Review Article | Open Access

Principles, Applications and Limitations of the Liquisolid System of Drug Delivery: A Review

    Daniel Ekpa Effiong

    Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, University of Uyo, Nigeria

    Godswill Chukwunweike Onunkwo

    Department of Pharmaceutical Technology and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, University of Nigeria, Nsukka, Nigeria


Received
19 Apr, 2024
Accepted
07 Jun, 2024
Published
08 Jun, 2024

The liquisolid system of drug delivery continues to find usefulness as a potent means to circumvent solubility and dissolution challenges commonly encountered during drug formulation and medicine development in the pharmaceutical industry. Originally invented by Spireas, the technique has gained acceptance and improved over the years, in the types of excipients used, nature and class of drug formulated and its applications not only in transforming lipophilic and poorly soluble drugs to solid dosage forms with enhanced bioavailability, but also now gaining popularity in modified delivery, photoprotection and minimizing the effect of pH on drug release. This review thus highlights the foundational principles of the liquisolid technique, examines current methods employed and modern applications of this technique. It also gives present knowledge of this delivery technique and its limitations. A systematic approach to the search of published literature in standard databases (Pubmed, Google Scholar, Researchgate) was carried out using specific search terms and operators that provided available works as information sources for this review. Critical evaluation of obtained literature from the database was adopted to extract data on the principles and current applications of the liquisolid systems beyond its intended initial use. The prevailing mechanism for enhancing bioavailability via the technique is improved wetting and drug presentation in well dispersed or soluble state; minimal pH influence is based on the phenomenon of saturation solubility of the drug in the nonvolatile vehicle; and photoprotection is due to high refractive and diffraction capacities of the coating materials used. This work concludes by presenting the identified and potential limitations of the liquisolid technique. It gives expert opinions on how these are being circumvented and future perspectives on this ever-unfolding promising drug delivery strategy. It will be an invaluable contribution to current literature in drug development research employing liquisolid techniques for drug delivery.

Copyright © 2024 Effiong and Onunkwo. This is an open-access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. 

INTRODUCTION

The growing interest in the use of liquisolid systems in drug formulation and delivery studies has been related to its invaluable contribution to circumventing poor drug solubility, enhancing bioavailability and presenting several delivery strategies with desirable therapeutic outcomes1.

Solubility, dissolution and bioavailability rank highly as parameters considered during the development of potential therapeutic agents for use as medicines because sooner or later, all medicines must be in solution to be absorbed or reach desired drug concentration in systemic circulation for therapeutic efficacy2,3. However, the successful transformation of a potent drug into medicine is often beset by challenges of these physicochemical parameters or those related to them. Solubility and dissolution rate of a drug can thus significantly influence its absorption, determine its bioavailability in the circulatory system and expectedly influence the expected pharmacological response1,4.

Drugs with poor water solubility are generally formulated and used as high-dose medicines with high dosage regimens so that, after administration (for example via oral route), the fraction of it that goes into solution could reach therapeutic plasma concentrations5. Sadly, high-dose medicines could mean exceeding the safety limits of the drugs and approaching toxicity, escalated untoward effects and increased production costs. As medicinal chemists and formulation scientists continually search for and employ strategies to improve the rate and amount of poorly soluble drugs that go into solution, the liquisolid system of drug delivery is now gaining attention. Thus in this paper, this technique will be well explored in terms of its principles, its application and limitations associated with it. This work will also address how these limitations are being overcome. First, a background of poor drug solubility will be presented, how widespread the problem is and common strategies that have been employed to improve it will be highlighted.

Generally, when discussing solubility and dissolution studies of drugs, the solvent of interest is usually an aqueous medium or one that is water-miscible because it is the main component of human body fluids where administered drugs will dissolve and subsequently be absorbed. This also explains why it is also the widely used solvent for liquid pharmaceutical formulations. In medicine development and manufacture of new chemical entities and candidates, the unprecedented incidence of low aqueous solubility is a major problem. One possible reason is that most drugs developed as medicines are either weakly acidic or weakly basic, thus having poor aqueous solubility. Yet these drugs must be present in an aqueous solution at the site of absorption to be taken up by the body and utilized.

The recent unlimited leap in the sphere of drug discovery resulting from the interplay of combinatorial chemistry, high throughput screening and computer-aided-drug designs and modeling has made possible rapid drug synthesis, optimization and drug-receptor simulation studies in silico6. This progress has churned out a number of drug molecules that can effectively and selectively interact with receptors (or ligands) within the body to elicit biological activity. These resulting available drug molecules or candidates, however, are highly hydrophobic, show a slow dissolution rate and are poorly water soluble2,7. No doubt, an indication of the magnitude of solubility challenges in the development and manufacture of medicines.

CHALLENGE OF POOR SOLUBILITY- HOW EXTENSIVE!

On the basis of their dissolution, water solubility and permeability in the intestinal tract, medicines are being classified by the Biopharmaceutical Classification System. Using this system and well-defined parameters, medicines are classified into Class I (well absorbed; that is have high solubility and permeability) Class II (limited solubility, high permeability low solubility) Class III (limited permeability, having high solubility but low permeability) and Class IV (poorly absorbed, having both solubility and permeability as being low)8. The parameters as defined of the dose of the medicine as follows; solubility (in 250 mL or less of water over a pH range of 1-8), dissolution rate (in dissolution apparatus 1 at 100 rpm or apparatus 2 at 50 rpm, in a volume of 900 mL buffer solutions of 0.1 N HCl/pH 4.5 buffer/pH 6.8 buffer with no enzymes) and permeability (using absorption rate of the prescribed dosage and is stability in the stomach).

Reportedly, some 4 out of every 10 medicines already in the market are poorly water soluble and about 90% of newly approved drug candidates in development are poorly water soluble. They fall in Class II and IV of the Biopharmaceutical Classification System and many of these fail to progress in the line to realization as medicines it is on record that almost 70-80% of the synthesized chemical entities have been classified as having poor solubility2,3,7. These statistics are clear indications of how extensive the issue of poorly water-soluble drugs and this is understandably a matter of serious concern to the formulation scientist, who constantly attempts to explore formulation strategies to circumvent this challenge but positively influence drugs to achieve medicines with enhanced delivery.

Solubility is not to be interchanged as dissolution or dissolution rate. Dissolution is the amount of a substance that goes into a solution at a time. Dissolution rate is described as the amount of a substance (drug) that goes into solution over a given time but solubility defines the total amount of the same drug that can go into solution under specified conditions2. Solubility is not a function of time. As a general rule, poor aqueous soluble substances have a slow dissolution rate, especially in coarse dispersions but an exception is hydroxypropyl methylcellulose, for instance, though highly soluble excipient for drug formulation has a very slow rate of dissolution because it takes a relatively longer time to be fully wetted to go into solution9. In the same vein, different dosage forms or delivery systems go into solution to exert therapeutic action at varying rates depending on how they were designed to function. Figure 1 presents different dosage forms and their rate of dissolution in increasing order in the direction of the arrow.

Strategies for improving solubility and dissolution- the associated limitations: For poorly soluble drugs or hydrophobic ones, several conventional methods and new formulation techniques have been employed by researchers and medicine manufacturers to overcome poor solubility and dissolution. These techniques and strategies as found in the published literature on improving solubility are numerous and can be grouped into physical techniques (by modification of particle sizes to micro sizes and nano-based systems thus increasing particle-specific surface area for improved dissolution rate); chemical modifications (use of different salt form, pH adjustment) and other formulation techniques. Table 1 presents a summary of such approaches, their application in particular drugs and their respective limitations.

Challenges associated with other techniques of improving poor solubility: As reflected in the table, these strategies to improve solubility and dissolution of drugs having challenging solubility profiles come with their limitations. Beyond those listed, some methods are even capital intensive, require sophisticated equipment, or demand technology. This is especially so in methods such as soft gelatin encapsulation, where poor soluble drugs are prepared in aqueous or oily solution form and then encapsulated as advanced solid dosage forms.

Fig. 1: Delivery systems showing increasing dissolution rate

Table 1: Summary of the techniques for enhanced solubility and their limitations
Solubility enhancing
techniques
Process involved Application Limitation Reference
Chemical modifications
Salt formation Conversion of drug to a salt
form that is water soluble
Diclofenac manufactured
as salts of sodium/potassium
Formation of new
product with likely
new properties and
Patil et al.10
  Amlodipine as
besylate salt
Theophylline,
Barbiturates
requiring safety
assessment
Samineni et al.8
pH adjustment Used to enhance solubility
of ionizable organic
medicines in solutions
Use of buffers
Aspirin tablets are used
as dissolving tablets by
preparing with sodium
bicarbonate and citric
acid as pH adjusting
buffers
Limited application to
drugs that are weakly
acidic or basic
Kalepu and
Nekkanti11
In situ salt formation Ciprofloxacin infusions
formed by adjusting pH
with lactic acid
 
Prodrug Approach converts a
pharmacologically active
substance into an inactive
one that gets activated in
the system after
absorption through
enzyme or chemical
action
Paclitaxel delivered as
Paclitaxel-disulfide
prodrug, for improved
anticancer activity
Chalcone delivered as
Chalcone-phosphate
Requires certain
specific conditions
for the active drug
the active drug to
be released from
the prodrug e.g.,
presence of an
hydrolytic enzyme
to release the drug
after administration
New moieties may
come with newer
physicochemical
properties
Jornada et al.12
Physical methods
Solid dispersion Hydrophobic drugs are
dispersed in solid
excipients matrix which
is hydrophilic
Delivery of Paclitaxel an
anticancer, delivery of
curcumin and curcumin
complex for acetylcholine
esterase inhibition and
anticancer, respectively
Limited commercial
application because
of poor stability on
storage and limited
understanding of
product solid-state
structure
Wu et al.13,
de Sá et al.14
and
Choi et al.15
Encapsulation of liquid
drug using soft gelatin
Drug in liquid form is
coated or protected
by capsule or covering
It has the highest and
most consistent
bioavailability, mainly due
to the fact that the drug is
already in solution
Lipid soluble vitamins Expensive and
uncommon
technology
Arango-Ruiz et al.16
Self nanoemulsion
(SNEDDS), self micro-
emulsifying drug
(SMEDDS) or self-
emulsifying drug
delivery system
(SEDDS)
Drug is prepared to be
carried within rapidly
forming emulsification
globules formed when in
contact with the fluid in
the gastrointestinal tract
The size of the globules
is used to classify the
delivery system as SEDDs
(if macro) SMEDDS (if
micron sized or SNEDDS
if nanosized
Atorvastatin self-
globules of emulsion
had better
Cyclosporine an
immunosuppressant
the management of
benign prostatic
hyperplasia
High cost and
complicated
procedure
Shen and Zhong17
Kalepu and
Nekkanti11 and
Agubata et al.18
Solid lipid nanoparticles Drugs are held within
the lipid matrices
Site-specific delivery
achievable
Ofloxacin was formulated
as SLN fenofibrate
successful incorporation
into SLN
High cost, a process
complicated
Xie et al.19
Microencapsulation A packaging engineering
te unique in that drugs are
coated with thing outer
polymer layer as shells to
form microcapsules
Delivery of doxorubicin and
heparin using chitosan for
encapsulation in managing
human papilloma carcinoma
Selecting suitable
coating polymer
requires good
technical skill
Chaturvedi and
Sharma20
Size reduction
Micronization and
nanonization
Micronization: Drug is
sized reduced by milling
to micrometer size ranges
precipitation to obtain
submicron size ranges so
as to increase the surface
area and enhance solubility
Micronization is employed in
the delivery of ibuprofen to
improve digestive absorption
and bioavailability

Micronization: High
tendency for
micronized hydrophobic
drug particles to
agglomerate. Physical
and thermal stress on
the drug, thus degrading
it Limited application

Han et al.21
Nanosuspension

Nanosuspension:
Drug suspension in
surfactant is wet milled and
homogenized to obtain a
colloidal dispersion of the
drug element at submicron
sizes with the surfactant as
a stabilizer

Nanosuspension:
Delivery of megesterol acetate
(Megace ES®) for rapid
dissolution and bioavailability
Delivery of fenofibrate tablet
(Tricor®) for improved (Tricor®)
for improved dissolution and
bioavailability
Nanosuspension:
Formulation design and
stabilization is complex
No controlled release
Kanikkannan6
Use of technological advancement
Inclusion complexation Involves inserting a
nonpolar molecule
(or poorly soluble drug)
into the cavity of another,
the host/companion
molecule (eg cyclodextrin)
and held by non-covalent
intermolecular forces
Delivery of poorly soluble
vericonazole (Pfizer, USA) as
intravenous solution for
systemic fungal infection
Delivery of omeprazole
(Betafarm, Germany) as
tablet for improved
bioavailability.
Improved delivery of nicotine
as sublingual tablet (Pharmacia,
Sweden)
Quantity of drug loading
and dosage form design
may be limiting factors
Challenge of scale up of
manufacturing process
Kanikkannan6 and
de Miranda et al.22
Crystal engineering Controlled crystallization of
drugs so as to produce
powders with crystal form
(crystalline or amorphous),
high purity, surface energy-
defined particle size
distribution and crystal
habit by manipulating
different solvents or change
in the stirring or adding other
components
Delivery of chloramphenicol
palmitate as meetastable
polymorph for enhanced
solubility and absorption
Delivery of oxytetracycline
using form B polymorph for
improved solubility
Savjani et al.3
Hot melt extrusion Process of dissolving poorly
soluble APIs into a polymer
matrix known to form an
amorphous solid dispersion
Delivery of oleanolic acid
for improved dissolution
Challenge to achieve
high drug loading
Not suitable for
thermosensitive
drugs and polymers
Limited choices of
pharmaceutical-grade
polymers and
surfactants
Gao et al.23
Lipid-based
microparticulate drug
delivery system (solid
lipid microparticles)
Drug delivery using
microsized drug carriers
with solid lipid matrices
such as fatty alcohol,
solid wax, fatty acids or
glycerides
Delivery of artemether
for improved improved
solubility, dissolution
permeability and
bioavailability
delivery of dihydroartemisinin
-piperaquine phosphate for
malaria
Potential for interaction
of lipid carrier with drug
structure or its
degradation limited
studies on the use of
lipids with high melting
points
The use of temperature
may limit the use of
thermolabile drugs
Agubata et al.24
Amarachi et al.25

Fig. 2: Classification of the liquisolid system

The liquisolid technique, as another promising strategy used to circumvent poor solubility, dissolution rate and bioavailability in medicine development, is now gaining wider attention and application as a drug delivery system. From its early introduction about 3 decades ago, much is now being made available by researchers who have explored this area of research in delivering drugs with poor water solubility. An up-to-date understanding of this technique is therefore critical for its use and application in improving the delivery of new and existing challenging drug molecules.

Liquisolid technique: Principle: First discovered and propagated by Spireas in his doctoral thesis and when employed to deliver steroids such as prednisolone, the liquisolid system is an advanced technique of solid oral dosage form for enhancing solubility and dissolution of poorly soluble drugs26,27. It has reportedly been valuable in overcoming the limitations associated with the other approaches for improving drug bioavailability such as hydrophobic agglomeration (common with micronizing), poor stability (associated with salt formation) uncommon technology (peculiar to nanotechnology-driven techniques) and the influence of pH.

The liquisolid technique has been considered an outgrowth of the powdered solution technology that was used in the delivery of liquid medication28. In its most simplistic definition, it is an approach of drug delivery where in a poorly soluble drug is formulated as liquid medicine but delivered in a solid dosage form. The poorly soluble solid drugs are initially made into liquid medicines by either dispersing or dissolving them in non-volatile solvents and then converted to flowable powders or granules that are non-adherent but compressible. This transition from liquid state to solid powders or granules is made possible through the use of carriers and coating materials. A simple illustration of the classification of the liquisolid system is shown in Fig. 2.

The formulation techniques and the resulting presentation further reveal the variety brought into this delivery system.

Liquisolid compact: These are liquisolid systems that are presented as tablets or capsules. They come as sustained or immediate-release formulations and have the inclusion of other excipients (disintegrants, lubricants, binders, etc.,) that make them readily compacted or encapsulated.

Liquisolid microsystem: These liquisolid systems are presented in multipellets or microsized granules or beads and encapsulated, usually contains adjuvant polyvinylpyrrolidone (PVP).

Theoretical principle: The liquisolid approach to drug delivery is based on the following fundamental pharmaceutics principles:

  Principle on flowability and compressibility: A given porous powder mass with acceptable flowability and compressibility can absorb (hold) a limited amount of liquid. This amount of retainable liquid is carrier-specific, coating material-specific and liquid-specific. This implies that the amount of liquid paraffin, for instance, that microcrystalline cellulose (a carrier) can hold and be flowable will be different if the liquid is changed to glycerol or the carrier is replaced with aerosol
  Based on this principle, in liquisolid systems, drug(s) in the liquid state are converted into apparently dry powder, with acceptable flow and compressible properties by simply introducing the liquid medication (either by blending with or spraying onto) selected excipient carriers and coating materials29
  That limit to the amount of liquid the powder mass can hold is a function of its specific surface area and absorptive capacity. Materials having higher values of specific surface area or absorptive capacity can hold a larger volume of the liquid medication. Hence, this principle provides guidance on the selection of powders for use as carriers and coating materials for optimal delivery. It must be mentioned too that, as reported in the literature, the incorporation of binders such as hypromellose or povidone reduced the amount of a blend of carrier-coating agents used to obtain flowable and compressible powder mass29

Principle of improving dissolution and solubility: The dissolution rate and solubility of a poorly soluble drug can be improved by increasing its wettability and surface availability to the dissolving medium. Wettability and surface availability can be increased by reducing the contact angle of the drug substance and the solvent using surface-acting agents or employing liquids with better wetting properties. When the drug (to achieve improved wetting) is thus dispersed in a solvent that is water miscible, it enhances faster dissolution and overall solubility of the drug in the aqueous medium. This principle finds its value in providing needed guidance on the choice of appropriate solvent for the optimal formulation of the liquid medicine in liquisolid delivery. Liquid medicine can be presented as a suspension, solution or emulsion of the drug28.

Principle of impact of excipient ratio: The compressibility and mechanical properties of the formulated solid dosage form (compacts or tablets) are dependent on the properties of the excipients (carrier in this case) used and their ratio to the coating material in the formulation. This highlights the importance of the ratio of carrier material to coating material in achieving a liquisolid compact having desirable properties. In literature the ratio of carrier to coating agent of between 10-20 to 1 but recent works have reported even higher ratios even up to 50:130,31.

Basing this technique of development of medicine on such foundational principles of pharmaceutics gives it some merit. Table 2 presents some of these alongside some disadvantages.

Mathematical concepts: To achieve a liquisolid system having the desired flow properties and compressibility, the liquid medication and excipients (the carrier and the coating agents) must be used in the right proportions. Some mathematical concepts are employed for accurate determinations of these proportions as follows.

Spireas and Bolton in 1999 as reported by Tiong and Elkordy32, introduced an empirical method to determine the amount of each excipient to be used in the formulation of the liquisolid compact on the basis that each excipient in a liquisolid system (carrier or coating material) possessed a flowable liquid-retention potential: For the carrier (Φ) and the coating materials (φ). This also holds when using different mass ratio mixes of carrier and coating materials. The values are required to calculate other necessary

Table 2: Merits and demerits of the liquisolid system
Merits of the liquisolid systems Demerits of liquisolid system
Versatility in delivery technique and useful for enhancing solubility
of poorly water-soluble drugs, slightly soluble ones and practically
insoluble drugs. It can now be applied to even soluble drugs to
sustain the release
In many cases, mainly applicable to drugs that come as low-
dose medicines. Drugs with higher dose strength will require
a larger amount of liquid for preparation of drug as liquid
medication, thus requiring more excipient use with results of
very large difficult-to-swallow tablet
Relatively cheap and simple technology to use in drug delivery Seemingly limited choice of dissolving/dispersing solvents
Other liquid vehicles are being considered for use
It leverages the tableting and encapsulating technology as the
popular oral route of administration
It requires necessary expertise in the application of the
mathematical concepts in determining the appropriate
quantities of excipients to be used in the formulation
It can be easily scalable and hence has good industrial application The non-volatile solvents used in preparing the liquid
medication can negatively influence the mechanical
strength of the compacts manufactured
Application of technique now expanded to modified delivery
system, circumvent photosensitivity reactions in drugs and
influence of pH on drug solubility during manufacture
The drug formulation though solid is actually a drug in solubilized
or well-dispersed liquid form hence allow faster wetting and
resultant enhanced dissolution
 

parameters of the system. The flowable liquid retention potential of a carrier, for example, is ascertained by gradually incorporating a liquid medication onto its bulk powder of the carrier, blending and then determining the flow properties of the bulk powder. This is repeated until when the optimal flow properties are obtained. The amount of liquid held at that point by the carrier is its flowable liquid retention potential. A particular flow property of significance and being used for such determination is the angle of the slide. The same is determined for the coating material. So the liquid load factor, Lf , the flowable liquid retention potential of the carrier, Φ, the same parameter for the coating material, φ and their mass ratio (R) for each liquid medication is related, according to Spireas by Equation 1 below32:

(1)

Secondly, the liquid load factor can affect the compressibility of the powder mass. The maximum liquid a bulk carrier powder can hold while maintaining optimal compressibility is termed the compressible liquid retention potential. This is specific for the carrier and coating agent. Lu et al.33 maintain that the compressible liquid retention potential is readily determined by pactisity. Hence, a valuable parameter of interest becomes compressible liquid retention potential for the carrier as well as the coating material. Equation 2 relates Lf , compressible liquid load factor, to the compressible liquid retention potential for the carrier (Ψ), compressible liquid retention potential for the coating material (ψ) and the mass ratios of the excipients (R) as follows33:

(2)

At the combination of carrier and coating material, the amount of liquid medication that can be held by such a system while remaining flowable and compressible becomes dependent on the excipient ratio, R. That excipient ratio (R) is the weight ratio of the carrier (Q) to the coating material (q) in the powder mix as given in this third equation below32:

(3)

In line with the third principle of the liuisolid technique, R is related to the compressibility, flow properties and mechanical properties of the liquisolid system hence an optimal value for R is desirable and recommended in literature to be from 10 to 2027,33,34. Recent works show, however, that R can go up to 35, even 50 and still achieve intended formulation outcome30.

The maximum amount of liquid that a given excipient ratio can hold while still maintaining flowability or compressibility is the liquid load factor (Lf). At such an optimal mix, Lf becomes Lo that is the optimal load factor. In mathematical terms, it is the ratio of weight of liquid medication (W) to weight of carrier material (Q) as presented here in Equation 434:

(4)

The Lf is also obtainable from the in Equation 1 or 2, whichever one of the two is the lower value. This value of as calculated is not the same as one that is usually determined experimentally because the former does not take into account the amount of drug in the liquid medication which will affect the total amount of liquid that will be available for absorption and adsorption by the carrier/coating material mix35.

So once the weight (W) of liquid medication and the loading factor (Lf) are known, the amount of carrier and coating materials to be used to obtain an optimal mix is readily determined so that the loaded amount of the liquid vehicle would not hinder the flowability and compressibility of the liquisolid system.

Once the different proportions of the ingredients of the liquisolid system are determined, the procedure for formulating the liquisolid system is followed strictly to obtain the desired results. These steps are well illustrated summarily in Fig. 3.

Formulation considerations of liquisolid dosage forms
Drug selection: The liquisolid technique of drug delivery was set out to solve the pharmaceutical problem of poor solubility. Thus originally, drugs selected for the liquisolid systems usually are those in the Class II and 1V of the Biopharmaceutical Classification System. Drugs in these classes have aqueous solubility challenges that the technique attempts to circumvent. Nevertheless, drugs in the other classes are now being considered when specific applications are intended (e.g., to overcome pH influence or achieve sustained drug release). Even in these later situations, solubility is still a valid concern as it influences the dissolution profile of the final compact and its size.

Fig. 3: Steps involved in converting a liquid medicine to liquisolid system

Another formulation consideration in drug selection is the dose and solubility of drugs delivered as a liquisolid system. To achieve a lower final tablet weight, the drug dose (loading dose) is usually of small weight (usually 1-50 mg) because the carriers and the coating materials generally constitute the larger percentage weight of the final tablet presentation. When drug dose is larger, this results in final tablet size and weight being too high for oral compact as the tablet size will be inconvenient for swallowing. Some of the drugs that have been formulated as such includes itraconazole, Olmesartan Medoxomil and Meloxicam36-38.

Liquid vehicle (non-volatile solvent): This component is used to dissolve/disperse the solid drug to form liquid medicine. The considerations for its use, beyond safety for human consumption and inertness, are solubility capacity, low viscosity, non-volatility and water miscibility. The intended application of the liquisolid system is also considered. It must be noted that the degree to which the liquid vehicle possesses these properties gives indication to how it will influence the final compact. For instance, liquid vehicle with low solubility capacity will have less molecular dispersion of the drug in it. This will translate to the use of more liquid vehicle to form the compact and larger carrier and coating agent used to take up the liquid and resulting tablet size likely to be large. Formulations with vehicles having low solubility capacity may favor a sustained release application. Commonly used liquid vehicles are glycerin, polyethylene glycol, PEG 200, 400 and 600, polysorbate (Tween) 20 and 80. They can be used singly or in combinations as co-solvents. The liquid vehicle even adds to the compactness of a liquisolid system likely due to the presence of hydrogen bonds and the degree of drug solubility or dispersibility in the liquid is directly related to percentage dissolved during dissolution studies33,39 . Although, originally, water miscible liuid vehicles were the desired in formulation using liquisolid systems as designed by Spireas non-water-miscible vehicles (such as castor oil and cremophor EL have now been applied in modern use40.

Carriers: The carriers in liquisolid systems are used to absorb the liquid medicine converting it to solid relatively flowable powders. To select a given material as a carrier, the following properties are considered:

  Specific surface area (SSA): This is the surface area possessed by a unit mass of the carrier. Higher value of SSA contributes to higher absorptive capacity to the carrier, reducing the quantity that will be used in each formulation and by extension the final compact size and weight
  Absorptive capacity: This is the amount of liquid medicine that can be held within a unit mass of the carrier. The SSA, nature of a given material and its surface characteristics determine its absorptive capacity

Thus the ratio of liquid phase to the carrier material had been reported to have an effect on the mechanical properties (hardness, friability, disintegration of tablet and tablet height) of the resulting liquisolid compacts formed29. Higher SSA and absorptive capacity are desirable properties in carriers beyond safety and inertness, to checkmate the final tablet weight. Commonly used examples of carriers for liquisolid systems and some of their are found in Table 3.

Applications of the liquisolid systems: The use of liquisolid technique has been reported in many works in the area of enhancing drug bioavailability through improved dissolution and solubility. This is not surprising as this is what the technique, as developed by Spireas was set out to achieve. Recent works, however, have expanded the techniques to preparing sustained release drug delivery, to overcome the influence of variation in pH for drugs whose dissolution rates are pH dependent and for photoprotection of light-sensitive drugs.

Liquisolid techniques as a delivery strategy to enhance dissolution, solubility and bioavailability of medicines: The bioavailability of a drug is related to its dissolution rate, aqueous solubility and intestinal permeability. Spireas and Sadu27 in his invention of the liquisolid technique was originally to improve drug dissolution and solubility, hence by extension bioavailability. Several authors have since applied the technique to achieve enhanced dissolution, solubility and bioavailability27,32,34,36-38. For example, Vranikova et al.29 applied liquisolid technique to deliver rosuvastatin (a hypolipidemic drug) using both the spray method and simple mixing method to incorporate the formulated liquid medication into the carrier-coating powder mass. The results showed that in both methods of incorporation, about 80% of the rosuvastatin was released within 5 min from the formulated liquisolid tablets. These researchers also found that rosuvastatin liquisolid tablets formed had hardness and friability that were affected by the ratio of the liquid medication to the carrier used in the formulation. They highlighted that the mode of introduction of the liquid medication was a factor to consider during the preparation of liquisolid systems as compacts made from the spray drying process of incorporation produced compacts with higher mechanical properties, better micropolitics and dosage uniformity.

Table 3: Commonly used carriers for liquisolid delivery and their peculiar characteristics41
Classes of carriers or
coating material
Specific examples Specific surface area
(m 2/g)
Absorption capacity
(mL/g)
Polysaccharides Starches (tuber starches) 0.6-4.35 0.96-1.5
(corn starches)   1.22-1.26
Microcrystalline cellulose(Avicel PH 101) 1 0.04-0.08
Amorphous cellulose 12-22
Lactose 0.35
Sugar alcohol Sorbitol 0.37-1.05
Phosphates Fujicalin® (anhydrous dibasic
calcium phosphate)
27~40 1.2
Dicalcium phosphate dihydrate (DCPD)
Florite®
0.3 1-1.3
Silicates Neusilin (magnesium aluminometasilicates)
Aerosil®200
300 3.4
Microsilica 200 1.7
78 2.9
Ordered mesoporous silicates 1030-1500 7.8

Similarly, Thakkar et al.36 attempted to enhance the bioavailability of itraconazole (a potent antifungal) using the liquisolid technique (PEG 600 as the non-volatile liquid, Aerosil 200 as coating material and branded microcrystalline cellulose [Alfacel PH 200] as carrier). Their optimized formulation (which had 10 mg drug in 150 mg liquid) had over 60% dissolution in less than 50 minutes, unlike the marketed brand which was about 40% in the same duration. They also noted that using a higher quantity of dissolution liquid (beyond 150 mg) in the formulations had no significant improvement in the percentage dissolution because saturation in solubility had occurred.

Table 4 below highlights several other examples of published research that has applied liquisolid technique with remarkable success for improved solubility, enhanced dissolution and drug bioavailability enhancement.

About three different mechanisms have been put forward as possible ways that the liquisolid systems bring about enhanced dissolution and bioavailability:

  By increasing the available effective surface area of the drug for dissolution: The drug is molecularly dispersed and held in that state onto the surface of the carrier and coating materials during blending. This surface area is then made available, on the administration of the dosage form, to the gastrointestinal fluid for faster dissolution and subsequent absorption42,43. Saeedi et al.42 even established in their work on dissolution enhancement of indomethacin by liquisolid system, that the fraction of the drug molecularly dispersed in the liquid medication of liquisolid systems was directly proportional to dissolution rate of the indomethacin compact

Table 4: Published works on liquisolid delivery systems showing enhanced dissolution, solubility and bioavailability
Drug Non-volatile
liquid used
Carrier/coating
material
Improvement over
marketed brand (%)
Presentation/
application
Reference
Valsartan Propylene
glycol
Avicel/aerosil
200
Over 15% in dissolution
efficiency
As liquisolid
compact
Chella et al.44
Rosuvastatin PEG 400 Neusilin US2/Aerosil
200
Fast disintegration and
enhanced dissolution
profiles
As liquisolid tablet Vraníková et al.29
Ritonavir PEG 400 MCC/crospovidone Improved dissolution
by over 40%
As liquisolid pellets de Espíndola et al.45
Telmisartan Transcutol HP Avicel PH02/Aerosil
200
Significant improvement Liquisolid compacts Chella et al.46
Efavirenz Transcutol HP Neusilin US2 and
corn starch/Aerosil
Improved dissolution
profile with almost 100%
release in 60 min
Liquisolid tablet Jaydip et al.47
Olmesartan
Medoxomil
Acrysol El 135
(Polyoxyl 35
castor oil)
Avicel PH 102,
Fujicalin and
Neusilin/Aerosil
Significant higher
drug release rates
Liquisolid compact Prajapati et al.37
Olanzapine Kolliphor EL Avicel/ Aerosil Formulations showed
good/excellent flow
properties and
compressibility AUC of
optimized liquisolid
formulation was higher
than marketed tablet
Liquisolid
tablet
Korni and Gonugunta48
Meloxicam PEG 400 Avicel PH102/Aerosil Higher dissolution
with more than 80%
drug release within
10 min
Liquisolid tablets Dias et al.38
Fexofenadine
hydrochloride
Propylene glycol
or Cremophor
®E L
Aerosil® 200
Avicel® PH102/
Increased oral drug
bioavailability by 62%
and reduced Tmax to
2.16 hrs
Liquisolid tablets Yehia et al.49
Itraconazole PEG 600 Afacel PH200/
Aerosil
Higher drug dissolution,
Cmax an AUC
Liquisolid compact Thakkar et al.36
Prednisolone PEG 400, glycerin,
propylene glycol
MCC/Silica Higher dissolution rate
with enhanced
bioavailability
Liquisolid compact Spireas and
Sadu27
Indomethacin PEG 200, glycerin Avicel PH101/
Nano-sized
amorphous silica
Liquisolid formulations
exhibited significantly
higher drug dissolution
rates than directly
compressed tablet
Liquisolid compact Saeedi et al.42
Famotidine Propylene glycol Avicel® PH 102/
Aerosil® 200
Optimized liquisolid
formula had 39% higher
release than directly
compressed tablets during
the first 10 min
Liquisolid tablet Fahmy and
Kassem50
Carbamazepine PEG 200, PEG 400 MCC or Lactose/
Nano-sized
amorphous silica
and PVP or HPMC
or PEG35000 added
to reduce quantity
of carrier-coating
mixture used
Liquisolid formulations
containing PVP showed
significantly higher drug
dissolution rates over
that prepared as directly
compressed
compacts
technique Improved
dissolution as carrier-
coating ratio reduced
from 20 to 10 but
decreased when reduced
to 5 10
Liquisolid compact Javadzadeh et al.34
Furosemide Castor oil,
cremophor EL
and PEG 400
in ratio 1:6:3
(Avicel PH101)
and coating
materials
(Aerosil 200)
2-folds increase in drug
release
SNEDDS delivered
as liuisolid tablets
Dalal et al.40
Atorvastatin PEG 400 Avicel PH 101 OR
Avicel PH 102 OR
Neusilin US2 as
carriers and
Aerosil 200 as
coating material
Optimized liquisolid
tablets showed higher
dissolution compared
to marketed tablets
Liquisolid tablets Windriyati et al.51
Clopidogrel Propylene glycol
and water in
ratio 2:1
Maize starch,
microcrystalline
cellulose/colloidal
silicondioxide
Formulation showed
higher solubility in
HCl buffer pH 2
Liquisolid tablet Ali et al.52

  Drug is presented in a dissolved state: Drugs administered orally undergo some steps (disintegration, deaggregation, then dissolution) before absorption in the gastrointestinal tract. But when the drug is already in the dissolved state in the liquid vehicle, adsorbed onto the carrier and coating material, these preliminary steps are readily passed and the dissolution rate is enhanced for improved bioavailability
  Improved wetting properties of the liquisolid compacts: A key factor that contributes to poor dissolution of solid oral compacts is their poor wettability in the dissolution medium has been explained to be a function of the high interfacial tension between the two phases. In liquisolid technique of drug delivery, the non-volatile liquid used to prepare the delivery system decreases the interfacial tension between the compact surface and the dissolution medium thus resulting in better wetting properties of the final solid dosage form as a first step towards enhanced dissolution42. Vranikova et al.29 reported this as one mechanism used for enhanced dissolution in their work involving rosusvastatin

As a means to minimize influence of pH variation on dissolution: Many drugs are weak acids, bases, or their salts. Hence their solubility and subsequent absorption in the gastrointestinal tract (GIT) is influenced by their ionization constant (pKa) and the pH of the local dissolving environment. After enteral administration, drugs move in the fluid secreted along the gastrointestinal tract. The pH of the gastrointestinal environment varies from the mouth to the large intestine and a large extent, this is a function of whether it is in a fasted or fed state. Thus these changing pH values in the GIT will affect the dissolution, solubility and bioavailability of these ionizable drugs as they gradually coast along in the gastrointestinal fluids. This variation in a person throughout his regimen and between persons could affect, the onset of action, therapeutic outcomes and expressed untoward effects. Drugs formulated via the liquisolid delivery system have now been reported to effectively minimize the impact of varying pH on drugs whose dissolution and solubility are affected by changing pH at the site of absorption.

El-Hammadi and Awad35 investigated the use of liquisolid technique to reduce the effect pH variations will have on the release of loratadine, employing propylene glycol, Avicel PH 102 and Aerosil 200 as non-volatile liquid, carrier and coating materials respectively. The formulated liquisolid systems were evaluated in 3 dissolution media, each of different pH (pH 1.2, 2.5 and 5). Results showed that, generally, the process of drug release becomes slower as the pH value of dissolution medium increased from 1 to 5. The optimized formulation of liquisolid systems showed significantly enhanced dissolution and drug release than the marketed brand at a higher pH of the dissolution medium. One mechanism used to explain this application of liquisolid technique is that liquid used to dissolve or disperse the drug in the formulation influences the saturation solubility thereby enabling maintenance of a good concentration gradient when in a dissolution medium of different pH thus maintaining dissolution35.

Table 5 presents a list of some of the successful claims in literature on this application of liquisolid technique in minimizing pH effect.

Liquisolid technique as a means to achieve photostability of photosensitive drugs: Photodegradation, loss of potency and subsequent likely production of toxic degradation molecules could readily result when photosensitive drugs are exposed to ultraviolet light. These consequent potential adverse effects can be devastating and hazardous to health. The liquisolid technique is gradually finding useful applications in photoprotection, as mentioned in the literature, to provide solution to the longstanding challenge of drug sensitivity to light. Khames54 for example, had reported the evaluation of amlodipine (a photosensitive antihypertensive and antianginal drug) formulated as liquisolid tablet (using propylene glycol as liquid vehicle and Avicel /amorphous silicon or nano-sized TiO2 as carrier/coating material respectively) under the influence of ultraviolet light to see the degree of photoprotection conferred on it by the technique. The optimized liquisolid system inhibited photodegradation under different light energies.

Table 5: List of examples of drugs that have been delivered as liquisolid systems to minimize the effect of pH variation
Drug Non-volatile
liquid used
Carrier/coating
material
Effect of pH
variation
Presentation/
application
Reference
Loratadine
(antihistamine,
used in rhinitis,
urticaria etc,)
Propylene glycol MCC/Silica Dissolution of liquisolid
tablets were significantly
higher than popular
marketed brand at higher
pH of dissolution medium
Liquisolid formulation less
affected by pH variation in
comparison with the directly
compressed tablets and
Clarityn®
liquisolid tablets El-Hammadi and
Awad35
Mosapride citrate
(gastroprokinetic:
Useful in dyspepsia
and acid reflux)
Glycerol Avicel PH 102,
lactose DC as
lactose DC as
carriers/Aerosil
200 as coating
material
Minimize the effect of pH
variation on drug release
along the gastrointestinal
tract using bio-relevant
media
Liquisolid tablets Badawy et al.53

Khazim et al.31 recently carried out similar research on Nimlodipine employing propylene glycol and Avicel, as nonvolatile solvents and carriers respectively but colloidal silicon dioxide, Cab-O-sil and titanium dioxide as the different coatings materials. They reported significantly improved photoprotection by the liquisolid systems formulated.

But how is photoprotection possible with the liquisolid technique? Researchers propose that the photoprotection by this strategy is based on photoprotective capacity of coating material (silicon dioxide) used in liquisolid system, having a high refractive index and diffraction for the light waves of different energies and wavelength so that the drug is protected from the effect of the light. Khames54 also noted that the photoprotective effect was inversely proportional to the excipients ratio (R). This application opens an opportunity to employ the liquisolid technique, with the coating material having good refractive and diffractive capacities as a good alternative to the conventional coating process. It will thus reduce the additional production step and cost of coating photosensitive solid dosage forms. Table 6 shows details of some photosensitive drugs that have been protected using the liquisolid technique.

liquisolid system used to achieve sustained release formulations: Another significant area of application of the liquisolid technique has been in modified-release formulations, especially sustained release. Hydrophobic carriers (e.g., ethyl cellulose, Eudragit® RL) when used in liquisolid compacts can confer reduced wetting on liquisolid formulation, thus modifying the drug’s release rate from the compact. If such carriers are combined with drug release retardants (such as hydrocolloids like hydroxypropyl methyl cellulose, HPMC) in matrix systems, the influence and degree of release modification are heightened to achieve prolonged or sustained drug release. Several authors have employed this and have reported success. Pavani et al.30 for example attempted to produce sustained release formulation of trimetazidine dihydrochloride liquisolid tablets. They employed ethyl cellulose, Eudragit® L 100 and

Table 6: Some published works on application of the liquisolid system to achieve protection for photosensitive drugs
Drug Non-volatile
liquid used
Carrier/coating
material
Degree of
photoprotection
Presentation/
application
Reference
Nimodipine propylene glycol Avicel/colloidal silicon
dioxide, Cab-O-sil and
titanium dioxide
Liquisolid significantly
improved photo
protection
Liquisolid tablet Khazim et al.31
Amlodipine propylene glycol Avicel/amorphous
silicon or nano-sized
TiO2
Inhibited the photodegradative
effect of different light energies
in all prepared liquisolid
formulations
Liquisolid tablet Khames54

Eudragit® RS 100 as retarding agents along with Avicel PH 200 as the carrier, coating material (Aerosil 200) and polysorbate 80 as the nonvolatile liquid to prepare liquisolid tablets. In the dissolution studies of the formulated liquisolid tablets as well as the marketed brands carried out in media of pH 1.2 for first 2 hrs and then in pH 7.4 for the next 12 hrs, they found out that while all the formulations showed potential, the one with Eudragit L100, optimized formulation, sustained the drug release significantly better than the marketed brand and followed a zero order drug release kinetics. That release profile was maintained even after storage for 6 months at temperature of 40±2°C and relative humidity of 75±25%. The possible mechanism of prolonged drug release here is by poor wetting which delays the disintegration stage for drug release to occur30,52.

Ali et al.52 in their work on clopidrogel formulated sustained release oral liquisolid tablets using hydrophilic carriers HPMC and PVP. The HPMC was found to sustain the release of clopidrogel for over 4 hrs in the 0.1 HCl dissolution medium and the sustaining release in the liquisolid compact was dependent on the quantity of the hydrophilic polymers employed. The mechanism as proposed by the researchers is by the formation of a gel by the hydrophilic polymer on absorption of gastrointestinal fluid thus retarding the amount and rate of release of the drug from the core rather than simply inhibiting wetting of the matrix. This proposed mechanism alligns with what other researchers working with hydrocolloids have used to explain the drug release mechanism from such polymers whether natural or modified biopolymers55,56.

Other factors that have been suggested to contribute to achieving sustained drug release in liquisolid systems include the use of carrier with lower specific surface area; ratio of drug in liquid vehicle; the ratio of carrier, retardant and coating material in the formulations investigated; choice of liquisolid vehicle for modulating the drug release rate and drug concentration in vehicle, where high concentration sustains release because at a higher drug concentration, the drug tends to precipitate within the dissolution medium42,57. Table 7 below presents some published literature on drugs that have been delivered as sustained release using the liquisolid system.

Limitations of the liquisolid system: Despite the current wide application of the liquisolid drug delivery technique in improving dissolution of poorly soluble drugs, limiting pH influence on drug release, protecting light-sensitive drugs and achieving sustained release, there are reported peculiar limitations. These are discussed in this section; using the conventional infrastructure for tableting gives the liquisolid techniques potential for industrial application. However, the scale-up of the procedure has been hampered by the challenge of poor and unpredictable flowability and compressibility of the produced liquisolid powder mixtures. This is reflected in the mechanical properties of the compacts usually produced. Another problem is that of mixing, especially when a viscous solvent is used to prepare the liquid medication, or when a small quantity of such liquid is to be incorporated into a large mass of carrier material.

Achieving complete mixing in such a situation may be a real challenge. At Industrial quantities, the challenge of free flow of the liquisolid mix may result in nonuniformity of doses. Another challenge could be in the method of incorporation of the liquid medication onto the carrier powder mass which significantly contributes to compaction behavior of liquisolid systems. In the work of Aleksic et al.60, who investigated the effect of formulation variables (liquid content, spray air pressure and liquid feed rate) on the compaction properties of liquisolid systems, they found that liquisolid system prepared using fluid bed processor gave better compaction behaviour while taking up higher liquid medication.

Table 7: List of published works of sample drugs showing application of liquisolid technique in achieving sustained release
Drug Nonvolatile liquid Carrier/coating agent Application Presentation References
Trimetazidine
dihydrochloride
Polysorbate 80 Ethyl cellulose, Eudragit
and RS100/Aerosil 200
Sustain release Liquisolid tablets Pavani et al.30
Theophylline Non-volatile
co-solvent
HPMC Presence of non-volatile
co-solvent was critical for
prolonging drug release.
The compared with the
directly compressed tablets
Liquisolid tablets Nokhodchi et al.58
Propranolol HCl Polysorbate 80 Eudragit RL 100 or RS
as the carrier/silica as
coating material
Sustained release polymer
chains coalesced better,
resulting in a fine polymer
network with lower
porosity and higher
tortuosity. liquisolid
formulation followed
zero order
As sustained-release
liquisolid tablets
Javadzadeh et al.34
Arthemeter-
Lumefanntrine
Precirol® ATO 5/
Transcutol® HP
Also, tallow fat/
Transcutol® HP
optimizedsystems
MCC/colloidal silicone
dioxide
Tablet compacts formulated
with Precirol® ATO 5/
Transcutol® HP-AL4
achieved higher LUM
release in simulated
intestinal fluid (84.32%)
than tallow fat/
ranscutol® HP-BL3
As sustained-release
liquisolid compact
Nnamani et al.59
Tramadol Propylene glycol Avicel PH 102/Aerosil
200 but HPMC(K4M)
used as release
retardant
Sustained release is
dependent on
percentage of HPMC
in the formulation and
release kinetics Korsmeyer
-Peppas and is non Fickian
release. Sustained longer
than that of marketed
tablets
Liquisolid
compact
Karmarkar et al.57
Clopidogrel
(antiplatelet)
Propylene glycol
and water (2:1)
Polyvinyl pyrollidone,
HPMC/
Formulation achieved
sustained release over that
prepared by the direct
compression
Liquisolid compact Ali et al.52

Achieving a balance between good flowability and compressibility of liquid drug powder mixture can be challenging. True, the established mathematical equations for calculation of the flowable and compressible mixture can be extrapolated for use. This can be technically demanding and cumbersome requiring good skill in formulation to achieve success. Moreover, the value for liquid retention potential and the liquid compressible potential are determined by repeated experimentation and measurements which may be cumbersome. Besides, the values as determined are different from the experimental value that will eventually be in use because the earlier determination did not incorporate the drug yet. More so there is a challenge of possible squeezing-out effect of the liquid medication during compression of the liquisolid powder mix. One way this is circumvented is the formulation of capsules in place of compressed tablets.

Application to drugs with small doses and limited amounts of liquid medication: Liquisolid technique was originally for application to low-dose medicines. The drug dose determines the amount of nonvolatile solvent used to dissolve or disperse it and will eventually translate to the size of the compact prepared.

Thus applying it to formulate high-dose drugs is a limitation as it portends larger size compact which may be impractical. Rokade et al.28 reported that once the therapeutic dose of a drug is greater than 50 mg, a low-level hydrophilic carrier and coating agent are used in the liquisolid technique. Higher drug doses will require a larger amount of carrier to achieve liquisolid powder mass that is free-flowing. This can result in a tablet weight that is more than 1 g and difficult to swallow61.

To overcome this limitation of suitability for delivering low therapeutic doses, the improvement pellets-have been investigated and used with remarkable success. The liquisolid pellet combines the concepts from the liquisolid technology and pelletization technology. It has the advantage of accommodating higher therapeutic doses of drugs for delivery than the liquisolid compact besides other benefits such as being a multi-particulate system over single-unit dosage forms of liquisolid compacts, better distribution along the gastrointestinal tract with enhanced bioavailability45,62. One possibility for achieving the use of a higher dose is the choice and amount of coating agent used. de Espindola et al.45 used crosspovidone at 30% concentration and maintained an R value (that is, the ratio of carrier to coating material in the formulation) of less than 1 and achieved liquisolid pellets of high drug dose and enhanced dissolution.

The release kinetics in liquisolid systems still have room for improvement and will need further studies. Some published studies reveal that classic liquisolid technique did not significantly affect the drug dissolution profile over conventional tablets as was seen in the advanced form, the liquiground. This interesting improvement on the liquisolid system in terms of improved dissolution rate is achievable by the liquiground technique-a combination of the liquisolid technique and the co-grinding technologies. Azharshekoufeh et al.63 applied this liquiground technique to deliver glibenclamide with significantly improved dissolution rate and enhanced bioavailability. This was possible because particle size reduction achieved by co-grinding of liquid medication was more effective in enhancing the dissolution rate of glibenclamide than the simple implementation of the liquisolid technique.

CONCLUSION AND FUTURE POSSIBILITIES

The liquisolid system has proven to be a versatile technology in drug delivery with improvements in drug release, overcoming poor solubility and dissolution while not altering chemical properties of the drug in the formulations. This review has presented the underlying pharmaceutics principles of the technique, provided ample examples of published works on the application in different areas of drug delivery and identified limitations associated with the formulation strategy. The current advancements to circumvent its presently identified limitations are welcomed to keep improving this highly invaluable technique expanding in applications to drugs of higher doses and to take up higher amounts of liquid medication. Also noteworthy is the fact that the combination of this liquisolid technology with other existing technologies brought about improvements in it. As more application is expected in the future with its combination with existing pharmaceutical technologies, formulation scientists can maximize the potential provided by this technique for optimal therapeutic outcomes.

SIGNIFICANCE STATEMENT

This work provides the underlying principles and scientific insight into published works that used the liquisolid technology for drug delivery and explores its current applications. This piece revealed that although the liquisolid technique was originally designed to enhance dissolution and bioavailability of poorly soluble drugs, at present, it has been applied with published examples in three other areas: Modified drug delivery (e.g., sustained releases), minimizing pH influence in drug release and in the protection of light-sensitive medicines. An understanding of the foundational principles of this technique approaches to achieve them, its limitations and its future possibilities are vital for the use of this relatively low-cost yet versatile pharmaceutical technique to design and develop modern innovative drug strategies for better therapeutic outcomes, patient acceptance and medicine availability.

REFERENCES

  1. Nagaich, U., 2018. Pharmaceutical applications of liquisolid technique. J. Adv. Pharm. Technol. Res., 9.
  2. Radke, R. and N.K. Jain, 2022. Enhancement of solubility and bioavailability of BCS class-II Ambrisentan: In vitro, in vivo and ex vivo analysis. Int. J. Appl. Pharm., 14: 67-74.
  3. Savjani, K.T., A.K. Gajjar and J.K. Savjani, 2012. Drug solubility: Importance and enhancement techniques. Int. Scholarly Res. Not., 2012.
  4. Vemula, V.R., V. Lagishetty and S. Lingala, 2010. Solubility enhancement techniques. Int. J. Pharm. Sci. Rev. Res., 5: 41-51.
  5. Varandal, A.B., D.D. Magar and R.B. Saudagar, 2013. Different approaches toward the enhancement of drug solubility: A review. J. Adv. Pharm. Educ. Res., 3: 415-426.
  6. Kanikkannan, N., 2018. Technologies to improve the solubility, dissolution and bioavailability of poorly soluble drugs. J. Anal. Pharm. Res., 7.
  7. Bhalani, D.V., B. Nutan, A. Kumar and A.K.S. Chandel, 2022. Bioavailability enhancement techniques for poorly aqueous soluble drugs and therapeutics. Biomedicines, 10.
  8. Samineni, R., J. Chimakurthy and S. Konidala, 2022. Emerging role of biopharmaceutical classification and biopharmaceutical drug disposition system in dosage form development: A systematic review. Turk. J. Pharm. Sci., 19: 706-713.
  9. Zhang, X., H. Xing, Y. Zhao and Z. Ma, 2018. Pharmaceutical dispersion techniques for dissolution and bioavailability enhancement of poorly water-soluble drugs. Pharmaceutics, 10.
  10. Patil, S.K., S.W. Kalpesh, B.P. Venkatesh, M.A. Anup and T.B. Dheeraj, 2011. Strategies for solubility enhancement of poorly soluble drugs. Int. J. Pharm. Sci. Rev. Res., 8: 74-80.
  11. Kalepu, S. and V. Nekkanti, 2015. Insoluble drug delivery strategies: Review of recent advances and business prospects. Acta Pharm. Sin. B, 5: 442-453.
  12. Jornada, D.H., G.F. dos Santos Fernandes, D.E. Chiba, T.R.F. de Melo, J.L. dos Santos and M.C. Chung, 2016. The prodrug approach: A successful tool for improving drug solubility. Molecules, 21.
  13. Wu, R., X. Mei, Y. Ye, T. Xue and J. Wang et al., 2019. Zn(II)-curcumin solid dispersion impairs hepatocellular carcinoma growth and enhances chemotherapy by modulating gut microbiota-mediated zinc homeostasis. Pharmacol. Res., 150.
  14. de Sá, I.S., A.P. Peron, F.V. Leimann, G.N. Bressan and B.N. Krum et al., 2019. In vitro and in vivo evaluation of enzymatic and antioxidant activity, cytotoxicity and genotoxicity of curcumin-loaded solid dispersions. Food Chem. Toxicol., 125: 29-37.
  15. Choi, J.S., N.H. Cho, D.H. Kim and J.S. Park, 2019. Comparison of paclitaxel solid dispersion and polymeric micelles for improved oral bioavailability and in vitro anti-cancer effects. Mater. Sci. Eng.: C, 100: 247-259.
  16. Arango-Ruiz, Á., Á. Martin, M.J. Cosero, C. Jiménez and J. Londoño, 2018. Encapsulation of curcumin using supercritical antisolvent (SAS) technology to improve its stability and solubility in water. Food Chem., 258: 156-163.
  17. Shen, H. and M. Zhong, 2006. Preparation and evaluation of self-microemulsifying drug delivery systems (SMEDDS) containing atorvastatin. J. Pharm. Pharmacol., 58: 1183-1191.
  18. Agubata, C.O., I.T. Nzekwe, N.C. Obitte, C.E. Ugwu, A.A. Attama and G.C. Onunkwo, 2014. Effect of oil, surfactant and co-surfactant concentrations on the phase behavior, physicochemical properties and drug release from self-emulsifying drug delivery systems. J. Drug Discovery Dev. Delivery, 1.
  19. Xie, S., L. Zhu, Z. Dong, X. Wang, Y. Wang, X. Li and W. Zhou, 2011. Preparation, characterization and pharmacokinetics of enrofloxacin-loaded solid lipid nanoparticles: Influences of fatty acids. Colloids Surf. B: Biointerfaces, 83: 382-387.
  20. Chaturvedi, P. and P. Sharma, 2024. A review on microencapsulation as method of drug delivery. BIO Web Conf., 86.
  21. Han, X., C. Ghoroi, D. To, Y. Chen and R. Davé, 2011. Simultaneous micronization and surface modification for improvement of flow and dissolution of drug particles. Int. J. Pharm., 415: 185-195.
  22. de Miranda, J.C., T.E.A. Martins, F. Veiga and H.G. Ferraz, 2011. Cyclodextrins and ternary complexes: Technology to improve solubility of poorly soluble drugs. Braz. J. Pharm. Sci., 47: 665-681.
  23. Gao, N., M. Guo, Q. Fu and Z. He, 2017. Application of hot melt extrusion to enhance the dissolution and oral bioavailability of oleanolic acid. Asian J. Pharm. Sci., 12: 66-72.
  24. Agubata, C.O., I.T. Nzekwe, A.A. Attama, C.C. Mueller-Goymann and G.C. Onunkwo, 2015. Formulation, characterization and anti-malarial activity of homolipid-based artemether microparticles. Int. J. Pharm., 478: 202-222.
  25. Amarachi, C.S., A.A. Attama and G.C. Onunkwo, 2022. Assessment of the anti-malarial properties of dihydroartemisinin-piperaquine phosphate solid lipid-based tablets. Recent Adv. Anti-Infect. Drug Discovery, 17: 103-117.
  26. Tayel, S.A., I.I. Soliman and D. Louis, 2008. Improvement of dissolution properties of carbamazepine through application of the liquisolid tablet technique. Eur. J. Pharm. Biopharm., 69: 342-347.
  27. Spireas, S. and S. Sadu, 1998. Enhancement of prednisolone dissolution properties using liquisolid compacts. Int. J. Pharm., 166: 177-188.
  28. Rokade, M., P. Khandagale and D. Phadtare, 2018. Liquisolid compact techniques: A review. Int. J. Curr. Pharm. Res., 10: 1-5.
  29. Vraníková, B., J. Gajdziok and D. Vetchý, 2015. Modern evaluation of liquisolid systems with varying amounts of liquid phase prepared using two different methods. BioMed Res. Int., 2015.
  30. Pavani, E., S. Noman and I.A. Syed, 2013. Liquisolid technique based sustained release tablet of trimetazidine dihydrochloride. Drug Invent. Today, 5: 302-310.
  31. Khazim, M.E., H.J. Hasan, N.M. Hanoon and H.A.H. Al-Sa’idy, 2021. Improvement of the photostability of nimodipine by using liquisolid compacts technique. Univ. Thi-Qar J. Sci., 8: 43-51.
  32. Tiong, N. and A.A. Elkordy, 2009. Effects of liquisolid formulations on dissolution of naproxen. Eur. J. Pharm. Biopharm., 73: 373-384.
  33. Lu, M., H. Xing, J. Jiang, X. Chen, T. Yang, D. Wang and P. Ding, 2017. Liquisolid technique and its applications in pharmaceutics. Asian J. Pharm. Sci., 12: 115-123.
  34. Javadzadeh, Y., B. Jafari-Navimipour and A. Nokhodchi, 2007. Liquisolid technique for dissolution rate enhancement of a high dose water-insoluble drug (carbamazepine). Int. J. Pharm., 341: 26-34.
  35. El-Hammadi, M. and N. Awad, 2012. Investigating the use of liquisolid compacts technique to minimize the influence of pH variations on loratadine release. AAPS PharmSciTech, 13: 53-58.
  36. Thakkar, H.P., D. Vasava, A.A. Patel and R.D. Dhande, 2020. Formulation and evaluation of liquisolid compacts of itraconazole to enhance its oral bioavailability. Ther. Delivery, 11: 83-96.
  37. Prajapati, S.T., H.H. Bulchandani, D.M. Patel, S.K. Dumaniya and C.N. Patel, 2013. Formulation and evaluation of liquisolid compacts for olmesartan medoxomil. J. Drug Delivery, 2013.
  38. Dias, R.J., S. Ranjan, K.K. Mali, V.S. Ghorpade and V.D. Havaldar, 2017. Liquisolid compacts of meloxicam: In-vitro and in-vivo evaluation. Egypt. Pharm. J., 16: 112-120.
  39. Leaner, C. and J. Dressman, 2000. Improving drug solubility for oral delivery using solid dispersions. Eur. J. Pharm. Biopharm., 50: 47-60.
  40. Dalal, L., A.W. Allaf and H. El-Zein, 2021. Formulation and in vitro evaluation of self-nanoemulsifying liquisolid tablets of furosemide. Sci. Rep., 11.
  41. Schlack, H., A. Bauer-Brandl, R. Schubert and D. Becker, 2001. Properties of Fujicalin®, a new modified anhydrous dibasic calcium phosphate for direct compression: Comparison with dicalcium phosphate dihydrate. Drug Dev. Ind. Pharm., 27: 789-801.
  42. Saeedi, M., J. Akbari, K. Morteza-Semnani, R. Enayati-Fard, S. Sar-Reshteh-dar and A. Soleymani, 2011. Enhancement of dissolution rate of indomethacin: Using liquisolid compacts. Iran. J. Pharm. Res., 10: 25-34.
  43. Pas, T., S. Verbert, B. Appeltans and G. van den Mooter, 2020. The influence of crushing amorphous solid dispersion dosage forms on the in-vitro dissolution kinetics. Int. J. Pharm., 573.
  44. Chella, N., N. Shastri and R.R. Tadikonda, 2012. Use of the liquisolid compact technique for improvement of the dissolution rate of valsartan. Acta Pharm. Sin. B, 2: 502-508.
  45. de Espíndola, B., A.O. Beringhs, D. Sonaglio, H.K. Stulzer, M.A.S. Silva, H.G. Ferraz and B.R. Pezzini, 2019. Liquisolid pellets: A pharmaceutical technology strategy to improve the dissolution rate of ritonavir. Saudi Pharm. J., 27: 702-712.
  46. Chella, N., N. Narra and T.R. Rao, 2014. Preparation and characterization of liquisolid compacts for improved dissolution of telmisartan. J. Drug Delivery, 2014.
  47. Jaydip, B., M. Dhaval, M.M. Soniwala and J. Chavda, 2020. Formulation and optimization of liquisolid compact for enhancing dissolution properties of efavirenz by using DoE approach. Saudi Pharm. J., 28: 737-745.
  48. Korni, R.D. and C.S.R. Gonugunta, 2024. Olanzapine liquisolid tablets using kolliphor el with improved flowability and bioavailability: In vitro and In vivo characterization. Turk. J. Pharm. Sci., 21: 52-61.
  49. Yehia, S.A., M.S. El-Ridi, M.I. Tadros and N.G. El-Sherif, 2015. Enhancement of the oral bioavailability of fexofenadine hydrochloride via cremophor® el-based liquisolid tablets. Adv. Pharm. Bull., 5: 569-581.
  50. Fahmy, R.H. and M.A. Kassem, 2008. Enhancement of famotidine dissolution rate through liquisolid tablets formulation: In vitro and in vivo evaluation. Eur. J. Pharm. Biopharm., 69: 993-1003.
  51. Windriyati, Y.N., M. Badriyah, D.A. Kusumaningtyas and R.L. Riesmalia, 2020. Liquisolid tablets formulation of atorvastatin calcium using polyethylene glycol 400 as solvent and some carrier materials. Indones. J. Pharm., 31: 305-311.
  52. Ali, B., A. Khan, H.S. Alyami, Majeed Ullah, A. Wahab, M. Badshah and A. Naz, 2021. Evaluation of the effect of carrier material on modification of release characteristics of poor water soluble drug from liquisolid compacts. PLoS ONE, 16.
  53. Badawy, M.A., A.O. Kamel and O.A. Sammour, 2016. Use of biorelevant media for assessment of a poorly soluble weakly basic drug in the form of liquisolid compacts: In vitro and in vivo study. Drug Delivery, 23: 808-817.
  54. Khames, A., 2013. Liquisolid technique: A promising alternative to conventional coating for improvement of drug photostability in solid dosage forms. Expert Opin. Drug Delivery, 10: 1335-1343.
  55. Akpabio E.I., T.O. Uwah, D.E. Effiong and J. Godwin, 2020. Evaluating hydrocolloids of Sida acuta as sustained release matrix for ibuprofen tablet. Global J. Med. Res., 40: 16-21.
  56. Uwah, T.O.O., E.I. Akpabio, T.C. Jackson, D.E. Effiong and E. Uduk, 2023. Potentials of Sterculia tragacantha Lindl seed husk gum as a release modifier in matrix tablet formulation. GSC Biol. Pharm. Sci., 25: 025-037.
  57. Karmarkar, A.B., I.D. Gonjari, A.H. Hosmani and P.N. Dhabale, 2010. Evaluation of in vitro dissolution profile comparison methods of sustained release tramadol hydrochloride liquisolid compact formulations with marketed sustained release tablets. Drug Discoveries Ther., 4: 26-32.
  58. Nokhodchi, A., R. Aliakbar, S. Desai and Y. Javadzadeh, 2010. Liquisolid compacts: The effect of cosolvent and HPMC on theophylline release. Colloids Surf. B: Biointerfaces, 79: 262-269.
  59. Nnamani, P.O., A.A. Ugwu, E.C. Ibezim, F.C. Kenechukwu and P.A. Akpa et al., 2016. Sustained-release liquisolid compact tablets containing artemether-lumefantrine as alternate-day regimen for malaria treatment to improve patient compliance. Int. J. Nanomed., 11: 6365-6378.
  60. Aleksić, I., I.G. Ilić, S. Cvijić and J. Parojčić, 2020. An investigation into the influence of process parameters and formulation variables on compaction properties of liquisolid systems. AAPS PharmSciTech, 21.
  61. Sinha, S., M. Ali, S. Baboota, A. Ahuja, A. Kumar and J. Ali, 2010. Solid dispersion as an approach for bioavailability enhancement of poorly water-soluble drug ritonavir. AAPS PharmSciTech, 11: 518-527.
  62. Lam, M., T. Ghafourian and A. Nokhodchi, 2019. Liqui-pellet: The emerging next-generation oral dosage form which stems from liquisolid concept in combination with pelletization technology. AAPS PharmSciTech, 20.
  63. Azharshekoufeh, L., J. Shokri, M. Barzegar-Jalali and Y. Javadzadeh, 2017. Liquigroud technique: A new concept for enhancing dissolution rate of glibenclamide by combination of liquisolid and co-grinding technologies. Bioimpacts, 7: 5-12.

How to Cite this paper?


APA-7 Style
Effiong, D.E., Onunkwo, G.C. (2024). Principles, Applications and Limitations of the Liquisolid System of Drug Delivery: A Review. Trends in Medical Research, 19(1), 178-198. https://doi.org/10.3923/tmr.2024.178.198

ACS Style
Effiong, D.E.; Onunkwo, G.C. Principles, Applications and Limitations of the Liquisolid System of Drug Delivery: A Review. Trends Med. Res 2024, 19, 178-198. https://doi.org/10.3923/tmr.2024.178.198

AMA Style
Effiong DE, Onunkwo GC. Principles, Applications and Limitations of the Liquisolid System of Drug Delivery: A Review. Trends in Medical Research. 2024; 19(1): 178-198. https://doi.org/10.3923/tmr.2024.178.198

Chicago/Turabian Style
Effiong, Daniel, Ekpa, and Godswill Chukwunweike Onunkwo. 2024. "Principles, Applications and Limitations of the Liquisolid System of Drug Delivery: A Review" Trends in Medical Research 19, no. 1: 178-198. https://doi.org/10.3923/tmr.2024.178.198