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 " c; G" x5 w0 @7 r8 W0 }
FDA Perspectives: Scientific Considerations of Forced Degradation Studies in ANDA W% v* ~) S/ v9 ^; m' C
Submissions3 [4 Y. r" R" B3 ^8 j/ n
The author outlines the scientific aspects of forced degradation studies that should be considered+ |, C6 A; a; d* P+ B0 r' k
in relation to ANDA submissions. 2 c0 \# E/ d* h3 KMay 2, 20125 `! j4 x# u1 S, s0 I- H$ S
By:Ragine Maheswaran + ]& ~9 y1 d% E+ |1 iPharmaceutical Technology4 U6 ?" s+ O* x- @1 X
Volume 36, Issue 5, pp. 73-80' d5 n, }! g- V! N/ H2 q% b
Forced degradation is synonymous with stress testing and purposeful degradation. Purposeful / c0 y/ \, u. H9 ] @' M/ n; Adegradation can be a useful tool to predict the stability of a drug substance or a drug product with* y% ~" w, `% P) D
effects on purity, potency, and safety. It is imperative to know the impurity profile and behavior of1 o" q/ g4 o$ N& S% |
a drug substance under various stress conditions. Forced degradation also plays an important role * r$ B% p& j) H2 N0 l5 min the development of analytical methods, setting specifications, and design of formulations under: A7 ?/ p' j& c8 h
the quality-by-design (QbD) paradigm. The nature of the stress testing depends on the individual, V& o% }6 h9 G" J% |, K
drug substance and the type of drug product (e.g., solid oral dosage, lyophilized powders, and5 t5 F# T8 Z9 R2 d
liquid formulations) involved (1)., I/ n/ d/ }6 j
The International Conference on Harmonization (ICH) Q1B guideline provides guidance for% T9 E7 J1 i" a0 E
performing photostability stress testing; however, there are no additional stress study 9 A, Z- [# q; Q6 Grecommendations in the ICH stability or validation guidelines (2). There is also limited) B8 e! V- T- F* }- X
information on the details about the study of oxidation and hydrolysis. The drug substance ' _, Z, I& F: p" @ |" M0 ?6 fmonographs of Analytical Profiles of Drug Substances and Excipients provide some information 6 v6 E; Z x" j0 G/ g4 O# D1 \with respect to different stress conditions of various drug substances (3).0 N0 ~; ^) Z* n# T, Z; Q
The forced degradation information provided in the abbreviated new drug application (ANDA) ) j; }" S5 q% r S2 Qsubmissions is often incomplete and in those cases deficiencies are cited. An overview of common! ^! ?8 C: n1 z& D: g1 {( S# Q
deficiencies cited throughout the chemistry, manufacturing, and controls (CMC) section of the5 U4 w: c: D! C' O5 ^! r0 |' E Z
ANDAs has been published (4–6). Some examples of commonly cited deficiencies related to) J, E5 P8 h9 x4 e) }' f9 a: Y
forced degradation studies include the following: & t: J3 i+ Z+ V# T3 J6 bYour drug substance does not show any degradation under any of the stress conditions. Please 5 X0 T5 c. F# T& D7 g9 h+ \repeat stress studies to obtain adequate degradation. If degradation is not achievable, please : L( W8 X. L( Xprovide your rationale. 4 R. V" X$ m. F9 h$ zPlease note that the conditions employed for stress study are too harsh and that most of your drug9 K8 d* Z! ]4 }3 E; ?+ p
substance has degraded. Please repeat your stress studies using milder conditions or shorter$ I+ [3 J' C5 v0 X D
exposure time to generate relevant degradation products. 9 j! q F5 c1 q: g% o/ d4 cIt is noted that you have analyzed your stressed samples as per the assay method conditions. For: b t# l. l: e' H/ T
the related substances method to be stability indicating, the stressed samples should be analyzed, y, ?9 W% g- C% S. V
using related substances method conditions., _5 u8 a r7 h9 N5 t
Please state the attempts you have made to ensure that all the impurities including the degradation5 p. L, x* o9 e5 z# T
products of the unstressed and the stressed samples are captured by your analytical method.) Q8 \: T$ t8 y6 `
Please provide a list summarizing the amount of degradation products (known and unknown) in2 d/ @/ D8 v2 Q
your stressed samples. 3 z+ I( w" S6 v! P- JPlease verify the peak height requirement of your software for the peak purity determination. ' ^: d- m# K9 p3 fPlease explain the mass imbalance of the stressed samples. ( @% e+ }( ?/ v' }- Y, i" nPlease identify the degradation products that are formed due to drug-excipient interactions.4 @6 P0 l: _1 n1 S0 f; ~
Your photostability study shows that the drug product is very sensitive to light. Please explain how" x+ Y( x; s* M J' I- B
this is reflected in the analytical method, manufacturing process, product handling, etc.9 M* ]4 q I' b% p
In an attempt to minimize deficiencies in the ANDA submissions, some general recommendations9 b# n2 Y% r3 G5 j, B; i9 s9 ]
to conduct forced degradation studies, to report relevant information in the submission, and to( I0 g$ q+ R$ g! s( S
utilize the knowledge of forced degradation in developing stability indicating analytical methods, ; E, f) q0 N e0 J8 }; _manufacturing process, product handling, and storage are provided in this article. " t- h$ P. b+ L( dStress conditions 5 _4 S! ?6 M0 y P7 {Typical stress tests include four main degradation mechanisms: heat, hydrolytic, oxidative, and1 |" @( g3 p. a# K, f% X
photolytic degradation. Selecting suitable reagents such as the concentration of acid, base, or" w* U V9 L" r3 X
oxidizing agent and varying the conditions (e.g., temperature) and length of exposure can achieve3 O+ d, I" |& C- D% V" }
the preferred level of degradation. Over-stressing a sample may lead to the formation of secondary 3 \. a( c7 D0 z! i& g0 w9 Tdegradants that would not be seen in formal shelf-life stability studies and under-stressing may not6 n U3 [7 J; ?, V
serve the purpose of stress testing. Therefore, it is necessary to control the degradation to a desired , G' V( X; j) s" F) K/ P( N9 vlevel. A generic approach for stress testing has been proposed to achieve purposeful degradation # q/ c1 a" z0 \( M+ p. z- N; ~that is predictive of long-term and accelerated storage conditions (7). The generally recommended / L. i' k) a/ f7 C7 y! l& c; i0 I5 wdegradation varies between 5-20% degradation (7–10). This range covers the generally6 O; z0 u- U# T
permissible 10% degradation for small molecule pharmaceutical drug products, for which the & B8 S: u( R5 Qstability limit is 90%-110% of the label claim. Although there are references in the literature that& Z7 _$ d6 @( s# \$ [
mention a wider recommended range (e.g., 10-30%), the more extreme stress conditions often " W8 n1 k: i0 X" V: w. \provide data that are confounded with secondary degradation products. 7 }! y& F ^& a6 RPhotostability.; G" S. G4 L ^' E0 Z
Photostability testing should be an integral part of stress testing, especially for photo-labile _8 W! c0 V( |8 f7 \6 U4 Zcompounds. Some recommended conditions for photostability testing are described in ICH Q1B% O. S7 n7 W% |
Photostability Testing of New Drug Substances and Products (2). Samples of drug substance, and0 F, K6 t4 l6 A% S! H7 [
solid/liquid drug product, should be exposed to a minimum of 1.2 million lux hours and 200 watt- W$ E# c8 d" |& a# [; J! q; O a
hours per square meter light. The same samples should be exposed to both white and UV light. To ; X9 d5 H3 g1 X9 pminimize the effect of temperature changes during exposure, temperature control may be . Y- f* C5 F" _ C- D, T2 `necessary. The light-exposed samples should be analyzed for any changes in physical properties 9 k% C& J9 S8 q4 @such as appearance, clarity, color of solution, and for assay and degradants. The decision tree 3 F; i6 L/ c2 ?1 Xoutlined in the ICH Q1B can be used to determine the photo stability testing conditions for drug * m5 d2 m1 M7 N- F' E2 Pproducts. The product labeling should reflect the appropriate storage conditions. It is also J# f X f' d7 Mimportant to note that the labeling for generic drug products should be concordant with that of the : o- F5 P a c+ B7 ?( P. Lreference listed drug (RLD) and with United States Pharmacopeia (USP) monograph 9 B; H+ N9 m' J) G9 Mrecommendations, as applicable. w* P6 j3 q0 _$ ?3 p# V) b
Heat. u3 x6 t6 ^( u. WThermal stress testing (e.g., dry heat and wet heat) should be more strenuous than recommended # ]; I5 l8 T, ]. s, kICH Q1A accelerated testing conditions. Samples of solid-state drug substances and drug products ' ^3 Z Y# x7 v, K# Gshould be exposed to dry and wet heat, whereas liquid drug products can be exposed to dry heat. It, g/ `9 T0 p$ D8 a+ ?% N; x
is recommended that the effect of temperature be studied in 10 °C increments above that for: @5 ]& \# \3 Z5 _5 s; D
routine accelerated testing, and humidity at 75% relative humidity or greater (1). Studies may be7 U& h5 \) Z8 M' a$ W: f( ?
conducted at higher temperatures for a shorter period (10). Testing at multiple time points could3 b' z b: A5 P! |
provide information on the rate of degradation and primary and secondary degradation products. ' p3 \4 ~$ I# W, a5 FIn the event that the stress conditions produce little or no degradation due to the stability of a drug / ~+ _, x" n$ G' lmolecule, one should ensure that the stress applied is in excess of the energy applied by , l" ?7 J: C$ L# x' Uaccelerated conditions (40 °C for 6 months) before terminating the stress study. ) n6 B# E7 e% \9 q8 m* A* zAcid and base hydrolysis. 4 S! `4 V9 p: I E+ b$ @% i9 nAcid and base hydrolytic stress testing can be carried out for drug substances and drug products in 9 z) M# L1 i% d; L: N5 nsolution at ambient temperature or at elevated temperatures. The selection of the type and. s, R N: i" J: X) I2 y, v
concentrations of an acid or a base depends on the stability of the drug substance. A strategy for 9 M3 O$ K2 t% e a) @generating relevant stressed samples for hydrolysis is stated as subjecting the drug substance, D! t/ V2 P% I: k* l1 w0 S t8 H" I
solution to various pHs (e.g., 2, 7, 10–12) at room temperature for two weeks or up to a maximum" e$ X- S, a& o' p' Y" v
of 15% degradation (7). Hydrochloric acid or sulfuric acid (0.1 M to 1 M) for acid hydrolysis and ?; L8 v0 G: G+ F3 f( Fsodium hydroxide or potassium hydroxide (0.1 M to 1 M) for base hydrolysis are suggested as. M' t6 _. W/ u* d# ~ f4 u
suitable reagents for hydrolysis (10). For lipophilic drugs, inert co-solvents may be used to 6 d7 \7 r1 ?9 k7 R4 y5 H" Rsolubilize the drug substance. Attention should be given to the functional groups present in the. D2 C8 k7 A9 |2 X7 ?
drug molecule when selecting a co-solvent. Prior knowledge of a compound can be useful in [) R2 I1 w* z3 s. P3 @+ R
selecting the stress conditions. For instance, if a compound contains ester functionality and is very + A; j6 @" c& s" a8 F9 qlabile to base hydrolysis, low concentrations of a base can be used. Analysis of samples at various ( c( y# {+ q mintervals can provide information on the progress of degradation and help to distinguish primary+ ]" G+ c4 j% `; _* @
degradants from secondary degradants.( l/ V$ p8 `8 [! E
Oxidation. ! g3 g) J0 T; r- \Oxidative degradation can be complex. Although hydrogen peroxide is used predominantly " }: I# }" B: z2 y8 N/ Sbecause it mimics possible presence of peroxides in excipients, other oxidizing agents such as " Q* F, x, j3 S% a7 D! m+ I. Tmetal ions, oxygen, and radical initiators (e.g., azobisisobutyronitrile, AIBN) can also be used.8 \- g6 a) }& J3 z I, T/ m
Selection of an oxidizing agent, its concentration, and conditions depends on the drug substance. $ C3 O# G: J8 c w7 t2 g* h$ XSolutions of drug substances and solid/liquid drug products can be subjected to oxidative : J. a3 Q- s4 @( T2 t; Tdegradation. It is reported that subjecting the solutions to 0.1%-3% hydrogen peroxide at neutral ' k Z# P: F+ }9 lpH and room temperature for seven days or up to a maximum 20% degradation could potentially / A5 E3 P) K& e) d. G2 Z9 c4 ngenerate relevant degradation products (10). Samples can be analyzed at different time intervals to' O7 ?9 S) Z1 B
determine the desired level of degradation.& y/ P( b: j- Y Y& t
Different stress conditions may generate the same or different degradants. The type and extent of 7 T k' G% H, S* Z; Wdegradation depend on the functional groups of the drug molecule and the stress conditions. 6 x/ m+ Q* R0 N+ jAnalysis method 6 ]) M$ d, K5 s! Q2 _1 P# L. \The preferred method of analysis for a stability indicating assay is reverse-phase9 m3 ], i2 F* a, g2 o( ]
high-performance liquid chromatography (HPLC). Reverse-phase HPLC is preferred for several : m! ]7 t+ _7 v3 `/ i6 {$ M; lreasons, such as its compatibility with aqueous and organic solutions, high precision, sensitivity, * I; w# p O9 E# M* ~and ability to detect polar compounds. Separation of peaks can be carried out by selecting 2 v+ a2 Q/ f3 a- F/ F6 Kappropriate column type, column temperature, and making adjustment to mobile phase pH. & {) D; o. K6 E4 j8 JPoorly-retained, highly polar impurities should be resolved from the solvent front. As part of; S& p( e# x# ]& T
method development, a gradient elution method with varying mobile phase composition (very low0 z* p4 {' s( ]% M h+ n& e
organic composition to high organic composition) may be carried out to capture early eluting! o$ S1 l- e& ]5 L+ ^6 [, \5 e+ T
highly polar compounds and highly retained nonpolar compounds. Stressed samples can also be8 M- F# J5 i: J% I" Q8 C
screened with the gradient method to assess potential elution pattern. Sample solvent and mobile 8 G! g& P3 T2 E- g- T- {& Aphase should be selected to afford compatibility with the drug substance, potential impurities, and5 M4 c- D* X! d- K; L
degradants. Stress sample preparation should mimic the sample preparation outlined in the w. L( e! Z7 Y9 l
analytical procedure as closely as possible. Neutralization or dilution of samples may be necessary ; [* t+ m2 a# \, N: K( dfor acid and base hydrolyzed samples. Chromatographic profiles of stressed samples should be 5 b* l4 ^6 [8 e# Tcompared to those of relevant blanks (containing no active) and unstressed samples to determine' ~0 j. [4 g# t2 _
the origin of peaks. The blank peaks should be excluded from calculations. The amount of 1 s9 c+ I8 ^1 c* cimpurities (known and unknown) obtained under each stress condition should be provided along . s6 w- C0 E. A6 A& Twith the chromatograms (full scale and expanded scale showing all the peaks) of blanks, ; S r2 L: {+ n: _5 h/ runstressed, and stressed samples. Additionally, chiral drugs should be analyzed with chiral: O4 N6 V$ h) l B) E5 H. j/ k3 M
methods to establish stereochemical purity and stability (11, 12).! a4 _ l e, R5 W( G O. P. o
The analytical method of choice should be sensitive enough to detect impurities at low levels (i.e., & a2 e6 j' S, R7 o$ h5 t7 X0.05% of the analyte of interest or lower), and the peak responses should fall within the range of ; N3 V8 Y) k7 f4 x' u* d& s7 udetector's linearity. The analytical method should be capable of capturing all the impurities formed5 h ~" F8 ^9 k7 Y7 ~' s, Y
during a formal stability study at or below ICH threshold limits (13, 14). Degradation product' p4 ~6 O& Z. O- E
identification and characterization are to be performed based on formal stability results in1 G) x! D& Y }6 ~0 t4 ~5 V! o
accordance with ICH requirements. Conventional methods (e.g., column chromatography) or * B) K6 D9 f% H2 h$ z0 N' fhyphenated techniques (e.g., LC–MS, LC–NMR) can be used in the identification and! g2 k2 Z5 F6 y5 C
characterization of the degradation products. Use of these techniques can provide better insight& w* s/ }! H" K
into the structure of the impurities that could add to the knowledge space of potential structural+ d' r- j6 k% B. d, n2 ~6 q
alerts for genotoxicity and the control of such impurities with tighter limits (12–17). It should be - g/ r4 t$ t" @5 e1 Nnoted that structural characterization of degradation products is necessary for those impurities that9 Q# q2 o6 `1 Y5 f9 q
are formed during formal shelf-life stability studies and are above the qualification threshold limit2 x' S" Q: C5 ^
(13). 6 s X, G$ t6 I; hVarious detection types can be used to analyze stressed samples such as UV and mass 8 ]% U, m& o" y% }spectroscopy. The detector should contain 3D data capabilities such as diode array detectors or " T6 O2 A7 A' X; l* s3 Rmass spectrometers to be able to detect spectral non-homogeneity. Diode array detection also . D5 j2 W$ G5 R* D6 Hoffers the possibility of checking peak profile for multiple wavelengths. The limitation of diode / g9 {) a7 j; J; j) K- H( q. Sarray arises when the UV profiles are similar for analyte peak and impurity or degradant peak and 2 P) c! `, X& K3 A* rthe noise level of the system is high to mask the co-eluting impurities or degradants. Compounds / _# @2 a6 ]# A/ o6 L, mof similar molecular weights and functional groups such as diastereoisomers may exhibit similar! F$ d4 ?9 ~, x, u5 ?
UV profiles. In such cases, attempts must be made to modify the chromatographic parameters to " l$ j$ B. F9 w8 o/ i3 r6 y4 oachieve necessary separation. An optimal wavelength should be selected to detect and quantitate6 `3 o7 v9 C! B
all the potential impurities and degradants. Use of more than one wavelength may be necessary, if _7 n( J6 x$ S( qthere is no overlap in the UV profile of an analyte and impurity or degradant peaks. A valuable - w5 R# q, y5 R0 w9 [- Ptool in method development is the overlay of separation signals at different wavelengths to8 J0 w& V6 H5 H5 P) A
discover dissimilarities in peak profiles.- Y* s( u( c$ R. y
Peak purity analysis.6 P' d! v% O2 ~: ]0 {# q1 w) P% P( L
Peak purity is used as an aid in stability indicating method development. The spectral uniqueness + T% Y- \6 t4 k2 C$ V6 V# G: xof a compound is used to establish peak purity when co-eluting compounds are present. 3 D3 Q+ V# Y9 _2 t3 s. lPeak purity or peak homogeneity of the peaks of interest of unstressed and stressed samples 3 P# F, \) ?; Eshould be established using spectral information from a diode array detector. When instrument ! w$ z! t3 Y) c% f- Q2 h3 [software is used for the determination of spectral purity of a peak, relevant parameters should be ' z8 t- e- I# |8 O+ x+ fset up in accordance with the manufacturer's guidance. Attention should be given to the peak 9 M3 h! \! c* K" `" B- e9 jheight requirement for establishing spectral purity. UV detection becomes non linear at higher : g8 |8 _' l' B1 q9 ]absorbance values. Thresholds should be set such that co-eluting peaks can be detected. Optimum , u6 q: y& t" z1 ?location of reference spectra should also be selected. The ability of the software to automatically % w3 g7 i! H6 Y5 R5 R }$ rcorrect spectra for continuously changing solvent background in gradient separations should be % V; n. ]& k& M/ O) h( f5 b/ ^ascertained. 7 G! ^7 C0 q2 t% g% c" j4 _ NEstablishing peak purity is not an absolute proof that the peak is pure and that there is no ! T* C2 ~. x! T6 P3 Q2 i) l9 _co-elution with the peak of interest. Limitations to peak purity arise when co-eluting peaks are ! i) h( U8 F" j1 G7 Xspectrally similar, or below the detection limit, or a peak has no chromophore, or when they are4 N$ Q# c3 c! {( F4 e( {+ q. K- l7 ]
not resolved at all.% l, y* M6 Y% s0 U
Mass balance.* `1 _8 _" K1 M8 l8 N, Y
Mass balance establishes adequacy of a stability indicating method though it is not achievable in - e5 t8 H& z4 S Yall circumstances. It is performed by adding the assay value and the amounts of impurities and ) k! ?/ u7 }2 N- s& C4 O, ~degradants to evaluate the closeness to 100% of the initial value (unstressed assay value) with due, I4 ], [* r) b5 r; ]
consideration of the margin of analytical error (1).( @# j- J* u) E) D9 z! Z' z# I
Some attempt should be made to establish a mass balance for all stressed samples. Mass # s& x- _9 x( n6 Nimbalance should be explored and an explanation should be provided. Varying responses of f9 S2 x7 b9 @& Q% `analyte and impurity peaks due to differences in UV absorption should also be examined by the : d( O9 N7 [, t$ B7 D/ A) @! puse of external standards. Potential loss of volatile impurities, formation of non-UV absorbing 8 M) a3 F& d2 A$ Wcompounds, formation of early eluants, and potential retention of compounds in the column " L, p* F) r9 K! d3 Nshould be explored. Alternate detection techniques such as RI LC/MS may be employed to; x4 Z5 E" I% `6 G" e
account for non-UV absorbing degradants. 2 A) d% k, c1 K3 DTermination of study , A6 m9 X# _6 @( _ cStress testing could be terminated after ensuring adequate exposure to stress conditions. Typical 7 ` @7 c' P6 Q U8 m2 yactivation energy of drug substance molecules varies from 12–24 kcal/mol (18). A compound may1 ^" v! I5 y1 F4 g, ^) C
not necessarily degrade under every single stress condition, and general guideline on exposure5 z$ j4 e# ]* n0 |" W/ ~
limit is cited in a review article (10). In circumstances where some stable drugs do not show any ^' v9 G- F1 b9 X' L& k
degradation under any of the stress conditions, specificity of an analytical method can be- O2 }: J. f$ e0 _0 D6 j) ~2 Q
established by spiking the drug substance or placebo with known impurities and establishing, ^! G& X a& W& E3 U q! |% V
adequate separation.1 }9 g k5 V; h- M% a
Other considerations0 b: v5 U1 |/ t" c2 z% K3 g
Stress testing may not be necessary for drug substances and drug products that have : F! [6 `) ]7 |3 U0 i* opharmacopeial methods and are used within the limitations outlined in USP <621>. In the case 3 Z4 e3 j0 p0 G/ L- G. |where a generic drug product uses a different polymorphic form from the RLD, the drug substance $ O6 V4 f1 g: L( i# k: k5 _should be subjected to stress testing to evaluate the physiochemical changes of the polymorphic) S3 F4 d7 y; C/ Y0 A ` d
form because different polymorphic forms may exhibit different stability characteristics. ( ^6 y# v) T2 {" C1 s3 `" h8 E7 {" RForced degradation in QbD paradigm9 \* q/ j- k3 ]% p9 ~7 L4 \
A systematic process of manufacturing quality drug products that meet the predefined targets for& x% C) p+ O' w* Y0 A
the critical quality attributes (CQA) necessitates the use of knowledge obtained in forced % B4 C K* M' d7 kdegradation studies. 6 D6 Y9 |( P6 z" j( x$ a+ AA well-designed, forced degradation study is indispensable for analytical method development in a 1 O+ {: C: a$ I! E7 A* Z7 fQbD paradigm. It helps to establish the specificity of a stability indicating method and to predict 6 c+ Z; M- M8 j% x* Tpotential degradation products that could form during formal stability studies. Incorporating all) ?0 g2 I0 ~6 Z
potential impurities in the analytical method and establishing the peak purity of the peaks of8 V b* t/ @ p5 s0 K0 H4 o( o
interest helps to avoid unnecessary method re-development and revalidation.0 c7 o3 l9 z+ U( p
Knowledge of chemical behavior of drug substances under various stress conditions can also6 b, C* I: S0 f" A
provide useful information regarding the selection of excipients for formulation development. 8 p L7 D; `2 g9 U; y) T3 dExcipient compatibility is an integral part of understanding potential formulation interactions 9 Y: r0 S) L) x: A- F0 B5 wduring product development and is a key part of product understanding. Degradation products due; v. A" y' \& @5 E
to drug-excipient interaction or drug-drug interaction in combination products can be examined by1 y3 Y# t& X9 E7 D9 _/ w) W
stressing samples of drug substance, drug product, and placebo separately and comparing the0 \9 [$ s+ |& G9 M9 f4 U
impurity profiles. Information obtained regarding drug-related peaks and non-drug-related peaks: t7 T( m+ B: w v, Q
can be used in the selection and development of more stable formulations. For instance, if a drug 5 u0 ]: {5 e. Y6 @5 qsubstance is labile to oxidation, addition of an antioxidant may be considered for the formulation. * b. A& m1 J! i" q6 v0 z& ~& BFor drug substances that are labile to acid or undergo stereochemical conversion in acidic medium, + O' Z. B. w A3 z8 ?# I- Odelayed-release formulations may be necessary. Acid/base hydrolysis testing can also provide7 x) g4 w9 _& U0 P
useful insight in the formulation of drug products that are liquids or suspensions.$ Q2 Y4 H: B& o& p$ P8 u- f l
Knowledge gained in forced degradation studies can facilitate improvements in the manufacturing& u: Q& v; H/ u9 x
process. If a photostability study shows a drug substance to be photolabile, caution should be + [8 w! g/ `% ?+ [- z# Y; X' Otaken during the manufacturing process of the drug product. Useful information regarding process! @ |! v9 d' G! a
development (e.g., wet versus dry processing, temperature selection) can be obtained from thermal" s' A+ o$ x) y2 A
stress testing of drug substance and drug product.! ]5 s3 P E1 F( r& Z3 W
Additionally, increased scientific understanding of degradation products and mechanisms may- ^* z6 ~% }6 t- d+ ~5 I0 T
help to determine the factors that could contribute to stability failures such as ambient temperature,6 A$ q, |$ s+ D- Z
humidity, and light. Appropriate selection of packaging materials can be made to protect against% J" G8 b$ d# w7 c/ Q7 D& B
such factors. . i3 t9 R* ?2 M1 L4 t9 R! R# tConclusion ; P2 ]2 L% D7 |+ P: S8 M# gAn appropriately-designed stress study meshes well with the QbD approaches currently being 9 J/ x4 y7 p9 epromoted in the pharmaceutical industry. A well-designed stress study can provide insight in , l' e: Y; L' W L2 tchoosing the appropriate formulation for a proposed product prior to intensive formulation 8 d; g( s$ I S8 I9 Udevelopment studies. A thorough knowledge of degradation, including mechanistic understanding$ g2 T! Q! L# @ T8 B0 ^
of potential degradation pathways, is the basis of a QbD approach for analytical method8 r( D+ T e/ H4 v# M3 I4 C
development and is crucial in setting acceptance criteria for shelf-life monitoring. Stress testing. H+ y1 M0 O9 t1 N6 V
can provide useful insight into the selection of physical form, stereo-chemical stability of a drug! u8 @5 Q, Y; _
substance, packaging, and storage conditions. It is important to perform stress testing for generic. D5 K" z- l3 d4 ]! X0 E
drugs due to allowable qualitative and quantitative differences in formulation with respect to the9 X5 u- j( i4 z( V
RLD, selection of manufacturing process, processing parameters, and packaging materials. 5 T- l. J" F/ {5 y+ a8 N( @Acknowledgments0 m! M) L; [9 C; {, K
The author would like to thank Bob Iser, Naiqi Ya, Dave Skanchy, Bing Wu, and Ashley Jung for* A9 T2 `7 d2 H+ z/ q
their scientific input and support.# s1 X* ^! A) r6 f: z8 ?/ o6 n
Ragine Maheswaran, PhD, is a CMC reviewer at the Office of Generic Drugs within the Office of4 c2 h. D' s. A8 G
Pharmaceutical Science, under the US Food and Drug Administration's Center for Drug + X4 U% I z# |Evaluation and Research, Ragine.Maheswaran@fda.hhs.gov ; O/ K" g: @1 v3 D6 e# pDisclaimer: The views and opinions in this article are only those of the author and do not& P/ p9 \: l$ w# ^; _# h) ^
necessarily reflect the views or policies of the US Food and Drug Administration. E2 s; b. a9 v3 t6 O9 y4 qReferences* K4 F( D! N/ D/ L
1. ICH, Q1A(R2) Stability Testing of New Drug Substances and Products (Geneva, Feb. 2003). 4 Y% K$ \8 a7 o; c( { `2. ICH, Q1B Stability Testing: Photostability Testing of New Drug Substances and Products* a5 U D/ f) Q1 Y" J: b
(Geneva, Nov. 1996).- n7 _. i0 N! R' W1 D
3. H. Brittain, Analytical Profiles of Drug Substances and Excipients (Academic Press, London,7 t2 u9 W, x5 w
2002).8 p- b, T3 ?0 R5 o; {
4. A. Srinivasan and R. Iser, Pharm. Technol. 34 (1), 50–59 (2010).1 k, n( A9 c, `' w2 y! \% _6 L
5. A. Srinivasan, R. Iser, and D. Gill, Pharm. Technol. 34(8), 45–51 (2010). {% U3 z9 O# l
6. A. Srinivasan, R. Iser, and D. Gill, Pharm. Technol. 35 (2), 58–67 (2011). # Z2 ~& M3 Z6 O5 J( t7. S. Klick, et al., Pharm.Technol. 29 (2) 48–66 (2005). ' c7 A/ `1 {) z# B- @4 u. L8. K. M. Alsante, L. Martin and S. W. Baertschi, Pharm.Technol. 27 (2) 60-72 (2003). , u0 V/ L: \6 Y) b9. D. W. Reynolds, et al., Pharm.Technol. 26 (2), 48–56 (2002). 1 t A& C0 I9 M, w; p4 F10. K. M. Alsante et al., Advanced Drug Delivery Reviews 59, 29–37 (2007).: X4 x9 u# K( {2 ^ h3 }& t
11. FDA, Guidance for Industry on Analytical Procedures and methods Validation Chemistry, : A: B# }# N. i: n4 Y- lManufacturing, and Controls Documentation (draft) (Rockville, MD, Aug. 2000).5 ^7 W! J2 ?( b$ l9 [, J
12. ICH, Q6A: Specifications: Test Procedures and Acceptance Criteria for New Drug Substances & W2 B! c7 ~- }. j- { \& i$ Cand New Drug Products: Chemical Substances (Geneva, Oct. 1999). 8 A9 z! V! Q1 V* A13. ICH, Q3A(R2) Impurities in New Drug Substances (Geneva, Oct. 2006). 0 |3 A4 }: q+ N5 o14. ICH, Q3B(R2) Impurities in New Drug Products (Geneva, June 2006).& b4 \$ e- p0 F5 @* {/ T
15. FDA, Guidance for Industry ANDAs: Impurities in Drug Substances (draft), (Rockville, MD, # N0 M8 T4 C" o. g4 VAug. 2005). 0 i& L* A: ]5 q# E: k" j16. FDA, Guidance for Industry ANDAs: Impurities in Drug Products (draft) (Rockville, MD, * H( M4 H; H9 S8 \ ENov. 2010).2 C( j% L* V* h( F# f8 L
17. EMA, Guideline on the Limits of Genotoxic Impurities, Committee for Medical Products for + ~# W) l* w' C( ?& ]9 Y6 S: mHuman Use (CHMP) (Doc. Ref EMA/CHMP/QWP/251344/2006) (Jan. 1, 2007). 9 |" T- J! }" n18. K. A. Conners et al., Chemical Stability of Pharmaceuticals, Wiley and Sons, New York, New4 K& _2 ?1 P) J% W, W) B ]
York, 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,