Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
J Urol ; 175(1): 57-62, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16406869

ABSTRACT

PURPOSE: WX-G250 is a chimeric monoclonal antibody that binds to carbonic anhydrase IX(G250/MN), which is present on greater than 95% of RCCs of the clear cell subtype. The suggested working mechanism of WX-G250 is by ADCC. Because the number of activated ADCC effector cells can be increased by a low dose interleukin-2 pulsing schedule, a multicenter study was initiated to investigate whether WX-G250 combined with LD-IL-2 could lead to an improved clinical outcome in patients with progressive RCC. MATERIALS AND METHODS: A total of 35 patients with progressive clear cell RCC received weekly infusions of WX-G250 for 11 weeks combined with a daily LD-IL-2 regimen. Patients were monitored longitudinally for ADCC capacity. Radiological assessment of metastatic lesions was performed at week 16 and regularly until disease progression. RESULTS: A durable clinical benefit was achieved in 8 of 35 patients (23%), including 3 with a partial response and 5 with stabilization at 24 weeks or greater. Mean survival was 22 months. In general treatment was well tolerated with little toxicity. The number of effector cells increased during treatment but lytic capacity per cell did not increase. ADCC and clinical outcome did not appear to correlate. CONCLUSIONS: WX-G250 combined with LD-IL-2 in patients with metastatic RCC is safe and well tolerated. With a substantial clinical benefit and a median survival of 22 months in patients with metastatic RCC who have progressive disease at study entry combination therapy showed increased overall survival compared to WX-G250 monotherapy. Survival was at least similar to that of currently used cytokine regimens but with a favorable toxicity profile.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Carcinoma, Renal Cell/drug therapy , Interleukin-2/administration & dosage , Kidney Neoplasms/drug therapy , Adult , Aged , Carcinoma, Renal Cell/pathology , Disease Progression , Drug Therapy, Combination , Female , Humans , Kidney Neoplasms/pathology , Male , Middle Aged , Prospective Studies
2.
Mol Cell Biol ; 21(7): 2570-80, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11259604

ABSTRACT

Ep-CAM is a new type of cell adhesion molecule (CAM) which does not structurally resemble the members of the four major families (cadherins, integrins, selectins, and CAMs of the immunoglobulin superfamily) and mediates Ca(2+)-independent, homophilic adhesions. The extracellular domain of Ep-CAM consists of a cysteine-rich region, containing two type II epidermal growth factor (EGF)-like repeats, followed by a cysteine-poor region. We generated mutated Ep-CAM forms with various deletions in the extracellular domain. These deletion mutants, together with monoclonal antibodies recognizing different epitopes in the extracellular domain, were used to investigate the role of the EGF-like repeats in the formation of intercellular contacts mediated by Ep-CAM molecules. We established that both EGF-like repeats are required for the formation of Ep-CAM-mediated homophilic adhesions, including the accumulation of Ep-CAM molecules at the cell-cell boundaries, and the anchorage of the Ep-CAM adhesion complex to F-actin via alpha-actinin. Deletion of either EGF-like repeat was sufficient to inhibit the adhesion properties of the molecule. The first EGF-like repeat of Ep-CAM is required for reciprocal interactions between Ep-CAM molecules on adjacent cells, as was demonstrated with blocking antibodies. The second EGF-like repeat was mainly required for lateral interactions between Ep-CAM molecules. Lateral interactions between Ep-CAM molecules result in the formation of tetramers, which might be the first and necessary step in the formation of Ep-CAM-mediated intercellular contacts.


Subject(s)
Antigens, Neoplasm/physiology , Cell Adhesion Molecules/physiology , Epidermal Growth Factor/genetics , Cell Adhesion/genetics , Cell Line , Epithelial Cell Adhesion Molecule , Epithelial Cells/cytology , Epithelial Cells/physiology , Gene Expression Regulation , Humans , Mutation , Tandem Repeat Sequences/genetics
3.
Cancer Res ; 60(7): 1921-6, 2000 Apr 01.
Article in English | MEDLINE | ID: mdl-10766181

ABSTRACT

Murine monoclonal antibodies (mAbs), when administered to patients, induce a human antimouse immunoglobulin immune response, especially when multiple infusions are required to obtain therapeutic efficacy. In a randomized Phase II clinical study, 83 patients with colorectal carcinoma of stage Dukes C were treated with the murine IgG2a mAb 17-1A (ab1) after curative surgery. The regimen consisted of a single infusion of 500mg of 17-1A within 2 weeks after surgery, followed by 100mg of mAbs four times every 4 weeks. Sera were taken every 2-3 weeks and screened for human antimouse antibodies (HAMA). HAMA were measured by a capture ELISA and an indirect antihuman immunoglobulin ELISA for the analysis of IgG and IgM isotypes. Anti-idiotypic antibodies (ab2) were detected by an inhibition ELISA, and anti-anti-idiotypic antibodies (ab3), recognizing the original antigen, were determined by flow cytometric analysis. About 20% of patients failed to develop HAMA; in the other patients, antibody titers were initially low after the first two infusions and reached their maximum only after a fifth infusion at 18-20 weeks after surgery. An analysis that differentiated between patients who developed recurrences and those who remained tumor-free did not show any difference in antibody titers between the two groups, neither for total HAMA nor for IgG, IgM, or ab2. The formation of ab3 was analyzed in eight patients and proved to be negative in all of them. HAMA remained detectable up to 2 years after the last treatment. In patients who experienced adverse events associated with therapy, HAMA titers tended to rise earlier; this difference, however, was not statistically significant. Thus, neither a beneficial nor a detrimental effect of HAMA formation could be determined for the clinical response to antibody therapy.


Subject(s)
Antibodies, Anti-Idiotypic/blood , Antibodies, Monoclonal/therapeutic use , Antibody Formation , Antigens, Neoplasm/immunology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/immunology , Anaphylaxis , Animals , Antibodies, Monoclonal/adverse effects , Colorectal Neoplasms/pathology , Colorectal Neoplasms/surgery , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , Mice , Neoplasm Staging , Predictive Value of Tests , Treatment Outcome
4.
Am J Pathol ; 155(2): 505-15, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10433943

ABSTRACT

Uterine cervix represents a convenient model for the study of the gradual transformation of normal squamous epithelium via low- to high-grade squamous intraepithelial lesions (SILs). Because SIL, on the basis of the cytokeratins expressed, are thought to originate from the reserve cells, we analyzed whether SILs also show a reserve cell phenotype with respect to intercellular interactions. The changes in expression and subcellular localization of the components of the adherens junction and desmosomal complexes were investigated in normal, metaplastic, and premalignant cervical epithelium, as well as in cell cultures derived from these tissues. The results suggest that 1) during progression of SILs, E-cadherin is suppressed, with its role in cell-cell connections diminishing; 2) P-cadherin, in contrast, becomes the predominant cadherin in high-grade SILs; 3) the level of cellular alpha-catenin is dramatically decreased in high-grade SILs; 4) the level of beta-catenin is decreased during progression of SILs, with plakoglobin suggestively becoming the predominant catenin mediating connection of cadherins to the cytoskeleton; 5) the assembly of desmosomes is affected during progression of SILs and is accompanied by a dramatically decreased expression for desmogleins and desmoplakins (I, II); and 6) expression of differentiation markers (involucrin, CK13) in high-grade SILs seems to be controlled by P-cadherin as opposed to E-cadherin in the normal tissue counterpart. We conclude that during development of cervical lesions substantial (both quantitative and qualitative) changes occur in cell-cell junctions, making the interactions of cells in lesions dissimilar from those of reserve cells, basal cells, or cells of immature squamous metaplasia, despite existing morphological similarity between all of these cell types and cells of high-grade lesions.


Subject(s)
Cadherins/physiology , Cytoskeletal Proteins/physiology , Trans-Activators , Uterine Cervical Dysplasia/metabolism , Uterine Cervical Neoplasms/metabolism , Biomarkers , Cadherins/analysis , Cell Adhesion Molecules/analysis , Cell Adhesion Molecules/physiology , Cells, Cultured , Cytoskeletal Proteins/analysis , Desmogleins , Desmoplakins , Desmosomes/metabolism , Disease Progression , Female , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Humans , Immunohistochemistry , Protein-Tyrosine Kinases/analysis , Protein-Tyrosine Kinases/physiology , Time Factors , Uterine Cervical Neoplasms/diagnosis , alpha Catenin , beta Catenin , gamma Catenin
5.
Exp Cell Res ; 246(1): 108-21, 1999 Jan 10.
Article in English | MEDLINE | ID: mdl-9882520

ABSTRACT

The epithelial cell adhesion molecule Ep-CAM is capable of mediating Ca2+-independent homotypic cell-cell adhesion when introduced into cells lacking their own means of cell-cell interactions. We used (confocal) immunofluorescent and (immuno-) electron microscopy to investigate the structural organization of Ep-CAM-mediated adhesions and their relation to other types of intercellular adhesions. Ep-CAM-transfected cell lines, cells of epithelial origin, and epithelial tissues were analyzed. In transfected L cells Ep-CAM brings the opposing intercellular membranes into a close proximity (approximately 10-14 nm) at sporadic contacts; however, no structures resembling junctional complexes were observed. In L cells cotransfected with Ep-CAM and E-cadherin, both molecules localize at the sites of cell-cell contact, forming independent adhesion sites with no Ep-CAM detectable within the structurally distinguishable cadherin-mediated adherens junctions. In well-differentiated carcinoma cell lines Ep-CAM colocalized with E-cadherin practically along the whole lateral domain; however, no colocalization was observed between Ep-CAM and the components of the tight junction complex (occludin and ZO-1), desmosomes (desmoplakins I/II), or cell-substrate adhesions (beta1 integrins). This was confirmed by analysis of polarized epithelium of normal colon where Ep-CAM was present at the lateral membrane including the adherens junction areas, but was fully excluded from the apical cell membrane (microvilli), tight junctions, and desmosomes. We conclude that (1) Ep-CAM does not form junctional complexes in L cells, (2) in epithelial cells, cell surface Ep-CAM is present at the lateral cell membrane, but is excluded from tight junctions and desmosomes, and (3) in epithelial cells, Ep-CAM is present within adhesions mediated by the classic cadherins (especially E-cadherin) with both types of molecules remaining as independent clusters. The colocalization with cadherins might be important for the modulating effect of Ep-CAM on cadherin-mediated adhesions.


Subject(s)
Antigens, Neoplasm/metabolism , Cadherins/metabolism , Cell Adhesion Molecules/metabolism , Cell Adhesion/physiology , Intercellular Junctions/metabolism , Animals , Antigens, Neoplasm/genetics , Cadherins/genetics , Cadherins/ultrastructure , Cell Adhesion Molecules/genetics , Cell Line , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Colon/cytology , Cytoskeletal Proteins/metabolism , Desmoplakins , Epithelial Cell Adhesion Molecule , Epithelial Cells , Humans , Integrin beta1/metabolism , Intercellular Junctions/ultrastructure , Membrane Proteins/metabolism , Mice , Microscopy, Immunoelectron , Models, Biological , Occludin , Phosphoproteins/metabolism , Pseudopodia/metabolism , Pseudopodia/ultrastructure , Transfection , Tumor Cells, Cultured , Zonula Occludens-1 Protein
6.
Br J Haematol ; 102(3): 783-90, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9722307

ABSTRACT

Interleukin 6 plays a key role in the pathogenesis of multiple myeloma (MM). Therefore we conducted a phase I dose-escalating study with chimaeric monoclonal anti-IL6 antibodies (cMab) in MM patients resistant to second-line chemotherapy. The cMab (CLB IL6/8; Kd 6.25 x 10(-12)M) was given in two cycles of 14 daily infusions, starting on day 1 and day 28, repectively, with a daily dose of 5 mg in patients 1-3, 10 mg in patients 4-6, 20 mg in patients 7-9 and 40mg in patients 10-12 (total dose 140 mg, 280mg, 560 mg and 1120 mg of anti-IL6, respectively). 11/12 patients had elevated pretreatment IL6 levels. Except for transient thrombocytopenia in two patients there was no toxicity. There were no changes in haemoglobin levels, granulocyte count, liver enzymes or renal function. No human anti-chimaeric antibodies were induced. This was also reflected in a long half-life time of the cMab (median 17.8 d), resulting in accumulation of the anti-IL6 cMab and high levels of circulating IL6. However, this was in the form of biologically inactive IL6/cMab complexes and did not result in acceleration of the disease. Although C-reactive protein (CRP) levels were decreased to below detection level in 11/12 patients, indicating effective IL6 blocking, none of the patients achieved a response according to the standard criteria. We conclude that this chimaeric anti-IL6 Mab has a low toxicity, low immunogenicity and a long T1/2. A dose of 40 mg/d for 14 d can safely be used in future phase II studies.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Interleukin-6/administration & dosage , Multiple Myeloma/therapy , Muscle Proteins , Aged , Agranulocytosis/etiology , Antibodies, Monoclonal/analysis , Antibodies, Monoclonal/pharmacokinetics , C-Reactive Protein/analysis , Connectin , Dose-Response Relationship, Immunologic , Female , Half-Life , Humans , Interleukin-6/immunology , Interleukin-6/pharmacokinetics , Male , Middle Aged , Multiple Myeloma/blood , Myeloma Proteins/analysis , Recombinant Fusion Proteins , Survival Analysis , Thrombocytopenia/etiology , Treatment Outcome , beta 2-Microglobulin/analysis
7.
Br J Cancer ; 78(4): 478-83, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9716030

ABSTRACT

Antibody-dependent cellular cytotoxicity (ADCC) is considered to be the major mechanism through which tumour cells, upon treatment with anti-tumour MAbs, are eliminated in vivo. However, the relative importance of various parameters that influence the efficacy of ADCC is unclear. Here we present in vitro data on the impact of MAb affinity and antigen density on ADCC, as obtained by comparison of two MAbs against the tumour-associated antigen Ep-CAM. The low-affinity MAb 17-1A (Ka = 5 x 10(7)M(-1)) currently used for therapy, and the high-affinity MAb 323/A3 (Ka = 2 x 10(9) M(-1)), were compared in ADCC experiments against murine and human tumour target cells transfected with the Ep-CAM cDNA under the control of an inducible promoter to enable regulation of the target antigen expression levels. Data obtained from these studies revealed that the high-affinity MAb, in contrast to the low-affinity MAb, could mediate killing of tumour cells with low antigen expression levels. Even at comparable MAb-binding levels, ADCC mediated by the high-affinity MAb was more effective. The kinetics of ADCC was also found to be determined by the level of antigen expression, and by the affinity and the concentration of the MAb used. The efficacy of ADCC with both low- and high-affinity MAbs further depended on adhesive interactions between effector and target cells mediated by CD18. However, at every given MAb concentration these interactions were of less importance for the high-affinity MAb than for the low-affinity MAb. As heterogeneity of a target antigen expression is a common feature of all tumours, and some tumour cells express very low levels of the antigen, the use of high-affinity MAbs in immunotherapy may significantly improve the clinical results obtained to the present date in the treatment of minimal residual disease.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibody Affinity , Antibody-Dependent Cell Cytotoxicity , Antigens, Neoplasm/immunology , Cell Adhesion Molecules/immunology , Neoplasms/immunology , Animals , Epithelial Cell Adhesion Molecule , Humans , Lymphocyte Function-Associated Antigen-1/immunology , Mice , Neoplasms/therapy , Transfection , Tumor Cells, Cultured
8.
Mol Cell Biol ; 18(8): 4833-43, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9671492

ABSTRACT

Ep-CAM, an epithelium-specific cell-cell adhesion molecule (CAM) not structurally related to the major families of CAMs, contains a cytoplasmic domain of 26 amino acids. The chemical disruption of the actin microfilaments, but not of the microtubuli or intermediate filaments, affected the localization of Ep-CAM at the cell-cell boundaries, suggesting that the molecule interacts with the actin-based cytoskeleton. Mutated forms of Ep-CAM were generated with the cytoplasmic domain truncated at various lengths. All of the mutants were transported to the cell surface in the transfectants; however, the mutant lacking the complete cytoplasmic domain was not able to localize to the cell-cell boundaries, in contrast to mutants with partial deletions. Both the disruption of the actin microfilaments and a complete truncation of the cytoplasmic tail strongly affected the ability of Ep-CAM to mediate aggregation of L cells. The capability of direct aggregation was reduced for the partially truncated mutants but remained cytochalasin D sensitive. The tail truncation did not affect the ability of the transfectants to adhere to solid-phase-adsorbed Ep-CAM, suggesting that the ability to form stable adhesions and not the ligand specificity of the molecule was affected by the truncation. The formation of intercellular adhesions mediated by Ep-CAM induced a redistribution to the cell-cell boundaries of alpha-actinin, but not of vinculin, talin, filamin, spectrin, or catenins. Coprecipitation demonstrated direct association of Ep-CAM with alpha-actinin. Binding of alpha-actinin to purified mutated and wild-type Ep-CAMs and to peptides representing different domains of the cytoplasmic tail of Ep-CAM demonstrates two binding sites for alpha-actinin at positions 289 to 296 and 304 to 314 of the amino acid sequence. The results demonstrate that the cytoplasmic domain of Ep-CAM regulates the adhesion function of the molecule through interaction with the actin cytoskeleton via alpha-actinin.


Subject(s)
Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/metabolism , Actinin/metabolism , Amino Acid Sequence , Animals , Antigens, Neoplasm/genetics , Binding Sites , Cell Adhesion , Cell Adhesion Molecules/genetics , Cell Extracts , Cell Line , Cell Line, Transformed , Cytochalasin D/pharmacology , Cytoplasm/metabolism , Detergents/pharmacology , Epithelial Cell Adhesion Molecule , Humans , Mice , Molecular Sequence Data , Mutagenesis , Nucleic Acid Synthesis Inhibitors/pharmacology , Octoxynol/pharmacology , Subcellular Fractions
9.
Leuk Lymphoma ; 31(5-6): 551-8, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9922045

ABSTRACT

Interleukin-6 (IL6) plays a major role in the pathogenesis of multiple myeloma. In patients with monoclonal gammopathy serum levels of sIL6R have been found to be increased. The role of IL6 in the regulation of soluble receptors is still unclear. In a phase I/II study we treated 12 myeloma patients with high-affinity chimeric anti-IL6 monoclonal antibodies. This treatment resulted in a total in vivo blockage of IL6 activity and as a result we had an unique opportunity to gain insight into the possible regulation effects of IL6 on these soluble IL6 receptors. Pre-treatment sIL6R levels were elevated in 9 of the 12 patients; pre-treatment sgp130 levels were significantly increased in all patients. Total blockage of IL6 activity by the high-affinity cMab did not influence sIL6R in 10 of these 12 patients and sgp130 levels remained stable in all patients. Of the 2 patients whose sIL6R levels increased during therapy, one had progressive disease and the other developed an acute infection. We conclude that in most end-stage myeloma patients sIL6R and sgp130 serum levels are elevated, but that there is no relation between IL6 activity and sIL6R or sgp130 levels.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, CD/blood , Immunization, Passive , Interleukin-6/antagonists & inhibitors , Membrane Glycoproteins/blood , Multiple Myeloma/therapy , Neoplasm Proteins/antagonists & inhibitors , Receptors, Interleukin-6/blood , Animals , Antibodies, Monoclonal/pharmacology , Cytokine Receptor gp130 , Disease Progression , Female , Humans , Interleukin-6/immunology , Interleukin-6/physiology , Male , Middle Aged , Multiple Myeloma/blood , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Neoplasm Proteins/analysis , Neoplasm Proteins/immunology , Neoplasm Proteins/physiology , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Solubility , Treatment Outcome
10.
J Immunother ; 20(6): 496-504, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9409456

ABSTRACT

The bispecific OC/TR monoclonal antibody (mAb) cross-links the CD3 molecule on T cells with the human folate-binding protein (FBP), which is highly expressed on nonmucinous ovarian carcinomas. Clinical trials of patients with ovarian carcinoma with the OC/TR mAb have shown some complete and partial responses. Most patients developed human anti-murine immunoglobulin antibodies (HAMA), which can inhibit OC/TR mAb-mediated lysis. We generated a chimeric version of the OC/TR mAb to decrease the immunogenicity of the OC/TR mAb and to allow more extended treatment schedules. Sp2/0 myeloma cells were transfected with chimeric heavy- and light-chain genes encoding the anti-CD3 mAb and the MOv18 mAb, respectively, which are reactive with FBP. The resulting cell line produced 80 micrograms/ml of total immunoglobulin G (IgG), of which 11.5% was the functionally active chimeric OC/TR mAb. Chimeric OC/TR F(ab')2 fragments mediated lysis of IGROV-1 ovarian carcinoma cells by human T cells at antibody concentrations of > or = 1 pg/ml. Specific lysis was still detectable at an effector-to-target cell ratio as low as 0.4. Two patients with ovarian carcinoma treated with F(ab')2 fragments of the murine OC/TR developed distinct HAMA titers, which were mainly anti-idiotypic and only partly directed against the murine antibody constant regions. However, of the two patients that were treated with the F(ab')2 fragments of the chimeric OC/TR mAb, only one developed a low transient HAMA response just above background level. In conclusion, the generation of chimeric OC/TR may allow more extended clinical studies of bispecific mAb-mediated immunotherapy of ovarian carcinoma.


Subject(s)
Antibodies, Bispecific/immunology , Antibodies, Monoclonal/immunology , Carrier Proteins/analysis , Cytotoxicity, Immunologic , Ovarian Neoplasms/immunology , Receptors, Cell Surface , Animals , Antibodies, Anti-Idiotypic/blood , Antibodies, Bispecific/genetics , Antibodies, Monoclonal/genetics , CD3 Complex/immunology , Cell Line , Female , Folate Receptors, GPI-Anchored , Gene Expression , Humans , Hybridomas , Immunoglobulin Fab Fragments/immunology , Immunotherapy , Mice , Recombinant Fusion Proteins , T-Lymphocytes/immunology , Transfection
11.
Metabolism ; 46(11): 1343-8, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9361697

ABSTRACT

Interleukin-6 (IL6) is believed to be involved in alterations of thyroid hormone metabolism in acute nonthyroidal illness. To evaluate the effects of IL6 on thyroid hormone metabolism in a chronic IL6-mediated disease, we measured thyroid hormone concentrations in multiple myeloma patients treated with intravenous anti-IL6 chimeric monoclonal antibodies ([cMabs] Kd = 6.25 x 10(-12) mol/L). Twelve patients were studied, receiving at least one complete treatment cycle of 14 days (daily dose: 5 mg, n = 3; 10 mg, n = 3; 20 mg, n = 3; and 40 mg, n = 3). Eight of them also completed a second treatment cycle of 14 days. Thyroid hormone concentrations were measured before, during, and after treatment with the anti-IL6 cMab. Even in the group with the lowest dosage, IL6 activity measured by the B9 bioassay was blocked completely. Compared with the reference ranges, 10 of 12 patients had one or more abnormal pretreatment values for thyroid hormone concentrations. Thyroid autoantibodies were negative in all patients. There was no correlation between thyroid hormone concentrations and IL6 levels, although plasma IL6 levels were increased in all but one subject. Moreover, neutralization of free IL6 by the anti-IL6 cMab did not affect thyroid hormone concentrations, although IL6-dependent C-reactive protein (CRP) levels decreased to undetectable levels in 11 of 12 patients. Two patients developed infectious complications resulting in increased free IL6 and CRP levels and in profound alterations of thyroid hormone levels consistent with an acute euthyroid sick syndrome. We conclude that IL6 is not a major determinant of thyroid hormone abnormalities in a chronic disease like multiple myeloma, but IL6 may be involved in thyroid hormone metabolism in acute diseases (probably in combination with other factors).


Subject(s)
Antibodies, Monoclonal/immunology , Interleukin-6/immunology , Multiple Myeloma/blood , Thyroid Hormones/blood , Thyrotropin/blood , Aged , Animals , Antibodies, Monoclonal/administration & dosage , Biological Assay , Female , Humans , Immunoglobulin G/administration & dosage , Immunoglobulin G/immunology , Infusions, Intravenous , Interleukin-6/administration & dosage , Interleukin-6/blood , Male , Mice , Middle Aged , Multiple Myeloma/immunology , Multiple Myeloma/physiopathology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/immunology , Thyroid Hormones/immunology , Thyroid Hormones/metabolism , Thyrotropin/immunology , Thyrotropin/metabolism , Thyroxine/blood , Thyroxine/immunology , Thyroxine/metabolism , Time Factors , Triiodothyronine/blood , Triiodothyronine/immunology , Triiodothyronine/metabolism
12.
Int J Cancer ; 73(2): 211-9, 1997 Oct 09.
Article in English | MEDLINE | ID: mdl-9335445

ABSTRACT

We have reported a 27% overall anti-tumor response using i.p. immunotherapy of advanced ovarian carcinoma with autologous, ex vivo expanded, T lymphocytes re-targeted with bi-specific monoclonal antibody OC/TR, combined with soluble OC/TR and low-dose recombinant interleukin-2 (IL-2). This treatment had no effect on extraperitoneal disease. Therefore we studied in 13 patients whether this immunotherapeutic protocol resulted only in local or also in systemic immunomodulation. The phenotype of the ex vivo expanded lymphocytes was mainly CD3+, 4-, 8+, 16-, 56-. Their OC/TR-re-targeted cytolytic activity against Igrov-1 ovarian-carcinoma cells was approximately as high in responders as in non-responders. Following most therapeutic cycles, the immunophenotype of lymphocytes recovered from the peritoneal fluid was similar to that of the infused T cells (i.e., mainly CD3+, 4-, 8+) and they were coated with OC/TR. However, cytolytic activity of the recovered lymphocytes against Igrov- 1 cells was low in direct assays, and only slightly increased after additional in vitro re-targeting with OC/TR. Systemically, the i.p. immunotherapy resulted in a transient lymphopenia lasting for about 7 days, low (i.e., 5 to 13 ng/ml) serum concentrations of free, functional OC/TR, and very weak coating of circulating T lymphocytes with OC/TR. These peripheral-blood T lymphocytes did not exert OC/TR-re-targeted cytolytic activity. Thus, locoregional OC/TR-re-targeted cellular immunotherapy resulted in substantial local immunomodulation and anti-tumor effects but virtually no systemic immunomodulation.


Subject(s)
Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal/therapeutic use , Immunotherapy , Ovarian Neoplasms/immunology , T-Lymphocytes/immunology , Ascitic Fluid/cytology , CD3 Complex/immunology , Cytotoxicity, Immunologic , Female , Flow Cytometry , Humans , Immunophenotyping , Infusions, Parenteral , Leukapheresis , Lymphocyte Activation/immunology , Lymphocyte Count , Ovarian Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology
13.
Hum Antibodies ; 8(4): 169-80, 1997.
Article in English | MEDLINE | ID: mdl-9395919

ABSTRACT

Crosslinking of immunoglobulin E molecules that are bound to the Fc epsilon receptors expressed on mast cells or basophils triggers activation of these cells, resulting in the development of a type I hypersensitivity. Targeting this potent immune reaction towards tumors by using IgE that reacts with a tumor-associated antigen, may induce a local inflammation at the tumor site, and may therefore promote tumor regression. We have previously shown that murine IgE bound to tumor cells can activate murine mast cells to release TNF-alpha and histamine. To further investigate the therapeutic potential of IgE-mediated immunotherapy of carcinomas, we have developed human/murine chimeric versions, containing the murine variable regions and human constant regions, of both G250 and 323/A3 IgE. These chimeric IgEs are reactive respectively with the G250 renal cell carcinoma antigen and the Ep-CAM molecule, which is highly expressed by most carcinomas. Transfection of the respective chimeric heavy and light chain genes into recipient Sp2/0 myeloma cells yielded chimeric IgE-producing clones. Chimeric G250 and 323/A3 IgE reacted with tumor cells expressing the G250 antigen or Ep-CAM, respectively. To generate a cell line that expresses Fc receptors for human or chimeric IgE, the rat basophilic leukemia cell line RBL-7 was transfected with the human Fc epsilon RI alpha chain (RBL-7TZ) and subsequently tested for binding of chimeric IgE. Functional assays showed that both chimeric IgEs activated RBL-7TZ cells to release TNF-alpha when cultured with tumor cells that express the respective specific antigen. Furthermore, both chimeric IgEs were able to activate freshly isolated human basophils.


Subject(s)
Antigens, Neoplasm , Immunoglobulin E/immunology , Receptors, IgE/metabolism , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Basophils/immunology , Humans , Hypersensitivity, Immediate/immunology , Immunoglobulin E/genetics , Immunoglobulin E/metabolism , In Vitro Techniques , Mast Cells/immunology , Mice , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Transfection , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism
14.
J Cell Biol ; 139(5): 1337-48, 1997 Dec 01.
Article in English | MEDLINE | ID: mdl-9382878

ABSTRACT

The contribution of noncadherin-type, Ca2+-independent cell-cell adhesion molecules to the organization of epithelial tissues is, as yet, unclear. A homophilic, epithelial Ca2+-independent adhesion molecule (Ep-CAM) is expressed in most epithelia, benign or malignant proliferative lesions, or during embryogenesis. Here we demonstrate that ectopic Ep-CAM, when expressed in cells interconnected by classic cadherins (E- or N-cadherin), induces segregation of the transfectants from the parental cell type in coaggregation assays and in cultured mixed aggregates, respectively. In the latter assay, Ep-CAM-positive transfectants behave like cells with a decreased strength of cell-cell adhesion as compared to the parental cells. Using transfectants with an inducible Ep-CAM-cDNA construct, we demonstrate that increasing expression of Ep-CAM in cadherin-positive cells leads to the gradual abrogation of adherens junctions. Overexpression of Ep-CAM has no influence on the total amount of cellular cadherin, but affects the interaction of cadherins with the cytoskeleton since a substantial decrease in the detergent-insoluble fraction of cadherin molecules was observed. Similarly, the detergent-insoluble fractions of alpha- and beta-catenins decreased in cells overexpressing Ep-CAM. While the total beta-catenin content remains unchanged, a reduction in total cellular alpha-catenin is observed as Ep-CAM expression increases. As the cadherin-mediated cell-cell adhesions diminish, Ep-CAM-mediated intercellular connections become predominant. An adhesion-defective mutant of Ep-CAM lacking the cytoplasmic domain has no effect on the cadherin-mediated cell-cell adhesions. The ability of Ep-CAM to modulate the cadherin-mediated cell-cell interactions, as demonstrated in the present study, suggests a role for this molecule in development of the proliferative, and probably malignant, phenotype of epithelial cells, since an increase of Ep-CAM expression was observed in vivo in association with hyperplastic and malignant proliferation of epithelial cells.


Subject(s)
Antigens, Neoplasm/metabolism , Cadherins/metabolism , Cell Adhesion Molecules/metabolism , Intercellular Junctions , Animals , Antigens, Neoplasm/genetics , Cell Adhesion Molecules/genetics , Cell Aggregation , Cell Line , Cytoskeleton/metabolism , Epithelial Cell Adhesion Molecule , Epithelial Cells , Humans , Mice , Protein Binding , Recombinant Proteins/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured
15.
J Clin Invest ; 98(6): 1441-8, 1996 Sep 15.
Article in English | MEDLINE | ID: mdl-8823310

ABSTRACT

In vitro as well as in vivo observations have shown that IL6 plays a key role in the pathogenesis of multiple myeloma. Therefore we started a phase I/II dose escalating study with chimeric monoclonal anti-IL6 antibodies (cMab) in multiple myeloma (MM) patients resistant to second-line chemotherapy. Here we describe the pharmacological data as well as a new method for calculating the endogenous IL6 production. The cMab (CLB IL6/8; Kd: 6.25 x 10(-12) M) was given in two cycles of 14 daily infusions, starting on day 1 and day 28. Daily dose: 5 mg in patients 1-3, 10 mg in patients 4-6, and 20 mg in patients 7-9 (total dose 140, 280, and 560 mg of anti-IL6, respectively). Using the pharmacokinetic data of free IL6 and the binding characteristics of the cMab, the endogenous IL6 production could be calculated from day to day using a one-compartment open model. The median half-life time of this antibody was 17.6 d. No human antichimeric antibodies were induced. Pre-treatment median endogenous IL6 production in the MM patients was 60 micrograms/d (range 13.8-230; normal controls < 7 micrograms/d). During treatment with anti-IL6 cMabs, the endogenous IL6 production immediately decreased in all patients to below 3 micrograms/d and never reached the pre-treatment value during the treatment period, except in two patients who developed an active infection, resulting in an IL6 production of 128 and 1,208 micrograms/d, respectively. We concluded that in MM patients endogenous IL6 production is 2-30 times higher than in healthy individuals. The anti-IL6 cMab strongly suppress this endogenous IL6 production, probably by blocking a positive feed-back loop, but this cMab does not prevent infection-induced IL6 production. The chimeric anti-IL6 Mabs have a long half-life time, a low immunogenicity, and are able to block IL6-dependent processes in vivo.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Interleukin-6/biosynthesis , Interleukin-6/immunology , Multiple Myeloma/metabolism , Multiple Myeloma/therapy , Aged , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacokinetics , Drug Resistance, Neoplasm , Female , Humans , Interleukin-6/pharmacokinetics , Male , Middle Aged , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/therapeutic use
16.
Br J Cancer ; 74(5): 735-44, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8795576

ABSTRACT

The monoclonal antibody (MAb) G250 binds to a tumour-associated antigen, expressed in renal cell carcinoma (RCC), which has been demonstrated to be a suitable target for antibody-mediated immunotherapy. A bispecific antibody having both G250 and anti-CD3 specificity can cross-link G250 antigen-expressing RCC target cells with T cells and can mediate lysis of such targets. Therapy studies with murine antibodies are limited by immune responses to the antibodies injected (HAMA response), which can be decreased by using chimeric antibodies. We generated a chimeric bispecific G250/anti CD3 MAb by transfecting chimeric genes of heavy and light chains for both the G250 MAb and the anti-CD3 MAb into a myeloma cell line. Cytotoxicity assays revealed that the chimeric bispecific MAb was capable of mediating lysis of RCC cell lines by cloned human CD8+T cells or by IL-2-stimulated peripheral blood lymphocytes (PBLs). Lysis mediated by the MAb was specific for target cells that expressed the G250 antigen and was effective at concentrations as low as 0.01 microgram ml-1. The chimeric bispecific G250/anti-CD3 MAb produced may be an effective adjuvant to the currently used IL-2-based therapy of advanced renal cell arcinoma.


Subject(s)
Antibodies, Bispecific/biosynthesis , Antibodies, Monoclonal/biosynthesis , CD3 Complex/immunology , Carcinoma, Renal Cell/immunology , Kidney Neoplasms/immunology , Recombinant Fusion Proteins/immunology , Animals , Antibodies, Bispecific/isolation & purification , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Antibody Specificity , Base Sequence , Cytotoxicity, Immunologic , Lymphocytes/immunology , Mice , Molecular Sequence Data , Multiple Myeloma/genetics , Multiple Myeloma/immunology , Protein Binding , RNA, Messenger/biosynthesis , RNA, Neoplasm/chemistry , Transfection , Tumor Cells, Cultured/immunology
17.
J Immunother Emphasis Tumor Immunol ; 19(4): 245-56, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8877719

ABSTRACT

To evaluate the role of affinity in monoclonal antibody (mAb)-mediated treatment of carcinomas, we compared the antibodies 17-1A and 323/A3 that bind with different affinities overlapping epitopes on the epithelial adhesion molecule Ep-CAM. This comparison was performed in several models for minimal residual disease in mice grafted with Ep-CAM transfected B16 melanoma cells originating from C57BL/6 mice. These cells were either grafted subcutaneously or injected intravenously into nude BALB/c mice, or grafted subcutaneously in immunocompetent C57BL/6 mice. In the BALB/c subcutaneous model, significant therapeutic results (p < 0.05) compared with the control mAb were obtained with both mAbs 17-1A and 323/A3. However, when treating lung metastases in nude BALB/c mice that had developed after intravenous injection of the B16/Ep-CAM tumor cells, only the high-affinity 323/A3 mAb could significantly (p < 0.05) reduce the number of metastases that appeared. In syngeneic C57BL/6 mice grafted subcutaneously with B16/ Ep-CAM cells, a single 323/A3 or 17-1A mAb injection had no effect, in contrast to that observed for the nude BALB/c mouse model. However, multiple injections of the 323/A3 mAb significantly (p < 0.005) reduced the mean tumor volume, although they did not prevent tumor development. The results show that in vivo antibody-mediated effector cell activation and subsequent tumor cell elimination is determined by mAb affinity and target antigen density. Therefore, treatment of minimal residual disease with high-affinity mAb 323/ A3 is expected to improve the clinical results obtained with mAb 17-1A.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibody Affinity/immunology , Immunotherapy/methods , Melanoma, Experimental/therapy , Neoplasm, Residual/therapy , Animals , Disease Models, Animal , Flow Cytometry , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Male , Melanoma, Experimental/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Skin Neoplasms/therapy , Transfection/genetics
18.
Am J Pathol ; 148(3): 865-75, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8774141

ABSTRACT

Ep-CAM, an epithelial adhesion molecule, is absent in normal squamous epithelia but can be detected in some squamous carcinomas. Using a panel of monoclonal antibodies to keratinocyte differentiation and proliferation markers, we investigated the association of EP-CAM expression with differentiation-related and/or neoplastic changes in cervical epithelium. Normal endocervical glandular epithelium (Both columnar and reserve cells) appeared strongly positive for EP-CAM, whereas ectocervical squamous epithelial cells did not express this molecule. Expression of Ep-CAM (in basal cells) was sometimes observed in morphologically normal ectocervical tissue but only in areas bordering cervical intraepithelial neoplasia (CIN) lesions. At the early stages of neoplasia the expression of Ep-CAM was regularly present in squamous epithelium, in general consistent with the areas of atypical, undifferentiated cells. Thus, in CIN grades I and II, the basal/suprabasal layers of the epithelia were positive, whereas in CIN grade III lesions, up to 100% of the cells over the whole thickness of the epithelium sometimes excluding the very upper layers, expressed Ep-CAM. A clear increase, not only in number of positive cells but also in levels of Ep-CAM expression (intensity) was observed during progression from CIN I to CIN III. Expression of Ep-CAM in ectocervical lesions did not coincide with a reappearance of the simple epithelium cytokeratins (CK8 and CK18). On the other hand, expression of Ep-CAM in atypical cells of CIN lesions correlated with the disappearance of CK13, which normally marks cells undergoing squamous differentiation. As was shown with Ki-67, a marker for proliferating cell populations, the areas of Ep-CAM expression were also the areas of enhanced proliferation. Cells expressing Ep-CAM did not express involucrin, a marker for cells committed to terminal differentiation. In the majority of both squamous and adenocarcinomas of the cervix a strong expression of Ep-CAM was observed, although some decrease in the expression (both the intensity and the number of positive cells), as compared with CIN III lesions, was observed in the areas of squamous differentiation. This study demonstrates that the expression of Ep-CAM in cervical squamous epithelium is associated with abnormal proliferation of cell populations that are not committed to terminal differentiation.


Subject(s)
Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/metabolism , Cervix Uteri/cytology , Cervix Uteri/metabolism , Biomarkers , Biomarkers, Tumor , Cell Differentiation , Cell Division , Cervix Uteri/pathology , Epithelial Cell Adhesion Molecule , Epithelium/metabolism , Female , Humans , Keratins/metabolism , Metaplasia , Uterine Cervical Dysplasia/metabolism , Uterine Cervical Neoplasms/metabolism
19.
Lab Invest ; 74(2): 467-75, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8780164

ABSTRACT

Immunoglobulin E (IgE) that specifically binds to antigens present on carcinoma cells may represent a useful tool to combat carcinomas. Induction of an inflammatory response at the tumor site by tumor-specific IgE may result in reduced tumor growth and tumor regression. Local mast cells may be activated to release TNF-alpha and other mediators that attract inflammatory cells, such as eosinophils and macrophages, to the tumor site. It may even be expected that eosinophils perform IgE-mediated lysis of tumor cells. The G250 IgE binds an antigen present on renal cell carcinoma. Mast cells were assayed for activation by G250 IgE in vitro in the presence of G250-positive tumor cells, by determination of the release of TNF-alpha and histamine. In parallel, G250 IgG1, IgG2a, and IgG2b, bound to tumor cells, were tested for their ability to activate mast cells. Tumor-specific IgE was capable of activating mast cells in the presence of tumor cells. This activation was specific and required the presence of the antigen on the tumor cell surface and recognition of the tumor cell by the IgE. G250 IgE mediated mast cell activation when present in the medium as well as being preloaded on either tumor cells or mast cells. Preincubation of mast cells with irrelevant IgE did not block specific G250 IgE-mediated mast cell activation. Upon activation, mast cells released histamine and TNF-alpha, as was detected in cytotoxicity assays with TNF-alpha-sensitive target cells (WEHI). G250 IgG2a also induced efficient mast cell activation, comparable to the effect of G250 IgE. Mast cells can be triggered to release mediators such as TNF-alpha by IgE in the presence of tumor cells expressing specific antigen. Whether mast cell activation contributes to antitumor effects observed during antibody-based immunotherapy of tumors deserves further investigation.


Subject(s)
Antigens, Neoplasm/immunology , Carcinoma, Renal Cell/immunology , Immunoglobulin E/immunology , Immunoglobulin E/pharmacology , Kidney Neoplasms/immunology , Mast Cells/physiology , Animals , Antibodies, Monoclonal/pharmacology , Histamine Release , Humans , Immunoglobulin Isotypes/pharmacology , Mast Cells/drug effects , Mice , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism
20.
Cancer Res ; 55(19): 4398-403, 1995 Oct 01.
Article in English | MEDLINE | ID: mdl-7671252

ABSTRACT

The therapeutic effects of the low and high affinity mAbs 17-1A and 323/A3 were investigated in nude mice xenografted with LS 180 human colorectal carcinoma cells. Treatment of mice grafted with dispersed tumor cells, before formation of a tumor nodule, was started 1 day after s.c. injection of tumor cells and consisted of a single i.p. injection of murine 17-1A or 323/A3 mAb. Tumor appearance after a single injection of either 17-1A or 323/A3 was delayed as compared to injection of an irrelevant mAb. Both 17-1A and 323/A3 reduced the tumor growth rate, and both mAbs decreased the total number of mice that eventually developed a tumor. In all experiments, 323/A3 showed consistently better treatment effects on xenografted mice than mAb 17-1A. For treatment of established tumors with mAb 17-1A or 323/A3 therapy was delayed until a tumor nodule was macroscopically detectable. One single i.p. injection of mAb 17-1A had no effect on further tumor growth and mean tumor size as compared to the control group injected with irrelevant mAb. One single i.p. injection with mAb 323/A3 reduced the tumor growth rate in some mice with established tumors and resulted in a significant difference of mean tumor size of this group as compared to the 17-1A treated mice and the control groups. Multiple injections with mAb 17-1A also had no effects on established tumors, in contrast to mAb 323/A3, where serial injections resulted in tumor growth reduction and, eventually, in some mice reduction in tumor size. In summary, we showed that in nude mice mAb 323/A3 (Ka = 2 x 10(9) M-1) is much more potent than mAb 17-1A (Ka = 5 x 10(7) M-1) in eradication of nonestablished tumor cells and treatment of small established tumors. These results suggest that high affinity mAbs like 323/A3 might dramatically improve the clinical results obtained thus far with the low affinity mAb 17-1A in the adjuvant treatment of surgically resected Dukes C colorectal cancer patients with minimal residual disease.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Colorectal Neoplasms/therapy , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibody Affinity , Antibody-Dependent Cell Cytotoxicity , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Tissue Distribution , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL