1
|
Salman DM, Mohammad TAM. siRNA-based therapy for gastric adenocarcinoma: what's next step? Pathol Res Pract 2024; 258:155328. [PMID: 38744002 DOI: 10.1016/j.prp.2024.155328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 05/16/2024]
Abstract
Gastric cancer continues to have a high death rate despite advancements in their diagnosis and treatment. Novel treatment techniques are thus desperately needed. This is where double-stranded RNA molecules known as small interfering RNA (siRNA), which may selectively target the mRNA of disease-causing genes, may find use in medicine. For siRNAs to function properly in the human body, they must be shielded from deterioration. Furthermore, in order to maintain organ function, they must only target the tumor and spare normal tissue. siRNAs have been designed using clever delivery mechanisms including polymers and lipids to achieve these objectives. Although siRNA protection is not hard to acquire, it is still challenging to target cancer cells with them. Here, we first discuss the basic characteristics of gastric cancer before describing the properties of siRNA and typical delivery methods created specifically for gastric tumors. Lastly, we provide a succinct overview of research using siRNAs to treat gastric tumors.
Collapse
Affiliation(s)
- Dyar Mudhafar Salman
- Department of Clinical Pharmacy, College of Pharmacy, Hawler Medical University, Erbil, Kurdistan Region, Iraq; Faculty of Pharmacy, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Talar Ahmad Merza Mohammad
- Department of Clinical Pharmacy, College of Pharmacy, Hawler Medical University, Erbil, Kurdistan Region, Iraq; Pharmacy department, School of Medicine, University of Kurdistan Hewlêr (UKH), Erbil, Kurdistan Region, Iraq.
| |
Collapse
|
2
|
Schmohl KA, Dolp P, Schug C, Knoop K, Klutz K, Schwenk N, Bartenstein P, Nelson PJ, Ogris M, Wagner E, Spitzweg C. Reintroducing the Sodium-Iodide Symporter to Anaplastic Thyroid Carcinoma. Thyroid 2017; 27:1534-1543. [PMID: 29032724 DOI: 10.1089/thy.2017.0290] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Anaplastic thyroid carcinoma (ATC), the most aggressive form of thyroid cancer, is unresponsive to radioiodine therapy. The current study aimed to extend the diagnostic and therapeutic application of radioiodine beyond the treatment of differentiated thyroid cancer by targeting the functional sodium-iodide symporter (NIS) to ATC. METHODS The study employed nanoparticle vectors (polyplexes) based on linear polyethylenimine (LPEI), shielded by polyethylene glycol (PEG) and coupled to the synthetic peptide GE11 as an epidermal growth factor receptor (EGFR)-specific ligand in order to target a NIS-expressing plasmid (LPEI-PEG-GE11/NIS) to EGFR overexpressing human thyroid carcinoma cell lines. Using ATC xenograft mouse models, transfection efficiency by 123I scintigraphy and potential for systemic radioiodine therapy after systemic polyplex application were evaluated. RESULTS In vitro iodide uptake studies in SW1736 and Hth74 ATC cells, and, for comparison, in more differentiated follicular (FTC-133) and papillary (BCPAP) thyroid carcinoma cells demonstrated high transfection efficiency and EGFR-specificity of LPEI-PEG-GE11/NIS that correlated well with EGFR expression levels. After systemic polyplex injection, in vivo 123I gamma camera imaging revealed significant tumor-specific accumulation of radioiodine in an SW1736 and an Hth74 xenograft mouse model. Radioiodine accumulation was found to be higher in SW1736 tumors, reflecting in vitro results, EGFR expression levels, and results from ex vivo analysis of NIS staining. Administration of 131I in LPEI-PEG-GE11/NIS-treated SW1736 xenograft mice resulted in significantly reduced tumor growth associated with prolonged survival compared to control animals. CONCLUSIONS The data open the exciting prospect of NIS-mediated radionuclide imaging and therapy of ATC after non-viral reintroduction of the NIS gene. The high tumor specificity after systemic application makes the strategy an attractive alternative for the treatment of highly metastatic ATC.
Collapse
Affiliation(s)
- Kathrin A Schmohl
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Patrick Dolp
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Christina Schug
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Kerstin Knoop
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Kathrin Klutz
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Nathalie Schwenk
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Peter Bartenstein
- 2 Department of Nuclear Medicine, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Peter J Nelson
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Manfred Ogris
- 3 Department of Pharmaceutical Chemistry, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), University of Vienna , Vienna, Austria
| | - Ernst Wagner
- 4 Department of Pharmaceutical Biotechnology, Department of Pharmacy, Center for System-Based Drug Research and Center for Nanoscience , LMU Munich, Munich, Germany
| | - Christine Spitzweg
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| |
Collapse
|
3
|
Berchel M, Akhter S, Berthe W, Gonçalves C, Dubuisson M, Pichon C, Jaffrès PA, Midoux P. Synthesis of α-amino-lipophosphonates as cationic lipids or co-lipids for DNA transfection in dendritic cells. J Mater Chem B 2017; 5:6869-6881. [DOI: 10.1039/c7tb01080j] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Cationic lipid/co-lipid combinations have been extensively explored in gene delivery as alternatives to viral vectors.
Collapse
Affiliation(s)
- Mathieu Berchel
- CEMCA
- UMR CNRS 6521
- University of Brest
- IBSAM
- 6 Avenue Victor Le Gorgeu
| | - Sohail Akhter
- Centre de Biophysique Moléculaire
- CNRS UPR4301
- rue Charles Sadron CS 80054 F-45071 Orléans Cedex 02 and University of Orléans
- France
- Le Studium® Loire Valley Institute for Advanced Studies
| | - Wilfried Berthe
- CEMCA
- UMR CNRS 6521
- University of Brest
- IBSAM
- 6 Avenue Victor Le Gorgeu
| | - Cristine Gonçalves
- Centre de Biophysique Moléculaire
- CNRS UPR4301
- rue Charles Sadron CS 80054 F-45071 Orléans Cedex 02 and University of Orléans
- France
| | - Marine Dubuisson
- Centre de Biophysique Moléculaire
- CNRS UPR4301
- rue Charles Sadron CS 80054 F-45071 Orléans Cedex 02 and University of Orléans
- France
| | - Chantal Pichon
- Centre de Biophysique Moléculaire
- CNRS UPR4301
- rue Charles Sadron CS 80054 F-45071 Orléans Cedex 02 and University of Orléans
- France
| | | | - Patrick Midoux
- Centre de Biophysique Moléculaire
- CNRS UPR4301
- rue Charles Sadron CS 80054 F-45071 Orléans Cedex 02 and University of Orléans
- France
| |
Collapse
|
4
|
Phase Transition Detection in Accumulation of a Potential Anticancer Drug Cl-IPBD with DNA: Supercoiled and Linear pUC19 Plasmids. Electrochim Acta 2016. [DOI: 10.1016/j.electacta.2016.05.159] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
5
|
Figueroa JA, Reidy A, Mirandola L, Trotter K, Suvorava N, Figueroa A, Konala V, Aulakh A, Littlefield L, Grizzi F, Rahman RL, Jenkins MR, Musgrove B, Radhi S, D'Cunha N, D'Cunha LN, Hermonat PL, Cobos E, Chiriva-Internati M. Chimeric antigen receptor engineering: a right step in the evolution of adoptive cellular immunotherapy. Int Rev Immunol 2015; 34:154-187. [PMID: 25901860 DOI: 10.3109/08830185.2015.1018419] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Cancer immunotherapy comprises different therapeutic strategies that exploit the use of distinct components of the immune system, with the common goal of specifically targeting and eradicating neoplastic cells. These varied approaches include the use of specific monoclonal antibodies, checkpoint inhibitors, cytokines, therapeutic cancer vaccines and cellular anticancer strategies such as activated dendritic cell (DC) vaccines, tumor-infiltrating lymphocytes (TILs) and, more recently, genetically engineered T cells. Each one of these approaches has demonstrated promise, but their generalized success has been hindered by the paucity of specific tumor targets resulting in suboptimal tumor responses and unpredictable toxicities. This review will concentrate on recent advances on the use of engineered T cells for adoptive cellular immunotherapy (ACI) in cancer.
Collapse
Affiliation(s)
- Jose A Figueroa
- Division of Hematology and Oncology, Texas Tech University Health Sciences Center , Lubbock, TX , USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Kostka L, Šubr V, Laga R, Chytil P, Ulbrich K, Seymour LW, Etrych T. Nanotherapeutics shielded with a pH responsive polymeric layer. Physiol Res 2015; 64:S29-40. [PMID: 26447593 DOI: 10.33549/physiolres.933139] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Efficient intravenous delivery is the greatest single hurdle, with most nanotherapeutics frequently found to be unstable in the harsh conditions of the bloodstream. In the case of nanotherapeutics for gene delivery, viral vectors are often avidly recognized by both the innate and the adaptive immune systems. So, most modern delivery systems have benefited from being coated with hydrophilic polymers. Self-assembling delivery systems can achieve both steric and lateral stabilization following surface coating, endowing them with much improved systemic circulation properties and better access to disseminated targets; similarly, gene delivery viral vectors can be 'stealthed' and their physical properties modulated by surface coating. Polymers that start degrading under acidic conditions are increasingly investigated as a pathway to trigger the release of drugs or genes once the carrier reaches a slightly acidic tumor environment or after the carrier has been taken up by cells, resulting in the localization of the polymer in acidic endosomes and lysosomes. Advances in the design of acid-degradable drug and gene delivery systems have been focused and discussed in this article with stress placed on HPMA-based copolymers. We designed a system that is able to "throw away" the polymer coat after successful transport of the vector into a target cell. Initial biological studies were performed and it was demonstrated that this principle is applicable for real adenoviral vectors. It was shown that the transfection ability of coated virus at pH 7.4 is 75 times lower then transfection at pH 5.4.
Collapse
Affiliation(s)
- L Kostka
- Department of Biomedicinal Polymers, Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Prague, Czech Republic.
| | | | | | | | | | | | | |
Collapse
|
7
|
Üzek R, Özkara S, Güngüneş H, Uzun L, Şenel S. Magnetic Nanoparticles for Plasmid DNA Purification through Hydrophobic Interaction Chromatography. SEP SCI TECHNOL 2014. [DOI: 10.1080/01496395.2014.905958] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
8
|
Murayama S, Kos P, Miyata K, Kataoka K, Wagner E, Kato M. Gene Regulation by Intracellular Delivery and Photodegradation of Nanoparticles Containing Small Interfering RNA. Macromol Biosci 2014; 14:626-31. [DOI: 10.1002/mabi.201300393] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 11/22/2013] [Indexed: 12/18/2022]
Affiliation(s)
- Shuhei Murayama
- Pharmaceutical Biotechnology; Center for System-based Drug Research, Center for NanoScience (CeNS), Ludwig-Maximilians-University; Butenandtstrasse 5-13 Munich Germany
- Graduate School of Pharmaceutical Sciences and GPLLI Program; The University of Tokyo; 7-3-1 Hongo Bunkyo-ku Tokyo 113-0033 Japan
- Molecular Imaging Center; National Institute of Radiological Sciences; 4-9-1 Anagawa Inage-ku Chiba 263-8555 Japan
| | - Petra Kos
- Pharmaceutical Biotechnology; Center for System-based Drug Research, Center for NanoScience (CeNS), Ludwig-Maximilians-University; Butenandtstrasse 5-13 Munich Germany
| | - Kanjiro Miyata
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine; The University of Tokyo; 7-3-1 Hongo Bunkyo-ku Tokyo 113-0033 Japan
| | - Kazunori Kataoka
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine; The University of Tokyo; 7-3-1 Hongo Bunkyo-ku Tokyo 113-0033 Japan
- Department of Materials Engineering, Graduate School of Engineering; The University of Tokyo; 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8656 Japan
| | - Ernst Wagner
- Pharmaceutical Biotechnology; Center for System-based Drug Research, Center for NanoScience (CeNS), Ludwig-Maximilians-University; Butenandtstrasse 5-13 Munich Germany
| | - Masaru Kato
- Graduate School of Pharmaceutical Sciences and GPLLI Program; The University of Tokyo; 7-3-1 Hongo Bunkyo-ku Tokyo 113-0033 Japan
| |
Collapse
|
9
|
Formulation development of lyophilized, long-term stable siRNA/oligoaminoamide polyplexes. Eur J Pharm Biopharm 2013; 85:294-305. [DOI: 10.1016/j.ejpb.2013.05.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2013] [Revised: 05/12/2013] [Accepted: 05/21/2013] [Indexed: 11/18/2022]
|
10
|
Gniazdowska A, Palinska-Saadi A, Krawczyk E, Elzanowska H, Maj-Zurawska M. Supercoiled and linear plasmid DNAs interactions with methylene blue. Bioelectrochemistry 2013; 92:32-41. [DOI: 10.1016/j.bioelechem.2013.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Revised: 02/14/2013] [Accepted: 02/21/2013] [Indexed: 11/28/2022]
|
11
|
Shim MS, Xia Y. A reactive oxygen species (ROS)-responsive polymer for safe, efficient, and targeted gene delivery in cancer cells. Angew Chem Int Ed Engl 2013; 52:6926-9. [PMID: 23716349 DOI: 10.1002/anie.201209633] [Citation(s) in RCA: 301] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2012] [Revised: 04/17/2013] [Indexed: 01/05/2023]
Affiliation(s)
- Min Suk Shim
- The Wallace H. Coulter Department of Biomedical Engineering, School of Chemistry & Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | | |
Collapse
|
12
|
Shim MS, Xia Y. A Reactive Oxygen Species (ROS)-Responsive Polymer for Safe, Efficient, and Targeted Gene Delivery in Cancer Cells. Angew Chem Int Ed Engl 2013. [DOI: 10.1002/ange.201209633] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
13
|
Alhakamy NA, Berkland CJ. Polyarginine molecular weight determines transfection efficiency of calcium condensed complexes. Mol Pharm 2013; 10:1940-8. [PMID: 23534410 PMCID: PMC4207646 DOI: 10.1021/mp3007117] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Cell penetrating peptides (CPPs) have been extensively studied in polyelectrolyte complexes as a means to enhance the transfection efficiency of plasmid DNA (pDNA). Increasing the molecular weight of CPPs often enhances gene expression but poses a risk of increased cytotoxicity and immunogenicity compared to low molecular weight CCPs. Conversely, low molecular weight CPPs typically have low transfection efficiency due to large complex size. Complexes made using low molecular weight CPPs were found to be condensed to a small size by adding calcium. In this study, complexes of low molecular weight polyarginine and pDNA were condensed with calcium. These complexes showed high transfection efficiency and low cytotoxicity in A549 carcinomic human alveolar basal epithelial cells. The relationships between transfection efficiency and polyarginine size (5, 7, 9, or 11 amino acids), polyarginine/pDNA charge ratios, and calcium concentrations were studied. Polyarginine 7 was significantly more effective than other polyarginines under most formulation conditions, suggesting a link between cell penetration ability and transfection efficiency.
Collapse
Affiliation(s)
- Nabil A. Alhakamy
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, USA 66047
| | - Cory J. Berkland
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, USA 66047
- Department of Chemical & Petroleum Engineering, University of Kansas, Lawrence, KS, USA 66047
| |
Collapse
|
14
|
Kumagai M, Shimoda S, Wakabayashi R, Kunisawa Y, Ishii T, Osada K, Itaka K, Nishiyama N, Kataoka K, Nakano K. Effective transgene expression without toxicity by intraperitoneal administration of PEG-detachable polyplex micelles in mice with peritoneal dissemination. J Control Release 2012; 160:542-51. [PMID: 22484197 DOI: 10.1016/j.jconrel.2012.03.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Revised: 02/21/2012] [Accepted: 03/23/2012] [Indexed: 12/13/2022]
Abstract
Block copolymer of poly(ethylene glycol)-block-poly{N-[N-(2-aminoethyl)-2-aminoethyl]aspartamide} (PEG-P[Asp(DET)]) has been originally introduced as a promising gene carrier by forming a nanomicelle with plasmid DNA. In this study, the polyplex micelle of PEG-SS-P[Asp(DET)], which disulfide linkage (SS) between PEG and cationic polymer can detach the surrounding PEG chains upon intracellular reduction, was firstly evaluated with respect to in vivo transduction efficiency and toxicity in comparison to that of PEG-P[Asp(DET)] in peritoneally disseminated cancer model. Intraperitoneal (i.p.) administration of PEG-SS-P[Asp(DET)] polyplex micelles showed a higher (P<0.05) transgene expression compared with PEG-P[Asp(DET)] in tumors. In contrast, the delivered distribution of the micelles was not different between the two polyplex micelles. PEG-SS-P[Asp(DET)] micelle encapsulating human tumor necrosis factor α (hTNF-α) gene exhibits a higher antitumor efficacy against disseminated cancer compared with PEG-P[Asp(DET)] or saline control. No hepatic and renal toxicities were observed by the administration of polyplex micelles. In conclusion, PEG-detachable polyplex micelles may represent an advantage in gene transduction in vivo over PEG-undetachable polyplex micelles after i.p. administration for peritoneal dissemination of cancer.
Collapse
Affiliation(s)
- Michiaki Kumagai
- Innovation Center for Medical Redox Navigation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
OPA quantification of amino groups at the surface of Lipidic NanoCapsules (LNCs) for ligand coupling improvement. Int J Pharm 2011; 419:266-70. [DOI: 10.1016/j.ijpharm.2011.07.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Revised: 07/15/2011] [Accepted: 07/18/2011] [Indexed: 01/07/2023]
|
16
|
Philipp A, Meyer M, Zintchenko A, Wagner E. Functional modification of amide-crosslinked oligoethylenimine for improved siRNA delivery. REACT FUNCT POLYM 2011. [DOI: 10.1016/j.reactfunctpolym.2010.10.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
17
|
Epidermal growth factor receptor-targeted (131)I-therapy of liver cancer following systemic delivery of the sodium iodide symporter gene. Mol Ther 2011; 19:676-85. [PMID: 21245850 DOI: 10.1038/mt.2010.296] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
We recently demonstrated tumor-selective iodide uptake and therapeutic efficacy of radioiodine in neuroblastoma tumors after systemic nonviral polyplex-mediated sodium iodide symporter (NIS) gene delivery. In the present study, we used novel polyplexes based on linear polyethylenimine (LPEI), polyethylene glycol (PEG), and the synthetic peptide GE11 as an epidermal growth factor receptor (EGFR)-specific ligand to target a NIS-expressing plasmid to hepatocellular carcinoma (HCC) (HuH7). Incubation of HuH7 cells with LPEI-PEG-GE11/NIS polyplexes resulted in a 22-fold increase in iodide uptake, which was confirmed in other cancer cell lines correlating well with EGFR expression levels. Using (123)I-scintigraphy and ex vivo γ-counting, HuH7 xenografts accumulated 6.5-9% injected dose per gram (ID/g) (123)I, resulting in a tumor-absorbed dose of 47 mGray/Megabecquerel (mGy/MBq) (131)Iodide ((131)I) after intravenous (i.v.) application of LPEI-PEG-GE11/NIS. No iodide uptake was observed in other tissues. After pretreatment with the EGFR-specific antibody cetuximab, tumoral iodide uptake was markedly reduced confirming the specificity of EGFR-targeted polyplexes. After three or four cycles of polyplex/(131)I application, a significant delay in tumor growth was observed associated with prolonged survival. These results demonstrate that systemic NIS gene transfer using polyplexes coupled with an EGFR-targeting ligand is capable of inducing tumor-specific iodide uptake, which represents a promising innovative strategy for systemic NIS gene therapy in metastatic cancers.
Collapse
|
18
|
Functional Polymer Conjugates for Medicinal Nucleic Acid Delivery. POLYMERS IN NANOMEDICINE 2011. [DOI: 10.1007/12_2011_148] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
19
|
Yu H, Deng C, Tian H, Lu T, Chen X, Jing X. Chemo-physical and biological evaluation of poly(L-lysine)-grafted chitosan copolymers used for highly efficient gene delivery. Macromol Biosci 2010; 11:352-61. [PMID: 21188696 DOI: 10.1002/mabi.201000283] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2010] [Revised: 10/08/2010] [Indexed: 12/31/2022]
Abstract
For the success of non-viral gene delivery, it is of great importance to develop gene vectors with high efficiency but low toxicity. We demonstrate that PLL-grafted chitosan copolymers combine the advantages of PLL with its good pDNA-binding ability and of chitosan with its good biocompatibility. The chemo-physical properties of the prepared Chi-g-PLL copolymers are thoroughly characterized. The in vitro transfection study shows that the copolymers have a much higher gene transfer ability than the starting materials chitosan and PLL. A positive correlation between PLL chain lengths and transfection efficiency of the copolymers is found. Our results suggest that these novel Chi-g-PLL copolymers are good candidates for gene delivery in vivo.
Collapse
Affiliation(s)
- Haijun Yu
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | | | | | | | | | | |
Collapse
|
20
|
RNA interference-mediated silencing of Foxo3 in antigen-presenting cells as a strategy for the enhancement of DNA vaccine potency. Gene Ther 2010; 18:372-83. [PMID: 21107437 DOI: 10.1038/gt.2010.146] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The transcription factor Forkhead box O3 (Foxo3) has a critical role in suppressing the expansion of antigen-specific effector T-cell populations; hence, Foxo3 is a potential target for enhancing the antitumor immunity of cancer vaccines. In this report, we evaluated the potential of RNA interference (RNAi)-mediated silencing of Foxo3 in antigen-presenting cells as an adjuvant for HER2/neu DNA cancer vaccines. Bicistronic plasmids expressing the N-terminal extracellular domain of human HER-2/neu and the Foxo3 short hairpin RNA (hN'-neu-Foxo3 shRNA) or the scrambled control (hN'-neu-scramble shRNA) were subcutaneously injected into mice by gene gun administration to elicit antitumor immunity against p185neu-overexpressing MBT-2 bladder tumor cells. We found that mice treated with hN'-neu-Foxo3 shRNA showed greater reductions in tumor growth and longer survival times than mice treated with hN'-neu-scramble shRNA, indicating that the silencing of Foxo3 enhanced the antitumor efficacy of the HER-2/neu cancer vaccine. Cytotoxicity analyses further revealed that the Foxo3 shRNA-enhanced antitumor effect was associated with significant increases in the number of functional CD8(+) T cells and in the levels of cytotoxic T lymphocytes activity. Interleukin-6 was induced by hN'-neu-Foxo3 shRNA treatment but did not have a critical role in the antitumor effect of the hN'-neu-Foxo3 shRNA vaccine. Moreover, in vivo lymphocyte depletion analyses confirmed that the antitumor efficacy of the hN'-neu-Foxo3 shRNA vaccine depended on functional CD8(+) T cells. Finally, Foxo3 suppression was shown to markedly improve the effect of the HER-2/neu DNA vaccine in limiting the growth and lung metastases of MBT-2 cells. Overall, these results support RNAi-mediated silencing of Foxo3 as an effective strategy to enhance the therapeutic antitumor effect of HER-2/neu DNA vaccines against p185neu-positive tumors.
Collapse
|
21
|
de Vrij J, Willemsen RA, Lindholm L, Hoeben RC, Bangma CH, Barber C, Behr JP, Briggs S, Carlisle R, Cheng WS, Dautzenberg IJC, de Ridder C, Dzojic H, Erbacher P, Essand M, Fisher K, Frazier A, Georgopoulos LJ, Jennings I, Kochanek S, Koppers-Lalic D, Kraaij R, Kreppel F, Magnusson M, Maitland N, Neuberg P, Nugent R, Ogris M, Remy JS, Scaife M, Schenk-Braat E, Schooten E, Seymour L, Slade M, Szyjanowicz P, Totterman T, Uil TG, Ulbrich K, van der Weel L, van Weerden W, Wagner E, Zuber G. Adenovirus-derived vectors for prostate cancer gene therapy. Hum Gene Ther 2010; 21:795-805. [PMID: 19947826 DOI: 10.1089/hum.2009.203] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Prostate cancer is a leading cause of death among men in Western countries. Whereas the survival rate approaches 100% for patients with localized cancer, the results of treatment in patients with metastasized prostate cancer at diagnosis are much less successful. The patients are usually presented with a variety of treatment options, but therapeutic interventions in prostate cancer are associated with frequent adverse side effects. Gene therapy and oncolytic virus therapy may constitute new strategies. Already a wide variety of preclinical studies has demonstrated the therapeutic potential of such approaches, with oncolytic prostate-specific adenoviruses as the most prominent vector. The state of the art and future prospects of gene therapy in prostate cancer are reviewed, with a focus on adenoviral vectors. We summarize advances in adenovirus technology for prostate cancer treatment and highlight areas where further developments are necessary.
Collapse
Affiliation(s)
- Jeroen de Vrij
- Department of Molecular Cell Biology, Leiden University Medical Center , 2300 RC Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Schaffert D, Kiss M, Rödl W, Shir A, Levitzki A, Ogris M, Wagner E. Poly(I:C)-mediated tumor growth suppression in EGF-receptor overexpressing tumors using EGF-polyethylene glycol-linear polyethylenimine as carrier. Pharm Res 2010; 28:731-41. [PMID: 20694527 DOI: 10.1007/s11095-010-0225-4] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Accepted: 07/19/2010] [Indexed: 11/28/2022]
Abstract
PURPOSE To develop a novel polyethylenimine (PEI)-based polymeric carrier for tumor-targeted delivery of cytotoxic double-stranded RNA polyinosinic:polycytidylic acid, poly(I:C). The novel carrier should be chemically less complex but at least as effective as a previously developed tetra-conjugate containing epidermal growth factor (EGF) as targeting ligand, polyethylene glycol (PEG) as shielding spacer, 25 kDa branched PEI as RNA binding and endosomal buffering agent, and melittin as endosomal escape agent. METHODS Novel conjugates were designed employing a simplified synthetic strategy based on 22 kDa linear polyethylenimine (LPEI), PEG spacers, and recombinant EGF. The efficacy of various conjugates (different PEG spacers, with and without targeting EGF) in poly(I:C)-mediated cell killing was evaluated in vitro using two human U87MG glioma cell lines. The most effective polyplex was tested for in vivo activity in A431 tumor xenografts. RESULTS Targeting conjugate LPEI-PEG2 kDa-EGF was found as most effective in poly(I:C)-triggered killing of tumor cells in vitro. The efficacy correlated with glioma cell EGFR levels. Repeated intravenous administration of poly(I:C) polypexes strongly retarded growth of A431 human tumor xenograft in mice. CONCLUSIONS The optimized LPEI-PEG2 kDa-EGF conjugate displays reduced chemical complexity and efficient poly(I:C)-mediated killing of EGFR overexpressing tumors in vitro and in vivo.
Collapse
Affiliation(s)
- David Schaffert
- Pharmaceutical Biotechnology, Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität Munich, Butenandtstr. 5-13, 81377, Munich, Germany.
| | | | | | | | | | | | | |
Collapse
|
23
|
Oskuee RK, Philipp A, Dehshahri A, Wagner E, Ramezani M. The impact of carboxyalkylation of branched polyethylenimine on effectiveness in small interfering RNA delivery. J Gene Med 2010; 12:729-38. [DOI: 10.1002/jgm.1490] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
24
|
A liposome-based platform, VacciMax, and its modified water-free platform DepoVax enhance efficacy of in vivo nucleic acid delivery. Vaccine 2010; 28:6176-82. [PMID: 20656034 DOI: 10.1016/j.vaccine.2010.07.025] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Revised: 06/10/2010] [Accepted: 07/08/2010] [Indexed: 11/24/2022]
Abstract
Nucleic acid vaccines represent a promising alternative to killed bacterial antigen, recombinant protein or peptide vaccines for infectious diseases and cancer immunotherapy. Although significant advances are made with DNA vaccines in animal studies, there are severe limitations to deliver these vaccines effectively and considerable reservations exist about current methods used. In this study, a liposome-based vaccine platform, VacciMax (VM), and its modified water-free version, DepoVax (DPX), were tested for their ability to improve in vivo delivery of plasmid DNA (pDNA), mRNA and siRNA. Subcutaneously injected pDNA for IL12 and pDNA as well as mRNA for green fluorescent protein (GFP) in VM/DPX significantly enhanced their in vivo expression. Enhanced IL12 secretion and GFP expression was restricted to CD11b(+) and CD11c(+) antigen-presenting cells, but not B cells. Further, significant inhibition of plasmid/antigen-induced IL12 secretion was seen after injection of IL12-siRNA in VM. These findings suggest VM and DPX to be promising means of delivering nucleic acid vaccines in vivo, and warrant further studies on their role in inducing effective immune responses.
Collapse
|
25
|
pH-sensitive multi-PEGylated block copolymer as a bioresponsive pDNA delivery vector. Pharm Res 2010; 27:2260-73. [PMID: 20300803 DOI: 10.1007/s11095-010-0092-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2009] [Accepted: 02/09/2010] [Indexed: 10/19/2022]
Abstract
PURPOSE A reversibly-PEGylated diblock copolymer, poly(aspartate-hydrazide-poly(ethylene glycol))-block-poly(aspartate-diaminoethane) (p[Asp(Hyd-PEG)]-b-p[Asp(DET)]) was reported here for enhanced gene transfection and colloidal stability. The diblock copolymer possessed a unique architecture based on a poly(aspartamide) backbone. The first block, p[Asp(Hyd)], was used for multi-PEG conjugations, and the second block, p[Asp(DET)], was used for DNA condensation and endosomal escape. METHODS p[Asp(Hyd-PEG)]-b-p[Asp(DET)] was synthesized and characterized by (1)H-NMR. Polyplexes were formed by mixing the synthesized polymers and pDNA. The polyplex size, ζ-potential, and in vitro transfection efficiency were determined by dynamic light scattering, ζ-potential measurements, and luciferase assays, respectively. pH-dependent release of PEG from the polymer was monitored by cationic-exchange chromatography. RESULTS The polyplexes were 70-90 nm in size, and the surface charge was effectively shielded by a PEG layer. The transfection efficiency of the reversibly PEGylated polyplexes was confirmed to be comparable to that of the non-PEGylated counterparts and 1,000 times higher than that of the irreversibly PEGylated polyplexes. PEG release was demonstrated to be pH-sensitive. Fifty percent of the PEG was released within 30 min at pH 5, while the polymer incubated at pH 7.4 could still maintain 50% of PEG after 8 h. CONCLUSION The reversibly PEGylated polyplexes were shown to maintain polyplex stability without compromising transfection efficiency.
Collapse
|
26
|
de Martimprey H, Bertrand JR, Malvy C, Couvreur P, Vauthier C. New core-shell nanoparticules for the intravenous delivery of siRNA to experimental thyroid papillary carcinoma. Pharm Res 2010; 27:498-509. [PMID: 20087631 DOI: 10.1007/s11095-009-0043-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2009] [Accepted: 12/17/2009] [Indexed: 10/20/2022]
Abstract
PURPOSE Development of efficient in vivo delivery nanodevices remains a major challenge to achieve clinical application of siRNA. The present study refers to the conception of core-shell nanoparticles aiming to make possible intravenous administration of chemically unmodified siRNA oriented towards the junction oncogene of the papillary thyroid carcinoma. METHODS Nanoparticles were prepared by redox radical emulsion polymerization of isobutylcyanoacrylate and isohexylcyanoacrylate with chitosan. The loading of the nanoparticles with siRNA was achieved by adsorption. The biological activity of the siRNA-loaded nanoparticles was assessed on mice bearing a papillary thyroid carcinoma after intratumoral and intravenous administration. RESULTS Chitosan-coated nanoparticles with a diameter of 60 nm were obtained by adding 3% pluronic in the preparation medium. siRNA were associated with the nanoparticles by surface adsorption. In vivo, the antisense siRNA associated with the nanoparticles lead to a strong antitumoral activity. The tumor growth was almost stopped after intravenous injection of the antisense siRNA-loaded nanoparticles, while in all control experiments, the tumor size was increased by at least 10 times. CONCLUSION This work showed that poly(alkylcyanoacrylate) nanoparticles coated with chitosan are suitable carriers to achieve in vivo delivery of active siRNA to tumor including after systemic administration.
Collapse
Affiliation(s)
- Henri de Martimprey
- Université de Paris-Sud-11, UMR CNRS 8612, 5 rue J.B. Clément, 92296, Châtenay-Malabry, France
| | | | | | | | | |
Collapse
|
27
|
Abstract
Plasmid DNA and siRNA have a large potential for use as therapeutic nucleic acids in medicine. The way to the target cell and its proper compartment is full of obstacles. Polymeric carriers help to overcome the encountered barriers. Cationic polymers can interact with the nucleic acid in a nondamaging way but still require optimization with regard to transfer efficiency and biocompatibility. Aiming at virus-like features, as viruses are the most efficient natural gene carriers, the design of bioresponsive polymers shows promising results regarding DNA and siRNA delivery. By specific chemical modifications dynamic structures are created, programmed to respond towards changing demands on the delivery pathway by cleavage of labile bonds or conformational changes, thus enhancing biocompatible gene delivery.
Collapse
|
28
|
Schwerdt A, Zintchenko A, Concia M, Roesen N, Fisher K, Lindner LH, Issels R, Wagner E, Ogris M. Hyperthermia-induced targeting of thermosensitive gene carriers to tumors. Hum Gene Ther 2009. [PMID: 19866491 DOI: 10.1089/hgt.2008.064] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Locoregional hyperthermia (HT) can be used for site-directed activation of macromolecular drug delivery systems. We have developed a gene delivery system based on thermosensitive block copolymers (TSCs) with a phase transition temperature of 42 degrees C [Zintchenko, A., Ogris, M., and Wagner, E. (2006). Bioconjug. Chem. 17, 766-772], in which the statistical copolymer of vinylpyrrolidinone and N-isopropylacryamide is grafted on polyethylenimine (PEI). Here we applied polyplexes consisting of plasmid DNA and TSCs systemically in A/J mice bearing a syngeneic Neuro2A neuroblastoma tumor subcutaneously in each hind limb. One limb was selectively treated by HT at 42 degrees C, at the same time that polyplexes were injected via the tail vein. Hyperthermia led to increased accumulation of thermosensitive polymer and aggregation of thermosensitive polyplexes in HT-treated tumors, resulting in up to 10-fold increased DNA deposition compared with non-HT-treated tumor. The level of transgene expression induced by TSC polyplexes in HT-treated tumors was significantly higher and selective for tumor tissue. With nonthermosensitive PEI polyplexes HT did not influence transgene deposition or expression in tumor.
Collapse
Affiliation(s)
- Alenka Schwerdt
- Pharmaceutical Biology-Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität, Munich, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Pangburn TO, Petersen MA, Waybrant B, Adil MM, Kokkoli E. Peptide- and aptamer-functionalized nanovectors for targeted delivery of therapeutics. J Biomech Eng 2009; 131:074005. [PMID: 19655996 DOI: 10.1115/1.3160763] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Targeted delivery of therapeutics is an area of vigorous research, and peptide- and aptamer-functionalized nanovectors are a promising class of targeted delivery vehicles. Both peptide- and aptamer-targeting ligands can be readily designed to bind a target selectively with high affinity, and more importantly are molecules accessible by chemical synthesis and relatively compact compared with antibodies and full proteins. The multitude of peptide ligands that have been used for targeted delivery are covered in this review, with discussion of binding selectivity and targeting performance for these peptide sequences where possible. Aptamers are RNA or DNA strands evolutionarily engineered to specifically bind a chosen target. Although use of aptamers in targeted delivery is a relatively new avenue of research, the current state of the field is covered and promises of future advances in this area are highlighted. Liposomes, the classic drug delivery vector, and polymeric nanovectors functionalized with peptide or aptamer binding ligands will be discussed in this review, with the exclusion of other drug delivery vehicles. Targeted delivery of therapeutics, from DNA to classic small molecule drugs to protein therapeutics, by these targeted nanovectors is reviewed with coverage of both in vitro and in vivo deliveries. This is an exciting and dynamic area of research and this review seeks to discuss its broad scope.
Collapse
Affiliation(s)
- Todd O Pangburn
- Department of Chemical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | |
Collapse
|
30
|
Klutz K, Russ V, Willhauck MJ, Wunderlich N, Zach C, Gildehaus FJ, Göke B, Wagner E, Ogris M, Spitzweg C. Targeted radioiodine therapy of neuroblastoma tumors following systemic nonviral delivery of the sodium iodide symporter gene. Clin Cancer Res 2009; 15:6079-86. [PMID: 19789324 DOI: 10.1158/1078-0432.ccr-09-0851] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE We recently reported the significant therapeutic efficacy of radioiodine therapy in various tumor mouse models following transcriptionally targeted sodium iodide symporter (NIS) gene transfer. These studies showed the high potential of NIS as a novel diagnostic and therapeutic gene for the treatment of extrathyroidal tumors. As a next crucial step towards clinical application of NIS-mediated radionuclide therapy we aim at systemic delivery of the NIS gene to target extrathyroidal tumors even in the metastatic stage. EXPERIMENTAL DESIGN In the current study, we used synthetic polymeric vectors based on pseudodendritic oligoamines with high intrinsic tumor affinity (G2-HD-OEI) to target a NIS-expressing plasmid (CMV-NIS-pcDNA3) to neuroblastoma (Neuro2A) cells. RESULTS Incubation with NIS-containing polyplexes (G2-HD-OEI/NIS) resulted in a 51-fold increase in perchlorate-sensitive iodide uptake activity in Neuro2A cells in vitro. Through (123)I-scintigraphy and ex vivo gamma counting Neuro2A tumors in syngeneic A/J mice were shown to accumulate 8% to 13% ID/g (123)I with a biological half-life of 13 hours, resulting in a tumor-absorbed dose of 247 mGy/MBq (131)I after i.v. application of G2-HD-OEI/NIS. Nontarget organs, including liver, lung, kidneys, and spleen revealed no significant iodide uptake. Moreover, two cycles of systemic NIS gene transfer followed by (131)I application (55.5 MBq) resulted in a significant delay in tumor growth associated with markedly improved survival. CONCLUSIONS In conclusion, our data clearly show the high potential of novel pseudodendritic polymers for tumor-specific NIS gene delivery after systemic application, opening the prospect of targeted NIS-mediated radionuclide therapy of nonthyroidal tumors even in metastatic disease.
Collapse
Affiliation(s)
- Kathrin Klutz
- Department of Internal Medicine II, Ludwig-Maximilians-University, Munich, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Meyer M, Dohmen C, Philipp A, Kiener D, Maiwald G, Scheu C, Ogris M, Wagner E. Synthesis and Biological Evaluation of a Bioresponsive and Endosomolytic siRNA−Polymer Conjugate. Mol Pharm 2009; 6:752-62. [DOI: 10.1021/mp9000124] [Citation(s) in RCA: 151] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Martin Meyer
- Pharmaceutical Biotechnology, Center for Drug Research, Department of Pharmacy, Department of Chemistry and Biochemistry, and Center for Nanoscience, Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Christian Dohmen
- Pharmaceutical Biotechnology, Center for Drug Research, Department of Pharmacy, Department of Chemistry and Biochemistry, and Center for Nanoscience, Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Alexander Philipp
- Pharmaceutical Biotechnology, Center for Drug Research, Department of Pharmacy, Department of Chemistry and Biochemistry, and Center for Nanoscience, Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Daniel Kiener
- Pharmaceutical Biotechnology, Center for Drug Research, Department of Pharmacy, Department of Chemistry and Biochemistry, and Center for Nanoscience, Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Gelja Maiwald
- Pharmaceutical Biotechnology, Center for Drug Research, Department of Pharmacy, Department of Chemistry and Biochemistry, and Center for Nanoscience, Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Christina Scheu
- Pharmaceutical Biotechnology, Center for Drug Research, Department of Pharmacy, Department of Chemistry and Biochemistry, and Center for Nanoscience, Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Manfred Ogris
- Pharmaceutical Biotechnology, Center for Drug Research, Department of Pharmacy, Department of Chemistry and Biochemistry, and Center for Nanoscience, Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Center for Drug Research, Department of Pharmacy, Department of Chemistry and Biochemistry, and Center for Nanoscience, Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, 81377 Munich, Germany
| |
Collapse
|
32
|
Chen Y, Huang L. Tumor-targeted delivery of siRNA by non-viral vector: safe and effective cancer therapy. Expert Opin Drug Deliv 2009; 5:1301-11. [PMID: 19040393 DOI: 10.1517/17425240802568505] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
RNA interference technology has been developed as a potential therapeutic agent for many indications, including cancer. Silencing a specific oncogene in tumor cells brings about cell death both in vitro and in vivo. However, there is a great need for powerful delivery strategies to enhance the therapeutic effect of small interfering RNA (siRNA). This review summarizes different signaling pathways inhibited by siRNA and the advantages of targeted siRNA as a delivery system.
Collapse
Affiliation(s)
- Yunching Chen
- University of North Carolina at Chapel Hill, Eshelman School of Pharmacy, Division of Molecular Pharmaceutics, Campus Box 7360 Kerr Hall, Chapel Hill, NC 27599, USA
| | | |
Collapse
|
33
|
Lentacker I, Wang N, Vandenbroucke RE, Demeester J, De Smedt SC, Sanders NN. Ultrasound Exposure of Lipoplex Loaded Microbubbles Facilitates Direct Cytoplasmic Entry of the Lipoplexes. Mol Pharm 2009; 6:457-67. [DOI: 10.1021/mp800154s] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Ine Lentacker
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicine, Faculty of Pharmaceutical Sciences, Ghent University, Harelbekestraat 72, B-9000 Ghent, Belgium, and Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, B-9820 Merelbeke, Belgium
| | - Nan Wang
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicine, Faculty of Pharmaceutical Sciences, Ghent University, Harelbekestraat 72, B-9000 Ghent, Belgium, and Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, B-9820 Merelbeke, Belgium
| | - Roosmarijn E. Vandenbroucke
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicine, Faculty of Pharmaceutical Sciences, Ghent University, Harelbekestraat 72, B-9000 Ghent, Belgium, and Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, B-9820 Merelbeke, Belgium
| | - Jo Demeester
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicine, Faculty of Pharmaceutical Sciences, Ghent University, Harelbekestraat 72, B-9000 Ghent, Belgium, and Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, B-9820 Merelbeke, Belgium
| | - Stefaan C. De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicine, Faculty of Pharmaceutical Sciences, Ghent University, Harelbekestraat 72, B-9000 Ghent, Belgium, and Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, B-9820 Merelbeke, Belgium
| | - Niek N. Sanders
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicine, Faculty of Pharmaceutical Sciences, Ghent University, Harelbekestraat 72, B-9000 Ghent, Belgium, and Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, B-9820 Merelbeke, Belgium
| |
Collapse
|
34
|
Siders WM, Shields J, Kaplan J, Lukason M, Woodworth L, Wadsworth S, Scaria A. Cytotoxic T lymphocyte responses to transgene product, not adeno-associated viral capsid protein, limit transgene expression in mice. Hum Gene Ther 2009; 20:11-20. [PMID: 18828728 PMCID: PMC2855252 DOI: 10.1089/hum.2008.055] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2008] [Accepted: 09/30/2008] [Indexed: 11/12/2022] Open
Abstract
The use of adeno-associated viral (AAV) vectors for gene replacement therapy is currently being explored in several clinical indications. However, reports have suggested that input capsid proteins from AAV-2 vector particles may result in the stimulation of cytotoxic T lymphocyte (CTL) responses that can result in a loss of transduced cells. To explore the impact of anti-AAV CTLs on AAV-mediated transgene expression, both immunocompetent C57BL=6 mice and B cell-deficient muMT mice were immunized against the AAV2 capsid protein (Cap) and were injected intravenously with an AAV-2 vector encoding alpha-galactosidase (alpha-Gal). C57BL=6 mice, which developed both CTL and neutralizing antibody responses against Cap, failed to show any detectable alpha-Gal expression. In contrast, serum alpha-Gal levels comparable to those of naive mice were observed in muMT mice despite the presence of robust CTL activity against Cap, indicating that preexisting Cap-specific CTLs did not have any effect on the magnitude and duration of transgene expression. The same strategy was used to assess the impact of CTLs against the alpha-Gal transgene product on AAV-mediated gene delivery and persistence of transgene expression. Preimmunization of muMT mice with an Ad=alpha-Gal vector induced a robust CTL response to alpha-Gal. When these mice were injected with AAV2=alpha-Gal vector, initial levels of alpha-Gal expression were reduced by more than 1 log and became undetectable by 2 weeks postinjection. Overall, our results indicate that CTLs against the transgene product as opposed to AAV capsid protein are more likely to interfere with AAV transgene expression.
Collapse
Affiliation(s)
| | | | - Johanne Kaplan
- Department of Immunotherapy Research, Genzyme, Framingham, MA 01701
| | - Michael Lukason
- Department of Molecular Biology, Genzyme, Framingham, MA 01701
| | - Lisa Woodworth
- Department of Immunotherapy Research, Genzyme, Framingham, MA 01701
| | - Sam Wadsworth
- Department of Molecular Biology, Genzyme, Framingham, MA 01701
| | - Abraham Scaria
- Department of Molecular Biology, Genzyme, Framingham, MA 01701
| |
Collapse
|
35
|
|
36
|
Schwerdt A, Zintchenko A, Concia M, Roesen N, Fisher K, Lindner LH, Issels R, Wagner E, Ogris M. Hyperthermia-Induced Targeting of Thermosensitive Gene Carriers to Tumors. Hum Gene Ther 2008; 19:1283-92. [DOI: 10.1089/hum.2008.064] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Alenka Schwerdt
- Pharmaceutical Biology-Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität, D-81377 Munich, Germany
| | - Arkadi Zintchenko
- Pharmaceutical Biology-Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität, D-81377 Munich, Germany
| | - Massimo Concia
- Pharmaceutical Biology-Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität, D-81377 Munich, Germany
| | - Nick Roesen
- Hybrid Systems, Cherwell Innovation Centre, Upper Heyford, OX25 5HD Oxfordshire, United Kingdom
| | - Kerry Fisher
- Hybrid Systems, Cherwell Innovation Centre, Upper Heyford, OX25 5HD Oxfordshire, United Kingdom
| | - Lars H. Lindner
- Department of Internal Medicine III, Klinikum Grosshadern Medical Center (KGMC), Ludwig-Maximilians-Universität, D-81377 Munich, Germany
- CCG Hyperthermia, Helmholtz Zentrum München-National Research Center for Environment and Health, D-81377 Munich, Germany
| | - Rolf Issels
- Department of Internal Medicine III, Klinikum Grosshadern Medical Center (KGMC), Ludwig-Maximilians-Universität, D-81377 Munich, Germany
- CCG Hyperthermia, Helmholtz Zentrum München-National Research Center for Environment and Health, D-81377 Munich, Germany
| | - Ernst Wagner
- Pharmaceutical Biology-Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität, D-81377 Munich, Germany
| | - Manfred Ogris
- Pharmaceutical Biology-Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität, D-81377 Munich, Germany
| |
Collapse
|
37
|
Does a targeting ligand influence nanoparticle tumor localization or uptake? Trends Biotechnol 2008; 26:552-8. [PMID: 18722682 DOI: 10.1016/j.tibtech.2008.06.007] [Citation(s) in RCA: 250] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2008] [Revised: 06/12/2008] [Accepted: 06/16/2008] [Indexed: 11/21/2022]
Abstract
Inclusion of a tumor-targeting molecule in nanosized delivery systems increases their in vivo efficacy. However, the biodistribution and pharmacokinetics of the uptake of such particles have not yet been well addressed. Several recent papers have suggested that tumor-targeting ligands function primarily to increase intracellular uptake of the nanocomplex and do not influence tumor localization. However, other reports indicate that they do play a role in the accumulation in the tumor. One difference might be the presence or absence of poly-[ethylene glycol] (PEG) in the complex and its impact on the enhanced permeability and retention (EPR) effect. Further studies are clearly needed to more fully elucidate the influence of composition on tumor-targeted, systemic delivery of nanoparticles.
Collapse
|
38
|
The silent (R)evolution of polymeric nucleic acid therapeutics. Pharm Res 2008; 25:2920-3. [PMID: 18679773 DOI: 10.1007/s11095-008-9689-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2008] [Accepted: 07/14/2008] [Indexed: 10/21/2022]
|
39
|
Dass CR, Khachigian LM, Choong PF. c-Jun knockdown sensitizes osteosarcoma to doxorubicin. Mol Cancer Ther 2008; 7:1909-12. [DOI: 10.1158/1535-7163.mct-08-0086] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
40
|
Sakaguchi N, Kojima C, Harada A, Koiwai K, Emi N, Kono K. Effect of transferrin as a ligand of pH-sensitive fusogenic liposome-lipoplex hybrid complexes. Bioconjug Chem 2008; 19:1588-95. [PMID: 18627196 DOI: 10.1021/bc800126s] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
We previously developed potent nonviral vectors based on complexation of lipoplexes and pH-sensitive fusogenic liposomes, which achieve efficient transfection through membrane fusion with intracellular acidic compartments such as endosomes. Because transferrin receptor is known to be overexpressed in cancer cells, in this study, we investigated the effect of transferrin as a ligand for transfection of various cancer-derived cell lines mediated by the liposome-lipoplex hybrid complexes. Results showed that these hybrid complexes with transferrin exhibited higher transfection efficiency toward these cells than complexes without transferrin, but the extent of the transferrin-induced enhancement was dependent on the cell line. Conjugation of transferrin increased their transfection activity for HeLa and KB cells, although it only slightly enhanced transfection for HT1080, HepG2, and K562. Transferrin receptors in HT1080, HepG2, and K562 cells were internalized slowly, whereas those in HeLa and KB cells were internalized quickly and actively. These results indicate that transfection mediated by the ligand-attached hybrid complex does not correlate with the amount of transferrin receptor in the cell surface but correlate with the activity of internalization of transferrin receptor into the cells.
Collapse
Affiliation(s)
- Naoki Sakaguchi
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Nakaku, Sakai, Osaka 599-8531, Japan
| | | | | | | | | | | |
Collapse
|
41
|
Juliano R, Alam MR, Dixit V, Kang H. Mechanisms and strategies for effective delivery of antisense and siRNA oligonucleotides. Nucleic Acids Res 2008; 36:4158-71. [PMID: 18558618 PMCID: PMC2475625 DOI: 10.1093/nar/gkn342] [Citation(s) in RCA: 294] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The potential use of antisense and siRNA oligonucleotides as therapeutic agents has elicited a great deal of interest. However, a major issue for oligonucleotide-based therapeutics involves effective intracellular delivery of the active molecules. In this Survey and Summary, we review recent reports on delivery strategies, including conjugates of oligonucleotides with various ligands, as well as use of nanocarrier approaches. These are discussed in the context of intracellular trafficking pathways and issues regarding in vivo biodistribution of molecules and nanoparticles. Molecular-sized chemical conjugates and supramolecular nanocarriers each display advantages and disadvantages in terms of effective and nontoxic delivery. Thus, choice of an optimal delivery modality will likely depend on the therapeutic context.
Collapse
Affiliation(s)
- Rudy Juliano
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC 27599, USA.
| | | | | | | |
Collapse
|
42
|
Zintchenko A, Philipp A, Dehshahri A, Wagner E. Simple modifications of branched PEI lead to highly efficient siRNA carriers with low toxicity. Bioconjug Chem 2008; 19:1448-55. [PMID: 18553894 DOI: 10.1021/bc800065f] [Citation(s) in RCA: 364] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Polymer carriers like PEI which proved their efficiency in DNA delivery were found to be far less effective for the applications with siRNA. In the current study, we generated a number of nontoxic derivates of branched PEI through modification of amines by ethyl acrylate, acetylation of primary amines, or introduction of negatively charged propionic acid or succinic acid groups to the polymer structure. The resulting products showed high efficiency in siRNA-mediated knockdown of target gene. In particular, succinylation of branched PEI resulted in up to 10-fold lower polymer toxicity in comparison to unmodified PEI. Formulations of siRNA with succinylated PEI were able to induce remarkable knockdown (80% relative to untreated cells) of target luciferase gene at the lowest tested siRNA concentration of 50 nM in Neuro2ALuc cells. The polyplex stability assay revealed that the efficiency of formulations which are stable in physiological saline is independent of the affinity of siRNA to the polymer chain. The improved properties of modified PEI as siRNA carrier are largely a consequence of the lower polymer toxicity. In order to achieve significant knockdown of target gene, the PEI-based polymer has to be applied at higher concentrations, required most probably for sufficient accumulation and proton sponge effects in endosomes. Unmodified PEI is highly toxic at such polymer concentrations. In contrast, the far less toxic modified analogues can be applied in concentrations required for the knockdown of target genes without side effects.
Collapse
Affiliation(s)
- Arkadi Zintchenko
- Center for Drug Research, Department of Pharmacy, and Center for NanoScience (CeNS), Ludwig-Maximilians-University, Butenandstr. 5-13, D-81377 Munich, Germany.
| | | | | | | |
Collapse
|
43
|
Abstract
Low efficiency, significant toxicity, polymer polydispersity and poorly understood delivery mechanisms have initially plagued the field of polymer-based gene therapy. Numerous strategies have helped to improve polyplexes, including the development of biodegradable polymers with reduced toxicity, incorporation of cell targeting, surface shielding and additional transport domains for effective and specific delivery, or improved chemistry for syntheses of polymers with uniform size and topology. Combined biooptical imaging and bioinformatics, providing insights into transfer bottlenecks, have helped to design improved polyplexes. Bioresponsive multifunctional polymers adapt in a dynamic manner to delivery barriers for efficient transfer of pDNA or siRNA to the target site.
Collapse
|
44
|
Tietze N, Pelisek J, Philipp A, Roedl W, Merdan T, Tarcha P, Ogris M, Wagner E. Induction of Apoptosis in Murine Neuroblastoma by Systemic Delivery of Transferrin-Shielded siRNA Polyplexes for Downregulation of Ran. Oligonucleotides 2008; 18:161-74. [DOI: 10.1089/oli.2008.0112] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Nicole Tietze
- Pharmaceutical Biotechnology, Department of Pharmacy, Center of Drug Research, LMU Munich, Germany
| | - Jaroslav Pelisek
- Pharmaceutical Biotechnology, Department of Pharmacy, Center of Drug Research, LMU Munich, Germany
| | - Alexander Philipp
- Pharmaceutical Biotechnology, Department of Pharmacy, Center of Drug Research, LMU Munich, Germany
| | - Wolfgang Roedl
- Pharmaceutical Biotechnology, Department of Pharmacy, Center of Drug Research, LMU Munich, Germany
| | - Thomas Merdan
- Departments of Advanced Drug Delivery, Abbott Laboratories, 100 Abbott Park Road, North Chicago IL
| | - Peter Tarcha
- Departments of Advanced Drug Delivery, Abbott Laboratories, 100 Abbott Park Road, North Chicago IL
| | - Manfred Ogris
- Pharmaceutical Biotechnology, Department of Pharmacy, Center of Drug Research, LMU Munich, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Center of Drug Research, LMU Munich, Germany
| |
Collapse
|
45
|
Takae S, Miyata K, Oba M, Ishii T, Nishiyama N, Itaka K, Yamasaki Y, Koyama H, Kataoka K. PEG-Detachable Polyplex Micelles Based on Disulfide-Linked Block Catiomers as Bioresponsive Nonviral Gene Vectors. J Am Chem Soc 2008; 130:6001-9. [DOI: 10.1021/ja800336v] [Citation(s) in RCA: 360] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
46
|
Soto ER, Ostroff GR. Characterization of multilayered nanoparticles encapsulated in yeast cell wall particles for DNA delivery. Bioconjug Chem 2008; 19:840-8. [PMID: 18376856 DOI: 10.1021/bc700329p] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Nonviral gene delivery technologies have been developed using layer-by-layer self-assembly of nanomaterials held together by electrostatic interactions in order to provide nanoparticulate materials that protect and deliver DNA to cells. Here we report a new DNA delivery technology based on the in situ layer-by-layer synthesis of DNA nanoparticles caged within hollow yeast cell wall particles (YCWP). YCWP provide protection and facilitate oral and systemic receptor-targeted delivery of DNA payloads to phagocytic cells. The nanoparticles inside YCWP consist of a core of tRNA/polyethylenimine (PEI) followed by a DNA layer that is finally coated with a protective outer layer of PEI. Using fluorescein and rhodamine labeling of tRNA, PEI, and DNA, the layer-by-layer formation of the nanoparticles was visualized by fluorescent microscopy and quantitated by fluorescence spectroscopy and flow cytometry. Optimal conditions (tRNA:YCWP, PEI:YCWP ratios and DNA load levels) to synthesize YCWP encapsulated nanoparticles were determined from these results. The high in vitro transfection efficiency of this encapsulated DNA delivery technology was demonstrated by the transfection of NIH3T3-D1 cells with YCWP-tRNA/PEI/gWizGFP/PEI formulations containing low amounts of the plasmid gWizGFP per particle to maximally express green fluorescent protein (GFP).
Collapse
Affiliation(s)
- Ernesto R Soto
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street, Worcester, Massachusetts 01605, USA
| | | |
Collapse
|
47
|
Srinivasachari S, Fichter KM, Reineke TM. Polycationic β-Cyclodextrin “Click Clusters”: Monodisperse and Versatile Scaffolds for Nucleic Acid Delivery. J Am Chem Soc 2008; 130:4618-27. [DOI: 10.1021/ja074597v] [Citation(s) in RCA: 181] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
| | - Katye M. Fichter
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio 45221-0172
| | - Theresa M. Reineke
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio 45221-0172
| |
Collapse
|
48
|
Pirollo KF, Chang EH. Targeted Delivery of Small Interfering RNA: Approaching Effective Cancer Therapies: Figure 1. Cancer Res 2008; 68:1247-50. [DOI: 10.1158/0008-5472.can-07-5810] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
49
|
|
50
|
DeRouchey J, Schmidt C, Walker GF, Koch C, Plank C, Wagner E, Rädler JO. Monomolecular Assembly of siRNA and Poly(ethylene glycol)−Peptide Copolymers. Biomacromolecules 2008; 9:724-32. [DOI: 10.1021/bm7011482] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Jason DeRouchey
- Department of Physics, Department of Chemistry and Pharmacy, and Center for NanoScience, Ludwig-Maximilians-Universität, 80539 Munich, Germany, Institute of Experimental Oncology, Technische Universität, 81675 Munich, Germany, and National Institutes of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20692
| | - Claudia Schmidt
- Department of Physics, Department of Chemistry and Pharmacy, and Center for NanoScience, Ludwig-Maximilians-Universität, 80539 Munich, Germany, Institute of Experimental Oncology, Technische Universität, 81675 Munich, Germany, and National Institutes of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20692
| | - Greg F. Walker
- Department of Physics, Department of Chemistry and Pharmacy, and Center for NanoScience, Ludwig-Maximilians-Universität, 80539 Munich, Germany, Institute of Experimental Oncology, Technische Universität, 81675 Munich, Germany, and National Institutes of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20692
| | - Christian Koch
- Department of Physics, Department of Chemistry and Pharmacy, and Center for NanoScience, Ludwig-Maximilians-Universität, 80539 Munich, Germany, Institute of Experimental Oncology, Technische Universität, 81675 Munich, Germany, and National Institutes of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20692
| | - Christian Plank
- Department of Physics, Department of Chemistry and Pharmacy, and Center for NanoScience, Ludwig-Maximilians-Universität, 80539 Munich, Germany, Institute of Experimental Oncology, Technische Universität, 81675 Munich, Germany, and National Institutes of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20692
| | - Ernst Wagner
- Department of Physics, Department of Chemistry and Pharmacy, and Center for NanoScience, Ludwig-Maximilians-Universität, 80539 Munich, Germany, Institute of Experimental Oncology, Technische Universität, 81675 Munich, Germany, and National Institutes of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20692
| | - Joachim O. Rädler
- Department of Physics, Department of Chemistry and Pharmacy, and Center for NanoScience, Ludwig-Maximilians-Universität, 80539 Munich, Germany, Institute of Experimental Oncology, Technische Universität, 81675 Munich, Germany, and National Institutes of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20692
| |
Collapse
|