activity of the hypoxia-activated prodrug, th-302, in ... · th-302 was also effective in advanced...

15
Cancer Therapy: Preclinical Activity of the Hypoxia-Activated Prodrug, TH-302, in Preclinical Human Acute Myeloid Leukemia Models Scott Portwood 1 , Deepika Lal 1 , Yung-Chun Hsu 1 , Rodrigo Vargas 2 , Megan K. Johnson 1 , Meir Wetzler 1 , Charles P. Hart 3 , and Eunice S. Wang 1,2 Abstract Purpose: Acute myeloid leukemia (AML) is an aggressive hematologic neoplasm. Recent evidence has shown the bone marrow microenvironment in patients with AML to be intrinsically hypoxic. Adaptive cellular responses by leukemia cells to survive under low oxygenation also confer chemoresistance. We therefore asked whether therapeutic exploitation of marrow hypoxia via the hypoxia-activated nitrogen mustard prodrug, TH-302, could effectively inhibit AML growth. Experimental Design: We assessed the effects of hypoxia and TH-302 on human AML cells, primary samples, and systemic xenograft models. Results: We observed that human AML cells and primary AML colonies cultured under chronic hypoxia (1% O 2 , 72 hours) exhibited reduced sensitivity to cytarabine-induced apoptosis as compared with normoxic controls. TH-302 treatment resulted in dose- and hypoxia-dependent apoptosis and cell death in diverse AML cells. TH-302 preferentially decreased proliferation, reduced HIF-1a expression, induced cell-cycle arrest, and enhanced double-stranded DNA breaks in hypoxic AML cells. Hypoxia-induced reactive oxygen species by AML cells were also diminished. In systemic human AML xenografts (HEL, HL60), TH-302 [50 mg/kg intraperitoneally (i.p.) 5 times per week] inhibited disease progression and prolonged overall survival. TH-302 treatment reduced the number of hypoxic cells within leukemic bone marrows and was not associated with hematologic toxicities in nonleukemic or leukemic mice. Later initiation of TH-302 treatment in advanced AML disease was as effective as earlier TH-302 treatment in xenograft models. Conclusions: Our results establish the preclinical activity of TH-302 in AML and provide the rationale for further clinical studies of this and other hypoxia-activated agents for leukemia therapy. Clin Cancer Res; 19(23); 6506–19. Ó2013 AACR. Introduction Acute myeloid leukemia (AML) is an aggressive immature hematologic neoplasm primarily affecting older adults. Chemotherapy for AML has not changed since the 1980s and continues to result in long-term cure in only 20% to 30% of patients. Recent data have shown that the bone marrow microenvironment, the principal site of AML initiation and expansion, is characterized by intrinsically low oxygen tension. This relative hypoxia within the bone marrow functions as a key mediator of normal hematopoi- esis, specifically hematopoietic stem cell biology (1–4). In fact, most primitive pluripotent hematopoietic stem cells (HSC) are preferentially located in distinct nonperfused specialized marrow niches characterized by the most severe hypoxia (5–7). The cytokine-rich milieu of these poorly oxygenated marrow niches has been shown to promote long-term cellular quiescence and self-renewal of HSCs and decrease levels of potentially toxic reactive oxygen species (ROS; refs. 8, 9). The hypoxic bone marrow microenvironment has also been shown to contribute to AML disease. Prior data in multiple animal models showed that acute leukemic pro- gression within the bone marrow is associated with further decreases in overall marrow oxygenation and expansion of hypoxic bone marrow areas (10–12). This has been attrib- uted, in part, to potentially compromised blood flow within a restricted noncompliant bony cavity (13, 14). Poorly per- fused hypoxic marrow niches could also affect the systemic exposure of AML cells to systemic chemotherapy agents as well as the circulation and recruitment of host immune effector cells mediating anti-tumor responses. Theoretically, such bone marrow microenvironmental changes may lead to the selective outgrowth of AML clones which are "better adapted" to survive within a severely hypoxic microenvi- ronment than normal hematopoietic cells (15–17). Pro- longed culturing of AML cells under hypoxia has been Authors' Afliations: 1 Departments of Medicine, 2 Immunology, Roswell Park Cancer Institute, Buffalo, New York; and 3 Threshold Pharmaceuticals Inc, South San Francisco, California Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). Corresponding Author: Eunice S. Wang, Department of Medicine, Ros- well Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263. Phone: 716-845-3544; Fax: 716-845-8741; E-mail: [email protected] doi: 10.1158/1078-0432.CCR-13-0674 Ó2013 American Association for Cancer Research. Clinical Cancer Research Clin Cancer Res; 19(23) December 1, 2013 6506 on August 21, 2020. © 2013 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from Published OnlineFirst October 2, 2013; DOI: 10.1158/1078-0432.CCR-13-0674

Upload: others

Post on 12-Jul-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Activity of the Hypoxia-Activated Prodrug, TH-302, in ... · TH-302 was also effective in advanced AML disease, the setting in which marrow hypoxia would be expected to be greatest

Cancer Therapy: Preclinical

Activity of the Hypoxia-Activated Prodrug, TH-302, inPreclinical Human Acute Myeloid Leukemia Models

Scott Portwood1, Deepika Lal1, Yung-Chun Hsu1, Rodrigo Vargas2, Megan K. Johnson1, Meir Wetzler1,Charles P. Hart3, and Eunice S. Wang1,2

AbstractPurpose: Acute myeloid leukemia (AML) is an aggressive hematologic neoplasm. Recent evidence has

shown the bone marrow microenvironment in patients with AML to be intrinsically hypoxic. Adaptive

cellular responses by leukemia cells to survive under low oxygenation also confer chemoresistance. We

therefore asked whether therapeutic exploitation of marrow hypoxia via the hypoxia-activated nitrogen

mustard prodrug, TH-302, could effectively inhibit AML growth.

Experimental Design: We assessed the effects of hypoxia and TH-302 on human AML cells, primary

samples, and systemic xenograft models.

Results:We observed that human AML cells and primary AML colonies cultured under chronic hypoxia

(1% O2, 72 hours) exhibited reduced sensitivity to cytarabine-induced apoptosis as compared with

normoxic controls. TH-302 treatment resulted in dose- and hypoxia-dependent apoptosis and cell death

in diverse AML cells. TH-302 preferentially decreased proliferation, reduced HIF-1a expression, induced

cell-cycle arrest, and enhanceddouble-strandedDNAbreaks inhypoxicAMLcells.Hypoxia-induced reactive

oxygen species byAMLcellswere alsodiminished. In systemic humanAMLxenografts (HEL,HL60), TH-302

[50 mg/kg intraperitoneally (i.p.) 5 times per week] inhibited disease progression and prolonged overall

survival. TH-302 treatment reduced thenumber of hypoxic cells within leukemic bonemarrows andwas not

associated with hematologic toxicities in nonleukemic or leukemic mice. Later initiation of TH-302

treatment in advanced AML disease was as effective as earlier TH-302 treatment in xenograft models.

Conclusions:Our results establish the preclinical activity of TH-302 in AML and provide the rationale

for further clinical studies of this and other hypoxia-activated agents for leukemia therapy. Clin Cancer

Res; 19(23); 6506–19. �2013 AACR.

IntroductionAcutemyeloid leukemia (AML) is an aggressive immature

hematologic neoplasm primarily affecting older adults.Chemotherapy for AML has not changed since the 1980sand continues to result in long-term cure in only 20% to30% of patients. Recent data have shown that the bonemarrow microenvironment, the principal site of AMLinitiation and expansion, is characterized by intrinsicallylow oxygen tension. This relative hypoxia within the bonemarrow functions as a key mediator of normal hematopoi-esis, specifically hematopoietic stem cell biology (1–4). Infact, most primitive pluripotent hematopoietic stem cells

(HSC) are preferentially located in distinct nonperfusedspecialized marrow niches characterized by the most severehypoxia (5–7). The cytokine-rich milieu of these poorlyoxygenated marrow niches has been shown to promotelong-term cellular quiescence and self-renewal of HSCs anddecrease levels of potentially toxic reactive oxygen species(ROS; refs. 8, 9).

The hypoxic bone marrow microenvironment has alsobeen shown to contribute to AML disease. Prior data inmultiple animal models showed that acute leukemic pro-gression within the bone marrow is associated with furtherdecreases in overall marrow oxygenation and expansion ofhypoxic bone marrow areas (10–12). This has been attrib-uted, in part, to potentially compromised bloodflowwithina restricted noncompliant bony cavity (13, 14). Poorly per-fused hypoxic marrow niches could also affect the systemicexposure of AML cells to systemic chemotherapy agents aswell as the circulation and recruitment of host immuneeffector cellsmediating anti-tumor responses. Theoretically,such bone marrow microenvironmental changes may leadto the selective outgrowth of AML clones which are "betteradapted" to survive within a severely hypoxic microenvi-ronment than normal hematopoietic cells (15–17). Pro-longed culturing of AML cells under hypoxia has been

Authors' Affiliations: 1Departments of Medicine, 2Immunology, RoswellPark Cancer Institute, Buffalo, New York; and 3Threshold PharmaceuticalsInc, South San Francisco, California

Note: Supplementary data for this article are available at Clinical CancerResearch Online (http://clincancerres.aacrjournals.org/).

Corresponding Author: Eunice S. Wang, Department of Medicine, Ros-well Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263.Phone: 716-845-3544; Fax: 716-845-8741; E-mail:[email protected]

doi: 10.1158/1078-0432.CCR-13-0674

�2013 American Association for Cancer Research.

ClinicalCancer

Research

Clin Cancer Res; 19(23) December 1, 20136506

on August 21, 2020. © 2013 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 2, 2013; DOI: 10.1158/1078-0432.CCR-13-0674

Page 2: Activity of the Hypoxia-Activated Prodrug, TH-302, in ... · TH-302 was also effective in advanced AML disease, the setting in which marrow hypoxia would be expected to be greatest

shown to favor quiescent immature stem cell–like clonesas well as cells with higher expression of signaling pathwayspromoting leukemia survival and expansion (3, 9, 18, 19).Similar to solid tumor cells, it now appears that molecularand biologic adaptations by AML cells to hypoxia alsoconfer resistance to chemotherapy. We and other investi-gators have reported that overexpression of hypoxic-induc-ible factor-1a (HIF-1a) in primary AML and acute lympho-cytic leukemia (ALL) marrow samples correlated with poorchemotherapy outcomes (20, 21). Hypoxia-induced upre-gulation of HIF-1a in human acute leukemia cells in vitrohas subsequently been shown to induce cell dormancy andresistance to chemotherapy (22, 23). For these reasons,hypoxia-activated agents, novel cytotoxic drugs preferen-tially activated under low oxygenation conditions, mayhave an active role in the treatment of AML and other bonemarrow malignancies.TH-302 is a 2-nitroimidazole hypoxia-activated prodrug

of the cytotoxin bromo-isophosphoramide mustard. TH-302 has been shown to exhibit broad-spectrum preclinicalactivity against solid tumor and multiple myeloma cellsunder low oxygenation (24–26). Clinical trials of TH-302alone and in combination with chemotherapy in patientswith solid and hematologic cancers have shown limitedtoxicities and evidence of clinical efficacy (27–29). Here, weinvestigated the preclinical activity of TH-302 in humanAML cells, primary samples, and xenograft models.

MethodsCell lines, patient samples, and reagentsHuman AML cell lines (HEL, HL60, Kasumi) were pur-

chased from the American Tissue Culture Collection. ML-2

cells containing MLL/11q23 were obtained from DSMZ(30). NB-4, MV4;11, LAMA, KG-1, and THP-1 were origi-nally obtained from DSMZ and were kindly provided byDr. C. Frangou, Department of Cancer Genetics, RPCI.Unless otherwise indicated, cells were grown in humidifiedincubators (37�C, 21% O2, 5% CO2) in RPMI-1640 medi-um containing 10% FBS, 2 mmol/L L-glutamine, 4.5 g/Lglucose, 1.5 g sodium bicarbonate, 1 u/mL penicillin/strep-tomycin, 10 mmol/L HEPES, and 1 nmol/L sodium pyru-vate. TH-302 was provided by Threshold Pharmaceuticals,Inc. Cytarabine, doxorubicin, and the NADPH oxidaseinhibitor, diphenyleneiodonium (DPI), were purchasedfrom Sigma-Aldrich. Concentration and duration of treat-ments are shown in individual figures and detailed in thefigure legends.

Hypoxic conditionsCells were plated in 6-well plates in an X-vivo hypoxia

chamber (BioSpherix) at 1%O2/5%CO2 level for indicatedtime points before harvesting for further analysis.

In vitro assaysViable cells were counted using a hemocytometer in

triplicate following dye exclusion (1:1 dilution in 0.4%trypan blue solution; Mediatech Inc.). Results were aver-aged among triplicate wells.

Flow cytometryLeukemia cells were plated in triplicate in 6-well plates at

a concentration of 5 � 105 cells/mL in the presence ofvehicle [dimethyl sulfoxide (DMSO)] or varying concentra-tions of TH-302. Cells were then incubated for 24 to 72hours under normoxic (20% O2) or hypoxic (1% O2)conditions followed by flow cytometric assays. Apoptosiswas assessed in asynchronously growing cells via anAnnexin V-PE Apoptosis Detection kit (BD Pharmingen).Double-stranded DNA breaks were assessed via measure-ments of phosphorylated histone H2A.X (EMD Millipore)and with the APO-BrdU TUNEL Assay Kit (MolecularProbes, Life Technologies) following manufacturer’sinstructions. Cell-cycle analysis was conducted on cellstreated for 24 hours and then washed, fixed with 70% ethylalcohol for 30minutes at 4�C, and centrifuged at 3,200 rpmfor 3 minutes. Cells were then treated with RNAse A treat-ment (0.5mg/mL) for another 30minutes at 37�C followedby the addition of propidium iodide (1 mg/mL; GenScript)for an additional 30minutes at 4�C in the dark followed byanalysis. To detect ROS in live cells, the fluorescein-baseddye CM-H2DCFDA (Invitrogen) was added to samples at afinal concentration of 10 mmol/L followed by incubation inthe dark with agitation at 37�C for 30 minutes. Sampleviability was assessed by 7-AAD (BD Pharmingen) andconfirmedby trypanblue exclusion under lightmicroscopy.All studies were conducted on a FACSCalibur instrument(BD) followed by analysis using Winlist Version 6.0 (VeritySoftware House) and FCS Express software (De Novo Soft-ware). Experiments were carried out independently at leasttwice.

Translational RelevanceAcutemyeloid leukemia (AML) is an aggressive hema-

tologic neoplasm affecting older adults. The primary siteof disease initiation and expansion in patients with AMLis the bone marrow. At baseline, the marrow microen-vironment is known to be intrinsically hypoxic. Oneconsequence of rapid leukemia growth is furthermarrowhypoxia. We show that the hypoxia-activated prodrug,TH-302, exerts significant anti-leukemic activity. TH-302overcomes chemoresistance conferred by hypoxia incultured human AML cells and primary AML samples.TH-302 induces hypoxic-dependent DNA damage, cell-cycle arrest, decreased reactive oxygen production, andcell death. In systemic human leukemia xenografts, TH-302 reduces disease burden and prolongs overall surviv-al without overt hematologic toxicity. Treatment withTH-302 was also effective in advanced AML disease, thesetting in which marrow hypoxia would be expected tobe greatest. These results support further clinical devel-opment of TH-302 as a novel approach for acute leuke-mia therapy.

TH-302 in Acute Myeloid Leukemia

www.aacrjournals.org Clin Cancer Res; 19(23) December 1, 2013 6507

on August 21, 2020. © 2013 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 2, 2013; DOI: 10.1158/1078-0432.CCR-13-0674

Page 3: Activity of the Hypoxia-Activated Prodrug, TH-302, in ... · TH-302 was also effective in advanced AML disease, the setting in which marrow hypoxia would be expected to be greatest

Protein analysesCells (1 � 106/mL) were plated in 6-well plates for drug

and/or hypoxic treatment followed by lysis in radioimmu-noprecipitation assay (RIPA) buffer [50 mmol/L Tris-HCl,pH 7.4, 150 mmol/L NaCl, 1 mmol/L EDTA, 1% Triton X-100, 1% (w/v) sodium deoxycholate, 0.1% (w/v) SDS, 1%(v/v)], supplemented with phosphatase inhibitor cocktail 1and 2 (Sigma-Aldrich), and 1% (v/v) protease inhibitorcocktail 1 and2 (Sigma-Aldrich). Protein concentrationwas

determined by Bio-Rad protein assay (Bio-Rad). Lysateswere separated by SDS-PAGE (12% for cleaved caspase-3,8% for the rest) and detected by chemiluminescence (ECLplus, GE Healthcare). Specific antibodies included anti-cleaved caspase-3 (#9661, Cell Signaling Technology) andanti-cleaved PARP (#9546, Cell Signaling Technology) usedat a final dilution ratio of 1:1,000. Antibody to actin(#CP01) was obtained from Calbiochem. For HIF-1aimmunoassays, protein samples were analyzed following

Figure 1. TH-302 preferentiallyinhibits in vitro growth of AML cellsunder hypoxia. A, humanAMLcells(HL60, HEL) were cultured in thepresence or absence of cytarabine100 nmol/L under normoxic (21%O2) or hypoxic (1% O2) conditionsfor up to 72 hours. Mean number ofapoptotic cells (� SEM) fromtriplicate samples under varyingconditions as assessed by 7-AAD/Annexin-V flow cytometry areshown. B, cytarabine-mediatedapoptosis under hypoxic versusnormoxic conditions was alsomeasured byWestern blot analysisfor cleaved PARP in AML (HL60)cells. C and D, TH-302 treatmentresulted in cell death of AML cells(HL60, HEL) in a dose-dependentand hypoxia-specific manner.Shown are the mean � SEMpercentages of dead cells asdetermined by flow cytometry for7-AAD/Annexin-V following 72hours of incubation with vehicle orTH-302 at doses ranging from 0.1to 5 mmol/L under normoxia (21%O2) or hypoxia (1%O2). (Continuedon the following page.)

Portwood et al.

Clin Cancer Res; 19(23) December 1, 2013 Clinical Cancer Research6508

on August 21, 2020. © 2013 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 2, 2013; DOI: 10.1158/1078-0432.CCR-13-0674

Page 4: Activity of the Hypoxia-Activated Prodrug, TH-302, in ... · TH-302 was also effective in advanced AML disease, the setting in which marrow hypoxia would be expected to be greatest

the manufacturer’s instructions (R&D Systems). Cells trea-ted with 100 mmol/L cobalt chloride (Sigma Aldrich) for 4hours served a positive control.

Primary AML clonogenic assaysPrimary AML patient cells (2 � 105) were plated in

triplicate in methylcellulose complete media containingrhSCF 50 ng/mL, rhGM-CSF 10 ng/mL, rhIL-3 10 ng/mL,and rhEpo 3U/mL (HSC003, R&DSystems) and cultured at37�C (21% O2, 5% CO2) for 14 days to allow for baselinegrowth. Following this, vehicle (PBS), cytarabine (0.01nmol/L), doxorubicin (0.5 nmol/L), or TH-302 (0.1–1nmol/L) was added. Plates were cultured for an additional72 hours under hypoxia or normoxia to determine theconcomitant effects of environmental and pharmacologic

interventions on established primary AML growth. Mean(�SEM) leukemia colony formation units (CFU-L) werequantified using an inverted microscope.

Leukemia xenograft experimentsAll animal experiments were carried out under an

approved IACUC protocol. Six- to 8-week-old female severecombined immunodeficient (SCID) mice (RPCI, Depart-ment of Laboratory Animal Research, Buffalo, NY) weresublethally irradiated (200 rad) followed by tail vein (1 �107 cells) injection of human HEL and HL60 cells stablytransfected with a pGL4 luciferase reporter vector (Pro-mega) as described previously (31). Mice were evaluatedweekly for evidence of leukemia engraftment and overallleukemia disease progression by serial bioluminescent

Figure 1. (Continued. ) E, primaryAML cells (2 � 105) from 3 patientswith AML were grown inmethylcellulose culture for 10 to14 days followed by treatmentwith vehicle (PBS), cytarabine(Ara-C, 0.01 nmol/L), or TH-302(0.5 mmol/L) for 72 hours underhypoxia or normoxia. Shown is themean (�SEM) number of leukemiacolony formation units (CFU) undereach condition as determined intriplicate plates by invertedmicroscopy. For all results, whitebars indicate normoxia; gray barsindicate hypoxia. All experimentswere carried out 3 separate timeswith triplicate samples percondition.P<0.05by t test analysiswas considered significant anddenoted by an asterisk.

TH-302 in Acute Myeloid Leukemia

www.aacrjournals.org Clin Cancer Res; 19(23) December 1, 2013 6509

on August 21, 2020. © 2013 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 2, 2013; DOI: 10.1158/1078-0432.CCR-13-0674

Page 5: Activity of the Hypoxia-Activated Prodrug, TH-302, in ... · TH-302 was also effective in advanced AML disease, the setting in which marrow hypoxia would be expected to be greatest

A

B

HL60 HEL

***

***

G1 G1 G1

G2 G2 G2

S S S

G1 G1 G1

G2 G2 G2

S S S

Cell

num

ber

Cell

num

ber

DNA content (PI) DNA content (PI) DNA content (PI)

DNA content (PI) DNA content (PI) DNA content (PI)

C H2AX phosphorylation D DNA fragmentation by TUNEL

Figure 2. TH-302 induceshypoxia-dependentcell-cycle arrest andDNAdamage inAMLcells.A, low-doseTH-302 treatment (0.1–1nmol/L) preferentially inhibitedthe proliferation of AML (HL60, HEL) cells under hypoxic conditions as determined by tritiated thymidine incorporation in triplicate wells. B, AML cells weretreatedwith vehicle (PBS) or TH-302 at the indicated concentrations for 24 hours under hypoxic (1%O2) or normoxic (21%O2) conditions in triplicatewells. Cellswere stained with propidium iodide staining and analyzed by flow cytometry. Representative cell-cycle effects and flow histograms are shown. Similarresults were obtained in 3 independent experiments. C, HEL cells were plated in triplicate wells and treated with vehicle or TH-302 for 24 hours under hypoxia(1% O2) or normoxia (21% O2). Cells were then harvested and stained for H2A.X phosphorylation followed by flow cytometry. Shown are the mean (�SEM)percentagesofH2A.X-positivecells undereachcondition.Representative resultsof2 independentexperimentsareshown.D,similarly treatedHELcellswerealsoassessed for expression of DNA fragmentation of apoptotic cells by TUNEL assay using flow cytometry. (Continued on the following page.)

Portwood et al.

Clin Cancer Res; 19(23) December 1, 2013 Clinical Cancer Research6510

on August 21, 2020. © 2013 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 2, 2013; DOI: 10.1158/1078-0432.CCR-13-0674

Page 6: Activity of the Hypoxia-Activated Prodrug, TH-302, in ... · TH-302 was also effective in advanced AML disease, the setting in which marrow hypoxia would be expected to be greatest

imaging (BLI; Xenogen IVIS 50 System,Caliper Life Science)following intraperitoneal D-luciferin injection (75 mg/kg).Dorsal and ventral images were acquired from each animal.Data were expressed as the average photon emission ¼photons per second per cm2 per steradian calculated fromdorsal and ventral images of each animal (Living ImagingSoftware, Calipers Life Sciences). Peripheral blood wascollected intermittently for complete blood cell measure-ments. For marrow hypoxia determinations, sublethally

irradiated SCID mice were injected in the tail vein withHEL-luciferase AML cells and serially followed for diseaseprogression by BLI at intervals after tumor inoculation.Twenty days after inoculation, cohorts of leukemia-engrafted animals (n ¼ 5–10 mice/group) were treatedwith vehicle (PBS), cytarabine 2 mg/kg intraperitoneally(i.p.) daily for 3 days, doxorubicin 35 mg/kg i.p. singledose, or aflibercept (VEGF Trap) 5 mg/kg single injection(31). Three days later, mice were injected i.p. with

EHIF-1α protein expression

F ROS generation

Normoxia Hypoxia

Vehicle

TH-302

0.05 µmol/L

TH-302

0.5 µmol/L

TH-302

5 µmol/L

G

Fluorescent intensity Fluorescent intensity

Cel

l co

un

tsC

ell c

ou

nts

Figure 2. (Continued. ) E, hypoxic AML cells treated with TH-302 (1–10 mmol/L) for 24 hours also expressed significantly less HIF-1a protein as determined byimmunoassay. Cobalt chloride–treated cells and normoxic AML cells cultured under the same conditions served as controls. F and G, production ofROSbyHL60AML cells following 48 hours of hypoxia (1%O2) were significantly abrogated by TH-302 treatment in a dose-dependentmanner, as determinedby flow cytometry for mean fluorescent intensity (CM-H2DCFDA staining). ROS levels in cells incubated under normoxia were unaffected by TH-302treatment. Analyses were conducted on gated viable cells only in triplicate wells. Each histogram records the fluorescent intensity of gated viable cells intriplicate. Outliers are observed primarily at higher concentrations of TH-302 treatment. Similar results were seen for HEL cells. At least 2 independentexperiments were carried out for all of the above experiments. White bars indicate normoxia, gray bars indicate hypoxia. Student t-test analysis comparedconditions versus controls. �, P < 0.05; ��, P < 0.001; ���, P � 0.0001.

TH-302 in Acute Myeloid Leukemia

www.aacrjournals.org Clin Cancer Res; 19(23) December 1, 2013 6511

on August 21, 2020. © 2013 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 2, 2013; DOI: 10.1158/1078-0432.CCR-13-0674

Page 7: Activity of the Hypoxia-Activated Prodrug, TH-302, in ... · TH-302 was also effective in advanced AML disease, the setting in which marrow hypoxia would be expected to be greatest

pimonidazole 60 mg/kg (Hypoxyprobe Store) and sacri-ficed after 30 minutes. Femurs were flushed with normalsaline with cell suspension evaluated by flow cytometry

for human and mouse (hCD33-PE, mCD45-PE, BD Phar-mingen) myeloid antigen expression as previouslydescribed (31). Experiments evaluating in vivo TH-302

TH

-302

Veh

icle

TH

-302

Veh

icle

Day 11 Day 14

Day 11Day 7 Day 14

Day 18

HL60A

B

* **

**

HEL

**

Figure 3. TH-302 treatment exerts in vivo anti-leukemic activity. Sublethally irradiated SCIDmice were injected via tail vein with human AML (HL60-luciferase,HEL-luciferase) cells. Systemic disease engraftment was confirmed by BLI approximately 7 to 10 days after tumor inoculation. Mice were then dividedinto groups of 5 to 10 animals and treated with vehicle (PBS) or TH-302 (50 mg/kg) i.p. 5 times a week for 3 weeks. A and B, representative whole-animalBLI on successive treatment days (expressed as total average photon flux per animal in each treatment group) is shown.P values from Student t-test analysisare denoted as � for P < 0.05 and �� for P < 0.001. (Continued on the following page.)

Table 1. Summary of the anti-leukemic activity of different TH-302 regimens

Human AMLxenograftmodel

Numberof miceper group TH-302 regimen

Totalweeklydose

Optimal % total fluxreduction vs. vehicle

Overall survivaldifference

HEL n ¼ 10 50 mg/kg (5�/week) 250 mg 83% (day 14, P ¼ 0.05) Yes (P ¼ 0.0285)HEL n ¼ 5 75 mg/kg (2�/week) 150 mg 93% (day 21, P ¼ 0.0398) No (P ¼ 0.0875)HEL n ¼ 15 100 mg/kg (once/week) 100 mg 75% (day 7, P ¼ 0.046) No (P ¼ NS)HL60 n ¼ 10 50 mg/kg (5�/week) 250 mg 81% (day 19, P ¼ 0.0003) Yes (P ¼ 0.0053)HL60 n ¼ 10 100 mg/kg (once/week) 100 mg 73% (day 14, P ¼ 0.09) No (P ¼ NS)

Portwood et al.

Clin Cancer Res; 19(23) December 1, 2013 Clinical Cancer Research6512

on August 21, 2020. © 2013 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 2, 2013; DOI: 10.1158/1078-0432.CCR-13-0674

Page 8: Activity of the Hypoxia-Activated Prodrug, TH-302, in ... · TH-302 was also effective in advanced AML disease, the setting in which marrow hypoxia would be expected to be greatest

effects were carried out in leukemia-na€�ve and systemicleukemia-engrafted animals. Mice were divided intogroups of 5 to 10 animals and treated with vehicle (PBS)or TH-302 (at doses ranging from 50–100 mg/kg i.p. oncea week up to 5 times a week). Mice were monitored 3times per week for moribund signs (i.e., weight loss,paralysis, respiratory distress) prompting sacrifice.

ImmunohistochemistryMurine sternums were fixed in zinc fixative for 48 hours,

decalcified for 48hours, and embedded in paraffin. Sectionswere cut at 5 mm, placed on charged slides, and dried in a60�C oven for 1 hour. Room temperature slides weredeparaffinized in 3 changes of xylene and rehydrated usinggraded alcohols. Endogenous peroxidase was quenchedwith aqueous 3% H2O2 for 10 minutes and washed withPBS with Tween 20 (PBS/T). Antigen retrieval was con-ducted in the microwave for 10 minutes in pH 6.0 citratebuffer, with a 15-minute cool down followed by a PBS/T

wash. Slides were then loaded on the DAKO autostainer(Dako). Casein 0.03% was used to block for 30 minutes,blownoff, and then theprimary antibodyHypoxyprobewasapplied at 1.4 mg/mL (1:50) to slides for 1 hour. An isotype-matched control (1.4 mg/mL mo IgG1) was used on aduplicate slide in place of the primary antibody as a negativecontrol. A PBS/T wash was followed by anti-mouse bioti-nylated secondary antibody and ABC reagent (Vector Labs)for 30 minutes each with PBS/T washes between reagents.The chromagen DABþ (Dako) was applied for 10 minutes.Slides were counterstained with hematoxylin, dehydrated,cleared, and coverslipped. Slides were evaluated using anOlympus Bx40 light microscope with 10�/0.30 and 20�/0.50 lenses and photographed with a Hitachi HV-C20Ucamera using Image Pro Plus Software (Media Cybernetics).

Statistical analysisResults of xenograft experiments are shown as mean BLI

(� SEM) at each time point for groups of 10 to 20 mice.

HL60 HEL

P = 0.005P = 0.024

Pim

onid

azo

le

×10

×20

VehicleD

C

TH-302

Figure 3. (Continued. ) C, overallsurvival analyses of HEL- andHL60-engrafted mice by treatmentgroup showsignificantly prolongedsurvival following TH-302treatment versus vehicle controls(Kaplan–Meier analysis, P < 0.05).D, bone marrow samples obtainedon treatment day 14 from HEL-luciferase–engrafted animalsshowed reduced numbers ofhypoxic (pimonidazole-positivebrown staining cells) marrow cellsfollowing TH-302 treatment ascompared with vehicle controls(10–20� magnification, OlympusBx40 light microscope). Allexperiments were independentlyrepeated at least 3 times.

TH-302 in Acute Myeloid Leukemia

www.aacrjournals.org Clin Cancer Res; 19(23) December 1, 2013 6513

on August 21, 2020. © 2013 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 2, 2013; DOI: 10.1158/1078-0432.CCR-13-0674

Page 9: Activity of the Hypoxia-Activated Prodrug, TH-302, in ... · TH-302 was also effective in advanced AML disease, the setting in which marrow hypoxia would be expected to be greatest

Statistical evaluation of differences in mean values wasconducted using 2-tailed Student t test analysis withunequal variance. All P < 0.05 was considered significant.Log-rank tests were used to calculate the statistical signifi-cance of the difference in Kaplan–Meier survival curves.These analyses were conducted using the GraphPad Prismsoftware program (Prism v5.0, GraphPad Software).

ResultsMarrow hypoxia increases with progressive leukemiadisease

Prior data in animal models showed that progressivegrowthof AML cellswithin themarrow spacewas associatedwith worsening hypoxia (10, 13, 14). To confirm this andthe hypoxic nature of themarrowmicroenvironment in ourmodels, we assessed the expression of the hypoxia markerpimonidazole in the bone marrow of irradiated SCID micebefore and at fixed time points following intravenous (tailvein) inoculation of human AML (HEL-luciferase) cells. Wenoted increased numbers of pimonidazole-positive cells inthe marrow over time which correlated with evidence ofincreasing in vivo leukemia disease by BLI (SupplementaryFig. S1) and marrow flow cytometry for human CD33antigen expressed by HEL cells (Supplementary Fig. S1).To test whether residual AML cells resistant to chemother-apy also expressed hypoxic markers, we established cohortsof leukemia-engrafted mice. Twenty days after leukemiainoculationwhen all animals had clear evidence of systemicAML disease by BLI, mice were treated with single doses ofvehicle (PBS), cytarabine, doxorubicin, or the VEGF inhib-itor aflibercept (VEGF Trap, Regeneron/Sanofi Aventis;ref. 31). Animals were sacrificed 3 days later for marrowimmunohistochemistry and flow cytometry. Leukemicbone marrows with evidence of residual AML disease (byflow cytometry for hCD33 expression) after treatment alsoall contained pimonidazole-positive cells (representativeimages in Supplementary Fig. S2).

TH-302 preferentially inhibits growth of AML cellsunder hypoxia

To determine whether a hypoxic microenvironment con-tributes to the chemoresistance of AML cells, we thenexamined the effects of a fixed dose of cytarabine (100nmol/L) on growth of human AML cells (HEL, HL60,ML-2) under prolonged hypoxic (1% O2) or normoxic(21%O2) conditions for up to72hours. The selectionof 1%O2 for the hypoxic condition was based on prior data thatthis oxygen tension may best reflect conditions withinmarrow hematopoietic niches (3, 32). The cytarabine dosewas selected on the basis of prior dose escalation experi-ments (Supplementary Fig. S3). As shown, AML cells (HEL,HL60, ML-2) propagated under chronic hypoxia for 48 to72 hours exhibited less cytarabine-induced apoptosis thannormoxic controls as determined by Annexin/PE/7-AADflow cytometry, Western blot analyses for apoptotic mar-kers, and cell viability assays with trypan blue exclusion(Fig. 1A–D; Supplementary Fig. S4). Of note, TH-302

treatment induced apoptosis and cell death of AML cells(HEL, HL60) under the same conditions (1% O2, 72hours) where resistance to cytarabine was noted. Addi-tional experiments confirmed the dose- and hypoxia-dependent apoptotic effects of TH-302 against a panelof several other human AML cell lines representingdiverse molecular and cytogenetic disease subtypes (Sup-plementary Table S2).

TH-302 inhibits primary AML colony formation underhypoxia

To assess the relevance of these results to primary AMLdisease, we establishedmethylcellulose colonies using bonemarrow samples from three relapsed patients with AML atour center. Patient 1 was a 78-year-old woman with sec-ondary AML characterized by complex karyotype. Patient 2was a 40-year-old woman with de novo normal karyotypeAML characterized by NPM-1 mutations with wild-typeFLT-3 status. Patient 3 was a 76-year-old woman withtherapy-related normal karyotype AML with unknownmutation status. Colonies were treated with vehicle (PBS),cytarabine, or TH-302 for 72 hours under normoxic (21%O2) or hypoxic (1% O2) conditions and quantified. Asshown, primary AML colonies were less responsive to thegrowth-inhibitory effects of cytarabine treatment underhypoxia versus normoxia (P ¼ 0.01). In contrast, TH-302treatment significantly reduced the number of primaryAML colonies under hypoxic versus normoxic conditions(Fig. 1E).

TH-302 inhibits growth of AML cells via multiplemechanisms

We then examined the potential mechanisms of action ofTH-302 on AML cells. We found that the in vitro prolifer-ation of human AML cells, as determined by tritiatedthymidine incorporation, was highly sensitive to very lowdoses of TH-302 treatment, with hypoxia-specific differ-ences detected after doses as low as 0.1 to 1 nmol/L (Fig.2A). At higher doses of TH-302 (ranging from 0.5 to 50mmol/L), AML cells (HEL, HL60) underwent progressivecell-cycle arrest in a dose-dependent manner with gradualaccumulation of cells in the G0–G1 phase similar to priorreports in other tumors (Fig. 2B, Supplementary Table S1;ref. 26). We then examined TH-302 induced cross-linkingof DNA as expressed by increased phosphorylation of thehistone variant (H2AX) on AML cells (HEL, HL60). Underdiffering oxygen conditions, TH-302 (1–10 mmol/L)treatment of AML cells resulted in significant hypoxia- anddose-dependent DNA damage (Fig. 2C, data not shown).These data were confirmed by measurement of DNAfragmentation via terminal deoxynucleotidyl transferase–mediated dUTPnick end labeling (TUNEL) assay inhypoxicAML cells under the same conditions (Fig. 2D). TH-302treatment in AML cells was also associated with dose-dependent decreases in HIF-1a protein, a master transcrip-tion factor typically upregulated in hypoxic cells (Fig. 2E).

We observed that AML cells cultured under chronichypoxia (1%O2, from24 to 72 hours) generated increasing

Portwood et al.

Clin Cancer Res; 19(23) December 1, 2013 Clinical Cancer Research6514

on August 21, 2020. © 2013 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 2, 2013; DOI: 10.1158/1078-0432.CCR-13-0674

Page 10: Activity of the Hypoxia-Activated Prodrug, TH-302, in ... · TH-302 was also effective in advanced AML disease, the setting in which marrow hypoxia would be expected to be greatest

levels of ROS, as detected by the fluorescent dyeCM-H2DCFDA (Supplementary Fig. S5A). ROS productionunder these conditions appeared to be dependent onNADPH oxidase and/or other NAD(P) enzymes as theywere attenuated in the presence of the inhibitor DPI(Supplementary Fig. S5B). We found that that TH-302decreased hypoxia-induced ROS generation by viableAML cells (HEL, HL60) in a dose-dependent manner atdrug concentrations where increased leukemia cell deathwas noted (0.5–5 mmol/L). Production of ROS by nor-moxic AML cells was not affected by TH-302 treatment(Fig. 2F and G, data not shown). Similar effects onhypoxia-induced ROS production were noted followingcytarabine treatment of AML cells at these same timepoints (Supplementary Fig. S5C).

TH-302 inhibits systemic AML growth in preclinicalmodelsWe then evaluated the in vivo activity of TH-302 in

SCID mice systemically engrafted with luciferase-trans-fected human AML (HEL, HL60) cells in the bone marrowand other extramedullary sites. Animals were treated withvehicle (PBS) or TH-302 administered at 3 differingdosing regimens based on prior reported literature(24–26). As summarized in Table 1, TH-302 treatmentsignificantly reduced overall leukemia disease burden, asreflected by differences in BLI, in both animal models in 3of 5 TH-302 treatment regimens (P < 0.05). However,only one regimen (TH-302 administered at 50 mg/kg 5times a week for a total weekly dose of 250 mg/kg)resulted in significantly prolonged overall survival in bothmodels as compared with vehicle controls (Table 1, Fig.3A–C). Pimonidazole staining of bone marrow from

leukemia-engrafted SCID mice following 3 weeks ofTH-302 (50 mg/kg 5 times a week) therapy showed fewerhypoxic cells than vehicle-treated mice. This differencewas specifically seen in the diaphyses of leukemia-engrafted mice (Fig. 3D). TH-302 exerted no obviouseffects on marrow vasculature, as reflected by VEGFR-3immunostaining of leukemic bone marrow sections (datanot shown).

Because normal pluripotent hematopoietic stem cellsalso reside within underperfused hypoxic marrow niches,we specifically investigated the potential effects of pro-longed in vivo TH-302 administration on normal hemato-poiesis. We found no significant differences in mean whiteblood cell, neutrophil, lymphocyte, or platelet counts orhemoglobin levels of nonleukemic or leukemia-engraftedmice following 2 weeks of vehicle versus TH-302 treatment(100 mg/kg i.p. once a week or 50 mg/kg i.p. 5 times aweek; Table 2).

TH-302 treatment was effective in a model of advancedAML disease

It has previously been reported that the in vivo efficacy ofTH-302 correlated with the degree of tumor hypoxia, asmeasured by pimonidazole staining, across eleven solidtumor xenograft models (24). On the basis of our findingthat bone marrow hypoxia increased with progressive AMLdisease in our models (Supplementary Fig. S1), we com-pared the relative in vivo efficacy of TH-302 in early (lesshypoxic) and late (more hypoxic) AML disease. We there-fore established cohorts of HEL-luciferase–engrafted micevia tail vein injection but varied the timing of treatmentinitiation with vehicle or TH-302. TH-302 therapy (50 mg/kg 5 times a week) was begun either on day 1 (1 day before

Table 2. Peripheral blood counts

Parameter Vehicle TH-302 P Normal range

A. Following TH-302 treatment in non-leukemic SCID micea

White blood cells (�108 cells/L) 2.0 � 0.3 1.4 � 0.2 0.10 1.8–15.0Neutrophils (�108 cells/L) 1.1 � 0.2 0.8 � 0.1 0.21 0.1–5.7Lymphocytes (�108 cells/L) 0.6 � 0.1 0.5 � 0.1 0.55 0.9–14.2Hemoglobin, g/dL 14 � 0.3 13 � 0.3 0.21 12.2–16.4Platelets (�109 cells/L) 477 � 37 576 � 38 0.09 100–1200B. On treatment day 14 in leukemia-engrafted SCID miceb

White blood cells (�108 cells/L) 8.2 � 2.9 8.9 � 3.5 0.90 1.8–15.0Neutrophils (�108 cells/L) 4.7 � 1.7 5.2 � 2.2 0.86 0.1–5.7Lymphocytes (�108 cells/L) 1.8 � 0.37 2.4 � 0.83 0.55 0.9–14.2Hemoglobin, g/dL 15 � 0.7 13 � 1.1 0.31 12.2–16.4Platelets (�109 cells/L) 790 � 21 930 � 50 0.80 100–1200

aMean � SE values of 6 mice per treatment group. No significant differences in white blood cell, hematocrit levels, platelet counts,neutrophil or lymphocyte percentages were noted in peripheral blood samples obtained after 7–14 days of treatment with vehicleversus TH-302 (50 mg/kg i.p. 5 times a week) in nonleukemic SCID mice (n ¼ 6 mice/group).bMean � SE values of 4 mice per treatment group. No significant differences in white blood cell, hematocrit levels, platelet counts,neutrophil or lymphocyte percentages were noted in peripheral blood samples obtained after 14 days of treatment with vehicle versusTH-302 (100 mg/kg i.p. once a week) in HEL-luciferase–engrafted SCID mice (n ¼ 4 mice/group).

TH-302 in Acute Myeloid Leukemia

www.aacrjournals.org Clin Cancer Res; 19(23) December 1, 2013 6515

on August 21, 2020. © 2013 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 2, 2013; DOI: 10.1158/1078-0432.CCR-13-0674

Page 11: Activity of the Hypoxia-Activated Prodrug, TH-302, in ... · TH-302 was also effective in advanced AML disease, the setting in which marrow hypoxia would be expected to be greatest

leukemia inoculation, termed "Early TH-302") or on day 10(8 days following leukemia inoculation, termed "Late TH-302). Despite the differences in treatment duration, both

"early" (20-day total duration) and "late" (10-day totalduration) TH-302 regimens resulted in similar degrees ofleukemia growth inhibition as determined by BLI

Day 1:

Vehicle

(PBS)

Day 2:

Inject

AML cells

Days 3–20: Vehicle (PBS) treatment

Day 1:

TH-302

Day 2:

Inject

AML cells

Days 3–20: TH-302 treatment

Days 10–20: TH-302 treatment

Day 20: Image mice

Vehicle

A

B

EarlyTH-302

LateTH-302

Observation

Day 20: Image mice

Day 20: Image mice

Day 2:

Inject

AML cells

Day 20 Day 20

* *TH-302 Early TH-302 Late

Vehicle

Figure 4. TH-302 treatment inhibits different stages of AML disease. A, experimental schema showing treatment regimens for different groups of irradiatedSCID mice (n ¼ 5). Vehicle controls (top row) received their first dose of PBS on day 1 followed by HEL-luciferase inoculation via tail vein on day 2and an additional 19 days of treatment (total 20 days). "Early" TH-302–treatedmice (middle row) received their first dose of TH-302 (50mg/kg i.p. 5�/week) onday 1 followed on day 2 by leukemia inoculation via tail vein and an additional 19 days of treatment (total 20 days). "Late" TH-302–treated mice (bottomrow) did not start TH-302 treatment until day 10 when systemic disease was documented by BLI. These animals received a total of 10 days of treatment.B and C, shown are representative BLI and total photon flux per animal per treatment group on day 20. These results show similar inhibition of AML growth inboth "early" and "late" TH-302 treatment groups as compared to vehicle controls by Student t-test analysis. P < 0.05 is denoted by an asterisk.

Portwood et al.

Clin Cancer Res; 19(23) December 1, 2013 Clinical Cancer Research6516

on August 21, 2020. © 2013 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 2, 2013; DOI: 10.1158/1078-0432.CCR-13-0674

Page 12: Activity of the Hypoxia-Activated Prodrug, TH-302, in ... · TH-302 was also effective in advanced AML disease, the setting in which marrow hypoxia would be expected to be greatest

measurement on day 20. Also of note, in these experiments,administration of the first dose of TH-302 up to 2 weeksbefore tumor cell inoculation did not appear to impacton the ability of HEL-luciferase AML cells to efficientlyengraft and propagate in SCID mice as determined bysuccessive BLI imaging (Fig. 4, data not shown).

DiscussionEmerging data have highlighted the intrinsic hypoxia of

the bone marrow microenvironment as a critical in vivomodulator of malignant hematopoietic growth (2–4).Preclinical evidence has shown that expansion of acuteleukemia cells within the bone marrow is associated withfurther decreases in local oxygenation (10, 13, 14). Weand other groups have shown that acute leukemia cellsplaced under hypoxia upregulate HIF-1a, undergo cell-cycle arrest, and exhibit resistance to chemotherapyagents such as cytarabine (22, 23).We postulated that TH-302, a hypoxia-activated nitrogen

mustard derivative prodrug, would be effective in eradicat-ing AML cells within a hypoxic bone marrow microenvi-ronment. Here, we show that TH-302 induced hypoxiaand dose-dependent cell death in human AML cell linesand primary AML samples under the same conditions (1%O2, 72 hours) where cytarabine resistance was observed.Mechanisms of action of TH-302 under hypoxia includedcell-cycle arrest, DNA strand breakage, and inhibition ofHIF-1a protein expression.It has been proposed that amajor benefit of a chronically

hypoxic marrow microenvironment to hematopoietic cellsis reduced exposure to potentially damaging ROSwhich caninduce genomic instability and DNA damage (8). In fact,under normoxia, chemotherapeutic agents, such as cytar-abine, typically induce ROS followed by cell death in AMLcells (33). In our experiments, we noted that human AMLcells culture under prolonged hypoxia (1% O2 for 48–72hours) expressed progressively higher levels of ROS overtime. Although the mechanisms underlying this ROS pro-duction are unclear, they appear to depend, in part, onNAPDH-oxidase. Treatment of AML with TH-302 (as wellas cytarabine) under these same conditions markedlydecreased ROS generation by hypoxic AML cells. Elevatedoxidative stress in primary leukemia blasts has recently beenlinked to AML progression, relapse, and poor prognosis,specifically in FLT-3–mutated AML (34, 35). Increased ROSmay also regulate leukemia self-renewal and survival. Forinstance, ROS generation contributes to leukemogenesismediated by BCR-ABL in hematopoietic cells and promotesthe growth and migration of cells expressing other onco-genic kinases (36). The negative effects of TH-302 on ROSgeneration therefore may be of therapeutic benefit inpatients with AML.Our results show that TH-302 treatment inhibited AML

disease and prolonged survival in 2 systemic human AMLmodels at drug doses similar to those previously evaluatedin human patients (27, 28). Because hypoxia is critical inregulating normal HSC biology, we examined whether

treatment induced any significant myelosuppression inleukemia-engrafted or leukemia-naive SCID mice. After 2weeks of therapy, no hematologic toxicities were noted.These latter results are in line with prior clinical trialsshowing limited hematological toxicity of TH-302 therapyin patients with solid tumors (27, 28). The differentialeffects of TH-302 on normal versus malignant hematopoi-etic cells could be explained by the selective hypoxic natureof this agent. Because acute leukemia growth markedlyexpands hypoxic marrow regions, TH-302 may preferen-tially eradicate tumor cells populating these lowoxygenatedregionswhile sparingHSCs in better perfused areas. TH-302may also selectively target leukemia cells via hypoxia-inde-pendent mechanisms; for instance, it has been shown thatsolid tumor cells deficient in homology-dependent repairexhibit enhanced sensitivity to TH-302 (25).

Hypoxia has also increasingly been implicated in leuke-mia stem cell biology (37). We noted that in vitro TH-302treatment decreased numbers of primary AML coloniesunder hypoxia. However, in contrast, in vivo TH-302treatment for up to 2 weeks before the inoculation ofhuman AML (HEL-luciferase) cells in irradiated SCID hostsdid not affect subsequent disease engraftment or propaga-tion. Because our experiments did not clearly distinguishbetween the effects of TH-302 on leukemia stem cells asopposed to bulk hypoxic AML cells, further studies areneeded.

Overall the results presented here, together with earlyresults of clinical activity in a phase I trial of TH-302 inhematologic cancers, provide the rationale for further clin-ical investigation of TH-302 for patients with AML (27–29).Our finding that chronic (5 times per week) administrationof TH-302 was associated with optimal in vivo activity inpreclinical AML models may be relevant for future dosingregimens. Further development of TH-302 and other hyp-oxia-activated agents for acute leukemia therapy, however,may need to take into account the unique mechanism ofaction of such drugs within the leukemic marrow micro-environment (11, 38–40). Because hypoxic regions withinthe marrow expand as a consequence of acute leukemiaprogression, TH-302 may be most effectively used in laterstage disease (10, 13, 14). Moreover, the presence of resid-ual cells expressing hypoxic markers in leukemic marrowsfollowing cytarabine or doxorubicin treatment suggests thatthe administrating TH-302 together with or following stan-dard chemotherapy may further improve outcomes. Newercombinations of TH-302 with other therapeutic agents(such as bortezomib) with potential synergistic antitumoractivity also hold promise for AML (41). Additional clinicalstudies are needed to verify these findings and potentiallydefine those subsets of patientswithAMLwhomight benefitfrom TH-302 therapy.

Disclosure of Potential Conflicts of InterestC.P. Hart is an employee and stockholder of Threshold Pharmaceuticals,

Inc. No potential conflicts of interest were disclosed by the other authors.

Authors' ContributionsConception and design: Y.-C. Hsu, C.P. Hart, E.S. Wang

TH-302 in Acute Myeloid Leukemia

www.aacrjournals.org Clin Cancer Res; 19(23) December 1, 2013 6517

on August 21, 2020. © 2013 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 2, 2013; DOI: 10.1158/1078-0432.CCR-13-0674

Page 13: Activity of the Hypoxia-Activated Prodrug, TH-302, in ... · TH-302 was also effective in advanced AML disease, the setting in which marrow hypoxia would be expected to be greatest

Development of methodology: Y.-C. Hsu, E.S. WangAcquisitionofdata (provided animals, acquired andmanagedpatients,provided facilities, etc.): D. Lal, M. Wetzler, E.S. WangAnalysis and interpretation of data (e.g., statistical analysis, biosta-tistics, computational analysis): S. Portwood, D. Lal, M.K. Johnson, E.S.WangWriting, review, and/or revision of the manuscript: S. Portwood, Y.-C.Hsu, M.K. Johnson, M. Wetzler, C.P. Hart, E.S. WangAdministrative, technical, or material support (i.e., reporting or orga-nizing data, constructing databases): S. Portwood, D. Lal, Y.-C. Hsu,R. Vargas, M.K. JohnsonStudy supervision: E.S. WangProvided primary patient samples and analyzed and interpreted data:M. WetzlerProvided reagent: C.P. Hart

AcknowledgmentsThe authors thank the Roswell Park Cancer Institute Core Facilities for

their contributions to thiswork, specifically the FlowCytometry and ImagingResource (Earl Timm), Department of Laboratory Animal Research, andPathology Research Core (Mary Vaughan, Angela Omilian). Other indivi-

duals who contributed to this work include Krista Pundt (RPCI, DeptPharmacology and Therapeutics) and R. Robert Vathanayagam (Dept Med-icine, RPCI). Thismanuscript is dedicated to thememory ofDr. JohnG.Curd(Threshold Pharmaceuticals, Inc) who provided valuable encouragementand suggestions.

Grant SupportResearch funding was provided by the Roswell Park Alliance Foundation

(Mr. and Mrs. E. LaFranier; Jacquie Hirsch Leukemia Research Fund; theSzefel Leukemia Research Fund), an American Cancer Society MentoredResearch Scientist Award (MRSG-06-044-01-LIB), and When Everyone Sur-vives Foundation grant. Core resource support was provided by the NCICancer Center Support Grant for RPCI (CA016156).

The costs of publication of this article were defrayed in part by thepayment of page charges. This article must therefore be hereby markedadvertisement in accordance with 18 U.S.C. Section 1734 solely to indicatethis fact.

ReceivedMarch11, 2013; revisedAugust 24, 2013; accepted September 13,2013; published OnlineFirst October 2, 2013.

References1. Konopleva MY, Jordan CT. Leukemia stem cells and microenviron-

ment: biology and therapeutic targeting. J Clin Oncol 2011;29:591–9.2. Colla S, Storti P, Donofrio G, Todoerti K, Bolzoni M, Lazzaretti M, et al.

Low bone marrow oxygen tension and hypoxia-inducible factor-1alpha overexpression characterize patients with multiple myeloma:role on the transcriptional and proangiogenic profiles of CD138(þ)cells. Leukemia 2010;24:1967–70.

3. Fiegl M, Samudio I, Clise-Dwyer K, Burks JK, Mnjoyan Z, Andreeff M.CXCR4 expression and biologic activity in acute myeloid leukemia aredependent on oxygen partial pressure. Blood 2009;113:1504–12.

4. Harrison JS, Rameshwar P, Chang V, Bandari P. Oxygen saturation inthe bone marrow of healthy volunteers. Blood 2002;99:394.

5. Chow DC, Wenning LA, Miller WM, Papoutsakis ET. Modeling pO(2)distributions in the bone marrow hematopoietic compartment. II.Modified Kroghian models. Biophys J 2001;81:685–96.

6. Parmar K, Mauch P, Vergilio JA, Sackstein R, Down JD. Distribution ofhematopoietic stem cells in the bone marrow according to regionalhypoxia. Proc Natl Acad Sci U S A 2007;104:5431–6.

7. Winkler IG, Barbier V, Wadley R, Zannettino AC, Williams S, LevesqueJP. Positioning of bone marrow hematopoietic and stromal cellsrelative to blood flow in vivo: serially reconstituting hematopoieticstem cells reside in distinct nonperfused niches. Blood 2010;116:375–85.

8. Eliasson P, Jonsson J. The hematopoietic stem cell niche: low inoxygen but a nice place to be. J Cell Physiol 2010;222:17–22.

9. Giuntoli S, Rovida E, Gozzini A, Barbetti V, Cipolleschi MG, OlivottoM,et al. Severe hypoxia defines heterogeneity and selects highly imma-ture progenitors within clonal erythroleukemia cells. Stem Cells (Day-ton, Ohio) 2007;25:1119–25.

10. Schaefer C, KrauseM, Fuhrhop I, Schroeder M, Algenstaedt P, FiedlerW, et al. Time-course-dependent microvascular alterations in a modelof myeloid leukemia in vivo. Leukemia 2008;22:59–65.

11. Benito J, Shi Y, Szymanska B, Carol H, Boehm I, Lu H, et al. Pro-nounced hypoxia in models of murine and human leukemia: highefficacy of hypoxia-activated prodrug PR-104. PLoS One 2011;6:e23108.

12. Chen Y, Jacamo R, Shi YX, Wang RY, Battula VL, Konoplev S, et al.Human extramedullary bone marrow in mice: a novel in vivo model ofgenetically controlled hematopoietic microenvironment. Blood2012;119:4971–80.

13. Mortensen BT, Jensen PO, Helledie N, Iversen PO, Ralfkiaer E, LarsenJK, et al. Changing bone marrow micro-environment during develop-ment of acute myeloid leukaemia in rats. Br J Haematol 1998;102:458–64.

14. Iversen PO, Thing-Mortensen B, Nicolaysen G, Benestad HB.Decreased blood flow to rat bone marrow, bone, spleen, and liver inacute leukemia. Leuk Res 1993;17:663–8.

15. Jensen PO, Mortensen BT, Hodgkiss RJ, Iversen PO, Christensen IJ,HelledieN, et al. Increased cellular hypoxia and reducedproliferation ofboth normal and leukaemic cells during progression of acute myeloidleukaemia in rats. Cell Prolif 2000;33:381–95.

16. Harris AL. Hypoxia–a key regulatory factor in tumour growth. Nat RevCancer 2002;2:38–47.

17. Bertout JA, Patel SA, Simon MC. The impact of O2 availability onhuman cancer. Nat Rev Cancer 2008;8:967–75.

18. Giuntoli S, Rovida E, Barbetti V, Cipolleschi MG, Olivotto M, DelloSbarba P. Hypoxia suppresses BCR/Abl and selects imatinib-insen-sitive progenitors within clonal CML populations. Leukemia 2006;20:1291–3.

19. Fiegl M, Samudio I, Mnjoyan Z, Korchin B, Fritsche H, Andreeff M.Physiological hypoxia promotes lipid raft and PI3K-dependent acti-vation of MAPK 42/44 in leukemia cells. Leukemia 2010;24:1364–7.

20. Deeb G, Vaughan MM, McInnis I, Ford LA, Sait SN, Starostik P, et al.Hypoxia-inducible factor-1alpha protein expression is associatedwithpoor survival in normal karyotype adult acute myeloid leukemia. LeukRes 2011;35:579–84.

21. Wellmann S, Guschmann M, Griethe W, Eckert C, von Stackelberg A,Lottaz C, et al. Activation of the HIF pathway in childhood ALL,prognostic implications of VEGF. Leukemia 2004;18:926–33.

22. Matsunaga T, Imataki O, Torii E, Kameda T, Shide K, Shimoda H, et al.Elevated HIF-1alpha expression of acute myelogenous leukemia stemcells in the endosteal hypoxic zonemay be a cause of minimal residualdisease in bone marrow after chemotherapy. Leuk Res 2012;36:e122–4.

23. FrolovaO, Samudio I, Benito JM, JacamoR,Kornblau SM,Markovic A,et al. Regulation ofHIF-1alpha signaling and chemoresistance in acutelymphocytic leukemia under hypoxic conditions of the bone marrowmicroenvironment. Cancer Biol Ther 2012;13:858–70.

24. Sun JD, Liu Q,Wang J, Ahluwalia D, Ferraro D,Wang Y, et al. Selectivetumor hypoxia targeting by hypoxia-activated prodrug TH-302 inhibitstumor growth in preclinical models of cancer. Clin Cancer Res2012;18:758–70.

25. Meng F, Evans JW, Bhupathi D, Banica M, Lan L, Lorente G, et al.Molecular and cellular pharmacology of the hypoxia-activatedprodrugTH-302. Mol Cancer Ther 2012;11:740–51.

26. HuJ,HandisidesDR, VanValckenborghE,DeRaeveH,MenuE, VandeBroek I, et al. Targeting the multiple myeloma hypoxic niche with TH-302, a hypoxia-activated prodrug. Blood 2010;116:1524–7.

27. Ganjoo KN, Cranmer LD, Butrynski JE, Rushing D, Adkins D, OkunoSH, et al. A phase I study of the safety and pharmacokinetics of thehypoxia-activated prodrug TH-302 in combination with doxorubicin inpatients with advanced soft tissue sarcoma. Oncology 2011;80:50–6.

28. Weiss GJ, Infante JR, Chiorean EG, Borad MJ, Bendell JC, Molina JR,et al. Phase 1 study of the safety, tolerability, and pharmacokinetics of

Portwood et al.

Clin Cancer Res; 19(23) December 1, 2013 Clinical Cancer Research6518

on August 21, 2020. © 2013 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 2, 2013; DOI: 10.1158/1078-0432.CCR-13-0674

Page 14: Activity of the Hypoxia-Activated Prodrug, TH-302, in ... · TH-302 was also effective in advanced AML disease, the setting in which marrow hypoxia would be expected to be greatest

TH-302, a hypoxia-activated prodrug, in patients with advanced solidmalignancies. Clin Cancer Res 2011;17:2997–3004.

29. Thomas D, Handisides DR, Lorente GA, Benito JM, Borthakur G,Jabbour E, et al. Phase 1 study of TH-302, a hypoxia activatedcytotoxic prodrug, in subjects with advanced leukemia. J Clin Oncol30, 2012 (suppl; abstr 6585).

30. Tanabe S, Zeleznik-Le NJ, Kobayashi H, Vignon C, Espinosa R III,LeBeauMM, et al. Analysis of the t(6;11)(q27;q23) in leukemia shows aconsistent breakpoint in AF6 in three patients and in the ML-2 cell line.Genes Chromosomes Cancer 1996;15:206–16.

31. Lal D, Park JA, Demock K, Marinaro J, Perez AM, Lin MH, et al.Aflibercept exerts antivascular effects and enhances levels of anthra-cycline chemotherapy in vivo in human acute myeloid leukemia mod-els. Mol Cancer Ther 2010;9:2737–51.

32. Desplat V, Faucher J, Mahon F, Dello Sbarba P, Praloran V, Ivanovic Z.Hypoxia modifies proliferation and differentiation of CD34(þ) CMLcells. Stem Cells (Dayton, Ohio) 2002;20:347–54.

33. Hedley DW, McCulloch EA. Generation of reactive oxygen intermedi-ates after treatment of blasts of acute myeloblastic leukemia withcytosine arabinoside: role of bcl-2. Leukemia 1996;10:1143–9.

34. Zhou FL, Zhang WG, Wei YC, Meng S, Bai GG, Wang BY, et al.Involvement of oxidative stress in the relapse of acute myeloid leu-kemia. J Biol Chem 2010;285:15010–5.

35. Sallmyr A, Fan J, Datta K, Kim KT, Grosu D, Shapiro P, et al. Internaltandem duplication of FLT3 (FLT3/ITD) induces increased ROS pro-duction, DNA damage, and misrepair: implications for poor prognosisin AML. Blood 2008;111:3173–82.

36. Hole PS, Darley RL, Tonks A. Do reactive oxygen species play a role inmyeloid leukemias? Blood 2011;117:5816–26.

37. Takubo K, Suda T. Roles of the hypoxia response system in hemato-poietic and leukemic stem cells. Int J Hematol 2012;95:478–83.

38. Houghton PJ, Lock R, Carol H, Morton CL, Phelps D, Gorlick R, et al.Initial testing of the hypoxia-activated prodrug PR-104 by the pediatricpreclinical testing program. Pediatr Blood Cancer 2011;57:443–53.

39. Hunter FW, Wang J, Patel R, Hsu HL, Hickey AJ, Hay MP, et al.Homologous recombination repair-dependent cytotoxicity of the ben-zotriazine di-N-oxide CEN-209: comparison with other hypoxia-acti-vated prodrugs. Biochem Pharmacol 2012;83:574–85.

40. Baumann RP, Ishiguro K, Penketh PG, Shyam K, Zhu R, Sartorelli AC.KS900: a hypoxia-directed, reductively activated methylating antitu-mor prodrug that selectively ablates O(6)-alkylguanine-DNA alkyl-transferase in neoplastic cells. Biochem Pharmacol 2011;81:1201–10.

41. Hu J, Van Valckenborgh E, Dehui X, Menu E, De RaeveH, De Bruyne E,et al. Synergistic induction of apoptosis in multiple myeloma cells bybortezomib andhypoxia-activated prodrug TH-302, in vivo and in vitro.Mol Cancer Ther Published OnlineFirst July 5, 2013.

TH-302 in Acute Myeloid Leukemia

www.aacrjournals.org Clin Cancer Res; 19(23) December 1, 2013 6519

on August 21, 2020. © 2013 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 2, 2013; DOI: 10.1158/1078-0432.CCR-13-0674

Page 15: Activity of the Hypoxia-Activated Prodrug, TH-302, in ... · TH-302 was also effective in advanced AML disease, the setting in which marrow hypoxia would be expected to be greatest

2013;19:6506-6519. Published OnlineFirst October 2, 2013.Clin Cancer Res   Scott Portwood, Deepika Lal, Yung-Chun Hsu, et al.   Human Acute Myeloid Leukemia ModelsActivity of the Hypoxia-Activated Prodrug, TH-302, in Preclinical

  Updated version

  10.1158/1078-0432.CCR-13-0674doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://clincancerres.aacrjournals.org/content/suppl/2013/10/02/1078-0432.CCR-13-0674.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://clincancerres.aacrjournals.org/content/19/23/6506.full#ref-list-1

This article cites 39 articles, 14 of which you can access for free at:

  Citing articles

  http://clincancerres.aacrjournals.org/content/19/23/6506.full#related-urls

This article has been cited by 5 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]

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

  Permissions

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

.http://clincancerres.aacrjournals.org/content/19/23/6506To request permission to re-use all or part of this article, use this link

on August 21, 2020. © 2013 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 2, 2013; DOI: 10.1158/1078-0432.CCR-13-0674