|
马上注册,结交更多好友,享用更多功能,让你轻松玩转社区
您需要 登录 才可以下载或查看,没有帐号?立即注册
x
. G2 a. @* Y& o2 P* j" o1 D+ ?
FDA Perspectives: Scientific Considerations of Forced Degradation Studies in ANDA* A: {6 v/ V# C3 g4 I- Y' Z# ]
Submissions
6 e: f/ E+ o. Y! F" oThe author outlines the scientific aspects of forced degradation studies that should be considered
3 Y% o# j) i) P- G" vin relation to ANDA submissions.+ E: N6 o% S. \; V8 Q# I0 B5 `
May 2, 2012
+ o6 Q4 B* E/ P; z" c4 V* u: dBy:Ragine Maheswaran8 d8 t9 L% w% L
Pharmaceutical Technology4 \; `/ q9 W. k- A/ @: ]7 p6 l9 a; P k
Volume 36, Issue 5, pp. 73-80) P2 `9 m- Q: Y+ T( @
Forced degradation is synonymous with stress testing and purposeful degradation. Purposeful: R' n" g* W3 y0 A1 B6 X4 p
degradation can be a useful tool to predict the stability of a drug substance or a drug product with% x; ?0 {( X" v9 p4 Z1 E8 Z5 U
effects on purity, potency, and safety. It is imperative to know the impurity profile and behavior of; \- K, U; D* |* N: I+ p4 F0 {, r
a drug substance under various stress conditions. Forced degradation also plays an important role
' p- T; |$ J- Y' f( I# Z7 Sin the development of analytical methods, setting specifications, and design of formulations under
0 v! J# o, E( }- f. f( B+ e( ethe quality-by-design (QbD) paradigm. The nature of the stress testing depends on the individual# ?) D4 ^ f3 j4 W) s" \
drug substance and the type of drug product (e.g., solid oral dosage, lyophilized powders, and
1 T; Z+ c& q1 }, jliquid formulations) involved (1).
" i8 N2 F9 u! Z! oThe International Conference on Harmonization (ICH) Q1B guideline provides guidance for# Z g& q0 }0 j( `3 m1 W6 g
performing photostability stress testing; however, there are no additional stress study" t9 P& @' f& m; m$ _& J
recommendations in the ICH stability or validation guidelines (2). There is also limited
* x5 G$ d# e& q' U! U9 {information on the details about the study of oxidation and hydrolysis. The drug substance" a) u* H5 e8 ^* P0 M# S
monographs of Analytical Profiles of Drug Substances and Excipients provide some information5 [$ j9 L4 u" k J+ l
with respect to different stress conditions of various drug substances (3).
% Z I! q( j* o# ^5 W: M, L9 mThe forced degradation information provided in the abbreviated new drug application (ANDA)
9 f3 M$ j7 U8 P4 f asubmissions is often incomplete and in those cases deficiencies are cited. An overview of common5 X6 e$ Q" g+ P( K
deficiencies cited throughout the chemistry, manufacturing, and controls (CMC) section of the' d, Z% G- w3 k2 C& M
ANDAs has been published (4–6). Some examples of commonly cited deficiencies related to, q4 [# K) N" v
forced degradation studies include the following:
- k. @$ I( j: a; {) gYour drug substance does not show any degradation under any of the stress conditions. Please
. r1 M9 e" I, `4 trepeat stress studies to obtain adequate degradation. If degradation is not achievable, please
3 D5 k. z3 {$ Y" Dprovide your rationale.
& U3 \. C! I1 TPlease note that the conditions employed for stress study are too harsh and that most of your drug) [% \/ l1 _0 f7 T
substance has degraded. Please repeat your stress studies using milder conditions or shorter# Y; l9 d( a* y) b! T& \8 O
exposure time to generate relevant degradation products.) Z0 Y% |! u; ~, p! [
It is noted that you have analyzed your stressed samples as per the assay method conditions. For
. i g. f8 m3 sthe related substances method to be stability indicating, the stressed samples should be analyzed( W+ P. y+ T n1 K1 {. _
using related substances method conditions.5 ]& E9 |$ M- \) Z7 [
Please state the attempts you have made to ensure that all the impurities including the degradation
0 _5 M9 n/ ^5 I, k: gproducts of the unstressed and the stressed samples are captured by your analytical method.
" c1 D9 F i2 |9 [, K& uPlease provide a list summarizing the amount of degradation products (known and unknown) in* H3 ], M7 F5 @
your stressed samples.$ S7 M$ o6 Y6 V' {; B
Please verify the peak height requirement of your software for the peak purity determination. I) T) z) J! n) _+ ], `, V8 X% h1 j
Please explain the mass imbalance of the stressed samples., ?3 s" f- Z3 E
Please identify the degradation products that are formed due to drug-excipient interactions.
$ W; {# q, n0 R1 q* Q! f1 jYour photostability study shows that the drug product is very sensitive to light. Please explain how4 v4 B- W" q# I9 ^) b5 O1 |
this is reflected in the analytical method, manufacturing process, product handling, etc.: Z' J: L8 P7 L3 M4 l. Z
In an attempt to minimize deficiencies in the ANDA submissions, some general recommendations0 P6 E; C [' J; p; A: ?- j7 j: i
to conduct forced degradation studies, to report relevant information in the submission, and to/ s* p- u. ~) i p$ a
utilize the knowledge of forced degradation in developing stability indicating analytical methods,, f' L% Q6 ]* o5 X5 S
manufacturing process, product handling, and storage are provided in this article." s/ o# L; [7 q2 c' _
Stress conditions
7 F, [" o9 r6 y- }3 {2 }( ]* b% CTypical stress tests include four main degradation mechanisms: heat, hydrolytic, oxidative, and! n% m( [* b, S8 S- w# H& U4 ]
photolytic degradation. Selecting suitable reagents such as the concentration of acid, base, or
- y, i' Z( ]& J+ k$ [) \4 ] X6 Joxidizing agent and varying the conditions (e.g., temperature) and length of exposure can achieve! o* P& I4 d1 ?, V
the preferred level of degradation. Over-stressing a sample may lead to the formation of secondary
) C! Y# v, H; pdegradants that would not be seen in formal shelf-life stability studies and under-stressing may not
V9 Z, o. J) G9 Dserve the purpose of stress testing. Therefore, it is necessary to control the degradation to a desired- l( y# S7 ?( L/ [
level. A generic approach for stress testing has been proposed to achieve purposeful degradation3 ~! u4 i: W/ K0 n
that is predictive of long-term and accelerated storage conditions (7). The generally recommended2 r! s' Z" z& S3 J M/ ^
degradation varies between 5-20% degradation (7–10). This range covers the generally0 }! `3 ^# G1 m& h: a, h2 X
permissible 10% degradation for small molecule pharmaceutical drug products, for which the/ f: Q9 }+ r2 q$ Q. l
stability limit is 90%-110% of the label claim. Although there are references in the literature that
3 G2 c% }: `5 X# R6 ~( Amention a wider recommended range (e.g., 10-30%), the more extreme stress conditions often$ R" S$ g9 J' x4 X1 m
provide data that are confounded with secondary degradation products.! I7 ^8 [: \# f2 T5 \* G
Photostability.
5 T/ q _! y7 o. ~* X( M& R gPhotostability testing should be an integral part of stress testing, especially for photo-labile
4 U4 Q! d4 m. |* h7 ^, Dcompounds. Some recommended conditions for photostability testing are described in ICH Q1B0 K# Y' m7 K% g2 y+ H
Photostability Testing of New Drug Substances and Products (2). Samples of drug substance, and1 s# Z( H8 x7 g
solid/liquid drug product, should be exposed to a minimum of 1.2 million lux hours and 200 watt
% i: s! T. [# k0 i8 shours per square meter light. The same samples should be exposed to both white and UV light. To
' K, q0 S! W% Y! X* j, Sminimize the effect of temperature changes during exposure, temperature control may be6 ~7 K3 I; U! N1 u! a% t
necessary. The light-exposed samples should be analyzed for any changes in physical properties+ Q8 e; ?$ T4 j# J& ?
such as appearance, clarity, color of solution, and for assay and degradants. The decision tree- K7 X! a3 R9 F8 D
outlined in the ICH Q1B can be used to determine the photo stability testing conditions for drug
- A% t L4 i" G0 B4 [products. The product labeling should reflect the appropriate storage conditions. It is also6 c; c# L' ?7 f
important to note that the labeling for generic drug products should be concordant with that of the
1 H8 o, w# o5 u' Treference listed drug (RLD) and with United States Pharmacopeia (USP) monograph
y- c, T) N# F, Drecommendations, as applicable.
3 H# n( P* V; n5 G, [. H& r% G* ZHeat.
2 u7 j) \9 w. F5 oThermal stress testing (e.g., dry heat and wet heat) should be more strenuous than recommended9 R# `: V0 X* o
ICH Q1A accelerated testing conditions. Samples of solid-state drug substances and drug products
/ j5 i3 \; [0 v/ n& c4 R- r0 nshould be exposed to dry and wet heat, whereas liquid drug products can be exposed to dry heat. It, @1 Z9 f5 ~$ ^5 a! m
is recommended that the effect of temperature be studied in 10 °C increments above that for0 r) i. T" b5 n
routine accelerated testing, and humidity at 75% relative humidity or greater (1). Studies may be# ~& [6 K2 w/ [; _0 {) s% E4 c! R
conducted at higher temperatures for a shorter period (10). Testing at multiple time points could
" W5 j3 J9 T' y1 T* O) x, K" V3 oprovide information on the rate of degradation and primary and secondary degradation products.
( u3 A0 q$ m8 ?In the event that the stress conditions produce little or no degradation due to the stability of a drug# i) E$ }$ q9 E* N: z9 b
molecule, one should ensure that the stress applied is in excess of the energy applied by
7 A1 `7 s% ~+ w8 ~. g( ?: T9 _1 Zaccelerated conditions (40 °C for 6 months) before terminating the stress study.
' |8 O3 o) F. s. LAcid and base hydrolysis./ M9 T7 I2 N* @( T. Q0 p
Acid and base hydrolytic stress testing can be carried out for drug substances and drug products in
9 o0 E2 B2 t. d; n4 S9 |solution at ambient temperature or at elevated temperatures. The selection of the type and/ x8 Q; b# {$ Y7 Q: i
concentrations of an acid or a base depends on the stability of the drug substance. A strategy for
& \9 b( I5 y0 x# ]) V) E2 kgenerating relevant stressed samples for hydrolysis is stated as subjecting the drug substance
) d K, @& |# C7 V- zsolution to various pHs (e.g., 2, 7, 10–12) at room temperature for two weeks or up to a maximum) U2 y- o/ s4 s
of 15% degradation (7). Hydrochloric acid or sulfuric acid (0.1 M to 1 M) for acid hydrolysis and
0 X* X3 ]2 o! z4 f, |# b: `sodium hydroxide or potassium hydroxide (0.1 M to 1 M) for base hydrolysis are suggested as$ Y; b7 _) g; w) j% z
suitable reagents for hydrolysis (10). For lipophilic drugs, inert co-solvents may be used to
3 }$ _: Q: ^3 Z+ k: r/ @: y4 Q) xsolubilize the drug substance. Attention should be given to the functional groups present in the
# z: D& n7 | u' R/ Edrug molecule when selecting a co-solvent. Prior knowledge of a compound can be useful in$ ]4 {! ~5 o2 U) H3 u M. ^
selecting the stress conditions. For instance, if a compound contains ester functionality and is very! C: X1 F1 a. A# Z
labile to base hydrolysis, low concentrations of a base can be used. Analysis of samples at various
# j& |6 U2 H, K0 D# ?# }, ]intervals can provide information on the progress of degradation and help to distinguish primary
q! f F7 @- `degradants from secondary degradants.) L# l( e2 w* {2 t8 l1 E
Oxidation., A, ~: l) A7 b* y
Oxidative degradation can be complex. Although hydrogen peroxide is used predominantly
4 q K9 V5 V/ z' H& i5 f" H4 }% obecause it mimics possible presence of peroxides in excipients, other oxidizing agents such as
; {" W6 M6 z5 b4 C! {8 G) ~metal ions, oxygen, and radical initiators (e.g., azobisisobutyronitrile, AIBN) can also be used.
8 t8 A' y1 j% B( o/ zSelection of an oxidizing agent, its concentration, and conditions depends on the drug substance.
; `; `6 g- u) y4 `# [# S( h& h' ^Solutions of drug substances and solid/liquid drug products can be subjected to oxidative
+ B/ d0 ]5 V, i# L2 Ndegradation. It is reported that subjecting the solutions to 0.1%-3% hydrogen peroxide at neutral
{- M7 N- g* t/ d- DpH and room temperature for seven days or up to a maximum 20% degradation could potentially
3 e- C( _) v$ j3 Dgenerate relevant degradation products (10). Samples can be analyzed at different time intervals to# I+ r$ V6 _3 ]0 ]/ M
determine the desired level of degradation.% o$ A W( q: n( t0 ^. v
Different stress conditions may generate the same or different degradants. The type and extent of
( N6 Z9 h. g- M; Fdegradation depend on the functional groups of the drug molecule and the stress conditions." o) F# H( X# s' T
Analysis method& g# E; F- C7 K2 k* j+ W3 P
The preferred method of analysis for a stability indicating assay is reverse-phase5 B& k8 I/ w6 B) m) c
high-performance liquid chromatography (HPLC). Reverse-phase HPLC is preferred for several
3 c5 O8 {, B7 G2 @# Lreasons, such as its compatibility with aqueous and organic solutions, high precision, sensitivity,
' {( H0 h* }; A; e' {8 Rand ability to detect polar compounds. Separation of peaks can be carried out by selecting4 h; g1 p" c: ^! v4 R h/ F# T
appropriate column type, column temperature, and making adjustment to mobile phase pH." ]) x2 o% F8 W% z2 P% ^, d% x
Poorly-retained, highly polar impurities should be resolved from the solvent front. As part of. k4 ?. `+ l5 B$ q$ Q4 s
method development, a gradient elution method with varying mobile phase composition (very low
' W, l* O9 r& ^/ x Corganic composition to high organic composition) may be carried out to capture early eluting
, E$ |# n2 U# u- A' whighly polar compounds and highly retained nonpolar compounds. Stressed samples can also be
# ?9 Y; r+ I; K' W: _/ w0 g- |screened with the gradient method to assess potential elution pattern. Sample solvent and mobile
& Z) V5 ^, A% L/ C1 Fphase should be selected to afford compatibility with the drug substance, potential impurities, and, |, _" Y3 s' h9 ~
degradants. Stress sample preparation should mimic the sample preparation outlined in the
1 @; q0 D( ?% m0 o. z7 i Zanalytical procedure as closely as possible. Neutralization or dilution of samples may be necessary
3 N% u1 Y' G. u+ M# g5 g. Gfor acid and base hydrolyzed samples. Chromatographic profiles of stressed samples should be! w* z/ W' L0 c. G* l4 K+ H: [
compared to those of relevant blanks (containing no active) and unstressed samples to determine
9 z/ @$ ]4 O& j" W( gthe origin of peaks. The blank peaks should be excluded from calculations. The amount of; c5 D! U+ `7 G% n
impurities (known and unknown) obtained under each stress condition should be provided along
1 F6 u1 \# w: s& z4 H4 O% K/ p8 Dwith the chromatograms (full scale and expanded scale showing all the peaks) of blanks,
% d* d9 g9 T, _0 j) ?* Tunstressed, and stressed samples. Additionally, chiral drugs should be analyzed with chiral
7 Z6 J" o" b7 n4 v* n1 fmethods to establish stereochemical purity and stability (11, 12).
3 h& x8 T7 C6 r1 q0 [6 c9 P) ^The analytical method of choice should be sensitive enough to detect impurities at low levels (i.e.,
i( P/ Y/ N/ U8 Q! p+ _+ O0.05% of the analyte of interest or lower), and the peak responses should fall within the range of$ Y9 c9 v, A$ e! O
detector's linearity. The analytical method should be capable of capturing all the impurities formed- U, g6 K' q4 a! a& l
during a formal stability study at or below ICH threshold limits (13, 14). Degradation product1 G+ S: r; A1 f2 O0 R8 ?
identification and characterization are to be performed based on formal stability results in0 O, @5 k& c2 I; f0 C6 L
accordance with ICH requirements. Conventional methods (e.g., column chromatography) or
; x! C5 B z# w& C- lhyphenated techniques (e.g., LC–MS, LC–NMR) can be used in the identification and
4 }6 y- I! q1 N% C) H- w. @: icharacterization of the degradation products. Use of these techniques can provide better insight; x* U& p; f3 q, t# P7 `3 H, r
into the structure of the impurities that could add to the knowledge space of potential structural
! r# p; I @2 D0 E, V4 ealerts for genotoxicity and the control of such impurities with tighter limits (12–17). It should be5 I2 I! g5 V$ d
noted that structural characterization of degradation products is necessary for those impurities that
5 @. {2 y5 e( i# o& z* Pare formed during formal shelf-life stability studies and are above the qualification threshold limit
5 @. [/ W9 X" Y/ m w* K(13).
- I; z6 T( ^. K, f0 {1 aVarious detection types can be used to analyze stressed samples such as UV and mass
2 D/ ~3 l: ], T& l, }spectroscopy. The detector should contain 3D data capabilities such as diode array detectors or( R7 {3 G( P! a) d8 Q! x, I7 |0 n
mass spectrometers to be able to detect spectral non-homogeneity. Diode array detection also5 V$ l- o, m: _# h" g
offers the possibility of checking peak profile for multiple wavelengths. The limitation of diode4 f2 }. m* F! y* n2 {+ g
array arises when the UV profiles are similar for analyte peak and impurity or degradant peak and" D( k) ]4 p$ ]+ g: \9 B5 j/ V% {
the noise level of the system is high to mask the co-eluting impurities or degradants. Compounds; R) P/ H9 k* N2 w" G- W9 S
of similar molecular weights and functional groups such as diastereoisomers may exhibit similar2 @3 g; a& R& m0 j% h
UV profiles. In such cases, attempts must be made to modify the chromatographic parameters to
1 i1 b2 z( q! W+ sachieve necessary separation. An optimal wavelength should be selected to detect and quantitate
& E1 Y9 H+ b6 Q: r9 eall the potential impurities and degradants. Use of more than one wavelength may be necessary, if6 K0 n- _# k% d
there is no overlap in the UV profile of an analyte and impurity or degradant peaks. A valuable( [/ s! K5 t8 m! j& R. Y9 A
tool in method development is the overlay of separation signals at different wavelengths to/ s" r3 C: y2 b- B2 ?/ S1 R6 Z
discover dissimilarities in peak profiles.
& F5 r+ Y& Y7 D3 U+ ?- wPeak purity analysis.
# @; H# y& \& X! KPeak purity is used as an aid in stability indicating method development. The spectral uniqueness* G1 g: I. w: ^
of a compound is used to establish peak purity when co-eluting compounds are present.! f0 g" l8 S8 d1 z7 k B5 n0 ~
Peak purity or peak homogeneity of the peaks of interest of unstressed and stressed samples( y( a2 w3 `# H4 ]+ Z L% C
should be established using spectral information from a diode array detector. When instrument
5 j5 F! a- P6 J( K( r& Isoftware is used for the determination of spectral purity of a peak, relevant parameters should be
* T: E/ y* m1 s4 {set up in accordance with the manufacturer's guidance. Attention should be given to the peak9 @- t/ l. \# i6 C9 t! @
height requirement for establishing spectral purity. UV detection becomes non linear at higher+ {: p( W1 ~6 ]% D1 |; B5 q
absorbance values. Thresholds should be set such that co-eluting peaks can be detected. Optimum( \: q* x9 ~, v9 e7 V! \( D
location of reference spectra should also be selected. The ability of the software to automatically: |/ h2 I' H. |1 J3 D2 P. G( x
correct spectra for continuously changing solvent background in gradient separations should be
, n, j+ j8 |7 q5 pascertained.
# ?0 h$ R1 L8 R, `0 C" iEstablishing peak purity is not an absolute proof that the peak is pure and that there is no7 Z+ q. Q& U2 A3 ?" a
co-elution with the peak of interest. Limitations to peak purity arise when co-eluting peaks are
. R5 i& K8 p1 N( Aspectrally similar, or below the detection limit, or a peak has no chromophore, or when they are: p1 E, Y0 E! `+ A
not resolved at all.; G* W6 J8 Z* s& w( ]7 s" a2 g
Mass balance.% U9 `9 m, Q6 K. S& ]' c I5 k& s
Mass balance establishes adequacy of a stability indicating method though it is not achievable in& }( i9 _! Y. `- ^ `; b2 F; S2 k( R
all circumstances. It is performed by adding the assay value and the amounts of impurities and7 _1 Z. M8 t3 F- |
degradants to evaluate the closeness to 100% of the initial value (unstressed assay value) with due/ d) }5 R! V) g3 B; C& M7 V: ~
consideration of the margin of analytical error (1).
. O+ n, ^3 h+ E) xSome attempt should be made to establish a mass balance for all stressed samples. Mass
/ S, S& }% k8 \/ simbalance should be explored and an explanation should be provided. Varying responses of
, w- V7 \5 |7 t- x9 l. Zanalyte and impurity peaks due to differences in UV absorption should also be examined by the
! C. p9 e6 Y* l( P K5 Z# uuse of external standards. Potential loss of volatile impurities, formation of non-UV absorbing0 q2 z9 @: X/ V1 o- e
compounds, formation of early eluants, and potential retention of compounds in the column- E+ E5 E# t- q C. p& B2 F
should be explored. Alternate detection techniques such as RI LC/MS may be employed to
0 s0 _ Z, z2 ]: G/ T: }account for non-UV absorbing degradants.& B# r8 [! p; Y Z- w1 B: D4 o
Termination of study' q( q. M. h4 @& w( O# u
Stress testing could be terminated after ensuring adequate exposure to stress conditions. Typical
* _& o" a. o ] Y9 d* q; zactivation energy of drug substance molecules varies from 12–24 kcal/mol (18). A compound may ~; T& ?9 @3 s+ _# d7 G6 \
not necessarily degrade under every single stress condition, and general guideline on exposure
" k8 |" |$ N! ]' tlimit is cited in a review article (10). In circumstances where some stable drugs do not show any( G) Y0 M2 d- o6 d+ t" U4 L
degradation under any of the stress conditions, specificity of an analytical method can be0 O+ C; S C/ r' E6 _+ |' `" }! w. ~1 c
established by spiking the drug substance or placebo with known impurities and establishing# x' l6 n6 E( P! Q: ~; h" n8 f L
adequate separation.
2 d; s/ P9 G% f, t) {Other considerations7 |1 {% A& K; o% y+ J7 t- V
Stress testing may not be necessary for drug substances and drug products that have
% `- t W. ]4 Y( v' tpharmacopeial methods and are used within the limitations outlined in USP <621>. In the case
3 V/ O7 l! I4 awhere a generic drug product uses a different polymorphic form from the RLD, the drug substance
/ W0 C" o. K9 |; S; C; s1 Z5 E% ?# xshould be subjected to stress testing to evaluate the physiochemical changes of the polymorphic
3 J% U+ y; d3 w, K1 uform because different polymorphic forms may exhibit different stability characteristics.
+ L5 l, F+ ?: k U7 s& `8 AForced degradation in QbD paradigm
0 Q, j, ]8 u0 p! l FA systematic process of manufacturing quality drug products that meet the predefined targets for
" g( U5 i' U8 Z- b: U6 j4 U/ M+ s0 W+ kthe critical quality attributes (CQA) necessitates the use of knowledge obtained in forced: Q' v% \' S) a
degradation studies.
7 ]9 k' x; l! k6 o2 F) Y1 _1 MA well-designed, forced degradation study is indispensable for analytical method development in a
0 J" \* ?' x$ X6 V5 v# HQbD paradigm. It helps to establish the specificity of a stability indicating method and to predict
/ k( i m1 l! V: @potential degradation products that could form during formal stability studies. Incorporating all
" { _' j3 l8 ?+ p. wpotential impurities in the analytical method and establishing the peak purity of the peaks of! B* M9 H+ T0 u w2 D! v
interest helps to avoid unnecessary method re-development and revalidation.( W. Y' }- H8 U* l3 L
Knowledge of chemical behavior of drug substances under various stress conditions can also
5 \3 o- p" d; K* w8 ]( i {provide useful information regarding the selection of excipients for formulation development.
( M; x5 ]& q! bExcipient compatibility is an integral part of understanding potential formulation interactions
1 [$ P* J$ e( _8 Nduring product development and is a key part of product understanding. Degradation products due
0 E! f' W6 M3 Y* ~# D% qto drug-excipient interaction or drug-drug interaction in combination products can be examined by/ S; e5 b8 d$ m8 `4 m
stressing samples of drug substance, drug product, and placebo separately and comparing the
% v) h* i1 J& m+ b3 [2 U7 u- \impurity profiles. Information obtained regarding drug-related peaks and non-drug-related peaks6 |% @; J" A4 A$ M# w! o" e
can be used in the selection and development of more stable formulations. For instance, if a drug
& e h6 w/ z$ G, {) I% }& xsubstance is labile to oxidation, addition of an antioxidant may be considered for the formulation.# N, h& I# U1 x- Y
For drug substances that are labile to acid or undergo stereochemical conversion in acidic medium,
& j Q" t+ x' Jdelayed-release formulations may be necessary. Acid/base hydrolysis testing can also provide
9 S$ l# \& D3 H3 juseful insight in the formulation of drug products that are liquids or suspensions.+ G8 }% B" O) g. Z/ G2 d; z4 K c* S. @6 t
Knowledge gained in forced degradation studies can facilitate improvements in the manufacturing
) P/ v! O# s( `3 g! l2 sprocess. If a photostability study shows a drug substance to be photolabile, caution should be4 s$ F* E4 g7 o4 s" v, C1 X+ E7 v
taken during the manufacturing process of the drug product. Useful information regarding process
' |2 i% Z! _9 }& }development (e.g., wet versus dry processing, temperature selection) can be obtained from thermal& i5 K6 d) K$ g- u0 u: `; Q
stress testing of drug substance and drug product.% w0 K) M6 J& P+ w/ P8 C8 H
Additionally, increased scientific understanding of degradation products and mechanisms may: Q- B) J2 E# P# L0 y* {
help to determine the factors that could contribute to stability failures such as ambient temperature,! o" g) ~3 \" d0 a$ I
humidity, and light. Appropriate selection of packaging materials can be made to protect against
" @* M3 E8 E" ~% s0 a5 gsuch factors.
E) R+ z8 V2 s: r+ qConclusion
- W2 p. ~1 y: y6 H5 V5 bAn appropriately-designed stress study meshes well with the QbD approaches currently being0 l8 V% j3 Q) H) x, s
promoted in the pharmaceutical industry. A well-designed stress study can provide insight in1 Y7 p1 s! O0 t5 V6 S& p
choosing the appropriate formulation for a proposed product prior to intensive formulation
) i0 E3 D+ R" P$ q- a+ F- \) ?0 S* {development studies. A thorough knowledge of degradation, including mechanistic understanding6 Z6 Z0 F% \5 d8 Q
of potential degradation pathways, is the basis of a QbD approach for analytical method
5 z7 p. x% Z8 X. D* U2 mdevelopment and is crucial in setting acceptance criteria for shelf-life monitoring. Stress testing
# a( x( K3 h0 z2 _- Lcan provide useful insight into the selection of physical form, stereo-chemical stability of a drug, x# W r4 n- K7 p4 ?
substance, packaging, and storage conditions. It is important to perform stress testing for generic
! U+ t) ^) T7 w$ _( Gdrugs due to allowable qualitative and quantitative differences in formulation with respect to the
$ z2 u7 q( ^, ORLD, selection of manufacturing process, processing parameters, and packaging materials.
! q T' i6 w* h5 ~1 W8 GAcknowledgments
0 Y& d0 Z# ~7 N4 Q6 @The author would like to thank Bob Iser, Naiqi Ya, Dave Skanchy, Bing Wu, and Ashley Jung for
% E, a. R8 D5 g* a$ A; i) Etheir scientific input and support.
+ p( R& S6 V9 J) fRagine Maheswaran, PhD, is a CMC reviewer at the Office of Generic Drugs within the Office of3 D" f; }# k9 V( Z F3 P
Pharmaceutical Science, under the US Food and Drug Administration's Center for Drug
1 `2 ^3 G6 q8 e, LEvaluation and Research, Ragine.Maheswaran@fda.hhs.gov0 U$ J7 [# C6 U7 c1 Z8 `
Disclaimer: The views and opinions in this article are only those of the author and do not5 y% H4 l$ P5 p' C
necessarily reflect the views or policies of the US Food and Drug Administration.; d% [$ G- Y5 U5 x: L5 p
References, B2 ^5 {7 V& H/ k' a# O3 |$ t3 Q5 n
1. ICH, Q1A(R2) Stability Testing of New Drug Substances and Products (Geneva, Feb. 2003).
$ R6 M# w6 E/ b' V* g9 y( d2. ICH, Q1B Stability Testing: Photostability Testing of New Drug Substances and Products
x1 s) N# ^# J(Geneva, Nov. 1996).
0 p" P. I1 f, B' i3. H. Brittain, Analytical Profiles of Drug Substances and Excipients (Academic Press, London,2 f3 |7 b4 V2 w* g7 C. N
2002).
, o/ L: P; G0 d4. A. Srinivasan and R. Iser, Pharm. Technol. 34 (1), 50–59 (2010).
( p1 g" ~9 s: T0 M5. A. Srinivasan, R. Iser, and D. Gill, Pharm. Technol. 34(8), 45–51 (2010).
* m1 |! [4 g* H6. A. Srinivasan, R. Iser, and D. Gill, Pharm. Technol. 35 (2), 58–67 (2011).: c% y4 E) N) K: ]9 K. G' I
7. S. Klick, et al., Pharm.Technol. 29 (2) 48–66 (2005).; o5 {+ a0 t, Y% U3 \; H7 O0 N% U
8. K. M. Alsante, L. Martin and S. W. Baertschi, Pharm.Technol. 27 (2) 60-72 (2003).9 q m) `+ r( Q
9. D. W. Reynolds, et al., Pharm.Technol. 26 (2), 48–56 (2002).
/ S) v1 G- O' J- g% N10. K. M. Alsante et al., Advanced Drug Delivery Reviews 59, 29–37 (2007).7 n* X8 D+ x& b) J
11. FDA, Guidance for Industry on Analytical Procedures and methods Validation Chemistry,; G3 ]" z3 {" n8 _: f* Y3 Z% g
Manufacturing, and Controls Documentation (draft) (Rockville, MD, Aug. 2000).
& C/ u$ M0 |* j* s+ b12. ICH, Q6A: Specifications: Test Procedures and Acceptance Criteria for New Drug Substances' Z+ n8 L! _" x
and New Drug Products: Chemical Substances (Geneva, Oct. 1999).
* [& [$ U( S1 k& f13. ICH, Q3A(R2) Impurities in New Drug Substances (Geneva, Oct. 2006).
7 `2 C) P$ {- k8 k14. ICH, Q3B(R2) Impurities in New Drug Products (Geneva, June 2006).$ ~/ U* Z/ V" B9 `
15. FDA, Guidance for Industry ANDAs: Impurities in Drug Substances (draft), (Rockville, MD,1 _3 E6 i7 L: z1 `, C
Aug. 2005).7 P5 a( d8 } \. k J \
16. FDA, Guidance for Industry ANDAs: Impurities in Drug Products (draft) (Rockville, MD,. x- ?" s9 N$ G2 ]! f- i
Nov. 2010).
' E' @# o5 c+ I: Z0 x17. EMA, Guideline on the Limits of Genotoxic Impurities, Committee for Medical Products for
6 v! K. b; g" |$ n9 V3 }- ~Human Use (CHMP) (Doc. Ref EMA/CHMP/QWP/251344/2006) (Jan. 1, 2007).9 s: Y4 X6 H( @( v3 l
18. K. A. Conners et al., Chemical Stability of Pharmaceuticals, Wiley and Sons, New York, New& ]" @$ Z) j7 Z, h
York, 2nd Ed., p. 19 (1986). |
|