12
Cancer Therapy: Preclinical Pralatrexate Is Synergistic with the Proteasome Inhibitor Bortezomib in In vitro and In vivo Models of T-Cell Lymphoid Malignancies Enrica Marchi 1 , Luca Paoluzzi 1 , Luigi Scotto 1 , Venkatraman E. Seshan 2 , Jasmine M. Zain 1 , Pier Luigi Zinzani 3 , and Owen A. O'Connor 1 Abstract Purpose: Pralatrexate (10-propargyl-10-deazaaminopterin) is an antifolate with improved cellular up- take and retention due to greater affinity for the reduced folate carrier (RFC-1) and folyl-polyglutamyl synthase. Based on the PROPEL data, pralatrexate was the first drug approved for patients with relapsed and refractory peripheral T-cell lymphoma. Bortezomib is a proteasome inhibitor that has shown some activity in patients with T-cell lymphoma. Experimental Design: Assays for cytotoxicity including mathematical analysis for synergism, flow cy- tometry, immunoblotting, and a xenograft severe combined immunodeficient-beige mouse model were used to explore the in vitro and in vivo activities of pralatrexate alone and in combination with bortezomib in T-cell lymphoid malignancies. Results: In vitro, pralatrexate and bortezomib exhibited concentration- and time-dependent cytotoxi- city against a broad panel of T-lymphoma cell lines. Pralatrexate showed synergism when combined with bortezomib in all cell lines studied. Pralatrexate also induced potent apoptosis and caspase activation when combined with bortezomib across the panel. Cytotoxicity studies on normal peripheral blood mononuclear cells showed that the combination was not more toxic than the single agents. Western blot assays for proteins involved in broad growth and survival pathways showed that p27, NOXA, HH3, and RFC-1 were all significantly modulated by the combination. In a severe combined immunodeficient-beige mouse model of transformed cutaneous T-cell lymphoma, the addition of pralatrexate to bortezomib enhanced efficacy compared with either drug alone. Conclusion: Collectively, these data suggest that pralatrexate in combination with bortezomib repre- sents a novel and potentially important platform for the treatment of T-cell malignancies. Clin Cancer Res; 16(14); 364858. ©2010 AACR. Malignancies derived from mature (post-thymic) T cells and natural killer cells, collectively referred to as peripher- al T-cell lymphomas (PTCL), encompass a variety of rare and often challenging diseases. PTCLs represent 10% to 15% of all nonHodgkin's lymphomas worldwide (1), accounting for 6,000 to 9,000 cases annually in the United States. CHOP (cyclophosphamide, doxorubicin, vincris- tine, and prednisone) and CHOP-like chemotherapy pro- grams are still the most commonly used regimens despite the suboptimal outcomes. Overall, CHOP-based chemo- therapies achieve overall response rates of 30% to 60% with an overall survival at 5 years of approximately 15% to 20%. Efforts to improve on these approaches with more dose-intense combination chemotherapy regimens (2, 3) have failed to show a benefit (62% versus 56%, respec- tively). Given the often dismal results seen in patients with PTCL compared with other subtypes of nonHodgkin's lymphoma, there is an urgent need to identify new agents with demonstrable activity in these T-cell neoplasms. Over the past few years, a number of promising new drugs have emerged, which seem to have marked single-agent activity in T-cell lymphomas, including gemcitabine (47), alemtu- zumab (8, 9), and the histone deacetylase (HDAC) inhibi- tors (10). More recently, pralatrexate, a novel antifol with improved cellular uptake and retention, has become the first drug approved by the U.S. Food and Drug Administra- tion for the treatment of relapsed/refractory PTCL. The results of the PROPEL registration study based on 109 evaluable patients showed an overall response rate Authors' Affiliations: 1 New York University Cancer Institute, NYU Langone Medical Center; 2 Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York; and 3 Institute of Hematology and Medical Oncology L. & A. Serágnoli,University of Bologna, Bologna, Italy Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). Corresponding Author: Owen A. O'Connor, Division of Hematologic Malignancies and Medical Oncology, New York University Cancer Insti- tute, NYU Langone Medical Center, New York, NY 10016. Phone: 212- 263-9884; Fax: 212-263-9210; E-mail: owen.o'[email protected]. doi: 10.1158/1078-0432.CCR-10-0671 ©2010 American Association for Cancer Research. Clinical Cancer Research Clin Cancer Res; 16(14) July 15, 2010 3648 Research. on July 8, 2020. © 2010 American Association for Cancer clincancerres.aacrjournals.org Downloaded from Published OnlineFirst May 25, 2010; DOI: 10.1158/1078-0432.CCR-10-0671

Research Pralatrexate Is Synergistic with the Proteasome Inhibitor ... · Pralatrexate Is Synergistic with the Proteasome Inhibitor Bortezomib in In vitro and In vivo Models of T-Cell

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Research Pralatrexate Is Synergistic with the Proteasome Inhibitor ... · Pralatrexate Is Synergistic with the Proteasome Inhibitor Bortezomib in In vitro and In vivo Models of T-Cell

3648

Published OnlineFirst May 25, 2010; DOI: 10.1158/1078-0432.CCR-10-0671

Cancer Therapy: Preclinical Clinical

Cancer

Research

Pralatrexate Is Synergistic with the Proteasome InhibitorBortezomib in In vitro and In vivo Models of T-CellLymphoid Malignancies

Enrica Marchi1, Luca Paoluzzi1, Luigi Scotto1, Venkatraman E. Seshan2, Jasmine M. Zain1,Pier Luigi Zinzani3, and Owen A. O'Connor1

Abstract

Authors' ALangoneBiostatisticYork; andSerágnoli,”

Note: SuppResearch O

CorresponMalignancitute, NYU263-9884;

doi: 10.115

©2010 Am

Clin Canc

Do

Purpose: Pralatrexate (10-propargyl-10-deazaaminopterin) is an antifolate with improved cellular up-take and retention due to greater affinity for the reduced folate carrier (RFC-1) and folyl-polyglutamylsynthase. Based on the PROPEL data, pralatrexate was the first drug approved for patients with relapsedand refractory peripheral T-cell lymphoma. Bortezomib is a proteasome inhibitor that has shown someactivity in patients with T-cell lymphoma.Experimental Design: Assays for cytotoxicity including mathematical analysis for synergism, flow cy-

tometry, immunoblotting, and a xenograft severe combined immunodeficient-beige mouse model wereused to explore the in vitro and in vivo activities of pralatrexate alone and in combination with bortezomibin T-cell lymphoid malignancies.Results: In vitro, pralatrexate and bortezomib exhibited concentration- and time-dependent cytotoxi-

city against a broad panel of T-lymphoma cell lines. Pralatrexate showed synergism when combined withbortezomib in all cell lines studied. Pralatrexate also induced potent apoptosis and caspase activationwhen combined with bortezomib across the panel. Cytotoxicity studies on normal peripheral bloodmononuclear cells showed that the combination was not more toxic than the single agents. Western blotassays for proteins involved in broad growth and survival pathways showed that p27, NOXA, HH3, andRFC-1 were all significantly modulated by the combination. In a severe combined immunodeficient-beigemouse model of transformed cutaneous T-cell lymphoma, the addition of pralatrexate to bortezomibenhanced efficacy compared with either drug alone.Conclusion: Collectively, these data suggest that pralatrexate in combination with bortezomib repre-

sents a novel and potentially important platform for the treatment of T-cell malignancies. Clin Cancer Res;

16(14); 3648–58. ©2010 AACR.

Malignancies derived from mature (post-thymic) T cellsand natural killer cells, collectively referred to as peripher-al T-cell lymphomas (PTCL), encompass a variety of rareand often challenging diseases. PTCLs represent 10% to15% of all non–Hodgkin's lymphomas worldwide (1),accounting for 6,000 to 9,000 cases annually in the UnitedStates. CHOP (cyclophosphamide, doxorubicin, vincris-tine, and prednisone) and CHOP-like chemotherapy pro-

ffiliations: 1New York University Cancer Institute, NYUMedical Center; 2Department of Epidemiology ands, Memorial Sloan Kettering Cancer Center, New York, New3Institute of Hematology and Medical Oncology “L. & A.University of Bologna, Bologna, Italy

lementary data for this article are available at Clinical Cancernline (http://clincancerres.aacrjournals.org/).

ding Author: Owen A. O'Connor, Division of Hematologices and Medical Oncology, New York University Cancer Insti-Langone Medical Center, New York, NY 10016. Phone: 212-Fax: 212-263-9210; E-mail: owen.o'[email protected].

8/1078-0432.CCR-10-0671

erican Association for Cancer Research.

er Res; 16(14) July 15, 2010

Researcon Julyclincancerres.aacrjournals.org wnloaded from

grams are still the most commonly used regimens despitethe suboptimal outcomes. Overall, CHOP-based chemo-therapies achieve overall response rates of 30% to 60%with an overall survival at 5 years of approximately 15%to 20%. Efforts to improve on these approaches with moredose-intense combination chemotherapy regimens (2, 3)have failed to show a benefit (62% versus 56%, respec-tively). Given the often dismal results seen in patients withPTCL compared with other subtypes of non–Hodgkin'slymphoma, there is an urgent need to identify new agentswith demonstrable activity in these T-cell neoplasms. Overthe past few years, a number of promising new drugs haveemerged, which seem to have marked single-agent activityin T-cell lymphomas, including gemcitabine (4–7), alemtu-zumab (8, 9), and the histone deacetylase (HDAC) inhibi-tors (10). More recently, pralatrexate, a novel antifol withimproved cellular uptake and retention, has become thefirst drug approved by the U.S. Food and Drug Administra-tion for the treatment of relapsed/refractory PTCL.The results of the PROPEL registration study based on

109 evaluable patients showed an overall response rate

h. 8, 2020. © 2010 American Association for Cancer

Page 2: Research Pralatrexate Is Synergistic with the Proteasome Inhibitor ... · Pralatrexate Is Synergistic with the Proteasome Inhibitor Bortezomib in In vitro and In vivo Models of T-Cell

Pralatrexate-Based Combination in T-Cell Malignancies

Published OnlineFirst May 25, 2010; DOI: 10.1158/1078-0432.CCR-10-0671

of 29% in patients with very heavily treated disease.4

Subgroup analyses have shown that the activity of pra-latrexate was equivalent irrespective of the amount ofprior therapy, age, prior autologous stem cell transplant,or subtype of PTCL. Pralatrexate belongs to the class offolate analogues known as 10-deazaaminopterins, whichhave shown markedly superior antitumor effects com-pared with methotrexate in severe combined immunode-ficient (SCID)-beige xenograft models of lymphoma (11,12). Pralatrexate was designed to have greater affinitythan methotrexate for the natural folate and antifolateprincipal transporters reduced folate carrier-1 (RFC-1;an oncofetal protein) and folyl-polyglutamyl synthase,leading to enhanced intracellular accumulation andpolyglutamylation in tumor cells.Recent preclinical studies by our group have established

that pralatrexate synergizes with other agents active inPTCL, including gemcitabine. These preclinical data (13)have been confirmed in a phase I clinical trial exploringthe schedule-dependent activity of pralatrexate and gemci-tabine in patients with non–Hodgkin's lymphoma. In ad-dition to PTCL, pralatrexate seems to be very active inpatients with drug-resistant cutaneous T-cell lymphoma(CTCL), producing an overall response rate of 55% in pa-tients who received a median of 6 prior regimens (range,1-25; ref. 14). Collectively, these data suggest that prala-trexate exhibits marked activity across different subtypesof T-cell neoplasms and could combine favorably with ahost of other agents known to be active in T-cell lympho-ma. The observation that bortezomib has now beenshown to exhibit significant activity across both CTCLand PTCL raises the interesting prospect that these agentscould also be combined in an effort to define a new plat-form for these diseases that is not CHOP dependent. Forexample, Zinzani et al. (15) have reported that bortezo-mib produces an ORR of 67% in patients with relapsedor refractory CTCL and PTCL, including a complete re-sponse rate of 17%.Given the relatively poor prognosis of patients with T-

cell malignancies and the relatively poor outcomes asso-ciated with conventional chemotherapy regimens such asCHOP, there is now a strong rationale to begin to com-bine and integrate these targeted agents into novel com-bination treatment regimens for patients with T-cellmalignancies that are not CHOP based. Based on thisrationale and the available clinical data, we sought toexplore the preclinical activity of pralatrexate and borte-zomib in models of T-cell lymphoma.

Materials and Methods

Cells and cell linesH9 (16) and HH (17) are CTCL cell lines, obtained

from the American Type Culture Collection. P12, PF382,

4 O'Connor OA, Pro B, Pinter-Brown L, et al. Pralatrexate in patients withrelapsed or refractory peripheral T-cell lymphoma (PTCL). Results from thepivotal PROPEL study. JCO, submitted March 2010.

www.aacrjournals.org

Researcon Julyclincancerres.aacrjournals.org Downloaded from

and CEM (18) are T-acute lymphoblastic leukemia (T-ALL) lines resistant to γ-secretase inhibitors, and KOKT-K1 (19), DND-41, and HPB-ALL (20) are T-ALL linessensitive to γ-secretase inhibitors. All leukemic cell lineswere provided by the laboratory of Dr. Adolfo Ferrando(Columbia University, New York, NY). Peripheral bloodmononuclear cells (PBMC) from healthy donors werepurchased from Allcells. All cell lines were grown aspreviously described (21).

MaterialsAll reagents for Western blotting were obtained from

Bio-Rad Laboratories and Pierce Biotechnology, Inc.;DMSO was obtained from Sigma. Pralatrexate was fromAllos Therapeutics, and bortezomib from Millennium.

Cytotoxicity assaysFor all in vitro assays, cells were counted and resus-

pended at an approximate concentration of 3 × 105 perwell in a 96-well plate (Becton Dickinson Labware) andincubated at 37°C in a 5% CO2 humidified incubatorfor up to 72 hours. Pralatrexate was added at concentra-tions from 100 pmol/L up to 200 μmol/L, whereas borte-zomib was tested at concentrations from 1 to 100 nmol/Lto determine growth inhibition curves for all cell lines. Incombination experiments, pralatrexate was added at con-centrations of 2 to 5.5 nmol/L, and bortezomib at 3 to6 nmol/L. These concentrations were selected to approxi-mate the IC25-50 (inhibitory concentration of 25-50% cellsfor each drug) for up to 72 hours. Following incubation at37°C in a 5% CO2 humidified incubator, 100 μL fromeach well were transferred to a 96-well opaque-walledplate; CellTiter-Glo reagent (Promega Corporation) wasused according to the manufacturer's instruction. Theplates were allowed to incubate at room temperature for10 minutes before recording luminescence with a SynergyHT Multi-Detection Microplate Reader (Biotek Instru-ments, Inc.). Each experiment was done in triplicate andrepeated at least twice.

Flow cytometry for apoptosisH9, HH, P12, and PF382 cells were seeded at a density of

3 × 105/mL and incubated with pralatrexate (2-5.5 nmol/L)and bortezomib (2-6 nmol/L) alone or in combination for48 or 72 hours. Aminimumof 1 × 105 events were acquiredfrom each sample. To quantitate apoptosis, Yo-Pro-1 andpropidium iodide (PI) were used (Vybrant apoptosis assaykit #4, Invitrogen) according to the manufacturer's instruc-tion. The fluorescence signals acquired by a FACSCaliburSystem were resolved by detection in the conventionalFL1 and FL3 channels. Cells were considered early apopto-tic if Yo-Pro-1 positive but PI negative, late apoptotic if Yo-Pro-1 and PI positive, and dead if only PI positive.

Caspase activation assaysH9 cells were seeded at a density of 1 × 106/mL and in-

cubated with pralatrexate and bortezomib alone or incombination at 4 to 6 nmol/L. After incubation for up

Clin Cancer Res; 16(14) July 15, 2010 3649

h. 8, 2020. © 2010 American Association for Cancer

Page 3: Research Pralatrexate Is Synergistic with the Proteasome Inhibitor ... · Pralatrexate Is Synergistic with the Proteasome Inhibitor Bortezomib in In vitro and In vivo Models of T-Cell

Marchi et al.

3650

Published OnlineFirst May 25, 2010; DOI: 10.1158/1078-0432.CCR-10-0671

to 24 hours, aliquots of 300 μL were obtained from eachsample and transferred into Eppendorf tubes. One micro-liter of FITC-IETD-FMK (CaspGLOW Fluorescin ActiveCaspase-8, Biovision) and 1 μL of Red-LEHD-FMK(CaspaseGLOW red Active Caspase-9, Biovision) wereadded to each tube. A negative control, the caspase inhi-bitor Z-VAD-FMK (1 μL/mL), and a positive control, eto-poside (100 nmol/L), were also evaluated in eachexperiment. The protocol followed the manufacturer's in-struction. All data from flow cytometry were analyzed byFlowJo software. Each experiment was done at least in du-plicate and repeated at least twice. Data are presented asaverage ± SD.

Western blot analysisCells were incubated with the same concentrations of

pralatrexate and/or bortezomib used in the apoptosisand caspase assays under normal growth conditions forup to 24 hours. Proteins from total cell lysates were re-solved on 4% to 20% SDS-PAGE and transferred onto ny-lon membranes. Membranes were blocked in PBS, 0.05%Tween 20 containing 5% skim milk powder and were thenprobed overnight with specific primary antibodies. Anti-bodies were detected with the corresponding horseradishperoxidase–linked secondary antibodies. Blots were devel-oped using SuperSignal West Pico chemiluminescent sub-strate detection reagents. The membranes were exposed toX-ray films for various time intervals. The images were cap-tured with a GS-800 calibrated densitometer (Bio-Rad),and the ratios were quantified by densitometric analyseswithin the linear range of each captured signal. The follow-ing monoclonal and polyclonal antibodies were used:p27, p100/p52, and acetylated histone H3 (from CellSignaling Technology), Noxa (Imgenex Corp.), RFC-1(SLC19A1 antibody from abcam), and acetylated α-tubulin(from Sigma).

Mouse xenograft modelsIn vivo experimentswere done as follows: 5- to 7-week-old

SCID-beige mice (Charles River Laboratories) were injecteds.c. with 20 × 106 HH cells on the flank. When tumor vo-lumes approached 50 mm3, mice were separated into treat-ment groups of 9 to 10 mice each. Tumors were assessedusing the two largest perpendicular axes (l, length;w, width)measured with standard calipers. Tumor volume was calcu-lated using the formula (4/3)r3, where r = (l +w) / 4. Tumor-bearing mice were assessed for weight loss and tumorvolume at least twice weekly. Animals were sacrificedwhen one-dimensional tumor diameter exceeded 2.0 cmor after loss of >10% body weight in accordance withinstitutional guidelines. Complete response (CR) was de-fined as nonpalpable tumor. Both pralatrexate and borte-zomib were given by i.p. injection. In the combinationexperiments, pralatrexate was administered at 15 mg/kg(1/4 of the maximum tolerated dose) on days 1, 4, 8,and 11; bortezomib (B) was administered at 0.5 mg/kgon days 1, 4, 8, and 11. Control groups were treated withthe vehicle solution alone.

Clin Cancer Res; 16(14) July 15, 2010

Researcon Julyclincancerres.aacrjournals.org Downloaded from

Statistical analysisFor the different in vitro experimental groups, permuta-

tion tests were done to determine whether any of the ex-perimental groups was superior to a control group. Theanalysis entailed comparing groups based on repetitions(typically 3) using ANOVA after a normalizing transforma-tion. Because multiple hypotheses were simultaneouslytested, all P values were adjusted using the Dunnett meth-od (22). For each cell line, the IC50 (refs. 23, 24) and thedrug-drug interaction in terms of synergism, additivity, orantagonism were computed using the Calcusyn software(Biosoft; ref. 25) for the cytotoxicity assay. A combinationindex (CI) of <1 defines synergism, CI = 1 additivity, andCI > 1 antagonism. For the apoptosis data, the drug-druginteractions were computed using the relative risk ratio(RRR) analysis (GraphPad; http://www.graphpad.com)with RRR < 1 defining synergism, RRR = 1 additivity,and RRR > 1 antagonism. For the mouse experiments,the tumor volumes and areas under the curve were logtransformed and evaluated using ANOVA for four-waycomparison and Wilcoxon test for pairwise comparisons.Median absolute deviation was used as a measurement ofvariability.

Results

Pralatrexate interacts synergistically with bortezomibin T-cell lymphoma linesThe IC50 values for pralatrexate alone at 48 and 72 hours

were generally in the low nanomolar range. Specifically,these values (nmol/L) at 48 and 72 hours, respectively,are as follows: H9, 1.1 and 2.5; P12, 1.7 and 2.4; CEM,3.2 and 4.2; PF-382, 5.5 and 2.7; KOPT-K1, 1 and 1.7;DND-41, 97.4 and 1.2; and HPB-ALL, 247.8 and 0.77.HH was relatively resistant after 48 hours of exposure,with the IC50 at 72 hours being 2.8 nmol/L (Table 1;Fig. 1A). Two CTCL lines (H9 and HH) and two T-ALL celllines (P12 and PF-382) were selected for combination ex-periments with bortezomib. Generally, the range of IC50sfor bortezomib in these cells was more similar. Forexample, the IC50s for bortezomib alone at 48 hours wereas follows: H9, 6 nmol/L; HH, 3.9 nmol/L (after 72 hoursof incubation); P12, 4.7 nmol/L; and PF382, 2.2 nmol/L.In the cytotoxicity assays, the combination of pralatrexateand bortezomib showed synergism in all cell lines: H9,CI ≤ 0.38; P12, CI ≤ 0.513; PF382, CI ≤ 0.352; and HH,CI ≤ 0.4 (Fig. 1A and B). These data support the mathe-matical synergy of these two agents in vitro.

Pralatrexate plus bortezomib enhances apoptosis inT-cell lymphomaTreatment with the combination of pralatrexate and

bortezomib for 48 hours exhibited significant inductionof apoptosis in H9, P12, PF382, and HH cell lines. For ex-ample, treatment of the CTCL line H9 with bortezomibalone at 5 nmol/L induced apoptosis in up to 28.6% ofthe population. When H9 cells were treated with pralatrex-ate at 3 or 4 nmol/L (IC50-60), 46% and 58% of the cells,

Clinical Cancer Research

h. 8, 2020. © 2010 American Association for Cancer

Page 4: Research Pralatrexate Is Synergistic with the Proteasome Inhibitor ... · Pralatrexate Is Synergistic with the Proteasome Inhibitor Bortezomib in In vitro and In vivo Models of T-Cell

1

Pralatrexate-Based Combination in T-Cell Malignancies

Published OnlineFirst May 25, 2010; DOI: 10.1158/1078-0432.CCR-10-0671

respectively, became apoptotic. The combination of prala-trexate (3 or 4 nmol/L) and bortezomib (5 nmol/L) pro-duced more than 70% induction of apoptosis, and theRRR for the combination of pralatrexate (3 nmol/L) andbortezomib was ≤0.8. Analysis of various pralatrexateand bortezomib concentrations revealed that the RRRseemed to be more sensitive to the relative concentrationof bortezomib. For example, 53% of H9 cells were apopto-tic when treated with 6 nmol/L bortezomib, whereas 46%and 58% of H9 cells were apoptotic when treated with 3and 4 nmol/L of pralatrexate, respectively. Treatment ofH9 with 6 nmol/L bortezomib and the highest concentra-tions of pralatrexate (4 nmol/L) produced nearly 90% in-duction of apoptosis. Furthermore, the RRR for the lattercombination was <0.6. Importantly, all combinationsstudied were statistically significant, favoring the combina-tion over the control and any single-agent exposure (Fig.2A; Table 2). In one of the more resistant cell lines, theCTCL HH line, the combination was superior to any singleagent. For example, treatment of HH with the combinationof pralatrexate (3 nmol/L) and bortezomib (2 nmol/L) for72 hours induced apoptosis in 53%, compared with 39%(pralatrexate) and 32% (bortezomib) of cells treated witheither drug alone (Fig. 2B; Table 2). Again, the RRR analysisconfirmed the synergistic interaction between the twoagents (RRR ≤0.66). Overall, these observations held upover the entire panel of T-cell malignancies screened. Whenthe T-ALL line P12 was treated with 5 nmol/L bortezomibor with 2 or 3 nmol/L pralatrexate alone, 57%, 25%, and49% of the cells became apoptotic, respectively. The com-bination of pralatrexate and bortezomib induced apopto-sis in 75% and 87% of cells, with more apoptosis beingobserved at the higher concentration of pralatrexate. TheRRRs of the combinations were ≤0.8 and ≤0.7, respectively,and the P values were statistically significant relative toboth the controls and the single-agent exposures (≤0.001;Fig. 3A; Table 2). When the T-ALL line PF382 was treated

www.aacrjournals.org

Researcon Julyclincancerres.aacrjournals.org Downloaded from

with 3 nmol/L bortezomib, 5.5 nmol/L pralatrexate, or thecombination, 72%, 65%, and 94% of the cells became ap-optotic, respectively. The RRR of the combination was 0.6(Fig. 3B; Table 2). These data support the observationsmade in the cytotoxicity experiments, confirming the syn-ergistic interaction of pralatrexate and bortezomib in T-celllymphoid malignancies.

Pralatrexate enhances the activity of bortezomib ininducing caspase-8 and caspase-9 activationTo evaluate the effect of the combination on the extrinsic

and intrinsic arms of the apoptositc cascade, we exploredthe effect of pralatrexate and bortezomib on the activationof caspase-8 and caspase-9 in the CTCL line H9. Incubationof H9 with pralatrexate alone at 4 nmol/L, bortezomibalone at 6 nmol/L, and the combination for 24 hours re-sulted in activation of caspase-8 in 14%, 10%, and 37%of cells, respectively. Similarly, activation of caspase-9was seen in 13%, 5%, and 39% of H9 cells, respectively(Fig. 4A). These data are again concordant with the obser-vations made in the cytotoxicity and apoptosis assays de-scribed above and confirmed that caspase activation withthe combination was also synergistic.

Pralatrexate does not increase the toxicity ofbortezomib in PMBCs from healthy donorsTo assess the toxicity of the combination in normal

PBMNC, we also investigated the induction of apoptosisof PBMCs from healthy donors. We explored the effectsof bortezomib at 3 and 5 nmol/L, pralatrexate at 2, 3,and 4 nmol/L, and the combination at all the possible per-mutations after 48 hours of exposure. Importantly, allcombination groups did not show any significant increasein apoptosis compared with the bortezomib-alone groupor between the combination group and pralatrexate (P >0.8). Pralatrexate also did not exhibit any increased cyto-toxicity relative to the untreated controls (Fig. 4B).

Table 1. Cytotoxicity of pralatrexate and methotrexate in eight T-cell lymphoma and leukemia cell lines

Cell line

Subtype Pralatrexate (nmol/L)

h. 8, 2020. © 2010 Am

Methotrexate (nmol/L)

IC50, 48 h (c.i.)

IC50, 72 h (c.i.) IC50, 48 h (c.i.)

Clin Cancer Res; 1

erican Association

IC50, 72 h (c.i.)

H9

CTCL 1.13 (0.1-8.9) 2.54 (0.5-12.9) 4.8 (2.5-9) 4.41 (2.9-6.7) HH N/A 2.8 (0.3-29.2) N/A 18.9 (0.09-3,538) P12 T-ALL (resistant to GSI) 1.73 (1.7-1.74) 2.43 (1.6-3.6) 4 (3.5-4.7) 16.8 (15.4-18.4) CEM 3.23 (1.6-6.5) 4.16 (3.8-4.6) 3.4 (2.3-5) 16.2 (13.1-20) PF382 5.47 (4.3-6 9) 2.72 (1.9-3 9) 21.6 (17-27 3) 19 (13.3-26.1) KOPT-K1 T-ALL (sensitive to GSI) 1 (0.8-1.3) 1.69 (1.3-2.2) 14.1 (13.2-14.5) 10 (7.3-13.8) DND-41 97.37 (25.8-368.2) 1.21 (1-1.3) 88.3 (77-458) 6.9 (2.4-19.7) HPB-ALL 247.78 (60.6-1,012 9) 0.77 (0.5-1.1) 56.7 (45.8-536) 11.1 (2.7-46.6) 1

NOTE: The table presents the IC50 values with the confidence interval for pralatrexate and methotrexate after 48 and 72 h ofincubation. In most of the cell lines, the IC50 values at 48 and 72 h of incubation are in the low nanomolar range. There is minimalcytotoxic activity appreciated at 24 h in all the cell lines studied.Abbreviations: c.i., confidence interval; GSI, γ-secretase inhibitor.

6(14) July 15, 2010 3651

for Cancer

Page 5: Research Pralatrexate Is Synergistic with the Proteasome Inhibitor ... · Pralatrexate Is Synergistic with the Proteasome Inhibitor Bortezomib in In vitro and In vivo Models of T-Cell

Marchi et al.

3652

Published OnlineFirst May 25, 2010; DOI: 10.1158/1078-0432.CCR-10-0671

Pralatrexate and bortezomib influence the expressionof proteins belonging to different pathways in theintracellular networkIn an effort to identify discrete pathways influenced by

the combination not markedly influenced by either drugalone, broad probes of the cell cycle and apoptotic ma-chinery were analyzed by Western blot. Treatment of theH9 and P12 cell lines with bortezomib and pralatrexaterevealed changes in a host of proteins known to be in-

Clin Cancer Res; 16(14) July 15, 2010

Researcon Julyclincancerres.aacrjournals.org Downloaded from

volved in cell cycle control, apoptosis, chromatin remodel-ing (histone acetylation), and cellular transport. Usingprecisely the same concentrations that effected synergy inthe in vitro assays, we identified significant changes in p27,NOXA, HH3, and RCF-1 in H9 after exposure to 5 nmol/Lbortezomib, 3 nmol/L pralatrexate, and the combination(after 16, 24, and 48 hours of exposure). These data (Sup-plementary Data A) showed that bortezomib increased theaccumulation of p27, NOXA, and HH3 acetylation in the

Fig. 1. A, cytotoxicity of pralatrexate in four T-cell lymphoma and leukemia cell lines. The plots graphically show the cytotoxicity curves for pralatrexateafter 48 h of incubation in the cell lines selected for the combination studies. B, pralatrexate interacts synergistically with bortezomib in T-cell leukemia andlymphoma cell lines. Isobologram analysis of the interaction between pralatrexate and bortezomib in H9, HH, P12, and PF382. All data points below the redline define synergistic interaction between the two drugs (CI < 1).

Clinical Cancer Research

h. 8, 2020. © 2010 American Association for Cancer

Page 6: Research Pralatrexate Is Synergistic with the Proteasome Inhibitor ... · Pralatrexate Is Synergistic with the Proteasome Inhibitor Bortezomib in In vitro and In vivo Models of T-Cell

Pralatrexate-Based Combination in T-Cell Malignancies

Published OnlineFirst May 25, 2010; DOI: 10.1158/1078-0432.CCR-10-0671

CTCL cell line H9, as seen in the bortezomib-alone andthe bortezomib plus pralatrexate combination groups.Pralatrexate alone had little to no effect on the accumula-tion of NOXA and HH3 acetylation, while clearly increas-ing p27 levels similar to that seen in the bortezomib-alonegroup. Moreover, pralatrexate seemed to increase the accu-mulation of RFC-1 in these cells, suggesting that the expo-sure to pralatrexate may induce an increase in its own

www.aacrjournals.org

Researcon Julyclincancerres.aacrjournals.org Downloaded from

intracellular accumulation. Supplementary Data B revealsthe expression of p27, NOXA, and HH3 in the T-ALL cellline P12 after exposure to 5 nmol/L bortezomib, 2 and3 nmol/L pralatrexate, and the possible combinations.These observations were similar to those in the H9 cellline, except that the increase in p27 seemed to be smaller.Whereas these drugs are likely to produce a number of ef-fects on these cell lines at both the transcriptional and the

Fig. 2. Enhanced apoptosis after treatment with pralatrexate plus bortezomib in CTCL (H9 and HH). A, H9 cells were incubated with pralatrexate (P),bortezomib (B), and their combination from 3 to 4 nmol/L and from 5 to 6 nmol/L, respectively, for 48 h (P ≤ 0.01). control Exp., expected. B, HH cells wereincubated with 3 nmol/L pralatrexate, 2 nmol/L bortezomib, and their combination for 72 h. Etop, etoposide. The histograms show the results after therelative risk (RR) analysis. All the figures also show the “expected apoptosis value” if the interaction between pralatrexate and bortezomib were just additive.

Table 2. Range of apoptosis induction for the single drug compared with the combination of pralatrexateand bortezomib in the CTCL and T-ALL lines studied

Cell line

Bortezomib (% apoptosis) Pralatrexate (% apoptosis)

h. 8, 2020. © 2010

Pralatrexate + bortezomib(% apoptosis − % expected)

Clin Cancer Res; 16(14) July 1

American Association for Cancer

RRR

H9

5 nmol/L (26-31%) 3 nmol/L (40-51%) 63-73% (58%) 0.7 6 nmol/L (43-63%) 3 nmol/L (40-51%) 73-92% (72%) 0.7 6 nmol/L (43-63%) 4 nmol/L (51-63%) 83-94% (78%) 0.6

HH

2 nmol/L (28-34%) 3 nmol/L (38-41%) 50-55% (46%) 0.6 P12 5 nmol/L (50-67%) 2 nmol/L (21-27%) 65-81% (58%) 0.8

5 nmol/L (50-67%)

3 nmol/L (42-55%) 84-92% (76%) 0.7 PF382 3 nmol/L (60-85%) 5.5 nmol/L (64-67%) 91-98% (89%) 0.6

NOTE: The table displays the range of apoptosis induction for the drugs alone or in combination and the RRR value after theanalysis. RRR < 1, synergy.

5, 2010 3653

Page 7: Research Pralatrexate Is Synergistic with the Proteasome Inhibitor ... · Pralatrexate Is Synergistic with the Proteasome Inhibitor Bortezomib in In vitro and In vivo Models of T-Cell

Marchi et al.

3654

Published OnlineFirst May 25, 2010; DOI: 10.1158/1078-0432.CCR-10-0671

translational levels, the survey of these proteins suggeststhat the combination of bortezomib and pralatrexate like-ly exerts its effect by influencing both cell cycle and apo-ptotic pathways.

Pralatrexate enhances the activity of bortezomib in aSCID-beige xenograft modelThe in vivo efficacy of pralatrexate combined with bor-

tezomib was investigated in a xenograft model of CTCL(HH; Supplementary Data B). HH was selected becauseit was the most resistant line based on the in vitro assays.After 30 days from the beginning of the experiment, theresults in the combination group treated with pralatrex-ate at a dose of 15 mg/kg (1/4 of the maximum tolera-ted dose) and bortezomib given on days 1, 4, 8, and 11at a dose of 0.5 mg/kg were statistically significant com-pared with pralatrexate alone (P = 0.002), bortezomibalone (P = 0.001), and the control (P = 0.001; Supple-mentary Data C). Interestingly, CRs were observed onlyin the combination cohort, where 6 of 10 mice experi-enced CR in the combination cohort at day 18, withtwo of those CRs being maintained beyond day 30. Nei-

Clin Cancer Res; 16(14) July 15, 2010

Researcon Julyclincancerres.aacrjournals.org Downloaded from

ther significant weight loss nor death was observed inany of the cohorts. These data support the in vitro experi-ments in establishing the superior efficacy of this combi-nation in T-cell malignancies.

Discussion

PTCLs are, with few exceptions, considered to be aggres-sive diseases with a poor prognosis. During the last fewyears, the proteasome inhibitor bortezomib has been ap-proved by the Food and Drug Administration for the treat-ment of multiple myeloma and mantle cell lymphomaand has been evaluated in a multicenter phase II clinicaltrial in patients with CTCL. Albeit a small study, an ORRof 67% including 2 CRs with a duration of response rang-ing from 7 to 14 months was reported (15). In addition toits single-agent activity, bortezomib has been found tosynergize with innumerable agents, including HDACinhibitors, Bcl-2–targeted agents (26), and conventionalcytotoxic agents (27). Similarly, pralatrexate has beenshown to exhibit marked activity across a panoply of B-and T-cell malignancies with unique activity in a subset

Fig. 3. Pralatrexate and bortezomib synergystically induce apoptosis after 48 h of incubation in T-ALL. A, P12 (T-ALL) cells were incubated with pralatrexateat doses of 2 and 3 nmol/L, bortezomib at a dose of 5 nmol/L, and their combination for 48 h (P ≤ 0.001). B, PF382 (T-ALL) cells were incubated withpralatrexate at a dose of 5.5 nmol/L, bortezomib at a dose of 3 nmol/L, and their combination for 48 h (P ≤ 0.001). All the figures also show the expectedapoptosis value if the interaction between pralatrexate and bortezomib were just additive.

Clinical Cancer Research

h. 8, 2020. © 2010 American Association for Cancer

Page 8: Research Pralatrexate Is Synergistic with the Proteasome Inhibitor ... · Pralatrexate Is Synergistic with the Proteasome Inhibitor Bortezomib in In vitro and In vivo Models of T-Cell

Pralatrexate-Based Combination in T-Cell Malignancies

Published OnlineFirst May 25, 2010; DOI: 10.1158/1078-0432.CCR-10-0671

of patients with T-cell lymphoma (28). While the precisemechanism for how these drugs complement each other islikely to be multifactorial, these agents seem to exhibit asynergistic interaction in T-cell–derived malignanciesbased on these preclinical data.The experiments presented here support the potent activity

of pralatrexate in a variety of T-cell lymphoma and leukemialines. At present, there are no cell lines or xenograft modelsrepresentative of PTCL. As a result, our studies are restrictedto the wider range possible of cell lines of T-cell lineage, in-cluding T-ALL and transformed mycosis fungoides (MF).

www.aacrjournals.org

Researcon Julyclincancerres.aacrjournals.org Downloaded from

The cytotoxicity assays suggested IC50 values in the na-nomolar range for the cutaneous lymphoma lines (H9 andHH) and the T-cell acute lymphoblastic leukemia lines(P12, PF382, and CEM; KOKPT K1, DND-41, and HPB-ALL) for both drugs. We also confirmed in this panel ofT-cell lines that pralatrexate is at least a log more potentthan methotrexate (Table 1). This study showed that theexposure of T-cell lines to even nanomolar concentrationsof pralatrexate and bortezomib produced potent induc-tion of apoptosis in these lines. For all the cell lines stud-ied (H9, P12, PF382, and HH), the drug-drug interaction

Fig. 4. Caspase-8 and caspase-9 activation in CTCL and lack of enhanced toxicity in PBMCs from healthy donors after treatment with pralatrexate ±bortezomib. A, the histograms show the percentage of cells with caspase-8 and caspase-9 activation after treatment with bortezomib (6 nmol/L) and/orpralatrexate (4 nmol/L) after 24 h of incubation. The histogram called “expected” reveals the degree of caspase activation assuming additivity of pralatrexateand bortezomib. The percentage of caspase-8/caspase-9 activation was calculated based on the percent of the cells with caspase activation in theuntretated cells (“0” limit) versus the percentage of cells with caspase activation treated with etoposide (100 nmol/L). B, at the same dilution that showedsynergistic effect, PMBCs were treated with bortezomib at 3 or 5 nmol/L and/or pralatrexate at 2, 3, and 4 nmol/L for 48 h. Each combination group was notsignificantly more cytotoxic than bortezomib given alone (P > 0.87).

Clin Cancer Res; 16(14) July 15, 2010 3655

h. 8, 2020. © 2010 American Association for Cancer

Page 9: Research Pralatrexate Is Synergistic with the Proteasome Inhibitor ... · Pralatrexate Is Synergistic with the Proteasome Inhibitor Bortezomib in In vitro and In vivo Models of T-Cell

Marchi et al.

3656

Published OnlineFirst May 25, 2010; DOI: 10.1158/1078-0432.CCR-10-0671

was described as synergistic based on the calculation of theCI and the RRR. To determine if the two drugs affect theiraction through the extrinsic or intrinsic apoptotic path-ways, flow cytometry was done to analyze the activationof caspase-8– and/or caspase-9–mediated apoptosis.Available published literature has shown that bortezomibitself is able to activate both the extrinsic (caspase-8) andthe intrinsic (caspase-9) apoptotic pathways (29). Interest-ingly, pralatrexate seems to activate the extrinsic and in-trinsic pathways of apoptosis as well. The combinationof these agents was not only synergistic in conventionalcytotoxicty assays but also seems to synergistically activateboth pathways of apoptosis. To establish that the synergyappreciated in the tumor cell lines did not extend to nor-mal lymphocytes, we explored the combination in PBMCsfrom healthy donors. The data showed that essentially thesame and even higher concentrations of pralatrexate andbortezomib shown to be synergistic in tumor cell lineshad no combined effect on normal PBMCs. The effect ofthe combination was not statistically more toxic againstPBMCs than either drug alone (P > 0.8), although theconcentration of bortezomib drove the toxicity againstPBMCs.A xenograft experiment of CTCL (HH) in SCID-beige

mice with bortezomib combined with pralatrexate wasdone and showed a statistically significant advantage forthe combination compared with the single agents andthe control after 18 days (P ≤ 0.002). Bortezomib was giv-en at a dose of 0.5 mg/kg and pralatrexate was adminis-tered at 15 mg/kg (1/4 of the maximum tolerated dose)on days 1, 4, 8, and 11 to maximize activity while mini-mizing toxicity (13). These doses were selected based onpreliminary animal toxicology studies which establishedthat higher doses of pralatrexate (30 and 60 mg/kg) incombination with bortezomib (0.5 mg/kg) were not toler-ated. Interestingly, CRs (6 of 10 animals) were only ob-served in the combination cohort. No significant toxicity(weight loss in excess of 10% or toxic death) was seen inany of the dose cohorts. The combination of bortezomiband pralatrexate at 1/4 of the maximum tolerated doseproduced significant activity with minimal toxicity in thexenograft model studied.Proteasome inhibition is known to affect a diverse array

of intracellular signaling pathways, including effects onNF-κB (impaired degradation of IB), cell cycle regulation(accumulation of p21/p27), modulation of Bcl-2 familymembers (upregulation of proapoptotic and BH3-onlymembers), and accumulation of p53 (30–32). In tworecent articles by our group (26, 27), AT-101, a small-molecule inhibitor of Bcl-2, Bcl-XL, and Mcl-1, potently sy-nergized with the conventional agents (cyclophosphamideand rituximab) and the irreversible proteasome inhibitorcarfilzomib in in vitro and in vivo models of mantle celllymphoma and diffuse large B-cell lymphoma. In addi-tion, the BH3-only mimetic ABT-737 synergized with theproteasome inhibitor bortezomib, which was mechanisti-cally attributed to an effect on both the relative balancebetween BAX/BAK binding to antiapoptotic proteins

Clin Cancer Res; 16(14) July 15, 2010

Researcon Julyclincancerres.aacrjournals.org Downloaded from

(Bcl-2, Bcl-XL, and Bcl-w) mediated by the BH3-only mi-metic and marked increases in the BH3-only proteinsNOXA and PUMA mediated by bortezomib. It is likelythat, whatever the ultimate mechanism of apoptosisinduced by pralatrexate, the lowering of the apoptoticthreshold with bortezomib serves to sensitize the T-celllines to the antifol. Despite the multitude of hypothesesabout the mechanism of action of proteasome inhibitors,it is difficult to be dogmatic regarding the relative contri-butions of one mechanism of action over another.Furthermore, while pralatrexate has marked activity in

patients with relatively drug-resistant T-cell lymphoma,the biological basis for the activity seen in T- over B-celllymphomas remains unclear. In an effort to identifyunique biomarkers of activity that might account for thesynergy between these two agents, a series of Western blotanalyses focused on proteins involved in cell cycle regula-tion and apoptosis were evaluated. Although there wereno significant changes in most of these potential biomar-kers, these data suggested that there were four proteins in-cluding p27, NOXA, RFC-1, and HH3 that seemed to befavorably modulated by these drugs used as single agentsand in combination. Preliminary data have suggested thatpralatrexate might actually lead to an increase in the ex-pression of RFC-1 on the target cells, potentially leadingto an “autocrine” increase in the intracellular uptake ofpralatrexate. Although there was no evidence that bortezo-mib had any effect on RFC-1, these data support the ob-servation that pralatrexate, at very low concentrations, maybe able to induce the expression of its own transporter.Pralatrexate treatment also leads to an accumulation ofp27 (likely cell cycle arrest effect) seen predominantly inH9 cells, whereas bortezomib increases p27, NOXA, andHH3. Thus, the combination of pralatrexate and bortezo-mib leads to augmented accumulation of RFC-1, p27,NOXA, and, surprisingly, HH3.Increase in NOXA is emerging as an interesting potential

biomarker of effect for proteasome inhibitors. Its accumu-lation following exposure to combination therapy may ex-plain potential synergistic effects, although admittedly, ithas not shed light on the important upstream pathways.NOXA belongs to the group of Bcl-2 homology domain3 (BH3) only proteins and seems to be crucial in fine-tuning cell death decisions by targeting the prosurvivalmolecule Mcl-1 for proteasomal degradation (33). Thisevent seems to be critical for cell death induction in lym-phoma cells treated with bortezomib and the combina-tion. Pralatrexate itself has been shown to selectivelyinduce apoptosis in human T-cell lymphotrophic virus-1–infected adult T-cell lymphoma/leukemia Pautrier mi-croabcesses from a patient treated with a single dose ofdrug (34). Biopsies of the patient's skin revealed selectivecaspase activation and apoptosis only in the malignantcells and not in the normal surrounding keratinocytes. Al-though NOXA was not specifically evaluated in the tissuebiopsies, these important observations confirm in a pa-tient that pralatrexate seems to induce apoptosis via cas-pase activation selectively in malignant T cells.

Clinical Cancer Research

h. 8, 2020. © 2010 American Association for Cancer

Page 10: Research Pralatrexate Is Synergistic with the Proteasome Inhibitor ... · Pralatrexate Is Synergistic with the Proteasome Inhibitor Bortezomib in In vitro and In vivo Models of T-Cell

Pralatrexate-Based Combination in T-Cell Malignancies

Published OnlineFirst May 25, 2010; DOI: 10.1158/1078-0432.CCR-10-0671

The increase in HH3 acetylation is surprising, and sug-gests that bortezomibmay have some properties as aHDACinhibitor. Previous data by Marcucci et al. have establishedthat bortezomib has an effect as a hypomethylating agent,and there is emerging data that hypomethylating agentsmay also increase histone acetylation (35). The observationof course raises the suspicion that this doublet of agentscould be combined with HDAC inhibitors, which also havesignificant activity in T-cell lymphomas, but it does not pro-vide specific insights into how this contributes to the cellkill noted.An emerging number of studies are beginning to con-

verge on the idea that proteasome inhibitors like bortezo-mib may mediate their effects through NOXA and p27 inT-cell malignancies. In one report by Ri et al. (36), bortezo-mib induced upregulation of NOXA at both the transcrip-tional and protein levels in a p53-independent manner inmature T cells (CTCL andadult T-cell lymphoma/leukemia).Repression of NOXA by small interfering RNA rescued theCTCL cells. These data confirmed that the time-dependentbinding of NOXA toMcl-1 led to loss of the transmembranemitochondrial potential, an observation we have similarlyvalidated inmantle cell lymphoma as well. These data, cou-pled with the recent observation by Heider et al. (37) thatboth HDAC inhibitor and bortezomib led to the upregula-tion of p21 and p27 in CTCL cells, which synergistically in-duced apoptosis in these cells, are starting to suggest apossible mechanism for these drugs in T-cell malignancies.Although the a priori rationale for combining drugs ac-

tive in T-cell malignances is at the moment largely focusedon agents with clinically documented single-agent activity,it is becoming increasing clear that important themes inthe underlying molecular pharmacology are emerging. Infact, it is likely that these themes appreciated at the molec-ular level and will become the immediate rationale for

www.aacrjournals.org

Researcon Julyclincancerres.aacrjournals.org Downloaded from

how to combine select classes of agents where other apriori rationales are limited. A convergence of data aroundthe reproducible observation on the modulation of p21/p27, NOXA, Mcl-2, and, unique to pralatrexate, RFC-1may begin to provide insights into potential biomarkersof activity and possibly response. Preclinical and clinicalstudies are now under way to evaluate how best to com-bine these agents in an effort to develop radically newplatforms that are not CHOP-centric for T-cell lymphoma.At the moment, these efforts are oriented toward under-standing the drug:drug interactions between pralatrexate,proteasome inhibitors, HDAC inhibitors, gemcitabine,forodesine, and Bcl-2–targeted agents. Phase I clinical trialsare now being designed to determine the dose-limitingtoxicity and maximum tolerated dose of pralatrexate givenin combination with proteasome and HDAC inhibitors.Collectively, both these preclinical and available clinicaldata suggest that innovative and highly effective plat-forms can be developed for patients with aggressive T-celllymphoma.

Disclosure of Potential Conflicts of Interest

O.A. O'Connor: commercial research grant, Allos Therapeutics, Inc.

Acknowledgments

We thank Adolfo Ferrando for providing the T-cell leukemia lines weused in the in vitro experiments.

Grant Support

Food and Drug Administration Orphan Product R01 Award(FD0003498-01; O.A. O'Connor).

Received 03/23/2010; revised 04/28/2010; accepted 05/19/2010;published OnlineFirst 05/25/2010.

References

1. Rudiger T, Weisenburger DD, Anderson JR, et al. Peripheral T-cell

lymphoma (excluding anaplastic large-cell lymphoma): results fromnon Hodgkin's Lymphoma Classification Project. Ann Oncol 2002;13:140–9.

2. Gressin R, Peoch M, Deconick E, et al. The VIP-ABVD regiment is notsuperior to the CHOP 21 for the treatment of non epidermotropic pe-ripheral T cell lymphoma. Final results of the LTP95 protocol of theGOELAMS [Abstract]. Blood 2006;108:Abstract #2464.

3. Escalón MP, Liu NS, Yang Y, et al. Prognostic factors and treatmentof patients with T-cell non-Hodgkin lymphomas: the M.D. AndersonCancer Center experience. Cancer 2005;103:2091–8.

4. Marchi E, Alinari L, Tani M, et al. Gemcitabine as frontline treatmentfor cutaneous T-cell lymphoma: phase II study of 32 patients. Cancer2005;104:2437–41.

5. Zinzani PL, Baliva G, Magagnoli M, et al. Gemcitabine treatment inpretreated cutaneous T-cell lymphoma: experience in 44 patients.J Clin Oncol 2000;18:2603–6.

6. Arkenau HT, Chong G, Cunningham D, et al. Gemcitabine, cisplatinand methylprednisolone for the treatment of patients with peripheralT-cell lymphoma: the Royal Marsden Hospital experience. Haemato-logica 2007;92:271–2.

7. Spencer A, Reed K, Arthur C. Pilot study of an outpatient-based ap-

proach for advanced lymphoma using vinorelbine, gemcitabine, andfilgrastim. Intern Med J 2007;37:760–6.

8. Enblad G, Hagberg H, Erlanson M, et al. A pilot study of alemtuzu-mab (anti-CD52 monoclonal antibody) therapy for patients with re-lapsed or chemotherapy-refractory peripheral T-cell lymphomas.Blood 2004;103:2920–4.

9. Moon J, Kim J, Sohn D, et al. Alemtuzumab plus CHOP as front-linechemotherapy for patients with peripheral T-cell lymphomas[abstract]. J Clin Oncol 2007;25:Abstract #8069.

10. Prince HM, Bishton MJ, Harrison SJ. Clinical studies of histone dea-cetylase inhibitors. Clin Cancer Res 2009;15:3958–69.

11. Sirotnak FM, DeGraw JI, Schmid FA, Goutas LJ, Moccio DM. Newfolate analogs of the 10-deaza-aminopterin series. Basis for thestructural design biochemical and pharmacologic properties. CancerChemother Pharmacol 1984;12:18–25.

12. Wang ES, O'Connor OA, She Y, Zelenetz AD, Sirotnak FM, Moore MA.Activity of a novel anti-folate (PDX, 10-propargyl10-deazaaminopterin)against human lymphoma is superior to metotrexate and correlateswith tumor RFC-1 gene expression. Leuk Lymphoma 2003;44:1027–35.

13. Toner LE, Vrhovac R, Smith EA, et al. The schedule-dependenteffects of the novel antifolate pralatrexate and gemcitabine are

Clin Cancer Res; 16(14) July 15, 2010 3657

h. 8, 2020. © 2010 American Association for Cancer

Page 11: Research Pralatrexate Is Synergistic with the Proteasome Inhibitor ... · Pralatrexate Is Synergistic with the Proteasome Inhibitor Bortezomib in In vitro and In vivo Models of T-Cell

Marchi et al.

3658

Published OnlineFirst May 25, 2010; DOI: 10.1158/1078-0432.CCR-10-0671

superior to methotrexate and cytarabine in models of human non-Hodgkin's lymphoma. Clin Cancer Res 2006;12:924–32.

14. Horwitz SM, Duvic M, et al. Pralatrexate (PDX) is active in cutaneousT-cell lymphoma (CTCL): results of a multicenter dose finding trial.ASH 2009. 2009;Abstract 919.

15. Zinzani PL, Musuraca G, Tani M, et al. Phase II trial of proteasomeinhibitor bortezomib in patients with relapsed or refractory cutaneousT-cell lymphoma. J Clin Oncol 2007;25:4293–7.

16. Gootenberg JE, Ruscetti FW, Mier JW, Gazdar A, Gallo RC. Humancutaneous T cell lymphoma and leukemia cell lines produce andrespond to T cell growth factor. J Exp Med 1981;154:1403–18.

17. Starkebaum G, Loughran TP, Jr., Waters CA, Ruscetti FW. Establish-ment of an IL-2 independent, human T-cell line possessing only thep70 IL-2 receptor. Int J Cancer 1991;49:246–53.

18. Foley GE, Lazarus H, Farber S, et al. Continuous culture of humanlymphoblasts from peripheral blood of child with acute leukemia.Cancer 1965;18:522–9.

19. Kojika S, Sugita K, Inukai T, et al. Mechanisms of glucocorticoid re-sistance in human leukemic cells: implication of abnormal 90 and 70kDa heat shock proteins. Leukemia 1996;10:994–9.

20. Sangster RN, Minowada J, Suciu-Foca N, et al. Rearrangement andexpression of the alpha, beta, and gamma chain T-cell receptorgenes in human thymic leukemia cells and functional T cells. J ExpMed 1986;163:1491–508.

21. O'Connor OA, Smith EA, Toner LE, et al. The combination of the pro-teasome inhibitor bortezomib and the bcl-2 antisense molecule ob-limersen sensitizes human B-cell lymphomas to cyclophosphamide.Clin Cancer Res 2006;12:2902–11.

22. Hockberg Y, Tamhane AC. Multiple comparison procedures. NewYork (NY): Wiley; 1987, p. 1–450.

23. Chou TC. Relationship between inhibition constants and fractionalinhibitions in enzyme-catalyzed reactions with different numbers ofreactants, different reaction mechanisms, and different types of me-chanisms of inhibition. Mol Pharmacol 1974;10:235–47.

24. Chou TC. Derivation and properties of Michaelis-Menten type and Hilltype equations for reference ligands. J Theoret Biol 1976;39:253–76.

25. Zhao L,Wientjes G, Au JLS. Evaluation of combination chemotherapy:integration of nonlinear regression, curve shift, isobologram, and com-bination index analyses. Clin Cancer Res 2004;10:7994–8004.

26. Paoluzzi L, GonenM, Bhagat G, et al. The BH3-only mimetic ABT-737

Clin Cancer Res; 16(14) July 15, 2010

Researcon Julyclincancerres.aacrjournals.org Downloaded from

synergizes the antineoplastic activity of proteasome inhibitors in lym-phoidmalignancies. Blood 2008;112:2906–16. Epub 2008 Jun 30.

27. Paoluzzi L, Gonen M, Gardner JR, et al. Targeting Bcl-2 family mem-bers with the BH3 mimetic AT-101 markedly enhances the therapeu-tic effects of chemotherapeutic agents in in vitro and in vivo modelsof B-cell lymphoma. Blood 111:5350–8.

28. O'Connor OA, Horwitz S, Hamlin P, et al. Phase II-I-II study of twodifferent doses and schedules of pralatrexate, a high-affinity sub-strate for the reduced folate carrier, in patients with relapsed or re-fractory lymphoma reveals marked activity in T-cell malignancies.J Clin Oncol 2009;27:4357–64. Epub 2009 Aug 3.

29. Zang QL, Wang L, Zhang YW, et al. The proteasome inhibitor borte-zomib interacts synergistically with the histone deacetylase inhibitorsuberoylanilide hydroxamic acid to induce T-leukemia/lymphomacells apoptosis. Leukemia 2009;23:1507–14. Epub 2009 Mar 12.

30. Palombella VJ, Rando OJ, Goldberg AL, Maniatis T. The ubiquitin-proteasome patway is required for processing the NF-κB1 pre-cursor protein and activation of NF-κB. Cell 1994;78:773–85.

31. Adams J. The proteasome: a suitable antineoplastic target. Nat RevCancer 2004;4:349–60.

32. Paoluzzi L, O'Connor OA. Mechanistic rationale and clinical evidencefor the efficacy of the proteasome inhibitors against indolent andmantle cell lymphomas. BioDrugs 2006;20:13–23.

33. Ploner C, Kofler R, Villunger A. Noxa: at the tip of the balance be-tween life and death. Oncogene 2009;27:S84–92.

34. Marneros AG, Grossman ME, Silvers DN, et al. Pralatrexate-inducedtumor cell apoptosis in the epidermis of a patient with HTLV-1 adultT-cell lymphoma/leukemia causing skin erosions. Blood 2009;113:6338–41. Epub 2009 Apr 23.

35. Liu S, Liu Z, Xie Z, et al. Bortezomib induces DNA hypomethyla-tion and silenced gene transcription by interfering with Sp1/NF-κB-dependent DNA methyltransferase activity in acute myeloidleukemia. Blood 2008;111:2364–73.

36. Ri M, Iida S, Ishida T, et al. Bortezomib-induced apoptosis in matureT-cell lymphoma cells partially depends on upregulation of NOXAand functional repression of Mcl-1. Cancer Sci 2008.

37. Heider U, Rademacher J, Lamottke B, et al. Synergistic interaction ofthe histone deacetylase inhibitor SAHA with the proteasome inhibitorbortezomib in cutaneous T cell lymphoma. Eur J Haematol 2009;82:440–9. Epub 2009 Feb 10.

Clinical Cancer Research

h. 8, 2020. © 2010 American Association for Cancer

Page 12: Research Pralatrexate Is Synergistic with the Proteasome Inhibitor ... · Pralatrexate Is Synergistic with the Proteasome Inhibitor Bortezomib in In vitro and In vivo Models of T-Cell

2010;16:3648-3658. Published OnlineFirst May 25, 2010.Clin Cancer Res   Enrica Marchi, Luca Paoluzzi, Luigi Scotto, et al.   Malignancies

Models of T-Cell LymphoidIn vivo and In vitroBortezomib in Pralatrexate Is Synergistic with the Proteasome Inhibitor

  Updated version

  10.1158/1078-0432.CCR-10-0671doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://clincancerres.aacrjournals.org/content/suppl/2010/05/24/1078-0432.CCR-10-0671.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://clincancerres.aacrjournals.org/content/16/14/3648.full#ref-list-1

This article cites 33 articles, 15 of which you can access for free at:

  Citing articles

  http://clincancerres.aacrjournals.org/content/16/14/3648.full#related-urls

This article has been cited by 10 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. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://clincancerres.aacrjournals.org/content/16/14/3648To request permission to re-use all or part of this article, use this link

Research. on July 8, 2020. © 2010 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst May 25, 2010; DOI: 10.1158/1078-0432.CCR-10-0671