Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Front Immunol ; 14: 1060905, 2023.
Article in English | MEDLINE | ID: mdl-36911670

ABSTRACT

New treatments are required to enhance current therapies for lung cancer. Mesothelin is a surface protein overexpressed in non-small cell lung cancer (NSCLC) that shows promise as an immunotherapeutic target in phase I clinical trials. However, the immunosuppressive environment in NSCLC may limit efficacy of these therapies. We applied time-of-flight mass cytometry to examine the state of circulating mononuclear cells in fourteen patients undergoing treatment for unresectable lung cancer. Six patients had earlier stage NSCLC (I-IVA) and eight had highly advanced NSCLC (IVB). The advanced NSCLC patients relapsed with greater frequency than the earlier stage patients. Before treatment, patients with very advanced NSCLC had a greater proportion of CD14- myeloid cells than patients with earlier NSCLC. These patients also had fewer circulating natural killer (NK) cells bearing an Fc receptor, CD16, which is crucial to antibody-dependent cellular cytotoxicity. We designed a high affinity tri-specific killer engager (TriKE®) to enhance NK cytotoxicity against mesothelin+ targets in this environment. The TriKE consisted of CD16 and mesothelin binding elements linked together by IL-15. TriKE enhanced proliferation of lung cancer patient NK cells in vitro. Lung cancer lines are refractory to NK cell killing, but the TriKE enhanced cytotoxicity and cytokine production by patient NK cells when challenged with tumor. Importantly, TriKE triggered NK cell responses from patients at all stages of disease and treatment, suggesting TriKE can enhance current therapies. These pre-clinical studies suggest mesothelin-targeted TriKE has the potential to overcome the immunosuppressive environment of NSCLC to treat disease.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Lung Neoplasms/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Mesothelin , Killer Cells, Natural/metabolism , Antibody-Dependent Cell Cytotoxicity , Immunosuppressive Agents/metabolism
2.
Vet Comp Oncol ; 18(4): 664-674, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32187827

ABSTRACT

We previously reported that eBAT, an EGF-targeted angiotoxin, was safe and it improved the overall survival for dogs with splenic haemangiosarcoma when added to the standard of care in a single cycle of three administrations in the minimal residual disease setting. Our objective for the SRCBST-2 trial was to assess whether increased dosing through multiple cycles of eBAT would be well tolerated and would further enhance the benefits of eBAT. Eligibility was expanded to dogs with stage 3 haemangiosarcoma, provided that gross lesions could be surgically excised. The interval between eBAT and the start of chemotherapy was reduced, and the experimental therapy was expanded to three cycles, each administered at the biologically active dose (50 µg/kg) on a Monday/Wednesday/Friday schedule following splenectomy, and scheduled 1 week prior to the first, second and fifth doxorubicin chemotherapy. Twenty-five dogs were enrolled; six experienced acute hypotension with two requiring hospitalization. Self-limiting elevation of ALT was observed in one dog. A statistically significant survival benefit was not seen in this study in eBAT-treated dogs compared with a Contemporary comparison group of dogs with stages 1-3 haemangiosarcoma treated with standard of care alone. Our results indicate that repeated dosing cycles of eBAT starting 1 week prior to doxorubicin chemotherapy led to greater toxicity and reduced efficacy compared with a single cycle given between surgery and a delayed start of chemotherapy. Further work is needed to understand the precise mechanisms of action of eBAT in order to optimize its clinical benefits in the treatment of canine haemangiosarcoma and other tumours. IACUC Protocols 1110A06186 and 1507-32804A.


Subject(s)
Chemotherapy, Adjuvant/veterinary , Dog Diseases/drug therapy , Epidermal Growth Factor/pharmacology , Hemangiosarcoma/veterinary , Splenic Neoplasms/veterinary , Animals , Antibiotics, Antineoplastic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Chemotherapy, Adjuvant/methods , Dog Diseases/pathology , Dogs , Doxorubicin/therapeutic use , Female , Hemangiosarcoma/drug therapy , Male , Splenic Neoplasms/drug therapy , Splenic Neoplasms/pathology , Treatment Outcome
3.
Clin Pharmacol ; 10: 113-121, 2018.
Article in English | MEDLINE | ID: mdl-30288129

ABSTRACT

PURPOSE: Human sarcomas are rare and difficult to treat cancerous tumors typically arising from soft tissue or bone. Conversely, carcinomas are the most common cancer subtype in humans and the primary cause of mortality across all cancer patients. While conventional therapeutic modalities can prolong disease-free intervals and survival in some cases, treatment of refractory or recurrent solid tumors is challenging, and tumor-related mortality remains unacceptably high. The identification of overexpressed cell surface receptors on sarcoma and carcinoma cells has provided a valuable tool to develop targeted toxins as an alternative anticancer strategy. Recent investigation of recombinant protein-linked toxins that specifically target these cancer receptors has led to the development of highly specific, cytotoxic, and deimmunized drugs that can kill cancer cells. METHODS: This study investigated a recombinant protein called epidermal growth factor bispecific angiotoxin (eBAT), which is designed to target the epidermal growth factor receptor (EGFR) on cancer cells and the urokinase plasminogen activator receptor (uPAR) on cancer cells and associated tumor vasculature. Both receptors are expressed by a variety of human sarcomas and carcinomas. Flow cytometry techniques were used to determine binding affinity of eBAT to cancer cells, and proliferation assays were performed to calculate tumor killing ability based on half-maximal inhibitory concentrations. RESULTS: eBAT demonstrated cytotoxicity against a variety of sarcoma and carcinoma cells that overexpress EGFR and uPAR in vitro and showed greater cell killing ability and binding affinity to cancer cells compared with its monospecific counterparts. CONCLUSION: The results of our study are promising, and further studies will be necessary to confirm the applicability of eBAT as a supplementary therapy for a variety of sarcomas, carcinomas, and possibly other refractory malignancies that express EGFR and uPAR.

4.
Toxins (Basel) ; 10(1)2018 01 06.
Article in English | MEDLINE | ID: mdl-29316610

ABSTRACT

Diphtheria toxin (DT) related targeted toxins are effective in cancer treatment, but efficacy diminishes in time because of their immunogenic potential and/or former vaccinations. In order to overcome this limitation for DT2219, a promising bispecific targeted toxin which targets CD19 and CD22, we deimmunized the DT moiety, and thereby developed an exciting improved drug (dDT2219) which still has the potential to sufficiently target B-cell malignancies but also limits clearance because of its reduced immunogenicity. The DT moiety was modified by inducing point mutations in prominent positions on the molecular surface. The new engineered dDT2219 was tested for activity, efficacy, and specificity using functional assays, proliferation assays, and flow cytometry. Furthermore, 12 samples of Chronic Lymphatic Leukemia (CLL) patients were used to assess binding. Immunogenicity was determined using a BALB/c mouse model. dDT2219 was efficient and specific against B-cell malignancies such as Bukitt-Lymphoma cell lines Daudi and Raji. dDT2219 showed specific binding on targets and on CLL samples. Intraperitoneal vaccination of immune competent mice showed that even after multiple administrations with increasing doses, induction of neutralizing antibodies was significantly lower in the dDT2219 treated animal group. The new dDT2219 combines potent anti-tumor cell activity with a reduced immunogenicity. With regard to the frequent development of neutralizing antibodies after multiple administrations with immunotoxins, dDT2219 shows promise to overcome this limitation and thus might maintain effectiveness even after multiple treatment cycles.


Subject(s)
Antineoplastic Agents/pharmacology , Diphtheria Toxin/pharmacology , Immunotoxins/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Lymphoma, B-Cell/drug therapy , Animals , Antibodies, Neutralizing/immunology , Antigens, CD19/immunology , Cell Line, Tumor , Diphtheria Toxin/genetics , Humans , Immunotoxins/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Lymphoma, B-Cell/immunology , Mice, Inbred BALB C , Sialic Acid Binding Ig-like Lectin 2/immunology
5.
Mol Cancer Ther ; 16(5): 956-965, 2017 05.
Article in English | MEDLINE | ID: mdl-28193671

ABSTRACT

Sarcomas differ from carcinomas in their mesenchymal origin. Therapeutic advancements have come slowly, so alternative drugs and models are urgently needed. These studies report a new drug for sarcomas that simultaneously targets both tumor and tumor neovasculature. eBAT is a bispecific angiotoxin consisting of truncated, deimmunized Pseudomonas exotoxin fused to EGF and the amino terminal fragment of urokinase. Here, we study the drug in an in vivo "ontarget" companion dog trial as eBAT effectively kills canine hemangiosarcoma and human sarcoma cells in vitro We reasoned the model has value due to the common occurrence of spontaneous sarcomas in dogs and a limited lifespan allowing for rapid accrual and data collection. Splenectomized dogs with minimal residual disease were given one cycle of eBAT followed by adjuvant doxorubicin in an adaptive dose-finding, phase I-II study of 23 dogs with spontaneous, stage I-II, splenic hemangiosarcoma. eBAT improved 6-month survival from <40% in a comparison population to approximately 70% in dogs treated at a biologically active dose (50 µg/kg). Six dogs were long-term survivors, living >450 days. eBAT abated expected toxicity associated with EGFR targeting, a finding supported by mouse studies. Urokinase plasminogen activator receptor and EGFR are targets for human sarcomas, so thorough evaluation is crucial for validation of the dog model. Thus, we validated these markers for human sarcoma targeting in the study of 212 human and 97 canine sarcoma samples. Our results support further translation of eBAT for human patients with sarcomas and perhaps other EGFR-expressing malignancies. Mol Cancer Ther; 16(5); 956-65. ©2017 AACR.


Subject(s)
ErbB Receptors/genetics , Hemangiosarcoma/drug therapy , Molecular Targeted Therapy , Receptors, Urokinase Plasminogen Activator/genetics , ADP Ribose Transferases/administration & dosage , ADP Ribose Transferases/chemistry , ADP Ribose Transferases/genetics , Animals , Bacterial Toxins/administration & dosage , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Cell Line, Tumor , Disease Models, Animal , Dogs , Doxorubicin/administration & dosage , Epidermal Growth Factor/chemistry , Epidermal Growth Factor/genetics , ErbB Receptors/antagonists & inhibitors , Exotoxins/administration & dosage , Exotoxins/chemistry , Exotoxins/genetics , Hemangiosarcoma/genetics , Hemangiosarcoma/pathology , Humans , Mice , Neoplasm Staging , Receptors, Urokinase Plasminogen Activator/antagonists & inhibitors , Urokinase-Type Plasminogen Activator/chemistry , Urokinase-Type Plasminogen Activator/genetics , Virulence Factors/administration & dosage , Virulence Factors/chemistry , Virulence Factors/genetics , Pseudomonas aeruginosa Exotoxin A
6.
Oncotarget ; 7(45): 73830-73844, 2016 Nov 08.
Article in English | MEDLINE | ID: mdl-27650544

ABSTRACT

BACKGROUND: The design of a highly effective anti-cancer immune-engager would include targeting of highly drug refractory cancer stem cells (CSC). The design would promote effective antibody-dependent cell-mediated cytotoxicity (ADCC) and simultaneously promote costimulation to expand and self-sustain the effector NK cell population. Based on our bispecific NK cell engager platform we constructed a tetraspecific killer engager (TetraKE) comprising single-chain variable fragments (scFvs) binding FcγRIII (CD16) on NK cells, EpCAM on carcinoma cells and CD133 on cancer stem cells in order to promote ADCC. Furthermore, an Interleukin (IL)-15-crosslinker enhanced NK cell related proliferation resulting in a highly active drug termed 1615EpCAM133. RESULTS: Proliferation assays showed TetraKE promoted proliferation and enhanced NK cell survival. Drug-target binding, NK cell related degranulation, and IFN-γ production was specific for both tumor related antigens in EpCAM and CD133 bearing cancer cell lines. The TetraKE showed higher killing activity and superior dose dependent degranulation. Cytokine profiling showed a moderately enhanced IFN-γ production, enhanced GM-CSF production, but no evidence of induction of excessive cytokine release. METHODS: Assembly and synthesis of hybrid genes encoding the TetraKE were performed using DNA shuffling and ligation. The TetraKE was tested for efficacy, specificity, proliferation, survival, and cytokine production using carcinoma cell lines and functional assays measuring NK cell activity. CONCLUSION: 1615EpCAM133 combines improved induction of ADCC with enhanced proliferation, limited cytokine response, and prolonged survival and proliferation of NK cells. By linking scFv-related targeting of carcinoma and CSCs with a sustaining IL-15 signal, our new construct shows great promise to target cancer and CSCs.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , Interleukin-15/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology , AC133 Antigen/genetics , AC133 Antigen/metabolism , Biomarkers , Cell Degranulation/immunology , Cell Line, Tumor , Cytokines/biosynthesis , Epithelial Cell Adhesion Molecule/genetics , Epithelial Cell Adhesion Molecule/metabolism , Humans , Interferon-gamma/biosynthesis , Interleukin-15/genetics , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , Protein Binding/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism
7.
Toxins (Basel) ; 7(10): 4067-82, 2015 Oct 10.
Article in English | MEDLINE | ID: mdl-26473923

ABSTRACT

BACKGROUND: Targeted toxins require multiple treatments and therefore must be deimmunized. We report a method of protein deimmunization based on the point mutation of highly hydrophilic R, K, D, E, and Q amino acids on the molecular surface of truncated diphtheria-toxin (DT390). METHODS: Based on their surface position derived from an X-ray-crystallographic model, residues were chosen for point mutation that were located in prominent positions on the molecular surface and away from the catalytic site. Mice were immunized with a targeted toxin containing either a mutated DT390 containing seven critical point mutations or the non-mutated parental toxin form. RESULTS: Serum analysis revealed a significant 90% reduction in anti-toxin antibodies in mice immunized with the mutant, but not the parental drug form despite multiple immunizations. The experiment was repeated in a second strain of mice with a different MHC-haplotype to address whether point mutation removed T or B cell epitopes. Findings were identical indicating that B cell epitopes were eliminated from DT. The mutant drug form lost only minimal activity in vitro as well as in vivo. CONCLUSION: These findings indicate that this method may be effective for deimmunizing of other proteins and that discovery of a deimmunized form of DT may lead to the development of more effective targeted toxin.


Subject(s)
Antineoplastic Agents/pharmacology , Diphtheria Toxin/genetics , Diphtheria Toxin/immunology , Drug Discovery/methods , Point Mutation , Recombinant Fusion Proteins/pharmacology , Animals , Antibodies, Neutralizing/blood , Antineoplastic Agents/immunology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , Humans , Immunoglobulin G/blood , Inhibitory Concentration 50 , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Plasmids , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/therapeutic use , Xenograft Model Antitumor Assays
8.
Mol Cancer Ther ; 11(12): 2674-84, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23075808

ABSTRACT

This study evaluates the mechanism by which bispecific and trispecific killer cell engagers (BiKEs and TriKEs) act to trigger human natural killer (NK) cell effector function and investigates their ability to induce NK cell cytokine and chemokine production against human B-cell leukemia. We examined the ability of BiKEs and TriKEs to trigger NK cell activation through direct CD16 signaling, measuring intracellular Ca²âº mobilization, secretion of lytic granules, induction of target cell apoptosis, and production of cytokine and chemokines in response to the Raji cell line and primary leukemia targets. Resting NK cells triggered by the recombinant reagents led to intracellular Ca²âº mobilization through direct CD16 signaling. Coculture of reagent-treated resting NK cells with Raji targets resulted in significant increases in NK cell degranulation and target cell death. BiKEs and TriKEs effectively mediated NK cytotoxicity of Raji targets at high and low effector-to-target ratios and maintained functional stability after 24 and 48 hours of culture in human serum. NK cell production of IFN-γ, TNF-α, granulocyte macrophage colony-stimulating factor (GM-CSF), interleukin (IL)-8, macrophage inflammatory protein (MIP)-1α, and regulated and normal T cell expressed and secreted (RANTES) was differentially induced in the presence of recombinant reagents and Raji targets. Moreover, significant increases in NK cell degranulation and enhancement of IFN-γ production against primary acute lymphoblastic leukemia and chronic lymphocytic leukemia targets were induced with reagent treatment of resting NK cells. In conclusion, BiKEs and TriKEs directly trigger NK cell activation through CD16, significantly increasing NK cell cytolytic activity and cytokine production against tumor targets, showing their therapeutic potential for enhancing NK cell immunotherapies for leukemias and lymphomas.


Subject(s)
Cytokines/immunology , Immunotherapy, Adoptive/methods , Killer Cells, Natural/immunology , Receptors, IgG/immunology , Antibodies, Bispecific/genetics , Antibodies, Bispecific/immunology , Antibodies, Bispecific/metabolism , Antigens, CD19/genetics , Antigens, CD19/immunology , Antigens, CD19/metabolism , Burkitt Lymphoma/immunology , Burkitt Lymphoma/therapy , Cell Line, Tumor , Cytokines/biosynthesis , Cytotoxicity, Immunologic , Epitopes/immunology , Humans , Immunoglobulin Fragments/genetics , Immunoglobulin Fragments/immunology , Lymphocyte Activation/immunology , Receptors, IgG/genetics , Receptors, IgG/metabolism , Signal Transduction
9.
Pancreas ; 41(5): 789-96, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22258068

ABSTRACT

OBJECTIVE: The objective was to test a bispecific ligand-directed toxin (BLT), with reduced immunogenicity for enhanced efficacy in targeting orthotopic pancreatic cancer in vivo. METHOD: A new BLT was created in which both human epidermal growth factor (EGF) and interleukin 4 cytokines were cloned onto the same single chain molecule with deimmunized pseudomonas exotoxin (dEGF4KDEL). Key amino acids dictating B-cell generation of neutralizing antitoxin antibodies were mutated. Bioassays were used to determine whether mutation reduced potency, and enzyme-linked immunosorbent assay studies were performed to determine whether antitoxin antibodies were reduced. A genetically altered luciferase MIA PaCa-2 xenograft model was used to image in real time and determine effects on systemic malignant human cancer. Bispecific ligand-directed toxins targeting B cells were used as specificity controls. RESULTS: Deimmunized EGF4KDEL was significantly effective after systemic injection against established orthotopic MIA PaCa-2 pancreatic cancer and selectively prevented metastasis. Mutagenesis significantly reduced antitoxin levels in vivo with no apparent activity loss in vitro. The drug was effective against 3 human pancreatic cancer lines in vitro, MIA PaCa-2, SW1990, and S2VP10. CONCLUSIONS: Despite the metastatic nature of the MIA PaCa-2 orthotopic tumor xenografted in nude mice, high percentages of tumors responded to extended dEGFKDEL treatment resulting in significant anticancer effects and disease-free survivors.


Subject(s)
Antineoplastic Agents/pharmacology , Immunotoxins/pharmacology , Pancreatic Neoplasms/drug therapy , Xenograft Model Antitumor Assays , Amino Acid Sequence , Animals , Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , Antineoplastic Agents/immunology , Cell Line, Tumor , Epidermal Growth Factor/genetics , Epidermal Growth Factor/immunology , Exotoxins/genetics , Exotoxins/immunology , Humans , Immunotoxins/genetics , Immunotoxins/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Ligands , Male , Mice , Mice, Nude , Mutation , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Pseudomonas/genetics , Pseudomonas/immunology , Time Factors , Tumor Burden/drug effects , Tumor Burden/immunology
10.
J Neurooncol ; 95(3): 331-342, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19517064

ABSTRACT

A bispecific ligand-directed toxin (BLT) consisting of human interleukin-13, epithelial growth factor, and the first 389 amino acids of diphtheria toxin was assembled in order to target human glioblastoma. In vitro, DTEGF13 selectively killed the human glioblastoma cell line U87-luc as well as other human glioblastomas. DTEGF13 fulfilled the requirement of a successful BLT by having greater activity than either of its monospecific counterparts or their mixture proving it necessary to have both ligands on the same single chain molecule. Aggressive brain tumors established intracranially (IC) in nude rats with U87 glioma genetically marked with a firefly luciferase reporter gene were treated with two injections of DTEGF13 using convection enhanced delivery resulting in tumor eradication in 50% of the rats which survived with tumor free status at least 110 days post tumor inoculation. An irrelevant BLT control did not protect establishing specificity. The bispecific DTEGF13 MTD dose was measured at 2 microg/injection or 0.5 microg/kg and toxicity studies indicated safety in this dose. Combination of monospecific DTEGF and DTIL13 did not inhibit tumor growth. ELISA assay indicated that anti-DT antibodies were not generated in normal immunocompetent rats given identical intracranial DTEGF13 therapy. Thus, DTEGF13 is safe and efficacious as an alternative drug for glioblastoma therapy and warrants further study.


Subject(s)
Brain Neoplasms/drug therapy , Drug Delivery Systems/methods , Glioblastoma/drug therapy , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/toxicity , Animals , Antibody Specificity , Brain Neoplasms/pathology , Cell Division/drug effects , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Genes, Reporter , Glioblastoma/pathology , Humans , Immunoglobulin G/blood , Ligands , Liver/drug effects , Luciferases/genetics , Neoplasm Transplantation , Rats , Rats, Nude , Recombinant Fusion Proteins/immunology , Xenograft Model Antitumor Assays
11.
Int J Cancer ; 120(2): 411-9, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17075792

ABSTRACT

A gene splicing technique was used to create a hybrid fusion protein DTAT encoding the 390 amino acid portion of diphtheria toxin (DT(390)), a linker, and the downstream 135-amino terminal fragment portion of human urokinase plasminogen activator. DTAT was assembled to target human glioblastoma cell lines in a murine intracranial model. Previously published in vitro studies demonstrated that DTAT was highly selective and toxic to human glioblastoma cell lines in a flank tumor model. The purpose of this study was to determine the toxicity, specificity and possible therapeutic efficacy of DTAT in an intracranial model. Convection enhanced delivery of DTAT resulted in about a 16-fold increase in maximum tolerated dose. Intracranial administration of DTAT on an every-other-day basis in nude mice with established U87 MG brain tumors resulted in significant reductions in tumor volume and significantly prolonged survival (p < 0.0001). Magnetic resonance imaging proved to be a powerful tool in mice and rats for demonstrating tumor growth in a xenograft intracranial model, assessing the efficacy of DTAT in tumor volume reduction and detecting DTAT-associated intracranial toxicity and vascular damage. These results suggest that the DTAT recombinant fusion protein is highly effective in an intracranial model and DTAT might be an effective treatment for glioblastoma.


Subject(s)
Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Recombinant Fusion Proteins/therapeutic use , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Female , Glioblastoma/pathology , Humans , Injections , Kidney/pathology , Liver/pathology , Magnetic Resonance Imaging , Mice , Mice, Nude , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/toxicity , Xenograft Model Antitumor Assays
12.
Int J Cancer ; 118(10): 2594-601, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16358262

ABSTRACT

A fusion protein consisting of human interleukin-13 and the first 389 amino acids of diphtheria toxin was assembled in order to target human glioblastoma cell lines in a murine intracranial model. In vitro studies to determine specificity indicated that the protein called DTIL13 was highly selective for human glioblastoma. In vivo, the maximum tolerated dose of DTIL13 was 1 microg/injection given every other day and repeated for 3 days. Doses that exceeded this amount resulted in weight loss and liver damage as determined by histology and enzyme assay. Experiments in IL-4 receptor knockout mice revealed that liver toxicity was receptor-related. This same dose given to nude mice with established U373 MG brain tumors resulted in significant reductions in tumor volume and significantly prolonged survival (p<0.0001). Magnetic resonance imaging (MRI) proved to be extremely useful in (i) determining the ability of DTIL13 to reduce tumor size and (ii) for studying toxicity since diffusion-weighted and gradient echo-weighted MRI revealed that vascular leak syndrome was not a limiting toxicity at this dose. These results suggest that DTIL13 is as effective in an intracranial rodent model as it was in a flank model in previous studies and that DTIL13 might be an effective treatment for glioblastoma multiforme.


Subject(s)
Brain Neoplasms/drug therapy , Diphtheria Toxin/therapeutic use , Glioblastoma/drug therapy , Interleukin-13/therapeutic use , Animals , Diffusion Magnetic Resonance Imaging , Diphtheria Toxin/adverse effects , Diphtheria Toxin/chemistry , Female , Humans , Interleukin-13/adverse effects , Interleukin-13/chemistry , Interleukin-4/genetics , Maximum Tolerated Dose , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptide Fragments , Treatment Outcome
13.
J Neurooncol ; 77(3): 257-66, 2006 May.
Article in English | MEDLINE | ID: mdl-16314943

ABSTRACT

DTAT13, a novel recombinant bispecific immunotoxin (IT) consisting of truncated diphtheria toxin, an amino-terminal (AT) fragment of the urokinase-type plasminogen activator (uPa), and a fragment of human IL-13 was assembled in order to target receptors on glioblastoma multiforme (GBM) and its associated neovasculature. Previous in vitro studies confirmed the efficacy of DTAT13 against various GBM cell lines expressing both IL-13 receptor or uPA receptor, and previous in vivo testing demonstrated the efficacy of DTAT13 in significantly inhibiting a range of xenograft tumors and showed that DTAT13 was 160- and 8-fold less toxic to the parental fusion IT, DTAT and DTIL13, respectively. To further understand the properties of DTAT13, pharmacokinetic/biodistribution experiments were performed. Binding analysis revealed that the IL-13 domain functioned independently of the uPA domain and that the K (d) for each binding domain was essentially the same as that of DTIL13 and DTAT. Flow cytometry studies indicated that DTAT13 bound better than DTAT or DTIL13. Analysis of the rate of protein synthesis inhibition in U87 MG cells by DTAT13 compared to DTAT revealed a faster rate of inhibition with DTAT13 compared to DTAT. The rate of protein synthesis inhibition of DTAT13 was identical to that of DTIL13 in U373 MG cells. Intracranial biodistribution studies revealed that DTAT13 was able to cross to the contralateral hemisphere unlike DTIL13 but similar to DTAT. These studies show that DTAT13 has properties encompassing those of both DTIL13 and DTAT and warrants further consideration for clinical development.


Subject(s)
Diphtheria Toxin/pharmacokinetics , Glioblastoma/metabolism , Immunotoxins/pharmacokinetics , Interleukin-13/pharmacokinetics , Neovascularization, Pathologic/immunology , Urokinase-Type Plasminogen Activator/pharmacokinetics , Animals , Brain/metabolism , Diphtheria Toxin/therapeutic use , Female , Glioblastoma/blood supply , Glioblastoma/drug therapy , Glioblastoma/immunology , Humans , Immunotoxins/therapeutic use , Interleukin-13/therapeutic use , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Protein Biosynthesis/drug effects , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/therapeutic use , Urokinase-Type Plasminogen Activator/therapeutic use
14.
Clin Cancer Res ; 11(10): 3879-88, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15897589

ABSTRACT

A novel bispecific single-chain fusion protein, DT2219, was assembled consisting of the catalytic and translocation domains of diphtheria toxin (DT(390)) fused to two repeating sFv subunits recognizing CD19 and CD22 and expressed in Escherichia coli. Problems with yield, purity, and aggregation in the refolding step were solved by incorporating a segment of human muscle aldolase and by using a sodium N-lauroyl-sarcosine detergent-based refolding procedure. Problems with reduced efficacy were addressed by combining the anti-CD19 and anti-CD22 on the same single-chain molecule. DT2219 had greater anticancer activity than monomeric or bivalent immunotoxins made with anti-CD19 and anti-CD22 sFv alone and it showed a higher level of binding to patient leukemia cells and to CD19(+)CD22(+) Daudi or Raji cells than did anti-CD19 and anti-CD22 parental monoclonal antibodies. The resulting DT2219, mutated to enhance its avidity, was cytotoxic to Daudi cells in vitro (IC(50) = 0.3 nmol/L). In vivo, DT2219 was effective in a flank tumor therapy model in which it significantly inhibited tumor growth (P < 0.05) and in a systemic model in which it significantly prolonged survival of severe combined immunodeficient mice with established Daudi (P < 0.008) compared with controls. DT2219 has broader reactivity in recognizing B-cell malignancies, has more killing power, and requires less toxin than using individual immunotoxin, which warrants further investigation as a new drug for treating B leukemia/lymphoma.


Subject(s)
Antigens, CD19/immunology , Antigens, CD/immunology , Antigens, Differentiation, B-Lymphocyte/immunology , Cell Adhesion Molecules/immunology , Immunotoxins/immunology , Immunotoxins/pharmacology , Lectins/immunology , Animals , Antibodies , Cell Death , Diphtheria Toxin/immunology , Escherichia coli/genetics , Escherichia coli/immunology , Female , Leukemia, B-Cell , Lymphoma, B-Cell , Mice , Mice, Nude , Mice, SCID , Molecular Conformation , Sialic Acid Binding Ig-like Lectin 2 , Survival Analysis , Transplantation, Heterologous , Tumor Cells, Cultured
15.
Leuk Res ; 29(3): 331-41, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15661270

ABSTRACT

A novel bivalent single chain fusion protein, Bic3, was assembled consisting of the catalytic and translocation domains of diphtheria toxin (DT(390)) fused to two repeating sFv molecules recognizing human CD3 epsilon of the human T-cell receptor. Historically, problems with these constructs include low yield, toxicity, and reduced efficacy. Instead of using conventional Gly(4)Ser linkers to connect heavy/light chains, aggregation reducing linkers (ARL) were used which when combined with a new SLS-based refolding method reduced aggregation and enhanced the yield of final product. Toxicity was reduced at least 25-fold by repeating the two sFv molecules and adding a portion of the hinge-CH2-CH3 human constant regions. The resulting Bic3 was just as cytotoxic to HPB-MLT.UM T leukemia cells in vitro (IC(50)=4 pmol) as a monovalent construct made with the same DT and sFv. In vivo, Bic3 was effective in a new and aggressive therapy model in which it significantly prolonged survival of scid mice with established human T-cell leukemia (p<0.0001 compared to controls). Importantly, no toxicity measured by weight loss, enzyme function, or histology was observed at the highest dose of Bic3 tested (2000 ug/kg). Bic3 warrants investigation as a new drug for treating T-cell malignancy and other T-cell related disorders.


Subject(s)
Immunotoxins/adverse effects , Immunotoxins/chemistry , Leukemia, T-Cell/drug therapy , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/chemical synthesis , Animals , Antibodies, Monoclonal , CD3 Complex/immunology , Disease Models, Animal , Humans , Immunoglobulin G , Kidney/pathology , Mice , Mice, SCID , Receptors, Antigen, T-Cell , Recombinant Fusion Proteins/chemistry , T-Lymphocytes/drug effects
16.
Protein Eng Des Sel ; 17(2): 157-64, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15047912

ABSTRACT

A bispecific immunotoxin (IT) called DTAT13 was synthesized in order to target simultaneously the urokinase-type plasminogen activator receptor (uPAR)-expressing tumor neovasculature and IL-13 receptor expressing glioblastoma cells with the goal of intratumoral administration for brain tumors. The recombinant hybrid was created using the non-internalizing N-terminal fragment (ATF) of uPA and the IL-13 molecule for binding plus the catalytic and translocation portion of diphtheria toxin (DT) for killing. The 71 kDa protein was highly selective for human glioblastoma in vitro showing no loss on binding compared with DTAT and DTIL13 controls. In vivo, DTAT13 caused the regression of small tumors when administered at 10 micro g/day given on a five-dose schedule every other day. DTAT13 was able to target both overexpressed uPAR and the vasculature, as demonstrated by its ability to kill HUVEC cells. Also, mortality studies indicated that DTAT13 was less toxic than DTAT or DTIL13. These findings indicate that bispecific IT may allow treatment of a broader subset of antigenically diverse patients while simultaneously reducing the exposure to toxin required than if two separate agents were employed.


Subject(s)
Immunotoxins/immunology , Immunotoxins/pharmacology , Receptors, Cell Surface/immunology , Receptors, Interleukin/metabolism , Xenograft Model Antitumor Assays , Animals , Antibodies, Bispecific , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Binding Sites , Cell Division/drug effects , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Female , Glioblastoma/drug therapy , Humans , Immunotoxins/genetics , Interleukin-13/genetics , Interleukin-13/metabolism , Interleukin-13 Receptor alpha1 Subunit , Kidney/drug effects , Mice , Mice, Nude , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Peptide Fragments , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Interleukin-13 , Receptors, Urokinase Plasminogen Activator , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Toxicity Tests
17.
Protein Eng ; 15(5): 419-27, 2002 May.
Article in English | MEDLINE | ID: mdl-12034862

ABSTRACT

Fusion proteins composed of tumor binding agents and potent catalytic toxins show promise for intracranial therapy of brain cancer and an advantage over systemic therapy. Glioblastoma multiforme (GBM) is the most common form of brain cancer and overexpresses IL-13R. Thus, we developed an interleukin-13 receptor targeting fusion protein, DT(390)IL13, composed of human interleukin-13 and the first 389 amino acids of diphtheria toxin. To measure its ability to inhibit GBM, DT(390)IL13 was tested in vitro and found to inhibit selectively the U373 MG GBM cell line with an IC(50) around 12 pmol/l. Cytotoxicity was neutralized by anti-human-interleukin-13 antibody, but not by control antibodies. In vivo, small U373 MG glioblastoma xenografts in nude mice completely regressed in most animals after five intratumoral injections of 1 microg of DT(390)IL13 q.o.d., but not by the control fusion protein DT(390)IL-2. DT(390)IL13 was also tested against primary explant GBM cells of a patient's excised tumor and the IC(50) was similar to that measured for U373 MG. Further studies showed a therapeutic window for DT(390)IL13 of 1-30 microg/injection and histology studies and enzyme measurements showed that the maximum tolerated dose of DT(390)IL13 had little effect on kidney, liver, spleen, lung and heart in non-tumor-bearing immunocompetent mice. Together, these data suggest that DT(390)IL13 may provide an important, alternative therapy for brain cancer.


Subject(s)
Antineoplastic Agents/administration & dosage , Diphtheria Toxin/administration & dosage , Glioblastoma/drug therapy , Interleukin-13/administration & dosage , Animals , Antineoplastic Agents/therapeutic use , Diphtheria Toxin/therapeutic use , Drug Delivery Systems , Drug Screening Assays, Antitumor , Female , In Vitro Techniques , Interleukin-13/therapeutic use , Interleukin-13 Receptor alpha1 Subunit , Kinetics , Maximum Tolerated Dose , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasm Transplantation , Receptors, Interleukin/metabolism , Receptors, Interleukin-13 , Recombinant Fusion Proteins/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...