Camptothecin and its Analogues as Putative Bruton Tyrosine Kinase (BTK) Inhibitors in Cancer Therapy - Biophysical Simulations of Wild Type and C481S Mutation

Authors

  • Abdul Rashid Issahaku Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4001, South Africa
  • Mahmoud Soliman Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4001, South Africa

DOI:

https://doi.org/10.62382/jcbt.v1i1.10

Keywords:

Camptothecin, Natural Anti-cancer, Chinese Traditional Medicine, Drug Repurposing, Bruton’s Tyrosine Kinase

Abstract

Camptothecin (CPT) and its derivatives are extracted from the Chinese tree Camptotheca acuminata and have been long known for their significant antitumor activity. The widely reported primary mechanism of their anti-cancer activity is through inhibition of topoisomerase I (Topo I), a key anticancer target. However, there is a lack of research on other possible mechanisms of action of CPT and its analogues (CPT/analogues). In this report, we investigated the potential inhibitory mechanism of CPT/derivatives against Bruton’s tyrosine kinase (BTK) ‒ a crucial protein for regulating various cellular functions and is essential for B-cell growth and cyclical division. The binding mechanism of CPT and its analogues (Irinotecan, Diflomotecan, and Topotecan) against BTK was investigated using molecular docking, molecular dynamics simulations and thermodynamic binding free energy calculations. NRX-0492, a potent orally active degrader of both wild-type and mutant BTK, was used as a reference/control inhibitor. We also included a set of active binders (actives) and non-binders (inactives) to distinguish between effective binders and false positives to ensure the validity of our results. Binding affinity analysis suggested that CPT and its analogues could potentially bind to BTK comparably to NRX-0492. The studied compounds demonstrated stable binding with the protein throughout the simulation time via hydrogen bonds, π-π interactions, van der Waals forces and π-sulfur interactions. Irinotecan exhibited a binding affinity of ΔGbinding = –42.4±5.3 kcal/mol, closely matching that of NRX-0492 (ΔGbinding = –48.8±3.4 kcal/mol). Furthermore, the activity of CPT/derivatives was examined against the C481S mutant. Although the activity was slightly decreased due to the mutation, the compounds retained a promising binding affinity when compared to the results of NRX-0492. The findings of this study provide foundation for future experimental investigation of unexplored potential anti-cancer mechanism of CPT/derivatives and offer a new perspective on repurposing of natural products in cancer therapy.

Downloads

Download data is not yet available.

References

Newman DJ, Cragg GM. Natural Products as Sources of New Drugs over the Nearly Four Decades from 01/1981 to 09/2019. Journal of Natural Products. 2020, 83(3), 770–803.

Li F, Jiang T, Li Q, Ling X. Camptothecin (CPT) and its derivatives are known to target topoisomerase I (Top1) as their mechanism of action: did we miss something in CPT analogue molecular targets for treating human disease such as cancer? American Journal of Cancer Research. 2017, 7(12), 2350-2394.

Moertel CG, Schutt AJ, Reitemeier RJ, Hahn RG. Phase II study of camptothecin (NSC-100880) in the treatment of advanced gastrointestinal cancer. Cancer Chemotherapy Reports. 1972, 56(1), 95-101.

Gottlieb JA, Guarino AM, Call JB, Oliverio VT, Block JB. Preliminary pharmacologic and clinical evaluation of camptothecin sodium (NSC-100880). Cancer Chemotherapy Reports. 1970, 54(6), 461-70.

Cao J, Qi F, Liu T. Adjuvant chemotherapy after curative resection for gastric cancer: A meta-analysis. Scandinavian Journal of Gastroenterology. 2014, 49, 690-704.

Almeida A, Fernandes E, Sarmento B, Lúcio M. A Biophysical Insight of Camptothecin Biodistribution: Towards a Molecular Understanding of Its Pharmacokinetic Issues. Pharmaceutics. 2021, 13(6), 869.

Liu LF, Desai SD, Li TK, Mao Y, Sun M, et al. Mechanism of action of camptothecin. Annals of the New York Academy of Sciences. 2000, 922, 1-10.

Legarza K, Yang LX. The Binding Site of the Cleavable Complex New Molecular Mechanisms of Action of Camptothecin-type Drugs. Anticancer Research. 2006, 26(5A), 3301-5.

Abelson HT, Penman S. Selective interruption of high molecular weight RNA synthesis in HeLa cells by camptothecin. Nature Cell Biology. 1972, 237(74), 144-6.

Kessel D. Effects of camptothecin on RNA synthesis in leukemia L1210 cells. Biochimica et Biophysica Acta (BBA). 1971, 246(2), 225-32.

Liu YQ, Li WQ, Morris-Natschke SL, Qian K, Yang L, et al. Perspectives on biologically active camptothecin derivatives. Medicinal Research Reviews. 2015, 35(4), 753-789.

Heng Y, Liang Y, Zhang J, Li L, Zhang W, et al. Camptothecin Inhibits Neddylation to Activate the Protective Autophagy Through NF- κ B/AMPK/mTOR/ULK1 Axis in Human Esophageal Cancer Cells. Frontiers in Oncology. 2021, 11, 671180.

Pommier Y. Topoisomerase I inhibitors: camptothecins and beyond. Nat Rev Cancer. 2006, 6(10), 789-802.

Mabb AM, Simon JM, King IF, Lee HM, An LK, et al. Topoisomerase 1 Regulates Gene Expression in Neurons through Cleavage Complex-Dependent and -Independent Mechanisms. PLoS One. 2016, 11(5), e0156439.

Roskoski RJ. A historical overview of protein kinases and their targeted small molecule inhibitors. Pharmacological Research. 2015, 100, 1-23.

Gross S, Rahal R, Stransky N, Lengauer C, Hoeflich KP. Targeting cancer with kinase inhibitors. Journal of Clinical Investigation. 2015, 125(5), 1780-1789.

Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011, 144(5), 646-74.

Rip J, Van Der Ploeg EK, Hendriks RW, Corneth OBJ. The Role of Bruton’s Tyrosine Kinase in Immune Cell Signaling and Systemic Autoimmunity. Critical ReviewsTM in Immunology. 2018, 38(1), 17-62.

Rip J, de Bruijn MJW, Appelman MK, Pal Singh S, Hendriks RW, et al. Toll-Like Receptor Signaling Drives Btk-Mediated Autoimmune Disease. Frontiers in Immunology. 2019, 10, 95.

Di Liberto V, Mudò G, Belluardo N. Crosstalk between receptor tyrosine kinases (RTKs) and G protein-coupled receptors (GPCR) in the brain: Focus on heteroreceptor complexes and related functional neurotrophic effects. Neuropharmacology. 2019, 152, 67-77.

Liu J, Guiadeen D, Krikorian A, Gao X, Wang J, et al. Discovery of 8-Amino-imidazo[1,5-a]pyrazines as Reversible BTK Inhibitors for the Treatment of Rheumatoid Arthritis. ACS Medicinal Chemistry Letters. 2016, 7(2), 198-203.

Pal Singh S, Dammeijer F, Hendriks RW. Role of Bruton’s tyrosine kinase in B cells and malignancies. Molecular Cancer. 2018, 17, 57.

Cameron F, Sanford M. Ibrutinib: first global approval. Drugs. 2014, 74(2), 263-71.

Blackmon A, O’Brien S. An update on acalabrutinib to treat chronic lymphocytic leukemia. Drugs Today (Barc). 2021, 57(7), 417-431.

Syed YY. Zanubrutinib: First Approval. Drugs. 2020, 80(1), 91-97.

Schafer PH, Kivitz AJ, Ma J, Korish S, Sutherland D, et al. Spebrutinib (CC-292) Affects Markers of B Cell Activation, Chemotaxis, and Osteoclasts in Patients with Rheumatoid Arthritis: Results from a Mechanistic Study. Rheumatology and Therapy. 2020, 7(1), 101-119.

Gillooly KM, Pulicicchio C, Pattoli MA, Cheng L, Skala S, et al. Bruton’s tyrosine kinase inhibitor BMS-986142 in experimental models of rheumatoid arthritis enhances efficacy of agents representing clinical standard-of-care. PLoS One. 2017, 12(7), e0181782.

Narita Y, Nagane M, Mishima K, Terui Y, Arakawa Y, et al. Phase I/II study of tirabrutinib, a second-generation Bruton’s tyrosine kinase inhibitor, in relapsed/refractory primary central nervous system lymphoma. Neuro-Oncology. 2021, 23(1), 122-133.

Cohen S, Tuckwell K, Katsumoto TR, Zhao R, Galanter J, et al. Fenebrutinib versus Placebo or Adalimumab in Rheumatoid Arthritis: A Randomized, Double-Blind, Phase II Trial (ANDES Study). Arthritis & Rheumatology. 2020, 72(9), 1435-1446.

Smith PF, Krishnarajah J, Nunn PA, Hill RJ, Karr D, et al. A phase I trial of PRN1008, a novel reversible covalent inhibitor of Bruton’s tyrosine kinase, in healthy volunteers. British Journal of Clinical Pharmacology. 2017, 83(11), 2367-2376.

Rozkiewicz D, Hermanowicz JM, Kwiatkowska I, Krupa A, Pawlak D. Bruton’s Tyrosine Kinase Inhibitors (BTKIs): Review of Preclinical Studies and Evaluation of Clinical Trials. Molecules. 2023, 28(5), 2400.

Goldwasser F, Shimizu T, Jackman J, O'Connor PM, Kohn KW, et al. Correlations between S and G2 Arrest and the Cytotoxicity of Camptothecin in Human Colon Carcinoma Cells. Cancer Research. 1996, 56(19), 4430-7.

Zhang D, Harris HM, Chen J, Judy J, James G, et al. NRX-0492 degrades wild-type and C481 mutant BTK and demonstrates in vivo activity in CLL patient-derived xenografts. Blood. 2023,141(13), 1584-1596.

Qi J, Endres S, Yosifov DY, Tausch E, Dheenadayalan RP, et al. Acquired BTK mutations associated with resistance to noncovalent BTK inhibitors. Blood Advances. 2023, 7(19), 5698-5702.

Elamin G, Aljoundi A, Alahmdi MI, Abo-Dya NE, Soliman MES. Battling BTK mutants with noncovalent inhibitors that overcome Cys481 and Thr474 mutations in Waldenström macroglobulinemia therapy: structural mechanistic insights on the role of fenebrutinib. Journal of Molecular Modeling. 2022, 28(11), 355.

Pettersen EF, Goddard TD, Huang CC, Couch GS, Greenblatt DM, et al. UCSF Chimera-A visualization system for exploratory research and analysis. Journal of Computational Chemistry. 2004, 25(13), 1605-12.

Cherinka B, Andrews BH, Sánchez-Gallego J, Brownstein J, Argudo-Fernández M, et al. Marvin: A Tool Kit for Streamlined Access and Visualization of the SDSS-IV MaNGA Data Set. The Astronomical Journal. 2019, 158, 74.

Hanwell MD, Curtis DE, Lonie DC, Vandermeersch T, Zurek E, et al. Avogadro: an advanced semantic chemical editor, visualization, and analysis platform. Journal of Cheminformatics. 2012, 4, 17.

Dallakyan S, Olson AJ. Small-molecule library screening by docking with PyRx. Methods in Molecular Biology. 2015, 1263, 243-50.

Trott O, Olson AJ. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. Journal of Computational Chemistry. 2010, 31(2), 455-461.

Song LF, Lee TS, Zhu C, York DM, Merz KM. Using AMBER18 for Relative Free Energy Calculations. Journal of Chemical Information and Modeling. 2019, 59(7), 3128-3135.

Maier JA, Martinez C, Kasavajhala K, Wickstrom L, Hauser KE, et al. ff14SB: Improving the Accuracy of Protein Side Chain and Backbone Parameters from ff99SB. Journal of Chemical Theory and Computation. 2015, 11(8), 3696-3713.

Wang J, Wolf RM, Caldwell JW, Kollman PA, Case DA. Development and testing of a general amber force field. Journal of Computational Chemistry. 2004, 25(9), 1157-74.

Bayly CI, Cieplak P, Cornell WD, Kollman PA. A well-behaved electrostatic potential based method using charge restraints for deriving atomic charges: The RESP model. Journal of Physical Chemistry. 1993, 97(40), 10269-10280.

Wang J, Wang W, Kollman PA, Case DA. Automatic atom type and bond type perception in molecular mechanical calculations. Journal of Molecular Graphics and Modelling. 2006, 25(2), 247-260.

Larini L, Mannella R, Leporini D. Langevin stabilization of molecular-dynamics simulations of polymers by means of quasisymplectic algorithms. The Journal of Chemical Physics. 2007, 126, 104101.

Lin Y, Pan D, Li J, Zhang L, Shao X. Application of Berendsen barostat in dissipative particle dynamics for nonequilibrium dynamic simulation. The Journal of Chemical Physics. 2017, 146(12), 124108.

Gonnet P. P-SHAKE: a quadratically convergent SHAKE in O (n2). Journal of Computational Physics. 2007, 220(2), 740-750.

Roe DR, Cheatham III TE. PTRAJ and CPPTRAJ: software for processing and analysis of molecular synamics trajectory data. Journal of Chemical Theory and Computation. 2013, 9, 3084-309595.

Agnihotry S, Pathak RK, Singh DB, Tiwari A, Hussain I. Protein structure prediction. Bioinformatics: Methods and Applications. 2021, 177-88.

Mateev E, Valkova I, Angelov B, Georgieva M, Zlatkov A. VALIDATION THROUGH RE-DOCKING, CROSS-DOCKING AND LIGAND ENRICHMENT IN VARIOUS WELL-RESOLUTED MAO-B RECEPTORS. International Journal of Pharmaceutical Sciences and Research. 2022, 13(3), 1099-1107.

Slavov S, Beger RD. Identification of structural factors that affect binding to cannabinoid receptor type 1. Journal of Molecular Structure. 2022, 1249, 131589.

Rajapakse M, Feng L. Predicting Peptide Binders of Flexible Lengths with Genetic Annealing Algorithm. Advanced Materials Research. 2013, 805-806, 1856.

Weiss DR, Karpiak J, Huang XP, Sassano MF, Lyu J, et al. Selectivity Challenges in Docking Screens for GPCR Targets and Antitargets. Journal of Medicinal Chemistry. 2018, 61(15), 6830-6845.

Miller BR, Mcgee TD, Swails JM, Homeyer N, Gohlke H, et al. MMPBSA.py : An E ffi cient Program for End-State Free Energy Calculations. Journal of Chemical Theory and Computation. 2012, 8(9),3314-3321.

Genheden S, Ryde U. The MM/PBSA and MM/GBSA methods to estimate ligand-binding affinities. Expert Opinion on Drug Discovery. 2015, 10(5), 449-461.

Downloads

Published

2024-07-07

How to Cite

Issahaku, A. R., & Soliman, M. (2024). Camptothecin and its Analogues as Putative Bruton Tyrosine Kinase (BTK) Inhibitors in Cancer Therapy - Biophysical Simulations of Wild Type and C481S Mutation. Journal of Cancer Biomoleculars and Therapeutics, 1(1), 59–71. https://doi.org/10.62382/jcbt.v1i1.10

Issue

Section

Articles