induction of potent nk cell-dependent anti-myeloma

16
Induction of potent NK cell-dependent anti-myeloma cytotoxic T cells in response to combined mapatumumab and bortezomib Paul J Neeson 1,2,3, * ,y , Andy K Hsu 1,2,y , Yin R Chen 1,2 , Heloise M Halse 1,2 , Joanna Loh 1,2 , Reece Cordy 1,2 , Kate Fielding 1,2 , Joanne Davis 1,2,3,4 , Josh Noske 1,2 , Alex J Davenport 1,2,4 , Camilla A Lindqvist-Gigg 1,2,4 , Robin Humphreys 5 , Tsin Tai 1,2 , H Miles Prince 2,6 , Joseph A Trapani 1,2,3 , Mark J Smyth 2,7,8 , and David S Ritchie 1,2,3,4,9 1 Cancer Immunology Research; Peter MacCallum Cancer Center; East Melbourne, VIC Australia; 2 Sir Peter MacCallum Department of Oncology; University of Melbourne; Parkville, VIC Australia; 3 The University of Melbourne; Parkville, VIC Australia; 4 The ACRF Translational Research Laboratory; Royal Melbourne Hospital; Parkville, VIC Australia; 5 Human Genome Sciences Inc.; Rockville, MD USA; 6 Department of Cancer Medicine; Peter MacCallum Cancer Center; East Melbourne, VIC Australia; 7 Immunology in Cancer and Infection Laboratory; Queensland Institute of Medical Research; Herston, QLD Australia; 8 School of Medicine; University of Queensland; Herston, Australia; 9 Department of Clinical Hematology and Bone Marrow Transplantation; Royal Melbourne Hospital; Parkville, VIC Australia y Indicates equal rst authorship. Keywords: dendritic cell, multiple myeloma, natural killer cell, proteasome inhibitor, T cell, tumor necrosis factor apoptosis inducing ligand and receptor Abbreviations: anti-myeloma cytotoxic lymphocyte, AMCL; bortezomib, btz; chymotrypsin, Ch; cytotoxic lymphocyte, CTL; dendritic cell, DC; human myeloma cell line, HMCL; low dose bortezomib, LDB; low dose bortezomib and mapatumumab, LDBCMapa; mapatumumab, Mapa; monocyte-derived dendritic cell, MoDC; Multiple myeloma, MM; TRAIL receptor 1, TRAIL-R1; tumor necrosis factor apoptosis inducing ligand, TRAIL There is increasing evidence that some cancer therapies can promote tumor immunogenicity to boost the endogenous antitumor immune response. In this study, we used the novel combination of agonistic anti-TRAIL-R1 antibody (mapatumumab, Mapa) with low dose bortezomib (LDB) for this purpose. The combination induced profound myeloma cell apoptosis, greatly enhanced the uptake of myeloma cell apoptotic bodies by dendritic cell (DC) and induced anti-myeloma cytotoxicity by both CD8 C T cells and NK cells. Cytotoxic lymphocyte expansion was detected within 24 h of commencing therapy and was maximized when myeloma-pulsed DC were co-treated with low dose bortezomib and mapatumumab (LDBCMapa) in the presence of NK cells. This study shows that Mapa has two distinct but connected modes of action against multiple myeloma (MM). First, when combined with LDB, Mapa produced powerful myeloma cell apoptosis; secondly, it promoted DC priming and an NK cell-mediated expansion of anti- myeloma cytotoxic lymphocyte (CTL). Overall, this study indicates that Mapa can be used to drive potent anti-MM immune responses. Introduction MM is incurable by either conventional chemotherapy or recently developed novel therapies including immunomodulatory drugs and proteasome inhibitors. 1,2,3 This underscores the need for the further development of therapies that target all aspects of myeloma control including cell survival, proliferation 4 and anti- myeloma immunity. 5 MM is associated with defects in both cel- lular and humoral immunity 6-9 that must be overcome to opti- mize immunotherapeutic approaches. 10 Ideal anti-MM therapy would be that which delivers maximal myeloma cell apoptosis whilst simultaneously promoting anti-myeloma immune responses by promoting immunogenic cell death. 11 Recent stud- ies in pre-clinical models, 12-16 and promising results in clinical trials 17-20 suggest that antibodies directed against plasma cells including those against SLAM-F7 (Elotuzumab, Bristol-Myers Squibb) or CD38 (Daratumumab, Janssen Biotech) have the potential to significantly improve immunotherapy against MM. The effect of these antibodies at promoting secondary cytotoxic cellular immune responses has not yet been full explored. Mono- clonal antibodies directed against the tumor necrosis factor- related apoptosis-inducing ligand receptor-1 (TRAIL-R1) may also offer an opportunity to induce myeloma cell apoptosis, pro- mote immunogenic cell death and generate cytotoxic cellular immune responses. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL or Apo2L) induces apoptosis by activation of either TRAIL-R1 (DR4) or TRAIL-R2 (DR5). 21-24 Soluble TRAIL *Correspondence to: Paul J Neeson; Email: [email protected] Submitted: 07/18/2014; Revised: 03/30/2015; Accepted: 04/01/2015 http://dx.doi.org/10.1080/2162402X.2015.1038011 www.tandfonline.com e1038011-1 OncoImmunology OncoImmunology 4:9, e1038011; September 2015; © 2015 Taylor & Francis Group, LLC ORIGINAL RESEARCH

Upload: others

Post on 28-Apr-2022

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Induction of potent NK cell-dependent anti-myeloma

Induction of potent NK cell-dependentanti-myeloma cytotoxic T cells in response

to combined mapatumumab and bortezomibPaul J Neeson1,2,3,*,y, Andy K Hsu1,2,y, Yin R Chen1,2, Heloise M Halse1,2, Joanna Loh1,2, Reece Cordy1,2, Kate Fielding1,2,Joanne Davis1,2,3,4, Josh Noske1,2, Alex J Davenport1,2,4, Camilla A Lindqvist-Gigg1,2,4, Robin Humphreys5, Tsin Tai1,2,

H Miles Prince2,6, Joseph A Trapani1,2,3, Mark J Smyth2,7,8, and David S Ritchie1,2,3,4,9

1Cancer Immunology Research; Peter MacCallum Cancer Center; East Melbourne, VIC Australia; 2Sir Peter MacCallum Department of Oncology; University of Melbourne;

Parkville, VIC Australia; 3The University of Melbourne; Parkville, VIC Australia; 4The ACRF Translational Research Laboratory; Royal Melbourne Hospital; Parkville, VIC Australia;5Human Genome Sciences Inc.; Rockville, MD USA; 6Department of Cancer Medicine; Peter MacCallum Cancer Center; East Melbourne, VIC Australia; 7Immunology in Cancer and

Infection Laboratory; Queensland Institute of Medical Research; Herston, QLD Australia; 8School of Medicine; University of Queensland; Herston, Australia;9Department of Clinical Hematology and Bone Marrow Transplantation; Royal Melbourne Hospital; Parkville, VIC Australia

yIndicates equal first authorship.

Keywords: dendritic cell, multiple myeloma, natural killer cell, proteasome inhibitor, T cell, tumor necrosis factor apoptosisinducing ligand and receptor

Abbreviations: anti-myeloma cytotoxic lymphocyte, AMCL; bortezomib, btz; chymotrypsin, Ch; cytotoxic lymphocyte,CTL; dendritic cell, DC; human myeloma cell line, HMCL; low dose bortezomib, LDB; low dose bortezomib and mapatumumab,

LDBCMapa; mapatumumab, Mapa; monocyte-derived dendritic cell, MoDC; Multiple myeloma, MM; TRAIL receptor 1,TRAIL-R1; tumor necrosis factor apoptosis inducing ligand, TRAIL

There is increasing evidence that some cancer therapies can promote tumor immunogenicity to boost theendogenous antitumor immune response. In this study, we used the novel combination of agonistic anti-TRAIL-R1antibody (mapatumumab, Mapa) with low dose bortezomib (LDB) for this purpose. The combination induced profoundmyeloma cell apoptosis, greatly enhanced the uptake of myeloma cell apoptotic bodies by dendritic cell (DC) andinduced anti-myeloma cytotoxicity by both CD8C T cells and NK cells. Cytotoxic lymphocyte expansion was detectedwithin 24 h of commencing therapy and was maximized when myeloma-pulsed DC were co-treated with low dosebortezomib and mapatumumab (LDBCMapa) in the presence of NK cells. This study shows that Mapa has two distinctbut connected modes of action against multiple myeloma (MM). First, when combined with LDB, Mapa producedpowerful myeloma cell apoptosis; secondly, it promoted DC priming and an NK cell-mediated expansion of anti-myeloma cytotoxic lymphocyte (CTL). Overall, this study indicates that Mapa can be used to drive potent anti-MMimmune responses.

Introduction

MM is incurable by either conventional chemotherapy orrecently developed novel therapies including immunomodulatorydrugs and proteasome inhibitors.1,2,3 This underscores the needfor the further development of therapies that target all aspects ofmyeloma control including cell survival, proliferation4 and anti-myeloma immunity.5 MM is associated with defects in both cel-lular and humoral immunity6-9 that must be overcome to opti-mize immunotherapeutic approaches.10 Ideal anti-MM therapywould be that which delivers maximal myeloma cell apoptosiswhilst simultaneously promoting anti-myeloma immuneresponses by promoting immunogenic cell death.11 Recent stud-ies in pre-clinical models,12-16 and promising results in clinical

trials17-20 suggest that antibodies directed against plasma cellsincluding those against SLAM-F7 (Elotuzumab, Bristol-MyersSquibb) or CD38 (Daratumumab, Janssen Biotech) have thepotential to significantly improve immunotherapy against MM.The effect of these antibodies at promoting secondary cytotoxiccellular immune responses has not yet been full explored. Mono-clonal antibodies directed against the tumor necrosis factor-related apoptosis-inducing ligand receptor-1 (TRAIL-R1) mayalso offer an opportunity to induce myeloma cell apoptosis, pro-mote immunogenic cell death and generate cytotoxic cellularimmune responses.

Tumor necrosis factor-related apoptosis-inducing ligand(TRAIL or Apo2L) induces apoptosis by activation of eitherTRAIL-R1 (DR4) or TRAIL-R2 (DR5).21-24 Soluble TRAIL

*Correspondence to: Paul J Neeson; Email: [email protected]: 07/18/2014; Revised: 03/30/2015; Accepted: 04/01/2015http://dx.doi.org/10.1080/2162402X.2015.1038011

www.tandfonline.com e1038011-1OncoImmunology

OncoImmunology 4:9, e1038011; September 2015; © 2015 Taylor & Francis Group, LLCORIGINAL RESEARCH

Page 2: Induction of potent NK cell-dependent anti-myeloma

induces apoptotic cell death in a wide range of tumors, includingMM, but not in normal cells, however its therapeutic utility islimited by its relatively short half-life. The agonistic humanizedIgG1 antibody to TRAIL-R1, Mapa, has been previously shownto induce apoptosis in MM cells;25 however, the immune poten-tiating effect of this antibody has not been previously explored.The combination of TRAIL ligand and bortezomib (btz) hasbeen shown to induce apoptosis in MM cell lines and primaryhuman samples.26 Furthermore, Mapa and Velcade (btz) havebeen used in a clinical trial with no additional effect of the anti-body above that of Velcade.27 This combination however maynot be optimal at the current bortezomib doses used clinically.btz has numerous immunosuppressive effects on the humanimmune system,28-30 contributing to a demonstrable increase inthe incidence of Varicella infections in btz-treated MMpatients.31 Monoclonal antibody combination therapies usingstandard dose btz, may therefore be limited in their ability to pro-mote immune responses by the immunosuppressive effects of btz.

In this study, we addressed this problem by adjusting the doseof btz to below that able to induce single agent myeloma cell apo-ptosis and then explored whether myeloma cells pre-treated withLDB potentiated Mapa-induced apoptosis. We also exploredwhether antibody-mediated tumor cell death induction also pro-moted antitumor immune responses and the mechanismsrequired to do so. Our results indicate that in an NK cell-depen-dent manner, the combination of LDBCMapa therapy inducesmyeloma cell immunogenic death and promotes potent anti-myeloma cytotoxic cell immunity.

Results

TRAIL receptor-1 expressing human multiple myeloma celllines are sensitive to Mapa plus low dose bortezomib

Six MM cell lines, when tested for sensitivity to titrated dosesof btz (Fig. S1), revealed two features. First, MM cell lines weresensitive to btz from a 5 nM dose and above. Secondly, the sub-optimal apoptosis-inducing dose was established for each cell lineand was 1 nM btz for RPMI8226, U266 and NCIH929;2.5 nM btz for OPM-2, JJN-03 and 5 nM for LP-1. This btzdose was used in subsequent studies to stress the MM cell with-out inducing apoptosis, and was termed “LDB”. MM cell lineexpression of TRAIL-R1 was shown to be variable; thus, LP-1,RPMI8226, NCI-H929, U266 and OPM-2 all express TRAIL-R1 (Fig. S2A) constitutively. In contrast, JJN-03 was consis-tently negative for TRAIL-R1. Subsequently, all cell lines weretreated with titrated doses of Mapa, and apoptosis assessed at48 h. The TRAIL-R1 expressing cell lines (OPM-2, U266 andLP-1) showed 60%, 30% and 25% apoptosis in response to10 mg/mL Mapa respectively (Fig. S2B). RPMI8226 is exqui-sitely sensitive to Mapa and undergoes apoptosis (90–100)% inresponse to 0.14 mg/mL Mapa (Fig. S2C). Two cell lines (NCI-H929 and JJN-03) were resistant to Mapa therapy in the doserange 0–10 mg/mL (Fig. S2B). Whilst TRAIL-R1 expressionwas required for the Mapa response, TRAIL-R1 expression leveldid not correlate with the extent of Mapa-induced apoptosis

(Fig. S2D), consistent with that observed previously by Menoretet al. 25 Thus, whilst OPM-2 was sensitive to Mapa therapy,OPM-2 constitutively expressed relatively lower levels of TRAIL-R1 than LP-1, a low responder to Mapa-induced apoptosis.

Subsequently, all six MM cell lines were treated with LDB(established above for each cell line) and titrated doses of Mapato establish whether combination therapy was more effectivethan Mapa alone. RPMI8226, OPM-2 and U266 were all signifi-cantly more sensitive to combination therapy rather than Mapaalone (Fig. 1); however, the Mapa-resistant cell lines (NCI-H929 and JJN-03) were also resistant to LDBCMapa therapy.Interestingly, LP-1 was only partially sensitive to combinationtherapy (30% apoptosis); there was no statistical significance inthe extent of LP-1 apoptosis induction by Mapa alone andLBMCM combination therapy.

We next asked whether sequential treatment (LDB followedby Mapa) would render the myeloma cells more sensitive toLDBCMapa-induced apoptosis. Pre-treatment of human mye-loma cell lines (HMCL) with LDB induced increased TRAIL-R1expression (compared to untreated cells) in LP-1 and OPM-2cells (Fig. 1B). To explore whether this LDB pre-treatmentcould significantly improve LDBCMapa-induced myeloma cellapoptosis, three HMCL’s (LP-1, NCI-H929 and JJN-03) weretested as they previously showed no increase in apoptosis inresponse to the LDBCMapa combination (compared to Mapaalone). Following pre-treatment with LDB, LP-1 cells were sig-nificantly more sensitive to LDBCMapa combination therapythan when treated with combination therapy alone (Fig. 1C). Incontrast, JJN-03 and NCI-H929 cells were resistant to the twophase ‘LDB’ followed by combination therapy (data not shown).

Taken together, this data showed the combinationLDBCMapa treatment effectively induced myeloma cell apopto-sis in vitro and that sequential treatment of myeloma cell lineswith LDB followed by Mapa also shows promising antitumoractivity.

Human dendritic cells treated with low dose bortezomibfunction normally

Human DC viability and function is compromised in vitro bybtz,28,29 this occurs from a 10 nM dose upwards (data notshown). Furthermore, a prior study using a xenotransplant modelof MM32 showed proteasome inhibition occurs in the peripheraltissues and lymphoid organs within 1 h of dosing. Lastly, the btzdose used in clinical practice (1.3 mg/m2/dose i.v.) results in pro-teasome inhibitor activity in the peripheral blood (PB), based onanimal studies and antitumor activity, and this is likely the casein peripheral tissues too. We examined whether using lower dosebtz combined with Mapa would retain anti-myeloma immuneactivity, including DC function. To do this, we performed aseries of studies examining DC function in increasingly stringentdrug conditions. Initially, inhibition of proteasome chymotryp-sin (Ch)-like activity was assessed on LDB, Mapa orLDBCMapa-treated monocyte-derived dendritic cells (MoDCs)(Fig. S3). This experiment showed that LDB and LDBCMapainhibited proteasome Ch-like activity by 10%, whereas Mapaalone had no effect.

e1038011-2 Volume 4 Issue 9OncoImmunology

Page 3: Induction of potent NK cell-dependent anti-myeloma

We then performed complemen-tary studies to examine MoDCphagocytosis of apoptotic myelomacells. First, live video microscopywas used to examine the kineticsand morphology of apoptotic mye-loma (apo-MM) phagocytosis byDCs (Fig. 2A). This study showedthat Apo-MM were phagocytosedby DCs as one large body within20 min of co-culture, and that by40 min the Apo-MM phagosomehad matured (drop in pH reflectedby pHrodobright fluorescence). ThepHrodobright Apo-MM remained ina mature phagosome for a further1.5 h. Second, FACS was used toexamine whether there was a quanti-tative difference inDC phagocytosisof Apo-MM depending on the drugused to induce MM cell apoptosis(Fig. 2B). Thus, DCs phagocytoseduntreated RPMI8226 cells (15.69§ 2.5)%; in contrast, a significantincrease in MM phagocytosis wasobserved when the RPMI8226 cellswere pre-treated with 1 nM Btzalone, Mapa alone or LDBCMapa(Fig. 2B). Interestingly, pre-treat-ment with Mapa alone orLDBCMapa induced a significantlyhigher level of DC phagocytosiscompared to LDB pre-treatmentalone, indicating that Mapa-medi-ated the dominant role in theincreased DC phagocytosisobserved with LDBCMapa treatedMM cells. Equivalent results wereobserved using treated U266 cells inthe DC phagocytosis assay (Fig.S4). Next, the effect of combinationdrug treatment on lipopolysaccha-ride (LPS)-induced DC maturationwas examined. Thus, whenMoDCswere stimulated with LPS, therewas no significant difference inDC maturation (CD80, CD86,CD83, HLA-ABC and HLADRexpression levels) betweenuntreated DCs or DCs treatedwith LDB, Mapa or LDBCMapa(Fig. 2C). There was also no sig-nificant difference in the level ofIL-12p70 or IL-4 induced byLPS in the presence or absenceof drug co-treatment (Fig. 2D).

Figure 1. Combination low dose bortezomib plus mapatumumab induce human myeloma cell apoptosis.(A) Six human myeloma cell lines were cultured for 48 h in triplicate wells in the previously determined sub-optimal btz dose, or increasing doses of Mapa (0.01–50 mg/mL) alone, or the combination of LDBCM. HMCLapoptosis was measured by annexin V binding and is presented as the mean § SE of three separate experi-ments, * designates a statistical significant difference (where p D <0.05) in HMCL apoptosis between Mapaalone and the combination of Mapa and low dose bortezomib. (B) HMCL were either untreated, or treatedfor 24 h with LDB. TRAIL-R1 expression was determined by FACS staining. Data is shown as histogram over-lays for each HMCL, and is representative of triplicate wells from three separate experiments showing consti-tutive TRAIL-R1 (thick black line), TRAIL-R1 after 24 h LDB treatment (dotted line), isotype control (shadedlight gray) or secondary only control (dashed line). In brackets are MFI values for HMCL TRAIL-R1 expressionafter 24 h LDB treatment (Pre-btz) or untreated (U/T). In C, LP-1 cells were either untreated (black) or pre-treated with 5 nM Btz (white) for 24 h prior to the addition of Mapa (0.01–50 mg/mL) for 48 h. LP-1 apopto-sis was measured by annexin V binding and is presented as the mean § SE of three separate experiments, *designates a statistical significant difference (student t-test where p D <0.05) in LP-1 apoptosis betweenuntreated versus LDB pre-treated cells.

www.tandfonline.com e1038011-3OncoImmunology

Page 4: Induction of potent NK cell-dependent anti-myeloma

Figure 2. For figure legend, see next page.

e1038011-4 Volume 4 Issue 9OncoImmunology

Page 5: Induction of potent NK cell-dependent anti-myeloma

Despite the immunosuppressive nature of apoptoticmyeloma cells, myeloma-pulsed dendritic cells induce ananti-myeloma immune response

Whilst LDB treatment did not inhibit LPS-induced MoDCmaturation, phagocytosis of apoptosis MM cells was associatedwith impairment of DC maturation. Prior co-culture of DCswith LDBCMapa-treated U266 or RPMI8226 cells significantlyinhibited LPS-induced DC maturation (Fig. S5), as assessed byCD80, CD83 and CD86 expression. LPS-induced MoDCCD80 and CD86 expression was more significantly inhibited byApo-RPMI8226 (p < 0.01) than by Apo-U266 (p < 0.05).

Using this information, a modified in vitro expansion cultureof anti-MM lymphocytes was devised using either no myelomacells, untreated U266, or LDBCMapa treated U266 as an anti-gen source. Furthermore, U266-pulsed DCs were then eithertreated with LDBCMapa or LPS. Autologous peripheral bloodmononuclear cells (PBMCs) were added to the MM-pulsedMoDCs for 7 d, and anti-myeloma cytotoxic lymphocyte(AMCL) expansion performed (as per methods) for a further 14

d. The MM killing assay showed untreated or LDBCMapa-treated U266 suppressed the ability of DCs to induce expansionof an anti-MM cytotoxic response (Fig. 3). In contrast, a signifi-cant expansion of MM-specific CTLs occurred when Apo-U266-pulsed DCs were also treated with LDBCMapa during the prim-ing phase of the culture (Fig. 3). The level of CTL expansionwas similar to that observed when Apo-U266-pulsed DCs wereactivated with LPS (Fig. 3). This cytotoxic response was specificto the HMCL, since autologous PBMCs were not lysed by effec-tor cells (Fig. 3). In addition, the AMCL expansion requiredDCs pulsed with myeloma antigen, as the ‘no antigen’ DC con-trol did not induce AMCL effectors (Fig. 3).

LDBCMapa combination therapy does not exert theimmune-modulating effect via a direct effect on dendritic cellsor by differential induction of TRAIL-R1 on immune subsets

To explore the mechanism/s whereby LDBCMapa expandedAMCLs, we first examined the direct effect of LDBCMapa onMoDC function. We know that MoDC effectively phagocytose

Figure 2 (See previous page). Combination LDBCM treatment of human myeloma cells induced increased MoDC cell phagocytosis, but does not affectMoDC response to the TLR ligand LPS. HMCL (U266 and RPMI8226) were labeled with the pH sensitive dye pHrodo, pre-treated with LD btz and Mapafor 48 h, then co-cultured with MoDCs (as per Methods). The kinetics of MoDC phagocytosis of apoptotic HMCLs was monitored by live video microscopy(A), then quantitated by flow cytometry (B). The effect of LDBCM combination or single drug treatment on MoDC responses to LPS were assessed in Cand D. In C, changes in MoDC expression of CD80, CD86, CD83, HLA-ABC and HLADR and CD83 were investigated. Data shown is representative histo-gram overlays, and collated mean fluorescence intensity (MFI) data from triplicate wells of a single experiment. In D, MoDC secretion of IL-12p70 and IL-4 is shown. Data presented is either representative data from three separate experiments (A) and (C). Quantitative data in B and D is the mean § SE forthree separate experiments, p values (student t-test) are provided in B for test conditions that are significantly different to the control, and in D **** rep-resents p < 0.0001.

Figure 3. Combination LDBCM induced increased effector cell killing of human myeloma cells. HLA-A2*01 MoDCs were pulsed with apoptotic U266 cells(induced by LDBCM treatment) in the presence or absence of LDBCM. MM pulsed-DCs were then cultured with autologous PBMCs and the responder Tcells expanded for 21 d in vitro (see Methods for details). The expansion of MM-specific effector cells was then assessed by CTL assay using Cr-labeledU266 cells as targets. Data presented is the percentage lysis (mean§ SE) of targets for three separate experiments on four HLA-A2*01 donors. Data seriesinclude four separate test conditions in the in vitro culture. These include DC pulsed with untreated or treated U266, DC pulsed with treated U266 in thepresence of btzCMapa, DC pulsed with treated U266 and then matured with LPS. Additional controls included ‘unloaded DC’ used at the priming phasein the culture (but U266 as targets in the killing assay), or control targets (autologous PBMC control) to show specificity of the AMCL response. A statisti-cal (student t-test) difference in U266 lysis between the test and control combinations was shown, where * p< 0.05, and ** p < 0.01.

www.tandfonline.com e1038011-5OncoImmunology

Page 6: Induction of potent NK cell-dependent anti-myeloma

Figure 4. For figure legend, see next page.

e1038011-6 Volume 4 Issue 9OncoImmunology

Page 7: Induction of potent NK cell-dependent anti-myeloma

LDBCMapa treated MM cells; how-ever, does this facilitate MoDC matura-tion/licensing in the absence of a toll-like receptor (TLR)-mediated signal?To address this question, we assessedwhether MoDC maturation wasaffected by co-treatment with LDB-Mapa in the presence or absence ofU266 MM cells (as depicted inFig. 4A). We examined MoDC CD80,CD83, CD40 and HLADR expressionlevels at 4 and 48 h after initiation ofLDBCMapa treatment. In the absenceof LPS, we showed no change inMoDC maturation kinetics in the con-text of LDBCMapa alone (Fig. 4B),whether the MoDCs were also pulsedwith U266 or not. In the presence ofLPS, MoDCs up-regulated CD80,CD40, CD83 and HLADR by 48 h,this response (CD80, CD83 andCD40) was significantly decreasedwhen the MoDCs were first pulsed withLDBCMapa-treated U266 (Fig. 4B).In addition, we explored whether theLDBCMapa co-treatment coulddirectly alter MoDC-secreted cytokinelevels. To address this, drug treatedMoDC culture supernatant was col-lected at early and late timepoints (0,4,8 and 24 h) and a cytokine bead arrayperformed for GM-CSF, IL-4, IL-6, IL-12p70, IL-1b, IL-6, IL-10 and sCD40L levels (Fig. 4C). We showed the LPS controlMoDCs secreted increased levels of all cytokines except IL-4. Inthe absence of LPS, apo-U266 pulsed MoDCs secreted higherlevels of IL-4 than non-pulsed MoDCs. This was true whetherthe MoDCs were also treated with LDB, Mapa or LDBCMapa(Fig. 4C). There was no data to indicate that LDBCMapa treat-ment of MoDCs induced increased IL-12p70 or Il-1b secretion.

An alternate potential mechanism for the LDBCMapa-treat-ment effect on CTL expansion was a differential effect on theeffector:suppressor ratio of T cells and their expression ofTRAIL-R1, the target for Mapa. To address the latter, humanPBMCs were directly treated with LDBCMapa and TRAIL-R1assessed at 4 and 48 hours (depicted in Fig. S6A). TRAIL-R1expression on regulatory CD4C T cells (Treg), CD4C or CD8C

T cells was not affected by LDBCMapa treatment during this

48 h time course (Fig. S6B). In addition, lymphocyte subset fre-quencies and their TRAIL-R1 expression were also assessed dur-ing the anti-myeloma expansion culture, using the expansionculture conditions described in Fig. S7. Data showed the per-centage of different lymphocyte subsets changed during theexpansion culture, although this was not specific to theLDBCMapa treatment group. For example, NK cell percent-age increased at one week after 1st restimulation; however,this was the case for all treatment groups. The percentage ofTRAIL-R1C Tregs increased in all culture conditions acrossthe timecourse; in contrast, NK and B cells had a specificTRAIL-R1 increase in the Apo-U266 MoDCCLPS control.Taken together, in the LDBCMapa versus other treatmentgroups, there was no data to indicate a relative increase inTeff or NK cell:Treg ratio, or a specific increased expressionof TRAIL-R1 on these subsets.

Figure 4 (See previous page). Combination LDBCMapa treatment does not directly alter MoDC maturation or cytokine secretion. MoDCs were culturedwith U266 myeloma cells and the effect of subsequent LDBCMapa treatment on MoDC maturation compared to MoDCs activated with LPS. MoDCswere harvested at 4 and 24 h post drug or LPS treatment (as depicted in A) and assessed for the changes in the MoDC maturation markers CD80, CD40,CD83 and HLADR (B). Data show is protein expression assessed by FACS in mean fluorescence intensity (MFI). This is pooled data of therapy responsesfrom three separate normal donor MoDC cultures. A statistically significant difference between test and control groups is represented by * (p < 0.05),**(p < 0.01), ***(p < 0.001). In addition, in C culture supernatant was collected from the co-cultures at 4, 8 and 24 h and cytokine levels (IL-12p70, IL-8,IL-4, GM-CSF, IL-1b, IL-6, IL-10 and sCD40 ligand) assessed by Luminex assay. In these co-culture conditions, ‘tr’ refers to cells treated with LDBCMapa.Data is from triplicate wells of a single experiment, representative of two experiments.

Figure 5. NK cells are required at the priming phase for efficient effector cell killing of human mye-loma cells. A myeloma-specific effector cell culture was established as for Fig. 3. Thus, MM-pulsedMoDCs were co-cultured with autologous PBMCs for 7 d. In addition, monocytes or NK cells weredepleted from the priming phase of the effector cell culture. MM-specific effector cells were expandedwith re-stimulation and expansion steps up to day 21. Harvested effector cells were used in a killingassay with Cr-labeled U266 as targets (E:T D 25:1). Results presented are percentage lysis (mean § SE),pooled data from three separate experiments. Where a statistical difference (student t-test) in U266killing was observed ** pD <0.005, ***pD <0.0005, ****p D <0.0001.

www.tandfonline.com e1038011-7OncoImmunology

Page 8: Induction of potent NK cell-dependent anti-myeloma

Figure 6. Myeloma induced activation of NK cells is increased by Mapatumumab pre-treatment. U266 myeloma cells were either untreated, or treatedwith LDB, Mapa or LDBCMapa for 48 h (as per methods) and the effect of drug treatment on myeloma cell immunogenicity assessed by FACS for expres-sion of CD80, CD86, CD83, HLA-ABC, HLA-DR (A). The effect of drug treatment on myeloma cell NK cell activating ligand expression was also investigated(B). NK activating ligands included the NKG2D ligands MIC-A, MIC-B, ULBP-1, ULBP-2,5,6 and the DNAM-1 ligands CD155 and CD112. Data is presented asrepresentative histogram overlays for the isotype control (filled light gray), untreated (filled dark gray), Mapa (black dotted line), LDB (black line), orLDBCMapa (black dashed line) treated U266 cells. In addition, in B a statistical comparison was made between treatment groups for NK cell activatingligand expression. In C at 48 h, the treated U266 cells were also co-cultured with HLA-A2+ PBLs. Cells were harvested at 4 (A) or 24 (B) h and the effect ofdrug treated myeloma cells on NK cell and T cell subset activation assessed by CD69 and CD25 expression. Data is presented for triplicate wells in a singleexperiment, representative of two separate experiments.

e1038011-8 Volume 4 Issue 9OncoImmunology

Page 9: Induction of potent NK cell-dependent anti-myeloma

Figure 7. For figure legend, see next page.

www.tandfonline.com e1038011-9OncoImmunology

Page 10: Induction of potent NK cell-dependent anti-myeloma

NK cells play a key role in expansion of anti-myelomacytotoxic lymphocytes

Having excluded an immediate direct effect of LDBCMapatreatment on DCs, we next explored whether the LDBCMapa-induced AMCL expansion occurred via an indirect effect at thepriming phase of the culture (Fig. 5). We postulated that FcgRexpressing NK cells or monocytes could be activated via Mapaand change the subsequent expansion of AMCL by an indirectmechanism. Thus, when NK cells (and not monocytes) weredepleted at the start of the culture from the autologous PBMCs,anti-myeloma cytotoxic function was significantly decreased(Fig. 5). Using the 51Cr-release assay to test anti-myeloma killingcapacity of the ensuing expanded cells, we showed that bothT cells (U266 killing in the presence of cold K562 targets) andNK cells participated in the myeloma target killing (Fig. 5).

Mapatumumab plays a key role in NK cell direct activationby LDBCMapa-treated myeloma cells

To explore how NK cells may be affected during the AMCLexpansion culture-priming phase, we examined potential directmolecular interactions between the drug-treated U266 andimmune system cells, with a focus on NK cells and their activa-tion. To do this, we first examined whether drug treatment ofU266 cells affects their immunogenicity or expression of NK-activating ligands. We showed that U266 cell immunogenicity(CD80, CD83, CD86, HLA-ABC and HLA-DR levels) was notaffected by single LDB, Mapa or LDBCMapa treatment(Fig. 6A). Furthermore, we observed that U266 cells constitu-tively expressed the NKG2D ligand MIC-A and low levels of theDNAM-1 ligand CD112; expression of these NK cell-activatingligands was not altered by single LDB, Mapa or combinationLDBCMapa treatment (Fig. 6B). When the drug-treated U266cells were then co-cultured with PBLs, NK cells were activated atboth the 4 and 24 h time points (Fig. 6C). Higher levels of NKactivation were observed in wells where PBLs were co-culturedwith Mapa- or LDBCMapa-treated U266 cells, suggesting thatMapa plays the key role in mediating the LDBCMapa mediatedNK cell activation (Fig. 6C). We also observed a lower but sig-nificant increase in NK cell activation in the context of untreatedU266 cells (vs. PBL alone control), this is likely due to recogni-tion of U266 NK activating ligands by the responding NK cell(ie.NKG2D/MIC-A and DNAM-1/CD112) (Fig. 6C).

Myeloma-pulsed DCs induce NK and CD8C T cellactivation and proliferation

In the final phase of this study, we explored whether theMoDCs (pulsed with drug-treated U266) induced activation ofNK and T cell subsets within 24 h of co-culture. To do this,

U266 cells were pre-treated with LDBCMapa for 48 h, MoDCswere pulsed with the drug-treated U266 and then co-culturedwith autologous PBLs. Co-cultured cells were assessed forchanges in the activation markers CD25 and CD69 at 4 and24 h. At 24 h, MoDCs induced an increased percentage of acti-vated (CD69CCD25– and CD69CCD25C) NK, CD4C andCD8C T cells when the MoDCs were pulsed with untreatedU266 (CLPS) or pulsed with LDBCMapa treated U266(Fig. 7A, B). Analysis of NK subset responses (Fig. 7B) showedan increased percentage of CD25–CD69C cells within both theCD16CCD56C and CD56hiCD16¡ NK cells when the MoDCswere pulsed with U266 cells (in the presence or absence ofLDBCMapa). In the same culture conditions, both CD4C andCD8C T cells showed an increased percentage of CD25CCD69¡

cells.Having revealed that U266 pulsed MoDCs activated NK and

T cell subsets within the first 24 h of the co-culture with autolo-gous PBLs, we next explored whether these activated cells prolif-erated over 7 d of culture. To do this, we labeled the autologousPBLs with Cell Trace Violet (CTV) and examined NK and Tsubset proliferation by CTV dilution assay plus their activationstatus. This study revealed that all NK cell subsets and CD8C Tcells, (but not CD4C T cells) undergo increased proliferationwhen co-cultured with MoDCs pulsed with U266, either afterLDBCMapa pre-treatment or with LPS activation (Fig. 8A). Inthe same co-cultures, all NK cell subsets and CD8C T cells hadan increased percentage of CD25Ccells, this correlated withincreased proliferation in the context of U266-pulsed MoDCs(Fig. 8A). Interestingly, CD4C T cells displayed a markedincrease in the percentage of CD25Ccells when PBLs were cul-tured alone, this decreased when the PBLs were co-cultured withU266 pulsed MoDCs. In addition, day 7 expanded cells fromthe co-culture were also re-stimulated with U266 cells to assesseffector function status at this time point (Fig. 8A). To assess thefunctional status of these activated proliferating NK and T cells,we co-cultured the cells with either LDBCMapa-treated oruntreated U266 cells for a 4 h period and assessed whether theydegranulated (CD107a), and/or secreted cytokines. TheLDBCMapa treated or untreated myeloma cells induced NK cellsubset and CD8C T cell degranulation (CD107aC), a subset ofthese cells also produced IFNg and TNF-a (Fig. 8B). Thisshows the U266-pulsed MoDCs induce effector NK and CD8C

T cells; in these culture conditions, there was no apparent differ-ence in responses by these cells when re-stimulated in the contextof untreated or LDBCMapa-treated U266.

In summary, LDBCMapa treatment of myeloma-pulsed DCsinduces a powerful CTL response, this does not occur via a directeffect on DCs, effector T cells or Tregs. Rather Mapa has two

Figure 7 (See previous page). Myeloma-pulsed DCs induce increased activation of autologous NK, CD4C and CD8C T cells. Monocyte-derived DCs werepulsed with untreated or drug (Mapa, LDB or LDBCMapa) treated U266 cells (as per methods) for 2 h, then co-cultured with autologous normal donor(n D 2) PBLs for a further 24 h. Cells were collected at 4 and 24 h, and activation of NK cells and T cell subsets assessed by CD69 and CD25 expression.Data presented in A are representative dot plots of NK and T cell activation in cultures from a single normal donor. Shown are PBL alone, PBLCDC,PBLCU266 pulsed MoDC (where U266 were pre-treated with LDBCMapa), and PBLCU266 pulsed MoDC in the context of 1 mg/mL LPS. Collated data inB is from duplicate co-culture wells (single normal donor representative of two normal donor experiments) and shows the percentage of CD25¡CD69C,CD25CCD69C and CD25CCD69¡ cells in NK cell subsets, CD4C and CD8C T cells.

e1038011-10 Volume 4 Issue 9OncoImmunology

Page 11: Induction of potent NK cell-dependent anti-myeloma

Figure 8. For figure legend, see next page.

www.tandfonline.com e1038011-11OncoImmunology

Page 12: Induction of potent NK cell-dependent anti-myeloma

distinct modes of action which co-opt the immune system indi-rectly. First, Mapa induces NK cell activation within 24 h ofcontact with LDBCMapa treated myeloma cells. Second, Mapapromotes DC phagocytosis of LDBCMapa treated myelomacells. We also know there is an ensuing antigen-specific responseby both NK and CD8C T cells within 24 h and proliferation by7 d to produce fully functional effector cells.

Discussion

Until recently, it has proven difficult to develop effectiveimmunotherapies against MM. This is in part due to the absenceof effective myeloma targeting antibodies. Recently, two mono-clonal antibodies, Elotuzumab (anti-SLAMF7) and Daratumu-mab (anti-CD38), have shown pre-clinical and early clinical trialsuccess. Whilst both clearly rely on NK cell function to inducemaximal plasma cell apoptosis, little is known about their down-stream ability to induce immunogenic cell death and prime cyto-toxic cellular immunity. We show that the agonistic TRAIL-R1antibody Mapa may also act as a myeloma-targeting antibody.We revealed that Mapa has at least two distinct modes of anti-myeloma activity. First, Mapa induces myeloma cell apoptosisdirectly, which is increased via pre- or concurrent treatment withLDB. These results compare favorably to studies where Mapawas used in vitro to induce HMCL and primary myeloma celldeath.25,33

Secondly, we show that Mapa induced plasma cell killing eli-cited a powerful anti-myeloma cytotoxic response. Critically, thisanti-myeloma CTL expansion occurred optimally when bothMapa and NK cells were present during the priming phase.Indeed, compared to LDB treatment, Mapa-treated myelomacells significantly increased NK cell activation and led to the max-imal uptake of apoptotic cells by DCs, which in turn resulted inreproducible expansion and activation of cytotoxic CD4C andCD8C T cells.

Prior studies have shown that apoptotic myeloma cells inhibitLPS-induced DC maturation due to a combination of IL-10,TGF-b and VEGF inducing activated phospho-STAT-3.34,35

Our study confirms this observation. However, despite this,when autologous PBMCs were primed in a co-culture systemcontaining LDBCMapa, myeloma targets, PBMC and DCs, anexpanded population of myeloma-specific cytotoxic cells wasconsistently generated. The importance of NK cells during theco-culture priming phase suggests cross-talk between NK cellsand the Apo-MM pulsed DCs. It is known that NK-DC crosstalk involves a number of mechanisms including soluble factors

IL-12, IL-18, IL-15 and type I interferon and contact-dependentsignals (NKG2DL-NKG2D, GITRL-GITR, LFA1-ICAM1),36

which can have a dramatic effect on the functional capacity ofthe DC and ensuing T and NK cell responses. We did not seeany effect of LDBCMapa treatment on cytokine secretion byMoDCs in the context of treated myeloma cells. However, thisdoes not exclude the possibility that the cross-talk occurs betweenNK cells and DCs at a later time point in our culture system. It islikely the NK cells are initially activated via interactions betweenactivating receptors and activating ligands expressed on myelomacells. In addition, Mapa-bound to either apoptotic or viable mye-loma cells will be bound by NK cell FcgRIII. The net effect ofthese interactions is increased NK cell activation, degranulationand production of cytokines,37 including IFNg, which has a DClicensing effect.38 This DC licensing is likely to have occurredfollowing cross-talk with activated NK cells present within theculture.39,40 Whilst FcgR-mediated phagocytosis of antigen isknown to be an efficient pathway by which to load DCs withantigen41,42 induction of an antigen-specific T cell responserequires a further licensing signal for the DCs. In our study, thisis efficiently achieved by Mapa-activated NK cells providing aDC licensing signal.

Taken together, our study showed Mapa is a novel anti-mye-loma antibody that has multiple effector arms. The rational com-bination with LDB enhanced optimal myeloma cell apoptosisleading to effective immunogenic cell death mediated by pre-served DC function and NK cell mediated licensing. Our studyprovides a strong rationale for the application of LDBCMapa inclinical trials of MM immunotherapy.

Methods and Materials

Cell lines and materialsHuman myeloma cell lines RPMI8226, U266 and NCI-

H929 cell lines were obtained from American Type Cell Collec-tion (ATCC); LP-1, OPM-2, and JJN-03 cell lines were fromDSMZ (Braunschweig, Germany). Cell lines were cultured inRPMI1640 supplemented with 10% heat-inactivated fetal bovineserum and 2 mM L-glutamine. btz was from Millenium Pharma-ceuticals, MA; Mapa from Human Genome Sciences Inc., NJ;Annexin V-FITC, 7-AAD, anti-mouse Ig-PE, the monoclonalantibodies CD11c-PECy7, CD83-APC and HLADR-FITCwere from BD Biosciences, CA and the IL-12p70 EIA kit fromBD PharMingen, CA. pHrodoTM (Invitrogen, Carlsbad, CA);lipopolysaccharide (from E.coli, SIGMA ALDRICH, St Louis,Mo).

Figure 8 (See previous page). Myeloma-pulsed DCs induce increased proliferation of NK cells, CD4C and CD8C T cells. Monocyte-derived DCs werepulsed with untreated or drug (Mapa, LDB or LDBCMapa) treated U266 cells (as per methods) for 2 h, then co-cultured with autologous normal donor(n D 2) PBLs (pre-labeled with Cell trace Violet, CTV) for 7 d. Cells were harvested, labeled with conjugated antibodies and analyzed on the FACS. In A,the proliferation and activation status of NK and T cell subsets was shown by gating first on lineage markers (NK subsets: CD56hiCD16¡, CD16-CD56lo,CD56loCD16C, and T cells CD3CCD4Cand CD3CCD8C) followed by CTV dilution (left panels) or CD69,CD25 expression (right panels). Cell proliferationdata is shown as individual histogram overlays with the PBL alone control (filled light gray) with the test groups (thick black line) either PBLCU266-pulsedMoDC or PBLCU266 pulsed MoDC (where U266 were pre-treated with LDBCMapa). In B day 7 cells were recovered from the co-culture and stimulatedby co-culture for 4 h, cells were stained for activation markers (CD25,CD69), degranulation (CD107a), and cytokines (IFNg and TNF-a). Data is presentedas dot plots showing CD107a expression and TNF-a (left panel) or IFNg (right panel) production by NK or T cell subsets.

e1038011-12 Volume 4 Issue 9OncoImmunology

Page 13: Induction of potent NK cell-dependent anti-myeloma

Myeloma therapy experimentsMyeloma cells (2 £ 106/mL) were treated with mono- or

combination therapy for 48 h in 24 well plates. Initially, drugswere used as monotherapy including btz (0–10 nM) or Mapa(0–10 mg/mL) and apoptosis induction assessed. To do this,after 48 h treatment, cells were harvested, washed once inDulbecco’s PBS and then stained with Annexin V-FITC and7AAD for 10 min at room temperature. Stained cells were thenanalyzed on the BD LSRII flow cytometer. For combinationtherapy, myeloma cell lines were treated with the highest dose ofbtz that did not induce apoptosis (termed ‘low dose’, this dosewas established from the monotherapy titration) combined withtitrated amounts of Mapa (from 0–10 mg/mL) for 48 h prior toevaluation of myeloma cell apoptosis.

Combination Mapa and low dose bortezomib therapy do notinhibit dendritic cell function

MoDCs were established from fresh PB monocytes asdescribed previously.43,44 Briefly, purified monocytes were cul-tured for 5 d in 10 ng/mL IL-4 and 20 ng/mL GM-CSF (Pepro-tech, NJ), the media was renewed at day 3. On day 5, non-adherent cells were removed by gentle aspiration from the cultureflask and the purity of immature MoDCs checked by FACSusing the phenotype CD11chi/CD14¡/CD11bint.

DC phagocytosis of treated myeloma cellsMoDCs were harvested from the above cultures and used for

myeloma cell phagocytosis assays. To induce myeloma cell apo-ptosis, RPMI8226 were either g-irradiated (30 Gy), or treatedwith 1 nM btz alone, 0.9 mg Mapa alone or a combination ofboth drugs for 48 h. Treated myeloma cells were stained with0.5 mg/mL pHrodo, washed with PBS twice before co-culturewith MoDCs (1:2 myeloma:DC ratio) for 4 h at 37�C. A nega-tive control co-culture was also performed at 4�C. Harvested cellswere then stained with anti-CD11c-PECy7 and Fluorogold(Invitrogen); stained cells were analyzed on the BD LSRII flowcytometer, 5000 CD11chi events were collected and the fre-quency of CD11chi/pHrodohi collected. In other experiments,the MoDC myeloma cell phagocytosis assay was performed inthe presence of 1 nM btz. pHrodoTM is a pH sensitive dye andundergoes a quantum yield in red fluorescence when stained apo-ptotic bodies are processed to the low pH environment of anendosome. Thus, cells that are CD11chi/pHrodohi are MoDCswith endocytosed myeloma cells and CD11chi/pHrodolo arelikely MoDCs with adherent myeloma apoptotic bodies. To ver-ify that the FACS data did indeed represent MoDC phagocytosisof treated myeloma cells we examined an aliquot of the same cellsby fluorescent live video microscopy. To do this, apoMM-pulsedMoDCs (1 £ 105) were added to optimally manufactured grids[125 mm (l) £ 125 mm (w) £ 60 mm (h)], which are individualpaddocks designed to contain the cells within the microscopefield of view (as described previously45 The grids were thenplaced in imaging chamber slides (Ibidi, DKSH, Martinsried,Germany). Images were taken on the Leica SP5 confocal micro-scope (Leica, Wetzlar, Germany). Voltage gain, offset and focuswere set on compensation wells before the first image was taken.

DCs were then added to wells and positions were selected tomaximise the wells in the field of view with DCs clearly visible.The tumor target cells were added (2 £ 105) just prior tothe image sequence starting. Images were taken at each positionevery 4–5mins for 3–4 h. The objective of the microscope was20£. The images were collated and analyzed using MetamorphSoftware (MDS Analytical Technologies, Downington USA),which allows single frame images to be viewed as a multi-framevideo.

LPS-induced DC responsesMoDCs were cultured with 1 mg/mL LPS for 48 h at 37�C in

the presence or absence of 1 nM btz. MoDCs were harvested andstained with antibodies to CD11c, CD83 and HLA-DR, cellswere analyzed on the BDLSRII flow cytometer to examine upre-gulation of CD83 and HLA-DR as an indicator of LPS-inducedMoDC maturation. In addition, culture supernatant was col-lected at 48 h of LPS treatment and an IL-12p70 assay per-formed (as per manufacturer’s instructions).

DC proteasome activity assay (chymotrypsin-like functionalsubunit)

MoDCs were cultured (as described above), proteasomes wereextracted from the DCs and treated with 1 nM btz alone, 1 mg/mL Mapa alone or a combination of both drugs for 4 h at 37�C.Treated proteasomes were pelleted and then a Ch-like protea-some assay performed (Millipore), as per manufacturer’s instruc-tions, and46,47 using Suc-Leu-Leu-Val-Tyr-MCA as the 20Sproteasome substrate.

Expansion of myeloma-specific cytotoxic lymphocytesin vitro and detection of cytotoxic function

U266 myeloma cells were either untreated, or treated withLDBCMapa for 48 h to induce apoptosis (Apo-U266). U266 orApo-U266 cells were washed twice in RPMI1640 containing10% fetal bovine serum, complete medium (CM). MoDCs(HLA-A2*01 normal donor) were pulsed with either untreatedor Apo-U266 cells for 4 h in the presence or absence ofLDBCMapa. Subsequently, the U266-pulsed MoDCs were co-cultured with autologous PBMCs for 7 d in CM containing20 U/mL IL-2, 10 ng/mL of IL-7 and IL-15 (priming phase).On days 7 and 14, cells were harvested and re-stimulated withautologous myeloma-pulsed MoDCs, expansion of CTL duringthese time points occurred in CM supplemented with 25 U IL-2. On day 21, cells were harvested and their anti-myeloma activ-ity assessed by a chromium-release assay. Controls for the CTLexpansion culture include unpulsed MoDCs, or U266-pulsedMoDCs that were activated with 1 mg/mL LPS. Chromium-release assay was performed as described previously.9 Briefly,chromium-labeled U266 cells were cultured (in triplicate wells)with effector cells from the CTL expansion culture at an effector-target ratio (E:T) from 6.25 to 50:1. After 4 h, culture superna-tant was collected from each well and the amount of Cr releasedby U266 cells measured on a g-counter (Wallac Wizard 1470,Perkin Elmer, MA, USA). The amount of U266 cell death (per-centage lysis) was compared to total U266 Cr-release wells

www.tandfonline.com e1038011-13OncoImmunology

Page 14: Induction of potent NK cell-dependent anti-myeloma

(U266 plus 2M HCl). Thus, percentage lysis was calculated usingthe formula (mean cpm – spontaneous cpm)/total cpm. To assessthe contribution of different effector cells (NK versus T cell) tothe killing of U266 cells, a ‘cold target assay’ was performed. Todo this, the Cr-release assay was performed as above, except thatin addition to the Cr-labeled U266 cells, equal numbers of unla-beled K562 cells (the cold target) were included. To assess the rel-ative importance of NK cells or monocytes at the priming phaseof the CTL expansion culture, NK cells (CD3¡/CD56C) ormonocytes (CD14C) were removed from the autologous PBMCsby FACS-sorting, purity was >95%. The NK- or monocyte-depleted PBMCs were then co-cultured with MM-pulsedMoDCs as above.

Direct effect of LDB§Mapa treatment on MoDCmaturation and cytokine secretion

Day five MoDCs were divided into two groups, one wasunpulsed and the other pulsed with LDBCMapa treated U266myeloma cells for 4 h. Both MoDC groups were then each fur-ther dissected into three arms (either untreated, treated withLDBCMapa or 1 mg/mL LPS) and cultured for 48 h as depictedin Fig. 5A. LDBCMapa treatment comprised 1 nM btz and1 mg/mL Mapa. Cells were harvested at 4 and 48 h to assessMoDC maturation at an early vs. late timepoint. Maturation wasassessed by FACS following labeling with antibodies to CD80,CD40, CD83 and HLADR. In addition, culture supernatantwas harvested at 0, 4, 8 and 24 h and a multiplex Luminex assayperformed to assess changes in cytokine levels (GM-CSF, IL-4,IL-8, IL-12p70, IL-1b, IL-6, IL-10 and sCD40L) in response tothe drug treatment.

Effect of drug treatment on human myeloma cellimmunogenicity and NK-activating ligand expression levels

U266 and RPMI8226 myeloma cells were either untreated, ortreated with LDB, Mapa or LDBCMapa for 48 h (as per meth-ods above) and the effect of drug treatment on myeloma cellimmunogenicity assessed. Treated myeloma cells were labeledwith antibodies to CD80-FITC, CD86-PE, CD83-APC, HLA-DR-APC-H7 (all from BD Biosciences) and HLA-ABC-V450(e-Bioscience) and changes in protein expression detected byFACS analysis. In addition, treated myeloma cells were labeledwith the antibodies CD155-FITC, MIC-A-PE, MIC-B-APC,CD112-FITC, ULBP-2,5,6-APC, ULBP-1-PE (all RnD Sys-tems) and changes in NK-activating ligand expression assessed byFACS analysis.

Assessment of direct activation of NK and T cells by drugtreated myeloma cells

Drug-treated U266 cells (prepared as above) were harvested at48 h, washed twice in media and then co-cultured with HLA-A2C PBLs. The PBLs from two normal donors were co-culturedwith treated U266 cells in separate cultures in triplicate wells.Cells were harvested 4 or 24 h later and labeled with antibodiesto CD45-BV510, CD3-BV711, CD4-BV605 (Biolegend)CD14- and CD19-APCH7, CD56-PE, CD8-PECy5.5, CD25-APC, CD69-PECy7 (BD Biosciences). Labeled cells were then

analyzed by FACS, and co-culture induced activation of NK orT cell subsets assessed following a devised gating strategy depictedin Fig. S8.

Detection of NK or T cell subset activation following co-culture with myeloma-pulsed MoDCs

Day 5 MoDCs were pulsed with drug-treated U266 cells at a1:1 ratio as in the protocol for CTL expansion culture (seeabove). After 4 h, cells were washed twice and left overnight.Recovered autologous PBLs were washed, counted and added tothe pulsed MoDCs at a 5:1 ratio. Co-cultured cells were recov-ered 4 and 24 h later. Four hours prior to each timepoint, Golgistop and CD107a-PeCy5 (BD) antibody were added to theappropriate wells for intracellular staining. Cells were washedand stained for NK, CD4 and CD8 T cell markers (CD3-BV711, CD4-BV605 (both Biolegend), CD56-PECy7, CD8-PerCPCy5.5, CD14/CD19-APCH7, CD16-PE (all BD Bio-sciences). Cells were then fixed and permeabilized (BD Cytofix/Cytoperm) before staining for intracellular cytokines (TNFa-APC and IFNg-FITC (both BD Biosciences). In addition, foractivation status, appropriate wells were stained with a separateantibody panel for NK, CD4 and CD8 T cell markers combinedwith activation markers (CD3-BV711, CD4-BV605 (Biole-gend), CD8-PerCPCy5.5, CD14/CD19-APCH7, CD56-PE,CD16-FITC, CD69-PECy7, CD25-APC (BD Biosciences).Labeled cells were acquired on the BD Fortessa FACS analyzer,FACS data was then analyzed using FlowJo analysis software anda gating strategy (depicted in Fig. S9) to derive the percentage ofNK and T cell subsets expressing activation markers (CD25 andCD69), the degranulation marker (CD107a) and cytokinesIFNg and TNF-a.

Detection of NK or T cell subset proliferation followingco-culture with myeloma-pulsed MoDCs

Day 5 MoDCs were pulsed with drug-treated U266 cells at a1:1 ratio as in the protocol for CTL expansion culture (asdescribed above). After 4 h, cells were washed twice with culturemedia and left overnight. Autologous PBLs (from two separatenormal donors) were washed, counted and stained with 2.5uMCTV (Life Technologies) for 20 min, washed and added to themyeloma pulsed MoDCs and cultured for 7 d at a 5:1 ratio in0.5 mL RPMI1640 plus 10% FCS, IL-2 (20 U/mL) in 48-wellplates. On day 4, culture media was replaced with fresh RPMIplus 10% FCS and low dose IL-2 (20 U/mL). Co-cultured cellswere harvested on day 7 and stained for NK, CD4, CD8 T celland activation markers (CDD3-BV785, CD4-BV605 (bothfrom Biolegend), CD8-PerCP Cy5.5, CD14/CD19-APCH7,CD16-FITC, CD56-PE, CD69-PECy7, and CD25-APC (allfrom BD Biosciences). Cells were analyzed by FACS and the levelof activation and proliferation assessed on NK and T cell subsetsusing the same gating strategy depicted in Fig. S9.

Detection of expansion culture NK and T cell effector statusDay 7 co-cultured cells (from above) were re-stimulated with

antigen (untreated or treated U266 cells) for 4 h prior to stain-ing. Golgi stop and CD107a-PeCy5 (BD) antibody were added

e1038011-14 Volume 4 Issue 9OncoImmunology

Page 15: Induction of potent NK cell-dependent anti-myeloma

to the appropriate wells. Staining panels were the same as for theNK, CD4 and CD8 activation protocol (above).

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Funding

This work was completed with funding from HumanGenome Sciences Inc. and National Health and MedicalResearch Council of Australia (program grants 454569 and1013667).

Supplemental Material

Supplemental data for this article can be accessed on thepublisher’s website.

Ethics Statement

Normal donor cells were acquired from Red Cross BloodBank donors following approval from the Peter MacCallum Can-cer Center Human Ethics Committee.

References

1. Attal M, Harousseau JL. Autologous peripheral bloodprogenitor cell transplantation for multiple myeloma.Baillieres Best Pract Res Clin Haematol 1999; 12:171-91; PMID:11000991; http://dx.doi.org/10.1053/beha.1999.0015

2. Child JA, Morgan GJ, Davies FE, Owen RG, Bell SE,Hawkins K, Brown J, Drayson MT, Selby PJ. High-dose chemotherapy with hematopoietic stem-cell rescuefor multiple myeloma. N Engl J Med 2003; 348:1875-83; PMID:12736280; http://dx.doi.org/10.1056/NEJMoa022340

3. Palumbo A, Anderson K. Multiple myeloma. N Engl JMed 2011; 364:1046-60; PMID:21410373; http://dx.doi.org/10.1056/NEJMra1011442

4. Ocio EM, Richardson PG, Rajkumar SV, Palumbo A,Mateos MV, Orlowski R, Kumar S, Usmani S, RoodmanD, Niesvizky R et al. New drugs and novel mechanismsof action in multiple myeloma in 2013: a report from theinternational myeloma working group (IMWG). Leuke-mia 2014; 28:525-42; PMID:24253022; http://dx.doi.org/10.1038/leu.2013.350

5. Donato F, Gay F, Bringhen S, Troia R, Palumbo A.Monoclonal antibodies currently in Phase II and III tri-als for multiple myeloma. Expert Opin Biol Ther2014; 14:1127-44; PMID:24749510; http://dx.doi.org/10.1517/14712598.2014.908848

6. Brimnes MK, Svane IM, Johnsen HE. Impaired func-tionality and phenotypic profile of dendritic cells frompatients with multiple myeloma. Clin Exp Immunol2006; 144:76-84; PMID:16542368; http://dx.doi.org/10.1111/j.1365-2249.2006.03037.x

7. Schutt P, Brandhorst D, Stellberg W, Poser M, EbelingP, Muller S, Buttkereit U, Opalka B, Lindemann M,Grosse-Wilde H et al. Immune parameters in multiplemyeloma patients: influence of treatment and correla-tion with opportunistic infections. Leuk Lymphoma2006; 47:1570-82; PMID:16966269; http://dx.doi.org/10.1080/10428190500472503

8. Chan AC, Neeson P, Leeansyah E, Tainton K, QuachH, Prince HM, Harrison SJ, Godfrey DI, Ritchie D,Berzins SP. Natural killer T cell defects in multiplemyeloma and the impact of lenalidomide therapy. ClinExp Immunol 2014; 175:49-58; PMID:24032527;http://dx.doi.org/10.1111/cei.12196

9. Hsu AK, Quach H, Tai T, Prince HM, Harrison SJ,Trapani JA, Smyth MJ, Neeson P, Ritchie DS. Theimmunostimulatory effect of lenalidomide on NK-cellfunction is profoundly inhibited by concurrent dexa-methasone therapy. Blood 2011; 117:1605-13;PMID:20978269; http://dx.doi.org/10.1182/blood-2010-04-278432

10. Rapoport AP, Stadtmauer EA, AquiN, Badros A, Cotte J,Chrisley L, Veloso E, Zheng Z,Westphal S, Mair R et al.Restoration of immunity in lymphopenic individualswith cancer by vaccination and adoptive T-cell transfer.Nat Med 2005; 11:1230-7; PMID:16227990; http://dx.doi.org/10.1038/nm1310

11. Tesniere A, Apetoh L, Ghiringhelli F, Joza N, PanaretakisT, KeppO, Schlemmer F, Zitvogel L, KroemerG. Immu-nogenic cancer cell death: a key-lock paradigm. CurrOpin Immunol 2008; 20:504-11; PMID:18573340;http://dx.doi.org/10.1016/j.coi.2008.05.007

12. Hsi ED, Steinle R, Balasa B, Szmania S, DraksharapuA, Shum BP, Huseni M, Powers D, Nanisetti A, ZhangY et al. CS1, a potential new therapeutic antibody tar-get for the treatment of multiple myeloma. Clin CancerRes 2008; 14:2775-84; PMID:18451245; http://dx.doi.org/10.1158/1078-0432.CCR-07-4246

13. Tai Y-T, Dillon M, Song W, Leiba M, Li X-F, BurgerP, Lee AI, Podar K, Hideshima T, Rice AG et al. Anti-CS1 humanized monoclonal antibody HuLuc63 inhib-its myeloma cell adhesion and induces antibody-depen-dent cellular cytotoxicity in the bone marrow milieu.Blood 2008; 112:1329-37; PMID:17906076; http://dx.doi.org/10.1182/blood-2007-08-107292

14. de Weers M, Tai YT, van der Veer MS, Bakker JM,Vink T, Jacobs DC, Oomen LA, Peipp M, Valerius T,Slootstra JW et al. Daratumumab, a novel therapeutichuman CD38 monoclonal antibody, induces killing ofmultiple myeloma and other hematological tumors. JImmunol 2011; 186:1840-8; PMID:21187443; http://dx.doi.org/10.4049/jimmunol.1003032

15. Nijhof IS, Groen RW, Noort WA, van Kessel B, deJong-Korlaar RA, Bakker JM, Lammerts-van BuerenJJ, Parren PW, Lokhorst HM, van de Donk NW et al.Preclinical evidence for the therapeutic potential ofCD38-targeted immuno-chemotherapy in multiplemyeloma patients refractory to lenalidomide and borte-zomib. Clin Cancer Res 2014; 21(12):1–9;PMID:25398450

16. van der Veer MS, de Weers M, van Kessel B, BakkerJM, Wittebol S, Parren PW, Lokhorst HM, Mutis T.Towards effective immunotherapy of myeloma:enhanced elimination of myeloma cells by combinationof lenalidomide with the human CD38 monoclonalantibody daratumumab. Haematologica 2011; 96:284-90; PMID:21109694; http://dx.doi.org/10.3324/haematol.2010.030759

17. Jakubowiak AJ, Benson DM, Bensinger W, Siegel DS,Zimmerman TM, Mohrbacher A, Richardson PG,Afar DE, Singhal AK, Anderson KC. Phase I trial ofanti-CS1 monoclonal antibody elotuzumab in combi-nation with bortezomib in the treatment of relapsed/refractory multiple myeloma. J Clin Oncol 2012;30:1960-5; PMID:22291084; http://dx.doi.org/10.1200/JCO.2011.37.7069

18. Lonial S, Vij R, Harousseau JL, Facon T, Moreau P,Mazumder A, Kaufman JL, Leleu X, Tsao LC, West-land C et al. Elotuzumab in combination with lenali-domide and low-dose dexamethasone in relapsed orrefractory multiple myeloma. J Clin Oncol 2012;30:1953-9; PMID:22547589; http://dx.doi.org/10.1200/JCO.2011.37.2649

19. Lokhorst HM, Laubach J, Nahi H, Plesner T, GimsingP, Hansson M, Minnema M, Lassen UN, Krejcik J,Ahmadi T et al. Dose-dependent efficacy of

daratumumab (DARA) as monotherapy in patientswith relapsed or refractory multiple myeloma (RRMM). ASCO Meet Abs 2014; 32:8513

20. Plesner T, Arkenau H-T, Lokhorst HM, Gimsing P,Krejcik J, Lemech CR, Minnema M, Lassen UN,Ahmadi T, Yeh H et al. Safety and efficacy of daratu-mumab with lenalidomide and dexamethasone inrelapsed or relapsed, refractory multiple myeloma.ASCO Meet Abstr 2014; 32:8533

21. Ashkenazi A, Pai RC, Fong S, Leung S, Lawrence DA,Marsters SA, Blackie C, Chang L, McMurtrey AE,Hebert A et al. Safety and antitumor activity of recom-binant soluble Apo2 ligand. J Clin Invest 1999;104:155-62; PMID:10411544; http://dx.doi.org/10.1172/JCI6926

22. Evdokiou A, Bouralexis S, Atkins GJ, Chai F, Hay S,Clayer M, Findlay DM. Chemotherapeutic agents sen-sitize osteogenic sarcoma cells, but not normal humanbone cells, to Apo2L/TRAIL-induced apoptosis. Int JCancer 2002; 99:491-504; PMID:11992538; http://dx.doi.org/10.1002/ijc.10376

23. Kothny-Wilkes G, Kulms D, Poppelmann B, LugerTA, Kubin M, Schwarz T. Interleukin-1 protects trans-formed keratinocytes from tumor necrosis factor-related apoptosis-inducing ligand. J Biol Chem 1998;273:29247-53; PMID:9786937; http://dx.doi.org/10.1074/jbc.273.44.29247

24. Lawrence D, Shahrokh Z, Marsters S, Achilles K, ShihD, Mounho B, Hillan K, Totpal K, DeForge L, SchowP et al. Differential hepatocyte toxicity of recombinantApo2L/TRAIL versions. Nat Med 2001; 7:383-5;PMID:11283636; http://dx.doi.org/10.1038/86397

25. Menoret E, Gomez-Bougie P, Geffroy-Luseau A, Dan-iels S, Moreau P, Le Gouill S, Harousseau JL, BatailleR, Amiot M, Pellat-Deceunynck C. Mcl-1L cleavage isinvolved in TRAIL-R1- and TRAIL-R2-mediated apo-ptosis induced by HGS-ETR1 and HGS-ETR2 humanmAbs in myeloma cells. Blood 2006; 108:1346-52;PMID:16638930; http://dx.doi.org/10.1182/blood-2005-12-007971

26. Mitsiades CS, Treon SP, Mitsiades N, Shima Y,Richardson P, Schlossman R, Hideshima T, AndersonKC. TRAIL/Apo2L ligand selectively induces apoptosisand overcomes drug resistance in multiple myeloma:therapeutic applications. Blood 2001; 98:795-804;PMID:11468181; http://dx.doi.org/10.1182/blood.V98.3.795

27. Belch A, Sharma A, Spencer A, Tarantolo S, Bahlis NJ,Doval D, Gallant G, Kumm E, Klein J, Chanan-KhanAA. A multicenter randomized phase II trial of mapatu-mumab, a TRAIL-R1 agonist monoclonal antibody, incombination with bortezomib in patients withrelapsed/refractory multiple myeloma (MM). ASHAnn Meet Abstr 2010; 116:5031

28. Straube C, Wehner R, Wendisch M, Bornhauser M,Bachmann M, Rieber EP, Schmitz M. Bortezomib sig-nificantly impairs the immunostimulatory capacity ofhuman myeloid blood dendritic cells. Leukemia 2007;

www.tandfonline.com e1038011-15OncoImmunology

Page 16: Induction of potent NK cell-dependent anti-myeloma

21:1464-71; PMID:17495970; http://dx.doi.org/10.1038/sj.leu.2404734

29. Subklewe M, Sebelin-Wulf K, Beier C, Lietz A, MathasS, Dorken B, Pezzutto A. Dendritic cell maturationstage determines susceptibility to the proteasome inhib-itor bortezomib. Hum Immunol 2007; 68:147-55;PMID:17349869; http://dx.doi.org/10.1016/j.humimm.2006.12.005

30. Blanco B, P�erez-Sim�on JA, S�anchez-Abarca LI, Carva-jal-Vergara X, Mateos J, Vidriales B, L�opez-HolgadoN, Maiso P, Alberca M, Villar�on E et al. Bortezomibinduces selective depletion of alloreactive T lympho-cytes and decreases the production of Th1 cytokines.Blood 2006; 107:3575-83; PMID:16282346; http://dx.doi.org/10.1182/blood-2005-05-2118

31. Chanan-Khan A, Sonneveld P, Schuster MW, Stadt-mauer EA, Facon T, Harousseau J-L, Ben-Yehuda D,Lonial S, Goldschmidt H, Reece D et al. Analysis ofherpes zoster events among bortezomib-treated patientsin the phase III APEX study. J Clin Oncol 2008;26:4784-90; PMID:18711175; http://dx.doi.org/10.1200/JCO.2007.14.9641

32. LeBlanc R, Catley LP, Hideshima T, Lentzsch S, Mit-siades CS, Mitsiades N, Neuberg D, Goloubeva O,Pien CS, Adams J et al. Proteasome inhibitor PS-341inhibits human myeloma cell growth in vivo and pro-longs survival in a murine model. Cancer Res 2002;62:4996-5000; PMID:12208752

33. Surget S, Chiron D, Gomez-Bougie P, Descamps G,M�enoret E, Bataille R, Moreau P, Le Gouill S, AmiotM, Pellat-Deceunynck C. Cell death via DR5, but notDR4, is regulated by p53 in myeloma cells. Cancer Res2012; 72:4562-73; PMID:22738917; http://dx.doi.org/10.1158/0008-5472.CAN-12-0487

34. Ratta M, Fagnoni F, Curti A, Vescovini R, Sansoni P,Oliviero B, Fogli M, Ferri E, Della Cuna GR, Tura Set al. Dendritic cells are functionally defective in

multiple myeloma: the role of interleukin-6. Blood2002; 100:230-7; PMID:12070032; http://dx.doi.org/10.1182/blood.V100.1.230

35. Yang D-H, Park J-S, Jin C-J, Kang H-K, Nam J-H,Rhee J-H, Kim Y-K, Chung S-Y, Choi S-J-N, Kim H-Jet al. The dysfunction and abnormal signaling pathwayof dendritic cells loaded by tumor antigen can be over-come by neutralizing VEGF in multiple myeloma. Leu-kemia Res 2009; 33:665-70; PMID:18922577; http://dx.doi.org/10.1016/j.leukres.2008.09.006

36. Barreira da Silva R, M€unz C. Natural killer cell activa-tion by dendritic cells: balancing inhibitory and activat-ing signals. Cell Mol Life Sci 2011; 68:3505-18;PMID:21861182; http://dx.doi.org/10.1007/s00018-011-0801-8

37. Fauriat C, Long EO, Ljunggren HG, Bryceson YT.Regulation of human NK-cell cytokine and chemokineproduction by target cell recognition. Blood 2010;115:2167-76; PMID:19965656; http://dx.doi.org/10.1182/blood-2009-08-238469

38. Piccioli D, Sbrana S, Melandri E, Valiante NM. Con-tact-dependent stimulation and inhibition of dendriticcells by natural killer cells. J Exp Med 2002; 195:335-41; PMID:11828008; http://dx.doi.org/10.1084/jem.20010934

39. Walzer T, Dalod M, Robbins SH, Zitvogel L, Vivier E.Natural-killer cells and dendritic cells: “l’union fait laforce”. Blood 2005; 106:2252-8; PMID:15933055;http://dx.doi.org/10.1182/blood-2005-03-1154

40. Walzer T, Dalod M, Vivier E, Zitvogel L. Natural killercell-dendritic cell crosstalk in the initiation of immuneresponses. Exp Opin Biol Ther 2005; 5(Suppl 1):S49-59; PMID:16187940; http://dx.doi.org/10.1517/14712598.5.1.S49

41. Kalergis AM, Ravetch JV. Inducing tumor immunitythrough the selective engagement of activatingFcgamma receptors on dendritic cells. J Exp Med

2002; 195:1653-9; PMID:12070293; http://dx.doi.org/10.1084/jem.20020338

42. Dhodapkar KM, Kaufman JL, Ehlers M, Banerjee DK,Bonvini E, Koenig S, Steinman RM, Ravetch JV, Dho-dapkar MV. Selective blockade of inhibitory Fcgammareceptor enables human dendritic cell maturation withIL-12p70 production and immunity to antibody-coated tumor cells. Proc Natl Acad Sci U S A 2005;102:2910-5; PMID:15703291; http://dx.doi.org/10.1073/pnas.0500014102

43. Dauer M, Obermaier B, Herten J, Haerle C, Pohl K,Rothenfusser S, Schnurr M, Endres S, Eigler A. Maturedendritic cells derived from human monocytes within48 hours: a novel strategy for dendritic cell differentia-tion from blood precursors. J Immunol 2003;170:4069-76; PMID:12682236; http://dx.doi.org/10.4049/jimmunol.170.8.4069

44. Sallusto F, Lanzavecchia A. Efficient presentation of solubleantigen by cultured human dendritic cells is maintained bygranulocyte/macrophage colony-stimulating factor plusinterleukin 4 and downregulated by tumor necrosis factoralpha. J Exp Med 1994; 179:1109-18; PMID:8145033;http://dx.doi.org/10.1084/jem.179.4.1109

45. Day D, Pham K, Ludford-Menting MJ, Oliaro J, IzonD, Russell SM, Gu M. A method for prolonged imag-ing of motile lymphocytes. Immunol Cell Biol 2009;87:154-8; PMID:18982018; http://dx.doi.org/10.1038/icb.2008.79

46. Adams J, Palombella VJ, Sausville EA, Johnson J, Des-tree A, Lazarus DD, Maas J, Pien CS, Prakash S, ElliottPJ. Proteasome inhibitors: a novel class of potent andeffective antitumor agents. Cancer Res 1999; 59:2615-22; PMID:10363983

47. Lightcap ES, McCormack TA, Pien CS, Chau V,Adams J, Elliott PJ. Proteasome inhibition measure-ments: clinical application. Clin Chem 2000; 46:673-83; PMID:10794750

e1038011-16 Volume 4 Issue 9OncoImmunology