Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Cancer Immunol Res ; 12(7): 921-943, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38683145

ABSTRACT

The prognosis of patients with acute myeloid leukemia (AML) is limited, especially for elderly or unfit patients not eligible for hematopoietic stem cell (HSC) transplantation. The disease is driven by leukemic stem cells (LSCs), which are characterized by clonal heterogeneity and resistance to conventional therapy. These cells are therefore believed to be a major cause of progression and relapse. We designed MP0533, a multispecific CD3-engaging designed ankyrin repeat protein (DARPin) that can simultaneously bind to three antigens on AML cells (CD33, CD123, and CD70), aiming to enable avidity-driven T cell-mediated killing of AML cells coexpressing at least two of the antigens. In vitro, MP0533 induced selective T cell-mediated killing of AML cell lines, as well as patient-derived AML blasts and LSCs, expressing two or more target antigens, while sparing healthy HSCs, blood, and endothelial cells. The higher selectivity also resulted in markedly lower levels of cytokine release in normal human blood compared to single antigen-targeting T-cell engagers. In xenograft AML mice models, MP0533 induced tumor-localized T-cell activation and cytokine release, leading to complete eradication of the tumors while having no systemic adverse effects. These studies show that the multispecific-targeting strategy used with MP0533 holds promise for improved selectivity toward LSCs and efficacy against clonal heterogeneity, potentially bringing a new therapeutic option to this group of patients with a high unmet need. MP0533 is currently being evaluated in a dose-escalation phase 1 study in patients with relapsed or refractory AML (NCT05673057).


Subject(s)
Leukemia, Myeloid, Acute , Neoplastic Stem Cells , T-Lymphocytes , Humans , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/therapy , Leukemia, Myeloid, Acute/pathology , Animals , Mice , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Interleukin-3 Receptor alpha Subunit/immunology , Interleukin-3 Receptor alpha Subunit/metabolism , Xenograft Model Antitumor Assays , Sialic Acid Binding Ig-like Lectin 3/metabolism , Sialic Acid Binding Ig-like Lectin 3/immunology , CD3 Complex/immunology , Cell Line, Tumor , Cytotoxicity, Immunologic
2.
MAbs ; 8(1): 141-9, 2016.
Article in English | MEDLINE | ID: mdl-26390837

ABSTRACT

Biologic treatment options such as tumor necrosis factor (TNF) inhibitors have revolutionized the treatment of inflammatory diseases, including rheumatoid arthritis. Recent data suggest, however, that full and long-lasting responses to TNF inhibitors are limited because of the activation of the pro-inflammatory TH17/interleukin (IL)-17 pathway in patients. Therefore, dual TNF/IL-17A inhibition is an attractive avenue to achieve superior efficacy levels in such diseases. Based on the marketed anti-TNF antibody adalimumab, we generated the bispecific TNF/IL-17A-binding FynomAb COVA322. FynomAbs are fusion proteins of an antibody and a Fyn SH3-derived binding protein. COVA322 was characterized in detail and showed a remarkable ability to inhibit TNF and IL-17A in vitro and in vivo. Through its unique mode-of-action of inhibiting simultaneously TNF and the IL-17A homodimer, COVA322 represents a promising drug candidate for the treatment of inflammatory diseases. COVA322 is currently being tested in a Phase 1b/2a study in psoriasis ( ClinicalTrials.gov Identifier: NCT02243787).


Subject(s)
Interleukin-17/antagonists & inhibitors , Psoriasis/drug therapy , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Antibodies, Bispecific/administration & dosage , Antibodies, Bispecific/immunology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Female , Humans , Interleukin-17/immunology , Male , Mice , Psoriasis/immunology , Tumor Necrosis Factor-alpha/immunology
3.
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
4.
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
5.
Immunobiology ; 214(6): 441-53, 2009.
Article in English | MEDLINE | ID: mdl-19157637

ABSTRACT

MT110 is an EpCAM/CD3-bispecific antibody construct in clinical development for the treatment of patients with adenocarcinoma expressing EpCAM (CD326). Like other members of this antibody class, MT110 can engage resting, polyclonal CD8(+) and CD4(+) T cells for highly potent redirected lysis of target cells. Here we further explored the mechanism of this action. Complete lysis of EpCAM(+) Kato III gastric cancer cells by previously unstimulated T cells was achieved within 48 h. During this period, a high percentage of CD4(+) and CD8(+) T cells became activated and increased expression of granzyme B. This apparently boosted the capacity for serial target cell lysis as studied at very low effector-to-target ratios. Elimination of cancer cells by MT110-redirected T cells involved membrane damage as was evident from nuclear uptake of propidium iodide and release of the cytosolic enzyme adenylate kinase. Redirected T cells also potently triggered programmed cell death in cancer cells as was evident by membrane blebbing, activation of procaspases 3 and 7, fragmentation of nuclear DNA and cleavage of the caspase substrate poly (ADP ribose) polymerase. Chelation of extracellular calcium fully protected cancer cells from lysis by MT110-redirected T cells, while the pan-caspase inhibitor Z-VAD-FMK blocked activation of procaspases, cleavage of poly (ADP ribose) polymerase and fragmentation of nuclear DNA in cancer cells, but could not prevent nuclear uptake of propidium iodide. Soluble factors did not significantly contribute to cancer cell death. Our study shows that MT110 can efficiently gear up the potential of CD8(+) and CD4(+) T cells for serial lysis, and mediate kill of cancer cells predominantly through poreforming and pro-apoptotic components of cytotoxic T cell granules.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cytotoxicity, Immunologic/drug effects , Adenylyl Cyclases/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Apoptosis , CD3 Complex/immunology , CD3 Complex/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Caspase Inhibitors , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Collagen Type XI/antagonists & inhibitors , Cytotoxicity, Immunologic/immunology , DNA Fragmentation/drug effects , Epithelial Cell Adhesion Molecule , Granzymes/genetics , Granzymes/immunology , Granzymes/metabolism , Humans , Lymphocyte Activation/drug effects , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Single-Chain Antibodies
6.
Cancer Immunol Immunother ; 58(1): 95-109, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18594818

ABSTRACT

MuS110 is a BiTE antibody bispecific for murine EpCAM (CD326) and murine CD3. A recent study has shown that microS110 has significant anti tumor activity at well-tolerated doses as low as 5 microg/kg in orthotopic breast and lung cancer models (Amann et al. in Cancer Res 68:143-151, 2008). Here, we have explored the safety profile of microS110 at higher doses. Escalation to 50 microg/kg microS110 caused in mice transient loss of body weight, and transient piloerection, hypomotility, hypothermia and diarrhoea. These clinical signs coincided with serum peaks of TNF-alpha, IL-6, IL-2, IFN-gamma and IL-4, and an increase of surface markers for T cell activation. Because activation of T cells in response to BiTE antibodies is typically dependent on target cells, we analyzed mouse blood for the presence of EpCAM(+) cells. Various mouse strains presented with a subpopulation of 2-3% EpCAM(+) blood cells, mostly B and T lymphocytes, which was not detected in human blood samples. In vitro experiments in which the number of EpCAM(+) cells in blood samples was either reduced or increased suggested that both T cell activation and cytokine release in response to microS110 was dependent on the number of target-expressing cells. In support for a role of EpCAM(+) lymphocytes in the observed side effects, reduction of EpCAM(+) blood cells in mice via a low-dose pre treatment with microS110 dramatically increased the tolerability of animals up to at least 500 microg/kg of the BiTE antibody. This high tolerability to microS110 occurred in the presence of non-compromised T cells. No damage to EpCAM(+) epithelial tissues was evident from histopathological examination of animals daily injected with 100 microg/kg microS110 for 28 days. In summary, these observations suggest that side effects of microS110 in mice were largely caused by an acute T cell activation that was triggered by a subpopulation of EpCAM(+) lymphocytes. Because humans have extremely low numbers of EpCAM(+) cells in blood, this toxicity of an EpCAM-specific BiTE may be specific for mice.


Subject(s)
Antigens, Neoplasm/blood , Cell Adhesion Molecules/blood , Epitopes, T-Lymphocyte/blood , Lymphocyte Subsets , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , CHO Cells , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Cricetinae , Cricetulus , Epithelial Cell Adhesion Molecule , Female , Humans , Lymphocyte Activation , Lymphocyte Subsets/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Recombinant Proteins/genetics , Species Specificity
7.
Breast Cancer Res Treat ; 117(3): 471-81, 2009 Oct.
Article in English | MEDLINE | ID: mdl-18819003

ABSTRACT

In the present study, the efficacy of a new drug, i.e. the bispecific single-chain antibody MT110 targeting the epithelial antigen EpCAM and the T-cell antigen CD3 was tested ex vivo in malignant pleural effusions (MPEs). EpCAM+ epithelial cells were found in 78% of the MPEs (n = 18). Ex vivo treatment of seven MPEs resulted in a dose-dependent specific lysis of 37 +/- 27% (+/- SD) EpCAM+ cells with 10 ng/ml (P = 0.03) and 57 +/- 29.5% EpCAM+ cells with 1,000 ng/ml MT110 (P = 0.016) after 72 h. As a prerequisite for redirected lysis, stimulation of the autologous CD4+ and CD8+ cells in MPE by 1,000 ng/ml MT110 resulted in 21 +/- 17% CD4+/CD25+ and 29.4 +/- 22% CD8+/CD25+ cells (P = 0.016, respectively) after 72 h. This was confirmed by a 22-fold release of TNF-alpha and 230-fold release of IFN-gamma (1,000 ng/ml, 48 h, P = 0.03, respectively). Thus, relapsed breast cancer patients resistant to standard treatment might benefit from targeted therapy using MT110.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antigens, Neoplasm/immunology , Breast Neoplasms/immunology , Cell Adhesion Molecules/immunology , Lymphocyte Activation/drug effects , Pleural Effusion, Malignant/immunology , T-Lymphocytes/drug effects , Antibodies, Bispecific/drug effects , Antibodies, Bispecific/immunology , CD3 Complex/immunology , Cytokines/biosynthesis , Epithelial Cell Adhesion Molecule , Female , Flow Cytometry , Humans , Immunohistochemistry , Lymphocyte Activation/immunology , Single-Chain Antibodies , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology
8.
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
9.
J Immunother ; 30(8): 798-807, 2007.
Article in English | MEDLINE | ID: mdl-18049331

ABSTRACT

Bispecific antibodies have been extensively studied in vitro and in vivo for their use in redirected tumor cell lysis. A particular challenge of bispecific antibody constructs that recognize the invariant CD3 signaling complex is a controlled polyclonal activation of T cells that, ideally, is exquisitely dependent on the presence of target cells. Otherwise, overt production of inflammatory cytokines and secondary reactions may occur as side effects, as can be observed with constitutively T-cell activating monoclonal antibodies to CD3 or CD28, and with bispecific antibodies bearing Fc gamma portions. Here we analyzed 2 distinct bispecific single-chain antibody constructs of the BiTE class, called MT110 and MT103 (or MEDI-538), for conditional T-cell activation. In the presence of target-expressing cell lines, low picomolar concentrations of the BiTE molecules were sufficient to stimulate a high percentage of peripheral human T cells to express cytokines and surface activation markers, enter into cell cycle, and induce redirected lysis of target cells. However, in the absence of target cells, the 2 BiTE molecules even at high concentrations did not detectably activate T cells. Our data show that T cell activation by monomeric forms of MT110 and MT103 is highly conditional in that it is strictly dependent on the presence of cells expressing the proper target antigen. BiTE molecules therefore qualify for a highly controlled polyclonal T-cell therapy of cancer.


Subject(s)
Antibodies, Bispecific/immunology , Antigens, Neoplasm/immunology , Cell Adhesion Molecules/immunology , Immunoglobulin Fragments/immunology , T-Lymphocytes/immunology , Amino Acid Substitution , Animals , Antibodies, Bispecific/genetics , Antigens, CD/metabolism , Antigens, CD19/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Antigens, Neoplasm/genetics , CD3 Complex/metabolism , CHO Cells , Cell Adhesion Molecules/genetics , Cell Proliferation , Cricetinae , Cricetulus , Cytokines/metabolism , Epithelial Cell Adhesion Molecule , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Humans , Immunoglobulin Fragments/genetics , Interleukin-2 Receptor alpha Subunit/metabolism , Lectins, C-Type , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lymphocyte Activation/immunology , Macaca fascicularis , Mice , Muromonab-CD3/metabolism , Recombinant Proteins/immunology , T-Lymphocytes/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Transfection
10.
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
11.
Cancer Immunol Immunother ; 56(4): 459-68, 2007 Apr.
Article in English | MEDLINE | ID: mdl-16937114

ABSTRACT

An important mode of action shared by human IgG1 antibody therapies is antibody-dependent cellular cytotoxicity (ADCC). ADCC relies on the interaction of the antibody's Fc portion with Fc-gama receptors (FcgammaR) on immune effector cells. The anti-tumor activity of human IgG1 antibodies is frequently assessed in mouse models. Binding of human IgG1 to murine FcgammaRs is however of reduced affinity. We here show that ADCC of adecatumumab (MT201), a fully human IgG1 antibody specific for epithelial cell adhesion molecule (EpCAM/CD326), is drastically lower if human peripheral blood mononuclear cells are replaced by murine splenocytes as effector cells. When the variable domains of adecatumumab were genetically fused to a murine IgG2a backbone (yielding mu-adecatumumab), ADCC with murine effector cells was much improved, but at the same time significantly reduced with human effector cells. The serum half-lives of adecatumumab and mu-adecatumumab were determined in mice and dosing schedules established that gave similar serum trough levels during a 4-week antibody treatment. The anti-tumor activities of adecatumumab and mu-adecatumumab were then compared side-by-side in a lung metastasis mouse model established with a syngeneic B16 melanoma line expressing human EpCAM at physiologically relevant levels. Treatment of mice with mu-adecatumumab led to an almost complete prevention of lung metastases, while the human version of the antibody was much less active. This shows that adecatumumab has high anti-tumor activity when tested in a form that is better compatible with the species' immune system. Moreover, our data suggest to routinely compare in mouse models human IgG1 and murine IgG2a versions of antibodies to properly assess the contribution of ADCC to overall anti-tumor activity.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antigens, CD/immunology , Antineoplastic Agents/pharmacology , Cell Adhesion Molecules/pharmacology , Disease Models, Animal , Lung Neoplasms/drug therapy , Receptors, IgG/immunology , Animals , Antibodies, Monoclonal, Humanized , Antibody-Dependent Cell Cytotoxicity , Antigens, Neoplasm/immunology , CHO Cells , Cell Adhesion Molecules/immunology , Cricetinae , Cricetulus , Epithelial Cell Adhesion Molecule , Humans , Lung Neoplasms/immunology , Mice , Neoplasm Metastasis/drug therapy , Species Specificity , Transplantation, Isogeneic
12.
Cancer Immunol Immunother ; 55(5): 503-14, 2006 May.
Article in English | MEDLINE | ID: mdl-16032400

ABSTRACT

BscCD19xCD3 is a bispecific single-chain antibody construct with exceptional cytotoxic potency in vitro and in vivo. Here, we have investigated the biological activity of bscCD19xCD3 in chimpanzee, the only animal species identified in which bscCD19xCD3 showed bispecific binding, redirected B-cell lysis and cytokine production comparable to human cells. Pharmacokinetic analysis following 2-h intravenous infusion of 0.06, 0.1 or 0.12 mug/kg of bscCD19xCD3 as part of a dose escalation study in a single female chimpanzee revealed a half-life of approximately 2 h and elimination of the bispecific antibody from circulation within approximately 8 h after the end of infusion. This short exposure to bscCD19xCD3 elicited a transient increase in serum levels of IFNgamma, IL-6, IL-2, soluble CD25, and transiently upregulated expression of CD69 and MHC class II on CD8-positive cells. Cytokine release and upregulation of T-cell activation markers were not observed with vehicle controls. A multiple-dose study using 5 weekly doses of 0.1 mug/kg in two animals also showed transient cytokine release and an activation of peripheral T cells with a first-dose effect, accompanied by a transient lymphopenia. While oscillations of T-cell counts were relatively even during repeated treatments, the amplitudes of peripheral B cells declined with every infusion, which was not observed in a vehicle control animal. Our data show that bscCD19xCD3 can be safely administered to chimpanzees at dose levels that cause fully reversible T-cell activation and, despite a very short exposure time, cumulative loss of peripheral B lymphocytes. A clinical trial testing prolonged administration of bscCD19xCD3 (MT103) for improving efficacy is currently ongoing.


Subject(s)
Antibodies, Bispecific/pharmacokinetics , B-Lymphocytes/drug effects , Lymphocyte Activation/drug effects , Lymphocyte Depletion , T-Lymphocytes/drug effects , Animals , Antigens, CD19/immunology , B-Lymphocytes/immunology , CD3 Complex/immunology , Cell Line, Tumor , Cells, Cultured , Cytotoxicity Tests, Immunologic , Female , Flow Cytometry , Humans , Immunotherapy , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Lymphocyte Activation/immunology , Male , Pan troglodytes , T-Lymphocytes/immunology
13.
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
14.
Cancer Immunol Immunother ; 55(7): 785-96, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16187083

ABSTRACT

Bispecific single-chain antibody constructs specific for human CD3 have been extensively studied for antitumor activity in human xenograft models using severe combined immunodeficient mice supplemented with human T cells. High efficacy at low effector-to-target ratios, independence of T cell costimuli and a potent activation of previously unstimulated polyclonal T cells were identified as hallmarks of this class of bispecific antibodies. Here we studied a bispecific single-chain antibody construct (referred to as 'bispecific T cell engager', BiTE) in an immunocompetent mouse model. This was possible by the use of a murine CD3-specific BiTE, and a syngeneic melanoma cell line (B16F10) expressing the human Ep-CAM target. The murine CD3-specific BiTE, called 2C11x4-7 prevented in a dose-dependent fashion the outgrowth of subcutaneously growing B16/Ep-CAM tumors with daily i.v. injections of 5 or 50 microg BiTE which was most effective. Treatment with 2C11x4-7 was effective even when it was started 10 days after tumor cell inoculation but delayed treatments showed a reduction in the number of cured animals. 2C11x4-7 was also highly active in a lung tumor colony model. When treatment was started on the day of intravenous tumor cell injection, seven out of eight animals stayed free of lung tumors, and three out of eight animals when treatment was started on day 5. Our study shows that BiTEs also have a high antitumor activity in immunocompetent mice and that there is no obvious need for costimulation of T cells by secondary agents.


Subject(s)
Antibodies, Bispecific/therapeutic use , Antigens, Neoplasm/immunology , CD3 Complex/immunology , Cell Adhesion Molecules/immunology , Immunotherapy , Melanoma, Experimental/therapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Animals , Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacokinetics , Antibody Specificity , Cell Line, Tumor/transplantation , Dose-Response Relationship, Immunologic , Drug Screening Assays, Antitumor , Epithelial Cell Adhesion Molecule , Humans , Immunocompetence , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Recombinant Fusion Proteins/immunology , Subcutaneous Tissue , Xenograft Model Antitumor Assays
15.
Vaccine ; 24(12): 2065-70, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16337719

ABSTRACT

A Phase I trial was performed to investigate the safety and immunogenicity of the third generation smallpox vaccine MVA-BN (IMVAMUNE), a highly attenuated clone derived from the Modified Vaccinia Virus Ankara strain 571, in naive and pre-immunized subjects. A total of 86 healthy subjects received the vaccine in five groups using different doses and routes of administration. All 38 subjects seroconverted in the groups receiving the highest dose (10(8) TCID50). All vaccinations were well tolerated with mainly mild or moderate pain at the injection site being the most frequent symptom. The results indicate that MVA-BN has the potential to be developed as an efficient and safe alternative to the conventional smallpox vaccines such as Lister-Elstree or Dryvax. Unique attributes render it a promising candidate for prophylactic mass immunization, even in subjects for whom conventional smallpox vaccines are contraindicated.


Subject(s)
Immunization, Secondary , Smallpox Vaccine/administration & dosage , Smallpox/prevention & control , Vaccinia virus/immunology , Adult , Double-Blind Method , Humans , Male , Middle Aged , Safety , Smallpox Vaccine/adverse effects , Smallpox Vaccine/genetics , Smallpox Vaccine/immunology
16.
Drug Discov Today ; 10(18): 1237-44, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16213416

ABSTRACT

Bispecific T-cell engager molecules (BiTEs) constitute a class of bispecific single-chain antibodies for the polyclonal activation and redirection of cytotoxic T cells against pathogenic target cells. BiTEs combine a unique set of properties that have not yet been reported for any other kind of bispecific antibody construct, namely extraordinary potency and efficacy against target cells at low T-cell numbers without the need for T-cell co-stimulation. Here we review novel insights into the mechanism of BiTE action, which help to explain the unique features of BiTEs, as well as data from various animal models demonstrating the outstanding therapeutic potential of BiTEs for the treatment of malignant diseases.


Subject(s)
Antibodies, Bispecific/therapeutic use , Neoplasms/therapy , T-Lymphocytes/immunology , Animals , Antibodies, Bispecific/pharmacology , Cytotoxicity, Immunologic , Humans , Lymphocyte Activation , Neoplasms/immunology
17.
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
18.
J Gen Virol ; 84(Pt 8): 2145-2151, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12867646

ABSTRACT

After passive transfer of measles virus (MV)-specific antibodies, vaccine-induced seroconversion and subsequent protection is inhibited in cotton rats (Sigmodon hispidus). In this system, an attenuated, recombinant vesicular stomatitis virus expressing the MV haemagglutinin (VSV-H) was found previously to induce neutralizing antibodies and protection against MV challenge after intranasal (i.n.) immunization. Here it is demonstrated that, after i.n. immunization, VSV-H is found in both lung and brain tissue in the absence of clinical signs. Intratracheal inoculation, which does not lead to infection of the brain, proved that immunization via the lung mucosa is sufficient to protect. To reduce or eliminate infection of the brain after i.n. inoculation, stepwise-attenuated VSV-H mutants with truncated cytoplasmic tails of the G protein were tested in cotton rats. A mutant with 9 aa in the G cytoplasmic tail was found at much lower levels in the brain and was protective in the absence or presence of MV-specific antibodies. A more attenuated mutant containing only 1 aa in its tail was not found in brain tissue after inoculation, but it still induced protective antibody to measles in the absence of MV-specific antibody. However, its ability to induce MV-neutralizing antibodies in the presence of passively transferred MV-specific antibodies and its protective capacity was abolished unless higher-dose immunizations were used. This study demonstrates that a lower degree of attenuation is required to be able to immunize in the presence of MV-specific antibodies.


Subject(s)
Administration, Intranasal , Antibodies, Viral/blood , Hemagglutinins, Viral/genetics , Measles Vaccine/administration & dosage , Measles/prevention & control , Vesicular stomatitis Indiana virus/pathogenicity , Animals , Antibodies, Viral/immunology , Brain/virology , Genetic Vectors , Hemagglutinins, Viral/metabolism , Immunization , Immunization, Passive , Measles/immunology , Measles Vaccine/immunology , Measles virus/immunology , Mutation , Recombination, Genetic , Sigmodontinae , Vaccines, Attenuated , Vesicular stomatitis Indiana virus/genetics , Vesicular stomatitis Indiana virus/immunology
19.
J Immunol ; 170(8): 4397-402, 2003 Apr 15.
Article in English | MEDLINE | ID: mdl-12682277

ABSTRACT

We have recently demonstrated that a recombinant single-chain bispecific Ab construct, bscCD19xCD3, in vitro induces rapid B lymphoma-directed cytotoxicity at picomolar concentrations with unstimulated peripheral T cells. In this study, we show that treatment of nonobese diabetic SCID mice with submicrogram doses of bscCD19xCD3 could prevent growth of s.c. human B lymphoma xenografts and essentially cured animals when given at an early tumor stage. The effect was dose dependent, dependent on E:T ratio and the time between tumor inoculation and administration of bscCD19xCD3. No therapeutic effect was seen in the presence of human lymphocytes alone, a vehicle control, or with a bispecific single-chain construct of identical T cell-binding activity but different target specificity. In a leukemic nonobese diabetic SCID mouse model, treatment with bscCD19xCD3 prolonged survival of mice in a dose-dependent fashion. The human lymphocytes used as effector cells in both animal models did not express detectable T cell activation markers at the time of coinoculation with tumor cells. The bispecific Ab therefore showed an in vivo activity comparable to that observed in cell culture with respect to high potency and T cell costimulus independence. These properties make bscCD19xCD3 superior to previously investigated CD19 bispecific Ab-based therapies.


Subject(s)
Antibodies, Bispecific/pharmacology , Antigens, CD19/immunology , Antineoplastic Agents/pharmacology , CD3 Complex/immunology , Growth Inhibitors/pharmacology , Leukemia, B-Cell/prevention & control , Lymphoma, B-Cell/prevention & control , T-Lymphocyte Subsets/immunology , Transplantation, Heterologous , Animals , Antibodies, Bispecific/genetics , Antineoplastic Agents/chemical synthesis , Cells, Cultured , Graft Survival/genetics , Graft Survival/immunology , Growth Inhibitors/chemical synthesis , Humans , Injections, Intravenous , Injections, Subcutaneous , Leukemia, B-Cell/genetics , Leukemia, B-Cell/immunology , Leukemia, B-Cell/pathology , Lymphocyte Activation/genetics , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Skin Neoplasms/prevention & control , Time Factors , Transplantation, Heterologous/methods , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...