12
Tumor-targeted induction of oxystress for cancer therapy J. FANG 1,† , H. NAKAMURA 1 & A. K. IYER 1,2 1 Laboratory of Microbiology and Oncology, Faculty of Pharmaceutical Sciences, Sojo University, Ikeda 4-22-1, Kumamoto 860-0082, Japan, and 2 Department of Nanoscience, Faculty of Engineering, Sojo University, Ikeda 4-22-1, Kumamoto 860-0082, Japan (Received 21 February 2007; accepted 7 June 2007) Abstract Reactive oxygen species (ROS), such as superoxide anion radicals (O z2 2 ) and hydrogen peroxide (H 2 O 2 ) are potentially harmful by-products of normal cellular metabolism that directly affect cellular functions. ROS is generated by all aerobic organisms and it seems to be indispensable for signal transduction pathways that regulate cell growth and reduction – oxidation (redox) status. However, overproduction of these highly reactive oxygen metabolites can initiate lethal chain reactions, which involve oxidation and damage to structures that are crucial for cellular integrity and survival. In fact, many antitumor agents, such as vinblastine, cisplatin, mitomycin C, doxorubicin, camptothecin, inostamycin, neocarzinostatin and many others exhibit antitumor activity via ROS-dependent activation of apoptotic cell death, suggesting potential use of ROS as an antitumor principle. Thus, a unique anticancer strategy named “oxidation therapy” has been developed by inducing cytotoxic oxystress for cancer treatment. This goal could be achieved mainly by two methods, namely, (i) inducing the generation of ROS directly to solid tumors and (ii) inhibiting the antioxidative enzyme (defense) system of tumor cells. Since 1950s, many strategies have been employed based on the first method, namely, administration of ROS per se (e.g. H 2 O 2 ) or ROS generating enzyme to tumor bearing animals. However no successful and practical results were obtained probably because of the lack of tumor selective ROS delivery and hence resulting in subsequent induction of severe side effects. To overcome these obstacles, we developed polyethylene glycol (PEG) conjugated O z2 2 or H 2 O 2 -generating enzymes, xanthine oxidase (XO) and D-amino acid oxidase (DAO) (PEG–DAO) respectively. More recently, a pegylated (PEG) zinc protoporphyrin (PEG–ZnPP) and a highly water soluble micellar formulation of ZnPP based on amphiphilic styrene maleic acid (SMA) copolymer, SMA–ZnPP, are prepared, which are potent inhibitors of heme oxygenase-1 (HO-1). HO-1 is a major antioxidative enzyme of tumors, that is different in mechanism of catalase or superoxide dismutase (SOD). Consequently, both PEG-enzymes and PEG–ZnPP exhibited superior in vivo pharmacokinetics than their parental molecules, particularly in tumor delivery by taking advantage of the EPR effect of macromolecular nature, and thus showed remarkable antitumor effects suggesting the potentials of this anticancer therapeutic for clinical application. Furthermore, it has been well known that many antioxidative enzymes such as catalase, SOD are down-regulated in most solid tumors in vivo. On the contrary, HO-1 is highly upregulated and it plays a very important role of antioxidation, because HO-1 generates biliverdin, which being converted to bilirubin exhibits a very potent antioxidative effect, and hence antiapoptosis in tumors. Thus this oxidation therapy, by inhibiting this HO-1 dependent antioxidant (bilirubin) formation by ZnPP, and by enhancing ROS generation, is expected to offer a powerful therapeutic modality for future anticancer therapy. Keywords: Reactive oxygen species, EPR effect, D-amino acid oxidase, heme oxygenase-1 Introduction Reactive oxygen species (ROS) is potentially danger- ous by-product of cellular metabolism that has direct effect on cell development, growth, survival, aging, and on the development of cancer (Davies 1995; Sundaresan et al. 1995). ROS is a group of free radicals, which contain molecules with one or more unpaired electrons. Electron acceptors, primarily molecular oxygen, react easily with free radicals, to become oxygen free radicals, named ROS. In aerobic life, because molecular oxygen is ubiquitous, ROS became the primary mediators of cellular free radical reactions which are generated during the production ISSN 1061-186X print/ISSN 1029-2330 online q 2007 Informa UK Ltd. DOI: 10.1080/10611860701498286 Correspondence: J. Fang, Laboratory of Microbiology and Oncology, Faculty of Pharmaceutical Sciences, Sojo University, Ikeda 4-22-1, Kumamoto 860-0082, Japan. Tel: 81 96 326 4137. Fax: 81 96 326 5048. E-mail: [email protected] Journal of Drug Targeting, August–September 2007; 15(7–8): 475–486 Journal of Drug Targeting Downloaded from informahealthcare.com by University of Connecticut on 01/05/14 For personal use only.

Tumor-targeted induction of oxystress for cancer therapy

  • Upload
    a-k

  • View
    222

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Tumor-targeted induction of oxystress for cancer therapy

Tumor-targeted induction of oxystress for cancer therapy

J. FANG1,†, H. NAKAMURA1 & A. K. IYER1,2

1Laboratory of Microbiology and Oncology, Faculty of Pharmaceutical Sciences, Sojo University, Ikeda 4-22-1, Kumamoto

860-0082, Japan, and 2Department of Nanoscience, Faculty of Engineering, Sojo University, Ikeda 4-22-1, Kumamoto

860-0082, Japan

(Received 21 February 2007; accepted 7 June 2007)

AbstractReactive oxygen species (ROS), such as superoxide anion radicals (Oz2

2 ) and hydrogen peroxide (H2O2) are potentiallyharmful by-products of normal cellular metabolism that directly affect cellular functions. ROS is generated by all aerobicorganisms and it seems to be indispensable for signal transduction pathways that regulate cell growth and reduction–oxidation(redox) status. However, overproduction of these highly reactive oxygen metabolites can initiate lethal chain reactions, whichinvolve oxidation and damage to structures that are crucial for cellular integrity and survival. In fact, many antitumor agents,such as vinblastine, cisplatin, mitomycin C, doxorubicin, camptothecin, inostamycin, neocarzinostatin and many othersexhibit antitumor activity via ROS-dependent activation of apoptotic cell death, suggesting potential use of ROS as anantitumor principle. Thus, a unique anticancer strategy named “oxidation therapy” has been developed by inducing cytotoxicoxystress for cancer treatment. This goal could be achieved mainly by two methods, namely, (i) inducing the generation ofROS directly to solid tumors and (ii) inhibiting the antioxidative enzyme (defense) system of tumor cells. Since 1950s, manystrategies have been employed based on the first method, namely, administration of ROS per se (e.g. H2O2) or ROS generatingenzyme to tumor bearing animals. However no successful and practical results were obtained probably because of the lack oftumor selective ROS delivery and hence resulting in subsequent induction of severe side effects. To overcome these obstacles,we developed polyethylene glycol (PEG) conjugated Oz2

2 or H2O2-generating enzymes, xanthine oxidase (XO) and D-aminoacid oxidase (DAO) (PEG–DAO) respectively. More recently, a pegylated (PEG) zinc protoporphyrin (PEG–ZnPP) and ahighly water soluble micellar formulation of ZnPP based on amphiphilic styrene maleic acid (SMA) copolymer, SMA–ZnPP,are prepared, which are potent inhibitors of heme oxygenase-1 (HO-1). HO-1 is a major antioxidative enzyme of tumors, thatis different in mechanism of catalase or superoxide dismutase (SOD). Consequently, both PEG-enzymes and PEG–ZnPPexhibited superior in vivo pharmacokinetics than their parental molecules, particularly in tumor delivery by taking advantageof the EPR effect of macromolecular nature, and thus showed remarkable antitumor effects suggesting the potentials of thisanticancer therapeutic for clinical application. Furthermore, it has been well known that many antioxidative enzymes such ascatalase, SOD are down-regulated in most solid tumors in vivo. On the contrary, HO-1 is highly upregulated and it plays a veryimportant role of antioxidation, because HO-1 generates biliverdin, which being converted to bilirubin exhibits a very potentantioxidative effect, and hence antiapoptosis in tumors. Thus this oxidation therapy, by inhibiting this HO-1 dependentantioxidant (bilirubin) formation by ZnPP, and by enhancing ROS generation, is expected to offer a powerful therapeuticmodality for future anticancer therapy.

Keywords: Reactive oxygen species, EPR effect, D-amino acid oxidase, heme oxygenase-1

Introduction

Reactive oxygen species (ROS) is potentially danger-

ous by-product of cellular metabolism that has direct

effect on cell development, growth, survival, aging,

and on the development of cancer (Davies 1995;

Sundaresan et al. 1995). ROS is a group of free

radicals, which contain molecules with one or more

unpaired electrons. Electron acceptors, primarily

molecular oxygen, react easily with free radicals, to

become oxygen free radicals, named ROS. In aerobic

life, because molecular oxygen is ubiquitous, ROS

became the primary mediators of cellular free radical

reactions which are generated during the production

ISSN 1061-186X print/ISSN 1029-2330 online q 2007 Informa UK Ltd.

DOI: 10.1080/10611860701498286

Correspondence: J. Fang, Laboratory of Microbiology and Oncology, Faculty of Pharmaceutical Sciences, Sojo University, Ikeda 4-22-1,Kumamoto 860-0082, Japan. Tel: 81 96 326 4137. Fax: 81 96 326 5048. E-mail: [email protected]

Journal of Drug Targeting, August–September 2007; 15(7–8): 475–486

Jour

nal o

f D

rug

Tar

getin

g D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Con

nect

icut

on

01/0

5/14

For

pers

onal

use

onl

y.

Page 2: Tumor-targeted induction of oxystress for cancer therapy

of ATP by aerobic metabolism in mitochondria. The

leakage of electrons from mitochondria during the

electron-transport steps of ATP production generates

the ROS, such as superoxide anion radical (Oz22 ).

Superoxide radicals is converted to hydrogen peroxide

(H2O2) by SOD, from which further hydroxyl radicals

(zOH) are generated in a reaction that either depends

on or is catalysed by Fe2þ or Cu2þ ions. Cells have

evolved a series of antioxidative defense systems to

handle these dangerous naturally occuring highly

reactive by-products. These defenses include intra-

cellular superoxide dismutases (SODs), catalase and

glutathione peroxidase that inactivate H2O2, and

other enzymes or compounds that contribute to

scavenging free radicals (e.g. heme oxygenase-1, HO-

1 and glutathione; Figure 1).

Because of their extremely high reactivity, ROS are

potentially harmful, by reacting with and damaging

protein (Davies 1993) and DNA (Lindahl 1993).

Under physiological conditions, damages to oxidisable

molecules caused by ROS are reduced by cellular

antioxidative defenses (e.g., SOD, catalase, HO-1,

glutathione peroxidase) and repair mechanism (Dem-

ple and Harrison 1994). However, over-burden of

ROS, while under pathological conditions such as

acute and chronic inflammation (Oda et al. 1989;

Cerutti and Trump 1991; Maeda and Akaike 1991)

and deficiency of antioxidant defense or repair

systems, will initiate lethal chain reactions that involve

oxidation and damage of cellular integrity and survival

(Davies 1995), by which reversible or irreversible

tissue injury will be induced, consequently leading to

various diseases, including cancer (Maeda and Akaike

1991; Dreher and Junod 1996; McCord 2000).

With regard to ROS and cancer, the prevailing

studies are focused on their roles on carcinogenesis.

In the past ten years, convincing evidence has been

accumulated that ROS is indeed considered an

endogenous class of carcinogens (Guyton and Kensler

1993; Feig et al. 1994; Cerutti 1994). In addition,

ROS is involved in each stage of cancer development,

including initiation, promotion, and progression

(Dreher and Junod 1996). As mentioned above,

overproduction of ROS induces increased damage to

DNA, which further triggers mutagenesis via DNA

base modifications and DNA helix alterations (Dreher

and Junod 1996). Moreover, oxidative stress can

stimulate the expansion of mutated cell clones by

modulating genes related to proliferation and trigger-

ing redox-responsive signaling cascades such as

epidermal growth factor (EGF), tyrosine phosphoryl-

ation and protein kinase C (PKC) (Droge 2002).

In 1990s, Maeda’s group found that during the

course of viral and bacterial infections, nitric oxide

synthase (iNOS) is induced at the site of infection and

inflammation in the same time course as Oz22

generation (either by XO activation and NADPH

oxidation) (Akaike et al. 1990, 2000, 2003; Umezawa

et al. 1997; Fujii et al. 1999). These two reactive

radicals, i.e. NO and Oz22 , react immediately and more

reactive derivative, peroxynitrite (ONOO2) is gener-

ated from nitric oxide (NO) and Oz22 under

pathological conditions and also in cancer. ONOO2

plays important roles on tumor angiogenesis and

metastasis by activating matrix metalloproteinases

(MMPs) as well as damaging RNA (virus) and DNA

(Okamoto et al. 2001; Wu et al. 2001; Sawa et al.

2003). In Maeda’s group, Yoshitake et al. (2004)

showed increase of viral mutation (Sendai virus/mouse

system) under such condition of extensive ONOO2

generation. Antioxidant canalol, a natural product

obtained from crude rape seed oil prevented the

mutation of Salmonella (Ames test) by ONOO2

(Kuwahara et al. 2004).

The effect of ROS on mutation is dose-dependent,

and in a dynamic equilibrium with antioxidant

defenses and cellular repair systems (Dreher and

Junod 1996); their production is a double-edged

sword. While NO is antioxidant per se, ROS makes NO

the most reactive. While low or intermediate levels of

oxidative stress are most effective for triggering

mutagenesis and promoting proliferation of cells,

high levels of oxidative stress not only inhibit cell

proliferation, but it is more damaging and causes

cytotoxic effect (Dreher and Junod 1996).

On the contrary, it is quite intriguing and more

important that the activities of various antioxidative

enzymes, such as catalase, glutathione peroxidase,

SOD, etc. have been found to be greatly reduced in

various tumor cells (Greenstein 1954; Yamanaka and

Deamer 1974; Sato et al. 1992; Hasegawa et al. 2002),

which will increase the vulnerability of tumor cells

Figure 1. ROS metabolism and the antioxidant defense system in

cancer cells. Cancer cells have high levels of metabolism that

generate ROS, and the corresponding antioxidative systems (e.g.

superoxide dismutase, SOD; heme oxygenase, HO-1) are essential

for cancer cells to fight against cytotoxic ROS. Thus, a new

antitumor strategy using potentially toxic ROS was challenged, both

by directly inducing ROS (O22 , H2O2) to tumors, and by targeting

these antioxidative enzymes.

J. Fang et al.476

Jour

nal o

f D

rug

Tar

getin

g D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Con

nect

icut

on

01/0

5/14

For

pers

onal

use

onl

y.

Page 3: Tumor-targeted induction of oxystress for cancer therapy

to ROS (Sawa et al. 2000; Fang et al. 2002). Thus,

one can imagine a unique antitumor tactics by

selectively delivering excess oxidative stress to tumor

cells, or preferentially disrupting the antioxidative

defense systems of tumor cells. Indeed, several

challenges were performed and verified the effect by

use of this concept of “oxidation therapy”, by

modulating the redox status in tumor cells (Ben-

Yoseph and Ross 1994; Yoshikawa et al. 1995;

Stegman et al. 1998; Huang et al. 2000). However,

the crucial problem of the concept is that specific and

tumor selective generation of ROS is not clearly

solved. To solve this problem, in Maeda’s laboratory

we developed a series of ROS-generating agents with

tumor-targeting nature, all of which showed potent

and satisfactory in vivo anticancer effects with little

side effect, suggesting a potential for this new

anticancer drug development (Sawa et al. 2000;

Fang et al. 2002, 2003a,b, 2004a,b, 2007). In this

review, the rationales of basic concept, and develop-

ment of tumor selective oxidation therapy, in which

EPR-effect based drug delivery system (DDS), with

high therapeutic efficacy were undertaken. Also, some

candidate agents are introduced.

History and development of oxidation therapy:

ROS generating system

During 1950s–1970s, researchers from different

laboratory tried to use H2O2, a highly cytotoxic and

relative stable ROS, for treating tumors in various

animal tumor models by injecting H2O2 into tumor or

into the circulation (Green and Westrop 1958; Sugiura

1958; Mealey 1965; Kaibara et al. 1971). Even though

no satisfactory antitumor effect was observed, they

became the pioneer of the ROS induced anticancer

therapeutic. This therapy was greatly developed in

1980s by Nathan and Cohn (1981), in which they

utilized glucose oxidase (GO), a H2O2 generating

enzyme, as a ROS donor, resulting in a significant

antitumor effect in a mice tumor model. This GO

method was further optimized and the concept of

oxidation therapy was first introduced by Ben-Yoseph

and Ross (1994). However, these researches were

unable to advance further because of the possible side

effects caused by the systemic generation of ROS, due

to ubiquitous presence of D-glucose.

Strikingly, utilization of D-amino acid oxidase

(DAO) as a ROS (H2O2) generator was a break-

through for this therapy because the production of

ROS can be easily controlled by controlling infusion of

substrate, D-amino acids (Stegman et al. 1998; Fang

et al. 2002, 2007). Besides H2O2, another ROS,

superoxide (Oz22 ) has also attracted considerable

attention. Oz22 generating enzyme xanthine oxidase

(XO) was thus developed and exhibited promising

results as a new anticancer agent (Yoshikawa et al.

1995; Sawa et al. 2000). Meantime, an alternative

strategy by inhibiting the antioxidative defense system

was verified effective by using SOD as a target for

selective killing of cancer cells (Huang et al. 2000).

However, one problem which remained unsolved was

how to target and keep the enzyme in the tumor. This

problem could be best solved by adapting macromol-

ecular drugs, polymeric micelles or nanoparticles

which exhibits EPR-effect in solid tumors.

In our laboratory, we observed that HO-1 is one of

the most potent antioxidative defense in tumor cells,

and thus we developed a series macromolecular

derivatives of HO-1 inhibitor zinc protoporphyrin

(ZnPP) for this goal and found remarkable anticancer

potentials of these agents (Fang et al. 2003a,b; Iyer

et al. 2007). The project for the oxidation therapy by

using polymeric DAO and ZnPP is still ongoing in our

laboratory, and these agents are expected to become

novel anticancer drugs in the future.

Key factors affecting the oxidation therapy:

Targeted generation of ROS in tumor

Choice of ROS-generating system

It is crucial for the oxidation therapy to select the

appropriate ROS and ROS-generating system. As is

well known, there are many ROS with potent

cytotoxicity, such as H2O2 in a broad sense, Oz22 ,

ONOO2 and HClO (both are products of ROS), zOH,

and lipid peroxylradical (LOOz) etc. Among these

ROS, H2O2 is a desirable candidate for this

therapeutic strategy because of its relatively mild

cytotoxic activity, which permits it more stability in

contrast to zOH radicals. Normally, H2O2 readily

crosses cell membrane. Ultimately, it causes oxidative

damage to DNA (Beckman and Ames 1997), proteins

(Berlett and Stadtman 1997), and lipids by direct

oxidation or in the presence of transition metal via

Haber–Weiss reaction to the extremely reactive

hydroxyl radical (Halliwell and Gutteridge 1984).

It was also reported more recently that H2O2 induces

apoptosis of a wide range of tumor cells in vitro

(Bladier et al. 1997; Suhara et al. 1998; Matsura et al.

1999; Yamakawa et al. 2000, Ren et al. 2001) via

activation of the caspase cascade. Of greater import-

ance, many antitumor agents, such as vinblastine,

doxorubicin, camptothecin, and inostamycin, exhibit

antitumor activity via H2O2-dependent activation of

apoptotic cell death, which suggests potential use of

H2O2 as an antitumor principle (Simizu et al. 1998).

However, H2O2 is relatively stable and small water-

soluble molecule, and thus diffuse or travel a long

distance and thus make it difficult to selective delivery

to and retain in tumor tissue. These characteristics

hamper the utility of H2O2 as an antitumor agent.

In fact, H2O2 used alone was ineffective when injected

into tumor or into the circulation (Green and

Westrop 1958; Sugiura 1958; Mealey 1965; Kaibara

et al. 1971), because of the aforesaid reasons. Further

Tumor-targeted induction of oxystress for cancer therapy 477

Jour

nal o

f D

rug

Tar

getin

g D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Con

nect

icut

on

01/0

5/14

For

pers

onal

use

onl

y.

Page 4: Tumor-targeted induction of oxystress for cancer therapy

it will be decomposed by catalase in erythrocytes

which exist in massive levels. Use of an H2O2-

generating enzyme has been proposed as an alternative

approach to developing an H2O2-dependent antitu-

mor system. Nathan and Cohn (1981) and Ben-

Yoseph and Ross (1994) reported that GO, which

generates H2O2 during oxidation of glucose, showed

antitumor activity in solid tumor models. However,

regulation of H2O2 production by exogenously

administered GO in tumor-bearing hosts is proble-

matic because the availability of its both substrates,

oxygen and glucose, cannot be effectively modulated.

Consequently, induction of severe systemic side

effects is anticipated. In fact, GO administration to

produce H2O2 required injection of antioxidants to

minimize systemic toxicity (Nathan and Cohn 1981;

Ben-Yoseph and Ross 1994).

To overcome this drawback, in our laboratory, we

selected another H2O2-generating enzyme, DAO, by

which the generation of H2O2 can be easily modulated

because of the scarcity of its natural substrate, D-

amino acids, in mammalian organisms (Konno and

Yasumura 1992). Therefore, as anticipated, the

preferential production of H2O2 more selectively in

tumor was accomplished, and thus efficient antitumor

activity was successfully achieved with little toxicity.

Besides H2O2, Oz22 is another good candidate for

this therapeutic strategy. It is well known that Oz22

plays an important role in the pathogenesis of various

diseases (McCord 2000; Akaike et al. 1990),

suggesting that it is a relatively active molecule with

highly cytotoxic nature. Accordingly, the Oz22 generat-

ing enzyme XO was also examined for the oxidation

therapy (Yoshikawa et al. 1995; Sawa et al. 2000).

Further, PEG–XO exhibited superior antitumor

activity without much toxicity (Sawa et al. 2000).

Antioxidative conditions of tumor

Another important approach for therapeutic maneu-

ver is to inhibit antioxidative systems, such as catalase,

SOD, glutathione peroxidase, and HO-1 that are

known to serve as the protective role against ROS in

normal cells. It is well known that tumor cells or

tissues adapt to anaerobic energy production system

(Warburg effect), which will lead to lesser oxygen

tension and oxystress in vivo. In any event, the defense

system against oxystress will be potentially protective

for tumor cells. Interestingly and fortunately, com-

pared with normal cells, these antioxidative enzyme

level was found extremely low, perhaps with an

exception of HO-1, instead it was upregulated in most

tumor cells. Thus HO-1 is the key for antioxidant

stress in tumor cells (Greenstein 1954; Yamanaka and

Deamer 1974; Sato et al. 1992; Doi et al. 1999;

Hasegawa et al. 2002; Tanaka et al. 2003). This means

suppression of HO-1 will greatly increase the

vulnerability of tumor cells to ROS. Among these

enzymes, catalase plays a central role in the

antioxidant defense of many healthy organisms, as

do other enzymes, as H2O2 is converted to H2O

(Mates et al. 1999). With a few exception in some

hepatomas (Mochizuki et al. 1971), most tumors has

been reported to show a significantly reduced catalase

activity (Greenstein 1954; Sato et al. 1992). This

might be due to a marked suppression of catalase gene

expression at the level of transcription (Sato et al.

1992). In this context, similar findings were observed

in our recent study by measuring the catalase activities

of three different solid tumors and the corresponding

normal tissues, i.e. liver, kidney and lung, in

comparison with both transplanted and chemically

induced tumor models, namely tumors showed a

decrease of catalase activity to less than 10%

compared to the normal tissues (Fang et al. 2007).

Comparison of the catalase activity between normal

tissues and tumors was summarized in Table I. This

tendency of low catalase level, however, is not

necessarily valid in tumor cells in culture.

On the other hand, many tumor tissues seem to

have extensive infiltration of leukocytes compared

with normal tissues, which might typically lead to

increased production of ROS. These leukocytes

(macrophage) in solid tumors comprises a part of

host defense response, and they generate ROS against

tumor cells. Therefore, the antioxidative systems, e.g.

HO-1, catalase, SOD, become much important in

tumor for defending against ROS in and around

tumor. Along similar context, cancer cells may be

damaged when these antioxidative enzymes are

inhibited. This hypothesis has been validated to be

practicable by Huang et al. (2000) via inhibiting SOD

by certain oestrogen derivatives in tumor cells. We

further verified the antitumor effect by inhibitng HO-

1 (Fang et al. 2003a,b), which will be discussed more

in details later in this review.

Table I. Comparison of catalase activities between tumors and normal tissues.

S180* B16* DMBA induced tumor* AH66† Ac2F† HepG2† Human hepatoma

Relative CAT act (%)‡ 0.95 2.59 6.87 0.80 1.77 3.37 ND{

See reference Fang et al. (2007) and Sato et al. (1992) for details; * S180, mouse sarcoma S180 tumor; B16, mouse B16 molenoma; DMBA

induced tumor, DMBA(7, 12-dimethylbenz (a) anthracene) induced rat breast cancer; † hepatoma cells; ‡ compared with the catalase activity

of mice or rat liver. CAT, catalase; {ND, not detected.

J. Fang et al.478

Jour

nal o

f D

rug

Tar

getin

g D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Con

nect

icut

on

01/0

5/14

For

pers

onal

use

onl

y.

Page 5: Tumor-targeted induction of oxystress for cancer therapy

Tumor-targeted generation of ROS based on EPR effect

Furthermore, in the oxidation therapy, to achieve

satisfactory antitumor effect, targeted delivery of ROS

to tumor tissues is indispensable for this strategy

because nonspecific production of ROS may cause

oxidative stress-induced systemic damage in normal

organs even though they have a high level of

antioxidation defense system such as catalase or

SOD. The concept of the EPR (enhanced per-

meability and retention) effect (Iwai et al. 1984;

Matsumura and Maeda 1986; Maeda 2001; Maeda

et al. 2001; Duncan 2003; Fang et al. 2003a,b; Greish

et al. 2003; Iyer et al. 2006) is also necessary, for the

delivery of ROS. Since the EPR effect is observed only

for macromolecules, polymeric nanoparticles and

lipids in most of solid tumors, ROS generating

macromolecules (i.e. the enzymes conjugated to

polymers) are thus ideal to be utilized in most solid

tumors (Figure 2).

The EPR effect is a complex phenomenon invol-

ving: (i) extensive angiogenesis and hence high

vascular density; (ii) extensive extravasation (vascular

permeability) induced by various vascular mediators,

such as bradykinin, NO, vascular permeability factor

(VPF)/vascular endothelial growth factor (VEGF),

prostaglandins involving cyclooxygenases and matrix

metalloproteinases (MMPs/collagenases); (iii) defec-

tive vascular architecture; and (iv) impaired lymphatic

clearance from the interstitial space of tumor tissues

(Iwai et al. 1984; Matsumura and Maeda 1986;

Maeda 2001; Maeda et al. 2001; Fang et al. 2003a,b;

Iyer et al. 2006). As a result of EPR effect,

biocompatible macromolecules and lipids preferen-

tially and spontaneously leak out of tumor vessels, and

remain there at high concentration for a long period of

time. This means it is more than passive targeting

because retention plays a crucial role. Further, the

molecular size-dependency of EPR effect is applicable

for the molecular size above 40 kDa (Matsumura and

Maeda 1986; Seymour et al. 1995; Noguchi et al.

1998). This dependency exhibits a reverse correlation

to the renal clearance of the compounds (Maeda et al.

2001). Therefore, EPR effect is observed for any

biocompatible macromolecule with a molecular size

larger than the renal excretion threshold. EPR effect

occurring in solid tumors is a universal phenomenon,

which has now been considered a “gold standard” in

the field of drug design for new anticancer drug

development (Muggia 1999; Maeda et al. 2001;

Duncan 2003; Vicent and Duncan 2006).

Poly(ethylene glycol) (PEG), one of the best

biocompatible polymers, is widely used for polymeric

drug preparation because of the favourable properties

such as increased solubility both in organic and

aqueous media, increased in vivo half-life, and

virtually no toxicity and immunogenicity, non-

biodegradability and easy excretion from body

Figure 2. Scheme of enhanced permeability and retention (EPR)-effect of macromolecules in solid tumor. For low molecular weight agents,

they distribute indiscriminately in normal and tumor tissues, and the concentration in each tissue is paralleled with the concentration in

circulation. Namely, the drug concentration in tumor drops quickly in parallel to the clearance of the drugs from blood. For high molecular

weight agents, they cannot enter normal tissues because of the big size, but will distribute in tumor tissue due to the anatomic and pathological

difference of tumor blood vessels. Moreover, they will retain in tumor tissues for long time because of the lack of lymphatic function in tumor

tissue. See text for details.

Tumor-targeted induction of oxystress for cancer therapy 479

Jour

nal o

f D

rug

Tar

getin

g D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Con

nect

icut

on

01/0

5/14

For

pers

onal

use

onl

y.

Page 6: Tumor-targeted induction of oxystress for cancer therapy

(Zalipsky 1995). Thus, it has been used as a covalent

modifier of a wide range of proteins/enzymes and

other drugs, to produce PEG conjugates (Harris

1992; Pasut et al. 2004). PEG-ademasew and PEG-

aspargasew are used for therapy of the severe

combined immunodeficiency disease and leukemia,

respectively (Duncan 2003; Iyer et al. 2006).

Pharmakokinetic superiority of PEG-interferon over

native interferon has almost replaced the use of native

interferon therapy (Duncan 2003; Vicent and Duncan

2006). Moreover, PEG–doxorubicin and PEG–

camptothecin have been reported to exhibit better

pharmacokinetics, tumor accumulation and in vivo

half-life compared with their native counterparts.

Further, reduction of antigenicity of the native agents

is possible (Harris and Chess 2003).

Similarly, for targeted delivery of ROS generating

system to tumor, we modified DAO and XO with

PEG, and conjugated a potent HO-1 inhibitor, ZnPP

to PEG. All these PEG conjugates showed superior

pharmacological characteristics to native proteins or

drugs (Sawa et al. 2000; Fang et al. 2002; Sahoo et al.

2002; Fang et al. 2003a,b). For example, pegylation

of ZnPP greatly increased the water-solubility of ZnPP,

which makes the clinical application of ZnPP possible.

In addition it exhibited macromolecular nature, thus

EPR-effect for tumor targeting could be anticipated.

Further significant extension of plasma half-life (t1/2)

and area under the concentration time curve (AUC)

could be achieved (Figure 3 A,B; Fang et al. 2003a,b).

Likewise, PEG–DAO exhibited tumor-selective

accumulation and increased plasma t1/2 after i.v.

injection to solid tumor-bearing mice (Figure 3C;

Fang et al. 2002). We also anticipate PEG-interferon

(Pegasus, MW 58 kDa) would show the EPR-effect in

solid tumor, but it is unknown in our knowledges.

Examples of oxystress induced anticancer

therapeutics

Poly(ethylene glycol) conjugated xanthine oxidase

(PEG–XO)

XO is an iron-containing metalloflavoprotien that

catalyzes the oxidation of purines, in which ROS

including superoxide anion radical (O22 ) and hydrogen

Figure 3. Pharmacokinetics of PEG–ZnPP and PEG–DAO. A and B show the plasma level and tumor accumulation of PEG–ZnPP

respectively; C shows the body distribution of PEG–DAO. Results are expressed as means; bars, ^ SE (n ¼ 3–4). *P , 0.01, PEG–ZnPP vs.

native ZnPP. See references for details (Fang et al. 2002; Fang et al. 2003a,b).

J. Fang et al.480

Jour

nal o

f D

rug

Tar

getin

g D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Con

nect

icut

on

01/0

5/14

For

pers

onal

use

onl

y.

Page 7: Tumor-targeted induction of oxystress for cancer therapy

peroxide (H2O2) are generated. Thus XO was

expected as a good candidate for oxystress induced

anticancer therapeutic and was reported in 1995

(Yoshikawa et al. 1995). However, the high binding

affinity of native XO to the wall of blood vessels would

cause systemic vascular damage and hence limits the

use of native XO in clinical settings (Yoshikawa et al.

1995). To overcome this drawback, and to achieve

selective delivery of the drug to tumor by EPR effect,

we converted XO to macromolecular nature with high

biocompatibility by conjugating with PEG (PEG–

XO) (Sawa et al. 2000). The 1-amino groups of lysine

residues of XO, which play a crucial role in its binding

to the vascular wall, was modified by PEG, in this case

49% of free amino groups were modified with PEG.

Pharmacokinetic study by using 125I-labeled deriva-

tives showed a 2.8-fold higher accumulation of PEG–

XO in solid tumor than that of native XO at 24 h after

administration, whereas only a slight or negligible

increase in accumulation of PEG–XO was found in

normal organs. Moreover, 24 h after i.v. injection of

PEG–XO to tumor-bearing mice, the highest PEG–

XO enzyme activity was detected in tumor compared

with normal organs expect blood; enzyme activity in

tumor was 5.0, 3.9, and 9.4 times higher than that in

liver, kidney, and spleen, respectively, and the

accumulation of PEG–XO in tumor retained for at

least 96 h. Administration of hypoxanthine, a sub-

strate of XO, via i.p. route 12 h after the injection of

PEG–XO resulted in significant suppression of tumor

growth, with no tumor growth even after 52 days

(Figure 4A). No or very little side effect, if any at all,

was observed after this treatment. These findings

suggest the validity of PEG–XO as a tumor tropic

novel anticancer agent.

Poly(ethylene glycol) conjugated D-amino acid oxidase

(PEG–DAO)

Along with PEG–XO, we explored another ROS-

generating enzyme, DAO, by PEG conjugation (Fang

et al. 2002). DAO is a flavoprotein that catalyzes the

stereoselective oxidative deamination of D-amino

acids to the corresponding a-keto acids. During this

oxidation reaction, molecular oxygen (O2) is used as

an electron acceptor, and H2O2 is generated (Yagi

Figure 4. Antitumor effect of PEG–XO, PEG–DAO, PEG–ZnPP, and PEG–ZnPP plus PEG–DAO in the S-180 tumor model. S-180 cells

(2 £ 106 cells) were implanted s.c. in ddY mice. Six to ten days after inoculation, mice were treated by each agents as indicated. A, PEG–XO

treatment; B, PEG–DAO treatment; C, PEG–ZnPP treatment; D, PEG–ZnPP plus PEG–DAO treatment. Data are means (n ¼ 4–8); bars,

SE. *P , 0.001. See references for details (Sawa et al. 2000; Fang et al. 2002, 2003a,b, 2004b).

Tumor-targeted induction of oxystress for cancer therapy 481

Jour

nal o

f D

rug

Tar

getin

g D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Con

nect

icut

on

01/0

5/14

For

pers

onal

use

onl

y.

Page 8: Tumor-targeted induction of oxystress for cancer therapy

1971). The natural substrate of DAO is D-amino

acids, with the exception of D-aspartic acid and

D-glutamic acid. Since D-amino acids do not usually

exist in mammalian organisms to a significant level

(Konno and Yasumura 1992), it is thus reasonable

that H2O2 generation can be regulated by the level of

exogenous administration of D-amino acids, thus

avoiding the possible induction or severe systemic side

effects because of systemic generation of H2O2.

However, DAO is relatively small protein (Mw

39 kDa), and it may be excreted gradually as

previously observed for other small proteins or

polymer drugs smaller than 40 kDa (Matsumura and

Maeda 1986; Seymour et al. 1995; Fang et al.

2003a,b). Pegylation of DAO resulted in a 63 kDa

molecule by calculating the number of amino groups

of DAO conjugated with PEG (Fang et al. 2002).

However, the apparent molecular size of PEG–DAO

in solution was determined to be 120–150 kDa by

SDS-PAGE analysis, while it existed as a micellar

form in aqueous solution with a mean hydrodynamic

diameter of 119 nm as determined by dynamic light

scattering studies (Fang et al. 2007). Accordingly, the

pharmacokinetic parameters after i.v. injection was

significantly improved in mice compared with the

native DAO; namely, its plasma t1/2 and AUC

increased 2.6- and 2.9-fold, respectively. Further-

more, PEG conjugated DAO dramatically improved

intratumoral accumulation as well as plasma level, i.e.

3.2-fold relative to native one and 7.4-fold to

untreated control in tumor (Figure 3C). In contrast,

PEG–DAO injection showed no increase on the

enzyme activity in normal organs and tissues.

Experiments in mice showed that administration of

PEG–DAO followed by administration of its substrate

D-proline, significantly suppressed tumor growth.

Growth suppression continued for at least 27 days

after tumor implantation (Figure 4B). In contrast, no

significant antitumor effect was observed in mice

treated with native DAO plus D-proline. In addition,

oxidative metabolites were significantly increased in

solid tumor by administration of PEG–DAO followed

by D-proline, as evidenced by thiobarbituric acid-

reactive substance (TBARS) assay, whereas it was not

increased in the liver and the kidney (Fang et al.

2002). These results strongly indicate the antitumor

potential of PEG–DAO, by selective generation of

H2O2 in tumor warrants further investigation towards

clinical application.

PEG conjugated zinc protoporphyrin IX (PEG–ZnPP)

Second method of oxidation therapy is a disruption of

the defense system against oxidation in tumor. For this

purpose we developed PEG conjugates of ZnPP,

which is a strong inhibitor of HO (Sahoo et al. 2002).

HO plays a key role in heme degradation: oxidation of

the porphyrin ring results in formation of biliverdin,

which is subsequently reduced by biliverdin reductase

yielding bilirubin, carbon monoxide, and free iron. All

three of them showed potent antioxidative effects

(Fang et al. 2004a). To date, three isoforms of

mammalian HO have been identified: HO-1, HO-2,

and HO-3, of which HO-1 is belongs to the family of

heat shock proteins (i.e. HSP32) and its expression is

triggered by diverse stress-inducing stimuli (Maines

et al 1988; Fang et al. 2004a; Figure 5).

HO-1 is suggested to serve as a key biological

molecule in the adaptation to and/or defense against

oxidative stress and cellular stress. More important,

it is interesting to note that several tumors, including

renal cell carcinoma (Goodman et al. 1997) and

Figure 5. Schematic representation of role of HO-1 in tumor growth and mechanism of PEG–ZnPP induced antitumor effect.

J. Fang et al.482

Jour

nal o

f D

rug

Tar

getin

g D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Con

nect

icut

on

01/0

5/14

For

pers

onal

use

onl

y.

Page 9: Tumor-targeted induction of oxystress for cancer therapy

prostate tumors (Maines and Abrahamsson 1996) in

humans, express a high level of HO-1. The high

HO-1 expression was also found in experimental

solid tumors, i.e. the rat hepatoma AH136B (Doi

et al. 1999) and the mouse sarcoma S180 (Sahoo

et al. 2002; Fang et al. 2003a,b). Administration of

the HO inhibitor zinc protoporphyrin (ZnPP), via

tumor-feeding artery significantly suppressed the

growth of AH136B tumors, which suggests a vital

role for HO-1 in tumor growth (Doi et al. 1999;

Tanaka et al. 2003). These findings also indicate a

potentially beneficial role of HO inhibitor, e.g. ZnPP

as a novel anticancer agent (Figure 5). However,

ZnPP is sparsely soluble in water, but may be

dissolved in non-physiological alkaline solution at

high pH, which limits its practical use as drug. To

solve this problem and to obtain better tumor-

targeting effect, PEG conjugation was carried out to

modified ZnPP, resulting in a highly water-soluble

compound PEG–ZnPP (Sahoo et al. 2002). More

importantly, pegylation of ZnPP greatly increased

the molecular size of ZnPP, from about 600 Da of

free ZnPP to more than 70 kDa of the PEG-

conjugates in aqueous media (Sahoo et al. 2002).

In vivo pharmacokinetic analysis revealed that PEG–

ZnPP administered i.v. had a circulation time that

was 40 times longer than that for nonconjugated

ZnPP (Figure 3A). More important, PEG–ZnPP

preferentially accumulated in solid tumor tissue in a

murine model (Figure 3B). In vivo treatment with

PEG–ZnPP remarkably suppressed the growth of

S180 tumors implanted in the dorsal skin of ddY

mice without any apparent side effects (Figure 4C).

In addition, this PEG–ZnPP treatment

produced tumor-selective suppression of HO activity

as well as induction of apoptosis (Fang et al.

2003a,b). These findings suggest that tumor-

targeted inhibition of HO by PEG–ZnPP will lead

to apoptosis in solid tumors, probably through

increased oxidative stress.

Furthermore, when PEG–ZnPP is combined with

ROS or ROS generating agents, additive or synergis-

tic effect on ROS-induced tumor cell killing is

envisaged. For instance, PEG–ZnPP treated

SW480 cells became vulnerable to insults caused by

various oxidative cytotoxic agents; the 50% inhibition

does (IC50) were reduced to 75, 61, 17 and 39% for

H2O2, t-butyl hydroperoxide, camptothecin and

doxorubicin, respectively. Cells treated with PEG–

ZnPP plus cytotoxic oxidants exhibited marked

production of intracellular ROS, which paralleled

the incidence of apoptosis. In vivo, PEG–ZnPP

pretreatment significantly suppressed the tumor

growth in mice receiving PEG–DAO/D-proline

compared to no PEG–ZnPP pretreatment

(Figure 4D; Fang et al. 2004b). These findings

suggest a definit potential of HO-1 be an attractive

target for chemotherapeutic intervention, and thus

the potential of PEG–ZnPP as a novel anticancer

drug by itself or in combination therapy.

Similar to PEG–ZnPP, we successfully prepared a

highly water-soluble micellar form of ZnPP recently,

by the use of amphiphilic styrene-maleic acid

copolymer (SMA), named SMA–ZnPP. SMA–

ZnPP existed as nanoparticles in aqueous solution

having a average hydrodynamic diameter of 176.5 nm

with relatively narrow size distribution profile. Static

light scattering and size exclusion chromatography

revealed that SMA–ZnPP micelles were associated to

form macromolecular complexes. The molecular size

of the SMA–ZnPP micelles was of the order of

144 kDa as determined by Sephacryl S-200 gel

chromatography. Thus the macromolecular nature of

these micelles may take advantage of EPR effect to

target tumor tissues selectively. SMA–ZnPP exhibited

similar HO inhibitory activity as that of native ZnPP

indicating its potential as a potent HO-1 inhibitor with

tumor targeting characteristics (Iyer et al. 2007).

From the preliminary in vitro and in vivo studies, we

found that SMA–ZnPP revealed a potent antitumor

effect, but without any apparent side effects even at

the dose of 200 mg/kg when injected i.v. in mice

(Iyer et al. 2007; Greish et al. unpublished data).

Thus we anticipate a pronounced therapeutic benefit

of PEG–ZnPP and SMA–ZnPP against tumor,

especially in combination with ROS-generating

systems including traditional antitumor agents.

Conclusion

ROS, a group of highly reactive molecules which

exhibit very important, yet paradoxical role on tumor.

On one hand, it can damage DNA, and thus initiate

and promote cancer development, yet on the other

hand it can serve as a tumor terminator if it is present

in excessive concentration due to its cytotoxic effect.

Moreover, although being not described here, the

highly elevated level of NO production in tumor will

further facilitate the antitumor effects of ROS by

generating more reactive species such as ONOO2,

which will be a combined effector in this anticancer

tactics. Our aim is to develop strategies for tumor-

selective generation of ROS. This could be achieved by

conjugating macromolecular carriers (polymers) to

appropriate enzymes or antioxidant inhibitors as

shown in this work and our previous works. Alternately

micellar formulations and nanonoparticles can also be

utilized to selectively deliver the drug to tumor site,

thereby successful antitumor effect can be achieved

without apparent side effects by taking advantage of

the EPR effect of macromolecules in solid tumor

(Figure 6). Another important rationale of this study is

that majority of tumor cells frequently possess very

little antioxidative enzymes, e.g. catalase, which makes

them very vulnerable to oxidative stresses. Thus, the

unique antitumor strategy ‘oxidation therapy’, might

Tumor-targeted induction of oxystress for cancer therapy 483

Jour

nal o

f D

rug

Tar

getin

g D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Con

nect

icut

on

01/0

5/14

For

pers

onal

use

onl

y.

Page 10: Tumor-targeted induction of oxystress for cancer therapy

be a promising approach for clinical anticancer trials in

the future, and warrants further investigation.

References

Akaike T, Ando M, Oda T, Doi T, Ijiri S, Araki S, Maeda H. 1990.

Dependence on O2- generation by xanthine oxidase of

pathogenesis of influenza virus infection in mice. J Clin Invest

85:739–745.

Akaike T, Fujii S, Kato A, Yoshitake J, Miyamoto Y, Sawa T,

Okamoto S, Suga M, Asakawa M, Nagai Y, Maeda H. 2000.

Viral mutation accelerated by nitric oxide production during

infection in vivo. FASEB J 14:1447–1454.

Akaike T, Okamoto S, Sawa T, Yoshitake J, Tamura F, Ichimori K,

Miyazaki K, Sasamoto K, Maeda H. 2003. 8-nitroguanosine

formation in viral pneumonia and its implication for pathogen-

esis. Proc Natl Acad Sci USA 100:685–690.

Beckman KB, Ames BN. 1997. Oxidative decay of DNA. J Biol

Chem 272:19633–19636.

Ben-Yoseph O, Ross BD. 1994. Oxidation therapy: The use of a

reactive oxygen species-generating enzyme system for tumour

treatment. Br J Cancer 70:1131–1135.

Berlett BS, Stadtman ER. 1997. Protein oxidation in aging, disease,

and oxidative stress. J Biol Chem 272:20313–20316.

Bladier C, Wolvetang EJ, Hutchinson P, de Haan JB, Kola I. 1997.

Response of a primary human fibroblast cell line to H2O2:

Senescence-like growth arrest or apoptosis? Cell Growth Differ

8:589–598.

Cerutti PA. 1994. Oxy-radicals and cancer. Lancet 344:862–863.

Cerutti PA, Trump BF. 1991. Inflammation and oxidative stress in

carcinogenesis. Cancer Cells 3:1–7.

Davies KJ. 1993. Protein modification by oxidants and the role of

proteolytic enzymes. Biochem Soc Trans 21:346–353.

Davies KJ. 1995. Oxidative stress: The paradox of aerobic life.

Biochem Soc Symp 61:1–31.

Demple B, Harrison L. 1994. Repair of oxidative damage to DNA:

Enzymology and biology. Annu Rev Biochem 63:915–948.

Doi K, Akaike T, Fujii S, Tanaka S, Ikebe N, Beppu T, Shibahara S,

Ogawa M, Maeda H. 1999. Induction of haem oxygenase-1 by

nitric oxide and ischaemia in experimental solid tumours and

implications for tumour growth. Br J Cancer 80:1945–1954.

Dreher D, Junod AF. 1996. Role of oxygen free radicals in cancer

development. Eur J Cancer 32A:30–38.

Droge W. 2002. Free radicals in the physiological control of cell

function. Physiol Rev 82:47–95.

Duncan R. 2003. The dawning era of polymer therapeutics. Nat Rev

Drug Discov 2:347–360.

Estensen RD, Levy M, Klopp SJ, Galbraith AR, Mandel JS,

Blomquist JA, Watenberg LW. 1999. N-acetylcysetine suppres-

sion of the proliferative index in the colon of patients with

previous adenomatous colonic polyps. Cancer Lett 147:

109–114.

Fang J, Sawa T, Akaike T, Maeda H. 2002. Tumor-targeted delivery

of polyethylene glycol-conjugated D-amino acid oxidase for

antitumor therapy via enzymatic generation of hydrogen

peroxide. Cancer Res 62:3138–3143.

Fang J, Sawa T, Maeda H. 2003a. Factors and mechanism of “EPR”

effect and the enhanced antitumor effects of macromolecular

drugs including SMANCS. Adv Exp Med Biol 519:29–49.

Fang J, Sawa T, Akaike T, Akuta T, Sahoo SK, Greish K, Hamada

A, Maeda H. 2003b. In vivo antitumor activity of pegylated zinc

protoporphyrin: Targeted inhibition of heme oxygenase in solid

tumor. Cancer Res 63:3567–3574.

Fang J, Akaike T, Maeda H. 2004a. Antiapoptotic role of heme

oxygenase (HO) and the potential of HO as a target in anticancer

treatment. Apoptosis 9:27–35.

Fang J, Sawa T, Akaike T, Greish K, Maeda H. 2004b.

Enhancement of chemotherapeutic response of tumor cells by

a heme oxygenase inhibitor, pegylated zinc protoporphyrin. Int J

Cancer 109:1–8.

Fang J, Nakamura H, Deng DW, Akuta T, Greish K, Iyer AK,

Maeda H. 2007. Oxystress inducing antitumor therapeutics via

targeted delivery of PEG-conjugated D-amino acid oxidase. Int J

Cancer, in press.

Figure 6. Scheme of the unique anticancer strategy: oxidation therapy. The ROS accumulation in tumor cells is induced by two approaches;

one is through exogenous delivery, which can be achieved by ROS-generating enzymes (e.g. PEG–DAO system); another way is by inhibiting

the antioxidative defense system of tumor cells (e.g. HO-1 inhibition via PEG–ZnPP). EPR effect is necessary for the targeted delivery of

ROS-generating system or inhibitors of HO-1, to tumor in both approaches. Thus increased ROS in tumor will kill tumor cells via apoptosis

related pathway. Consequently, remarkable and favorable antitumor effect is achieved without apparent side effect (see text for details).

J. Fang et al.484

Jour

nal o

f D

rug

Tar

getin

g D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Con

nect

icut

on

01/0

5/14

For

pers

onal

use

onl

y.

Page 11: Tumor-targeted induction of oxystress for cancer therapy

Feig DI, Reid TM, Loeb LA. 1994. Reactive oxygen species in

tumorigenesis. Cancer Res 54:1890s–1894s.

Fujii S, Akaike T, Maeda H. 1999. Role of nitric oxide in

pathogenesis of herpes simplex virus encephalitis in rats.

Virology 256:203–212.

Goodman AI, Choudhury M, da Silva JL, Schwartzman ML,

Abraham NG. 1997. Overexpression of the heme oxygenase

gene in renal cell carcinoma. Proc Soc Exp Biol Med 214:54–61.

Green HN, Westrop JW. 1958. Hydrogen peroxide and tumour

therapy. Nature 181:128–129.

Greenstein JP. 1954. Biochemistry of cancer. 2nd ed., New York:

Academic Press.

Greish K, Fang J, Inutsuka T, Nagamitsu A, Maeda H. 2003.

Macromolecular therapeutics: Advantages and prospects with

special emphasis on solid tumour targeting. Clin Pharmacokinet

42:1089–1105.

Guyton KZ, Kensler TW. 1993. Oxidative mechanisms in

carcinogenesis. Brit Med Bull 49:523–544.

Halliwell B, Gutteridge JM. 1984. Free radicals, lipid peroxidation,

and cell damage. Lancet 2:1095.

Harris JM. 1992. Poly(ethylene glycol) chemistry: Biotechnical and

biomedical applications. New York: Plenum Press.

Harris JM, Chess RB. 2003. Effect of pegylation on pharmaceu-

ticals. Nat Rev Drug Discov 2:214–221.

Hasegawa Y, Takano T, Miyauchi A, Matsuzuka F, Yoshida H,

Kuma K, Amino N. 2002. Decreased expression of glutathione

peroxidase mRNA in thyroid anaplastic carcinoma. Cancer Lett

182:69–74.

Huang P, Feng L, Oldham EA, Keating MJ, Plunkett W. 2000.

Superoxide dismutase as a target for the selective killing of

cancer cells. Nature 407:390–395.

Iwai K, Maeda H, Konno T. 1984. Use of oily contrast medium for

selective drug targeting to tumor: Enhanced therapeutic effect

and X-ray image. Cancer Res 44:2115–2221.

Iyer AK, Greish K, Fang J, Maeda H. 2006. Exploiting the

enhanced permeability and retention effect for tumor targeting.

Drug Discov Today 11:812–818.

Iyer AK, Greish K, Fang J, Murakami R, Maeda H. 2007. High-

loading nanosized micelles of copoly(styrene–maleic acid)-zinc

protoporphyrin for targeted delivery of a potent heme oxygenase

inhibitor. Biomaterials 28:1871–1881.

Kaibara N, Ikeda T, Hattori T, Inokuchi K. 1971. Experimental

studies on enhancing the therapeutic effect of mitomycin-C with

hydrogen peroxide. Jpn J Exp Med 41:323–329.

Konno R, Yasumura Y. 1992. D-Amino acid oxidase and its

physiological functions. Int J Biochem 24:519–524.

Kuwahara H, Kanazawa A, Wakamatu D, Morimura S, Kida K,

Akaike T, Maeda H. 2004. Antioxidative and antimutagenic

activities of 4-vinyl-2,6-dimethoxyphenol (canolol) isolated from

canola oil. J Agric Food Chem 52:4380–4387.

Lindahl T. 1993. Instability and decay of the primary structure of

DNA. Nature 362:709–751.

Maeda H. 2001. The enhanced permeability and retention (EPR)

effect in tumor vasculature: The key role of tumor-selective

macromolecular drug targeting. Adv Enzyme Regul 41:189–207.

Maeda H, Akaike T. 1991. Oxygen free radicals as pathogenic

molecules in viral diseases. Proc Soc Exp Biol Med 198:

721–727.

Maeda H, Sawa T, Konno T. 2001. Mechanism of tumor-targeted

delivery of macromolecular drugs, including the EPR effect in

solid tumor and clinical overview of the prototype polymeric

drug SMANCS. J Control Release 74:47–61.

Maines MD, Abrahamsson PA. 1996. Expression of heme

oxygenase-1 (HSP32) in human prostate: Normal, hyperplastic,

and tumor tissue distribution. Urology 47:727–733.

Maines MD. 1988. Heme oxygenase: function, multiplicity,

regulatory mechanisms, and clinical applications. FASEB J 2:

2557–2568.

Mates JM, Perez-Gomez C, Nunez de Castro I. 1999. Antioxidant

enzymes and human diseases. Clin Biochem 32:595–603.

Matsumura Y, Maeda H. 1986. A new concept for macromolecular

therapeutics in cancer chemotherapy: Mechanism of tumoritropic

accumulation of proteins and the antitumor agent SMANCS.

Cancer Res 46:6387–6392.

Matsura T, Kai M, Fujii Y, Yamada K. 1999. Hydrogen peroxide-

induced apoptosis in HL-60 cells requires caspase-3 activation.

Free Radic Res 30:73–83.

McCord JM. 2000. The evolution of free radicals and oxidative

stress. Am J Med 108:652–659.

Mealey J, Jr. 1965. Regional infusion of vinblastine and hydrogen

peroxide in tumor-bearing rats. Cancer Res 25:1839–1843.

Mochizuki Y, Hruban Z, Morris HP, Slesers A, Vigil EL. 1971.

Microbodies of Morris hepatomas. Cancer Res 31:763–773.

Muggia FM. 1999. Doxorubicin-polymer conjugates: Further

demonstration of the concept of enhanced permeability and

retention. Clin Cancer Res 5:7–8.

Nathan CF, Cohn ZA. 1981. Antitumor effects of hydrogen

peroxide in vivo. J Exp Med 154:1539–1553.

Noguchi Y, Wu J, Duncan R, Strohalm J, Ulbrich K, Akaike T,

Maeda H. 1998. Early phase tumor accumulation of macro-

molecules: A great difference in clearance rate between tumor

and normal tissues. Jpn J Cancer Res 89:307–314.

Oda T, Akaike T, Hamamoto T, Suzuki F, Hirano T, Maeda H.

1989. Oxygen radicals in influenza-induced pathogenesis and

treatment with pyran polymer-conjugated SOD. Science 244:

974–976.

Okamoto T, Akaike T, Sawa T, Miyamoto Y, van der Vliet A,

Maeda H. 2001. Activation of matrix metalloproteinases by

peroxynitrite-induced protein S-glutathiolation via disulfide

S-oxide formation. J Biol Chem 276:29596–29602.

Pasut G, Guiotto A, Veronese F. 2004. Protein, peptide and non-

peptide drug PEGylation for therapeutic application. Expert

Opin Ther Patents 14:859–894.

Ren JG, Xia LH, Just T, Dai YR. 2001. Hydroxyl radical-induced

apoptosis in human tumor cells is associated with telomere

shortening but not telomerase inhibition and caspase activation.

FEBS Lett 488:123–132.

Sahoo SK, Sawa T, Fang J, Tanaka S, Miyamoto Y, Akaike T,

Maeda H. 2002. Pegylated zinc protoporphyrin: A water-soluble

heme oxygenase inhibitor with tumor-targeting capacity.

Bioconjugate Chem 13:1031–1038.

Sato K, Ito K, Kohara H, Yamaguchi Y, Adachi K, Endo H. 1992.

Negative regulation of catalase gene expression in hepatoma

cells. Mol Cell Biol 12:2525–2533.

Sawa T, Wu J, Akaike T, Maeda H. 2000. Tumor-targeting

chemotherapy by a xanthine oxidase–polymer conjugate that

generates oxygen-free radicals in tumor tissue. Cancer Res 60:

666–671.

Sawa T, Akaike T, Ichimori K, Akuta T, Kaneko K, Nakayama H,

Stuehr DJ, Maeda H. 2003. Superoxide generation mediated by

8-nitroguanosine, a highly redox-active nucleic acid derivative.

Biochem Biophys Res Commun 311:300–306.

Seymour LW, Miyamoto Y, Maeda H, Brereton M, Strohalm J,

Ulbrich K, Duncan R. 1995. Influence of molecular weight on

passive tumour accumulation of a soluble macromolecular drug

carrier. Eur J Cancer 31A:766–770.

Simizu S, Takada M, Umezawa K, Imoto M. 1998. Requirement of

caspase-3 (-like) protease-mediated hydrogen peroxide pro-

duction for apoptosis induced by various anticancer drugs. J Biol

Chem 273:26900–26907.

Stegman LD, Zheng H, Neal ER, Ben-Yoseph O, Pollegioni L,

Pilone MS, Ross BD. 1998. Induction of cytotoxic oxidative

stress by D-alanine in brain tumor cells expressing Rhodotorula

gracilis D-amino acid oxidase: A cancer gene therapy strategy.

Hum Gene Ther 9:185–193.

Sugiura K. 1958. Effect of hydrogen peroxide on transplanted rat

and mouse tumours. Nature 182:1310–1311.

Tumor-targeted induction of oxystress for cancer therapy 485

Jour

nal o

f D

rug

Tar

getin

g D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Con

nect

icut

on

01/0

5/14

For

pers

onal

use

onl

y.

Page 12: Tumor-targeted induction of oxystress for cancer therapy

Suhara T, Fukuo K, Sugimoto T, Morimoto S, Nakahashi T, Hata S,

Shimizu M, Ogihara T. 1998. Hydrogen peroxide induces

up-regulation of Fas in human endothelial cells. J Immunol 160:

4042–4047.

Sundaresan M, Yu ZX, Ferrans VJ, Irani K, Finkel T. 1995.

Requirement for generation of H2O2 for platelet-derived growth

factor signal transduction. Science 270:296–299.

Tanaka S, Akaike T, Fang J, Beppu T, Ogawa M, Tamura F,

Miyamoto Y, Maeda H. 2003. Antiapoptotic effect of haem

oxygenase-1 induced by nitric oxide in experimental solid

tumour. Br J Cancer 88:902–909.

Umezawa K, Akaike T, Fujii S, Suga M, Setoguchi K, Ozawa A,

Maeda H. 1997. Induction of nitric oxide synthesis and xanthine

oxidase and their roles in the antimicrobial mechanism against

Salmonella typhimurium infection in mice. Infect Immun 65:

2932–2940.

Vicent MJ, Duncan R. 2006. Polymer conjugates:

Nanosized medicines for treating cancer. Trends Biotechnol

24:39–47.

Wu J, Akaike T, Hayashida K, Okamoto T, Okuyama A, Maeda H.

2001. Enhanced vascular permeability in solid tumor involving

peroxynitrite and matrix metalloproteinases. Jpn J Cancer Res

92:439–451.

Yagi K. 1971. Reaction mechanism of D-amino acid oxidase. Adv

Enzymol Relat Areas Mol Biol 34:41–78.

Yamakawa H, Ito Y, Naganawa T, Banno Y, Nakashima S,

Yoshimura S, Sawada M, Nishimura Y, Nozawa Y, Sakai N.

2000. Activation of caspase 9 and 3 during H2O2-induced

apoptosis of PC12 cells independent of ceramide formation.

Neurol Res 22:556–564.

Yamanaka N, Deamer D. 1974. Superoxide dismutase activity in

WI-38 cell cultures: Effect of age, trypsinization and SV-40

transformation. Physiol Chem Phys 6:95–106.

Yoshikawa T, Kokura S, Tanaka K, Naito Y, Kondo M. 1995.

A novel cancer therapy based on oxygen radicals. Cancer Res 55:

1617–1620.

Yoshitake J, Akaike T, Akuta T, Tamura F, Ogura T, Esumi H,

Maeda H. 2004. Nitric oxide as an endogenous mutagen for

Sendai virus without antiviral activity. J Virol 78:8709–8719.

Zalipsky S. 1995. Functionalized poly(ethylene glycol) for

preparation of biologically relevant conjugates. Bioconjugate

Chem 6:150–165.

J. Fang et al.486

Jour

nal o

f D

rug

Tar

getin

g D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Con

nect

icut

on

01/0

5/14

For

pers

onal

use

onl

y.