Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Eur J Cancer ; 172: 311-322, 2022 09.
Article in English | MEDLINE | ID: mdl-35816972

ABSTRACT

INTRODUCTION: Hepatoblastoma (HB) is the most common paediatric liver tumour, and epigenetic aberrations may be important in HB development. Recently, the Children's Hepatic Tumors International Collaboration-Hepatoblastoma Stratification (CHIC-HS) developed risk stratification based on clinicopathological factors. This study aimed to construct a more accurate model by integrating CHIC-HS with molecular factors based on DNA methylation. METHODS: HB tumour specimens (N = 132) from patients treated with the Japanese Pediatric Liver Tumors Group-2 protocol were collected and subjected to methylation analysis by bisulfite pyrosequencing. Associations between methylation status and clinicopathological factors, overall survival (OS), and event-free survival (EFS) were retrospectively analysed. We investigated the effectiveness of the evaluation of methylation status in each CHIC-HS risk group and generated a new risk stratification model. RESULTS: Most specimens (82%) were from post-chemotherapy tissue. Hypermethylation in ≥2 of the four genes (RASSF1A, PARP6, OCIAD2, and MST1R) was significantly associated with poorer OS and EFS. Multivariate analysis indicated that ≥2 methylated genes was an independent prognostic factor (hazard ratios of 6.014 and 3.684 for OS and EFS, respectively). Two or more methylated genes was also associated with poorer OS in the CHIC-very low (VL)-/low (L)-risk and CHIC-intermediate (I) risk groups (3-year OS rates were 83% vs. 98% and 50% vs. 95%, respectively). The 3-year OS rates of the VL/L, I, and high-risk groups in the new stratification model were 98%, 90%, and 62% (vs. CHIC-HS [96%, 82%, and 65%, respectively]), optimising CHIC-HS. CONCLUSIONS: Our proposed stratification system considers individual risk in HB and may improve patient clinical management.


Subject(s)
Hepatoblastoma , Liver Neoplasms , ADP Ribose Transferases/genetics , ADP Ribose Transferases/therapeutic use , Child , DNA , DNA Methylation , Hepatoblastoma/genetics , Hepatoblastoma/pathology , Humans , Liver Neoplasms/drug therapy , Neoplasm Proteins/genetics , Retrospective Studies , Risk Assessment
2.
Mol Pharm ; 18(6): 2285-2297, 2021 06 07.
Article in English | MEDLINE | ID: mdl-33998814

ABSTRACT

Cholecystokinin-2 receptor (CCK2R) has been proven to be a specific biomarker for colorectal malignancies. Immunotoxins are a valuable class of immunotherapy agents consisting of a targeting element and a bacterial or plant toxin. Previous work demonstrated that targeting CCK2R is a good therapeutic strategy for the treatment of colorectal cancer (CRC). In the present study, we developed a new version of CCK2R-targeting immunotoxin GD9P using a targeted peptide, GD9, as the binding motif and a truncated Pseudomonas exotoxin A (PE38) as the cytokiller. BALB/c nude mice were treated with different doses of GD9P, and pharmacodynamics, pharmacokinetic, and toxicological data were obtained throughout this study. Compared to the parental immunotoxin rCCK8PE38, GD9P exhibited about 1.5-fold yield, higher fluorescence intensity, and increased antitumor activity against human CRC in vitro and in vivo. The IC50 values of GD9P in vitro ranged from 1.61 to 4.55 nM. Pharmacokinetic studies were conducted in mice with a T1/2 of 69.315 min. When tumor-bearing nude mice were treated with GD9P at doses ≥2 mg/kg for five doses, a rapid shrinkage in tumor volume and, in some cases, complete remission was observed. A preliminary safety evaluation demonstrated a good safety profile of GD9P as a Pseudomonas exotoxin A-based immunotherapy. The therapy in combination with oxaliplatin can increase the antitumor efficacy and reduce the toxic side effects caused by chemotherapy. In conclusion, the data support the use of GD9P as a promising immunotherapy targeting CCK2R-expressing colorectal malignancies.


Subject(s)
ADP Ribose Transferases/pharmacology , Antineoplastic Agents/pharmacology , Bacterial Toxins/pharmacology , Colorectal Neoplasms/drug therapy , Exotoxins/pharmacology , Receptor, Cholecystokinin B/antagonists & inhibitors , Recombinant Fusion Proteins/pharmacology , Virulence Factors/pharmacology , ADP Ribose Transferases/genetics , ADP Ribose Transferases/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Bacterial Toxins/genetics , Bacterial Toxins/therapeutic use , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Exotoxins/genetics , Exotoxins/therapeutic use , Humans , Mice , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/therapeutic use , Tissue Distribution , Toxicity Tests, Acute , Virulence Factors/genetics , Virulence Factors/therapeutic use , Xenograft Model Antitumor Assays , Pseudomonas aeruginosa Exotoxin A
3.
Toxins (Basel) ; 12(12)2020 11 28.
Article in English | MEDLINE | ID: mdl-33260619

ABSTRACT

The epidermal growth factor receptor (EGFR) was found to be a valuable target on prostate cancer (PCa) cells. However, EGFR inhibitors mostly failed in clinical studies with patients suffering from PCa. We therefore tested the targeted toxins EGF-PE40 and EGF-PE24mut consisting of the natural ligand EGF as binding domain and PE40, the natural toxin domain of Pseudomonas Exotoxin A, or PE24mut, the de-immunized variant thereof, as toxin domains. Both targeted toxins were expressed in the periplasm of E.coli and evoked an inhibition of protein biosynthesis in EGFR-expressing PCa cells. Concentration- and time-dependent killing of PCa cells was found with IC50 values after 48 and 72 h in the low nanomolar or picomolar range based on the induction of apoptosis. EGF-PE24mut was found to be about 11- to 120-fold less toxic than EGF-PE40. Both targeted toxins were more than 600 to 140,000-fold more cytotoxic than the EGFR inhibitor erlotinib. Due to their high and specific cytotoxicity, the EGF-based targeted toxins EGF-PE40 and EGF-PE24mut represent promising candidates for the future treatment of PCa.


Subject(s)
ADP Ribose Transferases/therapeutic use , Bacterial Toxins/therapeutic use , Exotoxins/therapeutic use , Immunotoxins/therapeutic use , Prostatic Neoplasms/drug therapy , Virulence Factors/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , CHO Cells , Cell Line, Tumor , Cell Survival , Cricetulus , ErbB Receptors/antagonists & inhibitors , Humans , Male , PC-3 Cells , Recombinant Fusion Proteins/therapeutic use , Pseudomonas aeruginosa Exotoxin A
4.
Cancer Sci ; 111(12): 4548-4557, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33058342

ABSTRACT

Drug resistance represents an obstacle in colorectal cancer (CRC) treatment because of its association with poor prognosis. rBC2LCN is a lectin isolated from Burkholderia that binds cell surface glycans that have fucose moieties. Because fucosylation is enhanced in many types of cancers, this lectin could be an efficient drug carrier if CRC cells specifically present such glycans. Therefore, we examined the therapeutic efficacy and toxicity of lectin drug conjugate therapy in CRC mouse xenograft models. The affinity of rBC2LCN for human CRC cell lines HT-29, LoVo, LS174T, and DLD-1 was assessed in vitro. The cytocidal efficacy of a lectin drug conjugate, rBC2LCN-38 kDa domain of pseudomonas exotoxin A (PE38) was evaluated by MTT assay. The therapeutic effects and toxicity for each CRC cell line-derived mouse xenograft model were compared between the intervention and control groups. LS174T and DLD-1 cell lines showed a strong affinity for rBC2LCN. In the xenograft model, the tumor volume in the rBC2LCN-PE38 group was significantly reduced compared with that using control treatment alone. However, the HT-29 cell line showed weak affinity and poor therapeutic efficacy. No significant toxicities or adverse responses were observed. In conclusion, we demonstrated that rBC2LCN lectin binds CRC cells and that rBC2LCN-PE38 significantly suppresses tumor growth in vivo. In addition, the efficacy of the drug conjugate correlated with its binding affinity for each CRC cell line. These results suggest that lectin drug conjugate therapy has potential as a novel targeted therapy for CRC cell surface glycans.


Subject(s)
ADP Ribose Transferases/therapeutic use , Adenocarcinoma/drug therapy , Bacterial Toxins/therapeutic use , Colorectal Neoplasms/drug therapy , Exotoxins/therapeutic use , Immunoconjugates/therapeutic use , Lectins/therapeutic use , Virulence Factors/therapeutic use , ADP Ribose Transferases/adverse effects , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Bacterial Toxins/adverse effects , Burkholderia cenocepacia/chemistry , Cell Line, Tumor , Cell Survival , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Drug Carriers , Exotoxins/adverse effects , Fucose/metabolism , Fucosyltransferases/metabolism , HT29 Cells , Heterografts , Humans , Immunoconjugates/adverse effects , In Vitro Techniques , Lectins/isolation & purification , Lectins/metabolism , Mice , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/therapeutic use , Tumor Burden , Virulence Factors/adverse effects , Pseudomonas aeruginosa Exotoxin A
5.
Hepatology ; 71(5): 1696-1711, 2020 05.
Article in English | MEDLINE | ID: mdl-31520528

ABSTRACT

BACKGROUND AND AIMS: Treatment of hepatocellular carcinomas using our glypican-3 (GPC3)-targeting human nanobody (HN3) immunotoxins causes potent tumor regression by blocking protein synthesis and down-regulating the Wnt signaling pathway. However, immunogenicity and a short serum half-life may limit the ability of immunotoxins to transition to the clinic. APPROACH AND RESULTS: To address these concerns, we engineered HN3-based immunotoxins to contain various deimmunized Pseudomonas exotoxin (PE) domains. This included HN3-T20, which was modified to remove T-cell epitopes and contains a PE domain II truncation. We compared them to our previously reported B-cell deimmunized immunotoxin (HN3-mPE24) and our original HN3-immunotoxin with a wild-type PE domain (HN3-PE38). All of our immunotoxins displayed high affinity to human GPC3, with HN3-T20 having a KD value of 7.4 nM. HN3-T20 retained 73% enzymatic activity when compared with the wild-type immunotoxin in an adenosine diphosphate-ribosylation assay. Interestingly, a real-time cell growth inhibition assay demonstrated that a single dose of HN3-T20 at 62.5 ng/mL (1.6 nM) was capable of inhibiting nearly all cell proliferation during the 10-day experiment. To enhance HN3-T20's serum retention, we tested the effect of adding a streptococcal albumin-binding domain (ABD) and a llama single-domain antibody fragment specific for mouse and human serum albumin. For the detection of immunotoxin in mouse serum, we developed a highly sensitive enzyme-linked immunosorbent assay and found that HN3-ABD-T20 had a 45-fold higher serum half-life than HN3-T20 (326 minutes vs. 7.3 minutes); consequently, addition of an ABD resulted in HN3-ABD-T20-mediated tumor regression at 1 mg/kg. CONCLUSION: These data indicate that ABD-containing deimmunized HN3-T20 immunotoxins are high-potency therapeutics ready to be evaluated in clinical trials for the treatment of liver cancer.


Subject(s)
ADP Ribose Transferases/therapeutic use , Bacterial Toxins/therapeutic use , Carcinoma, Hepatocellular/therapy , Exotoxins/therapeutic use , Glypicans/antagonists & inhibitors , Immunotoxins/therapeutic use , Liver Neoplasms/therapy , Single-Domain Antibodies/therapeutic use , Virulence Factors/therapeutic use , ADP Ribose Transferases/chemistry , ADP Ribose Transferases/pharmacology , Animals , Bacterial Toxins/chemistry , Bacterial Toxins/pharmacology , Cell Line, Tumor , Exotoxins/chemistry , Exotoxins/pharmacology , Humans , Immunotoxins/chemistry , Immunotoxins/pharmacology , Mice , Mice, Nude , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/pharmacology , Virulence Factors/chemistry , Virulence Factors/pharmacology , Xenograft Model Antitumor Assays , Pseudomonas aeruginosa Exotoxin A
6.
Immunotherapy ; 11(13): 1117-1128, 2019 09.
Article in English | MEDLINE | ID: mdl-31361167

ABSTRACT

T regulatory cells (Tregs) are an important T cell population for immune tolerance, prevention of autoimmune diseases and inhibition of antitumor immunity. The tumor-promoting role played by Tregs in cancer has prompted numerous approaches to develop immunotherapeutics targeting Tregs. One approach to depletion of Treg cells is retargeting the highly potent cytotoxic activity of bacterial toxins. These agents capitalize on the well-characterized bacterial toxins, diphtheria toxin and Pseudomonas aeruginosa exotoxin A-both of which harbor membrane translocation domains and enzymatic domains that catalytically halt protein synthesis within intoxicated eukaryotic cells and act at picomolar or subpicomolar concentrations. In this review, we summarize the preclinical and clinical development of several Treg-depleting cancer immunotherapies based on these two bacterial toxins.


Subject(s)
ADP Ribose Transferases/therapeutic use , Bacterial Toxins/therapeutic use , Diphtheria Toxin/therapeutic use , Exotoxins/therapeutic use , Immunotherapy/methods , Lymphocyte Depletion/methods , Neoplasms/therapy , T-Lymphocytes, Regulatory/physiology , Virulence Factors/therapeutic use , Animals , Clinical Trials as Topic , Drug Evaluation, Preclinical , Humans , Immunity, Cellular/drug effects , Neoplasms/immunology , Tumor Microenvironment/drug effects , Pseudomonas aeruginosa Exotoxin A
7.
World J Microbiol Biotechnol ; 35(6): 94, 2019 Jun 11.
Article in English | MEDLINE | ID: mdl-31187291

ABSTRACT

Pseudomonas aeruginosa is the major infectious agent of concern for cystic fibrosis (CF) patients. Therefore, it is necessary to develop appropriate strategies for preventing colonization by this bacterium and/or neutralizing virulence factors. In this study, we formulated the encapsulation of exotoxin A into PLGA nanoparticles. The biological activities of the nanovaccine candidate were also characterized. Based on the results, ETA-PLGA can act as a suitable immunogen to stimulate the humoral and cellular immune response. The antibodies raised against ETA-PLGA significantly decreased bacterial titer in the spleens of the immunized mice after challenge with PAO1 strain, compared to the control groups. The encapsulation of PLGA into ETA led to a significantly higher production of INF-γ, TNF-α, IL-4, and IL-17A cytokine responses compared to the ETA group. ETA-PLGA enhanced IgG responses in immunized mice compared to ETA antigen. We concluded that encapsulation of Pseudomonas aeruginosa ETA to PLGA nanoparticles can increase its functional activity by decreasing the bacterial dissemination.


Subject(s)
ADP Ribose Transferases/immunology , Bacterial Toxins/immunology , Exotoxins/immunology , Immunization , Nanoconjugates , Polylactic Acid-Polyglycolic Acid Copolymer/immunology , Pseudomonas Infections/prevention & control , Pseudomonas aeruginosa/pathogenicity , Vaccines, Conjugate , Virulence Factors/immunology , ADP Ribose Transferases/therapeutic use , Animals , Bacterial Toxins/therapeutic use , Cytokines/metabolism , Disease Models, Animal , Exotoxins/therapeutic use , Female , Immunity, Cellular , Immunity, Humoral , Immunoglobulin G/blood , Interferon-gamma/metabolism , Interleukin-17/metabolism , Interleukin-4/metabolism , Mice , Mice, Inbred BALB C , Nanoparticles , Particle Size , Polylactic Acid-Polyglycolic Acid Copolymer/therapeutic use , Pseudomonas Infections/immunology , Spleen/immunology , Spleen/microbiology , Virulence Factors/therapeutic use , Pseudomonas aeruginosa Exotoxin A
8.
Int Immunopharmacol ; 72: 74-81, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30959374

ABSTRACT

Microvesicles (MVs) have been extensively identified in various biological fluids including bronchoalveolar lavage fluid (BALF), peripheral blood and ascitic fluids. Our previous study showed that MVs are responsible for acute lung injury, but the exact mechanism underlying MVs formation remains poorly understood. In the present study, we investigate the potential role of RhoA/Rock signaling in MVs generation and the biological activity of MVs in ventilator-induced lung injury (VILI). Our results revealed that high tide ventilation induced super MVs releasing into the lung and subsequently caused lung inflammation. Strikingly, intratracheal instillation of MVs that isolated from highly ventilated mice triggered significant lung inflammation in naive mice. The MVs production is strongly correlated with lung inflammation and the upregulation of RhoA, Rock and phospho-Limk (phosphorylation of Limk is the activated form). RhoA inhibitor decreased the expression of Rock and the phosphorylation of Limk, decreased MVs production and alleviated lung inflammation. Rock inhibitor also decreased the phosphorylation of Limk, decreased MVs production and alleviated lung inflammation. Our data demonstrated that the production of MVs requires RhoA/Rock signaling, and VILI might be potentially prevented by targeting RhoA/Rock signaling pathway.


Subject(s)
ADP Ribose Transferases/therapeutic use , Botulinum Toxins/therapeutic use , Cell-Derived Microparticles/drug effects , Ventilator-Induced Lung Injury/drug therapy , rhoA GTP-Binding Protein/antagonists & inhibitors , Amides/therapeutic use , Animals , Bronchoalveolar Lavage Fluid/immunology , Cytokines/immunology , Lung/drug effects , Lung/immunology , Lung/pathology , Mice, Inbred C57BL , Pyridines/therapeutic use , Ventilator-Induced Lung Injury/immunology , Ventilator-Induced Lung Injury/pathology , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/immunology , rhoA GTP-Binding Protein/immunology
9.
J Neurotrauma ; 35(9): 1049-1056, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29316845

ABSTRACT

Traumatic spinal cord injury (SCI) is associated with a lifetime of disability stemming from loss of motor, sensory, and autonomic functions; these losses, along with increased comorbid sequelae, negatively impact health outcomes and quality of life. Early decompression surgery post-SCI can enhance patient outcomes, but does not directly facilitate neural repair and regeneration. Currently, there are no U.S. Food and Drug Administration-approved pharmacological therapies to augment motor function and functional recovery in individuals with traumatic SCI. After an SCI, the enzyme, Rho, is activated by growth-inhibitory factors and regulates events that culminate in collapse of the neuronal growth cone, failure of axonal regeneration, and, ultimately, failure of motor and functional recovery. Inhibition of Rho activation is a potential treatment for injuries such as traumatic SCI. VX-210, an investigational agent, inhibits Rho. When administered extradurally after decompression (corpectomy or laminectomy) and stabilization surgery in a phase 1/2a study, VX-210 was well tolerated. Here, we describe the design of the SPRING trial, a multicenter, phase 2b/3, randomized, double-blind, placebo-controlled clinical trial to evaluate the efficacy and safety of VX-210 (NCT02669849). A subset of patients with acute traumatic cervical SCI is currently being enrolled in the United States and Canada. Medical, neurological, and functional changes are evaluated at 6 weeks and at 3, 6, and 12 months after VX-210 administration. Efficacy will be assessed by the primary outcome measure, change in upper extremity motor score at 6 months post-treatment, and by secondary outcomes that include question-based and task-based evaluations of functional recovery.


Subject(s)
ADP Ribose Transferases/therapeutic use , Botulinum Toxins/therapeutic use , Neuroprotective Agents/therapeutic use , Research Design , Spinal Cord Injuries/drug therapy , Cervical Vertebrae , Double-Blind Method , Enzyme Inhibitors/therapeutic use , Humans , rho-Associated Kinases/antagonists & inhibitors
10.
Mol Oncol ; 10(8): 1317-29, 2016 10.
Article in English | MEDLINE | ID: mdl-27507537

ABSTRACT

Mesothelin overexpression in lung adenocarcinomas correlates with the presence of activating KRAS mutations and poor prognosis. Hence SS1P, a mesothelin-targeted immunotoxin, could offer valuable treatment options for these patients, but its use in solid tumor therapy is hampered by high immunogenicity and non-specific toxicity. To overcome both obstacles we developed RG7787, a de-immunized cytotoxic fusion protein comprising a humanized SS1 Fab fragment and a truncated, B-cell epitope silenced, 24 kD fragment of Pseudomonas exotoxin A (PE24). Reactivity of RG7787 with sera from immunotoxin-treated patients was >1000 fold reduced. In vitro RG7787 inhibited cell viability of lung cancer cell lines with picomolar potency. The pharmacokinetic properties of RG7787 in rodents were comparable to SS1P, yet it was tolerated up to 10 fold better without causing severe vascular leak syndrome or hepatotoxicity. A pharmacokinetic/pharmacodynamic model developed based on NCI-H596 xenograft studies showed that for RG7787 and SS1P, their in vitro and in vivo potencies closely correlate. At optimal doses of 2-3 mg/kg RG7787 is more efficacious than SS1P. Even large, well established tumors (600 mm(3)) underwent remission during three treatment cycles with RG7787. Also in two patient-derived lung cancer xenograft models, Lu7336 and Lu7187, RG7787 showed anti-tumor efficacy. In monotherapy two treatment cycles were moderately efficacious in the Lu7336 model but showed good anti-tumor activity in the KRAS mutant Lu7187 model (26% and 80% tumor growth inhibition, respectively). Combination of RG7787 with standard chemotherapies further enhanced efficacy in both models achieving near complete eradication of Lu7187 tumors.


Subject(s)
ADP Ribose Transferases/therapeutic use , Bacterial Toxins/therapeutic use , Exotoxins/therapeutic use , GPI-Linked Proteins/metabolism , Lung Neoplasms/drug therapy , Protein Engineering , Pseudomonas/metabolism , Recombinant Fusion Proteins/therapeutic use , Virulence Factors/therapeutic use , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Liver/drug effects , Liver/pathology , Lung Neoplasms/pathology , Mesothelin , Mice, SCID , Models, Biological , Rats , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/pharmacology , Xenograft Model Antitumor Assays , Pseudomonas aeruginosa Exotoxin A
11.
Biochem Biophys Res Commun ; 475(1): 93-9, 2016 06 17.
Article in English | MEDLINE | ID: mdl-27178207

ABSTRACT

Immunotoxins have efficient anti-tumor activity due to their extreme potency. However, dose-limiting off-target toxicity and immunogenicity are the critical barriers for these immunotoxins to be used in a clinical setting. In this study, we designed a Pseudomonas exotoxin A (PE)-based human epidermal growth factor receptor-2 (HER2)-specific immunotoxin HER2-PE25-X7 by deleting most of domain II and introducing seven point mutations into domain III of the PE38 toxin. The anti-cancer activity, off-target toxicity and immunogenicity of this immunotoxin were carefully evaluated in vitro and in vivo. This new construct maintained the therapeutic potency of the original PE38-based immunotoxin HER2-PE38, with a greatly reduced off-target toxicity and immunogenicity. To compare with HER2-PE38, which resulted in the death of most of the mice after a single dose of 1.0 mg/kg, the new construct was completely tolerated at a dose of 10 mg/kg by the mice and almost completely depleted the tumor after treatment with five doses of 5 mg/kg of the immunotoxin. This work demonstrates a potentially attractive therapeutic modality for HER2-specific cancer treatment.


Subject(s)
ADP Ribose Transferases/therapeutic use , Antineoplastic Agents/therapeutic use , Bacterial Toxins/therapeutic use , Exotoxins/therapeutic use , Immunotoxins/therapeutic use , Neoplasms/drug therapy , Receptor, ErbB-2/immunology , Virulence Factors/therapeutic use , ADP Ribose Transferases/immunology , Animals , Antineoplastic Agents/immunology , Bacterial Toxins/immunology , Cell Line , Cell Line, Tumor , Exotoxins/immunology , Female , Humans , Immunotoxins/immunology , Mice, Inbred BALB C , Mice, Nude , Neoplasms/immunology , Pseudomonas/immunology , Virulence Factors/immunology , Pseudomonas aeruginosa Exotoxin A
12.
J Control Release ; 233: 48-56, 2016 07 10.
Article in English | MEDLINE | ID: mdl-27178808

ABSTRACT

DARPins fused with other proteins are promising non-immunoglobulin scaffolds for specific binding to target cells. In this study HER2-specific DARPin (DARPin_9-29) was used as a tumor-targeting moiety for the delivery of a cytotoxic agent - the fragment of Pseudomonas aeruginosa exotoxin A. It was determined that DARPin-PE40 possesses a considerable cytotoxic activity and induces apoptosis in HER2-positive cells. Cytotoxic effect of DARPin-PE40 strongly correlates with the HER2 expression level. The effect of intravenous administration of DARPin-PE40 was tested in the xenograft model of breast cancer. It was shown that treatment of animals with DARPin-PE40 caused strong and prolonged suppression of xenograft tumor growth.


Subject(s)
ADP Ribose Transferases/administration & dosage , Antineoplastic Agents/administration & dosage , Bacterial Toxins/administration & dosage , Exotoxins/administration & dosage , Immunotoxins/administration & dosage , Mammary Neoplasms, Experimental/drug therapy , Receptor, ErbB-2/metabolism , Recombinant Fusion Proteins/administration & dosage , Virulence Factors/administration & dosage , ADP Ribose Transferases/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Bacterial Toxins/therapeutic use , CHO Cells , Cell Line, Tumor , Cell Survival/drug effects , Cricetulus , Exotoxins/therapeutic use , Female , Humans , Immunotoxins/therapeutic use , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice, Nude , Recombinant Fusion Proteins/therapeutic use , Tumor Burden/drug effects , Virulence Factors/therapeutic use , Pseudomonas aeruginosa Exotoxin A
13.
J Natl Cancer Inst ; 108(8)2016 08.
Article in English | MEDLINE | ID: mdl-27075852

ABSTRACT

BACKGROUND: Triple-negative breast cancers (TNBCs) are typically more aggressive and result in poorer outcomes than other breast cancers because treatment options are limited due to lack of hormone receptors or amplified human epidermal growth factor receptor 2 (HER2). Many TNBCs overexpress the epidermal growth factor receptor (EGFR) or manifest amplification of theEGFRgene, supporting EGFR as a therapeutic target. While EGFR-directed small molecule inhibitors have shown limited effectiveness in clinical settings, use of EGFR as a mechanism of delivering enzymatic cytotoxins to TNBC has not been demonstrated. METHODS: Using the single-chain variable fragment (scFv) of the 806 antibody that binds only cells with overexpressed, misfolded, or mutant variants of the EGFR, a recombinant immunotoxin was engineered through gene fusion withPseudomonas aeruginosaExotoxin A (806-PE38). The potency of 806-PE38 on reducing TNBC cell growth in vitro and in xenograft models (n ≥ 6) was examined for six TNBC cell lines. All statistical tests were two-sided. RESULTS: 806-PE38 statistically significantly reduced the viability of all tested TNBC lines, with IC50values below 10 ng/mL for three of six cell lines, while not affecting cells with wild-type EGFR (IC50>300 ng/mL). Systemic treatments with 806-PE38 vs vehicle resulted in statistically significantly reduced tumor burdens (806-PE38 mean = 128 mm(3)[SD = 46 mm(3)] vs vehicle mean = 749 mm(3)[SD = 395 mm(3)], P = .001) and increased median survival (806-PE38 median = 82 days vs vehicle median = 50 days,P= .01) in a MDA-MB-468 TNBC mouse xenograft. Deletion of the catalytic residue eliminated both cytotoxic activity in vitro and the reduction in tumor burden and survival (P= .52). CONCLUSIONS: These data support the further development of the 806-PE38 immunotoxin as a therapeutic agent for the treatment of patients with EGFR-positive TNBC. Follow-up experiments with combination therapies will be attempted to achieve full remissions.


Subject(s)
ADP Ribose Transferases/therapeutic use , Bacterial Toxins/therapeutic use , ErbB Receptors/immunology , Exotoxins/therapeutic use , Immunologic Factors/therapeutic use , Immunotoxins/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/immunology , Virulence Factors/therapeutic use , ADP Ribose Transferases/pharmacology , Animals , Bacterial Toxins/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Drug Carriers , Epitopes/immunology , ErbB Receptors/genetics , Exotoxins/pharmacology , Female , Humans , Immunologic Factors/pharmacology , Immunotoxins/pharmacology , Mice , Mice, Nude , Molecular Targeted Therapy , Neoplasm Transplantation , Recombinant Proteins/therapeutic use , Survival Rate , Triple Negative Breast Neoplasms/pathology , Tumor Burden/drug effects , Virulence Factors/pharmacology , Pseudomonas aeruginosa Exotoxin A
14.
Clin Cancer Res ; 22(5): 1055-8, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26463707

ABSTRACT

Immunotoxins are targeted anticancer therapeutics that kill cancer cells using a cytotoxic bacterial toxin payload. Their development for use in solid tumor malignancies was delayed due to issues with their immunogenicity and limited therapeutic window. However, new research has rejuvenated the field. Coadministration with a lymphocyte-depleting regimen of pentostatin and cyclophosphamide can delay antidrug antibody formation, increasing the number of treatment cycles that patients can receive and resulting in durable responses in heavily pretreated patients. In addition, a new generation of immunotoxin molecules with reduced immunogenicity and nonspecific toxicity has been developed through protein engineering techniques, and one has recently entered the clinic. In preclinical studies in mouse models, these new agents are effective against many tumor types as single agents, and also produce synergistic antitumor responses in combination with chemotherapy. These new immunotoxins have renewed excitement in the field and may prove a promising addition to the targeted therapy repertoire.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Immunotherapy , Mesothelioma/drug therapy , ADP Ribose Transferases/genetics , ADP Ribose Transferases/immunology , ADP Ribose Transferases/therapeutic use , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Neoplasm/genetics , Antibodies, Neoplasm/immunology , Antibodies, Neoplasm/therapeutic use , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Bacterial Toxins/therapeutic use , Cyclophosphamide/therapeutic use , Disease Models, Animal , Exotoxins/genetics , Exotoxins/immunology , Exotoxins/therapeutic use , Humans , Lymphocytes/drug effects , Mesothelioma/genetics , Mesothelioma/pathology , Mice , Pentostatin/therapeutic use , Virulence Factors/genetics , Virulence Factors/immunology , Virulence Factors/therapeutic use , Pseudomonas aeruginosa Exotoxin A
15.
Ann Plast Surg ; 74(3): 350-3, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25643184

ABSTRACT

INTRODUCTION: Peripheral nerve regeneration over longer distances through conduits is limited. In the presented study, critical size nerve gap bridging with a poly-DL-lactide-ε-caprolactone (PLC) conduit was combined with application of C3 toxin to facilitate axonal sprouting. MATERIALS AND METHODS: The PLC filled with fibrin (n = 10) and fibrin gel loaded with 1-µg C3-C2I and 2-µg C2II (n = 10) were compared to autologous nerve grafts (n = 10) in a 15-mm sciatic nerve gap lesion model of the rat. Functional and electrophysiological analyses were performed before histological evaluation. RESULTS: Evaluation of motor function and nerve conduction velocity at 16 weeks revealed no differences between the groups. All histological parameters and muscle weight were significantly elevated in nerve graft group. No differences were observed in both PLC groups. CONCLUSIONS: The PLCs are permissive for nerve regeneration over a 15-mm defect in rats. Intraluminal application of C3 toxin did not lead to significant enhancement of nerve sprouting.


Subject(s)
ADP Ribose Transferases/therapeutic use , Bacterial Proteins/therapeutic use , Botulinum Toxins/therapeutic use , Guided Tissue Regeneration/methods , Peripheral Nervous System Agents/therapeutic use , Polyesters , Sciatic Neuropathy/therapy , Tissue Scaffolds , Animals , Biocompatible Materials , Combined Modality Therapy , Female , Nerve Regeneration/physiology , Random Allocation , Rats , Sciatic Nerve/injuries , Sciatic Nerve/pathology , Sciatic Nerve/physiopathology , Sciatic Nerve/surgery , Sciatic Neuropathy/pathology , Sciatic Neuropathy/physiopathology , Treatment Outcome
16.
Neoplasma ; 61(4): 392-400, 2014.
Article in English | MEDLINE | ID: mdl-25027740

ABSTRACT

MSH-PE38KDEL is a chimeric molecule composed of MSH, and fused to a truncated mutant form of Pseudomonas exotoxin (PE38KDEL). Our study aims to evaluate the specific cytotoxicity of recombinant immunotoxin MSH-PE38KDEL on melanoma cells A875 and B16 in vitro, as well as its inhibition of metastatic melanoma in vivo. MSH-PE38KDEL was expressed in Escherichia coli, and greater than 90% purity was obtained. The purified MSH-PE38KDEL was found to be selectively cytotoxic to MSH receptor-positive melanoma cells in vitro. The specific cytotoxicity of recombinant MSH-PE38KDEL to A875 and B16 was over 85% by cell viability assay; however, MSH-PE38KDEL had no cytotoxicity to the human 2BS cells. The anti-tumor activity of MSH-PE38KDEL was evaluated in mice with induced melanoma through intra-tumor or intravenous administration. The results showed that 90% melanoma growths were inhibited, and 40% of the tumors were disappeared completely. Histopathology results showed MSH-PE38KDEL can effectively inhibit intrahepatic metastasis. In conclusion, MSH-PE38KDEL had cytotoxic effects on MSH receptor-positive melanoma cells, and causes significant tumor growth inhibition. These results support a possible new approach for the treatment of melanoma.


Subject(s)
ADP Ribose Transferases/therapeutic use , Antineoplastic Agents/therapeutic use , Bacterial Toxins/therapeutic use , Exotoxins/therapeutic use , Immunotoxins/therapeutic use , Liver Neoplasms/drug therapy , Melanocyte-Stimulating Hormones/immunology , Melanoma, Experimental/drug therapy , Recombinant Fusion Proteins/therapeutic use , Virulence Factors/therapeutic use , Animals , Female , Humans , Immunoenzyme Techniques , In Vitro Techniques , Liver Neoplasms/immunology , Liver Neoplasms/secondary , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Nude , Tumor Cells, Cultured , Pseudomonas aeruginosa Exotoxin A
17.
World Neurosurg ; 82(3-4): e535-9, 2014.
Article in English | MEDLINE | ID: mdl-23298675

ABSTRACT

BACKGROUND: The Rho pathway has been shown to have a role in the pathophysiology of spinal cord injury (SCI). Upregulation of the Rho signaling pathway occurs as a result of SCI. Activation of Rho and its downstream effector kinases triggers growth cone collapse and represents a significant barrier to axon regeneration. Furthermore, there is evidence that Rho-ROCK signaling mediates the inhibitory effects of chondroitin sulfate proteoglycans on neurons, and that inhibition of Rho and ROCK can reverse chondroitin sulfate proteoglycan-mediated inhibition of neurite outgrowth. Work building on these findings suggests that inhibition of this pathway may boost neuroprotection and axonal regeneration after SCI. METHODS: A narrative review. RESULTS: Investigators have identified a C3 transferase, which selectively inhibits Rho without affecting other guanine triphosphatases. This has been shown to promote axonal sprouting and recovery of locomotor function after hemisection of the thoracic spinal cord in a mouse model of SCI. The neuroprotective properties of Rho inhibitors in animal models of SCI have been reinforced by studies carried out in vitro using retinal ganglion cells. In light of this, a Rho inhibitor known as Cethrin has been evaluated as a therapeutic intervention for SCI in a phase I/IIa clinical trial with promising results. CONCLUSIONS: The Rho pathway has been shown to have a role in the pathophysiology of SCI and preclinical and clinical work and is currently a promising target for the treatment of patients with SCI.


Subject(s)
Enzyme Inhibitors/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Spinal Cord Injuries/drug therapy , rho-Associated Kinases/antagonists & inhibitors , ADP Ribose Transferases/therapeutic use , Animals , Botulinum Toxins/therapeutic use , Humans , Neuroprotective Agents/therapeutic use , Signal Transduction/drug effects
18.
Acta Biochim Biophys Sin (Shanghai) ; 46(1): 6-14, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24240707

ABSTRACT

In order to develop more effective therapeutic vaccines against cancers with high-risk human papillomavirus (HPV) infection, it is crucial to enhance the immunogenicity, eliminate the oncogenicity of oncoproteins, and take a combination of E7- and E6-containing vaccines. It has been shown recently that PE(ΔIII)-E7-KDEL3 (E7), a fusion protein containing the HPV16 oncoprotein E7 and the translocation domain of Pseudomonas aeruginosa exotoxin A, is effective against TC-1 tumor cells inoculated in mice, therefore, we engineered PE(ΔIII)-E6-CRL-KDEL3 (E6), the de-oncogenic versions of the E7 and E6 fusion proteins [i.e. PE(ΔIII)-E7(d)-KDEL3, E7(d), and PE(ΔIII)-E6(d)-CRL-KDEL3, E6(d)] and tested the immunoefficacies of these fusion proteins as mono- and bivalent vaccines. Results indicated that the E7(d) get higher immunogenicity than its wild type and the E6 fusion proteins augmented the immunogenicity and antitumor effects of their E7 counterparts. Furthermore, the bivalent vaccine system E7(d) plus E6(d), in the presence of cisplatin, showed the best tumoristatic and tumoricidal effects against established tumors in vivo. Therefore, it can be concluded that this novel therapeutic vaccine system, upon further optimization, may shed new light on clinical management of HPV-related carcinomas.


Subject(s)
Cisplatin/therapeutic use , DNA-Binding Proteins/therapeutic use , Oncogene Proteins, Fusion/therapeutic use , Oncogene Proteins, Viral/therapeutic use , Papillomavirus E7 Proteins/therapeutic use , Papillomavirus Vaccines/therapeutic use , ADP Ribose Transferases/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Bacterial Toxins/therapeutic use , Cancer Vaccines/immunology , Cell Line, Tumor , Drug Synergism , Exotoxins/therapeutic use , Mice , Oncogene Proteins, Fusion/immunology , Papillomavirus Vaccines/immunology , Virulence Factors/therapeutic use , Pseudomonas aeruginosa Exotoxin A
19.
Int J Cancer ; 133(6): 1497-506, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23494569

ABSTRACT

Cytoreductive surgery and intraperitoneal (i.p.) chemotherapy constitute a curative treatment option in mucinous peritoneal surface malignancies of intestinal origin, but treatment outcome is highly variable and the search for novel therapies is warranted. Immunotoxins are attractive candidates for targeted therapy in the peritoneal cavity because of direct cytotoxicity, distinct mechanisms of action and tumor cell selectivity. The MOC31PE immunotoxin targets the tumor-associated adhesion protein EpCAM (Epithelial Cell Adhesion Molecule), and has been administered safely in early clinical trials. In our work, the efficacy of i.p. administration of MOC31PE alone and together with mitomycin C (MMC) was investigated in unique animal models of human mucinous peritoneal surface malignancies. In initial model validation experiments, clear differences in efficacy were demonstrated between MMC and oxaliplatin, favoring MMC in five investigated tumor models. Subsequently, MOC31PE and MMC were given as single i.p. injections alone and in combination. In the PMCA-2 model, moderate growth inhibition was obtained with both drugs, while the combination resulted in at least additive effects; whereas the PMP-2 model was highly sensitive to both drugs separately and in combination and intermediate sensitivity was found for the PMCA-3 model. Furthermore, results from ex vivo experiments on freshly obtained mucinous tumor tissue from animals and patients suggested that classic mechanisms of immunotoxin activity were involved, i.e., inhibition of protein synthesis and induction of apoptosis. The present results suggest that adding MOC31PE to MMC-based i.p. chemotherapy should be further explored for EpCAM-expressing peritoneal surface malignancies, and a phase I trial is in preparation.


Subject(s)
ADP Ribose Transferases/therapeutic use , Antigens, Neoplasm/immunology , Bacterial Toxins/therapeutic use , Cell Adhesion Molecules/immunology , Exotoxins/therapeutic use , Immunotoxins/therapeutic use , Peritoneal Neoplasms/drug therapy , Virulence Factors/therapeutic use , Animals , Disease Models, Animal , Epithelial Cell Adhesion Molecule , Female , Mice , Mice, Inbred BALB C , Mitomycin/therapeutic use , Organoplatinum Compounds/therapeutic use , Oxaliplatin , Peritoneal Neoplasms/pathology , Pseudomonas aeruginosa Exotoxin A
20.
Curr Pharm Des ; 19(24): 4400-10, 2013.
Article in English | MEDLINE | ID: mdl-23360272

ABSTRACT

Spinal cord injury (SCI) often results in permanent paralysis because there is little spontaneous repair. Neuronal injury in the central nervous system (CNS) causes breakage of axonal connections, release of myelin, inflammation and cell death at the lesion site. Many factors contribute to the failure of spontaneous repair after SCI, including the presence of growth inhibitory proteins in myelin, the inflammatory environment of the injured CNS, and the resulting signaling cascades that result in over-activation of Rho, a signaling switch in neurons and axons. In this review, we provide a general overview of growth inhibition in the CNS, and show evidence that most growth inhibitory proteins signal through a common intracellular pathway. Rho is a convergent signal for growth inhibition, and also for signaling some of the secondary consequences of inflammation after SCI. We review the preclinical evidence that targeting Rho is an effective way to stimulate axon regeneration and functional recovery in preclinical animal models. In the last part of the review, we describe the creation of Cethrin, a new investigational drug, and summarize the results of the Phase I/IIa clinical study to examine the safety, tolerability and efficacy of Cethrin in patients with acute SCI. We conclude with some insight for future clinical studies.


Subject(s)
ADP Ribose Transferases/therapeutic use , Botulinum Toxins/therapeutic use , Nerve Regeneration/drug effects , Neuroprotective Agents/therapeutic use , Spinal Cord Injuries/drug therapy , rho-Associated Kinases/antagonists & inhibitors , ADP Ribose Transferases/administration & dosage , ADP Ribose Transferases/adverse effects , Animals , Axons/drug effects , Axons/pathology , Botulinum Toxins/administration & dosage , Botulinum Toxins/adverse effects , Clinical Trials as Topic , Drug Evaluation, Preclinical , Humans , Motor Activity/drug effects , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/adverse effects , Spinal Cord Injuries/enzymology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...