Personalised Medicine and Molecular Docking: Tailoring Drug Discovery for Individual Patients (2024)

What is Open Access?

Open Access is an initiative that aims to make scientific research freely available to all. To date our community has made over 100 million downloads. It’s based on principles of collaboration, unobstructed discovery, and, most importantly, scientific progression. As PhD students, we found it difficult to access the research we needed, so we decided to create a new Open Access publisher that levels the playing field for scientists across the world. How? By making research easy to access, and puts the academic needs of the researchers before the business interests of publishers.

Our Authors and Editors

We are a community of more than 103,000 authors and editors from 3,291 institutions spanning 160 countries, including Nobel Prize winners and some of the world’s most-cited researchers. Publishing on IntechOpen allows authors to earn citations and find new collaborators, meaning more people see your work not only from your own field of study, but from other related fields too.

Content Alerts

Brief introduction to this section that descibes Open Access especially from an IntechOpen perspective

How it Works Manage preferences

Contact

Want to get in touch? Contact our London head office or media team here

Careers

Our team is growing all the time, so we’re always on the lookout for smart people who want to help us reshape the world of scientific publishing.

Home > Books > Unravelling Molecular Docking - From Theory to Practice [Working Title]

Personalised Medicine and Molecular Docking: Tailoring Drug Discovery for Individual Patients (2)Open access peer-reviewed chapter - ONLINE FIRST

Written By

Noopur Khare and Pragati Khare

Submitted: 24 December 2023 Reviewed: 05 January 2024 Published: 29 April 2024

DOI: 10.5772/intechopen.1004619

Personalised Medicine and Molecular Docking: Tailoring Drug Discovery for Individual Patients (3)

Unravelling Molecular Docking - From Theory to Practice

Edited by Črtomir Podlipnik

Chapter metrics overview

4 Chapter Downloads

View Full Metrics

Abstract

The combination of molecular docking with personalised medicine represents a paradigm shift in drug development, providing unmatched accuracy in customising therapeutic approaches for specific patients. This collaborative effort utilises cutting-edge computational methods, including molecular docking, in conjunction with genetic insights to optimise and anticipate drug-receptor interactions. Revolutionary achievements could be further amplified by integrating large-scale omics data, artificial intelligence, and structural biology discoveries. Molecular docking and personalised medicine are developing fields that could lead to treatments that take into account each patient’s unique molecular profile in addition to previously unheard-of levels of accuracy in disease diagnosis. This revolutionary landscape will be further enhanced by future developments in quantum computing, CRISPR-based gene editing, and biomarker discovery. These advances will enable the realisation of a healthcare paradigm in which interventions are not only precise but also proactive, thereby realising the full potential of customised therapeutic strategies for improved patient outcomes.

Keywords

  • personalised medicine
  • molecular docking
  • drug discovery
  • individualised therapeutics
  • genomic insights

Author Information

Show +

  • Noopur Khare

    • Bhai Gurdas Group of Institutions, Sangrur,Punjab, India
  • Pragati Khare*

    • Department of Pharmacy, Bhagwant University, Ajmer,Rajasthan, India

*Address all correspondence to: pragatikhare10@gmail.com

1. Introduction

A paradigm shift is occurring in the field of contemporary healthcare, bringing in an era where patients’ individual genetic composition will be precisely taken into account when designing medical therapies. The personalised medicine method is a new strategy that acknowledges the inherent variety in individuals’ genetic codes and biological reactions [1]. It deviates from the standard one-size-fits-all model. The symbiotic relationship between molecular docking, a complex dance of genetics and computational science, and personalised medicine, as we stand at the intersection of biology, technology, and medicine. This relationship has the potential to turn drug discovery into a highly individualised endeavour [2]. Understanding the genetic underpinnings of health and disease is the cornerstone of personalised medicine. The complexity of the human genome has been revealed by advances in genomics, which have also revealed a tapestry of genetic variations that underpin disease vulnerability and impact responses to therapeutic interventions. Simultaneously, the advent of omics technologies, which include transcriptomics, proteomics, metabolomics, and genomes, has enabled researchers to interpret the molecular subtleties that differentiate individual patients. Personalised medicine makes use of this abundance of data to divide patients into groups according to their distinct molecular profiles, which enables the development of more focused and efficient treatment plans [3].

In addition to this novel strategy, molecular docking—a computational method with roots in structural biology—has become a key component in the drug development process. Drugs with improved specificity and efficacy can be logically designed by molecular docking, which simulates the interaction between tiny drug-like molecules and their biological targets. The intersection of molecular docking and personalised medicine, examining how the combination of patient-specific genetic data and computational approaches is changing the face of drug discovery [4]. We take a close look at issues and possibilities for the future as well as an insightful look at case studies to explore a future in which medication is not only customised for each patient but also carefully built for their unique genetic makeup.

The complex interactions between molecular docking and personalised medicine, revealing how these two fields can work together to transform healthcare. The story will develop as we explore the methods and uses of molecular docking to demonstrate how this computational power may be used to create medications that perfectly fit each patient’s unique molecular makeup. Drug discovery procedures that use patient-specific genetic data have the potential to revolutionise healthcare by promoting preventative and predictive medicine in addition to improving treatment efficacy. This chapter attempts to add to the ongoing discussion on moving personalised medicine and molecular docking into a new frontier in healthcare innovation by exploring ethical considerations, technical hurdles, and potential trajectories [5].

Advertisem*nt

2. Foundations of personalised medicine

2.1 Genetic basis of personalised medicine

2.1.1 Role of genomics in understanding individual variations

A thorough grasp of the genetic mosaic that sets each person apart is fundamental to personalised therapy. The study of an organism’s complete gene pool, or genomics, offers a thorough road map for figuring out the complexities of individual genetic variants and how they affect health and disease. The human genome, which is made up of between 20,000–25,000 genes, acts as a database that determines a person’s biological composition. In-depth discussion of genomics’ function as the foundation of personalised medicine, along with an explanation of how genetic data is used to identify individual differences and adjust medical interventions accordingly. Finding genetic markers linked to treatment responses and illness risk is a crucial task for genomics. Thanks to developments in high-throughput sequencing technologies, it is now possible to efficiently and economically decode a person’s whole genome, which enables scientists to identify gene variants that may predispose people to particular diseases. The abundance of genetic data makes it easier to divide patients into groups according to genetic characteristics they have in common, opening the door to more specialised and efficient treatment plans [6].

The investigation of individual genetic variants covers drug metabolism and response in addition to illness propensity. A branch of genomics called pharmacogenomics studies how a person’s genetic makeup affects their response to medication, including how effective it is and how likely it is to cause side effects [7]. Clinicians can prescribe drugs that are not just suited to a patient’s particular ailment but also best suited to their individual genetic composition by having a thorough understanding of these genetic subtleties. The genetic strands that weave the story of individual differences as we traverse the complex terrain of genomics in personalised medicine. We set out on a trip that emphasises the revolutionary potential of deciphering the genetic code for customising medical therapies to the unique needs of each patient through a thorough assessment of the role of genomics in healthcare (Figure 1) [8].

Personalised Medicine and Molecular Docking: Tailoring Drug Discovery for Individual Patients (4)

2.1.2 Identification of genetic markers for disease susceptibility

Knowing the genetic foundations of illness susceptibility is essential to the goal of personalised medicine. Numerous genetic markers—distinct variations in DNA sequences—have been found through genomic research. These markers serve as markers that help researchers better understand an individual’s susceptibility to particular diseases [9]. This part delves into the complex terrain of determining genetic markers associated with illness vulnerability, highlighting the revolutionary implications of this understanding on customising medicinal therapies. Single nucleotide polymorphisms (SNPs) and more complicated structural variants are examples of genetic markers that act as genomic fingerprints to identify individuals. By utilising sophisticated sequencing methods and conducting extensive genome-wide association studies (GWAS), scientists are able to identify the markers linked to heightened vulnerability to a range of illnesses. These genetic discoveries have significant ramifications for early detection, risk assessment, and preventive measures [10].

In multifactorial disorders, when the total risk is influenced by both genetic and environmental variables, the identification of genetic markers is very clear. Complex genetic architectures are present in diseases like cancer, cardiovascular disease, and neurological illnesses. By identifying particular markers, patients can be categorised according to their genetic predispositions. In addition to improving prognosis, this stratification makes it easier to design tailored therapies that take unique risk profiles into account [11]. Personalised medicine is becoming more and more dependent on the ability to detect genetic markers for disease vulnerability. Clinicians can now proactively tailor prevention strategies, surveillance protocols, and therapeutic approaches by deciphering the genetic clues embedded in an individual’s DNA. This marks a new era in healthcare where interventions are precisely aligned with each patient’s unique genetic makeup [12].

2.2 Omics technologies

2.2.1 Genomics

With genomics at its core, omics technologies are revolutionising the field of personalised medicine by enabling the deciphering of the intricate workings of individual genetic codes. Genomics is a branch of omics that studies all of an organism’s genes in detail. It offers a thorough understanding of the genetic basis of health, disease susceptibility, and response to therapy [13]. Utilising high-throughput sequencing technology, like next-generation sequencing (NGS), which makes it possible to quickly and affordably sequence complete genomes, is at the core of genomics research. With the advancement of technology, scientists can now quickly decode the three billion base pairs that make up the human genome, bringing genomics into the mainstream of scientific study [14].

The identification of genetic differences, such as single nucleotide polymorphisms (SNPs) and structural changes, among heterogeneous populations is largely dependent on genomics. Large-scale programmes such as the Human Genome Project and others have accumulated a vast store of genomic data that makes it easier to identify genetic markers linked to treatment results and disease risk. In the effort to decipher the complex connection between genetics and personal health, these markers act as navigational aids [15]. Moreover, the discipline of pharmacogenomics, which studies how genetic variants affect drug responses, benefits from the extension of genomics. By customising medication prescriptions according to a person’s genetic composition, this field of genomics maximises treatment effectiveness and reduces side effects [16]. Genomic research serves as a cornerstone in the era of personalised medicine by offering the fundamental knowledge needed to understand the subtleties of individual genetic variants. The future of precision medicine and better patient outcomes is promised by the integration of genomics into healthcare, where medical therapies will be precisely tailored to each patient’s distinct genetic signature [17].

2.2.2 Transcriptomics

Transcriptomics is a dynamic field within the complicated field of omics technologies that is essential to understanding the intricate workings of gene expression within an organism. The study of all RNA transcripts—molecules that transfer genetic information from DNA to protein synthesis—is the focus of transcriptomics [18]. This field provides insights into the molecular orchestration of biological processes by helping to understand how genes are activated or silenced under different physiological situations. Transcriptomics has undergone a revolution thanks to high-throughput methods like RNA sequencing (RNA-Seq), which allow for the simultaneous investigation of thousands of transcripts from a variety of tissues and cell types. With the help of this technique, the dynamic transcriptome may be fully captured, illuminating the subtleties of non-coding RNA expression, alternative splicing, and gene regulation [19].

Transcriptomics reveals the molecular markers linked to certain diseases and the unique responses to treatment interventions, which has profound implications for personalised medicine. Transcriptomics helps to stratify patient groups and directs doctors in the selection of customised treatment methods by detecting gene expression patterns indicative of disease states or treatment efficacy. Transcriptomics is essentially a molecular storyteller that tells the complex stories of gene activity in cells. Transcriptomics, a key element of omics technologies, advances our knowledge of the intricate molecular processes underlying health and disease and opens the door to more specialised and individualised medical care [20].

2.2.3 Proteomics

Proteomics is the dominant field in the mosaic of omics technologies, exploring the in-depth analysis of an organism’s whole protein repertoire. Proteomics is essential for understanding the dynamic landscape of protein expression, changes, and interactions within cells. Proteins are the molecular workhorses that carry out the functions encoded by the genome. Proteomic investigations are made possible by sophisticated methods like mass spectrometry and two-dimensional gel electrophoresis, which allow thousands of proteins to be identified and quantified at once. A comprehensive picture of the proteome is offered by this high-throughput method, which captures the nuances of cellular functions, signalling cascades, and reactions to external stimuli [21]. By revealing protein patterns linked to certain diseases, proteomics makes a substantial contribution to personalised medicine by facilitating the discovery of biomarkers for early diagnosis and prognosis. Additionally, it clarifies the range of protein isoforms and post-translational changes, providing information on how therapy responses differ throughout individuals [22]. Proteomics’ incorporation into biomedical research enables a more thorough comprehension of the complex molecular mechanisms underlying health and illness. Proteomics, a fundamental component of omics technologies, opens the door for precision medicine, which promises more efficient and individualised treatment plans by customising therapeutic approaches to each patient’s own protein composition [23].

2.2.4 Metabolomics

Within the complex field of omics technologies, metabolomics is a potent field devoted to the in-depth analysis of an organism’s metabolome, or the entire collection of tiny molecules, or metabolites, that are involved in cellular functions. A snapshot of the physiological state is given in real time by metabolomics, which captures the results of cellular activity and their dynamic interactions [24]. Metabolomic analyses are powered by state-of-the-art analytical methods including mass spectrometry and nuclear magnetic resonance (NMR) spectroscopy, which allow for the identification and measurement of a wide range of metabolites. This high-throughput method provides information on biochemical processes, cellular homeostasis, and metabolic pathways [25].

Metabolomics plays a major role in personalised medicine by providing insights into the metabolic fingerprints linked to different diseases and how each patient reacts to different therapies. Metabolomics profiles the distinct fingerprint of metabolites in biological samples to help identify biomarkers, diagnose diseases, and optimise treatment approaches. Metabolomics, a crucial component of omics technologies, completes the whole picture of an organism’s molecular composition [26]. Enhancing our knowledge of biochemical networks and advancing precision medicine—where treatments are customised to each patient’s unique metabolic profile—are two benefits of this research. The addition of metabolomics to the omics toolkit represents a paradigm change in healthcare towards more individualised and complex interventions (Figure 2) [27].

Personalised Medicine and Molecular Docking: Tailoring Drug Discovery for Individual Patients (5)

2.3 Patient stratification and targeted therapies

2.3.1 Subpopulations and treatment responses

The deliberate identification and grouping of patients into subpopulations according to shared genetic, molecular, or clinical traits is a fundamental component of personalised medicine. A more sophisticated knowledge of individual differences in disease susceptibility and treatment responses is made possible by this patient stratification. Targeted medicines can be carefully adjusted to satisfy the unique needs of different subgroups by taking into account the heterogeneity within patient cohorts [28]. Patient stratification in the era of genetic medicine greatly depends on the knowledge obtained from omics technology. Subpopulations with differing susceptibilities to diseases and treatment responses are defined by molecular signatures that are identified through the use of genomic profiling, transcriptomics, proteomics, and metabolomics. This method goes beyond the conventional one-size-fits-all paradigm by acknowledging the distinct genetic composition that impacts each person’s particular health trajectory [29].

When it comes to targeted medicines, patient classification is very important. These treatments target certain pathways or substances linked to disease processes, acting at the molecular or cellular level. Targeted therapies, which represent a paradigm shift from conventional treatments, can achieve higher efficacy and fewer adverse effects by identifying subpopulations with shared molecular markers. Patient stratification is a significant step towards precision medicine, where treatment approaches are tailored to each patient’s unique needs. This trend is being driven by advancements in omics technologies. This customised approach not only improves treatment results but also establishes the framework for a more efficient and individualised healthcare system [30].

2.3.2 Tailoring therapies based on patient characteristics

The emergence of personalised medicine signals a paradigm shift in healthcare by highlighting the necessity of customising treatment procedures according to the unique characteristics of each patient. One of the main tenets of this paradigm is patient stratification, which is the methodical division of patients into groups according to their distinct clinical, molecular, or genetic characteristics [31]. In addition to revealing the innate variation among patient populations, this stratification paves the way for the creation of tailored targeted medicines that target particular traits unique to each grouping. Customising treatment plans according to the features of each patient represents a significant shift from the traditional one-size-fits-all approach. By merging data from omics technologies—genomics, transcriptomics, and proteomics, for example—clinicians can better comprehend the intricate molecular mechanisms underlying disease and how treatments affect patients’ outcomes. This information facilitates the discovery of biomarkers, genetic variants, and other characteristics unique to each patient that influence the choice of targeted treatments [32].

Personalised medicine is typified by targeted medicines, which precisely target specific molecules or pathways involved in disease processes at the molecular or cellular level. These treatments represent a paradigm change towards more effective and individualised healthcare by matching treatment plans to the unique features of patient subgroups [33]. They also increase efficacy while reducing side effects. The combination of targeted therapies and patient stratification is a novel approach that allows medical interventions to be customised to each patient’s specific characteristics. This is ushering in a new era of precision medicine that has the potential to completely transform patient care and treatment outcomes [34].

Advertisem*nt

3. Molecular docking in drug discovery

3.1 Overview of molecular docking

A key component of rational drug design is molecular docking, which offers a computer framework for predicting the favoured orientation and binding affinity of a small molecule (ligand) inside the target macromolecule’s (receptor) active site. This method is essential for optimising the interactions between drugs and receptors, which makes it easier to design and create new pharmaceutical agents [35]. Fundamentally, molecular docking is based on scoring functions and algorithms thatassess the geometry and energetics of putative ligand-receptor complexes. The procedure entails methodically examining the conformational space of the receptor and ligand in order to forecast the ideal binding pose in light of complimentary structural characteristics. In order to model and evaluate the stability of the ligand-receptor complex, the concepts of molecular docking take into account intermolecular forces such as hydrogen bonding, electrostatic interactions, and van der Waals forces [36].

Clarifying the binding mechanism and affinity between a drug candidate and its target is the main goal of molecular docking, since it provides important information about the viability and possible effectiveness of the interaction. By reducing the number of possible candidates for additional experimental validation, this in silico method greatly speeds up the drug discovery process and ultimately leads to the production of therapeutic medicines with improved specificity and affinity for their target targets (Figure 3) [37].

Personalised Medicine and Molecular Docking: Tailoring Drug Discovery for Individual Patients (6)

3.1.1 Importance in rational drug design

Molecular docking is essential to the logical design of pharmaceuticals because it acts as a computerised compass to navigate the complex process from lead identification to the creation of potent medicinal compounds. Molecular docking, at its core, offers a systematic and economical way to anticipate and optimise the interactions between small compounds and their target macromolecules, which is essential to rational drug design. Because it can virtually screen large compound libraries, molecular docking is important because it can speed up the drug discovery process [38]. Docking algorithms anticipate the most favourable binding conformations and energetically favourable interactions by modelling the binding of putative drug candidates to target receptors. Promising lead compounds are prioritised, their structures are optimised, and experimental efforts are rationalised for additional validation with the use of this prediction capability [39].

Additionally, the investigation of structure-activity relationships (SAR) and the comprehension of ligand-receptor interactions at the atomic level are made easier by molecular docking. It guides the iterative process of drug optimisation by enabling researchers to fine-tune chemical structures to improve binding affinities and specificity [40]. Molecular docking, which serves as a computational link between theoretical predictions and experimental validations, is fundamental to rational drug design. In addition to hastening the identification of possible treatments, its function in optimising the drug discovery process helps create safer, more effective medications that have a better chance of succeeding in clinical settings [41].

3.2 Techniques and algorithms

3.2.1 Ligand-based docking

A basic method of molecular docking that emphasises the features and attributes of the small molecule ligand is called “ligand-based docking.” This method is predicated on the idea that ligands with similar structures will have comparable binding patterns and bioactivities. In contrast to structure-based docking, which depends on the structure of the target receptor, ligand-based docking is useful in situations where experimental structures are not available and does not require receptor structural information [42]. In the process of ligand-based docking, algorithms compare the chemical structures of known ligands and the possible drug candidate in order to estimate the binding affinity based on properties that are common [43]. Molecular fingerprinting, pharmacophore modelling, and quantitative structure-activity relationship (QSAR) analysis are examples of common approaches. Scoring functions are used by molecular docking programmes like AutoDock and GOLD to rate possible ligand conformations and orientations within the binding site [44].

Particularly useful is ligand-based docking when the receptor structure is unclear or difficult to ascertain. It guides the search for possible therapeutic candidates by utilising the body of knowledge already available regarding ligand-receptor interactions and chemical similarities. Before moving on to more resource-intensive experimental validation, this method helps identify and optimise lead compounds with desirable pharmacological properties, which is crucial in the early phases of drug discovery [45].

3.2.2 Structure-based docking

A pillar of the molecular docking community is structure-based docking, which depends on the three-dimensional structures of the target receptor and the tiny ligand. Using computer algorithms, this method predicts the ideal binding shape and affinity between the ligand and the receptor, offering vital information for well-thought-out drug design. In structure-based docking, the search for ligand binding sites is directed by the crystallographic or computationally predicted structure of the target receptor [46]. The spatial orientations of the ligand within the binding pocket of the receptor are carefully explored using algorithms like the Lamarckian Genetic Algorithm and Monte Carlo simulations. To help determine the best binding poses, the scoring functions evaluate the energetics and complementarity of possible ligand-receptor complexes. This method works best in situations where comprehensive structural data about the target is accessible, allowing for an accurate comprehension of ligand-receptor interactions at the atomic level. It is essential for virtual screening of big compound libraries and lead optimisation. Structure-based docking is a valuable tool in drug development since it facilitates the identification of promising candidates for treatment and provides valuable insights for further experiments, ultimately leading to the rational design of pharmacological medicines with improved efficacy and specificity (Figure 4) [47].

Personalised Medicine and Molecular Docking: Tailoring Drug Discovery for Individual Patients (7)

3.3 Applications in drug discovery

3.3.1 Identification of potential drug candidates

Drug discovery benefits greatly from the use of molecular docking, especially in the early phases when finding viable treatment options is crucial. By predicting the binding affinity and orientation of tiny molecules within the active region of a target receptor, docking algorithms enable the systematic screening of large chemical libraries. Molecular docking helps find drugs that have good interactions with the target by means of virtual screening. This procedure speeds up the drug development process a great deal and allows researchers to concentrate their efforts on a small number of molecules that have the best chance of becoming successful. The algorithms enable the identification of lead candidates for additional experimental validation by prioritising compounds according to their estimated binding affinities [48].

Additionally, molecular docking guides the design and optimisation of new compounds with improved binding capabilities, assisting in the exploration of chemical space. It helps with the repeated improvement of chemical structures to increase specificity and efficacy by offering insightful information on the structure-activity relationships (SAR) of ligands. The use of molecular docking to find viable drug candidates is consistent with the rational drug design principles; it facilitates lead discovery and aids in the creation of novel pharmaceuticals that may prove to be therapeutically effective [49].

3.3.2 Optimization of drug-receptor interactions

Drug development relies heavily on molecular docking, which makes it easier to optimise how medications interact with their target receptors. Molecular docking algorithms are used to anticipate and fine-tune the binding modes, orientations, and affinities inside the target’s active site after possible drug candidates have been found. To improve the effectiveness and specificity of pharmaceutical agents, rational drug design includes optimising drug-receptor interactions as a critical step. Docking algorithms estimate the best binding poses by repeatedly exploring the ligand’s conformational space. Medicinal chemists use these predictions as a guidance when fine-tuning drug candidates’ chemical structures in order to increase binding affinity and maximise interactions with the target [50].

Moreover, molecular docking helps to clarify structure-activity relationships (SAR) by illuminating how modifications to a molecule’s structure affect binding. With this information, researchers can more effectively alter drug candidates to achieve better pharmaco*kinetics and less off-target effects. Molecular docking expedites the drug optimisation process and lowers lead development time and costs by fusing computational predictions with experimental confirmation. In the pursuit of novel and focused pharmaceutical treatments, this application makes sure that possible medicines are not only found but also optimised for their binding interactions. This ultimately leads to the development of safer, more effective medications [51].

Advertisem*nt

4. Integration of personalised medicine and molecular docking

4.1 Customised drug design

4.1.1 Incorporating patient-specific genetic information

The combination of molecular docking and personalised medicine has the potential to transform drug design through the use of genetic information specific to each patient. This methodology recognises the innate genetic variability among individuals and utilises omics data, encompassing genomics, to customise medication interventions with unparalleled accuracy. In this setting, molecular docking acts as a computational bridge, making predictions about how individual differences may affect drug-receptor interactions based on patient-specific genetic data. Docking algorithms can mimic the binding affinity and orientation of medication candidates within the setting of particular genetic markers or variants by taking into account the individual genetic composition of each patient. This customised strategy goes beyond traditional paradigms for medication design. It makes it possible to develop pharmaceuticals that are tailored to each patient’s unique genetic profile, potentially increasing therapeutic effectiveness while reducing side effects. For example, pharmacogenomic data can be analysed using algorithms to identify genetic markers that affect medication metabolism. This information can then be used to customise dose regimens based on the reactions of specific patients [52].

The integration of genetic data unique to each patient into molecular docking not only opens up new possibilities for unparalleled personalisation in drug design, but it also raises the possibility of safer and more effective treatments. The convergence of computational techniques and personalised medicine holds great promise for revolutionising precision medication design, enabling a paradigm shift towards customised treatments that take into account each patient’s own genetic landscape [53].

4.1.2 Designing drugs based on individual molecular profiles

Molecular docking in conjunction with personalised medicine ushers in a new era of drug discovery wherein pharmaceutical interventions are finely customised according to individual molecular profiles. Using the abundance of data from omics technologies—genomics, transcriptomics, proteomics, and metabolomics, for example—this novel method creates medications that are precisely tailored to the distinctive features of each patient’s molecular landscape. As a computational powerhouse, molecular docking plays a pivotal role in this process by forecasting the interactions between medications and particular biological targets in individual patients. Drugs that are designed for maximum efficacy and lowest side effects can be more easily designed because to docking algorithms, which analyse the complex aspects of an individual’s genetic composition, gene expression patterns, and other molecular markers. Beyond the conventional one-size-fits-all approach, this customised medication design approach acknowledges the intrinsic differences in patients’ molecular profiles that affect therapeutic reactions. The algorithms provide a personalised drug development roadmap by predicting the most successful drug-receptor interactions based on genetic markers, pathway activities, and other biological aspects [54].

The convergence of molecular docking and personalised medicine holds great promise for revolutionary developments in drug design. Individual molecular profiles are used to build customised medications that have the potential to dramatically enhance treatment outcomes, reduce side effects, and usher in a new era of precision medicine in which pharmaceutical interventions are as distinct as the persons they are intended to treat [55].

4.2 Target identification and validation

4.2.1 Utilising molecular docking in target selection

Target identification and validation are critical stages in the molecular docking and personalised medicine integration process, where molecular docking techniques are essential for the deliberate selection of therapeutic targets. In order to forecast and assess the binding interactions between possible drug candidates and a wide range of molecular targets, this procedure makes use of computational methods. Molecular docking facilitates the methodical investigation of the wide-ranging biological terrain by reducing the number of pertinent targets selected according to parameters like binding affinity and specificity. Docking algorithms use in silico simulations to forecast the possible interactions between a drug candidate and different molecular targets. This allows for the prediction of potential success in modifying specific pathways or biomolecules linked to a given disease. This strategy is particularly useful in the personalised medicine setting, where accurate target selection is required due to individual differences in genetic and molecular profiles. Molecular docking aids in the discovery of targets that are in line with the distinct features of each patient’s illness phenotype by integrating patient-specific data [56].

In addition to streamlining the drug development process, target selection based on molecular docking guarantees that therapeutic interventions are precisely tailored to the unique molecular settings of individual patients. The combination of computational methods with personalised medicine holds the potential to improve drug development’s effectiveness and efficiency while paving the way for more specialised and efficient therapies [57].

4.2.2 Validating targets for personalised therapies

Molecular docking approaches support the crucial stage of target discovery and validation in the personalised medicine-molecular docking integration, helping to rigorously validate targets for customised therapeutics. Molecular docking is a computer method that predicts and analyses the binding interactions between possible drug candidates and selected targets, assisting in the validation of the appropriateness of these targets. Molecular docking refines target selection based on binding affinity, specificity, and pharmacological significance by evaluating the viability of drug-receptor interactions using virtual screening and simulation analysis. In the context of personalised medicine, this procedure is especially helpful since it ensures that targets are validated and identified based on the distinct genetic and molecular traits of individual patients [58].

A thorough understanding of the ways in which particular molecular variants contribute to the aetiology of disease is necessary for the validation of targets for personalised therapeutics. This procedure is made easier by molecular docking, which predicts how well drug candidates would bind to targets linked to specific patient profiles and provides information about the therapeutic intervention’s likelihood of success. Molecular docking and personalised medicine working together to validate targets not only expedites the conversion of genomic and omics data into workable treatment plans but also guarantees that customised treatments are supported by substantial scientific data. In the era of precision medicine, this confluence signifies a transformative approach where computational approaches are essential to the validation of targets, opening the door to more efficient and customised treatments [59].

4.3 Examples of personalised medicine success using molecular docking

A number of notable case studies highlight the effective fusion of molecular docking with personalised medicine, illustrating the revolutionary potential of this cooperative strategy in practical therapeutic contexts. A good example is the treatment of some cancers, where it is essential to find genetic alterations unique to each patient in order to customise treatment. Researchers can simulate the interactions between medications and altered proteins to anticipate which targeted therapies will work best by using molecular docking. Patients who have used this strategy have had amazing results, including higher response rates and less side effects [60].

Molecular docking-guided personalised therapy has shown promise in treating infectious illnesses like HIV. To create antiretroviral medications that precisely target distinct viral weaknesses, researchers use docking algorithms to account for individual variances in viral strains and host variables. Drug resistance has decreased and treatment efficacy has increased as a result of this customised approach. Furthermore, the combination of molecular docking and personalised medicine has demonstrated promise in the identification of medications that particularly target individual genetic and molecular variables leading to neurodegenerative illnesses like Alzheimer’s disease. These case studies highlight the observable benefits of molecular docking and personalised medicine, demonstrating how this combined approach is changing the face of healthcare by providing tailored, efficient, and focused treatments for a range of illnesses [61].

4.3.1 Impact on patient outcomes

Molecular docking in conjunction with personalised therapy has shown impressive effects on patient outcomes in a variety of medical contexts. Molecular docking simulations have been used to identify individual genetic profiles, which have been used to adapt cancer treatments in oncology. This approach has resulted in considerable improvements in response rates and a decrease in side effects. Individualised therapy recipients benefit from increased treatment efficacy, longer survival times, and general improvements in their quality of life. Treatment techniques for infectious diseases have been revolutionised by personalised approaches informed by molecular docking, especially in the context of antiretroviral therapy for HIV. With better viral suppression, decreased resistance to therapy, and a greater chance of obtaining undetectable viral levels, the targeted targeting of patient-specific viral strains has improved long-term patient outcomes [62].

The possibility for slowing the progression of neurodegenerative illnesses has been demonstrated by the merging of molecular docking with personalised treatment. Patients with diseases such as Alzheimer’s disease have reported improved functional outcomes and cognitive stabilisation when therapies are customised to each patient’s unique genetic and molecular variables. Together, these case studies highlight the real influence on patient outcomes and highlight the revolutionary potential of molecular docking-driven personalised therapy. This comprehensive strategy is improving therapeutic precision and improving overall well-being and prognosis of patients receiving personalised interventions by tailoring treatments to each patient’s unique genetic and molecular profile [63].

Advertisem*nt

5. Challenges and future directions

5.1 Ethical considerations

5.1.1 Privacy concerns in genomic data

Growing developments in personalised medicine, particularly with regard to privacy in the handling of genomic information, are brought to light by the combination of genomic data and molecular docking. Concerns over possible misuse or unauthorised access to sensitive information are raised by genomic data since it is intrinsically personal and uniquely identifying. Risk of genetic data breaches and its possible effects on persons is one of the main challenges. Because genomic data is so rich, getting it without authorization could jeopardise someone’s privacy and put them at risk of stigmatisation, discrimination, or improper use of their genetic information [64].

Furthermore, it is crucial to make sure that patient data is de-identified and anonymized because personalised medicine depends on the compilation and analysis of massive genetic databases. A difficult ethical conundrum arises when attempting to strike a balance between the value of shared data for research purposes and protecting individual privacy. Strong data governance frameworks, strict security measures, and open informed consent processes are necessary to address these issues. The future of personalised medicine will require continued communication between scientists, physicians, legislators, and the general public to develop moral guidelines that protect individuals’ privacy and promote progress in the field of genetic medicine. As the field develops, handling genomic data in a proactive and moral manner will be essential to guaranteeing the ethical and responsible advancement of personalised medicine [65].

5.1.2 Informed consent and patient autonomy

One of the most important ethical issues in the rapidly developing fields of genetic data integration and personalised medicine is ensuring patient autonomy and obtaining informed permission. Patients must have a thorough comprehension of genomic information due to its complexity before consenting to engage in research or have genomic testing done on them. Maintaining ethical standards requires making sure people are properly educated about the possible consequences, hazards, and advantages of sharing their genetic information. There are questions about how genomic data is dynamic and how our understanding of genetics changes over time. Respecting patient autonomy requires continuing to communicate with patients, giving them information, and getting their consent again as knowledge expands [66].

Furthermore, the difficulty is striking a balance between personal freedom and the group advantages of combined genetic data. Finding the correct balance entails being open and honest about the possible effects on society of genetic information sharing for research, encouraging a feeling of shared responsibility while honouring personal preferences. The informed consent process needs to be improved, educational programmes need to be implemented, and technology needs to be embraced for dynamic consent models in the future. Giving patients a comprehensive knowledge of the consequences of sharing their genetic information helps to maintain moral principles, build confidence, and promote the ethical development of personalised medicine [67].

5.2 Technical challenges

5.2.1 Integration of large-scale omics data

In the field of personalised medicine, large-scale omics data integration presents formidable technical obstacles. A number of obstacles need to be overcome before genomic, transcriptomic, proteomic, and metabolomic data can be effectively used in patient treatment, given their increasing volume and complexity. Creating reliable computational frameworks that can handle a variety of omics datasets and extract insightful information is a major issue. Advanced bioinformatics tools and algorithms that can reconcile various data types, take variability into account, and identify pertinent patterns are necessary for the integration of multi-dimensional data [68].

The requirement for scalable and interoperable platforms that enable smooth integration across various healthcare systems and research institutes represents another technical challenge. By standardising data formats and protocols, personalised medicine projects can be expanded in scope and depth by facilitating the sharing, aggregation, and collective analysis of omics data. Real-time omics data integration into clinical processes also necessitates advancements in data storage, processing speed, and secure sharing protocols. In addition to improving personalised medicine’s accuracy, resolving these technological issues will expedite the conversion of omics research results into practical knowledge for medical professionals. In order to pave the way for a more technically sound and clinically significant era in personalised healthcare, future directions in personalised medicine will necessitate concentrated efforts in creating state-of-the-art informatics infrastructure, encouraging collaboration between computational biologists and clinicians, and establishing standards for data sharing [69].

5.2.2 Validation of molecular docking predictions

Ensuring rigorous confirmation of computational predictions is a continuous technical problem in the integration of molecular docking and personalised medicine. In silico drug design can be greatly enhanced by molecular docking, however there are challenges in guaranteeing the precision and dependability of its predictions. In molecular docking, validation is evaluating how well computational models forecast the binding interactions seen in real-world environments. Sophisticated methods like nuclear magnetic resonance spectroscopy or X-ray crystallography are frequently needed for experimental validation in order to ascertain the true structure of ligand-receptor complexes [55].

The complexity of biological systems and the wide range of variables affecting molecular interactions present a challenge. Complexity in validation is increased by variations in protein conformations, effects of solvents, and precise depiction of binding sites. Creating benchmark datasets, which use known ligand-receptor complexes as a foundation for assessing docking predictions, is necessary to address this difficulty. Molecular docking simulations must be continuously improved in order to increase the predicted accuracy of scoring functions and algorithms. Moving forward, the scientific community will need to adopt standardised protocols for validation, leverage machine learning techniques to increase docking accuracy, and advance experimental approaches for high-throughput validation. It is imperative to overcome these technical obstacles in order to validate the accuracy of molecular docking predictions and guarantee their application in trustworthy personalised medicine [50].

5.3 Future prospects

5.3.1 Advancements in technology and methodology

Personalised medicine has a bright future ahead of it thanks to expected advances in technology and practice. With the ongoing increase in processing power, new approaches and technologies have the potential to completely transform the industry. Developments in machine learning and artificial intelligence (AI) will be crucial. It is anticipated that AI algorithms would improve molecular docking prediction accuracy, allowing for more accurate drug-receptor interaction simulations. The utilisation of machine learning models is expected to aid in the discernment of complex patterns present in extensive omics datasets, hence expediting the identification of new therapeutic targets and biomarkers. Understanding cellular heterogeneity will change dramatically with the introduction of single-cell omics technology. With the help of this high-resolution method, illness states and treatment outcomes can be more accurately described and individual cell behaviour can be revealed in a way never possible before [70].

Moreover, the amalgamation of multi-omics data, encompassing transcriptomics, proteomics, metabolomics, and genomes, would provide a comprehensive perspective of a person’s molecular terrain. Physicians will be able to create thorough profiles for individualised diagnosis and treatment plans thanks to platforms for advanced analytics and data integration. Technologies based on CRISPR-mediated gene editing show promise for accurate genetic interventions, enabling the correction of genetic abnormalities that underlie a variety of diseases. This ground-breaking skill creates opportunities for really customised therapeutic approaches. In conclusion, a revolutionary era where healthcare is progressively customised to each individual’s own features is being fostered by the synergistic fusion of cutting-edge technologies and creative techniques that characterise the future of personalised medicine [71].

5.3.2 Potential breakthroughs in personalised medicine and molecular docking

Molecular docking and personalised medicine have enormous potential for significant advances that could completely change the way that healthcare is provided. The combination of quantum computing with molecular docking simulations holds promise for a breakthrough. The speed and accuracy of docking predictions could be greatly increased by the unmatched computational power of quantum computers, opening up new avenues for drug design. The accuracy of molecular docking predictions could be improved by higher-resolution insights into protein structures that can be obtained using structural biology techniques like cryo-electron microscopy. This may result in the more accurate determination of the best drug-receptor combinations, particularly for difficult targets. There is tremendous promise in the combination of CRISPR-based gene editing technology and personalised medicine [72].

Accurate genetic adjustments directed by molecular docking predictions may open the door to customised therapeutic interventions at the genomic level, tackling the underlying causes of hereditary illnesses. Advanced omics technologies provide promise for the identification of new molecular signatures in the field of biomarker discovery. The combination of machine learning techniques and multi-omics data may reveal patterns that were not previously identified, leading to the identification of reliable biomarkers for prognosis, therapy response, and illness diagnosis. With these developments, the combination of molecular docking and personalised medicine is set to usher in a new era of healthcare that not only customises treatment to each patient’s unique molecular profile but also uses state-of-the-art technologies to precisely optimise therapeutic outcomes [73].

Advertisem*nt

6. Conclusion

Pharmacological discovery is undergoing a radical metamorphosis at the dynamic nexus of molecular docking and personalised medicine, paving the way for a future in which healthcare is more individually customised. Molecular docking has become a key component in the logical design of personalised medicines thanks to the convergence of genetic knowledge, cutting-edge omics technology, and computational power. Personalised medicine is important because it has the potential to completely transform patient care as we negotiate this new terrain. Molecular docking’s predictive capability combined with the capacity to interpret individual genetic variations allows clinicians to not only diagnose patients with unprecedented precision but also to tailor treatment strategies to each patient’s unique molecular subtleties. This revolution is enhanced by molecular docking, a computational ally that acts as a link between theoretical predictions and experimental validations. It speeds up the process of finding new drugs by pointing scientists in the direction of the best possible drug-receptor interactions with amazing effectiveness. The combination of artificial intelligence, large-scale omics data, and structural biology advances increases the likelihood of molecular disease knowledge and targeting advances. The possibilities for the future are exciting: possible developments in biomarker identification, CRISPR-based gene editing, and quantum computing might completely change the field of personalised medicine. The idea of customising medication discovery for individual patients is becoming increasingly real as these technologies advance, offering a new paradigm in healthcare where interventions are proactive rather than reactive, taking into account each patient’s own molecular composition. To sum up, the combination of molecular docking and personalised medicine is a bright light that points the way towards a future in which healthcare is highly predictive, genuinely personalised, and deeply affects each patient’s quality of life.

References

  1. 1. Moerenhout T, Devisch I, Cornelis GC. E-health beyond technology: Analyzing the paradigm shift that lies beneath. Medicine, Health Care and Philosophy. 2018;21:31-41
  2. 2. Kunduru AR. Machine learning in drug discovery: A comprehensive analysis of applications, challenges, and future directions. International Journal on Orange Technologies. 2023;5(8):29-37
  3. 3. Ginsburg GS, Willard HF. The foundations of genomic and personalized medicine. In: Essentials of Genomic and Personalized Medicine. Academic Press; 2010. pp. 1-10
  4. 4. Khare N, Maheshwari SK, Rizvi SM, Albadrani HM, Alsagaby SA, Alturaiki W, et al. hom*ology modelling, molecular docking and molecular dynamics simulation studies of CALMH1 against secondary metabolites of bauhinia variegata to treat Alzheimer’s disease. Brain Sciences. 2022;12(6):770
  5. 5. Ahmed Z, Mohamed K, Zeeshan S, Dong X. Artificial intelligence with multi-functional machine learning platform development for better healthcare and precision medicine. Database. 2020;2020:baaa010
  6. 6. Sharma G. The human genome project and its promise. Journal of Indian College of Cardiology. 2012;2(1):1-3
  7. 7. Pirmohamed M. Personalized pharmacogenomics: Predicting efficacy and adverse drug reactions. Annual Review of Genomics and Human Genetics. 2014;15:349-370
  8. 8. Nassar SF, Raddassi K, Ubhi B, Doktorski J, Abulaban A. Precision medicine: Steps along the road to combat human cancer. Cell. 2020;9(9):2056
  9. 9. Redekop WK, Mladsi D. The faces of personalized medicine: A framework for understanding its meaning and scope. Value in Health. 2013;16(6):S4-S9
  10. 10. Teama S. DNA polymorphisms: DNA-based molecular markers and their application in medicine. In: Genetic Diversity and Disease Susceptibility. 2018. pp. 25-32
  11. 11. Kendler KS, Aggen SH, Czajkowski N, Røysamb E, Tambs K, Torgersen S, et al. The structure of genetic and environmental risk factors for DSM-IV personality disorders: A multivariate twin study. Archives of General Psychiatry. 2008;65(12):1438-1446
  12. 12. Ozomaro U, Wahlestedt C, Nemeroff CB. Personalized medicine in psychiatry: Problems and promises. BMC Medicine. 2013;11(1):1-35
  13. 13. Hasanzad M, Sarhangi N, Ehsani Chimeh S, Ayati N, Afzali M, Khatami F, et al. Precision medicine journey through omics approach. Journal of Diabetes & Metabolic Disorders. 2022;21(1):881-888
  14. 14. Zhang J, Chiodini R, Badr A, Zhang G. The impact of next-generation sequencing on genomics. Journal of Genetics and Genomics. 2011;38(3):95-109
  15. 15. LaFramboise T. Single nucleotide polymorphism arrays: A decade of biological, computational and technological advances. Nucleic Acids Research. 2009;37(13):4181-4193
  16. 16. Weinshilboum RM, Wang L. Pharmacogenomics: Precision medicine and drug response. In: Mayo Clinic Proceedings. Vol. 92, no. 11. Elsevier; 2017. pp. 1711-1722
  17. 17. Berliner JL, Cummings SA, Boldt Burnett B, Ricker CN. Risk assessment and genetic counseling for hereditary breast and ovarian cancer syndromes—Practice resource of the National Society of genetic Counselors. Journal of Genetic Counseling. 2021;30(2):342-360
  18. 18. Veenstra TD. Omics in systems biology: Current progress and future outlook. Proteomics. 2021;21(3-4):2000235
  19. 19. Garg M. RNA sequencing: A revolutionary tool for transcriptomics. In: Advances in Animal Genomics. Academic Press; 2021. pp. 61-73
  20. 20. Tsimberidou AM, Fountzilas E, Bleris L, Kurzrock R. Transcriptomics and solid tumors: The next frontier in precision cancer medicine. In: Seminars in Cancer Biology. Vol. 84. Academic Press; 2022. pp. 50-59
  21. 21. Das PP, Rana S, Muthamilarasan M, Kannan M, Ghazi IA. Omics approaches for understanding plant Defense response. Omics Technologies for Sustainable Agriculture and Global Food Security. 2021;1:41-83
  22. 22. Su M, Zhang Z, Zhou L, Han C, Huang C, Nice EC. Proteomics, personalized medicine and cancer. Cancers. 2021;13(11):2512
  23. 23. Manzoni C, Kia DA, Vandrovcova J, Hardy J, Wood NW, Lewis PA, et al. Genome, transcriptome and proteome: The rise of omics data and their integration in biomedical sciences. Briefings in Bioinformatics. 2018;19(2):286-302
  24. 24. Dai X, Shen L. Advances and trends in omics technology development. Frontiers in Medicine. 2022;9:911861
  25. 25. Segers K, Declerck S, Mangelings D, Heyden YV, Eeckhaut AV. Analytical techniques for metabolomic studies: A review. Bioanalysis. 2019;11(24):2297-2318
  26. 26. Singh S, Sarma DK, Verma V, Nagpal R, Kumar M. Unveiling the future of metabolic medicine: Omics technologies driving personalized solutions for precision treatment of metabolic disorders. Biochemical and Biophysical Research Communications. 2023;682(1):1-20
  27. 27. Angione C. Human systems biology and metabolic modelling: A review—From disease metabolism to precision medicine. BioMed Research International. 2019:1-19
  28. 28. Qoronfleh MW, Chouchane L, Mifsud B, Al Emadi M, Ismail S. THE FUTURE OF MEDICINE, healthcare innovation through precision medicine: Policy case study of Qatar. Life Sciences, Society and Policy. 2020;16(1):1-20
  29. 29. La Cognata V, Morello G, Cavallaro S. Omics data and their integrative analysis to support stratified medicine in neurodegenerative diseases. International Journal of Molecular Sciences. 2021;22(9):4820
  30. 30. Giordano S, Petrelli A. From single-to multi-target drugs in cancer therapy: When aspecificity becomes an advantage. Current Medicinal Chemistry. 2008;15(5):422-432
  31. 31. Pokorska-Bocci A, Stewart A, Sagoo GS, Hall A, Kroese M, Burton H. 'Personalized medicine': what’s in a name? Personalized Medicine. 2014;11(2):197-210
  32. 32. Seymour CW, Gomez H, Chang CC, Clermont G, Kellum JA, Kennedy J, et al. Precision medicine for all? Challenges and opportunities for a precision medicine approach to critical illness. Critical Care. 2017;21:1-1
  33. 33. Mastrangelo A, Armitage G, E, García A, Barbas C. Metabolomics as a tool for drug discovery and personalised medicine. A review. Current Topics in Medicinal Chemistry. 2014;14(23):2627-2636
  34. 34. Seyhan AA, Carini C. Are innovation and new technologies in precision medicine paving a new era in patients centric care? Journal of Translational Medicine. 2019;17:1-28
  35. 35. Abdolmaleki A, Ghasemi JB, Ghasemi F. Computer aided drug design for multi-target drug design: SAR/QSAR, molecular docking and pharmacophore methods. Current Drug Targets. 2017;18(5):556-575
  36. 36. Sivakumar KC, Haixiao J, Naman CB, Sajeevan TP. Prospects of multitarget drug designing strategies by linking molecular docking and molecular dynamics to explore the protein–ligand recognition process. Drug Development Research. 2020;81(6):685-699
  37. 37. Adelusi TI, Oyedele AQ , Boyenle ID, Ogunlana AT, Adeyemi RO, Ukachi CD, et al. Molecular modeling in drug discovery. Informatics in Medicine Unlocked. 2022;29:100880
  38. 38. Khare N, Maheshwari SK, Jha AK. Screening and identification of secondary metabolites in the bark of Bauhinia variegata to treat Alzheimer’s disease by using molecular docking and molecular dynamics simulations. Journal of Biomolecular Structure and Dynamics. 2021;39(16):5988-5998
  39. 39. Parween A, Singh PK, Anamika Y, Khare N, Rai NP, Bajpai M, et al. Molecular docking of quinolone against INHA to treat tuberculosis. International Journal for Research in Applied Science and Engineering Technology. 2020;8(6):2421-2427
  40. 40. Naqvi AA, Mohammad T, Hasan GM, Hassan MI. Advancements in docking and molecular dynamics simulations towards ligand-receptor interactions and structure-function relationships. Current Topics in Medicinal Chemistry. 2018;18(20):1755-1768
  41. 41. Ramírez D. Computational methods applied to rational drug design. The Open Medicinal Chemistry Journal. 2016;10:7
  42. 42. Mohan A, Krishnamoorthy S, Sabanayagam R, Schwenk G, Feng E, Ji HF, et al. Pharmacophore based virtual screening for identification of effective inhibitors to combat HPV 16 E6 driven cervical cancer. European Journal of Pharmacology. 2023;957:175961
  43. 43. Vázquez J, López M, Gibert E, Herrero E, Luque FJ. Merging ligand-based and structure-based methods in drug discovery: An overview of combined virtual screening approaches. Molecules. 2020;25(20):4723
  44. 44. Lee S, Barron MG. Development of 3D-QSAR model for acetylcholinesterase inhibitors using a combination of fingerprint, molecular docking, and structure-based pharmacophore approaches. Toxicological Sciences. 2015;148(1):60-70
  45. 45. Ballante F, Kooistra AJ, Kampen S, de Graaf C, Carlsson J. Structure-based virtual screening for ligands of G protein–coupled receptors: What can molecular docking do for you? Pharmacological Reviews. 2021;73(4):1698-1736
  46. 46. Di Filippo JI, Cavasotto CN. Guided structure-based ligand identification and design via artificial intelligence modeling. Expert Opinion on Drug Discovery. 2022;17(1):71-78
  47. 47. Lionta E, Spyrou G, Vassilatis K, D, Cournia Z. Structure-based virtual screening for drug discovery: Principles, applications and recent advances. Current Topics in Medicinal Chemistry. 2014;14(16):1923-1938
  48. 48. Sethi A, Joshi K, Sasikala K, Alvala M. Molecular docking in modern drug discovery: Principles and recent applications. Drug Discovery and Development-New Advances. 2019;2:1-21
  49. 49. Wang T, Wu MB, Lin JP, Yang LR. Quantitative structure–activity relationship: Promising advances in drug discovery platforms. Expert Opinion on Drug Discovery. 2015;10(12):1283-1300
  50. 50. Saikia S, Bordoloi M. Molecular docking: Challenges, advances and its use in drug discovery perspective. Current Drug Targets. 2019;20(5):501-521
  51. 51. Aouidate A, Ghaleb A, Ghamali M, Chtita S, Ousaa A, Choukrad MB, et al. Furanone derivatives as new inhibitors of CDC7 kinase: Development of structure activity relationship model using 3D QSAR, molecular docking, and in silico ADMET. Structural Chemistry. 2018;29:1031-1043
  52. 52. Sneha P, Doss CG. Molecular dynamics: New frontier in personalized medicine. Advances in Protein Chemistry and Structural Biology. 2016;102:181-224
  53. 53. Gayathiri E, Prakash P, Kumaravel P, Jayaprakash J, Ragunathan MG, Sankar S, et al. Computational approaches for modeling and structural design of biological systems: A comprehensive review. Progress in Biophysics and Molecular Biology. 2023;285(1):17-32
  54. 54. Waheed S, Li Z, Zhang F, Chiarini A, Armato U, Wu J. Engineering nano-drug biointerface to overcome biological barriers toward precision drug delivery. Journal of Nanobiotechnology. 2022;20(1):395
  55. 55. Moingeon P, Kuenemann M, Guedj M. Artificial intelligence-enhanced drug design and development: Toward a computational precision medicine. Drug Discovery Today. 2022;27(1):215-222
  56. 56. Agu PC, Afiukwa CA, Orji OU, Ezeh EM, Ofoke IH, Ogbu CO, et al. Molecular docking as a tool for the discovery of molecular targets of nutraceuticals in diseases management. Scientific Reports. 2023;13(1):13398
  57. 57. Ivasiv V, Albertini C, Gonçalves AE, Rossi M, Bolognesi ML. Molecular hybridization as a tool for designing multitarget drug candidates for complex diseases. Current Topics in Medicinal Chemistry. 2019;19(19):1694-1711
  58. 58. Özenver N, Efferth T. Integration of Phytochemicals and Phytotherapy into Cancer Precision Medicine. Approaching Complex Diseases: Network-Based Pharmacology and Systems Approach in Bio-Medicine; 2020. pp. 355-392
  59. 59. Hopkins CE, Brock T, Caulfield TR, Bainbridge M. Phenotypic screening models for rapid diagnosis of genetic variants and discovery of personalized therapeutics. Molecular Aspects of Medicine. 2023;91:101153
  60. 60. Hassan M, Awan FM, Naz A, deAndrés-Galiana EJ, Alvarez O, Cernea A, et al. Innovations in genomics and big data analytics for personalized medicine and health care: A review. International Journal of Molecular Sciences. 2022;23(9):4645
  61. 61. Melge AR, Kumar LG, Pavithran K, Nair SV, Manzoor K. Predictive models for designing potent tyrosine kinase inhibitors in chronic myeloid leukemia for understanding its molecular mechanism of resistance by molecular docking and dynamics simulations. Journal of Biomolecular Structure and Dynamics. 2019;37(18):4747-4766
  62. 62. Husnain A, Rasool S, Saeed A, Hussain HK. Revolutionizing pharmaceutical research: Harnessing machine learning for a paradigm shift in drug discovery. International Journal of Multidisciplinary Sciences and Arts. 2023;2(2):149-157
  63. 63. Gnanaraj C, Sekar M, Fuloria S, Swain SS, Gan SH, Chidambaram K, et al. In silico molecular docking analysis of karanjin against alzheimer’s and parkinson’s diseases as a potential natural lead molecule for new drug design, development and therapy. Molecules. 2022;27(9):2834
  64. 64. Najafi A, Emami N, Samad-Soltani T. Integration of genomics data and electronic health records toward personalized medicine: A targeted review. Frontiers in Health Informatics. 2021;10(1):86
  65. 65. Hulsen T, Jamuar SS, Moody AR, Karnes JH, Varga O, Hedensted S, et al. From big data to precision medicine. Frontiers in Medicine. 2019;6:34
  66. 66. Ormond KE, Cho MK. Translating personalized medicine using new genetic technologies in clinical practice: The ethical issues. Personalized Medicine. 2014;11(2):211-222
  67. 67. Bruynseels K, Santoni de Sio F, Van den Hoven J. Digital twins in health care: Ethical implications of an emerging engineering paradigm. Frontiers in Genetics. 2018;9:31
  68. 68. Grapov D, Fahrmann J, Wanichthanarak K, Khoomrung S. Rise of deep learning for genomic, proteomic, and metabolomic data integration in precision medicine. Omics: A Journal of Integrative Biology. 2018;22(10):630-636
  69. 69. Martinez-Garcia M, Hernández-Lemus E. Data integration challenges for machine learning in precision medicine. Frontiers in Medicine. 2022;8:784455
  70. 70. Arabi AA. Artificial intelligence in drug design: Algorithms, applications, challenges and ethics. Future. Drug Discovery. 2021;3(2):FDD59
  71. 71. Bhattacharjee G, Gohil N, Khambhati K, Mani I, Maurya R, Karapurkar JK, et al. Current approaches in CRISPR-Cas9 mediated gene editing for biomedical and therapeutic applications. Journal of Controlled Release. 2022;343:703-723
  72. 72. Auffray C, Chen Z, Hood L. Systems medicine: The future of medical genomics and healthcare. Genome Medicine. 2009;1:1-1
  73. 73. Strianese O, Rizzo F, Ciccarelli M, Galasso G, D’Agostino Y, Salvati A, et al. Precision and personalized medicine: How genomic approach improves the management of cardiovascular and neurodegenerative disease. Genes. 2020;11(7):747

Written By

Noopur Khare and Pragati Khare

Submitted: 24 December 2023 Reviewed: 05 January 2024 Published: 29 April 2024

© The Author(s). Licensee IntechOpen. This chapter is distributed under the terms of the Creative Commons Attribution 3.0 License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Personalised Medicine and Molecular Docking: Tailoring Drug Discovery for Individual Patients (2024)

FAQs

What is the tailoring of medical treatment to the individual characteristics of each patient? ›

Personalized medicine, also referred to as precision medicine, is a medical model that separates people into different groups—with medical decisions, practices, interventions and/or products being tailored to the individual patient based on their predicted response or risk of disease.

How can genetic information be used to tailor medical treatments for individual patients? ›

Our genes, or DNA, provide the blueprint for our bodies and the things that make us unique. In some cases, doctors can use this genetic information to help pick which medicines or doses of medicines may be safer or work better for a person. This helps people to get the right medicine at the right dose for them.

What are the challenges of tailoring medicine to individuals or small numbers of people? ›

The major obstacles for making personalized medicine available to patients requires basic, translational, and also regulatory science, especially in the areas of ethical, legal, and social issues (3).

What is the conclusion of personalized medicine? ›

In conclusion, personalized medicine, often referred to as precision medicine, offers a revolutionary approach to patient care by considering individual characteristics, from genomics to behavior, to customize interventions and treatments accordingly.

Is tailoring treatment to the medical characteristics of the individual patient? ›

Precision medicine, also known as personalized medicine, is a cutting-edge approach that takes into account a patient's unique genetic makeup, lifestyle, and environmental factors to create targeted and effective treatment plans.

What do we mean by tailoring of medical information during clinical interactions? ›

Tailoring information is often promoted to attune to individual differences. Clarity is needed regarding what tailoring during clinical visits actually means. Clinicians can tailor information based on what they assume to know about the patient. Preferably, they also tailor on what becomes known during the interaction.

Does insurance cover pharmacogenetic testing? ›

Depending on your policy and reasons for testing, some insurance companies may or may not cover pharmacogenomic testing. Contact your insurance provider about coverage prior to testing if this is a concern.

How can treatments be tailored to individuals? ›

Minimized Adverse Effects: By understanding a patient's genetic makeup, personalized treatment plans can identify potential risks or adverse reactions to certain medications. This knowledge enables healthcare providers to select alternative treatments or adjust dosages, minimizing the likelihood of adverse effects.

Is GeneSight testing legit? ›

The GeneSight test is not FDA approved. This means that the FDA has not done its own review of the product and found it to be successful at doing what it claims to do.

What is a drawback to personalized medicine? ›

Genetic testing and other personalized treatments can be expensive, making it difficult for some patients to access these treatments. Limited access: Precision medicine is not yet widely available, and not all doctors are trained to provide personalized treatments.

What are the ethical issues in personalized medicine? ›

There were eight main themes emerging from the point of view of the patients regarding ethical concerns and risks of precision medicine: privacy and security of patient data, economic impact on the patients, possible harms of precision medicine including psychosocial harms, risk for discrimination of certain groups, ...

What is the field that is tailoring medical treatments? ›

Pharmacogenomics is an important example of the field of precision medicine, which aims to tailor medical treatment to each person or to a group of people.

What is the ultimate goal of personalized medicine? ›

Personalized medicine, because it is based on each patient's unique genetic makeup, is beginning to overcome the limitations of traditional medicine. Increasingly it is allowing health care providers to: shift the emphasis in medicine from reaction to prevention. predict susceptibility to disease.

What are the challenges of personalized medicine? ›

Challenges for precision medicine

The anonymisation of data and its security is a big issue. So, ethical challenges are paramount. Noise in data is another distraction. Turnaround time for data analysis is at the earliest 26 h which is still slow for decisions in acute care settings.

What are the best examples of personalized medicine? ›

Examples of personalized medicine include the targeted therapies indicated for the treatment of human epidermal growth factor receptor 2–positive breast cancer and tumor marker testing to diagnose cancer.

What is tailoring in medical terms? ›

Tailored medicine also may be called “precision medicine,” “personalized medicine,” and sometimes “genomic medicine.” It uses your genetic information, lifestyle, and environment to create (or tailor) a treatment plan for your illness.

What does tailored mean in healthcare? ›

Listen to pronunciation. (TAY-lurd IN-ter-VEN-shun) The use of communication, drugs, or other types of treatments that are specific for an individual or a group to improve health or change behavior.

What is patient tailored therapies? ›

By giving greater attention to patient preferences and quality of life, we can tailor our approach in a way that reduces the burdens of cancer therapy while still providing effective care.

What is tailoring medicine? ›

Personalized medicine is the tailoring of medical treatment to the individual characteristics of each patient. The approach relies on scientific breakthroughs in our understanding of how a person's unique molecular and genetic profile makes them susceptible to certain diseases.

References

Top Articles
Latest Posts
Article information

Author: Carmelo Roob

Last Updated:

Views: 6574

Rating: 4.4 / 5 (65 voted)

Reviews: 88% of readers found this page helpful

Author information

Name: Carmelo Roob

Birthday: 1995-01-09

Address: Apt. 915 481 Sipes Cliff, New Gonzalobury, CO 80176

Phone: +6773780339780

Job: Sales Executive

Hobby: Gaming, Jogging, Rugby, Video gaming, Handball, Ice skating, Web surfing

Introduction: My name is Carmelo Roob, I am a modern, handsome, delightful, comfortable, attractive, vast, good person who loves writing and wants to share my knowledge and understanding with you.