Stability Studies and Testing of Pharmaceuticals: An Overview

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/342902068

Stability Studies and Testing of Pharmaceuticals: An Overview

Article  in  Lc Gc North America · June 2020

CITATIONS READS

0 37

2 authors:

Kim Huynh-Ba Michael W Dong


Pharmalytik 99 PUBLICATIONS   1,310 CITATIONS   
54 PUBLICATIONS   192 CITATIONS   
SEE PROFILE
SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Regulatory View project

A series of 3 white papers on uhplc View project

All content following this page was uploaded by Michael W Dong on 13 July 2020.

The user has requested enhancement of the downloaded file.


2  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  JUNE 2020WWW.CHROMATOGRAPHYONLINE.COM

K. Huynh-Ba and M. W. Dong, Stability Studies and Testing of Pharmaceuticals: An Overview, LCGC North Am.
38(6), 325-336, 2020.

PER SPEC TI V E S
IN MO DERN HPLC

Stability Studies and Testing of


Pharmaceuticals: An Overview
This installment is the first of a series of three white papers on stability studies and testing of pharmaceuticals, as well
as the development and validation of stability-indicating high performance liquid chromatography (HPLC) methods.
The series is co-authored by Kim Huynh-Ba, a subject-matter expert on stability testing and regulatory compliance, and
Michael Dong, the columnist on “Perspectives in Modern HPLC.” This first installment provides a comprehensive and
updated overview of stability studies and testing of small molecule drugs, current regulatory requirements, and industry
practices for forced degradation, as well as possible approaches for reduced testing and data evaluation to expedite
stability study timelines.
Kim Huynh-Ba and Michael W. Dong
Determining product shelf life is a ing of the science, best practices, and impurities), physical testing (pH, clarity,
regulatory requirement for pharmaceuti- regulatory expectations of stability pro- particulates), and sterility tests as appro-
cals and many other regulated consumer grams. priate.
products. The shelf life of medicines is set The stability profile, a critical qual-
following stringent regulations; therefore, ity attribute (CQA) of a pharmaceutical Stability Studies in
efficient application of stability science is entity, is based primarily on the physi- New Drug Development
critical. The shelf life (expiration dating cochemical properties of the drug sub- During drug discovery, the medicinal
or expiry) is displayed on labels of phar- stance (DS) and drug product (DP). The chemist focuses on synthesizing new
maceutical products to ensure the integ- DS is the active pharmaceutical ingredi- chemical entities (NCEs) that interact
rity, quality, and potency of the product ent (API), with small amounts of impuri- selectively with the molecular targets
when used within that time period. Shelf ties and degradation products. A DP typ- (such as receptors or enzymes), leading
life is established using data that are ically contains a single DS; when the DP to potential disease mitigation (1). The
generated to verify the label claim, and formulation has more than one DS, it is structural motifs of a leading NCE can-
approved by the regulatory agencies. An referred to as a combination product. For didate are then optimized for biological
expiration date is required by regional example, many over-the-counter (OTC) activities and safety by the synthesis of a
laws to ensure the safety, efficacy, and products contain multiple active ingredi- series of analogs. The optimization tools
quality of the drug products, and that ents. For small-molecule orally delivered include in vitro and in vivo animal studies,
these criteria are maintained throughout DPs, such as tablets or capsules, a typi- (such as biochemical target binding, bio-
the labeled shelf life of the pharmaceuti- cal expiration period is two to five years markers, or animal models), bioavailabil-
cal product. under room temperature storage. Table I ity (pharmacokinetics), and toxicological
Most companies have established summarizes the characteristics of stabil- evaluations (1,2). The desired NCEs need
standard operating procedures (SOPs) to ity studies and testing. Each is discussed to be reasonably stable to be viable
supplement regulatory guidelines to pro- further in this paper. drug candidates, which are scaled up for
vide more specific details, to ensure that In this paper, we use solid oral dosage clinical trials using simple formulations.
stability studies are appropriate for their forms, such as tablets or capsules, as Eventually, the final DS is formulated into
specific product types. Nevertheless, sta- examples. Four types of testing for orally a commercial drug product, and submit-
bility programs and their practices can available products, listed in Table II, are ted for regulatory approval.
vary widely, particularly between large discussed. Biological products are not In most pharmaceutical companies,
and small pharmaceutical companies, covered here. the nomination of an NCE to the status
often due to the depth of knowledge Other dosage forms such as parenter- of a drug development candidate trig-
and available resources. The primary aim als also have similar types of tests, includ- gers the formation of a multidisciplinary
of this paper is to increase understand- ing identity, chemical testing (assay and technical development team. This team
WWW.CHROMATOGRAPHYONLINE.COM JUNE 2020  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  3

is responsible for taking the drug can-


TABLE I: Summary of characteristics of stability studies and testing
didate into clinical trials in humans, and
eventually to drug approval, produc- Stability Study
Stability Testing Characteristics
Characteristics
tion, and commercialization. The team
is responsible for activities referred to in • Drug regulations require expiration dates to be posted
Stability studies must be
on the labels for all pharmaceutical products.
regulatory documents under the head- conducted to meet
• Stability data of DS and DP are submitted to regulatory
regulatory expectations
ing of “Chemistry, Manufacturing, and agencies.
Control” (CMC), and thus the team is • Formal stability studies are used to justify storage
often called the CMC team. conditions and the expiration dating of a packaged
product.
Stability studies for clinical trial materi- Stability studies fall • Other studies are secondary or those used to support
als (CTMs) are conducted to monitor their into different types the primary stability data set.
CQAs and help identify which formula- • Stress studies include photostability studies and studies
that support temperature excursions throughout the
tion will result in a successful candidate supply chain.
for regulatory submission. In the United
• Stability studies are conducted at several environmental
States, an expiration date is not required conditions based on the desired labeled storage conditions.
on the label of a CTM, but this is required • For products to be stored at room temperature, studies
in many countries, such as those in Studies are conducted are conducted at room temperature and accelerated
under multiple storage conditions. Intermediate condition can be studied if
Europe. Forced degradation studies (also conditions samples at accelerated
referred to as stress studies) are used to conditions may not meet room temperature specifications.
challenge the stability-indicating power • Refrigerated or freezing conditions are used for
products that are not chemically stable at room tem-
of the analytical method. The acceler- perature (such as biologics).
ated studies, which require storage
• Attributes that change over time are required to be
under higher temperature and humidity assessed in a stability study.
compared to normal room-temperature • The chemical, physical, microbiological, and
Stability-indicating
conditions, allow the samples to degrade performance characteristics of DP must meet
analytical procedures
acceptance criteria throughout the shelf life.
at a faster rate. Data from accelerated must be followed
• Forced degradation studies are performed to challenge
studies may be extrapolated to project the stability-indicating power of the method and to
the results of a longer-term, controlled- evaluate the major degradative pathways of the API.
room-temperature study. Most pharma- • Stability testing consumes a significant part of internal
Stability studies are
ceutical products exhibit a linear degra- analytical testing and outsourcing resources of
resource intensive
dation trend. Based on the stability data, pharmaceutical companies.

a retest period is assigned to the DS, and


an expiration date to the DP.
The CMC team typically consists of • conducting the necessary synthetic pro- and establishing commercial specifica-
scientists from various functions, such cess scale-up of the DS (process chemistry tions (QA and regulatory staff)
as process chemistry (synthetic organic staff) • manufacturing CTMs (CMC team and out-
chemistry), formulation (pharmaceutics), • performing detailed characterization of sourcing and project management staff)
analytical chemistry (analytical develop- the API, developing and validating analyti- • assembling the CMC submission package
ment and quality control [QC]), pharma- cal procedures for DS and DP, and iden- for regulatory filings (CMC team and regu-
cokinetics and drug metabolism (PKDM), tification of critical impurities and degra- latory staff).
outsourcing, regulatory affairs, supply dation products (analytical development In this series of white papers on sta-
chain, and project management. The staff) bility testing, we focus on the tasks per-
degree of involvement depends on the • finding the optimum solid-state form (salt, formed by the analytical development
development phase. For example, the crystallinity) or polymorph of the API (ana- and QC scientists within the CMC team.
analytical development chemist is more lytical staff) The first task for the analytical chemist
involved in the discovery and early- • conducting stability studies to support assigned to the CMC team is to develop
phases of development to support DS clinical development and registration a reasonable stability-indicating method
and DP processes. In contrast, quality (analytical and QC staff) for monitoring the quality of CTMs. This
control chemists and regulatory staff are • designing and developing processes for method is typically a “composite” ana-
more active in the later clinical phases for formulations of CTMs and the final com- lytical procedure that determines both
CTM manufacturing and regulatory filings mercial DP (pharmaceutics staff) the potency (API) and chemical impuri-
(3,4). Some of the team activities include: • setting acceptance criteria for CQAs ties of the DS. Ideally, it can also be used
(specifications) to monitor clinical quality for the assessment of DP samples. Once
a feasible analytical procedure is estab-
4  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  JUNE 2020WWW.CHROMATOGRAPHYONLINE.COM

methods will be covered in the sec-


TABLE II: Four categories of stability tests for tablet or capsule drug products
ond and third installments of the series.
Category Typical Tests Forced degradation and stability studies
The primary stability-indicating assay is typically an HPLC-UV are discussed in later sections here.
Chemical testing method for potency and chemical impurities. Others are tests
A better understanding of stability
for chiral impurities (HPLC-UV) and moisture (Karl Fisher)
studies and their regulatory requirements
Tests for appearance, color, solid-state characterization
Physical testing (crystallinity, polymorph by X-ray power diffraction [XRPD]),
is essential for the analytical chemist for
thermal methods) several reasons (6,7). First, these stud-
Dissolution (or disintegration) tests to measure ies require many different tests (shown
Performance testing
the rate of release of the solid dosage form. in Table II) on multiple batches stored at
Microbial limit testing Tests indicated in USP General Chapters <61> and <62> different storage conditions and pulled
at numerous time intervals. Second, the
process development or formulation
TABLE III: Storage conditions established by the ICH Q1A (R2) guideline for zone I/II. scientists may request many experimen-
Label tal batches of DS or formulations to be
Storage Conditions Storage Conditions to be Studied
Conditions placed on stability, leading to redundant
Long-term storage 25 ± 2 °C and 60 ± 5% RH testing, bringing little added value if not
Controlled
room Intermediate condition 30 ± 2 °C and 65 ± 5% RH managed with a science- and risk-based
temperature approach. Therefore, a better under-
Accelerated condition 40 ± 2 °C and 75 ± 5% RH
standing of the design, intent, and best
Refrigerated Refrigerated condition 5 ± 3 °C
practices of stability studies and a proper
Freezer Freezing condition –20 ± 5 °C
interpretation of associated regulations
can lead to more efficient implementa-
tion of stability programs.
TABLE IV: An example of stress conditions for drug substance

Factor Being Objectives of this Article


Conditions Time
Studied In this installment, we discuss:
a) Open dish • regulations and regulatory expecta-
Q1B exposure levels
Light b) In the immediate container tions of stability programs
(2x to 10x)
c) In the secondary container
• the goals of forced degradation stud-
Heat (solution) a) 60 °C Up to 24 h ies and examples of forced-degrada-
Heat (solid) a) 50 °C, 60 °C, 70 °C, 80 °C Up to 24 h tion protocols
Accelerated a) Open dish 40 °C/75% RH or greater • a strategy for conducting stability
Up to 7 d
stability b) In the immediate container studies to expedite regulatory filings
Peroxide
a) 3% peroxide at RT* a) 30 min to 2 h • modern predictive stability software
b) 3% peroxide at <40 °C b) 30 min to 2 d tools and statistical approaches to
a) 0.1–1 N HCl (at RT) a) 30 min to 2 h expedite stability studies in a science-
Acid (solution)
b) 0.1–1 N HCl (at 60 oC) b) 30 min to 2 d and risk-based approach.
a) 0.1–1 N NaOH (at RT) a) 30 min to 2 h
Base (solution)
b) 0.1–1 N NaOH (at 60 °C) b) 30 min to 2 d
Regulations and Regulatory
* RT = room temperature
Expectations of Stability Programs
Stability is a Critical Quality
lished, forced degradation studies are products (5). This method is validated to Attribute for Pharmaceuticals
performed to evaluate the specificity of ensure adequate analytical performance In the Quality by Design (QbD) approach,
the analytical procedure. This method is (specificity, linearity at 100% target con- CQAs describe the properties or attri-
used in the release testing of the early centration and expected impurities con- butes pertinent to product quality (8–11).
clinical batches and testing of samples centrations, accuracy, precision, sensi- For pharmaceutical products, CQAs are
from initial stability studies to support tivity, and solution stability) for “formal characteristics that impact the safety
clinical trials (3–5). stability studies” (defined as studies con- and efficacy of drug products. Accept-
The stability-indicating assay and ducted under Good Manufacturing Prac- able limits or product specifications must
impurities method is typically a gradient tices [GMP]) supporting the establish- be established and verified by release
reversed-phase HPLC method with UV ment of expiration dates for regulatory testing (11). Stability studies of DS and
detection that can separate the API and filings (5,6). Details on the development DP are conducted throughout the drug
all the known impurities and degradation and validation of the stability-indicating development process, from the pre-
WWW.CHROMATOGRAPHYONLINE.COM JUNE 2020  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  5

clinical stage to final product approval,


TABLE V: An elaborate forced degradation protocol used in an automated laboratory
with the study size dependent on the
phase of development. The initial ana- Stressing Temperature
Stressing Time (days)
Condition (°C)
lytical development activities include the
development of analytical procedures, Water 70 0, 0.25, 0.5, 0.75, 1, 1.5, 2, 3, 5, 7

establishment of acceptance criteria, 0.1 N HCl 70 0, 0.25, 0.5, 0.75, 1, 1.5, 2, 3, 5, 7


forced degradation studies, DS and DP 0.1 N NaOH 70 0, 0.25, 0.5, 0.75, 1, 1.5, 2, 3, 5, 7
method validation, and stress tests (3,4, 0.3% H2O2 Ambient 0, 0.25, 0.5, 0.75, 1, 1.5, 2, 3, 5, 7, 10, 14
12). In early phases, stability studies may Cool White Fluores-
Ambient 0, 0.25, 0.5, 0.75, 1, 1.5, 2, 3, 5, 7, 10, 14
be conducted under limited accelerated cent Light (solution)
conditions, and often without a fully vali- Cool White Fluores-
Ambient 0, 1, 2, 3, 5, 7, 9, 10, 11, 12, 13, 14
dated suite of analytical methods. CTMs cent Light (solid)
are released against a set of acceptance Suntest—UV and
Ambient 8 h (1xICH), 16 h (2xICH)
criteria (such as specifications) for clinical Visible Light (solid)
use by the Quality Assurance (QA) group. PH 2 Buffer 70 0, 1, 3, 5, 7
Once a product proceeds to Phase PH 4 Buffer 70 0, 1, 3, 5, 7
III development and preparation for PH 6 Buffer 70 0, 1, 3, 5, 7
product registration, GMP regulations
PH 8 Buffer 70 0, 1, 3, 5, 7
must be followed for finished pharma-
PH 10 Buffer 70 0, 1, 3, 5, 7
ceutical products starting from the reg-
Week 1: 60
istration batches (13). The registration In solid form at
Week 2: 70 0, 1, 3, 5, 7, 8, 10, 12, 14, 15, 17, 19, 21
batches and commercial products des- ambient humidity
Week 3: 80
tined to be distributed for patients’ use
Week 1: 60
must undergo stringent release testing In solid form
Week 2: 70 0, 1, 3, 5, 7, 8, 10, 12, 14, 15, 17, 19, 21
at 75%RH
by QC to meet the established com- Week 3: 80
mercial specifications for identity, purity,
potency, and quality of the finished prod-
uct. While QC performs release testing TABLE VI: Typical forced degradation studies for drug product of a solid-dosage form
of the registration batches and issues a
Forced degradation study Suggested conditions
Certificate of Analysis (CoA), it is officially
released and signed-off by the QA group Heat Expose drug products at 50 ºC for up to 1 month
(or by a “Qualified Person” [QP] in Euro-
Expose drug products to
pean Union pharmaceutical regulation) Humidity
40 ºC and 75% RH and 25 ºC and 90% RH
for market distribution after receiving
Expose drug product in a petri dish without
approval. A portion of the registration Photostability
cover at twice or three times the ICH Q1B exposure level
batches is generally used for “formal
(registration) stability studies,” and upon
commercialization, stability samples are
evaluated annually in marketed product TABLE VII: Typical stability studies to support early-phases clinical trials
stability programs.
Intended Storage Conditions to Be Studied Time Points (in Months)
Stability testing of CTMs typically
includes tests for appearance, identifi- 25 ºC and 60% RH Time point zero, 1, 2, 3, 6, 9,
Room temperature or 30 ºC and 65% RH and 12, or end of the study
cation and quantification (assay), impu-
rities, dissolution (for solid dosage 40 ºC and 75% RH 1, 2, 3, 6

forms), moisture, and additional physical Time point zero, 1, 2, 3, 6, 9,


2–8 ºC
Refrigerated and 12, or end of study
characterizations such as X-ray powder
diffraction (XRPD) tests for crystallinity 25 ºC and 60% RH 1, 2, 3, 6
and polymorph form. This set of tests
(customized for the specific DS or DP,
or compendial tests) is documented in
the product specifications. They must content, performance (dissolution), and In a stability study, representative
be used to assess the identity, physical microbial activity (4,14,15), to ensure that batches of pharmaceuticals are stored in
form (color, size, and crystallinity), purity drug quality is maintained throughout controlled stability chambers at different
(chemical and chiral purity) (3,4), water the stability studies. temperatures and relative humidity (RH)
6  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  JUNE 2020WWW.CHROMATOGRAPHYONLINE.COM

packaging selection to establish the


TABLE VIII: Example of a simple bracketing design
drug product’s shelf life, storage condi-
Strength 50 mg 100 mg 250 mg 500 mg tions, and commercial specifications.
Batch A B C A B C A B C A B C Post-marketing studies are conducted
to verify that any changes to raw mate-
50 mL T T T - - - - - - T T T rial, manufacturing steps, or container
Container
100 mL - - - - - - - - - - - - or closure systems have not affected the
size
250 mL T T T - - - - - - T T T DP through its expiration date based
on product commitment. Any planned
change of raw material, manufactur-
TABLE IX: Examples of factors to considering bracketing ing, or packaging must be evaluated to
determine if additional stability studies
or regulatory filings are needed.
1. Container size
2. Container wall thickness
The United States Food and Drug
3. The surface area of the volume Administration (FDA) has determined
4. The volume of the product that the stability profile is a critical attri-
5. The weight of the product
6. The permeability rate of water per dosage unit
bute of a pharmaceutical product. There-
7. The strength of a similar drug/excipient drug ratio fore, stability studies are necessary to
support any submission such as an Inves-
tigational New Drug (IND), New Drug
Application (NDA), or Abbreviated New
TABLE X: An example of a two-thirds factorial matrixing design
Drug Application (ANDA) filing (1). The
Timepoint Months on stability stability data packages within the CMC
Batch 0 3 6 9 12 18 24 36 section are typically the most exten-
sive non-clinical sections in these sub-
A T T - T T - T T missions. They include tabulated data,
Strength 1 graphs, narratives to summarize the
B T T T - T T - T
stability profile of the packaged product
C T - T T T T T T to justify specifications, and proposed
A T - T T T T T T expiry for the product.
Strength 2 B T T - T T - T T
C T T T - T T - T Harmonization of Regulations:
T = for stability testing scheduled for that timepoint
ICH Quality Guidelines
for Stability Studies
Given that stability testing is expensive
TABLE XI: Examples of factors for which matrixing can be considered and labor-intensive, it is imperative to
understand the regulatory requirements
1. Different batches and their intent to avoid unnecessary
2. Different concentrations studies. In addition, regulations vary sig-
3. Different filling
nificantly from country to country, which
4. Different container size
5. Different closure composition impacts the cost of new drug develop-
6. Different closure system ment and the registration timeline for a
global product launch. Therefore, in the
early 1990s, the International Council
levels in proposed packaging (containers, packaging conditions. Formal stability for Harmonisation of Technical Require-
closure systems) (6,7). These storage con- testing to support regulatory filings must ments for Pharmaceuticals for Human
ditions represent long-term, accelerated, be conducted with validated methods Use (ICH), consisting of representatives
or stress storage conditions. Samples are according to GMP regulations (13,16). from regulatory agencies and scientists
periodically removed from these cham- These stability data are also used to from pharmaceutical industries, was
bers according to a pre-approved sched- support the manufacturing, registration, formed. The council’s role is to harmonize
ule, and tested to verify that the phar- shipping, and transportation of commer- the quality requirements for pharmaceu-
maceutical products meet specifications cial products. Furthermore, stability data ticals to minimize redundant testing, and
within the established expiration date are collected through clinical formulation reduce the time and cost of new product
under the recommended storage and phases, process development, proposed development. The ICH stability guideline,
WWW.CHROMATOGRAPHYONLINE.COM JUNE 2020  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  7

TABLE XII: Example of a stability data record (6)

STABILITY ANALYTICAL RECORD


Sample Name: Manufacturing Date:
Storage condition:
Lot#: Manufacturing Site:
Sample Orienta-
Study #: Expiration Date:
tion (if applicable):
Protocol #: Testing Site:
Packaging Information:
Study Start Date: Packaging Site:
Packaging Date:
Study Purpose:
Acceptance Time
Test Name Method 1 Mo 2 Mo 3 Mo 6 Mo 9 Mo 12 Mo
Criteria Zero

Pull Date

Test Date

Appearance
Assay
Impurities
Individual
Total
Dissolution
Average
% RSD
Range
Moisture
Completed By: Date:
Reviewed By: Date:
Approved By: Date:

ICH Q1, was the first quality guidelines method development process, accord- ment of stable formulations and selec-
established by the council, indicating ing to ICH Q1A (R2) and ICH Q2 (R1) tion of suitably packaging
the significant need for harmonization of (8,16). Although regulations mandate • to obtain samples to verify method
global stability requirements. that these studies must be performed, specificity and structure elucidation of
The ICH initially harmonized three the guidance does not provide direc- significant degradation products
geographic regions: the United States, tions on procedures or conditions to use. • to allow the differentiation of impuri-
the European Union, and Japan, which The actual protocols employed appear to ties and degradation products from
encompassed Zone 1 and Zone 2 of the vary widely in different organizations. An the DS, excipients, and other interfer-
World Health Organization (WHO) cli- overview of the chemistry fundamentals ence
matic zones. However, many countries of drug forced degradation studies and • to facilitate the rapid establishment of
outside Zone 1 and 2 joined the ICH, best practices can be found in books, CQAs for selection or elimination of
or voluntarily adopted these guidelines articles, and other resources (17–20). selected testing.
later with various modifications. The cur- Forced degradation studies are per- • to rapidly identify any excipient incom-
rent stability guideline (Q1A [R2]) harmo- formed to investigate the major degra- patibility issues with the DS
nized the storage conditions, frequency dative pathways of the DS and DP and • to generate potential data to support
of required testing, and the minimum support the initial development of the the justification of specifications.
amount of data needed for registration DS stability-indicating methods (3,5,12). Forced degradation studies are con-
(6–8). Table III lists the stability storage High-stress conditions are employed to ducted under high temperature, humid-
conditions established by ICH Q1A (R2) subject DS and DP to conditions more ity, acid/base, oxidative, and light condi-
(8). severe than accelerated conditions to tions in both solid and solution states.
serve several purposes: Table IV lists an example set of stress
Forced Degradation Studies • to provide insight into the degrada- conditions for DS. An important goal of
Forced degradation studies, or stress tion pathways of DS and DP and their forced degradation studies is to gener-
testing, are required by regulations and mechanisms, facilitating the develop- ate a potential level of degradation prod-
scientific necessity during the initial ucts that may form during manufacturing
8  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  JUNE 2020WWW.CHROMATOGRAPHYONLINE.COM

tion protocol used by a pharmaceutical


TABLE XIII: Questions about the conclusions of a stability report
laboratory to collect data for most of
their NCEs using a four-temperature
► What is the stability profile of the product? thermal stressing condition matrix
► Are all lots within the stability trend of the product?
(ambient, 60 oC, 70 oC, 80 oC) for acid,
► Can differences be attributed to a particular factor such as packaging or strength?
► Do all tests meet specifications? base, oxidative, light, humidity, buff-
► Is there any test closely monitored? Is there any tests to be performed ered solution, and prolonged stressed
more often?
duration (up to 21 days). This sample-
► Is there any package closely monitored? Is there any package to be tested
more often? intensive protocol was made feasible
► Do all tests meet specifications? by this centralized and automated
► Do the data support the expiration date?
laboratory specializing in support-
► Are there any stability trends?
► Discuss any out-of-specification investigation. ing forced degradation studies for
► Discuss any statistical evaluation of the data set. the development site. The laboratory
► Would the stability database be satisfactory to the proposed or approved expiration
utilized automated sample storage
dating?
and retrieval system called “Powde-
rium” (a powder dispensing worksta-
tion), robotics, programmable liquid
and on stability storage. Typically, forced doing so is neither practical nor neces- handling, and refrigerated storage of
degradation studies are conducted until sary. A few papers have suggested a quenched samples (18,19).
a 5–20% loss of API is observed (18,19). series of extensive steps of pH changing Table VI provides a summary of forced
This range is set to produce a reason- from pH 2 to pH 10 buffer. It may not be degradation studies performed on solid
able amount of degradation products necessary as a routine practice, unless for dosage forms. Note that acid, base,
to facilitate method development and a specific DS or as part of a stress Design and oxidative exposures are not used,
to avoid secondary degradation prod- of Experiments (DoE) to establish a sta- because these conditions are not repre-
ucts from an unstable degradant (which bility model (21). sentative of realistic storage or exposure
are not observed under actual stability situations. The length of exposure should
conditions). It should be noted that the Photostability be evaluated carefully, depending on
conditions should be selected based on Photostability studies are essential, and the dosage forms or the formulation. For
the physicochemical properties of the must be performed to generate primary example, capsule formulations or film-
individual DS and DP. light-induced degradation products by coated tablets may not be able to with-
subjecting one representative batch of stand a temperature higher than 60 oC for
Thermal Degradation the DS to the light exposure listed in ICH an extended length of time.
The effect of temperature is studied by Q1B. The standard light exposure is 1.2 Similar to the DS, a goal of 5% to 20%
exposing the drug substance to high million lux hours of visible light and 200 loss of active ingredient is recommended.
temperatures in increments of 10 oC from lux hours of near UV, with storage condi- If DS or DP is stable and the desired deg-
50 to 80 oC. Thermal degradation is the tions controlled at room temperature. radation level cannot be reached under
primary degradation pathway of the DS These studies can be repeated when high-stress conditions, studies should be
in the solid state. there is a change of DS samples, such terminated. Overstressing a sample may
as a new synthetic route, with different lead to secondary degradation products
Solution State Acid/Base Degradation crystallinity form, different supplier, or a that are not present in formal stability
Solution-state degradation mechanisms change of DP formulation. studies. In contrast, understressing may
may be different, and should also be During the method development lead to insufficient degradation products
explored. phase, forced degradation samples are for method development or identifica-
If the drug substance is not soluble typically evaluated using a preliminary tion (12). For each condition of the forced
under aqueous conditions, then a small mass spectrometry (MS) compatible degradation study, the chromatogram is
amount (up to 10%) of organic solvent, HPLC method with a photodiode array compared with that of a control sample
acetonitrile, or methanol can be used to detector (PDA) to collect information to document the mass balance of the API
solubilize the DS, and the acid or base on peak area under the curve. The MS (% loss of API) and % degradation (nor-
can then be added to stress the samples. and PDA data are used for peak tracking, malized peak area %). For all conditions,
Attention should be given to the func- peak purity assessment, identification of the peak shapes and peak purity (by
tional groups present in the drug mol- degradation products, impurities, and PDA and MS) of all components should
ecule when selecting a co-solvent (20). interferences (3,5,19). be assessed for coelution or presence of
It is not advisable to use a high con- Table V shows an example of a more interferences (3–5).
centration of acid or base, because extensive, elaborate forced degrada- Accelerated and stress studies are
WWW.CHROMATOGRAPHYONLINE.COM JUNE 2020  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  9

performed to induce quicker degrada- ensure that sample removal is tracked Predictive Tools to
tion than what would be observed in rec- and controlled. Maintenance and service Establish Product Expiry
ommended storage conditions. Results records must be available for inspection Stability data are used to establish the
from these studies are used to estimate during audits. Downtime must be limited, expiry of finished products according to
the stability profile of the DS and DP at and a backup plan must be developed to US FDA Good Manufacturing Practices
long term storage conditions, establish avoid issues that can affect sample stor- regulation 21 CFR 211.137. The data
the actual degradation pathways, and age and data integrity. An annual cham- from the accelerated condition are used
demonstrate the intrinsic stability of the ber inventory program is also necessary to determine a tentative expiration date,
DS. It is recommended that these stud- to track the sample storage and disposi- but testing at full shelf life is necessary to
ies be implemented under GMP condi- tion at any time. confirm the approved expiry.
tions because the results of these studies In this section, we discuss examples
may be used as supporting information Stability Study Strategies of useful software tools and statistical
in regulatory document packages. Full to Expedite “First-in-Human” approaches to evaluate stability data and
shelf-life studies are still needed to verify Clinical Trials to expedite stability studies for establish-
the retest date of the DS and expiration Minimizing “Time to Market” is an essen- ing product expiry.
date of the DP. tial strategy for most pharmaceutical Most tools are science- and risk-based,
While discussions on forced degrada- companies (1). Given that by regulatory and have a robust statistical underpin-
tion studies for NCEs in NDA applications agencies expect to see three months ning that conforms to the principles of
are generally plentiful, those for abbrevi- of stability data in IND filings to enable QbD and DoE, which are endorsed by
ated NDA (ANDA) are often insufficient Phase 1 clinical trials, it is customary to regulatory agencies for pharmaceutical
and may result in regulatory deficiencies. perform preliminary accelerated stability development (9).
For instance, the impurity profiles may studies on the Good Laboratory Prac-
not be completely derived or sufficiently tices (GLP) toxicological DS and proto- Modern Predictive
discussed (20). In addition, the labeling type DP batches to provide supporting Methods and Approaches
for generic drug products should be con- data for quicker IND submissions. Early- One widely used approach is the Accel-
cordant with that of the reference listed phase stability studies are typically short, erated Stability Assessment Program
drug (RLD) and with a compendial mono- as shown in Table VII, and the study con- (ASAP), which uses a modified Arrhenius
graph, as applicable (20). ditions depend on where the clinical tri- approximation. This approach uses deg-
als will take place. During Phase 1, the radation kinetics at a few stressed con-
Stability Storage analytical procedures are usually not fully ditions in 1–2 weeks, and extrapolates
Stability samples are kept in a con- validated, though specificity, accuracy, to lower temperatures and humidity for
trolled temperature and humidity stabil- linearity, and precision studies should be longer timeframes. The advantages of
ity chambers. The ICH Q1A (R2) guide- completed. These time-saving strategies this strategy are that the length of these
line requires that these chambers be allow the pharmaceutical companies to experiments is short and the experi-
controlled within ±2 oC and ±5% RH start Phase I clinical trials as soon as pos- ments use fewer resources. The accuracy
for chambers operated at 25 °C/60% sible after the nomination of the drug of this model assumes a first-order reac-
RH, 30 °C/65% RH, and 40 °C/75% RH. candidates. tion, which is often the case for hydro-
The refrigerated condition is controlled Characterization studies of the API and lysis-based degradation (6,21). The dis-
at 5 oC ± 3 oC (2 to 8 oC) with ambient DP continue during formulation develop- advantage is that the presumed kinetic
humidity (monitored but not controlled). ment. It is recommended that the forced model limits the degradation to less than
These chambers are equipped with sen- degradation studies be repeated before a percent and requires extrapolation of
sors for continuous monitoring temper- the release testing and the initiation of three factors simultaneously: time, tem-
ature and relative humidity. These data the formal stability studies under GMP perature, and humidity. These disadvan-
are captured electronically or by chart regulations and ICH Q1A (R2) guideline tages are minimized when the DS and DP
recorders with backup power. Excur- (8). Examples of standard stability proto- have a simple kinetic pathway, and the
sions will be noted with an alarm system. cols that are listed for different dosage primary stability drivers are temperature
When the chambers are out of tolerance forms at different drug development and humidity (22). Another drawback
(see above), investigations must be per- phases are discussed elsewhere (6,7). could occur when the kinetic route is
formed to identify the cause of the issue Results of the formal stability studies complex or nonlinear or involves physical
and determine the impact of the stored used to determine the drug products’ changes, which may result in the linear
samples (7). expiration are included in the stability model not being accurate. Recently, this
The stability chambers are fully quali- section of the NDA or ANDA. approach was also used to select appro-
fied and maintained, and their access priate packaging based on its thermal
is limited to authorized personnel to and moisture-barrier characteristics, and
10  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  JUNE 2020WWW.CHROMATOGRAPHYONLINE.COM

to optimize the drug:excipient ratio of determination of shelf life through the sizes; thus, stability samples stored in
the formulation based on the moisture analysis of data based on a quantitative the 100 mL containers are not tested.
content (21). This approach has gained attribute that is expected to change over Similarly, the 100 mg and 250 mg tablet
popularity, mainly to set expiry for clini- time is to determine the period of time at strengths are bracketed by the 50 mg
cal materials, and has been accepted by which the 95% one-sided (or two-sided) and 500 mg strengths. Assuming that
multiple regulatory agencies (23). confidence limit for the mean curve inter- the stability profiles of these different
Another successful tool is the Predic- sects the acceptance criterion (26,27). The strength formulations are the same
tive Characterization Study (PCS), a rapid annual commitment lots should also be and the compositions of these tablets
development of robust stability models evaluated against the product trend to are similar, regardless of strength, then
using a semi-empirical design space. It assure the consistency of the stability pro- the testing of 100 mg and 250 mg tab-
is a systematic study that evaluates the file of the DP and verify if there is any unin- lets may not be needed. ICH Q1A (R2)
product data of samples exposed to tended change that may impact the DP. requires that three batches (A, B, C) be
a series of storage conditions to build made available for submission. Accord-
predictive stability models to establish Reduced Testing with ing to the guidelines, a pharmaceutical
the design space and control strategy Bracketing and Matrixing company could reduce its testing from
based on the QbD concept, such as Conducting stability programs is expen- potentially 36 configurations to only 12
setting specifications and selecting pro- sive and time consuming. To reduce configurations, resulting in significant
posed container–closure systems. This costs and increase the efficiency of the savings of resources and time (6,7).
approach is also found acceptable by stability program while maintaining reg- Table IX lists some factors to which
many regulatory agencies (22–24). ulatory compliance, many companies a bracketing approach can potentially
Improved modeling tools have employ bracketing or matrixing options apply, assuming that the container–clo-
enabled stability predictions with accel- that reduce the amount of testing and sure system and the headspace of the
erated timeframes compared to those of resources required (6,7). ICH Q1A (R2) containers do not have any impact. The
the traditional extrapolation approach. and ICH Q1D are guidelines to refer disadvantage of bracketing concept is
Besides providing a more accurate way to when reduced testing is applied. that when one of the results is out-of-
to justify an expiry assignment, these Although the bracketing and matrixing specification, then all bracketed con-
models are used to increase the under- concepts are included in multiple global figurations data could then be at risk.
standing of the critical factors that influ- stability guidelines, limited numbers of Full testing would need to be activated
ence the quality of the DS and DP, as published applications can be found. mid-study, complicating the data set,
demonstrated in empirical science and It is even more challenging to provide and negating the benefit of any testing
risk-based approaches detailed in ICH examples for the matrixing for an NCE. reduction. In addition, all bracketed fac-
Q8–Q11. These approaches are often Here, we explain the similarities and dif- tors (for example strengths/formulations
utilized by large pharmaceutical com- ferences between these approaches to and package configurations) will need to
panies with better modeling resources illustrate this application. be registered in all end markets, even
and expertise. A regulatory template if only a subset will be commercialized.
has been shared to standardize on criti- Bracketing Due to the risk, the bracketing concept
cal elements to use risk-based predictive Bracketing refers to a study design in is used more often for post-approval or
stability data for setting up shelf life to which only the extreme variables, such annual product monitoring stability stud-
support clinical development. (25) as extreme strengths, container sizes, ies rather than for DPs from NCEs.
or container fills, are tested. The plan
Trending Analysis assumes that the stability behaviors of Matrixing
The purpose of the stability program products manufactured or packaged at Matrixing is a statistical design of the sta-
is to establish a retest period for DS these extreme levels (such as, for exam- bility schedule in which a selected group
or shelf life for DP that will apply to all ple, highest vs. lowest strength, or larg- of samples of the total number of pos-
future commercial batches. Stability data est vs. smallest package size) encompass sible samples is tested at a specific time
of CQAs of individual batches should and represent the stability behaviors of point. At the next time point, a different
remain within specification throughout products of the intermediate levels, elim- group should be tested. At each time
the assigned retest period of the DS or inating the need for testing at the inter- point, the stability data of that group of
shelf life of DP. Therefore, the trending of mediate levels. A bracketing schedule samples will represent the stability of the
all stability data is very important. Most can be applied to multiple strengths of whole set of studies.
degradation trends of API are linear; identical or closely related formulations. Similar to the bracketing approach, this
however, the degradation pathway could Table VIII shows an example in which design also assumes that all the batches
be a linear, quadratic, or cubic function. the 100 mL container size is bracketed have similar stability profiles; thus, there
The typical ICH Q1E approach for the by the 50 mL and 250 mL container is no need to generate all the results. The
WWW.CHROMATOGRAPHYONLINE.COM JUNE 2020  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  11

long-term trends of all configurations are lines; however, a lack of knowledge of be evaluated to determine the consis-
approximately linear across the whole set matrixing among regulatory reviewers tency of the representative stability data
of all studies, and the comparative sta- and internal quality and regulatory pro- and trends with respect to the manufac-
bility of each presentation can be evalu- fessionals may hinder the use of this turing process and analytical method, by
ated with the reduced schedule. Many application to reduced stability testing. comparing the intercepts and the slopes
options are available with the matrixing of all batches (6,7,23).
concept, which could reduce the testing Stability Reports and
schedule by a third, a half, or two thirds, Regulatory Submissions Summary and Conclusions
depending on the aggressiveness of the Stability reports are required in all phases A robust and science-based stability pro-
statistical extrapolations and risk assess- of regulatory submissions regardless of gram is required for the registration and
ment for the design (6,7). lifecycle state (such as NDA, ANDA, or commercialization of any pharmaceutical
Table X shows an example of a two- supplement NDA). The stability data- product. Regulations in this area are well
thirds factorial matrixing design. In this base comprises laboratory data gen- established; however, the application
schedule, there are two strengths and erated and stored electronically or as and interpretation of the regulations vary
three batches made of each strength. printed reports. These stability data are between companies and regions. ICH
All presentations are tested at time zero, entered into stability reports, which are harmonizes the main regions to help
12 months, and the end of the study (36 essential to communicate information companies use a standardized approach.
months) for the highest possible preci- internally for audit purposes, product This article provides a high-level
sion as these time points are critical investigations, justification of specifica- summary of regulations and regula-
to the DP stability profile. For all other tions, to support product development, tory expectations of stability programs.
time points (3, 6, 9, 18, and 24 months), or simply as a communication tool. All It reviews the goals and practices of
only two-thirds of the presentations get detailed information in the report must forced degradation studies to generate
tested at a rotating schedule. Therefore, be verified for accuracy. For an NDA or sample data to confirm method speci-
all configurations are tested at different ANDA submission, the stability report is ficity. A strategy for conducting accel-
time points for the same amount of times typically the most extensive non-clinical erated stability studies on preliminary
in total. section to summarize the stability data batches to expedite initial regulatory
The matrixing approach requires that of the product during its shelf life (6,27). filing is described. Finally, predictive
all samples be placed in stability cham- Stability data can be presented in a stability software-based and statistical
bers, including at the timepoints that tabular format, in a graphical represen- approaches such as trending, bracketing,
are reduced. Therefore, when there is an tation, a narrative, or a combination of and matrixing are explained. 
instability issue, the testing schedule can formats. Table XII is an example of a sta-
be changed to test all configurations. If bility data record. The data tables can Acknowledgments
some data points are not available, there be generated manually in a document The authors express their gratitude to the
will be a minimum impact on the entire or printed out from a laboratory infor- following colleagues for their time and
study of multiple presentations. mation management system (LIMS) or efforts to provide timely reviews of the
Table XI lists some examples of factors stability management software. All infor- manuscript to improve content accuracy
where matrixing can be used to reduce mation and data must be reviewed and and clarity: Jane Weitzel of Zinata, Jing
testing. Matrixing also provides more verified. The conclusion of the stability Capucao of J&J, Mike Shifflet of John-
flexibility because different reduced report discusses whether the data indi- son and Johnson Consumer Health Care;
testing options can be performed for cate that the product continues to meet Adrijana Torbovska of Farmahem; Alice
various tests depending on the test- the quality specifications or acceptance Krumenaker of TW Metals, LLC; Mark
ing confidence. Realistically, matrixing criteria established for the study and Shapiro of MCS Pharma Consulting;
should be used only for label storage whether the data support the proposed Madhavi Mahavadi of BioMarin Pharma-
conditions. The accelerated stability data or approved expiration dating period ceuticals; Joshua Ayers of ASQ Solutions.
may provide some insight into the sta- (6,7).
bility profile of the product at the label Stability reports should include a sta- References
storage conditions. Because matrixing is tistical evaluation of the currently avail- (1) R.G. Hill and H.P. Rang, Eds., Drug Dis-
a statistical concept, evaluation of data able data and a discussion of the results covery and Development: Technology in
Transition (Churchill Livingston, Elsevier,
can be more involved, and assessment according to ICH Q1E or statements Edinburgh, Scotland, 2nd Ed., 2012)
of all the presentations as a complete that no overall trends and little variabil- (2) M.W. Dong, Drug Development Process,
set is more complicated, which explains ity are observed in the dataset. Table Short Course presented at Pittcon, Phila-
why this approach is not used widely. XIII lists typical questions that should delphia, Pennsylvania, March 2019.

These reduced testing approaches are be addressed in the stability conclusion. (3) M.W. Dong, LCGC North Am. 33(11),
764–775 (2015).
discussed in detail in many global guide- “Poolability” of different batches should
12  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  JUNE 2020WWW.CHROMATOGRAPHYONLINE.COM

(4) D. Kou, L. Wigman, P. Yehl, and M.W. Dong, Analysis, S. Ahuja and M. W. Dong, Eds.,
LCGC North Am. 33(12), 900–909, (2015). (Elsevier, Amsterdam, 2005), Chapter 6.
(5) M.W. Dong, HPLC and UHPLC for Practic- (19) M.W. Dong and H.T. Rasmussen, HPLC
ing Scientists (John Wiley & Sons, Hobo- Method Development Short Course,
ken, New Jersey, 2nd Ed., 2019), Chap- Eastern Analytical Symposium, Somer-
ters 9–11. set, New Jersey, 2004.
(6) K. Huynh-Ba, Ed., Handbook of Stabil- (20) R. Maheswaran, Pharm. Technol. 36(5),
ity Testing of Pharmaceutical Products: 1–6 (2012).
Regulations, Methodologies and Best (21) K. Waterman, Understanding and Pre-
Practices (Springer, New York, 2009). dicting Pharmaceutical product Shelf life,
(7) K. Huynh-Ba, Ed., Pharmaceutical Stabil- In Handbook of Stability Testing of Phar-
ity Testing to Support Global Markets maceutical Products: Regulations, Meth-
(Springer, New York, 2010). odologies and Best Practices (Springer,
(8) International Council for Harmonisation New York, 2009), Chapter 6.
of Technical Requirements for Pharma- (22) K. HuynhBa et al., Meeting Report,
ceuticals for Human Use (ICH) Q1A (R2), Analytical Approaches to Ensure Prod-
Stability Testing of New Pharmaceutical uct Quality – AAPS Joint Face-to-Face
Products (Geneva, Switzerland, 2003). Meeting of the Stability, the Pharmaceu- ABOUT THE CO-AUTHOR
(9) International Council for Harmonisation tical Impurities, and the CMC Statistics
of Technical Requirements for Pharma- Focus Group, AAPS Open 3 (1), 2017. Kim Huynh-Ba
ceuticals for Human Use (ICH) Q8(R2), https://aapsopen.springeropen.com/ is the managing director
Pharmaceutical Development (Geneva, articles/10.1186/s41120-017-0011-z of Pharmalytik LLC. (www.
Switzerland, 2009). (23) H. Williams et al., Pharm. Technol. 41(3), pharmaly tik.com), which
(10) International Council for Harmonisation 52–57 (2017). provides consulting ser-
of Technical Requirements for Pharma- (24) D. Lavrich, Rapid Development of Robust vices in Stability Sciences, Quality
ceuticals for Human Use (ICH), Q3A(R2), Stability Models Using Semi-Empirical Management Systems, and Analytical
Impurities in New Drug Substance, Design Space, AAPS Webinar, March 24, Development. She is an Adjunct Pro-
Q3B(R2), Impurities in New Drug Prod- 2016. https://aapsopen.springeropen. fessor at Temple University’s School
ucts (Geneva, Switzerland, 2006). com/articles/10.1186/s41120-017-0018- of Pharmacy and Illinois Institute of
(11) International Council for Harmonisa- 5#article-info Technology (IIT). Kim is a member of
tion of Technical Requirements for (25) H. Williams, Pharm Technol. 42(8), 42–47 the US Pharmacopeia’s Council of
Pharmaceuticals for Human Use (ICH) (2018). Experts, and the chair of the Chemical
Q6A, Specifications: Test Procedures (26) WHO Expert Committee on Specifica- Medicines Monograph IV Expert Com-
and Acceptance Criteria for New Drug tions for Pharmaceutical Preparations mittee, USP Good Documentation
Substances and New Drug Products, Fifty-second report, annex 10, Stability Practices Expert Panel, USP Organic
(Geneva, Switzerland, 1999). testing of active pharmaceutical ingredi- Impurities of Drug Substance and
(12) H. Bruemmer and N. Belikova, Pharm. ents and finished pharmaceutical prod- Drug Products Expert Panel. She is on
Technol. 41(9), 46–52 (2017). ucts (World Health Organization, 2018). the editorial board of AAPS Open, the
(13) 21 Code of Federal Regulations (CFR), (27) International Council for Harmonisation Journal of GXP Compliance, and the
Part 211.194(a), Current Good Manufac- of Technical Requirements for Pharma- Journal of Validation Technology. Kim
turing Practice for Finished Pharmaceu- ceuticals for Human Use (ICH) Q1E, Eval- has authored ~30 technical publica-
tical Products (US Government Printing uation of Stability Data (Geneva, Switzer- tions and is the editor of two books on
Office, Washington, DC, 2019). land, 2003). stability testing to support registration
(14) United States Pharmacopeia Conven- in global markets.
tion, USP 37/NF 32, General Chapter
USP <1225>, Validation of Compendial
Methods, (United States Pharmacopeial ABOUT THE AUTHOR
Convention, Rockville, Maryland, 2013).
Michael W. Dong
(15) United States Pharmacopeia Conven- is a principal of MWD
tion, USP 37/NF 32, General Chapter USP
Consulting, which provides
<1226>, Verification of Compendial Pro-
training and consulting
cedures (United States Pharmacopeial
Convention, Rockville, Maryland, 2013, services in HPLC and UHPLC,
revised 2018). method improvements, pharmaceutical
analysis, and drug quality. He was formerly
(16) International Council for Harmonisation
a Senior Scientist at Genentech, Research
of Technical Requirements for Pharma-
Fellow at Purdue Pharma, and Senior
ceuticals for Human Use (ICH) Q2 (R1),
Validation of Analytical Procedures: Staff Scientist at Applied Biosystems/
Methodology (Geneva, Switzerland, PerkinElmer. He holds a PhD in Analytical
November 1996, updated 2015). Chemistry from City University of New York.
He has more than 100 publications and
(17) S.W. Baertschi, K.M. Alsante and R.A.
a best-selling book in chromatography.
Reed, Eds., Pharmaceutical Stress Test-
ing: Predicting Drug Degradation (CRC He is an editorial advisory board
Press, Boca Raton, Florida, 2nd Ed., 2011). member of LCGC North America and
the Chinese American Chromatography
(18) H.T. Rasmussen, W. Li, D. Redlich, M.I.
Association. Direct correspondence to:
Jimidar, HPLC Method Development, In
LCGCedit@MMHGroup.com
Handbook of HPLC in Pharmaceutical

View publication stats

You might also like