Mitotic Spindle Disruption by Alternating Electric Fields Leads to Improper Chromosome Segregation and Mitotic Catastrophe in Cancer Cells

Moshe Giladi, Rosa S Schneiderman, Tali Voloshin, Yaara Porat, Mijal Munster, Roni Blat, Shay Sherbo, Zeev Bomzon, Noa Urman, Aviran Itzhaki, Shay Cahal, Anna Shteingauz, Aafia Chaudhry, Eilon D Kirson, Uri Weinberg, Yoram Palti, Moshe Giladi, Rosa S Schneiderman, Tali Voloshin, Yaara Porat, Mijal Munster, Roni Blat, Shay Sherbo, Zeev Bomzon, Noa Urman, Aviran Itzhaki, Shay Cahal, Anna Shteingauz, Aafia Chaudhry, Eilon D Kirson, Uri Weinberg, Yoram Palti

Abstract

Tumor Treating Fields (TTFields) are low intensity, intermediate frequency, alternating electric fields. TTFields are a unique anti-mitotic treatment modality delivered in a continuous, noninvasive manner to the region of a tumor. It was previously postulated that by exerting directional forces on highly polar intracellular elements during mitosis, TTFields could disrupt the normal assembly of spindle microtubules. However there is limited evidence directly linking TTFields to an effect on microtubules. Here we report that TTFields decrease the ratio between polymerized and total tubulin, and prevent proper mitotic spindle assembly. The aberrant mitotic events induced by TTFields lead to abnormal chromosome segregation, cellular multinucleation, and caspase dependent apoptosis of daughter cells. The effect of TTFields on cell viability and clonogenic survival substantially depends upon the cell division rate. We show that by extending the duration of exposure to TTFields, slowly dividing cells can be affected to a similar extent as rapidly dividing cells.

Conflict of interest statement

We wish to disclose that Yoram Palti holds stock in Novocure Ltd. All other authors, except Yoram Palti are paid employees of Novocure. There are no other known conflicts of interest associated with this publication.

Figures

Figure 1. TTFields Treatment Induce Severe Mitotic…
Figure 1. TTFields Treatment Induce Severe Mitotic Spindle Damage in Cancer Cell Lines.
(A–C) A549 cells were treated with TTFields for 24 hours. (A) Confocal fluorescence microscopy images of: Upper panel- normal metaphase alignment of chromosomes in control cell, Lower panel: metaphase alignment in TTFields treated cell. Blue, Dapi-stained DNA; Red, Phospho Histone 3 (PH3) stained chromosomes during mitosis; green, tubulin. The scale bar represents 5 μm. (B) Tubulin fluorescence images were inverted and pseudocolored so that increasing fluorescence intensity is indicated from blue to red (scale bar represent arbitrary units). Dashed lines define the region between the two spindle poles (white) and overall tubulin fluorescence within the cell (Red). (C) Box-and-whiskers plots show the percentage of fluorescence intensity defined by the spindle and poles in relation to the total sum of tubulin fluorescence within the cell. The boxes show the mean and the interquartile ranges, while the whiskers show the full range. (D) Z-stack based 2D visualization of the mitotic spindle structure in each cell following enhancement of the cytoskeletal filament networks. (E) Box-and-whiskers plots show the percentage of fluorescence intensity defined by the spindle and poles in relation to the total sum of microtubule fluorescence within the cell. (F,G) In a separate experiment cell lysates of A2780 cells were collected following 48 hours treatment with TTFields. (F) Immunoblot of polymerized (pol) and depolymerized (depol) tubulin fractions were compared to untreated cells and cells treated with paclitaxel or vinorelbin. (G) Graph represents the percentages of polymerized microtubules in relation to total tubulin. Horizontal bars indicate mean values with SD. (H) Immunoblot of polymerized (pol) and depolymerized (depol) tubulin fractions of cells treated with TTFields for 10 hours were compared to untreated cells, 16 hours after re-plating. (I) Graph represents the percentages of polymerized microtubules in relation to total tubulin. Horizontal bars indicate mean values with SD. 0.05 > *p > 0.01 and **p < 0.01 from control group.
Figure 2. TTFields Application Induce the Formation…
Figure 2. TTFields Application Induce the Formation of Multinuclear Cells and Chromosome Aneuploidy.
(A) Phase-contrast and fluorescent time-lapse analysis of nuclei formation in TTFields treated Tubulin-GFP HeLa cells. Dashed line defines cell nuclei following division. Numbers show elapsed time (hour:minute). The scale bar represents 10 μm. In a separate experiment, U-87 MG cells were treated with TTFields for 72 hours. (B) Upper panel, U-87 MG control cells. Lower panel, TTFields treated cells. Arrow indicates micronuclei structures. Blue, Dapi-stained DNA; Red, Phalloidin-stained Actin. The scale bar represent 10 μm. Dashed line defines cell boundaries. (C,D) Fischer rats inoculated intracranially with F-98 glioma cells were treated with 200 kHz TTFields for 7 days, 1 week after tumor inoculation. At the end of treatment, tumors were removed and evaluated for atypical mitotic figures. (C) Representative images of mitotic figures. White lines encircle mitotic cells. The scale bar represent 5 μm. (D) Quantification of normal mitotic figures is expressed as percentage. At least 120 cells in metaphase or anaphase were analyzed in each group. ***p < 0.001 from control group. (E) A2780 cells were treated with TTFields for 96 hours. Chromosome number was evaluated every 24 hours. Horizontal bars indicate median values (p < 0.0001 ; Brown-Forsythe test). (F–H) Spectral karyotyping of A2780 cells (46, xx, der(6)t(1;6)) showing numerical aberrations following TTFields treatment. (F) Control.(G) Hypodiploidy following treatment. (H) Tetraploidy following treatment.
Figure 3. TTFields Treatment Efficacy Is Cell…
Figure 3. TTFields Treatment Efficacy Is Cell Doubling Time Dependent.
Comparison of cell lines response to TTFields treatment. Cells were treated with TTFields for 72 hours at optimal frequency (Table 1). (A) Effect of TTFields treatment on cell count of various cancer cell lines. (B) Clonogenic survival of various cancer cell lines following TTFields treatment. (C) Correlation between cell doubling time and cell count (p = 0.008) and clonogenic response (p = 0.023) following TTFields treatment.
Figure 4. TTFields Treatment Efficacy is Dependent…
Figure 4. TTFields Treatment Efficacy is Dependent on Treatment Duration.
(A,B) A2780 cells were treated with TTFields for 72 hours. (A) Effect of TTFields treatment on number of A2780 cells. (B) Clonogenic survival of A2780 cells following TTFields treatment. (C,D) In a separate experiment, U-87 MG cells were treated with TTFields for 144 hours. (C) Effect of TTFields treatment on number of U-87 MG cells. (D) Clonogenic survival of U-87 MG cells following TTFields treatment. 0.05 > *p > 0.01, **p < 0.01, and ***p < 0.001 from control group.
Figure 5. Mitotic Arrest and Mitotic Cell…
Figure 5. Mitotic Arrest and Mitotic Cell Death Are Possible Outcomes of TTFields Treatment.
(A–C) Phase-contrast and fluorescent time-lapse analysis of mitosis in Tubulin-GFP HeLa cells. (A) Control cells. (B) TTFields treated cells (8 h). Numbers show elapsed time relative to entry to mitosis (hour: minute). Arrow indicates membrane blebbing. Cell death is indicated as D. The scale bar represents 10 μm. (C) Box-and-whiskers plots show the time that cells spent arrested in mitosis. The boxes show the mean and the interquartile ranges, while the whiskers show the full range. (D,E) In a separate experiment, A2780 cells were treated with TTFields for 72 hours. (D) Cell cycle analysis was performed using flow cytometry. (E) Graph represents the change in percentage of A2780 cells in M phase following TTFields application. (F) Fischer rats inoculated intracranially with F-98 glioma cells were treated with 200 kHz TTFields for 7 days 1 week after tumor inoculation. At the end of treatment, tumors were removed and evaluated for average mitotic rate. 0.05 > *p > 0.01, ***p < 0.001 from control group.
Figure 6. TTFields Kill Cancer Cells by…
Figure 6. TTFields Kill Cancer Cells by Triggering Caspase Mediated Apoptosis.
A2780 cells were treated with TTFields for 48 hours. (A–C) Cell samples were collected and evaluated for apoptosis. (A) Live cells. (B) Early apoptotic cells. (C) Late apoptotic cells. (D) Caspase activity was evaluated using flow cytometry analysis following 48 hours of TTFields application. (E,F) Effect of pan-caspase inhibition by Z-VAD-FMK on A2780 response to TTFields treatment. (E) Evaluation of number of cells (unpaired t-test). (F) Evaluation of apoptosis. 0.05 > *p > 0.01, and ***p < 0.001 from control group.
Figure 7. Effects of TTFields on replicating…
Figure 7. Effects of TTFields on replicating cells.
TTFields exert directional forces on polar microtubules and interfere with the assembly of the normal mitotic spindle. Such interference with microtubule dynamics results in abnormal spindle formation and subsequent mitotic arrest or delay, possibly due to improper attachment of chromosomes to the spindle fibers. Cells can die while in mitotic arrest, however, a more common outcome (highlighted by bold arrow) is progression to cell division. This can lead to the formation of either normal or abnormal aneuploid progeny. The formation of the tetraploid cells can occur either due to mitotic exit through slippage or can occur during improper cell division. Abnormal daughter cells can die in the subsequent interphase, can undergo a permanent arrest, or can proliferate through additional mitosis where they will be subjected to further TTFields assault.

References

    1. McCaig C. D., Rajnicek A. M., Song B. & Zhao M. Controlling cell behavior electrically: current views and future potential. Physiol. Rev. 85, 943–978 (2005).
    1. Markx G. H. The use of electric fields in tissue engineering: A review. Organogenesis 4, 11–17 (2008).
    1. Bassett C. A. The development and application of pulsed electromagnetic fields (PEMFs) for ununited fractures and arthrodeses. Clin. Plast. Surg. 12, 259–277 (1985).
    1. Repacholi M. H. & Greenebaum B. Interaction of static and extremely low frequency electric and magnetic fields with living systems: health effects and research needs. Bioelectromagnetics 20, 133–160 (1999).
    1. Elson E. Biologic effects of radiofrequency and microwave fields: in vivo and in vitro experimental results in The biomedical engineering handbook (ed. Bronzino J. D.) 1417–1423 (CRC Press, 1995).
    1. Kirson E. D. et al. Alternating electric fields arrest cell proliferation in animal tumor models and human brain tumors. Proc. Natl. Acad. Sci. USA 104, 10152–10157 (2007).
    1. Kirson E. D. et al. Disruption of cancer cell replication by alternating electric fields. Cancer Res. 64, 3288–3295 (2004).
    1. Davies A. M., Weinberg U. & Palti Y. Tumor treating fields: a new frontier in cancer therapy. Ann. N Y Acad. Sci. 1291, 86–95 (2013).
    1. Stupp R. et al. NovoTTF-100A versus physician’s choice chemotherapy in recurrent glioblastoma: a randomised phase III trial of a novel treatment modality. Eur. J. Cancer 48, 2192–2202 (2012).
    1. Mrugala M. M. et al. Clinical Practice Experience With NovoTTF-100A System for Glioblastoma: The Patient Registry Dataset (PRiDe). Semin. Oncol. 41 Suppl 6, S4–S13 (2014).
    1. Wong E. T., Lok E. & Swanson K. D. An Evidence-Based Review of Alternating Electric Fields Therapy for Malignant Gliomas. Curr. Treat. Options Oncol. 16, 353 (2015).
    1. Havelka D., Cifra M., Kucera O., Pokorny J. & Vrba J. High-frequency electric field and radiation characteristics of cellular microtubule network. J. Theor. Biol. 286, 31–40 (2011).
    1. Cifra M., Pokorny J., Havelka D. & Kucera O. Electric field generated by axial longitudinal vibration modes of microtubule. Biosystems 100, 122–131 (2010).
    1. Jordan M. A., Toso R. J., Thrower D. & Wilson L. Mechanism of mitotic block and inhibition of cell proliferation by taxol at low concentrations. Proc. Natl. Acad. Sci. USA 90, 9552–9556 (1993).
    1. Sorger P. K., Dobles M., Tournebize R. & Hyman A. A. Coupling cell division and cell death to microtubule dynamics. Curr. Opin. Cell Biol. 9, 807–814 (1997).
    1. Bollag D. M. et al. Epothilones, a new class of microtubule-stabilizing agents with a taxol-like mechanism of action. Cancer Res. 55, 2325–2333 (1995).
    1. Gascoigne K. E. & Taylor S. S. How do anti-mitotic drugs kill cancer cells? J. Cell Sci. 122, 2579–2585 (2009).
    1. Coleman M. L. et al. Membrane blebbing during apoptosis results from caspase-mediated activation of ROCK I. Nat. Cell Biol. 3, 339–345 (2001).
    1. Kline-Smith S. L. & Walczak C. E. Mitotic spindle assembly and chromosome segregation: refocusing on microtubule dynamics. Mol. Cell 15, 317–327(2004).
    1. Gordon M. B., Howard L. & Compton D. A. Chromosome movement in mitosis requires microtubule anchorage at spindle poles. J. Cell Biol. 152, 425–434 (2001).
    1. Vitale I., Galluzzi L., Castedo M. & Kroemer G. Mitotic catastrophe: a mechanism for avoiding genomic instability. Nat. Rev. Mol. Cell Biol. 12, 385–392 (2011).
    1. Musacchio A. & Hardwick K. G. The spindle checkpoint: structural insights into dynamic signalling. Nat. Rev. Mol. Cell Biol. 3, 731–741 (2002).
    1. Musacchio A. & Salmon E. D. The spindle-assembly checkpoint in space and time. Nat. Rev. Mol. Cell Biol. 8, 379–393 (2007).
    1. Weaver B. A. & Cleveland D. W. Decoding the links between mitosis, cancer, and chemotherapy: The mitotic checkpoint, adaptation, and cell death. Cancer Cell 8, 7–12 (2005).
    1. Demidenko Z. N. et al. Mechanism of G1-like arrest by low concentrations of paclitaxel: next cell cycle p53-dependent arrest with sub G1 DNA content mediated by prolonged mitosis. Oncogene 27, 4402–4410 (2008).
    1. Rieder C. L. & Maiato H. Stuck in division or passing through: what happens when cells cannot satisfy the spindle assembly checkpoint. Dev. Cell 7, 637–651 (2004).
    1. Hong F. D. et al. Taxol, vincristine or nocodazole induces lethality in G1-checkpoint-defective human astrocytoma U373 MG cells by triggering hyperploid progression. Carcinogenesis 20, 1161–1168 (1999).
    1. Sihn C. R. et al. p55CDC/hCDC20 mutant induces mitotic catastrophe by inhibiting the MAD2-dependent spindle checkpoint activity in tumor cells. Cancer Lett. 201, 203–210 (2003).
    1. Yamada H. Y. & Gorbsky G. J. Spindle checkpoint function and cellular sensitivity to antimitotic drugs. Mol. Cancer Ther. 5, 2963–2969 (2006).
    1. Elhajouji A., Cunha M. & Kirsch-Volders M. Spindle poisons can induce polyploidy by mitotic slippage and micronucleate mononucleates in the cytokinesis-block assay. Mutagenesis 13, 193–198 (1998).
    1. Giladi M. et al. Alternating electric fields (tumor-treating fields therapy) can improve chemotherapy treatment efficacy in non-small cell lung cancer both in vitro and in vivo. Semin. Oncol. 41 Suppl 6, S35–41 (2014).
    1. Miranda P. C., Mekonnen A., Salvador R. & Basser P. J. Predicting the electric field distribution in the brain for the treatment of glioblastoma. Phys. Med. Biol. 59, 4137–4147 (2014).
    1. Pless M. et al. A phase I/II trial of Tumor Treating Fields (TTFields) therapy in combination with pemetrexed for advanced non-small cell lung cancer. Lung Cancer 81, 445–450 (2013).
    1. Dumontet C. & Sikic B. I. Mechanisms of action of and resistance to antitubulin agents: microtubule dynamics, drug transport, and cell death. J. Clin. Oncol. 17, 1061–1070 (1999).
    1. Pearson C. G. et al. Measuring nanometer scale gradients in spindle microtubule dynamics using model convolution microscopy. Mol. Biol. Cell 17, 4069–4079 (2006).
    1. Herberich G., Windoffer R., Leube R. E. & Aach T. Signal and noise modeling in confocal laser scanning fluorescence microscopy. Med. Image Comput. Comput. Assist. Interv. 15, 381–388 (2012).
    1. Hung D. T., Jamison T. F. & Schreiber S. L. Understanding and controlling the cell cycle with natural products. Chem. Biol. 3, 623–639 (1996).
    1. Wilson L. & Jordan M. A. Microtubule dynamics: taking aim at a moving target. Chem. Biol. 2, 569–573 (1995).
    1. Jordan M. A. & Wilson L. Microtubules as a target for anticancer drugs. Nat. Rev. Cancer 4, 253–265 (2004).
    1. Chu S. W., Badar S., Morris D. L. & Pourgholami M. H. Potent inhibition of tubulin polymerisation and proliferation of paclitaxel-resistant 1A9PTX22 human ovarian cancer cells by albendazole. Anticancer Res. 29, 3791–3796 (2009).
    1. Belmont L. D., Hyman A. A., Sawin K. E. & Mitchison T. J. Real-time visualization of cell cycle-dependent changes in microtubule dynamics in cytoplasmic extracts. Cell 62, 579–589 (1990).
    1. Mooney D. J. et al. Extracellular matrix controls tubulin monomer levels in hepatocytes by regulating protein turnover. Mol. Biol. Cell 5, 1281–1288 (1994).
    1. Sun Z. et al. Kaposi’s Sarcoma-Associated Herpesvirus-Encoded LANA Can Induce Chromosomal Instability through Targeted Degradation of the Mitotic Checkpoint Kinase Bub1. J. Virol. 88 (2014).
    1. Turner S. G. et al. The effect of field strength on glioblastoma multiforme response in patients treated with the NovoTTF-100A system. World J. Surg. Oncol. 12, 162 (2014).
    1. Parry E. M. et al. Detection and characterization of mechanisms of action of aneugenic chemicals. Mutagenesis 17, 509–521 (2002).
    1. Silk A. D. et al. Chromosome missegregation rate predicts whether aneuploidy will promote or suppress tumors. Proc. Natl. Acad. Sci. USA 110, E4134–4141 (2013).
    1. Vignon C. et al. Flow cytometric quantification of all phases of the cell cycle and apoptosis in a two-color fluorescence plot. PLoS One 8, e68425 (2013).
    1. Ahmed A. A. et al. The extracellular matrix protein TGFBI induces microtubule stabilization and sensitizes ovarian cancers to paclitaxel. Cancer Cell 12, 514–527 (2007).
    1. Kirson E. D. et al. Chemotherapeutic treatment efficacy and sensitivity are increased by adjuvant alternating electric fields (TTFields). BMC Med. Phys. 9, 1 (2009).
    1. Romero Acuna L. et al. Vinorelbine and paclitaxel as first-line chemotherapy in metastatic breast cancer. J. Clin. Oncol. 17, 74–81 (1999).
    1. Ganguly A. et al. The role of microtubules and their dynamics in cell migration. J. Biol. Chem. 287 (2012).
    1. Yoon S. O., Shin S. & Mercurio A. M. Hypoxia stimulates carcinoma invasion by stabilizing microtubules and promoting the Rab11 trafficking of the alpha6beta4 integrin. Cancer Res. 65, 2761–2769 (2005).
    1. Zasadil L. M. et al. Cytotoxicity of paclitaxel in breast cancer is due to chromosome missegregation on multipolar spindles. Sci. Transl. Med. 6, 229–243, (2014).
    1. Li R. et al. Aneuploidy correlated 100% with chemical transformation of Chinese hamster cells. Proc. Natl. Acad. Sci. USA 94, 14506–14511 (1997).
    1. Marx J. Debate surges over the origins of genomic defects in cancer. Science 297, 544–546 (2002).
    1. Holland A. J. & Cleveland D. W. Boveri revisited: chromosomal instability, aneuploidy and tumorigenesis. Nat. Rev. Mol. Cell Biol. 10, 478–487 (2009).
    1. Boveri T. Concerning the origin of malignant tumours by Theodor Boveri. Translated and annotated by Henry Harris. J. Cell Sci. 121 Suppl 1, 1–84 (2008).
    1. Orticello M. et al. N-terminus-modified Hec1 suppresses tumour growth by interfering with kinetochore-microtubule dynamics. Oncogene 34, 3325–3335 (2015).
    1. Weaver B. A. et al. Aneuploidy acts both oncogenically and as a tumor suppressor. Cancer Cell 11, 25–36 (2007).
    1. Tang Y. C. & Amon A. Gene copy-number alterations: a cost-benefit analysis. Cell 152, 394–405 (2013).
    1. Thompson S. L. & Compton D. A. Proliferation of aneuploid human cells is limited by a p53-dependent mechanism. J. Cell Biol. 188, 369–381 (2010).
    1. Li M. et al. The ATM-p53 pathway suppresses aneuploidy-induced tumorigenesis. Proc. Natl. Acad. Sci. USA 107, 14188–14193 (2010).
    1. Orth J. D. et al. Quantitative live imaging of cancer and normal cells treated with Kinesin-5 inhibitors indicates significant differences in phenotypic responses and cell fate. Mol. Cancer Ther. 7, 3480–3489 (2008).
    1. Shi J., Orth J. D. & Mitchison T. Cell type variation in responses to antimitotic drugs that target microtubules and kinesin-5. Cancer Res. 68, 3269–3276 (2008).
    1. Huang H. C., Shi J., Orth J. D. & Mitchison T. J. Evidence that mitotic exit is a better cancer therapeutic target than spindle assembly. Cancer Cell 16, 347–358 (2009).
    1. Noguchi S. Predictive factors for response to docetaxel in human breast cancers. Cancer Sci. 97, 813–820 (2006).
    1. Langen K. J. et al. Comparison of iodotyrosines and methionine uptake in a rat glioma model. J. Nucl. Med. 39, 1596–1599 (1998).
    1. Saini M. et al. Morphometrical characterization of two glioma models in the brain of immunocompetent and immunodeficient rats. J. Neurooncol. 42, 59–67 (1999).
    1. Giladi M. et al. Mitotic disruption and reduced clonogenicity of pancreatic cancer cells in vitro and in vivo by tumor treating fields. Pancreatology 14, 54–63 (2014).

Source: PubMed

3
구독하다