Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
Adv Drug Deliv Rev ; 168: 134-146, 2021 01.
Article in English | MEDLINE | ID: mdl-32485207

ABSTRACT

Chronic infection with the hepatitis B virus (HBV) remains a significant worldwide medical problem. While diseases caused by HIV infection, tuberculosis and malaria are on the decline, new cases of chronic hepatitis B are on the rise. Because often fatal complications of cirrhosis and hepatocellular carcinoma are associated with chronic hepatitis B, the need for a cure is as urgent as ever. Currently licensed therapeutics fail to eradicate the virus and this is attributable to persistence of the viral replication intermediate comprising covalently closed circular DNA (cccDNA). Elimination or inactivation of the viral cccDNA is thus a goal of research aimed at hepatitis B cure. The ability to engineer nucleases that are capable of specific cleavage of a DNA sequence now provides the means to disable cccDNA permanently. The scientific literature is replete with many examples of using designer zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs) and RNA-guided endonucleases (RGENs) to inactivate HBV. However, important concerns about safety, dose control and efficient delivery need to be addressed before the technology is employed in a clinical setting. Use of in vitro transcribed mRNA to express therapeutic gene editors goes some way to overcoming these concerns. The labile nature of RNA limits off-target effects and enables dose control. Compatibility with hepatotropic non-viral vectors is convenient for the large scale preparation that will be required for advancing gene editing as a mode of curing chronic hepatitis B.


Subject(s)
Genetic Therapy/methods , Hepatitis B, Chronic/genetics , Hepatitis B, Chronic/therapy , RNA, Messenger/administration & dosage , Antiviral Agents/therapeutic use , Clustered Regularly Interspaced Short Palindromic Repeats , Dose-Response Relationship, Drug , Gene Editing , Genetic Vectors/administration & dosage , Hepatitis B Vaccines/administration & dosage , Hepatitis B, Chronic/drug therapy , Hepatitis B, Chronic/prevention & control , Nanoparticles/chemistry , Ribonucleases/administration & dosage , Transcription Activator-Like Effector Nucleases/metabolism , Zinc Finger Nucleases/metabolism
2.
Viruses ; 12(2)2020 02 05.
Article in English | MEDLINE | ID: mdl-32033253

ABSTRACT

Currently, no rabies virus-specific antiviral drugs are available. Ranpirnase has strong antitumor and antiviral properties associated with its ribonuclease activity. TMR-001, a proprietary bulk drug substance solution of ranpirnase, was evaluated against rabies virus in three cell types: mouse neuroblastoma, BSR (baby hamster kidney cells), and bat primary fibroblast cells. When TMR-001 was added to cell monolayers 24 h preinfection, rabies virus release was inhibited for all cell types at three time points postinfection. TMR-001 treatment simultaneous with infection and 24 h postinfection effectively inhibited rabies virus release in the supernatant and cell-to-cell spread with 50% inhibitory concentrations of 0.2-2 nM and 20-600 nM, respectively. TMR-001 was administered at 0.1 mg/kg via intraperitoneal, intramuscular, or intravenous routes to Syrian hamsters beginning 24 h before a lethal rabies virus challenge and continuing once per day for up to 10 days. TMR-001 at this dose, formulation, and route of delivery did not prevent rabies virus transit from the periphery to the central nervous system in this model (n = 32). Further aspects of local controlled delivery of other active formulations or dose concentrations of TMR-001 or ribonuclease analogues should be investigated for this class of drugs as a rabies antiviral therapeutic.


Subject(s)
Antiviral Agents/pharmacology , Rabies virus/drug effects , Ribonucleases/pharmacology , Virus Release/drug effects , Virus Replication/drug effects , Animals , Cell Line , Cells, Cultured , Chiroptera , Cricetinae , Female , Fibroblasts/virology , Mesocricetus , Mice , Rabies/prevention & control , Rabies virus/physiology , Ribonucleases/administration & dosage
3.
ACS Chem Biol ; 14(4): 599-602, 2019 04 19.
Article in English | MEDLINE | ID: mdl-30830748

ABSTRACT

A major hurdle in chemical biology is the delivery of native proteins into the cytosol of mammalian cells. Herein, we report that esterification of the carboxyl groups of an enzyme with a diazo compound enables not only its passage into the cytosol but also the retention of its catalytic activity there. This scenario is demonstrated with human ribonuclease 1, which manifests ribonucleolytic activity that can be cytotoxic. After internalization, the nascent esters are hydrolyzed in situ by endogenous esterases, making the process traceless. This strategy provides unprecedented opportunities for the delivery of functional enzymes into human cells.


Subject(s)
Esterases/administration & dosage , Ribonucleases/administration & dosage , Biocatalysis , Esterases/metabolism , Esterification , Humans , Proteolysis , Ribonucleases/metabolism
4.
J Control Release ; 263: 39-45, 2017 Oct 10.
Article in English | MEDLINE | ID: mdl-28153764

ABSTRACT

Developing safe and effective nanosystems to deliver active and therapeutic proteins to targeted cells and organs is an important tool for many biomedical applications. We present here a simple and efficient strategy for this purpose: delivering hyaluronic acid (HA)-modified RNase A (RNase A-HA) in nanocomplex with cationic lipid-like molecules (lipidoids) to cancer cells, resulting in targeted inhibition of cancer proliferation. The chemical conjugation of RNase A with HA both increased the supramolecular interaction with carrier lipidoids, promoting protein encapsulation efficacy, and facilitated cancer cell targeting via interaction with overexpressed CD44. Through confocal laser scanning microscopy and flow cytometry analysis, we demonstrated that protein/lipidoid nanoparticles could facilely enter cells with high CD44 expression, and inhibit cell proliferation in a dose-dependent manner.


Subject(s)
Hyaluronic Acid/administration & dosage , Lipids/administration & dosage , Nanoparticles/administration & dosage , Ribonucleases/administration & dosage , A549 Cells , Cell Survival/drug effects , Humans , Hyaluronan Receptors/metabolism , Hyaluronic Acid/chemistry , Lipids/chemistry , MCF-7 Cells , Nanoparticles/chemistry , Neoplasms/drug therapy , Neoplasms/metabolism , Ribonucleases/chemistry
5.
Biomaterials ; 122: 163-178, 2017 04.
Article in English | MEDLINE | ID: mdl-28126663

ABSTRACT

MicroRNAs (miRNAs) are active regulators in malignant growth and constitute potential targets for anticancer therapy. Consequently, considerable effort has focused on identifying effective ways to modulate aberrant miRNA expression. Here we introduce and assess a novel type of chemically engineered biomaterial capable of cleaving specific miRNA sequences, i.e. miRNA-specific artificial ribonucleases (hereafter 'miRNase'). The miRNase template presented here consists of the catalytic peptide Acetyl-[(LeuArg)2Gly]2 covalently attached to a miRNA-targeting oligonucleotide, which can be linear or hairpin. The peptide C-terminus is conjugated to an aminohexyl linker located at either the 3'- or 5'-end of the oligonucleotide. The cleavage efficacy, structural aspects of cleavage and biological relevance of a set of these designed miRNases was assayed with respect to highly oncogenic miR-21. Several miRNases demonstrated effective site-selective cleavage of miR-21 exclusively at G-X bonds. One of the most efficient miRNase was shown to specifically inhibit miR-21 in lymphosarcoma cells and lead to a reduction in their proliferative activity. This report provides the first experimental evidence that metallo-independent peptide-oligonucleotide chemical ribonucleases are able to effectively and selectively down-regulate oncogenic miRNA in tumour cells, thus suggesting their potential in development of novel therapeutics aimed at overcoming overexpression of disease-related miRNAs.


Subject(s)
Lymphoma, Non-Hodgkin/genetics , Lymphoma, Non-Hodgkin/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Oligonucleotides/administration & dosage , Peptides/administration & dosage , Ribonucleases/administration & dosage , Animals , Cell Line, Tumor , Mice , Oligonucleotides/chemistry , Peptides/chemistry , Ribonucleases/chemistry
6.
Acta Biochim Biophys Sin (Shanghai) ; 48(10): 894-901, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27590062

ABSTRACT

Onconase (Onc) is a cytotoxic ribonuclease derived from leopard frog oocytes or early embryos, and has been applied to the treatment of malignant mesothelioma in clinics. Onc also exhibits effective growth suppression of human non-small-cell lung cancer (NSCLC). Artemisinin (Art) and its derivatives are novel antimalarial drugs that exhibit antitumor and antivirus activities. In this study, we investigated the antitumor effects of combinations of Onc and an Art derivative, dihydroartemisinin (DHA), both in vitro and in vivo Isobologram analyses showed synergistic effects on the proliferation of NSCLC cells under the treatment with Onc and DHA. In vivo experiments also showed that the antitumor effect of Onc was markedly enhanced by DHA in mouse xenograft models. No obvious adverse effect was observed after the treatment. The density of microvasculature in the tumor tissues treated with Onc/DHA combination was lower than those treated with Onc or DHA alone. The above results are consistent with the results of the matrigel plug test for angiogenesis suppression using the Onc/DHA combination. These results imply that the anti-angiogenesis effects may make important contributions to the in vivo antitumor effects of the Onc/DHA combination treatment. The Onc/DHA combination therapy may have the potential to become a novel regimen for NSCLC and mesothelioma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Proliferation/drug effects , Lung Neoplasms/drug therapy , Mesothelioma/drug therapy , Neovascularization, Pathologic/prevention & control , A549 Cells , Animals , Antimalarials/administration & dosage , Antimalarials/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Artemisinins/administration & dosage , Artemisinins/pharmacology , Carcinoma, Non-Small-Cell Lung/blood supply , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line , Cell Line, Tumor , Drug Synergism , Humans , Immunohistochemistry , Lung Neoplasms/blood supply , Lung Neoplasms/metabolism , Male , Mesothelioma/blood supply , Mesothelioma/metabolism , Mesothelioma, Malignant , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/metabolism , Ribonucleases/administration & dosage , Ribonucleases/pharmacology , Tumor Burden/drug effects , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
7.
ACS Chem Biol ; 11(2): 319-23, 2016 Feb 19.
Article in English | MEDLINE | ID: mdl-26629587

ABSTRACT

The use of exogenous proteins as intracellular probes and therapeutic agents is in its infancy. A major hurdle has been the delivery of native proteins to an intracellular site of action. Herein, we report on a compact delivery vehicle that employs the intrinsic affinity of boronic acids for the carbohydrates that coat the surface of mammalian cells. In the vehicle, benzoxaborole is linked to protein amino groups via a "trimethyl lock." Immolation of this linker is triggered by cellular esterases, releasing native protein. Efficacy is demonstrated by enhanced delivery of green fluorescent protein and a cytotoxic ribonuclease into mammalian cells. This versatile strategy provides new opportunities in chemical biology and pharmacology.


Subject(s)
Boronic Acids/chemistry , Drug Carriers/chemistry , Green Fluorescent Proteins/administration & dosage , Ribonucleases/administration & dosage , Animals , Boronic Acids/metabolism , CHO Cells , Cell Line, Tumor , Cell Membrane Permeability , Cricetulus , Drug Carriers/metabolism , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/pharmacokinetics , Humans , Models, Molecular , Ribonucleases/chemistry , Ribonucleases/pharmacokinetics
8.
Viruses ; 7(9): 5133-44, 2015 Sep 21.
Article in English | MEDLINE | ID: mdl-26402693

ABSTRACT

The antiviral effect of a catalytic RNA-hydrolyzing antibody, 3D8 scFv, for intranasal administration against avian influenza virus (H1N1) was described. The recombinant 3D8 scFv protein prevented BALB/c mice against H1N1 influenza virus infection by degradation of the viral RNA genome through its intrinsic RNA-hydrolyzing activity. Intranasal administration of 3D8 scFv (50 µg/day) for five days prior to infection demonstrated an antiviral activity (70% survival) against H1N1 infection. The antiviral ability of 3D8 scFv to penetrate into epithelial cells from bronchial cavity via the respiratory mucosal layer was confirmed by immunohistochemistry, qRT-PCR, and histopathological examination. The antiviral activity of 3D8 scFv against H1N1 virus infection was not due to host immune cytokines or chemokines, but rather to direct antiviral RNA-hydrolyzing activity of 3D8 scFv against the viral RNA genome. Taken together, our results suggest that the RNase activity of 3D8 scFv, coupled with its ability to penetrate epithelial cells through the respiratory mucosal layer, directly prevents H1N1 virus infection in a mouse model system.


Subject(s)
Antibodies, Catalytic/administration & dosage , Antiviral Agents/administration & dosage , Epithelial Cells/immunology , Influenza A Virus, H1N1 Subtype/immunology , Orthomyxoviridae Infections/prevention & control , Ribonucleases/administration & dosage , Single-Chain Antibodies/administration & dosage , Administration, Intranasal , Animals , Antiviral Agents/pharmacokinetics , Hydrolysis , Mice, Inbred BALB C , RNA, Viral/metabolism , Single-Chain Antibodies/pharmacokinetics , Treatment Outcome
9.
J Control Release ; 212: 85-93, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26091921

ABSTRACT

The low efficiency of endosomal escape has been considered a bottleneck for the cytosolic delivery of biomacromolecules such as proteins and DNA. Although fusogenic peptides (FPs) such as HA2 have been employed to improve the intracellular delivery of biomacromolecules, the FPs studied thus far are not adequately efficient in enabling endosomal escape; therefore, novel FPs with higher activity are required. In this context, we focused on FPs derived from a sea urchin fertilization protein, bindin, which is involved in gamete recognition (B18, residues 103-120 and B55, residues 83-137 of mature bindin). We show that enhanced green fluorescent protein (EGFP)-fused B55 peptide binds to plasma membranes more strongly than EGFP-B18 and promotes the intracellular delivery of dextrans, which were co-administered using the trans method in a pH-dependent manner without affecting cell viability and proliferation, whereas conventional EGFP-HA2 did not affect dextran internalization. Furthermore, EGFP-B55 promoted the intracellular delivery of biomacromolecules such as antibodies, ribonuclease and plasmidic DNA using the trans method. Because the promotion of intracellular delivery by EGFP-B55 was suppressed by endocytosis inhibitors, EGFP-B55 is considered to have facilitated the endosomal escape of co-administered cargos. These results suggested that an FP that promotes the intracellular delivery of a variety of biomacromolecules with no detectable cytotoxicity should be useful for the cytosolic delivery of membrane-impermeable molecules for biomedical and biotechnological applications.


Subject(s)
Drug Delivery Systems , Green Fluorescent Proteins/chemistry , Peptides/chemistry , Receptors, Cell Surface/chemistry , Cell Survival/drug effects , DNA/administration & dosage , DNA/chemistry , Dextrans/administration & dosage , Dextrans/chemistry , Endocytosis , Endosomes/metabolism , Fertilization , HeLa Cells , Humans , Immunoglobulin G/administration & dosage , Immunoglobulin G/chemistry , Luciferases/genetics , Ribonucleases/administration & dosage , Ribonucleases/chemistry
10.
Int J Cancer ; 136(9): 2228-40, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25303768

ABSTRACT

Antibody therapy of solid cancers is well established, but suffers from unsatisfactory tumor penetration of large immunoglobulins or from low serum retention of antibody fragments. Oncolytic viruses are in advanced clinical development showing excellent safety, but suboptimal potency due to limited virus spread within tumors. Here, by developing an immunoRNase-encoding oncolytic adenovirus, we combine viral oncolysis with intratumoral genetic delivery of a small antibody-fusion protein for targeted bystander killing of tumor cells (viro-antibody therapy). Specifically, we explore genetic delivery of a small immunoRNase consisting of an EGFR-binding scFv antibody fragment fused to the RNase Onconase (ONC(EGFR)) that induces tumor cell death by RNA degradation after cellular internalization. Onconase is a frog RNase that combines lack of immunogenicity and excellent safety in patients with high tumor killing potency due to its resistance to the human cytosolic RNase inhibitor. We show that ONC(EGFR) expression by oncolytic adenoviruses is feasible with an optimized, replication-dependent gene expression strategy. Virus-encoded ONC(EGFR) induces potent and EGFR-dependent bystander killing of tumor cells. Importantly, the ONC(EGFR)-encoding oncolytic adenovirus showed dramatically increased cytotoxicity specifically to EGFR-positive tumor cells in vitro and significantly enhanced therapeutic activity in a mouse xenograft tumor model. The latter demonstrates that ONC(EGFR) is expressed at levels sufficient to trigger tumor cell killing in vivo. The established ONC(EGFR)-encoding oncolytic adenovirus represents a novel agent for treatment of EGFR-positive tumors. This viro-antibody therapy platform can be further developed for targeted/personalized cancer therapy by exploiting antibody diversity to target further established or emerging tumor markers or combinations thereof.


Subject(s)
Adenoviridae/genetics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/metabolism , Genetic Vectors/genetics , Oncolytic Viruses/genetics , Ribonucleases/administration & dosage , Ribonucleases/metabolism , Animals , Antibodies, Viral , Cell Line, Tumor , ErbB Receptors/metabolism , Female , Genetic Therapy/methods , Humans , Mice , Mice, Inbred BALB C , Oncolytic Virotherapy/methods , RNA/metabolism , Virus Replication/drug effects , Xenograft Model Antitumor Assays
11.
Bioorg Khim ; 41(5): 599-605, 2015.
Article in Russian | MEDLINE | ID: mdl-26762098

ABSTRACT

Targeted delivery of semiconductor quantum dots (Q Ds) to tumors overexpressing HER2 cancer marker has been. demonstrated on immunocompromised mice bearing human breast cancer xenografts. To obtain targeted QDs complexes we applied the approach based on the use of protein adaptor system, RNAase barnase and its inhibitor barstar. Specific binding to target cancer marker was achieved through bivalent fusion protein containing two fragments of4D5scFv recombinant antibody and a fragment of barnase. QDs were conjugated to barstar, and final assembly of targeted complexes was obtained through non-covalent specific interaction of barstar, attached to QD, and barnase, that is part of the recombinant targeting protein. The efficient delivery of QDs to HER2-expressing tumor demonstrates the possibilities and prospects of the approach for targeted delivery of nanoparticles to cancer cells in vivo as the way to improve the efficiency of diagnosis and promote development of therapies based on the use of nanoparticles.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Gene Targeting , Mammary Neoplasms, Experimental/genetics , Quantum Dots/administration & dosage , Receptor, ErbB-2/genetics , Recombinant Fusion Proteins/administration & dosage , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Bacterial Proteins/administration & dosage , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Cell Line, Tumor , Humans , Mice, Inbred BALB C , Mice, Nude , Quantum Dots/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Ribonucleases/administration & dosage , Ribonucleases/chemistry , Ribonucleases/genetics
12.
Exp Anim ; 63(3): 349-55, 2014.
Article in English | MEDLINE | ID: mdl-25077765

ABSTRACT

Reproductive engineering techniques, such as in vitro fertilization (IVF) and cryopreservation of embryos or spermatozoa, are essential for preservation, reproduction, and transportation of genetically engineered mice. However, it has not yet been elucidated whether these techniques can be applied for the generation of genome-edited mice using engineered nucleases such as transcription activator-like effector nucleases (TALENs). Here, we demonstrate the usefulness of frozen oocytes fertilized in vitro using frozen sperm for TALEN-mediated genome editing in mice. We examined side-by-side comparisons concerning sperm (fresh vs. frozen), fertilization method (mating vs. IVF), and fertilized oocytes (fresh vs. frozen) for the source of oocytes used for TALEN injection; we found that fertilized oocytes created under all tested conditions were applicable for TALEN-mediated mutagenesis. In addition, we investigated whether the ages in weeks of parental female mice can affect the efficiency of gene modification, by comparing 5-week-old and 8-12-week-old mice as the source of oocytes used for TALEN injection. The genome editing efficiency of an endogenous gene was consistently 95-100% when either 5-week-old or 8-12-week-old mice were used with or without freezing the oocytes. Thus, our report describes the availability of freeze-thawed oocytes and oocytes from female mice at various weeks of age for TALEN-mediated genome editing, thus boosting the convenience of such innovative gene targeting strategies.


Subject(s)
Animals, Genetically Modified/genetics , Cryopreservation/methods , Gene Targeting/methods , Genome/genetics , Oocytes , Reproductive Techniques , Ribonucleases/genetics , Animals , Female , Fertilization in Vitro , Male , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Microinjections , Ribonucleases/administration & dosage , Semen Preservation , Spermatozoa , Transcriptional Activation
13.
Int J Oncol ; 45(1): 419-25, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24789756

ABSTRACT

Although major advancements in antitumor treatment have been observed, several B cell-derived malignancies still remain incurable. A promising approach that involves targeting RNA either by the use of specific antisense oligonucleotides or cytostatic/cytotoxic ribonucleases (RNases) is being promoted. Two amphibian RNases, onconase (ONC; ranpirnase) and, more recently, r-amphinase (r-Amph), have already been tested, but thus far, mostly on solid tumors. In this study, for the first time we provide comprehensive data on ex vivo and in vivo cytotoxic activity of ONC or r-Amph against cancer cells from different B cell lymphoid malignancies, together with their detailed mode of antitumor action. Our data revealed strong pro-apoptotic activity of ONC and r-Amph in both chronic lymphocytic leukemia and aggressive B cell lymphomas, with less impact on acute lymphoblastic leukemia or multiple myeloma cells. Moreover, the antitumor action of ONC and r-Amph was markedly selective against neoplastic cells sparing normal, healthy control­derived lymphocytes.


Subject(s)
Antineoplastic Agents/administration & dosage , B-Lymphocytes/pathology , Doxorubicin/administration & dosage , Drug Resistance, Neoplasm/drug effects , Lymphoproliferative Disorders/pathology , Ribonucleases/administration & dosage , Adult , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols , Cell Line, Tumor , Dose-Response Relationship, Drug , Doxorubicin/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Middle Aged , Ribonucleases/pharmacology , Young Adult
14.
Postepy Hig Med Dosw (Online) ; 67: 1166-72, 2013 Dec 02.
Article in English | MEDLINE | ID: mdl-24379257

ABSTRACT

Ranpirnase (onconase; ONC) is an endoribonuclease obtained from the frog Rana pipiens. This enzyme exhibits anticancer properties mediated by degradation of cellular RNA and induction of apoptosis. In this study we assessed cytotoxicity of ONC in combination with currently used anticancer drugs on a human diffuse large B-cell lymphoma (DLBCL)-derived cell line (Toledo). Cytotoxic activity was measured by the exclusion of propidium iodide assay while apoptosis was assessed using the annexin-V binding method. Additionally, flow cytometry was used to assess the decline of mitochondrial potential and to determine activation of caspases 3, 8 and 9. It was observed that in vitro treatment with ONC in combination with rituximab, mafosfamide, vincristine, doxorubicin, and dexamethasone (drugs corresponding with elements of R-CHOP regimen) resulted in increased cytotoxicity. As a result ONC showed marked cytotoxicity against Toledo cells. Importantly, in combination of ONC with drugs imitating the R-CHOP regimen, this effect was significantly intensified. The main mechanism responsible for this event was induction of apoptosis along a mitochondrial dependent pathway. In conclusion, these data indicate that further preclinical and eventually clinical studies assessing activity of ONC+R-CHOP treatment are warranted.


Subject(s)
Antibodies, Antineutrophil Cytoplasmic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Lymphoma, Large B-Cell, Diffuse/drug therapy , Ribonucleases/administration & dosage , Antibodies, Monoclonal, Murine-Derived/administration & dosage , Apoptosis/drug effects , Cell Line , Cyclophosphamide/administration & dosage , Doxorubicin/administration & dosage , Humans , Prednisone/administration & dosage , Rituximab , Treatment Outcome , Vincristine/administration & dosage
15.
J Control Release ; 159(3): 346-52, 2012 May 10.
Article in English | MEDLINE | ID: mdl-22715504

ABSTRACT

The unfavorable pharmacokinetics and low tumor specificity hampered the potential clinical utility of Onconase, a promising modality in anticancer treatment with unique targets and novel mechanism of action. In this study, a modular and multi-stage drug delivery system (DDS) that can break down organ (renal accumulation), cellular (cancer cell specific uptake) and sub-cellular (endosomal escape) level barriers encountered by Onconase during its long journey from injection site to the cytoplasm of cancer cell was designed. Human serum albumin fusion extended the half-life of Onconase and significantly decreased its kidney accumulation. Epithelial cell adhesion molecular (EpCAM) specific antibody fragment appending enhanced binding and internalization of Onconase toward EpCAM positive cancer cell and increased its tumor accumulation and retention. Tethering Onconase to its carrier by cleavable disulfide linker prompted endosomal escape and restored its cytotoxicity. In vivo antitumor efficacy assay in human tumor xenograft model revealed that only when the entire organ, cellular and sub-cellular level barriers had been broken down, will Onconase turn into a potent antitumor agent.


Subject(s)
Antineoplastic Agents/administration & dosage , Cytosol/metabolism , Drug Delivery Systems/methods , Ribonucleases/administration & dosage , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Cell Survival/drug effects , Female , HT29 Cells , Humans , Kidney/drug effects , Kidney/metabolism , Kidney Function Tests , Liver/drug effects , Liver/metabolism , Liver Function Tests , Metabolic Clearance Rate , Mice , Mice, Inbred BALB C , Pichia/genetics , Recombinant Fusion Proteins/genetics , Ribonucleases/genetics , Ribonucleases/pharmacokinetics , Ribonucleases/pharmacology , Ribonucleases/toxicity , Time Factors , Tissue Distribution , Xenograft Model Antitumor Assays
16.
Inflamm Res ; 59(11): 971-8, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20473630

ABSTRACT

OBJECTIVE AND DESIGN: A 6-month, randomized clinical study was conducted to evaluate the effect of a ribonuclease-enriched lactoferrin (R-ELF) supplement on the circulating cytokine levels and bone health of postmenopausal women. SUBJECTS: Thirty-eight healthy postmenopausal women, aged 45-60 years, were randomized into placebo and R-ELF groups. TREATMENT: The R-ELF group was supplemented with R-ELF (2 × 125 mg/day) and calcium (100% RDA), while the placebo group received only the calcium supplement. METHODS: Serum levels of receptor activator for NF-κB ligand (RANKL), C-reactive protein (CRP) and various pro- and anti-inflammatory cytokines were determined by ELISA. RESULTS: Pro-inflammatory cytokines IL-6 and TNF-α decreased significantly (-44 and -10%, respectively) while anti-inflammatory IL-10 increased (140%) due to R-ELF supplementation at the end of study. RANKL and CRP were modestly reduced (-50%) relative to their placebo levels, although RANKL elevated initially. CONCLUSIONS: R-ELF supplementation showed beneficial effects towards improvement of inflammatory status in postmenopausal women.


Subject(s)
Dietary Supplements , Inflammation/immunology , Lactoferrin , Milk/enzymology , Postmenopause , Ribonucleases/administration & dosage , Animals , Cattle , Cytokines/blood , Cytokines/immunology , Female , Humans , Inflammation/blood , Lactoferrin/administration & dosage , Lactoferrin/chemistry , Middle Aged , Placebos
17.
Eur J Pharmacol ; 625(1-3): 181-9, 2009 Dec 25.
Article in English | MEDLINE | ID: mdl-19825371

ABSTRACT

Antitumor ribonucleases are small (10-28 kDa) basic proteins. They were found among members of both, ribonuclease A and T1 superfamilies. Their cytotoxic properties are conferred by enzymatic activity, i.e., the ability to catalyze cleavages of phosphodiester bonds in RNA. They bind to negatively charged cell membrane, enter cells by endocytosis and translocate to cytosol where they evade mammalian protein ribonuclease inhibitor and degrade RNA. Here, we discuss structures, functions and mechanisms of antitumor activity of several cytotoxic ribonucleases with particular emphasis to the amphibian Onconase, the only enzyme of this class that reached clinical trials. Onconase is the smallest, very stable, less catalytically efficient and more cytotoxic than most RNase A homologues. Its cytostatic, cytotoxic and anticancer effects were extensively studied. It targets tRNA, rRNA, mRNA as well as the non-coding RNA (microRNAs). Numerous cancer lines are sensitive to Onconase; their treatment with 10-100 nM enzyme leads to suppression of cell cycle progression, predominantly through G(1), followed by apoptosis or cell senescence. Onconase also has anticancer properties in animal models. Many effects of this enzyme are consistent with the microRNAs, one of its critical targets. Onconase sensitizes cells to a variety of anticancer modalities and this property is of particular interest, suggesting its application as an adjunct to chemotherapy or radiotherapy in treatment of different tumors. Cytotoxic RNases as exemplified by Onconase represent a new class of antitumor agents, with an entirely different mechanism of action than the drugs currently used in the clinic. Further studies on animal models including human tumors grafted on severe combined immunodefficient (SCID) mice and clinical trials are needed to explore clinical potential of cytotoxic RNases.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Ribonucleases/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Cell Cycle/drug effects , Dose-Response Relationship, Drug , Drug Delivery Systems , Humans , Neoplasms/physiopathology , Rana pipiens , Ribonucleases/administration & dosage
18.
J Immunol ; 183(6): 4013-20, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19717523

ABSTRACT

Eosinophil granule proteins are deposited in cutaneous lesions in many human diseases, but how these proteins contribute to pathophysiology is obscure. We injected eosinophil cationic protein (ECP or RNase 3), eosinophil-derived neurotoxin (EDN or RNase 2), eosinophil peroxidase (EPO), and major basic protein-1 (MBP1) intradermally into guinea pig and rabbit skin. ECP and EDN each induced distinct skin lesions at >or=2.5 microM that began at 2 days, peaking at approximately 7 days and persisting up to 6 wk. These lesions were ulcerated (ECP) or crusted (EDN) with marked cellular infiltration. EPO and MBP1 (10 microM) each produced perceptible induration and erythema with moderate cellular infiltration resolving within 2 wk. ECP and EDN localized to dermal cells within 2 days, whereas EPO and MBP1 remained extracellular. Overall, cellular localization and RNase activity of ECP and EDN were critical for lesion formation; differential glycosylation, net cationic charge, or RNase activity alone did not account for lesion formation. Ulcerated lesions from patients with the hypereosinophilic syndrome showed ECP and EDN deposition comparable to that in guinea pig skin. In conclusion, ECP and EDN disrupt skin integrity and cause inflammation. Their presence in ulcerative skin lesions may explain certain findings in human eosinophil-associated diseases.


Subject(s)
Eosinophil Granule Proteins/toxicity , Eosinophils/enzymology , Ribonucleases/toxicity , Skin Diseases/etiology , Animals , Eosinophil Cationic Protein/administration & dosage , Eosinophil Cationic Protein/toxicity , Eosinophil Granule Proteins/administration & dosage , Eosinophil Major Basic Protein/administration & dosage , Eosinophil Major Basic Protein/toxicity , Eosinophil Peroxidase/administration & dosage , Eosinophil Peroxidase/toxicity , Eosinophil-Derived Neurotoxin/administration & dosage , Eosinophil-Derived Neurotoxin/toxicity , Eosinophilia/pathology , Guinea Pigs , Humans , Rabbits , Ribonucleases/administration & dosage , Skin Diseases/pathology , Ulcer/etiology
19.
Pharm Res ; 26(8): 1838-46, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19415468

ABSTRACT

PURPOSE: Antineoplastic RNAse proteins, also known as Amphibinases, have been shown effective against various solid tumors but were found selectively neurotoxic to Purkinje cells in the cerebellum. This work describes the use of a waxy biodegradable poly(ricinoleic-co-sebacic acid) for the local controlled delivery of cytotoxic amphibinases in the parietal lobe of the brain in an attempt to overcome cerebellar neuronal toxicity while affecting glioma cells. METHODS: Amphibinase analogues were encapsulated in poly(ricinoleic-co-sebacic acid) formulations using mix-melt technology and loaded onto surgical foam. In-vitro release was monitored by BCA colorimetry and by RNAse specific bioactivity. The implants were inserted into rat brains bearing 9L glioma to assess toxicity and efficacy. RESULTS: The various formulations showed extended linear release for several weeks with minimal burst effect. Best in-vivo efficacy was obtained with ACC7201 containing implants, resulting in the extension of the median survival from 13 to 18 days with 13% long-term survivors. CONCLUSION: Antineoplastic proteins were released from a p(SA-RA) polyanhydride implants in a controlled manner, providing efficacy against 9L glioma, while evading neurotoxicity in the cerebellum. The controlled release of Amphibinases forms the potential for a new therapy against brain tumors.


Subject(s)
Antineoplastic Agents/administration & dosage , Brain Neoplasms/metabolism , Brain/metabolism , Glioma/metabolism , Ribonucleases/administration & dosage , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Brain Neoplasms/drug therapy , Cell Line, Tumor , Glioma/drug therapy , Rats , Ribonucleases/therapeutic use , Ribonucleases/toxicity
20.
Curr Pharm Biotechnol ; 9(3): 180-4, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18673283

ABSTRACT

The cytotoxic properties of naturally occurring or engineered RNases correlate well with their efficiency of cellular internalization and digestion level of cellular RNA. Cationized RNases are considered to adsorb to the anionic cellular surface by Coulombic interactions, and then become efficiently internalized into cells by an endocytosis-like pathway. The design of cytotoxic RNases by chemical modification of surface carboxylic residues is one of the powerful strategies for enhancing cellular internalization and is accompanied with a decreased sensitivity for the cytoplasmic RNase inhibitor. Although chemically modified cationized RNases showed decreased ribonucleolytic activity, improved endocytosis and decreased affinity to the endogenous RNase inhibitor conclusively contribute to their ability to digest cellular RNA. Furthermore, the cytotoxicity of cationized RNases can be drastically enhanced by co-endocytosis with an endosome-destabilizing peptide. Since efficient cellular internalization of proteins into living cells is an important technology for biotechnology, studies concerning the design of cytotoxic RNases provided general perceptions for protein-based drug design.


Subject(s)
Antineoplastic Agents , Drug Design , Endocytosis , Endosomes , Ribonucleases , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cations , Diamines/chemistry , Endocytosis/drug effects , Endosomes/drug effects , Endosomes/metabolism , Humans , Neoplasms/drug therapy , Polymers/chemistry , Protein Engineering , Ribonucleases/administration & dosage , Ribonucleases/chemistry , Ribonucleases/genetics , Ribonucleases/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...