Microsoft word - tech corner article

Bioequivalence of Topical Dermatological Drug Products
Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, 3001 Mercer University Drive, Mercer University, Atlanta, GA 30341 *To whom correspondence should be addressed: 1. Introduction
Topical dosage forms are liquid or semisolid dosage forms, which are not intended for systemic absorption. These dosage forms comprise solutions, lotions, gels, ointments, patches, and foams; that are applied onto the skin either to elicit therapeutic effect within the The Code of Federal Regulations: 21 CFR § 320.1 has the following definitions: a) Drug product means a finished dosage form, e.g., tablet, capsule, or solution, that
contains the active drug ingredient, generally, but not necessarily, in association with b) Bioavailability (BA) is the rate and extent to which the active ingredient or active moiety
is absorbed from a drug product and becomes available at the site of action. For drug products that are not intended to be absorbed into the bloodstream, BA may be assessed by measurements intended to reflect the rate and extent to which the active ingredient or active moiety becomes available at the site of action. c) Pharmaceutical equivalents (PE) means drug products in identical dosage forms that
contain identical amounts of the identical active drug ingredient, i.e., the same salt or ester of the same therapeutic moiety, or, in the case of modified release dosage forms that require a reservoir or overage or such forms as prefilled syringes where residual volume may vary, that deliver identical amounts of the active drug ingredient over the identical dosing period; do not necessarily contain the same inactive ingredients; and meet the identical compendial or other applicable standard of identity, strength, quality, and purity, including potency and, where applicable, content uniformity, disintegration times, d) Pharmaceutical alternatives (PA) means drug products that contain the identical
therapeutic moiety, or its precursor, but not necessarily in the same amount or dosage form or as the same salt or ester. Each such drug product individually meets either the identical or its own respective compendial or other applicable standard of identity, strength, quality, and purity, including potency and, where applicable, content uniformity, disintegration times and/or dissolution rates. e) Bioequivalence (BE) is the absence of a significant difference in the rate and extent to
which the active ingredient or active moiety in pharmaceutical equivalents (PE) or
pharmaceutical alternatives (PA) becomes available at the site of drug action when
administered at the same molar dose under similar conditions in an appropriately Establishing BE for Topical Dermatological Drug Products has been a topic of discussion for many years between scientific community and the regulatory agency. Despite great advances in addressing the issues related to topical bioequivalence, many challenges remain due to the complexity of drug transport through the skin from different formulations and lack of harmonized guidance documents. Siewert et al. (2003) have acknowledged that no single test procedure would be suitable for the development, biopharmaceutical characterization, and quality control of all semi-solid topical dosage forms. The FDA’s current approval strategy for most of the topical drug products (New Drug Application/Abbreviated New Drug Application) is based on clinical studies. Clinical trials to prove BE often lack sensitivity and require large and costly trials (FDA’s Critical Path Initiative § 4.3.3, 2004; Narkar, 2010). It may not be cost effective for generic manufacturers to conduct large clinical trials. Several surrogate methods have been in development to address this issue as summarized in this review. However, adoption by industry is limited. The reason could be less competition for a relatively smaller market of topical dosage forms when compared to other dosage forms. The Federal Trade Commission (FTC) has put conditions on Novartis AG's Acquisition of Fougera Holdings, Inc. in order to protect the
competition in the skin care market for certain products. The scope of this review is to summarize different methods that can facilitate establishing BE of Topical Dermatological Drug Products. 2. Regulations Governing Bioequivalence in USA
The Code of Federal Regulation (CFR), Title 21, describes how the Food and Drug Administration (FDA) regulates food, drugs, cosmetics, biologics, tobacco products, veterinary products, radiation emitting products, and medical devices in the US. As part of Department of Health and Human Services (DHHS), the FDA’s mandate is to protect and promote public health. There have been significant amendments to the Food, Drug and Cosmetics legislation ever since the Sulfanilamide Elixir tragedy in 1937 (Skelly, 2009; Table 1). The US enacted the Drug Price Competition and Patent Term Restoration Act of 1984 (DPCA 1984), widely known as the Hatch-Waxman Act, to lower the raising costs of prescription drugs by increasing the competition among manufacturers. The generic industry has flourished by the virtue of this legislation. However, due to lack of reliable alternative methods, clinical studies are required to establish BE of Topical Dermatological Drug Products (FDA’s Critical Path Initiative § 4.3.3, 2004). Topical BE has been a topic of much discussion among researchers, industry, and agency (Table 1). Table 1 Regulatory information pertaining to BE of Topical Dermatological Drug Products1
Year Comments
The Federal Food, Drug, and Cosmetic Act (FD&C Act) – required evidence of safety for market approval of new drug products The Kefauver-Harris Amendments established drug safety and effectiveness requirement from the manufacturers The Drug Price Competition and Patent Term Restoration Act of 1984 – Title I: Abbreviated New Drug Application; Title II: Patent Extension (Hatch-Waxman Act) Interim Guidance, Topical Corticosteroids: In Vivo BE and In Vitro Methods The FDA’s Guidance for Industry: Nonsterile Semisolid Dosage Forms Scale-Up and Post Approval Changes: Chemistry, Manufacturing, and Controls; In Vitro Release Testing and Bioequivalence (SUPAC-SS) The FDA’s Draft Guidance-Not for Implementation, Topical Dermatological Drug Product NDAs and ANDAs — In Vivo Bioavailability, Bioequivalence, In Vitro Release, and Associated Studies FDA Draft guidance on the Skin stripping method withdrawn due to contradictory results from two independent laboratories FDA’s Critical Path Initiative: To drive innovation in the scientific processes through which medical products are developed, evaluated, and manufactured. FDA Guidance recommends in vivo bioequivalence of topical dermatologic corticosteroids based on pharmacodynamics approach (Stoughton-McKenzie vasoconstrictor assay) FDA’s Critical Path Opportunities for Generic Drugs: 4.3.3 BE of Topical 3Mar 13 Workshop on the evaluation of Topical Drug Products  Development and 1http://www.fda.gov/RegulatoryInformation/default.htm 2Described DPK methodology for establishing BE for topical products 3Expected date; organized by Product Quality Research Institute (PQRI) 3. BE of Topical Dermatological Drug Products
In order to get an approval to market a generic copy of “pioneer or innovator drug product” must demonstrate bioequivalence to the Reference Listed Drug (RLD) in compliance with DPCA 1984. Simple solutions or liquid dosage forms may be exempted from this The new drug application relies on already existing safety and/or efficacy data of already approved drug product, but must demonstrate non-inferiority to the RLD, for example, seeking approval of cream dosage form to the RLD (ointment or 3.1.2. Abbreviated New Drug Application (ANDA) The new drug products intended for approval through the § 505(j) of DPCA 1984 (Table “Same drug product formulation means the formulation of the drug product submitted for
approval and any formulations that have minor differences in composition or method of manufacture from the formulation submitted for approval, but are similar enough to be relevant to the agency's determination of bioequivalence” (21 CFR § 320.1). In the case of any changes to the already approved product (same drug formulation), the NDA or ANDA holder may document BE with respect to unchanged formulation. The FDA’s Guidance for Industry: Nonsterile Semisolid Dosage Forms Scale-Up and Post Approval Changes: Chemistry, Manufacturing, and Controls; In Vitro Release Testing and Bioequivalence (SUPAC-SS, 1997) provides recommendations for pharmaceutical sponsors. The guidance defines “(1) the levels of change; (2) recommended chemistry, manufacturing, and controls (CMC) tests to support each level of change; (3) recommended in vitro release tests and/or in vivo bioequivalence tests to support each level of change; and (4) documentation to support 4. Demonstration of BE for Topical Dermatological Drug Products
Demonstration of PE and BE for Topical Dermatological Drug Products must be based on the intuitive choice from several available techniques. As envisaged by Franz (2011), a systematic approach for selecting an appropriate surrogate test can be adapted to establish BE (Figure 1). The choice primarily depends on the therapeutic target and secondly the type of vehicle in which drug is formulated with emphasis on Q3 equivalence (Lionberger, 2004). Skin being the largest organ of the body, has several biologically and chemically distinct layers. To simplify the structure of skin with respect to partitioning of drugs, the outermost layer (stratum corneum) is lipophilic and protects the underlying dermis, which is hydrophilic. Thus, drug release from various vehicles (creams, lotions, gels, ointments, and foams) and diffusion into or through the skin is complex. Figure 1 Systematic approach to selecting an appropriate surrogate test to establish
Bioequivalence of Topical Dermatological Drug Products (Franz TJ, AAPS Annual Meeting,
October 2011); IVRT: In Vitro Release Testing, TEWL: Trans Epidermal Water Loss, IVPT: In
Vitro Permeation Testing
Lionberger (2004) has classified the pharmaceutical equivalence of semisolid dosage  Q1 equivalence  qualitative similar components as RLD  Q2 equivalence  quantitatively similar components as RLD  Q3 equivalence  Q1 and Q2 with structural similarity as RLD IVRT utilizes widely accepted Franz diffusion cells to estimate rate of drug release from drug products. It involves the application of a drug product on to a membrane (synthetic membrane, excised animal skin, or excised human skin) that separates the donor and receiver chambers. The receiver chamber simulates sink conditions in vivo. The rate of delivery obtained from these studies is assumed to be similar to the in vivo situation. The method has been widely employed in discovery research for screening formulations and understanding mechanism of cutaneous drug transport (Narkar, 2010). However, it is not recognized as a surrogate for in vivo BA/BE of new drug products (SUPAC-SS, 1997). However, Franz et al. (2009) have reported substantial evidence of in vitro – in vivo correlation (maximum rate of absorption, total absorption, and time to maximum rate of absorption) using dermatomed cadaver human trunk skin (0.5 – 0.9 mm; finite dose model) for glucocorticoids and retinoid dosage forms of different vehicles when compared to respective RLD. (Glucocorticoid formulations: Alclometasone dipropionate cream and ointment 0.05%, Halobetasol cream and ointment 0.05%, Mometasone ointment 0.1%; Retinoid formulations: Tretinoin gels 0.01%, 0.025%) It is important to note that in vitro excised human skin model provided discriminatory evidence across different vehicles (Alclometasone dipropionate cream versus Ointment; Betamethasone valerate foam versus lotion) in contrast to non-discriminatory vasoconstriction assay (Franz TJ et al., 2009). In addition, Lehman et al., in their recent evaluation of literature, have concluded that using excised human skin would establish better in vitro – in vivo correlation, provided the study protocols are harmonized. SUPACSS (1997) provides a detailed description of the method along with recommended instructions in the event of any changes to the already approved drug Tape stripping provides information on drug uptake, apparent steady-state levels, and drug elimination from the stratum corneum based on a stratum corneum concentration- time curve (FDA’s Draft Guidance, 1998). This method is also known as the dermatopharmacokinetic (DPK) approach similar to blood, plasma, and urine analysis for drug concentrations as a function of time. Though the draft guidance document was withdrawn in 2002, the FDA has recommended it as surrogate method for certain class of drugs, for example antifungals that target the stratum corneum itself (Narkar, 2010). However, Au et al. (2010) have illustrated the potential of standardized TS methods; demonstrating the BE of two 0.05% clobetasol propionate cream formulations and bio-inequivalence of cream and ointment formulations. Pershing et al. (2003) have shown direct correlation between DPK parameters in healthy patients and clinical safety/efficacy of tretinoin gel products in Microdialysis is a continuous sampling technique in which the molecule of interest is collected from the target tissue; thus providing insight into the time course of drug action or biochemical monitoring of the tissue. The technique can be imagined as an artificial capillary, in which a hollow semipermeable probe is carefully inserted into the site of interest: brain, muscle, eye, and skin. Therefore, it provides valuable information of unbound drug concentrations or biomarkers at the site closer to the pharmacological action compared to the conventional plasma/blood drug concentration versus time. Though it was developed for neurological research, it has gained acceptance in other areas of research. Stenken et al. (2010), in their quest to answer, “how minimally invasive is microdialysis in human skin,” have concluded that “probe insertion in the skin leads to inflammatory responses, both acute and chronic, and an immunological probe rejection response, all of which have the potential to affect experimental microdialysis in different ways.” However, with respect to sampling of drug molecules from the skin, perturbation of blood flow to the local tissue is critical which would recover to normal in approximately two hours. The technique has been successfully adopted and demonstrated for dermatological research as well as for demonstrating the BE of topical dosage forms (Narkar, 2010, Stenken et al., 2010). The technique has shown promise in published and unpublished research in our laboratories for monitoring intradermal and subcutaneous tissue drug concentrations after application of transdermal drug formulations (Chaturvedula et al., 2005; Katikaneni et al., 2011; Paturi et al, 2010; Benfeldt et al. (2007) have estimated, based on the variability component of dermal microdialysis of Lidocaine cream and ointment products, the number of subjects as 27 for demonstrating BE with 90% CI and 80 –125% BE limits using two probes in each test area, or 18 subjects using three probes per formulation application site. The required number of subjects using dermal microdialysis is relatively smaller compared to traditional clinical efficacy trials requiring as many as 300 patients for demonstrating BE Dermal microdialysis technique is appealing, however, faces many limitations; such as technical difficulties, protein binding, variability associated with the recovery, and tissue Various spectroscopic methods, including ATR-FTIR, NIR, and Raman, have been investigated for non-invasive measurement of the drug in the skin. Not all molecules can have quantifiable spectral features, which can be used to distinguish from the stratum corneum (Narkar, 2010). NIR has been widely explored as one of the process analytical technology tools in the pharmaceutical industry. The promising feature of NIR is relatively rapid data acquisition and in vivo applicability. 4.6. Pharmacological Response to Demonstrate BE Pharmacodynamic approaches based on pharmacological response are more appropriate for dosage forms intended for local action at the site of application. Topical glucocorticoids (TG) are formulated in various vehicles to treat atopic dermatitis and psoriasis (Wiedersberg S, 2008). FDA Guidance recommends demonstrating in vivo BE of topical dermatologic corticosteroids based on the pharmacodynamics approach, Stoughton-McKenzie vasoconstrictor assay (Table 1). This test is based on the chromameter readings of skin blanching effect resulting from vasoconstrictive action of Several other tests were reported in the literature, for example, laser Doppler flow meter for measuring the blood flow to assess topical NSAIDs; Trans Epidermal Water Loss (TEWL) to evaluate absorption of retinoids; skin temperature increase by nicotinic acid esters (Narkar, 2010; Wiedersberg et al., 2008). 5. Conclusion
The intricacy of cutaneous drug delivery is very well addressed in the literature. Nevertheless, there is a knowledge gap between industry and regulatory agencies. It will not be possible to have a single step solution for demonstrating the BE of all Topical Dermatological Drug Products. However, FDA’s Guidance documents on surrogate methods could not only reduce US healthcare costs by encouraging competition among companies, but also increase the emphasis on product quality, in particular Q3 equivalence. 6. References
Note: Please refer the following references for detailed information.
Au WL, Skinner M, Kanfer I. Comparison of tape stripping with the human skin blanching assay for the bioequivalence assessment of topical clobetasol propionate formulations. J Pharm Pharm Sci. (2010) 13(1):11-20. Benfeldt E, Hansen SH, Vølund A, Menné T, Shah VP. Bioequivalence of topical formulations in humans: evaluation by dermal microdialysis sampling and the dermatopharmacokinetic method. J Invest Dermatol. (2007) 127(1): 170-8. Chaturvedula A, Joshi DP, Anderson C, Morris R, Sembrowich WL, Banga AK. Dermal, subdermal, and systemic concentrations of granisetron hydrochloride by iontophoretic delivery, Pharm Res. (2005) 22:1313-1319. Chen ML, Shah V, Patnaik R, Adams W, Hussain A, Conner D, Mehta M, Malinowski H, Lazor J, Huang SM, Hare D, Lesko L, Sporn D, Williams R. Bioavailability and bioequivalence: an FDA regulatory overview. Pharm Res. (2001) 18(12): 1645-50. CFR  Code of Federal Regulation, Title 21: Food and Drugs, Chapter I  Food and Drug Administration Department of Health and Human Services, Subchapter D  Drugs for human use, Part 320 Bioavailability and Bioequivalence requirements §320.1  definitions http://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfcfr/cfrsearch.cfm?cfrpart=320 (Accessed October 2012) Food and Drug Administration’s Critical Path Initiative § 4.3.3, (March 2004) http://www.fda.gov/ScienceResearch/SpecialTopics/CriticalPathInitiative/ucm076689.htm (Accessed October 2012) Food and Drug Administration’s Regulatory information, http://www.fda.gov/regulatoryinformation/legislation/default.htm (Accessed October 2012) Franz TJ, Lehman PA, Raney SG. Use of excised human skin to assess the bioequivalence of topical products. Skin Pharmacol Physiol. (2009) 22(5): 276-86. FTC File No. 121-0144, http://www.ftc.gov/opa/2012/09/novartis.shtm (Accessed October 2012) Generic competition and drug prices, http://www.fda.gov/AboutFDA/CentersOffices/OfficeofMedicalProductsandTobacco/CDER/ucm129385.htm (Accessed October 2012) Holmgaard R, Benfeldt E, Nielsen JB, Gatschelhofer C, Sorensen JA, Höfferer C, Bodenlenz M, Pieber TR, Sinner F. Comparison of open-flow microperfusion and microdialysis methodologies when sampling topically applied fentanyl and benzoic acid in human dermis ex vivo. Pharm Res. (2012) 29(7): 1808-20. Kamal K. Midha and Gordon McKay, Bioequivalence; Its History, Practice, and Future. The AAPS Journal, (2009) 11(4): 664-70. Katikaneni S, Kasha P, Badkar A, Banga A. Iontophoresis of a 13 kDa protein monitored by subcutaneous microdialysis in vivo, Bioanalysis (2011) 3(21): 2419-2426. Lehman PA, Raney SG, Franz TJ. Percutaneous absorption in man: in vitro-in vivo correlation. Skin Pharmacol Physiol. (2011) 24(4): 224-30. Lionberger R. Topical Bioequivalence Update at: Advisory Committee for Pharmaceutical Science, (April 2004) http://www.fda.gov/ohrms/dockets/AC/04/slides/4034S2_12_Lionberger.ppt. (Accessed October 2012) Narkar Y. Bioequivalence for topical products—an update. Pharm Research, (2010) 27(12): 2590-601. Paturi J, Kim HD, Chakraborty B, Friden PM, Banga AK. Transdermal and intradermal iontophoretic delivery of dexamethasone sodium phosphate: Quantification of the drug localized in skin, J. Drug Target. (2010) 18(2): 134-140. Pershing LK, Nelson JL, Corlett JL, Shrivastava SP, Hare DB, Shah VP. Assessment of dermatopharmacokinetic approach in the bioequivalence determination of topical tretinoin gel products. J Am Acad Dermatol. (2003) 48(5): 740-51. Shah VP, Flynn GL, Yacobi A, Maibach HI, Bon C, Fleischer NM, Franz TJ, Kaplan SA, Kawamoto J, Lesko LJ, Marty JP, Pershing LK, Schaefer H, Sequeira JA, Shrivastava SP, Wilkin J, Williams RL. Bioequivalence of topical dermatological dosage forms  methods of evaluation of bioequivalence. AAPS/FDA Workshop on 'Bioequivalence of Topical Dermatological Dosage Forms  Methods of Evaluating Bioequivalence', September 4-6, 1996, Bethesda, Md. Skin Pharmacol Appl Skin Physiol. (1998) 11(2): 117-24. Siddoju S, Sachdeva V, Friden PM, Banga AK. Iontophoretic delivery of acyclovir: Intradermal drug monitoring using microdialysis and quantification by skin extraction, PDA Journal of Pharmaceutical Sciences and Technology, (2011) 65 (5): 432-444. Singh GJ, Fleischer N, Lesko L, Williams R. Evaluation of the proposed FDA Pilot dose-response methodology for Topical Corticosteroid Bioequivalence Testing. Pharm Res. (1998) (1): 4-7. Siewert M, Dressman J, Brown CK, Shah VP. FIP; AAPS. FIP/AAPS guidelines to dissolution/in vitro release testing of novel/special dosage forms. AAPS PharmSciTech., (2003) 4(1): E7. Skelly JP. A history of biopharmaceutics in the Food and Drug Administration 1968-1993. AAPS J. (2010) 12(1): 44-50. Stenken JA, Church MK, Gill CA, Clough GF. How minimally invasive is microdialysis sampling? A cautionary note for cytokine collection in human skin and other clinical studies. AAPS J. (2010) 12(1): 73-8. Topical dermatological corticosteriods: in vivo bioequivalence, FDA (1995), Available at: http://www.fda.gov/cder/guidance/old098fn.pdf. (Accessed October 2012) Wiedersberg S, Leopold CS, Guy RH. Bioavailability and bioequivalence of topical glucocorticoids. Eur J Pharm Biopharm. (2008) 68(3): 453-66.

Source: http://www.aaps.org/uploadedFiles/Functional/AAPS_Section_Discussion_Board/Discussion_Board_FD/FDDTechArticleDec2012.pdf

C:\rewesquire\website\pennsylvania whistleblower statute has its protection and rewards

The Federal Statute-False Claims Act Affords Whistleblower Employee Protections and Remuneration In 1986, Congress added anti-retaliation protections to the False Claims Act. These provisions, which did not existpreviously, are contained in 31 U.S.C. Sec. 3730(h): Any employee who is discharged, demoted, suspended, threatened, harassed, or in any other mannerdiscriminated against in the terms

Yasmin_short form_sept 2013

Information for Vermont Prescribers of Prescription Drugs (Short Form) Yasmin (Drospirenone and Ethinyl Estradiol) • This list does not imply that the products on this chart are interchangeable or have the same efficacy or safety. Please refer to each product’s FDA-approved label and indication for further information. • The prices listed below are Average Wholesale Prices ("AW

Copyright © 2010-2014 Drug Shortages pdf