Monoclonal antibodies (MoAbs) that selectively identify Muc-1 core protein (MoAbs DF3-P, VU-4H5) determinants were used to identify the Muc-1 glycoform present on 7 multiple myeloma (MM) cell lines, 5 MM patient plasma cells, 12 MM patient B cells, as well as 32 non-MM cell lines and normal hematopoietic cells. Flow cytometry studies demonstrated that all MM cell lines, MM patient plasma cells, and MM patient B cells expressed Muc-1 core protein epitopes. Circulating B cells from 4 normal donors also expressed Muc-1 core protein. In contrast, Muc-1 core protein was absent on 28 of 32 non-MM neoplastic cell lines, 17 of which expressed Muc-1. Splenic and tonsillar B cells, CD34+ stem cells, resting T cells, and bone marrow plasma cells obtained from normal donors both lacked Muc-1 glycoforms. We next studied the effects of estrogen, progesterone, and glucocorticoid receptor agonists and antagonists on Muc-1 expression, because consensus sequences for the response elements of these steroids are present on the Muc-1 gene promoter. These studies showed that dexamethasone (Dex) induced Muc-1 expression on MM cell lines, as determined by both flow cytometry and Western blot analyses. Dex also induced upregulation of Muc-1 on prostate and ovarian cancer cell lines. Time and dose-response studies demonstrated that Dex induced maximal cell surface Muc-1 expression by 24 hours at concentrations of 10−8 mol/L. Dex induced Muc-1 upregulation could be blocked with a 10-fold excess of the glucocorticoid receptor antagonist RU486, confirming that Dex was acting via the glucocorticoid receptor. No changes in Muc-1 expression were observed on MM cells treated with estrogen and progesterone receptor agonists and antagonists or with RU486. These studies provide the framework for targeting Muc-1 core protein in vaccination and serotherapy trials in MM. In addition, the finding that Muc-1 expression on MM cells can be augmented by Dex at pharmacologically achievable levels suggests their potential utility in enhancing treatments targeting Muc-1 in MM.

ALTHOUGH MULTIPLE myeloma (MM) is sensitive to chemotherapy with a median survival 3 to 4 years, most patients are not cured and eventually succumb to MM. However, MM cells express both patient-specific (ie, idiotype) and myeloma-associated antigens that are ideal targets for immunotherapy. One such MM-associated antigen is the core protein of MUC-1 (polymorphic epithelial mucin, epithelial membrane antigen, DF3 antigen).1 Muc-1 is normally present on the luminal surface of secretory glands as a highly glycosylated transmembrane protein. Its extracellular domain is made up of 20 amino acid tandem repeats, with a variable number (range, 21 to 125) of copies within one Muc-1 molecule.2 The variable number of tandem repeat (VNTR) domain of Muc-1 is immunogenic: most of the 56 monoclonal antibodies (MoAbs) submitted to the TD-4 workshop bound to epitopes within the VNTR domain.3,4 The specificity of antibody binding to the VNTR domain depends largely on the extent of glycosylation of Muc-1.5-7 In contrast to normal glandular tissues, adenocarcinomas of the breast, ovary, pancreas, and lung express Muc-1 in an aberrant, hypoglycosylated form.1 Hypoglycosylated variants of Muc-1 appear to arise from mutations in glycosylation enzymes, resulting in the addition of shorter and less branched carbohydrate chains to the Muc-1 core protein backbone.8These differences in the glycosylation patterns of Muc-1 have permitted the identification of MoAbs that distinguish its various glycoforms.3-7 

Muc-1 is defined as an epithelial antigen. It is also expressed on MM cell lines, fresh patient MM cells, and plasmacytomas,9-13as well as other hematological malignancies, including Ki-1–positive B-cell lymphomas, T-cell lymphomas, Hodgkin’s disease, and malignant histiocytosis.9,10,14-16 The function of Muc-1 is not fully understood. Potential roles as an immunostimulatory12,17,18 or as an immunoinhibitory19,20 molecule have been proposed. Furthermore, Muc-1 binds to intercellular adhesion molecule-1 (ICAM-1), suggesting a role in cellular migration.21 Moreover, previous studies have identified consensus sequences for steroid response elements for estrogen, progesterone, and glucocorticoids on the Muc-1 promoter.22,23 Although a regulatory role for ovarian steroids on MUC-1 expression in mouse24and baboon25 uterus has been reported, no studies have yet examined the effect of steroids on MUC-1 expression on human cells.

In the present studies, we characterized the glycoform expression of Muc-1 on freshly obtained MM patient plasma cells and MM cell lines, as well as on normal donor plasma cells. Moreover, we examined Muc-1 glycoform expression on MM cells, as well as on hematologic and carcinoma cell lines. Because the MM clone extends from pre-B cells,26 sIgM+ preswitched B cells,27,28 to late stage B cells,29,30 we also studied Muc-1 glycoform expression on B cells from MM patients as well as normal donors. Given the steroid response elements on the Muc-1 promoter, we also examined the effect of treatment with estrogen, progesterone, and glucocorticoid receptor agonists and antagonists on Muc-1 expression on the cell surface of human MM cells. These studies demonstrate that MM plasma cells and B cells express Muc-1 core protein determinants and that cell surface expression of Muc-1 can be upregulated by dexamethasone. Our data therefore suggest the potential utility of incorporating glucocorticoids into immunotherapeutic treatment strategies targeting Muc-1 in MM.

Cell lines and culture conditions.

ARH-77, CESS, CMK, K562, MCF.7, RPMI 8226, U266, and ZR-75 cell lines were obtained from the American Type Culture Collection (Rockville, MD). OCI My-5 cells31 were kindly provided by Dr H.A. Messner (Ontario Cancer Institute, Toronto, Ontario, Canada); interleukin-6 (IL-6)–transfected S6B45 MM cells32 were kindly provided by Dr T. Kishimoto (Osaka University, Osaka, Japan). Jurkat, B-15, NALM 6, HZ, DHL6, DHL16, and RL cell lines were provided by Dr Lee Nadler (Dana-Farber Cancer Institute, Boston, MA). L5174T, SW480, HUTU80, HS294T, and AC5 cell lines were obtained from Dr Selwyn Broitman (Boston University Medical School, Boston, MA). AGSB gastric, T.Tu esophageal, and CAPAN-2 pancreatic cancer cells were provided by Drs Robert Hennihan, Anil Rustgi, and Andrew Warshaw, respectively (Massachusetts General Hospital, Boston, MA). 36M, CAOV-3, and OVC3 ovarian cancer cells were obtained from Dr Stephen Cannistra (Dana-Farber Cancer Institute). Lastly, DU145 and PC3 prostate cancer cells were kindly provided by Dr Arthur Pardee (Dana-Farber Cancer Institute). All MM (except as noted below), acute lymphocytic, non-Hodgkin’s lymphoma (NHL), lymphoblastic, leukemic, megakaryocytic, prostatic, and 786-0 renal cancer cell lines were cultured in RPMI 1640 medium (GIBCO, Grand Island, NY). OCI-My5 MM cells were cultured in Iscove’s Modified Dulbecco’s Medium (GIBCO). Breast, colon, duodenal, esophageal, gastric, melanoma, ovarian, A458, and ACHN renal cancer cell lines were cultured in Dulbecco’s Modified Eagle’s Medium. CAPAN-2 pancreatic and Caki-1 renal cancer cells were cultured in McCoy’s 5A medium. All media contained 10% fetal bovine serum (FBS; Hyclone, Logan, UT) and was supplemented with L-glutamine, penicillin, and streptomycin (GIBCO).

Patients.

Peripheral blood (PB) and/or bone marrow (BM) was obtained from 17 patients with MM after informed consent was obtained. MM patients included those at diagnosis, during intermittent chemotherapy, in stable phase, and in relapse. Peripheral blood was obtained from 6 healthy normal volunteer donors, and BM was obtained from 4 donors with nonmalignant disease, termed normal for the purpose of this study.

Antibodies.

DF3 and DF3-P MoAbs were the kind gift of Dr Donald Kufe (Dana-Farber Cancer Institute). The antigenic determinants for these antibodies have been previously characterized.7 VU-3C6 and VU-4H5 MoAbs were purified at our institution from hybridomas kindly provided by Dr J. Hilgers (Vrije Universiteit, Amsterdam, The Netherlands); the epitopes targeted by these antibodies have been extensively characterized.3,4 DF3 and VU-3C6 MoAbs bind to both Muc-1 glycosylated and core protein epitopes, whereas DF3-P and VU-4H5 MoAbs specifically bind to core protein epitopes within the VNTR domain of Muc-1.3,4 B4-FITC (CD19) MoAb was obtained from Coulter (Hialeah, FL). FMC63 (CD19) MoAb33 was conjugated to fluorescein isothiocyanate (FITC). Anti-CD38 (Leu17-phycoerythrin [PE]) MoAb was purchased from Becton Dickinson (San Jose, CA). FMC44 (CD45RA) MoAb34 was conjugated to FITC or PE and antihuman Ig (goat antihuman Ig [H+L]-FITC), and goat antimouse Ig-PE were purchased from Southern Biotechnology (Birmingham, AL).

Phenotypic analyses of cell lines and immunofluorescence.

MM and other neoplastic cell lines were characterized by flow cytometry for cell surface Muc-1 expression. Cells (106) were incubated with either anti–Muc-1 or control MoAbs (5.0 μg) for 45 minutes. Cells were then washed with phosphate-buffered saline (PBS) and incubated for 45 minutes with 2.0 μg of goat antimouse fluorescein-conjugated MoAb (Coulter). Cells were next washed and fixed with 1% formaldehyde PBS. Staining intensities were defined by comparing differences in log fluorescence for anti–Muc-1 MoAbs versus isotype control MoAbs, using the following criteria: (0) no difference; (+) <1 log fold difference ; (++) ≥1 but < 2 log difference; and (+++) ≥2 log difference.

Phenotypic analysis of patient cells and immunofluorescence.

PB or BM aspirates were collected into heparinized vacutainer tubes and centrifuged over a ficoll-hypaque gradient (Pharmacia, Uppsala, Sweden). To identify plasma cells from patients or normal donors, BM mononuclear cells (MC) were stained in three-color immunofluorescence using CD38-FITC and CD45RA-PE together with Muc-1 MoAbs or isotype-matched control MoAbs (indirect immunofluorescence). Files were gated for CD38hi45RA cells and the staining for Muc-1 epitopes plotted as a histogram as compared with an identically gated sample stained with an isotype-matched control MoAb. For PBMC samples, cells were stained in two-color immunofluorescence using CD19-FITC and Muc-1 or isotype-matched control MoAb (indirect immunofluorescence). The number of CD19+ B cells in MM patients and normal donors is identical when detected using either FMC63 or with B4 MoAb. Files were gated for CD19 cells and side scatter, and the staining for Muc-1 epitopes was plotted as a histogram as compared with identically gated isotype-matched control samples. For MM PBMC, CD19+ B cells as defined here have been previously shown to express CD19 and IgH transcripts with an IgH VDJ sequence identical to that of autologous BM plasma cells.33,35,36Normal T cells were isolated from PBMC using the E-Rosette technique.37 The purity of isolated T cells was confirmed by flow cytometric analysis for CD3 expression. CD34+ cells were isolated from the PB of healthy donors using Ceprate CD34 columns (Cell Pro, Bothell, WA).38 

Preparation of steroid agonists and antagonists.

17β estradiol (E2), tamoxifen (TAM), progesterone (PRG), RU486, and water-soluble dexamethasone (DEX) were obtained from Sigma Chemical (St Louis, MO). ICI 182,780 (ICI) was the kind gift from Zeneca Pharmaceuticals (Wilmington, DE). Stock solutions (10−2 mol/L in absolute alcohol) of E2, TAM, PRG, and RU486 were stored at −20°C. Water-soluble Dex was solubilized in PBS at a 10−2 mol/L stock solution and stored at 4°C. Cells were cultured for 24 to 72 hours with varying concentrations of steroids, which were freshly prepared for each experiment.

Immunoblotting assays for MUC-1.

Cell extracts from MM cell lines (RPMI 8226 and S6B45), breast cancer cell lines (MCF.7 and ZR-75), and prostatic cancer cells (DU145) were examined by Western blot analysis. Cells were lysed by incubation for 30 minutes in ice-cold lysis buffer (50 mmol/L Tris-HCl [pH 8.0], 150 mmol/L NaCl, 0.1% Nonidet P-40, 1 mmol/L EDTA, 5 μg/mL phenylmethyl sulfonyl fluoride [PMSF], 1 mmol/L Na3VO4, 2 μg/mL aprotinin, 2 μg/mL leupeptin, and 50 mmol/L NaF). Aliquots of lysates were electrophoresed on a 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). Protein was then transferred onto nitrocellulose membranes, and nonspecific binding was blocked by overnight incubation with 5% skim milk. Subsequently, the membranes were washed with TBS-Tween and incubated with either VU-3C6, VU-4H5, DF3, or DF3-P MoAbs (5 μg; diluted 1:200). DMA1 anti–α-tubulin MoAb (1 μg; diluted 1:1,000) was used as a control antibody. An enhanced chemiluminescence (ECL) kit (Amersham, Arlington Heights, IL) was used to visualize bound antibodies.

Expression of Muc-1 core protein on MM and other neoplastic cell lines.

In these studies, Muc-1 expression on 7 human MM cell lines, along with 32 other neoplastic cell lines, was defined by staining with MoAbs that bind Muc-1 glycosylated and core protein epitopes (VU-3C6, DF3) as well as MoAbs binding specifically to Muc-1 core protein (VU-4H5, DF3-P) in flow cytometric analysis. These studies demonstrated that 7 of 7 MM lines (ARH-77, HS Sultan, IM9, OCI My-5, RPMI 8226, S6B45, and U266) expressed Muc-1 on their cell surface, as evidenced by reactivity with the VU-3C6 MoAb. The intensities of Muc-1 expression using the VU-3C6 MoAb were as follows: low (U266), medium (ARH-77, HS Sultan, IM9), and high (OCI My-5, RPMI 8226, S6B45) (Fig 1and Table 1). These results paralleled those obtained with the DF3 MoAb, which identified Muc-1 on 6 of 7 MM lines (Table 1). In particular, DF3 staining was absent on HS Sultan cells, but was present on the remainder of MM cell lines with the following staining patterns: low (ARH-77, IM9) and high (OCI My-5, RPMI 8226, S6B45, U266) (Table 1). Muc-1 was also identified by either VU-3C6 or DF3 MoAbs on 17 of 32 non-MM neoplastic cell lines: Jurkat ALL; HZ NHL; MCF.7 and ZR-75 breast; CESS B-cell lymphoblasts; SW480 colon; T.Tu esophageal; CMK megakaryocytic; 36M, CAOV-3 and OVC3 ovarian; Capan-2 pancreatic; DU145 and PC3 prostatic; as well as A498, ACHN, and Caki-1 renal cancer cells. The intensities of Muc-1 staining among non-MM, Muc-1–positive cells was strongest on the breast, esophageal, ovarian, pancreatic, prostatic, and renal cancer cell lines (Table 1).

Fig. 1.

Expression of Muc-1 on human MM and breast cancer cell lines. Human MM (ARH-77, OCI My-5, RPMI 8226, S6B45 and U266) and breast cancer (ZR-75) cell lines were examined by flow cytometry for cell surface expression of Muc-1 glycosylated protein by staining with the VU-3C6 MoAb relative to an isotype control MoAb.

Fig. 1.

Expression of Muc-1 on human MM and breast cancer cell lines. Human MM (ARH-77, OCI My-5, RPMI 8226, S6B45 and U266) and breast cancer (ZR-75) cell lines were examined by flow cytometry for cell surface expression of Muc-1 glycosylated protein by staining with the VU-3C6 MoAb relative to an isotype control MoAb.

Close modal
Table 1.

Expression of Muc-1 on Cancer Cell Lines

Cell Line Type VU-3C6 DF3 VU-4H5 DF3P
ALL207 ALL, B-cell  0  0  0  0  
Jurkat  ALL, T-cell +  0  0  0/+  
B15  ALL, B-cell  0  0  0  
NALM 6  ALL, B-cell  0  0  0  0  
HL60 AML, Promyeloc.  0  0  0  0  
K562  AML, Erythroleuk.  0  0  0  0  
HZ  B-cell, NHL  +  +  0  
DHL6  B-cell, NHL  0  0  0  0  
DHL16 B-cell, NHL  0  0  0  +  
RL  B-cell, NHL  0  0  0  
BCBL-1  B-cell, PEL  0  ND  0  ND 
MCF.7  Breast  ++  ++  0  0  
ZR-75  Breast ++  ++  0  0  
CESS  B-lymphoblast  +  0  0  
L5174T  Colon  0  0  0  0  
SW480  Colon 0  +  0  0  
HUTU80  Duodenal  0  0  0  
T.Tu  Esophageal  0  ++  0  0  
AGSB  Gastric 0  0  0  0  
CMK  Megakaryocytic  +  0  0  
HS294T  Melanoma  0  0  0  0  
AC5 Melanoma  0  0  0  0  
ARH-77  Myeloma  ++ +  +  0  
HS Sultan  Myeloma  ++  0  +  
IM9  Myeloma  ++  +  ++  0  
OCI My-5  Myeloma +++  +++  +  +  
S6B45  Myeloma  +++  +++ ++  +  
RPMI 8226  Myeloma  +++  +++  ++  ++ 
U266  Myeloma  +  +++  +  +  
36M  Ovarian 0  ++  0  0  
CAOV-3  Ovarian  0  +  0  
OVC3  Ovarian  0  +  0  0  
Capan-2  Pancreas +++  +++  +  ND  
DU145  Prostate  ++  +++ 0  0  
PC3  Prostate  +  0  0  0  
A498 Renal  0  +  0  ND  
ACHN  Renal  +  +  ND  
Caki-1  Renal  0  ++  0  ND  
786-0 Renal  0  0  0  ND 
Cell Line Type VU-3C6 DF3 VU-4H5 DF3P
ALL207 ALL, B-cell  0  0  0  0  
Jurkat  ALL, T-cell +  0  0  0/+  
B15  ALL, B-cell  0  0  0  
NALM 6  ALL, B-cell  0  0  0  0  
HL60 AML, Promyeloc.  0  0  0  0  
K562  AML, Erythroleuk.  0  0  0  0  
HZ  B-cell, NHL  +  +  0  
DHL6  B-cell, NHL  0  0  0  0  
DHL16 B-cell, NHL  0  0  0  +  
RL  B-cell, NHL  0  0  0  
BCBL-1  B-cell, PEL  0  ND  0  ND 
MCF.7  Breast  ++  ++  0  0  
ZR-75  Breast ++  ++  0  0  
CESS  B-lymphoblast  +  0  0  
L5174T  Colon  0  0  0  0  
SW480  Colon 0  +  0  0  
HUTU80  Duodenal  0  0  0  
T.Tu  Esophageal  0  ++  0  0  
AGSB  Gastric 0  0  0  0  
CMK  Megakaryocytic  +  0  0  
HS294T  Melanoma  0  0  0  0  
AC5 Melanoma  0  0  0  0  
ARH-77  Myeloma  ++ +  +  0  
HS Sultan  Myeloma  ++  0  +  
IM9  Myeloma  ++  +  ++  0  
OCI My-5  Myeloma +++  +++  +  +  
S6B45  Myeloma  +++  +++ ++  +  
RPMI 8226  Myeloma  +++  +++  ++  ++ 
U266  Myeloma  +  +++  +  +  
36M  Ovarian 0  ++  0  0  
CAOV-3  Ovarian  0  +  0  
OVC3  Ovarian  0  +  0  0  
Capan-2  Pancreas +++  +++  +  ND  
DU145  Prostate  ++  +++ 0  0  
PC3  Prostate  +  0  0  0  
A498 Renal  0  +  0  ND  
ACHN  Renal  +  +  ND  
Caki-1  Renal  0  ++  0  ND  
786-0 Renal  0  0  0  ND 

Cell surface staining of neoplastic cell lines was assessed by flow cytometry using the VU-3C6 and DF3 antibodies that identify both Muc-1 glycosylated and core protein determinants and the VU-4H5 and DF3-P antibodies that specifically identify Muc-1 core protein. Intensity of staining is denoted as follows: (0) absent; (+) low; (++) medium; (+++) high.

Abbreviation: ND, not determined.

Interestingly, expression of Muc-1 core protein, determined by reactivity with the VU-4H5 and DF3 MoAbs, was more selective for MM cell lines (Fig 2 and Table 1). Using the VU-4H5 MoAb, Muc-1 core protein was detected on the cell surface of 7 of 7 MM cell lines with the following staining intensities: low (ARH-77, HS Sultan, OCI My-5, U266) and medium (IM9, RPMI 8226, S6B45) (Table 1). In addition, Muc-1 core protein was identified on 4 of 7 MM cell lines using the DF3-P MoAb with the following patterns of reactivity: low (U266, OCI My-5, S6B45) and medium (RPMI 8226) (Table1). In contrast to MM cells, 28 of 32 (88%) of non-MM neoplastic cell lines, including MCF.7 and 2R-75 breast as well as OVC.3 and 36M ovarian cell lines, lacked Muc-1 core protein, as determined by reactivity with the VU-4H5 or DF3-P MoAbs. Three of 4 non-MM cell lines that expressed Muc-1 core protein were of B-cell lineage, akin to the origin of MM cells, whereas the fourth cell line was CAPAN-2, which is a pancreatic cancer cell line. Expression of Muc-1 core protein in these 4 non-MM cell lines was of low intensity. As can be seen in Table1, expression of Muc-1 core protein was absent on breast, esophageal, ovarian, prostatic, and renal cancer cell lines, all of which expressed Muc-1 by use of the VU-3C6 and DF3 MoAbs.

Fig. 2.

Expression of Muc-1 core protein on human MM, breast cancer, and ovarian cancer cell lines. Human MM (ARH-77, OCI My-5, RPMI 8226, and S6B45), breast cancer (MCF.7 and ZR-75), and ovarian cancer (OVC.3 and 36M) cell lines were examined by flow cytometry for cell surface expression of Muc-1 core protein by staining with the VU-4H5 MoAb relative to an isotype control MoAb.

Fig. 2.

Expression of Muc-1 core protein on human MM, breast cancer, and ovarian cancer cell lines. Human MM (ARH-77, OCI My-5, RPMI 8226, and S6B45), breast cancer (MCF.7 and ZR-75), and ovarian cancer (OVC.3 and 36M) cell lines were examined by flow cytometry for cell surface expression of Muc-1 core protein by staining with the VU-4H5 MoAb relative to an isotype control MoAb.

Close modal
Expression of Muc-1 core protein on plasma cells from MM patients and normal donors.

To assess Muc-1 expression on freshly isolated plasma cells from MM patients, BMMC from 5 MM patients were stained in multicolor immunofluorescence using CD38, CD45RA, and MoAbs that detect both Muc-1 glycosylated and core protein epitopes (VU-3C6) or specifically identify core protein (VU-4H5). Of CD38hi45RA plasma cells from 5 of 5 MM patients, 55% ± 12% (range, 13% to 78%) express Muc-1 core protein (Fig 3 and Table 2). By use of the VU-3C6 MoAb, Muc-1 epitopes were identified on 38% ± 13% (range, 8% to 84%) of MM plasma cells from these 5 MM patients. In contrast, for 4 of 4 donors, normal plasma cells had very low or no expression of any Muc-1 glycoform using either MoAb (Fig 3 and Table 2).

Fig. 3.

Expression of Muc-1 core protein on MM patient and normal donor plasma cells. BM plasma cells from 2 representative MM patients (A) and 2 representative normal donors (B) are shown. Files were gated for CD38hi45RA plasma cells (15% to 87% and 1% to 5% were present in MM patients and normal donors, respectively). Muc-1 on plasma cells was plotted as a histogram, with bold lines indicating staining with the VU-4H5 or VU-3C6 anti–Muc-1 MoAbs and the thin lines representing staining by an identically gated isotype-matched control MoAb.

Fig. 3.

Expression of Muc-1 core protein on MM patient and normal donor plasma cells. BM plasma cells from 2 representative MM patients (A) and 2 representative normal donors (B) are shown. Files were gated for CD38hi45RA plasma cells (15% to 87% and 1% to 5% were present in MM patients and normal donors, respectively). Muc-1 on plasma cells was plotted as a histogram, with bold lines indicating staining with the VU-4H5 or VU-3C6 anti–Muc-1 MoAbs and the thin lines representing staining by an identically gated isotype-matched control MoAb.

Close modal
Table 2.

Muc-1 Epitopes on MM and Normal B-Lineage Cells in Blood and Bone Marrow

% of Subset Expressing Muc-1 Epitopes
VU-4H5 VU-3C6
Myeloma B-lineage subsets  
 MM plasma cells (BM) (5)  55 ± 12* 38 ± 13* 
 Range  15-78   8-84  
 MM B cells (PBMC) (12)  85 ± 3  57 ± 9 
 Range  58-95  15-98  
Normal B-lineage subsets  
 Plasma cells (BM) (4)  6 ± 2 1 ± 0.4  
 Range   1-12  0-2 
 B cells (PBMC) (4)  49 ± 10  64 ± 12 
 Range  25-74  33-90 
% of Subset Expressing Muc-1 Epitopes
VU-4H5 VU-3C6
Myeloma B-lineage subsets  
 MM plasma cells (BM) (5)  55 ± 12* 38 ± 13* 
 Range  15-78   8-84  
 MM B cells (PBMC) (12)  85 ± 3  57 ± 9 
 Range  58-95  15-98  
Normal B-lineage subsets  
 Plasma cells (BM) (4)  6 ± 2 1 ± 0.4  
 Range   1-12  0-2 
 B cells (PBMC) (4)  49 ± 10  64 ± 12 
 Range  25-74  33-90 

B and plasma cells were defined as indicated in Figs 3 and 4. Values are the mean ± SE.

*

P = .01 as compared with normal plasma cells.

Expression of Muc-1 core protein on B cells from MM patients and normal donors.

Previous work has shown that circulating B cells in MM patients are part of the malignant clone as defined by their expression of clonotypic IgH transcripts.33,35,36 PB B cells from 12 of 12 MM patients were analyzed for expression of Muc-1 epitopes (Fig 4A and Table 2). B cells were defined as CD19+ cells, including those cells with a high side scatter (SSchi).27,36 The SSchi set of MM B cells has been shown to be almost exclusively clonotypic, whereas the SSclow set includes both clonotypic and polyclonal B cells.33,35 The majority of CD19+B cells in MM patients (85% ± 3%) expressed Muc-1 core protein (Fig 4A and Table 2). Both the SSchi and SSclowsets of MM B cells were predominantly Muc-1+ (not shown). Polyclonal B cells (49% ± 10%) from 4 of 4 normal donors, defined as CD19+SSclow PBMC, also expressed Muc-1 core protein (Fig 4B and Table 2). In contrast to normal PB B cells, splenic and tonsillar B cells from 3 of 3 normal donors lacked Muc-1, by virtue of lack of reactivity with the VU-4H5 and VU-3C6 MoAbs (data not shown).

Fig. 4.

Expression of Muc-1 core protein on MM patient and normal donor B cells. Peripheral blood B cells from 2 representative MM patients (A) and 2 representative normal donors (B) are shown, with Muc-1 staining plotted as a histogram. The bold line shows Muc-1 staining on B cells, and the thin line shows isotype-matched control staining on an identically gated aliquot of cells. Shown are B cells (26% to 32% and 4% to 7% for MM patients and normal donors, respectively), which include SSchi gated PB B cells for MM patients, because these are clonotypic, having IgH and CD19 transcripts.32,34,35 In contrast, SSchi PB B cells are excluded from the B-cell gate in normal donors, because these are monocytes.35 

Fig. 4.

Expression of Muc-1 core protein on MM patient and normal donor B cells. Peripheral blood B cells from 2 representative MM patients (A) and 2 representative normal donors (B) are shown, with Muc-1 staining plotted as a histogram. The bold line shows Muc-1 staining on B cells, and the thin line shows isotype-matched control staining on an identically gated aliquot of cells. Shown are B cells (26% to 32% and 4% to 7% for MM patients and normal donors, respectively), which include SSchi gated PB B cells for MM patients, because these are clonotypic, having IgH and CD19 transcripts.32,34,35 In contrast, SSchi PB B cells are excluded from the B-cell gate in normal donors, because these are monocytes.35 

Close modal
Expression of Muc-1 on normal CD34+ hematopoietic progenitor cells and T cells.

We next examined whether Muc-1 was expressed on CD34+hematopoietic progenitor cells and on resting T cells obtained from 3 normal donors using the VU-4H5 and VU-3C6 MoAbs. Neither PB-derived CD34+ hematopoietic progenitor cells nor resting T cells from normal donors express Muc-1 (data not shown).

Effects of estrogen, progesterone, and glucocorticoid receptor agonists and antagonists action on Muc-1 expression.

Although consensus sequences of steroid response elements for estrogen, progesterone, and glucocorticoid receptors have been identified on the promoter of the Muc-1 gene,21,26 no functional analyses of these steroid response elements has been reported in human cells. We therefore treated S6B45, RPMI 8226, and OCI My-5 MM cells; MCF.7 and ZR-75 breast cancer cells; CAOV-3 and OVC-3 ovarian cancer cells; and DU145 prostate cancer cells with agonists and antagonists for estrogen (17β estradiol, diethylstilbestrol, tamoxifen, and ICI 182,780), progesterone (progesterone, RU486), and/or glucocorticoid (Dex, RU486) receptors and assayed for related changes in Muc-1 cell surface expression. Dex induced upregulation of Muc-1, as evidenced by reactivity with the VU-4H5, DF3-P, VU-3C6, and DF3 MoAbs, using flow cytometry and/or Western blot analyses of S6B45 and RPMI 8226 (Figs 5 and 6), of OCI My-5 MM cells, of CAOV-3 and OVC-3 ovarian cancer cells, and of DU145 prostate cancer cells (Figs 5 and 6). Dex induction of Muc-1 expression was most pronounced (≥1 log fold increase in fluorescence intensity) on S6B45 and RPMI 8226 MM cells and on DU145 prostatic cancer cells (Fig 5). Dose-response studies using Dex (10−12 to 10−6 mol/L) showed maximal induction of cell surface Muc-1 expression on S6B45 MM (Fig 7) and DU145 prostatic cancer cells at 10−8 mol/L. Dex induction of Muc-1 expression on S6B45, RPMI 8226, and OCI My-5 MM cells, as well as on DU145 prostate cancer cells was also evaluated at several intervals of Dex treatment (24, 48, and 72 hours) and shown to be maximal at 24 hours (data not shown). As can be seen in Fig 8, Dex induction of Muc-1 expression on two MM cell lines (S6B45 and RPMI 8226) was blocked by 10-fold excess of the glucocorticoid receptor antagonist RU486, consistent with a glucocorticoid receptor-mediated mechanism of Muc-1 induction by Dex.

Fig. 5.

Effect of Dex on cell surface Muc-1 expression. RPMI 8226 MM cells, S6B45 MM cells, and DU145 prostate cancer cells were cultured in media alone or with Dex (10−8 mol/L) for 24 hours. Muc-1 expression was assessed by flow cytometry by staining with the VU-4H5 and VU-3C6 MoAbs relative to isotype control MoAbs. Peaks shown are denoted as follows: (*) staining with isotype control MoAb; (**) staining of cells cultured in media alone using either the VU-4H5 or the VU-3C6 MoAbs; (***) staining of cells cultured with Dex using either the VU-4H5 or the VU-3C6 MoAbs. No change in isotype control MoAb staining was seen with Dex stimulation.

Fig. 5.

Effect of Dex on cell surface Muc-1 expression. RPMI 8226 MM cells, S6B45 MM cells, and DU145 prostate cancer cells were cultured in media alone or with Dex (10−8 mol/L) for 24 hours. Muc-1 expression was assessed by flow cytometry by staining with the VU-4H5 and VU-3C6 MoAbs relative to isotype control MoAbs. Peaks shown are denoted as follows: (*) staining with isotype control MoAb; (**) staining of cells cultured in media alone using either the VU-4H5 or the VU-3C6 MoAbs; (***) staining of cells cultured with Dex using either the VU-4H5 or the VU-3C6 MoAbs. No change in isotype control MoAb staining was seen with Dex stimulation.

Close modal
Fig. 6.

Immunoblotting assays for Muc-1 expression in human MM and prostate cancer cell lines stimulated with Dex. Cell lysates obtained from human MM (S6B45 and RPMI 8226) and prostate cancer (DU145) cells cultured with media alone or with Dex (10−8mol/L) for 24 hours were examined by Western blot analysis using the VU-4H5 MoAb. Dual bands seen in the lysates from S6B45 and DU145 cells show coallelic expression of Muc-1.1 The DMA1 MoAb to -tubulin was used as a control antibody.

Fig. 6.

Immunoblotting assays for Muc-1 expression in human MM and prostate cancer cell lines stimulated with Dex. Cell lysates obtained from human MM (S6B45 and RPMI 8226) and prostate cancer (DU145) cells cultured with media alone or with Dex (10−8mol/L) for 24 hours were examined by Western blot analysis using the VU-4H5 MoAb. Dual bands seen in the lysates from S6B45 and DU145 cells show coallelic expression of Muc-1.1 The DMA1 MoAb to -tubulin was used as a control antibody.

Close modal
Fig. 7.

Dose-response relationship for Dex induction of Muc-1 expression on S6B45 MM cells. S6B45 MM cells were cultured for 24 hours with Dex (10−12 to 10−6 mol/L) and analyzed by flow cytometry for Muc-1 expression using VU-4H5, VU-3C6 and isotype control MoAbs. *Cells stained with isotype control. **Cells cultured in media alone and stained with either the VU-4H5 or the VU-3C6 MoAbs. ***Cells cultured with Dex and stained with either the VU-4H5 or the VU-3C6 MoAbs.

Fig. 7.

Dose-response relationship for Dex induction of Muc-1 expression on S6B45 MM cells. S6B45 MM cells were cultured for 24 hours with Dex (10−12 to 10−6 mol/L) and analyzed by flow cytometry for Muc-1 expression using VU-4H5, VU-3C6 and isotype control MoAbs. *Cells stained with isotype control. **Cells cultured in media alone and stained with either the VU-4H5 or the VU-3C6 MoAbs. ***Cells cultured with Dex and stained with either the VU-4H5 or the VU-3C6 MoAbs.

Close modal
Fig. 8.

Effect of RU486 on Dex induction of Muc-1 expression on MM cells. S6B45 and RPMI 8226 MM cells were cultured in media alone or with Dex (10−8 mol/L) in the presence or absence of the glucocorticoid receptor antagonist RU486. (A) Changes in Muc-1 expression after culturing with Dex. (B) Changes in Muc-1 expression after culturing with both Dex and RU486. (C) Changes in isotype control MoAb staining in media alone, Dex alone, and Dex plus RU486 cultures. (a) Cells stained with isotype control MoAb. (b) Cells cultured with media alone and stained with the VU-4H5 MoAb. (c) Cells cultured with Dex and stained with the VU-4H5 MoAb. (d) Cells cultured with both Dex and RU486 and stained with the VU-4H5 MoAb.

Fig. 8.

Effect of RU486 on Dex induction of Muc-1 expression on MM cells. S6B45 and RPMI 8226 MM cells were cultured in media alone or with Dex (10−8 mol/L) in the presence or absence of the glucocorticoid receptor antagonist RU486. (A) Changes in Muc-1 expression after culturing with Dex. (B) Changes in Muc-1 expression after culturing with both Dex and RU486. (C) Changes in isotype control MoAb staining in media alone, Dex alone, and Dex plus RU486 cultures. (a) Cells stained with isotype control MoAb. (b) Cells cultured with media alone and stained with the VU-4H5 MoAb. (c) Cells cultured with Dex and stained with the VU-4H5 MoAb. (d) Cells cultured with both Dex and RU486 and stained with the VU-4H5 MoAb.

Close modal

No changes in Muc-1 expression were observed on MCF.7 and ZR-75 breast cancer cell lines treated with Dex, as assessed by both FACS and Western blot analyses (data not shown). In addition, treatment of S6B45, RPMI 8226, and OCI My-5 MM cells; MCF.7 and 2R75 breast cancer cells; and DU145 prostate cancer cells with estrogen and progesterone receptor agonists and antagonists, or alone with the glucocorticoid receptor antagonist RU486, did not alter Muc-1 cell surface expression (data not shown).

The use of immunotherapy for the treatment of MM has previously targeted cell surface antigens on MM cells such as CD16, CD38, CD54, and HM1.24.39-45 Unfortunately, these antigens are present (CD16, CD38, CD54) or have not been fully examined (HM1.24) on normal tissues. In these studies, we have identified Muc-1 core protein as a selective target for MM-directed immunotherapy. Muc-1 is a suitable target for immunotherapy, because epitopes that give rise to antibody and cytotoxic T-lymphocyte reactivity may be repeated 21 to 125 times within one molecule of Muc-1 due to genetic polymorphisms.1Both antibody46,47 and cytotoxic T-cell responses13,17,18 specific to Muc-1 have been observed in patients with Muc-1–bearing malignancies, and these responses can be elicited by vaccination with Muc-1 conjugated to mannan or keyhole limpet hemocyanin (KLH).48,49 The latter studies are of a phase I nature and show that Muc-1 vaccinations appear to be well tolerated.49 Although no clinical response data have so far been generated from those cancer patients immunized with Muc-1 vaccines, considerable preclinical data using tumor rejection models have shown that Muc-1–directed vaccinations are biologically active. Specifically, vaccinations against Muc-1 using Muc-1 cDNA,50 recombinant vaccinia virus containing Muc-1 gene sequences,51 Muc-1-KLH conjugated protein,52 or fusion cells made up of dendritic cells and Muc-1–bearing tumors53 can prevent establishment and dissemination of inoculated Muc-1–bearing tumors, as well as induce rejection of established Muc-1–positive tumors in mice. Muc-1 may also serve as a target for serotherapy.54,55 Athymic mice xenografted with Muc-1–bearing ovarian tumors show preferential tumor uptake of I125–conjugated DF3 MoAb.54 Moreover, the anti–Muc-1 MoAb HMFG1 conjugated to yttrium-90 has been used in phase I/II clinical trials involving ovarian cancer patients, with benefit suggested when used in an adjuvant setting in patients with no evident disease after surgery.55 

The presence of Muc-1 on the cell surface of MM cells has previously been identified by several investigators.9-13 However, these studies did not identify the Muc-1 glycoform present on MM cells. Because glycosylation patterns of Muc-1 vary among normal and malignant tissues, knowledge of the Muc-1 glycoform present on MM cells may be important in identifying selective epitopes to serve as targets for immunotherapy. In these studies, we have demonstrated that Muc-1 core protein is more selectively expressed on MM cell lines, on patient MM plasma cells and B cells, and on certain (ie, circulating) normal donor B cells. In contrast, Muc-1 core protein is absent on 28 of 32 (88%) non-MM neoplastic cell lines, including breast, esophageal, prostate, ovarian, and renal cancer cell lines that express Muc-1 in a glycosylated form. Moreover, a survey of CD34+ cells, resting T cells, splenic and tonsillar B cells, and plasma cells from normal donors did not show cell surface expression of Muc-1 core protein. Lastly, by immunohistochemistry normal human small intestine (VU-4H5), colon (VU-4H5), and breast (DF3-P, VU-4H5) tissues do not show reactivity when stained with the anti–Muc-1 core protein MoAbs used in these studies.7,56 Taken together, the above-described studies demonstrate that MM plasma and B cells, as well as circulating normal B cells, selectively express Muc-1 core protein on their cell surface. The finding that circulating MM B cells, in addition to plasma cells, express Muc-1 core protein is of particular relevance for Muc-1–directed immunotherapy strategies for MM, because MM B cells may be both clonotypic and act as a reservoir of chemotherapy resistance.26-30,35 

The significance of Muc-1 expression on MM plasma and B cells, along with circulating normal B cells, remains unclear at this time. A role in the migration of Muc-1–bearing malignant cells has previously been suggested, because Muc-1 binds to ICAM-1.21 Such binding may make it possible for Muc-1–bearing MM plasma cells and B cells to adhere to endothelium and bone marrow stroma expressing ICAM-121,57 or possibly another yet to be defined ligand(s). MM cells are known to make cytokines, including IL-1β and tumor necrosis factor-α (TNF-α),58,59which are potent inducers of ICAM-1 expression60,61 and may stimulate ICAM-1 on endothelial and bone marrow stromal cells. Moreover, Kaposi’s sarcoma-associated herpesvirus (KSHV), which may play a role in the pathogenesis of MM by infecting BM stromal cells,62 may lead to cytokine elaboration and induction of ICAM-1 expression on stromal cells, analogous to that seen in Kaposi’s sarcoma.61,63 In turn, binding of MM cells via Muc-1 to ICAM-1 on endothelial and BM stroma cells may lead to the release of IL-6, an important growth factor for MM cells.64 In addition, a role for Muc-1 in mediating evasion from immune systems has been suggested by several studies, wherein Muc-1 may either act through stearic hindrance to evade contact with immune cells or induce anergy through a yet to be defined mechanism.1,19,20 In such circumstances, malignant plasma cells that express Muc-1 would be able to escape immunosurveillance, unlike normal plasma cells that, in these studies, lacked Muc-1 expression.

Because Muc-1 may have a role in immunoregulation, as well as cell migration and adhesion, we were interested in identifying compounds that modulate Muc-1 expression. In particular, the presence of consensus sequences for several steroid response elements (estrogen, progesterone, and glucocorticoids)22,23 on the Muc-1 promoter suggested that steroids might alter Muc-1 expression. Although no functional analyses for these steroid response elements has been reported in human cells, in vitro studies involving mice and baboons suggest that estrogen and progesterone modulate uterine Muc-1 expression.24,25 In our study, Muc-1 expression was upregulated after Dex treatment of MM cell lines, with maximal stimulation occurred at 10−8 mol/L Dex, doses that correspond to serum levels of Dex that are readily achievable clinically.65 Muc-1 expression was also upregulated on DU145 prostatic cancer cells, as well as on CAOV-3 and OVC-3 ovarian cancer cells, demonstrating that Dex-induced upregulation of Muc-1 is not restricted to MM cells. However, no change in Muc-1 expression was seen on two breast cancer cell lines (MCF.7 and ZR-75) exposed to Dex, showing selectivity of induction of Muc-1 by Dex. In contrast to murine and baboon uterine Muc-1 studies,24,25 no changes in Muc-1 expression were triggered in MM, breast, and prostatic cancer cells treated with estrogen and progesterone receptor agonists and antagonists. These different sequelae may reflect species or tissue specificity for Muc-1 regulation by estrogen or progesterone. In addition, these animal studies involved in vivo treatment for longer periods of time (up to 12 days) with estrogen and progesterone, whereas our studies with cell lines involved in vitro exposure to steroids for up to 72 hours. Hence, we cannot exclude the possibility that longer intervals of exposure to estrogen and progesterone might influence Muc-1 expression on MM cells.

In summary, our studies form the basis for targeting Muc-1 core protein in vaccination and serotherapy trials for MM. The finding that Muc-1 expression can be augmented by Dex at pharmacologically achievable levels suggests the potential utility of incorporating glucocorticoids into immunotherapeutic strategies targeting Muc-1 in MM.

Supported by National Institutes of Health grant CA78378, the Lauri Strauss Leukemia Foundation and the International Myeloma Foundation, a Young Investigator Award from the American Society of Clinical Oncology (S.P.T.), and a grant from the Alberta Cancer Board Research Initiative Program.

The publication costs of this article were defrayed in part by page charge payment. This article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. section 1734 solely to indicate this fact.

1
Patton
S
Gendler
SJ
Spicer
AP
The epithelial mucin, Muc1, of milk, mammary gland and other tissues.
Biochim Biophys Acta
1241
1995
407
2
Gendler
SJ
Lancaster
CA
Taylor-Papademetriou
J
Duhig
T
Peat
N
Burchell
J
Pemberton
L
Lalani
EN
Wilson
D
Molecular cloning and expression of human tumor associated polymorphic epithelial mucin.
J Biol Chem
265
1990
15286
3
Price
MR
Rye
PD
Petrakou
E
Murray
A
Brady
K
et al
Summary report on the ISOBM TD-4 workshop: Analysis of 56 monoclonal antibodies against the Muc1 mucin.
Tumor Biol
19
1998
1
4
Petrakou
E
Murray
A
Price
MR
Epitope mapping of anti-Muc1 mucin protein core monoclonal antibodies.
Tumor Biol
19
1998
21
5
Dai
J
Allard
WJ
Davis
G
Yeung
KK
Effect of desialylation on binding, affinity, and specificity of 56 monoclonal antibodies against Muc1 mucin.
Tumor Biol
19
1998
100
6
Hanisch
FG
Specificity clusters of Muc1-reactive mouse monoclonal antibodies.
Tumor Biol
19
1998
111
7
Perey
L
Hayes
DF
Maimonis
P
Abe
M
O’Hara
C
Kufe
DW
Tumor selective reactivity of a monoclonal antibody prepared against a recombinant peptide derived from the DF3 human breast carcinoma associated antigen.
Cancer Res
52
1992
2563
8
Brockhausen
I
Yang
JM
Burchell
J
Whitehouse
C
Taylor-Papademetriou
J
Mechanisms underlying aberrant glycosylation of Muc1 mucin in breast cancer cells.
Eur J Biochem
233
1997
607
9
Delsol
G
Stein
H
Pulford
KAF
Gatter
KC
Erber
WN
Zinne
K
Mason
DY
Human lymphoid cells express epithelial membrane antigen: Implications for diagnosis of human neoplasms.
Lancet
2
1984
1124
10
Pinkus
GS
Kurtin
PJ
Epithelial membrane antigen—A diagnostic discriminant in surgical pathology.
Hum Pathol
16
1985
929
11
Zotter
S
Hageman
PC
Lossnitzer
A
Mooi
WJ
Hilgers
J
Tissue and tumor distribution of human polymorphic epithelial mucin.
Cancer Rev
11-12
1988
55
12
Duperray
C
Klein
B
Durie
BGM
Zhang
X
Jourdan
M
Poncelet
P
Favier
F
Vincent
C
Brochier
J
Lenoir
G
Bataille
R
Phenotypic analysis of human myeloma cell lines.
Blood
73
1989
566
13
Takahashi
T
Makiguchi
Y
Hinoda
Y
Kakiuchi
H
Nakagawa
N
Imai
K
Yachi
A
Expression of Muc1 on myeloma cells and induction of HLA unrestricted CTL against Muc1 from a multiple myeloma patient.
J Immunol
153
1994
2102
14
Al Saati
T
Caveriviere
P
Gorguet
B
Delsol
G
Gater
KC
Mason
DY
Epithelial membrane antigen in hematopoietic neoplasms.
Hum Pathol
17
1986
533
15
Delsol
G
Al Saati
T
Gatter
KC
Gerdes
J
Schwarting
R
Caveriviere
P
Rigal-Huguet
F
Robert
A
Stein
H
Mason
DY
Coexpression of epithelial membrane antigen (EMA), Ki-1, and interleukin-2 receptor by anaplastic large cell lymphomas.
Am J Pathol
130
1988
59
16
Mark
A
Mangkornkanok
M
B-cell lymphoma marking only with anti-epithelial membrane antigen.
Cancer
63
1989
2152
17
Barnd
DL
Lan
M
Metzgar
RS
Finn
OJ
Specific, MHC unrestricted recognition of tumor associated mucins by human cytotoxic T cells.
Proc Natl Acad Sci USA
86
1989
7159
18
Ioannides
CG
Fisk
B
Jerome
KR
Irimura
T
Wharton
JT
Finn
OJ
Cytotoxic T cells from ovarian malignant tumors can recognize polymorphic epithelial mucin core peptides.
J Immunol
151
1993
3693
19
Van de Wiel-Van Kemenade
E
Ligtenberg
MJL
de Boer
AJ
Buijis
F
Vos
KL
Melief
CJM
Hilkens
J
Figdor
CG
Episialin (Muc1) inhibits cytotoxic lymphocyte-target cell interaction.
J Immunol
151
1993
767
20
Agrawal
B
Krantz
MJ
Reddish
MA
Longenecker
BM
Cancer-associated Muc1 mucin inhibits human T-cell proliferation, which is reversible by IL-2.
Nat Med
4
1998
43
21
Regimbald
LH
Pilarski
LM
Longenecker
BM
Reddish
MA
Zimmermann
G
Hugh
JC
The breast mucin Muc1 as a novel adhesion ligand for endothelial intercellular adhesion molecule 1 in breast cancer.
Cancer Res
56
1996
4244
22
Tsarfaty
I
Hareuveni
M
Horev
J
Zaretsky
J
Weiss
M
Jeltsch
JM
Gernier
JM
Lathe
R
Keydar
I
Wreschner
DH
Isolation and characterization of an expressed hypervariable gene coding for a breast cancer associated antigen.
Gene
93
1990
313
23
Lancaster
CA
Peat
N
Duhig
T
Wislon
D
Taylor-Papademetriou
J
Gendler
SJ
Structure and expression of the human polymorphic epithelial mucin gene: An expressed VNTR unit.
Biochem Biophys Res Commun
173
1990
1019
24
Surveyor
GA
Gendler
SJ
Pemberton
L
Das
SK
Chakraborty
I
Julian
J
Pimental
RA
Wegner
CC
Dey
SK
Carson
DD
Expression and steroid hormonal control of Muc-1 in the mouse uterus.
Endocrinology
136
1995
3639
25
Hild-Petito
S
Fazleabas
AT
Julian
J
Carson
DD
Mucin (Muc-1) expression is differentially regulated in uterine luminal and glandular epithelia of the baboon.
Biol Reprod
54
1996
939
26
Jensen
GS
Mant
MJ
Belch
AR
Berensen
JR
Ruether
BA
Pilarski
LM
Selective expression of CD45 isoforms defines CALLA+ monoclonal B lineage cells in peripheral blood from myeloma patients as late stage B cells.
Blood
78
1991
711
27
Bergsagel
PL
Masellis Smith
A
Szczepek
A
Mant
MJ
Belch
AR
Pilarski
LM
In multiple myeloma, clonotypic B lymphocytes are detectable among CD19+ peripheral blood cells expressing CD38, CD56, and monotypic immunoglobulin light chain.
Blood
85
1995
436
28
Kubagawa
H
Vogler
LB
Capra
JD
Conrad
ME
Lawton
AR
Cooper
MD
Studies on the clonal origin of multiple myeloma.
J Exp Med
150
1979
792
29
Billadeau
D
Ahmann
G
Greipp
P
Van Ness
B
The bone marrow of multiple myeloma patients contains B cell populations at different stages of differentiation that are clonally related to the malignant plasma cell.
J Exp Med
178
1993
1023
30
Bakkus
MHC
van Riet
I
Van Camp
B
Thielemann
K
Evidence that the clonogenic cell in multiple myeloma originates from a pre-switched but somatically mutated B-cell.
Br J Haematol
87
1994
68
31
Hitzler
JK
Martinez-Valdez
H
Bergsagel
DB
Minden
MD
Messner
HA
Role of interleukin-6 in the proliferation of human multiple myeloma cell lines OCI-My 1 to 7 established from patients with advanced stage of the disease.
Blood
78
1991
1996
32
Okuno
Y
Takahashi
T
Suzuki
A
Fukumoto
M
Nakamura
K
Fukui
H
Koishihara
Y
Ohsugi
Y
Imura
H
Acquisition of growth autonomy and tumorigenicity by an interleukin-6 dependent human myeloma cell line transfected with interleukin-6 cDNA.
Exp Hematol
20
1992
395
33
Szczepek
AJ
Bergsagel
PL
Axelsson
L
Brown
CB
Belch
AR
Pilarski
LM
CD34+ cells in the blood of patients with multiple myeloma express CD19 and IgH mRNA and have patient specific IgH rearrangements.
Blood
89
1997
1824
34
Pilarski
LM
Gillitzer
R
Zola
H
Shortman
K
Scollay
R
Selective expression of CD45 (T200) antigens during human thymocyte differentiation.
Eur J Immunol
19
1988
589
35
Pilarski
LM
Szczepek
AJ
Belch
AR
Deficient drug transporter function of bone marrow localized and leukemic plasma cells in multiple myeloma.
Blood
90
1997
3751
36
Szczepek
AJ
Seeberger
K
Wizniak
J
Mant
MJ
Belch
AR
Pilarski
LM
A high frequency of circulating B cells share clonotypic IgH VDJ rearrangements with autologous bone marrow plasma cells in multiple myeloma, as measured by single cell and in-situ RT-PCR.
Blood
92
1998
2844
37
Hokland
P
Hokland
M
Heron
I
An improved technique for obtaining E rosettes with human lymphocytes and its use for B cell purification.
J Immunol Methods
13
1976
175
38
Urbano-Ispizua
A
Rozman
C
Martinez
C
Marin
P
Briones
J
Rovira
M
Feliz
P
Viguria
MC
Merino
A
Sierra
J
Mazzara
R
Carreras
E
Montserra
E
Rapid engraftment without significant graft versus host disease after allogeneic transplantation of CD34+ selected cells from peripheral blood.
Blood
89
1997
3967
39
Goldmacher
VS
Bourret
LA
Levine
BA
Rasmussen
RA
Pourshadi
M
Lambert
JM
Anderson
KC
Anti-CD-38-blocked ricin: An immunotoxin for the treatment of multiple myeloma.
Blood
84
1994
3017
40
Stevenson
FK
Bell
AJ
Cusack
R
Hamblin
TJ
Slade
CJ
Spellerberg
MB
Stevenson
GT
Preliminary studies for an immunotherapeutic approach to the treatment of human myeloma using chimeric anti-CD38 antibody.
Blood
77
1991
1071
41
Huang
YW
Richardson
JA
Vitetta
ES
Anti-CD54 (ICAM-1) has antitumour activity in SCID mice with human myeloma cells.
Cancer Res
55
1995
610
42
Ozaki
S
Kosaka
M
Wakatsuki
S
Abe
M
Koishihara
Y
Matsumoto
T
Immunotherapy of multiple myeloma with a monoclonal antibody directed against a plasma cell-specific antigen, HM1.24.
Blood
90
1997
3179
43
Vooijs
WC
Schuurman
HJ
Bast
EJ
deGast
GC
Evaluation of CD38 as a target for immunotherapy in multiple myeloma.
Blood
85
1995
2282
44
Voojis
WC
Post
J
Wijdenes
J
Schuurman
HJ
Bolognesi
A
Polito
L
Stirpe
F
Bast
EJ
deGast
GC
Efficacy and toxicity of plasma cell reactive monoclonal antibodies B-B2 and B-B4 and their immunotoxins.
Cancer Immunol Immunother
42
1996
319
45
Terstappen
LW
Shiang
H
Safford
M
Lansdorp
PM
Loken
MR
Sequential generations of hematopoietic colonies derived from single nonlineage-committed CD34+CD38− progenitor cells.
Blood
77
1991
1218
46
Kotera
Y
Fontenot
JD
Pecher
G
Metzgar
RS
Finn
OJ
Humoral immune response against a tandem repeat epitope of human mucin Muc-1 in sera from breast, pancreatic, and colon cancer patients.
Cancer Res
54
1994
2856
47
Gourevitch
MM
von Mensdorff-Pouilly
S
Litvinov
SV
Kenemans
P
van Kamp
GJ
Verstraeten
AA
Hilgers
J
Polymorphic epithelial mucin (Muc-1)-containing circulating immune complexes in carcinoma patients.
Br J Cancer
72
1995
934
48
Karanikas
V
Hwang
LA
Pearson
J
Ong
CS
Apostolopoulos
V
Vaughan
H
Xing
PX
Jamieson
G
Pietersz
G
Tait
B
Broadbent
R
Thynee
G
McKenzie
IF
Antibody and T cell responses of patients with adenocarcinoma immunized with mannan-MUC1 fusion protein.
J Clin Invest
100
1997
2783
49
Gilewski
T
Adluri
R
Zhang
S
Houghton
A
Norton
L
Livingston
P
MUC-1 keyhole limpet hemocyanin (KLH) conjugate plus QS-21 vaccination of high risk breast cancer patients with no evidence of disease.
Proc Am Soc Clin Oncol
16
1997
A1569
(abstr)
50
Graham
RA
Burchell
JM
Beverley
P
Taylor-Papademetriou
J
Intramuscular immunization with Muc1 cDNA can protect C57 mice challenged with Muc1-expressing syngeneic mouse tumour cells.
Int J Cancer
65
1996
664
51
Akagi
J
Hodge
JW
McLaughlin
JP
Gritz
L
Mazzara
G
Kufe
D
Schlom
J
Kantor
JA
Therapeutic antitumor response after immunization with an admixture of recombinant vaccinia viruses expressing a modified Muc1 gene and the murine T-cell costimulatory molecule B7.
J Immunother
20
1997
38
52
Zhang
S
Graeber
LA
Helling
F
Ragupathi
G
Adluri
S
Lloyd
KO
Livingston
P
Augmenting the immunogenicity of synthetic Muc1 peptide vaccines in mice.
Cancer Res
56
1996
3315
53
Gong
J
Chen
D
Kashiwaba
M
Kufe
D
Induction of antitumor activity by immunization with fusions of dendritic and carcinoma cells.
Nat Med
3
1997
558
54
Friedmann
EL
Hayes
DF
Kufe
DW
Reactivity of monoclonal antibody DF3 with a high molecular weight antigen expressed in human ovarian carcinomas.
Cancer Res
46
1986
5189
55
Hird
V
Maraveyas
A
Snook
D
Dhokia
B
Soutter
WP
Meares
C
Stewart
JSW
Mason
P
Lambert
HE
Epenetos
AA
Adjuvant therapy of ovarian cancer with radioactive monoclonal antibody.
Br J Cancer
68
1993
403
56
Cao
Y
Karsten
U
Hilgers
J
Immunohistochemical characterization of a panel of 56 antibodies with normal human, small intestine, colon, and breast tissues.
Tumor Biol
19
1998
88
57
Gregoretti
MG
Gottardi
D
Ghia
P
Bergui
L
Merico
F
Marchisio
PC
Caligaris-Cappio
F
Characterization of bone marrow stromal cells from multiple myeloma.
Leuk Res
18
1994
675
58
Carter
A
Merchav
S
Silvan-Draxler
I
Tatarsky
I
The role of interleukin-1 and tumor necrosis factor alpha in human multiple myeloma.
Br J Haematol
74
1990
424
59
Lu
ZY
Bataille
R
Poubelle
P
Rapp
MJ
Harousseau
JL
Klein
B
An interleukin 1 receptor antagonist blocks the IL-1 induced IL-6 paracrine production through a prostaglandin E2-related mechanism in multiple myeloma.
Stem Cells
113
1995
28
60
Dittel
BN
McCarthy
JB
Wayner
EA
LeBien
TW
Regulation of human B-cell precursor adhesion to bone marrow stromal cells by cytokines that exert opposing effects on the expresson of vascular cell adhesion molecule-1.
Blood
81
1993
2272
61
Yang
J
Xu
Y
Zhu
C
Hagan
MK
Lawley
T
Offerman
MK
Regulation of adhesion molecule expression in Kaposi’s Sarcoma cells.
J Immunol
152
1994
361
62
Rettig
MB
MA
HJ
Vescio
RA
Pold
M
Schiller
G
Belson
D
Savage
A
Nishikubo
C
Wu
C
Fraser
J
Said
JW
Berenson
J
Kaposi’s sarcoma associated herpesvirus infection of bone marrow dendritic cells from multiple myeloma patients.
Science
276
1997
1851
63
Offerman
MK
Lin
JC
Mar
EC
Shaw
R
Yang
J
Medford
RM
Antioxidant sensitive regulation of inflammatory response genes in Kaposi’s Sarcoma cells.
J AIDS Hum Retroviruses
13
1996
1
64
Treon
SP
Anderson
KC
Interleukin-6 in multiple myeloma and related plasma cell dyscrasias.
Curr Opin Hematol
5
1998
42
65
Brady
ME
Sartiano
GP
Rosenblum
SL
Zaglama
NE
Bauguess
CT
The pharmacokinetics of single high doses of dexamethasone in cancer patients.
Eur J Clin Pharm
32
1987
593

Author notes

Address reprint requests to Kenneth C. Anderson, MD, Division of Adult Oncology, Dana Farber Cancer Institute, 44 Binney St, Boston, MA 02115; e-mail: kenneth_anderson@dfci.harvard.edu.

Sign in via your Institution