标题: FDA 关于破坏实验的一些最新看法和要求 [打印本页] 作者: naren4545 时间: 2015-7-15 03:36 PM 标题: FDA 关于破坏实验的一些最新看法和要求
, w5 c8 B, q; w; [FDA Perspectives: Scientific Considerations of Forced Degradation Studies in ANDA9 g& |& j$ K6 i7 s( M8 X
Submissions $ x3 H& y0 w7 PThe author outlines the scientific aspects of forced degradation studies that should be considered ; b0 b h. @0 }1 _) j0 Pin relation to ANDA submissions.; W$ U+ W. b7 o& j. ^
May 2, 2012" ]0 c' H. @/ N5 a- X+ w! H9 O; a- @
By:Ragine Maheswaran ( i$ x# Q% q" K4 v4 X$ r& ZPharmaceutical Technology % h/ p4 o3 ^& A' BVolume 36, Issue 5, pp. 73-808 a, e) Z) c/ h4 Y
Forced degradation is synonymous with stress testing and purposeful degradation. Purposeful % @- ?$ v8 J! a) D+ pdegradation can be a useful tool to predict the stability of a drug substance or a drug product with 6 O* R) ?$ O+ \- l+ Q' t8 b+ G; [5 ceffects on purity, potency, and safety. It is imperative to know the impurity profile and behavior of: `( g% \& C+ t. m: f, X ~
a drug substance under various stress conditions. Forced degradation also plays an important role 4 @! e7 M. i9 ?1 }9 gin the development of analytical methods, setting specifications, and design of formulations under 6 t5 R# y3 b- t! l9 K+ ^the quality-by-design (QbD) paradigm. The nature of the stress testing depends on the individual 5 x# G; P9 z8 f$ w7 _" `$ Qdrug substance and the type of drug product (e.g., solid oral dosage, lyophilized powders, and e) q$ H1 q H# ^liquid formulations) involved (1).8 y# ~3 W+ J3 A1 `& g+ {
The International Conference on Harmonization (ICH) Q1B guideline provides guidance for ( o( W2 u' ^- q6 ^: c/ Rperforming photostability stress testing; however, there are no additional stress study! ^% D3 N3 ]/ W% w
recommendations in the ICH stability or validation guidelines (2). There is also limited ) a4 z0 P: g6 O1 L1 Linformation on the details about the study of oxidation and hydrolysis. The drug substance6 s; f Y) p8 P7 C7 Q
monographs of Analytical Profiles of Drug Substances and Excipients provide some information/ H, E; x s. Q: A
with respect to different stress conditions of various drug substances (3). & n! r; r8 e9 c. tThe forced degradation information provided in the abbreviated new drug application (ANDA) / F& r+ c3 J" C( Z, v/ xsubmissions is often incomplete and in those cases deficiencies are cited. An overview of common ' u! R% u; `& C+ fdeficiencies cited throughout the chemistry, manufacturing, and controls (CMC) section of the% s6 o- y& o6 l/ Z7 U3 v+ K- V% _
ANDAs has been published (4–6). Some examples of commonly cited deficiencies related to; u2 p: j |8 W9 b
forced degradation studies include the following:/ K; j$ [% `- Z
Your drug substance does not show any degradation under any of the stress conditions. Please & }( V# k. `8 {7 ~2 U: vrepeat stress studies to obtain adequate degradation. If degradation is not achievable, please 2 ]+ s+ O" {3 V# o! a; q' iprovide your rationale.: T1 c3 N/ j. I. d4 ]9 Q
Please note that the conditions employed for stress study are too harsh and that most of your drug , C2 X$ A; F. v7 s: Fsubstance has degraded. Please repeat your stress studies using milder conditions or shorter" D4 C6 d' O4 N* z' y) S ?, h0 ?& |
exposure time to generate relevant degradation products. y9 a: y2 v# B! ~It is noted that you have analyzed your stressed samples as per the assay method conditions. For 2 c, A% K( U3 I% a4 t0 Pthe related substances method to be stability indicating, the stressed samples should be analyzed1 A4 @; g% ]* W: t" W7 E
using related substances method conditions. $ k" B+ u* X9 XPlease state the attempts you have made to ensure that all the impurities including the degradation& M* Y2 _! s3 a9 m
products of the unstressed and the stressed samples are captured by your analytical method.( Q" n" R' ^/ [4 u
Please provide a list summarizing the amount of degradation products (known and unknown) in & ^* R, z* Y4 m5 [3 M; E' U; w: g p& w; a6 _your stressed samples.8 ^2 v" x2 y- l* R. T& o1 S
Please verify the peak height requirement of your software for the peak purity determination. 5 p& N% J0 y2 G& GPlease explain the mass imbalance of the stressed samples.2 R) _! |# A. j, W+ B- i/ H' ]! a
Please identify the degradation products that are formed due to drug-excipient interactions. 0 p( a% d9 `- U rYour photostability study shows that the drug product is very sensitive to light. Please explain how a: s: ]. m3 j/ u
this is reflected in the analytical method, manufacturing process, product handling, etc. b4 Z+ u0 q3 v- i* Y4 WIn an attempt to minimize deficiencies in the ANDA submissions, some general recommendations0 l# x- m# \8 W7 v
to conduct forced degradation studies, to report relevant information in the submission, and to+ u$ `; ^: `! o
utilize the knowledge of forced degradation in developing stability indicating analytical methods, ( J1 e* z! D" j& gmanufacturing process, product handling, and storage are provided in this article.% f" L+ a5 B* G g0 }% t
Stress conditions0 v0 S! @' o0 u
Typical stress tests include four main degradation mechanisms: heat, hydrolytic, oxidative, and % k. l2 Z& ~5 h( G. B9 hphotolytic degradation. Selecting suitable reagents such as the concentration of acid, base, or E# P( C l1 ^) [" z: }oxidizing agent and varying the conditions (e.g., temperature) and length of exposure can achieve( r. b4 `+ l+ T1 [
the preferred level of degradation. Over-stressing a sample may lead to the formation of secondary ! `+ z0 G- I/ @& vdegradants that would not be seen in formal shelf-life stability studies and under-stressing may not D) l9 R- b# P( K Tserve the purpose of stress testing. Therefore, it is necessary to control the degradation to a desired' O5 _3 l" S& B4 i) y! ?+ w
level. A generic approach for stress testing has been proposed to achieve purposeful degradation : M0 F3 _* y9 T3 J/ x2 Cthat is predictive of long-term and accelerated storage conditions (7). The generally recommended * W$ r8 G1 Z' Q% i* V% edegradation varies between 5-20% degradation (7–10). This range covers the generally ( ~; M# s+ j9 E* V1 wpermissible 10% degradation for small molecule pharmaceutical drug products, for which the3 |( }/ K! J1 y0 j: r+ {0 i8 u
stability limit is 90%-110% of the label claim. Although there are references in the literature that @) j* y" j* X3 g& ]6 {, pmention a wider recommended range (e.g., 10-30%), the more extreme stress conditions often0 c' A- y. N0 Z ^( d' y& u
provide data that are confounded with secondary degradation products.* k. M a) B; u/ ?( v7 L9 t
Photostability. 6 e9 t' k# Z, |3 yPhotostability testing should be an integral part of stress testing, especially for photo-labile % W* c2 j% ?' Q0 z6 dcompounds. Some recommended conditions for photostability testing are described in ICH Q1B" R2 [ y, n; g' v
Photostability Testing of New Drug Substances and Products (2). Samples of drug substance, and y1 v2 x' ^( E1 ~! F
solid/liquid drug product, should be exposed to a minimum of 1.2 million lux hours and 200 watt' q! g* J) s. S- D) I( t, @' e
hours per square meter light. The same samples should be exposed to both white and UV light. To4 h. m( N" r: G/ [- M
minimize the effect of temperature changes during exposure, temperature control may be ( P& x- }$ f8 _! z$ Hnecessary. The light-exposed samples should be analyzed for any changes in physical properties - S# m( ^$ N' I* r. L% Q3 \; V. o! @# Rsuch as appearance, clarity, color of solution, and for assay and degradants. The decision tree$ k5 p: `9 o$ C
outlined in the ICH Q1B can be used to determine the photo stability testing conditions for drug) |$ |1 X/ Z; V) M- q$ S3 n
products. The product labeling should reflect the appropriate storage conditions. It is also 4 p/ b0 R3 d( l# L, D8 Aimportant to note that the labeling for generic drug products should be concordant with that of the t) y" G# G* M$ mreference listed drug (RLD) and with United States Pharmacopeia (USP) monograph & D. ^# Z2 f" z, R0 x/ Erecommendations, as applicable.$ o# a+ f2 U4 X) v( ]; v' i9 y
Heat.3 Q2 _) r" M* t. w: W. P
Thermal stress testing (e.g., dry heat and wet heat) should be more strenuous than recommended , \+ N. Z/ t7 O7 ]* ^+ l0 M) m5 RICH Q1A accelerated testing conditions. Samples of solid-state drug substances and drug products 3 {3 E1 B4 {1 n2 k6 ~& ~, Cshould be exposed to dry and wet heat, whereas liquid drug products can be exposed to dry heat. It; B/ h7 R3 v6 D8 F: a7 A4 m: `
is recommended that the effect of temperature be studied in 10 °C increments above that for . I) j3 }. ^. j# Droutine accelerated testing, and humidity at 75% relative humidity or greater (1). Studies may be' d. S* P- D, A4 v4 A+ P
conducted at higher temperatures for a shorter period (10). Testing at multiple time points could & z* o E/ r1 C& i# rprovide information on the rate of degradation and primary and secondary degradation products. 2 t' z( g$ ^7 W. G0 BIn the event that the stress conditions produce little or no degradation due to the stability of a drug 6 {! A5 }, T$ ^! ]$ V: n$ Hmolecule, one should ensure that the stress applied is in excess of the energy applied by7 d5 U$ v1 D$ V, O
accelerated conditions (40 °C for 6 months) before terminating the stress study. % v! H* k d* D5 F5 k G% NAcid and base hydrolysis.( z0 r, @, K! f# Z* l, i7 o- A
Acid and base hydrolytic stress testing can be carried out for drug substances and drug products in - u9 b$ c- p/ r8 ]9 usolution at ambient temperature or at elevated temperatures. The selection of the type and& I1 b! K( c4 e3 S
concentrations of an acid or a base depends on the stability of the drug substance. A strategy for9 ?/ ]3 i# }9 @ A' P
generating relevant stressed samples for hydrolysis is stated as subjecting the drug substance+ c4 w: \% ]- b4 x* ]9 Z0 F
solution to various pHs (e.g., 2, 7, 10–12) at room temperature for two weeks or up to a maximum 2 ~/ u$ b( ?9 P9 p, g/ G1 S, s& ]; w9 n! zof 15% degradation (7). Hydrochloric acid or sulfuric acid (0.1 M to 1 M) for acid hydrolysis and + z( M: h/ Z4 E9 i# ^sodium hydroxide or potassium hydroxide (0.1 M to 1 M) for base hydrolysis are suggested as + f1 V5 A& @, [) p3 Bsuitable reagents for hydrolysis (10). For lipophilic drugs, inert co-solvents may be used to , s7 U# y: p% l n4 D% E" q7 X7 nsolubilize the drug substance. Attention should be given to the functional groups present in the; |. [" q8 A$ G4 N- ^
drug molecule when selecting a co-solvent. Prior knowledge of a compound can be useful in, G% Y4 G5 [+ o/ u4 Y
selecting the stress conditions. For instance, if a compound contains ester functionality and is very . w k$ a% w8 ?" Wlabile to base hydrolysis, low concentrations of a base can be used. Analysis of samples at various1 T6 D4 f5 F: U6 D7 G a% p4 }
intervals can provide information on the progress of degradation and help to distinguish primary8 u! h9 g# x" r7 O- A
degradants from secondary degradants. 9 \% \+ Z' p( a/ L( [Oxidation. ( F1 c: ]+ I- k9 ~$ X1 j/ ^8 h# JOxidative degradation can be complex. Although hydrogen peroxide is used predominantly" z# E+ b/ Y/ I: D0 M5 O. h. M
because it mimics possible presence of peroxides in excipients, other oxidizing agents such as" i" V# P( m' n. s9 o2 c( f2 y
metal ions, oxygen, and radical initiators (e.g., azobisisobutyronitrile, AIBN) can also be used.) N9 F1 g+ N' ]& W
Selection of an oxidizing agent, its concentration, and conditions depends on the drug substance. 1 l! `5 Z0 ~& aSolutions of drug substances and solid/liquid drug products can be subjected to oxidative' h( t: |% D7 w. q8 c7 v+ R$ [
degradation. It is reported that subjecting the solutions to 0.1%-3% hydrogen peroxide at neutral/ Y, I1 i9 x$ q( t( s, v
pH and room temperature for seven days or up to a maximum 20% degradation could potentially$ T$ S/ U6 Q# h1 o# I' Q5 V
generate relevant degradation products (10). Samples can be analyzed at different time intervals to" i' S6 l+ m+ Q0 S6 ^" D/ @. h7 a& F
determine the desired level of degradation.6 }2 O; D/ x- K8 ]* Q! F
Different stress conditions may generate the same or different degradants. The type and extent of ! e. m/ n+ o2 t8 ?8 [degradation depend on the functional groups of the drug molecule and the stress conditions.9 Q' C) A( b7 K) e# T
Analysis method+ m/ ^0 J: A- R; Z* \( c
The preferred method of analysis for a stability indicating assay is reverse-phase 5 S8 Y+ ~$ J1 zhigh-performance liquid chromatography (HPLC). Reverse-phase HPLC is preferred for several 1 M+ @! s2 [* P- m! c+ Z, }( breasons, such as its compatibility with aqueous and organic solutions, high precision, sensitivity, 3 Z7 h8 V: H! ~8 R2 @7 Dand ability to detect polar compounds. Separation of peaks can be carried out by selecting 2 y' t0 T0 x9 M. V2 V4 l( l/ Jappropriate column type, column temperature, and making adjustment to mobile phase pH. 5 A% q5 i5 A! O2 y- FPoorly-retained, highly polar impurities should be resolved from the solvent front. As part of. \3 h K$ ~* w( j5 v( R
method development, a gradient elution method with varying mobile phase composition (very low : o$ |5 s: V8 ]! Zorganic composition to high organic composition) may be carried out to capture early eluting2 x& \7 M% I8 B
highly polar compounds and highly retained nonpolar compounds. Stressed samples can also be ' }+ h) D6 c4 ?% X- m! W3 |screened with the gradient method to assess potential elution pattern. Sample solvent and mobile A) C5 w3 H$ a
phase should be selected to afford compatibility with the drug substance, potential impurities, and ' I0 b; ~+ C- G$ x3 s) A( {degradants. Stress sample preparation should mimic the sample preparation outlined in the 6 R8 x' s" e/ A" f6 d" nanalytical procedure as closely as possible. Neutralization or dilution of samples may be necessary% ]# @) Y& |9 u& r4 a; ]3 b
for acid and base hydrolyzed samples. Chromatographic profiles of stressed samples should be " B) J3 O" _% f( t3 }compared to those of relevant blanks (containing no active) and unstressed samples to determine ; i) s% C# j; N( Q( |the origin of peaks. The blank peaks should be excluded from calculations. The amount of5 V; q, V3 @, B
impurities (known and unknown) obtained under each stress condition should be provided along! D/ ^, o; E9 V
with the chromatograms (full scale and expanded scale showing all the peaks) of blanks, & q3 @8 l! F5 X7 [% o. Zunstressed, and stressed samples. Additionally, chiral drugs should be analyzed with chiral( t: e! x9 V T' F. `1 _
methods to establish stereochemical purity and stability (11, 12). 9 C' @& C3 Y U9 t8 M. nThe analytical method of choice should be sensitive enough to detect impurities at low levels (i.e.,0 l# [ G- J. B! Y" J
0.05% of the analyte of interest or lower), and the peak responses should fall within the range of9 i& @2 X" ^7 j& X. ?+ S' B
detector's linearity. The analytical method should be capable of capturing all the impurities formed . L+ U% @/ W4 ]& D" e8 P; x+ ]during a formal stability study at or below ICH threshold limits (13, 14). Degradation product, r. N! h9 m* |9 |1 h8 r
identification and characterization are to be performed based on formal stability results in4 G9 S2 q$ t( ~
accordance with ICH requirements. Conventional methods (e.g., column chromatography) or ! G3 p' L3 f& |! q( q3 vhyphenated techniques (e.g., LC–MS, LC–NMR) can be used in the identification and: P! _, R H# k- X; r8 g
characterization of the degradation products. Use of these techniques can provide better insight x; ~0 W' p6 |3 O8 p% \into the structure of the impurities that could add to the knowledge space of potential structural * t3 l# g$ ]) Q6 K0 M5 k% V: e' Calerts for genotoxicity and the control of such impurities with tighter limits (12–17). It should be9 z" L$ w( j# M" C: f. n+ W
noted that structural characterization of degradation products is necessary for those impurities that5 y& f' G: P8 j z
are formed during formal shelf-life stability studies and are above the qualification threshold limit 8 d' z3 m# [* ^ l(13). 8 \: }4 Z6 R3 z2 r! j4 F; pVarious detection types can be used to analyze stressed samples such as UV and mass+ D( N `9 s+ ]
spectroscopy. The detector should contain 3D data capabilities such as diode array detectors or 7 y6 R0 p+ O# X& r- v2 Jmass spectrometers to be able to detect spectral non-homogeneity. Diode array detection also$ Q m+ v! Z1 ]
offers the possibility of checking peak profile for multiple wavelengths. The limitation of diode " H. c7 ?! E% d4 z K* E6 ~array arises when the UV profiles are similar for analyte peak and impurity or degradant peak and 0 T# p5 U, k, I+ m0 t% kthe noise level of the system is high to mask the co-eluting impurities or degradants. Compounds * s5 \0 X/ e4 J: lof similar molecular weights and functional groups such as diastereoisomers may exhibit similar: A ]4 O% S( n* a
UV profiles. In such cases, attempts must be made to modify the chromatographic parameters to 8 g- G( }7 T' I- x" |! ^9 W6 C) {achieve necessary separation. An optimal wavelength should be selected to detect and quantitate% Q. {+ A" q, x* ^7 S5 A
all the potential impurities and degradants. Use of more than one wavelength may be necessary, if( y" [5 q: j) P# M
there is no overlap in the UV profile of an analyte and impurity or degradant peaks. A valuable5 P% w$ p, O# w% f
tool in method development is the overlay of separation signals at different wavelengths to 0 H$ I' q* w( ]9 }: }+ J5 s3 ?; a3 hdiscover dissimilarities in peak profiles.% W) p/ z0 {8 @3 M0 c4 {5 L9 H
Peak purity analysis. , g) I' M3 J5 ePeak purity is used as an aid in stability indicating method development. The spectral uniqueness# P; W, Y$ k4 O. ?5 V" z: [! f
of a compound is used to establish peak purity when co-eluting compounds are present. 2 C# L. d- {) ^- W1 j" m, rPeak purity or peak homogeneity of the peaks of interest of unstressed and stressed samples 3 `" t# d: ]2 i8 P# F. |should be established using spectral information from a diode array detector. When instrument' s3 }+ M; j) o$ [* m9 C
software is used for the determination of spectral purity of a peak, relevant parameters should be. ^6 o( j, [, ^( O, a
set up in accordance with the manufacturer's guidance. Attention should be given to the peak. D0 N5 Q: u+ z2 L# z
height requirement for establishing spectral purity. UV detection becomes non linear at higher 8 |. U4 \; g5 u- v S2 zabsorbance values. Thresholds should be set such that co-eluting peaks can be detected. Optimum 6 A( j% b9 q/ H' n! U1 W5 j# C& wlocation of reference spectra should also be selected. The ability of the software to automatically 3 u; G6 t5 a; Z) B5 icorrect spectra for continuously changing solvent background in gradient separations should be" c1 H6 n, u8 T7 c+ J
ascertained.( w2 R- q. I) i6 U) P y* p
Establishing peak purity is not an absolute proof that the peak is pure and that there is no7 X' o/ F; k6 ?
co-elution with the peak of interest. Limitations to peak purity arise when co-eluting peaks are2 x- i8 c$ M" n" f! e
spectrally similar, or below the detection limit, or a peak has no chromophore, or when they are ; C$ \- ]& H& Snot resolved at all. 1 ?3 A& ? a. Z4 @Mass balance. a6 u* i4 ]# t$ N) f" x3 |3 v2 a
Mass balance establishes adequacy of a stability indicating method though it is not achievable in' e6 f- }4 h3 ?4 ]1 x0 h
all circumstances. It is performed by adding the assay value and the amounts of impurities and 5 ~' W/ Z2 r+ Ldegradants to evaluate the closeness to 100% of the initial value (unstressed assay value) with due! B* e& D4 C% E
consideration of the margin of analytical error (1).& p9 z( E) B) H; k2 }. Z
Some attempt should be made to establish a mass balance for all stressed samples. Mass 0 M. h( k: H Z& v, Eimbalance should be explored and an explanation should be provided. Varying responses of 1 l0 Q! h' c6 O" k' n) danalyte and impurity peaks due to differences in UV absorption should also be examined by the $ }* w3 D; I& T# [- G9 Wuse of external standards. Potential loss of volatile impurities, formation of non-UV absorbing% r: w3 b" x% Y
compounds, formation of early eluants, and potential retention of compounds in the column& o# B+ M1 E* ~6 | R
should be explored. Alternate detection techniques such as RI LC/MS may be employed to+ f1 a3 i. P# J* g) g3 H# e- S1 r. J: l
account for non-UV absorbing degradants. 0 q0 [* Z( T: X, i3 K- v# lTermination of study 8 s, y# v# u- @3 J$ SStress testing could be terminated after ensuring adequate exposure to stress conditions. Typical: j( i* G4 s* e: Z8 v6 o: G) H% B! [
activation energy of drug substance molecules varies from 12–24 kcal/mol (18). A compound may 7 H4 W/ y9 L% }% e8 Cnot necessarily degrade under every single stress condition, and general guideline on exposure- l5 A4 w& n% W( |/ f/ R, I" K9 d u
limit is cited in a review article (10). In circumstances where some stable drugs do not show any ( D1 @; L3 |$ L* `( g& gdegradation under any of the stress conditions, specificity of an analytical method can be* v6 L% s& y) G" F- O
established by spiking the drug substance or placebo with known impurities and establishing 6 o: N ^2 i2 g. F; P" Oadequate separation. 5 @' {. B# h5 f3 H8 |Other considerations9 F% F8 M1 A* ^/ F! [* Q# v
Stress testing may not be necessary for drug substances and drug products that have; x) P) q1 I+ s& O
pharmacopeial methods and are used within the limitations outlined in USP <621>. In the case ) j( m& U- q2 W3 ]& g* ^) C; ]where a generic drug product uses a different polymorphic form from the RLD, the drug substance G3 E& d1 H) {' m9 K+ Rshould be subjected to stress testing to evaluate the physiochemical changes of the polymorphic: _# s; s p1 h4 x. g4 ^6 x- G
form because different polymorphic forms may exhibit different stability characteristics. : Z3 \- V5 L3 xForced degradation in QbD paradigm8 ~0 d/ {6 p: l
A systematic process of manufacturing quality drug products that meet the predefined targets for : z% E2 A. j& d9 Z! b- e! ^5 Q3 Athe critical quality attributes (CQA) necessitates the use of knowledge obtained in forced, N6 q6 \% m( O2 p
degradation studies. : q2 L4 c( H2 u" @' R8 WA well-designed, forced degradation study is indispensable for analytical method development in a; B( g& r* ? _% _. ~6 O* U
QbD paradigm. It helps to establish the specificity of a stability indicating method and to predict, o$ J& y9 U. x/ z) e; b3 A
potential degradation products that could form during formal stability studies. Incorporating all 3 @- ~* U" f9 g( p/ ?$ ~potential impurities in the analytical method and establishing the peak purity of the peaks of . z) C, w L! H R# F- uinterest helps to avoid unnecessary method re-development and revalidation. * p' X6 [3 I1 @ a" hKnowledge of chemical behavior of drug substances under various stress conditions can also 1 y4 z1 I9 i9 f- C( ^/ |" Rprovide useful information regarding the selection of excipients for formulation development.- S5 [) Z! J5 P0 D1 y+ [+ B
Excipient compatibility is an integral part of understanding potential formulation interactions7 O+ u, g" n/ i7 W
during product development and is a key part of product understanding. Degradation products due" H( O$ I; k/ I
to drug-excipient interaction or drug-drug interaction in combination products can be examined by 5 s9 P; J! L% G6 E" V0 y, K% O1 jstressing samples of drug substance, drug product, and placebo separately and comparing the" {% t* a$ K' C( D. Z
impurity profiles. Information obtained regarding drug-related peaks and non-drug-related peaks , N0 a- q0 K: H( }can be used in the selection and development of more stable formulations. For instance, if a drug # V1 D( n6 j2 ^3 {/ g5 C h0 tsubstance is labile to oxidation, addition of an antioxidant may be considered for the formulation." O/ E1 W; A$ L! l! k- q- }
For drug substances that are labile to acid or undergo stereochemical conversion in acidic medium, 4 b# @3 T1 l5 G0 P5 gdelayed-release formulations may be necessary. Acid/base hydrolysis testing can also provide4 ?. L5 z0 R3 ^1 t- X
useful insight in the formulation of drug products that are liquids or suspensions.% E! v! {* [0 e7 i! k7 q9 V) n" {
Knowledge gained in forced degradation studies can facilitate improvements in the manufacturing : p. P$ H, ^& N% N5 ]5 C uprocess. If a photostability study shows a drug substance to be photolabile, caution should be ! o+ {% R; g' Otaken during the manufacturing process of the drug product. Useful information regarding process ( C. S" m3 H* x+ S0 d. Gdevelopment (e.g., wet versus dry processing, temperature selection) can be obtained from thermal+ J6 f/ h# a+ D: G2 d7 ?4 {
stress testing of drug substance and drug product. . H3 k5 d* O: ?Additionally, increased scientific understanding of degradation products and mechanisms may . u% {( w+ G2 b6 Chelp to determine the factors that could contribute to stability failures such as ambient temperature, - T( M9 c _) X1 P# E; q9 ghumidity, and light. Appropriate selection of packaging materials can be made to protect against1 N: g# T9 H1 S _
such factors. ! f1 V: g! p2 |; f) {Conclusion4 `# m+ d6 C: {" U4 v
An appropriately-designed stress study meshes well with the QbD approaches currently being" W6 x" h: x2 Z" B
promoted in the pharmaceutical industry. A well-designed stress study can provide insight in 9 w$ O8 D* c- \4 p& t$ W8 C/ V0 hchoosing the appropriate formulation for a proposed product prior to intensive formulation 2 u7 |2 t- _. U$ A5 ?development studies. A thorough knowledge of degradation, including mechanistic understanding # i ]! m+ A2 ?! |of potential degradation pathways, is the basis of a QbD approach for analytical method7 b1 y6 ~$ D2 b9 r+ n' i
development and is crucial in setting acceptance criteria for shelf-life monitoring. Stress testing ) C" ^6 [9 R' A! G- ]! Xcan provide useful insight into the selection of physical form, stereo-chemical stability of a drug 0 ~7 C& h( L" u3 y \- Bsubstance, packaging, and storage conditions. It is important to perform stress testing for generic ! p$ c4 L( e: ?0 I& vdrugs due to allowable qualitative and quantitative differences in formulation with respect to the7 }) J1 i; z: [
RLD, selection of manufacturing process, processing parameters, and packaging materials. 1 ^. o" L! W9 W+ c; ?Acknowledgments / e- {" t, p- D8 C KThe author would like to thank Bob Iser, Naiqi Ya, Dave Skanchy, Bing Wu, and Ashley Jung for8 w, O/ q3 C/ N' \
their scientific input and support.; F- o3 V, r9 X; G- w, e6 q
Ragine Maheswaran, PhD, is a CMC reviewer at the Office of Generic Drugs within the Office of5 `" G4 y+ q% ]8 I) _2 _7 [
Pharmaceutical Science, under the US Food and Drug Administration's Center for Drug/ Q- R" w0 [0 Z& ~+ [; e' K
Evaluation and Research, Ragine.Maheswaran@fda.hhs.gov 6 `' O) {2 J% f7 ` m& b2 _' UDisclaimer: The views and opinions in this article are only those of the author and do not) o' T" w0 @" g: A
necessarily reflect the views or policies of the US Food and Drug Administration.- I) p* d; y. ]! D7 a5 p4 P+ h X' i! z
References$ K4 k0 r: i5 C0 n+ }6 G
1. ICH, Q1A(R2) Stability Testing of New Drug Substances and Products (Geneva, Feb. 2003).3 \. S# }! u" ?+ [7 n8 D, a
2. ICH, Q1B Stability Testing: Photostability Testing of New Drug Substances and Products ! P% Q/ a& c9 p% ?& F(Geneva, Nov. 1996). 8 a0 N" i/ ~9 A7 s3. H. Brittain, Analytical Profiles of Drug Substances and Excipients (Academic Press, London, / n V' e# d& R l( [) C, ^6 f7 G2002).. }) c% q8 O2 h) ?7 [ |1 ~
4. A. Srinivasan and R. Iser, Pharm. Technol. 34 (1), 50–59 (2010). 4 o7 O- o$ [0 G4 T, T' s! D$ p- [$ u5. A. Srinivasan, R. Iser, and D. Gill, Pharm. Technol. 34(8), 45–51 (2010). " G& z$ C8 }! s7 h/ K" s& ^6. A. Srinivasan, R. Iser, and D. Gill, Pharm. Technol. 35 (2), 58–67 (2011).- }, t% G( s4 `/ R# T* @; Q
7. S. Klick, et al., Pharm.Technol. 29 (2) 48–66 (2005).8 X* e4 u6 z ]: s
8. K. M. Alsante, L. Martin and S. W. Baertschi, Pharm.Technol. 27 (2) 60-72 (2003). . \% r) h5 w& M/ Y2 a4 G0 W9. D. W. Reynolds, et al., Pharm.Technol. 26 (2), 48–56 (2002).2 v& r. h9 A# H5 D- O3 y4 k# R5 ^
10. K. M. Alsante et al., Advanced Drug Delivery Reviews 59, 29–37 (2007).& C1 k+ e" G- h
11. FDA, Guidance for Industry on Analytical Procedures and methods Validation Chemistry,0 g" `6 B5 ~1 b
Manufacturing, and Controls Documentation (draft) (Rockville, MD, Aug. 2000). . k1 j v1 |$ z7 C12. ICH, Q6A: Specifications: Test Procedures and Acceptance Criteria for New Drug Substances ' X/ T0 M% O4 y C$ e! Aand New Drug Products: Chemical Substances (Geneva, Oct. 1999).: i; m4 I( N2 q6 C2 _; a
13. ICH, Q3A(R2) Impurities in New Drug Substances (Geneva, Oct. 2006). 0 j+ C. C$ R" \& }$ G' P7 T& a14. ICH, Q3B(R2) Impurities in New Drug Products (Geneva, June 2006). $ B4 v4 }! `) n+ J9 Y% ^& z+ ~15. FDA, Guidance for Industry ANDAs: Impurities in Drug Substances (draft), (Rockville, MD,, B2 i& m& p) O9 O$ W
Aug. 2005). & O* l& \# ~# ]2 {16. FDA, Guidance for Industry ANDAs: Impurities in Drug Products (draft) (Rockville, MD,. ^2 F2 {9 x* L9 z# R- {$ j# a5 {5 l; a
Nov. 2010). 2 `4 r/ t0 F7 o17. EMA, Guideline on the Limits of Genotoxic Impurities, Committee for Medical Products for ) h9 t0 n0 [+ ?! q" s, T( E4 ?+ rHuman Use (CHMP) (Doc. Ref EMA/CHMP/QWP/251344/2006) (Jan. 1, 2007).6 V3 u" r: o6 r4 B5 g
18. K. A. Conners et al., Chemical Stability of Pharmaceuticals, Wiley and Sons, New York, New 2 v1 e! z* ^3 |* @ I OYork, 2nd Ed., p. 19 (1986).作者: ruichao2005 时间: 2015-7-15 08:01 PM
学习一下!!!!!!!!!作者: 一花一世界 时间: 2015-7-22 04:02 PM
正看学习,算是很有帮助的,只是这个和稳定性考察中的降解试验 我有点混淆了,