药群论坛

 找回密码
 立即注册

只需一步,快速开始

查看: 1268|回复: 2
打印 上一主题 下一主题

[国际注册] FDA 关于破坏实验的一些最新看法和要求

[复制链接]
跳转到指定楼层
楼主
naren4545 发表于 2015-7-15 15:36:09 | 只看该作者 回帖奖励 |倒序浏览 |阅读模式

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

9 L" p$ E: d2 @5 }' a# @" PFDA Perspectives: Scientific Considerations of Forced Degradation Studies in ANDA
; W) R5 r0 U  D" l0 E3 PSubmissions
  q5 Z9 B0 G) T4 k0 V0 ?- jThe author outlines the scientific aspects of forced degradation studies that should be considered
' b/ E, i4 ~% o9 X, H% a7 Gin relation to ANDA submissions.
- l6 @9 V5 l: H# g' @# t* B7 qMay 2, 2012
& s# g2 D2 }9 V8 qBy:Ragine Maheswaran; x; Q- Y' @$ ?8 `* q' E% v+ {% z
Pharmaceutical Technology; d3 L4 A  l( @0 N3 g
Volume 36, Issue 5, pp. 73-80
, ]) a1 r( @5 z8 H* R4 NForced degradation is synonymous with stress testing and purposeful degradation. Purposeful
: x+ W1 F& O/ ^/ Z- ~. }degradation can be a useful tool to predict the stability of a drug substance or a drug product with: o4 f5 t* ]9 a
effects on purity, potency, and safety. It is imperative to know the impurity profile and behavior of
! j1 o' P! a- |6 r4 za drug substance under various stress conditions. Forced degradation also plays an important role
# R( g0 c' k. L* |/ ~2 W: W& _0 @+ kin the development of analytical methods, setting specifications, and design of formulations under
& t  \* ?" o) u3 b+ |the quality-by-design (QbD) paradigm. The nature of the stress testing depends on the individual+ H6 y3 r0 c7 a/ u: K- k* l
drug substance and the type of drug product (e.g., solid oral dosage, lyophilized powders, and
& p; s. ?) U* p7 E+ l9 \& n% A; jliquid formulations) involved (1).. [$ H& ?' x! |+ N: K4 D9 K
The International Conference on Harmonization (ICH) Q1B guideline provides guidance for
/ i+ H& d4 z: S$ Pperforming photostability stress testing; however, there are no additional stress study
$ u- K0 ^# m& C+ J5 Xrecommendations in the ICH stability or validation guidelines (2). There is also limited
  A6 }' n4 t& T) @5 E5 V+ c8 Ginformation on the details about the study of oxidation and hydrolysis. The drug substance
% r: n# N9 `  v/ E' @monographs of Analytical Profiles of Drug Substances and Excipients provide some information
4 L! z- d7 c; `3 S1 \with respect to different stress conditions of various drug substances (3).
6 a  N* V# \  y" j7 m5 {8 J- iThe forced degradation information provided in the abbreviated new drug application (ANDA)7 m0 I+ Y( _+ Z5 G: e3 e8 E
submissions is often incomplete and in those cases deficiencies are cited. An overview of common. l  W! N1 ?' ~* b1 R* |
deficiencies cited throughout the chemistry, manufacturing, and controls (CMC) section of the0 p+ B2 a2 x2 t# r0 D
ANDAs has been published (4–6). Some examples of commonly cited deficiencies related to3 \- ~; A$ |9 ~% d+ X# L
forced degradation studies include the following:
8 J% D% |6 z' ~2 D# L$ UYour drug substance does not show any degradation under any of the stress conditions. Please
5 @9 O, [: u& N2 M7 z% s) A3 srepeat stress studies to obtain adequate degradation. If degradation is not achievable, please
" S; [1 |" z5 d- B: q, Aprovide your rationale.4 C" A3 v0 b* S! ~* W
Please note that the conditions employed for stress study are too harsh and that most of your drug% f- b; R% \. i/ T
substance has degraded. Please repeat your stress studies using milder conditions or shorter
& q1 d$ p/ j- o: ~- ]" uexposure time to generate relevant degradation products.9 I) N) P+ z) E" o0 \' [) ~
It is noted that you have analyzed your stressed samples as per the assay method conditions. For% l0 }% l( P7 W, w6 M
the related substances method to be stability indicating, the stressed samples should be analyzed8 v! o! Y  R7 M( A5 f0 z6 F; b. S
using related substances method conditions.# H: i! Y) |% H
Please state the attempts you have made to ensure that all the impurities including the degradation4 C# o# u3 C& H. J6 _
products of the unstressed and the stressed samples are captured by your analytical method.
: G" r2 R$ _, L9 p  M1 N$ PPlease provide a list summarizing the amount of degradation products (known and unknown) in" Z6 K, R3 g8 X9 W$ \
your stressed samples.
# w/ D' q8 k& ^" }9 c7 FPlease verify the peak height requirement of your software for the peak purity determination.
4 t/ S2 d0 C- f$ o0 x# o9 |8 K/ U: ePlease explain the mass imbalance of the stressed samples.% h! T7 B/ t) O. S, P' [
Please identify the degradation products that are formed due to drug-excipient interactions.
; s9 O) D- O$ \; HYour photostability study shows that the drug product is very sensitive to light. Please explain how
' w5 T7 }8 m+ ?/ G. {* h. j' Sthis is reflected in the analytical method, manufacturing process, product handling, etc.  D! {0 F0 L- q2 W' J9 p7 J5 D
In an attempt to minimize deficiencies in the ANDA submissions, some general recommendations. z7 k- V- J# N9 @* H; `
to conduct forced degradation studies, to report relevant information in the submission, and to& o3 ~5 [# z: d, k3 B' S# x
utilize the knowledge of forced degradation in developing stability indicating analytical methods,
6 M) I+ J0 Z/ Q8 P+ @! w* K) r& imanufacturing process, product handling, and storage are provided in this article.# ]: q, i4 }6 E
Stress conditions
. ~- U8 m& L+ I! G2 Y0 }Typical stress tests include four main degradation mechanisms: heat, hydrolytic, oxidative, and. r6 e7 ]* U% g& D2 Q- w
photolytic degradation. Selecting suitable reagents such as the concentration of acid, base, or/ ?, z: M' N2 j4 j% m! r$ o
oxidizing agent and varying the conditions (e.g., temperature) and length of exposure can achieve
* S7 A4 c* k  s: Q2 ?( w. qthe preferred level of degradation. Over-stressing a sample may lead to the formation of secondary
. G" o8 x% n) x- D" {5 p2 Y; Z. i9 tdegradants that would not be seen in formal shelf-life stability studies and under-stressing may not
: k& Y* u7 x! Aserve the purpose of stress testing. Therefore, it is necessary to control the degradation to a desired! L- L, a$ K: a
level. A generic approach for stress testing has been proposed to achieve purposeful degradation
2 X: [7 U" j6 H4 m' Y6 [that is predictive of long-term and accelerated storage conditions (7). The generally recommended) v! _5 W. y/ A' C* _% V
degradation varies between 5-20% degradation (7–10). This range covers the generally' R+ @: |6 M) l; q. i; o9 L$ u
permissible 10% degradation for small molecule pharmaceutical drug products, for which the4 S9 c  R& i! B/ _
stability limit is 90%-110% of the label claim. Although there are references in the literature that& m/ X. a& l/ I' |+ H+ e& w
mention a wider recommended range (e.g., 10-30%), the more extreme stress conditions often
7 `7 l3 ^$ z" e% h# p+ n5 t2 xprovide data that are confounded with secondary degradation products.5 o5 D+ Q+ `3 C( v* a2 k' L- |
Photostability.
. H& r/ q' p/ Q3 W( V/ G: {0 e" EPhotostability testing should be an integral part of stress testing, especially for photo-labile* s6 s1 [$ x+ c$ }3 x* L. ?
compounds. Some recommended conditions for photostability testing are described in ICH Q1B
* d% b  x% I$ w8 ?2 {3 f6 XPhotostability Testing of New Drug Substances and Products (2). Samples of drug substance, and) S3 }. z$ @/ P0 r# w, H
solid/liquid drug product, should be exposed to a minimum of 1.2 million lux hours and 200 watt2 s, O/ Z( d2 f* q# I) H
hours per square meter light. The same samples should be exposed to both white and UV light. To
6 a6 A$ k! F9 J  e7 `minimize the effect of temperature changes during exposure, temperature control may be7 {; X, K) l4 d. y6 j! q; x( z
necessary. The light-exposed samples should be analyzed for any changes in physical properties
& o- P( l" ]$ g5 o& Wsuch as appearance, clarity, color of solution, and for assay and degradants. The decision tree
2 L7 o9 [! _: H! zoutlined in the ICH Q1B can be used to determine the photo stability testing conditions for drug6 J" l9 |1 ?: p
products. The product labeling should reflect the appropriate storage conditions. It is also5 C8 s4 X) d( o6 I/ b
important to note that the labeling for generic drug products should be concordant with that of the
0 B7 q) q# z. `! zreference listed drug (RLD) and with United States Pharmacopeia (USP) monograph* _4 l, i) {0 T. F- j( b; }7 W
recommendations, as applicable.
( l1 J4 r$ v. d* \  N( rHeat.9 [/ \+ b' n( m8 h0 J, s0 K8 S
Thermal stress testing (e.g., dry heat and wet heat) should be more strenuous than recommended5 Y# }8 D" `4 ]6 F
ICH Q1A accelerated testing conditions. Samples of solid-state drug substances and drug products! k: N, D% p. e" M
should be exposed to dry and wet heat, whereas liquid drug products can be exposed to dry heat. It( c5 z5 V6 P; O$ y- i% c
is recommended that the effect of temperature be studied in 10 °C increments above that for, A- q8 X$ F3 y/ k+ [
routine accelerated testing, and humidity at 75% relative humidity or greater (1). Studies may be
; X2 Z' |( d: w1 }5 _4 F1 Rconducted at higher temperatures for a shorter period (10). Testing at multiple time points could4 z  _& u1 x, ]/ h
provide information on the rate of degradation and primary and secondary degradation products.9 K) \0 }. x: ^! H9 N
In the event that the stress conditions produce little or no degradation due to the stability of a drug* U' j6 c) ^2 M0 x# T
molecule, one should ensure that the stress applied is in excess of the energy applied by
( e) k/ G; z  m8 Y4 ]1 y4 Eaccelerated conditions (40 °C for 6 months) before terminating the stress study.  u- H5 u, @8 f1 ], y) B* e5 U2 }
Acid and base hydrolysis.
4 v4 B& r! R9 T9 T8 [Acid and base hydrolytic stress testing can be carried out for drug substances and drug products in
4 p6 ?8 T- T8 q+ D5 f0 qsolution at ambient temperature or at elevated temperatures. The selection of the type and
6 {. {( J  K; K8 Xconcentrations of an acid or a base depends on the stability of the drug substance. A strategy for
9 f9 ?! J( I7 F3 U  m) v, Ggenerating relevant stressed samples for hydrolysis is stated as subjecting the drug substance1 F7 J" u! L0 e4 U4 F
solution to various pHs (e.g., 2, 7, 10–12) at room temperature for two weeks or up to a maximum
0 Z4 E( e  X2 Y: w. mof 15% degradation (7). Hydrochloric acid or sulfuric acid (0.1 M to 1 M) for acid hydrolysis and
" L5 D' Q! C* W( P4 c  }. f6 b4 `9 t  Asodium hydroxide or potassium hydroxide (0.1 M to 1 M) for base hydrolysis are suggested as
5 n8 l! ?( N+ \) g1 ksuitable reagents for hydrolysis (10). For lipophilic drugs, inert co-solvents may be used to. b* ~% J( B" A: \
solubilize the drug substance. Attention should be given to the functional groups present in the7 \* ?8 A8 C) s" \
drug molecule when selecting a co-solvent. Prior knowledge of a compound can be useful in6 o1 l* W' s2 ^  n' o7 G
selecting the stress conditions. For instance, if a compound contains ester functionality and is very- S/ l' ]; {4 U9 B% q7 r8 l
labile to base hydrolysis, low concentrations of a base can be used. Analysis of samples at various
' D! f* L, I* Z# Z* x7 @intervals can provide information on the progress of degradation and help to distinguish primary
* ~2 @5 I. W3 E) Edegradants from secondary degradants.! F) b7 o: W# y9 y1 g
Oxidation.5 K3 P/ q7 A9 K, O: k5 b
Oxidative degradation can be complex. Although hydrogen peroxide is used predominantly. U/ f+ S8 o6 s* U/ m% [
because it mimics possible presence of peroxides in excipients, other oxidizing agents such as1 g0 b2 s) f' Q3 G) A! p" N
metal ions, oxygen, and radical initiators (e.g., azobisisobutyronitrile, AIBN) can also be used.
9 f+ x2 {  U, u+ Z" uSelection of an oxidizing agent, its concentration, and conditions depends on the drug substance.' k1 ]3 F. [  O( G
Solutions of drug substances and solid/liquid drug products can be subjected to oxidative
; }* C2 F  |0 S# C8 r* Xdegradation. It is reported that subjecting the solutions to 0.1%-3% hydrogen peroxide at neutral6 A# ?7 M1 U9 J3 S/ v. E/ `0 g
pH and room temperature for seven days or up to a maximum 20% degradation could potentially' b* y& |# n# A
generate relevant degradation products (10). Samples can be analyzed at different time intervals to
. H: }* u# x  X9 I9 b3 S$ vdetermine the desired level of degradation.' F7 O3 n. V& j' `0 n4 v  R
Different stress conditions may generate the same or different degradants. The type and extent of
- ^7 ~5 R4 \: ~# pdegradation depend on the functional groups of the drug molecule and the stress conditions.
! M* d0 g8 O: oAnalysis method
/ p( p, k9 q# ?& yThe preferred method of analysis for a stability indicating assay is reverse-phase
/ ]% G7 F4 z- u/ C" Thigh-performance liquid chromatography (HPLC). Reverse-phase HPLC is preferred for several5 s. ^  E1 ?* i: v/ Z* y
reasons, such as its compatibility with aqueous and organic solutions, high precision, sensitivity,
) B% D7 o# D: r+ Iand ability to detect polar compounds. Separation of peaks can be carried out by selecting  M6 L4 v1 K9 U8 D) ^
appropriate column type, column temperature, and making adjustment to mobile phase pH.
$ ~! u. k: |( v; g# i1 c+ jPoorly-retained, highly polar impurities should be resolved from the solvent front. As part of
) ^8 T' Q6 H2 c$ w6 bmethod development, a gradient elution method with varying mobile phase composition (very low
. O% z$ E4 f2 g. C2 Torganic composition to high organic composition) may be carried out to capture early eluting/ n3 X# j$ E9 ?. _% H
highly polar compounds and highly retained nonpolar compounds. Stressed samples can also be
* x) E& o4 S4 Y* f; Rscreened with the gradient method to assess potential elution pattern. Sample solvent and mobile
  G% E8 |3 G/ m- wphase should be selected to afford compatibility with the drug substance, potential impurities, and3 C* B3 ~, O' s) Z8 e6 k/ J
degradants. Stress sample preparation should mimic the sample preparation outlined in the
, t" i/ f' q2 B- wanalytical procedure as closely as possible. Neutralization or dilution of samples may be necessary7 ]  Q" Y$ D6 |( h6 i# N
for acid and base hydrolyzed samples. Chromatographic profiles of stressed samples should be' j0 P3 o6 @1 N% d
compared to those of relevant blanks (containing no active) and unstressed samples to determine
' d8 X9 x. u* \9 m: ~the origin of peaks. The blank peaks should be excluded from calculations. The amount of: |8 m; R( m3 W4 ^! E7 O" h
impurities (known and unknown) obtained under each stress condition should be provided along5 ?& n) N/ g& e* C( V
with the chromatograms (full scale and expanded scale showing all the peaks) of blanks,
7 A( X' m6 }5 ]0 a$ a+ f4 e+ punstressed, and stressed samples. Additionally, chiral drugs should be analyzed with chiral3 G$ s* c, @  V+ K' x* T" t1 @
methods to establish stereochemical purity and stability (11, 12).
7 M3 p/ O4 r1 b7 {5 Q* mThe analytical method of choice should be sensitive enough to detect impurities at low levels (i.e.,
4 z  A: L6 ]1 X7 U4 |4 q6 |# J0.05% of the analyte of interest or lower), and the peak responses should fall within the range of
" F+ `$ C1 @5 P9 G; p& `detector's linearity. The analytical method should be capable of capturing all the impurities formed
; k% h: C$ {0 lduring a formal stability study at or below ICH threshold limits (13, 14). Degradation product
, I8 L. Z& R# S# V+ }identification and characterization are to be performed based on formal stability results in! C0 W7 v! d$ k, R: F, Z
accordance with ICH requirements. Conventional methods (e.g., column chromatography) or) O8 L, H% }& E& H
hyphenated techniques (e.g., LC–MS, LC–NMR) can be used in the identification and
* t; B1 T) u/ t4 S9 k8 D9 j/ X$ rcharacterization of the degradation products. Use of these techniques can provide better insight# c5 l9 e* V/ D) e3 C
into the structure of the impurities that could add to the knowledge space of potential structural8 G- S$ Y0 C8 L  R. t( T$ L
alerts for genotoxicity and the control of such impurities with tighter limits (12–17). It should be1 J3 t! h+ ~- H( n, U; o& s
noted that structural characterization of degradation products is necessary for those impurities that
, V) q( D% h8 M6 Dare formed during formal shelf-life stability studies and are above the qualification threshold limit
; }2 _" r& y9 r9 E% H(13).* a7 O: l: S% b, S% M  P
Various detection types can be used to analyze stressed samples such as UV and mass! @5 n2 d) `6 g. k
spectroscopy. The detector should contain 3D data capabilities such as diode array detectors or4 Q& W6 q0 D7 `. d- k
mass spectrometers to be able to detect spectral non-homogeneity. Diode array detection also
" C2 [* i( U8 P* G. v( X/ W  soffers the possibility of checking peak profile for multiple wavelengths. The limitation of diode
$ p8 g: v$ g0 I, |' Karray arises when the UV profiles are similar for analyte peak and impurity or degradant peak and# Y+ v  G  M' ~) n6 z, y6 Y3 S1 n
the noise level of the system is high to mask the co-eluting impurities or degradants. Compounds- r* h! Z2 m6 V
of similar molecular weights and functional groups such as diastereoisomers may exhibit similar
1 W+ z+ u) j  B7 m: mUV profiles. In such cases, attempts must be made to modify the chromatographic parameters to  }( X6 T# c: b+ V% S) p7 a* x2 q
achieve necessary separation. An optimal wavelength should be selected to detect and quantitate
  i; P# ~" D8 K% ]+ P; Qall the potential impurities and degradants. Use of more than one wavelength may be necessary, if
3 m+ }* d! c/ y: Rthere is no overlap in the UV profile of an analyte and impurity or degradant peaks. A valuable
9 A8 \* D6 ]$ L1 ^tool in method development is the overlay of separation signals at different wavelengths to
, S# o$ V) u; V, B4 Ydiscover dissimilarities in peak profiles.
- u! J- `# p6 y" `, e) {, W/ gPeak purity analysis.8 l/ J$ {# G; S3 c; l- d/ q2 t) C- R
Peak purity is used as an aid in stability indicating method development. The spectral uniqueness
4 a; v1 O* W# f3 L0 Q7 N7 Z7 Pof a compound is used to establish peak purity when co-eluting compounds are present.
! S. i2 T, s8 ]8 V9 e6 `Peak purity or peak homogeneity of the peaks of interest of unstressed and stressed samples
6 s8 Y7 j6 s2 Xshould be established using spectral information from a diode array detector. When instrument
5 d& }/ G2 b9 m$ `6 c. C. R3 Csoftware is used for the determination of spectral purity of a peak, relevant parameters should be* F  \# B/ e+ W/ A. c: \% R1 v' T
set up in accordance with the manufacturer's guidance. Attention should be given to the peak
: N: g( K2 X( X" B% _+ sheight requirement for establishing spectral purity. UV detection becomes non linear at higher
# x2 x; A* s: l. W8 Qabsorbance values. Thresholds should be set such that co-eluting peaks can be detected. Optimum
) z( V6 j) |. T& z. w4 g, Vlocation of reference spectra should also be selected. The ability of the software to automatically3 r1 J& W# y1 _% P' T
correct spectra for continuously changing solvent background in gradient separations should be
4 h. ]! g/ ]$ J) a; _2 a+ d7 }: Oascertained.2 \5 w7 L: R! O6 s7 Y+ b
Establishing peak purity is not an absolute proof that the peak is pure and that there is no2 r& l4 n0 L4 m$ y* Z* [
co-elution with the peak of interest. Limitations to peak purity arise when co-eluting peaks are
* J8 \+ U8 V0 |7 d+ G. Fspectrally similar, or below the detection limit, or a peak has no chromophore, or when they are: x1 O! X! L5 i( D3 B# P
not resolved at all.
& `4 Q8 D8 l7 s6 w0 M! f8 \Mass balance.  ?& \/ P* _, E/ C  ]0 v
Mass balance establishes adequacy of a stability indicating method though it is not achievable in
2 @( s; b& m" k; b- `! `) R* Jall circumstances. It is performed by adding the assay value and the amounts of impurities and0 j# Q1 Q& V' r! [0 c9 O" x
degradants to evaluate the closeness to 100% of the initial value (unstressed assay value) with due
+ ~9 c2 Z9 e; Z% @9 F- s& K# mconsideration of the margin of analytical error (1).* a/ R# D0 k/ d% B4 w. s& J5 z: X: W# M
Some attempt should be made to establish a mass balance for all stressed samples. Mass
5 E* P6 {0 k2 aimbalance should be explored and an explanation should be provided. Varying responses of7 P: T; h( F; ~5 B0 b, Z
analyte and impurity peaks due to differences in UV absorption should also be examined by the
1 l5 Q! A- V: o# R) c8 I. R6 Buse of external standards. Potential loss of volatile impurities, formation of non-UV absorbing4 D. a2 l! K3 g) e) s+ m( z
compounds, formation of early eluants, and potential retention of compounds in the column; l9 [7 D% ]7 I, i0 s
should be explored. Alternate detection techniques such as RI LC/MS may be employed to
$ [  ?; Y4 h; v8 ?4 G* R* N5 ?$ U2 Qaccount for non-UV absorbing degradants.) Z& E" B+ g- f  M2 @. P
Termination of study7 K6 }. n" J8 @+ ]
Stress testing could be terminated after ensuring adequate exposure to stress conditions. Typical1 k0 D9 I, p, p% S$ E  r
activation energy of drug substance molecules varies from 12–24 kcal/mol (18). A compound may
1 p, _& u6 N: b8 I' z6 ]not necessarily degrade under every single stress condition, and general guideline on exposure
3 F3 B* H: X% F/ ^$ L4 G; i- @limit is cited in a review article (10). In circumstances where some stable drugs do not show any& w5 \3 b  p* m+ o: e
degradation under any of the stress conditions, specificity of an analytical method can be
5 ^/ `+ E) S1 z' ~7 b3 K* n4 [: R2 Lestablished by spiking the drug substance or placebo with known impurities and establishing+ D, u8 h- g- q; h! S
adequate separation.6 w: W: l) |" C
Other considerations
: P5 `7 |# j6 G, j. t9 gStress testing may not be necessary for drug substances and drug products that have
  H- f( a* h% Z% i: Gpharmacopeial methods and are used within the limitations outlined in USP <621>. In the case8 g' h$ k9 F) ^! U9 g6 O5 x
where a generic drug product uses a different polymorphic form from the RLD, the drug substance# f# a) F7 ]1 e0 Q  o
should be subjected to stress testing to evaluate the physiochemical changes of the polymorphic, e5 x; D+ o3 [$ b
form because different polymorphic forms may exhibit different stability characteristics.  j$ R5 k( e& u) l8 i; k7 _
Forced degradation in QbD paradigm! m: n1 v: f4 Q3 S
A systematic process of manufacturing quality drug products that meet the predefined targets for
7 g( k& c( V% ^6 F( O, j' O' {the critical quality attributes (CQA) necessitates the use of knowledge obtained in forced
) X5 h" c2 x) }6 M1 xdegradation studies.4 V3 ~9 s" x9 D. @( B, s
A well-designed, forced degradation study is indispensable for analytical method development in a, _6 i3 w  t. U6 G+ d7 y  r
QbD paradigm. It helps to establish the specificity of a stability indicating method and to predict
' n# V# C3 w7 m. S% U/ i$ hpotential degradation products that could form during formal stability studies. Incorporating all" i) [* o! ^4 i* f
potential impurities in the analytical method and establishing the peak purity of the peaks of$ T* q/ e0 d3 K& Q
interest helps to avoid unnecessary method re-development and revalidation.8 |1 u5 S* F' v. g7 w* Z9 F
Knowledge of chemical behavior of drug substances under various stress conditions can also( k3 a2 `4 I; ?1 e
provide useful information regarding the selection of excipients for formulation development.
9 h* R1 @& @* w  SExcipient compatibility is an integral part of understanding potential formulation interactions
  K* \/ Z7 C3 B; Z& iduring product development and is a key part of product understanding. Degradation products due  H. w9 U; D# d) T- N- ]3 |
to drug-excipient interaction or drug-drug interaction in combination products can be examined by
3 S+ H# m4 G: N: a" f- J& v+ f; m+ rstressing samples of drug substance, drug product, and placebo separately and comparing the
& n. r. p1 p% K: i9 J2 a2 rimpurity profiles. Information obtained regarding drug-related peaks and non-drug-related peaks
, T5 d! B, i/ B4 Qcan be used in the selection and development of more stable formulations. For instance, if a drug7 i6 W6 T7 l1 r1 T
substance is labile to oxidation, addition of an antioxidant may be considered for the formulation.
  d8 g- F* F' g' o+ N4 r# W6 kFor drug substances that are labile to acid or undergo stereochemical conversion in acidic medium,) n) C" X3 L5 c; N) B- K
delayed-release formulations may be necessary. Acid/base hydrolysis testing can also provide
8 f- i$ u- M* Y0 p. ruseful insight in the formulation of drug products that are liquids or suspensions.% A$ W6 r/ S& B/ w0 P( H; w
Knowledge gained in forced degradation studies can facilitate improvements in the manufacturing
' m" {' W& n$ w6 @/ E! Nprocess. If a photostability study shows a drug substance to be photolabile, caution should be9 k+ L& |0 {0 y% F2 M5 J
taken during the manufacturing process of the drug product. Useful information regarding process7 n0 B# a1 W3 l
development (e.g., wet versus dry processing, temperature selection) can be obtained from thermal
  l! ?3 w- _1 j( F$ G9 Wstress testing of drug substance and drug product.5 r+ D9 M1 p4 e: |$ \
Additionally, increased scientific understanding of degradation products and mechanisms may
9 m- L. l9 e, T, b+ @help to determine the factors that could contribute to stability failures such as ambient temperature,2 i  a9 k' L% R. Z5 @
humidity, and light. Appropriate selection of packaging materials can be made to protect against1 i6 d! G5 E( H; v8 x# z# p% O
such factors.
$ ^' g3 L9 N3 i, K6 IConclusion
  S& s3 A0 B* RAn appropriately-designed stress study meshes well with the QbD approaches currently being* \' ^! G. O' i- C, }2 \! F; G: A
promoted in the pharmaceutical industry. A well-designed stress study can provide insight in5 _8 o# y7 D; x5 c! a/ ?$ P9 N
choosing the appropriate formulation for a proposed product prior to intensive formulation
' w7 |' s3 ^& R1 R, c7 Zdevelopment studies. A thorough knowledge of degradation, including mechanistic understanding1 C0 x3 l9 {6 N& l  `# c4 A1 w$ e
of potential degradation pathways, is the basis of a QbD approach for analytical method
$ u4 ^: Y- \! ]; O1 Zdevelopment and is crucial in setting acceptance criteria for shelf-life monitoring. Stress testing
+ v5 W1 e( n$ j* `! A, H# \can provide useful insight into the selection of physical form, stereo-chemical stability of a drug
5 S5 N$ V* m8 csubstance, packaging, and storage conditions. It is important to perform stress testing for generic; s+ `! F$ G6 ~& [
drugs due to allowable qualitative and quantitative differences in formulation with respect to the
4 j6 C2 f1 i. ?. C' h( L; aRLD, selection of manufacturing process, processing parameters, and packaging materials.
% H# y! c: n1 `$ |. _# XAcknowledgments
7 |5 l5 c+ J# H2 i- ]# I- f% O& IThe author would like to thank Bob Iser, Naiqi Ya, Dave Skanchy, Bing Wu, and Ashley Jung for  r3 u" @1 a% Q6 ~
their scientific input and support.
/ d8 @, I( \! X- q! R$ ]6 YRagine Maheswaran, PhD, is a CMC reviewer at the Office of Generic Drugs within the Office of
8 v" K5 _, U/ D% iPharmaceutical Science, under the US Food and Drug Administration's Center for Drug
+ B+ b' o# z/ S- n% Z% uEvaluation and Research, Ragine.Maheswaran@fda.hhs.gov
$ u* w  a: P! q& C# KDisclaimer: The views and opinions in this article are only those of the author and do not
  [6 A( s6 M( }1 Bnecessarily reflect the views or policies of the US Food and Drug Administration.6 ]$ ]% Z; ], M' J0 z) I
References
! w- H3 |- D4 e7 }8 q0 o1. ICH, Q1A(R2) Stability Testing of New Drug Substances and Products (Geneva, Feb. 2003).
  d0 b& c# ?8 V# W1 K# d2. ICH, Q1B Stability Testing: Photostability Testing of New Drug Substances and Products
6 S8 |% _3 V& F/ o3 q; A' _; X5 Y( K; p(Geneva, Nov. 1996).7 d2 b# V( Y" v
3. H. Brittain, Analytical Profiles of Drug Substances and Excipients (Academic Press, London,
& _/ m, W; L- n8 ^. T. s2002).
$ w, a& v( ~& }. d8 Y4. A. Srinivasan and R. Iser, Pharm. Technol. 34 (1), 50–59 (2010).
) ]  o/ c6 u- [* q. x/ w5. A. Srinivasan, R. Iser, and D. Gill, Pharm. Technol. 34(8), 45–51 (2010).
1 l9 M" Z0 a* C$ X9 q6. A. Srinivasan, R. Iser, and D. Gill, Pharm. Technol. 35 (2), 58–67 (2011).
9 K$ c7 ^, G& ?7. S. Klick, et al., Pharm.Technol. 29 (2) 48–66 (2005).
6 c4 r) N5 A3 H. z8. K. M. Alsante, L. Martin and S. W. Baertschi, Pharm.Technol. 27 (2) 60-72 (2003).
" j2 E# ], U: K( n3 l  a8 u8 O9. D. W. Reynolds, et al., Pharm.Technol. 26 (2), 48–56 (2002).
: D. {5 l, R8 N% @7 E6 t0 c10. K. M. Alsante et al., Advanced Drug Delivery Reviews 59, 29–37 (2007).
; P5 R/ R: c* [: \7 L. Z5 |+ i11. FDA, Guidance for Industry on Analytical Procedures and methods Validation Chemistry,
) ?) k: b& ~/ u, D* kManufacturing, and Controls Documentation (draft) (Rockville, MD, Aug. 2000).7 \6 L/ S) I$ ~5 J+ t6 d& {% |
12. ICH, Q6A: Specifications: Test Procedures and Acceptance Criteria for New Drug Substances: _5 f% o' \0 L  }2 I! [
and New Drug Products: Chemical Substances (Geneva, Oct. 1999)., Q1 e: H: G9 S4 h6 p6 i
13. ICH, Q3A(R2) Impurities in New Drug Substances (Geneva, Oct. 2006).
9 F3 z4 p  @8 p) @8 ~14. ICH, Q3B(R2) Impurities in New Drug Products (Geneva, June 2006).- o5 u$ o7 a5 n" K
15. FDA, Guidance for Industry ANDAs: Impurities in Drug Substances (draft), (Rockville, MD,
* v% Y0 p  h$ ^8 q' ]; {: ~' P+ AAug. 2005).9 k: T- \$ ]1 j! w8 A
16. FDA, Guidance for Industry ANDAs: Impurities in Drug Products (draft) (Rockville, MD,
: V( u7 \: K0 ~; ~8 KNov. 2010).
" V( h, c  o+ g( m17. EMA, Guideline on the Limits of Genotoxic Impurities, Committee for Medical Products for
4 ~6 q7 J& B1 VHuman Use (CHMP) (Doc. Ref EMA/CHMP/QWP/251344/2006) (Jan. 1, 2007).+ A, t5 p8 H6 s. ~3 o' ]
18. K. A. Conners et al., Chemical Stability of Pharmaceuticals, Wiley and Sons, New York, New
* ?& ]7 d+ w2 v& C% bYork, 2nd Ed., p. 19 (1986).
回复

使用道具 举报

板凳
一花一世界 发表于 2015-7-22 16:02:21 | 只看该作者
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,
回复 支持 反对

使用道具 举报

您需要登录后才可以回帖 登录 | 立即注册  

本版积分规则

QQ|手机版|药群论坛 ( 蜀ICP备15007902号 )

GMT+8, 2025-8-11 08:30 PM , Processed in 0.104620 second(s), 20 queries .

本论坛拒绝任何人以任何形式在本论坛发表与中华人民共和国法律相抵触的言论! X3.2

© 2011-2014 免责声明:药群网所有内容仅代表发表者个人观点,不代表本论坛立场。

快速回复 返回顶部 返回列表