24 results on '"Richard Harbottle"'
Search Results
2. Delivery of non-viral naked DNA vectors to liver in small weaned pigs by hydrodynamic retrograde intrabiliary injection
- Author
-
Tatjana Chan, Hiu Man Grisch-Chan, Philipp Schmierer, Ulrike Subotic, Nicole Rimann, Tanja Scherer, Udo Hetzel, Matthias Bozza, Richard Harbottle, James A. Williams, Barbara Steblaj, Simone K. Ringer, Johannes Häberle, Xaver Sidler, and Beat Thöny
- Subjects
pig ,liver gene therapy ,hydrodynamic intrabiliary injection ,non-viral ,DNA-vector ,bile duct ,Genetics ,QH426-470 ,Cytology ,QH573-671 - Abstract
Hepatic gene therapy by delivering non-integrating therapeutic vectors in newborns remains challenging due to the risk of dilution and loss of efficacy in the growing liver. Previously we reported on hepatocyte transfection in piglets by intraportal injection of naked DNA vectors. Here, we established delivery of naked DNA vectors to target periportal hepatocytes in weaned pigs by hydrodynamic retrograde intrabiliary injection (HRII). The surgical procedure involved laparotomy and transient isolation of the liver. For vector delivery, a catheter was placed within the common bile duct by enterotomy. Under optimal conditions, no histological abnormalities were observed in liver tissue upon pressurized injections. The transfection of hepatocytes in all tested liver samples was observed with vectors expressing luciferase from a liver-specific promoter. However, vector copy number and luciferase expression were low compared to hydrodynamic intraportal injection. A 10-fold higher number of vector genomes and luciferase expression was observed in pigs using a non-integrating naked DNA vector with the potential for replication. In summary, the HRII application was less efficient (i.e., lower luciferase activity and vector copy numbers) than the intraportal delivery method but was significantly less distressful for the piglets and has the potential for injection (or re-injection) of vector DNA by endoscopic retrograde cholangiopancreatography.
- Published
- 2022
- Full Text
- View/download PDF
3. Beta HPV38 oncoproteins act with a hit-and-run mechanism in ultraviolet radiation-induced skin carcinogenesis in mice.
- Author
-
Daniele Viarisio, Karin Müller-Decker, Rosita Accardi, Alexis Robitaille, Matthias Dürst, Katrin Beer, Lars Jansen, Christa Flechtenmacher, Matthias Bozza, Richard Harbottle, Catherine Voegele, Maude Ardin, Jiri Zavadil, Sandra Caldeira, Lutz Gissmann, and Massimo Tommasino
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Cutaneous beta human papillomavirus (HPV) types are suspected to be involved, together with ultraviolet (UV) radiation, in the development of non-melanoma skin cancer (NMSC). Studies in in vitro and in vivo experimental models have highlighted the transforming properties of beta HPV E6 and E7 oncoproteins. However, epidemiological findings indicate that beta HPV types may be required only at an initial stage of carcinogenesis, and may become dispensable after full establishment of NMSC. Here, we further investigate the potential role of beta HPVs in NMSC using a Cre-loxP-based transgenic (Tg) mouse model that expresses beta HPV38 E6 and E7 oncogenes in the basal layer of the skin epidermis and is highly susceptible to UV-induced carcinogenesis. Using whole-exome sequencing, we show that, in contrast to WT animals, when exposed to chronic UV irradiation K14 HPV38 E6/E7 Tg mice accumulate a large number of UV-induced DNA mutations, which increase proportionally with the severity of the skin lesions. The mutation pattern detected in the Tg skin lesions closely resembles that detected in human NMSC, with the highest mutation rate in p53 and Notch genes. Using the Cre-lox recombination system, we observed that deletion of the viral oncogenes after development of UV-induced skin lesions did not affect the tumour growth. Together, these findings support the concept that beta HPV types act only at an initial stage of carcinogenesis, by potentiating the deleterious effects of UV radiation.
- Published
- 2018
- Full Text
- View/download PDF
4. High-throughput Pyrosequencing™ of a phage display library for the identification of enriched target-specific peptides
- Author
-
Ahad Rahim, Charles Coutelle, and Richard Harbottle
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Gene therapy clinical trials have highlighted the importance of specific cellular/tissue targeting of gene delivery vectors. Phage display libraries are powerful tools for the selection of novel peptide ligands as targeting moieties because of their high-throughput screening potential. However, a severe rate-limiting step in this procedure in terms of time, numbers, and cost is the sequence identification of selected phages. Here we describe the application of PyrosequencingTM technology for sequencing phage isolates after panning a random 7-mer peptide expressing phage library against the A549 bronchial epithelial cell line to search for enrichment of possible targeting peptides. Pyrosequencing allows sequencing of 96 phages at one time in approximately 45 min at only a sixth of the cost of conventional sequencing methods. Using this technology, we have identified four sequences of interest. A phage binding assay revealed that three of the four sequences show a significant increase in binding abilities and specificity for A549 cells when compared to an unrelated cell line.
- Published
- 2003
- Full Text
- View/download PDF
5. Perivascular tenascin C triggers sequential activation of macrophages and endothelial cells to generate a pro-metastatic vascular niche in the lungs
- Author
-
Tsunaki Hongu, Maren Pein, Jacob Insua-Rodríguez, Ewgenija Gutjahr, Greta Mattavelli, Jasmin Meier, Kristin Decker, Arnaud Descot, Matthias Bozza, Richard Harbottle, Andreas Trumpp, Hans-Peter Sinn, Angela Riedel, and Thordur Oskarsson
- Subjects
Vascular Endothelial Growth Factor A ,Mice ,Cancer Research ,Lung Neoplasms ,Neovascularization, Pathologic ,Oncology ,Macrophages ,Animals ,Endothelial Cells ,Tenascin ,Lung - Abstract
Disseminated cancer cells frequently lodge near vasculature in secondary organs. However, our understanding of the cellular crosstalk invoked at perivascular sites is still rudimentary. Here, we identify intercellular machinery governing formation of a pro-metastatic vascular niche during breast cancer colonization in the lung. We show that specific secreted factors, induced in metastasis-associated endothelial cells (ECs), promote metastasis in mice by enhancing stem cell properties and the viability of cancer cells. Perivascular macrophages, activated via tenascin C (TNC) stimulation of Toll-like receptor 4 (TLR4), were shown to be crucial in niche activation by secreting nitric oxide (NO) and tumor necrosis factor (TNF) to induce EC-mediated production of niche components. Notably, this mechanism was independent of vascular endothelial growth factor (VEGF), a key regulator of EC behavior and angiogenesis. However, targeting both macrophage-mediated vascular niche activation and VEGF-regulated angiogenesis resulted in added potency to curb lung metastasis in mice. Together, our findings provide mechanistic insights into the formation of vascular niches in metastasis.
- Published
- 2022
- Full Text
- View/download PDF
6. Next-generation DNA vectors: is the nS/MARt platform a viable alternative to viruses for autologous T-cell immunotherapy?
- Author
-
Matthias Bozza and Richard Harbottle
- Subjects
General Economics, Econometrics and Finance - Published
- 2021
- Full Text
- View/download PDF
7. Abstract 1769: A combination approach of a cellular library and single cell microfluidics analysis for the rapid selection of CAR-T cells
- Author
-
Eren Boga, Luisa Berger, Alice De Roia, Inka Zörnig, Robert Embacher, Stefan B. Eichmüller, Dirk Jäger, Richard Harbottle, and Patrick Schmidt
- Subjects
Cancer Research ,Oncology - Abstract
Within the last decade CAR-T cells have changed the landscape of treatment regimen for leukemia and myeloma, which is reflected by the first FDA approval of this class of living drugs in 2017. As of today, the clinical trial situation aims on CAR-T applicability in solid tumors, which is more arduous due to antigen heterogeneity and limited CAR-T persistence. More personalized and multi-target oriented cellular products may offer a solution to overcome these problems but then CAR identification and selection display the major bottleneck in the drug development process. Usually, matching scFvs are selected from targeting screens of a phage-display library and hits are subsequently cloned in CAR backbones and tested for functionality and possible limiting factors as the occurrence of tonic signaling. This makes the whole process very time consuming and laborious. Within this project, we propose a novel CAR-T selection method that rapidly shortens the discovery procedure. We have developed a full length CAR library in nS/MARt DNA vectors that is electroporated in a Jurkat reporter cell line reflecting entirely its full diversity. By this, we can quickly identify the amount of tonic signaling CARs and exclude them from further selection. For on-target selection we first perform a bulk pre-selection followed by a single cell functionality screening using the Berkeley Lights Lightning™ device. This allows us to export hits as clonal viable cells that undergo long length Nanopore CAR-RNA sequencing. Our findings show the feasibility of our approach and that it can shorten the timeframe needed for the full selection process from weeks to days. Citation Format: Eren Boga, Luisa Berger, Alice De Roia, Inka Zörnig, Robert Embacher, Stefan B. Eichmüller, Dirk Jäger, Richard Harbottle, Patrick Schmidt. A combination approach of a cellular library and single cell microfluidics analysis for the rapid selection of CAR-T cells [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 1769.
- Published
- 2023
- Full Text
- View/download PDF
8. A nonviral, nonintegrating DNA nanovector platform for the safe, rapid, and persistent manufacture of recombinant T cells
- Author
-
Dirk Jäger, Inka Zörnig, Aileen Berger, Patrick Schmidt, Richard Harbottle, Alexandra Tuch, Matthias Bozza, Andreas Schmidt, Alice De Roia, and Margareta P. Correia
- Subjects
Computer science ,Transgene ,T-Lymphocytes ,Genetic Vectors ,Immunotherapy, Adoptive ,law.invention ,Cell therapy ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,law ,Neoplasms ,Humans ,Vector (molecular biology) ,Transgenes ,Molecular Biology ,Research Articles ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,SciAdv r-articles ,DNA ,In vitro ,Chimeric antigen receptor ,Genetically modified organism ,Cell biology ,chemistry ,Applied Sciences and Engineering ,030220 oncology & carcinogenesis ,Recombinant DNA ,human activities ,Research Article - Abstract
Autosomally replicating episomal DNA nanovectors allow clinical-scale CAR-T cell manufacturing., The compelling need to provide adoptive cell therapy (ACT) to an increasing number of oncology patients within a meaningful therapeutic window makes the development of an efficient, fast, versatile, and safe genetic tool for creating recombinant T cells indispensable. In this study, we used nonintegrating minimally sized DNA vectors with an enhanced capability of generating genetically modified cells, and we demonstrate that they can be efficiently used to engineer human T lymphocytes. This vector platform contains no viral components and is capable of replicating extrachromosomally in the nucleus of dividing cells, providing persistent transgene expression in human T cells without affecting their behavior and molecular integrity. We use this technology to provide a manufacturing protocol to quickly generate chimeric antigen receptor (CAR)–T cells at clinical scale in a closed system and demonstrate their enhanced anti-tumor activity in vitro and in vivo in comparison to previously described integrating vectors.
- Published
- 2021
9. Novel Non-integrating DNA Nano-S/MAR Vectors Restore Gene Function in Isogenic Patient-Derived Pancreatic Tumor Models
- Author
-
Williams James A, Martin R. Sprick, Alice De Roia, Vanessa Vogel, Richard Harbottle, Rienk Offringa, Edward W. Green, Corinna Klein, Matthias Bozza, and Elisa Espinet
- Subjects
0301 basic medicine ,Tumor suppressor gene ,lcsh:QH426-470 ,Transgene ,Cell ,Biology ,antibiotic-free ,isogenic cells ,Article ,S/MAR ,Transcriptome ,Insertional mutagenesis ,non-integrating ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Genetics ,medicine ,lcsh:QH573-671 ,Molecular Biology ,Mitosis ,Gene ,lcsh:Cytology ,tumor models ,gene supplementation ,nano-DNA vector ,Cell biology ,lcsh:Genetics ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,030220 oncology & carcinogenesis ,Molecular Medicine ,DNA - Abstract
We describe herein non-integrating minimally sized nano-S/MAR DNA vectors, which can be used to genetically modify dividing cells in place of integrating vectors. They represent a unique genetic tool, which avoids vector-mediated damage. Previous work has shown that DNA vectors comprising a mammalian S/MAR element can provide persistent mitotic stability over hundreds of cell divisions, resisting epigenetic silencing and thereby allowing sustained transgene expression. The composition of the original S/MAR vectors does present some inherent limitations that can provoke cellular toxicity. Herein, we present a new system, the nano-S/MAR, which drives higher transgene expression and has improved efficiency of establishment, due to the minimal impact on cellular processes and perturbation of the endogenous transcriptome. We show that these features enable the hitherto challenging genetic modification of patient-derived cells to stably restore the tumor suppressor gene SMAD4 to a patient-derived SMAD4 knockout pancreatic cancer line. Nano-S/MAR modification does not alter the molecular or phenotypic integrity of the patient-derived cells in cell culture and xenograft mouse models. In conclusion, we show that these DNA vectors can be used to persistently modify a range of cells, providing sustained transgene expression while avoiding the risks of insertional mutagenesis and other vector-mediated toxicity., Graphical Abstract, Bozza and et al. describe minimally sized DNA vectors that can be used to generate genetically modified isogenic human cells that produce persistent transgene expression while avoiding insertional mutagenesis and vector-mediated toxicity. They show the restoration of the tumor suppressor gene SMAD4 into patient-derived pancreatic cells reversing the cellular and molecular phenotype.
- Published
- 2020
10. Suppression of antitumor T cell immunity by the oncometabolite (R)-2-hydroxyglutarate
- Author
-
Iris Oezen, Michael Kilian, Michael O. Breckwoldt, Mario L. Suvà, Michael C. Burger, Mirco Friedrich, Khwab Sanghvi, Karl H. Plate, Magdalena Kramer, Sevin Turcan, Daniel Hänggi, Niklas Thon, Daniel Schrimpf, Stefan Kaulfuss, Katrin Deumelandt, Jochen Meyer, Richard Harbottle, Jürgen G. Okun, Theresa Bunse, Edward W. Green, Tobias Kessler, Miriam Ratliff, Michael Platten, Jana K. Sonner, Brandon Nicolay, Marion Dorsch, Michael Weller, Anna von Landenberg, Ruslan Al-Ali, Mya Steadman, Matthias Bozza, Holger Hess-Stumpp, Karen Bieback, Dongwei Zhu, Stefan Pusch, Cyril Neftel, Wolfgang Wick, Jessica Eisel, Benedikt Wiestler, Antje Habel, Minou Nadji-Ohl, Axel Benner, Christel Herold-Mende, Anna S. Berghoff, Dalia Alansary, Patrick N. Harter, Lukas Bunse, Kelly Marsh, Gernot Poschet, Barbara A. Niemeyer, Andreas von Deimling, Daniel P. Cahill, Felix Sahm, Simone Karcher-Bausch, Matthias Preusser, and Stefanie Uhlig
- Subjects
0301 basic medicine ,T-Lymphocytes ,T cell ,Receptors, Antigen, T-Cell ,Apoptosis ,Lymphocyte Activation ,General Biochemistry, Genetics and Molecular Biology ,Glutarates ,03 medical and health sciences ,Adenosine Triphosphate ,Antigen ,Cell Line, Tumor ,Paracrine Communication ,Histone methylation ,Polyamines ,medicine ,Animals ,Humans ,RNA, Messenger ,Cell Proliferation ,NFATC Transcription Factors ,Brain Neoplasms ,Chemistry ,Cell growth ,Immunity ,Glioma ,General Medicine ,Isocitrate Dehydrogenase ,Mice, Inbred C57BL ,030104 developmental biology ,Isocitrate dehydrogenase ,medicine.anatomical_structure ,Cell culture ,Mutation ,Cancer research ,Calcium ,Signal transduction ,Signal Transduction - Abstract
The oncometabolite (R)-2-hydroxyglutarate (R-2-HG) produced by isocitrate dehydrogenase (IDH) mutations promotes gliomagenesis via DNA and histone methylation. Here, we identify an additional activity of R-2-HG: tumor cell-derived R-2-HG is taken up by T cells where it induces a perturbation of nuclear factor of activated T cells transcriptional activity and polyamine biosynthesis, resulting in suppression of T cell activity. IDH1-mutant gliomas display reduced T cell abundance and altered calcium signaling. Antitumor immunity to experimental syngeneic IDH1-mutant tumors induced by IDH1-specific vaccine or checkpoint inhibition is improved by inhibition of the neomorphic enzymatic function of mutant IDH1. These data attribute a novel, non-tumor cell-autonomous role to an oncometabolite in shaping the tumor immune microenvironment.
- Published
- 2018
- Full Text
- View/download PDF
11. The expression of genes contributing to pancreatic adenocarcinoma progression is influenced by the respective environment
- Author
-
Martin R. Berger, Michael Zepp, Matthias Bozza, Frank Bergmann, Micah N Sagini, and Richard Harbottle
- Subjects
0301 basic medicine ,Cancer Research ,Biology ,tumor model ,03 medical and health sciences ,0302 clinical medicine ,Gene expression ,Genetics ,medicine ,Gene knockdown ,Cell growth ,gene expression profiles ,tumor micro-environment ,PDAC ,Cell migration ,medicine.disease ,Fold change ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer research ,Adenocarcinoma ,DNA fragmentation ,TGM2 ,Pancreas ,Research Paper - Abstract
Pancreatic adenocarcinoma is a highly aggressive malignancy with dismal prognosis and limited curative options. We investigated the influence of organ environments on gene expression in RNU rats by orthotopic and intraportal infusion of Suit2-007luc cells into the pancreas, liver and lung respectively. Tumor tissues from these sites were analyzed by chip array and histopathology. Generated data was analyzed by Chipster and Ingenuity Pathway Analysis (±1.5 expression fold change and p
- Published
- 2018
12. Maxcyte flow electroporation technology: a safe, reliable and effective method for engineering car T cells
- Author
-
Patrick Schmidt, A. Roig-Merino, P. Heine, Alexandra Tuch, J. Brady, Richard Harbottle, Matthias Bozza, and A. de Roia
- Subjects
Cancer Research ,Transplantation ,Materials science ,Oncology ,Flow (mathematics) ,Electroporation ,Immunology ,Immunology and Allergy ,Effective method ,Cell Biology ,Car t cells ,Genetics (clinical) ,Biomedical engineering - Published
- 2021
- Full Text
- View/download PDF
13. IMMU-41. HIGH-THROUGHPUT RETRIEVAL OF THERAPEUTIC T CELL RECEPTORS FROM GLIOMA
- Author
-
Richard Harbottle, Andreas von Deimling, Katharina A.M. Lindner, Matthias Bozza, Felix Sahm, Lukas Bunse, Khwab Sanghvi, Edward W. Green, Chin Leng Tan, Isabel Poschke, Wolfgang Wick, and Michael Platten
- Subjects
Cancer Research ,Oncology ,Chemistry ,Glioma ,Immunology ,T-cell receptor ,Cancer research ,medicine ,Neurology (clinical) ,medicine.disease ,Throughput (business) - Abstract
Gliomas are tumors with low mutational burden with the majority of them being resistant to checkpoint inhibition due to few immunogenic antigens. Multicenter vaccine trials targeting personalized neoantigens in gliomas demonstrated feasibility and illustrated the challenges of retrieving neoepitope-specific T cells based on the prediction of immunogenic neoepitopes. Here we took an entirely different T cell-centric approach and established a single cell sequencing-based high-throughput T cell receptor (TCR) retrieval platform, exploiting the therapeutic potential of spontaneous intratumoral T cell clonotypes for the development of adoptive cell therapy. We conducted direct ex vivo TCR single cell sequencing from freshly sorted human glioma-infiltrating T cell samples. High fidelity PCR was established to clone TCRs from single cell libraries directly into episomal expression vectors further optimized for T cell therapy. In parallel to standard therapy, patient-derived xenografts were developed and characterized. Tumor-reactivity of retrieved TCRs was demonstrated against patient-derived cell lines. Collectively, we provide a novel sequencing-based platform for high-throughput identification and validation of endogenous glioma-targeting TCRs and demonstrate their therapeutic applicability.
- Published
- 2020
- Full Text
- View/download PDF
14. Abstract 4717: Ly6-neurotoxin1 knockout in PDAC cells inhibits their growth in vitro and in vivo
- Author
-
Doaa M. Ali, Michael Zepp, Martin R. Berger, Maria Nikolova, Matthias Bozza, and Richard Harbottle
- Subjects
Cancer Research ,Gene knockdown ,Cell ,Clone (cell biology) ,Transfection ,Biology ,Molecular biology ,In vitro ,medicine.anatomical_structure ,Oncology ,In vivo ,Cell culture ,medicine ,MTT assay - Abstract
Ly6/neurotoxin1 (Lynx1) functions as a brake for nicotinic receptors and was defined as a tumor suppressor in lung cancer. As pancreatic cancer development may be slowed down by cholinergic signaling, we investigated the role of Lynx1 in pancreatic ductal adenocarcinoma (PDAC) cell lines, both in vitro and in vivo. Lynx1 knockout cell clones were generated by transfecting the CRISPRCas9 plasmid - (pSpCas9 (BB)-2A-Puro) into Miapacaluci and BXPC3luci PDAC cells using jet Pei and jet Prime as transfecting agents, respectively. The annealed gRNA was directed towards exon 3 of the Lynx1 gene. Control clones were transfected with the plasmid alone. Clones from both cell lines were tested by qRT-PCR and Western blot for knockout efficiency as well as by proliferation assay. In addition, the expression levels of p-MEK, p-MAPK, p-mTOR and p-Rictor were verified in two BXPC3 cell clones in relation to Lynx1 expression. Furthermore, 4x106 cells of four BXPC3 cell clones (one control, 3 knockout clones) were injected, respectively, into the portal vein of nude rats to control for a possibly reduced tumor growth in the liver. The knockdown of Lynx1 was incomplete at mRNA level and ranged from 20 to 60% in BXPC3 clones and from 0 to 30% in Miapaca clones. At protein level, the respective values ranged from 0 to 90% in BXPC3 and from 22 to 50% in Miapaca clones. However, Lynx1 protein levels increased at later time points. All BXPC3 clones proliferated less quickly than the respective control when tested by MTT assay. Miapaca clones, however, didn't show a significant difference from the respective control, although their growth was clearly disturbed shortly after transfection. Concomitantly with reduced Lynx1 protein levels, there was reduction of p-mTOR (90%), p-Rictor (30%) and p-MEK (40%) in BXPC3 cells. In vivo, the BXPC3 clones showed a lag period of 1 to 2 weeks till the appearance of a first bioluminescence signal indicating tumor growth. Rats injected with cells from the control clone showed a steady increase in the bioluminescence signal (n=7 of 10) as compared to the most sensitive knockdown clone, which didn't show any signal in any of 4 injected rats (p=0.05). Cells of 2 other BXPC3 clones showed a reduced growth rate at best in 2 of 2 rats used for each clone, respectively. In conclusion, knockout of Lynx1 was incomplete at both mRNA and protein levels. Nevertheless, the respective BXPC3 clones exhibited reduced proliferation in vitro, which was associated with diminished p-mTOR, p-Rictor and p-MEK levels. In addition, they failed to establish a tumor in vivo or showed a reduced tumor growth rate. These findings suggest that Lynx1 is a vital gene and may play an important role in the growth and establishment of PDAC cells. Citation Format: Doaa Ali, Michael Zepp, Matthias Bozza, Maria Nikolova, Richard Harbottle, Martin R. Berger. Ly6-neurotoxin1 knockout in PDAC cells inhibits their growth in vitro and in vivo [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 4717.
- Published
- 2020
- Full Text
- View/download PDF
15. Abstract 4066: A non-integrating, non-viral DNA Nanovector platform for the safe, persistent, and rapid manufacture of recombinant T-cells for Adoptive cell therapy
- Author
-
Alexandra Tuch, Alice De Roia, Aileen Berger, Matthias Bozza, Richard Harbottle, and Patrick Schmidt
- Subjects
Cancer Research ,Electroporation ,Transfection ,Biology ,Chimeric antigen receptor ,Viral vector ,law.invention ,Cell therapy ,Biopharmaceutical ,Oncology ,law ,Cancer research ,Recombinant DNA ,Vector (molecular biology) - Abstract
The capability to introduce Chimeric Antigen Receptors (CARs) into naïve Human T-Cells represents one of the most promising therapeutic strategies for the treatment of cancer. However, virus mediated adoptive cell therapy (ACT) remain severely limited by two factors: the long lead time and high cost of GMP virus manufacture, and the virus safety profiles. What if the entire ACT process could be sped up, made safer and more cost-effective by at least an order of magnitude? We have invented a novel DNA Vector platform based on scaffold/matrix attachment region (S/MAR) component that provides the opportunity to efficiently generate genetically engineered T-cells. This system is based on a nanovector technology. It contains no immunogenic and comprises only clinically approved sequences. It is easy, simple and cost-efficient to produce. Critically, it does not integrate and replicates autonomously and extrachromosomally in the nuclei of dividing primary human cells, thus avoiding the inherent risk of integrative mutagenesis. Through a process of iterative CpG depletion, selection marker minimalisation, empirical promoter design and elimination of cryptic eukaryotic signals our nano-S/MARt DNA Vector (nS/MARt) can be efficiently transfected into primary human T Cells. nS/MARt vectors are designed to remain stably expressed, and in addition to having the best in class safety profile, they also demonstrate enhanced performance as a biopharmaceutical. Human T-cells engineered to express the CAR receptor against the carcinoembryonic antigen (CEA) using a nS/MARt vector provide more effective killing of human cancer cells in vitro than those engineered with integrative lentivirus. These results hold in vivo, where nS/MARt transfected CAR Tcells outperform the lentivirally transduced cells, attenuating tumour growth and extending mouse survival. Moreover, in pre-clinical studies, the comparison with the FDA approved drug Kymriah®,T cells modified with nS/MARt vectors harbouring the expression of a CD19 CAR are comparable to those engineered with the viral vector. Notably, we have also taken steps to evaluate nS/MARt's scalability and have succeeded in manufacturing a clinically relevant number of CAR-T Cells (2 × 107CAR+ T-cells per kilo, we estimate the production for an individual of 80 Kg). The extension of the results from mice to patients-scale required a 1000x scale up for the processing of T-cell transfection while halving the time for production to hit a meaningful therapeutic window. We have developed a novel manufacturing protocol where nS/MARt vectors can be used "off the shelf" for CAR-T therapy to generate a clinically relevant number of modified cells in just seven days. The delivery of our DNA to CD3+ cells, reaches ~60-70% with cell viability of 60%, that increases in the days that follow the cell electroporation. Thus, the most significant benefit will be for the patients that will be able to access the nS/MARt mediated therapy in 1 week. To translate this technology into a clinical reality a fermentation process that allows the preparation of 2.6 g/L of pure, supercoiled DNA was optimised. There is a pressing need to offer ACT to more oncology patients, and we believe that this novel DNA Vector system provides a unique and innovative approach to this therapeutic strategy for cancer therapy. Citation Format: Matthias Bozza, Alice De Roia, Aileen Berger, Alexandra Tuch, Patrick Schmidt, Richard Harbottle. A non-integrating, non-viral DNA Nanovector platform for the safe, persistent, and rapid manufacture of recombinant T-cells for Adoptive cell therapy [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 4066.
- Published
- 2020
- Full Text
- View/download PDF
16. 3004 – IDENTIFICATION AND CHARACTERIZATION OF NOVEL FUNCTIONAL MARKERS DURING THE HEMATOPOIETIC STEM CELL SPECIFICATION PROCESS
- Author
-
David Hills, Irem Bayindir-Buchhalter, Marleen Büchler-Schäff, Wenjun Chang, Michael D. Milsom, Stella Pfaffenholz, Michèle Souyri, Julius Gräsel, Georges Lacaud, Sabrina Hanke, Roshana Thambyrajah, Wiebke Nadler, Alexander Medvinsky, Richard Harbottle, Milena Block, Paul Kaschutnig, Chrisoph Rösli, and Jakob Kremer
- Subjects
Cancer Research ,Cluster of differentiation ,Hematopoietic stem cell ,Cell Biology ,Hematology ,Embryoid body ,Biology ,Embryonic stem cell ,Cell biology ,Haematopoiesis ,chemistry.chemical_compound ,medicine.anatomical_structure ,RUNX1 ,chemistry ,Cell culture ,Genetics ,medicine ,Stem cell ,Molecular Biology - Abstract
Endothelial to hematopoietic transition (EHT) is a crucial step in the formation of definitive hematopoietic stem cells during embryonic development. Holding such a critical developmental role not many details regarding molecular changes and cell surface marker (CSM) expression on these transitional cells are known. We think that sub-segregating the process of EHT holds great therapeutic potential by giving new indications to understand the hematopoietic stem cell maturation. We generated embryonic stem (ES) cell lines from Hoxb4-YFP reporter mice (Hills, 2011), in which YFP expression marks functional HSCs in adult and embryonic mice. When these cell lines were subject to embryoid body differentiation assays, we observed a transient Hoxb4/YFP+ cell population, which corresponded to the emergence of hemogenic endothelial cells in culture. We then analyzed the gene expression profile of the Hoxb4+ cells and compared it to immediate precursor (Hoxb4-Flk1+) and daughter (Hoxb4-CD41+) cell populations. As well as documenting the expression of numerous molecular markers previously associated with EHT, we also observed an elevated inflammatory gene expression signature that has previously been characterized as a mediator of hematopoietic specification in vivo. We were additionally able to identify 45 novel cell surface markers that could potentially be used to prospectively isolate and sub segregate cells undergoing EHT. 26 of these targets were subsequently verified using MRM mass spectrometry. To establish if any of these markers were functionally relevant, we generated knockout (KO) cell lines using CRISPR/Cas9. Upon EB differentiation, we observed a profound block in hematopoietic differentiation for ES cells that were KO for the membrane proteins Evi2a and Lyve1. This block was manifest at the EHT stage, as verified by time-lapse video imaging, and was equivalent in magnitude to Runx1 KO, suggesting a crucial role of Evi2a and Lyve1 during the EHT stage of hematopoietic development. Notably, this defect could be rescued by genetic replacement of the deleted gene. When comparing our data to human fetal liver data sets Evi2a is highly expressed in fetal liver, as well as in HSCs isolated from murine AGM and fetal liver.
- Published
- 2020
- Full Text
- View/download PDF
17. Abstract A042: A novel nonviral, nonintegrative DNA vector system for T-cell engineering
- Author
-
Patrick Schmidt, Dirk Jaeger, Richard Harbottle, and Matthias Bozza
- Subjects
Cancer Research ,Electroporation ,medicine.medical_treatment ,Immunology ,Mutagenesis (molecular biology technique) ,Computational biology ,Immunotherapy ,Biology ,Sleeping Beauty transposon system ,Chimeric antigen receptor ,Cancer immunotherapy ,Cancer cell ,medicine ,Vector (molecular biology) - Abstract
Adoptive immunotherapy is one of the most encouraging therapeutic strategies for the treatment of a range of cancers. A particularly promising avenue of research is the functional introduction of chimeric antigen receptors (CARs) into naive human T-cells for autologous immunotherapy. Currently, the genetic engineering of these cells is achieved through the use of integrating vector systems such as lentiviruses or the sleeping beauty transposon system, which present a potential risk of genotoxicity associated with their random genomic integration. We have invented a novel DNA vector platform for the safe and efficient generation of genetically engineered T-cells for human immunotherapy. This DNA vector system contains no viral components and comprises only clinically approved sequences; it does not integrate into the target-cell’s genome but it can replicate autonomously and extrachromosomally in the nuclei of dividing human primary cells. These DNA vectors offer several advantages over currently used vector systems; they are not subject to commercial licenses, they are cheaper and easier to produce, and they can more quickly genetically modify human cells without the inherent risk of integrative mutagenesis. In preclinical experiments we have successfully generated genetically engineered human T-cells that sustain the expression of a reporter gene for over a month at persistently high levels without decline. We have also successfully modified these cells with a range of transgenic CAR receptors against several known cancer cell epitopes and we have demonstrated their viability and capability in the targeted killing of these human cancer cells. We showed that CAR-T-cells generated with our technology killed more efficiently target cells when compared to T-cells engineered with current state-of-the-art integrative lentivirus. The expression of functional CARs was detected over a period of two weeks of administration in culture and the anticancer activity of our DNA-CAR-T-cells was evaluated in vivo using xenotransplanted cell lines in immunodeficient mice. We are currently performing analyses in order to determine the molecular behavior of the vector in the cells and its impact on cellular viability. Furthermore, we are developing a protocol for large scale electroporation in order to manufacture a clinical grade CAR-T DNA product. We believe that this novel DNA vector system provides a unique and innovative approach to this exciting therapeutic strategy for cancer therapy. We estimate that this novel methodology will provide a simpler method of CAR T-cell manufacturing, resulting in a 10-fold reduction in the cost of the CAR-T product. Citation Format: Patrick Schmidt, Matthias Bozza, Dirk Jaeger, Richard Harbottle. A novel nonviral, nonintegrative DNA vector system for T-cell engineering [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A042.
- Published
- 2019
- Full Text
- View/download PDF
18. Abstract 3573: Novel DNA vectors encoding a chimeric antigen receptor mediate long term expression without genomic integration
- Author
-
Inka Zörnig, Aileen Berger, Claudia Luckner-Minden, Patrick Schmidt, Alexandra Tuch, Dirk Jäger, Richard Harbottle, and Matthias Bozza
- Subjects
Cancer Research ,chemistry.chemical_compound ,Oncology ,chemistry ,Encoding (memory) ,Biology ,DNA ,Chimeric antigen receptor ,Term (time) ,Cell biology - Abstract
Adoptive immunotherapy is one of the most encouraging therapeutic strategies for the treatment of a range of cancers. One particularly promising avenue of research is the functional introduction of Chimeric Antigen Receptors (CARs) into naive Human T-Cells for autologous-immunotherapy. Currently, the genetic engineering of these cells is achieved through the use of proprietary integrating vector systems such as lentiviruses or the sleeping beauty transposon system which present a risk of genotoxicity associated with their random genomic integration. We have invented a novel DNA Vector platform for the safe and efficient generation of genetically engineered T-Cells for Human Immunotherapy. This DNA vector system contains no viral components and comprises only clinically approved sequences, it does not integrate into the target cell's genome but it can replicate autonomously and extrachromosomally in the nucleus of dividing human primary cells. These DNA Vectors offer several advantages over currently used vector systems; they are not subject to commercial licences, they are cheaper and easier to produce, and they can more quickly genetically modify human cells without the inherent risk of integrative mutagenesis. In preclinical experiments we have successfully generated genetically engineered human T-Cells expressing the CAR receptor against several epitopes and have demonstrated their viability and capability in targeting and killing human cancer cells which express these epitopes. The long term anti-tumor activity of our DNA-CAR-T cells has been confirmed in vivo using xenotransplanted cell lines in immunodeficient mice. We believe that this novel DNA Vector system provides a unique and innovative approach to this exciting therapeutic strategy for cancer therapy. We estimate that this novel methodology will provide a simpler method of CAR T-cell manufacturing, resulting in a 10-fold reduction in the cost of the CART-product. Citation Format: Patrick Schmidt, Matthias Bozza, Aileen Berger, Claudia Luckner-Minden, Alexandra Tuch, Inka Zörnig, Dirk Jäger, Richard Harbottle. Novel DNA vectors encoding a chimeric antigen receptor mediate long term expression without genomic integration [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 3573.
- Published
- 2018
- Full Text
- View/download PDF
19. Abstract 644: Utilizing a novel luciferase labeling technique to establish and validate preclinical models of pancreatic cancer
- Author
-
Jenni Bernoulli, Richard Harbottle, Mari I. Suominen, Johanna Tuomela, Jussi M. Halleen, Matthias Bozza, Suzanne J. Dilly, Katja M. Fagerlund, and George Morris
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Reporter gene ,Cell growth ,Cancer ,Transfection ,Biology ,medicine.disease ,Gemcitabine ,Oncology ,Cell culture ,Pancreatic cancer ,medicine ,Cancer research ,Luciferase ,medicine.drug - Abstract
Bioluminescent-labeling imaging (BLI) allows sensitive non-invasive sequential imaging of tumor development and early metastasis. Current methods for the genetic modification of cells typically use integrating genotoxic viruses that can disrupt the molecular behavior of cancer cell lines due to their random nature of integration. A primary aim of the study was to utilize a non-integrating DNA vector that comprises an S/MAR (Scaffold/Matrix Attachment Region) element to stably genetically modify pancreatic cancer cells to persistently express the reporter gene luciferase without altering the molecular behavior of the cell or altering its sensitivity to therapeutic drug treatments. Once a novel isogenic cell line is generated the cells can subsequently be used in xenograft studies. A second aim was to validate these established models with gemcitabine and test the efficacy of VAL401, formulation of risperidone in rumenic acid. Human BxPC3, Capan-1, MiaPaCa-2 and Panc-1 pancreatic cancer cells were stably transfected with a pSMARt-UBC-Luc DNA vector and cultured for 4 weeks under selection. Colonies that formed after this period were isolated and expanded in normal medium and evaluated for luciferase expression and the molecular integrity of the DNA vector. Efficacy of gemcitabine was tested in these new luciferase expressing cell lines and VAL401 was tested in Capan-1-luc cells. For in vivo studies, BxPC3-luc cells were inoculated orthotopically into the pancreas of athymic nude mice and stratified into groups: control, gemcitabine, VAL401 (1mg/kg, p.o. daily) and VAL401 (2mg/kg, p.o. daily). In vitro validation results indicated that the luciferase transfected cells maintained their original properties with stable expression. Gemcitabine inhibited cell proliferation in all established cell lines. VAL401 inhibited cell proliferation of Capan-1-luc cells at 50 μM concentration. BxPC3-luc cells inoculated orthotopically into the pancreas were followed for 5 weeks with BLI by IVIS, and the results demonstrated high-quality follow-up of tumor growth. BxPC3-luc cells induced growth of pancreatic tumors with high take rate in all groups. Gemcitabine and both studied doses of VAL401 decreased tumor volume, and the same trend was seen in tumor weight and the BLI during the study. In conclusion, both gemcitabine and VAL401 decreased tumor volume and the same trend was observed using BLI. Our results demonstrated that S/MAR DNA vectors are able to produce genetically modified cells without the limitations of random genomic integration, whilst providing extra-chromosomal mitotic stability and high levels of sustained transgene expression. When utilized in orthotopic xenograft studies, these luciferase expressing cells formed a reliable and essentially non-invasive imaging platform that substantially improves the efficacy of testing anticancer drug candidates. Citation Format: Jenni Bernoulli, Matthias Bozza, Katja M. Fagerlund, Johanna Tuomela, Mari I. Suominen, Suzanne Dilly, George Morris, Jussi M. Halleen, Richard Harbottle. Utilizing a novel luciferase labeling technique to establish and validate preclinical models of pancreatic cancer. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 644.
- Published
- 2016
- Full Text
- View/download PDF
20. Abstract B69: VAL401 decreases tumor volume in a xenograft model of pancreatic cancer utilizing a novel improved luciferase labelling technique
- Author
-
George Morris, Jenni Bernoulli, Jussi M. Halleen, Johanna Tuomela, Mari I. Suominen, Suzanne J. Dilly, Matthias Bozza, Richard Harbottle, and Katja M. Fagerlund
- Subjects
Cancer Research ,Chemistry ,Cancer ,medicine.disease ,Gemcitabine ,Metastasis ,medicine.anatomical_structure ,Oncology ,In vivo ,Pancreatic cancer ,Immunology ,medicine ,Cancer research ,Bioluminescence imaging ,Luciferase ,Pancreas ,medicine.drug - Abstract
Bioluminescent-labelling allows sensitive non-invasive sequential imaging of tumor development and early metastasis. Current methods for the genetic modification of cells typically use integrating genotoxic viruses that can potentially disrupt the molecular behavior of cancer cell lines due to their random nature of integration. VAL401 is the reformulation of a clinical drug to enable use in the treatment of cancer. Preclinical data indicate potential use of the reformulated drug in lung cancer, where many subsets of patients have currently a high unmet medical need. We have utilized a non-viral DNA vector that comprises an S/MAR (Scaffold/Matrix Attachment Region) element to stably modify cells to be further used in xenograft studies to allow long term expression without affecting cell behavior or silencing over cell divisions. Human BxPC3 pancreatic cancer cells were stably transfected with a pSMARt-UBC-Luc and cultured for 4 weeks under selection. Colonies that formed after this period were isolated and expanded in normal medium and evaluated for luciferase expression and molecular integrity of the DNA vector. For in vivo studies, BxPC3 cells were inoculated orthotopically into the pancreas of athymic nude mice. Four experimental groups were included in the study: 1) Control group receiving vehicle; 2) Reference compound gemcitabine (60 mg/kg, q3dx4 i.p, one week pause, q3dx4 i.p.); 3) Test compound VAL401 (1 mg/kg, p.o. daily); 4) Test compound VAL401 (2 mg/kg, p.o. daily). The luciferase transfected cells maintained their original properties with stable expression. Luciferase-labelled BxPC3 cells inoculated orthotopically into the pancreas were successfully followed for 5 weeks with non-invasive bioluminescence imaging by IVIS, and the results demonstrated high-quality follow-up of tumor growth compared with tumor models using non-labelled cells. BxPC3-luc cells induced growth of pancreatic tumors with high take rate in all groups. Gemcitabine and both studied doses of VAL401 decreased tumor volume, and the same trend was seen in tumor weight and the BLI parameters (total flux, area and average radiance) during the study. In conclusion, both gemcitabine and VAL401 decreased tumor volume and same trend was seen in BLI. Our results demonstrated that S/MAR DNA vectors are able to produce genetically modified cells without the limitations of random genomic integration, whilst providing extra-chromosomal mitotic stability and sustained transgene expression at high level. When utilized in orthotopic xenograft studies, these luciferase expressing cells formed a reliable and essential non-invasive imaging platform that improves substantially efficacy testing of anticancer drug candidates. Citation Format: Mari I. Suominen, Jenni Bernoulli, Suzanne Dilly, Johanna Tuomela, Matthias Bozza, Katja M. Fagerlund, George Morris, Jussi M. Halleen, Richard Harbottle. VAL401 decreases tumor volume in a xenograft model of pancreatic cancer utilizing a novel improved luciferase labelling technique. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2015 Nov 5-9; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(12 Suppl 2):Abstract nr B69.
- Published
- 2015
- Full Text
- View/download PDF
21. Abstract 3246: Novel luciferase labelling technique to improve imaging of orthotopic prostate and pancreatic cancer models
- Author
-
George Morris, Jenni Bernoulli, Katja M. Fagerlund, Matthias Bozza, Jussi M. Halleen, Johanna Tuomela, Richard Harbottle, and Mari I. Suominen
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Cell ,Cancer ,Transfection ,Biology ,medicine.disease ,Metastasis ,Prostate cancer ,medicine.anatomical_structure ,Oncology ,Pancreatic cancer ,medicine ,Cancer research ,Bioluminescence imaging ,Luciferase - Abstract
Bioluminescent-labelling allows sensitive non-invasive sequential imaging of tumor development and early metastasis. However, current methods for the genetic modification of cells typically use integrating genotoxic viruses that can potentially disrupt the molecular behavior of cancer cell lines due to their random nature of integration. Here, we utilized a non-viral DNA vector that comprises an S/MAR (Scaffold/Matrix Attachment Region) element to stably modify cells to be further used in xenograft studies to allow long term expression without affecting cell behavior or silencing over cell divisions. Human PC-3 prostate cancer cells and BxPC-3 pancreatic cancer cells were stably transfected with a pCAG-LUC-S/MAR and cultured for 4 weeks under selection. Colonies that formed after this period were isolated and expanded in normal medium and evaluated for luciferase expression and molecular integrity of the DNA vector. For in vivo studies, PC-3 cells were inoculated orthotopically into the prostate, and BxPC-3 cells into the pancreas using athymic and BALB/c nude mice. For comparison, similar experiments with the corresponding study designs were performed with the non-labelled parental cell lines. The luciferase transfected cells maintained their original properties with stable expression. Luciferase-labelled PC-3 and BxPC-3 cells inoculated orthotopically into the prostate and pancreas, respectively, were successfully followed for 5 weeks with non-invasive bioluminescence imaging by IVIS. The results demonstrated high-quality follow-up of tumor growth compared with tumor models using non-labelled cells. In conclusion, S/MAR DNA vectors are able to produce genetically modified cells without the limitations of random genomic integration, whilst providing extra-chromosomal mitotic stability and sustained transgene expression at high level. When utilized in orthotopic xenograft studies, these luciferase expressing cells formed a reliable and essential non-invasive imaging platform that improves substantially efficacy testing of anticancer drug candidates. Citation Format: Jenni Bernoulli, Johanna Tuomela, Matthias Bozza, Katja M. Fagerlund, Mari I. Suominen, George Morris, Jussi M. Halleen, Richard Harbottle. Novel luciferase labelling technique to improve imaging of orthotopic prostate and pancreatic cancer models. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 3246. doi:10.1158/1538-7445.AM2015-3246
- Published
- 2015
- Full Text
- View/download PDF
22. Integrins and angiogenesis: unlocking the route to gene therapy
- Author
-
Ajay, Mistry, Richard, Harbottle, Steve, Hart, and Kairbaan M, Hodivala-Dilke
- Subjects
Integrins ,Neovascularization, Pathologic ,Genetic Vectors ,Humans ,Endothelium, Vascular ,Genetic Therapy ,Adenoviridae - Abstract
Angiogenesis is defined as the process of vascularization of a tissue, involving the development of new capillary blood vessels. Both the induction and inhibition of this process can have therapeutic benefits in various pathological conditions. Integrins are a structurally elaborate family of adhesion molecules; they participate in a wide range of biological processes, including angiogenesis. Endothelial cells are intimately involved in angiogenesis and are thought to mediate this function partially through the integrins on their cell membrane, which regulate cell-cell and cell-matrix contacts. Extensive research into elucidating the mechanisms involved in the angiogenesis process have led to the discovery of a growing number of genes encoding pro- and anti-angiogenic proteins. A variety of gene therapy approaches have been used to deliver many of these genes to induce or inhibit the angiogenesis process with varying levels of success. This review investigates whether targeting gene therapy vectors to integrin receptors found on endothelial cells is a viable means to improve the efficiency of the gene transfer process.
- Published
- 2004
23. IMMU-41. HIGH-THROUGHPUT RETRIEVAL OF THERAPEUTIC T CELL RECEPTORS FROM GLIOMA
- Author
-
Lindner K, Tan C, Bozza M, Sanghvi K, Poschke I, Richard Harbottle, von Deimling A, Wick W, Sahm F, Platten M, Green E, and Bunse L
24. Gene delivery and expression mediated by an integrin-binding peptide
- Author
-
Sl, Hart, Richard Harbottle, Cooper R, Miller A, Williamson R, and Coutelle C
- Subjects
Integrins ,Antigens, Polyomavirus Transforming ,Molecular Sequence Data ,Gene Transfer Techniques ,Gene Expression ,DNA ,Genetic Therapy ,Recombinant Proteins ,Cell Line ,Drug Design ,Colonic Neoplasms ,Tumor Cells, Cultured ,Humans ,Polylysine ,Amino Acid Sequence ,Luciferases ,Oligopeptides - Abstract
The ability to transfer sufficient DNA to specific target cells remains one of the main limitations to the development of gene therapy. For this reason much attention is being paid to the development of new gene delivery systems, both viral and non-viral. We describe gene transfer with a polycation-DNA complex which contains an integrin-binding domain. Integrin-mediated gene delivery has several potential advantages. Such complexes are less likely than other receptor-mediated gene delivery complexes to be constrained by the size of the complex. The ligands are small peptides, resembling naturally occurring integrin ligands, which minimises the possibility of complexes inducing an immune response in vivo.
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.