Next Article in Journal
Association of Vitamin D with Perfluorinated Alkyl Acids in Women with and without Non-Obese Polycystic Ovary Syndrome
Previous Article in Journal
SIMOA Diagnostics on Alzheimer’s Disease and Frontotemporal Dementia
Previous Article in Special Issue
Tanshinone IIA Facilitates Efficient Cartilage Regeneration under Inflammatory Factors Caused Stress via Upregulating LncRNA NEAT1_2
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Recent Advances in the Discovery of Novel Drugs on Natural Molecules

by
Laura Quintieri
1,*,
Leonardo Caputo
1 and
Orazio Nicolotti
2
1
Institute of Sciences of Food Production, National Research Council (CNR), Via G. Amendola, 122/O, 70126 Bari, Italy
2
Dipartimento di Farmacia—Scienze del Farmaco, Università degli Studi di Bari “Aldo Moro”, Via E. Orabona, 4, 70125 Bari, Italy
*
Author to whom correspondence should be addressed.
Biomedicines 2024, 12(6), 1254; https://doi.org/10.3390/biomedicines12061254
Submission received: 6 May 2024 / Accepted: 22 May 2024 / Published: 5 June 2024
(This article belongs to the Special Issue Recent Advances in the Discovery of Novel Drugs on Natural Molecules)
Natural products (NPs) are always a promising source of novel drugs for tackling unsolved diseases [1,2,3]. Natural molecules have addressed the rational design of many synthetic small-molecule drugs [4,5,6], despite their chemotypes having higher structural complexity, heavier molecular weights (often > 500), more sp3 carbon atoms which amplify tridimensionality and stereocenters, more oxygen atoms, fewer nitrogen and halogen atoms, more H-bond acceptors and donors, marked hydrophilicity, and greater molecular rigidity [7]. Natural molecules are likely still inspiring drug discovery due to their incredible scaffold diversity, making them extremely unique and selective in the chemistry field [8]. Thus, developing new natural bioactive compounds and repurposing approved natural drugs are hot topics in drug discovery and medicinal chemistry [9]. Since most compounds exhibit synergistic effects [10] or share multiple targets [11,12], traditional approaches to finding potential drug candidates, such as bioassay-guided fractionation, can reduce their therapeutic efficacy. As a result, new techniques are required to produce drugs with high and multi-target activity as well as improved bioavailability [13,14,15]. For example, molecular biological techniques can increase the availability of novel compounds produced by bacteria or yeasts [16,17], and virtual screening approaches can generate screening libraries of natural compounds resembling drug-like compounds [18,19,20,21] or predict their biological activity and related molecular targets with chemical accuracy [22,23]. Moreover, advances in metabolomics have also allowed us to identify active compounds from natural product mixtures as well as to reveal synergistic effects in complex mixtures [24,25].
We gathered twenty articles for this Special Issue that discuss the discovery of novel bioactive NPs with potential for medical purposes. The application of synthetic biology with all its multidisciplinary aspects (bioinformatics, data mining, pathway refactoring, cell factories, DNA editing, and computational chemistry) was preferred to allow the identification of novel drug molecules from microbial strains or bioresources that might escape classical top-down strategies.
The plant kingdom is a significant source of molecules with potential therapeutic benefits for humans; bioactive molecules derived from plants often exhibit clear therapeutic profiles and can be employed as drugs or starting points to derive synthetic drugs [26,27,28,29]. Several recent publications on the biological activity of plant compounds are collected in this Special Issue.
In particular, Sun et al. [30] extracted and purified the lipophilic diterpene Tanshinone IIA (TAN) from Salvia miltiorrhiz Bunge and evaluated its role in maintaining chondrocyte viability and promoting cartilage regeneration in osteoarthritis patients. TAN was already recognized in herbal medicine for its anti-inflammatory, antioxidant, and vascular endothelial cell-protective properties. Likewise, Schwarz et al. [31] investigated the mode of action of the steroid sapogenin diosgenin, previously identified in the Chinese plant Dioscoreae rhizoma, in dampening the autoimmune inflammatory response in T helper 17 (Th17)-driven pathologies. By combining methodological approaches including gene expression analysis and in silico analyses, the authors revealed diosgenin as an inverse agonist of the key transcription factors leading to Th17 cell differentiation and metabolism. Kim and colleagues [32] demonstrated that the plant-derived ferulic acid acts as a therapeutic agent for wound healing via inhibiting β-catenin in keratinocytes and activating Nrf2 in wound-induced inflammation.
Florets of Safflower (Carthamus tinctorius) were identified as a source of polyacetylene glycosides, which are responsible for preventing excessive lipid accumulation in obesity through the inhibition of adipocyte differentiation, reducing the transcription levels of mature adipocyte marker genes (Adipsin and Fabp4), promoting the expression of lipolytic genes, and downregulating the expression of lipogenic genes [33].
The application of plant-derived compounds (such as polyphenols) in therapies is often hampered by several factors including structural instability; poor bioavailability, gastric solubility, and residence time; and fast metabolization in the liver [34,35,36]. Through the combination of experimental (spectroscopy and calorimetry) and simulation techniques (docking and molecular dynamics simulations) the glycosyl derivate of the flavonoid rutin (quercetin-3-O-rutinose) was found to exhibit comparable potency to the parental molecule rutin and an estimated higher bioavailability. Thus, the results of this work were proposed as the basis for the development of quercetin-like antiviral compounds in coronavirus infection management [37].
Like quercetin, the alkaloid antitumoral camptothecin (CPT) shows weak pharmacokinetic and pharmacodynamics properties [38]. Nanotechnology is perfect for improving CPT bioavailability; thus, the review by Ghanbari-Movahed et al. provided a comprehensive and critical evaluation of the novel, efficient nano-CPT formulations being developed for cancer therapy [38]. By contrast, biotransformation of the antioxidant resveratrol (RSV) by the entomopathogenic fungus Beauveria bassiana yielded a safer RSV metabolite, the stilbene glycoside resvebassianol A [39].
As widely reported, NPs can exert multiple biological activities. For example, the anti-inflammatory phenolic compound Apocynin, an inhibitor of NADPH-dependent oxidase (NOX), was suggested to interact with plasmalemmal ionic channels by perturbing ionic currents in excitable cells [40]. However, more evidence is needed to understand the molecular-level nature of interactions affecting neuroendocrine, endocrine, or cardiac function.
Food proteins from animals and plants are widely exploited for cryptic bioactive peptides exhibiting multi-target activities [41,42,43,44,45,46,47,48,49,50]. Gambacorta et al. [51] evaluated the inhibitory activity of the whey-derived bioactive small peptides MHI, IAEK, and IPAVF against the SARS-CoV-2 3C-like protease (3CLpro) for the first time. These peptides were previously obtained by the enzymatic hydrolysis of whey proteins and displayed ACE-inhibitory activity [22]. The authors integrated theoretical and experimental techniques, first performing molecular docking studies to rationally evaluate the putative chance of binding and then in vitro testing for validation. The results confirmed the highest antiviral activity for IPAVF and IAEK, providing new opportunities for the development of dual-target small peptides endowed with antiviral 3CLpro- and ACE-inhibitory activities.
Using a machine learning approach, Casey et al. [52] predicted five novel anti-diabetic peptides (pep_1E99R5, pep_37MB3O, pep_ANUT7B, pep_RTE62G and pep_QT5XGQ) from a set of 109 peptides. Although further work is required to elucidate their bioavailability, mechanism of action, and clinical efficacy, the authors presented pep_1E99R5 as the most active peptide, affecting blood glucose metabolism. Bioactive peptide sequences can also be re-designed to obtain novel drugs in cancer therapy, as reported by [53,54,55]. Thus, an in silico peptide design optimization process was applied to identify active peptides from the C-terminal of azurin, an anticancer bacterial protein produced by Pseudomonas aeruginosa. Due to its molecular properties, CT-p19LC was predicted to exhibit the greatest anticancer activity. This was confirmed in experimental trials and, therefore, it was suggested for the development of novel anticancer strategies.
Multidisciplinary approaches effectively reveal the synthesis pathways of NPs [56,57] or discover new molecules that might be used as templates to develop novel biotherapeutics [58,59].
Rugen et al. [60] collected venom from the assassin bug Rhynocoris iracundus and investigated its composition and bioactivity in vitro and in vivo to exploit it for biomedical applications. Assassin bug venom induced neurolysis, caused the paralysis and melanization of Galleria mellonella larvae and pupae, and exhibited antibacterial activity. The combined proteo-transcriptomic approach could successfully identify molecules responsible for biological effects (redulysins, kininogens, chitinases, hemolysins, and Ptu1 family peptide toxins).
Among 35 phytochemicals, sennoside B from Cassia angustifolia was predicted as a TNF-α inhibitor by a competitive binding screening assay coupled with analytical size exclusion chromatography and liquid chromatography–tandem mass spectrometry (LC-MS). Molecular docking was also performed to determine the binding mode of sennoside B to TNF-α, confirming its activity in TNF-α-induced HeLa cell toxicity assays [61]. Similarly, molecular docking revealed that flavonoids (apigenin and luteolin) bound to histone deacetylases (HDACs), with important implications in epigenetic therapy to regulate cellular gene expression [62]. In addition, computational methods were applied to design modified flavonoids endowed with high monoamine oxidase (MAO) B affinity for neurological disorder treatment [63,64], as well as to identify new potential scaffolds against Cyclin-dependent kinase 7 (CDK7) for the development of novel antitumoral strategies [65].
The bioactive compounds found in natural products are also a valuable source of inspiration for new drug synthesis [14,66,67,68], such as the marine-inspired potent kinase inhibitors with antiproliferative activities described by [69]. NPs can also be used to chemically modify the molecular structure of existing drugs to improve their activity or pharmacokinetics properties. For example, Neganova et al. [70] found that the conjugation of sesquiterpene lactones, extracted from Inula helenium L. (Asteraceae), reduced the side effects of antitumoral canthracycline antibiotics.
In conclusion, the articles published in this Special Issue underline the advances and opportunities in using NPs in drug discovery. Several works show NPs’ key role in a wide array of biological activities, such as maintaining tissue integrity, regulating immune responses, and influencing complex processes in human diseases. Their current limitations and promising strategies to design and identify novel molecules are also discussed.

Author Contributions

Conceptualization, L.Q., L.C. and O.N.; writing—original draft preparation, L.Q.; writing—review and editing, L.C. and O.N. All authors have read and agreed to the published version of the manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Atanasov, A.G.; Zotchev, S.B.; Dirsch, V.M.; Supuran, C. Natural products in drug discovery: Advances and opportunities. Nat. Rev. Drug Discov. 2021, 20, 200–216. [Google Scholar] [CrossRef] [PubMed]
  2. Berdigaliyev, N.; Aljofan, M. An overview of drug discovery and development. Future Med. Chem. 2020, 12, 939–947. [Google Scholar] [CrossRef] [PubMed]
  3. Chopra, B.; Ashwani, K.D. Natural products: A lead for drug discovery and development. Phytother. Res. 2021, 35, 4660–4702. [Google Scholar] [CrossRef] [PubMed]
  4. Gambacorta, N.; Gasperi, V.; Guzzo, T.; Di Leva, F.S.; Ciriaco, F.; Sánchez, C.; Tullio, V.; Rozzi, D.; Marinelli, L.; Topai, A.; et al. Exploring the 1,3-Benzoxazine Chemotype for Cannabinoid Receptor 2 as a Promising Anti-Cancer Therapeutic. Eur. J. Med. Chem. 2023, 259, 115647. [Google Scholar] [CrossRef] [PubMed]
  5. Mangiatordi, G.F.; Trisciuzzi, D.; Alberga, D.; Denora, N.; Iacobazzi, R.M.; Gadaleta, D.; Catto, M.; Nicolotti, O. Novel chemotypes targeting tubulin at the Colchicine binding site and unbiasing P-glycoprotein. Eur. J. Med. Chem. 2017, 139, 792–803. [Google Scholar] [CrossRef] [PubMed]
  6. Ma, Y.S.; Xin, R.; Yang, X.L.; Shi, Y.; Zhang, D.D.; Wang, H.M.; Wang, P.Y.; Liu, J.B.; Chu, K.J.; Fu, D. Paving the way for small-molecule drug discovery. Am. J. Transl. Res. 2021, 13, 853–870. [Google Scholar] [PubMed]
  7. Dzobo, K. The role of natural products as sources of therapeutic agents for innovative drug discovery. Compr. Pharmacol. 2022, 408–422. [Google Scholar] [CrossRef]
  8. Amoroso, N.; Gambacorta, N.; Mastrolorito, F.; Togo, M.V.; Trisciuzzi, D.; Monaco, A.; Pantaleo, E.; Altomare, C.D.; Ciriaco, F.; Nicolotti, O. Making sense of chemical space network shows signs of criticality. Sci. Rep. 2023, 13, 21335. [Google Scholar] [CrossRef]
  9. Ciriaco, F.; Gambacorta, N.; Trisciuzzi, D.; Nicolotti, O. PLATO: A Predictive Drug Discovery Web Platform for Efficient Target Fishing and Bioactivity Profiling of Small Molecules. Int. J. Mol. Sci. 2022, 23, 5245. [Google Scholar] [CrossRef]
  10. Zou, H.; Ye, H.; Kamaraj, R.; Zhang, T.; Zhang, J.; Pavek, P. A review on pharmacological activities and synergistic effect of quercetin with small molecule agents. Phytomedicine 2021, 92, 153736. [Google Scholar] [CrossRef]
  11. Jha, A.K.; Sit, N. Extraction of bioactive compounds from plant materials using combination of various novel methods: A review. Trends Food Sci. Technol. 2022, 119, 579–591. [Google Scholar] [CrossRef]
  12. Chemat, F.; Vian, M.A.; Fabiano-Tixier, A.S.; Nutrizio, M.; Jambrak, A.R.; Munekata, P.E.; Lorenzo, J.M.; Barba, F.J.; Binello, A.; Cravotto, G. A review of sustainable and intensified techniques for extraction of food and natural products. Green Chem. 2020, 22, 2325–2353. [Google Scholar] [CrossRef]
  13. Gambacorta, N.; Ciriaco, F.; Amoroso, N.; Altomare, C.D.; Bajorath, J.; Nicolotti, O. CIRCE: Web-Based Platform for the Prediction of Cannabinoid Receptor Ligands Using Explainable Machine Learning. J. Chem. Inf. Model. 2023, 63, 5916–5926. [Google Scholar] [CrossRef]
  14. Najmi, A.; Javed, S.A.; Al Bratty, M.; Alhazmi, H.A. Modern approaches in the discovery and development of plant-based natural products and their analogues as potential therapeutic agents. Molecules 2022, 27, 349. [Google Scholar] [CrossRef] [PubMed]
  15. Zhu, Y.; Ouyang, Z.; Du, H.; Wang, M.; Wang, J.; Sun, H.; Kong, L.; Xu, Q.; Hongyue, M.; Sun, Y. New opportunities and challenges of natural products research: When target identification meets single-cell multiomics. Acta Pharm. Sin. B 2022, 12, 4011–4039. [Google Scholar] [CrossRef] [PubMed]
  16. Losurdo, L.; Quintieri, L.; Caputo, L.; Gallerani, R.; Mayo, B.; De Leo, F. Cloning and expression of synthetic genes encoding angiotensin-I converting enzyme (ACE)-inhibitory bioactive peptides in Bifidobacterium pseudocatenulatum. FEMS Microbiol. Lett. 2013, 340, 24–32. [Google Scholar] [CrossRef] [PubMed]
  17. Yang, D.; Park, S.Y.; Park, Y.S.; Eun, H.; Lee, S.Y. Metabolic engineering of Escherichia coli for natural product biosynthesis. Trends Biotechnol. 2020, 38, 745–765. [Google Scholar] [CrossRef]
  18. Sadybekov, A.V.; Katritch, V. Computational approaches streamlining drug discovery. Nature 2023, 616, 673–685. [Google Scholar] [CrossRef] [PubMed]
  19. Vázquez, J.; López, M.; Gibert, E.; Herrero, E.; Luque, F.J. Merging Ligand-Based and Structure-Based Methods in Drug Discovery: An Overview of Combined Virtual Screening Approaches. Molecules 2020, 25, 4723. [Google Scholar] [CrossRef]
  20. Oliveira, T.A.D.; Silva, M.P.D.; Maia, E.H.B.; Silva, A.M.D.; Taranto, A.G. Virtual screening algorithms in drug discovery: A review focused on machine and deep learning methods. Drugs Drug Candidates 2023, 2, 311–334. [Google Scholar] [CrossRef]
  21. Parvatikar, P.P.; Patil, S.; Khaparkhuntikar, K.; Patil, S.; Singh, P.K.; Sahana, R.; Kulkarni, R.V.; Raghu, A.V. Artificial intelligence: Machine learning approach for screening large database and drug discovery. Antivir. Res. 2023, 220, 105740. [Google Scholar] [CrossRef]
  22. Tondo, A.R.; Caputo, L.; Mangiatordi, G.F.; Monaci, L.; Lentini, G.; Logrieco, A.F.; Montaruli, M.; Nicolotti, O.; Quintieri, L. Structure-Based Identification and Design of Angiotensin Converting Enzyme-Inhibitory Peptides from Whey Proteins. J. Agric. Food Chem. 2020, 68, 541–548. [Google Scholar] [CrossRef] [PubMed]
  23. Gu, R.; Wu, F.; Huang, Z. Role of Computer-Aided Drug Design in Drug Development. Molecules 2023, 28, 7160. [Google Scholar] [CrossRef] [PubMed]
  24. Vidar, W.S.; Baumeister, T.U.H.; Caesar, L.K.; Kellogg, J.J.; Todd, D.A.; Linington, R.G.M.; Kvalheim, O.; Cech, N.B. Interaction Metabolomics to Discover Synergists in Natural Product Mixtures. J. Nat. Prod. 2023, 86, 655–671. [Google Scholar] [CrossRef] [PubMed]
  25. Palermo, A. Metabolomics-and systems-biology-guided discovery of metabolite lead compounds and druggable targets. Drug Discov. Today 2023, 28, 103460. [Google Scholar] [CrossRef] [PubMed]
  26. Chaachouay, N.; Zidane, L. Plant-Derived Natural Products: A Source for Drug Discovery and Development. Drugs Drug Candidates 2024, 3, 184–207. [Google Scholar] [CrossRef]
  27. Elshafie, H.S.; Camele, I.; Mohamed, A.A. A Comprehensive review on the biological, agricultural and pharmaceutical properties of secondary metabolites based-plant origin. Int. J. Mol. Sci. 2023, 24, 3266. [Google Scholar] [CrossRef] [PubMed]
  28. Lautie, E.; Russo, O.; Ducrot, P.; Boutin, J.A. Unraveling plant natural chemical diversity for drug discovery purposes. Front. Pharmacol. 2020, 11, 397. [Google Scholar] [CrossRef] [PubMed]
  29. Dias, M.C.; Pinto, D.C.; Silva, A.M. Plant flavonoids: Chemical characteristics and biological activity. Molecules 2021, 26, 5377. [Google Scholar] [CrossRef] [PubMed]
  30. Sun, J.; Chen, W.; Zhou, Z.; Chen, X.; Zuo, Y.; He, J.; Liu, H. Tanshinone IIA Facilitates Efficient Cartilage Regeneration under Inflammatory Factors Caused Stress via Upregulating LncRNA EAT1_2. Biomedicines 2023, 11, 3291. [Google Scholar] [CrossRef]
  31. Schwarz, P.F.; Perhal, A.F.; Schöberl, L.N.; Kraus, M.M.; Kirchmair, J.; Dirsch, V.M. Identification of the Natural Steroid Sapogenin Diosgenin as a Direct Dual-Specific RORα/γ Inverse Agonist. Biomedicines 2022, 10, 2076. [Google Scholar] [CrossRef]
  32. Kim, K.-H.; Jung, J.H.; Chung, W.-S.; Lee, C.-H.; Jang, H.-J. Ferulic Acid Induces Keratin 6α via Inhibition of Nuclear β-Catenin Accumulation and Activation of Nrf2 in Wound-Induced Inflammation. Biomedicines 2021, 9, 459. [Google Scholar] [CrossRef]
  33. Baek, S.C.; Yi, S.A.; Lee, B.S.; Yu, J.S.; Kim, J.-C.; Pang, C.; Jang, T.S.; Lee, J.; Kim, K.H. Anti-Adipogenic Polyacetylene Glycosides from the Florets of Safflower (Carthamus tinctorius). Biomedicines 2021, 9, 91. [Google Scholar] [CrossRef]
  34. Albuquerque, B.R.; Heleno, S.A.; Oliveira, M.B.P.; Barros, L.; Ferreira, I.C. Phenolic compounds: Current industrial applications, limitations and future challenges. Food Funct. 2021, 12, 14–29. [Google Scholar] [CrossRef]
  35. Zhang, Z.; Li, X.; Sang, S.; McClements, D.J.; Chen, L.; Long, J.; Jiao, A.; Jin, Z.; Qiu, C. Polyphenols as Plant-Based Nutraceuticals: Health Effects, Encapsulation, Nano-Delivery, and Application. Foods 2022, 11, 2189. [Google Scholar] [CrossRef]
  36. Bertelli, A.; Biagi, M.; Corsini, M.; Baini, G.; Cappellucci, G.; Miraldi, E. Polyphenols: From Theory to Practice. Foods 2021, 10, 2595. [Google Scholar] [CrossRef]
  37. Rizzuti, B.; Grande, F.; Conforti, F.; Jimenez-Alesanco, A.; Ceballos-Laita, L.; Ortega-Alarcon, D.; Vega, S.; Reyburn, H.T.; Abian, O.; Velazquez-Campoy, A. Rutin Is a Low Micromolar Inhibitor of SARS-CoV-2 Main Protease 3CLpro: Implications for Drug Design of Quercetin Analogs. Biomedicines 2021, 9, 375. [Google Scholar] [CrossRef]
  38. Ghanbari-Movahed, M.; Kaceli, T.; Mondal, A.; Farzaei, M.H.; Bishayee, A. Recent Advances in Improved Anticancer Efficacies of Camptothecin Nano-Formulations: A Systematic Review. Biomedicines 2021, 9, 480. [Google Scholar] [CrossRef]
  39. Ha, S.K.; Kang, M.C.; Lee, S.; Darlami, O.; Shin, D.; Choi, I.; Kim, K.H.; Kim, S.Y. Generation of Stilbene Glycoside with Promising Cell Rejuvenation Activity through Biotransformation by the Entomopathogenic Fungus Beauveria bassiana. Biomedicines 2021, 9, 555. [Google Scholar] [CrossRef]
  40. Chuang, T.-H.; Cho, H.-Y.; Wu, S.-N. Effective Accentuation of Voltage-Gated Sodium Current Caused by Apocynin (4′-Hydroxy-3′-methoxyacetophenone), a Known NADPH-Oxidase Inhibitor. Biomedicines 2021, 9, 1146. [Google Scholar] [CrossRef]
  41. Sharma, K.; Sharma, K.K.; Sharma, A.; Jain, R. Peptide-based drug discovery: Current status and recent advances. Drug Discov. Today 2023, 28, 103464. [Google Scholar] [CrossRef] [PubMed]
  42. Muttenthaler, M.; King, G.F.; Adams, D.J.; Alewood, P.F. Trends in peptide drug discovery. Nat. Rev. Drug Discov. 2021, 20, 309–325. [Google Scholar] [CrossRef] [PubMed]
  43. Trisciuzzi, D.; Villoutreix, B.O.; Siragusa, L.; Baroni, M.; Cruciani, G.; Nicolotti, O. Targeting protein-protein interactions with low molecular weight and short peptide modulators: Insights on disease pathways and starting points for drug discovery. Expert Opin. Drug Discov. 2023, 18, 737–752. [Google Scholar] [CrossRef] [PubMed]
  44. Trisciuzzi, D.; Siragusa, L.; Baroni, M.; Cruciani, G.; Nicolotti, O. An Integrated Machine Learning Model To Spot Peptide Binding Pockets in 3D Protein Screening. Chem. Inf. Model. 2022, 62, 6812–6824. [Google Scholar] [CrossRef]
  45. Trisciuzzi, D.; Siragusa, L.; Baroni, M.; Autiero, I.; Nicolotti, O.; Cruciani, G. Getting insights into structural and energetic properties of reciprocal peptide-protein interactions. J. Chem. Inf. Model. 2022, 62, 1113–1125. [Google Scholar] [CrossRef] [PubMed]
  46. Quintieri, L.; Caputo, L.; Monaci, L.; Cavalluzzi, M.M.; Denora, N. Lactoferrin-Derived Peptides as a Control Strategy against Skinborne Staphylococcal Biofilms. Biomedicines 2020, 8, 323. [Google Scholar] [CrossRef] [PubMed]
  47. Akbarian, M.; Khani, A.; Eghbalpour, S.; Uversky, V.N. Bioactive peptides: Synthesis, sources, applications, and proposed mechanisms of action. Int. J. Mol. Sci. 2022, 23, 1445. [Google Scholar] [CrossRef] [PubMed]
  48. Quintieri, L.; Fanelli, F.; Monaci, L.; Fusco, V. Milk and Its Derivatives as Sources of Components and Microorganisms with Health-Promoting Properties: Probiotics and Bioactive Peptides. Foods 2024, 13, 601. [Google Scholar] [CrossRef] [PubMed]
  49. Quintieri, L.; Nitride, C.; De Angelis, E.; Lamonaca, A.; Pilolli, R.; Russo, F.; Monaci, L. Alternative Protein Sources and Novel Foods: Benefits, Food Applications and Safety Issues. Nutrients 2023, 15, 1509. [Google Scholar] [CrossRef]
  50. Quintieri, L.; Zühlke, D.; Fanelli, F.; Caputo, L.; Liuzzi, V.C.; Logrieco, A.F.; Hirschfeld, C.; Becher, D.; Riedel, K. Proteomic analysis of the food spoiler Pseudomonas fluorescens ITEM 17298 reveals the antibiofilm activity of the pepsin-digested bovine lactoferrin. Food Microbiol. 2019, 82, 177–193. [Google Scholar] [CrossRef]
  51. Gambacorta, N.; Caputo, L.; Quintieri, L.; Monaci, L.; Ciriaco, F.; Nicolotti, O. Rational Discovery of Antiviral Whey Protein-Derived Small Peptides Targeting the SARS-CoV-2 Main Protease. Biomedicines 2022, 10, 1067. [Google Scholar] [CrossRef] [PubMed]
  52. Casey, R.; Adelfio, A.; Connolly, M.; Wall, A.; Holyer, I.; Khaldi, N. Discovery through Machine Learning and Preclinical Validation of Novel Anti-Diabetic Peptides. Biomedicines 2021, 9, 276. [Google Scholar] [CrossRef] [PubMed]
  53. Garizo, A.R.; Coelho, L.F.; Pinto, S.; Dias, T.P.; Fernandes, F.; Bernardes, N.; Fialho, A.M. The Azurin-Derived Peptide CT-p19LC Exhibits Membrane-Active Properties and Induces Cancer Cell Death. Biomedicines 2021, 9, 1194. [Google Scholar] [CrossRef] [PubMed]
  54. Jia, F.; Yu, Q.; Wang, R.; Zhao, L.; Yuan, F.; Guo, H.; Shen, Y.; He, F. Optimized antimicrobial peptide jelleine-I derivative Br-JI inhibits fusobacterium nucleatum to suppress colorectal cancer progression. Int. J. Mol. Sci. 2023, 24, 1469. [Google Scholar] [CrossRef] [PubMed]
  55. Moustafa, G.O.; Shalaby, A.; Naglah, A.M.; Mounier, M.M.; El-Sayed, H.; Anwar, M.M.; Nossier, E.S. Synthesis, Characterization, In Vitro Anticancer Potentiality, and Antimicrobial Activities of Novel Peptide–Glycyrrhetinic-Acid-Based Derivatives. Molecules 2021, 26, 4573. [Google Scholar] [CrossRef] [PubMed]
  56. Rattinam, R.; Basha, R.S.; Wang, Y.-L.; Wang, Z.-C.; Hsu, N.-S.; Lin, K.-H.; Zadeh, S.M.; Adhikari, K.; Lin, J.-P.; Li, T.-L. KasQ an Epimerase Primes the Biosynthesis of Aminoglycoside Antibiotic Kasugamycin and KasF/H Acetyltransferases Inactivate Its Activity. Biomedicines 2022, 10, 212. [Google Scholar] [CrossRef] [PubMed]
  57. Hou, S.; Du, W.; Hao, Y.; Han, Y.; Li, H.; Liu, L.; Zhang, K.; Zhou, M.; Sun, Z. Elucidation of the regulatory network of flavonoid biosynthesis by profiling the metabolome and transcriptome in tartary buckwheat. J. Agric. Food Chem. 2021, 69, 7218–7229. [Google Scholar] [CrossRef] [PubMed]
  58. Cheptea, C.; Sunel, V.; Morosanu, A.C.; Dimitriu, D.G.; Dulcescu-Oprea, M.M.; Angheluta, M.-D.; Miron, M.; Nechifor, C.D.; Dorohoi, D.O.; Malancus, R.N. Optimized Synthesis of New N-Mustards Based on 2-Mercaptobenzoxazole Derivatives with Antitumor Activity. Biomedicines 2021, 9, 476. [Google Scholar] [CrossRef] [PubMed]
  59. Mancini, I.; Vigna, J.; Sighel, D.; Defant, A. Hybrid Molecules Containing Naphthoquinone and Quinolinedione Scaffolds as Antineoplastic Agents. Molecules 2022, 27, 4948. [Google Scholar] [CrossRef]
  60. Rügen, N.; Jenkins, T.P.; Wielsch, N.; Vogel, H.; Hempel, B.-F.; Süssmuth, R.D.; Ainsworth, S.; Cabezas-Cruz, A.; Vilcinskas, A.; Tonk, M. Hexapod Assassins’ Potion: Venom Composition and Bioactivity from the Eurasian Assassin Bug Rhynocoris iracundus. Biomedicines 2021, 9, 819. [Google Scholar] [CrossRef]
  61. Peng, L.; Durai, P.; Park, K.; Pyo, J.J.; Choi, Y. A Novel Competitive Binding Screening Assay Reveals Sennoside B as a Potent Natural Product Inhibitor of TNF-α. Biomedicines 2021, 9, 1250. [Google Scholar] [CrossRef] [PubMed]
  62. Scafuri, B.; Bontempo, P.; Altucci, L.; De Masi, L.; Facchiano, A. Molecular Docking Simulations on Histone Deacetylases (HDAC)-1 and -2 to Investigate the Flavone Binding. Biomedicines 2020, 8, 568. [Google Scholar] [CrossRef] [PubMed]
  63. Gogineni, V.; Nael, M.A.; Chaurasiya, N.D.; Elokely, K.M.; McCurdy, C.R.; Rimoldi, J.M.; Cutler, S.J.; Tekwani, B.L.; León, F. Computationally Assisted Lead Optimization of Novel Potent and Selective MAO-B Inhibitors. Biomedicines 2021, 9, 1304. [Google Scholar] [CrossRef] [PubMed]
  64. Larit, F.; Elokely, K.M.; Chaurasiya, N.D.; Benyahia, S.; Nael, M.A.; León, F.; Abu-Darwish, M.S.; Efferth, T.; Wang, Y.H.; Belouahem-Abed, D.; et al. Inhibition of human monoamine oxidase A and B by flavonoids isolated from two Algerian medicinal plants. Phytomedicine 2018, 40, 27–36. [Google Scholar] [CrossRef] [PubMed]
  65. Kumar, V.; Parate, S.; Thakur, G.; Lee, G.; Ro, H.-S.; Kim, Y.; Kim, H.J.; Kim, M.O.; Lee, K.W. Identification of CDK7 Inhibitors from Natural Sources Using Pharmacoinformatics and Molecular Dynamics Simulations. Biomedicines 2021, 9, 1197. [Google Scholar] [CrossRef] [PubMed]
  66. Barba-Ostria, C.; Carrera-Pacheco, S.E.; Gonzalez-Pastor, R.; Heredia-Moya, J.; Mayorga-Ramos, A.; Rodríguez-Pólit, C.; Zúñiga-Miranda, J.; Arias-Almeida, B.; Guamán, L.P. Evaluation of Biological Activity of Natural Compounds: Current Trends and Methods. Molecules 2022, 27, 4490. [Google Scholar] [CrossRef]
  67. Quintieri, L.; Mani, S.; Lentini, G.; Maisetta, G. Advances in the discovery of natural molecules and their analogues against microbial infection-related biofilms. Front. Microbiol. 2022, 13, 1092209. [Google Scholar] [CrossRef] [PubMed]
  68. Guo, Z. The modification of natural products for medical use. Acta Pharm. Sin. B 2017, 7, 119–136. [Google Scholar] [CrossRef] [PubMed]
  69. Elsherbeny, M.H.; Elkamhawy, A.; Nada, H.; Abdellattif, M.H.; Lee, K.; Roh, E.J. Development of New Meridianin/Leucettine-Derived Hybrid Small Molecules as Nanomolar Multi-Kinase Inhibitors with Antitumor Activity. Biomedicines 2021, 9, 1131. [Google Scholar] [CrossRef]
  70. Neganova, M.; Semakov, A.; Aleksandrova, Y.; Yandulova, E.; Pukhov, S.; Anikina, L.; Klochkov, S. N-Alkylation of Anthracycline Antibiotics by Natural Sesquiterpene Lactones as a Way to Obtain Antitumor Agents with Reduced Side Effects. Biomedicines 2021, 9, 547. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Quintieri, L.; Caputo, L.; Nicolotti, O. Recent Advances in the Discovery of Novel Drugs on Natural Molecules. Biomedicines 2024, 12, 1254. https://doi.org/10.3390/biomedicines12061254

AMA Style

Quintieri L, Caputo L, Nicolotti O. Recent Advances in the Discovery of Novel Drugs on Natural Molecules. Biomedicines. 2024; 12(6):1254. https://doi.org/10.3390/biomedicines12061254

Chicago/Turabian Style

Quintieri, Laura, Leonardo Caputo, and Orazio Nicolotti. 2024. "Recent Advances in the Discovery of Novel Drugs on Natural Molecules" Biomedicines 12, no. 6: 1254. https://doi.org/10.3390/biomedicines12061254

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop