+ All Categories
Home > Documents > Mitochondrial-Targeting Nitrooxy-doxorubicin: A New Approach To Overcome Drug Resistance

Mitochondrial-Targeting Nitrooxy-doxorubicin: A New Approach To Overcome Drug Resistance

Date post: 10-Dec-2016
Category:
Upload: dario
View: 215 times
Download: 2 times
Share this document with a friend
14
Mitochondrial-Targeting Nitrooxy-doxorubicin: A New Approach To Overcome Drug Resistance Chiara Riganti,* ,,Barbara Rolando, § Joanna Kopecka, ,Ivana Campia, ,Konstantin Chegaev, § Loretta Lazzarato, § Antonella Federico, § Roberta Fruttero, § and Dario Ghigo ,Department of Oncology and Center for Experimental Research and Medical Studies (CeRMS), University of Torino, via Santena 5/bis, 10126, Torino, Italy § Department of Science and Drug Technology, University of Torino, via Pietro Giuria 9, 10125, Torino, Italy * S Supporting Information ABSTRACT: In previous studies, we showed that nitric oxide (NO) donors and synthetic doxorubicins (DOXs) modied with moieties containing NO-releasing groupssuch as nitrooxy-DOX (NitDOX) or 3-phenylsulfonylfuroxan-DOX (FurDOX)overcome drug resistance by decreasing the activity of ATP-binding cassette (ABC) transporters that can extrude the drug. Here, we have investigated the biochemical mechanisms by which NitDOX and FurDOX exert antitumor eects. Both NitDOX and FurDOX were more cytotoxic than DOX against drug-resistant cells. Interestingly, NitDOX exhibited a faster uptake and an extranuclear distribution. NitDOX was preferentially localized in the mitochondria, where it nitrated and inhibited the mitochondria-associated ABC transporters, decreased the ux through the tricarboxylic acid cycle, slowed down the activity of complex I, lowered the synthesis of ATP, induced oxidative and nitrosative stress, and elicited the release of cytochrome c and the activation of caspase-9 and -3 in DOX-resistant cells. We suggest that NitDOX may represent the prototype of a new class of multifunctional anthracyclines, which have cellular targets dierent from conventional anthracyclines and greater ecacy against drug-resistant tumors. KEYWORDS: multifunctional drugs, doxorubicin, nitric oxide, multidrug resistance, ATP-binding cassette transporters, mitochondria INTRODUCTION Doxorubicin (DOX), also known as adriamycin, is an antibiotic belonging to the class of anthracyclines, widely used to treat solid and hematological malignancies. The molecular mecha- nisms that underlie the DOX anticancer eects include DNA damage, inhibition of topoisomerase II, induction of oxidative stress [synthesis of reactive oxygen species (ROS) and reactive nitrogen species (RNS)], altered metabolism of Ca 2+ ions, and activation of the host immune system against the tumor. 1 The limitations to DOX ecacy in cancer therapy are of two types: cardiotoxicity, which can occur as both acute and chronic dose-related forms, and development of resistance through dierent mechanisms, the main of which is the overexpression of ATP-binding cassette (ABC) transporters, such as P- glycoprotein (Pgp/ABCB1), multidrug resistance-associated proteins (MRPs/ABCCs), and breast cancer resistance protein (BCRP/ABCG2), which actively extrude the drug from tumor cells. 2 Resistance to DOX is often part of a cross-resistance toward several anticancer drugs known as multidrug resistance (MDR), which aects up to 70% tumors at diagnosis and increases in relapses and metastasis. 3 MDR also aects new molecularly targeted drugs, 4 and until now, no satisfactory reversing strategies have been identied. Previous works from our group showed that nitric oxide (NO) donors, such as sodium nitroprusside (SNP), S- nitrosopenicillamine, S-nitrosoglutathione, 5,6 and furoxan de- rivatives (1,2,5-oxadiazole 2-oxides), 7 reverse MDR in human cancer cells. These compounds decreased the activity of Pgp/ ABCB1 and MRPs/ABCCs pumps by nitrating critical tyrosines, thus allowing the increase of intracellular DOX accumulation and toxicity. 5,6 NO is a small molecule involved in the regulation of vascular tone, aggregation of platelets, angiogenesis, cell death, dier- entiation, neurotransmission, and activity of the immune system. 8 It is synthesized from L-arginine by three NO synthase (NOS; EC 1.14.13.39) enzymes. 9 The role of NO in tumors, where it can be produced by cancer cells or by inltrating macrophages, 10 is a matter of huge debate. 11 At nanomolar concentrations, NO is a tumor-growth supporter. 12,13 At micromolar concentrations, as occurs after the up-regulation of inducible NOS (iNOS/NOS II), NO is an antitumor agent, which elicits oxidative damage on DNA, inactivates DNA- repairing systems, induces apoptosis and endoplasmic reticulum stress, hampers the mitochondrial energy metabolism, exerts antiangiogenic eects, and enhances the reaction of host Received: June 7, 2012 Revised: November 16, 2012 Accepted: November 27, 2012 Published: November 27, 2012 Article pubs.acs.org/molecularpharmaceutics © 2012 American Chemical Society 161 dx.doi.org/10.1021/mp300311b | Mol. Pharmaceutics 2013, 10, 161-174
Transcript
Page 1: Mitochondrial-Targeting Nitrooxy-doxorubicin: A New Approach To Overcome Drug Resistance

Mitochondrial-Targeting Nitrooxy-doxorubicin: A New Approach ToOvercome Drug ResistanceChiara Riganti,*,†,‡ Barbara Rolando,§ Joanna Kopecka,†,‡ Ivana Campia,†,‡ Konstantin Chegaev,§

Loretta Lazzarato,§ Antonella Federico,§ Roberta Fruttero,§ and Dario Ghigo†,‡

†Department of Oncology and ‡Center for Experimental Research and Medical Studies (CeRMS), University of Torino, via Santena5/bis, 10126, Torino, Italy§Department of Science and Drug Technology, University of Torino, via Pietro Giuria 9, 10125, Torino, Italy

*S Supporting Information

ABSTRACT: In previous studies, we showed that nitric oxide(NO) donors and synthetic doxorubicins (DOXs) modifiedwith moieties containing NO-releasing groupssuch asnitrooxy-DOX (NitDOX) or 3-phenylsulfonylfuroxan-DOX(FurDOX)overcome drug resistance by decreasing theactivity of ATP-binding cassette (ABC) transporters that canextrude the drug. Here, we have investigated the biochemicalmechanisms by which NitDOX and FurDOX exert antitumoreffects. Both NitDOX and FurDOX were more cytotoxic than DOX against drug-resistant cells. Interestingly, NitDOX exhibiteda faster uptake and an extranuclear distribution. NitDOX was preferentially localized in the mitochondria, where it nitrated andinhibited the mitochondria-associated ABC transporters, decreased the flux through the tricarboxylic acid cycle, slowed down theactivity of complex I, lowered the synthesis of ATP, induced oxidative and nitrosative stress, and elicited the release ofcytochrome c and the activation of caspase-9 and -3 in DOX-resistant cells. We suggest that NitDOX may represent theprototype of a new class of multifunctional anthracyclines, which have cellular targets different from conventional anthracyclinesand greater efficacy against drug-resistant tumors.

KEYWORDS: multifunctional drugs, doxorubicin, nitric oxide, multidrug resistance, ATP-binding cassette transporters, mitochondria

■ INTRODUCTION

Doxorubicin (DOX), also known as adriamycin, is an antibioticbelonging to the class of anthracyclines, widely used to treatsolid and hematological malignancies. The molecular mecha-nisms that underlie the DOX anticancer effects include DNAdamage, inhibition of topoisomerase II, induction of oxidativestress [synthesis of reactive oxygen species (ROS) and reactivenitrogen species (RNS)], altered metabolism of Ca2+ ions, andactivation of the host immune system against the tumor.1

The limitations to DOX efficacy in cancer therapy are of twotypes: cardiotoxicity, which can occur as both acute and chronicdose-related forms, and development of resistance throughdifferent mechanisms, the main of which is the overexpressionof ATP-binding cassette (ABC) transporters, such as P-glycoprotein (Pgp/ABCB1), multidrug resistance-associatedproteins (MRPs/ABCCs), and breast cancer resistance protein(BCRP/ABCG2), which actively extrude the drug from tumorcells.2 Resistance to DOX is often part of a cross-resistancetoward several anticancer drugs known as multidrug resistance(MDR), which affects up to 70% tumors at diagnosis andincreases in relapses and metastasis.3 MDR also affects newmolecularly targeted drugs,4 and until now, no satisfactoryreversing strategies have been identified.Previous works from our group showed that nitric oxide

(NO) donors, such as sodium nitroprusside (SNP), S-

nitrosopenicillamine, S-nitrosoglutathione,5,6 and furoxan de-rivatives (1,2,5-oxadiazole 2-oxides),7 reverse MDR in humancancer cells. These compounds decreased the activity of Pgp/ABCB1 and MRPs/ABCCs pumps by nitrating criticaltyrosines, thus allowing the increase of intracellular DOXaccumulation and toxicity.5,6

NO is a small molecule involved in the regulation of vasculartone, aggregation of platelets, angiogenesis, cell death, differ-entiation, neurotransmission, and activity of the immunesystem.8 It is synthesized from L-arginine by three NO synthase(NOS; EC 1.14.13.39) enzymes.9 The role of NO in tumors,where it can be produced by cancer cells or by infiltratingmacrophages,10 is a matter of huge debate.11 At nanomolarconcentrations, NO is a tumor-growth supporter.12,13 Atmicromolar concentrations, as occurs after the up-regulationof inducible NOS (iNOS/NOS II), NO is an antitumor agent,which elicits oxidative damage on DNA, inactivates DNA-repairing systems, induces apoptosis and endoplasmic reticulumstress, hampers the mitochondrial energy metabolism, exertsantiangiogenic effects, and enhances the reaction of host

Received: June 7, 2012Revised: November 16, 2012Accepted: November 27, 2012Published: November 27, 2012

Article

pubs.acs.org/molecularpharmaceutics

© 2012 American Chemical Society 161 dx.doi.org/10.1021/mp300311b | Mol. Pharmaceutics 2013, 10, 161−174

Page 2: Mitochondrial-Targeting Nitrooxy-doxorubicin: A New Approach To Overcome Drug Resistance

immune system against the tumor.14 Beside its MDR-reversingproperties, NO can enhance the efficacy of radiotherapy15 andchemotherapy.16,17

We have previously produced synthetic DOXs containingNO-releasing groups, such as nitrooxy-DOX (NitDOX;Supplementary Figure S1 in the Supporting Information) and3-phenylsulfonylfuroxan-DOX (FurDOX; Supplementary Fig-ure S1 in the Supporting Information), able to nitrate theMRP3/ABCC3 pump and to reduce the efflux of DOX in drug-resistant cells at low micromolar concentrations.18 The NO-releasing groups selected differ in terms of kinetics and extentof NO released. The furoxan system can release NO under theaction of endogenous thiols with a mechanism not yet fullyunderstood; by contrast, NO release from organic nitratesoccurs through enzymatic catalysis. A number of enzymes havebeen proposed for this conversion: in particular, the role ofmitochondrial aldehyde dehydrogenase and P450 enzyme(s)has been emphasized.18

The use of multitarget drugs or multifunctional drugs,namely, single compounds capable of interacting simulta-neously with more than one target, has a lower risk of drug−drug interactions, improves compliance by the patient, andshows a more predictable pharmacokinetic profile as comparedwith the association of drugs.19 The development of resistancetoward multitarget drugs is also expected to be more difficult.In addition, the NO-releasing multifunctional drugs should takeadvantage from the anticancer effects that both NO and DOXhave and maximize the antitumor efficacy.The aim of this work was to investigate how the presence of

either the nitrooxy- or the 3-phenylsulfonylfuroxan group mayimprove and/or change the anticancer activity of DOX. Wefound that NitDOX is a lead compound with an unexpectedand novel mechanism of action, which makes it ananthracycline exhibiting pharmacologic properties widely differ-ent from DOX.

■ EXPERIMENTAL SECTIONChemicals. Fetal bovine serum (FBS) and culture medium

were supplied by Invitrogen Life Technologies (Carlsbad, CA);plasticware for cell cultures was from Falcon (BectonDickinson, Franklin Lakes, NJ). Electrophoresis reagents wereobtained from Bio-Rad Laboratories (Hercules, CA); theprotein content of cell monolayers and lysates was assessedwith the BCA kit from Sigma Chemical Co. (St. Louis, MO).Those not specified were purchased from Sigma Chemical Co.The syntheses of NitDOX and FurDOX were performed asdescribed previously.18 The structures are reported inSupplementary Figure S1 in the Supporting Information,along with the methyl ester of acid containing nitrooxy moiety(NitE) and the methyl ester of acid containing phenyl-sulfonylfuroxan moiety (FurE).Cell Lines. Human colon cancer DOX-sensitive HT29 cells

were cultured in RPMI 1640 medium. A subpopulation ofDOX-resistant HT29 cells, named HT29-dx, was created aspreviously reported5 and subsequently cultured in RPMI 1640medium containing 200 nmol/L DOX. Human DOX-sensitivelung cancer A549 cells and human DOX-resistant lung cancerA549-dx cells (selected from the parental A549 cells after 30passages in the presence of 50 nmol/L DOX) were grown inHam's F12 medium. Human DOX-sensitive chronic leukemiaK562 cells and human DOX-resistant K562-dx cells (selectedfrom the parental cells after 30 passages in the presence of 25pmol/L DOX) were grown in RPMI 1640 medium. Non-

transformed mesothelial Met5A cells were cultured in RPMI1640; primary malignant mesothelioma MM98 cells, whichhave a constitutively resistant phenotype,20 were maintained inHam's F12 medium. Rat cardiomyocytes H9c2 cells weregrown in DMEM medium. Human epithelial colon CCD-18Cocells were cultured in MEM medium. All of the culture mediawere supplemented with 10% FBS, 1% penicillin−streptomy-cin, and 1% L-glutamine. Cell cultures were maintained in ahumidified atmosphere at 37 °C and 5% CO2.

Western Blot Analysis. Cells were treated with boiling 0.5mL of lysis buffer (10 mmol/L Tris, 100 mmol/L NaCl, 20mmol/L KH2PO4, 30 mmol/L EDTA, 1 mmol/L EGTA, and250 mmol/L sucrose, pH 7.5). After sonication, 1 mmol/LNaVO4, 1 mmol/L NaF, 10 mmol/L dithiothreitol (DTT), andthe inhibitor cocktail set III (100 mmol/L AEBSF, 80 mmol/Laprotinin, 5 mmol/L bestatin, 1.5 mmol/L E-64, 2 mmol/Lleupeptin, and 1 mmol/L pepstatin; Calbiochem, La Jolla, CA)were added, and cell lysates were centrifuged at 13000g for 15min. Thirty micrograms of whole cell proteins was separated bySDS-PAGE and probed with the following antibodies: anti-Pgp/ABCB1 (Santa Cruz Biotechnology, Santa Cruz, CA),anti-MRP1/ABCC1 (Abcam, Cambridge, MA), anti-MRP2/ABCC2 (Abcam), anti-BCRP/ABCG2 (Santa Cruz Biotech-nology), anti-SLC22A4 (Abcam), and anti-actin (SigmaChemical Co.). After an overnight incubation, the membranewas washed with 0.1% v/v PBS-Tween and subjected for 1 h toa peroxidase-conjugated secondary antibody (diluted 1:3000 in5% w/v PBS-Tween with milk). The membrane was washedagain with PBS-Tween, and proteins were detected byenhanced chemiluminescence (Immun-Star, Bio-Rad).Ten micrograms of proteins from nuclear extracts or

mitochondrial extracts (see below) was probed with the sameantibodies, using, respectively, an anti-TATA Box BindingProtein (Santa Cruz Biotechnology) or an anti-VDAC/porin(Abcam) antibody, to check the equal control loading.To analyze the presence of nitrated proteins, the whole cell

extract was subjected to immunoprecipitation using a rabbitpolyclonal antinitrotyrosine antibody (Millipore, Bedford, MA).Immunoprecipitated proteins were separated by SDS-PAGEand probed with anti-Pgp/ABCB1, anti-MRP1/ABCC1, anti-MRP2/ABCC2, or anti-BCRP/ABCG2 antibodies, as pre-viously described.

Extracellular Lactate Dehydrogenase (LDH) Activity.To verify the cytotoxic effect of DOX, the extracellular mediumwas centrifuged at 12000g for 15 min to pellet cellular debris,whereas cells were washed with fresh medium, detached withtrypsin/EDTA, resuspended in 0.2 mL of 82.3 mmol/Ltriethanolamine phosphate-HCl (pH 7.6), and sonicated onice with two 10 s bursts. The LDH activity was measured in theextracellular medium and in the cell lysate, as reported.20 Thereaction was followed for 6 min, measuring the absorbance at340 nm with Packard EL340 microplate reader (Bio-TekInstruments, Winooski, VT), and was linear throughout thetime of measurement. Both intracellular and extracellularenzyme activities were expressed in μmol NADH oxidized/min/dish, and then, extracellular LDH activity was calculated asa percentage of the total LDH activity in the dish.

Nitrite Production. Confluent monolayers in 35 mmdiameter Petri dishes were incubated in fresh medium underthe experimental conditions indicated in the Results. Nitriteproduction was measured by adding 0.15 mL of cell culturemedium to 0.15 mL of Griess reagent in a 96-well plate, andafter a 10 min incubation at 37 °C in the dark, the absorbance

Molecular Pharmaceutics Article

dx.doi.org/10.1021/mp300311b | Mol. Pharmaceutics 2013, 10, 161−174162

Page 3: Mitochondrial-Targeting Nitrooxy-doxorubicin: A New Approach To Overcome Drug Resistance

was detected at 540 nm with a Packard EL340 microplatereader (Bio-Tek Instruments). A blank was prepared for eachexperiment in the absence of cells, and its absorbance wassubtracted from the one obtained in the presence of cells. Thenitrite concentration was expressed as nmol/mg cell proteins.Intracellular DOX Accumulation. The amount of DOX of

whole cells lysates, nuclear, and mitochondrial extracts wasmeasured fluorimetrically as described,5 using a PerkinElmerLS-5 spectrofluorimeter (PerkinElmer, Waltham, MA). Ex-citation and emission wavelengths were 475 and 553 nm,respectively. A blank was prepared in the absence of cells ineach set of experiments, and its fluorescence was subtractedfrom the one measured in each sample. The fluorescence wasconverted in nmol DOX/mg cell proteins using a calibrationcurve prepared previously.Topoisomerase II Assay. The in vitro activity of

topoisomerase II was measured using the Topoisomerase IIDrug Screening Kit (Topogen Inc., Port Orange, FL), followingthe manufacturer's instructions. Two hundred fifty nanogramsof the supercoiled pHOT1 plasmid was incubated with 5 U ofpurified human Topoisomerase IIa (TOPOGen Inc.) at 37 °Cfor 30 min, in the presence of DOX, NitDOX, or etoposide.The reaction was stopped with 10% w/v SDS, and 1 μg ofproteinase K was added in each tube. The products wereresolved on a 1% w/v agarose gels, stained with 0.01% v/vethidium bromide. The appearance of a band corresponding tolinearized plasmid was taken as an index of active topoisomer-ase II. As control of the enzyme specificity, supercoiled pHOT1was incubated in the absence of topoisomerase II and treated asreported above.DOX Uptake. Drug uptake was measured as described in ref

21 with minor modifications. Cells were washed andpreincubated for 5 min at 37 °C in uptake buffer (125mmol/L NaCl, 20 mmol/L NaHCO3, 3 mmol/L KCl, 1.8mmol/L CaCl2, 1 mmol/L KH2PO4, 1.2 mmol/L MgSO4, 10mmol/L glucose, and 10 mmol/L Hepes, pH 7.4) and thenincubated for 10 min with DOX or NitDOX in the absence orpresence of tetraethylammonium (TEA) chloride. The assaywas stopped by diluting 1:10 the sample in ice-cold uptakebuffer. Cells were detached and centrifuged at 13000g for 30 s,washed three times with 1 mL of ice-cold uptake buffer, lysed in1 mL of ethanol/0.3 N HCl, and sonicated. The DOX amountwas measured fluorimetrically as reported above.Octanol/Water Partition Coefficient Determination.

The lipophilicity of DOX and NitDOX was estimated bymeasuring their partitioning between n-octanol and waterbuffered at pH 7.4. The distribution coefficients (log D7.4s)were obtained by shake-flask technique at room temperature.The organic (n-octanol) and aqueous phase (50 mmol/Lphosphate buffer, pH 7.4; ionic strength adjusted to 0.15 mol/Lwith KCl) were mutually saturated by shaking for 4 h. Thecompounds were solubilized in the buffered aqueous phase at aconcentration of about 0.1 mmol/L, and an appropriateamount of n-octanol was added. The two phases were shakenfor about 30 min, by which time the partitioning equilibrium ofsolutes was reached, and then centrifuged (10000g 10 min).The concentration of the solutes was measured in the aqueousphase by reverse phase-HPLC. Each log D value was an averageof at least six measurements. All of the experiments wereperformed avoiding exposure to light.Confocal Microscope Analysis. Five × 105 cells were

grown on sterile glass coverslips and treated as described in theResults, then rinsed with PBS, fixed with 4% w/v

paraformaldehyde for 15 min, washed three times with PBS,and incubated with 4′,6-diamidino-2-phenylindole dihydro-chloride (DAPI, diluted 1:20000) for 3 min at roomtemperature in the dark. Fluorescently labeled cells werewashed three times with PBS and once with water, and then,the slides were mounted with 4 μL of Gel Mount AqueousMounting and examined. In the experiments concerningNitDOX intracellular localization, cells were previously trans-fected with expression vectors encoding for the GFP-fused-leader sequence of E1α pyruvate dehydrogenase to labelmitochondria, the GFP-fused-KDEL sequence of calreticulin tolabel endoplasmic reticulum, the GFP-fused N-acetylgalactosa-minyltransferase 2 to label Golgi apparatus (Cell Light BacMan2.0, Invitrogen), then incubated for 6 h with 10 μM NitDOX,and prepared for the analysis by confocal microscope asreported above. For each experimental point, a minimum of fivemicroscopic fields were examined.

Cytosol-Nucleus Separation. The cytosolic and nuclearextracts were prepared as reported previously.20 After proteinquantification, samples were stored at −80 °C until use.

Mitochondria Isolation and Complex I−IV Activity.Mitochondria were extracted as described earlier.22 A 50 μLaliquot was sonicated and used for the measurement of proteincontent or Western blotting; the remaining part was stored at−80 °C until use. To confirm the presence of mitochondrialproteins in the extracts, 10 μg of each sonicated sample wassubjected to SDS-PAGE and probed with an anti-VDAC/porinantibody (data not shown).The activity of the mitochondrial complexes I−IV was

measured on 10 μg of nonsonicated mitochondrial extracts, asreported.23 Each reaction was followed for 5 min, using aLambda 3 spectrophotometer (PerkinElmer).

HPLC and LC-ESI-MS Analyses of Cell Fractions. DOXor NitDOX was incubated for 24 h in HT29 cells at 10 μmol/Lto operate at a concentration that granted the optimalsensitivity of HPLC analysis. The reverse-phase (RP) HPLCprocedure allowed the separation and quantitation ofcompounds and degradation products in cytosolic, nuclear,and mitochondrial fractions. Cellular fractions were obtained asdescribed above and diluted with equal volume of acetonitrileto deproteinize the sample. The sample was sonicated,vortexed, and then centrifuged for 10 min at 2150g, and theclear supernatant was filtered by 0.45 μm polytetrafluoro-ethylene filters and analyzed by RP-HPLC or LC-ESI-MSmethods.HPLC analyses were performed with a HP 1100 chromato-

graph system (Agilent Technologies, Palo Alto, CA) equippedwith a quaternary pump (model G1311A), a membranedegasser (G1379A), and a diode array detector (DAD)(model G1315B) integrated in the HP1100 system. Dataanalyses were processed using a HP ChemStation system(Agilent Technologies). The analytical column was a HyPurityElite C18 (250 mm × 4.6 mm, 5 μm; Hypersil, ThermoQuestCorporation, United Kingdom). The mobile phase wasacetonitrile/phosphate buffer (50 mmol/L, pH 2.0) at flowrate = 1.0 mL/min with gradient conditions: 70% PBS until 5min, from 70 to 35% PBS between 5 and 15 min, and from 35to 70% PBS between 15 and 20 min. The injection volume was100 μL (Rheodyne, Cotati, CA). The column effluent wasmonitored at 234 and 480 nm referenced against a 700 nmwavelength. Quantitation of DOX and NitDOX was done usingcalibration curves of compounds: the linearity of the calibrationcurves was determined in a concentration range of 1−50 μM

Molecular Pharmaceutics Article

dx.doi.org/10.1021/mp300311b | Mol. Pharmaceutics 2013, 10, 161−174163

Page 4: Mitochondrial-Targeting Nitrooxy-doxorubicin: A New Approach To Overcome Drug Resistance

(r2 > 0.99); quantitative analysis of their metabolites was alsoconducted using DOX (for doxorubicinol) and NitDOX (fordenitrated derivatives) as references.LC-ESI-MS analyses were performed with a Thermo

Finningan LCQ Deca XP plus system equipped with aquaternary pump, a Surveyor AS autosampler, and a Surveyorphotodiode array detector, and a vacuum degasser was used forLC/MS analyses (Thermo Electron Corporation, Waltham,MA). All of the chromatographic separations were performedon a Hypersil HyPURITY Elite C18 (5 μm, 250 mm × 4.6mm) (ThermoQuest Corporation) as a stationary phaseprotected by a C18 SecurityGuard (Phenomenex srl, CastelBolognese, Italy). Aliquots (20 μL) of supernatants obtainedfrom incubations were injected onto the system and eluted witha mobile phase (flow rate, 0.8 mL/min) consisting of A, 0.1%formic acid solution, and B, acetonitrile. The following gradientelution was used: 0−5 min (A = 65%, B = 35%), 5−17 min (A= 30%, B = 70%), 17−17.5 min (A = 65%, B = 35%), and17.5−25 min (A = 65%, B = 35%). The eluants were filtered

through a 0.45 μm pore size polyvinylidene difluoridemembrane filter before use. The eluate was injected into theelectrospray ion source (ESI), and MS spectra were acquiredand processed using Xcalibur software. The operatingconditions on the ion trap mass spectrometer were as follows:positive mode: spray voltage, 5.30 kV; source current, 80 μA;capillary temperature, 350 °C; capillary voltage, 12.00 V; tubelens offset, −30.00 V; multipole 1 offset, −6.75 V; multipole 2offset, −8.50 V; and sheath gas flow (N2), 60 Auxiliary Units.Data were acquired in full-scan and product ion scan modes(MS2) using a mass scan range of m/z 200−900. The collisionenergy was optimized at 25%.

Tricarboxylic Acid (TCA) Cycle Measurement. Cellswere washed with PBS, detached with trypsin/EDTA, andresuspended at 2 × 105 cells in 1 mL of Hepes buffer (145mmol/L NaCl, 5 mmol/L KCl, 1 mmol/L MgSO4, 10 mmol/LHepes, 10 mmol/L glucose, and 1 mmol/L CaCl2, pH 7.4)containing 2 μCi of [6-14C]glucose (55 mCi/mmol, Perki-nElmer). Cell suspensions were incubated for 1 h in a closed

Figure 1. Effects of DOX and NO-releasing DOXs in sensitive and resistant cells. (A) Expression of ABC transporters in DOX-sensitive humancolon cancer HT29 cells and DOX-resistant HT29-dx cells. Cells were lysed, and the whole cellular lysate was subjected to Western blotting for Pgp/ABCB1, MRP1/ABCC1, MRP2/ABCC2, and BCRP/ABCG2 proteins (see the Experimental Section). The expression of actin, as a product of ahousekeeping gene, was used as a control of equal protein loading. The figure is representative of three experiments with similar results. (B−D)HT29 and HT29-dx cells were cultured in fresh medium (0) or in the presence of 5 μmol/L DOX, NitDOX, or FurDOX for 1, 3, 6, and 24 h. Theintracellular accumulation of DOXs (B) was measured fluorimetrically in cell lysates, and the amount of extracellular nitrite (C) and the release ofLDH (D) were evaluated as reported in the Experimental Section. In panel C, the scale on the left y-axis refers to DOX and NitDOX, and the scaleon the right y-axis refers to FurDOX. Measurements were performed in triplicate, and data are presented as means ± SDs (n = 3). Versus untreatedcells (0), *p < 0.05; and versus DOX alone in the same data set, °p < 0.05.

Molecular Pharmaceutics Article

dx.doi.org/10.1021/mp300311b | Mol. Pharmaceutics 2013, 10, 161−174164

Page 5: Mitochondrial-Targeting Nitrooxy-doxorubicin: A New Approach To Overcome Drug Resistance

experimental system to trap the 14CO2 developed from[14C]glucose, and the reaction was stopped by injecting 0.5mL of 0.8 N HClO4. The amount of glucose transformed intoCO2 through the TCA cycle was calculated as described24 andexpressed as nmol CO2/h/mg of cell proteins.ATP Synthesis. Quantitative determination of ATP in

mitochondria was performed using the ATP BioluminescentAssay Kit (FL-AA, Sigma Aldrich Co.), following themanufacturer's instruction. The luminescence was recordedusing a Magic Lite Analyzer (Ciba Corning Diagnostic Corp.,Cambridge, MA). ATP was quantified as arbitrary light unitsand then converted into nmol ATP/mg mitochondrial proteins,according to a calibration curve previously set.ROS and RNS Measurement. For ROS measurement,

cells were rinsed with PBS and loaded with 10 μmol/L 5-(and-6)-chloromethyl-2′,7′-dichorodihydro-fluorescein diacetate-ace-toxymethyl ester (DCFDA-AM) for 10 min at 37 °C, thenwashed, and resuspended in 0.5 mL of PBS. The intracellularfluorescence was detected by flow cytometry, using aFACSCalibur system (Becton Dickinson), with a 530 nmbandpass filter. For each analysis, 100000 events were collectedand analyzed by the CellQuest software (Becton Dickinson).The amount of intracellular nitrotyrosine, considered as an

index of RNS generation,25 was measured by the NitrotyrosineELISA kit (Hycult Biotechnolohgy, The Netherlands),following the manufacturer's instructions. The absorbance wasread with a Packard EL340 microplate reader (Bio-TekInstruments) and converted into pmol/mg cell proteinsaccording to the titration curve.Lipid Peroxidation. Cells were washed with fresh medium,

detached, and resuspended in 1 mL of PBS. Lipid peroxidationwas detected by measuring the intracellular level ofmalonyldialdehyde (MDA), an end product of polyunsaturatedfatty acids and related esters breakdown, with the LipidPeroxidation Assay kit (Oxford Biomedical Research, Oxford,MI), which uses the reaction of N-methyl-2-phenylindole withMDA in the presence of hydrochloric acid to yield a stablechromophore with maximal absorbance at 586 nm. Theabsorbance was measured with a Packard EL340 microplatereader (Bio-Tek Instruments). Results were expressed as nmol/mg cell proteins, according to a titration curve previouslyprepared.Cytochrome c Release. Ten micrograms of proteins from

cytosolic and mitochondrial fraction was subjected to Westernblotting to assess the cytosolic release of cytochrome c, taken asan index of apoptosis,26 using an anticytochrome c antibody(Becton Dickinson).Caspase Activity. Cells were lysed in 0.5 mL of caspase

lysis buffer (20 mmol/L Hepes/KOH, 10 mmol/L KCl, 1.5mmol/L MgCl2, 1 mmol/L EGTA, 1 mmol/L EDTA 1, 1mmol/L DTT, 1 mmol/L PMSF, and 10 μg/mL leupeptin, pH7.5). Twenty micrograms of cell lysates was incubated for 1 h at37 °C with 20 μmol/L of the fluorogenic substrate of caspase-9Ac-Leu-Glu-His-Asp-7-amino-4-methylcoumarin (LEHD-AMC) or of caspase-3 Ac-Asp-Glu-Val-Asp-7-amino-4-methyl-coumarin (DEVD-AMC), in 0.25 mL of caspase assay buffer(25 mmol/L Hepes, 0.1% w/v 3-[(3-cholamidopropyl)-dimethylammonio]-1-propanesulfonate (CHAPS), 10% w/vsucrose, 10 mmol/L DTT, 0.01% w/v egg albumin, pH 7.5).The reaction was stopped by adding 0.75 mL of ice-cold 0.1%w/v trichloroacetic acid, and the fluorescence of AMC fragmentreleased by active caspases was read using a LS-5 spectro-fluorimeter (PerkinElmer). Excitation and emission wave-

lengths were 380 and 460 nm, respectively. Fluorescence wasconverted in pmol/μg cell proteins, using a calibration curveprepared previously with standard solutions of AMC.

Statistical Analysis. All data in text and figures areprovided as means ± SDs. The results were analyzed by a one-way analysis of variance (ANOVA) and Tukey's test. p < 0.05was considered significant.

■ RESULTSNitDOX Is More Effective than DOX in Drug-Resistant

Cancer Cells. To monitor the intracellular drug accumulationand toxicity of DOX, NitDOX, and FurDOX, we chose themodel of the drug-sensitive human colon cancer HT29 cellsand the drug-resistant counterpart HT29-dx cells, whichexpress higher levels of several ABC transporters extrudingDOX, such as Pgp/ABCB1, MRP1/ABCC1, MRP2/ABCC2,and BCRP/ABCG2, than HT29 cells (Figure 1A). At 5 μmol/L, a concentration of DOX that was cytotoxic in HT29 cells,but not in HT29-dx cells,5 all of the drugs accumulated timedependently in HT29 cells, with FurDOX > NitDOX > DOX(Figure 1B). DOX was not retained in HT29-dx cells, incontrast to NitDOX and FurDOX, whose accumulationincreased as a function of time also in drug-resistant cells.The content of FurDOX was higher than that of NitDOX ateach time point (Figure 1B). A similar trend was observed forthe extracellular nitrite amount (Figure 1C), taken as an indexof NO released by the compounds (for NitDOX and FurDOX)or synthesized by the cells themselves: DOX induced asignificant increase of nitrite only in HT29 cells after 24 h,whereas with NitDOX and FurDOX, the NO levels were higherthan with DOX, raised earlier and in both sensitive andresistant cells. FurDOX was a stronger NO releaser thanNitDOX (Figure 1C). Cell toxicity, measured as the release ofLDH in the extracellular medium, was produced by DOX onlyin HT29 cells and not in HT29-dx cells; on the opposite, theNO-releasing drugs were cytotoxic both in drug-sensitive and indrug-resistant cells (Figure 1D).To clarify whether NO was mainly responsible for the

cytotoxicity of the compounds, in a parallel experimental set,we used DOX in association with different NO donorsthat is,NitE, FurE, SNP, and spermine NONOate (NONO)eachshowing different kinetics and potency of NO release(Supplementary Figure S2A in the Supporting Information).All of the NO donors increased the intracellular retention ofDOX at 24 h (Supplementary Figure S2B in the SupportingInformation); the most potent NO donors (FurE and NONO)were also the most potent enhancers of the intracellularaccumulation of DOX (Supplementary Figure S2B in theSupporting Information). They were also cytotoxic per se inHT29 and HT29-dx cells (Supplementary Figure S2C in theSupporting Information). Of note, the presence of NO donorsrestored the cytotoxicity of DOX in drug-resistant HT29-dxcells (Supplementary Figure S2C in the Supporting Informa-tion).After 24 h, cells treated with NitE or FurE had similar levels

of nitrite when compared to cells treated with NitDOX orFurDOX (compare Supplementary Figure S2A in theSupporting Information and Figure 1C). Also, the DOXcontent was superimposable between the experimentalconditions DOX + NitE and NitDOX or DOX + FurE andFurE (compare Supplementary Figure S2B in the SupportingInformation and Figure 1B). Unexpectedly, the release of LDHwas higher with NitDOX than with DOX + NitE, as well as

Molecular Pharmaceutics Article

dx.doi.org/10.1021/mp300311b | Mol. Pharmaceutics 2013, 10, 161−174165

Page 6: Mitochondrial-Targeting Nitrooxy-doxorubicin: A New Approach To Overcome Drug Resistance

with FurDOX than with DOX + FurE (compare Supple-mentary Figure S2C in the Supporting Information and Figure1D), suggesting that NitDOX and FurDOX may haveadditional mechanisms of toxicity than DOX coincubatedwith the corresponding NO-releasing molecule.NitDOX and FurDOX were significantly more accumulated

than DOX also in other human cancer cells possessing anacquired drug-resistant phenotype, such as A549-dx and K562-dx cells (which, as expected, retained less DOX than thesensitive counterparts A549 and K562), and in primary cells ofmalignant mesothelioma (MM98), which again contained alower amount of DOX than the nontransformed mesothelialcells Met5A (Supplementary Figure S3A in the SupportingInformation).We checked in parallel the release of LDH from H9c2

cardiomyocytes: whereas DOX and NitDOX did not differ forthe intracellular accumulation and the toxicity, FurDOX wassignificantly more accumulated and elicited a stronger cytotoxiceffect (Supplementary Figure S3B in the SupportingInformation).Similarly, in nontransformed human colon epithelial CCD-

18Co cells, DOX and NitDOX did not differ in terms ofintracellular drug accumulation (Supplementary Figure S4A inthe Supporting Information), nitrite, and cytotoxicity (Supple-mentary Figure S4B in the Supporting Information), whereasFurDOX was the most retained within CCD-18Co cells, themost potent inducer of NO, and the most potent inducer ofcytotoxicity (Supplementary Figure S4A,B in the SupportingInformation).In consequence of the higher toxicity of FurDOX in

cardiomyocytes (Supplementary Figure S3B in the SupportingInformation) and nontransformed epithelial cells (Supplemen-tary Figure S4B in the Supporting Information), we withdrewFurDOX from the subsequent experiments and focused ourinvestigation on NitDOX, which was more effective than DOXin accumulating into all cell lines that we analyzed, withoutexerting a greater toxicity than DOX on cardiomyocytes andnontransformed colon cells.NitDOX Does Not Inhibit Topoisomerase II and Has a

Different Uptake and Intracellular Distribution thanDOX. Because one of the anticancer mechanisms of DOX is theinhibition of topoisomerase II, we first investigated whether theaddition of the nitrooxy group enhanced this property in an invitro assay (Figure 2). Purified topoisomerase II was inhibiteddose dependently by DOX, as well as by etoposide, the otherinhibitor profiled. NitDOX poorly inhibited topoisomerase II(Figure 2). This result led us to hypothesize that the celltoxicity of NitDOX relies on different mechanisms, such asmore favorable kinetics of uptake or cellular targets differentfrom nuclear topoisomerase II.NitDOX was more rapidly uptaken than DOX (Figure 3A).

The uptake of DOX was lower in HT29-dx cells (1.94 ± 0.19nmol/mg prot, corresponding to 0.41 ± 0.06 μmol/L) than inHT29 cells (3.94 ± 0.43 nmol/mg prot, corresponding to 0.91± 0.03 μmol/L), whereas the entry of NitDOX was the same indrug-sensitive (4.74 ± 0.58 nmol/mg prot, corresponding to1.14 ± 0.07 μmol/L) and drug-resistant cells (4.72 ± 0.91nmol/mg prot, corresponding to 1.01 ± 0.09 μmol/L). Inaddition, the uptake of DOX was reduced in the presence ofTEA (Figure 3A), an inhibitor of the SLC22A4 transporter, amember of the organic cation/zwitterion transporters familythat mediates the entry of DOX by facilitated transport.21

SLC22A4 was slightly higher in HT29 cells than in HT29-dx

cells (Figure 3B). Because the uptake of NitDOX was equal inboth cell lines and did not change in the presence of TEA(Figure 3A), it was likely a SLC22A4-independent process.NOendogenously produced or released from the nitrooxy

groupwas not responsible for the greater uptake of NitDOX,as suggested by the finding that the uptake of the compoundwas not reduced by NO scavengers, such as 2-phenyl-4,4,5,5-tetramethylimidazoline-1-oxyl 3-oxide (PTIO) or packed redblood cells. Similarly, NO scavengers did not affect the uptakeof DOX (data not shown).Another remarkable difference between DOX and NitDOX

was the intracellular distribution: whereas DOX accumulated inthe nuclei of HT29 cells, NitDOX exhibited a prominentextranuclear localization at 30 min and 1, 3, 6 (data not shown),and 24 h (Figure 3C). The quantitative fluorimetric analysis ofDOX and NitDOX in nuclear and cytosolic extracts confirmedwhat we had previously observed in whole HT29 cells (Figure1D). In HT29-dx cells, the amount of DOX was lower in bothnuclei and cytosol (Figure 3D) and was undetectable bymicroscope analysis (data not shown). NitDOX, although a bitless accumulated in HT29-dx cells, maintained the samedistribution pattern observed in HT29 cells, reaching thehighest concentration in cytosolic fractions (Figure 3D). Thecoincubation of DOX and NO donors increased the amount ofDOX in nuclear extracts, without significant differencesbetween each compound (Supplementary Figure S5A in theSupporting Information). Only a small increase of DOX wasdetectable in cytosolic extracts (Supplementary Figure S5A inthe Supporting Information).SLC224A was absent from nuclear extracts (data not shown),

and TEA was devoid of effect on the drugs distribution (datanot shown). Pgp/ABCB1, MRP1/ABCC1, and BCRP/ABCG2were present in nuclear extracts and were nitrated by DOX inHT29 cells and by NitDOX in HT29 and HT29-dx cells (datanot shown); however, PTIO, which prevented the nitration ofthe transporters, did not change the distribution of DOX orNitDOX (data not shown). These results indicate that theexclusion of NitDOX from nucleus was not due to a different

Figure 2. In vitro topoisomerase II inhibition by DOX and NitDOX.The activity of human purified topoisomerase II was measured afterincubating the enzyme with the supercoiled pHOT1 plasmid, in theabsence (C) or presence of DOX (1, 5, 25, and 50 μmol/L, DOX) orNitDOX (1, 5, 25, and 50 μmol/L, NitDOX). Etoposide (5, 50, and100 μmol/L, etop) was used as positive control of topoisomeraseinhibition. The reaction products were resolved on agarose gel. LinearpHOT1 (lane P) was used as a marker. To obtain a blank, supercoiledpHOT1 was incubated in the absence of topoisomerase II (lane C−).N, nicked pHOT1; L, linear pHOT1; and S, supercoiled pHOT1.

Molecular Pharmaceutics Article

dx.doi.org/10.1021/mp300311b | Mol. Pharmaceutics 2013, 10, 161−174166

Page 7: Mitochondrial-Targeting Nitrooxy-doxorubicin: A New Approach To Overcome Drug Resistance

uptake via SLC22A4 protein or to a different efflux fromnuclear envelope mediated by ABC transporters.DOX and NitDOX had a different lipohilicity: the octanol/

water distribution coefficient at pH 7.4 (log D7.4) was 0.47 ±0.08 for DOX and 2.72 ± 0.09 for NitDOX (n = 6), suggestingthat NitDOX should be more favored to enter cells by a simplediffusion mechanism. The different hydrophobicity may explainalso the different intracellular distribution of NitDOX andDOX. HPLC analysis of nuclear and cytosolic extracts fromHT29 cells confirmed that DOX was mainly present in thenucleus and NitDOX outside it (Table 1; SupplementaryFigure S6 in the Supporting Information).NitDOX Is Localized to Mitochondria and Impairs

Mitochondria Metabolism. To gather more informationabout the intracellular localization of NitDOX, we transfectedHT29 cells with GFP-fused expression vectors targeting specificorganelles: NitDOX was mainly found in mitochondria andendoplasmic reticulum, whereas it was absent from the Golgiapparatus (Figure 4A).Interestingly, NitDOX reached the same concentration in

mitochondria of HT29 and HT29-dx cells and was significantlyhigher than DOX (Figure 4B). Conversely, none of NO donorsincreased the intramitochondrial accumulation of DOX, withthe exception of a small but not significant increase promotedby NitE (Supplementary Figure S5B in the Supporting

Information). Mitochondria of HT29 and HT29-dx cells haveseveral ABC pumps transporting DOX, such as Pgp/ABCB1,MRP1/ABCC1, MRP2/ABCC2, and BCRP/ABCG2 (Figure4C). Interestingly, the amount of MRP1/ABCC1 was higher inHT29-dx cells (Figure 4C): this element may explain the loweraccumulation of DOX in the mitochondrial fraction of this cell

Figure 3. DOX and NitDOX uptake and intracellular distribution. (A) DOX-sensitive HT29 cells and DOX-resistant HT29-dx cells were incubatedfor 10 min with 5 μmol/L DOX or NitDOX, in the absence or presence of 100 μmol/L tetraethylammonium chloride (TEA), then washed, andanalyzed fluorimetrically for the intracellular drug content. Measurements were performed in triplicate, and data are presented as means ± SDs (n =3). HT29-dx DOX vs HT29 DOX, *p < 0.002; DOX vs DOX + TEA, °p < 0.05; and NitDOX vs the respective condition with DOX, ◊p < 0.002.(B) Expression of SLC22A4 in HT29 and HT29-dx cells. Whole cellular lysate was analyzed by Western blotting for SLC22A4 protein, using theexpression of actin as a control of equal protein loading. The figure is representative of three experiments with similar results. (C) Confocalmicroscope analysis of intracellular DOX. Nonpermeabilized HT29 cells were incubated for 24 h with 5 μmol/L DOX or NitDOX andcounterstained with DAPI. Image acquisition was performed with a Leica TCS SP2 AOBS confocal laser-scanning microscope (Leica Microsystems,Wetzlar, Germany) with a 63× oil immersion objective and 10× ocular lens.The micrographs are representative of three experiments with similarresults. (D) Nuclear and cytosolic content of DOXs. HT29 and HT29-dx cells were treated for 24 h with 5 μmol/L DOX or NitDOX; nuclear andcytosolic extracts were prepared as reported in the Experimental Section and checked for the intracellular drug content by fluorimetric assay.Measurements were performed in triplicate, and data are presented as means ± SDs (n = 3). NitDOX vs DOX in nucleus, *p < 0.001; and NitDOXvs DOX in cytosol, °p < 0.001.

Table 1. Quantitation by HPLC Analysis of DOX, NitDOX,and Metabolites in the Cellular Fractions (Cytosolic,Mitochondrial, and Nuclear Extracts) from HT29 CellsIncubated with 10 μmol/L DOX or NitDOX for 24 ha

detected compds after 24 h of incubation(nmol/mg prot.)

cellularfractions

incubatedcompds NitDOX DOX other metabolites

cytosol DOX NDb NDb

NitDOX 3.5 NDb 1.1 (NitDOX denitrate1 and 2)

mitochondria DOX 6.2 4.5 (DOX-ol)NitDOX 15.5 3.6 6.0 (NitDOX denitrate

1 and 2)nucleus DOX 8.8 5.3 (DOX-ol)

NitDOX 1.2 NDb 0.6 (NitDOX denitrate1 and 2)

aValues are the average of five experiments (n = 5, SD < 0.6). bND =not detectable (absent or <0.5 nmol/mg prot.).

Molecular Pharmaceutics Article

dx.doi.org/10.1021/mp300311b | Mol. Pharmaceutics 2013, 10, 161−174167

Page 8: Mitochondrial-Targeting Nitrooxy-doxorubicin: A New Approach To Overcome Drug Resistance

population (Figure 4B). DOX in HT29 and NitDOX in bothHT29 and HT29-dx cells elicited the tyrosine nitration ofMRP1/ABCC1 and BCRP/ABCG2, an event that wasprevented by the addition of PTIO (Figure 4C). SLC22A4was not detectable nor TEA affected the intramitochondrialaccumulation of DOX and NitDOX (data not shown).

In HT29-dx cells, the oxidative mitochondria metabolismwas more active than in HT29 cells, as demonstrated by thehigher flux through the TCA cycle (Figure 5A), the higherelectron flux through complex I (Figure 5B), and the highersynthesis of ATP (Figure 5F). In keeping with the differentintramitochondrial accumulation, DOX reduced TCA flux,complex I activity, and ATP synthesis in HT29 cells but wasdevoid of effect in HT29-dx cells (Figure 5A,B,F), where it waspoorly retained into mitochondria (Figure 4B). On thecontrary, NitDOX, which accumulated in mitochondria ofboth drug-sensitive and drug-resistant cells (Figure 4B), equallydecreased in these cells TCA cycle, complex I activity, and ATPsynthesis (Figure 5A,B,F). No changes occurred in the activityof other steps of mitochondrial electron chain, as determinedby the electron flux through the segment complex I to III(Figure 5C), through the segment complex II to III (Figure5D), and through complex IV (Figure 5E). These data suggestthat the effects elicited by the anthracyclines on mitochondrialmetabolism have rather specific targets. For both DOX inHT29 cells and NitDOX in HT29 and HT29-dx cells, thechanges on TCA activity, complex I, and ATP synthesis wereabrogated by the addition of PTIO (Figure 5A,B,F).Furthermore, the rate of DOX and NitDOX metabolism

within mitochondria was different: in mitochondrial extracts ofHT29 cells treated with DOX, we detected both DOX and amore hydrophilic metabolite, suggestive of doxorubicinol27

(DOX-ol, Table 1; Supplementary Figures S6 and S7 in theSupporting Information). On the contrary, in mitochondria ofNitDOX-treated cells, we found mainly NitDOX, together withDOX obtained from the hydrolysis of the ester group (about14%), and a considerable percentage (about 25%) of the twodenitrated metabolites deriving from NO release (NitDOXdenitrate 1 and 2, Supplementary Figures S6 and S7 in theSupporting Information). Structures of the metabolitesgenerated from DOX and NitDOX are summarized inSupplementary Figure S8 in the Supporting Information.

NitDOX Exerts a Strong Oxidative/Nitrosative Stressand Activates the Intrinsic Pathway of Apoptosis inResistant Cells. DOX increased the amount of ROS (Figure6A) and the amount of nitrotyrosine, an indicator of RNSgeneration25 (Figure 6B) in HT29 cells, but not in HT29-dxcells. These events, however, were not sufficient to producesignificant oxidative damage, because no marked change in lipidperoxidation was detected (Figure 6C). On the contrary,NitDOX effectively raised the amount of ROS and RNS inHT29 well as well as in HT29-dx cells (Figure 6A,B).Accordingly, only NitDOX produced a strong oxidative damagein both cell lines, as suggested by the significant increase ofMDA level (Figure 6C). NO was crucial in the genesis of thistype of damage, since the effect of NitDOX was reduced by theaddition of PTIO (Figure 6C).Finally, to determine whether the impairment of mitochon-

dria metabolism and the increased oxidative damages elicited byNitDOX effectively resulted in cell death, we analyzed therelease of cytochrome c into the cytosol and the subsequentactivation of the caspases involved in the intrinsic apoptoticpathway. Whereas DOX produced a small cytosolic release ofcytochrome c (Figure 7A), followed by an increased activity ofcaspase-9 (Figure 7B) and caspase-3 (Figure 7C) only in HT29cells, NitDOX was a stronger inducer of cytochrome c release(Figure 7A) and a stronger activator of caspase-9 (Figure 7B)and caspase-3 (Figure 7C) in both HT29 and HT29-dx cells.

Figure 4. Mitochondrial accumulation of DOX and NitDOX. (A)Subcellular localization of NitDOX. HT29 cells were incubated for 18h with the expression vectors encoding for the GFP-fused leadersequence of E1 α pyruvate dehydrogenase (to label mitochondria), theGFP-fused KDEL sequence of calreticulin (to label endoplasmicreticulum), and the GFP-fused N-acetylgalactosaminyltransferase 2 (tolabel Golgi apparatus), then incubated for 6 h with 10 μM NitDOX,and analyzed by Olympus FV300 laser scanning confocal microscope(Olympus Biosystems, Hamburg, Germany). Cells were imaged usinga 60× oil immersion objective and 10× ocular lens. Nomarski imageswere obtained by differential interference contrast optical componentsinstalled on an IX71 inverted microscope. The micrographs arerepresentative of three experiments with similar results. (B) HT29 andHT29-dx cells were incubated with 5 μmol/L DOX or NitDOX, in theabsence or presence of the NO scavenger PTIO (100 μmol/L), andthen fractioned. The mitochondria extracts were analyzed fluorimetri-cally to measure the DOXs amount. Measurements were performed intriplicate, and data are presented as means ± SDs (n = 3). NitDOX vsDOX, *p < 0.001; and NitDOX + PTIO vs NitDOX alone, °p < 0.002.(C) Western blot detection of nitrated mitochondrial ABC trans-porters. Mitochondrial extracts from cells untreated (ctrl) or treated asreported in panel B were lysed and immunoprecipitated with anantinitrotyrosine polyclonal antibody. The immunoprecipitatedproteins were subjected to Western blotting, using anti-Pgp/ABCB1,anti-MRP1/ABCC1, anti-MRP2/ABCC2, or BCRP/ABCG2 antibod-ies (see the Experimental Section). An aliquot of each sample wastested with the same antibodies before immunoprecipitation, tomeasure the amount of total ABC transporters. Porin was used as acontrol of equal protein loading. The figure is representative of threesimilar experiments.

Molecular Pharmaceutics Article

dx.doi.org/10.1021/mp300311b | Mol. Pharmaceutics 2013, 10, 161−174168

Page 9: Mitochondrial-Targeting Nitrooxy-doxorubicin: A New Approach To Overcome Drug Resistance

Again, the removal of NO by PTIO prevented all of theproapoptotic effects of DOX and NitDOX (Figure 7A−C).

■ DISCUSSIONIn the light of previous studies showing that NO donors arechemosensitizers,5,16,17 several NO-based strategies, such as theuse of the ABC transporters inhibitors conjugated with NO28

or the use of compounds with the dual property of inhibitingPgp and increasing the endogenous synthesis of NO,29 havebeen investigated. We have recently synthesized new multi-functional DOXs by joining themthrough an appropriatelinkerto a NO donor with known biological properties.These compounds, namely, NitDOX and FurDOX, overcameMDR in human colon cancer HT29-dx cells.18 Given thepleiotropism of anticancer effects of NO,14 we hypothesized

that NO-releasing DOXs may have additional effects thanparental DOX.Colon cancers are an interesting model to test this

hypothesis, because they are particularly sensitive to thecytotoxic effects of NO. For instance, NO-conjugated non-steroidal anti-inflammatory drugs reduce tumor growth ofcolon cancer and induce apoptosis with a potency significantlyhigher than the parental nonsteroidal anti-inflammatorydrugs.30,31 In our model of DOX-sensitive HT29 and DOX-resistant HT29-dx colon cancer cells incubated with DOX,NitDOX, or FurDOX, the degree of toxicity was proportionalto the amount of intracellular DOX and to the amount of NOreleased. Similarly, when we used DOX in association withdifferent NO donors, we increased in parallel the levels ofnitrite, the intracellular accumulation of DOX, and the

Figure 5. Effects of DOX and NitDOX on mitochondrial metabolism. HT29 and HT29-dx cells were grown for 24 h in fresh medium (ctrl) or with5 μmol/L DOX or NitDOX, in the absence or presence of the NO scavenger PTIO (100 μmol/L), and then subjceted to the followinginvestigations. (A) Tricarboxylic acid cycle was measured in living cells, labeled with [14-6C]-glucose, as reported in the Experimental Section.Measurements were performed in triplicate, and data are presented as means ± SDs (n = 3). Versus ctrl HT29, *p < 0.005; versus ctrl HT29-dx, °p <0.001; and DOX + PTIO or NitDOX + PTIO vs DOX or NitDOX alone, ◊p < 0.01. (C−E) Isolated mitochondria were analyzed for the activity ofcomplex I (B), complex I + II (C), complex II + III (D), and complex IV (E). Measurements were performed in triplicate, and data are presented asmeans ± SDs (n = 3). Versus ctrl HT29, *p < 0.01; versus ctrl HT29-dx, °p < 0.005; and DOX + PTIO or NitDOX + PTIO vs DOX or NitDOXalone, ◊p < 0.05. (F) ATP levels were detected by a chemiluminescence-based assay as described in the Experimental Section. Measurements wereperformed in triplicate, and data are presented as means ± SDs (n = 3). Versus ctrl HT29, *p < 0.02; versus ctrl HT29-dx, °p < 0.001; and DOX +PTIO or NitDOX + PTIO vs DOX or NitDOX alone, ◊p < 0.02.

Molecular Pharmaceutics Article

dx.doi.org/10.1021/mp300311b | Mol. Pharmaceutics 2013, 10, 161−174169

Page 10: Mitochondrial-Targeting Nitrooxy-doxorubicin: A New Approach To Overcome Drug Resistance

cytotoxicity, restoring the DOX-induced cytotoxicity also indrug-resistant HT29-dx cells. Both the increased retention ofDOX and the increased amount of NO, produced by iNOS inresponse to the intracellular DOX or released by thecompounds, may explain this restoration of cytotoxicity inDOX-resistant cells. NitDOX and FurDOX, being less extrudedthan DOX by ABC transporters,18 achieved a good

accumulation in both HT29 and HT29-dx cells. Their abilityto release NO had the dual effect to further reduce the drugextrusion through the ABC transporters and to enhance thedrug toxicity in resistant cells.Interestingly, the cytotoxicity elicited by NitDOX or

FurDOX was higher than the cytotoxicity elicited by thesame amount of DOX coincubated with the correspondingNO-releasing molecule (i.e., NitE and FurE, respectively). Thisresult suggested that the cytotoxicity of NitDOX and FurDOXwas not simply the sum of increased NO release and increasedretention of DOX but resulted from different pharmacody-namic effects.FurDOX, the most effective NO-releasing DOX against drug-

resistant cells was also highly cytotoxic for cardiomyocytes invitro, while conversely, NitDOX was not more toxic than DOX.The protective or toxic effects of NO on cardiomyocytes aredifficult to predict, because NO may act as either ROSscavenger or generator (favoring the synthesis of peroxynitrite),depending on the relative stoichiometry between NO andROS.8 This complexity may explain the discrepant reports onthe effects of NO in cardiomyocytes32−34 and the differentbehavior of NitDOX and FurDOX in H9c2 cells. Although wecannot infer any conclusion about the in vivo cardiotoxicityfrom these in vitro assays, it is noteworthy that NitDOX was amore potent cytotoxic agent than DOX and was capable toovercome drug resistance.18 In the light of these premises, itmight be administered at lower doses than DOX. Becausecardiotoxicity of DOX is dose-dependent, this feature ofNitDOX might decrease the risk of cumulative cardiac damagesof DOX in a putative administration in vivo.We cannot state that NitDOX was selective against cancer

cells, since it induced the same cytotoxicity of DOX innontransformed colon epithelial cells; however, being morecytotoxic than DOX against colon cancer cells,18 NitDOXmight be used in vivo at lower concentrations than DOX,exerting good efficacy against cancer cells and limited toxicityagainst colon epithelium.An intriguing difference between DOX and NitDOX is that

the latter was a less potent inhibitor of topoisomerase II, one ofthe known targets of anthracyclines. We postulate that thepresence of nitrooxy group makes NitDOX less able to bindtopoisomerase than DOX. On the other hand, this result led usto hypothesize that the cell toxicity of NitDOX wasindependent from topoisomerase II inhibition and could bedue to other mechanisms, such as a more favorable kinetics ofuptake and/or different cellular targets.The rate of DOX uptake was lower in drug-resistant cells

than in drug-sensitive ones and was dependent on both passivediffusion and facilitated diffusion via SLC22A4, as shown byemploying the SLC22A4 inhibitor TEA. High levels ofSLC22A4 confer sensitivity to DOX by increasing the amountof the drug uptake by cells,21 and indeed, the transporter wasslightly lower in HT29-dx cells than in HT29 cells. NitDOXuptake was equal in drug-sensitive and drug-resistant cells,TEA-insensitive, appearing independent of SLC22A4, andmore rapid than the uptake of DOX. NitDOX was morehydrophobic than DOX; this feature may facilitate its passivediffusion across the plasma membrane and explain its rapiduptake.At the same time, the higher hydrophobicity limited the

nuclear entry of NitDOX and favored the delivery throughhighly impermeant membranes, like the internal mitochondrialmembrane, highlighting a second difference between DOX and

Figure 6. Effects of DOX and NitDOX on the synthesis of ROS andRNS. HT29 and HT29-dx cells were grown for 24 h in fresh medium(ctrl) or with 5 μmol/L DOX or NitDOX. When indicated, the NOscavenger PTIO (100 μmol/L) was added. (A) ROS levels. Cells wereincubated for 20 min at 37 °C with the ROS-sensitive probe DCFDA-AM and then analyzed by flow cytometry in duplicate (n = 3). Thehistograms depict one representative experiment (gray curve, un-treated cells; dotted line, DOX-treated cells; and continuous line,NitDOX-treated cells). (B) RNS levels. The intracellular amount ofnitrotyrosine, taken as an index of RNS, was detected in cell lysates byan ELISA kit (see the Experimental Section). Measurements wereperformed in triplicate, and data are presented as means ± SDs (n =3). Versus ctrl HT29, *p < 0.001; and versus ctrl HT29-dx, °p < 0.02.(C) Lipoperoxidation measurement. The amount of MDA, a markerof lipid peroxidation, was measured in the cell lysates spectropho-metrically, as described in the Experimental Section. Measurementswere performed in triplicate, and data are presented as means ± SDs(n = 3). Versus ctrl HT29, *p < 0.001; and NitDOX + PTIO vsNitDOX alone, °p < 0.05.

Molecular Pharmaceutics Article

dx.doi.org/10.1021/mp300311b | Mol. Pharmaceutics 2013, 10, 161−174170

Page 11: Mitochondrial-Targeting Nitrooxy-doxorubicin: A New Approach To Overcome Drug Resistance

NitDOX. In keeping with our observation on the mitochondriallocalization of NitDOX, it has been reported that syntheticacyl-conjugated DOXs, more hydrophobic than the parentaldrug, are more uptaken by drug-resistant cells and display acytosolic rather than a nuclear localization.35

Also, the level of ABC transporters in subcellular compart-ments, which is highly variable and cell type-dependent,36−38

strongly affects the intracellular distribution of anthracyclines.In our model, the rate of nitration and extrusion activity ofPgp/ABCB1, MRP1/ABCC1, and BCRP/ABCG2 did notappear to underlie the nuclear-cytosolic partition of DOX andNitDOX. Interestingly, all of these ABC transporters weredetected in mitochondrial extracts of HT29 and HT29-dx cells,which had a greater amount of MRP1/ABCC1. Consideringthe nitration pattern of MRP1/ABCC1 and BCRP/ABCG2and the reversing effects of PTIO, which lowered the amount ofmitochondrial NitDOX, we postulate that the mitochondrialretention of NitDOX also relies on the reduced efflux throughABC transporters, subsequent to their nitration.The enhanced intramitochondrial accumulation of DOX was

not peculiar of all NO-releasing molecules: the NO donorscoincubated with DOX increased the content of DOX both inwhole cell and in the nucleus, likely as a consequence of thenitration of ABC transporters in HT29 and HT29-dx cells,5,6

but did not change the DOX retention within mitochondria.Only NitE produced a slight but not significant increase ofintramitochondrial DOX, which remained lower than the oneobtained with NitDOX. This result suggests that the nitrooxygroup conjugated with DOX is more potent as a nitrating agentof the mitochondrial ABC transporters than its ester derivative.In NitDOX, the nitrooxy group is part of a highly hydrophobicmolecule; therefore, it is particularly favored to cross themitochondrial membranes delivered by such a carrier. Onceentered, the mitochondrial aldehyde dehydrogenase responsibleof the metabolism of nitrates can produce NO from nitrooxygroup,39 increasing the intramitochondrial release of NO. Asevidenced by the analysis of NitDOX metabolites inmitochondrial extracts, about 25% of NitDOX was denitrated,suggesting that NO was released from the compound withinmitochondria. Of note, whereas DOX was highly convertedinto a more reduced metabolite suggestive of doxorubicinol27 inboth nucleus and mitochondria, the amount of NitDOXmetabolized was proportionally lower. This difference, whichcan be due to a lower affinity of NitDOX for the catabolizingenzymes, may contribute to maintain relatively high the amountof mitochondrial NitDOX.Overall, we suggest that the different intracellular distribution

of NitDOX versus DOX is the sum of at least two factors: the

Figure 7. Induction of apoptosis by NitDOX in sensitive and resistant cells. HT29 and HT29-dx cells were grown for 24 h in fresh medium (ctrl) orwith 5 μmol/L DOX or NitDOX, in the absence or presence of the NO scavenger PTIO (100 μmol/L, PT) and then subjected to the followinginvestigations. (A) Citochrome c release. Cytosol and mitochondrial fractions were separated and subjected to Western blotting analysis forcytochrome c. Porin and actin were used as equal loading controls for each fraction. The figure is representative of three experiments with similarresults. (B and C) The activity of caspase-9 (B) and caspase-3 (C) was measured fluorimetrically in cell lysates, as reported in the ExperimentalSection. Measurements were performed in triplicate, and data are presented as means ± SDs (n = 3). Versus ctrl, *p < 0.05; and DOX + PTIO orNitDOX + PTIO vs DOX or NitDOX alone, °p < 0.05.

Molecular Pharmaceutics Article

dx.doi.org/10.1021/mp300311b | Mol. Pharmaceutics 2013, 10, 161−174171

Page 12: Mitochondrial-Targeting Nitrooxy-doxorubicin: A New Approach To Overcome Drug Resistance

passive diffusion of the compound into subcellular compart-ments, which depends on its hydrophobicity, and the lower rateof extrusion, which relies on the activity of ABC transporters ineach cell compartment. The hydrophobicity of NitDOX andthe release of NO followed by the nitration of ABCtransporters in mitochondria both contribute to the highmitochondrial accumulation of NitDOX.The mitochondrial delivery of NitDOX, which occurred in

both sensitive and resistant cells, points out a novel andunexpected mechanism of cytotoxicity of this compound, inline with other works that propose a possible mitotoxic effectfor some anthracyclines. For instance, daunorubicin increasesthe mitochondrial respiration at low concentrations and inhibitsit at higher concentrations, thus acting as a mitotoxic drug.40

Recently, a liposomal daunorubicin targeting mitochondria hasbeen shown to exert high cytotoxicity in MCF7-Adr cells inassociation with the calcium channel blocker amlodipine, bydisrupting the mitochondrial potential, releasing the cyto-chrome c, and activating the caspases cascade.41 This findingconfirms that the mitochondrial metabolism is critical for thesurvival of drug-resistant cells and is in keeping with what weobserved in HT29-dx, where the TCA cycle, the electrontransport across complex I, and the synthesis of ATP werehigher than in HT29 cells. The preferential delivery of NitDOXwithin mitochondria may explain its greater efficacy in inducingcytotoxicity.It is well established that NO affects mitochondrial energy

metabolism.14 For instance, it elicits the tyrosine nitration ofTCA cycle enzymes like aconitase and isocitrate dehydrogen-ase, as well as enzymes important for the synthesis ofubiquinone and for the integrity of Fe−S clusters,42 andinhibits complexes I and IV.43 These mechanisms may accountfor the observed effects of NitDOX to decrease the TCA cycle,the activity of complex I, and the synthesis of ATP.Proper mitochondrial respiration protects cells against

oxygen toxicity.44 Conversely, high levels of NO irreversiblydamage mitochondrial respiratory chain components, resultingin oxidative and nitrosative stress, lipid peroxidation, loss ofcytochrome c from permeabilized mitochondria, and activationof caspases.45 All of these events were elicited by NitDOX andrelied on the release of NO from the compound, because theywere prevented by PTIO. Noteworthy, a significant activationof cytochrome c/caspase-9/caspase-3 axis was achieved equallyin drug-sensitive and drug-resistant cells.In conclusion, we propose NitDOX as a new multitarget lead

compound, whichoriginally designed as a MDR-reversingagenthas revealed a novel and unexpected mechanism ofaction. The simple addition of a NO-releasing group in theform of nitrooxy made NitDOX a functionally distinctanthracycline, with a more favorable toxicity profile and abetter efficacy against drug-resistant cells. In the context ofearlier attempts to use NO delivery strategies in cancertherapy,46,47 we believe that NitDOX is worthy of furtherinvestigations in preclinical and clinical settings.

■ ASSOCIATED CONTENT*S Supporting InformationStructures of DOX, NitDOX, FurDOX, NitE, and FurE(Supplementary Figure S1); the effects of the association ofDOX and NO donors (Supplementary Figure S2); thecytotoxic effects of DOX, NitDOX, and FurDOX in differentdrug-sensitive/drug-resistant cell lines and in cardiomyocytecultures (Supplementary Figure S3); the effects of DOX,

NitDOX, and FurDOX in nontransformed colon epithelial cells(Supplementary Figure S4); the effects of NO donors in theintracellular localization of DOX (Supplementary Figure S5);the representative chromatograms of the RP-HPLC analysis ofDOX, NitDOX, and their metabolites in cellular fractions(Supplementary Figure S6); the LC-ESI-MS analysis of DOX,NitDOX, and their metabolites in cellular fractions (Supple-mentary Figure S7); and the structures of the metabolitesgenerated from DOX and NitDOX (Supplementary Figure S8).This material is available free of charge via the Internet athttp://pubs.acs.org.

■ AUTHOR INFORMATIONCorresponding Author*Address: Department of Oncology, University of Torino, ViaSantena 5/bis, 10126 Torino, Italy. Tel: +39-011-6705857. Fax:+39-011-6705845. E-mail: [email protected] authors declare no competing financial interest.

■ ACKNOWLEDGMENTSWe are grateful to Costanzo Costamagna (Department ofOncology, University of Torino), Guido Serini (Institute forCancer Research and Treatment, Candiolo, Italy), and ErikaOrtolan (Department of Genetics, Biology and Biochemistry,University of Torino) for the technical assistance. We aregrateful to Dr. Silvio Aprile and Prof. Giorgio Grosa(Dipartimento di Scienze del Farmaco, Universita degli Studidel Piemonte Orientale A. Avogadro, Novara, Italy) for LC-MSanalysis, which allowed the characterization of metabolitesderiving from NitDOX. This work has been supported withgrants from the Italian Ministry of University and Research,Fondazione Internazionale Ricerche Medicina Sperimentale(FIRMS), Compagnia di San Paolo (“Progetto Oncologia”),and Italian Association for Cancer Research (AIRC; MFAG11475). We also thank Prof. Alberto Gasco for fruitfuldiscussions.

■ REFERENCES(1) Apetoh, L.; Mignot, G.; Panaretakis, T.; Kroemer, G.; Zitvogel, L.Immunogenicity of anthracyclines: Moving towards more personalizedmedicine. Trends Mol. Med. 2008, 14, 141−151.(2) Gottesman, M. M.; Fojo, T.; Bates, S. E. Multidrug resistance incancer: Role of ATP-dependent transporters. Nat. Rev. Cancer 2002, 2,48−58.(3) Takara, K.; Sakaeda, T.; Okumura, K. An update on overcomingMDR1-mediated multidrug resistance in cancer chemotherapy. Curr.Pharm. Des. 2006, 12, 273−286.(4) Haouala, A.; Rumpold, H.; Untergasser, G.; Buclin, T.; Ris, H. B.;Widmer., N.; Decosterd, L,A. siRNA-mediated knock-down of P-glycoprotein expression reveals distinct cellular disposition ofanticancer tyrosine kinases inhibitors. Drug Metab. Lett. 2010, 4,114−119.(5) Riganti, C.; Miraglia, E.; Viarisio, D.; Costamagna, C.;Pescarmona, G.; Ghigo, D.; Bosia, A. Nitric oxide reverts theresistance to doxorubicin in human colon cancer cells by inhibitingthe drug efflux. Cancer Res. 2005, 65, 516−525.(6) De Boo, S.; Kopecka, J.; Brusa, D.; Gazzano, E.; Matera, L.;Ghigo, D.; Bosia, A.; Riganti, C. iNOS activity is necessary for thecytotoxic and immunogenic effects of doxorubicin in human coloncancer cells. Mol. Cancer 2009, 8, e108.(7) Fruttero, R.; Crosetti, M.; Chegaev, K.; Guglielmo, S.; Gasco, A.;Berardi, F.; Niso, M.; Perrone, R.; Panaro, M. A.; Colabufo, N. A.Phenylsulfonylfuroxans as modulators of multidrug-resistance-associ-ated protein-1 and P-glycoprotein. J. Med. Chem. 2010, 53, 5467−575.

Molecular Pharmaceutics Article

dx.doi.org/10.1021/mp300311b | Mol. Pharmaceutics 2013, 10, 161−174172

Page 13: Mitochondrial-Targeting Nitrooxy-doxorubicin: A New Approach To Overcome Drug Resistance

(8) Wink, D. A.; Mitchell, J. R. Chemical biology of nitric oxide:Insights into regulatory, cytotoxic and cytoprotective mechanism ofnitric oxide. Free Radical Biol. Med. 1998, 25, 434−456.(9) Nathan, C.; Xie, Q. W. Regulation of biosynthesis of nitric oxide.J. Biol. Chem. 1994, 269, 13725−13728.(10) Fitzpatrick, B.; Mehibel, M.; Cowen, R. L.; Stratford, I. J. iNOSas a therapeutic target for treatment of human tumors. Nitric Oxide2008, 19, 217−224.(11) Singh, S.; Gupta, A. K. Nitric oxide: Role in tumour biology andiNOS/NO-based anticancer therapies. Cancer Chemother. Pharmacol.2011, 67, 1211−1224.(12) Coulter, J. A.; McCarthy, H. O.; Xiang, J.; Roedl, W.; Wagner,E.; Robson, T.; Hirst, D. G. Nitric oxideA novel therapeutic forcancer. Nitric Oxide 2008, 19, 192−198.(13) Engels, K.; Knauer, S. K.; Loibl, S.; Fetz, V.; Harter, P.;Schweitzer, A.; Fisseler-Eckhoff, A.; Kommoss, F.; Hanker, L.;Nekljudova, V.; Hermanns, I.; Kleinert, H.; Mann, W.; du Bois, A.;Stauber, R. H. NO signaling confers cytoprotectivity through thesurvivin network in ovarian carcinomas. Cancer Res. 2008, 68, 5159−5166.(14) Huerta, S.; Chilka, S.; Bonavida, B. Nitric oxide donors: Novelcancer therapeutics. Int. J. Oncol. 2008, 33, 909−927.(15) Bonavida, B.; Khineche, S.; Huerta-Yepez, S.; Garban, H.Therapeutic potential of nitric oxide in cancer. Drug Resist. Updates2006, 9, 157−173.(16) Muir, C. P.; Adams, M. A.; Graham, C. H. Nitric oxideattenuates resistance to doxorubicin in three-dimensional aggregates ofhuman breast carcinoma cells. Breast Cancer Res. Treat. 2006, 96, 169−176.(17) Frederiksen, L. J.; Sullivan, R.; Maxwell, L. R.; Macdonald-Goodfellow, S. K.; Adams, M. A.; Bennett, B. M.; Siemens, D. R.;Graham, C. H. Chemosensitization of cancer in vitro and in vivo bynitric oxide signaling. Clin. Cancer Res. 2007, 13, 2199−2206.(18) Chegaev, K.; Riganti, C.; Lazzarato, L.; Rolando, B.; Guglielmo,S.; Campia, I.; Fruttero, R.; Bosia, A.; Gasco, A. Nitric oxide donor−doxorubicin conjugates accumulate into doxorubicin resistant humancolon cancer cells inducing cytotoxicity. ACS Med. Chem. Lett. 2011, 2,494−497.(19) Morphy, R.; Rankovic, Z. Designed multiple ligands. Anemerging drug discovery paradigm. J. Med. Chem. 2005, 48, 6523−6543.(20) Riganti, C.; Orecchia, S.; Pescarmona, G.; Betta, P. G.; Ghigo,D.; Bosia, A. Statins revert doxorubicin resistance via nitric oxide inmalignant mesotelioma. Int. J. Cancer 2006, 119, 17−27.(21) Okabe, M.; Szakacs, G.; Reimers, M. A.; Suzuki, T.; Hall, M. D.;Abe, T.; Weinstein, J. N.; Gottesman, M. M. Profiling SLCO andSLC22 genes in the NCI-60 cancer cell lines to identify drug uptaketransporters. Mol. Cancer Ther. 2008, 7, 3081−3091.(22) Campia, I.; Lussiana, C.; Pescarmona, G.; Ghigo, D.; Bosia, A.;Riganti, C. Geranylgeraniol prevents the cytotoxic effects of mevastatinin THP-1 cells, without decreasing the beneficial effects on cholesterolsynthesis. Br. J. Pharmacol. 2009, 158, 1777−1786.(23) Wibom, R.; Hagenfeldt, L.; von Dobeln, U. Measurement ofATP production and respiratory chain enzyme activities inmitochondria isolated from small muscle biopsy samples. Anal.Biochem. 2002, 311, 139−151.(24) Riganti, C.; Gazzano, E.; Polimeni, M.; Costamagna, C.; Bosia,A.; Ghigo, D. Diphenyleneiodonium inhibits the cell reDOXmetabolism and induces oxidative stress. J. Biol. Chem. 2004, 279,47726−47731.(25) Ischiropoulos, H. Biological selectivity and functional aspects ofprotein tyrosine nitration. Biochem. Biophys. Res. Commun. 2003, 305,776−783.(26) Russo, P.; Catassi, A.; Malacarne, D.; Margaritora, S.; Cesario,A.; Festi, L.; Mule, A.; Ferri, L.; Granone, P. Tumor necrosis factorenhances sn38-mediated apoptosis in mesothelioma cells. Cancer2005, 103, 1503−1518.(27) Sakai Kato, K.; Saito, E.; Ishikura, K.; Kawanishi, T. Analysis ofintracellular doxorubicin and its metabolites by ultrahighperformance

liquid chromatography. J. Chromatogr., B: Anal. Technol. Biomed. LifeSci. 2010, 878, 1466−1470.(28) Rothweiler, F.; Michaelis, M.; Brauer, P.; Otte, J.; Weber, K.;Fehse, B.; Doerr, H. W.; Wiese, M.; Kreuter, J.; Al-Abed, Y.; Nicoletti,F.; Cinatl, J. Anticancer effects of the nitric oxide-modified saquinavirderivative saquinavir-NO against multidrug-resistant cancer cells.Carcinogenesis 2012, 12, 1023−1030.(29) Colabufo, N. A.; Contino, M.; Berardi, F.; Perrone, R.; Panaro,M. A.; Cianciulli, A.; Mitolo, V.; Azzariti, A.; Quatrale, A.; Paradiso, A.A new generation of MDR modulating agents with dual activity: P-gpinhibitor and iNOS inducer agents. Toxicol. in Vitro 2011, 25, 222−230.(30) Rao, C,V. Nitric oxide signaling in colon cancer chemo-prevention. Mutat. Res. 2004, 555, 107−119.(31) Hagos, G. K.; Carroll, R. E.; Kouznetsova, T.; Li, Q.; Toader, V.;Fernandez, P. A.; Swanson, S. M.; Thatcher, G. R. J. Colon cancerchemoprevention by a novel NO chimera that shows anti-inflammatory and antiproliferative activity in vitro and in vivo. Mol.Cancer Ther. 2007, 6, 2230−2239.(32) Aldieri, E.; Bergandi, L.; Riganti, C.; Costamagna, C.; Bosia, A.;Ghigo, D. Doxorubicin induces an increase of nitric oxide synthesis inrat cardiac cells that is inhibited by iron supplementation. Toxicol.Appl. Pharmacol. 2002, 185, 85−90.(33) Evig, C. B.; Kelley, E. E.; Weydert, C. J.; Chu, Y.; Buettner, G.R.; Burns, C. P. Endogenous production and exogenous exposure tonitric oxide augment doxorubicin cytotoxicity for breast cancer cellsbut not cardiac myoblasts. Nitric Oxide 2004, 10, 119−129.(34) Zhu, S. G.; Kukreja, R. C.; Das, A.; Chen, Q.; Lesnefsky, E. J.;Xi, L. Dietary nitrate supplementation protects against doxorubicin-induced cardiomyopathy by improving mitochondrial function. J. Am.Coll. Cardiol. 2011, 57, 2181−2189.(35) Effenberger-Neidnicht, K.; Breyer, S.; Mahal, K.; Sasse, F.;Schobert, R. Modification of uptake and subcellular distribution ofdoxorubicin by N-acylhydrazone residues as visualised by intrinsicfluorescence. Cancer Chemother. Pharmacol. 2012, 69, 85−90.(36) Solazzo, M.; Fantappie, O.; Lasagna, N.; Sassoli, C.; Nosi, D.;Mazzanti, R. P-gp localization in mitochondria and its functionalcharacterization in multiple drug-resistant cell lines. Exp. Cell Res.2006, 12, 4070−4078.(37) Paterson, J. K.; Gottesman, M. M. P-glycoprotein is not presentin mitochondrial membranes. Exp. Cell Res. 2007, 313, 3100−3105.(38) Solazzo, M.; Fantappie, O.; D'Amico, M.; Sassoli, C.; Tani, A.;Cipriani, G.; Bogani, C.; Formigli, L.; Mazzanti, R. Mitochondrialexpression and functional activity of breast cancer resistance protein indifferent multiple drug-resistant cell lines. Cancer Res. 2009, 69, 7235−7242.(39) Mayer, B.; Beretta, M. The enigma of nitroglycerin bioactivationand nitrate tolerance: news, views and troubles. Br. J. Pharmacol. 2008,155, 170−184.(40) Paul, M. K.; Patkari, M.; Mukhopadhayay, A. P. Existence of adistinct concentration window governing daunorubicin-inducedmammalian liver mitotoxicity-implication for determining therapeuticwindow. Biochem. Pharmacol. 2007, 74, 821−830.(41) Zhang, Y.; Li, R. J.; Ying, X.; Tian, W.; Yao, H. J.; Men, Y.; Yu,Y.; Zhang, L.; Ju, R. J.; Wang, X. X.; Zhou, J.; Chen, J. X.; Li, N.; Lu,W. L. Targeting therapy with mitosomal daunorubicin plus amlodipinehas the potential to circumvent intrinsic resistant breast cancer. Mol.Pharm. 2011, 8, 162−175.(42) Bhattacharjee, A.; Majumdar, U.; Maity, D.; Subhra Sarkar, T.;Mohan Goswami, A.; Sahoo, R.; Ghosh, S. In vivo protein tyrosinenitration in S. cerevisiae: Identification of tyrosine-nitrated proteins inmitochondria. Biochem. Biophys. Res. Commun. 2009, 388, 612−617.(43) Sarti, P.; Arese, M.; Forte, E.; Giuffre, A.; Mastronicola, D.Mitochondria and nitric oxide: Chemistry and pathophysiology. Adv.Exp. Med. Biol. 2012, 942, 75−92.(44) Sung, H. J.; Ma, W.; Wang, P.; Hynes, J.; O'Riordan, T. C.;Combs, C. A.; McCoy, J. P.; Bunz, F.; Kang, J. G.; Hwang, P. M.Mitochondrial respiration protects against oxygen-associated DNAdamage. Nat. Commun. 2010, 1, 5.

Molecular Pharmaceutics Article

dx.doi.org/10.1021/mp300311b | Mol. Pharmaceutics 2013, 10, 161−174173

Page 14: Mitochondrial-Targeting Nitrooxy-doxorubicin: A New Approach To Overcome Drug Resistance

(45) Kim, P. K. M.; Kwon, Y. G.; Chung, H. T.; Kim, Y. M.Regulation of caspases by nitric oxide. Ann. N.Y. Acad. Sci. 2002, 962,42−52.(46) Sonveaux, P.; Jordan, B. F.; Gallez, B.; Feron, O. Nitric oxidedelivery to cancer: Why and how? Eur. J. Cancer 2009, 45, 1352−1369.(47) Siemens, D. R.; Heaton, J. P.; Adams, M. A.; Kawakami, J.;Graham, C. H. Phase II study of nitric oxide donor for men withincreasing prostate-specific antigen level after surgery or radiotherapyfor prostate cancer. Urology 2009, 74, 878−883.

Molecular Pharmaceutics Article

dx.doi.org/10.1021/mp300311b | Mol. Pharmaceutics 2013, 10, 161−174174


Recommended