Search

Search
Generic filters

The United States Pharmacopoeia

The United States Pharmacopeia (or to give it its full name, The United States Pharmacopoeia and the National Formulary (USP–NF) is a pharmacopoeia published by the United States Pharmacopoeial Convention. It is the official compendia of the United States of America.

The USP-NF contains monographs and standards for medicines, finished dosage forms, active drug substances, excipients, biologics, compounded preparations, medical devices, dietary supplements, and many other therapeutic goods intended for use in healthcare.

The United States Pharmacopeial Convention (confusingly also called the USP) is the non-profit organization that owns the trademark and copyright to the USP-NF. It is charged with the responsibility for publishing the USP-NF and managing the process of developing official monographs as well as availing official reference standards.

The USP-NF does not enforce any laws – it merely carries official standards developed by the United States Pharmacopoeial Convention for medical articles manufactured and marketed in the United States. Enforcement is caried out by the U.S. Food and Drug Administration (FDA).

 

History of the United States Pharmacopoeia

The United States Pharmacopeia traces its origins to the early part of the 19th century. The War of Independence (1775-1783) had exposed the inadequacy of existing standards in controlling medicines required to meet the needs of American military hospitals.

Leading physicians of the time begun collating information on medicinal products and organising it into dispensatories and pharmacopoeias. In 1820, a group of 11 physicians met at the US Capitol in Washington DC, the result of which was the creation of the first United States Pharmacopeia (published as The Pharmacopoeia of The United States of America).

The first editions were very basic, and aimed only to serve as “authoritative” resources for the public to consult. They included a few hundred products that were deemed well understood and characterised at the time.

In 1848, the US Congress passed the Drug Importation Act in response to public outcry about imported patent medicines from Europe, which many felt substandard. This act recognised the United States Pharmacopeia as the official compendia for the United States of America.

Independently of the United States Pharmacopeia, the American Pharmaceutical Association established the National Formulary (NF) in 1888. The NF principally aimed to serve as a resource and a formulary for small-scale compounding of medicines.

In 1906, the Pure Food and Drugs Act further codified the United States Pharmacopeia and National Formulary standards, recognising both compendia for product strength, quality and purity. Subsequent legislation, including the 1938 Federal Food, Drug and Cosmetic Act, further cemented the role of these compendia in standardisation of quality of medicines in US commerce.

In 1975, the United States Pharmacopoeial Convention purchased the right to the National Formulary, bringing together the two compendia under one roof, thus creating the United States Pharmacopoeia-National Formulary that we know today.

 

Organisation of the United States Pharmacopoeia

Over the years, the USP-NF has evolved, not just in terms of national and international recognition but also in the scope of the information it covers.

Now in its 43rd edition (from May 2021), the current USP-NF remains true to its original mandate: to be an accessible source of information for quality of prescription, nonprescription, and compounded medicines; excipients; biologics; medical devices; and dietary supplements.

Actives are generally featured in the United State Pharmacopoeia section while excipients appear in the National Formulary. If an excipient is also used as an active it will be featured in the United States Pharmacopeia section.

It features close to 5000 monographs, of which there are around 1500 monographs for active ingredients, 450 for excipients, and over 2500 for finished dosage forms.

In addition to monographs, the USP-NF also features 350 “General Chapters” sections, which covers information on assays, tests, and procedures used in monographs.

 

Available Formats

The print version of the USP-NF is no longer available. The only available format now is online via a subscription.

The online format is highly convenient not only because of the convenience of access but any updates are automatically posted.

The online format also offers improved search functionalities, and moreover you can set alerts as well as bookmarks for pages of interest.

 

Subscriber Resources

 

References

Brinckmann R. M et al (2020). Quality Standards for Botanicals — Legacy of USP’s 200 Years of Contributions. HerbalGram (American Botanical Society) 126: 50-65. Accessed 10.11.2021.

 

The European Pharmacopoeia

The European Pharmacopoeia (Ph.Eur) is a pharmacopoeia published by the European Directorate for the Quality of Medicines and Healthcare (EDQM) under the auspices of the Council of Europe and all the signatory states to Treaty Number 050. The Ph.Eur is the de facto official pharmacopoeia of the European Union.

Through its monographs and general chapters, the Ph.Eur fosters public health by elaborating and communicating scientifically-valid standards required for assessing and controlling quality of medicines and excipients.

The European Pharmacopoeial Commission is the decision-making body at the EQQM responsible elaboration and maintenance of the Ph.Eur content, including revision and updates of different monographs and general chapters.

 

History of the European Pharmacopoeia

The Ph.Eur traces its origins to 1963, when the Public Health Committee of the Council of Europe adopted a draft Convention that lay the legal, technical and administrative foundations of the Ph.Eur. The following year, the Committee of Ministers adopted the Convention and the Rules of Procedure that would govern the European Pharmacopoeia Commission.

Between 1965 and 1966, a Technical Secretariat was expanded and a Commission appointed. Three years later, the first edition of the Ph.Eur was published, and contained just over 100 monographs. Six years later in 1975, the European Union adopted Council Directive 75/318/EEC, which made compliance with Ph.Eur monographs mandatory when applying marketing authorisations.

Now in its 10th edition, the Ph.Eur as well as the Convention has 39 signatory parties from across Europe, including the European Union, that participate and vote on sessions of the European Pharmacopoeia Commission.

 

Legal Framework

Several regulations form the legal basis for the Ph.Eur. They are:

 

Organisation of the European Pharmacopoeia

The Ph.Eur is arranged into different sections, including

  • General Chapters
  • General Monographs
  • Monographs for Vaccines, Immunosera, Radiopharmaceuticals, Sutures, Herbal products and Homeopathic products
  • Specific monographs on Active Pharmaceutical Ingredients and Excipients, and
  • Specific Monographs on Dosage Forms

The 10th Edition of the Ph.Eur (including Supplement 10.5) contains 2447 monographs (including dosage forms), 378 general texts (including general monographs and methods of analysis) and about 2800 descriptions of reagents.

All these standards are designed to meet the information needs of scientists and managers involved in research and quality control of medicines, regulatory authorities and those involved in the manufacture of medicinal products or individual components.

 

Available Formats

The Ph.Eur is available as a single reference volume that covers all relevant articles featured. The 10th Edition was released in July 2019 and will be updated with eight periodic supplements over the following three years (10.1 to 10.8).

Available in either English or French, the print version contains a subscription key (EPID code) that allows access to online archives. The compendium can also be accessed online via a licence (individual or shared access).

 

Subscriber Resources

The 2021 subscriptions to the European Pharmacopoeia Supplements 10.3-10.5 are available on the EDQM WebStore.

 

References

The European Medicines Agency: European Pharmacopoeia

Quality standards of the European Pharmacopoeia

 

Pharmaceutical Matrix Forming Excipients: Overview, Types, and Selection Criteria

As medical needs become more complex, the need for modified release pharmaceutical dosage forms becomes ever more salient. The matrix tablet technology, which uses matrix forming excipients, has been instrumental in this quest. But exactly what is a matrix former? How do they function, and how are they used? This technical note provides an overview, types and function of this important class of pharmaceutical ingredient.

What is a matrix forming excipient?

Matrix forming excipients are a class of materials used to fabricate sustained-release (prolonged release) pharmaceutical tablets. The matrix former creates a three-dimensional network that not only acts as a structural and functional scaffold for active ingredients but is the physical embodiment of the dosage form.

The matrix controls the entry of GI fluids into the tablet which retards the rate of diffusion of the active ingredient from the tablet, hence, its elimination half-life (time taken for the drug’s plasma concentration to reduce by 50%).

Matrix tablets have been around for more than half a century. Their benefits are easy to see when you consider the fact that 75 percent of all sustained release solid dosage forms used by patients today utilise matrix systems, a proportion that is larger than tablet film coatings, and many other approaches currently available for use to pharmaceutical scientists.

Why are matrix formers used in tablets?

The main reason for using tablet matrices is to achieve sustained release drug delivery. These dosage forms are designed to deliver active ingredients over a period of up to 24 hours. This results into longer-lasting therapeutic effects while reducing dosing frequency and side effects, while improving individual’s well-being.

The achievement of both goals improves patient adherence to medication, making treatments more successful, while also reducing costs of healthcare, especially for chronic conditions. This is increasingly important, due to changing demographics (aging), and children with medical complex needs. Click here to read about the importance of applying empathy in new drug development, and the challenges faced by patients with swallowing problems.

Matrix tablets are selected to formulate sustained release oral dosage forms because of ease and the economics of their manufacture. These products can be easily manufactured by direct compression or wet granulation depending on the type of polymer and additional excipients used in the formulation.

Generally, active drug substances best suited for incorporation into matrix systems exhibit the following characteristics:

  1. They exhibit neither very slow nor very fast rates of absorption and elimination. Drugs with slow rates of absorption or elimination half lives in excess of 10 hours are inherently self-retarding.
  2. Drug substances that have very short half-lives below 2 hours require special care during development because of potential huge fluctuations in plasms levels
  3. Drug substances whose solubility in aqueous media should be good and also maintain adequate residence time in the gastrointestinal tract.
  4. Active ingredients that are poorly absorbed or at unpredictable rates are also not good candidates for sustained release formulations.
  5. Finally, drug substances with very narrow therapeutic windows or really small doses are also poor candidates for sustained release formulations because of the difficulty of limiting control of release and preventing dose dumping.

Like what you're reading? Please share it to your network.

Types of matrix forming excipients

The different materials currently used to formulate matrix tablets can be classified into four groups on the basis of the basis of their drug retarding mechanism:

Hydrophobic matrix formers

Hydrophobic matrix materials are water-insoluble large molecular weight polymers for use in the fabrication of sustained release dosage forms. Examples of hydrophobic matrix formers include

  • Polyethylene
  • Polyvinyl chloride
  • Ethylcellulose, and
  • Acrylate copolymers

Sustained drug release is produced as a result of swelling and diffusion through a network of channels that exist between compacted polymer particles. The rate-controlling step in these formulations is speed of liquid entry into the matrix.

Lipid matrix formers

Lipid-based matrix formers are high melting point fatty acids and/or waxes capable of forming matrices for sustained release. Drug release from such matrices occurs through both pore diffusion and erosion.

Release characteristics are therefore more sensitive to digestive fluid composition than to purely insoluble polymer matrices.

Examples of lipid matrix formers include

  • Carnauba wax in combination with stearic acid,
  • Glyceryl dibehenate
  • Glyceryl tristearate
  • Tripalmitin
  • Trymyristin, and
  • Hard Fats

Lipid matrices are inert, non-eroding and non-dissolving systems that achieve drug release prolongation through the creation of a hydrophobic domains in the tablet. These domains not only slow down the hydration of the tablet but they also control the rate of dissolution and release of the drug into the aqueous milieu.

Hydrophilic matrix formers

Hydrophilic matrix formers are the most widely used materials of their kind owing to their ease of use, cost-effectiveness and wider regulatory acceptance. They are typically hydrophilic, high molecular weight polymers with high gelling capacities. Upon contact with water, they swell and gel, creating a moving barrier that regulated the rate of drug release. For this reason, they are also known as swelling controlled release systems.

Polymers used in the preparation of hydrophilic matrices are divided in to three broad groups

  1. Cellulose derivatives, such as methylcellulose, hydroxypropylcellulose, Hypromellose, and Sodium carboxymethylcellulose.
  2. Non cellulose natural or semi synthetic polymers such as Agar-Agar; Carob gum; Alginates; Xanthan gum, Pectin, Chitosan and Modified starches.
  3. Polymers of acrylic acid, such as Carbomer 934

Biodegradable matrix

The last category of matric forming materials are a special group of polymers that are biologically degraded or eroded by enzymes to generate simple metabolites that are eliminated through the usual processes. These polymers may be natural polymers such as proteins and polysaccharides, semi-synthetic polymers or fully synthetic systems, such as the well-known aliphatic poly (esters) and poly anhydrides.

Read about the key differences between Hypromellose and Polyethylene Oxide matrix forming excipients here:

Hypromellose v Polyethylene Oxide for the Formulation of Matrix Mini Tablets

Examples of drug products that use matrix former technology

Here are examples of tablets formulated as matrix systems for controlled release:

Theophylline

Theophylline is a methylxanthine, chemically similar to caffeine. It is one of the drugs used in the treatment of asthma and COPD due to its safety and low cost. Theophylline sustained release tablets use hydrophilic polymers such as hydroxyethylcellulose and hypromellose in combination with polyethylene glycol 6000 and cetostearyl alcohol (as a dissolution rate retardant).

Paliperidone

Paliperidone, which is also referred to as 9-hydroxyrisperidone, is the primary active metabolite of risperidone. It is a widely prescribed antipsychotic medication today. The oral dosage forms are available as sustained release tablets using polyethylene oxide and hydroxyethylecellulose as the matrix forming excipients.

Oxycodone

Oxycodone is a semisynthetic opioid drug substance derived from thebaine. It is similar to hydrocodone and morphine. It is currently used for moderate to severe pain due to its powerful analgesic and sedative properties. It exhibits excellent oral bioavailability and is commonly used and/or co-formulated with other analgesics. It is available as sustained release tablets, immediate release tablets, oral solutions and injectables. Matrix tablets when used as the drug release prolonging technology make use of ammoniomethacrylate copolymer (Eudragit® RS 30D), polyvinyl acetate and Glyceryl dibehenate.

Felodipine

Felodipine is a methyl and ethyl diester of 4-(2,3-dichlorophenyl)-2,6-dimethyl-1,4-dihydropyridine-3,5-dicarboxylic acid. Together with amlodipine and nifedipine, felodipine is a calcium-channel blocker that lowers blood pressure by reducing peripheral vascular resistance through a highly selective action on smooth muscle in arteriolar resistance vessels.

Felodipine sustained release tablets are variously formulated using either hydrophilic or hydrophobic matrix formers, such as Hypromellose K4M and hydrogenated castor oil.

Benefits of using matrix forming excipients

The reasons why particular matrix formers are selected for use by pharmaceutical formulation scientists are many and varied. There is no doubt, however, that as a technology class, these materials offer many advantages, which are outlined below:

  • Improve patient convenience and compliance by enabling the formulation of prolonged release dosage forms (e.g once-a-day tablets) that reduce dosing frequency
  • Highly versatile in terms of the active ingredients that can be formulated and the manufacturing methods
  • Enable the reduction of drug toxicity since the decrease plasma fluctuations and maintain drug within the therapeutic window
  • They can enhance product stability
  • Prolonged release drugs permit the use of lower amount of total active ingredients thereby reducing drug utilization and exposure
  • Improves bioavailability of some drugs
  • Matrix formers are relatively inexpensive as a technology

Summary about pharmaceutical matrix formers

Matrix tablets are widely used in the development of sustained release products. A wide range of inactive ingredients known as matrix formers, including hydrophilic, hydrophobic and biodegradable polymers, as well as high melting point lipids are used in matrix tablets. These systems are popular because they are well studied and are amenable for manufacture on standard equipment and processes.

Sources

Pharmacentral has a strict referencing policy and only uses peer-reviewed studies and reputable academic sources. We avoid use of personal anecdotes and opinions to ensure the content we present is accurate and reliable

Pharmaceutical Suspending Agents: Overview, Types, and Selection Criteria

Suspending agents are excipients added to disperse systems in order to maintain particulate ingredients in suspension or to prevent other forms of physical instability. In this technical note, we provide an introductory review of what suspending agents are, and outline the function they play in pharmaceutical formulations.

Definition of a Pharmaceutical Suspending Agent

Pharmaceutical suspending agents are a class of excipients added to disperse systems to ensure that any solid particles in the formulation are kept uniformly distributed within the continuous phase, thereby maintaining physical stability of the product.

Pharmaceutical dispersions include suspensions, creams and aerosols. They typically comprise dispersions of insoluble solids (drug and/or excipients) in an aqueous or non-aqueous liquid medium, known as the continuous phase.

Depending on the particle size of the disperse phase, disperse systems can be further categorised as:

  • Colloidal (the dispersed particles are in the submicron range), and
  • Coarse (dispersed particles are above colloidal size).

Note that the term ‘suspending agent’ is frequently used interchangeably with another, closely related term, ‘thickening agent’.

As we will see, while having some commonalities the two excipients groups are different. Very simply, thickeneing agents can be used as suspending agents but not all suspending agents are thickening agents.

This is best illustrated by the following commonly used OTC products:

Pepto Bismol® is an oral suspension of bismuth salicylate (active). It is formulated with aluminium magnesium silicate, methylcellulose, and gellan gum, which are the materials used to hold the active ingredient in suspension by increasing viscosity (thickeners).

Gaviscon® is an oral suspension of sodium bicarbonate, calcium carbonate, and sodium alginate. The sodium alginate functions both as a suspending excipient and active ingredient.

Calpol® is an oral suspension of paracetamol formulated with maltitol and sorbitol (syrups), microcrystalline cellulose, sodium carboxymethylcellulose, and xanthan gum, as the suspending agents. Suspension of the active is achieved through thickening of the dispersing phased.

In order to appreciate the utility of suspending agents, it’s necessary to quickly review the technical aspects of pharmaceutical disperse systems.

The process of dispersing solid particles in a liquid creates several problems, including:

  • Creaming
  • Sedimentation
  • Flocculation
  • Caking
  • Particle growth, and
  • Adhesion onto the walls of the container (in some formulations)

The above phenomenon constitute physical instabilities, and are generally dependent on three main factors:

  • Differences in density between the dispersed particles and the dispersion medium (∆ρ)
  • Particle size (radius) of the dispersed phase r
  • Viscosity of the dispersion medium (η)

The interrelationship between these three factors is given by Stokes’ law, which is shown below:

v=2gΔρr²/9η

where v is the rate of particle settling (or creaming, for that matter).

According to Stokes law, instability can be reduced by reducing particle size, decreasing density differences between the two phases or increasing the viscosity of the continuous phase.

It is worth knowing that particles will still be able to move about in the dispersion, however, the rate and intensity of collisions will be minimised. Furthermore, disperse systems are highly dynamic – dispersed particles frequently contact each other as a result of the following phenomenon:

  • Brownian motion
  • Creaming
  • Sedimentation due to gravitational forces
  • Convection

Given enough time, and in the absence of any other mitigating circumstances, the dispersed particles eventually come into close contact and may coalesce, form bonds and cake, destroying the balance of the disperse system.

Whether dispersed particles reach this point of no return depends on two factors:

  • Forces of attraction or repulsion, and
  • Nature of the surface of the dispersed particle

This is where suspending agents come into play, i.e they help minimise dispersed particle flux (sedimentation, creaming or convection) or interfere with particle-particle attraction.

We can therefore define pharmaceutical suspending agents as a specific category of excipients added to disperse systems to minimise disperse phase coalescence and instability.

Pharmaceutical suspending agents can be grouped into four main categories as outlined below:

Types of Pharmaceutical Suspending Agents

A). Wetting agents

For the disperse phase to freely and distribute in the continuous phase, it needs to be wetted by the liquid. The presence of entrapped air pockets on the particle surface or if the particles are hydrophobic is a hindrance to dispersion.

Wettability can be enhanced through the reduction of the interfacial tensions, namely solid-liquid and liquid-vapour interfaces.

1. Surface active agents

Surface active agents (surfactants for short) are excipients that lower the surface tension (or interfacial tension) between two liquids or between a liquid and a solid.

Surfactants with HLB values between 7 and 9 have been shown to be suitable wetting agents in pharmaceutical disperse systems.

This is because the apolar groups of the surfactant are able to adsorb onto hydrophobic parts of formulation particle while the polar groups project into the aqueous medium, resulting in a lowering of the interfacial tension between the solid and the liquid.

2. Hydrophilic colloids

Hydrophilic colloids are high molecular polysaccharide polymers or clays. They include acacia, bentonite, tragacanth, alginates and cellulose derivatives.

Being hydrophilic, they are able to coat particles to impart hydrophilic character to the disperse phase. In addition, hydrophilic colloids may also increase viscosity of the continuous phase, reducing the rate of sedimentation of particles.

3. Solvents

Common polar pharmaceutical solvents such as glycerol, propylene glycol and polyethylene glycol and alcohol are highly water-miscible and are able to reduce the liquid-air interfacial tension.

They enable water to infiltrate deep into power agglomerates, displacing entrapped air and enabling wetting of the dispersion to take place.

B). Flocculants

A well-formulated disperse system is one that exhibits the correct degree of flocculation. Systems that are underflocculated generally tend to settle and sediment very rapidly although the compacts formed are loose and redispersion is possible.

On the other hand, overflocculation results in products that while settle slowly the sediments pack tightly such that redispersion is not possible.

By controlling the degree of flocculation, it’s possible to reach a happy medium – in which there is a degree of flocculation and also deflocculation.

This can be achieved through, not only through particle size control and viscosity-modification, but also by using flocculating agents, including electrolytes, ionic surfactants and polymer flocculating agents.

1. Electrolytes

The addition of inorganic electrolytes to the solution changes the zeta potential of the dispersed particles, and provided this is lowered sufficiently enough, will produce a flocculated system.

2. Surfactants

Ionic surfactants can also be used to create flocculation in a disperse system by neutralising particle charges and creating a deflocculated system.

3. Polymeric flocculants

Several polymeric excipients are capable of forming gel-like networks in disperse systems thereby adsorbing onto and trapping dispersed particles and holding them in a flocculated state.

These materials are mainly hydrocolloids although not always, and include:

C). Viscosity modifiers

The deal disperse system is one that displays pseudoplastic or thixotropy behaviour, that is, exhibit high apparent viscosity at low shear rates.

Upon storage, the dispersed particles either settle slowly or, and preferably, remain suspended. When the product is sheared, for instance, when shaken by the consumer, the high apparent viscosity of the formulation should fall sufficiently for product to be dispensed easily.

The various materials currently used as viscosity modifiers are outlined below:

1. Polysaccharides

Examples of polysaccharide-based viscosity modifying excipients include the following:

2. Water-soluble cellulose ethers

Several cellulose ethers have the ability to increase viscosity of aqueous systems in which they are dispersed. They include:

The viscosity-increasing properties of cellulose ethers depends on the molecular weight and degree of substitution.

3. Hydrated silicates

Hydrated silicates are naturally-occurring siliceous clays that exist as colloids in water. This group of pharmaceutical grade clays includes:

  • Bentonite
  • Hectorite
  • Magnesium aluminium silicate (VEEGUM®)

Hydrated silicate materials exhibit thixotropic behaviour at low concentration in water and as gels at high concentration.

4. Carbomers

Carbomers are high molecular weight cross-linked polyacrylic acid polymers that swell in water to form viscous hydrogels depending on the degree of cross-linking.

Among carbomer excipients, carbomer 940 is the most commonly used suspending excipient in both topical and oral products.

If you would like to learn more about carbomers, here is a link to an article that gives an overview on these important excipients:

D). Density modifiers

From probing Stokes’ law above, it is clear that if the densities dispersed and dispersing medium are of the same magnitude sedimentation would be significantly slowed down.

Thus, changing the density of the dispersing medium, for example, addition of glycerol, propylene glycol, polyethylene glycol or sucrose-based syrups, can significantly modify densities and leveraged to control instability.

You read more about viscosity modifying excipients through this link:

Summary of Pharmaceutical Suspending Agents

Many pharmaceutical products are formulated as disperse systems in which particulate solids (active ingredients and/or excipients) are distributed throughout a continuous or disperse phase. Owing to the tendency towards settling or creaming by the disperse phase, the use of suspending agents is mandated.

There are many different types of pharmaceutical suspending agents; they can be grouped into four main classes depending on the mechanism of function:

Careful selection of suspending agents is essential in order to have a product that ensures the disperse phase does not settle rapidly, the particle do not settle into a cake, the system is easily re-dispersed into a uniform mixture when shaken, is easy to dispense from the container or use by the consumer.

Sources and Further Reading

Pharmaceutical Thickeners (Viscosity-Increasing Agents): Overview, Types, and Functions

Thickening agents are usually high molecular weight polymer excipients commonly used to increase viscosity of formulations. This can serve several objectives, including stability, ease of use and drug delivery. In this post, we provide an overview of materials frequently used to thicken pharmaceutical products.

Definition of Pharmaceutical Thickeners

Pharmaceutical thickening agents (thickeners, for short) are a diverse group of excipient materials used to increase the viscosity of liquid pharmaceutical systems, ideally without altering other fundamental properties.

They are also commonly known as viscosity-increasing agents.

Viscosity is the internal friction that occurs between the molecules within a liquid. It is a measure of the resistance to deformation in shear, such as flow or pouring, and is due to intermolecular friction and molecular adhesion-cohesion within the fluid.

Viscosity of a fluid decreases with increase in temperature due to increased atomic/molecular mobility, which decreases intermolecular cohesion and friction. The presence in solution of a high concentration of solutes can have an appreciable impact on viscosity of the solution.

This is why the terms “thick” (concentrated and high viscosity) and thin (dilute and low viscosity) are used as qualitative descriptions of the viscosity of liquid systems.

Many thickeners form weakly cohesive colloidal structures, which can exist as solutions, suspensions or gels when dissolved or dispersed in a suitable liquid medium.

Note that thickening agents are sometimes (and mistakenly) referred to as suspending agents. Although there is overlap in the type of excipients used for both product categories, there are sufficient differences to warrant a clear separation of the two categories, an approach we have taken in this article.

How Thickeners Increase Solution Viscosity

With respect to the use of polymers, the addition of a high molecular weight polymer increases viscosity of the liquid in which it is dissolved. The increase in viscosity is caused by strong internal friction between the randomly coiled and/or swollen macromolecules, and the surrounding solvent molecules, through hydrogen bonding¹. This is illustrated below:

How Thickeners Increase Solution Viscosity

With respect to the use of polymers, the addition of a high molecular weight polymer increases viscosity of the liquid in which it is dissolved. The increase in viscosity is caused by strong internal friction between the randomly coiled and/or swollen macromolecules, and the surrounding solvent molecules, through hydrogen bonding¹. This is illustrated below:

Functions of Pharmaceutical Thickeners

Viscosity is one of the most important properties associated with liquid systems, and formulations that possess high viscosity have many practical applications within the pharmaceutical space, as outlined below:

Drug delivery

The rate of drug delivery is influenced by viscosity of the milieu. Liquid and semisolid topical and oral formulations, as well as parenteral and matrix-based solid dosage forms frequently utilise high viscosity systems to slow down rates of drug diffusion.

Demulcent properties (soothing, irritation reduction & increase in slip)

Demulcents are a class of materials used to lubricate and protect mucous membranes, and more especially the oral, pharyngeal, oesophageal, and gastric mucosa. Their action depends on having sufficient tack and slip, both of which are obtained when polymeric excipients are dissolved in aqueous media. Click here to read about the challenges of dysphagia and oral medicines, and what action is required if we are to address it.

Substantivity and film formation

The ability of a formulation to adhere and remain in place (contact time) is a function of, among other factors, viscosity and film formation. Viscosity-increasing agents are routinely used to increase substantivity and contact time of topical and bioadhesive formulations.

Suspending and stability enhancement

Thickeners are typically used to minimise separation and settling that typically follow when insoluble ingredients are suspended in liquids

Improvement of appearance, consistency and quality

Thickening agents are added to skincare formulations to give skincare products a more appealing consistency and smoothness. Consistency is a particular formulation characteristic that describes the firmness or runniness of a product.

Gelling and texturizing

A key property of hydrocolloids (such as pectin and xanthan gum) is their ability to form gels and thicken liquid product. For instance, pectin, carrageenan and agar form gels under specific conditions, which opens up a myriad of applications and functionalities.

Like what you're reading? Share it with your network.

Types of Pharmaceutical Thickening Agents

A wide range of excipients can be used as thickening agents, with the most common being hydrocolloid gums and cellulose ethers.

Hydrocolloid Gums

The term ‘hydrocolloid’ is derived from two Greek words, ‘hydro = water, and ‘kolla’ = glue. Currently, the term hydrocolloid is used for macromolecular (polymers) hydrophilic substances (with affinity for water), and includes polysaccharides and proteins.

When dissolved or dispersed in water, hydrocolloids hydrate and swell (increase in volume) due to solvent molecules penetrating into void spaces that exist between polymer chains. Hydration is accompanied by an increase in viscosity.

Hydrocolloids can be classified into four groups:

Plant-derived hydrocolloids: including pectin, agar, Alginates, acacia, tragacanth, Karaya gum, guar gum, starches, cellulose and locust bean.

Products of fermentation: including xanthan gum, dextran, gellan gum and pullulan.

Semi-synthetic hydrocolloids: including modified starches, cellulose ethers (Methylcellulose, Hypromellose, Ethylcellulose, Hydroxypropylcellulose and hydroxyethylcellulose), amidated pectin and propylene glycol alginate.

Animal derived hydrocolloids: such as gelatin, chitosan and caseinates.

Vinyl polymers

Vinyl polymers is a generic term given to high molecular weight synthetic polymers based on the vinyl groups: XCH=CHY. In the pharmaceutical field, the most important vinyl polymers are:

Water soluble polyvinylpyrrolidones: This group includes povidone and copovidone of molecular weights ranging from 2500 to 1250000 g/mol.

Ethylene-vinyl acetates: Ethylene vinyl acetates (EVAs) are copolymers of ethylene and vinyl acetate monomers produced by free radical polymerisation under high temperature and pressure.

Varying vinyl acetate composition impacts polymer properties such as melting point, crystallinity and polarity, solubility, transparency, hardness and compatibility with active ingredients.

EVAs are mainly applied in the formulation of transdermal drug devices and Class I – IV medical devices, where they are used as and/or incorporated into matrix formers, rate controlling membranes and backing layers. EVAs provide tackiness, bioadhesion and flexibility.

Polyvinyl alcohol: Polyvinyl alcohol (PVA) is a water-soluble and biodegradable polymer obtained by polymerisation of vinyl acetate followed by partial hydrolysis. Pharmaceutical grades typically exhibit a degree of hydrolysis of 88% and molecular weight that vary from 20 000 g/ml to over 100 000 g/mol.

Clays (Aluminium Silicates)

Clays are fine-grained naturally-occurring aluminium silicates with traces of other inorganic oxides. They typically consist of plate-like crystal habits. The most well-known clays in the pharmaceutical field are bentonite, hectorite and magnesium aluminium silicate.

Owing to their platy habits, aluminium silicate particle planes possess different surface charge characteristics and complex modes of particle-particle interaction, depending on concentration and pH, among other factors.

In suspension, hydrodynamic forces or the influence of electroviscous effects of clay solutions impacts rheology and viscosity of clay suspensions, much the same way as polymers dispersions.

Carbomers

Carbomer are a group of closely related synthetic, high molecular weight, nonlinear polymers of polyacrylic acids, cross-linked with a polyalkenyl polyether. They differ in molecular weight, polymerization solvent or side-groups and contain between 56 and 68% w/w carboxylic acid groups.

Carbomer are highly versatile and multifunctional excipients for oral (solid and liquids) and topical formulations. When hydrated and neutralised, they yield highly viscous gels with viscosities upwards of 40,000 mPa s for concentrations as low as 0.5% w/v.

If you’re interested in learning more about carbomers, we have prepared a quick reference guide which you access through this link:

Carbomers: Overview, key properties and formulating tips

Polyols, sugar and oligosaccharide

Concentrated solutions of sugars, oligosaccharides and polyols behave very much like high molecular weight polymer solutions owing to increased intermolecular attractions between sugar molecules (hydrogen bonds) and the solvent. When sheared, the interactions create a drag which manifests as an increase in viscosity.

This class of viscosity-increasing agents include liquid Sorbitol, liquid Maltitol, sucrose, fructose, dextrose, maltodextrin and Polydextrose.

Miscellaneous Thickening Agents

Polyethylene glycol and polyethylene oxide: Polyethylene glycol and polyethylene oxide are related non-ionic polymers formed by the reaction of ethylene oxide and water under pressure in the presence of a catalyst. Low molecular weight polyethylene glycol grades are liquids and used primarily as solvents and co-solvents in oral and topical formulations. Solid polyethylene glycol grades are used suspending agents or to adjust the viscosity and consistency of other suspending vehicles.

Silica: Also known as colloidal silicon dioxide, silica is an oxide of silicon with the chemical formula SiO2. Silica grades with a high specific surface area are used to thicken polar liquids thereby converting them into transparent gels. This effect can be used as stabilising strategy for emulsions and semisolid preparations.

Other materials which can be used for thickening formulations include:

Summary of Thickening Agents

Many pharmaceutical excipients are often added to formulations to increase the viscosity or thicken liquid formulations. This property can be utilised to improve product properties, including drug delivery, stabilisation, demulcent effects, and texturizing. The range of materials that serve this function is very broad, but typically includes hydrocolloid gums, synthetic polymers, sugars and polyol syrups and many other miscellaneous materials.

Pharmaceutical Diluents and Fillers: Overview, Types, and Uses

Diluents and fillers are a ubiquitous class of materials in formulated products, including pharmaceuticals, medical devices, vaccines, nutraceuticals, cosmetics and industrial goods. But what exactly are diluents and fillers? Do they perform any function beyond filling up space? This technical note provides an introductory review of pharmaceutical diluents and fillers. The information will be particularly relevant to anyone interested in learning more about fillers and diluents.

Definition of Pharmaceutical Diluents and Fillers

For the vast majority of medical products in current use, the active pharmaceutical ingredient is rarely administered in its basic form. Often, this is because it is present in much lower amounts relative to the weight of the dosage form unit.

A specific type of excipients known as bulking agents are therefore added to product formulations to provide bulk and render the product convenient to process, manufacture or administer.

Alternative names for fillers and diluents are:

  • carriers
  • fillers
  • diluents
  • extenders
  • voluminising agents

Not every formulation requires a diluent – in product formulations where the active ingredient or other functional excipients are already added in sufficient proportions, for instance, greater than 70 – 90% a bulking agent may not be required.

When added, however, and despite their humble name, pharmaceutical fillers do a lot more than just fill space. Here are some of their functions:

  • add structure
  • aid application e.g., mouthfeel
  • impact appearance
  • influence many quantifiable properties that are important to a product’s performance.

In solid dosage forms, fillers and diluents are almost always required as they provide the foundation upon which the formulation is constructed.

For convenience of administration (holding and swallowing, for instance), a tablet should weigh no less 50mg, and where the active ingredient is present in very low quantities, a diluent with high carrying capacity is absolutely essential. This is an important consideration, particularly today given the increasingly high percentage (and growing) demographic of seniors across the world. Click here to read about the importance of applying empathy in new drug development.

As shown in the graphic below, diluents are often present in the highest concentration.

Diluents and fillers are not limited solid dosage forms only – their usage also applies to many other product types, including topical creams and lotions, parenteral products, dusting powders, aerosols, suppositories, and suspensions.

Types of Pharmaceutical Diluents and Fillers

Pharmaceutical fillers are very diverse group of excipients. They may be:

  • organic chemicals
  • mineral substances
  • metal oxides
  • polymers
  • simple inorganic chemicals,
  • liquids,
  • gases (aerosols)

They may be classified on the basis of

  • source (natural, mineral or synthetic)
  • dosage form in which they are used
  • physical and chemical properties and
  • functionality

A more practical approach is to classify fillers as either functional or non-functional.

Functional fillers are those excipients that extend the property gamut of the formulation, opening up new performance qualities and functionalities. For example, silica is frequently added to elastomers and rubbers to provide to the topical medical device strength and structure without compromising drug delivery properties.

Non-functional fillers, as the name suggests, only serve one thing – to provide bulk and fill void volume in the formulation, which is still an important requirement in the formulation for the reasons outlined previously.

The image galery below shows fillers that may be encountered in the pharmaceutical industry:

  • maltodextrin (used as a carrier for a pharmaceutical flavour)
  • native maize starch
  • compressed chewing gum base

Ideal Properties of a Pharmaceutical Filler and Diluent

When it comes to pharmaceutical formulations, there are a number of fundamentals properties that formulators seek in fillers:

  • Physiological inertness
  • Filler concentration
  • Particle size and size distribution of the filler
  • Shape and aspect ratio
  • Bulk density/carrying capacity
  • Low API binding capacity
  • Processability
  • Physical and chemical stability
  • Strength (low impact)
  • Cost-effectiveness

For solid dosage forms (tablets and capsules), the following factors (in no particular order) are the most important considerations when selecting a material to use:

  • Compressibility and compactibility
  • Flowability
  • Particle size and distribution
  • Moisture content and type of interactions
  • Bulk density
  • Compatibility
  • Solubility and effect on bioavailability
  • Abrasiveness (lubricity)
  • Stability
  • Physiological inertness
  • Cost and availability
  • Regulatory acceptance

The 10 Most Popular Filler-Diluents in Solid Dosage Forms

The following fillers are consistently the most commonly used materials in solid dosage forms:

Lactose

Lactose is a natural disaccharide obtained from milk. It consists of one galactose and one glucose moiety. It is listed in the USP-NF, JP and PhEur and is also GRAS listed and included in the FDA Inactive Ingredients Database (IM, and SC: powder for injections; oral: capsules and tablets: inhalation preparations; vaginal preparations).

Lactose is one of the most widely used filler and diluent in tablets and capsules, and to a more limited extent in lyophilized products and infant formulas. Lactose is also used as a diluent in dry-powder inhalation.

Find more about lactose through these links on our site:

Microcrystalline cellulose

Microcrystalline cellulose is purified, partially depolymerized cellulose and one of the most widely used ingredients in the pharmaceutical industry. It is used in many types of dosage forms, including as filler, binder, flow aid, lubricant, texturiser, among others.

Microcrystalline cellulose is listed in all major pharmacopoeia, including the USP-NF, Ph.Eur, BP, JP and IP. It is also GRAS listed and accepted for use as a food additive in Europe and the United States and included in the FDA Inactive Ingredients Database (oral: powders, suspensions, syrups and tablets; topical & vaginal products).

Find more about the uses of microcrystalline cellulose through these links on our site:

Starch

Starch is a polysaccharide consisting of two polymers: a linear polymer known as amylose and a branched polymer known as amylopectin. Both polymers are made up of glucose residues.

Pharmacopoeia have individual monographs for different starch derivatives, including native starch, pregelatinised starch, tapioca starch, pea starch and rice starch. Starch is also co-processed with lactose and microcrystalline cellulose.

Generally, as a material, starch is a highly versatile material, finding application in many different dosage forms. It is widely used as filler and diluents in solid dosage forms. It is also added to dusting powder and also in topical products and film coatings.

Find more about the uses of starch through these links:

Calcium phosphate

Calcium phosphate is actually a family of chemical substances made up of calcium ions and phosphate anions. These include anhydrous dibasic calcium phosphate, hydrated dibasic calcium phosphate, and tribasic calcium phosphate. All the three grades are listed in the main pharmacopoeia, are GRAS listed and included in the FDA Inactive Ingredients Database.

The main advantages of calcium phosphates as filler-diluents include:

  • Chemical inertness
  • Low hygroscopicity
  • High density
  • Source of calcium
  • Natural source

Find out more about uses of calcium phosphates through the following links:

Calcium carbonate

Calcium carbonate is the calcium salt of carbonic acid. It is listed in all the major pharmacopoeia and is GRAS listed. The FDA Inactive Ingredients Database lists calcium carbonate as an excipient for chewing gum, capsules and tablets.

It is one of the main fillers used in compressed tablets. Other uses are as a bulking agent in tablet sugar-coating processes and as an extender and opacifier in tablet film-coatings.

Sucrose

Sucrose is a naturally-occurring disaccharide made up of a glucose molecule joined to a fructose molecule. Pharmaceutical grade sucrose is currently obtained from sugar cane and sugar beet, and is listed in all the major pharmacopoeia and the FDA Inactive Ingredients Database (parenterals, tablets, capsules, syrups and topical products). It is a highly compressible filler in tablets and selected for its sweetness, low reactivity, safety profile and cost-effectiveness.

Maltodextrin

Maltodextrin is a saccharide consisting of mixture of polymers of D-glucose units. It is prepared by the partial hydrolysis of a starch. It is a non-sweet, odourless, white powder or granules.

Maltodextrin is used as a coating agent, tablet and capsule diluent, tablet binder and viscosity- increasing agent. Maltodextrin is also widely used in confectionery and food products, as well as personal care applications.

Mannitol

Mannitol (D-mannitol) is a polyol and an isomer of sorbitol. It occurs as a white, crystalline powder, or free-flowing granules, with a sweet taste. It is noted for its cooling, sweet sensation in the mouth. It is official in multiple pharmacopoeia as a filler diluent as well as a therapeutic agent.

Mannitol is used as a diluent (10 – 90% w/w) in tablet formulations. It’s specific advantage is its low hygroscopicity and high compressibility. However, it is a costly filler compared with lactose and selected for use where mouthfeel is important, for example, chewable tablets.

Sorbitol

Sorbitol is a polyol and an isomer of mannitol. It is supplied as a white or almost colourless, crystalline, hygroscopic powder. It is available in a wide range of grades and polymorphic forms, such as granules, powders and pellets for use in solid and liquid oral and topical products.

Sorbitol is used as a diluent in tablet formulations prepared by either wet granulation or direct compression. It is especially useful in chewable tablets owing to its pleasant, sweet taste and cooling sensation.

Sodium Chloride

Sodium chloride is an inorganic chloride salt having sodium as the exchangeable ion. It is listed in all the major pharmacopoeia, is GRAS and included in the FDA Inactive Ingredients Database (injections, inhalations, nasal, ophthalmic, oral, otic, rectal and topical products). It occurs as a white, crystalline powder with a saline taste.

Sodium chloride is used widely in parenteral products to produce isotonic solutions. In oral products, it functions as a diluent (capsules and direct compression tablets), and a porosity modifier in controlled-release coatings.

MaterialReason for PopularityUsage
Lactose (all grades)Highly compactibleUp to 60% w/w in tablets & capsules
Microcrystalline celluloseVersatile, inert and low costOptimum level is 15-20% w/w in tablets & capsules
Starch (all grades)Versatile, inert and low costUp to 40% w/w as a filler in tablets & capsules
Calcium phosphatesLow moisture content. Source of CalciumUp to 60% w/w in tablets & capsules
SucroseHighly compactible & Sweet tastingHighly compressible grades up to 90% w/w in tablets, capsules & oral liquids
MaltodextrinHighly compactibleUp to 50% w/w/ in tablets & capsules
MannitolMouthfeel & highly compactible and inertUp to 60% w/w in tablets & capsules
SorbitolMouthfeel & highly compactible and inertUp to 60% w/w in tablets, capsules & oral liquids

Summary about pharmaceutical Fillers and Diluents

Despite their ordinary name, pharmaceutical fillers do much more than simply fill up space. They provide structure, influence appearance, and impact many other measurable properties that ultimately determine product’s end uses.

For solid dosage forms, lactose and microcrystalline cellulose are the most commonly used diluents. Mannitol is used as a substitute for lactose, however it is costly and its use is only justified when other functional properties are sought in a formulation.

Sources Used

Pharmacentral has a strict referencing policy and only uses peer-reviewed studies and reputable academic sources. We avoid use of personal anecdotes and opinions to ensure the content we present is accurate and reliable.

  • A. Crouter, L. Briens, The effect of moisture on the flowability of pharmaceutical excipients, AAPS PharmSciTech, 15 (2014) 65-74. Pubmed
  • A. Lura, G. Tardy, P. Kleinebudde, J. Breitkreutz, Tableting of mini-tablets in comparison with conventionally sized tablets: A comparison of tableting properties and tablet dimensions, International journal of pharmaceutics: X, 2 (2020) 100061. Pubmed
  • N.O. Sierra-Vega, K.M. Karry, R.J. Romañach, R. Méndez, Monitoring of high-load dose formulations based on co-processed and non co-processed excipients, Int J Pharm, 606 (2021) 120910. Pubmed

What is an Active Pharmaceutical Ingredient?

Therapeutics, in its broadest definition, is the use of interventions aimed at alleviating the effects of disease in humans or animals.

The constitutive substances that elicit therapeutic effects are known by different names, including active, active pharmaceutical ingredient, active principle, drug substance, medicinal agents or therapeutic substance.

This article provides a comprehensive definition of active pharmaceutical ingredients and as well as answers to common FAQs.

Definition of Active Pharmaceutical Ingredients

An active pharmaceutical ingredient (or API) is defined as the chemical, biological mineral or any other entity or component responsible for the therapeutic (pharmacological, physiological, physical, etc) effects in a product, such as:

  • vaccine
  • pharmaceutical (medicine)
  • medical device

Active pharmaceutical ingredients are distinguishable from inactive pharmaceutical ingredients, commonly known as excipients or formulation aids. For a comparative discussion of what APIs are, click through this link for the World Health Organisation’s definition.

The European Medicines Agency, the US FDA and the International Conference on Harmonisation (Q7) all adopt the same definition of API as “any substance or mixture of substances intended to be used in the manufacture of drug (medicinal) products, and that, when used in the production of drug, becomes an active ingredient of the drug product.”

Many other terms are used interchangebly with active pharmaceutical ingredient. They include:

  • active
  • bulk pharmaceutical chemical
  • drug substance
  • active principle (especially for natural extracts)
  • therapeutic ingredient

You may want to take note that health authorities add qualifiers to the definition of actives, namely, that a substance becomes an active ingredient in the drug product when it’s used in the production of the drug product, and, actives are intended to provide pharmacological activity or any other direct effect that is important in the diagnosis, cure, prevention, treatment or prevention of a disease condition, or to modify the structure or function of the body.

The reason is that there are many substances that are used beyond therapeutics. For instance:

  • Insulin is also used in fermentation bioprocesses as well as an important medicine in diabetes
  • Warfarin is an aid in pest control (culling agent of Vampire bats) as well as an anticoagulant
  • Many other materials function as therapeutic substances as well as excipients. This list include simethicone which may be used as a processing aid or therapeutically as an anti-flatulent; docusate sodium is both an medicinal active (laxative) and a excipient (surfactant), and mannitol is used both as a filler in tablets and as a therapeutic substance in the treatment of glaucoma and kidney conditions.

Consider the fact that materials intended for use as pharmaceutical actives are subjected to very strict controls, with respect to quality controls during manufacturing, distribution and use, adding a qualifier to the definition allows regulators to apply the required standards to the relevant use category (API vs processing aid vs excipient), thus preventing dilution of standards.

A key attributes of active pharmaceutical ingredients is their ability to bind to receptors and elicit a physiological response that can also be advantageously used in the treatment of disease.

The image below is an illustration of a Tyrosine kinase receptor (TRK):

TRKs represent a widely studied class membrane receptors. They participate in many cellular functions, such as differentiation and apoptosis.

This has made them of particular interest in the search for anticancer agents, with more than 20 chemical agents successfully developed into therapeutic substances.

One of these is the active pharmaceutical ingredient, Imatinib, which is marketed as GLIVEC.

To find out more about how active pharmaceutical ingredients differ from excipients click on this link: Excipients Explained: Definition, Types, Uses, and Regulatory Controls.

What are the common sources of active pharmaceutical ingredients?

On the basis of origin, active pharmaceutical ingredients can be divided into four main categories as follows:

1. Synthetic organic chemistry as a source of drug substances

This group mainly includes small chemical substances, typically with a molecular weight of under 500 Daltons. The largest category of drug substances in use today are synthetic organic substances.

Examples of APIs obtained via synthetic organic chemistry include paracetamol (Panadol® – Glaxo) and artovastatin (Liptor® – Pfizer).

With better technology, it is also now possible to synthesise peptides having up to 20 amino acid residues in the laboratory for use as drug substances. Synthetic peptides include Somatostatin, Calcitonin (Fortical – )and Vasopressin (VASOSTRICT – Parpharm)

2. Natural organic molecules as sources of drug substances

From the time immemorial, humans have resorted to the nature as a source of therapeutic substances. This remains the case today.

For example, many antibiotics such as erythromycin and anticancer agents like paclitaxel (ABRAXANE® – BMS) are isolated from nature.

3. Recombinant DNA technology as a source of drug substances

Simply put, recombinant DNA technology is the process of altering gene of an organism and using the change to produce a biological molecule such as a large protein or chemical compound.

In just over a period of 40 years, recombinant DNA technology has grown to become one of the main sources of new drug substances today. Many important actives such as Human Insulin, Human Albumin, Erythropoietin and Human Growth Hormone are obtained this way, as are many vaccines and monoclonal antibodies.

4. Whole or Extractions from natural sources (animals)

There are still many therapeutic substances that can only be obtained from natural sources either as whole organisms or extracts from organisms.

Examples of these include blood and plasma, attenuated or live viruses used in vaccines and human immunoglobulins. The same applies to cells, tissues and organs used various in biotechnology modalities.

The table below summarises the main types of active pharmaceutical ingredients arranged by their source or origin:

Active TypeSourceExamples
Conventional small-molecule drugsSynthetic organic chemistryParacetamol, Ibuprofen, Imatinib
Conventional small-molecule drugsNatural productsPaclitaxel, Aminoglycoside antibiotics, Opiates & Ciclosporin
Conventional small-molecule drugsSemisynthetic compounds (derived from nature and modified in the laboratory)Penicillins (Ampicillin) & Simvastatin
Peptides & proteinsSyntheticVasopressin & Somatostatin
Peptides & proteinsExtracted from natural sources (human, animal or microbial)Insulin, Growth hormone & Human у-globulin
Peptides & proteinsRecombinant DNA technologyInsulin, Erythropoeitin, TNF-alpha
AntibodiesAnimal antisera & human immunoglobulinsSnake & Spider venoms & HNIG
AntibodiesMonoclonal antibodiesRituximab, Trastuzumab, etc
EnzymesRecombinant DNA technologyDornase, Galactosidase
VaccinesWhole organisms (live or attenuated)Smallpox, TB, tetanus, etc vaccines
VaccinesAntigen-based vaccines produced by recombinant DNA technologySeveral
DNA & mRNA productsRecombinant DNA technologyPatisiran
CellsHuman donor and engineered cellsVarious stem cell therapies
TissuesHuman & animal donors & engineered tissuesVarious
OrgansHuman donorsTransplant surgery

Irrespective of the type of drug substance, the process of isolating, preparing and purifying active ingredients is highly involved, and requires several painstaking steps.

In situation of high demand, this step-wise approach may not suit market requirements, for example, where the therapeutic good is in very high demand (Covid 19 vaccines?).

In recent decades, the pharmaceutical industry has sought to introduce technology aimed at improving synthetic yields of actives. When successfully applied, these technologies often result in major improvements in output over traditional processes.

Some technologies, though, promise much and deliver little. Click here to read about some of the technologies that promised much but have so far failed to improve drug discovery and development.

How are active pharmaceutical ingredients regulated?

EU legislation

The standards, requirements, and procedures that active pharmaceutical ingredients must meet before they can be used in marketing authorisations are primarily laid down in Directive 2001/83/EC, Regulation (EC) No 726/2004 and Directive 2011/62/EU. These regulations also set rules for the manufacture, distribution, and sale or advertising of medicinal products.

United States

In the United States, drug substances are controlled under the Federal Food, Drug, and Cosmetic Act of 1938 and subsequent amending statutes (which are all codified into Title 21 Chapter 9 of the United States Code). This law sets quality standards for drugs and medical devices manufactured and sold in the United States and provides for federal oversight and enforcement of these standards.

Did you know?

Did you know that the first synthetic active pharmaceutical ingredient is Chloral hydrate? It was synthesized by Justin Liebig in 1832 and introduced into medicine in 1869 as a sedative hypnotic. Although its use has declined, Chloral hydrate remains in use in some countries, particularly as a sedative for children.

Further Reading

Drug Discovery and Development: A Step by Step Guide

Drug discovery and development can be described as the sum total of steps taken by research-intensive entity to identify a new chemical or biological substance and transform it into a product approved for use by patients.

This highly knowledge and capital intensive process takes, on average, 10-15 years and over $2 billion (2021 figures), to pull off.

But exactly what is involved? Who does what and when? In this article, we outline key concepts of drug discovery and development, including target identification, clinical trials, pharmaceutical development and commercialisation.

Drug Development

Drug development covers all the activities undertaken to transform the compound obtained during drug discovery into a product that is approved for launch into the market by regulatory agencies. This is a pivotal process, and a lot rides on its success, thus, efficiency is absolutely critical, but mainly for two key points:

Firstly, development is expensive – accounting for 70% of the total R&D costs. Even though the number of projects is much smaller compared with those under discovery, the cost per project is significant, and increases exponentially as the project progresses through into the latter stages.

Secondly, speed is the essence in drug development as it determines how soon the company can start earning a return on the huge investment ploughed into the project. Besides, any delays eat away into exclusivity arrangements granted by patents and time before generic manufacturers can launch me-too products.

For clarity, drug development is presented as an operation that’s distinct from discovery. In reality, the distinction is not as clear cut. Often, different activities are being undertaken concurrently, and in deed, some processes that were traditionally undertaken much later on are increasingly being brought much forward. The idea is to identify compounds that have the highest chances of success much earlier and focus on those.

Components of drug development

The key activities involved in the development of a typical are summarised in the diagram below, showing the different tasks that are undertaken during this process. Generally, these tasks can be divided into three parts: technical; investigative and administrative.

General Perspective – New Drug Discovery and Development

The process of creating a new drug product can be broadly divided into three main phases:

  • Drug discovery – entailing the conceptualisation of the therapeutic into a molecule with known pharmacologic effects
  • Drug development – covering the steps taken to convert the molecule above into an approved and registered drug product
  • Commercialisation – which includes all the steps taken to convert the product into an approved therapeutic, launch into the market and to generate sales

These processes are schematically illustrated below, which is greatly oversimplified:

Historically, these functions were performed, respectively by the Research, Development and Marketing departments. Nowadays though, a number of these functions are outsourced to other companies that specialise in one or more aspects of these activities.

It’s worth emphasising that many activities described in the above scheme may proceed in parallel and other may spill out into other phases. For example, development activities, such as clinical trials or additional testing of formulations, generally continue well beyond registration of the drug product. Such tests may be driven by the requirement for more understanding about the new drug or the need to extend use beyond the main therapeutic applications.

The job of the discovery teams does not end with product registration and market launch. Many discovery scientists will carry out more research looking for other candidates to serves as backups in case the lead compound fails or as follow-on compounds that might have better safety or efficacy profiles over the lead compounds.

Finally, the three processes listed in the new drug development scheme above are not independent and consecutive. Rather, they are coordinated with each other because the performance of each process influences decisions taken in another stage.

Understandably, there are many competing interests as the new drug progresses through its journey. To successfully fit in and integrate all the different interests and cultures, effective project management skills are required.

Therapeutic Concept Selection

Therapeutic concept selection is about deciding whether or not to embark on a new 0project. Success at this stage is measured in terms of agreeing and signed off a drug discovery program with a clear-cut aim and timeline.

Exactly where the idea originates varies from company to company. Some companies have very strong exploratory research teams that undertake research internally and discover new knowledge on diseases and druggable targets. Others are more open minded, preferring to purchase new molecules in for further development. Often, it’s a mixture of both approaches.

Generally, the decision to select a particular program will be guided by three things: company strategy, technical capabilities and operational constraints.

  • Should the company do it? (Strategy)
  • Could the company do it? )scientific and technical capabilities)
  • Can the company deliver it? (operational constraints)

These three factors are summarised below:

Drug Discovery

The drug discovery process technically starts with choice of a disease area and a definition of the therapeutic need that should be addressed. Once this is done, the process proceeds to identification of the physiological mechanisms that need to be targeted, and ideally, identification of a specific molecular ‘drug target’.

During this phase, effort is focussed on identifying a lead chemical structure, designing, testing and fine-tuning it and ensure that it meets all the criteria required for development into a drug product.

An overview of the main stages that constitute a typical drug discovery project, from the point of identification of a target to the production of a candidate drug is shown below this process is shown below:

Discovery can at first appear like a shot in the dark. At the start, scientists will be dealing with a huge number of compounds (1020), which have to be filtered, mainly via computer simulations (in silico) into manageable number capable of being further optimised.

High throughput screening (HTS) is then applied to identify ‘HITS’ which demonstrate interesting activity. Since HTS can throw up a huge number of ‘HITS’ these are further optimised and validated to remove any artefacts or ‘noise’ from the screen.

A key aspect of validation stage is to find relationships between chemical structure and biological activity, and to find out if the compounds belong to any existing families of compounds (known as hits series).

Validated hits are therefore further studied, especially in terms of their pharmacokinetic profiles and toxicity. At this point, the number of compounds has reduced to a handful. It is these handful of substances that are subsequently entered into the lead optimisation programme.

Lead optimization is a critical process in drug discovery since it’s determines whether a suitable compound can be identified for taking forward into preclinical and clinical studies. Therefore, the goal of this stage is to scrutinise and fine-tune, typically in parallel, both the biological activity and the physicochemical properties of the lead series.

During this stage, rigorous data is generated in a precise, timely manner to quickly determine the compounds to progress the compounds, and the series, toward the ideal candidate profile. The higher the quality of these candidates, the higher the chances of successful progression into clinical trials.

“MAGIC BULLETS”

The term ‘magic bullet’ was coined by Paul Ehrlich (1854 – 1915), a Germany medical scientist and winner of the Nobel Prize in Physiology or Medicine in 1908. Ehrlich envisaged a compound (the bullet) capable of attacking a pathogen and destroying it while leaving its host intact. Nowadays, pharmaceutical scientists are developing targeted and personalised cancer therapies, and these, many argue, are modern realisations of Ehrlich’s idea.

Technical development – solving technical issues related to synthesis and formulation of the drug substance with the aim of ensuring the quality and safety of the drug product. The key functions involved here are chemical, manufacturing and formulation development.

Investigative studies – establishing the safety and efficacy of the product, including assessment of whether it’s pharmacokinetically suitable for clinical use. The main function involved here are safety pharmacology, toxicology and clinical development.

Administrative functions – coordinating and managing quality control, logistics, communications and decision making to ensure high quality data are generated and to minimise any delays. The main function involved here is project management.

In addition, there will be a team coordinating and liaising with regulatory authories, collating data, liaising with material suppliers and writing dossiers for presentation to authorities in order to gain approval in a timely manner.

Pharmaceutical Development

This stage is also known as pharmaceutical development. Since pure drug substances are rarely suited for clinic use, they need to be formulated; by combining them with excipients, into tablets, capsules, injections, etc.

Pharmaceutical development refers to all the different tasks undertaken to transform the drug substance identified as a candidate during the discovery phase into a dosage form that is able to reliably deliver the drug into the body in a safe and reliable way.

Designing a formulation can be as equally time consuming and complex as drug discovery. In order to mitigate some of the issues that crop up, initial studies are undertaken during lead optimisation, before development actually starts.

For conventional drug substances, the desired route of administration is the oral route. Alternatives are considered, particularly if sufficient bioavailability cannot be achieved orally.

The different tasks undertaken in pharmaceutical development can be grouped into two:

  • Preformulation studies which specifically investigate physical and chemical properties of the drug substance, such as solubility and dissolution rates; acidity and alkalinity (pKa), chemical and physical stability, lipophilicity, particle morphology, melting point and fracture behaviour, etc.
  • Formulation studies which are essentially, chemical engineering effort aimed at converting a powder or a liquid form of the drug substance into a stable and deliverable product. During this process, formulation scientists will take into account all the known properties of the drug substance and the desired drug delivery system that best meets the therapeutic objectives of the compound.

Clinical Development

Clinical development is an umbrella terms to describe the whole set of activities undertaken by the team in support of testing of a new drug substance in humans. It includes the following clinical trials, which refer to administration into man the new drug under controlled conditions to investigate bioavailability, efficacy, safety, tolerability and acceptability.

Clinical development of new drugs has been described as both a science and an art, since it requires technical expertise, sound judgement and commercial acumen.

Executed well, clinical development brings new medicines quickly and safely to patients who need them, while also managing to return on the financial investment.

At the time of writing this post In 2021, estimates of the investment required for clinical development studies vary, but most sources agree that, including the cost of failures and capital invested, the total cost of bringing a new drug to market is $2-3 billion, spread over a 12-year development cycle. Out of this cost, two-thirds is spent on clinical development.

The clinical development process is divided into four phases, summarised in the table below.

  • Phase I: first introduction and safety assessment in man, typically in healthy volunteers
  • Phase II: early exploratory and dose-finding studies in patients
  • Phase III: large scale studies in patients
  • Phase IV: post-marketing safety monitoring of patients.

In almost every country on earth today, clinical trials are a legal requirement before a new drug can be sold or claims made for its safety of benefits. This does not include alternative remedies, however. In addition, all clinical trials, include Phase I studies, are subject to international, national and in most cases, institutional regulations.

The different international regulations and requirements are set out in guidelines published by the International Conference on Harmonisation (ICH), an organisation that was set up to harmonise pharmaceutical regulations across Europe, United States and Japan.

It is a requirement that clinical development procedures be done under ICH guidelines if the results are to be accepted for registration in the three ICH guidelines. This does not mean that the studies need to be done there – they only have to comply with these guidelines.

In addition to ICH guidelines, human studies are required to be undertaken according to an ethical framework defined code, known as the The Declaration of Helsinki (2000). This code, in a nutshell, requires the principal investigator (physician) to protect life, health, privacy and dignity.

Regulatory Affairs

A fundamental maxim in pharmaceutical new product development is the basic division of responsibilities whereby the health authority, such as the US FDA, is responsible for safeguarding the public’s health against defective and unsafe products, and the pharmaceutical company being responsible for all aspects of drug product development (quality, safety and efficacy).

The regulatory authority develops regulations and guidelines for companies and others in the value chain to follow. The approval of a pharmaceutical product is a contract between the regulatory authority on behalf of the public, and the pharmaceutical company.

The regulatory authority is responsible for approving clinical trial applications, approving marketing authorisation applications, and monitoring safety and efficacy claims of the marketed drugs. Authorities can withdraw the approval at any point where there are cases of non-compliance.

The conditions of the approval are set out in a dossier and appear in the prescribing information. Changes to these terms have to be pre-approved and authorised before they can be implemented.

Within pharmaceutical companies, the regulatory affairs department is the one responsible for obtaining approval for the new pharmaceutical product and working to ensure that this approval is maintained for as long as the company desires so.

Regulatory affairs professionals work at the interface between the regulatory authority and the project team, and they’re often the channel of communication between external and internal stake holders with respect project’s regulatory standing and progress.

Milestones and Decision Points

The decision to advance a drug candidate into early development is the first of several key strategic decision points in a new drug development project. The timing, naming and decision-making process vary from company to company, the one conceptualised below was developed by Norvatis:

Early selection point (ESP

Is the decision to take the drug candidate molecule into early (preclinical) development.

Decision to develop in man (DDM)

Is the decision to enter the compound into Phase 1, based on information obtained during preclinical development phase. Once this decision is made, the company will aim to produce between 2 and 10kg of clinical grade material.

Full development decision point (FDP)

This point is reached after Phase I and Phase IIa studies have been completed. At this point, the company has some preliminary evidence about clinical efficacy in man. From here on, the project costs skyrocket and the company must be confident on commercial returns.

Submission decision point (SDP)

This is the final point when a decision is taken to apply for registration, based on the data collected and its quality.

Summary Points about Drug Discovery and Development

Pharmaceutical companies undertake research for commercial reasons and their overarching objective is a return on capital invested. This is not to say a few companies include altruistic motives, the fact of the matter is that it takes less priority.

For this reason, the research a company pursues has to be in line with its commercial goals. Curiosity-driven research is generally left to Universities and other institutes. That said, there are territories where the two universes overlap, namely, applied research. Many recent innovations in medicine, such as monoclonal antibodies, fall into this domain, and both pharmaceutical companies, and universities have contributed to their development.

Finally, it should be stated that drug discovery and development is unlike any other type of development or innovation process, such as developing a new car. Drug discovery and development carries far greater uncertainty, and the outcome is rarely assured.

Resources Used

Pharmacentral has a strict referencing policy and only uses peer-reviewed studies and reputable academic sources. We avoid use of personal anecdotes and opinions to ensure the content we present is accurate and reliable

  • R.G. Hill, H.P. Rang, Preface to 2nd Edition, in: R.G. Hill, H.P. Rang (Eds.) Drug Discovery and Development (Second Edition), Churchill Livingstone 2013
  • Orloff et a.,l The future of drug development: advancing clinical trial design. Nat Rev Drug Discov8, 949–957 (2009). https://doi.org/10.1038/nrd3025
orange coated tablets

Pharmaceutical Tablet Film Coatings

A pharmaceutical film coating is a polymer-based membrane applied onto solid dosage forms, such as tablets, capsules and granules, in order to impart certain desired attributes that are otherwise absent in the uncoated product.

In this post, we’ll tell you what film coatings are, why they are used in pharmaceutical products and how they are applied onto solid dosage forms.

What is a pharmaceutical film coating?

The term ‘coating’ usually implies a membrane formed directly on a substrate. It separates two phases between which fluids (gases, vapours or liquids) may be exchanged.

It is distinguished from ‘film’, a term used interchangeably with coating, which, however, refers to a preformed, stand-alone membrane (a film becomes a coating when applied onto a substrate) (Mwesigwa, 2006).

When used in relation to solid dosage forms, a film coating can be defined a polymer-based thin membrane applied onto the surface of tablets, capsules, granules and spheroidal unit dosage forms.

The coating generally imparts certain desirable attributes to the dosage that are otherwise absent in the uncoated form.

Such attributes include impartation of colour and branding marks, alteration of drug release properties, protection of sensitive ingredients or masking offensive odours or tastes, among others.

Pharmaceutical coatings are usually dense, non-porous films obtained from polymeric substances and are typically ≤ 100 μm thick (Hogan, 1995) although films with much higher thickness are not atypical.

Note that the use of polymer film coatings is not limited to solid dosage forms: many other products are routinely encased in polymer films of various types. For example, medical devices are coated with silicone resins to provide lubricity and protection.

Types of polymer film coatings

The practice of coating solid dosage forms (tablets, capsules, granules and spheroids) is a very old practice that has been undertaken for several centuries.

There are references to the use of mucilages to coat pills in Greco-Arab and Islamic herbal medical scripts.

As sugar became more widely available in the 19th century, the food and confectionery industry developed pan coating as a means for improving aesthetics of candy.

This technology was subsequently borrowed and adapted for use in the pharmaceutical industry, and remained the only means for coating until the early 1950 when the American pharmaceutical company, Abbott Laboratories marketed the first polymer film coated tablet.

This set a precedent for further introduction of many other polymers for use in pharmaceutical coatings. Presently, although sugar coated drug products are still widely marketed the vast majority of products utilise polymer film coatings.

Polymer coatings can be broadly categorized into three groups based on their functional roles:

(1) Conventional coatings

Conventional coatings are sometimes known as non-functional or immediate release coatings.

They are the most commonly encountered type of tablet coating.

The reason they are known as non-functional coatings is because they are designed to dissolve rapidly in the gastrointestinal tract without any significant impact on the rate of drug release in the gut.

Conventional coatings serve several purposes, including:

  • protective
  • aesthetics
  • masking taste or odours
  • improving mechanical strength of the core
  • facilitating product handling
  • reduction of the risk of interaction between incompatible components, and
  • reduction of dust and the associated explosion hazards

The main requirement for conventional coatings is pH independent solubility, as mentioned above, so as to have a minimum influence on drug release of the core.

(2) Delayed release coatings

Delayed release coatings are mainly of the enteric type.

They are designed to prevent disintegration of the dosage form in the stomach. However, on reaching the intestine, delayed release coatings disintegrate to release the drug for absorption or local action.

There are many reasons why an enteric coating may be applied to solid dosage forms:

  • Firstly, they may be utilised to protect certain drug substances which are hydrolysed in the stomach by gastric secretions (e.g., acids and enzymes),
  • They can be used to prevent gastric distress associated with acidic drugs (e.g., aspirin or other NSAIDs),
  • They can help deliver drugs intended for local action in the intestines, e.g., intestinal antiseptics, to deliver drugs that are optimally absorbed in the intestines to their prime absorption site, or to effect a delayed response to the dosage form.

Therefore, the ideal enteric-coating should be impermeable to gastric juices but easily susceptible to intestinal juices.

The majority of enteric polymers are have carboxyl ionizable groups that become protonated in acid media but form salts in alkaline pH enabling them to dissolve in the intestinal juices.

(3) Sustained release coatings

The third category is for covers coatings used to control the rate of release of drug substances from a dosage form and extend the release time over several hours.

Sustained release coatings are also known as controlled, extended or prolonged release coatings, although teach of these terms has specific meanings.

Generally, the most common mechanism of sustaining release is controlling the diffusion rate of the drug through the polymer membrane.

Depending on the diffucion mechanism sought, different release kinetics can be obtianed, such as zero-order (constant release), first-order release kinetics, or pulsatile or sigmoid release patterns.

Note that in the literature, the term ‘modified release’ coating is also sometimes used. This term is non-specific and covers both delayed release and sustained release coatings.

Controlled release coatings serve a variety of purposes:

  • They are used to achieve more effective therapy while eliminating the potential for both under- and overdosing.
  • Controlled release coatings are also used to achieve the slow release of highly soluble and or permeable drugs, fast release of low-solubility drugs, or delivery of drugs to specific sites, such as the colon.
  • Lastly, it is not uncommon that pharmaceutical companies to use controlled release technologies as a way of achieving brand line extension or to add marketing depth to an existing brand.

Materials used in polymer film coatings

A film coating is constituted from a variety of materials that include a polymer, a plasticizer, and other additives (surfactant, colorants/pigments and lubricants).

Once combined and dissolved or dispersed in a solvent or aqueous medium, the different ingredients are sprayed onto the substrate and allowed to form a thin, continuous envelope.

Polymers

The polymer is the film former and the main component on the formulation.

The ideal polymer is one capable of producing smooth, thin films that are reproducible under routine coating conditions.

The range of materials that can be used as film formers is very wide and covers materials that may be natural, synthetic or semi-synthetic.

Polymers used in conventional film coatings

This category includes:

  • cellulose ether derivatives, e.g., hypromellose (also called hydroxypropyl methylcellulose), methylcellulose, hydroxypropylcellulose and sodium carboxy methylcellulose
  • vinyl polymers, e.g., polyvinyl alcohol and polyvinylpyrrolidone
  • polymethacrylates, mainly copolymers of butylmethacrylate, (2-dimethylaminoethyl) methacrylate and methylmethacrylate
  • Other polymers e.g., zein, modified starches and polysaccharides

Polymers used in delayed release film coatings

  • This category mainly includes enteric polymers such as:
  • polymethacrylate derivatives (e.g., ethylmethacrylate copolymer)
  • polyvinyl acetate derivatives, e.g., poly(viny acetate phthalate), and
  • cellulose derivatives, e.g., cellulose acetate phthalate, cellulose acetate trimellitate, hydroxypropyl methylcellulose acetate phthalate, and hydroxypropyl methylcellulose acetate succinate.

Polymers used in extended/sustained-release film coatings

This category includes the following compounds:

  • cellulosic derivatives, e.g., ethylcellulose and cellulose acetate
  • polyacrylates, such as Eudragit R series of polymers; and
  • polylactides, poly(lactide-co-glycolides and polyorthoesters

Plasticizers

Plasticizers are low molecular weight organic esters added to coatings to facilitate processing.

They modify generic properties of the polymer rendering them easy to process and or to perform as membranes.

Although many compounds, covering diverse functions like phthalates, aliphatics, epoxides (vegetable oils), etc., and even polymers, possess plasticizing properties, only few are approved for pharmaceutical use.

These include polyols such as glycerol, propylene glycol and polyethylene glycols, organic esters such as phthalates, dibutyl sebacate, and triacetin, vegetable oils/glycerides such as castor oil.

Miscellaneous additives

Additives serve a variety of purposes: Colorants are used for individualization of the product by colour.

They may be pigments, e.g., amaranth, γ-carotene, or dyes such as lakes of aluminium or opacifiers, which are used to impart opacity. Talc and titanium dioxide are mainly used opacifiers.

Fillers and anti-tack agents are used to provide coating solids to the formulation and to reduce tackiness.

They include talc, microcrystalline cellulose, glyceryl monostearate, stearic acid and magnesium stearate.

Solvents

Solvents permit uniform film formation and application over the entire substrate.

Those which have been used for film coating include water, ethanol, ethanol/water, and various other solvent/solvent mixtures.

Their selection is based on the following criteria:

(i) empirically; according to the “like dissolves like” rule;

(ii) qualitatively, based on the assessment of intermolecular forces; and

(iii) quantitatively, based on solubility parameters

Up to the early 1970s, organic solvents were the main stay of film-coating applications. However, due to increasing costs, concerns about toxicity and effects on the environment, a move to aqueous based systems was prompted.

Therefore, water is the preferred coating medium today although a few companies still use organic solvents.

How Film Coatings Are Applied

Film coating of solid dosage forms in the pharmaceutical industry is undertaken in a specialised coating equipment, such as solid coating pans, perforated pans or fluidised coater.

The process involves loading the cores into the pan, which is spun at a low to moderate speed, while a stream of hot air is pumped into the bed of rotating cores.

A solution or dispersion of the coating (polymer, plasticizer, pigment, etc) is then pumped and sprayed into the moving bed at a pace matched with the rate of evaporation of the solvent, leading to the formation of a thin film on the core surface.

This is illustrated in the infographic below:

The different steps involved in a typical tablet film coating, as it is carried out presently, can be split into several stages:

  • Preparation of the coating dispersion

This step is required in order to effectively transform coating components into a medium that can be efficiently applied onto the coating substrates.

Typically, the coating materials are dissolved or dispersed in a water, water-alcohol solutions, acetone or ethyl acetate. Organic solvents are rarely used due to the potential hazards from flammability and toxicity.

  • Droplet generation/atomization and transfer onto substrates

Atomization is the process of breaking up the coating liquid into a spray of droplets. In film coating, atomization allows the polymer coating to be efficiently and uniformly transferred onto the substrate.

The main mechanism of atomization involves pumping the coating fluid through a pneumatic nozzle. As the liquid emerges at the nozzle tip the air stream that surrounds it disrupts its flow to create a fine mist.

  • Droplet drying, coalescence and adhesion

Upon contact with the substrates, heat and mass transfer take place between the droplets, heated air and the substrate. Water evaporates from the droplets and is transferred into the air by convection. The solid payload deposits onto the surface, adheres and dries on the surface to create a film.

The choice between water and solvent-based coatings

Even though organic solvents are much easier to process, there has been a steady move away from their use in pharmaceutical coatings due to various reasons.

  • The different reasons include:
  • rising costs of solvents
  • hazards and risks due to flammability
  • regulations
  • environmental concerns

These factors, together with the availability of better designed equipment and introduction of high performance, fully-formulated coating systems, mean that aqueous coatings are by far the most widely used systems today.

Examples of Film Coating Formulae

The following are gexamples of film coating formulae based on the concepts outlined above:

Convetional coatings

High gloss coating formula

Material% w/wFunction
Sodium carboxymethylcellulose40Film former
Maltodextrin18Extender
Dextrose15Extender
Lecithin10Surfactant/dispersant
Sodium citrate2Stabiliser
Pigments & oxides10Pigments
Colours & dyes5Colorants

General purpose coating (PVA-based)

Material% w/wFunction
Polyvinyl alcohol40Film former
Polyethylene glycol 335020.2Plasicizer
Talc14.8Antiadherent
Pigments25Colorants

General purpose (HPMC-based)

Material% w/wFunction
HPMC 60640Film former
Lactose monohydrate12Extender
Polyethylene glycol 335018Plasticizer
Triacetin5Plasticizer
Pigments25Colorants

Enteric coating

Material% w/wFunction
Methacrylic acid Copolymer Dispersion (30% aqueous dispersion)16.6Enteric polymer film former
Talc4Antiadherent
Polyethylene Glycol 6000 (10% squeous solution1.6Plasticizer
Antifoam emulsion0.2Foam suppressant
Water77.6Solvent

Summary about Pharmaceutical Film Coatings

The oral route of drug administration is the most popular means by which people take medication.

It allows the development of immediate and modified-release products with unparalleled ease.

One of the commonest means of modifying drug release in oral dosage forms is the use of polymeric film coatings. Polymers are selected for this purpose because they are able to form thin membranes or coatings that are not only strong but can also impart many useful properties to the dosage form.

It is also on the basis of the functional basis of polymer coatings that they are classified, thus, we have conventional or non-functional coatings, extended release (enteric coatings) and controlled release coatings.

Sources

Pharmacentral has a strict referencing policy and only uses peer-reviewed studies and reputable academic sources. We avoid use of personal anecdotes and opinions to ensure the content we present is accurate and reliable.

Definition and Importance of Excipients in Medicinal Products

Excipients are essential but integral components of medicines. They perform several functions that are important for the safe use of medical products. However, there still remains a lot of ambiguity on what excipients are, their safety and if they should be removed from products, especially products intended for very young children. This article aims to answer all these and many other questions you have always had but did not who to ask or where to find answers.

 

What is a Pharmaceutical Excipient?

An excipient is defined as any ingredient other than the active ingredient(s) included in formulations of therapeutic goods, such as medicines, vaccines and medical devices. Excipients are also used in some cosmetic products, and nutraceutical food supplements.

In most products, excipients constitute the largest component of the formulation, being anywhere from 40 to 99+ percent by weight. This is why excipients are often referred to as ‘bulking’ agents.

If we take the example of PANADOL tablets – a commonly used pain killer medicine, each PANADOL tablet contains 500mg of paracetamol, which is the active ingredient. However, there are other ingredients in the product, as outlined below:

All the listed ingredients with the exception of paracetamol are the excipients in this product. They are typically listed in in the SmPC document (Summary Product Characteristics), which you can find by searching in a public compendia, such as the Electronic Medical Compendium.

 

Functions of Excipients in Medicinal Products

Excipients aid processing, disintegration and dissolution in the body and help protect the drug substance against unfavourable conditions, both in vivo and in vitro.

They also provide bulk, so that the product can be easily picked by the patient and conveniently used. This can be a big challenge for elderly patients or very young children. Click here to read our article highlighting the importance of empathy in new drug development. We also recently discussed the importance of dysphagia and odynophagia (swallowing problems) and how they create health inequalities if not addressed by pharmaceutical companies, which you can read through this link.

Suffice to say all the different functions undertaken by excipients work together to permit companies such as Glaxo, Astra Zeneca or Pfizer (and many others which you can see listed here) to manufacture safe, efficacious drug products that meet the quality standards set by health authorities, such as the US Federal Drug Agency (FDA), European Medicines Agency (EMA), or the Medicines and Healthcare products Regulatory Agency, and countless others around the world.

The interrelationship between excipient functionality and the pharmacological activity of the active ingredient is illustrated in this infographic below:

<img src="excipientsapiinterrelationship.jpg" alt=excipients api interrelationship">

Origins of the word ‘excipient’

There are several (often conflicting) opinions on the origin of the word, excipient. What seems certain is its etymological basis, which is the Latin word excipiēns, the present participle of excipere. Excipere essentially means ‘to take out’ and translates into, in the modern sense, ‘everything else other than’. Thus, we can envisage excipients as materials other than active principles, used to aid the processing, elaboration or manufacture of the medicinal, cosmetic or nutraceutical products.

Such ingredients include materials used to protect, support or enhance the product’s quality, safety, stability, availability, acceptability and identity, or enhance any other attribute that allow products to be used safely and effectively.

Are Excipients Inactive?

An important question today, at least within the excipients space, is whether excipients are inactive, that is, passive bystanders that do nothing other than to hung about in products. Some have gone as far as to say that these materials are unnecessarily added into products for marketing purposes and should be avoided by consumers. This is understandable.

We can say for certain that for the most past, excipients are inert (or correctly, intended to be biologically inert). But this is not without exceptions. Generally, natural or partly-natural excipients, many of which are part of the normal human diet, are biologically inert, that is, they do not elicit a pharmacological effect. The definition of natural and nature-inspired excipients is provided in this article, which you can read by clicking through this link.

Examples of such excipients include, but are not limited to the below:

Many other fully synthetic substances, such as povidone, acrylic copolymers and inorganic minerals (such as calcium carbonate and calcium phosphate) are also established as inert.

Within the pharmaceutical sector, the principal requirement for any excipient is functionality, which simply refers to fitness for a named purpose. But fitness for one use does not mean fitness for use in another, and depending on factors such as route of administration, age, patient group or dosage, a material that is fine for oral use may be hazardous when injected or inhaled.

We also know for certain that, while adverse reactions to excipients are generally rare, they still do occur, and excipients can be hazardous especially when they are not used correctly. This is why consumers are advised to always read labels and liaise with health care professionals to avoid the use of products that might contain ingredients likely to cause adverse events. Click here to read about safety assessments of excipients.

Many excipients have caused problems in paediatric and adult patients, thus the idea that excipients are inactive materials is somewhat misleading, and possibly dangerous .

Examples of well-documented, non-biologically active or ‘problem’ excipients are listed in the table below:

Excipient name/classReported issues/problemsSources
SulfitesWheezing, dyspnoea and anaphylactoid reactionFlorence A T (2010), An Introduction to Clinical Pharmaceutics, Pharm Press
Benzalkonium chlorideBronchoconstrictionFlorence A T (2010), An Introduction to Clinical Pharmaceutics, Pharm Press
AspartameHeadaches and hypersensitivity Several
SaccharinDermatological reactions; should be avoided in children with sulfar allergiesSeveral
Benzyl alcoholIn high concentrations, potentially lethal in neonatesSeveral
Various dyesCross-sensitivity, headaches, urticaria, exacerbation of asthma symptomsSeveral
LactoseProblematic in lactase deficient individualsSeveral
Propylene glycolLocalised contact dermatitits when used topically, lactic acidosis after absorptionFlorence A T (2010), An Introduction to Clinical Pharmaceutics, Pharm Press
TalcAcute respiratory distress syndromeUS FDA
Tocopherol Polyethylene Glycol (TPGS)Inhibition of P-gp mediated transportBoman et al., J Pharm Sci (2003), 92: 1250-1261
PolysorbateInhibition of P-gp mediated transportBoman et al., J Pharm Sci (2003), 92: 1250-1261
polyethoxylated castor oilInhibition of P-gp mediated transportBoman et al., J Pharm Sci (2003), 92: 1250-1261
Polyethylene glycolInhibition of P-gp mediated transportBoman et al., J Pharm Sci (2003), 92: 1250-1261

Types of Excipients

The are many different classes of excipients as are product categories. The Handbook of Pharmaceutical Excipients, an influential reference on excipients, currently lists over 400 materials. They differ by chemical properties, function and source (mineral, natural or synthetic). They may be in the form of liquids, semi-solids, solids as well as gases.

A list of different materials classified by chemical group, function and source is shown below:

Chemical GroupExamples
Hydrocolloid gumsPectin, Xanthan gum, Acacia, Agar
Mineral claysBentonite, Kaolin
Cellulose polymersHypromellose, Ethylcellulose, Microcrystalline cellulose
StarchesMaize starch, Tapioca starch, Potato starch
Amino acids & ProteinsAlbumin, Glycine
Sugars & PolysaccharidesDextrose, Polydextrose, Fructose, Lactose
Vinyl polymersPolyvinyl alcohol
MineralsCalcium carbonate, Magnesium carbonate, Sodium chloride
PetrochemicalsLiquid paraffin
SiliconesDimethicone, Cyclomethicone, Simethicone
PolyolsSorbitol, Mannitol, Maltitol, Xylitol
Acrylic polymersCarbomers
Organic & Inorganic compoundsCitric acid, Ethylparaben
Triglycerides, Oleochemicals & DerivativesGlyceryl monostearate, Oleic aicd, Magnesium stearate
GlycolsPolyethylene glycol, Polyethylene oxide
Alkoxides & SilicatesSilica, Calcium silicate
FunctionExamples
Emulsifiers & SurfactantsSodium lauryl sulfate, Tween 80, Hydrogenated castor oil
Fillers & DiluentsLactose, Microcrystalline cellulose, Maize starch, Calcium phosphate
Film coatingsHypromellose, Ethylcellulose, Polyvinyl alcohol
BindersMaize starch, Povidone, Sodium carmellose
Sweeteners & FlavoursSucralose, Xylitol, Acesulfame K, Strawberry flavour
PlasticizersSorbitol, Triethylcitrate, Glycerine
Drug release modifiersPolyethylene oxide, Hypromellose, Ethylcellulose
Solvents & SolubilisersEthanol, Cyclomethicone
Viscosity modifiers & Suspending agentsXanthan gum, Sodium carmellose
Antioxidants & Stabilising agentsVitamin C, Vitamin E
Vaccine adjuvantsAluminium hydroxide
Gelling agentsCarbomers, Pectin, Carrageenan
Glidants, Lubricants & Anti-sticking agentsSilica, Magnesium stearate, Talc
PreservativesEthylparaben, Potassium sorbate, Benzalkonium chloride
Disintegrating agentsStarch, Sodium starch glycolate
Buffers & pH modifiersSodium hydrogen carbonate, Phosphoric acid
Excipient SourceExamples
Chemical synthesisSodium lauryl sulfate, Tween 80, Carbomer
MineralsTalcum, Kaolin, Calcium carbonate, Sodium chloride
Plant derivedMaize starch, Glucose, Mannitol
Animal derivedLactose, Gelatin
Products of fermentationXanthan gum

Excipients Used in Oral Solid Products

The oral route, and specifically, oral solids (mainly tablets, capsules and powders) is the most widely used route for administration of pharmaceutical products. Some studies indicate that the oral route accounts for over 80% of all medicines administered. Here is a list of the top 20 excipients most commonly used in oral solid dose formulations, including granulation binders, diluents for tablets and capsules, disintegrants, lubricants, antiadherents, glidants, colouring agents and flavours.

Common NameMain Function
AlginatesBinder and disintegrating agent
Calcium carbonateFiller, diluent, pigment & source of calcium. Opacifier & colorant
Calcium phosphate (all grades)Filler, diluent, pigment & source of calcium
GelatinCoating agent & binder
Croscarmellose sodiumDisintegrating agent
CrospovidoneDisintegrating agent
Povidone (PVP)Tablet binder
Silicon dioxideGlidant
Sodium starch glycolateDisintegrating agent
Dextrose (glucose)Filler, diluent & binder
Functional & Aesthetic coatingsAesthetics & modified drug release
Lactose (Spray-dried, monohydrate & anhydrous)Filler & diluent
Magnesium stearateLubricant
MannitolFiller & diluent
Native & modified starchesFiller, binder & disintegrating agent
SorbitolFiller & diluent
Microcrystalline celluloseFiller & dry binder
Sodium bicarbonateFiller & alkalising agent
HypromelloseBinder & modified-release agent
Sodium chlorideDiluent & tonicity agent

Excipients Used in Oral Liquid Products

Suspensions and oral liquids are an important dosage form particularly in children and the elderly or when drug products have poor solubility or cannot be formulated into tablets or capsules.

The top 12 excipients used in pharmaceutical suspensions, including syrup or dry suspensions are shown below.

Xanthan gumSuspending agent and viscosity modifier
Polyethylene glycol (mainly PEG 400)Solvent and Co-solvent
CarboxymethylcelluloseSuspending agent and viscosity-increasing agent
Flavours (mainly citrus, strawberry & vanilla)Taste masking and flavour enhancement of bitter products
GlucoseSweetener
MaltitolSuspending agent & diluent
SucroseSuspending agent, viscosity modifier and sweetener
EthanolSolvent & Co-solvent
SorbitolSweetener, Suspending agent & diluent
SucraloseTaste masking and flavour enhancement of bitter products
Propylene glycolSolvent & Co-solvent, flavour enhancer
PolysorbateSurfactant and dispersing aid
Parabens (All)Preservatives
Acesulfame PotassiumSweetener
Sodium/Potassium BenzoatePreservative

Excipients Used in Topical Products

Administration of medicines through the skin is referred to as ‘transdermal drug delivery’. It has grown in importance in recent decades owing to several advantages, including:

  • improved patient adherence
  • convenience
  • options for sustained and controlled release
  • avoidance of gastric irritation, and
  • avoidance of first-pass effect

Even though excipients used in topical products are not intended to be swallowed, they must be safe and comply with pharmacopoeia standards and meet expected quality standards.

A list of the top 15 commonly used topical excipients include:

ExcipientFunction
Carbomers (all)Viscosity increasing agents, Suspending agents, Emollients & Moisturizer
Petrolatum & Mineral OilsEmollient & Skin conditioner, Carrier
GlycerinMoisturiser and Co-solvent
Glyceryl MonostearateEmulsifier and Co-emulsifier
Cetostearyl AlcoholEmollient, Skin emulsifieir & Viscosity-increasing agent
Polyethylene GlycolGelling agent, Solvent & Co-solvent
Parabens (Methy, Propyl & Butyl)Preservatives
Tocopherol (Vitamin E)Antioxidant & Skin conditioner
Dimethicone & CyclomethiconeEmollient, Skin conditioner & Carrier
Waxes (Bees, Microcrystalline & Others)Refatting & Stiffening agent
Vegetable OilsEmollient & Carrier
Isopropyl MyristateEmollient, Skin penetrant & Carrier
Glyceryl MonooleateBioadhesive material, Emollient & Emulsifier
Hard FatsSuppository base
PolysorbatesSurfactants, Dispersants & Co-emulsifiers

Excipients Used in Injectable Products

The parenteral route covers all products administered by injection directly into the body’s issues.

This class of medicines can be divided into three groups:

  • injection into the skin (subcutaneous, intramuscular, & intracutaneous),
  • intraveneous infusions, and,
  • peridural and subarachnoid routes

Injectable products offer numerous advantages over other routes of administration, including precise and adjustable dosing, predictable bioavailability, and fast onset of action.

In acute-patient-care settings, injectable drugs are the main forms of drug administration.

Since injections introduce substances directly into the body bypassing many of the body’s defences, there are extra safeguards expected from excipients.

One of these is that they should be sterile, and if the volume of the product is large (>100ml), they should be free of pyrogens and endotoxins (fragments of bacterial cell walls).

A list of the 15 most commonly used excipients in injectable products and the roles in the formulation is below:

Excipient nameFunction
Arachis oilCarrier & Oleaginous vehicle
CresolPreservative
Benzyl alcoholPreservative
Dextrose (Glucose)Bulking agent & Cryoprotectant
EDTAChelating agent
Ethyl oleateCarrier and Solvent
Human serum albuminStabiliser
Hydrogenated castor oilSurfactant & Solubilising agent
Lecithin (and other phospholipids)Surfactant & Solubilising agent
Macrogol 15 HydroxystearateSurfactant & Solubilising agent
GlycerolCo-solvent
PolysorbateSurfactant & Solubilising agent
PolysorbatesSurfactant, Solubilising agent & Dispersant
Potassium or Sodium PhosphateBuffering agent
Sodium chlorideTonicity adjusting agent
Sodium hydroxidePH adjusting agent
Sodium citrateBuffering agent
Sodium metabisulfitePreservative & Antioxidant
Polyethylene glycolSolvent
Zinc chlorideTonicity adjusting agent
Hydrochloric acidPH adjuster

Excipient Safety Assessment

The issue of excipient safety is a major point of contention in public discourse, and possibly one of the major sources of misinformation on medical products.

Before any material can be used in a product as an excipient, regulatory agencies require manufacturers to undertake rigorous testing and guarantee that it is safe for use.

This has not always been the case – in fact, in the early days of the pharmaceutical industry, excipients were considered pharmacologically inert and rarely regulated.

This lack of control lead to many issues, including significant harm to the public. You can listen to our podcast on this through this link.

<iframe src=”https://anchor.fm/pharmacentral/embed/episodes/Introduction-Catastrophes-that-have-helped-shape-pharmaceutical-regulation-eq9ea1″ height=”102px” width=”400px” frameborder=”0″ scrolling=”no”></iframe>

Over the years, as products have become more complex and the ingredients used in them even more elaborate, safety testing of excipients is now mandatory.

Suffice to say, regulatory agencies, such the US FDA, MHRA and EMA, have approved lists for excipients, either in form of pharmacopoeia monographs or databases (e.g IIG Database, Dictionnaire Vidal or Japanese Pharmaceutical Excipients compendium) as well as standards for manufacture (e.g Good Manufacturing Practices) and distribution/handling (Good Distribution Practices) that manufacturers must comply with.

Read about excipient safety assessment here.

The bottom-line is that the pharmaceutical industry works through multiple checks and balances to ensure that quality and safety requirements are adhered to before any material can be used in a drug product.

 

Frequently Asked Questions About Excipients

What criteria are used to select excipients?

The main criteria for selection of an ingredient is functionality. Examples of the different functions were outlined above, but generally include:

  • maintainance of a product’s integrity (containment, stability, freedom from contamination, etc)
  • making up the volume
  • aiding in the release characteristics of the active ingredient
  • facilitating identification, etc.

But in addition to functional suitability, the excipient must be compatible with other excipients and the active. Incompatibilities can compromise product quality and safety of users.

Thus, the most suitable excipients for the job are selected, taking care that those that are selected have a genuine function in the product.

Indeed, product manufacturers are now required to state and justify the role of excipients in the product. It is no longer acceptable to add a material that does not have a clearly established and validated function.

For example, a preservative should not be included in sterile, single-dose products, such as injections or eye drops. As a general rule, only excipients that perform a needed function are required to be included int he formulation.

Which materials can be excipients?

In the USA, only materials officially recognised in the United States Pharmacopoeia-National Formulary (USP-NF) and/or listed in the US FDA Inactive Ingredients Database (IIG Database) can be used in pharmaceutical products as excipients.

The USP-NF defines the quality requirements for excipients in form of a monograph while the IIG Database shows which material is already in use and the types of products its used in.

The Europe Union does not have a bloc-wide list of approved excipients although individual countries have compendia that list approved materials for their jurisdictions.

There is however, a European Pharmacopoeia, which has excipient monographs similar to the USP-NF.

An important question is whether materials without a monograph or those not listed in official compendia be used as excipients?

Generally, a material not officially listed in a pharmacopoeia or recognised in compendia may still be used in pharmaceutical products. In deed there are various such products, including for example, butylene glycol and several others.

However, regulatory agencies require detailed assessment and evidence of that material’s safety, toxicity and quality profile. This can be time-consuming and expensive process, and for this reason, few manufacturers dare go down this route, preferring to use established/proven materials as excipients.

Which excipients should not be used in infants?

In 2008 the EU passed Regulation 1901/2006 on medicinal products for paediatric use, following which the European Medicines Agency published recommendations for pharmaceutical formulas intended for children (0-18 years old).

A few standout excipients in this regulation include:

ExcipientFunctionRegulatory Recommendation
Benzyl alcoholPreservativeNot to be given to neonates
EthanolSolvent and preservativeBest avoided in children under years old. The WHO proposes the following:Not more than 0.5% for children under 6 years old Not more than 5.0% for children between 6 and 12 years oldNot more than 10.0% for children over 12 years old
Propylene glycolSolventAvoid in children under 4 years old
Colouring agentsColouring & maskingShould be avoided in all paediatric products unless absolutely necessary. AZO dyes are banned

 

Which companies manufacture excipients?

Excipient manufacturers are a diverse group of companies, from public to private, small to large. You can see our compilation of the most innovative excipient manufacturers or the size of the excipients market by following through these links. Here, we provide a selection of the most important excipient companies currently:

CompanyCountry HQProduct Focus
BASFGermanyIbuprofen, Omega-3, Polxamer 188, Hydrogenated Castor oil, Povidone & Lactose
DupontUnited StatesSilicones & Elastomers
IFFUnited StatesMicrocrystalline cellulose, Flavours and Fragrances
ColorconUnited StatesFilm coatings
Evonik AGGermanySilica, Polymers, Carbomers and Antimicrobials
CARGILLUnited StatesCarbohydrate and polysaccharides
Lubrizol CorporationUnited StatesCarbomers
ShinEtsuJapanSilicones, Cellulose polymers
CrodaUnited KingdomOleochemicals
GattefosseFranceOleochemicals
Roquette FreiresFranceCarbohydrate and polysaccharides
Tereos Sugars & SweetenersFranceCarbohydrate and polysaccharides
JH NanhangChinaVinyl polymers (Povidones)
SolvayFranceSodium bicarbonate
AsahiJapanPolysaccharides & microcrystalline cellulose
Abitec CorporationUnited StatesOleochemicals
Angus Chemical CompanyUnited StatesBuffers and salts
Dow ChemicalUnited StatesPolymers & cellulosics
Meggle GroupGermanyPolysaccharides & microcrystalline cellulose
DFE PharmaThe NetherlandsPolysaccharides & microcrystalline cellulose
Tate & LyleUnited KingdomPolysaccharides & microcrystalline cellulose
CP KelcoUnited StatesHydrocolloids
AshlandUnited StatesFilm coatings, Povidones
JRS PharmaGermanyPolysaccharides & microcrystalline cellulose
SasolSouth AfricaOleofins
Eastman Chemical CompanyUnited StatesPolymers
Merck KGAAGermanyDiversified chemicals, Pigments & buffers
IOI OleochemicalsGermanyOleochemicals
FirmenichSwitzerlandFlavours and fragrances
Kerry GroupIrelandLactose, coatings & flavours
SPI PharmaUnited StatesODT systems, mannitol
CabotUnited StatesSilica
BudenheimGermanyCalcium salts
ImerysFranceTalc, calcium salts & kaolin
HallstarUnited StatesOleochemicals
BeneoGermanyPolysaccharides
InolexUnited StatesOleochemicals
Dr. Paul LohmannGermanyCalcium salts
Lonza AGSwitzerlandPreservatives & capsules
Nippon GohseiJapanPolyvinyl alcohol
CelaneseUnited StatesSweeteners
British SugarUnited KingdomSugar
WackerGermanySilicones and silica
WH GraceUnited StatesSilica
SeppicFranceFilm coatings
Novo NordiskDenmarkPreservatives
Mingtai Chemicals CoChina (Taiwan)Microcrystalline cellulose
AgranaAustriaStarch
CalumetCanadaPetrochemicals
NissoJapanHydroxypropylcellulose

 

Although PharmaCentral.com does not manufacture excipients, we have distribution arrangements and partnerships with many reputable firms. If you have any questions on any materials or would like to sample or purchase get in touch through here.

Should I avoid non-medical products that use excipients?

Since many excipients are chemically-derived industrial ingredients they may not sit well with some consumers of dietary nutraceutical products who prefer natural, eco-friendly and ‘clean’ labelled.

What’s useful to remember is that pharmaceutical excipient manufacturers mainly focus on achieving and maintaining functional performance and safety, which requires materials to undergo extra levels of processing and purification.

Ultimately, the choice to avoid dietary products with excipient-grade ingredients is down to personal choice and what people believe best suits their values and motives.

Where can I obtain more information on excipients?

There are many authoritative sites and places to get information on excipients:

You can of course, use our own PharmaCentral Products pages – where we have monographs of the most important and commonly used excipients listed. You will find all the information you need regarding any material. And the good thing is that it is 100% free.

 

Cited Literature and Sources Used

To ensure our content is accurate and scientifically sound, Pharmacentral implements a strict referencing policy. We only use peer-reviewed studies and reputable academic sources and authors.

Elder DP, Kuentz M, Holm R. Pharmaceutical excipients – quality, regulatory and biopharmaceutical considerations. Eur J Pharm Sci. 2016 May 25;87:88-99. doi: 10.1016/j.ejps.2015.12.018. Epub 2015 Dec 14. PMID: 26699228.

Fabiano V, Mameli C, Zuccotti GV. Paediatric pharmacology: remember the excipients. Pharmacol Res. 2011 May;63(5):362-5. Doi: 10.1016/j.phrs.2011.01.006. Epub 2011 Jan 15. PMID: 21241804.

Alison Haywood, Beverley D Glass. Pharmaceutical excipients – where do we begin? Australian Prescriber (Accessed October 2021).

Excipients – an overview (Science Direct)

Overview of pharmaceutical excipients used in tablets and capsules. (Accessed October 2021).

 

Back to Top
Product has been added to your cart