487
Views
1
CrossRef citations to date
0
Altmetric
Review

A patent review of topoisomerase I inhibitors (2016–present)

, , , , , & show all
Pages 473-508 | Received 05 Oct 2020, Accepted 18 Jan 2021, Published online: 29 Apr 2021

References

  • JB L, Champoux JJ. Human DNA topoisomerase I: relaxation, roles, and damage control. Chromosoma. 2005;114:75–85.
  • Martino E, Della Volpe S, Terribile E, et al. The long story of camptothecin: from traditional medicine to drugs. Bioorg Med Chem Lett. 2017;27(4):701–707.
  • Pommier Y. DNA topoisomerase I inhibitors: chemistry, biology, and interfacial inhibition. Chem Rev. 2009;109:2894–2902.
  • Reguera RM, Redondo CM, Gutierrez de Prado R, et al. DNA topoisomerase I from parasitic protozoa: a potential target for chemotherapy. Biochim Biophys Acta. 2006;1759:117–131.
  • Tse-Dinh YC. Targeting bacterial topoisomerase I to meet the challenge of finding new antibiotics. Future Med Chem. 2015;7:459–471.
  • Pommier Y. DNA topoisomerases and cancer. New York: Springer; 2012.
  • Wang JC. Interaction between DNA and an Escherichia coli protein ω. J Mol Biol. 1971;55:523–533.
  • Champoux JJ. DNA topoisomerases: structure, function, and mechanism. Annu Rev Biochem. 2001;70:369–413.
  • Pommier Y, Leo E, Zhang HL, et al. DNA topoisomerases and their poisoning by anticancer and antibacterial drugs. Chem Biol. 2010;17:421–433.
  • Barros FWA, Bezerra DP, Ferreira PMP, et al. Inhibition of DNA topoisomerase I activity and induction of apoptosis by thiazacridine derivatives. Toxicol Appl Pharmacol. 2013;268:37–46.
  • Kim S-H, Lee E, Baek KH, et al. Chalcones, inhibitors for topoisomerase I and cathepsin B and L, as potential anti-cancer agents. Bioorg Med Chem Lett. 2013;23:3320–3324.
  • Forterre P, Gribaldo S, Gadelle D, et al. Origin and evolution of DNA topoisomerases. Biochimie. 2007;89:427. (b) Schoeffler AJ, Berger JM. DNA topoisomerases: harnessing and constraining energy to govern chromosome topology. Q Rev Biophys 2008, 41, 41–101
  • Thomas A, Pommier Y. Targeting Topoisomerase I in the era of precision medicine. Clin Cancer Res. 2019;25:6581–6589.
  • Staker BL, Feese MD, Cushman M, et al. Structures of three classes of anticancer agents bound to the human Topoisomerase I−DNA covalent complex. Journal of Medicinal Chemistry. 2005;48(7):2336–2345.
  • Hsiang YH, Liu LF. Identification of mammalian DNA topoisomerase I as an intracellular target of the anticancer drug camptothecin. Cancer Res. 1988;48:1722–1726.
  • Hsiang YH, Hertzberg R, Hecht S, et al. Camptothecin induces protein‐linked DNA breaks via mammalian DNA topoisomerase I. J Biol Chem. 1985;27:14873–14878.
  • Postma C, Koopman M, Buffart TE, et al. DNA copy number profiles of primary tumors as predictors of response to chemotherapy in advanced colorectal cancer. Ann Oncol. 2009;20:1048–1056.
  • Hatefi A, Amsden B. Camptothecin delivery methods. Pharm Res. 2002;19:1389–1399.
  • Li F, Jiang T, Li Q, et al. Camptothecin (CPT) and its derivatives are known to target topoisomerase I (TopI) as their mechanism of action: did we miss something in CPT analogue molecular targets for treating human disease such as cancer? Am. J. Cancer Res.2017;7:2350–2394. .
  • Pommier Y. Topoisomerase I inhibitors: camptothecins and beyond. Nat Rev Cancer. 2006;6:789–802.
  • Ikeguchi M, Arai Y, Maeta Y, et al. Topoisomerase I expression in tumors as a biological marker for CPT-11 chemosensitivity in patients with colorectal cancer. Surg Today. 2011;41:1196–1199.
  • Wethington SL, Wright JD, Herzog TJ. Key role of topoisomerase I inhibitors in the treatment of recurrent and refractory epithelial ovarian carcinoma. Expert Rev Anticancer Ther. 2008;8:819–831.
  • Fu P, Zhuang Y, Wang Y, et al. New indolocarbazoles from a mutant strain of the marine-derived actinomycete Streptomyces fradiae 007M135. Org Lett. 2012;14:6194–6197.
  • Sánchez C, Méndez C, Salas JA. Indolocarbazole natural products: occurrence, biosynthesis, and biological activity. Nat Prod Rep. 2006;23:1007–1045.
  • Omura S, Iwai Y, Hirano A, et al. A new alkaloid AM-2282 OF Streptomyces origin. Taxonomy, fermentation, isolation and preliminary characterization. J Antibiot. 1977;30:275–282.
  • Saif MW, Sellers S, Diasio RB, et al. A phase I dose‐escalation study of edotecarin (J‐107088) combined with infusional 5‐fluorouracil and leucovorin in patients with advanced/metastatic solid tumors. Anti‐Cancer Drugs 2010;21:716–723.
  • Cagir A, Jones SH, Gao R, et al. A naturally occurring human DNA topoisomerase I poison. J Am Chem Soc. 2003;125:13628–13629.
  • Iwasaki H, Inafuku M, Taira N, et al. Tumor‐selective cytotoxicity of nitidine results from its rapid accumulation into mitochondria. BioMed. Res. Intl.. 2017;2017:1–10.
  • Ivanova B, Spiteller M. Substituted benzo[i]phenanthridines as promising topoisomerase-I non-camptothecin targeting agents: an experimental and theoretical study. Medicinal Chemistry Research. 2013;22(11):5204–5217.
  • Feng W, Satyanarayana M, Tsai Y-C, et al. LaVoie EJ.12-Substituted 2,3-dimethoxy-8,9-methylenedioxybenzo[i]phenanthridines as novel topoisomerase I-targeting antitumor agents. Bioorg Med Chem. 2009;17:2877–2885.
  • Clark RL, Deane FM, Anthony NG, et al. Exploring DNA topoisomerase I inhibition by the benzo[c]phenanthridines fagaronine and ethoxidine using steered molecular dynamics. Bioorg Med Chem. 2007;15:4741–4752.
  • Fang SD, Wang LK, Hecht SM. Inhibitors of DNA topoisomerase I isolated from the roots of zanthoxylum nitidum. The Journal of Organic Chemistry. 1993;58(19):5025–5027.
  • Arthur HR, Hui WH, Ng YL. An examination of the rutaceae of Hong Kong. Part III. The alkaloid, avicine, from Zanthoxylum avicennae. J Chem Soc. 1959;4007–4009.
  • Castelli S, Kathar P, Vassallo O, et al. Anti-cancer agents. Med Chem. 2013;13:356–363.
  • Marco E, Laine W, Tardy C, et al. Molecular determinants of topoisomerase I poisoning by lamellarins: comparison with camptothecin and structure‐activity relationships. J Med Chem. 2005;48:3807.
  • Facompre M, Tardy C, Bal-Mahieu C, et al. Lamellarin D: a novel potent inhibitor of topoisomerase I. Cancer Res. 2003;63:7392–7399.
  • Cinelli MA, Morrell AE, Dexheimer TS, et al. The structure-activity relationships of A-ring-substituted aromathecin topoisomerase I inhibitors strongly support a camptothecin-like binding mode. Bioorg Med Chem. 2010;18:5535–5552.
  • Kiselev E, Sooryakumar D, Agama K, et al. Optimization of the lactam side chain of 7‐azaindenoisoquinoline topoisomerase I inhibitors and mechanism of action studies in cancer cells. J Med Chem. 2014;57:1289–1298.
  • Beck DE, Reddy PVN, Lv W, et al. Investigation of the structure‐activity relationships of aza‐A‐ring indenoisoquinoline topoisomerase I poisons. J Med Chem. 2016;59:3840–3853.
  • Cushman M, Cheng L. Stereoselective oxidation by thionyl chloride leading to the indeno[1,2‐c]isoquinoline system. J Org Chem. 1978;43:3781–3783.
  • Beck DE, Agama K, Marchand C, et al. Synthesis and biological evaluation of new carbohydrate substituted indenoisoquinoline topoisomerase I inhibitors and improved syntheses of the experimental anticancer agents indotecan (LMP400) and indimitecan (LMP776). J Med Chem. 2014;57:1495–1512.
  • Burton JH, Mazcko C, Leblanc A, et al. NCI comparative oncology program testing of non-camptothecin indenoisoquinoline topoisomerase i inhibitors in naturally occurring canine lymphoma. Clin Cancer Res. 2018;24:5830–5840.
  • O’Sullivan Coyne GH, Kummar S, Meehan RS, et al. Phase I study of indenoisoquinolines LMP776 in adults with relapsed solid tumors and lymphomas. Journal of Clinical Oncology. 2017;35(15_suppl):2558.
  • Ruchelman AL, Singh SK, Wu X, et al. Diaza- and triazachrysenes: potent topoisomerase-targeting agents with exceptional antitumor activity against the human tumor xenograft, MDA-MB-435. Bioorg. Med Chem Lett. 2002;12:3333–3336.
  • Kiselev E, Dexheimer TS, Pommier Y, et al. Synthesis and Evaluation of Dibenzo[c,h][1,6]naphthyridines as Topoisomerase I inhibitors and potential anticancer agents. J Med Chem. 2010;53:8716–8726.
  • Alonso C, Fuertes M, Gonzalez M, et al. Synthesis and biological evaluation of 1,5-naphthyridines as topoisomerase I inhibitors. A new family of antiproliferative agents. Curr Top Med Chem. 2014;14:2722–2728.
  • Alonso C, Fuertes M, Gonzalez M, et al. Synthesis and biological evaluation of indeno[1,5]naphthyridines as topoisomerase I (TopI) inhibitors with antiproliferative activity. European Journal of Medicinal Chemistry. 2016;115: 179–190.
  • Tejeria A, Perez-Pertejo Y, Reguera RM, et al. Substituted 1,5-naphthyridines derivatives as novel Antileishmanial agents. Synthesis and biological evaluation. Eur J Med Chem. 2018;152:137–147.
  • Tejeria A, Perez-Pertejo Y, Reguera RM, et al. Antileishmanial effect of new indeno-1,5-naphthyridines, selective inhibitors of Leishmania infantum type IB DNA topoisomerase. Eur J Med Chem. 2016;124:740–749.
  • Andersen MB, Tesauro C, Gonzalez M, et al. Advantages of an optical nanosensor system for mechanistic analysis of a novel topoisomerase I targeting drug: a case story. Nanoscale. 2017;9:1886–1895.
  • Martín-Encinas E, Rubiales G, Knudsen BR, et al. Straightforward synthesis and biological evaluation as topoisomerase I inhibitors and antiproliferative agents of hybrid chromeno[4,3-b][1,5]naphthyridines and chromeno[4,3-b][1,5]naphthyridin-6-ones. Eur J Med Chem. 2019;178:752–766.
  • Martín-Encinas E, Selas A, Rubiales G, et al. Synthesis of novel hybrid quinolino[4,3-b][1,5]naphthyridines and quinolino[4,3-b][1,5]naphthyridin-6(5H)-one derivatives and biological evaluation as topoisomerase I inhibitors and antiproliferatives. Eur J Med Chem. 2020;195:112292.
  • Alonso C, Fuertes M, Martín-Encinas E, et al. Novel topoisomerase I inhibitors. Syntheses and biological evaluation of phosphorus substituted quinoline derivates with antiproliferative activity. Eur J Med Chem. 2018;149:225–237.
  • Tejeria A, Perez-Pertejo Y, Reguera RM, et al. Antileishmanial activity of new hybrid tetrahydroquinoline derivatives with phosphorus substituents. Eur J Med Chem. 2019;162:18–31.
  • Skok Z, Zidar N, Kikelj D, et al. Dual inhibitors of human DNA topoisomerase II and other cancer-related targets. J Med Chem. 2020;63:884–904.
  • Liang X, Wu Q, Luan S, et al. A comprehensive review of topoisomerase inhibitors as anticancer agents in the past decade. Eur J Med Chem. 2019;171:129–168.
  • Hevenern KE, Verstak TA, Lutat KE, et al. Recent developments in topoisomerase-targeted cancer chemotherapy. Acta Pharm Sin B. 2018;8:844–861.
  • Capranico G, Marinello J, Chillemi G. Type I DNA Topoisomerases. J Med Chem. 2017;60:2169–2192.
  • Kathiravan MK, Kale AN, Nilewar S. Discovery and development of topoisomerase inhibitors as anticancer agents. Mini-Rev Med Chem. 2016;16:1229.
  • D’Annessa I, Castelli S, Desideri A. Topoisomerase 1B as a target against Leishmaniasis. Mini-Rev Med Chem. 2015;15:203–210.
  • Khadka DB, Cho W-J. Topoisomerase inhibitors as anticancer agents: a patent update. Expert Opin Ther Pat. 2013;23:1033–1056.
  • Singh A, Kaur N, Singh G, et al. Topoisomerase I and II inhibitors: a patent review. Recent Pat. Anti-Cancer Drug Discov.. 2016;11:401–423.
  • Wang C, Shao C, Wang M, et al. Application of phenol derivatives in preparation of topoisomerase I inhibitor. Faming Zhuanli Shenqing CN 108938605, 2018.
  • Wang C, Shao C, Wang M, et al. Marine fungus separated from gorgonian and its application in preparing topoisomerase I inhibitor. Faming Zhuanli Shenqing CN 108929857, 2018.
  • Wang C, Shao C, Xin L, et al. Application of flavonoid compound in preparation of topoisomerase I inhibitor. Faming Zhuanli Shenqing CN 108926553, 2018.
  • Nishizawa M, Emura M, Kan Y, et al. Macrocarpals: HIV-RTase inhibitors of Eucalyptus globulus. Tetrahedron Lett. 1992;33:2983–2986.
  • Shou Q, Smith JE, Mon H, et al. Rhodomyrtals A-D, four unusual phloroglucinol-sesquiterpene adducts from Rhodomyrtus psidioides. RSC Adv. 2014;4:13514–13517.
  • S-P Y, X-W Z, Ai J, et al. Potent HGF/c-Met axis inhibitors from Eucalyptus globulus: the coupling of phloroglucinol and sesquiterpenoid is essential for the activity. J Med Chem. 2012;55:8183–8187.
  • Liu N, Ni W, Jin L, et al. Novel topoisomerase I inhibitor, and its pharmaceutical composition, preparation method and application in preparing drugs for treating cancer and complications and preparing functional health-care product. Faming Zhuanli Shenqing CN 107986951, 2018.
  • Tang Y, Duan J, Le S Quinoid chalcone-C-glycoside dimer compound having antitumor and antiinflammatory activity. PCT Int. Appl. WO 2017219510, 2017.
  • Tang Y, Yue S, Duan J, et al. Quinoid chalcone-C-glycoside dimer compound with antitumor and antiinflammatory activity. Faming Zhuanli Shenqing CN 106083788, 2016.
  • Tang Y, Duan J, Le S Quinoid chalcone and flavonol conjugate having antitumor activity and antiinflammatory activity, preparation method therefor, and application thereof. PCT Int. Appl. WO 2017219509, 2017.
  • Tang Y, Yue S, Duan J, et al. Preparation method of quinone chalcone and flavonol conjugate with antitumor and antiinflammatory activity, and application thereof. Faming Zhuanli Shenqing CN 106146581, 2016.
  • Guo M, Chen G, Tian Y, et al. Method for fast screening topoisomerase I inhibitor from natural product. Faming Zhuanli Shenqing CN 105717250, 2016.
  • Weng J, Kong Y, Pang K, et al. Preparation method of adamantane formamido ethyl formate as antitumor agents. Faming Zhuanli Shenqing CN 110423216, 2019.
  • Weng J, Kong Y, Wen Y, et al. 2-substituted-phenyl-3-(1-adamantane formamido)-4-thiazoline ketone compounds and its synthetic method and application. Faming Zhuanli Shenqing CN 110407766, 2019.
  • Weng J, Wen Y, Kong Y, et al. Adamantane formamide compound and its preparation method and application. Faming Zhuanli Shenqing CN 110790677, 2019.
  • Weng J, Ye F, Zhu Y, et al. Preparation of stilbene analogues with thiazole structure as antitumor agents. Faming Zhuanli Shenqing CN 110407767, 2019.
  • Yuan J, Novel GG I-type topoisomerase inhibitor. Faming Zhuanli Shenqing CN 110845389, 2020.
  • Wang J, Yun D, Yao J, et al. Design, synthesis and QSAR study of novel isatin analogues inspired Michael acceptor as potential anticancer compounds Jiabing. Eur J Med Chem. 2018;144:493–503.
  • Liu J, Geng G, Liang G, et al. A novel topoisomerase I inhibitor DIA-001 induces DNA damage mediated cell cycle arrest and apoptosis in cancer cell. Ann Transl Med. 2020;8:89–97.
  • Talukdar A, Das BB, Kundu B, et al. Bicycle topoisomerase I inhibiting compounds, process for preparation and use thereof. Indian Pat. Appl. IN 201811020003, PCT Int. Appl. WO2019229765A1, 2019.
  • Kundu B, Das SK, Chowdhuri SP, et al. Discovery and mechanistic study of tailor-made quinoline derivatives as Topoisomerase 1 poison with potent anticancer activity. J Med Chem. 2019;62:3428–3446.
  • Kerns RJ, Towle T, Hiasa H Quinolone-based compounds with anticancer activity. PCT Int. Appl. WO 2018107112, 2018.
  • Hergenrother PJ, Riley AP Topoisomerase inhibitors with antibacterial and anticancer activity. PCT Int. Appl. WO 2018237140, 2018.
  • Huang H-S, Yu D-S, Chen T-C Thiochromeno(2,3-c)quinolin-12-one derivatives, preparation method and application thereof. Can. Pat. Appl. CA 2866502, 2016.
  • Huang H-S, Yu D-S, Chen T-C Thiochromeno[2,3-c]quinolin-12-one derivatives and their use as topoisomerase inhibitors. Eur. Pat. Appl. EP 3002287, 2016.
  • Liu Y 10-Hydroxycamptothecin derivative, synthesis method and application thereof. Faming Zhuanli Shenqing CN 106588946, 2017.
  • Huang Y, Wu A, Zhang Y Synthetic method of topoisomerase I inhibitor. Faming Zhuanli Shenqing CN 106674106, 2017.
  • Cho WJ, Min SY, Le TN, et al. Synthesis of new 3-arylisoquinolinamines: effect on topoisomerase I inhibition and cytotoxicity. Bioorg Med Chem Lett. 2003;13:4451–4454.
  • Cho WJ Preparation of diarylisoquinolones and diarylisoquinolines as topoisomerase inhibitors Repub. Korean Kongkae Taeho Kongbo KR 2017049742, 2017.
  • Cushman MS, Pommier YG Alcohol-, diol-, and carbohydrate-substituted indenoisoquinolines as topoisomerase I inhibitors. U.S. Pat. Appl. Publ. US 20160318946, 2016.
  • Shapiro SL, Geiger K, Youlus J, et al. Indandiones A modified Dieckman reaction. J Org Chem. 1961;26:3580–3582.
  • Cushman MS, Morrell AE, Nagarajan PY N-substituted indenoisoquinolines and synthesis thereof. U.S. Pat. Appl. Publ. US 8053443, 2011.
  • Cushman MS, Lv P Prodrugs of anticancer agents indotecan and indimitecan. PCT Int. Appl. WO 2017136616, 2017.
  • Cushman MS, Beck DE N-substituted indenoisoquinolines and synthesis thereof. U.S. Pat. Appl. Publ. US 9796753, 2017.
  • Cushman MS, Beck DE, Pommier Y Aza-a-ring indenoisoquinolines as topoisomerase I poisons. PCT Int. Appl. WO 2017160898, 2017.
  • Cushman MS, Wang P, Pommier Y, et al. Azaindenoisoquinoline compounds and uses thereof. PCT Int. Appl. WO 2018118852, 2018.
  • Pan C, Kong S, Su G, et al. 2-(ω-Dialkylamino)aminoalkyl-3-(aryl-fused azolyl)quinoxaline compound as inhibiting topoisomerase I inhibitor useful in treatment of cancer and its preparation. Faming Zhuanli Shenqing. CN 108467392A, 2018.
  • Wu L, Zhang C A kind of β lapachols list star element heterozygote and preparation method thereof and medical usage. Faming Zhuanli Shenqing CN 105461641, 2016.
  • Wu L, Zhang C, Chao S O-Naphthoquinone derivative as topoisomerase I inhibitor and its preparation, pharmaceutical compositions and use in the treatment of cancer. Faming Zhuanli Shenqing CN 105541872, 2016.
  • Sheng C, Zhang W, Dong G, et al. Preparation method of 10-hydroxyevodiamine anti-tumor compound and its application in preparation of anticancer drugs and topoisomerase inhibitor. Faming Zhuanli Shenqing CN 105418610, 2016.
  • Sheng C, Dong G, Huang Y, et al. Rutaecarpin derivative and its preparation method and application with multiple target point antitumor activity. Faming Zhuanli Shenqing CN 108794474, 2018.
  • Dong G, Sheng C, Chen S, et al. Rutaecarpin derivative of multiple target point anti-tumor activity and the preparation method and application thereof. Faming Zhuanli Shenqing CN 110066281, 2019.
  • Epstein S, Zhang B, He S, et al. Preparation method of 4-methylamino acridine-N-phenylbenzamide as antitumor agents. Faming Zhuanli Shenqing CN 110407747, 2019.
  • Zhang B, He S, Wang N, et al. N-o-substituted phenyl benzamide 4-methylamino acridine compound and preparation method and application thereof. Faming Zhuanli Shenqing CN 111057004, 2020.
  • Song W, Wan M, Li S, et al. Phenanthridine derivative useful in treatment of cancer and its preparation. Faming Zhuanli Shenqing CN 108727397, 2018.
  • An LK, Zhang X, Wang H, et al. Oxynitidine derivatives useful as inhibitors of topoisomerase IB (TOP1) and tyrosyl-DNA phosphodiesterase 1 (TDP1). PCT Int. Appl.WO 2020023700, 2020.
  • Zhang X-R, Wang H-W, Tang W-L, et al. Discovery, synthesis, and evaluation of oxynitidine derivatives as dual inhibitors of DNA topoisomerase IB (TOP1) and tyrosyl-DNA phosphodiesterase 1 (TDP1), and potential antitumor agents. J Med Chem. 2018;61:9908–9930.
  • Clement B, Meier C, Steinhauer TN Novel pyrido-phenanthroline derivatives, production and use thereof as medicaments. Eur. Pat. Appl. EP 3330270; PCT Int. Appl. WO 2018099814, 2018.
  • Pan C, Su G, Kong S, et al. Naphtho[1,2-h][1,6]naphthyridin-3(4H)-one compounds as topoisomerase I inhibitors and their preparation, pharmaceutical compositions and use in the treatment of cancer. Faming Zhuanli Shenqing CN 106243103, 2016.
  • Zou H, Cai Y, Zhu H Preparing method and application of tetrahydro-angustine derivative including three R substituents. CN 107936021A, 2018.
  • Zou H, Cai Y, Zhu H Preparation of (S)-tetrahydrogenangustine derivative as antitumor drug. CN 107827888A, 2018.
  • Kim JS, Shin WS, Han JY Tumor targeting phototherapeutic agent and method for manufacturing thereof. Repub. Korea KR 1750658, 2017.
  • Uesugi M, Di M, Kawase E Preparation of 6-amino-9-phenyl-3H-xanthen-3-imine and 9-phenyl-3-oxo-3H-xanthen-6-ol derivatives as selective fluorescent probes for removing human pluripotent stem cells. PCT Int. Appl. WO 2018043567, 2018.
  • Mohan MA Compositions and methods for the treatment of cancer. PCT Int. Appl. WO 2016203352, 2016.
  • Kan P, Hung C, Hong K, et al. Pharmaceutical compositions of hydrophobic camptothecin derivatives. U.S. Pat. Appl. Publ. US 20190321356, 2019.
  • Santi DV, Fontaine S Synergistic cancer treatment. PCT Int. Appl. WO 2019140271, 2019.
  • Kuo DM Pharmaceutical composition containing topoisomerase inhibitor and antimetabolite for decreasing side effects of pancreatic cancer drug, and its preparation method. Taiwan. TW I634901, 2018.
  • Park JS, Jang MS, Oh WG Method for enhancing anticancer efficacy of DNA topoisomerase 1 inhibitor by using autophagocytosis regulating mechanism of methylenebis and related compounds. Repub. Korean Kongkae Taeho Kongbo KR 2019054423, 2019.
  • Camacho KM, Menegatti S, Kumar S, et al. Polymer-drug conjugates for combination anticancer therapy. PCT Int. Appl. WO 2016145096A1, 2016.
  • Friedman P, Eliasof S, Senderowicz A, et al. Treatment of cancer. PCT I nt. Appl. WO 2018102769A1, 2018.
  • Pan D, Geng B Graphene base for targeting DNA major groove and inhibiting topoisomerase as well as preparation method and application thereof. Faming Zhuanli Shenqing CN 110917212A, 2020.
  • Nandi A, Ghosh C, Basu S. Polymer conjugated graphene-oxide nanoparticles impair nuclear DNA and Topoisomerase I in cancer. Nanoscale Adv. 2019;4965.
  • Wang X, Yan X. Analyte-driven self-assembly of graphene oxide sheets onto hydroxycamptothecin-functionalized upconversion nanoparticles for the determination of type I topoisomerases in cell extracts. Anal Bioanal Chem. 2018;410:6761–6769.
  • Zuccaro L, Tesauro C, Kurkina T, et al. Real-time label-free direct electronic monitoring of topoisomerase enzyme binding kinetics on graphene. ACS Nano. 2015;9:11166–11176.
  • Goldenberg DM, Govindan SV Therapy of small-cell lung cancer (SCLC) with a topoisomerase-I inhibiting antibody-drug conjugate (ADC) targeting trop-2. PCT Int. Appl. WO 2018156634A1, 2018.
  • Goldenberg DM, Govindan SV Therapy of small-cell lung cancer with a topoisomerase-I inhibiting antibody-drug conjugate targeting Trop-2. U.S. Pat. Appl. Publ. US 20180185351A1, 2018.
  • Hwu P, McKenzie JA, Mbofung RN, et al. Combination of topoisomerase-I inhibitors with immunotherapy in the treatment of cancer. PCT Int. Appl. WO 2017049199A1, 2017.
  • Harki DA, Kaufmann SH, Perkins-Harki AL Methods and materials for assessing enzyme-nucleic acid complexes. U.S. Pat. Appl. Publ. US 20170003289A1, 2017.
  • Bacha JA, Brown DM, Steinoe A, et al. Use of dianhydrogalactitol or analogs and derivatives in combination with a p53 modulator or a PARP inhibitor. PCT Int. Appl. WO 2018160758A1, 2018.
  • Blanchette SF, Drummond DC, Fitzgerald JB, et al. Combination therapy for cancer treatment. PCT Int. Appl. WO 2017031445A1, 2017.
  • Blanchette SF, Drummond DC, Fitzgerald JB, et al. Combination therapy using liposomal irinotecan and a PARP inhibitor for cancer treatment. PCT Int. Appl. WO 2017031442A1, 2017.
  • Curley M, Louis CU Combination of an erbb3 inhibitor, topoisomerase I inhibitor, and an alkylating agent to treat cancer. PCT Int. Appl. WO 2018045256A1, 2018.
  • Brake RL, Kannan K, Xu Q Combination therapy including a RAF inhibitor for the treatment of colorectal cancer. PCT Int. Appl. WO 2017066664A1, 2017.
  • Strum JC, Bisi JE, Roberts PJ, et al. Treatment of Rb-negative tumors using topoisomerase inhibitors in combination with cyclin dependent kinase 4/6 inhibitors. PCT Int. Appl. WO 2016040848A1, 2016.
  • Chen L, Wang K, Qin G Method for predicting homologous recombination deletion mechanism and response of patient to cancer therapies. Faming Zhuanli Shenqing CN 107287285A, 2017.
  • Bruenner NA, Stenvang J, Budinska E, et al. Companion diagnostics for colorectal cancer. PCT Int. Appl.WO 2019068755A1, 2019.
  • Kobayashi Y, Watanabe N, Kurihara H, et al. Biomarker for camptothecin analog sensitivity as antitumor agent. Jpn. Kokai Tokkyo Koho JP 2016169945A, 2016.
  • Hergenrother PJ, Riley AP Topoisomerase inhibitors with antibacterial and anticancer activity. PCT Int. Appl. WO 2018237140A1, 2018.
  • Tse-Dinh Y-C SD Bacterial topoisomerase I inhibitors with antibacterial activity. U.S. Pat. Appl. Publ. US 20180079757A1, 2018.
  • Y-C T-D, Giulianotti MA, Houghten R Bacterial topoisomerase inhibitors and use thereof. U.S. Pat. Appl. Publ. US 20170298004A1, 2017.
  • Yang Z, Jiang T, Zhong H, et al. Inhibitor of bacterial type I topoisomerase and application. Faming Zhuanli Shenqing CN 106086023B, 2016.
  • Nagaraja V, Ekins S, Godbole AA Treating mycobacterium tuberculosis infection with topoisomerase I inhibitors. Indian Pat. Appl. IN 2015CH00652, 2016.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.