Nothing Special   »   [go: up one dir, main page]

2022-PHARM TECH II LAB MANUAL 1

Download as pdf or txt
Download as pdf or txt
You are on page 1of 117

2022

PHARMACEUTICAL
TECHNOLOGY II

Laboratory Manual

DEPARTMENT OF
PHARMACEUTICS
SCHOOL OF PHARMACY
UNIVERSITY OF NAMIBIA

Page |0
Index Page

Laboratory Safety 2

The Basics of Pharmaceutical Technology 4

Pharmaceutical Formulations (MCQ Exercise) 7

1. Good Manufacturing and Laboratory Practices 11

2. Pharmaceutical Research and Development 17

3. Manufacturing methods of dosage forms 20

4. Practical Application in Biotech 22

5. Drug Delivery 25

6. Practical Application in Specialty Pharmaceuticals 39

7. Small-scale Manufacture of Tablets by Wet Granulation 46

8. Solid Dosage Forms: 56


Powders and Granules
Capsules
Suppositories
9. Semi-solid Dosage Forms: 62
Gels
Creams
Pastes
10. Liquid Dosage Forms: 69
Solutions
Syrups
Elixirs
Suspensions
Emulsions
11. Quality Control of Dosage Forms 80

Journal Article Format - Formulation, In-process analysis & Discussion 89

Excipients in Drug Delivery and Tablet formulation -22-

References -24-
Appendices:
Article – Emulsions
Article/Experiment – Suspensions
Article – Quality Evaluation

Page |1
LAB SAFETY

In the laboratory, students should behave in accordance with the following general rules that are also displayed on
the walls in the laboratories:

▪ The practical classes begin promptly at the prescribed times. No student is allowed in the laboratory
before or after these times.
▪ Take note of the “prohibited” signs in the laboratory.
▪ Components that had been distributed must be handed in at the end of the practical.
▪ Connecting wire must also be returned where applicable.
▪ All electrical instruments must be switched off at the end of the practical.
▪ Chairs must be returned to their places after use.
▪ Tidy bench after use.
▪ Defective equipment must be entered in designated fault book in the laboratory.
▪ Please do not carry apparatus from one table to the other without permission of the demonstrator.
▪ No electrical circuit is to be switched on before it is checked by a demonstrator.
▪ Switch everything off after you have finished using it.
▪ Please be careful with gas and Bunsen burners. Make sure they are switched off after usage.
▪ Please leave the apparatus in neat order after completion of your experiment. Switch off all electrical
equipment and disconnect the leads.
▪ Report any problems with apparatus (apparatus out of order, etc.) without delay to a demonstrator.
▪ Handle all apparatus with respect.
▪ Please do not smoke in the laboratory and keep noise levels down.

Page |2
Page |3
The Basics of Pharmaceutical Technology
Pharmaceutical Technology explores the formulation and manufacturing of different dosage forms and the basic

principles of drug delivery. It also integrates experimental drug delivery systems and current trends in the

manufacture of drug delivery systems. It concerns all technologies and methods related to the development of

a pharmaceutical form using natural, semi-synthetic, and synthetic active and auxiliary substances, production

in industry, and use in patients. This includes:

• Drug chemistry

• Process technology

• Innovative instrumentation

• Drug manufacturing

• Drug analysis

• Drug production

• Drug delivery

• Management and analysis of data

• Test and quality control

• Regulatory affairs

Physicochemical principles concerning the preparation and properties of the dosage forms; preparation

techniques for therapeutics; Stability testing; processing of plant products, including freeze drying and micro-

capsulation; Packaging material; good manufacturing practice regulations and quality assurance all encompass

Pharmaceutical Technology. The course aims to teach theory-to-bench applications relating to industrial

operations concerned with the manufacturing of pharmaceutical products: aspects relevant for plant construction

and unit operations like heat transfer, evaporation, drying, refrigeration, distillation, extraction, crystallization,

filtration and centrifugation, mixing and agitation.

Page |4
Dissolution Tester - Optimisation
of therapeutic effectiveness during
product development and stability
assessment
Disintegration Tester - determine whether tablets or capsules disintegrate
within the prescribed time when placed in a liquid similar to the GIT

medium under the experimental conditions

Fluid Bed Dryer – introduction of air to remove


moisture and dry granules

Friability Tester - determine the durability of tablets during


packing processes and transit

Hardness Tester –
Determines the breaking point and structural integrity of e.g.
tablets "under conditions of storage, transportation, and
handling before usage" The breaking point of a tablet is
based on its shape.

Other processing instruments include but are not limited to:

o Agitators: To mix liquids, to promote chemical reactions and to increase heat or cooling
transfers.
o Blowers: Used in solvent recovery and other evaporation applications.
o Boilers: To create steam by applying heat energy to water.
o Capsule equipment: Different equipment is available to fill, polish, and sort capsules.
o Capsule and tablet printers: For printing information like drug names or dosage on the
capsules or tablets.
o Centrifuges: Used for separation of liquids of different densities, or for separation of
liquids from solids.
o Chillers: To quickly lower temperatures.
o Coaters: To coat tablets or capsules with films like a sugar film.
o Cooling towers: Used for cooling liquids or condensing steam.
Page |5
o Dryers and Granulators: For drying liquid preparations into powders or granules.
o Heat exchangers: Used to transfer heat from one medium to another.
o High Pressure Homogenizers: The most efficient fluid processing equipment for
particle size reduction and cell lysis.
o Inspection machines: Allow for visual inspection of the product as it is moved along by
rotating rollers.
o Metal detectors: For detecting tramp metal (bits of metal like nuts, screws, or broken
fragments of machinery) that may have contaminated the product.
o Mixers: For blending and particle size reduction.
o Ovens: For providing necessary heat or drying.
o Pulverizers / Cone mills: Particle size reduction equipment for granules.
o Tablet press: For producing tablets.
o Tablet deduster: For removing any dust created in the tablet press. Often also polishes
the tablet.
o Sifters: For sieving powders or granules.
o Spray coating machines: Used for spray coating liquid onto a powder.
o Tanks: For holding liquids.

Packaging equipment

o Blister packers: Designed to pack tablets, capsules, softgels, injection solutions, syringes
and other small medical items into blister packs. “Deblistering” machines are also
available to recover tablets, capsules or softgels from blister packs.
o Bottling and filling lines: For filling bottles with liquids or containers with tablets,
capsules or softgels.
o Cappers: Designed to place caps onto filled bottles or containers of medicines.
o Cartoners: These sophisticated machines can fill small medicine boxes with blisters
packs, fold and insert leaflets, and close, code, and seal the box.
o Counters: Counts capsules, tablets, softgels and any other small, solid items.
o Induction Sealers: Seals aluminum foil seals to the bottle mouth.
o Labeling equipment: For attaching or printing labels onto the packaging of boxes,
bottles, containers, tubes etc.
o Tube fillers: For filling and sealing tubes of ointments, creams and gels.

[https://www.beei.com/blog/pharmaceutical-manufacturing-equipment]

Page |6
Pharmaceutical Formulations: MCQ
1. Active pharmaceutical ingredients (API)
 Are components in a drug which provide pharmaceutical value
- The API is the component with the only therapeutic action (i.e. pharmaceutical value)
- Carriers are protein transporters or even bloodstream or excipients
 act as carriers to deliver drug to target site

2. Excipients are
Pharmacologically active substances used as carriers for the API
T √ F
Excipients are inactive.

3. Excipients may be determined by


 Stability
-Its chemical and physical properties determine physiological absorption of API in its appropriate form
(maintains API integrity)
It has no medicinal (biological) properties
 Biological action

4. Preservative is an excipient which


 Extends shelf-life and improve safety
-Preservative controls microbial bioburden which can influence drug safety and stability (introduction of
microbes deteriorates stability and pose a hazard upon drug intake)
 Increases drug stability

5. Diluents and glidants


 Improve dosage uniformity
 Do not have the same excipient function
-Diluents act as fillers to improve uniformity; glidants promote particle/powder flow.

6. A vehicle is ______ in syrups


 Different from an excipient
 One or more excipients
Page |7
- A vehicle is an excipient(s) to maintain stability, incorporate flavour etc.

7. The dosage form is the


 Pharmaceutical product with API and excipients
- Complete Unit dose / physical form of drug unit
- Drug delivery is approach to deliver dosage form/drug to site of action for therapeutic effect e.g. injection
 Drug delivery

8. Pharmaceutical dosage forms typically classify


 Solids, liquids and semi-solids
 Gas, liquids and solids – most accurate answer (first one excludes gases)
- Propellants are an example. Solid and liquid dosage forms can take both solid form (e.g. capsules)

9. A transdermal patch is an example of a


 External administrative route
 Solid dosage form
- Drug is administered through the skin hence it is not external; also it is made up of a solid dosage form (better
alternative to oral drugs)

10. Pharmaceutical products can


 Be contemporaneous or industrially manufactured
- Any product should be uniform; multi-forms are not possible across the entire spectrum of drug deliveries.
These products can be manufactured in a pharmacy (behind counter) or by a pharma company
 Be uniform or multi-form

11. A generic drug


 Is equivalent to a novel drug
- Generic and novel drug should be bioequivalent; it also cannot be pharmaceutically different because of its
dosage form, route of administration etc.
 Demonstrates reduced bioequivalence
 Can be clinically equivalent and pharmaceutically different

12. Compounded medicinal products


 Are equivalent to automatic formulated products

Page |8
 Differ in quality control and clinical specifications
- Compounded products are not subjected to automatic pharmacopoeia standards. Specifications can be
inaccurate or overlooked because it is a human (manual) process.

13. Formulations of drugs


 Determine bioavailability and activity
- Bioavailability and therapeutic action is the end goal of a formulation. Polymorphism is most commonly an
attribute of exposure to light (alters drug integrity).
 Indicate pH and polymorphism properties

14. A drug _______ in dosage units


 Vary
 Is consistent
- To ensure uniformity and batch control (dosage units)
 Uniform (uniform in the sense of api and excipient distribution in a single unit)
 Specific

15. Mixing is an example of a _____ operation


 Compiler
- The mixture of substances in one mass
 Basic
 Randomisation (this is merely the result of the process)
 Batch

16. Two main routes of drug administration


 Intravenous and dermal
 Systemic and local
- This covers intravenous, dermal, parenteral etc.

17. Inhalation is an example of a


 Systemic administration
- Inhalation enters circulation by oxygen
 External administration
 Respiratory application (no such term in pharmacy!)
Page |9
18. Transdermal is an example of _________ administration
 Local
 Topical route
 Local and systemic
 Systemic
- Transdermal releases API through into circulation – systemic delivery

19. Production of pharmaceutical tablets require


 Blending of solid powders
- To ensure tablets should have a homogenous mixture of API + Exc. (equal distribution)
 Mixing of API and excipient – this is applicable to any dosage form, not just tablets!
 Clinical validation – clinical validation does not form part of production, ONLY clinical trials.

20. Powder mixing in pharmacy employs


 Particulate systems
- The last three aspects below all form part of particle systems, incl. characterisation
 Particle engineering (simulation studies to investigate particle behaviour and motion)
 Particle size
 Particle adhesion

P a g e | 10
1. Good Manufacturing and Laboratory
Practices (GMP & GLP)

Good Manufacturing Practice (GMP) is a system for ensuring that products are consistently produced and controlled
according to quality standards. It is designed to minimize the risks involved in any pharmaceutical production that
cannot be eliminated through testing the final product.

GMP covers all aspects of production from the starting materials, premises, and equipment to the training and personal
hygiene of staff. Detailed written procedures are essential for each process that could affect the quality of the finished
product. There must be systems to provide documented proof that correct procedures are consistently followed at each
step in the manufacturing process - every time a product is made.

GMP refers to the Good Manufacturing Practice regulations promulgated by the US Food and Drug Administration
under the authority of the Federal Food, Drug, and Cosmetic Act (See Chapter IV for food, and Chapter V,
Subchapters A, B, C, D, and E for drugs and devices.) These regulations, which have the force of law, require that
manufacturers, processors, and packagers of drugs, medical devices, some food, and blood take proactive steps to
ensure that their products are safe, pure, and effective.

GMP regulations require a quality approach to manufacturing, enabling companies to minimize or eliminate instances of
contamination, mixups, and errors. This protects the consumer from purchasing a product which is not effective or even
dangerous. Failure of firms to comply with GMP regulations can result in very serious consequences including recall,
seizure, fines, and jail time.

GMP regulations address issues including record keeping,


personnel qualifications, sanitation, cleanliness,
equipment verification, process validation, and
complaint handling. Most GMP requirements are very
general and open-ended, allowing each manufacturer to
decide individually how to best implement the necessary
controls. This provides much flexibility, but also requires
that the manufacturer interpret the requirements in a
manner which makes sense for each individual business.

P a g e | 11
GMP is also sometimes referred to as "cGMP". The "c" stands for "current," reminding manufacturers that they must
employ technologies and systems which are up-to-date in order to comply with the regulation. Systems and equipment
used to prevent contamination, mix-ups, and errors, which may have been first-rate 20 years ago may be less than adequate
by current standards. [https://ispe.org/initiatives/regulatory-resources/gmp/what-is-gmp]

The principles of Good Laboratory Practice (GLP) define a set of rules and criteria for a quality system concerned
with the organisational process and the conditions under which non-clinical health and environmental safety studies are
planned, performed, monitored, recorded, reported and archived.

Good Laboratory Practice (GLP) – quality system of management controls for research laboratories and organizations
to try to ensure the uniformity, consistency, reliability, reproducibility, quality, and integrity of chemical (including
pharmaceuticals) non-clinical safety tests; from physio-chemical properties through acute to chronic toxicity tests.

GLP was first introduced in New Zealand and Denmark in 1972, and later in the US in 1978 in response to the
Industrial BioTest Labs scandal. It was followed a few years later by the Organization for Economic Co-operation and
Development (OECD) Principles of GLP in 1992; the OECD has since helped promulgate GLP to many countries.

GLP applies to non-clinical studies conducted for the assessment of the safety or efficacy of chemicals (including
pharmaceuticals) to man, animals and the environment.

GLP, a data quality system, should not be confused with standards for laboratory safety – appropriate gloves, glasses &
clothing to handle lab materials safely.

Principles of Good Laboratory Practice (GLP) ensure the generation of high quality and reliable test data related to
the safety of industrial chemical substances and preparations. The principles have been created in the context of
harmonising testing procedures for the Mutual Acceptance of Data (MAD). [https://gmpnews.net/glossary/glp/]

P a g e | 12
LAB Activity: Study the Pharmaceutics laboratory (design, equipment, documentation, etc), and
compile a report with recommendations and motivation, for the laboratory to meet (c)GMP and GLP
requirements, as an instructional laboratory appropriate to Industrial Pharmacy

Reference all work submitted.

Good Laboratories Practice (GLP) in Pharmaceuticals

Learn the main parts of Good Laboratories Practice (GLP) as Design, Location, Equipment, Chemicals &
Reagents, Documentation, Reports and Auditing.

In pharmaceutical laboratories, GLP should be followed. Following are the main points that should be considered
under GLP.

➢ The laboratory should be located designed, customized and maintained to suit the performance of all Q.C. test
and analysis required.
➢ Conveniently located to service the Manufacturing. Dept. but preferably separate to avoid vibration, dust,
internal and external traffic to protect the delicate instruments.
➢ As far as possible there must be separate wings for analytical, instruments, microbiology and sterility etc. and
all wings may be interconnected with the internal door.
➢ There must be an effective airlock, provisions for A.C. and fumigation chamber, the laboratory should be
designed with adequate provision of space for utility, water, solvent storage, extraction dust collection etc.
➢ Laboratory furniture designed such to provide for adaptability, tabletops must be covered properly resistant to
acid, alkali and solvent etc. The floor should be smooth, easy to clean and adequate drainage facility.

Equipment

➢ There must be a written standard operating procedure for each instrument. The instrument should be
located with an adequate place in a separate room under controlled temperature, Instrument must be handle
with almost care and keep it clean all the times. The surrounding also required to be cleaned.
➢ The calibration and maintenance/ service record must be kept and must be done periodically.
➢ The glassware must be calibrated with certified one before use. Particularly the glassware which is
supposed to be utilized for measuring purpose must need calibration before use. All the necessary
instruction regarding operating, handling and care should be display near the instruments.
➢ The light should be adequate.
➢ The electrical system in the laboratory must not be overloaded. Voltage stabilizer must be provided to
protect delicate instruments.
P a g e | 13
Chemicals and Reagents

➢ Storage of chemicals and reagent should be done in a manner it involved in the use, the container of all
chemicals and reagents must be properly labeled.
➢ Transfer of chemical must be done almost care. All analytical reagents and a prepared solution must be
labeled. Records of Molar Solutions entered in the register prepared for the same.
➢ “No chemical reagents pipettes out by mouth, rubber bulb must be used.”

Documentation

The document is a critical factor of the good laboratory Practice. Documentation is the accepted method of
recording information for future reference. The major documents that need to be provided are:

Protocols, logbook for usage, maintenance and calibration

Equipment - well established SOPs.

The following are some of the information that is routinely recorded in a laboratory: Receipt and storage of
samples

- Sampling

- Analytical testing

- Validation

- Calibration

- Data recording

- Operation of instruments

- Reagent preparation

- Training Records

- Organizational charts

- Sampling Procedure

P a g e | 14
- Analytical testing methodology

- inventory/list

- Instrument calibration Data

- Methods validation data

- Analytical testing results and reporting

Quality Control

➢ There must be a well-defined procedure, which covers all the aspects pertaining to the sample i.e. receipt
of the consignment, sampling techniques to be adopted, storage and handling of samples recording and
reporting of analysis.
➢ Every sample that is received must have a distinctive number, which should appear on the label of the
sample and should be stored in the prescribed conditions.
➢ There must be a well-defined sampling procedure in place, which should categorically specify in details
the sampling procedure. If the blending of the sample is permitted, how many can be blended together etc.

Protocols and conduct of a laboratory test

Each laboratory should develop a well –defined protocol to carry out the test and the protocol should
categorically mention.

Records and reports

Every laboratory should maintain records of all the tests performed any of the graphs pertaining to IR, HPLC, etc
should be stored along with the raw data.

For a quick reference, the access to records should be restricted to an authorized person and these records are
preferably stored under lock and key.

P a g e | 15
Safety

There should be adequate facilities and accessories to provide safety for personnel involved in drug testing,
required anti dose for possible accidents that occur, suitable equipment for fire extinguishing in case of an
accidental part.

Auditing Procedure

The quality Assurance Department of the laboratory should constitute a committee who has to regularly audit
their facilities to ensure compliance with GLP requirement.

https://www.pharmaguideline.com/2011/08/good-laboratories-practice-glp.html

P a g e | 16
2. Pharmaceutical Research & Development
Pharmaceutical R&D refers to the pharmaceutical research and development of new medicines. The process begins with
understanding the disease and selecting a target (usually a receptor site on a cell) that can potentially be affected by a drug
molecule (Institute of Medicine 2007; PhRMA 2009). Most commonly, researchers use high-speed screening of huge
libraries of molecules to identify a few hundred leading compounds, though sometimes they create a molecule or
genetically engineer one (PhRMA 2009). In the lab, the leading candidates are tested to see if they absorb, metabolize,
and excrete properly, without being too toxic, and to see if they are distributed to the proper site of action. Adjustments
are made to improve performance, and candidates are tested both in the lab and in animals before clinical trials begin with
humans.

Phase 1 trials measure responses to the drug-candidate in a small number of healthy subjects. Phase 2 trials measure a
candidate’s efficacy and short-term side effects in a few hundred subjects, and Phase 3 trials test these attributes in a few
thousand subjects. At each phase, companies withdraw candidates that are not performing as expected or whose business
prospects make further trials unprofitable. Although these are called “failures,” they are often withdrawals by the company
for commercial reasons. Finally, all the data and analyses from trials relevant to review for approval are submitted to the
regulator.

This extensive testing arose because pharmaceutical companies put very dangerous drugs on the market without proper
testing and explained, “We weren’t told we had to test” (Abraham 1995; Hilts 2003). Finally, in the 1960s they were.
Many of the final candidates they propose to market prove too toxic in the trials that are now required. But the bars for
efficacy and safety are low. New medicines need only prove they are better than an inert substance, not better than existing
medicines. In fact, the companies shaped regulatory rules so that regulators are prohibited from judging how much better
a new medicine is than existing ones, and companies can choose narrow criteria for testing and use surrogate or substitute
end points for clinical measures of improvement (Hilts 2003). Thus all new medicines are “better” than a placebo; but
when independent teams assess how many are better for patients, only 1 in 7 new drugs offers significant advantages,
while many have greater risk of adverse side effects (Carpenter et al. 2008; National Institute for Health Care Management
2000; Olson 2004; Prescrire International 2007). This indicates that most R&D is devoted to finding new medicines to
replace those going off patent in order to maintain high prices and profits – just as one would expect companies to do
when they profit most from finding a variation that can be patented. Nevertheless, 1-2 new drugs a year represent important
advances; so over 20 years that’s 20-40 important new drugs.

The bar for safety is low, because companies design trials using patients least likely to have adverse reactions and end
trials before many side effects develop; most trials are six months or less. They also often do not report patients in trials
who drop out because they cannot stand the side effects. Several other techniques are used to minimize evidence and
measurement of adverse effects (Light 2010a). Most of the time, there is little or no advantage to the new product to offset
its high risk. Thus the “benefit-risk ratio” (which is actually the benefit-harm ratio) is negative.[1]

P a g e | 17
Fees from companies pay for most or all of the costs for regulators to assess the safety and efficacy of new medicines,
and in return the companies demand short review times as a condition of that funding. The result is that about 1 in 7 new
drugs causes serious harm to patients that results in a Black Box warning (the most extreme warning the United States
Food and Drug Administration can require) or withdrawal (Lasser et al. 2002), and many more have mild but dysfunctional
side effects such as dizziness or sleepiness (Carpenter et al. 2008; Olson 2004). Because the causes of many diseases and
how drugs affect them are unclear companies can construct elaborate models about how their products work and widely
promote these theories without ever having to provide actual data to justify them. A few examples of this technique are:
the notion that depression is caused by low levels of serotonin and the SSRI psychotropic medicines work to raise
serotonin levels, that high cholesterol causes heart disease and statins reduce the risk of heart disease by lowering
cholesterol or that menopause is a high-risk condition for heart disease and cancer that needs hormone replacement therapy
(HRT). When complete and objective data have been gathered using clinical measures, all three of these constructed
disease models – and others – have been discredited (Light 2010b; Moynihan and Cassels 2005). Strong incentives to
minimize innovation, undetected risks, biased trials, and made-up or exaggerated health risks are some elements of the
Risk Proliferation Syndrome that has led to prescription drugs becoming the 4th leading cause of death and a major cause
of hospitalization (FDA CDER 2009).

[https://haiweb.org/encyclopaedia/pharmaceutical-research-and-development/]

LAB ACTIVITY: Using one type of dosage form of choice, discuss what research and development would be carried
as well as the methodology employed in this research and development, to develop a ‘blue-print’ for selected
dosage form.

This lab activity links with the next lab activity –manufacturing methods of dosage forms. The latter will be determined by this first lab activity, as it forms the

basis for any type of dosage form.

What to consider in research and development for dosage form:


 Derivative or compound (e.g. from local plant/herb/chemical extract etc.)/ drug discovery
 Type and class of dosage form, as well as a name/brand
 Therapeutic function (e.g. what type of condition it will treat and how it will treat it) & patient class
 Physical and chemical properties (i.e. strength, excipients, stability, shelf life, disintegration properties e.g.
extended release, or delayed…, taste, colour, etc.)
 Pre-formulation studies, experiments/trials that will be carried out to validate intended dosage form prior to
its manufacturing
 Requirements to be met in order to manufacture selected dosage form for the pharmaceutical market
[in summary - stages of drug discovery & development]
P a g e | 18
 Pictures, charts, flow diagrams, etc may be used to answer Lab activity
 All work including images used from other sources should be cited and referenced
 Submission format:
Title
Introduction & Background
Objectives
Methodology
Results (this would include criteria met from tests, experiments, etc. to comply for approval of dosage form –
these may be generated in line with pharmacopoeia standards and monographs)
Discussion
Conclusion and Recommendations (significance of selected dosage form)
References
Appendices (if applicable i.e. flow charts, diagrams, etc.) [50]

P a g e | 19
3. Manufacturing methods of dosage forms
Liposomes were discovered in the mid - 1960s [68] and were originally studied as cell membrane models. They
have since gained recognition in the field of drug delivery. Liposomes are formed by the self - assembly of
phospholipid molecules in an aqueous environment. The amphophilic phospholipid molecules form a closed
bilayer sphere in an attempt to shield their hydrophobic groups from the aqueous environment while still
maintaining contact with aqueous phase via the hydrophilic head group. The resulting closed sphere may
encapsulate aqueous soluble drugs within the central aqueous compartment or lipid - soluble drugs within the
bilayer membrane. Alternatively, lipid - soluble drugs may be complexed with other polymers (e.g.,
cyclodextrin) and subsequently encapsulated within the liposome aqueous compartment. The encapsulation
within/association of drugs with liposomes alters the drug pharmacokinetics.

• Liposomes are biocompatible

• Liposomes can entrap hydrophilic bioactive compounds in their internal compartment and hydrophobic into
the membrane

• Liposome - incorporated bioactives are protected from the inactivating effect of external conditions yet do not
cause undesirable side reactions

• Liposomes provide a unique opportunity to deliver pharmaceuticals into cells or even inside individual cellular
compartments

• The size, charge, and surface properties of liposomes can be easily changed by adding new ingredients to the
lipid mixture before liposome preparation and/or by variation of preparation methods. The clinical applications
of liposomes are well known. The initial success achieved with many liposome - based drugs has fueled further
clinical investigations. One of the drawbacks of the use of liposomes is the fast elimination from the blood and
capture of liposomal preparations by the cells of the reticuloendothelial system (RES), primarily in the liver.

There are a number of different types of liposomal vesicles:

• Multilamellar Vesicles These range in size from 500 to 5000 nm and consist of several concentric bilayers.

• Small Unilamellar Vesicles These are around 100 nm in size and are formed by a single bilayer.

• Large Unilamellar Vesicles These range in size from 200 to 800 nm.

• Long Circulating Liposomes Different methods have been suggested to achieve long circulation of liposomes
in vivo, including coating the liposome surface with inert, biocompatible polymers, such as polyethylene glycol

P a g e | 20
(PEG), which form a protective layer over the liposome surface and slow down its recognition by opsonins and
therefore subsequent clearance of liposomes. An important feature of protective polymers is their flexibility,
which allows a relatively smaller number of surface - grafted polymer molecules to create an impermeable layer
over the liposome surface. These types of modified liposomes demonstrate dose - dependent, nonsaturable , long
- linear kinetics, and increased bioavailability.

• Immunoliposomes To increase liposomal drug accumulation in the desired tissues and organs, the use of
targeted liposomes with surface - attached ligands capable of recognizing and binding to cells of interest has
been suggested. Immunoglobulins (Ig) of the IgG class and their fragments are the most widely used targeting
moieties for liposomes, which can be attached, without affecting liposomal integrity or the antibody properties,
by covalent bonding to the liposome surface or by hydrophobic insertion into the liposomal membrane after
modification with hydrophobic residues.

Injection Methods

Ethanol Injection: Small unilamellar vesicles (with diameter of 30 nm) can be prepared with the ethanol injection
technique [128] . Lipids are dissolved in ethanol and injected rapidly in the aqueous solution under stirring (fi
nal concentrations up to 7.5% (v/v) ethanol can be applied). The method is very easy, having the advantage of
avoiding chemical or physical treatment of lipids. However, there is an extra step to remove ethanol and the
concentration of vesicles produced is rather low. Also encapsulation of hydrophilic drugs is also low, due to the
high volumes used.

Ether Injection: The general principle of this method is the same as ethanol injection. The only difference is that
the lipid is injected slowly in the aqueous solution that is warm [129] . Furthermore, the concentrations used in
this case are somewhat higher (up to 10 m M ) compared to the ethanol injection approach.

Pharmaceutical Manufacturing Handbook Production and Processes, Wiley (2008)

LAB ACTIVITY: [Reference – LAB 2]

Using the methodology (or an alternative manual method) for the selected dosage form (previous LAB),
practically manufacture/create the desired dosage form

P a g e | 21
4. Practical Application in Biotech
LAB Assignment:

1) Identify a Biotech product relevant in Pharmacy, and describe the basic Biotechnology it applies.

2) Research on the product:

▪ Name two Biotech companies that produce the identified Biotech product/similar?
▪ What are they trying to discover? Which laboratory methods are used for the Biotech product and
why?
▪ Its significance in Pharmacy?

3) Prepare a 1.5 page “mini-report”, can have words &/pictures, NON-PLAGERIZED

4) Reference all work

What Is Biotechnology?
In a nutshell, biotechnology is an industry that focuses on novel drug development and clinical research aimed
at treating diseases and medical conditions. Biotechnology companies are almost always unprofitable (some
suggest that the distinction between "biotech" and "pharmaceutical" company lies in profitability), and many
have no real revenue at all.

Biotechnology is also characterized by long development lead times; it can take as much as a decade to get
a new drug from test tube to pharmacy shelf. What's more, there is an overwhelming likelihood of failure, as
85% to 95% of all prospective new drugs fail to reach approval. Still, for those that succeed, the rewards can be
tremendous and "daily doubles" are not unheard of.

(For a background reading, see The Ups and Downs of Biotechnology.)

Differences Between Biotech and Pharmaceuticals


There is more than a little gray area between what is "biotech" and what is "pharmaceutical." Nevertheless,
investors should keep a few general points in mind. From a philosophical standpoint, biotechnology is a risk-
taking enterprise, while the pharmaceutical industry is about managing and diversifying risk.

P a g e | 22
As most biotechs have insignificant revenue, to say nothing of income, dividends are exceptionally rare in
biotech. In contrast, dividends can comprise a significant part of the expected return from a pharmaceutical
stock.

Many biotech companies make no pretense of marketing their own drugs, as they see their expertise being
in research & development. By comparison, marketing and sales is the principal strength of many Big Pharma
companies. As more and more pharmaceutical companies fire scientists and pull back from basic research, they
increasingly become massive marketing machines that need an influx of new products from the biotech world.

The two industries also stand apart when it comes to valuation and business evaluation. Models and valuation
derived from cash flow are quite relevant in assessing pharmaceutical stocks; while many analysts gamely
attempt to construct discounted cash flow models for early-stage biotechs, the reality is that success is often
quite binary ("drug works" or "drug doesn't work").

When considering a potential biotechnology investment, there are several additional factors to keep in mind:

The Pipeline
A biotech's pipeline is everything, and it is the source of the company's presumed and projected value.
Generally speaking, investors should try to focus their attention on companies with multiple Phase 2 programs
(that is, multiple drugs in Phase 2 testing, not a single drug in multiple Phase 2 studies). It is true that single-
product biotechs can be big winners when they succeed, but the reverse is also true – they can suffer crushing
losses if that one and only product candidate fails.

Not All Diseases Are Equally Valuable


Some diseases are huge potential markets, but have ample competition and strict expectations for safety or
performance. For instance, while cancer and arthritis are major diseases with multi-billion dollar potential, there
are numerous drugs already approved and available – if new drugs do not offer something novel (better
efficacy, fewer side-effects, etc.), they may not even get approved, let alone find a large market.

On the other hand, less-common diseases can represent bigger opportunities than people realize. So called
"orphan drugs" target diseases that affect fewer than 200,000 people, but consider that getting just 20,000 users
of a drug costing $50,000 a year (not a bad price for a life-saving drug) means a billion-dollar revenue
opportunity. What's more, companies developing orphan drugs are given some additional assistance in the form
of market exclusivity and less stringent trial enrollment targets.

As a result, almost any disease target can pay off with the right drug. Few people had even thought of restless
leg syndrome as a disease, but drugs sold for this syndrome have done well. Likewise, there is a drug on the
market with the sole stated purpose of making eyelashes grow longer, which shows that one can never
completely dismiss an idea.
P a g e | 23
That said, investors should be careful with companies looking to crack certain diseases. Countless companies
have tried and failed miserably to develop effective drugs for sepsis, Alzheimer's and obesity. While there will
eventually be successes here, and the rewards will be great, there will likely be devastating failures as well, and
the odds are not in the investor's favor.

https://www.investopedia.com/articles/fundamental-analysis/11/primer-on-biotech-sector.asp

P a g e | 24
5. Drug Delivery
Drug delivery is a broad field of research on the development of novel materials or carrier systems for effective
therapeutic delivery of drugs. The drug delivery may be steady, controlled, or targeted drug delivery and is
commonly used methods. Since the advent of medical application systems, numerous drugs are being administered
through various conventional drug delivery dosage forms such as solutions, lotions, mixtures, creams, pastes,
ointments, powders, suppositories, suspensions, injectables, pills, immediate release capsules and tablets, etc., and
so on to treat various diseases. Instances of newer devices with tremendously improved therapeutic potential include
oral controlled release systems, fast dispersing dosage forms, liposomes, taste-masking systems, transdermal
patches, aerosols, and site-specific delivery systems. This chapter is an update on some of the existing drug delivery
technologies for oral controlled release, oral disintegrating dosage forms, test-masking formulations, liposomes,
and targeting drug delivery and transdermal drug delivery.

https://www.sciencedirect.com/topics/engineering/drug-delivery

P a g e | 25
LAB: Drug Delivery and Controlled Release

P a g e | 26
P a g e | 27
P a g e | 28
P a g e | 29
P a g e | 30
LAB (Basic concept): Controlled Release Drug Delivery from Hydrogels

1 Objective

The aim of this project will be to design a drug delivery system capable of delivering a drug over an extended
period of time at a known rate to a local area. The method of using implantable materials to treat tissues locally
is known as controlled release. You will be using gelatin as a controlled release material and food dyes as a
model drug. The gel concentration will be varied to show the effect of increased cross-linking on the rate drug
release. The drug release of each gel concentration will be considered for three conditions–no enzyme, low
enzyme concentration (1 mg/ml), and high enzyme concentration (5 mg/ml). An enzyme is a catalyst for
reactions involving biological molecules. Finally, you will use your data to design an optimal drug delivery
strategy for treating a brain tumour.

2 Background

Controlled release drug delivery is a new way to treat illnesses. The term controlled release refers to the ability
of a drug delivery system to release a drug over an extended period of time at a controlled rate. Over the last 20
years, it has become more popular as a way to treat diseases such as cancer and diabetes. It generally involves
implanting an engineered polymer directly into the organ or system that is affected by a disease. Since the
polymer is implanted directly into the tissues affected by disease, the side effects are often small compared to
systemic drug delivery (i.e. taking a pill or getting a shot). Brain diseases are particularly good candidates for
controlled release techniques because of a physiological feature known as the blood-brain barrier. The blood
brain barrier refers to a tight sheath of cells that surround the blood vessels in your brain. These cells make sure
that only specific types of molecules get into the brain. More specifically, only small (molecular weight less
than 1000 Da), water insoluble molecules can get into the brain. Consequently, the types of drugs that are
developed for brain disease must fit this criteria, which is unfortunate because many promising drugs are water
soluble or large. The use of controlled release techniques has led to tremendous breakthroughs in treating people
brain diseases.

In this activity we will be using a special type of polymer called a hydrogel. Briefly, a polymer is any molecule
made of repeating units, called monomers, that is bonded to itself many times. The most well-known types of
polymers are plastics. Polymers can take on lots of shapes such as rods, loose spaghetti like molecules (i.e.
proteins), helical coils (i.e. DNA), and meshes (Figure 1).
A mesh consists of linear molecules connected to each other by bonds called cross-links. The degree of cross-
linking is partly determined by the concentration of the polymer. At low concentrations a polymer will be a
loose mesh. As you increase concentration, and consequently cross-linking, the mesh will become tighter
(Figure 2). Hydrogels are a special type of mesh like polymer that have the ability to absorb large amounts of

P a g e | 31
water. In fact, most hydrogels are over 90% water by weight. The most well-known hydrogel is gelatin, or more
commonly known as Jell-O . Gelatin is a processed version of collagen which accounts for 1/3 of the protein in
your body1.
Proteins are polymers of molecules called amino acids. There are 22 amino acids and they make up the hundreds
of millions of proteins in biological systems. Enzymes are a special type of protein in your body that have very
specialized roles. One type of enzyme, called a protease, breaks down other proteins.

Figure 1: Examples of the structures of polymers used for drug delivery. Top left: Aerosol particle for drug delivery to lungs. Top right: Degradable

polymer fibres for tissue engineering (Langer and Peppas, 2003). Lower left: Characteristic protein structure. Lower right: DNA double helix amino acids

in proteins. In this activity meat tenderizer will break bonds in the Jell-O , and make it a looser mesh. The enzyme in meat tenderizer is called bromelain

and is found in pineapples. Bromelain is the reason you cannot put pineapples in your Jell-O fruit salad.

There are two ways that a drug can be released from a polymer implant: 1) Diffusion through the implant and into the
surrounding tissue. 2) Degradation of the implant by enzymes, water, or acidic/basic conditions coupled with diffusion
(Figure 3). Some systems are designed not to degrade and release is controlled only by diffusion. However, these systems
may require an extra surgery to remove the implant. Biodegradable systems only require one initial surgery and are
ultimately digested by the body.

1Collagen makes up the majority of your skin and is a constituent in connective tissue and bones. The collagen in
gelatin comes from processing animal carcasses.
P a g e | 32
Figure 2: The effect of concentration on the hydrogel mesh. Left: Low concentration.
Right: High concentration

Figure 3: Mechanisms of Drug Release

In this activity we will be simulating controlled release drug delivery using gelatin hydrogels. The gels will be loaded
with a known centration of a food dye which acts as the drug molecule. Following gelation, 5 mm3 cubes of gel will be
cut and placed into three different aqueous solutions. The first solution is plain water, which simulates a non-degradable
polymer matrix. Here release is due to diffusion through the implant only. The other two solutions will contain a low (1
mg/ml) and high concentration (5 mg/ml) of meat tenderizer. In these second two cases release will be due to both
diffusion and degradation. To quantify the release of the dye molecules from the gels we will use spectrophotometry.
Spectrophotometry is a method of characterizing solution concentration by measuring the amount of light that is
transmitted through a sample. In a clear sample, like a test tube of water, all of the light will be transmitted. In a darker
sample, like water with dye in it, some of the light will be absorbed. The amount of light absorbed can be correlated to
the concentration of the dye.

3 Formulating a Hypothesis

Before you start any scientific investigation it is important to formulate a hypothesis. There are several types of
hypotheses. One of the most common is to predict what you think will happen.
P a g e | 33
In your own words describe what the purpose of this activity is?

How do you think gel concentration, or the degree of cross-linking, will affect the release profile?

4 List of materials

Below is a list of materials you will need for this activity. Make sure you have everything you need before starting the
lab.

 Two packets of gelatin (≈15 g)

 100 mL food dye solution

 Meat tenderizer (≈1 g)

 Graduated cylinders (10 mL and 100 mL)

 Small beaker (50-200 mL)

 Stir rod or stir bar

 Hot plate

 Four-inch petri dish with top

 Razor blade or scalpel

 Parafilm

 Ten 10 mL test tubes that fit in spectrophotometer

5 Experimental Procedure

The procedure for this activity will take place over two class periods. The first period will involve making gels of varying
concentration with different molecules, in our case food dyes, and making the appropriate enzyme solutions. In the second
period the gels will be placed in the enzyme solutions and release will be measured by spectrophotometry. For best results
we are going to take data at 1 hour, 24 hours, and 48 hours.

6.1 Solution Calculations

You will make 31 mL of 5%, 10%, and 15% gelatin with a food dye solution. The first step is to calculate how much
gelatin we need. Fill in the blanks below.

5% ⇒ 31mL × 0.05 = 1.5g


10% ⇒ 31mL× = g
15% ⇒ 31mL× = g
P a g e | 34
The second solution is the meat tenderizer. Calculate the appropriate amount of meat tenderizer for 100 ml of 1 mg/ml
and 5 mg/ml solutions. Fill in the blanks below.

6.2 Making Hydrogels

1. Pour 31 mL of the food dye solution into a beaker.

2. Place on a hot plate and bring it to a boil.

3. Place a stir bar in the beaker and begin stirring or stir by hand with a stir rod.

4. Slowly add the gelatin.

5. Continue stirring until all of the gelatin is dissolved.

6. Pour the hot gelatin solution into the plastic dish and cover.

7. Place the plastic dish in the refrigerator and allow to gel overnight.

8. Repeat steps 1-7 for each gelatin concentration.

6.3 Making Enzyme Solutions

1. Weigh out the amount of meat tenderizer you calculated in the Solution Calculations

2. Pour 100 ml of water into a beaker

3. Add the meat tenderizer to the water and stir until dissolved

6.4 Controlled Release Experiment in Test Tubes

1. Select and clean 9 test tubes. Make sure to remove any fingerprints with alcohol.

2. In 3 test tubes add 5 ml of plain water

3. In 3 test tubes add 5 ml of 1 mg/ml meat tenderizer solution

4. In 3 test tubes add 5 ml of 5 mg/ml meat tenderizer solution


P a g e | 35
5. Cut 5 mm X 5 mm X 5 mm cubes of each of the gelatin concentrations

6. For each gelatin concentration place 1 cube into each of the solutions.

7. Cover the top of the test tube with a piece of parafilm.

8. Fill out the table below so that you can keep track of your solutions.

Table 1: Test tube contents.


Test Tube Gel concentration (%) Solution (mg/mL)

6.5 Calibrating the Spectrophotometer

1. Turn the spectrophotometer on and allow it to warm up for at least 5 minutes.

2. Turn top right knob to the appropriate wavelength (see Table 1 below).

3. Adjust the bottom left knob so that the transmission reads 0%.

4. Place a test tube of water only into the spectrophotometer.

5. Adjust the bottom right knob so the transmission reads 100%.

P a g e | 36
Table 2: Absorption Maxima for McCormick Food Colouring
dye wavelength (nm)
red 500
blue 610
green 625
yellow 430

6.6 Collecting Data with the Spectrophotometer

1 After you have calibrated the spectrophotometer, you are ready to take data.

2 Take a test tube and turn it upside down 3 times. This ensures that the contents are well mixed.

3 Place the test tube in the spectrophotometer and close the lid.

4 Record the Absorbance/Transmission in Table 3 in the next section.

5 Repeat this procedure for each test tube at each time point.

Data and Analysis

Table 3: Absorbance/Transmission data for each test tube.


time (hrs) 1 2 3 4 5 6 7 8 9

P a g e | 37
6.7 Drug Release Profile

Using Excel, enter in the data from Table 3. Normalize the data by dividing each data point by the maximum absorbance
for each test tube. Graph the normalized data (call it fractional release) versus time for each test tube. You should have a
total of nine curves. Fit a logarithmic trend line to each curve and display the equation on the graph.

Results and Conclusions

1. Did you observe any times of more rapid rise in concentration? If so, when? What might explain this?

2. How does the release profile change with gelatin concentration?

3. How does the release profile change with enzyme concentration?

4. The food dye used in this activity is a relatively small molecule. How do you think the release profile would look
if the drug was a much larger molecule?

5. The experiment was performed at room temperature (70 ◦F). How do you think the release profile would change at
body temperature (98 ◦F)? Why?

6. What other variables do you think could influence the release?

References

R. Langer and N. A. Peppas. Advances in biomaterials, drug delivery, and bionanotechnology. AIChE Journal,
49(12):2990–3006, 2003.
http://csip.cornell.edu/Curriculum_Resources/CSIP/Neeves/KNeeves_studentversion.pdf

P a g e | 38
6. Specialty Pharmaceuticals

LAB Assignment:

▪ Give one example of a medicine/medical product classed as a Specialty Pharmaceutical


▪ Briefly explain how this product is designed and formulated
▪ Briefly name & explain the laboratory methods employed to test the product (to meet requirements &
objective)
▪ What is the significance of ‘Specialty Pharmaceuticals’ in Pharmacy?

Specialty Pharmacy: What Pharmacy Students Should Know

Spurred by health care reform, the rise of specialty drugs continued in 2014 in an ascendance that is projected to
account for nearly half of all pharmacy spending within the next 2 years.

The growth of specialty medications means a growth in opportunities available to pharmacists. The increasing use
of specialty drugs will lead to more roles that will require a hightouch approach to patient management. As the
medications in the pipeline become more specialized, the skill sets of pharmacists must expand and evolve to
meet the needs of their patients initiating treatment with complex biologics.

Understanding current trends in specialty pharmacy will prepare pharmacy students to take advantage of these
new opportunities in the specialty space after graduation.

The following explores 10 major trends in specialty pharmacy as a new era of health care focuses the attention of
all players—patients, payers, physicians, and manufacturers—squarely on the specialty marketplace.

1. The Cost of Specialty Drugs Is Skyrocketing


The cost of specialty medications has achieved what some call “unsustainable growth,” with a wide-ranging
impact on patients and providers alike.

Catamaran Corporation reported that, in 2013, specialty medications accounted for 23.5% of total drug spending
by only 2.1% of patients. The average cost of a specialty prescription in 2013 was nearly $2900, which represents
a 17% increase from the previous year.

P a g e | 39
Additionally, spending on new specialty treatments increased 7.7%, to $7.5 billion in 2013 and accounted for
69% of total spending on new brand name drugs.

Whether that growth can be sustained in the long term by patients and providers is another question.

“Specialty drugs account for a disproportionate share of overall drug spending because of their extremely high
cost,” reports America’s Health Insurance Plans (AHIP), a trade association for the country’s health insurers.

AHIP suggests that a number of measures should be taken to mitigate spending growth while improving access to
specialty drugs, including encouraging alternative payments and incentive structures for new drugs and
technologies and shortening the exclusivity time for biologics.

2. The Specialty Marketplace Continues to Heat Up

Large companies are buying up independent specialty pharmacies as specialty medications take up a bigger piece
of the drug spending pie.

This past year saw a number of big players elbow their way into specialty pharmacy in response to the projected
growth.

“It’s hard to build a specialty pharmacy from scratch, so a lot of existing players are starting to acquire some of
the smaller, faster-growing companies that have already built specialty capabilities,” said Adam J. Fein, PhD,
president of Pembroke Consulting and chief executive officer of Drug Channels. “What we’re seeing is that
specialty pharmacy is really a set of services, not a distinct industry. So what we think of as pharmacy is going to
be merging, and is merging, with what people call specialty pharmacy.”

Dr. Fein added that this move toward an expanded specialty marketplace requires companies to have their eyes
firmly fixed on the future.

“Looking forward a few years, most of the drugs dispensed to patients are going to be low-cost generic drugs or
high-cost specialty drugs,” he noted. “Pharmacies need to figure out how they’re going to play that balanced
approach.”

3. The “Marriage” of Specialty Pharmacy and Accountable Care Organizations Continues to Strengthen

The goals of accountable care groups and specialty pharmacies are tightly aligned in supporting and caring for the
needs of patients.

P a g e | 40
With the passage of the Affordable Care Act (ACA) in 2010, accountable care organizations (ACOs) emerged
with the goal of reducing costs and improving patient adherence. To achieve those goals, ACOs are increasingly
looking to specialty pharmacies that are already armed with an extensive history of hands-on specialized care.

“Accountable care organizations aligned incentives around managing the whole patient and really integrating the
patient experience from the point of diagnosis with the doctor all the way through to successful therapy,” said
chief executive officer of Avella Specialty Pharmacy Rebecca M. Shanahan, esq, in a video interview on
SpecialtyPharmacyTimes.com. “What that is going to do is put a higher value and focus on the set of services that
specialty does. Specialty is often times both the integrator among different kinds of providers and the last link to
the patient relative to the success or failure they have in terms of understanding their disease and managing their
therapy.”

Despite the aligned goals between the 2 entities, ACOs are not anticipated to have a drastic impact on the
management of specialty medications or biopharmaceuticals, according to research by the Journal of Managed
Care Medicine.

The study indicated that ACOs would focus on efficiency and more general quality metrics rather than on
pharmaceutical access.

4. As Drug Costs Go Up, Adherence Goes Down

Reduced medication adherence is a by-product of escalating drug costs for patients feeling the economic pinch.

As specialty drugs become more prevalent and the costs continue to climb, so too does the potential for
nonadherence by a patient population unable to afford these high-priced treatments.

Analysis by CVS Health found that patients who use specialty drugs carry a total health care cost that is up to 8.5
times more than the cost for patients who do not use specialty drugs. This disparity is driven by the fact that
patients who use specialty drugs are more likely to have multiple diagnoses, see more specialists, fill more
prescriptions, and have more lab tests, emergency department visits, and hospitalizations than the average patient.

Additionally, the shift to high-deductible health care plans by many employers has the potential to lead to reduced
adherence and delayed or bypassed care due to high out-of-pocket costs.

Specialty pharmacies will need to remain aware of the impact of the ACA on coverage for specialty medications.

P a g e | 41
“An unfortunate consequence of many of the exchange plans people are purchasing under Obamacare is that the
enormous costs are being shifted to patients,” Dr. Fein said. “That is really not good for the health care system or
the patients.”

5. New Patients Continue to Flood the Specialty Space Through Health Care Reform

More patients are using health care services, which means more money will be spent in the specialty space.

With 32 million people projected to be newly insured by the end of the decade, the growing demand is expected
to drive specialty medications to account for approximately 40% of all pharmacy spending by 2016.

In response to this demand, manufacturers have focused their efforts to align with the growing specialty market.
Specialty medications currently comprise more than 50% of the pharmaceutical pipeline, with specialty products
accounting for 15 of 27 novel new drugs approved by the FDA in 2013. More than 30% of new drugs approved
last year were oncology drugs, which are projected to represent approximately 14% of the worldwide market
share by 2018, with targeted oncology therapies accounting for more than $69 billion in sales.

“Increasing access to insurance is increasing the demand for specialty drugs,” Dr. Fein said. “Overall, I think
getting insurance coverage is a good thing for these patients, but on the other hand, the way it’s been implemented
could ultimately drive up health care costs. There is a lot of work to be done to fix the reform that’s been
implemented.”

6. Growing Use of Technology Will Improve Patient Outcomes

Increasingly, specialty pharmacies are turning to technological breakthroughs to improve adherence and data
analytics.

The phrase “There’s an app for that” has moved beyond the commercialization of convenience tools and into the
realm of enhancing patient care and outcomes. Increasingly, providers are looking to technical solutions for some
of the age-old problems that have plagued specialty pharmacy. For example, Avella Specialty Pharmacy began
using a GlowCap, a device that fits on top of a pill bottle and wirelessly uses lights and sounds to remind patients
to take their medication.

Further innovation looms on the horizon, such as Next IT’s virtual health assistant platforms. “Why can’t we use
a virtual agent embedded in [smartphones] to help with reminders for medications, and answer questions about
the disease process?” asked Thomas Morrow, MD, chief medical officer for Next IT in a video interview on
SpecialtyPharmacyTimes.com. “An individual patient may have 2000 to 3000 potential questions about their

P a g e | 42
chronic illness…this is the ultimate Google-type of device, but specific to their needs.”

7. Health Care Is Evolving into a Patient-Centered Ecosystem

With health care reform putting the emphasis on patient management, specialty pharmacy moves to the forefront.

Specialty health care is evolving to focus less on medication management and more on the total patient. As a
result, providers are seeking to improve interaction between the physician, pharmacy benefit management, health
care plan, care management nurse, and specialty pharmacy to create a total care ecosystem that fits each patient’s
needs.

“Everything we do in specialty has a patient at the end of the activity,” said Randy Falkenrath, MBA, CVS Health
senior vice president of specialty pharmacy services, during the Armada summit. “It’s really all about the services
we provide in improving their care, improving their quality of life, and finding the opportunity, ideally, to
improve a length of their life span.”

In this new consumer-driven market, specialty pharmacy has already found itself uniquely situated with a strong
prior record of direct interaction with physicians and patients alike.

“Specialty pharmacy, just through the nature of the pharmacy business, has always been more patient-centered
than some of the large institutional organizations,” Dr. Fein said. “One of the things about specialty products is
that you’re treating a small patient population. That means it’s easier to devote resources to individuals who have
these conditions.”

8. Biosimilars Are Coming, Possibly Sooner than Expected

On the heels of the FDA accepting the first application filed for a biosimilar, the United States may finally be on
the brink of biologic price competition.

Novartis Group company Sandoz entered uncharted waters in US drug manufacturing on July 24, 2014, as it
became the first US company to file an application for a biosimilar with the FDA through the pathway created by
the Biologics Price Competition and Innovation Act of 2009.

Branded biologic oncology products alone currently represent more than $20 billion in global spending and are
anticipated to be the top target for biosimilar development over the next 5 years.

Estimates vary on just how much relief biosimilars will offer on the cost of specialty drugs, due to the fact that

P a g e | 43
manufacturers who have long since recouped their original investment could subsequently lower the price of the
reference product once a biosimilar competitor hits the market. Drug companies are still putting in their due
diligence to prepare for the impending flood, however. For example, Pfizer has extensive educational programs to
provide information for patients and physicians on the potential of biosimilars.

“We need to find a way to communicate the importance of biosimilars and the high potential they have, especially
the quality behind the molecules and how the entire concept of biosimilarity is based on the molecular structure of
the candidate drug. It is a big challenge,” Dr. Coindreau said.

9. Limited Distribution Networks Are Expanding for Greater Access to Specialty Drugs

Older strategies that limit distribution channels are evolving as more players enter the market.

Manufacturers previously limited distribution networks for a variety of legitimate concerns. Among them were
the ability to provide the appropriate level of patient care, special handling requirements for their products, a
small patient population, and making sure the products stay in legitimate channels to protect their patents.

With more major players entering the specialty space, manufacturers have been forced to rethink these prior
strategies that are no longer as applicable as they were even just a few short years ago.

“There are so many options available for specialty pharmacies, who are now often owned or controlled by other
entities the manufacturer already does business with,” Dr. Fein noted. “Manufacturers are taking advantage of the
diversity of offerings, while at the same time meeting the needs and demands of these large, powerful customers
they interact with. Specialty drugs today are being launched with networks twice or more as large as a few years
ago, so as more pharmacies and companies get into the specialty business, you’re going to see these networks get
bigger.”

10. The Cost Will Be Weighed Against the Cure

With the huge price tag for treatments, stakeholders in specialty pharmacy are evaluating whether a medication’s
efficacy is worth the high cost.

Controversy continues to surround Gilead’s hepatitis C treatment sofosbuvir (Sovaldi), which has achieved cure
rates of up to 95% while carrying a $1000 per pill price tag.

With the cost of a typical round of treatment being $84,000, the conversation about the drug has ignited a larger

P a g e | 44
debate about the escalating cost of specialty drugs and the sustainability of the system to continue paying for
them.

“If you put it in the context of a cure, then the value you’re receiving is beyond a price. I think it is priceless,” said
Stephen Vogt, PharmD, chief executive officer and president of BioPlus Specialty Pharmacy, in a video interview
with SpecialtyPharmacyTimes.com. “Yes, it will cost $84,000 to $100,000, but you’re cured. Think about that, in
my lifetime, we can say a patient is cured. They will not have liver cancer, they probably will be able to keep their
liver, it will decrease medical costs…Yes, the initial cost is heavy, but if you look at the long-term benefit for
that, I think in economics it pays for itself.”

https://www.pharmacytimes.com/publications/career/2014/pharmacycareers_november2014/southwestern-
oklahoma-state-university-college-of-pharamcy

P a g e | 45
7. Small Scale Manufacture of Tablets

Practical: Tablet Manufacture by Wet Granulation Method

Granulation is a unit operation in which small powder particles are gathered together to form agglomerates called
granules. To achieve cohesion between the powders, it is necessary to include adhesive substances called binders or
granulating agents within the formulation. It is a common practice to make use of a granulation solution since it is more
effective in comparison with the same quantum of the dry powder binder. Powder mixing, in conjunction with the
cohesive properties of the binder, enables the formation of granules which when duly compressed using tablet
press forms tablets with the desired properties.

Reasons for granulation


There are several reasons for converting powders or blends of powders into granules and they include:
1. To enhance the flow properties of powder mix.

2. To prevent segregation of powder components during tableting or storage.

3. To reduce the incidence of dust production.

4. To reduce cross-contamination and hazard associated with the generation of toxic dust that may arise during
manufacturing processes.

5. To improve the compression characteristics of drug substances.

6. To improve the appearance of the final product.

Ideal characteristics of granules -


For a successful manufacture of tablets, the granules must possess the following characteristics:
P a g e | 46
1. All formulation ingredients should be uniformly distributed in the granules.

2. A good granulation should be as near spherical in shape as possible to ensure reproducible flow which in turn
ensure constant tablet weight throughout the batch.

3. Granules of different sizes or density must not separate in the hopper as a result of machine vibration.

4. Granules should possess good disintegrating properties and lubrication to reduce die-wall friction.

5. The granules should have sufficient fines to fill empty spaces between coarse granules for better compression
characteristics.

6. A tablet granulation should have sufficient physical strength to form strong tablets when compacted.

Manufacture of tablets by wet granulation method

Wet granulation method is a process of size enlargement in which fine powder particles are agglomerated or brought
together into larger, strong and relatively permanent structure called granules using a suitable non-toxic granulating
fluid such as water, isopropanol or ethanol (or mixtures thereof). The granulating fluid can be used alone or as a solvent
containing binder or granulating agent. The choice of the granulating fluid depends greatly on the properties of the
materials to be granulated. Powder mixing, in conjunction with the cohesive properties of the granulating agent, enables
the formation of granules. The characteristics and performance of the final product, greatly depends on the extent to
which the powder particles interact with each other to form aggregates (granules).

Mechanisms of granule formation in wet granulation:

P a g e | 47
Mechanisms of granule formation in wet granulation, Image source: gruppotpp
The four key mechanisms of granule formation as originally outlined by Ennis include:

1. Wetting and nucleation


This is the first and an important phase in granule formation. It involves the initial wetting of powder bed and existing
granules by the granulating fluid to form nuclei. This step is largely influenced by spray rate or fluid distribution as well
as feed formulation properties, in comparison with mechanical mixing. It is worth noting that the nucleation process,
that is, the initial coalescence of primary particles in the immediate vicinity of the larger wetting drop is strongly linked
with the wetting stage.

2. Coalescence or ball growth


In the coalescence or ball growth stage, partially wetted primary particles and larger nuclei come together to form
granules composed of several particles. The more general term of coalescence refers to the successful collision of two
granules to form a new, larger granule.

3. Consolidation
As granules increase in size, they are consolidated by compaction forces due to bed agitation. The extent of the
consolidation depends on the agitation in the granulation equipment and the resistance of the granules to deformation.
This phase in granule formation controls internal granule porosity, and therefore final properties of the granules e.g.,
granule strength, hardness, or dissolution.

4. Attrition or breakage
At this stage, formed granules break into fragments which bind to other granules forming a layer of material over the
surviving granule.

The above mechanisms can occur simultaneously in all processes of wet granulation. However, certain mechanisms
may dominate in a particular manufacturing process depending on the type of equipment used.

P a g e | 48
Steps in wet granulation method of tablet production

Wet granulation method of tablet production involves the following processing steps:

Step 1: Weighing and mixing of formulation ingredients (excluding the lubricant).


This step involves the weighing, sifting and introduction of specified quantities of drug substance(s), bulking agent,
filler or diluent, and disintegrant into a powder mixer. These ingredients are mixed using either a planetary bowl mixer,
ribbon/ trough mixers, rotating drum mixer or high-speed mixer until a uniform powder mix is achieved. The mixing
efficiency can be enhanced by the use of powders that have similar average particle size, although this is often not the
case in many mixing operations.

There are many diluents available in commerce but those used in wet granulation method include lactose, microcrystalline
cellulose, starch, powdered sucrose, mannitol, fructose, sorbitol, calcium phosphate and calcium sulphate. Among these
diluents, the most widely used are lactose, because of its low cost, solubility and compatibility with most drug substances
and excipients and microcrystalline cellulose, because of its easy compaction, compatibility with most formulation
ingredients and consistent uniformity of supply. Diluents are usually selected based on the manufacturer’s experience
with the material, its relative cost, and its compatibility with the drug and other excipients.

Disintegrants used in wet granulation include croscarmellose, sodium starch glycolate, sodium carboxymethylcellulose,
polyvinylpyrrolidone (PVP), crospovidone, cation exchange resins, corn and potato starches, alginic acid and other
materials that counteract the effect of binders and the physical forces of compression used in forming the tablets.
Croscarmellose (2%) and sodium starch glycolate (5%) are often used because of their high water uptake and rapid
action.

Step 2: Preparing the damp mass


Here, the binder solution is mixed with the powder mixture to form an adhesive mass which can be granulated. The
amount of binding agent used as well as the quantity of fluid required to form a damp and coherent mass is part of the
operator’s skill; however, the resulting binder-powder mixture should compact when squeezed in the hand. The use of
P a g e | 49
insufficient binder tends to poor adhesion, capping and soft tablets. Excessive binder solution yields hard tablets with
slow disintegrating properties.

Among granulating agents are solutions of povidone, an aqueous preparation of cornstarch, molasses, methylcellulose,
carboxymethylcellulose, glucose solution and microcrystalline cellulose.

Dry binder or nonaqueous solution may be used for drug substances that are adversely affected by aqueous solution.
Colorants or flavoring agents may be added to the binding agent to prepare a granulation with an added feature.

Step 3: Wet screening/ Screening the dampened powder into pellets or granules
The wet massed powder blend is screened using 6- to 12- mesh screen to prepare wet granules. This may be done by
hand or with suitable equipment that prepares the granules by extrusion through perforations in the apparatus. The
granules formed are spread evenly on trays and dried in an oven.

Step 4: Drying of moist granules


The screened moist granules are dried in an oven at a controlled temperature not exceeding 550C to a consistent weight
or constant moisture content. The drying temperature and the duration of drying process depend on the nature of the
active ingredient and the level of moisture required for the successful production of satisfactory tablets. Shelf or tray
drier and fluidized-bed drier can be used for this purpose.
Step 5: Sizing the granulation by dry screening
The dried granules are passed through a screen of smaller size than that used to prepare the moist granules. The size of
the final granules is dependent on the size of the punches (and hence the final tablet size). Screens of 14- to 20- mesh
size are generally used for this purpose.

Step 6: Lubrication of granules


After dry screening, the dried and screened granules are separated into coarse and fine granules by shaking them on a
250 mesh sieve. Appropriate quantity of lubricant is passed through a 200 – mesh sieve. This is mixed with the fine
granules before the coarse granules are incorporated. The quantity of lubricant used varies from one formulation
scientist to another but usually ranges from about 0.1% to 5% of the weight of the granulation.

Examples of lubricants commonly used in wet granulation include magnesium stearate (most preferred), calcium
stearate, stearic acid, wax, hydrogenated vegetable oil, talc, and starch.

It is worth noting that disintegrant may be added in step 1 (intragranular) or in step 6 (extragranular) and sometimes in
both steps (intragranular – extragranular). Intragranular – extragranular incorporation appears to be the best method of
incorporation because the extragranularly added portion causes immediate disruption of the tablet into the previously
compressed granules while the portion added intragranuarly cause further erosion of the granules to the original powder
particles.

P a g e | 50
Step 7: Compression of granules into tablets
Here, the mixed granules are compressed in a single punch or multi-station tablet press fitted with the appropriate
punches and dies.

Compressed tablets may be coated if there is need to mask the taste of unpleasant drugs, increase the aesthetic appeal of
uncoated tablets, modify or control the release of therapeutic agents from tablets. This is achieved by enclosing or covering
the core tablet or granules with coating solutions.

Recent advances in wet granulation technology

In an urge to improve commercial output of pharmaceutical formulations, wet granulation process has witnessed various
technical and technological innovations such as:

1. Steam granulation

2. Moisture-Activated Dry Granulation/ moist granulation

3. Freeze granulation

4. Thermal adhesion granulation

5. Melt granulation/ thermoplastic granulation

6. Foam granulation and

7. Reverse wet granulation/ reverse-phase wet granulation

Advantages of wet granulation method of tablet manufacture

1. Wet granulation modifies the properties of formulation ingredients to overcome their tableting deficiencies.
Granules formed are relatively more spherical than the powders and have better flow properties.

2. Improved compressibility of powders resulting from wet granulation process allows the use of low pressure
during compression. This reduces machine wear and thus improves the life of the machine.

3. The process makes use of conventional excipients and therefore is not dependent on the inclusion of special
grades of excipients.

4. It ensures better content uniformity, especially for soluble low-dose drugs.

5. The process may improve the dissolution rate of poorly soluble drugs by imparting hydrophilic properties to the
surface of the granules.

6. Wet granulation prevents segregation of components of a homogenous powder mix during processing,
transferring, handling and/or storage, leading to reduced intra- and inter-batch variability.

7. Tablets manufactured by wet granulation are amenable to post-processing unit operations such as tablet coating.

P a g e | 51
8. Wet granulation reduces the level of dust present during manufacturing process thereby reducing the incidence of
cross-contamination and risk to workers.

9. Wet granulation reduces the amount of air entrapment thereby increasing powder compressibility.

Limitations of wet granulation

1. Wet granulation often requires several processing steps.

2. The cost of wet granulation is higher because of the time, labour, energy, equipment and space required for the
process.

3. The process is not suitable for thermolabile and moisture sensitive materials.

4. Migration of soluble dyes may occur during the drying process.

5. Incompatibilities between formulation ingredients will be aggravated by the granulating solvent which tends to
bring them into close contact.

6. There is a possibility of material loss during processing due to the transfer of material from one unit operation to
the other.

7. Dissolution rate of tablets manufactured by wet granulation may decrease with ageing.

In spite of all these limitations, the manufacture of tablet using wet granulation still persist due to the
following reasons:

1. Availability of extensive data on manufacture of existing products by wet granulation. This has made
pharmaceutical companies reluctant to adopt new techniques except there exist a compelling reason to do that.

2. Experience over the years in formulation has also shown that wet granulated granules and tablets assure good
content uniformity.

3. The drying process can be manipulated to produce granules with the desired moisture content.

LAB: Preparation of Paracetamol tablets by Wet Granulation

Granulation is a unit operation in which small powder particles are gathered together to form agglomerates called
granules. To achieve cohesion between the powders, it is necessary to include adhesive substances called binders or
granulating agents within the formulation. It is a common practice to make use of a granulation solution since it is more
effective in comparison with the same quantum of the dry powder binder. Powder mixing, in conjunction with the
cohesive properties of the binder, enables the formation of granules which when duly compressed using tablet press
forms tablets with the desired properties.

P a g e | 52
For a successful manufacture of tablets, the granules must possess the following characteristics: -

1. All formulation ingredients should be uniformly distributed in the granules.

2. A good granulation should be as near spherical in shape as possible to ensure reproducible flow which in turn
ensure constant tablet weight throughout the batch.

3. Granules of different sizes or density must not separate in the hopper as a result of machine vibration.

4. Granules should possess good disintegrating properties and lubrication to reduce die-wall friction.

5. The granules should have sufficient fines to fill empty spaces between coarse granules for better compression
characteristics.

6. A tablet granulation should have sufficient physical strength to form strong tablets when compacted.

Equipment

Analytical balance scale Wet and Dry granulator

Weighing boat Fluid bed dryer

Spatula Sieve

Measuring beaker Tablet Press

Measuring cylinder

Mortar & Pestle

Excipients and API

Paracetamol Corn starch

Lactose Water

MSC

Silicon Dioxide

Magnesium Stearate

P a g e | 53
Working Formulae: -

Ingredients Function Quantity (g) – Quantity (g) –


(1 tablet) (……tablets)

Paracetamol Active ingredient 6.28


Lactose Bulking agent 1.725
MSC Binder 0.176
Silicon Dioxide (colloidal) Glidant 0.440

Magnesium Stearate Lubricant 0.088


Corn Starch Disintegrant 0.440
Total weight 0.088

Procedure

[A] Weighing

1. Tare the analytical balance scale.


2. Weigh out the ingredients as per formulae on the analytical balance scale using the weighing boat, spatula and
measuring beaker.
3. Add all the weighed out ingredients a mortar.
4. Mix all the weighed ingredients in the mortar, using the pestle (no grinding).
4.1 Add a sufficient amount of water to allow the powder to mix, using a mortar and spatula – DON’T ADD
TOO MUCH WATER

[B] Granulation

5. Set up the granulation machine (choose an appropriate sieve size and rpm setting, based on the standards in the
British Pharmacopoeia).
6. Sift the granules to remove all the fine particles.

[C] Drying

7. Fluid bed dryer machine: Set up the machine by selecting an appropriate rpm and time setting, based on the
standards in the British Pharmacopoeia.
8. Add the granules into the Fluid Bed Dryer tubes (oppositely arranged)
9. Coat the granules with lubricant – don’t add too much lubricant
10. Mix

P a g e | 54
11. Place all the granules in the measuring cylinder of the fluid bed dryer apparatus and dry the granules for the set
time period.

[D] Tableting

12. Tablet Press: Set up the machine by transferring the dried granules into the hopper of the machine.
13. Let the machine run for the set time period (based on the British Pharmacopeia), while collecting the tablets in a
beaker

[E] Packaging and Labeling

14. Package the tablets into appropriate containers


15. Label the packaged tablets (with a legal tablet label, based on the Namibian Medicine and Controlled
Substances Act)

NOTES

1. It is important to ensure that not too much binder (water) is added to the mixture as this will only unnecessarily
prolong the drying process in the fluid bed dryer machine.
2. The excipients are multi-faceted. Thus they may included in a formulation for different multiple reasons. The
choice is based on the availability of the excipients.
3. Final packaging must be put in an appropriate container, based on the final product’s rheological and light
sensitivity properties.

Conclusion

The manufacture of tablets by wet granulation involves several unit operations. In order to manufacture tablets with
desired characteristics, it is important to have a good understanding of the processes involved.

https://www.pharmapproach.com/tabletmanufacturewetgranulationmethod/

P a g e | 55
8. Solid Dosage Forms:

Powders and Granules


Powder is a mixture of finely divided drugs and/or chemicals in dry form. Powders can be used internally and
externally (e.g., external applications to the skin). Dry powders, however, can be taken orally by some patients
who are unable to swallow other solid dosage forms such as capsules and tablets. Although powders per se are
not used extensively in therapeutics, they are widely used in preparation of various dosage forms.

Pharmaceutical powders are formulated to be exist as fine particles. The powders are then smooth to the touch
and nonirritating to the skin. Powders generally range from 0.1 to 10 micron in size. The size of the particles are
often expressed as a number which corresponds to the mesh screen size of a sieve. The screen size indicates the
number of openings in the mesh screen per inch. For example, a # 40 sieve has 40 openings per inch in the
screen mesh. Particles that can sift through that mesh are said to be "40 mesh" size.

Granules are agglomerates of powdered materials prepared into larger, free flowing particles. They typically fall
within the range of 850 μm (No. 20 sieve) to 4.75 mm (No. 4 sieve) size. Granules generally are made by first
blending the powders together and then moistening the mixture to form a pasty mass. The mass is passed through
a sieve and then dried in air or in an oven. They are prepared as a convenience for packaging, as a more stable
product due to less surface exposure, and as a popular dosage form. Granulations are also used as intermediates
in the preparation of capsules and tablets, since they flow more smoothly and predictably than do small powder
particles.
[https://drugs-bd.blogspot.com/]

P a g e | 56
P a g e | 57
P a g e | 58
Pharmaceutical Capsules
Capsules are gelatin containers that contain medicines. These are common pharmaceutical dosage
forms those are easy to manufacture. Capsules do not require any complex formulation to
manufacture.
Capsules are of following two types: 1. Soft gelatin capsules 2. Hard gelatin capsules

Soft Gelatin Capsules: These are also called as softgel. Soft gelatin capsules are manufactured as
single piece capsule shell. These capsules are suitable for a non-aqueous solution such as oils.
Active ingredients are dissolved or suspended in an oily base. Manufacturing and filling of soft
gelatin capsules are done same time on the same machine. The content of capsule is filled during
the manufacturing the shell. These capsules melt within the minutes in the stomach releasing the
content having within it. These contain 5% to 14% of moisture content. Sometimes soft gelatin
capsules are printed with brand names or strengths.
Hard Gelatin Capsules: Hard gelatin capsules consist of two parts, one is body and other is the cap
of capsules. These are supplies in closed condition but without locking. Generally, body and cap have
different colors. Hard gelatin capsules are dry in nature and active ingredients are filled in powder
form in the hard gelatin capsules. The body part of the capsule is filled, closed by the cap and locked
by pressing genteelly by the capsule machines or manually. These are disintegrated in 3 minutes and
release the content. These are hygroscopic in nature and contain 12% -16% of moisture. Sometimes
it is required to release the drug at any target then hard gelatin capsules are filled with target release
pellets. In some cases, tablets are also filled in the gelatin capsules.
(https://www.pharmaguideline.com/2013/11/types-and-size-of-capsules.html)

Practical: Capsules

1. Prepare _____ g of oral powder composed of:

API and excipient

2. Use formulated oral divided powder to make capsules with a consistent dose 3. Label and dispense accordingly

Compound Log:

Ingredients Official Formula (1 capsule) Working Formula = ____ capsules

API: . . . . . . . . . . . . . . 100 mg

Diluent 150 mg

Total 250 mg

P a g e | 59
Working formula is calculated based on number of capsules to be formulated/ i.e. 6 capsules,

100 mg x 6 capsules = 600 mg

150 mg x 6 capsules = 900 mg

1500 mg

Post-Lab Investigation

1. Briefly discuss the classification of powders

2. How would You evaluate the powder quality, ensuring that capsules meet required standards?

3. What other quality analyses are performed to ensure that the capsule product dispensed to patients are of high
quality?

Kindly reference Your work

P a g e | 60
Suppositories

P a g e | 61
9. Semi-solid Dosage Forms:

Ointments

Ointments are single phase, hydrophilic or hydrophobic systems with sufficient consistency to
allow the formulator to stably and homogeneously suspend dissolved or dispersed APIs.
Hydrophilic ointments have the advantage of being less greasy or oily during and after application
and they are easily washed off of skin after use. They also provide an excellent vehicle for the
topical delivery of hydrophilic drugs.

The biggest challenges for formulators of hydrophilic ointments are ensuring that APIs remain
solubilized or in a specific habit or polymorphic state throughout the life of the product, preventing
the separation of fluid from the formulation (i.e. weeping) and ensuring that the formulation has
acceptable spreadability on the skin. These challenges are overcome by the proper blending of
compatible fluids and solvents with higher melting point solids.
(https://pharmaceutical.basf.com/en/Drug-Formulation/Ointments.html).

LAB: Salicylic acid Ointment

P a g e | 62
LAB: Acetylsalicylic acid ointment

▪ API: Acetylsalicylic acid or Disprin tablets


▪ Excipient: Emulsifying BP Ointment (contains all ingredients listed in table above)
▪ Ratio of preparation – API:Exc. 3: 7
▪ *Package and label product

Aspirin has been one of the oldest drugs in the field of medicine, with a wide range of applications. In
dermatology, aspirin has shown benefit in a variety of disorders. Recently, reduction of melanoma risk with
aspirin has been demonstrated. Although an analgesic to begin with, aspirin has come a long way; after
cardiology, it is now found to be useful even in dermatology.

Aspirin although an old and inexpensive drug, has shown promise for a variety of dermatologic disorders.
With its role in reduction of melanoma risk aspirin has also found to have a place in oncology. With further
advances in the field of molecular pharmacology, in the near future more newer indications could be
discovered for aspirin, one of the oldest drugs in the field of medicine (Bubna A. K. (2015). Aspirin in
dermatology: Revisited. Indian dermatology online journal, 6(6), 428-35).

P a g e | 63
Pharmaceutical Pastes
Pharmaceutical pastes are generally composed of ointment bases that contain a high concentration
(frequently 50% w/w) of dispersed drug. The viscosity of pharmaceutical pastes is greater than that
of pharmaceutical ointments (https://phclub15.files.wordpress.com/2017/01/ointment.pdf).
Water‐in‐oil (W/O) bases (hydrophilic and non-hydrophilic) usually have a low surfactant content but
a significantly high oil content and are generally used to create occlusive films. Pastes by definition
have a high solids content.
Pastes (solid particles dispersed in semisolid) are one of the pharmaceutical dosage forms in which
the dose uniformity and thus the efficacy of drug products either local or systematic, depend on the
spreadability of the paste. However, the particle size of the drug and its shape along with its content in
the paste will affect the spreadability of the paste. Better spreading paste will provide more contact
area, which effects the penetration and absorption.

LAB: Salicylic acid /Zinc Oxide – Paste (Lassar’s Paste)

Materials:

▪ Mortar & pestle, balance and weigh boats, spatula and dispensing container

Method:

▪ Mix the ingredients together in a mortar and pestle, then add salicylic acid to mixture and then mix in
the white petrolatum.

▪ Dispense and label accordingly


P a g e | 64
Ingredient Ingredient weight (g) Ingredient weight (g) Practical
with Salicylic acid without salicylic acid Ingredient weight (g)

Zinc Oxide 25g 25g ________g

Starch 25g 25g ________g

White Petrolatum 48g 50g ________g

Salicylic Acid 2g 0g ________g

Post-Lab Questions

1. Tabulate the significant differences between an ointment, a paste and a cream


2. What methods are employed in Industrial pharmacy for the formulation of pastes? Briefly explain
3. How would you assess the quality of the paste? What QC tests would You perform and why?
4. Describe the significance of pastes as a vehicle in pharmacy
Kindly reference all work

P a g e | 65
Pharmaceutical Lotions
Lotions are dilute aqueous solutions or suspensions meant for external application to the body. They may be
applied to the skin, hair or eyes. They are applied without friction. They may contain: Humectants ; to retain
moisture on the skin after application Alcohol ; which evaporates quickly causes a cooling effect and leaving
the skin dry.

Experiment
AIM: To prepare and submit 10 ml of calamine lotion

Apparatus: Mortar pestle, beaker (250 ml), measuring cylinder (10 ml. 50 ml), Spatula and glass rod

Procedure:
I. Mix calamine and zinc oxide together in a mortar till a homogenous powder is obtained.
2. Pour glycerin and triturate to form a homogeneous paste.
3. Add a small portion of lime water and continue trituration till a very smooth levigate is obtained.
4. Dilute the smooth levigate with small portion of calcium hydroxide solution (lime water), then with large
portion following by trituration after each addition till attaining the final volume.
Uses: Soothing and protective in cases of skin irritation and dermatitis.
Lime water: Saturated calcium hydroxide solution (1.5g/L at 25ºC sparsely soluble).

P a g e | 66
Pharmaceutical Gel
A gel is a solid or semisolid system of at least two constituents, consisting of a condensed mass enclosing and
interpenetrated by a liquid.

Delivery of drugs to the skin is an effective and targeted therapy for local dermatological disorders. Topical
gel formulations provide a suitable delivery system for drugs because they are less greasy and can be easily
removed from the skin.

What is Nurofen gel used for?

Nurofen gel can be used to relieve pain and inflammation associated with:

• muscle or rheumatic pain


• backache
• sprains, strains and sports injuries
• severe throbbing or stabbing pain along a nerve and in the area supplied by the nerve (neuralgia)
• pain from non-serious arthritic conditions.

How does Nurofen gel work?

Nurofen gels contain the active ingredient ibuprofen, which is a type of medicine called a non-steroidal
anti-inflammatory drug (NSAID).

Ibuprofen works by blocking the action of an enzyme in the body called cyclo-oxygenase (COX).
In certain arthritic and rheumatic conditions, or if you have an injury, COX makes substances
called prostaglandins. These cause pain, swelling and inflammation. When you apply ibuprofen to the
skin, it stops the production of prostaglandins in the underlying tissues. This reduces inflammation and
pain in the local area.

Ibuprofen is absorbed into the bloodstream less when you apply it to the skin than when you take it by
mouth, which means gels and sprays are less likely to cause side effects than products you take by
mouth. However, absorption can still occur, particularly if you use large amounts on large areas of skin.

P a g e | 67
How do I use Nurofen gel?

• There are currently four Nurofen gel products: Nurofen gel, Nurofen maximum strength gel, Nurofen
joint and back pain relief gel and Nurofen joint and back pain relief max strength gel. Read the leaflet
provided with each product regarding how much gel to use.
• Apply the gel to the skin over the painful area and massage it in gently.
• Nurofen gels can be used up to four times in 24 hours, or as directed by a doctor. Leave at least four
hours between applications

[https://www.netdoctor.co.uk/medicines/aches-pains/a8282/nurofen-gel-ibuprofen/]

Practical: Ibuprofen Gel

Composition ration 1:2

Reference: Article

Procedure:

1 First dissolve Ibuprofen in water at 5-10 degrees Celsius


2 Mix the ibuprofen-water solution (while still liquid) with gel
3 Ensure that solution and gel are evenly mixed to ensure maximum distribution of API
4 Package and label product

P a g e | 68
10. Liquid Dosage Forms:
Are liquid preparations in which the therapeutic agent and the various excipients are dissolved in the chosen solvent
system.

Pharmaceutical solutions may contain a range of excipients, each with a defined pharmaceutical purpose. Examples
of these include

the vehicle, usually purified water


co-solvents, e.g. propylene glycol, glycerin, alcohol
agents specifically to enhance the solubility of the therapeutic agent in the vehicle, e.g. surface-active agents
preservatives, e.g. parahydroxybenzoate esters (methylhydrox- ybenzoate and propylhydroxybenzoate), boric
acid and borate salts, sorbic acid and sorbate salts, phenolics
sweeteners, e.g. glucose, saccharin, aspartame
rheology (viscosity) modifiers, e.g. hydrophilic polymers (cellulose derivatives, alginic acid,
polyvinylpyrrolidone)
antioxidants, e.g. sodium formaldehyde sulphoxylate, butylated hydroxyanisole, butylated hydroxytoluene
colours
flavours
buffers to regulate the pH of the formulation, e.g. citrate buffer

(http://pharmacypractice1.blogspot.com/2015/03/pharmaceutical-solutions.html)

P a g e | 69
Practical: PREPARATION OF SALICYLIC ACID SOLUTION 5%

Formulation:

Salicylic acid----- 5gms - ______________________________ = _____gms

Industrial methylated spirit 70%---- 100 ml - ______________________________ = ______ml

Preparation for 5% solution with a volume of 20 to 30 mls:

Dissolve the salicylic acid in the industrial methylated spirit.

Packaging:
Should be labeled for “external use only”
Salicylic acid solution should be packed in a well closed container. Salicylic acid solution is highly flammable and
should be stored in a cool place away from open flames. The solution should be used within 3 months. Expired salicylic
acid solution often has lower alcohol content due to evaporation of alcohol.
USES:
Salicylic acid solution is used for the treatment of acne. It has keratolytic properties.
Dose:
Apply the solution twice daily. Therapy generally needs to be continued for several months.
Pregnancy/breast feeding:
Teratogenic effects of salicylates in high oral doses have been shown in animal studies. But harmful effects in humans
from external use of salicylic acid have not been described. Evaluate the benefit/risk ratio before using salicylic acid
during pregnancy.
Following external use salicylic acid is excreted in breast milk. No adverse effects in the child have been reported
following the mother’s external use of salicylic acid.
Side effects:
Local irritation may occur, but is rare. When irritation or sensitization reactions develop, stop using this preparation and
do not use it again.
Intoxication:
Excessive or long-term use of salicylic acid containing preparations may cause systemic intoxication. This is unlikely to
occur following topical application of salicylic acid, unless for long-term use on large areas of the skin. Children are
more vulnerable to systemic intoxication because they have a relatively large skin surface. Systemic intoxication is
characterized by: slight intoxication: sweating, abdominal pains, dehydration and loss of hearing. More severe
intoxication: excitation, confusion, fever and convulsions.
Accidental ingestion of salicylic acid solution produces complex clinical symptoms, because at least three toxic
substances are involved: salicylic acid, alcohol and methanol. Induction of vomiting with syrup of ipecacuanha may be
initiated in addition to oral administration of 5% sodium bicarbonate solution.
Lower concentrations of salicylic acid, for example 2% can be prepared.

P a g e | 70
Syrups
Syrups are concentrated solutions of a sugar such as sucrose in water or other aqueous liquids. When purified water
alone is used in making the solution of sucrose, the preparation is known as syrup, or simple syrup. In addition to
sucrose, certain other polyols, such as glycerin or sorbitol, may be added to retard crystallization of sucrose or to
increase the solubility of added ingredients. When the aqueous preparation contains some added medicinal substance,
the syrup is called medicated syrup. Flavored syrup is one which is usually not medicated, but which contains various
aromatic or pleasantly flavored substances and is intended to be used as a vehicle or a flavor for prescriptions.

P a g e | 71
Elixirs
Elixirs are clear, pleasantly flavored, sweetened hydro- alcoholic liquids intended for oral use. They are used as flavors
and vehicles for drug substances and when such substances are incorporated into the specified solvents, they are classified
as medicated elixirs. The main ingredients in the elixir are ethanol and water but glycerin, sorbitol and propylene glycol,
flavoring agents, preservatives and syrups are often used in the preparation of the final products. The alcoholic content of
elixirs varies greatly from elixirs, only a small quantity to those that contains a considerable portion as a necessary aid to
solubility. For example; Aromatic Elixir USP contains 21 – 23% ethanol; Compound benzaldehyde elixir NF contains 3
- 5% ethanol. Elixirs may also contain glycerin and syrup. These may be added to increase the solubility of the medicinal
agent or for sweetening purpose. Some elixirs contain propylene glycol. This solvent is a satisfactory substitute for both
glycerin and ethanol. Sucrose increase viscosity and decreases the solubilizing properties of water and so must be added
after primary solution has been carried out. A high alcoholic content is maintained during preparation by adding the
aqueous phase to the alcoholic solution. Elixirs should always be brilliantly clear. They may be strained or filtered and if
necessary, subjected to the clarifying action of purified talc or siliceous earth.

Practical
AIM: To prepare and submit 10 ml of elixirs as non-aqueous preparation.

Apparatus: Beaker (250 ml), measuring cylinder (10 ml. 50 ml), Spatula and glass rod.

Materials: compound orange spirit, alcohol, glycerin, sucrose, purified water.

Formula: Rx

Compound orange spirit 10 ml.

Alcohol 100 ml.

Glycerin 200 ml.

Sucrose 320 g.

Purified water, q.s. 1000 ml.

Ft.: elix.

Procedure:

1) Take three quarter of water, warm on hot plate dissolve the sucrose into this warm water.

2) Mix the alcohol, glycerin, to it.

3) Filter the solvent mixture through a hard filter paper, returning, if necessary, the first portions of the filtrate until it
passes through clear.

4) Add enough of vehicles make the volume upto10 ml. Note (Alcohol content: 8 - 10 %)

P a g e | 72
Phenobarbitone Elixir

P a g e | 73
Suspensions
are coarse dispersions in which internal phase (therapeutically active ingredient) is dispersed
uniformly throughout the external phase. These are heterogeneous systems consisting of 2 phases. A
solid in liquid dispersion in which the particles are of colloidal size.
Pharmaceutical suspension is a biphasic system composed of finally divided insoluble solid material
suspended in liquid medium. The average size of suspended particles ranges from 0.5 µm to 5 µm in
most of the pharmaceutical suspensions. Suspensions essentially facilitate the administration of
insoluble and often distasteful substances in a form which is pleasant in taste.

Uses: It is used as media, during X-ray


examination.
Dose: whole amount in one time.
Storage: store in tight closed container.
Secondary label (auxiliary): shake well
before use

P a g e | 74
Practical: Preparation of various types suspensions and determination of their sedimentation
parameters

BACKGROUND

Mg. Carbonate is an insoluble but diffusible solid. In flocculated suspension, the practical settle more quickly than
practical of deflocculated one. But the sediments from a cake like structure in deflocculated suspension make
dispersibility by shaking a big problem. Comparing these 2 types of suspensions, the flocculated one is pharmaceutically
acceptable or preferable; however, a control flocculation is desirable to achieve a control sedimentation with ease of
dispersibility.
P a g e | 75
Electrolyte and ionic surfactant can be used as flocculating agents to produce a flocculated suspension. Suspension
prepared without flocculating agent is a deflocculated one.

The physical stability of the suspension is assist by determining the sedimentation volume or degree of flocculation.

Materials and apparatus:


− Mg.carbonate,
− Al.chloride,
− Water,
− Pestle and mortar
− Measuring cylinder
PROCEDURE
Step-1: Preparation of suspension
Deflocculated suspension
1 The light magnesium carbonate is powdered in a mortar.
2 Then water is added with trituration to make a cream and dilute sufficiently.
3 Then it is transferred to a measuring cylinder. The mortar is repeatedly rinsed with little water every time and the
rinsed mixture is added subsequently to adjust the volume.
Flocculated suspension
1 The light magnesium carbonate is powdered in mortar.
2 Aluminium chloride is dissolved in little water and added with trituration to make a cream. The cream is transferred to
the measuring cylinder after dilution with water.
3 The mortar is repeatedly rinsed with little amount of water every time and the rinsed mixture is added subsequently
to adjust the volume.
Step-2 Evaluation of suspension
1 The suspension in the cylinders are thoroughly shaken to make the dispersions uniform.
2 The cylinders are kept undisturbed on a flat surface after shaking.
3 The volume of sediment at different time: 0, 10, 20, 30 and 40 mins is measured.
Original volume of suspension is 100 ml
The degree of flocculation at one hr=sediment volume of flocculated suspension in one hour/sediment volume of
deflocculated volume in one hour.3
P a g e | 76
(Though degree of flocculation is related to the ultimate sediment volume in flocculated and deflocculated
suspension, it is not feasible to achieve or determine ultimate volume with in the practical period.)
The plot of sedimentation volume in Y axis and time in X axis.
(Sedimentation volume quickly decrease in flocculated suspension compared to deflocculated suspension initially
but ultimate sedimentation volume of flocculated will be higher).

Time Volume of Sedimentation Volume of Sedimentation


(min) sediment for volume for sediment for volume for
flocculated flocculated deflocculated deflocculated
suspension in suspension suspension in suspension
ml ml
0
10
20
30
40

CONCLUSION

The flocculated and deflocculated suspension is prepared and evaluated for sedimentation volume.

REFERENCES

More HN, Hajare AA. Practical Physical Pharmacy. Career Publications. 2010: 203-205. 1. Gaud RS, Gupta GD.
Practical Physical Pharmacy. CBS Publisher and Distributors. 2009: 81-84 2. Mohanta GP. Physical Pharmacy
Practical Text, Pharma Book Syndicate. 2006: 72-74. | Reference: Article

P a g e | 77
Emulsions
are heterogeneous, thermolabile biphasic liquids, containing two immiscible liquids which are made
miscible by adding emulsifying agent. Emulsions are a class of dispersed system in which a one
immiscible liquid is dispersed uniformly in a liquid dispersion medium.

Castor Oil Emulsion


Method of preparation

1. Select a sample-sized mortar and a suitable pestle.

2. Place in the mortar 2 g. acacia (very finely pulverized).

3. Measure 8 ml of oil in a dry measure and pour on the gum allowing some time for the measure to drain.

4. Triturate the oil and gum together for a few seconds.

5. Measure exactly 4 ml of water in another measure.

6. Begin to triturate the oil and, gum again using whipping motion, but not a grinding action. While triturating, add water
all at once.

7. As the water reaches the oil-gum mixture, increase the rate of trituration, taking care to maintain the whipping motion
and work in one direction only.

8. Continue trituration till the primary emulsion is formed. This is when the mixture becomes white in colour and
cracking sound is heard.

9. Continue to triturate for a little longer before attempting to dilute.

10. Measure small amount of water and add them drop wise, with continuous trituration, to the primary emulsion and
then dilute with remaining amount of water and transfer to a measure.

11. Rinse the mortar with more water and adjust the volume.

P a g e | 78
Practical: Water/Oil Pickering Emulsion [stabilized by Magnesium Oxide particles]
– a potential system with two active substances (Paracetamol & Griseofulvin/fulvicin)]

Materials
Oil
Mg Oxide
Water
Paracetamol
Griseofulvin
Petroleum jelly

I. Oil Phase: Mg oxide is added to the jelly by trituration until it is homogenous. Oil containing
Griseofulvin/antifungal is added to the initial mixture and agitated (shaking/stirrer) for 3 minutes.
II. Aqueous Phase: Dissolve paracetamol in distilled water. Homogenise the mixture using a stirrer.
III. Emulsification: Gradually add phase II to phase I. Add drops of water while mixing. Homogenise
mixture using a stirrer.

Reference: Article
Package and label product (as applicable)

P a g e | 79
11. Quality Control of Dosage Forms

Quality control testing of solid dosage forms:


Evaluation for visual appearance, labelling, odour, taste, texture, hardness and friability.
Moisture content: Limits are given in official compendia.
Standards and tests of identity: designed to demonstrate clearly that the specimens examined contain the
active ingredient(s) they purport to contain.
Standards and tests of homogeneity: Apply test for uniformity of weight.
Standards and test for purity: for potentially harmful degradation compounds that may be generated
during production and storage of dosage forms and for contaminants whose presence may indicate deviation
from GMP.
Standards and assays for the active ingredient(s) and for degradation products: It provides quantitatively
permitted range per tablet or capsule of average weight.
Uniformity of content: It involves individual analysis of for a given number of dosage forms to assess
possible variation. This test is particularly more important where the declared quantity of active ingredient in
a single tablet is less than 5 mg and in case of sugar-coated tablets 10 mg or less.
Standards and tests of performance: Designed to provide some assurance that the dosage form will
release its active ingredient as intended. Dissolution rate tests for poorly soluble drugs, potent drugs and
cases with dissolution problems; disintegration test to supplement dissolution rate tests.
Stability indicating tests: This is to take into account the deterioration in activity or strength of the drug
product that may occur because of degradation of the active ingredient in the dosage form as well as aspects
of physical instability of the product e.g. development of undue colour or colour instability.
Storage conditions.
[https://www.pharmapproach.com/quality-control-requirements-pharmaceutical-dosage-forms/]

Uniformity of diameter, thickness and hardness

Objectives:

To identify the uniformity of diameter, thickness and hardness for 10 samples of tablet.

To test the ability for tablet to withstand sufficient mechanical strength and fracture/ erosion during manufacturing and
handling.

All the tablets must undergo the quality testing experiment in order to ensure that all the designed tablet are uniform in
physiological properties to provide effective pharmacology effects to the patients. Diameter and thickness of the tablets
should be in suitable size in order to provide effective effect for patient and provide better oral intake. Hardness of tablet
is the resistance of the tablet to chipping, abrasion or breakage under conditions of storage, transportation and handling
before usage. Hardness test is a diametric compression test which require a force to break a tablet. Tablet thickness is
important for the evaluation of the properties of tablets: ability to withstand the shock of handling, packing and shipping,

P a g e | 80
physical parameter in the control of tablets and it is related to solubility. Tablet diameter is important for the consistency
of the appearance of the tablets.

Materials: 10 tablets

Apparatus: Weighing boat, Tablet Testing Instrument

By calculating the deviation of diameter, the uniformity of diameter of the tablets can be proven. According to theory,
tablets with diameter 12.5 mm or more than 12.5 mm would have not more than 3% of individual deviation from the
average diameter. The results we obtained showed that all the tablets have achieved percentage difference below than 3%
which means that each tablets had obeyed the theoretical value of standard diameter.

During manufacture of tablets, Diameter, thickness and hardness are three important aspects to be considered in order to
produce the quality tablet and also produce higher therapeutic effect.

Tablet friability

Objectives: To determine the tablet friability.

Theoretically, friability is the tendency for a tablet to chip, crumble or break following compression. This tendency is
normally confined to uncoated tablets and surfaces during handling or subsequent storage. It can be caused by a number
of factors including poor tablet design, low moisture content, insufficient binder, etc. For obvious reasons, tablets need to
be hard enough such that they do not break up in the bottle but friable enough that they disintegrate in the gastrointestinal
tract. The strength of a tablet plays a very important role in its marketing and dissolution. The mechanical strength of
tablet or granules can be determined by its hardness and through friability test. To examine this, tablets are subjected to a
uniform tumbling motion for specified time and weight loss is measured.

Materials: 10 tablets

Apparatus: Weighing boat, Tablet abrasion and friability tester

In this experiment, friability can be defined as the percentage of weight loss by tablets due to mechanical action during
the test. Tablets are weighed after and before the testing and the friability are expressed as the percentage loss. Friability
refers to the ability of the compressed tablet to avoid fracture and breaking during transport. According to the theory,
compressed tablet should not lose more than 1%. Therefore, we can say that the tablets are able to avoid fracture and
breaking during transport.

Uniformity of weight of tablets and capsules.

Objective: To test the uniformity of weight of tablets and capsules.

The safety and efficacy of drug products can be guaranteed when their quality is reliable and reproducible from batch to
batch. To ensure the requisite quality, drug manufacturers are required to test their products during and after
manufacturing and at various intervals during the shelf life of the product. Like all other drugs, tablets and capsules are
subjected to those pharmacopoeia standards which deal with “added substances” with respect to their toxicity, interference

P a g e | 81
with analytical methods. Such standards are found in the British Pharmacopoeia and United Pharmacopoeia and include
uniformity of weight (mass).

Tablets and capsules.


Test of uniformity of weight (mass) is carried out to tablets and capsules to ensure accurate and consistent
dosage form to be administered by patients. However, uniformity of weight test is not applicable to tablets and
capsules required to comply with test for uniformity of contents as drug substances present in lesser proportion
is demonstrated by content variation. Procedures in this experiments follow procedure in appendix XII C, BO
2011. Limit is the acceptable range of value of uniform weight.

In the experiment, since the average mass obtained for 20 tablets is 0.5908 g that is greater than 250mg, thus
minimum 18 tablets should not deviate from 0.5908 g by ±5%. The uniformity of weight of tablets is acceptable
as all 20 tablets do not deviate from ±5%. For capsule, the average mass obtained is 0.2947g that is more than
300mg, thus minimum 18 capsules must not deviate from average mass by ± 7.5%. The uniformity of capsule
is acceptable as all the 20 capsules do not deviate from ±7.5%. We assume that the concentration of drug, which
is the weight of drug per weight of dosage form, is uniform.

The results of the test of uniformity of weight may not be accurate as there is always substance left in the
capsules when they are emptied from the shells. The shell of capsule should be completely emptied before
weight. Besides, the balance should be calibrated first before doing the experiment.

Dosage performance tests

The performance test is one of a series of tests that compose the specification in a United States Pharmacopeia
(USP) dosage form monograph. For an orally administered, nonsolution dosage form, it is usually satisfied by
either a dissolution or disintegration procedure. Complete disintegration is defined as that state in which any
residue of the unit, except fragments of insoluble coating or capsule shell, remaining on the screen of the test
apparatus or adhering to the lower surface of the disc, if used, is a soft mass having no firm core. Dissolution
acceptance criteria are usually set in private negotiations between an applicant and a regulatory agency. With
information about this private agreement and other information provided in a sponsor’s Request for Revision
to USP, the USP’s Council of Experts elaborates a public dosage form monograph. Based on the relationship
between the regulatory decisions and the Request for Revision, the USP dissolution procedure links to a
regulatory judgment about bioavailability and bioequivalence and, ultimately, to a judgment about safety and
efficacy. The current dissolution procedure and acceptance criteria are perceived as having worked well over
the years and are generally accepted.
P a g e | 82
Objectives

The objectives for disintegration test is to determine how long the time taken needed to tablet for disintegrate
and whether it is disintegrating properly when placed in a liquid medium under the experimental condition in
this experiment.

The objectives for dissolution test is to determine the amount of active ingredient(s) released from a solid oral
dosage form, such as a tablet or a capsule, using a known volume of dissolution medium within a predetermined
length of time. This test method may not be applicable to certain oral dosage forms.

Apparatus and materials

For disintegration test:

 500 mL distilled water, 500 mL beaker, disintegration machine (include disc, mechanical device to
control up-down movement (28-32 cpm), device to control temperature (37 °C))

For dissolution test:

 1000mL cylindrical vessel, motor to regulate paddle speed, water bath (37°C)

Disintegration is the process of break-up of the tablet. It is important for a drug to be in solution form for it to
be absorbed from a solid dosage after oral administration.

The disintegration test is a measure of the time required under a given set of conditions for group of tablets to
disintegrate into particles which will pass through a 10 mesh screen. From the result obtained, observation
shows that after 60 minutes of the operation, all the 6…. Tablets disintegrate. Those tablets disintegrate after
43 minutes. There is a residue left which is the coating film seen. The time of the of disintegration is a measure
of the quality. This is because, for example, if the disintegration time is too high, it means that the tablet is too
highly compressed or the capsule shell gelatin is not pharmacopoeial quality. And also if the disintegration time
is not uniform in a set of samples being analysed, it indicates the excipients if the drug is not well distributed
or lack of batch uniformity.

In an experiment, we can’t predict how well the dosage form will release its active ingredient in vivo. This is
because it does not mimic the conditions of GIT. Studies have shown that the agitation of the gastric contents
during normal contractions is quite mild in contrast to the turbulent agitation produced in the disintegration test
apparatus. The low order magnitude of agitation in the stomach produces substantially higher disintegration in
vivo than those obtained using the USP apparatus. Furthermore, the particles of the disintegrated tablets are not
dispersed throughout the stomach but remains as aggregate. Thus, the tablet disintegration test is limited to
manufacturing control of variations in individual products and is not a measure of bioavailability. Nevertheless,
it is used to provide a simple ad useful means for monitoring and controlling the quality of tablet.

P a g e | 83
Assay: Content of API i.e. Ibuprofen
Objective: To calculate the amount of API

Apparatus :
Filter Funnel, Filter paper, Beaker, Retort stand, Erlenmeyer flask, Dryer, Burette, Weighing boat
Materials:

-Ibuprofen powder
-Chloroform
-96 % Ethanol
-Phenolphthalein
-0.1 M sodium hydroxide

Procedures:

1. 9925g of Ibuprofen powder is weighed using electronic balance (calculate the required amount based
on 7.94 g of the Ibuprofen powder contain 4g of Ibuprofen, the required amount of Ibuprofen is 0.5g,
hence 0.9925 g of Ibuprofen powder is taken)
2. The 0.9925 g Ibuprofen powder is mixed with 20ml chloroform measured by 50ml measuring cylinder
in an Erlenmeyer flask and is left for 15 minutes. After that the mixture is filtered using filter paper and
filter funnel and poured into the 100ml beaker.
3. The residue in the 100ml beaker is washed with 10ml chloroform three times and the filtrate is gently
evaporated by using hair dryer. Then, the residue is washed with 100ml of 96% ethanol. A few drops of
phenolphthalein solution are added to the filtrate as indicator.
4. The filtrate is titrated against the 0.1M sodium hydroxide solution to an end point with phenolphthalein
as indicator (which the solution turns pink).
5. The content of Ibuprofen is calculated if each ml of 0.1M sodium hydroxide used is equivalent to
0.02063 g of Ibuprofen (C13H18O2).
Observation: in titration, the solution changes from colourless to pink

The weight of Ibuprofen powder Ibuprofen = 7.94 g (contain 4g of Ibuprofen)

From the experiment :


The weight of Ibuprofen containing 0.5g active ingredients, y
Note : 7.94g of Ibuprofen powder contains 4g of ibuprofen.

Calculation :
y = (7.94 / 4) x 0.5
=0.9925 g

Titration of 0.1 M sodium hydroxide =22.0 ml

Amount of Ibuprofen (theory) =0.5 g

Note: Each amount of 0.1M sodium hydroxide is equivalent to 0.02063 g of ibuprofen.

Calculation:

The amount of Ibuprofen after experiment = 22.0 ml x 0.02063 g


=0.4539 g

P a g e | 84
(Exercise/Revision):

Questions

1. What are the objectives of the tests for uniformity of diameter and uniformity of content?

2. State the types of tablets and capsules that must be tested for uniformity of diameter and uniformity of content.

3. Give reasons for the non-compliance to test for uniformity of weight.


.
4. Why is dissolution test suitable to be used for batch-to-batch quality control?

5. Describe other apparatuses that are listed in pharmacopoeias that can also be used in dissolution testing of
tablets and capsules.

Aerosols:
Drug delivery to the respiratory tract locally has become an increasingly effective and important therapeutic method for
treating a variety of pulmonary disorders, including chronic abstractive disease, asthma, bronchitis, pneumonia, and cystic
fibrosis. Increase in prevalence respiratory disorders is a major factor driving the growth of global respiratory inhaler
devices market. The effectiveness of pharmaceutical aerosols therapeutic performance is affected by various factors such
as type and characteristics of propellants, whose properties are vapor pressure of propellants, viscosity and density flash
point and also other factors such as type and characteristics of active ingredients, containers, valves, and actuators, along
with percentage of moisture content and mechanism of emitted dose deposition, spray pattern, efficiency of valve crimping,
and measurement of particle size aerosols. The purpose of this book chapter is to discuss the in-process and finished product
quality control tests for pharmaceutical aerosols based on pharmacopeia standards and specifications.

[Abdo, R. W., Saadi, N., Hijazi, N. I., & Suleiman, Y. A. (2020). Quality control and testing evaluation of pharmaceutical
aerosols. Drug Delivery Systems, 579–614. https://doi.org/10.1016/B978-0-12-814487-9.00012-0]

Identification
This qualitative confirmation assessment is also known as the identity test it is applied to verify the type of the
pharmaceutically active ingredient(s) claimed to be found in the pharmaceutical aerosol. On the other hand, this
test can be also used to differentiate between the components that have similar or closely related structures that
might be existing in the formula. Identification tests should be specific for the drug substance, e.g., IR
spectroscopy, high-performance liquid chromatography/ultraviolet spectrophotometer (HPLC/UV),
HPLC/mass spectrometry (HPLC/MS), or gas chromatography/mass spectrometry (GC/MS) is generally
acceptable (Allen, 2013, Uddin et al., 2015, Uddin et al., 2016).

P a g e | 85
Assay
This quantitative active ingredient identity test is also known as the content test. It is used in the determination
of the strength or content of the pharmaceutically active ingredient(s) in the aerosol. Using ultraviolet
spectrophotometer, absorbance is determined for 1 mL of spray solution taken after adequate dilution.
Concentration is determined from the standard plot, and the drug content is calculated as a percentage of the
theoretical value (Parmar and Patel, 2017).

Drug content= Actual drug content ×100


Theoretical drug content
The amount of drug substance should be determined per weight unit or per volume unit for multidose products,
where the usual assay limits for medicinal products apply [EMEA (European Medicines Agency Inspections), 2006].

Relevant impurities
Characterization of potential impurities and degradation products that may result from the degradation of the
active ingredient in manufacturing or storage or as a result of the new drug synthesis is an important requirement
for aerosols safety evaluation (Allen, 2013, Uddin et al., 2015, Uddin et al., 2016).
Several methods are applicable in this issue, such as HPLC and gas chromatography, and one of the innovative
detectors is charged aerosol detection technology that delivers a good performance to measure analyte charge
that is in direct proportion to the amount of the analyte present (Zhang et al., 2013).

[https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7155167/]

Semi-solids:

 Evaluation for visual appearance, colour, odour, labelling, and homogeneity


 It must appear uniform and elegant.
 Particles of suspensions should be well distributed.
 No hard cake formation of particles.
 The suspension is poured in a transparent glass container and it should be checked if there is any coagulated material
adhering to the inside wall of the container
 This technique is mostly done for orally administrated suspensions.
 Variation in colour indicates poor distribution.
 Variation in taste is generally due to particle size and crystal habit.  Change in either colour, odour or taste indicates
chemical instability
 Loss of water
 Consistency
 Softening range (for suppositories)
 Viscosity

P a g e | 86
 This test is carried out to ensure that the final preparation is pourable and will not cause any problem during filling
and during handling by the patient
 Stability of a suspension is dependant on the sedimentation rate of dispersed phase which is dependant on the viscosity
of the dispersion medium.
 So this test is carried out to ensure optimum viscosity of the medium so a stable, redispersible suspension can be
formed
 The calculated values are compared with standard values and if any difference is found necessary corrective action is
taken to get optimized viscosity.  The viscosity can be measured by:- a)cup and bob viscometer b)cone and plate
viscometer
 Particle size distribution
Particle size is examined by a)optical microscopic method b)sedimentation method c)conductivity method
The suspension is examined for particle size distribution and crystal habit under microscope with camera.  It is mainly
used to differentiate between flocculated and deflocculated particles
 So this test is carried out by microscopically analysis and find out particle size range of drug then it is compared with
optimum particle size required.  If any difference is found, strict monitoring of micronization step is ensured
 pH
 So pH of the different vehicles, phases of suspension before mixing and after mixing are monitored and recorded time
to time to ensure optimum pH environment.
 Different methods are: a)Dip a piece of pH paper into the sample. b)pH meter
 Assay of active ingredients and of degradation products
 Identification test for active ingredient and possible contaminants
 Stability of the active ingredient in the dosage form
DILUTION TEST: •The emulsion is diluted with water. In case the emulsion remains stable after its dilution, it is o/w
emulsion. •The w/o emulsion breaks on its dilution with water but remains stable when diluted with oil. DYE TEST:
•The scarlet red dye is mixed with the emulsion. Place a drop of the emulsion on a microscopic slide, Cover it with a
cover- slip, and examine it under a microscope. If the disperse globules appear red and the ‘ground ’ colourless, the
emulsion is o/w type.
 Release of the active ingredient from the dosage form
 Storage conditions.
[https://www.pharmapproach.com/quality-control-requirements-pharmaceutical-dosage-forms/]

Liquids:
Evaluation for visual appearance, colour, taste, odour, labelling, and homogeneity,
Assay of active ingredients and of degradation products.
Pourability
Viscosity
Isotonicity
Particle size agglomeration and particle size distribution

P a g e | 87
Clarity
Crystallization and precipitation
Gas evolution
Relative density
pH
Surface tension
Microbial limit tests
Stability of the active ingredient(s), and identification tests
Light stability
Container and closure compatibility
Redispersibility
Suspensibility
Storage condition

[https://www.pharmapproach.com/quality-control-requirements-pharmaceutical-dosage-forms/]

Journal article format


- Formulation, In-process analysis & Discussion
See article attached (guide/template)
Experiments comprising of formulations, and quality evaluation will require reports written in an article format
Appendices:

A: Article - Emulsion
B: Article/Experiment – Suspension
C: Article - Quality Evaluation

P a g e | 88
Appendix A

P a g e | 89
P a g e | 90
P a g e | 91
Page |-1-
Page |-2-
Page |-3-
Appendix B
Preparation of various types suspensions and determination of their sedimentation parameters

BACKGROUND
Mg. Carbonate is an insoluble but diffusible solid. In flocculated suspension, the practical settle more
quickly than practical of deflocculated one. But the sediments from a cake like structure in deflocculated
suspension making dispersibility on shaking a big problem comparing these 2 types of suspension, the
flocculated one is pharmaceutically acceptable or preferable however, a control flocculation is desirable
to achieve a control sedimentation with ease of dispersibility.
Electrolyte and ionic surfactant can be used as flocculating agents to produce a flocculated suspension.
Suspension prepared without flocculating agent is a deflocculated one.
The physical stability of the suspension is assist by determining the sedimentation volume or degree of
flocculation.
Aim: To prepare a flocculated and deflocculated suspension of mg.carbonate and assess their stability.

REQUIREMENTS
Materials and apparatus:

o Mg. Carbonate,
o Al. chloride,
o Water,
o Pestle and mortar
o Measuring cylinder

PROCEDURE
Step-1: Preparation of suspension

Deflocculated suspension

1. The light magnesium carbonate is powered in a mortar.

2. The water is added with trituration to make a cream and diluted sufficiently.

3. Then it is transferred to a measuring cylinder. The mortar is repeatedly rinsed with little water every
time and the rinsed mixture is added subsequently to adjust the volume.

Flocculated suspension

1. The light magnesium carbonate is powered in mortar.


2. Aluminium chloride is dissolved in little water and added with trituration to make a cream. The
cream is transferred to the measuring cylinder after dilution of water.
3. The mortar is repeatedly rinsed with little amount of water every time and the rinsed mixture is
added subsequently to adjust the volume.

Page |-4-
Step-2 Evaluation of suspension

1. The suspension in the cylinders are thoroughly shaken to make the dispersions uniform.

2. The cylinders are kept undisturbed on a flat surface after shaking.

3. The volume of sediment at different time: 0,10,30,45, and 60 mins is measured.

Original volume of suspension is 100 ml


The degree of flocculation at one hr=sediment volume of flocculated suspension in one hour/sediment
volume of deflocculated volume in one hour. 3

(Though degree of flocculation is related to the ultimate sediment volume in flocculated and
deflocculated suspension, it is not feasible to achieve or determine ultimate volume with in the practical
period.)
The plot of sedimentation volume in Y axis and time in X axis.
(Sedimentation volume quickly decrease in flocculated suspension compared to deflocculated
suspension initially but ultimate sedimentation volume of flocculated will be higher)

Time in Sedimentation volume for Volume of sediment for Sedimentation


minutes flocculated suspension deflocculated suspension volume for
in ml deflocculated
suspension

0
10
30
45
60

CONCLUSION
The flocculated and deflocculated suspension is prepared and evaluated for sedimentation volume.

REFERENCES
1. More HN, Hajare AA. Practical Physical Pharmacy. Career Publications. 2010: 203-205.
2. Gaud RS, Gupta GD. Practical Physical Pharmacy. CBS Publisher and Distributors. 2009: 81-84
3. Mohanta GP. Physical Pharmacy Practical Text, Pharma Book Syndicate.
2006: 72-74.

Page |-5-
Appendix C:
Pharmacology & Pharmacy, 2014, 5, 49-60
Published Online January 2014 (http://www.scirp.org/journal/pp) http://dx.doi.org/10.4236/pp.2014.51009

The Quality and in Vitro Efficacy of


Amoxicillin/Clavulanic Acid Formulations in the Central
Region of Ghana
Henry Nettey, Grace Lovia Allotey-Babington, Philip Debrah, Ofosua Adi-Dako, Manal Shaick,
Isaac Kintoh, Francis Arnansi, Makafui Nyagblordzro, Marvin Holison

Department of Pharmaceutics and Microbiology, School of Pharmacy, University of Ghana, Legon, Ghana.
Email: hnettey@msn.com

Received November 11th, 2013; revised December 25th, 2013; accepted January 6th, 2014
Copyright © 2014 Henry Nettey et al. This is an open access article distributed under the Creative Commons Attribution
License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly
cited. In accordance of the Creative Commons Attribution License all Copyrights © 2014 are reserved for SCIRP and the owner
of the intellectual property Henry Nettey et al. All Copyright © 2014 are guarded by law and by SCIRP as a guardian.

ABSTRACT
Aim: To assess the quality and in vitro efficacy of five brands of amoxicillin/clavulanic acid tablet, suspension
and injectable preparations selected from pharmacies in the Central Region of Ghana. Method: Using a
Stratified Representation Sampling method, forty preparations (tablets, suspensions and injectable powders)
containing amoxicillin and clavulanic acid were sampled from nine different locations within the Central
Region of Ghana. To determine drug quality, several procedures, namely, content assay, disintegration and
dissolution testing were employed. In vitro drug efficacy was determined by comparing the Minimum
Inhibitory Concentrations (MIC’s) obtained with published values. Results: All tablets passed the
disintegration test, with disintegration time ranging between six (6) and fifteen (15) minutes. Analyses of all
the tablets for drug content showed 100% failure (14 out of 14) for amoxicillin and 14% failure (2 out of 14)
for clavulanic acid. Injectable formulations showed similar results. All four (4) samples analyzed for content
failed the amoxicillin content assay (0 out of 4) but all passed clavulanic acid assay (4 out of 4). For tablet
dissolution tests, there was a 93% (13 out of 14) pass rate for both amoxicillin and clavulanic acid. Content
analysis of all suspension formulations involved twenty-two (22) samples from five (5) brands. Only 41% (9
out of 22) passed for both amoxicillin and clavulanic acid. All the other samples failed for either amoxicillin,
clavulanic acid or both. Results obtained from drug quality tests were confirmed by in vitro efficacy tests
against selected microorganisms. Conclusion: The samples were therefore not of good quality, since content
assay is the most crucial test. It is hypothesized that this is due to poor storage conditions, and
recommendations, such as air conditioning and more structured procedures along the supply chain, are put
forward to counteract this.

KEYWORDS Amoxicillin; Clavulanic Acid; Minimum Inhibitory Concentration; Dissolution

Page |-6-
1. Introduction well. With the high demand for medication in
Ghana, and the high amount of revenue generated
Amoxicillin belongs to the class of medications called by the industry, counterfeit drugs have begun to
penicillin-like antibiotics. The penicillins belong to penetrate the Ghanaian market. In February 2013,
the beta-lactam group of antibiotics [1], which are the Food and Drugs Authority (FDA) in Ghana
the dominant class of agents currently used for the reported the importation of fake amoxicillin/cla-
chemotherapy of bacterial infections worldwide. vulanic acid tablets into Ghana [9]. Rox-Clav 625
Amoxicillin acts by inhibiting the synthesis of tablets available on the market were found to
bacterial cell walls [2]. It inhibits cross-linkage contain no clavulanic acid, thus making it
between the linear peptidoglycan polymer chains counterfeit. This is particularly dangerous because
that make up a major component of the cell wall of

Gram-positive bacteria [3]. It is usually the drug of without clavulanic acid, the product is highly
choice within the class because it is better absorbed inefficient.
following oral administration and it is resistant to
Unfortunately, administering counterfeit drugs
gastric acid [4]. Amoxicillin is susceptible to
could result in therapeutic failure, toxicity, allergic
degradation by beta-lactamase-producing bacteria,
reactions due to their content, drug resistance,
and so may be given with Clavulanic acid to decrease
prolonged illness, high cost of treatment and even
its susceptibility [5]. Clavulanic acid, a β-lactamase mortality. Furthermore, other secondary
inhibitor [6], is added to amoxicillin to inhibit β- implications are that the companies, pharmacies
lactamase and increase the antibacterial effect of and distributors involved may lose their credibility,
amoxicillin. It is however a less potent antibacterial and a lot of financial resources may be lost in
agent as compared to amoxicillin [6]. Clavulanic acid investigations and court cases. One should be aware
has a similar structure to the beta-lactam antibiotics that low quality medicines may not always be the
but binds irreversibly to the beta-lactamase result of problems at the manufacturing stage. They
enzymes [7]. Amoxicillin and Clavulanic acid could be because of problems with packaging,
combinations are available in oral solid dosage form, transportation, storage conditions and the
powder for reconstitution as suspension and distribution system.
injectable. These two drugs act synergistically to
Whatever the case, low quality medicines are
produce the desired therapeutic effect. Their
certainly not desirable on the market. Pharmacists,
potency depends on content of the active moiety in
regulatory officials and health inspectors have to be
these dosage forms. This combination is used as a
vigilant and perform quality assurance tests on
broad spectrum antibiotic for the treatment of a
medications available locally. This gives the public
wide range of bacterial infections, including upper
confidence in available medications, and helps to
and lower respiratory tract infections and infections
take substandard medications out of the system.
of the skin and soft tissue structures [5].
Increased quality assurance also prevents
One of the World Health Organization’s (WHO’s) counterfeit drugs from penetrating the market: for
main priorities is the quality of pharmaceuticals. fear that they will be detected.
Article 2 of the WHO Constitution states one of the
Quality assurance is generally conducted in the
organization’s functions as to “develop, establish
more prominent areas, such as the Greater Accra
and promote international standards with respect
Region in Ghana. This research project looks at
to food, biological, pharmaceutical and similar
amoxicillin/clavu- lanic acid formulations in the
products” [8].
Central Region of Ghana, and evaluates their quality
The quality of pharmaceuticals is of importance and efficacy. With the possibility of counterfeit
not just on the global scale, but within nations as drugs on the market, it is important that the

Page |-7-
pharmaceutical industry cultivates a culture of period of time when placed in a liquid medium
regular, quality assurance and efficient reporting under established experimental conditions. The end
pro- cesses. It is hoped that this research will of the test is detected when there is no residue of
contribute towards this practice. the tablet or capsule on the mesh of the test
apparatus aside from insoluble fragments of the
2. Materials and Methods tablet coating or the capsule gel [11]. The apparatus
consists of a two basket-rack arrangement of six
2.1. Sampling Method
open ended transparent tubes held upright by two
Five (5) brands of amoxicillin/clavulanic acid plastic plates with six holes equally spaced from
formulations available in nine (9) districts in the each other. A woven wire mesh made of stainless
Central region were purchased. The nine (9) steel having a square weave with holes is attached
districts were Kasoa (k), Agona Swedru (Ag), Apam to the lower plate and two 1000 ml beakers are
(Ap), Winneba (W), Mankessim (M), Elmina (E), placed in a rectangular shaped glass water tank. A
TwifuPraso (T), Assin Fosu (As) and Cape Coast (C). thermostat is used to control the temperature of
The five (5) brands were coded A, B, C, D and E. The the liquid medium between 35˚C and 39˚C, and a
samples were coded with the brand code first control knob for raising and lowering the basket
followed by the dosage form, the town of purchase, into the beaker of immersion fluid, distilled water,
and then a number subscript indicating the sample at a constant frequency rate ranging between 29 to
number. For example, if two tablet samples of the 39 cycles per minute.
brand A were purchased from two different
pharmacies in Kasoa, the codes will be AT-K1 and 2.3. Experimental Procedure
AT-K2. In purchasing the samples, a convenience 750 ml of distilled water was accurately measured
sampling method was used to select which districts using a 1000 ml measuring cylinder and transferred
to sample from. Convenience sampling involves into each of the 1000 ml beakers. Then the
drawing the sample from that part of the disintegration apparatus, QC-21 Disintegration Test
population which is close to hand. The central System (Hanson Research, California, USA), was
region is divided into seventeen districts. In calibrated to a temperature of
employing the method, the 9 districts were selected
because they had the highest number of 37˚C +/− 0.5˚C. To ensure that the temperature was
pharmacies/hospitals as recorded by the Pharmacy accurate, a glass thermometer was placed in each
Council of Ghana [10]. After selecting the 9 districts, beaker. Six tablets or capsules of each formulation
several pharmacies that were close at hand were of amoxicillin/clavulanic acid from a particular area
visited in the areas in order to find out which in the central region were individually weighed and
brands of amoxicillin/clavulanic acid were most each placed in a transparent tube of the first basket
popularly used. The 5 most popular brands, named rack. To the transparent tubes of the second basket
above, were thus selected. Then, tablets, powder rack, another set of six tablets/capsules was added.
for suspension and injectables of these brands were The two racks were gently immersed
purchased from more than one pharmacy in each simultaneously into the liquid medium and the run
district. In order to keep records and information started. This procedure was repeated for all
about each product, the pharmacist at each tablet/capsule samples obtained.
pharmacy from which the products were purchased
2.4. Amoxicillin and Clavulanic Acid Standard
filled a questionnaire to ensure easy traceability of
Preparation
the product.
All dissolution and HPLC analysis were done at the
2.2. Disintegration Test Food and Drug Authority (FDA) laboratories in
The disintegration test determines the ability of a Accra. Amoxicillin Trihydrate RS, lot number
tablet or capsule to disintegrate within a given P500127 and purity of 85.7% w/v was obtained

Page |-8-
from Sigma-Aldrich. Clavulanate Lithium USP, Lot injections are already in powder form, they cannot
number JOG109 and purity 95.7%, w/v was be weighed directly because of the risk of losing
obtained as a gift from the (FDA), Accra, Ghana. An some powder. Therefore each vial containing drug
approximate amount of 0.025 g of Amoxicillin powder was weighed. The vial was then filled with
trihydrate and 0.01 g of Clavulanate Lithium were distilled water and the contents transferred to a 100
weighed and transferred into a 50 ml volumetric ml volumetric flask. The vial was washed three
flask. Distilled water was then added up to the 50 times with distilled water and transferred to the
ml mark to produce a final concentration of 0.05% volumetric flask. The flask was finally made to
w/v or 0.5 mg/ml of amoxicillin trihydrate and volume with distilled water. The empty vial was
0.02% w/v or 0.2 mg/ml of clavulanate lithium. 20 dried, weighed, and the weight of the weight of the
µl volumes of both standards and samples were drug obtained by difference.
injected into High Performance Liquid
The volumetric flask was then placed in a Clifton
Chromatography (HPLC). An Agilent 1100 system
ultrasonic bath for 3 minutes for uniform mixing.
was used and conditions for all HPLC analysis were
Similarly, 5 ml of the resulting solution was drawn
as follows: Mobile Phase—5 volumes of methanol,
using a pipette, placed in a 50 ml volumetric flask,
95 volumes of 0.78% w/v Sodium dihydrogen
and topped up to the 50 ml mark with distilled
phosphate monohydride (pH 4.4); Flow rate of 2
water to obtain a final concentration of 0.05% w/v
ml/min using isocratic elution; 150 × 4.6 mm C18
of amoxicillin trihydrate and 0.02% w/v of
Supelco Column. Detection wavelength—220 nm.
potassium clavulanate. The 50 ml volumetric flask
Calculations were used to convert the amount of
was manually shaken for a few minutes and some
Potassium Clavulanate to Clavulanic acid and
drug solution drawn and filtered into an HPLC vial.
Amoxicillin Trihydrate to amoxicillin.
Two injections were made per sample.
2.5. Content Analysis For analysis of suspension formulations, the
The content of amoxicillin and clavulanic acid in samples were analyzed based on British
each formulation was determined using HPLC. For Pharmacopoeia (BP) requirements. Twenty-two (22)
each tablet pack three tablets were separately samples from five (5) brands were analyzed in all.
weighed and each placed in a 100 ml volumetric Reconstitution of the powder for suspension was
flask. 50 ml of distilled water was measured into made according to the manufacturer’s
each of the flasks, which was then placed in a specifications. The suspensions were further diluted
mechanical shaker for ten minutes to ensure that with distilled water to a final concentration of 0.05%
the tablet had completely disintegrated. Each of the w/v of amoxicillin trihydrate and 0.02% w/v of
flasks was thereafter made to volume with distilled potassium clavulanate. The amounts of clavulanic
water and placed in a Clifton ultrasonic bath for 3 acid and amoxicillin were calculated from potassium
minutes to obtain a uniformly mixed solution. 5 ml clavulanate and amoxicillin trihydrate respectively.
of the solution was then withdrawn, transferred
into a 50 ml volumetric flask and made to volume 2.6. Dissolution Test
with distilled water to give an expected final Two dissolution station apparatus: ERWEKA DT-600
concentration of 0.05% w/v of amoxicillin trihydrate and DT-800 were calibrated in the paddle mode and
and 0.02% w/v of potassium clavulanate. The 50 ml set to a temperature of 37˚C +/− 0.5˚C, time
volumetric flask was manually shaken for a few duration of 30 minutes and a frequency of 75
minutes and some drug solution drawn and filtered rotations per minute (rpm). A set volume of 900 ml
into an HPLC vial using a 0.45 µm filter. Two of dissolution medium, distilled water, was
injections were made per sample. accurately measured using a 1000 ml measuring
cylinder and poured into each of the six glass
The procedure for the injection formulations was
vessels and maintained at a temperature of 37˚C
similar to that for the tablets, however because the
Page |-9-
+/− 0.5˚C. Standard thermometers were placed in hours. Zones of inhibition (ZOI) were measured as
each vessel to crosscheck the temperature. Three the diameter of clear area of no growth around the
tablets of brand A, from one location were placed edges of the wells. The above procedure was
into three vessels and another three tablets of the repeated for the next three concentrations of the
same brand from a different area were placed into same sample and for the other samples. Plots of
the other three vessels. This was repeated for all drug concentration versus ZOI were made and the
the brands available, hence, six tablets of the same minimum inhibitory concentration (MIC) was
brand were placed into the buffer/dissolution obtained as the intercept on the concentration axis,
medium (distilled water) all at the same time. At the of the line of best fit. The MIC values obtained,
end of the run time, 10 ml was sampled from each though directly correlate with the susceptibility of
vessel into a labeled beaker with a calibrated an organism to the drug, are interpreted here to
syringe for further analysis. The drug solutions were also correspond to the concentration on the drug in
allowed to equilibrate to room temperature and solution. The identity of the drug molecule and the
portions filtered and transferred into agilent HPLC concentration in the various formulations has been
vials. Two injections were made per sample. established by in vitro content and dissolution tests.
Sample MIC values were compared with published
2.7. Antimicrobial Effect of MIC values for the various organisms [12]. A
Amoxicillin/Clavulanic Acid Formulations significantly lower sample MIC value is an indication
Bacillus subtilis and Klebsiellaspp were uncoded of low drug concentration.
clinical isolates obtained from the Korle-bu
Teaching Hospital in Accra, Ghana. Salmonella typhi 2.8. Statistical Analysis
(ATCC 33458), Escherichia coli (NCTC 1351) and The statistical tool used was the independent
Staphylococcus aureus (NCTC 6571) were gifts from samples T-test.
the Noguchi Memorial Institute, Legon, Ghana.
Stock suspensions of the antibiotics were prepared 3. Results
and used to make concentrations of 0.001%,
3.1. Disintegration Test
0.002%, 0.004% and 0.008% w/v in sterile water.
Nutrient agar was prepared and 25 ml portions The results of the disintegration test are presented
were transferred into test tubes. The test tubes in Figure 1. All the samples passed the B.P
were then sterilized in an autoclave for 15 minutes specification for disintegration test [13]. The highest
at 121˚C and stabilized in a water bath at 45˚C to disintegration time recorded was 14.11 minutes for
prevent the molten nutrient agar from solidifying. DT-E with the least being 6.43 minutes for ET-C. All
the other samples had variable disintegration times
Stabilized agar in test tubes was seeded with 1 ml but all fell within the specified time range. From the
each of a 24 hour broth cultured bacteria (1.2 × 106 results obtained it can be inferred that all the
cfu/ml). The bacteria used were B. subtilis, tablets will be able to readily release the active
Klebsiella sp, Salmonella sp, E. coli, and S. aureus. ingredients in aqueous medium or in
Seeded molten agar was aseptically transferred into gastrointestinal fluid.
petri dishes and allowed to set at room
temperature. Using the 10 mm diameter cork borer, 3.2. Content Assay and Dissolution Test
4 holes were made in the agar with reasonable In order to verify if the HPLC equipment was good
spacing. enough for carrying out experiments, the system
Three of the holes were each filled with the 0.1 suitability of the equipment was determined by
ml of antibiotic suspension and the fourth with calculating the relative standard deviation. Relative
sterile water. The plates were covered and left on standard deviations below 2% are acceptable [13]
the bench for an hour after which they were for the dissolution and content assay of the tablets
incubated in an upright position at 37˚C for 24 and injectables. The relative standard deviation for
P a g e | - 10 -
16
14
12
10
8
6
4
2
0
AT-C AT-K AT-S AT-W BT-C BT-K BT-S BT-As DT-K DT-E CT-C CT-K ET-C
ET-M
Code

Mean Disintegration time (min)

Figure 1. Disintegration time of samples.

Brand A
120

100

80

60

40

20

0
AS-C₁ AS-Ag AS-W AS-T AS-C₂ AS-E AS-K
AMOX 79.03 85.83 61.3 60.11 76.88 55.96 84.86
CLAV 90.37 80.85 78.92 99.04 99.28 81.06 76.17

Figure 2. Percentage of amoxicillin/clavulanic acid content of brand A suspensions.

both amoxicillin and clavulanic acid was below 2% Content analysis was performed for all the five (5)
in either the dissolution or content system brands of amoxicillin/clavulanic acid suspension
suitability test. obtained. Only one (1) out of seven (7) samples
(14.3%) of Brands A or B passed assay (Figures 2
According to the BP, the acceptable content
and 3). The two samples that passed were
range of a newly reconstituted suspension should
purchased from Agona Swedru and Cape Coast
be between 80% and 120% of the label claim when
respectively. Four (4) out of five (5) samples (80%)
stored at the appropriate temperature and period
stated for use. A sample has to pass for both
amoxicillin and clavulanic acid in order to be of Brand C passed (Figure 4). All (100%) of brands D
considered as having passed content analysis. and E passed (Figures 5 and 6). A summary of the

P a g e | - 11 -
results from the analysis of the suspension dosage (2) brands of injectables obtained from four (4)
forms showed that only nine (9) of the twenty-two pharmacies in three (3) districts. The acceptance
(22) samples passed contentanalysis (41%). A criteria for the assay of amoxicillin/clavulanic
statistically significance difference was observed tablets and injectables are between 90% and 120%
when the content of clavulanic acid was compared and each tablet should pass for both ingredients
in all suspension samples (p < 0.01); however [13]. All the tablets failed content analysis for
differences in amoxicillin content was not (p > 0.01) amoxicillin and twelve (12) out of fourteen (14)
(Table 1). tablet batches (86%) passed for clavulanic acid
alone (Figure 7). A statistically significance
Content assay was doneon all five (5) brands of
difference was observed when the content of
amo- xicillin/clavulanic acid tablets obtained from
amoxicillin was compared in all tablet samples (p <
fourteen (14) different pharmacies in seven (7)
0.01); however differences in clavulanic acid
districts as well as two
contentwas not (p > 0.01) (Table 1). Analysis of the
injectables showed all samples passed for

Brand B
140

120

100

80

60

40

20 Figure 3.
0

BS-W BS-Ap BS-T BS-C₁ BS-K₁ BS-K₂ BS-C₂

AMOX 69.29 62.06 70.68 73.18 77.14 73.68 83.31

CLAV 101.13 91.61 104.55 103.15 112.65 106.2 117.24


Percentage of amoxicillin/clavulanic acid content of brand B suspensions.

P a g e | - 12 -
Brand C
95

Figure 4.

90

85

80

75

CS-M CS-T CS-C₁ CS-K CS-C₂


70
AMOX 79.56 92.24 86.06 85.34 88.73
CLAV 78.59 93.55 89.42 88.48 91.43
Percentage of amoxicillin/clavulanic acid content of brand C suspensions.

Brand D
87
86
85
84
83
82
81
80
79
DS-K₁ DS-K₂
AMOX 83.68 82.04
CLAV 86.28 84.64

Figure 5. Percentage of amoxicillin/clavulanic acid content of brand D suspensions.

P a g e | - 13 -
Brand E
86

85

84

83

82

81

80

79
AMOX CLAV
ES-K 85.28 81.69

Figure 6. Percentage of amoxicillin/clavulanic acid contentof brand E suspension.

Table 1. One-sample test of amoxicillin/clavulanic acid formulations. Significance = 0.01. Test values are the lower limits of
reference concentration ranges.
N Mean Std. deviation t df p

Test value = 80

Suspension Amoxicillin 22 77.1018 10.17178 −1.336 21 0.196

Clavulanic acid 22 92.5591 11.52484 5.111 21 0.000

Test value = 90

Tablet Content Amoxicillin 14 75.585 5.71205 −9.442 13 0.000

Clavulanic acid 14 91.54 18.09205 0.318 13 0.755

Test value = 75

Tablet Dissolution Amoxicillin 14 79.8886 21.1786 0.864 13 0.403

Clavulanic acid 14 84.5981 23.56951 1.524 13 0.152

P a g e | - 14 -
120

100

80

60

40

20

0
AT-C AT-K
AT-S AT-W
BT-C BT-K BT-S BT-As
DT-K DT-E CT-C CT-K
Code ET-C ET-M
Figure 7. Percentage of amoxicillin/clavulanic acid content in tablet samples after content assay. The solid horizontal line
represents the lower acceptable limit for drug content per BP.
than 75% of the labeled amount of either
clavulanic acid but failed for amoxicillin content amoxicillin and clavulanic acid. Thirteen (13) of all
(Figure 8). Since no tablet or injectable passed both tablet batches tested (93%) passed the test (Figure
amoxicillin and clavulanic acid content per B.P 9). One batch (DT-E) failed for both amoxicillin and
criteria, there was 100% failure. A study by clavulanic acid, showing only an average of 3.36%
Sengaloundeth et al. in 2009 found a number of and 7.30% respectively. Statistical analysis of all
fake and substandard oral artesunate dosage forms tablet samples for dissolution showed that the
in Laos [14]. Later on in 2011, some of the authors difference among them was not significant (p >
in the Laos drug quality study thought it necessary 0.01) (Table 1).
to expand the scope of their investigations. They
determined in their search that most of the 3.3. Anti-Microbial Effect of
counterfeit and substandard anti-malarials found in Amoxicillin/Clavulanic Acid Samples
Africa have their origins in South East Asia. In that A subset of the tablet, injectable and suspension
study, they found numerous fake and substandard formulations of amoxicillin/clavulanic samples
anti-malarials in most of the African countries they obtained were used for in vitro anti-microbial
sampled from, including Ghana, They however, efficacy studies. Not all the samples obtained
stated a major limitation in their study as the lack of were tested because most had been used up in
dissolution studies [15]. This prompted the previous tests. A low MIC is highly significant and
researchers in the present study to include tablet indicative of a potent product which has an
dissolution studies as well as in vitro efficacy effective therapeutic outcome. The result of the
studied since we were dealing with antibiotics. Prior antimicrobial susceptibility test showed that all
to this study, a pilot study was done by one of the samples tested had significant inhibitory effect
researchers in Accra, a major city in Ghana. In that against B. subtilis except for samples AS-C1 and
study, some substandard Artemether-Lumefantrine DT-E which showed very minimal activity (Table
tablets were found in that region [16]. 2). The means of all samples, when

Dissolution test was done on all the tablets used


for drug content analysis. The B.P 2013 stipulates
that at 30 minutes, all tablets should have released
into the dissolution medium an amount not less

P a g e | - 15 -
100
90
100
9080
8070
70
6060
5050
40
3040
20
30
10
020
10 AT-C AT-K
AT-S AT-W
BT-C BT-K
0 BT-S BT-As
DT-K DT-E CT-C CT-K
AI-C Code ET-C ET-M
BI-C
BI-K
BI-Ap
Code

Figure 8. Percentage of amoxicillin/clavulanic acid content in injectable samples after content assay. The solid horizontal line
represents the lower acceptable limit for drug content per BP.

AI-C BI-C BI-K BI-Ap

Amoxycillin 76.45 83.35 84.2 84.56

Clavulanic 92.95 96.08 95.62 95.9

AT-C AT-K AT-S AT-W BT-C BT-K BT-S BT-As DT-K DT-E CT-C CT-K ET-C ET-M

Amoxycillin 83.49 80.61 84.29 85.93 83.35 84.44 85.26 83.25 81.78 7.3 90.72 93.86 86.86 87.3

Clavulanic Acid 89.073 87.3 89.02 91.25 93.31 91.7 92.2 91.63 83.57 3.36 91.81 96.23 92.55 91.37

Figure 9. Percentage of amoxicillin/clavulanic acid content in tablet samples after dissolution testing. The solid horizontal line
represents the lower acceptable limit for drug content per
BP.
Salmonella spand E. coliexcept DT-E which was
compared to the reference concentration of 4 least effective (Table 2). The mean differences
µg/ml showed differences that were statistically between the samples and reference were sta-
significant (p < 0.05) (Table 3). All samples tested tistically significant (p < 0.05) (Table 3).
showed inhibitory effect against Klebsiella sp, Staphylococcus aureus, with a reference
concentration of 4 µg/ml for susceptibility,

P a g e | - 16 -
showed the most resistance to the samples tested the bioavailability of drug because it is directly
(Table 2). The mean differences between the proportionalto the rate of dissolution of the
samples and reference were not statistically dosage form. The BP stipulates a disintegration
significant (p > 0.05) (Table 3). Also analysis time of not more than 15 minutes for uncoated
revealed relatively higher MIC values for sample tablets [13]. All the tablet formulations passed the
ax-std, which was amoxicillin powder and used as disintegration test and as such, it will be expected
a standard. This is because sample ax-std, had no that the active ingredient will be released in time
clavulanic acid and was subject to degradation by for dissolution to start in the stomach.
β-lactamase produce by gram positive bacteria. Assay of content of both tablet and injectable
formulations produced rather disappointing results.
4. Discussions
All the samples tested failed to meet B.P
Tablet disintegration is the first step for a drug to specifications for drug assay. The samples failed,
become bioavailable. A tablet must first not because they did not have the active
disintegrate and release the drug molecule to the ingredients, but rather the assayed amounts fell
gastrointestinal fluids. The active ingredient in the short of the label claim. All the samples had at least
disintegrated tablet must dissolve in 60% of either amoxicillin or clavulanic acid except
gastrointestinal fluid to be absorbed into the blood for sample DT-E which had clavulanic acid content
stream. Tablets should thus be sufficiently hard to of 30%. A host of factors may affect the content of a
withstand handling without crumbling or breaking drug product. Factors that may affect the quality of
but they should also be sufficiently soft for easy manufactured drug products include storage
disintegration in the stomach or intestine in order temperature conditions in which drug products are
to make the drug available to the body. Tablets stored, level of moisture at these storage sites
may be too hard and fail the disintegration test (humidity), exposure to light, microbial
because of poor manufacturing process or contamination, packaging materials used for the
incorrect storage. The disintegration test thus drug product, transportation factors associated
measures the time taken for tablets to disintegrate with the distribution of the drug product,
into particles. This is a necessary condition for drug components of drug composition and most
dissolution and could be the rate determining step importantly, the nature of the active ingredient
in the process of drug absorption. Tablet used
disintegration is thus a factor of consideration in

P a g e | - 17 -
Table 2. In vitro efficacy of amoxicillin/clavulanic acid tablets, Suspensions and injectables. Interpretation of MIC values for
Klebsiella sp, Salmonella sp, and E. coli are: ≤4 - 8 µg/ml = Susceptible (S); 9 - 16 µg/ml = intermediate (I); ≥17 - 32 µg/ml =
Resistant (R). MIC values for B. subtilis and S. aureus are: ≤2 - 4 µg/ml = Susceptible (S); ≥5 - 8 µg/ml = Resistant (R).
Organism name/sample concentration (µg/ml)
Sample code
B. subtilis Klebsiellasp Salmonella sp E.coli Staph. aureus
Ax std 4.11 0.00292 18.6
AT-K 2.78 0.98 2.98 2.61 2.85
AS-C1 8.4 2.35 3.76 0.292 9.19
AS-C2 0.0186 0.00104 0.0607 1.90 0.191
AS-Ag 0.93 0.53 2.15 0.91 1.39
AS-T 1.48 1.72 0.0057 0.43 3.01
AS-W 1.34 5.60 1.74 2.98 4.46
BT-C 1.05 1.48 0.048 2.64 9.22*10−10
BT-As 2.94 0.86 0.78 0.29 3.17
BT-S 2.73 0.065 1.73 4.05 1.31
BS-C 1.12 3.36 1.70 5.48 5.26
BS-K1 0.28 0.047 2.86 1.66 2.60
BS-K2 3.30 2.28 0.23 3.64 1.76
BS-W 2.6 1.65 0.00369 0.00375 3.01
BI-C 0.38 1.66 2.91 0.61 0.039
BI-K 1.33 0.52 1.20 4.05 1.31
CT-C 2.24 2.26 0.000497 1.51 0.716
CT-K 3.09 1.70 2.18 1.13 3.90
CS-C1 1.01 4.60 3.34 2.69 8.86
CS-C2 1.99 0.15 1.26 1.38 5.72
CS-M 1.05 2.45 1.43 2.65 8.11
CS-T 1.99 0.15 1.26 1.24 2.80
DT-C 1.9 2.01 0.625 1.86 0.00363
DT-E 13.45 14.12 14.12 6.14 14.51
DT-K 3.21 1.82 0.77 3.06 3.14
DS-K1 2.37 1.63 2.51 4.76 7.81
DS-K2 0.87 0.82 2.19 2.13 0.29
ET-C 1.18 1.81 1.99 2.83 4.01
ET–M 0.29 0.00032 1.39 0.699 0.399
ES-K 2.21 2.07 0.024 2.41 1.72
ES–K1 1.89 0.58 1.30 2.24 3.98
ES–K2 3.10 1.47 1.70 1.48 3.14

P a g e | - 18 -
Table 3. One-sample test of antimicrobial susceptibility to antibiotics. Test values are the lower limits of reference
susceptibility concentrations.
N Mean Std. deviation t df Sig. (1-tailed)

Test value = 4

B. subtilis 32 2.3946 2.53981 −3.576 31 0.001


S. aureus 32 3.9768 4.21354 −0.031 31 0.975
Test value = 8

Klebsiella sp 31 1.9595 2.5984 −12.943 30 0.000


Salmonella sp 31 1.879 2.50943 −13.581 30 0.000
E. coli 32 2.17993 1.55635 −21.154 31 0.000

for the formulation. All these factors eventually affect the drug product’s stability. Drug Stability refers to the
capacity of a drug substance or product to remain within established specifications of identity, strength,
quality, and purity in a specified period of time [17]. The most plausible explanation for the failure of the
samples is the poor storage conditions that these drugs might have been subjected to. Most of the drugs are
kept in trucks and vans under the scorching sun and without refrigeration for days. Content analysis of the
suspension dosage forms showed only 41% passed and this could also be attributed to poor storage and/or
manufacturing conditions. Dissolution of a drug is prerequisite for absorption of the drug into the body and
the dissolution rate is directly related to the bioavailability. For this reason, dissolution testing is commonly
used as a tool to compare the bioavailability of the same drugs manufactured by different companies.
Dissolution is the primary quality control test to determine whether a drug product can release its active
pharmaceutical ingredient(s) in a timely manner [18]. It is thus required for all solid oral dosage forms in
which absorption of the drug is necessary for the product to exert the desired therapeutic effect. If the rate
of dissolution is the rate-limiting step, then the dissolution rate determines the bioavailability. Dissolution is
thus used as a quality control measure for batch release, to ensure continued quality during the shelf life and
also to detect batches with poor quality without rejecting batches of adequate quality [19]. All the brands
studied passed for dissolution with the exception of sample DT-E. Sample DT-E contained an average of 3.36%
w/v of amoxicillin and 7.30% w/v of clavulanic acid. This may be attributed to factors that may have
influenced the dissolution of the active pharmaceutical ingredient (API) from the dosage form which may
include the nature of the excipients used in compounding or the formulation process. Although sample DT-E
passed disintegration test, it registered the longest time, and this could have influenced the rate of
dissolution. DT-E also produced the lowest API content among all samples analyzed which definitely affects
the results of the dissolution test. It is worthwhile to note that although all the tablet dosage forms analyzed
failed content analyses, 93% passed dissolution assay. A possible explanation for this observation is the less
stringent lower limit (75%) for a tablet to pass the dissolution test. One can safely infer from the results that
the drug products that passed the dissolution test may produce therapeutic levels of API. This will not always
be the case if API degradation, due to poor storage conditions, is the cause of the failed drug content. With
time, the API levels will reduce to such a level that dissolution tests will fail. Since dissolution tests are not
done for suspensions, we can safely conclude that those that failed content analysis will have reduced
bioavailability. In vitro efficacy tests confirmed results obtained for content analysis and dissolution tests.
Samples that generally showed lower content showed higher MIC values, hence will be less efficacious.

P a g e | - 19 -
5. Conclusion
All the samples analyzed for drug content failed to meet compendia requirements and are therefore
considered of poor quality. This conclusion can be drawn even though some of them passed
disintegration, dissolution or in vitro microbial growth inhibitory tests. Most of the brands had more than
sixty percent of either amoxicillin or clavulanic acid content and would likely be substandard rather than
counterfeit drugs. It is therefore necessary to enforce strict regulatory laws that will control the quality
of drugs from the time they are manufactured until they get to the consumer. Stability tests required for
the zone in which the drug would be marketed should be ensured. Random sampling of drugs on the
market should also be stepped up.

Acknowledgements
We wish to express our sincere thanks to the directors and technicians at the Ghana Food and Drugs
Authority for their immense assistance in our HPLC analysis and Dissolution tests.

REFERENCES
[1] M. E. Arroliga and L. Pien, “Penicillin Allergy: Consider Trying Penicillin Again,” Cleveland Clinic Journal of Medicine, Vol.
70, No. 4, 2003, pp. 313-314.
http://dx.doi.org/10.3949/ccjm.70.4.313
[2] C. Walsh, “Molecular Mechanisms That Confer Antibacterial Drug Resistance,” Nature, Vol. 406, No. 6797,
2000, pp. 775-781. http://dx.doi.org/10.1038/35021219
[3] A. Ashnagar and N. G. Naseri, “Analysis of Three Penicillin Antibiotics (Ampicillin, Amoxicillin and Cloxacillin) of Several
Iranian Pharmaceutical Companies by HPLC,” Journal of Chemistry, Vol. 4, No. 4, 2007, pp. 536-545.
[4] Å. Sullivan, C. Edlund and C. E. Nord, “Effect of Antim- icrobial Agents on the Ecological Balance of Human Microflora,” The
Lancet Infectious Diseases, Vol. 1, No. 2, 2001, pp. 101-114. http://dx.doi.org/10.1016/S1473-3099(01)00066-4
[5] C. M. Kaye, A. Allen, S. Perry, M. McDonagh, M. Davy, K. Storm, N. Bird and O. Dewit, “The Clinical Pharma- cokinetics of a
New Pharmacokinetically Enhanced For- mulation of Amoxicillin/Clavulanate,” Clinical Thera- peutics, Vol. 23, No. 4, 2001,
pp. 578-584.
http://dx.doi.org/10.1016/S0149-2918(01)80061-8

[6] S. M. Drawz and R. A. Bonomo, “Three Decades of β- Lactamase Inhibitors,” Clinical Microbiology Reviews,
Vol. 23, No. 1, 2010, pp. 160-201.
http://dx.doi.org/10.1128/CMR.00037-09
[7] M. Coll, J. Frau, B. Vilanova, J. Donoso, F. Munoz and F. G. Blanco, “Theoretical Study of the Alkaline Hydrolysis of a Bicyclic
Aza-β-Lactam,” The Journal of Physical Chemistry B, Vol. 104, No. 47, 2000, pp. 11389-11394.
http://dx.doi.org/10.1021/jp001989e
[8] World Health Organisation (WHO), “Quality Assurance of Pharmaceuticals: A Compendium of Guidelines and Related
Materials,” Vol. 2, Geneva, 2004.
[9] S. K. Opuni, “Alert: Osons Chemists, Roxin Ghana Import Fake Medicines from Ghana,” My Joy Online, 2013.
http://www.modernghana.com/news/445411/1/alert-osons
-chemists-roxin-ghana-import-fake-medic.html
[10] S. Ayim, “List of Hospitals and Pharmacies in the Central Region of Ghana as recorded by the Regional Health Directorate,”
Personal Interview, 2012.
[11] World Health Organisation (WHO), “The International Pharmacopoeia,” 4th Edition, 2nd Supplement, Geneva, 2008.
[12] Clinical and Laboratory Standards Institute, “Performance Standards for Antimicrobial Disk Susceptibility Test; Ap- proved
Standard,” CLSI Document M02-A10, 10th ed. 2009.
[13] British Pharmacopoeia Commission, “British Pharmaco- poeia 2013,” Renouf Publishing Company Limited, UK, 2013.

P a g e | - 20 -
[14] S. Sengaloundeth, M. D Green, F. M. Fernández, Ot. Manolin, K. Phommavong, V. Insixiengmay, C. Y. Hamp- ton, L. Nyadong,
D. C. Mildenhall, D. Hostetler, L. Khoun- saknalath, L. Vongsack, S. Phompida, V. Vanisaveth, L. Syhakhang and P. N. Newton,
“A Stratified Random Survey of the Proportion of Poor Quality Oral Artesunate
Sold at Medicine Outlets in the Lao PDR—Implications

for Therapeutic Failure and Drug Resistance,” Malaria Journal, Vol. 8, 2009, p. 172. http://dx.doi.org/10.1186/1475-
2875-8-172
[15] P. N. Newton, M. D. Green, D. C. Mildenhall, A. Plançon, H. Nettey, L. Nyadong, D. M. Hostetler, I. Swamidoss, G. A. Harris,
K. Powell, A. E. Timmermans, A. A. Amin, S. K. Opuni, S. Barbereau, C. Faurant, R. C. W. Soong, K. Faure, J. Thevanayagam,
P. Fernandes, H. Kaur, B. Angus, K. Stepniewska, P. J. Guerin and F. M. Fernández, “Poor Quality Vital Anti-Malarials in
Africa—An Urgent Neglected Public Health Priority,” Malaria Journal, Vol.
10, 2011, p. 352. http://dx.doi.org/10.1186/1475-2875-10-352
[16] E. A. Nyarko and H. Nettey, “Quality Assessment of Artemether/Lumefantrine Tablets Sampled from Pharma- cies in Accra,
Using the MVHimagePCv8.exe Color Soft- ware,” Pharmacology & Pharmacy, Vol. 4, 2013, pp. 567-572.
http://dx.doi.org/10.4236/pp.2013.48081
[17] J. Feng, X. Zhana, S. Qiaob, C. Wub and L. Xiaob, “A Mathematical Model for Calculating the Shelf Life of Ascorbic Acid
Solution under Given Conditions,” Drug Development and Industrial Pharmacy, Vol. 38, No. 3, 2012, pp. 264-270.
http://dx.doi.org/10.3109/03639045.2011.598537
[18] D. Q. Craig, “The Mechanisms of Drug Release from Solid Dispersions in Water-Soluble Polymers,” International Journal of
Pharmaceutics, Vol. 231, No. 2, 2002, pp. 131-144. http://dx.doi.org/10.1016/S0378-5173(01)00891-2
[19] S. A. Qureshi and J. Shabnam, “Cause of High Variability in Drug Dissolution Testing and Its Impact on Setting To- lerances,”
European Journal of Pharmaceutical Sciences, Vol. 12, No. 3, 2001, pp. 271-276. http://dx.doi.org/10.1016/S0928-
0987(00)00174-3

P a g e | - 21 -
P a g e | - 22 -
Additional Notes by Student:

P a g e | - 23 -
REFERENCES
▪ Bubna A. K. (2015). Aspirin in dermatology: Revisited. Indian dermatology online
journal, 6(6), 428-35
▪ R. Langer and N. A. Peppas. Advances in biomaterials, drug delivery, and
bionanotechnology. AIChE Journal, 49(12):2990–3006, 2003.
▪ Pharmaceutical Manufacturing Handbook Production and Processes, Wiley (2008)
▪ More HN, Hajare AA. Practical Physical Pharmacy. Career Publications. 2010: 203-
205. 1. Gaud RS, Gupta GD. Practical Physical Pharmacy. CBS Publisher and
Distributors. 2009: 81-84 2. Mohanta GP. Physical Pharmacy Practical Text, Pharma
Book Syndicate. 2006: 72-74.
▪ Abdo, R. W., Saadi, N., Hijazi, N. I., & Suleiman, Y. A. (2020). Quality control and
testing evaluation of pharmaceutical aerosols. Drug Delivery Systems, 579–614.
https://doi.org/10.1016/B978-0-12-814487-9.00012-0

1 https://www.beei.com/blog/pharmaceutical-manufacturing-equipment
2 https://ispe.org/initiatives/regulatory-resources/gmp/what-is-gmp
3 https://gmpnews.net/glossary/glp/
4 https://haiweb.org/encyclopaedia/pharmaceutical-research-and-development/
5 https://www.investopedia.com/articles/fundamental-analysis/11/primer-on-biotech-sector.asp
6 https://www.sciencedirect.com/topics/engineering/drug-delivery

P a g e | - 24 -
7 http://csip.cornell.edu/Curriculum_Resources/CSIP/Neeves/KNeeves_studentversion.pdf
8 https://www.pharmacytimes.com/publications/career/2014/pharmacycareers_november2014/so
uthwestern-oklahoma-state-university-college-of-pharamcy
9 https://www.pharmapproach.com/tabletmanufacturewetgranulationmethod/
10 https://drugs-bd.blogspot.com/
11 https://www.pharmaguideline.com/2013/11/types-and-size-of-capsules.html
12 https://pharmaceutical.basf.com/en/Drug-Formulation/Ointments.html
13 https://phclub15.files.wordpress.com/2017/01/ointment.pdf
14 https://www.netdoctor.co.uk/medicines/aches-pains/a8282/nurofen-gel-ibuprofen/
15 http://pharmacypractice1.blogspot.com/2015/03/pharmaceutical-solutions.html
16 https://www.pharmapproach.com/quality-control-requirements-pharmaceutical-dosage-forms/
17 https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7155167/
18 https://www.pharmapproach.com/quality-control-requirements-pharmaceutical-dosage-forms/
19 https://www.pharmapproach.com/quality-control-requirements-pharmaceutical-dosage-forms/

P a g e | - 25 -

You might also like