药群论坛

 找回密码
 立即注册

只需一步,快速开始

查看: 1052|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
: s( V& h" u# B- t
FDA Perspectives: Scientific Considerations of Forced Degradation Studies in ANDA
2 b* B# L. ]% s' n' ?* kSubmissions
7 ^- [  ~: U- Y1 K% LThe author outlines the scientific aspects of forced degradation studies that should be considered
* Q- R$ J' c- I- ^/ lin relation to ANDA submissions.
. r! T' T3 Y8 A+ M" uMay 2, 2012# p: |  B$ t5 U
By:Ragine Maheswaran
4 g/ I; d- H4 \3 d3 o- {1 m( g) MPharmaceutical Technology& F3 N1 C2 |- |/ b8 l% E
Volume 36, Issue 5, pp. 73-806 D' P8 w3 H" ^* H8 |* e0 i1 x6 }
Forced degradation is synonymous with stress testing and purposeful degradation. Purposeful* Q  `' d& ^8 x% O" H+ y5 `
degradation can be a useful tool to predict the stability of a drug substance or a drug product with8 Z% M6 V& S9 R
effects on purity, potency, and safety. It is imperative to know the impurity profile and behavior of+ g, P- c2 z0 H9 x; f9 N" a
a drug substance under various stress conditions. Forced degradation also plays an important role
: e6 W; I2 N# y1 `8 }2 O! [3 \" win the development of analytical methods, setting specifications, and design of formulations under
' Y7 e: {/ Y/ V/ q3 u  p  pthe quality-by-design (QbD) paradigm. The nature of the stress testing depends on the individual: D: G6 y# V, Z2 \) m! B/ p, Y" c& F
drug substance and the type of drug product (e.g., solid oral dosage, lyophilized powders, and
0 Y5 c: M: I. @! y* y! nliquid formulations) involved (1).
1 h% M' R, m3 r& B) Q6 [6 q2 a1 Y1 t& AThe International Conference on Harmonization (ICH) Q1B guideline provides guidance for4 n) P1 N7 f% E! N
performing photostability stress testing; however, there are no additional stress study3 W/ p/ v# @6 A6 K- c
recommendations in the ICH stability or validation guidelines (2). There is also limited4 v- `1 w0 \" i- ^' z* ?& o
information on the details about the study of oxidation and hydrolysis. The drug substance! L1 x+ P, u; {$ s4 p% e; R
monographs of Analytical Profiles of Drug Substances and Excipients provide some information& B6 }9 z7 R6 L, @) y
with respect to different stress conditions of various drug substances (3).+ X* D" K* O3 D( O
The forced degradation information provided in the abbreviated new drug application (ANDA)
" k* S) O" F* }( N$ C3 ^) xsubmissions is often incomplete and in those cases deficiencies are cited. An overview of common
- c1 g; z" j2 R. K  z1 \' z6 @; _deficiencies cited throughout the chemistry, manufacturing, and controls (CMC) section of the
9 t' m* G$ W4 V' z) qANDAs has been published (4–6). Some examples of commonly cited deficiencies related to& i3 Z- C# r. X) a' N
forced degradation studies include the following:
! a( S5 r/ Y. _1 h% @Your drug substance does not show any degradation under any of the stress conditions. Please
( \& N. _' D/ zrepeat stress studies to obtain adequate degradation. If degradation is not achievable, please
' `1 G; _/ H2 v, h/ ^provide your rationale.. [- \8 f4 }; o+ l$ [: y1 D
Please note that the conditions employed for stress study are too harsh and that most of your drug
( D. M# E4 Q8 G9 H5 w# s+ Fsubstance has degraded. Please repeat your stress studies using milder conditions or shorter: G( r; [$ e& N/ j$ A) h: L
exposure time to generate relevant degradation products.
8 T/ `% \- x. k/ }+ s2 Q) IIt is noted that you have analyzed your stressed samples as per the assay method conditions. For/ X! }" v0 Z( L, |9 ^
the related substances method to be stability indicating, the stressed samples should be analyzed
( U  f+ v  t5 z- p  f+ H: \2 Kusing related substances method conditions.
# X9 p) }, ~3 E# @8 A% ~: zPlease state the attempts you have made to ensure that all the impurities including the degradation' K! G% M" `* c$ F; p
products of the unstressed and the stressed samples are captured by your analytical method.) L7 {" h9 G- n
Please provide a list summarizing the amount of degradation products (known and unknown) in
& d$ k. u( q1 zyour stressed samples.
/ M& P8 k9 D7 wPlease verify the peak height requirement of your software for the peak purity determination.' ^! y( T" d: `) x, M
Please explain the mass imbalance of the stressed samples.+ q4 V! Y' Q5 ~4 h. B1 k4 e9 A9 s
Please identify the degradation products that are formed due to drug-excipient interactions.& E& p# E  S; L: V
Your photostability study shows that the drug product is very sensitive to light. Please explain how
0 v2 e+ n( p4 jthis is reflected in the analytical method, manufacturing process, product handling, etc.
9 _# _9 T  n; ?  m( VIn an attempt to minimize deficiencies in the ANDA submissions, some general recommendations; {$ X3 }5 M6 h, F) z/ W
to conduct forced degradation studies, to report relevant information in the submission, and to* |1 s) d$ O. h6 k
utilize the knowledge of forced degradation in developing stability indicating analytical methods,
! S4 k  I, {# ?manufacturing process, product handling, and storage are provided in this article.
2 U. l2 W, z5 d1 ?5 |Stress conditions
6 q, v% H$ d8 dTypical stress tests include four main degradation mechanisms: heat, hydrolytic, oxidative, and
# ^1 S  A# H2 y9 ~4 Tphotolytic degradation. Selecting suitable reagents such as the concentration of acid, base, or
0 d' `0 o" C2 D1 ^oxidizing agent and varying the conditions (e.g., temperature) and length of exposure can achieve5 \* m; d2 z" f( G- ]% ^
the preferred level of degradation. Over-stressing a sample may lead to the formation of secondary
3 N+ a- D$ o% t1 \$ G1 e* w6 }degradants that would not be seen in formal shelf-life stability studies and under-stressing may not; c$ a+ B, f, X$ z
serve the purpose of stress testing. Therefore, it is necessary to control the degradation to a desired
0 `% }6 T/ T5 H% _$ H- s1 Hlevel. A generic approach for stress testing has been proposed to achieve purposeful degradation
7 z; T) J& z+ u$ E" y" V- pthat is predictive of long-term and accelerated storage conditions (7). The generally recommended2 E* O$ a, ~) g0 U) x
degradation varies between 5-20% degradation (7–10). This range covers the generally' T4 f# L" t  H
permissible 10% degradation for small molecule pharmaceutical drug products, for which the- ?/ b2 W/ F3 Q
stability limit is 90%-110% of the label claim. Although there are references in the literature that1 M1 `+ }6 P- f. x
mention a wider recommended range (e.g., 10-30%), the more extreme stress conditions often
% S0 @3 @0 [' k2 v- ]provide data that are confounded with secondary degradation products.5 @" N& C# {% J( }0 q2 C
Photostability.
  g# b! y# a* S. P' v. l5 r$ RPhotostability testing should be an integral part of stress testing, especially for photo-labile
" s) @/ u- Q2 @$ qcompounds. Some recommended conditions for photostability testing are described in ICH Q1B/ L1 J# Q9 {7 f
Photostability Testing of New Drug Substances and Products (2). Samples of drug substance, and7 t* C" `$ o  \5 _+ e8 b
solid/liquid drug product, should be exposed to a minimum of 1.2 million lux hours and 200 watt
5 S2 o. W& o& e) a# S6 x, Ohours per square meter light. The same samples should be exposed to both white and UV light. To: ^: p& [# @7 q5 o
minimize the effect of temperature changes during exposure, temperature control may be
5 X! B$ c: {- R, Pnecessary. The light-exposed samples should be analyzed for any changes in physical properties
' e( Q( f8 E5 nsuch as appearance, clarity, color of solution, and for assay and degradants. The decision tree
$ @5 S( j- ]2 `outlined in the ICH Q1B can be used to determine the photo stability testing conditions for drug' O$ ~, p" L. ^, C
products. The product labeling should reflect the appropriate storage conditions. It is also& f2 v# {1 B' j5 v, L9 `
important to note that the labeling for generic drug products should be concordant with that of the
, ^3 [+ {4 y& k# ureference listed drug (RLD) and with United States Pharmacopeia (USP) monograph
7 R4 ]4 S4 A$ Orecommendations, as applicable.
' O+ ~+ y! ~! L8 r9 q, gHeat.: S- _9 H; j9 E8 g
Thermal stress testing (e.g., dry heat and wet heat) should be more strenuous than recommended
/ n2 ]. F& H  f% Q9 dICH Q1A accelerated testing conditions. Samples of solid-state drug substances and drug products
( H/ i, Q  }0 i( Pshould be exposed to dry and wet heat, whereas liquid drug products can be exposed to dry heat. It
) {' @! ?0 q( p' f% A2 bis recommended that the effect of temperature be studied in 10 °C increments above that for* l# z$ m. c' H8 m) I! n
routine accelerated testing, and humidity at 75% relative humidity or greater (1). Studies may be
% U( s5 G' H; f5 X( `+ [* q$ A9 _conducted at higher temperatures for a shorter period (10). Testing at multiple time points could
' C; u9 N0 {; ~  `6 z1 pprovide information on the rate of degradation and primary and secondary degradation products.; B% `, c1 B* {9 j: n& l# j6 D+ l
In the event that the stress conditions produce little or no degradation due to the stability of a drug
: D+ s5 u7 F5 K/ o# {molecule, one should ensure that the stress applied is in excess of the energy applied by  ?$ }3 X% g  c: @4 a8 T
accelerated conditions (40 °C for 6 months) before terminating the stress study.
+ D7 m0 h: T2 u, ~! c: TAcid and base hydrolysis.5 s8 {8 O7 F$ z/ A  I" V
Acid and base hydrolytic stress testing can be carried out for drug substances and drug products in
' U( Z6 i8 b) F5 g5 O9 V6 m5 usolution at ambient temperature or at elevated temperatures. The selection of the type and& G/ m4 W- Q3 F* O7 f
concentrations of an acid or a base depends on the stability of the drug substance. A strategy for
- [/ l5 m: a; r% [. Xgenerating relevant stressed samples for hydrolysis is stated as subjecting the drug substance
# P0 S% d2 c: D, K& o9 y8 qsolution to various pHs (e.g., 2, 7, 10–12) at room temperature for two weeks or up to a maximum
$ Z8 Z/ u/ l6 Jof 15% degradation (7). Hydrochloric acid or sulfuric acid (0.1 M to 1 M) for acid hydrolysis and
9 V  a9 a! T! ksodium hydroxide or potassium hydroxide (0.1 M to 1 M) for base hydrolysis are suggested as
! ?( w. U, |- J7 o2 h4 osuitable reagents for hydrolysis (10). For lipophilic drugs, inert co-solvents may be used to4 a9 i9 R/ [( n, ^7 t, K# h% R% D( h
solubilize the drug substance. Attention should be given to the functional groups present in the
5 T4 G# Q9 T! fdrug molecule when selecting a co-solvent. Prior knowledge of a compound can be useful in
4 I5 E' m5 y2 T8 A8 Dselecting the stress conditions. For instance, if a compound contains ester functionality and is very
. ~8 r* x" ^  p  W/ clabile to base hydrolysis, low concentrations of a base can be used. Analysis of samples at various
7 F; B5 \+ S: Nintervals can provide information on the progress of degradation and help to distinguish primary+ V% n6 F1 n$ {; Y; h. y- U- S
degradants from secondary degradants.
' Y0 B+ o1 }. f& VOxidation.
5 v: P9 l; c# r2 t& c, t' k/ m) Z( yOxidative degradation can be complex. Although hydrogen peroxide is used predominantly
- i/ @0 l. C: R3 M0 a, P- Cbecause it mimics possible presence of peroxides in excipients, other oxidizing agents such as
$ C& V7 ~$ m0 Q9 t' qmetal ions, oxygen, and radical initiators (e.g., azobisisobutyronitrile, AIBN) can also be used.
) D0 ~( u" F3 x4 l9 \Selection of an oxidizing agent, its concentration, and conditions depends on the drug substance.2 U+ c. p/ l" e: s! c
Solutions of drug substances and solid/liquid drug products can be subjected to oxidative
8 W9 s3 B3 I. }! l3 K/ a$ Odegradation. It is reported that subjecting the solutions to 0.1%-3% hydrogen peroxide at neutral
: u: Q1 t/ }8 W' {6 rpH and room temperature for seven days or up to a maximum 20% degradation could potentially: _3 A7 I* Z' ^1 J# r" m( d
generate relevant degradation products (10). Samples can be analyzed at different time intervals to* ?! q+ C9 V/ m6 j. W" A% x
determine the desired level of degradation.: t- g* [0 d1 h$ |, w
Different stress conditions may generate the same or different degradants. The type and extent of: a5 s  I, w) T' M/ G
degradation depend on the functional groups of the drug molecule and the stress conditions.! q7 i9 X% F, j7 N5 H$ C1 R! w
Analysis method
' W: }7 d2 n, E0 CThe preferred method of analysis for a stability indicating assay is reverse-phase
- a) x: x5 l5 m! whigh-performance liquid chromatography (HPLC). Reverse-phase HPLC is preferred for several
; Y7 g8 @5 Y5 d+ u" \1 E& G( dreasons, such as its compatibility with aqueous and organic solutions, high precision, sensitivity," v/ Y1 I; J# M% d
and ability to detect polar compounds. Separation of peaks can be carried out by selecting
: z$ d* G! f( Q" rappropriate column type, column temperature, and making adjustment to mobile phase pH.
$ {, K$ P" }# c* }; Y% i! Y6 ePoorly-retained, highly polar impurities should be resolved from the solvent front. As part of
$ k0 U/ V2 Z% p9 g3 q) Kmethod development, a gradient elution method with varying mobile phase composition (very low
2 g5 y2 W7 B% ~organic composition to high organic composition) may be carried out to capture early eluting
- G2 Y# e( E+ K1 E5 x7 d9 d% W8 bhighly polar compounds and highly retained nonpolar compounds. Stressed samples can also be& l- U0 T2 K, G1 `
screened with the gradient method to assess potential elution pattern. Sample solvent and mobile/ n; `" T6 {+ Q  b
phase should be selected to afford compatibility with the drug substance, potential impurities, and7 g3 U; ~! s& g. t0 E6 |, Q! R! H
degradants. Stress sample preparation should mimic the sample preparation outlined in the  a, r8 a( }' n* T  _
analytical procedure as closely as possible. Neutralization or dilution of samples may be necessary) J4 f' y* {' G; Q
for acid and base hydrolyzed samples. Chromatographic profiles of stressed samples should be
! I& }" J0 Q* E/ o6 Ycompared to those of relevant blanks (containing no active) and unstressed samples to determine
. x* b% W' [: z  n( Pthe origin of peaks. The blank peaks should be excluded from calculations. The amount of& ~: D( p! |4 t% \  L
impurities (known and unknown) obtained under each stress condition should be provided along3 e5 Y# E0 Z8 T0 i6 G
with the chromatograms (full scale and expanded scale showing all the peaks) of blanks,
3 \7 e6 o( y* v/ H) C6 Y3 tunstressed, and stressed samples. Additionally, chiral drugs should be analyzed with chiral
/ N. j0 i( e: k0 e& hmethods to establish stereochemical purity and stability (11, 12).. z9 p  t6 H# ?
The analytical method of choice should be sensitive enough to detect impurities at low levels (i.e.,; _" ^* j/ s8 E- \+ V
0.05% of the analyte of interest or lower), and the peak responses should fall within the range of) F3 e* D1 v4 S4 k
detector's linearity. The analytical method should be capable of capturing all the impurities formed
# Q* H6 u0 }- _- \2 y9 Oduring a formal stability study at or below ICH threshold limits (13, 14). Degradation product7 Y$ y0 b- I) T& G, G
identification and characterization are to be performed based on formal stability results in
, s- w, ?+ c( w) Z* Naccordance with ICH requirements. Conventional methods (e.g., column chromatography) or" l8 C9 [$ G1 b
hyphenated techniques (e.g., LC–MS, LC–NMR) can be used in the identification and& s. H; o0 u$ p
characterization of the degradation products. Use of these techniques can provide better insight* y# R) J5 C! t) A7 t, x- E
into the structure of the impurities that could add to the knowledge space of potential structural" ?$ M$ O, M2 V5 W) e1 q8 Z' y
alerts for genotoxicity and the control of such impurities with tighter limits (12–17). It should be3 B5 ^( v9 W6 f: Z/ [# ]
noted that structural characterization of degradation products is necessary for those impurities that
4 o3 i4 X) @5 P4 @4 @are formed during formal shelf-life stability studies and are above the qualification threshold limit
$ \/ h5 S* q* g3 r; K1 G6 c+ G6 ~(13).
- h. }1 d9 S/ W( ]6 ^5 V  c8 jVarious detection types can be used to analyze stressed samples such as UV and mass) \$ X# m6 h% W$ Q  x
spectroscopy. The detector should contain 3D data capabilities such as diode array detectors or' {: \! B. j. e5 e; ^/ c
mass spectrometers to be able to detect spectral non-homogeneity. Diode array detection also; L3 v; I- O  w* T7 o' c* Q
offers the possibility of checking peak profile for multiple wavelengths. The limitation of diode( k0 p9 s0 P$ L% s; o
array arises when the UV profiles are similar for analyte peak and impurity or degradant peak and
" ?! L6 A$ r. m4 G# Nthe noise level of the system is high to mask the co-eluting impurities or degradants. Compounds
/ U$ X  ?% u5 }& i) q2 k' A" G2 ]of similar molecular weights and functional groups such as diastereoisomers may exhibit similar" w* c& A  r6 F. T, S
UV profiles. In such cases, attempts must be made to modify the chromatographic parameters to
8 i# }( Q1 ^, q% D$ Nachieve necessary separation. An optimal wavelength should be selected to detect and quantitate
2 L) d4 H3 N# B, [4 call the potential impurities and degradants. Use of more than one wavelength may be necessary, if+ q% x) x( A* H
there is no overlap in the UV profile of an analyte and impurity or degradant peaks. A valuable4 x1 e$ G7 D( h+ ^( H, {' S" q
tool in method development is the overlay of separation signals at different wavelengths to: p! |) ~' V, s
discover dissimilarities in peak profiles.
/ k2 J2 \- _  t6 S* Y! o* X; sPeak purity analysis.
  b4 ?7 |4 S/ OPeak purity is used as an aid in stability indicating method development. The spectral uniqueness% E/ T8 w& g, C+ {  C
of a compound is used to establish peak purity when co-eluting compounds are present.
2 e" _( T' f7 S% pPeak purity or peak homogeneity of the peaks of interest of unstressed and stressed samples
2 r3 v" Y: w' N2 D# \should be established using spectral information from a diode array detector. When instrument
6 M" P# o; A* R; z0 x' V0 ~software is used for the determination of spectral purity of a peak, relevant parameters should be, b8 e2 x1 d. i* G* F
set up in accordance with the manufacturer's guidance. Attention should be given to the peak
: Q. _$ E1 T; Y; a# K5 Mheight requirement for establishing spectral purity. UV detection becomes non linear at higher% V) ]5 v( y+ n' ^
absorbance values. Thresholds should be set such that co-eluting peaks can be detected. Optimum
4 |. t9 Q: f5 x2 p- b. ]location of reference spectra should also be selected. The ability of the software to automatically2 [  S8 B' i9 K2 h. D
correct spectra for continuously changing solvent background in gradient separations should be
" x5 ?9 T: w# @4 O' ^' {ascertained.
5 h: X3 `6 J* m5 XEstablishing peak purity is not an absolute proof that the peak is pure and that there is no
6 D+ S- {7 p0 Z$ Dco-elution with the peak of interest. Limitations to peak purity arise when co-eluting peaks are( Y* \' w( U2 L" H( ^
spectrally similar, or below the detection limit, or a peak has no chromophore, or when they are
+ L6 ~1 ]3 S: P. \4 I; b8 Lnot resolved at all.+ f6 {6 _3 F# D$ b5 a+ ~  a
Mass balance.7 f5 L) k8 X% \$ @# D" f8 ]
Mass balance establishes adequacy of a stability indicating method though it is not achievable in
" ?' Y  G2 ~3 zall circumstances. It is performed by adding the assay value and the amounts of impurities and0 C, G6 O# B% z$ k' |8 Y( x
degradants to evaluate the closeness to 100% of the initial value (unstressed assay value) with due
1 I' p0 s; E/ `: Y+ Vconsideration of the margin of analytical error (1).* W0 Q2 o2 a. _, `/ l3 K! i$ _
Some attempt should be made to establish a mass balance for all stressed samples. Mass
7 K3 A+ X6 e. F' o; }imbalance should be explored and an explanation should be provided. Varying responses of
9 B; v2 b; E+ z; n5 Z6 ]analyte and impurity peaks due to differences in UV absorption should also be examined by the
& N' M" }3 G6 O$ ouse of external standards. Potential loss of volatile impurities, formation of non-UV absorbing
' h* {+ x0 Q/ Fcompounds, formation of early eluants, and potential retention of compounds in the column) v% s( ^: \; n2 i2 \! h2 ~
should be explored. Alternate detection techniques such as RI LC/MS may be employed to8 R0 J5 n: X0 Q* x) ]! Q8 Q5 s
account for non-UV absorbing degradants.
: a  ]1 E+ X* O0 b4 }9 DTermination of study& j* B+ O, t! l9 t4 n7 z
Stress testing could be terminated after ensuring adequate exposure to stress conditions. Typical. D2 p2 m+ c' M7 e
activation energy of drug substance molecules varies from 12–24 kcal/mol (18). A compound may$ U: w) a' v; T
not necessarily degrade under every single stress condition, and general guideline on exposure; k% g+ t; s1 @
limit is cited in a review article (10). In circumstances where some stable drugs do not show any
; R, q% b2 I: m8 y5 D6 Ldegradation under any of the stress conditions, specificity of an analytical method can be
' |5 O7 `# g, u9 [6 W/ Aestablished by spiking the drug substance or placebo with known impurities and establishing
: H  C) K& E2 r- V3 J4 zadequate separation.
" a0 [: V4 V1 n8 z0 iOther considerations
, i; o$ d- M6 b5 l6 y! zStress testing may not be necessary for drug substances and drug products that have0 ?" L% L2 U- q! ~! E* [* e  ]
pharmacopeial methods and are used within the limitations outlined in USP <621>. In the case4 Q9 L! ^1 N+ O
where a generic drug product uses a different polymorphic form from the RLD, the drug substance
$ p( p; _4 ?* o$ I9 k# eshould be subjected to stress testing to evaluate the physiochemical changes of the polymorphic3 j8 P5 a' v/ b: L" h" p
form because different polymorphic forms may exhibit different stability characteristics.# N3 \; [6 D) y, C% ?# `. Q/ g3 R
Forced degradation in QbD paradigm1 N; a9 q: S/ j0 W% R
A systematic process of manufacturing quality drug products that meet the predefined targets for
- h' }3 K% h, j6 E8 _$ `. Gthe critical quality attributes (CQA) necessitates the use of knowledge obtained in forced
; b5 E4 r! V, j- Pdegradation studies.
- b/ k8 |$ a) R  n$ JA well-designed, forced degradation study is indispensable for analytical method development in a& e5 ]* n) l6 P3 D' y6 W# P, K5 g$ T3 D
QbD paradigm. It helps to establish the specificity of a stability indicating method and to predict& J: k8 @+ N' e4 U1 I! y6 i' @3 M
potential degradation products that could form during formal stability studies. Incorporating all
1 q$ A" r( U2 A0 Q, epotential impurities in the analytical method and establishing the peak purity of the peaks of7 H1 `0 S) I7 K+ R$ x
interest helps to avoid unnecessary method re-development and revalidation.
) D$ e6 A6 ^" T7 y9 w2 PKnowledge of chemical behavior of drug substances under various stress conditions can also
/ [1 }( O7 z8 lprovide useful information regarding the selection of excipients for formulation development.9 T  A7 r' N7 n
Excipient compatibility is an integral part of understanding potential formulation interactions: ~' B& g# n3 z, b" ^! @& }
during product development and is a key part of product understanding. Degradation products due, K; X3 w+ _' I: g6 h4 ?
to drug-excipient interaction or drug-drug interaction in combination products can be examined by
* N( D2 k- W8 Y  T- W2 \stressing samples of drug substance, drug product, and placebo separately and comparing the+ K% d* K3 Y- e& s
impurity profiles. Information obtained regarding drug-related peaks and non-drug-related peaks/ o& r$ m0 L, r* g" x- s' m
can be used in the selection and development of more stable formulations. For instance, if a drug; \1 Y( z" p* y# ?, c, C: Y1 n
substance is labile to oxidation, addition of an antioxidant may be considered for the formulation.1 E5 n. c# P: A% E7 |
For drug substances that are labile to acid or undergo stereochemical conversion in acidic medium,
: a) [' d' s) I  j* r2 g+ _delayed-release formulations may be necessary. Acid/base hydrolysis testing can also provide$ [; ?% c+ z& w6 u) t
useful insight in the formulation of drug products that are liquids or suspensions.
, n' \0 {0 ]. ?* XKnowledge gained in forced degradation studies can facilitate improvements in the manufacturing
$ N/ M0 O3 D. i$ Q& D$ y! Yprocess. If a photostability study shows a drug substance to be photolabile, caution should be
. R7 y" j2 @: q" ^! q# utaken during the manufacturing process of the drug product. Useful information regarding process
& e4 z' {0 s" B+ c+ k* `5 cdevelopment (e.g., wet versus dry processing, temperature selection) can be obtained from thermal' d( c4 P6 X- C. d( }" ]
stress testing of drug substance and drug product.! @! ^5 t) G0 _1 Z* X& `9 y
Additionally, increased scientific understanding of degradation products and mechanisms may
' E2 ]. i- F+ X% xhelp to determine the factors that could contribute to stability failures such as ambient temperature,
' @7 S2 K& s" k7 i, c# D5 }: l; K% Qhumidity, and light. Appropriate selection of packaging materials can be made to protect against# [" ?$ n0 {( c9 l
such factors.- Z+ w: y* o6 o, t! Y7 y& ~. l1 Y' ]
Conclusion2 J+ r: |4 Y% \6 b( R
An appropriately-designed stress study meshes well with the QbD approaches currently being
4 n7 c0 c5 s' \$ j! Vpromoted in the pharmaceutical industry. A well-designed stress study can provide insight in
0 G5 K2 N1 @/ B# m! Y' Y$ Y0 v% f0 b: Hchoosing the appropriate formulation for a proposed product prior to intensive formulation
5 S% Z; T1 P3 tdevelopment studies. A thorough knowledge of degradation, including mechanistic understanding
' Y" C" I' F+ z9 G; }& W/ Lof potential degradation pathways, is the basis of a QbD approach for analytical method
# V3 f* T7 s6 x  s; cdevelopment and is crucial in setting acceptance criteria for shelf-life monitoring. Stress testing8 V5 \1 _% V6 x: ]
can provide useful insight into the selection of physical form, stereo-chemical stability of a drug. O5 B/ q" o1 o" N; T
substance, packaging, and storage conditions. It is important to perform stress testing for generic
0 u- k. n7 O& p+ b& {) o7 ^drugs due to allowable qualitative and quantitative differences in formulation with respect to the1 o; c7 I& s, g+ [
RLD, selection of manufacturing process, processing parameters, and packaging materials.) u# x7 Z9 U) O- F& Z1 T/ @
Acknowledgments
4 |) F+ |$ h, V, K+ aThe author would like to thank Bob Iser, Naiqi Ya, Dave Skanchy, Bing Wu, and Ashley Jung for$ D3 x" M3 B/ V1 j$ s
their scientific input and support.
7 @3 [$ C' ^8 u: @6 \6 f3 VRagine Maheswaran, PhD, is a CMC reviewer at the Office of Generic Drugs within the Office of
- J( m! H( |* y; G% }- zPharmaceutical Science, under the US Food and Drug Administration's Center for Drug
3 @) M* x, L. C) i+ U/ q9 nEvaluation and Research, Ragine.Maheswaran@fda.hhs.gov
1 c6 q4 E6 ^6 `' g3 ?2 m2 |Disclaimer: The views and opinions in this article are only those of the author and do not
. E( X4 ^( ^) M4 _necessarily reflect the views or policies of the US Food and Drug Administration.! _; g2 {2 D+ }8 W/ x: g
References2 N! p& P1 i# z$ K6 S' ]* {
1. ICH, Q1A(R2) Stability Testing of New Drug Substances and Products (Geneva, Feb. 2003).
* o9 l* R7 P( |7 f) L1 \! k2. ICH, Q1B Stability Testing: Photostability Testing of New Drug Substances and Products
) R( T- G& k. D, V(Geneva, Nov. 1996).
% j" ?" z9 a- y0 k7 `3. H. Brittain, Analytical Profiles of Drug Substances and Excipients (Academic Press, London,1 u/ S* A  Y- Y- k1 v: G
2002).' H5 x* i( e$ c; o7 g5 x" k
4. A. Srinivasan and R. Iser, Pharm. Technol. 34 (1), 50–59 (2010).
$ l' v/ S' j- {+ H* |5. A. Srinivasan, R. Iser, and D. Gill, Pharm. Technol. 34(8), 45–51 (2010).# U" Y  F9 I; `; ^: {2 ^
6. A. Srinivasan, R. Iser, and D. Gill, Pharm. Technol. 35 (2), 58–67 (2011).+ P3 \* T' k% I1 F' Y
7. S. Klick, et al., Pharm.Technol. 29 (2) 48–66 (2005).
, q" Y2 y5 k& `/ p( f6 A2 @% ~8. K. M. Alsante, L. Martin and S. W. Baertschi, Pharm.Technol. 27 (2) 60-72 (2003).
  }* K2 W8 ]% G, \9. D. W. Reynolds, et al., Pharm.Technol. 26 (2), 48–56 (2002).; T$ R% ]" R& Q. {; ~4 F8 q% J: h' K
10. K. M. Alsante et al., Advanced Drug Delivery Reviews 59, 29–37 (2007).( {8 v  }+ Y" e/ j  ~
11. FDA, Guidance for Industry on Analytical Procedures and methods Validation Chemistry,. i& V; _4 L3 n  k& x2 v
Manufacturing, and Controls Documentation (draft) (Rockville, MD, Aug. 2000).
+ {: j1 l! S: n" t+ y12. ICH, Q6A: Specifications: Test Procedures and Acceptance Criteria for New Drug Substances
- d6 d! Z! b5 W$ i  Nand New Drug Products: Chemical Substances (Geneva, Oct. 1999).* R% l( ]$ L9 |
13. ICH, Q3A(R2) Impurities in New Drug Substances (Geneva, Oct. 2006).
0 m5 |' D5 }3 Z3 ^14. ICH, Q3B(R2) Impurities in New Drug Products (Geneva, June 2006).9 c7 b6 C' h& L
15. FDA, Guidance for Industry ANDAs: Impurities in Drug Substances (draft), (Rockville, MD,
2 ?) M& [: @7 n" EAug. 2005).
3 R# q4 N: }; |% u, k  u16. FDA, Guidance for Industry ANDAs: Impurities in Drug Products (draft) (Rockville, MD,6 ]4 G/ y, \3 d8 a
Nov. 2010).
. @: i; I; V8 P( H8 ^; O1 p9 F17. EMA, Guideline on the Limits of Genotoxic Impurities, Committee for Medical Products for
# ]1 I: [0 R$ Y' X9 I, O7 IHuman Use (CHMP) (Doc. Ref EMA/CHMP/QWP/251344/2006) (Jan. 1, 2007).
( S1 e; ]7 v" h- P18. K. A. Conners et al., Chemical Stability of Pharmaceuticals, Wiley and Sons, New York, New
5 G0 P, i. G5 P( V( [; A: OYork, 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, 2024-11-29 07:58 PM , Processed in 0.095008 second(s), 18 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