You're using an outdated browser. Please upgrade to a modern browser for the best experience.
Genomic Features of Ph-Like ALL: Comparison
Please note this is a comparison between Version 2 by Nora Tang and Version 1 by Ilaria Iacobucci.

A wide spectrum of genetic alterations (>60), including translocations, cryptic rearrangements, sequence mutations and copy number changes have been described in Ph-like ALL, with slight differences in prevalence across age. These alterations drive constitutively active kinase or cytokine receptor signaling, many of which have been shown to be druggable with a variety of kinase inhibitors. The most commonly mutated pathways are the ABL and JAK-STAT pathways with multiple rearrangements and lesions that converge on downstream ABL/JAK-STAT signaling. Founder alterations may be grouped into three types: (i) JAK/STAT alterations including mutations activating cytokine receptors (e.g., CRLF2 and IL7R); gene fusions hijacking cytokine receptor expression (e.g.,

IGH-CRLF2

and

P2RY8–CRLF2

); gene fusions and/or mutations activating kinases (e.g.,

JAK1

,

JAK2

,

JAK3

,

TYK2

); and rearrangements hijacking and truncating cytokine receptor expression (e.g., cryptic

EPOR

rearrangements); (ii) fusions involving ABL-class genes (

ABL1

,

ABL2

,

CSF1R

,

LYN

,

PDGFRA

,

PDGFRB

); (iii) less common fusions (

FLT3

,

FGFR1

,

NTRK3

,

PTK2B

) whose number is growing with increasing sequencing studies of different cohorts.

  • acute lymphoblastic leukemia (ALL)
  • Ph-like ALL

1. JAK/STAT Alterations

Approximately 50% of patients with Ph-like ALL harbor rearrangements of the cytokine receptor- like factor 2 (

CRLF2

) gene, located on the pseudoautosomal region 1 (

PAR1) of chromosomes Xp22 and Yp11 [4,12,15,24]. In normal conditions CRLF2 dimerizes with the α- subunit of interleukin- 7 receptor (IL7RA) to form a heterodimeric thymic stromal lymphopoietin receptor (TSLPR) which actives downstream JAK2/STAT5 and thePI3K/AKT/mTOR pathways [26,27,28] and is implicated in early B-cell development [29]. 

) of chromosomes Xp22 and Yp11 [1][2][3][4]. In normal conditions CRLF2 dimerizes with the α- subunit of interleukin- 7 receptor (IL7RA) to form a heterodimeric thymic stromal lymphopoietin receptor (TSLPR) which actives downstream JAK2/STAT5 and thePI3K/AKT/mTOR pathways [5][6][7] and is implicated in early B-cell development [8]

CRLF2

 deregulation results from three main mechanisms: (1) a cryptic rearrangement that juxtaposes 

CRLF2

 to the immunoglobulin heavy chain locus (

IGH

); (2) a focal deletion in the pseudoautosomal region of the sex chromosomes resulting in P2Y receptor family member 8 (

P2RY8

)-

CRLF2

 fusion that positions 

CRLF2

 under the control of the

 P2RY8

 promoter; (3) and less frequently by an activating

CRLF2 point mutation, F232C [13,17,22,23,25,30,31,32,33]. Rearrangements of 

point mutation, F232C [9][10][11][12][13][14][15][16][17]. Rearrangements of 

CRLF2 account for 24% of pediatric patients with NCI SR Ph-like ALL [9], 55% of children with HR disease [10] and 50% to 60% of adolescent and adult patients with Ph-like ALL cases [4,11,12,13,17,18]. 

account for 24% of pediatric patients with NCI SR Ph-like ALL [18], 55% of children with HR disease [19] and 50% to 60% of adolescent and adult patients with Ph-like ALL cases [1][20][2][9][10][21]

P2RY8-CRLF2 fusions occur more commonly in younger children and in patients with Down syndrome (DS) ALL [22,25], while 

 fusions occur more commonly in younger children and in patients with Down syndrome (DS) ALL [11][13], while 

IGH-CRLF2 fusions are detected more frequently in older patients and patients of Hispanic ethnicity [34]. In a genome-wide association study of 

 fusions are detected more frequently in older patients and patients of Hispanic ethnicity [22]. In a genome-wide association study of 

CRLF2

-rearranged ALL, the inherited 

GATA3

 variant rs3824662 was associated with 

CRLF2

 rearrangement, 

JAK

 mutation, 

IKZF1

 deletion, variation in 

GATA3 expression and increased risk of relapse [35]. This variant is markedly more common in patients of Hispanic ethnicity (~40%) or Native American (~50%) genetic ancestry, while is it detected in only 14% of Europeans [35,36]. The point mutation changing phenylalanine 232 to cysteine in CRLF2 has been identified in 9% of DS-ALL patients [25] and 21% of adult B-ALL patients [23]. In in vitro assays, the expression of CRLF2 F232C in the absence of co-expression of mutant JAK2 promotes JAK2 signaling activation and cell transformation [23,25,37]. 

 expression and increased risk of relapse [23]. This variant is markedly more common in patients of Hispanic ethnicity (~40%) or Native American (~50%) genetic ancestry, while is it detected in only 14% of Europeans [23][24]. The point mutation changing phenylalanine 232 to cysteine in CRLF2 has been identified in 9% of DS-ALL patients [13] and 21% of adult B-ALL patients [12]. In in vitro assays, the expression of CRLF2 F232C in the absence of co-expression of mutant JAK2 promotes JAK2 signaling activation and cell transformation [12][13][25]

CRLF2

 rearrangement and overexpression is associated with worse outcome compared to cases with lack of 

CRLF2 alterations [15,34,38,39]. However, the poor prognostic impact of 

alterations [3][22][26][27]. However, the poor prognostic impact of 

CRLF2 

overexpression is overcome by 

BCR-ABL1

–like signature and 

IKZF1 deletion in the Dutch Childhood Oncology Group trials and German Cooperative ALL trials [37]. In about half of 

 deletion in the Dutch Childhood Oncology Group trials and German Cooperative ALL trials [25]. In about half of 

CRLF2

-rearranged pediatric Ph-like ALL cases, concomitant 

JAK1

 and 

JAK2 

(most commonly in the pseudokinase domain at R683) mutations occur. In adults, the frequency of

JAK

mutations in patients with 

CRLF2

 rearrangement is lower, with a ratio of 1:4 with 

JAK wild type [4,12,15,23]. In 

 wild type [1][2][3][12]. In 

JAK1

 the most common mutation is represented by V658F which is the homolog of JAK2 V617F, hotspot in myeloproliferative neoplasms. Other alterations leading to JAK/STAT activation target 

IL7RA

SH2B3

IL2RB

, and 

TYK2 genes. Collectively these alterations are approximately two-fold higher in children (14%) compared to adolescents (5.0%), and adults (7.3%) [4,12,15]. 

 genes. Collectively these alterations are approximately two-fold higher in children (14%) compared to adolescents (5.0%), and adults (7.3%) [1][2][3]

IL7RA mutations occur in exon 6 and are mainly in-frame insertion/deletions in the juxtamembrane-transmembrane domain or, rarely, a serine-to-cysteine substitution at amino acid 185 in the extracellular domain [38]. Independent of 

 mutations occur in exon 6 and are mainly in-frame insertion/deletions in the juxtamembrane-transmembrane domain or, rarely, a serine-to-cysteine substitution at amino acid 185 in the extracellular domain [26]. Independent of 

CRLF2

 rearrangements, JAK-STAT signaling activation can result from 

JAK2

 (~7%) or erythropoietin receptor (

EPOR,

 5%) -rearrangements.

Over 20 different 

JAK2

 gene fusion partners have been reported (most commonly 

EBF1

ETV6

PAX5

, and 

BCR

), making 

JAK2

 the most promiscuous gene in Ph-like ALL. All fusions preserve the JAK2 kinase domain and result in STAT5 activation and growth factor independence, making cells expressing these fusions amenable to JAK2 inhibitors.

Common 

EPOR

 rearrangements involve juxtaposition or less frequently translocation of the 

EPOR

 gene in proximity of a strong enhancer, such as that of the immunoglobulin heavy (

IGH

) or kappa (

IGK

) loci, that drives its expression. Less frequent rearrangements involve insertion of 

EPOR 

into the upstream region of 

LAIR1

 or the 

THADA loci [4,39]. All these rearrangements clip off the C-terminal cytoplasmic tail, thus preserving the proximal tyrosine requested for activation and removing almost all tyrosine sites required for shutting off the receptor signaling and down-regulate and internalize the receptor. This leads to transformation in in vivo models and sensitivity to a variety of different JAK2 inhibitors in in vitro and in vivo models. While 

 loci [1][27]. All these rearrangements clip off the C-terminal cytoplasmic tail, thus preserving the proximal tyrosine requested for activation and removing almost all tyrosine sites required for shutting off the receptor signaling and down-regulate and internalize the receptor. This leads to transformation in in vivo models and sensitivity to a variety of different JAK2 inhibitors in in vitro and in vivo models. While 

IGH-EPOR fusion due the translocation t(14;19)(q32;p13) can be detected by fluorescence in situ hybridization (FISH) [40], the other 

 fusion due the translocation t(14;19)(q32;p13) can be detected by fluorescence in situ hybridization (FISH) [28], the other 

EPOR

 rearrangements are cryptic and challenging to detect without using next-generation sequencing (NGS) technologies. The prevalence of 

EPOR rearrangements has a peak in young adults (9%) compared to children and adolescents (5% and 3%, respectively). They are rarely detected in adults (1%) [13,39]. 

 rearrangements has a peak in young adults (9%) compared to children and adolescents (5% and 3%, respectively). They are rarely detected in adults (1%) [9][27]

JAK2

 and 

EPOR rearrangements are associated with the poorest outcome compared with the other molecular Ph-like subtypes [12,13].

 rearrangements are associated with the poorest outcome compared with the other molecular Ph-like subtypes [2][9].

2. Fusions Involving ABL-Class Genes

The ABL-class gene fusions include rearrangements of ABL proto-oncogene 1 (

ABL1;

 e.g., to 

RCSD1, NUP214, LSM14A, ETV6, RANBP2, CENPC, FOXP1, SFPQ, SNX1, SNX2, SPTNA1, ZMIZ1, NUP153

), ABL proto-oncogene 2 (

ABL2;

 e.g., to 

RCSD2, PAG1, ZC3HAV1

), colony-stimulating factor 1 receptor (

CSF1R;

 e.g., to 

SSBP2

MEF2D, TBL1XR1

), platelet-derived growth factor receptor beta (

PDGFRB;

 e.g., to 

EBF1, ETV6, ATF7IP, SNX29, SSBP2, TNIP1, ZEB2, ZMYND8, NUMA1

) and 

LYN (GATAD2A-LYN, NCOR1-LYN [41]), with multiple partner genes, with 

 [29]), with multiple partner genes, with 

ABL1

 and 

PDGFRB being the most common. The prevalence of these rearrangements is 17% in children, 9% in adolescents, 10% young adults and 9% older adults [4,12,14,15]. Patients with ABL-class fusions respond poorly to chemotherapy regimens, and the 

 being the most common. The prevalence of these rearrangements is 17% in children, 9% in adolescents, 10% young adults and 9% older adults [1][2][30][3]. Patients with ABL-class fusions respond poorly to chemotherapy regimens, and the 

EBF1-PDGFRB fusion in particular is associated with induction failure [42,43,44]. All fusions preserve the tyrosine kinase of the ABL-class gene and promote constitutive kinase signaling that confers the ability to survive and grow independently of cytokine in vitro [45]. Imatinib, the dual ABL1/SRC inhibitor dasatinib or other TKIs inhibit the downstream signaling induced by each of these chimeric fusion proteins [4,46,47] and are currently used in clinical trials. The best and first example is provided by the inhibition of 

fusion in particular is associated with induction failure [31][32][33]. All fusions preserve the tyrosine kinase of the ABL-class gene and promote constitutive kinase signaling that confers the ability to survive and grow independently of cytokine in vitro [34]. Imatinib, the dual ABL1/SRC inhibitor dasatinib or other TKIs inhibit the downstream signaling induced by each of these chimeric fusion proteins [1][35][36] and are currently used in clinical trials. The best and first example is provided by the inhibition of 

EBF1-PDGFRB fusion by imatinib [44,46,47,48,49]. The emergence of kinase domain point mutations may represent a potential mechanism of relapse in 

 fusion by imatinib [33][35][36][37][38]. The emergence of kinase domain point mutations may represent a potential mechanism of relapse in 

EBF1-PDGFRB

 or other kinase driven-subtypes in Ph-like ALL. Recently, the T681I gatekeeper mutation has been demonstrated to be the most common resistant mutation in 

EBF1-PDGFRB Ph-like ALL to both imatinib and dasatinib in in vitro screens and it was associated with a trend towards increased risk of relapse in patients harboring T681I subclones at diagnosis compared to T681I-negative patients [50].

 Ph-like ALL to both imatinib and dasatinib in in vitro screens and it was associated with a trend towards increased risk of relapse in patients harboring T681I subclones at diagnosis compared to T681I-negative patients [39].

3. Other Kinase Fusions and Genetic Aberrations

Around 5% of Ph-like ALL cases harbor gene fusions or mutations involving 

NTRK3, BLNK, DGKH, PTK2B, FLT3, FGFR1, TYK2

 and 

SH2B3

. Among those, one percent of cases harbor the fusion between 

ETV6

 and 

NTRK3 encoding a member of the tropomyosin receptor tyrosine kinase (TRK) family [51]. This fusion is not unique of Ph-like ALL since it has been identified in a range of hematological malignancies, such as acute myeloid leukemia [52], infantile sarcoma [53,54] and solid tumors [55,56,57,58]. In preclinical models, ETV6-NTRK3 has been shown to promote the development of an aggressive B-ALL and to be exquisitely sensitive to the TRK inhibitors larotrectinib (LOXO-101) or PLX7486 (Plexxikon) in both patient derived xenograft models and in B-ALL patients with

 encoding a member of the tropomyosin receptor tyrosine kinase (TRK) family [40]. This fusion is not unique of Ph-like ALL since it has been identified in a range of hematological malignancies, such as acute myeloid leukemia [41], infantile sarcoma [42][43] and solid tumors [44][45][46][47]. In preclinical models, ETV6-NTRK3 has been shown to promote the development of an aggressive B-ALL and to be exquisitely sensitive to the TRK inhibitors larotrectinib (LOXO-101) or PLX7486 (Plexxikon) in both patient derived xenograft models and in B-ALL patients with

ETV6-NTRK3 [55,59,60]. Recently, a clinical response to larotrectinib has been reported in an adult Ph-like ALL with cryptic

[44][48][49]. Recently, a clinical response to larotrectinib has been reported in an adult Ph-like ALL with cryptic

ETV6-NTRK3

rearrangement and NRASGly12Asp mutation. The patient failed to respond to multiagent chemotherapy and relapse after investigational CD19-directed chimeric antigen receptor T-cell therapy with a clone positive for

ETV6-NTRK3 but not anymore for the NRASGly12Asp mutation. The relapsed leukemia progressed with further chemo- and immunotherapy but showed substantial leukemic cytoreduction using the TRK inhibitor larotrectinib [61]. Fusions of the B Cell Linker Protein (

but not anymore for the NRASGly12Asp mutation. The relapsed leukemia progressed with further chemo- and immunotherapy but showed substantial leukemic cytoreduction using the TRK inhibitor larotrectinib [50]. Fusions of the B Cell Linker Protein (

BLNK

) or 

SLP65

 gene to 

DNTT

 (also known as 

TDT) have been also described [13,62].

) have been also described [9][51].

BLNK encodes a cytoplasmic adapter protein important for B-cell development and function by activating BCR downstream signaling [63], while 

encodes a cytoplasmic adapter protein important for B-cell development and function by activating BCR downstream signaling [52], while 

DNTT encodes a encodes a template-independent DNA polymerase that catalyzes the addition of deoxynucleotides and that is highly expressed in normal and malignant pre-B and pre-T lymphocytes during early differentiation [64].

 encodes a encodes a template-independent DNA polymerase that catalyzes the addition of deoxynucleotides and that is highly expressed in normal and malignant pre-B and pre-T lymphocytes during early differentiation [53].

In addition to gene fusions, RAS pathway activating mutations or deletions (

KRAS, NRAS, NF1, PTPN11

) and copy number aberrations in genes involved in B-cell development (

IKZF1, PAX5, EBF1,

 and 

ETV6

) and cell cycle regulators (

CDKN2A/B

TP53

BTG1

, and 

RB1

) are recurrent. Deletions in 

IKZF1 occur in around 27% of pediatric cases and in approximately 70% of high-risk pediatric patients with ALL [4]. As in 

 occur in around 27% of pediatric cases and in approximately 70% of high-risk pediatric patients with ALL [1]. As in 

BCR-ABL positive ALL [6,59], 

 positive ALL [54][48]

IKZF1 deletions confer a poor prognostic outcome [20]. 

 deletions confer a poor prognostic outcome [55]

IKZF1 

deletions are significantly more common in patients carrying kinase or cytokine receptor rearrangement (

IGH-CRLF2) than a sequence mutation [4,13], especially in Hispanic/Latino (H/L) children with B-ALL (29% in H/L compared to 15% of non-Hispanic Whites) where both 

) than a sequence mutation [1][9], especially in Hispanic/Latino (H/L) children with B-ALL (29% in H/L compared to 15% of non-Hispanic Whites) where both 

IGH-CRLF2

 translocation and 

IKZF1 deletion provide a strong biological rationale for the higher death-rate H/L experience due to B-ALL [60].

 deletion provide a strong biological rationale for the higher death-rate H/L experience due to B-ALL [49].

 

References

  1. Roberts, K.G.; Li, Y.; Payne-Turner, D.; Harvey, R.C.; Yang, Y.-L.; Pei, D.; McCastlain, K.; Ding, L.; Lu, C.; Song, G.; et al. Targetable Kinase-Activating Lesions in Ph-like Acute Lymphoblastic Leukemia. N. Engl. J. Med. 2014, 371, 1005–1015.
  2. Jain, N.; Roberts, K.G.; Jabbour, E.; Patel, K.; Eterovic, A.K.; Chen, K.; Zweidler-McKay, P.; Lu, X.; Fawcett, G.; Wang, S.A.; et al. Ph-like acute lymphoblastic leukemia: A high-risk subtype in adults. Blood 2017, 129, 572–581.
  3. Loh, M.L.; Zhang, J.; Harvey, R.C.; Roberts, K.; Payne-Turner, D.; Kang, H.; Wu, G.; Chen, X.; Becksfort, J.; Edmonson, M.; et al. Tyrosine kinome sequencing of pediatric acute lymphoblastic leukemia: A report from the Children’s Oncology Group TARGET Project. Blood 2013, 121, 485–488.
  4. Russell, L.J.; Jones, L.; Enshaei, A.; Tonin, S.; Ryan, S.L.; Eswaran, J.; Nakjang, S.; Papaemmanuil, E.; Tubio, J.M.C.; Fielding, A.K.; et al. Characterisation of the genomic landscape ofCRLF2-rearranged acute lymphoblastic leukemia. Genes Chromosom. Cancer 2017, 56, 363–372.
  5. Isaksen, D.E.; Baumann, H.; Trobridge, P.A.; Farr, A.G.; Levin, S.D.; Ziegler, S.F. Requirement for stat5 in thymic stromal lymphopoietin-mediated signal transduction. J. Immunol. 1999, 163, 5971–5977.
  6. Wohlmann, A.; Sebastian, K.; Borowski, A.; Krause, S.; Friedrich, K. Signal transduction by the atopy-associated human thymic stromal lymphopoietin (TSLP) receptor depends on Janus kinase function. Biol. Chem. 2010, 391, 181–186.
  7. Tasian, S.K.; Doral, M.Y.; Borowitz, M.J.; Wood, B.L.; Chen, I.-M.; Harvey, R.C.; Gastier-Foster, J.M.; Willman, C.L.; Hunger, S.P.; Mullighan, C.G.; et al. Aberrant STAT5 and PI3K/mTOR pathway signaling occurs in human CRLF2-rearranged B-precursor acute lymphoblastic leukemia. Blood 2012, 120, 833–842.
  8. Scheeren, F.A.; Van Lent, A.U.; Nagasawa, M.; Weijer, K.; Spits, H.; Legrand, N.; Blom, B. Thymic stromal lymphopoietin induces early human B-cell proliferation and differentiation. Eur. J. Immunol. 2010, 40, 955–965.
  9. Roberts, K.G.; Gu, Z.; Payne-Turner, D.; McCastlain, K.; Harvey, R.C.; Chen, I.-M.; Pei, D.; Iacobucci, I.; Valentine, M.; Pounds, S.B.; et al. High Frequency and Poor Outcome of Philadelphia Chromosome–Like Acute Lymphoblastic Leukemia in Adults. J. Clin. Oncol. 2017, 35, 394–401.
  10. Herold, T.; Schneider, S.; Metzeler, K.H.; Neumann, M.; Hartmann, L.; Roberts, K.G.; Konstandin, N.P.; Greif, P.A.; Bräundl, K.; Ksienzyk, B.; et al. Adults with Philadelphia chromosome–like acute lymphoblastic leukemia frequently have IGH-CRLF2 and JAK2 mutations, persistence of minimal residual disease and poor prognosis. Haematologica 2016, 102, 130–138.
  11. Mullighan, C.G.; Collins-Underwood, J.R.; Phillips, L.A.; Loudin, M.L.; Liu, W.; Zhang, J.; Ma, J.; Coustan-Smith, E.; Harvey, R.C.; Willman, C.L.; et al. Rearrangement of CRLF2 in B-progenitor– and Down syndrome–associated acute lymphoblastic leukemia. Nat. Genet. 2009, 41, 1243–1246.
  12. Yoda, A.; Yoda, Y.; Chiaretti, S.; Bar-Natan, M.; Mani, K.; Rodig, S.J.; West, N.; Xiao, Y.; Brown, J.R.; Mitsiades, C.; et al. Functional screening identifies CRLF2 in precursor B-cell acute lymphoblastic leukemia. Proc. Natl. Acad. Sci. USA 2010, 107, 252–257.
  13. Hertzberg, L.; Vendramini, E.; Ganmore, I.; Cazzaniga, G.; Schmitz, M.; Chalker, J.; Shiloh, R.; Iacobucci, I.; Shochat, C.; Zeligson, S.; et al. 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 2010, 115, 1006–1017.
  14. Russell, L.J.; Enshaei, A.; Jones, L.; Erhorn, A.; Masic, D.; Bentley, H.; Laczko, K.S.; Fielding, A.K.; Goldstone, A.H.; Goulden, N.; et al. Translocations Are Prevalent in Teenagers and Young Adults with Acute Lymphoblastic Leukemia and Are Associated with a Poor Outcome. J. Clin. Oncol. 2014, 32, 1453–1462.
  15. Schmäh, J.; Fedders, B.; Panzer-Grümayer, R.; Fischer, S.; Zimmermann, M.; Dagdan, E.; Bens, S.; Schewe, D.; Moericke, A.; Alten, J.; et al. Molecular characterization of acute lymphoblastic leukemia with high CRLF2 gene expression in childhood. Pediatr. Blood Cancer 2017, 64, e26539.
  16. Konoplev, S.; Lu, X.; Konopleva, M.; Jain, N.; Ouyang, J.; Goswami, M.; Roberts, K.G.; Valentine, M.; Mullighan, C.G.; Bueso-Ramos, C.; et al. CRLF2-Positive B-Cell Acute Lymphoblastic Leukemia in Adult Patients: A Single-Institution Experience. Am. J. Clin. Pathol. 2017, 147, 357–363.
  17. Imamura, T.; Kiyokawa, N.; Kato, M.; Imai, C.; Okamoto, Y.; Yano, M.; Ohki, K.; Yamashita, Y.; Kodama, Y.; Saito, A.; et al. Characterization of pediatric Philadelphia-negative B-cell precursor acute lymphoblastic leukemia with kinase fusions in Japan. Blood Cancer J. 2016, 6, e419.
  18. Roberts, K.G.; Reshmi, S.C.; Harvey, R.C.; Chen, I.-M.; Patel, K.; Stonerock, E.; Jenkins, H.; Dai, Y.; Valentine, M.; Gu, Z.; et al. Genomic and outcome analyses of Ph-like ALL in NCI standard-risk patients: A report from the Children’s Oncology Group. Blood 2018, 132, 815–824.
  19. Reshmi, S.C.; Harvey, R.C.; Roberts, K.G.; Stonerock, E.; Smith, A.; Jenkins, H.; Chen, I.-M.; Valentine, M.; Liu, Y.; Li, Y.; et al. Targetable kinase gene fusions in high-risk B-ALL: A study from the Children’s Oncology Group. Blood 2017, 129, 3352–3361.
  20. Tasian, S.K.; Hurtz, C.; Wertheim, G.B.; Bailey, N.G.; Lim, M.S.; Harvey, R.C.; Chen, I.-M.; Willman, C.L.; Astles, R.; Zebrowski, A.; et al. High incidence of Philadelphia chromosome-like acute lymphoblastic leukemia in older adults with B-ALL. Leukemia 2017, 31, 981–984.
  21. Chiaretti, S.; Messina, M.; Grammatico, S.; Piciocchi, A.; Fedullo, A.L.; Di Giacomo, F.; Peragine, N.; Gianfelici, V.; Lauretti, A.; Bareja, R.; et al. Rapid identification ofBCR/ABL1-like acute lymphoblastic leukaemia patients using a predictive statistical model based on quantitative real time-polymerase chain reaction: Clinical, prognostic and therapeutic implications. Br. J. Haematologica 2018, 181, 642–652.
  22. Harvey, R.C.; Mullighan, C.G.; Chen, I.-M.; Wharton, W.; Mikhail, F.M.; Carroll, A.J.; Kang, H.; Liu, W.; Dobbin, K.K.; Smith, M.A.; et al. Rearrangement of CRLF2 is associated with mutation of JAK kinases, alteration of IKZF1, Hispanic/Latino ethnicity, and a poor outcome in pediatric B-progenitor acute lymphoblastic leukemia. Blood 2010, 115, 5312–5321.
  23. Perez-Andreu, V.; Roberts, K.G.; Harvey, R.C.; Yang, W.; Cheng, C.; Pei, D.; Xu, H.; Gastierfoster, J.M.; E, S.; Lim, J.Y.-S.; et al. Inherited GATA3 variants are associated with Ph-like childhood acute lymphoblastic leukemia and risk of relapse. Nat. Genet. 2013, 45, 1494–1498.
  24. Giddings, B.M.; Whitehead, T.P.; Metayer, C.; Miller, M.D. Childhood leukemia incidence in California: High and rising in the Hispanic population. Cancer 2016, 122, 2867–2875.
  25. van der Veer, A.; Waanders, E.; Pieters, R.; Willemse, M.E.; Van Reijmersdal, S.V.; Russell, L.J.; Harrison, C.J.; Evans, W.E.; van der Velden, V.H.; Hoogerbrugge, P.M.; et al. Independent prognostic value of BCR-ABL1-like signature and IKZF1 deletion, but not high CRLF2 expression, in children with B-cell precursor ALL. Blood 2013, 122, 2622–2629.
  26. Shochat, C.; Tal, N.; Gryshkova, V.; Birger, Y.; Bandapalli, O.R.; Cazzaniga, G.; Gershman, N.; Kulozik, A.E.; Biondi, A.; Mansour, M.R.; et al. Novel activating mutations lacking cysteine in type I cytokine receptors in acute lymphoblastic leukemia. Blood 2014, 124, 106–110.
  27. Iacobucci, I.; Li, Y.; Roberts, K.G.; Dobson, S.M.; Kim, J.C.; Payne-Turner, D.; Harvey, R.C.; Valentine, M.; McCastlain, K.; Easton, J.; et al. Truncating Erythropoietin Receptor Rearrangements in Acute Lymphoblastic Leukemia. Cancer Cell 2016, 29, 186–200.
  28. Russell, L.J.; De Castro, D.G.; Griffiths, M.; Telford, N.; Bernard, O.; Panzer-Grümayer, R.; Heidenreich, O.; Moorman, A.V.; Harrison, C.J. A novel translocation, t(14;19)(q32;p13), involving and the cytokine receptor for erythropoietin. Leukemia 2008, 23, 614–617.
  29. Dai, H.-P.; Yin, J.; Li, Z.; Yang, C.-X.; Cao, T.; Chen, P.; Zong, Y.-H.; Zhu, M.-Q.; Zhu, X.-M.; Xiao, S.; et al. Rapid Molecular Response to Dasatinib in a Pediatric Relapsed Acute Lymphoblastic Leukemia with NCOR1-LYN Fusion. Front. Oncol. 2020, 10, 359.
  30. Arber, D.A.; Orazi, A.; Hasserjian, R.; Thiele, J.; Borowitz, M.J.; Le Beau, M.M.; Bloomfield, C.D.; Cazzola, M.; Vardiman, J.W. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood 2016, 127, 2391–2405.
  31. Den Boer, M.L.; Cario, G.; Moorman, A.V.; Boer, J.M.; de Groot-Kruseman, H.A.; Fiocco, M.; Escherich, G.; Imamura, T.; Yeoh, A.; Sutton, R.; et al. Outcomes of paediatric patients with B-cell acute lymphocytic leukaemia with ABL-class fusion in the pre-tyrosine-kinase inhibitor era: A multicentre, retrospective, cohort study. Lancet Haematol. 2021, 8, e55–e66.
  32. Schwab, C.J.; Roberts, K.G.; Boer, J.M.; Gohring, G.; Steinemann, D.; Vora, A.; Macartney, C.; Hough, R.E.; Thorn, Z.; Dillon, R.; et al. SSBP2-CSF1R is a Recurrent Fusion in B-lineage Acute Lymphoblastic Leukaemia with Diverse Genetic Presentation and Variable Outcome. Blood 2020, 137, 1835–1838.
  33. Schwab, C.; Ryan, S.L.; Chilton, L.; Elliott, A.; Murray, J.; Richardson, S.; Wragg, C.; Moppett, J.; Cummins, M.; Tunstall, O.; et al. EBF1-PDGFRB fusion in pediatric B-cell precursor acute lymphoblastic leukemia (BCP-ALL): Genetic profile and clinical implications. Blood 2016, 127, 2214–2218.
  34. Roberts, K.G.; Yang, Y.-L.; Payne-Turner, D.; Lin, W.; Files, J.K.; Dickerson, K.; Gu, Z.; Taunton, J.; Janke, L.J.; Chen, T.; et al. Oncogenic role and therapeutic targeting of ABL-class and JAK-STAT activating kinase alterations in Ph-like ALL. Blood Adv. 2017, 1, 1657–1671.
  35. Horiuchi, M.; Yoshida, M.; Yamasaki, K.; Sakagami, R.; Aoyama, T.; Tatsumi, N.; Tsutsumi, M.; Nakaya, Y.; Fuseya, H.; Yoshimura, T.; et al. Effective treatment with imatinib for acute B-lymphoblastic leukaemia with EBF1-PDGFRB fusion. Ann. Hematol. 2020, 100, 1329–1331.
  36. Fazio, F.; Barberi, W.; Cazzaniga, G.; Fazio, G.; Messina, M.; Della Starza, I.; De Propris, M.S.; Mancini, F.; Mohamed, S.; Del Giudice, I.; et al. Efficacy of imatinib and chemotherapy in a pediatric patient with Philadelphia-like acute lymphoblastic leukemia with Ebf1-Pdgfrb fusion transcript. Leuk. Lymphoma 2020, 61, 469–472.
  37. Lengline, E.; Beldjord, K.; Dombret, H.; Soulier, J.; Boissel, N.; Clappier, E. Successful tyrosine kinase inhibitor therapy in a refractory B-cell precursor acute lymphoblastic leukemia with EBF1-PDGFRB fusion. Haematologica 2013, 98, e146–e148.
  38. Weston, B.W.; Hayden, M.A.; Roberts, K.G.; Bowyer, S.; Hsu, J.; Fedoriw, G.; Rao, K.W.; Mullighan, C.G. Tyrosine Kinase Inhibitor Therapy Induces Remission in a Patient with Refractory EBF1-PDGFRB–Positive Acute Lymphoblastic Leukemia. J. Clin. Oncol. 2013, 31, e413–e416.
  39. Tran, T.H.; Nguyen, J.V.; Stecula, A.; Akutagawa, J.; Moorman, A.V.; Braun, B.S.; Sali, A.; Mullighan, C.G.; Shah, N.P.; Dai, Y.; et al. The EBF1-PDGFRB T681I mutation is highly resistant to imatinib and dasatinib in vitro and detectable in clinical samples prior to treatment. Haematologica 2021.
  40. Roberts, K.G.; Janke, L.J.; Zhao, Y.; Seth, A.; Ma, J.; Finkelstein, D.; Smith, S.; Ebata, K.; Tuch, B.B.; Hunger, S.P.; et al. ETV6-NTRK3 induces aggressive acute lymphoblastic leukemia highly sensitive to selective TRK inhibition. Blood 2018, 132, 861–865.
  41. Kralik, J.M.; Kranewitter, W.; Boesmueller, H.; Marschon, R.; Tschurtschenthaler, G.; Rumpold, H.; Wiesinger, K.; Erdel, M.; Petzer, A.L.; Webersinke, G. Characterization of a newly identified ETV6-NTRK3 fusion transcript in acute myeloid leukemia. Diagn. Pathol. 2011, 6, 19.
  42. Kang, J.; Park, J.W.; Won, J.-K.; Bae, J.M.; Koh, J.; Yim, J.; Yun, H.; Kim, S.-K.; Choi, J.Y.; Kang, H.J.; et al. Clinicopathological findings of pediatric NTRK fusion mesenchymal tumors. Diagn. Pathol. 2020, 15, 1–11.
  43. Knezevich, S.R.; McFadden, D.E.; Tao, W.; Lim, J.F.; Sorensen, P.H. A novel ETV6-NTRK3 gene fusion in congenital fibrosarcoma. Nat. Genet. 1998, 18, 184–187.
  44. Leeman-Neill, R.J.; Bs, L.M.K.; Liu, P.; Brenner, A.V.; Leeman-Neill, R.J.; Bogdanova, T.I.; Evdokimova, V.N.; Hatch, M.; Zurnadzy, L.Y.; Nikiforova, M.N.; et al. ETV6-NTRK3 is a common chromosomal rearrangement in radiation-associated thyroid cancer. Cancer 2014, 120, 799–807.
  45. Brenca, M.; Rossi, S.; Polano, M.; Gasparotto, D.; Zanatta, L.; Racanelli, D.; Valori, L.; Lamon, S.; Tos, A.P.D.; Maestro, R. Transcriptome sequencing identifiesETV6-NTRK3as a gene fusion involved in GIST. J. Pathol. 2016, 238, 543–549.
  46. Otsubo, R.; Mussazhanova, Z.; Akazawa, Y.; Sato, A.; Matsuda, K.; Matsumoto, M.; Yano, H.; Matsuse, M.; Mitsutake, N.; Ando, T.; et al. Sporadic pediatric papillary thyroid carcinoma harboring the ETV6/NTRK3 fusion oncogene in a 7-year-old Japanese girl: A case report and review of literature. J. Pediatr. Endocrinol. Metab. 2018, 31, 461–467.
  47. Tognon, C.; Knezevich, S.R.; Huntsman, D.; Roskelley, C.D.; Melnyk, N.; Mathers, J.A.; Becker, L.; Carneiro, F.; MacPherson, N.; Horsman, D.; et al. Expression of the ETV6-NTRK3 gene fusion as a primary event in human secretory breast carcinoma. Cancer Cell 2002, 2, 367–376.
  48. Martinelli, G.; Iacobucci, I.; Storlazzi, C.T.; Vignetti, M.; Paoloni, F.; Cilloni, D.; Soverini, S.; Vitale, A.; Chiaretti, S.; Cimino, G.; et al. IKZF1 (Ikaros) Deletions in BCR-ABL1–Positive Acute Lymphoblastic Leukemia Are Associated with Short Disease-Free Survival and High Rate of Cumulative Incidence of Relapse: A GIMEMA AL WP Report. J. Clin. Oncol. 2009, 27, 5202–5207.
  49. Raca, G.; Abdel-Azim, H.; Yue, F.; Broach, J.; Payne, J.L.; Reeves, M.E.; Gowda, C.; Schramm, J.; Desai, D.; Dovat, E.; et al. Increased Incidence of IKZF1 deletions and IGH-CRLF2 translocations in B-ALL of Hispanic/Latino children—a novel health disparity. Leukemia 2021, 1–4.
  50. Nardi, V.; Ku, N.; Frigault, M.J.; Dubuc, A.M.; Tsai, H.K.; Amrein, P.C.; Hobbs, G.S.; Brunner, A.M.; Narayan, R.; Burke, M.E.; et al. Clinical response to larotrectinib in adult Philadelphia chromosome-like ALL with cryptic ETV6-NTRK3 rearrangement. Blood Adv. 2020, 4, 106–111.
  51. Gu, Z.; Churchman, M.; Roberts, K.; Li, Y.; Liu, Y.; Harvey, R.C.; McCastlain, K.; Reshmi, S.C.; Payne-Turner, D.; Iacobucci, I.; et al. Genomic analyses identify recurrent MEF2D fusions in acute lymphoblastic leukaemia. Nat. Commun. 2016, 7, 13331.
  52. Tan, J.E.-L.; Wong, S.-C.; Gan, S.K.-E.; Xu, S.; Lam, K.-P. The Adaptor Protein BLNK Is Required for B Cell Antigen Receptor-induced Activation of Nuclear Factor-κB and Cell Cycle Entry and Survival of B Lymphocytes. J. Biol. Chem. 2001, 276, 20055–20063.
  53. Benedict, C.L.; Gilfillan, S.; Thai, T.H.; Kearney, J.F. Terminal deoxynucleotidyl transferase and repertoire development. Immunol. Rev. 2000, 175, 150–157.
  54. Mullighan, C.G.; Su, X.; Zhang, J.; Radtke, I.; Phillips, L.A.; Miller, C.B.; Ma, J.; Liu, W.; Cheng, C.; Schulman, B.A.; et al. Deletion ofIKZF1and Prognosis in Acute Lymphoblastic Leukemia. N. Engl. J. Med. 2009, 360, 470–480.
  55. Roberts, K.G.; Pei, D.; Campana, D.; Payne-Turner, D.; Li, Y.; Cheng, C.; Sandlund, J.T.; Jeha, S.; Easton, J.; Becksfort, J.; et al. Outcomes of Children with BCR-ABL1–Like Acute Lymphoblastic Leukemia Treated with Risk-Directed Therapy Based on the Levels of Minimal Residual Disease. J. Clin. Oncol. 2014, 32, 3012–3020.
More
Academic Video Service