considerations of force ddegradation studies


2023年12月19日发(作者:solidarity的读音)

Pharmaceutical Technology: FDA Perspectives: Scientific Considerations of Page 1of 6

May 2, 2012

FDA Perspectives: Scientific Considerations of Forced

Degradation Studies in ANDA Submissions

By Ragine Maheswaran

The author outlines the scientific aspects of forced degradation studies that should be

considered in relation to ANDA submissions.

Forced degradation is synonymous with stress testing and purposeful degradation. Purposeful

degradation can be a useful tool to predict the stability of a drug substance or a drug product with effects

on purity, potency, and safety. It is imperative to know the impurity profile and behavior of a drug

substance under various stress conditions. Forced degradation also plays an important role in the

development of analytical methods, setting specifications, and design of formulations under the quality-by-design (QbD) paradigm. The nature of the stress testing depends on the individual drug substance

and the type of drug product (e.g., solid oral dosage, lyophilized powders, and liquid formulations)

involved (1).

The International Conference on Harmonization (ICH) Q1B guideline provides guidance for performing

photostability stress testing; however, there are no additional stress study recommendations in the ICH

stability or validation guidelines (2). There is also limited information on the details about the study of

oxidation and hydrolysis. The drug substance monographs of Analytical Profiles of Drug Substances and

Excipients provide some information with respect to different stress conditions of various drug

substances (3).

The forced degradation information provided in the abbreviated new drug application (ANDA)

submissions is often incomplete and in those cases deficiencies are cited. An overview of common

deficiencies cited throughout the chemistry, manufacturing, and controls (CMC) section of the ANDAs

has been published (4 –6). Some examples of commonly cited deficiencies related to forced degradation

studies include the following:

•Your drug substance does not show any degradation under any of the stress conditions. Please

repeat stress studies to obtain adequate degradation. If degradation is not achievable, please

provide your rationale.

•Please note that the conditions employed for stress study are too harsh and that most of your drug

substance has degraded. Please repeat your stress studies using milder conditions or shorter

exposure time to generate relevant degradation products.

•It is noted that you have analyzed your stressed samples as per the assay method conditions. For

the related substances method to be stability indicating, the stressed samples should be analyzed

using related substances method conditions.

•Please state the attempts you have made to ensure that all the impurities including the

degradation products of the unstressed and the stressed samples are captured by your analytical

method.

/user/?fuid=MTYzOTc3NTk%3D2012-7-9

Pharmaceutical Technology: FDA Perspectives: Scientific Considerations of Page 2of 6•Please provide a list summarizing the amount of degradation products (known and unknown) in

your stressed samples.

•Please verify the peak height requirement of your software for the peak purity determination.

•Please explain the mass imbalance of the stressed samples.

•Please identify the degradation products that are formed due to drug-excipient interactions.

•Your photostability study shows that the drug product is very sensitive to light. Please explain how

this is reflected in the analytical method, manufacturing process, product handling, etc.

In an attempt to minimize deficiencies in the ANDA submissions, some general recommendations to

conduct forced degradation studies, to report relevant information in the submission, and to utilize the

knowledge of forced degradation in developing stability indicating analytical methods, manufacturing

process, product handling, and storage are provided in this article.

Stress conditions

Typical stress tests include four main degradation mechanisms: heat, hydrolytic, oxidative, and

photolytic degradation. Selecting suitable reagents such as the concentration of acid, base, or oxidizing

agent and varying the conditions (e.g., temperature) and length of exposure can achieve the preferred

level of degradation. Over-stressing a sample may lead to the formation of secondary degradants that

would not be seen in formal shelf-life stability studies and under-stressing may not serve the purpose of

stress testing. Therefore, it is necessary to control the degradation to a desired level. A generic

approach for stress testing has been proposed to achieve purposeful degradation that is predictive of

long-term and accelerated storage conditions (7). The generally recommended degradation varies

between 5-20% degradation (7 –10). This range covers the generally permissible 10% degradation for

small molecule pharmaceutical drug products, for which the stability limit is 90%-110% of the label claim.

Although there are references in the literature that mention a wider recommended range (e.g., 10-30%),

the more extreme stress conditions often provide data that are confounded with secondary degradation

products.

Photostability. Photostability testing should be an integral part of stress testing, especially for photo-labile compounds. Some recommended conditions for photostability testing are described in ICH Q1B

Photostability Testing of New Drug Substances and Products (2). Samples of drug substance, and

solid/liquid drug product, should be exposed to a minimum of 1.2 million lux hours and 200 watt hours

per square meter light. The same samples should be exposed to both white and UV light. To minimize

the effect of temperature changes during exposure, temperature control may be necessary. The light-exposed samples should be analyzed for any changes in physical properties such as appearance,

clarity, color of solution, and for assay and degradants. The decision tree outlined in the ICH Q1B can be

used to determine the photo stability testing conditions for drug products. The product labeling should

reflect the appropriate storage conditions. It is also important to note that the labeling for generic drug

products should be concordant with that of the reference listed drug (RLD) and with United States

Pharmacopeia (USP) monograph recommendations, as applicable.

Heat. Thermal stress testing (e.g., dry heat and wet heat) should be more strenuous than recommended

ICH Q1A accelerated testing conditions. Samples of solid-state drug substances and drug products

should be exposed to dry and wet heat, whereas liquid drug products can be exposed to dry heat. It is

recommended that the effect of temperature be studied in 10 °C increments above that for routine

accelerated testing, and humidity at 75% relative humidity or greater (1). Studies may be conducted at

higher temperatures for a shorter period (10). Testing at multiple time points could provide information

on the rate of degradation and primary and secondary degradation products. In the event that the stress

conditions produce little or no degradation due to the stability of a drug molecule, one should ensure thatthe stress applied is in excess of the energy applied by accelerated conditions (40 °C for 6 months)

before terminating the stress study.

Acid and base hydrolysis. Acid and base hydrolytic stress testing can be carried out for drug

substances and drug products in solution at ambient temperature or at elevated temperatures. The

selection of the type and concentrations of an acid or a base depends on the stability of the drug

substance. A strategy for generating relevant stressed samples for hydrolysis is stated as subjecting the

drug substance solution to various pHs (e.g., 2, 7, 10 –12) at room temperature for two weeks or up to a

maximum of 15% degradation (7). Hydrochloric acid or sulfuric acid (0.1 M to 1 M) for acid hydrolysis

and sodium hydroxide or potassium hydroxide (0.1 M to 1 M) for base hydrolysis are suggested as

suitable reagents for hydrolysis (10). For lipophilic drugs, inert co-solvents may be used to solubilize the

drug substance. Attention should be given to the functional groups present in the drug molecule when

selecting a co-solvent. Prior knowledge of a compound can be useful in selecting the stress conditions.

/user/?fuid=MTYzOTc3NTk%3D2012-7-9

Pharmaceutical Technology: FDA Perspectives: Scientific Considerations of Page 3of 6For instance, if a compound contains ester functionality and is very labile to base hydrolysis, low

concentrations of a base can be used. Analysis of samples at various intervals can provide information

on the progress of degradation and help to distinguish primary degradants from secondary degradants.

Oxidation. Oxidative degradation can be complex. Although hydrogen peroxide is used predominantly

because it mimics possible presence of peroxides in excipients, other oxidizing agents such as metal

ions, oxygen, and radical initiators (e.g., azobisisobutyronitrile, AIBN) can also be used. Selection of an

oxidizing agent, its concentration, and conditions depends on the drug substance. Solutions of drug

substances and solid/liquid drug products can be subjected to oxidative degradation. It is reported that

subjecting the solutions to 0.1%-3% hydrogen peroxide at neutral pH and room temperature for seven

days or up to a maximum 20% degradation could potentially generate relevant degradation products

(10). Samples can be analyzed at different time intervals to determine the desired level of degradation.

Different stress conditions may generate the same or different degradants. The type and extent of

degradation depend on the functional groups of the drug molecule and the stress conditions.

Analysis method

The preferred method of analysis for a stability indicating assay is reverse-phase high-performance

liquid chromatography (HPLC). Reverse-phase HPLC is preferred for several reasons, such as its

compatibility with aqueous and organic solutions, high precision, sensitivity, and ability to detect polar

compounds. Separation of peaks can be carried out by selecting appropriate column type, column

temperature, and making adjustment to mobile phase pH. Poorly-retained, highly polar impurities should

be resolved from the solvent front. As part of method development, a gradient elution method with

varying mobile phase composition (very low organic composition to high organic composition) may be

carried out to capture early eluting highly polar compounds and highly retained nonpolar compounds.

Stressed samples can also be screened with the gradient method to assess potential elution pattern.

Sample solvent and mobile phase should be selected to afford compatibility with the drug substance,

potential impurities, and degradants. Stress sample preparation should mimic the sample preparation

outlined in the analytical procedure as closely as possible. Neutralization or dilution of samples may be

necessary for acid and base hydrolyzed samples. Chromatographic profiles of stressed samples should

be compared to those of relevant blanks (containing no active) and unstressed samples to determine the

origin of peaks. The blank peaks should be excluded from calculations. The amount of impurities (known

and unknown) obtained under each stress condition should be provided along with the chromatograms

(full scale and expanded scale showing all the peaks) of blanks, unstressed, and stressed samples.

Additionally, chiral drugs should be analyzed with chiral methods to establish stereochemical purity and

stability (11, 12).

The analytical method of choice should be sensitive enough to detect impurities at low levels (i.e., 0.05%

of the analyte of interest or lower), and the peak responses should fall within the range of detector's

linearity. The analytical method should be capable of capturing all the impurities formed during a formal

stability study at or below ICH threshold limits (13, 14). Degradation product identification and

characterization are to be performed based on formal stability results in accordance with ICH

requirements. Conventional methods (e.g., column chromatography) or hyphenated techniques (e.g., LC

–MS, LC –NMR) can be used in the identification and characterization of the degradation products. Use

of these techniques can provide better insight into the structure of the impurities that could add to the

knowledge space of potential structural alerts for genotoxicity and the control of such impurities with

tighter limits (12 –17). It should be noted that structural characterization of degradation products is

necessary for those impurities that are formed during formal shelf-life stability studies and are above the

qualification threshold limit (13).

Various detection types can be used to analyze stressed samples such as UV and mass spectroscopy.

The detector should contain 3D data capabilities such as diode array detectors or mass spectrometers to

be able to detect spectral non-homogeneity. Diode array detection also offers the possibility of checking

peak profile for multiple wavelengths. The limitation of diode array arises when the UV profiles are

similar for analyte peak and impurity or degradant peak and the noise level of the system is high to mask

the co-eluting impurities or degradants. Compounds of similar molecular weights and functional groups

such as diastereoisomers may exhibit similar UV profiles. In such cases, attempts must be made to

modify the chromatographic parameters to achieve necessary separation. An optimal wavelength should

be selected to detect and quantitate all the potential impurities and degradants. Use of more than one

wavelength may be necessary, if there is no overlap in the UV profile of an analyte and impurity or

degradant peaks. A valuable tool in method development is the overlay of separation signals at different

wavelengths to discover dissimilarities in peak profiles.

/user/?fuid=MTYzOTc3NTk%3D2012-7-9

Pharmaceutical Technology: FDA Perspectives: Scientific Considerations of Page 4of 6Peak purity analysis. Peak purity is used as an aid in stability indicating method development. The

spectral uniqueness of a compound is used to establish peak purity when co-eluting compounds are

present.

Peak purity or peak homogeneity of the peaks of interest of unstressed and stressed samples should be

established using spectral information from a diode array detector. When instrument software is used for

the determination of spectral purity of a peak, relevant parameters should be set up in accordance with

the manufacturer's guidance. Attention should be given to the peak height requirement for establishing

spectral purity. UV detection becomes non linear at higher absorbance values. Thresholds should be set

such that co-eluting peaks can be detected. Optimum location of reference spectra should also be

selected. The ability of the software to automatically correct spectra for continuously changing solvent

background in gradient separations should be ascertained.

Establishing peak purity is not an absolute proof that the peak is pure and that there is no co-elution with

the peak of interest. Limitations to peak purity arise when co-eluting peaks are spectrally similar, or

below the detection limit, or a peak has no chromophore, or when they are not resolved at all.

Mass balance. Mass balance establishes adequacy of a stability indicating method though it is not

achievable in all circumstances. It is performed by adding the assay value and the amounts of impurities

and degradants to evaluate the closeness to 100% of the initial value (unstressed assay value) with due

consideration of the margin of analytical error (1).

Some attempt should be made to establish a mass balance for all stressed samples. Mass imbalance

should be explored and an explanation should be provided. Varying responses of analyte and impurity

peaks due to differences in UV absorption should also be examined by the use of external standards.

Potential loss of volatile impurities, formation of non-UV absorbing compounds, formation of early

eluants, and potential retention of compounds in the column should be explored. Alternate detection

techniques such as RI LC/MS may be employed to account for non-UV absorbing degradants.

Termination of study

Stress testing could be terminated after ensuring adequate exposure to stress conditions. Typical

activation energy of drug substance molecules varies from 12 –24 kcal/mol (18). A compound may not

necessarily degrade under every single stress condition, and general guideline on exposure limit is cited

in a review article (10). In circumstances where some stable drugs do not show any degradation under

any of the stress conditions, specificity of an analytical method can be established by spiking the drug

substance or placebo with known impurities and establishing adequate separation.

Other considerations

Stress testing may not be necessary for drug substances and drug products that have pharmacopeial

methods and are used within the limitations outlined in USP <621>. In the case where a generic drug

product uses a different polymorphic form from the RLD, the drug substance should be subjected to

stress testing to evaluate the physiochemical changes of the polymorphic form because different

polymorphic forms may exhibit different stability characteristics.

Forced degradation in QbD paradigm

A systematic process of manufacturing quality drug products that meet the predefined targets for the

critical quality attributes (CQA) necessitates the use of knowledge obtained in forced degradation

studies.

A well-designed, forced degradation study is indispensable for analytical method development in a QbD

paradigm. It helps to establish the specificity of a stability indicating method and to predict potential

degradation products that could form during formal stability studies. Incorporating all potential impurities

in the analytical method and establishing the peak purity of the peaks of interest helps to avoid

unnecessary method re-development and revalidation.

Knowledge of chemical behavior of drug substances under various stress conditions can also provide

useful information regarding the selection of excipients for formulation development. Excipient

compatibility is an integral part of understanding potential formulation interactions during product

development and is a key part of product understanding. Degradation products due to drug-excipient

interaction or drug-drug interaction in combination products can be examined by stressing samples of

/user/?fuid=MTYzOTc3NTk%3D2012-7-9

Pharmaceutical Technology: FDA Perspectives: Scientific Considerations of Page 5of 6drug substance, drug product, and placebo separately and comparing the impurity profiles. Information

obtained regarding drug-related peaks and non-drug-related peaks can be used in the selection and

development of more stable formulations. For instance, if a drug substance is labile to oxidation, addition

of an antioxidant may be considered for the formulation. For drug substances that are labile to acid or

undergo stereochemical conversion in acidic medium, delayed-release formulations may be necessary.

Acid/base hydrolysis testing can also provide useful insight in the formulation of drug products that are

liquids or suspensions.

Knowledge gained in forced degradation studies can facilitate improvements in the manufacturing

process. If a photostability study shows a drug substance to be photolabile, caution should be taken

during the manufacturing process of the drug product. Useful information regarding process

development (e.g., wet versus dry processing, temperature selection) can be obtained from thermal

stress testing of drug substance and drug product.

Additionally, increased scientific understanding of degradation products and mechanisms may help to

determine the factors that could contribute to stability failures such as ambient temperature, humidity,

and light. Appropriate selection of packaging materials can be made to protect against such factors.

Conclusion

An appropriately-designed stress study meshes well with the QbD approaches currently being promoted

in the pharmaceutical industry. A well-designed stress study can provide insight in choosing the

appropriate formulation for a proposed product prior to intensive formulation development studies. A

thorough knowledge of degradation, including mechanistic understanding of potential degradation

pathways, is the basis of a QbD approach for analytical method development and is crucial in setting

acceptance criteria for shelf-life monitoring. Stress testing can provide useful insight into the selection of

physical form, stereo-chemical stability of a drug substance, packaging, and storage conditions. It is

important to perform stress testing for generic drugs due to allowable qualitative and quantitative

differences in formulation with respect to the RLD, selection of manufacturing process, processing

parameters, and packaging materials.

Acknowledgments

The author would like to thank Bob Iser, Naiqi Ya, Dave Skanchy, Bing Wu, and Ashley Jung for their

scientific input and support.

Ragine Maheswaran, PhD, is a CMC reviewer at the Office of Generic Drugs within the Office of

Pharmaceutical Science, under the US Food and Drug Administration's Center for Drug Evaluation and

Research, aran@ [aran@]

Disclaimer: The views and opinions in this article are only those of the author and do not necessarily

reflect the views or policies of the US Food and Drug Administration.

References

1. ICH, Q1A(R2) Stability Testing of New Drug Substances and Products (Geneva, Feb. 2003).

2. ICH, Q1B Stability Testing: Photostability Testing of New Drug Substances and Products (Geneva,

Nov. 1996).

3. H. Brittain, Analytical Profiles of Drug Substances and Excipients (Academic Press, London, 2002).

4. A. Srinivasan and R. Iser, Pharm. Technol. 34 (1), 50

–59 (2010).

5. A. Srinivasan, R. Iser, and D. Gill, Pharm. Technol. 34(8), 45 –51 (2010).

6. A. Srinivasan, R. Iser, and D. Gill, Pharm. Technol. 35 (2), 58 –67 (2011).

7. S. Klick, et al., l. 29 (2) 48 –66 (2005).

8. K. M. Alsante, L. Martin and S. W. Baertschi, l. 27(2) 60-72 (2003).

9. D. W. Reynolds, et al., l. 26 (2), 48 –56 (2002).

10. K. M. Alsante et al., Advanced Drug Delivery Reviews 59, 29 –37 (2007).

/user/?fuid=MTYzOTc3NTk%3D2012-7-9

Pharmaceutical Technology: FDA Perspectives: Scientific Considerations of Page 6of 611. FDA, Guidance for Industry on Analytical Procedures and methods Validation Chemistry,

Manufacturing, and Controls Documentation (draft) (Rockville, MD, Aug. 2000).

12. ICH, Q6A: Specifications: Test Procedures and Acceptance Criteria for New Drug Substances and

New Drug Products: Chemical Substances (Geneva, Oct. 1999).

13. ICH, Q3A(R2) Impurities in New Drug Substances (Geneva, Oct. 2006).

14. ICH, Q3B(R2) Impurities in New Drug Products (Geneva, June 2006).

15. FDA, Guidance for Industry ANDAs: Impurities in Drug Substances (draft), (Rockville, MD, Aug.

2005).

16. FDA, Guidance for Industry ANDAs: Impurities in Drug Products (draft) (Rockville, MD, Nov. 2010).

17. EMA, Guideline on the Limits of Genotoxic Impurities, Committee for Medical Products for Human

Use (CHMP) (Doc. Ref EMA/CHMP/QWP/251344/2006) (Jan. 1, 2007).

18. K. A. Conners et al., Chemical Stability of Pharmaceuticals, Wiley and Sons, New York, New York,

2nd Ed., p. 19 (1986).

2012 Advanstar Communications Inc.. Permission granted for up to 5 copies. All rights reserved.

You may forward this article or get additional permissions by typing /3.7458?icx_id=771905 into any web browser. Advanstar Communications Inc. and Pharmaceutical Technology logos are registered

trademarks of Advanstar Communications Inc. The iCopyright logo is a registered trademark of iCopyright, Inc.

/user/?fuid=MTYzOTc3NTk%3D2012-7-9


本文发布于:2024-09-21 14:35:31,感谢您对本站的认可!

本文链接:https://www.17tex.com/fanyi/14657.html

版权声明:本站内容均来自互联网,仅供演示用,请勿用于商业和其他非法用途。如果侵犯了您的权益请与我们联系,我们将在24小时内删除。

上一篇:force 释义
标签:读音   作者
留言与评论(共有 0 条评论)
   
验证码:
Copyright ©2019-2024 Comsenz Inc.Powered by © 易纺专利技术学习网 豫ICP备2022007602号 豫公网安备41160202000603 站长QQ:729038198 关于我们 投诉建议