Drug Res (Stuttg) 2024; 74(02): 81-88
DOI: 10.1055/a-2211-2218
Original Article

Structure-Based Drug Design for Targeting IRE1: An in Silico Approach for Treatment of Cancer

Alireza Poustforoosh
1   Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
,
Sanaz Faramarz
2   Applied Cellular and Molecular Research Center, Kerman University of Medical Sciences, Kerman, Iran
,
2   Applied Cellular and Molecular Research Center, Kerman University of Medical Sciences, Kerman, Iran
3   Department of Clinical Biochemistry, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
,
Mehdi Mahmoodi
2   Applied Cellular and Molecular Research Center, Kerman University of Medical Sciences, Kerman, Iran
,
Mahdiyeh Azadpour
4   Department of Chemical Engineering, Faculty of Engineering, Shahid Bahonar University of Kerman, Kerman, Iran
› Author Affiliations
Fundings Vice Chancellor for Research and Technology, Kerman University of Medical Sciences — http://dx.doi.org/10.13039/501100021077; 98001132 This study was granted by the Kerman University of Medical Sciences, Kerman, Iran by project No. 98001132.

Abstract

Background Endoplasmic Reticulum (ER) stress and Unfolded Protein Response (UPR) play a key role in cancer progression. The aggregation of incorrectly folded proteins in the ER generates ER stress, which in turn activates the UPR as an adaptive mechanism to fix ER proteostasis. Inositol-requiring enzyme 1 (IRE1) is the most evolutionary conserved ER stress sensor, which plays a pro-tumoral role in various cancers. Targeting its’ active sites is one of the most practical approaches for the treatment of cancers.

Objective In this study, we aimed to use the structure of 4μ8C as a template to produce newly designed compounds as IRE1 inhibitors.

Methods Various functional groups were added to the 4μ8C, and their binding affinity to the target sites was assessed by conducting a covalent molecular docking study. The potential of the designed compound for further in vitro and in vivo studies was evaluated using ADMET analysis.

Results Based on the obtained results, the addition of hydroxyl groups to 4μ8C enhanced the binding affinity of the designed compound to the target efficiently. Compound 17, which was constructed by the addition of one hydroxyl group to the structure of 4μ8C, can construct a strong covalent bond with Lys907. The outcomes of ADMET analysis indicated that compound 17 could be considered a drug-like molecule.

Conclusion Our results revealed that designed compound 17 could inhibit IRE1 activity. Therefore, this designed compound is a remarkable inhibitor of IRE1 and introduces a promising therapeutic strategy for cancer treatment.



Publication History

Received: 18 September 2023

Accepted: 13 November 2023

Article published online:
22 December 2023

© 2023. Thieme. All rights reserved.

Georg Thieme Verlag
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Raymundo DP, Doultsinos D, Guillory X, Carlesso A, Eriksson LA, Chevet E. Pharmacological targeting of IRE1 in cancer. Trends in cancer 2020; 6: 1018-1030
  • 2 Paydar P, Asadikaram G, Nejad HZ. et al Epigenetic modulation of BRCA-1 and MGMT genes, and histones H4 and H3 are associated with breast tumors. Journal of Cellular Biochemistry 2019; 120: 13726-13736
  • 3 Wang C-Q, Tang C-H, Tzeng H-E. et al Impacts of RETN genetic polymorphism on breast cancer development. Journal of Cancer 2020; 11: 2769
  • 4 Shai A, Galouk E, Miari R, Tareef H, Sammar M, Zeidan M, Rayan A, Falah M. Inhibiting mutant KRAS G12D gene expression using novel peptide nucleic acid‑based antisense: A potential new drug candidate for pancreatic cancer. Oncology Letters 2022; 23: 1-11
  • 5 Zhang L, Liu W, Wang Q, Li Q, Wang H, Wang J, Teng T, Chen M, Ji A, Li Y. New drug candidate targeting the 4A1 orphan nuclear receptor for medullary thyroid cancer therapy. Molecules 2018; 23: 565
  • 6 Falah M, Rayan M, Rayan A. A novel paclitaxel conjugate with higher efficiency and lower toxicity: a new drug candidate for cancer treatment. International Journal of Molecular Sciences 2019; 20: 4965
  • 7 Urra H, Dufey E, Avril T, Chevet E, Hetz C. Endoplasmic reticulum stress and the hallmarks of cancer. Trends in cancer 2016; 2: 252-262
  • 8 Roy A, Kumar A. ER stress and unfolded protein response in cancer cachexia. Cancers 2019; 11: 1929
  • 9 Wang Y, Wang K, Jin Y, Sheng X. Endoplasmic reticulum proteostasis control and gastric cancer. Cancer Letters 2019; 449: 263-271
  • 10 Limia CM, Sauzay C, Urra H, Hetz C, Chevet E, Avril T. Emerging roles of the endoplasmic reticulum associated unfolded protein response in cancer cell migration and invasion. Cancers 2019; 11: 631
  • 11 Lee J-H, Jeong J, Jeong EM, Cho S-Y, Kang JW, Lim J, Heo J, Kang H, Kim I-G, Shin D-M. Endoplasmic reticulum stress activates transglutaminase 2 leading to protein aggregation. International Journal of Molecular Medicine 2014; 33: 849-855
  • 12 Naidoo N. ER and aging—protein folding and the ER stress response. Ageing research reviews 2009; 8: 150-159
  • 13 Urra H, Dufey E, Lisbona F, Rojas-Rivera D, Hetz C. When ER stress reaches a dead end. Biochimica et Biophysica Acta (BBA)-Molecular Cell Research 2013; 1833: 3507-3517
  • 14 Sheng X, Nenseth HZ, Qu S, Kuzu OF, Frahnow T, Simon L, Greene S, Zeng Q, Fazli L, Rennie PS. IRE1α-XBP1s pathway promotes prostate cancer by activating c-MYC signaling. Nature communications 2019; 10: 323
  • 15 Beaupere C, Dinatto L, Wasko BM, Chen RB, VanValkenburg L, Kiflezghi MG, Lee MB, Promislow DE, Dang W, Kaeberlein M. Genetic screen identifies adaptive aneuploidy as a key mediator of ER stress resistance in yeast. Proceedings of the National Academy of Sciences 2018; 115: 9586-9591
  • 16 Leary M, Heerboth S, Lapinska K, Sarkar S. Sensitization of drug resistant cancer cells: a matter of combination therapy. Cancers 2018; 10: 483
  • 17 Shi W, Chen Z, Li L, Liu H, Zhang R, Cheng Q, Xu D, Wu L. Unravel the molecular mechanism of XBP1 in regulating the biology of cancer cells. Journal of Cancer 2019; 10: 2035
  • 18 Amawi H, Hussein NA, Ashby CR, Alnafisah R, Sanglard LM, Manivannan E, Karthikeyan C, Trivedi P, Eisenmann KM, Robey RW. Bax/tubulin/epithelial-mesenchymal pathways determine the efficacy of silybin analog HM015k in colorectal cancer cell growth and metastasis. Frontiers in pharmacology 2018; 9: 520
  • 19 Riaz TA, Junjappa RP, Handigund M, Ferdous J, Kim H-R, Chae H-J. Role of endoplasmic reticulum stress sensor IRE1α in cellular physiology, calcium, ROS signaling, and metaflammation. Cells 2020; 9: 1160
  • 20 Bouchecareilh M, Higa A, Fribourg S, Moenner M, Chevet E. Peptides derived from the bifunctional kinase/RNase enzyme IRE1α modulate IRE1α activity and protect cells from endoplasmic reticulum stress. The FASEB Journal 2011; 25: 3115-3129
  • 21 Siwecka N, Rozpędek-Kamińska W, Wawrzynkiewicz A, Pytel D, Diehl JA, Majsterek I. The structure, activation and signaling of IRE1 and its role in determining cell fate. Biomedicines 2021; 9: 156
  • 22 Carlesso A, Chintha C, Gorman AM, Samali A, Eriksson LA. Binding analysis of the Inositol-requiring enzyme 1 kinase domain. Acs Omega 2018; 3: 13313-13322
  • 23 Harrington PE, Biswas K, Malwitz D, Tasker AS, Mohr C, Andrews KL, Dellamaggiore K, Kendall R, Beckmann H, Jaeckel P. Unfolded protein response in cancer: IRE1α inhibition by selective kinase ligands does not impair tumor cell viability. ACS medicinal chemistry letters 2015; 6: 68-72
  • 24 Feldman HC, Tong M, Wang L, Meza-Acevedo R, Gobillot TA, Lebedev I, Gliedt MJ, Hari SB, Mitra AK, Backes BJ. Structural and functional analysis of the allosteric inhibition of IRE1α with ATP-competitive ligands. ACS chemical biology 2016; 11: 2195-2205
  • 25 Mehrabani M, Raeiszadeh M, Najafipour H, Esmaeli Tarzi M, Amirkhosravi A, Poustforoosh A, Mohammadi MA, Naghdi S, Mehrabani M. Evaluation of the Cytotoxicity, Antibacterial, Antioxidant, and Anti-inflammatory Effects of Different Extracts of Punica granatum var. pleniflora. Journal of Kerman University of Medical Sciences 2020; 27: 414-425
  • 26 Sanches M, Duffy NM, Talukdar M, Thevakumaran N, Chiovitti D, Canny MD, Lee K, Kurinov I, Uehling D, Al-Awar R. Structure and mechanism of action of the hydroxy–aryl–aldehyde class of IRE1 endoribonuclease inhibitors. Nature communications 2014; 5: 4202
  • 27 Tang C-HA, Ranatunga S, Kriss CL, Cubitt CL, Tao J, Pinilla-Ibarz JA, Del Valle JR, Hu C-CA. Inhibition of ER stress–associated IRE-1/XBP-1 pathway reduces leukemic cell survival. The Journal of clinical investigation 2014; 124: 2585-2598
  • 28 Cross BC, Bond PJ, Sadowski PG, Jha BK, Zak J, Goodman JM, Silverman RH, Neubert TA, Baxendale IR, Ron D. The molecular basis for selective inhibition of unconventional mRNA splicing by an IRE1-binding small molecule. Proceedings of the National Academy of Sciences 2012; 109: E869-E878
  • 29 Mihăşan M. What in silico molecular docking can do for the ‘bench-working biologists’. Journal of biosciences 2012; 37: 1089-1095
  • 30 Meng X-Y, Zhang H-X, Mezei M, Cui M. Molecular docking: a powerful approach for structure-based drug discovery. Current computer-aided drug design 2011; 7: 146-157
  • 31 Fan J, Fu A, Zhang L. Progress in molecular docking. Quantitative Biology 2019; 7: 83-89
  • 32 Poustforoosh A, Faramarz S, Nematollahi MH, Hashemipour H, Negahdaripour M, Pardakhty A. In silico SELEX screening and statistical analysis of newly designed 5mer peptide-aptamers as Bcl-xl inhibitors using the Taguchi method. Computers in Biology and Medicine 2022; 146: 105632
  • 33 Poustforoosh A, Hashemipour H, Tüzün B, Pardakhty A, Mehrabani M, Nematollahi MH. Evaluation of potential anti-RNA-dependent RNA polymerase (RdRP) drugs against the newly emerged model of COVID-19 RdRP using computational methods. Biophysical chemistry 2021; 272: 106564
  • 34 Poustforoosh A, Faramarz S, Nematollahi MH, Hashemipour H, Tüzün B, Pardakhty A, Mehrabani M. 3D-QSAR, molecular docking, molecular dynamics, and ADME/T analysis of marketed and newly designed flavonoids as inhibitors of Bcl-2 family proteins for targeting U-87 glioblastoma. Journal of Cellular Biochemistry 2022; 123: 390-405
  • 35 Rants'o TA, Van der Westhuizen CJ, van Zyl RL. Optimization of covalent docking for organophosphates interaction with Anopheles acetylcholinesterase. Journal of Molecular Graphics and Modelling 2022; 110: 108054
  • 36 Poustforoosh A, Hashemipour H, Tüzün B, Azadpour M, Faramarz S, Pardakhty A, Mehrabani M, Nematollahi MH. The impact of D614G mutation of SARS-COV-2 on the efficacy of anti-viral drugs: A comparative molecular docking and molecular dynamics study. Current microbiology 2022; 79: 241
  • 37 Taslimi P, Erden Y, Mamedov S, Zeynalova L, Ladokhina N, Tas R, Tuzun B, Sujayev A, Sadeghian N, Alwasel SH. The biological activities, molecular docking studies, and anticancer effects of 1-arylsuphonylpyrazole derivatives. Journal of biomolecular structure and dynamics 2021; 39: 3336-3346
  • 38 Pavlović N, Heindryckx F. Targeting ER stress in the hepatic tumor microenvironment. The FEBS Journal 2022; 289: 7163-7176
  • 39 Ricci D, Tutton S, Marrocco I, Ying M, Blumenthal D, Eletto D, Vargas J, Boyle S, Fazelinia H, Qian L. An interdomain helix in IRE1α mediates the conformational change required for the sensor's activation. Journal of Biological Chemistry 2021; 296: 100781
  • 40 Mahdizadeh SJ, Carlesso A, Eriksson LA. Deciphering the selectivity of inhibitor MKC9989 towards residue K907 in IRE1α; a multiscale in silico approach. RSC advances 2020; 10: 19720-19729
  • 41 Carlesso A, Eriksson LA. Selective inhibition of IRE1 signalling mediated by MKC9989: new insights from molecular docking and molecular dynamics simulations. ChemistrySelect 2019; 4: 3199-3203
  • 42 Carlesso A, Chintha C, Gorman AM, Samali A, Eriksson LA. Merits and pitfalls of conventional and covalent docking in identifying new hydroxyl aryl aldehyde like compounds as human IRE1 inhibitors. Scientific reports 2019; 9: 3407
  • 43 Dong J, Wang N-N, Yao Z-J, Zhang L, Cheng Y, Ouyang D, Lu A-P, Cao D-S. ADMETlab: a platform for systematic ADMET evaluation based on a comprehensively collected ADMET database. Journal of cheminformatics 2018; 10: 1-11
  • 44 Wang Y, Xing J, Xu Y, Zhou N, Peng J, Xiong Z, Liu X, Luo X, Luo C, Chen K. In silico ADME/T modelling for rational drug design. Quarterly reviews of biophysics 2015; 48: 488-515
  • 45 Wishart DS. Improving early drug discovery through ADME modelling: an overview. Drugs in R & D 2007; 8: 349-362
  • 46 Lipinski CA. Lead-and drug-like compounds: the rule-of-five revolution. Drug discovery today: Technologies 2004; 1: 337-341
  • 47 Jorgensen WL, Duffy EM. Prediction of drug solubility from structure. Advanced drug delivery reviews 2002; 54: 355-366
  • 48 Poustforoosh A, Nematollahi MH, Hashemipour H, Pardakhty A. Recent advances in Bio-conjugated nanocarriers for crossing the Blood-Brain Barrier in (pre-) clinical studies with an emphasis on vesicles. Journal of Controlled Release 2022; 343: 777-797
  • 49 Asadikaram G, Poustforoosh A, Pardakhty A, Torkzadeh-Mahani M, Nematollahi MH. Niosomal virosome derived by vesicular stomatitis virus glycoprotein as a new gene carrier. Biochemical and Biophysical Research Communications 2021; 534: 980-987