Stres Oxidativ

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Advanced Drug Delivery Reviews 61 (2009) 290–302

Contents lists available at ScienceDirect

Advanced Drug Delivery Reviews


j o u r n a l h o m e p a g e : w w w. e l s ev i e r. c o m / l o c a t e / a d d r

Therapeutic strategies by modulating oxygen stress in cancer and inflammation☆


Jun Fang a,⁎, Takahiro Seki b, Hiroshi Maeda a,⁎
a
Department of Microbiology & Oncology, Faculty of Pharmaceutical Sciences, Sojo University, Ikeda 4-22-1, Kumamoto 860-0082, Japan
b
Innovative Collaboration Organization, Kumamoto University, Kumamoto, Japan

a r t i c l e i n f o a b s t r a c t

Article history: Oxygen is the essential molecule for all aerobic organisms, and plays predominant role in ATP generation,
Received 14 November 2008 namely, oxidative phosphorylation. During this process, reactive oxygen species (ROS) including superoxide
Accepted 13 February 2009 anion (O•− 2 ) and hydrogen peroxide (H2O2) are produced as by-products, while it seems indispensable for
Available online 26 February 2009
signal transduction pathways that regulate cell growth and reduction–oxidation (redox) status. However,
during times of environmental stress ROS levels may increase dramatically, resulting in significant damage to
Keywords:
Reactive oxygen species
cell structure and functions. This cumulated situation of ROS is known as oxidative stress, which may,
Cancer however, be utilized for eradicating cancer cells.
Oxidative stress It is well known that oxidative stress, namely over-production of ROS, involves in the initiation and progression
Inflammation of many diseases and disorders, including cardiovascular diseases, inflammation, ischemia–reperfusion (I/R)
EPR effect injury, viral pathogenesis, drug-induced tissue injury, hypertension, formation of drug resistant mutant, etc.
Tumor-targeting Thus, it is reasonable to counter balance of ROS and to treat such ROS-related diseases by inhibiting ROS
Macromolecular therapeutics production. Such therapeutic strategies are described in this article, that includes polymeric superoxide
Oxidation therapy
dismutase (SOD) (e.g., pyran copolymer-SOD), xanthine oxidase (XO) inhibitor as we developed water soluble
form of 4-amino-6-hydroxypyrazolo[3,4-d]pyrimidine (AHPP), heme oxygenase-1 (HO-1) inducers (e.g.,
hemin and its polymeric form), and other antioxidants or radical scavengers (e.g., canolol).
On the contrary, because of its highly cytotoxic nature, ROS can also be used to kill cancer cells if one can
modulate its generation selectively in cancer. To achieve this goal, a unique therapeutic strategy was developed
named as “oxidation therapy”, by delivering cytotoxic ROS directly to the solid tumor, or alternatively inhibiting
the antioxidative enzyme system, such as HO-1 in tumor. This anticancer strategy was examined by use of O•− 2
or H2O2-generating enzymes (i.e., XO and D-amino acid oxidase [DAO] respectively), and by discovering the
inhibitor of HO-1 (i.e., zinc protoporphyrin [ZnPP] and its polymeric derivatives). Further for the objective of
tumor targeting and thus reducing side effects, polymer conjugates or micellar drugs were prepared by use of
poly(ethylene glycol) (PEG) or styrene maleic acid copolymer (SMA), which utilize EPR (enhanced
permeability and retention) effect for tumor-selective delivery. These macromolecular drugs further showed
superior pharmacokinetics including much longer in vivo half-life, particularly tumor targeted accumulation,
and thus remarkable antitumor effects.
The present review concerns primarily our own works, in the direction of “Controlling oxidative stress:
Therapeutic and delivery strategy” of this volume.
© 2009 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 291
1.1. ROS and cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 291
1.1.1. Endogenous defense system against ROS in cancer cells as a unique anticancer target . . . . . . . . . . . . . . . . . . . . 291
1.1.2. Another side of ROS in cancer. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 292
1.2. Other diseases related with ROS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 293
1.3. Concerns in ROS-mediated anticancer therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 293

☆ This review is part of the Advanced Drug Delivery Reviews theme issue on “Controlling Oxidative Stress: Therapeutic and Delivery Strategies”.
⁎ Corresponding authors. Fang is to be contacted at Tel.: +81 96 326 4137; fax: +81 96 326 4114. Maeda, Tel.: +81 96 3226 4114; fax: +81 96 326 4114.
E-mail addresses: fangjun@ph.sojo-u.ac.jp (J. Fang), hirmaeda@ph.sojo-u.ac.jp (H. Maeda).

0169-409X/$ – see front matter © 2009 Elsevier B.V. All rights reserved.
doi:10.1016/j.addr.2009.02.005
J. Fang et al. / Advanced Drug Delivery Reviews 61 (2009) 290–302 291

2. Therapeutic strategies by inhibiting ROS generation: the use of antioxidative agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . 293
2.1. Xanthine oxidase inhibitors and SOD . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 293
2.1.1. Pyrazolopyrimidine derivatives (AHPP): inhibitors of XO . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 293
2.1.2. SOD and its polymer conjugate. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 294
2.2. Heme oxygenase (HO). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 295
2.3. NO . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 296
2.4. Canolol, a ROS scavenging phenols from plant. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 296
3. Oxystress inducing anticancer therapy: oxidation therapy. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 297
3.1. Poly(ethylene glycol) conjugated xanthine oxidase (PEG–XO) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 297
3.2. Poly(ethylene glycol) conjugated D-amino acid oxidase (PEG–DAO) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 297
3.3. HO inhibitor: zinc protoporphyrin IX (ZnPP) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 298
3.3.1. Polymer conjugated ZnPP (PEG–ZnPP) and SMA–micelle of ZnPP (SMA–ZnPP) . . . . . . . . . . . . . . . . . . . . . . . 299
3.3.2. PEG–ZnPP/SMA–ZnPP for photodynamic therapy (PDT) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 299
3.3.3. Other aspects of ZnPP and its polymer derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 299
3.3.4. New insight into the mechanisms of PEG–ZnPP/SMA–ZnPP dependent cytotoxicity . . . . . . . . . . . . . . . . . . . . . 299
3.4. Caution for oxidation therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 299
4. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 299
Acknowledgement. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 300
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 300

1. Introduction ROS from aerobic respiration or ambient oxygen-derived ROS, thus


reduce the potential insults of ROS. When an antioxidative enzyme
Reactive oxygen species (ROS) is a collective term of oxygen- defective mutant (e.g. SOD (−)) is generated, the growth of the bacteria
derived species, including not only the oxygen radicals (superoxide in aerobic condition becomes suppressed. However, its growth in
anion radical O•− •
2 , hydroxyl radicals OH etc.) but also some non-radical anaerobic condition is normal [7]. So-called facultative bacteria can
derivatives of O2 (hydrogen peroxide H2O2, singlet oxygen, alkyl adjust their metabolism depending on oxygen pressure. Similar
peroxide etc.). They are generally very small molecules and are highly phenomenon could be found in cancer cells. Cancer cells, particularly
reactive due to the presence of unpaired valence shell electrons. ROS is in the center area of the tumor nodule or mass, where ambient oxygen
potentially hazardous and it can be a by-product of cellular pressure is low, i.e. hypoxic conditions, they adapt more like anaerobic
metabolism. It also involves cell development, growth, survival, cell bacteria, having low levels of mitochondrial oxidative phosphorylation.
killing, aging, drug metabolism, pathogenesis of viral infection and This has been well known as Warburg-effect for a long time [8]. Under
development of cancer [1,2]. During the production of ROS, molecular such hypoxic conditions, they produce ATP mainly by glycolysis, or even
oxygen usually acts as electron acceptors to become oxygen free fermentation of amino acids. Recently, it has also been reported that
radicals. In aerobic life, where molecular oxygen is ubiquitous, ROS acetyl-CoA synthetase played important roles in producing ATP for
becomes the primary mediators of free radical reactions in cells, which
are generated during the production of ATP by aerobic metabolism in
mitochondria. The leakage of electrons from mitochondria, during the
electron-transport steps of ATP production, generates ROS, e.g. O•− •
2 . O2

is converted by superoxide dismutase (SOD) to generate H2O2, from
which further •OH is generated in a reaction catalyzed by Fe2+ or Cu2+
ions. In addition to these ROS, oxides of nitrogen (•NO, •NO2) are also
free radicals, and they are frequently called reactive nitrogen species
(RNS), and play important roles in biology similar to ROS.
ROS is a group of highly reactive molecules, which can react quickly
and damage various types of biomolecules, including proteins [3], DNA
[4] and lipids [5]. Under physiological conditions, to overcome the
potential toxicity of ROS, cells have evolved a series of antioxidative
defense systems to counteract these highly dangerous and extremely
reactive insults. These defense systems include intracellular SOD,
catalase and glutathione peroxidase that eliminate O•− 2 and H2O2.
There are also other enzymes or compounds that contribute to
scavenging free radicals (e.g., heme oxygenase-1 (HO-1), ascorbate,
tocopherol and glutathione) (Fig. 1), by which the cellular insults
caused by ROS are reduced to a nontoxic level [6].
Fig. 1. Reactive oxygen species (ROS) and reactive nitrogen species (RNS) metabolism
and the antioxidant defense system in aerobic organisms. Oxidative free radicals, such
1.1. ROS and cancer as superoxide (O•− •
2 ), hydroxyl radicals ( OH), hydrogen peroxide (H2O2), nitric oxide
(NO), are generated during metabolism of cells, whereas, antioxidative defenses
1.1.1. Endogenous defense system against ROS in cancer cells as a unique including superoxide dismutase (SOD), heme oxygenase-1 (HO-1), catalase, glu-
tathione peroxidase, thioredoxin and various antioxidants (e.g., glutathione, Vitamin C,
anticancer target
E, canolol) serve as scavengers of these ROS and RNS. The balance between ROS (RNS)
As discussed above, the balance of ROS formation and antioxidative generation and antioxidative defenses is important for the versatile behaviors of cells.
defense level is crucial for cell survival and growth, and it is very Under physiological conditions, ROS is well controlled to tolerable levels, which will not
important for the cell to remove ROS properly for it to remain viable and harm the cells but it is rather involved in the process of cell development and growth.
maintain its vital function. Namely, normal cellular function will be On the contrary, imbalance of the ROS generating vs defending system, either by
overproduction of ROS or by deficits in antioxidative systems, will increase the level of
altered depending on this balance, which will in turn affect the fate of ROS, leading to cellular damages because of its highly toxic nature, which is the cause of
the cell. For example, most aerobic bacteria have adequate antioxidative many diseases. ONOO−, peroxynitrite; NOS, nitric oxide synthase; Arg, arginine. NAC,
systems, including SOD and catalase, which can eliminate hazardous N-acetylcysteine. Reproduced from Ref. [124] (modified) with permission.
292 J. Fang et al. / Advanced Drug Delivery Reviews 61 (2009) 290–302

tumor cells [9]. Namely, generation of ATP in the hypoxic tumor cells is parallel manner is also known in the same time course either by
not an oxygen-dependent phenomenon. Parallel with these facts, it is xanthine oxidase (XO) activation or NADPH oxidation to NO
very intriguing and more important that cancer cells are more generation [25–28]. NO and O•− 2 , react immediately and more reactive
frequently deficient in most crucial antioxidative enzymes, such as derivative, ONOO− is generated, which plays probably more impor-
catalase, glutathione peroxidase and SOD [10–13]. This means a high tant role in carcinogenesis [29–32], and also metastasis by activating
vulnerability of tumor cells to ROS will be observed [14,15]. In fact, many matrix metalloproteinases (MMPs) [33,34]. We further reported the
conventional anticancer drugs including vinblastine, doxorubicin, increased viral and bacterial mutation under such condition of
camptothecin, cisplatin and inostamycin, exhibit antitumor activity via extensive ONOO− generation [35–37].
ROS generation if not totally [16]. Accordingly, a unique antitumor Moreover, oxidative stress can stimulate the expansion of mutated
strategy named “oxidation therapy” was developed by delivering excess cell clones by modulating genes related to proliferation and triggering
oxidative stress into tumor cells or targetedly disrupting the antiox- redox-responsive signaling cascades such as epidermal growth factor
idative defense systems of tumor cells [17–20]. (EGF), tyrosine phosphorylation, protein kinase C (PKC) [38], and
transcription factors that regulate inflammation and apoptosis
1.1.2. Another side of ROS in cancer including NF-κB, Activator protein 1 (AP-1) and NF-E2-related factor
With regard to ROS and cancer, however, it should be noted that (Nrf2) [39–41]. In addition, apoptosis signal-regulating kinase 1
the biological effects of ROS in cancer are multiple and non-linear. (ASK1), which is a member of the mitogen-activated protein kinase
Namely high levels of oxidative stress exhibit cytotoxicity as kinase kinase (MAPKKK) family, plays crucial roles in ROS (e.g., H2O2)-
mentioned above, inhibiting cell proliferation and leading to apopto- mediated cellular responses [42,43]. Namely, ROS such as H2O2
tic/necrotic cell death; whereas low or intermediate levels of activates ASK1, which subsequently actives both p38 and c-Jun N-
oxidative stress are most effective in DNA damage, causing mutation, terminal kinase (JNK) pathway, inducing a wide variety of cellular
inflammatory reaction and promoting proliferation of cells, and responses such as proliferation, differentiation, senescence, and
ultimately inducing carcinogenesis via initiation, progressing to apoptosis [42–44]. ASK family, which consists of ASK1 and newly
cancer development [21]. In deed, convincing evidence indicated characterized ASK2, may probably be involved in ROS-mediated car-
that ROS is an endogenous class of carcinogens by triggering the cinogenesis and other human diseases, via MAPK signaling cascades
mutation of the cells [22–24]. Chronic infection of Helicobacter pylori, by triggering apoptosis and inflammation [44].
hepatitis C virus, human papilloma virus (HPV) and their carcinogen- In regards to these signal transduction pathways, it has been
esis can be explained by this mechanism. Namely, pathological levels reported by Sawa et al. more recently that 8-nitroguanosine 3′,5′-cyclic
of ROS induce increased damage to DNA, which accompanies trigger of monophosphate (8-nitro-cGMP) that is a NO dependent nitrated
mutagenesis via DNA base-modifications and mismatch repair [21]. In derivative of cGMP, plays an important role in signal transduction via
addition, RNS including nitric oxide (NO) and its derivatives such as the S-guanylation of a redox-sensor signaling protein Kelch-like ECH-
peroxynitrite (ONOO−) play important roles in the development of associated protein 1 (Keap1) [45]. The S-guanylation accompanying
cancer and viral infection. Maeda and other researchers found that inactivation of Keap1 by 8-nitro-cGMP will result in nuclear export of
during the course of viral and bacterial infections, production of NO is Nrf2 and activation of transcriptional activity of Nrf2, which subse-
enhanced through up-regulation of inducible nitric oxide synthase quently induces the expression of antioxidant enzymes such as HO-1
(iNOS) at the site of infection or inflammation, production of O•− 2 in [41] and regulates the progression of inflammation. Accordingly, 8-

Fig. 2. Schematic illustration of enhanced permeability and retention (EPR)-effect of macromolecules in solid tumors. Distribution of low molecular weight compounds (upper panel)
and high molecular weight compounds (lower panel) in tumors and inflammatory tissues are represented in time-dependent manner. Low molecular weight compounds rapidly
disappear from blood stream with very short plasma half-lives and no tumor (tissue) accumulation, whereas high molecular weight compounds gradually accumulate in solid tumor
(inflammatory tissues) by EPR. Reproduced from Ref. [124] (modified) with permission.
J. Fang et al. / Advanced Drug Delivery Reviews 61 (2009) 290–302 293

Table 1 tissues due to anatomical characteristics and pathophysiological


Factors involved in enhanced permeability and retention (EPR)-effect of macromolecules in reaction of these tissues. Namely, vascular mediatory factors such as
solid tumors.
bradykinin and NO play a key role in extravasation [62–64] (Table 1).
1 Active angiogenesis and high vascular density in tumora Thus EPR effect is now becoming a gold standard for anticancer drug
2 Defective vascular architecture, e.g. design. More recent reviews of EPR effect are seen in Refs [64,65].
Lack of smooth muscle cells
In addition to the tumor tropic mechanism by EPR effect, macro-
Reduced number of receptors for angiotensin II
Large gaps in endothelial cell–cell functions molecular therapeutics (e.g., PEGylated proteins) will offer better
Anomalous vascular conformation (branching or stretching) pharmacokinetics (i.e., longer plasma half-life), which is thus used for
3 Extensive production of vascular mediators that facilitate extravasation a, including various other diseases such as inflammation [64–66]. The macro-
Bradykinin
molecule-based ROS-mediated therapy will thus be warranted for the
Nitric oxide
VPF and VEGFb
clinical application.
Prostaglandins In this review, the therapeutic rationales of ROS will be discussed,
Matrix metalloproteinases, collagenase and some candidate agents, especially those with high therapeutic
peroxynitrite etc. response (e.g., PEGylated D-amino acid oxidase [DAO], PEGylated zinc
4 Impaired lymphatic clearance of macromolecules and lipids from interstitial tissue
protoporphyrin [ZnPP]) based on the EPR effect are introduced below.
(resulting in their retention)

See Refs. [61–66] and text for details.


a
2. Therapeutic strategies by inhibiting ROS generation: the use of
These phenomena are also found in inflammatory tissues.
b
VPF, vascular permeability factor; VEGF, vascular endothelial growth factor. antioxidative agents

Antioxidative agents include small ROS scavengers, inhibitors of ROS


nitro-cGMP seems to be a second messenger of NO, which may thus be generating enzymes, as well as antioxidative enzymes. The first
closely associated with ROS/RNS-induced inflammatory pathological antioxidative agent applied in clinic is 3-methyl-1-phenyl-2-pyrazolin-
process and the subsequent carcinogenesis. 5-one (MCI-186, edaravone, Radicut®), a small molecular ROS scavenger,
Accordingly, it is critical to control the amount of ROS carefully in which was approved for acute brain infarction in Japan, 2001 [67,68]. In
so-named oxidation therapy, because insufficient induction of ROS our group, we focused on inhibitors of XO (O•−2 generating enzyme), as
will inversely lead to increased growth of tumors. well as SOD and recently HO-1, which will be discussed in details below.

1.2. Other diseases related with ROS 2.1. Xanthine oxidase inhibitors and SOD

Besides cancer, ROS is known to be implicated in many diseases, 2.1.1. Pyrazolopyrimidine derivatives (AHPP): inhibitors of XO
because of its high reactivity and cytotoxicity. Under pathological O•−
2 could be produced from several different oxidase enzymes,
conditions such as acute and chronic infection and inflammation, including XO, NADPH-dependent oxidases, lipoxygenase, and NO
overproduction of these highly reactive metabolites will induce lethal synthases, among which XO/xanthine is one of the major sources of
damage to cellular integrity and survival [46–48], resulting in O•−
2 generation in vascular systems and in most inflammatory lesions
reversible or irreversible tissue injury. These pathological changes, [47,59,69,70]. It is therefore conceivable that suppression of O•− 2
namely ROS-related diseases and disorders include microbial infec- production with the use of XO inhibitors may be beneficial to the host
tions, inflammation, atherosclerosis, diabetes, ischemia–reperfusion as a therapeutic means.
(I/R) injury, neurologic disorders, Parkinson disease, hypertension, In this regard, allopurinol is a well-known XO inhibitor, which is a
anticancer drug-induced tissue injury, smoking-related diseases, and structural analogue of alloxanthine among pyrazolopyrimidine deriva-
aging [20,46–52]. Thus, it is reasonable to develop therapeutics for tives (Fig. 3). Allopurinol is used to treat gout that is caused by high
these ROS-related diseases by suppressing ROS generation. Indeed, levels of uric acid in the body. However, one problem inherent to the use
this therapeutic rationale has been challenged by many research of allopurinol is that the therapeutic effect of allopurinol is not dose-
groups, with the use of various antioxidative agents of low molecular dependent: at higher dose it becomes the substrate for XO, which will in
weight nature as well as enzymes, such as SOD and catalase. Further, turn produce O•− 2 and thus exacerbate the disease unless adequate dose
inhibitors of ROS generating enzyme XO, or inducers of antioxidative regimen is fulfilled [59,71]. This delicate dose optimization of allopurinol
enzyme HO-1 were being considered along this line [46,47,53–60]. will limit its wide application for the treatment of O•−
2 driven diseases.
These therapeutics, however, are opposite in mechanism to the Instead of allopurinol, we recently found a much potent XO
anticancer strategy of ROS-dependent cytotoxicity as described above. inhibitor, 4-amino-6-hydroxypyrazolo[3,4-d]pyrimidine (AHPP),
Although it seems to be controversial, it was found experimentally
practical if one can control the production of ROS at the local
pathological site, i.e. inducing the ROS generation in cancer tissue
selectively. For the treatment of inflammation and other ROS-related
diseases, suppression or elimination of ROS production can be a
remedy. Our approaches toward these goals are discussed below.

1.3. Concerns in ROS-mediated anticancer therapy

One crucial problem to be solved for anticancer strategy by ROS is


how to target the ROS production in cancer tissues selectively;
otherwise unexpected or systemic side effects will occur. Of great
importance, this problem could be effectively solved by enhanced
permeability and retention (EPR)-effect using macromolecular drugs,
polymeric micelles or nanoparticles [61–64] (Fig. 2). EPR effect is a
universal phenomenon seen in solid tumor for macromolecular drugs
in that the macromolecular or polymeric drugs will accumulate and Fig. 3. Chemical structures of xanthine and its analogues. (A), xanthine; (B), hypoxanthine;
remain selectively in solid tumor tissues, and also inflammatory (C), allopurinol; (D), AHPP; (E), alloxanthine.
294 J. Fang et al. / Advanced Drug Delivery Reviews 61 (2009) 290–302

which is more useful in terms of biochemistry (Fig. 3D). AHPP showed The main problem we encountered is, however, the very poor
an apparent Ki value of 0.17 µM, which is about 3-times lower than solubility of AHPP, which is hardly soluble in physiological solutions,
that of allopurinol (Ki of 0.5 µM), and it cannot be the substrate of XO making its therapeutic application impossible. To overcome this
at higher concentration [59]. drawback, a styrene maleic acid copolymer (SMA, Mw ~ 1280)
The therapeutic effect of AHPP was first examined in a sponta- conjugated AHPP (SMA–AHPP) was prepared, which shows relatively
neously hypertensive rat (SHR) model, in which we hypothesized that high water-solubility, meanwhile exhibits superior pharmacokinetic
the mechanism of hypertension is attributed to O•− 2 generated by XO parameter in vivo due to the macromolecular nature of the conjugates
in the vascular endothelium that would scavenge NO because of the [75]. Similar to AHPP, SMA–AHPP shows potent XO inhibitory activity,
rapid reaction between them [56]. Instead, NO reacts with O•− 2 at and most important, remarkable antihypertensive effect by both i.v. or
diffusion rate-limited manner, six-fold faster than the removal of O•− 2 oral administration in SHR [75]. In addition, XO is induced in many
by Cu, Zn–SOD [72,73]. Thus consumption of NO would result in infectious and inflammatory diseases such as influenza virus infection
hypertension [72,74]. Namely, vascular endothelial XO may counteract and inflammatory bowel disease. O•− 2 is then generated there, which is
against NO-induced vasorelaxation indirectly by O•− 2 generation. Thus the major pathological factor in these diseases [46–48,76]. Therefore
inhibition of XO activity may result in an antihypertensive effect the effect of SMA–AHPP was examined in a mouse colitis model,
(vasodilatation) by increasing the level of NO in vascular system. This which is induced by oral administration of dextran sodium sulfate
offers a new strategy against hypertension. As expected, AHPP (DSS). Our preliminary data showed that the symptom, as well as the
significantly augmented NO-mediated relaxation of aortic rings from survival rate of mice of this colitis model were greatly improved by
both rabbits and SHR in a dose-dependent manner; i.v. injection of SMA–AHPP (unpublished data).
AHPP reduced the blood pressure of SHR to 70% of the initial blood
pressure, which is almost the level of normal rats [59] (Fig. 4). These 2.1.2. SOD and its polymer conjugate
data strongly suggest the therapeutical potential of AHPP. Great interests have been focused on SOD for therapeutic use,
including Cu, Zn–SOD and Mn–SOD as the major scavenger of O•− 2 [21].
However, because of their poor pharmacological properties, such as an
extremely rapid plasma clearance time (e.g. plasma half-life b5 min in
mice), instability, and immunogenicity in vivo, the clinical application
of SOD as a therapeutic agent was very limited. The clinical application
of SOD is still problematic and remains unclear. The double-blind
controlled clinical trial has not reported positive effect of SOD in a
chronic inflammatory or autoimmune disease in humans. And it
seemed that SOD is not fully protective for myocardial reoxygenation
injury in open-chest dogs [21]. Many other trials of native/
recombinant SOD did not show efficacy in clinic. The reasons about
these problems may be explained by the following issue: SOD itself
catalyses dismutation of O•− 2 to yield H2O2, which will be then
converted to •OH via Fenton reaction by Cu2+ or other metals, and
more important Cu2+ is released from Cu, Zn–SOD [77]. This Cu2+
mediated generation of •OH may result in adverse effects of SOD
therapy. In this context, when combined with catalase, the effect of
SOD is greatly improved [21,53]. In addition, this SOD-medicated •OH
generation may be inhibited by chemical modification of SOD with Cu2+
chelating capacity, for example our findings from pyran copolymer–
SOD indicate that chelating effect of dicarboxylate of pyran may
suppress this reaction [65,78].
To improve these pharmacological and biochemical problems, a
wide variety of Cu, Zn–SOD conjugates were developed initially,
including pyran copolymer–SOD, polyethylene glycol (PEG)–SOD,
Ficoll–SOD, lecithinized SOD, styrene maleic acid copolymer (SMA)–
SOD, and gelatin or albumin conjugated SOD complexes. They all have
longer circulation half-lives than unconjugated SOD, high stability and
less immunogenicity [21,79,80].
In our laboratory, in the past 20 years we have studied on the role
of O•−
2 in various diseases, especially in virus infections, and developed
a pyran copolymer–SOD conjugate as most useful among others [46–
48]. In the pathogenesis of various viral diseases including influenza,
herpes simplex virus and Sendai virus, and bacterial infections
[27,41,46,47], source of O•−2 production is mainly from i) host's own
macrophages and granulocytes (neutrophils) in the “respiratory
burst”, and ii) XO, which is elevated enormously in serum and tissues
after influenza virus infection [46–48]. Namely, XO is 30–400 folds
higher in the lung of virus-infected mouse than healthy control mice
Fig. 4. Effect of AHPP on the blood pressure of spontaneous hypertensive rats (SHR). [48]. Therefore, treatment was undertaken by use of SOD or XO
(A) shows the antihypertensive effect of AHPP after i.v. administration in SHR, and the inhibitors. For this purpose, pyran copolymer–SOD conjugate was
effect of L-NAME on reduction of hypertension induced by AHPP in SHR is shown in developed in 1989 [46,47]. Compared with native SOD, pyran
(B). XO inhibitors (e.g., AHPP) were injected via the jugular vein (at 0 time), after
which mean arterial blood pressure was continuously measured. Data are means ± SE
copolymer–SOD showed much improved pharmacological properties,
(n = 8). ⁎P b 0.05; †P b 0.025; ‡P b 0.01. L-NAME, Nω-nitro-L-arginine methyl ester (NOS e.g., prolonged plasma half-life, 6 h vs about 4 min of native SOD,
inhibitor). Reproduced from Ref. [59] with permission. while retaining about 80% of the enzyme activity [79]. Daily i.v.
J. Fang et al. / Advanced Drug Delivery Reviews 61 (2009) 290–302 295

injection of pyran copolymer–SOD conjugate at the dose of 200 U/


mouse from day 5 to 8 after influenza virus infection protected 95% of
the mice from death while less than 5% survival without this
treatment (Fig. 5). It should be noted that, in this mouse model,
there was no detectable virus while the disease reached maximum as
judged by consolidation score of the lung and mortality as well as
infiltration of plasma cells in the alveolar compartment [46]. These
studies thus demonstrated for the first time that the real cause of this
viral disease was O•− 2 , but not the virus per se. Along this line, we
further developed a gelatin–SOD conjugate, which showed significant
suppressive effective against collagen-induced arthritis in mouse
model, which indicated that O•− 2 is also involved in the arthritis [80].
Meanwhile, many other research groups reported similar ther-
apeutic effect in various diseases including I/R injury, by use of various
SOD conjugates [21]. Among these, lecithinized Cu, Zn–SOD, which
was developed by Igarashi R et al. [81] exhibited promising
therapeutic efficacy against various ROS-related diseases, such
as ischemic injury, neuron injury, inflammation and HIV infection
Fig. 6. Schematic representation of cytoprotective role of HO-1 and mechanisms of
[81–85], and it is now under the clinical trail for ulcerative colitis and antitumor effect of ZnPP and its polymer (micelle) derivatives. Reproduced from Ref.
interstitial pneumonitis. [124] with permission.

2.2. Heme oxygenase (HO)


variety of stress-inducing stimuli including hypoxia, heavy metals, UV
HO is a key enzyme to regulate the intracellular heme level, which irradiation, ROS and RNS such as H2O2 and NO [60,87,88]. It has been
catalyzes the initial and rate-limiting step of heme degradation, reported that HO-1 stumulates cell growth and the proliferation of
resulting in the formation of biliverdin, carbon monoxide (CO), and various cells including epidermal keratinocytes, vascular endothelium
free iron [60,86]. Biliverdin is subsequently reduced by biliverdin and coronary endothelial cells, via its cytoprotective and antiapoptotic
reductase to produce bilirubin, a potent and crucial antioxidant (Fig. effect [60]. Studies with HO-1-deficient mice show embryonic loss and
6). Three isoforms have been identified for mammalian HO: HO-1, HO- subsequent death significantly. Further chronic inflammation in the
2 and HO-3 [60]. HO-1 is the inducible form enzyme, which is however kidney and the liver was observed in HO-1−/− adult mice, which
highly expressed in liver and spleen under physiological condition, and further support this role of HO [60]. The antioxidative and anti-
interestingly in most cancer cells; HO-2 is the constitutive isoform, apoptotic effect of HO-1 is proposed to be related to multiple
expressed mainly in brain and testis. HO-3 is a recently identified mechanisms: i) decreasing the prooxidant level (heme); ii) increasing
isoform, which has been detected in many organs; however, its activity the antioxidant level (bilirubin); iii) producing the antioxidative,
is very low compared to other two isoforms, and thus its physiological antiapoptotic molecule CO; iv) inducing ferritin, which removes
role remains unclear. and detoxifies free ferric ion, a potent source of •OH generation; and
Among these isoforms, HO-1 plays most important role in cell v) preventing over-stimulation of the immune response [60,89,90]
growth and cell death, by protecting cells against harmful oxidative (Fig. 6).
stress and apoptosis [60]. HO-1 also belongs to a member of the heat Because of the antioxidative and antiapoptotic effect, potent
shock protein family (Hsp-32), whose expression is triggered by a cytoprotective role of HO-1 was found subsequently to be involved
in various diseases and disorders, including atherosclerosis, hyperten-
sion, acute renal injury, toxic nephropathy, transplant rejection,
endotoxic shock, chronic obstructive lung disease, gastrointestinal
diseases, and Alzheimer's disease [60]. These facts suggest HO-1 as
a possible therapeutic target in these diseases. Indeed, inhibition of
HO-1 by specific HO inhibitor such as ZnPP or tin protoporphyrin
has resulted in the worsening of the diseases [60]. On the
other hand, administration of HO-1 inducers, such as cobalt proto-
porphyrin, hemin, and trinitrobenzene sulfonic acid or HO-1 gene
transfection showed significant therapeutic effect in many diseases
such as I/R injury, cisplatin nephrotoxicity and hypoxia-induced lung
injury [60].
Regarding the agent related to HO-1, hemin is a potent HO-1
inducer, and it has been used for acute intermittent porphyria in USA
and Europe as an orphan drug, however, it encountered with technical
difficulties and complications, and is associated with a high risk of
thrombophlebitis and coagulation disturbances [91]. A water soluble
form of hemin, heme arginate (Normosang®) was then developed,
however, its plasma half-life is relatively short (~10.8 h in human after
Fig. 5. Therapeutic effect of SOD and pyran–SOD conjugate on influenza virus-infected
mice. Male ddY mice, aging 4–6 weeks, were used in the experiments. Mice received the i.v. injection) mostly due to binding to hemopexin [91]. To overcome
influenza virus by inhalation of virus aerosol at 2 × LD50 dose. Ten mice were used in these problems and to obtain better pharmacokinetics, we synthe-
each treated group and 20 mice in the control group. SOD (200 and 1000 U per mouse) sized PEG conjugated hemin (PEG–hemin), which became water
and pyran–SOD conjugate (200 U per mouse) were given i.v. injection once daily for soluble molecules and behaved as water soluble micelles (unpublished
four consecutive days from 5 days after virus infection. ●, Control; ○, SOD (200 U per
mouse); e, SOD (1000 U per mouse); ▲, pyran–SOD conjugate (200 U per mouse). The
data). We thus anticipate a much longer plasma half-life of PEG–hemin
mortality rate of mice did not increase after day 15. Mice surviving on day 15 were as shown in many other PEGylated agents, such as PEG–interferon
considered as being cured. Reproduced from Ref. [46] with permission. [92,93]. In our preliminary experiments, addition of PEG–hemin to
296 J. Fang et al. / Advanced Drug Delivery Reviews 61 (2009) 290–302

cultured cells significantly induced the HO-1 expression; i.v. injection [96,97]. NO has been reported to exhibit remarkable cytoprotective
of PEG–hemin markedly alleviated the degree of liver injury in a rat I/R effect for I/R injuries [95,98]. Lack of NO may also contribute the
model, as evaluated by the value of liver enzymes in plasma (AST, ALT, pathogenesis of I/R because decrease of NO can trigger neutrophil
LDH) (unpublished data). Thus it became apparent that PEG–hemin adherence, platelet aggregation and exudates into the ischemia area,
might be a potentially useful agent for various ROS-related diseases, which exacerbates reperfusion injury. The cytoprotective and bene-
which warrants further investigations. ficial effects of NO (or nitrite) is extensively reviewed in this special
issue of the journal by Gladwin et al.
2.3. NO Regarding the cytoprotective effect of NO in I/R injuries, we
developed a S-nitrosated α1-protease inhibitor (S-NO-α1-PI), which
NO produced in biological systems exerts a variety of pathophy- has stable endogenous NO releasing activity, with long plasma half-
siological roles [94,95]. Excessive formation of NO at local site such as life and superior pharmacokinetics [99]. S-NO-α1-PI treatment
cancer and inflammation have pro-inflammatory effects such as significantly suppressed the elevation of liver enzymes (AST, ALT,
edema formation involving enhanced vascular permeability, inflam- LDH), neutrophil accumulation and apoptosis of liver cells in rat I/R
matory cell infiltration, and cytotoxicity, possibly through the model. Moreover, it improved the impaired hepatic blood flow to a
subsequent formation of ONOO− and other RNS [28]. In our great extent [100] (Fig. 7). Similarly, another NO donor, S-nitrosated
laboratory, we found NO is produced at the site of viral and bacteria albumin, showed remarkable cytoprotective effect against I/R injury
infections, by inducible NO synthase (iNOS) that is primarily of rat liver and bacterial infections [100]. These data suggest the
expressed in inflammatory leukocytes, from L-arginine as substrate. feasibility of these well designed NO donors for clinical application.
Inhibition of NO production by using NOS inhibitors or knockdown of
iNOS greatly improved the histopathological changes and suppressed 2.4. Canolol, a ROS scavenging phenols from plant
disease progression [25–27,35]. However, NO also was found
beneficial when it exists at low or moderate levels, including anti- Many flavonoids and phenolic compounds show potent antiox-
inflammatory activities, antiapoptotic and antioxidative effects idant activity [21]. Along this context, we recently isolated a phenolic

Fig. 7. Therapeutic effects of S-NO-α1-PI in a rat liver ischemia–reperfusion (I/R) model. Changes in serum levels of AST, ALT and hepatic blood flow are shown in (A) and (B),
respectively. (C) shows the neutrophil infiltration in rat liver after I/R, with or without S-NO-α1-PI treatment, and (D) shows apoptosis of hepatic cells induced by I/R in the presence
or absence of S-NO-α1-PI. ⁎P b 0.05, ⁎⁎P b 0.01. Reproduced from Ref. [96] with permission.
J. Fang et al. / Advanced Drug Delivery Reviews 61 (2009) 290–302 297

remarkable antitumor effects [14,15,106–109], which is to be dis-


cussed in details at below.

3.1. Poly(ethylene glycol) conjugated xanthine oxidase (PEG–XO)

As described earlier, XO, a major O•− 2 generating enzyme, is a


metalloflavoprotien (Mw 298 kDa, a heterodimer) that catalyzes the
Fig. 8. Chemical structure of canolol (Ref. [101]).
oxidation of xanthine/hypoxanthine to generate O•− 2 , which is
subsequently converted to H2O2 by SOD. XO was thus considered to
be a candidate of oxidation therapy [18]. However, when XO is used as
compound from crude canola (rape seed) oil, 4-vinyl-2,6-dimethox- native enzyme, its high affinity to blood vessels limits its application,
yphenol named canolol (Fig. 8), which exhibited potent antioxidant which will induce systemic vascular damage and hypertension [17,59].
activity [101]. Scavenging potency of canolol against alkylperoxyl This drawback can be avoided by PEGylation of XO to PEG–XO via
radical (ROO•) is much higher than well-known antioxidants, such as conjugating PEG to ɛ-amino groups of lysine residues of XO, which
α-tocopherol, vitamin C, β-carotene, rutin, and quercetin [102]. play a crucial role for binding to the luminal surface of vascular wall
Canolol also showed strong scavenging capacity against the endo- [14]. In addition, the EPR effect of PEG–XO and excellent pharmaco-
genous mutagen, ONOO−, thus greatly suppressing bacterial muta- kinetics were demonstrated and PEG–XO activity was retained in
tion, protecting DNA from oxidative damage, as well as inhibiting tumor for more than 96 h. However, for in vivo treatment we found
oxidation of lipids and proteins [102]. Moreover, canolol exerted the infusion of the substrates (xanthine or hypoxanthine) must be
cytoprotective activity against ONOO− induced cytotoxicity in human performed with adequate time interval after i.v. injection of PEG–XO,
kidney epithelial cells HEK293 and human bronchial epithelial cells to ensure tumoritropic accumulation of PEG–XO which will take
HBE140 (unpublished data). Canolol can also suppress the induction usually more than 24 h, otherwise the enzyme (XO) in circulation will
of iNOS and various inflammatory cytokines such as interleukin-1β generate O•−2 systemically because of incoming substrate, resulting
(IL-1β), tumor necrosis factor-α (TNF-α), interferon γ (IFN-γ) and systemic side effects. According to the above-mentioned protocol,
cyclooxygenase-2 (COX-2) [103]. As a consequence, gastric carcino- significant antitumor effect was achieved by PEG–XO plus subsequent
genesis was markedly suppressed in H. pylori-infected Mongolian infusion of the substrate, hypoxanthine; tumor growth was remark-
gerbils [103]. Interestingly, the viable H. pylori count was not changed ably suppressed up to at least 52 days, in which complete regression of
by feeding with the canolol containing diet. These results suggest that tumor growth was found in three of seven tumor-bearing mice.
the effect of canolol is to ameliorate the inflammation and Meanwhile, no apparent side effects were observed after this
carcinogenesis caused by ROS, but it dose not kill the bacteria. This treatment [14].
is the same mechanisms of pyran–SOD induced protection against
viral infection [46]. In addition, we recently found that canolol showed 3.2. Poly(ethylene glycol) conjugated D-amino acid oxidase (PEG–DAO)
remarkable therapeutic effect in a murine colitis model induced by
orally administration of DSS (unpublished data). These data suggest Along this line, we recently focused on H2O2-generating enzyme
the potential of canolol as a cancer preventive agent via anti- DAO. DAO is a flavoprotein (Mw 39 kDa) that catalyzes the
inflammatory effect. stereoselective oxidative deamination of D-amino acids to the
Regarding the activity of canolol, it should be noted that canolol corresponding α-keto acids, during which molecular oxygen is used
directly scavenges ROS (ONOO−, O•− 2 and ROO∙ etc.), but also as an electron acceptor, and H2O2 is generated [110]. An important
suppresses the induction of inflammatory mediators (e.g., IL-1β, merit of DAO is that the substrate (D-amino acids) exists very little
TNF-α, IFN-γ and COX-2) at mRNA level indirectly, as shown above under normal circumstance, and thus making it possible to control
[103], which is the same mechanism as seen in many flavonoids and H2O2 production by controlling exogenous infusion of D-amino acids.
other phenolic compounds from different plants [104,105]. We further conjugated DAO with PEG to become macromolecules (Mw
of ~63 kDa), which exhibited tumor-selective accumulation based on
3. Oxystress inducing anticancer therapy: oxidation therapy the EPR effect [15,111]. Accordingly, PEG–DAO/D-amino acids system
will yield better tumor-selective generation of H2O2 by a double
As discussed above, because of the toxic nature of ROS and targeting effect. Namely, D-proline was administered with a time
insufficient antioxidant enzymes in cancer cells, overproduction of interval (e.g., ~4 h) after PEG–DAO injection. During this lag time,
ROS will eliminate unwanted cancer cell. Based on this principle, a PEG–DAO is allowed to accumulate in tumor first. The subsequent
unique “oxidation therapy” was introduced [14,15,17–20]. One way to administration of D-proline will then be converted to H2O2 mostly in
achieve this anticancer therapy is to deliver ROS generating enzymes tumors. As expected, significant suppression of tumor growth was
to tumor tissues directly, such as glucose oxidase, XO and DAO, where observed by this treatment, with a significant increase of the survival
more selective delivery to tumor tissue can be accomplished by EPR rate in the tumor-bearing mice (Fig. 9A). PEG–DAO thus becomes
effect after pegylation of these oxidases [14,15,17–19]. Another more ideal candidate for oxidation therapy.
approach is to suppress the antioxidative systems in tumors using In addition, DAO is a monomeric simple protein and is easy to
inhibitors against for instance HO-1 which is highly upregulated in handle and produce by recombinant technology. Compared with DAO,
many tumors. These antioxidative systems are important for tumor XO is difficult to manufacture by recombinant technology because it is
cells to defend against the oxystress (e.g., ROS and RNS). Thus a relatively large molecule; it is a heterodimer with complex structure.
inhibition of the antioxidant enzymes, i.e. HO-1 in tumor can also be We recently reported production of recombinant porcine DAO using E.
an anticancer therapeutic [20,106]. coli with high yield (20 mg/l), and high enzyme activity (5.3 U/mg),
To exploit the oxidation therapy, we have developed several as well as reasonable stability [111].
polymer conjugates of ROS generating enzymes (XO and DAO) and Among the antioxidative enzymes, catalase plays a central role
HO-1 inhibitory compounds (ZnPP). Glucose oxidase was not selected against H2O2 [112]. Although some hepatomas has been found to
for this tactics because its substrate glucose is available ubiquitously in express high level of catalase [113], in most cases, significantly
vivo that will end up no regulation in ROS generation once this reduced catalase activity have been reported in many tumors [10,13].
enzyme is injected in vivo. The polymer conjugates exhibited tumor- Similar findings were observed in our group by measuring the catalase
targeting characteristics by taking advantage of EPR effect, resulting in activities in tumor tissues and normal tissues, such as the liver, kidney
298 J. Fang et al. / Advanced Drug Delivery Reviews 61 (2009) 290–302

Fig. 9. Antitumor effect of PEG–DAO (A), and PEG–ZnPP (B) in murine tumor models. 2 × 106 of mouse sarcoma S-180 cells (A) or colon 38 cancer cells (B) were implanted s.c. in ddY
mice or C57BL/6 mice respectively. Six to ten days after tumor inoculation, mice were treated by each agent as indicated. Inlet of (A) shows the survival rate of tumor-bearing mice
after treatment. Data are means (n = 4–8); bars, SE. ⁎, P b 0.001. Reproduced from Refs. [106,111] with permission.

and lung, in both experimental mouse tumor and chemically induced 3.3. HO inhibitor: zinc protoporphyrin IX (ZnPP)
rat tumors (Table 2). Coincided with these observations, we found a
significantly higher IC30 (the concentration of drugs when cell growth As described above, HO-1 serves as a key enzyme to defend against
is inhibited to 30% compared with untreated control cells) to PEG– oxidative stress [60,86]. More importantly, it has also been known
DAO/D-proline (H2O2) against normal cells; a great contrast to tumor upregulated and to play crucial roles in many tumors [57], including
cells (Table 3 and Ref. [101]). These findings further supported the renal cell carcinoma, prostate tumors in humans, and experimental
validity and safety of ROS-induced anticancer therapeutics, i.e. PEG– animal solid tumors, such as rat hepatoma AH136B and mouse
DAO/D-proline dependent treatment [15,111]. sarcoma S-180, to support tumor growth through its antiapoptotic and
Even though H2O2 was utilized as the antitumor principle in our antioxidative effects [60,87,88,107]. HO-1, also known as a heat shock
studies according to its potentially high cytotoxicity, it should also be protein (Hsp 32), is considered to be a survival factor of tumor cells.
noted H2O2 exists physiologically in most organisms, which is the We thus realized that impairment or inhibition of this defensive
resulting product of SOD mediated O•− 2 dismutation. With regard to molecule in tumor cells should be useful as therapeutic target, which
the physiological function of H2O2, it has been recently reported that is the second approach for oxidation therapy. Subsequently, we
H2O2 plays an important role in vasodilation, similar to the function of confirmed that this strategy worked in many experimental solid
NO [114,115]. Thus in our studies of PEG–DAO/D-proline, even though tumors, by use of a potent HO inhibitor ZnPP [87,88,106,109 and
incidental generation of H2O2 systemically may occur, it is however in
relatively low level compared to the targeted intense generation of
H2O2 in tumor that achieved by EPR effect and time-lagged infusion of Table 3
the substrate, which may be tolerated. Whereas, more important, the IC30 of PEG–DAO/D-proline against tumor cells and normal cells.
systemically generated small amount of H2O2 may increase the blood Tumor cellsa IC30 (mM)b Normal cellsa IC30 (mM)
flow of tumor because of the induced vasodilation as described above, DLD-1 1.7 BNL.CL2 11.8
which will further enhance the EPR effect, consequently improve the Sk-Hep 1.8 MDCK 10.0
therapeutic activity of PEG–DAO/D-proline. This notion however HT-29 3.1 CEF 4.8
needs further investigation to be clarified. A431 1.4 Hc 19.0
HeLa 0.9 HRPEC 40.0
CNE 1.8 CV1 8.2
ES2 1.4 HBE140 3.7
Table 2 Meth-A 3.2
Catalase activities in tumor tissues and normal tissues. SW480 0.9
Lxc 4.5
Tumor modelsa Catalase activity (Kat.f)b
KYSE510 0.3
Tumor Kidney Liver Lung Average 1.9 ± 0.4⁎ Average 13.9 ± 4.5⁎
S-180 53 7745 8495 2508
*P b 0.01.
B16 207 ± 12 10564 ± 788 7980 ± 834 1249 ± 194 a
DLD-1, HT-29, SW480, human colon cancer cells; Sk-Hep, human liver cancer cells;
DMBA induced tumor 559 ± 271 10499 8408 1017
A431, human lung cancer cells; HeLa, human cervical cancer cells; CNE, Lxc, human
Reprinted from Ref. [111] with permission. nasopharyngeal carcinoma cells; ES2, human ovarian cancer cells; Meth-A, mouse
a
S-180: mouse sarcoma S-180 tumor; B16: mouse B16 melanoma; DMBA induced fibrosarcoma cells; BNL.CL2, murine embryonic hepatocytes; MDCK, Madin–Darby
tumor: DMBA (12-dimethylbenz (a) anthracene) induced rat breast cancer. Canine Kidney cells; CEF, chick embryonic fibroblasts; Hc, human hepatic cells; HRPEC,
b
The animals bearing the tumor, which was around 10 mm diameter, were killed and human retinal pigment epithelium cells; CV1, monkey kidney cells; HBE140, human
then were subjected to reperfusion with saline containing heparin (5 U/ml). The tissues bronchi embryonic cells.
b
subsequently collected were homogenized and subjected to the catalase activity assay. IC30 was calculated by the concentration of D-proline in the presence of 10 mU/ml
Data are mean ± S.E., n = 3–6. PEG–DAO, when cell growth is inhibited to 30% compared to untreated control cells.
J. Fang et al. / Advanced Drug Delivery Reviews 61 (2009) 290–302 299

unpublished data], indicating a potentially beneficial role of HO conventional chemotherapeutic agents. Namely, PEG–ZnPP treated
inhibitor, i.e. ZnPP as a novel anticancer drug. SW480 cells in vitro became much more vulnerable to the insults
caused by ROS or ROS generating anticancer drugs doxorubicin and
3.3.1. Polymer conjugated ZnPP (PEG–ZnPP) and SMA–micelle of ZnPP camptothecin [118]. Namely, the IC50 values were reduced to 75%, 61%,
(SMA–ZnPP) 17%, and 39% for H2O2, t-butyl hydroperoxide, camptothecin and
ZnPP is almost insoluble in physiological aqueous solution, which doxorubicin, respectively, by pretreatment with PEG–ZnPP. In vivo
limits its practical use as drug. PEGylation of ZnPP was thus carried experiments further verified that this notion was valid with the use of
out, and PEG–ZnPP showed much increased water-solubility, and PEG–DAO/D-proline in addition to PEG–ZnPP [118].
more importantly, it formed macromolecular micelles of molecular
weight more than 70 kDa in aqueous media as demonstrated by 3.3.4. New insight into the mechanisms of PEG–ZnPP/SMA–ZnPP
Sephadex G-75 chromatography. Moreover, a hydrodynamic diameter dependent cytotoxicity
about 200 nm of PEG–ZnPP was observed indicating multimolecular With regard to the antitumor mechanism based on depletion of
associated form of micelles [107 and unpublished data]. When PEG– anti-ROS defense by inhibiting HO-1 in tumor cells, PEG–ZnPP or
ZnPP was administered i.v. it showed a much longer plasma residence SMA–ZnPP was found to down regulate the oncogene BCR/ABL in
time, 40 times of nonconjugated free ZnPP. In addition, PEG–ZnPP chronic myeloid leukemia (CML), which resulted in apoptosis of the
preferentially accumulated in solid tumor in murine tumor models CML cells [119,120]. Moreover, both PEG–ZnPP and SMA–ZnPP were
(about 5–10 times higher concentration than normal organs) due to also found effective on imatinib (Gleevec®) resistant CML as well as
EPR effect as expected [106]. acute lymphoblastic leukemia (ALL) [121,122]. These findings suggest
PEG–ZnPP remarkably suppressed the growth of S-180 and colon that HO-1 (Hsp-32) is an attractive molecular target for cancer
38 tumors implanted in ddY and BALB/c mice, respectively, whereas chemotherapy with versatile functions. Inhibition of HO-1 by PEG–
no or very little side effect at all was noticed, if any, during this ZnPP or SMA–ZnPP not only induces the ROS-related cytotoxicity, but
experiment (Fig. 7B). Moreover, PEG–ZnPP treatment induced tumor- also leads to the cancer cell death via other mechanisms without
selective suppression of HO activity significantly, even though HO-1 detrimental toxicity to the host in vivo, which will greatly increase the
protein seemed to increase after this treatment, leading to apoptosis therapeutic efficacy, and expand the clinical usage of PEG–ZnPP or
of tumor cells probably through increased oxidative stress [106]. SMA–ZnPP. In addition, there may be other mechanisms for PEG–
Besides PEG–ZnPP, we successfully prepared a highly water soluble ZnPP/SMA–ZnPP actions involving intracellular zinc signaling as
micellar form of ZnPP using SMA, which also revealed macromole- described by Hirano et al. [123], in which porphyrin may serve as
cular nature with the molecular weight and average micelle size of the carrier of zinc.
144 kDa and 176.5 nm, respectively, in physiological aqueous solutions
[108]. Similar to PEG–ZnPP, SMA–ZnPP showed a potent antitumor 3.4. Caution for oxidation therapy
effect in many solid tumor models including DMBA induced breast
cancer in rats and fibrosarcoma Meth-A, colon 38 cancer in mice, Even though the preclinical studies of oxidation therapy showed
whereas no apparent side effects was found even at the dose of 200 the promising results as shown above, it is critical and necessary to
mg/kg when injected i.v. [109 and unpublished data]. carefully control the amount of ROS. As described earlier, the
antitumor effect of ROS is dose-dependent; however, low or
3.3.2. PEG–ZnPP/SMA–ZnPP for photodynamic therapy (PDT) inadequate dose of ROS will trigger the progression of tumor. It is
It is well known that porphyrin derivatives are efficient photo- thus necessary to deliver excess amount of ROS into tumor, e.g.,
sensitizers for photodynamic therapy (PDT), and it is among milimolar concentration; otherwise adverse effect, namely tumor
promising and least invasive antitumor treatment modalities using progression, may occur. In our experiments using ROS generating
laser beam usually to generate singlet oxygen [116,117]. It is thus enzymes, relatively high dose of enzymes and their substrates (e.g., 2
feasible for ZnPP, especially its polymer (micelle) derivatives (e.g., U/mouse of PEG–DAO and 0.5 mmol/mouse of D-proline, cf. Ref. [111])
PEG–ZnPP, SMA–ZnPP) to be applied in PDT, which will further elevate were applied, and tumor targeted delivery of ROS was of course
the anticancer activities of these anticancer agents as discussed above. achieved, by using polymeric conjugate according to EPR effect and
One advantage of PEG–ZnPP/SMA–ZnPP over conventional photo- time-lagged infusion of the substrate [111], which will further increase
sensitizer (i.e. photopyrin) used for PDT, is that ZnPP does not require the intratumor concentration of ROS, and reduce the system side
laser beam whereas photopyrin requires laser beam at 630 nm. In effects. In fact, at the dose level of DAO and D-proline in vivo we have
contrast, ZnPP is active even under conventional xenon light and not observed toxicity such as death or significant body weight loss of
endoscopic light source. the mice. Accordingly, cautiously manipulation and development of
As expected, ZnPP efficiently generated highly reactive singlet effective tumor-targeting system are necessary to develop the
oxygen under illumination of visible light, laser, or xenon light [109]. oxidation therapy for clinical application.
Utilizing a tungsten–xenon light as the irradiation source, SMA–ZnPP/
PEG–ZnPP induced marked tumor regression, both in mouse tumor 4. Conclusions
xenograft and chemical induced rat beast cancers [109 and unpub-
lished data]. The light source used in this study for activating ZnPP is ROS, a group of highly reactive molecules, exhibits vital role in
the commonly used endoscopic light or bulb (Philips, TL-D 15W) used aerobic organisms as the indispensable factors of signal transduction
for icteric jaundice. The laser apparatus for conventional PDT is much pathway to regulate cell growth and drug metabolism [1,2], and it is
more expensive (~500, 000 US dollar) than that of xenon light source, like double-edge sword. Under physiological conditions, damages are
which is used in endoscope and almost available in all hospitals and reduced by cellular antioxidative defenses (e.g., SOD, catalase, HO-1)
clinics, the application of SMA–ZnPP or PEG–ZnPP under the and repair mechanisms [6]. However, under pathological circum-
irradiation by endoscopic light may be more convenient for certain stances such as inflammation [46–49], over-burden of ROS will lead to
cancers of such as esophagus, breast, bronchogenic origin (lung), reversible or irreversible tissue injury. Namely, ROS is potentially
colon, urinary bladder, and cervicus. harmful by reacting with proteins, DNA and other vital molecules, thus
damages the survival of cells [3–5]. Consequently it will induce various
3.3.3. Other aspects of ZnPP and its polymer derivatives diseases, including cancer [51,52,60,124]. It was thus suggested that
In combination with ROS or ROS-generation system (i.e., PEG– inhibiting or scavenging ROS would lead to the therapeutic modality
DAO), preliminary study showed a synergistic effect in PEG–ZnPP and for ROS-related diseases, utilizing either antioxidative compounds
300 J. Fang et al. / Advanced Drug Delivery Reviews 61 (2009) 290–302

(e.g., Radicut® and canolol) or antioxidant enzymes (e.g., SOD, HO-1). [20] P. Huang, L. Feng, E.A. Oldham, M.J. Keating, W. Plunkett, Superoxide dismutase as
a target for the selective killing of cancer cells, Nature 407 (2000) 390–395.
However, native SOD or low Mw ZnPP (HO inhibitor) did not show [21] B. Halliwell, J.M.C. Gutteridge (Eds.), Free Radicals in Biology and Medicine,
significant therapeutical potentials, but when they were made as Oxford University Press, NJ, USA, 1999, pp. 617–859.
polymer conjugates (PEGylation or micellar formation), to exert [22] K.Z. Guyton, T.W. Kensler, Oxidative mechanisms in carcinogenesis, Br. Med. Bull.
49 (1993) 523–544.
selective targeting and to improve their pharmacokinetics as well as
[23] D.I. Feig, T.M. Reid, L.A. Loeb, Reactive oxygen species in tumorigenesis, Cancer
in vivo half-life, they exhibited marked effects in vivo. Res. 54 (1994) 1890s–1894s.
On the other hand, cytotoxic ROS can also serve as a tumor [24] P.A. Gerutti, Oxy-radicals and cancer, Lancet 344 (1994) 862–863.
terminator if it is produced selectively in tumors. This anticancer [25] S. Fujii, T. Akaike, H. Maeda, Role of nitric oxide in pathogenesis of herpes simplex
virus encephalitis in rats, Virology 256 (1999) 203–212.
therapy was named as “oxidation therapy” and many examples
[26] T. Akaike, S. Okamoto, T. Sawa, J. Yoshitake, F. Tamura, K. Ichimori, K. Miyazaki,
presented here support this strategy. One of our recent promising K. Sasamoto, H. Maeda, 8-nitroguanosine formation in viral pneumonia and its
developments is PEG–ZnPP, which appears to have multiple antic- implication for pathogenesis, Proc. Natl. Acad. Sci. U. S. A. 100 (2003) 685–690.
ancer mechanisms including increase of susceptibility of cancer cells [27] K. Umezawa, T. Akaike, S. Fujii, M. Suga, K. Setoguchi, A. Ozawa, H. Maeda,
Induction of nitric oxide synthesis and xanthine oxidase and their roles in the
to ROS as well as modulation of signal transduction cascade. antimicrobial mechanism against Salmonella typhimurium infection in mice,
In conclusion, all the studies of controlling ROS generation give us Infect. Immun. 65 (1997) 2932–2940.
clues to treat various diseases including cancer and hypertension by [28] T. Akaike, H. Maeda, Pathophysiological effects of high-output production of
nitric oxide, in: L.J. Ignarro (Ed.), Nitric Oxide: Biology and Pathobiology,
modulating the ROS generation, which might be promising Academic Press, San Diego, 2000, pp. 733–745.
approaches toward clinical application in the future, which warrants [29] X. Ding, Y. Hiraku, N. Ma, T. Kato, K. Saito, M. Nagahama, R. Semba, K. Kuribayashi,
further investigation. S. Kawanishi, Inducible nitric oxide synthase-dependent DNA damage in mouse
model of inflammatory bowel disease, Cancer Sci. 96 (2005) 157–163.
[30] N. Ma, T. Tagawa, Y. Hiraku, M. Murata, X. Ding, S. Kawanishi, 8-Nitroguanine
Acknowledgement formation in oral leukoplakia, a premalignant lesion, Nitric Oxide 14 (2006)
137–143.
[31] P. Chaiyarit, N. Ma, Y. Hiraku, S. Pinlaor, P. Yongvanit, D. Jintakanon, M. Murata,
The works included in this paper are supported in part by Grant-in- S. Oikawa, S. Kawanishi, Nitrative and oxidative DNA damage in oral lichen
Aid from the Ministry of Education, Culture, Sports, Science and planus in relation to human oral carcinogenesis, Cancer Sci. 96 (2005) 553–559.
Technology of Japan (No. 20590049, No. 20015045 and No. S0801085). [32] S. Kawanishi, Y. Hiraku, Oxidative and nitrative DNA damage as biomarker for
carcinogenesis with special reference to inflammation, Antioxid. Redox Signal. 8
(2006) 1047–1058.
References [33] T. Okamoto, T. Akaike, T. Sawa, Y. Miyamoto, A. van der Vliet, H. Maeda, Activation
of matrix metalloproteinases by peroxynitrite-induced protein S-glutathiolation
[1] K.J. Davies, Oxidative stress: the paradox of aerobic life, Biochem. Soc. Symp. 61 via disulfide S-oxide formation, J. Biol. Chem. 276 (2001) 29596–29602.
(1995) 1–31. [34] J. Wu, T. Akaike, K. Hayashida, T. Okamoto, A. Okuyama, H. Maeda, Enhanced
[2] M. Sundaresan, Z.X. Yu, V.J. Ferrans, K. Irani, T. Finkel, Requirement for generation vascular permeability in solid tumor involving peroxynitrite and matrix
of H2O2 for platelet-derived growth factor signal transduction, Science 270 metalloproteinases, Jpn. J. Cancer Res. 92 (2001) 439–451.
(1995) 296–299. [35] J. Yoshitake, T. Akaike, T. Akuta, F. Tamura, T. Ogura, H. Esumi, H. Maeda, Nitric
[3] K.J. Davies, Protein modification by oxidants and the role of proteolytic enzymes, oxide as an endogenous mutagen for Sendai virus without antiviral activity,
Biochem. Soc. Trans. 21 (1993) 346–353. J. Virol. 78 (2004) 8709–8719.
[4] T. Lindahl, Instability and decay of the primary structure of DNA, Nature 362 [36] H. Kuwahara, A. Kanazawa, D. Wakamatu, S. Morimura, K. Kida, T. Akaike, H.
(1993) 709–751. Maeda, Antioxidative and antimutageneic activities of 4-vinyl-2,6-dimethox-
[5] B.A. Wagner, G.R. Buettner, C. Patrick Burns, Free radical-mediated lipid peroxida- yphenol (canolol) isolated from canola oil, J. Agric. Food Chem. 52 (2004)
tion in cells: oxidizability is a function of cell lipid bis-allyic hydrogen content, 4380–4387.
Biochemistry 33 (1994) 4449–4453. [37] H. Kuwahara, T. Kariu, J. Fang, H. Maeda, Generation of drug-resistant mutants of
[6] B. Demple, L. Harrison, Repair of oxidative damage to DNA: enzymology and helicobacter pylori in the presence of peroxynitrite, a derivative of nitric oxide at
biology, Annu. Rev. Biochem. 63 (1994) 915–948. pathophysiological concentration, Microbiol. Immunol. 53 (2009) 1–7.
[7] D.J. Hassett, H.P. Schweizer, D.E. Ohman, Pseudomonas aeruginosa sodA and sodB [38] W. Droge, Free radicals in the physiological control of cell function, Physiol. Rev.
mutants defective in manganese- and iron-cofactored superoxide dismutase 82 (2002) 47–95.
activity demonstrate the importance of the iron-cofactor from in aerobic [39] S.L. Camhi, J. Alam, G.W. Wiegand, B.Y. Chin, A.M.K. Choi, Transcriptional
metabolism, J. Bacteriol. 177 (1995) 6330–6337. activation of the HO-1 gene by lipopolysaccharide is mediated by 5′ distal
[8] O. Warburg, On the origin of cancer cells, Science 123 (1956) 309–314. enhancers: Role of reactive oxygen intermediates and AP-1, Am. J. Respir. Cell
[9] Y. Yoshii, T. Furukawa, H. Yoshii, T. Mori, Y. Kiyono, A. Waki, M. Kobayashi, T. Tsujikawa, Mol. Biol. 18 (1998) 226–234.
T. Kudo, H. Okazawa, Y. Yonekura, Y. Fujibayashi, Cytosolic acetyl-CoA synthetase [40] S.I. Kurata, M. Matsumoto, Y. Tsuji, H. Nakajima, Lipopolysaccharide activates
affected tumor cell survival under hypoxia: the possible function in tumor acetyl-CoA/ transcription of the heme oxygenase gene in mouse M1 cells through oxidative
acetate metabolism, Cancer Sci. (in press), doi:10.1111/j.1349-7006.2009.01099.x. activation of nuclear factor κB, Eur. J. Biochem. 239 (1996) 566–571.
[10] J.P. Greenstein (Ed.), Biochemistry of Cancer, 2nd ed., Academic Press, New York, [41] S.A. Rushworth, D.J. Macewan, HO-1 underlies resistance of AML cells to TNF-
1954. induced apoptosis, Blood 111 (2008) 3793–3801.
[11] Y. Hasegawa, T. Takano, A. Miyauchi, F. Matsuzuka, H. Yoshida, K. Kuma, N. Amino, [42] H. Ichijo, E. Nishida, K. Irie, P. ten Dijke, M. Saitoh, T. Moriguchi, M. Takagi, K.
Decreased expression of glutathione peroxidase mRNA in thyroid anaplastic Matsumoto, K. Miyazono, Y. Gotoh, Induction of apoptosis by ASK1, a mammalian
carcinoma, Cancer Lett. 182 (2002) 69–74. MAPKKK that activates SAPK/JUK and p38 signaling pathways, Science 275
[12] N. Yamanaka, D. Deamer, Superoxide dismutase activity in WI-38 cell cultures: (1997) 90–94.
effect of age, trypsinization and SV-40 transformation, Physiol. Chem. Phys. 6 [43] J. Matsukawa, A. Matsuzawa, K. Takeda, H. Ichijo, The ASK1-MAP kinase cascades
(1974) 95–106. in mammalian stress response, J. Biochem. 136 (2004) 261–265.
[13] K. Sato, K. Ito, H. Kohara, Y. Yamaguchi, K. Adachi, H. Endo, Negative regulation of [44] T. Fujisawa, K. Takeda, H. Ichijo, ASK family proteins in stress response and
catalase gene expression in hepatoma cells, Mol. Cell. Biol. 12 (1992) 2525–2533. disease, Mol. Biotechnol. 37 (2007) 13–18.
[14] T. Sawa, J. Wu, T. Akaike, H. Maeda, Tumor-targeting chemotherapy by a xanthine [45] T. Sawa, M.H. Zaki, T. Okamoto, T. Akuta, Y. Tokutomi, S. Kim-Mitsuyama, H. Ihara,
oxidase–polymer conjugate that generates oxygen-free radicals in tumor tissue, A. Kobayashi, M. Yamamoto, S. Fujii, H. Arimoto, T. Akaike, Protein S-guanylation
Cancer Res. 60 (2000) 666–671. by the biological signal 8-nitroguanosine 3′,5′-cyclic monophosphate, Nat. Chem.
[15] J. Fang, T. Sawa, T. Akaike, H. Maeda, Tumor-targeted delivery of polyethylene Biol. 3 (2007) 727–735.
glycol-conjugated D-amino acid oxidase for antitumor therapy via enzymatic [46] T. Oda, T. Akaike, T. Hamamoto, F. Suzuki, T. Hirano, H. Maeda, Oxygen radicals in
generation of hydrogen peroxide, Cancer Res. 62 (2002) 3138–3143. influenza-induced pathogenesis and treatment with pyran polymer-conjugated
[16] S. Simizu, M. Takada, K. Umezawa, M. Imoto, Requirement of caspase-3 (-like) SOD, Science 244 (1989) 974–976.
protease-mediated hydrogen peroxide production for apoptosis induced by [47] T. Akaike, M. Ando, T. Oda, T. Doi, S. Ijiri, S. Araki, H. Maeda, Dependence on O− 2
various anticancer drugs, J. Biol. Chem. 273 (1998) 26900–26907. generation by xanthine oxidase of pathogenesis of influenza virus infection in
[17] O. Ben-Yoseph, B.D. Ross, Oxidation therapy: the use of a reactive oxygen species- mice, J. Clin. Invest. 85 (1990) 739–745.
generating enzyme system for tumour treatment, Br. J. Cancer 70 (1994) [48] H. Maeda, T. Akaike, Oxygen free radicals as pathogenic molecules in viral
1131–1135. diseases, Proc. Soc. Exp. Biol. Med. 198 (1991) 721–727.
[18] T. Yoshikawa, S. Kokura, K. Tanaka, Y. Naito, M. Kondo, A novel cancer therapy [49] T. Akaike, H. Maeda, Nitric oxide and virus infection, Immunology 101 (2000)
based on oxygen radicals, Cancer Res. 55 (1995) 1617–1620. 300–308.
[19] L.D. Stegman, H. Zheng, E.R. Neal, O. Ben-Yoseph, L. Pollegioni, M.S. Pilone, B.D. [50] P.A. Cerutti, B.F. Trump. Inflammation and oxidative stress in carcinogenesis,
Ross, Induction of cytotoxic oxidative stress by D-alanine in brain tumor cells Cancer Cells 3 (1991) 1–7.
expressing Rhodotorula gracilis D-amino acid oxidase: a cancer gene therapy [51] J.M. McCord, The evolution of free radicals and oxidative stress, Am. J. Med. 108
strategy, Hum. Gene Ther. 9 (1998) 185–193. (2000) 652–659.
J. Fang et al. / Advanced Drug Delivery Reviews 61 (2009) 290–302 301

[52] D. Dreher, A.F. Junod, Role of oxygen free radicals in cancer development, Eur. J. [81] R. Igarashi, J. Hoshino, M. Takenaga, S. Kawai, Y. Morizawa, A. Yasuda, M. Otani,
Cancer 32A (1996) 30–38. Y. Mizushima, Lecithinization of superoxide dismutase potentiates its protective
[53] V.R. Muzykantov, E.N. Atochina, H. Ischiropoulos, S.M. Danilov, A.B. Fisher, effect against Forssman antiserum-induced elevation in guinea pig airway
Immunotargeting of antioxidant enzyme to the pulmonary endothelium, Proc. resistance, J. Pharmacol. Exp. Ther. 262 (1992) 1214–1219.
Natl. Acad. Sci. USA 93 (1996) 5213–5218. [82] T. Tsubokawa, V. Jadhav, I. Solaroglu, Y. Shiokawa, Y. Konishi, J.H. Zhang, Lecithinized
[54] B. Nieves-Cruz, A. Rivera, J. Cifuentes, G. Pataki, S. Matalon, W.A. Carlo, A.K. superoxide dismutase improves outcomes and attenuates focal cerebral ischemic
Tanswell, B. Freeman, Clinical surfactant preparations mediate SOD and catalase injury via antiapoptotic mechanisms in rats, Stroke 38 (2007) 1057–1062.
uptake by type II cells and lung tissue, Am. J. Physiol. 270 (1996) L659–667. [83] M. Takenaga, Y. Ohta, Y. Tokura, A. Hamaguchi, M. Nakamura, H. Okano, R. Igarashi,
[55] M. Takenaga, Y. Ohta, Y. Tokura, A. Hamaguchi, M. Nakamura, H. Okano, R. Lecithinized superoxide dismutase (PC-SOD) improved spinal cord injury-induced
Igarashi, Lecithinized superoxide dismutase (PC-SOD) improved spinal cord motor dysfunction through suppression of oxidative stress and enhancement of
injury-induced motor dysfunction through suppression of oxidative stress and neurotrophic factor production, J. Control. Release 110 (2006) 283–289.
enhancement of neurotrophic factor production, J. Control. Release 110 (2006) [84] S. Shimmura, R. Igarashi, H. Yaguchi, Y. Ohashi, J. Shimazaki, K. Tsubota, Lecithin-
283–289. bound superoxide dismutase in the treatment of noninfectious corneal ulcers,
[56] G.J. del Zoppo, S. Wagner, M. Tagaya, Trends and future developments in the Am. J. Ophthalmol. 135 (2003) 613–619.
pharmacological treatment of acute ischaemic stroke, Drugs 54 (1997) 9–38. [85] M. Premanathan, H. Nakashima, R. Igarashi, Y. Mizushima, K. Yamada,
[57] M.E. Anderson, Glutathione and glutathione delivery compound, Adv. Pharmacol. Lecithinized superoxide dismutase: an inhibitor of human immunodeficiency
38 (1996) 65–78. virus replication, AIDS Res. Hum. Retrovir. 13 (1997) 283–290.
[58] I. Cotgreave, N-Acetylcysteine: pharmacological considerations and experimental [86] M.D. Maines, Heme oxygenase: function, multiplicity, regulatory mechanisms,
and clinical applications, Adv. Pharmacol. 38 (1996) 205–227. and clinical applications, FASEB J. 2 (1988) 2557–2568.
[59] Y. Miyamoto, T. Akaike, M. Yoshida, S. Goto, H. Horie, H. Maeda, Potentiation of [87] K. Doi, T. Akaike, S. Fujii, S. Tanaka, N. Ikebe, T. Beppu, S. Shibahara, M. Ogawa,
nitric oxide-mediated vasorelaxation by xanthine oxidase inhibitors, Proc. Soc. H. Maeda. Induction of haem oxygenase-1 by nitric oxide and ischaemia in
Exp. Biol. Med. 211 (1996) 366–373. experimental solid tumours and implications for tumour growth, Br. J. Cancer
[60] J. Fang, T. Akaike, H. Maeda, Antiapoptotic role of heme oxygenase (HO) and the 80 (1999) 1945–1954.
potential of HO as a target in anticancer treatment, Apoptosis 9 (2004) 27–35. [88] S. Tanaka, T. Akaike, J. Fang, T. Beppu, M. Ogawa, F. Tamura, Y. Miyamoto, H. Maeda,
[61] Y. Matsumura, H. Maeda, A new concept for macromolecular therapeutics in Antiapoptotic effect of haem oxygenase-1 induced by nitric oxide in experimental
cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and solid tumours, Br. J. Cancer 88 (2003) 902–909.
the antitumor agent SMANCS, Cancer Res. 46 (1986) 6387–6392. [89] G. Balla, H.S. Jacob, J. Balla, M. Rosenberg, K. Nath, G. Apple, J.W. Eaton, G.M.
[62] H. Maeda, SMANCS and polymer-conjugated macromolecular drugs: advantages Vercellotti, Ferritin: a cytoprotective antioxidant strategem of endothelium,
in cancer chemotherapy, Adv. Drug Deliv. Rev. 46 (2001) 169–185. J. Biol. Chem. 267 (1992) 18148–18153.
[63] J. Fang, T. Sawa, H. Maeda, Factors and mechanism of “EPR” effect and the [90] T.S. Lee, L.Y. Chau, Heme oxygenase-1 medicates the anti-inflammatory effect of
enhanced antitumor effects of macromolecular drugs including SMANCS, Adv. interleukin-10 in mice, Nat. Med. 8 (2002) 240–246.
Exp. Med. Biol. 519 (2003) 29–49. [91] O. Tokola, R. Tenhunen, L. Volin, P. Mustajoki, Pharmacokinetics of intravenously
[64] T. Seki, J. Fang, H. Maeda, Tumor targeted macromolecular drug delivery based on administered haem arginate, Br. J. Clin. Pharmacol. 22 (1986) 331–335.
the enhanced permeability and retention effect in solid tumor, In: Pharmaceu- [92] R. Duncan, The dawning era of polymer therapeutics, Nat. Rev. Drug Discov. 2
tical Perspectives of Cancer Therapeutics, AAPS-Springer publishing (in press). (2003) 347–360.
[65] H. Maeda, G.Y. Bharate, J. Daruwalla, Polymeric drugs and nanomedicines for [93] A.K. Iyer, K. Greish, J. Fang, H. Maeda, Exploiting the enhanced permeability and
efficient tumor targeted drug delivery based on EPR-effect, Eur. J. Pharm. retention effect for tumor targeting, Drug Discov. Today 11 (2006) 812–818.
Biopharm. 71 (2009) 409–419. [94] L.J. Ignarro (Ed.), Nitric Oxide: Biology and Pathobiology, Academic Press, San
[66] H. Maeda, K. Greish, J. Fang, The EPR effect and polymeric drugs: a paradigm shift Diego, 2000.
for cancer chemotherapy in the 21st century, Adv. Polym. Sci. 193 (2006) 103–121. [95] R.F. Furchgott, P.M. Vanhoutte, Endothelium-derived relaxing and contracting
[67] T. Watanabe, M. Tanaka, K. Watanabe, Y. Takamatsu, A. Tobe, Research and factors, FASEB J. 3 (1989) 2007–2018.
development of the free radical scavenger edaravone as a neuroprotectant, [96] N. Ikebe, T. Akaike, Y. Miyamoto, K. Hayashida, J. Yoshitake, M. Ogawa, H. Maeda,
Yakugaku Zasshi 124 (2004) 99–111 (Japanese). Protective effect of S-nitrosated α1-protease inhibitor on hepatic ischemia–
[68] Edaravone Acute Infarction Study Group, Effect of a novel free radical scavenger, reperfusion injury, J. Pharmacol. Exp. Ther. 295 (2000) 904–911.
edaravone (MCI-186), on acute brain infarction. Randomized, placebo-con- [97] H. Rubbo, V. Darley-Usmar, B.A. Freeman, Nitric oxide regulation of tissue free
trolled, double-blind study at multicenters, Cerebrovasc. Dis. 15 (2003) 222–229. radical injury, Chem. Res. Toxicol. 9 (1996) 809–820.
[69] K.M. Channon, T.J. Guzik, Mechanisms of superoxide production in human blood [98] E.A. Konorev, M.M. Tarpey, J. Joseph, J.E. Baker, B. Kalyanaraman, S-nitrosoglu-
vessels: relationship to endothelial dysfunction, clinical and genetic risk factors, tathione improve functional recovery in the isolated rat heart after cardioplegic
J. Physiol. Pharmacol. 53 (2002) 515–524. ischemic arrest: Evidence for a cardioprotective effect of nitric oxide, J. Pharmacol.
[70] E.D. Jarasch, C. Grund, G. Bruder, H.W. Heid, T.W. Keenan, W.W. Franke, Exp. Ther. 274 (1995) 200–206.
Localization of xanthine oxidase in mammary-gland epithelium and capillary [99] Y. Miyamoto, T. Akaike, H. Maeda, S-Nitrosated human α1-protease inhibitor,
endothelium, Cell 25 (1981) 67–82. Biochim. Biophys. Acta 1477 (2000) 90–97.
[71] R.K. Robins, G.R. Revanker, D.E. O'Brein, R.H. Springer, T. Novinson, A. Albert, K. [100] Y. Ishima, T. Sawa, U. Kragh-Hansen, Y. Miyamoto, S. Matsushita, T. Akaike, M. Otagiri,
Senga, J.P. Miller, D.G. Streeter, Purine analog inhibitors of xanthine oxidase— S-Nitrosylation of human variant albumin Liprizzi (R410C) confers potent
structure activity relationships and proposed binding of the molybdenum antibacterial and cytoprotective properties, J. Pharmacol. Exp. Ther. 320 (2007)
cofactor, J. Heterocycl. Chem. 22 (1985) 601–634. 969–977.
[72] R.J. Gryglewski, R.M. Palmer, S. Moncada, Superoxide anion is involved in the [101] D. Wakamatu, S. Morimura, T. Sawa, K. Kida, C. Nakai, H. Maeda, Isolation,
breakdown of endothelium-derived vascular relaxing factor, Nature 320 (1986) identification, and structure of a potent alkyl–peroxyl radical scavenger in crude
454–456. canola oil, canolol, Biosci. Biotechnol. Biochem. 69 (2005) 1568–1574.
[73] R.E. Huie, S. Padmaja, The reaction of NO with superoxide, Free Radic. Res. [102] H. Kuwahara, A. Kanazawa, D. Wakamatu, S. Morimura, K. Kida, T. Akaike, H.
Commun. 18 (1993) 195–199. Maeda, Antioxidative and antimutageneic activities of 4-vinyl-2,6-dimethoxy-
[74] T. Akaike, M. Yoshida, Y. Miyamoto, K. Sato, M. Kohno, K. Sasamoto, K. Miyazaki, phenol (canolol) isolated from canola oil, J. Agric. Food Chem. 52 (2004)
S. Ueda, H. Maeda, Antagonistic action of imidazolineoxyl N-oxides against 4380–4387.
endothelium-derived relaxing factor/NO through a radical reaction, Biochem- [103] X. Cao, T. Tsukamoto, T. Seki, H. Tanaka, S. Morimura, L. Cao, T. Mizoshita, H. Ban, T.
istry 32 (1993) 827–832. Toyoda, H. Maeda, M. Tatematsu, 4-Vinyl-2,6-dimethoxyphenol (canolol) sup-
[75] J. Fang, A.K. Iyer, H. Nakamura, T. Seki, K. Greish, H. Maeda, SMA-copolymer conjugate presses oxidative stress and gastric carcinogenesis in Helicobacter pylori-infected
of AHPP: a polymeric inhibitor of xanthine oxidase with potential antihypertensive carcinogen-treated Mongolian gerbils, Int. J. Cancer 122 (2008) 1445–1454.
effect, J. Control. Release. (in press), doi:10.1016/j.jconrel.2009.01.006. [104] Y.J. Surh, Cancer chemoprevention with dietary phytochemicals, Nat. Rev. Cancer
[76] A. Rezaie, R.D. Parker, M. Abdollahi, Oxidative stress and pathogenesis of 3 (2003) 768–780.
inflammatory bowel disease: an epiphenomenon or the cause? Dig. Dis. Sci. 52 [105] A. Murakami, Y. Nakamura, K. Torikai, T. Tanaka, T. Koshiba, K. Koshimizu, S. Kuwahara,
(2007) 2015–2021. Y. Takahashi, K. Ogawa, M. Yano, H. Tokuda, H. Nishino, Y. Mimaki, Y. Sashida, S.
[77] K. Sato, T. Akaike, M. Kohno, M. Ando, H. Maeda, Hydroxyl radical production by Kitanaka, H. Ohigashi, Inhibitory effect of citrus nobiletin on phorbol ester-induced skin
H2O2 plus Cu,Zn-superoxide dismutase reflects the activity of free copper inflammation, oxidative stress, and tumor promotion in mice, Cancer Res. 60 (2000)
released from the oxidatively damaged enzyme, J. Biol. Chem. 267 (1992) 5059–5066.
25371–25377. [106] J. Fang, T. Sawa, T. Akaike, T. Akuta, S.K. Sahoo, K. Greish, A. Hamada, H. Maeda, In
[78] Y. Kojima, A. haruta, T. Imai, M. Otagiri, H. Maeda, Polymer conjugation to Cu, Zn– vivo antitumor activity of pegylated zinc protoporphyrin: targeted inhibition of
SOD and suppression of hydroxyl radical generation on exposure to H2O2: heme oxygenase in solid tumor, Cancer Res. 63 (2003) 3567–3674.
improved stability of SOD in vitro and in vivo, J. Bioact. Compat. Polym. 11 (1996) [107] S.K. Sahoo, T. Sawa, J. Fang, S. Tanaka, Y. Miyamoto, T. Akaike, H. Maeda, Pegylated
169–190. zinc portoporphyrin: a water-soluble heme oxygenase inhibitor with tumor-
[79] T. Ogino, M. Inoue, Y. Ando, M. Awai, H. Maeda, Y. Morino, Chemical modification targeting capacity, Bioconjug. Chem. 13 (2002) 1031–1038.
of superoxide dismutase. Extension of plasma half life of the enzyme through its [108] A.K. Iyer, K. Greish, J. Fang, R. Murakami, H. Maeda, High-loading nanosized micelles
reversible binding to the circulating albumin, Int. J. Pept. Protein Res. 32 (1988) of copoly(styrene-maleic acid)-zinc protoporphyrin for targeted delivery of a
153–159. potent heme oxygenase inhibitor, Biomaterials 28 (2007) 1871–1881.
[80] K. Kakimoto, Y. Kojima, K. Ishii, K. Onoue, H. Maeda, The suppressive effect of [109] A.K. Iyer, K. Greish, T. Seki, S. Okazaki, J. Fang, K. Takeshita, H. Maeda, Polymeric
gelatin-conjugated superoxide dismutase on disease development and severity micelles of zinc protoporphyrin for tumor targeted delivery based on EPR effect
of collagen-induced arthritis in mice, Clin. Exp. Immunol. 94 (1993) 241–246. and singlet oxygen generation, J. Drug Target. 15 (2007) 496–506.
302 J. Fang et al. / Advanced Drug Delivery Reviews 61 (2009) 290–302

[110] K. Yagi, Reaction mechanism of D-amino acid oxidase, Adv. Enzymol. Relat. Areas [119] R. Kondo, K.V. Gleixner, M. Mayerhofer, A. Vales, A. Gruze, P. Samorapoompichit, K.
Mol. Biol. 34 (1971) 41–78. Greish, M.T. Krauth, K.J. Aichberger, W.F. Pickl, H. Esterbauer, C. Sillaber, H. Maeda,
[111] J. Fang, H. Nakamura, D.W. Deng, T. Akuta, K. Greish, A.K. Iyer, H. Maeda, Oxystress P. Valent, Identification of heat shock protein 32 (Hsp32) as a novel survival factor
inducing antitumor therapeutics via targeted delivery of PEG-conjugated D-amino and therapeutic target in neoplastic mast cells, Blood 110 (2007) 661–669.
acid oxidase, Int. J. Cancer 122 (2008) 1135–1144. [120] E. Hadzijusufovic, L. Rebuzzi, K.V. Gleixner, V. Ferenc, B. Peter, R. Kondo, A. Gruze,
[112] J.M. Mates, C. Perez-Gomez, I. Nunez de Castro, Antioxidant enzymes and human M. Kneidinger, M.T. Krauth, M. Mayerhofer, P. Samorapoompichit, K. Greish, A.K.
diseases, Clin. Biochem. 32 (1999) 595–603. Iyer, W.F. Pickl, H. Maeda, M. Willmann, P. Valent, Targeting of heat-shock protein
[113] Y. Mochizuki, Z. Hruban, H.P. Morris, A. Slesers, E.L. Vigil, Microbodies of Morris 32/heme oxygenase-1 in canine mastocytoma cells is associated with reduced
hepatomas, Cancer Res. 31 (1971) 763–773. growth and induction of apoptosis, Exp. Hematol. 36 (2008) 1461–1470.
[114] K. Morikawa, H. Shimokawa, T. Matoba, H. Kubota, T. Akaike, M.A. Talukder, M. [121] M. Mayerhofer, K.V. Gleixner, J. Mayerhofer, G. Hoermann, E. Jaeger, K.J.
Hatanaka, T. Fujiki, H. Maeda, S. Takahashi, A. Takeshita, Pivotal role of Cu,Zn- Aichberger, R.G. Ott, K. Greish, H. Nakamura, S. Derdak, P. Samorapoompichit,
superoxide dismutase in endothelium-dependent hyperpolarization, J. Clin. W.F. Pickl, V. Sexl, H. Esterbauer, I. Schwarzinger, C. Sillaber, H. Maeda, P. Valent,
Invest. 112 (2003) 1871–1879. Targeting of heat shock protein 32 (Hsp32)/heme oxygenase-1 (HO-1) in
[115] H. Shimokawa, T. Matoba, Hydrogen peroxide as an endothelium-derived leukemic cells in chronic myeloid leukemia: a novel approach to overcome
hyperpolarizing factor, Pharmacol. Res. 49 (2004) 543–549. resistance against imatinib, Blood 111 (2008) 2200–2210.
[116] K. Schiwon, H.D. Brauer, B. Gerlach, C.M. Muller, F.P. Montforts, Potential [122] K.V. Gleixner, M. Mayerhofer, A. Vales, A. Gruze, W.F. Pickl, E. Lackner, C. Sillaber,
photosensitizers for photodynamic therapy. IV. Photophysical and photochemical C.C. Zielinski, H. Maeda, P. Valent, The Hsp32/HO-1-targeted drug SMA–ZnPP
properties of azaporphrin and azachlorin derivatives, J. Photochem. Photobiol., B counteracts the proliferation and viability of neoplastic cells in solid tumors and
Biol. 23 (1994) 239–243. hematologic neoplasms, J. Clin. Oncol. (Meet. Abstr.) 25 (2007) 14122.
[117] M. Regehly, K. Greish, F. Rancan, H. Maeda, F. Bohm, B. Roder, Water-soluble [123] M. Murakami, T. Hirano, Intracellular zinc homeostasis and zinc signaling, Cancer
polymer conjugates of ZnPP for photodynamic tumor therapy, Bioconjug. Chem. Sci. 99 (2008) 1515–1522.
18 (2007) 494–499. [124] J. Fang, H. Nakamura, A.K. Iyer, Tumor-targeted induction of oxystress for cancer
[118] J. Fang, T. Sawa, T. Akaike, K. Greish, H. Maeda, Enhancement of chemotherapeutic therapy, J. Drug Target. 15 (2007) 475–486.
response of tumor cells by a heme oxygenase inhibitor, pegylated zinc
protoporphyrin, Int. J. Cancer 109 (2004) 1–8.

You might also like