Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Gene Ther ; 17(7): 892-904, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20237509

ABSTRACT

Eighteen patients with refractory and progressive solid tumors were treated with a single round of triple modified oncolytic adenovirus (Ad5/3-Cox2L-D24). Ad5/3-Cox2L-D24 is the first non-Coxsackie-adenovirus receptor-binding oncolytic adenovirus used in humans. Grades 1-2 flu-like symptoms, fever, and fatigue were seen in most patients, whereas transaminitis or thrombocytopenia were seen in some. Non-hematological grades 3-5 side effects were seen in one patient with grade 3 ileus. Treatment resulted in high neutralizing antibody titers within 3 weeks. Virus appeared in serum 2-4 days after treatment in 83% of patients and persisted for up to 5 weeks. One out of five radiologically evaluable patients had partial response (PR), one had minor response (MR), and three had progressive disease (PD). Two patients scored as PD had a decrease in tumor density. Tumor reductions not measurable with Response Evaluation Criteria In Solid Tumors (RECIST) were seen in a further four patients. PR, MR, stable disease, and PD were seen in 12, 23.5, 35, and 29.5% of tumor markers analyzed, respectively (N=17). Ad5/3-Cox2L-D24 appears safe for treatment of cancer in humans and extended virus circulation results from a single treatment. Objective evidence of anti-tumor activity was seen in 11/18 (61%) of patients. Clinical trials are needed to extend these findings.


Subject(s)
Adenoviridae , Neoplasms/therapy , Oncolytic Virotherapy/methods , Adenoviridae/isolation & purification , Adult , Aged , Antibodies, Viral , Child, Preschool , Female , Humans , Liver/enzymology , Male , Middle Aged , Neoplasm Metastasis , Neoplasms/blood , Neoplasms/pathology , Neoplasms/virology , Oncolytic Virotherapy/adverse effects , Treatment Outcome
2.
Gene Ther ; 15(12): 921-9, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18401431

ABSTRACT

Prior infection has primed most adult humans for a rapid neutralizing antibody (NAb) response when re-exposed to adenovirus. NAb induction can severely limit the efficacy of systemic re-administration of adenoviral gene therapy. We hypothesized that changing the fiber knob could overcome NAb. Immune-competent mice were exposed to serotype 5 adenovirus (Ad5)(GL), Ad5/3luc1, Ad5lucRGD or Ad5pK7(GL). Mice immunized with Ad5(GL) featured reduced intravenous Ad5(GL) gene transfer to most organs, including the liver, lung and spleen. Ad5(GL) gene transfer was affected much less by exposure to capsid-modified viruses. Anti-Ad5(GL) NAb blocked intravenous Ad5(GL) gene transfer to orthotopic lung cancer xenografts, whereas capsid-modified viruses were not affected. When gene transfer to fresh cancer and normal lung explants was analyzed, we found that capsid-modified viruses allowed effective gene delivery to tumors in the presence of anti-Ad5(GL) NAb, whereas Ad5(GL) was blocked. In contrast, crossblocking by NAbs induced by different viruses affected gene delivery to normal human lung explants, suggesting the importance of non-fiber-knob-mediated infection mechanisms. We conclude that changing the adenovirus fiber knob is sufficient to allow a relative degree of escape from preexisting NAb. If confirmed in trials, this approach might improve the efficacy of re-administration of adenoviral gene therapy to humans.


Subject(s)
Adenoviridae/genetics , Genetic Therapy/methods , Genetic Vectors/genetics , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Animals , Antibodies, Viral/immunology , Antigens, Viral/genetics , Antigens, Viral/immunology , Capsid Proteins/genetics , Carcinoma, Non-Small-Cell Lung/therapy , Genetic Engineering , Genetic Vectors/administration & dosage , Humans , Lung/immunology , Lung/virology , Mice , Mice, Inbred ICR , Mice, Nude , Transduction, Genetic , Xenograft Model Antitumor Assays
3.
Gene Ther ; 14(19): 1380-8, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17611584

ABSTRACT

Arming oncolytic adenoviruses with therapeutic transgenes and enhancing transduction of tumor cells are useful strategies for eradication of advanced tumor masses. Herpes simplex virus thymidine kinase (TK) together with ganciclovir (GCV) has been promising when coupled with viruses featuring low oncolytic potential, but their utility is unknown in the context of highly effective infectivity-enhanced viruses. We constructed Ad5/3-Delta24-TK-GFP, a serotype 3 receptor-targeted, Rb/p16 pathway-selective oncolytic adenovirus, where a fusion gene encoding TK and green fluorescent protein (GFP) was inserted into 6.7K/gp19K-deleted E3 region. Ad5/3-Delta24-TK-GFP killed ovarian cancer cells effectively, which correlated with GFP expression. Delivery of GCV immediately after infection abrogated viral replication, which might have utility as a safety switch. Due to the bystander effect, killing of some cell lines in vitro was enhanced by GCV regardless of timing. In murine models of metastatic ovarian cancer, Ad5/3-Delta24-TK-GFP improved antitumor efficacy over the respective replication-deficient virus with GCV. However, GCV did not further enhance efficacy of Ad5/3-Delta24-TK-GFP in vivo. Simultaneous detection of tumor load and virus replication with bioluminescence and fluorescence imaging provided insight into the in vivo kinetics of oncolysis. In summary, TK/GCV may not add antitumor activity in the context of highly potent oncolysis.


Subject(s)
Genetic Therapy/methods , Oncolytic Virotherapy/methods , Ovarian Neoplasms/therapy , Protein-Tyrosine Kinases/genetics , Simplexvirus/enzymology , Animals , Antiviral Agents/metabolism , Antiviral Agents/therapeutic use , Carcinoma/secondary , Carcinoma/therapy , Cell Line, Tumor , Female , Ganciclovir/metabolism , Ganciclovir/therapeutic use , Gene Expression , Genetic Engineering , Green Fluorescent Proteins/genetics , Humans , Mice , Mice, Nude , Mice, SCID , Models, Animal , Neoplasm Transplantation , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/therapy , Protein-Tyrosine Kinases/metabolism , Virus Replication
4.
Gene Ther ; 14(11): 902-11, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17377596

ABSTRACT

Oncolytic viruses kill cancer cells by tumor-selective replication. Clinical data have established the safety of the approach but also the need of improvements in potency. Efficacy of oncolysis is linked to effective infection of target cells and subsequent productive replication. Other variables include intratumoral barriers, access to target cells, uptake by non-target organs and immune response. Each of these aspects relates to the location and degree of virus replication. Unfortunately, detection of in vivo replication has been difficult, labor intensive and costly and therefore not much studied. We hypothesized that by coinfection of a luciferase expressing E1-deleted virus with an oncolytic virus, both viruses would replicate when present in the same cell. Photon emission due to conversion of D-Luciferin is sensitive and penetrates tissues well. Importantly, killing of animals is not required and each animal can be imaged repeatedly. Two different murine xenograft models were used and intratumoral coinjections of luciferase encoding virus were performed with eight different oncolytic adenoviruses. In both models, we found significant correlation between photon emission and infectious virus production. This suggests that the system can be used for non-invasive quantitation of the amplitude, persistence and dynamics of oncolytic virus replication in vivo, which could be helpful for the development of more effective and safe agents.


Subject(s)
Adenoviridae/genetics , Genetic Therapy/methods , Luciferases/analysis , Microscopy, Fluorescence, Multiphoton , Neoplasms/therapy , Oncolytic Viruses/genetics , Adenoviridae/physiology , Animals , Female , Gene Expression , Genetic Vectors/administration & dosage , Image Processing, Computer-Assisted , Injections, Intraperitoneal , Luciferases/genetics , Mice , Mice, Nude , Models, Animal , Neoplasms/pathology , Oncolytic Virotherapy/methods , Oncolytic Viruses/physiology , Reverse Transcriptase Polymerase Chain Reaction , Transduction, Genetic/methods , Virus Replication
5.
Gene Ther ; 14(1): 58-67, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16900223

ABSTRACT

Conditionally replicating adenoviruses (CRAds) that replicate in tumor but less in normal cells are promising anticancer agents. A major determinant of their potency is their capacity for infecting target cells. The primary receptor for serotype 5 adenovirus (Ad5), the most widely used serotype in gene therapy, is the coxsackie-adenovirus receptor (CAR). CAR is expressed variably and often at low levels in various tumor types including advanced breast cancer. We generated a novel p16/retinoblastoma pathway-dependent CRAd, Ad5.pK7-Delta24, with a polylysine motif in the fiber C-terminus, enabling CAR-independent binding to heparan sulfate proteoglycans (HSPG). Ad5.pK7-Delta24 mediated effective oncolysis of all breast cancer cell lines tested. Further, we utilized noninvasive, fluorescent imaging for analysis of antitumor efficacy in an orthotopic model of advanced hormone refractory breast cancer. A therapeutic benefit was seen following both intratumoral and intravenous delivery. Murine biodistribution similar to Ad5, proven safe in trials, suggests feasibility of clinical safety testing. Interestingly, upregulation of CAR was seen in low-CAR M4A4-LM3 breast cancer cells in vivo, which resulted in better than expected efficacy also with an isogenic CRAd with an unmodified capsid. These results suggest utility of Ad5.pK7-Delta24 and the orthotopic model for further translational studies.


Subject(s)
Breast Neoplasms/therapy , Genetic Therapy/methods , Heparitin Sulfate/metabolism , Oncolytic Virotherapy/methods , Adenoviridae/genetics , Animals , Cell Line, Tumor , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Flow Cytometry , Gene Expression , Gene Targeting , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Heparitin Sulfate/analysis , Humans , Immunohistochemistry , Male , Mice , Mice, Nude , Models, Animal , Neoplasm Transplantation , Protein Binding , Receptors, Virus/genetics , Receptors, Virus/metabolism , Transduction, Genetic/methods , Virus Replication
6.
Gene Ther ; 12(15): 1198-205, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15800658

ABSTRACT

Conditionally replicating adenoviruses (CRAds) represent a novel approach for the treatment of cancers resistant to conventional therapies. The efficacy of CRAds might be further improved by using chemotherapeutic agents in a multimodal antitumor approach. We have evaluated the use of Ad5/3-Delta24, a serotype 3 receptor targeted Rb/p16 pathway selective CRAd, in combination with gemcitabine against human ovarian adenocarcinoma. The combination of these agents showed synergistic cell killing in vitro compared to single treatments. However, the effect was dependent on dose and sequencing of the agents. Our results also indicate that gemcitabine reduces the initial rate of Ad5/3-Delta24 replication without affecting the total amount of virus produced. Possible reasons for synergy between Ad5/3-Delta24 and gemcitabine include the chemosensitizing activity of E1A and/or altered replication kinetics. In an orthotopic murine model of peritoneally disseminated ovarian cancer, the combination increased the survival of mice over either agent alone, and almost 60% of treated mice were cured. Sequencing of the agents was critical for toxicity versus efficacy. Mice remained free from intraperitoneal disease, but some succumbed to treatment-related hepatic or bone marrow toxicity. This suggests that improved efficacy may uncover treatment-related toxicity, which needs to be monitored closely in clinical trials.


Subject(s)
Adenocarcinoma/therapy , Antiviral Agents/therapeutic use , Deoxycytidine/analogs & derivatives , Genetic Therapy/methods , Oncolytic Virotherapy/methods , Ovarian Neoplasms/therapy , Adenocarcinoma/drug therapy , Adenovirus E1A Proteins/genetics , Adenoviruses, Human/genetics , Animals , Antiviral Agents/adverse effects , Bone Marrow Cells/pathology , Bone Marrow Cells/virology , Cell Line, Tumor , Combined Modality Therapy , Deoxycytidine/adverse effects , Deoxycytidine/therapeutic use , Dose-Response Relationship, Drug , Female , Liver/pathology , Liver/virology , Mice , Neoplasms, Experimental , Oncolytic Virotherapy/adverse effects , Oncolytic Viruses , Ovarian Neoplasms/drug therapy , Virus Replication , Gemcitabine
7.
J Med Genet ; 40(10): 741-6, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14569119

ABSTRACT

BACKGROUND: The Schmid type of metaphyseal chondrodysplasia (MCDS) is generally due to mutations in COL10A1 encoding for type X collagen of cartilage. METHODS: We performed a study on the genes coding for the RNA components of RNase MRP (MRPR) and RNase P (H1RNA) among 20 patients with diagnosis of MCDS and no mutations in COL10A1. RESULTS: Two patients were found to be homozygous for a base substitution G for A at nucleotide 70 of RMRP, which is the major mutation causing cartilage-hair hypoplasia. No pathogenic mutations were detected in H1RNA. CONCLUSION: Cartilage-hair hypoplasia diagnosis should be considered in patients with metaphyseal chondrodysplasia even in the absence of any extra-skeletal manifestations if no mutation in COL10A1 can be found and the family history is compatible with autosomal recessive inheritance. Correct diagnosis is important for genetic counselling and for proper follow up of the patients.


Subject(s)
Endoribonucleases/genetics , Mutation , Osteochondrodysplasias/genetics , RNA, Catalytic/genetics , Child, Preschool , Humans , Male , Osteochondrodysplasias/diagnosis , Osteochondrodysplasias/diagnostic imaging , RNA/genetics , Radiography , Ribonuclease P
SELECTION OF CITATIONS
SEARCH DETAIL
...