Physicochemical properties play a crucial role in drug design and development. These properties determine how a drug interacts with the body, affecting its absorption, distribution, metabolism, and excretion. Understanding these properties helps medicinal chemists create effective and safe medications.

Key properties include , , , and . These factors influence how well a drug is absorbed, distributed throughout the body, and eliminated. By optimizing these properties, scientists can improve a drug's effectiveness and minimize potential side effects.

Properties affecting drug absorption

  • Absorption refers to the process by which a drug moves from the site of administration into the bloodstream
  • The physicochemical properties of a drug play a crucial role in determining its absorption characteristics
  • Understanding these properties helps medicinal chemists design drugs with optimal absorption profiles

Lipophilicity vs hydrophilicity

Top images from around the web for Lipophilicity vs hydrophilicity
Top images from around the web for Lipophilicity vs hydrophilicity
  • Lipophilicity refers to a drug's affinity for lipids or fats, while refers to its affinity for water
  • Highly lipophilic drugs tend to cross biological membranes more easily, as cell membranes are composed of lipid bilayers
  • However, extremely lipophilic drugs may have poor solubility in aqueous environments, limiting their absorption
  • Hydrophilic drugs are more soluble in water but may struggle to pass through lipid-rich cell membranes

Solubility in biological fluids

  • Solubility is the maximum amount of a drug that can dissolve in a given volume of solvent (e.g., water or gastrointestinal fluids)
  • Drugs must be soluble in the aqueous environment of the gastrointestinal tract to be absorbed
  • Poorly soluble drugs may precipitate in the gut, reducing their bioavailability
  • Strategies to improve solubility include salt formation, prodrugs, and formulation techniques (e.g., micronization, solid dispersions)

Ionization of drugs

  • Many drugs are weak acids or bases that can exist in ionized or unionized forms depending on the pH of their environment
  • The ionization state of a drug affects its solubility, permeability, and absorption
  • Unionized forms of drugs typically have higher permeability across cell membranes
  • The relates pH, , and the ratio of ionized to unionized drug: pH=pKa+log([A]/[HA])pH = pKa + log([A-]/[HA])

Partition coefficient

  • The () is a measure of a drug's lipophilicity, defined as the ratio of its concentration in an organic solvent (e.g., octanol) to its concentration in water at equilibrium
  • Drugs with higher log P values are more lipophilic and generally have better membrane permeability
  • However, extremely high log P values can lead to poor solubility and increased metabolic liability
  • The optimal log P range for oral drugs is typically between 1 and 5

Drug permeability

  • Permeability refers to a drug's ability to cross biological membranes, such as the intestinal epithelium or the blood-brain barrier
  • Factors affecting permeability include lipophilicity, molecular size, charge, and the presence of transporters
  • Passive diffusion is the primary mechanism for most drugs, favoring small, lipophilic molecules
  • Active transport and facilitated diffusion can also contribute to drug permeability, mediated by specific transporters (e.g., P-glycoprotein)

Properties affecting drug distribution

  • Distribution describes the process by which a drug moves from the bloodstream into various tissues and organs
  • The extent and pattern of drug distribution depend on its physicochemical properties and its interactions with blood components and tissue structures

Plasma protein binding

  • Many drugs bind reversibly to plasma proteins, such as albumin and alpha-1-acid glycoprotein
  • Protein binding can affect a drug's distribution, as only the unbound (free) fraction is available to diffuse into tissues
  • High protein binding can limit a drug's volume of distribution and reduce its clearance
  • However, it can also prolong a drug's half-life and provide a reservoir for sustained release

Tissue binding

  • Drugs may bind specifically or non-specifically to tissue components, such as proteins, lipids, or nucleic acids
  • Tissue binding can lead to drug accumulation in certain organs (e.g., liver, kidneys) or in fatty tissues
  • Extensive tissue binding may result in a large volume of distribution and a prolonged elimination half-life
  • Tissue binding can also contribute to drug toxicity if the bound drug disrupts normal cellular functions

Blood-brain barrier penetration

  • The blood-brain barrier (BBB) is a selective barrier that restricts the entry of substances from the bloodstream into the central nervous system (CNS)
  • To cross the BBB, drugs must be lipophilic, have a low molecular weight (< 500 Da), and not be strongly bound to plasma proteins
  • Drugs targeting CNS disorders (e.g., antidepressants, antipsychotics) must be designed to penetrate the BBB effectively
  • Conversely, drugs intended for peripheral targets should minimize BBB penetration to avoid central side effects

Properties affecting drug metabolism

  • Metabolism refers to the biochemical modification of drugs by enzymes, primarily in the liver
  • The metabolic fate of a drug is determined by its chemical structure and its interactions with metabolic enzymes

Metabolic stability

  • Metabolic stability is a measure of a drug's resistance to biotransformation by metabolic enzymes
  • Drugs with high metabolic stability have a longer duration of action and require less frequent dosing
  • However, metabolically stable drugs may accumulate in the body, leading to toxicity
  • Balancing metabolic stability with clearance is crucial for optimizing drug exposure and safety

Cytochrome P450 interactions

  • (CYP) enzymes are a superfamily of heme-containing proteins responsible for the metabolism of many drugs
  • CYP enzymes can be inhibited or induced by certain drugs, leading to drug-drug interactions
  • CYP inhibition can increase the exposure and toxicity of substrate drugs, while CYP induction can decrease their efficacy
  • Medicinal chemists aim to minimize CYP interactions by modifying drug structures or selecting compounds with favorable CYP profiles

Phase I vs Phase II metabolism

  • Drug metabolism is divided into two phases: Phase I (functionalization) and Phase II (conjugation)
  • Phase I reactions introduce or expose functional groups (e.g., hydroxylation, oxidation) and are primarily mediated by CYP enzymes
  • Phase II reactions attach hydrophilic moieties (e.g., glucuronic acid, sulfate) to the drug or its Phase I metabolites, increasing their water solubility and facilitating excretion
  • The balance between Phase I and Phase II metabolism can influence a drug's bioavailability, half-life, and toxicity

Prodrugs and active metabolites

  • Prodrugs are inactive compounds that are metabolically converted into active drugs within the body
  • Prodrug strategies can be used to improve solubility, permeability, or stability of the parent drug
  • Examples of prodrugs include ester derivatives (e.g., aspirin, oseltamivir) and phosphate esters (e.g., cyclophosphamide)
  • Some drugs are metabolized into active compounds that contribute to their pharmacological effects (e.g., codeine to morphine, tamoxifen to endoxifen)

Properties affecting drug excretion

  • Excretion is the process by which drugs and their metabolites are eliminated from the body
  • The primary routes of drug excretion are renal (via urine) and biliary (via feces), although other minor routes (e.g., sweat, saliva) may also contribute

Renal clearance

  • Renal clearance is the volume of blood that is completely cleared of a drug by the kidneys per unit time
  • Drugs are filtered, secreted, and reabsorbed in the nephron, the functional unit of the kidney
  • Factors affecting renal clearance include glomerular filtration rate, tubular secretion, and tubular reabsorption
  • Drugs with high renal clearance are rapidly eliminated from the body, while those with low renal clearance may accumulate and cause toxicity in patients with impaired kidney function

Biliary excretion

  • Biliary excretion is the process by which drugs and their metabolites are eliminated from the body via the bile into the feces
  • Drugs are actively transported from hepatocytes into the bile canaliculi by various transporters (e.g., P-glycoprotein, BCRP)
  • Factors affecting biliary excretion include molecular weight, polarity, and transporter affinity
  • Drugs with high biliary excretion may undergo enterohepatic recirculation, where they are reabsorbed from the intestine and returned to the liver, prolonging their half-life

Half-life and elimination rate

  • The elimination half-life (t1/2) is the time required for the plasma concentration of a drug to decrease by 50% during the elimination phase
  • The elimination rate constant (ke) is the fraction of a drug eliminated from the body per unit time and is related to the half-life by the equation: t1/2=ln(2)ket_{1/2} = \frac{ln(2)}{k_e}
  • Drugs with short half-lives require frequent dosing to maintain therapeutic concentrations, while those with long half-lives can be administered less frequently
  • The elimination rate and half-life are influenced by the drug's clearance and volume of distribution

Structure-property relationships

  • Structure-property relationships (SPRs) describe the connections between a drug's chemical structure and its physicochemical and pharmacokinetic properties
  • Understanding SPRs allows medicinal chemists to rationally design drugs with desired properties and optimize lead compounds

Functional groups and properties

  • Functional groups are specific arrangements of atoms within a molecule that confer characteristic properties
  • Examples of functional groups include hydroxyl (-OH), amino (-NH2), carboxyl (-COOH), and ester (-COO-)
  • The presence, absence, or modification of functional groups can significantly impact a drug's solubility, permeability, and metabolic stability
  • For instance, adding a hydroxyl group can increase a drug's water solubility, while an ester group can improve its lipophilicity and permeability

Molecular size and shape

  • The size and shape of a drug molecule can influence its absorption, distribution, and binding to targets
  • Smaller molecules (molecular weight < 500 Da) generally have better oral bioavailability and can cross biological membranes more easily
  • However, very small molecules (molecular weight < 200 Da) may have poor specificity and off-target effects
  • The shape of a drug molecule, determined by its conformation and flexibility, can affect its interactions with receptors, enzymes, and transporters

Stereochemistry and drug properties

  • refers to the three-dimensional arrangement of atoms in a molecule
  • Stereoisomers are compounds with the same molecular formula but different spatial arrangements (e.g., enantiomers, diastereomers)
  • Stereochemistry can significantly impact a drug's pharmacokinetic and pharmacodynamic properties
  • For example, the (S)-enantiomer of ibuprofen is more potent than the (R)-enantiomer, while the (R)-enantiomer of thalidomide is teratogenic

Quantitative structure-activity relationships (QSAR)

  • QSAR is a computational approach that relates a drug's chemical structure to its biological activity using mathematical models
  • QSAR models are built using a set of molecular descriptors (e.g., log P, molecular weight, topological indices) and statistical methods (e.g., multiple linear regression, partial least squares)
  • QSAR can be used to predict the properties of new compounds, guide the optimization of lead compounds, and identify key structural features responsible for drug activity
  • Examples of QSAR applications include the design of novel antibacterial agents and the prediction of ADME properties

Optimization of drug properties

  • Drug optimization is the process of improving the properties of a lead compound to create a drug candidate suitable for clinical development
  • The goal of optimization is to enhance the drug's efficacy, safety, and pharmacokinetic profile while maintaining its activity against the intended target

Strategies for improving solubility

  • Improving solubility is crucial for ensuring adequate drug absorption and bioavailability
  • Salt formation involves reacting the drug with an appropriate acid or base to create a more water-soluble ionic compound (e.g., sodium salt of ibuprofen)
  • Prodrug approaches, such as ester or phosphate derivatives, can increase solubility by masking hydrophobic groups (e.g., fosamprenavir, a phosphate ester prodrug of amprenavir)
  • Formulation techniques, such as micronization (reducing particle size), solid dispersions (dispersing the drug in a hydrophilic matrix), and cyclodextrin complexation, can enhance solubility without modifying the drug's chemical structure

Enhancing permeability and absorption

  • Improving permeability and absorption is essential for drugs targeting intracellular or CNS receptors
  • Increasing lipophilicity, for example, by adding non-polar groups or reducing hydrogen bond donors, can enhance passive diffusion across cell membranes
  • Reducing molecular size or polar surface area can also improve permeability by facilitating passage through tight junctions or lipid bilayers
  • Targeting specific transporters (e.g., amino acid or peptide transporters) can enable active uptake of drugs with suboptimal passive permeability

Reducing metabolic liability

  • Minimizing metabolic liability is important for prolonging drug exposure and avoiding toxic metabolites
  • Identifying and modifying metabolic "soft spots" (e.g., labile esters, oxidizable groups) can improve metabolic stability
  • Replacing metabolically labile groups with more stable analogs (e.g., amides instead of esters, deuterium instead of hydrogen) can reduce CYP-mediated metabolism
  • Designing compounds with reduced lipophilicity or increased polarity can decrease their affinity for metabolic enzymes and increase their excretion

Prolonging half-life and duration of action

  • Extending a drug's half-life and duration of action can improve patient compliance and therapeutic outcomes
  • Increasing plasma protein binding can create a reservoir of circulating drug, gradually releasing it over time
  • Designing prodrugs with slow bioactivation rates can prolong the exposure to the active compound
  • Incorporating structural features that reduce renal or biliary clearance (e.g., increasing molecular size, adding polar groups) can extend a drug's elimination half-life

Physicochemical profiling in drug discovery

  • Physicochemical profiling involves the experimental and computational assessment of a drug's properties at various stages of the discovery process
  • Profiling helps identify promising compounds, optimize lead structures, and predict in vivo performance

High-throughput screening assays

  • High-throughput screening (HTS) assays are used to rapidly evaluate the properties of large compound libraries
  • Solubility assays measure the concentration of a compound in aqueous buffer using UV/Vis spectroscopy or nephelometry
  • Permeability assays assess a compound's ability to cross artificial membranes (e.g., PAMPA) or cell monolayers (e.g., Caco-2)
  • Metabolic stability assays determine the rate of compound disappearance in the presence of liver microsomes or hepatocytes

In vitro ADME studies

  • In vitro ADME (absorption, distribution, metabolism, excretion) studies provide more detailed information on a compound's pharmacokinetic properties
  • Plasma protein binding is measured using equilibrium dialysis or ultrafiltration techniques
  • Blood-brain barrier penetration is assessed using in vitro models such as MDCK or LLC-PK1 cell lines expressing efflux transporters
  • CYP inhibition and induction studies evaluate the potential for drug-drug interactions using human liver microsomes or hepatocytes

In silico property prediction

  • In silico methods use computational models to predict a compound's properties based on its chemical structure
  • Quantitative structure-property relationship (QSPR) models correlate molecular descriptors with experimental property data using statistical techniques (e.g., multiple linear regression, artificial neural networks)
  • Molecular docking simulations can predict a compound's binding affinity to targets such as enzymes or transporters
  • Physiologically based pharmacokinetic (PBPK) models integrate in vitro and in silico data to predict a drug's absorption, distribution, metabolism, and excretion in humans

Druglikeness and lead optimization

  • Druglikeness refers to the set of molecular properties that are consistent with orally active drugs
  • Lipinski's Rule of Five is a widely used guideline for assessing druglikeness, based on molecular weight (≤ 500 Da), log P (≤ 5), hydrogen bond donors (≤ 5), and hydrogen bond acceptors (≤ 10)
  • Other druglikeness filters include the Veber rules (rotatable bonds ≤ 10, polar surface area ≤ 140 Ų) and the Ghose filter (molecular weight between 160 and 480 Da, log P between -0.4 and 5.6)
  • Lead optimization involves iterative cycles of synthesis, testing, and refinement to improve a compound's druglike properties while maintaining its potency and selectivity

Key Terms to Review (20)

Chirality: Chirality refers to the geometric property of a molecule that makes it non-superimposable on its mirror image, much like how left and right hands are mirror images but cannot perfectly align. This property is crucial in medicinal chemistry because the different spatial arrangements of atoms in chiral molecules can lead to vastly different biological activities. Understanding chirality is essential for analyzing conformational preferences, physicochemical properties, and the structure-activity relationship of various compounds, including alkaloids and pharmacophores.
Cytochrome P450: Cytochrome P450 refers to a family of enzymes that play a crucial role in the metabolism of various substances, including drugs, toxins, and endogenous compounds. These enzymes are found in many tissues but are most abundant in the liver, where they facilitate the oxidation of organic substances, aiding in their detoxification and elimination from the body. Their significance extends to understanding how physicochemical properties influence drug behavior, as well as their involvement in Phase I and Phase II metabolic processes.
Henderson-Hasselbalch Equation: The Henderson-Hasselbalch equation is a mathematical formula used to calculate the pH of a buffered solution based on the concentration of the acid and its conjugate base. It illustrates the relationship between pH, pKa, and the ratio of the concentrations of the deprotonated and protonated forms of an acid, providing crucial insights into the physicochemical properties of molecules in biological systems.
Hermann Emil Fischer: Hermann Emil Fischer was a prominent German chemist, known for his pioneering work in the field of medicinal chemistry and biochemistry, particularly in the study of sugars and purines. His contributions laid foundational principles for drug design and development, making significant impacts in areas like bioisosterism, physicochemical properties, and prodrug design, which are essential for understanding the behavior of pharmaceuticals within biological systems.
Hydrophilicity: Hydrophilicity refers to the property of a molecule that enables it to interact favorably with water, typically due to the presence of polar groups or charged regions. This characteristic plays a crucial role in various processes, such as solubility, drug formulation, and biological interactions, making it a key factor in understanding physicochemical properties.
Ionization: Ionization is the process by which an atom or molecule gains or loses electrons, resulting in the formation of ions. This process is crucial in understanding how substances interact in solutions and how they can influence the distribution and physicochemical properties of drugs within biological systems.
Kinetic stability: Kinetic stability refers to the resistance of a chemical species to undergo a change in its state or structure over time, particularly concerning reaction rates. This concept is important because it affects how long a compound will remain unchanged before transforming into a different product, which directly influences drug efficacy and shelf-life in medicinal chemistry.
Lipophilicity: Lipophilicity refers to the chemical affinity of a substance for lipids or fats, essentially indicating how well a compound can dissolve in non-polar solvents compared to polar solvents. This property plays a crucial role in determining how drugs are distributed in the body, how they are metabolized, and their overall efficacy as therapeutic agents. Compounds with high lipophilicity tend to penetrate cell membranes more easily, influencing their absorption and distribution, while also impacting drug design and the physicochemical properties that affect their action in biological systems.
Log p: Log p, or logarithm of the partition coefficient (P), is a measure of the lipophilicity of a compound, indicating how well it partitions between an organic solvent (usually octanol) and water. A higher log p value suggests greater hydrophobicity, which often influences drug absorption, distribution, metabolism, and excretion (ADME) properties. Understanding log p helps in predicting the behavior of drugs within biological systems and their potential effectiveness.
Mass Spectrometry: Mass spectrometry is an analytical technique used to measure the mass-to-charge ratio of ions, allowing for the identification and quantification of molecules in a sample. This method is essential in various fields for characterizing chemical compounds, analyzing biomolecules, and determining molecular structures, making it invaluable in early drug development, chemical property assessments, and exploring complex mixtures such as natural products.
Nmr spectroscopy: NMR spectroscopy, or nuclear magnetic resonance spectroscopy, is a powerful analytical technique used to determine the structure of organic compounds by observing the magnetic properties of atomic nuclei. This method provides detailed information about the molecular structure, dynamics, and environment of atoms, making it an essential tool in various fields including medicinal chemistry, where understanding molecular interactions is crucial for drug development and design.
Partition Coefficient: The partition coefficient is a ratio that describes how a compound distributes itself between two immiscible solvents, typically an organic solvent and water. This value is crucial in understanding the lipophilicity of drugs, influencing their absorption, distribution, metabolism, and excretion in biological systems. It is a fundamental physicochemical property that helps predict the behavior of compounds in living organisms.
Paul Ehrlich: Paul Ehrlich was a pioneering German scientist known for his contributions to immunology, chemotherapy, and the concept of targeted therapy in medicine. His work laid the foundation for the development of modern medicinal chemistry, particularly through his ideas around bioisosterism, physicochemical properties, and pharmacophores, which influence drug design and discovery.
Permeability: Permeability refers to the ability of a substance to allow molecules or ions to pass through it. This property is crucial in determining how drugs interact with biological membranes, influencing their absorption, distribution, and overall efficacy. High permeability typically indicates that a compound can easily traverse cellular barriers, while low permeability suggests a more challenging pathway, impacting drug delivery methods and mechanisms.
PKa: pKa is a quantitative measure of the strength of an acid in solution, defined as the negative logarithm of the acid dissociation constant (Ka). It indicates the tendency of a compound to donate a proton (H+) in a chemical reaction, which is crucial for understanding how substances behave in biological systems and drug interactions. The pKa value helps predict the ionization state of a molecule at a given pH, making it an essential concept in assessing physicochemical properties and guiding ligand-based drug design.
Quantitative structure-activity relationship (qsar): Quantitative structure-activity relationship (QSAR) is a computational method used to predict the biological activity of chemical compounds based on their molecular structure. It involves creating mathematical models that correlate specific molecular descriptors with observed biological activities, allowing researchers to identify and optimize potential drug candidates. This technique is crucial in medicinal chemistry for understanding how changes in chemical structure can affect drug efficacy and safety, linking it to concepts such as bioisosterism, physicochemical properties, and ADMET prediction.
Solubility: Solubility is the ability of a substance to dissolve in a solvent, forming a homogeneous solution. It plays a crucial role in various biological processes, drug formulation, and the design of pharmaceuticals, influencing how compounds are absorbed, distributed, metabolized, and excreted by the body.
Stereochemistry: Stereochemistry is the branch of chemistry that focuses on the spatial arrangement of atoms in molecules and how this arrangement affects their chemical behavior and properties. The three-dimensional structure of a molecule can significantly influence its interactions, reactivity, and the physicochemical properties such as boiling point, solubility, and optical activity.
Structure-Activity Relationship (SAR): Structure-Activity Relationship (SAR) refers to the relationship between the chemical or 3D structure of a molecule and its biological activity. Understanding SAR is crucial for optimizing drug design, as it helps identify which structural features influence the effectiveness and potency of a compound against a biological target, guiding modifications to enhance desired properties.
Thermodynamic stability: Thermodynamic stability refers to the tendency of a system to remain in a certain state or configuration under given conditions, often characterized by lower energy states being more stable than higher energy states. This concept is crucial for understanding how substances behave at different temperatures and pressures, influencing their solubility, reactivity, and phase transitions. It plays a significant role in the physicochemical properties of compounds, as substances with high thermodynamic stability tend to resist changes and reactions.
© 2024 Fiveable Inc. All rights reserved.
AP® and SAT® are trademarks registered by the College Board, which is not affiliated with, and does not endorse this website.