You are on page 1of 14

View Article Online / Journal Homepage / Table of Contents for this issue

NJC Dynamic Article Links

Cite this: New J. Chem., 2012, 36, 227–240

www.rsc.org/njc PERSPECTIVE
Dendrimer therapeutics: covalent and ionic attachmentsw
Saı̈d El Kazzouli,*a Serge Mignani,a Mosto Bousminaab and
Jean-Pierre Majoral*ac
Published on 08 September 2011. Downloaded by MIT Library on 2/10/2021 12:35:19 AM.

Received (in Montpellier, France) 31st May 2011, Accepted 18th August 2011
DOI: 10.1039/c1nj20459a

This concise review focuses on dendrimers functionalized by different linkers required for covalent
and ionic attachments of drugs to the dendrimer’s surface and highlights the importance of the
chemical nature of linkers for controlled release of free drugs. The stability of linkers under
physiological conditions and their lability under acidic conditions such as those of endosomes and
lysosomes or under enzymatic conditions will be discussed. Especially, we review functionality
of the most recently reported drug–dendrimer conjugates. Then, we give a short comparison of
dendrimer conjugates versus either dendrimer complexes or polymer therapeutics and finally we
briefly summarize the importance of both targeted and nontargeted covalently conjugated drugs.

a
Institute of Nanomaterials & Nanotechnology (INANOTECH), 1. Introduction
Moroccan Advanced Science, Innovation and Research (MAScIR)
Foundation, Avenue de l’Armée Royale, Madinat El Irfane, 10100, During the last decade, pertinent reviews on dendrimers
Rabat, Morocco. E-mail: s.elkazzouli@mascir.com;
Fax: +212 (0)537570880; Tel: +212 (0)537576194 synthesis and application in various areas have been reported,
b
Hassan II academy of Science and Technology, Rabat, Morocco mainly in the field of nanomedicine.1–31 The most attractive
c
Laboratoire de Chimie de Coordination du CNRS, and promising therapeutic applications of dendrimers are in
205 Route de Narbonne, 31077 Toulouse Cedex 4, France. gene delivery and imaging techniques, and as vaccine,
E-mail: majoral@lcc-toulouse.fr
w This article is part of the themed issue Dendrimers II, guest-edited antiviral, antibacterial, anticancer and anti-inflammatory
by Jean-Pierre Majoral. agents. For a recent and very impressive example, in vivo

Saı¨d El Kazzouli was born in Serge Mignani was born in


Beni Mellal (Morocco) in France. He joined the Catholic
1975. He obtained his MSc University of Louvain-la-
at the University of Orleans Neuve (Belgium) in 1978 for
(France) then in 2004 he a PhD training under the
received his PhD in organic supervision of Prof H. G. Viehe.
chemistry under the super- He obtained his PhD in 1981.
vision of Prof. G. Guillaumet In 1985–86, he joined the
and Prof. A. Mouaddib from University of Madison (USA)
the University of Orleans. In as a postdoctoral under the
2004 he worked at the same supervision of Prof B. M.
University as a postdoctoral Trost. In 1981, he joined
with Prof. L. Agrofoglio Rhône-Poulenc (currently
(nucleoside chemistry) then, Sanofi-Aventis) at the Vitry
Saı̈d El Kazzouli in 2005 with Prof. S. Berteina- Serge Mignani Research Center, where he
Raboin in collaboration with was a Head of the Medicinal
Syngenta firm (Switzerland). In 2006 he joined NCI, NIH in Chemistry Department and Scientific Director. He is the author
USA to work as a postdoctoral with Dr V. E. Marquez for of more than 90 publications and more than 100 patents. He is a
3 years on the design and synthesis of anticancer agents. In 2009 member of the ‘‘Comite´ National de la Chimie’’ (December
he became a scientist researcher at INANOTECH, Foundation 2009), of the ‘‘Board of Consulting Editors’’ of Bioorganic &
MAScIR in Rabat, Morocco, where he is working in dendrimer’s Medicinal Chemistry Letters and Bioorganic & Medicinal
group with Prof. Jean Pierre Majoral. His main research Chemistry (2003–2008), Vice-President of the ‘‘French-
interests are the design and synthesis of bioactive molecules, Japanese Society for Fine and Medicinal Chemistry’’, and
drug delivery as well as catalysis. recently he was nominated National Representative from France
to the IUPAC Division of Chemistry and Human Health.

This journal is c The Royal Society of Chemistry and the Centre National de la Recherche Scientifique 2012 New J. Chem., 2012, 36, 227–240 227
View Article Online

studies in mice showed that azabisphosphonate dendrimers controlled release. Thus, reaching a proper stability/lability
prevent complete ankle joint inflammation effects.32 balance for the linker between the drug and the dendrimer is
In the nanomedicine domain, dendrimers are gaining more critical. This can be achieved by adequate selection of both the
and more interest in the area of drug carriers and drug dendrimer end-groups and the linker chemical structure for
targeting, because they can combine the required character- each kind of targeted tissue.34 This concise review does not
istics of the efficient vectors for specific therapeutic targeting: intend to be exhaustive but aims to report and to discuss the
(i) surface functionalization with ligands for specific recogni- most recent developments in the field of the dendrimer-based
tion of receptors or antigens at the desired site, (ii) anchoring drug delivery by selecting pertinent and representative
of hydrophilic terminal surface groups to avoid the opsoniza- examples. The different kinds of linkers used so far (Fig. 1)
tion or the macrophages recognition and elimination by the as well as the chemical nature of the attachment of drugs to
Published on 08 September 2011. Downloaded by MIT Library on 2/10/2021 12:35:19 AM.

RES (reticular endothelial system),33 (iii) size tuning at the dendrimers will be discussed. In addition, we will comment on
nanometric scale (smaller than 100 nm) for prolonged the drug release process and biological activities of drug
circulating time, (iv) incorporation of probes for in vivo loaded-dendrimers versus the plain drug whenever they are
imaging and monitoring. Dendrimers are therefore powerful studied. We will also review and compare dendrimer
and efficient nanocarriers and their biodistribution and conjugates versus either dendrimer complexes or polymer
pharmacokinetic parameters can be controlled with respect conjugates. This review is therefore a literature updated
to the number and the nature of termini groups. supplement of the recent reviews reported by Haag et al.,6
Several dendrimer-based nanomedicines have already been Kojima,35 Jain et al.16 and Farokhzard et al.36
approved by the FDA. The major accomplishment in the
field is the phase I/II clinical trial development of Vivagels 2. Covalent drug binding to dendrimer termini
(SPL-7013) by StarPharma (Melbourne, Australia). Vivagels
is an anionic G4-poly(L-lysine)-type dendrimer functionalized Fig. 1 shows representative examples of drug–dendrimer
with thirty-two naphthalene disulfonate groups showing a conjugate linkers and the corresponding drugs. The main used
topical vaginal microbicidal activity. Completed phase I points of attachment are: amides, esters, disulfides, hydra-
dose-ranging studies in patients have demonstrated positive zones, thiol-maleimide and sulfinyls.
safety, tolerability, and pharmacokinetic properties of
2.1 Amide linkers
SPL-7013. Mild abdominal pain or discomfort are the main
adverse side effects. These studies represent a bridgehead for One of the earliest studies on drug-loaded dendrimers was
other clinical developments of dendrimers.5 reported by Zhuo et al.37 Poly(amido-amine) dendrimers,
Many reports on drug loaded-dendrimers have shown that with a 1,4,7,10-tetraazacyclododecane core, were partially
the release of the free drug can be enhanced by suitable choice acetylated leading to an enhancement of their water solubility.
of the linker between the dendrimer and the drug, especially, 5-Fluorouracil (5-Fu), an antimetabolite anticancer drug,
the linker/spacer length and flexibility. The stability of the typically administered with leucovorin and acting as an
bond between the dendrimer, linker and drug under physio- irreversible inhibitor of thymidylate synthase, was attached
logical conditions as well as the lability of the linker–drug by a simple amide linker to the acetylated dendrimers leading
bond in desired tissues are the key-factors for specific drug to 5-FU–dendrimer conjugates (Scheme 1). Assays on the

Mosto Bousmina was a full Jean-Pierre Majoral is an


professor and Canada research Emeritus Director of
Chair on Polymer Physics and Researches Exceptional Class
Nanomaterials at Laval at CNRS. After his PhD he
University, Canada, from worked as a Post-doc with
1994 to 2008. He then went A. Katritzky (Norwich, UK).
to Morocco, where he is He became Directeur de
presently the General Director Recherche at CNRS in 1978.
of INANOTECH and the He was in charge of a French-
Chancellor of the Hassan II Polish European Institute
Academy of Science and Tech- (LEA, 2000–2008). His
nology. He is the author research interest is mainly
of more than 150 scientific focused on main group
publications and 6 patents. elements, especially phosphorus
Mosto Bousmina He received various awards: Jean-Pierre Majoral in different areas of chemistry.
Louis-Pasteur Award in Presently, he is involved in the
1993, Innovation Award from the Quebec Ministry of preparation and properties of dendrimers and hyperbranched
Commerce and Technology in 1998, and Canada-top-20 polymers with emphasis on their applications in different fields
explorer Award in 2002 from CIAR (Canadian Institute for ranging from biology, nanomaterials and catalysis. He is a
Advanced Research), International Morand Lambla Award member of several Academy of Sciences worldwide and received
from the Polymer Processing Society in 2000 and the prestigious various international awards. He is the author of around 500
Steacie Award from NSERC-Canada in 2005. publications and 40 patents.

228 New J. Chem., 2012, 36, 227–240 This journal is c The Royal Society of Chemistry and the Centre National de la Recherche Scientifique 2012
Published on 08 September 2011. Downloaded by MIT Library on 2/10/2021 12:35:19 AM. View Article Online

Fig. 1 Linkers used for the attachment of different drugs onto dendrimers. The arrow indicates the side of site of attachment of the drug to the
dendrimer.

acting by intercalating the DNA, is linked to ST-PHPMA–


DOX (conjugate B) via an amide linkage. The final
ST-PHPMA–DOX dendrimers are obtained with good overall
yields. Additionally, linear N-(2-hydroxypropyl)methacryl-
amide copolymers containing DOX (HPMA–DOX) as well
as the DOX–HPMA copolymer conjugate via a tetrapeptide
sequence (GFLG–DOX, conjugate A) were also prepared. The
cytotoxicity of ST-PHPMA–DOX toward human ovarian
carcinoma A2780 cells was evaluated and compared to the
plain drug (DOX), HPMA–DOX and GFLG–DOX
conjugates. The biological results showed that cytotoxicities
of all conjugates were lower than the free DOX and the most
cytotoxic conjugate was ST-PHPMA–DOX followed by
GFLG–DOX and then HPMA–DOX. The authors proposed
that the differences in cytotoxic activities may result from the
difference in the rate of DOX released by lysosomal enzymes
via amido group cleavage and, probably, the cellular penetra-
tion rate (Scheme 2).
Another example was reported by Jiang’s group39 and
consists of the synthesis of different DOX–PEGylated
PAMAM conjugates. DOX was conjugated to differently
Scheme 1 Poly(amide-amine) dendrimer–5-FU conjugates.
PEGylated PAMAM dendrimers by two different amide link-
age types. The first linker is pH-sensitive (cis-aconityl linkage)
release of the conjugate have shown that hydrolysis in a and the second linker is pH-insensitive (succinic linkage),
phosphate buffer solution (pH 7.4) at 37 1C released free which leads to PCD and PPSD conjugates, respectively
5-FU. However, the amount of the released 5-FU is influenced (Scheme 3). In vitro evaluation studies have shown that
by the conjugates generation and the highest drug release was DOX release from pH-sensitive conjugates increased with
obtained with the highest conjugate generation. increasing PEGylation degree. In addition, cytotoxicity of
Kopecek and collaborators38 have reported the synthesis of pH-sensitive conjugates against ovarian cancer (SKOV-3) cells
original star-like HPMA copolymers, which were prepared by increased, while cellular uptake decreased with increasing
the reaction of semitelechelic poly[N-(2-hydroxypropyl)- PEGylation degree. In contrast, pH-insensitive conjugates
methacrylamide] macromolecules (ST-PHPMA), bearing released negligible drug concentration at any tested pH
chemically reactive ester groups, with amino end-groups of conditions explaining their low cytotoxicity. The conjugates
G2, G3, and G4 PAMAM dendrimers. Doxorubicin (DOX), were internalized in SKOV-3 cells via successively clathrin-
an anthracycline antibiotic, used in cancer chemotherapy and mediated and adsorptive endocytosis processes. DOX was

This journal is c The Royal Society of Chemistry and the Centre National de la Recherche Scientifique 2012 New J. Chem., 2012, 36, 227–240 229
Published on 08 September 2011. Downloaded by MIT Library on 2/10/2021 12:35:19 AM. View Article Online

Scheme 4 Synthesis of G2.5-PAMAM–MTX (conjugate A). (for


conjugate B, G3-PAMAM–MTX, a similar procedure was used).

amine-terminated G3 dendrimer and the carboxylic acid group


of MTX (conjugate B). Interestingly, 24-fold improvement of
the half maximal inhibitory concentration value (IC50) was
observed with conjugate A against MTX resistant CEM/MTX
and RII cell lines compared to free MTX. In contrast,
Scheme 2 Structure of HPMA copolymer–DOX conjugates.
conjugate B showed relatively little cytotoxicity versus
conjugate A. The authors argued that the difference in the
activities between the anionic and cationic conjugates is
probably due to the distinct intracellular release profiles of
the drug from the conjugates.

2.2 Ester linkers


D’Emanuele et al.41 have reported the synthesis and the
in vitro evaluation of new drug carrier systems based on
first-generation G1-PAMAM dendrimers. Terfenadine (Ter)
is an antihistaminic compound formerly used for the treatment
of allergies. Ter is a potent-glycoprotein (P-gp) efflux substrate
and a water-insoluble drug. Ter was attached to the dendrimer
surface using either a succinyl linker (single ester linker)
leading to the G1–suc–Ter conjugate or a succinicdiethylene
glycol linker (double ester linker) leading to the G1–suc-deg–
Ter conjugate (Scheme 5). Two lauroyl chains (L) were
attached to G1–suc-deg–Ter and give the L2–G1–suc-deg–Ter
Scheme 3 PPCD conjugate and PPSD conjugate. derivative which led to an enhancement of its permeability in
the Caco-2 model (the Caco-2 monolayers model for the
delivered by acidic lysosomes from pH-sensitive conjugates. prediction of intestinal drug absorption). All conjugates were
Then, DOX diffused into the nuclei of the cell to induce its more hydrophilic than the free drug and the biological assays
biological activity. Interestingly, pH-sensitive conjugates with indicated that the conjugates had no impact on the viability of
the highest PEGylation degree showed the best tumour accu- Caco-2 cells up to a concentration of 1 mM.
mulation in mice inoculated with SKOV-3 cells. Baker, Jr et al.42 have synthesized PAMAM dendrimer-
In another interesting report, Kannan and collaborators40 based multifunctional therapeutic devices. In addition to
have studied the cytotoxicity of dendrimers linked with MTX which was attached to the PAMAM dendrimers via
methotrexate (MTX). MTX is an antimetabolite anticancer an ester linker, imaging agents such as fluorescein isocyanate
drug acting as an inhibitor of dihydrofolate reductase. The (FI) or fluorescein isothiocyanate (FITC) were attached
authors have prepared two different PAMAM dendrimer– through urea or thiourea linkage. Moreover, folic acid (FA)
MTX conjugates (Scheme 4) using two different amide linkage was attached via an amide function and was used as a specific
types. The two starting molecules were the anionic half targeting agent (Scheme 6). In other reports, Baker, Jr and
generation G2.5 PAMAM dendrimer and the cationic G3 co-workers43,44 have shown that the G5–FI–FA–MTX conju-
PAMAM dendrimer. The first amide bond was achieved by gate binds, internalizes, and induces cytotoxicities specifically
the reaction between the carboxylic acid-terminated G2.5 in the overexpressed FA receptor-expressing KB cells in vitro.
dendrimer and the amine group of MTX (conjugate A). The This interesting result demonstrates clearly the applicability
second amide bond was the result of the reaction between the of dendrimers for the specific delivery of drugs into cells.

230 New J. Chem., 2012, 36, 227–240 This journal is c The Royal Society of Chemistry and the Centre National de la Recherche Scientifique 2012
Published on 08 September 2011. Downloaded by MIT Library on 2/10/2021 12:35:19 AM. View Article Online

Scheme 7 G5-PAMAM–RFn–MTXm conjugate.

(also known as Taxols) is a well known mitotic inhibitor used


in cancer chemotherapy. The synthesis started by the neutra-
lization of primary amino groups on the surface of the
G5-PAMAM dendrimer by partial acetylation, leading to
enhanced solubility of the dendrimer itself. Then, FITC, FA
and paclitaxel were successively conjugated to the remaining
nonacetylated primary amino groups using thiourea, amide
Scheme 5 G1–suc-deg–Ter and L2-G1–suc-deg–Ter conjugates.
and diester linkages, respectively (Scheme 8). These multi-
functional dendrimer conjugates have been tested in vitro for
targeted delivery of chemotherapeutic and imaging agents to
specific cancer cells. In vitro testing has shown uptake and the
cytotoxic effect of the conjugate.
In another interesting report, the kinetic rate of the hydro-
lysis of the dendrimer-Taxol conjugates in phosphate buffered
saline (PBS) was investigated.49 For the preparation of
G5–FITC–FA–Taxol conjugates, the Taxol moiety was linked
to the G5-PAMAM dendrimer unit using either succinic acid
(G5-PAMAM–FITC–su–Taxol) or glutaric acid (G5-PAMAM–
FITC–glu–Taxol) linkers (Scheme 9). This study showed that
the succinic acid linker–dendrimer conjugate released the
Taxol drug through bond hydrolysis in a time-dependent
fashion with a half-life of 10 hours. In contrast, the glutaric
acid linker–dendrimer conjugate was very stable and did not
release the Taxol compound even after seven days under
similar experimental conditions. Strongly, this result confirms
Scheme 6 Schematic representation of the G5–FITC–FA–MTX
conjugate.
the use of succinic acid as a useful linker.
Chai and collaborators50 have reported an elegant synthesis
of novel water-soluble and photostable dendrimers containing
Recently, G3–PAMAM–FI–MTX was synthesized by linking
the 3,4-dihydroxy-L-phenylalanine (L-DOPA) scaffold, which
both MTX and FI via ester bonds. The conjugate was able to
target FR-expressing cells without the need to conjugate FA.45
In a recent report, Haandel and Stobaugh46 have developed
a bioanalytical assay for the detection of the G5–MTX–FA
conjugate, and the released MTX as well as its main plasma
metabolite 7-hydroxymethotrexate. Recently, Baker’s group47
has published an interesting work on the synthesis of
G5–MTX conjugates via an ester linkage. They used riboflavin
(RF) as a specific targeting ligand (Scheme 7). Thus, the
G5–FITC–RF–MTX conjugate was synthesized and was
evaluated in vitro against KB cells. The biological results have
shown that the conjugate was carried into the cells through a
RF-receptor mediated mechanism, which can lead to a specific
release of MTX.
Following the same approach, Baker’s group48 has Scheme 8 Schematic representation of the G5–FITC–FA–Taxol
synthesized the G5–FI–FA–paclitaxel conjugate. Paclitaxel conjugate.

This journal is c The Royal Society of Chemistry and the Centre National de la Recherche Scientifique 2012 New J. Chem., 2012, 36, 227–240 231
View Article Online

Scheme 9 G5-PAMAM–FITC–su–Taxol and G5-PAMAM–FITC–


su–Taxol conjugates.

is a commonly used drug to treat Parkinson’s disease. The


Published on 08 September 2011. Downloaded by MIT Library on 2/10/2021 12:35:19 AM.

L-DOPA-dendrimers were synthesized by connection of indi-


vidual L-DOPA moieties to one another via hydrolysable Scheme 11 Schematic representation of the Taxol loaded triazine
diester linkages (Scheme 10). A G3-generation L-DOPA dendrimer.
dendrimer, containing 30 L-DOPA residues, showed a 20-fold
increase in aqueous solubility and enhanced photostability in These new dendrimer conjugates contain two Bolton–Hunter-
solutions over L-DOPA under identical experimental condi- type groups, used for biodistribution studies, and multiple
tions. L-DOPA-dendrimers represent a useful biocompatible Taxol groups attached by labile ester linkage. The conjugates
dendrimeric scaffold similar to the well known examples of were PEGylated leading to a water soluble drug–dendrimer
polyether-scaffold dendrimers,51 polyester-scaffold dendrimers52 conjugate.
and recently polypropylimine-scaffold dendrimers.53 In another study, Kannan et al.55 have reported the synthesis
Lim and Simanek54 have described the synthesis of triazine of simple hyperbranched polyol scaffolds and G4-OH PAMAM
dendrimers bearing the Taxol moiety on their surfaces (Scheme 11). dendrimers bearing methyl prednisolone (MP) (Scheme 12).

Scheme 10 Structure of the G3-L-DOPA dendrimer.

232 New J. Chem., 2012, 36, 227–240 This journal is c The Royal Society of Chemistry and the Centre National de la Recherche Scientifique 2012
Published on 08 September 2011. Downloaded by MIT Library on 2/10/2021 12:35:19 AM. View Article Online

Scheme 12 MP–G4-PAMAM dendrimer conjugate.


Scheme 13 Cationic G4-NH2 and anionic G3.5-COOH PAMAM
MP is an adrenocortical steroid typically used in the anti- dendrimers bearing NAC.
inflammatory field. In this study, the MP was conjugated to
PAMAM dendrimers and polyol scaffolds using either a anti-inflammatory agent, when compared to free NAC in vitro.
glutaric acid (GA) or a succinic acid (SA) spacer. The number Even at a low dose (0.5 mM of NAC) the NAC–dendrimer
of drug payload was the highest with polyol whatever the conjugate showed a high inhibitory rate. The same group59 has
spacer type, while in the case of the PAMAM dendrimer, the reported the synthesis of two different NAC–G4-PAMAM
higher payload was achieved with the GA than the SA spacer. dendrimer conjugates using two disulfide linkers (GSH-sensitive
The anti-inflammatory activity of these MP–polymer conju- linker): the first conjugate with a cationic G4-PAMAM–NH2
gates was assessed by the evaluation of the inhibition of and the second with anionic G3.5-PAMAM–COOH dendri-
prostaglandin synthesis. The inhibition potency follows the mers (Scheme 13).
order: MP–SA–dendrimer conjugate 4 MP–GA–polyol con- The results of NAC release from the conjugates have shown
jugate 4 MP–GA–dendrimer conjugate. In conclusion, in this that 70% of NAC payload was released at intracellular GSH
study, Kannan’s group has shown that drug release from concentration and only negligible NAC release was observed
hyperbranched polymer–drug conjugates and the subsequent at extracellular GSH levels. FITC-labeled conjugates showed
activity are influenced by the branching architecture and the rapid penetration and localization in the cytoplasm of lipo-
type of the linker. Interestingly, the same group has conju- polysaccharide (LPS)-activated microglial cells. Both G4-NH2
gated MP to the G4-PAMAM dendrimer using a diester and anionic G3.5-COOH PAMAM–NAC conjugates showed
linkage.56 The intranasal administration of the MP–dendrimer better nitric inhibition when compared to free NAC.
conjugate was effective in reducing the ovalbumin-induced
airway inflammatory response in a mouse model. Moreover, 2.4 Hydrazone linkers
significant efficacy of the drug was observed even at the lowest To date, few works on dendrimers bearing drugs
dose tested and the MP–dendrimer conjugate was able to through hydrazone bonds have been reported. Fréchet and
reduce the inflammatory reaction. It is noteworthy that the collaborators60 have synthesized dendritic polyester systems
MP–dendrimer conjugate has no undesired effects such as using 2,2-bis(hydroxymethyl)propanoic acid as the building
non-specific inflammation and has a longer residence time in block. This new family of dendrimers has excellent water
the lungs. solubility. To investigate the feasibility of the drug–dendrimer
In continuation of their work, Baker, Jr et al.57 have conjugate, authors have successfully attached DOX using a
reported the simultaneous conjugation of FA and MTX to hydrazone bond (pH sensitive linkage) (Scheme 14).
the G5-PAMAM dendrimer through ester linkages in a ‘‘one Recently, the same group61 has reported the synthesis of a
pot’’ approach (see Scheme 4). In vitro studies performed on new tubulysin analogue, a potent anticancer drug, which was
FA receptor-expressing KB cells have shown that the also conjugated via a hydrazone linker (acid labile bond) to the
G5–FA–MTX conjugate has a similar affinity and cytotoxic PEGylated dendrimer with a polypeptide core. The conjugate
potency to G5–FA–MTX synthesized using the traditional has a higher water solubility compared to the free tubulysin
multiple-step approach. analogue and shows no sign of toxicity, even at high doses of
the conjugate. In addition, the conjugate exhibited a longer
2.3 Disulfide linkers
lifespan (90% increase on average) (Scheme 15).
Kannan et al.58 have reported the synthesis of PAMAM- 5-Aminosalicylic acid (5-ASA), an anti-inflammatory agent,
COOH containing N-acetyl cysteine (NAC, Fluimicils). NAC was attached to the PAMAM dendrimer using two different
was used in different therapeutical indications such as spacers, p-aminobenzoic acid (PABA) and p-aminohippuric
anti-inflammatory and for the treatment of paracetamol acid (PAH). The new water-soluble PAMAM dendrimer–
(acetaminophen) overdose. The conjugate was prepared using 5-ASA conjugates with a designed spacer for colonic delivery62
a disulfide linker (glutathione sensitive linker) which enables have shown colon-specific and gradual release of 5-ASA after
relatively rapid intracellular release of the free drug. incubation with rat cecal contents at 37 1C (45.6% from
The new conjugate is non-toxic even at the highest concentra- PABA linker/dendrimer and 57% from PAH linker/dendrimer
tion tested in vitro and is a more effective antioxidant and conjugates). Based on these results Wiwattanapatapee et al.62

This journal is c The Royal Society of Chemistry and the Centre National de la Recherche Scientifique 2012 New J. Chem., 2012, 36, 227–240 233
Published on 08 September 2011. Downloaded by MIT Library on 2/10/2021 12:35:19 AM. View Article Online

Scheme 16 Glycopeptide dendrimer–colchicine conjugates.

Scheme 14 Dendritic polyester–DOX conjugate. have suggested that these dendrimers have potential for use as
colon-specific drug carriers.

2.5 Thiol-maleimide and sulfinyl linkages


Barth and coworkers63,64 have reported the first example of
urea linkage using thiol-maleimide coupling for the synthesis
of boronated monoclonal antibodies–dendrimer conjugates.
These conjugates were designed as delivery systems for
neutron capture therapy. Later, Wängler and collaborators65
have loaded DOTA onto the PAMAM dendrimer using the
same linker type.
Colchicine, which is a natural product with high cyto-
toxicity, was loaded onto the peptide dendrimer using sulfinyl
acetamide66 (Scheme 16). In contrast to free colchicine,
biological results have shown that colchicine–dendrimer
conjugates present selective cytotoxicities toward cancer cell
lines, such as HeLa cells, than normal cells.

3. Ionic attachment to dendrimer termini


(cisplatin and copper)
In drug delivery approaches by dendrimers developed so far,
cisplatin (trade names Platinol and Platinol-AQ) is the most
investigated metallodrug. Cisplatin is a potent anticancer
agent used in chemotherapy to treat various types of cancers.67
Thus, cisplatin was loaded into dendrimers using two
different coordination strategies (Scheme 17). (i) (NH2)2-Pt–
(dendrimer)2: the bonds between the platinum entity and the
dendrimer should be labile and should lead to the release of
the free drug, ‘Cl2Pt(NH2)2’.68 (ii) (Cl)2-Pt–(N-dendrimer)2:
Scheme 15 Tubulysin analogue–PEGylated dendrimer conjugate. the amine group on the dendrimer (at least one N–H moiety)

234 New J. Chem., 2012, 36, 227–240 This journal is c The Royal Society of Chemistry and the Centre National de la Recherche Scientifique 2012
Published on 08 September 2011. Downloaded by MIT Library on 2/10/2021 12:35:19 AM. View Article Online

Scheme 18 G0-PAMAM–py4–[PtCl2]4 and G0-PAMAM–py4–[CuCl2]7


Scheme 17 Synthesis of cisplatin dendrimer conjugates. conjugates.

and Chang liver and slightly active toward MOLT-4 cells.


could establish hydrogen bonding and the conjugates may bind
However, G0-PAMAM–py4–[CuCl2]7 displayed higher cyto-
directly to DNA without the release of the free cisplatin.69
toxicity than both platinum conjugates and free cisplatin itself
In early studies, Malik et al.70 conjugated G3.5 PAMAM-
against MOLT-4 and MCF-7 cells. This difference in cyto-
CO2H dendrimers with cisplatin giving PAMAM–platinate
toxicity may be attributed to the difference in solubility,
complexes (mono-, bi- and cross-linked-dentates). These
number of metals loaded onto the dendrimer and their
complexes are less toxic than cisplatin itself and release
coordination type with the ligands. Other types of conjugates
platinum slowly in vitro. In in vivo tests, the dendrimer–Pt
have also being reported using ionic attachments. For
displayed similar potency as cisplatin administered by the
example, cis-dichlorodiammineplatinum(II), a derivative of
intraperitoneal route against the L1210 leukemia tumour
cisplatin, was loaded onto dendrimer phthalocyanine and
model in mice. At high doses, dendrimer–Pt, given by the
poly(ethylene glycol)-block-poly(L-aspartic acid)74 which led
intraperitoneal route, showed good activity against the
to polymer–metal complex micelles.75
B16F10 melanoma model whereas cisplatin did not. Intravenous
administration of dendrimer–Pt induced potency against the
subcutaneous B16F10 model, while cisplatin was inactive. After
4. Comparison between different linkers
intravenous injection of dendrimer–Pt and cisplatin, selective
accumulation of dendrimer–Pt in solid tumour versus blood Various reports have investigated the difference between
was observed probably due to the EPR effect (vide supra). linkers and their effect for the release of drugs under similar
Biocompatible anionic linear-globular dendrimer–cis– experimental conditions. In this paragraph, we review the
platinum(II) conjugates have been described.71 The G2-citric- different studies reported so far.
acid-based dendrimer showed potent in vitro cytotoxic activities Baker et al.76 have reported the synthesis of the trifunctional
against HT1080 and CT26 cell lines (up to 8 fold versus plain G5-(Ac)96–FITC5–FA3–MTX4 conjugate which provide a
cisplatin) and, interestingly, against the Pt resistant cell line, targeting, imaging, and intracellular delivering system.
SKOV3 (B2 fold versus plain cisplatin), whereas the corres- MTX was conjugated either through an amide linkage (non
ponding G1 dendrimer showed lower potencies. Hemolytic biodegradable) or through an ester linkage (hydrolyzed in the
impacts and cell death mechanisms of these conjugates on endosome at low pH). Although both types of MTX triple
human blood and HT1080 cell line were investigated as well. conjugates were internalized into KB cells in vitro, only the
The same hemolysis behaviour (1.5, 3 and 6 mg ml1) was MTX conjugated through an ester linkage was cytotoxic. The
observed for all conjugates except for the G2–Pt complex at conjugates (G5–FITC–FA) with amine-surfaced nanodevices
the concentration of 6 mg ml1. Both apoptosis and necrosis showed minimal uptake at 30 nM with KB cells and G5–FITC
mechanisms (B2 fold versus cisplatin) were attributed to gave similar uptake, which proved that the binding of both
conjugates and cisplatin in a direct correlation between the conjugates (G5–FITC–FA and G5–FITC) was not specific to
concentration and the degree of cell death. the FA receptor. However, the modified surfaces containing
Kokotos and coworkers72 have synthesized poly-propylene- either carboxy, 2,3-dihydroxy propyl or acetamide moieties on
imines containing chiral amines with 8, 16 and 32 amino the nanodevice surfaces bind efficiently and rapidly to KB
groups on the surface. Dendrimers with cisplatin moieties at cells. In this case, the controlled nanodevice without FA did
the periphery were obtained by the reaction between the not bind to KB cells. It is noteworthy that an equimolar
free amine dendrimers and potassium tetrachloroplatinate(II) concentration of the free drug (MTX) was 4-fold less effective
leading to highly insoluble conjugates. in killing the tumour cells than the drug conjugated through
Chu and his group73 have recently prepared platinum and ester linkage. The authors observed also that a drug conju-
copper metallodendrimer conjugates (Scheme 18). gated through amide linkage, and the conjugate without a
The condensation reaction of platinum chloride and copper drug were not toxic at any tested concentration.
chloride afforded G0-PAMAM–py4–[PtCl2]4 and G0-PAMAM– Using polyester dendrimer–PEO hybrids, Fréchet and
py4–[CuCl2]7, respectively. The conjugate cytotoxicities were collaborators77 have reported the synthesis of DOX–[G3]-
studied toward leukemia cells, breast cancer cells and Chang (PEO5k)8-[G4]-(OH)16 bow-tie dendrimers using either
Liver cells. G0-PAMAM–py4–[PtCl2]4 was inactive to MCF-7 carbamate (stable) or acyl hydrazone (pH-labile) linkages.

This journal is c The Royal Society of Chemistry and the Centre National de la Recherche Scientifique 2012 New J. Chem., 2012, 36, 227–240 235
Published on 08 September 2011. Downloaded by MIT Library on 2/10/2021 12:35:19 AM. View Article Online

Scheme 19 G0-PAMAM–Naproxen conjugates.

The DOX–dendrimer conjugate through hydrazone linkage


was eliminated from serum with a half life of 16 hours and
its tumour uptake was nine-fold higher than free DOX at
48 hours. It was noticed that a single injection of the
DOX-dendrimer (20 mg kg1 DOX equivalents), 8 days after
tumour implantation, caused complete tumour regression and
100% survival of the mice over the 60-days experiment.
Scheme 20 Dendrimers 1–3 with 2 0 -ester and disulfide linkages.
Biological results have shown also that no cure was achieved
in tumour-implanted mice treated with free DOX at its
tolerated dose, a drug free dendrimer, and DOX–dendrimer In another report, Simanek et al.80 have reported the
through carbamate linkage. synthesis of triazine dendrimers bearing Taxol using either
D’Emanuele et al.78 have reported the synthesis and evalua- ester or ester/disulfide linkages (Scheme 20). Two different
tion of G0-PAMAM-Naproxen conjugates, which were PEG-N-hydroxysuccinimide (PEG-NHS) esters were used: for
designed in order to enhance drug solubility and bioavailability. conjugates 1 and 2, the PEG incorporates an ester-linked
Naproxen, a poorly water-soluble anti-inflammatory drug, succinic acid group, whereas, the PEG attached to conju-
was directly conjugated to the G0-PAMAM dendrimer via gate 3 incorporates an ether-linked acetic acid group
an amide bond or via an ester bon7d using either l-lactic acid (Scheme 20).
or diethylene glycol as a linker (Scheme 19). The synthesized In this study, it was shown that the three conjugates
conjugates have shown better water solubility than free (1, 2 and 3) appear to be well tolerated in vivo at doses equal
Naproxen. All conjugates were chemically stable over 48 h to 200% the maximum tolerated dose of Taxols.
of incubation at different pH values such as: hydrochloric acid In addition, the tumour localization of the conjugates was
buffer (pH 1.2), phosphate buffer (pH 7.4), and borate buffer observed at low levels and the elimination half-lives of the
(pH 8.5). However, Naproxen was enzymatically released three conjugates were similar.
from both ester conjugates in plasma (80% human plasma) Recently, Kannan and coworkers81 have published a very
and the release from the lactic ester conjugate was slower than interesting work comparing the drug release from polymer–
from the diethylene glycol ester conjugate. drug conjugates using different linkers. To understand the
As part of their research program, Kono and coworkers79 architecture and the linker effect on the release of ibuprofen, a
have synthesized G4-PAMAM dendrimers having a glutamic non-steroidal anti-inflammatory drug, they synthesized
acid (Glu) residue as the end group, then, they have attached G4-PAMAM dendrimer–ibuprofen conjugates using ester,
PEG chains to amino groups of Glu residues. DOX was amide, and peptide linkers, in addition to a linear mPEG–
conjugated to side chains of the Glu residues using either an ibuprofen conjugate. Comparing the amide linker versus the
amide bond, [PEGeGlu(DOX)-G4], or a hydrazone bond, ester linker, the amide was more stable against hydrolysis,
[PEGe Glu(NHNe DOX)-G4]. Biological results have shown whereas the ester-linked conjugates showed pH-dependent
that only dendrimers bearing DOX through hydrazone release. This study has shown also that direct linked conju-
linkage were stable under physiological conditions, and gates (amide- and ester) did not release ibuprofen enzymati-
released free DOX drug under acidic conditions such as cally in cathepsin B buffer and diluted human plasma. In
those of endosomes and lysosomes. These new conjugates contrast, the mPEG conjugate released 65% of its payload
showed much lower toxicity against HeLa cells than free within 12 h in diluted plasma by esterase activity, and the
DOX. In addition, PEGeGlu(NHNeDOX)-G4 was 7-fold peptide-linked dendrimer conjugate released 40% of its
cytotoxic than PEGeGlu(DOX)-G4, which demonstrates payload within 48 h by cathepsin B activity. It is noteworthy
the importance of pH-sensitive hydrazone linkage for high that the steric crowding at the surface of PAMAM
cytotoxicity. dendrimer–drug conjugates, along with linking chemistry

236 New J. Chem., 2012, 36, 227–240 This journal is c The Royal Society of Chemistry and the Centre National de la Recherche Scientifique 2012
View Article Online

5. Dendrimer conjugates versus dendrimer


complexes or polymer conjugates
5.1 Conjugates versus complexes?
In vitro and in vivo comparative studies of non-covalent drug
inclusion complexes and covalently conjugated drugs have
been carried out by different teams.
Briefly, one of the major potential advantages of covalent
attachment of drugs to the termini surface groups of
dendrimers in comparison with the encapsulation technique
Published on 08 September 2011. Downloaded by MIT Library on 2/10/2021 12:35:19 AM.

is the better control over drug release. In fact, Baker and


co-workers25 have studied the release kinetics of methotrexate
from the folate–G5-PAMAM-dendrimer–methotrexate conjugate
and folate–G5-PAMAM-dendrimer–methotrexate encapsula-
tion. The authors emphasized that the dendrimer–drug
Scheme 21 Surface modification using different amino acid moieties. conjugate was more suitable for targeted drug delivery, while
the complex improved the solubility of methotrexate in
govern the drug release mechanism as well as its kinetics. aqueous solution and displayed similar cytotoxicity against
Using amino acids as linkers, Kannan’s research group82 has KB cancer cells than plain methotrexate.
prepared new biocompatible dendrimer scaffolds which could In addition, in a recent report, Jiang and coworkers85
be used in drug delivery. The peripheries of G4-PAMAM presented the comparative evaluation of in vitro and in vivo
dendrimers were converted in one-step into diverse peripheral behaviors of methotrexate coupled to PEGylated or
groups (Scheme 21) which could be chemoselectively conju- non-PEGylated G4-PAMAM dendrimers both encapsulated
gated to different drugs. Dexamethasone and indomethacin, and conjugated. The in vitro studies have shown that there
which are widely used anti-inflammatory drugs, were conju- were no strong differences between complexes and free
gated to the functional dendrimers using ester and amide methotrexate in cytotoxicity against KB cell lines, whereas
linkages, respectively, while the imaging agent FITC was the conjugates showed lower potency. Remarkably, in vivo
attached through a thiourea linkage type. The in vitro studies in rodents have shown that the methotrexate–
cytotoxicity and hemolysis assays have shown that the hetero- G4-PAMAM-dendrimer–PEG-conjugate exhibited a good
bifunctional dendrimers were noncytotoxic even at high prolonged blood residence time (in normal rats, i.v. adminis-
concentrations (100 mg mL1 to 1 mg mL1). tration) and strongest antitumour activity (in S-180 tumour-
Good and collaborators83 have shown that sialic acid bearing mice, i.v. administration).
functionalized dendritic polymers can act as mimics of cell The need to conduct more systematic investigations for
surface sialic acid clusters and attenuate Ab-induced neuro- drugs complexed or conjugated with dendrimers on the long-
toxicity. In continuation of their efforts in the development term pharmacodynamic (PD) and pharmacokinetic (PK)
of treatments for Alzheimer’s disease, Good et al.83 have behaviors has been brought up by Cheng and Xu.86
developed sialic acid dendrimer conjugates. Thus, two
different attachments were used to link sialic acid to PAMAM
5.2 Polymer therapeutics or dendrimer therapeutics?
dendrimers. The first linker was the result of the coupling
between PAMAM termini and the anomeric hydroxyl group As shown in Table 1, Patel et al.87 have nicely summarized the
of sialic acid (physiological attachment). The second bond main properties of dendrimers versus linear polymers. Briefly,
was obtained by the reaction between the PAMAM many polymers are highly heterogeneous whereas size and
termini and carboxylic acid (nonphysiological attachment) molecular mass of dendrimers can be highly controlled. For
(Scheme 22). Biological results have shown that the use of these reasons, dendrimers display significant improvements
the attachment of sialic acid via the anomeric hydroxyl when compared to linear polymers related to their physical
instead of attachment via the carboxylic acid did not give a characteristics such as polydispersity, structure, structural
significant enhancement of their ability to mitigate Ab induced control, architecture and compressibility as well as their
neurotoxicity.84 chemical characteristics such as easy synthesis and aqueous
solubility. The architecture of dendrimers induces a large
proportion of group exposition at their surfaces affording a
very high molecular surface to volume ratio (41000 m2 g1) in
contrast to classical linear polymers (80–300 m2 g1).
The main biological applications of polymer therapeutics
(biopolymers) encompass polymer–protein conjugates and
drug–polymer conjugates.88 Numerous polymer–protein
conjugates have been developed in order to improve the
Scheme 22 Sialic acid PAMAM conjugates using two different stability and the pharmacokinetic behaviors (prolongation of
linkages (conjugate A: carboxylic acid, conjugate B: anomeric the biological half-life) and to reduce the immunogenicity of
hydroxyl group). considered native proteins. PEG-polymers have mainly been

This journal is c The Royal Society of Chemistry and the Centre National de la Recherche Scientifique 2012 New J. Chem., 2012, 36, 227–240 237
View Article Online

Table 1 Main properties of dendrimers and linear polymers (adapted from Patel et al.)85

Properties Dendrimers Linear polymers


a
Programmed release of drugs High ( ) reduced toxicity, increased bioavailability, simplified dosing schedule )
Penetration abilities High (high cell membrane penetration ) an increased cellular uptake level of High/low
the drugs complexed or conjugated)
Penetration and retention Enhanced ( ) preferential uptake of the dendrimers by cancer tissues) No EPR effect
(EPR) effect
Immunogenicity Low
a
Size, shape, surface properties of the polymeric carriers highly influence the pharmacodynamic (PD) and pharmacokinetic (PK) behaviors of
drugs encapsulated in/complexed to/conjugated to the dendrimers.
Published on 08 September 2011. Downloaded by MIT Library on 2/10/2021 12:35:19 AM.

the polymers developed for the construction of polymer– 6. Targeted or nontargeted covalently conjugated
protein conjugates. For instance, PEGylated L-asparaginase, drugs?
adenosine deaminase, interferon-a (INF-a), styrene-co-maleic
anhydride-neocarzinostatin (SMANCS) and granulocyte In drug delivery by dendrimers, two different approaches have
colony stimulating factor (PEG-G-CSF) were described.89 been developed: active targeted (few examples) and passive
Interestingly, the synthesis of the PEGylated 125I-EGF-OX26 non-targeted drug–dendrimer conjugates (numerous examples).
monoclonal antibody construct has been described for For example, in the oncology domain, both targeted and non-
imaging human brain tumours.90 targeted drug–dendrimer conjugates can successfully extra-
Development of drug–polymer conjugates with cleavable vasate across tumour’s leaky vasculature and accumulate in
linkers to improve the therapeutic index of several toxic drugs, the cancer tissue.
especially in the cancer chemotherapy, has been described. Briefly, targeted drug–dendrimer conjugates have the
Currently, phase I and phase II clinical trials are ongoing or advantage to selectively bind cancerous cells versus normal
completed with different drugs such as, doxorubicin, taxol, healthy cells. The receptor on the surface of the cancer cells
camptothecin, methotrexate and platinate(II) derivatives. The increases the residence time on the cell surface and enhances
main copolymers used are HPMA (N-(2-hydroxypropyl)- the internalization kinetics into cells.14 In addition, targeted
methacrylamide), PEG, poly-L-glutamate, albumin, dextran drug–dendrimer conjugates allow the release of higher drug
and 6-maleimidocaproyl hydrazone derivatives. Different dose loaded in the ‘‘right place’’ (not a premature release of the
linker types were employed, namely: Gly-Phe-Leu-Gly, drug under biological conditions). Interestingly, Baker and
Gly-6-aminohexanoyl-Gly, alanine ester, Gly-ester, ester and co-workers have carried out outstanding kinetic studies
acid-sensitive hydrazone.88,89 showing that the targeted dendrimer conjugates are preferen-
In addition, self-assembling block copolymer micelles have tially taken up by the targeted cancer cells resulting mainly due
also been explored as drug carriers. For instance, pluronic to their longer residence times versus the endocytic rate.93
block copolymer micelles including doxorubicin are able to
circumvent p-glycoprotein-mediated resistance which has
7. Concluding remarks and perspectives
shown promising results in phase II clinical trials. The drug
was noncovalently entrapped in the micelle.91 A dual doxo- The chemical nature of the linker between the drug and the
rubicin covalently bonded to the PEG-poly(aspartic acid) dendrimer has a pivotal role in the design of efficient drug
copolymer entrapping also free doxorubicin has been delivery systems. This review summarizes the recent develop-
described by Kataoka and colleagues.92 This doxorubicin- ments in the design of covalent linkers such as amides, esters,
copolymer accumulates preferentially in tumour tissue diesters, disulfides, hydrazones, thiol-maleimide and sulfinyls
through the EPR effect inducing its biological anti-cancer and their efficiency in terms of drug delivery and therapeutic
activity. biological activity. The cases of the ionic attachments for the
One other important biomedical application of biopolymers metallodrugs cisplatin and copper chloride were reported
is the destruction of angiogenic vasculature of solid tumours. and discussed as well. Some of the reviewed linkers are
Several targeted delivery systems were described such as: pH-sensitive and have proven to enhance intracellular release
polymer–angiogenesis inhibitor conjugates (e.g. HPMA- of the free drug. The biological evaluations have shown that,
TNF-470), antiendothelial immunoconjugates (e.g. L19-inter- in most cases, the conjugation of a drug with a dendrimer via a
leukin 2, L19-astatine 211), antiendothelial fusion proteins linker leads to great enhancement of the biological activity
(e.g. endostatin-human prolactin antagonist), antiendothelial along with the reduction in toxicity compared to free drugs.
peptide conjugates (e.g. NGRpeptide–doxorubicin) and anti- For example, in the case of nanodevices engineered for cancer
endothelial liposomes (e.g. APRPGpeptide–liposome– therapy, the pH-sensitive linkers can be cleaved more rapidly
DPP–CNDAC (antitumour nucleoside)). These approaches in cancer cells (endo-lysosomes) than under normal physio-
showed very promising results in in vitro and in vivo models logical conditions (normal cells). However, the nonspecific
and had apparently no specific toxicities. Combination of release of free drugs both in cells and tissues remains a major
polymeric therapeutics in cancer chemotherapy including challenge. To overcome the difficulty, new delivery systems
oxidative stress inducers or antiangiogenic agents has been containing the drug and targeting ligand, both connected to
also designed.88,89 the same dendrimer (preferentially PEGylated to guarantee

238 New J. Chem., 2012, 36, 227–240 This journal is c The Royal Society of Chemistry and the Centre National de la Recherche Scientifique 2012
View Article Online

biocompatibility and long blood circulating time with a focused on specific disease at the cellular level (e.g. cell mem-
sustained release effect), are under active investigation. brane, nucleus, endolysosomes etc.), organ level (e.g. lung,
In addition, the multi-functional nature of the dendrimer spleen, stomach, liver etc.) and/or tissue level (e.g. tumour
surface allows combining drugs, solubilising groups such as cells etc.).
PEG chains in the same surface unit. One of the important
additional advantages of the introduction of a PEG chain is
Acknowledgements
the inhibition of the blood serum opsonins by the PEGylated
dendrimer allowing its non-recognition by the phagocytic The Hassan II Academy of Science and Technology is warmly
cells. It is important to note that the surface-modified systems acknowledged for the financial support.
can operate differently than the native dendrimers. The type of
Published on 08 September 2011. Downloaded by MIT Library on 2/10/2021 12:35:19 AM.

linker used to covalently link drugs to the surface termini of


Notes and references
the dendrimers affects directly the biological activity of the
drug–dendrimer conjugates and can be used to control the rate 1 G. R. Newkome and C. Shreiner, Chem. Rev., 2010, 110, 6338.
2 D. Astruc, E. Boisselier and C. Ornelas, Chem. Rev., 2010,
of drug release (vide supra). The nanometric size of dendrimers
110, 1857.
induces their entry into hyper-permeable vasculature of 3 A. M. Caminade and J. P. Majoral, Chem. Soc. Rev., 2010,
tumour or inflamed tissues by the enhanced permeability 39, 2034.
and retention effect (EPR effect), whereas their high molecular 4 T. D. Patel, B. N. Parikh, G. D. Gothi, J. B. Dave and C. N. Patel,
J. Global Pharma Technol., 2010, 2, 5.
weight and the lymphatic dysfunctioning causes their specific 5 M. A. Mintzer and M. W. Grinstaff, Chem. Soc. Rev., 2010,
localization. In addition, the presence of hydrophobic terminal 40, 173.
surface groups on the dendrimers prevent their recognition by 6 M. Calderón, M. A. Quadir, M. Strumia and R. Haag, Biochimie,
macrophages and their elimination by the Reticuloendothelial 2010, 92, 1242.
7 C. M. Paleos, D. Tsiourvas, Z. Sideratou and L.-A. Tziveleka,
System (RES).14,94,95 Expert Opin. Drug Delivery, 2010, 7, 1387.
Moreover, the development of new drug-loaded dendrimers 8 J. M. Oliveira, A. J. Salgado, N. Sousa, J. F. Mano and R. L. Reis,
cleavable by specific enzymes and other biological molecules Prog. Polym. Sci., 2010, 35, 1163.
9 A.-M. Caminade, C.-O. Turrin and J.-P. Majoral, New J. Chem.,
present exclusively in the targeted cells is also a very attractive 2010, 34, 1512.
research area and may resolve the problem of non-specific 10 M. Raviña, P. Paolicelli, B. Seijo and A. Sanchez, Mini-Rev. Med.
release. These novel approaches open new avenues to cancer Chem., 2010, 10, 73.
and other disease treatments. It’s not only a supplement to the 11 S. Zhu, M. Hong, L. Zhang, G. Tang, Y. Jiang and Y. Pei, Pharm.
Res., 2010, 27, 161.
conventional chemotherapy and radiotherapy but also prevents 12 M. Zablocka, A. Hameau, A. M. Caminade and J. P. Majoral,
damage of normal tissues and prevents drug resistance. Adv. Synth. Catal., 2010, 352, 2341.
We expect that future research on the development of drug- 13 A. M. Caminade, A. Hameau and J. P. Majoral, Chem.–Eur. J.,
2009, 15, 9270.
linked dendrimers will face different important remaining
14 S. H. Medina and M. E. H. El-Sayed, Chem. Rev., 2009, 109, 3141.
challenges which could be summarized as follows: 15 J. R. Baker Jr., Am. Soc. Hematol., 2009, 708.
(a) Increase the degree of control in drug delivery by the 16 R. K. Tekade, P. V. Kumar and N. K. Jain, Chem. Rev., 2009,
design and the synthesis of novel specific linkers that will be 109, 49.
17 O. Rolland, C.-O. Turrin, A.-M. Caminade and J.-P. Majoral,
recognized and selectively cleaved by biological molecules New J. Chem., 2009, 33, 1809.
present exclusively in the desired targeted cells versus normal 18 G. R. Newkome and C. D. Shreiner, Polymer, 2008, 49, 1.
cells. The development of cell-based therapies for diseased 19 P. Niederhafner, M. Reinis, J. Sebestik and J. Jezek, J. Pept. Sci.,
tissues is probably one of the most promising research fields in 2008, 14, 556.
20 A. M. Caminade, C. O. Turrin and J. P. Majoral, Chem.–Eur. J.,
regenerative medicine. 2008, 14, 7422.
(b) Synthesis of dendrimer engineering towards tissues or 21 A. M. Caminade, P. Servin, R. Laurent and J. P. Majoral, Chem.
specific cellular compartments targeting (e.g. lung, skin, Soc. Rev., 2008, 37, 56.
22 D. A. Tomalia, L. A. Reyna and S. Svenson, Biochem. Soc. Trans.,
nucleus, mitochondria . . .). 2007, 35, 61.
(c) Development of clusters of dendrimers for the assembly 23 Y. Cheng, Z. Xu, M. Ma and T. Xu, J. Pharm. Sci., 2007, 97, 123.
of multifunctional therapeutic systems (e.g. combined anti- 24 E. R. Gillies and J. M. J. Fréchet, Drug Discovery Today, 2005,
cancer therapy . . .). 10, 35.
25 A. K. Patri, J. F. Kukowska-Latallo and J. R. Baker Jr., Adv. Drug
(d) Synthesis of biocompatible and biodegradable dendri- Delivery Rev., 2005, 57, 2203.
mers based on simplified and optimized synthetic pathways 26 T. D. McCarthy, P. Karellas, S. A. Henderson, M. Giannis,
reducing cost of their production. D. F. O’Keefe, G. Heery, J. R. A. Paull, B. R. Matthews and
G. Holan, Mol. Pharmaceutics, 2005, 2, 312.
(e) Set-up appropriate in vivo studies to evaluate the
27 E. E. Simanek and S. O. Gonzalez, J. Chem. Educ., 2002, 79, 1222.
biocompatibility and the therapeutic profiles of drug–dendrimer 28 A.-M. Caminade and J.-P. Majoral, Acc. Chem. Res., 2004,
conjugates (PK/PD behaviors). 37, 341.
(f) Ensure the preparation of dendrimers in large quantities 29 K. A. Jacobson, Trends Pharmacol. Sci., 2010, 10, 575.
30 J. Ogier, T. Arnauld and E. Doris, Future Med. Chem., 2009,
at clinical-grade purity for clinical trials (under GMP conditions). 4, 693.
(g) Implementation of dendrimers technology for the 31 (a) C. M. Paleos, D. Tsiourvas, Z. Sideratou and L.-A. Tziveleka,
deployment of personalized medicine via the development of Expert Opin. Drug Delivery, 2010, 7, 1387; (b) M. X. Tang,
combined companion diagnostics/therapeutics. C. T. Redemann and F. C. Szoka Jr., Bioconjugate Chem., 1996,
7, 703.
All these aspects should bring about increase of number of 32 M. Hayder, M. Poupot, M. Baron, D. Nigon, C.-O. Turrin,
commercialized dendrimers based drug delivery systems A.-M. Caminade, J.-P. Majoral, R. A. Eisenberg, J.-J. Fournié,

This journal is c The Royal Society of Chemistry and the Centre National de la Recherche Scientifique 2012 New J. Chem., 2012, 36, 227–240 239
View Article Online

A. Cantagrel, R. Poupot and J.-L. Davignon, Sci. Transl. Med., 63 F. Alam, A. H. Soloway, R. F. Barth, N. Mafune, D. M. Adams
2011, 3, 81. and W. H. Knoth, J. Med. Chem., 1989, 32, 2326.
33 S. Svenson and D. A. Tomalia, Adv. Drug Delivery Rev., 2005, 64 R. F. Barth, D. M. Adams, A. H. Soloway, F. Alam and
57, 2106. M. V. Darby, Bioconjugate Chem., 1994, 5, 58.
34 A. R. Menjoge, R. M. Kannan and D. A. Tomalia, Drug Discovery 65 C. Wängler, S. Maschauer, O. Prante, M. Schäfer, R. Schirrmacher,
Today, 2010, 15, 171. P. Bartenstein, M. Eisenhut and B. Wängler, ChemBioChem, 2010,
35 C. Kojima, Expert Opin. Drug Delivery, 2010, 7, 307. 11, 2168.
36 W. Gao, J. M. Chan and O. C. Farokhzard, Mol. Pharmaceutics, 66 T. Darbre and J.-L. Reymond, Acc. Chem. Res., 2006, 39, 925.
2010, 7, 1913. 67 A. V. Klein and T. W. Hambley, Chem. Rev., 2009, 109, 4911.
37 R. X. Zhuo, B. Du and Z. R. Lu, J. Controlled Release, 1999, 68 E. Gabano, M. Ravera and D. Osella, Curr. Med. Chem., 2009,
57, 249. 16, 4544.
38 D. Wang, P. Kopeckova, T. Minko, V. Nanayakkara and 69 P. J. Pellechia, J. Gao, Y. Gu, H. J. Ploehn and C. J. Murphy,
J. Kopecek, Biomacromolecules, 2000, 1, 313. Inorg. Chem., 2004, 43, 1421.
Published on 08 September 2011. Downloaded by MIT Library on 2/10/2021 12:35:19 AM.

39 S. Zhu, M. Hong, L. Zhang, G. Tang, Y. Jiang and Y. Pei, Pharm. 70 N. Malik, E. G. Evavgoraou and R. Duncan, Anti-Cancer Drugs,
Res., 2010, 27, 161. 1999, 10, 767.
40 S. Gurdag, J. Khandare, S. Stapels, L. H. Matherly and 71 I. Haririan, M. S. Alavidjeh, M. R. Khorramizadeh, M. S. Ardestani,
R. M. Kannan, Bioconjugate Chem., 2006, 17, 275. Z. Z. Ghane and H. Namazi, Int. J. Nanomed., 2010, 20, 63.
41 M. Najlah, S. Freeman, D. Attwood and A. D’Emanuele, Biocon- 72 E. Bellis, L. Hajba, B. Kovacs, K. Sandor, L. Kollar and
jugate Chem., 2007, 18, 937. G. Kokotos, J. Biochem. Biophys. Methods, 2006, 69, 151.
42 T. P. Thomas, I. J. Majoros, A. Kotlyar, J. F. Kukowska-Latallo, 73 X. Zhao, S. C. J. Loo, P. P.-F. Lee, T. T. Y. Tan and C. K. Chu,
A. Bielinska, A. Myc and J. R. Baker Jr., J. Med. Chem., 2005, J. Inorg. Biochem., 2010, 104, 105.
48, 3729. 74 W.-D. Jang, N. Nishiyama, G.-D. Zhang, A. Harada, D.-L. Jiang,
43 I. J. Majoros, T. P. Thomas, C. B. Mehta and J. R. Baker Jr., S. Kawauchi, Y. Morimoto, M. Kikuchi, H. Koyama, T. Aida and
J. Med. Chem., 2005, 48, 5892. K. Kataoka, Angew. Chem., Int. Ed., 2005, 44, 419.
44 J. F. Kukowska-Latallo, K. A. Candido, Z. Cao, S. S. Nigavekar, 75 J. Kim, H.-J. Yoon, S. Kim, K. Wang, T. Ishii, Y.-R. Kim and
I. J. Majoros, T. P. Thomas, T. P. Balogh, M. K. Khan and W.-D. Jang, J. Mater. Chem., 2009, 19, 4627.
J. R. Baker Jr., Cancer Res., 2005, 65, 5317. 76 A. Quintana, E. Raczka, L. Piehler, I. Lee, A. Myc, I. Majoros,
45 Y. Zhang, T. P. Thomas, K.-H. Lee, M. Li, H. Zong, A. M. Desai, A. K. Patri, T. Thomas, J. Mulé and J. R. Baker Jr., Pharm. Res.,
A. Kotlyar, B. Huang, M. M. Banaszak Holl and J. R. Baker Jr., 2002, 19, 1310.
Bioorg. Med. Chem., 2011, 19, 2557. 77 C. C. Lee, E. R. Gillies, M. E. Fox, S. J. Guillaudeu, J. M. J.
46 L. V. Haandel and J. F. Stobaugh, Anal. Bioanal. Chem., 2010, Fréchet, E. E. Dy and F. C. Szoka, Proc. Natl. Acad. Sci. U. S. A.,
397, 1841. 2006, 103, 16649.
47 T. P. Thomas, S. K. Choi, M.-H. Li, A. Kotlyar and J. R. Baker 78 M. Najlah, S. Freeman, D. Attwood and A. D’Emanuele, Int. J.
Jr., Bioorg. Med. Chem. Lett., 2010, 20, 5191. Pharm., 2006, 308, 175.
48 I. J. Majoros, A. Myc, T. Thomas, C. B. Mehta and J. R. Baker Jr., 79 K. Kono, C. Kojima, N. Hayashi, E. Nishisaka, K. Kiura,
Biomacromolecules, 2006, 7, 572. S. Watarai and A. Harada, Biomaterials, 2008, 29, 1664.
49 X. Bi, X. Shi, I. J. Majoros, R. Shukla and J. R. Baker Jr., 80 J. Lim, A. Chouai, S.-T. Lo, W. Liu, X. Sun and E. E. Simanek,
J. Comput. Theor. Nanosci., 2007, 4, 1187. Bioconjugate Chem., 2009, 20, 2154.
50 S. Tang, L. J. Martinez, A. Sharma and M. Chai, Org. Lett., 2006, 81 Y. E. Kurtoglua, M. K. Mishraa, S. Kannanb and R. M. Kannan,
8, 4421. Int. J. Pharm., 2010, 384, 189.
51 N. Malik, R. Wiwattanapatapee, R. Klopsch, K. Lorenz, H. Frey, 82 R. S. Navath, A. R. Menjoge, B. Wang, R. Romero, S. Kannan
J. W. Weener, E. W. Meijer, W. Paulus and R. Duncan, and R. M. Kannan, Biomacromolecules, 2010, 11, 1544.
J. Controlled Release, 2000, 65, 133. 83 D. A. Patel, J. E. Henry and T. A. Good, Biochim. Biophys. Acta,
52 H. R. Ihre, O. L. De Jesus, F. C. Szoka and J. M. J. Fréchet, Gen. Subj., 2006, 1760, 1802.
Bioconjugate Chem., 2002, 13, 443. 84 D. A. Patel, J. E. Henry and T. A. Good, Brain Res., 2007,
53 S. Jain, A. Kaur, R. Puri, P. Utreja, A. Jain, M. Bhide, R. Ratnam, 1161, 95.
V. Singh, A. S. Patil, N. Jayaraman, G. Kaushik, S. Yadav and 85 Y. Y. Jiang, G.-T. Tang, L.-H. Zhang, S.-Y. Kong, S.-J. Zhu and
K. L. Khanduja, Eur. J. Med. Chem., 2010, 45, 4997. Y.-Y. Pei, J. Drug Targeting, 2010, 18, 389.
54 J. Lim and E. E. Simanek, Org. Lett., 2008, 10, 201. 86 Y. Cheng and T. Xu, Eur. J. Med. Chem., 2008, 43, 2291.
55 O. Perumal, J. Khandare, P. Kolhe, S. Kannan, M. Lieh-Lai and 87 S. N. Patel, P. Sethi, K. Ansari, D. Tiwari, A. Sharma and
R. M. Kannan, Bioconjugate Chem., 2009, 20, 842. A. K. Singhai, Int. J. Pharm. Life Sci., 2010, 1, 382.
56 R. Inapagollaa, B. Raja Gurua, Y. E. Kurtoglua, X. Gaob, 88 R. Haag and F. Kratz, Angew. Chem., Int. Ed., 2006, 45, 1198.
M. Lieh-Laic, D. J. P. Bassett and R. M. Kannan, Int. J. Pharm., 89 R. Satchi-Fainaro, R. Duncan and C. M. Barnes, Adv. Polym. Sci.,
2010, 399, 140. 2006, 193, 1.
57 Y. Zhang, T. P. Thomas, A. Desai, H. Zong, P. R. Leroueil, 90 Y. Deguchi, A. Kurihara and W. M. Pardridge, Bioconjugate
I. J. Majoros and J. R. Baker Jr., Bioconjugate Chem., 2010, Chem., 1999, 10, 32.
21, 489. 91 E. V. Batrakova, T. Y. Dorodnych, E. Y. Klinkii,
58 B. Wang, R. S. Navath, R. Romero, S. Kannan and R. Kannan, E. N. Kliushnenkova, O. B. Shemchukova, O. N. Goncharova,
Int. J. Pharm., 2009, 377, 159. S. A. Arjakov, V. Y. Alakhov and A. V. Kabanov, Br. J. Cancer,
59 R. S. Navath, Y. E. Kurtoglu, B. Wang, S. Kannan, R. Romero 1996, 74, 1545.
and R. M. Kannan, Bioconjugate Chem., 2008, 19, 2446. 92 T. Nakanishi, S. Fukushima, K. Okamoto, M. Suzuki,
60 H. R. Ihre, O. L. Padilla De Jesus, F. C. Szoka Jr. and J. M. J. Y. Matsumura, M. Yokayama, T. Okano, Y. Sakarai and
Fréchet, Bioconjugate Chem., 2002, 13, 443. K. Kataoka, J. Controlled Release, 2001, 74, 295.
61 W. C. Floyd, G. K. Datta, S. Imamura, H. M. Kieler-Ferguson, 93 S. Hong, P. R. Leroueil, I. J. Majoros, B. G. Orr, J. R. Baker and
K. Jerger, A. W. Patterson, M. E. Fox, F. C. Szoka, J. M. J. M. M. B. Holl, Chem. Biol., 2007, 14, 107.
Fréchet and J. A. Ellman, ChemMedChem, 2011, 6, 49. 94 M. Najlah and A. D’Emanuele, Curr. Opin. Drug Discovery Dev.,
62 R. Wiwattanapatapee, L. Lomlim and K. Saramunee, J. Controlled 2007, 10, 756.
Release, 2003, 88, 1. 95 S. Svenson, Eur. J. Pharm. Biopharm., 2009, 71, 445.

240 New J. Chem., 2012, 36, 227–240 This journal is c The Royal Society of Chemistry and the Centre National de la Recherche Scientifique 2012

You might also like