View Article

Abstract

One important element affecting the pharmacokinetics and pharmacodynamics of medicinal drugs is protein binding. The basic processes of protein binding are examined in this overview, along with how medications and plasma proteins like albumin and alpha-1 acid glycoprotein interact. We look at how drug distribution, metabolism, and excretion are affected by protein binding, emphasizing how important it is in dictating the free drug concentration that can be used for pharmacological effect. Changes in protein binding brought about by pathogenic, physiological, or drug-related variables can drastically affect risk profiles and treatment outcomes. The therapeutic consequences of protein binding are reviewed in the review, along with how it affects drug interactions, how much to dose certain groups, and how to formulate drugs. This review emphasizes the significance of protein binding in enhancing drug discovery and customized treatment by fusing knowledge from basic and clinical pharmacology. To maximize the benefits of medications, reduce side effects, and provide better patient care overall, it is imperative to comprehend these dynamics.

Keywords

Protien Binding, Interactions, Memberane, Binding Sites

Introduction

Protein binding is a pivotal concept in pharmacokinetics, influencing the behavior and efficacy of therapeutic agents within the body. This phenomenon, wherein drugs interact with plasma proteins such as albumin and alpha-1-acid glycoprotein, plays a critical role in determining the pharmacological and toxicological outcomes of medications. Understanding protein binding is essential for optimizing drug development, improving clinical efficacy, and minimizing adverse effects. Drug molecules interact with various proteins in the bloodstream, primarily albumin and alpha-1 acid glycoprotein (AAG), influencing their pharmacokinetic profiles. Protein binding affects drug distribution, free drug concentration, and ultimately therapeutic outcomes. This review aims to provide a comprehensive examination of protein binding mechanisms, its effects on pharmacokinetics, and its clinical implications. By consolidating current knowledge and research findings, this review will enhance our understanding of how protein binding influences drug development and therapeutic outcomes, guiding more informed clinical decision-making and advancing the field of pharmacology. The interaction between medications and plasma proteins, specifically alpha-1-acid glycoprotein and albumin, is known as protein binding21. These proteins carry medications throughout the body in a carrier capacity.

Types of Protein Binding

  • Reversible Binding12: Most drugs bind reversibly to plasma proteins, primarily albumin. This binding is non-covalent21, involving hydrophobic interactions, hydrogen bonds, and electrostatic forces.
  • Irreversible Binding: Some drugs form covalent bonds with proteins, though this is less common and often associated with adverse effects.

Importance for Drug Metabolism: Protein binding influences how medications are absorbed and excreted from the body. The only drug molecules that have the ability to exhibit therapeutic effects or be digested and removed are those that are free, or unbound, molecules. Bound drug molecules are not able to pass through cell membranes to get to their intended locations and are momentarily dormant. 1

EFFECT ON DRUG DELIVERY KINETICS: Many pharmacokinetic parameters are influenced by the degree of protein binding, including:

 

  • Distribution17:

Because highly protein-bound medications are mostly restricted to the vascular compartment rather than the circulation, they typically have a wider volume of distribution compared to entering tissues.

  • Half-life:

A drug's half-life may be impacted by protein binding. Because they are released from protein binding sites and eliminated from the body more slowly, drugs that are heavily bound to proteins have longer half-lives.

  • Bioavailability:

A drug's bioavailability is impacted by protein binding. The only medication that can get past biological barriers and get to its target is the free drug. The amount of free medication that is accessible for distribution and metabolism can change due to modifications in protein binding.17

CLINICAL IMPLICATIONS

 Knowledge of protein binding is essential for;

  1. Deciding on the right dosage schedule for medications, particularly those with limited therapeutic windows.
  2. Forecasting interactions between drugs. Increased free drug concentrations and potential side effects can result from medications that compete for the same protein binding sites.

Medications that vie for the same protein binding sites may cause higher quantities of the free drug, as well as possible toxicity or reduced effectiveness. Developing treatment plans for patients whose protein binding has been disrupted by age-related alterations or illness conditions (such hypoalbuminemia).

THE MAIN PROCESSES OF PROTEIN BINDING ARE AS FOLLOWS:

Hydrophobic Relationships:

  1. A lot of medications have a lipophilic (loving fat) nature and can attach to hydrophobic pockets found on plasma proteins, especially albumin.2
  2. Hydrophobic interactions occur when non-polar drug molecule regions engage with non-polar protein surface amino acid residues (like leucine and phenylalanine).
  3. Because of the relative weakness and reversibility of these interactions, the medication is able to separate from the protein and still have its pharmacological effects.

 Electrostatic Interactions: 

  1. Charged groups on the drug molecule and oppositely charged amino acid residues on plasma proteins interact electrostatically2.
  2. For instance, medications with basic or acidic functional groups may interact with residues on albumin or alpha-1-acid glycoprotein that are negatively or positively charged.
  3. The stability and binding affinity of the drug-protein complex may be impacted by these interactions, which have the potential to be stronger than hydrophobic interactions.

Hydrogen Bonding:

Hydrogen bond donors or acceptors on the surface of proteins and hydrogen atoms connected to electronegative atoms (such as oxygen or nitrogen) in the drug molecule form a hydrogen bond .This kind of interaction may strengthen and increase the specificity of drug-protein binding .Although hydrogen bonds are often weaker than covalent connections, they are nonetheless very important for keeping the drug-protein combination stable.

Van der Waals Forces:

Because of temporary dipoles, molecules experience weak attractive forces known as Van der Waals forces. Through the optimization of the complimentary surface contacts, these forces contribute to the overall binding affinity between medicines and plasma proteins.

EFFECT ON THE AVAILABILITY AND DISTRIBUTION OF DRUGS:

  1. Distribution17:

Protein binding affects how medications are distributed among the many bodily parts. In contrast to fewer protein-bound medications, highly protein-bound 3pharmaceuticals have a greater volume of distribution within the vascular space and are more likely to stay in the bloodstream.

  1. Availability:

The only portion of a medication that may disperse to tissues, pass through cell membranes, and have therapeutic effects is the unbound (free) fraction. The amount of free medication that is available for distribution and metabolism is directly impacted by the degree of protein binding.3

  1. Half-life:

Because they are released from protein binding sites and eliminated from the body more slowly, drugs that are heavily bound to plasma proteins frequently have longer half-lives.

  1. Drug Interactions:

The amount of free drug in the circulation can be impacted by competition between medications for protein binding sites or by endogenous chemicals (such fatty acids). Potential drug-drug interactions5 or changed pharmacological effects may result from this competition.

The following are the main variables that affect protein binding:

1. Structure of Drugs:

Lipophilicity11:

Because albumin3 includes hydrophobic binding sites, drugs with a higher lipophilicity likely to bind to plasma proteins more extensively.

Molecular Size:

Although the accessibility and selectivity of these sites can vary, larger molecules may have more possible binding sites on proteins.

Functional Groups:

Certain functional groups, such as basic or acidic groups, might affect binding affinity by influencing electrostatic interactions with charged residues on proteins.

2. Concentrations of Plasma Proteins:

Albumin Concentration:

The primary binding protein in plasma is albumin11. A change in albumin level (such as hypoalbuminemia in liver illness) might affect a medication's ability to bind, which can modify the concentration of the free drug.

Alpha-1-Acid Glycoprotein (AAG):

AAG3 is an acute-phase reactant, which means that it can increase in concentration during stress or inflammation and influence the way that certain basic medicines interact to proteins.

 3. Aspects Physiological:

pH:

Variations in pH can impact a drug's ionization state as well as its propensity for binding to plasma proteins. For example, in alkaline pH11 circumstances, acidic medicines bind more strongly to albumin.

Temperature:

Although it is less clinically significant than other factors6, temperature variations can affect binding interactions and change the structure of proteins.

Disease States:

Protein concentrations and binding capabilities can be impacted by pathophysiological circumstances like hepatic or renal dysfunction, which can change how drugs are distributed.

4. Competition for Binding Sites:

Medications with comparable binding sites on proteins11 may vie for those sites, which could result in displacement and higher amounts of free drugs. This may change the toxicity or effectiveness of the medication. Endogenous compounds that compete with pharmaceuticals for protein binding sites include bilirubin and fatty acids, which can affect the pharmacokinetics3 of medications.

5. Genetic Variability:

 Proteins with genetic polymorphisms, such as albumin or AAG3, may have different binding affinities to different medications, which can cause inter-individual differences in toxicity and treatment response.

6. Binding Assay Conditions:

 The stated binding5 affinity values can be impacted by variables pertaining to the assay technique employed to quantify protein binding, such as temperature, buffer composition, and protein purity11. A variety of analytical approaches and procedures are used to quantify and describe protein binding. Here is a few popular techniques: 2

1. Equilibrium dialysis:

Principle:

Equilibrium dialysis2,15 allows drug molecules to become free (unbound) and bound (bound) by allowing them to cross a semipermeable membrane and find equilibrium.

Process:

A semipermeable membrane7-separated drug-protein combination is inserted into one dialysis cell compartment. Free drug molecules gradually permeate through the membrane and enter a different compartment16 that holds a buffer free of drugs. After thereafter, measurements are made to ascertain the amount of free drug present in each compartment. Understanding how medications interact with plasma proteins and how these interactions affect pharmacokinetics and pharmacodynamics7 is crucial to pharmacology and pharmaceutical development. This can be achieved through studying protein binding. Numerous analytical approaches and strategies are used to measure.

Advantages:

Because it can provide equilibrium conditions, this approach is regarded as the gold standard for protein binding investigations.

Requires meticulous experimental setup and validation; takes a lot of time.



       
            Screenshot 2024-08-18 125100.png
       

    


2.Centrifugal filtration, or ultrafiltration7:

Principle:

Centrifugation15 is used in ultrafiltration2 to separate free drug molecules from drug molecules bound to proteins according to their molecular sizes.

Method:

A drug-protein combination is run across a membrane that has a predetermined cut off molecular weight. Drug molecules that are free to flow through the membrane are separated from bigger complexes of drug molecules that are attached to proteins. Next, the amounts of bound and free drug in the retentate and filtrate are calculated, respectively.

Benefits:

This approach is quick and appropriate for high-throughput screening. Can handle tiny sample volumes.

Limitations:

Equilibrium dialysis1 may not be reached in its true state demands that the experimental setup and membrane characteristics be carefully considered.

3. Chromatographic techniques15, such as HPLC:

Basic Principle:

The difference in affinity between a stationary phase (such as column packing material) and a mobile phase (such as solvent) is the basis for the separation and quantification of free and bound drug molecules using chromatographic procedures.7 

Method:

A chromatographic2 column is injected with a drug-protein combination. Free drug molecules elute at different periods from protein-bound drug molecules because they have distinct affinities for the stationary phase. The concentration of both free and bound drug is shown by the region beneath the chromatographic peaks.3

Benefits:

Provides sensitivity and specificity for measuring drug fractions that are both free and bound can offer further details about drug interactions and metabolites. Requires calibration using established standards and chromatographic condition optimization. It might not be possible to test binding affinity directly.

4. UFAE15 or Ultrafast Affinity Extraction:

Principle:

Rapid equilibrium dialysis, followed by rapid ultrafiltration, is the basis of the more recent UFAE2 method, which measures protein binding.

Procedure:

Ultrafiltration7 is used to separate the drug that is bound and the free drug after the drug-protein mixture has been quickly equilibrated using dialysis. This technique combines the speed of ultrafiltration with the benefits of equilibrium dialysis, or conditions of equilibrium.

Benefits:

Offers quick and precise protein binding measurement.  Suitable for screening at high throughput

Restrictions:

Compared to conventional methods, the availability of equipment and methods may be restricted.

5. Alternative Methods:

Gel Filtration Chromatography:

Uses size exclusion principles to separate bound and free drug.

Dialysis Tubing15:

Semipermeable tubing is used in place of a dialysis cell in equilibrium dialysis, which is comparable.

Nuclear Magnetic Resonance (NMR)2,15: Used less frequently for quantitative binding investigations, although it offers structural information regarding drug-protein interactions.

Every method has advantages and disadvantages, and the best approach will rely on a number of variables, including the desired throughput, the amount of detail needed in the binding study, and the unique properties of the drug and protein.

Drugs' pharmacokinetic and pharmacodynamic 7characteristics are largely determined by protein binding, which also affects the drugs' safety, effectiveness, dosage recommendations, and possible interactions with other medications in therapeutic4 contexts. Here's how various features are affected by protein binding:

1.The effectiveness of drugs:

 Distribution and Target Site Availability11:

A drug is only pharmacologically active when it is free (unbound) and can penetrate biological barriers (such as the blood-brain barrier) to reach target tissues. Highly protein-bound medications may have a lower percentage of free medication available for therapeutic action4, which could have an impact on effectiveness.

Duration of Action:

Because they bind to proteins extensively, drugs with longer half-lives may have therapeutic effects that last longer and require fewer doses.

2. Drug Safety:

 Hazard of Toxicity:

Protein binding influences a drug's pharmacokinetics, or how it is distributed and eliminated from the body. A higher percentage of the drug may be bound and not easily removed if drug-protein binding is high, which raises the risk of drug build up and toxicity.

 

Therapeutic Index:

Drugs that have a narrow therapeutic index6—that is, a small range of doses that are both effective and toxic—are more vulnerable to modifications in protein binding because these changes can cause changes in the amounts of free drug, which could tip the scales in favor of toxicity11.

3.Dose Regimens5:

Individual Variation:

The ideal dose regimen for patients may be influenced by inter-individual variability in protein binding (caused, for example, by hereditary variables or illness conditions). Dosing modifications may be necessary for individuals with low plasma protein levels (such as hypoalbuminemia) in order to maximize therapeutic efficacy11 and minimize harm.

 Monitoring and Adjustments:

Physicians can more effectively adjust dosage schedules for patients depending on renal or hepatic function, age, and co-occurring drugs that might compete for protein binding sites by having a better understanding of protein binding qualities. 5

4. Drug-Drug Interactions:

Competition for Protein Binding Sites4: Medicines that share plasma protein binding sites, such as albumin, may vie for these sites, changing how each drug is distributed. Because of this competition, one or both medications may have higher free concentrations9, which could have beneficial effects but also raise toxicity.

Clinical Implications:

When prescribing a patient more than one medication, clinicians need to take into account the possibility of drug-drug interactions caused by protein binding4. To lessen these interactions, it could be essential to keep an eye on free drug concentrations or change dosages.5

 5. Clinical Illustrations:

 

Warfarin with non-steroidal anti-inflammatory medicines (NSAIDs)3:

Warfarin is an anticoagulant that is heavily attached to albumin. It can interact with NSAIDs, which also bind to albumin, raising the risk of bleeding because of displacement and increased warfarin free concentrations4.

Phenytoin with Valproic Acid:

Phenytoin11 is a highly protein-bound epilepsy medication. The displacement of phenytoin from protein binding sites caused by co-administration of valproic acid may result in elevated levels of free phenytoin and possible toxicity.  Protein binding is often a critical factor in the pharmacokinetics and pharmacodynamics5 of drug candidates, influencing their efficacy, safety, and ultimately their success or failure in clinical development. Here are some specific examples and case studies where protein binding played a significant role17:

1.         Aripiprazole (Abilify):

o          Background: Aripiprazole is an antipsychotic drug used to treat schizophrenia, bipolar disorder, and depression.

o          Case Study: Aripiprazole has moderate protein binding (about 99%), primarily to albumin. The extent of protein binding impacts its distribution, metabolism, and elimination. Understanding its protein binding characteristics is crucial for dosing regimens and avoiding potential drug interactions17.

2.         Warfarin (Coumadin):

o          Background: Warfarin11 is an anticoagulant used to prevent and treat blood clots.

o          Case Study: Warfarin is highly protein-bound (approximately 99%), mainly to albumin. Changes in protein binding due to factors like hypoalbuminemia or drug interactions (e.g., with NSAIDs) can significantly alter its pharmacokinetics and increase the risk of bleeding or thrombotic events. Clinicians must monitor free drug concentrations and adjust dosages accordingly to maintain therapeutic efficacy and safety.

3.         Diazepam (Valium):

o          Background: Diazepam is a benzodiazepine11 used to treat anxiety disorders, muscle spasms, and seizures.

o          Case Study: Diazepam exhibits high protein binding (approximately 98%) to albumin. Changes in protein binding capacity, such as in critically ill patients with hypoalbuminemia, can prolong the drug's effects due to increased free drug concentrations. This can lead to enhanced sedation and respiratory depression, necessitating dosage adjustments or alternative treatment approaches.

4.         Phenytoin (Dilantin):

o          Background: Phenytoin11 is an anticonvulsant used to control seizures.

o          Case Study: Phenytoin is highly protein-bound (approximately 90-95%) to albumin. Its binding is sensitive to changes in plasma protein levels and interactions with other drugs that also bind to albumin. For example, co-administration with valproic acid, which competes for albumin binding sites, can lead to increased free phenytoin levels and potential toxicity. Clinicians must carefully monitor free phenytoin concentrations to optimize therapeutic outcomes and minimize adverse effects.

5.         Furosemide (Lasix):

o          Background: Furosemide11 is a loop diuretic used to treat edema and hypertension.

o          Case Study: Furosemide has moderate protein binding (approximately 91-99%), primarily to albumin. Variability in protein binding can affect its distribution and elimination, impacting its diuretic efficacy. Understanding protein binding helps in adjusting doses based on factors like renal function and concurrent medications to achieve desired therapeutic effects.

In these case studies, protein binding influenced drug distribution, metabolism, and potential interactions, highlighting its critical role in clinical pharmacology and therapeutic management. Understanding and monitoring protein binding characteristics are essential for optimizing drug therapy and ensuring patient safety.

Here are some emerging trends, challenges, and future directions in this field:

1. Advanced Analytical Techniques:

High-Throughput Screening:

There is a growing demand for faster and more efficient methods to assess protein binding, especially in early drug discovery stages. Techniques such as rapid equilibrium dialysis and ultrafiltration coupled with high-performance liquid chromatography (HPLC) are being optimized for high-throughput screening applications.

Microfluidics:

Microfluidic platforms are being explored to miniaturize and automate protein binding assays, reducing sample volumes and improving assay throughput and efficiency.

2. Integration of Pharmacogenomics:

Genetic Variability:

Understanding how genetic polymorphisms affect protein structure and function, including those involved in drug binding (e.g., albumin variants), will enable more personalized dosing strategies.

Precision Medicine:

Protein binding data can be integrated with genomic information to predict individual drug responses and optimize treatment regimens based on genetic profiles.

3. Impact of Disease States and Comorbidities:

Hypo albuminemia and Other Conditions: Factors like hypo albuminemia, inflammation, and organ dysfunction can alter protein binding capacity. Future research will focus on developing strategies to account for these variations in clinical practice, potentially through adaptive dosing algorithms.

  1. Computational Modeling and Simulation:

Quantitative Structure-Activity Relationship (QSAR):

Computational models are being developed to predict drug-protein binding interactions based on molecular structure, aiding in drug design and optimization.

Physiologically-Based Pharmacokinetic (PBPK) Modeling :

PBPK models incorporate protein binding data to simulate drug distribution and predict drug-drug interactions under various physiological conditions, enhancing clinical decision-making.

5. Regulatory Considerations and Standardization:

Guidance and Guidelines:

Regulatory agencies are emphasizing the importance of understanding protein binding in drug development, requiring robust methodologies and standardized approaches for data generation and interpretation.

Harmonization Efforts:

Collaborative efforts are underway to establish consensus guidelines and recommendations for protein binding studies across different regulatory jurisdictions, ensuring consistency and reliability in data submission.

6. Clinical Translation and Application:

Patient-Centric Approaches:

Incorporating protein binding data into clinical decision support systems and electronic health records can facilitate real-time dose adjustment and improve medication safety and efficacy.

Therapeutic Drug Monitoring:

Utilizing protein binding information in therapeutic drug monitoring programs to optimize dosing regimens and minimize adverse effects, particularly in vulnerable patient populations.

7. Educational and Research Initiatives:

Training and Awareness:

Continued education and training programs are needed to enhance awareness among healthcare professionals and researchers about the importance of protein binding in drug therapy and its clinical implications.

Collaborative Research:

Multi-disciplinary collaborations between pharmacologists, clinicians, computational biologists, and industry partners will drive innovation and address complex challenges in protein binding research. In conclusion, the future of protein binding research and application in drug development and clinical practice is promising, with advancements in technology, pharmacogenomics, computational modelling , and regulatory frameworks contributing to personalized and effective therapeutic interventions. Addressing challenges such as variability in protein binding and optimizing analytical methodologies will be key to realizing these advancements and improving patient outcomes.

CONCLUSION:

Protein binding is a fundamental aspect of pharmacokinetics that profoundly influences drug development and therapeutic efficacy. Understanding the mechanisms of protein binding, including the types of interactions and the key proteins involved, is crucial for predicting a drug’s behavior in the body. This understanding impacts various pharmacokinetic parameters, such as drug distribution, metabolism, and excretion, all of which are essential for determining appropriate dosing regimens and ensuring therapeutic efficacy. The equilibrium between bound and free drug fractions is a critical determinant of a drug's pharmacological activity and toxicity. Variations in protein binding due to physiological changes, disease states, or drug interactions can significantly alter the free drug concentration, leading to potential therapeutic failures or adverse effects. Thus, accurate assessment of protein binding is indispensable for optimizing drug dosing and minimizing risks. In drug development, integrating protein binding studies early in the process aids in the design of safer and more effective therapeutic agents. It enables the prediction of drug interactions, adjustment of dosing in special populations, and the formulation of drugs to achieve desired pharmacokinetic profiles. Moreover, understanding protein binding helps in designing clinical trials and interpreting their outcomes, particularly for drugs with narrow therapeutic windows or those used in sensitive patient populations. Ultimately, a thorough comprehension of protein binding mechanisms and their effects on pharmacokinetics not only enhances drug development but also improves patient care. As pharmaceutical sciences advance, continued research into the nuances of protein binding will be vital for the development of innovative therapies that are both effective and safe. This holistic approach ensures that new drugs are better tailored to meet the complex needs of diverse patient populations, ultimately leading to more personalized and effective healthcare solutions.

REFERENCES

  1. Schmidt, S., Gonzalez, D., & Derendorf, H. (2010). Significance of Protein Binding in Pharmacokinetics and Pharmacodynamics. Journal of Pharmaceutical Sciences, 99(3), 1107–1122. doi:10.1002/jps.21916
  2. Vuignier, K., Schappler, J., Veuthey, JL. et al. Drug–protein binding: a critical review of analytical tools. Anal Bioanal Chem 398, 53–66 (2010). https://doi.org/10.1007/s00216-010-3737-1
  3. Olson, R. E., & Christ, D. D. (1996). Chapter 33. Plasma Protein Binding of Drugs. Annual Reports in Medicinal Chemistry, 327–336. doi:10.1016/s0065-7743(08)60472-8
  4. Kratochwil, N. A., Huber, W., Müller, F., Kansy, M., & Gerber, P. R. (2002). Predicting plasma protein binding of drugs: a new approach. Biochemical Pharmacology, 64(9), 1355–1374. doi:10.1016/s0006-2952(02)01074-2
  5. Roberts, J.A., Pea, F. & Lipman, J. The Clinical Relevance of Plasma Protein Binding Changes. Clin Pharmacokinet 52, 1–8 (2013). https://doi.org/10.1007/s40262-012-0018-5
  6. Trainor, G. L. (2007). The importance of plasma protein binding in drug discovery. Expert Opinion on Drug Discovery, 2(1), 51–64. https://doi.org/10.1517/17460441.2.1.51
  7. Howard, M., Hill, J., Galluppi, G., & McLean, M. (2010). Plasma Protein Binding in Drug Discovery and Development. https://doi.org/10.2174/138620710790596745.
  8. Rowland, M., & Tozer, T. N. (2011). Clinical Pharmacokinetics and Pharmacodynamics: Concepts and Applications. Lippincott Williams & Wilkins.
  9. Routledge, P. (1986), The plasma protein binding of basic drugs.. British Journal of Clinical Pharmacology, 22: 499-506. https://doi.org/10.1111/j.1365-2125.1986.tb02927.x
  10. Bohnert, T., & Gan, L.-S. (2013). Plasma protein binding: From discovery to development. Journal of Pharmaceutical Sciences
  11. Jusko, W. J., & Gretch, M. (1976). Plasma and Tissue Protein Binding of Drugs in Pharmacokinetics. Drug Metabolism Reviews, 5(1), 43–140. https://doi.org/10.3109/03602537608995839
  12. Oravcova´, J., Bo¨hs, B., & Lindner, W. (1996). Drug-protein binding studies new trends in analytical and experimental methodology. Journal of Chromatography B: Biomedical Sciences and Applications, 677(1), 1–28. doi:10.1016/0378-4347(95)00425-4
  13. Borgai Olof , Piafsky Kenneth M. , Nilsen Odd G. , (1977), Plasma protein binding of basic drugs, Clinical Pharmacology & Therapeutics, 22, doi: 10.1002/cpt1977225part1539.
  14. Vacobi Avraham, Udall John A., Levy Gerhard, (1976), Serum protein binding as a determinant of warfarin body clearance and anticoagulant effect, Clinical Pharmacology & Therapeutics, 19, doi: 10.1002/cpt1976195part1552.
  15. Sebille, B. (1990), Methods of drug protein binding determinations. Fundamental & Clinical Pharmacology, 4: 151s-161s. https://doi.org/10.1111/j.1472-8206.1990.tb00073.x        
  16. Banker, M., & Clark, T. (2008). Plasma / Serum Protein Binding Determinations. Current Drug Metabolism, 9(9), 854–859. doi:10.2174/138920008786485065
  17. McElnay, J.C., D’Arcy, P.F. Protein Binding Displacement Interactions and their Clinical Importance. Drugs 25, 495–513 (1983). https://doi.org/10.2165/00003495-198325050-00003
  18. Berggård, T., Linse, S. and James, P. (2007), Methods for the detection and analysis of protein–protein interactions. Proteomics, 7: 2833-2842. https://doi.org/10.1002/pmic.200700131
  19. Chignell, C.F. (1971). Physical Methods for Studying Drug-Protein Binding. In: Brodie, B.B., Gillette, J.R., Ackerman, H.S. (eds) Concepts in Biochemical Pharmacology. Handbuch der experimentellen Pharmakologie/Handbook of Experimental Pharmacology, vol 28 / 1. Springer, Berlin, Heidelberg. https://doi.org/
  20. Tillement, JP., Lhoste, F. & Giudicelli, J.F. Diseases and Drug Protein Binding. Clin Pharmacokinet 3, 144–154 (1978). https://doi.org/10.2165/00003088-197803020-00004
  21. Gideon Schreiber, Amy E Keating,Protein binding specificity versus promiscuity,Current Opinion in Structural Biology,Volume 21, Issue 1,2011,Pages 50-61,ISSN 0959-440X,https://doi.org/10.1016/j.sbi.2010.10.002.

Reference

  1. Schmidt, S., Gonzalez, D., & Derendorf, H. (2010). Significance of Protein Binding in Pharmacokinetics and Pharmacodynamics. Journal of Pharmaceutical Sciences, 99(3), 1107–1122. doi:10.1002/jps.21916
  2. Vuignier, K., Schappler, J., Veuthey, JL. et al. Drug–protein binding: a critical review of analytical tools. Anal Bioanal Chem 398, 53–66 (2010). https://doi.org/10.1007/s00216-010-3737-1
  3. Olson, R. E., & Christ, D. D. (1996). Chapter 33. Plasma Protein Binding of Drugs. Annual Reports in Medicinal Chemistry, 327–336. doi:10.1016/s0065-7743(08)60472-8
  4. Kratochwil, N. A., Huber, W., Müller, F., Kansy, M., & Gerber, P. R. (2002). Predicting plasma protein binding of drugs: a new approach. Biochemical Pharmacology, 64(9), 1355–1374. doi:10.1016/s0006-2952(02)01074-2
  5. Roberts, J.A., Pea, F. & Lipman, J. The Clinical Relevance of Plasma Protein Binding Changes. Clin Pharmacokinet 52, 1–8 (2013). https://doi.org/10.1007/s40262-012-0018-5
  6. Trainor, G. L. (2007). The importance of plasma protein binding in drug discovery. Expert Opinion on Drug Discovery, 2(1), 51–64. https://doi.org/10.1517/17460441.2.1.51
  7. Howard, M., Hill, J., Galluppi, G., & McLean, M. (2010). Plasma Protein Binding in Drug Discovery and Development. https://doi.org/10.2174/138620710790596745.
  8. Rowland, M., & Tozer, T. N. (2011). Clinical Pharmacokinetics and Pharmacodynamics: Concepts and Applications. Lippincott Williams & Wilkins.
  9. Routledge, P. (1986), The plasma protein binding of basic drugs.. British Journal of Clinical Pharmacology, 22: 499-506. https://doi.org/10.1111/j.1365-2125.1986.tb02927.x
  10. Bohnert, T., & Gan, L.-S. (2013). Plasma protein binding: From discovery to development. Journal of Pharmaceutical Sciences
  11. Jusko, W. J., & Gretch, M. (1976). Plasma and Tissue Protein Binding of Drugs in Pharmacokinetics. Drug Metabolism Reviews, 5(1), 43–140. https://doi.org/10.3109/03602537608995839
  12. Oravcova´, J., Bo¨hs, B., & Lindner, W. (1996). Drug-protein binding studies new trends in analytical and experimental methodology. Journal of Chromatography B: Biomedical Sciences and Applications, 677(1), 1–28. doi:10.1016/0378-4347(95)00425-4
  13. Borgai Olof , Piafsky Kenneth M. , Nilsen Odd G. , (1977), Plasma protein binding of basic drugs, Clinical Pharmacology & Therapeutics, 22, doi: 10.1002/cpt1977225part1539.
  14. Vacobi Avraham, Udall John A., Levy Gerhard, (1976), Serum protein binding as a determinant of warfarin body clearance and anticoagulant effect, Clinical Pharmacology & Therapeutics, 19, doi: 10.1002/cpt1976195part1552.
  15. Sebille, B. (1990), Methods of drug protein binding determinations. Fundamental & Clinical Pharmacology, 4: 151s-161s. https://doi.org/10.1111/j.1472-8206.1990.tb00073.x        
  16. Banker, M., & Clark, T. (2008). Plasma / Serum Protein Binding Determinations. Current Drug Metabolism, 9(9), 854–859. doi:10.2174/138920008786485065
  17. McElnay, J.C., D’Arcy, P.F. Protein Binding Displacement Interactions and their Clinical Importance. Drugs 25, 495–513 (1983). https://doi.org/10.2165/00003495-198325050-00003
  18. Berggård, T., Linse, S. and James, P. (2007), Methods for the detection and analysis of protein–protein interactions. Proteomics, 7: 2833-2842. https://doi.org/10.1002/pmic.200700131
  19. Chignell, C.F. (1971). Physical Methods for Studying Drug-Protein Binding. In: Brodie, B.B., Gillette, J.R., Ackerman, H.S. (eds) Concepts in Biochemical Pharmacology. Handbuch der experimentellen Pharmakologie/Handbook of Experimental Pharmacology, vol 28 / 1. Springer, Berlin, Heidelberg. https://doi.org/
  20. Tillement, JP., Lhoste, F. & Giudicelli, J.F. Diseases and Drug Protein Binding. Clin Pharmacokinet 3, 144–154 (1978). https://doi.org/10.2165/00003088-197803020-00004
  21. Gideon Schreiber, Amy E Keating,Protein binding specificity versus promiscuity,Current Opinion in Structural Biology,Volume 21, Issue 1,2011,Pages 50-61,ISSN 0959-440X,https://doi.org/10.1016/j.sbi.2010.10.002.

Photo
K.T. AKSHARA
Corresponding author

DEPARTMENT OF PHARMACEUTICAL ANALYSIS, AL SHIFA COLLEGE OF PHARMACY

Photo
PRASANTH S.S
Co-author

HEAD OF DEPARTMENT, DEPARTMENT OF PHARMACEUTICAL ANALYSIS, AL SHIFA COLLEGE OF PHARMACY

Photo
JISHA.U
Co-author

DEPARTMENT OF PHARMACEUTICAL ANALYSIS, AL SHIFA COLLEGE OF PHARMACY

Photo
SANOOJA P.K
Co-author

DEPARTMENT OF PHARMACEUTICAL ANALYSIS, AL SHIFA COLLEGE OF PHARMACY

Photo
SIBINA M.K
Co-author

DEPARTMENT OF PHARMACEUTICAL ANALYSIS, AL SHIFA COLLEGE OF PHARMACY

Photo
RIYA RAJAN
Co-author

DEPARTMENT OF PHARMACEUTICAL ANALYSIS, AL SHIFA COLLEGE OF PHARMACY

Photo
AJAY. A
Co-author

DEPARTMENT OF PHARMACEUTICAL ANALYSIS, AL SHIFA COLLEGE OF PHARMACY

Photo
MOHAMED FAROOQ. P
Co-author

DEPARTMENT OF PHARMACEUTICAL ANALYSIS, AL SHIFA COLLEGE OF PHARMACY

K.T. Akshara , Prasanth S. S. , Jisha U. , Sanooja P. K. , Sibina M. K. , Riya Rajan , Ajay A. , Mohammed Farooq , Protein Binding In Drug Development: A Systematic Review Of Mechanisms, Pharmacokinetics, And Clinical Implications, Int. J. of Pharm. Sci., 2024, Vol 2, Issue 8, 3369-3379. https://doi.org/10.5281/zenodo.13337309

More related articles
A Review On- Artificial Intelligence In Pharmacy &...
Meet Shah, Swapnil G. Kale, Kalyani Raut, Sayali Gandhi, Swapnil ...
Formulation And Evaluation Of Herbal Cough Syrup...
Lodwal Pavanraj Bhagwan, Tejaswini Kanhaiyasing Pardeshi, asst. p...
Cell and Gene Therapy in Cancer...
Swati Jadhav, Pavan khonde, Aman Upaganlawar, Chandrashekhar Upas...
Exploring the Hypoglycaemic Effects of herbal plant: A Potential Remedy for Diab...
Sonali Akshay Wagh, Ganesh Anil Vitukade, Kanish Mehta, ...
The Impact of Excipients on Pharmaceutical Product Quality: Review the role of e...
Prashant Ghayal , Hanumant Gutte, Adarsh Mane, Bhushan Kolekar, Prashant Bhoir, ...
Formulation And Evaluation Of Polyherbal Gel...
Palak, Dev Prakash Dahiya, Shalu Bharti, Chinu Kumari, ...
Related Articles
Recent Advances In Oral Solid Dosage Form Formulation And Development...
Ankita Anil walunj , Wagh Snehal A., Kale Megha K., Dhage Shubhangi S, Awari Monika S, Bhalekar S. M...
Formulation And Evaluation Of Herbal Shampoo With Goat Milk...
Prasad Rodge, Mayur Bagul, Nilesh Bagal, Utkarsh Bhamare, Utkarsha Chaudhari, ...
Review on Thyroid Disorder...
Anuja Jadhav, Apurva Torne, Sakshi Shahane, Prashant Singar , ...
The Alzheimer's Dieases...
Siddhant Sawatkar, Shivanee Tupkar, Dr. Gajanan Sanap, ...
A Review On- Artificial Intelligence In Pharmacy & Applications Of Ai In Pharmac...
Meet Shah, Swapnil G. Kale, Kalyani Raut, Sayali Gandhi, Swapnil Katkhade, Rohini Haral, Sneha S. Ka...
More related articles
A Review On- Artificial Intelligence In Pharmacy & Applications Of Ai In Pharmac...
Meet Shah, Swapnil G. Kale, Kalyani Raut, Sayali Gandhi, Swapnil Katkhade, Rohini Haral, Sneha S. Ka...
Formulation And Evaluation Of Herbal Cough Syrup...
Lodwal Pavanraj Bhagwan, Tejaswini Kanhaiyasing Pardeshi, asst. prof. Ram B Ingle, Kale Dipak Raju, ...
Cell and Gene Therapy in Cancer...
Swati Jadhav, Pavan khonde, Aman Upaganlawar, Chandrashekhar Upasani , ...
A Review On- Artificial Intelligence In Pharmacy & Applications Of Ai In Pharmac...
Meet Shah, Swapnil G. Kale, Kalyani Raut, Sayali Gandhi, Swapnil Katkhade, Rohini Haral, Sneha S. Ka...
Formulation And Evaluation Of Herbal Cough Syrup...
Lodwal Pavanraj Bhagwan, Tejaswini Kanhaiyasing Pardeshi, asst. prof. Ram B Ingle, Kale Dipak Raju, ...
Cell and Gene Therapy in Cancer...
Swati Jadhav, Pavan khonde, Aman Upaganlawar, Chandrashekhar Upasani , ...