Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Peter A. Pechan is active.

Publication


Featured researches published by Peter A. Pechan.


Brain Pathology | 1995

Gene Therapy for Brain Tumors

Christof M. Kramm; Miguel Sena-Esteves; Faith H. Barnett; Nikolai G. Rainov; Deborah E. Schuback; John S. Yu; Peter A. Pechan; Werner Paulus; E. Antonio Chiocca; Xandra O. Breakefield

Gene therapy has opened new doors for treatment of neoplastic diseases. This new approach seems very attractive, especially for glioblastomas, since treatment of these brain tumors has failed using conventional therapy regimens. Many different modes of gene therapy for brain tumors have been tested in culture and in vivo. Many of these approaches are based on previously established anti‐neoplastic principles, like prodrug activating enzymes, inhibition of tumor neovascularization, and enhancement of the normally weak anti‐tumor immune response. Delivery of genes to tumor cells has been mediated by a number of viral and synthetic vectors. The most widely used paradigm is based on the activation of ganciclovir to a cytotoxic compound by a viral enzyme, thymidine kinase, which is expressed by tumor cells, after the gene has been introduced by a retroviral vector. This paradigm has proven to be a potent therapy with minimal side effects in several rodent brain tumor models, and has proceeded to phase 1 clinical trials. In this review, current gene therapy strategies and vector systems for treatment of brain tumors will be described and discussed in light of further developments needed to make this new treatment modality clinically efficacious.


Gene Therapy | 1998

Pre-existing herpes simplex virus 1 (HSV-1) immunity decreases, but does not abolish, gene transfer to experimental brain tumors by a HSV-1 vector.

Ulrich Herrlinger; Christof M. Kramm; Aboody-Guterman Ks; Jonathan S. Silver; Keiro Ikeda; Johnston Km; Peter A. Pechan; Barth Rf; Dianne M. Finkelstein; E. A. Chiocca; David N. Louis; Xandra O. Breakefield

The influence of pre-existing anti-herpes simplex type 1 (HSV-1) immunity on HSV-1 vector-mediated gene transfer to glioma cells was analyzed in this gene marking study using intracranial D74 gliomas in syngeneic Fischer rats. The HSV-1 mutant virus used, hrR3, is defective in ribonucleotide reductase and bears the marker genes E. coli lacZ and HSV-1 thymidine kinase (HSVtk). Initial marker gene expression in tumors 12 h after direct virus injection was reduced in immunized animals to about 15% of that in nonimmunized animals. Marker gene expression in both sets stayed at initial levels for 2 days after intratumoral injection and declined markedly on day 5. Inflammatory infiltrates in the tumor were more prominent in HSV-1-immunized, as compared with nonimmunized animals, at 12 and 24 h, but appeared similar at 2–5 days after injection. By day 10, the immune reaction had subsided in immunized animals and macrophages remained only in nonimmunized animals. In conclusion, gene transfer to brain tumors using a HSV-1 vector was greatly reduced, but not competely abolished, under pre-immunization conditions. Pre-existing antibodies to HSV-1 may also serve a positive role in providing an increased margin of safety in intracranial application of HSV-1 vectors by limiting spread of the virus within the brain and to other tissues.


Neuroreport | 1995

Genetically modified fibroblasts producing NGF protect hippocampal neurons after ischemia in the rat.

Peter A. Pechan; Tazuka Yoshida; Nariman Panahian; Michael A. Moskowitz; Xandra O. Breakefield

The neuroprotective effect of nerve growth factor (NGF) on the pyramidal cells in the vulnerable CA1-CA2 sectors of the hippocampus was investigated in a rat model of transient forebrain ischemia. A genetically modified fibroblast line that secretes high levels of NGF was implanted 7 days before induction of ischemia between the hippocampal CA1-CA2 subfields in the right hemisphere. Rats were then subjected to 10 min of cerebral ischemia in a four vessel occlusion model. Morphological changes in the CA1 and CA2 subfields were evaluated 7 days after ischemia. Animals in the NGF-protected group had significantly higher numbers of normal appearing neurons in the right CA1 and CA2 regions, compared with their non-implanted left hemispheres, to non-implanted animals or to animals implanted with non-modified cells. The data confirmed that NGF can protect CA1-CA2 hippocampal neurons from ischemic damage by implantation of genetically engineered cells producing NGF.


Neuroreport | 1997

Green fluorescent protein as a reporter for retrovirus and helper virus-free HSV-1 amplicon vector-mediated gene transfer into neural cells in culture and in vivo

Aboody-Guterman Ks; Peter A. Pechan; Nikolai G. Rainov; Miguel Sena-Esteves; Andreas Jacobs; Evan Y. Snyder; Peter Wild; Elisabeth M. Schraner; Kurt Tobler; Xandra O. Breakefield; Cornel Fraefel

GREEN fluorescent protein (GFP) is an effective marker for retrovirus and herpes virus vector-mediated gene transfer into various central nervous system-derived cells, both proliferative and non-proliferative, in culture and in vivo. Retrovirus vectors were used to stably transduce several rat and human glioma lines, and a multi-potent mouse neural progenitor line in culture. Implantation of selected pools of transduced glioma cells into rodent brain allowed clear visualization of the tumor and the invading tumor edge. Helper virus-free HSV-1 amplicon vectors successfully transferred gfp into non-dividing primary neural cells in culture and in the rat brain. This study describes the versatility of GFP for: (i) labelling of glioma cells in experimental brain tumor models and neural progenitor cells by retrovirus vectors, and (ii) efficient, non-toxic delivery of genes to post mitotic cells of the nervous system using helper-virus free HSV-1 amplicon vectors.


Human Gene Therapy | 1999

Intraarterial Delivery of Adenovirus Vectors and Liposome± DNA Complexes to Experimental Brain Neoplasms

Nikolai G. Rainov; Keiro Ikeda; Nazir H. Qureshi; Shivani Grover; Ulrich Herrlinger; Peter A. Pechan; E. Antonio Chiocca; Xandra O. Breakefield; Faith H. Barnett

This study investigated the intraarterial delivery of genetically engineered replication-deficient adenovirus vectors (AVs) and cationic liposome-plasmid DNA complexes (lipoDNA) to experimental brain tumors. Adenovirus or lipoDNA was injected into the internal carotid artery (ICA) of F344 rats harboring intracerebral 9L gliosarcomas, using bradykinin (BK) to selectively permeabilize the blood-tumor barrier (BTB). Brain and internal organs of the animals were collected 48 hr after vector injection and stained for expression of the marker gene product, beta-galactosidase (beta-Gal). Intracarotid delivery of AV to 9L rat gliosarcoma without BTB disruption resulted in transgene expression in 3-10% of tumor cells distributed throughout the tumor. Virus-mediated expression of beta-gal gene products in this tumor model was particularly high in small foci (< or = 0.5 mm), which had invaded the normal brain tissue surrounding the main tumor mass. In these foci more than 50% of tumor cells were transduced. BK infusion increased the amount of transgene-expressing cells in larger tumor foci to 15-30%. In the brain parenchyma only a few endothelial cells expressed beta-gal owing to AV-mediated gene transfer. Intracarotid delivery of lipoDNA bearing a cytoplasmic expression cassette rendered more than 30% of the tumor cells positive for the marker gene without BTB disruption. The pattern of distribution was in general homogeneous throughout the tumor. BK infusion was able to increase further the number of transduced tumor cells to more than 50%. Although lipoDNA-mediated gene transfer showed increased efficacy as compared with AV-mediated gene transfer, it had less specificity since a larger number of endothelial and glial cells also expressed the transgene. AV and lipoDNA injections, in the absence and presence of BK, also resulted in transduction of peripheral organs. AV showed its known predilection for liver and lung. In the case of lipoDNA, parenchymal organs such as liver, lung, testes, lymphatic nodes, and especially spleen, were transduced. These findings indicate that intracarotid application of AV and lipoDNA vectors can effectively transduce tumor cells in the brain, and that BTB modulation by BK infusion can further increase the number of transgene-expressing tumor cells.


Neuroreport | 1992

bFGF induces its own gene expression in astrocytic and hippocampal cell cultures

Berenike Flott-Rahmel; W. Gerdes; Peter A. Pechan; Wolfgang Brysch; Karl-Hermann Schlingensiepen; Wilfried Seifert

Basic FGF mRNA induction by bFGF was investigated in cell cultures from rat brain, i.e. postnatal day 2 cortex and embryonic day 18 hippocampus. In situ hybridization shows that after bFGF treatment (10(-10) M) for 14 h neurones and glial cells show a remarkable increase in bFGF mRNA production. Incubation of astrocytes with antisense bFGF phosphorothioate oligodeoxynucleotides (bFGF-PTOs) resulted in an inhibition of both bFGF induced and serum induced proliferation. The results indicate that bFGF is capable of inducing its own mRNA production. This induction, i.e. new synthesis of bFGF mRNA, seems to be essential for the mitogenic effect of both bFGF and serum components.


Journal of NeuroVirology | 1998

Targeting gene therapy vectors to CNS malignancies

Matthew A. Spear; Ulrich Herrlinger; Nikolai G. Rainov; Peter A. Pechan; Ralph Weissleder; Xandra O. Breakefield

Gene therapy offers significant advantages to the field of oncology with the addition of specifically and uniquely engineered mechanisms of halting malignant proliferation through cytotoxicity or reproductive arrest. To confer a true benefit to the therapeutic ratio (the relative toxicity to tumor compared to normal tissue) a vector or the transgene it carries must selectively affect or access tumor cells. Beyond the selective toxicities of many transgene products, which frequently parallel that of contemporary chemotherapeutic agents, lies the potential utility of targeting the vector. This review presents an overview of current and potential methods for designing vectors targeted to CNS malignancies through selective delivery, cell entry, transport or transcriptional regulation. The topic of delivery encompasses physical and pharmaceutic means of increasing the relative exposure of tumors to vector. Cell entry based methodologies are founded on increasing relative uptake of vector through the chemical or recombinant addition of ligand and antibody domains which selectively bind receptors expressed on target cells. Targeted transport involves the potential for using cells to selectively carry vectors or transgenes into tumors. Finally, promoter and enhancer systems are discussed which have potential for selectivity activating transcription to produce targeted transgene expression or vector propagation.


Journal of Gene Medicine | 2000

HSV‐1 infected cell proteins influence tetracycline‐regulated transgene expression

Ulrich Herrlinger; Peter A. Pechan; Andreas Jacobs; Christian Woiciechowski; Nikolai G. Rainov; Cornel Fraefel; Werner Paulus; Steven A. Reeves

This study investigates elements of herpes simplex virus type 1 (HSV‐1) which influence transgene expression in tetracycline‐regulated expression systems.


Journal of Gene Medicine | 1999

Combined HSV-1 recombinant and amplicon piggyback vectors: replication-competent and defective forms, and therapeutic efficacy for experimental gliomas.

Peter A. Pechan; Ulrich Herrlinger; Manish K. Aghi; Andreas Jacobs; Xandra O. Breakefield

The versatility of HSV‐1 vectors includes large transgene capacity, selective replication of mutants in dividing cells, and availability of recombinant virus (RV) and plasmid‐derived (amplicon) vectors, which can be propagated in a co‐dependent, ‘piggyback’, manner.


Genetic Manipulation of the Nervous System | 1995

Gene delivery to the nervous system using retroviral vectors

Miguel Sena-Esteves; Manish K. Aghi; Peter A. Pechan; Edward M. Kaye; Xandra O. Breakefield

Publisher Summary Retrovirus vectors derived from Moloney murine leukemia virus (MMLV) have been the most utilized vector for gene delivery to the nervous system. Particles enter the cells by binding to glycoproteins on the plasma membrane of the host cell, followed by fusion of the virus envelope with the cell membrane. There is some cell specificity of virus entry conferred by the interaction between envelope elements in the virus particles and the cell membrane. It is shown that stable, albeit low level, transgene expression could be achieved following peripheral grafting of fibroblasts by using the “house-keeping” promoter for dihydrofolate reductase, which is active in most mammalian cell types. The function of particular neural proteins is elucidated by retrovirus-encoded antisense RNA, which interferes with expression of the protein. Antisense technology is still an incomplete art and one can expect to achieve only partial inhibition of translation of the targeted mRNA. Retrovirus-mediated gene transfer is a particularly applicable technique for brain tumors because of the selectivity of retroviruses for proliferating cells. The possibility of obtaining retroviruses with mixed phenotypes, through infection of the same cell by different viruses, from the same or different taxonomic groups is also elaborated.

Collaboration


Dive into the Peter A. Pechan's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Miguel Sena-Esteves

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

E. Antonio Chiocca

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Maureen Chase

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Manish K. Aghi

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge