11
Formation of 17-Allylamino-Demethoxygeldanamycin (17-AAG) Hydroquinone by NAD(P)H:Quinone Oxidoreductase 1: Role of 17-AAG Hydroquinone in Heat Shock Protein 90 Inhibition Wenchang Guo, Philip Reigan, David Siegel, Joseph Zirrolli, Daniel Gustafson, and David Ross Department of Pharmaceutical Sciences, School of Pharmacy and Cancer Center, University of Colorado Health Sciences Center, Denver, Colorado Abstract We have examined the role of NAD(P)H:quinone oxidoreduc- tase 1 (NQO1) in the bioreductive metabolism of 17-allyla- mino-demethoxygeldanamycin (17-AAG). High-performance liquid chromatography (HPLC) analysis of the metabolism of 17-AAG by recombinant human NQO1 revealed the formation of a more polar metabolite 17-AAGH 2 . The formation of 17-AAGH 2 was NQO1 dependent, and its formation could be inhibited by the addition of 5-methoxy-1,2-dimethyl-3-[(4- nitrophenoxy)methyl]indole-4,7-dione (ES936), a mechanism- based (suicide) inhibitor of NQO1. The reduction of 17-AAG to the corresponding hydroquinone 17-AAGH 2 was confirmed by tandem liquid chromatography-mass spectrometry. 17-AAGH 2 was relatively stable and only slowly underwent autooxidation back to 17-AAG over a period of hours. To examine the role of NQO1 in 17-AAG metabolism in cells, we used an isogenic pair of human breast cancer cell lines differing only in NQO1 levels. MDA468 cells lack NQO1 due to a genetic polymorphism, and MDA468/NQ16 cells are a stably transfected clone that express high levels of NQO1 protein. HPLC analysis of 17-AAG metabolism using cell sonicates and intact cells showed that 17-AAGH 2 was formed by MDA468/NQ16 cells, and formation of 17-AAGH 2 could be inhibited by ES936. No 17-AAGH 2 was detected in sonicates or intact MDA468 cells. Following a 4-hour treatment with 17-AAG, the MDA468/NQ16 cells were 12-fold more sensitive to growth inhibition compared with MDA468 cells. More importantly, the increased sensitivity of MDA468/NQ16 cells to 17-AAG could be abolished if the cells were pretreated with ES936. Cellular markers of heat shock protein (Hsp) 90 inhibition, Hsp70 induction, and Raf-1 degradation were measured by immunoblot analysis. Marked Hsp70 induction and Raf-1 degradation was observed in MDA468/NQ16 cells but not in MDA468 cells. Similarly, downstream Raf-1 signaling molecules mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK) kinase and ERK also showed decreased levels of phosphory- lation in MDA468/NQ16 cells but not in MDA468 cells. The ability of 17-AAG and 17-AAGH 2 to inhibit purified yeast and human Hsp90 ATPase activity was examined. Maximal 17-AAG–induced ATPase inhibition was observed in the presence of NQO1 and could be abrogated by ES936, showing that 17-AAGH 2 was a more potent Hsp90 inhibitor compared with 17-AAG. Molecular modeling studies also showed that due to increased hydrogen bonding between the hydroqui- none and the Hsp90 protein, 17-AAGH 2 was bound more tightly to the ATP-binding site in both yeast and human Hsp90 models. In conclusion, these studies have shown that reduction of 17-AAG by NQO1 generates 17-AAGH 2 , a relatively stable hydroquinone that exhibits superior Hsp90 inhibition. (Cancer Res 2005; 65(21): 10006-15) Introduction Heat shock protein (Hsp) 90 has been developed as a potential anticancer target and is an attractive target for several reasons (1–3). Hsp90 is a protein chaperone that uses the hydrolysis of ATP to assist in the folding of early nascent forms of client proteins to their mature, correctly folded forms. Once the client protein has been correctly folded, Hsp90 is released, and as such, it functions as a true protein ‘‘catalyst.’’ The basis for Hsp90 as an anticancer target is that this chaperone assists in the folding of many oncogenic proteins. Such proteins include ErbB2, Raf-1, mutant p53, estrogen, and steroid receptors. Thus, by inhibiting Hsp90, one can target a large number of downstream proteins and thereby attack the neoplastic process at several points (2–4). The first Hsp90 inhibitor used clinically was geldanamycin, which did not move forward in clinical trials due to liver toxicity. Second- generation derivatives, such as 17-allylamino-demethoxygeldana- mycin (17-AAG) and 17-dimethylaminogeldanamycin (17-DMAG), do not induce liver toxicity, have completed phase I, and are currently entering phase II clinical trials (2, 3, 5, 6). The product of cytochrome P 450–mediated dealkylation of 17-AAG and 17-DMAG at the 17 position, 17-AG, retains its quinone functionality and is also a Hsp90 inhibitor (7). Because 17-AAG and related benzoquinone ansamycins contain a quinone moiety, bioreduction to semiquinone and hydroquinone species is a possible metabolic pathway within tumor cells, and formation of these species will depend on the levels of bioreductive enzymes. Among the bioreductive enzymes expressed in cancer cells poised to have the greatest influence on 17-AAG metabolism is NAD(P)H:quinone oxidoreductase 1 (NQO1; DT-diaphorase, EC 1.6.99.2). This flavoenzyme can use either NADH or NADPH as reducing cofactors and can catalyze the direct two-electron reduction of quinones to hydroquinones (8). NQO1 is expressed at high levels in many human cancers, including lung, colon, stomach, pancreatic, and breast (9–11). NQO1 has also been shown to Requests for reprints: David Ross, Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Health Sciences Center, 4200 East 9th Avenue, Denver, CO 80262. Phone: 303-315-6077; Fax: 303-315-0274; E-mail: david. [email protected]. I2005 American Association for Cancer Research. doi:10.1158/0008-5472.CAN-05-2029 Cancer Res 2005; 65: (21). November 1, 2005 10006 www.aacrjournals.org Research Article Research. on January 25, 2020. © 2005 American Association for Cancer cancerres.aacrjournals.org Downloaded from

Formation of 17-Allylamino-Demethoxygeldanamycin (17-AAG ...Hydroquinone by NAD(P)H:Quinone Oxidoreductase 1: Role of 17-AAG Hydroquinone in Heat Shock Protein 90 Inhibition Wenchang

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Formation of 17-Allylamino-Demethoxygeldanamycin (17-AAG ...Hydroquinone by NAD(P)H:Quinone Oxidoreductase 1: Role of 17-AAG Hydroquinone in Heat Shock Protein 90 Inhibition Wenchang

Formation of 17-Allylamino-Demethoxygeldanamycin (17-AAG)

Hydroquinone by NAD(P)H:Quinone Oxidoreductase 1:

Role of 17-AAG Hydroquinone in Heat Shock

Protein 90 Inhibition

Wenchang Guo, Philip Reigan, David Siegel, Joseph Zirrolli,Daniel Gustafson, and David Ross

Department of Pharmaceutical Sciences, School of Pharmacy and Cancer Center, University ofColorado Health Sciences Center, Denver, Colorado

Abstract

We have examined the role of NAD(P)H:quinone oxidoreduc-tase 1 (NQO1) in the bioreductive metabolism of 17-allyla-mino-demethoxygeldanamycin (17-AAG). High-performanceliquid chromatography (HPLC) analysis of the metabolism of17-AAG by recombinant human NQO1 revealed the formationof a more polar metabolite 17-AAGH2. The formation of17-AAGH2 was NQO1 dependent, and its formation could beinhibited by the addition of 5-methoxy-1,2-dimethyl-3-[(4-nitrophenoxy)methyl]indole-4,7-dione (ES936), a mechanism-based (suicide) inhibitor of NQO1. The reduction of 17-AAG tothe corresponding hydroquinone 17-AAGH2 was confirmed bytandem liquid chromatography-mass spectrometry. 17-AAGH2

was relatively stable and only slowly underwent autooxidationback to 17-AAG over a period of hours. To examine the role ofNQO1 in 17-AAG metabolism in cells, we used an isogenic pairof human breast cancer cell lines differing only in NQO1 levels.MDA468 cells lack NQO1 due to a genetic polymorphism, andMDA468/NQ16 cells are a stably transfected clone that expresshigh levels of NQO1 protein. HPLC analysis of 17-AAGmetabolism using cell sonicates and intact cells showed that17-AAGH2 was formed by MDA468/NQ16 cells, and formationof 17-AAGH2 could be inhibited by ES936. No 17-AAGH2

was detected in sonicates or intact MDA468 cells. Following a4-hour treatment with 17-AAG, the MDA468/NQ16 cells were12-fold more sensitive to growth inhibition compared withMDA468 cells. More importantly, the increased sensitivity ofMDA468/NQ16 cells to 17-AAG could be abolished if the cellswere pretreated with ES936. Cellular markers of heat shockprotein (Hsp) 90 inhibition, Hsp70 induction, and Raf-1degradation were measured by immunoblot analysis. MarkedHsp70 induction and Raf-1 degradation was observed inMDA468/NQ16 cells but not in MDA468 cells. Similarly,downstream Raf-1 signaling molecules mitogen-activatedprotein kinase/extracellular signal-regulated kinase (ERK)kinase and ERK also showed decreased levels of phosphory-lation in MDA468/NQ16 cells but not in MDA468 cells. Theability of 17-AAG and 17-AAGH2 to inhibit purified yeastand human Hsp90 ATPase activity was examined. Maximal

17-AAG–induced ATPase inhibition was observed in thepresence of NQO1 and could be abrogated by ES936, showingthat 17-AAGH2 was a more potent Hsp90 inhibitor comparedwith 17-AAG. Molecular modeling studies also showed thatdue to increased hydrogen bonding between the hydroqui-none and the Hsp90 protein, 17-AAGH2 was bound moretightly to the ATP-binding site in both yeast and human Hsp90models. In conclusion, these studies have shown thatreduction of 17-AAG by NQO1 generates 17-AAGH2, a relativelystable hydroquinone that exhibits superior Hsp90 inhibition.(Cancer Res 2005; 65(21): 10006-15)

Introduction

Heat shock protein (Hsp) 90 has been developed as a potentialanticancer target and is an attractive target for several reasons(1–3). Hsp90 is a protein chaperone that uses the hydrolysis of ATPto assist in the folding of early nascent forms of client proteins totheir mature, correctly folded forms. Once the client protein hasbeen correctly folded, Hsp90 is released, and as such, it functions asa true protein ‘‘catalyst.’’ The basis for Hsp90 as an anticancertarget is that this chaperone assists in the folding of manyoncogenic proteins. Such proteins include ErbB2, Raf-1, mutantp53, estrogen, and steroid receptors. Thus, by inhibiting Hsp90,one can target a large number of downstream proteins and therebyattack the neoplastic process at several points (2–4). The firstHsp90 inhibitor used clinically was geldanamycin, which didnot move forward in clinical trials due to liver toxicity. Second-generation derivatives, such as 17-allylamino-demethoxygeldana-mycin (17-AAG) and 17-dimethylaminogeldanamycin (17-DMAG),do not induce liver toxicity, have completed phase I, and arecurrently entering phase II clinical trials (2, 3, 5, 6). The product ofcytochrome P450–mediated dealkylation of 17-AAG and 17-DMAGat the 17 position, 17-AG, retains its quinone functionality and isalso a Hsp90 inhibitor (7).Because 17-AAG and related benzoquinone ansamycins contain

a quinone moiety, bioreduction to semiquinone and hydroquinonespecies is a possible metabolic pathway within tumor cells, andformation of these species will depend on the levels of bioreductiveenzymes. Among the bioreductive enzymes expressed in cancercells poised to have the greatest influence on 17-AAG metabolismis NAD(P)H:quinone oxidoreductase 1 (NQO1; DT-diaphorase,EC 1.6.99.2). This flavoenzyme can use either NADH or NADPHas reducing cofactors and can catalyze the direct two-electronreduction of quinones to hydroquinones (8). NQO1 is expressed athigh levels in many human cancers, including lung, colon, stomach,pancreatic, and breast (9–11). NQO1 has also been shown to

Requests for reprints: David Ross, Department of Pharmaceutical Sciences,School of Pharmacy, University of Colorado Health Sciences Center, 4200 East 9thAvenue, Denver, CO 80262. Phone: 303-315-6077; Fax: 303-315-0274; E-mail: [email protected].

I2005 American Association for Cancer Research.doi:10.1158/0008-5472.CAN-05-2029

Cancer Res 2005; 65: (21). November 1, 2005 10006 www.aacrjournals.org

Research Article

Research. on January 25, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 2: Formation of 17-Allylamino-Demethoxygeldanamycin (17-AAG ...Hydroquinone by NAD(P)H:Quinone Oxidoreductase 1: Role of 17-AAG Hydroquinone in Heat Shock Protein 90 Inhibition Wenchang

increase the cytotoxicity of many quinone-containing antitumordrugs, such as AZQ, mitomycin C, EO9, streptonigrin, RH-1, andh-lapachone, by reduction to hydroquinone species (12–17). Clinicaltrials that target tumors expressing high levels of NQO1 withquinone-based agents are currently in progress.The role of NQO1 in benzoquinone ansamycin metabolism was

first described by Kelland et al. (18). In this work, the authorsshowed a positive correlation between 17-AAG sensitivity andNQO1 expression. 17-AAG was shown to undergo NQO1-mediatedreduction using purified NQO1, and human cancer cell linesexpressing NQO1 were more sensitive to 17-AAG (18). We haveextended these studies to examine the properties of the hydroqui-none formed after reduction of 17-AAG by purified NQO1. We alsoused the human breast cancer cell line MDA468 (NQO1 null) and anisogenic paired cell line MDA468/NQ16, differing only in NQO1levels, in combination with a mechanism-based inhibitor of NQO1[5-methoxy-1,2-dimethyl-3-[(4-nitrophenoxy)methyl]indole-4,7-dione (ES936)] to confirm the role of NQO1 in the bioreduction of17-AAG. Studies with purified yeast and human Hsp90 showed that17-AAGH2 was a more potent inhibitor of Hsp90, and molecularmodeling studies confirmed that 17-AAGH2 had a greater affinity forthe ATP-binding site in yeast and human Hsp90. Our studies showthat 17-AAG can be reduced to 17-AAGH2 by NQO1 and that thehydroquinone is a more potent Hsp90 inhibitor than 17-AAG.

Materials and Methods

Materials. 17-Allylamino-demethoxygeldanamycin (17-AAG) was pro-vided by the National Cancer Institute and Kosan Biosciences (Hayward,

CA). 2,6-Dichlorophenol-indophenol (DCPIP), NADH, NADPH, sodium

borohydride, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide(MTT), bovine serum albumin (BSA), h-lapachone, N-phenyl-1-naphthyl-amine, D(�)-penicillamine, and mouse monoclonal (IgG) and anti-actin

antibodies were obtained from the Sigma Chemical Co. (St. Louis MO).

Yeast Hsp90 and radicicol were obtained from Alexis (San Diego, CA).Human Hsp90 was a kind gift from Dr. David Toft (Mayo Clinic College of

Medicine, Rochester, MN). Recombinant human NQO1 (rhNQO1) was

purified from Escherichia coli as described previously (19). The activity of

rhNQO1 was 4.5 Amol DCPIP/min/mg protein. Malachite green phosphateassay kit was obtained from BioAssay Systems, Inc. (Hayward, CA). Mouse

anti-Hsp70 and rabbit anti-Raf-1 antibodies were obtained from Stressgen

(Vancouver, British Columbia, Canada). Rabbit anti–mitogen-activatedprotein kinase/extracellular signal-regulated kinase (ERK) kinase (MEK)/

phospho-MEK and ERK/phospho-ERK antibodies were obtained from Cell

Signaling (Beverly, MA).

Cell lines. The human breast cancer cell line MDA468 and the NQO1stably transfected cell line MDA468/NQ16 have been described previously

(20). Cells were grown in RPMI 1640 containing 10% (v/v) fetal bovine serum

and 1% (v/v) penicillin, streptomycin, and glutamine. Sonicates were

prepared from MDA468 and MDA468/NQ16 cell lines by probe sonication inice-cold 25 mmol/L Tris-HCl (pH 7.4) containing 250 mmol/L sucrose and

5 Amol/L flavin adenine dinucleotide. Protein concentrations were

determined by the method of Lowry et al. (21).

Inhibition of NAD(P)H:quinone oxidoreductase 1 by 5-methoxy-1,2-dimethyl-3-[(4-nitrophenoxy)methyl]indole-4,7-dione. ES936 was syn-

thesized by Christopher J. Moody (Department of Chemistry, University of

Exeter, Exeter, United Kingdom). ES936 has been shown previously to be apotent mechanism-based inhibitor of NQO1 (22). For these studies, a single

dose of 100 nmol/L ES936 was nontoxic and resulted in >96% inhibition of

NQO1 activity after 4 hours in MDA468/NQ16 cells. Inhibition of rhNQO1 by

ES936 was >98%.High-performance liquid chromatography analysis. The metabolism

of 17-AAG by NQO1 was analyzed by high-performance liquid chromatog-

raphy (HPLC) on a Luna C18 reverse-phase column (5 Am, 4.6 � 250 mm,

Phenomenex, Torrance, CA) at room temperature. HPLC conditions were

as follows: buffer A, 50 mmol/L ammonium acetate (pH 4) containing10 Amol/L D(�)-penicillamine; buffer B, acetonitrile (100%). Both buffers

were continuously bubbled with N2 gradient, 20% B to 85% B over

20 minutes and then 85% for 4 minutes ( flow rate of 1 mL/min). The sample

injection volume was 50 AL. LC-mass spectrometry (MS) was done using

positive ion electrospray ionization and the mass spectra were obtained

with a PE Sciex API-3000 triple quadrupole MS (Foster City, CA) with a

turbo ion spray source interfaced to a PE Sciex 200 HPLC system. Samples

were chromatographed on a Luna C18 reverse-phase column (5 Am, 50 �2 mm, Phenomenex) using a gradient elution consisting of a 2-minute initial

hold at 20% B followed by an increase to 80% B over 20 minutes at a flow

rate of 0.2 mL/min and a sample injection volume of 20 AL. HPLC

conditions were as follows: buffer A, 10 mmol/L ammonium acetate

containing 0.1% (v/v) acetic acid (pH 4.4): buffer B, 10 mmol/L ammonium

acetate in acetonitrile containing 0.1% (v/v) acetic acid. The MS settings

were turbo ion spray temperature of 250jC, spray needle voltage at 4,500 V,

declustering potential at 35 V, and focus plate at 125 V. Mass spectra were

continuously recorded from 150 to 1,000 amu every 3 seconds during the

chromatographic analysis.Growth inhibition assays. Growth inhibition was measured using the

MTT assay. Cells were seeded at 2 � 103 per well (96-well plate) in complete

medium overnight. The next morning, the cells were pretreated with

100 nmol/L ES936 or an equal amount of DMSO for 30 minutes and thenexposed to 17-AAG for 4 hours, after which cells were rinsed free of drug

and incubated in fresh medium for an additional 72 hours. Cell viability was

measured by the MTT assay as described previously (23).

17-Allylamino-demethoxygeldanamycin metabolism in MDA468 andMDA468/NQ16 cells. The metabolism of 17-AAG in cells was done under

the following conditions: Cells were seeded at 2 � 106 per 100-mm plate

(in duplicate) in complete medium for 3 days. Cells were then pretreated

with 100 nmol/L ES936 or an equal amount of DMSO for 30 minutesfollowed by 5 Amol/L 17-AAG for additional 4 hours at 37jC. Followingdrug treatment, cells (one plate) were washed extensively in PBS containing

1% (v/v) BSA and then lysed on the plate by the addition of 200 AL ice-coldacetonitrile containing 1 Ag/mL N -phenyl-1-naphthylamine (internal

standard). Samples were centrifuged at 13,000 rpm for 10 seconds and

the supernatant was immediately analyzed by HPLC. The cell number was

determined using the second plate of cells. Cells were washed in PBS andthen scraped in 200 AL PBS and counted (hemocytometer).

Immunoblot analysis. MDA468 and MDA468/NQ16 cells were grown in

100-mm plates in complete medium to f70% confluency. For Hsp70

and Raf-1 analysis, cells were treated with DMSO or 17-AAG (0.5-1 Amol/L)in 10 mL complete medium for 8 hours. For MEK and ERK analysis, cells

were serum starved for 24 hours and then treated with DMSO or 17-AAG

(0.5-1 Amol/L) in serum-free medium for 8 hours. Following drug treatment,cells were washed in PBS and then lysed by the addition of radio-

immunoprecipitation assay buffer [50 mmol/L Tris-HCl (pH 7.4), 0.5% (v/v)

NP40] containing 1 Mini protease tablet (protease inhibitor cocktail, Roche,

Indianapolis, IN) and phosphatase inhibitors (30 mmol/L NaF, 40 mmol/Lh-glycerophosphate, 20 mmol/L sodium pyrophosphate, 1 mmol/L ortho-

vanadate, 1 mmol/L EGTA). Lysates were probe sonicated (2 seconds) on ice

and then centrifuged to remove cellular debris. Protein concentrations were

determined on supernatant by the method of Lowry et al. (21). Sampleswere heated to 70jC in 2� Laemmli SDS sample buffer, and proteins were

separated by 12% SDS-PAGE (precast minigel, Bio-Rad, Hercules CA) and

then transferred to 0.4-Am polyvinylidene difluoride membranes. Mem-

branes were blocked in 10 mmol/L Tris-HCl (pH 8.0), 150 mmol/L NaCl,0.2% Tween 20, and 5% nonfat milk for a minimum of 1 hour at room

temperature. Anti-Hsp70, anti-Raf-1, anti-MEK, and anti-ERK antibodies

were added for 1 hour at room temperature. Anti-phospho-MEK and ERKantibodies were added overnight at 4jC. All primary antibodies were

diluted 1:1,000m except actin (1:5,000). Horseradish peroxidase–labeled

secondary antibodies (Jackson ImmunoResearch Labs, West Grove, PA)

were diluted 1:5,000 and added for 30 minutes. Proteins were visualizedusing enhanced chemiluminescence detection.

Heat shock protein 90 ATPase activity assay. Inhibition of Hsp90

ATPase was measured as described previously (24). Briefly, purified yeast or

Metabolism of 17-AAG by NQO1

www.aacrjournals.org 10007 Cancer Res 2005; 65: (21). November 1, 2005

Research. on January 25, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 3: Formation of 17-Allylamino-Demethoxygeldanamycin (17-AAG ...Hydroquinone by NAD(P)H:Quinone Oxidoreductase 1: Role of 17-AAG Hydroquinone in Heat Shock Protein 90 Inhibition Wenchang

human Hsp90 (2.5 Ag) was incubated in 100 mmol/L Tris-HCl (pH 7.4)

containing 20 mmol/L KCl, 6 mmol/L MgCl2, 400 Amol/L NADH, 17-AAGwith or without 3.3 Ag rhNQO1, and 20 Amol/L ES936. Reactions (25 AL)were started by the addition of 1 mmol/L ATP and allowed to proceed at

37jC for the indicated times. Reactions were then diluted with 225 AL of

100 mmol/L Tris-HCl (pH 7.4) containing 20 mmol/L KCl and 6 mmol/LMgCl2 mixed thoroughly, and 80 AL were transferred to each well (96-well

plate) followed by 20 AL malachite green reagent. After 10 minutes,

trisodium citrate (83 mmol/L) was added to stabilize the color and plates

were read at 650 nm.Molecular modeling of 17-allylamino-demethoxygeldanamycin and

17-AAGH2 in yeast and human heat shock protein 90 crystalstructures. Molecular modeling studies were done on a Silicon Graphics

Octane2 workstation. The crystallographic coordinates for the 2.5-Astructure of yeast Hsp90 (PDB: 1A4H; ref. 25) and the 1.9-A structure of

human Hsp90 (PDB: 1YET; ref. 26) were obtained from the Research

Collaboratory for Structural Bioinformatics Protein Data Bank.1 The Hsp90crystal structures were visualized using the InsightII software package

version 2000 (Accelrys, Inc., San Diego, CA). The Builder Module was used

to add hydrogens to the protein structure and the ionizable Asp, Arg, Glu,and Lys residues were corrected for physiologic pH. The 17-AAG and

the corresponding hydroquinone, 17-AAGH2, structures were constructed

and assigned the correct atom type and bond order, from the bound

geldanamycin structure, in each Hsp90 crystal structure. Once constructed,the ligands were in turn superpositioned onto the bound geldanamycin

structure using the coordinated system of the protein to correctly position

the ligand in the geldanamycin-binding domain of Hsp90. The geldanamy-

cin scaffold structure was then deleted from the protein structure and the‘‘docked’’ ligand assembly was associated with the Hsp90 protein structure.

For the molecular mechanics and molecular dynamics calculations, the

Discover Module was used and the potentials and charges of the Hsp90-

ligand complex were corrected using the consistent-valence force field. TheHsp90-ligand complex was then minimized using the conjugate gradient

method (1,000 iterations). The Docking Module was used to perform the

intermolecular energy calculation to determine the nonbonded interactionenergy between Hsp90 and the appropriate ligand. An interface 6-A radius

subset encompassing the ligand-binding domain was selected and both van

der Waals and electrostatic (coulombic) energies were calculated with a

specified cutoff of 8 A.

Figure 1. HPLC and LC-MS analysis of the reduction of 17-AAG by NQO1. HPLC analysis confirmed the formation of 17-AAGH2 following reduction of 17-AAG byrhNQO1 (A and B ) and inhibition of this reduction by ES936 (C ). Reaction conditions: 50 Amol/L 17-AAG, 200 Amol/L NADH, and 3.3 Ag rhNQO1 in 50 mmol/Lpotassium phosphate buffer (pH 7.4; 1 mL) containing 1 mg/mL BSA. After 40 minutes, reactions were stopped with an equal volume of acetonitrile containing internalstandard N -phenyl-1-naphthylamine (5 Ag/mL) and centrifuged and the supernatant was analyzed immediately by HPLC at 334 nm. A, 17-AAG and NADH;B, 17-AAG, NADH, and rhNQO1; C, 17-AAG, NADH, rhNQO1, and ES936 (1 Amol/L). The relatively small peak size of 17-AAGH2 compared with 17-AAG is due to lowabsorption at 334 nm for the hydroquinone. Inset, NQO1-mediated metabolism of 17-AAG (same conditions as in B) at a detection wavelength of 270 nm wherethe 17-AAG quinone and hydroquinone have approximately equal absorption. LC-MS was used to confirm 17-AAGH2 as the product of NQO1-mediated reduction of17-AAG (D ).

1 http://www.rcsb.org

Cancer Research

Cancer Res 2005; 65: (21). November 1, 2005 10008 www.aacrjournals.org

Research. on January 25, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 4: Formation of 17-Allylamino-Demethoxygeldanamycin (17-AAG ...Hydroquinone by NAD(P)H:Quinone Oxidoreductase 1: Role of 17-AAG Hydroquinone in Heat Shock Protein 90 Inhibition Wenchang

Results

In vitro studies with purified rhNQO1 clearly showed thatreduction of 17-AAG by NQO1 in combination with NADH orNADPH generated 17-AAGH2. HPLC analysis of aerobic incubationsof 17-AAG, NADH, and rhNQO1 resulted in loss of the 17-AAGpeak and generation of the more polar metabolite 17-AAGH2

(Fig. 1). Formation of 17-AAGH2 was NQO1 dependent and couldbe inhibited by addition of the NQO1 mechanism-based inhibitorES936. Identification of the more polar product as 17-AAGH2 wasconfirmed by LC-MS (Fig. 1D). 17-AAGH2 formed followingreduction of 17-AAG by NQO1 and NADH coeluted with themajor product generated during sodium borohydride reduction of17-AAG (data not shown). HPLC analysis showed that 90% of17-AAG was detected in the form of the hydroquinone afterreduction by NQO1, suggesting the generation of a relatively stablehydroquinone. During analysis, HLPC buffers were continuouslygassed with N2, and a copper chelator (see Materials and Methods)was included. Preliminary data (not included) showed thatinclusion of copper (CuSO4) could facilitate 17-AAGH2 oxidation.The stability of 17-AAGH2 in solution was measured by HPLCfollowing reduction of 17-AAG by rhNQO1 using stoichiometricequivalents of NADH and 17-AAG (Fig. 2). In these studies, theautooxidation of 17-AAGH2 was followed over time in an aerobicreaction. These results showed that 17-AAGH2 undergoes a slowrate of autooxidation back to 17-AAG over a period of hours.Whether 17-AAGH2 could be formed in cells following reduction

by NQO1 was investigated using the isogenic human breast cancercell lines MDA468 and MDA468/NQ16. These cell lines have beenused previously to examine the role of NQO1 in antitumor quinonemetabolism (20). The parental MDA468 cell lines is NQO1 null (<10nmol DCPIP/min/mg) due to homozygous expression of theNQO1*2 polymorphism. The stable transfection of the MDA468 cellline with human NQO1 generated the MDA468/NQ16 cell line,which has high NQO1 activity (>1,800 nmol DCPIP/min/mg). Initialexperiments were done using cell sonicates prepared from MDA468

and MDA468/NQ16 cells. HPLC analysis of these reactions wasnearly identical to results obtained with rhNQO1 (Fig. 1). Sonicatesprepared from MDA468/NQ16 cells readily generated 17-AAGH2

(Fig. 3A ). Generation of 17-AAGH2 was NADH or NADPH

Figure 2. Stability of 17-AAGH2. The stability of 17-AAGH2 was examined bymeasuring the autooxidation of 17-AAGH2 to 17-AAG over time by HPLC.Reaction conditions: 50 Amol/L quinone, 50 Amol/L NADH, and 3.3 Ag rhNQO1 in50 mmol/L potassium phosphate buffer (pH 7.4) containing 1 mg/mL BSA; totalvolume of 1 mL at 27jC. Reactions were stopped with an equal volume ofacetonitrile containing internal standard and samples were analyzed immediatelyby HPLC at 270 nm. Points, mean (n = 3); bars, SD.

Figure 3. HPLC analysis of 17-AAGH2 formation by MDA468 and MDA468/NQ16 cell sonicates and cells. HPLC analysis confirmed the formation of17-AAGH2 following reduction of 17-AAG by MDA468/NQ16 cell sonicates (A)and inhibition of this reduction by ES936 (B). Reaction conditions: 50 Amol/L17-AAG, 200 Amol/L NADH, and 500 Ag MDA468/NQ16 cell sonicates in50 mmol/L potassium phosphate buffer (pH 7.4; 1 mL) containing 1 mg/mL BSA.After 40 minutes, reactions were stopped with an equal volume of acetonitrilecontaining internal standard N-phenyl-1-naphthylamine (5 Ag/mL) andcentrifuged and the supernatant was analyzed immediately by HPLC at 334 nm.A, 17-AAG, NADH, and MDA468/NQ16 cell sonicates; B, 17-AAG, NADH, andMDA468/NQ16 cell sonicates and ES936 (1 Amol/L). C, 17-AAG (open columns )and 17-AAGH2 (filled columns ) were measured by HPLC at 270 nm in intactMDA468 and MDA468/NQ16 cells in the presence and absence of ES936. Cellswere pretreated with DMSO or ES936 (1 Amol/L) for 20 minutes and then treatedwith 17-AAG (5 Amol/L) for 4 hours. Following drug treatment, cells wereextensively washed, pelleted, and lysed by the addition of acetonitrile (200 AL)containing internal standard N -phenyl-1-naphthylamine (5 Ag/mL). The sampleswere centrifuged (13,000 rpm � 1 minute) and the supernatant was analyzedimmediately. Columns, mean (n = 3); bars, SD. *, P < 0.05, significantly differentfrom MDA468/NQ16 cells (one-way ANOVA with Tukey test for pairwisecomparison).

Metabolism of 17-AAG by NQO1

www.aacrjournals.org 10009 Cancer Res 2005; 65: (21). November 1, 2005

Research. on January 25, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 5: Formation of 17-Allylamino-Demethoxygeldanamycin (17-AAG ...Hydroquinone by NAD(P)H:Quinone Oxidoreductase 1: Role of 17-AAG Hydroquinone in Heat Shock Protein 90 Inhibition Wenchang

dependent and could be inhibited by ES936 (Fig. 3B). No 17-AAGH2

could be detected in sonicates from MDA468 cells. To furtherinvestigate the role of NQO1 in the reduction of 17-AAG, weexamined whether 17-AAGH2 could be generated by NQO1 inintact cells. In these experiments, MDA468 and MDA468/NQ16cells were treated with 17-AAG (5 Amol/L) for 4 hours and thenwashed free of drug and lysed by the addition of acetonitrilecontaining internal standard. Concentrations of 17-AAG and17-AAGH2 were then measured by HPLC, and the results clearlyshowed that 17-AAGH2 could be generated in MDA468/NQ16 cellsand formation of 17-AAGH2 could be inhibited by pretreatmentwith ES936 (Fig. 3C). No 17-AAGH2 could be detected in MDA468cells. In addition to generating 17-AAGH2, MDA468/NQ16 cellsalso accumulated more total drug (quinone plus hydroquinone)compared with cells treated with ES936 or parental MDA468 cells(Fig. 3C). Cell viability was >90% for both cell lines under thesetreatment condition (trypan blue exclusion assay).The role of NQO1 in growth inhibition induced by 17-AAG was

measured in MDA468 and MDA468/NQ16 cells. In these studies,cells were treated with 17-AAG for 4 hours in the presence andabsence of ES936 pretreatment. Cells were then washed free ofdrugs and growth inhibition was determined using the MTT assay

(Fig. 4). Results from these experiments showed that MDA468/NQ16 cells had increased sensitivity to 17-AAG (IC50, 0.86 F 0.16Amol/L) compared with parental MDA468 cells (IC50, 10.05 F 1.07Amol/L). The sensitivity to 17-AAG could be abrogated bypretreatment with ES936 (IC50, 7.67 F 1.36 Amol/L). Results weresimilar using an 8-hour exposure to 17-AAG with an f20-foldincrease in sensitivity in MDA468/NQ16 cells relative to MDA468cells, and growth inhibition in MDA468/NQ16 cells could alsobe abrogated by ES936. Experiments were also done using anonquinone Hsp90 inhibitor, radicicol, as a negative control. Thegrowth-inhibitory effects of radicicol were essentially identical inMDA468 and MDA468/NQ16 cells (data not shown), showing therequirement for a quinone functionality for increased growthinhibition in MDA468/NQ16 cells.The role of NQO1 in 17-AAG-induced Hsp90 inhibition in

MDA468 and MDA468/NQ16 cells was also examined. To measureHsp90 inhibition in these cells, we analyzed Hsp70 induction andRaf-1 degradation as markers of Hsp90 inhibition (Fig. 4). Thesestudies showed greater Hsp70 induction and increased Raf-1degradation in MDA468/NQ16 cells compared with MDA468 cells.We then examined downstream (Raf-1) signaling as reflected inMEK and ERK phosphorylation in MDA468 and MDA468/NQ16

Figure 4. Effect of 17-AAG on growth inhibition and Hsp90 client proteins in human breast cancer cells. Growth inhibition following 17-AAG treatment was measured byMTT analysis in MDA468 (NQO1-null) and MDA468/NQ16 (high NQO1) cell lines in the presence (filled symbols ) and absence (open symbols ) of ES936 (A).Points, mean (n = 3); bars, SD. B, effect of 17-AAG on Hsp70 and Raf-1 protein levels. Hsp70 and Raf-1 protein levels were analyzed by immunoblot analysis aftertreatment of MDA468 and MDA468/NQ16 cells with 17-AAG for 8 hours. Hsp70 immunoblot analysis was done on 25 Ag whole-cell sonicate. Raf-1 immunoblotanalysis was done on 50 Ag whole-cell sonicate. Results were confirmed in duplicate experiments. C, MEK and ERK phosphorylation was analyzed by immunoblotanalysis after treatment of MDA468 and MDA468/NQ16 cells with 17-AAG. Cells were serum starved for 24 hours and then treated with 17-AAG for 8 hours.Phospho-MEK (pMEK )/MEK immunoblot analysis was done on 25 Ag whole-cell sonicate. Phospho-ERK (pERK )/ERK immunoblot analysis was done on 50 Agwhole-cell sonicate. Results were confirmed in duplicate experiments. Similar data was obtained at a 16-hour time point (data not shown).

Cancer Research

Cancer Res 2005; 65: (21). November 1, 2005 10010 www.aacrjournals.org

Research. on January 25, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 6: Formation of 17-Allylamino-Demethoxygeldanamycin (17-AAG ...Hydroquinone by NAD(P)H:Quinone Oxidoreductase 1: Role of 17-AAG Hydroquinone in Heat Shock Protein 90 Inhibition Wenchang

cells treated with 17-AAG (Fig. 4). Decreased levels of bothMEK and ERK phosphorylation were observed in MDA468/NQ16cells compared with MDA468 cells following serum starvation. Nochanges in total MEK or ERK protein levels were observed (Fig. 4).These data suggested that a greater level of Hsp90 inhibition wasoccurring in NQO1-rich MDA468/NQ16 cells compared with NQO1-deficient MDA468 cells.To confirm the relationship between 17-AAGH2 formation and

increased Hsp90 inhibition, we examined the ability of 17-AAGand 17-AAGH2 to inhibit the ATPase activity of purified yeast andhuman Hsp90 (Fig. 5). In these assays, we incubated purified yeastor human Hsp90 with 17-AAG (2 Amol/L) and NADH (400 Amol/L)in the presence and absence of rhNQO1. Reactions were allowedto proceed for 3 hours (yeast Hsp90) or 12 hours (human Hsp90),after which reactions were terminated and the concentration ofinorganic phosphate was measured using the malachite green assay(24). Results from experiments using yeast Hsp90 showed that17-AAGH2 was the active Hsp90 inhibitor. In these studies, asubstantial decrease in ATPase activity was observed with 17-AAG

in the presence of NQO1 and this could be prevented by ES936. Noinhibition by 17-AAG was observed in the absence of NQO1. Resultsfrom experiments using human Hsp90 were qualitatively similarbut showed some quantitative differences. In these experiments,the addition of 17-AAG resulted in some inhibition of Hsp90ATPase activity. Inhibition was significantly increased, however, bythe inclusion of NQO1 and abrogated by inactivation of NQO1 byES936 (Fig. 5).Molecular modeling studies of 17-AAG and 17-AAGH2 docked

into the ATP-binding site of the yeast or human Hsp90 crystalstructure revealed significant differences in the binding energiesbetween the two compounds after minimization. The nonbondedinteraction energy or binding energy is the sum of the van der Waalsand electrostatic energies, the measure of the affinity betweenHsp90 and 17-AAG/17-AAGH2. In both yeast (Table 1) and human(Table 2) Hsp90 crystal structures, 17-AAGH2 had greater nonbond-ed interaction energy than the parent quinone. These data supportthe hypothesis that the hydroquinone form of 17-AAGH2 is a morepotent inhibitor of Hsp90. Following minimization, the Hsp90-ligand complex was visualized to identify important amino acidresidues in the ATP-binding domain that interact via hydrogenbonding with the ligand investigated (Fig. 6); there was nosignificant change in the global conformation of Hsp90. However,the Hsp90-17-AAGH2 complex revealed additional hydrogenbonding interactions between the hydroquinone moiety andHsp90 that resulted in a greater binding energy (Tables 1 and 2).In the ATP-binding domain of yeast Hsp90, the C21 ketone of

17-AAG hydrogen bonds with the amine of Lys98 and the C18ketone of the quinone ring system interacts with a water moleculethat in turn contacts Asp40, whereas in 17-AAGH2 the oxygen atomof the C21 hydroxyl does not seem to interact directly with Lys98.The hydrogen atom of the C18 hydroxyl interacts with an oxygenatom of the carboxylate side chain of Asp40, which subtly alters theoverall conformation of 17-AAGH2 and allows the amide of theansa ring to interact with the backbone nitrogen of Phe124.Interestingly, no interactions were observed with yeast Hsp90 andthe 17-(2-propenylamino)-substituent; the side chain is orientatedinto solvent and does not interfere with the binding of the ligand tothe target site.Similar interactions were observed in the ATP-binding domain of

human Hsp90 with 17-AAG; the C21 ketone hydrogen bonds withthe amine of Lys112 and the C18 ketone interacts with a watermolecule that makes contact with Asn51. In both 17-AAG and 17-AAGH2, the amide of the ansa ring interacts with the backbonenitrogen of Phe138 and the C11 hydroxyl group hydrogen bonds withthe amine of Lys58 in human Hsp90. However, there is arearrangement of protein-ligand interactions in the binding siteaccompanying the substitution of the hydroquinone for thebenzoquinone moiety. The interaction of the C21 hydroxyl andthe amine of the Lys112 is maintained, although at a greaterhydrogen bond distance, in contrast to that observed with yeastHsp90. The C18 hydroxyl interacts with Asp54; as a result, thehydroquinone moiety of 17-AAGH2 adopts a more compactconformation around the amino acid residues of helix 2. Thisallows hydrogen bonding between the 17-NH group of the 17-(2-propenylamino)-substituent and the carboxylate side chain of Asp54

and the side chain amine of Lys58. The hydrogen bonding betweenthe 17-amino group of 17-AAGH2 and Asp54 and Lys58 is the onlyprotein-ligand interaction provided by the 2-propenylamino group,because the remainder of the side chain is orientated away from thebinding site and into the solvent.

Figure 5. Inhibition of yeast and human Hsp90 by 17-AAG and 17-AAGH2.Yeast and human Hsp90 ATPase activity was measured in reactions with eithervehicle (DMSO) or 17-AAG in the presence and absence of rhNQO1. Reactionswith yeast Hsp90 (top ) were analyzed after 3 hours, whereas reactions withhuman Hsp90 (bottom ) were analyzed after 12 hours. Phosphate concentrationswere measured using the malachite green assay. Open columns, 17-AAG andNADH; filled columns, 17-AAG, NADH, and NQO1; hatched columns, 17-AAG,NADH, NQO1, and ES936. Columns, mean (n = 3); bars, SD. Hsp90 ATPaseactivity in incubates containing 17-AAG, NADH, and NQO1 were statisticallydifferent from incubates containing either 17-AAG and NADH or 17-AAG, NADH,NQO1, and ES936, *, P < 0.05, one-way ANOVA with Tukey for pairwisecomparison.

Metabolism of 17-AAG by NQO1

www.aacrjournals.org 10011 Cancer Res 2005; 65: (21). November 1, 2005

Research. on January 25, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 7: Formation of 17-Allylamino-Demethoxygeldanamycin (17-AAG ...Hydroquinone by NAD(P)H:Quinone Oxidoreductase 1: Role of 17-AAG Hydroquinone in Heat Shock Protein 90 Inhibition Wenchang

Discussion

Kelland et al. (18) in the first study of the role of NQO1 in themechanism of action of benzoquinone ansamycins showed arelationship between levels of NQO1 and sensitivity to 17-AAG.Kelland et al. also reported that NQO1 may play a role in thecellular metabolism and activity of Hsp90 inhibitors because they

observed increased 17-AAG and geldanamycin activity in NQO1-

transfected BE cells relative to NQO1-null BE parental cells (18).

This was an intriguing result and we examined the activity of 17-

AAG in the MDA468 and MDA468/NQ16 isogenic cell pair.

Consistent with the data of Kelland et al. (18), we found markedly

increased 17-AAG growth inhibition in MDA468/NQ16 cells with

Table 1. Total interaction energy, van der Waals, electrostatic energy, and hydrogen bonding interactions between yeastHsp90 and 17-AAG/17-AAGH2

Ligand Evdw

(kcal/mol)Eelect

(kcal/mol)E total

(kcal/mol)H-bond interactions H-bond

distance (A)

17-AAG �36.6 �25.0 �61.6 Asp79: carbamate NH2 2.03Lys98: quinone C=O 1.91

Phe124: amide (ansa) C=O 2.43

HOH: carbamate C=O 2.04HOH: methoxy (ansa) OCH3 2.47

HOH: carbamate OCONH2 2.32

HOH: hydroxy (ansa) OH 2.39

HOH: quinone C=O 1.83HOH: amide (ansa) NH 1.89

17-AAGH2 �39.1 �29.2 �68.2 Asp40: hydroquinone O-H 2.40

Asp79: carbamate NH2 2.00

Phe124: amide (ansa) C=O 2.35HOH: carbamate C=O 2.05

HOH: carbamate NH2 2.25

HOH: methoxy (ansa) OCH3 2.30

HOH: carbamate OCONH2 2.50HOH: hydroxy (ansa) OH 2.38

HOH: hydroquinone O-H 2.33

Abbreviations: E total, total interaction energy; Evdw, van der Waals; Eelect, electrostatic energy.

Table 2. Total interaction energy, van der Waals, electrostatic energy, and hydrogen bonding interactions between humanHsp90 and 17-AAG/17-AAGH2

Ligand Evdw

(kcal/mol)Eelect

(kcal/mol)E total

(kcal/mol)H-bond interactions H-bond

distance (A)

17-AAG �23.5 �4.6 �28.1 Lys58: hydroxy (ansa) O-H 1.69Asp93: carbamate NH2 2.06

Lys112: quinone C=O 1.84

Phe138: amide (ansa) C=O 2.08

HOH: carbamate NH2 2.32HOH: quinone C=O 2.10

HOH: carbamate C=O 2.11

HOH: methoxy (ansa) OCH3 2.37

17-AAGH2 �22.3 �15.2 �37.5 Asp54: hydroquinone O-H 1.96Asp54: amine NH 2.27

Lys58: hydroxy (ansa) O-H 1.72

Lys58: amine NH 2.44Asp93: carbamate NH2 2.04

Lys112: hydroquinone O-H 1.90

Phe138: amide (ansa) C=O 2.00

HOH: carbamate NH2 2.27HOH: carbamate C=O 2.05

HOH: carbamate OCONH2 2.47

HOH: methoxy (ansa) OCH3 2.32

Cancer Research

Cancer Res 2005; 65: (21). November 1, 2005 10012 www.aacrjournals.org

Research. on January 25, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 8: Formation of 17-Allylamino-Demethoxygeldanamycin (17-AAG ...Hydroquinone by NAD(P)H:Quinone Oxidoreductase 1: Role of 17-AAG Hydroquinone in Heat Shock Protein 90 Inhibition Wenchang

high NQO1 relative to the NQO1-deficient parental MDA468 cellline. The MDA468 cell line is deficient in NQO1 because thehomozygous NQO1*2 polymorphism that we have shown previ-ously leads to rapid degradation of NQO1*2 protein (27). Radicicol,a non-quinone-containing Hsp90 inhibitor, showed essentiallyidentical growth-inhibitory profiles in MDA468 and MDA468/NQ16 cells showing the requirement for a quinone functionality forincreased sensitivity in cells containing elevated NQO1 levels. Theinteresting aspect of our data was that we were able to extendprevious work by use of ES936, a mechanism-based inhibitor ofNQO1 (22). We showed that inhibition of NQO1 with ES936 couldabrogate the growth-inhibitory activity of 17-AAG in NQ16 cells,raising IC50 values back to approximately those observed inthe parental MDA468 cells. Increased client protein inhibition

and downstream Raf-1 mediated signaling could be shown inNQ16 cells relative to MDA468 cells. These observations arguedstrongly that 17-AAGH2 was a more potent inhibitor than 17-AAGitself.Using HPLC analysis, we showed the generation of 17-AAG

hydroquinone from 17-AAG, by recombinant NQO1, in NQ16sonicates and intact cells but not in MDA468 sonicates or cells.The generation of 17-AAGH2 by recombinant NQO1 was verified byLC-MS. The production of the hydroquinone of 17-AAG in NQ16sonicates and cells could be blocked by use of ES936, the suicideinhibitor of NQO1, which was entirely consistent with cellulargrowth inhibition and client protein inhibition data. In agreementwith previous data (28), it seems that aside from leading to theformation of 17-AAGH2 the presence of NQO1 in a cell also leads to

Figure 6. Molecular modeling of the Hsp90-17-AAG/17-AAGH2 complex. Flat ribbon representation of the yeast Hsp90 ATP-binding domain with (A ) 17-AAG and(B) 17-AAGH2 and human Hsp90 ATP-binding domain with (C ) 17-AAG and (D) 17-AAGH2 (ligands are yellow ; stick display style) displaying hydrogen bond contacts(green dashed lines ) with key amino acid residues and water molecules (stick display style). The carbon atoms are in gray , hydrogen atoms are in white , nitrogenatoms are in blue , and oxygen atoms are in red unless otherwise stated. The figures were constructed using Discovery Studio Viewer Professional Software (Accelrys).

Metabolism of 17-AAG by NQO1

www.aacrjournals.org 10013 Cancer Res 2005; 65: (21). November 1, 2005

Research. on January 25, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 9: Formation of 17-Allylamino-Demethoxygeldanamycin (17-AAG ...Hydroquinone by NAD(P)H:Quinone Oxidoreductase 1: Role of 17-AAG Hydroquinone in Heat Shock Protein 90 Inhibition Wenchang

increased total accumulation of (quinone plus hydroquinone)ansamycin equivalents. Presumably, this reflects the increasedwater solubility of 17-AAGH2 and a decreased propensity of thehydroquinone once generated in a cellular system to crossmembranes. Thus, reduction of 17-AAG by NQO1 may not onlylead to the generation of the more potent form of the Hsp90inhibitor but also result in the retention of the more activehydroquinone form in the cell (Scheme 1).To confirm our hypothesis that the hydroquinone was the more

potent Hsp90 inhibitor, we proceeded with two lines of experi-ments. In the first, we used purified yeast and human Hsp90 andemployed established methods to assess the ATPase activity ofthese purified proteins. We showed that in studies with both yeastand human Hsp90 the generation of 17-AAGH2 by NQO1 wasrequired for maximal ATPase inhibition. The inhibition of ATPaseactivity could be abrogated by use of ES936, the mechanism-basedinhibitor of NQO1, confirming the crucial role of 17-AAGH2 inHsp90 inhibition.The second line of experimentation we pursued was computer

modeling of the NH2-terminal domain of Hsp90 containing theATPase active site. Although the complete structure of the intactHsp90 protein has not yet been determined, crystal structures ofthe NH2-terminal domain identified by limited proteolysis havebeen described for yeast (25) and human (26) Hsp90. Thetertiary structures of these amino acid terminal domains are verysimilar, containing a h-sheet covered on one face by a-helices,with 69% identity yeast to human, consistent with the highhomology among all Hsp90 sequences. The helical face of theHsp90 NH2-terminal domain has a wide opening at its centerthat extends into the hydrophobic core of the structure andresults in a pronounced pocket, the binding site for the benzo-quinone ansamycin antitumor agents, geldanamycin and 17-DMAG (25, 26, 29).The 17-AAG and 17-AAGH2 structures consist of a 17-

(2-propenylamino)-substituted planar benzoquinone or hydroqui-

none, embedded in a closed ansa ring system (Fig. 6). The ansaring is folded under the benzoquinone/hydroquinone, forming aC-shaped conformation with the carbamate group at the base ofthe binding site and the benzoquinone/hydroquinone ring atthe solvent exposed pocket entrance. Multiple protein-ligandinteractions lock the macrocycle of 17-AAG and 17-AAGH2 in anoverall conformation similar to that of geldanamycin and 17-DMAG(25, 26, 29).The greater interaction energies of 17-AAGH2 is explained by the

stronger and greater number of hydrogen-bonding interactionswith key amino acid residues in the ATP-binding domain of Hsp90.As a result, 17-AAGH2 adopts a more compact C-shaped, clamp-like conformation around helix 2 due to the hydrogen bondinginteractions of the C18 hydroxyl group, of the hydroquinonemoiety, with Asp40 of yeast Hsp90 and Asp54 of human Hsp90,thereby increasing the electrostatic contribution to the nonbondedinteraction energy.It is clear that NQO1-mediated generation of 17-AAGH2 from

17-AAG results in increased Hsp90 inhibition as indicated byeffects on client proteins and growth inhibition and that NQO1 isa critical determinant of maintaining the benzoquinone ansamy-cin in its more potent hydroquinone form in tumors. Dependenton their structure and redox potential, however, hydroquinonesexhibit varying stability toward oxygen. The propensity forhydroquinones to undergo autooxidation varies and depends onoxygen tension, redox potential, and availability of redox activemetals, particularly iron and copper. Very little is known aboutthe stability of 17-AAGH2 and the hydroquinone forms of otheransamycins. Previous work focusing on geldanamycin reportedthe preparation of the geldanamycin hydroquinone by sodiumdithionite reduction, which was stable enough to be isolated andpurified but reverted to the hydroquinone slowly in the presenceof air (7). Our data show that the hydroquinone derivative of17-AAG is relatively stable and in agreement with the previousreport (7); we found that the hydroquinone slowly reoxidizes backto the parent quinone on exposure to air over a period of hours.This suggests that the pharmacokinetic and pharmacodynamicstudies of the effects of benzoquinone ansamycins need toconsider the quinone and hydroquinone as separate entities,particularly as they possess very different abilities to inhibitHsp90.Our work on the formation of 17-AAGH2 by NQO1 was presented

in abstract form (30), and at the same meeting, data were shownthat proposed the use of the hydroquinone of 17-AAG as a water-soluble prodrug of 17-AAG (31). The development of 17-AAGH2 as atherapeutic entity makes it even more important to study thebiological properties of the benzohydroquinone ansamycins, theirstability and their generation in human tumors.In summary, our data show that 17-AAGH2 can be generated

selectively in tumor cells containing high levels of NQO1 and thatthe hydroquinone form, based on target inhibition in both cell-freeand cellular systems and molecular modeling studies, represents amore potent inhibitor of Hsp90.

Acknowledgments

Received 6/14/2005; revised 7/26/2005; accepted 8/26/2005.Grant support: NIH grant CA51210 (W. Guo, P. Reigan, D. Siegel, and D. Ross) and

University of Colorado Cancer Center support grant (J. Zirrolli and D. Gustafson).The costs of publication of this article were defrayed in part by the payment of page

charges. This article must therefore be hereby marked advertisement in accordancewith 18 U.S.C. Section 1734 solely to indicate this fact.

Scheme 1. Proposed model of inhibition of cellular Hsp90 by 17-AAG and17-AAGH2. Major pathways are shown as solid lines ; minor pathways are shownas dotted lines .

Cancer Research

Cancer Res 2005; 65: (21). November 1, 2005 10014 www.aacrjournals.org

Research. on January 25, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 10: Formation of 17-Allylamino-Demethoxygeldanamycin (17-AAG ...Hydroquinone by NAD(P)H:Quinone Oxidoreductase 1: Role of 17-AAG Hydroquinone in Heat Shock Protein 90 Inhibition Wenchang

References1. Neckers L. Hsp90 inhibitors as novel cancer chemo-therapeutic agents. Trends Mol Med 2002;8:55–61.

2. Bagatell R, Whitesell L. Altered Hsp90 function incancer: a unique therapeutic opportunity. Mol CancerTher 2004;3:1021–30.

3. Workman P. Altered states: selectively drugging theHsp90 cancer chaperone. Trends Mol Med 2004;10:47–51.

4. Beliakoff J, Whitesell L. Hsp90: an emerging target forbreast cancer therapy. Anticancer Drugs 2004;15:651–62.

5. Hollingshead M, Alley M, Burger AM, et al. In vivoantitumor efficacy of 17-DMAG (17-dimethylaminoethy-lamino-17-demethoxygeldanamycin hydrochloride), awater-soluble geldanamycin derivative. Cancer Chemo-ther Pharmacol 2005;56:115–25.

6. Eiseman JL, Lan J, Lagattuta TF, et al. Pharmacoki-netics and pharmacodynamics of 17-demethoxy 17-[[(2-dimethylamino)ethyl]amino]geldanamycin (17DMAG,NSC 707545) in C.B-17 SCID mice bearing MDA-MB-231 human breast cancer xenografts. Cancer ChemotherPharmacol 2005;55:21–32.

7. Schnur RC, Corman ML, Gallaschun RJ, et al.Inhibition of the oncogene product p185erbB-2 in vitroand in vivo by geldanamycin and dihydrogeldanamycinderivatives. J Med Chem 1995;38:3806–12.

8. Ernster L. DT-diaphorase. Methods Enzymol 1967;10:309–17.

9. Siegel D, Franklin WA, Ross D. Immunohistochemicaldetection of NAD(P)H:quinone oxidoreductase in hu-man lung and lung tumors. Clin Cancer Res 1998;4:2065–70.

10. Siegel D, Ross D. Immunodetection of NAD(P)H:qui-none oxidoreductase 1 (NQO1) in human tissues. FreeRadic Biol Med 2000;29:246–53.

11. Cullen JJ, Hinkhouse MM, Grady M, et al. Dicumarolinhibition of NADPH:quinone oxidoreductase inducesgrowth inhibition of pancreatic cancer via a superoxidemediated mechanism. Cancer Res 2003;63:5513–20.

12. Siegel D, Gibson NW, Preusch PC, Ross D. Metabo-lism of diaziquone by NAD(P)H:(quinone acceptor)oxidoreductase (DT-diaphorase): role in diaziquone-

induced DNA damage and cytotoxicity in human coloncarcinoma cells. Cancer Res 1990;50:7293–300.

13. Siegel D, Gibson NW, Preusch PC, Ross D.Metabolism of mitomycin C by DT-diaphorase: rolein mitomycin C-induced DNA damage and cytotoxicityin human colon carcinoma cells. Cancer Res 1990;50:7483–9.

14. Walton MI, Smith PJ, Workman P. The role ofNAD(P)H:quinone reductase (EC 1.6.99.2, DT-diapho-rase) in the reductive bioactivation of the novelindoloquinone antitumor agent EO9. Cancer Commun1991;3:199–206.

15. Beall HD, Liu Y, Siegel D, Bolton EM, Gibson NW,Ross D. Role of NAD(P)H:quinone oxidoreductase (DT-diaphorase) in cytotoxicity and induction of DNAdamage by streptonigrin. Biochem Pharmacol 1996;51:645–52.

16. Winski SL, Hargreaves RH, Butler J, Ross D. A newscreening system for NAD(P)H:quinone oxidoreductase(NQO1)-directed antitumor quinones: identification of anew aziridinylbenzoquinone, RH1, as a NQO1-directedantitumor agent. Clin Cancer Res 1998;4:3083–8.

17. Pink JJ, Planchon SM, Tagliarino C, Varnes ME, SiegelD, Boothman DA. NAD(P)H:quinone oxidoreductaseactivity is the principal determinant of h-lapachonecytotoxicity. J Biol Chem 2000;275:5416–24.

18. Kelland LR, Sharp SY, Rogers PM, Myers TG,Workman P. DT-diaphorase expression and tumor cellsensitivity to 17-allylamino, 17-demethoxygeldanamy-cin, an inhibitor of heat shock protein 90. J Natl CancerInst 1999;91:1940–9.

19. Beall HD, Mulcahy RT, Siegel D, Traver RD, GibsonNW, Ross D. Metabolism of bioreductive antitumorcompounds by purified rat and human DT-diaphorases.Cancer Res 1994;54:3196–201.

20. Dehn DL, Winski SL, Ross D. Development of anew isogenic cell-xenograft system for evaluation ofNAD(P)H:quinone oxidoreductase-directed antitumorquinones: evaluation of the activity of RH1. Clin CancerRes 2004;10:3147–55.

21. Lowry OH, Rosebrough NJ, Farr AL, Randall RJ.Protein measurement with the Folin phenol reagent.J Biol Chem 1951;193:265–75.

22. Winski SL, Faig M, Bianchet MA, et al. Characteriza-tion of a mechanism-based inhibitor of NAD(P)H:qui-none oxidoreductase 1 by biochemical, X-raycrystallographic, and mass spectrometric approaches.Biochemistry 2001;40:15135–42.

23. Winski SL, Swann E, Hargreaves RH, et al. Relation-ship between NAD(P)H:quinone oxidoreductase 1(NQO1) levels in a series of stably transfected cell linesand susceptibility to antitumor quinones. BiochemPharmacol 2001;61:1509–16.

24. Rowlands MG, Newbatt YM, Prodromou C, Pearl LH,Workman P, Aherne W. High-throughput screeningassay for inhibitors of heat-shock protein 90 ATPaseactivity. Anal Biochem 2004;327:176–83.

25. Prodromou C, Roe SM, O’Brien R, Ladbury JE, PiperPW, Pearl LH. Identification and structural character-ization of the ATP/ADP-binding site in the Hsp90molecular chaperone. Cell 1997;90:65–75.

26. Stebbins CE, Russo AA, Schneider C, Rosen N, HartlFU, Pavletich NP. Crystal structure of an Hsp90-geldanamycin complex: targeting of a protein chaperoneby an antitumor agent. Cell 1997;89:239–50.

27. Siegel D, Anwar A, Winski SL, Kepa JK, ZolmanKL, Ross D. Rapid polyubiquitination and protea-somal degradation of a mutant form of NAD(P)H:-quinone oxidoreductase 1. Mol Pharmacol 2001;59:263–8.

28. Workman P. Auditing the pharmacological accountsfor Hsp90 molecular chaperone inhibitors: unfolding therelationship between pharmacokinetics and pharmaco-dynamics. Mol Cancer Ther 2003;2:131–8.

29. Jez JM, Chen JC, Rastelli G, Stroud RM, Santi DV.Crystal structure and molecular modeling of 17-DMAGin complex with human Hsp90. Chem Biol 2003;10:361–8.

30. Guo W, Siegel D, Reigan P, Zirrolli J, Gustafson D,Ross D. A potential role for NAD(P)H:quinone oxidore-ductase 1 (NQO1) in the mechanism of action of theHSP90 inhibitors geldanamycin and 17AAG. Proc AACR2005;46:4936.

31. Sydor JR, Pien CS, Zhang Y, et al. Anti-tumor activityof a novel, water soluble Hsp90 inhibitor IPI-504 inmultiple myeloma. Proc AACR 2005;46:6160.

Metabolism of 17-AAG by NQO1

www.aacrjournals.org 10015 Cancer Res 2005; 65: (21). November 1, 2005

Research. on January 25, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 11: Formation of 17-Allylamino-Demethoxygeldanamycin (17-AAG ...Hydroquinone by NAD(P)H:Quinone Oxidoreductase 1: Role of 17-AAG Hydroquinone in Heat Shock Protein 90 Inhibition Wenchang

2005;65:10006-10015. Cancer Res   Wenchang Guo, Philip Reigan, David Siegel, et al.   Shock Protein 90 InhibitionOxidoreductase 1: Role of 17-AAG Hydroquinone in Heat(17-AAG) Hydroquinone by NAD(P)H:Quinone Formation of 17-Allylamino-Demethoxygeldanamycin

  Updated version

  http://cancerres.aacrjournals.org/content/65/21/10006

Access the most recent version of this article at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/65/21/10006.full#ref-list-1

This article cites 31 articles, 12 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/65/21/10006.full#related-urls

This article has been cited by 32 HighWire-hosted articles. Access the articles at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected] at

To order reprints of this article or to subscribe to the journal, contact the AACR Publications

  Permissions

  Rightslink site. (CCC)Click on "Request Permissions" which will take you to the Copyright Clearance Center's

.http://cancerres.aacrjournals.org/content/65/21/10006To request permission to re-use all or part of this article, use this link

Research. on January 25, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from