You are on page 1of 46

Basic Principles of

Sterile Product
Formulation
Development
Maria Elvina Tresia Butar-Butar, M.Farm.
INTRODUCTION
 Development of sterile dosage forms not only includes the formulation but also the package and the
process. Glass, rubber, and plastic chemistry are covered to some extent, as well as packaging delivery
systems and devices, both traditional (e. g., vials, syringes) and more novel (e. g. needleless injectors, dual
chambered systems).
 The area of manufacturing includes chapters on process development and overview, contamination control,
facilities, water, air, personnel practices, preparation of components, sterilization, filtration, filling,
stoppering and sealing, lyophilization, aseptic processing, barrier technology, labeling and secondary
packaging, and some discussion of manufacturing advances.
 The area of quality and regulatory includes chapters on good manufacturing practice, the philosophy of
quality as it relates to the sterile dosage form, specific quality control tests unique to sterile products, and
some coverage of stability testing.
INTRODUCTION
 Sterile dosage forms have always been an important class of pharmaceutical products in disease diagnosis,
therapy, and nutrition. Certain pharmaceutical agents, particularly peptides, proteins, and many
chemotherapeutic agents, can be administered only by injection (with or without a needle), because they
are inactivated in the gastrointestinal tract when given by mouth.
 Administration of drugs by the parenteral (parenteral and injectable will be used interchangeably) route has
skyrocketed over the past several years and will continue to do so. A primary explanation for this enormous
growth lies with the advent of biotechnology, the products of which are biomolecules that cannot be readily
administered by any other route because of bioavailability and stability reasons. Since human insulin
became the first biotechnology drug approved by the Food and Drug Administration (FDA) in 1982, over
100 drug products of biotechnological origin have been approved and hundreds more will be approved in
the years ahead.
INTRODUCTION

 Most biotechnology drug products are administered only by the parenteral route. Science is advancing to
a time when it is likely that some of these drugs can or will be administered by other routes, primarily
pulmonary and perhaps someday even orally, but the mainstay route of administration for these
biopharmaceutical drugs will be by injection.
ARY of THE HISTORY of STERILE DRUG TECHNO
The drug injected was opium. While the poor human
receiving this injection may have had his pain
alleviated, he likely was going to die, eventually
from microbial and pyrogenic contamination
introduced using this crude means of injection.
Other drugs injected into humans during those early
days were jalap resins, arsenic, snail water, and
purging agents. It is improbable that the initial
pioneers of injectable therapy had much
appreciation about the needs for cleanliness and
purity when injecting these medications. After 1662,
injecting drug solutions into humans was not
commonly practiced until late in the 18th century.
Y of THE HISTORY of STERILE DRUG TECHNOLO

Intravenous (IV) therapy was first applied around 1831 when cholera was treated by the IV injection of a
solution containing sodium chloride and sodium bicarbonate in water. Normal saline was used by Thom Latt’s
to treat diarrhea in cholera patients using intravenous infusions. Intravenous feeding was first tried in 1843,
when Claude Bernard used sugar solutions, milk, and egg whites to feed animals. By the end of the 19th
century, the intravenous route of administration was a widely accepted practice. Injections of emulsified fat in
humans were first accomplished by Yamakawa in 1920 although, not surprisingly, major problems existed in
formulating and stabilizing fatty emulsions.
Y of THE HISTORY of STERILE DRUG TECHNOLO
 Robert Koch in 1888 developed the first syringe that could be sterilized and Karl Schneider built the first
all-glass syringe in 1896. Becton, Dickinson and Company created the first mass-produced disposable
glass syringe and needle, developed for Dr. Jonas Salk’s mass administration of one million American
children with the new Salk polio vaccine.

 Pasteur, Lister, and Koch all contributed to discovery of the germ theory of disease, concerns for sterility,
use of aseptic techniques, and development of sterilization methods during the 1860s. However, their
concerns for the need to sterilize and maintain sterility of injections were not accepted or implemented for
decades. It was not until 1884 that the autoclave was introduced by Charles Chamberland for sterilization
purposes. Gaseous sterilization was first discovered using formaldehyde in 1859 and ethylene oxide in 1944.
It was also in the early 1940s that radiation, beginning with ultraviolet light, was used as a means of
sterilization.
Injectable Drugs—Therapeutic Classes and Examples
22 11 21

1. Safety (freedom from adverse toxicological concerns)


2. Sterility (freedom from microbiological contamination)
3. Nonpyrogenic (freedom from pyrogenic—endotoxin—contamination)
4. Particle-free (freedom from visible particle contamination)
5. Stability (chemical, physical, microbiological)
6. Compatibility (formulation, package, other diluents)
7. Tonicity (isotonic with biological fluids)

Seven Basic Characteristics of Sterile Product


Dosage Forms
Parenteral products are defined as preparations
The first general chapter of the USP is entitled “<1>
intended for injection through the skin or other
INJECTIONS.” Within this section are the following
external boundary tissue where the active
subcategories with the content under each subcategory
ingredient is introduced directly into a blood
summarized. Of course, wording of these characterizations
vessel, organ, tissue, or lesion. Parenteral
might change over time so the reader must consult the
products are to be prepared scrupulously by
current edition of the USP for current wording.
methods designed to ensure that they meet
Pharmacopeial requirements for and, where
 Sterility appropriate, contain inhibitors of the growth of
 Particulate Matter
 Pyrogens microorganisms.
 Other Contaminants

CHARACTERISTICS OF STERILE DOSAGE FORMS


FROM THE UNITED STATES PHARMACOPEIA
FINITIONS INCLUDED in THE USP are as FOLLOW
Pharmacy Bulk Package: A pharmacy bulk Large- and Small-Volume Injections: The demarcation of
package is a single product containing a sterile drug volume differentiating a small from large-volume injection is 100
injection, sterile drug for injection, or sterile drug mL. Any product 100 mL or less is a small-volume injection. The
injectable emulsion (i.e., suspensions cannot be main purpose for differentiating large- from small-volume
contained in pharmacy bulk packages. A pharmacy injections is the method of sterilization. With perhaps a single
bulk package contains many single doses of the exception for blood products, all large-volume injections must be
active ingredient to be used for the preparation of terminally sterilized while most small-volume injections are not
admixtures for infusion, or, using a sterile transfer terminally sterilized.
device, for filling empty sterile syringes. The
Biologics: This definition simply states that pharmacopeial
closure of the bulk package shall be penetrated only
definitions for sterile preparations for parenteral use do not apply
once with a sterile device that will allow measured
to biologics because of their special nature and licensing
dispensing of the contents.
requirements. Biologic requirements are covered in USP <1041>
general chapter.
Information that is contained on a product label includes the following:
 For liquid products: Percentage content of the drug or amount of drug in a specified volume.
 For dry products: The amount of active ingredient.
 The route of administration.
 A statement of storage conditions and an expiration date.
 The name and place of business of the manufacturer, packer, or distributor.
 Identifying lot number—The lot number is capable of yielding the complete manufacturing history of the
specific package, including all manufacturing, filling, sterilizing, and labeling operations.

LABELING
TYPES of STERILE DOSAGE FORMS
Types of sterile
dosage forms
Sterile dosage forms basically can be
classified in three broad categories:

1. Conventional small volume


injectables
2. Conventional large volume
injectables
3. Modified release (depot)
injectables
Examples of injectable dosage forms.
(A) Solution. Source: Courtesy of Baxter Healthcare
Corporation.
(B) Suspension. Source: Courtesy of Dr. Gregory
Sacha, Baxter.
(C) Lyophilized powder (Gemzar). Source: Courtesy
of Eli Lilly and Company.
(D) Emulsion. Source: Courtesy of Teva
Pharmaceuticals
EXAMPLES OF COMMERCIALLY
AVAILABLE
Dextrose injections LARGE VOLUME INJECTIONS
 2.5% Dextrose injection, USP in glass container
 5% Dextrose injection, USP in glass container
 5% Dextrose injection, USP in VIAFLEX plastic container
 10% Dextrose injection, USP in VIAFLEX plastic container
Dextrose and electrolyte injections
 5% Dextrose and electrolyte no. 48 injection (multiple electrolytes and dextrose injection, Type 1, USP)
Dextrose and sodium chloride injections
 5% Dextrose and 0.2% sodium chloride injection, USP
 2.5% Dextrose and 0.45% sodium chloride injection, USP
 5% Dextrose and 0.9% sodium chloride injection, USP
 5% Dextrose and 0.45% sodium chloride injection, USP
 5% Dextrose and 0.33% sodium chloride injection, USP
Miscellaneous injections
 Ringer’s injection, USP
 Lactated Ringer’s injection, USP
 Sterile Water for Injection, USP (for drug diluent use only)
 5% Sodium bicarbonate injection, USP
 Sodium lactate injection, USP (M/6 sodium lactate)
 Ringer’s injection, USP
OSMITROL (Mannitol) injections in VIAFLEX plastic container
 10% OSMITROL injection (10% Mannitol injection, USP)
 15% OSMITROL injection (15% Mannitol injection, USP)
 20% OSMITROL injection (20% Mannitol injection, USP)
 5% OSMITROL injection (5% Mannitol injection, USP)
PLASMA-LYTE (electrolyte) replenishment solutions in VIAFLEX plastic container
 PLASMA-LYTE 148 injection (multiple electrolytes injection, Type 1, USP)
 PLASMA-LYTE A injection pH 7.4 (multiple electrolytes injection, Type 1, USP)
 PLASMA-LYTE 56 and 5% dextrose injection (multiple electrolytes and dextrose injection, Type 1, USP)
08 01 21

INJECTION
CATEGORIES
JECTION CATEGORIES

There are six main categories of injectable products:


1. Solutions ready for injection
2. Dry, soluble products ready to be combined with a solvent prior to use
3. Suspensions ready for injection
4. Dry, insoluble products ready to be combined with a vehicle prior to use
5. Emulsions
6. Liquid concentrates ready for dilution prior to administration.
SUSTAINED
RELEASE
INJECTABLE
DELIVERY
SYSTEMS
An explosion of advances and commercial successes in
controlling and/or sustaining the delivery of injectable
drugs has occurred in the past few years (2–10). Major
technologies developed for injectable controlled release

SUSTAINED include primarily microspheres, implants, or hydrogels. For


pharmaceutical protein controlled or sustained release,
RELEASE microsphere or hydrogel technologies are the most likely

INJECTABLE choices. These systems include classical microcrystalline

DELIVERY suspensions (e.g., NPH or Lente insulin formulations),


biodegradable microspheres, nondegradable implants, gel
SYSTEMS
1. Increased duration of release, reduced number of
systems, pegylated protein formulations, and

injections, and increased compliance hyperglycosylated protein formulations. Sustained- or


2. Localized delivery in the case of cancer therapy and
controlled-release injectable delivery systems are desirable
vaccinations
3. Protection against in vivo degradation of the active for three main reasons:
ingredient.
Polymeric implants are sterile, solid drug products manufactured by compression, melting, or sintering processes. The
implant consists of the drug and a biodegradable or replaceable polymeric system, with the polymeric system generally
being the rate-controlling key to sustained and prolonged drug delivery. Commercial examples of polymeric implants
include
1. Norplant®: Levonorgestrel in silastic capsules deposited
subdermally into the upper part of the arm within one week
of the onset of menses. Drug delivery can last up to five
years.
POLYMERIC 2. Duros®: A titanium cylindrical osmotic pump implanted in
the upper arm that delivers drug for weeks to months.
IMPLANTS 3.
Viadur® is an example.
Gliadel® wafer: Polifeprosan plus carmustine are
formulatedwith a biodegradable polyanhydride copolymer
with the wafer being 1.45 cm in diameter and 1 mm in
thickness. This wafer is implanted into the cavity created by
a brain tumor resectionwith up to eight wafers (61.6 mg
carmustine) implanted that provides up to three weeks of
antineoplastic therapy.
4. Compudose®: composed of silicone rubber for
subcutaneous estradiol implantation behind the ear of cattle.
POLYMERIC
The polymeric systems used to fabricate drug-containing microspheres operate under at least five different mechanisms
for sustained or controlled drug release.
1. Bioerodible release—The microsphere erodes layer-by-layer like an onion with equal amounts of drug localized
within each layer. Bioerodible polymers include hydrophobic materials such as poly(ortho esters) with acid-labile
linkages.
2. Biodegradable release—The microsphere erodes gradually as a whole (bulk erosion) with equal amounts of drug
released per unit time.
3. Swelling-controlled release—The microsphere hydrates and swells with drug diffusing out of the polymer due to
internal pressure produced by the swelling. There are dozens of swelling-controlled polymers including natural
materials such as alginates, chitosans, collagen, dextrans, and gelatin and synthetic polymers such as cross-linked
hydrophilic polymers like poly(2-hydroxyethylmethacrylate) and poly(N-isopropylacrylamide).
POLYMERIC
4. Osmotically controlled release—The microsphere consists of semipermeable membranes that swell, but do not
burst. The drug is propelled out of the polymer through an orifice in the polymer produced by a laser.
5. Diffusion-controlled release—The microsphere permits constant diffusion of the incorporated drug through the
polymeric membrane. Hydrophilic polymers such as hydroxypropyl cellulose or hyaluronic acid are examples of
diffusion-controlling polymers.
Lactide/Glycolide Injectable Microsphere Extended
Release Products
THE PRODUCT
DEVELOPMENT
PROCESS
Main Steps Involved in the Formulation of a New
Sterile Drug Product  Target dose? Of course, need to know the target dose
and/or range to design formulation experiments.
 Target patient population—This relates to knowing
the therapeutic activity of the active pharmaceutical
ingredient (API) and who will be receiving the
product. Will there be a pediatric indication that
could affect formulation component choices? Will
there be an elderly indication that could affect ease of
use of the medication?
 Route of administration and mode of therapy—How
will the drug product be administered. . . bolus dose,
intermittent dosing, infusion? This impacts toxicity,
safety, acute versus long-term usage of the drug
product.
 Type of delivery system (e.g., vial, syringe, and
infusion).
Main Steps Involved in the Formulation of a New
Sterile Drug
Safety concerns Product
. . . classification of the active  Initial formulations—depends on solubility, stability,
ingredient, material safety data sheet (MSDS), intended clinical usage. Let us assume that stability
personnel precautions. limitations require the drug to be freeze dried:
 Analytical method development - Freeze dry the drug alone, determine what happens
- Potency method - If excipients needed, start with commonly known
- Purity method excipients that
- Stability-indicating method a. Produce acceptable cakes with rapid reconstitution
- In-process assay rates
- Identity method b. Have minimal collapse temperatures
 Basic chemistry of active ingredient c. Provide the desired finished product with respect to
- Structure, hydrophilicity, stability questions, etc. the nature of the solid (crystalline vs. amorphous).
- Physical and chemical properties . . . salt form, pKa,  Formulations should have solids content between 5%
partition coefficient, solubility, etc. and 30% with a target of 10% to 15%.
- Solubility studies . . . structure dictates solvents to
study
- Solution stability—function of temperature, pH
 Compatibility with other materials (excipients,
packaging).
 Determine the maximum allowable temperature  Large molecules typically need
(chap. 10) permitted during freezing and primary - Bulking agents, stabilizers (protectants, surfactants,
drying buffers)
- (Te/Tg/Tc) of tentative formulation - Salts should be avoided (low Te and Tg ) plus
 Select the appropriate size of vial and product fill concentration effects on proteins
volume. - Tonicity modifiers (mannitol and/or sucrose best)
 Select the appropriate rubber closure  Once formulation and package tentatively selected,
- Low water vapor transmission, no oil vapor determine appropriate freeze dry process
absorption, top design minimizes shelf sticking parameters, for example,
 Conduct initial stress tests (e.g., freeze-thaw cycling, - Rate of freezing
agitation studies) to screen initial formulations. - Need for annealing
 Small molecules - Temperature and pressure during primary drying
- May not need an excipient (depends on nature and - Temperature and pressure during secondary drying
amount of active ingredient) - Sealing under vacuum or nitrogen
- Might need one or more of the following: bulking  Optimize formulation and process based on stability
agent, buffer, salt information both during and after lyophilization
- Most stability problems with small molecules are process and after storage in dry state.
moisture related, not the effects of freezing and/or
drying as is the case with large molecules
- Generally, the drier, the better
FORMULAT 22 11 21

ION
COMPONE
NTS
Sterile formulations, by necessity, must be as simple as possible. Safety considerations limit the number and choices of additives to
use in formulations besides the active and, if stability is sufficient, a vehicle. The ideal parenteral formulation would contain the
active ingredient and water and nothing else.
ADDED SUBSTANCES
The USP includes in this category all substances added to a preparation to improve or safeguard its quality. An added
substance may:
 Increase and maintain drug solubility. Examples include complexing agents and surface active agents. The most
commonly used complexing agents are the cyclodextrins, including Captisol® . The most commonly used surface-
active agents are polyoxy ethylene sorbitan monolaurate (Tween 20) and polyoxy ethylene sorbitans monooleate
(Tween 80).
 Provide patient comfort by reducing pain and tissue irritation, as do substances added to make a solution isotonic
or near physiological pH. Common tonicity adjusters are sodium chloride, dextrose, and glycerin.
 Enhance the chemical stability of a solution, as do antioxidants, inert gases, chelating agents, and buffers.
 Enhance the chemical and physical stability of a freeze-dried product, as do cryoprotectants and lyoprotectants.
 Enhance the physical stability of proteins by minimizing self-aggregation or interfacial induced aggregation.
Surface-active agents serve nicely in this capacity.
 Minimize protein interaction with inert surfaces such as glass and rubber and plastic. Competitive binders such as
albumin and surface-active agents are the best examples.
 Protect a preparation against the growth of microorganisms. The term preservative sometimes is applied only to
those substances that prevent the growth of microorganisms in a preparation. However, such limited use is
inappropriate, being better used for all substances that act to retard or prevent the chemical, physical, or biological
degradation of a preparation.
 While not covered in this chapter, other reasons for adding solutes to parenteral formulations include sustaining
and/or controlling drug release (polymers), maintaining the drug in a suspension dosage form (suspending agents,
usually polymers and surface-active agents), establishing emulsified dosage forms (emulsifying agents, usually
amphiphilic polymers and surface-active agents), and preparation of liposomes (hydrated phospholipids).
VEHICLES (SOLVENTS)
Most Commonly-Used Water-Miscible Co-Solvents in
The solvent in injectable formulations typically
Injectable Products (Percent Range Approved by FDA)
is the largest component. Of course, the
preferred solvent or vehicle is water for
injection (WFI). For drugs that are not
sufficiently soluble in water, water-miscible
organic co-solvents may be used with
limitations on the acceptable amounts from a
safety view point. For drugs completely
insoluble in water and not required to be
injected intravenously, oily (oleaginous) solvent
systems of vegetable origin may be used.
Dielectric Constants for Various Solvents
Example of co-solvent effect on drug solubility.

Examples of Added Solute Substances Used In Commercial


Sterile Dosage Forms To Increase Injectable Drug Solubility

Dielectric constant is a measure of the electric current conductivity property of solvents.


The higher the dielectric constant, the better electric current will travel through the
solvent. Thus, water has the highest ε while oil has the lowest. Poorly soluble drugs will
have greater solubility in solvents whose ε is not as high as water. Thus, mixtures of
water and one or more water-miscible co-solvents will solubilize slightly polar drugs.
Examples of Polysorbates Contained
in Commercial Protein Formulations
ANTIMICROBIAL AGENTS
Examples of Commercial Sterile Dosage Forms The USP states that antimicrobial agents in
Containing Antimicrobial Preservative Agents and Their bacteriostatic or fungistatic concentrations must
Concentrations be added to preparations contained in multiple-
dose containers. They must be present in
adequate concentration at the time of use to
prevent the multiplication of microorganisms
inadvertently introduced into the preparation
while withdrawing a portion of the contents with
a hypodermic needle and syringe. The USP
provides a test for antimicrobial preservative
effectiveness to determine that an antimicrobial
substance or combination adequately inhibits the
growth of microorganisms in a parenteral
product. Because antimicrobials may have
Protein pharmaceuticals, because of their cost and/or frequency of use, are preferred to be available as inherent toxicity for the patient, the USP
multiple-dose formulations (e.g., human insulin, human growth hormone, interferons, vaccines). However,
several proteins are reactive with antimicrobial preservative agents (e.g., tissue plasminogen activator,
prescribes maximum volume and concentration
sargramostim, interleukins) and, therefore, are only available as single dosage form units (see chap. 8). limits for those that are used commonly in
Phenol and benzyl alcohol are the two most common antimicrobial preservatives used in peptide and
protein products. Phenoxyethanol is the most frequently used preservative in vaccine products.
parenteral products
BUFFERS
Buffering agents are used primarily to stabilize a solution
against chemical degradation or, especially for proteins, Most Common Buffers Used in Sterile Drug Solutions
physical degradation, i.e., aggregation and precipitation
that might occur if the pH changes appreciably. Buffer
systems employed should normally have as low a
buffering capacity as feasible so as not to disturb
significantly the body’s buffering systems when injected.
In addition, the buffer type and concentration on the
activity of the active ingredient must be evaluated
carefully. Buffer components are known to catalyze
degradation of drugs. The acid salts most frequently
employed as buffers are citrates, acetates, and phosphates.
Typical buffers used in parenteral systems include phosphate, citrate, and acetate buffers. Sodium or potassium salts
are also commonly used. Buffer choice depends on compatibilities of the buffer system and the type of process
intended for manufacture. For example, phosphate buffers are typically not used for lyophilized materials because
the pH changes dramatically over the course of the very low temperatures experienced in the lyophilization process.
Similarly, acetate systems are not always used in lyophilization because the acetate buffer may tend to flash off
during the lyo process.
ANTIOXIDANT
Substances called antioxidants or reducing agents are required frequently to preserve products because of the ease
with which many drugs are oxidized. Sodium bisulfite and other sulfurous acid salts are used most frequently.
Ascorbic acid and its salts also are good antioxidants. The sodium salt of ethylenediaminetetraacetic acid (EDTA)
has been found to enhance the activity of antioxidants in some cases, apparently by chelating metallic ions that
would otherwise catalyze the oxidation reaction.

The oxidation process is initiated by the formation of a free radical due to the loss of a hydrogen atom that is
catalyzed by one or more of the following environmental or product factors:
- High temperature—Ambient temperature can be problematic for some oxygen sensitive drugs. Manufacturing
environments for processing oxygen-sensitive products should be in the temperature range of 15° to 21°C.
- High pH
TONICITY AGENTS
While it is the goal for every injectable product to be isotonic with physiologic fluids, this is not an essential
requirement for small-volume injectables that are administered intravenously. However, products administered by all
other routes, especially into the eye or spinal fluid must be isotonic. Injections into the subcutaneous tissue and
muscles also should be isotonic to minimize pain and tissue irritation. Tonicity-adjusting agents most commonly used
are electrolytes (sodium chloride most common), glycerin, and mono- or disaccharides.
OPROTECTANTS, LYOPROTECTANTS, BULKING A
Some products are so hydrolytically labile that they must be lyophilized to ensure long-term shelf life stability. Often,
the level of drug substance in these products is in milligram quantities and is not sufficient to provide an elegant
looking cake. Sometimes, the level of the drug substance is even microgram quantities and cannot even be seen in the
vial by the clinician. Therefore, an excipient is used to create the cake so that the vial appears to have product and give
a visual indication that the product is in good condition. These excipients are called “bulking agents.”

Bulking agents range from various amino acids to sodium chloride to a host of sugars. Glycine is an example of an
amino acid that is used for bulking. However, amino acids are expensive. Sodium chloride can be difficult to freeze-
dry, depending on the circumstances. Therefore, sugars are the most commonly used bulking agents.
These substances serve to protect biopharmaceuticals from adverse effects due to freezing and/or drying of the product
during freeze-dry processing. Sugars (nonreducing) such as sucrose or trehalose, amino acids such as glycine or lysine,
polymers such as liquid polyethylene glycol or dextran, and polyols such as mannitol or sorbitol all are possible cryo-
or lyoprotectants. Several theories exist to explain why these additives work to protect proteins against freezing and/or
drying effects. Excipients that are preferentially excluded from the surface of the protein are the best cryoprotectants,
and excipients that remain amorphous during and after freeze-drying serve best as lyoprotectants. These concepts of
additive stabilization of biopharmaceuticals during freezing, drying, and/or in the dry state
COMPETITIVE BINDERS
 These additives are used if the active ingredient is known to bind excessively to container and manufacturing
equipment surfaces. Such additives compete with the active ingredient for the surface-binding sites and keep the
active ingredient from losing potency or activity in the dosage form. Historically, the best or most commonly
used competitive binder has been human serum albumin (HSA) at concentrations ranging from 0.1% to 1.0%.
 Concerns used to exist over potential viral contamination of natural substances such as HSA. Attempts to
identify other potential competitive binding agents as effective as HAS have generally been unsuccessful,
although it has been reported that Polysorbate 80, albeit at fairly high concentrations, inhibited recombinant
Factor VIII adsorption at solid–water surfaces (17). Recombinant HSA removed the viral contamination fears
and is now used in commercial products.
SURFACTANTS
Surface-active agents (surfactants) exert their effect at surfaces of solid–solid, solid–liquid, liquid–liquid, and liquid–
air because of their chemical composition containing both hydrophilic and hydrophobic groups. Surfactants effectively
compete against proteins for these interfacial hydrophobic locations, thus helping to minimize protein adsorption and
potential aggregation.
COMPLEXING AGENTS
Protamine sulfate is an example of a complexing agent used to prepare suspensions. As excess protamine is
undesirable from an immunogenicity standpoint and may impact the stability of biphasic (solution: suspension)
mixtures by complexing some of the soluble component, the exact ratio required to completely complex all of the
available peptide or protein needs to be determined. Under appropriate conditions, no detectable free protamine or
peptide/protein remains in the supernatant.
Aluminum salts are complexing agents that were covered in the preceding text as adjuvants.
OTHER ADDITIVES
Other purposes for solute additives in sterile product formulations include bulking agents for freeze-dried products,
suspending agents and wetting agents for suspensions, emulsifying agents for emulsions, viscosity-inducing agents
for topical ophthalmic products, and the specialized polymers used to formulate advanced sustained-, prolonged-,
extended-, delayed-, or controlled-release dosage forms (microspheres, liposomes, gels, and other specialized
injectable delivery systems).
EXAMPLES OF ADDITIVES USED IN
SPECIALIZED STERILE
DOSAGE FORMS

You might also like