<<

CancerTherapy: Preclinical

Combining Milatuzumab with Bortezomib, , or Improves Responses in Cell Lines Rhona Stein,1Mitchell R. Smith,2 Susan Chen,1Maria Zalath,1and David M. Goldenberg1

Abstract Purpose: The humanized anti-CD74 monoclonal antibody, milatuzumab, is in clinical evaluation for the therapy of multiple myeloma (MM). The ability of milatuzumab to increase the efficacy of bortezomib, doxorubicin, and dexamethasone was examined in three human CD74+ MM cell lines, CAG, KMS11, KMS12-PE, and one CD74-MM cell line, OPM-2. Experimental Design: Activity of milatuzumab as a monotherapy and combined with the drugs was evaluated by studying in vitro cytotoxicity, signaling and apoptotic pathways, and in vivo therapeutic activity in severe combined immunodeficient (SCID) mouse models of MM. Results: Given as a monotherapy, cross-linked milatuzumab, but not milatuzumab alone, yielded sig- nificant antiproliferative effects in CD74+ cells.The combination of cross-linked milatuzumab with bortezomib, doxorubicin, or dexamethasone caused more growth inhibition than either cross-linked milatuzumab or drug alone, producing significant reductions in the IC50 of the drugs when combined. Efficacy of combined treatments was accompanied by increased levels of apoptosis measured by increases of activated caspase-3 and hypodiploid DNA. Both milatuzumab and bortezomib affect the nuclear factor-nB pathwayinCAGMMcells. In CAG-or KMS11-SCIDxenograftmodels ofdisseminated MM, milatuzumab more than doubled median survival time, compared withup to a 33% increase in median survival withbortezomib but no significant benefit with doxorubicin. Moreover, combining milatuzumabandbortezomibincreasedsurvivalsignificantlycomparedwitheither treatmentalone. Conclusions:The therapeutic efficacies of bortezomib, doxorubicin, and dexamethasone are en- hanced in MM cell lines when given in combination with milatuzumab, suggesting testing these combinations clinically.

Multiple myeloma (MM) is the second most prevalent blood transplantation is neither curative nor applicable to all patients cancer after non–Hodgkin lymphoma (1, 2). It represents and improved complete response rates have been seen in recent f1%of all cancers and 2%of all cancer deaths. Approximately clinical trials, the National Comprehensive Cancer Network 50,000 Americans currently have myeloma, and the American issued treatment guidelines recently that include recommenda- Cancer Society estimates 19,920 new cases of myeloma and tions for the use of newer treatment options for myeloma 10,690 deaths in 2008. Currently available therapies for patients. and bortezomib regimens involving myeloma include and stem cell transplantation, combination therapy with doxorubicin and/or dexamethasone as well as new emerging therapies and combination therapy are now recommended in initial therapy protocols for regimens that are being tested in clinical trials (2, 3). High-dose transplant candidates and for treatment of relapsed/refractory followed by autologous stem cell transplantation is disease. Combination of melphalan and with currently the standard of care because of its high response rate thalidomide or bortezomib are recommended as initial therapy and relatively low morbidity and mortality. Because stem cell for nontransplant candidates. Thus, there has been consider- able progress in the field of myeloma therapy. Although cures have not been documented, molecular complete responses Authors’Affiliations: 1Garden State Cancer Center, Center for Molecular Medicine 2 have been achieved with some of the new therapies. However, and Immunology, Belleville, New Jersey and Fox Chase Cancer Center, relapses still occur after molecular complete response, usually Philadelphia, Pennsylvania Received 7/28/08; revised 12/24/08; accepted 1/12/09; published OnlineFirst after a longer period of event-free survival. 4/7/09. Despite the substantial progress, the unmet need for Grant support: USPHS grants P01-CA103985 and R01-CA109474 from the improved therapy of MM has led to the evaluation of cell National Cancer Institute, and grants from the Thomas and Agnes Carvel surface targets expressed by the myeloma cells. Although Foundation and the Walter and Louise Sutcliffe Foundation. The costs of publication of this article were defrayed in part by the payment of page antigens such as CD19 and CD20 are important targets for charges. This article must therefore be hereby marked advertisement in accordance non–Hodgkin lymphoma, they are expressed in only a with 18 U.S.C. Section 1734 solely to indicate this fact. minority of myeloma cases. In contrast, the cell surface protein Requests for reprints: Rhona Stein, Center for Molecular Medicine and CD74 is expressed at fairly high levels in nearly 90%of MM Immunology, 520 Belleville Avenue, Belleville, NJ 07109. Phone: 973-844-7012; clinical specimens but not on most normal human cells (4). In Fax: 973-844-7020; E-mail: [email protected]. F 2009 American Association for Cancer Research. addition, the humanized anti-CD74 monoclonal antibody, doi:10.1158/1078-0432.CCR-08-1953 milatuzumab (hLL1), has in vitro growth inhibitory effects on

Clin Cancer Res 2009;15(8) April 15, 2009 2808 www.aacrjournals.org Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2009 American Association for Cancer Research. Milatuzumab Improves Drug Therapy of Myeloma

2B8 (anti-CD20) was purified from hybridoma supernatant of cells ob- Translational Relevance tained from the American Type Culture Collection. B-B4 (anti-CD138) was purchased from BD Biosciences. The function of CD74 as a survival receptor, combined Immunophenotyping. Determination of antigen expression levels on with the expression of CD74 on malignant B cells and MM cells was done by indirect immunofluorescence assays using FITC- limited expression on normal tissues, implicate CD74 as a goat anti-mouse IgG, purchased from Invitrogen Corporation, as potential therapeutic target. Based on our previous preclin- described previously (9). All flow cytometry experiments were done and analyzed using a FACSCalibur (Becton Dickinson). ical studies, the anti-CD74 humanized monoclonal anti- In vitro 3-(4,5-dimethyl thiazol-2-yl)-2,5-diphenyl tetrazolium body, milatuzumab, is currently in clinical evaluation for bromide assay. The cytotoxicity assay was based on the method of therapy of multiple myeloma. In this article, we show the Mosmann (10). Briefly, cell lines were plated at 1 to 2 Â 104 cells per ability of milatuzumab to increase the efficacy of bortezo- well (100 AL) in 96-well plates, to which antibodies and/or drugs were

mib, doxorubicin, and dexamethasone in multiple myeloma added (100 AL). After incubation for 4 d at 37jC in a humidified CO2 cell lines and animal models. The results presented here (5%) incubator, 25 AL of 5.0 mg/mL 3-(4,5-dimethylthiazol-2-yl)-2,5- now suggest testing these combinations clinically, with diphenyltetrazolium bromide (MTT) were added, and the cells were the goal of improving the outcome for patients with multi- incubated for an additional 4 h at 37jC. Plates were then centrifuged A ple myeloma. and supernatants were removed. Pellets were dissolved using 100 L DMSO per well and absorbance was measured at 570 nm on a microplate reader (Molecular Devices). Because unlabeled milatuzu- mab was reported previously to require cross-linking for cytotoxic MM cell lines and therapeutic effects in MM models (5, 6). activity (6), goat anti-human IgG (GAH) was added to some of the Based on these preclinical studies, milatuzumab is currently in wells. Milatuzumab was used at a final concentration of 5 Ag/mL and A clinical evaluation for therapy of MM. Here, we provide in vitro GAH was used at 20 g/mL. Percent growth inhibition and IC50 values and in vivo data corroborating the therapeutic efficacy of were determined using 4 replicates. milatuzumab in human MM model systems and expand our DNA fragmentation. Flow cytometric analysis of cellular DNA was observations to show that this monoclonal antibody is more done after propidium iodide staining (11, 12). Cells were placed in 24- Â 5 effective than key therapeutic agents for MM treatment, well plates (1.5 to 3 10 cells per well) and treated with drugs (at concentrations indicated for each experiment) or mAbs (5 Ag/mL) in including bortezomib, doxorubicin, and dexamethasone, when the presence or absence of a second antibody (20 Ag/mL). Percent each agent is given alone. In addition, the efficacies of the apoptotic cells (hypodiploid cells) was determined after a chemotherapeutics are enhanced when given in combination 48-h incubation. with milatuzumab. Cleaved caspase-3. Cells were incubated in the presence or absence of the drugs (at concentrations indicated for each experiment) and/or mAbs for 48 h. Changes in the intracellular levels of cleaved caspase-3 were measured using FITC-conjugated rabbit antiactivated caspase-3 Materials and Methods (BD Bioscience) as per the manufacturer’s directions. Analyses were done on the FACSCalibur. Cells. The cell lines were obtained as follows: KMS11 and KMS12- Western blots. Cells were cultured in the presence or absence of the PE from Dr. T. Otsuki (Kawasaki Medical School, Okayama, Japan), mAbs and/or drugs for the indicated times, pelleted, washed thrice in CAG from Dr. Joshua Epstein (University of Arkansas, Fayetteville, AR), PBS, then lysed in ice cold radioimmunoprecipitation assay buffer and OPM-2 from Dr. Kenji Oritani (Osaka University, Osaka, Japan). containing 100 Ag/mL phenylmethylsulfonyl fluoride and 1 Ag/mL The cells were grown as suspension cultures in DMEM (Life aprotinin on ice for 30 min. Bortezomib was used at 1 ng/mL. Technologies), supplemented with 10%fetal bovine serum, penicillin Milatuzumab was used at a final concentration of 5 Ag/mL and GAH (100 U/mL), streptomycin (100 Ag/mL), and L-glutamine (2 mmol/L). was used at 20 Ag/mL. The lysates were centrifuged at 1,200 Â g for Antibodies and drugs. Antibodies LL1 [the parental murine molec- ular antibody (mAb) formerly called EPB-1; ref. 7], milatuzumab (also called hLL1 or IMMU-115; the humanized IgG1 version; ref. 6), and hMN-14 (labetuzumab, anti-CD66e, anticarcinoembryonic antigen or -CEACAM5 IgG1, used here as a humanized isotype control; ref. 8) were provided by Immunomedics, Inc. Bortezomib was purchased from Millennium Pharmaceuticals. Doxorubicin and dexamethasone were purchased from Florida Infusion. The murine monoclonal antibody

Table 1. Antigen expression on MM cell lines

Control-Ag8 % Positive CD138-B-B4 (geometric mean FL) CD20-2B8 CD74-LL1 CAG 5.4 (6.7) 4.4 (6.0) 99.5 (123.3) 24.1 (11.7) KMS11 2.2 (4.9) 2.1 (4.9) 74.8 (22.0) 14.2 (7.5) KMS11-PE 1.8 (2.4) 2.7 (2.3) 86.7 (17.0) 55.2 (10.0) OPM-2 1.4 (5.4) 1.2 (5.3) 0.7 (5.2) 99.0 (255.0) Fig. 1. Effects of milatuzumab and GAH second antibody on proliferation of cell lines. Anti-proliferative effects of milatuzumab were determined by MTTassays. Cells were cultured with 5 Ag/mL milatuzumab with or without 20 Ag/mL second NOTE: Antigen expression was determined by flow cytometric antibody for cross-linking to mimic the role of effector cells or cross-linking analyses. molecules present in vivo. Percent inhibition is calculated relative to untreated control cells. Columns, mean of four replicates; bars, SD. *, not determined.

www.aacrjournals.org 2809 Clin Cancer Res 2009;15(8) April15, 2009 Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2009 American Association for Cancer Research. CancerTherapy: Preclinical

Fig. 2. In vitro cytotoxicity of bortezomib, dexamethasone, and doxorubicin with and without milatuzumab in CAG. Cells were cultured with the drugs, milatuzumab (5 Ag/mL), and GAH second antibody (20 Ag/mL) as shown, then assayed using MTT. Percent inhibition of absorbance at 570 nm, relative to media only, was measured. o, solid line, drug only; D, dashed line, drug + milatuzumab; 5, dotted line, drug + GAH; ., solid line, drug + milatuzumab + GAH. Points, mean of four replicates; bars, SD.

15 min at 4jC. Supernatants were removed and separated on 10%to Statistical considerations. For in vitro studies, means and SDs were 20%SDS-PAGE (Tris-HCl, Ready Gel; Bio-Rad) followed by transfer to calculated. Differences between treatment groups were compared using Immobilon-P membrane (Millipore). Membranes were blocked with the Student’s t test. In the in vivo studies, overall survival was evaluated 5%nonfat dry milk-0.05%Tween 20 (Sigma-Aldrich Corp.) in PBS for graphically by the methods of Kaplan and Meier. Pair-wise comparisons 1 h and then incubated overnight in the presence of antibody at 4j C. between specific groups were made using log-rank statistics. Signifi- Primary antibodies were from Cell Signaling Technology and were used cance for all tests was declared at P values of <0.05. at 1:1,000 dilution, except Bid from Trevigen also used at 1:1,000, and Mcl-1 from BD Pharmingen and Bcl-2 from Santa Cruz Biotechnology used at 1:500. The blots were incubated in secondary antibody Results conjugated to horseradish peroxidase (Amersham Biosciences) for In vitro 1 h, followed by chemiluminescent detection (Amersham). cytotoxicity of milatuzumab on MM cell lines. The In vivo studies in severe combined immunodeficient mice bearing cytotoxicity of milatuzumab was investigated on cell lines disseminated MM tumors. For studies on the therapeutic effect of the derived from MM patients, alone or cross-linked with GAH. mAbs and drugs in CAG-bearing severe combined immunodeficient CAG, KMS11, and KMS12-PE are positive for CD138 (synde- (SCID) mice (female, 6- to 8-wk-old; Charles River Laboratories), the can-1) and CD74, and CD20 negative. OPM-2 is CD138 mice were immunosuppressed by pretreatment with and positive but does not express cell surface CD74 or CD20. 6 3 d before i.v. injection of 5 to 10 Â 10 tumor cells, Antigen expression of these cell lines is tabulated in Table 1. As as previously described (6). Pretreatment with fludarabine and shown in Fig. 1, the CD74+ cell lines are sensitive to killing by cyclophosphamide was not used for implantation of KMS11 or cross-linked milatuzumab but not milatuzumab in the absence KMS12-PE. Dosing is described in figure legends. Mice were examined of cross-linking, or the cross-linking antibody (GAH) alone. For daily for signs of distress or hind leg paralysis, and weighed weekly. Paralysis of the hind legs or a weight loss of >20%was used as the example, in the representative data shown, milatuzumab+GAH F survival end point. Animals were euthanized at these end points. yielded 38.0% 8.9%inhibition (P = 0.0025) in CAG cells Experiments used groups of 8 to 10 mice. Animal studies were done compared with 5.5% F 12.8%inhibition in OPM-2 cells under protocols approved by the Institutional Animal Care and Use (P = 0.54) relative to untreated cells. The absence of cytotoxicity Committee. of cross-linked milatuzumab in OPM-2, which is CD74-, shows

Table 2. Combining milatuzumab with drugs in MM cell lines

IC50 (ng/mL) CAG KMS11 KMS12-PE OPM-2

- +-+-+-+ Doxorubicin 7.4 F 2.1 1.1 F 0.7 16.7 F 3.6 0.13 F 0.10 62.1 F 2.6 5.8 F 4.5 28.1 F 4.1 27.9 F 4.0 6.7-fold, P = 0.0063 128-fold, P = 0.0027 10.7-fold, P = 0.0205 Unchanged, P = 0.6360 Bortezomib 0.40 F 0.08 0.12 F 0.06 0.45 F 0.04 0.18 F 0.02 1.36 F 0.04 0.50 F 0.14 0.53 F 0.04 0.52 F 0.02 3.4-fold, P = 0.0015 2.5-fold, P = 0.0002 2.7-fold, P = 0.0006 Unchanged, P = 0.6953 Dexamethasone 20,038 F 550 145 F 86 >40,000 * >40,000 76 F 99 ND ND 13.8-fold, P = 0.0059

NOTE: IC50 values were determined using MTT cytotoxicity assays. Cells were incubated with the indicated drugs in the absence (-) or presence (+) of milatuzumab and GAH for 4 d. Abbreviation: ND, not determined. * IC50 value were not calculated because the combination of milatuzumab plus GAH yielded z50% inhibition of proliferation relative to untreated cells in the absence of drug.

Clin Cancer Res 2009;15(8) April 15, 2009 2810 www.aacrjournals.org Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2009 American Association for Cancer Research. Milatuzumab Improves Drug Therapy of Myeloma

specificity. Thus, cross-linked milatuzumab specifically and done to examine the effects of the drugs, alone and effectively kills CD74+ MM cells in vitro. combined with milatuzumab or milatuzumab+2nd antibody In vitro cytotoxicity of bortezomib, dexamethasone, and (GAH). Representative titrations are shown in Fig. 2 for CAG doxorubicin with and without milatuzumab. MTT assays were (data not shown for KMS11, KMS12-PE, and OPM-2). Results

Fig. 3. Apoptotic effect of mAbs on MM cell lines. Induction of apoptosis was evaluated by flow cytometry determination of (A) hypodiploid DNA and (B)activated caspase-3. Cells were incubated with drugs alone, milatuzumab alone, or combinations of drug + milatuzumab with and without a second antibody for cross-linking for 48 h. For determination of hypodiploid DNA, staining was done with propidium iodide. Changes in the intracellular levels of cleaved caspase-3 weremeasuredusing FITC-conjugated rabbit anti-activated caspase-3. Analyses were done on the FACSCalibur. o, solid line, drug only; D, dashed line, drug + milatuzumab; 5, dotted line, drug + GAH; ., solid line, drug + milatuzumab + GAH. Columns, mean of three replicates; bars, SD (some points are hidden by the symbol).

www.aacrjournals.org 2811 Clin Cancer Res 2009;15(8) April15, 2009 Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2009 American Association for Cancer Research. CancerTherapy: Preclinical

DNA by these drugs, with increases by the drugs alone remaining within 12%of baseline. In all cell lines tested, milatuzumab that was not cross-linked with second antibody did not affect the percent of hypodiploid DNA induced by the drug alone, percentages were generally within 1%to 5%of the drug only values. Similar results were obtained in the controls when second antibody was given in the absence of milatuzu- mab. However, milatuzumab + GAH led to statistically significant increased levels of hypodiploid DNA (P < 0.05). For example in CAG, levels of sub-G0 DNA were 8.4% F 0.5%, 8.0% F 0.3%, and 9.0% F 0.5%after treatment with 0.1 ng/ Fig. 4. Western blot analyses of lysates of CAG cell treated with milatuzumab and mL bortezomib alone, 0.1 ng/mL bortezomib + milatuzumab, bortezomib. CAG cells were cultured in the presence or absence of the indicated and 0.1 ng/mL bortezomib + GAH, respectively (no significant reagents for 30 min or 3 h, pelleted, then washed thrice in PBS, and lysed in ice cold radioimmunoprecipitation assay buffer containing 100 Ag/mL phenylmethylsulfonyl differences; P > 0.05). However, 0.1 ng/mL of bortezomib + A A fluoride and 1 g/mL aprotinin. Milatuzumab was used at 5 g/mL, GAH milatuzumab + GAH increased sub-G0 DNA to 34.5% F 0.3% A at 20 g/mL, and bortezomib at 1ng/mL. Equivalent amounts of protein were (P < 0.00001, versus drug alone). analyzed by Western blots and actin levels were compared as a control for protein loading. Lanes are as follows: 1, untreated control cells; 2, milatuzumab; 3, Induction of activated caspase-3 was similarly analyzed by milatuzumab + GAH; 4, milatuzumab + GAH + bortezomib; and 5, bortezomib. flow cytometry after incubation with increasing doses of bortezomib and doxorubicin. Induction of activated caspase-3 was observed (Fig. 3B). KMS12-PE was the least sensitive cell from these titrations were used to calculate IC50 values of the line in this test. Cross-linked milatuzumab yielded statistically drugs alone and in the presence of milatuzumab+GAH, significant increases in the induction of activated caspase-3 in shown in Table 2. Cross-linked milatuzumab, but not CAG and KMS11. For example, in KMS11 levels of activated milatuzumab alone, yielded significant reductions in the caspase-3 were 11.4% F 0.6%, 8.0% F 0.7%, and 13.1% F IC50 values of bortezomib and doxorubicin in CAG, KMS11, 0.3%after treatment with 0.1 ng/mL bortezomib alone, 0.1 ng/ and KMS12PE. For example, in CAG, cross-linked milatuzu- mL bortezomib + milatuzumab, and 0.1 ng/mL bortezomib + mab led to decreases of 6.7- 3.4-, and 13.8-fold in the IC50 GAH, respectively. However, 0.1 ng/mL of bortezomib + values of doxorubicin, bortezomib, and dexamethasone, milatuzumab + GAH yielded a value of 41.6 F 2.1%( P = respectively. Cross-linked milatuzumab did not affect the 0.0009, versus drug alone). IC50 values in the CD74-negative cell line, OPM-2. KMS11 Increases were not seen in OPM-2 and KMS12-PE, and KMS12-PE were relatively insensitive to dexamethasone. presumably due to the lack of CD74 in OPM-2 and the low The IC50 was not reached using dexamethasone concentra- caspase-3 induction by the drugs in KMS12-PE. These data tions up to 40 Ag/mL. Dexamethasone was not tested on show that doxorubicin and bortezomib induce apoptosis, as OPM-2. measured here by induction of hypodiploid DNA and Thus, the combination of cross-linked milatuzumab (mila- activated caspase-3. Moreover, cross-linked milatuzumab tuzumab+GAH) with bortezomib, doxorubicin, or dexameth- amplifies this effect, corroborating the benefit of using these asone causes more growth inhibition than either cross-linked agents in combination. milatuzumab or drug alone. This can be clearly observed by We have begun to examine the intracellular interactions of examining single submaximal points on the drug titration antibody and drug by investigating signaling and apoptotic curves. For example, growth inhibition of CAG cells was 44.6% pathways in CAG cells using Western blot analyses. No F 18.6%after incubation with 0.55 ng/mL bortezomib and alterations in Bid, Bax, Bcl-2, nor Mcl-1 were observed after 34.4% F 17.1%with milatuzumab+GAH, whereas incubation exposure of CAG cells to cross-linked milatuzumab (data not with these agents in combination increases the inhibition to shown). Thus, signaling through the extrinsic, but not the 84.0% F 1.4%( P < 0.025 versus either 0.55 ng/mL bortezomib mitochondrial, apoptotic pathways is suggested. or milatuzumab+GAH). CD74 activation by its ligand macrophage migration- Induction of apoptosis and signaling. Induction of apoptosis inhibitory factor signals through nuclear factor-nB (NF-nB) was evaluated by determining percentage of hypodiploid DNA in chronic lymphocytic leukemia cells (13). Moreover, stim- and induction of activated caspase-3 on the panel MM cell ulation of chronic lymphocytic leukemia cells with C-16, an lines. Cells were cultured with increasing doses of bortezomib agonistic anti-CD74 mAb, leads to NF-nB activation, and in- or doxorubicin in the presence or absence of milatuzumab or creased survival in chronic lymphocytic leukemia cells. Here, milatuzumab+GAH for 48 h, followed by DNA staining with we find decreases in phosphorylated InBa and phosphorylated propidium iodide. Cells were analyzed by flow cytometry, and NF-nB 30 minutes after incubation of CAG cells with cross- positive fluorescence below the G0-G1 region (hypodiploid) linked milatuzumab, with recovery of phosphorylated NF-nB represents DNA fragmentation and is a measure of apoptosis. to control levels at 3 hours (Fig. 4). In contrast, bortezomib The percentage of hypodiploid DNA induced by these treat- increased phosphorylated InBa and phosphorylated NF-nBat ments is shown in Fig. 3A. Baseline values of sub-G0 DNA in 3 hours, a result in contrast to the expected decreases in these untreated cells ranged from 2.7%to 8.3%.Bortezomib and values. The combination of cross-linked milatuzumab plus doxorubicin used as single agents increased hypodiploid DNA a bortezomib yielded an intermediate level of phosphorylated further 20%to 35%in CAG and KMS11, when used at NF-nB at 30 min. These data show that both milatuzumab concentrations up to 10 and 1,000 ng/mL, respectively. KMS12- binding to CD74 and bortezomib treatment induce intracel- PE and OPM-2 were less sensitive to induction of hypodiploid lular alterations of the NF-nB signal pathway in CAG

Clin Cancer Res 2009;15(8) April 15, 2009 2812 www.aacrjournals.org Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2009 American Association for Cancer Research. Milatuzumab Improves Drug Therapy of Myeloma

Fig. 5. Therapeutic efficacy of bortezomib plus milatuzumab in MM-bearing SCID mice. A, SCID mice were immunosuppressed by pretreatment with fludarabine phosphate and cyclophosphamide 3 d before injection of CAG cells.Treatments were given twice weekly for 4 wk, starting 1d after injection of the CAG cells. SCID mice bearing CAG- (B)orKMS11-(C) were treated with bortezomib, milatuzumab, and mixtures of milatuzumab + bortezomib. Treatments for experiments shown in B and C were given as 2 i.p. doses/wk for 3 wk, initiated on day 5 after injection of tumor cells.

myeloma cells, although in opposite directions. Future single agent, 1.0 mg/kg bortezomib caused an apparent studies examining total IKBa levels (e.g., by electrophoretic treatment-related death of 1 of 10 mice 12 days after mobility shift assays) are necessary to clarify the observed administration of the drug, and given in combination with effects on the NF-nB pathway under the various treatment 100 Ag milatuzumab, 2 of 10 mice died of treatment-related conditions. death (days 8 and 13). The 0.5 mg/kg dose was well-tolerated In vivo therapeutic activity of milatuzumab as a monotherapy with no loss of body weight. Median survival in untreated and combined with drugs. A dose-response therapy study control mice in this study was 33 days. Bortezomib alone in mice bearing the CAG cell line is shown in Fig. 5A and increased MST to 40 and 44 days, representing MST increases of Table 3. Milatuzumab was given at 30, 100, and 300 Ag/ 21.2%( P < 0.0001) and 33.3%( P = 0.0021) at 0.5 and 1.0 mg/kg, injection, twice weekly for 4 weeks, starting 1 day after respectively. Treatment with milatuzumab (100 Ag per mouse) injection of CAG cells. Animals were monitored for survival increasedtheMSTto73days.Whenbortezomiband and hind leg paralysis, and body weights were measured milatuzumab treatments were combined, MSTs were increased weekly. Results of this study showed that treatment with further to 79 and 93 days for 0.5 and 1.0 mg/kg bortezomib, milatuzumab yielded a significant survival benefit in this respectively (P = 0.0441 and P = 0.0065 for the 100 Ag model. Median survival times (MST) in both untreated CAG- milatuzumab+1.0 mg/kg bortezomib combination versus mila- bearing mice and mice given an unreactive isotype control tuzumab alone and 1.0 mg/kg bortezomib alone, respectively). mAb were 42 days. The median survival in the milatuzumab- The tumor growth curves for KMS11-bearing SCID mice treated groups was at least doubled, ranging between 84 and treated with bortezomib and milatuzumab are shown in 103 days (P < 0.0001 versus untreated). A dose-response was Fig. 5C and Table 5. Based on the results of the CAG study, not observed in this dose range. bortezomib was given at 0.8 mg/kg/dose. Milatuzumab was The effects of combining doxorubicin or bortezomib with administered at 2 dose levels, 35 and 100 Ag/dose. The milatuzumab were evaluated in SCID mice bearing CAG, 0.8 mg/kg dose was well-tolerated with no loss of body KMS11, and KMS12-PE cells. Bortezomib dosing was selected weight. Median survival in untreated KMS11 control mice in from reports of maximum tolerated dose values in SCID mice this study was 54 days. Bortezomib alone increased MST to (14). Treatments were given as 2 i.p. doses/week for 3 weeks, 61 days (P = 0.0667 versus untreated), representing a MST initiated on day 5 after injection of tumor cells. Figure 5B and increase of 13.0%. Treatment with 35 Ag milatuzumab alone Table 4 show the tumor growth curves for CAG-bearing SCID also increased MST to 61 days (P = 0.1518 versus untreated). mice treated with bortezomib and milatuzumab. Given as a MST was increased further to 86 days for the combination of

www.aacrjournals.org 2813 Clin Cancer Res 2009;15(8) April15, 2009 Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2009 American Association for Cancer Research. CancerTherapy: Preclinical

Table 3. Therapeutic efficacy of bortezomib plus milatuzumab in MM-bearing SCID mice

Group Median survival (d) P vs untreated % MST increase* Untreated 42 30 Ag milatuzumab 91 <0.0001 116.7% 100 Ag milatuzumab 103 <0.0001 145.2% 300 Ag milatuzumab 84 <0.0001 100.0% 300 Ag hMN-14 42 0.6850 0.0%

NOTE: Statistical summaries for data shown in Fig. 5. CAG-bearing SCID mice were given milatuzumab at 30, 100, and 300 Ag/injection, twice weekly for 4 wk, starting 1 d after injection of the CAG cells. Control animals were either left untreated or given hMN-14 (isotype control mAb at 300 Ag/injection by the same schedule). Survival data are plotted in Fig. 5A. *Relative to untreated control group.

35 Ag milatuzumab and 0.8 mg/kg bortezomib, representing yielded a minimum increase in median survival of 64%. a survival increase of 59.3%relative to untreated mice ( P < Statistical significance was not reached in this study, presum- 0.0001). The MST increase due to combination of 35 Ag ably due to the slow progression of tumor in this model. milatuzumab and 0.8 mg/kg bortezomib was statistically P values were 0.0962, 0.2126, and 0.1603 for milatuzumab + significant compared with untreated and both single agent bortezomib versus untreated, bortezomib alone, and milatu- treatment groups (P = 0.0040 and P = 0.0128 for the 35 Ag zumab alone, respectively. milatuzumab+0.8 mg/kg bortezomib combination versus Thus, when given as a monotherapy, milatuzumab is more milatuzumab alone and bortezomib alone, respectively). effective than bortezomib in the CAG and KMS11 MM models. There were 5 long-term survivors (>4 months) in the 35 Ag Moreover, combining milatuzumab and bortezomib increases milatuzumab+0.8 mg/kg bortezomib treatment group, com- survival compared with either treatment alone in the CAG, pared with 1 or none in the control and single agent KMS11, and KMS12-PE models. Importantly, these studies groups at these doses. Increasing the milatuzumab dose to confirm the therapeutic efficacy of milatuzumab previously 100 Ag/dose increased its effect. Median survival was not reported in MC/CAR-bearing SCID mice (6) in additional MM reached at 4 months; there were 7 long-term survivors each cell lines. with 100 Ag milatuzumab, and 100 Ag milatuzumab+0.8 mg/ The efficacy of milatuzumab also was compared with that of kg bortezomib. doxorubicin and a mixture of doxorubicin and milatuzumab in KMS12-PE is a slower growing tumor than CAG or KMS11. CAG-bearing SCID mice. Doxorubicin was administered at its Median survival in untreated KMS12-PE–bearing SCID mice is maximum tolerated dose in this model, 35 Ag, and at one- 110 days (survival curve not shown). At 6 months posttumor fourth the maximum tolerated dose, 8.65 Ag. Treatments were injection, 2 of 9 mice were still alive in the untreated group, given as a single i.p. dose 5 days after injection of tumor cells. compared with 3 of 9 given 0.8 mg/kg bortezomib (124 days Treatment with a single dose of milatuzumab (341 Ag per median survival), 3 of 9 given 100 Ag milatuzumab (110 days mouse) increased the MST from 33 to 69 days, with no median survival), and 6 of 9 given the combined treatment significant difference from the milatuzumab plus doxorubicin (MST not reached at 180 days). Thus, the combined treatment group, or the 100 Ag milatuzumab/mouse twice weekly for

Table 4. CAG-bearing SCID mice were treated with bortezomib, milatuzumab, and mixtures of milatuzumab + bortezomib

Group Median P vs %MST P vs 100 Mg %MST P vs %MST survival (d) untreated increase* milatuzumab increasec corresponding increaseb bortezomib concentration Untreated 33 0.5 mg/kg bortezomib 40 <0.0001 21.2% 1.0 mg/kg bortezomib 44 <0.0021 33.3% 100 Ag milatuzumab 73 <0.0001 121.2% 100 Ag milatuzumab + 79 <0.0001 139.4% 0.3110 8.2% <0.0001 97.5% 0.5 mg/kg bortezomib 100 Ag milatuzumab + 93 <0.0238 181.8% 0.0441 27.4% <0.0065 132.5% 1.0 mg/kg bortezomib

NOTE: Treatments were given as 2 i.p doses/wk for 3 wk, initiated on day 5 after injection of tumor cells. Survival data are plotted in Fig. 5B. *Relative to untreated control group. cRelative to 100 Ag milatuzumab group. bRelative to corresponding bortezomib concentration group.

Clin Cancer Res 2009;15(8) April 15, 2009 2814 www.aacrjournals.org Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2009 American Association for Cancer Research. Milatuzumab Improves Drug Therapy of Myeloma

Table 5. KMS-11-bearing SCID mice were treated with bortezomib, milatuzumab, and mixtures of milatuzumab + bortezomib

Group Median P vs %MST P vs P vs survival (d) untreated increase* corresponding bortezomibb milatuzumab concentrationc Untreated 54 0.8 mg/kg bortezomib 61 0.0667 13.0% 100 Ag milatuzumab >125 <0.0001 131.5% 100 Ag milatuzumab + 0.8 mg/kg bortezomib >125 <0.0001 131.5% 35 Ag milatuzumab 61 0.1518 13.0% 0.9542 8.2% 35 Ag milatuzumab + 0.8 mg/kg bortezomib 86 <0.0001 59.3% 0.0040 27.4%

NOTE: Treatments were given as 2 i.p. doses/wk for 3 wk, initiated on day 5 after injection of tumor cells. Survival data are plotted in Fig. 5C. * Relative to untreated control group. cRelative to 35 Ag milatuzumab group. bRelative to 0.8 mg/kg bortezomib group.

3 weeks dose schedule, shown in Fig. 5B. Given alone, cross-linking agents are necessary for milatuzumab activity in doxorubicin yielded little or no effect on survival compared in vitro studies but are not required for milatuzumab activity with untreated animals (data not shown). in vivo. Cross-linking with secondary antibodies is believed to function by generating stronger and/or more sustained Discussion signals than simple ligation of surface antigens with primary mAbs to the antigens alone (12). A requirement for extensive CD74 (invariant chain, Ii) is a type-II transmembrane cross-linking of surface molecules has been noted by several glycoprotein that associates with the major histocompatibilty groups studying lymphoma cell growth inhibition, class II a and h chains and directs the transport of the ahIi arrest, and/or apoptosis in vitro. Specifically, hypercross- complexes to endosomes and lysosomes (15–17). Because the linking with secondary antibodies has been used for in vitro chaperone function of CD74 was thought to be its primary evaluations of anti-CD20, anti-CD19, anit-CD22, anti–major function, it was not anticipated that an anti-CD74 mAb would histocompatibilty class II, anti-sIgM, and anti–APO-1 mAbs possess growth-inhibitory activity. However, it is now under- (12, 24–27). However, we and others have observed that stood that CD74 is the cellular receptor for the proinflamma- cross-linking by GAH is not needed for in vivo activity. tory cytokine, macrophage migration-inhibitory factor (18), Presumably the function of the secondary antibodies in vitro and that the binding of macrophage migration-inhibitory factor is fulfilled in vivo by other molecules present in the cellular to cell surface CD74 initiates a signaling cascade resulting in environment. These may include cross-linking with Fc- proliferation and survival (19). Shachar and coworkers (20) receptor–expressing cells (e.g., macrophages, monocytes, or have shown that after migration-inhibitory factor binding to dendritic cells), or other molecules involved in the signaling the CD74 extracellular domain, the cytosolic region of CD74 is pathway of the target antigen that cause further aggregation cleaved by a two-step process. The cytosolic fragment then after preliminary stimulation by the primary mAb. translocates to the cell nucleus, resulting in activation of NF-nB Based on the preclinical evidence of activity, milatuzumab (21), and induction of a survival cascade in which interleukin- is currently in clinical evaluation for therapy of MM, non– 8 (IL-8) levels are elevated, as well as levels of the antiapoptotic Hodgkin lymphoma, and chronic lymphocytic leukemia. In protein, Bcl-2 (13). In addition, Helicobacter pylori was shown this article, we showed the ability of milatuzumab to increase to bind CD74 on gastric epithelial cells and stimulate NF-nB the efficacy of bortezomib, doxorubicin, and dexamethasone and IL-8 production, thus corroborating the involvement of in MM cell lines. In vitro, cross-linked milatuzumab yielded these factors in the signal cascade initiated by CD74 activation significant cytotoxicity on CD74-positive MM cell lines when (22, 23). Milatuzumab apparently acts as an antagonistic given as a single agent and caused significant reductions in antibody, blocking this signal cascade by binding to cell-surface the IC50 values of the anti-MM drugs. In vivo, milatuzumab CD74. Using B-CLL patient specimens, Binsky et al. (13) given as a monotherapy was more effective than bortezomib showed that milatuzumab specifically blocked macrophage or doxorubicin, whereas the combination of milatuzumab migration-inhibitory factor–induced up-regulation of Bcl-2 and bortezomib was more effective than either agent alone. mRNA levels, inhibited IL-8 transcription, and increased the Bortezomib is a -inhibitor, whose mechanism of percentage of apoptotic cells. action is partly mediated through NF-nB (reviewed in refs. 28, The humanized anti-CD74 monoclonal antibody, milatu- 29). NF-nB is constitutively active in MM as well as other zumab, causes specific growth inhibition and induction of tumors and, by influencing transcription of multiple gene apoptosis in B-cell lines in the presence of a second cross- targets, supports proliferation and suppresses apoptosis. How- linking antibody. In addition, significant survival extensions ever, differential responses of NF-nB to bortezomib have been were observed in non–Hodgkin lymphoma– and MM- noted in MM as well as other hematologic and nonhematologic bearing SCID mice treated with naked milatuzumab without cancers and may contribute to heterogeneous responses to the need for an exogenous cross-linking agent (6). Thus, bortezomib therapy. Failure of bortezomib to decrease NF-nB

www.aacrjournals.org 2815 Clin Cancer Res 2009;15(8) April15, 2009 Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2009 American Association for Cancer Research. CancerTherapy: Preclinical activation or to actually cause increased NF-nB activation have markedly more effective than doxorubicin, and the doxoru- been recently reported in MM (30), mantle cell leukemia (31), bicin + milatuzumab treatment protocol administered did and endometrial (32), hepatocellular (33), and other carcino- not significantly improve the efficacy over that of milatuzu- mas, and therefore may be more prevalent than previously mab alone. However, it should be noted that other protocols thought. For example, in a recent study of primary tumor cells of doxorubicin administration, such as fractionated or from MM patients, Markovina et al. (30) found that all patients’ multiple dosing, have not yet been evaluated in this model, cells constitutively expressed NF-nB activity, and that bortezo- and may yield some efficacy in vivo. Moreover, in light of mib effectively blocked proteasome activity in all. However, in results of a phase III study in relapsed or refractory MM, 10 of 14 cases, bortezomib failed to inhibit NF-nB activity, and which showed that the combination of pegylated liposomal in 8 of these cases bortezomib augmented NF-nB activity. doxorubicin plus bortezomib is superior to bortezomib Thus, the combination of bortezomib with an agent such as monotherapy (35), combinations of milatuzumab with milatuzumab, which can independently down-regulate NF-nB, bortezomib plus doxorubicin will be tested. may be especially relevant. In conclusion, the function of CD74 as a survival receptor, Bortezomib may cause cell death through additional paths combined with the expression of CD74 on malignant B cells and not dependent on NF-nB. Thus, various scenarios can be limited expression on normal tissues, implicate CD74 as a envisioned to explain the enhancement of cytotoxicity by the potential therapeutic target. In vivo, milatuzumab alone or in combination of milatuzumab and bortezomib. If the tumor has combination with the drugs evaluated in this report is effective in constitutive NF-nB that is resistant to bortezomib, then MM therapy. Moreover, the therapeutic efficacies of bortezomib, bortezomib toxicity will only be due to apoptotic effects that doxorubicin, and dexamethasone are enhanced when given in are independent of NF-nB activation (32, 33). Down-regulation combination with milatuzumab. Although gaps remain in our of NF-nB by milatuzumab will add to the toxicity by understanding of the mechanisms of cytotoxicity of milatuzumab, introducing a second mechanism of killing. Alternatively, if it seems likely that interference with the survival functions of the tumor is one in which bortezomib does overcome NF-nB CD74 is involved. Future studies will focus on this issue as well as activation, then the combination of milatuzumab and borte- gaining a greater understanding of milatuzumab and drug zomib would be expected to yield greater decreases in NF-nB interactions at the subcellular level, to aid in the rational design activity, again leading to greater cell killing. Because myeloma of combined modality regimens. cell lines differ in baseline signaling characteristics (34), CD74 expression, and activation of tBid after signaling through caspase-8, as well as NF-nB response to bortezomib, a more Disclosure of Potential Conflicts of Interest detailed evaluation of concentration and time effects on a panel of cell lines will be necessary to fully characterize CD74 and D.M. Goldenberg declares financial interset (officer, stock, patents) in Immuno- medics, Inc.The other authors declare no financial conflict. bortezomib interactions at the subcellular level. Although our in vitro studies showed the sensitivity of CD74+ MM cell lines to doxorubicin, no significant effect Acknowledgments was observed when doxorubicin was used as a single agent in CAG-bearing SCID mice. Milatuzumab monotherapy was We thank Indira Joshi for excellent technical assistance.

References 1. Cancer Facts and Figures. Atlanta: American Cancer 10. Mosmann T. Rapid colorimetric assay for cellular 17. Roche PA,Teletski CL, Stang E, Bakke O, Long EO. Society, Inc.; 2008. growth and survival: application to proliferation and Cell surface HLA-DR-invariant chain complexes are 2. Kyle RA, Rajkumar SV. Multiple myeloma. Blood cytotoxicity assays. J Immunol Methods 1983;65: targeted to endosomes by rapid internalization. Proc 2008;111:2962 ^ 72. 55 ^ 63. Natl Acad Sci U S A 1993;90:8581 ^ 5. 3. Barlogie B, ShaughnessyJ,Tricot G, et al.Treatment of 11. Nicoletti I, Migliorati G, Pagliacci MC, Grignani F, 18. Leng L, Metz CN, FangY, et al. MIF signal transduc- multiple myeloma. Blood 2004;103:20 ^ 32. Riccardi C. A rapid and simple method for measur- tion initiated by binding to CD74. J Exp Med 2003; 4. Burton JD, Ely S, Reddy PK, et al. CD74 is expressed ing thymocyte apoptosis by propidium iodide stain- 197:14 67 ^ 76. by multiple myeloma cells and is a promising target for ing and flow cytometry. J Immunol Methods 1991; 19. Starlets D, GoreY, Binsky I, et al. Cell surface CD74 therapy. Clin Cancer Res 2004;10:6606 ^ 11. 139:271 ^ 9. initiates a signaling cascade leading to cell prolifera- 5. Stein R, Mattes MJ, CardilloTM, et al. CD74: a new 12. Shan D, Ledbetter JA, Press OW. Apoptosis of ma- tion and survival. Blood 2006;107:4807 ^ 16. candidate target for the immunotherapy of hematologi- lignant human B cells by ligation with monoclonal 20. Matza D, Lantner F, BogochY,Flaishon L, Hershkoviz calmalignancies.Clin CancerRes 2007;13:5556 ^ 63s. antibodies. Blood 1998;91:1644 ^ 52. R, Shachar I. Invariant chain induces B cell maturation 6. SteinR,QuZ,CardilloTM,etal.Anti-proliferativeac- 13. Binsky I, Haran M, Starlets D, et al. IL-8 secreted in a inaprocess thatisindependentof its chaperonicactivity. tivity of a humanized anti-CD74 monoclonal antibody, macrophage migration-inhibitory factor- and CD74- Proc Natl Acad Sci US A 2002;99:3018^23. hLL1, on B-cell malignancies. Blood 2004;104: dependent manner regulates B cell chronic lympho- 21. Becker-Herman S, Arie G, Medvedovsky H, Kerem 3705 ^ 11. cytic leukemia survival. Proc Natl Acad Sci U S A A, Shachar I. CD74 is a member of the regulated intra- 7. Pawlak-Byczkowska EJ, Hansen HJ, Dion AS, 2007;104:13408^13. membrane proteolysis-processed protein family. Mol Goldenberg DM. Two new monoclonal antibodies, 14. LeBlanc R, Catley LP,HideshimaT, et al. Proteasome Biol Cell 2005;16:5061^9. EPB-1 and EPB-2, reactive with human lymphoma. inhibitor PS-341 inhibits human myeloma cell growth 22. Beswick EJ, Bland DA, Suarez G, Barrera CA, Fan Cancer Res 1989;49:4568 ^ 77. in vivo and prolongs survival in a murine model. X, Reyes VE. Helicobacter pylori binds to CD74 on 8. Sharkey RM, Juweid M, ShevitzJ, et al. Evaluation of Cancer Res 2002;62:4996 ^ 5000. gastric epithelial cells and stimulates interleukin-8 pro- a complementarity-determining region-grafted (hu- 15. Claesson L, Larhammer D, Rask L, Peterson PA. duction. Infect Immun 2005;73:2736 ^ 43. manized) anti-carcinoembryonic antigen monoclonal cDNA clone for the human invariant gamma chain of 23. Beswick EJ, Pinchuk IV, Minch K, et al. The Helico- antibody in preclinical and clinical studies. Cancer class II histocompatibility antigens and its implications bacter pylori urease B subunit binds to CD74 on gas- Res 1995;55:5935^ 45s. for the protein structure. Proc Natl Acad Sci U S A tric epithelial cells and induces NF-nB activation and 9. Stein R, Chen S, Grossman W, Goldenberg DM. Hu- 19 8 3;8 0 :73 9 5 ^ 9. interleukin-8 production. Infect Immun 2006;74: man lung carcinoma monoclonal antibody specific for 16. Roche PA, Cresswell P. Intracellular transport of 114 8 ^ 5 5 . the Thomsen-Friedenreich antigen. Cancer Res 1989; class II MHC molecules directed by invarient chain. 24. Chaouchi N,Vazquez A, Galanaud P, Leprince C. B 49:32^7. Nature (London) 1990;345:615 ^ 8. cell antigen receptor-mediated apoptosis. Importance

Clin Cancer Res 2009;15(8) April 15, 2009 2816 www.aacrjournals.org Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2009 American Association for Cancer Research. Milatuzumab Improves Drug Therapy of Myeloma

ofaccessory molecules CD19and CD22, andof surface 28. Armand J-P, Burnett AK, Drach J, Harousseau J-L, inhibitors induce death but activate NF-kB on endo- IgM cross-linking. J Immunol 1995;154:3096 ^ 104. Lowenberg BL, San Miguel J. The emerging role of metrial carcinoma cell lines and primary culture 25. Dhein J, Daniel P, Trauth BC, Oehm A, Moller P, for hematologic malignancies: up- explants. J Biol Chem 2006;281:22118^ 30. Krammer PH. Induction of apoptosis by monoclonal date on bortezomib and . Oncologist 2007; 33. Chen K-F, Yeh P-Y,Yeh K-h, Lu Y-S, Huang S-Y, antibody anti-APO-1class switch variants is depen- 12 :2 81 ^ 9 0. Cheng A-L. Down-regulation of phospho-Akt is a dent on cross-linking of APO-1cell surface antigens. 29. Rajkumar SV, Richardson PG, HideshimaT, Anderson major molecular determinant of bortezomib-induced J Immunol 1992;149:3166 ^ 73. KC. Proteasome inhibition as a novel therapeutic target apoptosis in hepatocellular carcinoma cells. Cancer 26. Marches R, Racila E, Tucker TF, et al. Tumour dor- in human cancer. JClin Oncol 2005;23:630 ^ 9. Res 2008;68:6698^707. mancy and cell signaling-III: Role of hypercrosslinking 30. Markovina S, Callander NS, O’Connor SL, et al. 34. SandaT, Iida S, Ogura H, et al. Growth inhibition of of IgM and CD40 on the induction of cell cycle arrest Bortezomib-resistant nuclear factor-KB activity in multiple myeloma cells by a novel IkB kinase inhibitor. and apoptosis in B lymphoma cells. Therapeutic multiple myeloma cells. Mol Cancer Res 2008;6: Clin Cancer Res 2005;11:1974 ^ 82. Immunol 1995;2:125 ^ 36. 13 57^ 6 4. 35. Orlowski RZ, NaglerA, Sonneveld P, et al. Random- 27. Newell MK,VanderWall J, Beard KS, Freed JH. Liga- 31. Yang DT, Young KH, Kahl BS, Markovina S, ized Phase III study of pegylated liposomal doxorubi- tion of major histocompatibility complex class II mole- Miyamoto S. Prevalence of bortezomib-resistant cin plus bortezomib compared with bortezomib alone cules mediates apoptotic cell death in resting B constitutive NF-nB activity in . in relapsed or refractory multiple myeloma: combina- lymphocytes. Proc Natl Acad Sci U S A 1993;90: Mol Cancer 2008;7:doi:10.1186/476 ^ 4598 ^ 7-40. tion therapy improves time to progression. J Clin 10459^63. 32. Dolcet X, Llobet D, Encinas M, et al. Proteasome Oncol 2007;25:3892 ^ 901.

www.aacrjournals.org 2817 Clin Cancer Res 2009;15(8) April15, 2009 Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2009 American Association for Cancer Research. Combining Milatuzumab with Bortezomib, Doxorubicin, or Dexamethasone Improves Responses in Multiple Myeloma Cell Lines

Rhona Stein, Mitchell R. Smith, Susan Chen, et al.

Clin Cancer Res 2009;15:2808-2817.

Updated version Access the most recent version of this article at: http://clincancerres.aacrjournals.org/content/15/8/2808

Cited articles This article cites 33 articles, 26 of which you can access for free at: http://clincancerres.aacrjournals.org/content/15/8/2808.full#ref-list-1

Citing articles This article has been cited by 1 HighWire-hosted articles. Access the articles at: http://clincancerres.aacrjournals.org/content/15/8/2808.full#related-urls

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

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://clincancerres.aacrjournals.org/content/15/8/2808. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2009 American Association for Cancer Research.