1 Introduction
B-cell precursor acute lymphoblastic leukemia (BCP-ALL) is a blood cancer that originates from B-lymphoid progenitors [
1,
2]. Genetic susceptibility and somatic clonal expansion (tumor acquired) are the hallmarks and biological basis of BCP-ALL. The enrichment of chromosomal alterations, including genomic translocations and entire chromosome losses or gains (aneuploidy), DNA copy number variations (CNVs), and sequence mutations, are common in leukemic blast cells of BCP-ALLs [
3–
7]. In the past few years, numerous molecular subtypes with distinct genetic abnormalities and clinical significance have been identified in multicenter, global collaboration cohort studies of patients with BCP-ALL [
8–
12]. A recognition of these multifaceted genetic alterations contributing to leukemogenesis is vital in ensuring a precise risk stratification of the disease, which can then lead to improvements to cure rates in both adults and children with BCP-ALL [
5,
12–
26]. Before the 1980s, with the combined effectiveness of health-care systems and the absence of actionable risk stratification factors, the five-year overall survival rate was only approximately 50% in children patients with BCP-ALL [
27–
29]. In recent years, the released data from multiple large-cohort clinical trials indicate a five-year overall survival rate that is higher than 90% in children [
29–
42]. In addition, the rapid advancement in new drugs and agents, including target inhibitors, i.e., tyrosine kinase inhibitors (TKIs) and epigenetic inhibitors, immunomodulators, monoclonal antibodies, and chimeric antigen receptor T-cells (CAR-T), may further improve the prognosis of relapsed or refractory (R/R) BCP-ALL [
13,
43].
2 Overview of well-established molecular subtypes in BCP-ALL
The translocation t(9;22)(q34;q11.2) resulting in
BCR-
ABL1 (Philadelphia, Ph) fusion accounted for 15%–25% of adults and 2%–5% of children with BCP-ALL[
8,
9,
44].
BCR-
ABL1 fusion proteins are considered to be signaling regulators and can trigger kinase pathway activation [
45–
48]. Targeted treatments with TKIs have significantly improved the prognosis of patients with
BCR-
ABL1 [
8,
9,
13,
49,
50].
KMT2A (
MLL) rearrangements account for 3%–4% of children with BCP-ALL, particularly in infants (<1 year old), and in 4%–10% of adult patients, mainly>30 years old, with BCP-ALL [
8,
9,
18,
24,
51,
52]. Additionally, this rearrangement is typically associated with poor prognosis [
24,
52,
53]. The translocations t(4;11)(q21;q23), t(11;19)(q23;p13.3), and t(9;11)(p21;q23) resulting in
KMT2A-
AFF1,
KMT2A-
MLLT1, and
KMT2A-
MLLT3, respectively, account for more than 90% of
KMT2A fusions in BCP-ALL [
8,
9]. The translocation t(12;21)(p13;q22) resulting in
ETV6-
RUNX1 (also known as
TEL-
AML1) comprises approximately 15%–25% of children with BCP-ALL [
8,
9,
17,
18]. However, adults (<1%) and patients with
ETV6-
RUNX1 rarely have the same sensitivity to chemotherapy, and relapse and death events are less common [
5,
8,
9,
17,
54,
55]. In addition,
ETV6-
RUNX1 fusion is detectable in 5% healthy newborns based on DNA-based genomic inverse PCR for exploration of ligated breakpoint screening, but most never transform to BCP-ALL [
56]. Transcription factor 3 (
TCF3) is frequently involved in two types of translocations, namely, t(1;19)(q23;p13) and t(17;19)(q22;p13), which result in
TCF3-
PBX1 and
TCF3-
HLF fusions, respectively.
TCF3-
PBX1 accounts for approximately 4%–7% of children patients and 2%–5% of adult patients with BCP-ALL and is associated with intermediate outcomes [
8,
9,
12,
54].
TCF3-
HLF is a rare gene fusion (<1% of ALL), and relapse and death events are common [
25,
57].
HLF is also involved in translocation t(18;19)(q21;p13), thereby resulting in
TCF4-
HLF. Both
TCF3-
HLF and
TCF4-
HLF fusions retain the bZIP_2 domain and accompany the overexpression of
HLF [
8,
9,
25]. The complex intrachromosomal amplification of chromosome 21 (iAMP21), which was first reported in 2003, helped to define a distinct cytogenetic subgroup [
58,
59]. This amplification occurs in approximately 2%–3% of children with B-lineage ALL, especially in adolescence [
5,
9,
12]. This condition was originally considered a rare, high-risk subtype of BCP-ALL, but current intensive therapy has greatly improved its outcome [
59,
60]. High hyperdiploidy with a gain of at least five chromosomes is another independent predictor (along with
ETV6-
RUNX1) of favorable outcomes and accounts for approximately 15%–25% of patients with childhood BCP-ALL. However, high hyperdiploidy is rare in adults (<1%) [
8,
9]. Conversely, hypodiploid (<44 chromosomes) is uncommon (2%–3%) and associated with inferior outcomes compared with high hyperdiploidy BCP-ALL. In addition, hypodiploid (<44 chromosomes) is heterogeneous with distinct genetic abnormalities and gene expression profiles consisting of near-haploid BCP-ALL (25–29 chromosomes), low hypodiploid (32–39 chromosomes), and high hypodiploid (40–43 chromosomes) (Table 1) [
5,
12,
23].
The well-established molecular subtypes of BCP-ALL have been mostly integrated into the guidance of therapy options (e.g., dose and times of chemotherapy and types of targeted drugs) [
13]. Their integration has significantly improved the long-term survival of both adult and children patients. However, subtypes with poor/intermediate prognosis, including
BCR-
ABL1,
KMT2A fusions,
TCF3-
PBX1,
HLF fusions, and low hypodiploid, are needed in intensive chemotherapy [
5].
BCR-
ABL1 and
KMT2A fusions not only occur in BCP-ALL but also in other types of acute or chronic leukemia, including acute myeloid leukemia (AML), mixed phenotype acute leukemia (MPAL), and chronic myeloid leukemia [
61–
63]. A potential supposition is that the fusion of genes may occur at different stages of hematopoietic stem cell development.
3 Emerging molecular subtypes in BCP-ALL
3.1 DUX4, MEF2D, and ZNF384 gene fusions
Three new subtypes of adult and childhood BCP-ALL have been described recently [
16,
18,
20–
22,
64]. The subtypes are involved in the rearrangements of DNA binding factors, including double homeobox 4 gene (
DUX4), myocyte enhancer factor 2D (
MEF2D), and zinc finger protein 384 (
ZNF384), which separately account for approximately 4%–7%, 2%–4%, and 3%–5% of childhood BCP-ALL and 4%–7%, 2%–7%, and 3%–8% of adult BCP-ALL [
8,
9,
15–
18,
20–
22,
26,
65–
67].
DUX4-
r,
ZNF384-
r, and
MEF2D-
r BCP-ALL are associated with favorable, intermediate, and poor diagnoses, respectively. In most patients,
DUX4 overexpression is the consequence of
DUX4 fusion, typically
IGH but rarely
ERG [
17,
20,
21]. By contrast, the partner genes of
MEF2D and
ZNF384 are complex and diverse. A total of nine genes (
BCL9,
SS18,
FOXJ2,
CSF1R,
DAZAP1,
STAT6,
HNRNPUL1,
HNRNPH1, and
HNRNPM) and 11 genes (
EP300,
TCF3,
TAF15,
CREBBP,
EWSR1,
ARID1B,
SMARCA2,
SMARCA4,
SYNRG,
NIPBL, and
CLTC) have been reported as fusion partners of
MEF2D and
ZNF384 [
8,
15–
18,
20,
22,
66–
68], respectively. Translocations t(1;1)(q21;q22) and t(1;19)(q22;q13) resulting in
MEF2D-BCL9 and
MEF2D-
HNRNPUL1, respectively, are the top two items in
MEF2D-
r BCP-ALL, accounting for 70% and 15% of the cases.
EP300,
TCF3, and
TAF15 are the most common 5′ genes rearranged to
ZNF384 with translocations t(12;22)(p13;q13), t(12;19)(p13;p13), and t(12;17)(p13;q11), representing approximately 50%, 10%–20%, and 10% of
ZNF384 fusion-positive cases.
DUX4, located within a D4Z4 repeat array in the sub-telomeric region of chromosome 4, is a key transcription factor regulating embryonic development and is not expressed in normal B-cells. DUX4 can bind a large percentage of activated genes in early developing embryos and improve the accessibility of genes as early as the 2- to 4-cell stages [
20,
21,
69,
70]. Intragenic deletions of ERG were previously reported in approximately 5% of children with BCP-ALLs, but they were designated as the biological feature in
DUX4-
r cases [
5,
21]. Functional studies show that
DUX4 rearrangements, as an early initiating event in leukemogenesis, can bind to an intragenic region of
ERG and cause the overexpression of ERGalt with a noncanonical first exon and transcript. Aberrant ERGalt preserves the DNA binding and transactivating domains and encodes a truncated C-terminal
ERG protein. The transcriptional activity of wild-type
ERG is inhibited by the ERGalt protein-transforming leukemic blast cells [
5,
20,
21,
71]. Interestingly, despite the presence of approximately 40%–50% of genetic alterations in
IKZF1 deletions related to poor outcomes in other subtypes with BCP-ALL [
72],
DUX4-
r BCP-ALL can achieve excellent outcome [
5,
12,
17,
18,
21]. Recent studies have reported that
IGH-
DUX4 translocation occurs on the silenced IGH allele, thereby reducing the oncogenic stress of
DUX4’s high-level expression. The ERG deletions have a positive impact on the prognosis (ERG deletion positive/negative: five-year EFS 93%/68%,
P = 0.022; five-year OS 97%/75%,
P = 0.029) [
73,
74].
MEF2D is a transcription factor that can specifically bind to the myocyte-specific enhancer factor 2 (MEF2) element 5′-YTA[AT](4)TAR-3′. It belongs to the MEF2 gene family that contributes to the differentiation of muscle and neural cells, cardiac morphogenesis, formation of blood vessels, growth factor responsiveness, survival of neuronal cells, and acute leukemia [
75,
76]. As a member of the MEF2 gene family,
MEF2C is an activated oncogene in early T-cell precursor ALL, a subtype of high risk T-lineage ALL [
12,
77–
82]. In 2005,
MEF2D-
DAZAP1 was reported in the TS-2 cell line, as established from a three-year-old girl with ALL; it was found to contain t(1;19)(q23;p13.3) but was lacking the
TCF3-
PBX1 fusion [
83]. In 2016, multiple research groups who used RNA-seq recognized simultaneously the
MEF2D fusion in a subgroup of patients with BCP-ALL [
16,
18,
20,
64]. These studies determined that
MEF2D fusions can retain the MADS box domain required to mediate DNA binding with enhanced
MEF2D transcriptional activity. The heterogeneous nuclear ribonucleoproteins, including HNRNPUL1, HNRNPH1, and HNRNPM, are involved in
MEF2D-
r ALL [
8,
9,
15]. These proteins can bind RNA and are associated with pre-mRNA processing in the nucleus. Histone deacetylase 9 (
HDAC9), a target gene of
MEF2D, is significantly upregulated in
MEF2D-r patients, providing an optional therapeutic strategy by using histone deacetylase inhibitors, such as panobinostat [
5,
16,
18]. Staurosporine and venetoclax have recently been reported to be effective in inducing the caspase-dependent proteolysis of MEF2D-fusion proteins and apoptosis in MEF2D-fusion
+ ALL cells [
84].
ZNF384, also called
CIZ or
NMP4, can encode a C2H2-type zinc finger protein, whose function remains largely elusive, although it may function as a transcription factor.
ZNF384 fusions are often diagnosed as BCP-ALLs with an expression of cell surface markers of myeloid lineage (CD13 and CD33) or as B/myeloid (B/M) MPAL comprising approximately 50% of B/M MPAL [
63]. In 2002, seven cases with
ZNF384-
r acute leukemia and recurrent
EWSR1-
ZNF384 and
TAF15-
ZNF384 were reported [
85]. On the basis of large-cohort RNA-seq data analysis,
ZNF384-positive patients with BCP-ALL were identified as a prognostic subtype with distinct gene expression features [
8,
9,
16–
18,
20,
22,
65,
66,
86,
87]. All
ZNF384 fusions keep their entire coding region. Cell apoptotic response, MAPK signaling, and JAK-STAT signaling pathways are significantly upregulated in this subtype [
8,
22]. Cardiotrophin-like cytokine factor 1 (
CLCF1) is one of the most upregulated genes in
ZNF384-
r BCP-ALL that can bind to
CRLF1 to form a compound cytokine, thereby ultimately activating the JAK-STAT signaling pathway and B-cell proliferation
in vivo [
22,
88]. In addition, up to 60% of patients with
ZNF384 fusions also show alterations in their signaling molecules, such as
NRAS and
FLT3, and 40% have epigenetic mutations, particularly
SETD1B,
CREBBP, and
EZH2.
3.2 “Like” or “phenocopy” subtypes in BCP-ALL
Somatic mutations in the coding region and gene expression profile can help us recognize “like” or “phenocopy” subtypes that share similar gene expression features but lack consistent biomarkers. In recent years, several multi-omics and large-cohort studies have identified such subtypes, including Ph-like,
ETV6-
RUNX1-like,
KMT2A-like, and
ZNF384-like [
8,
9]. Integrated datasets showed that Ph-like and
ETV6-
RUNX1-like account for approximately 6%–15% and 2%–3% of children patients and 20%–25% and<1% of adult patients with BCP-ALL. However, both
KMT2A-like and
ZNF384-like are rare (<1%). Ph-like and
ETV6-
RUNX1-like are associated with poor and intermediate outcomes. The prognoses of
KMT2A-like and
ZNF384-like remain unclear because of limitation of sample size [
8,
9,
12].
The Ph-like subtype is
BCR-
ABL1 negative, but its gene expression feature is similar to that of
BCR-
ABL1-positive patients [
89–
93]. The Ph-like subtype was registered in the 2016 revision to the WHO classification of myeloid neoplasms and acute leukemia [
19]. Rearrangements of
CRLF2 (e.g.,
IGH-
CRLF2 and
P2RY8-
CRLF2 at
approximately 30%–50%),
ABL1/
ABL2 (10%),
JAK2 (10%), the erythropoietin receptor gene (
EPOR, 5%–10%), and
PDGFRB (5%) are the major chromosomal markers. Sequence mutations of signaling molecules, such as JAK-STAT signaling and Ras signaling (e.g.,
NRAS,
KRAS,
JAK2, and
PTPN11), account for up to 15%–20% of Ph-like cases [
8,
9].
BCR-
ABL1 and Ph-like are associated with poor response to chemotherapy but are sensitive to TKIs, such as imatinib and dasatinib.
The
ETV6-
RUNX1-like subtype was defined as
ETV6-
RUNX1 fusion negative and coexists with other
ETV6 and
IKZF1 alterations [
17], accounting for approximately 10%–20% of the
ETV6-
RUNX1/-like subtype. The
ETV6-
RUNX1-like subtype is also significantly enriched in children patients, and more than 80% of cases of
ETV6-
RUNX1-like subtype are children [
8,
9].
The
KMT2A-like and
ZNF384-like are recently defined subtypes, accounting for 5%–15% and 7%–10% of the
KMT2A/-like and
ZNF384/-like [
8,
9]. Rare fusions, such as
MED12-
HOXA9 and
AFF1-
TMEM156, are found in
KMT2A-like cases. Notably,
HOXA9 is deregulated in
KMT2A fusion-positive patients, and
AFF1 is the most common partner gene in
KMT2A fusions [
8,
9]. Recurrent
ZNF362 fusions (
SMARCA2-
ZNF362 and
TAF15-
ZNF362) are found in
ZNF384-like ALL. The fusion partner genes, including
SMARCA2 and
TAF15, and the sequence mutations of
ZEB2,
CREBBP, and
SETD1B, are found in both
ZNF384-r and
ZNF362-
r BCP-ALL. The zinc finger domains are retained in both fusion proteins [
8].
The Ph-like subtype can successfully exemplify significant improvements in prognosis by seeking the phenocopied events. However, extra effort is required to identify other possible phenocopied subtypes (e.g.,
HLF fusion-like and hypodiploid-like subtypes) [
8]. Moreover, the natural history and cell-of-origin of these “like” or “phenocopy” subtypes are still largely elusive. These gaps may prevent researchers from understanding the potential prognostic factors. A comprehensive comparison of the subtypes sharing similar gene expression profiles in many other aspects, such as cancer cell population, inherited or
de novo variants within a noncoding region, aberrant splicing, bacterial and viral infections, chromatin accessibility, and/or epigenetics alterations, may provide more hints [
27,
87,
94–
105].
3.3 PAX5-driven subtypes: PAX5alt and PAX5 p.Pro80Arg
Paired box 5 (
PAX5) is a member of the paired box (PAX) family and is an early B-lineage-specific transcriptional activator protein. Genetic alterations of
PAX5, including DNA CNVs, sequence mutations, and chromosomal translocations, are common in patients with BCP-ALL [
106]. CNVs and non-silent sequence mutations of
PAX5 occur in about 30% and 5%–9% of all BCP-ALL patients [
8,
9,
18]. Chromosomal translocations of
PAX5, which result in fusion genes, account for approximately 5%–7% of children and 2%–4% of adults with BCP-ALL [
8,
107].
PAX5 fusions are highly heterogeneous and complex. At least 24 partner genes (e.g.,
PAX5-
JAK2,
PAX5-
ETV6, and
PAX5-
NOL4L) of
PAX5 fusions are involved in
PAX5 rearrangements, thereby resulting in the expression of chimeric in-frame fusion transcript [
9]. Furthermore, 15%–25% of patients with
PAX5 fusions have presented Ph-like subtype characteristics, mainly those of
PAX5-JAK2 and
PAX5-
ZCCHC7. Nearly 20% of
PAX5 fusions coexist with other chromosomal alterations, including
CRLF2 fusions,
ETV6-
RUNX1,
TCF3-
PBX1,
KMT2A,
BCR-
ABL1, and iAMP21. For instance,
CRLF2 fusions coexist in up to 10% of all
PAX5 fusions, such as
PAX5-
NOL4L,
PAX5-
AUTS2,
PAX5-
NCOA5, and
P2RY8-
CRLF2 pairs. Notably, these fusions have shown different gene expression profiles compared with the
CRLF2 fusions clustered to the
BCR-
ABL1/Ph-like subtype and account for up to 20% of all
CRLF2 fusions [
8,
9].
In recent studies, two new subtypes (PAX5-driven) with specific
PAX5 alterations have been defined in BCP-ALL. The first subtype is PAX5alt with rearrangements, intragenic amplifications, or sequence mutations of
PAX5 [
8,
9]. The second subtype is the hotspot mutation PAX5 p.Pro80Arg (P80R) and biallelic
PAX5 alterations [
8,
9,
65,
86]. PAX5alt and PAX5 P80R occur in 7%–10% and 3%–4% of childhood and 4%–8% and 1% of adult patients with BCP-ALL. Moreover, both subtypes are associated with intermediate outcomes. Additionally, compared with adults with PAX5alt, adults with PAX5 P80R had more superior outcomes [
8,
9].
Diverse
PAX5 alterations were found in approximately 75% of patients in the PAX5alt subtype. The alterations include
PAX5 rearrangements (
PAX5-
ETV6,
PAX5-
NOL4L,
PAX5-
AUTS2,
PAX5-
CBFA2T3,
PAX5-
DACH1,
PAX5-
FOXP1, and
PAX5-
ZNF521), sequence mutations (PAX5 p.Pro32Ser (P32S), p.Pro34Leu (P34L), p.Arg38Cys (R38C)/p.Arg38His (R38H), and p.Arg140Leu (R140L)/p.Arg140Gln (R140Q)), and CNVs (mainly one copy loss and focal intragenic amplification of
PAX5). The ratio of CNVs that is positive for PAX5alt showed lesser frequency than that of other subtypes, but focal intragenic amplification (8/10) of
PAX5 (PAX5amp) was observed [
9]. Apart from
PAX5 alterations, other common genetic alterations in the PAX5alt subtype are the signaling molecules (e.g.,
NRAS,
KRAS, and
FLT3), cell-cycle regulator
CDKN2A deletions, B-cell development (
IKZF1 and
VPREB1 deletions), transcriptional factors (
ZFP36L2 and
ETV6), and epigenetics modifier (
KDM6A). The gene expression data showed that cytokine receptor genes (e.g.,
PDGFRB and
FLT3) are enriched in PAX5alt, which is in line with the activation mutations in signaling pathways [
8,
9].
Point mutations of PAX5 P80R in the DNA binding can affect the ability of
PAX5 to bind DNAs and regulate expressions. These mutations represent the first molecular subtype defined on the basis of homogeneous hotspot mutations and gene expression profiles [
8,
9,
65,
86,
108,
109]. Most patients with PAX5 P80R presented distinct gene expression profiles and uniform genetic alterations. PAX5 P80R patients can promote the biallelic alteration of
PAX5in vivo, and this alteration frequently coexists with the hemizygous loss of
PAX5. This scenario results in a higher mutation allele frequency compared with that of other
PAX5 point mutations [
9]. Hemizygous PAX5 P80R without the deletion of the wild-type
PAX5 allele may present gene expression features in other subtypes, such as the Ph-like and PAX5alt. Activating mutations in signaling (Ras and JAK/STAT pathways), including
PTPN11 and
IL7R, and inactivating mutations in epigenetic factor
SETD2 are the most common genetic alterations coexisting with PAX5 P80R [
8,
9,
65,
86].
PAX5-driven subtypes showed high heterogeneity in genetics, such as diverse patterning genes involved in
PAX5 fusions, and sequence mutations scattered on the coding region of
PAX5. This characteristic may further require diverse therapy agents, including a combination of chemotherapy and multi-inhibitors, and possibly immunotherapy. Additionally, different prognoses between adult and childhood PAX5alt indicates the presence of additional age-dependent factors that may affect the vitality, aggressiveness, and drug responsiveness of leukemic cells in this BCP-ALL subtype [
9].
3.4 NUTM1 gene fusion
The NUT midline carcinoma family member 1 (
NUTM1), also called nuclear protein in the testis, is located within chromosome 15q14. Previously,
NUTM1 fusions were mainly reported as translocations t(15;9)(q14;q34) and t(15;19)(q14;p13) that could result in
BRD3-NUTM1 and
BRD4-
NUTM1 [
110–
113]. In recent years, rearrangements of
NUTM1 involving
ACIN1 (14q11),
CUX1 (7q22),
AFF1 (4q21),
BRD9 (5p15),
ZNF618 (9q32),
IKZF (7p12), and
SLC12A6 (15q14) have been identified as a specific subtype (1%–2%) in BCP-ALL. This subtype is preliminarily considered a subtype of BCP-ALL associated with a favorable prognosis [
8,
9,
16–
18,
114,
115]. The
HOXA gene family, particularly
HOXA9, is upregulated in the
NUTM1-
r ALL, which has not been described in NUT midline carcinoma [
8].
KMT2A fusions can regulate leukemogenic gene expressions, particularly the
HOXA gene family, by modulating the acetylation of H3K27 and disturbing the telomeric silencing 1-like histone 3 lysine 79 (H3K79) methyltransferase DOT1L [
51,
116].
HOXA9 can cooperate with the JAK/STAT signaling pathway and drive leukemia development [
117]. The molecular mechanism of
NUTM1-
r ALL is still largely unknown and thus needs further study. Both
NUTM1 and
KMT2A fusions may drive leukemogenesis by disturbing epigenetic status and upregulating the
HOXA gene family.
3.5 Other gene fusions and point mutations
3.5.1 Rearrangements of BCL2/MYC and/or BCL6
Translocations t(14;18)(q32;q21), t(8;14)(q24;q32), and t(3;14)(q27;q32) that result in
BCL2-
IGH,
MYC-
IGH, and/or
BCL6-
IGH fusions occur in approximately 2% of adults (mostly>30 years old) with BCP-ALL and are less common in children. These translocations are associated with consistently poor responses in early treatment [
9]. Low levels of circulating t(14;18)-positive cells can be found in approximately 50%–70% of healthy individuals but never develop into a disease [
118–
121]. Chromosomal alterations in
BCL2,
MYC, and/or
BCL6 have been frequently reported in chronic lymphocytic leukemia and double- or triple-hit lymphoma, which are rare in ALL with a B-cell-precursor immunophenotype [
16,
121–
133]. The promoter regions of
BCL2,
MYC, and
BCL6 are usually affected by
IGH translocations, causing an overexpression of the rearranged allele compared with the germline allele. This characteristic can dramatically activate the proliferation of leukemogenic blast cells [
123]. At present, patients with the abovementioned alterations cannot be well cured because of the development of chemotherapy resistance [
9,
121]. This limitation poses a great challenge to patient treatment. The inhibition of cyclin-dependent kinase 7 (CDK7) has been reported as an optional target for reducing the resistance of BCL-2 in B-cell lymphoma models [
133]. The developed therapeutic agents in B-cell lymphoma may also be effective in BCP-ALL. Additional functional assays and clinical tests are urgently needed to improve the survival rate in the
BCL2/
MYC and/or
BCL6-
r BCP-ALL.
3.5.2 ZEB2 p.His1038Arg (H1038R) and IGH-CEBPE gene fusion
Zinc finger E-box binding homeobox 2 (
ZEB2), a member of the Zfh1 family of 2-handed zinc finger/homeodomain proteins, is a nuclear protein that can bind DNA and repress its transcriptional activity; this protein interacts with activated SMAD, a DNA binding protein, and recognizes an 8-bp palindromic sequence (GTCTAGAC) called the Smad binding element [
134]. CCAAT enhancer binding protein epsilon (CEPBE) is a bZIP transcription factor that binds to certain DNA regulatory regions by means of homodimer formation. In a recent study, a hotspot mutation of ZEB2 p.His1038Arg (H1038R) and translocation t(14;14) (q11;q32), which results in
IGH-
CEBPE with a truncation of the 3′ UTR, is defined as a rare subtype (<1%) sharing similar gene expression features. The rs2239630 G>A at the promoter of
CEBPE is associated with the
IGH-
CEBPE translocated ALL with increased
CEBPE expression [
135]. More than 50% of patients in this subtype also have
NRAS sequence mutations. The leukemic oncogene
LMO1 is significantly upregulated in this subtype and is an important component of a transcriptional complex that includes TAL1, TCF12/HEB, TCF3/E2A, MYB, RUNX1, GATA3, and LDB1. This transcription complex can form a positive interconnected autoregulatory circuit that impacts the transformation in approximately 60% of patients with T-cell ALL (T-ALL) [
77,
136]. In addition,
SMAD1 and
BMP2 are significantly downregulated in this subtype; these genes can regulate the signals of the bone morphogenetic proteins and are involved in a range of biological activities, including morphogenesis, cell growth, apoptosis, development, and immune responses [
137–
140]. Preliminarily,
ZEB2 mutation is associated with poor event-free survival and high relapse in patients with BCP-ALL [
141]. The clinical implications of ZEB2 p.His1038Arg (H1038R) and
IGH-
CEBPE gene fusion subtype are still largely unclear. Much larger sample sizes and functional tests are needed for this BCP-ALL subtype.
3.5.3 IKZF1 p.Asn159Tyr (N159Y)
IKZF1, also known as IKAROS, is a critical transcription factor related to the differentiation and maturation of the B-cell precursor. Somatic alterations of
IKZF1 are a hallmark of high-risk BCP-ALL with poor response to therapy [
72,
109,
142]. Point mutations of IKZF1 p.Asn159Tyr (N159Y) were recently recognized as a rare subtype (<1%) in BCP-ALL. In light of the limitation of sample size, the outcomes of IKZF1 N159Y are still undermined. IKZF1 N159Y, which is located in the DNA binding domain, may impact its capability for DNA binding and gene transcription regulation. Sequence mutations of
KRAS are recurrent in patients with IKZF1 N159Y but lack extra copy number alterations. The gene expression data showed that the transcriptional coactivator
YAP1 is significantly upregulated, which can drive
KRAS-induced transformation through rescued cell viability in
KRAS-dependent cells [
143]. The chromatin remodeling
SALL1 and the signaling factor
ARHGEF28 are also significantly upregulated in IKZF1 N159Y-positive patients. Meanwhile, the B-cell receptor signaling and JAK-STAT signaling pathways (e.g.,
FLT3,
FLT4, and
STAT5) are down-regulated in patients with IKZF1 N159Y. These two pathways are commonly activated in other BCP-ALLs [
8,
9,
144–
146]. At present, the public data show that IKZF1 N159Y-positive cases are still below 10. One case of IKZF1 N159T was reported in the chronic myelomonocytic leukemia cohort [
147]. Surprisingly,
de novo germline mutations, including IKZF1 N159S (
n = 6) and IKZF1 N159T (
n = 1), could exist at the same IKZF1 amino acid N159, which then would cause T-, B-, and myeloid cell-combined immunodeficiency, and a patient would develop T-ALL [
148].
Pneumocystis jirovecii pneumonia is positive in all of patients with N159S or N159T mutations. A patient died at 2 years old without leukemia phenotype although the child received hematopoietic stem cell transplants [
148]. Multi-center collaboration may accelerate the collection of patient data in this subtype for the further evaluation of its clinical significance (Table 1; Fig. 1A and 1B).
4 Diagnosis and molecular classification of BCP-ALL based on RNA-seq
Genetic variations with clinical significance in BCP-ALL are structurally heterogeneous [
18]. Multiple high-throughput sequencing approaches are highly recommended, as they can be utilized to accurately recognize the prognostic factors (e.g., fusion genes, gene expression-dependent subtypes, small sequence variants, and genomic duplications and deletions) and determine the strategic therapy [
149]. RNA-seq is a single and comprehensive platform for BCP-ALL diagnosis and genomic classification in the laboratory and clinical settings [
9,
43,
150–
152]. For example, most of the known fusion genes (e.g.,
BCR-
ABL1,
ETV6-
RUNX1, and
TCF3-
PBX1) and new fusion genes (e.g.,
TCF3/
4-
HLF,
NUTM1,
DUX4,
ZNF384/
ZNF362, and
MEF2D fusions) in BCP-ALL can be detected by RNA-seq [
149,
153]. These newly identified recurrent
DUX4,
ZNF384,
MEF2D, and
NUTM1 fusions have distinct clinical features [
8,
9,
15,
16,
18,
22,
64]. Notably, RNA-seq is also a reliable technology for simultaneously identifying the positive-fusion gene and the gene expression-dependent subtypes, including Ph-like,
ETV6-
RUNX1-like,
ZNF384-like, and
KMT2A-like. Small sequence variants and genomic deletions (e.g.,
IKZF1) are also detectable by RNA-seq in BCP-ALL [
8,
9,
77,
149,
151–
154]. For instance, by re-analyzing the RNA-seq data from different BCP-ALL cohorts, the molecular subtypes characterized by hotspot mutations (e.g., PAX5 P80R and ZEB2 H1038R) could be identified [
8]. Exon-level genomic deletions can cause differential transcripts expression (e.g.,
IKZF1 exons 4–7, 2–7, 2–8, and 4–8) and thus can be used to predict genomic deletion events [
153]. RNA-seq was also applied in AML diagnosis to accurately detect small sequence variants,
FLT3-internal tandem duplication (ITD), and
KMT2A-partial tandem duplication (PTD) events [
155–
157].
However, RNA-seq is more susceptible to bias factors originating from the samples, technology platform, and bioinformatics methodology (e.g., batch effect) compared with the DNA-based methods. Additional systematic benchmarks and further refinement of the methodology as a means of reducing the bias are needed to improve the stability and reducibility of the RNA-based methods [
149,
158,
159].
5 New therapeutic targets and agents in BCP-ALL
Intensive chemotherapy and allogeneic hematopoietic cell transplantation were the core options of BCP-ALL treatment in the past [
27]. Treatment toxicity with relapsed events induced by chemotherapy drugs has been one of the most critical concerns awaiting further resolution in BCP-ALL [
13,
27]. The advent of emerging inhibitors/antagonists and immunotherapeutic has launched a new era of target therapy in several molecular subtypes or unselected BCP-ALL patients (Table 2). This ongoing transformation may continuously reduce the use of chemotherapy drugs and consequently achieve less treatment-induced resistance events [
160]. Molecular target therapy and cellular immunotherapy in BCP-ALL are mainly dependent on the specific genetics and gene expression markers of the patients’ leukemic cells in various molecular subtypes (e.g.,
BCR-
ABL1, Ph-like, and
KMT2A fusions) or the shared or unselected cell surface marker (e.g., CD19 and CD22) (Fig. 2). Several molecular subtypes in BCP-ALL, including R/R subtypes, have benefited from the new therapeutic targets and agents, although a large number of biomarkers are still rarely used as therapy targets.
BCR-
ABL1 and Ph-like (e.g.,
BCR-
ABL1-negative with fusions or mutations involved in
ABL1/
ABL2,
PDGFRA/
B,
EPOR, and
CSF1R) have benefited from TKIs, including imatinib and/or dasatinib [
91]. The combination of dasatinib and c-JUN N-terminal kinase (JNK) inhibitor, i.e., JNK-IN-8, can significantly improve the survival of the
BCR-
ABL1-positive mice model [
161]. The cyclin-dependent kinases 8 (CDK8) inhibitor, YKL-06-101, combined with the mTOR inhibitor can induce cell death of human
BCR-
ABL1 leukemic cells [
162]. Besides, both CDK4/6 inhibitors and Bcl-2 inhibitor are two types of molecular inhibitors that have been tested in R/R BCP-ALL [
13,
163], such as CDK4/6 inhibitors palbociclib (NCT02310243, NCT03472573, NCT03515200, NCT03132454, and NCT03792256) and Bcl-2 inhibitor venetoclax (NCT03826992, NCT03319901, NCT03181126, NCT04029688, NCT03808610, and NCT03504644).
KMT2A fusions,
HLF fusions, and other R/R BCP-ALL may benefit from the inhibition of the cell cycle and apoptosis pathways. De-regulated DNA methylation or histone deacetylation has been found in several BCP-ALL subtypes (e.g.,
KMT2A and
MEF2D fusions). DOT1L inhibitors (e.g., pinometostat/EPZ-5676) have been tested in phase I trials (NCT01684150 and NCT02141828) for
KMT2A-
r leukemia [
164], and the phase 1b/II trials (NCT03724084 and NCT03701295) are currently recruiting patients. HDAC inhibitors (e.g., Vorinostat and Panobinostat) have also been proposed to be benefiting several BCP-ALL subtypes (NCT02553460), including
MEF2D fusions and R/R BCP-ALL [
16,
18,
64,
165]. A newly proposed bioavailable Menin (
MEN1)-
KMT2A interaction inhibitor, VTP50469, showed that it can improve survival in patient-derived tumor xenograft mouse models of
KMT2A-
r BCP-ALL by suppressing a subset of
KMT2A fusion target genes [
166]. Other activated critical cellular pathways regulating cell proliferation and apoptosis (i.e., pre-B/B-cell receptor, RAS, JAK-STAT, and mTOR/PI3K) [
13] are also promising targeted pathways. Numerous clinical trials have been registered. The test combining multiple molecular compounds or agents are still in progress. Nonetheless, additional work is required to verify whether the intensive dose of chemotherapy drugs can be reduced reasonably.
Apart from the inhibitors/antagonists, immunotherapeutic agents, such as CAR-T and monoclonal antibodies, are another promising strategy for targeting specific cell surface markers overexpressed in leukemic cells. Immunotherapeutic agents can be used in the therapy of most children and adults with R/R BCP-ALL. The cell membrane antigens CD19, CD20, and CD22 are the three most promising targets of immunotherapeutic agents for BCP-ALL [
167–
174]. Other bispecific agents (e.g., Combobox for CD19/CD22) are becoming the next-generation CAR-T treatment options. As one of the hottest fields in BCP-ALL, a series of clinical trials of CAR-T for R/R BCP-ALL is ongoing (e.g., NCT03330691, NCT03937544, NCT00450944, NCT04012879, and NCT04094766). The immunotherapeutic agents Inotuzumab and Blinatumomab were approved by the United States Food and Drug Authority for the therapy of adults with R/R BCP-ALL, which can help to improve in the future the overall survival of both children and adults with BCP-ALL. In addition, the combination of the TKIs imatinib or dasatinib with multiagent chemotherapy is currently used in multiple clinical trials of R/R BCP-ALL. This treatment has markedly improved the outcome in BCP-ALL subset, as the five-year overall survival of patients has increased to 75% from less than 50% [
12,
175]. Controlling early death, reducing therapy-induced resistance mutations, and comprehensive clinical management of adult patients are the potential key issues of medical precision in BCP-ALL treatment (Table 2) [
5,
8,
9,
12,
13,
25,
40,
60,
61,
160,
167,
169,
176–
182].
6 Conclusions
Through decades of collaboration, more than 95% of patients with BCP-ALL have been classified and labeled using detectable genetic alterations, including distinct translocations of chromosomes, aneuploidy, DNA copy number alterations, sequence mutations, and gene expression patterns [
8–
12,
183]. Refined risk stratification and the approval of new drugs and agents, including target inhibitors, monoclonal antibodies, and immunomodulators, and CAR-T, have resulted in an excellent five-year event-free survival and a five-year overall survival in children with BCP-ALL. However, the development of resistance and early death during treatments continue to pose daunting challenges for some subtypes of BCP-ALL, including hypodiploid (<44 chromosomes),
HLF-arranged,
BCL2/
MYC-
r,
BCR-
ABL1/Ph-like,
KMT2A-
r, and
MEF2D-
r BCP-ALL. Additionally, single-cell-based traces of cancer cell populations, chromatin accessibility, epigenetic alterations, genome-wide germline mutations, functional non-coding and synonymous mutations, and other abnormalities at the non-genomic levels, such as protein and metabolic levels, are still poorly understood. Information on these aspects can help in drug discovery and improvement in the prognoses of leukemia patients, including BCP-ALLs [
103–
105,
183–
188]. Through the comprehensive identification of prognostic biomarkers and the development of new techniques in diagnosis and target treatment, we can further improve the survival time and life quality of patients with BCP-ALL.
The Author(s) 2020. This article is published with open access at link.springer.com and journal.hep.com.cn