13
Tumor and Stem Cell Biology Activated KRAS Cooperates with MLL-AF4 to Promote Extramedullary Engraftment and Migration of Cord Blood CD34 þ HSPC But Is Insufcient to Initiate Leukemia Cristina Prieto 1 , Ronald W. Stam 2 , Antonio Agraz-Doblas 1,3 , Paola Ballerini 4 , Mireia Camos 5 , Julio Casta ~ no 1 , Rolf Marschalek 6 , Aldeheid Bursen 6 , Ignacio Varela 3 , Clara Bueno 1 , and Pablo Menendez 1,7 Abstract The MLLAF4 (MA4) fusion gene is the genetic hallmark of an aggressive infant proB-acute lymphoblastic leukemia (B-ALL). Our understanding of MA4-mediated transformation is very limited. Whole-genome sequencing studies revealed a silent mutational landscape, which contradicts the aggressive clinical outcome of this hematologic malignancy. Only RAS mutations were recurrently detected in patients and found to be associated with poorer outcome. The absence of MA4-driven B-ALL models further questions whether MA4 acts as a single oncogenic driver or requires cooperating mutations to manifest a malignant phenotype. We explored whether KRAS activation cooperates with MA4 to initiate leukemia in cord bloodderived CD34 þ hematopoietic stem/progenitor cells (HSPC). Clonogenic and differentiation/proliferation assays demonstrated that KRAS activation does not cooperate with MA4 to immortalize CD34 þ HSPCs. Intrabone marrow transplantation into immu- nodecient mice further showed that MA4 and KRAS G12V alone or in combination enhanced hematopoietic repopulation with- out impairing myeloidlymphoid differentiation, and that mutated KRAS did not cooperate with MA4 to initiate leukemia. However, KRAS activation enhanced extramedullary hematopoi- esis of MA4-expressing cell lines and CD34 þ HSPCs that was associated with leukocytosis and central nervous system inl- tration, both hallmarks of infant t(4;11) þ B-ALL. Transcriptional proling of MA4-expressing patients supported a cell migration gene signature underlying the mutant KRAS-mediated pheno- type. Collectively, our ndings demonstrate that KRAS affects the homeostasis of MA4-expressing HSPCs, suggesting that KRAS activation in MA4 þ B-ALL is important for tumor maintenance rather than initiation. Cancer Res; 76(8); 247889. Ó2016 AACR. Introduction The translocation t(4;11)(q21;q23) encodes the chimeric pro- tein MLLAF4 (MA4), the hallmark genetic abnormality associ- ated to ghastly infant proB-acute lymphoblastic leukemia (B- ALL) that arises prenatally during early hematopoiesis (1, 2). Newborn proB-ALL is highly aggressive with high tumor burdens and white blood cell (WBC) counts, therapy resistance, and an extremely short latency, raising the question of how it evolves so quickly (3). Our understanding of MA4-mediated transformation is limited, and unlike other MLL fusions, MA4-induced leukemo- genesis has been difcult to model. Current murine-human disease models do not faithfully recapitulate the pathogenesis/ phenotype (46). It could be argued that the absence of a disease model may be because of (i) a cell in a wrong developmental stage was targeted or (ii) the impact of other secondary hits has not been properly addressed. We have previously reported that the expres- sion of MA4 on its own enhanced the hematopoietic engraftment and clonogenic potential of human neonatal cord blood (CB)CD34 þ hematopoietic stem/progenitor cells (HSPC), but is not sufcient for leukemogenesis (7). MA4 also failed to transform CD34 þ HSPCs in combination with FLT3 activation, which has been proposed a candidate cooperating oncogenic insult (8). Recent whole-genome sequencing (WGSeq) studies on MLL- rearranged (MLL-r) infants revealed that this infant leukemia has one of the lowest frequencies of somatic mutations of any sequenced cancer, contradicting its aggressive clinical outcome (912). This, coupled to the absence of models reproducing this leukemia, feeds the debate of whether MA4 acts as a single-driver mutation in a critical target cell or cooperating mutations need to be accrued for a malignant phenotype. Despite the paucity of mutations observed in WGSeq studies, activating mutations in the RAS signaling pathway were found in half of the patients (912), and previous screenings of infants with MA4 þ proB-ALL also 1 Josep Carreras Leukemia Research Institute and Department of Bio- medicine, School of Medicine, University of Barcelona, Barcelona, Spain. 2 Pediatric Oncology/Hematology, Erasmus MC-Sophia Chil- dren's Hospital, Rotterdam, the Netherlands. 3 Institute of Biomedicine and Biotechnology of Cantabria (IBBTEC-CSIC-UNIVERSIDAD DE CANTABRIA-SODERCAN), Santander, Spain. 4 Pediatric Hematology Department, A. Trousseau Hospital, Paris, France. 5 Hematology Lab- oratory, Hospital Sant Joan de Deu, Barcelona, Spain. 6 Institute Phar- maceutical Biology, Goethe-University, Frankfurt/Main, Germany. 7 Instituci o Catalana Recerca i Estudis Avan¸ cats (ICREA), Barcelona, Spain. C. Bueno and P. Menendez contributed equally to this article. Corresponding Authors: Pablo Menendez, Instituci o Catalana Recerca i Estudis Avan¸ cats (ICREA), Carrer Casanova 143, 08036 Barcelona, Spain. Phone: 34-935572809; Fax: 34-933231751; E-mail: [email protected]; or Clara Bueno, E-mail: [email protected] doi: 10.1158/0008-5472.CAN-15-2769 Ó2016 American Association for Cancer Research. Cancer Research Cancer Res; 76(8) April 15, 2016 2478 on October 8, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from Published OnlineFirst February 2, 2016; DOI: 10.1158/0008-5472.CAN-15-2769

Activated KRAS Cooperates with MLL-AF4 to Promote ... · and previous screenings of infants with MA4þ pro–B-ALL also 1Josep Carreras Leukemia Research Institute and Department

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Activated KRAS Cooperates with MLL-AF4 to Promote ... · and previous screenings of infants with MA4þ pro–B-ALL also 1Josep Carreras Leukemia Research Institute and Department

Tumor and Stem Cell Biology

Activated KRAS Cooperates with MLL-AF4 toPromote Extramedullary Engraftment andMigration of Cord Blood CD34þ HSPC But IsInsufficient to Initiate LeukemiaCristina Prieto1, Ronald W. Stam2, Antonio Agraz-Doblas1,3, Paola Ballerini4,Mireia Camos5, Julio Casta~no1, Rolf Marschalek6, Aldeheid Bursen6,Ignacio Varela3, Clara Bueno1, and Pablo Menendez1,7

Abstract

The MLL–AF4 (MA4) fusion gene is the genetic hallmark of anaggressive infant pro–B-acute lymphoblastic leukemia (B-ALL).Our understanding of MA4-mediated transformation is verylimited. Whole-genome sequencing studies revealed a silentmutational landscape, which contradicts the aggressive clinicaloutcome of this hematologic malignancy. Only RAS mutationswere recurrently detected in patients and found to be associatedwith poorer outcome. The absence of MA4-driven B-ALL modelsfurther questions whether MA4 acts as a single oncogenic driveror requires cooperating mutations to manifest a malignantphenotype. We explored whether KRAS activation cooperateswith MA4 to initiate leukemia in cord blood–derived CD34þ

hematopoietic stem/progenitor cells (HSPC). Clonogenic anddifferentiation/proliferation assays demonstrated that KRASactivation does not cooperate with MA4 to immortalize

CD34þ HSPCs. Intrabone marrow transplantation into immu-nodeficient mice further showed that MA4 and KRASG12V aloneor in combination enhanced hematopoietic repopulation with-out impairing myeloid–lymphoid differentiation, and thatmutated KRAS did not cooperate with MA4 to initiate leukemia.However, KRAS activation enhanced extramedullary hematopoi-esis of MA4-expressing cell lines and CD34þ HSPCs that wasassociated with leukocytosis and central nervous system infil-tration, both hallmarks of infant t(4;11)þ B-ALL. Transcriptionalprofiling of MA4-expressing patients supported a cell migrationgene signature underlying the mutant KRAS-mediated pheno-type. Collectively, our findings demonstrate that KRAS affects thehomeostasis of MA4-expressing HSPCs, suggesting that KRASactivation in MA4þ B-ALL is important for tumor maintenancerather than initiation. Cancer Res; 76(8); 2478–89. �2016 AACR.

IntroductionThe translocation t(4;11)(q21;q23) encodes the chimeric pro-

tein MLL–AF4 (MA4), the hallmark genetic abnormality associ-ated to ghastly infant pro–B-acute lymphoblastic leukemia (B-ALL) that arises prenatally during early hematopoiesis (1, 2).Newbornpro–B-ALL is highly aggressivewithhigh tumor burdensand white blood cell (WBC) counts, therapy resistance, and an

extremely short latency, raising the question of how it evolves soquickly (3). Our understanding ofMA4-mediated transformationis limited, and unlike other MLL fusions, MA4-induced leukemo-genesis has been difficult to model. Current murine-humandisease models do not faithfully recapitulate the pathogenesis/phenotype (4–6). It could be argued that the absence of a diseasemodelmay be because of (i) a cell in awrong developmental stagewas targeted or (ii) the impact of other secondary hits has not beenproperly addressed. We have previously reported that the expres-sion of MA4 on its own enhanced the hematopoietic engraftmentand clonogenic potential of human neonatal cord blood (CB)–CD34þ hematopoietic stem/progenitor cells (HSPC), but is notsufficient for leukemogenesis (7). MA4 also failed to transformCD34þ HSPCs in combination with FLT3 activation, which hasbeen proposed a candidate cooperating oncogenic insult (8).

Recent whole-genome sequencing (WGSeq) studies on MLL-rearranged (MLL-r) infants revealed that this infant leukemia hasone of the lowest frequencies of somatic mutations of anysequenced cancer, contradicting its aggressive clinical outcome(9–12). This, coupled to the absence of models reproducing thisleukemia, feeds the debate of whether MA4 acts as a single-drivermutation in a critical target cell or cooperating mutations need tobe accrued for a malignant phenotype. Despite the paucity ofmutations observed inWGSeq studies, activatingmutations in theRAS signaling pathway were found in half of the patients (9–12),and previous screenings of infants with MA4þ pro–B-ALL also

1Josep Carreras Leukemia Research Institute and Department of Bio-medicine, School of Medicine, University of Barcelona, Barcelona,Spain. 2Pediatric Oncology/Hematology, Erasmus MC-Sophia Chil-dren's Hospital, Rotterdam, theNetherlands. 3Institute of Biomedicineand Biotechnology of Cantabria (IBBTEC-CSIC-UNIVERSIDAD DECANTABRIA-SODERCAN), Santander, Spain. 4Pediatric HematologyDepartment, A. Trousseau Hospital, Paris, France. 5Hematology Lab-oratory, Hospital Sant Joan de Deu, Barcelona, Spain. 6Institute Phar-maceutical Biology, Goethe-University, Frankfurt/Main, Germany.7Instituci�o Catalana Recerca i Estudis Avancats (ICREA), Barcelona,Spain.

C. Bueno and P. Menendez contributed equally to this article.

Corresponding Authors: Pablo Menendez, Instituci�o Catalana Recerca i EstudisAvancats (ICREA), Carrer Casanova 143, 08036 Barcelona, Spain. Phone:34-935572809; Fax: 34-933231751; E-mail: [email protected];or Clara Bueno, E-mail: [email protected]

doi: 10.1158/0008-5472.CAN-15-2769

�2016 American Association for Cancer Research.

CancerResearch

Cancer Res; 76(8) April 15, 20162478

on October 8, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 2, 2016; DOI: 10.1158/0008-5472.CAN-15-2769

Page 2: Activated KRAS Cooperates with MLL-AF4 to Promote ... · and previous screenings of infants with MA4þ pro–B-ALL also 1Josep Carreras Leukemia Research Institute and Department

found RAS mutations in approximately 25% of patients thatcorrelated with poor outcome. KRAS mutations have also beenassociated with higher WBC counts (13) and found subclonallypresent at birth in neonatal blood spots (14). Furthermore, in aMA4þ transgenic mouse model, activated KRAS accelerated leu-kemogenesis, although the latency/phenotype did not reproducethat seen in patients (15). Similarly, in a humanized MLL-AF10model, the coexpression of KRASG12Vwas required for a leukemicphenotype (16, 17).

Here, we explored whether activation of KRAS cooperates withMA4 to transform/immortalize CB-derived CD34þ HSPCs. Ourresults indicate that KRASG12V alone or in combination withMA4does not initiate leukemia. However, enforced KRASG12V expres-sion enhanced extramedullary hematopoiesis and invasion ofMA4-expressing CD34þHSPCs and MA4þ cell lines associated toelevated WBC counts and central nervous system (CNS) infiltra-tion, hallmarks ofMA4þ pro–B-ALL infants. These functional andclinical data indicate that activated KRAS does not cooperate withMA4 to initiate leukemia but influences the homeostasis ofMA4-expressing CD34þ HSPCs, suggesting that KRAS activation inMA4þ infant B-ALL may be important in tumor maintenancerather than initiation, thus supporting WGSeq studies showingthat RAS mutations in infant MLL-r leukemia are subclonal andlost at relapse. We conclude that MA4-mediated transformationmight depend on alternative (epi)-genetic cooperating lesionsand on a critical developmentally earlier window of stem cellvulnerability.

Materials and MethodsCB collection and CD34þ HSPCs isolation

Umbilical CB units (n ¼ 100) from healthy newborns wereobtained from the Catalonia Blood Tissue Bank following theinstitutional guidelines approved by our local InstitutionalReview Board. CBs were pooled to reduce variability amongindividual CB units, and mononuclear cells were isolatedby density gradient centrifugation using Ficoll–Hypaque. Afterlysing red cells, CD34þ cells were purified by magnetic beadseparation using the human CD34 MicroBead Kit and the Auto-MACS Pro-separator (Miltenyi Biotec) as per the manufacturer'sinstructions (Fig. 1A; refs.18, 19). Purity of the CD34þ fractionswas always assessed by flow cytometry using anti–CD34-PE(BD Biosciences), and only CD34þ fractions with purity >95%were used. The CD34� fraction was irradiated and used as acces-sory cells for cotransplantation with CD34þ HSPCs.

Plasmids and lentiviral transductionThe MA4 cDNA (7, 8) and the KRASG12V cDNA (kindly pro-

vided by professor Naoto Ishii, Tohoku University, Japan) weresubcloned into the pRRL-EF1a-PGK-GFP/dTo vector. The follow-ing lentivectors were used: pRRL-EF1a-PGK-GFP/dTo (emptyvector, EV), pRRL-EF1a-MA4-PGK-GFP (MA4), and pRRL-EF1a-KRASG12V-PGK-dTo (KRAS; Fig. 1B). Vesicular stomatitisvirus-G (VSV-G)–pseudotyped viral particles were generated on293T cells by calciumphosphate transfection and concentrated byultracentrifugation (7, 20). Human CD34þHSPCs (2� 106 cells)were infected overnight with concentrated viruses and polybrene(1 mg/mL; Sigma-Aldrich) and the hematopoietic cytokines StemCell Factor (100 ng/mL), FMS-like tyrosine kinase 3 ligand (100ng/mL), and IL3 (10 ng/mL, all from PeproTech). Viral superna-tant was removed 14 hours later, and transduced cells werewashed and maintained in culture in Stemspan medium (Stem

Cell Technologies; refs. 7, 21). For in vitro and in vivo experiments,transduced CD34þ cells were FACS-sorted (FACSAria) 36 hoursafter infection based on reporter expression: GFPþ for MA4-transduced CD34þ, dToþ for KRAS-transduced cells, and mergedyellow (GFPþdToþ) for MA4þKRAS cotransduced cells. The SEM(MA4þ) cell line was transduced with KRASG12V, FACS-purified,and xenografted as described below. SEM cells were obtainedfrom the DSMZ-German Collection Cell Bank (www.dsmz.de)and were routinely authenticated/characterized by PCR for MA4and FISH for MLL break-apart.

Mice transplantationNon-obese diabetic/LtSz-scid IL-2Rg�/� mice (NSG; n ¼ 50)

were used for in vivo assays (22, 23). Animals were housed underpathogen-free conditions, and the procedures were approved bythe Animal CareCommittee of Barcelona Parc Recerca Biomedica.Seven to 12-week-old mice were sublethally irradiated (2.25 Gy)6 to 16 hours before transplantation. They were anesthetized withisoflurane 3% inhalation, and intrabone marrow transplantation(IBMT) was performed (23, 24). A total of 3 � 104 transduced/sorted CD34þ HSPCs (along with 5 � 104 irradiated accessorycells) were transplanted in a 25-mL volume. For pain relief,buprenorphine:meloxicam was subcutaneously administeredduring the transplantation and 24 hours later. Mice were mon-itored throughout the entire experiment and killed at any sign ofdisease or 16 weeks after transplantation. For SEM assays, 2� 105

cells were IBM-transplanted.

Analysis of peripheral blood hematologic counts andmacroscopic parameters

Hematologic parameters, including absolute WBC, platelets,and hemoglobin levels, were determined in mice peripheralblood (PB) at sacrifice using the hematologic analyzer SysmexSX-800i (7). Spleen and liver were visualized macroscopically,measured, and weighted.

Analysis of engraftmentMice were killed 16 weeks after transplantation. Bonemarrow

(BM) from injected tibia (IT), contralateral tibia and femur(CL), liver, spleen, and PB were collected and analyzed forhuman chimerism. Cells were stained with anti-HLA.ABC-PEand CD45-APC.Cy7, and human chimerism was analyzedby FACS. All engrafted mice were assessed for multilineageanalysis using anti–CD33-APC for myeloid cells, anti–CD19-V450 for lymphoid cells, and anti–CD34-PE.Cy7 for immaturecells. Within the CD19þ cell subset, the percentage of CD34þ

and CD10þ cells was analyzed with anti–CD10-PerCP.Cy5.5to distinguish between pro-B and pre-B cells. In addition,the following CD34þ subsets were analyzed: immature pro-genitors (CD34þCD19�CD33�CD38�), myeloid progenitors(CD34þCD33þ), and B-cell progenitors (CD34þCD19þ).

In vitro liquid culture of transduced CD34þ cells, apoptosis,cell-cycle, and senescence assays

For liquid culture, transduced CD34þ HSPCs were FACS-sorted based on reporter expression. Purified GFPþ (MA4-expressing), dToþ (KRAS-expressing), or merged yellow (MA4-and KRAS-coexpressing) CD34þ HSPCs were allowed toexpand for a period of 40 to 50 days. To determine the growthkinetics, cells were counted every 5 days and replated at adensity of 1 � 105 cells/cm2 (7, 25). Furthermore, the expres-sion of CD34 antigen was analyzed every 3 to 4 days. To

KRAS Mutation and MLL–AF4 in Cord Blood CD34þ Cells

www.aacrjournals.org Cancer Res; 76(8) April 15, 2016 2479

on October 8, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 2, 2016; DOI: 10.1158/0008-5472.CAN-15-2769

Page 3: Activated KRAS Cooperates with MLL-AF4 to Promote ... · and previous screenings of infants with MA4þ pro–B-ALL also 1Josep Carreras Leukemia Research Institute and Department

analyze cell-cycle distribution, transduced CD34þ HSPCs werefixed in 70% ice-cold ethanol and stored at �20�C. Subse-quently, cells were suspended in propidium iodide (PI) buffercontaining 5 mg PI and 100 mg/mL RNAase. Cell-cycle distri-bution was analyzed on a FACSCanto-II cytometer using theFACSDiva software to discriminate among resting cells (G0–

G1), cycling cells (S phase), and G2–M cells (25). The apoptoticstatus of transduced CD34þ HSPCs was assessed using AnnexinV as previously detailed (25). Senescence was measured onfixed CD34þ HSPCs by b-galactosidase staining as previouslydescribed (26).

Clonogenic progenitor assayHuman clonogenic progenitor assays were performed by plat-

ing 1,000 sorted EV-, MA4-, KRAS-, or MA4þKRAS-expressingCD34þHSPCs inmethylcellulose (n¼ 7). Colonies were countedand scored at days 10 to 12 of the assay using standard morpho-logic criteria (7, 27, 28). For secondary replating, all the colonyforming units (CFU) from each experimental condition wereharvested, and a single-cell suspension was achieved and replatedas above. The resultingday 12CFUcultureswereharvested, single-cell dissociated, washed, and immunophenotyped using anti–CD33-APC and anti–CD15-BV510 antibodies (BD Biosciences).

Cell migration assaysMouse serum was harvested from healthy mice by blood

centrifugation. A total volume of 600 mL of RPMI supplementedwith 10%murine serumwas added to the lower chamber of a 24-well transwell (Corning). CD34þ or SEM (1 � 105) in 100 mL ofmedium were loaded to the upper chamber (pore size 8 mm) andwere allowed to migrate for 4 hours at 37�C. Migrating cells werecollected from the lower chamber, counted, and characterizedimmunophenotypically.

RNA extraction, cDNA synthesis, PCR, and RT-PCRRNA was isolated from in vitro cultures and BM of transplanted

mice with an RNeasy kit (QIAGEN) and treated with DNaseI.cDNA was synthesized using the High-Capacity cDNA ReverseTranscription Kit (Applied Biosystems). cDNA was used for con-ventional (MA4, KRAS, and GAPDH) and quantitative (HOXA9,PROM1, CXCR4, MET, NELIN, and DCL1) PCR. PCR for MA4,KRAS, and GAPDH was done using the following primers: MA4,fwd 50-CAGAGCAAACAGAAAAAAGTG-30 and rev 50-TCCACAG-TCCCTTCCAGAAC-30; GAPDH, fwd 50-GAAAGCCTGCCGGT-GACTAA-30 and rev 50-CTCCGGGTGATGCTTTTCCT-30; andKRAS, fwd 50-TGGACTACAAAGACGATGACG-30 and rev 50-CCCTCCCCAGTCCTCATGTA-30. PCR conditions were 95�C

p-ERK1/2 p-AKT

Corre

cted M

FI

EVKRAS

F

MNCisolation

Cord blood CD34+ MACS isolation

Lentiviral transduction

Transduced cell FACS purification

In vitroassays

In vivoassays

A

Day

GEVKRAS

Nº B

aF3 c

ells (

×105 )

B

EV KRASMA4 MA4KRAS

E

mRNA

relat

ive ex

pres

sion

D

GAPDH

C

KRAS

-dTo

105

102

10510MLLAF4-GFPGFP

dTo

105

102

105102

Untransduced

KRAS

-dTo

105

102

105102

GFP

KRAS-transduced

dTo

105

102

105102

MLLAF4-GFP

MA4-transduced MA4+KRAS-transduced

x2

x2

HOXA9PROM1

SIN 5’ LTR EF1a KRASG12V PGK dTo SIN 3’ LTR

SIN 5’ LTR EF1a PGK GFP SIN 3’ LTR

SIN 5’ LTR EF1a MA4 PGK GFP SIN 3’ LTR

KRAS

MA4

EV KRAS MA4MA4

KRASH2O

120 bp

253 bp

382 bp

15

10

5

0

1,000

800

600

400

200

0

80604020

1086420

5 10 15 200

Figure 1.Lentiviral-mediated expression and activation of MA4 and KRAS in CD34þ HSPCs. A, outline of the experimental design. B, schematic representation of thelentivectors used. MA4 and KRASG12V vectors express GFP and dTomato as reporter, respectively. C, representative flow cytometry plots showing howtransduced cellswere FACSpurified: GFPþ cells (green) inMA4-transduced, dToþ cells (red) in KRASG12V-transduced, andGFPþdToþ cells (yellow) in KRASG12V- andMA4-double transduced CD34þ HSPCs. Mock-transduced cells (left) were used as a reference control (n ¼ 12). D, RT-PCR confirming ectopic expressionofMA4 and KRASG12V in CB-CD34þ cells. E, RT-qPCR confirming approximately 2- to 10-fold upregulated expression of the MA4 downstream effectors HOXA9 andPROM1 in MA4-expressing CD34þ HSPCs. F, phosphoflow assay demonstrating activation of KRAS downstream effectors pERK1/2 and pAKT. G, KRASG12V

expression renders immortalization/IL3-independent growth of the pro–B-cell line BaF3 (n ¼ 2).

Prieto et al.

Cancer Res; 76(8) April 15, 2016 Cancer Research2480

on October 8, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 2, 2016; DOI: 10.1158/0008-5472.CAN-15-2769

Page 4: Activated KRAS Cooperates with MLL-AF4 to Promote ... · and previous screenings of infants with MA4þ pro–B-ALL also 1Josep Carreras Leukemia Research Institute and Department

(2minutes) followed by 40 cycles of 95�C (20 seconds), 60�C (20seconds), and 72�C (30 seconds) and a final extension of 72�C(10 minutes). The resulted amplicons (KRAS, 253 bp; MA4, 382bp; GAPDH, 120 bp) were resolved on agarose gels. For compar-ative expression of HOXA9, PROM1, CXCR4, MET, NELIN, andDCL1 genes, the following primers were used: HOXA9, fwd 50-AAGACCGAGCAAAAGACGAG-30 and rev 50-GGGTGAGAGAA-GGGAGAAGG-30; PROM1, fwd 50-CTTTCTCCTGCCTCCCGC-30

and rev 50-TTTATGACCCGGCTTCTGGG-30; CXCR4, fwd 50-ATAGTCCCCTGAGCCCATTT-30 and rev 50-AGCAGGTAGCAAA-GTGACG-30; MET, fwd 50-CCATCCAGTGTCTCCAGAAGTG-30

and rev 50-TTCCCAGTGATAACCAGTGTGTAG-30, NELIN, fwd50-CGACGAACAGAGGAGGAAC-30 and rev 50-TTCTCTGCTC-AATCCAAAGGT-30; and DLC1, fwd 50-GGACAGAGATGCCATT-GAGGCT-30 and rev 50-CACAAGGCTCATCCTCGTCTGA-30. qRT-PCR conditions were 95�C (10 minutes) followed by 40 cycles of95�C (30 seconds), 60�C (60 seconds), and 72�C (60 seconds).

Phosphoflow assays for AKT and ERK1/2For phosphosignaling studies, cells were resuspended in stain-

ing buffer, fixed with Cytofix buffer (BD Biosciences), and per-meabilized in prechilled Phosphoflow PermBuffer-III for 30minutes before staining with anti–phospho-ERK1/2 (pT302/pY204)-PE and anti–phospho-AKT (pS473)-PE. Data were ana-lyzed in a FACSCanto-II cytometer using FACSDiva. The meanfluorescence intensity was compared for KRASG12V-transducedversus EV-transduced CD34þ HSPCs.

BaF3 assayIL3-dependent pro–B-cell line BaF3 was used to confirm

KRASG12V activation. BaF3 cells (2� 106) were infected overnightwith concentrated EV- and KRASG12V-expressing lentivirus. Afterwashing viral particles, transduced BaF3 cells were recovered inRPMI þ 10% FBS þ 10 ng/mL mIL3 for 2 days. Then, mIL3 wasremoved and EV- and KRASG12V-BaF3 cells (2.5 � 104) wereplated without IL3 and counted every 5 days to determine IL3-independent growth.

CNS infiltrationMice skulls were retrieved at sacrifice and fixed in 10% neutral

buffered formalin (pH 6.8) for 2 weeks and then decalcified inEDTA (14%; pH 7.4) for 2 more weeks. Skulls were then embed-ded in paraffin, cut into 4 mm sections, rehydrated, and multiplesections were stained with hematoxylin and eosin (H&E) aspreviously described (29).

Gene expression profilingPB leukemic cells fromMA4-expressing pro–B-ALL infants (n¼

12) were purified for gene expression profiling (GEP). Five andseven patients harbored and lacked RAS mutations, respectively.Total RNA was extracted using TRIzol, and quantified on aspectrophotometer. The integrity of the RNA was assessed on anAgilent-2100 Bioanalyzer. High-quality RNA was reverse tran-scribed using T7-linked oligo-dT primers, and the obtained cDNAwas used as a template to synthesize biotinylated cDNA. LabeledcDNA was then fragmented and hybridized as duplicates toHU133plus2.0 GeneChips (Affymetrix) according to the manu-facturer's guidelines (30). Microarray data are deposited in NCBI(GSE19475). Hierarchical clustering of genes and samples wasperformed with the one minus correlation metric and theunweighted average distance. Because RAS mutations are sub-

clonal, a gene was considered differentially expressed when it was20% deregulated (up/down; P value < 0.05) as compared withthe control. Gene ontology (GO) term analysis was performedusing Gorilla (31–33) publicly available at http://cbl-gorilla.cs.technion.ac.il.

Statistical analysisData are expressed as mean � SEM of independent experi-

ments. Paired or impaired Student t tests were used as corre-sponding to perform statistical comparisons between condi-tions. Statistical significance was defined as P < 0.05.

ResultsSuccessful enforced coexpression of MA4 and KRASG12V in CB-derived CD34þ HSPCs

To determine whether KRASG12V cooperates with MA4 inregulating HSPCs, CD34þ cells were isolated from CB and trans-duced with a lentivector expressing (i) GFP reporter (EV), (ii)MA4-GFP (MA4), (iii) KRASG12V-dTomato (dTo) (KRAS), and(iv) MA4-GFP and KRAS-dTo, simultaneously (Fig. 1A and B).Transduced cellswere FACS-purified (purity>98%)48hours laterbased on GFP/dTo reporter and used for in vitro and in vivo assays(Fig. 1A and C). Proper transgene expression in HSPCs was alsoconfirmed by RT-PCR (Fig. 1D). The ectopic expression ofMA4 inCB-CD34þ HSPCs was comparable with that found in patientprimary blasts (data not shown), and induced the expression oftheMA4 target genesHOXA9 and PROM1 (Fig. 1E). Similarly, theectopic expression of KRASG12V resulted in robust RAS activationas demonstrated by phosphorylation of the downstream effectorsERK1/2 and AKT (Fig. 1F). Functionally, the immortalizationactivity of KRASG12V was confirmed by its capacity to conferIL3-independent growth to BaF3 cells (Fig. 1G).

KRASG12V does not cooperate with MA4 to immortalize in vitroCB-CD34þ cells.

MA4 was reported to enhance the in vitro clonogenic potentialwithout immortalizing CD34þ HSPCs (7, 8). We here examinedwhether KRAS cooperates with MA4 to immortalize CD34þ

HSPCs. The hematopoietic progenitor function of CD34þ

HSPCs was examined by quantitative and qualitative analysisof the CFU capacity of EV-, MA4-, KRAS-, and double-transducedcells (n ¼ 7). Scoring of primary CFUs revealed that progenitorsexpressing MA4 or KRAS alone displayed a higher clonogenicpotential (173 and 232 CFU, respectively, per 1,000 plated cells)compared with EV-transduced HSPCs (108 CFU/1,000 platedcells). However, KRAS did not synergize with MA4 to augmentthe clonogenic potential (Fig. 2A, left). Scoring of primary CFUsrevealed no significant differences in CFU types among condi-tions. Importantly, CFU replating assays revealed a significantdecrease in the secondary CFUs, indicating that KRAS does notcooperate with MA4 in progenitor immortalization (Fig. 2A,left). MA4-GFP and KRAS-dTo expressions were confirmedin CFUs by fluorescence microscopy and RT-PCR, (Fig. 2A, right).In addition, KRAS did not influence the MA4-mediated myeloidterminal differentiation, as demonstrated by similar proportionof CD33þCD15þ (45%–58%) and CD33þCD15� (42%–

55%; Fig. 2B), and identical kinetics of CD34 expression lossin liquid cultures (Fig. 2C), regardless of the genotype. Further-more, KRAS and MA4, either alone or combined, did notblock in vitro B-cell differentiation from CD34þ HSPCs on MS5

KRAS Mutation and MLL–AF4 in Cord Blood CD34þ Cells

www.aacrjournals.org Cancer Res; 76(8) April 15, 2016 2481

on October 8, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 2, 2016; DOI: 10.1158/0008-5472.CAN-15-2769

Page 5: Activated KRAS Cooperates with MLL-AF4 to Promote ... · and previous screenings of infants with MA4þ pro–B-ALL also 1Josep Carreras Leukemia Research Institute and Department

stromal cells using B-lineage growth conditions (data not shown;refs. 34, 35). Long-term liquid culture proliferation assaysrevealed that EV-, MA4-, KRAS-, and KRASþMA4-transducedCD34þ cultures grew similarly; cell growth gradually dropped

off from days 15 to 20, and by day 40, cultures were exhaustedregardless of the genotype (Fig. 2D). In line with the CFU assays,KRAS induced an enhanced transient expansion when activatedalone but not in cooperation with MA4, confirming that KRAS

D

A

EV MA4 KRAS MA4KRAS

No. C

FUs /

1,00

0 cell

s

EGMGMGEMM

(Re)plating 1st 2nd 1st 2nd 1st 2nd 1st 2nd

EV KRAS

MA4

MA4-

KRAS

10X

10X

10X

10X

KRASEV MA4MA4

KRAS H2O

KRAS

MA4

GAPDH 120 bp

253 bp

382 bp

**

No. C

ells ×

105

B

EV MA4 KRAS MA4KRAS

% C

D15+

with

in CD

33+

popu

lation

CD15+

CD15-

0

50

100

150

200

250

100

80

60

40

20

0

EVMA4KRASMA4-KRAS

Day

EV KRAS

MA4

MA4-

KRAS

10X

10X10X

10X

60

40

20

003010 0402

C

Day

% C

D34+

EVMA4KRASMA4-KRAS

100

80

60

40

20

05150 10

G

EV MA4 KRAS MA4-KRAS

% S

enes

cent

cells

Mean: 3.8% 9.2% 55.2% 56.2%

80

60

40

20

0

F

% A

popto

tic ce

lls

Day 9 Day 14

EVMA4KRASMA4-KRAS

% C

ells

G0–G1 G2 S–M

EVMA4KRASMA4-KRAS

E100

80

60

40

20

0

40

30

20

10

0

Figure 2.KRASG12V does not immortalize CD34þ HSPCs but impairs the clonogenic potential ofMA4-expressing CD34þ HSPCs. A, representative experiment (n¼ 7) of CFUsshowing that MA4 and KRASG12V do not support replating clonogenic potential. In MA4/KRASG12V cotransduced CD34þ HSPCs, the clonogenic potentialis partially impaired. E, erythroid; M, monocytic; G, granulocytic; GM, granulomonocytic; GEMM, granulocyte–erythroid–monocytic-megakaryocitic. � , P < 0.05. Topright, representative fluorescence image of CFU colonies (10�) harvested after 12 to 14 days. Bottom right, RT-PCR showing stable expression of MA4 andKRASG12V in individual CFUs. B, flow cytometry analysis at day 14 of CFU assays shows very similar content of CD33þCD15þ (neutrophils) and CD33þCD15�

(monocytes) for the indicated genotypes. C, differentiation over time, measured as loss of CD34 antigen, of the indicated genotypes of HSPCs seeded inliquid cultures (n ¼ 6). D, representative experiment (n ¼ 7) of in vitro cell growth kinetics over a period of 40 to 50 days of FACS-purified, EV-, MA4-,KRAS-, and MA4þKRAS–expressing CD34þ HSPCs. Right plots are representative fluorescence images confirming reporter expression on day 15 cultures. E and F,apoptotic levels (AnnexinVþ; E) and cell-cycle distribution (F) of the indicated genotypes of CD34þHSPCs in liquid culture (n¼ 3). G, percentage ofb-galþ senescentcells in the indicated genotypes of growing CD34þ HSPCs (n ¼ 2). Representative b-gal staining is shown.

Prieto et al.

Cancer Res; 76(8) April 15, 2016 Cancer Research2482

on October 8, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 2, 2016; DOI: 10.1158/0008-5472.CAN-15-2769

Page 6: Activated KRAS Cooperates with MLL-AF4 to Promote ... · and previous screenings of infants with MA4þ pro–B-ALL also 1Josep Carreras Leukemia Research Institute and Department

activation does not cooperate with MA4 to immortalize CD34þ

HSPCs. Interestingly, KRASG12V slightly impaired the clonogenicpotential of MA4-expressing CD34þ HSPCs. Apoptosis levels(Fig. 2E) and cell-cycle distribution (Fig. 2F) were not affectedamong genotypes. However, KRAS induced in vitro senescence ofCD34þ cells (Fig. 2G) and cooperated in vivo with MA4 to shiftthe CD34þ population toward a more immature and B-cellprogenitor phenotypes while compromising the myeloid phe-notype (Fig. 3D, bottom). This may explain, at least in part, theKRAS-driven lower clonogenic potential of MA4-expressingCD34þ HSPCs because human clonogenic assays represent amyeloid read-out.

KRAS activation does not cooperate with MA4 to enhancemultilineage hematopoietic engraftment and fails to initiateMA4-mediated leukemogenesis in vivo

To determine whether KRAS activation cooperates withMA4 inregulating HSPCs in vivo, purified CD34þHSPCs were transducedwith EV, MA4, KRAS, and MA4þKRAS, sorted based on reporterexpression, and 30,000 cells were IBMT-transplanted into suble-thally irradiated NSG mice (n ¼ 50). Animals were monitoredthroughout the entire experiment, and none of the mice showedany sign of disease 16 weeks after transplantation. Enforcedexpression of KRAS, alone or combined with MA4, enhanced thehematopoietic engraftment 3-fold as compared with EV-trans-duced cells (36% and 38%vs. 12%; Fig. 3A), indicating that KRASand MA4 do not synergize to promote more robust/sustainedengraftment potential. The ectopic expression of MA4 and/orKRAS was confirmed by RT-PCR in BM cells derived fromengrafted animals (Fig. 3B).

Next, the composition of the human graft was characterized byFACS, and multilineage repopulation was consistently observedin all engraftedmice, regardless of the expression ofMA4 or KRASand the tissue analyzed (Fig. 3C). The graft was consistentlylymphoid-biased (CD45þCD19þ; 70% � 6.0%), followed byCD45þCD33þ myeloid cells (19% � 4.9%) and CD45þCD34þ

immature cells (11% � 3.6%; Fig. 3D, top). Because leukemicblasts in t(4;11)þ B-ALL are characterized by a CD34þ

CD19þCD10� pro-B phenotype, we further analyzed the pheno-type of the CD45þCD19þ B-cell graft (Fig. 3D, middle). Withinthe CD45þCD19þ B-cell graft, approximately 7% of B-cell pro-genitors (CD19þCD10þCD34þ) coexisted with a predominantpre–B-cell population (CD19þCD10þCD34�; �58%) and moremature B cells (CD19þCD10�CD34�; �5%), confirming a nor-mal B-cell development. Importantly, KRAS cooperates withMA4 to shift the CD34þ population toward a more immaturephenotype (CD34þCD19�CD33�CD38�; 18% MA4þKRAS vs.10% other genotypes) and a B-cell progenitor phenotype(CD34þCD19þ; 72% vs. 58%) while compromising the myeloidphenotype (CD34þCD33þ; 16% MA4þKRAS vs. 33% othergenotypes; Fig. 3D, bottom).

To further confirm that KRAS did not contribute to MA4-mediated leukemia initiation, all the animals lacking diseasesignals after 16 weeks were sacrificed, and neither splenomegalynor hepatomegaly was observed regardless the genotype (Fig.3E). Besides, hemoglobin levels and platelet counts were nor-mal and similar between conditions (Fig. 3F). These resultssupport that enforced coexpression ofMA4 and KRASG12V is notsufficient for leukemogenesis, and KRAS does not cooperatewith MA4 to enhance multilineage hematopoietic engraftmentin vivo.

Activated KRAS cooperates with MA4 to promoteextramedullary engraftment andmigration of CB-CD34þHSPC

As previously reported (36), in our cohort of MA4þ B-ALLinfants (n ¼ 24), those harboring KRAS mutations presented 2-fold higher WBC counts at diagnosis than those with germlineKRAS (Fig. 4A). Similarly, the human WBC counts in PB ofxenografts transplanted with MA4þKRAS-transduced CB-CD34þ

cells were 4- to 8-fold higher than inmice transplantedwith singleMA4- or KRAS-transduced CD34þ HSPCs (Fig. 4B). The PBleukocytosis observed in t(4;11) patients harboring KRAS muta-tions and reproduced inNSGmice xenografted withMA4þKRAS-expressing CD34þ HSPCs suggests that RAS activation may con-tribute to the maintenance rather than to the initiation of theleukemia by altering the aggressiveness and migration ability ofMA4-expressing blasts and primary CD34þ HSPCs, respectively.

Direct injection of cells within the BM cavity provides theopportunity to assess the migration of transplanted cells in vivo.Thus, the levels of extramedullary hematopoiesis of transducedCD34þ HSPCs were assessed by analyzing the chimerism in theBM, PB, spleen, and liver. Regardless of the expression ofMA4 orKRAS, CD34þHSPCswere capable ofmigrating to and colonizingPB, spleen, and liver in all analyzed animals. Surprisingly, how-ever, MA4þKRAS-transduced CD34þ cells consistently displayeda 3- to 15-fold higher level of extramedullary engraftment in PB,spleen, and liver (Fig. 4C). To gain further insights, the cell lineSEM [t(4;11)þ and germline RAS] was transduced with EV andKRASG12V, FACS-sorted, and IBM-transplanted in NSG mice (n ¼12). After 3 weeks, BM engraftment was comparable between EVand KRAS conditions, whereas PB colonization was 3-fold higherby KRASG12V-expressing SEM (Fig. 4D, left). KRAS expression inengrafted mice was confirmed by fluorescence microscopy andRT-PCR (Fig. 4D, right).

Infant t(4;11)þ B-ALL commonly associates with CNS involve-ment (37). We thus analyzed whether KRAS activation facilitatesCNS infiltration inMA4-expressing leukemia cells that were IBM-transplanted. Three weeks after IBMT, brain-skull sections frommice engrafted with SEM-EV (n ¼ 6) or SEM-KRAS (n ¼ 6) wereanalyzed by histopathology, andH&E staining showed that KRASactivation consistently (100% of mice) conferred t(4;11)þ cells arobust ability to cross blood–brain barrier, as revealed by thepresence of leukemic infiltration within leptomeningeal space,indicative of CNS involvement (Fig. 4E). In vivo data with CB-CD34þ HSPCs and SEM cell line indicate that KRAS activationpromotes extramedullary hematopoiesis and CNS infiltration,suggesting a migratory role for KRASG12V in MA4-expressing(pre)-leukemia cells. To explore this further, in vitro migratorytranswell assays were performed with SEM (Fig. 4F and G) andCD34þ HSPCs (Fig. 4F and H). KRAS-expressing SEM andMA4þKRAS-transduced CB-CD34þ HSPCs migrated toward agradient of mouse serum 4-fold and 2-fold, respectively, morethan the comparing genotypes (Fig. 4F–H). Together, our in vitroand in vivo functional data suggest that KRAS activation confers anextramedullary migratory phenotype to MA4-expressing CB-CD34þ and leukemic cells.

Global transcriptional analysis of t(4;11)þ B-ALL infantssupports the KRAS-mediated migratory role/extramedullaryhematopoiesis

To identify patterns of gene expression that could explainmolecularly the impact of KRAS activation on MA4-expressingcells, we harnessed existing GEP data generated from PB leukemic

KRAS Mutation and MLL–AF4 in Cord Blood CD34þ Cells

www.aacrjournals.org Cancer Res; 76(8) April 15, 2016 2483

on October 8, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 2, 2016; DOI: 10.1158/0008-5472.CAN-15-2769

Page 7: Activated KRAS Cooperates with MLL-AF4 to Promote ... · and previous screenings of infants with MA4þ pro–B-ALL also 1Josep Carreras Leukemia Research Institute and Department

A

Gate on CD19+ Gate on CD34+

CD45

HLA-ABC

105

102

105102

CD33

CD19

105

102

105102

HLA-

ABC

CD34

105

102

105102

CD10

CD19

105

102

105102

CD33

CD19

105

102

105102

C

E

D

B

MA4

GAPDH

KRAS 253 bp

120 bp

382 bpMA4KRASEV

MA4KRAS H2O

Engr

aftme

nt co

mpos

ition CD34+ immature

CD33+ myeloid

CD19+ lymphoid

EV MA4 KRAS MA4KRAS

B-lin

eage

comp

ositio

n

CD19+CD10+CD19+CD10-

EV MA4 KRAS MA4KRAS

MA4KRASMA4EVKRAS

Splee

n weig

ht (g

)

Liver

weig

ht (g

)

MA4KRASMA4EVKRAS

MA4KRASMA4EVKRAS

MA4KRASMA4EVKRAS

Hg (g

/dL)

Plate

lets (

×1,00

0/dL)

F

MA4KRASMA4EVKRAS

MA4KRASMA4EVKRAS

% H

emato

poiet

ic en

graft

ment

KRASEV MA4-KRASMA4Mean: 36%12% 38%20%

0

20

40

60

80

100

100

50

0

100

50

0

0.15

0.10

0.05

0

2.0

1.5

1.0

0.5

18

16

14

12

10

2,000

1,500

1,000

500

0

CD34

+fra

ction

comp

ositio

n

EV MA4 KRAS MA4KRAS

100

50

0

CD34+CD33+

CD34+CD19-CD33-

CD34+CD19+

Figure 3.KRASG12V does not cooperate withMA4 to initiate leukemia in CB-CD34þHSPCs. A, long-term hematopoietic engraftment of CB-CD34þ expressingMA4, KRASG12V,or both MA4 and KRASG12V together (n ¼ 50 mice). B, RT-PCR confirming stable ectopic expression of MA4 and KRASG12V in xenografts killed 16 weeks aftertransplantation. C, representative flow cytometry analysis of engrafted mice. The human graft identified as CD45þ HLA-ABCþ comprises CD19þ B-lymphoid cells(CD10þ pre-B and CD10� pro-B), CD33þ myeloid cells, and CD34þ immature cells. D, top, graft composition confirming normal multilineage engraftment in micetransplanted with 30,000 sorted/transduced CD34þ HSPCs expressing the indicated genes. Middle, B-cell graft demonstrating normal, nonleukemic B-celldifferentiation. B-cell graft is comprised of a majority (�80%–90%) of pre-B cells (CD34�CD19þCD10þ) and a minority (�10%–20%) of the pro-B cells(CD34þCD19þCD10�). Bottom, engraftment composition of the CD34þ fraction, including the more immature CD34þ cells (CD34þCD19�CD33�CD38�),B-cell progenitors (CD34þCD19þ), and myeloid progenitors (CD34þCD33þ). KRASG12V cooperates with MA4 to half reduce the contribution of the myeloidengraftment (n ¼ 30 mice). E, weight (top) and macroscopic images (bottom) of spleen (left) and liver (right) showing lack of splenomegaly or hepatomegalyin transplanted mice (n ¼ 50). F, hemoglobin (left) and platelet (right) levels analyzed in the indicated mice groups (n ¼ 50) revealing no sign ofleukemia in reconstituted mice.

Prieto et al.

Cancer Res; 76(8) April 15, 2016 Cancer Research2484

on October 8, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 2, 2016; DOI: 10.1158/0008-5472.CAN-15-2769

Page 8: Activated KRAS Cooperates with MLL-AF4 to Promote ... · and previous screenings of infants with MA4þ pro–B-ALL also 1Josep Carreras Leukemia Research Institute and Department

EV KRASMA4 MA4-KRAS

3x

A

Infan

t pati

ent W

BC (1

09 /L)

KRASWT KRASmut

EB

*

*B

B

B

B

B

B

20x

40x

10x

20x

40x

10x

*

SEM-KRASSEM-EV

2x

Huma

n W

BC (1

09 /L) in

xeno

graft

s

EV MA4 KRAS MA4KRAS

~4-8x

% S

EM en

graft

ment

(CD4

5+ CD1

9+ )

SEM-EVSEM-KRAS

BM PB

DC

EV MA4 KRAS MA4KRAS

EV MA4 KRAS MA4KRAS

EV MA4 KRAS MA4KRAS

~3-6x

~3-15x

~ 5-10x

% H

emato

poiet

ic en

graft

ment

(Per

iphe

ral b

lood

)

F

Ø

Numb

er of

migr

ating

cells

MA4+KRAS0 10[Serum] (%)

SEM

0 10 0 10 0 10 0 10

Numb

er of

migr

ating

cells

[Serum] (%)

CB-CD34+

~ 4X~ 2X

G

% H

emato

poiet

ic en

graft

ment

(Live

r)%

Hem

atopo

ietic

engr

aftme

nt(S

plee

n)

H

EV KRAS

KRAS

GAPDH

EV KRAS

Upper compartment(Distinct cell genotypes)

InsertPorous membrane

Lower compartment(w/wo mouse serum)

MA40 10

Ø Ø

600

400

200

0

10

8

6

4

2

0

10

12

8

6

4

2

0

60

50

40

30

20

10

0

30

20

10

0

8

6

4

2

0

4×104

3×104

2×104

1×104

0

1×105

8×104

6×104

4×104

0

2×104

Figure 4.Coexpression ofMA4 andKRASG12V promotes extramedullary engraftment and/ormigration of CB-CD34þHSPCs andMA4þ cell lines. A, KRASmutMA4þ infantswithB-ALL show 2-fold higher WBC counts at diagnosis than KRASWT group. B, approximately 4- to 8-fold higher human absolute WBC counts in PB of micexenografted with CB-CD34þ cells coexpressing both MA4 and KRASG12V. C, mice xenografted with CB-CD34þ cells coexpressing both MA4 and KRASG12V display amuch higher engraftment in extramedullary hematopoietic sites, including liver (top), spleen (middle), and PB (bottom). D, expression of KRASG12V in thecell line SEM derived from a t(4;11)(q21;q23) infant B-ALL renders an increased engraftment in (colonization of) PB but not in the BM (n ¼ 6, left). Representativefluorescence image and RT-PCR confirming KRAS expression in transduced SEM cell line (right). E, photomicrographs of brain-skull sections stained withH&E from mice engrafted with SEM-EV or SEM-KRASG12V cells showing that expression of KRASG12V confers a robust ability to infiltrate the CNS (n ¼ 6 mice).The asterisk identifies leukemia infiltrate in leptomeningeal space. White arrowhead, the skull. B, brain tissue. F, overview of the transwell assay with SEMand CB-CD34þ HSPCs. G, 4-fold enhanced migration of SEM-KRASG12V cells toward mouse PB serum as compared with SEM-EV. H, CB-CD34þ HSPCs coexpressingboth MA4 and KRASG12V migrate toward mouse PB serum twice than those expressing MA4 or KRASG12V alone.

KRAS Mutation and MLL–AF4 in Cord Blood CD34þ Cells

www.aacrjournals.org Cancer Res; 76(8) April 15, 2016 2485

on October 8, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 2, 2016; DOI: 10.1158/0008-5472.CAN-15-2769

Page 9: Activated KRAS Cooperates with MLL-AF4 to Promote ... · and previous screenings of infants with MA4þ pro–B-ALL also 1Josep Carreras Leukemia Research Institute and Department

cells from t(4;11)/MA4þ pro–B-ALL infants (GSE19475; ref. 30).Twelve infants were analyzed, of which five harbored RAS muta-tions and seven were RAS germline. Because RAS mutations areoften subclonal, only patients with >40% RAS clonality wereconsidered for GEP. A heatmap representation of hierarchicalclustering of genes differentially expressed (20% dysregulated upor down; P value < 0.05) between t(4;11)þ patients with versuswithout KRASmutations is shown in Fig. 5A. A total of 764 geneswere differentially expressed between KRAS-mutated and KRAS-germline t(4;11)þpatients.Of them, 658 (86%)were upregulatedand 106 (14%)downregulated inKRAS-mutated t(4;11) patients,suggesting that KRAS functions as a global transcriptional activa-tor in t(4;11)/MA4þ infants (Fig. 5B). To get insight into thebiologic functions affected by differentially expressed genes, weusedGorilla software (38–40) to performGOanalysis comparingKRAS-mutated versus KRAS nonmutated t(4;11)þ patients.Among the top significant GO biologic processes enriched in theKRAS-mutated upregulated genes, we found "cell migration" and"cell motility" (Fig. 5C). In contrast, among the top most signif-icant GObiologic processes enriched in theKRAS-mutated down-regulated genes, "leukocyte cell adhesion" and "leukocyte aggre-gation" pathways as well as "mesenchymal–epithelial transition"pathway were observed (Fig. 5D).

To functionally support the t(4;11)þ patients-based GEPdata, we compared in MA4- versus MA4/KRAS-expressingCD34þ HSPCs harvested from xenograft extramedullary tissues(spleen and PB) the expression of CXCR4 and MET, mastergenes involved in HSPC retention to the BM and leukocyte celladhesion aggregation, respectively, and the expression ofNELIN (41) and DCL1 (42), master regulators of cell migrationand motility. A robust downregulation of CXCR4 and METcoupled to a significant upregulation of the migration-promot-ing genes NELIN and DCL1 was observed in CB-CD34þ HSPCscoexpressing KRAS and MA4. These data support the GOsignature found in PB t(4;11)þ patient blasts, suggesting aKRAS-mediated BM exit and extramedullary hematopoiesis ofMA4-expressing CB-CD34þ and SEM cells.

DiscussionDespite tremendous improvement in clinicalmanagement and

survival of childhood B-ALL, the outcome of infants with t(4;11)/MA4þ B-ALL remains dismal, with overall survival < 30% (43).Studies in monozygotic twins and archived blood spots haveprovided compelling evidence of a prenatal cellular origin astarget forMA4 fusion, explaining the brief latency of this leukemia(1, 2, 44, 45). Despite its aggressiveness/short latency, currentprogress about its etiology, pathogenesis, and cellular origin isvery limited as evidenced by the lack of models recapitulating thedisease phenotype/latency (4–6, 46). The lack of bona fideMA4þ

B-ALL disease models may be explained from a developmentalangle because developmental cues and the prenatal nature of thetarget cell remain elusive as they have not been properlyaddressed. To understand the biology underlying the initiation ofMA4þ B-ALL, two key questions must be addressed. First, it isunclear whether t(4;11)/MA4þ alone is sufficient for overt leu-kemia or secondary cooperating oncogenic insults are required(9–11). Second, the developmentally early nature of the cell oforigin of this leukemia remains unknown (34, 39).

In sharp contrast with its dismal clinical evolution, indepen-dent recent WGSeq studies have revealed a silent mutational

landscape in MLL-r infant B-ALL (9–12), supporting the possi-bility that MA4 functions as a single oncogenic driver that sufficesto spawn aggressive B-ALL. Despite the paucity of mutationsobserved in WGSeq studies, activation of FLT-3 and/or RAS wassubclonally found in 30% to 50% of patients (9–12) and corre-lated with poor outcome (36, 47). Using CB-CD34þ HSPCs, wehave previously reported that the expression of MA4 itself or incombination with activated FLT3 is not sufficient for leukemo-genesis (7). KRAS mutations have also been associated withhigher WBC counts and poor disease evolution (36) and havebeen found subclonally present at birth in matched neonatalblood spot diagnosis samples (14). Furthermore, in a MA4þ

transgenic mouse model, activated KRAS accelerated leukemo-genesis, although the phenotype did not reproduce that seen inpatients (15). Similarly, in a humanized MLL-AF10 model, coex-pression of KRASG12Vwas required for a leukemic phenotype (16,17). However, whether RAS activation cooperates with MA4 inleukemia initiation/maintenance has not been addressed in ahuman system. Here, we successfully developed a dual-reporter–based transduction strategy to stably overexpress both MA4 andKRASG12V in CB-CD34þ HSPCs.

Regardless of MA4 expression, expression of KRAS enhancedthe hematopoietic engraftment 3-fold, indicating that KRAS andMA4 do not synergize to promote more robust engraftmentpotential. Mice follow-up, characterization of the graft composi-tion, and hematopathology analyses revealed that enforced coex-pression of MA4 and KRASG12V is not sufficient to initiate leuke-mia in vivo. Similarly, in vitro assays showed that KRASG12V doesnot cooperate with MA4 to immortalize CB-CD34þ HSPCs, asdemonstrated by the inability of KRASG12V to confer CFU replat-ing potential or overgrowth to MA4-expressing CD34þ cells.Interestingly, KRASG12V slightly impairs the clonogenic potentialof MA4-expressing CD34þ HSPCs. Although no differences weredetectable in cell cycle and apoptosis, KRAS induced in vitrosenescence of CD34þ cells and cooperated with MA4 to shift theCD34þ population toward a nonmyeloid phenotype, explainingthe KRAS-driven lower clonogenic potential of MA4-expressingCD34þ HSPCs since human CFU assays only read-out myeloidclonogenic potential.

Our data support previous WGSeq studies in MLL-r B-ALLwhere KRASmutations are subclonal and lost at relapse, suggest-ing thatMA4-mediated transformation may rely on a critical fetaltarget cell or on epigenetic cooperating lesions (12). This stablemutational landscape is not unique to infant leukemia. It has alsobeen observed in other pediatric/infant tumors, such as rhabdoidtumors (48) andbrain/hindbrain ependymomas (49, 50), furthersupporting infant cancer as a developmental disease (51). Epige-netically enhanced promoter methylation is typically accompa-nied by global loss of methylation in nonpromoter regions of thegenome of cancer cells. However, MA4þ B-ALL cells display aglobal hypermethylated genomic state, both at promoter andnonpromoter regions (52). Global hypomethylation leads togenomic instability linked to cancer development, thus explain-ing the global genomic stability/silent mutational landscapefound in MA4þ infant B-ALL. Alternatively, AF4-MLL, the recip-rocal product of MA4, has been suggested to be an oncogenicdriver in t(4;11)þ B-ALL (6), even in the absence ofMA4.WhetherAF4-MLL is oncogenic in human CD34þ cells remains to beexplored.

Importantly, however, enforced KRASG12V expression in IBM-transplantedMA4-expressing CD34þ HSPCs and MA4þ cell lines

Prieto et al.

Cancer Res; 76(8) April 15, 2016 Cancer Research2486

on October 8, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 2, 2016; DOI: 10.1158/0008-5472.CAN-15-2769

Page 10: Activated KRAS Cooperates with MLL-AF4 to Promote ... · and previous screenings of infants with MA4þ pro–B-ALL also 1Josep Carreras Leukemia Research Institute and Department

KRASwt t(4;11) pts

Enrichment score-log (

P)

-log (

P)

Enrichment score-log (P)

Cellu

lar re

spon

se to

BMP

stim

ulus

Resp

onse

to B

MPRe

gulat

ion of

tran

scrip

tion

from

RNA

Pol II

prom

oter

Posit

ive re

gulat

ion of

cartil

age d

evelo

p.BM

P sig

nalin

g pa

thway

Fore

brain

deve

lopme

ntPo

sitive

regu

lation

of os

teobla

st dif

feren

tiatio

nRe

gulat

ion of

cartil

age

deve

lopme

ntRe

gulat

ionof

osteo

blast

differ

entia

tion

Rece

ptor p

rotei

n STK

sign

aling

path

way

Cellu

lar re

spon

se to

grow

th fac

tor st

imulu

sRe

gulat

ion

of ce

llmig

ratio

n

Cellp

rojec

tion

orga

nizati

onRe

gulat

ion

of ce

llmot

ility

CUpregulated in KRAS vs WT Downregulated in KRAS vs WT

D

Enrichment score

Enrichment score-log (P)

Hair f

ollicl

e pla

code

form

ation

Mese

nchy

mal-

epith

elial

cell

signa

ling

Ectod

erma

l plac

ode f

orma

tion

Multic

ellula

r org

anism

gro

wth

Homo

typic

cell-c

ell a

dhes

ionT-

cell a

ggre

gatio

nT-

cell a

ctiva

tion

Lym

phoc

yte a

ggre

gatio

nLe

ukoc

yte

aggr

egat

ion

Leuk

ocyt

e ce

ll-ce

ll ad

hesio

nSi

ngle

orga

nisma

l cell

-cell a

dhes

ionSi

ngle

orga

nism

cell a

dhes

ionCe

ll-Ce

ll adh

esio

nPo

sitive

regu

lation

of im

mune

syste

m pr

oces

sCe

ll adh

esio

nBi

olog

ical a

dhes

ion

A 1 2 3 4 5 6 7 8 9 10 11 12

KRASmut t(4;11) pts

BColor key and histogram

040

100

Coun

t

31-1-3

658(88%)

106(14%)

UpregulatedDownregulated

UnchangedChanged

764(1.8%)

40836(98.2%)

Total=41,600 Total=764

KRASmut vs. KRAS germline t(4;11)+ ptsGene regulationDifferentially expressed genes

E CB-CD34+

0.0

0.5

1.0

1.5

Relat

iveC

XC

R4

expr

essio

n(n

orma

lized

to G

AP

DH

)

MA4+KRASmut0.0

0.5

1.0

1.5

Relat

iveM

ET

expr

essio

n(n

orma

lized

to G

AP

DH

)

0.0

0.5

1.0

1.5

Relat

iveN

ELI

N ex

pres

sion

(nor

maliz

edto

GA

PD

H)

0.0

0.5

1.0

1.5

Relat

iveD

LC1

expr

essio

n(n

orma

lized

to G

AP

DH

)

SARK+4AM4AM mut SARK+4AM4AM mutMA4 MA4+KRASmutMA4

8

6

4

2

0

30

20

10

0

6

4

2

0

120

80

40

10

5

0

Figure 5.Gene expression profiling comparing KRAS-mutated versus KRAS nonmutated MA4þ B-ALL infants. A, heatmap diagram depicting the genes differentiallyexpressed (20%up- or downregulated;P<0.01) in PBblasts from t(4:11) patientswith (n¼ 5) orwithout (n¼ 7)KRASmutations. The right color bar codifies the geneexpression level in a log2 scale. B, scheme representing the number of genes differentially expressed in KRAS-mutated versus nonmutated t(4;11)þ infants(left). A total of 764 geneswere differentially expressed, of which 658 (86%) were upregulated and 106 (14%) were downregulated in KRAS-mutated t(4;11) patients(right). C and D, statistically significant GO biologic functions identified using Gorilla software of the genes differentially upregulated (C) or downregulated(D) in KRAS-mutated versus KRAS-germline PB blasts from t(4;11)þ infant B-ALL, ranked by enrichment score. Enrichment score, black bars (right y-axis);�log P value, filled red circle with red line (left y-axis). E, qRT-PCR confirming that coexpression of both MA4 and KRASG12V induces a pronounceddownregulation of CXCR4 and MET and an upregulation of NELIN and DLC1 in CB-CD34þ HSPCs.

www.aacrjournals.org Cancer Res; 76(8) April 15, 2016 2487

KRAS Mutation and MLL–AF4 in Cord Blood CD34þ Cells

on October 8, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 2, 2016; DOI: 10.1158/0008-5472.CAN-15-2769

Page 11: Activated KRAS Cooperates with MLL-AF4 to Promote ... · and previous screenings of infants with MA4þ pro–B-ALL also 1Josep Carreras Leukemia Research Institute and Department

induced highWBC counts, robustly enhanced in vivo extramedul-lary hematopoiesis and BM exit. Similarly, KRASG12V expressionconferred t(4;11)þ cells a robust ability to cross blood–brainbarrier, inducing CNS infiltration. In vitro migratory transwellassays performed with SEM cells and CB-CD34þ HSPCs con-firmed the migratory role for KRASG12V in MA4-expressing (pre)-leukemia cells. Furthermore, global gene expression profiling andGO analysis in PB blasts from t(4;11)þ patients revealed anupregulation of a migratory-motility genes coupled to down-regulation of cell adhesion aggregation and anchoring genes,further supporting a KRAS-mediated BM exit and extramedullaryhematopoiesis of MA4-expressing CB-CD34þ and SEM cells.Elevated WBC counts and CNS involvement are hallmark phe-notypes observed in MA4þ pro–B-ALL infants (37).

Our study indicates that KRAS does not cooperate with MA4 toinitiate leukemia in CB-CD34þHSPCs but affect the homeostasisof MA4-expressing HSPCs, suggesting that KRAS activation inMA4þ B-ALL may be important in tumor maintenance/dissem-ination rather than initiation. Our data support previous WGSeqstudies inMLL-r B-ALL where KRASmutations are subclonal andlost at relapse. We propose that MA4-mediated transformationdepends on alternative epigenetic cooperating lesions and on acritical target cell in a developmentally earlier windowof stem cellvulnerability.

Disclosure of Potential Conflicts of InterestNo potential conflicts of interest were disclosed.

Authors' ContributionsConception and design: C. Prieto, C. BuenoDevelopment of methodology: C. Prieto, C. Bueno, P. MenendezAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.):C. Prieto, R.W. Stam, P. Ballerini,M. Camos, A. BursenAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): C. Prieto, A. Agraz-Doblas, J. Casta~no, R. Marschalek,I. Varela, C. Bueno, P. MenendezWriting, review, and/or revision of the manuscript: C. Prieto, C. Bueno,P. MenendezAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): C. Prieto, C. Bueno, P. MenendezStudy supervision: C. Bueno, P. Menendez

Grant SupportThis work was supported by the European Research Council (P. Menendez),

The Spanish Ministry of Economy (ISCIII/FEDER-PI14/01119 to C. Bueno;SAF2013-43065R to P. Menendez; and SAF2012-31627 to I. Varela), TheSpanish Cancer Association (P. Menendez and C. Bueno), Fundaci�on Inocente(P.Menendez), and theDeutsche Jos�eCarreras Leuk€amie Stiftung (P.Menendezand R. Marschalek). C. Bueno is supported by aMiguel Servet contract (CPII13/00011). I. Varela is supported by Ram�on Cajal Program. C. Prieto is supportedby PFIS scholarship (FI12/00468). P. Menendez also acknowledges supportfrom Obra Social La Caixa-Fundaci�o Josep Carreras and Generalitat de Cata-lunya (SGR330).

The costs of publication of this articlewere defrayed inpart by the payment ofpage charges. This article must therefore be hereby marked advertisement inaccordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Received October 13, 2015; revised December 31, 2015; accepted January 8,2016; published OnlineFirst February 2, 2016.

References1. Macmahon B, Levy MA. Prenatal origin of childhood leukemia: Evidence

from twins. New Eng J Med 1964;270:1082–5.2. Gale KB, Ford AM, Repp R, Borkhardt A, Keller C, Eden OB, et al. Back-

tracking leukemia to birth: Identification of clonotypic gene fusionsequences in neonatal blood spots. Proc Natl Acad Sci 1997;94:13950–4.

3. Pui CH.Acute lymphoblastic leukemia in children. Current Opin Oncol2000;12:3–12.

4. Metzler M, Forster A, Pannell R, Arends MJ, Daser A, Lobato MN, et al. Aconditional model of MLL-AF4 B-cell tumourigenesis using invertor tech-nology. Oncogene 2006;25:3093–103.

5. Krivtsov AV, Feng Z, Lemieux ME, Faber J, Vempati S, Sinha AU, et al.H3K79 methylation profiles define murine and human MLL-AF4 leuke-mias. Cancer Cell 2008;14:355–68.

6. Bursen A, Schwabe K, Ruster B, Henschler R, Ruthardt M, Dingermann T,et al. The AF4.MLL fusion protein is capable of inducing ALL in micewithout requirement of MLL.AF4. Blood 2010;115:3570–9.

7. Montes R, Ayllon V, Gutierrez-Aranda I, Prat I, Hernandez-Lamas MC,Ponce L, et al. Enforced expression of MLL-AF4 fusion in cord bloodCD34þ cells enhances the hematopoietic repopulating cell function andclonogenic potential but is not sufficient to initiate leukemia. Blood 2011;117:4746–58.

8. Montes R, Ayllon V, Prieto C, Bursen A, Prelle C, Romero-Moya D, et al.Ligand-independent FLT3 activation does not cooperate with MLL-AF4 toimmortalize/transform cord blood CD34þ cells. Leukemia 2014;28:666–74.

9. Liang DC, Shih LY, Fu JF, Li HY, Wang HI, Hung IJ, et al. K-Rasmutations and N-Ras mutations in childhood acute leukemias with orwithout mixed-lineage leukemia gene rearrangements. Cancer 2006;106:950–6.

10. Dobbins SE, Sherborne AL, Ma YP, Bardini M, Biondi A, Cazzaniga G,et al. The silent mutational landscape of infant MLL-AF4 pro-B acutelymphoblastic leukemia. Genes, Chromosomes & Cancer 2013;52:954–60.

11. Prelle C, Bursen A, Dingermann T, Marschalek R. Secondary mutations in t(4;11) leukemia patients. Leukemia 2013;27:1425–7.

12. Andersson AK, Ma J, Wang J, Chen X, Gedman AL, Dang J, et al. Thelandscape of somatic mutations in infant MLL-rearranged acute lympho-blastic leukemias. Nature genet 2015;47:330–7.

13. Emerenciano M, Barbosa Tda C, de Almeida Lopes B, Meyer C, MarschalekR, Pombo-de-Oliveira MS. Subclonality and prenatal origin of RAS muta-tions in KMT2A (MLL)-rearranged infant acute lymphoblastic leukaemia.Br J Haematol 2015;170:268–71.

14. Emerenciano M, Renaud G, Sant'Ana M, Barbieri C, Passetti F, Pombo-de-Oliveira MS, et al. Challenges in the use of NG2 antigen as a marker topredict MLL rearrangements in multi-center studies. Leukemia Res 2011;35:1001–7.

15. Tamai H, Miyake K, Takatori M, Miyake N, Yamaguchi H, Dan K, et al.Activated K-Ras protein accelerates human MLL/AF4-induced leukemo-lymphomogenicity in a transgenic mouse model. Leukemia 2011;25:888–91.

16. Ono R, Kumagai H, Nakajima H, Hishiya A, Taki T, Horikawa K, et al.Mixed-lineage-leukemia (MLL) fusion protein collaborates with Ras toinduce acute leukemia through aberrantHox expression andRaf activation.Leukemia 2009;23:2197–209.

17. Moriya K, Suzuki M, Watanabe Y, Takahashi T, Aoki Y, Uchiyama T, et al.Development of a multi-step leukemogenesis model of MLL-rearrangedleukemia using humanized mice. PloS One 2012;7:e37892.

18. Bueno C, Catalina P, Melen GJ, Montes R, Sanchez L, Ligero G, et al.Etoposide induces MLL rearrangements and other chromosomal abnor-malities in human embryonic stem cells. Carcinogenesis 2009;30:1628–37.

19. Bueno C, Montes R, Martin L, Prat I, Hernandez MC, Orfao A, et al. NG2antigen is expressed in CD34þ HPCs and plasmacytoid dendritic cellprecursors: is NG2 expression in leukemia dependent on the target cellwhere leukemogenesis is triggered?Leukemia 2008;22:1475–8.

20. Real PJ, Ligero G, Ayllon V, Ramos-Mejia V, Bueno C, Gutierrez-Aranda I,et al. SCL/TAL1 regulates hematopoietic specification from human embry-onic stem cells. Mol ther 2012;20:1443–53.

21. Menendez P, Wang L, Chadwick K, Li L, Bhatia M. Retroviral transduc-tion of hematopoietic cells differentiated from human embryonic stem

Cancer Res; 76(8) April 15, 2016 Cancer Research2488

Prieto et al.

on October 8, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 2, 2016; DOI: 10.1158/0008-5472.CAN-15-2769

Page 12: Activated KRAS Cooperates with MLL-AF4 to Promote ... · and previous screenings of infants with MA4þ pro–B-ALL also 1Josep Carreras Leukemia Research Institute and Department

cell-derived CD45(neg)PFV hemogenic precursors. Mol ther 2004;10:1109–20.

22. Navarro-Montero O, Romero-Moya D, Montes R, Ramos-Mejia V, BuenoC, Real PJ, et al. Intrahepatic transplantation of cord blood CD34þ cellsinto newborn NOD/SCID-IL2Rgamma(null) mice allows efficient multi-organ and multi-lineage hematopoietic engraftment without accessorycells. Clinical Immunol 2012;145:89–91.

23. Bueno C, Montes R, de la Cueva T, Gutierrez-Aranda I, Menendez P. Intra-bone marrow transplantation of human CD34(þ) cells into NOD/LtSz-scid IL-2rgamma(null) mice permits multilineage engraftment withoutprevious irradiation. Cytotherapy 2010;12:45–9.

24. Levac K, Menendez P, Bhatia M. Intra-bone marrow transplantation facil-itates pauci-clonal human hematopoietic repopulation of NOD/SCID/beta2m(-/-) mice. Exp Hematol 2005;33:1417–26.

25. Romero-MoyaD,BuenoC,MontesR,Navarro-MonteroO, Iborra FJ, LopezLC, et al. Cord blood-derived CD34þ hematopoietic cells with lowmitochondrial mass are enriched in hematopoietic repopulating stem cellfunction. Haematologica 2013;98:1022–9.

26. Rodriguez R, Rubio R, Masip M, Catalina P, Nieto A, de la Cueva T, et al.Loss of p53 induces tumorigenesis in p21-deficient mesenchymal stemcells. Neoplasia 2009;11:397–407.

27. Ramos-Mejia V, Melen GJ, Sanchez L, Gutierrez-Aranda I, Ligero G, CortesJL, et al. Nodal/Activin signaling predicts humanpluripotent stem cell linesprone to differentiate toward the hematopoietic lineage. Mol Ther2010;18:2173–81.

28. Prosper F, Sola C, Hornedo J, Arbona C, Menendez P, Orfao A, et al.Mobilization of peripheral blood progenitor cells with a combinationof cyclophosphamide, r-metHuSCF and filgrastim in patients withbreast cancer previously treated with chemotherapy. Leukemia 2003;17:437–41.

29. Buonamici S, Trimarchi T, Ruocco MG, Reavie L, Cathelin S, Mar BG, et al.CCR7 signalling as an essential regulator of CNS infiltration in T-cellleukaemia. Nature 2009;459:1000–4.

30. Stam RW, Schneider P, Hagelstein JA, van der Linden MH, Stumpel DJ, deMenezes RX, et al. Gene expression profiling-based dissection of MLLtranslocated and MLL germline acute lymphoblastic leukemia in infants.Blood 2010;115:2835–44.

31. Bueno C, Montes R, Melen GJ, Ramos-Mejia V, Real PJ, Ayllon V, et al. Ahuman ESCmodel for MLL-AF4 leukemic fusion gene reveals an impairedearly hematopoietic-endothelial specification. Cell Res 2012;22:986–1002.

32. Rodriguez R, Rubio R, Gutierrez-Aranda I, Melen GJ, Elosua C, Garcia-Castro J, et al. Fus-Chop fusion protein expression coupled to P53 defi-ciency induces liposarcoma in mouse but not human adipose-derivedmesenchymal stem/stromal cells. Stem Cells 2011;29:179–92.

33. Rubio R, Gutierrez-Aranda I, Saez-Castillo AI, Labarga A, Rosu-Myles M,Gonzalez-Garcia S, et al. The differentiation stage of p53-Rb-deficient bonemarrow mesenchymal stem cells imposes the phenotype of in vivo sar-coma development.Oncogene 2013;32:4970–80.

34. Bueno C, Montes R, Catalina P, Rodriguez R, Menendez P. Insightsinto the cellular origin and etiology of the infant pro-B acute lym-phoblastic leukemia with MLL-AF4 rearrangement. Leukemia 2011;25:400–10.

35. French A, Yang CT, Taylor S, Watt SM, Carpenter L. Human inducedpluripotent stem cell-derived B lymphocytes express sIgM and can be

generated via a hemogenic endothelium intermediate. Stem Cells Dev2015;24:1082–95.

36. DriessenEM, vanRoonEH, Spijkers-Hagelstein JA, Schneider P, de LorenzoP, ValsecchiMG, et al. Frequencies andprognostic impact of RASmutationsin MLL-rearranged acute lymphoblastic leukemia in infants. Haematolo-gica 2013;98:937–44.

37. Pui CH, Mullighan CG, Evans WE, Relling MV. Pediatric acute lympho-blastic leukemia: where are we going and how do we get there?Blood2012;120:1165–74.

38. Rodriguez R, Rubio R, Gutierrez-Aranda I, Melen GJ, Elosua C, Garcia-Castro J, et al. FUS-CHOP fusion protein expression coupled to p53deficiency induces liposarcoma in mouse but not in human adipose-derived mesenchymal stem/stromal cells. Stem Cells 2011;29:179–92.

39. Bueno C, Montes R, Melen GJ, Ramos-Mejia V, Real PJ, Ayllon V, et al. Ahuman ESC model for MLL-AF4 leukemic fusion gene reveals animpaired early hematopoietic-endothelial specification. Cell Res 2012;22:986–1002.

40. Rubio R, Gutierrez-Aranda I, Saez-Castillo AI, Labarga A, Rosu-Myles M,Gonzalez-Garcia S, et al. The differentiation stage of p53-Rb-deficient bonemarrow mesenchymal stem cells imposes the phenotype of in vivo sar-coma development. Oncogene 2013;32:4970–80.

41. WangW, ZhangW, Han Y, Chen J, Wang Y, Zhang Z, et al. NELIN, a new F-actin associated protein, stimulates HeLa cell migration and adhesion.Biochem Biophys Res Commun 2005;330:1127–31.

42. Barras D,Widmann C. GAP-independent functions of DLC1 inmetastasis.Cancer Metastasis Rev 2014;33:87–100.

43. Biondi A, Cimino G, Pieters R, Pui CH. Biological and therapeutic aspectsof infant leukemia. Blood 2000;96:24–33.

44. Greaves MF, Maia AT, Wiemels JL, Ford AM. Leukemia in twins: Lessons innatural history. Blood 2003;102:2321–33.

45. Greaves MF, Wiemels J. Origins of chromosome translocations in child-hood leukaemia. Nat Rev Cancer 2003;3:639–49.

46. ChenW, Li Q,HudsonWA, Kumar A, Kirchhof N, Kersey JH. AmurineMll-AF4 knock-in model results in lymphoid and myeloid deregulation andhematologic malignancy. Blood 2006;108:669–77.

47. Chillon MC, Gomez-Casares MT, Lopez-Jorge CE, Rodriguez-Medina C,Molines A, SarasqueteME, et al. Prognostic significance of FLT3mutationalstatus and expression levels in MLL-AF4þ and MLL-germline acute lym-phoblastic leukemia. Leukemia 2012;26:2360–6.

48. Lee RS, Stewart C, Carter SL, Ambrogio L, Cibulskis K, Sougnez C, et al. Aremarkably simple genome underlies highlymalignant pediatric rhabdoidcancers. J Clin Invest 2012;122:2983–8.

49. Parker M, Mohankumar KM, Punchihewa C, Weinlich R, Dalton JD, Li Y,et al. C11orf95-RELA fusions drive oncogenic NF-kappaB signalling inependymoma. Nature 2014;506:451–5.

50. Mack SC,Witt H, Piro RM,Gu L, Zuyderduyn S, Stutz AM, et al. Epigenomicalterations define lethal CIMP-positive ependymomas of infancy. Nature2014;506:445–50.

51. Sanjuan-Pla A, Bueno C, Prieto C, Acha P, Stam RW, Marschalek R,Menendez P�. Revisiting the biology of infant t(4;11)/MLL-AF4þ B-cellacute lymphoblastic leukemia. Blood 2015;126:2676–85.

52. Stumpel DJ, Schneider P, van Roon EH, Pieters R, Stam RW. Absence ofglobal hypomethylation in promoter hypermethylated mixed lineageleukaemia-rearranged infant acute lymphoblastic leukaemia. Eur J Cancer2013;49:175–84.

www.aacrjournals.org Cancer Res; 76(8) April 15, 2016 2489

KRAS Mutation and MLL–AF4 in Cord Blood CD34þ Cells

on October 8, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 2, 2016; DOI: 10.1158/0008-5472.CAN-15-2769

Page 13: Activated KRAS Cooperates with MLL-AF4 to Promote ... · and previous screenings of infants with MA4þ pro–B-ALL also 1Josep Carreras Leukemia Research Institute and Department

2016;76:2478-2489. Published OnlineFirst February 2, 2016.Cancer Res   Cristina Prieto, Ronald W. Stam, Antonio Agraz-Doblas, et al.   HSPC But Is Insufficient to Initiate Leukemia

+Extramedullary Engraftment and Migration of Cord Blood CD34 Cooperates with MLL-AF4 to PromoteKRASActivated

  Updated version

  10.1158/0008-5472.CAN-15-2769doi:

Access the most recent version of this article at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/76/8/2478.full#ref-list-1

This article cites 52 articles, 11 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/76/8/2478.full#related-urls

This article has been cited by 7 HighWire-hosted articles. Access the articles at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected]

To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://cancerres.aacrjournals.org/content/76/8/2478To request permission to re-use all or part of this article, use this link

on October 8, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 2, 2016; DOI: 10.1158/0008-5472.CAN-15-2769