Biomodulatory Treatment With Azacitidine, All- trans Retinoic Acid and Pioglitazone Induces Differentiation of Primary AML Blasts Into Neutrophil Like Cells Capable of ROS Production and Phagocytosis

Sebastian Klobuch, Tim Steinberg, Emanuele Bruni, Carina Mirbeth, Bernhard Heilmeier, Lina Ghibelli, Wolfgang Herr, Albrecht Reichle, Simone Thomas, Sebastian Klobuch, Tim Steinberg, Emanuele Bruni, Carina Mirbeth, Bernhard Heilmeier, Lina Ghibelli, Wolfgang Herr, Albrecht Reichle, Simone Thomas

Abstract

Effective and tolerable salvage therapies for elderly patients with chemorefractory acute myeloid leukemia (AML) are limited and usually do not change the poor clinical outcome. We recently described in several chemorefractory elderly AML patients that a novel biomodulatory treatment regimen consisting of low-dose azacitidine (AZA) in combination with PPARγ agonist pioglitazone (PGZ) and all-trans retinoic acid (ATRA) induced complete remission of leukemia and also triggered myeloid differentiation with rapid increase of peripheral blood neutrophils. Herein, we further investigated our observations and comprehensively analyzed cell differentiation in primary AML blasts after treatment with ATRA, AZA, and PGZ ex vivo. The drug combination was found to significantly inhibit cell growth as well as to induce cell differentiation in about half of primary AML blasts samples independent of leukemia subtype. Notably and in comparison to ATRA/AZA/PGZ triple-treatment, effects on cell growth and myeloid differentiation with ATRA monotherapy was much less efficient. Morphological signs of myeloid cell differentiation were further confirmed on a functional basis by demonstrating increased production of reactive oxygen species as well as enhanced phagocytic activity in AML blasts treated with ATRA/AZA/PGZ. In conclusion, we show that biomodulatory treatment with ATRA/AZA/PGZ can induce phenotypical and functional differentiation of primary AML blasts into neutrophil like cells, which aside from its antileukemic activity may lower neutropenia associated infection rates in elderly AML patients in vivo. Clinical impact of the ATRA/AZA/PGZ treatment regimen is currently further investigated in a randomized clinical trial in chemorefractory AML patients (NCT02942758).

Keywords: acute myeloid leukemia; all-trans retinoic acid; azacitidine; differentiation; pioglitazone.

Figures

FIGURE 1
FIGURE 1
Treatment of HL-60 and U937 cells. (A) HL-60 cells (1.2 × 106) were incubated in RPMI supplemented with 10% FCS and as indicated with DMSO (open circles), 1 μM ATRA (closed circle), 5 μM PGZ (open square), 0.1 μM AZA (closed square), ATRA/PGZ (open diamond), ATRA/AZA (closed diamond), PGZ/AZA (open triangle), or ATRA/AZA/PGZ (closed triangle). On days 3, 5, and 7 during treatment, cells were counted by trypan blue staining. Data represent means of cell numbers from three independent experiments. Error bars indicate the standard deviation. (B) HL-60 cells were treated as described in (A). On day 5 of treatment, cells were characterized for surface-expression of the differentiation marker CD11b by flow cytometry. Shown are relative MFI values calculated by division of the MFI of antigen staining by the MFI of DMSO staining. Data represent means and standard deviations of three independent experiments. p-Values are shown for samples with significant differences in comparison to DMSO controls. Statistical analysis for group differences were performed using Friedman and Dunn’s multiple comparisons test. (C,D) U937 cells were incubated in RPMI supplemented with 10% FCS and as indicated with DMSO (open circles), 1 μM ATRA (closed circle), 5 μM PGZ (open square), 0.1 μM AZA (closed square), ATRA/PGZ (open diamond), ATRA/AZA (closed diamond), PGZ/AZA (open triangle), or ATRA/AZA/PGZ (closed triangle). From day 0 to day 4 during treatment, cells were analyzed for cell growth (C) and apoptosis (D). Data represent means from two independent experiments. Error bars indicate the standard deviation.
FIGURE 2
FIGURE 2
Cell growth and apoptosis of primary AML blasts. (A,B) Primary AML blasts (5 × 105) were incubated in AIM-V medium supplemented with 10% HS, G-CSF and SCF (each 50 ng/mL) and as indicated with medium, DMSO, ATRA, or ATRA/AZA/PGZ. On 14 of treatment, cell numbers were measured by conventional trypan blue staining. (A) Shows cell numbers normalized to medium (n = 14 per group), (B) shows Annexin positive cells normalized to medium (medium = 100%; n = 14 per group). Symbols represent individual patient samples and horizontal bars mark median values. Statistical analysis for group differences were performed using Friedman and Dunn’s multiple comparisons test. (C) Primary AML blasts (5 × 105) of patient sample AML144 (left panel) or AML111 (right panel) were incubated as described in (A). On days 7 and 14 of treatment, cell numbers were measured by conventional trypan blue staining. (D) AML blasts as described in (A) were characterized by Annexin V binding 14 days after treatment. Shown are % Annexin positive cells of AML111 (white bars) and AML144 (black bars).
FIGURE 3
FIGURE 3
Morphological differentiation of primary AML blasts. (A) Primary AML blasts (5 × 105) were treated as described in Figure 2. On day 14 after start of treatment, cells (5 × 104) were analyzed by light microscopy after centrifugation on microscope slides and May-Grünwald and Giemsa staining. Shown are AML cells of sample AML101 after treatment with ATRA/AZA/PGZ (I), ATRA (II), DMSO (III), or medium (IV), respectively. (B) Summary of 12 primary AML patient’s samples on day 14 of treatment as described in (A). Cells with morphological signs of differentiation were normalized to medium controls. Symbols represent individual patient samples and horizontal bars mark median values. (C) On day 14 of treatment with ATRA, PGZ, AZA, ATRA/AZA, ATRA/PGZ, PGZ/AZA, ATRA/AZA/PGZ, or DMSO sample AML101 was analyzed for morphological changes as described in (A). Cells with morphological signs of differentiation were normalized to medium controls. Data represent means of morphological changes from two independent experiments. Error bars indicate the standard deviation. (D) Primary AML samples (n = 14) at day 14 of treatment. Shown are the relative MFIs of CD11b staining normalized to medium. Symbols represent individual patient samples and horizontal bars mark median values. Statistical analysis for group differences were performed using Friedman and Dunn’s multiple comparisons test.
FIGURE 4
FIGURE 4
Reactive oxygen species production and phagocytosis of AML blasts. (A) Primary AML blasts were treated as described in Figure 2. On day 14 of treatment, 5 × 105 AML blasts were incubated with NBT (1 mg/mL) and PMA (200 ng/mL) for 90 min. Subsequently, cells were centrifuged on microscope slides for light microscopy. Shown are AML blasts of sample AML101 after treatment with ATRA/AZA/PGZ (I), ATRA (II), DMSO (III), or medium (IV), respectively. (B) Summary of primary AML samples (n = 11) on day 14 days of treatment as described in (A). NBT positive cells were counted in a blinded fashion and were normalized to medium. Symbols represent individual patient samples and horizontal bars mark median values. Statistical analysis for group differences were performed using Friedman and Dunn’s multiple comparisons test. (C,D) Primary AML blasts were treated as described in Figure 2. On day 14 of treatment, 2.5 × 105 AML blasts (n = 11 patients) were incubated with 6.25 × 106 GFP-labeled E. coli bacteria for 10 min. Subsequently, cells were analyzed by flow cytometry for GFP positive cells. (C) Summary of data (n = 11 AML samples) representing MFIs of GFP signal normalized to medium controls. Symbols represent individual patient samples and horizontal bars mark median values. Statistical analysis for group differences were performed using Friedman and Dunn’s multiple comparisons test. (D) Individual overlay plots and MFI of GFP signals measured with AML101 and AML147.
FIGURE 5
FIGURE 5
Biomodulatory treatment of AML cell lines in combination with midostaurin. (A) MV4-11 and (B) MOLM-13 cells were incubated in RPMI supplemented with 10% FCS and treated with single agent DMSO, ATRA, AZA, PGZ, MIDO or combinations thereof. On day 3 during treatment cells were counted by trypan blue staining. Data represent means of cell numbers from three independent experiments. Error bars indicate the standard deviation. Statistical analysis for group differences compared to DMSO were performed using Friedman and Dunn’s multiple comparisons test. ns p > 0.05, ∗p ≤ 0.05, ∗∗p ≤ 0.01, ∗∗∗p ≤ 0.001, ∗∗∗∗p ≤ 0.0001.

References

    1. Boyd A. L., Reid J. C., Salci K. R., Aslostovar L., Benoit Y. D., Shapovalova Z., et al. (2017). Acute myeloid leukaemia disrupts endogenous myelo-erythropoiesis by compromising the adipocyte bone marrow niche. Nat. Cell Biol. 19 1336–1347. 10.1038/ncb3625
    1. Burnett A. K., Hills R. K., Green C., Jenkinson S., Koo K., Patel Y., et al. (2010). The impact on outcome of the addition of all-trans retinoic acid to intensive chemotherapy in younger patients with nonacute promyelocytic acute myeloid leukemia: overall results and results in genotypic subgroups defined by mutations in NPM1, FLT3, and CEBPA. Blood 115 948–956. 10.1182/blood-2009-08-236588
    1. Cicconi L., Divona M., Ciardi C., Ottone T., Ferrantini A., Lavorgna S., et al. (2016). PML-RARα kinetics and impact of FLT3-ITD mutations in newly diagnosed acute promyelocytic leukaemia treated with ATRA and ATO or ATRA and chemotherapy. Leukemia 30 1987–1992. 10.1038/leu.2016.122
    1. Dini L., Coppola S., Ruzittu M. T., Ghibelli L. (1996). Multiple pathways for apoptotic nuclear fragmentation. Exp. Cell Res. 223 340–347. 10.1006/excr.1996.0089
    1. Döhner H., Estey E. H., Amadori S., Appelbaum F. R., Büchner T., Burnett A. K., et al. (2010). Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood 115 453–474. 10.1182/blood-2009-07-235358
    1. Dombret H., Seymour J. F., Butrym A., Wierzbowska A., Selleslag D., Jang J. H., et al. (2015). International phase 3 study of azacitidine vs conventional care regimens in older patients with newly diagnosed AML with 30% blasts. Blood 126 291–299. 10.1182/blood-2015-01-621664
    1. Estey E. H., Giles F. J., Kantarjian H., O’Brien S., Cortes J., Freireich E. J., et al. (1999). Molecular remissions induced by liposomal-encapsulated all-trans retinoic acid in newly diagnosed acute promyelocytic leukemia. Blood 94 2230–2235.
    1. Faber K., Bullinger L., Ragu C., Garding A., Mertens D., Miller C., et al. (2013). CDX2-driven leukemogenesis involves KLF4 repression and deregulated PPARgamma signaling. J. Clin. Invest. 123 299–314. 10.1172/JCI64745
    1. Flotho C., Claus R., Batz C., Schneider M., Sandrock I., Ihde S., et al. (2009). The DNA methyltransferase inhibitors azacitidine, decitabine and zebularine exert differential effects on cancer gene expression in acute myeloid leukemia cells. Leukemia 23 1019–1028. 10.1038/leu.2008.397
    1. Fujiki A., Imamura T., Sakamoto K., Kawashima S., Yoshida H., Hirashima Y., et al. (2012). All-trans retinoic acid combined with 5-Aza-2’-deoxycitidine induces C/EBPalpha expression and growth inhibition in MLL-AF9-positive leukemic cells. Biochem. Biophys. Res. Commun. 428 216–223. 10.1016/j.bbrc.2012.09.131
    1. Ghibelli L., Nosseri C., Coppola S., Maresca V., Dini L. (1995). The increase in H2O2-induced apoptosis by ADP-ribosylation inhibitors is related to cell blebbing. Exp. Cell Res. 221 470–477. 10.1006/excr.1995.1398
    1. Gille C., Spring B., Tewes L., Poets C. F., Orlikowsky T. (2006). A new method to quantify phagocytosis and intracellular degradation using green fluorescent protein-labeled Escherichia coli: comparison of cord blood macrophages and peripheral blood macrophages of healthy adults. Cytometry A 69 152–154. 10.1002/cyto.a.20222
    1. Gupta K., Gulen F., Sun L., Aguilera R., Chakrabarti A., Kiselar J., et al. (2012). GSK3 is a regulator of RAR-mediated differentiation. Leukemia 26 1277–1285. 10.1038/leu.2012.2
    1. Hollenbach P. W., Nguyen A. N., Brady H., Williams M., Ning Y., Richard N., et al. (2010). A comparison of azacitidine and decitabine activities in acute myeloid leukemia cell lines. PLoS One 5:e9001. 10.1371/journal.pone.0009001
    1. Hong C.-W. (2017). Current understanding in neutrophil differentiation and heterogeneity. Immune Netw. 17 298–306. 10.4110/in.2017.17.5.298
    1. Kiziltepe T., Hideshima T., Catley L., Raje N., Yasui H., Shiraishi N., et al. (2007). 5-Azacytidine, a DNA methyltransferase inhibitor, induces ATR-mediated DNA double-strand break responses, apoptosis, and synergistic cytotoxicity with doxorubicin and bortezomib against multiple myeloma cells. Mol. Cancer Ther. 6 1718–1727. 10.1158/1535-7163.MCT-07-0010
    1. Konopleva M., Elstner E., McQueen T. J., Tsao T., Sudarikov A., Hu W., et al. (2004). Peroxisome proliferator-activated receptor gamma and retinoid X receptor ligands are potent inducers of differentiation and apoptosis in leukemias. Mol. Cancer Ther. 3 1249–1262.
    1. Ley T. J., DeSimone J., Anagnou N. P., Keller G. H., Humphries R. K., Turner P. H., et al. (1982). 5-azacytidine selectively increases gamma-globin synthesis in a patient with beta+ thalassemia. N. Engl. J. Med. 307 1469–1475. 10.1056/NEJM198212093072401
    1. Liu Y. L., Yan Y., Webster C., Shao L., Lensing S. Y., Ni H., et al. (2016). Timing of the loss of Pten protein determines disease severity in a mouse model of myeloid malignancy. Blood 127 1912–1922. 10.1182/blood-2015-05-646216
    1. Lo-Coco F., Avvisati G., Vignetti M., Thiede C., Orlando S. M., Iacobelli S., et al. (2013). Retinoic acid and arsenic trioxide for acute promyelocytic leukemia. N. Engl. J. Med. 369 111–121. 10.1056/NEJMoa1300874
    1. Lübbert M., Kuendgen A. (2015). Combining DNA methyltransferase and histone deacetylase inhibition to treat acute myeloid leukemia/myelodysplastic syndrome: achievements and challenges. Cancer 121 498–501. 10.1002/cncr.29083
    1. Ma H. S., Greenblatt S. M., Shirley C. M., Duffield A. S., Bruner J. K., Li L., et al. (2016). All-trans retinoic acid synergizes with FLT3 inhibition to eliminate FLT3/ITD+ leukemia stem cells in vitro and in vivo. Blood 127 2867–2878. 10.1182/blood-2015-05-646786
    1. Marcucci G., Silverman L., Eller M., Lintz L., Beach C. L. (2005). Bioavailability of azacitidine subcutaneous versus intravenous in patients with the myelodysplastic syndromes. J. Clin. Pharmacol. 45 597–602. 10.1177/0091270004271947
    1. Milligan D. W., Wheatley K., Littlewood T., Craig J. I. O., Burnett A. K. (2006). Fludarabine and cytosine are less effective than standard ADE chemotherapy in high-risk acute myeloid leukemia, and addition of G-CSF and ATRA are not beneficial: results of the MRC AML-HR randomized trial. Blood 107 4614–4622. 10.1182/blood-2005-10-4202
    1. Noguera N. I., Piredda M. L., Taulli R., Catalano G., Angelini G., Gaur G., et al. (2016). PML/RARa inhibits PTEN expression in hematopoietic cells by competing with PU.1 transcriptional activity. Oncotarget 7 66386–66397. 10.18632/oncotarget.11964
    1. Palazzolo A. M., Suquet C., Konkel M. E., Hurst J. K. (2005). Green fluorescent protein-expressing Escherichia coli as a selective probe for HOCl generation within neutrophils. Biochemistry 44 6910–6919. 10.1021/bi047342s
    1. Patel L., Pass I., Coxon P., Downes C. P., Smith S. A., Macphee C. H. (2001). Tumor suppressor and anti-inflammatory actions of PPARgamma agonists are mediated via upregulation of PTEN. Curr. Biol. 11 764–768.
    1. Prost S., Relouzat F., Spentchian M., Ouzegdouh Y., Saliba J., Massonnet G., et al. (2015). Erosion of the chronic myeloid leukaemia stem cell pool by PPARgamma agonists. Nature 525 380–383. 10.1038/nature15248
    1. Raffoux E., Cras A., Recher C., Boelle P.-Y., Labarthe A., de Turlure P., et al. (2010). Phase 2 clinical trial of 5-azacitidine, valproic acid, and all-trans retinoic acid in patients with high-risk acute myeloid leukemia or myelodysplastic syndrome. Oncotarget 1 34–42. 10.18632/oncotarget.100518
    1. Saiki M., Hatta Y., Yamazaki T., Itoh T., Enomoto Y., Takeuchi J., et al. (2006). Pioglitazone inhibits the growth of human leukemia cell lines and primary leukemia cells while sparing normal hematopoietic stem cells. Int. J. Oncol. 29 437–443.
    1. Schaft N., Dörrie J., Müller I., Beck V., Baumann S., Schunder T., et al. (2006). A new way to generate cytolytic tumor-specific T cells: electroporation of RNA coding for a T cell receptor into T lymphocytes. Cancer Immunol. Immunother. 55 1132–1141. 10.1007/s00262-005-0098-2
    1. Schenk T., Chen W. C., Göllner S., Howell L., Jin L., Hebestreit K., et al. (2012). Inhibition of the LSD1 (KDM1A) demethylase reactivates the all-trans-retinoic acid differentiation pathway in acute myeloid leukemia. Nat. Med. 18 605–611. 10.1038/nm.2661
    1. Schlenk R. F., Döhner K., Kneba M., Götze K., Hartmann F., Del Valle F., et al. (2009). Gene mutations and response to treatment with all-trans retinoic acid in elderly patients with acute myeloid leukemia. Results from the AMLSG Trial AML HD98B. Haematologica 94 54–60. 10.3324/haematol.13378
    1. Schlenk R. F., Lübbert M., Benner A., Lamparter A., Krauter J., Herr W., et al. (2016). All-trans retinoic acid as adjunct to intensive treatment in younger adult patients with acute myeloid leukemia: results of the randomized AMLSG 07-04 study. Ann. Hematol. 95 1931–1942. 10.1007/s00277-016-2810-z
    1. Soriano A. O., Yang H., Faderl S., Estrov Z., Giles F., Ravandi F., et al. (2007). Safety and clinical activity of the combination of 5-azacytidine, valproic acid, and all-trans retinoic acid in acute myeloid leukemia and myelodysplastic syndrome. Blood 110 2302–2308. 10.1182/blood-2007-03-078576
    1. Stresemann C., Bokelmann I., Mahlknecht U., Lyko F. (2008). Azacytidine causes complex DNA methylation responses in myeloid leukemia. Mol. Cancer Ther. 7 2998–3005. 10.1158/1535-7163.MCT-08-0411
    1. Taylor S. M., Constantinides P. A., Jones P. A. (1984). 5-Azacytidine, DNA methylation, and differentiation. Curr. Top. Microbiol. Immunol. 108115–127.
    1. Thomas S., Schelker R., Klobuch S., Zaiss S., Troppmann M., Rehli M., et al. (2015). Biomodulatory therapy induces complete molecular remission in chemorefractory acute myeloid leukemia. Haematologica 100 e4–e6. 10.3324/haematol.2014.115055
    1. Verhagen H. J., Smit M. A., Rutten A., Denkers F., Poddighe P. J., Merle P. A., et al. (2016). Primary acute myeloid leukemia cells with overexpression of EVI-1 are sensitive to all-trans retinoic acid. Blood 127 458–463. 10.1182/blood-2015-07-653840
    1. Wald D. N., Vermaat H. M., Zang S., Lavik A., Kang Z., Peleg G., et al. (2008). Identification of 6-benzylthioinosine as a myeloid leukemia differentiation-inducing compound. Cancer Res. 68 4369–4376. 10.1158/0008-5472.CAN-07-6559

Source: PubMed

3
Subscribe