Next Article in Journal
Uric Acid: A New Perspective for Exploring the Pathological Process of Anthracycline-Induced Cardiotoxicity
Previous Article in Journal
Integrated BSA-Seq and WGCNA Analyses Reveal Candidate Genes Associated with Winter Bud Dormancy Maintenance in Fruit Mulberry (Morus spp.)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Molecular Pharmacology at the Crossroads of Precision Medicine

by
Alina Crenguţa Nicolae
1,
Ion-Bogdan Dumitrescu
2,* and
Cristina Manuela Drăgoi
1
1
Department of Biochemistry, Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, RO-020956 Bucharest, Romania
2
Department of Physics and Informatics, Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, RO-020956 Bucharest, Romania
*
Author to whom correspondence should be addressed.
Curr. Issues Mol. Biol. 2026, 48(1), 39; https://doi.org/10.3390/cimb48010039 (registering DOI)
Submission received: 20 December 2025 / Accepted: 24 December 2025 / Published: 27 December 2025
(This article belongs to the Section Molecular Pharmacology)

1. Molecular Pharmacology at the Intersection of Fundamental Molecular Science and Innovative Drug Development

Molecular pharmacology sits at the nexus of fundamental molecular science and therapeutic innovation. This field has seen remarkable progress over recent years, driven by advances in our understanding of drug–target interactions, disease pathways, and human genetics. Landmark achievements in molecular pharmacology have reshaped drug development paradigms, perhaps most notably through the shift from empirical, one-size-fits-all treatments to mechanism-guided targeted therapies and precision medicine approaches [1,2,3,4,5,6,7,8]. For example, recent analyses indicate that the majority of novel drugs approved in recent years qualify as targeted therapies, underscoring a decisive move toward agents that specifically modulate defined molecular drivers of disease. In parallel, over one-third of new FDA approvals now fall under personalized medicine, treatments tailored to patients’ individual biomarker or genetic profiles [9,10,11,12,13,14]. These trends highlight a fundamental evolution: modern pharmacology strives for the “five rights” of therapy—the right patient, drug, dose, time, and route—by leveraging molecular insights [15]. This transformation has only been possible through decades of fundamental research that elucidated the complex biology of receptors, enzymes, and signaling pathways and revealed how genetic variations influence drug response. It has become increasingly clear that pharmacogenomic factors—an individual’s genetic makeup affecting drug metabolism and targets—are critical to optimizing efficacy and safety. Indeed, pharmacogenomics has emerged as a cornerstone of precision pharmacology, tailoring drug selection and dosing to patients’ genetic features [15]. International consortia have even begun issuing clinical guidelines for genotype-guided prescribing, though their adoption in practice is still ongoing. Taken together, these developments mark a new era in which molecular pharmacology is not only about understanding how drugs work, but also about who they work for and why [16,17,18,19,20,21,22,23].

2. Privileged Scaffolds and Rational Drug Design

Amid this paradigm shift, drug discovery strategies have been refined by incorporating medicinal chemistry innovations. One concept gaining renewed prominence is the use of privileged scaffolds—structural motifs known to engage a variety of biological targets with high affinity. These scaffolds often possess favorable drug-like properties (e.g., suitable size, polarity, and metabolic stability), making them attractive templates in designing new therapeutics [4,24,25,26,27,28,29]. By cleverly modifying such privileged frameworks, researchers can efficiently generate libraries of candidate compounds with a higher prior probability of biological activity. The payoff of this approach is evidenced by its success across multiple domains, perhaps most visibly in antiviral and anticancer research. Privileged structures display versatile binding properties and can provide potent, selective ligands for diverse targets, yielding high-quality lead compounds and accelerating the development of novel drugs [4]. A striking recent example comes from the battle against COVID-19: researchers synthesized thiazole–pyridine hybrid molecules—built on a known bioactive scaffold—and demonstrated their efficacy in inhibiting the SARS-CoV-2 main protease in preclinical assays [30,31,32,33]. Such molecules, designed with structural insight, represent promising antiviral leads and underscore how scaffold-based drug design can rapidly respond to emerging health threats. More generally, leveraging privileged scaffolds allows medicinal chemists to simultaneously explore multiple therapeutic areas, repurposing core chemical structures to address different diseases. This strategy, grounded in fundamental structure–activity knowledge, exemplifies how rational drug design is superseding brute-force screening: researchers now design molecules with target binding and pharmacokinetic properties in mind from the outset, rather than relying purely on trial-and-error [34,35,36,37,38].

3. New Approaches Expanding the Pharmacological Arsenal

Another landmark achievement in molecular pharmacology has been the advent of entirely new therapeutic modalities beyond traditional small-molecule inhibitors. One notable innovation is the development of PROTACs (proteolysis-targeting chimeras) and related targeted protein degradation technologies. Unlike conventional drugs, which simply inhibit protein function, PROTACs recruit unwanted proteins to the cell’s own degradation machinery, thereby selectively eliminating disease-driving proteins. These chimeras represent an advanced therapeutic strategy that enables selective protein degradation, opening new possibilities in drug design [39,40], especially toward tackling targets previously deemed “undruggable,” such as scaffolding or transcriptional proteins without obvious active sites. Early PROTAC successes in preclinical models of cancer, viral infection, and neurodegenerative disease hint at their vast potential [41,42], and the first PROTAC-based candidates have now entered clinical trials, validating targeted protein degradation as more than a theoretical concept [43,44]. In a broader sense, molecular pharmacology has embraced an expanding toolkit that includes monoclonal antibodies, recombinant proteins, gene and cell therapies, and mRNA-based therapeutics. Each of these modalities stems from fundamental discoveries in molecular biology—from understanding the immune checkpoint receptors, yielding antibody immunotherapies, to deciphering mRNA translation mechanisms, paving the way for mRNA vaccines. The integration of these new therapeutic classes has blurred the boundaries between traditional pharmacology and biotechnology, creating a more integrative “pharmaco-biology.” Yet, all these advances share a common thread: they exploit molecular-level understanding of disease processes. For instance, the success of mRNA vaccines for COVID-19 demonstrated how precisely delivering genetic instructions to cells can induce pharmacological effects with unprecedented speed and specificity. As we harness these new approaches, molecular pharmacologists are adapting principles of drug action to novel contexts—whether the “drug” is a small organic molecule, a biomacromolecule, or even a living cell, the goal remains to modulate molecular pathways in a predictable, beneficial manner [45,46,47,48].

4. Precision and Personalized Pharmacology

Perhaps the most transformative progress has come in aligning therapies with patient-specific molecular profiles—the era of precision medicine. Fundamental research into genomic and phenotypic heterogeneity has revealed why patients with the same diagnosis can respond very differently to a given drug, fueling efforts to identify biomarkers (genetic mutations, receptor expression levels, metabolic signatures, etc.) that predict drug response or toxicity. The result is a growing roster of tailored therapies guided by companion diagnostics: from EGFR inhibitors given only to lung cancer patients with EGFR mutations, to CFTR modulators effective in cystic fibrosis patients with specific CFTR gene variants. Such targeted therapies exemplify how molecular pharmacology translates basic insights into clinical benefit. As noted, a majority of new drug approvals are now biomarker-driven agents, a testament to how deeply personalization has permeated pharmacological research and development. Personalized medicine considers each patient’s unique genetic and molecular makeup, disease subtype, environment, and lifestyle to craft the optimal treatment strategy [15,49]. Beyond selecting the right drug, this includes individualizing the dose (pharmacogenetics has taught us, for example, how variants in drug-metabolizing enzymes like CYP450s demand dose adjustments) and even the timing of administration [18,50,51,52,53,54,55]. The benefits of this approach are manifold: higher efficacy, by targeting drugs to those most likely to benefit; fewer adverse effects, by avoiding treatment of those likely to be harmed or non-responsive; and often cost-effectiveness for healthcare systems in the long run. A vivid illustration of precision pharmacology in action is the dramatic improvement in outcomes for certain cancers—oncogene-targeted therapies have turned once-lethal malignancies into manageable conditions for many patients [56,57,58,59,60]. Likewise, the explosion of checkpoint inhibitor immunotherapies, while not “targeted” to a mutation per se, relies on molecular understanding of tumor immune evasion and often uses biomarkers (like PD-L1 expression or MSI status) to guide use. Crucially, precision pharmacology extends beyond oncology: from cardiology to psychiatry, researchers are identifying genetic markers and endotypes of disease that correlate with drug response [8,61,62,63,64,65]. These advances owe a great debt to fundamental research in molecular genetics, clinical pharmacology, and systems biology, which continues to unravel the sources of inter-individual variability in drug effects.

5. Natural Products and Novel Chemical Matter from Fundamental Insights

It is worth noting that even as cutting-edge technologies flourish, classical pharmacology—often inspired by nature’s chemical diversity—remains an invaluable font of new therapeutic leads. Many “privileged” natural product scaffolds are inherently bioactive and have co-evolved with biological systems, making them excellent starting points for drug discovery. Natural molecules such as alkaloids, flavonoids, terpenes, and peptides have long been medicines or prototypes for synthetic drugs [37,66,67,68,69,70,71,72]. Today, with advanced tools to analyze complex extracts and modify natural structures, researchers are revisiting natural sources for new pharmacophores [73]. A striking example is the realization that fruit peels and other agricultural by-products, often discarded in massive quantities, contain a wealth of bioactive compounds with medicinal potential. Recent investigations showed that, in many cases, fruit peels harbor higher concentrations of beneficial phytochemicals and pharmacologically active molecules than fruit pulp itself [74]. Orange peel extracts, for instance, are rich in polymethoxylated flavones with anti-inflammatory and cholesterol-lowering effects, and citrus-peel-derived compounds have even entered trials as anti-cancer and cardioprotective agents. Hussain et al. comprehensively reviewed fruit peel constituents and highlighted their antimicrobial, antioxidant, anti-diabetic, and neuroprotective activities, advocating the repurposing of “food waste” into a resource for drug discovery [74,75]. Such work exemplifies how fundamental phytochemical and pharmacological research can transform our therapeutic armamentarium while also promoting sustainability. Another area of renewed interest is the pharmacology of dietary compounds and nutraceuticals. For example, polyphenols found in foods and beverages are being studied for their complex effects on human health [76,77,78,79,80]. Resveratrol and catechins garnered attention for cardioprotective and anti-aging properties, and more recently, red wine polyphenols have undergone rigorous evaluation [81,82]. A comprehensive review cataloged the myriad bioactivities of red-wine-derived polyphenols—including anti-inflammatory, anti-carcinogenic, and cardioprotective effects—in models of cardiovascular disease, cancer, and metabolic and gut microbiome modulation [83,84]. However, as the authors and others acknowledge, these nutraceutical effects can be subtle and context-dependent; indeed, years of study have yielded controversy and no clear consensus on some claims, underscoring the need for further fundamental and clinical research to validate benefits. The enduring message is that natural product pharmacology continues to inspire modern drug discovery, and rigorous molecular investigations are critical to separating genuine therapeutic potential from myths. By applying cutting-edge analytical techniques and mechanistic assays, researchers are translating traditional knowledge and chemical biodiversity into new pharmacological opportunities, often discovering privileged structures in the process [85,86,87,88,89].

6. Future Perspectives

As we look to the future of molecular pharmacology, it is clear that the field will become ever more interdisciplinary and technology-driven. One exciting frontier is the integration of artificial intelligence and machine learning into drug discovery and pharmacokinetic/pharmacodynamic modeling. Computational tools are now routinely used to predict molecular properties, identify drug–target interactions, and optimize lead compounds in silico, dramatically accelerating early-stage research. For example, deep learning models trained on large chemical and bioactivity datasets can predict a compound’s bioactivity or toxicity with increasing accuracy, guiding chemists away from less promising candidates [90,91,92,93,94]. Moreover, machine learning is enhancing precision medicine by enabling the integration of multi-omics data (genomic, proteomic, metabolomic) with clinical information to forecast individual drug responses. We foresee that clinical pharmacology will increasingly incorporate algorithms that suggest optimal drugs or doses for a patient based on their molecular profile, a vision already taking shape in oncology and expected to expand to other fields. Another burgeoning area is the application of systems pharmacology and network models to understand polypharmacy and complex drug effects. By modeling the human body as an interconnected network of pathways, researchers aim to predict how modulating one target affects others—crucial for tackling multifactorial diseases like neurodegeneration, where combination therapies might be needed [95,96,97]. Fundamental research remains the engine driving these innovations, but the toolbox of molecular pharmacology is constantly growing—from CRISPR-based genome editors as potential therapeutics, to microbiome-targeting agents, to nanomedicines that deliver drugs with cellular precision [98,99,100]. Each new technology will bring its own pharmacological principles to elucidate. Encouragingly, the field has shown an admirable capacity to evolve and integrate new knowledge. The COVID-19 pandemic illustrated how quickly molecular pharmacologists and drug developers could respond, producing novel antivirals and vaccines in record time, guided by fundamental virology and immunology insights. This agility bodes well for addressing other pressing health challenges, from antibiotic resistance to rare genetic disorders [101,102].
In summary, molecular pharmacology has matured into a central pillar of modern biomedical science, linking bench-top discoveries to bedside cures. The past few years alone have seen the consolidation of precision medicine, the rise of new drug modalities, and the creative repurposing of natural scaffolds—all underpinned by deep molecular understanding. Our community continues to celebrate landmark successes, such as life-saving targeted therapies and paradigm-shifting vaccines, even as we remain vigilant about areas of uncertainty and opportunity. Looking ahead, the field is poised to advance on multiple fronts: deeper mechanistic insights through fundamental research will feed the pipeline of drug targets; improved computational and experimental tools will refine how we design and test drugs; and personalized strategies will ensure treatments are more effective and safer for each individual. The future of molecular pharmacology is therefore both promising and demanding: novel therapies for currently intractable diseases are on the horizon, but developing and deploying them will require continued interdisciplinary collaboration, ethical use of genetic data, and a commitment to translating scientific discoveries into tangible health benefits [103]. As a scientific community, we stand ready to address these challenges. By building on the solid foundation of molecular and pharmacological knowledge, and by embracing innovation while remaining rooted in rigorous experimentation, molecular pharmacology will continue to thrive and deliver on its ultimate mission: to provide ever more precise, potent, and patient-centered therapeutics for the betterment of global health.

Author Contributions

Conceptualization, C.M.D. and A.C.N.; methodology, C.M.D. and A.C.N.; software, I.-B.D.; validation, C.M.D. and A.C.N. and I.-B.D.; formal analysis, C.M.D.; investigation, C.M.D. and I.-B.D.; resources, C.M.D.; data curation, A.C.N.; writing—original draft preparation, C.M.D.; writing—review and editing, C.M.D., A.C.N. and I.-B.D.; visualization, I.-B.D.; supervision, I.-B.D.; project administration, I.-B.D.; funding acquisition, C.M.D. and A.C.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. Dave, K.; Munteanu, C.R. IL-1R2 as a Precision Therapeutic Target in Sepsis: Molecular Insights into Immune Regulation. Curr. Issues Mol. Biol. 2025, 47, 429. [Google Scholar] [CrossRef] [PubMed]
  2. Vallée, A.; Lecarpentier, Y.; Vallée, J.N. Circadian Rhythms and Energy Metabolism Reprogramming in Parkinson’s Disease. Curr. Issues Mol. Biol. 2019, 31, 21–44. [Google Scholar] [CrossRef] [PubMed]
  3. Tang, K.; Tang, Z.; Niu, M.; Kuang, Z.; Xue, W.; Wang, X.; Liu, X.; Yu, Y.; Jeong, S.; Ma, Y.; et al. Allosteric targeted drug delivery for enhanced blood-brain barrier penetration via mimicking transmembrane domain interactions. Nat. Commun. 2025, 16, 3410. [Google Scholar] [CrossRef]
  4. Salama, B.M.; Helmy, M.W.; Fouad, H.; Shamaa, M.M.; Houssen, M.E. The Synergistic Antitumor Effect of Decitabine and Vorinostat Combination on HepG2 Human Hepatocellular Carcinoma Cell Line via Epigenetic Modulation of Autophagy–Apoptosis Molecular Crosstalk. Curr. Issues Mol. Biol. 2023, 45, 5935–5949. [Google Scholar] [CrossRef] [PubMed]
  5. Yuan, H.; Hou, Y.; Jiao, Y.; Lu, X.; Liu, L. An Integrated Network Pharmacology, Molecular Docking, Molecular Dynamics Simulation, and Experimental Validation Study to Investigate the Potential Mechanism of Isoliquiritigenin in the Treatment of Ischemic Stroke. Curr. Issues Mol. Biol. 2025, 47, 627. [Google Scholar] [CrossRef]
  6. Drăgoi, C.M.; Diaconu, C.C.; Nicolae, A.C.; Dumitrescu, I.B. Redox Homeostasis and Molecular Biomarkers in Precision Therapy for Cardiovascular Diseases. Antioxidants 2024, 13, 1163. [Google Scholar] [CrossRef]
  7. Pinoșanu, E.A.; Pîrșcoveanu, D.; Albu, C.V.; Burada, E.; Pîrvu, A.; Surugiu, R.; Sandu, R.E.; Serb, A.F. Rhoa/ROCK, mTOR and Secretome-Based Treatments for Ischemic Stroke: New Perspectives. Curr. Issues Mol. Biol. 2024, 46, 3484–3501. [Google Scholar] [CrossRef]
  8. Roointan, A.; Gheisari, Y.; Hudkins, K.L.; Gholaminejad, A. Non-invasive metabolic biomarkers for early diagnosis of diabetic nephropathy: Meta-analysis of profiling metabolomics studies. Nutr. Metab. Cardiovasc. Dis. 2021, 31, 2253–2272. [Google Scholar] [CrossRef]
  9. Marques, L.; Costa, B.; Pereira, M.; Silva, A.; Santos, J.; Saldanha, L.; Silva, I.; Magalhães, P.; Schmidt, S.; Vale, N. Advancing Precision Medicine: A Review of Innovative in Silico Approaches for Drug Development, Clinical Pharmacology and Personalized Healthcare. Pharmaceutics 2024, 16, 332. [Google Scholar] [CrossRef]
  10. Tiwari, P.; Park, K.I. Ginseng-Based Nanotherapeutics in Cancer Treatment: State-of-the-Art Progress, Tackling Gaps, and Translational Achievements. Curr. Issues Mol. Biol. 2025, 47, 250. [Google Scholar] [CrossRef]
  11. Drăgoi, C.M.; Nicolae, A.C.; Dumitrescu, I.B. Emerging Strategies in Drug Development and Clinical Care in the Era of Personalized and Precision Medicine. Pharmaceutics 2024, 16, 1107. [Google Scholar] [CrossRef] [PubMed]
  12. Maudsley, S. Unraveling Oxidative Threads: Stress, Pathology, and Resilience in Biochemistry, Molecular, and Cellular Biology (2023–2025). Curr. Issues Mol. Biol. 2025, 47, 1050. [Google Scholar] [CrossRef]
  13. Drăgoi, C.M.; Nicolae, A.C.; Ungurianu, A.; Margină, D.M.; Grădinaru, D.; Dumitrescu, I.B. Circadian Rhythms, Chrononutrition, Physical Training, and Redox Homeostasis—Molecular Mechanisms in Human Health. Cells 2024, 13, 138. [Google Scholar] [CrossRef] [PubMed]
  14. Axente, M.; Mirea, A.; Sporea, C.; Pădure, L.; Drăgoi, C.M.; Nicolae, A.C.; Ion, D.A. Clinical and Electrophysiological Changes in Pediatric Spinal Muscular Atrophy after 2 Years of Nusinersen Treatment. Pharmaceutics 2022, 14, 2074. [Google Scholar] [CrossRef] [PubMed]
  15. Lis, M.; Niedziela, N.; Adamczyk-Zostawa, J.; Zalejska-Fiolka, J.; Szczygieł, J.; Sowa, A.; Świętek, A.; Adamczyk-Sowa, M. Comparative Effects of Vitamin D Supplementation on Oxidative Stress in Relapsing–Remitting Multiple Sclerosis. Curr. Issues Mol. Biol. 2024, 46, 14119–14131. [Google Scholar] [CrossRef]
  16. Armeli, F.; Mengoni, B.; Laskin, D.L.; Businaro, R. Interplay among Oxidative Stress, Autophagy, and the Endocannabinoid System in Neurodegenerative Diseases: Role of the Nrf2- p62/SQSTM1 Pathway and Nutraceutical Activation. Curr. Issues Mol. Biol. 2024, 46, 6868–6884. [Google Scholar] [CrossRef]
  17. García-Domínguez, M. Neuroinflammation: Mechanisms, Dual Roles, and Therapeutic Strategies in Neurological Disorders. Curr. Issues Mol. Biol. 2025, 47, 417. [Google Scholar] [CrossRef]
  18. Park, J.J.; Cabel, G.Y.; Cheng, K.K.; Dang, J.; Ardati, A.K.; Han, J.; Lee, J.C. Genotype-guided prescribing predictors in CYP2C19 intermediate metabolizers receiving percutaneous coronary intervention. Pharmacogenomics 2024, 25, 293–298. [Google Scholar] [CrossRef]
  19. Chua, H.M.; Limenta, M.; Ng, C.Y.L.; Lo, E.A.G. Implementation of HLA-related genotype-guided prescribing in Singapore. Am. J. Health-Syst. Pharm. 2025, 82, e285–e293. [Google Scholar] [CrossRef]
  20. Cornelissen, G.; Watanabe, Y.; Beaty, L.A.; Otsuka, K. Toward a personalized chronotherapy of blood pressure. Biomed. J. 2025, 48, 100849. [Google Scholar] [CrossRef]
  21. Creangă, E.C.; Stan, R.; Nicolae, A.C.; Drăgoi, C.M.; Dumitrescu, I.B. Personalized Therapeutic Advances in Erythropoietin Signaling: From Anemia Management to Extensive Clinical Applications. Pharmaceutics 2025, 17, 1190. [Google Scholar] [CrossRef] [PubMed]
  22. Ritivoiu, M.E.; Drăgoi, C.M.; Matei, D.; Stan, I.V.; Nicolae, A.C.; Craiu, M.; Dumitrescu, I.B.; Ciolpan, A.A. Current and Future Therapeutic Approaches of Exocrine Pancreatic Insufficiency in Children with Cystic Fibrosis in the Era of Personalized Medicine. Pharmaceutics 2023, 15, 162. [Google Scholar] [CrossRef] [PubMed]
  23. Springer, N.; Echtler, L.; Volkmann, P.; Hühne-Landgraf, A.; Hochenbleicher, J.; Hoch, E.; Koller, G.; Landgraf, D. DAILY—A personalized circadian Zeitgeber therapy as an adjunctive treatment for alcohol use disorder patients: Results of a pilot trial. Front. Psychiatry 2025, 16, 1477895. [Google Scholar] [CrossRef] [PubMed]
  24. Singampalli, A.; Kumar, P.; Bandela, R.; Bellapukonda, S.M.; Nanduri, S.; Yaddanapudi, V.M. Indazole–an emerging privileged scaffold: Synthesis and its biological significance. RSC Med. Chem. 2025, 16, 5196–5221. [Google Scholar] [CrossRef]
  25. Zhang, H.; He, F.; Gao, G.; Lu, S.; Wei, Q.; Hu, H.; Wu, Z.; Fang, M.; Wang, X. Approved Small-Molecule ATP-Competitive Kinases Drugs Containing Indole/Azaindole/Oxindole Scaffolds: R&D and Binding Patterns Profiling. Molecules 2023, 28, 943. [Google Scholar]
  26. Hussain, K.; Jahangir Alam, M.; Hussain, A.; Ahmad Siddique, N. Therapeutic Significance of Indole Scaffold in Medicinal Chemistry. Int. J. Pharm. Sci. Res. 2020, 11, 5441. [Google Scholar]
  27. Naim, M.J.; Alam, O.; Alam, J.; Bano, F.; Alam, P.; Shrivastava, N. Recent Review on Indole: A Privileged Structure Scaffold. Int. J. Pharma Sci. Res. 2016, 7, 51–62. [Google Scholar]
  28. Mo, X.; Rao, D.P.; Kaur, K.; Hassan, R.; Abdel-Samea, A.S.; Farhan, S.M.; Bräse, S.; Hashem, H. Indole Derivatives: A Versatile Scaffold in Modern Drug Discovery—An Updated Review on Their Multifaceted Therapeutic Applications (2020–2024). Molecules 2024, 29, 4770. [Google Scholar] [CrossRef]
  29. Drăgoi, C.M.; Nicolae, A.C.; Dumitrescu, I.B.; Popa, D.E.; Ritivoiu, M.; Arsene, A.L. DNA targeting as a molecular mechanism underlying endogenous indoles biological effects. Farmacia 2019, 67, 367–377. [Google Scholar] [CrossRef]
  30. Yasir, M.; Park, J.; Han, E.T.; Han, J.H.; Park, W.S.; Chun, W. Investigating the Inhibitory Potential of Flavonoids against Aldose Reductase: Insights from Molecular Docking, Dynamics Simulations, and gmx_MMPBSA Analysis. Curr. Issues Mol. Biol. 2024, 46, 11503–11518. [Google Scholar] [CrossRef]
  31. Alghamdi, A.; Abouzied, A.S.; Alamri, A.; Anwar, S.; Ansari, M.; Khadra, I.; Zaki, Y.H.; Gomha, S.M. Synthesis, Molecular Docking, and Dynamic Simulation Targeting Main Protease (Mpro) of New, Thiazole Clubbed Pyridine Scaffolds as Potential COVID-19 Inhibitors. Curr. Issues Mol. Biol. 2023, 45, 1422–1442. [Google Scholar] [CrossRef] [PubMed]
  32. Suceveanu, A.I.; Mazilu, L.; Suceveanu, A.P.; Parepa, I.; Dumitrescu, I.B.; Drăgoi, C.M.; Nicolae, A.C.; Botea, F.; Voinea, F.; Burcea-Dragomiroiu, G.T.A. Assertion for montelukast in the COVID-19 pandemics. Farmacia 2020, 68, 579–585. [Google Scholar] [CrossRef]
  33. Arsene, A.L.; Dumitrescu, I.B.; Dragoi, C.M.; Udeanu, D.I.; Lupuliasa, D.; Jinga, V.; Drăgănescu, D.; Dinu-Pîrvu, C.E.; Burcea Dragomiroiu, G.T.A.; Blejan, I.E.; et al. A new era for the therapeutic management of the ongoing COVID-19 pandemic. Farmacia 2020, 68, 185–196. [Google Scholar] [CrossRef]
  34. Wang, Q.; Lu, Q.; Guo, Q.; Teng, M.; Gong, Q.; Li, X.; Du, Y.; Liu, Z.; Tao, Y. Structural basis of the ligand binding and signaling mechanism of melatonin receptors. Nat. Commun. 2022, 13, 454. [Google Scholar] [CrossRef]
  35. Arsene, A.L.; Uivaroși, V.; Mitrea, N.; Drăgoi, C.M.; Nicolae, A.C. The binding properties of some novel ruthenium (III) complexes with human serum transferrin. Biopolym. Cell 2011, 27, 141–146. [Google Scholar] [CrossRef]
  36. Deen, M.; Hougaard, A.; Hansen, H.D.; Schain, M.; Dyssegaard, A.; Knudsen, G.M.; Ashina, M. Association Between Sumatriptan Treatment During a Migraine Attack and Central 5-HT 1B Receptor Binding. JAMA Neurol. 2019, 76, 834. [Google Scholar] [CrossRef]
  37. Lis, K.; Bartuzi, Z. Selected Technical Aspects of Molecular Allergy Diagnostics. Curr. Issues Mol. Biol. 2023, 45, 5481–5493. [Google Scholar] [CrossRef]
  38. He, B.; Nohara, K.; Park, N.; Park, Y.S.; Guillory, B.; Zhao, Z.; Garcia, J.M.; Koike, N.; Lee, C.C.; Takahashi, J.S.; et al. The Small Molecule Nobiletin Targets the Molecular Oscillator to Enhance Circadian Rhythms and Protect against Metabolic Syndrome. Cell Metab. 2016, 23, 610–621. [Google Scholar] [CrossRef]
  39. Tran, N.L.; Leconte, G.A.; Ferguson, F.M. Targeted Protein Degradation: Design Considerations for PROTAC Development. Curr. Protoc. 2022, 2, e611. [Google Scholar] [CrossRef]
  40. Kubryń, N.; Fijałkowski, Ł.; Nowaczyk, J.; Jamil, A.; Nowaczyk, A. PROTAC Technology as a New Tool for Modern Pharmacotherapy. Molecules 2025, 30, 2123. [Google Scholar] [CrossRef]
  41. Scheau, C.; Caruntu, A. Physiology Reimagined: Molecular, Metabolic, and Microenvironmental Forces Driving Epithelial Cancers. Curr. Issues Mol. Biol. 2025, 47, 1064. [Google Scholar] [CrossRef]
  42. Yang, Z.; Qi, Y.; Wang, Y.; Chen, X.; Wang, Y.; Zhang, X. Identifying Network Biomarkers in Early Diagnosis of Hepatocellular Carcinoma via miRNA–Gene Interaction Network Analysis. Curr. Issues Mol. Biol. 2023, 45, 7374–7387. [Google Scholar] [CrossRef] [PubMed]
  43. Mageau, E.; Derbowka, R.; Dickinson, N.; Lefort, N.; Kovala, A.T.; Boreham, D.R.; Tai, T.C.; Thome, C.; Tharmalingam, S. Molecular Mechanisms of Radiation Resistance in Breast Cancer: A Systematic Review of Radiosensitization Strategies. Curr. Issues Mol. Biol. 2025, 47, 589. [Google Scholar] [CrossRef] [PubMed]
  44. Kim, H.S.; Jang, S.H.; Baek, G.O.; Yoon, M.G.; Shim, J.; Han, J.E.; Kim, S.S.; Cheong, J.Y.; Eun, J.W. CTC-537E7.3 as a Liver-Specific Biomarker for Hepatocellular Carcinoma: Diagnostic and Prognostic Implications. Curr. Issues Mol. Biol. 2025, 47, 563. [Google Scholar] [CrossRef]
  45. Park, D.H.; Ramos, S.C.; Sim, H.B.; Lee, J.B.; Jang, H.Y.; Jeong, B.G.; Park, K.W.; Kang, K.Y.; Kim, J.J. Therapeutic Effects of Cephalotaxus harringtonia Leaf Extract on Hepatocellular Carcinoma via Regulation of the Intrinsic Apoptosis Pathway and Cell Cycle. Curr. Issues Mol. Biol. 2025, 47, 994. [Google Scholar] [CrossRef]
  46. Gaiaschi, L.; Bottone, M.G.; De Luca, F. Towards Effective Treatment of Glioblastoma: The Role of Combination Therapies and the Potential of Phytotherapy and Micotherapy. Curr. Issues Mol. Biol. 2024, 46, 14324–14350. [Google Scholar] [CrossRef]
  47. Niculae, D.; Dusman, R.; Leonte, R.A.; Chilug, L.E.; Dragoi, C.M.; Nicolae, A.; Serban, R.M.; Niculae, D.A.; Dumitrescu, I.B.; Draganescu, D. Biological Pathways as Substantiation of the Use of Copper Radioisotopes in Cancer Theranostics. Front. Phys. 2021, 8, 568296. [Google Scholar] [CrossRef]
  48. Kobroob, A.; Kongkaew, A.; Wongmekiat, O. Melatonin Reduces Aggravation of Renal Ischemia–Reperfusion Injury in Obese Rats by Maintaining Mitochondrial Homeostasis and Integrity through AMPK/PGC-1α/SIRT3/SOD2 Activation. Curr. Issues Mol. Biol. 2023, 45, 8239–8254. [Google Scholar] [CrossRef]
  49. Rongala, G.; Rongala, D.S.; Rongala, A.N. The Future of Precision Medicine: Targeted Therapies, Personalized Medicine and Formulation Strategies. J. Pharm. BioTech Ind. 2025, 2, 19. [Google Scholar] [CrossRef]
  50. Seripa, D.; Lozupone, M.; Miscio, G.; Stella, E.; La Montagna, M.; Gravina, C.; Maria Urbano, M.; di Mauro, L.; Daniele, A.; Greco, A.; et al. CYP2D6 genotypes in revolving door patients with bipolar disorders. Medicine 2018, 97, e11998. [Google Scholar] [CrossRef]
  51. Nicolae, A.C.; Arsene, A.L.; Vuță, V.; Popa, D.E.; Sîrbu, C.A.; Burcea-Dragomiroiu, G.T.A.; Dumitrescu, I.B.; Velescu, B.S.; Gofiță, E.; Drăgoi, C.M.; et al. In vitro P-gp expression after administration of CNS active drugs. Farmacia 2016, 64, 844–850. [Google Scholar]
  52. Zairi, I.; Bejar, M.A.; Ben Mrad, I.; Mzoughi, K.; Kraiem, S. Effect of intermittent fasting and chronotherapy on blood pressure control in hypertensive patients during Ramadan. Arter. Hypertens. 2022, 26, 67–72. [Google Scholar] [CrossRef]
  53. Zhang, Y.; Zhao, Z.; Li, W.; Tang, Y.; Wang, S. Mechanism of Taxanes in the Treatment of Lung Cancer Based on Network Pharmacology and Molecular Docking. Curr. Issues Mol. Biol. 2023, 45, 6564–6582. [Google Scholar] [CrossRef]
  54. Dragoi, C.M.; Yang, Z.; Fekry, B.; Brenna, A. Editorial: Chronobiology in cardiometabolic health and disease. Front. Pharmacol. 2025, 15, 1544963. [Google Scholar] [CrossRef] [PubMed]
  55. Drăgoi, C.M.; Moroșan, E.; Dumitrescu, I.B.; Nicolae, A.C.; Arsene, A.L.; Drăgănescu, D.; Lupuliasa, D.; Ioniţă, A.C.; Stoian, A.P.; Nicolae, C.; et al. Insights into chrononutrition: The innermost interplay amongst nutrition, metabolism and the circadian clock, in the context of epigenetic reprogramming. Farmacia 2019, 67, 557–571. [Google Scholar] [CrossRef]
  56. Chen, C.Y.; Wu, K.H.; Guo, B.C.; Lin, W.Y.; Chang, Y.J.; Wei, C.W.; Lin, M.J.; Wu, H.P. Personalized Medicine in Severe Asthma: From Biomarkers to Biologics. Int. J. Mol. Sci. 2023, 25, 182. [Google Scholar] [CrossRef]
  57. Yamamoto, Y.; Kanayama, N.; Nakayama, Y.; Matsushima, N. Current Status, Issues and Future Prospects of Personalized Medicine for Each Disease. J. Pers. Med. 2022, 12, 444. [Google Scholar] [CrossRef]
  58. Ben-Eltriki, M.; Gayle, E.J.; Walker, N.; Deb, S. Pharmacological Significance of Heme Oxygenase 1 in Prostate Cancer. Curr. Issues Mol. Biol. 2023, 45, 4301–4316. [Google Scholar] [CrossRef]
  59. Kluska, M.; Piastowska-Ciesielska, A.W.; Tokarz, P. Cell Cycle Status Influences Resistance to Apoptosis Induced by Oxidative Stress in Human Breast Cancer Cells, Which Is Accompanied by Modulation of Autophagy. Curr. Issues Mol. Biol. 2023, 45, 6325–6338. [Google Scholar] [CrossRef]
  60. Li, W.; Guo, L.; Xing, Z.; Fang, X.; Liang, H.; Zhang, S.; Shi, L.; Kuang, C.; Shi, L.; Zheng, Y.; et al. Forty-three key gene expressions involved in the effect of indoleamine 2,3-dioxygenase 1 expression on cancer prognosis may be a potential indoleamine 2,3-dioxygenase 1 inhibitor biomarker. Clin. Transl. Med. 2021, 11, e330. [Google Scholar] [CrossRef]
  61. Barbu, C.G.; Arsene, A.L.; Florea, S.; Albu, A.; Sirbu, A.; Martin, S.; Nicolae, A.C.; Burcea-Dragomiroiu, G.T.A.; Popa, D.E.; Velescu, B.S.; et al. Cardiovascular risk assessment in osteoporotic patients using osteoprotegerin as a reliable predictive biochemical marker. Mol. Med. Rep. 2017, 16, 6059–6067. [Google Scholar] [CrossRef] [PubMed]
  62. Kim, J.; Park, H.; Lee, S. Mitochondrial Dysfunction and Redox Imbalance in Neurodegeneration. Curr. Issues Mol. Biol. 2024, 47, 310–325. [Google Scholar]
  63. Hrabak, M.; Moon, C.; Bolaños-Guzmán, C.A.; Steiner, H. Vilazodone, a Selective Serotonin Reuptake Inhibitor with Diminished Impact on Methylphenidate-Induced Gene Regulation in the Striatum: Role of 5-HT1A Receptor. Mol. Neurobiol. 2024, 61, 1907–1919. [Google Scholar] [CrossRef] [PubMed]
  64. Diaconu, C.C.; Drăgoi, C.M.; Bratu, O.G.; Neagu, T.P.; Pantea Stoian, A.; Cobelschi, P.C.; Nicolae, A.C.; Iancu, M.A.; Hainăroșie, R.; Stănescu, A.M.A.; et al. New approaches and perspectives for the pharmacological treatment of arterial hypertension. Farmacia 2018, 66, 408–415. [Google Scholar] [CrossRef]
  65. Diaconu, C.C.; Cozma, M.A.; Dobrică, E.C.; Gheorghe, G.; Jichitu, A.; Ionescu, V.A.; Nicolae, A.C.; Drăgoi, C.M.; Găman, M.A. Polypharmacy in the Management of Arterial Hypertension—Friend or Foe? Medicina 2021, 57, 1288. [Google Scholar] [CrossRef]
  66. Spinelli, S.; Remigante, A.; Morabito, R. Therapeutic Effects of Natural Bioactive Compounds in the Management of Human Diseases. Curr. Issues Mol. Biol. 2025, 47, 1036. [Google Scholar] [CrossRef]
  67. Hsu, Y.T.; Wu, H.C.; Tsai, C.C.; Tsai, Y.C.; Kuo, C.Y. Plant Extracts and Natural Compounds for the Treatment of Urinary Tract Infections in Women: Mechanisms, Efficacy, and Therapeutic Potential. Curr. Issues Mol. Biol. 2025, 47, 591. [Google Scholar] [CrossRef]
  68. Drăgoi, C.M.; Olaru, O.T.; Dinu, M.; Popescu, C.; Arsene, A.L.; Dune, A.; Nicolae, A.C.; Ancuceanu, R.V.; Zbârcea, C.E.; Negreș, S.; et al. Characterisation, pharmacotoxicological and biochemical studies on Morus alba L. extract and its fractions. Farmacia 2018, 66, 120–128. [Google Scholar]
  69. Canli, K.; Turu, D.; Benek, A.; Bozyel, M.E.; Simsek, Ö.; Altuner, E.M. Biochemical and Antioxidant Properties as well as Antimicrobial and Antibiofilm Activities of Allium scorodoprasum subsp. jajlae (Vved.) Stearn. Curr. Issues Mol. Biol. 2023, 45, 4970–4984. [Google Scholar] [CrossRef]
  70. Cortes-Torres, A.G.; López-Castillo, G.N.; Marín-Torres, J.L.; Portillo-Reyes, R.; Luna, F.; Baca, B.E.; Sandoval-Ramírez, J.; Carrasco-Carballo, A. Cymbopogon citratus Essential Oil: Extraction, GC–MS, Phytochemical Analysis, Antioxidant Activity, and In Silico Molecular Docking for Protein Targets Related to CNS. Curr. Issues Mol. Biol. 2023, 45, 5164–5179. [Google Scholar] [CrossRef]
  71. Suárez-González, E.; Sandoval-Ramírez, J.; Flores-Hernández, J.; Carrasco-Carballo, A. Ginkgo biloba: Antioxidant Activity and In Silico Central Nervous System Potential. Curr. Issues Mol. Biol. 2023, 45, 9674–9691. [Google Scholar] [CrossRef] [PubMed]
  72. Zhang, Y.; Liu, Y.; Yang, S.; Yin, B.; Zhao, Z.; Huang, Z.; Wu, J.; Lin, S.; Wang, X. Water Extract of Portulaca Oleracea Inhibits PEDV Infection-Induced Pyrolysis by Caspase-1/GSDMD. Curr. Issues Mol. Biol. 2023, 45, 10211–10224. [Google Scholar] [CrossRef]
  73. Toiu, A.; Vlase, L.; Drăgoi, C.M.; Vodnar, D.; Oniga, I. Phytochemical analysis, antioxidant and antibacterial activities of Hypericum humifusum L. (Hypericaceae). Farmacia 2016, 64, 663–667. [Google Scholar]
  74. Hussain, H.; Mamadalieva, N.Z.; Hussain, A.; Hassan, U.; Rabnawaz, A.; Ahmed, I.; Green, I.R. Fruit Peels: Food Waste as a Valuable Source of Bioactive Natural Products for Drug Discovery. Curr. Issues Mol. Biol. 2022, 44, 1960–1994. [Google Scholar] [CrossRef] [PubMed]
  75. Klyushova, L.S.; Kandalintseva, N.V.; Grishanova, A.Y. Antioxidant Activity of New Sulphur- and Selenium-Containing Analogues of Potassium Phenosan against H2O2-Induced Cytotoxicity in Tumour Cells. Curr. Issues Mol. Biol. 2022, 44, 3131–3145. [Google Scholar] [CrossRef]
  76. Martínez-Huélamo, M.; Rodríguez-Morató, J.; Boronat, A.; De la Torre, R. Modulation of Nrf2 by Olive Oil and Wine Polyphenols and Neuroprotection. Antioxidants 2017, 6, 73. [Google Scholar] [CrossRef]
  77. Shaalan, W.; Hassan, N.; Ibrahim, M.G.; Schäfgen, B.; Haßdenteufel, K.; Gallwas, J.; Kiesel, L.; Schüring, A.N.; Götte, M. NUMB in Endometrial Pathology: From Adenomyosis Expression Patterns to Endometrial Cancer Survival Implications. Curr. Issues Mol. Biol. 2025, 47, 1027. [Google Scholar] [CrossRef]
  78. Qi, G.; Mi, Y.; Liu, Z.; Fan, R.; Qiao, Q.; Sun, Y.; Ren, B.; Liu, X. Dietary tea polyphenols ameliorate metabolic syndrome and memory impairment via circadian clock related mechanisms. J. Funct. Foods 2017, 34, 168–180. [Google Scholar] [CrossRef]
  79. Dumitrescu, I.B.; Drăgoi, C.M.; Nicolae, A.C. From Soil to Brain: Olive Oil Attributes, Consumer Choices, Intermittent Fasting, and Their Impact on Health. Nutrients 2025, 17, 1905. [Google Scholar] [CrossRef]
  80. Castaldo, L.; Narváez, A.; Izzo, L.; Graziani, G.; Gaspari, A.; Di Minno, G.; Ritieni, A. Red Wine Consumption and Cardiovascular Health. Molecules 2019, 24, 3626. [Google Scholar] [CrossRef]
  81. Ungurianu, A.; Margină, D.; Mihai, D.P.; Nicolae, A.C.; Drăgoi, C.M.; Grădinaru, D.; Zanfirescu, A. Caloric restriction mimetics: Pinostilbene versus resveratrol regarding SIRT1 and SIRT6 interaction. Adv. Med. Sci. 2025, 70, 44–50. [Google Scholar] [CrossRef] [PubMed]
  82. Sun, L.; Wang, Y.; Song, Y.; Cheng, X.R.; Xia, S.; Rahman, M.R.T.; Shi, Y.; Le, G. Resveratrol restores the circadian rhythmic disorder of lipid metabolism induced by high-fat diet in mice. Biochem. Biophys. Res. Commun. 2015, 458, 86–91. [Google Scholar] [CrossRef] [PubMed]
  83. Buljeta, I.; Pichler, A.; Šimunović, J.; Kopjar, M. Beneficial Effects of Red Wine Polyphenols on Human Health: Comprehensive Review. Curr. Issues Mol. Biol. 2023, 45, 782–798. [Google Scholar] [CrossRef] [PubMed]
  84. Kim, J.; Jung, S.; Kim, G.; Kim, J.; Son, B.; Shin, H. Modulation of Gut Microbiota by Cacao: Insights from an In Vitro Model. Curr. Issues Mol. Biol. 2025, 47, 414. [Google Scholar] [CrossRef]
  85. Nicolae, A.C.; Mitrea, N.; Arsene, A.L.; Constantinescu, M.Z.; Vuță, V.; Drăgoi, C.M. In vitro P-glycoprotein inhibition assay on N2a murine cell line. Farmacia 2013, 61, 481–491. [Google Scholar]
  86. Dumitrescu, I.B.; Lupuliasa, D.; Drăgoi, C.M.; Nicolae, A.C.; Pop, A.; Șaramet, G.; Drăgănescu, D. The age of pharmaceutical 3d printing. Technological and therapeutical implications of additive manufacturing. Farmacia 2018, 66, 365–389. [Google Scholar] [CrossRef]
  87. Martínez-García, E.; Rubio, A.; Torres, J. Role of Gut Microbiota in Metabolic Inflammation and Atherosclerosis. Curr. Issues Mol. Biol. 2023, 46, 1542–1560. [Google Scholar]
  88. Creangă, E.C.; Ott, C.; Nicolae, A.C.; Drăgoi, C.M.; Stan, R. Roxadustat as a Hypoxia-Mimetic Agent: Erythropoietic Mechanisms, Bioanalytical Detection, and Regulatory Considerations in Sports Medicine. Curr. Issues Mol. Biol. 2025, 47, 734. [Google Scholar] [CrossRef]
  89. Drăgoi, C.M.; Zanfirescu, A.; Dumitrescu, I.B.; Ungurianu, A.; Margină, D.M.; Nicolae, A.C. An Experimental Dynamic Investigation of the Influence of Melatonin, Serotonin and Tryptophan on the Stability of the DNA Structure. Chemistry 2024, 6, 922–940. [Google Scholar] [CrossRef]
  90. Naidoo, V.; Achilonu, I.; Mirza, S.; Hull, R.; Kandhavelu, J.; Soobben, M.; Penny, C. Computational Modelling of Tunicamycin C Interaction with Potential Protein Targets: Perspectives from Inverse Docking with Molecular Dynamic Simulation. Curr. Issues Mol. Biol. 2025, 47, 339. [Google Scholar] [CrossRef]
  91. Ali, M.M.; Ashraf, S.; Nure-e-Alam, M.; Qureshi, U.; Khan, K.M.; Ul-Haq, Z. Identification of Selective BRD9 Inhibitor via Integrated Computational Approach. Int. J. Mol. Sci. 2022, 23, 13513. [Google Scholar] [CrossRef] [PubMed]
  92. Shan, Z.; Tang, W.; Shi, Z.; Shan, T. Ferroptosis: An Emerging Target for Bladder Cancer Therapy. Curr. Issues Mol. Biol. 2023, 45, 8201–8214. [Google Scholar] [CrossRef] [PubMed]
  93. Gu, Y.; Xia, S.; Ouyang, Q.; Zhang, Y. Bioactivity Deep Learning for Complex Structure-Free Compound-Protein Interaction Prediction. J. Chem. Inf. Model. 2025, 65, 9910–9926. [Google Scholar] [CrossRef] [PubMed]
  94. Gupta, R.; Srivastava, D.; Sahu, M.; Tiwari, S.; Ambasta, R.K.; Kumar, P. Artificial intelligence to deep learning: Machine intelligence approach for drug discovery. Mol. Divers. 2021, 25, 1315–1360. [Google Scholar] [CrossRef]
  95. Pouryahya, M.; Oh, J.H.; Mathews, J.C.; Belkhatir, Z.; Moosmüller, C.; Deasy, J.O.; Tannenbaum, A.R. Pan-Cancer Prediction of Cell-Line Drug Sensitivity Using Network-Based Methods. Int. J. Mol. Sci. 2022, 23, 1074. [Google Scholar] [CrossRef]
  96. Hazra, D.; Kim, M.R.; Byun, Y.C. Generative Adversarial Networks for Creating Synthetic Nucleic Acid Sequences of Cat Genome. Int. J. Mol. Sci. 2022, 23, 3701. [Google Scholar] [CrossRef]
  97. Zeng, L.; Lu, Y.; Yan, W.; Yang, Y. A Protein Co-Conservation Network Model Characterizes Mutation Effects on SARS-CoV-2 Spike Protein. Int. J. Mol. Sci. 2023, 24, 3255. [Google Scholar] [CrossRef]
  98. Dong, M.B.; Tang, K.; Zhou, X.; Zhou, J.J.; Chen, S. Tumor immunology CRISPR screening: Present, past, and future. Trends Cancer 2022, 8, 210–225. [Google Scholar] [CrossRef]
  99. Matalqah, S.; Lafi, Z.; Asha, S.Y. Hyaluronic Acid in Nanopharmaceuticals: An Overview. Curr. Issues Mol. Biol. 2024, 46, 10444–10461. [Google Scholar] [CrossRef]
  100. Laurent, M.; Geoffroy, M.; Pavani, G.; Guiraud, S. CRISPR-Based Gene Therapies: From Preclinical to Clinical Treatments. Cells 2024, 13, 800. [Google Scholar] [CrossRef]
  101. Li, S.A.; Cadelis, M.M.; Sue, K.; Blanchet, M.; Vidal, N.; Brunel, J.M.; Bourguet-Kondracki, M.L.; Copp, B.R. 6-Bromoindolglyoxylamido derivatives as antimicrobial agents and antibiotic enhancers. Bioorg. Med. Chem. 2019, 27, 2090–2099. [Google Scholar] [CrossRef] [PubMed]
  102. Touabi, L.; Ismail, N.S.M.; Bakkar, M.R.; McLean, G.R.; Abo-zeid, Y. Antiviral Activity of Rhamnolipids Nano-Micelles Against Rhinoviruses—In Silico Docking, Molecular Dynamic Analysis and In-Vitro Studies. Curr. Issues Mol. Biol. 2025, 47, 333. [Google Scholar] [CrossRef] [PubMed]
  103. Zugaro, S.; Benedetti, E.; Caioni, G. Garlic (Allium sativum L.) as an Ally in the Treatment of Inflammatory Bowel Diseases. Curr. Issues Mol. Biol. 2023, 45, 685–698. [Google Scholar] [CrossRef] [PubMed]
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

Nicolae, A.C.; Dumitrescu, I.-B.; Drăgoi, C.M. Molecular Pharmacology at the Crossroads of Precision Medicine. Curr. Issues Mol. Biol. 2026, 48, 39. https://doi.org/10.3390/cimb48010039

AMA Style

Nicolae AC, Dumitrescu I-B, Drăgoi CM. Molecular Pharmacology at the Crossroads of Precision Medicine. Current Issues in Molecular Biology. 2026; 48(1):39. https://doi.org/10.3390/cimb48010039

Chicago/Turabian Style

Nicolae, Alina Crenguţa, Ion-Bogdan Dumitrescu, and Cristina Manuela Drăgoi. 2026. "Molecular Pharmacology at the Crossroads of Precision Medicine" Current Issues in Molecular Biology 48, no. 1: 39. https://doi.org/10.3390/cimb48010039

APA Style

Nicolae, A. C., Dumitrescu, I.-B., & Drăgoi, C. M. (2026). Molecular Pharmacology at the Crossroads of Precision Medicine. Current Issues in Molecular Biology, 48(1), 39. https://doi.org/10.3390/cimb48010039

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop