药群论坛

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
! a. N/ j/ x5 f. g# V
FDA Perspectives: Scientific Considerations of Forced Degradation Studies in ANDA
" O# p' X9 X3 w( vSubmissions: Q. _6 ], l! ~4 K$ u
The author outlines the scientific aspects of forced degradation studies that should be considered/ a. s; H8 e4 ~  J
in relation to ANDA submissions.
. {& x6 }( V* x. [7 hMay 2, 20126 ^8 r; d' |" E& q' y) z- |# ?
By:Ragine Maheswaran
- T2 o/ u, b$ ^" s9 h: CPharmaceutical Technology7 Q# s1 e0 S4 L; ?. [% G5 M& T
Volume 36, Issue 5, pp. 73-805 ^7 g3 O1 p- s7 G9 ?; `% H
Forced degradation is synonymous with stress testing and purposeful degradation. Purposeful
2 T7 w2 ?! r6 }, P1 ^2 H7 Kdegradation can be a useful tool to predict the stability of a drug substance or a drug product with
+ u  l! _( }6 x4 U1 K2 peffects on purity, potency, and safety. It is imperative to know the impurity profile and behavior of% U* f  A1 d$ O- Z3 t
a drug substance under various stress conditions. Forced degradation also plays an important role: d4 ]8 x: f% S" N
in the development of analytical methods, setting specifications, and design of formulations under3 R. y9 Q" e" I; m( Y
the quality-by-design (QbD) paradigm. The nature of the stress testing depends on the individual! H$ m/ G( N' A
drug substance and the type of drug product (e.g., solid oral dosage, lyophilized powders, and- d9 f4 s5 C0 r) H# _
liquid formulations) involved (1).5 u$ Q6 G9 k: \( M  c0 c/ B+ ^
The International Conference on Harmonization (ICH) Q1B guideline provides guidance for
. v0 A. G$ ?% g$ \, Pperforming photostability stress testing; however, there are no additional stress study5 t7 _& V3 ]! Q
recommendations in the ICH stability or validation guidelines (2). There is also limited
! S2 r+ `+ P' @5 Y4 T5 Yinformation on the details about the study of oxidation and hydrolysis. The drug substance. p' ?/ {( `( A2 E! K/ r8 Q
monographs of Analytical Profiles of Drug Substances and Excipients provide some information
  J( s0 Q& {$ E! bwith respect to different stress conditions of various drug substances (3).
# _( P- e- U" P0 rThe forced degradation information provided in the abbreviated new drug application (ANDA)
/ F4 d. r  @" {4 [8 Qsubmissions is often incomplete and in those cases deficiencies are cited. An overview of common
7 l8 a% n3 ~0 R2 E% ^deficiencies cited throughout the chemistry, manufacturing, and controls (CMC) section of the2 a  n$ g3 `( F: H; m3 R$ V
ANDAs has been published (4–6). Some examples of commonly cited deficiencies related to7 T, q3 o2 k& ]" j' z8 U! H
forced degradation studies include the following:
* a3 p- L/ E; Y* _Your drug substance does not show any degradation under any of the stress conditions. Please( H1 }" U* K+ m- n# c' [0 P; g+ C
repeat stress studies to obtain adequate degradation. If degradation is not achievable, please: n0 y' J) ~/ ]+ s+ c
provide your rationale.; |/ I% F7 i; H, @& D, e  K- e1 {
Please note that the conditions employed for stress study are too harsh and that most of your drug- ^1 o! P, s; M7 k5 ]0 L, {+ w8 J
substance has degraded. Please repeat your stress studies using milder conditions or shorter# u9 i  }# n0 A" f$ m
exposure time to generate relevant degradation products.
" D0 E- e2 C: F% _  qIt is noted that you have analyzed your stressed samples as per the assay method conditions. For7 G/ S, J$ Y$ d3 ^4 J
the related substances method to be stability indicating, the stressed samples should be analyzed
. g) E& }! |% D# Yusing related substances method conditions.6 Y" k  f2 C. w# U
Please state the attempts you have made to ensure that all the impurities including the degradation
: I" A) n* P. R3 @: p/ wproducts of the unstressed and the stressed samples are captured by your analytical method.# Y. |1 s2 @8 E9 f: V  U
Please provide a list summarizing the amount of degradation products (known and unknown) in3 y& g: J$ D, x& B$ z/ f+ e6 g7 G
your stressed samples.
3 F& }/ q6 J3 [* `) y+ e& s% O2 ~  APlease verify the peak height requirement of your software for the peak purity determination.
3 c7 t( [1 ?& W' k7 g. fPlease explain the mass imbalance of the stressed samples.1 C  h( V% c+ s( _
Please identify the degradation products that are formed due to drug-excipient interactions.
& F7 h7 G6 I+ b* R8 j( N5 jYour photostability study shows that the drug product is very sensitive to light. Please explain how& O: B- t) t8 C6 G
this is reflected in the analytical method, manufacturing process, product handling, etc.; o/ y. M- R1 y5 A+ B
In an attempt to minimize deficiencies in the ANDA submissions, some general recommendations6 {4 z8 B2 p& ?+ ^' P' r1 p
to conduct forced degradation studies, to report relevant information in the submission, and to
$ H9 j2 j& X% w6 z- l" @4 outilize the knowledge of forced degradation in developing stability indicating analytical methods,, W( }* X' _% Q% T8 ]' p4 ?* p
manufacturing process, product handling, and storage are provided in this article.
' a: Q( r8 z  v+ p1 J. ]Stress conditions
) K0 e- n2 y- {+ R) f0 P7 L/ FTypical stress tests include four main degradation mechanisms: heat, hydrolytic, oxidative, and( j# T8 h0 k8 ~: Q. I& O% n3 v/ K
photolytic degradation. Selecting suitable reagents such as the concentration of acid, base, or
1 c4 z* S; g  Z1 ~. uoxidizing agent and varying the conditions (e.g., temperature) and length of exposure can achieve
1 l8 D3 |  R6 S2 ~! dthe preferred level of degradation. Over-stressing a sample may lead to the formation of secondary5 c) w% m( D& f1 Y3 ~+ f
degradants that would not be seen in formal shelf-life stability studies and under-stressing may not
4 N/ j& H& h5 Q+ n0 o; qserve the purpose of stress testing. Therefore, it is necessary to control the degradation to a desired
4 i, k1 R# p5 e, L! X3 ]3 flevel. A generic approach for stress testing has been proposed to achieve purposeful degradation
. j+ o. D( {9 Uthat is predictive of long-term and accelerated storage conditions (7). The generally recommended8 g# K0 D! K  T" r6 |- }3 Q$ b
degradation varies between 5-20% degradation (7–10). This range covers the generally$ z& ]+ K( X% R  D# S* i7 M' b
permissible 10% degradation for small molecule pharmaceutical drug products, for which the
9 e, Y; O/ o& B2 E3 @stability limit is 90%-110% of the label claim. Although there are references in the literature that: L9 G' ]" B8 {$ A- K* s6 h( o
mention a wider recommended range (e.g., 10-30%), the more extreme stress conditions often8 \4 E9 q* \6 ^( H+ G! V
provide data that are confounded with secondary degradation products.3 S0 q9 C7 ^# ?/ `8 W
Photostability.
0 _# y1 w( u5 g- a8 l0 WPhotostability testing should be an integral part of stress testing, especially for photo-labile
+ i3 t% {3 [# [, scompounds. Some recommended conditions for photostability testing are described in ICH Q1B; H5 s* |: M: s  i. Z! u5 _
Photostability Testing of New Drug Substances and Products (2). Samples of drug substance, and" c: `- e; H6 s# g$ {% n( L" ?6 P
solid/liquid drug product, should be exposed to a minimum of 1.2 million lux hours and 200 watt
5 t# b: k$ r3 i# _hours per square meter light. The same samples should be exposed to both white and UV light. To
, `& h5 e, h) J2 \minimize the effect of temperature changes during exposure, temperature control may be& N8 h& @6 @8 z
necessary. The light-exposed samples should be analyzed for any changes in physical properties& Y- c* F+ n/ ?0 x( U: L' O" w3 q
such as appearance, clarity, color of solution, and for assay and degradants. The decision tree$ g: ^; b+ E) ]8 w9 a0 V4 \
outlined in the ICH Q1B can be used to determine the photo stability testing conditions for drug' M; k4 A, w5 T, @
products. The product labeling should reflect the appropriate storage conditions. It is also! T3 ]! Y1 O+ ]; w# e+ `2 a/ N4 L
important to note that the labeling for generic drug products should be concordant with that of the
- d% k; r7 i6 J* Areference listed drug (RLD) and with United States Pharmacopeia (USP) monograph
* X* e% n8 }# F% ~0 K! U  X- Krecommendations, as applicable.6 W) b( w9 k9 J( G* Q
Heat.
  Z' l; @& q  x' V. M7 M& EThermal stress testing (e.g., dry heat and wet heat) should be more strenuous than recommended
" Q  y6 D8 N' |. iICH Q1A accelerated testing conditions. Samples of solid-state drug substances and drug products
, l/ Y3 N  m+ _1 z* e# Wshould be exposed to dry and wet heat, whereas liquid drug products can be exposed to dry heat. It
( A: `1 g: J+ m5 kis recommended that the effect of temperature be studied in 10 °C increments above that for
& E% V2 `- K2 H( vroutine accelerated testing, and humidity at 75% relative humidity or greater (1). Studies may be
! W( g* t  X7 D2 A& J& |0 Uconducted at higher temperatures for a shorter period (10). Testing at multiple time points could! M5 |4 m* Q' p% ?# v
provide information on the rate of degradation and primary and secondary degradation products.
: l) g9 ?$ A, t+ EIn the event that the stress conditions produce little or no degradation due to the stability of a drug
# u# ~) @  W/ d3 V5 _- X6 Z3 qmolecule, one should ensure that the stress applied is in excess of the energy applied by
* y3 l7 ]; c" \: `9 Zaccelerated conditions (40 °C for 6 months) before terminating the stress study.
" _, j; p" }' u* aAcid and base hydrolysis.. t+ Y% Y' j9 a* ~* r7 r7 g' |
Acid and base hydrolytic stress testing can be carried out for drug substances and drug products in' z. I5 V8 S  [/ L3 `9 `& z3 w1 a
solution at ambient temperature or at elevated temperatures. The selection of the type and! {0 j' g0 s* A3 m- h
concentrations of an acid or a base depends on the stability of the drug substance. A strategy for
! Z) j! i8 s  O3 u1 i, M) `. tgenerating relevant stressed samples for hydrolysis is stated as subjecting the drug substance7 N% S. x' A# l- {9 u4 M" o7 {. H
solution to various pHs (e.g., 2, 7, 10–12) at room temperature for two weeks or up to a maximum
# Z- Z* a% b! z7 @/ oof 15% degradation (7). Hydrochloric acid or sulfuric acid (0.1 M to 1 M) for acid hydrolysis and8 Q$ i# q$ H9 X: E+ b' [& H
sodium hydroxide or potassium hydroxide (0.1 M to 1 M) for base hydrolysis are suggested as/ X+ D6 }5 T5 M
suitable reagents for hydrolysis (10). For lipophilic drugs, inert co-solvents may be used to5 _, f) w/ @% i+ y1 r
solubilize the drug substance. Attention should be given to the functional groups present in the
# [3 H9 L2 \& `7 T; H- n6 [9 zdrug molecule when selecting a co-solvent. Prior knowledge of a compound can be useful in
% A4 Q: b, t6 ^9 O! K# l3 B: v& Vselecting the stress conditions. For instance, if a compound contains ester functionality and is very
$ m0 W% A7 w! q. r) M( E; ^) f% x1 vlabile to base hydrolysis, low concentrations of a base can be used. Analysis of samples at various5 R9 }- d7 K) [2 M; l
intervals can provide information on the progress of degradation and help to distinguish primary
! ]8 }0 }- m2 }3 X4 v* }degradants from secondary degradants.
( E& p$ g) a8 pOxidation.
2 W9 I% G2 B# _5 Y; gOxidative degradation can be complex. Although hydrogen peroxide is used predominantly
6 A& H' Z* l. q0 Q2 sbecause it mimics possible presence of peroxides in excipients, other oxidizing agents such as
& J7 Q6 B* \! e" m* X. o- h! _metal ions, oxygen, and radical initiators (e.g., azobisisobutyronitrile, AIBN) can also be used.
. V" m, I- C/ G2 k: E1 T9 K5 JSelection of an oxidizing agent, its concentration, and conditions depends on the drug substance.0 x% k, E- Q; N  [: e" B4 s
Solutions of drug substances and solid/liquid drug products can be subjected to oxidative# ]! G& t. C/ ~, J
degradation. It is reported that subjecting the solutions to 0.1%-3% hydrogen peroxide at neutral
1 v& e5 {' W; q9 B% UpH and room temperature for seven days or up to a maximum 20% degradation could potentially3 M" f4 H! |; L9 J% W8 H2 U
generate relevant degradation products (10). Samples can be analyzed at different time intervals to
$ S' t1 U) {% z9 Wdetermine the desired level of degradation./ z) }* @/ j7 ?6 A& Q$ D
Different stress conditions may generate the same or different degradants. The type and extent of) H  o1 C: q0 K" h
degradation depend on the functional groups of the drug molecule and the stress conditions.2 P4 @. H+ @+ r' d4 a: s5 X
Analysis method
- I, g3 ?- n1 E0 u6 |# CThe preferred method of analysis for a stability indicating assay is reverse-phase
* m. J! G4 G& ^7 M) ~  D5 [high-performance liquid chromatography (HPLC). Reverse-phase HPLC is preferred for several
8 v/ q- F" ]. M1 dreasons, such as its compatibility with aqueous and organic solutions, high precision, sensitivity,
$ s4 Z) r9 j& F- S  y4 F: t1 Oand ability to detect polar compounds. Separation of peaks can be carried out by selecting$ v2 S0 d. ?5 O! A! N0 c6 s
appropriate column type, column temperature, and making adjustment to mobile phase pH.  u8 Z# J6 w8 r# f, k) f
Poorly-retained, highly polar impurities should be resolved from the solvent front. As part of; D6 _- b/ `! a5 o7 v
method development, a gradient elution method with varying mobile phase composition (very low
' u" |9 E4 F% V* M( Torganic composition to high organic composition) may be carried out to capture early eluting
" d) C/ H" B: h, q) k& Yhighly polar compounds and highly retained nonpolar compounds. Stressed samples can also be
4 @; O; j: s) O2 d& iscreened with the gradient method to assess potential elution pattern. Sample solvent and mobile) y- g9 L6 O2 s: p3 l& E" ~
phase should be selected to afford compatibility with the drug substance, potential impurities, and
2 g, ~* J% G9 @+ l  ldegradants. Stress sample preparation should mimic the sample preparation outlined in the
* B/ b0 R' [( W, ^analytical procedure as closely as possible. Neutralization or dilution of samples may be necessary% F4 O7 `1 Z/ V& w3 Z/ V
for acid and base hydrolyzed samples. Chromatographic profiles of stressed samples should be' m1 n# s& }; b3 d# Y) O( m
compared to those of relevant blanks (containing no active) and unstressed samples to determine
  Z0 a" |- M% n. Zthe origin of peaks. The blank peaks should be excluded from calculations. The amount of" L; H( \4 J/ ]$ p7 T* Z! ~7 i
impurities (known and unknown) obtained under each stress condition should be provided along
6 k0 J: s$ J+ \4 dwith the chromatograms (full scale and expanded scale showing all the peaks) of blanks,
4 j( v" C6 u; u/ x" `0 f  D0 Bunstressed, and stressed samples. Additionally, chiral drugs should be analyzed with chiral
. b; Z% K, B! c! A& j( @6 ~methods to establish stereochemical purity and stability (11, 12).
5 C9 G7 Y( A8 a" S8 q2 C% X' {7 oThe analytical method of choice should be sensitive enough to detect impurities at low levels (i.e.,
' Y% o$ ^. l: j/ J0.05% of the analyte of interest or lower), and the peak responses should fall within the range of' ^7 F' F# {+ h/ m8 N( v3 w7 R
detector's linearity. The analytical method should be capable of capturing all the impurities formed8 l5 p& k8 ~+ t2 l/ p' l) @$ f( H# b
during a formal stability study at or below ICH threshold limits (13, 14). Degradation product% K; M8 K  E- U( t% O
identification and characterization are to be performed based on formal stability results in3 g# b* Q+ H, T. ?2 R& L
accordance with ICH requirements. Conventional methods (e.g., column chromatography) or+ [  P6 p$ S' Y' `+ E: k; X
hyphenated techniques (e.g., LC–MS, LC–NMR) can be used in the identification and, S- Y( q" d5 {, i  ^/ @$ P0 U0 _
characterization of the degradation products. Use of these techniques can provide better insight
% C2 e4 N7 L+ n. ?+ Q! t1 Hinto the structure of the impurities that could add to the knowledge space of potential structural
# ]' p; ^1 q; ~2 H" _% U% y4 lalerts for genotoxicity and the control of such impurities with tighter limits (12–17). It should be
6 o3 y# N! u) Z: u: T0 a* j' jnoted that structural characterization of degradation products is necessary for those impurities that
4 P  G" ]. V* y: _are formed during formal shelf-life stability studies and are above the qualification threshold limit9 J) L. ^/ M8 H1 p0 e, p' w
(13).
/ s# Y7 J- @' u4 J1 {  bVarious detection types can be used to analyze stressed samples such as UV and mass
4 p( w- }- Q' {, ?, ]spectroscopy. The detector should contain 3D data capabilities such as diode array detectors or
/ H  e7 Q1 t( Omass spectrometers to be able to detect spectral non-homogeneity. Diode array detection also
+ _( x& k6 Z: Y  q2 E9 zoffers the possibility of checking peak profile for multiple wavelengths. The limitation of diode
" U5 l- `* Y' F% P0 S+ o" |5 W$ carray arises when the UV profiles are similar for analyte peak and impurity or degradant peak and
6 c6 X. j) V( P! K7 nthe noise level of the system is high to mask the co-eluting impurities or degradants. Compounds7 ^1 S: w5 I' r+ c
of similar molecular weights and functional groups such as diastereoisomers may exhibit similar+ l9 s5 g; y5 ]7 e! A
UV profiles. In such cases, attempts must be made to modify the chromatographic parameters to/ o& u$ c7 t" V" z4 Z) x7 C- j
achieve necessary separation. An optimal wavelength should be selected to detect and quantitate
1 J2 E- C. Q) k4 call the potential impurities and degradants. Use of more than one wavelength may be necessary, if! G/ X7 [" j6 e9 W1 f' M- S
there is no overlap in the UV profile of an analyte and impurity or degradant peaks. A valuable) y3 z3 I0 A$ {; O
tool in method development is the overlay of separation signals at different wavelengths to
3 |  p; o" R( ?$ P# [+ ~; u- bdiscover dissimilarities in peak profiles.2 w$ W6 ]. H/ h* u$ z# o0 M/ C' |: R
Peak purity analysis.
9 e/ h3 P, O0 w$ l! |2 VPeak purity is used as an aid in stability indicating method development. The spectral uniqueness
: Q( E3 a1 x" I, k- m) zof a compound is used to establish peak purity when co-eluting compounds are present.
3 n4 ]+ c% Z, F# Q4 ]' ?3 P9 NPeak purity or peak homogeneity of the peaks of interest of unstressed and stressed samples! V+ i  P" a: i/ \! x' D5 y
should be established using spectral information from a diode array detector. When instrument7 ]; m- Y1 }$ x' I+ F: e( n
software is used for the determination of spectral purity of a peak, relevant parameters should be
. v4 z3 z1 R. H" pset up in accordance with the manufacturer's guidance. Attention should be given to the peak2 W7 g5 l' Z  s+ [7 H
height requirement for establishing spectral purity. UV detection becomes non linear at higher
# @; K) V% P3 Wabsorbance values. Thresholds should be set such that co-eluting peaks can be detected. Optimum
' Z7 {' {! Z7 t( Y" flocation of reference spectra should also be selected. The ability of the software to automatically- X5 l' r1 j& i6 O, n' A
correct spectra for continuously changing solvent background in gradient separations should be5 l( E8 j- b; Q! q' z( `3 s' h& j
ascertained.6 w( n% p# N2 ~- y- j; ]
Establishing peak purity is not an absolute proof that the peak is pure and that there is no
! b8 A! U1 J* R; K- xco-elution with the peak of interest. Limitations to peak purity arise when co-eluting peaks are
6 J( m1 r9 |! t  Rspectrally similar, or below the detection limit, or a peak has no chromophore, or when they are; U; t- B4 ~9 ]4 {
not resolved at all.) x3 Y: t3 F; m
Mass balance.% e! T2 n( T3 @1 X
Mass balance establishes adequacy of a stability indicating method though it is not achievable in
. E1 b* ]  D6 \  Uall circumstances. It is performed by adding the assay value and the amounts of impurities and
* G7 Y6 T8 l9 @2 Xdegradants to evaluate the closeness to 100% of the initial value (unstressed assay value) with due7 ^% I( C9 Z* F
consideration of the margin of analytical error (1).
: [8 o( h; L$ A, A9 L! r$ QSome attempt should be made to establish a mass balance for all stressed samples. Mass
) z- K- v0 |7 Q: S% o2 l' Pimbalance should be explored and an explanation should be provided. Varying responses of$ V  e0 L; F1 D! Y% n" Y- d  d
analyte and impurity peaks due to differences in UV absorption should also be examined by the0 o2 G3 x) u- Q) c1 A' [: a. \
use of external standards. Potential loss of volatile impurities, formation of non-UV absorbing
3 o5 L7 W! R( A7 S' L3 b$ ?. wcompounds, formation of early eluants, and potential retention of compounds in the column
% X; ^% S: g( ~: f+ T- I* i9 v3 Tshould be explored. Alternate detection techniques such as RI LC/MS may be employed to5 Q  y$ W7 F: y0 O! E) j3 N
account for non-UV absorbing degradants.4 k1 G: {! Z2 n5 z% E" e/ a% |# t- o
Termination of study. E0 J% A+ T% o1 }+ w; D
Stress testing could be terminated after ensuring adequate exposure to stress conditions. Typical
7 v7 B' J" |0 Tactivation energy of drug substance molecules varies from 12–24 kcal/mol (18). A compound may5 W! G# h& S2 k4 y, M2 t+ g$ v
not necessarily degrade under every single stress condition, and general guideline on exposure) n& h2 S. n' q# F
limit is cited in a review article (10). In circumstances where some stable drugs do not show any$ A; C' g- c$ K6 j# T
degradation under any of the stress conditions, specificity of an analytical method can be; n0 s7 G6 g2 P4 r: H9 ?
established by spiking the drug substance or placebo with known impurities and establishing
5 r5 t8 |1 P6 R1 C! K; B# {% uadequate separation.% k/ W0 K9 H( x1 U" ?
Other considerations
: S1 H& e! L% o' I3 F5 p/ |7 a2 f, \Stress testing may not be necessary for drug substances and drug products that have" g6 Z5 W4 z- D, P* }! f0 R' |
pharmacopeial methods and are used within the limitations outlined in USP <621>. In the case
  M& y- X9 x; ~5 n5 ?* }* Bwhere a generic drug product uses a different polymorphic form from the RLD, the drug substance
$ I3 Y' M& n* Q  Sshould be subjected to stress testing to evaluate the physiochemical changes of the polymorphic( U( y& b( P5 m$ n! d
form because different polymorphic forms may exhibit different stability characteristics.. ]( a' F! Z( U# T
Forced degradation in QbD paradigm
/ Y; b% o2 Q: S/ m% F2 CA systematic process of manufacturing quality drug products that meet the predefined targets for$ C) i7 s) G4 x! c
the critical quality attributes (CQA) necessitates the use of knowledge obtained in forced2 m* `& A$ U6 l
degradation studies.) m) ]7 A5 F( R
A well-designed, forced degradation study is indispensable for analytical method development in a+ u% V; ~6 Y$ m" X+ _: j
QbD paradigm. It helps to establish the specificity of a stability indicating method and to predict8 s7 W( H6 k" ?
potential degradation products that could form during formal stability studies. Incorporating all
" ]! w7 n* z2 b9 x1 jpotential impurities in the analytical method and establishing the peak purity of the peaks of; ~1 r7 c: Q% s# M" H* `7 }
interest helps to avoid unnecessary method re-development and revalidation.. D$ c4 q/ B& d' d0 z) X
Knowledge of chemical behavior of drug substances under various stress conditions can also
/ f) V. O5 \3 i, Z" p" wprovide useful information regarding the selection of excipients for formulation development.1 g' m) h) H8 i1 e& D
Excipient compatibility is an integral part of understanding potential formulation interactions
: h& L: t+ J+ [0 E: {0 S3 wduring product development and is a key part of product understanding. Degradation products due& |! x& [& q- f* R- q; y/ k
to drug-excipient interaction or drug-drug interaction in combination products can be examined by8 w2 Z' S4 K' U1 S( ^
stressing samples of drug substance, drug product, and placebo separately and comparing the
9 k+ b: x( P' s1 _- Jimpurity profiles. Information obtained regarding drug-related peaks and non-drug-related peaks
+ z2 ~0 Y' H7 X) H5 w% W6 p+ }can be used in the selection and development of more stable formulations. For instance, if a drug
3 B% z- @7 `8 ^6 W, H  Wsubstance is labile to oxidation, addition of an antioxidant may be considered for the formulation.8 D+ M1 a' ?8 a% t; R% `/ C0 E
For drug substances that are labile to acid or undergo stereochemical conversion in acidic medium,
. `- e" d5 }, m' M; w7 hdelayed-release formulations may be necessary. Acid/base hydrolysis testing can also provide1 I  v9 ?" e) d6 z
useful insight in the formulation of drug products that are liquids or suspensions.
3 C% k; W" M: W. i4 FKnowledge gained in forced degradation studies can facilitate improvements in the manufacturing0 n6 I+ m4 d7 C& u1 L
process. If a photostability study shows a drug substance to be photolabile, caution should be9 X6 c0 M0 }* j: u$ D. b! l
taken during the manufacturing process of the drug product. Useful information regarding process
, l2 [1 J" N2 M; e& \: n! I0 D/ Pdevelopment (e.g., wet versus dry processing, temperature selection) can be obtained from thermal8 W: n' U: h- ]* K% ?
stress testing of drug substance and drug product.
5 _: g2 @! O8 z) a) Z" w1 D& f' pAdditionally, increased scientific understanding of degradation products and mechanisms may
7 X  {! I3 N/ v3 a( \help to determine the factors that could contribute to stability failures such as ambient temperature,
6 t3 h7 h7 n$ Ihumidity, and light. Appropriate selection of packaging materials can be made to protect against
- @  `" m6 E; r" Usuch factors.
: Y2 `: `+ _0 L1 @8 T9 F; `Conclusion. l' F: y* Z: I* p7 g
An appropriately-designed stress study meshes well with the QbD approaches currently being# q: `" Y# M8 r7 L9 w. l7 @3 h9 A8 x4 T
promoted in the pharmaceutical industry. A well-designed stress study can provide insight in
& ^% G6 u- w( ~, Ychoosing the appropriate formulation for a proposed product prior to intensive formulation
: l+ S$ j! d$ V6 ^$ x" o' mdevelopment studies. A thorough knowledge of degradation, including mechanistic understanding
! y( K/ X  ?. s: h8 ?  w3 ^- Qof potential degradation pathways, is the basis of a QbD approach for analytical method. ^$ [2 A" h' H7 ]- u
development and is crucial in setting acceptance criteria for shelf-life monitoring. Stress testing
! W( L2 u, o/ m3 e5 g4 D3 Vcan provide useful insight into the selection of physical form, stereo-chemical stability of a drug
- R- e* m5 p! U0 P% xsubstance, packaging, and storage conditions. It is important to perform stress testing for generic
2 O- X; v+ P; m$ h4 _: z  Q+ Ddrugs due to allowable qualitative and quantitative differences in formulation with respect to the1 m" h) a; H; |
RLD, selection of manufacturing process, processing parameters, and packaging materials.
1 M$ {7 F& C7 Z5 {: ?. M( O4 rAcknowledgments
/ u* ^# J, t1 P$ `0 J7 a# OThe author would like to thank Bob Iser, Naiqi Ya, Dave Skanchy, Bing Wu, and Ashley Jung for
3 _% b- m) z7 rtheir scientific input and support.! k7 U1 f$ P/ A4 p
Ragine Maheswaran, PhD, is a CMC reviewer at the Office of Generic Drugs within the Office of, }$ y" @# J9 p4 s, h; z
Pharmaceutical Science, under the US Food and Drug Administration's Center for Drug+ [9 g; y* a- T5 {" I" N
Evaluation and Research, Ragine.Maheswaran@fda.hhs.gov: B% \4 |% m" M0 @8 [6 B0 w8 \
Disclaimer: The views and opinions in this article are only those of the author and do not" a2 [; b8 ~8 b5 L
necessarily reflect the views or policies of the US Food and Drug Administration.3 e$ n8 f4 G$ L5 s1 R
References
% k3 S. M8 P5 d) b- w* s- p, ]9 |7 G/ h1. ICH, Q1A(R2) Stability Testing of New Drug Substances and Products (Geneva, Feb. 2003).3 y  N, ^1 V( w2 Z$ F4 {, A8 c
2. ICH, Q1B Stability Testing: Photostability Testing of New Drug Substances and Products
; t' R* Z: F7 _, d1 i( w(Geneva, Nov. 1996).) F- F. Z  m& A0 ^8 e9 n
3. H. Brittain, Analytical Profiles of Drug Substances and Excipients (Academic Press, London,
: O7 Y/ G, N8 s- d2002).
2 R8 P$ F7 n/ B/ E: I4. A. Srinivasan and R. Iser, Pharm. Technol. 34 (1), 50–59 (2010).! a4 ~0 ?0 m+ H
5. A. Srinivasan, R. Iser, and D. Gill, Pharm. Technol. 34(8), 45–51 (2010).) X, e$ T8 X3 n) n! G
6. A. Srinivasan, R. Iser, and D. Gill, Pharm. Technol. 35 (2), 58–67 (2011).
0 r1 [$ t% @( K; a7. S. Klick, et al., Pharm.Technol. 29 (2) 48–66 (2005).
9 w# l9 R5 n- Q% J* w0 H/ n8. K. M. Alsante, L. Martin and S. W. Baertschi, Pharm.Technol. 27 (2) 60-72 (2003).$ I5 T3 f! P; e5 H1 p* E
9. D. W. Reynolds, et al., Pharm.Technol. 26 (2), 48–56 (2002).) v4 _( g8 j' c1 M
10. K. M. Alsante et al., Advanced Drug Delivery Reviews 59, 29–37 (2007).
' O. m" V1 l5 U; \' e7 l11. FDA, Guidance for Industry on Analytical Procedures and methods Validation Chemistry,
9 O& X6 h8 i  S) a+ [Manufacturing, and Controls Documentation (draft) (Rockville, MD, Aug. 2000).
6 ]4 a/ L( l% e! w( t2 e$ i2 Z, N12. ICH, Q6A: Specifications: Test Procedures and Acceptance Criteria for New Drug Substances( Q- o1 G) a4 P0 \- B
and New Drug Products: Chemical Substances (Geneva, Oct. 1999).+ ^# _5 I) N* O# c
13. ICH, Q3A(R2) Impurities in New Drug Substances (Geneva, Oct. 2006).
0 d: P! }* H. }; u14. ICH, Q3B(R2) Impurities in New Drug Products (Geneva, June 2006).- c) B1 L6 m# [: {) \1 e+ r+ s9 O+ U
15. FDA, Guidance for Industry ANDAs: Impurities in Drug Substances (draft), (Rockville, MD,
, z  o; v0 w# }" E: o5 z+ GAug. 2005).0 W* ~. k" o$ l$ Z! ~. a
16. FDA, Guidance for Industry ANDAs: Impurities in Drug Products (draft) (Rockville, MD,
2 K9 O. c9 D" Q& Y7 O4 Y$ zNov. 2010).7 j$ h4 f3 x) n" z3 |/ d
17. EMA, Guideline on the Limits of Genotoxic Impurities, Committee for Medical Products for
6 a$ J: r- g0 k/ s0 _7 |( L, oHuman Use (CHMP) (Doc. Ref EMA/CHMP/QWP/251344/2006) (Jan. 1, 2007).
" W$ j. k  |2 g, u18. K. A. Conners et al., Chemical Stability of Pharmaceuticals, Wiley and Sons, New York, New
& |. ~8 c: F3 nYork, 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,




欢迎光临 药群论坛 (http://yaoqun.net/) Powered by Discuz! X3.2