Implementation of pharmacogenetics for treatment of patients with acute lymphoblastic leukemia

Violeta Graiqevci-Uka1, Emir Behluli1, Rifat Hadziselimovic2, Thomas Liehr3, Gazmend Temaj4

1Department of Pediatrics, University Clinical Center, Prishtina, Kosovo

2Faculty of Science, University of Sarajevo, 71000 Sarajevo, Bosnia and Herzegovina

3Jena University Hospital, Friedrich Schiller University, Institute of Human Genetics, Jena, Germany

4UBT College, Faculty of Pharmacy, Prishtina, Kosovo

Corresponding author: Gazmend Temaj (gazmend.temaj@ubt-uni.net)

Abstract

Introduction: Acute lymphoblastic leukemia (ALL) is the most frequent pediatric leukemia; it can be defined according to chromosomic and genomic data. Cytogenetic analyses and determination of chromosomal numbers (such as hypo- or hyperdiploidy) and/or specific chromosomal rearrangements are basic for ALL classification and treatment. Even though cure rates of childhood ALL are at ~95%, pharmacogenetic aspects are of raising importance.

Material and Methods: We have analyzed the literature for ALL subtypes, corresponding therapy options, and pharmacogenetic implications.

Results: Data for ALL subtypes such as B-ALL, T-ALL, Ph-like ALL, DS-ALL, ETP-ALL, BCR-ABL1-like ALL are presented here. The gene polymorphism which lead to metabolizability of 6-MP are ITPA variants (94C>A) and IVS2+21A>C, in conjunction with TPMT (238G>C, TPMT*3B 460G>A and *3C 719A>G and NUDT15 (415C>T). For methotrexate metabolism gene polymorphisms are found for gene MTHFR as C677T and A1298C.

Conclusion: In the last decade in many hospital laboratories, pharmacogenetic aspects gain more and more importance. Application of many molecular biology methods provided progress in treatment and diagnosis of ALL patients. Combination therapy is proposed as an alternative to single drug treatments.

Graphical abstract

Keywords: B-ALL, T-ALL, Ph-Like ALL, methotrexate, 6-mercaptopurine

Introduction

Acute lymphoblastic leukemia (ALL) is a rare heterogeneous hematological malignancy, which is characterized by uncontrolled proliferation of premature lymphoid cells. Recently, progress was achieved by developing many strategies to improve the therapeutic results for adults with ALL, by introduction of tyrosine-kinases inhibitor-, CAR- (chimeric antigen receptor), T-cell-, antibody-drug conjugation-, monoclonal and specific antibody-therapies. Besides, ALL is the most common cancer in children, being responsible for about 30% of childhood cancers worldwide (Graiqevci-Uka et al. 2022). Here, cure rates of up to 94% were achieved. However, significant interindividual variability in drug toxicity and treatment outcome was observed. Thus, pharmacogenetics and identification of underlying genetic polymorphisms become more and more important (Evans et al. 2013).

There are different types of protocols for treatment of ALL patients. The typical treatment starts with the induction phase through applying anthracyclines such as doxorubicin (DOX) or daunorubicin (DAU), L-Asparaginase (LASPA), glucocorticoids such as prednisone (PRED) or prednisolone (PRDL), and vinca-alkaloids such as vincristine (VINC) to eradicate leukemic cells. The consolidation phase mainly comprises methotrexate (MTX) with 6-mercaptopurine (6-MP) treatment to kill residual leukemia cells. This phase may be followed by a re-induction cycle with dexamethasone (DEXA) and VINC depending on the presence of residual cells. Finally, to maintain remission, patients are treated with 6-MP and MTX with rotating cycle of DEXA, VINC or cyclophosphamide (CTX) (Stanulla et al. 2009).

Pharmacogenetics is increasingly taken into consideration in childhood leukemia, specifically ALL (Lopez-Lopez et al. 2014; Mei et al. 2015; Al-Mahayri et al. 2017; Lee et al. 2017; Rudin et al. 2017). Genetic predisposition needs to be considered in single drug admission as well as in combination therapy (e.g. of 6-MP and MTX) and can influence such hepatotoxicity or myelo-suppression (Schmiegelow et al. 2014).

Here, chromosomic and genomic based treatment options of ALL together with pharmacogenetic aspects are reviewed.

Material and Methods

The studies selected and evaluated here studies were published between 2013-2022. The keywords which were used were “treatment of ALL” and “pharmacogenetics of ALL”. We screened approximately 100 papers, and we selected every paper based on gene mutations being important for therapy.

Results and Discussion

Chromosomic and genomic subtypes

ALL is a genetically heterogeneous lymphoid neoplasm derived from T or B-cells. Based on genetic and morphologic characteristics, ALL cases are divided into subgroups (Hoelzer et al. 2016). Diagnosis of ALL is based on bone marrow biopsy and aspiration followed by morphological analysis, flow cytometry immunophenotype, karyotyping, FISH (fluorescence in situ hybridization) and molecular genetics to determine numerical and structural chromosomal aberrations and/or rearrangements on gene level (Faderl et al. 2010).

The German Multicenter Study Group on Adult Acute Lymphoblastic leukemia characterized differences between T- and B- cell ALL based on immunophenotype and molecular genetics. B-ALL comprises approximately 75% of all ALL cases. The B-ALL typically shows expression of cluster differentiation of CD19, CD20, CD22 and CD79a, whereas T-ALL expresses CD3, CD1a, CD2, CD5, CD7, CD56 and TdT. B-ALL has many subtypes, characterized by different chromosomal abnormalities (Faderl et al. 2010; Künz et al. 2022; Moorman et al. 2022). Patients with complex karyotypes and with hypodiploidy/near triploidy have adverse prognoses (Moorman et al. 2022).

The study by Moorman et al. (2022) revealed that patients with complex karyotype and with hypodiploidy/near triploidy show higher rates of relapse and death than other patients.

B- ALL

The ALL patients undergo a generalized treatment. Genomic features of ALL have been classified into several-subtypes (see Table 1).

Table 1.

Download as

_XLSX_

_CSV_

The genome subtype of ALL

Type

Characterization

Gene mutations

B-ALL (B-cell acute lymphoblastic leukemia)

highest prevalence of Acute Leukemia

DUX4; EPOR; ZNF384; ZNF362; NUTM1; ZNF618; ARID5B; CEBPE; GATA3; RUNX1; PAX5; ETV6; IKZF1; TCF3-HLF; KMT2; ETS

T-ALL (T-cell acute lymphoblastic leukemia)

breakpoints in T-cell receptor gene

TAL1; TAL2; LYL1; TLX1; TLX3; NKX2-5, LMO1; LMO2; MYB; BCL11B; SP11; CDKN2; FBXW7; GATA3; IKZF1; RUNX1; ETV6

Ph-like ALL (Philadelphia chromosome–like acute lymphoblastic leukemia)

break-points cluster region in ABL1 gene

BCR-ABL1; IGH-CRLF2; P2RY8-CRLF2; JAK1; JAK2; JAK3; TYK2; ABL1; ABL2; CSF1R; LYN; PDGFRA; PDGFRB; FLT3; FGFR1; NTRK3; PTK2B; KMT2A

DS-ALL (Down Syndrome-Acute Lymphoblastic Leukemia)

children with Down syndrome have high risk for Acute Lymphoblastic Leukemia and Acute Myeloid Leukemia

 

ETS; JAK2; P2RY8-CRLF2; RAS; CRLF2; PTPN11

ETP-ALL (Early T-cell precursor acute lymphoblastic leukemia/lymphoma)

the premature arrest of T-cell differentiation

RUNX1; IKZF1; ETV6; GATA3; EP300; BRAF; FLT3; IGFR1; JAK1; JAK3; KRAS; NRAS; IL7r; EED; SUZ12; EZH2

BCR-ABL1-like ALL (BCR-ABL1-like acute lymphoblastic leukemia)

the major features are genetic changes in kinase activities and IL (interleukine) kinase

ABL1; ABL2; CSFIR; PDGFRB; PDGFRA; CRLF2; EPOR; TSLP; IL2B; TYK2; KRAS; NRAS; NF1; PTNP11; CBL1; BRAF; NTRK3; PTK2B; FLT3; BLNK; FGFR1; DGKH CSF1R; JAK2; CRLF2

Relapsed Acute Lymphoblastic Leukemia   

increased risk for male children, CNS disturbances, T-cell lineage

NT5C; TP53 Alterations; ETV6/RUNX1 Mutations; BCR-ABL Fusion Gene

Two major types with higher risk level of ALL are B-cell acute lymphoblastic leukemia (B-ALL) and T-cell acute lymphoblastic leukemia (T-ALL), and include over thirty distinct subtypes characterized by genetic alternation in germline and somatic cells, which have the ability to cover distinct gene expression profiles (Iacobucci et al. 2017; Pui et al. 2018; Schwab et al. 2018; Gu et al. 2019; Mullighan et al. 2019; Li et al. 2021; Montefiori et al. 2021).

These subtypes are defined by chromosome gains and losses (hyperploidy and hypoploidy), chromosomal rearrangements, such as gene fusions as for example of DUX4 and EPOR, BXL11B –rearrangements in T-ALL (Montefiori et al. 2021), and as subtypes with similar gene expression profile but different alternations, such as BCR-ABL1-like ALL and ETV6-RUNX1-like ALL (Roberts et al. 2012; Roberts et al. 2014; Li et al. 2018; Gu et al. 2019; Kimura et al. 2020; Graiqevci-Uka et al. 2023).

ZNF384 or ZNF362 are rearranged acute leukemia and are typical for another subtype which is present in approximately 6% of children, and 7.3% of adult, 15% of AYA B-ALL, and 48% of B/myeloid mixed phenotype acute leukemia (MPAL) (Gu et al. 2019; Li et al. 2018; Zaliova et al. 2019; Alexander et al. 2018; Hirabayashi et al. 2019). In NUTM1 gene (nuclear protein in testis midline carcinoma family 1), mutations are found in approximately <2% of children with B-ALL (Li et al. 2018; Gu et al. 2019; Hormann et al. 2019; McEvoy et al. 2020; Ueno et al. 2020; Boer et al. 2021).

Germline mutations and polymorphism in AT-rich at domain 5B (ARID5B), CCAAAT/enhancer binding protein epsilon (CEBPE), GATA binding protein 3 (GATA3), paired-box containing (PAX5), Ets variant gene 6 (ETV6) and IKZF1 gene enhance vulnerability to B-ALL (Perez-Andreu et al. 2013; Perez-Andreu et al. 2015; Wu et al. 2018). Transcription factor 1 (RUNX1) can be translocated in association with many tumor suppressor genes, accounting for about 25% of childhood B-ALL (Parker et al. 2008).

Another type of B-ALL is intra-chromosomal amplification of chromosome (iAMP21); this is associated with a constitutional Robertsonian rearrangement being more prevalent in elder children over nine years (Li et al. 2014). Childhood B-ALL also involves translocation of Histone-lysine N-methyltransferase 2A to KMT2, most commonly in 3-month-old patients (Pieters et al. 2007). The sPI3K, MEK and Ras signaling pathways are shown to play pivotal role in enhancing mutation of KMT-2A gene. Translocation of this gene is involved in different epigenetic processes, such as histone methylation or histone deacetylation. Hence, decitabine, 5-azacytidine, panobinostat etc. that block epigenetic modulation have been suggested as suitable therapeutics targeting patients with B-ALL.

DS (Down Syndrome -ALL)

DS-ALL refers to Acute Lymphoblastic Leukemia (ALL) that is associated with Down syndrome (DS). Down syndrome can be associated with B-ALL, and children with ALL and DS have very poor outcomes (Izraeli et al. 2014; Behluli et al. 2022). Here are the key points regarding DS-ALL based on the provided search results:

1)   Unique characteristics: DS-ALL presents unique characteristics compared to non-DS ALL (NDS-ALL). In DS, the increased risk of ALL is primarily limited to the B-cell precursor phenotype. Additionally, acquired polysomy of chromosome 21 is predominantly found in B-cell precursor ALL in children without DS (Izraeli et al. 2014).

2)   Genetic variability: DS-ALL is not a single biological entity but rather exhibits heterogeneity confirmed by gene expression and cytogenetic analyses. Notably, common cytogenetic subgroups and unfavorable translocations like BCR-ABL and MLL-AF4 are less common in DS-ALL. Approximately 40% of DS-ALL cases have a normal karyotype compared to only 7% of NDS-ALL cases (Izraeli et al. 2014).

3)   Cytokine receptor abnormalities: Abnormal expression of the cytokine receptor CRLF2 has been identified in around 60% of DS-ALL cases. This abnormality, together with other mutations downstream, provides a pathway relevant to a significant proportion of DS-ALL cases that may be targeted by specific inhibitors (Izraeli et al. 2014).

4)   Treatment implications: The genetic differences between DS and NDS ALL have implications for therapy outcomes. Stem cell transplant (SCT) can be considered for DS-ALL patients in good general health with a good response to relapse induction chemotherapy. A radiation-free approach may be preferred due to concerns about central nervous system toxicity (Izraeli et al. 2014; Davis et al. 2023; Duffield et al. 2023).  [Understanding the distinct features of DS-ALL, including genetic variations and treatment implications, is crucial for optimizing therapeutic strategies and improving outcomes for individuals with DS-ALL.

ETP-acute lymphoblastic leukemia (ETP-ALL)

Early T-cell precursor acute lymphoblastic leukemia (ETP-ALL) is a rare and distinct subtype of T-cell acute lymphoblastic leukemia (T-ALL) characterized by specific genetic and immunophenotypic features. Here are the key insights based on the provided search results:

1)   Immunophenotypic characteristics: ETP-ALL is identified by a unique immunophenotypic signature, including CD7+, with CD34+ and/or CD13+/CD33+, while being negative for CD1, CD4, and CD8. This specific immunophenotypic profile aids in the accurate identification of ETP-ALL cases, with 94% specificity (Tarantini et al. 2021).

2)   Genetic aberrations: ETP-ALL exhibits a distinct genetic landscape compared to other T-ALL subtypes. Mutations in genes encoding transcription factors, kinase signaling molecules, and epigenetic modifiers are common in ETP-ALL. Notably, mutations in FLT3, DMNT3A, IL7R, JAK3, RUNX1, EZH2, and EED are more prevalent in ETP-ALL cases (Sin et al. 2021).

3)   Chromosomal abnormalities: While no clear association with specific chromosomal abnormalities is found in ETP-ALL, the disease is characterized by high genomic instability. Some reported chromosomal aberrations include a slightly higher frequency of deletion 13q and a translocation t(2;14)(q22;q32) associated with ZEB2 deregulation (Tarantini et al. 2021).

4)   Clinical aggressiveness: ETP-ALL is known for its clinical aggressiveness and historically poor outcomes. The unique genetic and immunophenotypic features contribute to the challenging nature of this subtype. Allogeneic stem cell transplant (SCT) has shown potential benefits in improving outcomes for ETP-ALL patients (Jain et al. 2016). Understanding the distinct characteristics of ETP-ALL, including its immunophenotypic profile, genetic mutations, and clinical behavior, is crucial for tailored treatment approaches and prognostication in individuals diagnosed with this rare subtype of T-cell acute lymphoblastic leukemia.

T-ALL

It is known that T-ALL cells express a subset of T-cell markers such as CD3, cyCD3, CD2, CD5, CD7, CD8 (Iacobucci et al. 2021). The different cases of T-ALL may be subclassified according to aberrant expression and dysregulated pathway of different transcription factors such as basic helix-loop-helix (bHLH) factors (TAL1, TAL2, LYL1), homeobox genes (TLX1 (HOX11), TLX3 (HOX11L2), NKX2-5, HOXA), LMO1, LMO2, MYB, BCL11B and SPI1 (Liu et al. 2017; Seki et al. 2017; Montefiori et al. 2021).

These subtypes are defined with expression profiles by WGS or microarray; however, almost half of these leukemia-initiating alterations in T-ALL show intergenic breakpoints (Roberts et al. 2016; Liu et al. 2017).

Compared to B-ALL, T-ALL has well-defined chemotherapy guidelines, including immunophenotype targets with the potential to minimize T-ALL pathology (Vora et al. 2016). Mutations observed in T-ALL patients affect NOTCH1, CDKN2 (cyclin dependent kinase inhibitor 2A) and F-box WD repeat domain containing 7 (FBXW7) which stimulate the PI3K/AKT signaling pathway during cell proliferation of cancer cells (Paganin and Ferrando 2011). Chromosomal rearrangements are found to be very common in T-ALL; T-cell receptors fused to “T-cell leukemia homeobox 1-T-cell receptor δ (TLX1-TCRδ)” and STIL-TAL1 oncogenes (Tasian et al. 2015) are observed. Mutations in gene TLX have distinct connection with X-chromosome, and occur more often in pediatric male population. X-chromosome’s deletion at the plant homeodomain finger 6 (PHF6) gene plays pivotal role during tumor suppression, histone remodeling and prevent cell proliferation via its direct effect on transcription (Van Vlierberghe et al. 2010).

An “early T-cell precursor” (ETP) cell subtype has also been shown to have influence at up-regulation of immune reactivity; increasing cluster differentiation 3 (CD3) expression and suppressing CD1a and CD (Coustan-Smith et al. 2009). The primary gene mutation for ETP were in GATA3, IKZF1, RUNX1 and ETV6, responsibly for myeloid and stem cell development (Pui 2015).

Ph-like ALL

It is known that 10% of all ALL belong to the category of Philadelphia chromosome-like (BCR-ABL1-like) or better represented as “Ph-like” group. Mutation in the breakpoint region - Abelson murine leukemia gene 1 – is shown to cause resistance to the treatment with tyrosine kinase inhibitors. In the Brazilian patients, there were studies on BCR-ABL1 gene mutation. In 193 patients, two simultaneous mutations (E189G/V299L and E255K/T315I) were observed in 2/5 of patients while other mutations were found in sequential samples of the other three patients (Y253Y/T315I, T315I/E255K and E255K/T315I). This molecular characterization contributes to the identification of resistance profile to TKI inhibition in Brazilian patients (Costa et al. 2018). Tanasi et al. 2019 dentified the ABL-class fusion was identified at initial diagnosis work up and TKI was introduced during front line treatment. In 5 patients, ABL-class fusions were diagnosed at relapse and TKI was introduced in association with salvage therapy This is made by decision of physicians for TKI, TKI dosage and combination also. Fourteen patients were treated with imatinib, 9 with dasatinib, and one – with ponatinib. Three patients first were treated with imatinib and then were switched to dasatinib during the first phase of treatment. The same group reported on the largest cohort of patients with ABL kinase rearrangement exposed to frontline tyrosine kinase inhibitor (TKI) therapy, and showed promising outcomes, reminiscent of those observed in early treatment with imatinib and chemotherapy combination in Ph-ALL patients.

The most common signaling pathway mutated is JAK/STAT; but other alternations are found and are grouped into three types such as: 1) JAK/STAT alteration including mutation in cytokine receptor, e.g. CRLF2 and IL7R; gene rearrangement deregulating cytokine receptor expression, e.g., IGH-CRLF2 and P2RY8-CRLF2 (Mullighan et al. 2009; Yoda et al. 2009; Hertzberg et al. 2010; Russell et al. 2017); gene fusion or mutated activated kinases, e.g., JAK1, JAK2, JAK3, TYK2; and rearrangement of truncated cytokine receptor expression such as cryptic EPOR rearrangement (Iacobucci et al. 2016); 2) fusion involving other genes such as ABL-class genes (ABL1, ABL2, CSF1R, LYN, PDGFRA, PDGFRB) and 3) less common fusion genes such as FLT3, FGFR1, NTRK3, PTK2B (Iacobucci et al. 2017; Roberts 2017).

The study by Raimondi et al. (1990) show mutation of TKI and cytokine receptor genes. The JAK signaling pathway and gene fusion such as “platelet-derived growth factor receptor beta (PDGFRB)”, ABL1, ABL2 and colony stimulation factor 1 receptor (CSF1R) show mutation, as well as gene mutation.

Overall, the optimal therapy has not yet been defined for Ph-like ALL, and prospective trials evaluating TKIs in combination with chemotherapy are currently underway.

Relapsed ALL

Relapsed ALL are a challenge for treatment of children. Until today, treatment options have been limited to incisive cytotoxic chemotherapy with or without site directed radiotherapy and allogenic hematopoietic stem cell transplantation (HSCT) (Hunger and Raetz 2020). In the past decade, several immunotherapeutic methods were developed, changing the treatment of children with relapsed ALL.  Currently, the incorporation of immunotherapeutics is being explored as a treatment approach into salvage regiments, and survival rates and side effects are being investigated. This treatment may offer real promise for less toxic and very effective therapy for children with relapsed ALL (Hu et al. 2022; Lv et al. 2022), Figure 1.

Figure 1. Genetic and other factors which are reported to be involved and to have impact on chemotherapy efficacy and toxicity. Note: ABC – ATP binding cassette. GSH – Glutathione –s-transferase; hMLH1 – mutL homolog 1; MTHFD1 – Methylentetrahydrofolate dehydrogenase; MTHFR – Methylentetrahydrofolate reductase; MDR – multidrug resistance protein; SULT – sulfotransferase; TS – Thymydilate synthase; TPMT – Thiopurine methyl transferase; UGT – UDP glucuronyl transferase; RFC – Reduced folate carrier; XPE   damage-specific DNA binding protein 1; XRCC1 – X-ray repair complementing defective repair in Chinese hamster cell 1.

Genetic polymorphism of ALL

The most reported genetic polymorphism being of potential importance for personalized medicine are genes NUDT15 c.415 C>T (15*3), TPMT*3C, ITPA c.94 C>A, and MRP4 c.2269 C>T (Pai et al. 2021). Also, it has been shown that 6-mercaptopurine toxicity is influenced by genetic polymorphism of genes ITPA 94C>A and IVS2+21A>C, TMPT*2 238G>C, TPMT*3B460G>A and *3C719A>G, and NUDT15 415C>T (Moradveisi et al. 2019; Mao et al. 2021).

MTX

MTX is a folate analog which is a widely used form for cancer treatment (Chabner et al. 1985; Schmiegelow et al. 2014). MTX is pro-drug, being processed by folylpolyglutamyl synthetase (FPGS) to gamma-linked glutamic acid residues. Methotrexate (MTX) is the most effective drug which is used for treatment of pediatric ALL patients which have better overall survival. Delayed MTX clearance can lead to prolonged, elevated exposure, causing increased risks for nephrotoxicity, mucositis, seizures, and neutropenia. So far several genes have been identified which are involved in the MTX clearance. Taylor et al. (2021) during the search for genes involved in the MTX metabolism found that 24 different genes are involved in the folate transport pathway, but the only one gene that reliably demonstrate an effect on MTX is SLCO1B1 (reviewed by Taylor et al. 2021). Ramalingam et al. (2022) show that SLC19A1 gene facilitates cyclic dinucleotide import and folate reductions and help for better management MTX in the ALL-patients who are treated with this type of drug.

Shen et al. (2021) analyzed the effects of MTHFR C677T and A1298C polymorphisms on MTX elimination and toxicities. It was shown that patients with the genotype MTHFR C677T TT could tolerate a significantly higher MTX dose than patients who have genotype CC/CT. But, in the patients with genotype C677T TT, the risk for hypokalemia was shown to increase (1.369 to CC and 1.409 to CT types). The rate of MTX infusion in the patients who have the genotype MTHFR A1298C AC is shown to be slightly lower than in the patients who have genotype CC or AA. The patients with the genotype A1298C AA were shown to have a higher risk for hepatotoxicity – approximately 1.405-fold than those with the AC genotype (P>0.05) (Shen et al. 2021).

The patients who had genotype C677T after treatment with MTX were shown to have oral mucositis, leucopenia, and thrombocytopenia, while patients with genotype A1298C were shown to have a low risk for vomiting, but were more likely to develop anemia and leucopenia (Lu et al. 2021).

The polymorphism of MTHFR C677T and A1298C was shown to lack marker for MTX-related toxicity and/or outcome in pediatric ALL (Umerez et al. 2017).

6-Mercaptopurine

Mercaptopurine is well established for treatment of a wide range of diseases such as leukemia and other inflammatory diseases in general. This drug has a quick rate of absorption and elimination, but it has shown to have different effects on individuals (Perez-Andreu et al. 2013). Seven genes (NUDT15, SUCLA2, TPMT, IPTA, IMPGH1, MTHFR, PACSIN2, and MRP4) are associated with mercaptopurine intolerance (Zhou et al. 2020; Wang et al. 2021). NUDT15 (rs116855232) and IMPGH1 (rs2278293) were associated with a 5.50-fold and 5.80-fold higher risk of leukopenia, respectively (Zhou et al. 2020).

The data from the study of Moradveisi et al. (2019) confirm the important role of TPMT, NUDT15, and ITPA genes in the 6-MP intolerance in Middle Eastern countries for children diagnosed with ALL. Based on the higher frequency of ITPA gene variants found in the study by Maradveisi et al. and higher significant association of genes with 6-MP dose intensity mentioned above, the same author recommends that physicians must consider genotyping of ITPA variants (94C>A (rs1127354)) and IVS2+21A>C (rs7270101)), in conjunction with TPMT (238G>C (rs1800462), TPMT*3B 460G>A (rs1800460) and *3C 719A>G (rs1142345)) and NUDT15 (415C>T (rs116855232)) prior to 6-MP therapy in the children diagnosed with ALL (Moradveisi et al. 2019).

Hao et al. (2021) showed the genetic polymorphism of TMPT gene (CC genotype for both TPMT*2 and TPMT*3B) in the ALL patients. The genetic polymorphism for TPMT*3C (TT and TC) reach 96.76% and 3.24%, respectively. The allele frequency was 1.62% for TPMT*3C and NUDT15 has an effect on the tolerance of 6-MP in the treatment of adult ALL (Hao et al. 2021), Figure 2.

Figure 2. The intercellular pathway of 6-MP (6-mercaptopurine).
Note: SAM – S-adenosylmethionine; SAH-S adenosylmethionine; SAH – S-adenosylhomocysteine; TPMT – thiopurine S-methyltransferase; XO – xanthine oxidase; 6-Me-MP – 6-methyl-mercaptopurine; 6-TIMP – 6-thioinosine triphosphate; ITPA – inosine triphosphate pyrophosphatase; 6-MMPN – 6-methyl-mercaptopurine nucleoside; 6-TGN – 6-thioguanine nucleoside.

Dexamethasone and ALL pharmacogenetics

Dexamethasone is known to be a very important component for treatment of childhood with acute lymphoblastic leukemia (ALL). Such genes as ABCB1, GSTM1, GSTT1, BCL2, BCL2L11, and MCL1 have a potential effect on dexamethasone treatment of ALL patients (Jackson et al. 2016; Jackson et al. 2019; Zou et al. 2021).

The first five days of treatment of children with dexamethasone showed an increase in their nighttime sleeping; and during the daytime, the napping was observed in the young children with ALL. The increases in sleep duration return to baseline one day after the discontinuation of dexamethasone (Rosen et al. 2015).

Cytarabine

Cytarabine is a type of drug which is known by brand name Ara C. and in the activation form is as 1-β-D-arabinofuranoxyl cytosine-5’-triphosphate, and in a dose of 100 mg/m2 is a well established drug for leukemia (Krajinovic et al. 2002). Cytarabine has an ability to block DNA replication via inhibition of DNA polymerases (Wiley et al. 1982).

It is shown that Cytarabine (Ara-c) and EIP synergistically reduce viability of ALL cells with high ERG expression, which may be achieved by promoting their apoptosis and inhibiting their nesting (Cheng et al. 2021). Individual variability among patients diagnosed with leukemia appears to be associated with accumulation of Ara-CTP due to genetic variants of metabolic enzymes. In review by Di Francia et al. (2021), they report about response association of cytarabine and new drugs which are optimized in pharmacokinetics aspects. The study of candidate genes is focused on the sequence variation, alternative splicing, and expression of the key cytarabine gene pathways, including SCL29A1, DCK, CDA, and NT5C2. The SNPs (single nucleotide polymorphism) in the coding region of these genes have shown to participate in the protein production with amino acid changes (Lamba, 2009; Cao et al. 2018; Zhu et al. 2018). The DCK (deoxycytidine kinase) plays the pivotal role in activation of Cytarabine; this gene has several SNPs (-125G>T, -201C>T, -289T>A, -360C>G, and -740G>C) and three others are non-synonymous coding changes Ile24Val, Ala119Gly, and Pro122Ser in activation of cytarabine and metabolization (Lamba et al. 2007). Also Cytarabine catabolism is made during the deamination process, which involves CDA and CMPD enzymes, and dephosphorylation by NT5C family enzymes.

Conclusion

In conclusion of this review, we report about the subtypes of ALL, the genes mutations which are found by using different methods for diagnosis and treatment of patients with ALL, and genes which are involved in drug administration together with their polymorphisms. Based on literature which we have reviewed, we can see that in many cases gene fusions in all types of ALL. Some of literature resource them have been focused on the treatment of ALL patients (B-ALL, T-ALL and Ph-Like ALL) and genes which are responsible for these types of ALL. In this review, we have considered all data published together, and we show the growth evidence of the involvement of drugs such as MTX, 6-MP, Cytarabine, and Dexamethasone in the treatment of patients diagnosed with ALL. On the other hand, considering the polymorphism for B-ALL, T-ALL and Ph-like ALL treatment and toxicities, it would be very interesting to analyze gene polymorphism such as DUX4 and EPOR, BXL11B, BCR-ABL1-like ALL, ETV6-RUNX (TLX1 (HOX11), TLX3 (HOX11L2), NKX2-5, HOXA), LMO1, LMO2, MYB, BCL11B and SPI1. The development field of pharmacogenetics has started to produce the promise results and genetic variants have great potential to be used as biomarkers for further studies, which are very important to increase the power of patients cure diagnosed before.

Conflict of interest

The authors declare no conflict of interests.

Financial support

The authors declare no financial support received.

Data availability

All of the data that support the findings of this study are available in the main text.

References

·      Al-Mahayri ZN, Patrinos GP, Ali BR (2017) Pharmacogenomics in pediatric acute lymphoblastic leukemia: promises and limitations. Pharmacogenomics 18(7): 687–699. https://doi.org/10.2217/pgs-2017-0005 [PubMed]

·      Alexander TB, Gu Z, Iacobucci I, Dickerson K, Choi JK, Xu B, Payne-Turner D, Yoshihara H, Loh ML, Horan J, Buldini B, Basso G, Elitzur S, de Haas V, Zwaan CM, Yeoh A, Reinhardt D, Tomizawa D, Kiyokawa N, Lammens T, De Moerloose B, Catchpoole D, Hori H, Moorman A, Moore AS, Hrusak O, Meshinchi S, Orgel E, Devidas M, Borowitz M, Wood B, Heerema NA, Carrol A, Yang YL, Smith MA, Davidsen TM, Hermida LC, Gesuwan P, Marra MA, Ma Y, Mungall AJ, Moore RA, Jones SJM, Valentine M, Janke LJ, Rubnitz JE, Pui CH, Ding L, Liu Y, Zhang J, Nichols KE, Downing JR, Cao X, Shi L, Pounds S, Newman S, Pei D, Guidry Auvil JM, Gerhard DS, Hunger SP, Inaba H, Mullighan CG (2018) The genetic basis and cell of origin of mixed phenotype acute leukaemia. Nature 562(7727): 373–379. https://doi.org/10.1038/s41586-018-0436-0 [PubMed] [PMC]

·      Behluli E, Nuhii N, Liehr T, Temaj G (2022) Suspicions regarding the genetic inheritance of acute lymphoblastic leukemia in patients with down syndrome. Journal of Mother and Child 26(1): 104–110. https://doi.org/10.34763/jmotherandchild.20222601.d-22-00002 [PubMed] [PMC]

·      Boer JM, Valsecchi MG, Hormann FM, Antić Ž, Zaliova M, Schwab C, Cazzaniga G, Arfeuille C, Cavé H, Attarbaschi A, Strehl S, Escherich G, Imamura T, Ohki K, Grüber TA, Sutton R, Pastorczak A, Lammens T, Lambert F, Li CK, Carrillo de Santa Pau E, Hoffmann S, Möricke A, Harrison CJ, Den Boer ML, De Lorenzo P, Stam RW, Bergmann AK, Pieters R (2021) Favorable outcome of NUTM1-rearranged infant and pediatric B cell precursor acute lymphoblastic leukemia in a collaborative international study. Leukemia 35(10): 2978–2982. https://doi.org/10.1038/s41375-021-01333-y [PubMed] [PMC]

·      Cao Y, Wang X, Cao Z, Wu C, Wu D, Cheng X (2018) Genetic polymorphisms of MBL2 and tuberculosis susceptibility: A metaanalysis of 22 case-control studies. Archives of Medical Sciences 14(6): 1212–1232. https://doi.org/10.5114/aoms.2017.65319 [PubMed] [PMC]

·      Chabner BA, Allegra CJ, Curt GA, Clendeninn NJ, Baram J, Koizumi S, et al. (1985) Polyglutamation of methotrexate. Is methotrexate a prodrug? Journal of Clinical Investigation 76(3): 907–912. https://doi.org/10.1172/JCI112088 [PubMed] [PMC]

·      Cheng L, Zeng S, Yan D, Tu L, Yang Y, Wang X, Zheng X (2021) Cytarabine and EIP co-administration synergistically reduces viability of acute lymphoblastic leukemia cells with high ERG expression. Leukemia Research 109: 106649. https://doi.org/10.1016/j.leukres.2021.106649 [PubMed]

·      Costa HZ, Pereira NF, Kaminski L, Pasquini R, Funke VAM, Mion ALV (2018) Mutations in the breakpoint cluster region-Abelson murine leukemia 1 gene in Brazilian patients with chronic myeloid leukemia. Hematology, Transfusion and Cell Therapy 40(4): 363–367. https://doi.org/10.1016/j.htct.2018.03.005 [PubMed] [PMC]

·      Coustan-Smith E, Mullighan CG, Onciu M, Behm FG, Raimondi SC, Pei D, Cheng C, Su X, Rubnitz JE, Basso G, Biondi A, Pui CH, Downing JR, Campana D (2009) Early T-cell precursor leukaemia: A subtype of very high-risk acute lymphoblastic leukaemia. Lancet Oncology 10(2): 147–156. https://doi.org/10.1016/S1470-2045(08)70314-0 [PubMed] [PMC]

·      Davis K, Sheikh T, Aggarwal N (2023) Emerging molecular subtypes and therapies in acute lymphoblastic leukemia. Seminars in Diagnostic Pathology 40(3): 202215. https://doi.org/10.1053/j.semdp.2023.04.003 [PubMed]

·      Den Boer ML, van Slegtenhorst M, De Menezes RX, Cheok MH, Buijs-Gladdines JG, Peters ST, Van Zutven LJ, Beverloo HB, Van der Spek PJ, Escherich G, Horstmann MA, Janka-Schaub GE, Kamps WA, Evans WE, Pieters R (2009) A subtype of childhood acute lymphoblastic leukaemia with poor treatment outcome: A genome-wide classification study. Lancet Oncology 10(2): 125134. https://doi.org/10.1016/S1470-2045(08)70339-5 [PubMed] [PMC]

·      Di Francia R, Crisci S, De Monaco A, Cafiero C, Re A, Iaccarino G, De Filippi R, Frigeri F, Corazzelli G, Micera A, Pinto A (2021) Response and toxicity to cytarabine therapy in leukemia and lymphoma: From dose puzzle to pharmacogenomic biomarkers. Cancers 13(5): 966. https://doi.org/10.3390/cancers13050966 [PubMed] [PMC]

·      Duffield AS, Mullighan CG, Borowitz MJ (2023) International consensus classification of acute lymphoblastic leukemia/lymphoma. Virchows Archiv 482(1): 11–26. https://doi.org/10.1007/s00428-022-03448-8 [PubMed] [PMC]

·      Evans WE, Crews KR, Pui CH (2013) A health-care system perspective on implementing genomic medicine: pediatric acute lymphoblastic leukemia as a paradigm. Clinical Pharmacology Therapy 94(2): 224–229. https://doi.org/10.1038/clpt.2013.9 [PubMed] [PMC]

·      Faderl S, O’Brien S, Pui CH, Stock W, Wetzler M, Hoelzer D, Kantarjian HM (2010) Adult acute lymphoblastic leukemia. Cancer 116(5): 1165–1176. https://doi.org/10.1002/cncr.24862 [PubMed] [PMC]

·      Graiqevci-Uka V, Behluli E, Spahiu L, Liehr T, Temaj G (2023) A new case of childhood acute lymphoblastic B-Cell leukemia from Pristina. Acta Medica Bulgarica 50(1): 59–62. https://doi.org/10.2478/amb-2023-0009   

·      Graiqevci-Uka V, Behluli E, Spahiu L, Liehr T, Temaj G (2023) Targeted treatment and immunotherapy in high-risk and relapsed/refractory pediatric acute lymphoblastic leukemia. Current Pediatric Review 19(12): 150–156. https://doi.org/10.2174/1573396318666220901165247 [PubMed] [PMC]

·      Gu Z, Churchman ML, Roberts KG, Moore I, Zhou X, Nakitandwe J, Hagiwara K, Pelletier S, Gingras S, Berns H, Payne-Turner D, Hill A, Iacobucci I, Shi L, Pounds S, Cheng C, Pei D, Qu C, Newman S, Devidas M, Dai Y, Reshmi SC, Gastier-Foster J, Raetz EA, Borowitz MJ, Wood BL, Carroll WL, Zweidler-McKay PA, Rabin KR, Mattano LA, Maloney KW, Rambaldi A, Spinelli O, Radich JP, Minden MD, Rowe JM, Luger S, Litzow MR, Tallman MS, Racevskis J, Zhang Y, Bhatia R, Kohlschmidt J, Mrózek K, Bloomfield CD, Stock W, Kornblau S, Kantarjian HM, Konopleva M, Evans WE, Jeha S, Pui CH, Yang J, Paietta E, Downing JR, Relling MV, Zhang J, Loh ML, Hunger SP, Mullighan CG (2019) PAX5-driven subtypes of B-progenitor acute lymphoblastic leukemia. Nature Genetics 51(2): 296–307. https://doi.org/10.1038/s41588-018-0315-5 [PubMed] [PMC]

·      Hao QS, Wang Z, Fang QY, Gong XY, Liu KQ, Li Y, Wei H, Wang Y, Li QH, Wang M, Tian Z, Wang JX, Mi YC (2021) Effect of genetic polymorphism of TPMT and NUDT15 on the tolerance of 6-mercaptopurine therapy in adult acute lymphoblastic leukemia. Zhonghua Xue Ye Xue Za Zhi 42(11): 911916. https://doi.org/10.3760/cma.j.issn.0253-2727.2021.11.005 [PubMed] [PMC] [in Chinese]

·      Hertzberg L, Vendramini E, Ganmore I, Cazzaniga G, Schmitz M, Chalker J, Shiloh R, Iacobucci I, Shochat C, Zeligson S, Cario G, Stanulla M, Strehl S, Russell LJ, Harrison CJ, Bornhauser B, Yoda A, Rechavi G, Bercovich D, Borkhardt A, Kempski H, te Kronnie G, Bourquin JP, Domany E, Izraeli S (2010) Down syndrome acute lymphoblastic leukemia, a highly heterogeneous disease in which aberrant expression of CRLF2 is associated with mutated JAK2: A report from the International BFM Study Group. Blood 115(5): 1006–1017. https://doi.org/10.1182/blood-2009-08-235408 [PubMed]

·      Hetzel S, Mattei AL, Kretzmer H, Qu C, Chen X, Fan Y, Wu G, Roberts KG, Luger S, Litzow M, Rowe J, Paietta E, Stock W, Mardis ER, Wilson RK, Downing JR, Mullighan CG, Meissner A (2022) Acute lymphoblastic leukemia displays a distinct highly methylated genome. Nature Cancer 3(6): 768782. https://doi.org/10.1038/s43018-022-00370-5 [PubMed] [PMC]

·      Hirabayashi S, Butler ER, Ohki K, Kiyokawa N, Bergmann AK, Möricke A, Boer JM, Cavé H, Cazzaniga G, Yeoh AEJ, Sanada M, Imamura T, Inaba H, Mullighan C, Loh ML, Norén-Nyström U, Pastorczak A, Shih LY, Zaliova M, Pui CH, Haas OA, Harrison CJ, Moorman AV, Manabe A (2021) Clinical characteristics and outcomes of B-ALL with ZNF384 rearrangements: A retrospective analysis by the Ponte di Legno Childhood ALL Working Group. Leukemia 35(11): 3272–3277. https://doi.org/10.1038/s41375-021-01199-0 [PubMed] [PMC]

·      Hoelzer D, Bassan R, Dombret H, Ribera J, Buske C (2016) Acute lymphoblastic leukemia in adult patients: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Annals of Hematology and Oncology 27(suppl 5): 69–82. https://doi.org/10.1093/annonc/mdw025 [PubMed]

·      Hoppmann AL, Chen Y, Landier W, Hageman L, Evans WE, Wong FL, Relling MV, Bhatia S (2021) Individual prediction of nonadherence to oral mercaptopurine in children with acute lymphoblastic leukemia: Results from COG AALL03N1. Cancer 127(20): 38323839. https://doi.org/10.1002/cncr.33760 [PubMed] [PMC]

·      Hormann FM, Hoogkamer AQ, Beverloo HB, Boeree A, Dingjan I, Wattel MM, Stam RW, Escherich G, Pieters R, den Boer ML, Boer JM (2019) NUTM1 is a recurrent fusion gene partner in B-cell precursor acute lymphoblastic leukemia associated with increased expression of genes on chromosome band 10p12.31-12.2. Haematologica 104(10): e455–e459. https://doi.org/10.3324/haematol.2018.206961 [PubMed] [PMC]

·      Hu G, Cheng Y, Zuo Y, Chang Y, Suo P, Jia Y, Lu A, Wang Y, Jiao S, Zhang L, Sun Y, Yan C, Xu L, Zhang X, Liu K, Wang Y, Zhang L, Huang X (2022) Comparisons of long-term survival and safety of haploidentical hematopoietic stem cell transplantation after CAR-T cell therapy or chemotherapy in pediatric patients with first relapse of B-cell acute lymphoblastic leukemia based on MRD-guided treatment. Frontiers in Immunology 13: 915590. https://doi.org/10.3389/fimmu.2022.915590 [PubMed] [PMC]

·      Hunger SP (1996) Chromosomal translocations involving the E2A gene in acute lymphoblastic leukemia: Clinical features and molecular pathogenesis. Blood 87(4): 12111224. [PubMed]

·      Hunger SP, Raetz EA (2020) How I treat relapsed acute lymphoblastic leukemia in the pediatric population. Blood 136(16): 18031812. https://doi.org/10.1182/blood.2019004043 [PubMed]

·      Iacobucci I, Kimura S, Mullighan CG (2021) Biologic and therapeutic implications of genomicalterations in acute lymphoblastic leukemia. Journal of Clinical Medicine 10(17): 3792. https://doi.org/10.3390/jcm10173792 [PubMed] [PMC]

·      Iacobucci I, Li Y, Roberts KG, Dobson SM, Kim JC, Payne-Turner D, Harvey RC, Valentine M, McCastlain K, Easton J, Yergeau D, Janke LJ, Shao Y, Chen IM, Rusch M, Zandi S, Kornblau SM, Konopleva M, Jabbour E, Paietta EM, Rowe JM, Pui CH, Gastier-Foster J, Gu Z, Reshmi S, Loh ML, Racevskis J, Tallman MS, Wiernik PH, Litzow MR, Willman CL, McPherson JD, Downing JR, Zhang J, Dick JE, Hunger SP, Mullighan CG (2016) Truncating erythropoietin receptor rearrangements in acute lymphoblastic leukemia. Cancer Cell 29(2): 186–200. https://doi.org/10.1016/j.ccell.2015.12.013 [PubMed] [PMC]

·      Iacobucci I, Mullighan CG (2017) Genetic basis of acute lymphoblastic leukemia. Journal of Clinical Oncology 35(9): 975–983. https://doi.org/10.1200/JCO.2016.70.7836 [PubMed] [PMC]

·      Izraeli S, Vora A, Zwaan CM, Whitlock J (2014) How I treat All in Down's syndrome: Pathobiology and management. Blood 123(1): 3540. https://doi.org/10.1182/blood-2013-07-453480 [PubMed]

·      Jackson RK, Irving JA, Veal GJ (2016) Personalization of dexamethasone therapy in childhood acute lymphoblastic leukaemia. British Journal of Hematology 173(1): 1324. https://doi.org/10.1111/bjh.13924 [PubMed]

·      Jackson RK, Liebich M, Berry P, Errington J, Liu J, Parker C, Moppett J, Samarasinghe S, Hough R, Rowntree C, Goulden NJ, Vora A, Kearns PR, Saha V, Hempel G, Irving JAE, Veal GJ (2019) Impact of dose and duration of therapy on dexamethasone pharmacokinetics in childhood acute lymphoblastic leukaemia: A report from the UKALL 2011 trial. European Journal of Cancer 120: 7585. https://doi.org/10.1016/j.ejca.2019.07.026 [PubMed]

·      Jain N, Lamb AV, O'Brien S, Ravandi F, Konopleva M, Jabbour E, Zuo Z, Jorgensen J, Lin P, Pierce S, Thomas D, Rytting M, Borthakur G, Kadia T, Cortes J, Kantarjian HM, Khoury JD (2016) Early T-cell precursor acute lymphoblastic leukemia/lymphoma (ETP-ALL/LBL) in adolescents and adults: A high-risk subtype. Blood 127(15): 1863–1869. https://doi.org/10.1182/blood-2015-08-661702 [PubMed] [PMC]

·      Kandikonda P, Bostrom B (2019) Methotrexate polyglutamate values in children and adolescents with acute lymphoblastic leukemia during maintenance therapy. Journal of Pediatric Hematology/Oncology 41(6): 429432. https://doi.org/10.1097/MPH.0000000000001530 [PubMed]

·      Kawedia JD, Liu C, Pei D, Cheng C, Fernandez CA, Howard SC, Campana D, Panetta JC, Bowman WP, Evans WE, Pui CH, Relling MV (2012) Dexamethasone exposure and asparaginase antibodies affect relapse risk in acute lymphoblastic leukemia. Blood 119(7): 1658–1664. https://doi.org/10.1182/blood-2011-09-381731 [PubMed] [PMC]

·      Kimura S, Mullighan CG (2020) Molecular markers in ALL: Clinical implications. Best Practice & Research Clinical Haematology 33(3): 101193. https://doi.org/10.1016/j.beha.2020.101193 [PubMed] [PMC]

·      Krajinovic M, Costea I, Chiasson S (2002) Polymorphism of the thymidylate synthase gene and outcome of acute lymphoblastic leukaemia. Lancet 359(9311): 10331034. https://doi.org/10.1016/S0140-6736(02)08065-0 [PubMed]

·      Künz T, Hauswirth AW, Hetzenauer G, Rudzki J, Nachbaur D, Steiner N (2022) Changing landscape in the treatment of adult acute lymphoblastic leukemia (ALL). Cancers 14(17): 4290. https://doi.org/10.3390/cancers14174290 [PubMed] [PMC]

·      Lamba JK (2009) Genetic factors influencing cytarabine therapy. Pharmacogenomics 10(10): 1657–1674. https://doi.org/10.2217/pgs.09.118 [PubMed] [PMC]

·      Lamba JK, Crews K, Pounds S, Schuetz EG, Gresham J, Gandhi V, Plunkett W, Rubnitz J, Ribeiro R (2007) Pharmacogenetics of deoxycytidine kinase: Identification and characterization of novel genetic variants. Journal of Pharmacological and Experimental Therapy 323(3): 935–945. https://doi.org/10.1124/jpet.107.128595 [PubMed]

·      Larsen RH, Hjalgrim LL, Grell K, Kristensen K, Pedersen LG, Brünner ED, Als-Nielsen B, Schmiegelow K, Nersting J (2020) Pharmacokinetics of tablet and liquid formulations of oral 6 mercaptopurine in children with acute lymphoblastic leukemia. Cancer Chemotherapy and Pharmacology 86(1): 2532. https://doi.org/10.1007/s00280-020-04097-x [PubMed]

·      Lee SHR, Yang JJ (2017) Pharmacogenomics in acute lymphoblastic leukemia. Best Practice & Research Clinical Haematology 3(3): 229–236. https://doi.org/10.1016/j.beha.2017.07.007 [PubMed]

·      Li J, Dai Y, Wu L, Zhang M, Ouyang W, Huang J, Chen S (2021) Emerging molecular subtypes and therapeutic targets in B-cell precursor acute lymphoblastic leukemia. Frontiers in Medicine 15(3): 347–371. https://doi.org/10.1007/s11684-020-0821-6 [PubMed]

·      Li JF, Dai YT, Lilljebjörn H, Shen SH, Cui BW, Bai L, Liu YF, Qian MX, Kubota Y, Kiyoi H, Matsumura I, Miyazaki Y, Olsson L, Tan AM, Ariffin H, Chen J, Takita J, Yasuda T, Mano H, Johansson B, Yang JJ, Yeoh AE, Hayakawa F, Chen Z, Pui CH, Fioretos T, Chen SJ, Huang JY (2018) Transcriptional landscape of B cell precursor acute lymphoblastic leukemia based on an international study of 1223 cases. Proceedings of the National Academy of Sciences of the United States of America 115(50): E11711–E11720. https://doi.org/10.1073/pnas.1814397115 [PubMed] [PMC]

·      Li Y, Schwab C, Ryan S, Papaemmanuil E, Robinson HM, Jacobs P, Moorman AV, Dyer S, Borrow J, Griffiths M, Heerema NA, Carroll AJ, Talley P, Bown N, Telford N, Ross FM, Gaunt L, McNally RJQ, Young BD, Sinclair P, Rand V, Teixeira MR, Joseph O, Robinson B, Maddison M, Dastugue N, Vandenberghe P, Stephens PJ, Cheng J, Van Loo P, Stratton MR, Campbell PJ, Harrison CJ (2014) Constitutional and somatic rearrangement of chromosome 21 in acute lymphoblastic leukaemia. Nature 508(7494): 98102. https://doi.org/10.1038/nature13115 [PubMed] [PMC]

·      Liu Y, Easton J, Shao Y, Maciaszek J, Wang Z, Wilkinson MR, McCastlain K, Edmonson M, Pounds SB, Shi L, Zhou X, Ma X, Sioson E, Li Y, Rusch M, Gupta P, Pei D, Cheng C, Smith MA, Auvil JG, Gerhard DS, Relling MV, Winick NJ, Carroll AJ, Heerema NA, Raetz E, Devidas M, Willman CL, Harvey RC, Carroll WL, Dunsmore KP, Winter SS, Wood BL, Sorrentino BP, Downing JR, Loh ML, Hunger SP, Zhang J, Mullighan CG (2017) The genomic landscape of pediatric and young adult T-lineage acute lymphoblastic leukemia. Nature Genetics 49(8): 1211–1218. https://doi.org/10.1038/ng.3909 [PubMed] [PMC]

·      Liu YF, Wang BY, Zhang WN, Huang JY, Li BS, Zhang M, Jiang L, Li JF, Wang MJ, Dai YJ, Zhang ZG, Wang Q, Kong J, Chen B, Zhu YM, Weng XQ, Shen ZX, Li JM, Wang J, Yan XJ, Li Y, Liang YM, Liu L, Chen XQ, Zhang WG, Yan JS, Hu JD, Shen SH, Chen J, Gu LJ, Pei D, Li Y, Wu G, Zhou X, Ren RB, Cheng C, Yang JJ, Wang KK, Wang SY, Zhang J, Mi JQ, Pui CH, Tang JY, Chen Z, Chen SJ (2016) Genomic profiling of adult and pediatric b-cell acute lymphoblastic leukemia. EBioMedicine 8: 173–183. https://doi.org/10.1016/j.ebiom.2016.04.038 [PubMed] [PMC]

·      Lopez-Lopez E, Gutierrez-Camino A, Bilbao-Aldaiturriaga N, Pombar- Gomez M, Martin-Guerrero I, Garcia-Orad A (2014) Pharmacogenetics of childhood acute lymphoblastic leukemia. Pharmacogenomics 15(10): 1383–1398. https://doi.org/10.2217/pgs.14.106 [PubMed]

·      Lu S, Zhu X, Li W, Chen H, Zhou D, Zhen Z, Sun F, Huang J, Zhu J, Wang J, Zhang Y, Sun X (2021) Influence of methylenetetrahydrofolate reductase C677T and A1298C polymorphism on high-dose methotrexate-related toxicities in pediatric non-Hodgkin lymphoma patients. Frontiers in Oncology 11: 598226. https://doi.org/10.3389/fonc.2021.598226 [PubMed] [PMC]

·      Lv M, Zhuang X, Shao S, Li X, Cheng Y, Wu D, Wang X, Qiao T (2022) Immunotherapy for pediatric acute lymphoblastic leukemia: Recent advances and future perspectives. Journal of Immunology Research 13: 921894. https://doi.org/10.3389/fimmu.2022.921894 [PubMed] [PMC]

·      Mao X, Yin R, Sun G, Zhou Y, Yang C, Fang C, Wu Y, Cui T, Liu L, Gan J, Tian X (2021) Effects of TPMT, NUDT15, and ITPA genetic variants on 6-mercaptopurine toxicity for pediatric patients with acute lymphoblastic leukemia in Yunnan of China. Frontiers in Pediatrics 9: 719803. https://doi.org/10.3389/fped.2021.719803 [PubMed] [PMC]

·      McEvoy CR, Fox SB, Prall OWJ (2020) Emerging entities in NUTM1-rearranged neoplasms. Genes, Chromosomes and Cancer 59(6): 375–385. https://doi.org/10.1002/gcc.22838 [PubMed]

·      Mei L, Ontiveros EP, Griffiths EA, Thompson JE, Wang ES, Wetzler M (2015) Pharmacogenetics predictive of response and toxicity in acute lymphoblastic leukemia therapy. Blood Review 29(4): 243–249. https://doi.org/10.1016/j.blre.2015.01.001 [PubMed] [PMC]

·      Mendoza-Santiago A, Becerra E, Garay E, Bah M, Berumen-Segura L, Escobar-Cabrera J, Hernández-Pérez A, García-Alcocer G (2019) Glutamic acid increased methotrexate polyglutamation and cytotoxicity in a CCRF-SB acute lymphoblastic leukemia cell line. Medicina (Kaunas) 55(12): 758. https://doi.org/10.3390/medicina55120758 [PubMed] [PMC]

·      Montefiori LE, Bendig S, Gu Z, Chen X, Pölönen P, Ma X, Murison A, Zeng A, Garcia-Prat L, Dickerson K, Iacobucci I, Abdelhamed S, Hiltenbrand R, Mead PE, Mehr CM, Xu B, Cheng Z, Chang TC, Westover T, Ma J, Stengel A, Kimura S, Qu C, Valentine MB, Rashkovan M, Luger S, Litzow MR, Rowe JM, den Boer ML, Wang V, Yin J, Kornblau SM, Hunger SP, Loh ML, Pui CH, Yang W, Crews KR, Roberts KG, Yang JJ, Relling MV, Evans WE, Stock W, Paietta EM, Ferrando AA, Zhang J, Kern W, Haferlach T, Wu G, Dick JE, Klco JM, Haferlach C, Mullighan CG (2021) Enhancer hijacking drives oncogenic BCL11B expression in lineage ambiguous stem cell leukemia. Cancer Discover 11(11): 2846–2867. https://doi.org/10.1158/2159-8290.CD-21-0145 [PubMed] [PMC]

·      Moorman AV, Barretta E, Butler ER, Ward EJ, Twentyman K, Kirkwood AA, Enshaei A, Schwab C, Creasey T, Leongamornlert D, Papaemmanuil E, Patrick P, Clifton-Hadley L, Patel B, Menne T, McMillan AK, Harrison CJ, Rowntree CJ, Marks DI, Fielding AK (2022) Prognostic impact of chromosomal abnormalities and copy number alterations in adult B-cell precursor acute lymphoblastic leukaemia: A UKALL14 study. Leukemia 36(3): 625–636. https://doi.org/10.1038/s41375-021-01448-2 [PubMed] [PMC]

·      Moradveisi B, Muwakkit S, Zamani F, Ghaderi E, Mohammadi E, Zgheib NK (2019) ITPA, TPMT, and NUDT15 genetic polymorphisms predict 6-mercaptopurine toxicity in Middle Eastern children with acute lymphoblastic leukemia. Frontiers in Pharmacology 10: 916. https://doi.org/10.3389/fphar.2019.00916 [PubMed] [PMC]

·      Mullighan CG (2019) How advanced are we in targeting novel subtypes of ALL? Best Practice & Research Clinical Haematology 32(4): 101095. https://doi.org/10.1016/j.beha.2019.101095 [PubMed] [PMC]

·      Mullighan CG, Collins-Underwood JR, Phillips LA, Loudin MG, Liu W, Zhang J, Ma J, Coustan-Smith E, Harvey RC, Willman CL, Mikhail FM, Meyer J, Carroll AJ, Williams RT, Cheng J, Heerema NA, Basso G, Pession A, Pui CH, Raimondi SC, Hunger SP, Downing JR, Carroll WL, Rabin KR (2009) Rearrangement of CRLF2 in B-progenitor–and Down syndrome–associated acute lymphoblastic leukemia. Nature Genetics 41(11): 1243–1246. https://doi.org/10.1038/ng.469 [PubMed] [PMC]

·      Mullighan CG, Su X, Zhang J, Radtke I, Phillips LA, Miller CB, Ma J, Liu W, Cheng C, Schulman BA, Harvey RC, Chen IM, Clifford RJ, Carroll WL, Reaman G, Bowman WP, Devidas M, Gerhard DS, Yang W, Relling MV, Shurtleff SA, Campana D, Borowitz MJ, Pui CH, Smith M, Hunger SP, Willman CL, Downing JR; Children’s Oncology Group (2009) Deletion of IKZF1 and prognosis in acute lymphoblastic leukemia. The New England Journal of Medicine 360(5): 470480. https://doi.org/10.1056/NEJMoa0808253 [PubMed] [PMC]

·      Nakano T, Kobayashi R, Matsushima S, Hori D, Yanagi M, Suzuki D, Kobayashi K (2021) Risk factors for delayed elimination of high-dose methotrexate in childhood acute lymphoblastic leukemia and lymphoma. International Journal of Hematology 113(5): 744750. https://doi.org/10.1007/s12185-020-03071-w [PubMed]

·      Nersting J, Nielsen SN, Grell K, Paerregaard M, Abrahamsson J, Lund B, Jonsson OG, Pruunsild K, Vaitkeviciene G, Kanerva J, Schmiegelow K; Nordic Society of Paediatric Haematology and Oncology (NOPHO) (2019) Methotrexate polyglutamate levels and co-distributions in childhood acute lymphoblastic leukemia maintenance therapy. Cancer Chemotherapy and Pharmacology 83(1): 5360. https://doi.org/10.1007/s00280-018-3704-7 [PubMed]

·      Paganin M, Ferrando A (2011) Molecular pathogenesis and targeted therapies for NOTCH1-induced T-cell acute lymphoblastic leukemia. Blood Reviews 25(2): 8390. https://doi.org/10.1016/j.blre.2010.09.004 [PubMed] [PMC]

·      Pai AA, Young-Sing Q, Bera S, Maheshwari K, Misra A (2022) Analysis of clinical and patient-reported outcomes in post-ELAPE perineal reconstruction with IGAP flap - A 5-year review. JPRAS Open 34:1020. https://doi.org/10.1016/j.jpra.2022.06.002 [PubMed] [PMC] 

·      Paietta E, Roberts KG, Wang V, Gu Z, Buck GAN, Pei D, Cheng C, Levine RL, Abdel-Wahab O, Cheng Z, Wu G, Qu C, Shi L, Pounds S, Willman CL, Harvey R, Racevskis J, Barinka J, Zhang Y, Dewald GW, Ketterling RP, Alejos D, Lazarus HM, Luger SM, Foroni L, Patel B, Fielding AK, Melnick A, Marks DI, Moorman AV, Wiernik PH, Rowe JM, Tallman MS, Goldstone AH, Mullighan CG, Litzow MR (2021) Molecular classification improves risk assessment in adult BCR-ABL1-negative B-ALL. Blood Advances 138(11): 948–958. https://doi.org/10.1182/blood.2020010144 [PubMed] [PMC]

·      Parker H, An Q, Barber K, Case M, Davies T, Konn Z, Stewart A, Wright S, Griffiths M, Ross FM, Moorman AV, Hall AG, Irving JA, Harrison CJ, Strefford JC (2008) The complex genomic profile of ETV6-RUNX1 positive acute lymphoblastic leukemia highlights a recurrent deletion of TBL1XR1. Genes Chromosomes Cancer 47(12): 11181125. https://doi.org/10.1002/gcc.20613 [PubMed]

·      Perez-Andreu V, Roberts KG, Xu H, Smith C, Zhang H, Yang W, Harvey RC, Payne-Turner D, Devidas M, Cheng IM, Carroll WL, Heerema NA, Carroll AJ, Raetz EA, Gastier-Foster JM, Marcucci G, Bloomfield CD, Mrózek K, Kohlschmidt J, Stock W, Kornblau SM, Konopleva M, Paietta E, Rowe JM, Luger SM, Tallman MS, Dean M, Burchard EG, Torgerson DG, Yue F, Wang Y, Pui CH, Jeha S, Relling MV, Evans WE, Gerhard DS, Loh ML, Willman CL, Hunger SP, Mullighan CG, Yang JJ (2015) A genome-wide association study of susceptibility to acute lymphoblastic leukemia in adolescents and young adults. Blood 125(4): 680686. https://doi.org/10.1182/blood-2014-09-595744 [PubMed] [PMC]

·      Perez-Andreu V, Xu H, Yang JJ (2013) The novel susceptibility variants for childhood acute lymphoblastic leukemia. Journal of the National Cancer Institute 105(19): 15121513. https://doi.org/10.1093/jnci/djt230 [PubMed] [PMC]

·      Phillips NS, Cheung YT, Glass JO, Scoggins MA, Liu W, Ogg RJ, Mulrooney DA, Pui CH, Robison LL, Reddick WE, Hudson MM, Krull KR (2020) Neuroanatomical abnormalities related to dexamethasone exposure in survivors of childhood acute lymphoblastic leukemia. Pediatric Blood & Cancer 3: e27968. https://doi.org/10.1002/pbc.27968 [PubMed] [PMC]

·      Pieters R, Schrappe M, De Lorenzo P, Hann I, De Rossi G, Felice M, Hovi L, LeBlanc T, Szczepanski T, Ferster A, Janka G, Rubnitz J, Silverman L, Stary J, Campbell M, Li CK, Mann G, Suppiah R, Biondi A, Vora A, Valsecchi MG (2007) A treatment protocol for infants younger than 1 year with acute lymphoblastic leukaemia (Interfant-99): An observational study and a multicentre randomised trial. Lancet 370(9583): 240250. https://doi.org/10.1016/S0140-6736(07)61126-X [PubMed]

·      Pui CH (2015) Genomic and pharmacogenetic studies of childhood acute lymphoblastic leukemia. Frontiers of Medicine 9: 19. https://doi.org/10.1007/s11684-015-0381-3 [PubMed]

·      Pui CH, Nichols KE, Yang JJ (2018) Somatic and germline genomics in paediatric acute lymphoblastic leukaemia. Nature Reviews Clinical Oncology 16(4): 227–240. https://doi.org/10.1038/s41571-018-0136-6 [PubMed]

·      Raimondi SC, Behm FG, Roberson PK, Williams DL, Pui CH, Crist WM, Look AT, Rivera GK (1990) Cytogenetics of pre-B-cell acute lymphoblastic leukemia with emphasis on prognostic implications of the t(1;19). Journal of Clinical Oncology 8: 13801388. https://doi.org/10.1200/JCO.1990.8.8.1380 [PubMed]

·      Roberts KG (2017) The biology of Philadelphia chromosome-like ALL. Best Practice & Research Clinical Haematology 30(3): 212–221. https://doi.org/10.1016/j.beha.2017.07.003 [PubMed]

·      Roberts KG, Li Y, Payne-Turner D, Harvey RC, Yang YL, Pei D, McCastlain K, Ding L, Lu C, Song G, Ma J, Becksfort J, Rusch M, Chen SC, Easton J, Cheng J, Boggs K, Santiago-Morales N, Iacobucci I, Fulton RS, Wen J, Valentine M, Cheng C, Paugh SW, Devidas M, Chen IM, Reshmi S, Smith A, Hedlund E, Gupta P, Nagahawatte P, Wu G, Chen X, Yergeau D, Vadodaria B, Mulder H, Winick NJ, Larsen EC, Carroll WL, Heerema NA, Carroll AJ, Grayson G, Tasian SK, Moore AS, Keller F, Frei-Jones M, Whitlock JA, Raetz EA, White DL, Hughes TP, Guidry Auvil JM, Smith MA, Marcucci G, Bloomfield CD, Mrózek K, Kohlschmidt J, Stock W, Kornblau SM, Konopleva M, Paietta E, Pui CH, Jeha S, Relling MV, Evans WE, Gerhard DS, Gastier-Foster JM, Mardis E, Wilson RK, Loh ML, Downing JR, Hunger SP, Willman CL, Zhang J, Mullighan CG (2014) Targetable kinase-activating lesions in Ph-like acute lymphoblastic leukemia. The New England Journal of Medicine 371(11): 1005–1015. https://doi.org/10.1056/NEJMoa1403088 [PubMed] [PMC]

·      Roberts KG, Morin RD, Zhang J, Hirst M, Zhao Y, Su X, Chen SC, Payne-Turner D, Churchman ML, Harvey RC, Chen X, Kasap C, Yan C, Becksfort J, Finney RP, Teachey DT, Maude SL, Tse K, Moore R, Jones S, Mungall K, Birol I, Edmonson MN, Hu Y, Buetow KE, Chen IM, Carroll WL, Wei L, Ma J, Kleppe M, Levine RL, Garcia-Manero G, Larsen E, Shah NP, Devidas M, Reaman G, Smith M, Paugh SW, Evans WE, Grupp SA, Jeha S, Pui CH, Gerhard DS, Downing JR, Willman CL, Loh M, Hunger SP, Marra MA, Mullighan CG (2012) Genetic alterations activating kinase and cytokine receptor signaling in high-risk acute lymphoblastic leukemia. Cancer Cell 22(2): 153–166. https://doi.org/10.1016/j.ccr.2012.06.005 [PubMed] [PMC]

·      Roberts KG, Pei D, Campana D, Payne-Turner D, Li Y, Cheng C, Sandlund JT, Jeha S, Easton J, Becksfort J, Zhang J, Coustan-Smith E, Raimondi SC, Leung WH, Relling MV, Evans WE, Downing JR, Mullighan CG, Pui CH (2014) Outcomes of children with BCR-ABL1-like acute lymphoblastic leukemia treated with risk-directed therapy based on the levels of minimal residual disease. Journal of Clinical Oncology 32(27): 30123020. https://doi.org/10.1200/JCO.2014.55.4105 [PubMed] [PMC]

·      Rosen G, Harris AK, Liu M, Dreyfus J, Krueger J, Messinger YH (2015) The effects of dexamethasone on sleep in young children with acute lymphoblastic leukemia. Sleep Medicine 16(4): 503509. https://doi.org/10.1016/j.sleep.2014.11.002 [PubMed] [PMC]

·      Rudin S, Marable M, Huang RS (2017) The promise of pharmacogenomics in reducing toxicity during acute lymphoblastic leukemia maintenance treatment. Genomics Proteomics Bioinformatics 15(2): 82–93. https://doi.org/10.1016/j.gpb.2016.11.003 [PubMed] [PMC]

·      Russell LJ, Capasso M, Vater I, Akasaka T, Bernard OA, Calasanz MJ, Chandrasekaran T, Chapiro E, Gesk S, Griffiths M, Guttery DS, Haferlach C, Harder L, Heidenreich O, Irving J, Kearney L, Nguyen-Khac F, Machado L, Minto L, Majid A, Moorman AV, Morrison H, Rand V, Strefford JC, Schwab C, Tönnies H, Dyer MJ, Siebert R, Harrison CJ (2009) Deregulated expression of cytokine receptor gene, CRLF2, is involved in lymphoid transformation in B-cell precursor acute lymphoblastic leukemia. Blood 114(13): 26882698. https://doi.org/10.1182/blood-2009-03-208397 [PubMed]

·      Russell LJ, Jones L, Enshaei A, Tonin S, Ryan SL, Eswaran J, Nakjang S, Papaemmanuil E, Tubio JM, Fielding AK, Vora A, Campbell PJ, Moorman AV, Harrison CJ (2017) Characterisation of the genomic landscape ofCRLF2-rearranged acute lymphoblastic leukemia. Genes, Chromosomes and Cancer 56(5): 363–372. https://doi.org/10.1002/gcc.22439 [PubMed] [PMC]

·      Schmiegelow K (2009) Advances in individual prediction of methotrexate toxicity: A review. British Journal of Hematology 146(5): 489503. https://doi.org/10.1111/j.1365-2141.2009.07765.x [PubMed]

·      Schmiegelow K, Nielsen SN, Frandsen TL, Nersting J (2014) Mercaptopurine/ Methotrexate maintenance therapy of childhood acute lymphoblastic leukemia: Clinical facts and fiction. Journal of Pediatric Hematology/Oncology 36(7): 503–517. https://doi.org/10.1097/MPH.0000000000000206 [PubMed] [PMC]

·      Schwab C, Harrison CJ (2018) Advances in B-cell precursor acute lymphoblastic leukemia genomics. HemaSphere 2(4): e53. https://doi.org/10.1097/HS9.0000000000000053 [PubMed] [PMC]

·      Seki M, Kimura S, Isobe T, Yoshida K, Ueno H, Nakajima-Takagi Y, Wang C, Lin L, Kon A, Suzuki H, Shiozawa Y, Kataoka K, Fujii Y, Shiraishi Y, Chiba K, Tanaka H, Shimamura T, Masuda K, Kawamoto H, Ohki K, Kato M, Arakawa Y, Koh K, Hanada R, Moritake H, Akiyama M, Kobayashi R, Deguchi T, Hashii Y, Imamura T, Sato A, Kiyokawa N, Oka A, Hayashi Y, Takagi M, Manabe A, Ohara A, Horibe K, Sanada M, Iwama A, Mano H, Miyano S, Ogawa S, Takita J (2017) Recurrent SPI1 (PU.1) fusions in high-risk pediatric T cell acute lymphoblastic leukemia. Nature Genetics 49(8): 1274–1281. https://doi.org/10.1038/ng.3900 [PubMed]

·      Shen Y, Wang Z, Zhou F, Jin R (2021) The influence of MTHFR genetic polymorphisms on methotrexate therapy in pediatric acute lymphoblastic leukemia. Open Life Science 16(1): 12031212. https://doi.org/10.1515/biol-2021-0121 [PubMed] [PMC]

·      Sin CF, Man PM (2021) Early T-Cell precursor acute lymphoblastic leukemia: Diagnosis, updates in molecular pathogenesis, management, and novel therapies. Frontiers in Oncology 11: 750789. https://doi.org/10.3389/fonc.2021.750789 [PubMed] [PMC]

·      Stanulla M, Schrappe M, Yang SK (2009) Treatment of childhood acute lymphoblastic leukemia. Seminar Hematology 46(1): 52–63. https://doi.org/10.1053/j.seminhematol.2008.09.007 [PubMed]

·      Tanasi I, Ba I, Sirvent N, Braun T, Cuccuini W, Ballerini P, Duployez N, Tanguy-Schmidt A, Tamburini J, Maury S, Doré E, Himberlin C, Duclos C, Chevallier P, Rousselot P, Bonifacio M, Cavé H, Baruchel A, Dombret H, Soulier J, Landman-Parker J, Boissel N, Clappier E (2019) Efficacy of tyrosine kinase inhibitors in Ph-like acute lymphoblastic leukemia harboring ABL-class rearrangements. Blood 134(16): 1351–1355. https://doi.org/10.1182/blood.2019001244 [PubMed]

·      Tarantini F, Cumbo C, Anelli L, Zagaria A, Specchia G, Musto P, Albano F (2021) Inside the biology of early T-cell precursor acute lymphoblastic leukemia: the perfect trick. Biomark Research 9(1): 89. https://doi.org/10.1186/s40364-021-00347-z [PubMed] [PMC]

·      Tasian SK, Loh ML, Hunger SP (2015) Childhood acute lymphoblastic leukemia: Integrating genomics into therapy. Cancer 121(20): 35773590. https://doi.org/10.1002/cncr.29573 [PubMed] [PMC]

·      Tolbert JA, Bai S, Abdel-Rahman SM, August KJ, Weir SJ, Kearns GL, Neville KA (2017) Pharmacokinetics of two 6-mercaptopurine liquid formulations in children with acute lymphoblastic leukemia. Pediatric Blood & Cancer 8: https://doi.org/10.1002/pbc.26465 [PubMed]

·      Ueno H, Yoshida K, Shiozawa Y, Nannya Y, Iijima-Yamashita Y, Kiyokawa N, Shiraishi Y, Chiba K, Tanaka H, Isobe T, Seki M, Kimura S, Makishima H, Nakagawa MM, Kakiuchi N, Kataoka K, Yoshizato T, Nishijima D, Deguchi T, Ohki K, Sato A, Takahashi H, Hashii Y, Tokimasa S, Hara J, Kosaka Y, Kato K, Inukai T, Takita J, Imamura T, Miyano S, Manabe A, Horibe K, Ogawa S, Sanada M (2020) Landscape of driver mutations and their clinical impacts in pediatric B-cell precursor acute lymphoblastic leukemia. Blood Advances 4(20): 5165–5173. https://doi.org/10.1182/bloodadvances.2019001307 [PubMed] [PMC]

·      Umerez M, Gutierrez-Camino Á, Muñoz-Maldonado C, Martin-Guerrero I, Garcia-Orad A (2017) MTHFR polymorphisms in childhood acute lymphoblastic leukemia: Influence on methotrexate therapy. Pharmgenomics and Personalized Medicine 10: 69-78. https://doi.org/10.2147/PGPM.S107047 [PubMed] [PMC]

·      Van Vlierberghe P, Palomero T, Khiabanian H, Van der Meulen J, Castillo M, Van Roy N, De Moerloose B, Philippé J, González-García S, Toribio ML, Taghon T, Zuurbier L, Cauwelier B, Harrison CJ, Schwab C, Pisecker M, Strehl S, Langerak AW, Gecz J, Sonneveld E, Pieters R, Paietta E, Rowe JM, Wiernik PH, Benoit Y, Soulier J, Poppe B, Yao X, Cordon-Cardo C, Meijerink J, Rabadan R, Speleman F, Ferrando A (2010) PHF6 mutations in T-cell acute lymphoblastic leukemia. Nature Genetics 42(4): 338342. https://doi.org/10.1038/ng.542 [PubMed] [PMC]

·      Vora A, Goulden N, Wade R, Mitchell C, Hancock J, Hough R, Rowntree C, Richards S (2013) Treatment reduction for children and young adults with low-risk acute lymphoblastic leukaemia defined by minimal residual disease (UKALL 2003): A randomised controlled trial. Lancet Oncology 14(3): 199209. https://doi.org/10.1016/S1470-2045(12)70600-9 [PubMed]

·      Wang DS, Yu CH, Lin CY, Chang YH, Lin KH, Lin DT, Jou ST, Lu MY, Chang HH, Lin SW, Chen HY, Yang YL (2021) Childhood acute lymphoblastic leukemia mercaptopurine intolerance is associated with NUDT15 variants. Pediatric Research 89(1): 217222. https://doi.org/10.1038/s41390-020-0868-8 [PubMed]

·      Wiley JS, Jones SP, Sawyer WH, Paterson AR (1982) Cytosine arabinoside influx and nucleoside transport sites in acute leukemia. Journal of Clinical Investigation 69(2): 479489. https://doi.org/10.1172/jci110472 [PubMed] [PMC]

·      Winter SS, Dunsmore KP, Devidas M, Eisenberg N, Asselin BL, Wood BL, Leonard Rn MS, Murphy J, Gastier-Foster JM, Carroll AJ, Heerema NA, Loh ML, Raetz EA, Winick NJ, Carroll WL, Hunger SP (2015) Safe integration of nelarabine into intensive chemotherapy in newly diagnosed T-cell acute lymphoblastic leukemia: children’s oncology group study AALL0434. Pediatric Blood & Cancer 62(7): 11761183. https://doi.org/10.1002/pbc.25470 [PubMed] [PMC]

·      Wu C, Li W (2018) Genomics and pharmacogenomics of pediatric acute lymphoblastic leukemia. Critical Reviews in Oncology/Hematology 126: 100111. https://doi.org/10.1016/j.critrevonc.2018.04.002 [PubMed]

·      Yang W, Cai J, Shen S, Gao J, Yu J, Hu S, Jiang H, Fang Y, Liang C, Ju X, Wu X, Zhai X, Tian X, Wang N, Liu A, Jiang H, Jin R, Sun L, Yang M, Leung AWK, Pan K, Zhang Y, Chen J, Zhu Y, Zhang H, Li C, Yang JJ, Cheng C, Li CK, Tang J, Zhu X, Pui CH (2021) Pulse therapy with vincristine and dexamethasone for childhood acute lymphoblastic leukaemia (CCCG-ALL-2015): an open-label, multicentre, randomised, phase 3, non-inferiority trial. Lancet Oncology 22(9): 13221332. https://doi.org/10.1016/S1470-2045(21)00328-4 [PubMed] [PMC]

·      Yasuda T, Tsuzuki S, Kawazu M, Hayakawa F, Kojima S, Ueno T, Imoto N, Kohsaka S, Kunita A, Doi K, Sakura T, Yujiri T, Kondo E, Fujimaki K, Ueda Y, Aoyama Y, Ohtake S, Takita J, Sai E, Taniwaki M, Kurokawa M, Morishita S, Fukayama M, Kiyoi H, Miyazaki Y, Naoe T, Mano H (2016) Recurrent DUX4 fusions in B cell acute lymphoblastic leukemia of adolescents and young adults. Nature Genetics 48(5): 569–574. https://doi.org/10.1038/ng.3535 [PubMed]

·      Yoda A, Yoda Y, Chiaretti S, Bar-Natan M, Mani K, Rodig SJ, West N, Xiao Y, Brown JR, Mitsiades C, Sattler M, Kutok JL, DeAngelo DJ, Wadleigh M, Piciocchi A, Dal Cin P, Bradner JE, Griffin JD, Anderson KC, Stone RM, Ritz J, Foà R, Aster JC, Frank DA, Weinstock DM (2009) Functional screening identifies CRLF2 in precursor B-cell acute lymphoblastic leukemia. Proc Natl Acad Sci U S A 107(1): 252–257. https://doi.org/10.1073/pnas.0911726107 [PubMed] [PMC]

·      Zaliova M, Stuchly J, Winkowska L, Musilova A, Fiser K, Slamova M, Starkova J, Vaskova M, Hrusak O, Sramkova L, Stary J, Zuna J, Trka J (2019) Genomic landscape of pediatric B-other acute lymphoblastic leukemia in a consecutive European cohort. Haematologica 104(7): 1396–1406. https://doi.org/10.3324/haematol.2018.204974 [PubMed] [PMC]

·      Zhang J, Mullighan CG, Harvey RC, Wu G, Chen X, Edmonson M, Buetow KH, Carroll WL, Chen IM, Devidas M, Gerhard DS, Loh ML, Reaman GH, Relling MV, Camitta BM, Bowman WP, Smith MA, Willman CL, Downing JR, Hunger SP (2011) Key pathways are frequently mutated in high-risk childhood acute lymphoblastic leukemia: A report from the Children’s Oncology Group. Blood 118(11): 30803087. https://doi.org/10.1182/blood-2011-03-341412 [PubMed] [PMC]

·      Zhou Y, Wang L, Zhai XY, Wen L, Tang F, Yang F, Liu XT, Dong L, Zhi LJ, Shi HY, Hao GX, Zheng Y, Jacqz-Aigrain E, Wang TY, Zhao W (2020) Precision therapy of 6-mercaptopurine in Chinese children with acute lymphoblastic leukaemia. British Journal of Clinical Pharmacology 86(8): 15191527. https://doi.org/10.1111/bcp.14258 [PubMed] [PMC]

·      Zhu KW, Chen P, Zhang DY, Yan H, Liu H, Cen LN, Liu YL, Cao S, Zhou G, Zeng H, Chen SP, Zhao XL, Chen XP (2018) Association of genetic polymorphisms in genes involved in Ara-C and dNTP metabolism pathway with chemosensitivity and prognosis of adult acute myeloid leukemia (AML). Journal of Translational Medicine 16(1): 90. https://doi.org/10.1186/s12967-018-1463-1 [PubMed] [PMC]

·      Zou Y, Mei D, Yuan J, Han J, Xu J, Sun N, He H, Yang C, Zhao L (2021) Preparation, characterization, pharmacokinetic, and therapeutic potential of novel 6-mercaptopurine-loaded oral nanomedicines for acute lymphoblastic leukemia. International Journal of Nanomedicine 16: 11271141. https://doi.org/10.2147/IJN.S290466 [PubMed] [PMC]

Author Сontributions

Violeta Grajqevci-Uka, PhD in Medicine, Faculty of Medicine, Department of Pediatrics, University Clinical Center, Kosovo; e-mail: violetagrajqevci@hotmail.com; ORCID ID https://orcid.org/0000-0002-3653-1407. This author designed the idea of the study, formulated its concept, analyzed the results, formulated the conclusions and wrote the manuscript.

Emir Behluli, PhD in Medicine, Faculty of Medicine, Department of Pediatrics, University Clinical Center, Kosovo; e-mail: ebehluli_19@hotmail.com; ORCID ID https://orcid.org/0000-0002-0889-5510. This author designed the idea of the study, formulated its concept, analyzed the results, formulated the conclusions and wrote the manuscript.

Rifat Hadziselimovic, PhD in Genetics, Professor, Faculty of Sciences, Department of Biology, University of Sarajevo, Bosnia and Herzegovina; e-mail: rifat.hadziselimovic@gmx.net; ORCID ID https://orcid.org/0000-0002-6590-8824. This author supervised the entire study from the idea conception to conclusions.

Thomas Liehr, PhD der Naturwiessenschaft in Professor of Molecular Cytogenetics, Faculty of Medicine, Jena University Hospital, Friedrich Schiller University, Institute of Human Genetics, Germany; e-mail: thomas.liehr@med.uni-jena.de; ORCID ID https://orcid.org/0000-0003-1672-3054. This author reviewed and edited the manuscript.

Gazmend Temaj, PhD in Molecular Medicine, Professor, Faculty of Pharmacy Department of Human Genetics, UBT College, Kosovo; e-mail: gazmend.temaj@ubt-uni.net; ORCID ID https://orcid.org/0000-0003-4807-2938. This author supervised the entire study from the idea conception to conclusions.