Acute promyelocytic leukemia (APL) is often associated with activating FLT3 signaling mutations. These are highly related to hyperleukocytosis, a major adverse risk factor with chemotherapy-based regimens. APL is a model for oncogene-targeted therapies: all-trans retinoic acid (ATRA) and arsenic both target and degrade its ProMyelocytic Leukemia/Retinoic Acid Receptor α (PML/RARA) driver. The combined ATRA/arsenic regimen now cures virtually all patients with standard-risk APL. Although FLT3-internal tandem duplication (ITD) was an adverse risk factor for historical ATRA/chemotherapy regimens, the molecular bases for this effect remain unknown. Using mouse APL models, we unexpectedly demonstrate that FLT3-ITD severely blunts ATRA response. Remarkably, although the transcriptional output of initial ATRA response is unaffected, ATRA-induced PML/RARA degradation is blunted, as is PML nuclear body reformation and activation of P53 signaling. Critically, the combination of ATRA and arsenic fully rescues therapeutic response in FLT3-ITD APLs, restoring PML/RARA degradation, PML nuclear body reformation, P53 activation, and APL eradication. Moreover, arsenic targeting of normal PML also contributes to APL response in vivo. These unexpected results explain the less favorable outcome of FLT3-ITD APLs with ATRA-based regimens, and stress the key role of PML nuclear bodies in APL eradication by the ATRA/arsenic combination.

Acute promyelocytic leukemia (APL) is caused by the t(15;17) translocation, which involves the ProMyelocytic Leukemia (PML) and Retinoic Acid Receptor α (RARA) genes.1,2 PML/RARA expression drives leukemogenesis by deregulating differentiation and self-renewal of myeloid progenitors.3  In multiple animal transgenic or knock-in models, PML/RARA expression yields typical APL, although at variable penetrance.4-7 

Retinoic acid (RA) receptor α is a transcription factor activated by All-Trans Retinoic Acid (ATRA).8  Some RARA targets regulate myeloid differentiation and granulopoiesis.9  PML is a nuclear protein that forms speckles known as PML nuclear bodies (NBs). These dynamic structures recruit p53 and most of its regulators.10-12  PML NBs are believed to facilitate posttranslational modifications, notably sumoylation and ubiquitinylation,13-15  and play a key role in senescence through their control of p53 and E2F signaling.14,16  The PML/RARA fusion protein binds corepressors and histone deacetylases,17-21  opposing RA-accelerated myeloid maturation. PML/RARA may sometimes activate transcription of some of its primary targets.22  In addition, PML/RARA heterodimerizes with PML, causing the disruption of PML NBs and redistribution of PML in a microspeckled pattern.23-25  This presumably abrogates PML-controlled pathways, and may thus also contribute to APL pathogenesis.26-28 

Patients with APL treated with chemotherapy alone had an overall poor prognosis. The addition of ATRA to anthracycline-based chemotherapy was a decisive advance in APL therapy, dramatically increasing complete remission rates and cures.29  Initially, the therapeutic effect of ATRA was believed to reflect its ability to reverse transcriptional repression of myeloid differentiation genes. Yet experiments carried out in APL mice have suggested that blast differentiation may be uncoupled from loss of leukemia-initiating cells self-renewal,30-32  shedding a renewed interest on PML/RARA degradation on ATRA treatment.32-35  Indeed, ATRA-initiated PML/RARA proteolysis allows PML NB reassembly, triggering p53 activation and senescence-like features, and ultimately drives elimination of leukemia-initiating cells.36,37 

Another therapy, arsenic trioxide, targets the PML moiety of PML/RARA, inducing its degradation and driving NB association.33,38,39  Arsenic is an extremely efficient and highly selective APL drug that cures up to 70% of patients with APL as a single agent.40,41  The molecular and biochemical bases for arsenic activity involve direct binding onto a specific site in PML driving NB formation and conjugations by Small Ubiquitin-like MOdifier (SUMO), followed by ubiquitin and proteasome-mediated PML/RARA degradation (reviewed in de Thé et al42  and Dos Santos et al43 ). Recently, the frontline combination of ATRA and arsenic in non–high-risk APL allowed the cure of most patients without the need for DNA-damaging chemotherapy.44-48 

Internal tandem duplication (ITD) of the fms-like tyrosine kinase 3 gene (FLT3) occurs with a much higher frequency in APL (35%-40%) than in other AMLs (20%-25%).49-56  FLT3 activation is tightly associated with a high white blood cell count at presentation, cases designated as high-risk APL. Although hyperleukocytosis is a well-known adverse risk factor for conventional APL therapy, the prognostic significance of FLT3-ITD per se remains somehow controversial.57-63  There is converging clinical evidence that arsenic therapy can overcome the poor prognosis associated with FLT3-ITD.45,47,64,65  Yet the basis for the relative ATRA resistance of FLT3-ITD APLs has not been comprehensively explored. Knock-in alleles of FLT3-ITD enhance self-renewal and drive a chronic myeloproliferative disease.66  Retroviral transduction of FLT3-ITD in PML/RARA transgenic cells accelerates APL development in mice,67  and in that setting, ATRA response is accelerated by FLT3 inhibitors.68,69  Here we reassessed the role of FLT3-ITD in APL development and therapy response. We demonstrate that FLT3-ITD impedes ATRA-induced APL differentiation and clearance in vivo. Such resistance was abrogated by combination with arsenic. This mechanistically explains the potency of the frontline ATRA/arsenic combination in these APLs.

Ethics statement

All experiments in mice were performed in accordance with a protocol approved by the Comité d’Ethique en Expérimentation Animale Paris-Nord no. 121, (project no. 02577.02).

Animal models

Female FVB/N and C57Bl/6Nj mice (6 weeks old) were purchased from Janvier Labs. Bone marrows from MRP8-PML/RARA transgenics70  (P/R) crossed with FTL3ki66  (P/R-FLT3ki) were injected in CD45.2 congenic FVB/N-strain syngenic mice. PML/RARAC212S immortalization of primary hematopoietic progenitors was performed as previously.71  Briefly, lineage-depleted (Lin) mouse bone marrow hematopoietic cells collected from 5-fluorouracil-treated C57Bl/6Nj Pml+/+ or Pml/ mice were infected with retroviruses obtained by transient transfection of Plat-E cells with pMSCV-PML/RARAC212S and pMIE-FLT3-ITD. After spinoculation, transduced cells were injected in C57Bl/6Nj syngenic mice. For experiments, 1 × 106 leukemic bone marrow cells were intravenously injected into recipient mice. Retinoic acid (ATRA) was administered by subcutaneous implantation of 21-day-release 10-mg pellets (Innovative Research of America). Arsenic (concentration 1 mg/mL, injected dose 5 µg/g body weight; ACROS Organics) was administered by daily intraperitoneal injection.

Retroviral transduction

Plat-E cells were transfected with retroviral vectors. Leukemic cells or Lin progenitors were then transduced with the obtained retroviral supernatants. After spinoculation, transduced cells were injected into syngenic mice.

FACS analysis

Immunophenotypic assays were performed by first blocking cellular Fc receptors with normal rat immunoglobulin G (Fc Block, Clone 93 anti-CD16 / 32, eBioscience) and then using fluorochrome-conjugated monoclonal antibodies (1:1000): APC-conjugated anti-CD117 (Clone 2B8, eBioscience), APC-Cy7-conjugated anti-CD11b (Clone M1/70, BioLegend), and phycoerythrin-conjugated anti-Gr1 (Clone RB6-8C5, eBioscience). Staining was performed overnight at 4°C. Cells were washed and resuspended in phosphate-buffered saline (PBS) with paraformaldehyde 0.2%. Apoptosis assays were carried out according to the suppliers’ recommendations with APC-Annexin-V (BD Biosciences) and phycoerythrin-active-caspase-3 apoptosis kits (BD Biosciences). Fluorescence-activated cell sorter (FACS) analyses are performed on BD LSRFortessa Cell Analyzer (BD Biosciences).

Western immunoblotting

Total cell extract was obtained from bone marrow cells by direct lysis in Laemmli buffer. Extracts were resolved on 4% to 15% precast sodium dodecyl sulfate-polyacrylamide gel electrophoresis gels (BIO-RAD) and transferred onto nitrocellulose membranes (BioTrace NT, PALL Corp.). PML/RARA was detected with rabbit polyclonal antibodies to human PML (homemade antibody, 1:10 000), RARA with rabbit anti-RARA (C-20) polyclonal antibodies (sc-551, Santa Cruz Biotechnology, 1:1000), and vinculin with mouse monoclonal antibodies (sc-73614, Santa Cruz Biotechnology, 1:1000) as a loading control. Detection was performed with the chemiluminiscent substrate SuperSignal West Femto and Dura (Thermo Fisher Scientific).

Immunofluorescence

Next, 5 × 104 bone marrow cells were cytospinned, fixed with 10% paraformaldehyde, and permeabilized in PBS 0.2% Triton-X100 for 15 minutes. Then blocking was performed with PBS 0.05% Triton-X100 1% BSA for 30 minutes. Used primary antibodies were rabbit anti-human PML (homemade, 1:1000), Alexa Fluor A488-conjugated antimurine PML (homemade, 1:2000) for 1 hour. Rabbit secondary antibodies (1:2000) conjugated to Alexa Fluor A488 or A647 fluorochromes incubated for 30 minutes. A drop of Mowiol mounting medium containing DAPI (1 µg/mL) was finally added.

Transcriptomic analysis

Total RNA was extracted from green fluorescent protein (GFP)-sorted APL bone marrow cells (RNeasy Mini Kit, Qiagen). Transcriptomic analyses were performed at the Institut Universitaire d’Hématologie plate-form. Samples were hybridized on GeneChip Mouse Transcriptome Assay 1.0. Gene set enrichment analysis (GSEA) was performed.

A FLT3-ITD knock-in allele does not accelerate PML/RARA-driven APL development

We and others had previously observed that when a FLT3-ITD was expressed from a retrovirus in PML/RARA transgenic bone marrow,67,69  activated FLT3 accelerated APL development. To further analyze the effect of the FLT3-ITD mutation on APL development, we first crossed animals bearing a FLT3-ITD knock-in allele with our MRP8-hPML/RARA transgenics. FLT3-ITD line (referred below as FLT3ki) was obtained in a C57BL/6J (B6) background, whereas hPML/RARA animals (P/R) were in the FVB/n background. Bone marrow cells from F1 P/R-FLT3ki animals were transplanted into lethally irradiated recipients. Intriguingly, there was only a trend toward earlier illness in the P/R-FLT3ki animals, with no statistically significant acceleration of leukemia development when compared with P/R alone (Figure 1A). We thus backcrossed the FLT3-ITD allele into the FVB/n background for 7 generations and generated P/R-FLT3ki animals whose bone marrow cells were transplanted into irradiated syngenic FVB/n animals. Comparing the overall and acute leukemia-free survival curves for the transplanted P/R-FLT3ki cohort along with the similar P/R cohort again showed that the P/R-FLT3ki combination did not accelerate leukemia development when compared with P/R alone (data not shown). Histopathologic findings in the P/R and P/R-FLTki leukemias showed increased immature myeloid cells occupying large areas of the bone marrow and spleen and infiltrating other organs including liver and kidney (supplemental Figure 1, available on the Blood Web site).

Figure 1.

Effect of the FLT3-ITD knock-in allele on APL initiation by PML/RARA. (A) Overall survival and acute leukemia-free survival of lethally irradiated recipients transplanted with P/R-FLT3ki, FLT3ki, or P/R bone marrow cells. (B) Strategy to obtain P/R and P/R-FLT3ki cells in a FVB background, subsequently tagged with GFP by retroviral transduction. (C) GSEA analysis for 50 hallmark gene sets between P/R and P/R-FLT3ki mice. Transcriptomic analysis performed on RNA extracted from GFP-positive leukemic cells. Only significant pathways were represented (with false discovery rate q values [FDR.q.val] <0.05) according to normalized enrichment score (NES) value.

Figure 1.

Effect of the FLT3-ITD knock-in allele on APL initiation by PML/RARA. (A) Overall survival and acute leukemia-free survival of lethally irradiated recipients transplanted with P/R-FLT3ki, FLT3ki, or P/R bone marrow cells. (B) Strategy to obtain P/R and P/R-FLT3ki cells in a FVB background, subsequently tagged with GFP by retroviral transduction. (C) GSEA analysis for 50 hallmark gene sets between P/R and P/R-FLT3ki mice. Transcriptomic analysis performed on RNA extracted from GFP-positive leukemic cells. Only significant pathways were represented (with false discovery rate q values [FDR.q.val] <0.05) according to normalized enrichment score (NES) value.

Close modal

To compare the leukemic cells from these P/R and the P/R-FLT3ki models, we retrovirally transduced leukemia blasts (from leukemias that arose in the predominantly FVB/n background) with GFP, transferred these cells into FVB/n mice (Figure 1B), sorted GFP-positive cells from the marrows of resultant leukemic animals, and compared the transcriptomes of these leukemias. As expected, P/R-FLT3ki blasts exhibited activated STAT5, K-RAS, or tumor necrosis factor/NF-kB signaling, as determined by GSEA analysis.72-77  Conversely, interferon α or γ pathways were strongly downregulated in the FLT3-ITD context78  (Figure 1C). Differentiation statuses, as assessed by May-Grünwald-Giemsa (MGG) stains and cell surface analyses (Kit, CD11b, GR1), were similar (Figure 2A). Thus, although this model exhibited the expected changes downstream of FLT3 signaling, it did not lead to a sharp acceleration of APL development.67-69 

Figure 2.

Cell differentiation and blast clearance in P/R and P/R-FLT3ki APL on ATRA treatment. (A) ATRA-induced differentiation of P/R (top) and P/R-FLT3ki (bottom) bone marrow cells, as assessed by MGG staining (scale bar, 10 µm). FACS analysis after 2, 6, and 12 days of in vivo ATRA treatment. GFP-positive leukemic cells (pink) and GFP-negative normal cells (blue) are shown. (B) Percentages of GFP-positive APL cells in bone marrow of untreated (UT) and ATRA-treated P/R (red bars) and P/R-FLT3ki (blue bars) mice at 2 to 12 days posttreatment. Data are expressed as mean ± standard deviation of 2 independent experiments. (C) Survival of secondary recipients transplanted with total APL bone marrow cells from ATRA 6-day-treated primary APL mice in both models (n ≥ 10 for each model).

Figure 2.

Cell differentiation and blast clearance in P/R and P/R-FLT3ki APL on ATRA treatment. (A) ATRA-induced differentiation of P/R (top) and P/R-FLT3ki (bottom) bone marrow cells, as assessed by MGG staining (scale bar, 10 µm). FACS analysis after 2, 6, and 12 days of in vivo ATRA treatment. GFP-positive leukemic cells (pink) and GFP-negative normal cells (blue) are shown. (B) Percentages of GFP-positive APL cells in bone marrow of untreated (UT) and ATRA-treated P/R (red bars) and P/R-FLT3ki (blue bars) mice at 2 to 12 days posttreatment. Data are expressed as mean ± standard deviation of 2 independent experiments. (C) Survival of secondary recipients transplanted with total APL bone marrow cells from ATRA 6-day-treated primary APL mice in both models (n ≥ 10 for each model).

Close modal

FLT3-ITD blunts APL cell differentiation and blast clearance on ATRA treatment

In P/R APLs, ATRA initiated terminal granulocytic differentiation assessed by MGG staining and FACS analysis (Gr1 and Mac/CD11b), already evident after 2 days.32,79  In sharp contrast, P/R-FLT3ki APLs only displayed clear features of differentiation at day 6 (Figure 2A-B). Moreover, loss of c-kit/CD117, a marker of leukemic cell self-renewal, occurred at day 2 in P/R mice, whereas c-kit expression persisted at day 6 in P/R-FLT3ki (Figure 2A-B). Examining APL clearance by GFP, we observed that ATRA cleared P/R APLs at day 8, with normal bone marrow replacing leukemia as soon as day 6.32,79  In keeping with their delayed differentiation, P/R-FLT3ki APL cell elimination was hindered at day 6 (Figure 2A-B). Moreover, ATRA treatment never eliminated P/R-FLT3ki APL cells, which actually aggressively regrew at day 10 (Figure 2B-C). As expected, loss of APL-initiating activity at day 6 was impeded in the FLT3-ITD model (Figure 2D). Altogether, FLT3-ITD mutation blocks therapeutic response to ATRA in APL mice.

FLT3-ITD impedes PML/RARA degradation and NB-reformation, not transcriptional control

To explore the basis for ATRA resistance conferred by FLT3ki expression, we first performed transcriptomic analyses at 9 and 24 hours to explore PML/RARA-mediated gene activation. Unexpectedly, robust activation of many canonical PML/RARA-target genes was observed (such as Pram1 and Cyp26a1), implying that ATRA-initiated transcription was unaffected (data not shown). Accordingly, pathway analysis showed that very similar signaling networks were activated early on in the 2 models (Figure 3A).

Figure 3.

Transcriptional analysis and PML NB reformation on ATRA treatment. (A) GSEA analysis for 50 hallmark gene sets in P/R and P/R-FLT3ki mice treated or not during 9 or 24 hours with ATRA. Only significant pathways were represented (with FDR.q.val <0.05) according to NES value. (B) PML NB reformation assessed by immunofluorescence analysis of mPML (green) and human PML/RARA (red) with DAPI (blue) in bone marrow cells of P/R and P/R-FLT3ki APL mice treated with ATRA after 2 days (scale bar, 10 µm). (C) GSEA analysis of the p53 pathway in P/R and P/R-FLT3ki mice treated during 9 or 24 hours with ATRA. NES and FDR are noted. FDR.q.val <0.05 are in bold.

Figure 3.

Transcriptional analysis and PML NB reformation on ATRA treatment. (A) GSEA analysis for 50 hallmark gene sets in P/R and P/R-FLT3ki mice treated or not during 9 or 24 hours with ATRA. Only significant pathways were represented (with FDR.q.val <0.05) according to NES value. (B) PML NB reformation assessed by immunofluorescence analysis of mPML (green) and human PML/RARA (red) with DAPI (blue) in bone marrow cells of P/R and P/R-FLT3ki APL mice treated with ATRA after 2 days (scale bar, 10 µm). (C) GSEA analysis of the p53 pathway in P/R and P/R-FLT3ki mice treated during 9 or 24 hours with ATRA. NES and FDR are noted. FDR.q.val <0.05 are in bold.

Close modal

Previous studies have stressed the role of PML NB reformation in APL regression.32,36,37,80  We thus compared NB status on ATRA treatments in vivo in our 2 models. In both untreated P/R and P/R-FLT3ki APLs, human PML/RARA and murine PML (mPML) proteins appeared as microspeckles (Figure 3B).23,25  In P/R-FLT3ki APLs, PML/RARA was often detected in the cytoplasm, as described in some patients with APL.23  After 2 days of ATRA treatment, mPML was reorganized into normal-looking NB in P/R cells. In sharp contrast, this reorganization was absent from P/R-FLT3ki APLs, where microspeckles remained apparent (Figure 3B). Failure of NB reorganization might underlie ATRA resistance, and previous studies demonstrated that p53 is a key downstream effector of PML NB.36  Indeed, the network of p53 targets was activated in P/R APLs, but not in P/R-FLT3ki ones (Figure 3B-C).

We formerly demonstrated that ATRA-induced NB-reorganization was a result of PML/RARA degradation, releasing the ability of normal PML to reform NBs.81  We thus compared PML/RARA degradation in our 2 APL models in vivo. As expected, ATRA initiated PML/RARA degradation in P/R APLs (Figure 4A). In contrast, in P/R-FLT3ki APLs, PML/RARA expression was maintained by both western blot and immunofluorescence at least up to day 6 (Figure 4A-B). Inefficient PML/RARA degradation in P/R-FLT3ki APLs is the most likely origin of therapy resistance. To investigate the basis for ATRA inability to clear PML/RARA, we examined the status of the normal RARA protein, also targeted by ATRA-initiated proteolysis.81  Although RARA was rapidly downregulated in P/R APL cells, it was not in P/R-FLT3ki ones (Figure 4C).

Figure 4.

Effect of FLT3-ITD on PML/RARA degradation on ATRA treatment. (A) Western blots of hPML/RARA (P/R, arrowheaded) and vinculin (Vcl) expression in the bone marrow of P/R (top) and P/R-FLT3ki (bottom) APL untreated (UT) and ATRA-treated mice from 2 to 12 days. Antihuman PML antibody. The percentage of GFP-positive cells is indicated for each sample. (B) Immunofluorescence analysis of human PML/RARA (green, P/R) with DAPI (blue) in bone marrow cells of P/R and P/R-FLT3ki APL mice treated with ATRA after 2 and 6 days (scale bar, 10 µm). (C) Western blot analysis of RARA degradation on in vivo ATRA treatment of the indicated time in both APL models (left). Graph shows quantification of RARA/Vcl ratio, indicative of RARA degradation (right).

Figure 4.

Effect of FLT3-ITD on PML/RARA degradation on ATRA treatment. (A) Western blots of hPML/RARA (P/R, arrowheaded) and vinculin (Vcl) expression in the bone marrow of P/R (top) and P/R-FLT3ki (bottom) APL untreated (UT) and ATRA-treated mice from 2 to 12 days. Antihuman PML antibody. The percentage of GFP-positive cells is indicated for each sample. (B) Immunofluorescence analysis of human PML/RARA (green, P/R) with DAPI (blue) in bone marrow cells of P/R and P/R-FLT3ki APL mice treated with ATRA after 2 and 6 days (scale bar, 10 µm). (C) Western blot analysis of RARA degradation on in vivo ATRA treatment of the indicated time in both APL models (left). Graph shows quantification of RARA/Vcl ratio, indicative of RARA degradation (right).

Close modal

Combined RA/As treatment overcomes FLT3-ITD-triggered resistance

Multiple clinical studies have now demonstrated the potency of the ATRA/arsenic combination, including in FLT3-mutant APLs.44,45,64,65,79,82  We thus examined the effects of the ATRA/arsenic combination in our 2 APL models. In keeping with previous studies reporting antagonism between ATRA and arsenic for differentiation,32,34  a slight delay in ATRA-initiated blast maturation was observed in P/R treated with the combination (Figures 2A and 5A). Remarkably, as early as day 2, massive cellular differentiation was now observed in both APL models treated with the combination (Figure 5A). Terminal differentiation was followed by rapid APL clearance, as determined by GFP positivity in the bone marrow, and restoration of normal hematopoiesis at day 6 in P/R-FLT3ki APLs (Figure 5B). Rapid loss of Kit, a key marker of self-renewal, may explain why addition of arsenic-promoted clearance of P/R-FLT3ki APLs. We also assessed apoptosis induction using Annexin-V and active Caspase-3 labeling (Figure 5C). Remarkably, P/R-FLT3ki APL did not exhibit enhanced apoptosis on ATRA therapy, whereas both P/R-FLT3ki and P/R APLs displayed clear apoptosis induction when treated with the arsenic/ATRA combination (Figure 5C). As expected from the nonoverlapping degrons,83  PML/RARA degradation was accelerated by the combined treatment in P/R-FLT3ki mice (Figure 5D). Adding arsenic to ATRA allowed very early restoration of mPML NBs at day 2, with a similar pattern in the 2 APL models (Figure 5E).

Figure 5.

Arsenic drives elimination of FLT3ki APLs. (A) ATRA/As-induced differentiation of P/R (top) and P/R-FLT3ki (bottom) bone marrow cells, as assessed by MGG staining (scale bar 10, µm) and FACS analysis after 2, 6, 8, and 12 days of in vivo treatment. GFP-positive leukemic cells (pink) and GFP-negative normal cells (blue) are shown. (B) Percentages of GFP-positive APL cells in bone marrow of untreated (UT) and ATRA/As-treated P/R (red bars) and P/R-FLT3ki (blue bars) mice at 2 to 12 days posttreatment. (C) Proportion of annexin-V (left) and active caspase-3 (right)-positive spleen cells in APL mice treated for 4 days, as indicated. (D) Western blot of PML/RARA (P/R, arrowheaded) and vinculin (Vcl) expression in the bone marrow of P/R-FLT3ki APL UT and ATRA/As-treated mice from 2 to 12 days. (E) PML NB reformation assessed by immunofluorescence analysis of mPML (green) and human PML/RARA (red) with 4′,6-diamidino-2-phenylindole (DAPI) (blue) in bone marrow cells of P/R and P/R-FLT3ki APL mice treated with ATRA/As after 2 days (scale bar, 10 µm).

Figure 5.

Arsenic drives elimination of FLT3ki APLs. (A) ATRA/As-induced differentiation of P/R (top) and P/R-FLT3ki (bottom) bone marrow cells, as assessed by MGG staining (scale bar 10, µm) and FACS analysis after 2, 6, 8, and 12 days of in vivo treatment. GFP-positive leukemic cells (pink) and GFP-negative normal cells (blue) are shown. (B) Percentages of GFP-positive APL cells in bone marrow of untreated (UT) and ATRA/As-treated P/R (red bars) and P/R-FLT3ki (blue bars) mice at 2 to 12 days posttreatment. (C) Proportion of annexin-V (left) and active caspase-3 (right)-positive spleen cells in APL mice treated for 4 days, as indicated. (D) Western blot of PML/RARA (P/R, arrowheaded) and vinculin (Vcl) expression in the bone marrow of P/R-FLT3ki APL UT and ATRA/As-treated mice from 2 to 12 days. (E) PML NB reformation assessed by immunofluorescence analysis of mPML (green) and human PML/RARA (red) with 4′,6-diamidino-2-phenylindole (DAPI) (blue) in bone marrow cells of P/R and P/R-FLT3ki APL mice treated with ATRA/As after 2 days (scale bar, 10 µm).

Close modal

To investigate transcriptional signaling on dual ATRA/arsenic treatment in P/R and P/R-FLT3ki APLs, we profiled GFP-sorted cells 9 and 24 hours after in vivo treatment. As before, the 2 models showed essentially similar signaling pathways modulation (Figure 6A). Yet, critically, arsenic rescued the defective activation of p53 signaling in P/R-FLT3ki APLs at 9 hours (Figure 6A-B). Accordingly, the ATRA/arsenic combination strongly impeded APL transplantability (Figure 6C compared to Figure 2C). Collectively, these data establish that resistance conferred by FLT3-ITD can be fully reversed by the addition of arsenic to ATRA treatment.

Figure 6.

Combined ATRA/As treatment overcomes FLT3-ITD-triggered resistance. (A) GSEA analysis for 50 hallmark gene sets between P/R and P/R-FLT3ki mice treated during 9 or 24 hours with ATRA/As. Only significant pathways were represented (with FDR.q.val <0.05) according to NES value. (B) GSEA analysis of the p53 pathway in P/R and P/R-FLT3ki mice treated during 9 or 24 hours with ATRA/As. NES and FDR are noted. FDR.q.val <0.05 are in bold. (C) Survival of secondary recipients transplanted with APL bone marrow cells from ATRA/As 6-day-treated primary APL mice in both models (n ≥ 8 for each model). (D) Comparison of spleen weight (left), differentiation (CD11+/Gr1+, middle) and blast clearance (GFP+, right) of untreated (UT) or As 7-day-treated APL mice with MRP8-PML/RARAC212S in Pml+/+ or Pml−/− genetic background. Data are expressed as mean ± standard deviation of at least 2 independent experiments.

Figure 6.

Combined ATRA/As treatment overcomes FLT3-ITD-triggered resistance. (A) GSEA analysis for 50 hallmark gene sets between P/R and P/R-FLT3ki mice treated during 9 or 24 hours with ATRA/As. Only significant pathways were represented (with FDR.q.val <0.05) according to NES value. (B) GSEA analysis of the p53 pathway in P/R and P/R-FLT3ki mice treated during 9 or 24 hours with ATRA/As. NES and FDR are noted. FDR.q.val <0.05 are in bold. (C) Survival of secondary recipients transplanted with APL bone marrow cells from ATRA/As 6-day-treated primary APL mice in both models (n ≥ 8 for each model). (D) Comparison of spleen weight (left), differentiation (CD11+/Gr1+, middle) and blast clearance (GFP+, right) of untreated (UT) or As 7-day-treated APL mice with MRP8-PML/RARAC212S in Pml+/+ or Pml−/− genetic background. Data are expressed as mean ± standard deviation of at least 2 independent experiments.

Close modal

Arsenic targets both hPML/RARA and mPML, possibly explaining its potency, even with the relative ATRA resistance conferred by FLT3 activation. To decipher the contribution of each partner, we generated APL models by infection of bone marrow progenitors with retroviruses encoding FLT3-ITD and PML/RARAC212S, a protein that is defective for arsenic binding.84  We also assessed arsenic response in APLs derived from Pml+/+ or Pml−/− progenitors.85  Using both spleen weight and GFP-positive cells as end points, Pml−/− APLs were completely resistant to arsenic when compared with Pml+/+ ones, stressing the role of normal PML in therapy response (supplemental Figure 2). Unexpectedly, APLs expressing PML/RARAC212S remained partially sensitive to arsenic (Figure 6D), demonstrating the key role of arsenic targeting of normal PML protein in therapy response.37,80,86 

Previous studies found that FLT3 cooperated with PML/RARA to accelerate APL development,67  and that FLT3 inhibitors synergized with ATRA to initiate differentiation in these models. Yet these studies did not explore/report ATRA response.69  Our experiments demonstrate that a knock-in FLT3-ITD allele does not accelerate APL development, but dramatically deters ATRA response. The reasons underlying these discrepancies may relate to higher levels of FLT3-ITD expression on retroviral expression in the previous studies. Yet most of our results were duplicated in a distinct model of FLT3-ITD APL mice, where the FLT3-ITD gene was directly transduced in P/R cells (supplemental Figure 3). Our observations shed a new light on multiple clinical observations that this common alteration is associated with an increased probability of relapses with ATRA/chemotherapy regimen. Interestingly, amplitude of ex vivo differentiation was linked to survival with suboptimal ATRA/chemotherapy associations, a finding that could now be interpreted as the possible effect of FLT3-ITD on ATRA-induced ex vivo differentiation.87  Mechanistically, FLT3 activation impedes ATRA-induced PML/RARA and RARA degradation. This consequently inhibits PML NB reformation and p53 activation, strongly supporting our model for a key role of this cascade in APL therapy by ATRA.36,37  Previous studies demonstrated multiple cross talks between ATRA or RARA and kinases activated downstream of FLT3.88,89  Biochemical attempts to involve them in inefficient ATRA-induced PML/RARA degradation were not successful. It is also formally possible that the delayed PML/RARA degradation also reflects FLT3-ITD-enhanced APL cell self-renewal, constantly restoring a new pool of PML/RARA-expressing cells. In that respect, STAT5, a key pathway of AML self-renewal, was clearly activated by FLT3-ITD (Figure 1C).90 

We unambiguously demonstrate that ATRA resistance in P/R-FLT3ki APLs can be circumvented by arsenic, which restores PML/RARA degradation, NB-reformation, p53 activation, and loss of self-renewal in vivo. This likely reflects the ability of arsenic to target PML/RARA for destruction by biochemical pathways distinct from ATRA-activated ones. Yet arsenic also directly targets normal PML for NB reformation. Critically, we provide evidence strongly supporting implication of normal PML in murine APL arsenic response. Not only are APLs obtained in a Pml−/− background completely arsenic-resistant, but transduction of the arsenic-resistant PML/RARAC212S mutant84  in Pml+/+ background nevertheless exhibits some response to arsenic in vivo. Indeed, in P/R-FLT3ki APLs treated with the ATRA/arsenic combination, NB reformation was more rapidly restored than PML/RARA degradation (Figure 5D-E). Note that mutations in the arsenic binding site of normal PML were observed in therapy-resistant patients,52,53,80,86  pointing to the human relevance of our observations. Clinical evidence suggested that addition of arsenic to APL therapy (either frontline or later in treatment) can erase the poor prognosis associated with FLT3 activation and/or hyperleukocytosis.45,47,64,65,91,92  Our studies imply that first-line arsenic should be used in FLT3-ITD or high-risk patients (in combination or not with some chemotherapy). More broadly, our findings could point to possible deregulation of basal ATRA/RARA signaling in tumors with activated kinase signaling.89,93,94  Our findings also provide some evidence for a role of arsenic in targeting the normal PML allele independently from PML/RARA, raising the possibility that arsenic may be active through PML targeting in selected conditions, even where PML is not rearranged.95 

All renewable reagents are available on request to the corresponding author. Transcriptomic data are publicly available at European Bioinformatics Institute, Array Express E-MTAB-7566.

The online version of this article contains a data supplement.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

The authors thank N. Setterblad and Institut Universitaire d’Hématologie technology platforms for imaging, fluorescence-activated cell sorter analysis, and transcriptomic assays; V. Parietti and M. Chopin for the animal house; and laboratory members for critical reading of the manuscript.

The H.d.T. laboratory is supported by the Ligue Nationale contre le Cancer, INSERM, Centre National de la Recherche Scientifique, University Paris Diderot, Institut National du Cancer, the Association pour la Recherche contre le Cancer (Prix Griffuel), European Research Council Senior grant 268729-STEMAPL (to H.d.T.), and TRANSCAN (DRAMA project). The contributions of C.G. and S.K. were supported by National Institutes of Health, National Cancer Institute grant R01-CA95274.

Contribution: C.E, R.R., and K.L.R. performed and analyzed in vivo experiments on P/R and P/R-FLT3ki mouse models; C.B. and A.-L.M. performed in vivo experiments on PML/RARAC212S in Pml+/+ and Pml−/− mice; C.G. generated and characterized P/R-FLT3ki mice; S.Q. performed the bioinformatics analysis; S.K. provided P/R-FLT3ki murine bone marrows; C.E, R.R., S.K, and H.d.T reviewed the data and wrote the manuscript with the assistance and final approval of all authors; and H.d.T. designed the study.

Conflict-of-interest disclosure: H de Thé consults for Vectorlabs Gmbh. The remaining authors declare no competing financial interests.

The current affiliation for K.L.R. is Phylogica, Subiaco, WA, Australia.

The current affiliation for C.G. is Genentech Inc, South San Francisco, CA.

Correspondence: Hugues de Thé, INSERM 944 Hôpital St. Louis 1 Ave Vellefaux, Paris, 75475 France; e-mail: hugues.dethe@inserm.fr.

1.
de Thé
H
,
Chomienne
C
,
Lanotte
M
,
Degos
L
,
Dejean
A
.
The t(15;17) translocation of acute promyelocytic leukaemia fuses the retinoic acid receptor alpha gene to a novel transcribed locus
.
Nature
.
1990
;
347
(
6293
):
558
-
561
.
2.
de Thé
H
,
Lavau
C
,
Marchio
A
,
Chomienne
C
,
Degos
L
,
Dejean
A
.
The PML-RAR alpha fusion mRNA generated by the t(15;17) translocation in acute promyelocytic leukemia encodes a functionally altered RAR
.
Cell
.
1991
;
66
(
4
):
675
-
684
.
3.
Du
C
,
Redner
RL
,
Cooke
MP
,
Lavau
C
.
Overexpression of wild-type retinoic acid receptor alpha (RARalpha) recapitulates retinoic acid-sensitive transformation of primary myeloid progenitors by acute promyelocytic leukemia RARalpha-fusion genes
.
Blood
.
1999
;
94
(
2
):
793
-
802
.
4.
Grisolano
JL
,
Wesselschmidt
RL
,
Pelicci
PG
,
Ley
TJ
.
Altered myeloid development and acute leukemia in transgenic mice expressing PML-RAR alpha under control of cathepsin G regulatory sequences
.
Blood
.
1997
;
89
(
2
):
376
-
387
.
5.
He
LZ
,
Tribioli
C
,
Rivi
R
, et al
.
Acute leukemia with promyelocytic features in PML/RARalpha transgenic mice
.
Proc Natl Acad Sci USA
.
1997
;
94
(
10
):
5302
-
5307
.
6.
Pollock
JL
,
Westervelt
P
,
Kurichety
AK
,
Pelicci
PG
,
Grisolano
JL
,
Ley
TJ
.
A bcr-3 isoform of RARalpha-PML potentiates the development of PML-RARalpha-driven acute promyelocytic leukemia
.
Proc Natl Acad Sci USA
.
1999
;
96
(
26
):
15103
-
15108
.
7.
Zimonjic
DB
,
Pollock
JL
,
Westervelt
P
,
Popescu
NC
,
Ley
TJ
.
Acquired, nonrandom chromosomal abnormalities associated with the development of acute promyelocytic leukemia in transgenic mice
.
Proc Natl Acad Sci USA
.
2000
;
97
(
24
):
13306
-
13311
.
8.
Chambon
P
.
A decade of molecular biology of retinoic acid receptors
.
FASEB J
.
1996
;
10
(
9
):
940
-
954
.
9.
Kastner
P
,
Lawrence
HJ
,
Waltzinger
C
,
Ghyselinck
NB
,
Chambon
P
,
Chan
S
.
Positive and negative regulation of granulopoiesis by endogenous RARalpha
.
Blood
.
2001
;
97
(
5
):
1314
-
1320
.
10.
Fogal
V
,
Gostissa
M
,
Sandy
P
, et al
.
Regulation of p53 activity in nuclear bodies by a specific PML isoform
.
EMBO J
.
2000
;
19
(
22
):
6185
-
6195
.
11.
Guo
A
,
Salomoni
P
,
Luo
J
, et al
.
The function of PML in p53-dependent apoptosis
.
Nat Cell Biol
.
2000
;
2
(
10
):
730
-
736
.
12.
Pearson
M
,
Carbone
R
,
Sebastiani
C
, et al
.
PML regulates p53 acetylation and premature senescence induced by oncogenic Ras
.
Nature
.
2000
;
406
(
6792
):
207
-
210
.
13.
Ishov
AM
,
Sotnikov
AG
,
Negorev
D
, et al
.
PML is critical for ND10 formation and recruits the PML-interacting protein daxx to this nuclear structure when modified by SUMO-1
.
J Cell Biol
.
1999
;
147
(
2
):
221
-
234
.
14.
Lallemand-Breitenbach
V
,
de Thé
H
.
PML nuclear bodies
.
Cold Spring Harb Perspect Biol
.
2010
;
2
(
5
):
a000661
.
15.
Lallemand-Breitenbach
V
,
de Thé
H
.
PML nuclear bodies: from architecture to function
.
Curr Opin Cell Biol
.
2018
;
52
:
154
-
161
.
16.
Vernier
M
,
Ferbeyre
G
.
Complete senescence: RB and PML share the task
.
Cell Cycle
.
2014
;
13
(
5
):
696
.
17.
Kamashev
D
,
Vitoux
D
,
De Thé
H
.
PML-RARA-RXR oligomers mediate retinoid and rexinoid/cAMP cross-talk in acute promyelocytic leukemia cell differentiation
.
J Exp Med
.
2004
;
199
(
8
):
1163
-
1174
.
18.
Licht
JD
.
Reconstructing a disease: What essential features of the retinoic acid receptor fusion oncoproteins generate acute promyelocytic leukemia?
Cancer Cell
.
2006
;
9
(
2
):
73
-
74
.
19.
Martens
JH
,
Brinkman
AB
,
Simmer
F
, et al
.
PML-RARalpha/RXR alters the epigenetic landscape in acute promyelocytic leukemia
.
Cancer Cell
.
2010
;
17
(
2
):
173
-
185
.
20.
Minucci
S
,
Maccarana
M
,
Cioce
M
, et al
.
Oligomerization of RAR and AML1 transcription factors as a novel mechanism of oncogenic activation
.
Mol Cell
.
2000
;
5
(
5
):
811
-
820
.
21.
Zhu
J
,
Nasr
R
,
Pérès
L
, et al
.
RXR is an essential component of the oncogenic PML/RARA complex in vivo
.
Cancer Cell
.
2007
;
12
(
1
):
23
-
35
.
22.
Viale
A
,
De Franco
F
,
Orleth
A
, et al
.
Cell-cycle restriction limits DNA damage and maintains self-renewal of leukaemia stem cells
.
Nature
.
2009
;
457
(
7225
):
51
-
56
.
23.
Daniel
MT
,
Koken
M
,
Romagné
O
, et al
.
PML protein expression in hematopoietic and acute promyelocytic leukemia cells
.
Blood
.
1993
;
82
(
6
):
1858
-
1867
.
24.
Koken
MH
,
Daniel
MT
,
Gianni
M
, et al
.
Retinoic acid, but not arsenic trioxide, degrades the PLZF/RARalpha fusion protein, without inducing terminal differentiation or apoptosis, in a RA-therapy resistant t(11;17)(q23;q21) APL patient
.
Oncogene
.
1999
;
18
(
4
):
1113
-
1118
.
25.
Koken
MH
,
Puvion-Dutilleul
F
,
Guillemin
MC
, et al
.
The t(15;17) translocation alters a nuclear body in a retinoic acid-reversible fashion
.
EMBO J
.
1994
;
13
(
5
):
1073
-
1083
.
26.
de Thé
H
,
Chen
Z
.
Acute promyelocytic leukaemia: novel insights into the mechanisms of cure
.
Nat Rev Cancer
.
2010
;
10
(
11
):
775
-
783
.
27.
Voisset
E
,
Moravcsik
E
,
Stratford
EW
, et al
.
Pml nuclear body disruption cooperates in APL pathogenesis and impairs DNA damage repair pathways in mice
.
Blood
.
2018
;
131
(
6
):
636
-
648
.
28.
Occhionorelli
M
,
Santoro
F
,
Pallavicini
I
, et al
.
The self-association coiled-coil domain of PML is sufficient for the oncogenic conversion of the retinoic acid receptor (RAR) alpha
.
Leukemia
.
2011
;
25
(
5
):
814
-
820
.
29.
Chen
SJ
,
Zhou
GB
,
Zhang
XW
,
Mao
JH
,
de Thé
H
,
Chen
Z
.
From an old remedy to a magic bullet: molecular mechanisms underlying the therapeutic effects of arsenic in fighting leukemia
.
Blood
.
2011
;
117
(
24
):
6425
-
6437
.
30.
Ablain
J
,
de The
H
.
Revisiting the differentiation paradigm in acute promyelocytic leukemia
.
Blood
.
2011
;
117
(
22
):
5795
-
5802
.
31.
Ablain
J
,
Leiva
M
,
Peres
L
,
Fonsart
J
,
Anthony
E
,
de Thé
H
.
Uncoupling RARA transcriptional activation and degradation clarifies the bases for APL response to therapies
.
J Exp Med
.
2013
;
210
(
4
):
647
-
653
.
32.
Nasr
R
,
Guillemin
MC
,
Ferhi
O
, et al
.
Eradication of acute promyelocytic leukemia-initiating cells through PML-RARA degradation [published correction appears in Nat Med. 2009;15(1):117]
.
Nat Med
.
2008
;
14
(
12
):
1333
-
1342
.
33.
Zhu
J
,
Koken
MH
,
Quignon
F
, et al
.
Arsenic-induced PML targeting onto nuclear bodies: implications for the treatment of acute promyelocytic leukemia
.
Proc Natl Acad Sci USA
.
1997
;
94
(
8
):
3978
-
3983
.
34.
Shao
W
,
Fanelli
M
,
Ferrara
FF
, et al
.
Arsenic trioxide as an inducer of apoptosis and loss of PML/RAR alpha protein in acute promyelocytic leukemia cells
.
J Natl Cancer Inst
.
1998
;
90
(
2
):
124
-
133
.
35.
Licht
JD
.
Acute promyelocytic leukemia–weapons of mass differentiation
.
N Engl J Med
.
2009
;
360
(
9
):
928
-
930
.
36.
Ablain
J
,
Rice
K
,
Soilihi
H
,
de Reynies
A
,
Minucci
S
,
de Thé
H
.
Activation of a promyelocytic leukemia-tumor protein 53 axis underlies acute promyelocytic leukemia cure
.
Nat Med
.
2014
;
20
(
2
):
167
-
174
.
37.
de Thé
H
,
Pandolfi
PP
,
Chen
Z
.
Acute promyelocytic leukemia: a paradigm for oncoprotein-targeted cure
.
Cancer Cell
.
2017
;
32
(
5
):
552
-
560
.
38.
Lallemand-Breitenbach
V
,
Zhu
J
,
Puvion
F
, et al
.
Role of promyelocytic leukemia (PML) sumolation in nuclear body formation, 11S proteasome recruitment, and As2O3-induced PML or PML/retinoic acid receptor alpha degradation
.
J Exp Med
.
2001
;
193
(
12
):
1361
-
1371
.
39.
Lallemand-Breitenbach
V
,
Jeanne
M
,
Benhenda
S
, et al
.
Arsenic degrades PML or PML-RARalpha through a SUMO-triggered RNF4/ubiquitin-mediated pathway
.
Nat Cell Biol
.
2008
;
10
(
5
):
547
-
555
.
40.
Ghavamzadeh
A
,
Alimoghaddam
K
,
Rostami
S
, et al
.
Phase II study of single-agent arsenic trioxide for the front-line therapy of acute promyelocytic leukemia
.
J Clin Oncol
.
2011
;
29
(
20
):
2753
-
2757
.
41.
Mathews
V
,
George
B
,
Chendamarai
E
, et al
.
Single-agent arsenic trioxide in the treatment of newly diagnosed acute promyelocytic leukemia: long-term follow-up data
.
J Clin Oncol
.
2010
;
28
(
24
):
3866
-
3871
.
42.
de Thé
H
,
Le Bras
M
,
Lallemand-Breitenbach
V
.
The cell biology of disease: Acute promyelocytic leukemia, arsenic, and PML bodies
.
J Cell Biol
.
2012
;
198
(
1
):
11
-
21
.
43.
Dos Santos
GA
,
Kats
L
,
Pandolfi
PP
.
Synergy against PML-RARa: targeting transcription, proteolysis, differentiation, and self-renewal in acute promyelocytic leukemia
.
J Exp Med
.
2013
;
210
(
13
):
2793
-
2802
.
44.
Lo-Coco
F
,
Avvisati
G
,
Vignetti
M
, et al
;
Study Alliance Leukemia
.
Retinoic acid and arsenic trioxide for acute promyelocytic leukemia
.
N Engl J Med
.
2013
;
369
(
2
):
111
-
121
.
45.
Burnett
AK
,
Russell
NH
,
Hills
RK
, et al
;
UK National Cancer Research Institute Acute Myeloid Leukaemia Working Group
.
Arsenic trioxide and all-trans retinoic acid treatment for acute promyelocytic leukaemia in all risk groups (AML17): results of a randomised, controlled, phase 3 trial
.
Lancet Oncol
.
2015
;
16
(
13
):
1295
-
1305
.
46.
Zhu
HH
,
Qin
YZ
,
Huang
XJ
.
Resistance to arsenic therapy in acute promyelocytic leukemia
.
N Engl J Med
.
2014
;
370
(
19
):
1864
-
1866
.
47.
Zhu
HH
,
Liu
YR
,
Jia
JS
,
Qin
YZ
,
Zhao
XS
,
Lai
YY
.
Oral arsenic and all-trans retinoic acid for high-risk acute promyelocytic leukemia
.
Blood
.
2018
;
131
(
26
):
2987
-
2989
.
48.
Lo-Coco
F
,
Di Donato
L
,
Schlenk
RF
GIMEMA
;
German–Austrian Acute Myeloid Leukemia Study Group and Study Alliance Leukemia
.
Targeted therapy alone for acute promyelocytic leukemia
.
N Engl J Med
.
2016
;
374
(
12
):
1197
-
1198
.
49.
Beitinjaneh
A
,
Jang
S
,
Roukoz
H
,
Majhail
NS
.
Prognostic significance of FLT3 internal tandem duplication and tyrosine kinase domain mutations in acute promyelocytic leukemia: a systematic review
.
Leuk Res
.
2010
;
34
(
7
):
831
-
836
.
50.
Kainz
B
,
Heintel
D
,
Marculescu
R
, et al
.
Variable prognostic value of FLT3 internal tandem duplications in patients with de novo AML and a normal karyotype, t(15;17), t(8;21) or inv(16)
.
Hematol J
.
2002
;
3
(
6
):
283
-
289
.
51.
Kiyoi
H
,
Naoe
T
,
Yokota
S
, et al
;
Leukemia Study Group of the Ministry of Health and Welfare (Kohseisho)
.
Internal tandem duplication of FLT3 associated with leukocytosis in acute promyelocytic leukemia
.
Leukemia
.
1997
;
11
(
9
):
1447
-
1452
.
52.
Lehmann-Che
J
,
Bally
C
,
Letouzé
E
, et al
.
Dual origin of relapses in retinoic-acid resistant acute promyelocytic leukemia
.
Nat Commun
.
2018
;
9
(
1
):
2047
.
53.
Madan
V
,
Shyamsunder
P
,
Han
L
, et al
.
Comprehensive mutational analysis of primary and relapse acute promyelocytic leukemia
.
Leukemia
.
2016
;
30
(
8
):
1672
-
1681
.
54.
Miller
CA
,
Wilson
RK
,
Ley
TJ
.
Genomic landscapes and clonality of de novo AML
.
N Engl J Med
.
2013
;
369
(
15
):
1473
.
55.
Ronchini
C
,
Brozzi
A
,
Riva
L
, et al
.
PML-RARA-associated cooperating mutations belong to a transcriptional network that is deregulated in myeloid leukemias
.
Leukemia
.
2017
;
31
(
9
):
1975
-
1986
.
56.
Welch
JS
,
Ley
TJ
,
Link
DC
, et al
.
The origin and evolution of mutations in acute myeloid leukemia
.
Cell
.
2012
;
150
(
2
):
264
-
278
.
57.
Breccia
M
,
Loglisci
G
,
Loglisci
MG
, et al
.
FLT3-ITD confers poor prognosis in patients with acute promyelocytic leukemia treated with AIDA protocols: long-term follow-up analysis
.
Haematologica
.
2013
;
98
(
12
):
e161
-
e163
.
58.
Callens
C
,
Chevret
S
,
Cayuela
JM
, et al
;
European APL Group
.
Prognostic implication of FLT3 and Ras gene mutations in patients with acute promyelocytic leukemia (APL): a retrospective study from the European APL Group
.
Leukemia
.
2005
;
19
(
7
):
1153
-
1160
.
59.
Hong
SD
,
Kim
YK
,
Kim
HN
, et al
.
Treatment outcome of all-trans retinoic acid/anthracycline combination chemotherapy and the prognostic impact of FLT3/ITD mutation in acute promyelocytic leukemia patients
.
Korean J Hematol
.
2011
;
46
(
1
):
24
-
30
.
60.
Lucena-Araujo
AR
,
Kim
HT
,
Jacomo
RH
, et al
.
Internal tandem duplication of the FLT3 gene confers poor overall survival in patients with acute promyelocytic leukemia treated with all-trans retinoic acid and anthracycline-based chemotherapy: an International Consortium on Acute Promyelocytic Leukemia study
.
Ann Hematol
.
2014
;
93
(
12
):
2001
-
2010
.
61.
Schnittger
S
,
Bacher
U
,
Haferlach
C
,
Kern
W
,
Alpermann
T
,
Haferlach
T
.
Clinical impact of FLT3 mutation load in acute promyelocytic leukemia with t(15;17)/PML-RARA
.
Haematologica
.
2011
;
96
(
12
):
1799
-
1807
.
62.
Souza Melo
CP
,
Campos
CB
,
Dutra
AP
, et al
.
Correlation between FLT3-ITD status and clinical, cellular and molecular profiles in promyelocytic acute leukemias
.
Leuk Res
.
2015
;
39
(
2
):
131
-
137
.
63.
Xue
MX
,
Qiu
HY
,
Feng
YF
, et al
.
[Prevalence and clinical significance of FLT3 mutations in acute promyelocytic leukemia]
.
Zhonghua Xue Ye Xue Za Zhi
.
2008
;
29
(
11
):
757
-
761
.
64.
Iland
HJ
,
Collins
M
,
Bradstock
K
, et al
;
Australasian Leukaemia and Lymphoma Group
.
Use of arsenic trioxide in remission induction and consolidation therapy for acute promyelocytic leukaemia in the Australasian Leukaemia and Lymphoma Group (ALLG) APML4 study: a non-randomised phase 2 trial
.
Lancet Haematol
.
2015
;
2
(
9
):
e357
-
e366
.
65.
Iland
HJ
,
Bradstock
K
,
Supple
SG
, et al
;
Australasian Leukaemia and Lymphoma Group
.
All-trans-retinoic acid, idarubicin, and IV arsenic trioxide as initial therapy in acute promyelocytic leukemia (APML4)
.
Blood
.
2012
;
120
(
8
):
1570
-
1580, quiz 1752
.
66.
Lee
BH
,
Tothova
Z
,
Levine
RL
, et al
.
FLT3 mutations confer enhanced proliferation and survival properties to multipotent progenitors in a murine model of chronic myelomonocytic leukemia
.
Cancer Cell
.
2007
;
12
(
4
):
367
-
380
.
67.
Kelly
LM
,
Kutok
JL
,
Williams
IR
, et al
.
PML/RARalpha and FLT3-ITD induce an APL-like disease in a mouse model
.
Proc Natl Acad Sci USA
.
2002
;
99
(
12
):
8283
-
8288
.
68.
Lee
BD
,
Sevcikova
S
,
Kogan
SC
.
Dual treatment with FLT3 inhibitor SU11657 and doxorubicin increases survival of leukemic mice
.
Leuk Res
.
2007
;
31
(
8
):
1131
-
1134
.
69.
Sohal
J
,
Phan
VT
,
Chan
PV
, et al
.
A model of APL with FLT3 mutation is responsive to retinoic acid and a receptor tyrosine kinase inhibitor, SU11657
.
Blood
.
2003
;
101
(
8
):
3188
-
3197
.
70.
Brown
D
,
Kogan
S
,
Lagasse
E
, et al
.
A PMLRARalpha transgene initiates murine acute promyelocytic leukemia
.
Proc Natl Acad Sci USA
.
1997
;
94
(
6
):
2551
-
2556
.
71.
Shen
Y
,
Fu
YK
,
Zhu
YM
, et al
.
Mutations of epigenetic modifier genes as a poor prognostic factor in acute promyelocytic leukemia under treatment with all-trans retinoic acid and arsenic trioxide
.
EBioMedicine
.
2015
;
2
(
6
):
563
-
571
.
72.
Choudhary
C
,
Brandts
C
,
Schwable
J
, et al
.
Activation mechanisms of STAT5 by oncogenic Flt3-ITD
.
Blood
.
2007
;
110
(
1
):
370
-
374
.
73.
Mizuki
M
,
Fenski
R
,
Halfter
H
, et al
.
Flt3 mutations from patients with acute myeloid leukemia induce transformation of 32D cells mediated by the Ras and STAT5 pathways
.
Blood
.
2000
;
96
(
12
):
3907
-
3914
.
74.
Mizuki
M
,
Schwable
J
,
Steur
C
, et al
.
Suppression of myeloid transcription factors and induction of STAT response genes by AML-specific Flt3 mutations
.
Blood
.
2003
;
101
(
8
):
3164
-
3173
.
75.
Kupsa
T
,
Vanek
J
,
Vasatova
M
, et al
.
Evaluation of cytokines and soluble adhesion molecules in patients with newly diagnosed acute myeloid leukemia: the role of TNF-alpha and FLT3-ITD
.
Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub
.
2016
;
160
(
1
):
94
-
99
.
76.
Mead
AJ
,
Neo
WH
,
Barkas
N
, et al
.
Niche-mediated depletion of the normal hematopoietic stem cell reservoir by Flt3-ITD-induced myeloproliferation
.
J Exp Med
.
2017
;
214
(
7
):
2005
-
2021
.
77.
Schnetzke
U
,
Fischer
M
,
Spies-Weisshart
B
, et al
.
The E3 ubiquitin ligase TRAF2 can contribute to TNF-α resistance in FLT3-ITD-positive AML cells
.
Leuk Res
.
2013
;
37
(
11
):
1557
-
1564
.
78.
Wallace
JA
,
Kagele
DA
,
Eiring
AM
, et al
.
miR-155 promotes FLT3-ITD-induced myeloproliferative disease through inhibition of the interferon response
.
Blood
.
2017
;
129
(
23
):
3074
-
3086
.
79.
Lallemand-Breitenbach
V
,
Guillemin
MC
,
Janin
A
, et al
.
Retinoic acid and arsenic synergize to eradicate leukemic cells in a mouse model of acute promyelocytic leukemia
.
J Exp Med
.
1999
;
189
(
7
):
1043
-
1052
.
80.
Lehmann-Che
J
,
Bally
C
,
de Thé
H
.
Resistance to therapy in acute promyelocytic leukemia
.
N Engl J Med
.
2014
;
371
(
12
):
1170
-
1172
.
81.
Zhu
J
,
Gianni
M
,
Kopf
E
, et al
.
Retinoic acid induces proteasome-dependent degradation of retinoic acid receptor alpha (RARalpha) and oncogenic RARalpha fusion proteins
.
Proc Natl Acad Sci USA
.
1999
;
96
(
26
):
14807
-
14812
.
82.
Hu
J
,
Liu
YF
,
Wu
CF
, et al
.
Long-term efficacy and safety of all-trans retinoic acid/arsenic trioxide-based therapy in newly diagnosed acute promyelocytic leukemia
.
Proc Natl Acad Sci USA
.
2009
;
106
(
9
):
3342
-
3347
.
83.
Zhu
J
,
Lallemand-Breitenbach
V
,
de Thé
H
.
Pathways of retinoic acid- or arsenic trioxide-induced PML/RARalpha catabolism, role of oncogene degradation in disease remission
.
Oncogene
.
2001
;
20
(
49
):
7257
-
7265
.
84.
Jeanne
M
,
Lallemand-Breitenbach
V
,
Ferhi
O
, et al
.
PML/RARA oxidation and arsenic binding initiate the antileukemia response of As2O3
.
Cancer Cell
.
2010
;
18
(
1
):
88
-
98
.
85.
Minucci
S
,
Monestiroli
S
,
Giavara
S
, et al
.
PML-RAR induces promyelocytic leukemias with high efficiency following retroviral gene transfer into purified murine hematopoietic progenitors
.
Blood
.
2002
;
100
(
8
):
2989
-
2995
.
86.
Iaccarino
L
,
Ottone
T
,
Divona
M
, et al
.
Mutations affecting both the rearranged and the unrearranged PML alleles in refractory acute promyelocytic leukaemia
.
Br J Haematol
.
2016
;
172
(
6
):
909
-
913
.
87.
Cassinat
B
,
Chevret
S
,
Zassadowski
F
, et al
;
European APL Group
.
In vitro all-trans retinoic acid sensitivity of acute promyelocytic leukemia blasts: a novel indicator of poor patient outcome
.
Blood
.
2001
;
98
(
9
):
2862
-
2864
.
88.
Gianni
M
,
Peviani
M
,
Bruck
N
, et al
.
p38αMAPK interacts with and inhibits RARα: suppression of the kinase enhances the therapeutic activity of retinoids in acute myeloid leukemia cells
.
Leukemia
.
2012
;
26
(
8
):
1850
-
1861
.
89.
Bruck
N
,
Vitoux
D
,
Ferry
C
, et al
.
A coordinated phosphorylation cascade initiated by p38MAPK/MSK1 directs RARalpha to target promoters
.
EMBO J
.
2009
;
28
(
1
):
34
-
47
.
90.
Chung
KY
,
Morrone
G
,
Schuringa
JJ
,
Wong
B
,
Dorn
DC
,
Moore
MA
.
Enforced expression of an Flt3 internal tandem duplication in human CD34+ cells confers properties of self-renewal and enhanced erythropoiesis
.
Blood
.
2005
;
105
(
1
):
77
-
84
.
91.
Abaza
Y
,
Kantarjian
H
,
Garcia-Manero
G
, et al
.
Long-term outcome of acute promyelocytic leukemia treated with all-trans-retinoic acid, arsenic trioxide, and gemtuzumab
.
Blood
.
2017
;
129
(
10
):
1275
-
1283
.
92.
Zhu
HH
,
Huang
XJ
.
Oral arsenic and retinoic acid for non-high-risk acute promyelocytic leukemia
.
N Engl J Med
.
2014
;
371
(
23
):
2239
-
2241
.
93.
Alsafadi
S
,
Even
C
,
Falet
C
, et al
.
Retinoic acid receptor alpha amplifications and retinoic acid sensitivity in breast cancers
.
Clin Breast Cancer
.
2013
;
13
(
5
):
401
-
408
.
94.
Rochette-Egly
C
.
Retinoic acid signaling and mouse embryonic stem cell differentiation: Cross talk between genomic and non-genomic effects of RA
.
Biochim Biophys Acta
.
2015
;
1851
(
1
):
66
-
75
.
95.
Kozono
S
,
Lin
YM
,
Seo
HS
, et al
.
Arsenic targets Pin1 and cooperates with retinoic acid to inhibit cancer-driving pathways and tumor-initiating cells
.
Nat Commun
.
2018
;
9
(
1
):
3069
.

Author notes

*

C.E., R.R., and K.L.R. contributed equally to this study.

Supplemental data

Sign in via your Institution