e-TGN increased from 280 nmol/mmol hemoglobin (Hb) after 12 weeks of standard MT to 440 after 12 weeks of MT with addition of allopurinol 50 mg/ m2 (P<0.001). Mean e-MeMP decreased simultaneously from 9,481 nmol/mmol Hb to 2,791 (P<0.001) and mean alanine aminotransferase declined by almost 50%. Primary endpoint, defined as e-TGN >200 nmol/mmol Hb, was reached for 91% of the patients after 12 weeks of allopurinol (week 25) compared to 67% before (week 13) (P<0.001). This level was chosen as the median e-TGN in a previous NOPHO ALL-2008 study was just below 200 nmol/mmol Hb. During weeks on allopurinol a slightly higher proportion of the patients had a white blood cell count within target 1.5-3.0×109/L. Allopurinol did not increase severe adverse events and no life-threatening events were reported. In conclusion, allopurinol add-on treatment is safe and leads to increased e-TGN and reduced e-MeMP also in ALL-patients without previous signs of skewed thiopurine metabolism and is a promising approach to increase antileukemic effect and reduce toxicity.
Collapse
Affiliation(s)
- Jonatan Källström
- Children's Cancer Centre, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Pediatrics, Institute for Clinical Sciences, University of Gothenburg
| | - Riita Niinimäki
- Department of Pediatrics and Adolescents, Oulu University Hospital
| | | | - Hartmut Vogt
- Department of Pediatrics, Linköping University Hospital
| | | | - Anders Castor
- Department of Pediatrics, Skåne University Hospital, Lund
| | - Josefine Palle
- Department of Woman's and Children's Health, Uppsala University
| | - Arja Harila
- Department of Woman's and Children's Health, Uppsala University
| | - Magnus Borssén
- Department of Pediatrics, Norrlands University Hospital, Umeå
| | - Jonas Abrahamsson
- Children's Cancer Centre, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Pediatrics, Institute for Clinical Sciences, University of Gothenburg
| | - Torben Ek
- Children's Cancer Centre, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Pediatrics, Institute for Clinical Sciences, University of Gothenburg.
| |
Collapse
|
5
|
Maillard M, Nishii R, Yang W, Hoshitsuki K, Chepyala D, Lee SHR, Nguyen JQ, Relling MV, Crews KR, Leggas M, Singh M, Suang JLY, Yeoh AEJ, Jeha S, Inaba H, Pui CH, Karol SE, Trehan A, Bhatia P, Antillon Klussmann FG, Bhojwani D, Haidar CE, Yang JJ. Additive effects of TPMT and NUDT15 on thiopurine toxicity in children with acute lymphoblastic leukemia across multiethnic populations. J Natl Cancer Inst 2024; 116:702-710. [PMID: 38230823 PMCID: PMC11077315 DOI: 10.1093/jnci/djae004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 12/22/2023] [Accepted: 01/04/2024] [Indexed: 01/18/2024] Open
Abstract
BACKGROUND Thiopurines such as mercaptopurine (MP) are widely used to treat acute lymphoblastic leukemia (ALL). Thiopurine-S-methyltransferase (TPMT) and Nudix hydrolase 15 (NUDT15) inactivate thiopurines, and no-function variants are associated with drug-induced myelosuppression. Dose adjustment of MP is strongly recommended in patients with intermediate or complete loss of activity of TPMT and NUDT15. However, the extent of dosage reduction recommended for patients with intermediate activity in both enzymes is currently not clear. METHODS MP dosages during maintenance were collected from 1768 patients with ALL in Singapore, Guatemala, India, and North America. Patients were genotyped for TPMT and NUDT15, and actionable variants defined by the Clinical Pharmacogenetics Implementation Consortium were used to classify patients as TPMT and NUDT15 normal metabolizers (TPMT/NUDT15 NM), TPMT or NUDT15 intermediate metabolizers (TPMT IM or NUDT15 IM), or TPMT and NUDT15 compound intermediate metabolizers (TPMT/NUDT15 IM/IM). In parallel, we evaluated MP toxicity, metabolism, and dose adjustment using a Tpmt/Nudt15 combined heterozygous mouse model (Tpmt+/-/Nudt15+/-). RESULTS Twenty-two patients (1.2%) were TPMT/NUDT15 IM/IM in the cohort, with the majority self-reported as Hispanics (68.2%, 15/22). TPMT/NUDT15 IM/IM patients tolerated a median daily MP dose of 25.7 mg/m2 (interquartile range = 19.0-31.1 mg/m2), significantly lower than TPMT IM and NUDT15 IM dosage (P < .001). Similarly, Tpmt+/-/Nudt15+/- mice displayed excessive hematopoietic toxicity and accumulated more metabolite (DNA-TG) than wild-type or single heterozygous mice, which was effectively mitigated by a genotype-guided dose titration of MP. CONCLUSION We recommend more substantial dose reductions to individualize MP therapy and mitigate toxicity in TPMT/NUDT15 IM/IM patients.
Collapse
Affiliation(s)
- Maud Maillard
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Rina Nishii
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Wenjian Yang
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Keito Hoshitsuki
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Divyabharathi Chepyala
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Shawn H R Lee
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Children’s Research Hospital, Memphis, TN, USA
- Khoo Teck Puat-National University Children’s Medical Institute, National University Hospital, National University Health System, Singapore, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jenny Q Nguyen
- Personalized Care Program, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Mary V Relling
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Kristine R Crews
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Mark Leggas
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Meenu Singh
- Haematology-Oncology Unit, Department of Paediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Joshua L Y Suang
- Khoo Teck Puat-National University Children’s Medical Institute, National University Hospital, National University Health System, Singapore, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Allen E J Yeoh
- Khoo Teck Puat-National University Children’s Medical Institute, National University Hospital, National University Health System, Singapore, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sima Jeha
- Department of Oncology, St Jude Children’s Research Hospital, Memphis, TN, USA
- Department of Global Pediatric Medicine, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Hiroto Inaba
- Department of Oncology, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Ching-Hon Pui
- Department of Oncology, St Jude Children’s Research Hospital, Memphis, TN, USA
- Department of Global Pediatric Medicine, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Seth E Karol
- Department of Oncology, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Amita Trehan
- Haematology-Oncology Unit, Department of Paediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Prateek Bhatia
- Haematology-Oncology Unit, Department of Paediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | | | - Deepa Bhojwani
- Cancer and Blood Disease Institute, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Cyrine E Haidar
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Jun J Yang
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Children’s Research Hospital, Memphis, TN, USA
- Department of Oncology, St Jude Children’s Research Hospital, Memphis, TN, USA
| |
Collapse
|
6
|
Franca R, Stocco G, Kiren V, Tessitore A, Fagioli F, Quarello P, Bertorello N, Rizzari C, Colombini A, Bettini LR, Locatelli F, Vinti L, Girardi K, Silvestri D, Valsecchi MG, Decorti G, Rabusin M. Impact of Mercaptopurine Metabolites on Disease Outcome in the AIEOP-BFM ALL 2009 Protocol for Acute Lymphoblastic Leukemia. Clin Pharmacol Ther 2023; 114:1082-1092. [PMID: 37550838 DOI: 10.1002/cpt.3022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 08/01/2023] [Indexed: 08/09/2023]
Abstract
In the maintenance phase of Associazione Italiana di Ematologia e Oncologia Pediatrica (AIEOP)- Berlin-Frankfurt-Muenster (BFM) acute lymphoblastic leukemia (ALL) 2009 protocol, mercaptopurine (MP) is given at the planned dose of 50 mg/m2 /day; however, dose adjustments are routinely performed to target patients' white blood cells to the optimal range of 2,000-3,000 cells/μL. Pediatric patients with ALL (n = 290, age: median (1st-3rd quartile): 4.8 (3.0-8.1) years; boys: 56.9%) were enrolled mainly in 4 medium-large Italian pediatric hospitals; 14.1% of patients relapsed after a median (1st-3rd quartile) follow-up time of 4.43 (3.82-5.46) years from maintenance beginning. MP metabolites (thionucleotide (TGN) and methyl-derivatives (MMPN)) were measured in the erythrocytes of 387 blood samples of 200 patients by high performance liquid chromatography with ultraviolet detection. Single-nucleotide polymorphisms (SNPs; (rs1800462, rs1800460, and rs1142345 in TPMT gene, rs116855232 in NUDT15, rs1127354, rs7270101, rs6051702 in ITPA, and rs2413739 in PACSIN2) were characterized by Taqman SNP genotyping assays. Cox proportional hazard models did not show an impact of TGN levels and variability on relapse. In contrast, after multivariate analysis, relapse hazard ratio (HR) increased in children with ALL of the intermediate risk arm compared with those in standard risk arm (3.44, 95% confidence interval (CI), 1.31-9.05, P = 0.012), and in carriers of the PACSIN2 rs2413739 T allele compared with those with the CC genotype (heterozygotes CT: HR, 2.32, 95% CI, 0.90-5.97, P = 0.081; and homozygous TT: HR, 4.14, 95% CI, 1.54-11.11, P = 0.005). Future studies are needed to confirm the lack of impact of TGN levels and variability on relapse in the AIEOP-BFM ALL trials, and to clarify the mechanism of PACSIN2 rs2413739 on outcome.
Collapse
Affiliation(s)
- Raffaella Franca
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Gabriele Stocco
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
- Institute for Maternal and Child Health - IRCCS Burlo Garofolo, Trieste, Italy
| | - Valentina Kiren
- Institute for Maternal and Child Health - IRCCS Burlo Garofolo, Trieste, Italy
| | - Antimo Tessitore
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Franca Fagioli
- Paediatric Onco-Haematology Department, Regina Margherita Children's Hospital, Turin, Italy
- Department of Public Health and Pediatrics, University of Turin, Turin, Italy
| | - Paola Quarello
- Paediatric Onco-Haematology Department, Regina Margherita Children's Hospital, Turin, Italy
- Department of Public Health and Pediatrics, University of Turin, Turin, Italy
| | - Nicoletta Bertorello
- Paediatric Onco-Haematology Department, Regina Margherita Children's Hospital, Turin, Italy
| | - Carmelo Rizzari
- University of Milano-Bicocca, Milan, Italy
- Pediatric Hematology Oncology Unit, MBBM Foundation, ASST Monza, Monza, Italy
| | - Antonella Colombini
- Pediatric Hematology Oncology Unit, MBBM Foundation, ASST Monza, Monza, Italy
| | - Laura Rachele Bettini
- University of Milano-Bicocca, Milan, Italy
- Pediatric Hematology Oncology Unit, MBBM Foundation, ASST Monza, Monza, Italy
| | - Franco Locatelli
- Pediatric Hematology and Oncology, IRCCS Ospedale Pediatrico Bambin Gesù, Rome, Italy
| | - Luciana Vinti
- Pediatric Hematology and Oncology, IRCCS Ospedale Pediatrico Bambin Gesù, Rome, Italy
| | - Katia Girardi
- Pediatric Hematology and Oncology, IRCCS Ospedale Pediatrico Bambin Gesù, Rome, Italy
| | - Daniela Silvestri
- Pediatric Hematology Oncology Unit, MBBM Foundation, ASST Monza, Monza, Italy
| | - Maria Grazia Valsecchi
- Bicocca Centre of Bioinformatics, Biostatistics and Bioimaging, School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Giuliana Decorti
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
- Institute for Maternal and Child Health - IRCCS Burlo Garofolo, Trieste, Italy
| | - Marco Rabusin
- Institute for Maternal and Child Health - IRCCS Burlo Garofolo, Trieste, Italy
| |
Collapse
|
7
|
Zupanec S, Herriage T, Landier W. Children's Oncology Group 2023 blueprint: Nursing discipline. Pediatr Blood Cancer 2023; 70 Suppl 6:e30575. [PMID: 37470719 PMCID: PMC10655901 DOI: 10.1002/pbc.30575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 07/21/2023]
Abstract
In contrast to other Children's Oncology Group (COG) committees, the COG nursing discipline is unique in that it provides the infrastructure necessary for nurses to support COG clinical trials and implements a research agenda aimed at scientific discovery. This hybrid focus of the discipline reflects the varied roles and expertise within pediatric oncology clinical trials nursing that encompass clinical care, leadership, and research. Nurses are broadly represented across COG disease, domain, and administrative committees, and are assigned to all clinically focused protocols. Equally important is the provision of clinical trials-specific education and training for nurses caring for patients on COG trials. Nurses involved in the discipline's evidence-based practice initiative have published a wide array of systematic reviews on topics of clinical importance to the discipline. Nurses also develop and lead research studies within COG, including stand-alone studies and aims embedded in disease/ treatment trials. Additionally, the nursing discipline is charged with responsibility for developing patient/family educational resources within COG. Looking to the future, the nursing discipline will continue to support COG clinical trials through a multifaceted approach, with a particular focus on patient-reported outcomes and health equity/disparities, and development of interventions to better understand and address illness-related distress in children with cancer.
Collapse
Affiliation(s)
- Sue Zupanec
- Hospital for Sick Children, Toronto, Ontario, Canada
| | | | - Wendy Landier
- University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
8
|
Wadhwa A, Chen Y, Hageman L, Hoppmann A, Angiolillo A, Dickens DS, Neglia JP, Ravindranath Y, Ritchey AK, Termuhlen A, Wong FL, Landier W, Bhatia S. Poverty and relapse risk in children with acute lymphoblastic leukemia: a Children's Oncology Group study AALL03N1 report. Blood 2023; 142:221-229. [PMID: 37070673 PMCID: PMC10375268 DOI: 10.1182/blood.2023019631] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/16/2023] [Accepted: 03/16/2023] [Indexed: 04/19/2023] Open
Abstract
The association between individual-level poverty and relapse in children receiving maintenance treatment for acute lymphoblastic leukemia (ALL) remains unclear. In a secondary analysis of COG-AALL03N1, we used data from US Census Bureau to categorize patients living below year-specific federal poverty thresholds, calculated using self-reported annual household income and size of household. Participants with federal poverty thresholds above 120% of their yearly household income were categorized as living in extreme poverty. Hazard of relapse was estimated using multivariable proportional subdistributional hazards regression for patients living in extreme poverty while receiving ALL maintenance therapy after adjusting for relevant predictors. Among 592 patients in this analysis, 12.3% of the patients were living in extreme poverty. After a median follow-up of 7.9 years, the cumulative incidence of relapse at 3 years from study enrollment among those living in extreme poverty was significantly higher (14.3%) than those not living in extreme poverty (7.6%). Multivariable analysis demonstrated that children living in extreme poverty had a 1.95-fold greater hazard of relapse than those not living in extreme poverty; this association was mitigated after the inclusion of race/ethnicity in the model, likely because of collinearity between race/ethnicity and poverty. A greater proportion of children living in extreme poverty were nonadherent to mercaptopurine (57.1% vs 40.9%); however, poor adherence did not completely explain the association between poverty and relapse risk. Future studies need to understand the mechanisms underlying the association between extreme poverty and relapse risk. This trial was registered at www.clinicaltrials.gov as #NCT00268528.
Collapse
Affiliation(s)
- Aman Wadhwa
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
- Division of Pediatric Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL
| | - Yanjun Chen
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Lindsey Hageman
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Anna Hoppmann
- Division of Pediatric Hematology/Oncology, University of South Carolina School of Medicine, Columbia, SC
| | | | - David S. Dickens
- Division of Pediatric Hematology/Oncology, University of Iowa, Iowa City, IA
| | - Joseph P. Neglia
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN
| | | | - A. Kim Ritchey
- Division of Hematology/Oncology, Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Amanda Termuhlen
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN
| | - F. Lennie Wong
- Department of Population Sciences, City of Hope, Duarte, CA
| | - Wendy Landier
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
- Division of Pediatric Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL
| | - Smita Bhatia
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
- Division of Pediatric Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
9
|
Zeng XL, Heneghan MB, Badawy SM. Adherence to Oral Chemotherapy in Acute Lymphoblastic Leukemia during Maintenance Therapy in Children, Adolescents, and Young Adults: A Systematic Review. Curr Oncol 2023; 30:720-748. [PMID: 36661705 PMCID: PMC9858168 DOI: 10.3390/curroncol30010056] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/26/2022] [Accepted: 12/31/2022] [Indexed: 01/08/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most common malignancy in children and young adults. Treatment is long and involves 2-3 years of a prolonged maintenance phase composed of oral chemotherapies. Adherence to these medications is critical to achieving good outcomes. However, adherence is difficult to determine, as there is currently no consensus on measures of adherence or criteria to determine nonadherence. Furthermore, there have been few studies in pediatric B-ALL describing factors associated with nonadherence. Thus, we performed a systematic review of literature on oral chemotherapy adherence during maintenance therapy in ALL following PRISMA guidelines. Published studies demonstrated various objective and subjective methods of assessing adherence without generalizable definitions of nonadherence. However, the results of these studies suggested that nonadherence to oral maintenance chemotherapy was associated with increased risk of relapse. Future studies of B-ALL therapy should utilize a uniform assessment of adherence and definitions of nonadherence to better determine the impact of nonadherence on B-ALL outcomes and identify predictors of nonadherence that could yield targets for adherence improving interventions.
Collapse
Affiliation(s)
- Xiaopei L. Zeng
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Division of Hematology, Oncology and Stem Cell Transplantation, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA
| | - Mallorie B. Heneghan
- Division of Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, University of Utah, Salt Lake City, UT 84132, USA
| | - Sherif M. Badawy
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Division of Hematology, Oncology and Stem Cell Transplantation, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA
| |
Collapse
|
10
|
Wadhwa A, Chen Y, Hageman L, Hoppmann AL, Angiolillo A, Dickens DS, Lew G, Neglia JP, Ravindranath Y, Ritchey AK, Termuhlen A, Wong FL, Landier W, Bhatia S. Body mass index during maintenance therapy and relapse risk in children with acute lymphoblastic leukemia: A Children's Oncology Group report. Cancer 2023; 129:151-160. [PMID: 36369905 PMCID: PMC10173700 DOI: 10.1002/cncr.34529] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/28/2022] [Accepted: 08/01/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND Obesity at diagnosis of childhood acute lymphoblastic leukemia (ALL) is associated with greater risk of relapse; whether this association extends to obesity during maintenance is unstudied. METHODS This study used data from AALL03N1 to calculate median body mass index (BMI) for 676 children over 6 consecutive months during maintenance therapy; BMI percentile (BMI%ile) were operationalized as normal/underweight (<85%ile), overweight/obese (85%-98%ile), and extreme obesity (≥99%ile). Hazard of relapse was estimated using multivariable proportional subdistributional hazards regression after adjusting for all relevant demographic and clinical predictors. RESULTS Median age at study enrollment was 6 years and median length of follow-up was 7.9 years. Overall, 43.3% of the cohort was underweight/normal weight, 44.8% was overweight/obese, and 11.8% had extreme obesity. Cumulative incidence of relapse at 4 years from study enrollment was higher among those with extreme obesity (13.6% ± 4.5%) compared to those with underweight/normal weight (9.0% ± 2.1%). Multivariable analysis revealed that children with extreme obesity had a 2.4-fold (95% confidence interval [CI], 1.1-5.0; p = .01) greater hazard of relapse compared to those who were underweight/normal weight. Overweight/obese patients were at comparable risk to those who were underweight/normal weight (hazard ratio, 0.8; 95% CI, 0.4-1.6). Erythrocyte thioguanine nucleotide (TGN) levels were significantly lower among children with extreme obesity compared to those with underweight/normal weight (141.6 vs. 168.8 pmol/8 × 108 erythrocytes; p = .0002), however, the difference in TGN levels did not explain the greater hazard of relapse among those with extreme obesity. CONCLUSIONS Extreme obesity during maintenance therapy is associated with greater hazard of relapse in children with ALL. Underlying mechanisms of this association needs further investigation. LAY SUMMARY Findings from this study demonstrate that extreme obesity during maintenance therapy is associated with a greater hazard of relapse among children with acute lymphoblastic leukemia. We show that children with obesity have lower levels of erythrocyte thioguanine nucleotides even after adjusting for adherence to oral chemotherapy. However, these lower levels do not explain the greater hazard of relapse, paving the way for future studies to explore this association.
Collapse
Affiliation(s)
- Aman Wadhwa
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Division of Pediatric Hematology/Oncology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Yanjun Chen
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Lindsey Hageman
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Anna L. Hoppmann
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Division of Pediatric Hematology/Oncology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Anne Angiolillo
- Division of Pediatric Hematology/Oncology, Children’s National Medical Center, Washington, District of Columbia, USA
| | - David S. Dickens
- Division of Pediatric Hematology/Oncology, University of Iowa, Iowa City, Iowa, USA
| | - Glen Lew
- Division of Pediatric Hematology/Oncology, Children’s Healthcare of Atlanta, Emory University, Atlanta, Georgia, USA
| | - Joseph P. Neglia
- Division of Pediatric Hematology/Oncology, University of Minnesota Masonic Children’s Hospital, Minnesota, Minnesota, USA
| | | | - A. Kim Ritchey
- Division of Hematology/Oncology, Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Amanda Termuhlen
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - F. Lennie Wong
- Department of Population Sciences, City of Hope, Duarte, California, USA
| | - Wendy Landier
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Division of Pediatric Hematology/Oncology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Smita Bhatia
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Division of Pediatric Hematology/Oncology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
11
|
Kahn JM, Stevenson K, Beauchemin M, Koch VB, Cole PD, Welch JJG, Gage-Bouchard E, Karsenty C, Silverman LB, Kelly KM, Bona K. Oral Mercaptopurine Adherence in Pediatric Acute Lymphoblastic Leukemia: A Survey Study From the Dana-Farber Cancer Institute Acute Lymphoblastic Leukemia Consortium. JOURNAL OF PEDIATRIC HEMATOLOGY/ONCOLOGY NURSING 2023; 40:17-23. [PMID: 36221984 PMCID: PMC9982234 DOI: 10.1177/27527530221122685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Background: Oral chemotherapy nonadherence is a challenge in clinical oncology. During therapy for acute lymphoblastic leukemia (ALL), poor adherence to 6-mercaptopurine (6MP) increases relapse risk. Clinically significant nonadherence is reported in 30% of children treated for ALL on Children's Oncology Group (COG) trials. Whether nonadherence rates vary across regimens with different treatment schedules and modes of administration is unknown. Methods: We conducted an exploratory, cross-sectional survey study on parents of children (1-18 years) receiving continuation therapy on, or as per Dana-Farber Cancer Institute (DFCI) ALL Consortium Protocol 11-001. Treatment required weekly visits to the clinic and 14 days of oral 6MP every 3 weeks. Survey assessed self-reported sociodemographics, medication-taking, chemotherapy comprehension, and 6MP adherence; adherence survey items were developed from published surveys. Patients were grouped as nonadherent if they endorsed missing one 6MP dose during the last cycle, or more than one dose during prior cycles, for nonmedical reasons. Results: Sixty-two families completed the surveys, all of whom had evaluable adherence data. In total, 25% of patients met the study definition of nonadherence. Twenty-three percent reported that it was "not easy" to follow administration guidelines around the dairy intake and 57% requested more teaching and educational resources. Conclusion: Self-reported nonadherence to oral 6MP in the DFCI ALL Consortium is high, with rates similar to those observed in the COG. This suggests that the additional contact during weekly infusions on the DFCI is insufficient to address barriers affecting oral chemotherapy adherence.
Collapse
Affiliation(s)
- Justine M. Kahn
- Division of Pediatric Hematology, Oncology and Stem Cell
Transplantation, Columbia University Irving Medical
Center, New York, NY, USA
| | - Kristen Stevenson
- Department of Biostatistics and
Computational Biology, Dana-Farber Cancer Institute, Harvard Medical
School, Boston, MA, USA
| | - Melissa Beauchemin
- Division of Pediatric Hematology, Oncology and Stem Cell
Transplantation, Columbia University Irving Medical
Center, New York, NY, USA
- Columbia University School of Nursing, New York, NY, USA
| | - Victoria B. Koch
- Department of Pediatric Oncology,
Dana-Farber Cancer Institute, Boston, MA, USA
| | - Peter D. Cole
- Rutgers Cancer Institute of New
Jersey, New Brunswick, NJ, USA
| | - Jennifer J. G. Welch
- Pediatric Hematology Oncology, Hasbro Children’s Hospital/Brown University, Providence, RI, USA
| | - Elizabeth Gage-Bouchard
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer
Center, Buffalo, NY, USA
| | - Cecile Karsenty
- Department of Pediatrics, Baylor College of Medicine, Houston, TX,
USA
- Texas Children’s Cancer and Hematology
Centers, Texas Children’s Hospital, Houston, TX, USA
| | - Lewis B. Silverman
- Department of Pediatric Oncology,
Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kara M. Kelly
- Department of Pediatrics, Roswell Park Comprehensive Cancer
Center, Buffalo, NY, USA
| | - Kira Bona
- Department of Pediatric Oncology,
Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
12
|
Saultier P, Simonin M, Beaumais TAD, Rialland F, Alby-Laurent F, Lubnau M, Desplantes C, Jacqz-Aigrain E, Rohrlich P, Reguerre Y, Rabian F, Sirvent N, Plat GW, Petit A. [Practical management during maintenance therapy of pediatric acute lymphoblastic leukemia: Recommendations of the French Society for Childhood and Adolescent Cancer and Leukemia (SFCE)]. Bull Cancer 2022; 109:1132-1143. [PMID: 35863954 DOI: 10.1016/j.bulcan.2022.05.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/17/2022] [Accepted: 05/31/2022] [Indexed: 11/24/2022]
Abstract
Maintenance therapy is the last phase of treatment for acute lymphoblastic leukemia in children and adolescents. Although maintenance therapy is associated with toxicities and specific management issues, it is an essential phase of treatment that reduces the risk of relapse. The objective of this work is to propose a guide for the initiation, administration, and monitoring of maintenance therapy, and for the management of food, schooling, leisure, community life, risk of infection and links with family medicine.
Collapse
Affiliation(s)
- Paul Saultier
- Hôpital de la Timone Enfants, APHM, service d'hématologie, immunologie et oncologie pédiatrique, Marseille, France.
| | - Mathieu Simonin
- AP-HP, hôpital Armand-Trousseau, Sorbonne université, service d'hématologie et oncologie pédiatrique, Paris, France
| | | | - Fanny Rialland
- CHU de Nantes, service d'onco-hématologie pédiatrique, Nantes, France
| | - Fanny Alby-Laurent
- AP-HP, hôpital Armand-Trousseau, Sorbonne université, service d'hématologie et oncologie pédiatrique, Paris, France
| | - Marion Lubnau
- CHU de Nancy, service d'onco-hématologie pédiatrique, Nancy, France
| | | | - Evelyne Jacqz-Aigrain
- AP-HP, hôpital Saint-Louis, département de pharmacologie et pharmacogénétique, Paris, France
| | - Pierre Rohrlich
- CHU de Nice, service d'hématologie pédiatrique, Nice, France
| | - Yves Reguerre
- CHU de la Réunion, service d'hémato-oncologie pédiatrique, Réunion, France
| | - Florence Rabian
- AP-HP, hôpital Saint-Louis, service d'hématologie adolescents et jeunes adultes, Paris, France
| | - Nicolas Sirvent
- CHU de Montpellier, service d'hématologie et oncologie pédiatrique, Montpellier, France
| | - Geneviève Willson Plat
- CHU de Toulouse, service d'hématologie oncologie et immunologie pédiatrique, Toulouse, France
| | - Arnaud Petit
- AP-HP, hôpital Armand-Trousseau, Sorbonne université, service d'hématologie et oncologie pédiatrique, Paris, France
| |
Collapse
|
13
|
Pyke-Grimm KA, Franck LS, Halpern-Felsher B, Goldsby RE, Rehm RS. Day-to-Day Decision Making by Adolescents and Young Adults with Cancer. JOURNAL OF PEDIATRIC HEMATOLOGY/ONCOLOGY NURSING 2022; 39:290-303. [PMID: 35538622 PMCID: PMC9807778 DOI: 10.1177/27527530211068718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Background: Adolescents and young adults (AYAs) with cancer must negotiate the transition between childhood and adulthood while dealing with a life-threatening illness. AYA involvement in decision making varies depending on the type of decision and when decisions occur during treatment, and evidence suggests that AYAs want to be involved in decision making. Objective: To explore involvement of AYAs with cancer in day-to-day decisions affected by their cancer and treatment. Methods: This qualitative study used interpretive focused ethnography within the sociologic tradition, informed by symbolic interactionism. Semi-structured interviews and informal participant observation took place at two quaternary pediatric oncology programs. Results: Thirty-one interviews were conducted with 16 AYAs ages 15 to 20 years. Major day to day decision-making categories identified included: (1) mental mindset, (2) self-care practices, (3) self-advocacy, and (4) negotiating relationships. Participants described how they came to grips with their illness early on and decided to fight their cancer. They described decisions they made to protect their health, how they advocated for themselves and decisions they made about relationships with family and friends. Conclusions: Through day-to-day decisions, participants managed the impact of cancer and its treatment on their daily lives. Research should focus on developing and implementing interventions to empower AYAs to participate in day-to-day decisions that will affect how they manage their cancer, its treatment and ultimately their outcomes. Implications for Practice: Healthcare providers can facilitate AYA's participation in day-to-day decision making through encouraging autonomy and self-efficacy by providing support and through effective communication.
Collapse
Affiliation(s)
- Kimberly A. Pyke-Grimm
- Stanford Children's Health, Department of Nursing Research and
Evidence-Based Practice, Palo Alto, CA, USA,Division of Hematology/Oncology, Department of Pediatrics, Stanford
University School of Medicine,Department of Family Health Care Nursing, San Francisco School of
Nursing, University of California, San Francisco, CA, USA,Kimberly A. Pyke-Grimm, PhD, RN, CNS,
CPHON, Department of Nursing Research and Evidence-Based Practice, Stanford
Children's Health, 750 Welch Road, Palo Alto, CA 94304, USA.
| | - Linda S. Franck
- Department of Family Health Care Nursing, San Francisco School of
Nursing, University of California, San Francisco, CA, USA
| | - Bonnie Halpern-Felsher
- Department of Pediatrics, Division of Adolescent Medicine, Stanford University, School of Medicine, Stanford, CA, USA
| | | | - Roberta S. Rehm
- Department of Family Health Care Nursing, San Francisco School of
Nursing, University of California, San Francisco, CA, USA,*Professor Emeritus
| |
Collapse
|
14
|
Maintenance therapy for acute lymphoblastic leukemia: basic science and clinical translations. Leukemia 2022; 36:1749-1758. [PMID: 35654820 PMCID: PMC9252897 DOI: 10.1038/s41375-022-01591-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 01/21/2023]
Abstract
Maintenance therapy (MT) with oral methotrexate (MTX) and 6-mercaptopurine (6-MP) is essential for the cure of acute lymphoblastic leukemia (ALL). MTX and 6-MP interfere with nucleotide synthesis and salvage pathways. The primary cytotoxic mechanism involves the incorporation of thioguanine nucleotides (TGNs) into DNA (as DNA-TG), which may be enhanced by the inhibition of de novo purine synthesis by other MTX/6-MP metabolites. Co-medication during MT is common. Although Pneumocystis jirovecii prophylaxis appears safe, the benefit of glucocorticosteroid/vincristine pulses in improving survival and of allopurinol to moderate 6-MP pharmacokinetics remains uncertain. Numerous genetic polymorphisms influence the pharmacology, efficacy, and toxicity (mainly myelosuppression and hepatotoxicity) of MTX and thiopurines. Thiopurine S-methyltransferase (encoded by TPMT) decreases TGNs but increases methylated 6-MP metabolites (MeMPs); similarly, nudix hydrolase 15 (encoded by NUDT15) also decreases TGNs available for DNA incorporation. Loss-of-function variants in both genes are currently used to guide MT, but do not fully explain the inter-patient variability in thiopurine toxicity. Because of the large inter-individual variations in MTX/6-MP bioavailability and metabolism, dose adjustments are traditionally guided by the degree of myelosuppression, but this does not accurately reflect treatment intensity. DNA-TG is a common downstream metabolite of MTX/6-MP combination chemotherapy, and a higher level of DNA-TG has been associated with a lower relapse hazard, leading to the development of the Thiopurine Enhanced ALL Maintenance (TEAM) strategy-the addition of low-dose (2.5-12.5 mg/m2/day) 6-thioguanine to the 6-MP/MTX backbone-that is currently being tested in a randomized ALLTogether1 trial (EudraCT: 2018-001795-38). Mutations in the thiopurine and MTX metabolism pathways, and in the mismatch repair genes have been identified in early ALL relapses, providing valuable insights to assist the development of strategies to detect imminent relapse, to facilitate relapse salvage therapy, and even to bring about changes in frontline ALL therapy to mitigate this relapse risk.
Collapse
|
15
|
Tang N, Kovacevic A, Zupanec S, Sivananthan A, Patel R, Patel P, Vennettilli A, Paw Cho Sing E, Alexander S, Sung L, Dupuis LL. Perceptions of parents of pediatric patients with acute lymphoblastic leukemia on oral chemotherapy administration: A qualitative analysis. Pediatr Blood Cancer 2022; 69:e29329. [PMID: 34492743 DOI: 10.1002/pbc.29329] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 08/22/2021] [Indexed: 01/12/2023]
Abstract
OBJECTIVE To describe the experiences and perspectives of parents of pediatric patients with acute lymphoblastic leukemia (ALL) regarding oral chemotherapy administration during maintenance therapy. METHODS English-speaking parents of patients 4 to <18 years who were receiving ALL maintenance oral chemotherapy were eligible to participate in this mixed methods study. Using semi-structured interviews, we asked participants how difficult they found oral chemotherapy administration. We also probed regarding barriers and facilitators of oral chemotherapy administration and strategies used to overcome challenges. Lastly, we asked participants for their advice to future parents giving oral chemotherapy to their children. RESULTS Twenty-three participants were interviewed. One-fifth of participants stated that oral chemotherapy administration at home was hard or very hard. Common factors influencing oral chemotherapy administration were product-related (e.g., formulation) and treatment-related adverse effects (e.g., nausea), lifestyle adjustment (e.g., fitting in with family schedule), and attitudes (e.g., onus of medication administration). Strategies to address oral chemotherapy administration included several administration techniques, scheduling of medication administration, and normalization of medication taking. CONCLUSIONS Oral chemotherapy administration during ALL maintenance therapy was hard for some parents. Identification of these parents and discussion of strategies to facilitate adherence to oral chemotherapy regimens may optimize patient outcomes.
Collapse
Affiliation(s)
- Natalie Tang
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Anja Kovacevic
- Department of Pharmacy, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Sue Zupanec
- Department of Nursing, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Araby Sivananthan
- Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Rikesh Patel
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.,Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Priya Patel
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.,Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Ashlee Vennettilli
- Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Edric Paw Cho Sing
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.,Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Sarah Alexander
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Lillian Sung
- Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada.,Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - L Lee Dupuis
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.,Department of Pharmacy, The Hospital for Sick Children, Toronto, Ontario, Canada.,Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
16
|
Parents' Experiences with Home-Based Oral Chemotherapy Prescribed to a Child Diagnosed with Acute Lymphoblastic Leukemia: A Qualitative Study. Curr Oncol 2021; 28:4377-4391. [PMID: 34898538 PMCID: PMC8628767 DOI: 10.3390/curroncol28060372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 11/16/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most common type of cancer in children. Treatment includes home-based oral chemotherapies (OCs) (e.g., 6-mercaptopurine and dexamethasone) taken for 2 to 3 years. The management of OC can be challenging for children and their parents. However, the multifaceted experience of families with children taking OC for ALL is largely undescribed. We report the experience with these OCs from the parents’ perspective. We conducted a qualitative descriptive study. Semi-structured interviews were conducted with the parents of children with ALL aged < 15 years, followed in a specialized university-affiliated center. The interviews were fully transcribed and thematically analyzed. Thirteen of the seventeen eligible parents (76.5%) participated in the study. The parents’ motivation to follow the recommendations provided by the multidisciplinary care team regarding OC was very high. The quantity and the quality of the information received were judged adequate, and the parents reported feeling knowledgeable enough to take charge of the OC at home. Adapting to the consequences of OC on family daily life was collectively identified as the biggest challenge. This includes developing and maintaining a strict daily routine, adapting to the child’s neurobehavioral changes during dexamethasone days and adapting family social life. Our findings have several implications for enhancing the support offered to families with home-based OC for ALL. Supportive interventions should consider the family as a whole and their needs should be regularly monitored. Specific attention should be paid to the development and maintenance of a routine, to the parental burden, and to the emotional impact, especially regarding dexamethasone.
Collapse
|
17
|
Franca R, Braidotti S, Stocco G, Decorti G. Understanding thiopurine methyltransferase polymorphisms for the targeted treatment of hematologic malignancies. Expert Opin Drug Metab Toxicol 2021; 17:1187-1198. [PMID: 34452592 DOI: 10.1080/17425255.2021.1974398] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Thiopurine methyltransferase (TPMT) catalyzes the S-methylation of thiopurines (mercaptopurine (MP) and tioguanine (TG)), chemotherapeutic agents used in the treatment of acute lymphoblastic leukemia (ALL). Polymorphisms in TPMT gene encode diminished activity enzyme, enhancing accumulation of active metabolites, and partially explaining the inter-individual differences in patients' clinical response. AREAS COVERED This review gives an overview on TPMT gene and function, and discusses the pharmacogenomic implications of TPMT variants in the prevention of severe thiopurine-induced hematological toxicities and the less known implication on TG-induced sinusoidal obstruction syndrome. Additional genetic and non-genetic factors impairing TPMT activity are considered. Literature search was done in PubMed for English articles published since1990, and on PharmGKB. EXPERT OPINION To titrate thiopurines safely and effectively, achieve the right degree of lymphotoxic effect and avoid excessive myelosuppression, the optimal management will combine a preemptive TPMT genotyping to establish a safe initial dose with a close phenotypic monitoring of TPMT activity and/or of active metabolites during long-term treatment. Compared to current ALL protocols, replacement of TG by MP during reinduction phase in TPMT heterozygotes and novel individualized TG regimens in maintenance for TPMT wild-type subjects could be investigated to improve outcomes while avoiding risk of severe hepatotoxicity.
Collapse
Affiliation(s)
- R Franca
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - S Braidotti
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - G Stocco
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - G Decorti
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy.,Institute for Maternal & Child Health (I.r.c.c.s) Burlo Garofolo, Trieste, Italy
| |
Collapse
|
18
|
Hoppmann AL, Chen Y, Landier W, Hageman L, Evans WE, Wong FL, Relling MV, Bhatia S. Individual prediction of nonadherence to oral mercaptopurine in children with acute lymphoblastic leukemia: Results from COG AALL03N1. Cancer 2021; 127:3832-3839. [PMID: 34161608 DOI: 10.1002/cncr.33760] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/06/2021] [Accepted: 04/26/2021] [Indexed: 11/06/2022]
Abstract
BACKGROUND Poor mercaptopurine (6MP) adherence (mean adherence rate < 90%) increases the relapse risk among children with acute lymphoblastic leukemia (ALL). 6MP adherence remains difficult to measure in real time. Easily measured patient-level factors could identify patients at risk for poor adherence. METHODS The authors measured 6MP adherence via electronic monitoring for 6 months per patient. Using data from month 3, they created a risk prediction model for 6MP nonadherence in 407 children with ALL (mean age, 7.7 ± 4.4 years); they used receiver operating characteristic analyses in the training set (n = 250) and replicated this in the test set (n = 157). RESULTS Age, race/ethnicity, 6MP dose intensity, absolute neutrophil count, 6MP ingestion patterns, and household structure were retained in the prediction model. The model yielded areas under the receiver operating characteristic curve (AUCs) of 0.79 (95% confidence interval [CI], 0.71-0.85) and 0.74 (95% CI, 0.63-0.85) in the training and test sets, respectively. The model performed better for those who were ≥12 years old (AUC, 0.79; 95% CI, 0.59-0.99) than those <12 years old (AUC, 0.70; 95% CI, 0.58-0.81). Using the predicted probability of nonadherence based on receiver operating characteristic analysis, the authors developed a binary risk classifier to classify patients with a high or low probability of nonadherence. The sensitivity and specificity of the binary risk classifier were 71% and 76%, respectively. Adjusted for clinical prognosticators, the risk of relapse was 2.2-fold higher (95% CI, 0.94-5.1; P = .07) among patients with a high probability of nonadherence in comparison with those with a low probability, as identified by the risk prediction model. CONCLUSIONS The risk prediction model identified patients with a high probability of nonadherence and could be used in real time to personalize recommendations and interventions in the clinic. LAY SUMMARY The vast majority of children with acute lymphoblastic leukemia, the most common childhood cancer, are cured. The treatment of acute lymphoblastic leukemia includes taking an oral chemotherapy medicine (mercaptopurine) for approximately 2 years. Children who miss doses of this medicine (specifically children who take the medicine less than 90% of the time that it is prescribed) are more likely to suffer leukemia relapse. The authors of this article have measured mercaptopurine adherence with electronic bottle caps to determine characteristics of patients that predict nonadherence, and they have created a prediction tool that could allow physicians to identify and intervene with patients at high risk of nonadherence.
Collapse
Affiliation(s)
- Anna L Hoppmann
- Institute for Cancer Outcomes and Survivorship, Division of Pediatric Hematology and Oncology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Yanjun Chen
- Institute for Cancer Outcomes and Survivorship, Division of Pediatric Hematology and Oncology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Wendy Landier
- Institute for Cancer Outcomes and Survivorship, Division of Pediatric Hematology and Oncology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Lindsey Hageman
- Institute for Cancer Outcomes and Survivorship, Division of Pediatric Hematology and Oncology, University of Alabama at Birmingham, Birmingham, Alabama
| | | | | | - Mary V Relling
- St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Smita Bhatia
- Institute for Cancer Outcomes and Survivorship, Division of Pediatric Hematology and Oncology, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
19
|
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
|
20
|
Abstract
The last decade has witnessed great advances in our understanding of the genetic and biological basis of childhood acute lymphoblastic leukemia (ALL), the development of experimental models to probe mechanisms and evaluate new therapies, and the development of more efficacious treatment stratification. Genomic analyses have revolutionized our understanding of the molecular taxonomy of ALL, and these advances have led the push to implement genome and transcriptome characterization in the clinical management of ALL to facilitate more accurate risk-stratification and, in some cases, targeted therapy. Although mutation- or pathway-directed targeted therapy (e.g., using tyrosine kinase inhibitors to treat Philadelphia chromosome [Ph]-positive and Phlike B-cell-ALL) is currently available for only a minority of children with ALL, many of the newly identified molecular alterations have led to the exploration of approaches targeting deregulated cell pathways. The efficacy of cellular or humoral immunotherapy has been demonstrated with the success of chimeric antigen receptor T-cell therapy and the bispecific engager blinatumomab in treating advanced disease. This review describes key advances in our understanding of the biology of ALL and optimal approaches to risk-stratification and therapy, and it suggests key areas for basic and clinical research.
Collapse
Affiliation(s)
- Hiroto Inaba
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN; Hematological Malignancies Program, St. Jude Children's Research Hospital, Memphis, TN.
| | - Charles G Mullighan
- Hematological Malignancies Program, St. Jude Children's Research Hospital, Memphis, TN; Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN.
| |
Collapse
|
21
|
Karol SE, Yang JJ. Pharmacogenomics and ALL treatment: How to optimize therapy. Semin Hematol 2020; 57:130-136. [PMID: 33256902 DOI: 10.1053/j.seminhematol.2020.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 01/28/2023]
Abstract
Inherited genetic variations may alter drug sensitivity in patients with acute lymphoblastic leukemia, predisposing to adverse treatment side effects. In this review, we discuss evidence from children and young adults with acute lymphoblastic leukemia to review the available pharmacogenomic data with an emphasis on clinically actionable and emerging discoveries, for example, genetic variants in thiopurine methyltransferase and NUDT15 that alter 6-mercaptopurine dosing. We also highlight the need for ongoing pharmacogenomic research to validate the significance of recent findings. Further research in young adults, as well as with novel therapeutics, is needed to provide optimal therapy in future trials.
Collapse
Affiliation(s)
- Seth E Karol
- Departments of Oncology, St. Jude Children's Research Hospital, Memphis, TN.
| | - Jun J Yang
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN
| |
Collapse
|
22
|
Conneely SE, Cooper SL, Rau RE. Use of Allopurinol to Mitigate 6-Mercaptopurine Associated Gastrointestinal Toxicity in Acute Lymphoblastic Leukemia. Front Oncol 2020; 10:1129. [PMID: 32766146 PMCID: PMC7378397 DOI: 10.3389/fonc.2020.01129] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 06/05/2020] [Indexed: 01/20/2023] Open
Abstract
An essential component of acute lymphoblastic leukemia (ALL) therapy is the prolonged maintenance phase with daily 6-mercaptopurine (6-MP) as the cornerstone. While 6-MP is generally well-tolerated, some patients suffer from significant side effects such as gastrointestinal (GI) toxicity, including hepatitis, hypoglycemia, nausea, and pancreatitis, which can substantially limit the tolerated dose of 6-MP. These toxicities are thought to result from skewed metabolism of 6-MP leading to an accumulation of the 6-methylmercaptopurine (6-MMP) metabolite. Here, we describe current knowledge behind the use of allopurinol to modify 6-MP metabolism and improve tolerance to therapy. This method has been successfully used in adults with inflammatory bowel disease refractory to purine therapy and has been modified for use in children with GI toxicities related to 6-MP in maintenance therapy for ALL. Use of allopurinol for 6-MP related toxicities should be reserved for patients in which an alternative cause of signs or symptoms has been excluded and for whom non-pharmacologic measures have failed. When allopurinol is used, simultaneous dose reduction of 6-MP is required to avoid severe myelosuppression and related side effects, though overall combination therapy appears to be well-tolerated and effective when instituted appropriately.
Collapse
Affiliation(s)
- Shannon E Conneely
- Department of Pediatric Hematology and Oncology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, United States
| | - Stacy L Cooper
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Rachel E Rau
- Department of Pediatric Hematology and Oncology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, United States
| |
Collapse
|
23
|
Wiernikowski JT, Bernhardt MB. Review of nutritional status, body composition, and effects of antineoplastic drug disposition. Pediatr Blood Cancer 2020; 67 Suppl 3:e28207. [PMID: 32083372 DOI: 10.1002/pbc.28207] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 01/22/2020] [Accepted: 01/23/2020] [Indexed: 12/22/2022]
Abstract
The overall survival for children with cancer in high income countries is excellent. However, there are many disparities that may negatively affect survival, which are particularly problematic in low income countries, such as nutritional status at diagnosis and throughout therapy. Nutritional status as well as concomitant foods, supplements, and medications may play a role in overall exposure and response to chemotherapy. Emerging science around the microbiome may also play a role and should be further explored as a contributor to disease progression and therapeutic response. This article highlights some of these issues and proposes additional areas of research relevant to nutritional status and pharmacology that are needed in pediatric oncology.
Collapse
Affiliation(s)
- John T Wiernikowski
- Division of Paediatric Haematology/Oncology, Department of Paediatrics, McMaster Children's Hospital, McMaster University, Hamilton, Ontario, Canada
| | - Melanie Brooke Bernhardt
- Section of Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
24
|
Ramanathan S, Narula G, Prasad M, Vora T, Chinnaswamy G, Banavali S. Parvoviral disease in childhood cancer: Clinical outcomes and impact on therapy at a tertiary cancer center in India. Pediatr Blood Cancer 2018; 65:e27357. [PMID: 30058287 DOI: 10.1002/pbc.27357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 06/16/2018] [Accepted: 06/18/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIM Parvovirus-B19 disease in immunocompromised children can cause myelosuppression and therapeutic delays. We studied the clinical profiles of children having symptoms suggestive of parvoviral disease at our institution, a large tertiary cancer center. METHODS Children below age 15 years undergoing treatment for malignancies with clinical features suggestive of parvoviral infection, and/or unexplained drop in hemoglobin, and/or prolonged cytopenia were screened for parvovirus infection using DNA-PCR for parvovirus-B19 (PB19) in the peripheral blood. Patients testing positive from September 2014 till February 2017 were studied. RESULTS Of the 59 patients (36 patients with hematolymphoid malignancies, 23 with solid tumors) screened for suspected parvoviral infections, 27 tested positive. Median age was 9.6 years (2.25-15 years), 18 (66%) had hematolymphoid malignancies, while 7 (33%) had solid tumors. Six patients (26%) were on intensive phases, 16 (60%) patients developed the symptoms during maintenance chemotherapy, and 4 (15%) after completion of therapy. Isolated anemia was the commonest feature seen in 10 patients (37%) while bicytopenia and pancytopenia were noticed in 8 (30%) and 9 (33%) patients respectively. Fifty percent of patients those who received rituximab (3/6) developed persistent parvoviremia (>4 weeks) as compared with 24% (5/21) of those who did not. Two patients (7%) developed hemophagocytic lymphohistiocytosis (HLH). Treatment delay by more than 14 days was encountered in a majority (62%), with 5 patients requiring treatment modification or even suspension. CONCLUSIONS Parvoviral infection in children who are on or have recently completed chemotherapy can lead to multiple cytopenias and significant treatment delays. Rituximab exposure may lead to persistent parvoviral disease (p < 0.05). HLH, though occasional, can be a serious complication.
Collapse
Affiliation(s)
- Subramaniam Ramanathan
- Department of Pediatric Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India.,Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Gaurav Narula
- Department of Pediatric Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India.,Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Maya Prasad
- Department of Pediatric Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India.,Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Tushar Vora
- Department of Pediatric Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India.,Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Girish Chinnaswamy
- Department of Pediatric Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India.,Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Shripad Banavali
- Department of Pediatric Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India.,Homi Bhabha National Institute, Mumbai, Maharashtra, India
| |
Collapse
|
25
|
Acute lymphoblastic leukaemia in 2017: Immunotherapy for ALL takes the world by storm. Nat Rev Clin Oncol 2017; 15:69-70. [PMID: 29255238 DOI: 10.1038/nrclinonc.2017.176] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
26
|
Alsous M, Abu Farha R, Alefishat E, Al Omar S, Momani D, Gharabli A, McElnay J, Horne R, Rihani R. Adherence to 6-Mercaptopurine in children and adolescents with Acute Lymphoblastic Leukemia. PLoS One 2017; 12:e0183119. [PMID: 28877179 PMCID: PMC5587295 DOI: 10.1371/journal.pone.0183119] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 07/28/2017] [Indexed: 01/17/2023] Open
Abstract
Objective Studies on children with Acute Lymphoblastic Leukemia (ALL) reported non-adherence in 2–54% of cases. The primary objective of this study was to assess rates of adherence to 6-MP using two different methods in children and adolescents with ALL. Secondary aim was to identify factors that influence adherence to 6-MP in children with ALL. Methods All eligible children with ALL who are (≤ 19) years old and receive 6-MP therapy for at least 1 month were approached to participate in the study. A total of 52 children with ALL and their primary caregivers were recruited. Adherence measures included an objective method (measuring 6-MP metabolites in packed Red Blood Cells (RBCs)) and a subjective method (using parent and child self-report via the Medication Adherence Report Scale; MARS; Adherence was defined as 90% or greater). Results Rates of adherence varied across the measurement methods. Packed RBCs sample analysis indicated forty-four patients (84.6%) to be adherent. Using the MARS questionnaires, a total of 49 children (94.2%) were classified as being adherent according to the parental MARS questionnaire scores, while all the 15 children (100%) who answered the MARS (child) questionnaire were classified as adherent. Overall adherence rate was 80.8% within the studied population. Conclusion MARS scale was shown to overestimate adherence compared to measurement of 6-MP metabolites in the blood. A combination of both methods led to increased detection of non-adherence to thiopurine in children with ALL.
Collapse
Affiliation(s)
- Mervat Alsous
- Department of Clinical Pharmacy and Therapeutics, Faculty of Pharmacy, Applied Science Private University, Amman, Jordan
- * E-mail:
| | - Rana Abu Farha
- Department of Clinical Pharmacy and Therapeutics, Faculty of Pharmacy, Applied Science Private University, Amman, Jordan
| | - Eman Alefishat
- Department of Biopharmaceutics and Clinical Pharmacy, Faculty of Pharmacy, the University of Jordan, Amman, Jordan
| | - Suha Al Omar
- Clinical Pharmacy Department, King Hussein Cancer Center, Amman, Jordan
| | - Deema Momani
- Clinical Pharmacy Department, King Hussein Cancer Center, Amman, Jordan
| | - Alia Gharabli
- Clinical Pharmacy Department, King Hussein Cancer Center, Amman, Jordan
| | - James McElnay
- Clinical and Practice Research Group, School of Pharmacy, Queen’s University Belfast, Belfast, United Kingdom
| | - Robert Horne
- Centre for Behavioural Medicine, UCL School of Pharmacy, University College London, London, United Kingdom
| | - Rawad Rihani
- Department of Paediatric Oncology, Paediatric Bone Marrow and Stem Cell Transplantation, King Hussein Cancer Center, Amman, Jordan
| |
Collapse
|