You are on page 1of 12

Advanced Drug Delivery Reviews 57 (2005) 2203 – 2214

www.elsevier.com/locate/addr

Targeted drug delivery with dendrimers: Comparison of the release


kinetics of covalently conjugated drug and non-covalent drug
inclusion complexB
Anil K. Patri, Jolanta F. Kukowska-Latallo, James R. Baker Jr. *
Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Health System, Ann Arbor, MI 48109-0648, USA
Received 3 March 2005; accepted 13 September 2005
Available online 14 November 2005

Abstract

Dendrimers have unique characteristics including monodispersity and modifiable surface functionality, along with highly
defined size and structure. This makes these polymers attractive candidates as carriers in drug delivery applications. Drug delivery
can be achieved by coupling a drug to polymer through one of two approaches. Hydrophobic drugs can be complexed within the
hydrophobic dendrimer interior to make them water-soluble or drugs can be covalently coupled onto the surface of the dendrimer.
Using both methods we compared the efficacy of generation 5 PAMAM dendrimers in the targeted drug delivery of methotrexate
coupled to the polymer. The amine-terminated dendrimers bind to negatively charged membranes of cells in a non-specific manner
and can cause toxicity in vitro and in vivo. To reduce toxicity and to increase aqueous solubility, modifications were made to the
surface hydroxyl groups of the dendrimers. For targeted drug delivery, the dendrimer was modified to have a neutral terminal
functionality for use with surface-conjugated folic acid as the targeting agent. The complexation of methotrexate within a
dendrimer changes the water insoluble drug into a stable and readily water-soluble compound. When this dendrimer complexed
drug, however, was placed in a solution of phosphate buffered saline, the methotrexate was immediately released and displayed
diffusion characteristics identical to free methotrexate. Covalently coupled methotrexate dendrimer conjugates were stable under
identical conditions in water and buffered saline. Cytotoxicity tests showed that methotrexate as the dendrimer inclusion complex
had an activity identical to the free drug in vitro. In contrast, folic acid targeted dendrimer with covalently conjugated methotrexate
specifically killed receptor-expressing cells by intracellular delivery of the drug through receptor-mediated endocytosis. This
study demonstrates that while drug as a dendrimer inclusion complex is readily released and active in vitro, covalently conjugated
drug to dendrimer is better suited for specifically targeted drug delivery.
D 2005 Elsevier B.V. All rights reserved.

Keywords: Dendrimers; Drug delivery; Inclusion complex; Methotrexate; Folic acid; Folate receptor

B
This review is part of the Advanced Drug Delivery Reviews theme issue on bDendrimers: a Versatile Targeting PlatformQ, Vol. 57/15, 2005.
* Corresponding author. Department of Internal Medicine Chief, Division of Allergy and Clinical Immunology, Nanotechnology Institute for
Medicine and Biological Sciences, 9220 MSRB III Ann Arbor, MI 48109-0648, USA. Tel.: +1 734 647 2777; fax: +1 734 936 2990.
E-mail address: jbakerjr@umich.edu (J.R. Baker).
URL: http://www.nano.med.umich.edu (J.R. Baker).

0169-409X/$ - see front matter D 2005 Elsevier B.V. All rights reserved.
doi:10.1016/j.addr.2005.09.014
2204 A.K. Patri et al. / Advanced Drug Delivery Reviews 57 (2005) 2203–2214

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2204
2. Experimental . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2205
2.1. Materials and methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2205
2.2. Cell culture and cytotoxicity measurement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2206
2.3. Synthesis of dendrimer conjugates . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2206
2.3.1. G5–FA–MTX conjugate . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2206
2.3.2. G5–MTX conjugate . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2206
2.3.3. Inclusion complex . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2207
2.4. Gel filtration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2207
3. Results and discussion. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2207
3.1. Methotrexate release studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2209
3.2. Targeted cytotoxicity of MTX–dendrimer inclusion complexes and conjugates . . . . . . . . . . . . . 2209
4. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2212
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2212
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2212

1. Introduction ity. This has led to investigations of novel carrier


systems [4] that overcome the problems, have longer
Targeted drug delivery directs a therapeutic agent circulation times and offer the potential to carry larger
in a site-specific manner to a particular cell type or amounts of drug. The aim of these applications is that
tissue. It could be beneficial in cancer therapy where the macromolecular carrier remains soluble and
cytotoxic drugs can kill tumor cells while sparing complexed to both the therapeutic and targeting
healthy tissue overcoming the toxic side effects agents until it reaches the targeting site. Any
associated with chemotherapy. In cancer therapy degradation of the carrier or premature release of free
several methods have been proposed to target tumors. drug before reaching the desired location defeats the
One employs the enhanced permeability and retention purpose of the carrier and reduces targeting efficacy,
(EPR) phenomenon in tumors [1] where microvascu- leading to increased side effects. For this reason, a
lature allows permeation of molecules and the absence stable system is required for any macromolecular
of lymphatic drainage leads to retention and accumu- carrier system for targeted drug delivery.
lation of the delivered compounds in a tumor. This has Dendritic polymers, or dendrimers, have been
the potential to concentrate drugs to some extent in proposed as one type of carrier for use in targeted
tumors when using polymeric drug carriers of a size drug delivery [5,6]. The unique characteristics of
that pervade tumor vasculature [2,3]. In EPR target- dendrimers such as uniform and controlled size,
ing, the therapeutic agent is subsequently released monodispersity, and modifiable surface group func-
locally and taken up by tumor cells to achieve its tionality make these molecules appealing for biomed-
effect. In contrast, tumor receptor or antigen targeting ical applications [5–7]. The ability to functionalize
specifically aims the therapeutic agent at tumor cells. their terminal groups with various targeting, thera-
Targeted tumor cells express or over-express unique peutic, and imaging agents in a specific and control-
receptors or antigens. Once identified, they can be lable manner leads to the potential to use them as
targeted using antibodies, small molecules or peptides carriers for targeted drug delivery. The utility of the
that can recognize and bind to the sites. In this internal void volume of dendrimers to encapsulate
approach, a therapeutic or a bioactive agent can be hydrophobic guest molecules and drugs has been
coupled directly to a targeting moiety for selective demonstrated by several research groups [8–11]. The
delivery. There are limitations, however, posed by resulting problems with aqueous solubility, biocom-
these conjugates, such as reduced specificity, de- patibility, and availability, however, have limited drug
creased solubility, and limited drug carrying capabil- delivery investigations to very few classes of den-
A.K. Patri et al. / Advanced Drug Delivery Reviews 57 (2005) 2203–2214 2205

drimers. The PAMAM dendrimers, which fulfill most tion 5 PAMAM dendrimer. The terminology
requirements for use in in vivo applications are being binclusion complexQ was used to depict dendrimer
considered extensively for medical applications containing a free drug bound by non-covalent
[12,13]. PAMAM dendrimers are being investigated interaction and bconjugateQ was used to represent
as carriers in gene transfection [14], MRI contrast a drug covalently conjugated to dendrimer. The
agents[15–17], boron–neutron capture therapy drug release characteristics from unmodified
[18,19], and drug delivery applications [7,20,21]. PAMAM dendrimer–inclusion complexes and cova-
The internal tertiary amines of PAMAM dendrimers lently conjugated drugs are evaluated in vitro for
are available for acid–base interactions and hydrogen the first time under the conditions replicating those
bonding as well as for other non-covalent interactions necessary for drug delivery.
with encapsulated guest molecules, thus making the
polymers effective agents for solubilizing hydropho-
bic drugs [22,23]. 2. Experimental
Research efforts on drug encapsulation in den-
drimers led to the synthesis of polyethylene glycol 2.1. Materials and methods
(PEG) functionalized dendrimers to impart aqueous
solubility, biocompatibility, and increased drug Folic acid, methotrexate, EDC, glycidol, hydrox-
loading with greater size of the dendrimer [24,25]. yl terminated PAMAM dendrimer, methanol and
Ibuprofen encapsulated, G3 and G4 amine function- dimethyl sulfoxide were purchased from Aldrich
alized PAMAM dendrimers were investigated for (St. Louis, MO). An Isco Low Pressure Liquid
their uptake into cells [26]. In all these efforts, Chromatography (LPLC) system with UA-6 UV-Vis
however, the encapsulated drug molecules were detector and type 11 (254 nm) or type 12 (214 nm)
released readily in isotonic solutions, rendering optical units in conjunction with a Sephadex G-25
them ineffective as carriers for targeted drug column was used for purification and detection of
delivery. On the other hand, efforts towards eluents by gel filtration. UV measurements were
covalent attachment of drugs to the surface of carried out on a Perkin Elmer Lambda 20
polyaryl ether dendrimers resulted in the conju- spectrometer and NMR measurements on a Bruker
gates’ becoming insoluble at physiological pH [27]. 500 MHz spectrometer. Generation 5 PAMAM
Addition of the PEG (polyethylene glycol) function dendrimers were synthesized as described below
to polyaryl ether dendrimers limited the solubility and characterized by 1H and 13C NMR, GPC, and
of drug–dendrimer conjugates at physiological con- MALDI-TOF Mass Spectrometry.
ditions because of the hydrophobic aromatic den- Unless specified, purification of all the dendrimer
dritic architecture coupled with hydrophobic drugs conjugates was carried out by dialysis in a 10 K
on the surface [28]. MWCO regenerated cellulose bag (Slide-A-Lyzer
We have investigated targeted drug delivery dialysis cassette; Pierce, Rockford, IL), with the initial
using a PAMAM dendrimer platform. Modification buffer exchanges performed with phosphate buffered
of the positively charged terminal amine function- saline (PBS pH 7.4) and then several times with
ality to neutral, acetylated or hydroxylated surfaces deionized water. The release of methotrexate from
decreased non-specific uptake of these dendrimers, either an inclusion complex or a covalent conjugate
improved aqueous solubility of various conjugates, was monitored by UV spectroscopy. The inclusion
and reduced aggregation [29,30]. Highly reproduc- complex or conjugate was introduced into a Micro
ible results for targeting with folic acid (FA), both DispoDialyzerk (8 kD MWCO; Spectrum Laborato-
in vitro and in vivo, encouraged further investiga- ries Inc., Rancho Dominguez, CA) and dialyzed in
tions into imaging and drug delivery [31]. In the PBS or water directly into a cuvette while stirring. The
current study, we synthesized highly soluble tar- UV spectra of permeates were taken at 15-min
geted dendrimer conjugates and compared the intervals, and the absorbance was compared from a
efficacy of covalently bound methotrexate (MTX) concentration curve of methotrexate to determine the
with an MTX inclusion complex utilizing genera- amount of released methotrexate.
2206 A.K. Patri et al. / Advanced Drug Delivery Reviews 57 (2005) 2203–2214

2.2. Cell culture and cytotoxicity measurement O


a N C FA
NH2 H
KB is a human cell line that over-expresses folate 12 8
NH2
receptor (FAR) when grown in folate-deficient medium G5-NH2 G5-FA-NH2
[32]. Cells were purchased from the American Type
Culture Collection (ATCC, Manassas, VA) and main- b c O
tained at 37 8C with 5% CO2 in folate-deficient RPMI C FA
NH
1640 supplemented with penicillin (100 units/mL), N OH
streptomycin (100 Ag/mL), and 10% heat-inactivated HO
2 N OH
FBS. For toxicity studies cells were plated in 96-well G5-OH HO
G5-FA 2
plates at 5000 cells/well in 100 AL of medium. After 24
h incubation of cells, conjugates were applied in 5 AL d
of medium at a final concentration range from 1 nM to
d O
C FA
50 AM of MTX. The concentration of compounds was NH
N OH
adjusted based on the number of MTX molecules HO
RN 2
present in the inclusion complex or conjugate. The N OH
RN HO
control dendrimers without MTX were used at the HO
2

molar concentrations of dendrimer present in the HO


O
inclusion complex and/or conjugate. Cells were pre- O
incubated with the conjugates for 1.5 h, then the MTX
O MTX
O
medium was removed and the cells were washed with
additional volumes of culture medium. After additional G5-MTX G5-FA-MTX
incubation for 72 h cell viability was determined using Scheme 1. Synthesis of dendrimer conjugates. a) Folic acid, EDC,
a colorimetric XTT based assay (Roche Diagnostics, DMSO; b) Glycidol, DMSO; c) Glycidol, DMSO/PBS; d)
Indianapolis, IN). The assay measures the conversion Methotrexate, EDC, DMSO. Conjugates 3 and 4 were used to
of XTT (sodium 3V-[1-(phenylamino-carbonyl)-3,4- make inclusion complexes. Conjugates 5 and 6 are covalent
tetrazolium]-bis(4-methoxy-6-nitro) benzene sulfonic conjugates.
acid hydrate] labeling reagent) into water soluble
formazan salt by mitochondrial dehydrogenase activity mixture was dialyzed and lyophilized to yield 280 mg
of viable cells [33]. The absorbance of the samples was of the folate–hydroxyl–terminated dendrimer G5–FA.
measured at 492 nm using a SPECTRA MAX 340 Methotrexate (7 mg; 15.3 Amol) was activated with
microtiter plate reader (Molecular Devices Corp., EDC (4 mg; 18.4 Amol) for 30 min in DMSO and added
Sunnyvale, CA). drop-wise under argon to a solution of G5–FA (66 mg;
1.5 Amol) in 5 mL of anhydrous DMSO and stirred for
2.3. Synthesis of dendrimer conjugates 8 h at room temperature. The resultant mixture was
purified by dialysis and lyophilized, yielding 60 mg of
2.3.1. G5–FA–MTX conjugate purified conjugate G5–FA–MTX.
Generation 5 amine terminated PAMAM dendrimer
G5–NH2 (Scheme 1) was used for the current studies. 2.3.2. G5–MTX conjugate
The synthesis of folic acid conjugated dendrimer G5– To a solution of generation 5 PAMAM dendrimer
FA–NH2 was carried out using a previously published G5–NH2 (250 mg; 10 Amol) in anhydrous DMSO (5
procedure [29]. The unreacted folic acid and urea mL) excess glycidol (300 mg; 4 mmol) was added and
byproducts were removed by ultrafiltration using a stirred at room temperature under nitrogen. The
Pellicon device. To a solution of G5–FA–NH2 (250 mg; reaction mixture was then dialyzed. The purified
8.9 Amol) in a 1 : 1 mixture of DMSO / pH 8.5 PBS (8 conjugate was lyophilized and yielded 260 mg of
mL), excess glycidol (158 mg; 2.1 mmol) was added G5–OH conjugate. Methotrexate (8.1 mg; 17.82 Amol)
and stirred for 3 h. An additional aliquot (158 mg; 2.1 was activated with EDC (4.1 mg; 21 Amol) in
mmol) of glycidol was added and after 3 h the resultant anhydrous DMSO (0.5 mL) for 30 min, added drop-
A.K. Patri et al. / Advanced Drug Delivery Reviews 57 (2005) 2203–2214 2207

wise to a solution of G5–OH (50 mg; 1.2 Amol) in characterized using NMR and UV spectroscopy and
DMSO (3 mL) and stirred for 8 h under nitrogen. The was determined to have an average of 5 folic acid
mixture was dialyzed and lyophilized, yielding 53 mg moieties per dendrimer. The surface of the dendrimers
of G5–MTX conjugate. was modified to achieve neutral (acetylated or
hydroxylated substitution) or negatively charged
2.3.3. Inclusion complex (carboxylic acid substitution) terminal group function-
Dendrimer inclusion complexes were prepared by alities because the amine-terminated dendrimers bind
adding 0.6 Amol of methotrexate to 66 nmol of non-specifically to cells. The binding characteristics
dendrimer solution in deionized water (0.5 mL), of all of the modified dendrimers, with or without
vortexing followed by stirring for 2 h and then conjugated folic acid, were compared using a folic
lyophilizing the resultant inclusion complex. The acid receptor (FAR) expressing cell line [30]. The
complex was then re-dissolved in 0.2 mL of deionized acetylated and hydroxylated dendrimers were both
water and an aliquot of 50 AL was dialyzed (8 kD found to have low non-specific binding and high
MWCO) at room temperature as described earlier and aqueous solubility. Although the conjugation of
the cumulative release of methotrexate was moni- hydrophobic groups to dendrimers decreases their
tored. Alternately, the release studies were carried out aqueous solubility, the hydroxyl-functionalized den-
in a dialysis cassette (10 K MWCO) as described drimers impart aqueous solubility even after partial
above. This method, requiring a larger amount of conjugation with hydrophobic groups. The hydroxyl
material, had a faster release rate. groups on the surface facilitate ester conjugation of
the carboxylic acid containing drugs as well. These
2.4. Gel filtration ester conjugates are acid labile and are stable in
neutral buffers. We have chosen to test the efficacy as
Gel filtration of the dendrimer inclusion complexes a difference in cytotoxicity measured in vitro of the
and covalently coupled conjugates were conducted on drug delivered in the form of ester-conjugated MTX
a G-25 sephacryl column pre-equilibrated and eluted or as an inclusion complex containing MTX.
with phosphate buffered saline. Samples were applied The folate–dendrimer conjugate G5–FA–NH2 was
to the column, and eluting fractions were detected at reacted with excess glycidol to functionalize the
210 nm and isolated using a fraction collector. terminal primary amines to a hydroxyl-terminated
G5–FA dendrimer. This conjugate is highly soluble in
aqueous buffers. The MTX was then conjugated to G5–
3. Results and discussion FA by EDC coupling in anhydrous DMSO to form an
ester-linked MTX–dendrimer–FA conjugate (G5–FA–
The use of folic acid-conjugated PAMAM den- MTX). This conjugate was purified by dialysis. The 1H
drimer for targeting and imaging applications was NMR in D2O had broad overlapping signals in the
reported previously by several groups including ours aliphatic region between d 4 to 2 ppm corresponding to
[15,30,34]. The well-documented conjugation of folic the dendrimer C H 2 protons and aromatic signals at d
acid to dendrimer was chosen to test different delivery 6.7, 7.6, and 8.5 ppm. The UV spectrum had over-
approaches through targeting. When limited numbers lapping absorptions due to the presence of both FA and
of folic acid molecules (5 per dendrimer) are MTX (Fig. 2b) covalently bound to dendrimer, which
conjugated to G5 PAMAM dendrimer, the conjugate are different from conjugates containing only FA or
retains solubility in water [30]. The use of phosphate MTX. As a control without FA, generation 5 PAMAM
buffered saline solution (PBS) during the initial phase dendrimer was surface-functionalized with glycidol to
of ultrafiltration removes complexed small molecules give the hydroxyl-terminated dendrimer G5–OH. The
from the inclusion complex, but does not disrupt the MTX was conjugated to this dendrimer using EDC
covalent drug linkage. Subsequent washings with coupling as described earlier and purified by dialysis.
deionized water remove the buffer salts. This proce- The 1H NMR of this G5–MTX conjugate showed
dure of purification assures a high purity of covalent overlapping broad signals in the aliphatic region
dendrimer conjugate. The G5–FA–NH2 conjugate was between d 4–2 ppm for the dendrimers methylene
2208 A.K. Patri et al. / Advanced Drug Delivery Reviews 57 (2005) 2203–2214

protons and three signals in the aromatic region (Fig. 1b). As expected, the first fraction with
between d 6.6 to 8.5 ppm. The UV spectrum of this absorbance was analyzed to be the dendrimer–FA
conjugate has a characteristic absorption similar to that conjugate (G5–FA) with characteristic absorption for
of free MTX in addition to an intense absorption at 210 FA at ODmax 286, while the second fraction appeared
nm for the dendrimer, confirming the conjugation. to be the free MTX. This indicates that charge-based
Dendrimer conjugates were tested for purity by gel drug inclusion complexes can be disrupted by
filtration using LPLC. Initially the MTX inclusion phosphate-buffered saline solution and that the den-
complex with G5–FA conjugate was filtered through a drimer–drug conjugates can be purified by gel-
Sephadex G-25 column using LPLC. Two peaks were filtration. The dendrimer conjugate with covalently
detected (Fig. 1a) and analyzed by UV spectroscopy coupled FA and MTX (G5–FA–MTX) was stable

b
1

0.9

0.8

0.7
Fraction 1
Absorbance

0.6 Fraction 2
0.5

0.4

0.3

0.2

0.1

0
200 250 300 350 400 450 500
Wavelength (nm)

Fig. 1. (a) LPLC chromatogram of methotrexate inclusion complex with G5–FA upon gel filtration. First isolated fraction was G5–FA conjugate
and the second, later eluted fraction was free methotrexate (Fig. 1b). (b) UV spectra of two fractions isolated upon gel filtration of a dendrimer–
methotrexate inclusion complex. The UV spectrum of fraction 1 is identical to the G5–FA conjugate and fraction 2 is identical to that of free
methotrexate.
A.K. Patri et al. / Advanced Drug Delivery Reviews 57 (2005) 2203–2214 2209

under the gel-filtration conditions and did not yield dialyzed in PBS (Fig. 3). More than 70% of the
any free methotrexate. The only fraction detected (Fig. included MTX was detected in PBS within 2.5 h. The
2a) was analyzed by UV spectroscopy (Fig. 2b) and release rate of free MTX from the dialyzer in PBS did
contained the dendrimer with both FA and MTX. The not differ from MTX released from the dendrimer
non-targeted dendrimer–MTX conjugate (G5–MTX) inclusion complex under the same conditions. The
without folate was also found to be stable and did not release rate of MTX from the inclusion complex with
contain detectable free MTX (data not shown). G5–FA showed a similar pattern, with 50% of the
drug being released from the dialyzer in PBS in
3.1. Methotrexate release studies 2.5 h whereas there was no release of MTX from this
compound when dialyzed in water. This trend of
The dendrimer–drug inclusion complex is soluble MTX release from the inclusion complexes with
and stable in water with minimal release of free MTX dendrimers in PBS is consistent and reproducible
(less than 5%) when dialyzed against water. The with either hydroxyl, acetylated, or amine surface
hydroxyl-terminated G5 dendrimer G5–OH, however, dendrimers, although the rate of release is slightly
releases MTX from its inclusion complex when different, depending on the method and the surface
composition. This indicates that the interactions
between the dendrimer and MTX are not strong
a enough to retain MTX in an inclusion complex when
the inter-molecular forces are neutralized in a
buffered salt solution. In contrast MTX covalently
conjugated to dendrimer is neither released in water
nor in phosphate-buffered saline.

3.2. Targeted cytotoxicity of MTX–dendrimer inclu-


sion complexes and conjugates

Targeting through the high-affinity receptor (FR)


for the vitamin folic acid (FA), also known as the
folate binding protein, has been achieved by delivery
of carriers with conjugated folic acid [35]. The FR is
over-expressed in many cancers and is internalized
b into cells after ligand-binding [36,37]. We have
1.2
previously demonstrated the highly selective binding
1 and internalization in vitro of PAMAM dendrimers
Fraction 1
covalently conjugated with FA in KB tumor cells that
Absorbance

0.8
over-express FR [30]. The KB tumor human cell line
0.6 over-expresses the FR when maintained in a folate-
deficient medium [38]. This FR expression can be
0.4
down regulated by culturing KB cells in a thousand-
0.2 fold excess of FA or can be transiently blocked by the
addition of free FA into the medium [30]. The
0 antineoplastic drug methotrexate (MTX) is a folic
200 300 400 500
Wavelength (nm) acid antagonist with several-hundred-fold lower af-
finity for FR than folic acid [39–41]. The cytotoxicity
Fig. 2. (a) LPLC chromatogram of a G5–FA–MTX conjugate. A of inclusion complexes obtained with KB cells over-
single fraction was isolated and no detectable free methotrexate was
observed. (b) UV spectrum of the single fraction isolated upon gel
expressing FR (FAR+) is similar to the cytotoxicity of
filtration of a G5–FA–MTX conjugate. The spectrum shows free MTX in a concentration range of drug from 0.05
absorption for both folic acid and methotrexate. to 0.1 AM (Fig. 4). The MTX is released from
2210 A.K. Patri et al. / Advanced Drug Delivery Reviews 57 (2005) 2203–2214

Release Profile of MTX from various Dendrimer Conjugates and Inclusion


Complexes in both PBS and Water
100
MTX in PBS
90

80 MTX inclusion with


G5-OH in PBS
70
MTX inclusion with
Percent Released

60 G5-FA in PBS
G5-FA-MTX conjugate
50
in PBS
40 G5-MTX conjugate in
PBS
30
MTX inclusion with
20 G5-OH in water
10 MTX inclusion with
G5-FA in water
0
0 0.5 1 1.5 2 2.5 3
Time (hrs)

Fig. 3. Comparative release profile of methotrexate (MTX) from inclusion complexes and conjugates in water and phosphate-buffered saline
(PBS). Methotrexate in inclusion complexes in PBS was released to the same degree as free MTX. The MTX inclusion complexes in water and
covalent conjugates in PBS did not release free methotrexate.

KB (FAR+) 1.5h preincubation


120
MTX

100 G5-OH control


Cell Viability (% control)

G5-FA control
80
G5-MTX
conjugate
60
G5-FA-MTX
conjugate
40 MTX Inclusion
with G5-OH
MTX Inclusion
20 with G5-FA
Control
0
0.001 0.01 0.1 1 10 100
Concentration (µM)

Fig. 4. Cytotoxicity of covalent conjugates with MTX and dendrimer inclusion complexes with MTX tested on KB cells over-expressing FA
receptor (FAR+). Cells were pre-incubated for 1.5 h. with methotrexate containing dendrimers, and dendrimers without drug, washed with medium
and further incubated for 72 h. While the dendrimer control conjugates G5–OH and G5–FA without methotrexate are not cytotoxic, the
methotrexate inclusion complexes with G5–OH and G5–FA were as equally toxic as free methotrexate in a concentration range of MTX from 0.05
to 0.1 AM. The G5–MTX conjugate without folic acid is cytotoxic at higher (about 1.0 AM) concentration of MTX, compared to the targeted G5–
FA–MTX conjugate at 0.3 AM of MTX.
A.K. Patri et al. / Advanced Drug Delivery Reviews 57 (2005) 2203–2214 2211

KB (FAR+) 1.5h preincubation blocked with free FA


120
MTX

100 G5-OH
control

G5-FA control
Cell viability (% control)

80
G5-MTX
conjugate
60
G5-FA-MTX
conjugate

40 MTX inclusion
with G5-OH
MTX inclusion
20 with G5-FA

Control

0
0.001 0.01 0.1 1 10 100
Concentration (µM)

Fig. 5. Specificity of delivering FA-targeted MTX conjugates and inclusion complexes. Cytotoxicity was tested using KB (FAR+) cells after
initial blocking of cell culture with free folic acid (9 AM) followed by the pre-incubation with compounds for 1.5 h, wash and further incubation
for 72 h. The free methotrexate and inclusion complexes were toxic only at a higher (10 fold) concentrations of MTX (1 AM) compared to non-
blocked KB (FAR+) cells (Fig. 4). While the G5–FA–MTX conjugate showed toxicity at 10.0 AM of MTX, the control non-targeted G5–MTX
conjugate was not toxic at the same concentration of MTX.

KB (FAR-) 1.5 h preincubation


120
MTX

100 G5-OH control


Cell Viability (% control)

80 G5-FA control

G5-MTX
60
conjugate
G5-FA-MTX
40 conjugate
MTX inclusion
20 with G5-OH
Control

0
0.001 0.01 0.1 1 10 100
Concentration (µM)

Fig. 6. Specificity of delivering MTX via FA receptor using KB cells with low expression of the FA receptor (FAR ). Compared with Fig. 5, a
10-fold higher concentration of drug was required to achieve similar cytotoxicity. Compared to the FAR (+) cells, methotrexate was toxic at
higher concentrations (N1 AM) and similar to the MTX in the inclusion complex. The G5–FA–MTX and G5–MTX conjugates had toxicity at
N10 AM concentration of MTX.
2212 A.K. Patri et al. / Advanced Drug Delivery Reviews 57 (2005) 2203–2214

inclusion complexes within 1.5 h of pre-incubation inclusion complex improves its solubility in water,
with cells and is as effective as free MTX. The effective a cleavable, covalently linked dendrimer conjugate
cytotoxic concentration of the conjugated MTX was is better suited for targeted drug delivery as it does
from 0.3 to 1.0 AM (Fig. 4). Similar patterns of not release the drug prematurely in biological
cytotoxicity but at 10- to 20-fold higher concentrations conditions. Although in the in vitro cell culture
of MTX were observed for inclusion complexes (1.0 assays used here, the cytotoxicity of the inclusion
AM) and the conjugates (10.0 AM) when the initial 1.5 complex is identical to the free methotrexate on
h uptake was blocked with an excess of free folic acid, FAR (+), FAR (+) blocked with free folic acid, and
indicating receptor-mediated uptake (Fig. 5). The FAR ( ) cells, the viability of the cells depends on
specific uptake of covalently conjugated methotrexate the presence of the receptor. The specificity of the
through receptor-mediated internalization has to be internalization of the covalent conjugate is clearly
followed by the hydrolysis of free drug to effect discernable on KB cells over-expressing FR and is
cytotoxicity. The ester-linked MTX, FA-targeted con- further supported by the blocking of toxicity with
jugate G5–FA–MTX is required at about 10-fold higher the free folic acid or by the lack of cytotoxic effect
concentration to have the same effect as the inclusion in FAR ( ) cells.
complex as the result of the slower release of the
conjugated MTX after receptor-mediated internaliza-
tion of the conjugate (Figs. 4 and 5). In contrast, much Acknowledgements
smaller inhibition from folic acid was observed with
the drug inclusion complex and free MTX (Fig. 5). We thank Sonya Krasilshchikova, David Leavitt,
These observations were further confirmed using KB Adam Was and Andrea East for their technical
cells with down-regulated FR (FAR ). The toxicity assistance. This project has been funded in whole
pattern remained similar in inclusion complexes and or in part with Federal Funds from the U.S. Army
conjugates and at the very high concentrations of MTX Medical Research and Material Command, Depart-
(5.0–10.0 AM) the differences could not be clearly ment of Defense, under grant DAMD17-02-1-0096
discerned (Fig. 6). and National Cancer Institute, National Institutes of
Health, under Contract #NOI-CO-97111.

4. Conclusion
References
We have synthesized generation 5 PAMAM [1] H. Maeda, J. Wu, T. Sawa, Y. Matsumura, K. Hori, Tumor
dendrimer conjugates with folic acid and modified vascular permeability and the EPR effect in macromolecular
surface to give respectively a hydrophilic, neutral, therapeutics: a review, J. Control. Release 65 (2000) 271 – 284.
hydroxyl surface. Methotrexate inclusion complexes [2] H.R. Ihre, O.L.P. De Jesus, F.C. Szoka, J.M.J. Frechet,
were formed in water and their cytotoxicity was Polyester dendritic systems for drug delivery applications:
design, synthesis, and characterization, Bioconjug. Chem. 13
compared with MTX covalently conjugated by an (2002) 443 – 452.
ester linkage that mediates release of the active [3] O.L. Padilla De Jesus, H.R. Ihre, L. Gagne, J.M. Frechet, F.C.
drug through hydrolysis. Even though free metho- Szoka Jr., Polyester dendritic systems for drug delivery
trexate is retained within the drug complex when applications: in vitro and in vivo evaluation, Bioconjug.
Chem. 13 (2002) 453 – 461.
dialyzed against deionized water, it is readily
[4] R. Langer, New methods of drug delivery, Science 249 (1990)
released in phosphate-buffered saline (PBS) and, 1527 – 1533.
in the cell culture, is available in a manner similar [5] G.R. Newkome, C.N. Moorefield, F. Vogtle, Dendrimers and
to the free drug. The activity of MTX is Dendrons: Concepts, Syntheses, Applications, Wiley-VCH,
proportional to the number of MTX molecules in Weinheim, Germany, 2001.
the inclusion complex. All the covalently coupled [6] J.M.J. Fréchet, D.A. Tomalia (Eds.), Dendrimers and other
Dendritic Polymers, Wiley, West Sussex, UK, 2001.
drug–dendrimer conjugates were stable in both [7] A.K. Patri, I. Majoros, J.R.J. Baker, Dendritic polymer
water and PBS buffer solution. This suggests that, macromolecular carriers for drug delivery, Curr. Opin. Chem.
while complexing a drug with dendrimer as an Biol. 6 (2002) 466 – 471.
A.K. Patri et al. / Advanced Drug Delivery Reviews 57 (2005) 2203–2214 2213

[8] A.M. Naylor, W.A. Goddard III, G.E. Kiefer, D.A. Tomalia, water soluble PAMAM derivatives, Tetrahedron 59
Starburst dendrimers: 5. Molecular shape control, J. Am. (2003) 3873 – 3880.
Chem. Soc. 111 (1989) 2339 – 2341. [23] L.J. Twyman, A.E. Beezer, R. Esfand, M.J. Hardy, J.C.
[9] G.R. Newkome, C.N. Moorefield, G.R. Baker, M.J. Saunders, Mitchell, The synthesis of water soluble dendrimers, and their
S.H. Grossman, Unimolecular micelles, Angew. Chem., Int. application as possible drug delivery systems, Tetrahedron
Ed. Engl. 30 (1991) 1178 – 1180. Lett. 40 (1999) 1743 – 1746.
[10] J.F.G.A. Jansen, E.M.M. de Brabander-van den Berg, E.W. [24] M. Liu, K. Kono, J.M. Frechet, Water-soluble dendritic
Meijer, Encapsulation of guest molecules into a dendritic box, unimolecular micelles: their potential as drug delivery agents,
Science 266 (1994) 1226 – 1229. J. Control. Release 65 (2000) 121 – 131.
[11] J.F.G.A. Jansen, E.W. Meijer, E.M.M. de Brabander-van den [25] C. Kojima, K. Kono, K. Maruyama, T. Takagishi, Synthesis of
Berg, The dendritic box: shape-selective liberation of encap- polyamidoamine dendrimers having poly(ethylene glycol)
sulated guests, J. Am. Chem. Soc. 117 (1995) 4417 – 4418. grafts and their ability to encapsulate anticancer drugs,
[12] D.A. Tomalia, H. Baker, J.R. Dewald, M. Hall, G. Kallos, S. Bioconjug. Chem. 11 (2000) 910 – 917.
Martin, J. Roeck, J. Ryder, P. Smith, A new class of polymers: [26] P. Kolhe, E. Misra, R.M. Kannan, S. Kannan, M. Lieh-Lai,
StarburstR—dendritic macromolecules, Polym. J. 17 (1985) Drug complexation, in vitro release and cellular entry of
117 – 132. dendrimers and hyperbranched polymers, Int. J. Pharm. 259
[13] D.A. Tomalia, A.M. Naylor, W.A. Goddard III, Starburst (2003) 143 – 160.
dendrimers: molecular-level control of size, shape, surface [27] K. Kono, M. Liu, J.M. Frechet, Design of dendritic macro-
chemistry, topology and flexibility in the conversion of atoms molecules containing folate or methotrexate residues, Bio-
to macroscopic materials, Angew. Chem., Int. Ed. Engl. 29 conjug. Chem. 10 (1999) 1115 – 1121.
(1990) 138 – 175. [28] M. Liu, K. Kono, J.M. Frechet, Water soluble dendrimer-
[14] J.F. Kukowska-Latallo, A.U. Bielinska, J. Johnson, R. poly(etylene glycol) starlike conjugates as potential drug
Spindler, D.A. Tomalia, J.R. Baker Jr., Efficient transfer of carriers, J. Polym. Sci. Polym. Chem. 37 (1999) 3492 – 3503.
genetic material into mammalian cells by using Starburst [29] J.R. Baker Jr., A. Quintana, L.T. Piehler, M. Banaszak Holl,
polyamidoamine dendrimers, Proc. Natl. Acad. Sci. 93 (1996) D. Tomalia, E. Raczka, The synthesis and testing of anti-
4897 – 4902. cancer therapeutic nanodevices, Biomed. Microdevices 3
[15] S.D. Konda, M. Aref, S. Wang, M. Brechbiel, E.C. Wiener, (2001) 61 – 69.
Specific targeting of folate-dendrimer MRI contrast agents to [30] A. Quintana, E. Raczka, L. Piehler, I. Lee, A. Myc, I. Majoros,
the high affinity folate receptor expressed in ovarian tumor A.K. Patri, T. Thomas, J. Mule, J.R. Baker Jr., Design and
xenografts, Magma 12 (2001) 104 – 113. function of a dendrimer-based therapeutic nanodevice targeted
[16] E.C. Wiener, M.W. Brechbiel, H. Brothers, R.L. Magin, to tumor cells through the folate receptor, Pharm. Res. 19
O.A. Gansow, D.A. Tomalia, P.C. Lauterbur, Dendrimer- (2002) 1310 – 1316.
based metal chelates: a new class of magnetic [31] S. Wang, R.J. Lee, G. Cauchon, D.G. Gorenstein, P.S. Low,
resonance imaging contrast agents, Magn. Reson. Med. Delivery of antisense oligodeoxyribonucleotides against the
31 (1994) 1 – 8. human epidermal growth-factor receptor into cultured KB cells
[17] H. Kobayashi, S. Kawamoto, S.K. Jo, H.L. Bryant Jr., M.W. with liposomes conjugated to folate via polyethylene–glycol,
Brechbiel, R.A. Star, Macromolecular MRI contrast agents Proc. Natl. Acad. Sci. U. S. A. 92 (1995) 3318 – 3322.
with small dendrimers: pharmacokinetic differences between [32] E.C. Wiener, S. Konda, A. Shadron, M. Brechbiel, O. Gansow,
sizes and cores, Bioconjug. Chem. 14 (2003) 388 – 394. Targeting dendrimer–chelates to tumors and tumor cells
[18] R.F. Barth, D.M. Adams, A.H. Soloway, F. Alam, M.V. Darby, expressing the high-affinity folate receptor, Invest. Radiol.
Boronated StarburstR dendrimer-monoclonal antibody immu- 32 (1997) 748 – 754.
noconjugates: evaluation as a potential delivery system for [33] N.W. Roehm, G.H. Rodgers, S.M. Hatfield, A.L. Glasebrook,
neutron capture therapy, Bioconjug. Chem. 5 (1994) 58 – 66. An improved colorimetric assay for cell-proliferation and
[19] G. Wu, R.F. Barth, W. Yang, M. Chatterjee, W. Tjarks, M.J. viability utilizing the tetrazolium salt Xtt, J. Immunol.
Ciesielski, R.A. Fenstermaker, Site-specific conjugation of Methods 142 (1991) 257 – 265.
boron-containing dendrimers to anti-EGF receptor monoclonal [34] S.D. Konda, S. Wang, M. Brechbiel, E.C. Wiener, Biodis-
antibody cetuximab (IMC-C225) and its evaluation as a tribution of a 153 Gd-folate dendrimer, generation = 4, in mice
potential delivery agent for neutron capture therapy, Biocon- with folate-receptor positive and negative ovarian tumor
jug. Chem. 15 (2004) 185 – 194. xenografts, Invest. Radiol. 37 (2002) 199 – 204.
[20] R. Esfand, D.A. Tomalia, Poly(amidoamine) (PAMAM) [35] C.P. Leamon, J.A. Reddy, Folate-targeted chemotherapy, Adv.
dendrimers: from biomimicry to drug delivery and biomedical Drug Deliv. Rev. 56 (2004) 1127 – 1141.
applications, Drug Discov. Today 6 (2001) 427 – 436. [36] S.D. Weitman, R.H. Lark, L.R. Coney, D.W. Fort, V. Frasca,
[21] M. Liu, J.M. Frechet, Designing dendrimers for drug delivery, V.R. Zurawski Jr., Distribution of the folate receptor GP38 in
Pharm. Sci. Technol. Today 2 (1999) 393 – 401. normal and malignant cell lines and tissues, Cancer Res. 52
[22] A.E. Beezer, A.S.H. King, I.K. Martin, J.C. Mitchel, (1992) 3396 – 3401.
L.J. Twyman, C.F. Wain, Dendrimers as potential drug [37] J.F. Ross, P.K. Chaudhuri, M. Ratnam, Differential regulation
carriers; encapsulation of acidic hydrophobes within of folate receptor isoforms in normal and malignant tissues in
2214 A.K. Patri et al. / Advanced Drug Delivery Reviews 57 (2005) 2203–2214

vivo and in established cell lines. Physiologic and clinical [40] A.C. Antony, M.A. Kane, R.M. Portillo, P.C. Elwood, J.F.
implications, Cancer 73 (1994) 2432 – 2443. Kolhouse, Studies of the role of a particulate folate-
[38] J.J. Turek, C.P. Leamon, P.S. Low, Endocytosis of folate– binding protein in the uptake of 5-methyltetrahydrofolate
protein conjugates: ultrastructural localization in KB cells, J. by cultured human KB cells, J. Biol. Chem. 260 (1985)
Cell Sci. 106 (Pt 1) (1993) 423 – 430. 14911 – 14917.
[39] J.C. Deutsch, P.C. Elwood, R.M. Portillo, M.G. Macey, J.F. [41] T.P. Thomas, I.J. Majoros, A. Kotlyar, J.F. Kukowska-Latallo,
Kolhouse, Role of the membrane-associated folate binding A.U. Bielinska, A. Myc, J.R. Baker Jr., Targeting and
protein (folate receptor) in methotrexate transport by human inhibition growth by an engineered dendritic nanodevice, J.
KB cells, Arch. Biochem. Biophys. 274 (1989) 327 – 337. Med. Chem. 48 (2005) 3729 – 3735.

You might also like