Abstract
The incidence of acute lymphoblastic leukaemia (ALL) in adults is about 6500 cases per year in the United States alone, making it the second most prevalent form of acute leukaemia in adults. Chromosomal abnormalities and genetic changes that are involved in the differentiation and proliferation of lymphoid precursor cells are the defining characteristics of allogeneic lymphoma (ALL). 75% of cases in adults develop from progenitors of the B-cell lineage, whereas the remaining instances consist of malignant T-cell precursors. This percentage is based on clinical observations. Historically, risk classification has been determined by clinical criteria such as age, white blood cell count, and response to chemotherapy. However, the identification of recurrent genetic abnormalities has assisted in refining individual prognosis and guiding therapeutic techniques. Multi-agent chemotherapy with vincristine, corticosteroids, and an anthracycline, along with allogeneic stem cell transplantation for individuals who are qualified, continues to be the cornerstone of treatment, despite the fact that there have been advancements in medication administration. It is common for elderly individuals to be unable to take such regimens, and their prognosis is especially adverse in this regard. Acute lymphocytic leukaemia, also known as ALL, is a kind of cancer that is frequently detected in youngsters. Additionally, there has been a consistent rise in the number of cases of ALL as well as treatment failures, despite the fact that technical improvements have been made to improve treatment and survival rates. Specifically, the pathogenic combination between genetic and environmental variables that leads to ALL in children is the topic of discussion in this work. It conducts an analysis of the previously established therapy guidelines as well as the significant hurdles that lead to treatment toxicities, recurrence, and resistance. An analysis of a ten-year trend in the management guidelines of paediatric patients is presented in this paper. Furthermore, the outcome of ALL treatments and the general incidence of the disease are strongly influenced by a number of risk factors, including the interaction between hereditary and environmental factors. Furthermore, enormous financial costs have continued to be a very big obstacle in terms of the consequences.
Full text article
References
Horowitz WM, Messerer D, Hoelzer D. Chemotherapy compared with bone marrow transplantation for adults with acute lymphoblastic leukemia in first remission. Am Int Med 1991;115:13-18.
Larson RA, Stock W, Hoelzer DF, Kantarjian H. Acute lymphoblastic leukemia in adults. Educational Brochure, American Society of Hematology, 1998.
Pui CH, Evans WE. Acute lymphoblastic leukemia. N Engl J Med 1998;339:605-615. Thandla S, Aplan PD. Molecular biology of acute lymphocytic leukemia. Semin Oneal 1997 ;24:45-56.
Salzer WL, Burke MJ, Devidas M, et al.: Impact of intrathecal triple therapy versus intrathecal methotrexate on disease-free survival for high-risk B-lymphoblastic leukemia: Children's Oncology Group Study AALL1131. J Clin Oncol. 2020, 38:2628-38.
Zhang BH, Wang J, Xue HM, Chen C: Impact of chemotherapy-related hyperglycemia on prognosis of child acute lymphocytic leukemia. Asian Pac J Cancer Prev. 2014, 15:8855-9.
Berry DA, Zhou S, Higley H, et al.: Association of minimal residual disease with clinical outcome in pediatric and adult acute lymphoblastic leukemia: a meta-analysis. JAMA Oncol. 2017, 3:e170580.
Maloney KW, Devidas M, Wang C, et al.: Outcome in children with standard-risk B-cell acute lymphoblastic leukemia: results of Children's Oncology Group Trial AALL0331. J Clin Oncol. 2020, 38:602-12.
Minimal residual disease evaluation in childhood acute lymphoblastic leukemia: an economic analysis . Ont Health Technol Assess Ser. 2016, 16:1-83.
Li W, Zhang Y, Kankala RK, Zou L, Chen Z: Antibody and cellular-based therapies for pediatric acute lymphoblastic leukemia: mechanisms and prospects. Pharmacology. 2022, 107:368-75.
Rivera GK, Ribeiro RC: Improving treatment of children with acute lymphoblastic leukemia in developing countries through technology sharing, collaboration and partnerships. Expert Rev Hematol. 2014, 7:649-57.
Burkhardt B, Mueller S, Khanam T, Perkins SL: Current status and future directions of T-lymphoblastic lymphoma in children and adolescents. Br J Haematol. 2016, 173:545-59.
Dunsmore KP, Winter SS, Devidas M, et al.: Children's Oncology Group AALL0434: a phase III randomized clinical trial testing nelarabine in newly diagnosed T-cell acute lymphoblastic leukemia. J Clin Oncol. 2020, 38:3282-93.
Fournier-Goodnight AS, Ashford JM, Clark KN, et al.: Disseminability of computerized cognitive training: performance across coaches. Appl Neuropsychol Child. 2019, 8:113-22.
American Cancer Society: treatment of children with acute lymphocytic leukemia (ALL) . (2019). Accessed: August 12, 2023:
Jędraszek K, Malczewska M, Parysek-Wójcik K, Lejman M: Resistance mechanisms in pediatric B-cell acute lymphoblastic leukemia. Int J Mol Sci. 2022, 23:
Horton TM, McNeer JL: Treatment of Acute Lymphoblastic Leukemia/Lymphoma in Children and Adolescents. UpToDate. Newburger P, Rosmarin A (ed): Wolters Kluwer Health, Waltham, MA; 2023. 12:
Domenech C, Suciu S, De Moerloose B, et al.: Dexamethasone (6 mg/m2/day) and prednisolone (60 mg/m2/day) were equally effective as induction therapy for childhood acute lymphoblastic leukemia in the EORTC CLG 58951 randomized trial. Haematologica. 2014, 99:1220-7.
Mahadeo KM, Khazal SJ, Abdel-Azim H, et al.: Management guidelines for paediatric patients receiving chimeric antigen receptor T cell therapy. Nat Rev Clin Oncol. 2019, 16:45-63.
Thomas DA, O'Brien S, Jorgensen JL, Cortes J, Faderl S, Garcia-Manero G et al. Prognostic significance of CD20 expression in adults with de novo precursor B-lineage acute lymphoblastic leukemia. Blood 13: 6330–6337.
Wierda WG, Padmanabhan S, Chan GW, Gupta IV, Lisby S, Österborg A et al. Ofatumumab is active in patients with fludarabine-refractory CLL irrespective of prior rituximab: results from the phase 2 international study. Blood 2011; 118: 5126–5129.
Teeling JL, French RR, Cragg MS, v.d. Brakel J, Pluyter M, Huang H et al. Characterization of new human CD20 monoclonal antibodies with potent cytolytic activity against non-Hodgkin lymphomas. Blood 2004; 104: 1793–1800.
Pawluczkowycz AW, Beurskens FJ, Beum PV, Lindorfer MA, v.d. Winkel JGJ, Parren PWHI et al. Binding of Submaximal C1q Promotes ComplementDependent Cytotoxicity (CDC) of B Cells Opsonized with Anti-CD20 mAbs Ofatumumab (OFA) or Rituximab (RTX): Considerably Higher Levels of CDC Are Induced by OFA than by RTX. J Immunol 2009; 183: 749–758.
Jabbour E, Hagop K, Thomas D, Garcia-Manero G, Hoehn D, Garris R et al. Phase II Study Of The Hyper-CVAD Regimen In Combination With Ofatumumab As Frontline Therapy For Adults With CD-20 Positive Acute Lymphoblastic Leukemia (ALL). Blood 2013; 122: 2664.
Sasaki K, Kantarjian HM, Ravandi F, Daver N, Kadia TM, Khouri RB et al. Frontline Ofatumumab in Combination with Hyper-CVAD for Adult Patients with CD-20 Positive Acute Lymphoblastic Leukemia (ALL): Interim Result of a Phase II Clinical Trial. Poster presented at the American Society of Hematology 58th Annual Meeting and Exposition. 2 December 2016, San Diego, CA, USA, 2016.
Thomas DA, O'Brien S, Faderl S, Garcia-Manero G, Ferrajoli A, Wierda W et al. Chemoimmunotherapy With a Modified Hyper-CVAD and Rituximab Regimen Improves Outcome in De Novo Philadelphia Chromosome–Negative Precursor B-Lineage Acute Lymphoblastic Leukemia. J Clin Oncol 2010; 28: 3880–3889.
Herter S, Herting F, Mundigl O, Waldhauer I, Weinzierl T, Fauti T et al. Preclinical activity of the type II CD20 antibody GA101 (obinutuzumab) compared with rituximab and ofatumumab in vitro and in xenograft models. Mol Cancer Ther 2013; 12: 2031–2042.
Awasthi A, Ayello J, Van de Ven C, Elmacken M, Sabulski A, Barth MJ et al. Obinutuzumab (GA101) compared to rituximab significantly enhances cell death and antibody-dependent cytotoxicity and improves overall survival against CD20(+) rituximab-sensitive/-resistant Burkitt lymphoma (BL) and precursor B-acute lymphoblastic leukaemia (pre-B-ALL): potential targeted therapy in patients with poor risk CD20(+) BL and pre-B-ALL. Br J Haematol 2015; 171: 763–775.
Goede V, Fischer K, Busch R, Engelke A, Eichhorst B, Wendtner CM et al. Obinutuzumab plus Chlorambucil in Patients with CLL and Coexisting Conditions. N Engl J Med 2014; 370: 1101–1110.
Smith EJ, Olson K, Haber LJ, Varghese B, Duramad P, Tustian AD et al. A novel, native-format bispecific antibody triggering T-cell killing of B-cells is robustly active in mouse tumor models and cynomolgus monkeys. Scientific Reports, Published online 2015; 5: 17943, 2015.
Bannerji R, Brown JR, Advani RH, Arnason J, O'Brien SM, Allan JN et al. Phase 1 Study of REGN1979, an Anti-CD20 x Anti-CD3 Bispecific Monoclonal Antibody, in Patients with CD20+ B-Cell Malignancies Previously Treated with CD20-Directed Antibody Therapy. Blood 2016; 128: 621.
Fujimoto M, Poe JC, Jansen PJ, Sato S, Tedder TF. CD19 amplifies B lymphocyte signal transduction by regulating Src-family protein tyrosine kinase activation. J Immunol 1999; 162: 7088–7094.
Otero DC, Omori SA, Rickert RC. CD19-dependent Activation of Akt Kinase in B-lymphocytes. J Biol Chem 2001; 276: 1474–1478.
Chung EY, Psathas JN, Yu D, Li Y, Weiss MJ, Thomas-Tikhonenko A. CD19 is a major B cell receptor-independent activator of MYC-driven B-lymphomagenesis. J Clin Invest 2012; 122: 2257–2266.
Ning BT, Tang YM, Chen YH, Shen HQ, Qian BQ. Comparison between CD19 and CD20 expression patterns on acute leukemic cells. Zhongguo Shi Yan Xue Ye Xue Za Zhi 2005; 13: 943–947.
Widdison Wayne C, Wilhelm Sharon D, Cavanagh Emily E, Whiteman Kathleen R, Leece Barbara A, Kovtun Yelena et al. Semisynthetic Maytansine Analogues for the Targeted Treatment of Cancer. J Med Chem 2006; 49: 4392–4408.
Authors
Copyright (c) 2024 https://creativecommons.org/licenses/by/4.0/

This work is licensed under a Creative Commons Attribution 4.0 International License.