Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Publication year range
1.
Mol Ther ; 31(12): 3478-3489, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-37919902

ABSTRACT

Mutations in the fukutin-related protein (FKRP) gene cause dystroglycanopathy, with disease severity ranging from mild LGMD2I to severe congenital muscular dystrophy. Recently, considerable progress has been made in developing experimental therapies, with adeno-associated virus (AAV) gene therapy and ribitol treatment demonstrating significant therapeutic effect. However, each treatment has its strengths and weaknesses. AAV gene therapy can achieve normal levels of transgene expression, but it requires high doses, with toxicity concerns and variable distribution. Ribitol relies on residual FKRP function and restores limited levels of matriglycan. We hypothesized that these two treatments can work synergistically to offer an optimized therapy with efficacy and safety unmatched by each treatment alone. The most effective treatment is the combination of high-dose (5e-13 vg/kg) AAV-FKRP with ribitol, whereas low dose (1e-13 vg/kg) AAV-FKRP combined with ribitol showed a 22.6% increase in positive matriglycan fibers and the greater improvement in pathology when compared to low-dose AAV-FKRP alone. Together, our results support the potential benefits of combining ribitol with AAV gene therapy for treating FKRP-related muscular dystrophy. The fact that ribitol is a metabolite in nature and has already been tested in animal models and clinical trials in humans without severe side effects provides a safety profile for it to be trialed in combination with AAV gene therapy.


Subject(s)
Muscular Dystrophies , Pentosyltransferases , Animals , Humans , Pentosyltransferases/genetics , Pentosyltransferases/metabolism , Pentosyltransferases/therapeutic use , Ribitol/metabolism , Ribitol/therapeutic use , Dependovirus/genetics , Dependovirus/metabolism , Dystroglycans/metabolism , Muscular Dystrophies/drug therapy , Genetic Therapy/methods , Mutation , Muscle, Skeletal/metabolism
2.
Cancer Gene Ther ; 20(6): 358-65, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23722591

ABSTRACT

Gene-directed enzyme prodrug therapy (GDEPT), or suicide gene therapy, has shown promise in clinical trials. In this preclinical study using stable cell lines and xenograft tumor models, we show that a triple-suicide-gene GDEPT approach produce enhanced therapeutic efficacy over previous methods. Importantly, all the three genes (thymidine kinase, cytosine deaminase and uracil phosphoribosyltransferase) function simultaneously as effectors for GDEPT and markers for multimodality molecular imaging (MMI), using positron emission tomography, magnetic resonance spectroscopy and optical (fluorescent and bioluminescent) techniques. It was demonstrated that MMI can evaluate the distribution and function/activity of the triple suicide gene. The concomitant expression of these genes significantly enhances prodrug cytotoxicity and radiosensitivity in vitro and in vivo.


Subject(s)
Cytosine Deaminase/therapeutic use , Genes, Transgenic, Suicide , Neoplasms/therapy , Pentosyltransferases/therapeutic use , Thymidine Kinase/therapeutic use , Cell Line, Tumor , Clinical Trials as Topic , Cytosine Deaminase/genetics , Genetic Therapy , Humans , Magnetic Resonance Spectroscopy , Neoplasms/genetics , Pentosyltransferases/genetics , Positron-Emission Tomography , Prodrugs/therapeutic use , Radiotherapy , Thymidine Kinase/genetics , Transfection
3.
Bioorg Khim ; 39(6): 745-8, 2013.
Article in Russian | MEDLINE | ID: mdl-25696936

ABSTRACT

In preparation of the therapeutic genetic constructs aimed to the gene-programmed enzymatic transformation of the non-toxic prodrug into toxin within cancer cells the right choice of regulatory elements (promoters and enhancers) is essential. This is widely accepted that the efficiency of the gene therapy constructions is dependent, in particular, on the strength of promoters driving the expression of the therapeutic genes. In this work we demonstrated, using the melanoma-specific promoters and enhancers of human melanoma inhibitory activity and mouse tyrosinase gene, that for the development of cytotoxic effect the promoter strength is not of primary importance. In the case of HSVtk, coding for the herpes simplex virus thymidine kinase, and FCU1, coding for cytosine deaminase/uracil phosphoribosyltransferase hybrid protein genes, their cytotoxic activity was determined by the quantity of the added prodrug.


Subject(s)
Cytosine Deaminase/genetics , Genetic Therapy , Melanoma, Experimental/genetics , Pentosyltransferases/genetics , Animals , Cytosine Deaminase/therapeutic use , Genes, Transgenic, Suicide , Humans , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Pentosyltransferases/therapeutic use , Prodrugs/therapeutic use , Promoter Regions, Genetic , Simplexvirus/enzymology , Simplexvirus/genetics
4.
Exp Neurol ; 237(1): 170-9, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22721770

ABSTRACT

After spinal cord injury, proteoglycans with growth-inhibitory glycosaminoglycan (GAG-) side chains in scar tissue limit spontaneous axonal sprouting/regeneration. Interventions that reduce scar-related inhibition facilitate an axonal growth response and possibly plasticity-based spinal cord repair. Xylosyltransferase-1 (XT-1) is the enzyme that initiates GAG-chain formation. We investigated whether intravenous administration of a deoxyribozyme (DNA enzyme) to XT-1 mRNA (DNAXT-1as) would elicit plasticity after a clinically relevant contusion of the spinal cord in adult rats. Our data showed that systemic DNAXT-1as administration resulted in a significant increase in sensorimotor function and serotonergic axon presence caudal to the injury. DNAXT1as treatment did not cause pathological or toxicological side effects. Importantly, intravenous delivery of DNAXT-1as did not exacerbate contusion-induced neuropathic pain. Collectively, our data demonstrate that DNAXT-1as is a safe neurotherapeutic, which holds promise to become an integral component of therapies that aim to improve the quality of life of persons with spinal cord injury.


Subject(s)
DNA, Catalytic/administration & dosage , Pentosyltransferases/administration & dosage , Spinal Cord Injuries/therapy , Animals , DNA, Catalytic/genetics , DNA, Catalytic/therapeutic use , Female , Injections, Intravenous , Pentosyltransferases/genetics , Pentosyltransferases/therapeutic use , RNA, Messenger/administration & dosage , RNA, Messenger/therapeutic use , Rats , Rats, Inbred F344 , Recovery of Function/genetics , Spinal Cord Injuries/enzymology , Spinal Cord Injuries/genetics , UDP Xylose-Protein Xylosyltransferase
5.
J Gene Med ; 14(7): 445-58, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22576955

ABSTRACT

BACKGROUND: Transcriptional targeted suicide gene (SG) therapy driven by the insulinoma-associated 1 (INSM1) promoter makes it possible to target suicide toxin production and cytotoxicity exclusively to small cell lung cancer (SCLC) cells and tumors. It remains to be determined whether acquired chemoresistance, as observed in the majority of SCLC patients, desensitizes SCLC cells to INSM1 promoter-driven SG therapy. METHODS: A panel of SCLC cell lines resistant to clinically relevant chemotherapeutics was characterized regarding the expression of proteins involved in response to chemotherapy and regarding INSM1 promoter activity. Sensitivity towards INSM1 promoter-driven SG therapy was tested using different systems: Yeast cytosine deaminase-uracil phosphoribosyl transferase (YCD-YUPRT) in combination with the prodrug 5-fluorocytosine (5-FC) or Escherichia coli nitroreductase (NTR) together with the bromomustard prodrug SN27686. RESULTS: The chemoresistant cell lines displayed heterogeneous expression profiles of molecules involved in multidrug resistance, apoptosis and survival pathways. Despite this, the INSM1 promoter activity was found to be unchanged or increased in SCLC chemoresistant cells and xenografts compared to chemosensitive variants. INSM1 promoter-driven SG therapy with YCD-YUPRT/5-FC or NTR/SN27686, was found to induce high levels of cytotoxicity in both chemosensitive and chemoresistant SCLC cells. Moreover, the combination of INSM1 promoter-driven YCD-YUPRT/5-FC therapy and chemotherapy, as well as the combination of INSM1 promoter-driven YCD-YUPRT/5-FC and NTR/SN27686 therapy, was observed to be superior to single agent therapy in chemoresistant SCLC cells. CONCLUSIONS: Collectively, the present study demonstrates that targeted SG therapy is a potent therapeutic approach for chemoresistant SCLC patients, with the highest efficacy achieved when applied as combination SG therapy or in combination with standard chemotherapy.


Subject(s)
Genes, Transgenic, Suicide/genetics , Genetic Therapy/methods , Lung Neoplasms/therapy , Repressor Proteins/genetics , Small Cell Lung Carcinoma/therapy , Animals , Apoptosis , Cell Line, Tumor , Cell Survival , Chemotherapy, Adjuvant , Cytosine Deaminase/genetics , Cytosine Deaminase/therapeutic use , Drug Resistance, Neoplasm , Drug Therapy, Combination , Escherichia coli/enzymology , Escherichia coli/genetics , Flucytosine/therapeutic use , Humans , Male , Mice , Nitroreductases/therapeutic use , Pentosyltransferases/genetics , Pentosyltransferases/therapeutic use , Promoter Regions, Genetic/genetics , Yeasts/enzymology , Yeasts/genetics
6.
Mol Cancer Ther ; 10(8): 1327-36, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21610170

ABSTRACT

Several antiangiogenic drugs targeting VEGF/VEGF receptor (VEGFR) that were approved by the Food and Drug Administration for many cancer types, including colorectal and lung cancer, can effectively reduce tumor growth. However, targeting the VEGF signaling pathway will probably influence the normal function of endothelial cells in maintaining homeostasis and can cause unwanted adverse effects. Indeed, emerging experimental evidence suggests that VEGF-targeting therapy induced less tumor cell-specific cytotoxicity, allowing residual cells to become more resistant and eventually develop a more malignant phenotype. We report an antitumor therapeutic EndoCD fusion protein developed by linking endostatin (Endo) to cytosine deaminase and uracil phosphoribosyltransferase (CD). Specifically, Endo possesses tumor antiangiogenesis activity that targets tumor endothelial cells, followed by CD, which converts the nontoxic prodrug 5-fluorocytosine (5-FC) to the cytotoxic antitumor drug 5-fluorouracil (5-FU) in the local tumor area. Moreover, selective targeting of tumor sites allows an increasing local intratumoral concentration of 5-FU, thus providing high levels of cytotoxic activity. We showed that treatment with EndoCD plus 5-FC, compared with bevacizumab plus 5-FU treatment, significantly increased the 5-FU concentration around tumor sites and suppressed tumor growth and metastasis in human breast and colorectal orthotropic animal models. In addition, in contrast to treatment with bevacizumab/5-FU, EndoCD/5-FC did not induce cardiotoxicity leading to heart failure in mice after long-term treatment. Our results showed that, compared with currently used antiangiogenic drugs, EndoCD possesses potent anticancer activity with virtually no toxic effects and does not increase tumor invasion or metastasis. Together, these findings suggest that EndoCD/5-FC could become an alternative option for future antiangiogenesis therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Cytosine Deaminase/therapeutic use , Endostatins/therapeutic use , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Pentosyltransferases/therapeutic use , Animals , Antineoplastic Agents/toxicity , Cell Line, Tumor , Cytosine Deaminase/genetics , Disease Models, Animal , Endostatins/genetics , Genetic Therapy , Genetic Vectors/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Pentosyltransferases/genetics , Prodrugs/metabolism , Prodrugs/pharmacology , Prodrugs/toxicity , Recombinant Fusion Proteins/therapeutic use
7.
Hum Cell ; 15(3): 138-50, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12703544

ABSTRACT

In order to develop an effective therapeutic intervention for patients with pancreatic cancer, we examined the genetic alternations of pancreatic cancer. Based on these results, we are developing a new gene therapy targeting the genetic character of pancreatic cancer using mutant adenoviruses selectively replication-competent in tumor cells. Loss of heterozygosity (LOH) of 30% or more were observed on chromosome arms 17p (47%), 9p (45%), 18q (43%), 12q (34%), and 6q (30%). LOH of 12q, 17p, and 18q showed the significant association with poor prognosis. These data strongly suggest that mutation of the putative suppressor genes, TP53 and SMAD4 play significant roles in the disease progression. Based on this rationale, we are developing a new gene therapy targeting tumors without normal TP53 function. E1B-55kDa-deleted adenovirus (AxE1AdB) can selectively replicate in TP53-deficient human tumor cells but not cells with functional TP53. We evaluated the therapeutic effect of this AxE1AdB on pancreatic cancer without normal TP53 function. The growth of human pancreatic tumor in SCID mice model was markedly inhibited by the consecutive injection of AxE1AdB. Furthermore, AxE1AdB is not only the strong weapon but also useful carrier of genes possessing anti-tumor activities as a virus vector specific to tumors without normal TP53 function. It was reported that uracil phosphoribosyl transferase (UPRT) overcomes 5FU resistance. UPRT catalyzes the synthesis of 5-fluorouridine monophosphate (FUMP) from Uracil and phosphoribosylpyrophosphate (PRPP). The antitumor effect of 5FU is enhanced by augmenting 5-fluorodeoxyuridine monophosphate (FdUMP) converted from FUMP, which inhibits Thymidylate Synthetase (TS). The therapeutic advantage of restricted replication competent adenovirus that expresses UPRT (AxE1AdB-UPRT) was evaluatedin an intra-peritoneal disseminated tumor model. To study the anti-tumor effect of AxE1AdB-UPRT/5FU, mice with disseminated AsPC-1 tumors were administered the adenovirus, followed by the 5FU treatment. It was shown that the treatment with AxE1AdB-UPRT/5FU caused a dramatic reduction of the disseminated tumor burden without toxicity in normal tissues. These results revealed thatthe AxE1AdB-UPRT/5FU system is a promising tool for intraperitoneal disseminated pancreatic cancer.


Subject(s)
Adenoviridae , Gene Targeting/methods , Genes, Tumor Suppressor , Genetic Therapy/methods , Genetic Vectors , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/therapy , Virus Replication , Adenoviridae/genetics , Adenoviridae/physiology , Animals , Fluorouracil/therapeutic use , Genes, Retinoblastoma , Genes, p53/genetics , Humans , Loss of Heterozygosity , Mice , Mutation , Pancreatic Neoplasms/pathology , Pentosyltransferases/therapeutic use , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...