Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 509
Filter
1.
Proc Natl Acad Sci U S A ; 119(24): e2200200119, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35675429

ABSTRACT

The human transferrin receptor (TFR) is overexpressed in most breast cancers, including preneoplastic ductal carcinoma in situ (DCIS). HB21(Fv)-PE40 is a single-chain immunotoxin (IT) engineered by fusing the variable region of a monoclonal antibody (HB21) against a TFR with a 40 kDa fragment of Pseudomonas exotoxin (PE). In humans, the administration of other TFR-targeted immunotoxins intrathecally led to inflammation and vascular leakage. We proposed that for treatment of DCIS, intraductal (i.duc) injection of HB21(Fv)-PE40 could avoid systemic toxicity while retaining its potent antitumor effects on visible and occult tumors in the entire ductal tree. Pharmacokinetic studies in mice showed that, in contrast to intravenous injection, IT was undetectable by enzyme-linked immunosorbent assay in blood following i.duc injection of up to 3.0 µg HB21(Fv)-PE40. We demonstrated the antitumor efficacy of HB21(Fv)-PE40 in two mammary-in-duct (MIND) models, MCF7 and SUM225, grown in NOD/SCID/gamma mice. Tumors were undetectable by In Vivo Imaging System (IVIS) imaging in intraductally treated mice within 1 wk of initiation of the regimen (IT once weekly/3 wk, 1.5 µg/teat). MCF7 tumor-bearing mice remained tumor free for up to 60 d of observation with i.duc IT, whereas the HB21 antibody alone or intraperitoneal IT treatment had minimal/no antitumor effects. These and similar findings in the SUM225 MIND model were substantiated by analysis of mammary gland whole mounts, histology, and immunohistochemistry for the proteins Ki67, CD31, CD71 (TFR), and Ku80. This study provides a strong preclinical foundation for conducting feasibility and safety trials in patients with stage 0 breast cancer.


Subject(s)
ADP Ribose Transferases , Bacterial Toxins , Breast Neoplasms , Carcinoma, Intraductal, Noninfiltrating , Exotoxins , Immunotoxins , Molecular Targeted Therapy , Receptors, Transferrin , Virulence Factors , ADP Ribose Transferases/administration & dosage , ADP Ribose Transferases/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Bacterial Toxins/administration & dosage , Breast Neoplasms/therapy , Carcinoma, Intraductal, Noninfiltrating/therapy , Exotoxins/administration & dosage , Female , Humans , Immunotoxins/administration & dosage , MCF-7 Cells , Mice , Mice, Inbred NOD , Mice, SCID , Receptors, Transferrin/metabolism , Virulence Factors/administration & dosage , Pseudomonas aeruginosa Exotoxin A
2.
Proc Natl Acad Sci U S A ; 118(29)2021 07 20.
Article in English | MEDLINE | ID: mdl-34253615

ABSTRACT

We investigated the role of mesothelin (Msln) and thymocyte differentiation antigen 1 (Thy1) in the activation of fibroblasts across multiple organs and demonstrated that Msln-/- mice are protected from cholestatic fibrosis caused by Mdr2 (multidrug resistance gene 2) deficiency, bleomycin-induced lung fibrosis, and UUO (unilateral urinary obstruction)-induced kidney fibrosis. On the contrary, Thy1-/- mice are more susceptible to fibrosis, suggesting that a Msln-Thy1 signaling complex is critical for tissue fibroblast activation. A similar mechanism was observed in human activated portal fibroblasts (aPFs). Targeting of human MSLN+ aPFs with two anti-MSLN immunotoxins killed fibroblasts engineered to express human mesothelin and reduced collagen deposition in livers of bile duct ligation (BDL)-injured mice. We provide evidence that antimesothelin-based therapy may be a strategy for treatment of parenchymal organ fibrosis.


Subject(s)
Cholestasis/drug therapy , Fibroblasts/immunology , Immunotherapy , Liver Cirrhosis/drug therapy , Animals , Cholestasis/genetics , Cholestasis/immunology , Collagen/immunology , Fibroblasts/drug effects , Humans , Immunotoxins/administration & dosage , Liver Cirrhosis/genetics , Liver Cirrhosis/immunology , Mesothelin/genetics , Mesothelin/immunology , Mice , Thy-1 Antigens/genetics , Thy-1 Antigens/immunology
3.
Sci Rep ; 11(1): 14925, 2021 07 21.
Article in English | MEDLINE | ID: mdl-34290365

ABSTRACT

Prostate cancer cells are characterized by a remarkably low proliferative rate and the production of high levels of prostate-specific proteases. Protein-based toxins are attractive candidates for prostate cancer therapy because they kill cells via proliferation-independent mechanisms. However, the non-specific cytotoxicity of these potent cytotoxins must be redirected to avoid toxicity to normal tissues. Prostate-Specific Membrane Antigen (PSMA) is membrane-bound carboxypeptidase that is highly expressed by prostate cancer cells. Potent dipeptide PSMA inhibitors have been developed that can selectively deliver and concentrate imaging agents within prostate cancer cells based on continuous PSMA internalization and endosomal cycling. On this basis, we conjugated a PSMA inhibitor to the apoptosis-inducing human protease Granzyme B and the potent Pseudomonas exotoxin protein toxin fragment, PE35. We assessed selective PSMA binding and entrance into tumor cell to induce cell death. We demonstrated these agents selectively bound to PSMA and became internalized. PSMA-targeted PE35 toxin was selectively toxic to PSMA producing cells in vitro. Intratumoral and intravenous administration of this toxin produced marked tumor killing of PSMA-producing xenografts with minimal host toxicity. These studies demonstrate that urea-based PSMA inhibitors represent a simpler, less expensive alternative to antibodies as a means to deliver cytotoxic proteins to prostate cancer cells.


Subject(s)
Drug Delivery Systems , Immunotoxins/administration & dosage , Kallikreins , Prostate-Specific Antigen , Prostatic Neoplasms/drug therapy , Urea , Carboxypeptidases/metabolism , Cell Line, Tumor , Humans , Kallikreins/antagonists & inhibitors , Kallikreins/metabolism , Male , Prostate-Specific Antigen/antagonists & inhibitors , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/metabolism
4.
Immunopharmacol Immunotoxicol ; 43(2): 230-238, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33657977

ABSTRACT

Neuropilin-1 (NRP-1) regulates a range of physiological and pathological processes, including angiogenesis. Targeting of NRP1 is considered a significant approach in cancer therapy. In the present study, a novel antiNRP1 immunotoxin (αNRP1 IT) was developed by genetic fusion of a single domain (VHH) anti-NRP-1 antibody fragment to a truncated diphtheria toxin. The αNRP1 IT was expressed into bacterial cells as an inclusion body (IB). Expression of αNRP1 IT was confirmed by SDS-PAGE and western blotting. Recombinant αNRP1 IT was purified using nickel affinity chromatography. Toxicity and antiangiogenesis effect of αNRP1 IT was investigated both in vitro and in vivo. Results showed that αNRP1 IT significantly reduced the viability of human umbilical vein endothelial cell line (HUVEC) (p < .05). The αNRP1 IT significantly inhibited tube formation of HUVEC cells (p < .001). Furthermore, αNRP1 IT inhibited angiogenesis in Chick Chorioallantoic Membrane (CAM) Assay. These data suggest the potential of αNRP1 IT as a novel therapeutic in targeted cancer therapy.


Subject(s)
Diphtheria Toxin/administration & dosage , Human Umbilical Vein Endothelial Cells/drug effects , Immunotoxins/administration & dosage , Neovascularization, Pathologic/prevention & control , Neuropilin-1/antagonists & inhibitors , Single-Domain Antibodies/administration & dosage , Animals , Camelus , Cell Survival/drug effects , Cell Survival/immunology , Chickens , Dose-Response Relationship, Drug , HEK293 Cells , Human Umbilical Vein Endothelial Cells/immunology , Humans , MCF-7 Cells , Male , Neovascularization, Pathologic/immunology , Neuropilin-1/immunology
5.
Life Sci ; 265: 118866, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33301810

ABSTRACT

AIMS: GnRH-DFF40 (gonadotropin releasing hormone-DNA fragmentation factor 40) humanized recombinant immunotoxin serves as a prospective candidate for targeted therapy of malignancies with over-expressed gonadotropin releasing hormone receptor (GnRHR). In this study, we attempted to generate a GnRH-based chimeric protein composed of human DFF40 fused with GnRH which encodes an apoptotic nuclease and specifically targets cancer cells displaying GnRH receptor overexpression. MATERIALS AND METHODS: A codon optimized, synthetic GnRH-DFF40 fusion gene and its single counterpart (DFF40) were constructed in pET28a expression vector. Cytotoxicity of these expressed proteins were evaluated on three breast cancer cell lines (MCF7, MDA-MB231, and SKBR3). The stability and biological activity of the recombinant proteins were investigated in the treated cell line and cell-free system. Also, the ability of this fusion and its single form in inducing apoptosis, and inhibiting metastasis and migration were evaluated by flow cytometry, migration assay and wound healing analysis, respectively. In silico analyses were also done to understand the specific interactions between GnRH and its receptor. KEY FINDINGS: GnRH-DFF40 fusion protein and DFF40 were successfully expressed. The purified chimeric protein showed dose-dependent cytotoxicity against all three cell lines. The recombinant fusion protein was biologically active with nucleolytic functionality and apoptosis induction ability. Moreover, the fusion could inhibit the invasion property of MDA-MB-231 cells. In silico analysis also showed that four residues from GnRH domain and 11 GnRHR residues had the most interaction sites for specific targeted delivery of the immunotoxin in cancer cells. SIGNIFICANCE: Fusion construct could be a prospective candidate for targeted therapy of cancers upregulating GnRH receptor.


Subject(s)
Breast Neoplasms/therapy , Deoxyribonucleases/genetics , Immunotoxins/pharmacology , Poly-ADP-Ribose Binding Proteins/genetics , Receptors, LHRH/genetics , Recombinant Fusion Proteins/pharmacology , Apoptosis/physiology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell-Free System , Computer Simulation , Dose-Response Relationship, Drug , Female , Flow Cytometry , Humans , Immunotoxins/administration & dosage , MCF-7 Cells , Molecular Targeted Therapy , Recombinant Fusion Proteins/administration & dosage
6.
Proc Natl Acad Sci U S A ; 117(36): 22090-22100, 2020 09 08.
Article in English | MEDLINE | ID: mdl-32839344

ABSTRACT

The application of proteinaceous toxins for cell ablation is limited by their high on- and off-target toxicity, severe side effects, and a narrow therapeutic window. The selectivity of targeting can be improved by intein-based toxin reconstitution from two dysfunctional fragments provided their cytoplasmic delivery via independent, selective pathways. While the reconstitution of proteins from genetically encoded elements has been explored, exploiting cell-surface receptors for boosting selectivity has not been attained. We designed a robust splitting algorithm and achieved reliable cytoplasmic reconstitution of functional diphtheria toxin from engineered intein-flanked fragments upon receptor-mediated delivery of one of them to the cells expressing the counterpart. Retargeting the delivery machinery toward different receptors overexpressed in cancer cells enables selective ablation of specific subpopulations in mixed cell cultures. In a mouse model, the transmembrane delivery of a split-toxin construct potently inhibits the growth of xenograft tumors expressing the split counterpart. Receptor-mediated delivery of engineered split proteins provides a platform for precise therapeutic and experimental ablation of tumors or desired cell populations while also greatly expanding the applicability of the intein-based protein transsplicing.


Subject(s)
Bacterial Toxins/administration & dosage , Bacterial Toxins/chemistry , Cytoplasm/metabolism , Drug Delivery Systems/methods , Inteins , Neoplasms/drug therapy , Animals , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Cell Line, Tumor , Cytoplasm/genetics , Diphtheria Toxin/administration & dosage , Diphtheria Toxin/chemistry , Diphtheria Toxin/genetics , Diphtheria Toxin/metabolism , Female , Heterografts , Humans , Immunotoxins/administration & dosage , Immunotoxins/chemistry , Immunotoxins/genetics , Immunotoxins/metabolism , Mice , Mice, Nude , Neoplasms/genetics , Neoplasms/metabolism , Protein Domains , Protein Transport , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism
7.
Sci Rep ; 10(1): 12255, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32703972

ABSTRACT

Immunotoxins appear as promising therapeutic molecules, alternative to allergen-specific-immunotherapy. In this work, we achieved the development of a protein chimera able to promote specific cell death on effector cells involved in the allergic reaction. Der p 1 allergen was chosen as cell-targeting domain and the powerful ribotoxin α-sarcin as the toxic moiety. The resultant construction, named proDerp1αS, was produced and purified from the yeast Pichia pastoris. Der p 1-protease activity and α-sarcin ribonucleolytic action were effectively conserved in proDerp1αS. Immunotoxin impact was assayed by using effector cells sensitized with house dust mite-allergic sera. Cell degranulation and death, triggered by proDerp1αS, was exclusively observed on Der p 1 sera sensitized-humRBL-2H3 cells, but not when treated with non-allergic sera. Most notably, equivalent IgE-binding and degranulation were observed with both proDerp1αS construct and native Der p 1 when using purified basophils from sensitized patients. However, proDerp1αS did not cause any cytotoxic effect on these cells, apparently due to its lack of internalization after their surface IgE-binding, showing the complex in vivo panorama governing allergic reactions. In conclusion, herein we present proDerp1αS as a proof of concept for a potential and alternative new designs of therapeutic tools for allergies. Development of new, and more specific, second-generation of immunotoxins following proDerp1αS, is further discussed.


Subject(s)
Allergens/immunology , Antigens, Dermatophagoides/immunology , Arthropod Proteins/immunology , Cysteine Endopeptidases/immunology , Dermatophagoides pteronyssinus/immunology , Hypersensitivity/immunology , Hypersensitivity/therapy , Immunotoxins/administration & dosage , Animals , Basophils/immunology , Basophils/metabolism , Cell Degranulation , Cell Line , Cells, Cultured , Desensitization, Immunologic , Humans , Immunoglobulin E/blood , Immunoglobulin E/immunology , Recombinant Proteins/immunology
8.
Cancer Immunol Immunother ; 69(8): 1535-1548, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32300857

ABSTRACT

With an infection rate of 60-90%, the human cytomegalovirus (HCMV) is very common among adults but normally causes no symptoms. When T cell-mediated immunity is compromised, HCMV reactivation can lead to increased morbidity and mortality. HCMV antigens are processed and presented as peptides on the cell surface via HLA I complexes to the T cell receptor (TCR) of T cells. The generation of antibodies against HCMV peptides presented on HLA complexes (TCR-like antibodies) has been described, but is without therapeutic applications to date due to the polygenic and polymorphic nature of HLA genes. We set out to obtain antibodies specific for HLA/HCMV-peptides, covering the majority of HLA alleles present in European populations. Using phage display technology, we selected 10 Fabs, able to bind to HCMV-peptides presented in the 6 different HLA class I alleles A*0101, A*0201, A*2402, B*0702, B*0801 and B*3501. We demonstrate specific binding of all selected Fabs to HLA-typed lymphoblastoid cell lines (EBV-transformed B cells) and lymphocytes loaded with HCMV-peptides. After infection with HCMV, 4/10 tetramerized Fabs restricted to the alleles HLA-A*0101, HLA-A*0201 and HLA-B*0702 showed binding to infected primary fibroblasts. When linked to the pseudomonas exotoxin A, these Fab antibodies induce highly specific cytotoxicity in HLA matched cell lines loaded with HCMV peptides. TCR-like antibody repertoires therefore represent a promising new treatment modality for viral infections and may also have applications in the treatment of cancers.


Subject(s)
Cytomegalovirus Infections/prevention & control , Cytomegalovirus/immunology , Fibroblasts/immunology , HLA Antigens/immunology , Immunoglobulin Fab Fragments/administration & dosage , Peptide Fragments/immunology , T-Lymphocytes, Cytotoxic/immunology , Antigens, Viral/immunology , Cell Survival , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/metabolism , Cytomegalovirus Infections/virology , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fab Fragments/metabolism , Immunotoxins/administration & dosage , Melanoma/immunology , Melanoma/metabolism , Melanoma/pathology , Melanoma/prevention & control , Receptors, Antigen, T-Cell/immunology , Viral Proteins/immunology
10.
Pharmacol Rep ; 72(4): 1032-1046, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32141025

ABSTRACT

BACKGROUND: Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL/Apo2L) has been perceived as a promising anti-cancer agent because of its unique ability to kill cancer cells while sparing normal cells. However, translation of TRAIL to clinical studies was less successful as a large number of cancer cells acquire resistance to TRAIL-based monotherapies. An ideal strategy to overcome TRAIL resistance is to combine it with potential sensitizing agents. OBJECTIVE: To investigate the TRAIL-sensitizing effect of curcumin in leukemia. METHODS: The mechanism underlying TRAIL sensitization by curcumin was studied by flow cytometric analysis of TRAIL receptors in leukemic cell lines and patient samples, and immunoblot detection of TRAIL-apoptosis signaling proteins. RESULTS: Curcumin augments TRAIL-apoptotic signaling in leukemic cells by upregulating the expression of DR4 and DR5 along with suppression of cFLIP and anti-apoptotic proteins Mcl-1, Bcl-xl, and XIAP. Curcumin pre-treatment significantly (p < 0.01) enhanced the sensitivity of leukemic cell lines to TRAIL recombinant proteins. IL2-TRAIL peptide in the presence of curcumin induced potent apoptosis (p < 0.001) as compared to TRAIL and IL2-TRAIL protein in leukemic cell lines with IC50 < 0.1 µΜ. Additionally, the combination of IL2-TRAIL peptide and curcumin showed significant cytotoxicity in patient peripheral blood mononuclear cells (PBMCs) with an efficacy of 90% in acute myeloid leukemia (AML), but 100% in acute lymphoblastic leukemia (ALL), chronic lymphocytic leukemia (CLL) and chronic myelomonocytic leukemia (CMML). CONCLUSION: Overall, our results suggest that curcumin potentiates TRAIL-induced apoptosis through modulation of death receptors and anti-apoptotic proteins which significantly enhances the therapeutic efficacy.


Subject(s)
Antineoplastic Agents/administration & dosage , Curcumin/administration & dosage , Immunotoxins/administration & dosage , Leukemia/metabolism , TNF-Related Apoptosis-Inducing Ligand/administration & dosage , Adolescent , Adult , Aged , Apoptosis/drug effects , Apoptosis/physiology , Child , Child, Preschool , Dose-Response Relationship, Drug , Female , HL-60 Cells , Humans , Infant , K562 Cells , Leukemia/drug therapy , Leukemia/immunology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Young Adult
11.
Proc Natl Acad Sci U S A ; 117(11): 6086-6091, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32123080

ABSTRACT

Recombinant immunotoxins (RITs) are chimeric proteins composed of an Fv and a protein toxin being developed for cancer treatment. The Fv brings the toxin to the cancer cell, but most of the RITs do not reach the tumor and are removed by other organs. To identify cells responsible for RIT removal, and the pathway by which RITs reach these cells, we studied SS1P, a 63-kDa RIT that targets mesothelin-expressing tumors and has a short serum half-life. The major organs that remove RIT were identified by live mouse imaging of RIT labeled with FNIR-Z-759. Cells responsible for SS1P removal were identified by immunohistochemistry and intravital two-photon microscopy of kidneys of rats. The primary organ of SS1P removal is kidney followed by liver. In the kidney, SS1P passes through the glomerulus, is taken up by proximal tubular cells, and transferred to lysosomes. In the liver, macrophages are involved in removal. The short half-life of SS1P is due to its very rapid filtration by the kidney followed by degradation in proximal tubular cells of the kidney. In mice treated with SS1P, proximal tubular cells are damaged and albumin in the urine is increased. SS1P uptake by kidney is reduced by coadministration of l-lysine. Our data suggests that l-lysine administration to humans might prevent SS1P-mediated kidney damage, reduce albumin loss in urine, and alleviate capillary leak syndrome.


Subject(s)
Albuminuria/pathology , Antibodies, Monoclonal/pharmacokinetics , Capillary Leak Syndrome/pathology , Immunotoxins/pharmacokinetics , Kidney Tubules, Proximal/drug effects , Albuminuria/chemically induced , Albuminuria/prevention & control , Albuminuria/urine , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/toxicity , Capillary Leak Syndrome/chemically induced , Capillary Leak Syndrome/prevention & control , Capillary Leak Syndrome/urine , Disease Models, Animal , Female , Fluorescent Dyes/chemistry , Half-Life , Humans , Immunotoxins/administration & dosage , Immunotoxins/chemistry , Immunotoxins/toxicity , Intravital Microscopy , Kidney Glomerulus/metabolism , Kidney Tubules, Proximal/diagnostic imaging , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Lysine/administration & dosage , Mesothelin , Mice , Microscopy, Fluorescence , Neoplasms/drug therapy , Recombinant Proteins/administration & dosage , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/toxicity , Renal Elimination/drug effects , Serum Albumin/analysis , Serum Albumin/metabolism , Staining and Labeling
12.
Int J Cancer ; 146(2): 449-460, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31584195

ABSTRACT

Bladder cancer is the sixth most common cancer in the United States, and it exhibits an alarming 70% recurrence rate. Thus, the development of more efficient antibladder cancer approaches is a high priority. Accordingly, this work provides the basis for a transformative anticancer strategy that takes advantage of the unique characteristics of the bladder. Unlike mucin-shielded normal bladder cells, cancer cells are exposed to the bladder lumen and overexpress EGFR. Therefore, we used an EGF-conjugated anthrax toxin that after targeting EGFR was internalized and triggered apoptosis in exposed bladder cancer cells. This unique agent presented advantages over other EGF-based technologies and other toxin-derivatives. In contrast to known agents, this EGF-toxin conjugate promoted its own uptake via receptor microclustering even in the presence of Her2 and induced cell death with a LC50 < 1 nM. Furthermore, our data showed that exposures as short as ≈3 min were enough to commit human (T24), mouse (MB49) and canine (primary) bladder cancer cells to apoptosis. Exposure of tumor-free mice and dogs with the agent resulted in no toxicity. In addition, the EGF-toxin was able to eliminate cells from human patient tumor samples. Importantly, the administration of EGF-toxin to dogs with spontaneous bladder cancer, who had failed or were not eligible for other therapies, resulted in ~30% average tumor reduction after one treatment cycle. Because of its in vitro and in vivo high efficiency, fast action (reducing treatment time from hours to minutes) and safety, we propose that this EGF-anthrax toxin conjugate provides the basis for new, transformative approaches against bladder cancer.


Subject(s)
Antigens, Bacterial/administration & dosage , Antineoplastic Agents/administration & dosage , Bacterial Toxins/administration & dosage , Epidermal Growth Factor/administration & dosage , Immunotoxins/administration & dosage , Urinary Bladder Neoplasms/drug therapy , Administration, Intravesical , Animals , Antigens, Bacterial/adverse effects , Antineoplastic Agents/adverse effects , Apoptosis/drug effects , Bacterial Toxins/adverse effects , Cell Line, Tumor , Dogs , Drug Screening Assays, Antitumor , Epidermal Growth Factor/adverse effects , Female , Humans , Immunotoxins/adverse effects , Male , Mice , Primary Cell Culture , Receptor, ErbB-2/metabolism , Treatment Outcome , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/veterinary
13.
J Pharm Sci ; 109(1): 104-115, 2020 01.
Article in English | MEDLINE | ID: mdl-31669121

ABSTRACT

Immunotoxins (ITs) are attractive anticancer modalities aimed at cancer-specific delivery of highly potent cytotoxic protein toxins. An IT consists of a targeting domain (an antibody, cytokine, or another cell-binding protein) chemically conjugated or recombinantly fused to a highly cytotoxic payload (a bacterial and plant toxin or human cytotoxic protein). The mode of action of ITs is killing designated cancer cells through the effector function of toxins in the cytosol after cellular internalization via the targeted cell-specific receptor-mediated endocytosis. Although numerous ITs of diverse structures have been tested in the past decades, only 3 ITs-denileukin diftitox, tagraxofusp, and moxetumomab pasudotox-have been clinically approved for treating hematological cancers. No ITs against solid tumors have been approved for clinical use. In this review, we discuss critical research and development issues associated with ITs that limit their clinical success as well as strategies to overcome these obstacles. The issues include off-target and on-target toxicities, immunogenicity, human cytotoxic proteins, antigen target selection, cytosolic delivery efficacy, solid-tumor targeting, and developability. To realize the therapeutic promise of ITs, novel strategies for safe and effective cytosolic delivery into designated tumors, including solid tumors, are urgently needed.


Subject(s)
Antineoplastic Agents/metabolism , Immunotoxins/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Bacterial Toxins/administration & dosage , Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Clinical Trials as Topic/methods , Cytosol/drug effects , Cytosol/metabolism , Exotoxins/administration & dosage , Exotoxins/chemistry , Exotoxins/metabolism , Humans , Immunotoxins/administration & dosage , Immunotoxins/chemistry , Protein Binding/physiology , Protein Structure, Secondary
14.
Methods Mol Biol ; 2021: 201-215, 2019.
Article in English | MEDLINE | ID: mdl-31309507

ABSTRACT

Proteus mirabilis is a major cause of complicated urinary tract infections (UTIs). P. mirabilis' urease activity hydrolyzes urea and raises urine pH levels, which can catalyze bladder and kidney stone formation. This urolithiasis leads to harder-to-treat infections, possible urinary blockage, and subsequent septicemia. Development of a mucosal vaccine against P. mirabilis urinary tract infections is critical to protect against this potentially deadly infection process. Here, we describe the methodology necessary to produce a vaccine candidate conjugated to cholera toxin, administer the vaccine via the intranasal route, and test efficacy in a murine transurethral P. mirabilis infection model.


Subject(s)
Antibodies, Bacterial/metabolism , Immunotoxins/administration & dosage , Proteus Infections/prevention & control , Proteus mirabilis/immunology , Urinary Tract Infections/prevention & control , Administration, Intranasal , Animals , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/immunology , Cholera Toxin/administration & dosage , Cholera Toxin/immunology , Disease Models, Animal , Female , Humans , Immunotoxins/immunology , Mice , Mice, Inbred CBA , Vaccination
15.
Clin Colorectal Cancer ; 18(3): 192-199.e1, 2019 09.
Article in English | MEDLINE | ID: mdl-31345777

ABSTRACT

BACKGROUND: Mesothelin (MSLN) is a cell surface glycoprotein expressed at a high level on many malignancies, including pancreatic adenocarcinoma, serous ovarian cancer, and epithelioid mesothelioma. MSLN-targeted recombinant immunotoxins (RITs) consist of an anti-MSLN Fv fused to the catalytic domain of Pseudomonas exotoxin A. Recent data has also shown that MSLN is expressed at clinically relevant levels on the surface of colorectal cancer (CRC). In this study, CRC cell lines were tested for MSLN expression and susceptibility to MSLN-targeted RITs. MATERIALS AND METHODS: CRC cell lines were tested for membranous MSLN expression via flow cytometry. Cell lines expressing MSLN were tested by WST-8 cell viability assay for sensitivity to various RITs and chemotherapeutic agents. CRC cell line SW-48 was tested in a mouse model for response to RIT as a single agent or in combination with actinomycin D and oxaliplatin. RESULTS: CRC cell lines were susceptible to anti-MSLN RITs at half maximal inhibitory concentration levels comparable with those previously described in pancreatic cancer cell lines. In a nude mouse model, MSLN-targeted RIT treatment of SW48 CRC tumors resulted in a significant decrease in tumor volume. Although combination therapy with standard of care chemotherapeutic oxaliplatin did not improve tumor regressions, combination therapy with actinomycin D resulted in > 90% tumor volume reduction with 50% complete regressions. CONCLUSIONS: These data support the development of anti-MSLN RITs as well as other MSLN-targeted therapies for CRC.


Subject(s)
ADP Ribose Transferases/administration & dosage , Antineoplastic Agents, Immunological/administration & dosage , Bacterial Toxins/administration & dosage , Colorectal Neoplasms/drug therapy , Exotoxins/administration & dosage , GPI-Linked Proteins/antagonists & inhibitors , Immunotoxins/administration & dosage , Virulence Factors/administration & dosage , ADP Ribose Transferases/genetics , ADP Ribose Transferases/immunology , Animals , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Antineoplastic Agents, Immunological/immunology , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Cell Line, Tumor , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Exotoxins/genetics , Exotoxins/immunology , Female , GPI-Linked Proteins/immunology , GPI-Linked Proteins/metabolism , Humans , Immunotoxins/genetics , Immunotoxins/immunology , Mesothelin , Mice , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Virulence Factors/genetics , Virulence Factors/immunology , Xenograft Model Antitumor Assays , Pseudomonas aeruginosa Exotoxin A
16.
Sci Rep ; 9(1): 5339, 2019 03 29.
Article in English | MEDLINE | ID: mdl-30926865

ABSTRACT

Staphylococcus aureus pneumonia is associated with high mortality irrespective of antibiotic susceptibility. Both MRSA and MSSA strains produce powerful cytotoxins: alpha-hemolysin(Hla) and up to five leukocidins - LukSF-PV, HlgAB, HlgCB, LukED and LukGH (LukAB) - to evade host innate defense mechanisms. Neutralizing cytotoxins has been shown to provide survival benefit in rabbit S. aureus pneumonia models. We studied the mechanisms of protection of ASN100, a combination of two human monoclonal antibodies (mAbs), ASN-1 and ASN-2, that together neutralize Hla and the five leukocidins, in rabbit MRSA and MSSA pneumonia models. Upon prophylactic passive immunization, ASN100 displayed dose-dependent increase in survival and was fully protective against all S. aureus strains tested at 5 or 20 mg/kg doses. Macroscopic and microscopic lung pathology, edema rate, and bacterial burden were evaluated 12 hours post infection and reduced by ASN100. Pharmacokinetic analysis of ASN100 in bronchoalveolar-lavage fluid from uninfected animals detected efficient penetration to lung epithelial lining fluid reaching peak levels between 24 and 48 hours post dosing that were comparable to the mAb concentration measured in serum. These data confirm that the ASN100 mAbs neutralize the powerful cytotoxins of S. aureus in the lung and prevent damage to the mucosal barrier and innate immune cells.


Subject(s)
Antibodies, Neutralizing/immunology , Immunoglobulin G/immunology , Immunotoxins/immunology , Pneumonia, Staphylococcal/immunology , Pneumonia, Staphylococcal/prevention & control , Staphylococcus aureus/immunology , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/immunology , Biopsy , Disease Models, Animal , Dose-Response Relationship, Immunologic , Immunization, Passive , Immunohistochemistry , Immunotoxins/administration & dosage , Methicillin-Resistant Staphylococcus aureus/immunology , Pneumonia, Staphylococcal/mortality , Pneumonia, Staphylococcal/pathology , Prognosis , Rabbits
17.
Proc Natl Acad Sci U S A ; 116(8): 3100-3105, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30718426

ABSTRACT

Denileukin diftitox (DAB-IL-2, Ontak) is a diphtheria-toxin-based fusion protein that depletes CD25-positive cells including regulatory T cells and has been approved for the treatment of persistent or recurrent cutaneous T cell lymphoma. However, the clinical use of denileukin diftitox was limited by vascular leak toxicity and production issues related to drug aggregation and purity. We found that a single amino acid substitution (V6A) in a motif associated with vascular leak induction yields a fully active, second-generation biologic, s-DAB-IL-2(V6A), which elicits 50-fold less human umbilical vein endothelial cell monolayer permeation and is 3.7-fold less lethal to mice by LD50 analysis than s-DAB-IL-2. Additionally, to overcome aggregation problems, we developed a production method for the fusion toxin using Corynebacterium diphtheriae that secretes fully folded, biologically active, monomeric s-DAB-IL-2 into the culture medium. Using the poorly immunogenic mouse B16F10 melanoma model, we initiated treatment 7 days after tumor challenge and observed that, while both s-DAB-IL-2(V6A) and s-DAB-IL-2 are inhibitors of tumor growth, the capacity to treat with higher doses of s-DAB-IL-2(V6A) could provide a superior activity window. In a sequential dual-therapy study in tumors that have progressed for 10 days, both s-DAB-IL-2(V6A) and s-DAB-IL-2 given before checkpoint inhibition with anti-programmed cell death-1 (anti-PD-1) antibodies inhibited tumor growth, while either drug given as monotherapy had less effect. s-DAB-IL-2(V6A), a fully monomeric protein with reduced vascular leak, is a second-generation diphtheria-toxin-based fusion protein with promise as a cancer immunotherapeutic both alone and in conjunction with PD-1 blockade.


Subject(s)
Diphtheria Toxin/administration & dosage , Interleukin-2/administration & dosage , Melanoma, Experimental/drug therapy , Programmed Cell Death 1 Receptor/genetics , Amino Acid Substitution/genetics , Antibodies/administration & dosage , Cell Proliferation/drug effects , Corynebacterium diphtheriae/chemistry , Corynebacterium diphtheriae/pathogenicity , Diphtheria Toxin/chemistry , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Immunosuppressive Agents/administration & dosage , Immunotoxins/administration & dosage , Interleukin-2/chemistry , Interleukin-2 Receptor alpha Subunit/drug effects , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Receptors, Interleukin-2/genetics , Receptors, Interleukin-2/immunology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/chemistry , T-Lymphocytes, Regulatory/drug effects
18.
Proc Natl Acad Sci U S A ; 116(10): 4592-4598, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30782819

ABSTRACT

Multiple myeloma (MM) is a B cell malignancy for which new treatments are urgently needed. The B cell maturation antigen (BCMA) is a lineage-restricted differentiation protein highly expressed on myeloma. Recombinant immunotoxins (RITs) are proteins composed of the Fv or Fab portion of an antibody fused to a bacterial toxin. We previously treated H929 myeloma s.c. tumors with anti-BCMA immunotoxins, very active on killing cultured cells, and observed tumor growth inhibition but not complete tumor responses. To determine if immunotoxins were more active against cells growing in the bone marrow (BM), the normal location of myeloma cells, we developed a BM mouse model that is more relevant to human disease. H929 cells were transfected with luciferase and GFP, enriched by flow, recycled through the BM of a mouse, and injected IV into nonobese diabetic scid γ mice (NSG) mice. A second myeloma mouse model used the MM.1S-GFP-luc cell line. Mice were treated IV with immunotoxins, and the tumor burden was assessed using bioluminescence imaging. We achieved complete durable remissions when treating mice with H929-GFP-luc cells with anti-BCMA RITs both leptomycin B-75 (LMB-75) [anti-BCMA-disulfide-stabilized (ds)-Fv-PE24] (where PE represents Pseudomonas exotoxin A) or LMB-70 (anti-BCMA-Fab-PE24) given every other day for 5-d (QOD×5) doses beginning on day 4 or day 8. Mice were disease free at 3 months; untreated mice became moribund around day 40. We also achieved long-term responses using the MM.1S-GFP-luc myeloma cell line. Treatment with an 1.5 mg/kg LMB-75 QOD×5 anti-BCMA RIT beginning on day 4 caused the complete disappearance of tumors for 80 days. To summarize, LMB-75 and LMB-70, our anti-BCMA RITs, induced complete durable responses in two myeloma models.


Subject(s)
Antibodies, Monoclonal/administration & dosage , B-Cell Maturation Antigen/immunology , Immunotherapy , Immunotoxins/administration & dosage , Multiple Myeloma/therapy , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Multiple Myeloma/genetics , Multiple Myeloma/immunology , Multiple Myeloma/physiopathology , Remission Induction
19.
Drugs ; 78(16): 1763-1767, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30357593

ABSTRACT

Moxetumomab pasudotox-tdfk (LUMOXITI™), an anti CD22 recombinant immunotoxin, has been developed by MedImmune and its parent company AstraZeneca for the treatment of hairy cell leukaemia. The product, discovered at the National Cancer Institute, is an optimised version of immunotoxin CAT-3888. Moxetumomab pasudotox is composed of the Fv fragment of an anti-CD22 monoclonal antibody fused to a 38 kDa fragment of Pseudomonas exotoxin A, PE38. The Fv portion of moxetumomab pasudotox binds to CD22, a cell surface receptor expressed on a variety of malignant B-cells, thereby delivering the toxin moiety PE38 directly to tumour cells. Once internalised, PE38 catalyses the ADP ribosylation of the diphthamide residue in elongation factor-2 (EF-2), resulting in the rapid fall in levels of the anti-apoptotic protein myeloid cell leukaemia 1 (Mcl-1), leading to apoptotic cell death. This article summarizes the milestones in the development of moxetumomab pasudotox leading to this first approval for the treatment of adults with relapsed or refractory hairy cell leukaemia who received at least two prior systemic therapies, including treatment with a purine nucleoside analogue. Development of moxetumomab pasudotox for non-Hodgkin's lymphoma, chronic lymphocytic leukaemia and precursor cell lymphoblastic leukaemia/lymphoma was discontinued.


Subject(s)
Antineoplastic Agents , Bacterial Toxins , Exotoxins , Leukemia, Lymphoid/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Antineoplastic Agents, Immunological , Bacterial Toxins/administration & dosage , Bacterial Toxins/adverse effects , Bacterial Toxins/therapeutic use , Exotoxins/administration & dosage , Exotoxins/adverse effects , Exotoxins/therapeutic use , Humans , Immunoglobulin Variable Region , Immunotoxins/administration & dosage , Immunotoxins/adverse effects , Immunotoxins/chemistry , Immunotoxins/therapeutic use , Middle Aged , Sialic Acid Binding Ig-like Lectin 2/antagonists & inhibitors
20.
Theranostics ; 8(13): 3544-3558, 2018.
Article in English | MEDLINE | ID: mdl-30026865

ABSTRACT

Cervical cancer, the second most common cause of cancer death in women worldwide, is significantly associated with infection of high-risk human papillomaviruses (HPVs), especially the most common genotype, HPV 16. To date, there is no established noninvasive therapy to treat cervical cancer. Methods: Here, we report a novel affitoxin that targets HPV16 E7 protein, one of the primary target proteins in molecular targeted therapy for HPV-induced cervical cancer. The affitoxin, ZHPV16E7 affitoxin384 was generated by fusing the modified Pseudomonas Exotoxin A (PE38KDEL) to the HPV16 E7-specific affibody. The expressed and purified ZHPV16E7 affitoxin384 was characterized using numerous methods. SPR assay, indirect immunofluorescence assay, and near-infrared (NIR) optical imaging were respectively performed to assess the targeting ability of ZHPV16E7 affitoxin384 to HPV16 E7 protein both in vitro and in vivo. Cell viability assays and SiHa tumor-bearing nude mice were used to evaluate the efficacy of ZHPV16 E7 affitoxin384 in vitro and in vivo, respectively. Results: Using in vitro methods the SPR assay and indirect immunofluorescence assay showed that ZHPV16E7 affitoxin384 targeted HPV16 E7 with high binding affinity and specificity. Significant reduction of cell viability in HPV16 positive cells was observed in the presence of ZHPV16 E7 affitoxin384. By NIR optical imaging, ZHPV16 E7 affitoxin384 specifically targeted HPV16 positive tumors in vivo. ZHPV16E7 affitoxin384 showed significant in vivo antitumor efficacy in two kinds of tumor-bearing nude mouse models. Conclusions: ZHPV16E7 affitoxin384 is a potent anti-cervical cancer therapeutic agent that could be effective against HPV16 positive tumors in humans.


Subject(s)
Antineoplastic Agents/administration & dosage , Immunotherapy/methods , Immunotoxins/administration & dosage , Molecular Targeted Therapy/methods , Papillomavirus E7 Proteins/antagonists & inhibitors , Uterine Cervical Neoplasms/drug therapy , Animals , Antibodies, Viral/genetics , Antibodies, Viral/metabolism , Antineoplastic Agents/pharmacology , Bacterial Toxins/genetics , Bacterial Toxins/toxicity , Cell Line, Tumor , Cell Survival , Disease Models, Animal , Female , Humans , Immunotoxins/genetics , Immunotoxins/pharmacology , Mice , Mice, Nude , Pseudomonas/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/toxicity , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...