CC BY 4.0 · Indian J Med Paediatr Oncol 2024; 45(02): 147-152
DOI: 10.1055/s-0043-1774780
Original Article

Hepatic Dysfunction during Induction Chemotherapy in Children with Acute Lymphoblastic Leukemia and Lymphoblastic Lymphoma and Its Effects on Subsequent Therapy and Outcome

Nalla Anuraag Reddy
1   Paediatric Hematology and Oncology Department, KMC Hospital, Mangalore, Karnataka, India
,
Keerthi Raj
2   Department of Paediatrics, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
,
Harsha Prasada Lashkari
2   Department of Paediatrics, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
› Author Affiliations
Funding None declared.

Abstract

Introduction The overall survival rate for childhood acute lymphoblastic leukemia (ALL) and lymphoblastic lymphoma (LBL) has shown tremendous growth in the recent years. Hepatic dysfunction is one of the complications seen during therapy and can add to the underlying morbidity of the disease, delay in chemotherapy, modification of drugs, and rarely fulminant hepatic failure.

Objective This article aims to find out the prevalence of hepatic dysfunction during induction chemotherapy for ALL and LBL.

Materials and Methods This was a retrospective study, where the data of all children between 1 and 18 years of age with ALL and LBL treated at our center as per the UK-ALL 2003 protocol between December 2013 and December 2021 have been included from the medical records. Hepatic dysfunction was defined as grade 3 and 4 alanine transaminase (ALT) and aspartate transaminase (AST) levels as per Common Terminology Criteria for Adverse Events v5.0 and hyperbilirubinemia as ≥ 1.4 mg/dL as the chemotherapy modification begins at this cutoff. Data from children with hepatic dysfunction was compared with those without hepatic dysfunction using chi-squared test and Student's t-tests. Those variables with a p-value of < 0.2 were analyzed with multivariate regression analysis. Kaplan–Meier survival estimates were used to calculate the event-free survival (EFS).

Results A total of 142 children were included in the study. Thirty-one (21.8%) children developed hepatic dysfunction, 14 (9.9%) of them with ALT/AST elevation and 27 (19%) with bilirubin elevation. Weight (mean 25 ± 13.5, p 0.01), body surface area (mean 0.87 ± 0.29, p 0.02), and National Cancer Institute high risk (p 0.005) were associated with hepatic dysfunction in univariate analysis but none of them were significant in multivariate regression analysis. Treatment modification was required in 14/31 children with hepatic dysfunction. Death in induction was more among children with hepatic dysfunction (p < 0.001). There was no significant impact on minimal residual disease outcomes. Five-year EFS (death or relapse) was 59.93 ± 9% in children with hepatic dysfunction as opposed to 72 ± 5.0% in those without hepatic dysfunction (95% confidence interval, p = 0.07).

Conclusion One in five children with ALL and LBL on induction therapy developed hepatic dysfunction. Almost half of those with hepatic dysfunction required chemotherapy modifications.

Patient Consent

None declared.


Authorship

The manuscript has been read and approved by all the authors, that the requirements for authorship have been met, and each author believes that the manuscript represents honest work.


Authors' Contributions

N.A.R.: Contributed to conception of design, literature search, data collection, data and statistical analysis, manuscript writing, editing, and manuscript review.


K.R.: Contributed to data collection, literature search, and manuscript review.


H.P.L.: Contributed to conception of design, data and statistical analysis, manuscript editing, and manuscript review.


Sources of Support

None declared.




Publication History

Article published online:
27 February 2024

© 2024. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)

Thieme Medical and Scientific Publishers Pvt. Ltd.
A-12, 2nd Floor, Sector 2, Noida-201301 UP, India

 
  • References

  • 1 Arora RS, Arora B. Acute leukemia in children: a review of the current Indian data. South Asian J Cancer 2016; 5 (03) 155-160
  • 2 Hunger SP, Lu X, Devidas M. et al. Improved survival for children and adolescents with acute lymphoblastic leukemia between 1990 and 2005: a report from the Children's Oncology Group. J Clin Oncol 2012; 30 (14) 1663-1669
  • 3 Teachey DT, Hunger SP, Loh ML. Optimizing therapy in the modern age: differences in length of maintenance therapy in acute lymphoblastic leukemia. Blood 2021; 137 (02) 168-177
  • 4 Bassan R, Maino E, Cortelazzo S. Lymphoblastic lymphoma: an updated review on biology, diagnosis, and treatment. Eur J Haematol 2016; 96 (05) 447-460
  • 5 Mejía-Aranguré E, Reyes-López A, Juárez-Villegas LE. et al. Costs associated with adverse events from remission induction for children with acute lymphoblastic leukemia (ALL). BMC Health Serv Res 2022; 22 (01) 1522
  • 6 Yang W, Karol SE, Hoshitsuki K. et al. Association of inherited genetic factors with drug-induced hepatic damage among children with acute lymphoblastic leukemia. JAMA Netw Open 2022; 5 (12) e2248803
  • 7 Vora AJ, Mitchell C, Goulden N. et al. UKALL 2003, a randomised trial investigating treatment reduction for children and young adults with minimal residual disease defined low risk acute lymphoblastic leukaemia. Blood 2010; 116: 496
  • 8 Lashkari HP, Faheem M, Sridevi Hanaganahalli B. et al. Resource limited centres can deliver treatment for children with acute lymphoblastic leukaemia with risk-stratified minimal residual disease based UKALL 2003 protocol with no modification and a good outcome. Expert Rev Hematol 2020; 13 (10) 1143-1151
  • 9 Common Terminology Criteria for Adverse Events (CTCAE) Version 5. Published: November 27. US Department of Health and Human Services, National Institutes of Health, National Cancer Institute. Common Terminology Criteria for Adverse Events (CTCAE). Accessed August 23, 2023 at: cancer.gov
  • 10 Denton CC, Rawlins YA, Oberley MJ, Bhojwani D, Orgel E. Predictors of hepatotoxicity and pancreatitis in children and adolescents with acute lymphoblastic leukemia treated according to contemporary regimens. Pediatr Blood Cancer 2018; 65 (03) DOI: 10.1002/pbc.26891.
  • 11 Hashmi SK, Navai SA, Chambers TM. et al. Incidence and predictors of treatment-related conjugated hyperbilirubinemia during early treatment phases for children with acute lymphoblastic leukemia. Pediatr Blood Cancer 2020; 67 (02) e28063
  • 12 Felice MS, Hammermuller E, De Dávila MT. et al. Acute lymphoblastic leukemia presenting as acute hepatic failure in childhood. Leuk Lymphoma 2000; 38 (5-6): 633-637
  • 13 McCord RG, Gilbert EF, Joo PJ. Acute leukemia presenting as jaundice with acute liver failure. Clin Pediatr (Phila) 1973; 12 (12) 17A , passim
  • 14 Conway Jr EE, Santorineou M, Mitsudo S. Fulminant hepatic failure in a child with acute lymphoblastic leukemia. J Pediatr Gastroenterol Nutr 1992; 15 (02) 194-197
  • 15 Segal I, Rassekh SR, Bond MC, Senger C, Schreiber RA. Abnormal liver transaminases and conjugated hyperbilirubinemia at presentation of acute lymphoblastic leukemia. Pediatr Blood Cancer 2010; 55 (03) 434-439
  • 16 Mekonnen AT, Wondmeneh TG. Evaluation of liver function tests to identify hepatotoxicity among acute lymphoblastic leukemia patients who are receiving chemotherapy induction. Sci Rep 2022; 12 (01) 13215