Aditya S. Kakad
1
Molecular Pharmaceutics ( NTDS )
1) Targeted Drug Delivery System : Concepts & Biological process
involved in drug Targeting.
 Concepts
TDDS is a special form of DDS where medicament is selectively
targeted or delivered only to its site of action or absorption and not to
the non-target organs or tissues or cells.
It is a method Patient in a g delivering medication to a manner that
increases the concentration of the medication in parts of the body
relative to others.
Need of TDDS
a) Pharmacokinetic Reason
 Poor absorption
 host half-life
 Large volume of distribution.
b) Pharmaco dynamic Reason
 Low specificity
 low therapeutic index
c) Pharmaceutical Reasons
 Drug instability .
 Low Solubility
Concept of Targeting
The concept of drug targeting has been originated from the
perception of paul ehrlich, who proposed dong delivery to be as a
"magic bullet"
Gregoriadis, 1981 described drug targeting using novel DDS as
"old drugs in new clothes"
 Biological process involved in drug Targeting.
Events and Biological Process involved in Dreng targeting.
a) Cellular uptake and processing
b) Transport across the epithelial barrier.
c) extravasations
d) Lymphatic uptake...
Aditya S. Kakad
2
a) Cellular uptake and processing
Low molar mass drugs can enter into or pass through Various Cells
by simple diffusion process
Targeted DD usually have macro molecular assemblies hence
cannot enter by such simple process, hence taken up by A process Called
endocytosis.
 Steps involved endocytosis :-
i. Intermaligation of the plasma membrane
ii. Concomitant with engulment of extra Cellular material.
 Types of endocytosis :-
i. Phagocytosis
Compared with
phagocytosis pinocytosis is
a universal Phenomenon in all the cells, Pinocytosis does not
require any external stimuli.
ii. Pinocytosis
Types :-
 fluid phase pinocytosis
 Absorptive pinocytosis.
Compared with phagocytosis fluid phase pinocytosis Capture
molecules slowly being directly proportional to the concentration
and Size dependant.
b) Transport across the epithelial barrier.
 The oral, Buccal, nasal, vaginal and rectal Cavities. are internally
lined with one or more layers of epithelial cells.
 Depending on the position and function in the body epithelial cells
Can be varied forms.
 Epithelial
 Lamia propria
 Basal lamina.
 low molar mass drugs Cross the above by passive diffusion Carrier
mediated Systems and selective and nou selective endocytosis
 The polar materials diffuse through tight junctions of epithelial
cells.
 Passive transport is usually higher in damaged mucosa where as
active transport depends on structural integrity of epithelial cells.
 Positively charged particles showed increased uptake than
negatively charged Counterparts.
 Absorption of drugs from buccal via transcellular and paracellular
later being dominant
Aditya S. Kakad
3
c) Extravasations
Many disease are Known that Hesult from dysfunction Cells
outside the Cardiovascular System, thus for a for a drug to exert its
therapeutic effect, it must come out from the Central Circulation and
interact with extra cellular or intracellular targets.
The process of trans cellular exchange is called as
extravasation, which is governed by blood. Capillary Walls.
Permeability of Capillaries are Controlled by :
• Structure of the Capillary wall
• Pathological Condition
• Rate of blood and lymph supply
• Physicochemical factors of drug.
Depending on the morphology and Continuity of the
endothelial layer and the basement membrane blood Capillaries are
divided into :
• Continous
• Fenestraded
• Sinusoidal
d) Lymphatic uptake...
 extravasion dong molecules Can be either reabsorbed into the
blood stream directly or enter into the Lymphatic system and
return with the Lymph to the blood Circulation
 Drugs administered by subcutaneous intracellular transdermal
peritoneal routes Can reach the Systemic Circulation by lymphatic
system
2) Brain Specific delivery.
Drug delivery to the brain is the process of passing therapeutically
active molecules across the Blood- Brain-Barrier.
This is a complex process that must take into account the complex
anatomy of the brain as well as the restrictions imposed by the special
functions of the BBB
Various routes of administration as well as conjugations of drugs, e.g.
with liposome and nanoparticles are Considered.
 Novel Approaches
A. Invasive approaches or Neurosurgical approaches:
i. Intra - cerebral injection/implant
ii. Intra - cerebro ventricular (ICV) infusion
iii. Disruption of the BBB
iv. Convection-enhanced delivery (CED)
Aditya S. Kakad
4
B. Non - Invasive approaches :
i. Pharmacological techniques:
1. Chemical Techniques
a) Prodrugs
b) Drug Conjugates
2. Colloidal Techniques
a) Nanoparticles
b) Liposomes
ii. Biological / Physiological Techniques:
1. Pseudo nutrients
2. Antibody
3. Chimeric peptides
C. Miscellaneous approaches :
i. Intranasal delivery
ii. Iontophoretic delivery
 Blood Brain Barrier
BBB is a highly selective permeability barrier that separates the
Circulating blood from the brain extracellular fluid (BECF) in the CNS.
BBB is Composed of high density cells, restricting passage of substances
from the blood to the brain.
Parameters Considered optimum for a compound to transport across
the BBB are :
• Compound should be un ionized
• Approx. log p value must be 2
• Molecular weight must be less than 400 Da
• Cumulative no of H-bonds must not go beyond 8-10
• It is estimated only 2% of small molecular wt. drug will cross BBB. .
 Strategies :
Aditya S. Kakad
5
3) Nano Particles : Types, Preparation & evaluation.
Nanoparticles are defined as particulate dispersions or Solid particles with a size in
the range of 10-1000nm.
Drug is confined to a cavity surrounded by a unique polymer layer / membrane
called nano Capsules. While nanosphores are matrix system in which the drug is
physically and uniformly dispersed.
NANO CAPSULES : The nano Capsules are system in which the drug is confined to
Surrounded by a Cavity unique polymer membrane
NANOSPHERES: The nanosphere are matrix system in which the physically and
uniformly drug is dispersed.
 Ideal properties of Nanoparticles :
• Should be stable in blood
• Should be biodegradable
• Should be non toxic
• Should be non-immunogenic
• Should be non- thrombogenic
• Should be non-inflammatory
 Types of nanoparticles
• One dimensional nanoparticles.
• Two dimensional nanoparticles
• Three dimensional nanoparticles fullerenes
• Quantum dots.
 Preparation of Nanoparticles.
1. SOLVENT EVAPORATION METHOD
 In this method firstly nano emulsion formulation are Prepared
 Polymer is dissolved after the 1st step in organic solvent
(dichloromethane, chloroform or ethyl acetate)
 Drug is dispersed in the solution prepared Then this mixture is
emulsified in an aqueous phase which contains surfactant
(polysorbates, Poloxamers, Polyvinyl alcohol.)
 An oil/water emulsion is prepared by using mechanical stirring,
sonification or micro fluidization.
2. DOUBLE EMULSIFICATION METHOD
 Emulsification and evaporation method have limitation of poor
entrapment of hydrophilic drugs, hence double emulsification
technique is used.
 Firstly w/o emulsion is prepared by addition of aqueous drug
solution to organic polymer with Co with continuous stirring.
 This prepared emulsion is mixed with another aqueous phase
with vigorous stirring, Hesultanting w/o/w emulsion
 Organic solvents are removed by high speed centrifugation.
Aditya S. Kakad
6
3. EMULSIONS - DIFFUSION METHOD
 it is a modified form of salting out method.
 In this technique, polymer is dissolved in water miscible
solvent (propylene Carbonate, benzyl alcohol, and is saturated
with water.
 Polymer-water saturated solvent phase is then emulsified in an
aqueous solution conteining stabilizer
 Afterwards, solvent is removed by evaporation or filtration.
4. NANO PRECIPITATION METHOD
 In this method precipitation of polymer and drug obtained
from organic solvent and the organic solvent diffused in to the
aqueous medium with or without presence of surfactant
 Firstly drug was Co-solvent was dissolved in water, and then
added into the solution
 The another solution of polymer (ethyl cellulose) and
propylene glycol with chloroform is prepared and this solution
was dispersed to the drug solution
 This dispersion was slowly added to 10ml of 7o % aqueous
ethanol-solution
 After 5 minutes of mixing, the organic solvents. were removed
by evaporation at 35° under normal pressure, nanoparticles
were separated by using cooling centrifuge (10000 rpm for 20
min)
 Supernatants were removed and nanoparticles are washed
with water and dried at room temp, in a desicator
5. COACERVATION METHOD
 In this method, drug and protein -solution (2 % w/v) is
incubated for one hour at room temperature and pH adjusted
to 5.5
 Ethanol was added to the prepared solution in 2:1 ratio (v/v)
 Resultant Coacerved is left to hardened with 25 %
glutaraldehyde (1.56 ug/mg) for 2 hours which allow Cross
Linking of protein
 Organic solvents are removed by notary vacuum evaporation
at reduced pressure are and nanoparticles Collected and
purified by centrifugation
 Pellets of nanoparticles were then suspended in phosphate
buffer and lyophilized with mannitol
Aditya S. Kakad
7
6. SALTING OUT METHOD
 Salting out method is Very close to solvent diffusion method.
 This technique is based on the separation of water-miscible
solvent from aqueous solution by salting out effect.
 Generally acetone is used because it is totally miscible with
water and easily removed
 Polymer and drug are dissolved in a solvent which emulsified
into a aqueous solution Containing salting out agent.
7. DIALY SIS
 Dialysis is an effective method for preparation of nanoparticles
 In this method, firstly polymer (such as poly (benzyl-
glutamate)-b-poly (ethylene oxide), poly (lactide) and drug
dissolved in an organic Solvent
 This solution is added to a dialysis tube and a dialysis is
performed.
 Evaluation OF Nanoparticles.
Nano particles are evaluated by the following Method:
1. Particle Size
2. Surface area
3. Surface charge
4. Density
5. Molecular weight
6. Nano Particle yield
7. Dry entrapment efficiency
8. Invitro release.
4) Monoclonal Antibodies : Types, Preparation, Evaluation &
Application
 Monoclonal antibodies Can be defined as a type of antibody derived
from hybridoma Cells.
 Monoclonal antibodies are "antibodies that are identical because they
are produced by Single B-Cell clone.
 Monoclonal antibodies are homogenous immunological regents of
defined specificity, So that these can be utilized for diagnosis and
screening with ease and Certainty.
 Types :
1. Murine.
2. Chimeric.
3. Humanized.
4. Fully Human.
Aditya S. Kakad
8
 Preparation :
 Monoclonal antibodies production (mAb) is produced by Cell lines or
clones obtained from the immunized animals with the substances to be
studied.
 Cell lines are Produced by fusing B-Cells from the immunized animal with
myeloma Cells.
 To produce the desired MAB, the celle must be grown in either of two
ways:
 By injection into the peritoneal Cavity in mouse (In Vivo)
 By in vitro tissue Culture
 The Vitro tissue Culture is the method used when the Cells are placed in
culture outside the mouse the mouse's body in flask.
 Evaluation :
1. Characterisation of monoclonal antibodies
 Physicochemical characterisation
 Immunological properties
 Biological activity
 Purity, impurity and contaminants
 Quantity
2. Specifications
 Identity
 Purity and impurities
 Potency
 Quantity
 General tests
 Application
1. Measuring protein and drug level in serum
2. Identifying infectious agents.
3. Identifying and quantifying hormones
4. Biochemical analysis - Pregnancy , cancer
5. Identifying tumors agent & also antibodies
6. MAbs as targeting agents.
7. Diagnostic imaging
8. Protein Purification
5) Niosomes : Types, Preparation, Evaluation & Application
 Niosomes are novel drug delivery system in which the medication is
encapsulated in a Vesicle.
 The Vesicle is composed of a bilayer non-ionic surface active agents and
hence the name niosomes.
 Niosomes are very small and microscopic in size. Their size lies in the
nanometric scale. Although Structurally similar to liposomes, they offer
several advantages over them.
Aditya S. Kakad
9
 Types :
1. SUV (small unilameller vesicles)
2. LUV (large unilameller vesicles)
3. MLV (multi lammeler vesicles)
 Method of preparation:
Ether injection Method
 Surfactant + Cholesterol is dissolved in directly ether
 Then injected in warm water maintained at 60 o
C through a 14
gauze needle
 Ether is vaporized to form single layered niosomes.
Hand Shaking Method (thin film hydration technique)
 Surfactant + Cholesterol + Solvent
 Remove organic Solvent at room temperature
 Thin layer formed on the walls of flask
 film Can be rehydrated to form multilameller niosomes.
Sonication Method:
 Way in buffes + Surfactant / cholesterol in 10ml of aqueous
phase
 Above mixture is Sonicated for 3 minutes at 60 oC using
titanium probe yielding niosomes.
Multiple membrane extrusion method.
 Mixture of surfactant, Cholesterol in chloroform is made into
thin film by evaporation.
 The film is hydrated with aqueous drug solution and the
resultant suspension extruded through polycarbonate
membrane. It is a good method for Controlling niosomes size.
The bubble method
 It is novel technique for the one step preparation of
liposomes and niosomes without the use of organic Solvents.
 The bubbling unit Consists of round-bottomed flask with three
neck positioned in water bath to Control the temperature.
 Water Cooled reflux and thermometer is positioned in the
first and second neck and nitrogen Supply through the third
neck.
 Cholesterol and Surfactant are dispersed together in the
buffer (pH 7.4) at 70oC, the dispersion mixed for 15 Seconds
with high shear homogenizer & immediately afterward
bubbled at 70oC using nitrogen gas
Aditya S. Kakad
10
 Evaluation :
A. Size, Shape and Morphology
Freeze Fracture Electron Microscopy:- Visualize the vesicular
structure of surfactant based vesicles.
Photon Correlation Spectroscopy :- Determine mean diameter of the
vesicles. Electron Microscopy :- Morphological studies of vesicles.
B. Entrapment efficiency
After preparing niosomal dispersion, unentrapped drug is
separated by dialysis and the drug remained entrapped in niosomes is
determined by complete vesicle disruption using 50% n-propanol or
0.1% Triton X-100 and analysing the resultant solution by appropriate
assay method for the drug.
C. Vesicle Surface Charge
Determined by measurement of electrophoretic mobility and
expressed in expressed in terms of zeta potential
D.In vitro studies
 Applications
1. Drug Targeting
2. In Diagnosis
3. Anti Neoplastic Treatment
4. Delivery of peptide drug
5. Niosomes are carriers of Hemoglobuline
6) Aerosols: Propellents (Nomenclature rule with Eg.);
Containers types, Preparation & Evaluation
 Propellants
Propellants are the driving force in aerosol systems, responsible
for expelling the product from its container. They are classified into
three main types:
1. Liquefied Gas Propellants
 These are gases that become liquid under pressure. Common
examples include
 Hydrocarbons (eg, propane, butane, isobutane)
 Chlorofluorocarbons (CFCs) (eg, trichlorofluoromethane,
dichlorodifluoromethane, dichlorotetrafluoroethane)
 Hytirofluorocarbons (HFCs) (eg, 1,1,1,2-tetrafluoroethane, 1.1-
difluoroethanel
Aditya S. Kakad
11
2. Compressed Gas Propellants
 These are gases compressed into their containers and include
 Carbon dioxide (CO)
 Nitrogen (N)
 Nitrous oxide (NO)
3. Hybrid Propellants
 Combination of liquefied gas and compressed gas propellants to
achieve specific properties
 Containers
Aerosol containers are designed to withstand the pressure of the
propellant and ensure product stability. Types include:
1. Tin-Plated Steel Cans
 Used for a variety of aerosol products
 Offer durability and are corrosion resistant when lined.
2. Aluminum Cans
 Lightweight corrosion resistant, and used for pharmaceuticals
and food products.
3. Glass Containers:
 Used less frequently due to fragility, but suitable for certain
pharmaceuticals and cosmetic aerosols
 Typically used with protective outer coverings or coatings
 Plastic Containers
 Made from materials like polyethylene or polypropylene
 Limited use due to permeation and compatibility issues with
certain propellants.
 Preparation of Aerosols
1. Formulation:
 Active ingredient is dissolved or suspended in a suitable solvent.
 Propellant is selected based on product requirements.
 An appropriate container and valve system are chosen.
2. Filling:
 Cold Filling: Product concentrate and propellant are cooled to -30
to -60°C, then filled into the container.
Aditya S. Kakad
12
 Pressure Filling: Product concentrate is filled at room
temperature, followed by adding the propellant under high
pressure,
3. Sealing:
 The container is sealed with a valve crimped onto the can.
4. Testing:
 Leakage and pressure tests to ensure integrity and safety.
 Homogeneity and particle size distribution tests to ensure
consistent product delivery.
 Evaluation of Aerosols
1. Spray Pattern:
 Ensures that the aerosol dispenses uniformly.
2. Particle Size Distribution:
 Evaluated using methods like laser diffraction to ensure
therapeutic efficacy (for inhalation aerosols).
3. Dose Uniformity:
 Ensures consistent delivery of the active ingredient per actuation.
4. Leakage Test:
 Ensures the container does not leak propellant or product over
time,
5. Pressure Test:
 Confirms the container can withstand the pressure of the
propellant.
6. Valve Delivery Rate:
 Measures the amount of product delivered per actuation to
ensure proper dosing.
7) Ex-vivo & In-vivo gene therapy.
 Ex-Vivo Gene Therapy
Ex-vivo gene therapy involves the genetic modification of cells
outside the body and then reintroducing them into the patient. This
method is particularly useful for cells that can be easily extracted,
cultured, and reintroduced, such as blood cells.
Aditya S. Kakad
13
 Steps in Ex-vivo Gene Therapy:
1. Cell Collection:
 Cells are harvested from the patient. Common sources include
hematopoietic stem cells from bone marrow or peripheral blood.
2. Genetic Modification:
 The collected cells are cultured and genetically modified in vitro.
This can be done using various vectors, such as:
 Viral Vectors: Retroviruses, lentiviruses, adenoviruses, and
adeno-associated viruses (AAV) are commonly used to deliver the
therapeutic gene.
 Non-viral Methods: Techniques like electroporation, liposome-
mediated transfer, and CRISPR/Cas9 can be used.
3. Selection and Expansion:
 Modified cells are selected and expanded in culture to increase
their number.
4. Reintroduction:
 The genetically modified cells are reintroduced into the patient,
typically through infusion.
5. Engraftment and Function:
 The modified cells engraft into the patient's tissues and begin to
produce the therapeutic effect.
 Applications of Ex-vivo Gene Therapy:
 Hematological Disorders: Treatment of severe combined
immunodeficiency (SCID), beta- thalassemia, and sickle cell
disease.
 Metabolic Disorders: Enzyme replacement in conditions like
adenosine deaminase deficiency.
 Cancer: CAR-T cell therapy for certain types of leukemia and
lymphoma.
 Advantages:
 Controlled environment for genetic modification.
 Ability to select and expand modified cells.
 Reduced risk of immune response against the vector.
Aditya S. Kakad
14
 Challenges:
 Complex and time-consuming process.
 Requires specialized facilities and expertise.
 Limited to cells that can be extracted, cultured, and
reintroduced.
 In-Vivo Gene Therapy
In-vivo gene therapy involves delivering genetic material directly
into the patient's body. This approach targets cells within the body and
does not require cell extraction and reintroduction.
 Steps in In-vivo Gene Therapy:
1. Vector Design:
 A suitable vector is designed to carry the therapeutic gene.
Common vectors include:
 Viral Vectors: Adenoviruses, AAV, lentiviruses, and herpes
simplex viruses (HSV).
 Non-viral Vectors: Naked DNA, plasmids, nanoparticles, and
liposomes.
2. Delivery:
 The vector carrying the therapeutic gene is administered to the
patient. Delivery routes can vary:
 Intravenous (IV): For systemic delivery.
 Intramuscular (IM): For targeting muscle tissue.
 Intrathecal: For delivery to the central nervous system.
 Inhalation: For respiratory diseases.
 Local Injection: For specific tissues or organs.
3. Cellular Uptake and Expression:
 The vector transduces the target cells, and the therapeutic gene
is expressed, leading to the desired therapeutic effect.
 Applications of In-vivo Gene Therapy:
 Genetic Disorders: Treatment of Duchenne muscular dystrophy,
hemophilia, and cystic fibrosis.
 Ocular Diseases: Gene therapy for Leber's congenital amaurosis
and retinitis pigmentosa.
Aditya S. Kakad
15
 Neurological Disorders: Treatment of spinal muscular atrophy
and Parkinson's disease.
 Cardiovascular Diseases: Delivery of angiogenic genes for
ischemic heart disease.
 Advantages:
 Direct delivery to target tissues.
 Suitable for a wide range of tissues and organs.
 Simpler and faster compared to ex-vivo methods.
 Challenges:
 Potential for immune responses against the vector.
 Difficulty in targeting specific cells or tissues.
 Risk of off-target effects and insertional mutagenesis.
8) Potential target disease for gene therapy (Inherited disorder
& cancer)
Gene therapy is a cutting-edge medical approach that involves
modifying a person’s genes to treat or prevent disease. It holds promise
for a variety of diseases, particularly inherited disorders and cancer.
 Inherited Disorders
1. Cystic Fibrosis (CF):
o Problem: Affects the lungs and digestive system due to a defective
gene that causes mucus to become thick and sticky.
o Gene Therapy Goal: Introduce a healthy version of the CFTR gene
to help produce normal mucus and improve lung function.
2. Hemophilia:
o Problem: A bleeding disorder caused by a deficiency in specific
clotting factors due to faulty genes.
o Gene Therapy Goal: Deliver a working copy of the defective gene
to help the body produce the missing clotting factors and prevent
excessive bleeding.
3. Duchenne Muscular Dystrophy (DMD):
o Problem: A severe muscle-wasting disease caused by mutations in
the dystrophin gene.
o Gene Therapy Goal: Introduce a functional version of the
dystrophin gene to help build and repair muscle tissue.
Aditya S. Kakad
16
4. Sickle Cell Disease:
o Problem: Red blood cells become misshapen and break down due
to a mutation in the hemoglobin gene.
o Gene Therapy Goal: Correct or replace the defective hemoglobin
gene to produce healthy red blood cells.
 Cancer
1. Leukemia:
o Problem: A type of cancer that affects blood and bone marrow.
o Gene Therapy Goal: Use modified immune cells (CAR-T cells) to
target and destroy cancerous cells in the blood.
2. Glioblastoma:
o Problem: An aggressive form of brain cancer.
o Gene Therapy Goal: Introduce genes that make cancer cells more
sensitive to treatments like chemotherapy or trigger the immune
system to attack the tumor.
3. Melanoma:
o Problem: A dangerous form of skin cancer that can spread to
other parts of the body.
o Gene Therapy Goal: Employ gene editing techniques to modify
immune cells so they can better recognize and attack melanoma
cells.
4. Prostate Cancer:
o Problem: A common cancer in men that affects the prostate
gland.
o Gene Therapy Goal: Introduce genes that target and kill cancer
cells or make them more responsive to other treatments.
9) Liposomal gens delivery systems
Liposomal gene delivery systems are a way to transport genetic
material (like DNA or RNA) into cells using tiny, fat-like particles called
liposomes. This method helps deliver genes safely and effectively to
treat or prevent diseases.
 How Liposomal Gene Delivery Works
1. Creating Liposomes:
o Liposomes are small, spherical vesicles made from phospholipids,
which are the same types of fats found in cell membranes.
o These vesicles can encase genetic material, protecting it from
degradation.
Aditya S. Kakad
17
2. Loading the Genes:
o The genetic material, such as a piece of DNA or RNA, is placed
inside the liposome.
o This can include genes needed to replace faulty ones, or RNA that
can silence harmful genes.
3. Delivery to Cells:
o Once loaded, the liposomes are introduced into the body.
o Because they are made of similar materials to cell membranes,
liposomes can merge with cells and release their genetic payload
inside.
4. Gene Expression:
o After the genetic material is delivered into the cell, it can be used
by the cell’s machinery.
o This can result in the production of a needed protein, correction
of a genetic defect, or silencing of a harmful gene.
 Benefits of Liposomal Gene Delivery
 Protection: Liposomes protect genetic material from being broken down
by enzymes in the body.
 Efficiency: They facilitate the entry of genes into cells more efficiently
compared to direct injection.
 Biocompatibility: Made from natural materials, liposomes are generally
non-toxic and less likely to trigger an immune response.
 Targeting: Liposomes can be modified to target specific types of cells,
improving the precision of the treatment.
 Applications
1. Inherited Disorders:
o Liposomal systems can be used to deliver functional genes to
correct genetic defects, such as in cystic fibrosis or hemophilia.
2. Cancer:
o They can transport genes that make cancer cells more susceptible
to treatment or enhance the body’s immune response to cancer.
3. Vaccines:
o Liposomes can deliver RNA vaccines, such as those used for
COVID-19, to instruct cells to produce antigens and trigger an
immune response.
Aditya S. Kakad
18
10)Therapeutic Antisense Molecules (with application)
Therapeutic antisense molecules are a type of treatment that can
specifically target and block the activity of certain genes. These
molecules are designed to bind to the messenger RNA (mRNA) produced
by a gene, preventing it from making a protein that could cause disease.
 How Antisense Molecules Work
1. Structure:
o Antisense molecules are short, single strands of DNA or RNA.
o They are complementary to a specific mRNA sequence, meaning they
can bind to it perfectly.
2. Binding to mRNA:
o When an antisense molecule binds to its target mRNA, it prevents the
mRNA from being used to make a protein.
o This process is like putting a "block" on the instructions for making the
protein.
3. Blocking Protein Production:
o By blocking the mRNA, antisense molecules can reduce or eliminate the
production of harmful proteins.
o This can help treat diseases caused by the overproduction or abnormal
production of certain proteins.
 Applications of Therapeutic Antisense Molecules
1. Genetic Disorders:
o Example: Duchenne Muscular Dystrophy (DMD)
 How it helps: Antisense molecules can help restore the
production of a functional dystrophin protein, which is missing
or defective in DMD patients.
 Outcome: Improved muscle function and slowed disease
progression.
2. Cancer:
o Example: Bcl-2 Protein in Cancer
 How it helps: Antisense molecules can target and reduce the
production of Bcl-2, a protein that helps cancer cells survive.
 Outcome: Increased effectiveness of cancer treatments and
reduced tumor growth.
3. Viral Infections:
o Example: Cytomegalovirus (CMV) Infections
 How it helps: Antisense molecules can inhibit the
replication of viral mRNA, reducing the viral load in infected
individuals.
 Outcome: Better control of the infection and fewer
symptoms.
Aditya S. Kakad
19
4. Cholesterol Management:
o Example: High Cholesterol (Familial Hypercholesterolemia)
 How it helps: Antisense molecules can target and reduce
the production of apolipoprotein B (ApoB), a key
component in the formation of bad cholesterol (LDL).
 Outcome: Lower levels of LDL cholesterol and reduced risk
of heart disease.
 Benefits of Therapeutic Antisense Molecules
 Precision: They can specifically target and silence disease-causing genes
without affecting other genes.
 Versatility: Applicable to a wide range of diseases, including genetic
disorders, cancers, and infections.
 Personalized Medicine: Treatments can be tailored to individual genetic
profiles and specific disease mechanisms.
11)Intra Nasal Route Delivery System :- Preparation
The intra nasal route delivery system is a method of
administering medications through the nose. This route is used because
it allows for quick absorption of the drug into the bloodstream,
bypassing the digestive system and providing rapid effects. Here’s how it
is prepared in simple terms:
 Steps to Prepare an Intra Nasal Delivery System
1. Choosing the Medication:
o Select a suitable drug: The medication should be effective and
safe for nasal delivery.
o Form: The drug can be in the form of a liquid, gel, or powder.
2. Formulation Development:
o Solution Preparation: If the drug is a liquid, dissolve it in a suitable
solvent to make a nasal spray solution.
o Stabilizers and Preservatives: Add ingredients to keep the
solution stable and free from contamination.
o pH Adjustment: Adjust the pH to match the nasal environment for
comfort and better absorption.
o Viscosity Agents: Add agents to adjust the thickness of the
solution to ensure it stays in the nasal cavity long enough for
absorption.
Aditya S. Kakad
20
3. Sterilization:
o Sterilize the formulation: Use methods like filtration, heat, or
radiation to ensure the solution is free from microbes.
4. Packaging:
o Nasal Spray Bottles: Fill the solution into nasal spray bottles,
which have a special nozzle for delivering the drug into the nasal
cavity.
o Powder Inhalers: For powder formulations, use devices designed
to deliver the drug as a fine mist.
5. Quality Control:
o Testing: Perform tests to ensure the formulation is effective, safe,
and stable. This includes checking the drug concentration, pH,
sterility, and particle size (for powders).
6. Labeling and Instructions:
o Labeling: Properly label the packaging with the drug’s name,
dosage, expiration date, and instructions for use.
o Instructions: Provide clear instructions on how to use the nasal
delivery system correctly.
 Key Points for Intra Nasal Delivery System
 Quick Absorption: The nasal route allows for rapid absorption of the
drug into the bloodstream, providing quick relief.
 Non-Invasive: It is a non-invasive method, making it easier and more
comfortable for patients to use.
 Bypasses Digestive System: The drug does not pass through the
digestive system, reducing the chance of degradation by stomach acids
or enzymes.
12)Aquasomes : Preparation & Application
Aquasomes are a type of nanoparticle used to deliver drugs,
proteins, and other therapeutic agents in a stable and efficient manner.
They are known for preserving the biological activity of their payloads
due to their water-like properties. Here’s a simple explanation of how
they are prepared and their applications.
 Preparation of Aquasomes
1. Core Formation:
o Material: Typically made from ceramic materials like calcium
phosphate or biodegradable polymers.
o Shape: The core particles are usually spherical in shape and very
small (nanometer scale).
Aditya S. Kakad
21
2. Coating the Core:
o Layer of Carbohydrates: The core is coated with a layer of
carbohydrates (sugars) like trehalose or cellobiose. This layer
helps to protect and stabilize the core and the eventual payload.
o Method: The coating is usually done by adsorption, where the
carbohydrate molecules stick to the core particles.
3. Loading the Drug:
o Immobilization: The drug or therapeutic protein is immobilized
onto the carbohydrate-coated core.
o Non-Covalent Bonding: The loading involves non-covalent
interactions such as hydrogen bonding, van der Waals forces, or
electrostatic interactions, which preserve the biological activity of
the drug.
4. Final Formulation:
o Stabilization: Additional steps may include lyophilization (freeze-
drying) to further stabilize the aquasomes and prepare them for
storage or administration.
 Applications of Aquasomes
1. Drug Delivery:
o Purpose: To deliver drugs in a stable and controlled manner.
o Examples: Used for delivering insulin, antibiotics, and anti-cancer
drugs, ensuring they reach their target site without degradation.
2. Protein and Peptide Delivery:
o Purpose: To deliver fragile biological molecules like proteins and
peptides without losing their activity.
o Examples: Enzymes, hormones (like insulin), and vaccines can be
delivered effectively using aquasomes.
3. Gene Delivery:
o Purpose: To deliver genetic material such as DNA or RNA for gene
therapy.
o Examples: Used in experimental treatments for genetic disorders
and cancers by delivering therapeutic genes directly into cells.
4. Vaccines:
o Purpose: To enhance the stability and delivery of vaccine antigens.
o Examples: Aquasomes can be used to deliver viral proteins or
inactivated viruses, enhancing the immune response and ensuring
the vaccine remains effective.
 Benefits of Aquasomes
 Preservation of Activity: Aquasomes protect the biological activity of the
loaded drug or protein, ensuring it remains effective.
Aditya S. Kakad
22
 Biocompatibility: Made from materials that are generally safe and
compatible with the body.
 Controlled Release: Provides a controlled and sustained release of the
therapeutic agent, improving its efficacy.
 Stability: Enhances the stability of drugs, proteins, and genes, especially
those that are sensitive to environmental conditions.
13)Short Note : Tumor targeting
Tumor targeting is a medical approach used to deliver treatments
directly to cancer cells while minimizing damage to healthy cells. This
method enhances the effectiveness of cancer treatments and reduces
side effects.
 How Tumor Targeting Works
1. Identifying Target Molecules:
o Tumor Markers: Cancer cells often have specific proteins or
markers on their surface that are different from normal cells.
o Target Selection: Researchers identify these unique markers to
target the cancer cells specifically.
2. Delivery Methods:
o Antibodies: Special proteins called antibodies can be designed to
recognize and bind to the tumor markers.
o Nanoparticles: Tiny particles (like liposomes or other
nanocarriers) can be engineered to carry drugs and deliver them
directly to the tumor.
o Ligands: Molecules that bind to specific receptors on cancer cells,
guiding the drug to its target.
3. Mechanism of Action:
o Direct Binding: Antibodies or ligands attach to the tumor markers,
guiding the drug to the cancer cells.
o Release of Drugs: Once attached, the drug is released directly into
the cancer cell, maximizing its effect.
 Benefits of Tumor Targeting
1. Increased Effectiveness:
o Concentration: Higher concentrations of the drug can reach the
tumor, enhancing its ability to kill cancer cells.
Aditya S. Kakad
23
o Specificity: Targeting specific markers ensures that the treatment
is more effective against cancer cells.
2. Reduced Side Effects:
o Minimized Damage: By focusing on cancer cells, less of the drug
affects healthy cells, reducing harmful side effects.
o Precision Medicine: Treatments can be tailored to the specific
type of cancer and its unique markers.
3. Improved Drug Delivery:
o Enhanced Uptake: Targeted delivery systems can improve the
uptake of drugs by cancer cells, making treatments more efficient.
o Controlled Release: Nanoparticles can provide controlled and
sustained release of the drug over time.
 Applications of Tumor Targeting
1. Chemotherapy:
o Example: Liposomal doxorubicin is a nanoparticle-based
chemotherapy that targets cancer cells while sparing healthy cells.
o Benefit: Reduces common side effects like hair loss and nausea.
2. Immunotherapy:
o Example: Monoclonal antibodies (like trastuzumab) target specific
proteins on cancer cells.
o Benefit: Enhances the immune system's ability to recognize and
attack cancer cells.
3. Radiotherapy:
o Example: Radiolabeled antibodies deliver radiation directly to
cancer cells.
o Benefit: Directly irradiates cancer cells, sparing healthy tissue.
4. Gene Therapy:
o Example: Targeted delivery of therapeutic genes to cancer cells to
repair or replace faulty genes.
o Benefit: Provides a more precise approach to correcting genetic
abnormalities in cancer cells.
14)Short Note : Liposomes
Liposomes are tiny, spherical vesicles made from lipids (fats) that
are used to deliver drugs and other substances in the body. They are like
small bubbles with a water-loving (hydrophilic) center and a fat-loving
(lipophilic) outer layer.
Aditya S. Kakad
24
 How Liposomes Work
1. Structure:
o Outer Layer: Made of phospholipids, similar to the materials in
cell membranes.
o Inner Core: Can hold water-soluble drugs, while the outer layer
can carry fat-soluble drugs.
2. Drug Encapsulation:
o Water-Soluble Drugs: Encapsulated in the watery core of the
liposome.
o Fat-Soluble Drugs: Incorporated into the lipid layer of the
liposome.
3. Delivery to Cells:
o Fusion with Cell Membranes: Liposomes can merge with cell
membranes, releasing their contents directly into the cell.
o Endocytosis: Cells can engulf liposomes, taking them in and
releasing the drug inside the cell.
 Benefits of Liposomes
1. Targeted Delivery:
o Site-Specific: Liposomes can be designed to target specific tissues
or organs, reducing side effects and increasing the drug's
effectiveness.
o Prolonged Release: They can release drugs slowly over time,
providing a sustained therapeutic effect.
2. Enhanced Stability:
o Protection: Liposomes protect drugs from degradation in the
body, ensuring they reach their target intact.
o Improved Solubility: They can improve the solubility of drugs that
are poorly soluble in water.
3. Reduced Toxicity:
o Minimized Side Effects: By delivering drugs directly to the target
site, liposomes can reduce the exposure of healthy tissues to the
drug, minimizing side effects.
 Applications of Liposomes
1. Cancer Treatment:
o Example: Doxorubicin, a chemotherapy drug, is delivered using
liposomes (Doxil) to target cancer cells more effectively and
reduce toxicity to the heart.
2. Infectious Diseases:
Aditya S. Kakad
25
o Example: Liposomal amphotericin B is used to treat fungal
infections, providing better efficacy and fewer side effects
compared to the conventional form.
3. Vaccines:
o Example: Liposomes are used in some vaccine formulations to
deliver antigens and enhance the immune response.
4. Gene Therapy:
o Example: Liposomes can carry genetic material (like DNA or RNA)
to cells, offering a method for correcting genetic disorders.
15)Short Note : Micro-capsule
Micro-capsules are tiny, spherical containers that can hold and
release substances like drugs, fragrances, or other active ingredients in a
controlled way. They are much smaller than a grain of sand and are used
in various industries, including pharmaceuticals, food, and cosmetics.
 How Micro-Capsules Work
1. Structure:
o Core: The core of a micro-capsule contains the active ingredient,
such as a drug or fragrance.
o Shell: The core is surrounded by a protective outer shell made
from materials like gelatin, polymers, or other biocompatible
substances.
2. Encapsulation:
o Process: The active ingredient is encapsulated within the shell
using various techniques like spray drying, coacervation, or
interfacial polymerization.
o Purpose: This encapsulation protects the active ingredient from
environmental factors and controls its release.
3. Release Mechanism:
o Controlled Release: The shell can be designed to break down
under specific conditions, such as changes in pH, temperature, or
mechanical pressure.
o Targeted Delivery: The micro-capsule releases its contents exactly
where and when it is needed.
 Benefits of Micro-Capsules
1. Protection:
Aditya S. Kakad
26
o Stability: The shell protects the active ingredient from
degradation due to light, heat, moisture, or other environmental
factors.
o Preservation: This extends the shelf life of the product and
maintains its effectiveness.
2. Controlled Release:
o Timed Release: The active ingredient can be released slowly over
time or under specific conditions, ensuring a sustained effect.
o Targeted Action: Ensures that the active ingredient is delivered
exactly where it is needed, improving efficiency and reducing
waste.
3. Versatility:
o Wide Applications: Used in various fields, including medicine,
food, cosmetics, and agriculture.
o Customization: The size, composition, and release mechanism of
micro-capsules can be tailored to suit specific needs.
 Applications of Micro-Capsules
1. Pharmaceuticals:
o Example: Controlled-release medications that provide a steady
dose of the drug over an extended period.
o Benefit: Improves patient compliance and ensures consistent
therapeutic effects.
2. Food Industry:
o Example: Encapsulation of vitamins, flavors, or probiotics to
protect them from degradation and release them at the right
time.
o Benefit: Enhances the nutritional value and sensory properties of
food products.
3. Cosmetics:
o Example: Fragrances encapsulated in micro-capsules that release
scent upon application to the skin.
o Benefit: Provides a long-lasting fragrance and better product
stability.
4. Agriculture:
o Example: Encapsulated pesticides or fertilizers that release their
contents slowly over time.
o Benefit: Reduces the frequency of application and minimizes
environmental impact.
Aditya S. Kakad
27
16)Short Note : Micro-Sphere
Micro-spheres are tiny, spherical particles used to deliver drugs
and other substances in a controlled manner. They are similar to micro-
capsules but typically have a solid matrix structure instead of a core and
shell design. These particles can be made from various materials and are
used in many fields, especially in medicine.
 How Micro-Spheres Work
1. Structure:
o Solid Matrix: Micro-spheres are composed of a solid matrix that
can hold and release the active ingredient.
o Size: They are very small, usually ranging from 1 to 1000
micrometers in diameter.
2. Encapsulation:
o Incorporation of Active Ingredients: The active ingredient is
evenly distributed within the solid matrix during the formation of
the micro-sphere.
o Materials: They can be made from biocompatible materials like
polymers, proteins, or lipids.
3. Release Mechanism:
o Controlled Release: The active ingredient is released slowly as the
micro-sphere degrades or through diffusion from the matrix.
o Stimuli-Responsive: Some micro-spheres are designed to release
their contents in response to specific triggers like changes in pH,
temperature, or enzyme activity.
 Benefits of Micro-Spheres
1. Controlled Release:
o Sustained Effect: Micro-spheres can provide a sustained release of
the active ingredient over a prolonged period.
o Reduced Dosing Frequency: This means fewer doses are needed,
improving patient compliance and convenience.
2. Targeted Delivery:
Aditya S. Kakad
28
o Specificity: Micro-spheres can be engineered to deliver their
contents to specific tissues or organs, enhancing treatment
effectiveness.
o Minimized Side Effects: By targeting the active ingredient to
where it's needed, micro-spheres can reduce side effects on
healthy tissues.
3. Protection:
o Stability: Micro-spheres protect the active ingredient from
degradation by environmental factors like light, heat, or enzymes.
o Improved Shelf Life: This enhances the stability and shelf life of
the product.
 Applications of Micro-Spheres
1. Pharmaceuticals:
o Example: Extended-release formulations for drugs like pain
relievers, hormones, or antibiotics.
o Benefit: Provides consistent therapeutic levels of the drug over
time.
2. Vaccines:
o Example: Micro-spheres can be used to deliver antigens in a
controlled manner, improving immune response and vaccine
efficacy.
o Benefit: Enhances the effectiveness and stability of vaccines.
3. Cosmetics:
o Example: Micro-spheres containing moisturizers or active
ingredients in skincare products.
o Benefit: Provides a controlled release of active ingredients,
improving the product's performance.
4. Biomedical Engineering:
o Example: Delivery of growth factors or other bioactive substances
to promote tissue regeneration.
o Benefit: Supports controlled and localized treatment, enhancing
the healing process.
17)Short Note : Aquasomes
 It was first developed by NIR KOSSOVSKY
 Aquasomes are spherical in shape with 60-300 nm (less than
1000nm)
Aditya S. Kakad
29
 particle size
 These are nano particulate Carrier Systems and are three layered self
assembled structures.
 There structures are self assembled by non Covalent and ionic bonds.
 The delivery system has been successfully utilized for the delivery of
insulin, hemoglobin and enzymes like Serratiopeptidase etc.
 Aquasomes are nano particulate Carries system but instead of being
Simple nanoparticles these are three layered Self assembled
structures, Comprised of a solid phase nanocrystalline Core Coated
with oligomeric.
 Preparation
Three steps of preparation by using the principle of assembly. :-
1. Preparation of Core
i. Co-precipitation
ii. Self precipitation
iii. Sonication
iv. PAMAM
2. Carbohydrate Coating of core
3. Immobilization of drug molecules.
 Application
1. Oxygen Carrier
2. For immuno therapy
3. for oral route
4. for immune potentiation
5. Anti thrombic activity
6. Antigen delivery
7. Delivery of poorly soluble drug
8. Enzyme delivery
9. Insulin delivery
10.Vaccine delivery.
18)Short Note : Phytosomes
Phytosomes are advanced forms of herbal extracts that enhance
the absorption and bioavailability of plant-based compounds in the
body. They are created by combining natural plant extracts with
phospholipids, which are fat-like substances found in cell membranes.
 How Phytosomes Work
1. Structure:
o Phytoconstituents: The active compounds extracted from plants
(e.g., flavonoids, terpenoids).
Aditya S. Kakad
30
o Phospholipids: Natural fats that help integrate the plant
compounds into cell membranes, improving their absorption.
2. Formation:
o Complexation: The plant extracts are bonded to phospholipids,
creating a phytosome complex.
o Encapsulation: This process encapsulates the active ingredients
within a lipid-compatible form, making them easier for the body
to absorb.
 Benefits of Phytosomes
1. Improved Absorption:
o Enhanced Bioavailability: Phytosomes improve the absorption of
plant compounds, making them more effective at lower doses.
o Better Uptake: The phospholipid complex helps the active
ingredients pass through cell membranes more efficiently.
2. Increased Stability:
o Protection: Phytosomes protect the active plant compounds from
degradation by digestive enzymes and stomach acid.
o Longer Shelf Life: The encapsulation process also helps to extend
the shelf life of the herbal extract.
3. Targeted Delivery:
o Efficient Delivery: Phytosomes ensure that more of the active
ingredient reaches the target tissues in the body.
o Reduced Dosage: Because of the increased bioavailability, lower
doses can achieve the desired therapeutic effect.
 Applications of Phytosomes
1. Nutraceuticals:
o Example: Curcumin phytosomes used for anti-inflammatory and
antioxidant benefits.
o Benefit: Enhanced absorption of curcumin, improving its
effectiveness in reducing inflammation and oxidative stress.
2. Herbal Medicine:
o Example: Milk thistle phytosomes used for liver protection.
o Benefit: Better delivery of silymarin, the active ingredient in milk
thistle, supporting liver health more effectively.
3. Cosmetics:
o Example: Green tea phytosomes used in skincare products.
o Benefit: Improved absorption of the antioxidants in green tea,
enhancing skin protection and anti-aging effects.
Aditya S. Kakad
31
19)Short Note : Aptamers
Aptamers are short, single-stranded DNA or RNA molecules that
can bind to specific targets with high precision, much like antibodies.
They are designed to attach to proteins, small molecules, or even cells,
making them useful in various medical and scientific applications.
 How Aptamers Work
1. Structure:
o Nucleic Acids: Aptamers are made of nucleic acids (DNA or RNA).
o Specific Shapes: They fold into unique three-dimensional shapes
that allow them to bind tightly and specifically to their target
molecules.
2. Selection Process:
o SELEX Method: Aptamers are selected from a large pool of
random sequences using a process called SELEX (Systematic
Evolution of Ligands by EXponential enrichment). This involves
multiple rounds of binding, separation, and amplification to find
the best-fitting sequences.
o High Affinity: The selected aptamers have high affinity and
specificity for their target, meaning they bind very tightly and
selectively.
 Benefits of Aptamers
1. High Specificity:
o Precision Targeting: Aptamers can distinguish between very
similar molecules, allowing for precise targeting of specific
proteins or cells.
2. Versatility:
o Multiple Targets: They can be designed to bind to a wide range of
targets, from small molecules to large proteins and cells.
o Functional Flexibility: Aptamers can be easily modified to
enhance their stability, binding strength, or other properties.
3. Low Immunogenicity:
o Reduced Immune Response: Unlike antibodies, aptamers are less
likely to trigger an immune response in the body, making them
safer for therapeutic use.
4. Easy Production:
o Synthetic Manufacture: Aptamers can be synthesized chemically,
which is generally easier and more cost-effective than producing
antibodies.
Aditya S. Kakad
32
 Applications of Aptamers
1. Medical Diagnostics:
o Example: Aptamers can be used in diagnostic tests to detect the
presence of specific proteins or pathogens in a sample.
o Benefit: Provides highly accurate and sensitive detection.
2. Therapeutics:
o Example: Aptamers can be used to block the activity of disease-
related proteins or deliver drugs to specific cells.
o Benefit: Targets treatments precisely to diseased cells, reducing
side effects.
3. Biosensors:
o Example: Aptamer-based sensors can detect contaminants or
toxins in food, water, or environmental samples.
o Benefit: Offers quick and reliable detection of harmful substances.
4. Research Tools:
o Example: Aptamers can be used to study protein interactions and
functions in the laboratory.
o Benefit: Provides a versatile tool for understanding biological
processes.
20)Short Note : Bio distribution & Pharmacokinetics.
 Biodistribution & Pharmacokinetics
Biodistribution and pharmacokinetics are terms used to describe how drugs
move through the body and where they go.
 Biodistribution
Biodistribution refers to the way a drug or other substance spreads
throughout the body after it is administered. It looks at where the drug goes
and how much of it reaches different tissues and organs.
 Key Points About Biodistribution
1. Administration:
o The drug can be given in various ways, such as orally,
intravenously, or through injection.
2. Distribution:
o Once in the body, the drug travels through the bloodstream to
reach different tissues and organs.
Aditya S. Kakad
33
o Some drugs may accumulate in certain tissues more than others,
depending on their properties and the blood flow to those tissues.
3. Target Tissues:
o Understanding biodistribution helps researchers know if the drug
reaches its intended target in the right amount.
o It also helps identify any potential areas where the drug might
cause unwanted side effects.
 Pharmacokinetics
Pharmacokinetics (PK) is the study of how a drug is absorbed, distributed,
metabolized, and excreted from the body. It’s often summarized by the
acronym ADME.
 Key Points About Pharmacokinetics
1. Absorption:
o How the drug enters the bloodstream. This can vary based on the
method of administration (e.g., oral, injection).
o Example: A pill dissolves in the stomach and intestines, and its
active ingredients are absorbed into the bloodstream.
2. Distribution:
o How the drug spreads throughout the body, similar to
biodistribution.
o Example: Once in the bloodstream, the drug travels to various
organs and tissues.
3. Metabolism:
o How the drug is broken down by the body, usually in the liver.
o Example: Enzymes in the liver convert the drug into metabolites,
which might be active or inactive.
4. Excretion:
o How the drug and its metabolites are eliminated from the body,
usually through urine or feces.
o Example: The kidneys filter out the metabolites, which are then
excreted in the urine.
 Why Biodistribution and Pharmacokinetics Matter
1. Effectiveness:
o Ensuring that the drug reaches the right place in the right amount
to have the desired effect.
2. Safety:
o Understanding where the drug goes helps identify and minimize
potential side effects.
Aditya S. Kakad
34
3. Dosing:
o Helps determine the correct dose and frequency to maintain
effective drug levels in the body without causing harm.
4. Drug Development:
o Critical in developing new drugs and improving existing ones by
providing insights into how the drug behaves in the body.
 Summary
Biodistribution is about where a drug goes in the body after administration,
while pharmacokinetics studies how the drug is absorbed, distributed,
metabolized, and excreted. Both are essential for ensuring that drugs are
effective and safe, helping researchers determine the best dosages and
administration methods.

Molecular Pharmaceutics M.Pharmacy Sem-2

  • 1.
    Aditya S. Kakad 1 MolecularPharmaceutics ( NTDS ) 1) Targeted Drug Delivery System : Concepts & Biological process involved in drug Targeting.  Concepts TDDS is a special form of DDS where medicament is selectively targeted or delivered only to its site of action or absorption and not to the non-target organs or tissues or cells. It is a method Patient in a g delivering medication to a manner that increases the concentration of the medication in parts of the body relative to others. Need of TDDS a) Pharmacokinetic Reason  Poor absorption  host half-life  Large volume of distribution. b) Pharmaco dynamic Reason  Low specificity  low therapeutic index c) Pharmaceutical Reasons  Drug instability .  Low Solubility Concept of Targeting The concept of drug targeting has been originated from the perception of paul ehrlich, who proposed dong delivery to be as a "magic bullet" Gregoriadis, 1981 described drug targeting using novel DDS as "old drugs in new clothes"  Biological process involved in drug Targeting. Events and Biological Process involved in Dreng targeting. a) Cellular uptake and processing b) Transport across the epithelial barrier. c) extravasations d) Lymphatic uptake...
  • 2.
    Aditya S. Kakad 2 a)Cellular uptake and processing Low molar mass drugs can enter into or pass through Various Cells by simple diffusion process Targeted DD usually have macro molecular assemblies hence cannot enter by such simple process, hence taken up by A process Called endocytosis.  Steps involved endocytosis :- i. Intermaligation of the plasma membrane ii. Concomitant with engulment of extra Cellular material.  Types of endocytosis :- i. Phagocytosis Compared with phagocytosis pinocytosis is a universal Phenomenon in all the cells, Pinocytosis does not require any external stimuli. ii. Pinocytosis Types :-  fluid phase pinocytosis  Absorptive pinocytosis. Compared with phagocytosis fluid phase pinocytosis Capture molecules slowly being directly proportional to the concentration and Size dependant. b) Transport across the epithelial barrier.  The oral, Buccal, nasal, vaginal and rectal Cavities. are internally lined with one or more layers of epithelial cells.  Depending on the position and function in the body epithelial cells Can be varied forms.  Epithelial  Lamia propria  Basal lamina.  low molar mass drugs Cross the above by passive diffusion Carrier mediated Systems and selective and nou selective endocytosis  The polar materials diffuse through tight junctions of epithelial cells.  Passive transport is usually higher in damaged mucosa where as active transport depends on structural integrity of epithelial cells.  Positively charged particles showed increased uptake than negatively charged Counterparts.  Absorption of drugs from buccal via transcellular and paracellular later being dominant
  • 3.
    Aditya S. Kakad 3 c)Extravasations Many disease are Known that Hesult from dysfunction Cells outside the Cardiovascular System, thus for a for a drug to exert its therapeutic effect, it must come out from the Central Circulation and interact with extra cellular or intracellular targets. The process of trans cellular exchange is called as extravasation, which is governed by blood. Capillary Walls. Permeability of Capillaries are Controlled by : • Structure of the Capillary wall • Pathological Condition • Rate of blood and lymph supply • Physicochemical factors of drug. Depending on the morphology and Continuity of the endothelial layer and the basement membrane blood Capillaries are divided into : • Continous • Fenestraded • Sinusoidal d) Lymphatic uptake...  extravasion dong molecules Can be either reabsorbed into the blood stream directly or enter into the Lymphatic system and return with the Lymph to the blood Circulation  Drugs administered by subcutaneous intracellular transdermal peritoneal routes Can reach the Systemic Circulation by lymphatic system 2) Brain Specific delivery. Drug delivery to the brain is the process of passing therapeutically active molecules across the Blood- Brain-Barrier. This is a complex process that must take into account the complex anatomy of the brain as well as the restrictions imposed by the special functions of the BBB Various routes of administration as well as conjugations of drugs, e.g. with liposome and nanoparticles are Considered.  Novel Approaches A. Invasive approaches or Neurosurgical approaches: i. Intra - cerebral injection/implant ii. Intra - cerebro ventricular (ICV) infusion iii. Disruption of the BBB iv. Convection-enhanced delivery (CED)
  • 4.
    Aditya S. Kakad 4 B.Non - Invasive approaches : i. Pharmacological techniques: 1. Chemical Techniques a) Prodrugs b) Drug Conjugates 2. Colloidal Techniques a) Nanoparticles b) Liposomes ii. Biological / Physiological Techniques: 1. Pseudo nutrients 2. Antibody 3. Chimeric peptides C. Miscellaneous approaches : i. Intranasal delivery ii. Iontophoretic delivery  Blood Brain Barrier BBB is a highly selective permeability barrier that separates the Circulating blood from the brain extracellular fluid (BECF) in the CNS. BBB is Composed of high density cells, restricting passage of substances from the blood to the brain. Parameters Considered optimum for a compound to transport across the BBB are : • Compound should be un ionized • Approx. log p value must be 2 • Molecular weight must be less than 400 Da • Cumulative no of H-bonds must not go beyond 8-10 • It is estimated only 2% of small molecular wt. drug will cross BBB. .  Strategies :
  • 5.
    Aditya S. Kakad 5 3)Nano Particles : Types, Preparation & evaluation. Nanoparticles are defined as particulate dispersions or Solid particles with a size in the range of 10-1000nm. Drug is confined to a cavity surrounded by a unique polymer layer / membrane called nano Capsules. While nanosphores are matrix system in which the drug is physically and uniformly dispersed. NANO CAPSULES : The nano Capsules are system in which the drug is confined to Surrounded by a Cavity unique polymer membrane NANOSPHERES: The nanosphere are matrix system in which the physically and uniformly drug is dispersed.  Ideal properties of Nanoparticles : • Should be stable in blood • Should be biodegradable • Should be non toxic • Should be non-immunogenic • Should be non- thrombogenic • Should be non-inflammatory  Types of nanoparticles • One dimensional nanoparticles. • Two dimensional nanoparticles • Three dimensional nanoparticles fullerenes • Quantum dots.  Preparation of Nanoparticles. 1. SOLVENT EVAPORATION METHOD  In this method firstly nano emulsion formulation are Prepared  Polymer is dissolved after the 1st step in organic solvent (dichloromethane, chloroform or ethyl acetate)  Drug is dispersed in the solution prepared Then this mixture is emulsified in an aqueous phase which contains surfactant (polysorbates, Poloxamers, Polyvinyl alcohol.)  An oil/water emulsion is prepared by using mechanical stirring, sonification or micro fluidization. 2. DOUBLE EMULSIFICATION METHOD  Emulsification and evaporation method have limitation of poor entrapment of hydrophilic drugs, hence double emulsification technique is used.  Firstly w/o emulsion is prepared by addition of aqueous drug solution to organic polymer with Co with continuous stirring.  This prepared emulsion is mixed with another aqueous phase with vigorous stirring, Hesultanting w/o/w emulsion  Organic solvents are removed by high speed centrifugation.
  • 6.
    Aditya S. Kakad 6 3.EMULSIONS - DIFFUSION METHOD  it is a modified form of salting out method.  In this technique, polymer is dissolved in water miscible solvent (propylene Carbonate, benzyl alcohol, and is saturated with water.  Polymer-water saturated solvent phase is then emulsified in an aqueous solution conteining stabilizer  Afterwards, solvent is removed by evaporation or filtration. 4. NANO PRECIPITATION METHOD  In this method precipitation of polymer and drug obtained from organic solvent and the organic solvent diffused in to the aqueous medium with or without presence of surfactant  Firstly drug was Co-solvent was dissolved in water, and then added into the solution  The another solution of polymer (ethyl cellulose) and propylene glycol with chloroform is prepared and this solution was dispersed to the drug solution  This dispersion was slowly added to 10ml of 7o % aqueous ethanol-solution  After 5 minutes of mixing, the organic solvents. were removed by evaporation at 35° under normal pressure, nanoparticles were separated by using cooling centrifuge (10000 rpm for 20 min)  Supernatants were removed and nanoparticles are washed with water and dried at room temp, in a desicator 5. COACERVATION METHOD  In this method, drug and protein -solution (2 % w/v) is incubated for one hour at room temperature and pH adjusted to 5.5  Ethanol was added to the prepared solution in 2:1 ratio (v/v)  Resultant Coacerved is left to hardened with 25 % glutaraldehyde (1.56 ug/mg) for 2 hours which allow Cross Linking of protein  Organic solvents are removed by notary vacuum evaporation at reduced pressure are and nanoparticles Collected and purified by centrifugation  Pellets of nanoparticles were then suspended in phosphate buffer and lyophilized with mannitol
  • 7.
    Aditya S. Kakad 7 6.SALTING OUT METHOD  Salting out method is Very close to solvent diffusion method.  This technique is based on the separation of water-miscible solvent from aqueous solution by salting out effect.  Generally acetone is used because it is totally miscible with water and easily removed  Polymer and drug are dissolved in a solvent which emulsified into a aqueous solution Containing salting out agent. 7. DIALY SIS  Dialysis is an effective method for preparation of nanoparticles  In this method, firstly polymer (such as poly (benzyl- glutamate)-b-poly (ethylene oxide), poly (lactide) and drug dissolved in an organic Solvent  This solution is added to a dialysis tube and a dialysis is performed.  Evaluation OF Nanoparticles. Nano particles are evaluated by the following Method: 1. Particle Size 2. Surface area 3. Surface charge 4. Density 5. Molecular weight 6. Nano Particle yield 7. Dry entrapment efficiency 8. Invitro release. 4) Monoclonal Antibodies : Types, Preparation, Evaluation & Application  Monoclonal antibodies Can be defined as a type of antibody derived from hybridoma Cells.  Monoclonal antibodies are "antibodies that are identical because they are produced by Single B-Cell clone.  Monoclonal antibodies are homogenous immunological regents of defined specificity, So that these can be utilized for diagnosis and screening with ease and Certainty.  Types : 1. Murine. 2. Chimeric. 3. Humanized. 4. Fully Human.
  • 8.
    Aditya S. Kakad 8 Preparation :  Monoclonal antibodies production (mAb) is produced by Cell lines or clones obtained from the immunized animals with the substances to be studied.  Cell lines are Produced by fusing B-Cells from the immunized animal with myeloma Cells.  To produce the desired MAB, the celle must be grown in either of two ways:  By injection into the peritoneal Cavity in mouse (In Vivo)  By in vitro tissue Culture  The Vitro tissue Culture is the method used when the Cells are placed in culture outside the mouse the mouse's body in flask.  Evaluation : 1. Characterisation of monoclonal antibodies  Physicochemical characterisation  Immunological properties  Biological activity  Purity, impurity and contaminants  Quantity 2. Specifications  Identity  Purity and impurities  Potency  Quantity  General tests  Application 1. Measuring protein and drug level in serum 2. Identifying infectious agents. 3. Identifying and quantifying hormones 4. Biochemical analysis - Pregnancy , cancer 5. Identifying tumors agent & also antibodies 6. MAbs as targeting agents. 7. Diagnostic imaging 8. Protein Purification 5) Niosomes : Types, Preparation, Evaluation & Application  Niosomes are novel drug delivery system in which the medication is encapsulated in a Vesicle.  The Vesicle is composed of a bilayer non-ionic surface active agents and hence the name niosomes.  Niosomes are very small and microscopic in size. Their size lies in the nanometric scale. Although Structurally similar to liposomes, they offer several advantages over them.
  • 9.
    Aditya S. Kakad 9 Types : 1. SUV (small unilameller vesicles) 2. LUV (large unilameller vesicles) 3. MLV (multi lammeler vesicles)  Method of preparation: Ether injection Method  Surfactant + Cholesterol is dissolved in directly ether  Then injected in warm water maintained at 60 o C through a 14 gauze needle  Ether is vaporized to form single layered niosomes. Hand Shaking Method (thin film hydration technique)  Surfactant + Cholesterol + Solvent  Remove organic Solvent at room temperature  Thin layer formed on the walls of flask  film Can be rehydrated to form multilameller niosomes. Sonication Method:  Way in buffes + Surfactant / cholesterol in 10ml of aqueous phase  Above mixture is Sonicated for 3 minutes at 60 oC using titanium probe yielding niosomes. Multiple membrane extrusion method.  Mixture of surfactant, Cholesterol in chloroform is made into thin film by evaporation.  The film is hydrated with aqueous drug solution and the resultant suspension extruded through polycarbonate membrane. It is a good method for Controlling niosomes size. The bubble method  It is novel technique for the one step preparation of liposomes and niosomes without the use of organic Solvents.  The bubbling unit Consists of round-bottomed flask with three neck positioned in water bath to Control the temperature.  Water Cooled reflux and thermometer is positioned in the first and second neck and nitrogen Supply through the third neck.  Cholesterol and Surfactant are dispersed together in the buffer (pH 7.4) at 70oC, the dispersion mixed for 15 Seconds with high shear homogenizer & immediately afterward bubbled at 70oC using nitrogen gas
  • 10.
    Aditya S. Kakad 10 Evaluation : A. Size, Shape and Morphology Freeze Fracture Electron Microscopy:- Visualize the vesicular structure of surfactant based vesicles. Photon Correlation Spectroscopy :- Determine mean diameter of the vesicles. Electron Microscopy :- Morphological studies of vesicles. B. Entrapment efficiency After preparing niosomal dispersion, unentrapped drug is separated by dialysis and the drug remained entrapped in niosomes is determined by complete vesicle disruption using 50% n-propanol or 0.1% Triton X-100 and analysing the resultant solution by appropriate assay method for the drug. C. Vesicle Surface Charge Determined by measurement of electrophoretic mobility and expressed in expressed in terms of zeta potential D.In vitro studies  Applications 1. Drug Targeting 2. In Diagnosis 3. Anti Neoplastic Treatment 4. Delivery of peptide drug 5. Niosomes are carriers of Hemoglobuline 6) Aerosols: Propellents (Nomenclature rule with Eg.); Containers types, Preparation & Evaluation  Propellants Propellants are the driving force in aerosol systems, responsible for expelling the product from its container. They are classified into three main types: 1. Liquefied Gas Propellants  These are gases that become liquid under pressure. Common examples include  Hydrocarbons (eg, propane, butane, isobutane)  Chlorofluorocarbons (CFCs) (eg, trichlorofluoromethane, dichlorodifluoromethane, dichlorotetrafluoroethane)  Hytirofluorocarbons (HFCs) (eg, 1,1,1,2-tetrafluoroethane, 1.1- difluoroethanel
  • 11.
    Aditya S. Kakad 11 2.Compressed Gas Propellants  These are gases compressed into their containers and include  Carbon dioxide (CO)  Nitrogen (N)  Nitrous oxide (NO) 3. Hybrid Propellants  Combination of liquefied gas and compressed gas propellants to achieve specific properties  Containers Aerosol containers are designed to withstand the pressure of the propellant and ensure product stability. Types include: 1. Tin-Plated Steel Cans  Used for a variety of aerosol products  Offer durability and are corrosion resistant when lined. 2. Aluminum Cans  Lightweight corrosion resistant, and used for pharmaceuticals and food products. 3. Glass Containers:  Used less frequently due to fragility, but suitable for certain pharmaceuticals and cosmetic aerosols  Typically used with protective outer coverings or coatings  Plastic Containers  Made from materials like polyethylene or polypropylene  Limited use due to permeation and compatibility issues with certain propellants.  Preparation of Aerosols 1. Formulation:  Active ingredient is dissolved or suspended in a suitable solvent.  Propellant is selected based on product requirements.  An appropriate container and valve system are chosen. 2. Filling:  Cold Filling: Product concentrate and propellant are cooled to -30 to -60°C, then filled into the container.
  • 12.
    Aditya S. Kakad 12 Pressure Filling: Product concentrate is filled at room temperature, followed by adding the propellant under high pressure, 3. Sealing:  The container is sealed with a valve crimped onto the can. 4. Testing:  Leakage and pressure tests to ensure integrity and safety.  Homogeneity and particle size distribution tests to ensure consistent product delivery.  Evaluation of Aerosols 1. Spray Pattern:  Ensures that the aerosol dispenses uniformly. 2. Particle Size Distribution:  Evaluated using methods like laser diffraction to ensure therapeutic efficacy (for inhalation aerosols). 3. Dose Uniformity:  Ensures consistent delivery of the active ingredient per actuation. 4. Leakage Test:  Ensures the container does not leak propellant or product over time, 5. Pressure Test:  Confirms the container can withstand the pressure of the propellant. 6. Valve Delivery Rate:  Measures the amount of product delivered per actuation to ensure proper dosing. 7) Ex-vivo & In-vivo gene therapy.  Ex-Vivo Gene Therapy Ex-vivo gene therapy involves the genetic modification of cells outside the body and then reintroducing them into the patient. This method is particularly useful for cells that can be easily extracted, cultured, and reintroduced, such as blood cells.
  • 13.
    Aditya S. Kakad 13 Steps in Ex-vivo Gene Therapy: 1. Cell Collection:  Cells are harvested from the patient. Common sources include hematopoietic stem cells from bone marrow or peripheral blood. 2. Genetic Modification:  The collected cells are cultured and genetically modified in vitro. This can be done using various vectors, such as:  Viral Vectors: Retroviruses, lentiviruses, adenoviruses, and adeno-associated viruses (AAV) are commonly used to deliver the therapeutic gene.  Non-viral Methods: Techniques like electroporation, liposome- mediated transfer, and CRISPR/Cas9 can be used. 3. Selection and Expansion:  Modified cells are selected and expanded in culture to increase their number. 4. Reintroduction:  The genetically modified cells are reintroduced into the patient, typically through infusion. 5. Engraftment and Function:  The modified cells engraft into the patient's tissues and begin to produce the therapeutic effect.  Applications of Ex-vivo Gene Therapy:  Hematological Disorders: Treatment of severe combined immunodeficiency (SCID), beta- thalassemia, and sickle cell disease.  Metabolic Disorders: Enzyme replacement in conditions like adenosine deaminase deficiency.  Cancer: CAR-T cell therapy for certain types of leukemia and lymphoma.  Advantages:  Controlled environment for genetic modification.  Ability to select and expand modified cells.  Reduced risk of immune response against the vector.
  • 14.
    Aditya S. Kakad 14 Challenges:  Complex and time-consuming process.  Requires specialized facilities and expertise.  Limited to cells that can be extracted, cultured, and reintroduced.  In-Vivo Gene Therapy In-vivo gene therapy involves delivering genetic material directly into the patient's body. This approach targets cells within the body and does not require cell extraction and reintroduction.  Steps in In-vivo Gene Therapy: 1. Vector Design:  A suitable vector is designed to carry the therapeutic gene. Common vectors include:  Viral Vectors: Adenoviruses, AAV, lentiviruses, and herpes simplex viruses (HSV).  Non-viral Vectors: Naked DNA, plasmids, nanoparticles, and liposomes. 2. Delivery:  The vector carrying the therapeutic gene is administered to the patient. Delivery routes can vary:  Intravenous (IV): For systemic delivery.  Intramuscular (IM): For targeting muscle tissue.  Intrathecal: For delivery to the central nervous system.  Inhalation: For respiratory diseases.  Local Injection: For specific tissues or organs. 3. Cellular Uptake and Expression:  The vector transduces the target cells, and the therapeutic gene is expressed, leading to the desired therapeutic effect.  Applications of In-vivo Gene Therapy:  Genetic Disorders: Treatment of Duchenne muscular dystrophy, hemophilia, and cystic fibrosis.  Ocular Diseases: Gene therapy for Leber's congenital amaurosis and retinitis pigmentosa.
  • 15.
    Aditya S. Kakad 15 Neurological Disorders: Treatment of spinal muscular atrophy and Parkinson's disease.  Cardiovascular Diseases: Delivery of angiogenic genes for ischemic heart disease.  Advantages:  Direct delivery to target tissues.  Suitable for a wide range of tissues and organs.  Simpler and faster compared to ex-vivo methods.  Challenges:  Potential for immune responses against the vector.  Difficulty in targeting specific cells or tissues.  Risk of off-target effects and insertional mutagenesis. 8) Potential target disease for gene therapy (Inherited disorder & cancer) Gene therapy is a cutting-edge medical approach that involves modifying a person’s genes to treat or prevent disease. It holds promise for a variety of diseases, particularly inherited disorders and cancer.  Inherited Disorders 1. Cystic Fibrosis (CF): o Problem: Affects the lungs and digestive system due to a defective gene that causes mucus to become thick and sticky. o Gene Therapy Goal: Introduce a healthy version of the CFTR gene to help produce normal mucus and improve lung function. 2. Hemophilia: o Problem: A bleeding disorder caused by a deficiency in specific clotting factors due to faulty genes. o Gene Therapy Goal: Deliver a working copy of the defective gene to help the body produce the missing clotting factors and prevent excessive bleeding. 3. Duchenne Muscular Dystrophy (DMD): o Problem: A severe muscle-wasting disease caused by mutations in the dystrophin gene. o Gene Therapy Goal: Introduce a functional version of the dystrophin gene to help build and repair muscle tissue.
  • 16.
    Aditya S. Kakad 16 4.Sickle Cell Disease: o Problem: Red blood cells become misshapen and break down due to a mutation in the hemoglobin gene. o Gene Therapy Goal: Correct or replace the defective hemoglobin gene to produce healthy red blood cells.  Cancer 1. Leukemia: o Problem: A type of cancer that affects blood and bone marrow. o Gene Therapy Goal: Use modified immune cells (CAR-T cells) to target and destroy cancerous cells in the blood. 2. Glioblastoma: o Problem: An aggressive form of brain cancer. o Gene Therapy Goal: Introduce genes that make cancer cells more sensitive to treatments like chemotherapy or trigger the immune system to attack the tumor. 3. Melanoma: o Problem: A dangerous form of skin cancer that can spread to other parts of the body. o Gene Therapy Goal: Employ gene editing techniques to modify immune cells so they can better recognize and attack melanoma cells. 4. Prostate Cancer: o Problem: A common cancer in men that affects the prostate gland. o Gene Therapy Goal: Introduce genes that target and kill cancer cells or make them more responsive to other treatments. 9) Liposomal gens delivery systems Liposomal gene delivery systems are a way to transport genetic material (like DNA or RNA) into cells using tiny, fat-like particles called liposomes. This method helps deliver genes safely and effectively to treat or prevent diseases.  How Liposomal Gene Delivery Works 1. Creating Liposomes: o Liposomes are small, spherical vesicles made from phospholipids, which are the same types of fats found in cell membranes. o These vesicles can encase genetic material, protecting it from degradation.
  • 17.
    Aditya S. Kakad 17 2.Loading the Genes: o The genetic material, such as a piece of DNA or RNA, is placed inside the liposome. o This can include genes needed to replace faulty ones, or RNA that can silence harmful genes. 3. Delivery to Cells: o Once loaded, the liposomes are introduced into the body. o Because they are made of similar materials to cell membranes, liposomes can merge with cells and release their genetic payload inside. 4. Gene Expression: o After the genetic material is delivered into the cell, it can be used by the cell’s machinery. o This can result in the production of a needed protein, correction of a genetic defect, or silencing of a harmful gene.  Benefits of Liposomal Gene Delivery  Protection: Liposomes protect genetic material from being broken down by enzymes in the body.  Efficiency: They facilitate the entry of genes into cells more efficiently compared to direct injection.  Biocompatibility: Made from natural materials, liposomes are generally non-toxic and less likely to trigger an immune response.  Targeting: Liposomes can be modified to target specific types of cells, improving the precision of the treatment.  Applications 1. Inherited Disorders: o Liposomal systems can be used to deliver functional genes to correct genetic defects, such as in cystic fibrosis or hemophilia. 2. Cancer: o They can transport genes that make cancer cells more susceptible to treatment or enhance the body’s immune response to cancer. 3. Vaccines: o Liposomes can deliver RNA vaccines, such as those used for COVID-19, to instruct cells to produce antigens and trigger an immune response.
  • 18.
    Aditya S. Kakad 18 10)TherapeuticAntisense Molecules (with application) Therapeutic antisense molecules are a type of treatment that can specifically target and block the activity of certain genes. These molecules are designed to bind to the messenger RNA (mRNA) produced by a gene, preventing it from making a protein that could cause disease.  How Antisense Molecules Work 1. Structure: o Antisense molecules are short, single strands of DNA or RNA. o They are complementary to a specific mRNA sequence, meaning they can bind to it perfectly. 2. Binding to mRNA: o When an antisense molecule binds to its target mRNA, it prevents the mRNA from being used to make a protein. o This process is like putting a "block" on the instructions for making the protein. 3. Blocking Protein Production: o By blocking the mRNA, antisense molecules can reduce or eliminate the production of harmful proteins. o This can help treat diseases caused by the overproduction or abnormal production of certain proteins.  Applications of Therapeutic Antisense Molecules 1. Genetic Disorders: o Example: Duchenne Muscular Dystrophy (DMD)  How it helps: Antisense molecules can help restore the production of a functional dystrophin protein, which is missing or defective in DMD patients.  Outcome: Improved muscle function and slowed disease progression. 2. Cancer: o Example: Bcl-2 Protein in Cancer  How it helps: Antisense molecules can target and reduce the production of Bcl-2, a protein that helps cancer cells survive.  Outcome: Increased effectiveness of cancer treatments and reduced tumor growth. 3. Viral Infections: o Example: Cytomegalovirus (CMV) Infections  How it helps: Antisense molecules can inhibit the replication of viral mRNA, reducing the viral load in infected individuals.  Outcome: Better control of the infection and fewer symptoms.
  • 19.
    Aditya S. Kakad 19 4.Cholesterol Management: o Example: High Cholesterol (Familial Hypercholesterolemia)  How it helps: Antisense molecules can target and reduce the production of apolipoprotein B (ApoB), a key component in the formation of bad cholesterol (LDL).  Outcome: Lower levels of LDL cholesterol and reduced risk of heart disease.  Benefits of Therapeutic Antisense Molecules  Precision: They can specifically target and silence disease-causing genes without affecting other genes.  Versatility: Applicable to a wide range of diseases, including genetic disorders, cancers, and infections.  Personalized Medicine: Treatments can be tailored to individual genetic profiles and specific disease mechanisms. 11)Intra Nasal Route Delivery System :- Preparation The intra nasal route delivery system is a method of administering medications through the nose. This route is used because it allows for quick absorption of the drug into the bloodstream, bypassing the digestive system and providing rapid effects. Here’s how it is prepared in simple terms:  Steps to Prepare an Intra Nasal Delivery System 1. Choosing the Medication: o Select a suitable drug: The medication should be effective and safe for nasal delivery. o Form: The drug can be in the form of a liquid, gel, or powder. 2. Formulation Development: o Solution Preparation: If the drug is a liquid, dissolve it in a suitable solvent to make a nasal spray solution. o Stabilizers and Preservatives: Add ingredients to keep the solution stable and free from contamination. o pH Adjustment: Adjust the pH to match the nasal environment for comfort and better absorption. o Viscosity Agents: Add agents to adjust the thickness of the solution to ensure it stays in the nasal cavity long enough for absorption.
  • 20.
    Aditya S. Kakad 20 3.Sterilization: o Sterilize the formulation: Use methods like filtration, heat, or radiation to ensure the solution is free from microbes. 4. Packaging: o Nasal Spray Bottles: Fill the solution into nasal spray bottles, which have a special nozzle for delivering the drug into the nasal cavity. o Powder Inhalers: For powder formulations, use devices designed to deliver the drug as a fine mist. 5. Quality Control: o Testing: Perform tests to ensure the formulation is effective, safe, and stable. This includes checking the drug concentration, pH, sterility, and particle size (for powders). 6. Labeling and Instructions: o Labeling: Properly label the packaging with the drug’s name, dosage, expiration date, and instructions for use. o Instructions: Provide clear instructions on how to use the nasal delivery system correctly.  Key Points for Intra Nasal Delivery System  Quick Absorption: The nasal route allows for rapid absorption of the drug into the bloodstream, providing quick relief.  Non-Invasive: It is a non-invasive method, making it easier and more comfortable for patients to use.  Bypasses Digestive System: The drug does not pass through the digestive system, reducing the chance of degradation by stomach acids or enzymes. 12)Aquasomes : Preparation & Application Aquasomes are a type of nanoparticle used to deliver drugs, proteins, and other therapeutic agents in a stable and efficient manner. They are known for preserving the biological activity of their payloads due to their water-like properties. Here’s a simple explanation of how they are prepared and their applications.  Preparation of Aquasomes 1. Core Formation: o Material: Typically made from ceramic materials like calcium phosphate or biodegradable polymers. o Shape: The core particles are usually spherical in shape and very small (nanometer scale).
  • 21.
    Aditya S. Kakad 21 2.Coating the Core: o Layer of Carbohydrates: The core is coated with a layer of carbohydrates (sugars) like trehalose or cellobiose. This layer helps to protect and stabilize the core and the eventual payload. o Method: The coating is usually done by adsorption, where the carbohydrate molecules stick to the core particles. 3. Loading the Drug: o Immobilization: The drug or therapeutic protein is immobilized onto the carbohydrate-coated core. o Non-Covalent Bonding: The loading involves non-covalent interactions such as hydrogen bonding, van der Waals forces, or electrostatic interactions, which preserve the biological activity of the drug. 4. Final Formulation: o Stabilization: Additional steps may include lyophilization (freeze- drying) to further stabilize the aquasomes and prepare them for storage or administration.  Applications of Aquasomes 1. Drug Delivery: o Purpose: To deliver drugs in a stable and controlled manner. o Examples: Used for delivering insulin, antibiotics, and anti-cancer drugs, ensuring they reach their target site without degradation. 2. Protein and Peptide Delivery: o Purpose: To deliver fragile biological molecules like proteins and peptides without losing their activity. o Examples: Enzymes, hormones (like insulin), and vaccines can be delivered effectively using aquasomes. 3. Gene Delivery: o Purpose: To deliver genetic material such as DNA or RNA for gene therapy. o Examples: Used in experimental treatments for genetic disorders and cancers by delivering therapeutic genes directly into cells. 4. Vaccines: o Purpose: To enhance the stability and delivery of vaccine antigens. o Examples: Aquasomes can be used to deliver viral proteins or inactivated viruses, enhancing the immune response and ensuring the vaccine remains effective.  Benefits of Aquasomes  Preservation of Activity: Aquasomes protect the biological activity of the loaded drug or protein, ensuring it remains effective.
  • 22.
    Aditya S. Kakad 22 Biocompatibility: Made from materials that are generally safe and compatible with the body.  Controlled Release: Provides a controlled and sustained release of the therapeutic agent, improving its efficacy.  Stability: Enhances the stability of drugs, proteins, and genes, especially those that are sensitive to environmental conditions. 13)Short Note : Tumor targeting Tumor targeting is a medical approach used to deliver treatments directly to cancer cells while minimizing damage to healthy cells. This method enhances the effectiveness of cancer treatments and reduces side effects.  How Tumor Targeting Works 1. Identifying Target Molecules: o Tumor Markers: Cancer cells often have specific proteins or markers on their surface that are different from normal cells. o Target Selection: Researchers identify these unique markers to target the cancer cells specifically. 2. Delivery Methods: o Antibodies: Special proteins called antibodies can be designed to recognize and bind to the tumor markers. o Nanoparticles: Tiny particles (like liposomes or other nanocarriers) can be engineered to carry drugs and deliver them directly to the tumor. o Ligands: Molecules that bind to specific receptors on cancer cells, guiding the drug to its target. 3. Mechanism of Action: o Direct Binding: Antibodies or ligands attach to the tumor markers, guiding the drug to the cancer cells. o Release of Drugs: Once attached, the drug is released directly into the cancer cell, maximizing its effect.  Benefits of Tumor Targeting 1. Increased Effectiveness: o Concentration: Higher concentrations of the drug can reach the tumor, enhancing its ability to kill cancer cells.
  • 23.
    Aditya S. Kakad 23 oSpecificity: Targeting specific markers ensures that the treatment is more effective against cancer cells. 2. Reduced Side Effects: o Minimized Damage: By focusing on cancer cells, less of the drug affects healthy cells, reducing harmful side effects. o Precision Medicine: Treatments can be tailored to the specific type of cancer and its unique markers. 3. Improved Drug Delivery: o Enhanced Uptake: Targeted delivery systems can improve the uptake of drugs by cancer cells, making treatments more efficient. o Controlled Release: Nanoparticles can provide controlled and sustained release of the drug over time.  Applications of Tumor Targeting 1. Chemotherapy: o Example: Liposomal doxorubicin is a nanoparticle-based chemotherapy that targets cancer cells while sparing healthy cells. o Benefit: Reduces common side effects like hair loss and nausea. 2. Immunotherapy: o Example: Monoclonal antibodies (like trastuzumab) target specific proteins on cancer cells. o Benefit: Enhances the immune system's ability to recognize and attack cancer cells. 3. Radiotherapy: o Example: Radiolabeled antibodies deliver radiation directly to cancer cells. o Benefit: Directly irradiates cancer cells, sparing healthy tissue. 4. Gene Therapy: o Example: Targeted delivery of therapeutic genes to cancer cells to repair or replace faulty genes. o Benefit: Provides a more precise approach to correcting genetic abnormalities in cancer cells. 14)Short Note : Liposomes Liposomes are tiny, spherical vesicles made from lipids (fats) that are used to deliver drugs and other substances in the body. They are like small bubbles with a water-loving (hydrophilic) center and a fat-loving (lipophilic) outer layer.
  • 24.
    Aditya S. Kakad 24 How Liposomes Work 1. Structure: o Outer Layer: Made of phospholipids, similar to the materials in cell membranes. o Inner Core: Can hold water-soluble drugs, while the outer layer can carry fat-soluble drugs. 2. Drug Encapsulation: o Water-Soluble Drugs: Encapsulated in the watery core of the liposome. o Fat-Soluble Drugs: Incorporated into the lipid layer of the liposome. 3. Delivery to Cells: o Fusion with Cell Membranes: Liposomes can merge with cell membranes, releasing their contents directly into the cell. o Endocytosis: Cells can engulf liposomes, taking them in and releasing the drug inside the cell.  Benefits of Liposomes 1. Targeted Delivery: o Site-Specific: Liposomes can be designed to target specific tissues or organs, reducing side effects and increasing the drug's effectiveness. o Prolonged Release: They can release drugs slowly over time, providing a sustained therapeutic effect. 2. Enhanced Stability: o Protection: Liposomes protect drugs from degradation in the body, ensuring they reach their target intact. o Improved Solubility: They can improve the solubility of drugs that are poorly soluble in water. 3. Reduced Toxicity: o Minimized Side Effects: By delivering drugs directly to the target site, liposomes can reduce the exposure of healthy tissues to the drug, minimizing side effects.  Applications of Liposomes 1. Cancer Treatment: o Example: Doxorubicin, a chemotherapy drug, is delivered using liposomes (Doxil) to target cancer cells more effectively and reduce toxicity to the heart. 2. Infectious Diseases:
  • 25.
    Aditya S. Kakad 25 oExample: Liposomal amphotericin B is used to treat fungal infections, providing better efficacy and fewer side effects compared to the conventional form. 3. Vaccines: o Example: Liposomes are used in some vaccine formulations to deliver antigens and enhance the immune response. 4. Gene Therapy: o Example: Liposomes can carry genetic material (like DNA or RNA) to cells, offering a method for correcting genetic disorders. 15)Short Note : Micro-capsule Micro-capsules are tiny, spherical containers that can hold and release substances like drugs, fragrances, or other active ingredients in a controlled way. They are much smaller than a grain of sand and are used in various industries, including pharmaceuticals, food, and cosmetics.  How Micro-Capsules Work 1. Structure: o Core: The core of a micro-capsule contains the active ingredient, such as a drug or fragrance. o Shell: The core is surrounded by a protective outer shell made from materials like gelatin, polymers, or other biocompatible substances. 2. Encapsulation: o Process: The active ingredient is encapsulated within the shell using various techniques like spray drying, coacervation, or interfacial polymerization. o Purpose: This encapsulation protects the active ingredient from environmental factors and controls its release. 3. Release Mechanism: o Controlled Release: The shell can be designed to break down under specific conditions, such as changes in pH, temperature, or mechanical pressure. o Targeted Delivery: The micro-capsule releases its contents exactly where and when it is needed.  Benefits of Micro-Capsules 1. Protection:
  • 26.
    Aditya S. Kakad 26 oStability: The shell protects the active ingredient from degradation due to light, heat, moisture, or other environmental factors. o Preservation: This extends the shelf life of the product and maintains its effectiveness. 2. Controlled Release: o Timed Release: The active ingredient can be released slowly over time or under specific conditions, ensuring a sustained effect. o Targeted Action: Ensures that the active ingredient is delivered exactly where it is needed, improving efficiency and reducing waste. 3. Versatility: o Wide Applications: Used in various fields, including medicine, food, cosmetics, and agriculture. o Customization: The size, composition, and release mechanism of micro-capsules can be tailored to suit specific needs.  Applications of Micro-Capsules 1. Pharmaceuticals: o Example: Controlled-release medications that provide a steady dose of the drug over an extended period. o Benefit: Improves patient compliance and ensures consistent therapeutic effects. 2. Food Industry: o Example: Encapsulation of vitamins, flavors, or probiotics to protect them from degradation and release them at the right time. o Benefit: Enhances the nutritional value and sensory properties of food products. 3. Cosmetics: o Example: Fragrances encapsulated in micro-capsules that release scent upon application to the skin. o Benefit: Provides a long-lasting fragrance and better product stability. 4. Agriculture: o Example: Encapsulated pesticides or fertilizers that release their contents slowly over time. o Benefit: Reduces the frequency of application and minimizes environmental impact.
  • 27.
    Aditya S. Kakad 27 16)ShortNote : Micro-Sphere Micro-spheres are tiny, spherical particles used to deliver drugs and other substances in a controlled manner. They are similar to micro- capsules but typically have a solid matrix structure instead of a core and shell design. These particles can be made from various materials and are used in many fields, especially in medicine.  How Micro-Spheres Work 1. Structure: o Solid Matrix: Micro-spheres are composed of a solid matrix that can hold and release the active ingredient. o Size: They are very small, usually ranging from 1 to 1000 micrometers in diameter. 2. Encapsulation: o Incorporation of Active Ingredients: The active ingredient is evenly distributed within the solid matrix during the formation of the micro-sphere. o Materials: They can be made from biocompatible materials like polymers, proteins, or lipids. 3. Release Mechanism: o Controlled Release: The active ingredient is released slowly as the micro-sphere degrades or through diffusion from the matrix. o Stimuli-Responsive: Some micro-spheres are designed to release their contents in response to specific triggers like changes in pH, temperature, or enzyme activity.  Benefits of Micro-Spheres 1. Controlled Release: o Sustained Effect: Micro-spheres can provide a sustained release of the active ingredient over a prolonged period. o Reduced Dosing Frequency: This means fewer doses are needed, improving patient compliance and convenience. 2. Targeted Delivery:
  • 28.
    Aditya S. Kakad 28 oSpecificity: Micro-spheres can be engineered to deliver their contents to specific tissues or organs, enhancing treatment effectiveness. o Minimized Side Effects: By targeting the active ingredient to where it's needed, micro-spheres can reduce side effects on healthy tissues. 3. Protection: o Stability: Micro-spheres protect the active ingredient from degradation by environmental factors like light, heat, or enzymes. o Improved Shelf Life: This enhances the stability and shelf life of the product.  Applications of Micro-Spheres 1. Pharmaceuticals: o Example: Extended-release formulations for drugs like pain relievers, hormones, or antibiotics. o Benefit: Provides consistent therapeutic levels of the drug over time. 2. Vaccines: o Example: Micro-spheres can be used to deliver antigens in a controlled manner, improving immune response and vaccine efficacy. o Benefit: Enhances the effectiveness and stability of vaccines. 3. Cosmetics: o Example: Micro-spheres containing moisturizers or active ingredients in skincare products. o Benefit: Provides a controlled release of active ingredients, improving the product's performance. 4. Biomedical Engineering: o Example: Delivery of growth factors or other bioactive substances to promote tissue regeneration. o Benefit: Supports controlled and localized treatment, enhancing the healing process. 17)Short Note : Aquasomes  It was first developed by NIR KOSSOVSKY  Aquasomes are spherical in shape with 60-300 nm (less than 1000nm)
  • 29.
    Aditya S. Kakad 29 particle size  These are nano particulate Carrier Systems and are three layered self assembled structures.  There structures are self assembled by non Covalent and ionic bonds.  The delivery system has been successfully utilized for the delivery of insulin, hemoglobin and enzymes like Serratiopeptidase etc.  Aquasomes are nano particulate Carries system but instead of being Simple nanoparticles these are three layered Self assembled structures, Comprised of a solid phase nanocrystalline Core Coated with oligomeric.  Preparation Three steps of preparation by using the principle of assembly. :- 1. Preparation of Core i. Co-precipitation ii. Self precipitation iii. Sonication iv. PAMAM 2. Carbohydrate Coating of core 3. Immobilization of drug molecules.  Application 1. Oxygen Carrier 2. For immuno therapy 3. for oral route 4. for immune potentiation 5. Anti thrombic activity 6. Antigen delivery 7. Delivery of poorly soluble drug 8. Enzyme delivery 9. Insulin delivery 10.Vaccine delivery. 18)Short Note : Phytosomes Phytosomes are advanced forms of herbal extracts that enhance the absorption and bioavailability of plant-based compounds in the body. They are created by combining natural plant extracts with phospholipids, which are fat-like substances found in cell membranes.  How Phytosomes Work 1. Structure: o Phytoconstituents: The active compounds extracted from plants (e.g., flavonoids, terpenoids).
  • 30.
    Aditya S. Kakad 30 oPhospholipids: Natural fats that help integrate the plant compounds into cell membranes, improving their absorption. 2. Formation: o Complexation: The plant extracts are bonded to phospholipids, creating a phytosome complex. o Encapsulation: This process encapsulates the active ingredients within a lipid-compatible form, making them easier for the body to absorb.  Benefits of Phytosomes 1. Improved Absorption: o Enhanced Bioavailability: Phytosomes improve the absorption of plant compounds, making them more effective at lower doses. o Better Uptake: The phospholipid complex helps the active ingredients pass through cell membranes more efficiently. 2. Increased Stability: o Protection: Phytosomes protect the active plant compounds from degradation by digestive enzymes and stomach acid. o Longer Shelf Life: The encapsulation process also helps to extend the shelf life of the herbal extract. 3. Targeted Delivery: o Efficient Delivery: Phytosomes ensure that more of the active ingredient reaches the target tissues in the body. o Reduced Dosage: Because of the increased bioavailability, lower doses can achieve the desired therapeutic effect.  Applications of Phytosomes 1. Nutraceuticals: o Example: Curcumin phytosomes used for anti-inflammatory and antioxidant benefits. o Benefit: Enhanced absorption of curcumin, improving its effectiveness in reducing inflammation and oxidative stress. 2. Herbal Medicine: o Example: Milk thistle phytosomes used for liver protection. o Benefit: Better delivery of silymarin, the active ingredient in milk thistle, supporting liver health more effectively. 3. Cosmetics: o Example: Green tea phytosomes used in skincare products. o Benefit: Improved absorption of the antioxidants in green tea, enhancing skin protection and anti-aging effects.
  • 31.
    Aditya S. Kakad 31 19)ShortNote : Aptamers Aptamers are short, single-stranded DNA or RNA molecules that can bind to specific targets with high precision, much like antibodies. They are designed to attach to proteins, small molecules, or even cells, making them useful in various medical and scientific applications.  How Aptamers Work 1. Structure: o Nucleic Acids: Aptamers are made of nucleic acids (DNA or RNA). o Specific Shapes: They fold into unique three-dimensional shapes that allow them to bind tightly and specifically to their target molecules. 2. Selection Process: o SELEX Method: Aptamers are selected from a large pool of random sequences using a process called SELEX (Systematic Evolution of Ligands by EXponential enrichment). This involves multiple rounds of binding, separation, and amplification to find the best-fitting sequences. o High Affinity: The selected aptamers have high affinity and specificity for their target, meaning they bind very tightly and selectively.  Benefits of Aptamers 1. High Specificity: o Precision Targeting: Aptamers can distinguish between very similar molecules, allowing for precise targeting of specific proteins or cells. 2. Versatility: o Multiple Targets: They can be designed to bind to a wide range of targets, from small molecules to large proteins and cells. o Functional Flexibility: Aptamers can be easily modified to enhance their stability, binding strength, or other properties. 3. Low Immunogenicity: o Reduced Immune Response: Unlike antibodies, aptamers are less likely to trigger an immune response in the body, making them safer for therapeutic use. 4. Easy Production: o Synthetic Manufacture: Aptamers can be synthesized chemically, which is generally easier and more cost-effective than producing antibodies.
  • 32.
    Aditya S. Kakad 32 Applications of Aptamers 1. Medical Diagnostics: o Example: Aptamers can be used in diagnostic tests to detect the presence of specific proteins or pathogens in a sample. o Benefit: Provides highly accurate and sensitive detection. 2. Therapeutics: o Example: Aptamers can be used to block the activity of disease- related proteins or deliver drugs to specific cells. o Benefit: Targets treatments precisely to diseased cells, reducing side effects. 3. Biosensors: o Example: Aptamer-based sensors can detect contaminants or toxins in food, water, or environmental samples. o Benefit: Offers quick and reliable detection of harmful substances. 4. Research Tools: o Example: Aptamers can be used to study protein interactions and functions in the laboratory. o Benefit: Provides a versatile tool for understanding biological processes. 20)Short Note : Bio distribution & Pharmacokinetics.  Biodistribution & Pharmacokinetics Biodistribution and pharmacokinetics are terms used to describe how drugs move through the body and where they go.  Biodistribution Biodistribution refers to the way a drug or other substance spreads throughout the body after it is administered. It looks at where the drug goes and how much of it reaches different tissues and organs.  Key Points About Biodistribution 1. Administration: o The drug can be given in various ways, such as orally, intravenously, or through injection. 2. Distribution: o Once in the body, the drug travels through the bloodstream to reach different tissues and organs.
  • 33.
    Aditya S. Kakad 33 oSome drugs may accumulate in certain tissues more than others, depending on their properties and the blood flow to those tissues. 3. Target Tissues: o Understanding biodistribution helps researchers know if the drug reaches its intended target in the right amount. o It also helps identify any potential areas where the drug might cause unwanted side effects.  Pharmacokinetics Pharmacokinetics (PK) is the study of how a drug is absorbed, distributed, metabolized, and excreted from the body. It’s often summarized by the acronym ADME.  Key Points About Pharmacokinetics 1. Absorption: o How the drug enters the bloodstream. This can vary based on the method of administration (e.g., oral, injection). o Example: A pill dissolves in the stomach and intestines, and its active ingredients are absorbed into the bloodstream. 2. Distribution: o How the drug spreads throughout the body, similar to biodistribution. o Example: Once in the bloodstream, the drug travels to various organs and tissues. 3. Metabolism: o How the drug is broken down by the body, usually in the liver. o Example: Enzymes in the liver convert the drug into metabolites, which might be active or inactive. 4. Excretion: o How the drug and its metabolites are eliminated from the body, usually through urine or feces. o Example: The kidneys filter out the metabolites, which are then excreted in the urine.  Why Biodistribution and Pharmacokinetics Matter 1. Effectiveness: o Ensuring that the drug reaches the right place in the right amount to have the desired effect. 2. Safety: o Understanding where the drug goes helps identify and minimize potential side effects.
  • 34.
    Aditya S. Kakad 34 3.Dosing: o Helps determine the correct dose and frequency to maintain effective drug levels in the body without causing harm. 4. Drug Development: o Critical in developing new drugs and improving existing ones by providing insights into how the drug behaves in the body.  Summary Biodistribution is about where a drug goes in the body after administration, while pharmacokinetics studies how the drug is absorbed, distributed, metabolized, and excreted. Both are essential for ensuring that drugs are effective and safe, helping researchers determine the best dosages and administration methods.