hoxa9 cooperates with activated jak/stat signaling to …...2018/04/09  · expression in 3 of 5...

17
OF1 | CANCER DISCOVERY MAY 2018 www.aacrjournals.org HOXA9 Cooperates with Activated JAK/STAT Signaling to Drive Leukemia Development Charles E. de Bock 1,2 , Sofie Demeyer 1,2 , Sandrine Degryse 1,2 , Delphine Verbeke 1,2 , Bram Sweron 1,2 , Olga Gielen 1,2 , Roel Vandepoel 1,2 , Carmen Vicente 1,2 , Marlies Vanden Bempt 1,2 , Antonis Dagklis 1,2 , Ellen Geerdens 1,2 , Simon Bornschein 1,2 , Rik Gijsbers 3 , Jean Soulier 4 , Jules P. Meijerink 5 , Merja Heinäniemi 6 , Susanna Teppo 7 , Maria Bouvy-Liivrand 6 , Olli Lohi 7 , Enrico Radaelli 1,2 , and Jan Cools 1,2 RESEARCH ARTICLE ABSTRACT Leukemia is caused by the accumulation of multiple genomic lesions in hemato- poietic precursor cells. However, how these events cooperate during oncogenic transformation remains poorly understood. We studied the cooperation between activated JAK3/ STAT5 signaling and HOXA9 overexpression, two events identified as significantly co-occurring in T-cell acute lymphoblastic leukemia. Expression of mutant JAK3 and HOXA9 led to a rapid development of leukemia originating from multipotent or lymphoid-committed progenitors, with a significant decrease in disease latency compared with JAK3 or HOXA9 alone. Integrated RNA sequencing, chromatin immu- noprecipitation sequencing, and Assay for Transposase-Accessible Chromatin using sequencing (ATAC- seq) revealed that STAT5 and HOXA9 have co-occupancy across the genome, resulting in enhanced STAT5 transcriptional activity and ectopic activation of FOS/JUN (AP1). Our data suggest that onco- genic transcription factors such as HOXA9 provide a fertile ground for specific signaling pathways to thrive, explaining why JAK/STAT pathway mutations accumulate in HOXA9-expressing cells. SIGNIFICANCE: The mechanism of oncogene cooperation in cancer development remains poorly char- acterized. In this study, we model the cooperation between activated JAK/STAT signaling and ectopic HOXA9 expression during T-cell leukemia development. We identify a direct cooperation between STAT5 and HOXA9 at the transcriptional level and identify PIM1 kinase as a possible drug target in mutant JAK/STAT/HOXA9-positive leukemia cases. Cancer Discov; 8(5); 1–16. ©2018 AACR. 1 KU Leuven, Center for Human Genetics, Leuven, Belgium. 2 VIB, Center for Cancer Biology, Leuven, Belgium. 3 Laboratory for Viral Vector Technol- ogy and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium. 4 U944 INSERM and Hematology laboratory, St-Louis Hospital, APHP, Hematology University Institute, Uni- versity Paris-Diderot, Paris, France. 5 Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands. 6 Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland. 7 Tampere Centre for Child Health Research, University of Tampere and Tampere University Hospital, Tampere, Finland. Note: Supplementary data for this article are available at Cancer Discovery Online (http://cancerdiscovery.aacrjournals.org/). Corresponding Author: Jan Cools, Laboratory for Molecular Biology of Leukemia, VIB-KU Leuven Center for Cancer Biology, KU Leuven Cam- pus Gasthuisberg O&N 4, 3000 Leuven, Belgium. Phone: 32-16-33-00-82; E-mail: [email protected] doi: 10.1158/2159-8290.CD-17-0583 ©2018 American Association for Cancer Research. Research. on January 15, 2021. © 2018 American Association for Cancer cancerdiscovery.aacrjournals.org Downloaded from Published OnlineFirst March 1, 2018; DOI: 10.1158/2159-8290.CD-17-0583

Upload: others

Post on 22-Sep-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: HOXA9 Cooperates with Activated JAK/STAT Signaling to …...2018/04/09  · expression in 3 of 5 JAK3-mutant cases, that was up to 10-fold higher when compared with HOXA10 and HOXA11

OF1 | CANCER DISCOVERY MAY 2018 www.aacrjournals.org

HOXA9 Cooperates with Activated JAK/STAT Signaling to Drive Leukemia Development Charles E. de Bock1,2, Sofie Demeyer1,2, Sandrine Degryse1,2, Delphine Verbeke1,2, Bram Sweron1,2, Olga Gielen1,2, Roel Vandepoel1,2, Carmen Vicente1,2, Marlies Vanden Bempt1,2, Antonis Dagklis1,2, Ellen Geerdens1,2, Simon Bornschein1,2, Rik Gijsbers3, Jean Soulier4, Jules P. Meijerink5, Merja Heinäniemi6, Susanna Teppo7, Maria Bouvy-Liivrand6, Olli Lohi7, Enrico Radaelli1,2, and Jan Cools1,2

ReseaRch aRticle

abstRact Leukemia is caused by the accumulation of multiple genomic lesions in hemato­poietic precursor cells. However, how these events cooperate during oncogenic

transformation remains poorly understood. We studied the cooperation between activated JAK3/STAT5 signaling and HOXA9 overexpression, two events identified as significantly co­occurring in T­cell acute lymphoblastic leukemia. Expression of mutant JAK3 and HOXA9 led to a rapid development of leukemia originating from multipotent or lymphoid­committed progenitors, with a significant decrease in disease latency compared with JAK3 or HOXA9 alone. Integrated RNA sequencing, chromatin immu­noprecipitation sequencing, and Assay for Transposase­Accessible Chromatin using sequencing (ATAC­seq) revealed that STAT5 and HOXA9 have co­occupancy across the genome, resulting in enhanced STAT5 transcriptional activity and ectopic activation of FOS/JUN (AP1). Our data suggest that onco­genic transcription factors such as HOXA9 provide a fertile ground for specific signaling pathways to thrive, explaining why JAK/STAT pathway mutations accumulate in HOXA9­expressing cells.

SIGNIFICANCE: The mechanism of oncogene cooperation in cancer development remains poorly char­acterized. In this study, we model the cooperation between activated JAK/STAT signaling and ectopic HOXA9 expression during T­cell leukemia development. We identify a direct cooperation between STAT5 and HOXA9 at the transcriptional level and identify PIM1 kinase as a possible drug target in mutant JAK/STAT/HOXA9­positive leukemia cases. Cancer Discov; 8(5); 1–16. ©2018 AACR.

1KU Leuven, Center for Human Genetics, Leuven, Belgium. 2VIB, Center for Cancer Biology, Leuven, Belgium. 3Laboratory for Viral Vector Technol­ogy and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium. 4U944 INSERM and Hematology laboratory, St­Louis Hospital, APHP, Hematology University Institute, Uni­versity Paris­Diderot, Paris, France. 5Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands. 6Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland. 7Tampere Centre for Child Health Research, University of Tampere and Tampere University Hospital, Tampere, Finland.

Note: Supplementary data for this article are available at Cancer Discovery Online (http://cancerdiscovery.aacrjournals.org/).Corresponding Author: Jan Cools, Laboratory for Molecular Biology of Leukemia, VIB­KU Leuven Center for Cancer Biology, KU Leuven Cam­pus Gasthuisberg O&N 4, 3000 Leuven, Belgium. Phone: 32­16­33­00­82; E­mail: [email protected]: 10.1158/2159­8290.CD­17­0583©2018 American Association for Cancer Research.

Research. on January 15, 2021. © 2018 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst March 1, 2018; DOI: 10.1158/2159-8290.CD-17-0583

Page 2: HOXA9 Cooperates with Activated JAK/STAT Signaling to …...2018/04/09  · expression in 3 of 5 JAK3-mutant cases, that was up to 10-fold higher when compared with HOXA10 and HOXA11

MAY 2018 CANCER DISCOVERY | OF2

iNtRODUctiON

T-cell acute lymphoblastic leukemia (T-ALL) is an aggres-sive hematologic disease, which arises from the malignant transformation of developing T-cell progenitors. The best- characterized genetic defects in T-ALL include inactivation of cell-cycle regulators (p16/p15), overexpression of tran-scription factors (TLX1, TLX3, TAL1, HOXA), mutations that activate the NOTCH1 and PI3K signaling cascades, and hyperactivation of signaling pathways by mutant kinases such as JAK3 mutants or the NUP214–ABL1 fusion (1–3). In T-ALL, there are an average of 10 to 20 damaging genomic lesions per case (4–6). These mutations target critical cel-lular pathways including lymphoid development, cell-cycle regulation, tumor suppression, lymphoid signaling, and drug responsiveness (6, 7). Recent studies using next-generation sequencing have identified mutations in the IL7R/JAK3/STAT5 pathway in 28% of T-ALL cases with JAK3 tyrosine kinase mutations being the most frequent mutations found in up to 16% of T-ALL cases (4, 5, 8, 9).

In normal T-cell biology, the JAK3 and JAK1 kinases are essential components of the heterodimeric IL7 receptor com-plex. JAK3 binds the common gamma chain (IL2RG), and JAK1 binds the IL7Rα chain (10). When the receptor complex

is bound by its ligand IL7, there is phosphorylation of both JAK1 and JAK3. This leads to the recruitment and phospho-rylation of STAT5, which then translocates to the cell nucleus to regulate gene expression. The binding of IL7 to its receptor is essential for the survival and proliferation of double-neg-ative thymocytes and is also important in the homeostasis, differentiation, and activity of mature T cells (11, 12). The JAK3 mutations found in patients with T-ALL are able to cir-cumvent this tightly controlled signaling process and activate STAT5 in the absence of cytokine, transform cells in vitro, and drive the development of T-ALL in vivo using a murine bone marrow transplant leukemia model (13).

Although the validation of JAK3 mutations as bona fide “driver” mutations using a one-oncogene murine model is significant, from a clinical perspective, patients have addi-tional mutations that may cooperate for malignant cell trans-formation. For example, it has been shown that BCR–ABL and NUP98–HOXA9 can cooperate synergistically in acute myeloid leukemia, and modeling this cooperation identified compounds that selectively target leukemic stem cells (14, 15). More recently, the existence of multiple “mini-driver” mutations has also been suggested that might substitute for a major driver change (16), and therefore given the high num-ber of genetic changes within T-ALL, it is likely that leukemia

Research. on January 15, 2021. © 2018 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst March 1, 2018; DOI: 10.1158/2159-8290.CD-17-0583

Page 3: HOXA9 Cooperates with Activated JAK/STAT Signaling to …...2018/04/09  · expression in 3 of 5 JAK3-mutant cases, that was up to 10-fold higher when compared with HOXA10 and HOXA11

de Bock et al.RESEARCH ARTICLE

OF3 | CANCER DISCOVERY MAY 2018 www.aacrjournals.org

development and progression require synergistic modulation of downstream signaling pathways by cooperating oncogenic lesions in a polygenic manner.

Some cooperating lesions in T-ALL have already been iden-tified and include Arf inactivation in combination with Lmo2 overexpression significantly accelerating the development of the T-cell malignancies and acquisition of Notch1 mutations (17). Similar work in our laboratory found that loss of the phosphatase PTPN2 enhanced the kinase activity of the NUP214–ABL1 fusion or JAK1 mutants, thereby sensitiz-ing cells to leukemogenic transformation (18, 19). However, the vast majority of co-occurring lesions and their ability to cooperate in driving tumorigenesis still require functional validation.

In the current work, we set about identifying cooperating oncogenes in the context of a JAK3 mutant–driven leukemia and functionally validating these in vivo using a two- oncogene model. We found that T-ALL cases with JAK3 mutations are often HOXA+, specifically expressing high levels of HOXA9, and together can drive an aggressive leukemia using mouse models.

ResUltsJAK3 Mutations Are Significantly Associated with HOXA+ T-ALL Cases

Based on results of large-scale sequencing studies in T-ALL (4, 5, 8, 9, 20), it is becoming clear that some mutations frequently co-occur, whereas others are mutually exclusive. We analyzed genetic data for 155 T-ALL cases (Vicente data-set; ref. 5) in which IL7R/JAK/STAT5 mutations occur in 28% of the cases and are found more frequently in HOXA+, immature, and TLX1/3-positive cases. Focusing specifically on JAK3 mutations alone, these account for the most fre-quent type of mutation found in 16% of the cases (5). Upon analyzing different T-ALL subgroups, these JAK3 muta-tions were found to significantly associate with those T-ALL cases designated as HOXA+ (P value = 0.018) and negatively associate with those designated as TAL1/LMO2–positive (P value = 0.002; Fig. 1B). The designation of HOXA+ included cases with chromosomal rearrangements known to either directly or indirectly upregulate genes within the HOXA cluster (i.e., CALM-AF10, SET-NUP214, NUP98-RAP1GDS, and TCRB-HOXA; refs. 5, 21, 22). Analysis of a second independ-ent cohort of T-ALL cases (Liu dataset; ref. 9) showed 22% of the patients had IL7R/JAK/STAT5 mutations (data included mutations within JAK1, JAK3, IL7R, PTPN2, FLT3, STAT5B, and SH2B3; Fig. 1A). These mutations were enriched within the HOXA+, TLX1/3+, and LMO2/LYL1+ cases. Furthermore, focusing specifically on JAK3-mutant cases again revealed a significant positive association with HOXA+ and negative association with TAL1/TAL2/LMO2 cases (Fig. 1B). Moreo-ver, associated gene expression data showed that HOXA+ cases have overall higher JAK3 expression levels compared with the majority of other T-ALL cases (Fig. 1C). Activation of the HOXA cluster in T-ALL typically results in upregula-tion of both HOXA9 and HOXA10. Analysis of primary T-ALL samples (Supplementary Table S1) revealed strong HOXA9 expression in 3 of 5 JAK3-mutant cases, that was up to 10-fold higher when compared with HOXA10 and HOXA11

(Fig. 1D), suggesting that HOXA9 is the most important upregulated gene of the HOXA cluster in T-ALL.

JAK3 Mutations and HOXA9 Coexpression Transform Hematopoietic Stem and Progenitor Cells and Lead to Rapid Leukemia Development In Vivo Using a Bone Marrow Transplant Model

The most common JAK3 mutation found within T-ALL cases is the M511I mutation just upstream of the pseu-dokinase domain (5, 9, 13). Therefore, we next sought to determine whether mutant JAK3 signaling (using the JAK3M511I mutant) can cooperate with HOXA9 in the trans-formation of hematopoietic stem progenitor cells (HSPC) and leukemia development. Two separate retroviral vectors containing JAK3M511I (also expressing GFP) or HOXA9 (also expressing mCHERRY) were used for the cotransduc-tion of murine HSPCs ex vivo and yielded a mixture of nontransduced, single-transduced, or double-transduced cells as assessed by GFP and mCHERRY fluorescence (Fig. 2A). HSPCs transduced with both JAK3M511I and HOXA9 rapidly transformed to cytokine-independent growth and expanded over 20 days in the absence of all cytokines. All nontransduced and single-transduced cells either were outcompeted and/or did not grow (Fig. 2B). The vari-ous JAK3 mutations activate downstream STAT5 signaling with the majority also requiring binding to a functional cytokine receptor complex (13, 23). Constitutively active STAT5BN642H mutations have also been identified and func-tionally characterized in T-ALL (24, 25). Therefore, the same experimental setup was repeated using STAT5BN642H and HOXA9 retroviral expression constructs. Again, only the double-transduced STAT5BN642H and HOXA9 cells expanded in the absence of all cytokines, phenocopying the JAK3M511I and HOXA9 result (Fig. 2C). Taken together, these results show that oncogenic JAK3/STAT5 activation is necessary and sufficient for cooperation with HOXA9 in ex vivo HSPC cell transformation.

Given this evident cooperation in HSPC transforma-tion ex vivo, we next investigated the cooperation between JAK3M511I and HOXA9 during in vivo leukemia development. Mouse HSPCs were isolated and transduced with retroviral vectors containing JAK3M511I or HOXA9 or a mixture of both and then injected into sublethally irradiated synge-neic recipient mice (BALB/c or C57BL/6). JAK3M511I+HOXA9 recipient BALB/c mice rapidly developed a leukemia char-acterized by a rapid increase in peripheral white blood cell (WBC) counts in excess of 10,000 cells/mm3 within 30 days compared with JAK3M511I only (Fig. 2D). For each recipi-ent, the double transduced JAK3M511I and HOXA9 leukemic clone outcompeted all single transduced clones (Fig. 2E). This rapid leukemia development resulted in significantly decreased disease-free survival (DFS; median DFS = 25 days, P value < 0.0001) when compared with JAK3M511I alone (median DFS = 126.5 days; Fig. 2F). In contrast, clones coexpressing TAL1 and JAK3M511I were counter-selected in vivo with mice succumbing to a JAK3M511I-only disease (Fig.  2F; Supplementary Fig. S1) and reconciled with JAK3 mutations negatively associating with TAL1 in T-ALL (Fig. 1A). Notably, when the same experimental approach was used for HOXA10 together with JAK3M511, there was only a

Research. on January 15, 2021. © 2018 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst March 1, 2018; DOI: 10.1158/2159-8290.CD-17-0583

Page 4: HOXA9 Cooperates with Activated JAK/STAT Signaling to …...2018/04/09  · expression in 3 of 5 JAK3-mutant cases, that was up to 10-fold higher when compared with HOXA10 and HOXA11

HOXA9 and Mutant JAK3 Cooperation in Leukemia RESEARCH ARTICLE

MAY 2018 CANCER DISCOVERY | OF4

Figure 1.  JAK3 mutations and ectopic expression of HOXA9 are significantly associated in clinical T­ALL cases. A, Frequency of IL7R/JAK/STAT mutations in 155 T­ALL cases (Vicente dataset) and 264 T­ALL cases (Liu dataset) within different defined transcription factor subgroups. Dotted line denotes average percentage of IL7R/JAK/STAT mutations across entire dataset. B, Association between JAK3 mutations only and defined transcription factor subgroups within the Vicente and Liu datasets. C, RNA­seq FPKM levels for JAK3 and HOXA9 in the Liu dataset. D, Real­time qPCR of primary T­ALL samples for HOXA9, HOXA10, and HOXA11.

JAK3 wild-type(n = 130)

JAK3 mutant(n = 25)

HOXA+ P = 0.018TAL1/LMO2+ P = 0.002

HOXA+

TLX1/3+

TAL1/LMO2+

12%15%

32%

24%36%

32%41%

8%

XB

37X

12X

B47

X15

X17

XB

41X

13X

10X

B34

X09

XA

32X

C57

X16

1Q94

839U

9H60

863I

O62

3X

117Q

0538

9E38

2L

0

2

4

6

8

10

50

100

150

CN

RQ

Mut JAK3

HOXA10

HOXA9

HOXA11

Other

A B

0 50 100

TAL1+

(n = 54)

TLX1/TLX3+

(n = 41)

HOXA+

(n = 10)

Immature (n = 15)

IL7R/JAK/STAT5 pathway mutation (Vicente dataset)

Percentage

28

0 22 50 100

NKX2_1(n = 14)

TAL1/TAL2(n = 95)

TLX1/TLX3+

(n = 72)a

HOXA+

(n = 33)

LMO1/2(n = 10)

LMO2_LYL1(n = 18)

IL7R–JAK–STAT5 pathway mutation (Liu dataset)

Percentage

HOXA+TLX1/3

LMO2LYL1

TAL1/TAL2/LMO2

Other

JAK3 wild-type(n = 244)

JAK3 mutant(n = 20)

HOXA+ P = 0.006TAL1/TAL2/LMO2+ P = 0.0047

C D

HO

XA

LMO

2_LY

L1

NK

X2_

1

TLX

1

TLX

3

Unk

now

n

TA

L1/2

LMO

1/2

0

20

40

60

0

20

40

60

80

RN

A-s

eq F

PK

M

RN

A-s

eq F

PK

M

HOXA9 JAK3

HO

XA

LMO

2_LY

L1

NK

X2_

1

TLX

1

TLX

3

Unk

now

n

TA

L1/2

LMO

1/2

27%

11%

13%

7%

35%25%

10%

20%42%10%

HOXA+Other

TLX1/3+

TAL1/LMO2+

HOXA+

TLX1/3

LMO2LYL1

TAL1/TAL2/LMO2

Other

Research. on January 15, 2021. © 2018 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst March 1, 2018; DOI: 10.1158/2159-8290.CD-17-0583

Page 5: HOXA9 Cooperates with Activated JAK/STAT Signaling to …...2018/04/09  · expression in 3 of 5 JAK3-mutant cases, that was up to 10-fold higher when compared with HOXA10 and HOXA11

de Bock et al.RESEARCH ARTICLE

OF5 | CANCER DISCOVERY MAY 2018 www.aacrjournals.org

Figure 2.  Coexpression of JAK3M511I and HOXA9 transforms hematopoietic progenitor cells and leads to rapid leukemia development in vivo. A, Schematic of the two retroviral expression vectors used in the transduction of murine HSPCs for the ex vivo transformation assay and bone marrow transplant model. Dual transduction results in single, double, and nontransduced cells for direct clonal competition analyses ex vivo and in vivo. B, Cell proliferation of HSPCs transduced with both JAK3M511I and HOXA9 over 20 days in the absence of all cytokines with matching FACS profiles. C, Cell pro­liferation of HSPCs transduced with both STAT5N642H and HOXA9 over 20 days in the absence all cytokines with matching FACS profiles. D, WBC counts in recipient BALB/C mice that received lin– cells transduced with JAK3M511I and HOXA9 retrovirus or JAK3M511I­only retrovirus. E, Clonal analysis by FACS for GFP and mCHERRY reporters representing JAK3M511I and HOXA9, respectively, at injection start and end stages. F, Kaplan–Meier DFS for JAK3M511I/HOXA9 mice (median DFS = 25 days, P value < 0.0001), JAK3M511I alone (median DFS = 126.5 days), and JAK3M511I/TAL1 (median DFS = 144.5 days). DFS determined by WBC <30,000 cells/mm3. G and H, Reciprocal experiments in C57BL/6 mice coexpressing JAK3M511I and HOXA9 for WBC and clonal analysis by FACS for GFP and mCHERRY reporters representing JAK3M511I and HOXA9 respectively at injection start and end stages. I, Kaplan–Meier DFS for STAT5BN642H/HOXA9 (median DFS = 12 days), JAK3M511I/HOXA9 (median DFS = 40 days), JAK3M511I (median DFS = 150 days), and HOXA9 only (median DFS = 182 days) recipient C57BL/6 mice.

Retroviral spinfectionof Lin− cells

5′LTR 3′LTRJAK3 (M511I) IRES GFP

5′LTR 3′LTRHOXA9 IRES mCHERRY

Tail vein injectionsublethally irradiated mice

(BALB/c; C57BL/6)

WB

C (

cells

/mm

3 )

HO

XA

9

JAK3 (M511I)

Days post-transplantSta

rtEnd

0

20

40

60

80

100

JAK3

HOXA9

JAK3 (M511I)/HOXA9

JAK3 (M511I)

JAK3 (M511I)/HOXA9

JAK3 (M511I)/TAL1*

Dis

ease

-fre

e su

rviv

al (

%)

Days post-transplant

Flu

orop

hore

(%

)

0

20

40

60

80

100

JAK3

HOXA9

JAK3 (M511I)/HOXA9

Flu

orop

hore

(%

)

0 100 200 3000

50,000

100,000

150,000

200,000200,000

300,000

400,000

Days post-transplant

WB

C (

cells

/mm

3 )

JAK3 (M511I)

HOXA9

JAK3 (M511I)/HOXA9

STAT5B (N642H)/ HOXA9

Dis

ease

-fre

e su

rviv

al (

%)

0 100 200 300

Days post-transplant

0

50

100JAK3 (M511I) (n = 5)

HOXA9 (n = 5)

JAK3 (M511I)/HOXA9 (n = 9)

P < 0.0001

***

0 50 100 150 200 2500

20,000

40,000

60,000

80,000

100,000100,000

200,000

300,000

400,000

500,000

600,000JAK3 (M511I) (n = 10)JAK3 (M511I)/HOXA9 (n = 9)

BA

LB/c

C57

BL/

6

0 100 200 3000

50

100

Days post-transduction

Day 1

HO

XA

9-IR

ES

-mC

HE

RR

Y

JAK3 (M511I)-IRES-GFP

HO

XA

9-IR

ES

-mC

HE

RR

Y

STAT5B (N642H)-IRES-GFP

19 25

45 11

14 53

10 22

3 89

0.14 8

17 37

33 13

21 61

5 13

3 92

0.35 4

0 5 10 15 20 25

HOXA9JAK3 (M511I)

JAK3 (M511I) + HOXA9

Nontransduced

0 5 10 15 20 25

HOXA9STAT5B (N642H)

STAT5B (N642H) + HOXA9

Nontransduced

1.5 × 108

1.0 × 108

5.0 × 107

0

1.5 × 108

1.0 × 108

5.0 × 107

0

Cel

l num

ber

Cel

l num

ber

Cellulartransformation assay

Bone marrowtransplant

A

B

C

D E F

G H I

P < 0.0001

***

Day 10 Day 20

Day 1 Day 10 Day 20

101

101 102 103

102

103

101

101 102 103

102

103

101

101 102 103

102

103

101

101 102 103

102

103

101

101 102 103

102

103

101

101 102 103

102

103

Start

End

Research. on January 15, 2021. © 2018 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst March 1, 2018; DOI: 10.1158/2159-8290.CD-17-0583

Page 6: HOXA9 Cooperates with Activated JAK/STAT Signaling to …...2018/04/09  · expression in 3 of 5 JAK3-mutant cases, that was up to 10-fold higher when compared with HOXA10 and HOXA11

HOXA9 and Mutant JAK3 Cooperation in Leukemia RESEARCH ARTICLE

MAY 2018 CANCER DISCOVERY | OF6

moderate and less profound cooperative effect compared with HOXA9 (median DFS = 100 days; Supplementary Fig. S1).

To negate any mouse strain–specific effects, the same experi-mental setup was repeated on a C57BL/6 background. Similar to BALB/c, recipient C57BL/6 mice receiving cotransduced JAK3M511I/HOXA9 had a rapid increase in WBC compared with JAK3M511I or HOXA9 alone (Fig. 2G), and in all cases, the double-transduced clone was the major clone at end stage (Fig. 2H). This resulted in a significant decrease in DFS (median DFS = 40 days) compared with JAK3M511I (median DFS = 150 days) or HOXA9 (median DFS = 182 days) only recipient mice (Fig. 2I). Moreover, when JAK3M511I was substituted for the con-stitutive active STAT5BN642H and cotransduced with HOXA9, this further reduced the DFS (median DFS = 12 days).

At end-stage disease, the leukemia development in vivo was characterized by both myeloid and lymphoid expan-sion. Phenotypic FACS staining and pathology analysis of the JAK3M511I/HOXA9-driven leukemia revealed that leuke-mic mice had polyclonal expansion of cells displaying either myeloid (CD16/32+, CD11b+, Gr1+/−) or lymphoid (CD8+) line-age markers and occurred at different frequencies (Fig 3A). At end-stage disease, histopathologic and immunohistochemical analysis revealed the development of a complex hematolym-phoid neoplasm characterized by the coexistence of different populations of atypical cells displaying both lymphoid and myeloid differentiation. There was significant leukemic infil-tration into spleen, lymph nodes, and bone marrow organs by atypical lymphocytes (CD3+, TdT-) and myeloid/granulocytic (myeloperoxidase-positive) cells (Fig. 3B; Supplementary Fig. S2). Western blot analysis confirmed expression of JAK3 and HOXA9 in vivo with robust phosphorylation of STAT5 (Fig. 3C). These cells proliferated for up to 30 days ex vivo in the absence of all cytokines with clonal selection of those clones expressing high levels of HOXA9 and JAK3M511I as assessed by GFP and mCHERRY expression (Fig 3D and E). Moreo-ver, sorted JAK3M511I/HOXA9 CD8+ T-ALL clones were able to engraft secondary recipient mice with higher frequency compared with age-matched JAK3M511I CD8+ leukemic clones (Supplementary Fig. S3).

To determine whether the resulting leukemia remained dependent on activated JAK/STAT5 signaling directly, JAK3M511I/HOXA9 leukemic mice were treated for 20 days with the JAK3 inhibitor tofacitinib (40 mg/kg/day; Fig. 3F). Within 3 days of treatment with tofacitinib, there was a significant decrease in the levels of pSTAT5 compared with vehicle-treated mice (Fig 3G). After 20 days of tofacitinib treatment, there was a significant attenuation in leukemia WBC expansion reduced splenomegaly (Fig 3H–J). Staining of the spleen at end stage also showed a significant reduction in the number of proliferative cells as assessed by Ki67 staining (Fig. 3K). Tofacitinib is approved for rheumatoid arthritis and known to cause decreased numbers of neutrophils, lymphocytes, and natural killer (NK) cells but rarely to an extent that leads to serious adverse effects in patients (26). Indeed, in our own studies, wild-type mice treated at 40 mg/kg/day showed a minor decrease in NK-cell frequency but no significant altera-tions in CD4+ or CD8+ T-cell frequencies (Supplementary Fig. S4). Taken together, these results first provide strong valida-tion that ectopic expressions of HOXA9 together with JAK3/STAT5 activation are bona fide cooperating events in leukemia

development. Second, there is a degree of transcription factor specificity for cooperation with JAK3 mutations, because JAK3 mutations do not cooperate with the oncogenic transcription factor TAL1. Third, the resulting leukemia remains dependent on activated JAK3 signaling.

A Novel Retroviral Vector to Direct Transgene Expression within Developing Lymphoid Cells Demonstrates Cooperation of JAK3M511I/HOXA9 in T-ALL Development

The previous data illustrate the potential of combined expression of JAK3M511I and HOXA9 to generate mixed mye-loid/lymphoid leukemia originating from multipotent hemat-opoietic precursor cells. However, HOXA9 expression is often activated in T-ALL by T cell–specific enhancers that become active only in lymphoid-committed progenitors (e.g., TCRB-HOXA9; refs. 21, 22). To convincingly demonstrate coop-eration between JAK3M511I and HOXA9 during T-cell leukemia development, and to specifically model lymphoid-specific acti-vation of HOXA9 expression as observed in human T-ALL, we developed a strategy to limit HOXA9 expression to developing lymphoid cells. We tested a previously published strategy with 3′ UTR mir-223 sites (27), but in the context of JAK3M511I/HOXA9, this strategy was unable to prevent overt myeloid expansion and eventual acute myeloid leukemia (AML) expan-sion that we speculate to indicate endogenous miR-223 satu-ration in the presence of high levels of transcript driven by the strong MSCV promoter (Supplementary Fig. S5).

We therefore developed a retroviral vector in which one or two oncogenes are initially cloned in an antisense orientation, and thus remain completely inactive. The expression of the oncogenes is activated only upon Cre-mediated inversion of the LOX66/71 flanked construct (Fig. 4A). By use of hemat-opoietic cells isolated from cell type–specific Cre mice, we could then direct expression of the oncogenes to specific hematopoi-etic cell types. The novel vector with JAK3M511I+HOXA9 was first validated in Ba/F3 cells and found to transform the cells into cytokine-independent growth only when Cre-recombinase was present and did not display ectopic “leaky” expression or cell transformation in Ba/F3 wild-type cells over an 8-day period (Fig. 4B). Western blot analysis confirmed the expres-sion of JAK3 and HOXA9 in the Cre-positive Ba/F3 cells and highlighted that although there was less JAK3M511I expression compared with constitutive expression of JAK3, both had equivalent levels of phospho-STAT5 (Fig. 4C). The novel retro-viral vector strategy was also validated in vivo using a well-estab-lished NOTCH1-ICN bone marrow transplant model, but now with ICN cloned in the antisense direction between the LOX66/71 sites. When the viral vector was transduced on cells derived from CD4-Cre mice, the recipient mice succumbed to the anticipated CD4+/CD8+ T-ALL leukemia (median overall survival = 43 days; Supplementary Fig. S6).

Using this new strategy, we next tested whether combined expression of JAK3M511I and HOXA9 via CD2-Cre– mediated activation in developing lymphoid cells (ref. 28; Fig. 4D) could generate T-cell leukemia. Mice that received transplantation with inducible JAK3M511I-P2A-HOXA9 HSPCs developed T-ALL–like leukemia with decreased latency when compared with JAK3M511I-P2A-BFP only (median DFS = 135 days vs. 244 days). Notably, both JAK3M511I and JAK3M511I-P2A-HOXA9

Research. on January 15, 2021. © 2018 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst March 1, 2018; DOI: 10.1158/2159-8290.CD-17-0583

Page 7: HOXA9 Cooperates with Activated JAK/STAT Signaling to …...2018/04/09  · expression in 3 of 5 JAK3-mutant cases, that was up to 10-fold higher when compared with HOXA10 and HOXA11

de Bock et al.RESEARCH ARTICLE

OF7 | CANCER DISCOVERY MAY 2018 www.aacrjournals.org

Figure 3.  Constitutive expression of JAK3M511I and HOXA9 leads to development of both AML and T­ALL in vivo. A, Representative peripheral blood FACS of expanding leukemic clones expressing JAK3M511I and HOXA9. B, Histopathology for MPO and CD3 staining of bone marrow, spleen, and lymph nodes. Scale bars: bone marrow, 40 μm; spleen and lymph nodes, 80 μm. C, Western blot analysis of spleen and bone marrow leukemic cells. D and E, Ex vivo growth assay of leukemia clones and matching FACS with JAK3M511I and HOXA9 levels measured by GFP and mCherry expression, respectively. F, Schematic of in vivo treatment of primary bone marrow transplant recipients. Mice were treated daily for 20 days from day 14 after transplantation with either 40 mg/kg/day tofacitinib (oral gavage) or vehicle control. G, Phospho­flow cytometry of pSTAT5 levels in JAK3M511I/HOXA9 leukemic cells in peripheral blood after 3­day tofacitinib (tofa) treatment compared with vehicle control. H, Change in WBC count before and after the treatment. I, Total WBC counts at day 35. J, Spleen weights of tofacitinib­treated vs. vehicle­treated mice at day 35. K, Ki67 staining of spleens.

82 0

8 10

100 0

0 0

3 48

8 41

0.5 0

99 0.5

M127- Day 63

HO

XA

9

JAK3 (M511I)

CD

8

CD4 CD135 CD11b

CD

16/3

2

Gr1

14%

103

101 102 103

102

101

103

101 102 103

102

101

103

101 102 103

102

101

103

101 102 103

102

101

103

101 102 103

102

101

103

101 102 103

102

101

103

101 102 103

102

101

38%12 0

88 0

2 0

93 4

MPO CD3

Bonemarrow

Spleen

Lymphnode

Spl

een

Bon

e m

arro

w

pJAK3pJAK1

JAK3

pSTAT5 (Y694)

STAT5

HOXA9 0 2 4 6 8 100

1.5 × 107

1.0 × 107

5.0 × 106

1 × 106

Spleen BM

Day

Cel

l num

ber

Spleen

Bone marrow

Day 1 Day 35

HO

XA

9(m

Che

rry)

JAK3(GFP)

A

C

F

H I J K

G

D E

B

Primary JAK3(M511I)/HOXA9Day 14 Day 35

Vehicle

Tofacitinib (40 mg/kg/day)

Day 14 Day 350

10

20

30

40

50100

200

300

400

500

WB

C (

×1,0

00)

(cel

ls/m

m3 ) V

ehicleT

ofacitinib

Vehicl

e

Tofacit

inib

Vehicl

e

Tofacit

inib

10,000

100,000

WB

C (

cells

/mm

3 )

**

P = 0.006

0

200

400

600

800

1,000

Wei

ght (

mg)

***

P < 0.001 Ki67

Vehicle

Tofacitinib

P = 0.08

pSTAT5-APC

MFI = 557

MFI = 696

MFI = 296

MFI = 424

Veh

icle

Tof

a

Research. on January 15, 2021. © 2018 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst March 1, 2018; DOI: 10.1158/2159-8290.CD-17-0583

Page 8: HOXA9 Cooperates with Activated JAK/STAT Signaling to …...2018/04/09  · expression in 3 of 5 JAK3-mutant cases, that was up to 10-fold higher when compared with HOXA10 and HOXA11

HOXA9 and Mutant JAK3 Cooperation in Leukemia RESEARCH ARTICLE

MAY 2018 CANCER DISCOVERY | OF8

Figure 4.  Novel retroviral strategy to restrict JAK3M511I/HOXA9 expression to developing lymphoid cells results in significantly decreased leukemia latency. A, Schematic of new retroviral vector incorporating antiparallel LOX66/71 sites for unidirectional inversion in the presence of Cre­recombinase. B and C, Cell proliferation of Ba/F3 or Ba/F3_Cre­recombinase cells transduced with the retroviral constructs encoding JAK3M511I or JAK3M511I_P2A_HOXA9 in between the LOX66/71 sites. C, Western blot analysis of the transduced Ba/F3. D, Model of improved bone marrow transplant utilizing Cre­donor mice in combination with novel retroviral vector. E, Kaplan–Meier DFS cure for mice with hCD2­iCre–driven inversion of JAK3M511I and HOXA9 and the cell surface phenotype frequency for the resulting leukemia (DFS = WBC <30,000 cells/mm3). F, Genomic PCR detection of the inversion of the retroviral construct.

5’LTR 3’LTR5’LTR 3’LTRLox66 Lox71

GFP

JAK3(M511I)-P2A-HOXA9

CD2-Cre donor

Retroviral spinfection of Lin− cells

Injection into sublethally irradiated wild-type recipients

Leukemia latency/disease progression assessment

JAK3(M511I)-P2A-BFPJAK3((((M511I)-P))) 2A-BFP

JAK3(((M511I)-P)) 2A-HOXA9

Days post-transplant

Dis

ease

-fre

e su

rviv

al

0 100 200 3000

50

100 JAK3(M511I)-P2A-BFP (n = 12)

JAK3(M511I)-P2A-HOXA9 (n = 13)

**P = 0.0014

23% Mixed CD8+/DN100% CD8+

77% CD8+

JAK3(M511I)-P2A-BFP JAK3(M511I)-P2A-HOXA9

0 2 4 6 80

2

4

6

8

10

Days (-IL3)

Cel

l pro

lifer

atio

n (f

old

chan

ge) JAK3(M511I)

0 2 4 6 80

2

4

6

8

10

Days (-IL3)

Cel

l pro

lifer

atio

n (f

old

chan

ge)

Ba/F3 Ba/F3_Cre

[JAK3(M511I)-P2A-HOXA9]Lox66/71

[JAK3(M5111I)-P2A-BFP]Lox66/71

phospho-JAK1

phospho-JAK3

JAK3

phospho-STAT5

STAT5

HOXA9

GFP

β-actin

IL3:Cre:

− + +− − + + +−

− − −

MSCV-J

AK3(M

511I

)

[JAK3(

M51

1I)-P

2A-B

FP]Lox

66/7

1

[JAK3(

M51

1I)-P

2A-H

OXA9]Lo

x66/

71

MSCV-J

AK3(M

511I

)

[JAK3(

M51

1I)-P

2A-B

FP]Lox

66/7

1

[JAK3(

M51

1I)-P

2A-H

OXA9]Lo

x66/

71

JAK3(M511I)

[JAK3(M511I)-P2A-HOXA9]Lox66/71

[JAK3(M5111I)-P2A-BFP]Lox66/71

3’LTR3’LTRGFPLoxPDMLox

5’LTR JAK3 (M511I) P2A HOXA9HOXA9JAK3 (M51(( 1I) )))))) P2A

F1 F2R1 R2

M12

: JA

K3-

P2A

-BF

P

M13

: JA

K3-

P2A

-BF

P

M14

: JA

K3-

P2A

-BF

P

M18

: JA

K3-

P2A

-HO

XA

9

M20

: JA

K3-

P2A

-HO

XA

9

M57

: JA

K3-

P2A

-HO

XA

9

F1+R1

F2+R2

A B

C D

E F

IRES

IRES

Research. on January 15, 2021. © 2018 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst March 1, 2018; DOI: 10.1158/2159-8290.CD-17-0583

Page 9: HOXA9 Cooperates with Activated JAK/STAT Signaling to …...2018/04/09  · expression in 3 of 5 JAK3-mutant cases, that was up to 10-fold higher when compared with HOXA10 and HOXA11

de Bock et al.RESEARCH ARTICLE

OF9 | CANCER DISCOVERY MAY 2018 www.aacrjournals.org

mice now developed a CD8+ or CD4−CD8− T-ALL, without myeloid expansion (Fig. 4E). For all mice, CD2-mediated recombination was confirmed by PCR (Fig. 4F). These data not only illustrate that JAK3 mutants cooperate with HOXA9 to induce leukemia when expressed in common lymphoid progenitors, but also demonstrate the utility and efficacy of this novel Cre-inducible retroviral vector for cell type–specific expression in the hematopoietic system.

STAT5 and HOXA9 Co-occupy Similar Genetic Loci Resulting in Increased JAK/STAT Signaling in Leukemia Cells

To determine the underlying molecular mechanism driving the enhanced oncogenic potential of JAK3M511I in the presence of HOXA9, RNA-sequencing (RNA-seq) analysis was carried out on age-matched sorted leukemic CD8+ JAK3M511I, CD8+ JAK3M511I/HOXA9 as well as sorted normal thymic CD8+ T cells (Supplementary Table S2). The addition of HOXA9 led to skewed upregulation of genes when compared with JAK3M511I-only T-ALL (Fig. 5A). Notably, principal component analysis revealed distinct clustering of myeloid and T-cell leukemias (Supplementary Fig. S7). The normal CD8+ T cells formed a tight cluster away from the JAK3M511I/HOXA9 CD8+ T ALL cells and in between the JAK3M511I-only cells. Furthermore, within the myeloid cell cluster, the JAK3M511I/HOXA9-driven AML cells clustered separately from MEIS1/HOXA9 and MLL-AF9 AML (29), suggesting different mechanisms driving these leukemias, and indeed there was no increased MEIS1 expression in our leukemia, suggesting that the cooperation is independent of MEIS1. FLT3 ITD-driven leukemia (30) clustered closer to the JAK3M511I/HOXA9 and could in part reflect the downstream activation of STAT5 in both leukemias (Supplementary Fig. S7).

Chromatin immunoprecipitation sequencing (ChIP-seq) analysis of CD8+ JAK3M511I/HOXA9 leukemia showed sig-nificant co-occupancy for both STAT5 and HOXA9 across the genome. Regions cobound by STAT5 and HOXA9 were asso-ciated with H3K27Ac and H3K4me3 marks indicating colo-calization at proximal promoters rather than distal enhancers (Fig. 5B and C). Comparison of the STAT5 signal in JAK3M511I versus JAK3M511I/HOXA9 showed strong overlap, suggest-ing that HOXA9 does not significantly reposition STAT5 to de novo sites (Supplementary Fig. S8). Specific analysis of canonical STAT5 target genes illustrated the strong co-occupancy of both STAT5 and HOXA9. For example, PIM1 kinase, a known target gene of both STAT5 (31) and HOXA9 (32), showed co-occupancy of the same DNA region for both STAT5 and HOXA9. Similarly, Bcl2, Osm, Cish, and Il7r also showed co-occupancy by STAT5 and HOXA9 (Fig. 5D; Supplementary Fig. S8). Analysis of relevant human T-ALL samples also showed strong co-occupancy of HOXA9 with STAT5 peaks at known STAT5-regulated genes including BCL6, MYC, SOCS, and PIM1 (Supplementary Fig. S9).

This strong STAT5/HOXA9 colocalization prompted the question of whether a direct protein–protein interaction occurs between STAT5 and HOXA9. Indeed, a direct interaction between HOXA9 and STAT5 is suggested based on available mass spec-trometry data (33). However, we were unable to confirm a robust protein–protein interaction between HOXA9 and STAT5 using coimmunoprecipitation, nor in an independent assay using a recombinant strepII-tagged HOXA9. Only a weak interaction

with the constitutively active STAT5BN642H mutant was observed, but not with wild-type STAT5 (Supplementary Fig. S10).

Global analysis of the regions with cobinding of HOXA9 and STAT5 found an association with diminished levels of repres-sive H3K27me3 marks at upregulated genes and concomitantly associated with increased levels of active H3K27Ac marks (Fig. 5E). Integrating the ChIP-seq analysis with the RNA-seq differ-entially expressed genes also revealed that STAT5 and HOXA9 loading was higher in upregulated genes compared with down-regulated or unchanged genes (Fig. 5F), illustrating that the presence of HOXA9 leads to increased gene activation. Indeed, gene set enrichment analysis (GSEA) pathway analysis com-paring CD8+ JAK3M511I T-ALL versus CD8+ T cells and CD8+ JAK3M511I/HOXA9 versus CD8+ JAK3M511I found a significant and increasingly positive enrichment for JAK/STAT signal-ing (Fig. 5G; Supplementary Table S3). These data show that ectopic expression of HOXA9 enhances STAT5 transcriptional activity in CD8+ leukemic cells rather than inducing a de novo gene expression program complimentary to STAT5.

PIM1 kinase is overexpressed in a number of different can-cers and is an emerging cancer drug target (34, 35). Consistent with our RNA-seq and ChIP-seq data, we confirmed increased Pim1 expression in JAK3M511I/HOXA9 leukemic mouse sam-ples by qPCR (Fig. 6A). Analysis of primary human T-ALL patient samples showed the highest levels of PIM1 expression in HOXA+ cases and those cases with IL7R/JAK3/JAK1 muta-tions (Fig. 6B). Furthermore, PIM1 RNA and protein levels were rapidly downregulated in a HOXA9+ patient-derived xenograft (PDX) sample with active JAK/STAT signaling after treatment with the JAK1 inhibitor ruxolitinib (Fig. 6C). This provided further evidence for a functional signaling net-work of PIM1 regulation downstream of JAK/STAT signaling. Exploiting this observation, we next sought to determine whether dual inhibition of PIM1 and JAK1 would be benefi-cial in JAK/STAT-mutant T-ALL samples. Here, we observed a synergistic response in JAK3-mutant T-ALL samples when treated with a JAK kinase inhibitor (ruxolitinib) in combina-tion with a PIM1 inhibitor (AZD1208) within ex vivo cell cul-ture and a significant attenuation in leukemia burden in vivo using a PDX sample (Fig. 6D and E). Taken together, these data suggest that the ectopic expression of HOXA9 cobinds with STAT5 to enhance the JAK/STAT, and targeting of both PIM1 and JAK1 provides a strong therapeutic benefit.

Chromatin Accessibility by ATAC-seq Analysis Reveals a Role for AP1 Activation

To further investigate and identify additional factors and mechanisms underlying the JAK3/HOXA9 cooperation, chro-matin accessibility was assessed at a global level using Assay for Transposase-Accessible Chromatin using sequencing (ATAC-seq) comparing JAK3M511I with JAK3M511I/HOXA9 leu-kemia. This revealed significant changes of chromatin archi-tecture with appearing peaks associated with upregulated gene expression and loading of STAT5 and HOXA9 (Fig. 7A and B). Surprisingly, many upregulated sites were bound by neither STAT5 or HOXA9. We interpreted this finding as acti-vation by secondary events downstream of activated STAT5/HOXA9 signaling. Motif scanning using i-cisTarget revealed these peaks were highly enriched for FOS and JUN motifs (Fig. 7C). RNA-seq data further confirmed increased levels of

Research. on January 15, 2021. © 2018 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst March 1, 2018; DOI: 10.1158/2159-8290.CD-17-0583

Page 10: HOXA9 Cooperates with Activated JAK/STAT Signaling to …...2018/04/09  · expression in 3 of 5 JAK3-mutant cases, that was up to 10-fold higher when compared with HOXA10 and HOXA11

HOXA9 and Mutant JAK3 Cooperation in Leukemia RESEARCH ARTICLE

MAY 2018 CANCER DISCOVERY | OF10

Figure 5.  HOXA9 and STAT5 co­occupy similar genomic regions and increase JAK/STAT signaling. A, Volcano plot of differential gene expression analysis of age­matched CD8+ T­ALL JAK3M511I/HOXA9 vs. JAK3M511I­only leukemia cells. B, JAK3M511I/HOXA9 leukemia ChIP­seq density heat maps cen­tered on STAT5 signal alongside HOXA9 and associated H3K27Ac, H3K27me3, H3K4me3, and H3K4me1 histone marks. C, Frequency of STAT5/HOXA9 co­occupied DNA regions with promoters (H3K4me3­positive) and enhancers (H3K27Ac­positive/H3K4me3­negative). D, Individual ChIP­seq tracks for canonical STAT5 target genes Pim1 and Bcl2. E, GSEA analysis of RNA­seq differentially expressed genes for JAK3M511I/HOXA9 vs. JAK3M511I and loss of the repressive H3K27me3 chromatin marks or gain in H3K27ac activation mark. F, Transcription factor loading of STAT5 and HOXA9 in upregulated genes, downregulated genes, or genes that do not change. G, Heat map and GSEA pathway of JAK/STAT signaling pathway genes in CD8+ control T­cell vs. JAK3M511I vs. JAK3M511I/HOXA9 leukemia.

Rank in ordered dataset

Dataset: CD8+ vs. JAK3(M511I)Gene set: JAK/STAT SIGNALING PATHWAY KEGG HSA04630

0 17,500

0 17,500

NES: 1.525P: 0.007FDR q-value: 0.135

Dataset: JAK3(M511I) vs. JAK3(M511I) + HOXA9Gene set: JAK/STAT SIGNALING PATHWAY KEGG HSA04630

NES: 1.659P: 0.018FDR q-value: 0.098

IL2RBIL7RPIK3R5IFNGR1JAK3IL4RPIM1STAT3IL21RPIK3R1SOCS3STAT4TYK2CBLBCREBBPIL10RBIL6RSTAT2PIK3CGBCL2L1SOS2SOCS1IL10RACISHIL12RB1CRLF2PIAS3OSMIFNGCSF2RAIL15RAIL3RAMYCIL2RASOCS2IL20RBCSF3RCSF2RBSPRY2SPRY4LIFLIFRIL13RA1CSF2IL5RAIL12AIL15EPORIL10IL22RA2IL6

CD8JA

K3(M

511I

)

JAK3(

M51

1I) +

HOXA9

A

−8 −6 −4 −2 0 2 4 6 80

5

10

15

203035404550

Log2 fold change

Log 1

0(P

adj)

HOXA9

B

Summit

+5 k

b

−5 k

b

Sco

re-r

anke

d S

TAT

5 C

hIP

-seq

pea

ks

cent

ered

at t

he s

umm

its

HOXA9STAT5 H3K27Ac H3K4me3H3K27me3 H3K4me1

E

G

−2.0 2.00.0

Dataset: JAK3(M511I)/HOXA9 vs. JAK3(M511I)Gene set: Appearing H3K27Ac peaks

Dataset: JAK3(M511I)/HOXA9 vs. JAK3(M511I)Gene set: Disappearing H3K27me3 peaks

Rank in ordered dataset0 25,000 0 25,000

NES: 1.32P: 0.029FDR q-value: 0.032

Rank in ordered dataset

D

NES: 1.43P: 0.018FDR q-value: 0.038

H3K27me3H3K4me3H3K4me1

STAT5

H3K27Ac

H3K27me3H3K4me3H3K4me1

STAT5

H3K27Ac

HOXA9

[0-100]

[0-50]

[0-1000]

[0-200]

[0-100]

[0-100]

[0-50]

[0-1000]

[0-200]

[0-100][0-40]

JAK

3(M

511I

)JA

K3(

M51

1I)

+ H

OX

A9

5 kb

Pim1

H3K27me3H3K4me3H3K4me1

STAT5

H3K27Ac

H3K27me3H3K4me3H3K4me1

STAT5

H3K27Ac

HOXA9

[0-100]

[0-50]

[0-1000]

[0-200]

[0-100]

[0-100]

[0-50]

[0-1000]

[0-200]

[0-100][0-40]

JAK

3(M

511I

)JA

K3(

M51

1I)

+ H

OX

A9

Bcl2 Kdsr Vps4b

50 kb

UpDownNeutral

STAT5 signal

−4kb +4kb−2kb +2kb

2

4

6

8

10

12

Sig

nal (

× 10

3 )

UpDownNeutral

HOXA9 signal

0.5

1

1.5

2

2.5

3

Sig

nal (

× 10

4 )

TS

S −4kb +4kb−2kb +2kb

TS

S

Promoters

Enhancers

80%

18%

Summit

+5 k

b

−5 k

b Summit

+5 k

b

−5 k

b Summit

+5 k

b

−5 k

b Summit

+5 k

b

−5 k

b Summit

+5 k

b

−5 k

b

Other

STAT5/HOXA9overlapping

peaks

C

F

Rank in ordered dataset

Research. on January 15, 2021. © 2018 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst March 1, 2018; DOI: 10.1158/2159-8290.CD-17-0583

Page 11: HOXA9 Cooperates with Activated JAK/STAT Signaling to …...2018/04/09  · expression in 3 of 5 JAK3-mutant cases, that was up to 10-fold higher when compared with HOXA10 and HOXA11

de Bock et al.RESEARCH ARTICLE

OF11 | CANCER DISCOVERY MAY 2018 www.aacrjournals.org

members of the AP1 complex including FOS and JUN (Fig. 7D) and increased expression of AP1 target genes (Fig. 7E). Although some of these target genes such as Fasl were bound by STAT5 and HOXA9, many others such as App, Klf4, and Csf1 had very weak or no binding (Fig. 7F) and represent examples of genes activated by AP1 independent of STAT5.

DiscUssiONThe development of T-ALL is a stepwise process via the accu-

mulation of somatic mutations (36). Although we and others have identified and confirmed various important driver muta-

tions in T-ALL, this has often been in the context of a single oncogene, either within transgenic (e.g., TLX1; ref. 37) or bone marrow transplant mouse models (e.g., JAK3M511I; ref. 13). How-ever, patients present with multiple mutations at diagnosis, and we therefore hypothesized that mutations that significantly associate with one another are potentially cooperating events in driving T-ALL. In a recent analysis, signi ficant associations were found between mutations in the IL7R/JAK signaling path-way and epigenetic regulators (5). Here, we extend this analysis and find that those cases with JAK3 mutations are significantly associated with cases designated HOXA+. In particular, we show that HOXA9 expression levels are significantly elevated

Figure 6.  PIM1 expression in increased in JAK3M511I/HOXA9 leukemia. A, qPCR analysis of PIM1 levels in normal CD8+ T cells, CD8+ JAK3M511I, and CD8+ JAK3M511I/HOXA9 cells B, Analysis of clinical T­ALL samples for PIM1 expression levels and those cases with JAK3/IL7R/STAT mutations (red dots). C, Analysis of PIM1 RNA and protein expression in HOXA9­positive PDX samples ex vivo after 1 μmol/L ruxolitinib over 6 hours. D, Combinatorial treat­ment with small molecular inhibitors against JAK kinases (ruxolitinib) and PIM1 kinase (AZD1208) in DND41(IL7R mutant) and JAK3­mutant PDX samples. E, In vivo combination treatment with ruxolitinib and AZD1208 (7 days) of the JAK3­mutant XC65 PDX sample.

Vehicl

eRux

oAZD

Combi

0

1

2

3

4

BLI - absolute difference (Day 7 vs. Day 1)

XC65-Day 7

VehicleRuxolitinib

(50 mg/kg/day)AZD1208

(30 mg/kg/day) Combination

RO

I (×

109 ;

p/s

)

6.0 × 107

4.0 × 107

2.0 × 107

CORE ENRICHMENT

0

1

2

3

4

A

D

E

CP

IM1

expr

essi

on (

CN

RQ

)

**

6

7

8

9

10

11

2091

93_a

t PIM

1 (R

MA

, Log

2) JAK3/JAK1/IL7R mutation

0

50

100

150 X10(CI = n/a)

0

50

100

150XC57

(CI = n/a)

0

50

100

150 XC65(CI = 0.53)

Via

bilit

y

0

50

100

150 X11(CI = 0.29)

AZD1208Ruxolitinib

Via

bilit

y

Via

bilit

y

Via

bilit

y

AZD1208Ruxolitinib

JAK3 mutant (PDX) JAK3 wild-type (PDX)

0− + − +− − + +

− + − +− − + +

− + − +− − + +

− + − +− − + +

− + − +− − + +

50

100

150

DND41(CI = 0.16)

Pro

lifer

atio

n

AZD1208Ruxolitinib

*

**

0

0.5

1.0

1.5

PIM

mR

NA

(C

NR

Q)

DM

SO

RU

XO

DM

SO

RU

XO

DM

SO

RU

XO

2h 4h 6h

PDX(XE89) : HOXA9-positive

pSTAT5

PIM1

β-actin

CD8+ T

cell

CD8+ JA

K3(M

511I

)

CD8+ JA

K3(M

511I

)/HOXA9

HOXATAL1

IMM

ATURETLX

1TLX

3

2h D

MSO

2h R

UXO

4h D

MSO

4h D

MSO

6h D

MSO

6h R

UXO

B

Research. on January 15, 2021. © 2018 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst March 1, 2018; DOI: 10.1158/2159-8290.CD-17-0583

Page 12: HOXA9 Cooperates with Activated JAK/STAT Signaling to …...2018/04/09  · expression in 3 of 5 JAK3-mutant cases, that was up to 10-fold higher when compared with HOXA10 and HOXA11

HOXA9 and Mutant JAK3 Cooperation in Leukemia RESEARCH ARTICLE

MAY 2018 CANCER DISCOVERY | OF12

0 25,000

0 25,000

Rank in ordered dataset

Dataset: JAK3(M511I)/HOXA9 vs. JAK3(M511I)Gene set: Appearing ATAC-seq peaks

A B

NES: 1.83P <0.001FDR q-value <0.001

Rank in ordered dataset

Dataset: JAK3(M511I)/HOXA9 vs. JAK3(M511I)Gene set: Disappearing ATAC-seq peaks

NES: −1.69P <0.001FDR q-value <0.001

JUN (NES = 6.29)FOSL2 (NES = 6.34)

CD8+

JAK3(

M51

1I)

JAK3(

M51

1I)/H

OXA98

10

12

14

16

18

Nor

mal

ized

cou

nts Fosl2

81012141618

Nor

mal

ized

cou

nts Fos

CD8+

JAK3(

M51

1I)

JAK3(

M51

1I)/H

OXA910

12

14

16

18

Nor

mal

ized

cou

nts

Jun

10

12

14

16

18

Nor

mal

ized

cou

nts Jund

CD8+

JAK3(

M51

1I)

JAK3(

M51

1I)/H

OXA9CD8

+

JAK3(

M51

1I)

JAK3(

M51

1I)/H

OXA9

STAT5HOXA9STAT5ATACATAC

JAK3(M511I)

JAK3(M511I)/HOXA9

CD8+

JAK3(M511I)

JAK3(M511I)/HOXA9

JAK3(M511I)

ATAC-seq

JAK3(M511I)/HOXA9

ATAC-seq

JAK3(M511I)

STAT5 ChIP

JAK3(M511I)/HOXA9

STAT5 ChIP HOXA9 ChIP

Disappearing ATAC-seq peaks

Appearing ATAC-seq peaks

CEBPB

CEBPACSF1

FGL2FASL

GZMB

APPKLF

4

0

5

10

15

Nor

mal

ized

cou

nts

AP1 target genes

Fasl

5 kb 50 kb

JAK3(M511I)JAK3(M511I)/HOXA9

STAT5HOXA9STAT5ATACATAC

JAK3(M511I)

JAK3(M511I)/HOXA9

JAK3(M511I)JAK3(M511I)/HOXA9

Klf4

5 kb

STAT5HOXA9STAT5ATACATAC

JAK3(M511I)

JAK3(M511I)/HOXA9

JAK3(M511I)JAK3(M511I)/HOXA9

App

STAT5HOXA9STAT5ATACATAC

JAK3(M511I)

JAK3(M511I)/HOXA9

JAK3(M511I)JAK3(M511I)/HOXA9

5 kb

Csf1

A

C

D

F

E

0

500

1,000

1,500

2,000

2,500

3,000

0

500

1,000

1,500

2,000

2,500

3,000

0

500

1,000

1,500

2,000

2,500

3,000

0

500

1,000

1,500

2,000

2,500

3,000

0

500

1,000

1,500

2,000

2,500

3,000−5,000 −2,500 2,500 5,0000 −5,000 −2,500 2,500 5,0000 −5,000 −2,500 2,500 5,0000 −5,000 −2,500 2,500 5,0000 −5,000 −2,500 2,500 5,0000

2.0

Bits 1.0

0.0

2.0

Bits 1.0

0.05′ 5′ 3′3′1 2 3 4 5 6 7 8 9 1 2 3 4 5 6 7 8 910 11 12

Figure 7.  ATAC­seq reveals role for AP1 complex in JAK3M511I/HOXA9­driven leukemia. A, GSEA results showing appearing and disappearing ATAC­seq peaks correlated with upregulated and downregulated genes. B, HOXA9 and STAT5 bound genomic regions relative to appearing and disappearing ATAC­seq peaks. C, i­cisTarget results for DNA­binding motifs within the appearing ATAC­seq peaks. D, Relative expression of members of the AP1 complex including Fos, Jun, Jund, and Fosl2. E, Relative expression of AP1 target genes in normal CD8+ cells and JAK3M511I or JAK3M511I/HOXA9 leukemias. F, ChIP­seq analysis tracks for AP1 targets such as Fasl (bound by STAT5 and HOXA9) and Csf1, App, and Klf4 not bound by STAT5 or HOXA9 (all track heights = 0–100).

Research. on January 15, 2021. © 2018 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst March 1, 2018; DOI: 10.1158/2159-8290.CD-17-0583

Page 13: HOXA9 Cooperates with Activated JAK/STAT Signaling to …...2018/04/09  · expression in 3 of 5 JAK3-mutant cases, that was up to 10-fold higher when compared with HOXA10 and HOXA11

de Bock et al.RESEARCH ARTICLE

OF13 | CANCER DISCOVERY MAY 2018 www.aacrjournals.org

compared with either HOXA10 or HOXA11 in HOXA+ cases, and coexpression of HOXA9 and JAK3M511I leads to the rapid development of leukemia in vivo.

In our initial mouse model, the developing leukemia in vivo was characterized by overt myeloid and lymphoid expansion due to the constitutive expression of both JAK3M511I and HOXA9 within the HSPCs. Traditionally, the modeling of T-cell leukemia in mice and the role on oncogenes have often used transgenic mice using tissue-specific promoters to limit the expression within developing lymphoid cells (e.g., CD2-LMO2, ref. 38; LCK-TLX1, ref. 37). However, generat-ing transgenic knock-in mice remains technically laborious and time consuming. To this end, we developed a retroviral expression strategy for lineage-specific expression using the bone marrow transplant system that is compatible with the numerous well-characterized Cre mice now available, and we demonstrated the efficacy of this retroviral vector to rap-idly generate mouse T-ALL models. Moreover, this system allows a more physiologic comparison of cellular competition between wild-type and transgene-expressing cells during dif-ferentiation and potential leukemia development.

From a mechanistic perspective, the integrated ChIP-seq and RNA-seq of JAK3M511I/HOXA9-driven leukemias revealed that HOXA9 expression leads to a significant amplification of STAT5 transcriptional output and to addiction of leuke-mia cells to STAT5 activation. This reconfirms STAT5 as a major central player in T-ALL pathogenesis and identifies STAT5 target genes (such as PIM1) as potential targets for therapy. PIM1 is a constitutive serine threonine kinase over-expressed in a number of cancers and regulates a number of different oncogenic processes including cell survival and apoptosis, making it an attractive target for inhibition (39). Indeed, PIM1 inhibition has been shown to be effective in a triple-negative breast cancer model in vivo (40), and more recently also in T-ALL cell lines and human T-cell leukemia virus type 1–associated adult T-cell leukemia and T-cell lymphoma (41, 42). Secondly, HOXA9 through cobinding with STAT5 also leads to the upregulation of FOS and JUN and AP1 target genes. Members of the AP1 complex are overexpressed in numerous lymphoid malignancies, including T-cell leukemia, and have been recently reviewed (43) and therefore could further contrib-ute to the enhanced leukemogenesis. The addition of HOXA9 with JAK3 mutations thereby upregulates the AP1 complex at the transcriptional levels directly rather than the canonical AP1 activation by the upstream MAPK signaling pathway.

HOXA9 has a well-established role in both hematopoiesis and leukemia. HOXA9 loss-of-function studies in mice show deficits in committed myeloid, erythroid, and B-cell progeni-tors and perturbations in T-cell development (44, 45). Con-versely, overexpression of HOXA9 in transgenic mice using lymphoid-specific regulatory elements results in mild CD8 expansion (46). Early evidence for HOXA9 in the development of leukemia was the identification of recurrent translocation t(7;11) producing the NUP98–HOXA9 fusion (47) with sub-sequent studies showing MLL fusions and NPM1 also result-ing in HOXA9 overexpression (48). Early studies of ectopic HOXA9 expression in mice showed weak leukemogenic poten-tial with either AML developing after a long latency using retroviral transduction/transplantation (49, 50) or a T-cell malignancy (preT LBL) using a transgenic approach (51). Our

bone marrow transplant studies presented here showed high leukemia penetrance for MSCV-based HOXA9 expression and reconcile with a recent study using a similar retroviral strategy (52) highlighting the role of promoter strength and viral titers in dictating leukemia latency.

Nevertheless, what is now well understood is that the ability for HOXA9 to drive rapid leukemia development and cellular transformation is dependent on the expression of different cofactors. Most of these cofactors belong to the “three loop amino-acid-loop extension” homeobox family (48). Early work over a decade ago showed an essential role for MEIS1 as a cofactor of HOXA9-driven AML in mice (49, 53), and since this time, many additional HOXA9-interacting factors have been identified including STAT5 in AML (33, 48). Indeed, STAT5B activation is important in AML, and loss of STAT5B decreases the leukemia-initiating cell frequency in HOXA9/MN1-induced leukemias (54). In our current work, we find a striking growth advantage and cellular transformation of HSPCs transduced with JAK3M511I and HOXA9 that is pheno-copied by constitutive active STAT5BN642H coexpressed with HOXA9. Moreover, in our bone marrow transplant model, we see the rapid development of both AML and T-ALL clones in vivo showing that sustained STAT5 activation and HOXA9 expression are bona fide cooperative events in driving leuke-mia development in both lineages. Significantly, the levels of MEIS1 expression are very low in our STAT5/HOXA9-driven leukemias and, together with the principal component analysis segregating HOXA9/MEIS1 AML away from both JAK3M511I/HOXA9 AML and T-ALL, suggest that MEIS1 is not important in our JAK3M511I/HOXA9 leukemia models. Fur-thermore, we showed that the resulting JAK3M511I/HOXA9-driven leukemia was sensitive to tofacitinib, suggesting these leukemias remain dependent on the STAT5 signaling and are not purely HOXA9-driven leukemias. Taken together, our data identify STAT5 as a novel cofactor of HOXA9 in T-ALL.

Interestingly, transformed progenitor cells expressing HOXA9 and MEIS1a but not HOXA9 induced the expression of FLT3 and IL7R (50), both of which activate STAT5 down-stream, suggesting that our leukemia model in part bypasses the requirement of MEIS1 via direct activation of STAT5. The link between HOX cluster activation and activated STAT sign-aling is also observed in acute megakaryoblastic leukemia with HoxR cells also carrying an activating mutation of the MPL receptor driving the phosphorylation of STAT5 (55). Similarly, MLL intragenic abnormalities in AML upregulate HOXA9 and also correlated with FLT3 mutations that activate STAT5 (56).

In conclusion, we show that HOXA9 and JAK3/STAT5 are bona fide cooperating factors in driving leukemia develop-ment and that the cooperation is situated at the transcrip-tional level where STAT5 and HOXA9 co-occupy similar genomic regions. Furthermore, our results have significance not only for T-ALL but also for other leukemias where acti-vated STAT5 would potentially cooperate with HOXA9.

MethODsExpression Plasmids and Retrovirus Production

The MSCV-JAK3M511I-IRES-GFP retroviral vector is as previ-ously described (13). The human HOXA9 cDNA was synthesized by Genscript and cloned into the MSCV-IRES-mCHERRY vector.

Research. on January 15, 2021. © 2018 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst March 1, 2018; DOI: 10.1158/2159-8290.CD-17-0583

Page 14: HOXA9 Cooperates with Activated JAK/STAT Signaling to …...2018/04/09  · expression in 3 of 5 JAK3-mutant cases, that was up to 10-fold higher when compared with HOXA10 and HOXA11

HOXA9 and Mutant JAK3 Cooperation in Leukemia RESEARCH ARTICLE

MAY 2018 CANCER DISCOVERY | OF14

The LOX66/71-IRES GFP vector was generated by cloning in the LOX66/71 sites directly into the multiple cloning site of MSCV-GFP retroviral vector. Retrovirus production using 293T cells and retrovi-ral transduction of Ba/F3 cells and hematopoietic lineage–negative cells were performed as previously described (13, 57).

RNA Extractions and qPCRRNA was extracted from tissue and cells using either the illustra

RNAspin Mini Kit (GE Healthcare Life Sciences) or the Maxwell 16 LEV Simply RNA Purification Kit (Promega) as per the manufac-turer’s instructions. cDNA synthesis was carried out using GoScript (Promega) and real-time quantitative performed using the GoTaq qPCR master mix (Promega) with the ViiA7 Real Time PCR system (Applied Biosystem). Quality control, primer efficiency, and data analysis were carried out using qbase+ software (Biogazelle). All gene expression was normalized using two housekeeping reference genes. Primers used for qPCR are listed in Supplementary Table S4.

RNA-seqThe single-end RNA-seq data were first cleaned (i.e., removal of

adapters and low-quality parts) with the fastq-mcf software after which a quality control was performed with FastQC. The reads were then mapped to the Mus Musculus (mm10) genome with Tophat2. To identify the gene expression, HTSeq-count was used to count the number of reads per gene. These read count numbers were then normalized to the sample size. Differential gene expression analysis was performed with the R-package DESeq2 (Supplementary Table S2). Pathway enrichment and other gene set enrichment analy-ses were done with GSEA. GSEA datasets were downloaded from http://download.baderlab.org/EM_Genesets/. The data are accessible through GEO Series accession number GSE109653 (https://www.ncbi. nlm.nih.gov/geo/query/acc.cgi?acc=GSE109653).

Western BlottingCells were lysed in cold lysis buffer containing 5 mmol/L

NA3VO4 and protease inhibitors (Complete EDTA-free, Roche). The proteins were separated on NuPAGE NOVEX Bis-Tris 4%–12% gels (Life Technologies) and transferred to PVDF membranes. Subsequent Western blot analysis was performed using antibod-ies directed against phospho-JAK1/JAK3 (SC-16773), STAT5 (SC-1081), JAK1 (Millipore; 05-1154), phospho-STAT5 (Cell Signaling Technology; #9359), HOXA9 (Millipore; 07-178), and beta-actin (Sigma; A1978). Anti–phospho-JAK1 antibody was used to detect both phosphorylated JAK1 and JAK3. Western blot detection was performed with secondary antibodies conjugated with horseradish peroxidase (GE Healthcare). Bands were visualized using a cooled charge-coupled device camera system (ImageQuant LAS-4000; GE Health Care).

Murine Bone Marrow TransplantationMale C57BL/6 or BALB/c mice were purchased from Charles River

Laboratories. Male mice were sacrificed, and bone marrow cells were harvested from femur and tibia. Lineage-negative cells were enriched (STEMCELL Technologies) and cultured overnight in RPMI with 20% FCS with IL3 (10 ng/mL; Peprotech), IL6 (10 ng/mL; Peprotech), SCF (50 ng/mL; Peprotech), and penicillin–streptomycin. The follow-ing day, 1 × 106 cells were transduced by spinoculation (90 minutes at 2,500 rpm) with viral supernatant and 8 μg/mL polybrene. The fol-lowing day, the cells were washed in PBS and injected (1 × 106 cells/ 0.3 mL) into the lateral tail vein of sublethally irradiated (5 Gy) syngeneic female recipient mice. Mice were housed in individually ventilated cages and monitored daily. End-point analysis for DFS was defined at WBC >30,000 cells/mm3. In vivo treatment of mice with tofacitinib was carried out as previously described (13).

Ex Vivo Cell Culture Transformation AssayThe Lin– cells were spin infected with two retroviral constructs

leading to lineage-negative cells with all four combinations— nontransduced, single transduced JAK3M511I only, HOXA9 only, and double transduced JAK3M511I/HOXA9. The day after transduction, the cells were split into media containing only 20% FBS/RPMI, removing the SCF, IL6, and IL3 that were present. The cells were then left to grow in this cytokine-free media and split when appropriate. There was no sorting of cells prior to cytokine withdrawal.

In Vivo and Ex Vivo Treatment of PDX SamplesPDX samples were transplanted in 8-week-old NSG mice through

tail-vein injection. Human leukemic cell expansion was monitored through human CD45 staining on blood samples. Single cells were isolated from the spleen, which at time of sacrifice contained >85% human CD45+ cells. Spleen cells were seeded in 96-well plate (5 × 105 cells/well) and incubated with vehicle (DMSO) or inhibitor. Cell viability was assessed at 48 hours using ATP-Lite. CompuSyn was used to calculate the combination index. For in vivo treatment stud-ies, the XC65 was transduced overnight with lentivirus pCH-SFFV-eGFP-P2A-fLuc. The GFP-positive cells were then sorted using the S3 Sorter (Bio-Rad) and retransplanted back into recipient NSG mice. Upon confirmation that XC65 was greater than >95% GFP positive, leukemic cells were isolated from the spleen and reinjected into a larger cohort of NSG mice for acute 7-day in vivo treatment. Ruxoli-tinib (HY-50858, MedChem Express) was dissolved in 0.5% methylcel-lulose, AZD1208 was dissolved in 50% PEG400/0.5% methylcellulose, and both were administered by oral gavage.

Flow Cytometry AnalysesSingle-cell suspensions were prepared from peripheral blood, bone

marrow, spleen, thymus, and lymph nodes. Cells were analyzed on either a FACS Canto flow cytometer (BD Biosciences) or MACSQuant Vyb (Miltenyi) with the following antibodies: CD3e, CD8a, CD45, CD4, CD45R, CD44, CD25, CD11b, Gr1/Ly6G, CD127, CD117, and TCRbeta. These antibodies were conjugated to phycoerythrin (PE), allophycocyanin (APC), or peridinin chlorophyll protein Cyanine5.5 (PerCP-Cy5), VioBlue, PE-Vio770, or APC-Vio770. For intracellular phospho-STAT5 staining, cells were first fixed in a 1:1 ration with IC fix buffer (Invitrogen; #00-8222-49) and then permeabilized in ice-cold 100% methanol. Staining was then carried out in PBS/2% FCS with 1:20 dilution pSTAT5-APC (eBioscience; #4334964) or IgG1 kappa isotype (eBioscience; #17-4714-41) control. Data were analyzed with the FlowJo software (Tree Star).

ChIPmentation, ChIP-Seq, and CUT&RUN ChIP-SeqChIPmentation ChIP-seq on mouse leukemic samples was carried

out as described with modifications (58). ALL-SIL cells were origi-nally sourced from DSMZ in 2004 and authenticated using short tandem repeat analysis and PCR for the NUP214–ABL1 fusion in 2017. Briefly, 20 to 40 million cells were washed in PBS and cross-linked with 1% formaldehyde for 10 minutes at room temperature and then quenched by addition of glycine (125 mmol/L final con-centration). For nuclei isolation, cells were resuspended in 1× RSB buffer (10 mmol/L Tris, pH 7.4, 10 mmol/L NaCl, 3 mmol/L MgCl2) and left on ice for 10 minutes to swell. Cells were collected by cen-trifugation and resuspended in RSBG40 buffer (10 mmol/L Tris, pH 7.4, 10 mmol/L NaCl, 3 mmol/L MgCl2, 10% glycerol, 0.5% NP40) with 1/10 v/v of 10% detergent (3.3% w/v sodium deoxycholate, 6.6% v/v Tween-40). Nuclei were collected by centrifugation and resuspended in L3B+ buffer (10 mmol/L Tris-Cl, pH 8.0, 100 mmol/L NaCl, 1 mmol/L EDTA, 0.5 mmol/L EGTA, 0.1% Na-Deoxycholate, 0.5% N-Lauroylsarcosine, 0.2% SDS). Chromatin was fragmented to 200 to 400 bp using 20 cycles (30 seconds on, 30 seconds off,

Research. on January 15, 2021. © 2018 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst March 1, 2018; DOI: 10.1158/2159-8290.CD-17-0583

Page 15: HOXA9 Cooperates with Activated JAK/STAT Signaling to …...2018/04/09  · expression in 3 of 5 JAK3-mutant cases, that was up to 10-fold higher when compared with HOXA10 and HOXA11

de Bock et al.RESEARCH ARTICLE

OF15 | CANCER DISCOVERY MAY 2018 www.aacrjournals.org

High Setting) using the Bioruptor (Diagenode). Chromatin immu-noprecipitation was carried out overnight in the presence of anti-bodies (H3K27me3 07-449 Millipore or H3K27me3 Cat# 39155 Active Motif Lot#06517018; H3K4me3 Cat# 39159 Active Motif Lot#15617005; H3K4me1 Cat#39297 Active Motif Lot#01714002; Active Motif H3K27Ac Cat#4729 Lot#GR251958-1, or H3K27Ac Cat#39134 Active Motif Lot#20017009; STAT5 Cell Signaling Cat#8363 Lot9; HOXA9 Sigma Atlas Antibodies Cat#HPA061982 Lot#86503) coupled to magnetic protein A/G beads (Millipore). Tagmentation and library preparation was carried out as described (http://www.medical-epigenomics.org/papers/schmidl2015/). DNA was purified using triple-sided SPRI bead cleanup using 1.0X, 0.65X, and 0.9X ratios (Agencourt AMPure Beads, Beckman Coulter) and analyzed by Illumina HiSeq 2000 (Illumina). Raw sequencing data were mapped to the human reference genome (GRCh37/h19) using Bowtie. Peak calling was performed using MACS 1.4. A custom Python script was used to generate the centered heat maps. To find the enriched motifs in the peaks, we made use of i-cisTarget (https://gbiomed.kuleuven.be/apps/lcb/i-cisTarget/). For the patient sample XE89, CUT&RUN ChIP-seq was carried out as described (59) using HOXA9 Sigma Atlas Antibodies (Cat#HPA061982 Lot#86503).

Ethical AspectsAnimal experiments were approved by the ethical committee on

animal experimentation of KU Leuven. Human leukemia samples were collected at the University Hospital Leuven (UZ Leuven) after approval by the ethical committee. Informed consent was obtained from all patients or their parents, according to the Declaration of Helsinki.

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

Authors’ ContributionsConception and design: C.E. de Bock, J. CoolsDevelopment of methodology: C.E. de Bock, M. Vanden Bempt, S. BornscheinAcquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): C.E. de Bock, S. Degryse, D. Verbeke, B. Sweron, O. Gielen, R. Vandepoel, M. Vanden Bempt, R. Gijsbers, J. Soulier, J.P. Meijerink, M. Heinäniemi, S. Teppo, E. RadaelliAnalysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): C.E. de Bock, S. Demeyer, D. Verbeke, C. Vicente, A. Dagklis, J.P. Meijerink, O. Lohi, E. Radaelli, J. CoolsWriting, review, and/or revision of the manuscript: C.E. de Bock, S. Demeyer, J.P. Meijerink, M. Heinäniemi, M. Bouvy-Liivrand, O. Lohi, E. Radaelli, J. CoolsAdministrative, technical, or material support (i.e., reporting or organizing data, constructing databases): C.E. de Bock, B. Sweron, R. Vandepoel, E. Geerdens, S. Bornschein, J.P. MeijerinkStudy supervision: C.E. de Bock, M. Heinäniemi, J. CoolsOther (development and production of viral vectors used in the study): R. Gijsbers

AcknowledgmentsThis work was supported by grants from KU Leuven (PF/10/016

SymBioSys), FWO-Vlaanderen, Foundation against Cancer, Swiss Bridge Foundation, and an ERC-consolidator grant (to J. Cools). M. Vanden Bempt holds an SB Fellowship of the Research Founda-tion-Flanders. D. Verbeke and S. Bornschein were supported by a fellowship from FWO-Vlaanderen, and A. Dagklis was supported by a fellowship from Kom op Tegen Kanker.

Received May 24, 2017; revised January 26, 2018; accepted February 22, 2018; published first March 1, 2018.

REFERENCES 1. Van Vlierberghe P, Ferrando A. The molecular basis of T cell acute

lymphoblastic leukemia. J Clin Invest 2012;122:3398–406. 2. Sanchez-Martin M, Ferrando A. The NOTCH1-MYC highway toward

T-cell acute lymphoblastic leukemia. Blood 2017;129:1124–33. 3. Girardi T, Vicente C, Cools J, De Keersmaecker K. The genetics and

molecular biology of T-ALL. Blood 2017;129:1113–23. 4. De Keersmaecker K, Atak ZK, Li N, Vicente C, Patchett S, Girardi T,

et al. Exome sequencing identifies mutation in CNOT3 and riboso-mal genes RPL5 and RPL10 in T-cell acute lymphoblastic leukemia. Nat Genet 2013;45:186–90.

5. Vicente C, Schwab C, Broux M, Geerdens E, Degryse S, Demeyer S, et  al. Targeted sequencing identifies association between IL7R-JAK mutations and epigenetic modulators in T-cell acute lymphoblastic leukemia. Haematologica 2015;100:1301–10.

6. Mullighan CG, Goorha S, Radtke I, Miller CB, Coustan-Smith E, Dalton JD, et al. Genome-wide analysis of genetic alterations in acute lymphoblastic leukaemia. Nature 2007;446:758–64.

7. Mullighan CG, Downing JR. PBX3 is an important cofactor of HOXA9 in leukemogenesis. Semin Hematol 2009;46:3–15.

8. Li Y, Buijs-Gladdines JGCAM, Canté-Barrett K, Stubbs AP, Vroegindeweij EM, Smits WK, et  al. IL-7 receptor mutations and steroid resistance in pediatric T cell acute lymphoblastic leukemia: a Genome Sequencing Study. PLoS Med 2016;13:e1002200.

9. Liu Y, Easton J, Shao Y, Maciaszek J, Wang Z, Wilkinson MR, et al. The genomic landscape of pediatric and young adult T-lineage acute lymphoblastic leukemia. Nat Genet 2017;63:5329.

10. Mazzucchelli R, Durum SK. Interleukin-7 receptor expression: intel-ligent design. Nat Rev Immunol 2007;7:144–54.

11. Koch U, Radtke F. Mechanisms of T cell development and transfor-mation. Annu Rev Cell Dev Biol 2011;27:539–62.

12. Ribeiro D, Melão A, Barata JT. IL-7R-mediated signaling in T-cell acute lymphoblastic leukemia. Adv Biol Regul 2013;53:211–22.

13. Degryse S, de Bock CE, Cox L, Demeyer S, Gielen O, Mentens N, et al. JAK3 mutants transform hematopoietic cells through JAK1 activa-tion, causing T-cell acute lymphoblastic leukemia in a mouse model. Blood 2014;124:3092–100.

14. Ashton JM, Balys M, Neering SJ, Hassane DC, Cowley G, Root DE, et al. Gene sets identified with oncogene cooperativity analysis regu-late in vivo growth and survival of leukemia stem cells. Cell Stem Cell 2012;11:359–72.

15. Richardson SE. Modelling how initiating and transforming onco-genes cooperate to produce a leukaemic cell state. Dis Model Mech 2013;6:3–5.

16. Castro-Giner F, Ratcliffe P, Tomlinson I. The mini-driver model of polygenic cancer evolution. Nat Rev Cancer 2015;15:680–5.

17. Treanor LM, Volanakis EJ, Zhou S, Lu T, Sherr CJ, Sorrentino BP. Functional interactions between Lmo2, the Arf tumor suppressor, and Notch1 in murine T-cell malignancies. Blood 2011;117:5453–62.

18. Kleppe M, Lahortiga I, Chaar El T, De Keersmaecker K, Mentens N, Graux C, et al. Deletion of the protein tyrosine phosphatase gene PTPN2 in T-cell acute lymphoblastic leukemia. Nat Genet 2010;42:530–5.

19. Kleppe M, Soulier J, Asnafi V, Mentens N, Hornakova T, Knoops L, et  al. PTPN2 negatively regulates oncogenic JAK1 in T-cell acute lymphoblastic leukemia. Blood 2011;117:7090–8.

20. Zhang J, Ding L, Holmfeldt L, Wu G, Heatley SL, Payne-Turner D, et al. The genetic basis of early T-cell precursor acute lymphoblastic leukaemia. Nature 2012;481:157–63.

21. Speleman F, Cauwelier B, Dastugue N, Cools J, Verhasselt B, Poppe B, et  al. A new recurrent inversion, inv(7)(p15q34), leads to transcriptional activation of HOXA10 and HOXA11 in a subset of T-cell acute lymphoblastic leukemias. Leukemia 2005;19:358–66.

22. Soulier J, Clappier E, Cayuela J-M, Regnault A, García-Peydró M, Dombret H, et  al. HOXA genes are included in genetic and bio-logic networks defining human acute T-cell leukemia (T-ALL). Blood 2005;106:274–86.

23. Losdyck E, Hornakova T, Springuel L, Degryse S, Gielen O, Cools J, et  al. Distinct acute lymphoblastic leukemia (ALL)-associated

Research. on January 15, 2021. © 2018 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst March 1, 2018; DOI: 10.1158/2159-8290.CD-17-0583

Page 16: HOXA9 Cooperates with Activated JAK/STAT Signaling to …...2018/04/09  · expression in 3 of 5 JAK3-mutant cases, that was up to 10-fold higher when compared with HOXA10 and HOXA11

HOXA9 and Mutant JAK3 Cooperation in Leukemia RESEARCH ARTICLE

MAY 2018 CANCER DISCOVERY | OF16

Janus Kinase 3 (JAK3) mutants exhibit different cytokine-receptor requirements and JAK inhibitor specificities. J Biol Chem 2015;290: 29022–34.

24. Bandapalli OR, Schuessele S, Kunz JB, Rausch T, Stütz AM, Tal N, et al. The activating STAT5B N642H mutation is a common abnor-mality in pediatric T-cell acute lymphoblastic leukemia and confers a higher risk of relapse. Haematologica 2014;99:e188–92.

25. Kontro M, Kuusanmäki H, Eldfors S, Burmeister T, Andersson EI, Bruserud O, et  al. Novel activating STAT5B mutations as putative drivers of T-cell acute lymphoblastic leukemia. Leukemia 2014;28: 1738–42.

26. Winthrop KL. The emerging safety profile of JAK inhibitors in rheu-matic disease. Nat Rev Rheumatol 2017;13:234–43.

27. Brown BD, Gentner B, Cantore A, Colleoni S, Amendola M, Zingale A, et al. Endogenous microRNA can be broadly exploited to regulate transgene expression according to tissue, lineage and differentiation state. Nat Biotechnol 2007;25:1457–67.

28. Siegemund S, Shepherd J, Xiao C, Sauer K. hCD2-iCre and Vav-iCre mediated gene recombination patterns in murine hematopoietic cells. PLoS ONE 2015;10:e0124661.

29. Zhu N, Chen M, Eng R, DeJong J, Sinha AU, Rahnamay NF, et  al. MLL-AF9- and HOXA9-mediated acute myeloid leukemia stem cell self-renewal requires JMJD1C. J Clin Invest 2016;126:997–1011.

30. Meyer SE, Qin T, Muench DE, Masuda K, Venkatasubramanian M, Orr E, et al. DNMT3A haploinsufficiency transforms FLT3ITD mye-loproliferative disease into a rapid, spontaneous, and fully penetrant acute myeloid leukemia. Cancer Discov 2016;6:501–15.

31. Matikainen S, Sareneva T, Ronni T, Lehtonen A, Koskinen PJ, Julkunen I. Interferon-alpha activates multiple STAT proteins and upregulates proliferation-associated IL-2Ralpha, c-myc, and pim-1 genes in human T cells. Blood 1999;93:1980–91.

32. Hu Y-L, Passegué E, Fong S, Largman C, Lawrence HJ. Evidence that the Pim1 kinase gene is a direct target of HOXA9. Blood 2007;109: 4732–8.

33. Huang Y, Sitwala K, Bronstein J, Sanders D, Dandekar M, Collins C, et  al. Identification and characterization of Hoxa9 binding sites in hematopoietic cells. Blood 2012;119:388–98.

34. Tursynbay Y, Zhang J, Li Z, Tokay T, Zhumadilov Z, Wu D, et al. Pim-1 kinase as cancer drug target: an update. Biomed Rep 2016;4:140–6.

35. Narlik-Grassow M, Blanco-Aparicio C, Carnero A. The PIM family of serine/threonine kinases in cancer. Med Res Rev 2014;34:136–59.

36. Martincorena I, Campbell PJ. Somatic mutation in cancer and nor-mal cells. Science 2015;349:1483–9.

37. De Keersmaecker K, Real PJ, Gatta GD, Palomero T, Sulis ML, Tosello V, et al. The TLX1 oncogene drives aneuploidy in T cell transforma-tion. Nat Med 2010;16:1321–7.

38. McCormack MP, Young LF, Vasudevan S, de Graaf CA, Codrington R, Rabbitts TH, et al. The Lmo2 oncogene initiates leukemia in mice by inducing thymocyte self-renewal. Science 2010;327:879–83.

39. Merkel AL, Meggers E, Ocker M. PIM1 kinase as a target for cancer therapy. Expert Opin Investig Drugs 2012;21:425–36.

40. Horiuchi D, Camarda R, Zhou AY, Yau C, Momcilovic O, Balakrishnan S, et al. PIM1 kinase inhibition as a targeted therapy against triple-negative breast tumors with elevated MYC expression. Nat Med 2016;22:1321–9.

41. Padi SKR, Luevano LA, An N, Pandey R, Singh N, Song JH, et  al. Targeting the PIM protein kinases for the treatment of a T-cell acute lymphoblastic leukemia subset. Oncotarget 2017;8:30199–216.

42. Bellon M, Lu L, Nicot C. Constitutive activation of Pim1 kinase is a therapeutic target for adult T-cell leukemia. Blood 2016;127:2439–50.

43. Papoudou-Bai A, Hatzimichael E, Barbouti A, Kanavaros P. Expres-sion patterns of the activator protein-1 (AP-1) family members in lymphoid neoplasms. Clin Exp Med 2016;117:5019–14.

44. Lawrence HJ, Helgason CD, Sauvageau G, Fong S, Izon DJ, Humphries RK, et al. Mice bearing a targeted interruption of the homeobox gene HOXA9 have defects in myeloid, erythroid, and lymphoid hemat-opoiesis. Blood 1997;89:1922–30.

45. Izon DJ, Rozenfeld S, Fong ST, Kömüves L, Largman C, Lawrence HJ. Loss of function of the homeobox gene Hoxa-9 perturbs early T-cell development and induces apoptosis in primitive thymocytes. Blood 1998;92:383–93.

46. Thorsteinsdottir U, Mamo A, Kroon E, Jerome L, Bijl J, Lawrence HJ, et al. Overexpression of the myeloid leukemia-associated Hoxa9 gene in bone marrow cells induces stem cell expansion. Blood 2002; 99:121–9.

47. Nakamura T, Largaespada DA, Lee MP, Johnson LA, Ohyashiki K, Toyama K, et al. Fusion of the nucleoporin gene NUP98 to HOXA9 by the chromosome translocation t(7;11)(p15;p15) in human myeloid leukaemia. Nat Genet 1996;12:154–8.

48. Collins CT, Hess JL. Role of HOXA9 in leukemia: dysregulation, cofactors and essential targets. Oncogene 2015;35:1090–8.

49. Kroon E, Krosl J, Thorsteinsdottir U, Baban S, Buchberg AM, Sauvageau G. Hoxa9 transforms primary bone marrow cells through specific collaboration with Meis1a but not Pbx1b. EMBO J 1998;17: 3714–25.

50. Wang GG, Pasillas MP, Kamps MP. Meis1 programs transcription of FLT3 and cancer stem cell character, using a mechanism that requires interaction with Pbx and a novel function of the Meis1 C-terminus. Blood 2005;106:254–64.

51. Beachy SH, Onozawa M, Silverman D, Chung YJ, Rivera MM, Aplan PD. Isolated Hoxa9 overexpression predisposes to the development of lymphoid but not myeloid leukemia. Exp Hematol 2013;41:518–529.e5.

52. Mohr S, Doebele C, Comoglio F, Berg T, Beck J, Bohnenberger H, et al. Hoxa9 and Meis1 cooperatively induce addiction to Syk signal-ing by suppressing miR-146a in acute myeloid leukemia. Cancer Cell 2017;31:549–562.e11.

53. Thorsteinsdottir U, Kroon E, Jerome L, Blasi F, Sauvageau G. Defin-ing roles for HOX and MEIS1 genes in induction of acute myeloid leukemia. Mol Cell Biol 2001;21:224–34.

54. Heuser M, Sly LM, Argiropoulos B, Kuchenbauer F, Lai C, Weng A, et  al. Modeling the functional heterogeneity of leukemia stem cells: role of STAT5 in leukemia stem cell self-renewal. Blood 2009; 114:3983–93.

55. de Rooij JDE, Branstetter C, Ma J, Li Y, Walsh MP, Cheng J, et  al. Pediatric non–down syndrome acute megakaryoblastic leukemia is characterized by distinct genomic subsets with varying outcomes. Nat Genet 2017;1–8.

56. Renneville A, Roumier C, Biggio V, Nibourel O, Boissel N, Fenaux P, et al. Cooperating gene mutations in acute myeloid leukemia: a review of the literature. Leukemia 2008;22:915–31.

57. Kleppe M, Mentens N, Tousseyn T, Wlodarska I, Cools J. MOHITO, a novel mouse cytokine-dependent T-cell line, enables studies of oncogenic signaling in the T-cell context. Haematologica 2011;96: 779–83.

58. Schmidl C, Rendeiro AF, Sheffield NC, Bock C. ChIPmentation: fast, robust, low-input ChIP-seq for histones and transcription factors. Nat Methods 2015;12:963–5.

59. Skene PJ, Henikoff S. An efficient targeted nuclease strategy for high-resolution mapping of DNA binding sites. Elife 2017;6:576.

Research. on January 15, 2021. © 2018 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst March 1, 2018; DOI: 10.1158/2159-8290.CD-17-0583

Page 17: HOXA9 Cooperates with Activated JAK/STAT Signaling to …...2018/04/09  · expression in 3 of 5 JAK3-mutant cases, that was up to 10-fold higher when compared with HOXA10 and HOXA11

Published OnlineFirst March 1, 2018.Cancer Discov   Charles E. de Bock, Sofie Demeyer, Sandrine Degryse, et al.   Drive Leukemia DevelopmentHOXA9 Cooperates with Activated JAK/STAT Signaling to

  Updated version

  10.1158/2159-8290.CD-17-0583doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://cancerdiscovery.aacrjournals.org/content/suppl/2018/03/01/2159-8290.CD-17-0583.DC1

Access the most recent supplemental material at:

   

   

   

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

  Subscriptions

Reprints and

  [email protected] at

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

  Permissions

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

.http://cancerdiscovery.aacrjournals.org/content/early/2018/04/09/2159-8290.CD-17-0583To request permission to re-use all or part of this article, use this link

Research. on January 15, 2021. © 2018 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst March 1, 2018; DOI: 10.1158/2159-8290.CD-17-0583