Sie sind auf Seite 1von 22

Drug Delivery: Controlled Release

Yie W. Chien
Research and Development, Kaohsiung Medical University,
Kaohsiung, Taiwan

Senshang Lin
College of Pharmacy and Allied Health Professions, St. John’s University,
Jamaica, New York, U.S.A.

INTRODUCTION 3. Feedback-regulated drug delivery systems.


4. Site-targeting drug delivery systems.
Over the past decades, the treatment of illness has been
accomplished by administering drugs to the human In this article, the scientific concepts and technical
body via various pharmaceutical dosage forms, like principles behind the development of this new
tablets. These traditional pharmaceutical products are generation of drug-delivery systems are outlined and
still commonly seen today in the prescription and discussed.
over-the-counter drug marketplace. To achieve and
maintain the drug concentration in the body within
the therapeutic range required for a medication, it is RATE-PREPROGRAMMED DRUG
often necessary to take this type of drug delivery sys- DELIVERY SYSTEMS
tem several times a day. This yields an undesirable
‘‘seesaw’’ drug level in the body (Fig. 1). In this group of CrDDSs, the release of drug molecules
A number of advancements have been made from the delivery systems has been preprogrammed at
Drug Delivery
Buccal–Mono

recently in the development of new techniques for drug a specific rate profile. This is accomplished by system
delivery. These techniques are capable of regulating the design, which controls the molecular diffusion of drug
rate of drug delivery, sustaining the duration of thera- molecules in and/or across the barrier medium within
peutic action, and/or targeting the delivery of drug to or surrounding the delivery system. Fick’s laws of dif-
a specific tissue.[1–6] These advancements have already fusion are often followed. These CrDDSs can further
led to the development of several novel drug delivery be classified as follows:
systems that could provide one or more of the follow-
ing benefits: 1. Polymer membrane permeation-controlled drug
delivery systems.
1. Controlled administration of a therapeutic dose 2. Polymer matrix diffusion-controlled drug deliv-
at a desirable rate of delivery. ery systems.
2. Maintenance of drug concentration within an 3. Polymer (membrane/matrix) hybrid-type drug
optimal therapeutic range for prolonged delivery systems.
duration of treatment. 4. Microreservoir partition-controlled drug deliv-
3. Maximization of efficacy-dose relationship. ery systems.
4. Reduction of adverse side effects.
5. Minimization of the needs for frequent dose
intake. Polymer Membrane Permeation-Controlled
6. Enhancement of patient compliance. Drug Delivery Systems

Based on the technical sophistication of the In this type of CrDDS, a drug formulation is either
controlled-release drug delivery systems (CrDDSs) that totally or partially encapsulated in a drug reservoir
have been marketed so far, or that are under active compartment whose drug-releasing surface is covered
development, the CrDDSs can be classified (Fig. 2) by a rate-controlling polymeric membrane. The drug
as follows: reservoir can be drug solid particles, a dispersion of
drug solid particles, or a concentrated drug solution in
1. Rate-preprogrammed drug delivery systems. a liquid- or solid-type dispersing medium. The poly-
2. Activation-modulated drug delivery systems. meric membrane can be fabricated from a homogeneous
Encyclopedia of Pharmaceutical Technology DOI: 10.1081/E-EPT-100001051
1082 Copyright # 2007 by Informa Healthcare USA, Inc. All rights reserved.
Drug Delivery: Controlled Release 1083

A C
A4 Adverse side effects Sphere Sheet
Polymer
coating
Drug concentration

Toxic level Drug


A3 reservoir
A2 B
A1 Therapeutic range
Cylinder Pore
B
Drug
Minimum effective Drug release impermeable
concentration Drug barrier
reservoir Nonporous
No therapeutic membrane
effects
D Diffusion layer Porous membrane
1 2 3 4 Cp
Cp
Frequencies of dosing Pd
Pm Cm Cb Sink
Fig. 1 Drug concentration profiles in the systemic circu- Cs
Dm Da Elution medium
lation as a result of taking a series of multiple doses of a con-
ventional drug-delivery system (A1, A2, . . . ) in comparison hd
with the ideal drug concentration profile (B). (Adapted from hm
Ref.[6].)
Fig. 3 Release of drug from various shapes of polymer
membrane permeation-controlled drug-delivery systems: (A)
or a heterogeneous non-porous polymeric material or sphere-type, (B) cylinder-type, and (C) sheet-type. In (D),
a microporous or semipermeable membrane. The the drug concentration gradients across the rate-controlling
polymeric membrane and hydrodynamic diffusion layer exist
encapsulation of drug formulation inside the reservoir
in series. Both the polymer membrane, which is either porous
compartment can be accomplished by molding, cap-
or non-porous, and the diffusion layer have a controlled
sulation, microencapsulation, or other techniques. thickness (hm and hd, respectively).
Different shapes and sizes of drug delivery systems
can be fabricated (Fig. 3).
The release of drug from this type of CrDDSs Dm, and Dd are, respectively, the diffusion coefficients

Drug Delivery
Buccal–Mono
should be at a constant rate (Q/t), which is defined in the rate-controlling membrane with a thickness of
by the following general equation: hm, and in the aqueous diffusion layer with a thickness
of hd. For microporous membrane, the porosity, and
Q Km=r Ka=m Dd Dm tortuosity of the pores in the membrane should be
¼ CR ð1Þ included in the estimation of Dm and hm. CR is the drug
t Km=r Dm hd þ Ka=m Dd hm
concentration in the reservoir compartment.
The release of drug molecules from this type of
where Km/r and Ka/m are, respectively, the partition- CrDDS is controlled at a preprogrammed rate by mod-
coefficients for the interfacial partitioning of drug ulating the partition coefficient and the diffusivity of
molecules from the reservoir to the membrane and drug molecule and the rate-controlling membrane
from the membrane to the aqueous diffusion layer; and the thickness of the membrane. Several CrDDSs

A B C D
Rate-controlling Rate-controlling Rate-controlling Drug Rate-controlling
Drug Drug Drug
surface surface surface reservoir surface
reservoir reservoir reservoir

Drug Drug
Drug Drug
Site-
targeting
moiety

Energy sensor Biochemical responsive/ Biochemical responsive/


Energy sensor Energy sensor

Fig. 2 The four major classes of controlled-release drug delivery systems: (A) Rate-preprogrammed DDS; (B) Activation-modu-
lated DDS; (C) Feedback-regulated DDS and (D) Site-trageting DDS.
1084 Drug Delivery: Controlled Release

of this type have been successfully marketed for thera- 305μ


13.4mm 74 μ
peutical uses and some representatives are outlined
later for illustration.

5.7mm
Ethylene/vinyl acetate membrane
Pilocarpine-core reservoir
Titanium dioxide-white ring
ProgestasertÕ IUD
60
In this controlled-release intrauterine device, the drug

Pilocarpine release rate


reservoir exists as a dispersion of progesterone crystals
in silicone (medical grade) fluid encapsulated in the 40

(mcg/hr)
vertical limb of a T-shaped device walled by a non-
porous membrane of ethylene–vinyl acetate copolymer
(Fig. 4). It is engineered to release continuously a daily 20
dose of 65 mg progesterone inside the uterine cavity to
achieve contraception for one year.[6] The same tech-
nology has been utilized in the development of the 0
MirenaÕ system, a plastic T-shaped frame with a ster- 0 1 2 3 4 5 6 7 8 9
Time (days)
oid reservoir containing 52 mg levonorgestrel, which is
designed to release a daily dose of levonorgestrel at Fig. 5 Diagrammatic illustration of a unit of OcusertÕ sys-
20 mg/day for achieving effective contraception for tem, showing various structural components, and the ocular
five years.[7–9] release rate profile of pilocarpine from the Ocusert pilo-20
system. (From Ref.[11].)
OcusertÕ system

In this controlled-release ocular insert, the drug reser- reservoir compartment to dissolve pilocarpine from the
voir is a thin disc of pilocarpine–alginate complex complex. Pilocarpine molecules are then released at a
sandwiched between two transparent discs of micro- constant rate of 20 or 40 mg/h for a 4- to 7-day man-
porous membrane fabricated from ethylene–vinyl agement of glaucoma.[1,6,10,11]
Drug Delivery
Buccal–Mono

acetate copolymer (Fig. 5). The microporous mem-


branes permit the tear fluid to penetrate into the drug Transderm-NitroÕ system

In this controlled-release transdermal therapeutic


system, the drug reservoir, which is a dispersion of
A
Polyethylene nitroglycerin–lactose triturate in a silicone (medical
grade) fluid, is encapsulated in an ellipsoid-shaped thin
patch. The drug reservoir is sandwiched between a drug-
impermeable metallic plastic laminate, as the backing
Ethylene membrane, and a constant surface of drug-permeable,
vinylacetate rate-controlling membrane of ethylene–vinyl acetate
copolymer copolymer (Fig. 6). This device is fabricated by an
38 mg of progesterone microcrystals injection-molding process. A thin layer of silicone
(and barium sulfate) adhesive is further coated on the drug-permeable
suspended in silicone oil
membrane in order that an intimate contact of the
B drug-releasing surface with the skin surface is achieved
In Vitro In Vivo and maintained. It is engineered to have nitroglycerin
100
delivered transdermally at a rate of 0.5 (mg/cm2)/day
95% Confidence Level
80
100 for a daily relief of angina.[2,3]
μg/day

80
60 60 The same technology has been utilized in the develop-
40 40 ment of the following: 1) the EstradermÕ system, which
20 20
0 0 administers a controlled dose of estradiol transdermally
0 100 200 300 400 0 100 200 300 400 over 3–4 days for the relief of postmenopausal syndrome
Days Days and osteoporosis;[12–14] 2) the DuragesicÕ system, which
Fig. 4 Diagrammatic illustration of a unit of Progestasert provides a transdermal-controlled administration of
IUD, showing various structural components (A) and the fentanyl, a potent narcotic analgesic, for 72-h relief
in vitro and in vivo delivery rate profiles of progesterone of chronic pain;[14] and 3) the AndrodermÕ system,
for up to 400 days (B). which provides a transdermal-controlled delivery of
Drug Delivery: Controlled Release 1085

Drug reservoir Drug-impermeable by either 1) blending a dose of finely ground drug


metallic plastic laminate particles with a viscous liquid (or a semisolid) poly-
mer, followed by a crosslinking of polymer chains
or 2) mixing drug solids with a melted polymer at
Rate-controlling
polymeric membrane
an elevated temperature. The resultant drug-polymer
dispersion is then molded or extruded to form drug-
Adhesive layer
delivery devices of various shapes and sizes designed
for a specific application (Fig. 8). It can also be
fabricated by dissolving the drug and the polymer in a
0.25 common solvent, followed by solvent evaporation, at an
elevated temperature and/or under a vacuum, in a mold.
Plasma nitroglycerin conc.

The release profile of drug from this matrix dif-


0.20
fusion-controlled CrDDS is not constant, because the
(ng/ml ± SD)

rate of drug release is time dependent as defined by:


0.15
Q
Night Period ð2Þ
0.10 t1=2 ¼ ð2ACR DP Þ1=2

where A is the initial loading dose of drug dispersed in


0.05
the polymer matrix; CR is the drug solubility in the
polymer, which is also the drug reservoir concentration
0 in the polymer matrix; and Dp is the diffusivity of the
0 5 10 15 20 25
drug molecules in the polymer matrix.
Time (h)
The release of drug molecules from this type of
CrDDSs may be controlled at a preprogrammed rate
Fig. 6 Cross-sectional view of a unit of Transderm-NitroÕ by controlling the loading level and the polymer
system, showing various structural components, and plasma solubility of the drug and its diffusivity in the polymer

Drug Delivery
concentration profiles of nitroglycerin in 14 human volunteers, matrix. Several CrDDSs of this type have been suc-

Buccal–Mono
each receiving one unit of Transderm-Nitro system (20 cm2, cessfully marketed for therapeutical uses, and some
with a delivery rate of 10 mg/day) for 24 h. (From Refs.[11,55].)
representatives are outlined later for illustration.

testosterone, through non-scrotal skin, for the 24 h Nitro-DurÕ system


replacement therapy of testosterone-deficient patients.[14]
This controlled-release transdermal therapeutic system
NorplantÕ subdermal implant is fabricated by first heating an aqueous solution of
water-soluble polymer, glycerol, and polyvinyl alcohol
The controlled-release subdermal implant is fabricated and then lowering the temperature of the mixture to
from a non-porous silicone (medical-grade) tubing, by form a polymer gel. Nitroglycerin/lactose triturate is
sealing both ends with silicone (medical-grade) dispersed in the gel, and the mixture is then solidified
adhesive to encapsulate either levonorgestrel crystals at room temperature to form a medicated polymer disc
alone (generation I) or a solid dispersion of levonorges- by a molding and slicing technique. After assembly
trel in silicone elastomer matrix (generation II). It is onto a drug-impermeable metallic plastic laminate, a
designed to attain a continuous release of levonorges- patch-type transdermal therapeutic system is produced
trel, at a daily dosage rate of 30 mg, to each subject (fol- with an adhesive rim surrounding the medicated disc
lowing the subcutaneous implantation of either 6 units (Fig. 9). It is designed for application onto an intact
of I or 2 units of II); (Fig. 7) for up to 7 years.[15–18] skin to provide a continuous transdermal infusion of
nitroglycerin, at a daily dose of 0.5 mg/cm2, for the
prevention of angina pectoris.[2,19]
Polymer Matrix Diffusion-Controlled The drug reservoir can also be formulated by
Drug Delivery Systems directly dispersing the drug in an adhesive polymer,
such as poly(isobutylene) or poly(acrylate) adhesive,
In this type of CrDDS, the drug reservoir is produced and then spreading the medicated adhesive by solvent
from the homogeneous dispersion of drug particles in casting or hot melt, onto a flat sheet of drug-impermeable
either a lipophilic or a hydrophilic polymer matrix. The backing support to form a single- or multiple-layer drug
drug dispersion in the polymer matrix is accomplished reservoir. This type of transdermal CrDDS (TDD)
1086 Drug Delivery: Controlled Release

Milligrams Milligrams
216 total load of levonorgestrel 216
~
~ ~
~
90 daily dose 30 μg 90
70 70

50 50
30 30

10 10
1 2 3 4 5 6
Years of use

Concentration of
Levonorgestrel in plasma

ORAL
1.00
Peak (mean)
0.75

0.50
NONPLANT SUBCERNAL INPLANTS
Mean value and
0.25 95% confidence 2.4 mm
intervals
Trough (mean) 34 mm

24 hr 1 2 3 4 5
Time of use (years)
Drug Delivery
Buccal–Mono

Fig. 7 Diagrammatic illustration of the subcutaneous implantation of NorplantÕ implants. The subcutaneous release profile of
levonorgestrel in female volunteers for up to 6 years and the resultant plasma profile as compared to those obtained by oral
administration. (Adapted from Refs.[15–18].)

is best illustrated by the development and marketing delivery of 17b-estradiol for transdermal permeation
of an isosorbide dinitrate-releasing TDD system, for once-weekly treatment of vasomotor systems[14]
named FrandolÕ tape, by Toaeiyo/Yamanouchi in associated with menopause.
Japan, and of a nitroglycerin-releasing TDD system,
named Nitro-DurÕ II system by Key in the United CompudoseÕ implant
States, for once-a-day medication for angina pectoris.
This second generation of TDD system (NitroDur II) This controlled-release subdermal implant is fabricated
has also received FDA approval for marketing. Nitro- by dispersing micronized estradiol crystals in a viscous
Dur II compares favorably with Nitro-Dur (Fig. 10) mixture of silicone elastomer and catalyst and then
and has gradually replaced the first-generation coating the estradiol-polymer dispersion around a rigid
Nitro-Dur from the marketplace. The same technical (drug-free) silicone rod by an extrusion technique to
basis has been also utilized in the development of form a cylinder-shaped implant (Fig. 11). This implant
the following: 1) HabitrolÕ and NicotrolÕ systems, is designed for subcutaneous implantation in the
which provide a controlled dose of nicotine transder- steer’s ear flap for a duration of 200 or 400 days, dur-
mally over 24 h for smoking cessation;[14] 2) MinitranÕ ing which a controlled quantity of estradiol is released
system, which administers a controlled dose of nitrogly- daily for growth promotion.[20]
cerin transdermally over 24 h for the relief of anginal To improve the Q versus t1/2 drug release profiles
attacks;[14] 3) TestodermÕ system, which administers a [Eq. (2)], this polymer matrix diffusion-controlled
controlled delivery of testosterone for transdermal per- CrDDS can be modified to have the drug-loading
meation through a scrotal skin[14] for the replacement level varied, in an incremental manner, to form a gradi-
therapy of testosterone-deficient patients for 24 h; and ent of drug reservoir along the diffusional path in the
4) ClimaraÕ system, which provides a controlled polymer matrix. A constant drug release profile is thus
Drug Delivery: Controlled Release 1087

A B Furthermore, it was recently demonstrated that the


Drug reservoir release of a drug, such as propranolol, from the multila-
(Dispersion) minate adhesive-based TDD system can be maintained
Drug release at zero-order kinetics by controlling the particle size
distribution of drug crystals in the various laminates
of adhesive matrix.[22]

Drug depletion Gel layer


zone Polymer (Membrane/Matrix) Hybrid-Type
Drug Delivery Systems
Drug release
This type of CrDDS is developed with the objective of
combining the constant drug release kinetics of poly-
mer membrane permeation-controlled drug delivery
systems with the mechanical superiority of polymer
C
matrix diffusion-controlled drug delivery systems.
Receding boundary
depletion zone The release profile of drug from a sandwich-type drug
Drug reservoir delivery system (Fig. 13) is constant, and the instan-
Diffusion layer taneous rate of drug release is defined by:
dQ ACp Dp
A Matrix Elution medium ¼  2 ð4Þ
dt Dp Km ð1=Pm þ 1=Pd þ 4ACp Dp t1=2
CR
Dp
Perfect sink where A is the initial amount of drug solid impregnated
in a unit volume of polyer matrix with solubility Cp
hd and diffusivity Dp; Km is the partition coefficient for
the interfacial partitioning of drug molecules from
hp + dhp
polymer matrix toward polymer coating membrane;

Drug Delivery
Pm is the permeability coefficient of the polymer coat-

Buccal–Mono
Fig. 8 Release of drug from the polymer matrix diffusion-
controlled drug delivery systems with drug reservoir exists ing membrane with thickness hm; and Pd is the per-
as a homogeneous dispersion in (A) lipophilic, non-swellable meability coefficient of the hydrodynamic diffusion
polymer matrix, with a growing thickness of drug depletion layer with thickness hd.
zone, or (B) a hydrophilic, swellable polymer matrix, with a The hybrid system is exemplified by the develop-
growing thickness of drug-depleted gel layer. In (C), the drug ment of clonidine-releasing and scopolamine-releasing
concentration gradients across the time-dependent drug transdermal therapeutic systems (Catapres-TTSÕ
depletion zone, with a growing thickness (hp þ dhp), and and Transderm-ScopÕ) (Fig. 14), in which a rate-
the hydrodynamic diffusion layer, with a controlled thickness
controlling non-medicated polymeric membrane is
(hd), are shown in series.
added to coat the surface of the drug-dispersing poly-
mer matrix, and the release of drug molecules thus
achieved, and the rate of drug release from this drug becomes controlled by membrane permeation instead
reservoir gradient-controlled drug delivery system is of matrix diffusion. The same technology has been
defined by: utilized in the development of levonorgestrel-releasing
subdermal implants (NorplantÕ II).
dQ Ka=r Da
¼ Cp ðha Þ ð3Þ
dt ha ðtÞ Microreservoir Partition-Controlled
Drug Delivery Systems
in which the time-dependent thickness [ha(t)] of the
diffusional path for drug molecules to diffuse through, In this type of CrDDS, the drug reservoir is a suspen-
which is increasing with time, is compensated by the sion of drug solid particles in an aqueous solution of a
proportional increase in the drug-loading level [Cp(ha)], water-miscible polymer, like polyethylene glycols. This
and a constant drug release profile is thus obtained. This forms a homogeneous dispersion of many discrete,
type of CrDDS is best illustrated by the nitroglycerin- unleachable, microscopic drug reservoirs in a biocom-
releasing DeponitÕ system (Fig. 12), first marketed patible polymer, like silicone elastomers (Fig. 15). The
by Pharma-Schwartz/Lohmann in Europe.[21] Wyeth- microdispersion is achieved by applying a high-energy
Ayerst has received FDA approval for marketing this dispersion technique.[13,23] Different shapes and sizes of
system in the United States. drug-delivery devices can be fabricated from this
1088 Drug Delivery: Controlled Release

Absorbent pad
Occlusive baseplate Impermeable backing
(aluminum foil) (polyethylene coverstrip)

Adhesive rim
(microporous acrylic polymer tape) Drug reservoir
(drug/hydrophilic polymer matrix)

0.8
Plasma nitroglycerin conc.

0.6
(ng/ml ± SEM)

Night Period
0.4 off

0.2
Drug Delivery
Buccal–Mono

0
0 5 10 15 20 25
Time (h)

Fig. 9 Cross-sectional view of a unit of Nitro-DurÕ system, showing various structural components, and the plasma nitrogly-
cerin concentration profiles in six human volunteers, each receiving 1 unit of Nitro-DurÕ system (20 cm2, with a delivery rate of
10 mg/day) for 24 h. (From Refs.[55–56].)

microreservoir-type CrDDS by molding or extrusion the polymer coating membrane over drug solubility
techniques. Depending upon the physicochemical in the same polymer. Kl, Km, and Kp are, respectively,
properties of drugs and the desired rate of drug release, the partition coefficients for the interfacial partitioning
the device can be further coated with a layer of bio- of drug from the liquid compartments to the polymer
compatible polymer to modify the mechanism and matrix, from the polymer matrix to the polymer coat-
the rate of drug release. ing membrane, and from the polymer coating mem-
The rate of drug release (dQ/dt) from this type of brane to the elution solution, whereas Dl, Dp, and Dd
CrDDS is defined by: are, respectively, the diffusivities of the drug in the
liquid layer surrounding the drug particles, the poly-
dQ Dp Dd mKp
¼ mer coating membrane enveloping the polymer matrix,
dt Dp hd þ Dd hp mKp and the hydrodynamic diffusion layer surrounding the
  
Dl Sl ð1  nÞ 1 1 polymer coating membrane with respective thicknesses
 nSp  þ ð5Þ of hl, hp, and hd (Fig. 15); and Sl and Sp are the solubi-
ht Kl Km
lities of the drug in the liquid compartments and in the
where m ¼ a/b and n is the ratio of drug concen- polymer matrix, respectively.
tration at the inner edge of the interfacial barrier over The release of drug from the microreservoir-type
the drug solubility in the polymer matrix,[1,6] in which a CrDDS can follow either a dissolution- or a matrix
is the ratio of drug concentration in the bulk of elution diffusion-control process, depending upon the relative
solution over drug solubility in the same medium and b magnitude of Sl and Sp.[24] Representatives of this type
is the ratio of drug concentration at the outer edge of of CrDDS is outlined below.
Drug Delivery: Controlled Release 1089

Drug-loaded Impermeable film Silicone rod


adhesive

Release liner
1000
Estradiol-releasing
500 polymer matrix
Nitroglycerin (pg/ml)

250
75%
1.0
100 40%

50 0.8

25 20%

Q (mg/cm2)
0.6
10
0 5 10 15 20 25
Time (h)
0.4
Fig. 10 Cross-sectional view of Nitro-Dur II, showing
various structural components, and the comparative 24 h
plasma nitroglycerin concentration profiles in 24 healthy 0.2
male volunteers, each receiving randomly 1 unit of Nitro-
Dur II (open circle) or Nitro-Dur (closed circle), 20 cm2 each,
with a delivery rate of 10 mg/day, over the chest for 24 h 0.0
(the arrow indicates unit removal). (From Ref.[56].) 0 1 2 3 4
(Days)½

Drug Delivery
Buccal–Mono
Fig. 11 Diagrammatic illustration of a unit of CompudoseÕ
Nitrodisc system Õ subdermal implant and in vitro release profiles of estradiol
from the implants immersed in aqueous solution containing
various volume fractions of polyethylene glycol 400.
In this transdermal CrDDS (Fig. 16), the drug reser-
voir is a suspension of nitroglycerin/lactose triturate
in an aqueous solution of 40% polyethylene glycol
400. It is dispersed homogeneously by a high-energy the suspension is delivered into a silicone medical-grade
mixing technique, with isopropyl palmitate, a skin tubing, which serves as the mold as well as the coating
permeation enhancer, in a mixture of viscous silicone membrane, and then polymerized in situ. The polymer-
elastomer and catalyst.[25] The resultant drug-polymer ized drug-polymer composition is then cut into a
dispersion is then formed in situ into a solid medicated cylinder-shaped implant with its ends staying open
disc on a drug-impermeable metallic plastic laminate, (Fig. 17). This tiny cylindrical implant is designed to
with an adhesive rim, by an injection-molding tech- be inserted into the subcutaneous tissue of the live-
nique and application of an instantaneous heating. It stock’s ear flap; norgestomet is released continuously
is engineered to provide a transdermal administration into the subcutaneous tissue for up to 20 days for the
of nitroglycerin at a daily rate of 0.5 mg/cm2 for control and synchronization of estrus and ovulation
once-a-day medication of angina pectoris.[2,26] AQ and up to 160 days for growth promotion. A constant
versus t1/2 (matrix diffusion-controlled) release profile Q versus t (dissolution-controlled) release profile has
is obtained. been achieved, as compared to the Q versus t1/2 release
profile (matrix diffusion-controlled drug release) for
Syncro-Mate-C implant the Syncro-Mate-B implant and the Nitrodisc system
discussed above.
This subdermal controlled-release implant is fabricated
by dispersing the drug reservoir, which is a suspension Transdermal contraceptive device
of norgestomet in an aqueous solution of PEG 400, in
a viscous mixture of silicone elastomers by a high- The transdermal contraceptive device is based on a
energy dispersion technique.[24] After adding catalyst, patentable micro-drug-reservoir technique[26] to achieve
1090 Drug Delivery: Controlled Release

Drug-impermeable chemical, or biochemical processes and/or facilitated by


metallic plastic laminate an energy supplied externally (Fig. 2). The rate of drug
release is then controlled by regulating the process
applied or energy input. Based on the nature of the
process applied or the type of energy used, these
R1
activation-modulated CrDDSs can be classified into
R2 Drug reservoir the following categories:
gradient layers
R3 (R1 > R2 > R3)
1. Physical means
a. Osmotic pressure-activated drug delivery
Adhesive layer systems
b. Hydrodynamic pressure-activated drug
400 delivery systems
Cmax = 255 ± 151 pg/ml (tmax = 3.6 ± 3.5 h) c. Vapor pressure-activated drug delivery
Css = 125 ± 50 pg/ml (8-24 h) systems
Plasma nitroglycerin conc. (pg/ml)

AUC = 3.3 ± 1.6 ng·h/ml


d. Mechanical force-activated drug delivery
200 systems
e. Magnetics-activated drug delivery systems
f. Sonophoresis-activated drug delivery systems
g. Iontophoresis-activated drug delivery systems
100 h. Hydration-activated drug delivery systems

80 2. Chemical means

60 Dose = 5.0 ± 0.7 mg/day (n = 6) a. pH-activated drug delivery systems


4.5 ± 0.8 mg/day (n = 17) b. pH-activated drug delivery systems
c. Ion-activated drug delivery systems
40 d. Hydrolysis-activated drug delivery systems
Drug Delivery

0 4 8 12 16 20 24
Buccal–Mono

Duration of Device/Skin contact (h) 3. Biochemical means


Fig. 12 Cross-sectional view of a unit of DeponitÕ system, a. Enzyme-activated drug delivery systems
showing various structural components, and the plasma b. Biochemical-activated drug delivery systems
nitroglycerin concentration profiles in six human volunteers,
each receiving 1 unit of Deponit system (16 cm2, with a deliv- Several CrDDSs have been successfully developed
ery rate of 5 mg/day) for 24 h. (Plasma profiles are plotted and applied clinically to the controlled delivery of
from data from Ref.[21].) pharmaceuticals and biopharmaceuticals. These are
outlined and discussed below.

a dual-controlled release of levonorgestrel, a potent


synthetic progestin, and estradiol, a natural estrogen, Osmotic Pressure-Activated Drug
at constant and enhanced rates, continuously, for a Delivery Systems
period of 7 days.[5] By applying 1 unit (10 or 20 cm2)
of transdermal contraceptive device per week, beginning In this type of CrDDSs, the drug reservoir, which can
on day 5 of an individual’s menstrual cycle, for 3 be either a solution or a solid formulation, is con-
consecutive weeks (3 weeks on and 1 week off), steady- tained within a semipermeable housing with a con-
state serum levels of levonorgestrel have been obtained, trolled water permeability. The drug in solution is
and the secretion of gonadotropins and progesterone released through a special laser-drilled delivery orifice
have been effectively suppressed. at a constant rate under a controlled gradient of
osmotic pressure.
For a solution-type osmotic pressure-activated
CrDDS, the intrinsic rate of drug delivery (Q/t) is
ACTIVATION-MODULATED DRUG defined by:
DELIVERY SYSTEMS

In this group of CrDDSs, the release of drug molecules Q Pw Am


¼ ðps  pe Þ ð6Þ
from the delivery systems is activated by some physical, t hm
Drug Delivery: Controlled Release 1091

Sphere

Cylinder

hp(t) hm hd

Drug Delivery
Buccal–Mono
A Drug
reservoir Dp Dm Dd
CR
Perfect sink
pm pd (Cb = 0)

Polymer matrix Drug Polymer Diffusion Solution


depletion coating layer bulk
zone membrane

Fig. 13 The controlled release of drug molecules from a (membrane-matrix) hybrid-type drug delivery system in which solid
drug is homogeneously dispersed in a polymer matrix, which is then encapsulated inside a polymeric membrane, where D, P,
and h are the diffusivity, permeability, and thickness, respectively, and the subscripts p, m, and d denote the drug depletion zone
in the polymer matrix, polymer coating membrane, and diffusion layer, respectively.

For a solid-type osmotic pressure-activated CrDDS,


the intrinsic rate of drug delivery should also be a DRUG-IMPERMEABLE
BACKING LAMINATE
constant and is defined by:
ADHESIVE LAYERS
DRUG-DISPERSING

MICROPOROUS

Q P w Am
¼ ðps  pe ÞSd ð7Þ
MEMBRANE

t hm

where Pw, Am, and hm are, respectively, the water per-


meability, the effective surface area, and the thickness
DRUG MOLECULES
of the semipermeable housing; (ps  pe) is the differen-
tial osmotic pressure between the drug-delivery system Fig. 14 Cross-section view of various structural components
with an osmotic pressure of ps and the environment in the Transderm-ScopÕ and Catapres-TTSÕ systems.
1092 Drug Delivery: Controlled Release

Polymer matrix
(cross-linked, solid)

Drug reservoir
(microscopic liquid
compartments)

Coating membrane

Polymer/Solution interface
Drug Delivery
Buccal–Mono

δl δm δd
Interfacial
barrier

Polymer matrix
Dp
Liquid Dm Ds
layer
Solution
sink
Cp
Dl Cp' Cm
Cp'

Drug
particle
Cd
Cb = O
Sl Cl Cm'

Polymer Diffusion
coating layer
membrane

Fig. 15 Microscopic view of a microreservoir-type drug-delivery system, which shows the microscopic structure of various com-
ponents, and the physical model developed for the mechanistic analysis of the controlled release of drug. (Adapted from Refs.[1,57].)

with an osmotic pressure of pe; and Sd is the aqueous effective surface area of the semipermeable housing as
solubility of the drug component in the solid reservoir. well as the osmotic pressure gradient. Several CrDDSs
The release of drug molecules from this type of of this type have been successfully marketed for thera-
CrDDS is activated by osmotic pressure and controlled peutical uses and some representatives are outlined
at a rate determined by the water permeability and the later.
Drug Delivery: Controlled Release 1093

Occlusive baseplate Adhesive foam pad Drug


(aluminum foil disc) (flexible polyurethane) reservoir
Polymer coating
membrane

Microscopic drug reservoirs


(drug/co-solvents)
Adhesive rim
(acrylic polymer coating) Polymer matrix Open ends Medicated MDD core
(silicone elastomer)
100
0.6

Fraction of drug released (%)


0.5 80
Plasma nitroglycerin
conc. (ng/ml ± SEM)

Night Period
0.4
60
0.3

0.2 40
(C plasma)ss
0.1
20
0.0
0 4 8 12 16 20 24 28 32
Time (h) 0
0 5 10 15 20 25 30
Fig. 16 Cross-sectional view of a unit of Nitrodisc system, Õ Days of implantation
showing various structural components, and the plasma
Fig. 17 Syncro-Mate-C implant, a subdermal implant fabri-
nitroglycerin concentration profiles in 12 human volunteers,
cated from the microreservoir dissolution-controlled drug-
each receiving 1 unit of Nitrodisc system (16 cm2, with a
delivery system, and subcutaneous controlled release of nor-
delivery rate of 10 mg/day) for 32 h. (From Ref.[23].)
gestomet, a potent synthetic progestin, at constant rate for 20
days. The open ends on the implant do not affect the zero-
order in vivo drug release profile. (Adapted from Ref.[57].)

Drug Delivery
Buccal–Mono
Alzet osmotic pump
AcutrimÕ tablet
In this implantable or insertable CrDDS, the drug res-
ervoir, which is normally a solution formulation, is In this oral CrDDS, the drug reservoir, which is a solid
contained within a collapsible, impermeable polyester tablet of water-soluble and osmotically-active phenyl-
bag whose external surface is coated with a layer of propanolamine (PPA) HCl, is enclosed within a semi-
osmotically active salt, for example, sodium chloride. permeable membrane of cellulose triacetate.[2,30] The
This reservoir compartment is then totally sealed inside surface of the semipermeable membrane is further
a rigid housing walled with a semipermeable mem- coated with a thin layer of immediately releasable
brane (Fig. 19). At an implantation site, the water PPA dose (Fig. 20). In the alimentary tract, the gastro-
content in the tissue fluid will penetrate through the intestinal fluid will dissolve away the immediate release
semipermeable membrane at a controlled rate and layer of PPA to provide an initial dose of PPA and
dissolve the osmotically active salt. This creates an then penetrate through the semipermeable membrane
osmotic pressure in the narrow spacing between the to dissolve the sustained-release dose of PPA. Under
flexible reservoir wall and the rigid semipermeable the osmotic pressure created [Eq. (7)], the PPA solution
housing. Under the osmotic pressure created [Eq. (6)], is released continuously at a controlled rate, through
the reservoir compartment is thus reduced in volume an orifice pre-drilled by a laser beam.[2,30,31] It is
and the drug solution is forced to release through the designed to provide a controlled delivery of PPA over
flow moderator at a controlled rate.[27,28] By varying a duration of 16 h for appetite suppression in a weight-
the drug concentration in the solution, different doses control program.[31] The same delivery system has
of drug can be delivered at a constant rate for a period also been utilized for the oral controlled delivery of
of 1–4 weeks. indomethacin. An extension of this technology is the
In addition to its application in the subcutaneous development of a push-pull type osmotic pressure-
controlled administration of drugs for pharmacologi- activated CrDDS for the oral controlled delivery of
cal studies, this technology has recently been extended nifedipine and metroprolol.[27] It has been further
to the controlled administration of drugs in the rectum extended to the delayed-onset and controlled oral
by zero-order kinetics. The hepatic first-pass metab- delivery of verapamil[14] to produce a maximum
olism of drugs is thus bypassed.[29] plasma concentration in the morning hours.
1094 Drug Delivery: Controlled Release

A hydrophilic polymer layer is sandwiched between the


288 drug reservoir compartment and the housing. In the
10 Sq. cm - Patch (n=6) gastrointestinal tract, the laminate will imbibe the gas-
Concentration (pg/ml ± S.E.)

240 20 Sq. cm - Patch (n=6) trointestinal fluid through the annular openings at the
lower end of the housing and become swollen. This
192 generates a hydrodynamic pressure in the system.
The hydrodynamic pressure, thus created, forces the
144
drug reservoir compartment to reduce in volume and
96 causes the liquid drug formulation to release through
the delivery orifice.[32] The drug release rate is
48 defined by:

0 Q Pf Am
0 168 336 504 672 ¼ ðys  ye Þ ð8Þ
m mm mm m p p
t hm
Duration of study (h)
where Pf, Am, and hm are the fluid permeability, the
B effective surface area, and the thickness of the wall
30
Subject Code MM Subject Code MG
with annular openings, respectively; and ys  ye, is
the difference in hydrodynamic pressure between the
Serum progesterone (ng/ml)

(Group A: 1 TCD patch) (Group B: 2 TCD patch)


25 drug delivery system (ys) and the environment (ye).
Pretreatment Pretreatment
Treatment Treatment The release of drug molecules from this type of
20
CrDDS is activated by hydrodynamic pressure and
15 controlled at a rate determined by the fluid per-
meability and effective surface area of the wall with
10 annular openings as well as by the hydrodynamic
Tmax
Tmax pressure gradient.
5
Drug Delivery
Buccal–Mono

0 Vapor Pressure-Activated
0 10 20 30 40 0 10 20 30 40
Day of menstrual cycle Drug Delivery Systems

Fig. 18 (Upper panel) The 4–week serum levonorgestrel In this type of CrDDS, the drug reservoir, which is a
profiles in 12 human volunteers, each receiving 1 or 2 units solution formulation, is contained inside the infusion
of a transdermal contraceptive system (10 cm2, with daily compartment. It is physically separated from the
dosage of 28.3 mg/day) once a week, consecutively for 3 pumping compartment by a freely movable partition
weeks, and the same size of placebo on week 4. (Lower panel)
(Fig. 21). The pumping compartment contains a vapor-
Comparative serum concentration profiles of progesterone
during the pretreatment and treatment cycles in two subjects,
izable fluid, such as fluorocarbon, which vaporizes at
each as the representative for group A (receiving 10 cm2) and body temperature and creates a vapor pressure. Under
group B (receiving 20 cm2), respectively. The suppression of the vapor pressure created, the partition moves upward
progesterone peak during the treatment cycle is an indication and forces the drug solution in the infusion compart-
of effective fertility control. ment to be delivered, through a series of flow regulator
and delivery cannula, into the blood circulation at a
constant flow rate.[1,6,33] The process is defined by:
Hydrodynamic Pressure-Activated
Drug Delivery Systems Q d4 dP
¼ ð9Þ
t 40:74ml
In addition to the osmotic pressure systems discussed
above, hydrodynamic pressure has also been explored where d and l are, respectively, the inner diameter and
as the potential source of energy to modulate the deliv- thelength of the delivery cannula; Dp is the pressure
ery of therapeutic agents.[2] difference between the vapor pressure in the pumping
A hydrodynamic pressure-activated drug-delivery compartment and the pressure at the implantation site;
system can be fabricated by placing a liquid drug for- and m is the viscosity of the drug formulation.
mulation inside a collapsible, impermeable container The delivery of drug from this type of CrDDS is
to form a drug reservoir compartment. This is then activated by vapor pressure and controlled at a rate
contained inside a rigid, shape-retaining housing. A determined by the differential vapor pressure, the for-
laminate of an absorbent layer and a swellable, mulation viscosity, and the size of the delivery cannula.
Drug Delivery: Controlled Release 1095

A B
200
Urine volume
(% of Pretreatment control + S.D.)
Drug solution leaving
175 via delivery portal

150
Daily urine volume

Pump Pump
Removable cap
125 implanted removed Flange
Flow moderator
100
Neck Plug
75

50

25 Flexible impermeable
reservoir wall
0
0 3 6 9 12 15 Osmotic agent

3000
Urine osmolality

2500
(mOsm/ kg H2O + S.D.)

Semipermeable
Urine osmolalilty

2000 membrane
Water entering
1500
semipermeable
membrane
1000
Pump Pump Reservoir
implanted removed
500

0
0 3 6 9 12 15

Drug Delivery
Time (day)

Buccal–Mono
Fig. 19 (A) Cross-sectional view of the AlzetÕ osmotic pump, an osmotic pressure-activated drug-delivery system. (B) The effect
of 7 days of subcutaneous delivery of antidiuretic hormone (vasopressin) on the daily volume of urinary excretion and urine
osmolality in the Brattleboro rats with diabetes insipidus.

A typical example is the development of InfusaidÕ, its synthetic analogs, such as buserelin. Through nasal
an implantable infusion pump by Metal Bellows, for absorption, the hepatic first-pass elimination of these
the constant infusion of heparin in anticoagulation peptide drugs is thus avoided.[24]
treatment,[34] of insulin in the normoglycermic control
of diabetics,[33] and of morphine for patients suffering
from the intensive pain of a terminal cancer.[35] Magnetic-Activated Drug Delivery Systems

Macromolecular drugs, such as peptides, have been


Mechanical Force-Activated known to release only at a relatively low rate from a
Drug Delivery Systems polymer-controlled drug-delivery system. This low rate
of release can be improved by incorporating an electro-
In this type of CrDDS, the drug reservoir is a solution magnetism-triggering vibration mechanism into the
formulation in a container equipped with a mechani- polymeric delivery device. With a hemispheric-shaped
cally activated pumping system. A metered dose of design, a zero-order drug-release profile is achieved.[36]
drug formulation can be reproducibly delivered into By combining these two approaches, a subdermally
a body cavity, such as the nose, through the spray head implantable, magnetic-activated hemispheric drug-
upon manual activation of the drug-delivery pumping delivery device is developed. It is fabricated by first
system. The volume of solution delivered is fixed and positioning a tiny doughnut-shaped magnet at the cen-
is independent of the force and duration of activation. ter of a drug-dispersing biocompatible polymer matrix
A typical example of this type of drug-delivery sys- and then coating the external surface of the medicated
tem is the development of a metered-dose nebulizer for polymer matrix, with the exception of one cavity at the
the intranasal administration of a precision dose of center of the flat surface, with a pure polymer, for
luteinizing hormone-releasing hormone (LHRH) and instance, ethylene–vinyl acetate copolymer or silicone
1096 Drug Delivery: Controlled Release

Delivery orifice molecule across a biological membrane, such as the


Semi-permeable coating
skin, in a manner similar to passive diffusion under a
Drug reservoir/ Controlled-release concentration gradient but at a much facilitated rate.
osmotically active dose Immediate releasing layer The iontophoresis-facilitated skin permeation rate of
solutes (initial dose)
a charged molecule i consists of three components
and is expressed by:
Cumulative % loading dose released

100 12.16 atm


30.16 atm

80
54.16 atm
Jiisp ¼ J p þ J e þ J c
114.0 atm    
dC Zi Di Fi dE
¼ Ks Ds Ci þ ðkCs Id Þ ð10Þ
60 hs RT hs

40 where J p, J e, and J c represent, respectively, the flux for


the skin permeation by passive diffusion, for the elec-
20 trical current-driven permeation, and for the convec-
tive flow-driven skin permeation; Ks is the partition
0 coefficient for interfacial partitioning from the donor
4 8 12 16 20 24
Time (h) solution to the stratum comeum; Ds and Di are,
respectively, the diffusivity across the skin and the
Fig. 20 Cross-sectional view of a unit of AcutrimÕ tablet, a diffusivity of ionic species i in the skin; Ci and Cs are,
solid-type osmotic pressure-activated drug delivery system, respectively, the donor concentration of ionic species i
and the effect of increased osmotic pressure in the dissolution and the concentration in the skin tissue; dE/hs is the
medium on the release profiles of phenylpropanolamine HCl
electrical potential gradient across the skin; dC/hs is
from the Acutrim tablet at intestinal condition. (Adapted
the concentration gradientacross the skin; Zi is the
from Refs.[31,58].)
electrical valence of ionic species i; Id is thecurrent
density applied; F, k, and R are, respectively, the fara-
elastomers. This uncoated cavity is designed for allow- day, proportionality, and gas constant; and T is the
Drug Delivery

ing a peptide drug to release. absolute temperature.


Buccal–Mono

The hemispheric magnetic delivery device produced A typical example of this type of activation-
can release macromolecular drugs, like bovine serum controlled CrDDS is the development of an iontophore-
albumin, at a low basal rate, by diffusion process, and tic drug delivery system, named Phoresor by Motion
under a non-triggering condition, or it can release the Control, to facilitate the percutaneous penetration of
same drug at a much higher rate, when the magnet is acti- antiinflammatory drugs, such as dexamethasone sodium
vated, to vibrate by an external electromagnetic field. phosphate,[39–41] to surface tissues.
Further development of the iontophoresis-activated
drug delivery technique has yielded a new design of
Sonophoresis-Activated Drug iontophoretic drug delivery system—the transdermal
Delivery Systems periodic iontotherapeutic system (TPIS). This new sys-
tem, which is capable of delivering a physiologically-
This type of activation-controlled drug delivery system acceptable pulsed direct current, in a periodic manner,
utilizes ultrasonic energy to activate (or trigger) the with a special combination of waveform, intensity, fre-
delivery of drugs from a polymeric drug delivery quency, and on/off ratio, for a specific duration, has
device. The system can be fabricated from either a significantly improved the efficiency of transdermal
non-degradable polymer, such as ethylene–vinyl acet- delivery of peptide and protein drugs.[4] A typical
ate copolymer, or a bioerodible polymer, such as poly example is the iontophoretic transdermal delivery of
[bis(p-carboxyphenoxy)alkane anhydride].[37] The insulin, a protein drug, in the control of hyperglycemia
potential application of sonophoresis (or phonophor- in diabetic animals.
esis) to regulate the delivery of drugs was recently
reviewed.[38]

Hydration-Activated Drug Delivery Systems


Iontophoresis-Activated Drug
Delivery Systems In this type of CrDDS, the drug reservoir is homoge-
neously dispersed in a swellable polymer matrix fabri-
This type of CrDDS use electrical current to activate cated from a hydrophilic polymer. The release of drug
and to modulate the diffusion of a charged drug is activated and modulated by hydration-induced
Drug Delivery: Controlled Release 1097

Bacterial Inlet
filter septum
assembly Needle stop

Fluorocarbon Flow
fluid regulator
filling tube
2.4 cm Silicone
polymer
coating

Empty Infusate Bellows


weight = 181 g Fluorocarbon
chamber
fluid
chamber

8.6 cm

1000
A
Pump
800
implanted

600

400
n=7
Dose units/kg per day

200

0
1000
B

Drug Delivery
Buccal–Mono
800

600

400
n = 25
200

0
0 8 16 24 32 40 48 56
Weeks of infusion

Fig. 21 Cross-sectional view of a unit of InfusaidÕ system, a vapor pressure-activated drug-delivery system, and daily heparin
dose (mean  S.E.) delivered to 25 dogs for 6 months and to 7 dogs for 12 months. (Adapted from Ref.[34].)

swelling of the polymer matrix. Representatives of this engineered to deliver norgestomet, at a rate of
type of CrDDS are outlined below. 504 mg/cm2/day1/2, in the subcutaneous tissue for up
to 16 days for the control and synchronization of
Syncro-Male-B implant estrus in livestock.[13]

This subcutaneous CrDDS is fabricated by dissolving ValreleaseÕ tablet


norgestomet, a potent progestin for estrus synchro-
nization, in an alcoholic solution of linear ethylene This oral CrDDS is prepared by granulating Valium, an
glycomethacrylate polymer (Hydron S). The drug- antidepression drug, with hydrocolloids (20–75 wt%)
polymer mixture is then cross-linked by adding and pharmaceutical excipients. The granules are then
ethylene dimethacrylate, in the presence of an oxidizing compressed to form an oral tablet. After oral intake,
catalyst, to form a cylinder-shaped subdermally the hydrocolloids absorb the gastric fluid and are
implantable implant.[1,6] This tiny subdermal implant activated to form a colloid gel matrix surrounding the
can be activated by tissue fluid to swell and can be tablet surface (Fig. 22). The release of Valium molecules
1098 Drug Delivery: Controlled Release

is then controlled by diffusion through the gel barrier, intestine, and the intestinal fluid-soluble component
while the tablet remains buoyant in the stomach, due in the coating membrane is dissolved away by the
to a density difference between the gastric fluid intestinal fluid (pH >7.5). This produces a micropor-
(d > 1) and the gelling tablet (d < 1).[2,3] ous membrane of intestinal fluid-insoluble polymer
to control the release of drug from the core tablet.
The drug is thus delivered in a controlled manner in
pH-Activated Drug Delivery Systems the intestine by a combination of drug dissolution
in the core and diffusion through the pore channels.
For a drug labile to gastric fluid or irritating to gastric By adjusting the ratio of the intestinal fluid-soluble
mucosa, this type of CrDDS has been developed to tar- polymer to the intestinal fluid-insoluble polymer in
get the delivery of the drug only in the intestinal tract, the membrane, the rate of drug delivery can be regu-
not in the stomach.[2] It is fabricated by coating a core lated. Representative application of this type of CrDDS
tablet ofthe gastric fluid-sensitive drug with a combi- is in the oral controlled delivery of potassium chloride,
nation of intestinal fluid-insoluble polymer, like ethyl which is highly irritating to gastric epithelium.
cellulose, and intestinal fluid-soluble polymer, like
hydroxylmethyl cellulose phthalate (Fig. 23).
In the stomach, the coating membrane resists the Ion-Activated Drug Delivery Systems
degrading action of gastric fluid (pH <3), and the drug
molecules are thus protected from the acidic degradation. For controlling the delivery of an ionic or an ionizable
After gastric emptying, the CrDDS travels to the small drug, this type of CrDDS has been developed.[2]
Because the gastrointestinal fluid has regularly main-
tained a relatively constant level of ions, the delivery
A
of drug by this type of CrDDS can be modulated, the-
oretically, at a constant rate.
Hydrocolloids
Such a CrDDS is prepared by first complexing an
(20–75% w/w)
ionizable drug with an ion-exchange resin, such as
Gastric fluid (d > 1)
Drug Delivery
Buccal–Mono

Stomach
(pH < 3)
Colloid gel barrier
d< 1 Gastric emptying

B Coating of

Gastric fluid- Intestinal fluid-insoluble polymer


2000 labile drug
Intestinal fluid-soluble polymer
Radioactivity in stomach

1000
800
Valrelease Intestinal fluid
600 (pH > 7.5)
(Placebo)
400

Valium
200 (Placebo) Microporous membrane
of intestinal fluid-
Gastric fluid- insoluble polymer
100 labile drug
0 1 2 3 4 5 6
Time (h)

Fig. 22 (A) Schematic illustration of ValreleaseÕ tablet, a Drug


swelling-activated drug-delivery system, and the hydration-
induced formation of colloid gel barrier. (B) Comparison in Fig. 23 Schematic illustration of a pH-activated drug-
the gastric residence profile between Valrelease with the delivery system and the pH-dependent formation of micro-
conventional Valium capsule. (From Ref.[58].) porous membrane in the intestinal tract.
Drug Delivery: Controlled Release 1099

complexing a cationic drug with a resin containing made possible by the enzymatic hydrolysis of biopoly-
SO3 group or an anionic drug with a resin containing mers by a specific enzyme in the target tissue.[46–48] A
N(CH3)3þ group. The granules of the drug–resin com- typical example is the development of albumin micro-
plex are further treated with an impregnating agent, spheres, which release 5-fluorouracil, in a controlled
like polyethylene glycol 4000, for reducing the rate of manner, by protease-activated biodegradation.
swelling upon contact with an aqueous medium. They
are then coated by an air-suspension coating technique
with a water-insoluble but water-permeable polymeric FEEDBACK-REGULATED DRUG
membrane, such as ethylcellulose. This membrane DELIVERY SYSTEMS
serves as a rate-controlling barrier to modulate the
release of drug from the CrDDS. In the GI tract, In this group of CrDDSs, the release of drug molecules
hydronium and chloride ions diffuse into the CrDDS is activated by a triggering agent, such as a biochemical
and interact with the drug–resin complex to trigger substance, in the body via some feedback mechanisms
the dissociation and release of ionic drug (Fig. 24). (Fig. 2). The rate of drug release is regulated by the con-
This type of CrDDS is exemplified by the devel- centration of a triggering agent detected by a sensor
opment of PennkineticÕ system (by Pennwalt Phar- built into the CrDDS.
maceuticals), which permits the formulation of oral
liquid-type dosage forms with sustained release of a
combination of hydrocodone and chlorpheniramine Bioerosion-Regulated Drug
(TussionexÕ).[14,42–44] Delivery Systems

The feedback-regulated drug delivery concept has been


Hydrolysis-Activated Drug applied to the development of a bioerosion-regulated
Delivery Systems CrDDS by Heller and Trescony.[49] This CrDDS con-
sists of a drug-dispersed bioerodible matrix fabricated
This type of CrDDS depends on the hydrolysis process from poly(vinyl methyl ether) half-ester, which was
to activate the release of drug molecules. In this sys- coated with a layer of immobilized urease (Fig. 26).
tem, the drug reservoir is either encapsulated in micro- In a solution with near neutral pH, the polymer only

Drug Delivery
capsules or homogeneously dispersed in microspheres

Buccal–Mono
erodes very slowly. In the presence of urea, urease at
or nanoparticles. It can also be fabricated as an
implantable device. All these systems are prepared
from a bioerodible or biodegradable polymer, such as Drug-resin complex particles
polylactide, poly(lactide–glycolide) copolymer, poly
Resin – SO3– Drug+
(orthoester), or poly(anhydride). The release of a drug + ]Drug–
Resin [N(CH3)3
from the polymer matrix is activated by the hydrolysis-
induced degradation of polymer chains, and the rate of Polyethylene glycol treatment
drug delivery is controlled by polymer degradation
rate.[45] A typical example is the development of Ethyl cellulose coating
Lupron DepotÕ, an injectable microspheres for the
subcutaneous controlled delivery of luprolide, a potent
biosynthetic analog of gonadotropin-releasing hormone Blood
Gut wall
(GnRH) for the treatment of gonadotropin-dependent Polymer
cancers, such as prostate carcinoma in men and endome- Drug
triosis in the females, for up to 4 months. Another
example is the development of ZoladexÕ system, an
implantable cylinder for the subcutaneous controlled
Ion
delivery of goserelin, also a potent biosynthetic analog Coating
of GnRH for the treatment of patients with prostate membrane
cancer (Fig. 25) for up to 3 months.[14]
H + + Resin – SO3– Drug+ Resin–SO3– H + + Drug +
Enzyme-Activated Drug Delivery Systems
CI – + Resin [N(CH3)3 + ] Drug – Resin [N(CH3)3+ ] CI – + Drug–

In this type of CrDDS, the drug reservoir is either phys- Fig. 24 Cross-sectional view of an ion-activated drug-
ically entrapped in microspheres or chemically bound delivery system, showing various structural components,
to polymer chains fabricated from biopolymers, such and diagrammatic illustration of ion-activated drug release.
as albumins or polypeptides. The release of drugs is (Adapted from Ref.[58].)
1100 Drug Delivery: Controlled Release

Drug-dispersed Poly (vinyl methyl ether) half-ester


polymer matrix u u u u u (monolithic matrix)

Hydrolytic erosion u u
Phase I: surface erosion u Hydrocortisone u
Micropores Urease
Phase II: bulk erosion u u
(Immobilized)
u u u u u
goserelin
urease
Glp His Trp Ser Tyr (D)
Leu Arg Pro Azgly NH urea 2NH4+ + HCO3− + OH–
Ser H 2O
t-Bu alkaline
polymer erosion
pH
Serum Testosterone
Serum LH (lU/L)

40 30 Hydrocortisone
(nmol/L)

20 15
100

Hydrocortisone released (%)


0 0
80
0 4 8 12 0 4 8 12
Weeks Weeks
60
Fig. 25 Amino acid sequence of goserelin, a biosynthetic
analog of gonadotropin-releasing hormone, and the effect urea (0.1 M)
40
of subcutaneous controlled release of goserelin from the bio-
degradable poly(lactide-glycolide) implant on the serum
20
levels of luteinizing hormone and testosterone.

0
0 20 40 60 80 100 120 140 160
the surface of the drug delivery system metabolizes
Time (h)
urea to form ammonia. This causes the pH to increase
and activates a rapid degradation of polymer matrix as
Drug Delivery

Fig. 26 Cross-sectional view of a bioerosion-regulated


Buccal–Mono

well as the release of drug molecules. hydrocortisone delivery system, a feedback-regulated drug
delivery system, showing the drug-dispersed monolithic
bioerodible polymer matrix with surface-immobilized
Bioresponsive Drug Delivery Systems ureases. The mechanism of release and time course for the
urea-activated release of hydrocortisone are also shown.
The feedback-regulated drug delivery concept has also (From Ref.[49].)
been applied to the development of a bioresponsive
CrDDS by Horbett et al.[50]. In this CrDDS, the drug
reservoir is contained in a device enclosed by a biore- Self-Regulating Drug Delivery Systems
sponsive polymeric membrane whose permeability to
drug molecules is controlled by the concentration of This type of feedback-regulated CrDDS depends on a
a biochemical agent in the tissue where the CrDDS is reversible and competitive binding mechanism to acti-
located. vate and to regulate the release of drug. In this CrDDS,
A typical example of this bioresponsive CrDDS is the drug reservoir is a drug complex encapsulated
the development of a glucose-triggered insulin delivery within a semipermeable polymeric membrane. The
system, in which the insulin reservoir is encapsulated release of drug from the CrDDS is activated by the
within a hydrogel membrane containing pendant membrane permeation of a biochemical agent from
NR2 groups (Fig. 27). In an alkaline solution, the the tissue where the CrDDS is located.
NR2 groups exist at neutral state and the membrane Kim et al. first applied the mechanism of reversible
is unswollen and thus impermeable to insulin. As binding of sugar molecules with lectin into the design
glucose penetrates into the membrane, it is oxidized of self-regulating CrDDS.[51] For this CrDDS, a biolo-
enzymatically by the glucose oxidase entrapped in the gically-active insulin derivative, in which insulin is
membrane to form gluconic acid. This process triggers coupled with a sugar (e.g., maltose), was first prepared
the protonation of NR2 groups to form NR2Hþ, and and then conjugated with lectin to form an insulin–
the hydrogel membrane becomes swollen and is thus sugar–lectin complex. The complex is then encapsu-
permeable to insulin molecules (Fig. 27). The amount lated within a semipermeable membrane to produce
of insulin delivered is bioresponsive to the concen- CrDDS. As blood glucose diffuses into the CrDDS, it
tration of glucose penetrating into the CrDDS. binds, competitively, with the binding sites in the lectin
Drug Delivery: Controlled Release 1101

molecules and activates the release of the insulin–sugar Self-Regulating Insulin Delivery Systems
derivatives from the binding sites. The released insulin-
sugar derivatives diffuse out of the CrDDS, and the (Biochemical Approach)
amount of insulin-sugar derivatives released depends
on the concentration of glucose. Thus, a self-regulating Concanavalin A Glycosylated (G) insulin
drug delivery is achieved. However, a potential prob-
lem has remained to be resolved: that is, the release + Glucose in
Polymer
of insulin is non-linear in response to the changes in membrane
G-Insulin out
glucose level.[52]
Further development of the self-regulating insulin
delivery system has utilized the complex of glycosy-
lated insulin–concanavalin A, which is encapsulated Pancreatectomized dogs
inside a polymer membrane.[53] As glucose penetrates

Blood glucose level (mg/dl)


into the system, it activates the release of glycosylated 300
insulin from the complex for a controlled release from
the system (Fig. 28). The amount of insulin released is
thus self-regulated by the concentration of glucose that 200
has penetrated into the insulin delivery system.

100 Normoglycemic
level
SITE-TARGETING DRUG DELIVERY SYSTEMS
F F F F F F = Feeding

Delivery of a drug to a target tissue that needs medi- 9am 3pm 9pm 3am 9am 3am 9pm
cation is known to be a complex process that consists Time of day

Fig. 28 Various components of a self-regulating insulin


delivery system, a feedback-regulated drug delivery system,
Amine-containing
hydrogel membrane and its control of blood glucose level in the pancreatecto-

Drug Delivery
Buccal–Mono
Glucose oxidase mized dogs. (From Ref.[53].)
NR2 ......
.. ..
.... ..... Glucose
NR
NR2 . 2 of multiple steps of diffusion and partitioning. The
........
.
NR2 CrDDSs outlined above generally address only the first
..........
NR2 step of this complex process. Essentially, these
CrDDSs have been designed to control the rate of drug
Insulin Glucose Oxidase Gluconic acid
release from the delivery systems, but the path for the
reservoir transport of drug molecules from the delivery system
–NR2 H+ +
–N R2H
Acidic pH to the target tissue remains largely uncontrolled.
Hydrogel membrane swells
Ideally, the path of drug transport should also be
under control. Then, the ultimate goal of optimal treat-
ment with maximal safety can be achieved. This can be
+ N Enzyme
N R2 ... reasonably accomplished by the development of a
H ...............
CrDDS with a site-targeting specificity (Fig. 2). An
........... +
.. N R2 ideal site-targeting CrDDS has been proposed by
+ H
N R2 Ringsdorf.[54] A model, which is shown in Fig. 29, is
H
Insulin constructed from a non-immunogenic and biodegrad-
+
N R2 able polymer and acts as the backbone to contain three
H .......... +
.. N R2 types of attachments: 1) a site-specific targeting moiety,
H
which is capable of leading the drug delivery system to
Swollen membrane the vicinity of a target tissue (or cell); 2) a solubilizer,
which enables the drug delivery system to be trans-
Fig. 27 Cross-sectional view of a bioresponsive insulin
delivery system, a feedback-regulated drug delivery system,
ported to and preferentially taken up by a target tissue;
showing the glucose oxidase-entrapped hydrogel membrane and 3) a drug moiety, which is convalently bonded to
constructed from amine-containing hydrophilic polymer. the backbone, through a spacer, and contains a linkage
The mechanism of insulin release, in response to the influx that is cleavable only by a specific enzyme(s) at the
of glucose, is also illustrated. (From Ref.[50].) target tissue.
1102 Drug Delivery: Controlled Release

Polymer backbone of Pharmacy, Proceedings Published in Drug Develop. &


(nonimmunogenic Ind. Pharm. 1983; 9, 1077–1396.
and blodegradable) 4. Chien, Y.W. International Pharmaceutical R&D
Symposium on Advances in Transdermal Controlled Drug
Administration for Systemic Medication. Piscataway, New
Jersey, June 20&21, 1985; Rutgers University, College of
Pharmacy, Proceedings Published in Transdermal Controlled
Systemic Medications; Chien, Y.W., Ed.; Marcel Dekker, Inc.:
Site-specific Spacer Solubilizer New York, 1987.
targeting
moiety 5. Chien, Y.W. Rate-control drug delivery systems: controlled
Cleavable Facilitate systemic release vs. sustained release. Med. Prog. Technol. 1989,
group distribution 15 (1–2), 21–46.
and tissue uptake 6. Chien, Y.W. Novel Drug Delivery Systems: Second Edition,
Revised and Expanded; Marcel Dekker, Inc.: New York,
1992.
Enzyme Drug 7. Luukkainen, T.; Allonen, H.; Haukkamaa, M.;
(at target tissue) Lahteenmaki, P.; Nilsson, C.G.; Toivonen, J. Five years’
experience with levonorgestrel-releasing IUDs. Contracep-
tion 1986, 33 (2), 139–148.
Cell of Drug 8. Andersson, K.; Odlind, V.; Rybo, G. Levonorgestrel-
target tissue releasing and copper-releasing (Nova T) IUDs during five
years of use: a randomized comparative trial. Contracep-
tion 1994, 49 (1), 56–72.
Drug 9. Sivin, I.; Stern, J. Health during prolonged use of levonor-
gestrel 20 micrograms/d and the copper TCu 380Ag
Cell membrane intrauterine contraceptive devices: a multicenter study.
International committee for contraception research
(ICCR). Fertil. Steril. 1994, 61 (1), 70–77.
10. Robinson, J.R. Sustained and Controlled Release Drug
Delivery Systems; Marcel Dekker, Inc.: New York, 1978.
11. Baker, R.W.; Lonsdale, H.K. Controlled delivery—an
emerging use for membranes. Chemtech. 1975, 5, 668.
12. Chien, Y.W. Methods to achieve sustained drug delivery.
The physical approach: implants. In Sustained and Con-
Fig. 29 An ideal site-targeting controlled-release drug trolled Release Drug Delivery Systems; Robinson, J.R.,
delivery. (From Ref.[54].) Ed.; Marcel Dekker, Inc.: New York, 1978.
13. Chien, Y.W. Microsealed Pharmaceutical Delivery Device.
Drug Delivery
Buccal–Mono

US Patent 3,992,518, Nov 16, 1976.


14. Physicians’ Desk Reference, 53rd Ed.; Medical Economics
Unfortunately, this ideal site-targeting CrDDS is Company. Montvale, 1999.
only in the conceptual stage. Its construction remains 15. Segal, S.J. The development of NORPLANT implants.
largely unresolved and is still a challenging task in Stud. Fam. Plann. 1983, 14 (6–7), 159–163.
16. Diaz, S.; Pavez, M.; Miranda, P.; Robertson, D.N.; Sivin, I.;
the biomedical and pharmaceutical sciences. Croxatto, H.B. A five-year clinical trial of levonorgestrel
silastic implants (Norplant2). Contraception 1982, 25 (5),
447–456.
CONCLUSIONS 17. Weiner, E.; Victor, A.; Johansson, E.D. Plasma levels of D-
norgestrel after oral administration. Contraception 1976,
14 (5), 563–570.
The controlled-release drug delivery systems outlined 18. Croxatto, H.B.; Diaz, S.; Miranda, P.; Elamsson, K.;
here have been steadily introduced into the biomedical Johansson, E.D. Plasma levels of levonorgestrel in women
during longterm use of Norplant. Contraception 1981,
community since the middle of the 1970s. There is a 23 (2), 197–209.
growing belief that many more of the conventional 19. Keith, A.D. Polymer matrix considerations for transdermal
drug delivery systems we have been using for decades devices. Drug Develop. & Ind. Pharm. 1983, 9, 605–625.
20. Hsieh, D.S.T. Subcutaneous controlled delivery of estradiol
will be gradually replaced in the coming years by these by compudose implants: in vitro and in vivo evaluations.
CrDDSs. Drug Develop. & Ind. Pharm. 1987, 13, 2651–2666.
21. Wolff, M.; Cordes, G.; Luckow, V. In vitro and in vivo
release of nitroglycerin from a new transdermal therapeutic
system. Pharm. Res. 1985, 1, 23–29.
REFERENCES 22. Corbo, M.; Liu, J.C.; Chien, Y.W. Transdermal controlled
delivery of propranolol from a multilaminate adhesive
1. Chien, Y.W. Novel Drug Delivery Systems: Fundamental, device. Pharm. Res. 1989, 6 (9), 753–758.
Developmental Concepts and Biomedical Assessments; 23. Microsealed Pharmaceutical Delivery device. US Patent
Marcel Dekker, Inc.: New York, 1982. 4,053,580, Oct 11, 1977.
2. Chien, Y.W. Industrial Pharmaceutical R&D Symposium 24. Chien, Y.W. Microsealed drug delivery systems: Fabrication
on Transdermal Controlled Release Medication. Piscataway, and performance. In Methods in Enzymology; Widder, K.J.,
New Jersey, Jan 14&15, 1982; Rutgers University, College of Green, R., Eds.; Academic Press: New York, 1985; 461–470.
Pharmacy, Proceedings Published in Drug Develop. & Ind. 25. Sanvordeker, D.R.; Cooney, J.G.; Wester, R.C. Transder-
Pharm. 1983; 9, 497–744. mal Nitroglycerin Pad. US Patent 4,336,243, June 22, 1982.
3. Chien, Y.W. Industrial Pharmaceutical R&D Symposium 26. Karim, A. Transdermal absorption: a unique opportunity
on Oral Controlled Drug Administrations. Piscataway, for constant delivery of nitroglycerin. Drug Develop. &
New Jersey, Jan 19&20, 1983; Rutgers University, College Ind. Pharm. 1983, 9, 671.
Drug Delivery: Controlled Release 1103

27. Theeuwes, F.; Yum, S.I. Principles of the design and oper- 44. Raghunathan, Y.; Amsel, L.; Hinsvark, O.; Bryant, W.
ation of generic osmotic pumps for the delivery of semisolid Sustained-release drug delivery system I: coated ion-
or liquid drug formulations. Ann. Biomed. Eng. 1976, 4 (4), exchange resin system for phenylpropanolamine and other
343–353. drugs. J. Pharm. Sci. 1981, 70 (4), 379–384.
28. Theeuwes, F. Elementary osmotic pump. J. Pharm. Sci. 45. Heller, J. Biodegradable polymers in controlled drug deliv-
1975, 64 (12), 1987–1991. ery. Crit. Rev. Ther. Drug Carrier Syst. 1984, 1 (1), 39–90.
29. De Leede, L.G.J. Rate-Controlled and Site-Specified Rectal 46. Morimoto, Y.; Fujimoto, S. Albumin microspheres as drug
Drug Delivery; Ph.D. Thesis; State University of Leiden: carriers. Crit. Rev. Ther. Drug Carrier Syst. 1985, 2 (1),
Leiden, The Netherlands, 1983. 19–63.
30. Theeuwes, F. Oros-osmotic system development. Drug 47. Sezaki, H.; Hashida, M. Macromolecule-drug conjugates in
Develop. & Ind. Pharm. 1983, 9, 1331–1357. targeted cancer chemotherapy. Crit. Rev. Ther. Drug
31. Liu, J.-C.; Farber, M.; Chien, Y.W. Comparative release of Carrier Syst. 1984, 1 (1), 1–38.
phenylpropranolamine HCL for long-acting appetite sup- 48. Heller, J.; Pengburn, S.H. A triggered bioerodible naltrex-
pressant products: acutrim vs. dexatrim. Drug Develop. & one delivery system. Proc. Int. Symp. Control. Rel. Bioact.
Ind. Pharm. 1984, 10, 1639–1661. Mater. 1986, 13, 35–36.
32. Michaels, A.S. Device for Delivering Drug to Biological 49. Heller, J.; Trescony, P.V. Controlled drug release by poly-
Environment. US Patent 4,180,073, Dec. 25, 1979. mer dissolution. II: Enzyme-mediated delivery device.
33. Blackshear, P.J.; Rohde, T.D.; Grotting, J.C.; Dorman, J. Pharm. Sci. 1979, 68 (7), 919–921.
F.D.; Perkins, P.R.; Varco, R.L.; Buchwald, H. Control 50. Horbett, T.A.; Ratner, B.D.; Kost, J.; Singh, M. A biore-
of blood glucose in experimental diabetes by means of a sponsive membrane for insulin delivery. In Recent
totally implantable insulin infusion device. Diabetes 1979, Advances in Drug Delivery Systems; Anderson, J.M.,
28 (7), 634–639. Kim, S.W., Eds.; Plenum Press: New York, 1984; 209–220.
34. Blackshear, P.J.; Rohde, T.D.; Varco, R.L.; Buchwald, H. 51. Kim, S.W.; Jeong, S.Y.; Sato, S.; McRea, J.C.; Feijan, J.
One year of continuous heparinization in the dog using a Self-regulating insulin delivery systema—a chemical
totally implantable infusion pump. Surg. Gynecol. Obstet. approach. In Recent Advances in Drug Delivery Systems;
1975, 141 (2), 176–186. Anderson, J.M., Kim, S.W., Eds.; Plenum Press: New York,
35. American pharmacy, implantable pump for morphine. 1983; 123.
NS24:20 1984. 52. Baker, R.W. Controlled release of biologically active
36. Hsieh, D.S.T.; Langer, R. Zero-order drug delivery systems agents; J. Wiley & Sons: New York, 1987.
with magnetic control. In Controlled Release Delivery Sys- 53. Jeong, S.Y.; Kim, S.W.; Eenink, M.J.D.; Feijen, J. Self-
tems; Roseman, T.J., Mansdorf, S.Z., Eds.; Marcel Dekker, regulating insulin delivery systems. I. Synthesis and charac-
Inc.: New York, 1983. terization of glycosylated insulin. J. Controlled Release
37. Kost, J. Ultrasound for controlled delivery of therapeutics. 1984, 1, 57–66.
Clin. Mater. 1993, 13 (1–4), 155–161. 54. Ringsdorf, H. Synthetic polymeric drugs. In Polymeric
38. Tyle, P.; Agrawala, P. Drug delivery by phonophoresis. Delivery Systems; Kostelnik, R.J., Ed.; Gordon and Brech:
Pharm. Res. 1989, 6 (5), 355–361. New York, 1978.
39. Bertolucci, L.E. Introduction of anti-inflammatory drugs 55. Chien, Y.W. Logics of transdermal controlled drug

Drug Delivery
Buccal–Mono
by iontophophoresis: double blind study. J. Orthopaed. & administration. Drug Develop. & Ind. Pharm. 1983, 9,
Sports Phys. Ther. 1982, 4, 103. 497–520.
40. Glass, J.M.; Stephen, R.L.; Jacobson, S.C. The quantity 56. Noonan, P.K.; Gonzalez, M.A.; Ruggirello, D.; Tomlinson,
and distribution of radiolabeled dexamethasone delivered J.; Babcock-Atkinson, E.; Ray, M.; Golub, A.; Cohen, A.
to tissue by iontophoresis. Int. J. Dermatol. 1980, 19 (9), Relative bioavailability of a new transdermal nitroglycerin
519–525. delivery system. J. Pharm. Sci. 1986, 75 (7), 688–691.
41. Harris, P.R. Iontophoresis: clinical research in musculoske- 57. Chien, Y.W. Microsealed drug delivery systems: theoretical
letal inflammatory conditions. J. Orthopaed. & Sports Phys. aspects and biomedical assessments. In Recent Advances in
Ther. 1982, 4, 109. Drug Delivery Systems; Anderson, J.M., Kim, S.W., Eds.;
42. Controlled Release Suspensions, APhA/APS Midwest Reg. Plenum Press: New York, 1984; 367–387.
Meet, Chicago, Illinois, April, 1984. 58. Chien, Y.W. Potential developments and new approaches in
43. Raghunathan, Y. Prolonged Release Pharmaceutical oral controlled release drug delivery systems. Drug
Preparations. US Patent 4,221,778, Sept 9, 1980. Develop. & Ind. Pharm. 1983, 9, 1291–1330.

Das könnte Ihnen auch gefallen