Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Int J Cancer ; 139(4): 916-27, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27037627

ABSTRACT

Chondroitin sulfate proteoglycan 4 (CSPG4) has been identified as a highly promising target antigen for immunotherapy of triple-negative breast cancer (TNBC). TNBC represents a highly aggressive heterogeneous group of tumors lacking expression of estrogen, progesterone and human epidermal growth factor receptor 2. TNBC is particularly prevalent among young premenopausal women. No suitable targeted therapies are currently available and therefore, novel agents for the targeted elimination of TNBC are urgently needed. Here, we present a novel cytolytic fusion protein (CFP), designated αCSPG4(scFv)-MAP, that consists of a high affinity CSPG4-specific single-chain antibody fragment (scFv) genetically fused to a functionally enhanced form of the human microtubule-associated protein (MAP) tau. Our data indicate that αCSPG4(scFv)-MAP efficiently targets CSPG4(+) TNBC-derived cell lines MDA-MB-231 and Hs 578T and potently inhibits their growth with IC50 values of ∼200 nM. Treatment with αCSPG(scFv)-MAP resulted in induction of the mitochondrial stress pathway by activation of caspase-9 as well as endonuclease G translocation to the nucleus, while induction of the caspase-3 apoptosis pathway was not detectable. Importantly, in vivo studies in mice bearing human breast cancer xenografts revealed efficient targeting to and accumulation of αCSPG4(scFv)-MAP at tumor sites resulting in prominent tumor regression. Taken together, this preclinical proof of concept study confirms the potential clinical value of αCSPG4(scFv)-MAP as a novel targeted approach for the elimination of CSPG4-positive TNBC.


Subject(s)
Antibodies, Monoclonal/genetics , Chondroitin Sulfate Proteoglycans/metabolism , Membrane Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Single-Chain Antibodies/genetics , Triple Negative Breast Neoplasms/metabolism , tau Proteins/metabolism , Animals , Biomarkers , Biomarkers, Tumor , Caspase 3/metabolism , Caspase 9/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Chondroitin Sulfate Proteoglycans/genetics , Disease Models, Animal , Female , Gene Expression , Humans , Membrane Proteins/genetics , Mice , Molecular Targeted Therapy , Protein Binding , Protein Transport , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Tubulin/metabolism , Xenograft Model Antitumor Assays , tau Proteins/genetics
2.
Cancer Immunol Immunother ; 64(12): 1623-35, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26515978

ABSTRACT

Chimeric antigen receptor (CAR)-modified T cells emerged as effective tools in the immunotherapy of cancer but can produce severe on-target off-tissue toxicities. This risk can conceivably be overcome, at least partially, by transient transfection. The design of CARs, however, has so far not been optimized for use in non-permanent T cell modification. Here we compared the performance of T cells modified with three different first- and second-generation CARs, each specific for MCSP (HMW-MAA) which is commonly expressed by melanoma cells. Upon RNA transfer, the expression of all receptors was limited in time. The second-generation CARs, which combined CD28-CD3ζ signaling, were expressed at higher levels and more prolonged than first-generation CARs with CD3ζ only. The CD28 domain increased the cytokine production, but had only an indirect effect on the lytic capacity, by prolonging the CAR expression. Especially for the second-generation CARs, the scFv clearly impacted the level and duration of CAR expression and the T cell performance. Thus, we identified a CAR high in both expression and anti-tumor cell reactivity. T cells transfected with this CAR increased the mean survival time of mice after challenge with melanoma cells. To facilitate clinical application, this CAR was used to redirect T cells from late-stage melanoma patients by RNA transfection. These T cells mediated effective antigen-specific tumor cell lysis and release of pro-inflammatory cytokines, even after cryoconservation of the transfected T cells. Taken together, the analysis identified a CAR with superior anti-melanoma performance after RNA transfer which is a promising candidate for clinical exploration.


Subject(s)
Mitochondrial Proteins/immunology , Receptors, Antigen, T-Cell/immunology , Animals , Carrier Proteins/immunology , Cell Line, Tumor , Disease Models, Animal , Gene Expression Regulation, Neoplastic/immunology , Humans , Melanoma/immunology , Melanoma/physiopathology , Mice , Mitochondrial Proteins/genetics , Protein Stability , Protein Structure, Tertiary , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Single-Chain Antibodies/metabolism
3.
PLoS One ; 8(11): e79047, 2013.
Article in English | MEDLINE | ID: mdl-24244415

ABSTRACT

B lymphocytes are an important cell population of the immune system. However, until recently it was not possible to transduce resting B lymphocytes with retro- or lentiviral vectors, making them unsusceptible for genetic manipulations by these vectors. Lately, we demonstrated that lentiviral vectors pseudotyped with modified measles virus (MV) glycoproteins hemagglutinin, responsible for receptor recognition, and fusion protein were able to overcome this transduction block. They use either the natural MV receptors, CD46 and signaling lymphocyte activation molecule (SLAM), for cell entry (MV-LV) or the vector particles were further modified to selectively enter via the CD20 molecule, which is exclusively expressed on B lymphocytes (CD20-LV). It has been shown previously that transduction by MV-LV does not induce B lymphocyte activation. However, if this is also true for CD20-LV is still unknown. Here, we generated a vector specific for another B lymphocyte marker, CD19, and compared its ability to transduce resting B lymphocytes with CD20-LV. The vector (CD19ds-LV) was able to stably transduce unstimulated B lymphocytes, albeit with a reduced efficiency of about 10% compared to CD20-LV, which transduced about 30% of the cells. Since CD20 as well as CD19 are closely linked to the B lymphocyte activation pathway, we investigated if engagement of CD20 or CD19 molecules by the vector particles induces activating stimuli in resting B lymphocytes. Although, activation of B lymphocytes often involves calcium influx, we did not detect elevated calcium levels. However, the activation marker CD71 was substantially up-regulated upon CD20-LV transduction and most importantly, B lymphocytes transduced with CD20-LV or CD19ds-LV entered the G1b phase of cell cycle, whereas untransduced or MV-LV transduced B lymphocytes remained in G0. Hence, CD20 and CD19 targeting vectors induce activating stimuli in resting B lymphocytes, which most likely renders them susceptible for lentiviral vector transduction.


Subject(s)
Antigens, CD19/metabolism , Antigens, CD20/metabolism , B-Lymphocytes/metabolism , Genetic Vectors , Lymphocyte Activation , Measles virus , Transduction, Genetic , Antigens, CD19/genetics , Antigens, CD20/genetics , B-Lymphocytes/cytology , G1 Phase/genetics , HEK293 Cells , Humans
4.
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
5.
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
6.
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
7.
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
8.
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
9.
Blood ; 116(13): 2277-85, 2010 Sep 30.
Article in English | MEDLINE | ID: mdl-20566893

ABSTRACT

The use of dendritic cells (DCs) in therapeutic cancer vaccination requires their loading with tumor-specific antigen(s). DEC-205, a phagocytosis receptor mediating antigen uptake, is associated with CD8(+) T-cell responses in mice. Here we fused an anti-DEC-205scFv to an HLA-DP4-restricted epitope from the tumor antigen MAGE-A3, and examined the suitability and efficacy of DEC-205 to deliver a helper epitope to human monocyte-derived DCs (moDCs). The construct specifically bound DEC-205 on human moDCs without negative impact on DC phenotype and function. We measured antigen presentation with specific autologous CD4(+) T cells, generated by TCR-RNA transfection. DEC-205 targeting resulted in significant major histocompatibility complex class II-restricted antigen presentation, and was superior to loading DCs by electroporation of mRNA encoding endosome-targeted MAGE-A3-DCLAMP or by direct peptide pulsing. Anti-DEC-205scFv-MAGE-A3 was presented 100 times more efficiently than the control constructs. DC maturation before or during incubation with anti-DEC-205scFv-MAGE-A3 reduced the interleukin-10/interleukin-2 ratio. Moreover, we successfully applied the DEC-205 targeting strategy to moDCs from malignant melanoma patients. Again, DEC-205-targeted mature DCs (mDCs) presented the antigen more efficiently than peptide-pulsed DCs and maintained their stimulatory capacity after cryoconservation. Thus, DEC-205 targeting represents a feasible and effective method to deliver helper epitopes to DCs in anticancer vaccine strategies, which may also be suitable for DC targeting in vivo.


Subject(s)
Antigen Presentation/immunology , Antigens, CD/immunology , Dendritic Cells/immunology , Histocompatibility Antigens Class II/metabolism , Lectins, C-Type/immunology , Receptors, Cell Surface/immunology , Amino Acid Sequence , Antibodies/administration & dosage , Antibodies/genetics , Antigens, CD/metabolism , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cytokines/biosynthesis , Dendritic Cells/cytology , Epitopes/administration & dosage , Epitopes/genetics , HLA-DP Antigens/metabolism , HLA-DP beta-Chains , Humans , Immunotherapy, Active , In Vitro Techniques , Lectins, C-Type/metabolism , Melanoma/immunology , Melanoma/therapy , Minor Histocompatibility Antigens , Molecular Sequence Data , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Receptors, Cell Surface/metabolism , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology
10.
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
11.
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
12.
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
13.
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
...