Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 11(12): 2664-73, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23041545

ABSTRACT

For treatment of patients with prostate cancer (PCa), we developed a novel T cell-engaging (BiTE) antibody designated AMG 212 or BAY2010112 that is bispecific for prostate-specific membrane antigen (PSMA) and the CD3 epsilon subunit of the T cell receptor complex. AMG 212/BAY2010112 induced target cell-dependent activation and cytokine release of T cells, and efficiently redirected T cells for lysis of target cells. In addition to Chinese hamster ovary cells stably expressing human or cynomolgus monkey PSMA, T cells redirected by AMG 212/BAY2010112 also lysed human PCa cell lines VCaP, 22Rv1, MDA PCa 2b, C4-2, PC-3-huPSMA, and LnCaP at half maximal BiTE concentrations between 0.1 and 4 ng/mL (1.8-72 pmol/L). No lysis of PSMA-negative human PCa cell lines PC-3 and DU145 was observed. The subcutaneous (s.c.) formation of tumors from PC-3-huPSMA cells in NOD/SCID mice was significantly prevented by once daily intravenous (i.v.) injection of AMG 212/BAY2010112 at a dose level as low as 0.005 mg/kg/d. Rapid tumor shrinkage with complete remissions were observed in NOD/SCID mice bearing established s.c. 22Rv1 xenografts after repeated daily treatment with AMG 212/BAY2010112 by either the i.v. or s.c. route. Of note, 22Rv1 tumors were grown in the absence of human T cells followed by intraperitoneal injection of T cells 3 days before BiTE treatment. No effects on tumor growth were observed in the absence of human T cells or AMG 212/BAY2010112. On the basis of these preclinical results, AMG 212/BAY2010112 appears as a promising new BiTE antibody for the treatment of patients with PSMA-expressing PCa.


Subject(s)
Antibodies, Bispecific/pharmacology , Antigens, Surface/immunology , CD3 Complex/immunology , Glutamate Carboxypeptidase II/immunology , Prostatic Neoplasms/immunology , Prostatic Neoplasms/therapy , Animals , Antibodies, Bispecific/genetics , Antibodies, Bispecific/immunology , CD3 Complex/genetics , CHO Cells , Cricetinae , Cross Reactions , Female , Haplorhini , Humans , Immunization, Passive/methods , Macaca fascicularis , Male , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Prostatic Neoplasms/pathology , Transfection , Xenograft Model Antitumor Assays
2.
J Clin Oncol ; 29(18): 2493-8, 2011 Jun 20.
Article in English | MEDLINE | ID: mdl-21576633

ABSTRACT

PURPOSE: Blinatumomab, a bispecific single-chain antibody targeting the CD19 antigen, is a member of a novel class of antibodies that redirect T cells for selective lysis of tumor cells. In acute lymphoblastic leukemia (ALL), persistence or relapse of minimal residual disease (MRD) after chemotherapy indicates resistance to chemotherapy and results in hematologic relapse. A phase II clinical study was conducted to determine the efficacy of blinatumomab in MRD-positive B-lineage ALL. PATIENTS AND METHODS: Patients with MRD persistence or relapse after induction and consolidation therapy were included. MRD was assessed by quantitative reverse transcriptase polymerase chain reaction for either rearrangements of immunoglobulin or T-cell receptor genes, or specific genetic aberrations. Blinatumomab was administered as a 4-week continuous intravenous infusion at a dose of 15 µg/m2/24 hours. RESULTS: Twenty-one patients were treated, of whom 16 patients became MRD negative. One patient was not evaluable due to a grade 3 adverse event leading to treatment discontinuation. Among the 16 responders, 12 patients had been molecularly refractory to previous chemotherapy. Probability for relapse-free survival is 78% at a median follow-up of 405 days. The most frequent grade 3 and 4 adverse event was lymphopenia, which was completely reversible like most other adverse events. CONCLUSION: Blinatumomab is an efficacious and well-tolerated treatment in patients with MRD-positive B-lineage ALL after intensive chemotherapy. T cells engaged by blinatumomab seem capable of eradicating chemotherapy-resistant tumor cells that otherwise cause clinical relapse.


Subject(s)
Antibodies, Bispecific/therapeutic use , Drug Delivery Systems , Immunotherapy/methods , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , T-Lymphocytes, Cytotoxic/drug effects , Adult , Agammaglobulinemia/chemically induced , Aged , Aged, 80 and over , Antibodies, Bispecific/adverse effects , Antibodies, Bispecific/pharmacokinetics , Antibody Specificity , Antigens, CD19/immunology , Antigens, Neoplasm/immunology , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Bone Marrow/pathology , CD3 Complex/immunology , Cell Lineage , Combined Modality Therapy , Disease-Free Survival , Drug Resistance, Neoplasm , Female , Humans , Kaplan-Meier Estimate , Lymphocyte Activation , Lymphopenia/chemically induced , Male , Middle Aged , Molecular Targeted Therapy , Neoplasm, Residual , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Remission Induction , T-Cell Antigen Receptor Specificity/drug effects , T-Cell Antigen Receptor Specificity/immunology , T-Lymphocytes, Cytotoxic/immunology , Young Adult
3.
Proc Natl Acad Sci U S A ; 107(28): 12605-10, 2010 Jul 13.
Article in English | MEDLINE | ID: mdl-20616015

ABSTRACT

Epidermal growth factor receptor (EGFR)-specific monoclonal antibodies predominantly inhibit colorectal cancer (CRC) growth by interfering with receptor signaling. Recent analyses have shown that patients with CRC with mutated KRAS and BRAF oncogenes do not profit from treatment with such antibodies. Here we have used the binding domains of cetuximab and pantitumumab for constructing T cell-engaging BiTE antibodies. Both EGFR-specific BiTE antibodies mediated potent redirected lysis of KRAS- and BRAF-mutated CRC lines by human T cells at subpicomolar concentrations. The cetuximab-based BiTE antibody also prevented at very low doses growth of tumors from KRAS- and BRAF-mutated human CRC xenografts, whereas cetuximab was not effective. In nonhuman primates, no significant adverse events were observed during treatment for 3 wk at BiTE serum concentrations inducing, within 1 d, complete lysis of EGFR-overexpressing cancer cells. EGFR-specific BiTE antibodies may have potential to treat CRC that does not respond to conventional antibodies.


Subject(s)
Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , ErbB Receptors/antagonists & inhibitors , Antibodies, Monoclonal , Antibodies, Monoclonal, Humanized , Cetuximab , Colorectal Neoplasms/pathology , ErbB Receptors/genetics , ErbB Receptors/metabolism , Genes, ras/drug effects , Humans , Mutation/drug effects , Neoplasms/genetics , Proto-Oncogene Proteins B-raf , Risk Factors
4.
J Immunother ; 32(5): 452-64, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19609237

ABSTRACT

muS110 is a BiTE antibody bispecific for murine EpCAM (CD326) and murine CD3. MT110, its human-specific analog, is in a clinical phase 1 trial for treatment of patients with adenocarcinoma of the lung or gastrointestinal tract. Recent studies have shown a therapeutic window for muS110, have explored single-dose toxicity of muS110, and have found that a 1-week low-dose treatment dramatically increased the tolerability of mice to very high doses of muS110 (Cancer Immunol. Immunother. 2009;58:95-109). Here we analyzed the impact of long-term, high-dose treatment of mice with muS110 on antitumor activity and functionality of T cells. After an initial self-limiting cytokine release, the 1-week adaptation period effectively blunted further cytokine production in response to a subsequent high-dose treatment with muS110. The much-increased tolerability of mice adapted to muS110 was not because of anergy of T cells. T cells isolated from chronically muS110-treated mice fully retained their cytotoxic potential, proliferative capacity, and responsiveness to stimulation by either muS110 or anti-CD3/anti-CD28/interleukin-2 when compared with T cells from control mice. Unimpaired T-cell performance was also evident from the effective prevention of orthotopic 4T1 breast tumor outgrowth in mice treated long term with escalating doses of muS110. Finally, we show that muS110 and MT110 recognize orthologous epitopes on mouse and human EpCAM proteins, suggesting that the target-related safety profile of muS110 in mice may be predictive for MT110 in humans.


Subject(s)
Antibodies, Bispecific/administration & dosage , Antigens, Neoplasm/immunology , CD3 Complex/immunology , Cell Adhesion Molecules/immunology , Immunotherapy , Mammary Neoplasms, Experimental/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Bispecific/adverse effects , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , CD3 Complex/genetics , CD3 Complex/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Proliferation/drug effects , Clonal Anergy/drug effects , Cytokines/metabolism , Epithelial Cell Adhesion Molecule , Female , Humans , Immunologic Factors/administration & dosage , Immunologic Factors/adverse effects , Lymphocyte Activation/drug effects , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/therapy , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Protein Engineering , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Time Factors
5.
J Immunother ; 32(4): 341-52, 2009 May.
Article in English | MEDLINE | ID: mdl-19342971

ABSTRACT

Carcinoembryonic antigen (CEA, CD66e) is a well-characterized tumor-associated antigen that is frequently overexpressed in tumors. Phospholipases release CEA from tumor cells resulting in high circulating serum levels of soluble CEA (sCEA) that has been validated as marker for progression of colorectal, breast, and lung cancers. sCEA also acts as a competitive inhibitor for anticancer strategies targeting membrane-bound CEA. As a novel therapeutic approach for treatment of tumors expressing CEA on their cell surface, we constructed a series of bispecific single-chain antibodies (bscAb) combining various single-chain variable fragments recognizing human CEA with a deimmunized single-chain variable fragments recognizing human CD3. CEA/CD3-bscAbs redirected human T cells to lyse CEA-expressing tumor cells in vitro and in vivo. Efficient tumor cell lysis was achieved in vitro at bscAb concentrations from 1 pg/mL (19 fM) to 8.9 pg/mL with preactivated CD8 T cells, and 200 to 500 pg/mL with unstimulated peripheral blood mononuclear cell. The cytotoxic activity of a subset of CEA/CD3-bscAbs was not competitively inhibited by sCEA at concentrations that exceeded levels found in the serum of most cancer patients. Treatment with CEA/CD3-bscAbs prevented the growth of human colorectal cancer lines in a severe combined immunodeficiency mouse model modified to show human T cell killing of tumors. A murine surrogate CEA/CD3-bscAb capable of recruiting murine T cells for redirected tumor lysis in immunocompetent mice prevented the growth of lung tumors expressing human CEA. Together, our results reveal a unique opportunity for targeting cytotoxic T cells toward CEA-expressing tumors without being competitively inhibited by sCEA and establish CEA/CD3-bscAb as a promising and potent therapeutic approach.


Subject(s)
Antibodies, Bispecific/therapeutic use , CD3 Complex/immunology , CD8-Positive T-Lymphocytes/immunology , Carcinoembryonic Antigen/immunology , Colorectal Neoplasms/therapy , Recombinant Fusion Proteins/therapeutic use , Animals , Antibodies, Bispecific/immunology , CD8-Positive T-Lymphocytes/metabolism , CHO Cells , Carcinoembryonic Antigen/blood , Cricetinae , Cricetulus , Humans , Immunotherapy , Mice , Mice, SCID , Recombinant Fusion Proteins/immunology , Tetrahydrofolate Dehydrogenase/genetics , Tetrahydrofolate Dehydrogenase/immunology
6.
Leuk Res ; 33(3): 465-73, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18835037

ABSTRACT

We have compared the cytotoxic activity of rituximab with that of blinatumomab (MT103/MEDI-538), a single-chain CD19-/CD3-bispecific antibody engaging human T cells. Blinatumomab consistently led to a higher degree of lysis of human lymphoma lines than rituximab, and was active at much lower concentration. The cytotoxicity mediated by blinatumomab and rituximab both caused a potent activation of pro-caspases 3 and 7 in target cells, a key event in induction of granzyme-mediated apoptotic cell death. Combination of rituximab with blinatumomab was found to greatly enhance the activity of rituximab, in particular at low effector-to-target cell ratios and at low antibody concentration.


Subject(s)
Antibodies, Bispecific/pharmacology , Antibodies, Monoclonal/pharmacology , Cytotoxicity, Immunologic/drug effects , Antibodies, Monoclonal, Murine-Derived , Antigens, CD19/immunology , Antineoplastic Combined Chemotherapy Protocols , CD3 Complex/immunology , Caspase 3/metabolism , Caspase 7/metabolism , Drug Synergism , Granzymes , Humans , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/pathology , Rituximab , Tumor Cells, Cultured
7.
Cancer Res ; 68(1): 143-51, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18172306

ABSTRACT

EpCAM (CD326) is one of the most frequently and highly expressed tumor-associated antigens known and recently has also been found on cancer stem cells derived from human breast, colon, prostate, and pancreas tumors. However, like many other tumor-associated antigens used for antibody-based immunotherapeutic approaches, EpCAM is expressed on normal tissues including epithelia of pancreas, colon, lung, bile ducts, and breast. To assess the therapeutic window of an EpCAM/CD3-bispecific single-chain antibody construct of the bispecific T-cell engager (BiTE) class, we constructed murine surrogate of MT110 (muS110) from single-chain antibodies specific for murine EpCAM and CD3 antigens. Immunhistochemical analysis showed that, with minor differences, the expression of EpCAM protein on a large variety of tissues from man and mouse was similar with respect to distribution and level. MuS110 exhibited significant antitumor activity at as low as 5 microg/kg in both syngeneic 4T1 orthotopic breast cancer and CT-26 lung cancer mouse models. Dosing of muS110 for several weeks up to 400 microg/kg by intraanimal dose escalation was still tolerated, indicating existence of a significant therapeutic window for an EpCAM-specific BiTE antibody in mice. MuS110 was found to have similar in vitro characteristics and in vivo antitumor activity as MT110, a human EpCAM/human CD3-bispecific BiTE antibody that currently is in formal preclinical development.


Subject(s)
Antibodies, Bispecific/pharmacokinetics , Antibodies, Monoclonal/pharmacokinetics , Antigens, Neoplasm/immunology , CD3 Complex/immunology , Cell Adhesion Molecules/immunology , Neoplasms/drug therapy , Animals , Antibodies, Bispecific/immunology , Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/analysis , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , CD3 Complex/analysis , Cell Adhesion Molecules/analysis , Cricetinae , Cytotoxicity, Immunologic , Epithelial Cell Adhesion Molecule , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Mice , Neoplasms/immunology , Single-Chain Antibodies , Tissue Distribution
8.
Cancer Res ; 67(8): 3927-35, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17440108

ABSTRACT

The EphA2 receptor tyrosine kinase is frequently overexpressed and functionally altered in malignant cells and thus provides opportunities for selective targeting of tumor cells. We describe here the development of a novel, bispecific single-chain antibody (bscAb) referred to as bscEphA2xCD3. This molecule simultaneously targets EphA2 on tumor cells and the T-cell receptor/CD3 complex on T cells and possesses structural and functional characteristics of the recently developed BiTE technology. An EphA2-specific single-chain antibody was selected for recognition of an epitope that is preferentially exposed on malignant cells based on the concept of epitope exclusion; this was fused to a CD3-specific single-chain antibody to generate bscEphA2xCD3. The resultant bscAb redirected unstimulated human T cells to lyse EphA2-expressing tumor cells both in vitro and in vivo. In separate experiments, efficient tumor cell lysis was achieved in vitro at drug concentrations

Subject(s)
Antibodies, Bispecific/pharmacology , CD3 Complex/immunology , Immunization, Passive/methods , Neoplasms/therapy , Receptor, EphA2/immunology , Animals , Antibodies, Bispecific/immunology , Antibody Specificity , CHO Cells , Cell Growth Processes/immunology , Cell Line, Tumor , Cricetinae , Cricetulus , Epitopes/immunology , Humans , Intercellular Junctions/immunology , Lymphocyte Activation , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms/immunology , Neoplasms/pathology , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
9.
Mol Immunol ; 43(8): 1129-43, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16139892

ABSTRACT

We have developed a novel single-chain Ep-CAM-/CD3-bispecific single-chain antibody construct designated MT110. MT110 redirected unstimulated human peripheral T cells to induce the specific lysis of every Ep-CAM-expressing tumor cell line tested. MT110 induced a costimulation independent polyclonal activation of CD4- and CD8-positive T cells as seen by de novo expression of CD69 and CD25, and secretion of interferon gamma, tumor necrosis factor alpha, and interleukins 2, 4 and 10. CD8-positive T cells made the major contribution to redirected tumor cell lysis by MT110. With a delay, CD4-positive cells could also contribute presumably as consequence of a dramatic upregulation of granzyme B expression. MT110 was highly efficacious in a NOD/SCID mouse model with subcutaneously growing SW480 human colon cancer cells. Five daily doses of 1 microg MT110 on days 0-4 completely prevented tumor outgrowth in all mice treated. The bispecific antibody construct also led to a durable eradication of established tumors in all mice treated with 1 microg doses of MT110 on days 8-12 after tumor inoculation. Finally, MT110 could eradicate patient-derived metastatic ovarian cancer tissue growing under the skin of NOD/SCID mice. MT110 appears as an attractive bispecific antibody candidate for treatment of human Ep-CAM-overexpressing carcinomas.


Subject(s)
Antibodies, Bispecific/immunology , Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Neoplasms/drug therapy , Animals , Antibody Specificity , Antigens, CD19/metabolism , CD3 Complex/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cytotoxicity, Immunologic , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Kinetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms/pathology , Recombinant Proteins/isolation & purification , Single-Chain Antibodies
10.
Cancer Res ; 65(7): 2882-9, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15805290

ABSTRACT

Bispecific T-cell engager (BiTE) are a class of bispecific single-chain antibodies that can very effectively redirect cytotoxic T cells for killing of tumor target cells. Here, we have assessed the in vivo efficacy of one representative, called bscEp-CAMxCD3, with specificity for tumors overexpressing epithelial cell adhesion molecule (Ep-CAM) in human xenograft models. Cells of the human colon carcinoma line SW480 were mixed at a 1:1 ratio with unstimulated human peripheral mononuclear cells, s.c. injected in nonobese diabetes/severe combined immunodeficiency (NOD/SCID) mice, and animals were treated with bscEp-CAMxCD3. Five daily i.v. injections of as little as 100 ng per mouse of bscEp-CAMxCD3 completely prevented tumor outgrowth when treatment was started at the day of tumor cell inoculation. BscEp-CAMxCD3 was also efficacious when administered up to 8 days after xenograft injection. Established tumors could be eradicated in all animals by five 10 microg doses given between days 8 and 12 after tumor cell inoculation. To test the efficacy of bscEp-CAMxCD3 in a more physiologic model, pieces of primary metastatic tumor tissue from ovarian cancer patients were implanted in NOD/SCID mice. Partial tumor engraftment and growth was observed with four of six patient samples. Treatment of established tumors with daily 5 microg doses led to a significant reduction and, in some cases, eradication of human tumor tissue. These effects obviously relied on the tumor-resident T cells reactivated by bscEp-CAMxCD3. Our data show that the class of single-chain bispecific antibodies has very high antitumor efficacy in vivo and can use previously unstimulated T cells at low effector-to-target ratios.


Subject(s)
Antibodies, Bispecific/pharmacology , Antigens, Neoplasm/immunology , CD3 Complex/immunology , Cell Adhesion Molecules/immunology , Colonic Neoplasms/therapy , Ovarian Neoplasms/therapy , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/therapy , Animals , Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacokinetics , Antibody Specificity , Cell Line, Tumor , Colonic Neoplasms/immunology , Epithelial Cell Adhesion Molecule , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/immunology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL