Fixed Dose Combination (FDC)
Products: Introduction, Development and Regulations
Hamid
Khan1*, Javed Ali2
1JK College of Pharmacy, Bilaspur,
CG, India-495001
2Department of Pharmaceutics, Faculty of Pharmacy, Hamdard University, New Delhi, India-110062
*Corresponding
Author E-mail: khanhamid770@gmail.com
ABSTRACT:
A fixed dose combination (FDC) is a formulation of two
or more active ingredients combined in a single dosage form available in
certain fixed doses. Combination therapy with two or more agents having
complementary mechanisms of action represents a type of incremental innovation
that has extended the range of therapeutic options in the treatment of almost
every human disease. Combining two or more active pharmaceutical ingredients in
a single-dosage form can increase a drug’s efficacy and improve patient
compliance. Several difficulties arise during formulation development,
manufacturing and regulations of FDC products. Two or more active ingredients
in the FDCs must be physically and chemically compatible along with their excipients. In this review article authors provided the
brief information regarding advantages, formulation development and regulations
of FDC products.
KEYWORDS:
INTRODUCTION:
According
to WHO definition, new fixed-ratio combination products are regarded as new
drugs in their own right and they are acceptable only when (a) the dosage of
each ingredient meets the requirements of a defined population group, and (b)
the combination has a proven advantage over single compounds administered
separately in terms of therapeutic effect, safety or compliance. They should
not be treated as generic versions of single-component products [1].
Combination therapy is commonly used in treatment of almost every area of
diseases, especially hypertension, tuberculosis, malaria, diabetes and pain
management, etc. Ideally, combination products can provide a synergistic effect
of individual drugs with reduced side effects. From a compliance point of view,
combination products provide a single pill, reducing the number of pills taken
on a daily basis and therefore enhancing patient compliance.
Reducing the number of pills diminishes the complexity
of the regimen, so that improved patient adherence is expected with combination
products. To combine drugs belonging to different classes and having mechanisms
of action that is complementary to each other, for example antihypertensive
drugs, Telmisartan and hydrochlorothiazide. Using FDC
products in tuberculosis and malaria control simplifies the doctor's
prescription and patient's drug intake, helps patients adhere to treatment,
precludes inadvertent monotherapy by the patient and
therefore prevents the emergence of drug resistance due to missed dose of a
constituent drug for example, Isoniazide and Rifampicin. Several fixed-dosed FDC products are
commercially available with complementary mechanisms of action to improve glycemic control in type 2 diabetes patients, for example: Metformin and Glimepiride, Pioglitazone and glimepiride etc.
FDC products of two or more analgesics with different modes of action activate
multiple pain-inhibitory pathways and thus provide more effective pain relief,
for example: Aceclofenac and Paracetamol, Diclofenac
and Paracetamol etc. However some disadvantages are also associated with FDC
products such as dosage alteration of one drug is not possible without
alteration of the other drug, i. e. dose adjustment
to the patient’s needs. Some FDCs when combined leads to increased toxicity,
example, anti-TB drugs have the side effects oto and nephro-toxicity. If the biological half-life of different
compounds in FDC is different, it may considerably affect the pattern of drug
availability in the plasma, and hence the over all
efficacy of the product is affected, example, Rifampicin
FDC products [2-7].
Development of FDC Products
In relation to formulation development very similar
principles apply to fixed dose combination finished products as apply to single
entity products. However, there are additional complexities arising due to the
presence of two or more drug substances. These complexities are related to
compatibility of drug substances, assay, stability, physicochemical properties
(for example dissolution rate) and bioavailability/bioequivalence, analytical
methods and acceptance criteria for impurities, stress testing and
determination of shelf lives of FDC products. Chemical and physicochemical
compatibility of the APIs in an FDC with one another as well as with possible excipients. Physicochemical properties and stability of
each of the drug in an FDC will define the formulation approaches to maintain
stability during the shelf life of the combined drug product. For example, the hygroscopicity of one active drug substance can influence
the stability of the other drug in the combination product unless they are
completely separated by way of multiple techniques. On the basis of formulation strategy, the
manufacturing of an FDC can be as simple as mixing the two or more active drug
substances provided that they are physically and chemically compatible and
stable with excipients required for encapsulation, or
tableting and compression followed by coating or
encapsulation. If the active drugs are incompatible with each other, they would
need to be formulated in a single pill using a number of different options,
such as multilayer tablets, compression-coated tablet, multiparticulates
in capsules, combinations of coated and uncoated beads, or pellets of one placed
with a powder of another active. The manufacturing process is more complex with
bilayer, three-layer, or tablet-in-a-tablet
manufacturing. This could involve multiple granulations and compressing these
layers using suitable multilayer tablet presses or a special press for tablet
in a tablet [1, 8].
Analytical
method development for FDCs
Method development by HPLC-UV for two or more
compounds and their related impurities becomes very complex if the UV profiles
are not similar. Hence need to be analyzed by other sophisticated techniques
such as LC-MS, UPLC-PDA, UPLC /Q-TOF-MS etc. Developing and validating a
stability-indicating assay method becomes more challenging when multiple drugs
are present in a drug product. Since developing and marketing new chemical
entities (NCEs) for multiple indications is difficult task, pharmaceutical
companies are looking into creating products by combining two or more known,
compatible APIs to treat multiple diseases and achieve better patient
compliance. Method development for two or more compounds and their related
impurities becomes very complex if the solubility and the pKa
values vary greatly and the UV profiles are not similar. If the solubility and
the pKa values of the APIs involved are similar,
where all the active components are totally soluble in water, the method
development is much easier [9-11]. In the case of two APIs, new impurities seen
in stability samples, the samples must be evaluated carefully and the
impurities reported to their origins. For routine analysis in a stability
program, a stability-indicating method is required for analyzing both the API
and impurities. Analytical methods should be validated for each active
ingredient in the presence of related process impurities and degradation
products. The interference by degradation products can be controlled by peak
purity testing by HPLC-UV, HPLC-PDA, HPLC-MS, UPLC-PDA and UPLC/QTOF
techniques. The acceptance criteria for impurities in FDC products are
expressed with reference to the parent active ingredient and not with reference
to the total content of active ingredients. During stress testing, the active
ingredients are combined in the same ratio as in the final product. The expiry
date is determined on the basis of stability of the least stable active
ingredient [12-17].
Regulation of FDC products
The Drug Controller General of India (DCGI) had given
marketing approvals for 40 FDCs in January 2002. It is an accepted fact that
FDC is treated as a new drug, because by combining two or more drugs, the safety,
efficacy, and bioavailability of the individual active pharmaceutical
ingredient (API) may change. As per the Drugs and Cosmetic Act, 1940, any new
drug and the permission to market a drug is to be given by the DCGI. Before the
approval of any drug, the Central Drugs Standard Control Organization (CDSCO)
undergoes a process with respect to their quality, safety and efficacy. The
DCGI monitors the drug formulations, including the combinations of drugs, from
the angle of safety, effectiveness and rationality. In India, the CDSCO is the
main controlling department for drug approval. The CDSCO regulates this process
through DCGI. List of some
marketed FDCs approved by DCGI is presented in Table 1. The DCGI
controlling the drug regulations through drug commissioners and drug inspectors
in different states of India. Recently CDSCO, adopted guidelines for industry
on fixed dose combinations in April 2010. These guidelines provide the
information about various aspects of formulation, manufacture and marketing of
FDC products in India [8].
Internationally, there is an increasing trend to license fixed-dose
combination products for the market. Currently there are no specific
international guidelines for FDCs but WHO published the guidelines for
registration of fixed-dose combination medicinal products in the year 2005.
Some national authorities have developed their own guidelines, some for
specific classes of medicines [1]. Guidelines for registration of FDCs products
are summarized in Table 2.
Table 1. List of some marketed FDCs approved by DCGI
Guidelines |
Description |
Scientific and technical principles for FDC
products. Botswana, 22 April 2004 |
Described the registration, quality, efficay, and safety requirements for FDCs. |
Fixed-combination medicinal products. CPMP April
1996, CPMP/EWP/240/95, II/5773/94 formerly known as Testing combination; and
licensing criteria for FDC products. |
Require circumstances in which FDCs are
acceptable, describe the considerations of pharmacokinetic and pharmcodynamic interactions, evidence for safety and
efficacy. |
WHO guidelines for registration of fixed-dose
combination medicinal products. (Annex 5, 39th report). TRS No. 929, Geneva,
2005 |
Described the advantages, disadvantages, quality,
safety and marketing authorization of FDCs. |
Guidance for industry on fixed dose combinations
(FDCs), CDSCO, New Delhi, April 2010 |
Guidelines to manufacture, import, and marketing
approval of FDCs as per Drugs and Cosmetics Act and Rules. |
Table 2. Guidelines for
registration of FDCs products
Category |
FDC products |
Category |
FDC products |
Antihypertensive |
Losartan + Hydrochlorothiazide tablets Valsartan + Hydrochlorothiazide
tablets Telmisartan + Hydrochlorothiazide
tablets Atenolol + Hydrochlorothiazide tablets |
Hypercholesterolemia (Lipid
lowering) |
Atorvastatin + Ezetmibe
tablets Simbastatin + Ezetmibe
tablets |
Antidiabetics |
Glipizide + Metformin
tablets Glimepiride + Metformin
tablets Pioglitazone + Metformin
tablets Pioglitazone +Glimepiride
+ Metformin tablets |
Antitubercular |
Isoniazide + Rifampicin
tablets Isoniazide + Ethambutol
tablets Isoniazide + Rifampicin
+ Ethambutol tablets |
NSAIDS, Analgesic and antipyretic |
Diclofenac
+ Paracetamol tablets Ibuprofen
+ Paracetamol tablets Aceclofenac +
Paracetamol tablets |
Antimalarials |
Sulfadoxine + Pyrimethamine
tablets Artemether + Lumefantrine
tablets |
CONCLUSION:
The FDC products provide the increased drug efficacy
and improved patient compliance. Drugs in FDC products provide complementary
mechanism of action to each other, therefore extended range of therapeutic
effects can be achieved. Several pharmaceutical companies now investing for the
development and marketing these products. The use of FDC products becoming
worldwide, hence the systematic approach is required for the design, development
and its regulations. For the development of FDC products several factors must
be taken in to consideration such as Chemical and physicochemical compatibility
of the drug substances in an FDC with one another as well as with possible excipients. Other factors are their stability,
bioavailability, analytical methods, acceptance criteria for impurities and
determination of shelf lives of FDC products. In the presented review article,
advantages of FDC products have been discussed. Strategies for formulation
development, analytical method development and regulatory guidelines for FDC
products have been described in brief.
REFERENCES:
1.
WHO
Guidelines for registration of fixed-dose combination medicinal products. 2005,
Available from http://www.who.int/medicines/publications/pharmprep/TRS_937.pdf.
2.
Chakraborti A. Fixed dose combinations in therapy. Express Pharma. 2 (19); 2007: 62-63.
3.
Sen A. Indian market's fixation with fixed dose combinations. Rational Drug
Bulletin. 12 (1); 2002: 1-2.
4.
Gautam CS, Saha L. Fixed dose drug combinations
(FDCs): rational or irrational: a view point. British Journal of Clinical
Pharmacology. 65(5); 2008:795-796.
5.
Panda
J, Tiwari P, Uppal R.
Evaluation of rationality of some FDC: Focus on antihypertensive drugs. Indian
Journal of Pharmaceutical Sciences. 68(5); 2006: 649-653.
6.
Sreedhar D, Janodia MD, Ligade VS, Mohapatra S, Ganguly R, Udupa N. Fixed dose
combinations: Rational or irrational. Current Science. 95 (5); 2008: 581-583.
7.
Wertheimer
AI, Morrison A. Combination drugs: Innovation in pharmacotherapy. Pharmacology
and Therapeutics. 27 (1); 2002: 44-49.
8.
Draft
guidelines on fixed dose combination products. Central drugs standard control
organization (CDSCO). Directorate general of health services, ministry of
health and family welfare, govt. of India. April 2010; CDSCO, Delhi, India.
9.
International
Conference on Harmonization, ICH Q2 (R1), Validation of Analytical Procedures:
Text and methodology, Geneva, 2005.
10. International Conference on Harmonization,
ICH Q1A (R2), Stability testing of New Drug Substances and Products, Geneva, 2003.
11. Singh S, Bakshi M.
Development of stability-indicating assay methods. Journal of Pharmaceutical
and Biomedical Analysis. 28 (6); 2002: 1011-1040.
12. Lee H, Shena S, Grinberg N. Identification and control of impurities for
drug substance development using LC/MS and GC/MS. J. Liq. Chromatogr.
Relat. Technol. 31(15); 2008: 2235-2252.
13. Ahuja S. Impurities evaluation of
pharmaceuticals, Marcel Dekker, New York. 2006.
14. Qiua F, Norwood DL. Identification of pharmaceutical
impurities. J. Liq. Chromatogr. Relat.
Technol. 30(5); 2007: 877-935.
15. Vyas VK, Ghate M, Ukawala RD. Recent advances in characterization of
impurities-Use of hyphenated LC-MS technique. Current Pharmaceutical Analysis.
6(4); 2010: 299-306.
16. Roy J. Pharmaceutical impurities- A review.
AAPS Pharm. Sci. Tech. 3(2); 2002:1-8.
17. Rahman N, Azmi SN, Wu
HN. Importance of impurity analysis in pharmaceutical products. Accred. Qual. Assur. 11(2); 2006:
69-74.
Received on 21.05.2016 Modified on 15.06.2016
Accepted on 05.07.2016 ©A&V Publications All right reserved
Res. J. Pharm.
Dosage Form. & Tech. 2016; 8(3): 207-210.
DOI: 10.5958/0975-4377.2016.00028.8