JAK2V617F myeloproliferative neoplasm eradication by a novel interferon/arsenic therapy involves PML

Tracy Dagher, Nabih Maslah, Valérie Edmond, Bruno Cassinat, William Vainchenker, Stéphane Giraudier, Florence Pasquier, Emmanuelle Verger, Michiko Niwa-Kawakita, Valérie Lallemand-Breitenbach, Isabelle Plo, Jean-Jacques Kiladjian, Jean-Luc Villeval, Hugues de Thé, Tracy Dagher, Nabih Maslah, Valérie Edmond, Bruno Cassinat, William Vainchenker, Stéphane Giraudier, Florence Pasquier, Emmanuelle Verger, Michiko Niwa-Kawakita, Valérie Lallemand-Breitenbach, Isabelle Plo, Jean-Jacques Kiladjian, Jean-Luc Villeval, Hugues de Thé

Abstract

Interferon α (IFNα) is used to treat JAK2V617F-driven myeloproliferative neoplasms (MPNs) but rarely clears the disease. We investigated the IFNα mechanism of action focusing on PML, an interferon target and key senescence gene whose targeting by arsenic trioxide (ATO) drives eradication of acute promyelocytic leukemia. ATO sharply potentiated IFNα-induced growth suppression of JAK2V617F patient or mouse hematopoietic progenitors, which required PML and was associated with features of senescence. In a mouse MPN model, combining ATO with IFNα enhanced and accelerated responses, eradicating MPN in most mice by targeting disease-initiating cells. These results predict potent clinical efficacy of the IFNα+ATO combination in patients and identify PML as a major effector of therapy, even in malignancies with an intact PML gene.

Conflict of interest statement

Disclosures: B. Cassinat reported a pending patent to WO2018134260. W. Vainchenker reported a patent to JAK2V617F licensed "Quiagen." V. Lallemand-Breitenbach reported a pending patent to WO2018134260. J. Kiladjian reported personal fees from AOP Orphan during the conduct of the study and personal fees from Novartis outside the submitted work. In addition, J. Kiladjian had a pending patent to WO2018134260 naming Hugues de Thé, Bruno Cassinat, Valérie Lallemand-Breitenbach, Isabelle Plo, Jean-Luc Villeval, and Jean-Jacques Kiladjian as inventors. H. de Thé reported personal fees from Vectorlab outside the submitted work; in addition, H. de Thé had a pending patent to WO2018134260. No other disclosures were reported.

© 2020 Dagher et al.

Figures

Graphical abstract
Graphical abstract
Figure 1.
Figure 1.
IFNα+ATO enforces potent NB formation in JAK2V617F cells.(a) Representative immunofluorescence images (upper panel) and quantification of PML-NB numbers and intensities (lower panel) in CD34+ cells from a healthy donor (control) and a JAK2V617F MF patient, not treated (NT) or treated in vitro with ATO (1 µM) for 1 h, IFNα (1,000 IU/ml) for 24 h, or both (lower panel represents the pooled results of three patients and controls). Two-tailed unpaired Student’s t test. Scale bars = 5 µm. (b) Same as panel a in LK cells from Jak2V617F KI mice (VF) or Jak2WT littermates (WT) not treated or treated in vitro with ATO (0.3 µM) for 1 h, IFNα (1,000 IU/ml) for 22 h, or both (n = 6). Scale bars = 10 µm. (c) Western blot analysis of PML in LK cells from Jak2V617F KI, Jak2WT littermate, or Pml−/ mice not treated or treated with ATO for 1 h, IFNα for 22 h, or both (n = 3). The values shown are mean ± SEM; **P< 0.01; ***P< 0.001; ****P< 0.0001.
Figure S1.
Figure S1.
Arsenic potently enhances NB formation and IFN-driven growth suppression in JAK2V617F-expressing cells. (a) Representative immunofluorescence images (upper panel) and quantification of PML-NB numbers and intensities per cell (lower panel) in JAK2WT (WT) and JAK2V617F (VF) UT-7 cells untreated (NT) or treated with ATO (0.5 µM) for 2 h, IFNα (1,000 IU/ml) for 24 h, or the combination of both drugs in these conditions. Three independent experiments with at least 20 cells analyzed per condition. Two-tailed unpaired Student’s t test. Scale bars = 5 µm. (b) Proliferation of JAK2WT (WT) or JAK2V617F (VF) UT-7 cells after 72 h of treatment with increasing doses of ATO alone or combined with IFNα 200 IU/ml. Three independent experiments. (c) Top panels: example of cell cycle analysis (BrdU/DAPI staining) of either JAK2WT (WT) or JAK2V617F (VF) UT-7 cells 1 d after being untreated or treated with ATO (0.25 µM), IFNα (1,000 IU/ml), or the combination of both. Bottom plot: summary and statistical analysis of three independent experiments. (d) Absolute numbers of JAK2WT/WT (WT), JAK2WT/V617F (WT/VF), or JAK2V617F/V617F (VF) colonies counted after 14 d of culture of CD34+ cells from seven PV patients. The effect of increasing doses of IFNα (200 IU/ml, 500 IU/ml, and 1,000 IU/ml) is shown on the upper panel, and the effect of increasing doses of IFNα (0 IU/ml, 200 IU/ml, 500 IU/ml, and 1,000 IU/ml) in combination with a constant dose of ATO (0.1 µM) is shown on the lower panel. Seven patient samples in duplicate cultures. Two-tailed unpaired Student’s t test. (e) Percentages of erythroid colonies relative to untreated controls after 14 d of culture of CD34+ cells from five healthy donors untreated or treated with ATO (0.1 µM), IFNα (200 IU/ml), or both drugs in triplicate cultures. Two-tailed unpaired Student’s t test. The values shown are mean ± SEM. *P< 0.05; **P< 0.01; ***P< 0.001; ****P< 0.0001; ns, not significant; CFC, colony-forming cell.
Figure 2.
Figure 2.
Arsenic enhances IFNα-driven growth suppression in JAK2V617F cells in vitro.(a) Relative proliferation of JAK2WT (WT) UT-7 cells or UT-7 cells expressing ectopic JAK2V617F (VF) treated with ATO (0.25 µM), IFNα (1,000 IU/ml), or both for 3 d (n = 3). (b) Numbers of JAK2WT (WT, light shade) and JAK2V617F-positive (VF, dark shade) erythroid colonies from CD34+ patient cells cultured for 14 d. Triplicate cultures from 10 JAK2V617F MF patients untreated (NT) or treated with ATO (0.1 µM), IFNα (200 IU/ml), or both. (c) Numbers of JAK2WT/WT (WT; light shade), JAK2WT/V617F (WT/VF; patterned), and JAK2V617F/V617F (VF; dark shade) erythroid and myeloid colonies counted from CD34+ patient cells cultured for 14 d. Results represent duplicate cultures from seven PV patients untreated or treated with ATO (0.1 µM), IFNα (1,000 IU/ml), or both. (d) Erythroid colonies formed from CD34+ cells isolated from a PV patient taken after 14 d of culture: untreated or treated with ATO (0.1 µM), IFNα (200 or 1,000 IU/ml), or both. Scale bars = 50 µm. (e) Numbers of erythroid and myeloid colonies after 7 d of culture of 5 × 104 BM cells collected from Jak2V617F; UbiGFP KI mice (left), from Jak2WT littermate mice (right), or a 1:1 mixture from both, mimicking the situation found in patients (bottom). Cultures were untreated or treated with ATO (0.1 µM), IFNα (200 or 1,000 IU/ml), or both. Three independent experiments (except for ATO n = 1) treated with ATO (0.2 µM), IFNα (200 or 1,000 IU/ml as indicated) with or without ATO. Two-way ANOVA. Error bars represent the mean ± SEM. *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001. ns, not significant; CFC, colony-forming cell.
Figure S2.
Figure S2.
Experimental design of the preclinical MPN mouse model. Recipient mice were lethally irradiated and transplanted with 3 × 106 40:60 mix of Jak2V617F/UbiGFP and Jak2WT BM cells. 4 wk after BM transplantation (BMT), mice were treated with ATO (5 mg/kg/2 d), IFNα (3 × 104 IU/d), or both for 8 wk. At completion of treatment, some mice were sacrificed, and their BM cells were used to assess disease-initiating cells in secondary transplantations, also with 3 × 106 BM cells; the remaining mice were kept alive to assess disease relapse around 9 wk without further treatment.
Figure 3.
Figure 3.
The IFNα+ATO combination efficiently targets Jak2V617F disease in vivo.(a) Hematological parameters of mice bearing Jak2V617F disease were obtained from four independent experiments (untreated, n = 25; ATO, n = 19; IFNα, n = 28; and IFNα+ATO, n = 29; Fig. S2 for protocol details). One-way ANOVA with multiple comparisons. (b) Percentage of GFP-positive Jak2V617F Gr-1+/CD11b+ cells (granulocytes), CD42+ cells (platelets), and Ter119+ cells (RBCs) in chimeric recipients during treatment. (c) Spleen weight after treatment discontinuation obtained from four independent experiments (untreated [NT], n = 12; ATO, n = 8; IFNα, n = 11; and IFN+ATO, n = 19). Unpaired two-sided Student’s t test. (d) Percentage of Jak2V617F cells (untreated, n = 5; ATO, n = 4; IFNα, n = 4; and IFNα+ATO, n = 9). Unpaired two-sided Student’s t test. (e) Percentage of healthy secondary recipients evaluated at 8–15 wk after BM transplantation (four independent experiments: untreated, n = 17; ATO, n = 12; IFNα, n = 22; and IFNα + ATO, n = 18). Mice were considered healthy based on the hematocrit (<50%) and the absence of Jak2V617F cells (GFP labeled) in granulocytes (Gr-1+/CD11b+) and platelets (CD42+). One-way ANOVA with multiple comparisons. (f) Percentage of relapses 9 wk after treatment completion in primary recipients. Results from four independent experiments (untreated, n = 11; ATO, n = 10; IFNα, n = 15; and IFNα + ATO, n = 15). One-way ANOVA with multiple comparisons. The values shown are mean ± SEM. *P< 0.05; **P< 0.01; ***P< 0.001.
Figure 4.
Figure 4.
PML drives IFNα+ATO response.(a) Absolute numbers of JAK2WT (WT) and JAK2V617F (VF) erythroid colonies after 14 d of culture of shSCR–treated (PML expressing) or shPML-treated (PML down-regulated) CD34+ cells from four JAK2V617F MF patients untreated (NT) or treated with the combination of IFNα (200 IU/ml) and ATO (0.1 µM). Unpaired two-sided Student’s t test. (b) Lethally irradiated mice were grafted with 50% BM cells from Jak2V617F KI/UbiGFP mice and 50% BM cells from Jak2V617F KI Pml−/− mice. Mice were treated with IFNα alone or IFNα+ATO 4 wk after transplantation. (c) Blood parameters (WBCs, platelets, and hematocrit) every 2 wk during the treatment. n = 5 for each treatment; one-way ANOVA with multiple comparisons. (d) Percentage of Pml−/− Gr-1+ cells (granulocytes), Pml−/− CD42+ cells (platelets), and Pml−/− Ter119+ cells (RBCs). n = 5 for each treatment; one-way ANOVA with multiple comparisons. Color code as in panel c. The values shown are mean ± SEM. **P< 0.01; ***P< 0.001; ****P< 0.0001. BMT, BM transplantation.
Figure S3.
Figure S3.
IFN/ATO drives senescence more efficiently in the JAK2V617F context.(a) Percentages of JAK2WT (WT) or JAK2V617F (VF) UT-7 cells untreated (NT) or treated with ATO (0.25 µM), IFNα (1,000 IU/ml), or both drugs for 7 d stained positively for SA-β-Gal. (b) Relative expression to untreated control of the senescence-associated genes PML, PAI1, and CDKN1A in UT-7 cells. Color code as in panel a. Error bars represent mean ± SEM of three independent experiments; unpaired two-sided Student’s t test. (c) SA-β-Gal staining assay of CD34+-derived cells from three MF patients or three healthy controls after 10 d of megakaryocytic differentiation and untreated or treated with the combination of IFNα (1,000 IU/ml) and ATO (0.25 µM). The values shown are mean ± SEM. *P< 0.05; **P< 0.01; ***P< 0.001.
Figure 5.
Figure 5.
IFNα+ATO drives PML-dependent senescence.(a) Percentages of SA-β-Gal+ cells after CD34+ cells from three healthy donors’ (control) and three JAK2V617F MF patients’ samples (patient) were treated with ATO (0.25 µM), IFNα (1,000 IU/ml), or both for 10 d. Unpaired two-sided Student’s t test. (b) Relative expression to untreated control of senescence-associated genes PML, PAI1, CDKN1A, and GDF15 of patient samples described in panel a. Color code as in panel a. Unpaired two-sided Student’s t test. (c) SA-β-Gal staining of shSCR–treated (PML expressing) or shPML-treated (PML down-regulated) CD34+-derived cells from two MF patients after 10 d of megakaryocytic differentiation and untreated (NT) or treated with the combination IFNα (1,000 IU/ml) and ATO (0.25 µM). (d) Percentages of SA-β-Gal+ cells of shSCR–treated (PML-expressing) or shPML-treated (PML down-regulated) CD34+-derived cells from three MF patient samples after 10 d of megakaryocytic differentiation untreated or treated with the combination IFNα (1,000 IU/ml) and ATO (0.25 µM). (e) Relative expression of PML, PAI1, CDKN1A, and GDF15 of samples described in panel d. Unpaired two-sided Student’s t test. (f) Expression of Cdkn2a, Pai1, and Gdf15 in mouse Lin− BM cells derived from Jak2V617F KI (VF) or littermate Jak2WT (WT) mice deficient (hatched bars) or not (not hatched bars) in Pml treated in vitro with IFNα (1,000 IU/ml)+ATO (0.3 µM) for 24 h. The values shown are mean ± SEM. *P< 0.05; **P< 0.01; ***P< 0.001; ****P< 0.0001.

References

    1. Ablain, J., Rice K., Soilihi H., de Reynies A., Minucci S., and de Thé H.. 2014. Activation of a promyelocytic leukemia-tumor protein 53 axis underlies acute promyelocytic leukemia cure. Nat. Med. 20:167–174. 10.1038/nm.3441
    1. Amodeo, V., A D., Betts J., Bartesaghi S., Zhang Y., Richard-Londt A., Ellis M., Roshani R., Vouri M., Galavotti S., et al. . 2017. A PML/Slit Axis Controls Physiological Cell Migration and Cancer Invasion in the CNS. Cell Rep. 20:411–426. 10.1016/j.celrep.2017.06.047
    1. Austin, R.J., Straube J., Bruedigam C., Pali G., Jacquelin S., Vu T., Green J., Gräsel J., Lansink L., Cooper L., et al. . 2020. Distinct effects of ruxolitinib and interferon-alpha on murine JAK2V617F myeloproliferative neoplasm hematopoietic stem cell populations. Leukemia. 34:1075–1089. 10.1038/s41375-019-0638-y
    1. Besancenot, R., Chaligné R., Tonetti C., Pasquier F., Marty C., Lécluse Y., Vainchenker W., Constantinescu S.N., and Giraudier S.. 2010. A senescence-like cell-cycle arrest occurs during megakaryocytic maturation: implications for physiological and pathological megakaryocytic proliferation. PLoS Biol. 8:e1000476 10.1371/journal.pbio.1000476
    1. Croker, B.A., Metcalf D., Robb L., Wei W., Mifsud S., DiRago L., Cluse L.A., Sutherland K.D., Hartley L., Williams E., et al. . 2004. SOCS3 is a critical physiological negative regulator of G-CSF signaling and emergency granulopoiesis. Immunity. 20:153–165. 10.1016/S1074-7613(04)00022-6
    1. de Thé, H., Pandolfi P.P., and Chen Z.. 2017. Acute Promyelocytic Leukemia: A Paradigm for Oncoprotein-Targeted Cure. Cancer Cell. 32:552–560. 10.1016/j.ccell.2017.10.002
    1. Giorgi, C., Ito K., Lin H.K., Santangelo C., Wieckowski M.R., Lebiedzinska M., Bononi A., Bonora M., Duszynski J., Bernardi R., et al. . 2010. PML regulates apoptosis at endoplasmic reticulum by modulating calcium release. Science. 330:1247–1251. 10.1126/science.1189157
    1. Gisslinger, H., Klade C., Georgiev P., Krochmalczyk D., Gercheva-Kyuchukova L., Egyed M., Rossiev V., Dulicek P., Illes A., Pylypenko H., et al. . PROUD-PV Study Group . 2020. Ropeginterferon alfa-2b versus standard therapy for polycythaemia vera (PROUD-PV and CONTINUATION-PV): a randomised, non-inferiority, phase 3 trial and its extension study. Lancet Haematol. 7:E196–E208. 10.1016/S2352-3026(19)30236-4
    1. Hasan, S., Lacout C., Marty C., Cuingnet M., Solary E., Vainchenker W., and Villeval J.L.. 2013. JAK2V617F expression in mice amplifies early hematopoietic cells and gives them a competitive advantage that is hampered by IFNα. Blood. 122:1464–1477. 10.1182/blood-2013-04-498956
    1. Hsu, K.S., and Kao H.Y.. 2018. PML: Regulation and multifaceted function beyond tumor suppression. Cell Biosci. 8:5 10.1186/s13578-018-0204-8
    1. Ivanschitz, L., Takahashi Y., Jollivet F., Ayrault O., Le Bras M., and de Thé H.. 2015. PML IV/ARF interaction enhances p53 SUMO-1 conjugation, activation, and senescence. Proc. Natl. Acad. Sci. USA. 112:14278–14283. 10.1073/pnas.1507540112
    1. Jeanne, M., Lallemand-Breitenbach V., Ferhi O., Koken M., Le Bras M., Duffort S., Peres L., Berthier C., Soilihi H., Raught B., et al. . 2010. PML/RARA oxidation and arsenic binding initiate the antileukemia response of As2O3. Cancer Cell. 18:88–98. 10.1016/j.ccr.2010.06.003
    1. Kchour, G., Tarhini M., Kooshyar M.M., El Hajj H., Wattel E., Mahmoudi M., Hatoum H., Rahimi H., Maleki M., Rafatpanah H., et al. . 2009. Phase 2 study of the efficacy and safety of the combination of arsenic trioxide, interferon alpha, and zidovudine in newly diagnosed chronic adult T-cell leukemia/lymphoma (ATL). Blood. 113:6528–6532. 10.1182/blood-2009-03-211821
    1. Kiladjian, J.J., Cassinat B., Turlure P., Cambier N., Roussel M., Bellucci S., Menot M.L., Massonnet G., Dutel J.L., Ghomari K., et al. . 2006. High molecular response rate of polycythemia vera patients treated with pegylated interferon α-2a. Blood. 108:2037–2040. 10.1182/blood-2006-03-009860
    1. Kiladjian, J.J., Cassinat B., Chevret S., Turlure P., Cambier N., Roussel M., Bellucci S., Grandchamp B., Chomienne C., and Fenaux P.. 2008. Pegylated interferon-alfa-2a induces complete hematologic and molecular responses with low toxicity in polycythemia vera. Blood. 112:3065–3072. 10.1182/blood-2008-03-143537
    1. Kiladjian, J.J., Giraudier S., and Cassinat B.. 2016. Interferon-alpha for the therapy of myeloproliferative neoplasms: targeting the malignant clone. Leukemia. 30:776–781. 10.1038/leu.2015.326
    1. Lallemand-Breitenbach, V., and de Thé H.. 2018. PML nuclear bodies: from architecture to function. Curr. Opin. Cell Biol. 52:154–161. 10.1016/j.ceb.2018.03.011
    1. Lallemand-Breitenbach, V., Zhu J., Puvion F., Koken M., Honoré N., Doubeikovsky A., Duprez E., Pandolfi P.P., Puvion E., Freemont P., and de Thé H.. 2001. 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. 193:1361–1372. 10.1084/jem.193.12.1361
    1. Lallemand-Breitenbach, V., Jeanne M., Benhenda S., Nasr R., Lei M., Peres L., Zhou J., Zhu J., Raught B., and de Thé H.. 2008. Arsenic degrades PML or PML-RARalpha through a SUMO-triggered RNF4/ubiquitin-mediated pathway. Nat. Cell Biol. 10:547–555. 10.1038/ncb1717
    1. Lehmann-Che, J., Bally C., and de Thé H.. 2014. Resistance to therapy in acute promyelocytic leukemia. N. Engl. J. Med. 371:1171–1172.
    1. Lu, M., Xia L., Li Y., Wang X., and Hoffman R.. 2014. The orally bioavailable MDM2 antagonist RG7112 and pegylated interferon α 2a target JAK2V617F-positive progenitor and stem cells. Blood. 124:771–779. 10.1182/blood-2013-11-536854
    1. Lui, W.C., Chan Y.F., Chan L.C., and Ng R.K.. 2014. Cytokine combinations on the potential for ex vivo expansion of murine hematopoietic stem cells. Cytokine. 68:127–132. 10.1016/j.cyto.2014.04.008
    1. Marty, C., Lacout C., Droin N., Le Couédic J.P., Ribrag V., Solary E., Vainchenker W., Villeval J.L., and Plo I.. 2013. A role for reactive oxygen species in JAK2 V617F myeloproliferative neoplasm progression. Leukemia. 27:2187–2195. 10.1038/leu.2013.102
    1. Mascarenhas, J., Lu M., Kosiorek H., Virtgaym E., Xia L., Sandy L., Mesa R., Petersen B., Farnoud N., Najfeld V., et al. . 2019. Oral idasanutlin in patients with polycythemia vera. Blood. 134:525–533. 10.1182/blood.2018893545
    1. Mullally, A., Poveromo L., Schneider R.K., Al-Shahrour F., Lane S.W., and Ebert B.L.. 2012. Distinct roles for long-term hematopoietic stem cells and erythroid precursor cells in a murine model of Jak2V617F-mediated polycythemia vera. Blood. 120:166–172. 10.1182/blood-2012-01-402396
    1. Mullally, A., Bruedigam C., Poveromo L., Heidel F.H., Purdon A., Vu T., Austin R., Heckl D., Breyfogle L.J., Kuhn C.P., et al. . 2013. Depletion of Jak2V617F myeloproliferative neoplasm-propagating stem cells by interferon-α in a murine model of polycythemia vera. Blood. 121:3692–3702. 10.1182/blood-2012-05-432989
    1. Niwa-Kawakita, M., Ferhi O., Soilihi H., Le Bras M., Lallemand-Breitenbach V., and de Thé H.. 2017. PML is a ROS sensor activating p53 upon oxidative stress. J. Exp. Med. 214:3197–3206. 10.1084/jem.20160301
    1. Pasquier, F., Cabagnols X., Secardin L., Plo I., and Vainchenker W.. 2014. Myeloproliferative neoplasms: JAK2 signaling pathway as a central target for therapy. Clin. Lymphoma Myeloma Leuk. 14(Suppl):S23–S35. 10.1016/j.clml.2014.06.014
    1. Pearson, M., Carbone R., Sebastiani C., Cioce M., Fagioli M., Saito S., Higashimoto Y., Appella E., Minucci S., Pandolfi P.P., and Pelicci P.G.. 2000. PML regulates p53 acetylation and premature senescence induced by oncogenic Ras. Nature. 406:207–210. 10.1038/35018127
    1. Quignon, F., De Bels F., Koken M., Feunteun J., Ameisen J.-C., and de Thé H.. 1998. PML induces a novel caspase-independent death process. Nat. Genet. 20:259–265. 10.1038/3068
    1. Sahin, U., Ferhi O., Jeanne M., Benhenda S., Berthier C., Jollivet F., Niwa-Kawakita M., Faklaris O., Setterblad N., de Thé H., and Lallemand-Breitenbach V.. 2014. Oxidative stress-induced assembly of PML nuclear bodies controls sumoylation of partner proteins. J. Cell Biol. 204:931–945. 10.1083/jcb.201305148
    1. Salomoni, P., and Pandolfi P.P.. 2002. The role of PML in tumor suppression. Cell. 108:165–170. 10.1016/S0092-8674(02)00626-8
    1. Schaefer, B.C., Schaefer M.L., Kappler J.W., Marrack P., and Kedl R.M.. 2001. Observation of antigen-dependent CD8+ T-cell/ dendritic cell interactions in vivo. Cell. Immunol. 214:110–122. 10.1006/cimm.2001.1895
    1. Seeler, J.S., and Dejean A.. 2017. SUMO and the robustness of cancer. Nat. Rev. Cancer. 17:184–197. 10.1038/nrc.2016.143
    1. Spivak, J.L. 2019. How I treat polycythemia vera. Blood. 134:341–352. 10.1182/blood.2018834044
    1. Stadler, M., Chelbi-Alix M.K., Koken M.H.M., Venturini L., Lee C., Saïb A., Quignon F., Pelicano L., Guillemin M.-C., Schindler C., and de Thé H.. 1995. Transcriptional induction of the PML growth suppressor gene by interferons is mediated through an ISRE and a GAS element. Oncogene. 11:2565–2573.
    1. Takahashi, Y., Lallemand-Breitenbach V., Zhu J., and de Thé H.. 2004. PML nuclear bodies and apoptosis. Oncogene. 23:2819–2824. 10.1038/sj.onc.1207533
    1. Vainchenker, W., and Kralovics R.. 2017. Genetic basis and molecular pathophysiology of classical myeloproliferative neoplasms. Blood. 129:667–679. 10.1182/blood-2016-10-695940
    1. Verger, E., Soret-Dulphy J., Maslah N., Roy L., Rey J., Ghrieb Z., Kralovics R., Gisslinger H., Grohmann-Izay B., Klade C., et al. . 2018. Ropeginterferon alpha-2b targets JAK2V617F-positive polycythemia vera cells in vitro and in vivo. Blood Cancer J. 8:94 10.1038/s41408-018-0133-0
    1. Vernier, M., and Ferbeyre G.. 2014. Complete senescence: RB and PML share the task. Cell Cycle. 13:696 10.4161/cc.28090
    1. Vernier, M., Bourdeau V., Gaumont-Leclerc M.F., Moiseeva O., Bégin V., Saad F., Mes-Masson A.M., and Ferbeyre G.. 2011. Regulation of E2Fs and senescence by PML nuclear bodies. Genes Dev. 25:41–50. 10.1101/gad.1975111
    1. Zhang, X.W., Yan X.J., Zhou Z.R., Yang F.F., Wu Z.Y., Sun H.B., Liang W.X., Song A.X., Lallemand-Breitenbach V., Jeanne M., et al. . 2010. Arsenic trioxide controls the fate of the PML-RARalpha oncoprotein by directly binding PML. Science. 328:240–243. 10.1126/science.1183424
    1. Zhu, J., Koken M.H.M., Quignon F., Chelbi-Alix M.K., Degos L., Wang Z.Y., Chen Z., and de Thé H.. 1997. Arsenic-induced PML targeting onto nuclear bodies: implications for the treatment of acute promyelocytic leukemia. Proc. Natl. Acad. Sci. USA. 94:3978–3983. 10.1073/pnas.94.8.3978
    1. Zhu, H.H., Liu Y.R., Jia J.S., Qin Y.Z., Zhao X.S., and Lai Y.Y.. 2018. Oral arsenic and all-trans retinoic acid for high-risk acute promyelocytic leukemia. Blood. 131:2987–2989. 10.1182/blood-2018-02-834051
    1. Zitvogel, L., Galluzzi L., Kepp O., Smyth M.J., and Kroemer G.. 2015. Type I interferons in anticancer immunity. Nat. Rev. Immunol. 15:405–414. 10.1038/nri3845

Source: PubMed

Подписаться