Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Oncoimmunology ; 7(9): e1472195, 2018.
Article in English | MEDLINE | ID: mdl-30228941

ABSTRACT

A number of agents designed for immunotherapy of Acute Myeloid Leukemia (AML) are in preclinical and early clinical development. Most of them target a single antigen on the surface of AML cells. Here we describe the development and key biological properties of a tri-specific agent, the dual-targeting triplebody SPM-2, with binding sites for target antigens CD33 and CD123, and for CD16 to engage NK cells as cytolytic effectors. Primary blasts of nearly all AML patients carry at least one of these target antigens and the pair is particularly promising for the elimination of blasts and leukemia stem cells (LSCs) from a majority of AML patients by dual-targeting agents. The cytolytic activity of NK cells mediated by SPM-2 was analyzed in vitro for primary leukemic cells from 29 patients with a broad range of AML-subtypes. Blasts from all 29 patients, including patients with genomic alterations associated with an unfavorable genetic subtype, were lysed at nanomolar concentrations of SPM-2. Maximum susceptibility was observed for cells with a combined density of CD33 and CD123 above 10,000 copies/cell. Cell populations enriched for AML-LSCs (CD34pos and CD34pos CD38neg cells) from 2 AML patients carried an increased combined antigen density and were lysed at correspondingly lower concentrations of SPM-2 than unsorted blasts. These initial findings raise the expectation that SPM-2 may also be capable of eliminating AML-LSCs and thus of prolonging survival. In the future, patients with a broad range of AML subtypes may benefit from treatment with SPM-2.

2.
Oncotarget ; 7(50): 83392-83408, 2016 Dec 13.
Article in English | MEDLINE | ID: mdl-27825135

ABSTRACT

Triplebodies are antibody-derived recombinant proteins carrying 3 antigen-binding domains in a single polypeptide chain. Triplebody SPM-1 was designed for lysis of CD19-bearing malignant B-lymphoid cells through the engagement of CD16-expressing cytolytic effectors, including NK and γδ T cells.SPM-1 is an optimized version of triplebody ds(19-16-19) and includes humanization, disulfide stabilization and the removal of potentially immunogenic sequences. A three-step chromatographic procedure yielded 1.7 - 5.5 mg of purified, monomeric protein per liter of culture medium. In cytolysis assays with NK cell effectors, SPM-1 mediated potent lysis of cancer-derived B cell lines and primary cells from patients with various B-lymphoid malignancies, which surpassed the ADCC activity of the therapeutic antibody Rituximab. EC50-values ranged from 3 to 86 pM. Finally, in an impedance-based assay, SPM-1 mediated a particularly rapid lysis of CD19-bearing target cells by engaging and activating both primary and expanded human γδ T cells from healthy donors as effectors.These data establish SPM-1 as a useful tool for a kinetic analysis of the cytolytic reactions mediated by γδ T and NK cells and as an agent deserving further development towards clinical use for the treatment of B-lymphoid malignancies.


Subject(s)
Antigens, CD19/immunology , Antineoplastic Agents, Immunological/pharmacology , Cytotoxicity, Immunologic/drug effects , Intraepithelial Lymphocytes/drug effects , Killer Cells, Natural/drug effects , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphoma, B-Cell/drug therapy , Antineoplastic Agents, Immunological/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Intraepithelial Lymphocytes/immunology , Killer Cells, Natural/immunology , Kinetics , Lymphocyte Activation/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Rituximab/pharmacology , Tumor Cells, Cultured
3.
Analyst ; 141(7): 2284-95, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-26958659

ABSTRACT

Cancer therapy via redirected lysis mediated by antibodies and antibody-derived agents relies on the availability of substantial numbers of sufficiently active immune effector cells. To monitor antitumor responses before and during therapy, sensitive methods are needed, capable of quantitating specific lysis of target cells. Here we present a chip-based single-cell cytometric assay, which uses adherent human target cells arrayed in structured micro-fields. Using a fluorescent indicator of cell death and time-lapse microscopy in an automated high-throughput mode, we measured specific target cell lysis by activated human NK cells, mediated by the therapeutic single chain triplebody SPM-2 (33-16-123). This antibody-derived tri-specific fusion protein carries binding sites for the myeloid antigens CD33 and CD123 and recruits NK cells via a binding site for the Fc-receptor CD16. Specific lysis increased with increasing triplebody concentration, and the single-cell assay was validated by direct comparison with a standard calcein-release assay. The chip-based approach allowed measurement of lysis events over 16 hours (compared to 4 hours for the calcein assay) and required far smaller numbers of primary cells. In addition, dynamic properties inaccessible to conventional methods provide new details about the activation of cytolytic effector cells by antibody-derived agents. Thus, the killing rate exhibited a dose-dependent maximum during the reaction interval. In clinical applications ex vivo monitoring of NK activity of patient's endogenous cells will likely help to choose appropriate therapy, to detect impaired or recovered NK function, and possibly to identify rare subsets of cancer cells with particular sensitivity to effector-cell mediated lysis.


Subject(s)
Killer Cells, Natural/cytology , Microchip Analytical Procedures/methods , Single-Cell Analysis/methods , Single-Chain Antibodies/metabolism , Cell Death , Cell Line, Tumor , HEK293 Cells , Humans , Time Factors
4.
Oncotarget ; 7(16): 22579-89, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-26981773

ABSTRACT

Simultaneous targeting of multiple tumor-associated antigens (TAAs) in cancer immunotherapy is presumed to enhance tumor cell selectivity and to reduce immune escape.The combination of B lymphoid marker CD19 and myeloid marker CD33 is exclusively present on biphenotypic B/myeloid leukemia cells. Triplebody 33-3-19 binds specifically to both of these TAAs and activates T cells as immune effectors. Thereby it induces specific lysis of established myeloid (MOLM13, THP-1) and B-lymphoid cell lines (BV173, SEM, Raji, ARH77) as well as of primary patient cells. EC50 values range from 3 pM to 2.4 nM. In accordance with our hypothesis, 33-3-19 is able to induce preferential lysis of double- rather than single-positive leukemia cells in a target cell mixture: CD19/CD33 double-positive BV173 cells were eliminated to a significantly greater extent than CD19 single-positive SEM cells (36.6% vs. 20.9% in 3 hours, p = 0.0048) in the presence of both cell lines. In contrast, equivalent elimination efficiencies were observed for both cell lines, when control triplebody 19-3-19 or a mixture of the bispecific single chain variable fragments 19-3 and 33-3 were used. This result highlights the potential of dual-targeting agents for efficient and selective immune-intervention in leukemia patients.


Subject(s)
Antigens, Neoplasm/drug effects , Antineoplastic Agents/pharmacology , Immunotherapy/methods , Leukemia, Biphenotypic, Acute , Single-Chain Antibodies/pharmacology , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/immunology , Antigens, CD19/immunology , Antigens, Differentiation, Myelomonocytic/drug effects , Antigens, Differentiation, Myelomonocytic/immunology , Humans , Sialic Acid Binding Ig-like Lectin 3/immunology
5.
Oncotarget ; 5(15): 6466-83, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25115385

ABSTRACT

Triplebody 19-3-19, an antibody-derived protein, carries three single chain fragment variable domains in tandem in a single polypeptide chain. 19-3-19 binds CD19-bearing lymphoid cells via its two distal domains and primary T cells via its CD3-targeting central domain in an antigen-specific manner. Here, malignant B-lymphoid cell lines and primary cells from patients with B cell malignancies were used as targets in cytotoxicity tests with pre-stimulated allogeneic T cells as effectors. 19-3-19 mediated up to 95 % specific lysis of CD19-positive tumor cells and, at picomolar EC50 doses, had similar cytolytic potency as the clinically successful agent Blinatumomab. 19-3-19 activated resting T cells from healthy unrelated donors and mediated specific lysis of both autologous and allogeneic CD19-positive cells. 19-3-19 led to the elimination of 70 % of CD19-positive target cells even with resting T cells as effectors at an effector-to-target cell ratio of 1 : 10. The molecule is therefore capable of mediating serial lysis of target cells by a single T cell. These results highlight that central domains capable of engaging different immune effectors can be incorporated into the triplebody format to provide more individualized therapy tailored to a patient's specific immune status.


Subject(s)
Lymphocytes/immunology , Lymphoma, B-Cell/therapy , Single-Chain Antibodies/pharmacology , T-Lymphocytes/immunology , Aged , Aged, 80 and over , Antibody-Dependent Cell Cytotoxicity , Female , HEK293 Cells , Humans , Immunization, Passive/methods , Lymphocyte Activation , Lymphoma, B-Cell/immunology , Male , Middle Aged , Single-Chain Antibodies/immunology , Young Adult
6.
MAbs ; 6(1): 286-96, 2014.
Article in English | MEDLINE | ID: mdl-24135631

ABSTRACT

The single-chain triplebody HLA-ds16-hu19 consists of three single-chain Fv (scFv) antibody fragments connected in a single polypeptide chain. This protein with dual-targeting capacity mediated preferential lysis of antigen double positive(dp) over single-positive (sp) leukemic cells by recruitment of natural killer (NK) cells as effectors. The two distal scFv modules were specific for the histocompatibility protein HLA-DR and the lymphoid antigen CD19, the central one for the Fc gamma receptor CD16. In antibody-dependent cellular cytotoxicity (ADCC) experiments with a mixture of leukemic target cells comprising both HLA-DR sp HuT-78 or Kasumi-1 cells and (HLA-DR plus CD19) dp SEM cells, the triplebody mediated preferential lysis of the dp cells even when the sp cells were present in ≤ 20-fold numerical excess.The triplebody promoted equal lysis of SEM cells at 2.5-fold and 19.5-fold lower concentrations than the parental antibodies specific for HLA-DR and CD19, respectively. Finally, the triplebody also eliminated primary leukemic cells at lower concentrations than an equimolar mixture of bispecific single-chain Fv fragments (bsscFvs) separately addressing each target antigen (hu19-ds16 and HLA-ds16). The increased selectivity of targeting and the preferential lysis of dp over sp cells achieved by dual-targeting open attractive new perspectives for the use of dual-targeting agents in cancer therapy.


Subject(s)
Antibodies, Neoplasm/pharmacology , Antigens, CD19/immunology , Antineoplastic Agents/pharmacology , Drug Delivery Systems , HLA-DR Antigens/immunology , Immunity, Cellular/drug effects , Killer Cells, Natural/immunology , Neoplasm Proteins/immunology , Single-Chain Antibodies/pharmacology , Animals , Antibodies, Neoplasm/genetics , Antibodies, Neoplasm/immunology , Antigens, CD19/genetics , Antineoplastic Agents/immunology , CHO Cells , Cricetinae , Cricetulus , Female , HLA-DR Antigens/genetics , Humans , Immunity, Cellular/immunology , Killer Cells, Natural/pathology , Male , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology
7.
J Transl Med ; 11: 289, 2013 Nov 16.
Article in English | MEDLINE | ID: mdl-24237598

ABSTRACT

BACKGROUND: The capacity of patient's Natural Killer cells (NKs) to be activated for cytolysis is an important prerequisite for the success of antibody-derived agents such as single-chain triplebodies (triplebodies) in cancer therapy. NKs recovered from AML patients at diagnosis are often found to be reduced in peripheral blood titers and cytolytic activity. Here, we had the unique opportunity to compare blood titers and cytolytic function of NKs from an AML patient with those of a healthy monozygotic twin. The sibling's NKs were compared with the patient's drawn either at diagnosis or in remission after chemotherapy. The cytolytic activities of NKs from these different sources for the patient's autologous AML blasts and other leukemic target cells in conjunction with triplebody SPM-2, targeting the surface antigens CD33 and CD123 on the AML cells, were compared. METHODS: Patient NKs drawn at diagnosis were compared to NKs drawn in remission after chemotherapy and a sibling's NKs, all prepared from PBMCs by immunomagnetic beads (MACS). Redirected lysis (RDL) assays using SPM-2 and antibody-dependent cellular cytotoxicity (ADCC) assays using the therapeutic antibody RituximabTM were performed with the enriched NKs. In addition, MACS-sorted NKs were analyzed for NK cell activating receptors (NCRs) by flow cytometry, and the release of TNF-alpha and IFN-gamma from blood samples of both siblings after the addition of the triplebody were measured in ELISA-assays. RESULTS: Patient NKs isolated from peripheral blood drawn in remission produced comparable lysis as NKs from the healthy twin against the patient's autologous bone marrow (BM) blasts, mediated by SPM-2. The NCR receptor expression profiles on NKs from patient and twin were similar, but NK cell titers in peripheral blood were lower for samples drawn at diagnosis than in remission. CONCLUSIONS: Peripheral blood NK titers and ex vivo cytolytic activities mediated by triplebody SPM-2 were comparable for cells drawn from an AML patient in remission and a healthy twin. If these results can be generalized, then NKs from AML patients in remission are sufficient in numbers and cytolytic activity to make triplebodies promising new agents for the treatment of AML.


Subject(s)
Cytotoxicity, Immunologic , Killer Cells, Natural/immunology , Leukemia, Myeloid, Acute/immunology , Remission Induction , Twins, Monozygotic , Adult , Antibody-Dependent Cell Cytotoxicity , Female , Flow Cytometry , Humans , Young Adult
8.
MAbs ; 4(1): 45-56, 2012.
Article in English | MEDLINE | ID: mdl-22327429

ABSTRACT

To test the hypothesis that dual-targeting confers the novel ability of selective binding to antigen double-positive over antigen single-positive cells, a single-chain triplebody (sctb), HLA-ds16-hu19, was produced and characterized. The molecule carries three single-chain Fv (scFv) antibody fragments in a single polypeptide chain, the two distal ones specific for the human histocompatibility protein HLA-DR and the B-lymphoid cell surface protein CD19, the central one for CD16, the human low affinity Fc-receptor FcγRIII. For comparison, the bispecific scFvs (bsscFv) hu19-ds16 and HLA-ds16 were also produced. All CD16 binding modules are disulfide-stabilized (ds). The sctb bound simultaneously to both CD19 and HLA-DR on the same cancer cell and, thus, showed functional dual-targeting. In a mixing-experiment with HLA-DR single-positive HUT-78 cells and (HLA-DR plus CD19) double-positive SEM cells, the triplebody showed preferential binding to the double-positive cells, even when the single-positive cells were present in a numerical excess of up to 20-fold. In antibody-dependent cellular cytotoxicity experiments with mononuclear cells as effector cells, the sctb promoted equal lysis of Raji cells, an antigen double-positive cell line, at 130-fold lower concentrations than the bsscFv hu19-ds16, indicating that both distal scFvs of the sctb contributed to tumor cell lysis. A panel of stably-transfected HEK293 cell lines was generated that included CD19- and HLA-DR single-positive and (HLA-DR plus CD19) double-positive lines with antigen-surface densities varying over a broad range. Using a pair of cell lines with matching densities, the sctb eliminated double-positive target cells preferentially single-positive cells. This ability of preferential or selective targeting of antigen double-positive over single-positive cells opens attractive new perspectives for the use of dual-targeting sctbs in cancer therapy.


Subject(s)
Antibodies, Bispecific/immunology , Antibody Specificity/immunology , Antigens, CD19/immunology , HLA-DR Antigens/immunology , Receptors, IgG/immunology , Recombinant Fusion Proteins/immunology , Animals , Antibodies, Bispecific/genetics , Antibodies, Bispecific/metabolism , Antibody-Dependent Cell Cytotoxicity/immunology , Antigens, CD19/metabolism , CHO Cells , Cell Line , Cricetinae , Cricetulus , GPI-Linked Proteins/immunology , GPI-Linked Proteins/metabolism , HEK293 Cells , HLA-DR Antigens/metabolism , Humans , Killer Cells, Natural/immunology , Protein Binding , Receptors, IgG/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Single-Chain Antibodies/genetics , Single-Chain Antibodies/metabolism
9.
J Immunol Methods ; 373(1-2): 67-78, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-21855548

ABSTRACT

Protein- or glyco-engineering of antibody molecules can be used to enhance Fc-mediated effector functions. ScFv-Fc fusion proteins (scFv-Fc) represent interesting antibody derivatives due to their relatively simple design and increased tissue penetration. Here, the impact of protein- and glyco-engineering on ADCC potency of a panel of human IgG1-based scFv-Fc was tested. Three matched sets of scFv-Fc variants targeting CD7, CD20 or HLA class II and optimized for CD16a binding by mutagenesis, lack of core-fucose, or their combination, were generated and functionally tested in comparison to the corresponding wild type scFv-Fc. Antigen binding activity was not compromised by altered glycosylation or Fc mutagenesis, whereas Fc binding to CD16a was significantly enhanced in the order: non-core fucosylated/Fc-mutated double-engineered≫Fc-mutated≥non-core-fucosylated>wild-type IgG1-Fc. All engineered variants triggered potent ADCC with up to 100-fold reduced EC50 values compared to non-engineered variants. Interestingly, double-engineered variants were similarly effective in triggering ADCC compared to single-engineered variants irrespective of their 1 log greater CD16a binding affinity. Thus, these data demonstrate that protein- and glyco-engineering enhances NK-cell mediated ADCC of scFv-Fc similarly and show that enhancing CD16a affinity beyond a certain threshold does not result in a further increase of NK-cell mediated ADCC.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/immunology , Immunoglobulin Fc Fragments/immunology , Killer Cells, Natural/immunology , Receptors, IgG/immunology , Single-Chain Antibodies/immunology , Animals , Antibody Affinity/immunology , Antigens, CD20/immunology , Antigens, CD20/metabolism , Antigens, CD7/immunology , Antigens, CD7/metabolism , Blotting, Western , CHO Cells , Cell Line , Cell Line, Tumor , Cricetinae , Cricetulus , Flow Cytometry , Fucose/metabolism , Glycoproteins/genetics , Glycoproteins/immunology , Glycoproteins/metabolism , HLA-D Antigens/immunology , HLA-D Antigens/metabolism , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Immunoglobulin G/immunology , Killer Cells, Natural/metabolism , Mutagenesis , Protein Binding , Protein Engineering/methods , Receptors, Fc/immunology , Receptors, Fc/metabolism , Receptors, IgG/genetics , Receptors, IgG/metabolism , Single-Chain Antibodies/genetics , Single-Chain Antibodies/metabolism
10.
Cancer Lett ; 303(2): 128-39, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21339041

ABSTRACT

Bispecific scFv antibody-derivatives (bsscFvs) recruiting natural killer (NK) cells for the lysis of malignant cells have therapeutic potential. However, a bsscFv specific for the B-lymphoid tumor antigen CD19 and the trigger molecule CD16 on NK cells had similar affinities for both antigens (42 and 58nM, respectively) and was not optimal for cytotoxicity. Therefore, a bispecific tribody (bsTb) was constructed with two binding sites for CD19 and one for CD16. This bsTb contained a CD19-specific Fab fragment carrying a CD16-specific scFv fused to its light chain and a CD19-specific scFv fused to its heavy chain. The bsTb was compared with a bispecific bibody (bsBb) lacking the CD19-specific scFv. The bsTb had 3-fold greater avidity for CD19 than the bsBb (8 and 24nM, respectively), while both had equal affinity for CD16 (56nM). Both molecules mediated antibody-dependent cellular cytotoxicity (ADCC) of leukemia-derived SEM cells and primary cells from leukemia patients. The bsTb showed half-maximum effective concentrations (EC(50)) of 55pM and promoted equal lysis as the bsBb and the bsscFv at 6- and 12-fold lower concentrations, respectively. Among these three molecules the bsTb showed the most promising in vitro properties which are anticipated to be displayed also in vivo.


Subject(s)
Antibodies, Bispecific/chemistry , Antigens, CD19/chemistry , Lymphoma, B-Cell/immunology , Receptors, IgG/chemistry , Antibody-Dependent Cell Cytotoxicity/immunology , Cell Line, Tumor , Dimerization , Flow Cytometry/methods , Humans , Immunoglobulin Fragments , Immunotherapy/methods , Kinetics , Leukocytes, Mononuclear/cytology , Lymphoma, B-Cell/therapy , Protein Binding , Recombinant Fusion Proteins/chemistry
11.
MAbs ; 3(1): 21-30, 2011.
Article in English | MEDLINE | ID: mdl-21081841

ABSTRACT

A single-chain triplebody (sctb) 33-ds16-ds19 comprising two distal single-chain Fv fragments (scFvs) specific for the lymphoid antigen CD19 and the myeloid antigen CD33 flanking a central scFv specific for CD16, which is the low affinity Fc-receptor (FcγRIII) present on natural killer cells and macrophages, was produced and its properties were investigated. CD33 and CD19 in combination are present on acute leukemiablasts with mixed lineage phenotype, but not on normal human hematopoietic cells. For comparison, two bispecific scFvs (bsscFvs), ds19-ds16 and 33-ds16, with monovalent binding to CD19 and CD33, respectively, were also studied. The sctb 33-ds16-ds19 specifically interacted with all 3 antigens. On the antigen double-positive cell line BV-173, the sctb bound with 2-fold greater avidity than bsscFv ds19-ds16 (KD = 21 vs. 42 nM) and with 1.4-fold greater avidity than bsscFv 33-ds16 (KD = 29 nM). All 3 fusion proteins had similar affinity for CD16 and sufficient thermic stability in human serum. In antibody-dependent cellular cytotoxicity (ADCC) reactions with human mononuclear cells as effectors, the sctb promoted lysis of BV-173 cells at 23-fold lower concentrations than bsscFv ds19-ds16 and at 1.4-fold lower concentrations than bsscFv 33-ds16. The sctb also mediated potent ADCC of the antigen double-positive mixed lineage leukemia cell line SEM, and the half-maximal concentration EC50 for BV-173 cells was 7 pM. Therefore, CD19 and CD33 are present on the surface of these leukemic cell lines such that they can be connected by a single sctb molecule, permitting the recruitment of NK cells via CD16 and tumor cell lysis.


Subject(s)
Antigens, CD19/immunology , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/immunology , Single-Chain Antibodies/immunology , Animals , Antibody Specificity/immunology , Antigens, CD/metabolism , Antigens, CD19/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Blotting, Western , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Cytotoxicity, Immunologic/immunology , Flow Cytometry , HEK293 Cells , Humans , Kinetics , Leukemia/immunology , Leukemia/pathology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Protein Binding/immunology , Protein Stability , Receptors, IgG/immunology , Receptors, IgG/metabolism , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Sialic Acid Binding Ig-like Lectin 3 , Single-Chain Antibodies/genetics , Single-Chain Antibodies/metabolism , Temperature
12.
Mol Cancer ; 9: 301, 2010 Nov 23.
Article in English | MEDLINE | ID: mdl-21092273

ABSTRACT

BACKGROUND: Advanced melanoma is characterized by a pronounced resistance to therapy leading to a limited patient survival of ~6 - 9 months. Here, we report on a novel bifunctional therapeutic fusion protein, designated anti-MCSP:TRAIL, that is comprised of a melanoma-associated chondroitin sulfate proteoglycan (MCSP)-specific antibody fragment (scFv) fused to soluble human TRAIL. MCSP is a well-established target for melanoma immunotherapy and has recently been shown to provide important tumorigenic signals to melanoma cells. TRAIL is a highly promising tumoricidal cytokine with no or minimal toxicity towards normal cells. Anti-MCSP:TRAIL was designed to 1. selectively accrete at the cell surface of MCSP-positive melanoma cells and inhibit MCSP tumorigenic signaling and 2. activate apoptotic TRAIL-signaling. RESULTS: Treatment of a panel of MCSP-positive melanoma cell lines with anti-MCSP:TRAIL induced TRAIL-mediated apoptotic cell death within 16 h. Of note, treatment with anti-MCSP:sTRAIL was also characterized by a rapid dephosphorylation of key proteins, such as FAK, implicated in MCSP-mediated malignant behavior. Importantly, anti-MCSP:TRAIL treatment already inhibited anchorage-independent growth by 50% at low picomolar concentrations, whereas > 100 fold higher concentrations of non-targeted TRAIL failed to reduce colony formation. Daily i.v. treatment with a low dose of anti-MCSP:TRAIL (0.14 mg/kg) resulted in a significant growth retardation of established A375 M xenografts. Anti-MCSP:TRAIL activity was further synergized by co-treatment with rimcazole, a σ-ligand currently in clinical trials for the treatment of various cancers. CONCLUSIONS: Anti-MCSP:TRAIL has promising pre-clinical anti-melanoma activity that appears to result from combined inhibition of tumorigenic MCSP-signaling and concordant activation of TRAIL-apoptotic signaling. Anti-MCSP:TRAIL alone, or in combination with rimcazole, may be of potential value for the treatment of malignant melanoma.


Subject(s)
Chondroitin Sulfate Proteoglycans/metabolism , Melanoma/drug therapy , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/therapeutic use , TNF-Related Apoptosis-Inducing Ligand/therapeutic use , Animals , Apoptosis/drug effects , Cell Line , Cell Line, Tumor , Cells, Cultured , Chondroitin Sulfate Proteoglycans/genetics , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Male , Melanocytes/drug effects , Melanocytes/metabolism , Melanoma/metabolism , Mice , Mice, Nude , Proteome/drug effects , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Xenograft Model Antitumor Assays
13.
Br J Haematol ; 150(5): 574-86, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20636437

ABSTRACT

Two trivalent constructs consisting of single-chain Fv antibody fragments (scFvs) specific for the interleukin-3 receptor alpha chain (CD123), CD33 and the Fcgamma-receptor III (CD16) were designed and characterized for the elimination of acute myeloid leukaemia (AML) cells. The dual targeting single-chain Fv triplebody (sctb) [123 x ds16 x 33] and the mono targeting sctb [123 x ds16 x 123] both specifically bound their respective target antigens and were stable in human serum at 37 degrees C for at least 5 d. Both constructs induced potent antibody-dependent cellular cytotoxicity (ADCC) of two different AML-derived CD33- and CD123 double-positive cell lines in the low picomolar range using isolated mononuclear cells (MNCs) as effector cells. In these experiments the dual targeting molecule produced significantly stronger lysis than the mono targeting agent. In addition, the sctbs showed a high potency in mediating ADCC of primary leukaemia cells isolated from peripheral blood or bone marrow of seven AML patients. Hence, these novel molecules displayed potent anti-leukaemic effects against AML cells in vitro and represent attractive candidates for further preclinical development.


Subject(s)
Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/immunology , Interleukin-3 Receptor alpha Subunit/immunology , Leukemia, Myeloid, Acute/immunology , Single-Chain Antibodies/immunology , Antibody-Dependent Cell Cytotoxicity/immunology , GPI-Linked Proteins , Humans , Immunoglobulin Fragments/immunology , Killer Cells, Natural/immunology , Leukemia, Myeloid, Acute/pathology , Receptors, IgG/immunology , Recombinant Proteins/immunology , Sialic Acid Binding Ig-like Lectin 3 , Tumor Cells, Cultured
14.
J Immunother ; 33(6): 599-608, 2010.
Article in English | MEDLINE | ID: mdl-20551837

ABSTRACT

Single-chain Fv triplebodies (sctb), consisting of a single polypeptide chain with 3 single-chain antibody variable fragments connected in tandem, were generated as antileukemic agents. A CD19-specific sctb of this format has previously been shown to be superior to a bispecific single-chain Fv antibody fragment (bsscFv) for the elimination of leukemic B-lineage cells, but corresponding targeted agents for the treatment of acute myeloid leukemia are still lacking. For this purpose, both a bsscFv and a sctb specific for CD33 and the trigger molecule CD16 (FcgammaRIII) were produced. The sctb displayed 3.5-fold greater avidity for CD33 than the bsscFv 33xds16, whereas both had close to equal affinity for CD16. In antibody-dependent cellular cytotoxicity (ADCC) reactions with human mononuclear cells as effectors, both the bsscFv 33xds16 and the sctb induced lysis of tumor cells with half maximum effective concentrations (EC50) in the low picomolar range. It is interesting to note that the sctb promoted equal lysis of human leukemia-derived cell lines at 10 to 200-fold lower concentrations than the bsscFv. Both molecules mediated ADCC of primary patient cells. In conclusion, both the bsscFv 33xds16 and the sctb 33xds16x33 eliminated acute myeloid leukemia cells in ADCC reactions, but the novel sctb format showed significantly greater specific activity.


Subject(s)
Antibodies, Bispecific/pharmacology , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/immunology , Immunotherapy , Leukemia, Myeloid, Acute/immunology , Receptors, IgG/immunology , Recombinant Fusion Proteins/pharmacology , Animals , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/genetics , Antibody Affinity , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/immunology , Antigens, CD/biosynthesis , Antigens, CD/genetics , Antigens, Differentiation, Myelomonocytic/biosynthesis , Antigens, Differentiation, Myelomonocytic/genetics , CHO Cells , Computer Simulation , Cricetinae , Cricetulus , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Protein Engineering , Protein Stability , Receptors, IgG/genetics , Receptors, IgG/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Sialic Acid Binding Ig-like Lectin 3
15.
Br J Haematol ; 148(6): 879-89, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20064159

ABSTRACT

Four new single-chain Fv antibody fragments (scFvs) specific for the human leucocyte surface antigen CD123 (interleukin-3 receptor alpha) were generated to achieve preferential targeting of leukaemia stem cells (LSCs) in acute myeloid leukaemia (AML). The scFvs were isolated from a phage display library generated with spleen RNA from mice, immunized with a fusion protein consisting of the extracellular domain of CD123 and the Fc domain of a human immunoglobulin G1. The scFvs displayed CD123-specific binding on tumour cells (binding constants (K(D)) 4.5-101 nmol/l). The scFv with the highest affinity was used to design two cell death-inducing molecules. First, an immunotoxin, a fusion protein with truncated Pseudomonas Exotoxin A, induced potent apoptosis of AML-derived MOLM-13 and SKNO-1 cells at nanomolar concentrations. Second, the fusion to another scFv, specific for the low affinity Fcgamma-receptor III (CD16), created a bispecific single chain Fv (bsscFv). This bsscFv [123 x ds16] mediated potent lysis of AML-derived MOLM-13, THP-1 and SKNO-1 cells in antibody-dependent cellular cytotoxicity (ADCC) reactions at picomolar concentrations. The recruitment of CD16-positive effector cells for the lysis of AML cells via CD123 represents a novel combination with attractive prospects for future clinical testing.


Subject(s)
Immunoglobulin Fragments/immunology , Interleukin-3 Receptor alpha Subunit/immunology , Leukemia, Myeloid, Acute/immunology , Single-Chain Antibodies/immunology , ADP Ribose Transferases/immunology , Animals , Antibody-Dependent Cell Cytotoxicity , Apoptosis/immunology , Bacterial Toxins/immunology , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Immunologic , Epitopes , Exotoxins/immunology , Humans , Immunotoxins/immunology , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred BALB C , Tumor Cells, Cultured , Virulence Factors/immunology , Pseudomonas aeruginosa Exotoxin A
16.
J Immunol ; 184(3): 1210-7, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20042573

ABSTRACT

Bispecific Abs offer new perspectives for cancer immunotherapy. In this study, we describe a recombinant bispecific single-chain fragment variable (bsscFv) directed against Fc alpha RI (CD89) on polymorphonuclear neutrophils (PMNs) or monocytes/macrophages and HLA class II on lymphoma target cells. Fc alpha RI and HLA class II-directed single-chain fragment variable (scFv) fragments were isolated from phage display libraries, established from the hybridomas A77 and F3.3, respectively. The two scFv molecules were connected with a 20 aa flexible linker sequence. After expression in SF21 insect cells and chromatographic purification, the bispecific molecule showed specific binding to both Ags at K(D) values of 148 +/- 42 nM and 113 +/- 25 nM for the anti-Fc alpha RI and anti-HLA class II scFv components in the bsscFv, respectively. In Ab-dependent cytotoxicity assays with PMNs as effectors and a series of lymphoma-derived cell lines (ARH-77, RAJI, REH, NALM-6, RS4;11), the bsscFv was significantly more cytotoxic than the parental murine IgG1 and its chimeric IgG1 derivative. When targeting primary tumor cell isolates from six patients with B cell malignancies, the killing capacity of the (Fc alphaRI x HLA class II) bsscFv compared favorably to conventional HLA class II mAb. Importantly, the cell lines NALM-6 and RS411, as well as two primary tumor cell isolates, were exclusively lysed by the bsscFv. To our knowledge, this is the first report of an Fc alpha RI-directed bsscFv effectively recruiting PMNs for redirected cytotoxicity against human B cell malignancies. Our data show that an (Fc alpha RI x HLA class II) bsscFv is an interesting candidate for further engineering of small, modular immunopharmaceuticals.


Subject(s)
Antibodies, Bispecific/physiology , Antibody-Dependent Cell Cytotoxicity , Antigens, CD/physiology , B-Lymphocyte Subsets/immunology , Cell Movement/immunology , HLA-D Antigens/immunology , Immunoglobulin Fragments/physiology , Immunoglobulin Variable Region/physiology , Neutrophils/immunology , Receptors, Fc/physiology , Recombinant Fusion Proteins/immunology , Animals , Antibodies, Bispecific/genetics , Antibody-Dependent Cell Cytotoxicity/genetics , Antigens, CD/genetics , B-Lymphocyte Subsets/metabolism , B-Lymphocyte Subsets/pathology , Cell Line , Cell Line, Tumor , Cell Movement/genetics , Cricetinae , HLA-D Antigens/genetics , Humans , Immunoglobulin Fragments/genetics , Immunoglobulin Variable Region/genetics , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Lymphoma, B-Cell/therapy , Mice , Neutrophils/metabolism , Neutrophils/pathology , Protein Binding/genetics , Protein Binding/immunology , Receptors, Fc/antagonists & inhibitors , Receptors, Fc/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Tumor Cells, Cultured
17.
Protein Eng Des Sel ; 22(3): 135-47, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19188138

ABSTRACT

A single-chain Fv (scFv) fragment derived from the murine antibody 4G7, specific for human lymphocyte CD19, was engineered for stability and expression in Escherichia coli in view of future use as a therapeutic protein. We compared two orthogonal knowledge-based procedures. In one approach, we designed a mutant with 14 single amino-acid substitutions predicted to correct destabilizing residues in the 4G7-wt sequence to create 4G7-mut. In the second variant, the murine CDRs were grafted to the human acceptor framework huVkappa3-huV(H)3, with 11 additional point mutations introduced to obtain a better match between CDR graft and acceptor framework, to arrive at 4G7-graft. Compared to 4G7-wt, 4G7-mut showed greater thermodynamic stability in guanidinium chloride-induced equilibrium denaturation experiments and somewhat greater stability in human serum. The loop graft maintained the comparatively high stability of the murine loop donor, but did not improve it further. Our analysis indicates that this is due to subtle strain introduced between CDRs and framework, mitigating the otherwise highly favorable properties of the human acceptor framework. This slight strain in the loop graft is also reflected in the binding affinities for CD19 on leukemic cells of 8.4 nM for 4G7-wt, 16.4 nM for 4G7-mut and 30.0 nM for 4G7-graft. This comparison of knowledge-based mutation and loop-grafting-based approaches will be important, when moving molecules forward to therapeutic applications.


Subject(s)
Antigens, CD19/genetics , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Variable Region/genetics , Point Mutation/genetics , Recombinant Fusion Proteins/genetics , Amino Acid Sequence , Amino Acid Substitution/genetics , Animals , Antibody Affinity/genetics , Antigens, CD19/chemistry , Antigens, CD19/metabolism , Chromatography, Gel , Escherichia coli/genetics , Genetic Engineering , Humans , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Heavy Chains/metabolism , Immunoglobulin Variable Region/chemistry , Immunoglobulin Variable Region/metabolism , Mice , Models, Molecular , Molecular Sequence Data , Protein Denaturation/genetics , Protein Stability , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Solubility , Thermodynamics
18.
J Immunother ; 31(9): 871-84, 2008.
Article in English | MEDLINE | ID: mdl-18833000

ABSTRACT

A novel bispecific antibody-derived recombinant protein targeting leukemias and lymphomas was designed, a single-chain Fv triple body (sctb) consisting of 1 polypeptide chain with 3 scFvs connected in tandem. The distal scFvs were specific for the tumor antigen CD19, and the central scFv for the trigger molecule CD16 (FcgammaRIII) on natural killer (NK) cells and macrophages. We had previously built a disulphide stabilized (ds) bsscFv [19 x 16] with monovalent binding for CD19 from ds components. The sctb ds[19 x 16 x 19] also used ds components and displayed 3-fold greater avidity for CD19 than the bsscFv (KD = 13 vs. 42 nM), whereas both had equal affinity for CD16 (KD = 58 nM). Plasma half-lives in mice were 4 and 2 hours for the sctb and the bsscFv, respectively. In antibody-dependent cellular cytotoxicity reactions with human mononuclear cells as effectors, the sctb promoted equal lysis of leukemic cell lines and primary cells from leukemia and lymphoma patients at 10-fold to 40-fold lower concentrations than the bsscFv. This new format may also be applicable to a variety of other tumor antigens and effector molecules. With half-maximum effective concentrations (EC50) in the low picomolar range, the sctb ds[19 x 16 x 19] is an attractive candidate for further preclinical evaluation.


Subject(s)
Antibodies, Bispecific/immunology , Antibody-Dependent Cell Cytotoxicity , Antigens, CD19/immunology , Immunoglobulin Fragments/immunology , Killer Cells, Natural/immunology , Leukemia/therapy , Receptors, IgG/immunology , Animals , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/genetics , Antibodies, Bispecific/therapeutic use , Antigens, CD19/genetics , Antigens, CD19/metabolism , CHO Cells , Cloning, Molecular , Cricetinae , Cricetulus , Humans , Immunoglobulin Fragments/chemistry , Immunoglobulin Fragments/genetics , Immunoglobulin Fragments/metabolism , Immunoglobulin Fragments/therapeutic use , Immunotherapy , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Leukemia/immunology , Leukemia/pathology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Receptors, IgG/genetics , Receptors, IgG/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/therapeutic use
19.
Cell ; 134(4): 577-86, 2008 Aug 22.
Article in English | MEDLINE | ID: mdl-18691745

ABSTRACT

Evaluation of the therapeutic potential of RNAi for HIV infection has been hampered by the challenges of siRNA delivery and lack of suitable animal models. Using a delivery method for T cells, we show that siRNA treatment can dramatically suppress HIV infection. A CD7-specific single-chain antibody was conjugated to oligo-9-arginine peptide (scFvCD7-9R) for T cell-specific siRNA delivery in NOD/SCIDIL2rgamma-/- mice reconstituted with human lymphocytes (Hu-PBL) or CD34+ hematopoietic stem cells (Hu-HSC). In HIV-infected Hu-PBL mice, treatment with anti-CCR5 (viral coreceptor) and antiviral siRNAs complexed to scFvCD7-9R controlled viral replication and prevented the disease-associated CD4 T cell loss. This treatment also suppressed endogenous virus and restored CD4 T cell counts in mice reconstituted with HIV+ peripheral blood mononuclear cells. Moreover, scFvCD7-9R could deliver antiviral siRNAs to naive T cells in Hu-HSC mice and effectively suppress viremia in infected mice. Thus, siRNA therapy for HIV infection appears to be feasible in a preclinical animal model.


Subject(s)
HIV Infections/genetics , HIV Infections/therapy , RNA Interference , RNA, Small Interfering/metabolism , T-Lymphocytes/metabolism , Animals , Antigens, CD7/metabolism , Disease Models, Animal , Gene Expression , HIV-1/genetics , HIV-1/metabolism , Humans , Immunoglobulin Fragments/metabolism , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/metabolism , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Mice , Mice, Inbred NOD , Mice, SCID , RNA, Viral/metabolism
20.
Melanoma Res ; 18(2): 73-84, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18337643

ABSTRACT

A recombinant immunotoxin was constructed by fusing a single chain fragment variable antibody fragment, specific for the melanoma-associated chondroitin sulfate proteoglycan (MCSP), to a truncated variant of Pseudomonas exotoxin A (ETA'), carrying a C-terminal KDEL-peptide for improved retrograde intracellular transport. The resulting immunotoxin MCSP-ETA' was periplasmatically expressed in Escherichia coli and purified under native conditions by affinity chromatography resulting in a yield of approximately 30 mug/l bacterial culture. This immunotoxin induced antigen-specific apoptosis in the cultured human melanoma-derived cell lines A2058 and A375M, and treatment with a single dose of the agent eliminated up to 80% of these cells within 72 h. The dose needed for half-maximum killing (EC50) was approximately 1 nmol/l for both cell lines. MCSP-ETA' also displayed cytotoxic activity against cultured primary melanoma cells from patients with advanced disease (pathologic stages IIIC and IV), with net cell death reaching up to 70% within 96 h after treatment with a single dose of 14 nmol/l. MCSP-ETA' induced cell death synergistically with cyclosporin A, both in established human melanoma cell lines and cultured primary melanoma cells. The distinctive antigen-restricted induction of apoptosis and the synergy with cyclosporin A justify further evaluation of this novel agent with regard to its potential application for the treatment of malignant melanoma.


Subject(s)
Apoptosis , Chondroitin Sulfate Proteoglycans/immunology , Immunotoxins/pharmacology , Melanoma/pathology , ADP Ribose Transferases/immunology , ADP Ribose Transferases/metabolism , Bacterial Toxins/immunology , Bacterial Toxins/metabolism , Cell Line, Tumor , Chondroitin Sulfate Proteoglycans/metabolism , Cyclosporine/pharmacology , Cytotoxicity, Immunologic , Exotoxins/immunology , Exotoxins/metabolism , Humans , Immunosuppressive Agents/pharmacology , Immunotherapy , Immunotoxins/immunology , Immunotoxins/isolation & purification , Immunotoxins/metabolism , Melanoma/immunology , Melanoma/metabolism , Melanoma/therapy , Recombinant Fusion Proteins/immunology , Tumor Cells, Cultured , Virulence Factors/immunology , Virulence Factors/metabolism , Pseudomonas aeruginosa Exotoxin A
SELECTION OF CITATIONS
SEARCH DETAIL