Lovsin - PDE Ocular
Lovsin - PDE Ocular
Lovsin - PDE Ocular
Ester Lovsin Barle, Jean-Claude Bizec, Milica Glogovac, Kamila Gromek &
Gian Christian Winkler
To cite this article: Ester Lovsin Barle, Jean-Claude Bizec, Milica Glogovac, Kamila Gromek &
Gian Christian Winkler (2017): Determination and Application of the Permitted Daily Exposure
(PDE) for Topical Ocular Drugs in Multipurpose Manufacturing Facilities, Pharmaceutical
Development and Technology, DOI: 10.1080/10837450.2017.1312442
a
Novartis Pharma AG, Postfach, CH-4002 Basel, Switzerland
b
Novartis Pharma AG NIBR, Postfach, CH-4002 Basel, Switzerland
*Corresponding author:
Ester Lovsin Barle, Novartis Pharma AG, Postfach, CH-4002 Basel, Switzerland,
[email protected]; Tel: +41613246256
D
Co-Authors:
TE
Jean-Claude Bizec, Novartis Pharma AG NIBR, Postfach, CH-4002 Basel, Switzerland,
[email protected], Tel: +41798743462
EP
Milica Glogovac, Novartis Pharma AG, Postfach, CH-4002 Basel, Switzerland,
[email protected]; Tel: +41616966851
C
D
risk assessments for manufacturing of topical ocular drugs. Similarly, the
methods apply to systemically administered drugs, if their production precedes
TE
manufacturing of a topical ocular drug. We have examined pharmacokinetic (PK)
properties of topical ocular drugs and compared them to the PK parameters of
systemically administered drugs. Furthermore, we examined possible adverse
EP
effects of the carry-over in topical ocular drugs at therapeutic doses.
C
and Veterinary Use ANNEX 15 (PIC/S, 2015) describes the principles of qualification
and validation applicable to the facilities, equipment, utilities and processes used for
manufacturing of medicinal products. Section 9.5 states that the "limits for the carry-
product specific permitted daily exposure (PDE) value“ and should be documented in a
risk assessment. The PDE represents a dose that is likely to be without an appreciable
risk of deleterious effects to the potential patient population, by any route, during an
average lifetime. This limit should be calculated in accordance with the revised GMP
D
guideline and EMA’s "Guideline on setting health based exposure limits for use in risk
TE
identification in the manufacture of different medicinal products in shared facilities"
(EMA 2014). The concept of the PDE is similar to limits developed by other regulatory
EP
or authoritative bodies (as well as individual manufacturers) to define “tolerable” or
effects mainly after oral, intravenous (IV) or inhalation exposure. To ensure patients
safety, the toxicological evaluation should have been performed also for locally
ST
PDE value (PDEocular) specifically for active pharmaceutical ingredients (APIs), applied
via the topical ocular route (as drops or gels/creams). Drugs applied intravitreally and
high molecular weight APIs (>40kD) are not in scope of this manuscript. Additionally,
after the production of drugs for systemic administration. Selection of adequate PDEs
Methodology/Background
PDE calculation
In a first step, the calculation of a PDE involves the hazard identification
(reviewing all relevant preclinical and clinical data), identification of critical health
effects (effects most likely to be relevant for the target population and target route of
exposure) and selection of the point-of-departure (PoD). Next, a reliable and robust no-
D
effect level in humans has to be extrapolated by applying Composite Adjustment
TE
Factors (CAF) to the most relevant PoD. CAF is a result of multiplication of several
[TABLE 1]
Many clinically used topical ocular drugs are small molecules and fairly
lipophilic. The limiting molecular weight for conjunctival penetration is between 20 and
JU
40 kDa. After topical ocular administration, they are absorbed rapidly (within a few
minutes) to the systemic circulation (Boddu et al. 2014, Järvinen et al. 1995, Urtti
2006). This process may take place directly from the conjunctival sac via local blood
capillaries or the nasal cavity, which is extensively vascularized and thin. Only a few
studies investigating systemic absorption in humans can be found in the literature, e.g.
for timolol (almost 100% ocular absorption) or pilocarpine (up to 80%) (Urtti and
Salminen 1993). As the most conservative general approach, it is assumed that around
90% of a drug applied topically to the eye will end up in the circulation (Kim et al.
2014, Zafar et al. 2016). Systemic effects after topical ocular administration must be
identified and characterized in the comprehensive health hazard assessment for each
Generally, the total amount of the applied drug is so small, that the level in the
blood is below detection limit or such data are not available (Kompella et al. 2010).
However, some exceptions have been described for molecules acting on receptors
which are not unique to the eye e.g. beta-blockers (e.g. timolol) or muscarinic
D
antagonists (e.g. atropine, scopolamine) (Princelle et al. 2013, Izazola-Conde et al.
2011). TE
EP
Some topical ocular drugs are metabolized during or after absorption e.g.
pilocarpine, levobunolol and epinephrine (Nakano et al. 2014, Järvinen et al. 1995,
C
mechanisms, which ensure proper functioning of the eye and by other concomitant
factors. For example drainage of the instilled solutions, lacrimation and tear turnover,
ST
is less than 5% (Kim et al. 2014, Petal et al. 2013, Kompella et al. 2010, Cholkar et al.
2014) and depends on drop volume as well (Keister et al. 1991). Some publications
state that only 1% or less of a topically applied drug will be absorbed across the cornea
and thus reach the anterior segment of the eye (Saettone 2002). From the aqueous
humor the drug has easy access to the iris and ciliary body, where the drug may bind to
melanin. Melanin bound drugs may form a reservoir that is released gradually to the
surrounding cells, thereby potentially lowering the activity (Cmax), but possibly
prolonging the drug activity (no changes in AUC) (Agrahari et al. 2016). There is a
Individuals with deep brown irides may have up to four times more ocular melanin than
individuals with light blue eyes. This may impact therapy by the ocular route (Abelson
and McLaughlin 2014, Siegfried et al. 2011). Many classes of ocular drugs, including
alpha- and beta-adrenergics, antibiotics and corticosteroids, have been shown to bind to
general, due to the disposition in the anterior chamber and short biological half-life, the
D
risk of bioaccumulation is low, even for individuals with high melanin level.
TE
The selection of AFs for LOAEL (Lowest Observed Adverse Effect Level) to
EP
NOAEL (No Observed Adverse Effect Level) for extrapolation and duration of
exposure may not be different for the calculation of the PDEocular and the calculation of
C
the PDE for systemically administered drugs. The severity of effect AF must consider
AC
local as well as systemic critical health effects based on the whole dataset.
interspecies adjustment factor has to be applied. For ocular drugs, the most relevant
ST
species will be rabbit or monkey (Eskes et al. 2014). The relevance of the PK
parameters in the eye is compared between topical ocular drugs and systemically
JU
[TABLE 2]
of another topical ocular drug. In this case the PDEocular must be selected for the
manufacturing of a drug intended for systemic administration (e.g. IV, oral, inhalation).
In this case the correct PDE (e.g. PDEintravenous, PDEoral, PDEinhalation) must be selected
which reflects the route of systemic administration of the next drug produced in the
shared equipment.
(e.g. IV, oral, inhalation) followed by manufacturing of a topical ocular drug. In this
D
case the PDEocular must be selected for the MSC calculation.
[FIGURE 1]
TE
EP
First scenario: Manufacturing of topical ocular drug followed by manufacturing
of another topical ocular drug
C
This scenario results in much less uncertainty for PDE calculation than the other
AC
two scenarios, because both drugs have known safety profiles, posology and the same
route of administration. Human data are the most reliable for any PDE calculation.
ST
Therefore, the minimal topical ocular therapeutic dose (MinDD) or the lowest
effects observed (Bercu et al. 2016). Critical adverse effects must be recognized and
potential and its dose level for local or systemic effects of topically applied drugs. With
the application of adequate AFs, the calculated PDEocular will provide sufficient
protection from critical adverse effects in the eye (even when applied to both eyes), as
second topical ocular drug (B). The assumption is that the drug load in the second drug
product is 100%.
MinDD ocular A
PDEocular [µg/eye/day] = (2)
CAF
D
Where:
In general, the most relevant PoD for PDE calculation is the MinDD in the most
sensitive subpopulation, relevant for the intended route of exposure (Bercu et al. 2016).
For some topical ocular drugs, it is a challenge to select the adequate PoD and predict
critical effects after systemic exposure, if the systemic route of administration has not
been examined as well. Should the available dataset not cover all endpoints, such as
fertility or reproductive toxicity, the most sensible approach is comparison with other
drugs of the same class. Pharmacokinetics of some drugs administered to the eye may
resemble the intravenous route rather than the oral or enteral route of administration, but
usually the amount of the applied drug is below detection limit. Therefore, ocular
therapeutic doses will be over conservative, if used as a PoD for systemic PDE
calculation. Alternatively, the lowest ocular therapeutic dose may be used as a NOAEL
and no additional AF is applied. Frequently, systemic data are available e.g. for steroids,
antibiotics and analgesics. In this case the systemic NO(A)ELs are used as a PoD as
D
(e.g. IV, oral, inhalation) followed by manufacturing of a topical ocular drug
TE
Especially, if a topical ocular drug is manufactured by a contract manufacturing
organization (CMO), it is possible that the preceding drug produced has been one
EP
intended only for systemic administration. For such a drug, a standard health hazard
assessment will not include a PDEocular. Only adverse health effects to the eye after
C
eye effects will be apparent after ocular administration due to its low ocular
bioavailability. For topical ocular drugs, it is known that they do not reach the posterior
segment drug targets (retina, vitreous, choroid). These targets may be treated by high
ST
2014, Kompella et al. 2010). Therefore, adverse effects which appear after systemic
compartmental drug therapy (Urtti 2006). The human eye (in adults) is an
approximately globular structure with a diameter of 24 mm, and a mass of about 7.5
grams. The eyes occupy less than 0.05% of the total body weight (Kim et al. 2014),
which represents a factor of approx. 3000 when compared to the weight of the whole
body (assuming: 7.5 g per eye = 15 g weight of both eyes; the body weight of the
patient: 50 kg, therefore the conversion factor would be: 50 kg/15 g = 3333, rounded to
3000).
D
If we apply the same criteria as for clinically used ocular topical drugs, it is
TE
expected that drugs intended for systemic administration will have a bioavailability in
the eye of 5% or less due to the presence of natural barriers (Reshma et al. 2015, Petal
EP
et al. 2013,). Additionally, drug absorption into the systemic circulation decreases the
drug concentration in lacrimal fluid extensively, resulting in a very short biological half-
C
life in the anterior chamber. Low bioavailability and short biological half-life reduce the
AC
Based on arguments regarding the low ocular bioavailability and the weight of
ST
the eyes, the systemic PDEiv may be modified for topical ocular administration
the safe dose for lifetime exposure considering 100% bioavailability. Consequently, if
the drug would be systemically absorbed with the dose of the PDEiv, no adverse
systemic effect would be expected. However, since 1-5% of the drug reaches the eye,
bioavailability may be adjusted to extrapolate the PDEiv to the safe dose for ocular
5 % (topical)
Bioavailability factor (BF) = (5)
100 % (iv)
Eq. 4-6: The use of bioavailability and PDEiv [µg/day] as an alternative formula to
D
Conclusions
TE
The topical ocular route is the most common route of ocular drug administration
EP
to the eyes. Eye-drops are convenient dosage forms, which account for 90% of currently
accessible ophthalmic formulations (Petal et al. 2013). The unique anatomy and
C
physiology of the eye make it a highly protected and sensitive organ. Pharmacokinetic
AC
systemically administered drugs. In this paper, we have discussed the essential factors
side effects (systemic and local). PDEocular should be always ≥ PDEiv, considering the
The purpose of this investigation was to identify and describe a method for
calculating a PDE value for topical ocular drugs (PDEocular) and present appropriate
intended for systemic administration is produced before a topical ocular drug, the factor
of 150 can be used to extrapolate a PDEocular from the PDEiv. This extrapolated PDEocular
is then used as the safe dose in the cleaning validation process for of the ocular drug.
Selection criteria for appropriate PDE values in cleaning validation risk assessments are
presented in table 3:
[TABLE 3]
Acknowledgements
The authors would like to thank Ronald Newton, Maarten Prause and Mark
D
Declaration of Interest
The authors report no declarations of interest.
References
TE
EP
Abelson MB, McLaughlin J. 2014. The Colors of Ocular Health The genetics and
C
aesthetics of eye color, and how its components affect ocular therapy. Review of
AC
https://www.reviewofophthalmology.com/article/the-colors-of-ocular-health
ST
Selection for the Derivation of Acceptable Daily Exposure (ADE) Value for Active
Boddu SHS, Gupta H, Patel S. 2014. Drug Delivery to the Back of the Eye Following
D
Cholkar K, Vadlapudi AD, Trinh HM, Mitra AK. Compositions, formulation,
TE
pharmacology, pharmacokinetics, and toxicity of topical, periocular, and intravitreal
EP
ophthalmic drugs. Methods in Pharmacology and Toxicology (2014) pp. 91-118 in:
http://www.ecetoc.org/index.php?mact=Newsroom,cntnt01,details,0&cntnt01documenti
d=146&cntnt01returnid=76
JU
from: http://echa.europa.eu/guidance-documents/guidance-on-information-
requirements-and-chemical-safety-assessment
European Food Safety Authority (EFSA). 2012. Scientific Opinion: Guidance on
selected default values to be used by the EFSA Scientific Committee, Scientific Panels
and Units in the absence of actual measured data. EFSA Journal 10:1–32 [Internet].
European Medicines Agency (EMA). 1998. ICH Topic Q 3 C (R3), Impurities: Residual
Solvents, CPMP/ICH/283/95.
European Medicines Agency (EMA). 2014. Guideline on setting health based exposure
D
limits for use in risk identification in the manufacture of different medicinal products in
TE
shared facilities EMA/CHMP/ CVMP/ SWP/169430/2012 [Internet]. Available from:
EP
http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2014/11/
WC500177735.pdf
C
AC
Eskes C, Vliet E , Schäffer M, Zuang V. Ocular Toxicity (2014). pp. 169-197 in: In
of Pharmaceuticals for Human Use (ICH). 1998. ICH Harmonized Tripartite Guideline:
Duration of Chronic Toxicity Testing in Animals (Rodent and Non Rodent Toxicity
http://www.ich.org/fileadmin/Public_Web_Site/ICH_Products/Guidelines/Safety/S4/Ste
p4/S4_Guideline.pdf
International Society for Pharmaceutical Engineering (ISPE). 2010. Risk-Based
guides/risk-mapp
Systemic Adverse Effects of Ophthalmic and Non Ophthalmic Medications. Proc. West.
D
Järvinen K, Järvinen T, Urtti A. 1995. Ocular absorption following topical delivery,
Kompella UB, Kadam RS, Lee VHL. 2010. Recent advances in ophthalmic drug
JU
Nakano M, Lockhart CM, Kelly JE, Rettie AE. 2014. Ocular cytochrome P450s and
transporters: roles in disease and endobiotic and xenobiotic disposition, Drug Metab
Rev. 1-14.
Naumann BD, Weideman PA. 1995. Scientific basis for uncertainty factors used to
establish occupational exposure limits for pharmaceutical active ingredients. Hum Ecol
Patel A, Cholkar K, Agrahari V, Mitra AK. 2013. Ocular drug delivery systems: An
Good Manufacturing Practice for Medicinal Products, Annex 15. PS/INF 11/2015. 1
D
April 2015 [Internet]. Available from:
TE
http://www.picscheme.org/bo/commun/upload/document/ps-inf-11-2015-pics-gmp-
EP
revised-annex-15.pdf
C
effects of topical ocular instillation of atropine in two children. Arch Pediatr. 20(4):391-
4.
ST
Reichard JF, Maier MA, Naumann BD, Pfister T, Sandhu R, Sargent EV, Streeter AJ,
Toxicity in Rabbits after Sub Acute Exposure to Ocular Irritant Chemicals. Toxicol.
Res. 31(1):49-59
Saettone MF. 2002. Progress and Problems in Ophthalmic Drug Delivery. Future Drug
References used in ma
D
Siegfried CJ, Shui YB, Holekamp NM, Bai F, Beebe DC. 2011. Racial Differences in
TE
Ocular Oxidative Metabolism. Arch Opthalmol. 129(7):849-854.
EP
Sussman R, Naumann B, Pfister T, Sehner C, Seaman C, Weideman P. 2016. A
C
U.S. Food and Drug Administration (FDA). 2005. Guidance for Industry: Estimating
ST
the Maximum Safe Starting Dose in Initial Clinical Trials for Therapeutics in Adult
Healthy Volunteers. Center for Drug Evaluation and Research (CDER), (July).
JU
http://www.fda.gov/downloads/Drugs/Guidances/UCM078932.pdf
U.S. Environmental Protection Agency (U.S. EPA). 2002. A Review of the Reference
concentration-processes
D
WHO. 2001. Guidance Document for the Use of Data in Development of Chemical-
TE
Specific Adjustment Factors (CSAFs ) for Interspecies Differences and Human
Zafar A, Ahmad J, Akhter S, Addo TR. 2016. Nanotechnology for Transcorneal Drug
Targeting in Glaucoma: Challenges and Progress. 75- 100. From Addo AT (ed.) Ocular
Publishing.
JU
JU
ST
AC
C
EP
TE
D
Table 1. Adjustment factors (AFs) typically used to account for uncertainties in the
derivation of the PDE for systemically administered drugs (Sussman et al. 2016).
D
Intraspecies Default Differences between individuals: EFSA
variability value: TE
physiological, biochemical or social Scientific
EP
AF=10 or aspects Committee
Weiderman
1995.
JU
2016,
Naumann and
Weiderman
1995
D
insufficient for PDE calculation) 2012, EFSA
TE
As a default: AF= 1 (a lifetime daily Scientific
EP
exposure, human therapy) Committee
2012,
C
Sussman et al.
AC
2016.
identified)
identified
/ Dosing interval
D
used as a PoD)
used)
adjustment
JU
factor (CAF)
topical ocular drugs do not reach the posterior segment drug targets (retina, vitreous,
by intravitreal administration.
D
Distribution The volumes of distribution are Generally information is available
difficult to determine due to the for evaluation and assessment
slow equilibration of drug in the
ocular tissues
TE
case-by case
EP
Metabolism There are significant levels of Orally applied drugs can be a
various esterases, peptidases, subject of extensive first-pass
C
To → Ocular DS Systemic DS
↓From
next DS
PDEIV/150
D
TE
EP
C
AC
ST
JU