A Dual Targeting Dendrimer-Mediated siRNA Delivery System for Effective Gene Silencing in Cancer TherapyClick to copy article linkArticle link copied!
- Yiwen DongYiwen DongState Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, Center of Advanced Pharmaceutics and Biomaterials, China Pharmaceutical University, 24 Tong Jia Xiang, 210009 Nanjing, People’s Republic of ChinaMore by Yiwen Dong
- Tianzhu YuTianzhu YuAix-Marseille Université, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, UMR 7325, “Equipe Labellisée Ligue Contre le Cancer”, 13288 Marseille, FranceMore by Tianzhu Yu
- Ling DingLing DingAix-Marseille Université, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, UMR 7325, “Equipe Labellisée Ligue Contre le Cancer”, 13288 Marseille, FranceMore by Ling Ding
- Erik LauriniErik LauriniMolecular Biology and Nanotechnology Laboratory (MolBNL@UniTS), DEA, University of Trieste, 34127 Trieste, ItalyMore by Erik Laurini
- Yuanyu HuangYuanyu HuangAdvanced Research Institute of Multidisciplinary Science and School of Life Science, Beijing Institute of Technology, Beijing 100081, People’s Republic of ChinaSchool of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, People’s Republic of ChinaMore by Yuanyu Huang
- Mengjie ZhangMengjie ZhangAdvanced Research Institute of Multidisciplinary Science and School of Life Science, Beijing Institute of Technology, Beijing 100081, People’s Republic of ChinaMore by Mengjie Zhang
- Yuhua WengYuhua WengAdvanced Research Institute of Multidisciplinary Science and School of Life Science, Beijing Institute of Technology, Beijing 100081, People’s Republic of ChinaMore by Yuhua Weng
- Shuting LinShuting LinState Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, Center of Advanced Pharmaceutics and Biomaterials, China Pharmaceutical University, 24 Tong Jia Xiang, 210009 Nanjing, People’s Republic of ChinaMore by Shuting Lin
- Peng ChenPeng ChenState Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, Center of Advanced Pharmaceutics and Biomaterials, China Pharmaceutical University, 24 Tong Jia Xiang, 210009 Nanjing, People’s Republic of ChinaMore by Peng Chen
- Domenico MarsonDomenico MarsonMolecular Biology and Nanotechnology Laboratory (MolBNL@UniTS), DEA, University of Trieste, 34127 Trieste, ItalyMore by Domenico Marson
- Yifan JiangYifan JiangAix-Marseille Université, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, UMR 7325, “Equipe Labellisée Ligue Contre le Cancer”, 13288 Marseille, FranceMore by Yifan Jiang
- Suzanne GiorgioSuzanne GiorgioAix-Marseille Université, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, UMR 7325, “Equipe Labellisée Ligue Contre le Cancer”, 13288 Marseille, FranceMore by Suzanne Giorgio
- Sabrina PriclSabrina PriclMolecular Biology and Nanotechnology Laboratory (MolBNL@UniTS), DEA, University of Trieste, 34127 Trieste, ItalyMore by Sabrina Pricl
- Xiaoxuan Liu*Xiaoxuan Liu*[email protected]State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, Center of Advanced Pharmaceutics and Biomaterials, China Pharmaceutical University, 24 Tong Jia Xiang, 210009 Nanjing, People’s Republic of ChinaAix-Marseille Université, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, UMR 7325, “Equipe Labellisée Ligue Contre le Cancer”, 13288 Marseille, FranceMore by Xiaoxuan Liu
- Palma RocchiPalma RocchiInserm, Aix-Marseille Université, Institut Paoli-Calmettes, CNRS, CRCM, 13009 Marseille, FranceMore by Palma Rocchi
- Ling Peng*Ling Peng*[email protected]Aix-Marseille Université, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, UMR 7325, “Equipe Labellisée Ligue Contre le Cancer”, 13288 Marseille, FranceMore by Ling Peng
Abstract
Small interfering RNA (siRNA) is emerging as a novel therapeutic for treating various diseases, provided a safe and efficient delivery is available. In particular, specific delivery to target cells is critical for achieving high therapeutic efficacy while reducing toxicity. Amphiphilic dendrimers are emerging as novel promising carriers for siRNA delivery by virtue of the combined multivalent cooperativity of dendrimers with the self-assembling property of lipid vectors. Here, we report a ballistic approach for targeted siRNA delivery to cancer cells using an amphiphilic dendrimer equipped with a dual targeting peptide bearing an RGDK warhead. According to the molecular design, the amphiphilic dendrimer was expected to deliver siRNA effectively, while the aim of the targeting peptide was to home in on tumors via interaction of its warhead with integrin and the neuropilin-1 receptor on cancer cells. Coating the positively charged siRNA/dendrimer delivery complex with the negatively charged segment of the targeting peptide via electrostatic interactions led to small and stable nanoparticles which were able to protect siRNA from degradation while maintaining the accessibility of RGDK for targeting cancer cells and preserving the ability of the siRNA to escape from endosomes. The targeted system had enhanced siRNA delivery, stronger gene silencing, and more potent anticancer activity compared to nontargeted or covalent dendrimer-based systems. In addition, neither acute toxicity nor induced inflammation was observed. Consequently, this delivery system constitutes a promising nonviral vector for targeted delivery and can be further developed to provide RNAi-based personalized medicine against cancer. Our study also gives new perspectives on the use of nanotechnology based on self-assembling dendrimers in various biomedical applications.
Introduction
Results and Discussion
Formation of Small and Stable Peptide-Decorated siRNA/AD/E16G6RGDK Complexes
Accessible RGDK Targeting Moieties in the Stable Peptide-Coated Complexes
Enhanced Cell Uptake via Dual Receptor-Mediated Interactions Followed by Effective Endosomal Escape
Targeted Delivery Increases Gene Silencing and Anticancer Activity in Vitro
Targeted Delivery Enhances Gene Silencing and Antitumor Activity in Vivo
The Targeted Delivery System Is Safe and Devoid of Toxicity
Conclusions
Supporting Information
The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/jacs.8b10021.
Materials and methods as well as all experimental protocols for TEM, DLS, computational details, ITC, cell uptake and endosome release, siRNA delivery, gene silencing, anticancer activity and toxicity, etc. (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
Financial support from ARTP (L.P., X.L.), La Ligue Nationale Contre le Cancer (EL2016.LNCC/LPP) (L.P.), and the French National Research Agency under the frame of EuroNanoMed II (ANR-15-ENM2-0006-02, ANR-16-ENM2-0004-02) (L.P.), National Natural Science Foundation of China (No. 51773227, 81701815) (X.L.), Natural Science Foundation of Jiangsu Province (BK20170735) (X.L.), Recruitment Program for Youth Talents (X.L.), the Program for Jiangsu Province Innovative Research Talents (X.L.), the Program for Jiangsu Province Innovative Research Team (X.L.), the Jiangsu Specially-Appointed Professors Program (X.L.), the Six Talent Peaks Project of Jiangsu Province of China (X.L.), the State Key Laboratory of Natural Medicines at China Pharmaceutical University (SKLNMKF201703, SKLNMZZRC201804) (L.P., X.L.), the Italian Association for Cancer Research (AIRC, IG 17413 to S.P.), Beijing Institute of Technology Research Fund Program for Young Scholars (Y.H.), the Fundamental Research Funds for the Central Universities (Y.H.), the Hunan Provincial Natural Science Foundation of China (2018JJ1019) (Y.H.), and Huxiang Young Talent Program (Y.H.) is gratefully acknowledged. L.D. is supported by the China Scholarship Council and Y.J. by the Fondation de Recherche Médicale (SPF20160936294).
References
This article references 52 other publications.
- 1Fire, A.; Xu, S.; Montgomery, M.; Kostas, S.; Driver, S.; Mello, C. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature 1998, 391, 806– 811, DOI: 10.1038/35888Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXhtlCju74%253D&md5=7bd291ca75d455bbbe1dee41c3362208Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegansFire, Andrew; Xu, SiQun; Montgomery, Mary K.; Kostas, Steven A.; Driver, Samuel E.; Mello, Craig C.Nature (London) (1998), 391 (6669), 806-811CODEN: NATUAS; ISSN:0028-0836. (Macmillan Magazines)Exptl. introduction of RNA into cells can be used in certain biol. systems to interfere with the function of an endogenous gene. Such effects have been proposed to result from a simple antisense mechanism that depends on hybridization between the injected RNA and endogenous mRNA transcripts. RNA interference has been used in the nematode C. elegans to manipulate gene expression. Here we investigate the requirements for structure and delivery of the interfering RNA. To our surprise, we found that double-stranded RNA was substantially more effective at producing interference than was either strand individually. After injection into adult animals, purified single strands had at most a modest effect, whereas double-stranded mixts. caused potent and specific interference. The effects of this interference were evident in both the injected animals and their progeny. Only a few mols. of injected double-stranded RNA were required per affected cell, arguing against stoichiometric interference with endogenous mRNA and suggesting that there could be a catalytic or amplification component in the interference process.
- 2Hannon, G. RNA interference. Nature 2002, 418, 244– 251, DOI: 10.1038/418244aGoogle Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XltFGmu7Y%253D&md5=8e218d4b8d0880adfea123dd2e77df11RNA interferenceHannon, Gregory J.Nature (London, United Kingdom) (2002), 418 (6894), 244-251CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)A review. A conserved biol. response to double-stranded RNA, known variously as RNA interference (RNAi) or post-transcriptional gene silencing, mediates resistance to both endogenous parasitic and exogenous pathogenic nucleic acids, and regulates the expression of protein-coding genes. RNAi has been cultivated as a means to manipulate gene expression exptl. and to probe gene function on a whole-genome scale.
- 3Castanotto, D.; Rossi, J. The promises and pitfalls of RNA-interference-based therapeutics. Nature 2009, 457, 426– 433, DOI: 10.1038/nature07758Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXotlOgsA%253D%253D&md5=0574a639adbec5f45553d2248b807fceThe promises and pitfalls of RNA-interference-based therapeuticsCastanotto, Daniela; Rossi, John J.Nature (London, United Kingdom) (2009), 457 (7228), 426-433CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)A review. The discovery that gene expression can be controlled by the Watson-Crick base-pairing of small RNAs with mRNAs contg. complementary sequence - a process known as RNA interference - has markedly advanced our understanding of eukaryotic gene regulation and function. The ability of short RNA sequences to modulate gene expression has provided a powerful tool with which to study gene function and is set to revolutionize the treatment of disease. Remarkably, despite being just one decade from its discovery, the phenomenon is already being used therapeutically in human clin. trials, and biotechnol. companies that focus on RNA-interference-based therapeutics are already publicly traded.
- 4Haussecker, D.; Kay, M. RNA interference. Drugging RNAi. Science 2015, 347, 1069– 1070, DOI: 10.1126/science.1252967Google ScholarThere is no corresponding record for this reference.
- 5Wittrup, A.; Lieberman, J. Knocking down disease: a progress report on siRNA therapeutics. Nat. Rev. Genet. 2015, 16, 543– 552, DOI: 10.1038/nrg3978Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtlKnsLnF&md5=6475cb83ab2f8ab205e93d26b89952acKnocking down disease: a progress report on siRNA therapeuticsWittrup, Anders; Lieberman, JudyNature Reviews Genetics (2015), 16 (9), 543-552CODEN: NRGAAM; ISSN:1471-0056. (Nature Publishing Group)A review. Small interfering RNAs (siRNAs), which downregulate gene expression guided by sequence complementarity, can be used therapeutically to block the synthesis of disease-causing proteins. The main obstacle to siRNA drugs - their delivery into the target cell cytosol - has been overcome to allow suppression of liver gene expression. Here, we review the results of recent clin. trials of siRNA therapeutics, which show efficient and durable gene knockdown in the liver, with signs of promising clin. outcomes and little toxicity. We also discuss the barriers to more widespread applications that target tissues besides the liver and the most promising avenues to overcome them.
- 6Ledford, H. Gene-silencing technology gets first drug approval after 20-year wait. Nature 2018, 560, 291– 292, DOI: 10.1038/d41586-018-05867-7Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsFahtrvO&md5=39e63d6837dbb66e4d057a96d615fa84Gene-silencing technology gets first drug approval after 20-year waitLedford, HeidiNature (London, United Kingdom) (2018), 560 (7718), 291-292CODEN: NATUAS; ISSN:0028-0836. (Nature Research)The US Food and Drug Administration's decision breathes new life into RNA-interference therapies.
- 7Whitehead, K.; Langer, R.; Anderson, D. Knocking down barriers: advances in siRNA delivery. Nat. Rev. Drug Discovery 2009, 8, 129– 138, DOI: 10.1038/nrd2742Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtFOqt70%253D&md5=f3eecb4c59f7f3b233c3cce4823e7c72Knocking down barriers: advances in siRNA deliveryWhitehead, Kathryn A.; Langer, Robert; Anderson, Daniel G.Nature Reviews Drug Discovery (2009), 8 (2), 129-138CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. In the 10 years that have passed since the Nobel prize-winning discovery of RNA interference (RNAi), billions of dollars have been invested in the therapeutic application of gene silencing in humans. Today, there are promising data from ongoing clin. trials for the treatment of age-related macular degeneration and respiratory syncytial virus. Despite these early successes, however, the widespread use of RNAi therapeutics for disease prevention and treatment requires the development of clin. suitable, safe and effective drug delivery vehicles. Here, we provide an update on the progress of RNAi therapeutics and highlight novel synthetic materials for the encapsulation and intracellular delivery of nucleic acids.
- 8Kanasty, R.; Dorkin, J.; Vegas, A.; Anderson, D. Delivery materials for siRNA therapeutics. Nat. Mater. 2013, 12, 967– 977, DOI: 10.1038/nmat3765Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhs1Grt7rM&md5=fdc0612869582a97b13673c0ca9c0cf5Delivery materials for siRNA therapeuticsKanasty, Rosemary; Dorkin, Joseph Robert; Vegas, Arturo; Anderson, DanielNature Materials (2013), 12 (11), 967-977CODEN: NMAACR; ISSN:1476-1122. (Nature Publishing Group)A review. RNA interference (RNAi) has broad potential as a therapeutic to reversibly silence any gene. To achieve the clin. potential of RNAi, delivery materials are required to transport short interfering RNA (siRNA) to the site of action in the cells of target tissues. This Review provides an introduction to the biol. challenges that siRNA delivery materials aim to overcome, as well as a discussion of the way that the most effective and clin. advanced classes of siRNA delivery systems, including lipid nanoparticles and siRNA conjugates, are designed to surmount these challenges. The systems that we discuss are diverse in their approaches to the delivery problem, and provide valuable insight to guide the design of future siRNA delivery materials.
- 9Yin, H.; Kanasty, R.; Eltoukhy, A.; Vegas, A.; Dorkin, J.; Anderson, D. Non-viral vectors for gene-based therapy. Nat. Rev. Genet. 2014, 15, 541– 555, DOI: 10.1038/nrg3763Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtFGqsbfF&md5=eb763f2d3fe0b8cde33599f802f1db1aNon-viral vectors for gene-based therapyYin, Hao; Kanasty, Rosemary L.; Eltoukhy, Ahmed A.; Vegas, Arturo J.; Dorkin, J. Robert; Anderson, Daniel G.Nature Reviews Genetics (2014), 15 (8), 541-555CODEN: NRGAAM; ISSN:1471-0056. (Nature Publishing Group)Gene-based therapy is the intentional modulation of gene expression in specific cells to treat pathol. conditions. This modulation is accomplished by introducing exogenous nucleic acids such as DNA, mRNA, small interfering RNA (siRNA), microRNA (miRNA) or antisense oligonucleotides. Given the large size and the neg. charge of these macromols., their delivery is typically mediated by carriers or vectors. In this Review, we introduce the biol. barriers to gene delivery in vivo and discuss recent advances in material sciences, nanotechnol. and nucleic acid chem. that have yielded promising non-viral delivery systems, some of which are currently undergoing testing in clin. trials. The diversity of these systems highlights the recent progress of gene-based therapy using non-viral approaches.
- 10Juliano, R. The delivery of therapeutic oligonucleotides. Nucleic Acids Res. 2016, 44, 6518– 6548, DOI: 10.1093/nar/gkw236Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC28bgsFWluw%253D%253D&md5=22ea7e38834cf7494c35687df25f23a8The delivery of therapeutic oligonucleotidesJuliano Rudolph LNucleic acids research (2016), 44 (14), 6518-48 ISSN:.The oligonucleotide therapeutics field has seen remarkable progress over the last few years with the approval of the first antisense drug and with promising developments in late stage clinical trials using siRNA or splice switching oligonucleotides. However, effective delivery of oligonucleotides to their intracellular sites of action remains a major issue. This review will describe the biological basis of oligonucleotide delivery including the nature of various tissue barriers and the mechanisms of cellular uptake and intracellular trafficking of oligonucleotides. It will then examine a variety of current approaches for enhancing the delivery of oligonucleotides. This includes molecular scale targeted ligand-oligonucleotide conjugates, lipid- and polymer-based nanoparticles, antibody conjugates and small molecules that improve oligonucleotide delivery. The merits and liabilities of these approaches will be discussed in the context of the underlying basic biology.
- 11Pack, D.; Hoffman, A.; Pun, S.; Stayton, P. Design and development of polymers for gene delivery. Nat. Rev. Drug Discovery 2005, 4, 581– 593, DOI: 10.1038/nrd1775Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXmsFGjtbo%253D&md5=ed3dcd52a8b297ee3d0c1cbb7d5ae43bDesign and development of polymers for gene deliveryPack, Daniel W.; Hoffman, Allan S.; Pun, Suzie; Stayton, Patrick S.Nature Reviews Drug Discovery (2005), 4 (7), 581-593CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. The lack of safe and efficient gene-delivery methods is a limiting obstacle to human gene therapy. Synthetic gene-delivery agents, although safer than recombinant viruses, generally do not possess the required efficacy. In recent years, a variety of effective polymers have been designed specifically for gene delivery, and much has been learned about their structure-function relationships. With the growing understanding of polymer gene-delivery mechanisms and continued efforts of creative polymer chemists, it is likely that polymer-based gene-delivery systems will become an important tool for human gene therapy.
- 12Lachelt, U.; Wagner, E. Nucleic acid therapeutics using polyplexes: a journey of 50 years (and beyond). Chem. Rev. 2015, 115, 11043– 11078, DOI: 10.1021/cr5006793Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2MjktVagtg%253D%253D&md5=f317ecbf3554fbcb4ebae59ef0d3d48fNucleic Acid Therapeutics Using Polyplexes: A Journey of 50 Years (and Beyond)Lachelt Ulrich; Wagner Ernst; Lachelt Ulrich; Wagner ErnstChemical reviews (2015), 115 (19), 11043-78 ISSN:.There is no expanded citation for this reference.
- 13Tseng, Y.; Mozumdar, S.; Huang, L. Lipid-based systemic delivery of siRNA. Adv. Drug Delivery Rev. 2009, 61, 721– 731, DOI: 10.1016/j.addr.2009.03.003Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXmsFyns7Y%253D&md5=535a7456fd5f7ab5503a19dfa7a66f7dLipid-based systemic delivery of siRNATseng, Yu-Cheng; Mozumdar, Subho; Huang, LeafAdvanced Drug Delivery Reviews (2009), 61 (9), 721-731CODEN: ADDREP; ISSN:0169-409X. (Elsevier B.V.)A review. RNAi technol. has brought a new category of treatments for various diseases including genetic diseases, viral diseases, and cancer. Despite the great versatility of RNAi that can down regulate almost any protein in the cells, the delicate and precise machinery used for silencing is the same. The major challenge indeed for RNAi-based therapy is the delivery system. In this review, we start with the uniqueness and mechanism of RNAi machinery and the utility of RNAi in therapeutics. Then we discuss the challenges in systemic siRNA delivery by dividing them into two categories-kinetic and phys. barriers. At the end, we discuss different strategies to overcome these barriers, esp. focusing on the step of endosome escape. Toxicity issues and current successful examples for lipid-based delivery are also included in the review.
- 14Wagner, E. Polymers for siRNA delivery: inspired by viruses to be targeted, dynamic, and precise. Acc. Chem. Res. 2012, 45, 1005– 1013, DOI: 10.1021/ar2002232Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhs1GltrfO&md5=7b0c369312a3d10cb5a8f7fe5dd21f82Polymers for siRNA Delivery: Inspired by Viruses to be Targeted, Dynamic, and PreciseWagner, ErnstAccounts of Chemical Research (2012), 45 (7), 1005-1013CODEN: ACHRE4; ISSN:0001-4842. (American Chemical Society)A review. Synthetic small interfering RNA (siRNA) presents an exciting novel medical opportunity. Although researchers agree that siRNA could have a great therapeutic impact, the required extracellular and intracellular delivery of these mols. into the disease-assocd. target cells presents the primary roadblock for the broader translation of these mols. into medicines. Thus, the design of adequate delivery technologies has utmost importance. Viruses are natural masterpieces of nucleic acid delivery and present chemists and drug delivery experts with a template for the design of artificial carriers for synthetic nucleic acids such as siRNA. They have been developed into gene vectors and have provided convincing successes in gene therapy. Optimized by biol. evolution, viruses are programmed to be dynamic and bioresponsive as they enter living cells, and they carry out their functions in a precisely defined sequence. However, because they are synthesized within living cells and with naturally available nucleotides and amino acids, the chem. of viruses is limited. With the use of diverse synthetic mols. and macromols., chemists can provide delivery solns. beyond the scope of the natural evolution of viruses. This Account describes the design and synthesis of "synthetic siRNA viruses." These structures contain elements that mimic the delivery functions of viral particles and surface domains that shield against undesired biol. interactions and enable specific host cell receptor binding through the presentation of multiple targeting ligands. For example, cationic polymers can reversibly package one or more siRNA mols. into nanoparticle cores to protect them against a degradative bioenvironment. After internalization by receptor-mediated endocytosis into the acidifying endosomes of cells, synthetic siRNA can escape from these vesicles through the activation of membrane-disruption domains as viruses do and reach the cytoplasm, the location of RNA interference. This multistep task presents an attractive challenge for chemists. Similar to the design of prodrugs, the functional domains of these systems have to be activated in a dynamic mode, triggered by conformational changes or bond cleavages in the relevant microenvironment such as the acidic endosome or disulfide-reducing cytoplasm. These chem. analogs of viral domains are often synthetically simpler and more easily accessible mols. than viral proteins. Their precise assembly into multifunctional macromol. and supramol. structures is facilitated by improved anal. techniques, precise orthogonal conjugation chemistries, and sequence-defined polymer syntheses. The chem. evolution of microdomains using chem. libraries and macromol. and supramol. evolution could provide key strategies for optimizing siRNA carriers to selected medical indications.
- 15Wittrup, A.; Ai, A.; Liu, X.; Hamar, P.; Trifonova, R.; Charisse, K.; Manoharan, M.; Kirchhausen, T.; Lieberman, J. Visualizing lipid-formulated siRNA release from endosomes and target gene knockdown. Nat. Biotechnol. 2015, 33, 870– 876, DOI: 10.1038/nbt.3298Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtF2murjF&md5=1e896c5372fff738d373ef1deb2ac412Visualizing lipid-formulated siRNA release from endosomes and target gene knockdownWittrup, Anders; Ai, Angela; Liu, Xing; Hamar, Peter; Trifonova, Radiana; Charisse, Klaus; Manoharan, Muthiah; Kirchhausen, Tomas; Lieberman, JudyNature Biotechnology (2015), 33 (8), 870-876CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)A central hurdle in developing small interfering RNAs (siRNAs) as therapeutics is the inefficiency of their delivery across the plasma and endosomal membranes to the cytosol, where they interact with the RNA interference machinery. With the aim of improving endosomal release, a poorly understood and inefficient process, we studied the uptake and cytosolic release of siRNAs, formulated in lipoplexes or lipid nanoparticles, by live-cell imaging and correlated it with knockdown of a target GFP reporter. siRNA release occurred invariably from maturing endosomes within ∼5-15 min of endocytosis. Cytosolic galectins immediately recognized the damaged endosome and targeted it for autophagy. However, inhibiting autophagy did not enhance cytosolic siRNA release. Gene knockdown occurred within a few hours of release and required <2,000 copies of cytosolic siRNAs. The ability to detect cytosolic release of siRNAs and understand how it is regulated will facilitate the development of rational strategies for improving the cytosolic delivery of candidate drugs.
- 16Lee, C.; MacKay, J.; Frechet, J.; Szoka, F. Designing dendrimers for biological applications. Nat. Biotechnol. 2005, 23, 1517– 1526, DOI: 10.1038/nbt1171Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXht1yisrnE&md5=d1306ae17979250c01f2fb51fa76a080Designing dendrimers for biological applicationsLee, Cameron C.; MacKay, John A.; Frechet, Jean M. J.; Szoka, Francis C.Nature Biotechnology (2005), 23 (12), 1517-1526CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)A review. Dendrimers are branched, synthetic polymers with layered architectures that show promise in several biomedical applications. By regulating dendrimer synthesis, it is possible to precisely manipulate both their mol. wt. and chem. compn., thereby allowing predictable tuning of their biocompatibility and pharmacokinetics. Advances in our understanding of the role of mol. wt. and architecture on the in vivo behavior of dendrimers, together with recent progress in the design of biodegradable chemistries, has enabled the application of these branched polymers as anti-viral drugs, tissue repair scaffolds, targeted carriers of chemotherapeutics and optical oxygen sensors. Before such products can reach the market, however, the field must not only address the cost of manuf. and quality control of pharmaceutical-grade materials, but also assess the long-term human and environmental health consequences of dendrimer exposure in vivo.
- 17Khandare, J.; Calderon, M.; Dagia, N.; Haag, R. Multifunctional dendritic polymers in nanomedicine: opportunities and challenges. Chem. Soc. Rev. 2012, 41, 2824– 2848, DOI: 10.1039/C1CS15242DGoogle Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xjs1CksLg%253D&md5=2096b686d6657325955ec41bbbc006f2Multifunctional dendritic polymers in nanomedicine: opportunities and challengesKhandare, Jayant; Calderon, Marcelo; Dagia, Nilesh M.; Haag, RainerChemical Society Reviews (2012), 41 (7), 2824-2848CODEN: CSRVBR; ISSN:0306-0012. (Royal Society of Chemistry)A review. Nanotechnol. has resulted in materials that have greatly improved the effectiveness of drug delivery because of their ability to control matter on the nanoscale. Advanced forms of nanomedicine have been synthesized for better pharmacokinetics to obtain higher efficacy, less systemic toxicity, and better targeting. These criteria have long been the goal in nanomedicine, in particular, for systemic applications in oncol. disorders. Now, the "holy grail" in nanomedicine is to design and synthesize new advanced macromol. nanocarriers and to translate them from lab to clinic. This review describes the current and future perspectives of nanomedicine with particular emphasis on the clin. targets in cancer and inflammation. The advanced forms of liposomes and polyethylene glycol (PEG) based nanocarriers, as well as dendritic polymer conjugates will be discussed with particular attention paid to designs, synthetic strategies, and chem. pathways. In this crit. review, we also report on the current status and perspective of dendritic polymer nanoconjugate platforms (e.g. polyamidoamine dendrimers and dendritic polyglycerols) for cellular localization and targeting of specific tissues (192 refs.).
- 18Liu, X.; Rocchi, P.; Peng, L. Dendrimers as non-viral vectors for siRNA delivery. New J. Chem. 2012, 36, 256– 263, DOI: 10.1039/C1NJ20408DGoogle ScholarThere is no corresponding record for this reference.
- 19Liu, X.; Liu, C.; Catapano, C.; Peng, L.; Zhou, J.; Rocchi, P. Structurally flexible triethanolamine-core poly(amidoamine) dendrimers as effective nanovectors to deliver RNAi-based therapeutics. Biotechnol. Adv. 2014, 32, 844– 852, DOI: 10.1016/j.biotechadv.2013.08.001Google ScholarThere is no corresponding record for this reference.
- 20Reebye, V.; Sætrom, P.; Mintz, P.; Huang, K.; Swiderski, P.; Peng, L.; Liu, C.; Liu, X.; Lindkaer-Jensen, S.; Zacharoulis, D. Novel RNA oligonucleotide improves liver function and inhibits liver carcinogenesis in vivo. Hepatology 2014, 59, 216– 227, DOI: 10.1002/hep.26669Google ScholarThere is no corresponding record for this reference.
- 21Yu, T.; Liu, X.; Bolcato-Bellemin, A.; Wang, Y.; Liu, C.; Erbacher, P.; Qu, F.; Rocchi, P.; Behr, J.; Peng, L. An amphiphilic dendrimer for effective delivery of small interfering RNA and gene silencing in vitro and in vivo. Angew. Chem., Int. Ed. 2012, 51, 8478– 8484, DOI: 10.1002/anie.201203920Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtVyjs7rO&md5=176386f529e80f5778008b8aba3c2714An Amphiphilic Dendrimer for Effective Delivery of Small Interfering RNA and Gene Silencing In Vitro and In VivoYu, Tianzhu; Liu, Xiaoxuan; Bolcato-Bellemin, Anne-Laure; Wang, Yang; Liu, Cheng; Erbacher, Patrick; Qu, Fanqi; Rocchi, Palma; Behr, Jean-Paul; Peng, LingAngewandte Chemie, International Edition (2012), 51 (34), 8478-8484, S8478/1-S8478/33CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)An amphiphilic dendrimer bearing a hydrophobic alkyl chain and hydrophilic poly(amidoamine) dendrons is able to combine the advantageous features of lipid and dendrimers vectors to deliver a heat shock protein 27 siRNA and produce potent gene silencing and anticancer activity in vitro and in vivo in a prostate cancer model. This dendrimer can be used alternatively for treating various diseases.
- 22Liu, X.; Zhou, J.; Yu, T.; Chen, C.; Cheng, Q.; Sengupta, K.; Huang, Y.; Li, H.; Liu, C.; Wang, Y.; Posocco, P.; Wang, M.; Cui, Q.; Giorgio, S.; Fermeglia, M.; Qu, F.; Pricl, S.; Shi, Y.; Liang, Z.; Rocchi, P.; Rossi, J.; Peng, L. Adaptive amphiphilic dendrimer-based nanoassemblies as robust and versatile siRNA delivery systems. Angew. Chem., Int. Ed. 2014, 53, 11822– 11827, DOI: 10.1002/anie.201406764Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhsFCisrvF&md5=5cb14bd9e508eb76c4b008b6752ed806Adaptive Amphiphilic Dendrimer-Based Nanoassemblies as Robust and Versatile siRNA Delivery SystemsLiu, Xiaoxuan; Zhou, Jiehua; Yu, Tianzhu; Chen, Chao; Cheng, Qiang; Sengupta, Kheya; Huang, Yuanyu; Li, Haitang; Liu, Cheng; Wang, Yang; Posocco, Paola; Wang, Menghua; Cui, Qi; Giorgio, Suzanne; Fermeglia, Maurizio; Qu, Fanqi; Pricl, Sabrina; Shi, Yanhong; Liang, Zicai; Rocchi, Palma; Rossi, John J.; Peng, LingAngewandte Chemie, International Edition (2014), 53 (44), 11822-11827CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)SiRNA delivery remains a major challenge in RNAi-based therapy. Here, we report for the first time that an amphiphilic dendrimer is able to self-assemble into adaptive supramol. assemblies upon interaction with siRNA, and effectively delivers siRNAs to various cell lines, including human primary and stem cells, thereby outperforming the currently available nonviral vectors. In addn., this amphiphilic dendrimer is able to harness the advantageous features of both polymer and lipid vectors and hence promotes effective siRNA delivery. Our study demonstrates for the first time that dendrimer-based adaptive supramol. assemblies represent novel and versatile means for functional siRNA delivery, heralding a new age of dendrimer-based self-assembled drug delivery in biomedical applications.
- 23Chen, C.; Posocco, P.; Liu, X.; Cheng, Q.; Laurini, E.; Zhou, J.; Liu, C.; Wang, Y.; Tang, J.; Col, V.; Yu, T.; Giorgio, S.; Fermeglia, M.; Qu, F.; Liang, Z.; Rossi, J.; Liu, M.; Rocchi, P.; Pricl, S.; Peng, L. siRNA delivery: mastering dendrimer self-assembly for efficient siRNA delivery: from conceptual design to in vivo efficient gene silencing. Small 2016, 12, 3667– 3676, DOI: 10.1002/smll.201503866Google ScholarThere is no corresponding record for this reference.
- 24Liu, X.; Wang, Y.; Chen, C.; Tintaru, A.; Cao, Y.; Liu, J.; Ziarelli, F.; Tang, J.; Guo, H.; Rosas, R.; Giorgio, S.; Charle, L.; Rocchi, P.; Peng, L. A fluorinated bola-amphiphilic dendrimer for on-demand delivery of siRNA, via specific response to reactive oxygen species. Adv. Funct. Mater. 2016, 26, 8594– 8603, DOI: 10.1002/adfm.201604192Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhslGitLjE&md5=931ea4546e4a792fc32c41dd25b69a8aA Fluorinated Bola-Amphiphilic Dendrimer for On-Demand Delivery of siRNA, via Specific Response to Reactive Oxygen SpeciesLiu, Xiaoxuan; Wang, Yang; Chen, Chao; Tintaru, Aura; Cao, Yu; Liu, Juan; Ziarelli, Fabio; Tang, Jingjie; Guo, Hongbo; Rosas, Roseline; Giorgio, Suzanne; Charles, Laurence; Rocchi, Palma; Peng, LingAdvanced Functional Materials (2016), 26 (47), 8594-8603CODEN: AFMDC6; ISSN:1616-301X. (Wiley-VCH Verlag GmbH & Co. KGaA)Functional materials capable of responding to stimuli intrinsic to diseases are extremely important for specific drug delivery at the disease site. However, developing on-demand stimulus-responsive vectors for targeted delivery is highly challenging. Here, a stimulus-responsive fluorinated bola-amphiphilic dendrimer is reported for on-demand delivery of small interfering RNA (siRNA) in response to the characteristic high level of reactive oxygen species (ROS) in cancer cells. This dendrimer bears a ROS-sensitive thioacetal in the hydrophobic core and pos. charged poly(amidoamine) dendrons at the terminals, capable of interacting and compacting the neg. charged siRNA into nanoparticles to protect the siRNA and promote cellular uptake. The ROS-sensitive feature of this dendrimer boosts specific and efficient disassembly of the siRNA/vector complexes in ROS-rich cancer cells for effective siRNA delivery and gene silencing. Moreover, the fluorine tags in the vector enable 19F-NMR anal. of the ROS-responsive delivery process. In addn., this ingenious and distinct bola-amphiphilic dendrimer is also able to combine the advantageous delivery features of both lipid and dendrimer vectors. Therefore, it represents an innovative on-demand stimulus-responsive delivery platform.
- 25Percec, V.; Wilson, D.; Leowanawat, P.; Wilson, C.; Hughes, A.; Kaucher, M.; Hammer, D.; Levine, D.; Kim, A.; Bates, F. Self-assembly of Janus dendrimers into uniform dendrimersomes and other complex architectures. Science 2010, 328, 1009– 1014, DOI: 10.1126/science.1185547Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXmtFCnsLo%253D&md5=f27fa5e6ae26f177e85c36ca5e46047eSelf-Assembly of Janus Dendrimers into Uniform Dendrimersomes and Other Complex ArchitecturesPercec, Virgil; Wilson, Daniela A.; Leowanawat, Pawaret; Wilson, Christopher J.; Hughes, Andrew D.; Kaucher, Mark S.; Hammer, Daniel A.; Levine, Dalia H.; Kim, Anthony J.; Bates, Frank S.; Davis, Kevin P.; Lodge, Timothy P.; Klein, Michael L.; De Vane, Russell H.; Aqad, Emad; Rosen, Brad M.; Argintaru, Andreea O.; Sienkowska, Monika J.; Rissanen, Kari; Nummelin, Sami; Ropponen, JarmoScience (Washington, DC, United States) (2010), 328 (5981), 1009-1014CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Self-assembled nanostructures obtained from natural and synthetic amphiphiles serve as mimics of biol. membranes and enable the delivery of drugs, proteins, genes, and imaging agents. Yet the precise mol. arrangements demanded by these functions are difficult to achieve. Libraries of amphiphilic Janus dendrimers, prepd. by facile coupling of tailored hydrophilic and hydrophobic branched segments, have been screened by cryogenic transmission electron microscopy, revealing a rich palette of morphologies in water, including vesicles, denoted dendrimersomes, cubosomes, disks, tubular vesicles, and helical ribbons. Dendrimersomes marry the stability and mech. strength obtainable from polymersomes with the biol. function of stabilized phospholipid liposomes, plus superior uniformity of size, ease of formation, and chem. functionalization. This modular synthesis strategy provides access to systematic tuning of mol. structure and of self-assembled architecture.
- 26Sherman, S.; Xiao, Q.; Percec, V. Mimicking complex biological membranes and their programmable glycan ligands with dendrimersomes and glycodendrimersomes. Chem. Rev. 2017, 117, 6538– 6631, DOI: 10.1021/acs.chemrev.7b00097Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXmtVyqur8%253D&md5=7448c0318ceb9b46a0b12963822a166bMimicking Complex Biological Membranes and Their Programmable Glycan Ligands with Dendrimersomes and GlycodendrimersomesSherman, Samuel E.; Xiao, Qi; Percec, VirgilChemical Reviews (Washington, DC, United States) (2017), 117 (9), 6538-6631CODEN: CHREAY; ISSN:0009-2665. (American Chemical Society)A review. Synthetic vesicles have been assembled and co-assembled from phospholipids, their modified versions, and other single amphiphiles into liposomes, and from block copolymers into polymersomes. Their time-consuming synthesis and prepn. as stable, monodisperse, and biocompatible liposomes and polymersomes called for the elaboration of new synthetic methodologies. Amphiphilic Janus dendrimers (JDs) and glycodendrimers (JGDs) represent the most recent self-assembling amphiphiles capable of forming monodisperse, stable, and multifunctional unilamellar and multilamellar onion-like vesicles denoted dendrimersomes (DSs) and glycodendrimersomes (GDSs), dendrimercubosomes (DCs), glycodendrimercubosomes (GDCs), and other complex architectures. Amphiphilic JDs consist of hydrophobic dendrons connected to hydrophilic dendrons and can be thought of as monodisperse oligomers of a single amphiphile. They can be functionalized with a variety of mols. such as dyes, and, in the case of JGDs, with carbohydrates. Their iterative modular synthesis provides efficient access to sequence control at the mol. level, resulting in topologies with specific epitope sequence and d. DSs, GDSs, and other architectures from JDs and JGDs serve as powerful tools for mimicking biol. membranes and for biomedical applications such as targeted drug and gene delivery and theranostics. This Review covers all aspects of the synthesis of JDs and JGDs and their biol. activity and applications after assembly in aq. media.
- 27Bertrand, N.; Wu, J.; Xu, X.; Kamaly, N.; Farokhzad, O. Cancer nanotechnology: the impact of passive and active targeting in the era of modern cancer biology. Adv. Drug Delivery Rev. 2014, 66, 2– 25, DOI: 10.1016/j.addr.2013.11.009Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXisV2hug%253D%253D&md5=0980da1a1b75cde64cc1cf82184e01b7Cancer nanotechnology: The impact of passive and active targeting in the era of modern cancer biologyBertrand, Nicolas; Wu, Jun; Xu, Xiaoyang; Kamaly, Nazila; Farokhzad, Omid C.Advanced Drug Delivery Reviews (2014), 66 (), 2-25CODEN: ADDREP; ISSN:0169-409X. (Elsevier B.V.)Cancer nanotherapeutics are progressing at a steady rate; research and development in the field has experienced an exponential growth since early 2000's. The path to the commercialization of oncol. drugs is long and carries significant risk; however, there is considerable excitement that nanoparticle technologies may contribute to the success of cancer drug development. The pace at which pharmaceutical companies have formed partnerships to use proprietary nanoparticle technologies has considerably accelerated. It is now recognized that by enhancing the efficacy and/or tolerability of new drug candidates, nanotechnol. can meaningfully contribute to create differentiated products and improve clin. outcome. This review describes the lessons learned since the commercialization of the first-generation nanomedicines including DOXIL and Abraxane. It explores our current understanding of targeted and non-targeted nanoparticles that are under various stages of development, including BIND-014 and MM-398. It highlights the opportunities and challenges faced by nanomedicines in contemporary oncol., where personalized medicine is increasingly the mainstay of cancer therapy. We revisit the fundamental concepts of enhanced permeability and retention effect (EPR) and explore the mechanisms proposed to enhance preferential "retention" in the tumor, whether using active targeting of nanoparticles, binding of drugs to their tumoral targets or the presence of tumor assocd. macrophages. The overall objective of this review is to enhance our understanding in the design and development of therapeutic nanoparticles for treatment of cancers.
- 28Dai, L.; Liu, J.; Luo, Z.; Li, M.; Cai, K. Tumor therapy: targeted drug delivery system. J. Mater. Chem. B 2016, 4, 6758– 6772, DOI: 10.1039/C6TB01743FGoogle Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsVWktr3F&md5=43534ec2f361c7181059c074132608d4Tumor therapy: targeted drug delivery systemsDai, Liangliang; Liu, Junjie; Luo, Zhong; Li, Menghuan; Cai, KaiyongJournal of Materials Chemistry B: Materials for Biology and Medicine (2016), 4 (42), 6758-6772CODEN: JMCBDV; ISSN:2050-7518. (Royal Society of Chemistry)Recently, targeted drug delivery systems (TDDSs) have been extensively studied as a promising therapeutic for tumor therapy. In this review, we investigate the typical targeting mechanisms of TDDSs, covering both passively and actively targeting DDSs for tumor therapy. We highlight the popular active targeting strategies for different sites of action, including tumor cytomembrane or various organelles. Finally, we present some recent representative TDDSs that are under testing in preclin./clin. trials and have shown excellent clin. potential as the alternate treatment strategy for tumor therapy. Although TDDSs are proving to be promising therapeutic nanoplatforms for tumor therapy, extended investigations should be considered in the landscape for highly efficient tumor therapy with good biosafety.
- 29Davis, M.; Zuckerman, J.; Choi, C.; Seligson, D.; Tolcher, A.; Alabi, C.; Yen, Y.; Heidel, J.; Ribas, A. Evidence of RNAi in humans from systemically administered siRNA via targeted nanoparticles. Nature 2010, 464, 1067– 1070, DOI: 10.1038/nature08956Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXjs1ehur8%253D&md5=b49a61e3acc1245274c2314fb6bcc99aEvidence of RNAi in humans from systemically administered siRNA via targeted nanoparticlesDavis, Mark E.; Zuckerman, Jonathan E.; Choi, Chung-Hang J.; Seligson, David; Tolcher, Anthony; Alabi, Christopher A.; Yen, Yun; Heidel, Jeremy D.; Ribas, AntoniNature (London, United Kingdom) (2010), 464 (7291), 1067-1070CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Therapeutics that are designed to engage RNA interference (RNAi) pathways have the potential to provide new, major ways of imparting therapy to patients. Long, double-stranded RNAs were first shown to mediate RNAi in Caenorhabditis elegans, and the potential use of RNAi for human therapy has been demonstrated by the finding that small interfering RNAs (siRNAs; approx. 21-base-pair double-stranded RNA) can elicit RNAi in mammalian cells without producing an interferon response. We are at present conducting the first in-human phase I clin. trial involving the systemic administration of siRNA to patients with solid cancers using a targeted, nanoparticle delivery system. Here we provide evidence of inducing an RNAi mechanism of action in a human from the delivered siRNA. Tumor biopsies from melanoma patients obtained after treatment show the presence of intracellularly localized nanoparticles in amts. that correlate with dose levels of the nanoparticles administered (this is, to our knowledge, a first for systemically delivered nanoparticles of any kind). Furthermore, a redn. was found in both the specific mRNA (M2 subunit of ribonucleotide reductase (RRM2)) and the protein (RRM2) levels when compared to pre-dosing tissue. Most notably, we detect the presence of an mRNA fragment that demonstrates that siRNA-mediated mRNA cleavage occurs specifically at the site predicted for an RNAi mechanism from a patient who received the highest dose of the nanoparticles. Together, these data demonstrate that siRNA administered systemically to a human can produce a specific gene inhibition (redn. in mRNA and protein) by an RNAi mechanism of action.
- 30Sugahara, K.; Teesalu, T.; Karmali, P.; Kotamraju, V.; Agemy, L.; Greenwald, D.; Ruoslahti, E. Co-administration of a tumor-penetrating peptide enhances the efficacy of cancer drugs. Science 2010, 328, 1031– 1035, DOI: 10.1126/science.1183057Google ScholarThere is no corresponding record for this reference.
- 31Wong, P.; Tang, K.; Coulter, A.; Tan, S.; Baker, J.; Choi, S. Multivalent dendrimer vectors with DNA intercalation motifs for gene delivery. Biomacromolecules 2014, 15, 4134– 4415, DOI: 10.1021/bm501169sGoogle Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhs1yisr7K&md5=8c21541c6d33bbcad02f41564d5fe0d6Multivalent Dendrimer Vectors with DNA Intercalation Motifs for Gene DeliveryWong, Pamela T.; Tang, Kenny; Coulter, Alexa; Tang, Shengzhuang; Baker, James R.; Choi, Seok KiBiomacromolecules (2014), 15 (11), 4134-4145CODEN: BOMAF6; ISSN:1525-7797. (American Chemical Society)Poly(amido amine) (PAMAM) dendrimers constitute an important class of nonviral, cationic vectors in gene delivery. Here we report on a new concept for dendrimer vector design based on the incorporation of dual binding motifs: DNA intercalation, and receptor recognition for targeted delivery. We prepd. a series of dendrimer conjugates derived from a fifth generation (G5) PAMAM dendrimer, each conjugated with multiple folate (FA) or riboflavin (RF) ligands for cell receptor targeting, and with 3,8-diamino-6-phenylphenanthridinium ("DAPP")-derived ligands for anchoring a DNA payload. Polyplexes of each dendrimer with calf thymus dsDNA were made and characterized by surface plasmon resonance (SPR) spectroscopy, dynamic light scattering (DLS) and zeta potential measurement. These studies provided evidence supporting polyplex formation based on the observation of tight DNA-dendrimer adhesion, and changes in particle size and surface charge upon coincubation. Further SPR studies to investigate the adhesion of the polyplex to a model surface immobilized with folate binding protein (FBP), demonstrated that the DNA payload has only a minimal effect on the receptor binding activity of the polyplex: KD = 0.22 nM for G5(FA)(DAPP) vs. 0.98 nM for its polyplex. Finally, we performed in vitro transfection assays to det. the efficiency of conjugate mediated delivery of a luciferase-encoding plasmid into the KB cancer cell line and showed that RF-conjugated dendrimers were 1 to 2 orders of magnitude more effective in enhancing luciferase gene transfection than a plasmid only control. In summary, this study serves as a proof of concept for DNA-ligand intercalation as a motif in the design of multivalent dendrimer vectors for targeted gene delivery.
- 32Sugahara, K.; Teesalu, T.; Karmali, P.; Kotamraju, V.; Agemy, L.; Girard, O.; Hanahan, D.; Mattrey, R.; Ruoslahti, E. Tissue-penetrating delivery of compounds and nanoparticles into tumors. Cancer Cell 2009, 16, 510– 520, DOI: 10.1016/j.ccr.2009.10.013Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXht1eqsbY%253D&md5=f42c01e8e751526318903d28d70ce4c4Tissue-penetrating delivery of compounds and nanoparticles into tumorsSugahara, Kazuki N.; Teesalu, Tambet; Karmali, Priya Prakash; Kotamraju, Venkata Ramana; Agemy, Lilach; Girard, Olivier M.; Hanahan, Douglas; Mattrey, Robert F.; Ruoslahti, ErkkiCancer Cell (2009), 16 (6), 510-520CODEN: CCAECI; ISSN:1535-6108. (Cell Press)Poor penetration of drugs into tumors is a major obstacle in tumor treatment. We describe a strategy for peptide-mediated delivery of compds. deep into the tumor parenchyma that uses a tumor-homing peptide, iRGD (CRGDK/RGPD/EC). I.v. injected compds. coupled to iRGD bound to tumor vessels and spread into the extravascular tumor parenchyma, whereas conventional RGD peptides only delivered the cargo to the blood vessels. IRGD homes to tumors through a three-step process: the RGD motif mediates binding to αv integrins on tumor endothelium and a proteolytic cleavage then exposes a binding motif for neuropilin-1, which mediates penetration into tissue and cells. Conjugation to iRGD significantly improved the sensitivity of tumor-imaging agents and enhanced the activity of an antitumor drug.
- 33Desgrosellier, J.; Cheresh, D. Integrins in cancer: biological implications and therapeutic opportunities. Nat. Rev. Cancer 2010, 10, 9– 22, DOI: 10.1038/nrc2748Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhsF2hsLzI&md5=43d0b526ca38021e780aba991a58451bIntegrins in cancer: biological implications and therapeutic opportunitiesDesgrosellier, Jay S.; Cheresh, David A.Nature Reviews Cancer (2010), 10 (1), 9-22CODEN: NRCAC4; ISSN:1474-175X. (Nature Publishing Group)A review. The integrin family of cell adhesion receptors regulates a diverse array of cellular functions crucial to the initiation, progression and metastasis of solid tumors. The importance of integrins in several cell types that affect tumor progression has made them an appealing target for cancer therapy. Integrin antagonists, including the αvβ3 and αvβ5 inhibitor cilengitide, have shown encouraging activity in Phase II clin. trials and cilengitide is currently being tested in a Phase III trial in patients with glioblastoma. These exciting clin. developments emphasize the need to identify how integrin antagonists influence the tumor and its microenvironment.
- 34Marelli, U.; Rechenmacher, F.; Sobahi, T.; Mas-Moruno, C.; Kessler, H. Tumor targeting via integrin ligands. Front. Oncol. 2013, 3, 222– 233, DOI: 10.3389/fonc.2013.00222Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2cflt1Gktg%253D%253D&md5=b09d0b738b25909e8b68d42d90ad638eTumor Targeting via Integrin LigandsMarelli Udaya Kiran; Rechenmacher Florian; Sobahi Tariq Rashad Ali; Mas-Moruno Carlos; Kessler HorstFrontiers in oncology (2013), 3 (), 222 ISSN:2234-943X.Selective and targeted delivery of drugs to tumors is a major challenge for an effective cancer therapy and also to overcome the side-effects associated with current treatments. Overexpression of various receptors on tumor cells is a characteristic structural and biochemical aspect of tumors and distinguishes them from physiologically normal cells. This abnormal feature is therefore suitable for selectively directing anticancer molecules to tumors by using ligands that can preferentially recognize such receptors. Several subtypes of integrin receptors that are crucial for cell adhesion, cell signaling, cell viability, and motility have been shown to have an upregulated expression on cancer cells. Thus, ligands that recognize specific integrin subtypes represent excellent candidates to be conjugated to drugs or drug carrier systems and be targeted to tumors. In this regard, integrins recognizing the RGD cell adhesive sequence have been extensively targeted for tumor-specific drug delivery. Here we review key recent examples on the presentation of RGD-based integrin ligands by means of distinct drug-delivery systems, and discuss the prospects of such therapies to specifically target tumor cells.
- 35Teesalu, T.; Sugahara, K.; Kotamraju, V.; Ruoslahti, E. C-end rule peptides mediate neuropilin-1-dependent cell, vascular, and tissue penetration. Proc. Natl. Acad. Sci. U. S. A. 2009, 106, 16157– 16162, DOI: 10.1073/pnas.0908201106Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXht1OjsbbI&md5=3eb210213cfccc091c4aba949d437b42C-end rule peptides mediate neuropilin-1-dependent cell, vascular, and tissue penetrationTeesalu, Tambet; Sugahara, Kazuki N.; Kotamraju, Venkata Ramana; Erkki RuoslahtiProceedings of the National Academy of Sciences of the United States of America (2009), 106 (38), 16157-16162, S16157/1-S16157/15CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Screening of phage libraries expressing random peptides for binding to prostate cancer cells primarily yielded peptides that had a C-terminal arginine (or rarely lysine) residue, usually in a consensus context R/KXXR/K. Phage expressing these sequences and synthetic nanoparticles coated with them bound to and were internalized into cells. The C-terminal arginine (or lysine) was essential to the activity; adding another amino acid, or even blocking the free carboxyl group of this arginine residue by amidation, eliminated the binding and internalizing activity. An internal R/KXXR/K can be exposed and switched on by a cleavage by a protease. The strict requirement for C-terminal exposure of the motif prompted the authors to term the phenomenon the C-end rule (CendR). Affinity chromatog. showed that the CendR peptides bind to neuropilin-1 (NRP-1) on the target cells. NRP-1 is a cell-surface receptor that plays an essential role in angiogenesis, regulation of vascular permeability, and the development of the nervous system. VEGF-A165 and other ligands of NRP-1 possess a C-terminal CendR sequence that interacts with the b1 domain of NRP-1 and causes cellular internalization and vascular leakage. The authors' CendR peptides have similar effects, particularly when made multivalent through coupling to a particle. The authors also noted a unique and important activity of these peptides: penetration and transportation through tissues. The peptides were able to take payloads up to the nanoparticle size scale deep into extravascular tissue. The authors' observations have implications in drug delivery and penetration of tissue barriers and tumors.
- 36Liu, X.; Liu, C.; Chen, C.; Bentobji, M.; Cheillan, F.; Piana, J.; Qu, F.; Rocchi, P.; Peng, L. Targeted delivery of Dicer-substrate siRNAs using a dual targeting peptide decorated dendrimer delivery system. Nanomedicine 2014, 10, 1627– 1636, DOI: 10.1016/j.nano.2014.05.008Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtFOgtL3K&md5=27ce93c828721bcad7351c4edde95339Targeted delivery of Dicer-substrate siRNAs using a dual targeting peptide decorated dendrimer delivery systemLiu, Xiaoxuan; Liu, Cheng; Chen, Chao; Bentobji, Melanie; Cheillan, Francine Azario; Piana, Jeanne Thomassin; Qu, Fanqi; Rocchi, Palma; Peng, LingNanomedicine (New York, NY, United States) (2014), 10 (8), 1627-1636CODEN: NANOBF; ISSN:1549-9634. (Elsevier)Small interfering RNAs (siRNA) are emerging as novel therapeutic agents, providing competent delivery systems that are available. Dendrimers, a special family of synthetic macromols., represent an exciting delivery platform by virtue of their well-defined dendritic structure and unique multivalency and cooperativity confined within a nanoscale vol. Here, we report a Dicer-substrate siRNA (dsiRNA) which, when delivered using a structurally flexible triethanolamine-core poly(amidoamine) dendrimer of generation 5 as the nanocarrier, gives rise to a much greater RNAi response than that produced with conventional siRNA. Further decoration of the dsiRNA/dendrimer complexes with a dual targeting peptide simultaneously promoted cancer cell targeting through interacting with integrins and cell penetration via the interaction with neuropilin-1 receptors, which led to improved gene silencing and anticancer activity. Altogether, our results disclosed here open a new avenue for therapeutic implementation of RNAi using dendrimer nanovector based targeted delivery.
- 37Svenson, S. The dendrimer paradox--high medical expectations but poor clinical translation. Chem. Soc. Rev. 2015, 46, 4131– 4144, DOI: 10.1039/C5CS00288EGoogle ScholarThere is no corresponding record for this reference.
- 38Cao, Y.; Liu, X.; Peng, L. Molecular engineering of dendrimer nanovecters for siRNA delivery and gene silencing. Front. Chem. Sci. Eng. 2017, 11, 663– 675, DOI: 10.1007/s11705-017-1623-5Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXlvFyjsLs%253D&md5=72a76c778f9c6878d0f125e3b57bcb88Molecular engineering of dendrimer nanovectors for siRNA delivery and gene silencingCao, Yu; Liu, Xiaoxuan; Peng, LingFrontiers of Chemical Science and Engineering (2017), 11 (4), 663-675CODEN: FCSEA3; ISSN:2095-0187. (Springer)A review. Small interfering RNA (siRNA) therapeutics hold great promise to treat a variety of diseases, as long as they can be delivered safely and effectively into cells. Dendrimers are appealing vectors for siRNA delivery by virtue of their well-defined mol. architecture and multivalent cooperativity. However, the clin. translation of RNA therapeutics mediated by dendrimer delivery is hampered by the lack of dendrimers that are of high quality to meet good manufg. practice std. In this context, we have developed small amphiphilic dendrimers that self-assemble into supramol. structures, which mimic high-generation dendrimers synthesized with covalent construction, yet are easy to produce in large amt. and superior quality. This short review presents a brief introduction to RNAi therapeutics, the obstacles to their delivery and the advantages of dendrimer delivery vectors as well as our bio-inspired structurally flexible dendrimers for siRNA delivery. We then highlight our efforts in creating selfassembling amphiphilic dendrimers to construct supramol. dendrimer nanosystems for effective siRNA delivery as well as the related structural alterations to enhance delivery efficiency. The advent of self-assembling supramol. dendrimer nanovectors holds great promise and heralds a new era of dendrimer-mediated delivery of RNA therapeutics in biomedical applications.
- 39Zhou, J.; Liu, J.; Cheng, C.; Patel, T.; Weller, C.; Piepmeier, J.; Jiang, Z.; Saltzman, W. Biodegradable poly(amine-co-ester) terpolymers for targeted gene delivery. Nat. Mater. 2012, 11, 82– 90, DOI: 10.1038/nmat3187Google Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhsFGkurfN&md5=937db7f417aafb60789ec0f760f27d49Biodegradable poly(amine-co-ester) terpolymers for targeted gene deliveryZhou, Jiangbing; Liu, Jie; Cheng, Christopher J.; Patel, Toral R.; Weller, Caroline E.; Piepmeier, Joseph M.; Jiang, Zhaozhong; Saltzman, W. MarkNature Materials (2012), 11 (1), 82-90CODEN: NMAACR; ISSN:1476-1122. (Nature Publishing Group)Many synthetic polycationic vectors for non-viral gene delivery show high efficiency in vitro, but their usually excessive charge d. makes them toxic for in vivo applications. Here we describe the synthesis of a series of high mol. wt. terpolymers with low charge d., and show that they exhibit efficient gene delivery, some surpassing the efficiency of the com. transfection reagents Polyethylenimine and Lipofectamine 2000. The terpolymers were synthesized via enzyme-catalyzed copolymn. of lactone with dialkyl diester and amino diol, and their hydrophobicity adjusted by varying the lactone content and by selecting a lactone comonomer of specific ring size. Targeted delivery of the pro-apoptotic TRAIL gene to tumor xenografts by one of the terpolymers results in significant inhibition of tumor growth, with minimal toxicity both in vitro and in vivo. Our findings suggest that the gene delivery ability of the terpolymers stems from their high mol. wt. and increased hydrophobicity, which compensates for their low charge d.
- 40Grandclement, C.; Borg, C. Neuropilins: a new target for cancer therapy. Cancers 2011, 3, 1899– 1928, DOI: 10.3390/cancers3021899Google ScholarThere is no corresponding record for this reference.
- 41Siegel, R.; Miller, K.; Jemal, A. Cancer Statistics, 2017. Ca-Cancer J. Clin. 2017, 67, 7– 30, DOI: 10.3322/caac.21387Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1c7isFWgtA%253D%253D&md5=e439f3424d5a773e4ae289c392236ad6Cancer Statistics, 2017Siegel Rebecca L; Miller Kimberly D; Jemal AhmedinCA: a cancer journal for clinicians (2017), 67 (1), 7-30 ISSN:.Each year, the American Cancer Society estimates the numbers of new cancer cases and deaths that will occur in the United States in the current year and compiles the most recent data on cancer incidence, mortality, and survival. Incidence data were collected by the Surveillance, Epidemiology, and End Results Program; the National Program of Cancer Registries; and the North American Association of Central Cancer Registries. Mortality data were collected by the National Center for Health Statistics. In 2017, 1,688,780 new cancer cases and 600,920 cancer deaths are projected to occur in the United States. For all sites combined, the cancer incidence rate is 20% higher in men than in women, while the cancer death rate is 40% higher. However, sex disparities vary by cancer type. For example, thyroid cancer incidence rates are 3-fold higher in women than in men (21 vs 7 per 100,000 population), despite equivalent death rates (0.5 per 100,000 population), largely reflecting sex differences in the "epidemic of diagnosis." Over the past decade of available data, the overall cancer incidence rate (2004-2013) was stable in women and declined by approximately 2% annually in men, while the cancer death rate (2005-2014) declined by about 1.5% annually in both men and women. From 1991 to 2014, the overall cancer death rate dropped 25%, translating to approximately 2,143,200 fewer cancer deaths than would have been expected if death rates had remained at their peak. Although the cancer death rate was 15% higher in blacks than in whites in 2014, increasing access to care as a result of the Patient Protection and Affordable Care Act may expedite the narrowing racial gap; from 2010 to 2015, the proportion of blacks who were uninsured halved, from 21% to 11%, as it did for Hispanics (31% to 16%). Gains in coverage for traditionally underserved Americans will facilitate the broader application of existing cancer control knowledge across every segment of the population. CA Cancer J Clin 2017;67:7-30. © 2017 American Cancer Society.
- 42Grasso, C.; Wu, Y.; Robinson, D.; Cao, X.; Dhanasekaran, S.; Khan, A.; Quist, M.; Jing, X.; Lonigro, R.; Brenner, J.; Asangani, I.; Ateeq, B.; Chun, S.; Siddiqui, J.; Sam, L.; Anstett, M.; Mehra, R.; Prensner, J.; Palanisamy, N.; Ryslik, G.; Vandin, F.; Raphael, B.; Kunju, L.; Rhodes, D.; Pienta, K.; Chinnaiyan, A.; Tomlins, S. The mutational landscape of lethal castration-resistant prostate cancer. Nature 2012, 487, 239– 243, DOI: 10.1038/nature11125Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtVWisLfP&md5=15b1780d9cd2189325baf4e4fec84687The mutational landscape of lethal castration-resistant prostate cancerGrasso, Catherine S.; Wu, Yi-Mi; Robinson, Dan R.; Cao, Xuhong; Dhanasekaran, Saravana M.; Khan, Amjad P.; Quist, Michael J.; Jing, Xiaojun; Lonigro, Robert J.; Brenner, J. Chad; Asangani, Irfan A.; Ateeq, Bushra; Chun, Sang Y.; Siddiqui, Javed; Sam, Lee; Anstett, Matt; Mehra, Rohit; Prensner, John R.; Palanisamy, Nallasivam; Ryslik, Gregory A.; Vandin, Fabio; Raphael, Benjamin J.; Kunju, Lakshmi P.; Rhodes, Daniel R.; Pienta, Kenneth J.; Chinnaiyan, Arul M.; Tomlins, Scott A.Nature (London, United Kingdom) (2012), 487 (7406), 239-243CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Characterization of the prostate cancer transcriptome and genome has identified chromosomal rearrangements and copy no. gains and losses, including ETS gene family fusions, PTEN loss and androgen receptor (AR) amplification, which drive prostate cancer development and progression to lethal, metastatic castration-resistant prostate cancer (CRPC). However, less is known about the role of mutations. Here we sequenced the exomes of 50 lethal, heavily pre-treated metastatic CRPCs obtained at rapid autopsy (including three different foci from the same patient) and 11 treatment-naive, high-grade localized prostate cancers. We identified low overall mutation rates even in heavily treated CRPCs (2.00 per megabase) and confirmed the monoclonal origin of lethal CRPC. Integrating exome copy no. anal. identified disruptions of CHD1 that define a subtype of ETS gene family fusion-neg. prostate cancer. Similarly, we demonstrate that ETS2, which is deleted in approx. one-third of CRPCs (commonly through TMPRSS2:ERG fusions), is also deregulated through mutation. Furthermore, we identified recurrent mutations in multiple chromatin- and histone-modifying genes, including MLL2 (mutated in 8.6% of prostate cancers), and demonstrate interaction of the MLL complex with the AR, which is required for AR-mediated signaling. We also identified novel recurrent mutations in the AR collaborating factor FOXA1, which is mutated in 5 of 147 (3.4%) prostate cancers (both untreated localized prostate cancer and CRPC), and showed that mutated FOXA1 represses androgen signaling and increases tumor growth. Proteins that phys. interact with the AR, such as the ERG gene fusion product, FOXA1, MLL2, UTX (also known as KDM6A) and ASXL1 were found to be mutated in CRPC. In summary, we describe the mutational landscape of a heavily treated metastatic cancer, identify novel mechanisms of AR signaling deregulated in prostate cancer, and prioritize candidates for future study.
- 43Gundem, G.; Van, P.; Kremeyer, B.; Alexandrov, L.; Tubio, J.; Papaemmanuil, E.; Brewer, D.; Kallio, H.; Hoegnas, G.; Annala, M.; Kivinummi, K.; Goody, V.; Latimer, C.; O’Meara, S.; Dawson, K.; Isaacs, W.; Emmert-Buck, M.; Nykter, M.; Foster, C.; Kote-Jarai, Z.; Easton, D.; Whitaker, H.; Neal, D.; Cooper, C.; Eeles, R.; Visakorpi, T.; Campbell, P.; McDermott, U.; Wedge, D.; Bova, G. The evolutionary history of lethal metastatic prostate cancer. Nature 2015, 520, 353– 357, DOI: 10.1038/nature14347Google ScholarThere is no corresponding record for this reference.
- 44Lorente, D.; Mateo, J.; Perez-Lopez, R.; de, J.; Attard, G. Sequencing of agents in castration-resistant prostate cancer. Lancet Oncol. 2015, 16, 279– 292, DOI: 10.1016/S1470-2045(15)70033-1Google ScholarThere is no corresponding record for this reference.
- 45Cornford, P.; Dodson, A.; Parsons, K.; Desmond, A.; Woolfenden, A.; Fordham, M.; Neoptolemos, J.; Ke, Y.; Foster, C. Heat shock protein expression independently predicts clinical outcome in prostate cancer. Cancer Res. 2000, 60, 7099– 7105Google ScholarThere is no corresponding record for this reference.
- 46Rocchi, P.; So, A.; Kojima, S.; Signaevsky, M.; Beraldi, E.; Fazli, L.; Hurtado-Coll, A.; Yamanaka, K.; Gleave, M. Heat shock protein 27 increases after androgen ablation and plays a cytoprotective role in hormone-refractory prostate cancer. Cancer Res. 2004, 64, 6595– 6602, DOI: 10.1158/0008-5472.CAN-03-3998Google ScholarThere is no corresponding record for this reference.
- 47Jego, G.; Hazoume, A.; Seigneuric, R.; Garrido, C. Targeting heat shock proteins in cancer. Cancer Lett. 2013, 332, 275– 285, DOI: 10.1016/j.canlet.2010.10.014Google Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXlvVWitLs%253D&md5=4e1dcc15387cbb33d4a2d18f5defdd16Targeting heat shock proteins in cancerJego, Gaetan; Hazoume, Adonis; Seigneuric, Renaud; Garrido, CarmenCancer Letters (New York, NY, United States) (2013), 332 (2), 275-285CODEN: CALEDQ; ISSN:0304-3835. (Elsevier)A review. Heat shock proteins (HSPs) HSP27, HSP70 and HSP90 are powerful chaperones. Their expression is induced in response to a wide variety of physiol. and environmental insults including anti-cancer chemotherapy, thus allowing the cell to survive to lethal conditions. Different functions of HSPs have been described to account for their cytoprotective function, including their role as mol. chaperones as they play a central role in the correct folding of misfolded proteins, but also their anti-apoptotic properties. HSPs are often overexpressed in cancer cells and this constitutive expression is necessary for cancer cells' survival. HSPs may have oncogene-like functions and likewise mediate "non-oncogene addiction" of stressed tumor cells that must adapt to a hostile microenvironment, thereby becoming dependent for their survival on HSPs. HSP-targeting drugs have therefore emerged as potential anti-cancer agents. This review describes the different mols. and approaches being used or proposed in cancer therapy based on the in inhibition of HSP90, HSP70 and HSP27.
- 48Wei, T.; Chen, C.; Liu, J.; Liu, C.; Posocco, P.; Liu, X.; Cheng, Q.; Huo, S.; Liang, Z.; Fermeglia, M.; Pricl, S.; Liang, X.; Rocchic, P.; Peng, L. Anticancer drug nanomicelles formed by self-assembling amphiphilic dendrimer to combat cancer drug resistance. Proc. Natl. Acad. Sci. U. S. A. 2015, 112, 2978– 2983, DOI: 10.1073/pnas.1418494112Google Scholar48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXjtFemur4%253D&md5=ba3fcc69507c6da884cfeeabd95fb782Anticancer drug nanomicelles formed by self-assembling amphiphilic dendrimer to combat cancer drug resistanceWei, Tuo; Chen, Chao; Liu, Juan; Liu, Cheng; Posocco, Paola; Liu, Xiaoxuan; Cheng, Qiang; Huo, Shuaidong; Liang, Zicai; Fermeglia, Maurizio; Pricl, Sabrina; Liang, Xing-Jie; Rocchi, Palma; Peng, LingProceedings of the National Academy of Sciences of the United States of America (2015), 112 (10), 2978-2983CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Drug resistance and toxicity constitute challenging hurdles for cancer therapy. The application of nanotechnol. for anticancer drug delivery is expected to address these issues and bring new hope for cancer treatment. In this context, we established an original nanomicellar drug delivery system based on an amphiphilic dendrimer (AmDM), which could generate supramol. micelles to effectively encapsulate the anticancer drug doxorubicin (DOX) with high drug-loading capacity (>40%), thanks to the unique dendritic structure creating large void space for drug accommodation. The resulting AmDM/DOX nanomicelles were able to enhance drug potency and combat doxorubicin resistance in breast cancer models by significantly enhancing cellular uptake while considerably decreasing efflux of the drug. In addn., the AmDM/DOX nanoparticles abolished significantly the toxicity related to the free drug. Collectively, our studies demonstrate that the drug delivery system based on nanomicelles formed with the self-assembling amphiphilic dendrimer constitutes a promising and effective drug carrier in cancer therapy.
- 49Posocco, P.; Liu, X.; Laurini, E.; Marson, D.; Chen, C.; Liu, C.; Fermeglia, M.; Rocchi, P.; Pricl, S.; Peng, L. Impact of siRNA overhangs for dendrimer-mediated siRNA delivery and gene silencing. Mol. Pharmaceutics 2013, 10, 3262– 3273, DOI: 10.1021/mp400329gGoogle Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtVyjur%252FI&md5=bc7721f224b9006c6e075bf8a0ff401fImpact of siRNA Overhangs for Dendrimer-Mediated siRNA Delivery and Gene SilencingPosocco, Paola; Liu, Xiaoxuan; Laurini, Erik; Marson, Domenico; Chen, Chao; Liu, Cheng; Fermeglia, Maurizio; Rocchi, Palma; Pricl, Sabrina; Peng, LingMolecular Pharmaceutics (2013), 10 (8), 3262-3273CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)Small interfering RNA (siRNA) have attracted considerable attention, as compelling therapeutics providing safe and competent delivery systems are available. Dendrimers are emerging as appealing siRNA delivery vectors thanks to their unique, well-defined architecture and the resulting cooperativity and multivalency confined within a nanostructure. We have recently disclosed the structurally flexible fifth-generation TEA-core PAMAM dendrimer (G5) as an effective nanocarrier for delivery of sticky siRNA bearing long complementary sequence overhangs (dA)n/(dT)n (n = 5 or 7). Here, using combined exptl./computational approaches, we successfully clarified (i) the underlying mechanisms of interaction between the dendrimer nanovector G5 and siRNA mols. bearing either complementary or noncomplementary sequence overhangs of different length and chem. and (ii) the impact of siRNA overhangs contributing toward the improved delivery potency. Using siRNA with complementary overhangs offer the best action in term of gene silencing through the formation of concatemers, i.e., supramol. structures resulting from synergistic and cooperative binding via (dA)n/(dT)n bridges (n = 5 or 7). On the other hand, although siRNA bearing long, noncomplementary overhangs (dA)n/(dA)n or (dT)n/(dT)n (n = 5 or 7) are endowed with considerably higher gene silencing potency than normal siRNA with (dT)2/(dT)2, they remain less effective than their sticky siRNA counterparts. The obsd. gene silencing potency depends on length, nature, and flexibility of the overhangs, which behave as a sort of clamps that hold and interact with the dendrimer nanovectors, thus impacting siRNA delivery performance and, ultimately, gene silencing. Our findings can be instrumental in designing siRNA entities with enhanced capability to achieve effective RNA interference for therapeutic applications.
- 50Albanyan, B.; Laurini, E.; Posocco, P.; Pricl, S.; Smith, D. Self-assembled multivalent (SAMul) polyanion binding—impact of hydrophobic modifications in the micellar core on DNA and heparin binding at the peripheral cationic ligands. Chem. - Eur. J. 2017, 23, 6391– 6397, DOI: 10.1002/chem.201700177Google Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXlvVCgsLk%253D&md5=e971234081cca25156b7bcde61149d98Self-Assembled Multivalent (SAMul) Polyanion Binding-Impact of Hydrophobic Modifications in the Micellar Core on DNA and Heparin Binding at the Peripheral Cationic LigandsAlbanyan, Buthaina; Laurini, Erik; Posocco, Paola; Pricl, Sabrina; Smith, David K.Chemistry - A European Journal (2017), 23 (26), 6391-6397CODEN: CEUJED; ISSN:0947-6539. (Wiley-VCH Verlag GmbH & Co. KGaA)This paper reports a small family of cationic surfactants designed to bind polyanions such as DNA and heparin. Each mol. has the same hydrophilic cationic ligand and a hydrophobic aliph. group with eighteen carbon atoms with one, two, or three alkene groups within the hydrophobic chain (C18-1, C18-2 and C18-3). Dynamic light scattering indicates that more alkenes lead to geometric distortion, giving rise to larger self-assembled multivalent (SAMul) nanostructures. Mallard Blue and Ethidium Bromide dye displacement assays demonstrate that heparin and DNA have markedly different binding preferences, with heparin binding most effectively to C18-1, and DNA to C18-3, even though the mol. structural differences of these SAMul systems are buried in the hydrophobic core. Multiscale modeling suggests that adaptive heparin maximises enthalpically favorable interactions with C18-1, while shape-persistent DNA forms a similar no. of interactions with each ligand display, but with slightly less entropic cost for binding to C18-3-fundamental thermodn. differences in SAMul binding of heparin or DNA. This study therefore provides unique insight into electrostatic mol. recognition between highly charged nanoscale surfaces in biol. relevant systems.
- 51Fechner, L.; Albanyan, B.; Vieira, V.; Laurini, E.; Posocco, P.; Pricl, S.; Smith, D. Electrostatic binding of polyanions using self-assembled multivalent (SAMul) ligand displays - structure-activity effects on DNA/heparin binding. Chem. Sci. 2016, 7, 4653– 4659, DOI: 10.1039/C5SC04801JGoogle Scholar51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xms1Kks7o%253D&md5=2663d127c7871258d0687f089ec63624Electrostatic binding of polyanions using self-assembled multivalent (SAMul) ligand displays - structure-activity effects on DNA/heparin bindingFechner, Loryn E.; Albanyan, Buthaina; Vieira, Vania M. P.; Laurini, Erik; Posocco, Paola; Pricl, Sabrina; Smith, David K.Chemical Science (2016), 7 (7), 4653-4659CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)This paper reports that modifying the ligands in self-assembled multivalent (SAMul) displays has an impact on apparent binding selectivity towards two nanoscale biol. polyanions - heparin and DNA. For the nanostructures assayed here, spermidine ligands are optimal for heparin binding but spermine ligands are preferred for DNA. Probing subtle differences in such nanoscale binding interfaces is a significant challenge, and as such, several exptl. binding assays - competition assays and isothermal calorimetry - are employed to confirm differences in affinity and provide thermodn. insights. Given the dynamic nature and hierarchical binding processes involved in SAMul systems, we employed multiscale modeling to propose reasons for the origins of polyanion selectivity differences. The modeling results, when expressed in thermodn. terms and compared with the exptl. data, suggest that DNA is a shape-persistent polyanion, and selectivity originates only from ligand preferences, whereas heparin is more flexible and adaptive, and as such, actively reinforces ligand preferences. As such, this study suggests that inherent differences between polyanions may underpin subtle binding selectivity differences, and that even simple electrostatic interfaces such as these can have a degree of tunability, which has implications for biol. control and regulation on the nanoscale.
- 52Qi, C.; Su, Y.; Peng, Y.; Tian, E.; Sun, G.; Zhou, J.; Sun, G.; Liu, X.; Chao, C.; Murai, K.; Zhao, C.; Azizian, K.; Yang, L.; Warden, C.; Wu, X.; D’Apuzzo, M.; Brown, C.; Badie, B.; Peng, L.; Riggs, D.; Rossi, J.; Shi, Y. Downregulation of TLX induces TET3 expression and inhibits glioblastoma stem cell self-renewal and tumorigenesis. Nat. Commun. 2016, 7, 10637– 10651, DOI: 10.1038/ncomms10637Google ScholarThere is no corresponding record for this reference.
Cited By
This article is cited by 171 publications.
- Yue Gao, Aiyu Li, Yanying Li, Honghua Guo, Liangyu He, Kangan Li, Dzmitry Shcharbin, Xiangyang Shi, Mingwu Shen. Dendrimer/Copper(II) Complex-Mediated siRNA Delivery Disrupts Lactate Metabolism to Reprogram the Local Immune Microenvironment against Tumor Growth and Metastasis. Biomacromolecules 2024, 25
(12)
, 7995-8005. https://doi.org/10.1021/acs.biomac.4c01249
- Yuki Koba, Masahiko Nakamoto, Masanori Nagao, Yoshiko Miura, Michiya Matsusaki. Intrinsic Synergy and Selectivity for the Inhibition of Cancer Cell Growth Generated by a Polymer Ligand of Proximal Enzymes. Nano Letters 2024, 24
(45)
, 14206-14214. https://doi.org/10.1021/acs.nanolett.4c03334
- Andrea Mio, Domenico Marson, Gabriele Cavalieri, Maurizio Fermeglia, Erik Laurini, Sabrina Pricl. Beyond the Veil: Free Energy Profiles and Partition Coefficients for Antimelanoma Drugs in Self-Assembled Nanomicelles via COSMOmic and Atomistic Molecular Dynamics Simulations. Journal of Chemical & Engineering Data 2024, 69
(10)
, 3450-3461. https://doi.org/10.1021/acs.jced.4c00076
- Weiqiang Zhou, Zefeng Long, Chuan Xu, Junge Zhang, Xin Zhou, Xianghai Song, Pengwei Huo, Yi Guo, Wei Xue, Quan Wang, Chen Zhou. Advances in Functionalized Biocomposites of Living Cells Combined with Metal–Organic Frameworks. Langmuir 2024, 40
(29)
, 14749-14765. https://doi.org/10.1021/acs.langmuir.4c00404
- Dapeng Zhang, Qi Xiao, Mehrnoush Rahimzadeh, Matthew Liu, Cesar Rodriguez-Emmenegger, Yusuke Miyazaki, Wataru Shinoda, Virgil Percec. Self-Assembly of Glycerol-Amphiphilic Janus Dendrimers Amplifies and Indicates Principles for the Selection of Stereochemistry by Biological Membranes. Journal of the American Chemical Society 2023, 145
(7)
, 4311-4323. https://doi.org/10.1021/jacs.3c00389
- Zhiqing Zhang, Dongyan Xu, Jiawei Wang, Ruyan Zhang, Huan Du, Ting Zhou, Xiufeng Wang, Fang Wang. Rolling Circle Amplification-Based DNA Nano-Assembly for Targeted Drug Delivery and Gene Therapy. Biomacromolecules 2023, 24
(1)
, 439-448. https://doi.org/10.1021/acs.biomac.2c01271
- Dokkari Nagalaxmi Yadav, Mohammad Sadik Ali, Ajinkya Madhukar Thanekar, Sunil Venkanna Pogu, Aravind Kumar Rengan. Recent Advancements in the Design of Nanodelivery Systems of siRNA for Cancer Therapy. Molecular Pharmaceutics 2022, 19
(12)
, 4506-4526. https://doi.org/10.1021/acs.molpharmaceut.2c00811
- Yuki Koba, Masahiko Nakamoto, Michiya Matsusaki. Fabrication of a Polymeric Inhibitor of Proximal Metabolic Enzymes in Hypoxia for Synergistic Inhibition of Cancer Cell Proliferation, Survival, and Migration. ACS Applied Materials & Interfaces 2022, 14
(46)
, 51790-51797. https://doi.org/10.1021/acsami.2c16454
- Wathsala Liyanage, Tony Wu, Sujatha Kannan, Rangaramanujam M. Kannan. Dendrimer–siRNA Conjugates for Targeted Intracellular Delivery in Glioblastoma Animal Models. ACS Applied Materials & Interfaces 2022, 14
(41)
, 46290-46303. https://doi.org/10.1021/acsami.2c13129
- Jiaxuan Chen, Dandan Zhu, Xiaoxuan Liu, Ling Peng. Amphiphilic Dendrimer Vectors for RNA Delivery: State-of-the-Art and Future Perspective. Accounts of Materials Research 2022, 3
(5)
, 484-497. https://doi.org/10.1021/accountsmr.1c00272
- Wei Guo, Wenchang Liu, Pengqi Wan, Hao Wang, Chunsheng Xiao, Li Chen, Xuesi Chen. Cationic Amphiphilic Dendrons with Anticancer Activity. ACS Biomaterials Science & Engineering 2022, 8
(5)
, 2121-2130. https://doi.org/10.1021/acsbiomaterials.2c00181
- Dapeng Zhang, Elena N. Atochina-Vasserman, Juncheng Lu, Devendra S. Maurya, Qi Xiao, Matthew Liu, Jasper Adamson, Nathan Ona, Erin K. Reagan, Houping Ni, Drew Weissman, Virgil Percec. The Unexpected Importance of the Primary Structure of the Hydrophobic Part of One-Component Ionizable Amphiphilic Janus Dendrimers in Targeted mRNA Delivery Activity. Journal of the American Chemical Society 2022, 144
(11)
, 4746-4753. https://doi.org/10.1021/jacs.2c00273
- Hao Wang, Ning Liu, Fuxu Yang, Nannan Hu, Mingyue Wang, Meiying Cui, Nico Bruns, Xingang Guan. Bioengineered Protein Nanocage by Small Heat Shock Proteins Delivering mTERT siRNA for Enhanced Colorectal Cancer Suppression. ACS Applied Bio Materials 2022, 5
(3)
, 1330-1340. https://doi.org/10.1021/acsabm.1c01221
- Siming Huang, Wangshu Zhu, Fang Zhang, Guosheng Chen, Xiaoxue Kou, Xieqing Yang, Gangfeng Ouyang, Jun Shen. Silencing of Pyruvate Kinase M2 via a Metal–Organic Framework Based Theranostic Gene Nanomedicine for Triple-Negative Breast Cancer Therapy. ACS Applied Materials & Interfaces 2021, 13
(48)
, 56972-56987. https://doi.org/10.1021/acsami.1c18053
- Dapeng Zhang, Elena N. Atochina-Vasserman, Devendra S. Maurya, Matthew Liu, Qi Xiao, Juncheng Lu, George Lauri, Nathan Ona, Erin K. Reagan, Houping Ni, Drew Weissman, Virgil Percec. Targeted Delivery of mRNA with One-Component Ionizable Amphiphilic Janus Dendrimers. Journal of the American Chemical Society 2021, 143
(43)
, 17975-17982. https://doi.org/10.1021/jacs.1c09585
- Dapeng Zhang, Elena N. Atochina-Vasserman, Devendra S. Maurya, Ning Huang, Qi Xiao, Nathan Ona, Matthew Liu, Hamna Shahnawaz, Houping Ni, Kyunghee Kim, Margaret M. Billingsley, Darrin J. Pochan, Michael J. Mitchell, Drew Weissman, Virgil Percec. One-Component Multifunctional Sequence-Defined Ionizable Amphiphilic Janus Dendrimer Delivery Systems for mRNA. Journal of the American Chemical Society 2021, 143
(31)
, 12315-12327. https://doi.org/10.1021/jacs.1c05813
- Mingshu Chen, Yunqiu Miao, Kun Qian, Xin Zhou, Linmiao Guo, Yu Qiu, Rui Wang, Yong Gan, Xinxin Zhang. Detachable Liposomes Combined Immunochemotherapy for Enhanced Triple-Negative Breast Cancer Treatment through Reprogramming of Tumor-Associated Macrophages. Nano Letters 2021, 21
(14)
, 6031-6041. https://doi.org/10.1021/acs.nanolett.1c01210
- Lei Chen, Gaigai Li, Xingxing Wang, Jinbo Li, Yan Zhang. Spherical Nucleic Acids for Near-Infrared Light-Responsive Self-Delivery of Small-Interfering RNA and Antisense Oligonucleotide. ACS Nano 2021, 15
(7)
, 11929-11939. https://doi.org/10.1021/acsnano.1c03072
- Haisi Dong, Guangqi Song, Danhui Ma, Tiedong Wang, Shisong Jing, Haimiao Yang, Ye Tao, Yong Tang, Yan Shi, Zhen Dai, Ji-min Zhu, Taotao Liu, Bingmei Wang, Xiangyang Leng, Xizhong Shen, Changfeng Zhu, Yicheng Zhao. Improved Antiviral Activity of Classical Swine Fever Virus-Targeted siRNA by Tetrahedral Framework Nucleic Acid-Enhanced Delivery. ACS Applied Materials & Interfaces 2021, 13
(25)
, 29416-29423. https://doi.org/10.1021/acsami.1c08143
- Qiuhong Zhang, Junmin Zhang, Jun Song, Yizhen Liu, Xiangzhong Ren, Yanli Zhao. Protein-Based Nanomedicine for Therapeutic Benefits of Cancer. ACS Nano 2021, 15
(5)
, 8001-8038. https://doi.org/10.1021/acsnano.1c00476
- Jiangfan Cao, Orysia T. Zaremba, Qi Lei, Evelyn Ploetz, Stefan Wuttke, Wei Zhu. Artificial Bioaugmentation of Biomacromolecules and Living Organisms for Biomedical Applications. ACS Nano 2021, 15
(3)
, 3900-3926. https://doi.org/10.1021/acsnano.0c10144
- Serge Mignani, Xiangyang Shi, Maria Zablocka, Jean-Pierre Majoral. Dendritic Macromolecular Architectures: Dendrimer-Based Polyion Complex Micelles. Biomacromolecules 2021, 22
(2)
, 262-274. https://doi.org/10.1021/acs.biomac.0c01645
- Yichuan Zhang, Muhammed Üçüncü, Alessia Gambardella, Assel Baibek, Jin Geng, Shuo Zhang, Jessica Clavadetscher, Inga Litzen, Mark Bradley, Annamaria Lilienkampf. Bioorthogonal Swarming: In Situ Generation of Dendrimers. Journal of the American Chemical Society 2020, 142
(52)
, 21615-21621. https://doi.org/10.1021/jacs.0c07869
- Zhenbin Lyu, Ling Ding, Aura Tintaru, Ling Peng. Self-Assembling Supramolecular Dendrimers for Biomedical Applications: Lessons Learned from Poly(amidoamine) Dendrimers. Accounts of Chemical Research 2020, 53
(12)
, 2936-2949. https://doi.org/10.1021/acs.accounts.0c00589
- Yanjing Yang, Yao He, Zhiwei Deng, Jiacheng Li, Jin Huang, Shian Zhong. Intelligent Nanoprobe: Acid-Responsive Drug Release and In Situ Evaluation of Its Own Therapeutic Effect. Analytical Chemistry 2020, 92
(18)
, 12371-12378. https://doi.org/10.1021/acs.analchem.0c02099
- Xiyu Ke, Lillie Shelton, Yizong Hu, Yining Zhu, Emily Chow, Haoyu Tang, José Luis Santos, Hai-Quan Mao. Surface-Functionalized PEGylated Nanoparticles Deliver Messenger RNA to Pulmonary Immune Cells. ACS Applied Materials & Interfaces 2020, 12
(32)
, 35835-35844. https://doi.org/10.1021/acsami.0c08268
- Raut Bholakant, Hongliang Qian, Junmei Zhang, Xin Huang, Dechun Huang, Jan Feijen, Yinan Zhong, Wei Chen. Recent Advances of Polycationic siRNA Vectors for Cancer Therapy. Biomacromolecules 2020, 21
(8)
, 2966-2982. https://doi.org/10.1021/acs.biomac.0c00438
- Mengjie Zhang, Yuhua Weng, Ziyang Cao, Shuai Guo, Bo Hu, Mei Lu, Weisheng Guo, Tongren Yang, Chunhui Li, Xianzhu Yang, Yuanyu Huang. ROS-Activatable siRNA-Engineered Polyplex for NIR-Triggered Synergistic Cancer Treatment. ACS Applied Materials & Interfaces 2020, 12
(29)
, 32289-32300. https://doi.org/10.1021/acsami.0c06614
- Marc Heitz, Susanna Zamolo, Sacha Javor, Jean-Louis Reymond. Fluorescent Peptide Dendrimers for siRNA Transfection: Tracking pH Responsive Aggregation, siRNA Binding, and Cell Penetration. Bioconjugate Chemistry 2020, 31
(6)
, 1671-1684. https://doi.org/10.1021/acs.bioconjchem.0c00231
- Wei Zhou, Guogui Sun, Zhen Zhang, Libo Zhao, Li Xu, Hongyu Yuan, Shumu Li, Zaizai Dong, Yongmei Song, Xiaohong Fang. Proteasome-Independent Protein Knockdown by Small-Molecule Inhibitor for the Undruggable Lung Adenocarcinoma. Journal of the American Chemical Society 2019, 141
(46)
, 18492-18499. https://doi.org/10.1021/jacs.9b08777
- Jiantao Yu, Jing Li, Shiyao Zhai, Li Lin, Kui Wang, Bin Tang, Hong Meng, Leilei Tian. Enzymatically Synthesized DNA Polymer as Co-carrier for Enhanced RNA Interference. ACS Applied Bio Materials 2019, 2
(11)
, 5204-5215. https://doi.org/10.1021/acsabm.9b00862
- Rachit Sapra, Ram P. Verma, Govind P. Maurya, Sameer Dhawan, Jisha Babu, V. Haridas. Designer Peptide and Protein Dendrimers: A Cross-Sectional Analysis. Chemical Reviews 2019, 119
(21)
, 11391-11441. https://doi.org/10.1021/acs.chemrev.9b00153
- Natesan Thirumalaivasan, Parthiban Venkatesan, Ping-Shan Lai, Shu-Pao Wu. In Vitro and In Vivo Approach of Hydrogen-Sulfide-Responsive Drug Release Driven by Azide-Functionalized Mesoporous Silica Nanoparticles. ACS Applied Bio Materials 2019, 2
(9)
, 3886-3896. https://doi.org/10.1021/acsabm.9b00481
- Marc Heitz, Sacha Javor, Tamis Darbre, Jean-Louis Reymond. Stereoselective pH Responsive Peptide Dendrimers for siRNA Transfection. Bioconjugate Chemistry 2019, 30
(8)
, 2165-2182. https://doi.org/10.1021/acs.bioconjchem.9b00403
- Yajun Wang, Tian Zhang, Cuilan Hou, Menghang Zu, Yi Lu, Xianbin Ma, Die Jia, Peng Xue, Yuejun Kang, Zhigang Xu. Mitochondria-Specific Anticancer Drug Delivery Based on Reduction-Activated Polyprodrug for Enhancing the Therapeutic Effect of Breast Cancer Chemotherapy. ACS Applied Materials & Interfaces 2019, 11
(32)
, 29330-29340. https://doi.org/10.1021/acsami.9b10211
- Janhavi Bhavsar, Kaustubh Kasture, Bhagyashree V Salvi, Pravin Shende. Strategies for transportation of peptides across the skin for treatment of multiple diseases. Therapeutic Delivery 2025, 16
(1)
, 63-86. https://doi.org/10.1080/20415990.2024.2411943
- Maya Beena, Purnimajayasree Ramesh, Arunkumar Palaniappan. Poly(amidoamine)-based dendrimers for biomedical applications. 2025, 105-132. https://doi.org/10.1016/B978-0-323-95233-0.00017-0
- Wangwang Zhang, Leyang Song, Yi Zhou, Jishang Sun, Cuiyao Li, Baoqin Han, Jing Chang, Baosan Han, Ting Wang. Study on the inhibition of non-small cell lung cancer mediated by chitosan-based gene carrier delivering STAT3-shRNA. International Journal of Biological Macromolecules 2025, 284 , 138211. https://doi.org/10.1016/j.ijbiomac.2024.138211
- Jose Garcia Coll, Pauline Trousselier, Sachin Dattram Pawar, Yannick Bessin, Laure Lichon, Jeanne Chain Leblond, Emmanuelle Sachon, Nadir Bettache, Sébastien Ulrich. Amphiphilic Dynamic Covalent Polymer Vectors of siRNA. Chemical Science 2025, https://doi.org/10.1039/D4SC07668K
- Caden M. Henningfield, Neelakshi Soni, Ryan W. Lee, Rishi Sharma, Jeffrey L. Cleland, Kim N. Green. Selective targeting and modulation of plaque associated microglia via systemic hydroxyl dendrimer administration in an Alzheimer’s disease mouse model. Alzheimer's Research & Therapy 2024, 16
(1)
https://doi.org/10.1186/s13195-024-01470-3
- Bilan Wang, Shiqi Hu, Yan Teng, Junli Chen, Haoyuan Wang, Yezhen Xu, Kaiyu Wang, Jianguo Xu, Yongzhong Cheng, Xiang Gao. Current advance of nanotechnology in diagnosis and treatment for malignant tumors. Signal Transduction and Targeted Therapy 2024, 9
(1)
https://doi.org/10.1038/s41392-024-01889-y
- Philemon Ubanako, Sheefa Mirza, Paul Ruff, Clement Penny. Exosome-mediated delivery of siRNA molecules in cancer therapy: triumphs and challenges. Frontiers in Molecular Biosciences 2024, 11 https://doi.org/10.3389/fmolb.2024.1447953
- Nazgol Karimi Dastgerdi, Nazanin Karimi Dastgerdi, Hulya Bayraktutan, Gabriella Costabile, Fatemeh Atyabi, Rassoul Dinarvand, Giuseppe Longobardi, Cameron Alexander, Claudia Conte. Enhancing siRNA cancer therapy: Multifaceted strategies with lipid and polymer-based carrier systems. International Journal of Pharmaceutics 2024, 663 , 124545. https://doi.org/10.1016/j.ijpharm.2024.124545
- Amrita Ghosh Majumdar, Biswajit Pany, Sankha Subhra Parua, Debdeepto Mukherjee, Agnihotri Panda, Madhuchhanda Mohanty, Biswadeep Das, Satyabrata Si, Priti S. Mohanty. Stimuli-Responsive Nanogel/Microgel Hybrids as Targeted Drug Delivery Systems: A Comprehensive Review. BioNanoScience 2024, 14
(3)
, 3496-3521. https://doi.org/10.1007/s12668-024-01577-9
- Vildan Torun, Elif Degerli, Demet Cansaran-Duman. Revealing the Molecular Signatures of miR-185-5p on Breast Cancer Cells Using Proteomic Analysis. Protein & Peptide Letters 2024, 31
(9)
, 681-695. https://doi.org/10.2174/0109298665322427240906060626
- Lingxiao Xu, Zhenxuan Shao, Xia Fang, Zengfeng Xin, Shenzhi Zhao, Hongbo Zhang, Yu Zhang, Wenbiao Zheng, Xiaohua Yu, Zengjie Zhang, Lingling Sun. Exploring precision treatments in immune‐mediated inflammatory diseases: Harnessing the infinite potential of nucleic acid delivery. Exploration 2024, 21 https://doi.org/10.1002/EXP.20230165
- Rohit Bhatia, Amandeep Singh, Sukhwinder Singh, Navneesh, Ravindra Kumar Rawal. Emerging trends in nano-carrier based gene delivery systems for targeted cancer therapy. Journal of Drug Delivery Science and Technology 2024, 95 , 105546. https://doi.org/10.1016/j.jddst.2024.105546
- Jingting Wu, Xiaoqi Zheng, Wenqing Lin, Linhuan Chen, Zai‐Sheng Wu. Persistent Targeting DNA Nanocarrier Made of 3D Structural Unit Assembled from Only One Basic Multi‐Palindromic Oligonucleotide for Precise Gene Cancer Therapy. Advanced Healthcare Materials 2024, 13
(11)
https://doi.org/10.1002/adhm.202303865
- Ehsan Lotfi, Azam Kholghi, Fereshteh Golab, Ali Mohammadi, Mahmood Barati. Circulating miRNAs and lncRNAs serve as biomarkers for early colorectal cancer diagnosis. Pathology - Research and Practice 2024, 255 , 155187. https://doi.org/10.1016/j.prp.2024.155187
- Saurabh Shah, Paras Famta, Dadi A. Srinivasarao, Syed Shahrukh, Naitik Jain, Akshay Shinde, Sajja Bhanu Prasad, Ganesh Vambhurkar, Giriraj Pandey, Rajendra Kumar, Shailendra Saraf, Saurabh Srivastava. Nanomedicine-RNAi interface: Architypes in prostate cancer therapeutics. Journal of Drug Delivery Science and Technology 2024, 92 , 105349. https://doi.org/10.1016/j.jddst.2024.105349
- K Sai Manogna, B Deva Prasad Raju, G Rajasekhara Reddy, Parashuram Kallem, Mannur Ismail Shaik, N John Sushma. Investigations on anticancer activity of Eu3+ doped hydroxyapatite nanocomposites against MCF7 and 4T1 breast cancer cell lines: A structural and luminescence Perspective. Heliyon 2024, 10
(3)
, e25064. https://doi.org/10.1016/j.heliyon.2024.e25064
- Miqdam M. Obaid AL-Jumaili. Transcription Silencing and CpGs Hypermethylation as Therapeutic Gene Editing in Clinical Colorectal Adenocarcinoma Repression. The Korean Journal of Gastroenterology 2024, 83
(1)
, 6-16. https://doi.org/10.4166/kjg.2023.132
- Susanna Zamolo, Elena Zakharova, Lise Boursinhac, Florian Hollfelder, Tamis Darbre, Jean-Louis Reymond. Peptide dendrimers transfecting CRISPR/Cas9 plasmid DNA: optimization and mechanism. RSC Chemical Biology 2024, 8 https://doi.org/10.1039/D4CB00116H
- Savannah R. Brown, Emilie E. Vomhof-DeKrey, Sarmad Al-Marsoummi, Nicholas D. Brown, Kole Hermanson, Marc D. Basson. Schlafen Family Intra-Regulation by IFN-α2 in Triple-Negative Breast Cancer. Cancers 2023, 15
(23)
, 5658. https://doi.org/10.3390/cancers15235658
- Pragati Ramesh Kumbhar, Prakash Kumar, Aarti Lasure, Ravichandiran Velayutham, Debabrata Mandal. An updated landscape on nanotechnology-based drug delivery, immunotherapy, vaccinations, imaging, and biomarker detections for cancers: recent trends and future directions with clinical success. Discover Nano 2023, 18
(1)
https://doi.org/10.1186/s11671-023-03913-6
- Longjie Li, Yukai Deng, Yonghui Zeng, Bei Yan, Yulian Deng, Ziyang Zheng, Siqi Li, Yuhang Yang, Jinwei Hao, Xianjin Xiao, Xinyu Wang. The application advances of dendrimers in biomedical field. VIEW 2023, 4
(6)
https://doi.org/10.1002/VIW.20230023
- Yuhe Zhang, Jiahao Shi, Bin Ma, Ya-Nan Zhou, Haiyang Yong, Jianzhong Li, Xiangyi Kong, Dezhong Zhou. Functionalization of polymers for intracellular protein delivery. Progress in Polymer Science 2023, 146 , 101751. https://doi.org/10.1016/j.progpolymsci.2023.101751
- Jong Won Lee, Man Kyu Shim, Hyosuk Kim, Hochung Jang, Yuhan Lee, Sun Hwa Kim. RNAi therapies: Expanding applications for extrahepatic diseases and overcoming delivery challenges. Advanced Drug Delivery Reviews 2023, 201 , 115073. https://doi.org/10.1016/j.addr.2023.115073
- Valentina Andretto, Silvia Dusi, Serena Zilio, Mathieu Repellin, David Kryza, Stefano Ugel, Giovanna Lollo. Tackling TNF-α in autoinflammatory disorders and autoimmune diseases: From conventional to cutting edge in biologics and RNA- based nanomedicines. Advanced Drug Delivery Reviews 2023, 201 , 115080. https://doi.org/10.1016/j.addr.2023.115080
- Peng Zhang, Zhi Li, Weiling Cao, Jingjie Tang, Yi Xia, Ling Peng, Jing Ma. A PD‐L1 Antibody‐Conjugated PAMAM Dendrimer Nanosystem for Simultaneously Inhibiting Glycolysis and Promoting Immune Response in Fighting Breast Cancer. Advanced Materials 2023, 35
(41)
https://doi.org/10.1002/adma.202305215
- Jie Wang, Ping Li, Chao Wang, Ning Liu, Dongming Xing. Molecularly or atomically precise nanostructures for bio-applications: how far have we come?. Materials Horizons 2023, 10
(9)
, 3304-3324. https://doi.org/10.1039/D3MH00574G
- Anirban Goutam Mukherjee, Uddesh Ramesh Wanjari, Abilash Valsala Gopalakrishnan, Pragya Bradu, Antara Biswas, Raja Ganesan, Kaviyarasi Renu, Abhijit Dey, Balachandar Vellingiri, Achraf El Allali, Alsamman M. Alsamman, Hatem Zayed, C. George Priya Doss. Evolving strategies and application of proteins and peptide therapeutics in cancer treatment. Biomedicine & Pharmacotherapy 2023, 163 , 114832. https://doi.org/10.1016/j.biopha.2023.114832
- Xue Jiang, Jiaojiao Zhang, Pik Kwan Lo, Zhengwei Mao. Polyphenol‐Based Assembly Nanosystems as Multifunctional Biopharmaceutical Platform. Advanced NanoBiomed Research 2023, 3
(6)
https://doi.org/10.1002/anbr.202200168
- Xiaobin Li, Junyu Liu, Haihong Chen, Yaxin Chen, Yi Wang, Can Yang Zhang, Xin-Hui Xing. Multi-functional engineered polypeptide-based drug delivery systems for improved cancer therapy. Green Chemical Engineering 2023, 4
(2)
, 173-188. https://doi.org/10.1016/j.gce.2022.07.010
- Jiaxuan Chen, Dandan Zhu, Baoping Lian, Kangjie Shi, Peng Chen, Ying Li, Wenyi Lin, Ling Ding, Qiulin Long, Yang Wang, Erik Laurini, Wenjun Lan, Yun Li, Aura Tintaru, Caoyun Ju, Can Zhang, Sabrina Pricl, Juan Iovanna, Xiaoxuan Liu, Ling Peng. Cargo-selective and adaptive delivery of nucleic acid therapeutics by bola-amphiphilic dendrimers. Proceedings of the National Academy of Sciences 2023, 120
(21)
https://doi.org/10.1073/pnas.2220787120
- Dongdong Guo, Xiaoyong Dai, Kewei Liu, Yuhong Liu, Jiamin Wu, Kun Wang, Shengwei Jiang, Fen Sun, Lijun Wang, Bing Guo, Dongye Yang, Laiqiang Huang. A Self‐Reinforcing Nanoplatform for Highly Effective Synergistic Targeted Combinatary Calcium‐Overload and Photodynamic Therapy of Cancer. Advanced Healthcare Materials 2023, 12
(12)
https://doi.org/10.1002/adhm.202202424
- Yiqing Liu, Peiyong Guo, Xinhao Dong, Yina Xu, Dan Li, Hua Zheng, Jianhong Liao. Synchronized delivery of dual-drugs for potentiating combination chemotherapy based on smart triple-responsive polymeric micelles. Biomaterials Advances 2023, 147 , 213344. https://doi.org/10.1016/j.bioadv.2023.213344
- Dolly Jain, Shiv Kumar Prajapati, Ankit Jain, Rohit Singhal. Nano-formulated siRNA-based therapeutic approaches for cancer therapy. Nano Trends 2023, 1 , 100006. https://doi.org/10.1016/j.nwnano.2023.100006
- Yingqi Liu, Yunyun Wu, Zhong Luo, Menghuan Li. Designing supramolecular self-assembly nanomaterials as stimuli-responsive drug delivery platforms for cancer therapy. iScience 2023, 26
(3)
, 106279. https://doi.org/10.1016/j.isci.2023.106279
- Ahmed Khaled Abosalha, Waqar Ahmad, Jacqueline Boyajian, Paromita Islam, Merry Ghebretatios, Sabrina Schaly, Rahul Thareja, Karan Arora, Satya Prakash. A comprehensive update of siRNA delivery design strategies for targeted and effective gene silencing in gene therapy and other applications. Expert Opinion on Drug Discovery 2023, 18
(2)
, 149-161. https://doi.org/10.1080/17460441.2022.2155630
- Anabela Gonçalves, Mário Fernandes, Margarida Lima, João Pedro Gomes, Filipa Silva, Sara Castro, Filipa Sampaio, Andreia Castro Gomes. Nanotechnology to the Rescue: Therapeutic Strategies Based on Brown Algae for Neurodegenerative Diseases. Applied Sciences 2023, 13
(3)
, 1883. https://doi.org/10.3390/app13031883
- Hallie Gaitsch, Andrew M. Hersh, Safwan Alomari, Betty M. Tyler. Dendrimer Technology in Glioma: Functional Design and Potential Applications. Cancers 2023, 15
(4)
, 1075. https://doi.org/10.3390/cancers15041075
- Adina Căta, Ioana Maria Carmen Ienașcu, Mariana Nela Ştefănuț, Dan Roșu, Oana-Raluca Pop. Properties and Bioapplications of Amphiphilic Janus Dendrimers: A Review. Pharmaceutics 2023, 15
(2)
, 589. https://doi.org/10.3390/pharmaceutics15020589
- Jie Shen, Jingwen Wang, Qiulian Hao, Weihong Ji, Huan Peng, Yanyue Wu, Ruichen Zhao, Xinyu Qiu, Wenli Wang, Jun Yang, Yan Li, Tianlu Zhang, Zhiguo Lu, Xin Zhang. Transformable Nanotheranostics Capable of Sensoring Nucleic Acid Drugs’ Spatiotemporal Behaviors. Advanced Functional Materials 2023, 33
(5)
https://doi.org/10.1002/adfm.202209749
- Tao Sun, Chufeng Li, Xuwen Li, Haolin Song, Boyu Su, Haoyu You, Tongyu Zhang, Chen Jiang. Pharmaceutical Nanotechnology. 2023, 179-283. https://doi.org/10.1007/978-981-16-8984-0_10
- Xingya Jiang, Kimia Abedi, Jinjun Shi. Polymeric nanoparticles for RNA delivery. 2023, 555-573. https://doi.org/10.1016/B978-0-12-822425-0.00017-8
- Helena Tomás, João Rodrigues. Dendrimers and dendrimer-based nano-objects for oncology applications. 2023, 41-78. https://doi.org/10.1016/B978-0-323-85671-3.00002-6
- Maria Russi, Rachele Valeri, Domenico Marson, Chiara Danielli, Fulvia Felluga, Aura Tintaru, Natasa Skoko, Suzana Aulic, Erik Laurini, Sabrina Pricl. Some things old, new and borrowed: Delivery of dabrafenib and vemurafenib to melanoma cells via self-assembled nanomicelles based on an amphiphilic dendrimer. European Journal of Pharmaceutical Sciences 2023, 180 , 106311. https://doi.org/10.1016/j.ejps.2022.106311
- Ahmed Khaled Abosalha, Jacqueline Boyajian, Waqar Ahmad, Paromita Islam, Merry Ghebretatios, Sabrina Schaly, Rahul Thareja, Karan Arora, Satya Prakash. Clinical pharmacology of siRNA therapeutics: current status and future prospects. Expert Review of Clinical Pharmacology 2022, 15
(11)
, 1327-1341. https://doi.org/10.1080/17512433.2022.2136166
- Rongying Zhu, Mengying Hou, Yang Zhou, Huan Ye, Lei Chen, Chenglong Ge, Shanzhou Duan, Lichen Yin, Yongbing Chen. Spherical α-helical polypeptide-mediated E2F1 silencing against myocardial ischemia-reperfusion injury (MIRI). Biomaterials Science 2022, 10
(21)
, 6258-6266. https://doi.org/10.1039/D2BM01075E
- Fei Kuang, De Luo, Mengjia Zhou, Juan Du, Jie Yang, . lncRNAs AC156455.1 and AC104532.2 as Biomarkers for Diagnosis and Prognosis in Colorectal Cancer. Disease Markers 2022, 2022 , 1-13. https://doi.org/10.1155/2022/4872001
- Simona Camorani, Silvia Tortorella, Lisa Agnello, Chiara Spanu, Annachiara d’Argenio, Roberto Nilo, Antonella Zannetti, Erica Locatelli, Monica Fedele, Mauro Comes Franchini, Laura Cerchia. Aptamer-Functionalized Nanoparticles Mediate PD-L1 siRNA Delivery for Effective Gene Silencing in Triple-Negative Breast Cancer Cells. Pharmaceutics 2022, 14
(10)
, 2225. https://doi.org/10.3390/pharmaceutics14102225
- Barbara Ziemba, Maciej Borowiec, Ida Franiak-Pietryga. There and back again: a dendrimer’s tale. Drug and Chemical Toxicology 2022, 45
(5)
, 2169-2184. https://doi.org/10.1080/01480545.2021.1915327
- Yunfang Xiong, Ran Ke, Qingyu Zhang, Wenjun Lan, Wanjun Yuan, Karol Nga Ieng Chan, Tom Roussel, Yifan Jiang, Jing Wu, Shuai Liu, Alice Sze Tsai Wong, Joong Sup Shim, Xuanjun Zhang, Ruiyu Xie, Nelson Dusetti, Juan Iovanna, Nagy Habib, Ling Peng, Leo Tsz On Lee. Small Activating RNA Modulation of the G Protein‐Coupled Receptor for Cancer Treatment. Advanced Science 2022, 9
(26)
https://doi.org/10.1002/advs.202200562
- Rajat Goyal, Hitesh Chopra, Inderbir singh, Kamal Dua, Rupesh K. Gautam. Insights on prospects of nano-siRNA based approaches in treatment of Cancer. Frontiers in Pharmacology 2022, 13 https://doi.org/10.3389/fphar.2022.985670
- V. Karthick, Lok Kumar Shrestha, V. Ganesh Kumar, Pranjali Pranjali, Dinesh Kumar, Aniruddha Pal, Katsuhiko Ariga. Nanoarchitectonics horizons: materials for life sciences. Nanoscale 2022, 14
(30)
, 10630-10647. https://doi.org/10.1039/D2NR02293A
- Xiaowen Guan, Fanqi Meng, Hongwei Tan, Xiaoni Wang, Jingjing Li, Juanjuan Wei, Jin Ouyang, Na Na. Modular and hierarchical self-assembly of siRNAs into supramolecular nanomaterials for soft and homogeneous siRNA loading and precise and visualized intracellular delivery. Chemical Science 2022, 13
(29)
, 8657-8666. https://doi.org/10.1039/D2SC02488H
- Kaili Deng, Dongxue Yang, Yuping Zhou. Nanotechnology-Based siRNA Delivery Systems to Overcome Tumor Immune Evasion in Cancer Immunotherapy. Pharmaceutics 2022, 14
(7)
, 1344. https://doi.org/10.3390/pharmaceutics14071344
- Wen Nie, Jing Chen, Bilan Wang, Xiang Gao. Nonviral vector system for cancer immunogene therapy. MedComm – Biomaterials and Applications 2022, 1
(1)
https://doi.org/10.1002/mba2.10
- Ma. Concepción Peña-Juárez, Omar Rodrigo Guadarrama-Escobar, José Juan Escobar-Chávez. Transdermal Delivery Systems for Biomolecules. Journal of Pharmaceutical Innovation 2022, 17
(2)
, 319-332. https://doi.org/10.1007/s12247-020-09525-2
- Orode UV Aniejurengho, Steve T Meikle, Matteo Santin. Poly(epsilon-lysine) dendrons and nucleic acids complexes for non-viral delivery of bacteriophage DNA into bacterial cells. Proceedings of the Institution of Mechanical Engineers, Part N: Journal of Nanomaterials, Nanoengineering and Nanosystems 2022, 1 , 239779142211022. https://doi.org/10.1177/23977914221102291
- Xin Song, Xuefeng Zhu, Hao Yao, Weili Shang, Cong Du, Wensheng Lu, Minghua Liu, Wei Tian. Topological-skeleton controlled chirality expression of supramolecular hyperbranched and linear polymers. Fundamental Research 2022, 2
(3)
, 422-428. https://doi.org/10.1016/j.fmre.2021.10.006
- Boyang Hu, Ruijie Liu, Qingyue Liu, Zi'an Lin, Yiwei Shi, Jun Li, Lijun Wang, Longjie Li, Xianjin Xiao, Yuzhou Wu. Engineering surface patterns on nanoparticles: new insights into nano-bio interactions. Journal of Materials Chemistry B 2022, 10
(14)
, 2357-2383. https://doi.org/10.1039/D1TB02549J
- Deblin Jana, Yanli Zhao. Strategies for enhancing cancer chemodynamic therapy performance. Exploration 2022, 2
(2)
https://doi.org/10.1002/EXP.20210238
- Chi Ma, Dandan Zhu, Wenyi Lin, Ying Li, Yuanzheng Huang, Huiling Zhu, Mengyun Ye, Yang Wang, Ling Peng, Xiaoxuan Liu. A biodegradable amphiphilic poly(aminoester) dendrimer for safe and effective siRNA delivery. Chemical Communications 2022, 58
(26)
, 4168-4171. https://doi.org/10.1039/D1CC06655B
- Xiuna Jia, Mengmeng Lv, Yunwei Fei, Qing Dong, Hao Wang, Qiong Liu, Dan Li, Jin Wang, Erkang Wang. Facile one-step synthesis of NIR-Responsive siRNA-Inorganic hybrid nanoplatform for imaging-guided photothermal and gene synergistic therapy. Biomaterials 2022, 282 , 121404. https://doi.org/10.1016/j.biomaterials.2022.121404
- Fengjuan Xie, Rongxin Li, Weikang Shu, Liang Zhao, Jingjing Wan. Self-assembly of Peptide dendrimers and their bio-applications in theranostics. Materials Today Bio 2022, 14 , 100239. https://doi.org/10.1016/j.mtbio.2022.100239
- V. Karthick, Katsuhiko Ariga. Nanostructured Biocompatible Materials. 2022, 135-151. https://doi.org/10.1039/9781788019613-00135
- Supusson Pengnam, Samarwadee Plianwong, Boon-ek Yingyongnarongkul, Prasopchai Patrojanasophon, Praneet Opanasopit. Delivery of small interfering RNAs by nanovesicles for cancer therapy. Drug Metabolism and Pharmacokinetics 2022, 42 , 100425. https://doi.org/10.1016/j.dmpk.2021.100425
- Jinho Yoon, Minkyu Shin, Ji-Young Lee, Sang-Nam Lee, Jin-Ha Choi, Jeong-Woo Choi. RNA interference (RNAi)-based plasmonic nanomaterials for cancer diagnosis and therapy. Journal of Controlled Release 2022, 342 , 228-240. https://doi.org/10.1016/j.jconrel.2022.01.012
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
References
This article references 52 other publications.
- 1Fire, A.; Xu, S.; Montgomery, M.; Kostas, S.; Driver, S.; Mello, C. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature 1998, 391, 806– 811, DOI: 10.1038/358881https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXhtlCju74%253D&md5=7bd291ca75d455bbbe1dee41c3362208Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegansFire, Andrew; Xu, SiQun; Montgomery, Mary K.; Kostas, Steven A.; Driver, Samuel E.; Mello, Craig C.Nature (London) (1998), 391 (6669), 806-811CODEN: NATUAS; ISSN:0028-0836. (Macmillan Magazines)Exptl. introduction of RNA into cells can be used in certain biol. systems to interfere with the function of an endogenous gene. Such effects have been proposed to result from a simple antisense mechanism that depends on hybridization between the injected RNA and endogenous mRNA transcripts. RNA interference has been used in the nematode C. elegans to manipulate gene expression. Here we investigate the requirements for structure and delivery of the interfering RNA. To our surprise, we found that double-stranded RNA was substantially more effective at producing interference than was either strand individually. After injection into adult animals, purified single strands had at most a modest effect, whereas double-stranded mixts. caused potent and specific interference. The effects of this interference were evident in both the injected animals and their progeny. Only a few mols. of injected double-stranded RNA were required per affected cell, arguing against stoichiometric interference with endogenous mRNA and suggesting that there could be a catalytic or amplification component in the interference process.
- 2Hannon, G. RNA interference. Nature 2002, 418, 244– 251, DOI: 10.1038/418244a2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XltFGmu7Y%253D&md5=8e218d4b8d0880adfea123dd2e77df11RNA interferenceHannon, Gregory J.Nature (London, United Kingdom) (2002), 418 (6894), 244-251CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)A review. A conserved biol. response to double-stranded RNA, known variously as RNA interference (RNAi) or post-transcriptional gene silencing, mediates resistance to both endogenous parasitic and exogenous pathogenic nucleic acids, and regulates the expression of protein-coding genes. RNAi has been cultivated as a means to manipulate gene expression exptl. and to probe gene function on a whole-genome scale.
- 3Castanotto, D.; Rossi, J. The promises and pitfalls of RNA-interference-based therapeutics. Nature 2009, 457, 426– 433, DOI: 10.1038/nature077583https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXotlOgsA%253D%253D&md5=0574a639adbec5f45553d2248b807fceThe promises and pitfalls of RNA-interference-based therapeuticsCastanotto, Daniela; Rossi, John J.Nature (London, United Kingdom) (2009), 457 (7228), 426-433CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)A review. The discovery that gene expression can be controlled by the Watson-Crick base-pairing of small RNAs with mRNAs contg. complementary sequence - a process known as RNA interference - has markedly advanced our understanding of eukaryotic gene regulation and function. The ability of short RNA sequences to modulate gene expression has provided a powerful tool with which to study gene function and is set to revolutionize the treatment of disease. Remarkably, despite being just one decade from its discovery, the phenomenon is already being used therapeutically in human clin. trials, and biotechnol. companies that focus on RNA-interference-based therapeutics are already publicly traded.
- 4Haussecker, D.; Kay, M. RNA interference. Drugging RNAi. Science 2015, 347, 1069– 1070, DOI: 10.1126/science.1252967There is no corresponding record for this reference.
- 5Wittrup, A.; Lieberman, J. Knocking down disease: a progress report on siRNA therapeutics. Nat. Rev. Genet. 2015, 16, 543– 552, DOI: 10.1038/nrg39785https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtlKnsLnF&md5=6475cb83ab2f8ab205e93d26b89952acKnocking down disease: a progress report on siRNA therapeuticsWittrup, Anders; Lieberman, JudyNature Reviews Genetics (2015), 16 (9), 543-552CODEN: NRGAAM; ISSN:1471-0056. (Nature Publishing Group)A review. Small interfering RNAs (siRNAs), which downregulate gene expression guided by sequence complementarity, can be used therapeutically to block the synthesis of disease-causing proteins. The main obstacle to siRNA drugs - their delivery into the target cell cytosol - has been overcome to allow suppression of liver gene expression. Here, we review the results of recent clin. trials of siRNA therapeutics, which show efficient and durable gene knockdown in the liver, with signs of promising clin. outcomes and little toxicity. We also discuss the barriers to more widespread applications that target tissues besides the liver and the most promising avenues to overcome them.
- 6Ledford, H. Gene-silencing technology gets first drug approval after 20-year wait. Nature 2018, 560, 291– 292, DOI: 10.1038/d41586-018-05867-76https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsFahtrvO&md5=39e63d6837dbb66e4d057a96d615fa84Gene-silencing technology gets first drug approval after 20-year waitLedford, HeidiNature (London, United Kingdom) (2018), 560 (7718), 291-292CODEN: NATUAS; ISSN:0028-0836. (Nature Research)The US Food and Drug Administration's decision breathes new life into RNA-interference therapies.
- 7Whitehead, K.; Langer, R.; Anderson, D. Knocking down barriers: advances in siRNA delivery. Nat. Rev. Drug Discovery 2009, 8, 129– 138, DOI: 10.1038/nrd27427https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtFOqt70%253D&md5=f3eecb4c59f7f3b233c3cce4823e7c72Knocking down barriers: advances in siRNA deliveryWhitehead, Kathryn A.; Langer, Robert; Anderson, Daniel G.Nature Reviews Drug Discovery (2009), 8 (2), 129-138CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. In the 10 years that have passed since the Nobel prize-winning discovery of RNA interference (RNAi), billions of dollars have been invested in the therapeutic application of gene silencing in humans. Today, there are promising data from ongoing clin. trials for the treatment of age-related macular degeneration and respiratory syncytial virus. Despite these early successes, however, the widespread use of RNAi therapeutics for disease prevention and treatment requires the development of clin. suitable, safe and effective drug delivery vehicles. Here, we provide an update on the progress of RNAi therapeutics and highlight novel synthetic materials for the encapsulation and intracellular delivery of nucleic acids.
- 8Kanasty, R.; Dorkin, J.; Vegas, A.; Anderson, D. Delivery materials for siRNA therapeutics. Nat. Mater. 2013, 12, 967– 977, DOI: 10.1038/nmat37658https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhs1Grt7rM&md5=fdc0612869582a97b13673c0ca9c0cf5Delivery materials for siRNA therapeuticsKanasty, Rosemary; Dorkin, Joseph Robert; Vegas, Arturo; Anderson, DanielNature Materials (2013), 12 (11), 967-977CODEN: NMAACR; ISSN:1476-1122. (Nature Publishing Group)A review. RNA interference (RNAi) has broad potential as a therapeutic to reversibly silence any gene. To achieve the clin. potential of RNAi, delivery materials are required to transport short interfering RNA (siRNA) to the site of action in the cells of target tissues. This Review provides an introduction to the biol. challenges that siRNA delivery materials aim to overcome, as well as a discussion of the way that the most effective and clin. advanced classes of siRNA delivery systems, including lipid nanoparticles and siRNA conjugates, are designed to surmount these challenges. The systems that we discuss are diverse in their approaches to the delivery problem, and provide valuable insight to guide the design of future siRNA delivery materials.
- 9Yin, H.; Kanasty, R.; Eltoukhy, A.; Vegas, A.; Dorkin, J.; Anderson, D. Non-viral vectors for gene-based therapy. Nat. Rev. Genet. 2014, 15, 541– 555, DOI: 10.1038/nrg37639https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtFGqsbfF&md5=eb763f2d3fe0b8cde33599f802f1db1aNon-viral vectors for gene-based therapyYin, Hao; Kanasty, Rosemary L.; Eltoukhy, Ahmed A.; Vegas, Arturo J.; Dorkin, J. Robert; Anderson, Daniel G.Nature Reviews Genetics (2014), 15 (8), 541-555CODEN: NRGAAM; ISSN:1471-0056. (Nature Publishing Group)Gene-based therapy is the intentional modulation of gene expression in specific cells to treat pathol. conditions. This modulation is accomplished by introducing exogenous nucleic acids such as DNA, mRNA, small interfering RNA (siRNA), microRNA (miRNA) or antisense oligonucleotides. Given the large size and the neg. charge of these macromols., their delivery is typically mediated by carriers or vectors. In this Review, we introduce the biol. barriers to gene delivery in vivo and discuss recent advances in material sciences, nanotechnol. and nucleic acid chem. that have yielded promising non-viral delivery systems, some of which are currently undergoing testing in clin. trials. The diversity of these systems highlights the recent progress of gene-based therapy using non-viral approaches.
- 10Juliano, R. The delivery of therapeutic oligonucleotides. Nucleic Acids Res. 2016, 44, 6518– 6548, DOI: 10.1093/nar/gkw23610https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC28bgsFWluw%253D%253D&md5=22ea7e38834cf7494c35687df25f23a8The delivery of therapeutic oligonucleotidesJuliano Rudolph LNucleic acids research (2016), 44 (14), 6518-48 ISSN:.The oligonucleotide therapeutics field has seen remarkable progress over the last few years with the approval of the first antisense drug and with promising developments in late stage clinical trials using siRNA or splice switching oligonucleotides. However, effective delivery of oligonucleotides to their intracellular sites of action remains a major issue. This review will describe the biological basis of oligonucleotide delivery including the nature of various tissue barriers and the mechanisms of cellular uptake and intracellular trafficking of oligonucleotides. It will then examine a variety of current approaches for enhancing the delivery of oligonucleotides. This includes molecular scale targeted ligand-oligonucleotide conjugates, lipid- and polymer-based nanoparticles, antibody conjugates and small molecules that improve oligonucleotide delivery. The merits and liabilities of these approaches will be discussed in the context of the underlying basic biology.
- 11Pack, D.; Hoffman, A.; Pun, S.; Stayton, P. Design and development of polymers for gene delivery. Nat. Rev. Drug Discovery 2005, 4, 581– 593, DOI: 10.1038/nrd177511https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXmsFGjtbo%253D&md5=ed3dcd52a8b297ee3d0c1cbb7d5ae43bDesign and development of polymers for gene deliveryPack, Daniel W.; Hoffman, Allan S.; Pun, Suzie; Stayton, Patrick S.Nature Reviews Drug Discovery (2005), 4 (7), 581-593CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. The lack of safe and efficient gene-delivery methods is a limiting obstacle to human gene therapy. Synthetic gene-delivery agents, although safer than recombinant viruses, generally do not possess the required efficacy. In recent years, a variety of effective polymers have been designed specifically for gene delivery, and much has been learned about their structure-function relationships. With the growing understanding of polymer gene-delivery mechanisms and continued efforts of creative polymer chemists, it is likely that polymer-based gene-delivery systems will become an important tool for human gene therapy.
- 12Lachelt, U.; Wagner, E. Nucleic acid therapeutics using polyplexes: a journey of 50 years (and beyond). Chem. Rev. 2015, 115, 11043– 11078, DOI: 10.1021/cr500679312https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2MjktVagtg%253D%253D&md5=f317ecbf3554fbcb4ebae59ef0d3d48fNucleic Acid Therapeutics Using Polyplexes: A Journey of 50 Years (and Beyond)Lachelt Ulrich; Wagner Ernst; Lachelt Ulrich; Wagner ErnstChemical reviews (2015), 115 (19), 11043-78 ISSN:.There is no expanded citation for this reference.
- 13Tseng, Y.; Mozumdar, S.; Huang, L. Lipid-based systemic delivery of siRNA. Adv. Drug Delivery Rev. 2009, 61, 721– 731, DOI: 10.1016/j.addr.2009.03.00313https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXmsFyns7Y%253D&md5=535a7456fd5f7ab5503a19dfa7a66f7dLipid-based systemic delivery of siRNATseng, Yu-Cheng; Mozumdar, Subho; Huang, LeafAdvanced Drug Delivery Reviews (2009), 61 (9), 721-731CODEN: ADDREP; ISSN:0169-409X. (Elsevier B.V.)A review. RNAi technol. has brought a new category of treatments for various diseases including genetic diseases, viral diseases, and cancer. Despite the great versatility of RNAi that can down regulate almost any protein in the cells, the delicate and precise machinery used for silencing is the same. The major challenge indeed for RNAi-based therapy is the delivery system. In this review, we start with the uniqueness and mechanism of RNAi machinery and the utility of RNAi in therapeutics. Then we discuss the challenges in systemic siRNA delivery by dividing them into two categories-kinetic and phys. barriers. At the end, we discuss different strategies to overcome these barriers, esp. focusing on the step of endosome escape. Toxicity issues and current successful examples for lipid-based delivery are also included in the review.
- 14Wagner, E. Polymers for siRNA delivery: inspired by viruses to be targeted, dynamic, and precise. Acc. Chem. Res. 2012, 45, 1005– 1013, DOI: 10.1021/ar200223214https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhs1GltrfO&md5=7b0c369312a3d10cb5a8f7fe5dd21f82Polymers for siRNA Delivery: Inspired by Viruses to be Targeted, Dynamic, and PreciseWagner, ErnstAccounts of Chemical Research (2012), 45 (7), 1005-1013CODEN: ACHRE4; ISSN:0001-4842. (American Chemical Society)A review. Synthetic small interfering RNA (siRNA) presents an exciting novel medical opportunity. Although researchers agree that siRNA could have a great therapeutic impact, the required extracellular and intracellular delivery of these mols. into the disease-assocd. target cells presents the primary roadblock for the broader translation of these mols. into medicines. Thus, the design of adequate delivery technologies has utmost importance. Viruses are natural masterpieces of nucleic acid delivery and present chemists and drug delivery experts with a template for the design of artificial carriers for synthetic nucleic acids such as siRNA. They have been developed into gene vectors and have provided convincing successes in gene therapy. Optimized by biol. evolution, viruses are programmed to be dynamic and bioresponsive as they enter living cells, and they carry out their functions in a precisely defined sequence. However, because they are synthesized within living cells and with naturally available nucleotides and amino acids, the chem. of viruses is limited. With the use of diverse synthetic mols. and macromols., chemists can provide delivery solns. beyond the scope of the natural evolution of viruses. This Account describes the design and synthesis of "synthetic siRNA viruses." These structures contain elements that mimic the delivery functions of viral particles and surface domains that shield against undesired biol. interactions and enable specific host cell receptor binding through the presentation of multiple targeting ligands. For example, cationic polymers can reversibly package one or more siRNA mols. into nanoparticle cores to protect them against a degradative bioenvironment. After internalization by receptor-mediated endocytosis into the acidifying endosomes of cells, synthetic siRNA can escape from these vesicles through the activation of membrane-disruption domains as viruses do and reach the cytoplasm, the location of RNA interference. This multistep task presents an attractive challenge for chemists. Similar to the design of prodrugs, the functional domains of these systems have to be activated in a dynamic mode, triggered by conformational changes or bond cleavages in the relevant microenvironment such as the acidic endosome or disulfide-reducing cytoplasm. These chem. analogs of viral domains are often synthetically simpler and more easily accessible mols. than viral proteins. Their precise assembly into multifunctional macromol. and supramol. structures is facilitated by improved anal. techniques, precise orthogonal conjugation chemistries, and sequence-defined polymer syntheses. The chem. evolution of microdomains using chem. libraries and macromol. and supramol. evolution could provide key strategies for optimizing siRNA carriers to selected medical indications.
- 15Wittrup, A.; Ai, A.; Liu, X.; Hamar, P.; Trifonova, R.; Charisse, K.; Manoharan, M.; Kirchhausen, T.; Lieberman, J. Visualizing lipid-formulated siRNA release from endosomes and target gene knockdown. Nat. Biotechnol. 2015, 33, 870– 876, DOI: 10.1038/nbt.329815https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtF2murjF&md5=1e896c5372fff738d373ef1deb2ac412Visualizing lipid-formulated siRNA release from endosomes and target gene knockdownWittrup, Anders; Ai, Angela; Liu, Xing; Hamar, Peter; Trifonova, Radiana; Charisse, Klaus; Manoharan, Muthiah; Kirchhausen, Tomas; Lieberman, JudyNature Biotechnology (2015), 33 (8), 870-876CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)A central hurdle in developing small interfering RNAs (siRNAs) as therapeutics is the inefficiency of their delivery across the plasma and endosomal membranes to the cytosol, where they interact with the RNA interference machinery. With the aim of improving endosomal release, a poorly understood and inefficient process, we studied the uptake and cytosolic release of siRNAs, formulated in lipoplexes or lipid nanoparticles, by live-cell imaging and correlated it with knockdown of a target GFP reporter. siRNA release occurred invariably from maturing endosomes within ∼5-15 min of endocytosis. Cytosolic galectins immediately recognized the damaged endosome and targeted it for autophagy. However, inhibiting autophagy did not enhance cytosolic siRNA release. Gene knockdown occurred within a few hours of release and required <2,000 copies of cytosolic siRNAs. The ability to detect cytosolic release of siRNAs and understand how it is regulated will facilitate the development of rational strategies for improving the cytosolic delivery of candidate drugs.
- 16Lee, C.; MacKay, J.; Frechet, J.; Szoka, F. Designing dendrimers for biological applications. Nat. Biotechnol. 2005, 23, 1517– 1526, DOI: 10.1038/nbt117116https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXht1yisrnE&md5=d1306ae17979250c01f2fb51fa76a080Designing dendrimers for biological applicationsLee, Cameron C.; MacKay, John A.; Frechet, Jean M. J.; Szoka, Francis C.Nature Biotechnology (2005), 23 (12), 1517-1526CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)A review. Dendrimers are branched, synthetic polymers with layered architectures that show promise in several biomedical applications. By regulating dendrimer synthesis, it is possible to precisely manipulate both their mol. wt. and chem. compn., thereby allowing predictable tuning of their biocompatibility and pharmacokinetics. Advances in our understanding of the role of mol. wt. and architecture on the in vivo behavior of dendrimers, together with recent progress in the design of biodegradable chemistries, has enabled the application of these branched polymers as anti-viral drugs, tissue repair scaffolds, targeted carriers of chemotherapeutics and optical oxygen sensors. Before such products can reach the market, however, the field must not only address the cost of manuf. and quality control of pharmaceutical-grade materials, but also assess the long-term human and environmental health consequences of dendrimer exposure in vivo.
- 17Khandare, J.; Calderon, M.; Dagia, N.; Haag, R. Multifunctional dendritic polymers in nanomedicine: opportunities and challenges. Chem. Soc. Rev. 2012, 41, 2824– 2848, DOI: 10.1039/C1CS15242D17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xjs1CksLg%253D&md5=2096b686d6657325955ec41bbbc006f2Multifunctional dendritic polymers in nanomedicine: opportunities and challengesKhandare, Jayant; Calderon, Marcelo; Dagia, Nilesh M.; Haag, RainerChemical Society Reviews (2012), 41 (7), 2824-2848CODEN: CSRVBR; ISSN:0306-0012. (Royal Society of Chemistry)A review. Nanotechnol. has resulted in materials that have greatly improved the effectiveness of drug delivery because of their ability to control matter on the nanoscale. Advanced forms of nanomedicine have been synthesized for better pharmacokinetics to obtain higher efficacy, less systemic toxicity, and better targeting. These criteria have long been the goal in nanomedicine, in particular, for systemic applications in oncol. disorders. Now, the "holy grail" in nanomedicine is to design and synthesize new advanced macromol. nanocarriers and to translate them from lab to clinic. This review describes the current and future perspectives of nanomedicine with particular emphasis on the clin. targets in cancer and inflammation. The advanced forms of liposomes and polyethylene glycol (PEG) based nanocarriers, as well as dendritic polymer conjugates will be discussed with particular attention paid to designs, synthetic strategies, and chem. pathways. In this crit. review, we also report on the current status and perspective of dendritic polymer nanoconjugate platforms (e.g. polyamidoamine dendrimers and dendritic polyglycerols) for cellular localization and targeting of specific tissues (192 refs.).
- 18Liu, X.; Rocchi, P.; Peng, L. Dendrimers as non-viral vectors for siRNA delivery. New J. Chem. 2012, 36, 256– 263, DOI: 10.1039/C1NJ20408DThere is no corresponding record for this reference.
- 19Liu, X.; Liu, C.; Catapano, C.; Peng, L.; Zhou, J.; Rocchi, P. Structurally flexible triethanolamine-core poly(amidoamine) dendrimers as effective nanovectors to deliver RNAi-based therapeutics. Biotechnol. Adv. 2014, 32, 844– 852, DOI: 10.1016/j.biotechadv.2013.08.001There is no corresponding record for this reference.
- 20Reebye, V.; Sætrom, P.; Mintz, P.; Huang, K.; Swiderski, P.; Peng, L.; Liu, C.; Liu, X.; Lindkaer-Jensen, S.; Zacharoulis, D. Novel RNA oligonucleotide improves liver function and inhibits liver carcinogenesis in vivo. Hepatology 2014, 59, 216– 227, DOI: 10.1002/hep.26669There is no corresponding record for this reference.
- 21Yu, T.; Liu, X.; Bolcato-Bellemin, A.; Wang, Y.; Liu, C.; Erbacher, P.; Qu, F.; Rocchi, P.; Behr, J.; Peng, L. An amphiphilic dendrimer for effective delivery of small interfering RNA and gene silencing in vitro and in vivo. Angew. Chem., Int. Ed. 2012, 51, 8478– 8484, DOI: 10.1002/anie.20120392021https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtVyjs7rO&md5=176386f529e80f5778008b8aba3c2714An Amphiphilic Dendrimer for Effective Delivery of Small Interfering RNA and Gene Silencing In Vitro and In VivoYu, Tianzhu; Liu, Xiaoxuan; Bolcato-Bellemin, Anne-Laure; Wang, Yang; Liu, Cheng; Erbacher, Patrick; Qu, Fanqi; Rocchi, Palma; Behr, Jean-Paul; Peng, LingAngewandte Chemie, International Edition (2012), 51 (34), 8478-8484, S8478/1-S8478/33CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)An amphiphilic dendrimer bearing a hydrophobic alkyl chain and hydrophilic poly(amidoamine) dendrons is able to combine the advantageous features of lipid and dendrimers vectors to deliver a heat shock protein 27 siRNA and produce potent gene silencing and anticancer activity in vitro and in vivo in a prostate cancer model. This dendrimer can be used alternatively for treating various diseases.
- 22Liu, X.; Zhou, J.; Yu, T.; Chen, C.; Cheng, Q.; Sengupta, K.; Huang, Y.; Li, H.; Liu, C.; Wang, Y.; Posocco, P.; Wang, M.; Cui, Q.; Giorgio, S.; Fermeglia, M.; Qu, F.; Pricl, S.; Shi, Y.; Liang, Z.; Rocchi, P.; Rossi, J.; Peng, L. Adaptive amphiphilic dendrimer-based nanoassemblies as robust and versatile siRNA delivery systems. Angew. Chem., Int. Ed. 2014, 53, 11822– 11827, DOI: 10.1002/anie.20140676422https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhsFCisrvF&md5=5cb14bd9e508eb76c4b008b6752ed806Adaptive Amphiphilic Dendrimer-Based Nanoassemblies as Robust and Versatile siRNA Delivery SystemsLiu, Xiaoxuan; Zhou, Jiehua; Yu, Tianzhu; Chen, Chao; Cheng, Qiang; Sengupta, Kheya; Huang, Yuanyu; Li, Haitang; Liu, Cheng; Wang, Yang; Posocco, Paola; Wang, Menghua; Cui, Qi; Giorgio, Suzanne; Fermeglia, Maurizio; Qu, Fanqi; Pricl, Sabrina; Shi, Yanhong; Liang, Zicai; Rocchi, Palma; Rossi, John J.; Peng, LingAngewandte Chemie, International Edition (2014), 53 (44), 11822-11827CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)SiRNA delivery remains a major challenge in RNAi-based therapy. Here, we report for the first time that an amphiphilic dendrimer is able to self-assemble into adaptive supramol. assemblies upon interaction with siRNA, and effectively delivers siRNAs to various cell lines, including human primary and stem cells, thereby outperforming the currently available nonviral vectors. In addn., this amphiphilic dendrimer is able to harness the advantageous features of both polymer and lipid vectors and hence promotes effective siRNA delivery. Our study demonstrates for the first time that dendrimer-based adaptive supramol. assemblies represent novel and versatile means for functional siRNA delivery, heralding a new age of dendrimer-based self-assembled drug delivery in biomedical applications.
- 23Chen, C.; Posocco, P.; Liu, X.; Cheng, Q.; Laurini, E.; Zhou, J.; Liu, C.; Wang, Y.; Tang, J.; Col, V.; Yu, T.; Giorgio, S.; Fermeglia, M.; Qu, F.; Liang, Z.; Rossi, J.; Liu, M.; Rocchi, P.; Pricl, S.; Peng, L. siRNA delivery: mastering dendrimer self-assembly for efficient siRNA delivery: from conceptual design to in vivo efficient gene silencing. Small 2016, 12, 3667– 3676, DOI: 10.1002/smll.201503866There is no corresponding record for this reference.
- 24Liu, X.; Wang, Y.; Chen, C.; Tintaru, A.; Cao, Y.; Liu, J.; Ziarelli, F.; Tang, J.; Guo, H.; Rosas, R.; Giorgio, S.; Charle, L.; Rocchi, P.; Peng, L. A fluorinated bola-amphiphilic dendrimer for on-demand delivery of siRNA, via specific response to reactive oxygen species. Adv. Funct. Mater. 2016, 26, 8594– 8603, DOI: 10.1002/adfm.20160419224https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhslGitLjE&md5=931ea4546e4a792fc32c41dd25b69a8aA Fluorinated Bola-Amphiphilic Dendrimer for On-Demand Delivery of siRNA, via Specific Response to Reactive Oxygen SpeciesLiu, Xiaoxuan; Wang, Yang; Chen, Chao; Tintaru, Aura; Cao, Yu; Liu, Juan; Ziarelli, Fabio; Tang, Jingjie; Guo, Hongbo; Rosas, Roseline; Giorgio, Suzanne; Charles, Laurence; Rocchi, Palma; Peng, LingAdvanced Functional Materials (2016), 26 (47), 8594-8603CODEN: AFMDC6; ISSN:1616-301X. (Wiley-VCH Verlag GmbH & Co. KGaA)Functional materials capable of responding to stimuli intrinsic to diseases are extremely important for specific drug delivery at the disease site. However, developing on-demand stimulus-responsive vectors for targeted delivery is highly challenging. Here, a stimulus-responsive fluorinated bola-amphiphilic dendrimer is reported for on-demand delivery of small interfering RNA (siRNA) in response to the characteristic high level of reactive oxygen species (ROS) in cancer cells. This dendrimer bears a ROS-sensitive thioacetal in the hydrophobic core and pos. charged poly(amidoamine) dendrons at the terminals, capable of interacting and compacting the neg. charged siRNA into nanoparticles to protect the siRNA and promote cellular uptake. The ROS-sensitive feature of this dendrimer boosts specific and efficient disassembly of the siRNA/vector complexes in ROS-rich cancer cells for effective siRNA delivery and gene silencing. Moreover, the fluorine tags in the vector enable 19F-NMR anal. of the ROS-responsive delivery process. In addn., this ingenious and distinct bola-amphiphilic dendrimer is also able to combine the advantageous delivery features of both lipid and dendrimer vectors. Therefore, it represents an innovative on-demand stimulus-responsive delivery platform.
- 25Percec, V.; Wilson, D.; Leowanawat, P.; Wilson, C.; Hughes, A.; Kaucher, M.; Hammer, D.; Levine, D.; Kim, A.; Bates, F. Self-assembly of Janus dendrimers into uniform dendrimersomes and other complex architectures. Science 2010, 328, 1009– 1014, DOI: 10.1126/science.118554725https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXmtFCnsLo%253D&md5=f27fa5e6ae26f177e85c36ca5e46047eSelf-Assembly of Janus Dendrimers into Uniform Dendrimersomes and Other Complex ArchitecturesPercec, Virgil; Wilson, Daniela A.; Leowanawat, Pawaret; Wilson, Christopher J.; Hughes, Andrew D.; Kaucher, Mark S.; Hammer, Daniel A.; Levine, Dalia H.; Kim, Anthony J.; Bates, Frank S.; Davis, Kevin P.; Lodge, Timothy P.; Klein, Michael L.; De Vane, Russell H.; Aqad, Emad; Rosen, Brad M.; Argintaru, Andreea O.; Sienkowska, Monika J.; Rissanen, Kari; Nummelin, Sami; Ropponen, JarmoScience (Washington, DC, United States) (2010), 328 (5981), 1009-1014CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Self-assembled nanostructures obtained from natural and synthetic amphiphiles serve as mimics of biol. membranes and enable the delivery of drugs, proteins, genes, and imaging agents. Yet the precise mol. arrangements demanded by these functions are difficult to achieve. Libraries of amphiphilic Janus dendrimers, prepd. by facile coupling of tailored hydrophilic and hydrophobic branched segments, have been screened by cryogenic transmission electron microscopy, revealing a rich palette of morphologies in water, including vesicles, denoted dendrimersomes, cubosomes, disks, tubular vesicles, and helical ribbons. Dendrimersomes marry the stability and mech. strength obtainable from polymersomes with the biol. function of stabilized phospholipid liposomes, plus superior uniformity of size, ease of formation, and chem. functionalization. This modular synthesis strategy provides access to systematic tuning of mol. structure and of self-assembled architecture.
- 26Sherman, S.; Xiao, Q.; Percec, V. Mimicking complex biological membranes and their programmable glycan ligands with dendrimersomes and glycodendrimersomes. Chem. Rev. 2017, 117, 6538– 6631, DOI: 10.1021/acs.chemrev.7b0009726https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXmtVyqur8%253D&md5=7448c0318ceb9b46a0b12963822a166bMimicking Complex Biological Membranes and Their Programmable Glycan Ligands with Dendrimersomes and GlycodendrimersomesSherman, Samuel E.; Xiao, Qi; Percec, VirgilChemical Reviews (Washington, DC, United States) (2017), 117 (9), 6538-6631CODEN: CHREAY; ISSN:0009-2665. (American Chemical Society)A review. Synthetic vesicles have been assembled and co-assembled from phospholipids, their modified versions, and other single amphiphiles into liposomes, and from block copolymers into polymersomes. Their time-consuming synthesis and prepn. as stable, monodisperse, and biocompatible liposomes and polymersomes called for the elaboration of new synthetic methodologies. Amphiphilic Janus dendrimers (JDs) and glycodendrimers (JGDs) represent the most recent self-assembling amphiphiles capable of forming monodisperse, stable, and multifunctional unilamellar and multilamellar onion-like vesicles denoted dendrimersomes (DSs) and glycodendrimersomes (GDSs), dendrimercubosomes (DCs), glycodendrimercubosomes (GDCs), and other complex architectures. Amphiphilic JDs consist of hydrophobic dendrons connected to hydrophilic dendrons and can be thought of as monodisperse oligomers of a single amphiphile. They can be functionalized with a variety of mols. such as dyes, and, in the case of JGDs, with carbohydrates. Their iterative modular synthesis provides efficient access to sequence control at the mol. level, resulting in topologies with specific epitope sequence and d. DSs, GDSs, and other architectures from JDs and JGDs serve as powerful tools for mimicking biol. membranes and for biomedical applications such as targeted drug and gene delivery and theranostics. This Review covers all aspects of the synthesis of JDs and JGDs and their biol. activity and applications after assembly in aq. media.
- 27Bertrand, N.; Wu, J.; Xu, X.; Kamaly, N.; Farokhzad, O. Cancer nanotechnology: the impact of passive and active targeting in the era of modern cancer biology. Adv. Drug Delivery Rev. 2014, 66, 2– 25, DOI: 10.1016/j.addr.2013.11.00927https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXisV2hug%253D%253D&md5=0980da1a1b75cde64cc1cf82184e01b7Cancer nanotechnology: The impact of passive and active targeting in the era of modern cancer biologyBertrand, Nicolas; Wu, Jun; Xu, Xiaoyang; Kamaly, Nazila; Farokhzad, Omid C.Advanced Drug Delivery Reviews (2014), 66 (), 2-25CODEN: ADDREP; ISSN:0169-409X. (Elsevier B.V.)Cancer nanotherapeutics are progressing at a steady rate; research and development in the field has experienced an exponential growth since early 2000's. The path to the commercialization of oncol. drugs is long and carries significant risk; however, there is considerable excitement that nanoparticle technologies may contribute to the success of cancer drug development. The pace at which pharmaceutical companies have formed partnerships to use proprietary nanoparticle technologies has considerably accelerated. It is now recognized that by enhancing the efficacy and/or tolerability of new drug candidates, nanotechnol. can meaningfully contribute to create differentiated products and improve clin. outcome. This review describes the lessons learned since the commercialization of the first-generation nanomedicines including DOXIL and Abraxane. It explores our current understanding of targeted and non-targeted nanoparticles that are under various stages of development, including BIND-014 and MM-398. It highlights the opportunities and challenges faced by nanomedicines in contemporary oncol., where personalized medicine is increasingly the mainstay of cancer therapy. We revisit the fundamental concepts of enhanced permeability and retention effect (EPR) and explore the mechanisms proposed to enhance preferential "retention" in the tumor, whether using active targeting of nanoparticles, binding of drugs to their tumoral targets or the presence of tumor assocd. macrophages. The overall objective of this review is to enhance our understanding in the design and development of therapeutic nanoparticles for treatment of cancers.
- 28Dai, L.; Liu, J.; Luo, Z.; Li, M.; Cai, K. Tumor therapy: targeted drug delivery system. J. Mater. Chem. B 2016, 4, 6758– 6772, DOI: 10.1039/C6TB01743F28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsVWktr3F&md5=43534ec2f361c7181059c074132608d4Tumor therapy: targeted drug delivery systemsDai, Liangliang; Liu, Junjie; Luo, Zhong; Li, Menghuan; Cai, KaiyongJournal of Materials Chemistry B: Materials for Biology and Medicine (2016), 4 (42), 6758-6772CODEN: JMCBDV; ISSN:2050-7518. (Royal Society of Chemistry)Recently, targeted drug delivery systems (TDDSs) have been extensively studied as a promising therapeutic for tumor therapy. In this review, we investigate the typical targeting mechanisms of TDDSs, covering both passively and actively targeting DDSs for tumor therapy. We highlight the popular active targeting strategies for different sites of action, including tumor cytomembrane or various organelles. Finally, we present some recent representative TDDSs that are under testing in preclin./clin. trials and have shown excellent clin. potential as the alternate treatment strategy for tumor therapy. Although TDDSs are proving to be promising therapeutic nanoplatforms for tumor therapy, extended investigations should be considered in the landscape for highly efficient tumor therapy with good biosafety.
- 29Davis, M.; Zuckerman, J.; Choi, C.; Seligson, D.; Tolcher, A.; Alabi, C.; Yen, Y.; Heidel, J.; Ribas, A. Evidence of RNAi in humans from systemically administered siRNA via targeted nanoparticles. Nature 2010, 464, 1067– 1070, DOI: 10.1038/nature0895629https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXjs1ehur8%253D&md5=b49a61e3acc1245274c2314fb6bcc99aEvidence of RNAi in humans from systemically administered siRNA via targeted nanoparticlesDavis, Mark E.; Zuckerman, Jonathan E.; Choi, Chung-Hang J.; Seligson, David; Tolcher, Anthony; Alabi, Christopher A.; Yen, Yun; Heidel, Jeremy D.; Ribas, AntoniNature (London, United Kingdom) (2010), 464 (7291), 1067-1070CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Therapeutics that are designed to engage RNA interference (RNAi) pathways have the potential to provide new, major ways of imparting therapy to patients. Long, double-stranded RNAs were first shown to mediate RNAi in Caenorhabditis elegans, and the potential use of RNAi for human therapy has been demonstrated by the finding that small interfering RNAs (siRNAs; approx. 21-base-pair double-stranded RNA) can elicit RNAi in mammalian cells without producing an interferon response. We are at present conducting the first in-human phase I clin. trial involving the systemic administration of siRNA to patients with solid cancers using a targeted, nanoparticle delivery system. Here we provide evidence of inducing an RNAi mechanism of action in a human from the delivered siRNA. Tumor biopsies from melanoma patients obtained after treatment show the presence of intracellularly localized nanoparticles in amts. that correlate with dose levels of the nanoparticles administered (this is, to our knowledge, a first for systemically delivered nanoparticles of any kind). Furthermore, a redn. was found in both the specific mRNA (M2 subunit of ribonucleotide reductase (RRM2)) and the protein (RRM2) levels when compared to pre-dosing tissue. Most notably, we detect the presence of an mRNA fragment that demonstrates that siRNA-mediated mRNA cleavage occurs specifically at the site predicted for an RNAi mechanism from a patient who received the highest dose of the nanoparticles. Together, these data demonstrate that siRNA administered systemically to a human can produce a specific gene inhibition (redn. in mRNA and protein) by an RNAi mechanism of action.
- 30Sugahara, K.; Teesalu, T.; Karmali, P.; Kotamraju, V.; Agemy, L.; Greenwald, D.; Ruoslahti, E. Co-administration of a tumor-penetrating peptide enhances the efficacy of cancer drugs. Science 2010, 328, 1031– 1035, DOI: 10.1126/science.1183057There is no corresponding record for this reference.
- 31Wong, P.; Tang, K.; Coulter, A.; Tan, S.; Baker, J.; Choi, S. Multivalent dendrimer vectors with DNA intercalation motifs for gene delivery. Biomacromolecules 2014, 15, 4134– 4415, DOI: 10.1021/bm501169s31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhs1yisr7K&md5=8c21541c6d33bbcad02f41564d5fe0d6Multivalent Dendrimer Vectors with DNA Intercalation Motifs for Gene DeliveryWong, Pamela T.; Tang, Kenny; Coulter, Alexa; Tang, Shengzhuang; Baker, James R.; Choi, Seok KiBiomacromolecules (2014), 15 (11), 4134-4145CODEN: BOMAF6; ISSN:1525-7797. (American Chemical Society)Poly(amido amine) (PAMAM) dendrimers constitute an important class of nonviral, cationic vectors in gene delivery. Here we report on a new concept for dendrimer vector design based on the incorporation of dual binding motifs: DNA intercalation, and receptor recognition for targeted delivery. We prepd. a series of dendrimer conjugates derived from a fifth generation (G5) PAMAM dendrimer, each conjugated with multiple folate (FA) or riboflavin (RF) ligands for cell receptor targeting, and with 3,8-diamino-6-phenylphenanthridinium ("DAPP")-derived ligands for anchoring a DNA payload. Polyplexes of each dendrimer with calf thymus dsDNA were made and characterized by surface plasmon resonance (SPR) spectroscopy, dynamic light scattering (DLS) and zeta potential measurement. These studies provided evidence supporting polyplex formation based on the observation of tight DNA-dendrimer adhesion, and changes in particle size and surface charge upon coincubation. Further SPR studies to investigate the adhesion of the polyplex to a model surface immobilized with folate binding protein (FBP), demonstrated that the DNA payload has only a minimal effect on the receptor binding activity of the polyplex: KD = 0.22 nM for G5(FA)(DAPP) vs. 0.98 nM for its polyplex. Finally, we performed in vitro transfection assays to det. the efficiency of conjugate mediated delivery of a luciferase-encoding plasmid into the KB cancer cell line and showed that RF-conjugated dendrimers were 1 to 2 orders of magnitude more effective in enhancing luciferase gene transfection than a plasmid only control. In summary, this study serves as a proof of concept for DNA-ligand intercalation as a motif in the design of multivalent dendrimer vectors for targeted gene delivery.
- 32Sugahara, K.; Teesalu, T.; Karmali, P.; Kotamraju, V.; Agemy, L.; Girard, O.; Hanahan, D.; Mattrey, R.; Ruoslahti, E. Tissue-penetrating delivery of compounds and nanoparticles into tumors. Cancer Cell 2009, 16, 510– 520, DOI: 10.1016/j.ccr.2009.10.01332https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXht1eqsbY%253D&md5=f42c01e8e751526318903d28d70ce4c4Tissue-penetrating delivery of compounds and nanoparticles into tumorsSugahara, Kazuki N.; Teesalu, Tambet; Karmali, Priya Prakash; Kotamraju, Venkata Ramana; Agemy, Lilach; Girard, Olivier M.; Hanahan, Douglas; Mattrey, Robert F.; Ruoslahti, ErkkiCancer Cell (2009), 16 (6), 510-520CODEN: CCAECI; ISSN:1535-6108. (Cell Press)Poor penetration of drugs into tumors is a major obstacle in tumor treatment. We describe a strategy for peptide-mediated delivery of compds. deep into the tumor parenchyma that uses a tumor-homing peptide, iRGD (CRGDK/RGPD/EC). I.v. injected compds. coupled to iRGD bound to tumor vessels and spread into the extravascular tumor parenchyma, whereas conventional RGD peptides only delivered the cargo to the blood vessels. IRGD homes to tumors through a three-step process: the RGD motif mediates binding to αv integrins on tumor endothelium and a proteolytic cleavage then exposes a binding motif for neuropilin-1, which mediates penetration into tissue and cells. Conjugation to iRGD significantly improved the sensitivity of tumor-imaging agents and enhanced the activity of an antitumor drug.
- 33Desgrosellier, J.; Cheresh, D. Integrins in cancer: biological implications and therapeutic opportunities. Nat. Rev. Cancer 2010, 10, 9– 22, DOI: 10.1038/nrc274833https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhsF2hsLzI&md5=43d0b526ca38021e780aba991a58451bIntegrins in cancer: biological implications and therapeutic opportunitiesDesgrosellier, Jay S.; Cheresh, David A.Nature Reviews Cancer (2010), 10 (1), 9-22CODEN: NRCAC4; ISSN:1474-175X. (Nature Publishing Group)A review. The integrin family of cell adhesion receptors regulates a diverse array of cellular functions crucial to the initiation, progression and metastasis of solid tumors. The importance of integrins in several cell types that affect tumor progression has made them an appealing target for cancer therapy. Integrin antagonists, including the αvβ3 and αvβ5 inhibitor cilengitide, have shown encouraging activity in Phase II clin. trials and cilengitide is currently being tested in a Phase III trial in patients with glioblastoma. These exciting clin. developments emphasize the need to identify how integrin antagonists influence the tumor and its microenvironment.
- 34Marelli, U.; Rechenmacher, F.; Sobahi, T.; Mas-Moruno, C.; Kessler, H. Tumor targeting via integrin ligands. Front. Oncol. 2013, 3, 222– 233, DOI: 10.3389/fonc.2013.0022234https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2cflt1Gktg%253D%253D&md5=b09d0b738b25909e8b68d42d90ad638eTumor Targeting via Integrin LigandsMarelli Udaya Kiran; Rechenmacher Florian; Sobahi Tariq Rashad Ali; Mas-Moruno Carlos; Kessler HorstFrontiers in oncology (2013), 3 (), 222 ISSN:2234-943X.Selective and targeted delivery of drugs to tumors is a major challenge for an effective cancer therapy and also to overcome the side-effects associated with current treatments. Overexpression of various receptors on tumor cells is a characteristic structural and biochemical aspect of tumors and distinguishes them from physiologically normal cells. This abnormal feature is therefore suitable for selectively directing anticancer molecules to tumors by using ligands that can preferentially recognize such receptors. Several subtypes of integrin receptors that are crucial for cell adhesion, cell signaling, cell viability, and motility have been shown to have an upregulated expression on cancer cells. Thus, ligands that recognize specific integrin subtypes represent excellent candidates to be conjugated to drugs or drug carrier systems and be targeted to tumors. In this regard, integrins recognizing the RGD cell adhesive sequence have been extensively targeted for tumor-specific drug delivery. Here we review key recent examples on the presentation of RGD-based integrin ligands by means of distinct drug-delivery systems, and discuss the prospects of such therapies to specifically target tumor cells.
- 35Teesalu, T.; Sugahara, K.; Kotamraju, V.; Ruoslahti, E. C-end rule peptides mediate neuropilin-1-dependent cell, vascular, and tissue penetration. Proc. Natl. Acad. Sci. U. S. A. 2009, 106, 16157– 16162, DOI: 10.1073/pnas.090820110635https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXht1OjsbbI&md5=3eb210213cfccc091c4aba949d437b42C-end rule peptides mediate neuropilin-1-dependent cell, vascular, and tissue penetrationTeesalu, Tambet; Sugahara, Kazuki N.; Kotamraju, Venkata Ramana; Erkki RuoslahtiProceedings of the National Academy of Sciences of the United States of America (2009), 106 (38), 16157-16162, S16157/1-S16157/15CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Screening of phage libraries expressing random peptides for binding to prostate cancer cells primarily yielded peptides that had a C-terminal arginine (or rarely lysine) residue, usually in a consensus context R/KXXR/K. Phage expressing these sequences and synthetic nanoparticles coated with them bound to and were internalized into cells. The C-terminal arginine (or lysine) was essential to the activity; adding another amino acid, or even blocking the free carboxyl group of this arginine residue by amidation, eliminated the binding and internalizing activity. An internal R/KXXR/K can be exposed and switched on by a cleavage by a protease. The strict requirement for C-terminal exposure of the motif prompted the authors to term the phenomenon the C-end rule (CendR). Affinity chromatog. showed that the CendR peptides bind to neuropilin-1 (NRP-1) on the target cells. NRP-1 is a cell-surface receptor that plays an essential role in angiogenesis, regulation of vascular permeability, and the development of the nervous system. VEGF-A165 and other ligands of NRP-1 possess a C-terminal CendR sequence that interacts with the b1 domain of NRP-1 and causes cellular internalization and vascular leakage. The authors' CendR peptides have similar effects, particularly when made multivalent through coupling to a particle. The authors also noted a unique and important activity of these peptides: penetration and transportation through tissues. The peptides were able to take payloads up to the nanoparticle size scale deep into extravascular tissue. The authors' observations have implications in drug delivery and penetration of tissue barriers and tumors.
- 36Liu, X.; Liu, C.; Chen, C.; Bentobji, M.; Cheillan, F.; Piana, J.; Qu, F.; Rocchi, P.; Peng, L. Targeted delivery of Dicer-substrate siRNAs using a dual targeting peptide decorated dendrimer delivery system. Nanomedicine 2014, 10, 1627– 1636, DOI: 10.1016/j.nano.2014.05.00836https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtFOgtL3K&md5=27ce93c828721bcad7351c4edde95339Targeted delivery of Dicer-substrate siRNAs using a dual targeting peptide decorated dendrimer delivery systemLiu, Xiaoxuan; Liu, Cheng; Chen, Chao; Bentobji, Melanie; Cheillan, Francine Azario; Piana, Jeanne Thomassin; Qu, Fanqi; Rocchi, Palma; Peng, LingNanomedicine (New York, NY, United States) (2014), 10 (8), 1627-1636CODEN: NANOBF; ISSN:1549-9634. (Elsevier)Small interfering RNAs (siRNA) are emerging as novel therapeutic agents, providing competent delivery systems that are available. Dendrimers, a special family of synthetic macromols., represent an exciting delivery platform by virtue of their well-defined dendritic structure and unique multivalency and cooperativity confined within a nanoscale vol. Here, we report a Dicer-substrate siRNA (dsiRNA) which, when delivered using a structurally flexible triethanolamine-core poly(amidoamine) dendrimer of generation 5 as the nanocarrier, gives rise to a much greater RNAi response than that produced with conventional siRNA. Further decoration of the dsiRNA/dendrimer complexes with a dual targeting peptide simultaneously promoted cancer cell targeting through interacting with integrins and cell penetration via the interaction with neuropilin-1 receptors, which led to improved gene silencing and anticancer activity. Altogether, our results disclosed here open a new avenue for therapeutic implementation of RNAi using dendrimer nanovector based targeted delivery.
- 37Svenson, S. The dendrimer paradox--high medical expectations but poor clinical translation. Chem. Soc. Rev. 2015, 46, 4131– 4144, DOI: 10.1039/C5CS00288EThere is no corresponding record for this reference.
- 38Cao, Y.; Liu, X.; Peng, L. Molecular engineering of dendrimer nanovecters for siRNA delivery and gene silencing. Front. Chem. Sci. Eng. 2017, 11, 663– 675, DOI: 10.1007/s11705-017-1623-538https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXlvFyjsLs%253D&md5=72a76c778f9c6878d0f125e3b57bcb88Molecular engineering of dendrimer nanovectors for siRNA delivery and gene silencingCao, Yu; Liu, Xiaoxuan; Peng, LingFrontiers of Chemical Science and Engineering (2017), 11 (4), 663-675CODEN: FCSEA3; ISSN:2095-0187. (Springer)A review. Small interfering RNA (siRNA) therapeutics hold great promise to treat a variety of diseases, as long as they can be delivered safely and effectively into cells. Dendrimers are appealing vectors for siRNA delivery by virtue of their well-defined mol. architecture and multivalent cooperativity. However, the clin. translation of RNA therapeutics mediated by dendrimer delivery is hampered by the lack of dendrimers that are of high quality to meet good manufg. practice std. In this context, we have developed small amphiphilic dendrimers that self-assemble into supramol. structures, which mimic high-generation dendrimers synthesized with covalent construction, yet are easy to produce in large amt. and superior quality. This short review presents a brief introduction to RNAi therapeutics, the obstacles to their delivery and the advantages of dendrimer delivery vectors as well as our bio-inspired structurally flexible dendrimers for siRNA delivery. We then highlight our efforts in creating selfassembling amphiphilic dendrimers to construct supramol. dendrimer nanosystems for effective siRNA delivery as well as the related structural alterations to enhance delivery efficiency. The advent of self-assembling supramol. dendrimer nanovectors holds great promise and heralds a new era of dendrimer-mediated delivery of RNA therapeutics in biomedical applications.
- 39Zhou, J.; Liu, J.; Cheng, C.; Patel, T.; Weller, C.; Piepmeier, J.; Jiang, Z.; Saltzman, W. Biodegradable poly(amine-co-ester) terpolymers for targeted gene delivery. Nat. Mater. 2012, 11, 82– 90, DOI: 10.1038/nmat318739https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhsFGkurfN&md5=937db7f417aafb60789ec0f760f27d49Biodegradable poly(amine-co-ester) terpolymers for targeted gene deliveryZhou, Jiangbing; Liu, Jie; Cheng, Christopher J.; Patel, Toral R.; Weller, Caroline E.; Piepmeier, Joseph M.; Jiang, Zhaozhong; Saltzman, W. MarkNature Materials (2012), 11 (1), 82-90CODEN: NMAACR; ISSN:1476-1122. (Nature Publishing Group)Many synthetic polycationic vectors for non-viral gene delivery show high efficiency in vitro, but their usually excessive charge d. makes them toxic for in vivo applications. Here we describe the synthesis of a series of high mol. wt. terpolymers with low charge d., and show that they exhibit efficient gene delivery, some surpassing the efficiency of the com. transfection reagents Polyethylenimine and Lipofectamine 2000. The terpolymers were synthesized via enzyme-catalyzed copolymn. of lactone with dialkyl diester and amino diol, and their hydrophobicity adjusted by varying the lactone content and by selecting a lactone comonomer of specific ring size. Targeted delivery of the pro-apoptotic TRAIL gene to tumor xenografts by one of the terpolymers results in significant inhibition of tumor growth, with minimal toxicity both in vitro and in vivo. Our findings suggest that the gene delivery ability of the terpolymers stems from their high mol. wt. and increased hydrophobicity, which compensates for their low charge d.
- 40Grandclement, C.; Borg, C. Neuropilins: a new target for cancer therapy. Cancers 2011, 3, 1899– 1928, DOI: 10.3390/cancers3021899There is no corresponding record for this reference.
- 41Siegel, R.; Miller, K.; Jemal, A. Cancer Statistics, 2017. Ca-Cancer J. Clin. 2017, 67, 7– 30, DOI: 10.3322/caac.2138741https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1c7isFWgtA%253D%253D&md5=e439f3424d5a773e4ae289c392236ad6Cancer Statistics, 2017Siegel Rebecca L; Miller Kimberly D; Jemal AhmedinCA: a cancer journal for clinicians (2017), 67 (1), 7-30 ISSN:.Each year, the American Cancer Society estimates the numbers of new cancer cases and deaths that will occur in the United States in the current year and compiles the most recent data on cancer incidence, mortality, and survival. Incidence data were collected by the Surveillance, Epidemiology, and End Results Program; the National Program of Cancer Registries; and the North American Association of Central Cancer Registries. Mortality data were collected by the National Center for Health Statistics. In 2017, 1,688,780 new cancer cases and 600,920 cancer deaths are projected to occur in the United States. For all sites combined, the cancer incidence rate is 20% higher in men than in women, while the cancer death rate is 40% higher. However, sex disparities vary by cancer type. For example, thyroid cancer incidence rates are 3-fold higher in women than in men (21 vs 7 per 100,000 population), despite equivalent death rates (0.5 per 100,000 population), largely reflecting sex differences in the "epidemic of diagnosis." Over the past decade of available data, the overall cancer incidence rate (2004-2013) was stable in women and declined by approximately 2% annually in men, while the cancer death rate (2005-2014) declined by about 1.5% annually in both men and women. From 1991 to 2014, the overall cancer death rate dropped 25%, translating to approximately 2,143,200 fewer cancer deaths than would have been expected if death rates had remained at their peak. Although the cancer death rate was 15% higher in blacks than in whites in 2014, increasing access to care as a result of the Patient Protection and Affordable Care Act may expedite the narrowing racial gap; from 2010 to 2015, the proportion of blacks who were uninsured halved, from 21% to 11%, as it did for Hispanics (31% to 16%). Gains in coverage for traditionally underserved Americans will facilitate the broader application of existing cancer control knowledge across every segment of the population. CA Cancer J Clin 2017;67:7-30. © 2017 American Cancer Society.
- 42Grasso, C.; Wu, Y.; Robinson, D.; Cao, X.; Dhanasekaran, S.; Khan, A.; Quist, M.; Jing, X.; Lonigro, R.; Brenner, J.; Asangani, I.; Ateeq, B.; Chun, S.; Siddiqui, J.; Sam, L.; Anstett, M.; Mehra, R.; Prensner, J.; Palanisamy, N.; Ryslik, G.; Vandin, F.; Raphael, B.; Kunju, L.; Rhodes, D.; Pienta, K.; Chinnaiyan, A.; Tomlins, S. The mutational landscape of lethal castration-resistant prostate cancer. Nature 2012, 487, 239– 243, DOI: 10.1038/nature1112542https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtVWisLfP&md5=15b1780d9cd2189325baf4e4fec84687The mutational landscape of lethal castration-resistant prostate cancerGrasso, Catherine S.; Wu, Yi-Mi; Robinson, Dan R.; Cao, Xuhong; Dhanasekaran, Saravana M.; Khan, Amjad P.; Quist, Michael J.; Jing, Xiaojun; Lonigro, Robert J.; Brenner, J. Chad; Asangani, Irfan A.; Ateeq, Bushra; Chun, Sang Y.; Siddiqui, Javed; Sam, Lee; Anstett, Matt; Mehra, Rohit; Prensner, John R.; Palanisamy, Nallasivam; Ryslik, Gregory A.; Vandin, Fabio; Raphael, Benjamin J.; Kunju, Lakshmi P.; Rhodes, Daniel R.; Pienta, Kenneth J.; Chinnaiyan, Arul M.; Tomlins, Scott A.Nature (London, United Kingdom) (2012), 487 (7406), 239-243CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Characterization of the prostate cancer transcriptome and genome has identified chromosomal rearrangements and copy no. gains and losses, including ETS gene family fusions, PTEN loss and androgen receptor (AR) amplification, which drive prostate cancer development and progression to lethal, metastatic castration-resistant prostate cancer (CRPC). However, less is known about the role of mutations. Here we sequenced the exomes of 50 lethal, heavily pre-treated metastatic CRPCs obtained at rapid autopsy (including three different foci from the same patient) and 11 treatment-naive, high-grade localized prostate cancers. We identified low overall mutation rates even in heavily treated CRPCs (2.00 per megabase) and confirmed the monoclonal origin of lethal CRPC. Integrating exome copy no. anal. identified disruptions of CHD1 that define a subtype of ETS gene family fusion-neg. prostate cancer. Similarly, we demonstrate that ETS2, which is deleted in approx. one-third of CRPCs (commonly through TMPRSS2:ERG fusions), is also deregulated through mutation. Furthermore, we identified recurrent mutations in multiple chromatin- and histone-modifying genes, including MLL2 (mutated in 8.6% of prostate cancers), and demonstrate interaction of the MLL complex with the AR, which is required for AR-mediated signaling. We also identified novel recurrent mutations in the AR collaborating factor FOXA1, which is mutated in 5 of 147 (3.4%) prostate cancers (both untreated localized prostate cancer and CRPC), and showed that mutated FOXA1 represses androgen signaling and increases tumor growth. Proteins that phys. interact with the AR, such as the ERG gene fusion product, FOXA1, MLL2, UTX (also known as KDM6A) and ASXL1 were found to be mutated in CRPC. In summary, we describe the mutational landscape of a heavily treated metastatic cancer, identify novel mechanisms of AR signaling deregulated in prostate cancer, and prioritize candidates for future study.
- 43Gundem, G.; Van, P.; Kremeyer, B.; Alexandrov, L.; Tubio, J.; Papaemmanuil, E.; Brewer, D.; Kallio, H.; Hoegnas, G.; Annala, M.; Kivinummi, K.; Goody, V.; Latimer, C.; O’Meara, S.; Dawson, K.; Isaacs, W.; Emmert-Buck, M.; Nykter, M.; Foster, C.; Kote-Jarai, Z.; Easton, D.; Whitaker, H.; Neal, D.; Cooper, C.; Eeles, R.; Visakorpi, T.; Campbell, P.; McDermott, U.; Wedge, D.; Bova, G. The evolutionary history of lethal metastatic prostate cancer. Nature 2015, 520, 353– 357, DOI: 10.1038/nature14347There is no corresponding record for this reference.
- 44Lorente, D.; Mateo, J.; Perez-Lopez, R.; de, J.; Attard, G. Sequencing of agents in castration-resistant prostate cancer. Lancet Oncol. 2015, 16, 279– 292, DOI: 10.1016/S1470-2045(15)70033-1There is no corresponding record for this reference.
- 45Cornford, P.; Dodson, A.; Parsons, K.; Desmond, A.; Woolfenden, A.; Fordham, M.; Neoptolemos, J.; Ke, Y.; Foster, C. Heat shock protein expression independently predicts clinical outcome in prostate cancer. Cancer Res. 2000, 60, 7099– 7105There is no corresponding record for this reference.
- 46Rocchi, P.; So, A.; Kojima, S.; Signaevsky, M.; Beraldi, E.; Fazli, L.; Hurtado-Coll, A.; Yamanaka, K.; Gleave, M. Heat shock protein 27 increases after androgen ablation and plays a cytoprotective role in hormone-refractory prostate cancer. Cancer Res. 2004, 64, 6595– 6602, DOI: 10.1158/0008-5472.CAN-03-3998There is no corresponding record for this reference.
- 47Jego, G.; Hazoume, A.; Seigneuric, R.; Garrido, C. Targeting heat shock proteins in cancer. Cancer Lett. 2013, 332, 275– 285, DOI: 10.1016/j.canlet.2010.10.01447https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXlvVWitLs%253D&md5=4e1dcc15387cbb33d4a2d18f5defdd16Targeting heat shock proteins in cancerJego, Gaetan; Hazoume, Adonis; Seigneuric, Renaud; Garrido, CarmenCancer Letters (New York, NY, United States) (2013), 332 (2), 275-285CODEN: CALEDQ; ISSN:0304-3835. (Elsevier)A review. Heat shock proteins (HSPs) HSP27, HSP70 and HSP90 are powerful chaperones. Their expression is induced in response to a wide variety of physiol. and environmental insults including anti-cancer chemotherapy, thus allowing the cell to survive to lethal conditions. Different functions of HSPs have been described to account for their cytoprotective function, including their role as mol. chaperones as they play a central role in the correct folding of misfolded proteins, but also their anti-apoptotic properties. HSPs are often overexpressed in cancer cells and this constitutive expression is necessary for cancer cells' survival. HSPs may have oncogene-like functions and likewise mediate "non-oncogene addiction" of stressed tumor cells that must adapt to a hostile microenvironment, thereby becoming dependent for their survival on HSPs. HSP-targeting drugs have therefore emerged as potential anti-cancer agents. This review describes the different mols. and approaches being used or proposed in cancer therapy based on the in inhibition of HSP90, HSP70 and HSP27.
- 48Wei, T.; Chen, C.; Liu, J.; Liu, C.; Posocco, P.; Liu, X.; Cheng, Q.; Huo, S.; Liang, Z.; Fermeglia, M.; Pricl, S.; Liang, X.; Rocchic, P.; Peng, L. Anticancer drug nanomicelles formed by self-assembling amphiphilic dendrimer to combat cancer drug resistance. Proc. Natl. Acad. Sci. U. S. A. 2015, 112, 2978– 2983, DOI: 10.1073/pnas.141849411248https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXjtFemur4%253D&md5=ba3fcc69507c6da884cfeeabd95fb782Anticancer drug nanomicelles formed by self-assembling amphiphilic dendrimer to combat cancer drug resistanceWei, Tuo; Chen, Chao; Liu, Juan; Liu, Cheng; Posocco, Paola; Liu, Xiaoxuan; Cheng, Qiang; Huo, Shuaidong; Liang, Zicai; Fermeglia, Maurizio; Pricl, Sabrina; Liang, Xing-Jie; Rocchi, Palma; Peng, LingProceedings of the National Academy of Sciences of the United States of America (2015), 112 (10), 2978-2983CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Drug resistance and toxicity constitute challenging hurdles for cancer therapy. The application of nanotechnol. for anticancer drug delivery is expected to address these issues and bring new hope for cancer treatment. In this context, we established an original nanomicellar drug delivery system based on an amphiphilic dendrimer (AmDM), which could generate supramol. micelles to effectively encapsulate the anticancer drug doxorubicin (DOX) with high drug-loading capacity (>40%), thanks to the unique dendritic structure creating large void space for drug accommodation. The resulting AmDM/DOX nanomicelles were able to enhance drug potency and combat doxorubicin resistance in breast cancer models by significantly enhancing cellular uptake while considerably decreasing efflux of the drug. In addn., the AmDM/DOX nanoparticles abolished significantly the toxicity related to the free drug. Collectively, our studies demonstrate that the drug delivery system based on nanomicelles formed with the self-assembling amphiphilic dendrimer constitutes a promising and effective drug carrier in cancer therapy.
- 49Posocco, P.; Liu, X.; Laurini, E.; Marson, D.; Chen, C.; Liu, C.; Fermeglia, M.; Rocchi, P.; Pricl, S.; Peng, L. Impact of siRNA overhangs for dendrimer-mediated siRNA delivery and gene silencing. Mol. Pharmaceutics 2013, 10, 3262– 3273, DOI: 10.1021/mp400329g49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtVyjur%252FI&md5=bc7721f224b9006c6e075bf8a0ff401fImpact of siRNA Overhangs for Dendrimer-Mediated siRNA Delivery and Gene SilencingPosocco, Paola; Liu, Xiaoxuan; Laurini, Erik; Marson, Domenico; Chen, Chao; Liu, Cheng; Fermeglia, Maurizio; Rocchi, Palma; Pricl, Sabrina; Peng, LingMolecular Pharmaceutics (2013), 10 (8), 3262-3273CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)Small interfering RNA (siRNA) have attracted considerable attention, as compelling therapeutics providing safe and competent delivery systems are available. Dendrimers are emerging as appealing siRNA delivery vectors thanks to their unique, well-defined architecture and the resulting cooperativity and multivalency confined within a nanostructure. We have recently disclosed the structurally flexible fifth-generation TEA-core PAMAM dendrimer (G5) as an effective nanocarrier for delivery of sticky siRNA bearing long complementary sequence overhangs (dA)n/(dT)n (n = 5 or 7). Here, using combined exptl./computational approaches, we successfully clarified (i) the underlying mechanisms of interaction between the dendrimer nanovector G5 and siRNA mols. bearing either complementary or noncomplementary sequence overhangs of different length and chem. and (ii) the impact of siRNA overhangs contributing toward the improved delivery potency. Using siRNA with complementary overhangs offer the best action in term of gene silencing through the formation of concatemers, i.e., supramol. structures resulting from synergistic and cooperative binding via (dA)n/(dT)n bridges (n = 5 or 7). On the other hand, although siRNA bearing long, noncomplementary overhangs (dA)n/(dA)n or (dT)n/(dT)n (n = 5 or 7) are endowed with considerably higher gene silencing potency than normal siRNA with (dT)2/(dT)2, they remain less effective than their sticky siRNA counterparts. The obsd. gene silencing potency depends on length, nature, and flexibility of the overhangs, which behave as a sort of clamps that hold and interact with the dendrimer nanovectors, thus impacting siRNA delivery performance and, ultimately, gene silencing. Our findings can be instrumental in designing siRNA entities with enhanced capability to achieve effective RNA interference for therapeutic applications.
- 50Albanyan, B.; Laurini, E.; Posocco, P.; Pricl, S.; Smith, D. Self-assembled multivalent (SAMul) polyanion binding—impact of hydrophobic modifications in the micellar core on DNA and heparin binding at the peripheral cationic ligands. Chem. - Eur. J. 2017, 23, 6391– 6397, DOI: 10.1002/chem.20170017750https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXlvVCgsLk%253D&md5=e971234081cca25156b7bcde61149d98Self-Assembled Multivalent (SAMul) Polyanion Binding-Impact of Hydrophobic Modifications in the Micellar Core on DNA and Heparin Binding at the Peripheral Cationic LigandsAlbanyan, Buthaina; Laurini, Erik; Posocco, Paola; Pricl, Sabrina; Smith, David K.Chemistry - A European Journal (2017), 23 (26), 6391-6397CODEN: CEUJED; ISSN:0947-6539. (Wiley-VCH Verlag GmbH & Co. KGaA)This paper reports a small family of cationic surfactants designed to bind polyanions such as DNA and heparin. Each mol. has the same hydrophilic cationic ligand and a hydrophobic aliph. group with eighteen carbon atoms with one, two, or three alkene groups within the hydrophobic chain (C18-1, C18-2 and C18-3). Dynamic light scattering indicates that more alkenes lead to geometric distortion, giving rise to larger self-assembled multivalent (SAMul) nanostructures. Mallard Blue and Ethidium Bromide dye displacement assays demonstrate that heparin and DNA have markedly different binding preferences, with heparin binding most effectively to C18-1, and DNA to C18-3, even though the mol. structural differences of these SAMul systems are buried in the hydrophobic core. Multiscale modeling suggests that adaptive heparin maximises enthalpically favorable interactions with C18-1, while shape-persistent DNA forms a similar no. of interactions with each ligand display, but with slightly less entropic cost for binding to C18-3-fundamental thermodn. differences in SAMul binding of heparin or DNA. This study therefore provides unique insight into electrostatic mol. recognition between highly charged nanoscale surfaces in biol. relevant systems.
- 51Fechner, L.; Albanyan, B.; Vieira, V.; Laurini, E.; Posocco, P.; Pricl, S.; Smith, D. Electrostatic binding of polyanions using self-assembled multivalent (SAMul) ligand displays - structure-activity effects on DNA/heparin binding. Chem. Sci. 2016, 7, 4653– 4659, DOI: 10.1039/C5SC04801J51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xms1Kks7o%253D&md5=2663d127c7871258d0687f089ec63624Electrostatic binding of polyanions using self-assembled multivalent (SAMul) ligand displays - structure-activity effects on DNA/heparin bindingFechner, Loryn E.; Albanyan, Buthaina; Vieira, Vania M. P.; Laurini, Erik; Posocco, Paola; Pricl, Sabrina; Smith, David K.Chemical Science (2016), 7 (7), 4653-4659CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)This paper reports that modifying the ligands in self-assembled multivalent (SAMul) displays has an impact on apparent binding selectivity towards two nanoscale biol. polyanions - heparin and DNA. For the nanostructures assayed here, spermidine ligands are optimal for heparin binding but spermine ligands are preferred for DNA. Probing subtle differences in such nanoscale binding interfaces is a significant challenge, and as such, several exptl. binding assays - competition assays and isothermal calorimetry - are employed to confirm differences in affinity and provide thermodn. insights. Given the dynamic nature and hierarchical binding processes involved in SAMul systems, we employed multiscale modeling to propose reasons for the origins of polyanion selectivity differences. The modeling results, when expressed in thermodn. terms and compared with the exptl. data, suggest that DNA is a shape-persistent polyanion, and selectivity originates only from ligand preferences, whereas heparin is more flexible and adaptive, and as such, actively reinforces ligand preferences. As such, this study suggests that inherent differences between polyanions may underpin subtle binding selectivity differences, and that even simple electrostatic interfaces such as these can have a degree of tunability, which has implications for biol. control and regulation on the nanoscale.
- 52Qi, C.; Su, Y.; Peng, Y.; Tian, E.; Sun, G.; Zhou, J.; Sun, G.; Liu, X.; Chao, C.; Murai, K.; Zhao, C.; Azizian, K.; Yang, L.; Warden, C.; Wu, X.; D’Apuzzo, M.; Brown, C.; Badie, B.; Peng, L.; Riggs, D.; Rossi, J.; Shi, Y. Downregulation of TLX induces TET3 expression and inhibits glioblastoma stem cell self-renewal and tumorigenesis. Nat. Commun. 2016, 7, 10637– 10651, DOI: 10.1038/ncomms10637There is no corresponding record for this reference.
Supporting Information
Supporting Information
The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/jacs.8b10021.
Materials and methods as well as all experimental protocols for TEM, DLS, computational details, ITC, cell uptake and endosome release, siRNA delivery, gene silencing, anticancer activity and toxicity, etc. (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.