0% found this document useful (0 votes)
27 views

CADD Unit 5

The document discusses quantum mechanics and molecular mechanics, emphasizing their roles in understanding molecular behavior and interactions, particularly in drug discovery. Quantum mechanics provides insights into electronic structures and molecular properties through principles like wave-particle duality and the Schrödinger Equation, while molecular mechanics utilizes classical physics to model molecular systems and predict energies. Additionally, energy minimization and conformational analysis are highlighted as essential techniques for optimizing molecular structures and enhancing drug design accuracy.
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
0% found this document useful (0 votes)
27 views

CADD Unit 5

The document discusses quantum mechanics and molecular mechanics, emphasizing their roles in understanding molecular behavior and interactions, particularly in drug discovery. Quantum mechanics provides insights into electronic structures and molecular properties through principles like wave-particle duality and the Schrödinger Equation, while molecular mechanics utilizes classical physics to model molecular systems and predict energies. Additionally, energy minimization and conformational analysis are highlighted as essential techniques for optimizing molecular structures and enhancing drug design accuracy.
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
You are on page 1/ 14

QUANTUM MECHANICS

Quantum mechanics defines the behaviour of nuclei and electrons and it also explain the molecular
interactions in terms of distribution and motion.
It is necessary to describe the quantized energy level to understand the bonding orbitals of atoms and
molecules. Quantum methods utilize the principles of particle physics to examine structure as a
function of electron distribution.
It is based on the relisation that election and all material particles exhibit wave like properties.
Quantum mechanical (QM) methods are employed for conformer optimization, partial charge
calculation, and descriptor generation, enabling better scoring functions and binding affinity
predictions.
Quantum mechanical (QM) calculations can accurately predict ligand geometries, charges, tautomers,
and protonation states.
Principles of Quantum Mechanics in Chemistry
1. Wave-Particle Duality:
o Electrons and atoms behave both like particles and waves.
o This duality is described by wavefunctions (ψ), solutions to the Schrödinger
Equation.
2. Schrödinger Equation:
o A foundational equation of quantum mechanics:
H^ψ=Eψ
Ψ – Time dependent wave function
E ψ - Represent total potential and kinetic energy of all particles in structure
H – Hamiltonium operator acting on the wave function
o It gives the energy (E) and wavefunction (ψ) of a system.
o Helps predict the electronic structure, orbital shapes, and reactivity of molecules.
3. Quantization:
o Electrons occupy discrete energy levels (quantized orbitals).
o Transitions between levels explain absorption/emission spectra.
4. Pauli Exclusion Principle:
o No two electrons in an atom can have the same quantum state — key for
understanding electronic configuration.
5. Heisenberg Uncertainty Principle:
o You can't simultaneously know both the position and momentum of an electron
precisely.
o Important in modeling electron densities and probabilities.
6. Molecular Orbital Theory:
o Describes bonding using combinations of atomic orbitals into molecular orbitals.
o Important for drug-receptor interaction modeling.
The Schrödinger Equation describes how the quantum state (wavefunction, ψ\psiψ) of a system
evolves. It’s essentially the quantum version of Newton’s laws, but for subatomic particles like
electrons.
Time-independent QM methods are widely used in drug design to determine ligand conformations and
optimize geometries
There are two main forms:
1. Time-Dependent Schrödinger Equation (TDSE)

Equation:

• ψ(r,t) The wavefunction that depends on position r\mathbf{r}r and time ttt
• H^: The Hamiltonian operator (total energy operator = kinetic + potential energy)
• i: Imaginary unit
• ℏ: Reduced Planck’s constant
Time-Dependent Schrödinger Equation Describes how quantum states evolve over time.
• It’s essential when studying dynamic systems—for example, when electrons move or
molecules vibrate in real time.
Application in Chemistry:
• Used in spectroscopy, quantum dynamics, and reaction kinetics.
• Helps simulate how a molecule’s wavefunction changes when exposed to light or other fields
(like in photoactivation of drugs).
2. Time-Independent Schrödinger Equation (TISE)

Equation:

• Now the wavefunction ψ depends only on position—not time.


• E is the energy eigenvalue associated with a stationary state.
Equation Solves for stationary states (states that do not change in time).
• Provides the energy levels and spatial distribution of electrons in atoms/molecules.
Application in Drug Design:
• Density Functional Theory (DFT) and ab initio methods solve this equation to:
o Predict molecular geometries
o Calculate electrostatic potentials
o Identify reactive sites on drug molecules
o Model enzyme active sites using QM/MM

Applications in Predicting Chemical Behavior

Quantum mechanics helps in:

• Predicting molecular geometry and electron distributions (via DFT and ab initio
methods).

• Understanding reaction mechanisms, transition states, and activation energy.

• Calculating molecular properties like dipole moments, ionization potentials, and orbital
energies.

• Spectroscopy predictions (NMR, IR, UV-Vis spectra).

Role of Quantum Mechanics in Drug Discovery


Both documents emphasize how quantum mechanics has revolutionized computer-aided drug
design (CADD):
1. Ligand Optimization:
o QM refines ligand geometries and electrostatic potentials for better docking accuracy.
o QM-derived properties enhance virtual screening.
2. Protein-Ligand Binding:
o Used in QM/MM hybrid methods (Quantum Mechanics/Molecular Mechanics) to
study active sites.
o Enables understanding of electronic interactions in binding pockets.
3. Prediction of Reactivity and Metabolism:
o Identifies sites of metabolism, oxidation, or hydrolysis.
o Used in toxicity predictions and drug metabolism studies.
4. Free Energy Calculations:
o High-accuracy QM calculations contribute to binding free energy prediction, crucial
for hit-to-lead optimization.
5. Tautomer and Protonation State Prediction:
o QM helps resolve bioactive forms of molecules, crucial for docking accuracy.
MOLECULAR MECHANICS
Molecular mechanics describes the energy of a molecule in terms of simple function which accounts
for distortion from “ideal” bond distances and angles as well as for non bonded, vander waals and
coulombic interactions.
Principles of Molecular Mechanics
Molecular Mechanics (MM) is a classical approach to model molecular systems using Newtonian
physics. It treats atoms as spheres and bonds as springs. Instead of solving quantum mechanical
equations, it uses empirical equations (force fields) to describe molecular energies.
Molecular mechanics breaks down pair wise interaction into Bonded interaction (internal
coordination) i.e. Atoms that are connected via one I to three bonds and Non bonded interaction i.e.
Electrostatic and Van der waals component.
At its core, MM calculates the potential energy (E) of a system based on the positions of atoms using
a predefined force field:
E=E bonded+ E non-bonded
Bonded interactions
• Used for better approximate the interaction of the adjacent atoms.
• Calculations in the molecular mechanics is similar to the Newtonians Jaw of classical mechanics and
geometry as a function of steric energy. it will calculate
• f=kx, Hooke's law is applied here
F = force on the spring needed to stretch an ideal spring is proportional to its elongation x, and where
k is the force constant or spring constant of the spring.
EBonded= EBond + EAngle + EDihedral
Non bonded interaction
• Nearly applied to all pairs of atoms.
The nonbonded interaction terms usually include electrostatic interactions and van der waals
interaction, which are expressed as coloumbic interaction as well as Lennard-Jones type potentials,
respectively.
Enonbounded = Eelectrostatic + E vander waals
This is expanded as:

Force Fields
A force field refers to the functional forn and parameter sets used to describe the potential energy of a
system of particles.
The molecular mechanics energy expression consists of a simple algebraic equation for the energy of
the compound.
A set of the equations with their associated constants which are the energy expression is called a force
field.
Such equations describe the various aspects of the equation like stretching, bending, torsions,
electronic interactions van der waals forces and hydrogen bonding.
Importance of Force field:
Analysis of the energy contributions at the level of individual or classes of interactions.
Energy expression is the equation that describes the potential energy surface of a particular model as
a function of its atomic coordinates.
Key Parameters in Molecular Mechanics
1. Bond Stretching Energy (E_bond):
o Treated like a spring (Hooke’s Law).

o r: bond length, r0: equilibrium bond length, kb: bond force constant.
2. Angle Bending Energy (E_angle):

o Models changes in bond angles from their equilibrium values.


3. Dihedral/Torsional Energy (E_dihedral):

o Models rotation around bonds.


4. Van der Waals (E_vdW):
o Described using Lennard-Jones potential.

5. Electrostatic Interactions (E_electrostatic):


Applications in Predicting Behavior of Chemical Systems

1. Conformational Analysis:
o MM helps identify the most stable conformation of molecules.
2. Energy Minimization:
o Used to find low-energy structures (stable states) of molecules.
3. Molecular Dynamics:
o MM parameters are used in simulating the motion of molecules over time.
4. Prediction of Physical Properties:
o Geometry, bond lengths, angles, and strain energies can be predicted.
5. Macromolecule Modeling:
o Particularly useful in proteins, nucleic acids, and polymers.

Role of Molecular Mechanics in Drug Discovery

1. Lead Optimization:
o Predicts how small structural changes affect binding and stability.
2. Ligand-Protein Interactions:
o Evaluates binding affinities via docking and scoring functions.
3. Structure-Based Drug Design (SBDD):
o Uses MM to refine ligand-protein complexes post-docking.
4. Virtual Screening:
o Ranks compounds based on MM energy calculations.
5. Force Field Application:
o Different force fields (e.g., AMBER, CHARMM, OPLS) are used to model
biological macromolecules in a realistic manner.
6. Solvation Models:
o MM incorporates solvent effects implicitly (e.g., Generalized Born) or explicitly
for accurate predictions.
ENERGY MINIMIZATION
Energy minimization is a computational technique used to convert a molecular structure from a
potentially unrealistic or high-energy conformation to a more physically accurate, stable (low-energy)
structure. This process ensures that the 3D geometry of molecules is optimized, which is essential for
accurate prediction of molecular properties and interactions.

Why it’s important:

• Molecules in nature adopt minimum energy conformations.


• To compare computational predictions with experimental data, structures must be energy
minimized.
• It eliminates atomic clashes, unnatural strains, and inaccurate geometries.
• Used before simulations or property predictions in drug design, QSAR, and molecular
docking.

Methods Used in Energy Minimization


1. Steepest Descent Method
• Atoms are moved in the direction opposite to the force acting on them.
• Fast per step but less efficient overall, often needing many steps.
• Best for initial stages when atoms are far from equilibrium.
• Assumes constant second derivatives (Hessian not computed).
• Computationally inexpensive but can trapped in local minima.
• Example: Used to minimize energy of Glyburide from 157.36 to 29.52 kcal/mol.
2. Conjugate Gradient Method
• More efficient than steepest descent.
• Uses information from previous steps to guide the direction of minimization.
• Good for systems closer to the energy minimum.
• Fastest convergence & better handling of local minima.
3. Newton-Raphson Method
• Very accurate near the minimum.
• Uses second derivatives (Hessian matrix), so it's computationally expensive.
• Fast convergence but not used for large systems due to cost.
4. Simulated Annealing
• Combines random sampling and energy minimization.
• Helps to find the global minimum by avoiding traps in local minima.
• Temperature is gradually reduced to settle into a low-energy state.
5. Molecular Dynamics Based Minimization
• Uses time-dependent simulation data to guide structure relaxation.
• Useful for complex systems like proteins and nucleic acids.
• Computationally expensive
Parameters Involved in Molecular Mechanics (for Energy Calculation)
Energy minimization relies on force fields that define mathematical functions to calculate the total
energy. Parameters include:
1. Bond Stretching – Deviation from ideal bond lengths.
2. Angle Bending – Changes in bond angles from equilibrium.
3. Dihedral/Torsional Angles – Rotations around bonds.
4. Improper Torsion – Maintains planarity or chirality.
5. Van der Waals Interactions – Non-bonded interactions based on atom distances.
6. Electrostatics – Coulombic forces between partial charges.
7. Hydrogen Bonds – Special type of electrostatic interaction.
8. Torsion-Stretch & Bend-Bend Coupling – Complex effects due to connected movements.
These terms are summed to compute total potential energy of the molecule.
Role in Drug Discovery
Energy minimization plays a critical role in computer-aided drug design (CADD):
• Produces realistic 3D conformations of drug molecules.
• Prepares ligand and receptor structures for docking.
• Improves binding predictions and interaction analysis.
• Helps in structure-activity relationship (SAR) studies by aligning low-energy conformers.
• Enables exploration of conformational space of drug candidates.
• Contributes to lead optimization by assessing how structure affects energy and binding.
CONFORMATIONAL ANALYSIS

What is Conformational Analysis?

Conformational analysis is the study of the three-dimensional (3D) spatial arrangements of atoms in a
molecule that arise due to rotation around single (sigma) bonds. These different spatial arrangements
are called conformers or rotamers.
Unlike structural isomers, conformers do not involve breaking bonds but result from internal
rotations. Since atoms are not static in space, conformational analysis evaluates all the possible low-
energy shapes a molecule can adopt.

Purpose and Importance of Conformational Analysis

1. Determination of Stable Geometry:


o Molecules can adopt multiple conformations, but under physiological conditions, they
tend to exist in low-energy or stable conformations. Conformational analysis helps
identify these stable forms.
2. Influence on Biological Activity:
o Many biomolecular interactions (e.g., drug-receptor binding) depend heavily on the
correct conformation of the ligand (drug) and receptor. An incorrect conformation
may prevent binding or reduce efficacy.
3. Structure–Activity Relationship (SAR):
o Conformation plays a vital role in SAR studies, where even minor changes in shape
can significantly affect activity, selectivity, and toxicity.
4. Pharmacophore Modeling:
o Defines the spatial arrangement of essential features (hydrogen bond
donors/acceptors, hydrophobic regions, aromatic rings) necessary for biological
activity.

Key Strategies in Conformational Analysis

1. Rigid Geometry Approximation:


o Assumes that bond lengths and bond angles remain constant.
o Only torsional angles (around single bonds) are varied to generate conformers.
o Simplifies computational cost while focusing on key flexible regions.
2. Rigid Body Rotation:
o Rotates groups of atoms or entire fragments (as rigid bodies) around bonds.
o Especially useful in docking studies where ligands need to fit into rigid active sites.
3. Conformational Clustering:
o After generating many conformations, clustering techniques are used to group similar
conformations.
o Representative conformers from each cluster are chosen for further analysis, saving
computational resources.
4. Conformational Restriction:
o Involves chemically locking a molecule in its active conformation (e.g., through ring
formation or double bonds).
o Prevents the molecule from adopting inactive forms, improving specificity and
potency.

Electronic Properties in Conformational Analysis

Electronic properties help predict how molecules behave in chemical and biological
environments:
• Molecular Orbitals: HOMO (highest occupied molecular orbital) and LUMO (lowest
unoccupied molecular orbital) predict reactivity.
• Electron Density: Indicates the probability of finding electrons around atoms.
• Electrostatic Potential (ESP):
o Indicates regions of positive and negative charge.
o Used to determine how molecules will interact via hydrogen bonding, dipole-dipole,
and ionic interactions.

Applications in Drug Design

1. Structure-Based Drug Design (SBDD):


o Conformational analysis helps in accurate molecular docking by predicting the most
likely binding pose of a ligand.
o Ensures that the ligand binds in the bioactive conformation.
2. Ligand Flexibility in Docking:
o Ligands can adopt many conformations; evaluating these helps in understanding all
possible binding modes with a receptor.
3. Lead Optimization:
o Helps modify lead compounds to lock the active conformation, thereby enhancing
binding affinity and selectivity.
o Reduces the entropic cost of binding.
4. Virtual Screening:
o Multiple conformations of a ligand are screened to identify which conformer has the
best fit in the receptor binding pocket.
5. Toxicity Prediction:
o Some conformations may allow off-target binding, leading to toxicity. Avoiding such
conformers can help improve safety profiles.
6. Pharmacokinetics and ADMET:
o Molecular conformation can influence solubility, permeability, and metabolism.
Example: Conformational Analysis of Cyclohexane
• Cyclohexane exists in chair, boat, and twist-boat conformations.
• The chair conformation is the most stable due to minimal torsional strain.
• Substituents prefer the equatorial position to minimize 1,3-diaxial interactions.
• This principle is extrapolated in drug molecules with ring systems.

Half chair Twisted boat

GLOBAL CONFORMATION MINIMA DETERMINATION


Concept of Global Minima
In the context of molecular modeling and drug design, the concept of global minima is crucial in
understanding the most stable conformation of a molecule.

What is a Global Minimum?

• A global minimum refers to the lowest energy conformation that a molecule can adopt among
all its possible conformations.
• Molecules can exist in various rotational or spatial arrangements due to the flexibility of
bonds, especially in complex drug molecules.
• Each conformation has an associated potential energy, and when plotted, this forms a
Potential Energy Surface (PES).
• It reflects the most thermodynamically stable structure under a given set of conditions (e.g.,
in vacuum, solvent, or biological environment).
• Finding this conformation allows scientists to:
o Understand how a drug behaves in a free state.
o Use it as a starting point for docking and QSAR studies.
o Compare it with the bioactive conformation.
How is Global Minimum Conformation Determined?
The process involves systematic conformational search and energy minimization using molecular
mechanics or quantum mechanics tools.

1. Generation of Conformers

First, the molecule must be allowed to explore its conformational space:


• Systematic Search: Rotates all rotatable bonds in increments (e.g., every 30°) to generate all
possible conformers.
• Random Search: Randomly samples torsion angles.
• Monte Carlo Methods: Use random perturbations with statistical acceptance.
• Molecular Dynamics (MD): Simulates how the molecule moves over time, capturing diverse
conformations.
Molecular, dynamics is a molecular mechanics program designed to mimic the movement of
atoms within a molecule . The force acting on each atom are then calculated by considering
bond lengths, bond angles, torsinol term and non bonded interaction with surrounding atoms.

• Genetic Algorithms: Mimic biological evolution to generate optimized structures.

2. Energy Evaluation of Each Conformer

Each generated structure is evaluated for its total potential energy using a force field (e.g., MMFF,
AMBER, CHARMM):
• Includes:
o Bond stretching
o Angle bending
o Torsional rotation
o Van der Waals interactions
o Electrostatics (Coulombic interactions)
3. Energy Minimization

Each conformer is subjected to local energy minimization using optimization algorithms:


• Steepest Descent: Fast but less accurate; used for rough minimization.
• Conjugate Gradient: Better convergence; commonly used.
• Newton-Raphson: Very precise; used for small systems or final polishing.
Minimization continues until the system reaches a local minimum (where the gradient of energy = 0).

4. Comparison and Identification of Global Minimum

• Among all minimized conformers, the one with the lowest total potential energy is
considered the global minimum.
• Energy graphs or Potential Energy Surfaces (PES) help visualize where each conformer
lies.
• In reality, several conformers may be close in energy — hence clustering and Boltzmann
distribution can also be considered.

Energy Landscape:

• The PES represents a multidimensional landscape where:


o Valleys represent low-energy conformations (minima),
o Peaks represent high-energy conformations (transition states or unstable
conformations).
• Among all the minima, the global minimum is the deepest valley — the most stable structure
in terms of thermodynamics.

Importance in Drug Design:

• Determining the global minimum is crucial because:


o It provides insights into the most probable conformation a drug molecule would adopt
in solution.
o It aids in accurate molecular docking, QSAR studies, and pharmacophore modeling.
Relationship Between Global Minimum Conformation and Bioactive Conformation
While the global minimum represents the most stable conformation in isolation, the bioactive
conformation is the specific 3D shape a drug adopts when bound to its target (like an enzyme or
receptor).

Key Differences:

Feature Global Minimum Conformation Bioactive Conformation

Definition Lowest energy conformation in Conformation when bound to biological


isolation target

Stability Thermodynamically most stable May not be the most stable


Environment Determined in solvent or vacuum Determined within the target's binding site

Relationship and Significance:

• The bioactive conformation may or may not be the global minimum.


• Often, drug molecules adopt higher-energy conformations to fit into the binding site of a
receptor — this is possible due to the binding energy compensating for the conformational
energy penalty.
• The binding interaction (hydrogen bonds, hydrophobic interactions, etc.) can stabilize these
higher-energy conformations within the receptor.

Example:

• A drug might have a global minimum conformation in water due to solvent interactions, but
when it binds to a hydrophobic pocket in a protein, it might adopt a different conformation
(the bioactive one) that better fits the binding site.

Implications in Drug Design:

• Understanding this relationship helps medicinal chemists to:


o Modify drugs to reduce the energy gap between global minimum and bioactive forms.
o Design rigid analogs that prefer the bioactive conformation, increasing binding
efficiency.
o Use conformational analysis and molecular dynamics to simulate and predict the
drug’s behavior both in free form and in complex with targets.

You might also like