middlesex university research repository for the review of ato.pdf · levels, in one experiment,...

13
Middlesex University Research Repository An open access repository of Middlesex University research Akhtar, Anam, Wang, Scarlet Xiaoyan, Ghali, Lucy ORCID: https://orcid.org/0000-0003-3410-6615, Bell, Celia M. ORCID: https://orcid.org/0000-0002-3270-7081 and Wen, Xuesong ORCID: https://orcid.org/0000-0001-6518-8962 (2017) Recent advances in arsenic trioxide encapsulated nanoparticles as drug delivery agents to solid cancers. Journal of Biomedical Research, 31 (2) . pp. 1-12. ISSN 1674-8301 (doi:10.7555/JBR.31.20160059) Published version (with publisher’s formatting) This version is available at: Copyright: Middlesex University Research Repository makes the University’s research available electronically. Copyright and moral rights to this work are retained by the author and/or other copyright owners unless otherwise stated. The work is supplied on the understanding that any use for commercial gain is strictly forbidden. A copy may be downloaded for personal, non-commercial, research or study without prior permission and without charge. Works, including theses and research projects, may not be reproduced in any format or medium, or extensive quotations taken from them, or their content changed in any way, without first obtaining permission in writing from the copyright holder(s). They may not be sold or exploited commercially in any format or medium without the prior written permission of the copyright holder(s). Full bibliographic details must be given when referring to, or quoting from full items including the author’s name, the title of the work, publication details where relevant (place, publisher, date), pag- ination, and for theses or dissertations the awarding institution, the degree type awarded, and the date of the award. If you believe that any material held in the repository infringes copyright law, please contact the Repository Team at Middlesex University via the following email address: [email protected] The item will be removed from the repository while any claim is being investigated. See also repository copyright: re-use policy:

Upload: others

Post on 01-Jun-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Middlesex University Research Repository for the review of ATO.pdf · levels, in one experiment, glutathione content was increased by treating the cells with N-acetylcysteine. The

Middlesex University Research RepositoryAn open access repository of

Middlesex University research

http://eprints.mdx.ac.uk

Akhtar, Anam, Wang, Scarlet Xiaoyan, Ghali, Lucy ORCID:https://orcid.org/0000-0003-3410-6615, Bell, Celia M. ORCID:

https://orcid.org/0000-0002-3270-7081 and Wen, Xuesong ORCID:https://orcid.org/0000-0001-6518-8962 (2017) Recent advances in arsenic trioxide

encapsulated nanoparticles as drug delivery agents to solid cancers. Journal of BiomedicalResearch, 31 (2) . pp. 1-12. ISSN 1674-8301 (doi:10.7555/JBR.31.20160059)

Published version (with publisher’s formatting)

This version is available at: http://eprints.mdx.ac.uk/20854/

Copyright:

Middlesex University Research Repository makes the University’s research available electronically.

Copyright and moral rights to this work are retained by the author and/or other copyright ownersunless otherwise stated. The work is supplied on the understanding that any use for commercial gainis strictly forbidden. A copy may be downloaded for personal, non-commercial, research or studywithout prior permission and without charge.

Works, including theses and research projects, may not be reproduced in any format or medium, orextensive quotations taken from them, or their content changed in any way, without first obtainingpermission in writing from the copyright holder(s). They may not be sold or exploited commercially inany format or medium without the prior written permission of the copyright holder(s).

Full bibliographic details must be given when referring to, or quoting from full items including theauthor’s name, the title of the work, publication details where relevant (place, publisher, date), pag-ination, and for theses or dissertations the awarding institution, the degree type awarded, and thedate of the award.

If you believe that any material held in the repository infringes copyright law, please contact theRepository Team at Middlesex University via the following email address:

[email protected]

The item will be removed from the repository while any claim is being investigated.

See also repository copyright: re-use policy: http://eprints.mdx.ac.uk/policies.html#copy

Page 2: Middlesex University Research Repository for the review of ATO.pdf · levels, in one experiment, glutathione content was increased by treating the cells with N-acetylcysteine. The

Original article

Recent advances in arsenic trioxide encapsulated nanoparticlesas drug delivery agents to solid cancers

Anam Akhtar, Scarlet Xiaoyan Wang, Lucy Ghali, Celia Bell, Xuesong Wen✉,Δ

Abstract

Since arsenic trioxide (ATO) was first approved as the front line therapy for acute promyelocytic leukemia (APL)20 five years ago, its anti-cancer properties for various malignancies have been under intense investigation. However,the clinical successes of ATO in treating hematological cancers have not been translated to solid cancers. This is dueto arsenic’s rapid clearance by the body’s immune system before reaching the tumor site. Several attempts havehenceforth been made to increase its bioavailability toward solid cancers without increasing its dosage albeit withoutmuch success. This review summarizes the past and current utilization of ATO in the medical field with primary focuson the implementation of nanotechnology for ATO delivery to solid cancer cells. Different approaches that have beenemployed to increase arsenic’s efficacy, specificity and bioavailability to solid cancer cells were evaluated andcompared. The potential of combining different approaches or tailoring delivery vehicles to target specific types ofsolid cancers according to individual cancer characteristics and arsenic chemistry was proposed and discussed.

Keywords: arsenic trioxide, solid cancer, nanotechnology, drug delivery, liposome

Introduction

Arsenic is a naturally occurring metalloid found as atrace element in the earth’s crust [1]. Instead of beingpresent in its elemental forms; it exists as highly toxicarsenates of metals like calcium, sodium and potassiumor as oxides and sulphides. Among its three inorganicforms, ATO manifests as white arsenic and isindustrially produced as a by-product while roastingarsenic containing ores and purifying the smoke [2].The transformation of ATO from being the “king ofpoisons” to a “broad spectrum anti-cancer drug” is oneof the most exciting stories in clinical oncology [3].Despite being the mainstay of materiamedica since

the past 2400 years, ATO faced an escalating decline inits use as a therapeutic agent in the twentieth century

due to an increasing awareness of its toxicity [4]. Itsresurgence transpired in the late twentieth century whena group of Chinese physicians recognized it as the mainanti-leukemic ingredient in traditional Chinese medi-cine and decided to evaluate its therapeutic potency intreating patients with acute promyelocytic leukemia(APL), a subtype of acute myeloid leukemia [5].Clinical trials using ATO to treat APL patients as asingle agent or in combination with conventionalchemotherapy yielded highly encouraging results [5].ATO was consequently approved by Food and DrugAdministration (FDA) as frontline therapy for APL in2000 [6]. Similar clinical successes were subsequentlytranslated for other hematological malignancies, forinstance, acute myeloid leukemia, chronic myelogenousleukemia, Hodgkins disease and multiple myeloma

ΔThe authors reported no conflict of interests✉Corresponding author: Dr. Xuesong Wen,Department of Natural

Sciences, School of Science and Technology, Middlesex University,The Burroughs, London, NW4 4BT Email:[email protected].

Received 3 May 2016, Revised 29 July 2016, Accepted 22 August2016, Epub 20 September 2016CLC number: R730.51, Document code: A

Available online at www.jbr-pub.org

Open Access at PubMed Central

The Journal of Biomedical Research, 2016 31(1): 1–12

© 2016 by the Journal of Biomedical Research. All rights reserved doi:10.7555/JBR.31.20160059

1

5

10

15

20

25

30

35

40

45

50

1

5

10

15

20

25

30

35

40

45

50

JBR-16059-AA.3d 14/9/016 16:40:32

Page 3: Middlesex University Research Repository for the review of ATO.pdf · levels, in one experiment, glutathione content was increased by treating the cells with N-acetylcysteine. The

although not significantly for the solid tumors [7]. Rapidclearance of ATO and its products from the blood byreticuloendothelial system (RES) limits the therapeuticdose reaching the tumor site. Increasing ATO dosagecould not be a solution as it leads to systemic toxicitiessuch as long-term memory damage, liver failure, cardiacfailure and peripheral neuropathy [8].Since then, several attempts have henceforth been

carried out to utilize arsenic’s significant anti-cancerproperties for treating solid tumors by increasing itsbioavailability and specificity for carcinoma cells whilereducing its administered dosage [9]. These includesensitizing the cells prior to ATO treatment, employingit in combination therapy with other conventionalchemotherapeutic agents to explore their synergisticactions or employing nanotechnology to increasebioavailability of ATO while reducing its systemictoxicity [10–13].This review aims to summarize the advances that

have been made till date regarding therapeutic applica-tions of ATO drug with an emphasis on the ongoinginvolvement of nanotechnology in the current ATOtherapy in treating various malignancies.

Arsenic trioxide (ATO) history: rise, declineand resurgence

ATO presents a rich history in medicinal applicationsdating back to more than 2400 years [14]. It was used intraditional Chinese medicine to treat syphilis, psoriasisand rheumatism [2]. Other applications were alsoreported with ATO being effective as an anti-spasmodic,anti-pyretic, anti- periodic, caustic and sedative [14].However, the awareness of its toxicity began to risewhen people realized that long-term chronic exposure toATO could cause hyperpigmentation of the skin,cancers of lung, kidney, bladder and prostate alongwith rare cases of neuropathy, encephalopathy, leuco-penia and diabetes [6]. Consequently, by the turn oftwentieth century, its medicinal use started to declinedue to its acquired reputation of being a toxic substanceand a carcinogen.ATO resurged as a therapeutic compound in 1990s

following an initial study at Harbin Medical Universitywhere its anti-leukemic properties were identified intreating APL [14]. A group of physicians at ShanghaiSecond Medical University continued the study andremarkable results were observed from the clinical trialwith patients of relapsed and refractory APL after verylow doses of intravenous ATO were administered [15].It was a major breakthrough in treating APL relapsedpatients as the mortality rate with routine treatment i.e.all-trans retinoic acid (ATRA) and anthracycline based

chemotherapy was 20-30% and bone marrow trans-plantation was the only option [14]. The findings, whichwere later successfully replicated in United States, led tothe approval of ATO (TrisenoxTM) by FDA as a frontlinedrug for relapsed and refractory APL in 2000 [6].Although some successes of ATO therapy have also

been observed in other hematological cancers, thetherapeutic dosage administered is relatively low, andhence the commonly reported side effects are low gradeand reversible after ceasing the ATO therapy. However,the same kind of clinical success has not been translatedfor solid cancers in humans despite ongoing clinicaltrials to investigate arsenic’s efficacy on a variety ofsolid cancers [15] (http://www.clinicaltrials.gov). Thisis primarily due to the rapid clearance of ATO and itsmetabolites from the blood by RES. Compromisedtherapeutic dose of ATO reach the tumor site, therefore,an increased dosage is required which might, in turn,lead to systemic toxicities and long-term memorydamage and hormonal imbalance [8,16]. Therefore,there is a crucial need for analyzing various possibleapproaches of increasing the bioavailability of ATOwithout raising its dosage.

Therapeutic strategies in using ATO alone orATO combined agents

Anti-cancer activity of free ATO has been tested on avariety of solid tumor cell lines with positive experi-mental results but limited clinical success. Zhao andcoworkers [17] administered direct intra-tumoral injec-tions of ATO to human esophageal carcinoma xeno-grafts in mice and investigated ATO distribution and itsefficacy within the tumor. Tumour growth inhibition of13.56, 62.37 and 76.92% was observed with adminis-tration of 1, 5 and 10 mg of ATO respectively withoutappreciable systemic side effects. These results pro-vided encouraging evidence for potential clinical utilityof direct intra-tumoral injections of ATO as one form oftreatment for solid cancers. However, there is anescalating need to investigate other approaches whereATO can be delivered selectively to its site of action andto penetrate inside the tumor without being toxic tosurrounding healthy tissue. Some of the approaches thathave been tested or proposed to use are outlined below(Fig. 1).

Glutathione depleting agents

One of the key targets of ATO is glutathione (GSH), atripeptide molecule which protects the cells fromoxidative stress. It is involved in a number of bio-reductive reactions and in the detoxification of xeno-biotics [11]. Glutathione titrates intracellular arsenic by

2 Akhtar A et al. J Biomed Res, 2017, 31

1

5

10

15

20

25

30

35

40

45

50

1

5

10

15

20

25

30

35

40

45

50

JBR-16059-AA.3d 14/9/016 16:40:32

Page 4: Middlesex University Research Repository for the review of ATO.pdf · levels, in one experiment, glutathione content was increased by treating the cells with N-acetylcysteine. The

forming a complex As(GS)3 [11]. This interaction isexothermic and spontaneous at pH 7.4 and temperature37°C (ΔG = -8.8 kcal/mol). The formation of thiscomplex is preceded by its export out of the cell viamultidrug resistance export proteins (MRP1-10) foundin cells [8]. Hence, substances which deplete glu-tathione can sensitize the cells to ATO anti-tumoractivity by preventing its export out of the cells. Toverify the importance of modulation of glutathionelevels, in one experiment, glutathione content wasincreased by treating the cells with N-acetylcysteine.The anti-tumor activity of ATO was subsequentlyobserved to be suppressed [12]. This makes it evidentthat simultaneously depleting intracellular glutathionelevels might be an effective strategy to intensify ATOaction.Gartenhaus et al. [13] reported ascorbic acid to be one

such compound which depletes intracellular glutathionelevels. Ascorbic acid further auto-oxidises to releaseH2O2 which might enhance pro-apoptotic effects ofarsenic. L-Buthioninesulphoximine (BSO) is anotherdrug which inhibits a critical step in glutathionesynthesis by inhibiting the rate limiting enzyme γ-

glutamyl cysteine synthetase along with generatingreactive oxygen species (ROS). This sensitizes the cellsto ATO treatment and was reported to enhance drug’sefficacy in inducing apoptosis on a number of cell linesarising from cervix, bladder, prostrate, kidney, colon,pancreas and breast cancers [18]. Further biochemicalstudies indicated that this combination treatment blocksthe H2O2 scavenging system in the cells by interferingwith glutathione along with catalase and glutathioneoxidase enzymes. Therefore this combination of anROS producing drug and ROS removing inhibitor hasthe potential to offer selective and efficient anti-cancertreatment [18].

Ionising radiation

When ATO is used in conjunction with ionisingradiation, radiation response was reported to beintensified in tumor cells. ATO acts as a radiosensitizerto tumor cells by increasing their susceptibility towardapoptosis. Combined with a low-dose irradiation, thedual treatment has been reported to hold a promise foreffective cancer therapy. Kang and Lee [9] observedsynergistic effect when human cervical cancer cells

Fig. 1 Key therapeutic strategies employed in the attempt to increase the efficacy of ATO as a drug in treating solid tumors withoutescalating its dosage [9,11,12,18–39].

Drug delivery of encapsulated arsenic trioxide for solid cancers 3

1

5

10

15

20

25

30

35

40

45

50

1

5

10

15

20

25

30

35

40

45

50

JBR-16059-AA.3d 14/9/016 16:40:32

Page 5: Middlesex University Research Repository for the review of ATO.pdf · levels, in one experiment, glutathione content was increased by treating the cells with N-acetylcysteine. The

were treated with ATO along with ionising radiation.The treatment caused regression in tumor growth byincreasing ROS, destabilizing mitochondrial membraneand activating caspases in cancer cells.

Non-steroidal anti-inflammatory drugs (NSAID)

These drugs are known to enhance the susceptibilityof cancer cells toward various further anti-cancertreatments such as biologic treatment, chemotherapyand radiotherapy. One such drug sulindac has beenreported to sensitize the cells due to its apoptosisinducing and anti-angiogenic properties [40]. Itincreases the radiation sensitivity of cancer cells byinterfering with the tumor neovascularisation [40].Sulindac also exhibits synergistic augmentation ofcytotoxicity when used alongside paclitaxel [41].Similarly, when sulindacwas used alongside ATO onhuman lung cancer NCI-H157 cell line, it enhanced thecaspase activation via generation of ROS [19]. Thissynergistic action allowed much lower concentration ofATO to be effective in carrying out its anti-cancerproperties.Another NSAID drug, indomethacin is a known

inhibitor of cyclooxygenase-2 (COX-2), whose expres-sion is significantly increased in a variety of cancers[19]. Hence, by acting as a COX-2 enzyme inhibitor,indomethacin exerts its cytotoxic effects on tumor cells.When it was used in combination with ATO, very lowdoses of both drugs achieve significant cytotoxicity[42]. The synergism in their anti-cancer action wasclearly evident as either drug was not as toxic when itwas administered at the same concentration alone [42].

Encapsulation of ATO: as a single agent orco-encapsulated with another drug

The advent of nanotechnology has already changedthe landscape of drug delivery. Improving the bioavail-ability of poorly water soluble drugs, selectivelydelivering the drug in a cell or tissue specific manner,co-delivering two or more drugs, transporting the drugto intracellular compartments/ site of action andvisualizing the site of delivery using imaging modalitiesare just few of the promises that nanotechnology holdsfor drug delivery [43] (Fig. 2). ATO, along with fewother organic arsenicals has already been encapsulatedin nanoparticles and their efficacy is summarized andevaluated in this review (Table 1).

Liposome encapsulation

Liposomes are lipid nanoparticles encapsulating ahydrophilic interior with a hydrophobic membrane.They are formed via self-assembly when certainphospholipids are hydrated in water [44]. The hydro-phobic membrane can entrap strongly lipophilic drugswhereas the hydrophilic interiors can encapsulate watersoluble molecules like ATO. Zhao et al. [17] preparedliposomes encapsulating ATO and intravenouslyinjected them to investigate their therapeutic effect onC6 gliomas established in rat brains. Liposomalpreparation led to a 5-fold increase in arsenic concen-tration in rat brains as compared to ATO solutions,inducing apoptosis and inhibiting tumor angiogenesisby interfering in the expression of vascular endothelialgrowth factor (VEGF) with fewer side effects [17].

Fig. 2 Implementation of nanotechnology for ATO drug delivery. Amphiphilic phospholipids and block co-polymers self-assemble toform liposomes and polymersomes respectively which can effectively encapsulate ATO in their cores. Polyethylene glycol (PEG) layeracts as a brush to endow stealth, prevent immune recognition and recognition by serum proteins and increases their circulation half-lifein vivo. Targeting ligands such as antibodies, peptides, proteins and other small molecules attached to the membrane bilayer or PEGfacilitate the receptor-mediated endocytosis. Fluorophores attached promote the imaging of the nanoparticles and their fate in vivo.

4 Akhtar A et al. J Biomed Res, 2017, 31

1

5

10

15

20

25

30

35

40

45

50

1

5

10

15

20

25

30

35

40

45

50

JBR-16059-AA.3d 14/9/016 16:40:33

Page 6: Middlesex University Research Repository for the review of ATO.pdf · levels, in one experiment, glutathione content was increased by treating the cells with N-acetylcysteine. The

Table1

Applicationsof

deliveringATO

into

cancercells

usingnan

otechnolog

y.Deliveryvehicle

Co-encapsulated

agent

Targetinglig

and

Type

ofcancer

tested

TreatmentRespo

nse

References

Liposom

esManganese

(Mn2

+)

F(ab’) 2fragmentsof

PGN635,

human

mon

oclonalph

osph

otidyl

serine

(PS)-

targetingantib

ody

Glio

blastomamultiform

(GBM)

EncapsulatedAs3

+-M

n2+dissociatesto

releaseAs3

+as

activ

edrug

andMn2

+as

T1contrastagentforMRI

imaging.

[10]

Liposom

es(N

anob

ins)

Transition

metal

ion

(Ni,Cu,

Zn,

Co)

NA

Lym

phom

a;Breastcancer;

SU-D

HL-4

human

lymphom

acelllin

es

Liposom

alATO

isless

harm

fulto

theov

arianfunctio

nsof

femalepatients.

[20–22

]

Liposom

esTransition

metal

ion

(Ni,Cu,

Zn,

Co)

Urokinase

plasminogen

activ

ator

antib

ody

Ovarian

cancer

Uptakeof

targeted

liposom

esby

cancer

cells

is4-fold

high

erthan

non-targeted

ones.

[23]

Liposom

esNA

NA

NA

Liposom

alATO

ismoreeffectivethan

free

form

inreducing

theoncogene

level(H

PV-16)

inHPV

positiv

ecervical

cancersandinducing

apoptosis

[24]

Liposom

es(therm

osensitiv

e)Transition

metal

ion

(Ni,Cu,

Zn,

Co)

NA

NA

Drugreleaseachieved

byasm

allbu

trapidincrease

intemperature

(<4°C)

[25]

Liposom

esTransition

metal

ion

(Ni,Cu,

Zn,

Co)

Folic

acid

FRpo

sitiv

ehu

man

nasoph

aryn

gealandcervical

cancer

celllin

es

Enh

ancedanti-cancer

activ

ityob

served

with

thetargeted

liposom

es[26]

Liposom

es(incorporatin

garsenono

lipid)

NA

NA

Three

malignant

celllin

es(H

L-60,

C6,

andGH3)

Inducedmorphological

changesandinhibitedsurvival

ofcancer

celllin

eswhile

beingno

n-toxicto

controlcells.

[27]

Polym

ersomes

(PEGylated

PLGA

nano

particles)

NA

NA

Murineperitoneal

macrophages

Uptakeby

macrophages

reducedindicatin

glonger

circulationhalf-lifeof

nanoparticles

[28]

Polym

ersomes

NA

anti-CD44

v6sing

lechainvariable

fragment

Pancreatic

cancer

celllin

e,PA

NC-1

Targeted

nanoparticlesdisplayenhanced

accumulation

intumor

site

andsuccessfully

induce

apoptosis.

[29]

Polym

ersomes

with

thiol

grou

psforloadingof

phenylarsine

oxide(PAO)

utilizing

thiol-arsenicchem

istry

NA

NA

Breastcancer,MDA-M

B-435

celllin

eThe

polymeric

system

displays

similartoxicity

asfree

PAO

onthecelllin

etested.

[30]

Nanoparticlesform

edby

linking

sugar-lip

oaminoacid

toPA

O

NA

NA

Breastcancer

MCF-7

cell

lines

andHT-29

coloncancer

celllin

es

Inhibitio

nof

cellproliferationandapoptosisin

MCF-7

butnotin

HT-29

celllin

esshow

inghigh

selectivity

with

inthesearsenicals.

[31]

Drug delivery of encapsulated arsenic trioxide for solid cancers 5

1

5

10

15

20

25

30

35

40

45

50

1

5

10

15

20

25

30

35

40

45

50

JBR-16059-AA.3d 14/9/016 16:40:34

Page 7: Middlesex University Research Repository for the review of ATO.pdf · levels, in one experiment, glutathione content was increased by treating the cells with N-acetylcysteine. The

Table1Applicationsof

deliveringATO

into

cancer

cells

usingnan

otechnology.(contin

ued)

Deliveryvehicle

Co-encapsulated

agent

Targetinglig

and

Type

ofcancer

tested

TreatmentRespo

nse

References

pHrespon

sive

polymer

networksurrou

ndingATO

encapsulatinglip

osom

e

Ni2+catio

nNA

Hum

ancervical

HeL

acell

lines

Sim

ultaneousreleaseof

both

Ni2+andAs3

+,with

the

form

erdiminishing

anti-oxidantslevelsaiding

the

apop

totic

effect

ofAs3

+.

[32]

Liposom

es(therm

osensitiv

e)Mn 0

.5Zn 0

.5Fe 2O4

magnetic

nano

particles

NA

MDA-M

B-231

breast

cancer

celllin

eThe

magnetic

nano

particlesserveas

thesource

ofhypertherm

iacausingthelip

osom

eto

destabilize

atits

phasetransitio

ntemperature

andreleaseAs3

+to

indu

ceapoptosis.

[33]

Nanoparticles

(Arsenic

platinum

complex)

Pt2+

NA

Colon

HCT-116and

glioblastomaU-87celllin

eCom

binatio

nof

cisplatin

andATO

prov

esmoreeffective

than

either

ofthedrug

.[34]

Magnetitedo

pedmesop

orou

ssilicanano

particles(M

SN)

Drugcamptothecinin

thepo

resof

MSN

alongwith

ATO

onits

surfaceusingthiol

bind

ing

NA

Pancreatic

cancer

cells

(BxP

C-3)

Displaysasynergistic

effect

andmagnetite

superparam

agnetic

crystalscanbe

used

asacontrast

agentforMRI

[35]

Polym

ersomeencapsulating

arsenic

Co-treatm

entwith

si-RNA

polyplex

NA

Hum

anpancreatic

cancer

Synergistic

effect

with

si-RNA

silencingKRAS

oncogene

andAs3

+inducing

apoptosis

[36]

Fe 3O4magnetic

nanoparticles

complexed

with

As 2O3

Iron

oxide

nano

particles

NA

Hum

ancervical

cancer

cell

line(H

eLa)

Inhibitcancer-related

proteins

expression

when

combinedwith

hypertherm

ia[37]

Liposom

esPlatin

umFolic

acid

Folic

acid

positiv

etumor

cell

line

Enhanceduptake

andapoptosisinductionof

targeted

liposom

es[26]

ATO

nanoparticles

Coatedwith

chito

sanand

2,3-dimercaptosuccinic

acid

NA

Hum

anandrog

endepend

ent

andindependentprostate

cancer

celllin

es

Coatednanoparticlesaremorestable

andless

toxicthan

bare

As 2O3nanoparticles.

[38]

6 Akhtar A et al. J Biomed Res, 2017, 31

1

5

10

15

20

25

30

35

40

45

50

1

5

10

15

20

25

30

35

40

45

50

JBR-16059-AA.3d 14/9/016 16:40:34

Page 8: Middlesex University Research Repository for the review of ATO.pdf · levels, in one experiment, glutathione content was increased by treating the cells with N-acetylcysteine. The

However, the liposomes formulated were stable for amaximum of three days, which limited their therapeuticuse.

Nanobins

ATO forms arsenous acid, As (OH)3 when dissolvedin water at physiologic pH [8]. The latter is able torapidly cross the lipid membranes; thereby the lipo-somes formulated with ATO encapsulated are highlyunstable with a stability life span of around 3 days. Theissue of such “leaky liposomes” was solved when Chenet al. [26] developed a novel method to stablyencapsulate ATO in liposomes utilizing a metal-iongradient loading mechanism. Arsenic complexes withtransition metal ions such as NiII, CoII, PtII and forms aprecipitate. Utilizing this property, liposomes formedare first loaded with acetate solution of a transition metalion, followed by loading of ATO to the liposomesformulated [22]. Efflux of protonated acetic acid drivesthe ATO to get efficiently encapsulated within theliposomes where it stably precipitates with the transitionmetal ion in the form of a solid, crystalline substance[8].Loading of ATO via this mechanism was found to be

very efficient and the lipid layer surrounding the drugattenuates its toxicity around 4 times compared with thefree drug. These liposomes exhibit a pH dependency byreleasing the arsenic payload on encountering lower pHin either acidic tumor milieu or the endosomes afterbeing taken up by the cells. Ahnet al. [21] tested theefficacy of these nanobins for orthotopic model of triplenegative breast cancer. Avisible shrinkage of tumor wasobserved in rats which were subjected to the nanobintreatment. This could be attributed to the prolongedpharmacokinetics due to enhanced drug exposure to thetumor caused by better penetration of liposomes alongwith a reduced clearance of encapsulated arsenic withinPEGylated liposomes. The shelf life of nanobins wasgreater than 6 months, probably due to the stabilityprovided by direct arsenic-metal bond [8].Most recently in our laboratories, Wang and collea-

gues [24] have also established that treatment withliposomal ATO is distinctively effective for HPVinfected cervical cancer cells, by reducing the expres-sion of HPV-18 E6 oncogene and inducing apoptosismore effectively than free ATO.When acetate solution of aqua-cisplatin was

employed for arsenic complexation, the liposomesformed had both arsenic and platinum species seques-tered inside. Cisplatin (cis-diamine dichloroplatinumII)is widely used to treat a variety of solid tumors such asbladder, ovarian, testicular carcinomas [44]. On being

intracellularly hydrolysed, it yields aqua cisplatin,which induces apoptotic cell death in cancer cells bycreating intra and inter-DNA strands cross-links [34].This method hence provides an efficient co-encapsula-tion strategy to load both ATO and platinum drugswithin the liposomal aqueous core. To further improveanti-cancer activity of the resulting nanobin, Chen et al.[26] conducted a study using folic acid as a targetingligand to introduce ATO into malignant epithelial cancercell lines which overexpressed folic acid receptors (FR).The efficacy of such “targeted” liposomes was analyzedon FR positive KB (human nasopharyngeal) cell lineand cellular uptake of ATO via FR mediated endocy-tosis was observed to be much more than FR negativeMCF-7 (breast cancer) cell line. Moreover, the targetedliposomal uptake was 3-6 times higher than theuntargeted ATO liposomes [26].The targeting via nano-carriers opens a new horizon

for selectively delivering the drug to specific cancercells. Zhang et al. [23] utilized the overexpression ofurokinase system in ovarian epithelial cells andconsequently conjugated urokinase plasminogen acti-vator (uPA) antibodies to nanobins. The resultingtargeted liposomes were reported to exhibit more than4-fold increase in their uptake by the ovarian cancercells than their non-targeted counterparts. They alsoinhibited tumor cell growth both in vitro and in vivo andinduced apoptosis along with downregulating stem cellmarker expression. Hence, utilizing the specific cancercell surface characteristics and identifying suitableligands to be attached to ATO encapsulating liposomesare essential in enhancing the efficacy of ATO therapy.

Thermosensitive liposomes

Mild hyperthermia has frequently been employed as astimulus to release the drug encapsulated either in theaqueous core or in the lipid bilayer of the liposome [45].Local mild heating can be applied directly within anarrow temperature range. Moreover, it renders the cellssusceptible toward oxidative stress caused by ATOtreatment. Needham et al. [46] observed that inclusionof small fraction of lysolipids in double tail lipidliposomes causes the drug to be released quickly atphase transition temperature Tm. Winter et al. [25]described a liposomal formulation for encapsulation ofATO which efficiently released the drug on being heatedfrom 27°C to 35°C. This was achieved by incorporatinglysolipid such as monopalmitoylphosphotidylcholine(MPPC) in liposomes composed of dipalmitoylpho-sphotidylcholine (DPPC). 10 mol% of MPPC enabled avery rapid release of encapsulated arsenic within thefirst hour of temperature change. The ATO loading was

Drug delivery of encapsulated arsenic trioxide for solid cancers 7

1

5

10

15

20

25

30

35

40

45

50

1

5

10

15

20

25

30

35

40

45

50

JBR-16059-AA.3d 14/9/016 16:40:34

Page 9: Middlesex University Research Repository for the review of ATO.pdf · levels, in one experiment, glutathione content was increased by treating the cells with N-acetylcysteine. The

accomplished using the same principle as described inthe nanobin above.

Arsonoliposomes

Arsonoliposomes are nanosized liposomes consti-tuted from arsonolipids, which are analogs of phospho-nolipids where arsenic replaces the phosphorus in thelipid polar head group [47]. This type of nanoparticles isdesigned to combine the anti-cancer properties ofarsenic with the enhanced efficacy and reduced toxicityof liposomes. Arsonolipids, when employed on theirown, do not possess anti-cancer activity adequateenough to be of therapeutic value [48]. However, onbeing incorporated in liposomes, they demonstrateincreased toxicity to cancer cells while being lesstoxic to normal cells. Phosphotidyl choline (PC)-basedarsonoliposomes suffered from rapid clearance fromblood circulation after in vivo administration, alongwith their aggregating and fusing into larger particles.However, the PEGylation of arsonoliposomes was ableto enhance their bioavailability and pharmacokineticprofile and was shown to be effective against brainglioma, pituitary and leukemia cell lines [27].

Polymersome encapsulation

Polymersomes are nanoparticles constituted of blockcopolymers that are organized in a bilayer, similar toliposomes, with the hydrophobic polymer wall sur-rounding an aqueous core [44]. PEGylated PLGAnanoparticles were synthesized as drug delivery vehi-cles for ATO. Loading the nanoparticles with ATO wasachieved by slightly modified spontaneous emulsifica-tion solvent diffusion (SESD) method after transform-ing the amorphous ATO into cubic crystal form andthereby, increasing its solubility in organic solvents[28].Qian et al. [29] synthesized PEG-PDLLA vesicles

from amphiphilic diblock copolymer poly (D, L-lactide)and encapsulated arsenite ion in the nanoparticles’ core.To further improve the specificity, they conjugated anantibody fragment, i.e. anti-CD44v6 single chainvariable fragment onto the vesicles which demonstratedhigh specificity and affinity toward membrane antigenCD44v6 overexpressed on a number of epitheliumderived cancer cells. The formulated nanoparticles werethen tested against human pancreatic cancer cell linePANC-1 and were found to be very efficient indelivering the payload within the tumor site incomparison to the non-targeted ones.A polymeric micelle system was developed recently

by Zhang et al. [30] utilizing a biodegradable block

copolymer. Free thiol groups on the hydrophobic blockof the polymer backbone were used to conjugate aneven more toxic form of trivalent arsenic speciescompared to ATO, i.e. phenylarsine oxide (PAO). 65%of free thiol groups were reported to conjugate withPAO whose release from the formulated nanoparticleswas triggered by the presence of glutathione, whichperhaps acted as a competing agent for thiol group. PAOencapsulation further served to improve micellarstability and prevented micellar dissociation in wateror in the presence of detergents like SDS [17].In addition to the above, a polymeric shell was also

employed in caging ATO encapsulating liposome“nanobin” as described previously. The pH responsivepolymeric network surrounding the nano-sized lipo-some allowed the simultaneous release of membranepermeable trivalent arsenic ions along with membraneimpermeable Ni cations to which they form a complex.When the pH is reduced to the levels found in lateendosomes or tumor acidic milieu, both ions werereleased simultaneously, unlike nanobins, where nickelions remained inside the liposome while the arsenicdiffused out [49]. This is an attractive strategy becausenickel ions have been known to deplete intracellularglutathione and ascorbate levels, thereby rendering thecells even more susceptible toward ATO toxicity [26].The cytotoxicity of these polymer caged nanobins wastested against HeLa cells and they were found to bemore potent in inducing apoptosis than the plainliposomes. The triggered release from pH change wasdue to the collapse of the cross-linked polymericnetwork which creates pressure points, hence pressingliposomes and consequently destabilizing their lipidbilayer membrane, making them release their payload[49].

Nanoparticle mediated siRNA and arsenicco-therapy

Zeng et al. [36] demonstrated the potential synergisticeffect of silencing mutant KRAS gene and arsenictherapy on pancreatic tumor cell lines with excellentresults. Mutant KRAS gene plays an important role incell transformation and cancer development and isknown to be activated in a number of pancreaticcancers. Therefore, downregulating it with si-RNA genetherapy becomes an attractive treatment which gavepositive results such as inhibition of proliferative,migratory and invasive pancreatic tumor cells [50].When co-treated with ATO, the apoptotic effect of thelatter was substantially improved. Arsenic was encap-sulated in poly(ethylene glycol)-block-poly(DL-lactide)

8 Akhtar A et al. J Biomed Res, 2017, 31

1

5

10

15

20

25

30

35

40

45

50

1

5

10

15

20

25

30

35

40

45

50

JBR-16059-AA.3d 14/9/016 16:40:34

Page 10: Middlesex University Research Repository for the review of ATO.pdf · levels, in one experiment, glutathione content was increased by treating the cells with N-acetylcysteine. The

vesicles and siRNA-complexed polyplexes wereformed from poly(ethylene glycol)-block-poly(L-lysine) polymer. Both of the polymer nanoparticleswere simultaneously administered to the human pan-creatic cancer cell lines PANC-1 and BXPC-3 and theircytotoxicity was analyzed and substantiated [50].

Other nanoparticles

ATO Magnetic Nanoparticles

Encapsulating magnetic nanoparticles along with ananti-cancer drug has many advantages. These nanopar-ticles enable targeting under the influence of magneticfield and improve drug’s pharmacokinetics profile invivo. In addition, these ferromagnetic nanoparticles canalso be utilized for inducing hyperthermia in the tumortissue. Magnetic fluid (fluid comprised of superpar-amagnetic nanoparticles) absorbs much higher energythan usual materials and transfers all of it to the tumortissue where temperature may rise to 45°C. Thisconcept, magnetic fluid hyperthermia (MFH), wheremagnetic nanoparticles are heated by applying alternat-ing magnetic field is gradually becoming an attractiveanti-cancer treatment [33]. By behaving as localizedpoints of heat, they may prevent the damage to healthytissues as is commonly induced in conventionalhyperthermia treatment.In a previous study, Fe3O4 magnetic nanoparticles

were synthesized with ATO utilizing co-precipitationand impregnation techniques to form nanosized As2O3-Fe3O4 complexes. These nanoparticles were directlyinjected into the xenograft cancer tissue of humancervical cancer HeLa cells in nude mice and were foundto be internalised by the cancer cells. When combinedwith MFH, these particles attained a steady rise intemperature ranging from 42° to 65°C which success-fully ablated the tissue [37]. In another study, As2O3-MgFe2O4nanoparticles were synthesized via solventdisplacement methods and characterized [51].When such a drug- magnetic nanoparticle complex

was co-encapsulated in a thermosensitive liposome, theheat from these magnetic nanoparticles could lead todestabilizing of the liposomal membrane, and the drugcould then be released at the specified location. Li et al.[33] successfully used magnetic hyperthermia andthermosensitive liposomes in conjunction by co-encap-sulating Mn-Zn ferrite magnetic nanoparticles withATO in liposomes. Upon exposure to alternatingmagnetic field, the Mn-Zn ferrite nanoparticles servedas the heating source, causing the lipids in liposomalmembrane to reach their transition temperatures andthereby release ATO. Their efficacy was tested againstthe human breast cancer MDA-MB 231 cell line and a

significant inhibition of the cancer cells growth wasobserved [33].Song et al. [52] prepared magnetic core-polymer shell

drug delivery system encapsulating ATO. The magneticcore employed was a small magnetic nanoparticle likeFe3O4and the shell was composed of a biocompatibleand biodegradable polymer. This ATO drug deliveringsystem has been utilizing double emulsion-solventevaporation technique by coating the magnetite withthe polymer poly lactic acid (PLA). These hybridnanoparticles had high encapsulation efficiency andwere effective in achieving anti-cancer response alongwith magnetic targeting.

ATO Albumin Microspheres

Bovine serum albumin microspheres containing ATOwere prepared employing chemical crosslink andsolidification method [53]. The drug loading experi-ments exhibited 29% encapsulation efficiency and therelease experiments in vitro suggested a slower releaseof ATO after an initial burst from the microspheres, witha cumulative percentage release close to 95% [54].When conjugated with cell penetrating peptides, themicrospheres could display an increased cellular uptakeand consequent intracellular delivery of ATO into thecells. For this purpose, trans-activating transcriptionalactivator (Tat) peptide was employed and covalentlylinked to the albumin microspheres for successfuldelivering ATO into the bladder cells [55].

ATO loaded Silica Nanoparticles

Magnetite doped mesoporous silica nanoparticles(MSN) were recently fabricated where both the internalporous surface and external surface could be utilized forloading different carcinogenic drugs. Moreover, theypossessed further attractive features including a largesurface area, ease of surface functionalization, biocom-patibility and stimuli responsive drug release. Muham-mad et al. [34] loaded camptothecin, a poorlyhydrophobic drug, into the pores while conjugatingATO onto the surfaces of the designed silica nanopar-ticles utilizing the interaction between arsenic andsulphydryl groups. Besides, superparamagnetic magne-tite nanocrystals tethered onto their surface served twoimportant purposes. They allowed the nanoparticles todeliver drug payload to the site of action and alsoenabled monitoring of therapeutic response via mag-netic resonance imaging (MRI). This dual drug nano-formulation exhibited synergism with very low con-centrations of nanoparticles showing cytotoxic effect tocancer cells.

Drug delivery of encapsulated arsenic trioxide for solid cancers 9

1

5

10

15

20

25

30

35

40

45

50

1

5

10

15

20

25

30

35

40

45

50

JBR-16059-AA.3d 14/9/016 16:40:34

Page 11: Middlesex University Research Repository for the review of ATO.pdf · levels, in one experiment, glutathione content was increased by treating the cells with N-acetylcysteine. The

Arsenoplatins

Arsenoplatins are small molecule complexes ofaqueous form of ATO linked chemically to Pt(II).They are formed when cisplatin, an inorganic drug, isheated with ATO in an acetonitrile mixture for threedays [34]. The rationale behind their synthesis is thatcisplatin, cis-diamminedichloroplatinum(II), widelyused to treat ovarian, head, neck, bladder and testicularcancers, shows synergistic activity when used alongsidewith ATO. Both of them induce apoptosis in cancer cellsalbeit with different mechanisms, cisplatin by causinginter and intra-DNA crosslinking and arsenic by themechanisms listed above. Miodragović et al. [34]reported the formation of arsenoplatins, which arestable in solution, display superior anti-cancer activityand also overcome drug resistance which is a majorlimitation of platinum drugs. The As-Pt core is suitablefor ligand substitution reactions and its efficacy hasbeen demonstrated for a variety of cisplatin-resistantcancer cell lines, such as cisplatin resistant ovarian,colon and glioblastoma cancer cell lines.

Others

Wimmer et al. [31] developed a glycol lipid arsenicaland reported its anti-proliferative effects on breastcancer cells MCF-7.This compound was formed bychemically attaching lipoamino acid to sugar. Theformer increased the lipophilicity of the compound andaugmented arsenic’s toxic effects. In addition, realgar(As4S4) quantum dots have been produced recently bothas a colloidal dispersion and as a hydrogel; the latter ispH responsive and both forms have been reported topossess superior anti-tumor properties [8]. Mostrecently, chitosan, a well-known surface modifier, wasreported to be used for coating ATO nanoparticles. Theresulting nanoparticles were biocompatible and had animproved stability and toxicity profile against prostatecancer [38]. Additionally, these nanoparticles inducedcell cycle arrest by p21 WAF/CIP1 expression viaepigenetic remodelling in androgen dependent andindependent cell lines [56].

Conclusion

Despite an increasing awareness of toxicity arisingfrom arsenic compounds, ATO has established itself asan effective, broad spectrum anti-cancer drug. Follow-ing its successful establishment as the front linetreatment for APL patients, researchers and clinicianshave been working toward improving its effectivenessin treatment and reducing its side effects for solid

cancers. Its poor bioavailability due to rapid clearancefrom blood by RES limits the therapeutic dose ofarsenic reaching the tumor site. Therefore, usingnanotechnology to facilitate ATO delivery into solidtumors becomes the focus in this field. Up to date,immense progress has been made in delivering ATOcompounds specifically to the cancer cells. ATO alongwith other arsenic compounds has successfully beenencapsulated in liposomes, polymersomes and othernanoparticles. In addition, ligands unique to thereceptors overexpressed on specific cancer cells havebeen conjugated to these nanoparticles. Hence, it ispromising that delivering ATO using nanotechnologywould be employed as an anti-cancer drug option intreating a variety of solid cancers with better efficacyand much less toxicity.

References:

[1] Shen S, Li XF, Cullen WR, et al. Arsenic binding to proteins.

Chem Rev, 2013, 113(10): 7769–7792.

[2] Miller WHJr, Schipper HM, Lee JS, et al. Mechanisms of action

of arsenic trioxide. Cancer Res, 2002, 62(14): 3893–3903.

[3] Wang S, Wu X, Tan M, et al. Fighting fire with fire: poisonous

Chinese herbal medicine for cancer therapy. J Ethnopharmacol,

2012, 140(1): 33–45.

[4] Evens AM, Tallman MS, Gartenhaus RB. The potential of

arsenic trioxide in the treatment of malignant disease: past,

present, and future. Leuk Res, 2004, 28(9): 891–900.

[5] Shen ZX, Chen GQ, Ni JH, et al. , and the Clinical Efficacy and

Pharmacokinetics in Relapsed Patients. Use of arsenic trioxide

(As2O3) in the treatment of acute promyelocytic leukemia

(APL): II. Clinical efficacy and pharmacokinetics in relapsed

patients. Blood, 1997, 89(9): 3354–3360.

[6] Antman KH. Introduction: the history of arsenic trioxide in

cancer therapy. Oncologist, 2001, 6(2Suppl 2): 1–2.

[7] Zhang P, Wang SY, Hu XH. Arsenic trioxide treated 72 cases of

acute promyelocytic leukemia. Chinese Journal of Hematology,

1996, 17(1): 58–62.

[8] Swindell EP, Hankins PL, Chen H, et al. Anticancer activity of

small-molecule and nanoparticulate arsenic(III) complexes.

InorgChem, 2013, 52(21): 12292–12304.

[9] Kang YH, Lee SJ. Role of p38 MAPK and JNK in enhanced

cervical cancer cell killing by the combination of arsenic

trioxide and ionizing radiation. Oncol Rep, 2008, 20(3): 637–

643.

[10] Zhang L, Zhang Z, Mason RP, et al. Convertible MRI contrast:

Sensing the delivery and release of antigliomanano-drugs. Sci

Rep, 2015, 5: 1–13.

[11] Emadi A, Gore SD. Arsenic trioxide- An old drug rediscovered.

Blood Rev, 2010, 24(4-5): 191–199.

[12] Grad JM, Bahlis NJ, Reis I, et al. Ascorbic acid enhances

arsenic trioxide-induced cytotoxicity in multiple myeloma cells.

10 Akhtar A et al. J Biomed Res, 2017, 31

1

5

10

15

20

25

30

35

40

45

50

1

5

10

15

20

25

30

35

40

45

50

JBR-16059-AA.3d 14/9/016 16:40:34

Page 12: Middlesex University Research Repository for the review of ATO.pdf · levels, in one experiment, glutathione content was increased by treating the cells with N-acetylcysteine. The

Blood, 2001, 98(3): 805–813.

[13] Gartenhaus RB, Prachand SN, Paniaqua M, et al. Arsenic

trioxide cytotoxicity in steroid and chemotherapy-resistant

myeloma cell lines: enhancement of apoptosis by manipulation

of cellular redox state. Clin Cancer Res, 2002, 8(2): 566–572.

[14] Waxman S, Anderson KC. History of the development of

arsenic derivatives in cancer therapy. Oncologist, 2001, 6

(2Suppl 2): 3–10.

[15] Murgo AJ. Clinical trials of arsenic trioxide in hematologic and

solid tumors: overview of the National Cancer Institute

Cooperative Research and Development Studies. Oncologist,

2001, 6(2Suppl 2): 22–28.

[16] Platanias LC. Biological responses to arsenic compounds. J

BiolChem, 2009, 284(28): 18583–18587.

[17] Zhao S, Zhang X, Zhang J, et al. Intravenous administration of

arsenic trioxide encapsulated in liposomes inhibits the growth

of C6 gliomas in rat brains. J Chemother, 2008, 20(2): 253–

262.

[18] Maeda H, Hori S, Ohizumi H, et al. Effective treatment of

advanced solid tumors by the combination of arsenic trioxide

and L-buthionine-sulfoximine. Cell Death Differ, 2004, 11(7):

737–746.

[19] Kim HR, Kim EJ, Yang SH, et al. Combination treatment with

arsenic trioxide and sulindac augments their apoptotic potential

in lung cancer cells through activation of caspase cascade and

mitochondrial dysfunction. Int J Oncol, 2006, 28(6): 1401–

1408.

[20] Ahn RW, Barrett SL, Raja MR, et al. Nano-encapsulation of

arsenic trioxide enhances efficacy against murine lymphoma

model while minimizing its impact on ovarian reserve in vitro

and in vivo. PLoS One, 2013, 8(3): e58491.

[21] Ahn RW, Chen F, Chen H, et al. A novel nanoparticulate

formulation of arsenic trioxide with enhanced therapeutic

efficacy in a murine model of breast cancer. Clin Cancer Res,

2010, 16(14): 3607–3617.

[22] Chen H, MacDonald RC, Li S, et al. Lipid encapsulation of

arsenic trioxide attenuates cytotoxicity and allows for con-

trolled anticancer drug release. J Am ChemSoc, 2006, 128(41):

13348–13349.

[23] Zhang Y, Kenny HA, Swindell EP, et al. Urokinase plasmino-

gen activator system-targeted delivery of nanobins as a novel

ovarian cancer therapy. Mol Cancer Ther, 2013, 12(12): 2628–

2639.

[24] Wang X, Li D, Ghali L, et al. Therapeutic potential of delivering

arsenic trioxide into HPV-infected cervical cancer cells using

liposomal nanotechnology. Nanoscale Res Lett, 2016, 11(1):

94.

[25] Winter ND, Murphy RKJ, O’Halloran TV, et al. Development

and modeling of arsenic-trioxide-loaded thermosensitive lipo-

somes for anticancer drug delivery. J Liposome Res, 2011, 21

(2): 106–115.

[26] Chen H, Ahn R, Van den Bossche J, et al. Folate-mediated

intracellular drug delivery increases the anticancer efficacy of

nanoparticulate formulation of arsenic trioxide. Mol Cancer

Ther, 2009, 8(7): 1955–1963.

[27] Gortzi O, Papadimitriou E, Kontoyannis CG, et al. Arsonolipo-

somes, a novel class of arsenic-containing liposomes: effect of

palmitoyl-arsonolipid-containing liposomes on the viability of

cancer and normal cells in culture. Pharm Res, 2002, 19(1): 79–

86.

[28] Wang Z, Liu W, Xu H, et al. Preparation and in vitro Studies of

Stealth PEGylated PLGA Nanoparticles as Carriers for Arsenic

Trioxide. Chin J ChemEng, 2007, 15(6): 795–801.

[29] Qian C, Wang Y, Chen Y, et al. Suppression of pancreatic

tumor growth by targeted arsenic delivery with anti-CD44v6

single chain antibody conjugated nanoparticles. Biomaterials,

2013, 34(26): 6175–6184.

[30] Zhang Q, Vakili MR, Li XF, et al. Polymeric micelles for GSH-

triggered delivery of arsenic species to cancer cells. Biomater-

ials, 2014, 35(25): 7088–7100.

[31] Wimmer N, Robinson JA, Gopisetty-Venkatta N, et al. Novel

Glyco-lipid-arsenicals (III) with Anti-proliferative Effects on

MCF-7 Human Breast Cancer Cells. Understanding Biology

Using Peptides.Springer New York, 2006, 365–366.

[32] Lee SM, Chen H, O’Halloran TV, et al. “Clickable” polymer-

caged nanobins as a modular drug delivery platform. J Am

ChemSoc, 2009, 131(26): 9311–9320.

[33] Wang L, Wang Z, Liu J, et al. Preparation of a new nanosized

As2O3/Mn0.5Zn0.5Fe2O4 thermosensitive magnetoliposome and

its antitumor effect on MDA_MB_231 cells. J NanosciNano-

technol, 2011, 11(12): 10755–10759.

[34] Miodragović ÐU, Quentzel JA, Kurutz JW, et al. Robust

structure and reactivity of aqueous arsenous acid-platinum(II)

anticancer complexes. AngewChemInt Ed Engl, 2013, 52(41):

10749–10752.

[35] Muhammad F, Zhao J, Wang N, et al. Lethal drug combination:

arsenic loaded multiple drug mesoporous silica for theranostic

applications. Colloids Surf B Biointerfaces, 2014, 123: 506–

514.

[36] Zeng L, Li J, Wang Y, et al. Combination of siRNA-directed

Kras oncogene silencing and arsenic-induced apoptosis using a

nanomedicine strategy for the effective treatment of pancreatic

cancer. Nanomedicine, 2014, 10(2): 463–472.

[37] Du Y, Zhang D, Liu H, et al. Thermochemotherapy effect of

nanosized As2O3/Fe3O4 complex on experimental mouse

tumors and its influence on the expression of CD44v6, VEGF-C

and MMP-9. BMC Biotechnol, 2009, 9(1): 84.

[38] Jadhav V, Sachar S, Chandra S, et al. Synthesis and

Characterization of Arsenic Trioxide Nanoparticles and Their

In Vitro Cytotoxicity Studies on Mouse Fibroblast and Prostate

Cancer Cell Lines. J NanosciNanotechnol, 2015, 15: 1–7.

[39] Zhao Z, Zhang H, Chi X, et al. Silica nanovehicles endow

arsenic trioxide with an ability to effectively treat cancer cells

and solid tumours. J Mater Chem B Mater Biol Med, 2014, 2

(37): 6313–6323.

[40] Milas L. Cyclooxygenase-2 (COX-2) enzyme inhibitors as

Drug delivery of encapsulated arsenic trioxide for solid cancers 11

1

5

10

15

20

25

30

35

40

45

50

1

5

10

15

20

25

30

35

40

45

50

JBR-16059-AA.3d 14/9/016 16:40:34

Page 13: Middlesex University Research Repository for the review of ATO.pdf · levels, in one experiment, glutathione content was increased by treating the cells with N-acetylcysteine. The

potential enhancers of tumor radioresponse. SeminRadiatOn-

col, 2001, 11(4): 290–299.

[41] Soriano AF, Helfrich B, Chan DC, et al. Synergistic effects of

new chemopreventive agents and conventional cytotoxic agents

against human lung cancer cell lines. Cancer Res, 1999, 59(24):

6178–6184.

[42] Mandegary A, Torshabi M, Seyedabadi M, et al. Indomethacin-

Enhanced Anticancer Effect of Arsenic Trioxide in A549 Cell

Line: Involvement of Apoptosis and Phospho-ERK and p38

MAPK Pathways. BioMed Research International,2013, 2013:

1–9.

[43] Farokhzad OC, Langer R. Impact of nanotechnology on drug

delivery. ACS Nano, 2009, 3(1): 16–20.

[44] Soussan E, Cassel S, Blanzat M, et al. Drug delivery by soft

matter: matrix and vesicular carriers. AngewChemInt Ed Engl,

2009, 48(2): 274–288.

[45] Griffin RJ, Monzen H, Williams BW, et al. Arsenic trioxide

induces selective tumour vascular damage via oxidative stress

and increases thermosensitivity of tumours. Int J Hyperthermia,

2003, 19(6): 575–589.

[46] Needham D, Ponce AM, Wright A, et al. Targeted bioavail-

ability of drugs by triggered release from liposomes. Future

Lipidol, 2006, 1(1): 25–34.

[47] Koutsopoulos S, Fatouros DG, Ioannou PV, et al. Thermal

behavior of novel non-sonicated arsonolipid-containing lipo-

somes. BiophysChem, 2006, 121(2): 150–154.

[48] Fatouros DG, Ioannou PV, Antimisiaris SG. Arsonoliposomes:

novel nanosized arsenic-containing vesicles for drug delivery. J

NanosciNanotechnol, 2006, 6(9-10): 2618–2637.

[49] Salnikow K, Donald SP, Bruick RK, et al. Depletion of

intracellular ascorbate by the carcinogenic metals nickel and

cobalt results in the induction of hypoxic stress. J BiolChem,

2004, 279(39): 40337–40344.

[50] Jones S, Zhang X, Parsons DW, et al. Core signaling pathways

in human pancreatic cancers revealed by global genomic

analyses. Science, 2008, 321(5897): 1801–1806.

[51] Yang Guo-Fu, LXH, Zhao Zhe, et al. Characterization and

Tissue Pharmacokinetics in Mice of Arsenic Trioxide Magnetic

Nanoparticles. Chemical Journal of Chinese Universi-

ties,2010, 5: 010.

[52] Song X, You J, Wang J, et al. Preparation and investigation of

arsenic trioxide-loaded polylactic acid/magnetic hybrid nano-

particles. Chem Res Chin Univ, 2014, 30(2): 326–332.

[53] Yang Z, Yang M, Peng J. Evaluation of arsenic trioxide-loaded

albumin nanoparticles as carriers: preparation and antitumor

efficacy. Drug Dev Ind Pharm, 2008, 34(8): 834–839.

[54] Zhou J, Zeng F, Xiang G, et al. Preparation of arsenic trioxide

albumin microspheres and its release characteristics in vitro.

[Medical Sciences]. J HuazhongUnivSciTechnolog Med Sci,

2005, 25(3): 310–312., 319.

[55] Zhou J, Wang QH, Liu JH, et al. Effects of Tat peptide on

intracellular delivery of arsenic trioxide albumin microspheres.

Anticancer Drugs, 2012, 23(3): 303–312.

[56] Jadhav V, Ray P, Sachdeva G, et al. Biocompatible arsenic

trioxide nanoparticles induce cell cycle arrest by p21(WAF1/

CIP1) expression via epigenetic remodeling in LNCaP and PC3

cell lines. Life Sci, 2016, 148: 41–52.

12 Akhtar A et al. J Biomed Res, 2017, 31

1

5

10

15

20

25

30

35

40

45

50

1

5

10

15

20

25

30

35

40

45

50

JBR-16059-AA.3d 14/9/016 16:40:35