|
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
5 H! ]1 a# }5 K5 E5 p' d: v( j
FDA Perspectives: Scientific Considerations of Forced Degradation Studies in ANDA* P9 y3 \" u* E1 @- p/ Z9 c: s
Submissions
v* H8 M3 H/ m8 fThe author outlines the scientific aspects of forced degradation studies that should be considered
6 h% F/ V A% s$ r& |' R R' F" uin relation to ANDA submissions.+ m0 g, S6 Z; K& W
May 2, 2012# s. \. a- t, \: h+ V
By:Ragine Maheswaran; K& Z% W% f" g7 S7 V: X/ e. b
Pharmaceutical Technology1 H/ o' M3 G6 S' ]0 b
Volume 36, Issue 5, pp. 73-80+ Z% F* ^$ e3 s+ b: M6 t( _+ k0 k
Forced degradation is synonymous with stress testing and purposeful degradation. Purposeful" k! @9 ^, N# O4 u/ `( Z+ @
degradation can be a useful tool to predict the stability of a drug substance or a drug product with8 C9 R" u2 B# v( ]) L( G, a, ^
effects on purity, potency, and safety. It is imperative to know the impurity profile and behavior of/ P5 Q/ |- U1 X: u) d7 A! m
a drug substance under various stress conditions. Forced degradation also plays an important role+ Z$ O6 l" x; p2 h* ~
in the development of analytical methods, setting specifications, and design of formulations under1 B: i/ m/ P. G/ ]; q
the quality-by-design (QbD) paradigm. The nature of the stress testing depends on the individual
% p1 r5 g6 s5 P \; Ydrug substance and the type of drug product (e.g., solid oral dosage, lyophilized powders, and' @7 \9 p9 j1 |- c+ g6 `
liquid formulations) involved (1).
* ]2 r. A6 j8 F( AThe International Conference on Harmonization (ICH) Q1B guideline provides guidance for
* F! V6 x8 V; n3 [; R9 b$ R1 ?performing photostability stress testing; however, there are no additional stress study8 f; g" m" a# p9 I% @
recommendations in the ICH stability or validation guidelines (2). There is also limited+ b7 s5 ]( e5 u
information on the details about the study of oxidation and hydrolysis. The drug substance, `, B- r4 N5 |
monographs of Analytical Profiles of Drug Substances and Excipients provide some information
& X7 z4 `7 q2 T3 Y* ]. @6 o7 @with respect to different stress conditions of various drug substances (3).
. O) t6 X% n1 I2 bThe forced degradation information provided in the abbreviated new drug application (ANDA)
4 ^0 i- t0 \- gsubmissions is often incomplete and in those cases deficiencies are cited. An overview of common8 x' z* N. _% Y/ [% _
deficiencies cited throughout the chemistry, manufacturing, and controls (CMC) section of the% o" \4 ]/ R0 w; e4 x7 H
ANDAs has been published (4–6). Some examples of commonly cited deficiencies related to
7 a) ^4 o& k3 M0 Iforced degradation studies include the following:
: t& h! d7 z- h' \Your drug substance does not show any degradation under any of the stress conditions. Please/ l) x3 G$ O F; ~8 S1 k: p
repeat stress studies to obtain adequate degradation. If degradation is not achievable, please% r( M2 X1 r1 i
provide your rationale.
' W6 U8 R/ X7 y, O- RPlease note that the conditions employed for stress study are too harsh and that most of your drug7 p @0 F" p& N2 ?; E) l2 [; @
substance has degraded. Please repeat your stress studies using milder conditions or shorter$ {$ U" |8 o: H
exposure time to generate relevant degradation products.
+ y# j$ ]1 z9 a0 T! i" O) YIt is noted that you have analyzed your stressed samples as per the assay method conditions. For
- X+ U. U* U @" m5 ~8 {7 Othe related substances method to be stability indicating, the stressed samples should be analyzed
Y- ^2 G! q$ o! K# k" s( Husing related substances method conditions.
4 z) M8 G! d% JPlease state the attempts you have made to ensure that all the impurities including the degradation
( B% \, b9 `* R: lproducts of the unstressed and the stressed samples are captured by your analytical method.3 M1 N8 C. \. y
Please provide a list summarizing the amount of degradation products (known and unknown) in1 g3 G$ [( v3 \
your stressed samples.
4 f- P8 I# a) F0 h4 {Please verify the peak height requirement of your software for the peak purity determination.; c& a( |* X' d
Please explain the mass imbalance of the stressed samples.
" I O, H3 p4 _! J JPlease identify the degradation products that are formed due to drug-excipient interactions.# I! t6 l2 T. o4 _- p' n
Your photostability study shows that the drug product is very sensitive to light. Please explain how
& o2 `. s, X1 s7 J9 wthis is reflected in the analytical method, manufacturing process, product handling, etc.
6 j8 i8 i% h0 I( f$ YIn an attempt to minimize deficiencies in the ANDA submissions, some general recommendations
$ \( F+ @; h9 Z* m& p+ }3 Dto conduct forced degradation studies, to report relevant information in the submission, and to
8 B6 p1 j$ y+ `utilize the knowledge of forced degradation in developing stability indicating analytical methods,% h% O* K* {; \* s
manufacturing process, product handling, and storage are provided in this article., t' A0 E6 d' a! e1 c! [& H
Stress conditions
6 t2 I$ R# O: ZTypical stress tests include four main degradation mechanisms: heat, hydrolytic, oxidative, and9 r8 Z7 H' W; N/ K4 x
photolytic degradation. Selecting suitable reagents such as the concentration of acid, base, or
. G9 O. B: k& e: W: loxidizing agent and varying the conditions (e.g., temperature) and length of exposure can achieve7 G7 Z; T. X7 Z2 V: f9 C
the preferred level of degradation. Over-stressing a sample may lead to the formation of secondary
% _, h+ K- a2 r% ~# W$ [degradants that would not be seen in formal shelf-life stability studies and under-stressing may not0 ]6 Y# {# T- j8 G
serve the purpose of stress testing. Therefore, it is necessary to control the degradation to a desired O: \1 ?1 [ Z! V: ?' L! k0 a
level. A generic approach for stress testing has been proposed to achieve purposeful degradation: o: z1 n3 d, F* t. Z
that is predictive of long-term and accelerated storage conditions (7). The generally recommended9 |7 G; ^- f9 o* T5 o& g
degradation varies between 5-20% degradation (7–10). This range covers the generally
9 u8 ?- i2 n+ D2 E e4 P9 |permissible 10% degradation for small molecule pharmaceutical drug products, for which the$ _3 I) K, b: w/ @" X" A& G
stability limit is 90%-110% of the label claim. Although there are references in the literature that
$ b& |) N3 i# k% {) ^$ g/ W: Ymention a wider recommended range (e.g., 10-30%), the more extreme stress conditions often8 c; G; v7 |: [7 P
provide data that are confounded with secondary degradation products." Z2 k8 L3 i1 h* a9 D6 [! F
Photostability.
$ |- |4 N0 X9 l0 c' {; D! Y7 WPhotostability testing should be an integral part of stress testing, especially for photo-labile
& W9 W0 \3 f5 j8 q+ d1 ~compounds. Some recommended conditions for photostability testing are described in ICH Q1B
; \2 [7 l6 f6 {+ q5 M4 D6 sPhotostability Testing of New Drug Substances and Products (2). Samples of drug substance, and I- B9 T- F& z7 X' T# D) H1 h
solid/liquid drug product, should be exposed to a minimum of 1.2 million lux hours and 200 watt0 t5 _+ n& L" p3 j" {
hours per square meter light. The same samples should be exposed to both white and UV light. To0 M( C) H' W: H8 `& X6 w; M* N5 C n5 I4 S
minimize the effect of temperature changes during exposure, temperature control may be
! g+ W6 N# }+ Enecessary. The light-exposed samples should be analyzed for any changes in physical properties
1 n. T: @" G, T- J" ksuch as appearance, clarity, color of solution, and for assay and degradants. The decision tree
1 j+ D- ]8 S; g+ Houtlined in the ICH Q1B can be used to determine the photo stability testing conditions for drug
0 ]8 | l- `# oproducts. The product labeling should reflect the appropriate storage conditions. It is also
1 X% K3 c* F7 J# `2 dimportant to note that the labeling for generic drug products should be concordant with that of the9 x' D! @8 ]: |$ R
reference listed drug (RLD) and with United States Pharmacopeia (USP) monograph" x# f& q: y* ^7 E# u
recommendations, as applicable.
- b- S( ~4 F& g* j: IHeat.
1 U0 L' ^3 s( n$ uThermal stress testing (e.g., dry heat and wet heat) should be more strenuous than recommended
/ p; A0 F3 |; H1 m6 Q, wICH Q1A accelerated testing conditions. Samples of solid-state drug substances and drug products4 _ s; d) h* k0 L2 ?
should be exposed to dry and wet heat, whereas liquid drug products can be exposed to dry heat. It+ D' M7 R7 o( K: ~3 _
is recommended that the effect of temperature be studied in 10 °C increments above that for
1 |6 }2 C& |+ [6 v5 f2 Mroutine accelerated testing, and humidity at 75% relative humidity or greater (1). Studies may be6 L2 X/ p& t7 ~8 n, o1 D
conducted at higher temperatures for a shorter period (10). Testing at multiple time points could
$ i1 X7 h' ~9 d1 J% ?provide information on the rate of degradation and primary and secondary degradation products.
$ ~% ~) ^: B& [. x8 q7 ]. eIn the event that the stress conditions produce little or no degradation due to the stability of a drug
$ T. L a& h/ y w/ vmolecule, one should ensure that the stress applied is in excess of the energy applied by
- M8 P- N% @4 x3 ^8 \2 p; n& [accelerated conditions (40 °C for 6 months) before terminating the stress study.; Y5 g, o( m+ P" Y
Acid and base hydrolysis.+ {/ B+ z( ?8 b, Y' L
Acid and base hydrolytic stress testing can be carried out for drug substances and drug products in! t1 L- k$ f, ~1 l/ D; S2 o" p9 A' n
solution at ambient temperature or at elevated temperatures. The selection of the type and+ m6 l6 M7 [3 t2 Y% [. U
concentrations of an acid or a base depends on the stability of the drug substance. A strategy for
- Y# {* }. S8 A5 ggenerating relevant stressed samples for hydrolysis is stated as subjecting the drug substance
/ K" b& N" b# l& f2 ? `$ z( ~# m# d( wsolution to various pHs (e.g., 2, 7, 10–12) at room temperature for two weeks or up to a maximum
* C' Z6 ?) a( A: N7 yof 15% degradation (7). Hydrochloric acid or sulfuric acid (0.1 M to 1 M) for acid hydrolysis and
F- v; t- u, @0 _: u6 @! O) X" Rsodium hydroxide or potassium hydroxide (0.1 M to 1 M) for base hydrolysis are suggested as
1 W0 _, v& S) _ Dsuitable reagents for hydrolysis (10). For lipophilic drugs, inert co-solvents may be used to, Y- P1 c+ h$ R& i
solubilize the drug substance. Attention should be given to the functional groups present in the
8 R; d9 Q% i3 D# f: ydrug molecule when selecting a co-solvent. Prior knowledge of a compound can be useful in
. w f8 e. @2 d5 e+ H- B9 e* mselecting the stress conditions. For instance, if a compound contains ester functionality and is very
( S% Y8 b2 n& Z1 z( M: m5 J7 elabile to base hydrolysis, low concentrations of a base can be used. Analysis of samples at various! r% y; Q! v( `; X w
intervals can provide information on the progress of degradation and help to distinguish primary6 c" a ]. m" Z' ?: ~: y2 I# t1 j
degradants from secondary degradants.: m- v( e5 n; j# n3 O& Y% z
Oxidation.
0 S7 c+ u* g s4 d: EOxidative degradation can be complex. Although hydrogen peroxide is used predominantly6 l. {+ ^. J% h. u8 ^
because it mimics possible presence of peroxides in excipients, other oxidizing agents such as6 m9 o! |- A8 H0 O& W
metal ions, oxygen, and radical initiators (e.g., azobisisobutyronitrile, AIBN) can also be used. R2 A! b( \! p
Selection of an oxidizing agent, its concentration, and conditions depends on the drug substance.. M, F, C0 e" U- M3 c% l8 \2 R
Solutions of drug substances and solid/liquid drug products can be subjected to oxidative; o5 m2 C& B9 ?6 T% v
degradation. It is reported that subjecting the solutions to 0.1%-3% hydrogen peroxide at neutral
3 \+ O0 B6 P- K0 y* }: z, t" wpH and room temperature for seven days or up to a maximum 20% degradation could potentially
" d) w6 _3 r# V; t) ~5 }generate relevant degradation products (10). Samples can be analyzed at different time intervals to
; E* r* f* S( R- Q2 g* r+ @- G& Ddetermine the desired level of degradation.
6 @( j+ u# G; X8 C6 h0 i% g3 ]6 HDifferent stress conditions may generate the same or different degradants. The type and extent of
# |* c6 ^ C: `9 cdegradation depend on the functional groups of the drug molecule and the stress conditions.
) M. E; b6 i, d) X; N3 y9 n; GAnalysis method/ X5 T; O) @& Q% F% [% P" W7 b
The preferred method of analysis for a stability indicating assay is reverse-phase
# C! m1 D: E$ a1 Yhigh-performance liquid chromatography (HPLC). Reverse-phase HPLC is preferred for several
1 f& O3 q1 j. n, w2 i1 l$ Vreasons, such as its compatibility with aqueous and organic solutions, high precision, sensitivity,4 ]* a4 ]1 T" t
and ability to detect polar compounds. Separation of peaks can be carried out by selecting; w# h, D2 X2 N
appropriate column type, column temperature, and making adjustment to mobile phase pH.
) I, U6 r. `- E- d6 l! q1 o% \Poorly-retained, highly polar impurities should be resolved from the solvent front. As part of) p$ I, Q3 m, L' U* V% b1 a
method development, a gradient elution method with varying mobile phase composition (very low
. V7 r2 C4 d$ ]* ]; torganic composition to high organic composition) may be carried out to capture early eluting1 Z8 u# f, r. h$ i1 P# w
highly polar compounds and highly retained nonpolar compounds. Stressed samples can also be+ y8 r+ F) |) Y4 X% u8 k' _
screened with the gradient method to assess potential elution pattern. Sample solvent and mobile
0 H5 q0 f0 \4 V3 W' dphase should be selected to afford compatibility with the drug substance, potential impurities, and* s6 [4 ` g% C# |! O$ g/ m
degradants. Stress sample preparation should mimic the sample preparation outlined in the
8 f8 F! x2 I6 Q1 z. u4 v( g# L' Danalytical procedure as closely as possible. Neutralization or dilution of samples may be necessary
6 ?1 P8 L6 z N% I' q5 B8 a! kfor acid and base hydrolyzed samples. Chromatographic profiles of stressed samples should be4 n2 C% N4 |1 t. u" @3 A. w
compared to those of relevant blanks (containing no active) and unstressed samples to determine
* R z2 j9 l2 ?the origin of peaks. The blank peaks should be excluded from calculations. The amount of6 G& Q7 u) [3 R: }6 P2 ~# q
impurities (known and unknown) obtained under each stress condition should be provided along2 n: b* @' V; Q& L6 v9 j
with the chromatograms (full scale and expanded scale showing all the peaks) of blanks,7 X! ~( X" p8 U3 f6 O
unstressed, and stressed samples. Additionally, chiral drugs should be analyzed with chiral4 O) y+ h. G$ a9 W
methods to establish stereochemical purity and stability (11, 12).
/ y8 o4 J# E- Q& X8 V jThe analytical method of choice should be sensitive enough to detect impurities at low levels (i.e.,
) P5 q" H" e: u0 F- e) z% N6 Q4 l0.05% of the analyte of interest or lower), and the peak responses should fall within the range of6 R1 `$ l# h# O- N. p
detector's linearity. The analytical method should be capable of capturing all the impurities formed8 b/ Z1 H' m- R5 s
during a formal stability study at or below ICH threshold limits (13, 14). Degradation product" T% Q1 H5 R$ }- x" L& G/ G
identification and characterization are to be performed based on formal stability results in
/ A% B: z. ?# N; E5 l8 Faccordance with ICH requirements. Conventional methods (e.g., column chromatography) or
2 q' } s% K- Qhyphenated techniques (e.g., LC–MS, LC–NMR) can be used in the identification and( F! f# o- F: [" d4 }
characterization of the degradation products. Use of these techniques can provide better insight9 D" K/ f. V+ {8 f1 [& A9 k
into the structure of the impurities that could add to the knowledge space of potential structural- p) c, c) x* w4 U) `1 }
alerts for genotoxicity and the control of such impurities with tighter limits (12–17). It should be0 B) `/ A# c$ A
noted that structural characterization of degradation products is necessary for those impurities that
) K0 N1 u2 z H7 J) U: s- I( ware formed during formal shelf-life stability studies and are above the qualification threshold limit# O/ \, e/ ]4 g( W
(13).; B$ t9 ?3 w- g9 Z- c7 R
Various detection types can be used to analyze stressed samples such as UV and mass
9 }/ r; V, r% e6 V6 b! t" R% mspectroscopy. The detector should contain 3D data capabilities such as diode array detectors or
B; E4 m' n! t' t5 { Fmass spectrometers to be able to detect spectral non-homogeneity. Diode array detection also1 @" Z$ d7 p A% Y) `& @& G
offers the possibility of checking peak profile for multiple wavelengths. The limitation of diode
$ ^) U- r% Z* `) A5 i9 Q5 Earray arises when the UV profiles are similar for analyte peak and impurity or degradant peak and: t) y. R9 X$ j0 m& d/ c
the noise level of the system is high to mask the co-eluting impurities or degradants. Compounds
# H7 @$ q: O; S8 \of similar molecular weights and functional groups such as diastereoisomers may exhibit similar- o! c9 U3 \& O5 J5 `3 u; o% r
UV profiles. In such cases, attempts must be made to modify the chromatographic parameters to; i; k4 a& K( q. g- ~% w( }. M
achieve necessary separation. An optimal wavelength should be selected to detect and quantitate
% T. d- w( f# wall the potential impurities and degradants. Use of more than one wavelength may be necessary, if
$ `! z8 v2 j% Ithere is no overlap in the UV profile of an analyte and impurity or degradant peaks. A valuable
% G6 W' M l. A2 b9 ]1 x& Gtool in method development is the overlay of separation signals at different wavelengths to
" W% U( b& N; K+ ~discover dissimilarities in peak profiles.- A- ~7 |( A" F# u5 i h( W u
Peak purity analysis.
1 b' E7 U1 a0 ?2 fPeak purity is used as an aid in stability indicating method development. The spectral uniqueness
! c5 a8 J. F4 V" ]! z a+ p! }- Yof a compound is used to establish peak purity when co-eluting compounds are present.$ V+ L1 o5 x& P2 M. u$ }) G
Peak purity or peak homogeneity of the peaks of interest of unstressed and stressed samples
' y+ D( I* B+ ^$ Ishould be established using spectral information from a diode array detector. When instrument
5 @; y- V, h2 N7 q9 O2 jsoftware is used for the determination of spectral purity of a peak, relevant parameters should be8 L- j, y C: O3 b" o
set up in accordance with the manufacturer's guidance. Attention should be given to the peak
5 X2 |2 v; I o4 P2 nheight requirement for establishing spectral purity. UV detection becomes non linear at higher: Y3 |7 d8 Q7 T) c; h5 B
absorbance values. Thresholds should be set such that co-eluting peaks can be detected. Optimum+ B$ Q! @! C; E
location of reference spectra should also be selected. The ability of the software to automatically, I" `5 i& E& j6 ~, U. o7 ^8 Y2 E/ S2 a
correct spectra for continuously changing solvent background in gradient separations should be7 c! ^/ h z0 }4 D t
ascertained.' V) a8 s9 W3 \/ W$ Y
Establishing peak purity is not an absolute proof that the peak is pure and that there is no$ ^8 H( S1 v: D) s) y
co-elution with the peak of interest. Limitations to peak purity arise when co-eluting peaks are
- k3 p+ W# u" R% _spectrally similar, or below the detection limit, or a peak has no chromophore, or when they are
& H3 J( }. C2 j P- Mnot resolved at all.
2 W& ^( l; C) o2 u5 EMass balance.
) I Y4 `3 v/ y- iMass balance establishes adequacy of a stability indicating method though it is not achievable in0 _8 ?$ y" d) q" @: R
all circumstances. It is performed by adding the assay value and the amounts of impurities and
/ u/ e# ?( {- d6 A, t% R( gdegradants to evaluate the closeness to 100% of the initial value (unstressed assay value) with due
2 S8 y9 q" i& |4 O7 c0 [consideration of the margin of analytical error (1).) D0 C1 O e3 z
Some attempt should be made to establish a mass balance for all stressed samples. Mass
* `8 I( i& G4 X" Z' L# B- dimbalance should be explored and an explanation should be provided. Varying responses of
) @: q( h! ]/ ~analyte and impurity peaks due to differences in UV absorption should also be examined by the- B' {. P; K- w9 m
use of external standards. Potential loss of volatile impurities, formation of non-UV absorbing- G( B4 n) C! T. _; J4 q' g: w( _
compounds, formation of early eluants, and potential retention of compounds in the column
; K: m3 u' b6 s( ?; Oshould be explored. Alternate detection techniques such as RI LC/MS may be employed to+ Z& f& U. k% I
account for non-UV absorbing degradants.
* R- F* q6 w, s$ bTermination of study4 [8 A# x! g* c9 _3 c
Stress testing could be terminated after ensuring adequate exposure to stress conditions. Typical
- \2 T) B0 n& X% k2 [) d0 Factivation energy of drug substance molecules varies from 12–24 kcal/mol (18). A compound may' _0 Q" K3 f8 N2 a1 Z
not necessarily degrade under every single stress condition, and general guideline on exposure* ]1 w. _; U L. O; D! T! l
limit is cited in a review article (10). In circumstances where some stable drugs do not show any9 b( s7 j) @, ~$ f J* [
degradation under any of the stress conditions, specificity of an analytical method can be5 t: W q1 _$ ~0 Q
established by spiking the drug substance or placebo with known impurities and establishing! B8 t! L% y, ^ n! I
adequate separation.
5 {% e1 ]3 S5 ?3 YOther considerations7 c6 x5 s! p5 C2 m$ B: D
Stress testing may not be necessary for drug substances and drug products that have: ~7 M2 m) @: e9 i& o
pharmacopeial methods and are used within the limitations outlined in USP <621>. In the case4 N3 W+ D5 B7 C7 F1 ?5 k3 E F
where a generic drug product uses a different polymorphic form from the RLD, the drug substance) L& K8 p( u1 R' }$ K2 i
should be subjected to stress testing to evaluate the physiochemical changes of the polymorphic) q, i, ?4 Y8 y4 }, s6 f
form because different polymorphic forms may exhibit different stability characteristics.
! V# b8 ?: S9 D( p/ x7 E4 \/ ^/ PForced degradation in QbD paradigm2 N6 B; S* t4 X4 u$ V3 {5 E4 n
A systematic process of manufacturing quality drug products that meet the predefined targets for
; Q3 r. o" q0 {. C3 Jthe critical quality attributes (CQA) necessitates the use of knowledge obtained in forced
* L' i) o" x8 J. K/ B2 ?6 |1 T/ ]degradation studies.
/ V8 H! T0 ^3 N3 i0 A* lA well-designed, forced degradation study is indispensable for analytical method development in a
: x! {# @2 {1 ~, e4 IQbD paradigm. It helps to establish the specificity of a stability indicating method and to predict
& @, X) j6 m9 \/ ipotential degradation products that could form during formal stability studies. Incorporating all
3 ?0 X4 d4 R* s/ qpotential impurities in the analytical method and establishing the peak purity of the peaks of6 @: W+ c- X; [7 ?
interest helps to avoid unnecessary method re-development and revalidation.8 \' [; `/ d) W) ^+ k
Knowledge of chemical behavior of drug substances under various stress conditions can also
8 W4 Z6 j' D8 W+ Q, Rprovide useful information regarding the selection of excipients for formulation development.
0 r! d; ?" U3 M, \) u9 Y8 o; Q% _Excipient compatibility is an integral part of understanding potential formulation interactions% F4 x! r" d4 `9 y2 N% m
during product development and is a key part of product understanding. Degradation products due
4 u! L0 \. G, i% J' d$ z ^to drug-excipient interaction or drug-drug interaction in combination products can be examined by
4 o* k3 i9 q" e O8 `3 mstressing samples of drug substance, drug product, and placebo separately and comparing the
9 i( N7 q$ {$ d7 S% kimpurity profiles. Information obtained regarding drug-related peaks and non-drug-related peaks- ]: u* h4 M& J- `! F
can be used in the selection and development of more stable formulations. For instance, if a drug
Z g8 E7 d+ Z! [substance is labile to oxidation, addition of an antioxidant may be considered for the formulation.$ L# w/ o/ F: G. ^
For drug substances that are labile to acid or undergo stereochemical conversion in acidic medium,# Q3 n( L1 K, o' G1 I
delayed-release formulations may be necessary. Acid/base hydrolysis testing can also provide8 H$ r& d9 O$ f$ z
useful insight in the formulation of drug products that are liquids or suspensions.
* F$ @, c! l8 C" x5 `Knowledge gained in forced degradation studies can facilitate improvements in the manufacturing
+ M6 x5 m6 d: @( @! C: Fprocess. If a photostability study shows a drug substance to be photolabile, caution should be
: \+ h( p* E. }! k- P# ]6 z, \! ^taken during the manufacturing process of the drug product. Useful information regarding process
" K9 { A3 V) N: z/ M6 O# Pdevelopment (e.g., wet versus dry processing, temperature selection) can be obtained from thermal7 q; } D. C* u, W2 _0 f% c: O
stress testing of drug substance and drug product. N/ P0 F* x7 O* a4 y* i( W- r
Additionally, increased scientific understanding of degradation products and mechanisms may
, w H& W1 B8 M7 J. @help to determine the factors that could contribute to stability failures such as ambient temperature,5 }' @* q5 j- Y+ k/ E
humidity, and light. Appropriate selection of packaging materials can be made to protect against
5 A3 \; n U# w/ a0 ksuch factors.% i+ r6 @7 d* y" A/ o4 D* A
Conclusion7 D0 R: C3 {. a5 \9 m) y1 s# ~
An appropriately-designed stress study meshes well with the QbD approaches currently being5 s# g3 W: }+ S( @4 F! `
promoted in the pharmaceutical industry. A well-designed stress study can provide insight in% D) s2 q8 E( s) j4 o) Q0 N7 U& ~
choosing the appropriate formulation for a proposed product prior to intensive formulation
( X; Q; d% A$ x7 n8 G. u I; Sdevelopment studies. A thorough knowledge of degradation, including mechanistic understanding+ s) Z( }+ m/ E( _) r
of potential degradation pathways, is the basis of a QbD approach for analytical method
( t6 [3 c+ c. Idevelopment and is crucial in setting acceptance criteria for shelf-life monitoring. Stress testing6 `7 R2 C B1 |6 l- J, `$ T
can provide useful insight into the selection of physical form, stereo-chemical stability of a drug- y. g6 I! V! [( e0 p$ j* i
substance, packaging, and storage conditions. It is important to perform stress testing for generic- e0 ^# J0 Z, L0 t) ?" h
drugs due to allowable qualitative and quantitative differences in formulation with respect to the5 f6 E' @( ~( K; x- P
RLD, selection of manufacturing process, processing parameters, and packaging materials.( d) v; v3 @0 f$ _
Acknowledgments
2 j( v$ S/ U3 K0 n0 bThe author would like to thank Bob Iser, Naiqi Ya, Dave Skanchy, Bing Wu, and Ashley Jung for9 R! n* M t; e" ^' B
their scientific input and support.6 K L" `$ z0 a v9 |
Ragine Maheswaran, PhD, is a CMC reviewer at the Office of Generic Drugs within the Office of
- t% j, {& u) K+ R% a! w& |Pharmaceutical Science, under the US Food and Drug Administration's Center for Drug
9 i% {" \, Q1 q0 F, }% REvaluation and Research, Ragine.Maheswaran@fda.hhs.gov
|$ c9 e# }6 F1 w# xDisclaimer: The views and opinions in this article are only those of the author and do not( _, u1 W& g- i6 x! q7 B
necessarily reflect the views or policies of the US Food and Drug Administration.
N) t" H# x: C( BReferences2 s% E- j8 {6 J/ g# S' ?
1. ICH, Q1A(R2) Stability Testing of New Drug Substances and Products (Geneva, Feb. 2003).
, ^0 g q; Q2 r8 `. B2. ICH, Q1B Stability Testing: Photostability Testing of New Drug Substances and Products4 o8 b: Q. z* c8 B# W
(Geneva, Nov. 1996).: Q/ `3 L% p4 p- i2 W$ y1 c# v1 o
3. H. Brittain, Analytical Profiles of Drug Substances and Excipients (Academic Press, London,
. U! n5 `2 I" @2 R$ @9 I/ z# b2002).
6 x- v( N& k$ n' l5 D% K8 J4. A. Srinivasan and R. Iser, Pharm. Technol. 34 (1), 50–59 (2010).$ a. ^; v1 Z- v% r4 k' O: m
5. A. Srinivasan, R. Iser, and D. Gill, Pharm. Technol. 34(8), 45–51 (2010).
B. I7 G: G8 c& a: W* R4 y S* Q3 {6. A. Srinivasan, R. Iser, and D. Gill, Pharm. Technol. 35 (2), 58–67 (2011)., y! x, m( o& n5 {6 ~
7. S. Klick, et al., Pharm.Technol. 29 (2) 48–66 (2005).
% g* }4 y/ |8 r+ n. A; y8. K. M. Alsante, L. Martin and S. W. Baertschi, Pharm.Technol. 27 (2) 60-72 (2003).
) D) b# @8 P; F9. D. W. Reynolds, et al., Pharm.Technol. 26 (2), 48–56 (2002).$ U. n6 n% x1 H: d ]" }
10. K. M. Alsante et al., Advanced Drug Delivery Reviews 59, 29–37 (2007)., U$ L3 [6 a _" K( f$ Y
11. FDA, Guidance for Industry on Analytical Procedures and methods Validation Chemistry,- C" R* |7 L1 O; C1 ?* E
Manufacturing, and Controls Documentation (draft) (Rockville, MD, Aug. 2000).1 }' l8 J/ C/ ~- a# W
12. ICH, Q6A: Specifications: Test Procedures and Acceptance Criteria for New Drug Substances
) N$ r; x5 x2 ^and New Drug Products: Chemical Substances (Geneva, Oct. 1999).6 n4 x: z6 f, ^, q
13. ICH, Q3A(R2) Impurities in New Drug Substances (Geneva, Oct. 2006).; A# |5 t8 x. d$ y) f
14. ICH, Q3B(R2) Impurities in New Drug Products (Geneva, June 2006).4 r; y( {6 D6 m2 c8 I! `
15. FDA, Guidance for Industry ANDAs: Impurities in Drug Substances (draft), (Rockville, MD,8 K( d" {8 p% c) b
Aug. 2005).
- Y# ], r8 K" O: B4 U16. FDA, Guidance for Industry ANDAs: Impurities in Drug Products (draft) (Rockville, MD,# f( \6 I; N: M7 v9 Z: F
Nov. 2010).$ y# i, Y5 v, r- l4 s
17. EMA, Guideline on the Limits of Genotoxic Impurities, Committee for Medical Products for
4 C' U2 c% ?2 o- NHuman Use (CHMP) (Doc. Ref EMA/CHMP/QWP/251344/2006) (Jan. 1, 2007)./ c& N: q6 u+ K3 b$ s7 Y" U! f d
18. K. A. Conners et al., Chemical Stability of Pharmaceuticals, Wiley and Sons, New York, New
+ b0 w3 C: t4 t. tYork, 2nd Ed., p. 19 (1986). |
|