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
4 [6 d; m5 O2 x1 C* v
FDA Perspectives: Scientific Considerations of Forced Degradation Studies in ANDA5 y6 ~- F6 J, K# h
Submissions
- X7 t- U3 m" |1 J6 S3 X6 K- wThe author outlines the scientific aspects of forced degradation studies that should be considered
& l1 R3 E/ R0 G: Iin relation to ANDA submissions.0 b: T" G+ q. X' @. L: N0 Z
May 2, 2012
3 p' u0 m; p3 k% _0 o+ A' ?By:Ragine Maheswaran
0 ]0 n8 h4 B1 KPharmaceutical Technology
0 R$ C7 m+ u( b6 TVolume 36, Issue 5, pp. 73-80* J9 W) e6 O' ~
Forced degradation is synonymous with stress testing and purposeful degradation. Purposeful
( F, c& k+ @! E' x9 `degradation can be a useful tool to predict the stability of a drug substance or a drug product with2 h0 b/ @+ M: ~: Z' r. Z
effects on purity, potency, and safety. It is imperative to know the impurity profile and behavior of! C8 w; t. `5 S# U* l
a drug substance under various stress conditions. Forced degradation also plays an important role
) i2 t3 L8 n* ^* Xin the development of analytical methods, setting specifications, and design of formulations under9 p# x$ ?% m. c1 R* A
the quality-by-design (QbD) paradigm. The nature of the stress testing depends on the individual4 J% G/ W6 H: R. r3 h8 z
drug substance and the type of drug product (e.g., solid oral dosage, lyophilized powders, and8 Z9 }0 |# ]9 |3 }0 e
liquid formulations) involved (1).
7 j0 O' h) X, T% {) M! ?2 S* DThe International Conference on Harmonization (ICH) Q1B guideline provides guidance for1 N& e/ s6 b' Z% k, r- G1 M
performing photostability stress testing; however, there are no additional stress study% W' R; G0 J) y4 f2 a' q6 Q
recommendations in the ICH stability or validation guidelines (2). There is also limited" d& p$ a( M: g4 P* f0 `
information on the details about the study of oxidation and hydrolysis. The drug substance
/ S, I' }2 j( N, J6 s' I( _monographs of Analytical Profiles of Drug Substances and Excipients provide some information& a6 y) H3 q7 M& l- f+ v
with respect to different stress conditions of various drug substances (3).: m' ^5 T/ [4 B2 P- l* E) l7 d! I: h5 n
The forced degradation information provided in the abbreviated new drug application (ANDA). c, l  w0 K% B9 j# c& h
submissions is often incomplete and in those cases deficiencies are cited. An overview of common
5 o& r; K5 m. ~0 H+ g0 Zdeficiencies cited throughout the chemistry, manufacturing, and controls (CMC) section of the3 s+ {- i- ?+ }7 E2 B9 T# X
ANDAs has been published (4–6). Some examples of commonly cited deficiencies related to
- b: I6 }' B3 n1 K2 C4 ?# Iforced degradation studies include the following:
( Y% |. e7 ~( W* g" V# |Your drug substance does not show any degradation under any of the stress conditions. Please1 F) ^8 |8 r7 H1 e: j/ A( |. f! B
repeat stress studies to obtain adequate degradation. If degradation is not achievable, please; P; v; g( I2 r5 v5 U5 X% Q; p5 W
provide your rationale.( {( U" j; S* \! S+ f
Please note that the conditions employed for stress study are too harsh and that most of your drug
5 q4 m- A" ~: L% }substance has degraded. Please repeat your stress studies using milder conditions or shorter
4 c+ n( B; D: M$ k# v' kexposure time to generate relevant degradation products.
& o9 ]2 {* U+ t- F1 y9 PIt is noted that you have analyzed your stressed samples as per the assay method conditions. For
0 o1 A( q2 [2 N  w& qthe related substances method to be stability indicating, the stressed samples should be analyzed( F$ G1 _( H, s0 R2 d" B- y
using related substances method conditions.% w$ h5 K5 P' @7 m8 G
Please state the attempts you have made to ensure that all the impurities including the degradation' g) ?; Q* }. i& Z4 v! B
products of the unstressed and the stressed samples are captured by your analytical method.
3 v5 n; ?" |& U' q4 d  RPlease provide a list summarizing the amount of degradation products (known and unknown) in
9 ~+ }" e/ }* v. y5 a/ n/ ]9 ]' Wyour stressed samples.; \( t: y- Q! {" X% u
Please verify the peak height requirement of your software for the peak purity determination.( R4 h8 `9 ]" ~0 z
Please explain the mass imbalance of the stressed samples.3 k, a* b& ?$ B3 W1 A8 |$ ~+ ?
Please identify the degradation products that are formed due to drug-excipient interactions.6 p# E, c% P* \
Your photostability study shows that the drug product is very sensitive to light. Please explain how- ^3 N/ Q/ ~$ {& Q! y- f0 i' Y, B
this is reflected in the analytical method, manufacturing process, product handling, etc.
/ J( _' q: Y% N) aIn an attempt to minimize deficiencies in the ANDA submissions, some general recommendations: T' N7 p; a  T# O8 L
to conduct forced degradation studies, to report relevant information in the submission, and to, J0 }: G9 h+ X, G' ?- D
utilize the knowledge of forced degradation in developing stability indicating analytical methods,
8 @5 W3 G, H+ `7 h9 M# t5 Umanufacturing process, product handling, and storage are provided in this article., k# z4 c% w' K( y" t# R
Stress conditions& [2 ]# ]. [' V3 {" N
Typical stress tests include four main degradation mechanisms: heat, hydrolytic, oxidative, and( d. M8 |' g$ q# |4 k  m
photolytic degradation. Selecting suitable reagents such as the concentration of acid, base, or
; ]; a2 x0 {0 }6 ]+ M; @$ O$ a; Goxidizing agent and varying the conditions (e.g., temperature) and length of exposure can achieve
( e" y1 h: v" p! Q/ L: v+ V: ^0 m. i" |the preferred level of degradation. Over-stressing a sample may lead to the formation of secondary% A5 g2 B6 ^. M3 o, Z
degradants that would not be seen in formal shelf-life stability studies and under-stressing may not" H. g2 }; b8 h0 p8 N8 E$ L' d
serve the purpose of stress testing. Therefore, it is necessary to control the degradation to a desired
$ ?1 M! k6 T; \; `+ ?$ X4 W  ?0 Nlevel. A generic approach for stress testing has been proposed to achieve purposeful degradation6 v* T1 v6 f; l$ x
that is predictive of long-term and accelerated storage conditions (7). The generally recommended
# N4 Z+ l$ e2 [$ pdegradation varies between 5-20% degradation (7–10). This range covers the generally
, @' h8 [* V* A2 Y( Dpermissible 10% degradation for small molecule pharmaceutical drug products, for which the
! |. d. s+ F  Q- Mstability limit is 90%-110% of the label claim. Although there are references in the literature that" e0 e. c  e' e% C, o) q# d' F
mention a wider recommended range (e.g., 10-30%), the more extreme stress conditions often* }) b! x, l4 M1 Y* {& |% [) P: ~
provide data that are confounded with secondary degradation products.
6 k, O' C0 E9 V9 G$ ]/ ^5 x) l! P% fPhotostability.
; w- y9 i  `8 s* s: @Photostability testing should be an integral part of stress testing, especially for photo-labile
; a% O) t- T% }. lcompounds. Some recommended conditions for photostability testing are described in ICH Q1B
+ [9 z( z1 c1 L+ C, C! n$ Q: ^6 LPhotostability Testing of New Drug Substances and Products (2). Samples of drug substance, and+ c/ f: s1 ^1 n* j% w4 z' h& A& i% s
solid/liquid drug product, should be exposed to a minimum of 1.2 million lux hours and 200 watt( B% F2 r+ N  z9 l
hours per square meter light. The same samples should be exposed to both white and UV light. To
3 S* Z* W6 s0 K, @minimize the effect of temperature changes during exposure, temperature control may be
# C& A% R3 [( z% ]" _necessary. The light-exposed samples should be analyzed for any changes in physical properties
' d# i6 ^  d# d9 Gsuch as appearance, clarity, color of solution, and for assay and degradants. The decision tree
0 r: \6 t0 d2 B) j7 d3 O9 ioutlined in the ICH Q1B can be used to determine the photo stability testing conditions for drug9 c: @# g5 U" E  Z, {* b9 {
products. The product labeling should reflect the appropriate storage conditions. It is also
) U  q& e9 \+ ~$ d6 k5 [important to note that the labeling for generic drug products should be concordant with that of the
+ f" v5 s! h/ T: Zreference listed drug (RLD) and with United States Pharmacopeia (USP) monograph
3 i7 r! g! n. X/ R, }) Urecommendations, as applicable.6 L/ v* Q6 f. j& Q7 i. w+ T
Heat.
# R" c( a1 s  VThermal stress testing (e.g., dry heat and wet heat) should be more strenuous than recommended) z5 k) D; y/ z+ _3 I
ICH Q1A accelerated testing conditions. Samples of solid-state drug substances and drug products9 W  d4 Y' S7 y
should be exposed to dry and wet heat, whereas liquid drug products can be exposed to dry heat. It
( c5 {, _" P  R( R- [is recommended that the effect of temperature be studied in 10 °C increments above that for6 Y6 b( B. T+ x; c* ]
routine accelerated testing, and humidity at 75% relative humidity or greater (1). Studies may be
) x- w/ N6 L' d, s4 }conducted at higher temperatures for a shorter period (10). Testing at multiple time points could
7 Z' Q. D% j( fprovide information on the rate of degradation and primary and secondary degradation products.
: U# ~' [0 Y3 w* kIn the event that the stress conditions produce little or no degradation due to the stability of a drug  O9 v7 A7 i6 p8 M& B
molecule, one should ensure that the stress applied is in excess of the energy applied by
- r2 {. e* R8 q; P/ Raccelerated conditions (40 °C for 6 months) before terminating the stress study.4 I1 ^' j+ \* n2 E, F8 X
Acid and base hydrolysis.$ ~: g. }8 @% L2 W, ^. S
Acid and base hydrolytic stress testing can be carried out for drug substances and drug products in/ W2 `$ F3 d( k9 t5 H% Q2 s
solution at ambient temperature or at elevated temperatures. The selection of the type and4 Q' F, F6 b1 v4 Z
concentrations of an acid or a base depends on the stability of the drug substance. A strategy for( n! G0 N7 r4 o  p' D
generating relevant stressed samples for hydrolysis is stated as subjecting the drug substance
4 w4 @. A, j+ [, {, _solution to various pHs (e.g., 2, 7, 10–12) at room temperature for two weeks or up to a maximum
9 I6 F+ E+ n. q, N; V8 Mof 15% degradation (7). Hydrochloric acid or sulfuric acid (0.1 M to 1 M) for acid hydrolysis and: A7 G) v. V& K) V* y4 y
sodium hydroxide or potassium hydroxide (0.1 M to 1 M) for base hydrolysis are suggested as8 m' ?; {; _8 I) j! B
suitable reagents for hydrolysis (10). For lipophilic drugs, inert co-solvents may be used to( ^* F2 Y. t; ?7 P2 k
solubilize the drug substance. Attention should be given to the functional groups present in the( r# I9 U! w* P# Y" _0 c: O
drug molecule when selecting a co-solvent. Prior knowledge of a compound can be useful in
. w% ?/ D, ]- c3 Eselecting the stress conditions. For instance, if a compound contains ester functionality and is very
1 L& x  G/ \4 Q: M) `5 elabile to base hydrolysis, low concentrations of a base can be used. Analysis of samples at various4 \+ ?8 o: o# i/ Q
intervals can provide information on the progress of degradation and help to distinguish primary
  L. O# H5 h3 Y9 j6 [9 l0 C: J! Cdegradants from secondary degradants.
/ H4 v1 z# |* p& {& g4 ~Oxidation.
, Q* K% p+ R' t" ~( I. fOxidative degradation can be complex. Although hydrogen peroxide is used predominantly
  a1 ~* |! z  }9 j$ q* Bbecause it mimics possible presence of peroxides in excipients, other oxidizing agents such as
9 Z. I5 Z& t. U& n8 Y3 J  S6 `metal ions, oxygen, and radical initiators (e.g., azobisisobutyronitrile, AIBN) can also be used.1 b. }, L( _7 [. B1 A
Selection of an oxidizing agent, its concentration, and conditions depends on the drug substance.1 F5 ]4 _+ r0 e6 J
Solutions of drug substances and solid/liquid drug products can be subjected to oxidative  o+ N" U3 M+ O: u$ Q2 k  e
degradation. It is reported that subjecting the solutions to 0.1%-3% hydrogen peroxide at neutral! h9 p+ \! [5 z6 M  v4 d
pH and room temperature for seven days or up to a maximum 20% degradation could potentially
- g0 g$ i. p) L' G. [generate relevant degradation products (10). Samples can be analyzed at different time intervals to
! a8 L. ^) R$ {; Vdetermine the desired level of degradation.8 O  L- J3 J, E
Different stress conditions may generate the same or different degradants. The type and extent of
( z3 Z3 Q, [- n1 Cdegradation depend on the functional groups of the drug molecule and the stress conditions.9 I9 S# Q- o! U0 z3 |' \
Analysis method6 x9 L) n  ^% C; Q  N4 u# I
The preferred method of analysis for a stability indicating assay is reverse-phase4 t) w  v& C: h! P1 a  A# K
high-performance liquid chromatography (HPLC). Reverse-phase HPLC is preferred for several
( J: @3 D, P  a8 |) D8 hreasons, such as its compatibility with aqueous and organic solutions, high precision, sensitivity,
/ w/ x) i8 j6 @3 Z1 V' l' `and ability to detect polar compounds. Separation of peaks can be carried out by selecting
# V2 x7 y9 o0 e" J$ ~8 Y3 e  Sappropriate column type, column temperature, and making adjustment to mobile phase pH.! e% H1 Y6 \0 Q; v7 A8 {5 t" k" @
Poorly-retained, highly polar impurities should be resolved from the solvent front. As part of
5 L9 N# R# [% q8 pmethod development, a gradient elution method with varying mobile phase composition (very low- m2 e0 c3 j5 u: _
organic composition to high organic composition) may be carried out to capture early eluting
/ s& U' |3 M' Q* W3 ^" ghighly polar compounds and highly retained nonpolar compounds. Stressed samples can also be& L7 s# h) O& `3 z- @; Y# r9 g# q+ c
screened with the gradient method to assess potential elution pattern. Sample solvent and mobile
6 O; e' L1 e* u  p, c& [phase should be selected to afford compatibility with the drug substance, potential impurities, and# R9 i6 Q! z) r
degradants. Stress sample preparation should mimic the sample preparation outlined in the6 U% M/ x& j5 n+ k& O% Z
analytical procedure as closely as possible. Neutralization or dilution of samples may be necessary
! [4 f1 A$ [" Tfor acid and base hydrolyzed samples. Chromatographic profiles of stressed samples should be: U) x" [- E6 k
compared to those of relevant blanks (containing no active) and unstressed samples to determine! `/ O; W# d$ \5 ^1 s4 l- |! _
the origin of peaks. The blank peaks should be excluded from calculations. The amount of' p4 \" @5 T# F7 t3 g
impurities (known and unknown) obtained under each stress condition should be provided along
, _( Y2 w0 C3 D5 u7 iwith the chromatograms (full scale and expanded scale showing all the peaks) of blanks,- o% w0 r( P; Y' C
unstressed, and stressed samples. Additionally, chiral drugs should be analyzed with chiral# C* E% l5 d. j# J. M
methods to establish stereochemical purity and stability (11, 12).
. ?# m% H. }/ }The analytical method of choice should be sensitive enough to detect impurities at low levels (i.e.,
: b4 ^" Y1 Z2 |7 L( }; u6 K( f0.05% of the analyte of interest or lower), and the peak responses should fall within the range of
, [4 Z5 |" r5 u- U8 q* Pdetector's linearity. The analytical method should be capable of capturing all the impurities formed) \5 o9 {5 S) f4 H* S2 M! b
during a formal stability study at or below ICH threshold limits (13, 14). Degradation product
) B: k: w: h% w6 x! U& C: F- D8 Fidentification and characterization are to be performed based on formal stability results in
7 J8 F0 u1 Q( \" X+ D& H7 k- paccordance with ICH requirements. Conventional methods (e.g., column chromatography) or# ~+ f+ @% \3 ^
hyphenated techniques (e.g., LC–MS, LC–NMR) can be used in the identification and& o1 V$ H4 s' S6 G; y7 r& B
characterization of the degradation products. Use of these techniques can provide better insight* h2 J9 C0 I( H6 f& L$ E+ N
into the structure of the impurities that could add to the knowledge space of potential structural
" |6 b2 G- f" Z) J1 |alerts for genotoxicity and the control of such impurities with tighter limits (12–17). It should be8 b" L1 G+ T. _* z
noted that structural characterization of degradation products is necessary for those impurities that& L) r$ X$ C) u9 @  H
are formed during formal shelf-life stability studies and are above the qualification threshold limit( U) H) h4 p. J8 Y0 V/ A
(13).' c( T4 M2 m* r$ _
Various detection types can be used to analyze stressed samples such as UV and mass
2 A! Z/ x: t0 B" I6 I+ h; L+ \spectroscopy. The detector should contain 3D data capabilities such as diode array detectors or
: }( o: `6 k, D6 n& l* [mass spectrometers to be able to detect spectral non-homogeneity. Diode array detection also
8 e" y5 L: D8 P5 d$ q7 Woffers the possibility of checking peak profile for multiple wavelengths. The limitation of diode3 E7 n9 D2 M/ U) s. x- Q
array arises when the UV profiles are similar for analyte peak and impurity or degradant peak and
+ s; a- R3 Z7 S; Q0 Sthe noise level of the system is high to mask the co-eluting impurities or degradants. Compounds" B, s5 C3 D6 n6 b9 u
of similar molecular weights and functional groups such as diastereoisomers may exhibit similar
3 u* B- {+ H: l4 Q- ^UV profiles. In such cases, attempts must be made to modify the chromatographic parameters to
* F9 d! n: W  d( \, f8 x- ]achieve necessary separation. An optimal wavelength should be selected to detect and quantitate
& |6 x* @& W/ Z4 M/ p2 jall the potential impurities and degradants. Use of more than one wavelength may be necessary, if7 J4 ^* b/ x0 h0 J8 Q
there is no overlap in the UV profile of an analyte and impurity or degradant peaks. A valuable. b! @3 F7 H0 p: ^
tool in method development is the overlay of separation signals at different wavelengths to
6 g3 C9 O0 X; D1 t9 Vdiscover dissimilarities in peak profiles.
3 M/ [' l! x0 Z' D7 V. {& v$ p/ d9 _Peak purity analysis.
  R: u8 ^) B3 V) d' s  SPeak purity is used as an aid in stability indicating method development. The spectral uniqueness) ^6 l5 @; X3 N5 ?2 o/ _5 K3 P
of a compound is used to establish peak purity when co-eluting compounds are present.
  D9 e. L: u( l, _8 c& [Peak purity or peak homogeneity of the peaks of interest of unstressed and stressed samples$ u1 q+ z$ S4 O2 M/ I  ^
should be established using spectral information from a diode array detector. When instrument
/ i4 G' a% E! q# m/ D4 I/ j) zsoftware is used for the determination of spectral purity of a peak, relevant parameters should be7 Y1 K0 f9 F# K
set up in accordance with the manufacturer's guidance. Attention should be given to the peak
! G& N. t: d( q$ S# E" j; d; G  Vheight requirement for establishing spectral purity. UV detection becomes non linear at higher
9 |; i9 |1 @9 S7 m/ dabsorbance values. Thresholds should be set such that co-eluting peaks can be detected. Optimum
/ s7 @/ @$ S2 L7 w6 Clocation of reference spectra should also be selected. The ability of the software to automatically$ q, a4 w: r0 Y" y& S) j
correct spectra for continuously changing solvent background in gradient separations should be
8 x0 {* n9 E* `( v& @, X1 Iascertained.8 }$ s" }5 n; i9 {: u4 y5 y
Establishing peak purity is not an absolute proof that the peak is pure and that there is no
4 s. B: E2 s( A7 ^2 `co-elution with the peak of interest. Limitations to peak purity arise when co-eluting peaks are- a9 ?" T* m% p- ^
spectrally similar, or below the detection limit, or a peak has no chromophore, or when they are
' K8 D8 c/ ?  b( Hnot resolved at all.% H5 a7 r( F& r" T
Mass balance.1 t3 l8 W2 k& p" z- {
Mass balance establishes adequacy of a stability indicating method though it is not achievable in$ _9 L( ?1 A5 w" b8 r) ^
all circumstances. It is performed by adding the assay value and the amounts of impurities and
3 X0 B, A9 o$ a, D* ndegradants to evaluate the closeness to 100% of the initial value (unstressed assay value) with due
8 D" Q  l( B; c* Econsideration of the margin of analytical error (1).4 w0 _7 o8 ^. t9 s* a
Some attempt should be made to establish a mass balance for all stressed samples. Mass
' l1 i8 H' v2 ?/ @7 D8 ]imbalance should be explored and an explanation should be provided. Varying responses of  ]) R& Q# Q# y: C
analyte and impurity peaks due to differences in UV absorption should also be examined by the7 ^# _, y' k3 \! u
use of external standards. Potential loss of volatile impurities, formation of non-UV absorbing0 M: T5 S8 j8 C1 a
compounds, formation of early eluants, and potential retention of compounds in the column
  d2 X7 H2 ?! }7 @6 p9 ?should be explored. Alternate detection techniques such as RI LC/MS may be employed to4 _; d- A5 n& W* A! P- z: }( ]. d, W
account for non-UV absorbing degradants.
0 g2 P4 s" ~; {0 f/ _Termination of study7 }0 J1 k  I' V" E5 j
Stress testing could be terminated after ensuring adequate exposure to stress conditions. Typical8 o) o0 M4 A5 {  q* ]1 ~8 e
activation energy of drug substance molecules varies from 12–24 kcal/mol (18). A compound may
" i( g; p, ^9 L  Y9 Enot necessarily degrade under every single stress condition, and general guideline on exposure+ B( u9 H+ C6 D( z& N
limit is cited in a review article (10). In circumstances where some stable drugs do not show any  O9 O4 ~9 a* e, v
degradation under any of the stress conditions, specificity of an analytical method can be
& u) R6 j3 W- U! Hestablished by spiking the drug substance or placebo with known impurities and establishing1 |- P3 s( ~. `$ S8 X0 [; E$ R- k
adequate separation.
: O- i: |* c" x0 y( dOther considerations) p: b" e# m* R; E3 z
Stress testing may not be necessary for drug substances and drug products that have% a9 l2 X; f" }% F8 @
pharmacopeial methods and are used within the limitations outlined in USP <621>. In the case0 j1 u7 ~. ~( e+ O$ Q- y
where a generic drug product uses a different polymorphic form from the RLD, the drug substance
" F) a5 V8 Z2 {5 dshould be subjected to stress testing to evaluate the physiochemical changes of the polymorphic
6 s' N% |# D0 x! L; F  sform because different polymorphic forms may exhibit different stability characteristics.
2 C- h- Q6 a2 PForced degradation in QbD paradigm2 m8 d/ ~1 W) g3 R# V8 g" O
A systematic process of manufacturing quality drug products that meet the predefined targets for
# P5 B7 Y0 J& M! wthe critical quality attributes (CQA) necessitates the use of knowledge obtained in forced. {: Y4 a# |. i8 |
degradation studies.
' b! i+ J3 [3 g* \, e6 M. q5 fA well-designed, forced degradation study is indispensable for analytical method development in a
$ M2 {7 v' L. uQbD paradigm. It helps to establish the specificity of a stability indicating method and to predict
  i' ~5 e. a, Apotential degradation products that could form during formal stability studies. Incorporating all9 Z% y2 G; |- ^
potential impurities in the analytical method and establishing the peak purity of the peaks of
5 T+ x. c3 D3 @* D4 _" }$ uinterest helps to avoid unnecessary method re-development and revalidation.
) Y5 X  n' o9 Y* h+ rKnowledge of chemical behavior of drug substances under various stress conditions can also
, _% Z& I$ V. pprovide useful information regarding the selection of excipients for formulation development.
5 j: |# l0 G4 EExcipient compatibility is an integral part of understanding potential formulation interactions
9 ]6 s- r' o( c0 `! e; `5 Jduring product development and is a key part of product understanding. Degradation products due& ?0 O9 H! s6 X% t4 p& t- `
to drug-excipient interaction or drug-drug interaction in combination products can be examined by
: E. C' d, V  Q3 U- Sstressing samples of drug substance, drug product, and placebo separately and comparing the
* d7 @6 ?/ E$ _3 |impurity profiles. Information obtained regarding drug-related peaks and non-drug-related peaks5 }3 n6 V: _( ^% {6 i
can be used in the selection and development of more stable formulations. For instance, if a drug
/ K! d* R! p" d* \9 Zsubstance is labile to oxidation, addition of an antioxidant may be considered for the formulation.
8 W8 s3 m; h) oFor drug substances that are labile to acid or undergo stereochemical conversion in acidic medium,9 Z9 G7 q: u6 g' j9 E3 f
delayed-release formulations may be necessary. Acid/base hydrolysis testing can also provide
% P/ \  Z& j- R# l* |: _useful insight in the formulation of drug products that are liquids or suspensions.
7 {7 P% O5 ?4 jKnowledge gained in forced degradation studies can facilitate improvements in the manufacturing9 [) T4 p  U0 F0 U( ~
process. If a photostability study shows a drug substance to be photolabile, caution should be, A+ v  Z0 K8 a. G4 _- Q! D0 S
taken during the manufacturing process of the drug product. Useful information regarding process
; j1 q; a/ ]1 Z! F" e- S! f6 Edevelopment (e.g., wet versus dry processing, temperature selection) can be obtained from thermal
8 a4 \$ t" v+ H$ k$ ?( \0 Nstress testing of drug substance and drug product.
% g2 C/ O: V5 {+ m- ?( cAdditionally, increased scientific understanding of degradation products and mechanisms may
( G. @3 M% Y, Xhelp to determine the factors that could contribute to stability failures such as ambient temperature,
/ J/ f# Z2 D. V' [+ r* t; \+ Hhumidity, and light. Appropriate selection of packaging materials can be made to protect against) c: d* n8 y2 B  X' s
such factors.
. I4 s+ H2 f6 k5 e2 fConclusion" T% z: i! h6 f& F4 {7 @
An appropriately-designed stress study meshes well with the QbD approaches currently being, Q, D; [3 @3 d- x$ l
promoted in the pharmaceutical industry. A well-designed stress study can provide insight in. F8 o5 z1 ^; u  x' P  n8 C: r
choosing the appropriate formulation for a proposed product prior to intensive formulation- m3 E: R6 I- _0 q/ n
development studies. A thorough knowledge of degradation, including mechanistic understanding
! b$ r2 F# t7 _2 iof potential degradation pathways, is the basis of a QbD approach for analytical method
  q8 ?+ ?  A7 ^' Jdevelopment and is crucial in setting acceptance criteria for shelf-life monitoring. Stress testing
6 f8 R$ Q3 P- |4 o7 O) q: fcan provide useful insight into the selection of physical form, stereo-chemical stability of a drug% W. V' `$ H0 M6 ]4 K  V
substance, packaging, and storage conditions. It is important to perform stress testing for generic
- P- h; i( _6 f! hdrugs due to allowable qualitative and quantitative differences in formulation with respect to the
' ^: i1 t$ h3 }' I! F8 y$ SRLD, selection of manufacturing process, processing parameters, and packaging materials.. i6 M# w% p- V( l% N
Acknowledgments5 C2 n9 u0 |2 ^4 N: I6 T
The author would like to thank Bob Iser, Naiqi Ya, Dave Skanchy, Bing Wu, and Ashley Jung for6 T/ E1 g; d5 E' _
their scientific input and support.- k5 _, C1 K+ v- B, C( t4 o) |, \
Ragine Maheswaran, PhD, is a CMC reviewer at the Office of Generic Drugs within the Office of! o' h$ i: C6 k
Pharmaceutical Science, under the US Food and Drug Administration's Center for Drug
1 h  B. [8 M) @% n, YEvaluation and Research, Ragine.Maheswaran@fda.hhs.gov
% Z% B" q5 i5 J/ T0 `Disclaimer: The views and opinions in this article are only those of the author and do not
/ m- a* ^, x0 d  C- a( Snecessarily reflect the views or policies of the US Food and Drug Administration.
+ _, j5 Q- I* X9 }9 g6 qReferences- y# Q3 m* R. b
1. ICH, Q1A(R2) Stability Testing of New Drug Substances and Products (Geneva, Feb. 2003).2 B9 f0 M4 P' o0 @2 o( Z0 A; p. D5 T" q
2. ICH, Q1B Stability Testing: Photostability Testing of New Drug Substances and Products+ s6 w% O" D8 D2 w( @
(Geneva, Nov. 1996).
# P" d0 _; ^# `5 w2 f3. H. Brittain, Analytical Profiles of Drug Substances and Excipients (Academic Press, London,
! f# C* T4 c) v# {5 w5 l7 `2002).
! F! C. j' m! p1 I) K& v! d$ D) F4. A. Srinivasan and R. Iser, Pharm. Technol. 34 (1), 50–59 (2010).# R6 F$ M/ ~; [  x; Q
5. A. Srinivasan, R. Iser, and D. Gill, Pharm. Technol. 34(8), 45–51 (2010).
0 x) b$ d9 Q3 `% x- i1 A9 k6. A. Srinivasan, R. Iser, and D. Gill, Pharm. Technol. 35 (2), 58–67 (2011).4 M- z" W' k& v) ]
7. S. Klick, et al., Pharm.Technol. 29 (2) 48–66 (2005).) j3 q! ]3 m! Z" W) m( k; L) C
8. K. M. Alsante, L. Martin and S. W. Baertschi, Pharm.Technol. 27 (2) 60-72 (2003).& V; `( O, ~  @3 n4 c
9. D. W. Reynolds, et al., Pharm.Technol. 26 (2), 48–56 (2002).
% K8 m% F8 J4 Q! N: A9 S) |10. K. M. Alsante et al., Advanced Drug Delivery Reviews 59, 29–37 (2007).
' v4 C. I3 e3 ~2 t9 }9 g. i11. FDA, Guidance for Industry on Analytical Procedures and methods Validation Chemistry,
, e: }7 y; w$ T7 VManufacturing, and Controls Documentation (draft) (Rockville, MD, Aug. 2000).. V9 G! ^; Y8 _6 C! S- T
12. ICH, Q6A: Specifications: Test Procedures and Acceptance Criteria for New Drug Substances  C- s8 T$ k8 A* [; h
and New Drug Products: Chemical Substances (Geneva, Oct. 1999).
8 J0 ?: I7 X6 M4 g9 u13. ICH, Q3A(R2) Impurities in New Drug Substances (Geneva, Oct. 2006).
# P6 X0 U4 n0 g+ M14. ICH, Q3B(R2) Impurities in New Drug Products (Geneva, June 2006).
; N8 B4 Z0 k/ Q0 ^/ d, \$ T' B15. FDA, Guidance for Industry ANDAs: Impurities in Drug Substances (draft), (Rockville, MD,
  V. a: C1 v! b4 v6 a9 W+ A* a6 fAug. 2005)." @! g, o3 e3 J" q  @
16. FDA, Guidance for Industry ANDAs: Impurities in Drug Products (draft) (Rockville, MD,
$ l- T6 ~# v9 S, \# y4 xNov. 2010).
5 I8 |, U  ]5 ^1 U- a17. EMA, Guideline on the Limits of Genotoxic Impurities, Committee for Medical Products for
0 I3 {$ i0 \7 T2 I, R" OHuman Use (CHMP) (Doc. Ref EMA/CHMP/QWP/251344/2006) (Jan. 1, 2007).3 C3 ^, _; i( |* r& X1 e
18. K. A. Conners et al., Chemical Stability of Pharmaceuticals, Wiley and Sons, New York, New
4 x  u6 F" l5 cYork, 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://www.yaoqun.net/) Powered by Discuz! X3.2