Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

PERSPECTIVE

Challenges for Metals in Medicine:


How Nanotechnology May Help
To Shape the Future
Nicolas P. E. Barry* and Peter J. Sadler*

Department of Chemistry, University of Warwick, Coventry CV4 7AL, U.K.

ABSTRACT Encapsulation of the platinum(IV) prodrug mitaplatin in block


copolymer nanoparticles increases drug circulation time in the blood and reduces
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.
Downloaded via UNIV FED DO RIO GRANDE DO SUL on September 3, 2021 at 20:45:50 (UTC).

accumulation in the kidneys, as reported by Lippard and colleagues in this issue of


ACS Nano. Importantly, controlled drug release from the nanoparticles produces
long-term anticancer efficacy, with the prospect of reduced side effects. We
highlight the potential that such a strategy holds for the future development of
metallodrugs. Metal coordination complexes offer the prospect of novel mecha-
nisms of activity on account of their unique architectures, as well as potential
activation mechanisms, including ligand substitution and metal- and ligand-centered redox properties. Nanoparticles offer exciting prospects for improving
delivery, cell uptake, and targeting of metallodrugs, especially anticancer drugs, to make them more effective and safer.

I
norganic medicinal chemistry is in the anticancer drugs (used as a component of
early days of its development, although nearly 50% of all cancer treatments). To
there are now a significant number of date, three platinum(II) compounds, cispla-
clinical trials involving metal compounds or tin, carboplatin, and oxaliplatin, have been
other agents that interfere with metabolic approved by the FDA (FDA approvals in
pathways for metals, both for therapy and 1978, 1989, and 2002, respectively, Chart 1).
for diagnosis.1 There is an urgent need for Nonetheless, side effects associated with
the discovery of drugs with novel mecha- these complexes and the development of
nisms of action, particularly because some tumor resistance have led to the search for
diseases and conditions develop resistances new generations of platinum-based antic-
to current drugs. Metal coordination com- ancer agents.2
plexes offer biological and chemical diver- Octahedral low-spin 5d6 PtIV complexes
sity that is distinct from that of organic are known to be relatively inert toward
drugs. This diversity arises not only from ligand substitution but can be activated
the choice of the metal itself and its oxida- chemically by reduction. Hence, they have
tion state, but also from the types and potential advantages as anticancer pro-
numbers of coordinated ligands and the drugs. They often have higher aqueous
coordination geometry of the complex. solubility than square-planar PtII complexes,
a feature exploited long ago by Tobe et al.
who synthesized iproplatin (CHIP, JM9, cis,
Metal coordination trans,cis-[PtCl2(OH)2(isopropylamine)2],
Chart 1).3 Iproplatin entered phase I and II
complexes offer biological clinical trials, and even phase III, but ulti-
and chemical diversity that is mately was found to be less active than
* Address correspondence to
cisplatin and so was not registered for clin- N.Barry@warwick.ac.uk,
distinct from that of organic ical use.4 Platinum(IV)-based drugs can re- P.J.sadler@warwick.ac.uk.

drugs. lease active PtII species as well as bioactive


ligands on chemical reduction in the envi-
Published online July 09, 2013
ronment of cancerous cells, for example by 10.1021/nn403220e
Interest in metallodrugs has been stimu- ascorbate or by a thiol such as glutathione.
lated by the recent success of platinum An interesting example of such a PtIV prodrug C 2013 American Chemical Society

BARRY AND SADLER VOL. 7 ’ NO. 7 ’ 5654–5659 ’ 2013 5654


www.acsnano.org
PERSPECTIVE
copolymer self-assembled into mi-
cellar NPs (see Chart 2 for molecular
structure of poly(D,L-lactide)), which
can entrap paclitaxel, an organic
drug used for the treatment of
various cancers, including lung,
ovarian, and breast. This micellar
paclitaxel formulation, named
Genexol-PM (Chart 2), has been
approved by the FDA for use in
patients with breast cancer. The co-
Chart 1. Molecular structures of three square-planar PtII anticancer drugs, two polymer increases the water solubi-
octahedral PtIV anticancer compounds, and the enzyme inhibitor dichloroacetate lity of paclitaxel and allows delivery
(a ligand in mitaplatin).
of higher doses than those achiev-
able with paclitaxel alone while
is mitaplatin, developed by Dhar well as by passive diffusion avoiding the use of adjuvants, which
and Lippard (Chart 1).5 Mitaplatin across the membrane. might have side effects. Another
releases two equivalents of dichloro- (iii) Targeting. The nanoparticle paclitaxel formulation, NK105, con-
acetate (DCA, Chart 1), an inhibitor might be designed so that it sisting of PEG and modified polyas-
of the enzyme pyruvate dehydro- has vectors on its surface partate (50% of the carboxylic groups
genase kinase, and one equivalent that can target specific cell of the polyaspartate block are ester-
of cisplatin upon reduction in can- receptors as well as have the ified with 4-phenyl-1-butanol, Chart 2)
cer cells. Dichloroacetate is an FDA capacity to encapsulate the is currently undergoing phase II clin-
orphan drug currently in clinical drug, thus reducing side ef- ical trials for treatment of advanced
phase II trials for head and neck fects and limiting attack to stomach cancer.10 Metallodrugs have
carcinoma treatment. target cells or organelles only. also been encapsulated into NPs. For
Nanotechnology, which has been (iv) Multidrug delivery and ther- instance, cisplatin has been formu-
defined as the engineering and anostics. More than one drug lated in micelles composed of PEG
manufacturing of materials at the might be encapsulated for and poly(γ-benzyl-L-glutamate) (PEG-
atomic and molecular scale,6 offers combination therapy. Diag- PGlu, Chart 2), a formulation named
unique tools for developing safer nostic aids (e.g., imaging tags) NC-6004 or Nanoplatin, which is un-
and more effective medicines (nano- and multiple therapeutic drugs der phase I/II clinical evaluation for
medicines), and provides several might also be incorporated the treatment of pancreatic cancer.
potential advantages for drug for- into a single nanoparticle. Other examples include micel-
mulation and delivery. Nanoparticles made of polymers lar metal-based magnetic reso-
(i) Control of drug solubility. (NPs) are of particular interest as nance imaging (MRI) contrast or
This might involve either in- drug delivery systems because of single-photon emission computed
creasing the aqueous solubility their synthetic versatility as well as tomography/X-ray computed tomog-
of highly lipophilic complexes their tunable properties (e.g., ther- raphy imaging agents.11,12
or decreasing the solubility of mosensitivity and pH-response). As Drug loading into NPs can be
complexes that might other- early as 1994, it was demonstrated achieved by three techniques: (i)
wise be rapidly excreted: a that nanospheres, synthesized from covalent attachment to the polymer
“slow-release” strategy that amphiphilic copolymers composed backbone, (ii) adsorption to the
may engender less toxicity of two biocompatible blocks (including polymer surface, or (iii) entrapment
and improve the therapeutic a polyethylene glycol(PEG)-block, in the polymer matrix during pre-
response compared to a burst Chart 2), exhibit dramatically in- paration of the NPs.9 In most cases
release. creased blood circulation times and (Table 1), metallodrugpolymer
(ii) Modulation of drug distribu- low liver accumulation in mice.7 This systems have been formulated
tion. The uptake of drugs discovery was followed by numer- by covalent attachment of the
encapsulated in nanoparti- ous reports of PEGylated NPs with metal-based drug to the polymer
cles is likely to depend on various architectures,8 and even- backbone (formulations usually
the shape, size, and surface tually led to PEG being listed as termed “polymermetal complex”
recognition of the nanopar- ''Generally Recognized as Safe'' or “polymerdrug conjugates”).
ticles by cells rather than on (GRAS) by the FDA, and to the clin- Although attractive because it
the characteristics of the drug. ical translation of a number of NPs.9 offers facile and reproducible drug
For example, cells can take up Among them, the biodegradable polymer conjugates with high metal-
particles by endocytosis as poly(ethylene glycol)-poly(D,L-lactide) lodrug loading percentages, this

BARRY AND SADLER VOL. 7 ’ NO. 7 ’ 5654–5659 ’ 2013 5655


www.acsnano.org
PERSPECTIVE
Chart 2. Molecular structures of some of the polymers discussed.

a
TABLE 1. Examples of Platinum and Ruthenium Anticancer Complexes Encapsulated into Polymer Nanoparticles (NPs)

block copolymer metal complex formulation outcome ref

PEG-PGlu nanoplatin: complex entrapped in NPs pancreatic cancer: phase I/II clinical trials 13
(sponsor Nanocarrier Co., Ltd.)
PEG-PDEM, PEG-PCL, cisplatin complex entrapped in NPs in vivo: better activity than cisplatin 14
PCL-PDEM
PEG-PAsp {(NH3)2-Pt}2þ polymermetal conjugation in vivo: minimal nephrotoxicity, higher accumulation 15
(dicarboxylated polymer(COO)2Pt in tumors than cisplatin, similar activity
chelation)
PLGA-PEG mitaplatin complex entrapped in NPs in vivo: slow-release of mitaplatin, longer circulation 16
time, similar tumor growth inhibition and less
accumulation in kidneys than mitaplatin
PEG-PLAMCC dinuclear Pt(II) polymermetal conjugation in vivo: longer blood circulation time and higher 17
complex (carboxylated polymerCOOPt) antitumor efficacy than oxaliplatin
{di-DACH-Pt2}4þ
PEG-PGlu polymermetal conjugation in vivo: longer plasma half-life than oxaliplatin in 18
(dicarboxylated polymer(COO)2Pt chelation) micelles; high specificity for tumor tissue; strong
{DACH-Pt}2þ antitumor activity in mice
amidomalonato ProLindac (AP5346): polymermetal head and neck cancer: randomized, pilot clinical trials 19
chelator-HPMA conjugation (amidomalonato chelation) (sponsor University of California, San Diego)
PhenISA {Ru(phen)2}2þ polymermetal conjugation (phenanthroline in vitro: cellular uptake by endocytic pathway 20
chelation)
a
Acronyms: PCL, polycaprolactone; PAsp, poly(aspartic acid); PMA, poly(methacrylicacid); PDEM, poly[2-(N,N-dimethylamino)ethyl methacrylate]; PLAMCC, poly(L-lactide-co-
2-methyl-2-carboxylpropylenecarbonate); PGLA, poly(D,L-lactic-co-glycolic acid); HPMA, hydroxypropylmethacrylamide; phen, 1,10-phenanthroline. See Chart 2 for molecular
structures. DACH = diaminocyclohexane.

BARRY AND SADLER VOL. 7 ’ NO. 7 ’ 5654–5659 ’ 2013 5656


www.acsnano.org
PERSPECTIVE
Scheme 1. Diagrammatic representation of the double emulsion method for encapsulating mitaplatin in PLGA-PEG
nanoparticles. PVA = poly(vinyl alcohol). Reproduced from ref 16. Copyright 2013 American Chemical Society.

method presents the drug as a pro- PLGA).16 Because the presence of of this approach using nanoparticle
drug which must then be released, axial dichloroacetate ligands is a treatment.
with inherent disadvantages: the key structural feature of mitaplatin, Future Challenges. We now high-
drug is structurally modified, which an unusual synthetic strategy was light three future challenges for
can impact its biological activity; the employed for its encapsulation in medicinal inorganic chemists that
release of the drug from the poly- the NPs. Instead of classical drug might be overcome using polymer
mer backbone needs to be achiev- loading methods, such as nanopre- NPs.
able under physiological conditions; cipitation or conjugation, a water/ Targeting Specific Classes of Cells
upon cleavage, the drug must retain oil/water double emulsion path- or Specific Cell Components. To date,
its therapeutic properties; poisoning way was employed (Scheme 1). both clinically validated therapeutic
and contamination by catalysts or The encapsulation process was and imaging NPs usually target can-
other reagents must be avoided dur- optimized to increase platinum cer cells in a passive way. Such a
ing the drugpolymer-conjugation loading and to minimize particle passive drug targeting and delivery
synthesis, which also underlies the diameter. Both mitaplatin-loaded strategy is achieved by taking advan-
difficulties surrounding the character- and mitaplatin-free NPs were tage of the enhanced permeability
ization of covalently modified NPs. synthesized. and retention (EPR) effect in tumor
Such systems will not be described In vivo studies on these NPs tissues.21 Tumor vasculature is highly
in detail in this Perspective; instead, showed that the encapsulation of disorganized as compared to the
we focus on the entrapment of intact mitaplatin prolongs retention of vasculature in normal tissues, and
metallodrugs through noncovalent platinum in the bloodstream as the vascular endothelium in tumors
hydrophobic or polar interactions, compared to administration of mi- proliferates rapidly and discontinu-
that is, without grafting of the taplatin alone, which correlates with ously. This results in a higher number
metal-based drug onto the polymer in vitro release studies. The tumor of fenestrations and open junctions
chains. This method allows the re- burden of mice carrying MDA-MB- (from 200 nm to 1.2 μm) than in
tention of the structural integrity of 468 triple-negative breast cancer normal vessels. Therefore, particles
the loaded drug (ligands, geometry, xenografts was also reduced by with a typical size of a few hundred
metal oxidation state, and stereo- both mitaplatin alone and mitaplatin- nanometers can passively cross the
chemistry), which is of the utmost loaded-NPs (no effect for mitaplatin- tumor endothelial barrier through
importance for metal-based drug free-NPs), and the biodistribution of fenestrations, and accumulate at
candidates. the nanodelivery platinum is signif- particular sites through blood hemo-
In this issue of ACS Nano, Lippard icantly shifted from accumulation in dynamic forces and diffusion mech-
and colleagues report the encapsu- the kidneys to the liver. Increasing anisms.22 This leads to the passive
lation of the octahedral platinum(IV) the circulation time of platinum in targeting of cancer cells. The EPR
compound mitaplatin in a poly(D,L- the bloodstream, reducing accumu- effect may be used to enhance the
lactic-co-glycolic acid)-block-poly- lation in the kidneys, and the con- uptake of specific drugs into cancer
(ethylene glycol) (PLGA-PEG) NP (see trolled release of the drug over time, cells with high selectivity as com-
Chart 2 for molecular structure of are all key potential advantages pared to normal cells. Nevertheless,

BARRY AND SADLER VOL. 7 ’ NO. 7 ’ 5654–5659 ’ 2013 5657


www.acsnano.org
PERSPECTIVE
the delivery of the active pharmaceu- inorganic drugs is worthy of further of intact metal complexes directly
tical ingredient (API) into specific in- exploration and exploitation in the into cells by nanoparticles (as distinct
tracellular sites in cancer cells requires near future. from metalpolymer conjugates)
active targeting. Actively targeted provides a means of avoiding changes
NPs may be internalized through to the speciation of metal complexes
One of the
clathrin-dependent endocytosis before they enter cells, while prevent-
pathways, caveolin-assistance, cell- achievements reported ing nonspecific binding of NP surfaces
adhesion-molecule directed, or lipid- to blood components and avoiding
raft-associated mechanisms, leading
in the article by Lippard clearance by the mononuclear phago-
to endosome formation, which ulti- and colleagues is the cyte system (MPS), or by the reticu-
mately leads to lysosomes.23 loendothelial system (RES), as distinct
demonstration that NP
Control of the API release from from the behavior of liposomes.
NPs depends on the hydrophilicity encapsulation can At the subcellular level, there is
of the API. Hydrophobic small mol- currently a lack of techniques power-
reduce unwanted
ecules may be released within the ful enough for studying the specia-
endosome, resulting in permeation accumulation of tion of metal complexes directly in
within the intracellular components, cells. Nanotechnology tools, if ap-
while release of bioactive macromo-
platinum from the plied for specifically delivering in-
lecules such as nucleic acids (DNA, platinum(IV) prodrug tact metallodrugs to subcellular
siRNA, miRNA) or charged hydrophi- organelles, might help to simplify
mitaplatin in the
lic small molecules requires first the this problem. Indeed, advances in
escape of the NPs from the endo- kidneys. metal analysis and especially spe-
some, and then fusing with lyso- ciation techniques are expected
somes in order to reach the desired within a few years, and the restric-
subcellular organelle. Numerous re- Speciation of Metal Complexes in tion of localization of metal com-
ports comparing targeted and non- Cells. Understanding the mechan- plexes in specific organelles would
targeted NPs have confirmed that the ism of the action of metallodrugs, help such speciation analysis. This
primary role of the targeting ligand is not only to optimize activity but work, spanning chemistry, biology,
to enhance cellular uptake into target also to reduce side effects, requires and physics, might also be fruitfully
cells. The encapsulation of metallo- knowledge of the speciation of metal- applied to the identification of tar-
drugs in nanoparticles thus provides based drugs, both en route to the get sites and understanding of me-
a degree of protection to the metal- tumor cells and in cells. This is a tabolic pathways.
based drug from reductants and nu- difficult task;metal complexes in In conclusion, this Perspective is
cleophiles by physically preventing general are susceptible to ligand based on results reported in this
interaction between these agents exchange reactions on a wide ranging issue of ACS Nano by Lippard and
and the encapsulated metal complex. time scale (years to nanoseconds, or colleagues, highlighting the role
Reducing the Side Effects of even less if photoexcitation is used), that polymer nanoparticles can play
Drugs. Nanotechnology tools also and to metal- and ligand-centered in the slow release of platinum com-
open up new prospects for the con- redox reactions. The choice made pounds into cancer cells, shift of
trol of the biodistribution of metal- by Lippard and colleagues16 of a biodistribution of platinum from
lodrugs. One of the achievements platinum(IV) complex instead of a kidneys to liver, increase of circula-
reported in the article by Lippard platinum(II) is based on the fact that tion time in blood, and retention of
and colleagues16 is the demonstra- PtIV complexes with their low-spin the anticancer activity of the intact
tion that NP encapsulation can re- 5d6 outer shell electronic configura- encapsulated metal complex.16 It is,
duce unwanted accumulation of tion are more inert to substitution however, clear that inorganic com-
platinum from the platinum(IV) reactions than square-planar PtII pounds offer different mechanisms
prodrug mitaplatin in the kidneys. complexes. Hence, they are likely of drug action depending on the
Unencapsulated mitaplatin displays to undergo fewer side reactions en metal(s) used, their structures, and
a greater acute effect, but the long- route to the tumor, although once their redox properties. Thus, they
term tumor growth inhibition rea- they arrive they need to undergo can be utilized for the design of
lized by both treatments is the reduction to PtII (e.g., by intracellular novel drugs in the treatment of a
same. Other reports in the recent glutathione) to be active. In this broad range of diseases.1 For in-
literature also show that metallo- case, there is dual activity arising from stance, lithium salts, used clinically
drugs loaded in NPs do not cause the release of the axial dichloroace- for stabilization of bipolar disorders,
as much damage as the drugs on tate ligands (which can stimulate mi- might take advantage of slow re-
their own.24 This advantageous nano- tochondrial function) and from attack lease by entrapment in polymer
particle formulation strategy for on DNA by the PtII product. Delivery particles. The shift of biodistribution

BARRY AND SADLER VOL. 7 ’ NO. 7 ’ 5654–5659 ’ 2013 5658


www.acsnano.org
PERSPECTIVE
away from kidneys might be of inter- Orphan Drug Dichloroacetate. Proc. Polymer Therapeutic. Adv. Drug Deliv-
est for a variety of gold complexes Natl. Acad. Sci. U.S.A. 2009, 106, ery Rev. 2009, 61, 1214–1219.
22199–22204. 20. Maggioni, D.; Fenili, F.; D'Alfonso, L.;
used for the treatment of rheumatoid 6. Farokhzad, O. C.; Langer, R. Impact Donghi, D.; Panigati, M.; Zanoni, I.;
arthritis. The safety enhancement ob- of Nanotechnology on Drug Deliv- Marzi, R.; Manfredi, A.; Ferruti, P.;
tainable by passive and active target- ery. ACS Nano 2009, 3, 16–20. D'Alfonso, G.; et al. Luminescent Rhe-
7. Gref, R.; Minamitake, Y.; Peracchia, nium and Ruthenium Complexes of an
ing of metallodrugs encapsulated M. T.; Trubetskoy, V.; Torchilin, V.; Amphoteric Poly(amidoamine) Func-
into NPs is of potential interest for Langer, R. Biodegradable Long- tionalized with 1,10-Phenanthroline.
a range of metal-based drug candi- Circulating Polymeric Nanospheres. Inorg. Chem. 2012, 51, 12776–12788.
Science 1994, 263, 1600–1603. 21. Konno, T.; Maeda, H.; Iwai, K.; Tashiro,
dates, while a high metal-complex 8. Jokerst, J. V.; Lobovkina, T.; Zare, S.; Maki, S.; Morinaga, T.; Mochinaga,
loading might be an advantage for R. N.; Gambhir, S. S. Nanoparticle M.; Hiraoka, T.; Yokoyama, I. Effect
the delivery of gadolinium MRI con- PEGylation for Imaging and Therapy. of Arterial Administration of High-
Nanomedicine 2011, 6, 715–728. Molecular-Weight Anticancer Agent
trast agents. 9. Kamaly, N.; Xiao, Z.; Valencia, P. M.; SMANCS with Lipid Lymphographic
Furthermore, bioinspired, bioen- Radovic-Moreno, A. F.; Farokhzad, Agent on Hepatoma: A Preliminary
gineered, and biomimetic NPs with O. C. Targeted Polymeric Therapeu- Report. Eur. J. Cancer Clin. Oncol.
tic Nanoparticles: Design, Develop- 1983, 19, 1053–1065.
shapes other than spherical micelles25 ment and Clinical Translation. Chem. 22. Wang, X.; Guo, Z. Targeting and
as well as the understanding of release Soc. Rev. 2012, 41, 2971–3010. Delivery of Platinum-Based Antican-
mechanisms and metabolism are now 10. Matsumura, Y. Poly (Amino Acid) cer Drugs. Chem. Soc. Rev. 2013, 42,
Micelle Nanocarriers in Preclinical 202–224.
beginning to be explored. Such under- 23. Bareford, L. M.; Swaan, P. W. Endo-
and Clinical Studies. Adv. Drug De-
standing should enable the design of livery Rev. 2008, 60, 899–914. cytic Mechanisms for Targeted Drug
new NPs with optimized blood circu- 11. Haxton, K. J.; Burt, H. M. Polymeric Delivery. Adv. Drug Delivery Rev.
Drug Delivery of Platinum-Based 2007, 59, 748–758.
lation times, able to target specific 24. Butler, J. S.; Sadler, P. J. Targeted
Anticancer Agents. J. Pharm. Sci.
cells and organelles. The diversity and 2009, 98, 2299–2316. Delivery of Platinum-Based Antican-
versatility of metallodrugs along with 12. Hoang, B.; Lee, H.; Reilly, R. M.; Allen, cer Complexes. Cur. Opin. Chem.
C. Noninvasive Monitoring of the Biol. 2013, 17, 175–188.
the technological progress made in 25. Geng, Y.; Dalhaimer, P.; Cai, S.; Tsai,
Fate of 111In-Labeled Block Copoly-
the design of polymer nanocarriers mer Micelles by High Resolution and R.; Tewari, M.; Minko, T.; Discher, D. E.
may provide opportunities for tack- High Sensitivity MicroSPECT/CT Imag- Shape Effects of Filaments versus
ing. Mol. Pharm. 2009, 6, 581–592. Spherical Particles in Flow and Drug
ling medical challenges in the near Delivery. Nat. Nanotechnol. 2007, 2,
13. Plummer, R.; Wilson, R. H.; Calvert, H.;
future. Boddy, A. V.; Griffin, M.; Sludden, J.; 249–255.
Conflict of Interest: The authors de- Tilby, M. J.; Eatock, M.; Pearson, D. G.;
clare no competing financial interest. Ottley, C. J.; et al. A Phase I Clinical
Study of Cisplatin-Incorporated Poly-
Acknowledgment. We thank the Swiss meric Micelles (NC-6004) in Patients
National Science Foundation (Grant No. with Solid Tumors. Br. J. Cancer 2011,
PA00P2-145308 to NPEB), the ERC (Grant 104, 593–598.
No. 247450 to P.J.S.), EPSRC (Grant No. 14. Xu, P.; VanKirk, E. A.; Murdoch, W. J.;
EP/F034210/1) and EC COST Action CM1105 Zhan, Y.; Isaak, D. D.; Radosz, M.;
for support. Shen, Y. Anticancer Efficacies of
Cisplatin-Releasing pH-Responsive
Nanoparticles. Biomacromolecules
REFERENCES AND NOTES 2006, 7, 829–835.
1. Barry, N. P. E.; Sadler, P. J. Exploration 15. Yokoyama, M.; Okano, T.; Sakurai, Y.;
of the Medical Periodic Table: To- Suwa, S.; Kataoka, K. Development
wards New Targets. Chem. Commun. of the Polymer Micelle Carrier Sys-
2013, 49, 5106–5131. tem for Doxorubicin. J. Controlled
2. Graf, N.; Lippard, S. J. Redox Activa- Release 1996, 39, 351–356.
tion of Metal-Based Prodrugs as a 16. Johnstone, T. C.; Kulak, N.; Pridgen,
Strategy for Drug Delivery. Adv. Drug E. M.; Farokhzad, O. C.; Langer, R.;
Delivery Rev. 2012, 64, 993–1004. Lippard, S. J. Nanoparticle Encapsu-
3. Braddock, P. D.; Connors, T. A.; Jones, lation of Mitaplatin and the Effect
M.; Khokhar, A. R.; Melzack, D. H.; Thereof on in Vivo Properties. ACS
Tobe, M. L. Structure and Activity Nano 2013, 10.1021/nn401905g.
Relations of Platinum Complexes 17. Wang, R.; Hu, X.; Xiao, H.; Xie, Z.;
with Antitumor Activity. Chem. Biol. Huang, Y.; Jing, X. Polymeric Dinuc-
Interact. 1975, 11, 145–161. lear Platinum(II) Complex Micelles
4. Bonomi, P. D.; Finkelstein, D. M.; for Enhanced Antitumor Activity.
Ruckdeschel, J. C.; Blum, R. H.; Green, J. Mater. Chem. B 2013, 1, 744–748.
M. D.; Mason, B.; Hahn, R.; Tormey, 18. Cabral, H.; Nishiyama, N.; Okazaki, S.;
D. C.; Harris, J.; Comis, R. Combina- Koyama, H.; Kataoka, K. Preparation
tion Chemotherapy Versus Single and Biological Properties of Dichloro-
Agents Followed by Combination (1,2-diaminocyclohexane)platinum(II)
Chemotherapy in Stage IV Non-Small- (DACHPt)-Loaded Polymeric Micelles.
Cell Lung Cancer: A Study of the J. Controlled Release 2005, 101, 223–
Eastern Cooperative Oncology Group. 232.
J. Clin. Oncol. 1989, 7, 1602–1613. 19. Nowotnik, D. P.; Cvitkovic, E. ProL-
5. Dhar, S.; Lippard, S. J. Mitaplatin, a indac (AP5346): A Review of the Devel-
Potent Fusion of Cisplatin and the opment of an HPMA DACH Platinum

BARRY AND SADLER VOL. 7 ’ NO. 7 ’ 5654–5659 ’ 2013 5659


www.acsnano.org

You might also like