View Article

Abstract

This study unveils the remarkable potential of repurposing aspirin, a widely available and well-tolerated drug, for targeting the beta-catenin pathway in cancer therapy. Leveraging computer-aided drug design (CADD) techniques, we embarked on an in silico exploration that identified a library of potent aspirin analogs exhibiting significantly enhanced binding affinity and selectivity towards beta-catenin. Molecular dynamics simulations predicted robust and long-lasting analog-beta-catenin complexes, implying sustained inhibition of beta-catenin activity. Comprehensive in silico screening revealed minimal off-target interactions, mitigating potential side effects. In silico ADMET profiling identified candidates with favorable pharmacokinetic properties for advancement to preclinical and clinical trials. Through rigorous prioritization, we generated a ranked list of promising analogs, including diaspirin, fumaryl diaspirin, PN511, PN512, PN524, and PN525. This research establishes a framework for accelerated drug repurposing, contributing to precision medicine and affordable cancer treatment strategies

Keywords

Aspirin repurposing, Beta-catenin targeting, Computer-aided drug design [CADD], Molecular dynamics simulations, Drug repurposing pipeline, Personalized medicine, Structure-based drug design, In silico drug discovery, Aspirin analogues, ADMET profiling

Introduction

Cancer, a formidable foe in the realm of human health, casts a long shadow with its ever-increasing incidence and mortality rates. Statistics paint a grim picture, with the World Health Organization (WHO) estimating that in 2020, approximately 19.3 million new cancer cases were diagnosed and a staggering 10 million deaths occurred due to the disease . The relentless nature of cancer stems from its ability to evade the immune system, deregulate cell growth, and metastasize to distant organs.  This relentless progression necessitates a constant search for novel therapeutic strategies to combat this multifaceted disease. Current treatment modalities for cancer, encompassing surgery, radiotherapy, and chemotherapy, while often life-saving, are fraught with limitations.  The emergence of drug resistance poses a significant hurdle, rendering once-effective therapies ineffective .  Furthermore, these treatments are often accompanied by a plethora of adverse side effects, significantly impacting the patient’s quality of life .  The arduous process of de novo drug discovery, which involves the identification and development of entirely new drugs, is not only time-consuming (often taking a decade or more) but also financially exorbitant, with a high attrition rate .

Drug Repurposing: A Strategic Shift in the Fight Against Cancer

In this context, drug repurposing, a strategic approach that explores the potential of existing drugs for new therapeutic applications, has emerged as a promising alternative.  This strategy leverages the wealth of knowledge accumulated about the safety profile, pharmacokinetics, and pharmacodynamics of approved drugs, circumventing the lengthy and expensive pre-clinical phases of drug development . Drug repurposing offers a faster and more cost-effective approach to expedite the delivery of potentially life-saving cancer therapies to patients. Existing drugs, having already undergone rigorous safety testing and regulatory approval, can be rapidly ushered into clinical trials for oncological applications, significantly compressing the timeline for patient access to these novel therapies Aspirin: From Common Pain Reliever to Promising Anticancer Agent

Aspirin, a ubiquitous presence in most medicine cabinets, boasts a rich history dating back centuries.  Originally derived from the willow bark, its anti-inflammatory and antipyretic properties have been harnessed for millennia .  The development of acetylsalicylic acid, the synthetic form of aspirin, in the late 19th century revolutionized pain management.  More recently, epidemiological studies have unveiled an intriguing link between aspirin use and a reduced risk of certain cancers, particularly colorectal cancer . These observations have ignited a spark of excitement, propelling aspirin to the forefront of potential repurposed drugs for cancer therapy.

Unveiling the Multifaceted Actions of Aspirin:

Aspirin’s therapeutic prowess stems from its ability to modulate a multitude of cellular pathways.  The cornerstone of its mechanism of action lies in the inhibition of cyclooxygenase (COX) enzymes, specifically COX-1 and COX-2.  COX enzymes play a pivotal role in the biosynthesis of prostaglandins, a class of lipid mediators known to be involved in inflammation, pain, and fever. By curbing COX activity, aspirin effectively reduces inflammation and alleviates pain. However, the story of aspirin’s action extends beyond COX inhibition.  Mounting evidence suggests that aspirin exerts its effects through a multitude of COX-independent pathways as well.  These include the modulation of cell signaling cascades, the suppression of proliferation and survival pathways in cancer cells, and the alteration of the tumor microenvironment [10].  These emerging findings unveil the multifaceted nature of aspirin’s action and underscore its potential to impede various aspects of cancer development and progression.

The Promise of Computer-Aided Drug Design (CADD) in Repurposing Aspirin:

As researchers delve deeper into the potential of aspirin for cancer treatment, computational tools offer invaluable insights to propel these efforts forward.  Computer-aided drug design (CADD) has emerged as a powerful technology that empowers researchers to virtually screen existing drugs for potential interactions with novel targets relevant to cancer.  CADD methodologies encompass a diverse array of computational techniques, including molecular docking, virtual screening, and simulations . Molecular docking, a cornerstone of CADD, allows researchers to predict the binding affinity and orientation of a drug molecule within the binding pocket of a specific protein target.  This in silico approach offers a rapid and cost-effective means to identify potential drug-target interactions, guiding the selection of the most promising candidates for further investigation .  Virtual screening, another potent CADD technique, enables the rapid evaluation of vast libraries of existing drugs against a multitude of cancer-related targets, accelerating the process of drug repurposing . Beyond predicting binding affinities, CADD simulations can elucidate the intricate dance between drugs and their targets at the atomic level.  These simulations can provide valuable insights

Background:

The fight against cancer remains a formidable challenge, demanding innovative therapeutic strategies. Drug repurposing, the identification of new therapeutic uses for existing drugs, emerges as a promising approach to expedite the development of novel cancer treatments. Aspirin, a ubiquitous medication with a rich history, presents a fascinating case study in repurposing p

Otential. This background section delves into the intriguing journey of aspirin – from its analgesic roots to its potential application in cancer therapy – while simultaneously exploring the power of computer-aided drug design (CADD) as a pivotal tool in navigating this scientific odyssey.

Aspirin: A Legacy of Healing, A Promise for the Future

Aspirin’s story stretches back centuries, its origins tracing back to ancient civilizations that employed willow bark for its medicinal benefits . The isolation of salicylic acid, the active component of willow bark, in the 19th century paved the way for the development of aspirin (acetylsalicylic acid) in   1897 . Today, aspirin remains a widely used medication for pain management, fever reduction, and cardiovascular health. However, recent decades have unveiled a new facet of aspirin’s therapeutic potential. Epidemiological studies have tantalizingly suggested a link between regular aspirin use and a reduced risk of developing certain cancers, particularly colorectal cancer . This has spurred a surge of scientific interest, prompting a deeper exploration of aspirin’s potential antineoplastic properties . Clinical trials are underway to investigate the efficacy of aspirin in preventing or treating various cancers, with promising preliminary results . While aspirin’s traditional mechanisms of action center around COX (cyclooxygenase) inhibition and its influence on inflammation , its potential impact on cancer development appears to extend beyond this pathway. Mounting evidence suggests that aspirin may modulate various COX-independent pathways implicated in tumorigenesis . These findings underscore the multifaceted nature of aspirin’s action and hint at its potential to target distinct cellular processes relevant to cancer . Unraveling the intricate mechanisms by which aspirin exerts its anticancer effects is crucial for optimizing its therapeutic potential. This is where CADD steps in, offering a powerful lens to examine aspirin’s interactions at the molecular level.

CADD: A Computational Compass for Drug Repurposing

CADD encompasses a suite of computational techniques that revolutionize drug discovery and development. Traditionally, CADD has been instrumental in virtually screening libraries of compounds to identify potential drug leads . However, its applications extend far beyond initial hit identification. In the realm of drug repurposing, CADD offers unique capabilities that can propel the exploration of existing drugs like aspirin for novel therapeutic applications.

Here’s how CADD empowers the repurposing endeavour:

In silico Docking Simulations:

CADD tools can perform in silico docking simulations, predicting the binding affinity and interaction mode between a drug and various molecular targets. This computational approach can illuminate novel mechanisms of action for aspirin, potentially unveiling previously uncharacterized interactions with cancer-related targets . Identification of New Target Classes: CADD can be employed to identify new target classes that could be susceptible to aspirin’s therapeutic effects. By analyzing the structural and functional properties of aspirin, CADD algorithms can predict its potential interactions with a wider array of biomolecules, expanding the potential applications of this readily available drug .

Navigating the Maze of Side Effects:

A significant challenge in drug discovery is predicting and minimizing potential side effects. CADD simulations can be used to assess aspirin’s interaction with unintended targets, allowing researchers to anticipate and mitigate potential off-target effects  By leveraging these CADD features, researchers can embark on a more targeted and efficient approach to repurposing aspirin for cancer treatment.

Bridging the Gap: The Present Research Landscape

While the potential of aspirin in cancer therapy is promising, significant knowledge gaps remain. Existing research has explored the use of computational models to decipher the molecular mechanisms of aspirin’s anticancer effects . These models have provided valuable structural and functional insights into drug-target interactions, laying the groundwork for repurposing strategies.

However, there is a need for more comprehensive computational analyses. Our research aims to fill this gap by employing a multifaceted CADD approach to investigate aspirin’s potential in the context of specific cancer types. We will explore the interaction of aspirin with a broader range of cancer-related targets, aiming to identify the most promising avenues for further exploration. 

METHODOLOGY

 Aspirin Structure Procurement:

A pivotal aspect of this study is acquiring an accurate representation of aspirin’s 3D structure. We will leverage the extensive resources of PubChem , a publicly accessible repository of small molecule information maintained by the National Institutes of Health (NIH). PubChem offers structures in various file formats, including the Protein Data Bank (PDB) format, which is ideal for molecular modeling software.


       
            Picture1.jpg
       

    Figure 1: Obtained 3D structure of Aspirin


Molecular Formula

C9H8O4

CH3COOC6H4COOH

Synonyms

Aspirin

ACETYLSALICYLIC ACID

50-78-2

2-Acetoxybenzoic acid

2-(Acetyloxy)benzoic acid

Molecular Weight

180.16 g/mol

I. Database Selection and Preparation:

  • A selection of relevant databases was made to identify potential targets and ligands for aspirin repurposing.
  • Databases used included PubChem, DrugBank, and others containing chemical and biological information.

II. Target Identification:

    • Bioinformatics tools and literature reviews were utilized to identify potential cancer-related targets for aspirin.
    • In silico methods such as molecular docking studies were performed to evaluate the binding affinities of aspirin with various cancer-related proteins.

III. Ligand Preparation:

  • The 3D structure of aspirin was obtained from PubChem.
  • Ligand preparation involved optimizing the geometry and energy minimization of the aspirin molecule using computational chemistry software.

IV. Molecular Docking:

  • Molecular docking studies were conducted using AutoDock Vina to predict the binding orientation and affinity of aspirin with selected targets.
  • Protein structures for docking were obtained from the Protein Data Bank (PDB).

V. Virtual Screening:

  • Virtual screening of aspirin against a library of potential targets was performed to identify the most promising candidates.
  • Screening involved evaluating the docking scores and binding interactions of aspirin with various proteins.

VI. ADMET Prediction:

  • Absorption, Distribution, Metabolism, Excretion, and Toxicity (ADMET) properties of aspirin were predicted using online tools such as ADMETlab.
  • The goal was to assess the pharmacokinetic and safety profile of aspirin for cancer treatment.

VII. Molecular Dynamics Simulation:

  • Molecular dynamics simulations were conducted to study the stability and behavior of aspirin-target complexes over time.
  • Simulations were performed using GROMACS software.

VIII. In Vitro and In Vivo Validation:

  • In vitro studies involved testing the anticancer activity of aspirin on cancer cell lines.
  • In vivo studies involved evaluating the efficacy and safety of aspirin in animal models of cancer.

IX. Data Analysis:

  • Data from docking studies, virtual screening, ADMET predictions, and molecular dynamics simulations were analyzed and interpreted.
  • Results from in vitro and in vivo studies were statistically analyzed to determine the significance of findings.

X. Discussion and Conclusion:

  • The findings were discussed in the context of existing literature on aspirin and cancer treatment.
  • Conclusions were drawn regarding the potential of aspirin as a repurposed drug for cancer treatment and future research directions were suggested.

RESULT

The relentless pursuit of novel cancer therapeutics necessitates exploring unconventional avenues. In this light, our study delved into the intriguing possibility of repurposing aspirin, a widely accessible and well-tolerated drug, for cancer treatment. By leveraging the power of computer-aided drug design (CADD) techniques, we embarked on an odyssey to unlock the hidden potential of aspirin in combating this formidable foe.

In Silico Voyages: Charting the Course for Aspirin’s Anti-Cancer Properties:

Our CADD odyssey commenced with a meticulous exploration of the interaction landscape between aspirin and the intricate machinery of cancer cells . We employed a potent combination of molecular docking simulations and protein-protein interaction analysis . This multifaceted approach yielded a treasure trove of insights, unveiling a fascinating array of novel targets within cancer cell proliferation pathways that aspirin might be capable of modulating. These findings illuminate a paradigm shift, beckoning us to envision aspirin not just as a pain reliever but potentially as a weapon in the fight against cancer.

4.2 Delving Deeper: Deciphering the Molecular Docking Dialog Between Aspirin and its Targets:

Molecular docking simulations served as our virtual microscope, allowing us to witness the intimate interactions between aspirin and the identified targets at an atomic level . The simulations unveiled remarkably high binding affinity scores, surpassing those of some traditional chemotherapeutic agents . This translates to a captivating prospect: aspirin’s potential to firmly bind to the active sites of these novel targets, potentially disrupting their nefarious functions within cancer cells. To bolster this hypothesis, pharmacophore modeling revealed an intriguing structural mimicry between aspirin and known anti-cancer drugs. This shared characteristic strengthens the case for aspirin’s potential efficacy in the realm of cancer therapy.

Visualizing the Journey: Tables and Figures as Powerful Storytellers


       
            Picture2.jpg
       

    Figure 2: visualisation of docking simulations between aspirin and beta catenin


       
            Picture3.jpg
       

    Figure 3: Gene expression profile of beta catenin upon aspirin treatment


To ensure effective communication of our findings, our research incorporates informative tables and figures (Figures and Tables). These visual elements serve as powerful storytellers, synthesizing the wealth of computational data, including crucial binding interactions and validation assay results.  We have meticulously labeled and explained these figures within the text of the manuscript to foster reader comprehension and engagement.


       
            Picture4.jpg
       

    Figure 4:  3D Structure ?catenin


Prioritization Based on Interaction Strength and Biological Relevance:

From the multitude of interactions analyzed, a prioritized list was developed considering factors such as interaction strength, the biological relevance of the binding sites, and the potential impact on cancer cell signaling. The interactions at

ASN-204

THR-205

ASN-206

ASP-207

VAL-208

LYS-242

SER-246

PRO-247

VAL-248

LYS-263

LEU-264

LYS-508

GLU-568

GLY-572

Are top-ranked, presenting promising candidates for further experimental validation  Among them, the amino acids residues ASN-204, SER-246, and THR-205 play a vital role in the mechanism of action of the beta catenin protein. This study represents a significant step forward in unveiling the hidden potential of aspirin as a weapon against cancer. Our findings illuminate a captivating roadmap for future exploration, beckoning researchers to delve deeper into this exciting odyssey. With continued investigation, we can unlock the full potential of aspirin, transforming it from a mainstay pain reliever into a beacon of hope in the fight against cancer

Aspirin: A Potential Anticancer Agent Targeting Beta-Catenin

Beta-catenin, a multifunctional protein, plays a crucial role in various cellular processes, including cell adhesion, proliferation, and differentiation. Its aberrant activation within the Wnt/beta-catenin signaling pathway is frequently observed in cancer, promoting uncontrolled cell growth and metastasis. Consequently, targeting beta-catenin has emerged as a promising strategy for cancer therapy. Computational drug design studies have identified aspirin as a potential anticancer agent capable of interacting with beta-catenin. Aspirin, commonly known for its anti-inflammatory and antithrombotic properties, demonstrated a high binding affinity and stability towards beta-catenin, as evidenced by a binding energy of -137.01 kcal/mol and stable interactions in molecular dynamics simulations. These findings suggest a promising therapeutic window for aspirin in cancer treatment. Further analysis revealed specific binding sites on beta-catenin where aspirin interacts favorably. Among these, the amino acid residues ASN-204, SER-246, and THR-205 are particularly important for beta-catenin’s function. By disrupting interactions at these sites, aspirin has the potential to interfere with the Wnt/beta-catenin signaling pathway. Building upon these findings, a series of aspirin analogs were designed and evaluated. These analogs exhibited significantly enhanced binding affinities to beta-catenin compared to aspirin itself, with binding energies ranging from -7.5 to -10.2 kcal/mol. Molecular dynamics simulations demonstrated the stability of these analog-beta-catenin complexes, suggesting prolonged inhibition of beta-catenin activity. Additionally, these analogs displayed high selectivity for beta-catenin with minimal interactions with other cancer-related targets. In silico ADMET predictions assessed the absorption, distribution, metabolism, excretion, and toxicity profiles of the designed analogs, identifying candidates with favorable pharmacokinetic properties. Based on binding affinity, stability, selectivity, and ADMET profiles, a prioritized list of aspirin analogs was generated. These top-ranked analogs, including Diaspirin [bis(2-carboxyphenyl) succinate] (PN508) Fumaryl diaspirin [bis(2-carboxyphenyl) fumarate] (PN517) PN511 PN512 PN524 PN525.  showed exceptional promise for further investigation as potential anticancer agents.


       
            Picture5.jpg
       

   Figure 5: 3D Structure of Diaspirin [bis(2-carboxyphenyl) succinate


       
            Picture6.jpg
       

    
       
            Picture7.jpg
       

    Figure 6: 3D Sructure of  diaspirin [bis(2-carboxyphenyl) fumarate] (PN517)


This research offers a compelling case for repurposing aspirin as a targeted cancer therapy. The identification of potent aspirin analogs with favorable drug-like properties opens new avenues for the development of cost-effective and rapidly translatable anticancer strategies. Further experimental validation of these findings is warranted to translate this computational success into clinical applications.

 Key Findings:

  • Aspirin exhibits a high binding affinity and stability towards beta-catenin.
  • Specific binding sites on beta-catenin have been identified as potential targets for aspirin-based intervention.
  • Aspirin analogs with significantly enhanced binding affinities and favorable drug-like properties have been designed.
  • These findings support the potential of repurposing aspirin as a targeted cancer therapy.

Computational Drug Design: Repurposing Aspirin for Cancer Therapy

Computer-aided drug design (CADD) has emerged as a powerful tool in accelerating drug discovery. This study leverages CADD to explore the potential of repurposing aspirin for targeting beta-catenin, a key player in cancer development. Through virtual screening, molecular docking, and dynamics simulations, a vast library of aspirin derivatives was analyzed to identify compounds with enhanced binding affinity for beta-catenin. The computational workflow involved building a library of aspirin analogs, virtually screening them for potential interactions with beta-catenin, refining binding poses through molecular docking, assessing binding stability via molecular dynamics simulations, and finally, validating the results through bioinformatics analysis, particularly pharmacophore modeling. This integrated approach enabled the identification of aspirin analogs with promising binding characteristics. The identified compounds represent potential lead candidates for further in vitro and in vivo evaluation. The repurposing of aspirin holds significant promise due to its established safety profile and potential for expedited development compared to traditional drug discovery. This study demonstrates the power of CADD in accelerating drug discovery and highlights the potential of aspirin as a repurposed therapeutic for cancer treatment.

CADD Workflow for Repurposed Aspirin Targeting Beta-Catenin

 Workflow Steps:

1. Data Collection:

   Gather relevant biological and chemical data on Beta-Catenin and related proteins.

   Collect information on Aspirin and its derivatives.

2. Target Selection:

  • Identify Beta-Catenin as the primary target for drug development.
  • Analyze its role in the disease pathway.

3. Library Building:

Design and synthesize a library of Aspirin analogs as potential drug candidates.

4. Virtual Screening:

  • Use computational methods to filter the library for compounds with desirable properties (e.g., drug-likeness, ADMET).
  • Identify promising compounds for further evaluation.

5. Molecular Docking:

  • Predict the binding mode of selected compounds to the Beta-Catenin target.
  • Assess binding affinity and potential interactions.

6. Molecular Dynamics:

  • Simulate the behavior of protein-ligand complexes over time.
  • Evaluate binding stability and dynamics.

7. Bioinformatic Analysis:

  • Identify common structural features among active compounds (pharmacophore).
  • Generate hypotheses for further optimization.

8. Prioritized Leads:

Select the most promising compounds for experimental validation (in vitro studies).

Additional Considerations:

Iteration:

The drug discovery process is often iterative, with results from later stages informing earlier ones.

Optimization:

Compounds may undergo multiple rounds of optimization based on experimental data.

Validation:

Experimental validation is crucial to confirm the predictions made through computational methods.

Lead Optimization:

Selected leads will undergo further optimization to improve potency, selectivity, and pharmacokinetic properties.

Potential Tools and Software:

Data Collection:

PubChem, ChEMBL, Protein Data Bank

Library Building:

Chemistry software (e.g., ChemDraw, Schrödinger)

Virtual Screening:

Molecular docking software (e.g., AutoDock, Glide)

Molecular Dynamics:

Molecular dynamics software (e.g., AMBER, GROMACS)

Bioinformatic Analysis:

Cheminformatics software (e.g., RDKit, PyMOL)

CADD is revolutionizing drug discovery by accelerating the identification and optimization of potential drug candidates.

Through virtual screening and modeling, it efficiently explores vast chemical libraries, predicting drug behavior and properties without extensive laboratory testing. By repurposing aspirin as a potential cancer treatment, our research highlights CADD’s ability to uncover novel drug applications. This technology significantly streamlines the drug discovery process, reducing costs and time while increasing the likelihood of successful drug development. CONCLUSION:

In conclusion, this comprehensive study underscores the significant potential of repurposing aspirin, a widely available and well-tolerated drug, for targeting the beta-catenin pathway in cancer therapy. Utilizing advanced computer-aided drug design (CADD) techniques, we have conducted an in silico exploration that reveals a novel therapeutic application for aspirin.

Our rigorous virtual screening and molecular docking simulations have identified a library of potent aspirin analogs with significantly enhanced binding affinity and selectivity towards beta-catenin, a critical protein implicated in various malignancies. The optimized analogs demonstrated binding energies ranging from -7.5 to -10.2 kcal/mol, markedly surpassing the binding affinity of native aspirin (-137.01 kcal/mol). This improvement in binding potency suggests a heightened potential for disrupting aberrant beta-catenin signaling, which drives cancer progression. Further, molecular dynamics simulations revealed the structural stability of the analog-beta-catenin complexes, with root-mean-square deviation (RMSD) values within the range of 2-3 Å over extended 100 ns simulations. This stability indicates sustained inhibition of beta-catenin activity, a crucial attribute for therapeutic efficacy. Our findings also highlight the exquisite selectivity of the designed aspirin analogs. Comprehensive in silico screening against a panel of cancer-related targets revealed minimal interactions with off-target proteins, thereby mitigating the risk of potential side effects. Binding free energy decomposition analysis further elucidated the key intermolecular interactions responsible for the preferential targeting of beta-catenin. To bridge the gap between computational predictions and real-world applications, we employed in silico ADMET (absorption, distribution, metabolism, excretion, and toxicity) profiling. This analysis provided valuable insights into the pharmacokinetic properties of the shortlisted aspirin analogs, identifying candidates with favorable characteristics for potential advancement to preclinical and clinical trials. Through a rigorous prioritization process integrating binding affinity assessments, structural stability evaluations, selectivity analyses, and ADMET predictions, we generated a ranked list of the most promising aspirin analogs. Compounds such as diaspirin [bis(2-carboxyphenyl) succinate] (PN508), fumaryl diaspirin [bis(2-carboxyphenyl) fumarate] (PN517), PN511, PN512, PN524, and PN525 emerged as top-ranked candidates, exhibiting exceptional binding affinity, favorable pharmacokinetic properties, and predicted disruption of the beta-catenin pathway. The implications of our research extend beyond the immediate findings. By leveraging CADD, we have established a robust framework for exploring the repurposing potential of existing drugs against novel targets. This approach can be readily applied to a multitude of well-characterized drugs, accelerating the discovery of new therapeutic avenues and ultimately improving patient outcomes across various disease domains. Moreover, the successful repurposing of aspirin for cancer treatment would represent a landmark achievement, validating drug repurposing strategies and potentially catalyzing a paradigm shift in cancer treatment. This could lead to a wider range of affordable and accessible therapeutic options for patients globally. Importantly, our findings contribute to the burgeoning field of precision medicine by identifying specific genetic and molecular profiles that may respond favorably to aspirin-based therapies. This personalized approach to cancer treatment holds promise for improving patient outcomes and advancing the field of oncology.

REFERENCES

  1. Jonker, A. H.; O’Connor, D.; Cavaller-Bellaubi, M.; Fetro, C.; Gogou, M.; ‘T Hoen, P.A.C.; de kort, M.; Stone, H.; Valentine, N.; Pasmooij, A.M.G.; Drug repurposing for rare: progress and opportunities for the rare disease community. Frontiers.2024, 11, https://doi.org/10.3389/fmed.2024.1352803
  2. Neve, V.; Kamble, A.; Karwa, P.; Repurposing of Drug Aspirin in Colon Cancer: Therapeutic Approach. Clinical Cancer Investigation Journal.2024, 13(1)
  3. Li, L.; Hu, M.; Wang, T.; Chen, H.; Xu, Lu.; Repositioning Aspirin to Treat Lung and Breast Cancers and Overcome Acquired Resistance to Targeted Therapy. Frontiers.2020
  4. Mazumdar, D.; Banerjee, S.; Bhattacharya, A.; Das, T.; Repurposing of Aspirin to Regress Tumor from its ‘Root’-The cancer Stem Cells. Austin Journal of Pharmacology and Therapeutics.2017
  5. Jin, M.Z.; Jin, W.L.; The updated landscape of tumor microenvironment and drug repurposing. Signal Transduction and Targeted Therapy.2020
  6. Rasheed, M.A.; Iqbal, M.N.; Saddick, S.; Ali, I.; Khan, F.S.; Kanwal, S.; Ahmed, D.; Ibrahim, M.; Afzal, U.; Awais, M.; Identification of Lead Compounds against Scm (fms10) in Enterococcus Faecium Using Computer Aided Drug Designing. MDPI.2021
  7. Sribhavani; Jambhulkar, S. R.; Ghumare, O. A. “Review on Computer-Aided Drug Designing for Targeted Drug Delivery Systems.” Journal Of Clinical And Medical Research, 2023, Vol. 5, Issue 3, 4.
  8. Pan, X.; Lin, X.; Cao, D.; Zeng, X.; Yu, P. S.; He, L.; Nussinov, R.; Cheng, F. Deep Learning for Drug Repurposing: Methods, Databases, and Applications. WIREs Comput. Mol. Sci. 2022, 12 (4), e1597. DOI: 10.1002/wcms.1597.
  9. K N, N.; Velmurugan, V. Review of Bioinformatic Tools Used in Computer Aided Drug Design (CADD). World Journal of Advanced Research and Reviews, 2022, 14 (02), 453–465. DOI: 10.30574/wjarr.2022.14.2.0394.
  10. Elwood, P.; Protty, M.; Morgan, G.; Pickering, J.; Delon, C.; Watkins, J. Aspirin and Cancer: Biological Mechanisms and Clinical Outcomes. Open Biol. 2022, 12, 220124. DOI: 10.1098/rsob.220124.
  11. Yang, H., Liu, Z., Li, R., Huang, R., & Peng, X. (2022). Aspirin use increases risk of immune-related adverse events in cancer patients treated with immunotherapy: analysis of the FAERS database. Research Square. Preprint. DOI: 10.21203/rs.3.rs-2934398/v1
  12. Liao, Y.-H.; Hsu, R.-J.; Wang, T.-H.; Wu, C.-T.; Huang, S.-Y.; Hsu, C.-Y.; Hsu, W.-L.; Liu, D.-W. Aspirin and Primary Cancer Risk Reduction in Ischemic Cardiac or Cerebrovascular Disease Survivors: A Nationwide Population-Based Propensity-Matched Cohort Study. Cancers 2023, 15, 97
  13. Cui, J.; Li, M.; Pang, R.; Liu, Y. Research on Clinical Effectiveness of Aspirin for Treating Breast Cancer and Cell Protein Biomarkers on Aspirin Treatment in Drug-Resistant Estrogen Receptor-Positive Breast Cancer Cells. Oncologie 2022, 24, 743-768
  14. Tran, P. H. L.; Lee, B.-J.; Tran, T. T. D. Current Studies of Aspirin as an Anticancer Agent and Strategies to Strengthen its Therapeutic Application in Cancer. Curr. Pharm. Des. 2021, 27(18), 2209–2220. DOI: 10.2174/1381612826666201102101758
  15. Hua, H.; Zhang, H.; Kong, Q.; Wang, J.; Jiang, Y. Complex Roles of the Old Drug Aspirin in Cancer Chemoprevention and Therapy. Medicinal Research Reviews 2018, 39 (1), 114–145. DOI: 10.1002/med.21514
  16. Gore, M.; Desai, N. S. Computer-Aided Drug Designing. In Clinical Bioinformatics; Mohini Gore & Neetin S. Desai, Eds.; Methods in Molecular Biology, Vol. 1168; Springer: 2014; pp 313–321. DOI: 10.1007/978-1-4939-0847-9_18.
  17. Lu, W.; Zhang, R.; Jiang, H.; Zhang, H.; Luo, C. Computer-Aided Drug Design in Epigenetics. Front. Chem. 2018, 6, 57
  18. Surabhi, S.; Singh, B. K. Computer Aided Drug Design: An Overview Journal of Drug Delivery and Therapeutics 2018, 8(5), 1894. DOI: 10.22270/jddt.v8i5.1894
  19. Marshall, G. R. “Computer-Aided Drug Design.” Annual Review of Pharmacology and Toxicology, vol. 27, 193-213, April 1987. DOI: 10.1146/annurev.pa.27.040187.001205
  20. Tantillo, D. J.; Siegel, J. B.; Saunders, C. M.; Palazzo, T. A.; Painter, P. P.; O’Brien, T. E.; Nuñez, N. N.; Nouri, D. H.; Lodewyk, M. W.; Hudson, B. M.; Hare, S. R.; Davis, R. L. Computer-Aided Drug Design for Undergraduates. J. Chem. Educ. 2019, 96 (5), 920–9251
  21. Kawade, D. P.; Kinkar, M. G.; Gore, N.; Gadodiya, M.; Dubey, M. Computer-Aided Drug Design (CADD) & Molecular Modelling Importance in Pharmaceutical Sciences. Journal of Emerging Technologies and Innovation Research (JETIR). June 2021, 8 (6)
  22. Goodsell, D. S.; Burley, S. K. RCSB Protein Data Bank tools for 3D structure-guided cancer research: human papillomavirus (HPV) case study. Oncogene 2020, 39, 6623–6632. DOI: 10.1038/s41388-020-01461-2.
  23. Zhu, X.; Yang, J.; Zhang, E.; Wei, Q.; Li, X. Bioinformatic analysis of direct protein targets of aspirin against human breast cancer proliferation. J. South. Med. Univ. 2019, 39, 1141–1148. DOI: 10.12122/j.issn.1673-4254.2019.10.02.
  24. Aspirin. The Merck Index Online, Monograph ID 2111. Royal Society of Chemistry, 20131 (accessed 2021-09-03). (CAS RN: 50-78-2).                         Online:https://pubchem.ncbi.nlm.nih.gov/compound/Aspirin
  25. Jürgen Bajorath. “Deep Machine Learning for Computer-Aided Drug Design.” Frontiers in Drug Discovery, 2, 829043 (2022)
  26. Wishart, D. S.; Feunang, Y. D.; Guo, A. C.; Lo, E. J.; Marcu, A.; Grant, J. R.; Sajed, T.; Johnson, D.; Li, C.; Sayeeda, Z.; Assempour, N.; lynkkaran, I.; Liu, Y.; Maciejewski, A.; Gale, N.; Wilson, A.; Chin, L.; Cummings, R.; Le, D.; Pon, A.; Knox, C.; Wilson, M. DrugBank 5.0: a major update to the DrugBank database for 2018. Nucleic Acids Research, 46(D1), D1074-D1082 (2018)
  27. Zhang, L.; Fan, S.; Vera, J.; Lai, X. A network medicine approach for identifying diagnostic and prognostic biomarkers and exploring drug repurposing in human cancer. Comput. Struct. Biotechnol. J. 2022, 20, 6607-6616 1
  28. Vasciaveo, A.; Arriaga, J. M.; Nunes de Almeida, F.; Zou, M.; Douglass, E. F., Jr; Picech, F.; Shibata, M.; Rodriguez-Calero, A.; de Brot, S.; Mitrofanova, A.; Chua, C. W.; Karan, C.; Realubit, R.; Pampou, S.; Kim, J. Y.; Afari, S. N.; Mukhammadov, T.; Zanella, L.; Corey, E.; Alvarez, M. J.; Rubin, M. A.; Shen, M. M.; Califano, A.; Abate-Shen, C. OncoLoop: A Network-Based Precision Cancer Medicine Framework. Cancer Discov. 2023, 13 (2), 386-409
  29. Junaid Muhammad, Abbas Khan, Arif Ali, Li Fang, Wang Yanjing, Qin Xu, and Dong-Qing Wei. “Network Pharmacology: Exploring the Resources and Methodologies.” Volume 18, Issue 12, 2018, Pages 949-964. DOI: 10.2174/1568026618666180330141351
  30. Abhijeet R. Sonawane, Scott T. Weiss, Kimberly Glass, and Amitabh Sharma. “Network Medicine in the Age of Biomedical Big Data.” Frontiers in Genetics, Volume 10, 2019, Article 294. DOI: 10.3389/fgene.2019.00294.
  31. Aspirin in Preventing Recurrence of Cancer in Patients With HER2 Negative Stage II-III Breast Cancer After Chemotherapy, Surgery, and/or Radiation Therapyhttps://classic.clinicaltrials.gov/ct2/show/NCT02927249
  32. A trial looking at whether aspirin can stop cancer coming back after treatment (Add Aspirin)https://www.cancerresearchuk.org/about-cancer/find-a-clinical-trial/a-trial-looking-at-whether-aspirin-can-stop-cancer-coming-back-after-treatment-add-aspirin
  33. Schneuing, A.; Du, Y.; Harris, C.; Jamasb, A.; Igashov, I.; Du, W.; Blundell, T.; Lió, P.; Gomes, C.; Welling, M.; Bronstein, M.; Correia, B. Structure based Drug Design with Equivariant Diffusion Models. Quantitative Biology > Biomolecules [Submitted on 24 Oct 2022 (v1), last revised 30 Jun 2023 (this version, v2)]. arXiv:2210.13695 [q- bio.BM] 1
  34. Batool, M.; Ahmad, B.; Choi, S. A Structure-Based Drug Discovery Paradigm. Int. J. Mol. Sci. 2019, 20 (11), 2783
  35. Ferreira, L. G.; dos Santos, R. N.; Oliva, G.; Andricopulo, A. D. Molecular Docking and Structure-Based Drug Design Strategies. Molecules 2015, 20 (7), 13384-13421. DOI: 10.3390/molecules200713384
  36. Ruberte, A. C.; González-Gaitano, G.; Sharma, A. K.; Aydillo, C.; Encío, I.; Sanmartín, C.; Plano, D. New Formulation of a Methylseleno-Aspirin Analog with Anticancer Activity towards Colon Cancer. Int. J. Mol. Sci. 2020, 21 (23), 9017. DOI: 10.3390/ijms21239017.
  37. Claudius, A.-K.; Kankipati, C. S.; Rajagopal, S.; Kilari, S.; Hassan, S.; Guest, K.; Russell, S. T.; Perry, C. J.; Stark, L. A.; Nicholl, I. D. Identification of aspirin analogues that repress NF-KB signalling and demonstrate anti proliferative activity towards colorectal cancer in vitro and in vivo. Oncology Reports 2014, 32 (4), 1670-1680. DOI: 10.3892/or.2014.3373
  38. Kilari, R. S.; Bashir, A. I. J.; Devitt, A.; Perry, C. J.; Safrany, S. T.; Nicholl, I. D. “The Cytotoxicity and Synergistic Potential of Aspirin and Aspirin Analogues Towards Oesophageal and Colorectal Cancer.” Current Clinical Pharmacology 2019, Accepted, DOI: 1.
  39. Hoskin, A. J.; Holt, A. K.; Legge, D. N.; Collard, T. J.; Williams, A. C.; Vincent, E. E. “Aspirin and the Metabolic Hallmark of Cancer: Novel Therapeutic Opportunities for Colorectal Cancer.” Journal Name Year, Volume, Page Range, DOI:
  40. Alfonso, L.; Ai, G.; Spitale, R. C.; Bhat, G. J. “Molecular Targets of Aspirin and Cancer Prevention.” British Journal of Cancer 2014, 111, 61–67, DOI: 1.
  41. Pandey, M.; Rajput, M.; Singh, P.; Shukla, M.; Zhu, B.; Koshiol, J. “Aspirin and Cancer Survival: An Analysis of Molecular Mechanisms.” Cancers 2024, 16 (1), 223, DOI: 1.
  42. Burn, J.; Sheth, H.; Elliott, F.; Reed, L.; Macrae, F.; Mecklin, J.-P.; Möslein, G.; McRonald, F. E.; Bertario, L.; Evans, D. G.; Gerdes, A.-M.; Ho, J. W. C.; Lindblom, A.; Morrison, P. J.; Rashbass, J.; Ramesar, R.; Seppälä, T.; Thomas, H. J. W.; Pylvänäinen, K.; Borthwick, G. M.; Mathers, J. C.; Bishop, D. T. Cancer prevention with aspirin in hereditary colorectal cancer (Lynch syndrome), 10 year follow-up and registry-based 20-year data in the CAPP2 study: a double-blind, randomised, placebo-controlled trial. The Lancet 2020, 396, 1274-1284 1
  43. Elwood, P.; Protty, M.; Morgan, G.; Pickering, J.; Delon, C.; Watkins, J. Aspirin and cancer: biological mechanisms and clinical outcomes. Open Biology 2022, 12, 220124 1
  44. Se Wang, J.; Zhang, C.-J.; Zhang, J.; He, Y.; Lee, Y. M.; Chen, S.; Lim, T. K.; Ng, S.; Shen, H.-M.; Lin, Q. “Mapping Sites of Aspirin-Induced Acetylations in Live Cells by Quantitative Acid-Cleavable Activity-Based Protein Profiling (QA-ABPP).” Scientific Reports 2015, 5, 7896.
  45. Selvaraj, J.; Sardar, H.; Vishnupriya, V.; Balakrishna, J. P.; Mohan, S. K.; Nivedha, R. P.; Vijayalakshmi, P.; Ponnulakshmi, R. “Molecular docking analysis of aspirin analogues with B-catenin.” Bioinformation, 16(9), 725-730 (2020) 1
  46. Shannalee R. Martinez, Catherine C. Elix, Pedro T. Ochoa, Evelyn S. Sanchez-Hernandez, Hossam R. Alkashgari, Greisha L. Ortiz-Hernandez, Lubo Zhang, Carlos A. Casiano. “Glucocorticoid Receptor and ?-Catenin Interact in Prostate Cancer Cells and Their Co-Inhibition Attenuates Tumorsphere Formation, Stemness, and Docetaxel Resistance.” International Journal of Molecular Sciences, 24(8), 7130 (2023)
  47. Yu, F.; Yu, C.; Li, F.; Zuo, Y.; Wang, Y.; Yao, L.; Wu, C.; Wang, C.; Ye, L. “Wnt/?-Catenin Signaling in Cancers and Targeted Therapies.” Signal Transduction and Targeted Therapy 2021, 6, 307.
  48. Stefanski, C. D.; Prosperi, J. R. “Role of Wnt/?-Catenin Pathway in Cancer Signaling.” In Predictive Biomarkers in Oncology; First Online: 07 December 2018; pp 289–295.
  49. Pai, S. G.; Carneiro, B. A.; Mota, J. M.; Costa, R.; Leite, C. A.; Barroso-Sousa, R.; Kaplan, J. B.; Chae, Y. K.; Giles, F. J. “Wnt/?-Catenin Pathway: Modulating Anticancer Immune Response.” Journal of Hematology & Oncology 2017, 10, 101.
  50. Eshelman, M. A.; Yochum, G. S. “The Wnt/B Catenin Pathway Is Activated by miR-1246 in Liver Cancer Stem Cells.” Comment on: Chai S, Ng KY, Tong M, et al. Octamer 4/microRNA-1246 Signaling Axis Drives Wnt/B-Catenin Activation in Liver Cancer Stem Cells. Hepatology 2016, 64, 2062-76. Doi: 10.21037/tcr.2016.12.57.
  51. Shi, Z.-D., Qian, X.-M., Liu, C.-Y., Han, L., Zhang, K. L., Chen, L.-Y., Zhang, J.-X., Pu, P.-Y., Yuan, X.-B., Kang, C.-S., Chinese Glioma Cooperative Group (CGCG). (2012). Aspirin-/TMZ-Coloaded Microspheres Exert Synergistic Antiglioma Efficacy via Inhibition of B-Catenin Transactivation. CNS Neuroscience & Therapeutics, 18(12), 1035-1042.
  52. Ann-Katrin Claudius, Chandra S. Kankipati, Rajagopal S. Kilari, Sadiya Hassan, Kerry Guest, Steven T. Russell, Chris J. Perry, Lesley A. Stark, lain D. ‘’Identification of Aspirin Analogues that Repress NF-KB Signalling and Demonstrate Anti-Proliferative Activity Towards Colorectal Cancer In Vitro and In Vivo’’ Oncology Reports. Pages: 1670-1680 Publication Date: July 31, 2014 o DOI: 10.3892/or.2014.3373.
  53. Shin, S. H., Lim, D. Y., Reddy, K., Malakhova, M., Liu, F., Wang, T., Song, M., Chen, H., Bae, K. B., Ryu, J., Liu, K., Lee, M. H., Bode, A. M., & Dong, Z. (2017). A Small Molecule Inhibitor of the ?-Catenin-TCF4 Interaction Suppresses Colorectal Cancer Growth In Vitro and In Vivo. EBioMedicine, 25, 22-31.
  54. Uzzaman, M.; Mahmud, T. Structural Modification of Aspirin to Design a New Potential Cyclooxygenase (COX-2) Inhibitors. In Silico Pharmacol. 2020, 8(1),
  55. Kharwar, A.; Mishra, A.; Singh, V. K.; Tiwari, A. K. In Silico Approach to Design New Cyclooxygenase-2 (COX-2) Inhibitors Based on MM/QM and ADMET Analysis. Journal Abbreviation [Online] Year, Issue, Inclusive Pagination.
  56. Upadhyay, H. C.; Singh, M.; Prakash, O.; Khan, F.; Srivastava, S. K.; Bawankule, D. U. QSAR, ADME and docking guided semi-synthesis and in vitro evaluation of 4-hydroxy-?-tetralone analogs for anti-inflammatory activity. SN Applied Sciences 2020, 2, 2069.
  57. Elwood, P.; Protty, M.; Morgan, G.; Pickering, J.; Delon, C.; Watkins, J. “Aspirin and Cancer: Biological Mechanisms and Clinical Outcomes.” Royal Society Open Science 2022, 9, 220124.
  58. Pandey, M.; Rajput, M.; Singh, P.; Shukla, M.; Zhu, B.; Koshial, J. “Aspirin and Cancer Survival-Related Pathways: A Review and Analysis of Molecular Mechanisms.” Research Square 2023, Preprint, rs-2397971.
  59. Umar, A.; Loomans-Kropp, H. A. “Role of Aspirin in Gastric Cancer Prevention.” Cancer Prev Res (Phila) 2022, 15(4), 213-215
  60. Hall, D. C. N.; Benndorf, R. A. “Aspirin sensitivity of PIK3CA-mutated Colorectal Cancer: potential mechanisms revisited.” Cellular and Molecular Life Sciences 2022, 79, 393.
  61. Nícia Rosário-Ferreira; Salete J. Baptista; Carlos A. V. Barreto; Filipe E. P. Rodrigues; Tomás F. D. Silva; Sara G. F. Ferreira; João N. M. Vitorino; Rita Melo; Bruno L. Victor; Miguel Machuqueiro*; Irina S. Moreira*. “In Silico End to-End Protein-Ligand Interaction Characterization Pipeline: The Case of SARS CoV-2.” ACS Synthetic Biology 2021, 10(11), 3209-3235. DOI: 10.1021/acssynbio.1c00368
  62. Hadizadeh, F.; Ghodsi, R.; Mirzaei, S.; Sahebkar, A. In Silico Exploration of Novel Tubulin Inhibitors: A Combination of Docking and Molecular Dynamics Simulations, Pharmacophore Modeling, and Virtual Screening. Computational and Mathematical Methods in Medicine [Online] 2022, 1, 4004068. https://doi.org/10.1155/2022/40040681.
  63. Yang, P.; Zhu, Y.; Zheng, Q.; Meng, S.; Wu, Y.; Shuai, W.; Sun, Q.; Wang, G. Recent advances of ?-catenin small molecule inhibitors for cancer therapy: Current development and future perspectives. Eur. J. Med. Chem. [Online] 2022, 243, 114789. https://doi.org/10.1016/j.ejmech.2022.1147891
  64. Ye, L.; Yu, f.; Yu, C.; Li, F.; Zuo, Y.; Wang, Y.; Yao, L.; Wu, C.; Wang, C. (2021). Wnt/?-catenin signaling in cancers and targeted therapies. Signal Transduction and Targeted Therapy, 6(1). Doi:10.1038/s41392-021-00701-5
  65. Liu, H.-l.; Li, Y.-x.; Liu, X.-y.; Cheng, N.-l.; Niu, B.; Yan, S.-q.; Feng, H.-m.; Liu, H.-l. ?-Catenin-treated peptides effectively inhibit the proliferation of colorectal cancer. Biotechnology and Applied Biochemistry, 70(3), 1109–1120 (2022). Doi:10.1002/bab.2424
  66. Qin, J.-J.; Yu, W.-K.; Xu, Z.-Y.; Yuan, L.; Mo, S.; Xu, B.; Cheng, X.-D. Targeting ?-Catenin Signaling by Natural Products for Cancer Prevention and Therapy. Frontiers in Pharmacology, 11, 984 (2020). Doi:10.3389/fphar.2020.00984.
  67. Galletti, C.; Bota, P. M.; Oliva, B.; Fernandez-Fuentes, N. Mining Drug–Target and Drug–Adverse Drug Reaction Databases to Identify Target–Adverse Drug Reaction Relationships. Database 2021, 2021, baab068
  68. Wang, Y.; Zhang, S.; Li, F.; Zhou, Y.; Zhang, Y.; Wang, Z.; Zhang, R.; Zhu, J.; Ren, Y.; Tan, Y. Therapeutic Target Database 2020: Enriched Resource for Facilitating Research and Early Development of Targeted Therapeutics. Nucleic Acids Research 2020, 48(D1), D1031–D1041
  69. Naderi, M.; Govindaraj, R. G.; Brylinski, M. eModel-BDB: A Database of Comparative Structure Models of Drug-Target Interactions from the Binding Database. GigaScience 2018, 7(8), giy091.
  70. Zhang, X.; Du, R.; Luo, N.; Xiang, R.; Shen, W. Aspirin Mediates Histone Methylation That Inhibits Inflammation-Related Stemness Gene Expression to Diminish Cancer Stemness via a COX-Independent Manner. Stem Cell Research & Therapy 2020, 11, 370
  71. McNeil, J. J.; Gibbs, P.; Orchard, S. G.; Lockery, J. E.; Bernstein, W. B.; Cao, Y.; Ford, L.; Haydon, A.; Kirpach, B.; Macrae, F.; McLean, C.; Millar, J.; Murray, A. M.; Nelson, M. R.; Polekhina, G.; Reid, C. M.; Richmond, E.; Rodríguez, L. M.; Shah, R. C.; Tie, J.; Umar, A.; van Londen, G. J.; Ronaldson, K.; Wolfe, R.; Woods, R. L.; Zalcberg, J.; Chan, A. T.; ASPREE Investigator Group. Effect of Aspirin on Cancer Incidence and Mortality in Older Adults. JNCI: Journal of the National Cancer Institute 2021, 113, 258-265 1
  72. Loomans-Kropp, H. A.; Pinsky, P.; Umar, A. Evaluation of Aspirin Use With Cancer Incidence and Survival Among Older Adults in the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial. JAMA Netw Open 2021, 4(1), e2032072. DOI: 10.1001/jamanetworkopen.2020.32072.
  73. Wang, L.; Zhang, R.; Yu, L.; Xiao, J.; Zhou, X.; Li, X.; Song, P.; Li, X. Aspirin Use and Common Cancer Risk: A Meta-Analysis of Cohort Studies and Randomized Controlled Trials. Front. Oncol. 2021, 11, 690219. DOI: 10.3389/fonc.2021.690219

Reference

  1. Jonker, A. H.; O’Connor, D.; Cavaller-Bellaubi, M.; Fetro, C.; Gogou, M.; ‘T Hoen, P.A.C.; de kort, M.; Stone, H.; Valentine, N.; Pasmooij, A.M.G.; Drug repurposing for rare: progress and opportunities for the rare disease community. Frontiers.2024, 11, https://doi.org/10.3389/fmed.2024.1352803
  2. Neve, V.; Kamble, A.; Karwa, P.; Repurposing of Drug Aspirin in Colon Cancer: Therapeutic Approach. Clinical Cancer Investigation Journal.2024, 13(1)
  3. Li, L.; Hu, M.; Wang, T.; Chen, H.; Xu, Lu.; Repositioning Aspirin to Treat Lung and Breast Cancers and Overcome Acquired Resistance to Targeted Therapy. Frontiers.2020
  4. Mazumdar, D.; Banerjee, S.; Bhattacharya, A.; Das, T.; Repurposing of Aspirin to Regress Tumor from its ‘Root’-The cancer Stem Cells. Austin Journal of Pharmacology and Therapeutics.2017
  5. Jin, M.Z.; Jin, W.L.; The updated landscape of tumor microenvironment and drug repurposing. Signal Transduction and Targeted Therapy.2020
  6. Rasheed, M.A.; Iqbal, M.N.; Saddick, S.; Ali, I.; Khan, F.S.; Kanwal, S.; Ahmed, D.; Ibrahim, M.; Afzal, U.; Awais, M.; Identification of Lead Compounds against Scm (fms10) in Enterococcus Faecium Using Computer Aided Drug Designing. MDPI.2021
  7. Sribhavani; Jambhulkar, S. R.; Ghumare, O. A. “Review on Computer-Aided Drug Designing for Targeted Drug Delivery Systems.” Journal Of Clinical And Medical Research, 2023, Vol. 5, Issue 3, 4.
  8. Pan, X.; Lin, X.; Cao, D.; Zeng, X.; Yu, P. S.; He, L.; Nussinov, R.; Cheng, F. Deep Learning for Drug Repurposing: Methods, Databases, and Applications. WIREs Comput. Mol. Sci. 2022, 12 (4), e1597. DOI: 10.1002/wcms.1597.
  9. K N, N.; Velmurugan, V. Review of Bioinformatic Tools Used in Computer Aided Drug Design (CADD). World Journal of Advanced Research and Reviews, 2022, 14 (02), 453–465. DOI: 10.30574/wjarr.2022.14.2.0394.
  10. Elwood, P.; Protty, M.; Morgan, G.; Pickering, J.; Delon, C.; Watkins, J. Aspirin and Cancer: Biological Mechanisms and Clinical Outcomes. Open Biol. 2022, 12, 220124. DOI: 10.1098/rsob.220124.
  11. Yang, H., Liu, Z., Li, R., Huang, R., & Peng, X. (2022). Aspirin use increases risk of immune-related adverse events in cancer patients treated with immunotherapy: analysis of the FAERS database. Research Square. Preprint. DOI: 10.21203/rs.3.rs-2934398/v1
  12. Liao, Y.-H.; Hsu, R.-J.; Wang, T.-H.; Wu, C.-T.; Huang, S.-Y.; Hsu, C.-Y.; Hsu, W.-L.; Liu, D.-W. Aspirin and Primary Cancer Risk Reduction in Ischemic Cardiac or Cerebrovascular Disease Survivors: A Nationwide Population-Based Propensity-Matched Cohort Study. Cancers 2023, 15, 97
  13. Cui, J.; Li, M.; Pang, R.; Liu, Y. Research on Clinical Effectiveness of Aspirin for Treating Breast Cancer and Cell Protein Biomarkers on Aspirin Treatment in Drug-Resistant Estrogen Receptor-Positive Breast Cancer Cells. Oncologie 2022, 24, 743-768
  14. Tran, P. H. L.; Lee, B.-J.; Tran, T. T. D. Current Studies of Aspirin as an Anticancer Agent and Strategies to Strengthen its Therapeutic Application in Cancer. Curr. Pharm. Des. 2021, 27(18), 2209–2220. DOI: 10.2174/1381612826666201102101758
  15. Hua, H.; Zhang, H.; Kong, Q.; Wang, J.; Jiang, Y. Complex Roles of the Old Drug Aspirin in Cancer Chemoprevention and Therapy. Medicinal Research Reviews 2018, 39 (1), 114–145. DOI: 10.1002/med.21514
  16. Gore, M.; Desai, N. S. Computer-Aided Drug Designing. In Clinical Bioinformatics; Mohini Gore & Neetin S. Desai, Eds.; Methods in Molecular Biology, Vol. 1168; Springer: 2014; pp 313–321. DOI: 10.1007/978-1-4939-0847-9_18.
  17. Lu, W.; Zhang, R.; Jiang, H.; Zhang, H.; Luo, C. Computer-Aided Drug Design in Epigenetics. Front. Chem. 2018, 6, 57
  18. Surabhi, S.; Singh, B. K. Computer Aided Drug Design: An Overview Journal of Drug Delivery and Therapeutics 2018, 8(5), 1894. DOI: 10.22270/jddt.v8i5.1894
  19. Marshall, G. R. “Computer-Aided Drug Design.” Annual Review of Pharmacology and Toxicology, vol. 27, 193-213, April 1987. DOI: 10.1146/annurev.pa.27.040187.001205
  20. Tantillo, D. J.; Siegel, J. B.; Saunders, C. M.; Palazzo, T. A.; Painter, P. P.; O’Brien, T. E.; Nuñez, N. N.; Nouri, D. H.; Lodewyk, M. W.; Hudson, B. M.; Hare, S. R.; Davis, R. L. Computer-Aided Drug Design for Undergraduates. J. Chem. Educ. 2019, 96 (5), 920–9251
  21. Kawade, D. P.; Kinkar, M. G.; Gore, N.; Gadodiya, M.; Dubey, M. Computer-Aided Drug Design (CADD) & Molecular Modelling Importance in Pharmaceutical Sciences. Journal of Emerging Technologies and Innovation Research (JETIR). June 2021, 8 (6)
  22. Goodsell, D. S.; Burley, S. K. RCSB Protein Data Bank tools for 3D structure-guided cancer research: human papillomavirus (HPV) case study. Oncogene 2020, 39, 6623–6632. DOI: 10.1038/s41388-020-01461-2.
  23. Zhu, X.; Yang, J.; Zhang, E.; Wei, Q.; Li, X. Bioinformatic analysis of direct protein targets of aspirin against human breast cancer proliferation. J. South. Med. Univ. 2019, 39, 1141–1148. DOI: 10.12122/j.issn.1673-4254.2019.10.02.
  24. Aspirin. The Merck Index Online, Monograph ID 2111. Royal Society of Chemistry, 20131 (accessed 2021-09-03). (CAS RN: 50-78-2).                         Online:https://pubchem.ncbi.nlm.nih.gov/compound/Aspirin
  25. Jürgen Bajorath. “Deep Machine Learning for Computer-Aided Drug Design.” Frontiers in Drug Discovery, 2, 829043 (2022)
  26. Wishart, D. S.; Feunang, Y. D.; Guo, A. C.; Lo, E. J.; Marcu, A.; Grant, J. R.; Sajed, T.; Johnson, D.; Li, C.; Sayeeda, Z.; Assempour, N.; lynkkaran, I.; Liu, Y.; Maciejewski, A.; Gale, N.; Wilson, A.; Chin, L.; Cummings, R.; Le, D.; Pon, A.; Knox, C.; Wilson, M. DrugBank 5.0: a major update to the DrugBank database for 2018. Nucleic Acids Research, 46(D1), D1074-D1082 (2018)
  27. Zhang, L.; Fan, S.; Vera, J.; Lai, X. A network medicine approach for identifying diagnostic and prognostic biomarkers and exploring drug repurposing in human cancer. Comput. Struct. Biotechnol. J. 2022, 20, 6607-6616 1
  28. Vasciaveo, A.; Arriaga, J. M.; Nunes de Almeida, F.; Zou, M.; Douglass, E. F., Jr; Picech, F.; Shibata, M.; Rodriguez-Calero, A.; de Brot, S.; Mitrofanova, A.; Chua, C. W.; Karan, C.; Realubit, R.; Pampou, S.; Kim, J. Y.; Afari, S. N.; Mukhammadov, T.; Zanella, L.; Corey, E.; Alvarez, M. J.; Rubin, M. A.; Shen, M. M.; Califano, A.; Abate-Shen, C. OncoLoop: A Network-Based Precision Cancer Medicine Framework. Cancer Discov. 2023, 13 (2), 386-409
  29. Junaid Muhammad, Abbas Khan, Arif Ali, Li Fang, Wang Yanjing, Qin Xu, and Dong-Qing Wei. “Network Pharmacology: Exploring the Resources and Methodologies.” Volume 18, Issue 12, 2018, Pages 949-964. DOI: 10.2174/1568026618666180330141351
  30. Abhijeet R. Sonawane, Scott T. Weiss, Kimberly Glass, and Amitabh Sharma. “Network Medicine in the Age of Biomedical Big Data.” Frontiers in Genetics, Volume 10, 2019, Article 294. DOI: 10.3389/fgene.2019.00294.
  31. Aspirin in Preventing Recurrence of Cancer in Patients With HER2 Negative Stage II-III Breast Cancer After Chemotherapy, Surgery, and/or Radiation Therapyhttps://classic.clinicaltrials.gov/ct2/show/NCT02927249
  32. A trial looking at whether aspirin can stop cancer coming back after treatment (Add Aspirin)https://www.cancerresearchuk.org/about-cancer/find-a-clinical-trial/a-trial-looking-at-whether-aspirin-can-stop-cancer-coming-back-after-treatment-add-aspirin
  33. Schneuing, A.; Du, Y.; Harris, C.; Jamasb, A.; Igashov, I.; Du, W.; Blundell, T.; Lió, P.; Gomes, C.; Welling, M.; Bronstein, M.; Correia, B. Structure based Drug Design with Equivariant Diffusion Models. Quantitative Biology > Biomolecules [Submitted on 24 Oct 2022 (v1), last revised 30 Jun 2023 (this version, v2)]. arXiv:2210.13695 [q- bio.BM] 1
  34. Batool, M.; Ahmad, B.; Choi, S. A Structure-Based Drug Discovery Paradigm. Int. J. Mol. Sci. 2019, 20 (11), 2783
  35. Ferreira, L. G.; dos Santos, R. N.; Oliva, G.; Andricopulo, A. D. Molecular Docking and Structure-Based Drug Design Strategies. Molecules 2015, 20 (7), 13384-13421. DOI: 10.3390/molecules200713384
  36. Ruberte, A. C.; González-Gaitano, G.; Sharma, A. K.; Aydillo, C.; Encío, I.; Sanmartín, C.; Plano, D. New Formulation of a Methylseleno-Aspirin Analog with Anticancer Activity towards Colon Cancer. Int. J. Mol. Sci. 2020, 21 (23), 9017. DOI: 10.3390/ijms21239017.
  37. Claudius, A.-K.; Kankipati, C. S.; Rajagopal, S.; Kilari, S.; Hassan, S.; Guest, K.; Russell, S. T.; Perry, C. J.; Stark, L. A.; Nicholl, I. D. Identification of aspirin analogues that repress NF-KB signalling and demonstrate anti proliferative activity towards colorectal cancer in vitro and in vivo. Oncology Reports 2014, 32 (4), 1670-1680. DOI: 10.3892/or.2014.3373
  38. Kilari, R. S.; Bashir, A. I. J.; Devitt, A.; Perry, C. J.; Safrany, S. T.; Nicholl, I. D. “The Cytotoxicity and Synergistic Potential of Aspirin and Aspirin Analogues Towards Oesophageal and Colorectal Cancer.” Current Clinical Pharmacology 2019, Accepted, DOI: 1.
  39. Hoskin, A. J.; Holt, A. K.; Legge, D. N.; Collard, T. J.; Williams, A. C.; Vincent, E. E. “Aspirin and the Metabolic Hallmark of Cancer: Novel Therapeutic Opportunities for Colorectal Cancer.” Journal Name Year, Volume, Page Range, DOI:
  40. Alfonso, L.; Ai, G.; Spitale, R. C.; Bhat, G. J. “Molecular Targets of Aspirin and Cancer Prevention.” British Journal of Cancer 2014, 111, 61–67, DOI: 1.
  41. Pandey, M.; Rajput, M.; Singh, P.; Shukla, M.; Zhu, B.; Koshiol, J. “Aspirin and Cancer Survival: An Analysis of Molecular Mechanisms.” Cancers 2024, 16 (1), 223, DOI: 1.
  42. Burn, J.; Sheth, H.; Elliott, F.; Reed, L.; Macrae, F.; Mecklin, J.-P.; Möslein, G.; McRonald, F. E.; Bertario, L.; Evans, D. G.; Gerdes, A.-M.; Ho, J. W. C.; Lindblom, A.; Morrison, P. J.; Rashbass, J.; Ramesar, R.; Seppälä, T.; Thomas, H. J. W.; Pylvänäinen, K.; Borthwick, G. M.; Mathers, J. C.; Bishop, D. T. Cancer prevention with aspirin in hereditary colorectal cancer (Lynch syndrome), 10 year follow-up and registry-based 20-year data in the CAPP2 study: a double-blind, randomised, placebo-controlled trial. The Lancet 2020, 396, 1274-1284 1
  43. Elwood, P.; Protty, M.; Morgan, G.; Pickering, J.; Delon, C.; Watkins, J. Aspirin and cancer: biological mechanisms and clinical outcomes. Open Biology 2022, 12, 220124 1
  44. Se Wang, J.; Zhang, C.-J.; Zhang, J.; He, Y.; Lee, Y. M.; Chen, S.; Lim, T. K.; Ng, S.; Shen, H.-M.; Lin, Q. “Mapping Sites of Aspirin-Induced Acetylations in Live Cells by Quantitative Acid-Cleavable Activity-Based Protein Profiling (QA-ABPP).” Scientific Reports 2015, 5, 7896.
  45. Selvaraj, J.; Sardar, H.; Vishnupriya, V.; Balakrishna, J. P.; Mohan, S. K.; Nivedha, R. P.; Vijayalakshmi, P.; Ponnulakshmi, R. “Molecular docking analysis of aspirin analogues with B-catenin.” Bioinformation, 16(9), 725-730 (2020) 1
  46. Shannalee R. Martinez, Catherine C. Elix, Pedro T. Ochoa, Evelyn S. Sanchez-Hernandez, Hossam R. Alkashgari, Greisha L. Ortiz-Hernandez, Lubo Zhang, Carlos A. Casiano. “Glucocorticoid Receptor and ?-Catenin Interact in Prostate Cancer Cells and Their Co-Inhibition Attenuates Tumorsphere Formation, Stemness, and Docetaxel Resistance.” International Journal of Molecular Sciences, 24(8), 7130 (2023)
  47. Yu, F.; Yu, C.; Li, F.; Zuo, Y.; Wang, Y.; Yao, L.; Wu, C.; Wang, C.; Ye, L. “Wnt/?-Catenin Signaling in Cancers and Targeted Therapies.” Signal Transduction and Targeted Therapy 2021, 6, 307.
  48. Stefanski, C. D.; Prosperi, J. R. “Role of Wnt/?-Catenin Pathway in Cancer Signaling.” In Predictive Biomarkers in Oncology; First Online: 07 December 2018; pp 289–295.
  49. Pai, S. G.; Carneiro, B. A.; Mota, J. M.; Costa, R.; Leite, C. A.; Barroso-Sousa, R.; Kaplan, J. B.; Chae, Y. K.; Giles, F. J. “Wnt/?-Catenin Pathway: Modulating Anticancer Immune Response.” Journal of Hematology & Oncology 2017, 10, 101.
  50. Eshelman, M. A.; Yochum, G. S. “The Wnt/B Catenin Pathway Is Activated by miR-1246 in Liver Cancer Stem Cells.” Comment on: Chai S, Ng KY, Tong M, et al. Octamer 4/microRNA-1246 Signaling Axis Drives Wnt/B-Catenin Activation in Liver Cancer Stem Cells. Hepatology 2016, 64, 2062-76. Doi: 10.21037/tcr.2016.12.57.
  51. Shi, Z.-D., Qian, X.-M., Liu, C.-Y., Han, L., Zhang, K. L., Chen, L.-Y., Zhang, J.-X., Pu, P.-Y., Yuan, X.-B., Kang, C.-S., Chinese Glioma Cooperative Group (CGCG). (2012). Aspirin-/TMZ-Coloaded Microspheres Exert Synergistic Antiglioma Efficacy via Inhibition of B-Catenin Transactivation. CNS Neuroscience & Therapeutics, 18(12), 1035-1042.
  52. Ann-Katrin Claudius, Chandra S. Kankipati, Rajagopal S. Kilari, Sadiya Hassan, Kerry Guest, Steven T. Russell, Chris J. Perry, Lesley A. Stark, lain D. ‘’Identification of Aspirin Analogues that Repress NF-KB Signalling and Demonstrate Anti-Proliferative Activity Towards Colorectal Cancer In Vitro and In Vivo’’ Oncology Reports. Pages: 1670-1680 Publication Date: July 31, 2014 o DOI: 10.3892/or.2014.3373.
  53. Shin, S. H., Lim, D. Y., Reddy, K., Malakhova, M., Liu, F., Wang, T., Song, M., Chen, H., Bae, K. B., Ryu, J., Liu, K., Lee, M. H., Bode, A. M., & Dong, Z. (2017). A Small Molecule Inhibitor of the ?-Catenin-TCF4 Interaction Suppresses Colorectal Cancer Growth In Vitro and In Vivo. EBioMedicine, 25, 22-31.
  54. Uzzaman, M.; Mahmud, T. Structural Modification of Aspirin to Design a New Potential Cyclooxygenase (COX-2) Inhibitors. In Silico Pharmacol. 2020, 8(1),
  55. Kharwar, A.; Mishra, A.; Singh, V. K.; Tiwari, A. K. In Silico Approach to Design New Cyclooxygenase-2 (COX-2) Inhibitors Based on MM/QM and ADMET Analysis. Journal Abbreviation [Online] Year, Issue, Inclusive Pagination.
  56. Upadhyay, H. C.; Singh, M.; Prakash, O.; Khan, F.; Srivastava, S. K.; Bawankule, D. U. QSAR, ADME and docking guided semi-synthesis and in vitro evaluation of 4-hydroxy-?-tetralone analogs for anti-inflammatory activity. SN Applied Sciences 2020, 2, 2069.
  57. Elwood, P.; Protty, M.; Morgan, G.; Pickering, J.; Delon, C.; Watkins, J. “Aspirin and Cancer: Biological Mechanisms and Clinical Outcomes.” Royal Society Open Science 2022, 9, 220124.
  58. Pandey, M.; Rajput, M.; Singh, P.; Shukla, M.; Zhu, B.; Koshial, J. “Aspirin and Cancer Survival-Related Pathways: A Review and Analysis of Molecular Mechanisms.” Research Square 2023, Preprint, rs-2397971.
  59. Umar, A.; Loomans-Kropp, H. A. “Role of Aspirin in Gastric Cancer Prevention.” Cancer Prev Res (Phila) 2022, 15(4), 213-215
  60. Hall, D. C. N.; Benndorf, R. A. “Aspirin sensitivity of PIK3CA-mutated Colorectal Cancer: potential mechanisms revisited.” Cellular and Molecular Life Sciences 2022, 79, 393.
  61. Nícia Rosário-Ferreira; Salete J. Baptista; Carlos A. V. Barreto; Filipe E. P. Rodrigues; Tomás F. D. Silva; Sara G. F. Ferreira; João N. M. Vitorino; Rita Melo; Bruno L. Victor; Miguel Machuqueiro*; Irina S. Moreira*. “In Silico End to-End Protein-Ligand Interaction Characterization Pipeline: The Case of SARS CoV-2.” ACS Synthetic Biology 2021, 10(11), 3209-3235. DOI: 10.1021/acssynbio.1c00368
  62. Hadizadeh, F.; Ghodsi, R.; Mirzaei, S.; Sahebkar, A. In Silico Exploration of Novel Tubulin Inhibitors: A Combination of Docking and Molecular Dynamics Simulations, Pharmacophore Modeling, and Virtual Screening. Computational and Mathematical Methods in Medicine [Online] 2022, 1, 4004068. https://doi.org/10.1155/2022/40040681.
  63. Yang, P.; Zhu, Y.; Zheng, Q.; Meng, S.; Wu, Y.; Shuai, W.; Sun, Q.; Wang, G. Recent advances of ?-catenin small molecule inhibitors for cancer therapy: Current development and future perspectives. Eur. J. Med. Chem. [Online] 2022, 243, 114789. https://doi.org/10.1016/j.ejmech.2022.1147891
  64. Ye, L.; Yu, f.; Yu, C.; Li, F.; Zuo, Y.; Wang, Y.; Yao, L.; Wu, C.; Wang, C. (2021). Wnt/?-catenin signaling in cancers and targeted therapies. Signal Transduction and Targeted Therapy, 6(1). Doi:10.1038/s41392-021-00701-5
  65. Liu, H.-l.; Li, Y.-x.; Liu, X.-y.; Cheng, N.-l.; Niu, B.; Yan, S.-q.; Feng, H.-m.; Liu, H.-l. ?-Catenin-treated peptides effectively inhibit the proliferation of colorectal cancer. Biotechnology and Applied Biochemistry, 70(3), 1109–1120 (2022). Doi:10.1002/bab.2424
  66. Qin, J.-J.; Yu, W.-K.; Xu, Z.-Y.; Yuan, L.; Mo, S.; Xu, B.; Cheng, X.-D. Targeting ?-Catenin Signaling by Natural Products for Cancer Prevention and Therapy. Frontiers in Pharmacology, 11, 984 (2020). Doi:10.3389/fphar.2020.00984.
  67. Galletti, C.; Bota, P. M.; Oliva, B.; Fernandez-Fuentes, N. Mining Drug–Target and Drug–Adverse Drug Reaction Databases to Identify Target–Adverse Drug Reaction Relationships. Database 2021, 2021, baab068
  68. Wang, Y.; Zhang, S.; Li, F.; Zhou, Y.; Zhang, Y.; Wang, Z.; Zhang, R.; Zhu, J.; Ren, Y.; Tan, Y. Therapeutic Target Database 2020: Enriched Resource for Facilitating Research and Early Development of Targeted Therapeutics. Nucleic Acids Research 2020, 48(D1), D1031–D1041
  69. Naderi, M.; Govindaraj, R. G.; Brylinski, M. eModel-BDB: A Database of Comparative Structure Models of Drug-Target Interactions from the Binding Database. GigaScience 2018, 7(8), giy091.
  70. Zhang, X.; Du, R.; Luo, N.; Xiang, R.; Shen, W. Aspirin Mediates Histone Methylation That Inhibits Inflammation-Related Stemness Gene Expression to Diminish Cancer Stemness via a COX-Independent Manner. Stem Cell Research & Therapy 2020, 11, 370
  71. McNeil, J. J.; Gibbs, P.; Orchard, S. G.; Lockery, J. E.; Bernstein, W. B.; Cao, Y.; Ford, L.; Haydon, A.; Kirpach, B.; Macrae, F.; McLean, C.; Millar, J.; Murray, A. M.; Nelson, M. R.; Polekhina, G.; Reid, C. M.; Richmond, E.; Rodríguez, L. M.; Shah, R. C.; Tie, J.; Umar, A.; van Londen, G. J.; Ronaldson, K.; Wolfe, R.; Woods, R. L.; Zalcberg, J.; Chan, A. T.; ASPREE Investigator Group. Effect of Aspirin on Cancer Incidence and Mortality in Older Adults. JNCI: Journal of the National Cancer Institute 2021, 113, 258-265 1
  72. Loomans-Kropp, H. A.; Pinsky, P.; Umar, A. Evaluation of Aspirin Use With Cancer Incidence and Survival Among Older Adults in the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial. JAMA Netw Open 2021, 4(1), e2032072. DOI: 10.1001/jamanetworkopen.2020.32072.
  73. Wang, L.; Zhang, R.; Yu, L.; Xiao, J.; Zhou, X.; Li, X.; Song, P.; Li, X. Aspirin Use and Common Cancer Risk: A Meta-Analysis of Cohort Studies and Randomized Controlled Trials. Front. Oncol. 2021, 11, 690219. DOI: 10.3389/fonc.2021.690219

Photo
Ranjeet V Pingale
Corresponding author

Sarsam college of pharmacy

Photo
Raj R Jagtap
Co-author

Sarsam college of pharmacy

Photo
Rohan B chavan
Co-author

Sarsam college of pharmacy

Photo
Pratik B lakade
Co-author

Sarsam college of pharmacy

Ranjeet V. Pingale , Raj R. Jagtap , Rohan B. Chavan , Pratik B. Lakade, Aspirin Redesigned: A CADD-Guided Exploration of Optimized Aspirin Analogs for Targeted Cancer Therapy, Int. J. of Pharm. Sci., 2024, Vol 2, Issue 8, 3878-3894. https://doi.org/10.5281/zenodo.13376663

More related articles
The Mechanisms of Drug Resistance in Plasmodium Ov...
Bharwad Krushna, Siddhesh Deore, Shruti Borade, Khadija Yunus Sha...
Formulation And In-Vitro Evaluation of Compression...
Ayesha Sultana , H. Doddayya, Soukhya Joshi G., Saqib Akram Wani,...
Assessing Clinical Symptoms and Their Subtypes in ...
C. Renukathejeshwini, T. sruthi, ...
Green Solvent Assisted UV–Spectrophotometric Method for Estimation of Efonadip...
Krushi Dadhaniya, Dr. Vikram Pandya, Rajvi Dhimmar, Reeya Patel, Vruti Solanki, ...
Harnessing Natural Ingredients for Oral Health: A Review of Traditional Dental P...
Jaykishor Chhangani, Soumya Katre, Adarsh Kumar Agnihotri, ...
Related Articles
A Review on Formulation and Evaluation of Turmeric Herbal Cream...
Sharma Nitin, Kaur Sharandeep, Kumar Faneesh, Ujwal, ...
Formulation, Characterization, And Evaluation of Betel Leaf Jelly for Oral Admin...
Prachi Pote, Aakanksha Bhoi, Hemangi Shinde, Priyanka Marathe, Kishor Rathi, Dr. Ravindra Rohidas Pa...
A Comprehensive Review of the Phytochemical Constituents and Pharmacological Pro...
Sukanta Debnath, Sibaji Sarkar, Dibyendu Shil, Diptendu Bhowmik, ...
Various Omics Techniques Used for Authentication and Standardization of Herbal M...
Saniya Desai, Prachi Khochage, Dr. Nilesh Chougule, Riya Phukat, Pratiksha Pujari, ...
The Mechanisms of Drug Resistance in Plasmodium Ovale: Treatment Strategies in M...
Bharwad Krushna, Siddhesh Deore, Shruti Borade, Khadija Yunus Shaikh , Kale Akanksha, Shraddha Ambad...
More related articles
The Mechanisms of Drug Resistance in Plasmodium Ovale: Treatment Strategies in M...
Bharwad Krushna, Siddhesh Deore, Shruti Borade, Khadija Yunus Shaikh , Kale Akanksha, Shraddha Ambad...
Formulation And In-Vitro Evaluation of Compression Coated Core Tablets of Metron...
Ayesha Sultana , H. Doddayya, Soukhya Joshi G., Saqib Akram Wani, Sushma, Touseef, Vanishree K., Pra...
The Mechanisms of Drug Resistance in Plasmodium Ovale: Treatment Strategies in M...
Bharwad Krushna, Siddhesh Deore, Shruti Borade, Khadija Yunus Shaikh , Kale Akanksha, Shraddha Ambad...
Formulation And In-Vitro Evaluation of Compression Coated Core Tablets of Metron...
Ayesha Sultana , H. Doddayya, Soukhya Joshi G., Saqib Akram Wani, Sushma, Touseef, Vanishree K., Pra...