You are on page 1of 13

Pharmaceutical Development and Technology

ISSN: 1083-7450 (Print) 1097-9867 (Online) Journal homepage: http://www.tandfonline.com/loi/iphd20

Formulation design, challenges, and development


considerations for fixed dose combination (FDC) of
oral solid dosage forms

Divyakant Desai, Jennifer Wang, Hong Wen, Xuhong Li & Peter Timmins

To cite this article: Divyakant Desai, Jennifer Wang, Hong Wen, Xuhong Li & Peter Timmins
(2013) Formulation design, challenges, and development considerations for fixed dose combination
(FDC) of oral solid dosage forms, Pharmaceutical Development and Technology, 18:6, 1265-1276,
DOI: 10.3109/10837450.2012.660699

To link to this article: https://doi.org/10.3109/10837450.2012.660699

Published online: 16 Feb 2012.

Submit your article to this journal

Article views: 2247

View related articles

Citing articles: 17 View citing articles

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=iphd20
Pharmaceutical Development and Technology, 2013; 18(6): 1265–1276
© 2013 Informa Healthcare USA, Inc.
ISSN 1083-7450 print/ISSN 1097-9867 online
DOI: 10.3109/10837450.2012.660699

REVIEW: PHARMACEUTICAL Development Fundamentals

Formulation design, challenges, and development


considerations for fixed dose combination (FDC) of oral
solid dosage forms
Divyakant Desai1, Jennifer Wang1, Hong Wen2, Xuhong Li3, and Peter Timmins4

Drug Product Science and Technology, Bristol-Myers Squibb Co., New Brunswick, NJ, USA, 2Pentian Pharmaceuticals
1

Company, Shanghai, China, 3CDER, FDA, Silver Spring, MD, USA, and 4Drug Product Science and Technology,
Bristol-Myers Squibb, Co., Moreton, Merseyside, UK

Abstract
Fixed dose combination (FDC) products are common in the treatment of hypertension, diabetes, human
immunodeficiency virus, and tuberculosis. They make it possible to combine two or more drug molecules with
different modes of pharmacological actions in a single dosing unit and optimize the treatment. From a patient
perspective, they offer convenience, reduced dosing unit burden, and cost savings. From a clinical perspective, aging
population in developed countries will need multiple medications to treat age related diseases and co-morbidities.
FDC products simplify dosing regimen and enhance patient compliance. As outlined in the article, the number of
FDC products has grown over the years and the trend is likely to continue. This review article gives an overview to
pharmaceutical scientists about recent trends in the formulation development of the FDC products and provides
decision trees to select most optimum formulation development strategy. While some formulation technologies
such as multi-layer tablets, multiparticulate systems, active film coating, and hot-melt granulation are discussed in
more detail, a few specialized technologies are also introduced briefly to the readers.
Keywords:  Fixed dose combination, FDC, formulations, an active coating, HME, bi-layer tablet

Overview of fixed dose combination drug components, only 18 have three drug components,
products of oral dosage forms and only one human immunodeficiency virus (HIV)
Fixed dose combination (FDC) drug products are very FDC has four drug components.[4] Among cardiovascular
common for almost all therapeutic areas. Many different agents, nearly 80% of the FDCs are for hypertension[5];
types of oral, parenteral, and inhalation FDC formulations FDCs for HIV/AIDS ranked top in infection; most of FDCs
are commercially available.[1] Reviews of the Physicians’ in metabolic diseases are for diabetes.[6] Among the routes
Desk Reference (PDR) and United States Pharmacopeia of administration for the FDC products, oral delivery
(USP) in 2005 revealed that there were 150 FDC products, ranked much higher than any others, followed by inhala-
mainly cardiovascular and cough medicines, listed in tion. This review article is mainly focused on the FDC oral
PDR and 80 in USP.[2] The difference in numbers for both solid dosage formulation and process development.
compendia was attributed to the lag time for a marketed According to the Drugs@FDA website, 26 out of the
product to be a USP product. Nevertheless, both numbers 101 Food and Drug Administration (FDA)-approved New
showed continued growth of the FDC products. Similarly, it Drug Applications (NDAs) are FDC products in 2010. A
was shown that the top five therapeutic areas for approved list of the FDC products at different development stages
FDCs are infectious, cardiovascular, hormone, allergies in 2009 and 2010 have been tabulated in Table 1 in order
and pain by surveying the oral FDC products listed in the to assess the current trends in the FDC product develop-
2010 PDR.[3] Majority of the FDCs are composed of two ment. Even though the list is incomplete, it is clear that

Address for Correspondence: Dr. Divyakant Desai, Ph.D., Drug Product Science and Technology, Bristol-Myers Squibb Co., PO Box 191,
New Brunswick, NJ 08903-0191, USA. Tel: (732)-227–6458. Email: divyakant.desai@bms.com
(Received 09 September 2011; revised 20 January 2012; accepted 20 January 2012)

1265
1266  D. Desai et al.

Table 1.  Partial list of FDCs at different development stages reported in 2009 and 2010.
Company Indication Combination drugs Status
Takeda Hypertension Candesartan and amlodipine besylate NDA submission in Japan
Takeda Hypertension Candesartan and hydrochlorothiazide Approved by EMEA
Takeda Type 2 diabetes Competact® (pioglitazone HCl and Application withdrawn
metformin HCl) from EMEA.
Bristol-Myers Squibb Type 2 diabetes Kombiglyze XR (saxagliptin and metformin Approved by FDA
Company and AstraZeneca HCl XR)
Kombiglyze (saxagliptin and metformin) Approved by EMEA
Takeda Type 2 diabetes Pioglitazone and sulfonylurea NDA submission in Japan
Novo Nordisk and Sciele Type 2 diabetes Repaglinide and metformin HCl Approved by FDA
Pharma
QRxPharma Pain Morphine and oxycodone Phase III
Abbott Cholesterol Simcor (niacin and simvastatin) Approved by FDA
Schering-Plough Asthma Mometasone furoate and formoterol Validated by EMEA
fumarate
Pfizer and Sigma-Tau Malaria Eurartesim® (dihydroartemisinin + Marketing in Africa
piperaquine)
AstraZeneca and Pozen Relief of signs and symptoms of Vimovo™ (naproxen and esomeprazole Approved by FDA
osteoarthritis, rheumatoid magnesium)
arthritis and ankylosing
spondylitis, and to decrease the
risk of developing gastric ulcers
in patients at risk of developing
NSAID-associated gastric ulcers
Gilead HIV Truvada® and TMC278 Formulation BE achieved
GlaxoSmithKline Benign prostatic hyperplasia Approved by Swissmedic
Duodart® (dutasteride and tamsulosin)
and EMEA
BE, bioequivalence; BPH, benign prostatic hyperplasia; EMEA, European Medicines Agency; FDA, US Food and Drug Administration;
HIV, human immunodeficiency virus; JP, NDA, New drug application.

treatment for both hypertension and type 2 diabetes has antiplatelet agents, dipyridamole and aspirin, which
attracted a lot of attention in FDC product development. showed better efficacy than the co-administration of
Current pipeline in the pharmaceutical industry is rela- these drugs.[7]
tively empty after a burst of several innovative new com- 2. The FDC of amoxicillin and potassium clavulanate
pounds; FDCs are the only new therapies that will achieve enhances the effectiveness of amoxicillin, because
very significant growth in sales. Overall, FDCs have been potassium clavulanate is an inhibitor of β lacta-
developed in many different therapeutic areas, and will mases, protecting amoxicillin from degradation by
continue to be a valuable option in drug development. β lactamases produced by many microbial organ-
isms.[8] This combination effectively extends the anti-
Advantages and disadvantages of FDC products bacterial spectrum of amoxicillin.
FDC products containing two or more active pharma- 3. Sometimes, the drugs are combined to minimize
ceutical ingredients (APIs) in a single oral dosage form the potential drug abuse, such as Suboxone®
are playing important roles in treating various kinds of (buprenorphine and naloxone) and Lomotil®
diseases, such as cardiovascular, HIV/AIDS, malaria, (diphenoxylate and atropine). Buprenorphine and
and tuberculosis, where polypharmacy is a norm (1). The naloxone are combined in Suboxone® to prevent the
FDC products, however, have both non-therapeutic and possibility of injecting the product by opioid addicts
therapeutic advantages and disadvantages. to get high on buprenorphine, a partial opioid ago-
nist. Naloxone, an opioid antagonist, will generate
Therapeutic advantages of FDC products withdrawal symptoms if Suboxone® is injected.[9]
The FDC products provide the opportunities to combine Similarly, diphenoxylate/atropine combination is
more than one medicine with different pharmacological used to counter opioid-like effect of diphenoxylate
mechanisms into a single dosing unit. This has added at high doses with anticholinergic effects of atro-
significant clinical values in some therapeutic areas pine.[10]
such as anti-hypertensive, anti-viral, anti-glycemic, and 4. The combination of short-acting pseudoephed-
anti-cholesterol treatment. The clinical benefits of FDC rine and long-acting loratidine for the coverage
include but not limited to: of extended period in allergy products such as
Claritin-D®.
1. Synergistic therapeutic effects of two molecules. This 5. Minimizing the drug-resistant microorganisms,
was noted for Aggrenox®, a combination two oral especially for anti-viral and anti-tubercular drugs.

 Pharmaceutical Development and Technology


Formulation design and development of fixed dose combinations  1267
For the HIV treatment, efavirenz, emtricitabine, and when the serum creatinine is 1.5 mg/dL and 1.4 mg/
tenofovir are combined in Atripla®, a single tablet. dL, respectively.[14] This stipulation may be easily
For the treatment of tuberculosis, FDC products missed for FDC products containing metformin.
containing rifampicin, isoniazid, pyrazinamide, and 4. Pharmacists and physicians can easily overlook the
ethambutol are available.[11] limit of certain drugs in FDCs. An example is acet-
6. Sometimes, one drug is combined with another aminophen for which the maximum daily dose is 4 g/
drug to improve safety/tolerability. For example, day.[14] Many patients, in addition to acetaminophen,
in Arthrotec® tablet, misoprostol, a prostaglandin can be on more than one FDCs of acetaminophen
analog, is combined with diclofenac, a non-steroidal with codeine, oxycodone, or hydrocodone for pain
anti-inflammatory drug (NSAID).[2] Misoprostol management, the limit can be missed.
has gastro-protective property which reduces gas- 5. Since the FDC contains multiple drugs in one tablet,
trointestinal (GI) irritation/ulceration caused by sometimes, the tablet size may be too big to swal-
diclofenac. low for pediatric and elderly patients. For example,
7. The FDC products improve patient compliance for type 2 diabetes, metformin is the main pillar of
by reducing dosing unit burden. For example, the the therapy with the dose range of 500 mg–2000 mg.
well-known cocktail therapies in HIV/AIDS patients When any other antidiabetic agents are combined
involve the combination of several active ingredients with metformin, the tablet size can be too big to swal-
to create multiple obstacles to HIV replication and low easily.
to reduce the possibility of drug resistance.[12] The
patient compliance is very important to avoid devel- Regulatory considerations and guidance on
opment of the drug resistance. FDC products
The regulatory authorities have recognized the impor-
Non-therapeutic advantages of the FDC products tance of FDC drug products on public health by devel-
oping relevant guidelines. In the United States, the basic
1. The evolving FDC for anti-retroviral products signifi- requirements for the FDC prescription drugs for humans
cantly improves HIV patient’s compliance, especially have been defined in the code of federal registration 21
for those in the third world countries, not only due to CFR 300.50. A FDC is defined as “Two or more drugs may
the reduced dosing burden, but also due to the fact be combined in a single dosage form when each compo-
that the FDC products tend to be cheaper than mul- nent makes a contribution to the claimed effects and the
tiple individual products. dosage of each component (amount, frequency, dura-
2. The FDC products reduce manufacturing cost com- tion) is such that the combination is safe and effective for
pared with the costs of manufacturing separate prod- a significant patient population requiring such concur-
ucts. At the same time, the FDC products simplify the rent therapy as defined in the labeling for the drug.”
logistics of distribution. There was a draft guidance for the pharmaceutical
3. Finally, the FDC products offer an opportunity for the industry regarding the clinical evaluation of estrogen/
pharmaceutical companies to sustain their product progestin combination drug products published in
pipeline as the industry faces a slowdown in block- 2003.[15] The first FDC related guidance for the pharma-
busters drugs.[13] This was evidenced by the fact that a ceutical industry was implemented for anti-retroviral
significant amount of newly approved drug products products by FDA in 2006.[16] This guidance was “intended
are FDCs. to encourage sponsors to submit applications to the FDA
for approval of the FDC products and co-packaged ver-
Disadvantages of the FDC products sions of previously approved anti-retroviral therapies
In spite of above-mentioned advantages, there are some for the treatment of HIV”. After reviewing the guidance,
disadvantages of FDC products. readers would understand “To which products does this
guidance apply?”, “What regulatory procedures apply to
1. FDC products reduce dosing flexibility. To compen- FDC and co-packaged HIV products?” and “What are
sate the reduced dosing flexibility, multiple strength the characteristics of the potential regiments for FDC or
combinations are developed to cover common co-packaged HIV therapies?” Considerations for clinical,
doses for each drug component of the FDC product. clinical pharmacology and biopharmaceutics, chemistry,
However, some disease treatments or some patients manufacturing, and controls, microbiology/virology and
require frequent dose adjustments where the FDC labeling were provided in this document. Other regulatory
products are not very useful. considerations, such as exclusivity, user fees, pediatric
2. It is more difficult to pin point the cause of the adverse studies and post-approval issues were addressed as well.
drug reactions of the FDC products as more than one There are two possible NDA pathways for FDC prod-
drug is administered in a single dosing unit. ucts for drugs already approved by the FDA. They depend
3. FDC products could be challenging for pharmacists on the source of the data. For 505 (b) (1), a sponsor gen-
to monitor patients’ drug therapy. For example, erated all necessary data or has a permission to use the
metformin is contraindicated for men and women data. For 505 (b) (2), a sponsor did not generate necessary

© 2013 Informa Healthcare USA, Inc.


1268  D. Desai et al.
data or has right to reference the data, but would rely on process development easier. In addition, discussion with
relevant published data or literature. The details are pro- regulatory authorities may be critical in clinical design or
vided in the FDA guidance.[17] The main advantage of 505 even determining the feasibility of a drug combination.
(b) (1) is the longer product exclusivity compared to the
limited exclusivity of 505 (b) (2). Extension of patent life
Until recently, FDC products were only approved for One common strategy to protect a drug with pending
drugs previously approved as single entity. However, it patent expiration is to develop its FDC with another new
has been shown that certain diseases such as hepatitis C, drug that has longer patent protection. For example,
viral resistance to single molecule can develop quickly. valsartan (Diovan®), a blockbuster anti-hypertensive
Therefore, it is more beneficial to co-develop molecules drug with annual sales of more than $8 billion, will lose
starting very early in the development. With advances in compound patent protection in the United States in
genomics and cell biology, regulation has also advanced 2012. Aliskiren (Valturna®) is the 2007 FDA-approved
in concert.[18] Most recently, in December 2010, a new new anti-hypertensive drug. The FDC tablet contain-
FDA draft guidance regarding the co-development of ing valsartan and aliskiren will not only provide clinical
two or more unmarketed investigational drugs for use in benefits, but may also help to retain some of the Diovan®
combination was published in the Federal Register and market share after its patent expiration. The usefulness
is soliciting public comments.[19] This guidance has pro- of FDC to preserve market share of the individual best-
vided recommendations to the industry in “determin- selling drugs has mixed produced mixed results so far.
ing whether or not, a co-development is an appropriate For example, Caduet®, a combination of two multibil-
development option.” This document has also listed a lion dollar drugs, Norvasc® (amlodipine) and Lipitor®
number of regulatory procedures to be considered in co- (atorvastatin), only managed to have a few million dol-
development of drugs for FDC products. lars in annual sale.
For the regulatory landscape outside of the United
States, the interested readers can refer to the World Health Formulation development challenges and strategies for
Organization’s Guidelines for Registration of Fixed Dose FDC oral solid dosage forms
Combination Medicinal Products for general advices.[11] Formulation design and process development for FDC
In March 2009, the World Health Organization approved products are typically more challenging than corre-
the expedited process to review FDC medicines and co- sponding single entity products.[21] Many factors can
packaging of existing therapies for the treatment of HIV/ contribute to the complexity in formulation design and
AIDS in developing countries.[20] process development. If combined drug strengths are too
high (>1000–1500 mg), the tablet size of the FDC product
Formulation development strategies for can become critical in achieving patient acceptance.
FDC oral solid dosage forms Disproportionate drug dose combinations can be
In general, drugs are selected for FDC development very challenging in achieving good content uniformity,
based on fundamental understanding of their pharma- especially in weight control of low dose layer of multi-
cological mechanisms, drug–drug interactions, clinical layer tablets that contains low dose in the second layer.
experience, and pharmacokinetic profiles and manufac- For example, drugs like metformin where dose as high
turability. Synergistic therapeutic effects are desired, but as 1000 mg in one layer is combined with sulfonylureas
very difficult to demonstrate. with the low dose of 2.5 mg in the second layer. Chemical
Establishing bioequivalence (BE) of drugs in FDC to compatibility among all drugs as well as excipients in
drugs co-administered as individual entities is a very FDCs are always the first step in formulation design and
common approach compared to therapeutic equivalence process development. Efforts should be made to stay with
(TE) as a clinical strategy for FDC development. The main the same dosage form as that of the single entities. For
reason is that the BE studies are shorter and less resource example, if the single entity is a tablet, then select tablet
intensive compared to the TE studies. From a clinical as the dosage form in monolithic or bi-layer tablet. A bi-
perspective, drug–drug interactions in the FDCs need to layer tablet is needed if there is incompatibility between
be evaluated carefully. The possible food effects on the the drugs or different drug release profiles are needed.
drug components in the FDC may be different as well. For some reasons, if a tablet dosage form is not feasible
then multiparticulate dosage form should be pursued
Dose proportionality consideration for bio-waiver (Figure 1).
FDCs often require multiple dose strength combinations
based on the mono therapy regime. The multitude of Clinical challenges
dose strength combinations will not only impact formu- Instead of waiting for the FDC formulation develop-
lation design, but also increase BE complexity. If dose ment to be completed, clinical studies may be initiated
proportion is feasible, it is possible to obtain bio-waivers by co-administering two individual single entity tablets.
for the lower strength combinations after establishing the Once the FDC product is developed, then a BE study can
BE for the highest strength combination of the FDC. Dose be conducted to demonstrate that the co-administered
proportion may also make the formulation design and components are bioequivalent to those from a FDC

 Pharmaceutical Development and Technology


Formulation design and development of fixed dose combinations  1269

Figure 1.  Decision tree of the formulation design of FDC.

product. This strategy removes the FDC product develop- is finite volume. Therefore, for this approach to succeed
ment from the rate-limiting path in the overall develop- the doses have to be small or drugs should have high lipid
ment program. However, if the potential risk for BE failure solubility.
is high then this strategy is not advisable. The BE failure
risk assessment needs to be conducted using the frame Formulation design and process development for FDC
work of biopharmaceutical classification system (BCS) oral products
and clinical experience with two drugs identified for Various formulations and manufacturing processes have
FDC. The BE failure risk is higher for BCS Class 4 drugs, been successfully used in commercial FDC products.
the drugs with low solubility and low permeability, com- Several commonly used approaches are reviewed briefly
pared to other BCS classes. The potential BE failure risk in light of their inherent advantages and challenges. An
also needs to be taken into consideration in selecting the example decision tree on how to select the manufactur-
fixed dose formulation approach. Ideally, the dissolution ing process for a FDC product is shown in Figure 2.
profiles of the components from a FDC should be similar
to those of respective single entity tablets. Monolithic systems
Another challenge is how to dose FDC product with When two or more drugs are chemically compatible
regard to meal. For example, metformin is dosed after the with each other, a monolithic system in tablet solid dos-
meal for better GI tolerance. In contrast, sulfonylureas age form may be considered first during formulation
are dosed prior to meal to avoid possible hypoglycemia. development. Figure 1 briefly describes an example
Interestingly, the metformin combination products are decision tree in determining whether a FDC product can
given with meals. In addition, some drugs are known be developed into a monolithic system in a much sim-
to have positive or negative food effects. In case of posi- plified scenario. A monolithic FDC product is similar to
tive food effects, the drug bioavailability is increased single entity formulation except it contains at least two
significantly when given with a high fat meal compared active ingredients. However, whether or not being able to
to when administered in a fasted state. For the negative achieve BE using monolithic for all drugs in FDCs have
food effect, the bioavailability is decreased significantly to be carefully evaluated. For example, when combining
when given with high fat meals. FDCs of such drugs one BCS II drug and one BCS III drug in one FDC, due
could be very challenging. The only known formulation to different dissolution profiles and absorption mecha-
approach to overcome positive food effect is a softgel for- nisms, even though they may be chemically compatible,
mulation containing long chain and/or medium chain monolithic system may not be a suitable choice. For
fatty acids. The major limitation of this approach for FDC example, a formulation matrix used for a BCS Class III

© 2013 Informa Healthcare USA, Inc.


1270  D. Desai et al.
containing calcium carbonate, magnesium oxide, mag-
nesium carbonate, or sodium phosphate monobasic
was evaluated as the buffer layer to prevent the chemi-
cal reactions between pravastatin and aspirin in two
separate layers in a FDC tri-layer tablet.[27] Additionally,
the drug–drug interaction from the two separate layers
with an interface may manifest in such a way that the
in vitro dissolution release profile of one API is affected
due to the change of the microenvironment caused
by the other API or excipients in the second layer. It is
always valuable to evaluate whether or not one drug
will impact another drug’s in-vitro dissolution profile
with or without pH changes.
Once the chemical stability profiles of all drugs in a
FDC are satisfactory, multi-layer tablets present some
unique challenges in terms of obtaining acceptable
tablet physical characteristics compared to the conven-
tional monolithic tablets. The challenges may include
insufficient overall tablet tensile strength which may
lead to excessive friability, delamination at the interface
between layers, or capping within individual layers. In
Figure 2. Decision tree for manufacturing process selection addition, unsatisfactory weight control for individual
for FDC. layers or overall tablet may lead to problematic content
uniformity of the APIs.
drug may impair wetting and dispersion of a BCS Class
II drug, it may make its bioavailability highly variable. In Layer formulation composition similarity  In order to miti-
such cases, a monolithic system is not desired. gate the challenges and to avoid the pitfalls of multi-layer
tablet formulation development, a preferred approach
Multi-layer tablets is to maintain certain level of formulation similarity
As an extension of the well-known conventional single- between layers. This view point is further elaborated using
layer tablets, multi-layer tablets provide a simplified bi-layer tablets as an example. If sufficient similarities
treatment regimens, greater patient convenience and can be maintained between the two layers, there would
compliance. be less potential for the aforementioned bi-layer tablet
process issues to occur. Depending on the difference
Chemical stability of layers  From drug product develop- of the drug dose that has to be included in each layer, it
ment perspective, bi-layer tablets, sometimes tri-layer would be beneficial to formulate both layers with similar
tablets, are a convenient way to formulate incompat- composition and weight. When the drug loading is high
ible drugs in a single tablet. It is the approach that in only one layer for clinical reasons (e.g. 300–600 mg),
often results in significantly improved overall chemical or when the weight of one layer has to be kept high for
stability of the drug product. In addition, it is a conve- formulation reasons (e.g. 800–1200 mg), it is necessary to
nient approach to enable the inclusion of an immedi- keep the overall tablet weight low enough (e.g. 1000–1500
ate release loading dose and another controlled release mg) for good patient compliance. In such cases, when
dose for sustained action in a multi-layer tablet in the weights of both layers have to be significantly differ-
which the API included in all layers could be the same ent, one should try to make compositions of both layers
or different from each other.[22–24] However, in some to share some common excipients. Additionally, it is a
cases, multi-layer tablets are not specifically developed good practice to keep the weight ratio of the two layers
for FDC products when there is only one API included not more than 1:6. The similarities of the weight and/or
as the middle layer sandwiched with two swellable composition of the two layers subsequently lead to simi-
and floatable composite matrix layers to provide a larities of the physical properties of the granular/powder
zero-order drug delivery or bimodal drug release pro- materials for tablet compression such as particle size and
files in the fast/slow/fast mode.[25,26] distribution, density, and flow. Consequently, it would
be more achievable to obtain the similarities of the tab-
Chemical instability and buffer layer  If the chemical let compaction profiles of the two layers, which benefits
stability of the APIs in a bi-layer tablet system is still not the physical integrity of a bi-layer tablet. It has also been
acceptable due to the interaction at the layer interface, discovered that different levels of layer swelling capac-
an alternative is to add a third buffer layer between the ity in a bi-layer tablet may lead to layer separation at the
two API-containing layers in order to further reduce interface, especially when exposed to a higher relative
the chemical reactions. For example, an alkaline layer humidity environment (e.g. relative humidity of 75% and

 Pharmaceutical Development and Technology


Formulation design and development of fixed dose combinations  1271
higher). The reason may be the increased level of shear adjusting the compression zone in the die, especially for
stress at the layer interface caused by the different extent the second layer, to a shallower position (closer to the
of swelling of individual layers (unpublished data). upper surface of the die) can reduce the occurrence of
the delamination/capping. Lastly, the layer of smaller
Process parameter and physical integrity  When different drug dose or press weight is considered to be compressed
formulations have to be used for each layer in a multi- first for the purpose of obtaining satisfactory API content
layer tablet, it is often imperative to adjust the tablet uniformity.
compression process in order to achieve satisfactory It is advisable to optimize the first layer compression
tablet physical quality attributes. Sufficient adhesion at force in order to minimize the delamination potential.
the interface of the two layers is necessary to maintain Subsequently, the second layer weight variation caused by
the physical integrity of a bi-layer tablet. Insufficient the volume variation of the first layer can be reduced. On
adhesion could lead to delamination which is defined the other hand, the sequence of layer compression is also
as the distinct layer separation along the interface. It was evaluated in combination with the compaction properties
found that the compression force for the first layer was of individual layers. Fortunately, the technology advance-
a critical factor affecting the propensity of bi-layer tablet ment in tablet press has offered great assurance in bi-layer
delamination (unpublished data). The function of the tablet compression which is one of the most important
first layer compression force, often in the range of 2–18 unit operations for this dosage form. Bi-layer tablet presses
kN, is to tamp the material in order to moderately reduce have been routinely used to manufacture commercial scale
the volume and smooth the surface of the first layer, and bi-layer tablets since 1990’s.[31] Many bi-layer tablet presses,
make room for deposition of the second layer material. such as Piccola, Oystar Manesty, Fette, Killian, Korsh, and
In general, higher compression force results in higher Curtory, are available to serve both product development
tensile strength of the first layer and smoother layer sur- and commercial production purpose. These tablet presses
face before the second layer material is fed into the die. are often featured with critical functions including pre-
However, a decrease of the surface roughness of the first compression/tamping station for each layer, automatic
layer sometimes results in an increase of the delamina- first layer sampling, or automatic layer weight control for
tion potential mainly by reducing the mechanical inter- all layers during multi-layer tablet compression.
locking of the interparticulate adhesion between the two
layers. Such phenomena have been generally observed Friability and tensile strength testing  As the physical
regardless whether both layers have the same or different integrity is a primary concern for multi-layer tablets, the
formulation compositions, or whether each formulation conventional tablet friability test using a friability tester is
consists of single ingredient (e.g. microcrystalline cellu- still widely used. There has been some progress in char-
lose) or multiple ingredients (e.g. microcrystalline cellu- acterizing multi-layer tablets. X-ray tomography was used
lose and lactose monohydrate) (unpublished data). as a non-destructive testing to visualize the delamina-
The delamination and capping could occur within one tion, cracks, voids, and other fracture patterns inside the
of the layers (intralayer), or between layers (interlayer). tablets and explore the mechanical failure mechanisms
The primary reasons for capping could be insufficient of multi-layer tablets by acquiring three-dimensional
cohesive/adhesive interaction among granules/ingredi- images for bi-layer tablets (unpublished data). The com-
ents, low level of moisture content, air entrapment dur- paction behavior of binary mixtures and bi-layer tablets
ing compression, or excessive compression force applied of microcrystalline cellulose and lactose was investigated
during compression. The delamination tendencies of with this method.[32] It was shown that for binary and bi-
bi-layer tablets have been reported by Podczeck[28] and layer tablets with the identical compositions consisting
especially the interfacial relaxation during tablet ejection of microcrystalline cellulose and lactose, the apparent
has been covered nicely by Anuar et al.[29] Another aspect tensile strength of the compact was generally comparable
of the bi-layer tablet compression is about the sequence for the powder systems studied and the fracture patterns
of layer compression for the same purpose of reducing depended upon the ingredient weight ratios and process
the potential of interlayer delamination or intralayer conditions including the sequence of the layer compres-
capping. A general rule of thumb for determining the sion. Another method, non-contact laser profilometry,
sequence of bi-layer tablets compression is to compress was used to characterize the topographic profiles and
the materials with higher fragmentation tendency as quantitative roughness of various surfaces of the bi-
the first layer, and the materials with more deformable layer tablets.[33,34] The results suggested that the localized
capacity as the second layer. The compaction character- stored energy released by volume expansion in the radial
istics of individual layers can be obtained by evaluating direction maybe responsible for the fracture of tablets
the granule/powder compactibility profiles such as the along the interface for a bi-layer tablet. Additionally,
compact tensile strength versus solid fraction curve.[30] the air-coupled acoustic technique, a non-contact/non-
Thirdly, it is an effective method to use one-way or two- destructive method, was used to determine the mechani-
way tapered die to reduce the level of air entrapment cal properties of the bi-layer tablets.[35] The mechanical
during bi-layer tablet compression. On the same token, properties, such as Young’s modulus, Poisson’s ratio, and
the use of pre-compression force for individual layers or mass density, were determined based on the resonance

© 2013 Informa Healthcare USA, Inc.


1272  D. Desai et al.
frequency measurements and computational calcula- also be combined to bring together an immediate release
tions. It is not only that this technique had the capability and extended release dosage form together. For example,
of detecting the weak bonding at the interface, but also extended release beads of propranolol can be combined
it had the potential to monitor a bi-layer tablet com- with an immediate release hydrochlorothiazide table in
pression process online. More in-depth understanding, a capsule.[2]
characterization, and modeling of the bi-layer tablets, In practice, many concepts of the development and
especially the establishment of a priori method in mate- manufacture of the multiparticulate dosage forms con-
rial selection and fracture mechanics prediction in for- taining one drug may be borrowed for FDC products. The
mulation and process development are of great interest common scheme of using multiparticulate system for
to many researchers. FDC products is to combine the immediate release and
controlled release components in one dosage form.[47]
Multiparticulate systems However, the applications have not been much used for
In theory, a multiparticulate system could be a good the FDCs, especially for marketed FDC drug products.
choice suitable for the development and manufacture The main contributing factors may lie in the fact that the
of FDC products. The ways of combining multiple APIs technical complexities of the product development and
to obtain various release profiles in one dosage form the requirements of more unique pellet manufacturing
presents fewer limitations from a pharmaceutical devel- equipment and process. Although the pelletization for
opment perspective. The most used multiparticulate the manufacture of multiparticulate drug delivery sys-
system for oral solid dosage form contains pellets and/ tems, such as extrusion-spheronization, is considered
or powders compressed into tablets or encapsulated well established, they are far from simple. Small changes
into capsules. One or several APIs may be introduced in the formulation and process may cause significant
into a pulsatile drug delivery system or alike during the effects on the quality attributes of final products.[48–50]
manufacturing process. The main technology of making Other technologies with less demanding requirements,
such systems is extrusion/spheronization to form pellets such as multi-layer tablets, have provided good alterna-
followed by one layer or multiple layers of film coating. tives for the multiparticulate dosage forms for FDC drug
Based on the physicochemical properties of the APIs and products.
excipients, many variations in this technology have been
used. The preparation of pellets or spherical granules by Hot-melt extrusion approach
extrusion/spheronization is a more established method HME has been well established in the plastic and food
with the advantages of narrower particle size distribution industries since 1930’s. It has gained increasing atten-
of the pellets with lower friability suitable for film coating tion in the last 10 years in the pharmaceutical industry.
from product manufacture stand point.[36] The extrusion/ Traditionally and currently, hot-melt extrusion (HME)
spheronization process includes four steps: granulation technology was mainly used to make amorphous drugs
to prepare the wet mass, extrusion to shape the wet mass in drug development. HME technology has been used
into cylinder-shaped extrudates, spheronization to break in pharmaceutical industry to make not only sustained
the extrudates and round off the particles into spheres, release formulations, but also immediate release for-
and drying of the pellets. The equipment requirements mulations. A new hot-melt granulation approach to
and manufacturing process critical points for the extru- densify the granulation and improve compressibility
sion/spheronization process, as well as for other meth- using HME technology has been reported as well.[51]
ods of making pellets, are well summarized by various It is well suited for the immediate release FDC prod-
researchers.[36–40] Another important aspect of making ucts containing one or two drugs of higher dose (e.g.
multiparticulate systems is how to incorporate APIs into 300–1000 mg). The drug may exist in either crystalline
the pellets. In most cases, the APIs may be mixed with the state or amorphous state depending on the process
ingredients during the process of making pellets.[41] conditions.
The main working principle of the melt granulation is
Pelletization by drug layering  Another way could be lay- to reduce polymer viscosities at higher temperature and
ering the drug solution or suspension onto sugar pellets, increasing its surface area resulting in improved com-
followed by coating of a swelling layer and a controlled pression characteristics of the granules containing drug
release layer.[42,43] Some ready-to-use pellets made with and polymer. The processing temperatures are typically
sugar (e.g. Suglets®), microcrystalline cellulose (e.g. set between the glass transition temperature (Tg) of the
Ethispheres®), or starch (e.g. Corn Starch Spheres®) are polymer and melting temperature of the drug substance.
also commercially available. Drug can also be incorpo- As a result, the drug substance remains in crystalline state
rated into the highly porous pellets (33.2% porosity) by in most cases. Several polymers like hydroxypropyl cel-
soaking the pellets into a drug solution or a drug-contain- lulose, hydroxypropylmethylcellulose, and Poloxamer®
ing supercritical fluid.[44] As a variation of the pellets with are commonly used in the melt granulation technology.
or without the controlled release film coat, mini-tablets Other excipients, especially plasticizers, can impact the
having immediate or controlled release profiles can be required process temperature significantly. The process
used in the multiparticulate systems.[45,46] The pellet can conditions, such as torque force and feeding speed, may

 Pharmaceutical Development and Technology


Formulation design and development of fixed dose combinations  1273
make actual process temperatures much higher than
the set temperature. Compared to other conventional
granulation technologies, the melt granulation is par-
ticularly suitable for the formulations with higher drug
loading requirements. This could be particularly benefi-
cial for some bi-layer FDC products if both drug doses
are high. In some cases, the drug loading was increased
from 50% w/w to 80–90% w/w by using the melt granu- Figure 3. Active film coating for fixed dose combination
lation technology. For example, if both drug doses are tablets.
300 mg in a bi-layer FDC product, the total tablet weight
was reduced from 1200 mg (50% w/w drug loading) to Compression sensitive molecules  Degradation of some
750–667 mg (80–90% w/w drug loading), which is more molecules is augmented by commonly used pharma-
patient friendly (unpublished data). ceutical operations such as dry or wet granulation,
It was also reported that the melt granulation dem- milling, or tablet compression. A FDC tablet formula-
onstrated enhanced chemical and physical stabilities for tion involving such a molecule is even more challeng-
moisture sensitive compounds.[52] The output of Leistritz ing. One possible solution is to formulate a traditional
50 mm extruder can reach 50–80 kg/h, and Leistritz tablet formulation for a molecule which is not sensitive
100 mm extruder can be even higher. Compared with to common pharmaceutical operations as the core
wet granulation, melt granulation has shown improved tablets followed by depositing the sensitive molecule
processability and quality control. However, the melt on the core tablets by active coating. By using such
granulation has its own disadvantages, such as the chal- approach, common pharmaceutical operations for a
lenges with thermo labile compounds, physical stability sensitive molecule can be avoided.[56]
of extrudates. Another frequently faced challenge is the
black specks in granules and tablets, which may be due Stabilization of acid and base labile molecules Some
to the prolonged excipient residence time in an extruder molecules are prone to acid as well as base catalyzed
during the processing, causing some charring. This can degradation. This challenge is further aggravated
be overcome through optimizing the formulation and when such a molecule needs to be formulated in a
process conditions. In addition, cleaning of the extruder FDC. One proposed solution is to make tablet core
is much more time consuming than for other granulation of the relatively stable molecule and to use an active
equipment. coating approach for the acid or base labile molecule.
The melt granulation technology has been success- One inherent advantage of the active coating is that
fully used in developing multiple commercial FDC prod- it provides relatively higher drug to excipient ratio
ucts by various pharmaceutical companies. As a new compared to having such molecule in the tablet core.
granulation approach, there is no clear decision process For example, 1 mg of the acid and base labile mol-
regarding the selection of polymers, plasticizers, and ecule in 100 mg core tablet will have drug to excipient
other excipients. More in-depth investigation and under- ratio of 1/100 compared to having such molecule in
standing of this technology is needed. 10 mg coating material. The later will have drug to
excipient ratio of 1/10. With higher drug to excipient
Active film-coating approach ratio, the number of excipient sites available to react
An active film-coating technology is often used to prepare a with the drug molecule is reduced providing better
FDC tablet formulations. For example in Claritin-D™, lorata- stability.[57]
dine and pseudoephedrine sulfate are coated on extended
release pseudoephedrine cores.[53] Upon oral administra- Challenges for active film coating  For traditional tab-
tion, the coating dissolves immediately to release loratadine let formulations developed using direct compression,
and pseudoephedrine to provide initial dose followed by wet granulation, or dry granulation technology, API is
extended release of pseudoephedrine from the tablet core weighed and mixed with other excipients as a part of the
matrix.[53] Similar approach was also adopted for Advicor™ manufacturing process. In the active coating approach,
where lovastatin was coated on extended release niacin API is sprayed on the tablet cores. There are two main
tablets.[54] One important advantage of the active coat- challenges in this approach. First, how to determine the
ing technology is that the release profiles of two different coating end point so that when the coating is stopped,
drugs are maintained.[55] There are FDC tablet formulations the tablets will have targeted potency. Second, given
where the main objective is to keep two actives apart to the variability inherent in the coating operation, how
minimize chemical interactions. In such cases, either one to make sure that all the tablets will have satisfactory
active could be included in the core tablets followed by the content uniformity.
active coating of another active. Alternatively, both actives
can be coated on the inert cores by applying two separate Determining the coating end point  For traditional cos-
coating layers separated by an inert coating layer, if needed metic coating, the coating end point is determined
as illustrated in Figure 3.[56] either based on the amount of suspension sprayed or

© 2013 Informa Healthcare USA, Inc.


1274  D. Desai et al.
the weight gain of the core tablets. This approach has second drug. This technology cannot be used for heat
been further modified for the active coating. During sensitive molecules. Moreover, the maximum amount
the active coating, tablet samples are taken periodically applied can be limited to 5 mg and there are specific
and analysed not only for weight gain but also for the requirements for core and coating materials in terms of
amount of API deposited by performing an in-process conductivity.
assay. As shown in Figure 4, after 50% coating, a linear
relationship has been observed between the weight Compression coating technology
gained and amount of API deposited.[56] Normally, at In this approach, a single tablet containing an immediate
about 75%–80% amount sprayed, the amount of API or controlled release drug core tablet is enveloped by a
deposited is determined. Based on that information, compression-coated compartment containing a second
additional amount of suspension needed to get the tar- drug with same or different in-vitro dissolution profile.
get potency is determined. This technology may be suitable for heat and solvent sen-
sitive drugs.
Content uniformity  For the traditional tablet formu-
lations, the amount of API needed for the batch is Co-processing approach
weighed and mixed with the formulation excipients The co-processing approach where two excipients or a
followed by dry or wet granulation. The API is locked drug and an excipient are crystallized together from a
in the granulation step with the rest of the excipients. solvent to either improve compressibility[64] or dissolu-
Since the API is intimately mixed with the excipients, tion, this approach has been extended for the develop-
the proper control on the weight variation of the tablet ment of FDC. The main challenge in this approach is
ensures satisfactory tablet content uniformity. On the to identify solvent and anti-solvent which do not alter
other hand, in the active coating, the API is sprayed polymorphic form of the drug substances. Mohammed
along with the coating materials on already formed core et  al. used the co-processing approach to combine
tablet. Therefore, the factors governing the content uni- nevirapine and stavudine to overcome content unifor-
formity of active coated tablets are very much different mity issue. These two co-processed drugs were com-
than those of the traditional tablets.[58] Control on the bined with lamivudine to form a triple combination
spraying operation is vital for the satisfactory content tablet.[65]
uniformity of the finished products. Essentially, when-
ever tablets come in front of the spray zone, the API Enrobing technology
and coating materials get deposited on them. Recent This patented technology from the Banner Technology
development in the active coating technology has enrobes tablets with either a gelatin or material with
made it possible for formulators to use this technology non-animal origin.[66] The enrobement process uses coat-
to develop FDCs.[59–63] ing die techniques in which the tablets to be enrobed
are introduced individually between two sealable films
Electrostatic coating positioned between opposing matching dies. With the
This technology was developed by the Phoqus Company. flexibility to include the second drug included in the
Tablet core with some conductive property with one of enrobing film, this technology offers proprietary imme-
the FDC drugs is prepared. Using electro-magnetic field, diate/modified release technologies for incorporation of
the opposite charge is applied to another drug with some drug in the FDC products.
coating material. Since the charge of the core and coating
is opposite, the coating gets deposited on the core tab- Liquid dispensing technology
lets electro statically. The coated tablets are heated up to This is a proprietary technology developed by Glaxo
about 70–100°C for the coating adhesion. This technique Smith Kline(GSK).[67] In this technology, the tablet can
has been shown to be able to deposit 10 mcg–5 mg of the be an inert core or may contain an API. On one side of
the core surface, there is a small well or recess. In this
well, drug and polymer dissolved in an organic solvent
is deposited using a micro pipette. The organic sol-
vent evaporates leaving behind a thin film of drug and
polymer. On this film, just coating material dissolved
in an organic solvent is deposited. The organic solvent
evaporates leaving behind a thin film coat. Similar
procedures can be repeated on the other side of the
tablet core. Thus, FDC of two drugs can be developed,
if desired, with the inclusion of one or more drugs in
tablet core, a triple combination can be developed. This
technology is very suitable for potent molecules where
Figure 4. Middle (active) layer monitoring (2.5 mg API coated drug containment is highly desired since drug is always
tablets). deposited in solution.

 Pharmaceutical Development and Technology


Formulation design and development of fixed dose combinations  1275

Conclusion symptoms recommendations for clinical evaluation. Rockville,


MD, USA.
From a clinical perspective, aging population in many 16. FDA Guidance for Industry. (2006). Fixed Dose combinations,
developed countries will need multiple medications to treat Co-Packaged Drug Products, and Single-Entity Versions of Previously
Approved Antiretrovirals for the Treatment of HIV. Rockville, MD, USA.
age related diseases and co-morbidities. FDC products 17. Guidance for Industry. (1999). Application covered by section
will continue to be a useful tool to combine medications 505b2, CDER, Editor. Rockville, MD.
with different mode of action to treat disease conditions 18. Woodcock J, Griffin JP, Behrman RE. Development of novel
with fewer dosing units. As outlined in the article, the combination therapies. N Engl J Med 2011;364:985–987.
number of FDC products has grown over the years and the 19. FDA Guidance for Industry. (2010). Co-development of Two or
More Unmarketed Investigational Drugs for Use in Combination.
trend is likely to continue. With the increase in growth of Rockville, MD, USA.
the FDC products, new formulation approaches will also 20. World Health Organization Expert Committee. (2005). Fixed-dose
be invented by pharmaceutical companies. It is hoped that combination medicinal products. Geneva, 94–142.
this review article will give an overview to pharmaceutical 21. Wechsler J. Combination products raise manufacturing challenges.
scientists about the challenges and opportunities in the Pharmaceut Tech 2005;29:32–40.
22. Nirmal J, Saisivam S, Peddanna C, Muralidharan S, Godwinkumar
formulation development of the FDC products.
S, Nagarajan M. Bilayer tablets of atorvastatin calcium and
nicotinic acid: formulation and evaluation. Chem Pharm Bull
2008;56:1455–1458.
Declaration of interest 23. Shiyani B, Gattani S, Surana S. Formulation and evaluation of
The authors report no conflicts of interest. bi-layer tablet of metoclopramide hydrochloride and ibuprofen.
AAPS PharmSciTech 2008;9:818–827.
24. Mandal U, Pal TK. Formulation and in vitro studies of a fixed-dose
combination of a bilayer matrix tablet containing metformin HCl
References as sustained release and glipizide as immediate release. Drug Dev
Ind Pharm 2008;34:305–313.
 1. Luo Y, Zu Y, Ahmad S. Challenges of fixed dose combination 25. Liu Q, Fassihi R. Zero-order delivery of a highly soluble, low dose
product development. American Pharmaceutical Review 2007, drug alfuzosin hydrochloride via gastro-retentive system. Int J
10(2): 120–126. Pharm 2008;348:27–34.
  2. Timmins P. (2006). Developing fixed combination drug products– 26. Streubel A, Siepmann J, Peppas NA, Bodmeier R. Bimodal drug
Technical challenges and opportunities. In: The 10th Annual Drug release achieved with multi-layer matrix tablets: transport
Delivery Partnerships. Phoenix, AZ, USA. mechanisms and device design. J Control Release 2000;69:455–468.
  3. Pudipeddi M. (2010). Biopharmaceutical challenges in 27. Benkerrour L, Galley O, Quinet F, Abebe A, Timmins P. Multilayered
development of fixed-dose combinations. In: Eastern tablet containing pravastatin and aspirin and method, 2004, US
Pharmaceutical Technology Meeting. Somerset, New Jersey, USA. patent, US2004/0115265.
  4. Karali TT. Available at: (http://www.pharmacircle.com/ 28. Podczeck F. Theoretical and experimental investigations into
presentations/fixed dose.pdf ) the delamination tendencies of bilayer tablets. Int J Pharm
 5. Gupta AK, Arshad S, Poulter NR. Compliance, safety, and 2011;408:102–112.
effectiveness of fixed-dose combinations of antihypertensive 29. Anuar MS, Briscoe BJ. Interfacial elastic relaxation during the
agents: a meta-analysis. Hypertension 2010;55:399–407. ejection of bi-layered tablets. Int J Pharm 2010;387:42–47.
  6. Bailey CJ, Day C. Fixed-dose single tablet antidiabetic combinations. 30. Tye CK, Sun CC, Amidon GE. Evaluation of the effects of tableting
Diabetes Obes Metab 2009;11:527–533. speed on the relationships between compaction pressure, tablet tensile
  7. Serebruany VL, Malinin AI, Sane DC, Jilma B, Takserman A, Atar D, strength, and tablet solid fraction. J Pharm Sci 2005;94:465–472.
Henneckens CH. Aggrenox and aspirin in patients after ischemic 31. Li SP, Karth MG, Feld KM, Di Paolo LC, Pendharkar CM, Williams
stroke: the Aggrenox versus aspirin therapy evaluation trial. Eur J RO. Evaluation of bilayer tablet machines–a case study. Drug Dev
Pharmacol 2004;3:315–324. Ind Pharm 1995;21:571–590.
  8. Easton J, Noble S, Perry CM, Cuffini AM, Jacobs MR, Subba Rao SD, 32. Wu CY, Jonathan P,Seville K. A comparative study of compaction
Pichichero ME, Thornberry C. Amoxicillin/clavunic acid: a review properties of binary and bilayer tablets. Powder Tech
of its use in the management of paediatric patients with acute otitis 2009;189:285–294.
media. Drugs 2003;23:311–340. 33. Inman SJ, Briscoe BJ, Pitt KG, Shiu C. The non-uniformity of
 9. Vicknasingam B, Mazlan M, Schottenfeld RS, Chawarski MC. microcrystalline cellulose bilayer tablets. Powder Tech 2009;
Injection of buprenorphine and buprenorphine/naloxone tablets 188:283–294.
in Malaysia. Drug Alcohol Depend 2010;111:44–49. 34. Inman SL, Briscoe BJ, Pitt KG, Shiu C. Topographic characterization
10. Coleman JJ, Schuster CR, Dupont RL. Reducing the abuse potential of cellulose bilayered tablets interfaces. Chem Eng Res Des 2007;
of controlled substances. Pharmaceut Med 2010;24:21–36. 85:1005–1012.
11. World Health Organization Expert Committee. (2000). The use of 35. Akseli I, Dey D, Cetinkaya C. Mechanical property characterization
essential drugs including the revised model list of essential drugs. of bilayered tablets using nondestructive air-coupled acoustics.
Geneva. AAPS PharmSciTech 2010;11:90–102.
12. Friedland GH, Williams A. Attaining higher goals in HIV treatment: 36. Gandhi R, Lal Kaul C, Panchagnula R. Extrusion and spheronization
the central importance of adherence. AIDS 1999;13 Suppl in the development of oral controlled-release dosage forms. Pharm
1:S61–S72. Sci Technol Today 1999;4:160–170.
13. Dublin CH. Combination products: strategies for continuous 37. Gainotti A, Bettini R, Gazzaniga A, Colombo P, Giordano F. Drug-β-
pipeline. Drug Deliv Tech 2005;5:41–46. cyclodextrin containing pellets prepared with a high-shear mixer.
14. Lacy CF, Armstrong LL, Goldman MP, Lance LL. (2006). Drug Drug Dev Ind Pharm 2004;30:1061–1068.
Information Handbook. 14th edition ed. Hudson, Ohio, USA: Lexi- 38. Mezreb N, Charrueau C, Boy P, Allain P, Chaumeil JC. Production
Comp Inc. of Carbopol 974P and Carbopol 971P pellets by extrusion-
15. FDA Guidance for Industry. (2003). Estrogen/progestin drug spheronization: optimization of the processing parameters and
products to treat vasomoto symptoms and vulvar vaginal atrophy water content. Drug Dev Ind Pharm 2004;30:481–490.

© 2013 Informa Healthcare USA, Inc.


1276  D. Desai et al.
39. Poelvoorde N, Huyghebaert N, Vervaet C, Remon JP. Optimisation 53. Kwan H, Liebowitz S. (1994). Stable extended release oral dosage
of an enteric coated, layered multi-particulate formulation for ileal composition comprising loratidine and pseudoephedrine. United
delivery of viable recombinant Lactococcus lactis. Eur J Pharm States, 2009, US Patent 5314697.
Biopharm 2008;69:969–976. 54. Bova J, DunneJ. Combinations of HMG-COA reductase
40. Abdul S, Chandewar AV, Jaiswal SB. A flexible technology for inhibitors and nicotinic acid compounds and methods for
modified-release drugs: multiple-unit pellet system (MUPS). J treating hyperlipidemia once a at night, 2009, Publication No:
Control Release 2010;147:2–16. WO/1999/006046
41. Kok PJAH, Von kP, Hoekzema MA, Kossen NWF. Development 55. Charlton ST, Nicholson SJ. A novel immediate release active film
of particulate pulse-release formulation and their mathematical coat formulation for use with an extended release gel matrix tablet
description. Powder Tech 2001;119:33–44. core. J Pharm Pharmacol 2010;62:1397–1398.
42. Dashevsky A, Mohamad A. Development of pulsatile 56. Lipper RD, Desai D,Kiang S. (2006). Case Study: Implementation of
multiparticulate drug delivery system coated with aqueous Design Space. In: Real World Applications of PAT and QbD in Drug
dispersion Aquacoat ECD. Int J Pharm 2006;318:124–131. Process Development and Approval. Arlington, Virginia, USA.
43. Baki G, Bajdik J, Djuric D, Knop K, Kleinebudde P, Pintye-Hódi K. Role 57. Desai D, Rao V, Guo H, Li D, Stein D, Hu FY et al. An active film-
of surface free energy and spreading coefficient in the formulation coating approach to enhance chemical stability of a potent drug
of active agent-layered pellets. Eur J Pharm Biopharm 2010;74: molecule. Pharm Dev Technol 2010;11:1–9.
324–331. 58. Carstensen JT, Koff A, Johnson JB, Rubin SH. Tablet-to-tablet
44. Cosijns A, Nizet D, Nikolakakis I, Vervaet C, De Beer T, Siepmann F variation of drug content of sugar-coated tablets containing drug
et al. Porous pellets as drug delivery system. Drug Dev Ind Pharm in the sugar coat. J Pharm Sci 1970;59:553–555.
2009;35:655–662. 59. Chen W, Chang SY, Kiang S, Marchut A, Lyngberg O, Wang J et al.
45. Kranz H, Le Brun V, Wagner T. Development of a multi particulate Modeling of pan coating processes: prediction of tablet content
extended release formulation for ZK 811 752, a weakly basic drug. uniformity and determination of critical process parameters. J
Int J Pharm 2005;299:84–91. Pharm Sci 2010;99:3213–3225.
46. Riis T, Bauer-Brandl A, Wagner T, Kranz H. pH-independent drug 60. Hagrasy AE, Chang SY, Desai D, Kiang S. Application of Raman
release of an extremely poorly soluble weakly acidic drug from Spectroscopy for quantitative in-line monitoring of tablet coating.
multiparticulate extended release formulations. Eur J Pharm Am Pharmaceut Rev 2006;9:40–45.
Biopharm 2007;65:78–84. 61. Hagrasy AE, Chang SY, Desai D, Kiang S. Raman Spectroscopy for
47. Qiu Y, Gupta P, Briskin J, Cheskin H, Semla S. Sustained-release the determination of coating uniformity of tablets: assessment of
multiparticulate formlations of Zileuton. I. In vitro and in vivo product quality and coating pan mixing efficiency during scale-up.
evaluation. Int J Pharm 1996;143:179–185. J Pharmaceut Innovat 2006;1:37–42.
48. Wesdyk R, Joshi YM,Vincentis JD,Newman AW,Jain NB. Factors 62. Chen W, Chang SY, Kiang S, Early W, Paruchuri S, Desai D.
affecting differences in film thickness of beads coated in fluidized The measurement of spray quality for pan coating processes. J
bed units. Int J Pharm 1993;93:101–109. Pharmaceut Innovat 2008;3:1872–1875.
49. McConnell EL, Macfarlane CB, Basit AW. An observational study 63. Fichana D, Marchut AJ, Ohlsson PH, Chang SY, Lyngberg O, Dougherty
on the influence of solvent composition on the architecture of J et  al. Experimental and model-based approaches to studying
drug-layered pellets. Int J Pharm 2009;380:67–71. mixing in coating pans. Pharm Dev Technol 2009; 14:173–184.
50. Talukder R, Fassihi R. Gastroretentive delivery systems: hollow 64. Gohel MC, Jogani PD. A review of co-processed directly
beads. Drug Dev Ind Pharm 2004;30:405–412. compressible excipients. J Pharm Pharm Sci 2005;8:76–93.
51. Vasanthavada M, Wang Y, Haefele T, Lakshman JP, Mone M, Tong 65. Mohammed GA, Puri V, Bansal AK. Coprocessing of
W et  al. Application of melt granulation technology using twin- nevirapine and stavudine by spray drying. Pharm Dev Technol
screw extruder in development of high-dose modified-release 2008;13:299–310.
tablet formulation. J Pharm Sci 2011;100:1923–1934. 66. Sadek H, Dietel GL. Film-enrobed unitary-core medicament and
52. Kowalski J. (2008). Novel opportunities for solid oral dosage the like. U.S. Patent 5,146,730. Editor 1992.
form development using a twin screw extruder. In: American 67. Li Y, Clarke AJ. (2009). A novel liquid dispensing technology for
Association of Pharmaceutical Scientist Annual Meeting. Atlanta, manufacturing low dose immediate release tablets. In: 2009 AAPS
GA, USA. Annual meeting.

 Pharmaceutical Development and Technology

You might also like