Ponatinib promotes a G1 cell-cycle arrest of merlin/NF2-deficient human schwann cells

Alejandra M Petrilli, Jeanine Garcia, Marga Bott, Stephani Klingeman Plati, Christine T Dinh, Olena R Bracho, Denise Yan, Bing Zou, Rahul Mittal, Fred F Telischi, Xue-Zhong Liu, Long-Sheng Chang, D Bradley Welling, Alicja J Copik, Cristina Fernández-Valle, Alejandra M Petrilli, Jeanine Garcia, Marga Bott, Stephani Klingeman Plati, Christine T Dinh, Olena R Bracho, Denise Yan, Bing Zou, Rahul Mittal, Fred F Telischi, Xue-Zhong Liu, Long-Sheng Chang, D Bradley Welling, Alicja J Copik, Cristina Fernández-Valle

Abstract

Neurofibromatosis type 2 (NF2) is a genetic syndrome that predisposes individuals to multiple benign tumors of the central and peripheral nervous systems, including vestibular schwannomas. Currently, there are no FDA approved drug therapies for NF2. Loss of function of merlin encoded by the NF2 tumor suppressor gene leads to activation of multiple mitogenic signaling cascades, including platelet-derived growth factor receptor (PDGFR) and SRC in Schwann cells. The goal of this study was to determine whether ponatinib, an FDA-approved ABL/SRC inhibitor, reduced proliferation and/or survival of merlin-deficient human Schwann cells (HSC). Merlin-deficient HSC had higher levels of phosphorylated PDGFRα/β, and SRC than merlin-expressing HSC. A similar phosphorylation pattern was observed in phospho-protein arrays of human vestibular schwannoma samples compared to normal HSC. Ponatinib reduced merlin-deficient HSC viability in a dose-dependent manner by decreasing phosphorylation of PDGFRα/β, AKT, p70S6K, MEK1/2, ERK1/2 and STAT3. These changes were associated with decreased cyclin D1 and increased p27Kip1levels, leading to a G1 cell-cycle arrest as assessed by Western blotting and flow cytometry. Ponatinib did not modulate ABL, SRC, focal adhesion kinase (FAK), or paxillin phosphorylation levels. These results suggest that ponatinib is a potential therapeutic agent for NF2-associated schwannomas and warrants further in vivo investigation.

Keywords: PDGFR; SRC; STAT3; neurofibromatosis type 2; schwannoma.

Conflict of interest statement

CONFLICTS OF INTEREST

The authors reported no potential conflicts of interest.

Figures

Figure 1. Ponatinib decreases HSC viability
Figure 1. Ponatinib decreases HSC viability
(A) Characterization of primary HSC. Confocal Images of HSC expressing human and SC linage markers: human nuclear antigen (HNA, red), S100 (green), GAP43 (red), proteolipid protein (PLP, green), negative nestin (red), DAPI stained nuclei (blue) and F-actin was visualized with phaloidin-Alexa633 (white). Scale bar: 50μm. (B) Representative Western blots of primary HSC and merlin-deficient HSC (MD-HSC) lysates, merlin silencing increased levels of phosphorylated SRC and PDGFRα/β. (C) Western blotting for merlin and β-actin in control HSC and merlin deficient cells at increasing cell passages. (D) Western blot for merlin and β-actin in three control HSC lines and four merlin-deficient (knock-down) HSC lines. (E) Ponatinib dose-response CellTiter-Fluor viability assay. Control HSC lines: SCR-HSC, GFP-HSC and WT-HSC and merlin-deficient HSC: #45, 74, 75, 77 were treated with increasing concentrations of ponatinib in constant 0.1% DMSO or vehicle alone for 48h. Viability is presented as a % of the DMSO control. Graph represents the mean ± SEM of three independent experiments. (F-G) WT-HSC and MD-HSC (#45) were maintained in serum free medium for 5-10 days and then treated with 0.25μM ponatinib for one week. Relative cell numbers was assessed using a crystal violet assay: (F) Representative 20X phase contrast images of cells. Scale bar= 100μm. (G) Cell viability calculated as a % of their respective DMSO control. Graph represent mean ± SEM of three independent experiments (** p<0.01, unpaired t-test, two tailed). (H) Viability of primary human VS cells treated for 48h with increasing ponatinib concentrations. Relative cell viability was assessed using a crystal violet assay and presented as % viability normalized to DMSO group. Plot of mean ± SEM of 6 replicates. VS1 (heterozygous deletion of 23 nucleotides in exon 8 of the NF2 gene, non-irradiated, passage 2); VS2 (heterozygous missense c.1460T>A and p. I487N in exon 14 of the NF2 gene, non-irradiated, passage 2).
Figure 2. Ponatinib decreased merlin-deficient HSC viability…
Figure 2. Ponatinib decreased merlin-deficient HSC viability independent of ABL/SRC/FAK pathway inhibition
Representative ponatinib dose-response Western blots (n=3). MD- HSC#45 plated in 12-well plates were treated with increasing concentrations of ponatinib for 2h as indicated. Cells were harvested, lysed, resolved by SDS-PAGE and blotted for: (A) p-ABL-Tyr452, c-ABL and β-actin as a loading control; (B) p-SRC-Tyr416 and total SRC, FYN, YES, p-FAK-Tyr576, p-FAK-Tyr577, total FAK, p-Paxillin-Tyr118, and total paxillin. The β-actin levels were used as loading controls.
Figure 3. Ponatinib inhibited PDGFRα/β phosphorylation in…
Figure 3. Ponatinib inhibited PDGFRα/β phosphorylation in merlin-deficient HSC
Representative ponatinib dose-response Western blots (n=3). MD-HSC#45 plated in 12-well plates were treated with increasing concentrations of ponatinib for 2h as indicated. Cells were harvested, lysed, resolved by SDS-PAGE and blotted for p-PDGFRα/β-Tyr849/Tyr857, p-PDGFRβ-Tyr740, p-PDGFRβ-Tyr771, p-PDGFRβ-Tyr1021 and total PDGFRα and PDGFRβ. The β-actin levels were used as loading controls.
Figure 4. Downstream signaling pathways inhibited by…
Figure 4. Downstream signaling pathways inhibited by ponatinib in merlin-deficient HSC
Western blots of extracts prepared from merlin-deficient HSC#45 treated with increasing ponatinib concentrations. Quantitation was done by fluorescence intensity analysis, normalized to β-actin, and plotted as mean ± SEM (n=3). One-way analysis of variance and Dunnett's multiple comparison post-test were used for statistical analysis (* p<0.1; ** p<0.01 and *** p<0.001). Representative Western blots of dose response experiments at 2h for: (A) p-AKT-Thr308 and AKT; (B) p-p70S6K-Thr229; (C) MEK1/2, p-MEK1/2-Ser217/Ser221, ERK1/2, and p-ERK1/2-Thr202/Tyr204; (D) p-STAT3-Tyr705, and total STAT3. (E) Ponatinib dose-responseCellTiter-Fluor viability assay. MD- HSC#45 were treated with semi-log serial dilutions of ponatinib, NSC-74859, selumetinib, perifosine in 0.1% DMSO for 48h, or vehicle alone. Viability is presented as % of the DMSO control. Graph represents the mean ± SEM of three independent experiments.
Figure 5. Ponatinib arrests merlin-deficient HSC at…
Figure 5. Ponatinib arrests merlin-deficient HSC at the G1 phase of the cell cycle
(A) Representative Western blots for Cyclin D1of lysates prepared from merlin-deficient HSC#45 treated 24h with increasing concentrations of ponatinib. Plotted below as mean ± SEM (n=3). One-way analysis of variance and Dunnett's multiple comparison post-test were used for statistical analysis (** p<0.01 and *** p<0.001). (B,C) Merlin-deficient HSC were treated with 3 and 5μM ponatinib for 24h and during the last 3h, 10μM EdU was added. Cells were harvested, labeled with live/dead fixable dye, and analyzed by flow cytometry. (B) Representative plots of the distribution of EdU- and FxCycle-labelled cells of 0.1% DMSO vehicle control and ponatinib treated cells. (C) Graph of the distribution of the cell cycle phases (gated for the live population) of all the experiments as mean ± SEM, n=4; **p<0.01 and ***p<0.001 were determined by two-way ANOVA and Bonferroni multiple comparisons post-test. (D) Representative plots of the distribution of live and dead cell population in these experiments with increasing concentrations of ponatinib as indicated.
Figure 6. Analysis of G1 regulatory proteins…
Figure 6. Analysis of G1 regulatory proteins during the cell-cycle in merlin-deficient HSC treated with ponatinib
MD-HSC were treated with 3μM ponatinib or vehicle control for 24h. Cells were harvested, fixed, and permeabilized. DNA was stained with FxCycle, and intracellular G1 regulatory proteins were immunostained and analyzed by flow cytometry. (A) Distribution plots of cells with positive/negative p27Kip1 immunostain vs DNA content. Data shown are representative plots of four independent experiments. (B) Distribution of cells analyzed by flow cytometry with positive/negative cyclin D1 immunostain vs DNA content. Shown are representatives of four independent experiments. (C) Diagram of signaling pathways inhibited by ponatinib in merlin-deficient HSC. Merlin deficiency leads to activation of PDGFR, SRC and PI3K. Activation of PI3K potentiates AKT and p70S6K phosphorylation and leads to cell survival, growth and G1 cell cycle progression. PDGFR activity triggers ERK and STAT3 activation, leading to cyclin D1 expression and cell proliferation. SRC activation of FAK and paxillin is not modulated by ponatinib. Ponatinib decreases cell viability through downstream inhibition of AKT, ERK, and STAT3.
Figure 7. PDGFRα/β, SRC, MEK and STAT3…
Figure 7. PDGFRα/β, SRC, MEK and STAT3 are overactive in human schwannomas
(A) Phospho-RTK membrane profile of schwannomas and control cultured primary HSC. (B) Bar graph of the quantitation ofp-PDGFRα and p-PDGFRβ membrane dot intensity normalized to positive controls. **p<0.005; ****p<0.0001 determined using unpaired t-test of control HSC vs. VS populations, two-tailed. (C) Phospho-kinase membrane profile of schwannomas and control cultured primary HSC samples. (D) Bar graph of the quantitation of phospho-SRC, phospho-MEK1/2 and phospho-STAT3 membrane intensity normalized to positive controls. *p<0.05; ***p<0.001 determined using unpaired t-test of control HSC vs. VS populations, two-tailed.

References

    1. Blakeley JO, Plotkin SR. Therapeutic advances for the tumors associated with neurofibromatosis type 1, type 2, and schwannomatosis. Neuro-oncology. 2016;18:624–38.
    1. Rouleau GA, Merel P, Lutchman M, Sanson M, Zucman J, Marineau C, Hoang-Xuan K, Demczuk S, Desmaze C, Plougastel B, et al. Alteration in a new gene encoding a putative membrane-organizing protein causes neuro-fibromatosis type 2. Nature. 1993;363:515–521.
    1. Trofatter JA, MacCollin MM, Rutter JL, Murrell JR, Duyao MP, Parry DM, Eldridge R, Kley N, Menon AG, Pulaski K, et al. A novel moesin-, ezrin-, radixin-like gene is a candidate for the neurofibromatosis 2 tumor suppressor. Cell. 1993;72:791–800.
    1. Petrilli AM, Fernandez-Valle C. Role of Merlin/NF2 inactivation in tumor biology. Oncogene. 2015;35:537–48.
    1. Farschtschi S, Kollmann P, Dalchow C, Stein A, Mautner VF. Reduced dosage of bevacizumab in treatment of vestibular schwannomas in patients with neurofibromatosis type 2. Eur Arch Otorhinolaryngol. 2015;272:3857–3860.
    1. Karajannis MA, Ferner RE. Neurofibromatosis-related tumors: emerging biology and therapies. Current opinion in pediatrics. 2015;27:26–33.
    1. Ammoun S, Hanemann CO. Emerging therapeutic targets in schwannomas and other merlin-deficient tumors. Nature reviews Neurology. 2011;7:392–399.
    1. Andrae J, Gallini R, Betsholtz C. Role of platelet-derived growth factors in physiology and medicine. Genes Dev. 2008;22:1276–1312.
    1. Roskoski R., Jr Src protein-tyrosine kinase structure, mechanism, and small molecule inhibitors. Pharmacol Res. 2015;94:9–25.
    1. Ammoun S, Flaiz C, Ristic N, Schuldt J, Hanemann CO. Dissecting and targeting the growth factor-dependent and growth factor-independent extracellular signal-regulated kinase pathway in human schwannoma. Cancer Res. 2008;68:5236–5245.
    1. Lallemand D, Manent J, Couvelard A, Watilliaux A, Siena M, Chareyre F, Lampin A, Niwa-Kawakita M, Kalamarides M, Giovannini M. Merlin regulates transmembrane receptor accumulation and signaling at the plasma membrane in primary mouse Schwann cells and in human schwannomas. Oncogene. 2009;28:854–865.
    1. Mukherjee J, Kamnasaran D, Balasubramaniam A, Radovanovic I, Zadeh G, Kiehl TR, Guha A. Human schwannomas express activated platelet-derived growth factor receptors and c-kit and are growth inhibited by Gleevec (Imatinib Mesylate) Cancer Res. 2009;69:5099–5107.
    1. Ammoun S, Cunliffe CH, Allen JC, Chiriboga L, Giancotti FG, Zagzag D, Hanemann CO, Karajannis MA. ErbB/HER receptor activation and preclinical efficacy of lapatinib in vestibular schwannoma. Neuro-oncology. 2010;12:834–843.
    1. Fraenzer JT, Pan H, Minimo L, Jr, Smith GM, Knauer D, Hung G. Overexpression of the NF2 gene inhibits schwannoma cell proliferation through promoting PDGFR degradation. Int J Oncol. 2003;23:1493–1500.
    1. Houshmandi SS, Emnett RJ, Giovannini M, Gutmann DH. The neurofibromatosis 2 protein, merlin, regulates glial cell growth in an ErbB2- and Src-dependent manner. Molecular and cellular biology. 2009;29:1472–1486.
    1. Zhou L, Hanemann CO. Merlin, a multi-suppressor from cell membrane to the nucleus. FEBS Lett. 2012;586:1403–1408.
    1. Zhou T, Commodore L, Huang WS, Wang Y, Thomas M, Keats J, Xu Q, Rivera VM, Shakespeare WC, Clackson T, Dalgarno DC, Zhu X. Structural mechanism of the Pan-BCR-ABL inhibitor ponatinib (AP24534): lessons for overcoming kinase inhibitor resistance. Chem Biol Drug Des. 2011;77:1–11.
    1. Blanc J, Geney R, Menet C. Type II kinase inhibitors: an opportunity in cancer for rational design. Anticancer Agents Med Chem. 2013;13:731–747.
    1. O’Hare T, Shakespeare WC, Zhu X, Eide CA, Rivera VM, Wang F, Adrian LT, Zhou T, Huang WS, Xu Q, Metcalf CA, 3rd, Tyner JW, Loriaux MM, et al. AP24534, a pan-BCR-ABL inhibitor for chronic myeloid leukemia, potently inhibits the T315I mutant and overcomes mutation-based resistance. Cancer Cell. 2009;16:401–412.
    1. Zhou L, Ercolano E, Ammoun S, Schmid MC, Barczyk MA, Hanemann CO. Merlin-deficient human tumors show loss of contact inhibition and activation of Wnt/beta-catenin signaling linked to the PDGFR/Src and Rac/PAK pathways. Neoplasia. 2011;13:1101–1112.
    1. Ammoun S, Ristic N, Matthies C, Hilton DA, Hanemann CO. Targeting ERK1/2 activation and proliferation in human primary schwannoma cells with MEK1/2 inhibitor AZD6244. Neurobiology of disease. 2010;37:141–146.
    1. Hossain S, Fragoso G, Mushynski WE, Almazan G. Regulation of peripheral myelination by Src-like kinases. Exp Neurol. 2010;226:47–57.
    1. Kumar A, Jaggi AS, Singh N. Pharmacology of Src family kinases and therapeutic implications of their modulators. Fundam Clin Pharmacol. 2015;29:115–130.
    1. McLean GW, Carragher NO, Avizienyte E, Evans J, Brunton VG, Frame MC. The role of focal-adhesion kinase in cancer - a new therapeutic opportunity. Nat Rev Cancer. 2005;5:505–515.
    1. Calalb MB, Polte TR, Hanks SK. Tyrosine phosphorylation of focal adhesion kinase at sites in the catalytic domain regulates kinase activity: a role for Src family kinases. Molecular and cellular biology. 1995;15:954–963.
    1. Turner CE. Paxillin interactions. J Cell Sci. 2000;113(Pt 23):4139–4140.
    1. Heldin CH, Lennartsson J. Structural and functional properties of platelet-derived growth factor and stem cell factor receptors. Cold Spring Harb Perspect Biol. 2013;5:a009100.
    1. Alessi DR, Andjelkovic M, Caudwell B, Cron P, Morrice N, Cohen P, Hemmings BA. Mechanism of activation of protein kinase B by insulin and IGF-1. EMBO J. 1996;15:6541–6551.
    1. Alessi DR, Kozlowski MT, Weng QP, Morrice N, Avruch J. 3-Phosphoinositide-dependent protein kinase 1 (PDK1) phosphorylates and activates the p70S6 kinase in vivo and in vitro. Curr Biol. 1998;8:69–81.
    1. Bowman T, Broome MA, Sinibaldi D, Wharton W, Pledger WJ, Sedivy JM, Irby R, Yeatman T, Courtneidge SA, Jove R. Stat3-mediated Myc expression is required for Src transformation and PDGF-induced mitogenesis. Proc Natl Acad Sci U S A. 2001;98:7319–7324.
    1. Kortylewski M, Jove R, Yu H. Targeting STAT3 affects melanoma on multiple fronts. Cancer Metastasis Rev. 2005;24:315–327.
    1. Wang YZ, Wharton W, Garcia R, Kraker A, Jove R, Pledger WJ. Activation of Stat3 preassembled with platelet-derived growth factor beta receptors requires Src kinase activity. Oncogene. 2000;19:2075–2085.
    1. Kim DJ, Chan KS, Sano S, Digiovanni J. Signal transducer and activator of transcription 3 (Stat3) in epithelial carcinogenesis. Mol Carcinog. 2007;46:725–731.
    1. Meloche S, Pouyssegur J. The ERK1/2 mitogen-activated protein kinase pathway as a master regulator of the G1- to S-phase transition. Oncogene. 2007;26:3227–3239.
    1. Jakel H, Peschel I, Kunze C, Weinl C, Hengst L. Regulation of p27 (Kip1) by mitogen-induced tyrosine phosphorylation. Cell Cycle. 2012;11:1910–1917.
    1. Ammoun S, Schmid MC, Zhou L, Ristic N, Ercolano E, Hilton DA, Perks CM, Hanemann CO. Insulin-like growth factor-binding protein-1 (IGFBP-1) regulates human schwannoma proliferation, adhesion and survival. Oncogene. 2012;31:1710–1722.
    1. Gwanmesia PM, Romanski A, Schwarz K, Bacic B, Ruthardt M, Ottmann OG. The effect of the dual Src/Abl kinase inhibitor AZD0530 on Philadelphia positive leukaemia cell lines. BMC Cancer. 2009;9:53.
    1. Donato NJ, Wu JY, Stapley J, Lin H, Arlinghaus R, Aggarwal BB, Shishodia S, Albitar M, Hayes K, Kantarjian H, Talpaz M. Imatinib mesylate resistance through BCR-ABL independence in chronic myelogenous leukemia. Cancer Res. 2004;64:672–677.
    1. Huang WS, Zhu X, Wang Y, Azam M, Wen D, Sundaramoorthi R, Thomas RM, Liu S, Banda G, Lentini SP, Das S, Xu Q, Keats J, et al. 9-(Arenethenyl) purines as dual Src/Abl kinase inhibitors targeting the inactive conformation: design, synthesis, and biological evaluation. J Med Chem. 2009;52:4743–4756.
    1. Jabbour E, Kantarjian H, Cortes J. Use of second- and third-generation tyrosine kinase inhibitors in the treatment of chronic myeloid leukemia: an evolving treatment paradigm. Clin Lymphoma Myeloma Leuk. 2015;15:323–334.
    1. Ammoun S, Schmid MC, Triner J, Manley P, Hanemann CO. Nilotinib alone or in combination with selumetinib is a drug candidate for neurofibromatosis type 2. Neuro-oncology. 2011;13:759–766.
    1. Giovannini M, Robanus-Maandag E, van der Valk M, Niwa-Kawakita M, Abramowski V, Goutebroze L, Woodruff JM, Berns A, Thomas G. Conditional biallelic Nf2 mutation in the mouse promotes manifestations of human neurofibromatosis type 2. Genes Dev. 2000;14:1617–1630.
    1. Jessen KR, Mirsky R, Morgan L. Role of cyclic AMP and proliferation controls in Schwann cell differentiation. Ann N Y Acad Sci. 1991;633:78–89.
    1. Zorick TS, Lemke G. Schwann cell differentiation. Curr Opin Cell Biol. 1996;8:870–876.
    1. Atanasoski S, Boentert M, De Ventura L, Pohl H, Baranek C, Beier K, Young P, Barbacid M, Suter U. Postnatal Schwann cell proliferation but not myelination is strictly and uniquely dependent on cyclin-dependent kinase 4 (cdk4) Mol Cell Neurosci. 2008;37:519–527.
    1. Mirsky R, Woodhoo A, Parkinson DB, Arthur-Farraj P, Bhaskaran A, Jessen KR. Novel signals controlling embryonic Schwann cell development, myelination and dedifferentiation. J Peripher Nerv Syst. 2008;13:122–135.
    1. Atanasoski S, Shumas S, Dickson C, Scherer SS, Suter U. Differential cyclin D1 requirements of proliferating Schwann cells during development and after injury. Mol Cell Neurosci. 2001;18:581–592.
    1. Kim HA, Pomeroy SL, Whoriskey W, Pawlitzky I, Benowitz LI, Sicinski P, Stiles CD, Roberts TM. A developmentally regulated switch directs regenerative growth of Schwann cells through cyclin D1. Neuron. 2000;26:405–416.
    1. Heldin CH, Westermark B. Mechanism of action and in vivo role of platelet-derived growth factor. Physiol Rev. 1999;79:1283–1316.
    1. Meier C, Parmantier E, Brennan A, Mirsky R, Jessen KR. Developing Schwann cells acquire the ability to survive without axons by establishing an autocrine circuit involving insulin-like growth factor, neurotrophin-3, and platelet-derived growth factor-BB. J Neurosci. 1999;19:3847–3859.
    1. Peulve P, Laquerriere A, Paresy M, Hemet J, Tadie M. Establishment of adult rat Schwann cell cultures: effect of b-FGF, alpha-MSH, NGF, PDGF, and TGF-beta on cell cycle. Exp Cell Res. 1994;214:543–550.
    1. James MF, Stivison E, Beauchamp R, Han S, Li H, Wallace MR, Gusella JF, Stemmer-Rachamimov AO, Ramesh V. Regulation of mTOR complex 2 signaling in neurofibromatosis 2-deficient target cell types. Mol Cancer Res. 2012;10:649–659.
    1. Petrilli AM, Fuse MA, Donnan MS, Bott M, Sparrow NA, Tondera D, Huffziger J, Frenzel C, Malany CS, Echeverri CJ, Smith L, Fernandez-Valle C. A chemical biology approach identified PI3K as a potential therapeutic target for neurofibromatosis type 2. American journal of translational research. 2014;6:471–493.
    1. Rong R, Tang X, Gutmann DH, Ye K. Neurofibromatosis 2 (NF2) tumor suppressor merlin inhibits phosphatidylinositol 3-kinase through binding to PIKE-L. Proc Natl Acad Sci U S A. 2004;101:18200–18205.
    1. Masuda M, Suzui M, Yasumatu R, Nakashima T, Kuratomi Y, Azuma K, Tomita K, Komiyama S, Weinstein IB. Constitutive activation of signal transducers and activators of transcription 3 correlates with cyclin D1 overexpression and may provide a novel prognostic marker in head and neck squamous cell carcinoma. Cancer Res. 2002;62:3351–3355.
    1. Sinibaldi D, Wharton W, Turkson J, Bowman T, Pledger WJ, Jove R. Induction of p21WAF1/CIP1 and cyclin D1 expression by the Src oncoprotein in mouse fibroblasts: role of activated STAT3 signaling. Oncogene. 2000;19:5419–5427.
    1. Hari SB, Perera BG, Ranjitkar P, Seeliger MA, Maly DJ. Conformation-selective inhibitors reveal differences in the activation and phosphate-binding loops of the tyrosine kinases Abl and Src. ACS Chem Biol. 2013;8:2734–2743.
    1. Hoy SM. Ponatinib: a review of its use in adults with chronic myeloid leukaemia or Philadelphia chromosome-positive acute lymphoblastic leukaemia. Drugs. 2014;74:793–806.
    1. Moslehi JJ, Deininger M. Tyrosine Kinase Inhibitor-Associated Cardiovascular Toxicity in Chronic Myeloid Leukemia. J Clin Oncol. 2015;33:4210–4218.
    1. Millot F, Guilhot J, Baruchel A, Petit A, Leblanc T, Bertrand Y, Mazingue F, Lutz P, Verite C, Berthou C, Galambrun C, Nicolas S, Yacouben K, et al. Growth deceleration in children treated with imatinib for chronic myeloid leukaemia. Eur J Cancer. 2014;50:3206–3211.
    1. Mombach JC, Bugs CA, Chaouiya C. Modelling the onset of senescence at the G1/S cell cycle checkpoint. BMC Genomics. 2014;15:S7.
    1. Pucci B, Kasten M, Giordano A. Cell cycle and apoptosis. Neoplasia. 2000;2:291–299.
    1. Ehrhardt H, Wachter F, Grunert M, Jeremias I. Cell cycle-arrested tumor cells exhibit increased sensitivity towards TRAIL-induced apoptosis. Cell Death Dis. 2013;4:e661.
    1. Petrilli A, Copik A, Posadas M, Chang LS, Welling DB, Giovannini M, Fernandez-Valle C. LIM domain kinases as potential therapeutic targets for neurofibromatosis type 2. Oncogene. 2014;33:3571–3582.
    1. Petrilli A, Bott M, Fernandez-Valle C. Inhibition of SIRT2 in merlin/NF2-mutant Schwann cells triggers necrosis. Oncotarget. 2013;4:2354–2365. doi: 10.18632/oncotarget.1422.
    1. Darzynkiewicz Z, Gong J, Juan G, Ardelt B, Traganos F. Cytometry of cyclin proteins. Cytometry. 1996;25:1–13.

Source: PubMed

3
Subscribe