You are on page 1of 46

10_1439PP_CH04 10/11/05 2:53 pm Page 93

4
Drug stability

4.1 The chemical decomposition of 4.4 Factors influencing drug stability 113
drugs 94 4.5 Stability testing and prediction of shelf-
4.2 Kinetics of chemical decomposition in life 127
solution 102 Summary 136
4.3 Solid dosage forms: kinetics of chemical References 137
decomposition 110

Many drugs are susceptible to some form of chemical decomposition when formulated in either
liquid or even solid dosage forms. Such degradation not only leads to a loss of potency of the
drug but may, in some cases, cause changes in the physical appearance of the dosage forms, for
example, discoloration following the photochemical decomposition of the drug.
In this chapter we will identify those classes of drugs which are particularly susceptible to
chemical breakdown and examine some of the precautions which can be taken to minimise the
loss of activity. It is important to be able to determine a time interval over which the drug retains
sufficient potency for it to be used. Usually the shelf-life is assumed to be the time taken for the
concentration of the drug to be reduced to 95% of its value when originally prepared. To be able
to make shelf-life predictions we should first understand the kinetics of the decomposition process.
We will look at how reactions can be classified into various ‘orders’, and how we can calculate
the rate constant for a reaction under a given set of environmental conditions. Methods for
accelerating the drug breakdown using elevated temperatures will be examined and we will see
how it is possible to estimate the stability of the drug for the required storage conditions from these
measurements. Although such experiments cannot replace the rigorous stability testing pro-
cedures on the product in the form in which it is finally to be marketed, they do lead to consider-
able saving of time at the product development stage when it is necessary to identify rapidly a
formulation in which the stability is at an acceptable level.
10_1439PP_CH04 10/11/05 2:53 pm Page 94

4.1 The chemical decomposition of 4.1.1 Hydrolysis


drugs
Drugs susceptible to hydrolytic degradation
In this section we examine various ways in How can we tell whether a drug is likely to be
which drugs in both liquid and solid formu- susceptible to this type of degradation? If the
lations can lose their activity, so that we can drug is a derivative of carboxylic acid or con-
be aware of those chemical groups which, tains functional groups based on this moiety,
when present in drug molecules, can cause for example an ester, amide, lactone, lactam,
stability problems. We will later see how to imide or carbamate (see Scheme 4.1), then we
prevent or minimise the chemical breakdown are dealing with a drug which is liable to
for each type of decomposition process. undergo hydrolytic degradation. We will
Although each decomposition scheme is consider some examples.
considered separately, it should be noted that Drugs that contain ester linkages include
with some drug molecules more than one type acetylsalicylic acid (aspirin), physostigmine,
of decomposition may be occurring at the methyldopate, tetracaine and procaine. Ester
same time; this, of course, complicates the hydrolysis is usually a bimolecular reaction
analysis of the system. involving acyl–oxygen cleavage. For example, the
hydrolysis of procaine is shown in Scheme 4.2.
The hydrolysis of amides involves the
Structure Chemical class cleavage of the amide linkage as for example,
in the breakdown of the local anaesthetic
O O
Imide
cinchocaine (Scheme 4.3).
H This type of link is also found in drugs
RC N CR
such as chloramphenicol, ergometrine and
RCH CO benzylpenicillin sodium.
Lactam Finally, as examples of lactam ring hydroly-
(CH2)n NH sis we can consider the decomposition of
nitrazepam and chlordiazepoxide, which is
RCH CO
Lactone
discussed in more detail later (section 4.2.7).
(CH2)n O Other drugs, apart from the benzodiazepines,
which are susceptible to hydrolysis include
O the penicillins and cephalosporins.
Ester
RC OR
Controlling drug hydrolysis in solution
O
Amide
RC NH2
Optimisation of formulation
Hydrolysis is frequently catalysed by hydrogen
Scheme 4.1 Examples of chemical groups susceptible to ions (specific acid-catalysis) or hydroxyl ions
hydrolysis. (specific base-catalysis) and also by other acidic

OH
H
NH2 C O CH2 CH2 N(C2H5)2 NH2 C  HO CH2 CH2 N(C2H5)2
OH
O
O

Scheme 4.2 Hydrolysis of the ester group of procaine.


10_1439PP_CH04 10/11/05 2:53 pm Page 95

The chemical decomposition of drugs 95

CO NH (CH2)2 N(C2H5)2 COO

OH
 H2N(CH2)2 N(C2H5)2

N O (CH2)3 CH3 N O(CH2)3 CH3

Scheme 4.3 Hydrolysis of the amide linkage of cinchocaine.

or basic species that are commonly encoun- where k and k0 are the rate constants for the
tered as components of buffers. This latter type reaction of the substituted and unsubstituted
of catalysis is referred to as general acid–base compounds, respectively, σ is the Hammett
catalysis. Both types of catalysis will be dealt substituent constant (which is determined
with in greater depth in section 4.4.1. Several by the nature of the substituents and is
methods are available to stabilise a solution of a independent of the reaction), and ρ is the
drug which is susceptible to acid–base catalysed reaction constant, which is dependent on
hydrolysis. The usual method is to determine the reaction, the conditions of reaction and
the pH of maximum stability from kinetic the nature of the side-chains undergoing reac-
experiments at a range of pH values and to tion. Thus, a plot of log k against the Hammett
formulate the product at this pH (section constant (values are readily available in the
4.4.1). Alteration of the dielectric constant by literature1) is linear if this relationship is
the addition of nonaqueous solvents such as obeyed, with a slope of ρ. This concept has
alcohol, glycerin or propylene glycol may in been used, for example, in the production of
many cases reduce hydrolysis (section 4.4.1). the best substituents for allylbarbituric acids
Since only that portion of the drug which is to obtain optimum stability.2
in solution will be hydrolysed, it is possible to
suppress degradation by making the drug less
soluble. The stability of penicillin in procaine– 4.1.2 Oxidation3
penicillin suspensions was significantly
increased by reducing its solubility by using After hydrolysis, oxidation is the next most
additives such as citrates, dextrose, sorbitol common pathway for drug breakdown.
and gluconate. Adding a compound that However, whereas the hydrolytic degradation
forms a complex with the drug can increase of drugs has been thoroughly studied, their
stability. The addition of caffeine to aqueous oxidative degradation has received compara-
solutions of benzocaine, procaine and tetra- tively little attention. Indeed, in cases where
caine was shown to decrease the base-catalysed simultaneous hydrolytic and oxidative degra-
hydrolysis of these local anaesthetics in this dation can occur, the oxidative process has
way. In many cases solubilisation of a drug usually been eliminated by storage under
by surfactants protects against hydrolysis, as anaerobic conditions without an investigation
discussed in section 4.4.1. of the oxidative mechanism.

Modification of chemical structure of drug


The control of drug stability by modifying Oxidation processes
chemical structure using appropriate sub-
Oxidation involves the removal of an electro-
stituents has been suggested for drugs for
positive atom, radical or electron, or the addi-
which such a modification does not reduce
tion of an electronegative atom or radical.
therapeutic efficacy. The Hammett linear free
Oxidative degradation can occur by autoxida-
energy relationship for the effect of substituents
tion, in which reaction is uncatalysed and
on the rates of aromatic side-chain reactions,
proceeds quite slowly under the influence of
such as the hydrolysis of esters, is given by
molecular oxygen, or may involve chain
log k =  log k0 + σρ (4.1) processes consisting of three concurrent
10_1439PP_CH04 10/11/05 2:53 pm Page 96

96 Chapter 4 • Drug stability

reactions – initiation, propagation and termi- Steroids and sterols represent an important
nation. Initiation can be via free radicals class of drugs that are subject to oxidative
formed from organic compounds by the degradation through the possession of carbon–
action of light, heat or transition metals such carbon double bonds (alkene moieties) to
as copper and iron which are present in trace which peroxyl radicals can readily add. Simi-
amounts in almost every buffer. The propaga- larly, polyunsaturated fatty acids, commonly
tion stage of the reaction involves the combi- used in drug formulations, are particularly
nation of molecular oxygen with the free susceptible to oxidation and care must be
radical R‹ to form a peroxy radical ROO ‹, exercised to minimise degradation in formula-
which then removes H from a molecule of the tions containing high concentrations of, for
organic compound to form a hydroperoxide, example, vegetable oils.5 For drugs, such as the
ROOH, and in so doing creates a new free cholesterol-lowering agent simvastatin (I), that
radical (Scheme 4.4). contain conjugated double bonds, addition of
The reaction proceeds until the free radicals peroxyl radicals may lead to the formation of
are destroyed by inhibitors or by side-reactions polymeric peroxides (simvastatin polymerises
which eventually break the chain. The rancid up to a pentamer6), cleavage of which pro-
odour which is a characteristic of oxidised fats duces epoxides which may further degrade
and oils is due to aldehydes, ketones and short- into aldehydes or ketones.
chain fatty acids which are the breakdown Polyene antibiotics, such as amphotericin B
products of the hydroperoxides. Peroxides (II) which contains seven conjugated double
(ROOR,) and hydroperoxides (ROOH) are bonds (heptaene moiety), are subject to attack
photolabile, breaking down to hydroxyl (HO ‹) by peroxyl radicals, leading to aggregation and
andor alkoxyl (RO ‹) radicals, which are them- loss of activity.7
selves highly oxidising species. The presence of The oxidation of phenothiazines to the
residual peroxides in polyoxyethylene glycols sulfoxide involves two single-electron transfer
(PEGs) is a cause for concern when these reactions involving a radical cation intermedi-
excipients are used in formulation, as for ate as shown in Scheme 4.5. The sulfoxide is
example in the case of fenprostalene.4 subsequently formed by reaction of the cation
with water.
The ether group in drugs such as econazole
Drugs susceptible to oxidation
nitrate (III) and miconazole nitrate (IV) is sus-
We will consider some examples of drugs and ceptible to oxidation. The process involves
excipients that are subject to oxidative degra- removal of hydrogen from the C–H bonds in
dation owing to the possession of functional the α-position to the oxygen to produce radi-
groups that are particularly sensitive to oxida- cals, which further degrade to α-hydroperox-
tion. ides and eventually to aldehydes, ketones,
alcohols and carboxylic acids.

HO O
• •
Initiation: X  RH R  XH
O
• • O
Propagation: R  O2 ROO
• •
ROO  RH ROOH  R
H
• •
Termination: ROO  ROO stable product
• •
ROO  R stable product
• •
R  R stable product

Scheme 4.4 Simplified oxidation scheme involving a chain


process. Structure I Simvastatin
10_1439PP_CH04 10/11/05 2:53 pm Page 97

The chemical decomposition of drugs 97

OH
O OH HO OH OH O
HO O Me

HO2C

HO Me
Me
OH

HO
O

H2N Me

OH

Structure II Amphotericin B

R R R

N N N
e  e
•
e e
S S S

Scheme 4.5 Oxidation of phenothiazines.

N • HNO3
N • HNO3

N N
Cl

O O

Cl Cl Cl Cl
Cl Cl

Structure III Econazole nitrate Structure IV Miconazole nitrate

Stabilisation against oxidation


propagation of the chain reaction may be pre-
Various precautions should be taken during vented or delayed by adding low concen-
manufacture and storage to minimise oxida- trations of compounds that act as inhibitors.
tion. The oxygen in pharmaceutical contain- Such compounds are called antioxidants and
ers should be replaced with nitrogen or carbon interrupt the propagation by interaction with
dioxide; contact of the drug with heavy-metal the free radical. The antioxidant free radical so
ions such as iron, cobalt or nickel, which cata- formed is not sufficiently reactive to maintain
lyse oxidation, should be avoided; and storage the chain reaction and is eventually annihi-
should be at reduced temperatures. lated. The structures of some commonly used
antioxidants are given in Scheme 4.6.
Antioxidants Reducing agents such as sodium metabisulfite
It is very difficult to remove all of the oxygen may also be added to formulations to prevent
from a container and even traces of oxygen are oxidation. These compounds are more readily
sufficient to initiate the oxidation chain. The oxidised than the drug and so protect it from
10_1439PP_CH04 10/11/05 2:53 pm Page 98

98 Chapter 4 • Drug stability

OH
OH OH
HO OH
t -Bu t -Bu

t -Bu
OMe Me O O
Butylated hydroxyanisole (BHA) Butylated hydroxytoluene (BHT) Propyl gallate

R3
Vitamin E
HO
a-Tocopherol (R1  R2  R3  Me)
b-Tocopherol (R1  R3  Me, R2  H)
R2 O g-Tocopherol (R1  R2  Me, R3  H)
d-Tocopherol (R1  Me, R2  R3  H)
R1

CH2OH CH2OC(O)(CH2)14 Me
OH OH
O O O O
H H
O
HO SH
SH
HO OH HO OH OH HO
Ascorbic acid Ascorbyl palmitate Thioglycerol Thioglycolic acid
(vitamin C)

O O O O

S S S S
NaO OH NaO ONa NaO O ONa
Sodium bisulfite Sodium sulfite Sodium metabisulfite

Scheme 4.6 Structures of some common antioxidants.

oxidation. Oxidation is catalysed by unpro- In acidic conditions the tetracyclines


tonated amines such as aminophylline, and undergo epimerisation at carbon atom 4 to
hence admixture of susceptible drugs with form an equilibrium mixture of tetracycline
such compounds should be avoided. and the epimer, 4-epi-tetracycline (Scheme
4.7). The 4-epi-tetracycline is toxic and its
content in medicines is restricted to not more
4.1.3 Isomerisation than 3%. The epimerisation follows the kinet-
ics of a first-order reversible reaction (see
Isomerisation is the process of conversion of a equation (4.24)). The degradation rate is pH-
drug into its optical or geometric isomers. dependent (maximum epimerisation occurring
Since the various isomers of a drug are fre-
quently of different activity, such a conver- H N(CH3)2 (H3C)2N H
sion may be regarded as a form of degradation, OH OH
often resulting in a serious loss of therapeutic
activity. For example, the appreciable loss of
CONH2 CONH2
activity of solutions of adrenaline at low pH
has been attributed to racemisation – the O O

conversion of the therapeutically active form, Partial structure of Partial structure of


4-epi-tetracycline natural tetracycline
in this case the levorotary form, into its less-
active isomer. Scheme 4.7 Epimerisation of tetracyclines.
10_1439PP_CH04 10/11/05 2:53 pm Page 99

The chemical decomposition of drugs 99

at pH 3.2) and is also catalysed by phosphate oxidised to the aldehyde and then isomerised
and citrate ions. to yield 11-cis-retinal (Scheme 4.9), which
Cephalosporin esters are widely used as has a decreased activity compared with the
intermediates in cephalosporin synthesis and all-trans molecule.
as prodrugs for oral administration of par-
enteral cephalosporins. These esters undergo
reversible base-catalysed isomerisation accord- 4.1.4 Photochemical decomposition8
ing to the mechanism shown in Scheme 4.8. A
proton in the 2-position is abstracted by a base Many pharmaceutical compounds, including
(B) and the resulting carbanion can be repro- the phenothiazine tranquillizers, hydrocorti-
tonated in the 4-postion, giving a Δ 2-ester. sone, prednisolone, riboflavin, ascorbic acid
On hydrolysis, Δ 2-cephalosporin esters yield and folic acid, degrade when exposed to light.
Δ 2-cephalosporins, which are biologically As a result there will be a loss of potency of
inactive. the drug, often accompanied by changes in
Cis–trans isomerisation may be a cause of loss the appearance of the product, such as dis-
of potency of a drug if the two geometric coloration or formation of a precipitate.
isomers have different therapeutic activities. Photodecomposition might occur not only
Vitamin A (all-trans-retinol) is enzymatically during storage but also during use of the

O B O O

R1 C NH S
H R1 C NH S
H R1 C NH S
H

HB H
H
N N  N
O R2 O R2 O R2

COOR COOR H COOR

Scheme 4.8 Proposed mechanism for the base-catalysed isomerisation of cephalosporin esters.
Reproduced from W. F. Richter, Y. H. Chong and V. J. Stella, J. Pharm. Sci., 79, 185 (1990).

CH3 CH3
H3C CH3
CH2OH
all-trans-retinol

CH3

Oxidation Isomerization

CH3
H3C CH3

11-cis-retinal
CH3 H3C

C
O H

Scheme 4.9 Isomerisation of vitamin A.


10_1439PP_CH04 10/11/05 2:53 pm Page 100

100 Chapter 4 • Drug stability

product. For example, sunlight is able to pene- photochemical process. In formulations that
trate the skin to a sufficient depth to cause contain low drug concentrations, the primary
photodegradation of drugs circulating in the photochemical reaction follows first-order
surface capillaries or in the eyes of patients kinetics; the kinetics are more complicated at
receiving the drug. higher concentrations and in the solid state
Primary photochemical reaction occurs because most of the light is then absorbed
when the wavelength of the incident light is near the surface of the product.
within the wavelength range of absorption of Although it is difficult to predict which
the drug (usually within the ultraviolet range, drugs are likely to be prone to photodegrada-
unless the drug is coloured), so that the drug tion, there are certain chemical functions that
molecule itself absorbs radiation and degrades. are expected to introduce photoreactivity,
Photodegradation may also occur with drugs including carbonyl, nitroaromatic and N-oxide
that do not directly absorb the incident radi- functions, aryl halides, alkenes, polyenes and
ation, as a consequence of absorption of sulfides.9 The mechanisms of photodegrada-
radiation by excipients in the formulation tion are of such complexity as to have been
( photosensitisers) which transfer the absorbed fully elucidated in only a few cases. We will
energy to the drug, causing it to degrade. In consider two examples – chlorpromazine and
assessing the photostability of a product it is ketoprofen.
therefore necessary to consider the final for- The phenothiazine chlorpromazine (CLP) is
mulation rather than simply the drug itself. rapidly decomposed under the action of ultra-
The rate of the photodegradation is depen- violet light, the decomposition being accom-
dent on the rate at which light is absorbed by panied by discoloration of the solutions
the system and also the efficiency of the (Scheme 4.10).

(CH2)3 N (CH3)2 (CH2)3 N (CH3)2


Ultraviolet
N Cl light N Cl
e 
e 
S S
CLP R

(CH2)3 N (CH3)2

N Cl

2R  CLP

S
P

(CH2)3 N (CH3)2

N Cl

P  H2O  2H

O
CPO

Scheme 4.10 The effect of ultraviolet light on chlorpromazine (CLP).


The first step of the photodegradation is the loss of an electron to yield the semiquinone free radical R. Further stages in
the degradation yield the phenazathonium ion P, which is thought to react with water to yield chlorpromazine sulfoxide
(CPO). The chlorpromazine sulfoxide is itself photolabile and further decomposition occurs. Other products of the
photooxidation include chlorpromazine N-oxide and hydroxychlorpromazine.
Reproduced from F. H. Merkle and C. A. Discher, J. Pharm. Sci., 53, 620 (1964).
10_1439PP_CH04 10/11/05 2:53 pm Page 101

The chemical decomposition of drugs 101

CH3 CH3
N

CH2

CH2

CH2

S N S

N S N

CH2 CH2

CH2 CH2

CH2 CH2

N N
CH3 CH3 CH3 CH3
n

Structure V Polymer produced by the ultraviolet irradiation of chlorpromazine under anaerobic conditions

Chlorpromazine behaves differently result of the liberation of HCl during photo-


towards ultraviolet irradiation under anaero- decomposition.
bic conditions. A polymerisation process has The photodegradation of ketoprofen can
been proposed10 which involves the liberation involve decarboxylation to form an inter-
of HCl in its initial stages. The polymer (V) mediate which then undergoes reduction,
was isolated, and upon intracutaneous injec- or dimerisation of the ketoprofen itself as
tion it produced a bluish-purple discoloration illustrated in Scheme 4.11.
typical of that observed in some patients
receiving prolonged chlorpromazine medica-
Stabilisation against photochemical decomposition
tion. It was suggested that the skin irritation
that accompanies the discoloration may be a Pharmaceutical products can be adequately

O O

COOH (1)

(3) (2)

O OH

H
O

Scheme 4.11 Photodegradation of ketoprofen.


Photodegradation of ketoprofen by decarboxylation (reaction 1) and subsequent reduction (reaction 2), and also by
dimerisation of the ketoprofen (reaction 3).
Reproduced from H. H. Tønnesen, Int. J. Pharm., 225, 1 (2001).
10_1439PP_CH04 10/11/05 2:53 pm Page 102

102 Chapter 4 • Drug stability

protected from photoinduced decomposition formed (Scheme 4.12). The process can con-
by the use of coloured glass containers and tinue to form higher polymers. Such poly-
storage in the dark. Amber glass excludes light meric substances have been shown to be
of wavelength `470 nm and so affords con- highly antigenic in animals and they are con-
siderable protection of compounds sensitive sidered to play a part in eliciting pencilloyl-
to ultraviolet light. Coating tablets with a specific allergic reactions to ampicillin in
polymer film containing ultraviolet absorbers humans. The dimerising tendency of the
has been suggested as an additional method aminopenicillins increases with the increase
for protection from light. In this respect, a film in the basicity of the side-chain group, the
coating of vinyl acetate containing oxyben- order, in terms of increasing rates, being cycla-
zone as an ultraviolet absorber has been cillin << ampicillin ` epicillin ` amoxycillin.
shown11 to be effective in minimising the The hydrate of formaldehyde, HOCH2 OH,
discoloration and photolytic degradation of may under certain conditions polymerise in
sulfasomidine tablets. aqueous solution to form paraformaldehyde,
HOCH2(OCH2)n OCH2 OH, which appears as a
white deposit in the solution. The polymeris-
4.1.5 Polymerisation ation may be prevented by adding to the
solution 10–15% of methanol.
Polymerisation is the process by which two
or more identical drug molecules combine
together to form a complex molecule. It has 4.2 Kinetics of chemical decomposition
been demonstrated that a polymerisation in solution
process occurs during the storage of concen-
trated aqueous solutions of aminopenicillins,
such as ampicillin sodium. The reactive β- Before we can predict the shelf-life of a dosage
lactam bond of the ampicillin molecule is form it is essential to determine the kinetics of
opened by reaction with the side-chain of a the breakdown of the drug under carefully
second ampicillin molecule and a dimer is controlled conditions. Unfortunately, drug

Lactam ring
S S
CH CO NH Hydrolysis CH CO NH

NH2 C N NH2 C HN
O COO O COO
O
NH2
S Dimerisation
Opened lactam ring
CH CO NH

C N
O COO

S
CH CO NH
Ampicillin
NH2 C HN dimer
O COO
NH
Amide link S
CH CO NH

C N
O COO
Scheme 4.12 Dimerisation and hydrolysis of ampicillin.
Reproduced from H. Bundgaard, Acta Pharm. Suec., 13, 9 (1976).
10_1439PP_CH04 10/11/05 2:53 pm Page 103

Kinetics of chemical decomposition in solution 103

decomposition often does not follow simple or


reaction schemes and in this section we will
dx
look not only at the traditional ways of  =  k[A][B] (4.3)
classifying reactions, but also at some of the dt
complications which can arise with pharma- where the proportionality constant, k, is called
ceutical preparations, which confuse this the rate constant. This is an example of a
simple classification. second-order reaction since the order of reac-
tion is the sum of the exponents of the
concentration terms in the rate equation. As
4.2.1 Classifying reactions: the order of we will see (section 4.4.1), many hydrolysis
reaction reactions are catalysed by H!, OH0 or buffer
components and so we can write equation
Reactions are classified according to number (4.3) as, for example,
of reacting species whose concentration deter- dx
mines the rate at which the reaction occurs,  =  k2[A][H +] (4.4)
dt
i.e. the order of reaction. We will concentrate
mainly on zero-order reactions, in which the k2 is a second-order rate constant and has
breakdown rate is independent of the con- units of (concentration)01(time)01, for example
centration of any of the reactants; first-order (mol dm03)01 min01. When the solution is
reactions, in which the reaction rate is deter- buffered at constant pH, [H!] is constant and
mined by one concentration term, and second- we can write equation (4.4) as
order reactions, in which the rate is deter- dx
mined by the concentrations of two reacting  =  k1[A] = k1(a − x) (4.5)
species. dt
Experimentally we can monitor the rate of where k1 # k2[H!]. Since the rate of reaction
breakdown of the drug either by its decrease in now effectively depends on one concen-
concentration with time or alternatively by tration term it is a first-order reaction or, more
the rate of appearance of one of the break- correctly in this case, a pseudo first-order
down products. If we represent the initial con- reaction (see section 4.2.3). The majority of
centration of drug A as a mol dm03 and if we decomposition reactions involving drugs fall
find experimentally that x mol dm03 of the into this category, either because the species
drug has reacted in time t, then the amount of reacting with the drug is maintained constant
drug remaining at a time t is (a 0 x) mol dm03 by buffering or because, as in the case of
and the rate of reaction is uncatalysed hydrolysis reactions, the water is
in such large excess that any change in its
−d[A] −d(a − x) concentration is negligible.
 =  =  dx/dt (4.2)
dt dt If as well as maintaining a constant amount
Notice that the term a is a constant and there- of water in a reaction, we also maintain a
fore disappears during differentiation. We will fixed drug concentration, then equation (4.3)
use dxdt to describe the reaction rate in this becomes
section. dx
If we assume that a typical reaction between  =  k0 (4.6)
dt
a drug molecule A and a reactant B occurs
when two molecules are in collision, then we where k0 # k1[A] # k2[A][B].
might expect that the number of collisions, This type of reaction, which is called a zero-
and hence the reaction rate, would be propor- order reaction, can often occur in suspensions
tional to the concentration of the two reacting of poorly soluble drugs. In these systems the
molecules, i.e. suspended drug slowly dissolves as the drug
decomposes and so a constant drug concen-
Rate ∝ [A][B] tration in solution is maintained.
10_1439PP_CH04 10/11/05 2:53 pm Page 104

104 Chapter 4 • Drug stability

We will now examine the ways in which we 100


can determine the rate constants for these
three types of reaction.
O OH

4.2.2 Zero-order reactions


O CH3

Percentage C9H8O4 remaining


In this type of reaction the decomposition O
proceeds at a constant rate and is independent
80
of the concentrations of any of the reactants.
The rate equation is given by equation (4.6)
as
dx
 =  k0
dt
Integration, noting that x # 0 at t # 0, gives
x t
  dx =  k0   dt (4.7) 60
0 0

i.e.
20 40 50
x =  k0 t (4.8)
Time (days)
A plot of the amount remaining (as ordinate) Figure 4.1 Hydrolysis of a suspension of acetylsalicylic
against time (as abscissa) is linear with a slope acid at 34°C.
of k0 (concentration·time01). Reproduced from K. C. James, J. Pharm. Pharmacol., 10, 363 (1958) with
Many decomposition reactions in the solid permission.

phase (section 4.3) or in suspensions appar-


ently follow zero-order kinetics. Figure 4.1 Or, rearranging into a linear form,
shows the hydrolysis of a suspension of acetyl- 2.303 2.303
salicylic acid. t =   log a −  log(a − x) (4.11)
k1 k1
According to equation (4.11), a plot of the log-
4.2.3 First-order reactions arithm of the amount of drug remaining (as
ordinate) as a function of time (as abscissa) is
The rate of first-order reactions is determined linear if the decomposition follows first-order
by one concentration term and may be kinetics. The first-order rate constant may be
written using equation (4.5) as obtained from the slope of the plot (slope #
0k12.303). k1 has the dimensions of time01.
dx The time taken for half of the reactant to
 =  k1(a − x)
dt decompose is referred to as the half-life of the
reaction, t0.5. An expression for t0.5 for a first-
Since x # 0 at the start of the measurements order reaction may be derived from equa-
(that is, when t # 0) tion (4.10), noting that when t # t0.5, x # a2:
x t
dx
  
(a − x)
= k1   dt (4.9) t0.5 =
2.303
 log 
a
(4.12)
0 0 k1 a/2
Thus,
2.303 a 2.303 0.693
k1 =  log  (4.10) t0.5 =  log 2 = (4.13)
t a−x k1 k1
10_1439PP_CH04 10/11/05 2:53 pm Page 105

Kinetics of chemical decomposition in solution 105

The half-life is therefore independent of the Show that the hydrolysis follows first-order
initial concentration of reactants. kinetics and calculate (a) the rate constant,
Even in the case of a reaction involving more and (b) the half-life.
than one reacting species, the rate may still
follow first-order kinetics. The most common Answer
example of this occurs when one of the reac- (a) The reaction will be first-order if a plot
tants is in such a large excess that any change of the logarithm of the amount of
in its concentration is negligible compared homatropine remaining against time is
with changes in the concentration of the other linear.
reactants. This type of reaction is termed a
pseudo first-order reaction. Such reactions are Log percentage 1.97 1.93 1.88 1.80 1.72 1.62
often met in stability studies of drugs that remaining
Time(h) 1.38 3.0 6.0 8.6 12 17
hydrolyse in solution, the water being in such
excess that changes in its concentration are Figure 4.2 shows a linear plot with a
negligible and hence the rate of reaction is slope # 0(1.96 0 1.55)(20 0 2) # 02.278
dependent solely on the drug concentration. " 1002 h01

Slope = −k1/2.303
EXAMPLE 4.1 Calculation of first-order rate con-
stant and half-life Therefore,

The following data were obtained for the k1 = 5.25 × 10 −2 h −1
hydrolysis of homatropine in 0.226 mol dm03
(b) From equation (4.13)
HCl at 90°C:
Percentage 93.4 85.2 75.9 63.1 52.5 41.8 t0.5 = 0.693/k1 = 13.2 h
homatropine
remaining The half-life of the reaction is 13.2 h.
Time (h) 1.38 3.0 6.0 8.6 12 17

2.0

Me O
1.9 N
OH
O
Log percentage ester

1.8 Ph

1.7

1.6

1.5

2 4 6 8 10 12 14 16 18 20
Time (h)

Figure 4.2 First-order plot for hydrolysis of homatropine in hydrochloric acid (0.226 mol dm03) at 90°C.
Data from M. H. Krasowska, S. Schytt Larsen and K. Ilver, Dansk. Tidsskr. Farm., 42, 170 (1968) with permission.
10_1439PP_CH04 10/11/05 2:53 pm Page 106

106 Chapter 4 • Drug stability

4.2.4 Second-order reactions yields


1 1 1 x
The rate of a second-order reaction is deter-
mined by the concentrations of two reacting
species. The general rate equation is given by
t = 

  −  =
k2 a − x a

k2 a(a − x)
from which it is seen that a plot of t (ordinate)
(4.20)

equation (4.3) as against xa(a 0 x) (abscissa) yields a linear plot


dx of slope 1k2 .
 =  k2[A][B] (4.14) The half-life of a reaction which follows
dt equation (4.20) is given by
If the initial concentrations of reactants A and 1
B are a and b, respectively, equation (4.14) t0.5 = (4.21)
may be written k2 a

dx Unlike t0.5 for the first-order reactions, the


 =  k2(a − x)(b − x) (4.15) half-life of the second-order reaction is depen-
dt dent on the initial concentration of reactants.
where x is the amount of A and B decomposed It is not possible to derive a simple expression
in time t. Integration of equation (4.15) by the for the half-life of a second-order reaction
method of partial fractions yields with unequal initial concentrations.

2.303 b(a − x)
k2 =  log  (4.16)
t(a − b) a(b − x) 4.2.5 Third-order reactions
Rearranging into a linear form suitable for
plotting gives Third-order reactions are only rarely encoun-
tered in drug stability studies involving, as
2.303 b 2.303 (a − x) they do, the simultaneous collision of three
t =  = log  +  log 
k2(a − b) a k2(a − b) (b − x) reactant molecules. The overall rate of ampi-
(4.17) cillin breakdown by simultaneous hydrolysis
and polymerisation may be represented by an
k2 can then be obtained from the slope, 2.303 equation of the form
k2(a 0 b), of a plot of t (as ordinate) against
log[(a 0 x)(b 0 x)] (as abscissa). −d[A]
 =  ka[A] + kb[A] 2 + kc[A] 3 (4.22)
Examination of equation (4.17) shows that dt
the second-order rate constant is dependent where ka, kb and kc are the pH-dependent
on the units used to express concentration; apparent rate constant for hydrolysis, uncata-
the units of k2 are concentration01 time01. lysed polymerisation and the general acid–
For reactions in which both concentration base-catalysed polymerisation of ampicillin,
terms refer to the same reactant we may write respectively.12 As seen from equation (4.22)
−d[A] the decomposition rate shows both second-
 =  k2[A] 2 (4.18) order and third-order dependency on the total
dt
ampicillin concentration [A].
and
dx
 =  k2(a − x) 2 (4.19) 4.2.6 Determination of the order of reaction
dt
A similar equation applies to second-order The most obvious method of determining the
reactions in which the initial concentrations order of a reaction is to determine the amount
of the two reactants are the same. of drug decomposed after various intervals
Integration of equation (4.19) between the and to substitute the data into the integrated
limits of t from 0 to t and of x from 0 to x equations for zero-, first- and second-order
10_1439PP_CH04 10/11/05 2:53 pm Page 107

Kinetics of chemical decomposition in solution 107

reactions. The equation giving the most con- The intercept of the graph (that is, the value
sistent value of k for a series of time intervals is of log t0.5 at log C0 # 0) is 2.60. Thus, from
that corresponding most closely to the order equation (4.23),
of the reaction. Alternatively, the data may be
log[(2 − 1)/k] = 2.60
displayed graphically according to the linear
equations for the various orders of reactions and
until a straight-line plot is obtained. Thus, for
k =  2.51 × 10 −3 (mol dm −3) −1 min −1
example, if the data yield a linear graph when
plotted as t against log(a 0 x) the reaction is
then taken to be first-order.
Fitting data to the standard rate equations
may, however, produce misleading results if a 4.2.7 Complex reactions
fractional order of reaction applies. An alter-
native method of determining the order of There are many examples of drugs in which
reaction, which avoids this problem, is based decomposition occurs simultaneously by two
on equation (4.23): or more pathways, or involves a sequence of
decomposition steps or a reversible reaction.
2 n −1 − 1

 
log t0.5 = log + (1 − n) log C0 Indeed, the degradative pathways of some
k(n − 1) drugs include examples of each of these types
(4.23) of complex reactions. Modification of the
rate equations is necessary whenever such
The half-life of the reaction is determined for a reactions are encountered.
series of initial drug concentrations, C0 , and
the order, n, is calculated from the slope of
plots of log t0.5 as a function of log C0 . Reversible reactions
Treatment of the kinetics of a reversible reac-
tion involves two rate constants; one, kf, to
EXAMPLE 4.2 Determination of the order of describe the rate of the forward reaction and
reaction
the other, kr, to describe the rate of the reverse
The kinetics of decomposition of a drug in reaction. For the simplest example in which
aqueous solution were studied using a series of both of these reactions are first-order, that is
solutions of different initial drug concentra- kf
A g
k
B
tions, C0 . For each solution the time taken for r

half the drug to decompose (that is, t0.5) was the rate of decomposition of reactant is
determined with the following results:
−d[A]
C0 (mol dm03) 4.625 1.698 0.724 0.288  =  kf[A] − kr[B] (4.24)
t0.5 (min) 87.17 240.1 563.0 1414.4 dt
Determine the order of reaction and calculate The integrated form of the rate equation is
the rate constant.
2.303 A0 − Aeq
t =   log  (4.25)
Answer kf + kr A − Aeq
Application of equation (4.23) requires values where A0, A and Aeq represent the initial con-
for log C0 and log t0.5; thus centration, the concentration at time t and
log C0 0.665 0.230 00.140 00.540 the equilibrium concentration of reactant A,
log t0.5 1.94 2.38 2.75 3.15 respectively. Equation (4.25) indicates that a
A plot of log t0.5 against log C0 is linear (Fig. plot of t (as ordinate) against log[(A0 0 Aeq)
4.3) with slope (1 0 n) # 01.01. Hence n # 2.01, (A 0 Aeq)] should be linear with a slope of
i.e. the reaction is second-order. 2.303(kf ! kr). kf and kr may be calculated
10_1439PP_CH04 10/11/05 2:53 pm Page 108

108 Chapter 4 • Drug stability

3.4

3.2

3.0

2.8
Log t0.5

2.6

2.4

2.2

2.0

0.6 0.4 0.2 0 0.2 0.4 0.6 0.8


Log C0

Figure 4.2 Example 4.2: plot of log of half-life (t0.5) against log of initial drug concentration (C0).

separately if the equilibrium constant K is also In other cases decomposition may occur
determined, since simultaneously by two different processes, as
Beq 1 − Aeq kf in the simultaneous hydrolysis and epimerisa-
K =  = = (4.26) tion of pilocarpine (see Example 4.3).
Aeq Aeq kr The overall rate equation for a parallel reac-
where Beq is the equilibrium concentration of tion is the sum of the constants for each
product B. pathway. For example, for a decomposition of
The epimerisation of tetracycline (see section a drug X involving two pathways, each of
4.1.3) is an example of a first-order reversible which is first-order,
decomposition reaction. kA A

X
Parallel reactions
kB B
The decomposition of many drugs involves
two or more pathways, the preferred route of the rate equation is
reaction being dependent on reaction condi- −d[X]
tion. Nitrazepam (VI) decomposes in two  =  (kA + kB)[X] = kexp[X] (4.27)
pseudo first-order parallel reactions giving dif- dt
ferent breakdown products in solution and in where kA and kB are the rate constants for the
the solid state, as illustrated in Scheme 4.13. formation of A and B, respectively, and kexp is
Decomposition of nitrazepam tablets in the the experimentally determined rate constant.
presence of moisture will occur by both routes, Values of the rate constants kA and kB may be
the ratio of the two products being dependent evaluated separately by determining the ratio
on the amount of water present. R of the concentration of products formed by
10_1439PP_CH04 10/11/05 2:54 pm Page 109

Kinetics of chemical decomposition in solution 109

NH2

O
O2N C

aq.
H
O
N C
(VII)
CH2

O2N C N H

solid
N O
state C

O2N C NH2
(VI)

(VIII)

Scheme 4.13 Simplified decomposition scheme for nitrazepam (VI) in the solid state and aqueous solution. The main
decomposition product is 2-amino-5-nitrobenzophenone (VII) in aqueous solution and 3-amino-6-nitro-4-phenyl-2(1H )-
quinolone (VIII) in the solid state.
Reproduced from W. Meyer, S. Erbe and R. Voigt, Pharmazie, 27, 32 (1972).

each reaction: 6.96 " 102 (mol dm 03)01 h01. Analysis has shown
[A] kA that at equilibrium the percentage of the
R =  = (4.28) epimerised form of pilocarpine (isopilocarpine)
[B] kB
at 25°C is 20.6%. Calculate the rate constants
Since for hydrolysis, kH, and epimerisation, kE.
kexp = kA + kB (4.29)
Answer
kB The ratio R of pilocarpine to isopilocarpine is
kexp = kA + (4.30)
R
R =  79.38/20.62 = 3.85
Solving for kA gives
From equation (4.31),
R
kA = kexp  (4.31) kH = (6.96 × 10 2)(3.85/4.85)
(R + 1)
i.e.
Similarly,
kexp kH = 5.48 × 10 2 (mol dm −3) −1 h −1
kB = (4.32)
(R + 1) From equation (4.32),
kE = (6.96 × 10 2)/4.85
EXAMPLE 4.3 Calculation of rate constants of i.e.
parallel reactions
kE = 1.44 × 10 2 (mol dm −3) −1 h −1
Pilocarpine has been shown to undergo simul- Thus the rate constants for hydrolysis and
taneous hydrolysis and epimerisation in epimerisation are 5.48 " 102 and 1.44 " 102
aqueous solution. The experimentally deter- (mol dm03)01 h01, respectively.
mined rate constant, kexp, at 25°C is
10_1439PP_CH04 10/11/05 2:54 pm Page 110

110 Chapter 4 • Drug stability

Consecutive reactions Since, at any time,


The simplest example of a consecutive reaction [A]0 = [A] + [B] + [C] (4.39)
is that described by a sequence
then
kA kB
A 2 B 2  C [C] =  [A]0 − [A] − [B]
where each step is a nonreversible first-order 1
reaction. The hydrolysis of chlordiazepoxide
follows a first-order decomposition scheme
similar to that described in this equation

 =  [A]0 × 1 +  (k  e −kAt − kA e −kBt)
kA − kB B
(4.40)

(Scheme 4.14). Equations (4.36), (4.37) and (4.38) may be
The rate of decomposition of A is used to estimate the rate constants kA and kB
−d[A] and also the concentration of the breakdown
 =  kA[A] (4.33) product C.
dt
The rate of change of concentration of B is
d[B] 4.3 Solid dosage forms: kinetics of
 =  kA[A] − kB[B] (4.34)
dt chemical decomposition
and that of C is
In spite of the importance of solid dosage
d[C]
 =  kB[B] (4.35) forms, there have been relatively few attempts
dt to evaluate the detailed kinetics of decompo-
Integration of the rate equation (4.33) yields sition. Most of the earlier work was carried out
with the sole objective of predicting stability,
[A] = [A]0 e −kAt (4.36) and data were treated using the rate equations
Substitution of equation (4.36) into equa- derived for reaction in solution. More
tion (4.34) gives recently, the mechanisms that were developed
to describe the kinetics of decomposition of
d[B] pure solids have been applied to pharma-
= kA[A]0 e −kAt − kB[B] (4.37)
dt ceutical systems and some rationalisation of
decomposition behaviour has been possible. A
which upon integration gives
comprehensive account of this topic has been
kA[A]0 presented by Carstensen13, 14 on which the
[B] =  [e −kAt − e −kB t] (4.38) following summary is based.
(kB − kA)

NHCH3 H O
N N NH2
HOH HOH
H, HA, A H O
Cl N Cl N Cl C
O O

(IX) or (IXHⴙ) (X) (XI)

Scheme 4.14 Decomposition scheme for chlordiazepoxide. The neutral or cationic chlordiazepoxide (IX or IXH!) is
transformed to the lactam (X) and, finally, in acidic solutions, to the yellow benzophenone (XI).
Reproduced from H. V. Maudling et al., J. Pharm. Sci., 64, 278 (1975).
10_1439PP_CH04 10/11/05 2:54 pm Page 111

Solid dosage forms: kinetics of chemical decomposition 111

It is convenient to divide single component For the contracting sphere model,


systems into two categories: those solids that
k
decompose to a solid product and a gas, and (1 − x) 1/3 = 1 −  t (4.42)
those that decompose to give a liquid and a gas. r0
There are a few pharmaceutical examples of
Solids that decompose to give a solid and a gas compounds that decompose by topochemical
reaction. Thus, the decomposition of aspirin
An example of this category is p-aminosali- at elevated temperatures has been shown to
cylic acid, which decomposes to a solid conform to equation (4.41) (Fig. 4.5).
( p-aminophenol) and a gas (carbon dioxide): A similarity between equations (4.41) and
NH2 C6 H3(OH)COOH 2  (4.42) and the first-order rate equations was
NH2 C6 H4 OH + CO2 pointed out by Carstensen,13 who suggested
that this similarity might account for the fact
The decomposition curves which result from that many decompositions in solid dosage
such a reaction show either (a) an initial rapid forms appear to follow first-order kinetics.
decomposition followed by a more gradual The sigmoidal decomposition curves can be
decomposition rate, or (b) an initial lag period, interpreted using the Prout and Tompkins
giving a sigmoidal appearance. The shape pro- model. This model assumes that the decom-
duced by (a) can usually be accounted for by position is governed by the formation and
topochemical (or contracting geometry) reac- growth of active nuclei which occur on the
tions and that produced in (b) by nucleation surface as well as inside the crystals. The for-
theories. mation of product molecules sets up further
The model used in the treatment of topo- strains in the crystal since the surface array of
chemical decomposition is that of a cylinder product molecules has a different unit cell
or sphere (Fig. 4.4), in which it is assumed that from the original substance. The strains are
the radius of the intact chemical substance relieved by the formation of cracks. Reaction
decreases linearly with time. For the con- takes place at the mouth of these cracks owing
tracting cylinder model, the mole fraction x to lattice imperfections and spreads down into
decomposed at time t is given by the crevices. Decomposition on these surfaces
k produces further cracking and so the chain
(1 − x) 1/2 = 1 −  t (4.41) reaction spreads.
r0
The equation proposed to describe decom-
position by this process is of the form
x k

 
Decomposed
ln =  t + C (4.43)
(1 − x) r0
where C is a lag-time term.
ro The decomposition curves of p-aminosali-
cylic acid are sigmoidal (see Fig. 4.6) and linear
r
plots are produced when the data are plotted
according to equation (4.43). Stability meas-
urements made inadvertently in the lag
periods of this type of decomposition would
Intact
suggest zero-order kinetics.

Solids that decompose to give a liquid and a gas

Figure 4.4 Model of a sphere or a cylinder used in An example of a solid in this category is p-
theoretical treatment of topochemical reactions. aminobenzoic acid, which decomposes into
Reproduced from J. T. Carstensen, J. Pharm. Sci., 63, 1 (1974). aniline and carbon dioxide. Decomposition
10_1439PP_CH04 10/11/05 2:54 pm Page 112

112 Chapter 4 • Drug stability

0.12
47.5C

0.10
50C

0.08
1(1x)1/2

0.06

0.04

0.02

1 2 3 4
Time (days)

Figure 4.5 Decomposition of aspirin at elevated temperatures in tablets containing sodium bicarbonate plotted according
to equation (4.41).
Reproduced from E. Nelson et al., J. Pharm. Sci., 63, 755 (1974) with permission.

80C 75 C
60

50
Percentage decomposition

40
70C

30
O OH

20
OH

10
H2N

80 160 240 320 400 480 560 640 720


Time (h)

Figure 4.6 Degradation of powdered p-aminosalicylic acid in a dry atmosphere at elevated temperatures.
Reproduced from S. S. Kornblum and B. J. Sciarrone, J. Pharm. Sci., 53, 935 (1964) with permission.
10_1439PP_CH04 10/11/05 2:54 pm Page 113

Factors influencing drug stability 113

OH
145 C
40
140C H2N

135C
Decomposition (mole %)

30

20

10

130C

100 200 300


Time (h)

Figure 4.7 Decomposition curves of p-aminobenzoic acid.


Reproduced from J. T. Carstensen and M. N. Musa, J. Pharm. Sci., 61, 1113 (1972).

causes a layer of liquid to form around the dosage forms separately, although, as we will
solid which dissolves the solid. The decom- see, there are similarities in the influence of
position curves show an initial lag period several factors on drug breakdown in both.
(Fig. 4.7), which corresponds to the establish-
ment of the liquid layer. Beyond this region,
the plot represents the first-order decomposi- 4.4.1 Liquid dosage forms
tion of the solid in solution in its liquid
decomposition products. There are thus two pH
rate constants, that for the initial decomposi-
tion of the solid itself, and that for the decom- pH is perhaps the most important parameter
position of the solid in solution. which affects the hydrolysis rate of drugs in
liquid formulations; it is certainly the one
which has been most widely examined.
Studying the influence of pH on degradation
4.4 Factors influencing drug stability rate is not as simple as might at first be imag-
ined. If the hydrolysis rate of the drug in a
Before we can suggest ways in which we might series of solutions buffered to the required pH
prevent the decomposition of drugs, we is measured and the hydrolytic rate constant is
should first consider the various factors that then plotted as a function of pH, a pH–rate
accelerate the decomposition processes. For profile will be produced, but this will almost
convenience we will consider liquid and solid certainly be influenced by the buffers used to
10_1439PP_CH04 10/11/05 2:54 pm Page 114

114 Chapter 4 • Drug stability

prepare the solutions. It is probable that a For a complete evaluation of the stability of
different pH–rate profile would be obtained the drug, we need to evaluate the catalytic
using a different buffer. To understand why coefficients for specific acid and base catalysis
this should be, we have to consider not only and also to determine the catalytic coefficients
the catalytic effect of hydrogen and hydroxyl of possible buffers which we might wish to use
ions, which is called specific acid–base catalysis, in the formulation.
but also the possible accelerating effect of the First we will examine how to achieve a
components of the buffer system, which we buffer-independent pH–rate profile, since this
refer to as general acid–base catalysis. These two will show us at which pH the stability is great-
types of acid–base catalysis can be combined est. By way of illustration we can consider a
together in a general expression as follows: specific example of a stability study which
kobs = k0 + kH+[H +] + kOH−[OH −] has been reported for the antihypertensive
vasodilator ciclosidomine. Experiments carried
+ kHX[HX] + kX−[X −] (4.44)
out at constant temperature and constant
In this equation, kobs is the experimentally ionic strength using a series of different buffers
determined hydrolytic rate constant, k0 is the over the pH range 3–6 produced the graphs
uncatalysed or solvent catalysed rate constant, shown in Fig. 4.8. These plots show that an
kH+ and kOH− are the specific acid- and base- increase of buffer concentration, particularly at
catalysis rate constants respectively, kHX and pH 3, had a marked effect on the hydrolysis
kX− are the general acid- and base-catalysis rate rate. The effect of the phosphate buffer on this
constants respectively, and [HX] and [X0] system became less pronounced with increase
denote the concentrations of protonated and of pH and was found to have a negligible effect
unprotonated forms of the buffer. above pH 7.5.

0.0050
pH 3
(phosphate) O
0.0045

0.0040 N

N
0.0035 N
Observed rate constant (h1)

O
0.0030
N
pH 4
0.0025 (acetate) C O
pH 5
0.0020 (acetate)

pH 6
0.0015 (phosphate)

0.0010

0.0005

0.0000
0.00 0.02 0.04 0.06 0.08 0.10 0.12
Buffer concentration (mol dm3)

Figure 4.8 Effect of buffer concentration on the hydrolytic rate constant for ciclosidomine at 60°C as a function of pH.
Reproduced from C. F. Carney, J. Pharm. Sci., 76, 393 (1987) with permission.
10_1439PP_CH04 10/11/05 2:54 pm Page 115

Factors influencing drug stability 115

To remove the influence of the buffer, the [OH0] at high pH, the gradient will be the
reaction rate should be measured at a series of rate constant for base-catalysed hydrolysis.
buffer concentrations at each pH and the data Example 4.4 illustrates the calculation of these
extrapolated back to zero concentration as catalytic coefficients.
shown in Fig. 4.8. If these extrapolated rate
constants are plotted as a function of pH, the
required buffer-independent pH–rate profile E X A M P L E 4 . 4 Calculation of rate constants for
will be obtained. Figure 4.9 illustrates the base-catalysed hydrolysis
simple type of pH–rate profile which is
obtained with codeine sulfate. The following data were obtained for the
As we can see from Fig. 4.9, this drug is very hydrolytic rate constant, kobs of codeine sulfate
stable in unbuffered solution over a wide pH in aqueous buffer-free solution at 80°C
range but degrades relatively rapidly in the 107kobs (s01) 5.50 4.40 2.30 1.25.0 0.70
presence of strong acids or bases. Since the pH 11.63 11.53 11.23 10.93 10.63
influence of buffer components has been
Determine graphically (a) the catalytic coeffi-
removed, this plot allows us to calculate the
cient for base-catalysis, kOH−, and also (b) the
rate constants for specific acid and base cataly-
coefficient for solvent catalysis, k0.
sis. Removing the terms for the effect of buffer
from equation (4.44), we have Answer
kobs = k0 + kH+[H +] + kOH−[OH −] (4.45) At high pH,

and consequently a plot of measured rate con- kobs = k0 + kOH−[OH −]


stant kobs against the hydrogen ion concentra- where [OH0] is calculated from
tion [H!] at low pH will have a gradient equal
to the rate constant for acid catalysis. p[OH −] = −log[OH −] = 12.63 − pH
Similarly, of course, if we plot kobs against (Note: pKw # 12.63 at 80°C.)

4

5

6
MeO
Log k (s1)

7
Me
N

O
8

H
9 HO

10

0 4 8 12
pH

Figure 4.9 Log rate–pH profile for the degradation of codeine sulfate in buffer-free solutions at 60°C.
Reproduced from M. F. Powell, J. Pharm. Sci., 75, 901 (1986) with permission.
10_1439PP_CH04 10/11/05 2:54 pm Page 116

116 Chapter 4 • Drug stability

A plot of kobs against [OH0] has a gradient of [H2 PO−4/BT], will have an intercept at
kOH− and an intercept of k0. [H2 PO−4/BT] = 0 equal to kHPO42−. Furthermore,
From a graph of the above data, the k, value at [H2 PO−4/BT] = 1 is the other
catalytic coefficient, kH2PO4−.
k0 =  2.0 × 10 −8 s −1
kOH− =  5.2 × 10 −6(mol dm −3) −1 s −1
EXAMPLE 4.5 Calculation of the catalytic coeffi-
cients for buffer species
The degradation of codeine is particularly
susceptible to the effects of buffers. Its hydroly- The following data were obtained for the
sis rate in 0.05 mol dm03 phosphate buffer hydrolytic rate constant k of codeine sulfate at
at pH 7 is almost 20 times faster than in 100°C in phosphate buffers of varying total
unbuffered solution at this pH, so this is a concentration BT at pH values of 6 and 8:
good drug to use as an example of the deter-
mination of the influence of buffer compo- BT (mol dm03) 00.03 00.06 00.09 00.12
107k (s01) at pH 6 6.1 11.20 16.30 21.40
nents on the breakdown rate.
107k (s01) at pH 8 12.90 25.00 37.00 49.00
In phosphate buffers of neutral pH, the
major buffer species are H2 PO−4 and HPO2− 4 , If the fraction of [H2 PO−4] present in buffer
either of which may act as a catalyst for solutions at pH 6 is 0.74 and at pH 8 is 0.23,
codeine degradation. To find out which of determine graphically the catalytic coeffi-
these is the stronger catalyst, we can treat the cients for the buffer species (a) H2 PO−4 and
experimental data in the following way. In (b) HPO2−
4 .
neutral pH solutions we can write the follow-
ing expression for the observed rate constant, Answer
From equation (4.46), a plot of kobs against BT
kobs = k0 + kH2PO4−[H2 PO−4] + kHPO42−[HPO2−
4 ] has an intercept k0 and a gradient k,. Therefore
(4.46) plot kobs against BT from the given data at each
or pH and measure the gradient of the graph.
kobs = k0 + k’BT From the graph:

where kH2PO4− and kHPO42− are the rate constants k, at pH 6 =  1.7 × 10 −5 (mol dm −3) −1 s −1
for catalysis by H2 PO−4 and HPO2− 4 ions respec-
k, at pH 8 =  4.0 × 10 −5 (mol dm −3) −1 s −1
tively, and BT is the total concentration of
From equation (4.47), a plot of k, against the
phosphate buffer. Notice that the terms for fraction of acidic buffer component,
specific acid- and base-catalysis have little [H2 PO−4]/BT, has an intercept of kHPO42−.
effect at this pH and we need not consider Also when [H2 PO−4]/BT = 1,
them in this treatment.
From equation (4.46), a plot of kobs against k, =  kH2PO4−
BT will have an intercept k0 and a gradient k,.
From the graph:
To find values for the catalytic coefficients, we
rearrange the equation into the following kHPO42− =  5.1 × 10 −5 (mol dm −3) −1 s −1
linear form:
kH2PO4− =  0.5 × 10 −5 (mol dm −3) −1 s −1
(kobs − k0) kH2PO−4[H2 PO−4]
k, =  =
BT BT
The relationship between the ability of a
BT − [H2 PO−4]

 
 + kHPO42− (4.47) buffer component to catalyse hydrolysis,
BT denoted by the catalytic coefficient, k, and its
dissociation constant, K, may be expressed by
We can now see that a second plot of the
the Brønsted catalysis law as
apparent rate constant k, against the fraction
of the acid buffer component present, i.e. kA = aKαAfor a weak acid (4.48)
10_1439PP_CH04 10/11/05 2:54 pm Page 117

Factors influencing drug stability 117

and differing susceptibility of the unionised and


ionised forms of the drug molecule to
kB = bKβBfor a weak base (4.49)
hydrolysis.
where a, b, α and β are constants characteristic By way of illustration we will look at the case
of the solvent and temperature. α and β are of the hydrolysis of mecillinam (XII), which is
positive and vary between 0 and 1. an antimicrobially active amidopenicillamic
In our treatment of the degradation of acid. This amphoteric drug can exist as a
codeine sulfate we have not yet considered cation, which we can write as MH+2, as a
any effect which changes in its ionisation zwitterion MH& or as an anion M0. Figure 4.10
might have on its stability. Codeine has a pKa shows the pH–rate profile at zero buffer con-
of 8.2 at 25°C and so its ionisation state will centration. The reason this plot is so much
change over the pH range 6–10. With this more complex than that of codeine sulfate is
particular drug, the stability was not affected that each of the species present in solution can
by any such changes. This is not the case with undergo specific acid–base catalysis to varying
many drugs, however, and complex pH–rate extents and so each contributes to the overall
profiles are often produced because of the profile shown in Fig. 4.10.

0.8

0.4

0.4

0.8
Log k (h1)

1.2

1.6

2.0

2.4

2.8

0 2 4 6 8 10 12 14
pH

Figure 4.10 Log k–pH profile for the degradation of mecillinam in aqueous solution at 35°C (ionic strength #
0.5 mol kg01), where k is the apparent first-order rate constant for degradation in buffer-free solutions or in buffers showing
no effect on rate of degradation.
Reproduced from C. Larsen and H. Bundgaard, Arch Pharm. Chem., 5, 66 (1977).
10_1439PP_CH04 10/11/05 2:54 pm Page 118

118 Chapter 4 • Drug stability

we can for mecillinam, we can still, of course,


S choose the pH at which to buffer the drug
N CH N
solution for maximum stability. In the case of
N mecillinam this would be between pH 4 and 6.
O COOH
We should also note that the oxidative degra-
dation of some drugs in solution may be pH-
Structure XII Mecillinam dependent; for example, the oxidation of
prednisolone is base-catalysed. Similarly, the
The various reactions which are thought to oxidation of morphine occurs more rapidly in
occur are alkaline or neutral solution than in acid solu-
kH
MH+2 + H + 2  products 1 tion. The reason for this may be the effect of
k’H
 pH on the oxidation–reduction potential, E0,
MH & + H + 2 products 2 of the drug.
k0
&
MH + H2 O 2  products 3 The photodegradation of several drugs is

k’OH

also pH-dependent. For example, the photo-
MH & + OH − 2 products 4 chemical decomposition of the benzodiazepine
kOH
− −
M + OH  2  products 5 (4.50) derivative midazolam (XIII) increases with
pH,15 and ciprofloxacin (XIV) is most sensitive
The part of the pH–rate profile at very low pH to photodegradation at slightly basic pH where
(below pH 1.5) is given entirely by reaction the drug is in zwitterionic form, the stability
(1). This is because the mecillinam exists as increasing when the pH is lowered to 3–416
the cation MH+2, the hydrolysis of which will (Fig. 4.11).
be acid-catalysed over this pH range. The
shoulder in the pH–rate profile at around pH 2 Temperature
coincides approximately with the first pKa
value of mecillinam and indicates an increased Increase in temperature usually causes a very
acid-catalysis of the zwitterion relative to the pronounced increase in the hydrolysis rate of
cationic species. The decrease of hydrolysis
rate constant with increasing pH up to pH 4 Me
N
can be explained by assuming that both
reactions (1) and (2) are occurring. We can see N

from Fig. 4.10 that the hydrolysis rate is


almost constant between pH 4 and 6, and this Cl
suggests that the hydrolysis is water-catalysed N
(reaction 3) over this pH region. The hydro- F
lysis rate now starts to increase with increasing
pH, which indicates that base-catalysis is now
the dominating factor. Between pH 6.5 and
the pKa for ionisation of the amidino side-
chains (pKa2 # 8.79), it is reaction (4) which Structure XIII Midazolam
describes the hydrolysis rate. The plot changes
slope at around pH 8 because it is affected by O O
the changes which are occurring in the state of F
ionisation of the amidino side-chain. Above OH
pH 12, the mecillinam exists in solution as the
anion and this final part of the graph is N N
described entirely by the base-catalysis of this HN
species (reaction 5).
Even though we may not always be able to
explain the pH–rate profile as completely as Structure XIV Ciprofloxacin
10_1439PP_CH04 10/11/05 2:54 pm Page 119

Factors influencing drug stability 119

100
4

80
pH 4.0
5
Ciprofloxacin (%)

60
6
pH 5.0
pH 10.6

Log k (s1)
40 pH 6.0 7 100C

80 C

20 8
pH 8.6 60 C

9
0
0 20 40 60 80 100 120 140
Time (min)
10 25 C
Figure 4.11 The effect of pH on the photodegradation of
ciprofloxacin. Radiation source: mercury lamp at wave-
length of 313 nm.
Reproduced from K. Torniainen, S. Tammilehto and V. Ulvi, Int. J. Pharm., 132, 0 4 8 12
pH
53 (1996).
Figure 4.12 Log rate–pH profiles for the degradation of
drugs in solution, a fact which is used to good codeine sulfate in buffer-free solutions at several tempera-
tures. The dashed line is the log rate–pH profile calculated
effect in the experimental studies of drug
from the Arrhenius equation.
stability described above. Such studies are Reproduced from M. F. Powell, J. Pharm. Sci., 75, 901 (1986).
usually carried out at high temperatures, say
60 or 80°C, because the hydrolysis rate is reactant molecules collide. A is the frequency
greater at these temperatures and can there- factor and this is assumed to be independent
fore be measured more easily. Of course, if a of temperature for a given reaction. R is the gas
formulation has to be heat sterilized then its constant (8.314 J mol01 K01) and T is the tem-
stability will, in any case, have to be measured perature in kelvins. We can see from equation
at elevated temperatures. Figure 4.12 shows (4.51) that a plot of the log of the rate constant
the pH–rate profiles for the degradation of k against the reciprocal of the temperature
codeine sulfate at several temperatures and should be linear with a gradient of 0Ea2.303R.
also the calculated values at 25°C. We will Therefore, assuming that there is not a change
now see how these calculated values can be in the order of the reaction as the temperature
obtained. is changed, we can extrapolate plots of log k
The equation which describes the effect of against 1T to any required temperature and
temperature on decomposition, and which so determine the rate of breakdown at that
shows us how to calculate the rate of break- temperature. We can also, of course, calculate
down at room temperature from measure- the activation energy from the gradient of this
ments at much higher temperatures, is the plot. Figure 4.13 shows Arrhenius plots for the
Arrhenius equation. breakdown of the drug ciclosidomine at
Ea several pH values.
log k =  log A − (4.51) When it is clear from stability determi-
2.303RT
nations that a drug is particularly unstable at
In this equation, Ea is the activation energy, room temperature, then of course it will need
that is the energy barrier which has to be over- to be labelled with instructions to store in a
come if reaction is going to occur when two cool place. This is the case, for example, with
10_1439PP_CH04 10/11/05 2:54 pm Page 120

120 Chapter 4 • Drug stability

0.1
0.06
0.03

0.01

Observed rate constant (h1)


0.006
0.003 pH 1

0.001
0.0006
0.0003

pH 3
0.0001
0.00006
0.00003
pH 6

0.00001
0.00290 0.00305 0.00320 0.00335
1/T (K1)

Figure 4.13 Arrhenius plots for the hydrolysis of ciclosidomine in buffer solutions at several pH values.
Reproduced from C. F. Carney, J. Pharm. Sci., 76, 393 (1987).

injections of penicillin, insulin, oxytocin and Ionic strength


vasopressin.
We often need to add electrolytes to drug solu-
tions, for example to control their tonicity.
Consequently we must pay particular atten-
EXAMPLE 4.6 Calculation using the Arrhenius
tion to any effect they may have on stability.
equation
In fact, the stability experiments that we con-
The following values were determined for the sidered earlier in this section should all be
specific acid-catalytic constants kH+ for an anti- carried out at constant electrolyte concen-
inflammatory drug: tration to avoid any confusion arising from
possible differences in electrolyte effects
Temp (°C) 95 90 85 75 65
103 kH + (mol  dm03)01 s01) 8.15 4.85 2.76 1.02 0.29
between different systems.
The equation which describes the influence
Determine graphically (a) the rate constant for of electrolyte on the rate constant is the
acid-catalysis at 25°C, (b) the activation energy. Brønsted–Bjerrum equation,

Answer log k =  log k0 + 2AzA zB μ 1/2 (4.52)


According to the Arrhenius equation, a plot of
In this equation, zA and zB are the charge
log k against 1T has a gradient of 0Ea 2.303R.
numbers of the two interacting ions and A is a
From the graph:
constant for a given solvent and temperature
(a) At 1T # 3.356 " 1003 K01, log k # 05.85. (see equation 3.36). μ is the ionic strength of
Therefore, k at 25°C # 1.41 " 1006 (mol the solution, which we can calculate from
dm03)01 s01.
μ =  12 Σ (mz 2) = 12(mA z2A + mB z2B + ) (4.53)
(b) Gradient # 05.91 " 103 K.
Therefore Ea # 113 kJ mol01. For example, if we have a monovalent drug
ion of concentration 0.01 mol kg01 in the
10_1439PP_CH04 10/11/05 2:54 pm Page 121

Factors influencing drug stability 121

presence of 0.001 mol kg01 of Ca2! ions then values plotted according to equation (4.54).
the ionic strength of the solution will be The gradients of these plots should be propor-
tional to the product of the charge carried by
μ =  12[(0.01 × 1 2) + (0.001 × 2 2)]
the reactive species. Whether the gradient is
 =  0.007 mol kg −1 positive or negative depends on the reaction
If the drug ion and the electrolyte ion are involved. Reactions between ions of similar
both monovalent, then the ionic strength will charge, for example the acid-catalysed hydroly-
be equal to the total molality of the solution. sis of a cationic drug ion, will produce plots of
We can see from equation (4.52) that if we positive slope (i.e. the reaction rate will be
determine the rate constant of a reaction in increased by electrolyte addition), whereas the
the presence of a series of different concen- base-catalysed hydrolysis of positively charged
trations of the same electrolyte and plot log k drug species will produce negative gradients.
against μ12, then the plot should be linear Investigations of the influence of ionic
with an intercept of log k0 and a gradient of strength on reaction rate may therefore be
2AzAzB. This is frequently found to be the case used to provide confirmation of the type of
even in solutions of high ionic strength, reaction which is occurring. For example, the
although equation (4.52) is strictly valid only gradients of the two plots of Fig. 4.14 are 0.260
for ionic strengths of less than 0.01 mol kg01. and 01.759 at pH 3.1 and 7.2 respectively.
At higher ionic strengths (up to about 0.1 mol Since the value of 2A at 90°C is 1.174, we can
kg01) it is preferable to use a modified form of calculate values of the product zA zB as 0.221
the Brønsted–Bjerrum equation in which we and 1.498 at pH 3.1 and 7.2 respectively.
plot log k against μ12(1 ! μ12): These values are not integers as they should
μ 1/2 be if the reactions involved were simple acid-
log k =  log k0 + 2AzA zB

Figure 4.14 shows the degradation of phen-
(1 + μ 1/2)  (4.54) and base-catalysed reactions. This has been
explained by suggesting complex reactions
between the buffer species and the phentol-
tolamine hydrochloride at two different pH amine at each pH.

0.0
pH 7.2

0.5
H
N
CH2 N
OH
Log k (h1)

N
1.0

Me
1.5
pH 3.1

2.0
0.3 0.4 0.5
m1/2/(1 m1/2)

Figure 4.14 The effect of ionic strength, μ, on the hydrolytic rate constant, k, for phentolamine hydrochloride in buffer
solutions of pH 3.1 and 7.2 at 90°C.
10_1439PP_CH04 10/11/05 2:54 pm Page 122

122 Chapter 4 • Drug stability

drug ion and the interacting species are the


E X A M P L E 4.7 Calculation using the Brønsted– same, then we can see that the gradient of the
Bjerrum equation line will be negative. In this case, if we replace
the water with a solvent of lower dielectric
The following values of the rate constant, k,
constant then we will achieve the desired
were obtained at 60°C for the hydrolysis of
effect of reducing the reaction rate. If the drug
penicillin in phosphate buffer at pH 8.8 in a
ion and the interacting ion are of opposite
series of solutions of varying ionic strength, μ:
signs, however, then the slope will be positive
k(h01) 0.078 0.068 0.056 0.049 and the choice of a nonpolar solvent will only
μ (mol kg01) 0.490 0.380 0.270 0.200` result in an increase of decomposition.
Plot the data in accordance with the modified
Brønsted-Bjerrum equation (equation (4.54)) Oxygen
and determine the gradient of the plot.
Comment on whether this value would be Since molecular oxygen is involved in many
expected for the acid-catalysis of the proton- oxidation schemes, we could use oxygen as a
ated penicillin ion. (Note: Assume a value of challenge to find out whether a particular
unity for A at this temperature.) drug is likely to be affected by oxidative break-
down. We would do this by storing solutions
Answer of the drug in ampoules purged with oxygen
The gradient of the plot of log k against and then comparing their rate of breakdown
μ12(1 ! μ12) is !2.0. As expected for a reaction with similar solutions stored under nitrogen.
between ions of like charge (acid and Formulations that are shown to be susceptible
protonated penicillin ion), the gradient is to oxidation can be stabilized by replacing the
positive. oxygen in the storage containers with nitro-
gen or carbon dioxide, by avoiding contact
with heavy metal ions, and by adding anti-
oxidants (see section 4.1.2).
Solvent effects
Since we are considering the hydrolysis of
Light
drugs it might seem that an obvious way to
reduce the breakdown would be to replace Photolabile drugs are usually stored in con-
some or all of the water in the system with a tainers which exclude ultraviolet light, since
solvent such as alcohol or propylene glycol. As exposure to light in this wavelength range is
we will see in this section, however, this is the most usual cause of photodegradation (see
effective only in certain systems and in others section 4.1.4) Amber glass is particularly effec-
it can, in fact, increase the rate of breakdown. tive in this respect because it excludes light of
The equation that allows us to predict the wavelength of less than about 470 nm. As an
effect of the solvent on the hydrolysis rate is added precaution, it is always advisable to
store photolabile drugs in the dark.
KzA zB
log k =  log kε = ∞ − (4.55)
ε
Surfactants
In this equation, K is a constant for a given
system at a given temperature. We can see that As might be expected, the presence of sur-
a plot of log k as a function of the reciprocal of factants in micellar form has a modifying
the dielectric constant, ε, of the solvent should effect on the rate of hydrolysis of drugs. The
be linear with a gradient of magnitude KzA zB magnitude of the effect depends on the differ-
and an intercept equal to the logarithm of the ence in the rate constant when the drug is in
rate constant in a theoretical solvent of infin- aqueous solution and when it is solubilised
ite dielectric constant. If the charges on the within the micelle, and also on the extent of
10_1439PP_CH04 10/11/05 2:54 pm Page 123

Factors influencing drug stability 123

solubilisation. Thus, or cream base used in the formulation. Hydro-


cortisone in a series of commercially available
kobs = km fm + kw fw (4.56)
bases exhibits maximum decomposition in poly-
where kobs , km and kw are the observed, micellar ethylene glycol base.18, 19 The reported shelf-life
and aqueous rate constants, respectively, and was only 6 months in this base, which makes
fm and fw are the fractions of drug associated manufacture on a commercial basis an un-
with the micelles and aqueous phase, respect- reasonable proposition, considering the length
ively. The value of km is dependent on the of time involved in distribution of the drug from
location of the drug within the micelle. A wholesaler to patient.
solubilisate may be incorporated into the Not only should possible stability problems
micelle in a variety of locations. Nonpolar com- be borne in mind in the choice of ointment
pounds are thought to be solubilised within the base at the formulation stage, but also similar
lipophilic core and, as such, are likely to be care should be exercised if the ointment is
more effectively removed from the attacking diluted at a later stage. Such dilution is, unfor-
species than those compounds that are located tunately, common practice in cases where the
close to the micellar surface. Where the drug is practitioner wishes to reduce the potency of
located near to the micellar surface, and there- highly active topical preparations, particularly
fore still susceptible to attack, the ionic nature steroids. The pharmaceutical and biopharma-
of the surfactant is an important influence on ceutical dangers of this procedure have been
decomposition rate. For base-catalysed hydroly- stressed.20 Of particular interest here are the
sis, solubilisation into anionic micelles affords problems of drug stability which can occur
an effective stabilisation due to repulsion of through the use of unsuitable diluents. An
OH0 by the micelles. Conversely, solubilisation example has been cited of the dilution of
into cationic micelles might be expected to betamethasone valerate cream with a cream
cause an enhanced base-catalysed hydrolysis. base having a neutral to alkaline pH. Under
Many drugs associate to form micelles in such conditions, conversion of the 17-ester to
aqueous solution (see section 6.3) and several the less-active betamethasone 21-ester can
studies have been reported of the effect of this occur. Similarly, diluents containing oxidising
self-association on stability. In micellar solu- agents could cause chemical degradation of
tions of benzylpenicillin (500 000 units cm03) fluocinolone acetate to less-active compounds.
the apparent rate of the hydrogen-ion-catal- Incorporation of drugs into gel structures
ysed degradation was increased twofold, but frequently leads to a change in their stability,
that of water- and hydroxide-ion-catalysed such as increased degradation of benzylpeni-
hydrolysis was decreased twofold to three- cillin sodium in hydrogels of various natural
fold.17 Consequently, the pH profile was and semisynthetic polymers.21 At pH 6 in
shifted to higher pH values and the pH of Carbopol hydrogels, the percentage of unde-
minimum degradation was found to be 7.0 composed pilocarpine at equilibrium is a simple
compared to 6.5 for monomeric solution function of the apparent viscosity of the
(8000 units cm03). When compared at the medium.22 The rate constant for degradation
respective pH–rate profile minima, micellar was not, however, significantly affected by
benzylpenicillin was reported to be 2.5 times changes in viscosity. Little influence of viscosity
as stable as the monomeric solutions under on the rate of oxidation of ascorbic acid in solu-
conditions of constant pH and ionic strength. tions of gels of Polysorbate 80 has been noted.23

4.4.3 Solid dosage forms


4.4.2 Semisolid dosage forms
Moisture
The chemical stability of active ingredients incor-
porated into ointments or creams is frequently Water-soluble drugs present in a solid dosage
dependent on the nature of the ointment form will dissolve in any moisture which has
10_1439PP_CH04 10/11/05 2:54 pm Page 124

124 Chapter 4 • Drug stability

adsorbed on the solid surface. The drug will In all cases it is important to minimise
now be in an aqueous environment and its access of moisture during manufacture and
decomposition could be influenced by many storage. The correct selection of packaging is
of the factors which we have already discussed obviously important, although this is not as
when dealing with liquid dosage forms. For straightforward as you might at first think. For
example, decomposition could now occur by example, tablets containing a water-labile
hydrolytic cleavage of ester or amide linkages drug were found to be more stable in a water-
in the drug molecule and hence will be permeable blister package at 50°C than in a
affected by the pH of the adsorbed moisture sealed glass bottle and yet the situation was
film. It is not surprising, therefore, that reversed at room temperature and 70% rela-
moisture is considered to be one of the most tive humidity.24 The reason for this behaviour
important factors that must be controlled in was attributed to the loss of considerable
order to minimise decomposition. amounts of water through the film at 50°C, so
We can get an idea of the extent to which improving stability, and the reverse diffusion
moisture adsorption affects stability from the at room temperature, so decreasing stability.
results shown in Fig. 4.15. We can see from
this figure that an increase in the water vapour
Excipients
pressure (and therefore an increase in the
amount of moisture associated with the drug) One of the main ways in which the excipients
greatly increases the percentage decompo- of the solid dosage form can affect the degra-
sition at any given time. The problem is even dation of drugs is by increasing the moisture
more pronounced with drugs which are content of the preparation. Excipients such as
hygroscopic or which decompose to give starch and povidone have particularly high
hygroscopic products. In such cases it is worth water contents (povidone contains about 28%
trying to prepare a less hygroscopic salt of the equilibrium moisture at 75% relative humid-
drug. ity). However, whether this high moisture

144 torr
60 118 torr

50

40
Per cent decomposed

30

52 torr

20

10

0 120 240 360


Time (h)

Figure 4.15 The effect of water vapour pressure on the decomposition of aminosalicylic acid.
Reproduced from S. S. Kornblum and B. J. Sciarrone, J. Pharm. Sci., 53, 935 (1964).
10_1439PP_CH04 10/11/05 2:54 pm Page 125

Factors influencing drug stability 125

level has an effect on stability depends on how while 0.5% magnesium stearate increased the
strongly it is bound and whether the moisture breakdown rate dramatically.
can come into contact with the drug. Mag- Other studies on the influence of tablet
nesium trisilicate causes increased hydrolysis excipients on drug decomposition have iden-
of aspirin in tablet form because, it is thought, tified problems with stearate salts and it has
of its high water content. been suggested that these salts should be
Many examples of the effects of tablet excip- avoided as tablet lubricants if the active com-
ients on drug decompositions are reported in ponent is subject to hydroxide-ion-catalysed
the pharmaceutical literature. Chemical inter- degradation. The degradative effect of the
action between components in solid dosage alkali stearates is inhibited in the presence of
forms may lead to increased decomposition. malic, hexamic or maleic acids owing, it is
Replacement of the phenacetin in compound thought, to competition for the lubricant
codeine and APC tablets by paracetamol in cation between the drug and the additive acid.
NHS formulations in Australia in the 1960s The base used in the formulation of sup-
(because of the undesirable side-effects of positories can often affect the rate of decom-
phenacetin), led to an unexpected decreased position of the active ingredients. Aspirin
stability of the tablets. The cause was later decomposes in several polyoxyethylene glycols
attributed to a transacetylation reaction which are often incorporated into suppository
between aspirin and paracetamol and also a bases.25 Degradation was shown to be due in
possible direct hydrolysis of the paracetamol part to transesterification, giving the decom-
(Scheme 4.15). position products salicylic acid and acetylated
Figure 4.16 shows the increased generation polyethylene glycol. The rate of decom-
of free salicylic acid at 37°C in the tablets position, which followed pseudo first-order
containing paracetamol. It is interesting to kinetics, was considerably greater than when
note from this figure the effect on stability of a fatty base such as cocoa butter was
tablet excipients. Addition of 1% talc caused used.26 Analysis of commercial batches of
only a minimal increase in the decomposition, 100 mg indometacin–polyethylene glycol

OH

COOH
OCOCH3

NHCOCH3
Aspirin
Hydrolysis Paracetamol
Transacetylation

OH OCOCH3 COOH

OH

CH3COOH  

NH2 NHCOCH3
p -Aminophenol Diacetyl-p -Aminophenol Salicylic acid

Scheme 4.15 Reactions showing the postulated transacetylation between aspirin and paracetamol and the direct
hydrolysis of paracetamol.
Reproduced from B. G. Boggianno et al., Aust. J. Pharm., 51, S14 (1970).
10_1439PP_CH04 10/11/05 2:54 pm Page 126

126 Chapter 4 • Drug stability

Aspirin–paracetamol–
codeine–talc–magnesium stearate Aspirin–paracetamol–
codeine–talc Aspirin–
0.8 paracetamol–
codeine
Free salicylic acid (%) 0.7

0.6 BP 1968 free salicylic acid limit

0.5

0.4

0.3

0.2
Aspirin–
0.1 phenacetin–codeine

0 1 2 3 4 5 6
Time (months)

Figure 4.16 Development of free salicylic acid in aspirin–paracetamol–codeine and aspirin–phenacetin–codeine tablets
at 37°C.
Reproduced from B. G. Boggianno et al., Aust. J. Pharm., 51, S14 (1970).

suppositories27 showed that approximately phenolic impurities in tablet binding agents


2%, 3.5% and 4.5% of the original amount such as povidone, disintegrants such as
of indometacin was esterified with polyoxy- crospovidone and viscosity-modifying agents
ethylene glycol 300 (XV) after storage times of such as alginates can lead to photodegra-
1, 2 and 3 years, respectively. dation. Similarly, coloured products may be
Excipients present in tablet formulations formed by the reaction of aldehydes formed
can have an impact on the photostability of during spray-drying or autoclaving of lactose
the product, the effect arising in many cases with primary amine groups in the product.
from impurities present in the excipients.8
For example, free radical reactions involving
Temperature

O The effect of temperature change on the sta-


bility of solid dosage forms can be complicated
CH3O CH2CO(CH2CH2O)nH
for many possible reasons. The drug or one of
the excipients may, for example, melt or
N CH3 n  3–10 change its polymorphic form as temperature is
increased, or it may contain loosely bound
C O water which is lost at higher temperatures. We
should also remember that the relative humid-
ity will change with temperature and so we
must take care to keep this at a constant value.
Despite these possible complications, many
Cl authors have found that the effect of temper-
ature on decomposition rate can be described
by an Arrhenius-type equation; i.e. plots of
Structure XV Polyethylene glycol esters of indometacin
identified in stored suppositories (n # number of ethylene
log k against 1T are linear. This then enables
oxide units) the stability to be predicted at room
10_1439PP_CH04 10/11/05 2:54 pm Page 127

Stability testing and prediction of shelf-life 127

temperature from measurements made at Light and oxygen


elevated temperatures. Also, of course, we can
We have examined above the stability prob-
calculate an apparent activation energy, Ea,
lems which arise with drugs which are sus-
from the gradient of the plots, but we must
ceptible to photodecomposition or oxidation.
remember that this does not have the same
We will not reconsider them here but merely
meaning as the activation energy for reactions
re-emphasise that we should take all the nec-
in solution. The Ea value in the solid state is
essary precautions to exclude light or oxygen
affected, for example, by changes not only in
when storing these drugs. In this respect we
the solubility of the drug in the moisture layer
should remember that water contains dis-
but also in the intrinsic rate of reaction. We
solved oxygen and so the presence of moisture
cannot, however, use the Arrhenius equation
on the surface of solid preparations may
in cases where decomposition shows an
increase the oxidation of susceptible drugs;
approach to equilibrium. Examples of such
such drugs must, therefore, be stored under
systems include vitamin A in gelatin beadlets
dry conditions.
and vitamin E in lactose base tablets. In these
cases we can often use the van’t Hoff equation
to describe the effect of temperature on break-
down. We determine the equilibrium con- 4.5 Stability testing and prediction of
centrations of products and reactants at a shelf-life
series of temperatures and then plot the
logarithm of the equilibrium constant K
against the reciprocal of the temperature (see It is clearly most important to be able to
Fig. 4.17) according to the equation ensure that a particular formulation when
packaged in a specific container will remain
−ΔH within its physical, chemical, microbiological,
ln K =  + constant (4.57)
RT therapeutic and toxicological specifications

Me

Me O (CH2)3 (CH2)3 (CH2)3


O CHMe2
Me
Me Me
HO2C O

Me

2

3
In K

4

2.8 2.9 3.0 3.1


103/T (K1)

Figure 4.17 Van’t Hoff plot for vitamin E succinate decomposition in lactose base tablets.
Reproduced from J. T. Carstensen et al., J. Pharm. Sci., 57, 23 (1968) with permission.
10_1439PP_CH04 10/11/05 2:54 pm Page 128

128 Chapter 4 • Drug stability

on storage for a specified time. In order to A convenient, approximate method which


make such an assurance, we obviously need is useful for estimation of decomposition rates
to conduct a rigorous stability testing pro- at room temperature makes use of the ratio of
gramme on the product in the form that is rate constants at room temperature (T1 ) and at
finally to be marketed. Since the testing period a higher temperature (T2 ). If we subtract the
can be as long as two years, it has become Arrhenius equations at temperatures T1 and
essential to devise a more rapid technique T2 , assuming that the log A term is the same at
which can be used during product develop- each temperature, we obtain
ment to speed up the identification of the
k2 Ea 1 1
most suitable formulation. In this section we
shall examine ways of predicting the chemical
stability of a formulation during preformu- or
log

k1
= −  
 −
2.303R T2 T1
 (4.59)

lation studies using accelerated storage tests.


k2 E (T − T1)
We will not be considering the toxicological
or microbiological studies or the determina-
tion of physical stability, all of which are
log
 = a 2
k1 2.303RT2 T1
We must, of course, have a value of Ea in order
(4.60)

essential elements in the overall stability to be able to use these equations for the calcu-
testing of the formulation but which are lation of the room temperature rate constant
outside the scope of this chapter. Although k1. If we only require a rough estimation of k1
most of this section will be concerned with the then we can assume a mid-range value of Ea,
prediction of the effect of temperature on drug say 75 kJ mol01, for these calculations.
decomposition, other environmental factors
will also be briefly considered.
EXAMPLE 4.8 Application of the Arrhenius equation
The first-order rate constant for the hydrolysis
4.5.1 Effect of temperature on stability of sulfacetamide at 120°C is 9 " 1006 s01 and
the activation energy is 94 kJ mol01. Calculate
We have already considered the basic method the rate constant at 25°C.
of accelerating the chemical decomposition by
raising the temperature of the preparations. Answer
We will briefly reconsider essential steps in the Using equation (4.60) with an Ea value of
process. The order of reaction can be deter- 94 kJ mol01 we have
mined by plotting stability data at several ele- k120 94 × 10 3 × (393 − 298)
vated temperatures according to the equations
relating decomposition to time for each of
the orders of reaction, until linear plots are
log
 
k25
 =
2.303 × 8.314 × (393 × 298)
 =  3.98
obtained. We can now calculate values of rate Removing the logarithms, k120k25 # 9.55 " 103.
constant at each temperature from the gradi- Therefore, k25 # 9 " 10 069.55 " 10 3 # 9.4 "
ent of these plots, and plot the logarithm of k 10010 s01.
against reciprocal temperature according to
the Arrhenius equation:
An alternative method of data treatment is
Ea to plot the logarithm of the half-life t0.5 as a
log k =  log A − (4.58) function of reciprocal temperature. From
2.303RT
equation (4.13), t0.5 # 0.693k. Therefore,
The required value of k can be interpolated log k =  log 0.693 − log t0.5 (4.61)
from this plot at room temperature, and the
activation energy Ea can be calculated from and substituting into equation (4.51) gives
the gradient, which is 0Ea 2.303R. Values of Ea log t0.5 = log 0.693 − log A + (Ea/2.303RT)
are usually within the range 50–96 kJ mol01. (4.62)
10_1439PP_CH04 10/11/05 2:54 pm Page 129

Stability testing and prediction of shelf-life 129

Once the rate constant is known at the The expiry date is thus 40.9 months after
required storage temperature, it is a simple initial preparation.
matter to calculate a shelf-life for the product
based on an acceptable degree of decomposi-
Although accelerated storage testing based
tion. The equations which we can use for 10%
on the use of the Arrhenius equation has
loss of activity are obtained by substituting
resulted in a very significant saving of time, it
x # 0.1a in the zero- and first-order equations
still involves the time-consuming step of the
(equations 4.8 and 4.10), giving
initial determination of the order of reaction
[D]0 for the decomposition. While most investi-
t0.9 = 0.1 (zero order) (4.63) gators have emphasised the need for a know-
k0
ledge of the exact kinetic pathway of
and degradation, some have bypassed this initial
0.105 step by assuming a particular decomposition
t0.9 = (first order) (4.64) model. At less than 10% degradation and
k1
within the limits of experimental error
where [D]0 is the initial concentration of drug. involved in stability studies, it is not possible
Although t0.9 is usually used as an estimate of to distinguish between zero-, first- or simple
shelf-life, other percentage decompositions second-order kinetics using curve-fitting tech-
may be required, for example when the niques; consequently, the assumption of first-
decomposition products produce discol- order kinetics for any decomposition reaction
oration or have undesirable side-effects. The should involve minimum error. In fact, it was
required equations for these may be derived shown that there was a linear relationship
by substituting in the relevant rate equations. between the logarithm of t0.9 (the time taken
for the concentration of the reactant to decom-
pose to 90% of its original value) and the reci-
EXAMPLE 4.9 Calculation of shelf-life procal temperature, which was independent of
the order of reaction for the decomposition of
The initial concentration of active principle in a series of drugs.28 On the basis of these find-
an aqueous preparation was 5.0 " 1003 g cm03. ings it was suggested that the use of such linear
After 20 months the concentration was shown plots to determine t0.9 at the required tempera-
by analysis to be 4.2 " 1003 g cm03. The drug is ture would provide a rapid, and yet sufficiently
known to be ineffective after it has decom- accurate, means of studying decomposition
posed to 70% of its original concentration. rate during the development stage.
Assuming that decomposition follows first- Even with the modifications suggested
order kinetics, calculate the expiry date of the above, the method of stability testing based
drug preparation. on the Arrhenius equation is still time-
consuming, involving as it does the separate
Answer determination of rate constants at a series of
Substituting into the first-order equation elevated temperatures. Experimental tech-
(equation 4.10) niques have been developed29, 30 which enable
k =  (2.303/20) log[(5 × 10 −3)/(4.2 × 10 −3)] the decomposition rate to be determined
from a single experiment. Such methods
k =  8.719 × 10 −3 month −1 involve raising the temperature of the
70% of the initial concentration # 3.5 " 1003 g product in accordance with a predetermined
cm03 temperature–time programme and are conse-
quently referred to as nonisothermal stability
t =  (2.303/8.719 × 10 −3) studies.
× log[(5 × 10 −3)/(3.5 × 10 −3)] Any suitable temperature–time programme
t =  40.9 months may be used. In the method of Rogers25 the
10_1439PP_CH04 10/11/05 2:54 pm Page 130

130 Chapter 4 • Drug stability

rise of temperature was programmed so that reactants at the beginning of the experiment,
the reciprocal of the temperature varied loga- and at and bt are their concentrations at time
rithmically with time according to t. The value of the final term of equa-
tion (4.70) rapidly tends to zero as kt becomes
1 1
−  =  2.303b log(1 + t) (4.65) greater than k0. Thus a graph of log f (c) against
T0 Tt log(1 ! t) will be linear from that time after
where T0 and Tt  are the temperatures at zero which k0 is negligible in comparison with kt.
time and at time t, respectively, and b is any The slope of the line is (1 ! Ea bR), enabling Ea
suitable proportionality constant. Applying to be determined. The rate constant k0 may
the Arrhenius equation at both temperatures then be calculated from the intercept when
and subtracting gives log(1 ! t) # 0, which is equal to log k0 0
log(1 ! Ea bR). The rate constant at any other
Ea 1 1
 
temperature may be calculated from k0 and Ea.
log kt =  log k0 +   − (4.66)
2.303R T0 Tt
Substituting equation (4.65) into equation
E X A M P L E 4 . 1 0 Accelerated storage testing
(4.66) gives
using temperature–time programme
Ea b
log kt =  log k0 +  log(1 + t) (4.67) In a study of the first-order decomposition of
R riboflavin in 0.05 mol dm03 NaOH using accel-
Therefore, erated storage techniques, the temperature was
programmed to rise from 12.5 to 55°C using a
kt =  k0(1 + t) Eab/R (4.68) programme constant, b, of 2.171 " 1004 K01.
For first-order reactions 0dcdt # kc, where c is The initial concentration, c0, of riboflavin
concentration. Substituting for k from equa- was 1004 mol dm03, and the concentration ct
tion (4.68) and integrating gives remaining at time t was as follows:
ct t t (h) 0.585 0.996 1.512 2.163 2.982 4.013 5.312 6.946
dc
−     =  k0   (1 + t) Eab/R dt (4.69) 105 ct 9.881 9.763 9.532 9.109 8.371 6.902 4.931 2.435
c0 c 0 (mol dm03)

where c0 and ct are the concentrations at zero Calculate the activation energy and the rate
time and at time t, respectively. constant at 20°C.
Therefore,
Ea b E b Answer
log f(c) = log k0 − log 1 +
 R  + 1 + a

k0
R
1+R/Eab
For first-order reactions the data are plotted
according to equations (4.70) and (4.71).

  
× log (1 + t) + log 1 − t log(1 ! t ) log[2.303 log(c0ct)]
kt 0.585 0.2 01.94
(4.70) 0.996 0.3 01.62
1.512 0.4 01.32
where 2.163 0.5 01.03
c0 2.982 0.6 00.75
f(c) = 2.303 log  (4.71) 4.013 0.7 00.43
ct
5.321 0.8 00.15
A similar equation applies to second-order 6.946 0.9 !0.15
reactions with A plot of log[2.303 log(c0ct)] against log(1 ! t)
2.303 a 2.303 b is linear (see Fig. 4.18) with a slope of 2.95.
log f(c) =  log  t +  log  0
a0 − b0 bt a0 − b0 a0 From equation (4.70),
(4.72) Ea b
Slope = 1 +
where a0 and b0 are the concentrations of the R
10_1439PP_CH04 10/11/05 2:54 pm Page 131

Stability testing and prediction of shelf-life 131

Therefore, form of equation (4.66):


Ea = (1.95 × 8.314)/(2.171 × 10 −4) log kt =  −2.08 + [74 680/(2.303 × 8.314)]
= 74.68 kJ mol −1  ×  [(1/285.5) − (1/293)]
Intercept at log(1 ! t) # 0 is 02.55. From equa- = −2.08 + 0.3497 = −1.730
tion (4.70),
and
Ea b
Intercept = log k0 − log 1 +
R
where the final term on the right of equa-
  kt =  1.86 × 10 −2 h −1
The rate constant at 20°C is thus 0.0186 h01.
tion (4.70) is neglected.
Therefore, The advantages of this method over the
conventional method of stability testing are
log k0 = −2.55 + log 2.95 = −2.08 that (a) the data required to calculate the
and stability are obtained in a single one-day
experiment rather than from a series of experi-
k0 = 0.0083 h −1
ments which may last for several weeks; (b) no
That is, the rate constant at temperature T0 preliminary experiments are required to deter-
(12.5°C) is 0.0083 h01. mine the optimum temperatures for the accel-
The rate constant at 20°C may then be erated storage test; and (c) the linearity of the
calculated from the Arrhenius equation in the plot of log f (c) against log(1 ! t) confirms that

1
O

Me N
log [2.303 log (c0 /ct )]

NH

Me N N O
2 HO

HO OH

OH

3

0 0.2 0.4 0.6 0.8 1.0


Log (1  t )

Figure 4.18 Example 4.10: accelerated storage plot for the decomposition of riboflavin in 0.05 mol dm03 NaOH using
data from reference 29.
10_1439PP_CH04 10/11/05 2:54 pm Page 132

132 Chapter 4 • Drug stability

the correct order of reaction has been decomposition processes involving parallel or
assumed. consecutive reactions, there may be a change
Several improvements on the original non- in the relative contributions of the compo-
isothermal stability testing methods have nent reactions as the temperature is increased.
been suggested. Rather than subjecting the
drug formulation to a predetermined fixed
Suspensions
time–temperature profile, the temperature
may be changed during the course of the One complication which arises when we are
experiment at a rate consistent with the carrying out stability testing of suspensions is
analytical results from the experiment.30 The the changes in the solubility of the suspended
resultant time–temperature data are fitted to a drug with increase in temperature. With sus-
polynomial expression of sufficient degree to pensions, the concentration of the drug in
describe the changes. This relationship and solution usually remains constant because, as
the experimental data are then combined and the decomposition reaction proceeds, more of
utilised to compute a series of degradation the drug dissolves to keep the solution satu-
pathways corresponding to a series of values of rated. As we have seen, this situation usually
activation energy. The curves are matched leads to zero-order release kinetics. If the
with the experimental analytical data to actual decomposition of dissolved drug is first-
obtain the correct activation energy for the order, then we can express the decrease of
reaction. Using this activation energy and the concentration, c, with time, t, as
analytical data, the reaction rate and stability
may be calculated. Computational procedures dc
−  =  k0 (4.73)
whereby the activation energy and frequency dt
factor of the Arrhenius equation may be where k0 # k1 S and S is the solubility of the
determined from simple nonisothermal exper- drug.
iments with a fixed temperature–time profile The problem that arises with these systems
have been described.31, 32 is that an increase of temperature causes not
An improvement in the design of stability only the usual increase in rate constant but
tests which avoids the difficulties inherent in also an increase in solubility. Application of
the nonlinear curve-fitting procedures outlined the Arrhenius equation to the data involves
above has been suggested.33 The experimental the measurement of the changes in solubility
procedure involves changing the temperature of the drug over the temperature range
of the samples being studied until degradation involved. An alternative method which does
is rapid enough to proceed at a convenient not necessitate the determination of solubility
rate for isothermal studies to be carried out. uses the relationship between solubility and
The analytical information obtained during the temperature:
nonisothermal and isothermal portions of
the experiment is utilised in calculating the ΔHf
log S = − + constant (4.74)
activation energy and determining the order of 2.303RT
reaction and the reaction rate and predicting
stability at any required temperature. Since k0 # k1 S,
Although accelerated storage testing has log k0 = log k1 + log S
proved invaluable in the development of
stable formulations, it is important that we If we substitute for log k1, using the Arrhenius
consider some of the limitations of this tech- equation, we obtain
nique. We must take care that the order of (−Ea + ΔHf)
reaction is not different at the higher tem- log k0 = + constant (4.75)
2.303RT
peratures from that which occurs at room
temperature. There are several cases where this where ΔHf is the molar heat of fusion. You can
might be so. For example, with complex now see that we can plot log k0 against 1T
10_1439PP_CH04 10/11/05 2:54 pm Page 133

Stability testing and prediction of shelf-life 133

and extrapolate to give a room temperature temperature; and (c) that a separate, sealed
value of k0. It is important that we remember ampoule should be taken for each assay point
that this treatment assumes that drug degrad- and water determination, thus avoiding dis-
ation in solution follows first-order kinetics turbance of water equilibrium on opening the
and that the kinetics are not limited by the container.
dissolution rate. As has been discussed above, we can often
use the Arrhenius equation to predict stability
Solid state in the solid state, even though the kinetics of
breakdown are different from those in solu-
The main problems arising in stability testing tion. The exception to this is when equilibrium
of solid dosage forms are24 (a) that the analyt- reactions occur, in which case we can often use
ical results tend to have more scatter because the van’t Hoff equation (equation 4.57) to
tablets and capsules are distinct dosage units predict room-temperature stability.
rather than the true aliquots encountered with
stability studies on drugs in solution, and
(b) that these dosage forms are heterogeneous 4.5.2 Other environmental factors affecting
systems often involving a gas phase (air and stability
water vapour), a liquid phase (adsorbed mois-
ture) and the solid phase itself. The compo- Light
sitions of all of these phases can vary during
an experiment. Photostability testing of drug substances
The first of these problems can be overcome usually involves the initial stress testing of the
by ensuring uniformity of the dosage form drug to determine its overall photostability
before commencing the stability studies. The and the identification of any degradation
problems arising from the heterogeneity are products. In this process the sample is irra-
more difficult to overcome. The main compli- diated at all absorbing wavelengths using a
cating factor is associated with the presence of broad-spectrum light source. Those drugs or
moisture. As we have seen in section 4.4.3, formulations which are shown to be photo-
moisture can have a significant effect on sensitive are then subjected to more formal
the kinetics of decomposition and this may photostability testing in which they are chal-
produce many experimental problems during lenged with light of wavelength comparable
stability testing. For example, with gelatin to that to which the formulations are exposed
capsules the water in the capsule shell must in practical situations. During their shelf-life it
equilibrate with that in the formulation and is most likely that the products will be exposed
surrounding air and this may require an appre- to fluorescent light, direct daylight and day-
ciable time. The prediction of stability is diffi- light filtered through window glass, and the
cult in solid dosage forms in which there is stability testing procedures are designed to
chemical interaction between components, or cover these possibilities. A specific protocol for
chemical equilibrium phenomena. In fact, the testing the photostability of new drugs and
data for stability studies involving the latter products is described in the ICH Guideline.34
are often plotted using a van’t Hoff plot rather
than an Arrhenius plot.
Oxygen
To reduce some of these problems, parti-
cularly those associated with moisture, during The stability of an oxidisable drug in a liquid
stability testing, the following have been dosage form is generally a function of the effi-
suggested:24 (a) the use of tightly sealed con- ciency of any antioxidant included in the for-
tainers, except where the effect of packaging is mulation. Exaggeration of the effect of oxygen
to be investigated; (b) that the amount of on stability may be achieved by an increase in
water present in the dosage form should the partial pressure of oxygen in the system. It
be determined, preferably at each storage is not often easy, however, to make decisions
10_1439PP_CH04 10/11/05 2:54 pm Page 134

134 Chapter 4 • Drug stability

on what would be the normal access of oxygen 4.5.3 Protocol for stability testing
during storage and a meaningful extrapolation
of the acquired data may be difficult. A recently agreed stability-testing requirement
for a Registration Application within the three
areas of the EC, Japan and the USA exemplifies
Moisture content
the core stability data package required for
The stability of solid dosage forms is usually new drug substances and associated drug
very susceptible to the moisture content of the products.36 Under this agreement, infor-
atmosphere in the container in which they are mation on stability generated in any one of
stored (see section 4.4.3) A linear relationship these three areas which meets the appropriate
between log k and the water vapour pressure requirements of this guideline is mutually
for vitamin A palmitate beadlets in sugar- acceptable in both of the other two areas. The
coated tablets has been found.35 Similarly, a following summarises some of the main
linear relationship between the logarithm of points of the guideline as it affects the stability
the rate constant for the decomposition of testing of both drug substances and drug
nitrazepam in the solid state and the relative products; the original document should of
humidity has been established (Fig. 4.19). course be consulted if a more detailed account
The need for consideration of the effect of is required.
moisture on stability has been stressed by
Carstensen,13 who stated that stability pro-
Drug substances
grammes should always include samples that
have been artificially stressed by addition of Stability information from accelerated and
moisture. One purpose of a stability pro- long-term testing is required to be provided on
gramme should be to define the stability of at least three batches manufactured to a mini-
the dosage form as a function of moisture mum of pilot plant scale by the same synthetic
content. route and using a method of manufacture and

83.4C

3.0 73.8C
105 log k

55.0C
2.0

42.5C

1.0

0
30 40 50 60 70
Relative humidity (% )

Figure 4.19 Logarithm of the nitrazepam decomposition constant, k, as a function of relative humidity at various
temperatures.
Reproduced from D. Genton and U. W. Kesselring, J. Pharm. Sci., 66, 676 (1977) with permission.
10_1439PP_CH04 10/11/05 2:54 pm Page 135

Stability testing and prediction of shelf-life 135

procedure that simulates the final process to sufficient to establish the stability character-
be used on a manufacturing scale. In this istics of the drug substance; under the long-
context, ‘pilot plant scale’ is taken to mean a term conditions this will normally be every 3
minimum scale of one tenth that of the full months over the first year, every 6 months
production process. The containers to be used over the second year and then annually.
in the long-term evaluation should be the
same as, or simulate, the actual packaging
Drug product
used for storage and distribution. The overall
quality of the batches of drug substance sub- The design of the stability programme for the
jected to stability testing should be representa- finished product is based on the knowledge of
tive of both the quality of the material used in the behaviour and properties of the drug sub-
preclinical and clinical studies and the quality stance and the experience gained from clinical
of material to be made on a manufacturing formulation studies and from stability studies
scale. on the drug substance. Stability information
The testing should be designed to cover from long-term and accelerated testing is
those features susceptible to change during required to be presented on three batches of
storage and likely to influence quality, safety the same formulation and dosage form in the
andor efficacy, including, as necessary, the containers and closure proposed for market-
physical, chemical and microbiological char- ing. Two of the three batches should be at least
acteristics. The length of the studies and the pilot scale; the third batch may be smaller, for
storage conditions should be sufficient to example 25 000–50 000 tablets or capsules for
cover storage, shipment and subsequent use. solid dosage forms. Data on laboratory-scale
The specifications for the long-term testing are batches are not acceptable as primary stability
a temperature of 25 & 2°C and 60 & 5% relative information. It is stipulated that the manu-
humidity (RH) for a period of 12 months. For facturing process to be used should meaning-
accelerated testing the temperature is specified fully simulate that which would be applied to
as 40 & 2°C and RH as 75 & 5% for a period of 6 large-scale batches for marketing and should
months. Other storage conditions are allow- provide product of the same quality intended
able if justified; in particular, temperature- for marketing, and meeting the same quality
sensitive drugs should be stored at a lower specification as to be applied for release of
temperature, which then becomes the desig- material. Where possible, batches of the
nated long-term testing temperature. The 6- finished product should be manufactured
month accelerated testing should then be using identifiably different batches of drug
carried out at a temperature at least 15°C substance.
above this designated temperature together As with the stability testing of drug sub-
with the relative humidity appropriate to that stance, the testing of the product should cover
temperature. Where ‘significant change’ occurs those features susceptible to change during
during the 6-month accelerated storage storage and likely to influence quality, safety
testing, additional testing at an intermediate andor efficacy. The range of testing should
temperature (such as 30 & 2°C60% & 5% RH) cover not only chemical and biological sta-
should be conducted for drug substances to be bility but also loss of preservative, physical
used in the manufacture of dosage forms properties and characteristics, organoleptic
tested for long-term stability at 25°C60% RH. properties and, where required, microbiologi-
‘Significant change’ at 40°C75% RH or cal attributes. The conditions and time periods
30°C60% RH is defined as failure to meet the for long-term and accelerated storage testing
specification. are the same as those outlined above for drug
The long-term testing is required to be substances but with special considerations
continued for a sufficient period beyond arising from the nature of the drug product. If
12 months to cover all appropriate re-test it is necessary to store the product at a lower
periods. The frequency of testing should be temperature because of its heat sensitivity,
10_1439PP_CH04 10/11/05 2:54 pm Page 136

136 Chapter 4 • Drug stability

then consideration should be given to any controlled by adjusting the pH to that of


physical or chemical change in the product maximum stability or in some cases by the
which might occur at this temperature; for addition of nonaqueous solvents.
example, suspensions or emulsions may sedi- ● Oxidative degradation is a problem with
ment or cream, while oils and semi-solid drugs possessing carbon–carbon double
preparations may show an increased viscosity. bonds such as the steroids, polyunsaturated
Storage under conditions of high relative fatty acids and polyene antibiotics. Such
humidity applies particularly to solid dosage drugs can be stabilised by replacing the
forms. For products such as solutions and oxygen in the system with inert gases such
suspensions contained in packs designed to as nitrogen; by avoiding contact with metals
provide a permanent barrier to water loss, such as iron, cobalt and nickel, and by
specific storage under conditions of high RH is adding antioxidants or reducing agents to
not necessary, but the same range of temper- the solution. Some oxidative degradations
atures should be applied. It is recognised that are pH-dependent and can be stabilised by
low RH (10–20%) can adversely affect prod- buffering the system.
ucts packed in semipermeable containers such ● Loss of activity of solutions of some drugs
as solutions in plastic bags and nose drops in such as the tetracyclines can occur because
small plastic containers, and consideration of epimerisation of the drug molecule,
should be given to appropriate testing under while others such as vitamin A lose activity
such conditions. because of geometrical isomerisation.
In the case of drug products, ‘significant ● Photochemical decomposition can be a
change’ at the accelerated condition is defined problem with drugs such as the pheno-
as thiazine tranquillisers and can cause discol-
oration of the solution and loss of activity.
● A 5% potency loss from the initial assay
Such systems have to be stored in amber
value of a batch
glass containers, which remove the ultra-
● Any specified degradant exceeding its spec-
violet components of light.
ification limit ● Reactions can be classified according to
● The product exceeding its pH limits
their order of reaction; the breakdown of
● Dissolution exceeding the specification
drugs in the majority of preparations in
limits for 12 capsules or tablets
which the drug is dissolved in aqueous
● Failure to meet specifications for appear-
solution follows first-order or pseudo first-
ance and physical properties, e.g. colour,
order kinetics. There are, however, many
phase separation, resuspendability, delivery
cases of drugs in which decomposition
per actuation, caking and hardness
occurs simultaneously by two or more
If significant change occurs at 40°C75% RH pathways (parallel reactions), or involves a
then it is necessary to submit a minimum of 6 sequence of decomposition steps (consecu-
months’ data from an ongoing one-year study tive reactions) or a reversible reaction.
at 30°C60% RH using the same criteria for ● The hydrolysis rate of drugs in liquid dosage
‘significant change’. forms is strongly influenced by the pH of the
solution and can be catalysed not only by
H! and OH0 ions (specific acid–base cataly-
sis) but also by the components of the buffer
Summary
used (general acid–base catalysis). We have
looked at the ways in which the effect of the
● The most common cause of degradation of buffer components can be removed so that
drugs in aqueous systems is hydrolysis and the pH of maximum stability of the solution
the most susceptible drugs are those con- can be determined from the pH–rate profile
taining ester, amide, lactone, lactam, imide and the rate constants for specific acid–base
or carbamate groups. Hydrolysis can be catalysis can be calculated.
10_1439PP_CH04 10/11/05 2:54 pm Page 137

References 137

● Temperature increase usually causes a pro- influence the decomposition of drugs in


nounced increase of hydrolytic degrada- aqueous solution. Excipients of the solid
tion. We have seen how to calculate the dosage form can affect the drug breakdown
hydrolytic rate constant at room temper- by increasing the moisture content of the
ature from data at elevated temperatures dosage form.
using the Arrhenius equation. ● Temperature increase causes an increase in
● The addition of electrolyte can increase the the rate of breakdown of drugs in solid
hydrolysis rate if the reaction involves the dosage forms, which can often be described
interaction of the drug ion with an ion of by the Arrhenius equation, although the
similar charge. Similarly, a change of effect of temperature change is usually far
solvent to one of lower dielectric constant more complicated than for liquid formu-
will stabilise only this type of system but lations. This equation cannot be used for
not one involving the reaction between systems that show an approach to equi-
ions of opposite sign. librium. The van’t Hoff equation is often
● In solid dosage forms containing drugs that useful to describe the effect of temperature
are susceptible to hydrolysis, decomposi- on the decomposition of these systems.
tion of the drug can occur if moisture is ● In liquid formulations the shelf-life of a for-
allowed to adsorb on the surface of the mulation can be estimated by application
dosage form. Careful selection of packaging of the Arrhenius equation. The protocol for
is important to reduce this possibility. Drug stability testing of drug substances and drug
which dissolves in this surface layer will products has been discussed.
be affected by many of the factors which

References

1. P. R. Wells. Linear free energy relationships. Chem. 8. H. H. Tønnesen. Formulation and stability testing
Rev., 63, 171–219 (1963) of photolabile drugs. Int. J. Pharm., 225, 1–14
(2001)
2. J. T. Cartensen, E. G. Serenson and J. J. Vance. Use
of Hammett graphs in stability programs. J. Pharm. 9. J. V. Greenhill and M. A. McLelland. Photo-
Sci., 53, 1547–8 (1964) decomposition of drugs. Prog. Med. Chem., 27,
51–121 (1990)
3. S. W. Hovorka and C. Schöneich. Oxidative degra-
dation of pharmaceuticals: theory, mechanisms 10. C. L. Huang and F. L. Sands. Effect of ultraviolet
and inhibition. J. Pharm. Sci., 90, 253–69 (2001) irradiation on chlorpromazine. II. Anaerobic
condition. J. Pharm. Sci., 56, 259–64 (1967)
4. D. M. Johnson and W. F. Taylor. Degradation of
fenprostalene in polyethylene glycol 400 solution. 11. Y. Matsuda, H. Inouye and R. Nakanishi.
J. Pharm. Sci., 73, 1414–7 (1984) Stabilization of sulfisomidine tablets by use of film
coating containing UV absorber: Protection of
5. L. Halbaut, C. Barbé, M. Aróztegui and C. de la
coloration and photolytic degradation from
Torre. Oxidative stability of semi-solid excipient
exaggerated light. J. Pharm. Sci., 67, 196–201
mixtures with corn oil and its implication in the
(1978)
degradation of vitamin A. Int. J. Pharm., 147, 31–40
(1997) 12. H. Bundgaard. Polymerization of penicillins:
kinetics and mechanism of di- and polymerization
6. G. B. Smith, L. DiMichele, L. F. Colwell, et al.
of ampicillin in aqueous solution. Acta Pharm.
Autooxidation of simvastatin. Tetrahedron, 49,
Suec., 13, 9–26 (1976)
4447–62 (1993)
13. J. T. Carstensen. Stability of solids and solid dosage
7. M. T. Lamy-Freund, V. F. N. Ferreira, A. Faljoni-
Alário and S. Schreier. Effect of aggregation on the forms. J. Pharm. Sci., 63, 1–14 (1974)
kinetics of autoxidation of the polyene antibiotic 14. J. T. Carstensen. Drug Stability. Principles and
amphotericin B. J. Pharm. Sci., 82, 162–6 (1993) Practices 2nd edn, Marcel Dekker, New York, 1995
10_1439PP_CH04 10/11/05 2:54 pm Page 138

138 Chapter 4 • Drug stability

15. R. Andersin and S. Tammilehto. Photochemical 26. C. W. Whitworth, L. A. Luzzi, B. B. Thompson and
decomposition of midazolam. IV. Study of pH- H. W. Jun. Stability of aspirin in liquid and semi-
dependent stability by high-performance liquid solid bases. II. Effect of fatty additives on stability
chromatography. Int. J. Pharm., 123, 229–35 in a polyethylene glycol base. J. Pharm. Sci., 62,
(1995) 1372–4 (1973)
16. K. Torniainen, S. Tammilehto and V. Ulvi. The 27. R. Ekman, L. Liponkoski and P. Kahela. Formation
effect of pH, buffer type and drug concentration of indomethacin esters in polyethylene glycol
on the photodegradation of ciprofloxacin. Int. J. suppositories. Acta Pharm. Suec., 19, 241–6 (1982)
Pharm., 132, 53–61 (1996) 28. A. K. Amirjahed. Simplified method to study
17. J. T. H. Ong and H. B. Kostenbauder. Effect of self- stability of pharmaceutical preparations. J. Pharm.
association on rate of penicillin G degradation in Sci., 66, 785 (1977)
concentrated aqueous solutions. J. Pharm. Sci., 64,
29. A. R. Rogers. An accelerated storage test with pro-
1378–80 (1975)
grammed temperature rise. J. Pharm. Pharmacol.,
18. A. E. Allen and V. Das Gupta. Stability of hydro- 15, 101T (1963)
cortisone in polyethylene glycol ointment base. J.
30. H. V. Maudling and M. A. Zoglio. Flexible non-
Pharm. Sci., 63, 107–9 (1974)
isothermal stability studies. J. Pharm. Sci., 59,
19. V. Das Gupta. Effect of vehicles and other active 333–7 (1970)
ingredients on stability of hydrocortisone. J.
31. B. W. Madsen, R. A. Anderson, D. Herbison-Evans
Pharm. Sci., 67, 299–302 (1978)
and W. Sneddon. Integral approach to noniso-
20. M. J. Busse. Dangers of dilution of topical steroids. thermal estimation of activation energies. J. Pharm.
Pharm. J., 220, 25 (1978) Sci., 63, 777–81 (1974)
21. E. Ullmann, K. Thoma and G. Zelfel. The stability 32. A. I. Kay and T. H. Simon. Use of an analog com-
of sodium penicillin G in the presence of ionic puter to simulate and interpret data obtained from
surfactants, organic gel formers, and preservatives. linear nonisothermal stability studies. J. Pharm.
Pharm. Acta. Helv., 38, 577–86 (1963) Sci., 60, 205–8 (1971)
22. B. Testa and J. C. Etter. Hydrolysis of pilocarpine in 33. M. A. Zoglio, H. V. Maudling, W. H. Streng and W.
Carbopol hydrogels. Can. J. Pharm. Sci., 10, 16–20 C. Vincek. Nonisothermal kinetic studies III: rapid
(1975) nonisothermal-isothermal method for stability
23. R. I. Poust and J. C. Colaizzi. Copper-catalyzed prediction. J. Pharm. Sci., 64, 1381 (1975)
oxidation of ascorbic acid in gels and aqueous 34. ICH Harmonised Tripartite Guideline. Q1B:
solutions of polysorbate 80. J. Pharm. Sci., 57, Photostability Testing of New Drug Substances and
2119–25 (1968) Products. Federal Register, 62, 27115 (1997)
24. J. Tingstad and J. Dudzinski. Preformulation 35. L. Lachman. Physical and chemical stability
studies. II. Stability of drug substances in solid testing of tablet dosage forms. J. Pharm. Sci., 54,
pharmaceutical systems. J. Pharm. Sci., 62, 1856–60 1519–26 (1965)
(1973)
36. Stability testing of new drug substances and
25. H. W. Jun, C. W. Whitworth and L. A. Luzzi. products (ICH), MCA EuroDirect Publication No.
Decomposition of aspirin in polyethylene glycols. 333592.
J. Pharm. Sci., 61, 1160–2 (1972)

You might also like