Targeting DNA Repair and Chromatin Crosstalk in Cancer Therapy
Abstract
:Simple Summary
Abstract
1. Introduction
2. DNA Repair Is Dysfunctional in Certain Developmental Diseases and Cancers
3. MRN and ATM/ATR Repair Signal Recognition Factors as Cancer Treatment Targets
4. Targeting Cell-Cycle Checkpoints in Cancer
5. The Use of Synthetic Lethality in Cancer Therapeutics and Its Limitations
6. Challenges Associated with Targeted Therapies and a Novel Strategy to Achieve Pan DNA Repair Inhibition
7. Chromatin Interactions Associated with Repair, Transcription, and Replication and Implications for Cancer Therapy
8. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kitano, K. Structural mechanisms of human RecQ helicases WRN and BLM. Front. Genet. 2014, 5, 366. [Google Scholar] [CrossRef]
- Goto, M. Werner’s syndrome: From clinics to genetics. Clin. Exp. Rheumatol. 2000, 18, 760–766. [Google Scholar] [PubMed]
- Manthei, K.A.; Keck, J.L. The BLM dissolvasome in DNA replication and repair. Cell. Mol. Life Sci. 2013, 70, 4067–4084. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- German, J. Bloom’s syndrome. Dermatol. Clin. 1995, 13, 7–18. [Google Scholar] [CrossRef]
- Aggarwal, M.; Sommers, J.A.; Shoemaker, R.H.; Brosh, R.M., Jr. Inhibition of helicase activity by a small molecule impairs Werner syndrome helicase (WRN) function in the cellular response to DNA damage or replication stress. Proc. Natl. Acad. Sci. USA 2011, 108, 1525–1530. [Google Scholar] [CrossRef] [Green Version]
- Hengel, S.R.; Spies, M.A.; Spies, M. Small-Molecule Inhibitors Targeting DNA Repair and DNA Repair Deficiency in Research and Cancer Therapy. Cell Chem. Biol. 2017, 24, 1101–1119. [Google Scholar] [CrossRef] [Green Version]
- Mirzoeva, O.K.; Petrini, J.H. DNA damage-dependent nuclear dynamics of the Mre11 complex. Mol. Cell. Biol. 2001, 21, 281–288. [Google Scholar] [CrossRef] [Green Version]
- Kastan, M.B. DNA damage responses: Mechanisms and roles in human disease: 2007 G.H.A. Clowes Memorial Award Lecture. Mol. Cancer Res. 2008, 6, 517–524. [Google Scholar] [CrossRef] [Green Version]
- O’Driscoll, M. Diseases associated with defective responses to DNA damage. Cold Spring Harb. Perspect. Biol. 2012, 4. [Google Scholar] [CrossRef] [Green Version]
- Spycher, C.; Miller, E.S.; Townsend, K.; Pavic, L.; Morrice, N.A.; Janscak, P.; Stewart, G.S.; Stucki, M. Constitutive phosphorylation of MDC1 physically links the MRE11-RAD50-NBS1 complex to damaged chromatin. J. Cell Biol. 2008, 181, 227–240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nakanishi, K.; Taniguchi, T.; Ranganathan, V.; New, H.V.; Moreau, L.A.; Stotsky, M.; Mathew, C.G.; Kastan, M.B.; Weaver, D.T.; D’Andrea, A.D. Interaction of FANCD2 and NBS1 in the DNA damage response. Nat. Cell Biol. 2002, 4, 913–920. [Google Scholar] [CrossRef] [PubMed]
- Waltes, R.; Kalb, R.; Gatei, M.; Kijas, A.W.; Stumm, M.; Sobeck, A.; Wieland, B.; Varon, R.; Lerenthal, Y.; Lavin, M.F.; et al. Human RAD50 deficiency in a Nijmegen breakage syndrome-like disorder. Am. J. Hum. Genet. 2009, 84, 605–616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lavin, M.F. Ataxia-telangiectasia: From a rare disorder to a paradigm for cell signalling and cancer. Nat. Rev. Mol. Cell. Biol. 2008, 9, 759–769. [Google Scholar] [CrossRef]
- Lavin, M.F. ATM and the Mre11 complex combine to recognize and signal DNA double-strand breaks. Oncogene 2007, 26, 7749–7758. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huang, R.X.; Zhou, P.K. DNA damage response signaling pathways and targets for radiotherapy sensitization in cancer. Signal Transduct. Target. Ther. 2020, 5, 60. [Google Scholar] [CrossRef]
- Durant, S.T.; Zheng, L.; Wang, Y.; Chen, K.; Zhang, L.; Zhang, T.; Yang, Z.; Riches, L.; Trinidad, A.G.; Fok, J.H.L.; et al. The brain-penetrant clinical ATM inhibitor AZD1390 radiosensitizes and improves survival of preclinical brain tumor models. Sci. Adv. 2018, 4, eaat1719. [Google Scholar] [CrossRef] [Green Version]
- Shiotani, B.; Zou, L. ATR signaling at a glance. J. Cell Sci. 2009, 122, 301–304. [Google Scholar] [CrossRef] [Green Version]
- Ball, H.L.; Myers, J.S.; Cortez, D. ATRIP binding to replication protein A-single-stranded DNA promotes ATR-ATRIP localization but is dispensable for Chk1 phosphorylation. Mol. Biol. Cell 2005, 16, 2372–2381. [Google Scholar] [CrossRef] [Green Version]
- Vassin, V.M.; Anantha, R.W.; Sokolova, E.; Kanner, S.; Borowiec, J.A. Human RPA phosphorylation by ATR stimulates DNA synthesis and prevents ssDNA accumulation during DNA-replication stress. J. Cell Sci. 2009, 122, 4070–4080. [Google Scholar] [CrossRef] [Green Version]
- Ma, M.; Rodriguez, A.; Sugimoto, K. Activation of ATR-related protein kinase upon DNA damage recognition. Curr. Genet. 2020, 66, 327–333. [Google Scholar] [CrossRef]
- Ubhi, T.; Brown, G.W. Exploiting DNA Replication Stress for Cancer Treatment. Cancer Res. 2019, 79, 1730–1739. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tanaka, A.; Weinel, S.; Nagy, N.; O’Driscoll, M.; Lai-Cheong, J.E.; Kulp-Shorten, C.L.; Knable, A.; Carpenter, G.; Fisher, S.A.; Hiragun, M.; et al. Germline mutation in ATR in autosomal- dominant oropharyngeal cancer syndrome. Am. J. Hum. Genet. 2012, 90, 511–517. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lewis, K.A.; Mullany, S.; Thomas, B.; Chien, J.; Loewen, R.; Shridhar, V.; Cliby, W.A. Heterozygous ATR mutations in mismatch repair-deficient cancer cells have functional significance. Cancer Res. 2005, 65, 7091–7095. [Google Scholar] [CrossRef] [Green Version]
- Moiseeva, T.N.; Yin, Y.; Calderon, M.J.; Qian, C.; Schamus-Haynes, S.; Sugitani, N.; Osmanbeyoglu, H.U.; Rothenberg, E.; Watkins, S.C.; Bakkenist, C.J. An ATR and CHK1 kinase signaling mechanism that limits origin firing during unperturbed DNA replication. Proc. Natl. Acad. Sci. USA 2019, 116, 13374–13383. [Google Scholar] [CrossRef] [Green Version]
- Qvist, P.; Huertas, P.; Jimeno, S.; Nyegaard, M.; Hassan, M.J.; Jackson, S.P.; Borglum, A.D. CtIP Mutations Cause Seckel and Jawad Syndromes. PLoS Genet. 2011, 7, e1002310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- O’Driscoll, M.; Ruiz-Perez, V.L.; Woods, C.G.; Jeggo, P.A.; Goodship, J.A. A splicing mutation affecting expression of ataxia-telangiectasia and Rad3-related protein (ATR) results in Seckel syndrome. Nat. Genet. 2003, 33, 497–501. [Google Scholar] [CrossRef] [Green Version]
- Alderton, G.K.; Joenje, H.; Varon, R.; Borglum, A.D.; Jeggo, P.A.; O’Driscoll, M. Seckel syndrome exhibits cellular features demonstrating defects in the ATR-signalling pathway. Hum. Mol. Genet. 2004, 13, 3127–3138. [Google Scholar] [CrossRef]
- Yazinski, S.A.; Zou, L. Functions, Regulation, and Therapeutic Implications of the ATR Checkpoint Pathway. Annu. Rev. Genet. 2016, 50, 155–173. [Google Scholar] [CrossRef]
- Marechal, A.; Zou, L. DNA damage sensing by the ATM and ATR kinases. Cold Spring Harb. Perspect. Biol. 2013, 5. [Google Scholar] [CrossRef]
- Yang, J.; Xu, Z.P.; Huang, Y.; Hamrick, H.E.; Duerksen-Hughes, P.J.; Yu, Y.N. ATM and ATR: Sensing DNA damage. World J. Gastroenterol. 2004, 10, 155–160. [Google Scholar] [CrossRef] [PubMed]
- Brown, E.J.; Baltimore, D. ATR disruption leads to chromosomal fragmentation and early embryonic lethality. Genes Dev. 2000, 14, 397–402. [Google Scholar] [PubMed]
- Ward, I.M.; Chen, J. Histone H2AX is phosphorylated in an ATR-dependent manner in response to replicational stress. J. Biol. Chem. 2001, 276, 47759–47762. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shiotani, B.; Zou, L. Single-stranded DNA orchestrates an ATM-to-ATR switch at DNA breaks. Mol. Cell 2009, 33, 547–558. [Google Scholar] [CrossRef] [Green Version]
- Tomimatsu, N.; Mukherjee, B.; Burma, S. Distinct roles of ATR and DNA-PKcs in triggering DNA damage responses in ATM-deficient cells. EMBO Rep. 2009, 10, 629–635. [Google Scholar] [CrossRef] [Green Version]
- Chanoux, R.A.; Yin, B.; Urtishak, K.A.; Asare, A.; Bassing, C.H.; Brown, E.J. ATR and H2AX cooperate in maintaining genome stability under replication stress. J. Biol. Chem. 2009, 284, 5994–6003. [Google Scholar] [CrossRef] [Green Version]
- Goodwin, J.F.; Knudsen, K.E. Beyond DNA repair: DNA-PK function in cancer. Cancer Discov. 2014, 4, 1126–1139. [Google Scholar] [CrossRef] [Green Version]
- Davidson, D.; Amrein, L.; Panasci, L.; Aloyz, R. Small Molecules, Inhibitors of DNA-PK, Targeting DNA Repair, and Beyond. Front. Pharmacol. 2013, 4, 5. [Google Scholar] [CrossRef] [Green Version]
- Rasco, D.W.; Papadopoulos, K.P.; Pourdehnad, M.; Gandhi, A.K.; Hagner, P.R.; Li, Y.; Wei, X.; Chopra, R.; Hege, K.; DiMartino, J.; et al. A First-in-Human Study of Novel Cereblon Modulator Avadomide (CC-122) in Advanced Malignancies. Clin. Cancer Res. 2019, 25, 90–98. [Google Scholar] [CrossRef] [Green Version]
- Carr, M.I.; Zimmermann, A.; Chiu, L.Y.; Zenke, F.T.; Blaukat, A.; Vassilev, L.T. DNA-PK Inhibitor, M3814, as a New Combination Partner of Mylotarg in the Treatment of Acute Myeloid Leukemia. Front. Oncol. 2020, 10, 127. [Google Scholar] [CrossRef]
- Wise, H.C.; Iyer, G.V.; Moore, K.; Temkin, S.M.; Gordon, S.; Aghajanian, C.; Grisham, R.N. Activity of M3814, an Oral DNA-PK Inhibitor, In Combination with Topoisomerase II Inhibitors in Ovarian Cancer Models. Sci. Rep. 2019, 9, 18882. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, Q.; Guntuku, S.; Cui, X.S.; Matsuoka, S.; Cortez, D.; Tamai, K.; Luo, G.; Carattini-Rivera, S.; DeMayo, F.; Bradley, A.; et al. Chk1 is an essential kinase that is regulated by Atr and required for the G(2)/M DNA damage checkpoint. Genes Dev. 2000, 14, 1448–1459. [Google Scholar] [PubMed]
- Molinari, M. Cell cycle checkpoints and their inactivation in human cancer. Cell Prolif. 2000, 33, 261–274. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Hunter, T. Roles of Chk1 in cell biology and cancer therapy. Int. J. Cancer 2014, 134, 1013–1023. [Google Scholar] [CrossRef] [Green Version]
- Visconti, R.; Della Monica, R.; Grieco, D. Cell cycle checkpoint in cancer: A therapeutically targetable double-edged sword. J. Exp. Clin. Cancer Res. 2016, 35, 153. [Google Scholar] [CrossRef] [Green Version]
- Sausville, E.; Lorusso, P.; Carducci, M.; Carter, J.; Quinn, M.F.; Malburg, L.; Azad, N.; Cosgrove, D.; Knight, R.; Barker, P.; et al. Phase I dose-escalation study of AZD7762, a checkpoint kinase inhibitor, in combination with gemcitabine in US patients with advanced solid tumors. Cancer Chemother. Pharmacol. 2014, 73, 539–549. [Google Scholar] [CrossRef] [Green Version]
- Tang, Y.; Dai, Y.; Grant, S.; Dent, P. Enhancing CHK1 inhibitor lethality in glioblastoma. Cancer Biol. Ther. 2012, 13, 379–388. [Google Scholar] [CrossRef] [Green Version]
- Haince, J.F.; McDonald, D.; Rodrigue, A.; Dery, U.; Masson, J.Y.; Hendzel, M.J.; Poirier, G.G. PARP1-dependent kinetics of recruitment of MRE11 and NBS1 proteins to multiple DNA damage sites. J. Biol. Chem. 2008, 283, 1197–1208. [Google Scholar] [CrossRef] [Green Version]
- Malyuchenko, N.V.; Kotova, E.Y.; Kulaeva, O.I.; Kirpichnikov, M.P.; Studitskiy, V.M. PARP1 Inhibitors: Antitumor drug design. Acta Nat. 2015, 7, 27–37. [Google Scholar] [CrossRef]
- Roy, R.; Chun, J.; Powell, S.N. BRCA1 and BRCA2: Different roles in a common pathway of genome protection. Nat. Rev. Cancer 2011, 12, 68–78. [Google Scholar] [CrossRef] [Green Version]
- Li, H.; Liu, Z.Y.; Wu, N.; Chen, Y.C.; Cheng, Q.; Wang, J. PARP inhibitor resistance: The underlying mechanisms and clinical implications. Mol. Cancer 2020, 19, 107. [Google Scholar] [CrossRef] [PubMed]
- Lord, C.J.; Ashworth, A. PARP inhibitors: Synthetic lethality in the clinic. Science 2017, 355, 1152–1158. [Google Scholar] [CrossRef] [PubMed]
- Jannetti, S.A.; Zeglis, B.M.; Zalutsky, M.R.; Reiner, T. Poly(ADP-Ribose)Polymerase (PARP) Inhibitors and Radiation Therapy. Front. Pharmacol. 2020, 11, 170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Coleman, R.L.; Oza, A.M.; Lorusso, D.; Aghajanian, C.; Oaknin, A.; Dean, A.; Colombo, N.; Weberpals, J.I.; Clamp, A.; Scambia, G.; et al. Rucaparib maintenance treatment for recurrent ovarian carcinoma after response to platinum therapy (ARIEL3): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017, 390, 1949–1961. [Google Scholar] [CrossRef] [Green Version]
- Karnitz, L.M.; Zou, L. Molecular Pathways: Targeting ATR in Cancer Therapy. Clin. Cancer Res. 2015, 21, 4780–4785. [Google Scholar] [CrossRef] [Green Version]
- Petroni, M.; Sardina, F.; Infante, P.; Bartolazzi, A.; Locatelli, E.; Fabretti, F.; Di Giulio, S.; Capalbo, C.; Cardinali, B.; Coppa, A.; et al. MRE11 inhibition highlights a replication stress-dependent vulnerability of MYCN-driven tumors. Cell Death Dis. 2018, 9, 895. [Google Scholar] [CrossRef]
- Batey, M.A.; Zhao, Y.; Kyle, S.; Richardson, C.; Slade, A.; Martin, N.M.; Lau, A.; Newell, D.R.; Curtin, N.J. Preclinical evaluation of a novel ATM inhibitor, KU59403, in vitro and in vivo in p53 functional and dysfunctional models of human cancer. Mol. Cancer Ther. 2013, 12, 959–967. [Google Scholar] [CrossRef] [Green Version]
- Dillon, M.T.; Boylan, Z.; Smith, D.; Guevara, J.; Mohammed, K.; Peckitt, C.; Saunders, M.; Banerji, U.; Clack, G.; Smith, S.A.; et al. PATRIOT: A phase I study to assess the tolerability, safety and biological effects of a specific ataxia telangiectasia and Rad3-related (ATR) inhibitor (AZD6738) as a single agent and in combination with palliative radiation therapy in patients with solid tumours. Clin. Transl. Radiat. Oncol. 2018, 12, 16–20. [Google Scholar] [CrossRef]
- Yap, T.A.; O’Carrigan, B.; Penney, M.S.; Lim, J.S.; Brown, J.S.; de Miguel Luken, M.J.; Tunariu, N.; Perez-Lopez, R.; Rodrigues, D.N.; Riisnaes, R.; et al. Phase I Trial of First-in-Class ATR Inhibitor M6620 (VX-970) as Monotherapy or in Combination With Carboplatin in Patients With Advanced Solid Tumors. J. Clin. Oncol. 2020, 38, 3195–3204. [Google Scholar] [CrossRef]
- Nasser, M.I.; Zhu, S.; Chen, C.; Zhao, M.; Huang, H.; Zhu, P. A Comprehensive Review on Schisandrin B and Its Biological Properties. Oxid. Med. Cell. Longev. 2020, 2020, 2172740. [Google Scholar] [CrossRef] [Green Version]
- Evans, T.; Matulonis, U. PARP inhibitors in ovarian cancer: Evidence, experience and clinical potential. Ther. Adv. Med. Oncol. 2017, 9, 253–267. [Google Scholar] [CrossRef] [PubMed]
- Brown, J.S.; Kaye, S.B.; Yap, T.A. PARP inhibitors: The race is on. Br. J. Cancer 2016, 114, 713–715. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dees, E.C.; Baker, S.D.; O’Reilly, S.; Rudek, M.A.; Davidson, S.B.; Aylesworth, C.; Elza-Brown, K.; Carducci, M.A.; Donehower, R.C. A phase I and pharmacokinetic study of short infusions of UCN-01 in patients with refractory solid tumors. Clin. Cancer Res. 2005, 11, 664–671. [Google Scholar] [PubMed]
- Daud, A.I.; Ashworth, M.T.; Strosberg, J.; Goldman, J.W.; Mendelson, D.; Springett, G.; Venook, A.P.; Loechner, S.; Rosen, L.S.; Shanahan, F.; et al. Phase I dose-escalation trial of checkpoint kinase 1 inhibitor MK-8776 as monotherapy and in combination with gemcitabine in patients with advanced solid tumors. J. Clin. Oncol. 2015, 33, 1060–1066. [Google Scholar] [CrossRef]
- Calvo, E.; Braiteh, F.; Von Hoff, D.; McWilliams, R.; Becerra, C.; Galsky, M.D.; Jameson, G.; Lin, J.; McKane, S.; Wickremsinhe, E.R.; et al. Phase I Study of CHK1 Inhibitor LY2603618 in Combination with Gemcitabine in Patients with Solid Tumors. Oncology 2016, 91, 251–260. [Google Scholar] [CrossRef]
- Biau, J.; Devun, F.; Jdey, W.; Kotula, E.; Quanz, M.; Chautard, E.; Sayarath, M.; Sun, J.S.; Verrelle, P.; Dutreix, M. A preclinical study combining the DNA repair inhibitor Dbait with radiotherapy for the treatment of melanoma. Neoplasia 2014, 16, 835–844. [Google Scholar] [CrossRef] [Green Version]
- Biau, J.; Devun, F.; Verrelle, P.; Dutreix, M. Dbait: An innovative concept to inhibit DNA repair and treat cancer. Bull. Cancer 2016, 103, 227–235. [Google Scholar] [CrossRef]
- Biau, J.; Chautard, E.; Berthault, N.; de Koning, L.; Court, F.; Pereira, B.; Verrelle, P.; Dutreix, M. Combining the DNA Repair Inhibitor Dbait With Radiotherapy for the Treatment of High Grade Glioma: Efficacy and Protein Biomarkers of Resistance in Preclinical Models. Front. Oncol. 2019, 9, 549. [Google Scholar] [CrossRef]
- Ferreira, S.; Dutreix, M. DNA repair inhibitors to enhance radiotherapy: Progresses and limitations. Cancer Radiother. 2019, 23, 883–890. [Google Scholar] [CrossRef]
- Ferreira, S.; Foray, C.; Gatto, A.; Larcher, M.; Heinrich, S.; Lupu, M.; Mispelter, J.; Boussin, F.D.; Pouponnot, C.; Dutreix, M. AsiDNA Is a Radiosensitizer with no Added Toxicity in Medulloblastoma Pediatric Models. Clin. Cancer Res. 2020, 26, 5735–5746. [Google Scholar] [CrossRef]
- Le Tourneau, C.; Dreno, B.; Kirova, Y.; Grob, J.J.; Jouary, T.; Dutriaux, C.; Thomas, L.; Lebbe, C.; Mortier, L.; Saiag, P.; et al. First-in-human phase I study of the DNA-repair inhibitor DT01 in combination with radiotherapy in patients with skin metastases from melanoma. Br. J. Cancer 2016, 114, 1199–1205. [Google Scholar] [CrossRef] [PubMed]
- Devun, F.; Bousquet, G.; Biau, J.; Herbette, A.; Roulin, C.; Berger, F.; Sun, J.S.; Robine, S.; Dutreix, M. Preclinical study of the DNA repair inhibitor Dbait in combination with chemotherapy in colorectal cancer. J. Gastroenterol. 2012, 47, 266–275. [Google Scholar] [CrossRef] [PubMed]
- Horwitz, S.B. Taxol (paclitaxel): Mechanisms of action. Ann. Oncol. 1994, 5 (Suppl. 6), S3–S6. [Google Scholar]
- Dasari, S.; Tchounwou, P.B. Cisplatin in cancer therapy: Molecular mechanisms of action. Eur. J. Pharmacol. 2014, 740, 364–378. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhu, Q.; Wani, A.A. Histone modifications: Crucial elements for damage response and chromatin restoration. J. Cell. Physiol. 2010, 223, 283–288. [Google Scholar] [CrossRef] [Green Version]
- Soria, G.; Polo, S.E.; Almouzni, G. Prime, repair, restore: The active role of chromatin in the DNA damage response. Mol. Cell 2012, 46, 722–734. [Google Scholar] [CrossRef] [Green Version]
- Tjeertes, J.V.; Miller, K.M.; Jackson, S.P. Screen for DNA-damage-responsive histone modifications identifies H3K9Ac and H3K56Ac in human cells. EMBO J. 2009, 28, 1878–1889. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xie, W.; Song, C.; Young, N.L.; Sperling, A.S.; Xu, F.; Sridharan, R.; Conway, A.E.; Garcia, B.A.; Plath, K.; Clark, A.T.; et al. Histone h3 lysine 56 acetylation is linked to the core transcriptional network in human embryonic stem cells. Mol. Cell 2009, 33, 417–427. [Google Scholar] [CrossRef] [Green Version]
- Hamperl, S.; Bocek, M.J.; Saldivar, J.C.; Swigut, T.; Cimprich, K.A. Transcription-Replication Conflict Orientation Modulates R-Loop Levels and Activates Distinct DNA Damage Responses. Cell 2017, 170, 774–786. [Google Scholar] [CrossRef] [Green Version]
- Crossley, M.P.; Bocek, M.; Cimprich, K.A. R-Loops as Cellular Regulators and Genomic Threats. Mol. Cell 2019, 73, 398–411. [Google Scholar] [CrossRef] [Green Version]
- Allison, D.F.; Wang, G.G. R-loops: Formation, function, and relevance to cell stress. Cell Stress 2019, 3, 38–46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sollier, J.; Cimprich, K.A. Breaking bad: R-loops and genome integrity. Trends Cell Biol. 2015, 25, 514–522. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rodriguez, A.; D’Andrea, A. Fanconi anemia pathway. Curr. Biol. 2017, 27, R986–R988. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhao, H.; Zhu, M.; Limbo, O.; Russell, P. RNase H eliminates R-loops that disrupt DNA replication but is nonessential for efficient DSB repair. EMBO Rep. 2018, 19. [Google Scholar] [CrossRef] [PubMed]
- Tharkar-Promod, S.; Johnson, D.P.; Bennett, S.E.; Dennis, E.M.; Banowsky, B.G.; Jones, S.S.; Shearstone, J.R.; Quayle, S.N.; Min, C.; Jarpe, M.; et al. HDAC1,2 inhibition and doxorubicin impair Mre11-dependent DNA repair and DISC to override BCR-ABL1-driven DSB repair in Philadelphia chromosome-positive B-cell precursor acute lymphoblastic leukemia. Leukemia 2018, 32, 49–60. [Google Scholar] [CrossRef]
- Kakarougkas, A.; Downs, J.A.; Jeggo, P.A. The PBAF chromatin remodeling complex represses transcription and promotes rapid repair at DNA double-strand breaks. Mol. Cell. Oncol. 2015, 2, e970072. [Google Scholar] [CrossRef]
- Clouaire, T.; Legube, G. A Snapshot on the Cis Chromatin Response to DNA Double-Strand Breaks. Trends Genet. 2019, 35, 330–345. [Google Scholar] [CrossRef] [Green Version]
- Shanbhag, N.M.; Rafalska-Metcalf, I.U.; Balane-Bolivar, C.; Janicki, S.M.; Greenberg, R.A. ATM-dependent chromatin changes silence transcription in cis to DNA double-strand breaks. Cell 2010, 141, 970–981. [Google Scholar] [CrossRef] [Green Version]
- Kakarougkas, A.; Ismail, A.; Chambers, A.L.; Riballo, E.; Herbert, A.D.; Kunzel, J.; Lobrich, M.; Jeggo, P.A.; Downs, J.A. Requirement for PBAF in transcriptional repression and repair at DNA breaks in actively transcribed regions of chromatin. Mol. Cell 2014, 55, 723–732. [Google Scholar] [CrossRef] [Green Version]
- Vissers, J.H.; van Lohuizen, M.; Citterio, E. The emerging role of Polycomb repressors in the response to DNA damage. J. Cell Sci. 2012, 125, 3939–3948. [Google Scholar] [CrossRef] [Green Version]
- Johnson, D.P.; Spitz, G.S.; Tharkar, S.; Quayle, S.N.; Shearstone, J.R.; Jones, S.; McDowell, M.E.; Wellman, H.; Tyler, J.K.; Cairns, B.R.; et al. HDAC1,2 inhibition impairs EZH2- and BBAP-mediated DNA repair to overcome chemoresistance in EZH2 gain-of-function mutant diffuse large B-cell lymphoma. Oncotarget 2015, 6, 4863–4887. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Johnson, D.P.; Spitz-Becker, G.S.; Chakraborti, K.; Bhaskara, S. Assessment of epigenetic mechanisms and DNA double-strand break repair using laser micro-irradiation technique developed for hematological cells. EBioMedicine 2019, 43, 138–149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bhaskara, S.; Jacques, V.; Rusche, J.R.; Olson, E.N.; Cairns, B.R.; Chandrasekharan, M.B. Histone deacetylases 1 and 2 maintain S-phase chromatin and DNA replication fork progression. Epigenet. Chromatin 2013, 6, 27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sirbu, B.M.; Couch, F.B.; Feigerle, J.T.; Bhaskara, S.; Hiebert, S.W.; Cortez, D. Analysis of protein dynamics at active, stalled, and collapsed replication forks. Genes Dev. 2011, 25, 1320–1327. [Google Scholar] [CrossRef] [Green Version]
- Moldovan, G.L.; Pfander, B.; Jentsch, S. PCNA, the maestro of the replication fork. Cell 2007, 129, 665–679. [Google Scholar] [CrossRef] [Green Version]
- Wells, C.E.; Bhaskara, S.; Stengel, K.R.; Zhao, Y.; Sirbu, B.; Chagot, B.; Cortez, D.; Khabele, D.; Chazin, W.J.; Cooper, A.; et al. Inhibition of histone deacetylase 3 causes replication stress in cutaneous T cell lymphoma. PLoS ONE 2013, 8, e68915. [Google Scholar] [CrossRef]
- Ruan, K.; Yamamoto, T.G.; Asakawa, H.; Chikashige, Y.; Kimura, H.; Masukata, H.; Haraguchi, T.; Hiraoka, Y. Histone H4 acetylation required for chromatin decompaction during DNA replication. Sci. Rep. 2015, 5, 12720. [Google Scholar] [CrossRef] [Green Version]
- Sirbu, B.M.; McDonald, W.H.; Dungrawala, H.; Badu-Nkansah, A.; Kavanaugh, G.M.; Chen, Y.; Tabb, D.L.; Cortez, D. Identification of proteins at active, stalled, and collapsed replication forks using isolation of proteins on nascent DNA (iPOND) coupled with mass spectrometry. J. Biol. Chem. 2013, 288, 31458–31467. [Google Scholar] [CrossRef] [Green Version]
- Summers, A.R.; Fischer, M.A.; Stengel, K.R.; Zhao, Y.; Kaiser, J.F.; Wells, C.E.; Hunt, A.; Bhaskara, S.; Luzwick, J.W.; Sampathi, S.; et al. HDAC3 is essential for DNA replication in hematopoietic progenitor cells. J. Clin. Investig. 2013, 123, 3112–3123. [Google Scholar] [CrossRef] [Green Version]
- Rondinelli, B.; Gogola, E.; Yucel, H.; Duarte, A.A.; van de Ven, M.; van der Sluijs, R.; Konstantinopoulos, P.A.; Jonkers, J.; Ceccaldi, R.; Rottenberg, S.; et al. EZH2 promotes degradation of stalled replication forks by recruiting MUS81 through histone H3 trimethylation. Nat. Cell Biol. 2017, 19, 1371–1378. [Google Scholar] [CrossRef]
- Quivy, J.P.; Gerard, A.; Cook, A.J.; Roche, D.; Almouzni, G. The HP1-p150/CAF-1 interaction is required for pericentric heterochromatin replication and S-phase progression in mouse cells. Nat. Struct. Mol. Biol. 2008, 15, 972–979. [Google Scholar] [CrossRef] [PubMed]
- Collins, N.; Poot, R.A.; Kukimoto, I.; Garcia-Jimenez, C.; Dellaire, G.; Varga-Weisz, P.D. An ACF1-ISWI chromatin-remodeling complex is required for DNA replication through heterochromatin. Nat. Genet. 2002, 32, 627–632. [Google Scholar] [CrossRef] [PubMed]
- Fu, H.; Maunakea, A.K.; Martin, M.M.; Huang, L.; Zhang, Y.; Ryan, M.; Kim, R.; Lin, C.M.; Zhao, K.; Aladjem, M.I. Methylation of histone H3 on lysine 79 associates with a group of replication origins and helps limit DNA replication once per cell cycle. PLoS Genet. 2013, 9, e1003542. [Google Scholar] [CrossRef]
- Unterberger, A.; Andrews, S.D.; Weaver, I.C.; Szyf, M. DNA methyltransferase 1 knockdown activates a replication stress checkpoint. Mol. Cell. Biol. 2006, 26, 7575–7586. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cortez, D. Proteomic Analyses of the Eukaryotic Replication Machinery. Methods Enzymol. 2017, 591, 33–53. [Google Scholar] [CrossRef] [Green Version]
- Bhaskara, S.; Knutson, S.K.; Jiang, G.; Chandrasekharan, M.B.; Wilson, A.J.; Zheng, S.; Yenamandra, A.; Locke, K.; Yuan, J.L.; Bonine-Summers, A.R.; et al. Hdac3 is essential for the maintenance of chromatin structure and genome stability. Cancer Cell 2010, 18, 436–447. [Google Scholar] [CrossRef] [Green Version]
- Bhaskara, S.; Hiebert, S.W. Role for histone deacetylase 3 in maintenance of genome stability. Cell Cycle 2011, 10, 727–728. [Google Scholar] [CrossRef] [Green Version]
- Bhattacharya, S.; Piya, S.; Borthakur, G. Bromodomain inhibitors: What does the future hold? Clin. Adv. Hematol. Oncol. 2018, 16, 504–515. [Google Scholar]
- Yang, L.; Zhang, Y.; Shan, W.; Hu, Z.; Yuan, J.; Pi, J.; Wang, Y.; Fan, L.; Tang, Z.; Li, C.; et al. Repression of BET activity sensitizes homologous recombination-proficient cancers to PARP inhibition. Sci. Transl. Med. 2017, 9. [Google Scholar] [CrossRef] [Green Version]
- Pericole, F.V.; Lazarini, M.; de Paiva, L.B.; Duarte, A.; Vieira Ferro, K.P.; Niemann, F.S.; Roversi, F.M.; Olalla Saad, S.T. BRD4 Inhibition Enhances Azacitidine Efficacy in Acute Myeloid Leukemia and Myelodysplastic Syndromes. Front. Oncol. 2019, 9, 16. [Google Scholar] [CrossRef] [Green Version]
- Mio, C.; Gerratana, L.; Bolis, M.; Caponnetto, F.; Zanello, A.; Barbina, M.; Di Loreto, C.; Garattini, E.; Damante, G.; Puglisi, F. BET proteins regulate homologous recombination-mediated DNA repair: BRCAness and implications for cancer therapy. Int. J. Cancer 2019, 144, 755–766. [Google Scholar] [CrossRef] [PubMed]
- Muvarak, N.E.; Chowdhury, K.; Xia, L.; Robert, C.; Choi, E.Y.; Cai, Y.; Bellani, M.; Zou, Y.; Singh, Z.N.; Duong, V.H.; et al. Enhancing the Cytotoxic Effects of PARP Inhibitors with DNA Demethylating Agents—A Potential Therapy for Cancer. Cancer Cell 2016, 30, 637–650. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maifrede, S.; Martinez, E.; Nieborowska-Skorska, M.; Di Marcantonio, D.; Hulse, M.; Le, B.V.; Zhao, H.; Piwocka, K.; Tempera, I.; Sykes, S.M.; et al. MLL-AF9 leukemias are sensitive to PARP1 inhibitors combined with cytotoxic drugs. Blood Adv. 2017, 1, 1467–1472. [Google Scholar] [CrossRef] [PubMed]
- Karakashev, S.; Zhu, H.; Yokoyama, Y.; Zhao, B.; Fatkhutdinov, N.; Kossenkov, A.V.; Wilson, A.J.; Simpkins, F.; Speicher, D.; Khabele, D.; et al. BET Bromodomain Inhibition Synergizes with PARP Inhibitor in Epithelial Ovarian Cancer. Cell Rep. 2017, 21, 3398–3405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wong, M.; Polly, P.; Liu, T. The histone methyltransferase DOT1L: Regulatory functions and a cancer therapy target. Am. J. Cancer Res. 2015, 5, 2823–2837. [Google Scholar] [PubMed]
- Wood, K.; Tellier, M.; Murphy, S. DOT1L and H3K79 Methylation in Transcription and Genomic Stability. Biomolecules 2018, 8, 11. [Google Scholar] [CrossRef] [Green Version]
- Veloso, A.; Kirkconnell, K.S.; Magnuson, B.; Biewen, B.; Paulsen, M.T.; Wilson, T.E.; Ljungman, M. Rate of elongation by RNA polymerase II is associated with specific gene features and epigenetic modifications. Genome Res. 2014, 24, 896–905. [Google Scholar] [CrossRef] [Green Version]
- MacLeod, G.; Bozek, D.A.; Rajakulendran, N.; Monteiro, V.; Ahmadi, M.; Steinhart, Z.; Kushida, M.M.; Yu, H.; Coutinho, F.J.; Cavalli, F.M.G.; et al. Genome-Wide CRISPR-Cas9 Screens Expose Genetic Vulnerabilities and Mechanisms of Temozolomide Sensitivity in Glioblastoma Stem Cells. Cell Rep. 2019, 27, 971–986. [Google Scholar] [CrossRef] [Green Version]
- Dai, Y.; Zhang, A.; Shan, S.; Gong, Z.; Zhou, Z. Structural basis for recognition of 53BP1 tandem Tudor domain by TIRR. Nat. Commun. 2018, 9, 2123. [Google Scholar] [CrossRef]
- Zgheib, O.; Pataky, K.; Brugger, J.; Halazonetis, T.D. An oligomerized 53BP1 tudor domain suffices for recognition of DNA double-strand breaks. Mol. Cell. Biol. 2009, 29, 1050–1058. [Google Scholar] [CrossRef] [Green Version]
- Botuyan, M.V.; Lee, J.; Ward, I.M.; Kim, J.E.; Thompson, J.R.; Chen, J.; Mer, G. Structural basis for the methylation state-specific recognition of histone H4-K20 by 53BP1 and Crb2 in DNA repair. Cell 2006, 127, 1361–1373. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Paquin, K.L.; Howlett, N.G. Understanding the Histone DNA Repair Code: H4K20me2 Makes Its Mark. Mol. Cancer Res. 2018, 16, 1335–1345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kari, V.; Raul, S.K.; Henck, J.M.; Kitz, J.; Kramer, F.; Kosinsky, R.L.; Ubelmesser, N.; Mansour, W.Y.; Eggert, J.; Spitzner, M.; et al. The histone methyltransferase DOT1L is required for proper DNA damage response, DNA repair, and modulates chemotherapy responsiveness. Clin. Epigenet. 2019, 11, 4. [Google Scholar] [CrossRef] [Green Version]
- Zhu, B.; Chen, S.; Wang, H.; Yin, C.; Han, C.; Peng, C.; Liu, Z.; Wan, L.; Zhang, X.; Zhang, J.; et al. The protective role of DOT1L in UV-induced melanomagenesis. Nat. Commun. 2018, 9, 259. [Google Scholar] [CrossRef] [PubMed]
- Liu, W.; Deng, L.; Song, Y.; Redell, M. DOT1L inhibition sensitizes MLL-rearranged AML to chemotherapy. PLoS ONE 2014, 9, e98270. [Google Scholar] [CrossRef]
- Oksenych, V.; Zhovmer, A.; Ziani, S.; Mari, P.O.; Eberova, J.; Nardo, T.; Stefanini, M.; Giglia-Mari, G.; Egly, J.M.; Coin, F. Histone methyltransferase DOT1L drives recovery of gene expression after a genotoxic attack. PLoS Genet. 2013, 9, e1003611. [Google Scholar] [CrossRef]
Target | Drugs | Side Effects (Clinical Trial Identifier if Applicable) |
---|---|---|
ATM | AZD1390, KU-59403 | AZD1390—seizures, lung problems, muscle pain, dark urine, irregular heartbeat, low blood pressure, light sensitivity, pancreatic or abdominal pain, drop in blood cells counts, increased risk of infection and bleeding problems (NCT03423618) KU-59403—no major toxicity in mice, no clinical trials to date [57] |
ATR | AZD6738, VX-970 (M6620), schisandrin B | AZD6738—fatigue, anemia, thrombocytopenia, nausea (NCT02223923) [58] VX-970—no dose-limiting toxicities reported to date in phase I (NCT02487095) [59] schisandrin B—gastric distress, reduced appetite when used as a health supplement, no clinical trial data [60] |
PARP | rucaparib, olaparib, niraparib | Similar for all—fatigue, nausea and vomiting, anemia, constipation, increased cholesterol, liver and kidney problems, diarrhea, abdominal pain, decreased appetite (NCT00753545, NCT02655016 and many more) [61,62] |
CHK1/CHK2 | UCN-01, MK-8776, AZD7762, LY2603618 | UCN-01—nausea, vomiting, hypotension, hyperglycemia; has not moved past phase II trials due to low specificity (NCT00003289) [63] MK-8776—QTc prolongation, fatigue, nausea, constipation (NCT00779584) [64] AZD7762—failed phase I clinical trial due to cardiac toxicity (NCT00413686) [46] LY2603618—fatigue, decreased platelets, nausea, decreased neutrophils, decreased hemoglobin; has not moved past phase II as it was not more effective than standard of care (NCT01341457) [65] |
DNA-PK | VX-984, M3814, AZD7648, avadomide (CC-122) | VX-984—no data available from ongoing phase I trial (NCT02644278) M3814—fatigue, nausea, constipation, vomiting, decreased appetite, dysphagia, back and chest pain, diarrhea, mucosal inflammation (NCT02516813) AZD7648—no dose-limiting toxicity reported to date in ongoing phase I trial (NCT03907969) avadomide—fatigue, neutropenia, diarrhea [39] |
MRN (MRE11) | Mirin | no side effects reported in mice [56]; no human trials have begun |
DNA damage response (DDR) Pathways | AsiDNA (DT01) | no side effects yet reported in mice; in humans’ reversible grade 1 and 2 injection site reactions observed but no dose limiting toxicity in ongoing trials (NCT01469455, NCT03579628) [66,67,68,69,70] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Johnson, D.P.; Chandrasekharan, M.B.; Dutreix, M.; Bhaskara, S. Targeting DNA Repair and Chromatin Crosstalk in Cancer Therapy. Cancers 2021, 13, 381. https://doi.org/10.3390/cancers13030381
Johnson DP, Chandrasekharan MB, Dutreix M, Bhaskara S. Targeting DNA Repair and Chromatin Crosstalk in Cancer Therapy. Cancers. 2021; 13(3):381. https://doi.org/10.3390/cancers13030381
Chicago/Turabian StyleJohnson, Danielle P., Mahesh B. Chandrasekharan, Marie Dutreix, and Srividya Bhaskara. 2021. "Targeting DNA Repair and Chromatin Crosstalk in Cancer Therapy" Cancers 13, no. 3: 381. https://doi.org/10.3390/cancers13030381
APA StyleJohnson, D. P., Chandrasekharan, M. B., Dutreix, M., & Bhaskara, S. (2021). Targeting DNA Repair and Chromatin Crosstalk in Cancer Therapy. Cancers, 13(3), 381. https://doi.org/10.3390/cancers13030381