1
|
C M, Pasha TY, Rahamathulla M, H P G, B L K, K M G, K N P, Hussain SM, Ahmed MM, Shivanandappa TB, Pasha I. Epidermal growth factor receptors unveiled: a comprehensive survey on mutations, clinical insights of global inhibitors, and emergence of heterocyclic derivatives as EGFR inhibitors. J Drug Target 2025; 33:933-951. [PMID: 39756062 DOI: 10.1080/1061186x.2024.2449495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/09/2024] [Accepted: 12/21/2024] [Indexed: 01/07/2025]
Abstract
Mutations that overexpress the epidermal growth factor receptor (EGFR) are linked to cancers like breast (15-20%), head and neck (10-15%), colorectal (5-8%), and non-small cell lung cancer (10-50%), especially in East Asian populations. EGFR activation stimulates 'RAS/RAF/MEK/ERK, PI3K/Akt, and MAPK' pathways, which enhance cell division, survival, angiogenesis, and tumour growth while inhibiting apoptosis and metastasis. Secondary mutations (e.g. 'T790M', 'C797S'), off-target effects, and resistance due to alternate pathway activation reduce the efficacy of currently available EGFR inhibitors. To address these issues, 'novel heterocyclic inhibitors with structural versatility were developed to improve selectivity and binding affinity for mutant EGFR forms'. These new EGFR reduce side effects, enhance pharmacokinetics, and enhance therapeutic efficacy at low concentrations. This review focuses on 'EGFR mutations in various cancers' detailing the biochemical effects, clinical profiles, and binding interactions of globally approved EGFR inhibitors. Furthermore, it focuses into recent progress in nano-formulations and the development of heterocyclic derivatives that can successfully 'target mutant EGFRs' through varied synthesis methods. These inhibitors have the potential to have better binding affinities, selectivity's, and less side-effect. Further research required to refine the structures and develop nanoformulations of EGFR-targeted therapeutics in order to improve therapeutic efficiency and, provide more effective cancer treatments.
Collapse
Affiliation(s)
- Manojmouli C
- Department of Pharmaceutical Chemistry, Sri Adichunchanagiri College of Pharmacy, Adichunchanagiri University, B. G. Nagara, Karnataka, India
| | - T Y Pasha
- Department of Pharmaceutical Chemistry, Sri Adichunchanagiri College of Pharmacy, Adichunchanagiri University, B. G. Nagara, Karnataka, India
| | - Mohamed Rahamathulla
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Al Faraa 62223, Abha, Saudi Arabia
| | - Gagana H P
- Department of Pharmaceutical Chemistry, Sri Adichunchanagiri College of Pharmacy, Adichunchanagiri University, B. G. Nagara, Karnataka, India
| | - Kavya B L
- Department of Pharmaceutical Chemistry, Sri Adichunchanagiri College of Pharmacy, Adichunchanagiri University, B. G. Nagara, Karnataka, India
| | - Gagana K M
- Department of Pharmaceutical Chemistry, Sri Adichunchanagiri College of Pharmacy, Adichunchanagiri University, B. G. Nagara, Karnataka, India
| | - Purushotham K N
- Department of Pharmaceutical Chemistry, Sri Adichunchanagiri College of Pharmacy, Adichunchanagiri University, B. G. Nagara, Karnataka, India
| | - Shalam M Hussain
- Department of Clinical Pharmacy, College of Nursing and Health Science. Al-Rayyan Medical College, Madinah, Saudi Arabia
| | - Mohammed Muqtader Ahmed
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | | | - Ismail Pasha
- Department of Pharmacology, Orotta College of Medicine and Health Sciences, Asmara University, Asmara, State of Eritrea
| |
Collapse
|
2
|
Saleh NS, El-Sayed NNE, Saleh OA, Allam HA, Mohamed NM, Abbas SES, Said MF. 6,7-Dimethoxy-2-methyl-4-substituted quinazolines: Design, synthesis, EGFR inhibitory activity, in vitro cytotoxicity, and in silico studies. Eur J Med Chem 2025; 290:117502. [PMID: 40120497 DOI: 10.1016/j.ejmech.2025.117502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/25/2025] [Accepted: 03/10/2025] [Indexed: 03/25/2025]
Abstract
Six series of 2,4,6,7-tetrasubstituted quinazolines 4a-c, 6a-c, 8a-c, 10a-d, 13a-d along with quinazoline-tetrahydropyrimidine hybrids 15a-c were designed and synthesized based on keeping the essential key binding pattern of some EGFR inhibitors to appraise their EGFR inhibition and anticancer activity. Twelve compounds out of twenty displayed a significant EGFR inhibition in a subnanomolar level (IC50 = 0.143-0.946 nM) compared to afatinib (IC50 = 0.102 nM). The most potent derivatives 4a, 6c, 8b, 13a and 15b (IC50 = 0.143-0.313 nM) were further screened for their anticancer activity against lung (A549) and colon (HCT116) cancer cell lines, in addition to, normal fibroblast cells (WI-38). It was found that, compounds 6c and 13a show a nearly equipotent to superior cytotoxicity towards (A549) (IC50 = 0.020 and 0.006 μM; respectively) and (IC50 = 0.020 and 0.038 μM; respectively) against HCT116 in comparison to afatinib (IC50 = 0.025 and 0.030 μM; respectively). Also, compounds 6c and 13a caused a cell cycle arrest at S phase and induced apoptosis in A549 and HCT116; respectively. Moreover, in silico studies clarified the binding pattern of the potent compounds in EGFR enzyme active site and confirmed their ability to gratify the structural features meted for binding and rationalized their selectivity. Furthermore, the most active candidates possess promising predicted pharmacokinetic properties.
Collapse
Affiliation(s)
- Nermin S Saleh
- Egyptian Drug Authority (EDA), 51 Wezaret El-Zeraa St., Giza, 35521, Egypt
| | - Nahed N E El-Sayed
- Egyptian Drug Authority (EDA), 51 Wezaret El-Zeraa St., Giza, 35521, Egypt
| | - Ola A Saleh
- Medicinal and Pharmaceutical Chemistry Department, Pharmaceutical Industries Research Institute, National Research Centre, Egypt
| | - Heba Abdelrasheed Allam
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, P.O. Box 11562, Kasr El-Aini Street, Cairo, Egypt
| | - Nada M Mohamed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Modern University for Technology and Information MTI, Cairo, Egypt
| | - Safinaz E-S Abbas
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, P.O. Box 11562, Kasr El-Aini Street, Cairo, Egypt
| | - Mona F Said
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, P.O. Box 11562, Kasr El-Aini Street, Cairo, Egypt.
| |
Collapse
|
3
|
Das A, Biggs MA, Hunt HL, Mahabadi V, Goncalves BG, Phan CAN, Banerjee IA. Design and investigation of novel iridoid-based peptide conjugates for targeting EGFR and its mutants L858R and T790M/L858R/C797S: an in silico study. Mol Divers 2025; 29:2517-2541. [PMID: 39424745 DOI: 10.1007/s11030-024-11007-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 09/29/2024] [Indexed: 10/21/2024]
Abstract
In this work, we designed novel peptide conjugates with plant-based iridoid and lichen-derived depside derivatives to target the wild-type EGFR (WT) and its mutants, L858R and T790M/L858R/C797S triple mutant. These mutations are often expressed in multiple cancers, particularly lung cancer. Specifically, the iridoids included 7-deoxyloganetic acid (7-DGA) and loganic acid (LG), while the depside derivative was sekikaic acid (SK). These compounds are known for their innate anticancer properties and were conjugated with two separate peptide sequences KLPGWSG (K) and YSIPKSS (Y). These sequences have been shown to target EGFR in previous phage display library screening, although the mechanism is unknown. Thus, we created the di-conjugates for dual targeting and investigated their interactions of the di-conjugates and that of the neat peptides with the kinase domain of EGFR (WT) and the two mutants using molecular docking, molecular dynamics (MD) simulations, and MM-GBSA analysis. Docking studies revealed that the (7-DGA)2-K showed the highest binding affinity at - 9.3 kcal/mol with the L858R mutant, while (LG)2-Y displayed the highest binding affinity at - 9.0 kcal/mol for the triple mutant receptor. Our results indicated that several of the conjugates interacted with crucial residues of the kinase domain, including ASP855 and THR854 (activation loop), MET793 and PRO794 (hinge region), ARG841 (catalytic loop), and LYS728 and LEU718 of the glycine-rich P-loop. Interestingly, strong hydrophobic interactions were also observed with the C-terminal tail residues, such as PHE997 and ALA1000 as well as with ARG999 for the YSIPKSS peptide and most of the conjugates. The hydroxyl group of the cyclopentane ring and the oxygen of the pyran ring of the (7-DGA)2-peptide conjugates contributed to binding particularly in the hinge region, while the peptide components formed an extended structure that bound well into the C-lobe. The (SK)2-Y di-conjugate and KLPGWSG peptide formed hydrogen bonds with the SER797 residue of the triple mutant. Overall, our results show that the (7-DGA)2-K, di-conjugate, the (7-DGA)2-Y di-conjugate, and the neat YSIPKSS demonstrated strong and stable binding with the L858R mutant and the highly resistant triple mutant EGFR, respectively. The novel designed conjugates demonstrate potential for further optimization for laboratory studies aimed at developing new therapeutics for targeting specific EGFR mutant expressing cells.
Collapse
Affiliation(s)
- Amrita Das
- Department of Chemistry and Biochemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Mary A Biggs
- Department of Chemistry and Biochemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Hannah L Hunt
- Department of Chemistry and Biochemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Vida Mahabadi
- Department of Chemistry and Biochemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Beatriz G Goncalves
- Department of Chemistry and Biochemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Chau Anh N Phan
- Department of Chemistry and Biochemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Ipsita A Banerjee
- Department of Chemistry and Biochemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA.
| |
Collapse
|
4
|
Kolcuoglu Y, Bekircan O, Ustalar N, Türe A, Akdemir A, Sanlier SH. Synthesis, In Vitro Evaluation, and Molecular Docking Studies of Novel 3,5-Diphenyl-1H-1,2,4-Triazole Derivatives as Potential hEGFR Inhibitors. Arch Pharm (Weinheim) 2025; 358:e70007. [PMID: 40359331 DOI: 10.1002/ardp.70007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 03/10/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025]
Abstract
EGFR, an important target in cancer chemotherapy, is an important component of the signaling system that regulates important cellular processes such as growth, differentiation, metabolism, and apoptosis in response to both internal and external stimuli. Based on this approach, comprehensive modeling studies targeting the EGFR protein were performed, and synthesized molecules were proposed. For this purpose, the synthesis of new 3,5-diphenyl-1H-1,2,4-triazole derivatives containing semicarbazide, thiosemicarbazide, 1,2,4-triazole-3-thione, and 1,2,4-triazole-3-one units was carried out. Among these compounds, 6a-6i presented in the present study exhibited EGFR inhibition in the nanomolar range. In addition, molecules 5e and 6e showed significant IC50 values. Compound 6e showed the closest IC50 value to gefitinib, a well-known EGFR inhibitor, with its noncompetitive inhibition mode. The Ki value of compound 6e was determined as 0.174 µM.
Collapse
Affiliation(s)
- Yakup Kolcuoglu
- Department of Chemistry, Faculty of Science, Karadeniz Technical University, Trabzon, Türkiye
| | - Olcay Bekircan
- Department of Chemistry, Faculty of Science, Karadeniz Technical University, Trabzon, Türkiye
| | - Narin Ustalar
- Department of Chemistry, Faculty of Science, Karadeniz Technical University, Trabzon, Türkiye
| | - Aslı Türe
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Marmara University, Istanbul, Türkiye
| | - Atilla Akdemir
- Faculty of Pharmacy, Department of Pharmacology, Istinye University, Istanbul, Türkiye
| | | |
Collapse
|
5
|
Paul JK, Azmal M, Haque Shohan MN, Mrinmoy M, Newaz Been Haque ANMS, Talukder OF, Ghosh A. Identification of natural phytochemicals as AKT2 inhibitors using molecular docking and dynamics simulations as potential cancer therapeutics. Heliyon 2025; 11:e41897. [PMID: 39897896 PMCID: PMC11783009 DOI: 10.1016/j.heliyon.2025.e41897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 01/09/2025] [Accepted: 01/09/2025] [Indexed: 02/04/2025] Open
Abstract
The PI3K/AKT/mTOR pathway is central in regulating key cellular processes such as proliferation, survival, metabolism, and angiogenesis. Dysregulation of this pathway, particularly in the AKT2 isoform, is commonly observed in cancers such as breast, ovarian, and pancreatic cancers, leading to enhanced tumor progression, metastasis, and therapeutic resistance. Therefore, targeting AKT2 for inhibition is a promising strategy for cancer therapy. This study utilized molecular docking and dynamics simulations to identify natural phytochemicals that inhibit AKT2. Molecular docking results revealed that millettone (CID 442810) exhibited the highest binding affinity to AKT2, with a docking score of -9.5 kcal/mol, followed by uzarigenin (CID 92760), dihydrobiochanin A (CID 439784), and abyssinone I (CID 442152) with docking scores of -9.0 kcal/mol, -8.9 kcal/mol, and -8.7 kcal/mol respectively, outperforming the control inhibitor, ipatasertib (CID 24788740) docking score of -7.56 kcal/mol. Molecular dynamics simulations indicated that millettone, uzarigenin, and dihydrobiochanin A demonstrated strong binding affinities and stable interactions with AKT2, suggesting their potential as therapeutic agents for cancers that involve AKT2 hyperactivation. Notably, uzarigenin's superior stability, as evidenced by its lower root mean square deviation (RMSD), which measures structural stability, and solvent-accessible surface area (SASA), which indicates molecular compactness, highlights its promise as a potent inhibitor of AKT2. Future in vitro and in vivo studies will be crucial to confirm the efficacy of these inhibitors in reducing tumor progression and their potential applications. Given that AKT2 also plays a role in neuronal survival and plasticity, these compounds may have potential applications in neurodegenerative diseases such as Alzheimer's, warranting further investigation into their dual therapeutic relevance.
Collapse
Affiliation(s)
- Jibon Kumar Paul
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Mahir Azmal
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Md Naimul Haque Shohan
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Mohua Mrinmoy
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - ANM Shah Newaz Been Haque
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Omar Faruk Talukder
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Ajit Ghosh
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| |
Collapse
|
6
|
Hunt HL, Goncalves BG, Biggs MA, Rico MI, Murray ME, Lebedenko CG, Banerjee IA. Design and investigation of interactions of novel peptide conjugates of purine and pyrimidine derivatives with EGFR and its mutant T790M/L858R: an in silico and laboratory study. Mol Divers 2024; 28:3683-3711. [PMID: 38240950 DOI: 10.1007/s11030-023-10772-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 11/17/2023] [Indexed: 12/21/2024]
Abstract
Peptide-based therapeutics have been gaining attention due to their ability to actively target tumor cells. Additionally, several varieties of nucleotide derivatives have been developed to reduce cell proliferation and induce apoptosis of tumor cells. In this work, we have developed novel peptide conjugates with newly designed purine analogs and pyrimidine derivatives and explored the binding interactions with the kinase domain of wild-type EGFR and its mutant EGFR [L858R/ T790M] which are known to be over-expressed in tumor cells. The peptides explored included WNWKV (derived from sea cucumber) and LARFFS, which in previous work was predicted to bind to Domain I of EGFR. Computational studies conducted to explore binding interactions include molecular docking studies, molecular dynamics simulations and MMGBSA to investigate the binding abilities and stability of the complexes. The results indicate that conjugation enhanced binding capabilities, particularly for the WNWKV conjugates. MMGBSA analysis revealed nearly twofold higher binding toward the T790M/L858R double mutant receptor. Several conjugates were shown to have strong and stable binding with both wild-type and mutant EGFR. As a proof of concept, we synthesized pyrimidine conjugates with both peptides and determined the KD values using SPR analysis. The results corroborated with the computational analyses. Additionally, cell viability and apoptosis studies with lung cancer cells expressing the wild-type and double mutant proteins revealed that the WNWKV conjugate showed greater potency than the LARFFS conjugate, while LARFFS peptide alone showed poor binding to the kinase domain. Thus, we have designed peptide conjugates that show potential for further laboratory studies for developing therapeutics for targeting the EGFR receptor and its mutant T790M/L858R.
Collapse
Affiliation(s)
- Hannah L Hunt
- Department of Chemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Beatriz G Goncalves
- Department of Chemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Mary A Biggs
- Department of Chemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Mia I Rico
- Department of Chemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Molly E Murray
- Department of Chemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Charlotta G Lebedenko
- Department of Chemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Ipsita A Banerjee
- Department of Chemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA.
| |
Collapse
|
7
|
Azmy L, Ibraheem IBM, Alsalamah SA, Alghonaim MI, Zayed A, Abd El-Aleam RH, Mohamad SA, Abdelmohsen UR, Elsayed KNM. Evaluation of Cytotoxicity and Metabolic Profiling of Synechocystis sp. Extract Encapsulated in Nano-Liposomes and Nano-Niosomes Using LC-MS, Complemented by Molecular Docking Studies. BIOLOGY 2024; 13:581. [PMID: 39194519 DOI: 10.3390/biology13080581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/10/2024] [Accepted: 07/18/2024] [Indexed: 08/29/2024]
Abstract
Liposomes and niosomes can be considered excellent drug delivery systems due to their ability to load all compounds, whether hydrophobic or hydrophilic. In addition, they can reduce the toxicity of the loaded drug without reducing its effectiveness. Synechocystis sp. is a unicellular, freshwater cyanobacteria strain that contains many bioactive compounds that qualify its use in industrial, pharmaceutical, and many other fields. This study investigated the potential of nano-liposomes (L) and nano-niosomes (N) for delivering Synechocystis sp. extract against cancer cell lines. Four different types of nanoparticles were prepared using a dry powder formulation and ethanol extract of Synechocystis sp. in both nanovesicles (N1 and N2, respectively) and liposomes (L1 and L2, respectively). Analysis of the formed vesicles using zeta analysis, SEM morphological analysis, and visual examination confirmed their stability and efficiency. L1 and L2 in this investigation had effective diameters of 419 and 847 nm, respectively, with PDI values of 0.24 and 0.27. Furthermore, the zeta potentials were found to range from -31.6 mV to -43.7 mV. Regarding N1 and N2, their effective diameters were 541 nm and 1051 nm, respectively, with PDI values of 0.31 and 0.35, and zeta potentials reported from -31.6 mV to -22.2 mV, respectively. Metabolic profiling tentatively identified 22 metabolites (1-22) from the ethanolic extract. Its effect against representative human cancers was studied in vitro, specifically against colon (Caco2), ovarian (OVCAR4), and breast (MCF7) cancer cell lines. The results showed the potential activities of the prepared N1, N2, L1, and L2 against the three cell lines, where L1 had cytotoxicity IC50 values of 19.56, 33.52, and 9.24 µg/mL compared to 26.27, 56.23, and 19.61 µg/mL for L2 against Caco2, OVCAR4, and MCF7, respectively. On the other hand, N1 exhibited IC50 values of 9.09, 11.42, and 2.38 µg/mL, while N2 showed values of 15.57, 18.17, and 35.31 µg/mL against Caco2, OVCAR4, and MCF7, respectively. Meanwhile, the formulations showed little effect on normal cell lines (FHC, OCE1, and MCF10a). All of the compounds were evaluated in silico against the epidermal growth factor receptor tyrosine kinase (EGFR). The molecular docking results showed that compound 21 (1-hexadecanoyl-2-(9Z-hexadecenoyl)-3-(6'-sulfo-alpha-D-quinovosyl)-sn-glycerol), followed by compounds 6 (Sulfoquinovosyl monoacylgycerol), 7 (3-Hydroxymyristic acid), 8 (Glycolipid PF2), 12 (Palmitoleic acid), and 19 (Glyceryl monostearate), showed the highest binding affinities. These compounds formed good hydrogen bond interactions with the key amino acid Lys721 as the co-crystallized ligand. These results suggest that nano-liposomes and nano-niosomes loaded with Synechocystis sp. extract hold promise for future cancer treatment development. Further research should focus on clinical trials, stability assessments, and pharmacological profiles to translate this approach into effective anticancer drugs.
Collapse
Affiliation(s)
- Lamya Azmy
- Botany and Microbiology Department, Faculty of Science, Beni-Suef University, Beni-Suef 62511, Egypt
| | - Ibraheem B M Ibraheem
- Botany and Microbiology Department, Faculty of Science, Beni-Suef University, Beni-Suef 62511, Egypt
| | - Sulaiman A Alsalamah
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh 11623, Saudi Arabia
| | - Mohammed Ibrahim Alghonaim
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh 11623, Saudi Arabia
| | - Ahmed Zayed
- Pharmacognosy Department, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Rehab H Abd El-Aleam
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Modern University for Technology and Information MTI, Cairo 11571, Egypt
| | - Soad A Mohamad
- Clinical Pharmacy Department, Faculty of Pharmacy, Deraya University, New Minia 61111, Egypt
| | - Usama Ramadan Abdelmohsen
- Deraya Center for Scientific Research, Deraya University, New Minia 61111, Egypt
- Pharmacognosy Department, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Khaled N M Elsayed
- Botany and Microbiology Department, Faculty of Science, Beni-Suef University, Beni-Suef 62511, Egypt
| |
Collapse
|
8
|
Giugliano G, Gajo M, Marforio TD, Zerbetto F, Mattioli EJ, Calvaresi M. Identification of Potential Drug Targets of Calix[4]arene by Reverse Docking. Chemistry 2024; 30:e202400871. [PMID: 38777795 DOI: 10.1002/chem.202400871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/20/2024] [Accepted: 05/22/2024] [Indexed: 05/25/2024]
Abstract
Calixarenes are displaying great potential for the development of new drug delivery systems, diagnostic imaging, biosensing devices and inhibitors of biological processes. In particular, calixarene derivatives are able to interact with many different enzymes and function as inhibitors. By screening of the potential drug target database (PDTD) with a reverse docking procedure, we identify and discuss a selection of 100 proteins that interact strongly with calix[4]arene. We also discover that leucine (23.5 %), isoleucine (11.3 %), phenylalanines (11.3 %) and valine (9.5 %) are the most frequent binding residues followed by hydrophobic cysteines and methionines and aromatic histidines, tyrosines and tryptophanes. Top binders are peroxisome proliferator-activated receptors that already are targeted by commercial drugs, demonstrating the practical interest in calix[4]arene. Nuclear receptors, potassium channel, several carrier proteins, a variety of cancer-related proteins and viral proteins are prominent in the list. It is concluded that calix[4]arene, which is characterized by facile access, well-defined conformational characteristics, and ease of functionalization at both the lower and higher rims, could be a potential lead compound for the development of enzyme inhibitors and theranostic platforms.
Collapse
Affiliation(s)
- Giulia Giugliano
- Dipartimento di Chimica "Giacomo Ciamician", Alma Mater Studiorum - Università di Bologna, Via Francesco Selmi 2, 40126, Bologna, Italy E-Mail
| | - Margherita Gajo
- Dipartimento di Chimica "Giacomo Ciamician", Alma Mater Studiorum - Università di Bologna, Via Francesco Selmi 2, 40126, Bologna, Italy E-Mail
| | - Tainah Dorina Marforio
- Dipartimento di Chimica "Giacomo Ciamician", Alma Mater Studiorum - Università di Bologna, Via Francesco Selmi 2, 40126, Bologna, Italy E-Mail
| | - Francesco Zerbetto
- Dipartimento di Chimica "Giacomo Ciamician", Alma Mater Studiorum - Università di Bologna, Via Francesco Selmi 2, 40126, Bologna, Italy E-Mail
| | - Edoardo Jun Mattioli
- Dipartimento di Chimica "Giacomo Ciamician", Alma Mater Studiorum - Università di Bologna, Via Francesco Selmi 2, 40126, Bologna, Italy E-Mail
| | - Matteo Calvaresi
- Dipartimento di Chimica "Giacomo Ciamician", Alma Mater Studiorum - Università di Bologna, Via Francesco Selmi 2, 40126, Bologna, Italy E-Mail
| |
Collapse
|
9
|
Truong DT, Ho K, Nhi HTY, Nguyen VH, Dang TT, Nguyen MT. Imidazole[1,5-a]pyridine derivatives as EGFR tyrosine kinase inhibitors unraveled by umbrella sampling and steered molecular dynamics simulations. Sci Rep 2024; 14:12218. [PMID: 38806555 PMCID: PMC11133355 DOI: 10.1038/s41598-024-62743-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 05/21/2024] [Indexed: 05/30/2024] Open
Abstract
Although the use of the tyrosine kinase inhibitors (TKIs) has been proved that it can save live in a cancer treatment, the currently used drugs bring in many undesirable side-effects. Therefore, the search for new drugs and an evaluation of their efficiency are intensively carried out. Recently, a series of eighteen imidazole[1,5-a]pyridine derivatives were synthetized by us, and preliminary analyses pointed out their potential to be an important platform for pharmaceutical development owing to their promising actions as anticancer agents and enzyme (kinase, HIV-protease,…) inhibitors. In the present theoretical study, we further analyzed their efficiency in using a realistic scenario of computational drug design. Our protocol has been developed to not only observe the atomistic interaction between the EGFR protein and our 18 novel compounds using both umbrella sampling and steered molecular dynamics simulations, but also determine their absolute binding free energies. Calculated properties of the 18 novel compounds were in detail compared with those of two known drugs, erlotinib and osimertinib, currently used in cancer treatment. Inspiringly the simulation results promote three imidazole[1,5-a]pyridine derivatives as promising inhibitors into a further step of clinical trials.
Collapse
Affiliation(s)
- Duc Toan Truong
- Laboratory for Chemical Computation and Modeling, Institute for Computational Science and Artificial Intelligence, Van Lang University, Ho Chi Minh City, 70000, Vietnam
- Faculty of Applied Technology, School of Technology, Van Lang University, Ho Chi Minh City, 70000, Vietnam
| | - Kiet Ho
- Institute for Computational Science and Technology (ICST), Quang Trung Software City, Ho Chi Minh City, 70000, Vietnam
| | - Huynh Thi Yen Nhi
- Laboratory for Chemical Computation and Modeling, Institute for Computational Science and Artificial Intelligence, Van Lang University, Ho Chi Minh City, 70000, Vietnam
| | - Van Ha Nguyen
- Faculty of Chemistry, VNU University of Science, Vietnam National University, Hanoi, 19 Le Thanh Tong, Hoan Kiem, Hanoi, 11021, Vietnam
| | - Tuan Thanh Dang
- Faculty of Chemistry, VNU University of Science, Vietnam National University, Hanoi, 19 Le Thanh Tong, Hoan Kiem, Hanoi, 11021, Vietnam
| | - Minh Tho Nguyen
- Laboratory for Chemical Computation and Modeling, Institute for Computational Science and Artificial Intelligence, Van Lang University, Ho Chi Minh City, 70000, Vietnam.
- Faculty of Applied Technology, School of Technology, Van Lang University, Ho Chi Minh City, 70000, Vietnam.
| |
Collapse
|
10
|
Said AAE, Mahmoud BK, Helmy AM, Mohamed NM, Attia EZ, Samy MN, Abdelmohsen UR, Fouad MA. Niosomes as promising approach for enhancing the cytotoxicity of Hemimycale sp. total crude extract supported with in-silico studies. Sci Rep 2024; 14:2546. [PMID: 38291122 PMCID: PMC10827731 DOI: 10.1038/s41598-024-52918-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 01/25/2024] [Indexed: 02/01/2024] Open
Abstract
The crude extract of Hemimycale sp. marine sponge was evaluated as a cytotoxic drug against different cell lines; whereas it exhibited promising selective activity toward the breast cancer cell line only with IC50 value 199.6 ± 0.00512 µg/ml. Moreover, its cytotoxic activity against the breast cancer cell line was reevaluated upon forming total extract-loaded niosomes. This revealed an IC50 value of 44.35 ± 0.011128 µg/ml, indicating the potential contribution of niosomes in boosting cell penetration and activity as a result. Owing to highlight the bioactive constituents responsible for the cytotoxic activity, metabolomics profiling of Hemimycale sp. was performed using liquid chromatography coupled with high-resolution electrospray ionization mass spectrometry (LC-HR-ESI-MS) revealing tentative identification of phytoconstituents clusters like as, diterpenes, sesterterpenes and sterols. Additionally, the cytotoxic activity of the crude extract was explained on the molecular level, whereas the dereplicated compounds were evaluated in silico against the Epidermal Growth Factor Receptor tyrosine kinase (EGFR). The sesterterpenoid derivatives phorbaketal A acetate (12) and secoepoxy ansellone A (13) together with mycalol-522 (17) showed the best binding energy.
Collapse
Affiliation(s)
- Asmaa Abo Elgoud Said
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| | - Basma Khalaf Mahmoud
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| | - Abdelrahman M Helmy
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Deraya University, Minya, Egypt
- Pharmaceutical Engineering and 3D Printing (PharmE3D) Lab, Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Nada M Mohamed
- Pharmaceutical Chemistry Department, Modern University for Technology and Information (MTI), Cairo, Egypt
| | - Eman Zekry Attia
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| | - Mamdouh Nabil Samy
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| | - Usama Ramadan Abdelmohsen
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt.
- Department of Pharmacognosy, Faculty of Pharmacy, Deraya University, Universities Zone, New Minia City, 61111, Egypt.
| | - Mostafa A Fouad
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| |
Collapse
|
11
|
Foschi F, Tinivella A, Crippa V, Pinzi L, Mologni L, Passarella D, Rastelli G. Structure-activity exploration of a small-molecule allosteric inhibitor of T790M/L858R double mutant EGFR. J Enzyme Inhib Med Chem 2023; 38:239-245. [PMID: 36373202 PMCID: PMC9665079 DOI: 10.1080/14756366.2022.2145284] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
EGFR is a protein kinase whose aberrant activity is frequently involved in the development of non-small lung cancer (NSCLC) drug resistant forms. The allosteric inhibition of this enzyme is currently one among the most attractive approaches to design and develop anticancer drugs. In a previous study, we reported the identification of a hit compound acting as type III allosteric inhibitor of the L858R/T790M double mutant EGFR. Herein, we report the design, synthesis and in vitro testing of a series of analogues of the previously identified hit with the aim of exploring the structure-activity relationships (SAR) around this scaffold. The performed analyses allowed us to identify two compounds 15 and 18 showing improved inhibition of double mutant EGFR with respect to the original hit, as well as interesting antiproliferative activity against H1975 NSCLC cancer cells expressing double mutant EGFR. The newly discovered compounds represent promising starting points for further hit-to-lead optimisation.
Collapse
Affiliation(s)
- Francesca Foschi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy.,Department of Chemistry, University of Milano, Milano, Italy
| | - Annachiara Tinivella
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Valentina Crippa
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Luca Pinzi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Luca Mologni
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | | | - Giulio Rastelli
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
12
|
Taher Mohie El-Dien R, Mahmoud BK, Abdelwahab MF, Khedr AIM, Kamel MS, Yahia R, Mohamed NM, Zawily AE, Kamel ES, Salem AK, Abdelmohsen UR, Fouad MA. Paralemnalia thyrsoides-associated fungi: phylogenetic diversity, cytotoxic potential, metabolomic profiling and docking analysis. BMC Microbiol 2023; 23:308. [PMID: 37884900 PMCID: PMC10601334 DOI: 10.1186/s12866-023-03045-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 10/06/2023] [Indexed: 10/28/2023] Open
Abstract
BACKGROUND Cancer continues to be one of the biggest causes of death that affects human health. Chemical resistance is still a problem in conventional cancer treatments. Fortunately, numerous natural compounds originating from different microbes, including fungi, possess cytotoxic characteristics that are now well known. This study aims to investigate the anticancer prospects of five fungal strains that were cultivated and isolated from the Red Sea soft coral Paralemnalia thyrsoides. The in vitro cytotoxic potential of the ethyl acetate extracts of the different five isolates were evaluated using MTS assay against four cancer cell lines; A549, CT-26, MDA-MB-231, and U87. Metabolomics profiling of the different extracts using LC-HR-ESI-MS, besides molecular docking studies for the dereplicated compounds were performed to unveil the chemical profile and the cytotoxic mechanism of the soft coral associated fungi. RESULTS The five isolated fungal strains were identified as Penicillium griseofulvum (RD1), Cladosporium sphaerospermum (RD2), Cladosporium liminiforme (RD3), Penicillium chrysogenum (RD4), and Epicoccum nigrum (RD5). The in vitro study showed that the ethyl acetate extract of RD4 exhibited the strongest cytotoxic potency against three cancer cell lines A549, CT-26 and MDA-MB-231 with IC50 values of 1.45 ± 8.54, 1.58 ± 6.55 and 1.39 ± 2.0 µg/mL, respectively, also, RD3 revealed selective cytotoxic potency against A549 with IC50 value of 6.99 ± 3.47 µg/mL. Docking study of 32 compounds dereplicated from the metabolomics profiling demonstrated a promising binding conformation with EGFR tyrosine kinase that resembled its co-crystallized ligand albeit with better binding energy score. CONCLUSION Our results highlight the importance of soft coral-associated fungi as a promising source for anticancer metabolites for future drug discovery.
Collapse
Affiliation(s)
- Radwa Taher Mohie El-Dien
- Department of Pharmacognosy, Faculty of pharmacy, New Valley University, New Valley City, Egypt
- Department of Pharmacognosy, Faculty of Pharmacy, Deraya University, 61111, New Minia City, Minia, Egypt
| | - Basma Khalaf Mahmoud
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, 61519, Minia, Egypt
| | - Miada F Abdelwahab
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, 61519, Minia, Egypt
| | - Amgad I M Khedr
- Department of Pharmacognosy, Faculty of Pharmacy, Port Said University, 42526, Port Said, Egypt
| | - Mohamed Salah Kamel
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, 61519, Minia, Egypt
- Department of Pharmacognosy, Faculty of Pharmacy, Deraya University, 61111, New Minia City, Minia, Egypt
| | - Ramadan Yahia
- Department of Microbiology and immunology, Faculty of Pharmacy, Deraya University, 61111, New Minia City, Minia, Egypt
| | - Nada M Mohamed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo, Egypt
| | - Amr El Zawily
- Department of Plant and Microbiology, Faculty of Science, Damanhour University, 22511, Damanhour, Egypt.
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, 52242, USA.
| | - Eman S Kamel
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, 52242, USA
| | - Aliasger K Salem
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, 52242, USA
| | - Usama Ramadan Abdelmohsen
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, 61519, Minia, Egypt.
- Department of Pharmacognosy, Faculty of Pharmacy, Deraya University, 61111, New Minia City, Minia, Egypt.
| | - Mostafa A Fouad
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, 61519, Minia, Egypt
| |
Collapse
|
13
|
Shama Bhat G, Shaik Mohammad F. Computational Fragment-Based Design of Phytochemical Derivatives as EGFR Inhibitors. Chem Biodivers 2023; 20:e202300681. [PMID: 37399183 DOI: 10.1002/cbdv.202300681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/30/2023] [Accepted: 07/03/2023] [Indexed: 07/05/2023]
Abstract
Epidermal growth factor receptor (EGFR) is a potential target with disease modifying benefits against Alzheimer's disease (AD). Repurposing of FDA approved drugs against EGFR have shown beneficial effect against AD but are confined to quinazoline, quinoline and aminopyrimidines. Futuristically, the possibility of acquiring drug resistance mutation as seen in the case of cancer could also hamper AD treatment. To identify novel chemical scaffolds, we rooted on phytochemicals identified from plants such as Acorus calamus, Bacopa monnieri, Convolvulus pluricaulis, Tinospora cordifloia, and Withania somnifera that have well-established records in the treatment of brain disorders. The rationale was to mimic the biosynthetic metabolite extension process observed in the plants for synthesizing new phytochemical derivates. Thus, novel compounds were designed computationally by fragment-based method followed by extensive in silico analysis to pick potential phytochemical derivates. PCD1, 8 and 10 were predicted to have better blood brain barrier permeability. ADMET and SoM analysis suggested that these PCDs exhibited druglike properties. Further simulation studies showed that PCD1 and PCD8 stably interact with EGFR and have the potential to be used even in cases of drug-resistance mutations. With further experimental evidence, these PCDs could be leveraged as potential inhibitors of EGFR.
Collapse
Affiliation(s)
- Gayathri Shama Bhat
- Department of Biotechnology, Manipal Institute of Technology, Manipal, 576104, Karnataka, India
| | - Fayaz Shaik Mohammad
- Department of Biotechnology, Manipal Institute of Technology, Manipal, 576104, Karnataka, India
| |
Collapse
|
14
|
Gudasi S, Gharge S, Koli R, Patil K. Antioxidant properties and cytotoxic effects of Oxalis corniculata on human Hepatocarcinoma (Hep-G2) cell line: an in vitro and in silico evaluation. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2023. [DOI: 10.1186/s43094-023-00476-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023] Open
Abstract
Abstract
Background
Oxalis corniculata is a well-known medicinal plant used in folk medicine for the management of many diseases. The aim is to determine the physico-chemical properties, microscopic study, phytochemical properties, in-silico, in-vitro antioxidant and anticancer activity on human Hepatocarcinoma (Hep-G2) cell line of O. corniculata plant extract.
Results
Microscopical study reaveals that presence of pericyclic fibres, starch gains, trichomes etc, and phytochemical screening is carried to find out secondary metabolites. The molecular docking study concluded that some of the phytocompounds showed inhibition of epidermal growth factor receptor tyrosine kinase domain (PDB ID: 1M17) inhibitor. Furthermore, ADMET and drug likeness study hints some of phytocompounds may act as lead for anticancer drug discovery and development. Among selected phytocompounds, compound apigenin possesses − 7.90 kcal/mol as compared to standard drug doxorubicin possesses − 7.63 kcal/mol against the epidermal growth factor receptor tyrosine kinase. The plant extract shown antioxidant activities based on the different tests were performed. The hydroalcoholic plant extracts were found to be selectively cytotoxic in vitro to human Hepatocarcinoma (Hep-G2) cell line with IC50values 34.494 ± 0.42 µg/ml and EAF showed at IC50 value 30.245 ± 0.58 µg/ml of the cells were inhibited at the concentration of 50 µg/ml as compared with standard doxorubicin at IC50 value 24.8939 ± 0.25 µg/ml, respectively.
Conclusion
The present study concluded that O. corniculata possesses potential antioxidant and cytotoxic properties based upon the computer aided drug design models and in-vitro activity.
Collapse
|
15
|
Sattler M, Mambetsariev I, Fricke J, Tan T, Liu S, Vaidehi N, Pisick E, Mirzapoiazova T, Rock AG, Merla A, Sharma S, Salgia R. A Closer Look at EGFR Inhibitor Resistance in Non-Small Cell Lung Cancer through the Lens of Precision Medicine. J Clin Med 2023; 12:jcm12051936. [PMID: 36902723 PMCID: PMC10003860 DOI: 10.3390/jcm12051936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/22/2023] [Accepted: 02/26/2023] [Indexed: 03/05/2023] Open
Abstract
The development of EGFR small-molecule inhibitors has provided significant benefit for the affected patient population. Unfortunately, current inhibitors are no curative therapy, and their development has been driven by on-target mutations that interfere with binding and thus inhibitory activity. Genomic studies have revealed that, in addition to these on-target mutations, there are also multiple off-target mechanisms of EGFR inhibitor resistance and novel therapeutics that can overcome these challenges are sought. Resistance to competitive 1st-generation and covalent 2nd- and 3rd-generation EGFR inhibitors is overall more complex than initially thought, and novel 4th-generation allosteric inhibitors are expected to suffer from a similar fate. Additional nongenetic mechanisms of resistance are significant and can include up to 50% of the escape pathways. These potential targets have gained recent interest and are usually not part of cancer panels that look for alterations in resistant patient specimen. We discuss the duality between genetic and nongenetic EGFR inhibitor drug resistance and summarize current team medicine approaches, wherein clinical developments, hand in hand with drug development research, drive potential opportunities for combination therapy.
Collapse
Affiliation(s)
- Martin Sattler
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave., Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
- Correspondence:
| | - Isa Mambetsariev
- Department of Medical Oncology and Therapeutics Research, City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA
| | - Jeremy Fricke
- Department of Medical Oncology and Therapeutics Research, City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA
| | - Tingting Tan
- Department of Medical Oncology and Therapeutics Research, City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA
| | - Sariah Liu
- Department of Medical Oncology and Therapeutics Research, City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA
| | - Nagarajan Vaidehi
- Department of Computational and Quantitative Medicine, City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA
| | - Evan Pisick
- City of Hope Chicago, 2520 Elisha Avenue, Zion, IL 60099, USA
| | - Tamara Mirzapoiazova
- Department of Medical Oncology and Therapeutics Research, City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA
| | - Adam G. Rock
- Department of Medical Oncology and Therapeutics Research, City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA
| | - Amartej Merla
- Department of Medical Oncology and Therapeutics Research, City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA
| | - Sunil Sharma
- Division of Applied Cancer Research and Drug Discovery, Translational Genomic Research Institute (Tgen), 445 N 5th St, Phoenix, AZ 85004, USA
| | - Ravi Salgia
- Department of Medical Oncology and Therapeutics Research, City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA
| |
Collapse
|
16
|
Qureshi R, Zou B, Alam T, Wu J, Lee VHF, Yan H. Computational Methods for the Analysis and Prediction of EGFR-Mutated Lung Cancer Drug Resistance: Recent Advances in Drug Design, Challenges and Future Prospects. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:238-255. [PMID: 35007197 DOI: 10.1109/tcbb.2022.3141697] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Lung cancer is a major cause of cancer deaths worldwide, and has a very low survival rate. Non-small cell lung cancer (NSCLC) is the largest subset of lung cancers, which accounts for about 85% of all cases. It has been well established that a mutation in the epidermal growth factor receptor (EGFR) can lead to lung cancer. EGFR Tyrosine Kinase Inhibitors (TKIs) are developed to target the kinase domain of EGFR. These TKIs produce promising results at the initial stage of therapy, but the efficacy becomes limited due to the development of drug resistance. In this paper, we provide a comprehensive overview of computational methods, for understanding drug resistance mechanisms. The important EGFR mutants and the different generations of EGFR-TKIs, with the survival and response rates are discussed. Next, we evaluate the role of important EGFR parameters in drug resistance mechanism, including structural dynamics, hydrogen bonds, stability, dimerization, binding free energies, and signaling pathways. Personalized drug resistance prediction models, drug response curve, drug synergy, and other data-driven methods are also discussed. Recent advancements in deep learning; such as AlphaFold2, deep generative models, big data analytics, and the applications of statistics and permutation are also highlighted. We explore limitations in the current methodologies, and discuss strategies to overcome them. We believe this review will serve as a reference for researchers; to apply computational techniques for precision medicine, analyzing structures of protein-drug complexes, drug discovery, and understanding the drug response and resistance mechanisms in lung cancer patients.
Collapse
|
17
|
Nasrollahpour H, Khalilzadeh B, Hasanzadeh M, Rahbarghazi R, Estrela P, Naseri A, Tasoglu S, Sillanpää M. Nanotechnology‐based electrochemical biosensors for monitoring breast cancer biomarkers. Med Res Rev 2022; 43:464-569. [PMID: 36464910 DOI: 10.1002/med.21931] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 10/01/2022] [Accepted: 11/04/2022] [Indexed: 12/07/2022]
Abstract
Breast cancer is categorized as the most widespread cancer type among women globally. On-time diagnosis can decrease the mortality rate by making the right decision in the therapy procedure. These features lead to a reduction in medication time and socioeconomic burden. The current review article provides a comprehensive assessment for breast cancer diagnosis using nanomaterials and related technologies. Growing use of the nano/biotechnology domain in terms of electrochemical nanobiosensor designing was discussed in detail. In this regard, recent advances in nanomaterial applied for amplified biosensing methodologies were assessed for breast cancer diagnosis by focusing on the advantages and disadvantages of these approaches. We also monitored designing methods, advantages, and the necessity of suitable (nano) materials from a statistical standpoint. The main objective of this review is to classify the applicable biosensors based on breast cancer biomarkers. With numerous nano-sized platforms published for breast cancer diagnosis, this review tried to collect the most suitable methodologies for detecting biomarkers and certain breast cancer cell types.
Collapse
Affiliation(s)
- Hassan Nasrollahpour
- Department of Analytical Chemistry, Faculty of Chemistry University of Tabriz Tabriz Iran
| | - Balal Khalilzadeh
- Stem Cell Research Center Tabriz University of Medical Sciences Tabriz Iran
| | - Mohammad Hasanzadeh
- Pharmaceutical Analysis Research Center Tabriz University of Medical Sciences Tabriz Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center Tabriz University of Medical Sciences Tabriz Iran
- Department of Applied Cellular Sciences, Faculty of Advanced Medical Sciences Tabriz University of Medical Sciences Tabriz Iran
| | - Pedro Estrela
- Centre for Biosensors, Bioelectronics and Biodevices (C3Bio) and Department of Electronic and Electrical Engineering University of Bath Bath UK
| | - Abdolhossein Naseri
- Department of Analytical Chemistry, Faculty of Chemistry University of Tabriz Tabriz Iran
| | - Savas Tasoglu
- Koç University Translational Medicine Research Center (KUTTAM) Rumeli Feneri, Sarıyer Istanbul Turkey
| | - Mika Sillanpää
- Environmental Engineering and Management Research Group Ton Duc Thang University Ho Chi Minh City Vietnam
- Faculty of Environment and Labour Safety Ton Duc Thang University Ho Chi Minh City Vietnam
| |
Collapse
|
18
|
Identification of a novel nitroflavone-based scaffold for designing mutant-selective EGFR tyrosine kinase inhibitors targeting T790M and C797S resistance in advanced NSCLC. Bioorg Chem 2022; 129:106219. [DOI: 10.1016/j.bioorg.2022.106219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/05/2022] [Accepted: 10/17/2022] [Indexed: 11/17/2022]
|
19
|
Zhao Z, Bourne PE. Harnessing systematic protein-ligand interaction fingerprints for drug discovery. Drug Discov Today 2022; 27:103319. [PMID: 35850431 DOI: 10.1016/j.drudis.2022.07.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 07/04/2022] [Accepted: 07/12/2022] [Indexed: 12/15/2022]
Abstract
Determining protein-ligand interaction characteristics and mechanisms is crucial to the drug discovery process. Here, we review recent progress and successful applications of a systematic protein-ligand interaction fingerprint (IFP) approach for investigating proteome-wide protein-ligand interactions for drug development. Specifically, we review the use of this IFP approach for revealing polypharmacology across the kinome, predicting promising targets from which to design allosteric inhibitors and covalent kinase inhibitors, uncovering the binding mechanisms of drugs of interest, and demonstrating resistant mechanisms of specific drugs. Together, we demonstrate that the IFP strategy is efficient and practical for drug design research for protein kinases as targets and is extensible to other protein families.
Collapse
Affiliation(s)
- Zheng Zhao
- School of Data Science and Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22904, USA.
| | - Philip E Bourne
- School of Data Science and Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22904, USA.
| |
Collapse
|
20
|
Wang F, Du H, Li B, Luo Z, Zhu L. Unlocking phenotypic plasticity provides novel insights for immunity and personalized therapy in lung adenocarcinoma. Front Genet 2022; 13:941567. [PMID: 36147496 PMCID: PMC9486167 DOI: 10.3389/fgene.2022.941567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Unlocking phenotype plasticity (UPP) has been shown to have an essential role in the mechanism of tumor development and therapeutic response. However, the clinical significance of unlocking phenotypic plasticity in patients with lung adenocarcinoma is unclear. This study aimed to explore the roles of unlocking phenotypic plasticity in immune status, prognosis, and treatment in patients with lung adenocarcinoma (LUAD). Methods: Differentially expressed genes (DEGs) and clinical information of UPP were selected from the cancer genome atlas (TCGA) database, and the GO, KEGG enrichment analyses were performed. The independent prognostic genes were determined by univariate and multivariate Cox regression, and the UPP signature score was constructed. Patients with LUAD were divided into high- and low-risk groups according to the median of score, and the immunocytes and immune function, the gene mutation, and drug sensitivities between the two groups were analyzed. Finally, the results were validated in the GEO database. Results: Thirty-nine significantly DEGs were determined. Enrichment analysis showed that UPP-related genes were related to protein polysaccharides and drug resistance. The prognostic results showed that the survival of patients in the high-risk group was poorer than that in the low-risk group (p < 0.001). In the high- and low-risk groups, single nucleotide polymorphism (SNP) and C > T are the most common dissent mutations. The contents of immune cells were significantly different between high- and low-risk groups. And the immune functions were also significantly different, indicating that UPP affects the immunity in LUAD. The results from TCGA were validated in the GEO. Conclusion: Our research has proposed a new and reliable prognosis indicator to predict the overall survival. Evaluation of the UPP could help the clinician to predict therapeutic responses and make individualized treatment plans in patients with LUAD.
Collapse
Affiliation(s)
- Feng Wang
- Department of Oncology, Chongqing General Hospital, Chongqing, China
| | - Hongjuan Du
- Department of Oncology, Chongqing General Hospital, Chongqing, China
| | - Bibo Li
- Department of Oncology, Chongqing General Hospital, Chongqing, China
| | - Zhibin Luo
- Department of Oncology, Chongqing General Hospital, Chongqing, China
| | - Lei Zhu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Lei Zhu,
| |
Collapse
|
21
|
Mahalapbutr P, Leechaisit R, Thongnum A, Todsaporn D, Prachayasittikul V, Rungrotmongkol T, Prachayasittikul S, Ruchirawat S, Prachayasittikul V, Pingaew R. Discovery of Anilino-1,4-naphthoquinones as Potent EGFR Tyrosine Kinase Inhibitors: Synthesis, Biological Evaluation, and Comprehensive Molecular Modeling. ACS OMEGA 2022; 7:17881-17893. [PMID: 35664590 PMCID: PMC9161259 DOI: 10.1021/acsomega.2c01188] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/21/2022] [Indexed: 05/02/2023]
Abstract
Epidermal growth factor receptor (EGFR) has been recognized as one of the attractive targets for anticancer drug development. Herein, a set of anilino-1,4-naphthoquinone derivatives (3-18) was synthesized and investigated for their anticancer and EGFR inhibitory potentials. Among all tested compounds, three derivatives (3, 8, and 10) were selected for studying EGFR inhibitory activity (in vitro and in silico) due to their most potent cytotoxic activities against six tested cancer cell lines (i.e., HuCCA-1, HepG2, A549, MOLT-3, MDA-MB-231, and T47D; IC50 values = 1.75-27.91 μM), high selectivity index (>20), and good predicted drug-like properties. The experimental results showed that these three promising compounds are potent EGFR inhibitors with nanomolar IC50 values (3.96-18.64 nM). Interestingly, the most potent compound 3 bearing 4-methyl substituent on the phenyl ring displayed 4-fold higher potency than the known EGFR inhibitor, erlotinib. Molecular docking, molecular dynamics simulation, and MM/GBSA-based free energy calculation revealed that van der Waals force played a major role in the accommodations of compound 3 within the ATP-binding pocket of EGFR. Additionally, the 4-CH3 moiety of the compound was noted to be a key chemical feature contributing to the highly potent EGFR inhibitory activity via its formations of alkyl interactions with A743, K745, M766, and L788 residues as well as additional interactions with M766 and T790.
Collapse
Affiliation(s)
- Panupong Mahalapbutr
- Department
of Biochemistry, and Center for Translational Medicine, Faculty of
Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Ronnakorn Leechaisit
- Department
of Chemistry, Faculty of Science, Srinakharinwirot
University, Bangkok 10110, Thailand
| | - Anusit Thongnum
- Department
of Physics, Faculty of Science, Srinakharinwirot
University, Bangkok 10110, Thailand
| | - Duangjai Todsaporn
- Structural
and Computational Biology Research Unit, Department of Biochemistry,
Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Veda Prachayasittikul
- Center
of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand
| | - Thanyada Rungrotmongkol
- Structural
and Computational Biology Research Unit, Department of Biochemistry,
Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
- Program
in Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok 10330, Thailand
| | - Supaluk Prachayasittikul
- Center
of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand
| | - Somsak Ruchirawat
- Laboratory
of Medicinal Chemistry and Program in Chemical Sciences, Chulabhorn Research Institute, Chulabhorn Graduate
Institute, Bangkok 10210, Thailand
- Commission
on Higher Education, Ministry of Education, Center of Excellence on Environmental Health and Toxicology (EHT), Bangkok 10400, Thailand
| | - Virapong Prachayasittikul
- Department
of Clinical Microbiology and Applied Technology, Faculty of Medical
Technology, Mahidol University, Bangkok 10700, Thailand
| | - Ratchanok Pingaew
- Department
of Chemistry, Faculty of Science, Srinakharinwirot
University, Bangkok 10110, Thailand
| |
Collapse
|
22
|
Structural Insight and Development of EGFR Tyrosine Kinase Inhibitors. Molecules 2022; 27:molecules27030819. [PMID: 35164092 PMCID: PMC8838133 DOI: 10.3390/molecules27030819] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/23/2022] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
Lung cancer has a high prevalence, with a growing number of new cases and mortality every year. Furthermore, the survival rate of patients with non-small-cell lung carcinoma (NSCLC) is still quite low in the majority of cases. Despite the use of conventional therapy such as tyrosine kinase inhibitor for Epidermal Growth Factor Receptor (EGFR), which is highly expressed in most NSCLC cases, there was still no substantial improvement in patient survival. This is due to the drug’s ineffectiveness and high rate of resistance among individuals with mutant EGFR. Therefore, the development of new inhibitors is urgently needed. Understanding the EGFR structure, including its kinase domain and other parts of the protein, and its activation mechanism can accelerate the discovery of novel compounds targeting this protein. This study described the structure of the extracellular, transmembrane, and intracellular domains of EGFR. This was carried out along with identifying the binding pose of commercially available inhibitors in the ATP-binding and allosteric sites, thereby clarifying the research gaps that can be filled. The binding mechanism of inhibitors that have been used clinically was also explained, thereby aiding the structure-based development of new drugs.
Collapse
|
23
|
Elrayess R, Darwish KM, Nafie MS, El-Sayyed GS, Said MM, Yassen ASA. Quinoline–hydrazone hybrids as dual mutant EGFR inhibitors with promising metallic nanoparticle loading: rationalized design, synthesis, biological investigation and computational studies. NEW J CHEM 2022. [DOI: 10.1039/d2nj02962f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A novel quinoline–hydrazone hybrid induced apoptosis in MCF-7 cells through dual mutant EGFR inhibition with promising metallic nanoparticle loading.
Collapse
Affiliation(s)
- Ranza Elrayess
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Khaled M. Darwish
- Medicinal Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Mohamed S. Nafie
- Chemistry Department, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt
| | - Gharieb S. El-Sayyed
- Microbiology and Immunology Department, Faculty of Pharmacy, Galala University, New Galala City, Suez, Egypt
- Drug Radiation Research Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| | - Mohamed M. Said
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Asmaa S. A. Yassen
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
24
|
Bashant MM, Mitchell SM, Hart LR, Lebedenko CG, Banerjee IA. In silico studies of interactions of peptide-conjugated cholesterol metabolites and betulinic acid with EGFR, LDR, and N-terminal fragment of CCKA receptors. J Mol Model 2021; 28:16. [PMID: 34961887 DOI: 10.1007/s00894-021-05007-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 12/14/2021] [Indexed: 11/25/2022]
Abstract
In this work, we designed three new ligands by conjugating cholesterol metabolites 3-hydroxy-5-cholestenoic acid (3-HC) and 3-oxo-4-cholestenoic acid (3-OC) and the natural tri-terpenoid betulinic acid with the tumor-targeting peptide YHWYGYTPQNVI. Molecular interactions with the unconjugated peptide and the conjugates were examined with three receptors that are commonly overexpressed in pancreatic adenocarcinoma cells using ligand docking and molecular dynamics. This study demonstrated the utility of the designed conjugates as a valuable scaffold for potentially targeting EGFR and LDLR receptors. Our results indicate that the conjugates showed strong binding affinities and formation of stable complexes with EGFR, while the unconjugated peptide, BT-peptide conjugate, an 3-HC-peptide conjugate showed the formation of fairly stable complexes with LDLR receptor. For EGFR, two receptor kinase domains were explored. Interactions with the N-terminal domain of CCKA-R were relatively weaker. For LDLR, binding occurred in the beta-propeller region. For the N-terminal fragment of CCKA-R, the conjugates induced significant conformational changes in the receptor. The molecular dynamic simulations for 100 ns demonstrate that BT-peptide conjugates and the unconjugated peptide had the highest binding and formed the most stable complexes with EGFR. RMSD and trajectory analyses indicate that these molecules transit to a dynamically stable configuration in most cases within 60 ns. NMA analysis indicated that amongst the conjugates that showed relatively higher interactions with the respective receptors, the highest potential for deformability was seen for the N-terminal-47 amino acid region of the CCKA-R receptor with and the lowest for the LDLR-receptor. Thus, the newly designed compounds may be evaluated in the future toward developing drug delivery materials for targeting tumor cells overexpressing LDLR or EGFR.
Collapse
Affiliation(s)
- Madeline M Bashant
- Department of Chemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Saige M Mitchell
- Department of Chemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Lucy R Hart
- Department of Chemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Charlotta G Lebedenko
- Department of Chemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA
| | - Ipsita A Banerjee
- Department of Chemistry, Fordham University, 441 East Fordham Road, Bronx, NY, 10458, USA.
| |
Collapse
|
25
|
Kiriwan D, Seetaha S, Jiwacharoenchai N, Tabtimmai L, Sousa SF, Songtawee N, Choowongkomon K. Identification of tripeptides against tyrosine kinase domain of EGFR for lung cancer cell inhibition by in silico and in vitro studies. Chem Biol Drug Des 2021; 99:456-469. [PMID: 34923743 DOI: 10.1111/cbdd.14010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/10/2021] [Accepted: 12/11/2021] [Indexed: 12/30/2022]
Abstract
Epidermal growth factor receptor tyrosine kinase domain (EGFR-TK) has been one of the prominent targets for therapeutics of several human cancers, in particular non-small cell lung cancer. Although several small chemical compounds targeting EGFR-TK have been approved by FDA for treatment of such a cancer, the discovery of a new class of EGFR-TK inhibitors, for example, small peptides, is still desired. In this study, using molecular docking-based virtual screening, we selected five small peptides with high docking scores from eight thousand peptides as candidate compounds against EGFR-TK. Among five, the tripeptide WFF had the most potency to suppress the survival of non-small cell lung cancer cells but had the least toxicity to human liver cancer cells. Our in vitro kinase assays showed that WFF exhibited much lower inhibitory activity against purified EGFR-TK than the drug erlotinib (i.e., IC50 values of ≈ 0.62 μM vs ≈ 7.57 nM, respectively). The relative free binding energies estimated from molecular dynamic simulations were consistent with the in vitro experiments in which the WFF bound had a lower affinity than erlotinib bound to EGFR-TK (i.e., ΔGbind values of -20.3 kJ/mol vs ≈ -126.8 kJ/mol, respectively). In addition, the simulation analyses demonstrated the difference in EGFR binding preference between the drug and tripeptide in which erlotinib was stably bound in the ATP-binding pocket for 4-anilinoquinazoline class of inhibitors, while WFF moved out of that pocket to interact with polar amino acid residues on the αC-helix, activation loop, and substrate-binding region. Our findings suggest preferable interactions of the potential tripeptide on enzyme inhibition that are useful for further development of a new class of inhibitors targeting EGFR-TK.
Collapse
Affiliation(s)
- Duangnapa Kiriwan
- Genetic Engineering and Bioinformatics Program, Kasetsart University, Bangkok, Thailand
| | - Supaphorn Seetaha
- Department of Biochemistry, Faculty of Science, Kasetsart University, Bangkok, Thailand
| | | | - Lueacha Tabtimmai
- Department of Biotechnology, Faculty of Applied Science, King Mongkut's University of Technology North Bangkok, Bangkok, Thailand
| | - Sérgio F Sousa
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculdade de Medicina, Universidade do Porto, Porto, Portugal.,UCIBIO-Applied Molecular Biosciences Unit, BioSIM-Departamento de Biomedicina, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| | - Napat Songtawee
- Department of Clinical Chemistry, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, Thailand
| | - Kiattawee Choowongkomon
- Department of Biochemistry, Faculty of Science, Kasetsart University, Bangkok, Thailand.,Center for Advanced Studies in Nanotechnology for Chemical, Food and Agricultural Industries, KU Institute for Advanced Studies, Kasetsart University, Bangkok, Thailand
| |
Collapse
|
26
|
Abourehab MAS, Alqahtani AM, Youssif BGM, Gouda AM. Globally Approved EGFR Inhibitors: Insights into Their Syntheses, Target Kinases, Biological Activities, Receptor Interactions, and Metabolism. Molecules 2021; 26:6677. [PMID: 34771085 PMCID: PMC8587155 DOI: 10.3390/molecules26216677] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/28/2021] [Accepted: 11/02/2021] [Indexed: 02/06/2023] Open
Abstract
Targeting the EGFR with small-molecule inhibitors is a confirmed valid strategy in cancer therapy. Since the FDA approval of the first EGFR-TKI, erlotinib, great efforts have been devoted to the discovery of new potent inhibitors. Until now, fourteen EGFR small-molecule inhibitors have been globally approved for the treatment of different types of cancers. Although these drugs showed high efficacy in cancer therapy, EGFR mutations have emerged as a big challenge for these drugs. In this review, we focus on the EGFR small-molecule inhibitors that have been approved for clinical uses in cancer therapy. These drugs are classified based on their chemical structures, target kinases, and pharmacological uses. The synthetic routes of these drugs are also discussed. The crystal structures of these drugs with their target kinases are also summarized and their bonding modes and interactions are visualized. Based on their binding interactions with the EGFR, these drugs are also classified into reversible and irreversible inhibitors. The cytotoxicity of these drugs against different types of cancer cell lines is also summarized. In addition, the proposed metabolic pathways and metabolites of the fourteen drugs are discussed, with a primary focus on the active and reactive metabolites. Taken together, this review highlights the syntheses, target kinases, crystal structures, binding interactions, cytotoxicity, and metabolism of the fourteen globally approved EGFR inhibitors. These data should greatly help in the design of new EGFR inhibitors.
Collapse
Affiliation(s)
- Mohammed A. S. Abourehab
- Department of Pharmaceutics, Faculty of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
| | - Alaa M. Alqahtani
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Bahaa G. M. Youssif
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt;
| | - Ahmed M. Gouda
- Department of Medicinal Chemistry, Faculty of pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| |
Collapse
|
27
|
Antiproliferative activity, enzymatic inhibition and apoptosis-promoting effects of benzoxazole-based hybrids on human breast cancer cells. Bioorg Chem 2021; 109:104752. [PMID: 33657444 DOI: 10.1016/j.bioorg.2021.104752] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 01/26/2021] [Accepted: 02/15/2021] [Indexed: 12/30/2022]
Abstract
New benzoxazole derivatives containing 1,3,4-oxadiazole, 1,2,4-triazole or triazolothiadiazine rings were synthesized and screened for their in vitro antiproliferative activities against MCF-7 and MDA-MB-231 breast cancer cell lines using MTT assay. Doxorubicin, cisplatin and 2-(4-aminophenyl)benzothiazole (CJM 126) were used as references. The most active compounds 7a, 8d, 8e and 10c were screened for their antiproliferative activities against MCF-10A normal breast cells where compounds 8e and 7a were the most selective towards MCF-7 and MDA-MB-231 cell lines, respectively compared to CJM 126. In vitro enzymatic inhibition assays of epidermal growth factor receptor (EGFR) and aromatase (ARO) enzymes were performed. Compound 7a showed inhibition of EGFR comparable to that of erlotinib while compound 8e exhibited nearly half the inhibitory activity of erlotinib towards EGFR and was more potent inhibitor of ARO than letrozole. Caspase-9 activation assay, cell cycle analysis and Annexin-V/ Propidium iodide assay performed for compounds 7a, 8d, 8e and 10c demonstrated over expression of caspase-9 protein level, pre G1 apoptosis and high annexin V binding affinity. Therefore, these compounds are considered as potent apoptosis-promoting agents. The predicted docking studies and in silico chemo-informatic properties of compounds 7a and 8e were appropriate. Compounds 7a and 8e are promising anti-breast cancer agents exhibiting potent apoptosis-promoting properties.
Collapse
|
28
|
Zhao Z, Bourne PE. Structural Insights into the Binding Modes of Viral RNA-Dependent RNA Polymerases Using a Function-Site Interaction Fingerprint Method for RNA Virus Drug Discovery. J Proteome Res 2020; 19:4698-4705. [PMID: 32946692 PMCID: PMC7640976 DOI: 10.1021/acs.jproteome.0c00623] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Indexed: 01/18/2023]
Abstract
The coronavirus disease of 2019 (COVID-19) pandemic speaks to the need for drugs that not only are effective but also remain effective given the mutation rate of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). To this end, we describe structural binding-site insights for facilitating COVID-19 drug design when targeting RNA-dependent RNA polymerase (RDRP), a common conserved component of RNA viruses. We combined an RDRP structure data set, including 384 RDRP PDB structures and all corresponding RDRP-ligand interaction fingerprints, thereby revealing the structural characteristics of the active sites for application to RDRP-targeted drug discovery. Specifically, we revealed the intrinsic ligand-binding modes and associated RDRP structural characteristics. Four types of binding modes with corresponding binding pockets were determined, suggesting two major subpockets available for drug discovery. We screened a drug data set of 7894 compounds against these binding pockets and presented the top-10 small molecules as a starting point in further exploring the prevention of virus replication. In summary, the binding characteristics determined here help rationalize RDRP-targeted drug discovery and provide insights into the specific binding mechanisms important for containing the SARS-CoV-2 virus.
Collapse
Affiliation(s)
- Zheng Zhao
- School
of Data Science, University of Virginia, Charlottesville, Virginia 22904, United States of America
- Department
of Biomedical Engineering, University of
Virginia, Charlottesville, Virginia 22904, United States of America
| | - Philip E. Bourne
- School
of Data Science, University of Virginia, Charlottesville, Virginia 22904, United States of America
- Department
of Biomedical Engineering, University of
Virginia, Charlottesville, Virginia 22904, United States of America
| |
Collapse
|
29
|
Zhao Z, Bourne PE. Revealing Acquired Resistance Mechanisms of Kinase-Targeted Drugs Using an on-the-Fly, Function-Site Interaction Fingerprint Approach. J Chem Theory Comput 2020; 16:3152-3161. [PMID: 32283024 DOI: 10.1021/acs.jctc.9b01134] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Although kinase-targeted drugs have achieved significant clinical success, they are frequently subject to the limitations of drug resistance, which has become a primary vulnerability to targeted drug therapy. Therefore, deciphering resistance mechanisms is an important step in designing more efficacious, antiresistant drugs. Here we studied two FDA-approved kinase drugs: Crizotinib and Ceritinib, which are first- and second-generation anaplastic lymphoma kinase (ALK) targeted inhibitors, to unravel drug-resistance mechanisms. We used an on-the-fly, function-site interaction fingerprint (on-the-fly Fs-IFP) approach, combining binding free-energy surface calculations with the Fs-IFPs. Establishing the potentials of mean force and monitoring the atomic-scale protein-ligand interactions, before and after L1196M-induced drug resistance, revealed insights into drug-resistance/antiresistant mechanisms. Crizotinib prefers to bind the wild-type ALK kinase domain, whereas Ceritinib binds more favorably to the mutated ALK kinase domain, in agreement with experimental results. We determined that ALK kinase-drug interactions in the region of the front pocket are associated with drug resistance. Additionally, we find that the L1196M mutation does not simply alter the binding modes of inhibitors but also affects the flexibility of the entire ALK kinase domain. Our work provides an understanding of the mechanisms of ALK drug resistance, confirms the usefulness of the on-the-fly Fs-IFP approach, and provides a practical paradigm to study drug-resistance mechanisms in prospective drug discovery.
Collapse
|
30
|
Schmalhorst PS, Bergner A. A Grid Map Based Approach to Identify Nonobvious Ligand Design Opportunities in 3D Protein Structure Ensembles. J Chem Inf Model 2020; 60:2178-2188. [DOI: 10.1021/acs.jcim.0c00051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Philipp S. Schmalhorst
- Department of Medicinal Chemistry, Boehringer Ingelheim RCV GmbH & Co KG, Dr.-Boehringer-Gasse 5-11, 1121 Vienna, Austria
| | - Andreas Bergner
- Department of Medicinal Chemistry, Boehringer Ingelheim RCV GmbH & Co KG, Dr.-Boehringer-Gasse 5-11, 1121 Vienna, Austria
| |
Collapse
|
31
|
Large-Scale Virtual Screening Against the MET Kinase Domain Identifies a New Putative Inhibitor Type. Molecules 2020; 25:molecules25040938. [PMID: 32093126 PMCID: PMC7070486 DOI: 10.3390/molecules25040938] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 02/13/2020] [Accepted: 02/14/2020] [Indexed: 12/13/2022] Open
Abstract
By using an ensemble-docking strategy, we undertook a large-scale virtual screening campaign in order to identify new putative hits against the MET kinase target. Following a large molecular dynamics sampling of its conformational space, a set of 45 conformers of the kinase was retained as docking targets to take into account the flexibility of the binding site moieties. Our screening funnel started from about 80,000 chemical compounds to be tested in silico for their potential affinities towards the kinase binding site. The top 100 molecules selected—thanks to the molecular docking results—were further analyzed for their interactions, and 25 of the most promising ligands were tested for their ability to inhibit MET activity in cells. F0514-4011 compound was the most efficient and impaired this scattering response to HGF (Hepatocyte Growth Factor) with an IC50 of 7.2 μM. Interestingly, careful docking analysis of this molecule with MET suggests a possible conformation halfway between classical type-I and type-II MET inhibitors, with an additional region of interaction. This compound could therefore be an innovative seed to be repositioned from its initial antiviral purpose towards the field of MET inhibitors. Altogether, these results validate our ensemble docking strategy as a cost-effective functional method for drug development.
Collapse
|
32
|
Kharkar PS. Computational Approaches for the Design of (Mutant-)Selective Tyrosine Kinase Inhibitors: State-of-the-Art and Future Prospects. Curr Top Med Chem 2020; 20:1564-1575. [PMID: 32357816 DOI: 10.2174/1568026620666200502005853] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/10/2020] [Accepted: 03/26/2020] [Indexed: 02/08/2023]
Abstract
Kinases remain one of the major attractive therapeutic targets for a large number of indications such as cancer, rheumatoid arthritis, cardiac failure and many others. Design and development of kinase inhibitors (ATP-competitive, allosteric or covalent) is a clinically validated and successful strategy in the pharmaceutical industry. The perks come with limitations, particularly the development of resistance to highly potent and selective inhibitors. When this happens, the cycle needs to be repeated, i.e., the design and development of kinase inhibitors active against the mutated forms. The complexity of tumor milieu makes it awfully difficult for these molecularly-targeted therapies to work. Every year newer and better versions of these agents are introduced in the clinic. Several computational approaches such as structure-, ligand-based or hybrid ones continue to live up to their potential in discovering novel kinase inhibitors. New schools of thought in this area continue to emerge, e.g., development of dual-target kinase inhibitors. But there are fundamental issues with this approach. It is indeed difficult to selectively optimize binding at two entirely different or related kinases. In addition to the conventional strategies, modern technologies (machine learning, deep learning, artificial intelligence, etc.) started yielding the results and building success stories. Computational tools invariably played a critical role in catalysing the phenomenal progress in kinase drug discovery field. The present review summarized the progress in utilizing computational methods and tools for discovering (mutant-)selective tyrosine kinase inhibitor drugs in the last three years (2017-2019). Representative investigations have been discussed, while others are merely listed. The author believes that the enthusiastic reader will be inspired to dig out the cited literature extensively to appreciate the progress made so far and the future prospects of the field.
Collapse
Affiliation(s)
- Prashant S Kharkar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Nathalal Parekh Marg, Matunga, Mumbai 400 019, India
| |
Collapse
|