Leveraging aptamers for targeted protein degradation.

[1]  Qiang Cheng,et al.  Lipid Nanoparticle (LNP) Enables mRNA Delivery for Cancer Therapy , 2023, Advanced materials.

[2]  J. Zhang,et al.  Targeting L‐Selectin Lymphocytes to Deliver Immunosuppressive Drug in Lymph Nodes for Durable Multiple Sclerosis Treatment , 2023, Advanced science.

[3]  Céline Cano,et al.  A beginner's guide to current synthetic linker strategies towards VHL-recruiting PROTACs. , 2023, Bioorganic & medicinal chemistry.

[4]  H. T. Soh,et al.  A massively parallel screening platform for converting aptamers into molecular switches , 2023, Nature communications.

[5]  Kun Xu,et al.  c-Myc-Targeting PROTAC Based on a TNA-DNA Bivalent Binder for Combination Therapy of Triple-Negative Breast Cancer. , 2023, Journal of the American Chemical Society.

[6]  Yan Xu,et al.  The aptamer-based RNA-PROTAC. , 2023, Bioorganic & medicinal chemistry.

[7]  H. Harashima,et al.  On the mechanism of tissue-selective gene delivery by lipid nanoparticles. , 2023, Journal of controlled release : official journal of the Controlled Release Society.

[8]  Zhi Zhu,et al.  Frontispiece: Aptamer‐LYTACs for Targeted Degradation of Extracellular and Membrane Proteins , 2023, Angewandte Chemie International Edition.

[9]  M. Manoharan,et al.  Chemistry, structure and function of approved oligonucleotide therapeutics , 2023, Nucleic acids research.

[10]  C. Crews,et al.  Protein degraders enter the clinic — a new approach to cancer therapy , 2023, Nature Reviews Clinical Oncology.

[11]  Hai-long Zhang Current status and patent prospective of lipid nanoparticle for mRNA delivery , 2023, Expert opinion on therapeutic patents.

[12]  Songbo Xie,et al.  Peptide-based PROTACs: Current Challenges and Future Perspectives. , 2023, Current medicinal chemistry.

[13]  D. Zhong,et al.  Selective degradation of the p53‐R175H oncogenic hotspot mutant by an RNA aptamer‐based PROTAC , 2023, Clinical and translational medicine.

[14]  J. Ran,et al.  Inducible Degradation of Oncogenic Nucleolin Using an Aptamer-Based PROTAC. , 2023, Journal of medicinal chemistry.

[15]  Y. Takakura,et al.  Extracellular vesicle-based therapeutics: Extracellular vesicles as therapeutic targets and agents. , 2023, Pharmacology & therapeutics.

[16]  F. Kaye,et al.  PROTAC therapy as a new targeted therapy for lung cancer , 2022, Molecular therapy : the journal of the American Society of Gene Therapy.

[17]  Xiaobing Zhang,et al.  Aptamer Inhibits Tumor Growth by Leveraging Cellular Proteasomal Degradation System to Degrade c-Met in Mice. , 2022, Angewandte Chemie.

[18]  M. Oren,et al.  Drugging p53 in cancer: one protein, many targets , 2022, Nature Reviews Drug Discovery.

[19]  Fernando Salangsang,et al.  Modular cytokine receptor-targeting chimeras for targeted degradation of cell surface and extracellular proteins , 2022, Nature Biotechnology.

[20]  Yan Xu,et al.  A heterobifunctional molecule recruits cereblon to an RNA scaffold and activates its PROTAC function , 2022, Cell Reports Physical Science.

[21]  S. Knapp,et al.  PROTAC degraders as chemical probes for studying target biology and target validation. , 2022, Chemical Society reviews.

[22]  Jun Zhou,et al.  Targeting Undruggable Transcription Factors with PROTACs: Advances and Perspectives. , 2022, Journal of medicinal chemistry.

[23]  Quanyin Hu,et al.  Proteolysis-targeting chimera (PROTAC) delivery system: advancing protein degraders towards clinical translation. , 2022, Chemical Society reviews.

[24]  B. Pei,et al.  Target and tissue selectivity of PROTAC degraders. , 2022, Chemical Society reviews.

[25]  Wenchen Pu,et al.  Proteolysis-targeting chimeras (PROTACs) in cancer therapy , 2022, Molecular Cancer.

[26]  Zeyu Cai,et al.  Rational Design for Nitroreductase (NTR)-Responsive Proteolysis Targeting Chimeras (PROTACs) Selectively Targeting Tumor Tissues. , 2022, Journal of medicinal chemistry.

[27]  Xing Sun,et al.  Development of a novel PROTAC using the nucleic acid aptamer as a targeting ligand for tumor selective degradation of nucleolin , 2022, bioRxiv.

[28]  D. Langley,et al.  PROTAC targeted protein degraders: the past is prologue , 2022, Nature Reviews Drug Discovery.

[29]  L. Cerchia,et al.  Profiling Cancer Cells by Cell-SELEX: Use of Aptamers for Discovery of Actionable Biomarkers and Therapeutic Applications Thereof , 2021, Pharmaceutics.

[30]  Xiujun Cai,et al.  Targeting mutant p53 for cancer therapy: direct and indirect strategies , 2021, Journal of Hematology & Oncology.

[31]  L. Jaykus,et al.  Generation of Nucleic Acid Aptamer Candidates against a Novel Calicivirus Protein Target , 2021, Viruses.

[32]  Wenyi Wei,et al.  Folate-Guided Protein Degradation by Immunomodulatory Imide Drug-Based Molecular Glues and Proteolysis Targeting Chimeras. , 2021, Journal of medicinal chemistry.

[33]  Guoqiang Dong,et al.  Aptamer-PROTAC Conjugates (APCs) for Tumor-specific Targeting in Breast Cancer. , 2021, Angewandte Chemie.

[34]  S. Luo,et al.  Degradation of lipid droplets by chimeric autophagy-tethering compounds , 2021, Cell Research.

[35]  Y. Xiong,et al.  Advancing targeted protein degradation for cancer therapy , 2021, Nature Reviews Cancer.

[36]  J. L. La Clair,et al.  Unraveling the Role of Linker Design in Proteolysis Targeting Chimeras. , 2021, Journal of medicinal chemistry.

[37]  D. Trauner,et al.  Optical control of targeted protein degradation. , 2021, Cell chemical biology.

[38]  Yuquan Wei,et al.  Role of lysosomes in physiological activities, diseases, and therapy , 2021, Journal of Hematology & Oncology.

[39]  Wenyi Wei,et al.  Cancer Selective Target Degradation by Folate-Caged PROTACs. , 2021, Journal of the American Chemical Society.

[40]  C. Crews,et al.  Targeted protein degradation: a promise for undruggable proteins. , 2021, Cell chemical biology.

[41]  Da Han,et al.  Bispecific Aptamer Chimeras Enable Targeted Protein Degradation on Cell Membranes. , 2021, Angewandte Chemie.

[42]  Lixia Chen,et al.  Rational Design and Synthesis of Novel Dual PROTACs for Simultaneous Degradation of EGFR and PARP. , 2021, Journal of medicinal chemistry.

[43]  C. Bertozzi,et al.  LYTACs that engage the asialoglycoprotein receptor for targeted protein degradation , 2021, Nature Chemical Biology.

[44]  M. Eisenstein,et al.  Engineering Aptamer Switches for Multifunctional Stimulus‐Responsive Nanosystems , 2020, Advanced materials.

[45]  D. Yadav,et al.  PEGYLATION: an important approach for novel drug delivery system , 2020, Journal of biomaterials science. Polymer edition.

[46]  Carolyn R. Bertozzi,et al.  Lysosome-targeting chimaeras for degradation of extracellular proteins , 2020, Nature.

[47]  Meral Yüce,et al.  Current Perspectives on Aptamers as Diagnostic Tools and Therapeutic Agents , 2020, Pharmaceutics.

[48]  Shuaijian Ni,et al.  Recent Progress in Aptamer Discoveries and Modifications for Therapeutic Applications. , 2020, ACS applied materials & interfaces.

[49]  Daohong Zhou,et al.  PROteolysis TArgeting Chimeras (PROTACs) as emerging anticancer therapeutics , 2020, Oncogene.

[50]  A. Levine p53: 800 million years of evolution and 40 years of discovery , 2020, Nature Reviews Cancer.

[51]  T. Golub,et al.  The CDK inhibitor CR8 acts as a molecular glue degrader that depletes cyclin K , 2020, Nature.

[52]  N. Gray,et al.  Light-induced control of protein destruction by opto-PROTAC , 2020, Science Advances.

[53]  C. Crews,et al.  Proteolysis-Targeting Chimeras as Therapeutics and Tools for Biological Discovery , 2020, Cell.

[54]  G. Winter,et al.  Targeted protein degradation: current and future challenges. , 2020, Current opinion in chemical biology.

[55]  W. Gu,et al.  Mutant p53 on the Path to Metastasis. , 2020, Trends in cancer.

[56]  T. Akaike,et al.  AUTACs: Cargo-Specific Degraders Using Selective Autophagy. , 2019, Molecular cell.

[57]  A. Ballabio,et al.  Lysosomes as dynamic regulators of cell and organismal homeostasis , 2019, Nature Reviews Molecular Cell Biology.

[58]  S. M. Taghdisi,et al.  Therapeutic applications of AS1411 aptamer, an update review. , 2019, International journal of biological macromolecules.

[59]  Yi Xiao,et al.  Universal Design of Structure-Switching Aptamers with Signal Reporting Functionality. , 2019, Analytical chemistry.

[60]  Philip P. Chamberlain,et al.  Development of targeted protein degradation therapeutics , 2019, Nature Chemical Biology.

[61]  S. Muller,et al.  Lysosomes as a therapeutic target , 2019, Nature Reviews Drug Discovery.

[62]  B. Sullenger,et al.  Anti-PEG Antibodies Inhibit the Anticoagulant Activity of PEGylated Aptamers. , 2019, Cell chemical biology.

[63]  Nandini Kundu,et al.  Mirror-Image Oligonucleotides: History and Emerging Applications. , 2019, Chemistry.

[64]  M. Naito,et al.  SNIPERs-Hijacking IAP activity to induce protein degradation. , 2019, Drug discovery today. Technologies.

[65]  Roberto A Barrero,et al.  Three decades of nucleic acid aptamer technologies: Lessons learned, progress and opportunities on aptamer development. , 2019, Biotechnology advances.

[66]  Xiaoyuan Chen,et al.  Aptamer‐based targeted therapy , 2018, Advanced drug delivery reviews.

[67]  R. Eritja,et al.  AS1411-decorated niosomes as effective nanocarriers for Ru(iii)-based drugs in anticancer strategies. , 2018, Journal of materials chemistry. B.

[68]  D. DiMaio,et al.  Regulation of C-C chemokine receptor 5 (CCR5) stability by Lys197 and by transmembrane protein aptamers that target it for lysosomal degradation , 2018, The Journal of Biological Chemistry.

[69]  K. Wong,et al.  USP15-dependent lysosomal pathway controls p53-R175H turnover in ovarian cancer cells , 2018, Nature Communications.

[70]  James E. Bradner,et al.  The dTAG system for immediate and target-specific protein degradation , 2018, Nature Chemical Biology.

[71]  Jianliang Zhang,et al.  Novel DNA Aptamers for Parkinson’s Disease Treatment Inhibit α-Synuclein Aggregation and Facilitate its Degradation , 2018, Molecular therapy. Nucleic acids.

[72]  K. Wiman,et al.  Targeting mutant p53 for efficient cancer therapy , 2017, Nature Reviews Cancer.

[73]  Melina Schuh,et al.  A Method for the Acute and Rapid Degradation of Endogenous Proteins , 2017, Cell.

[74]  B. Sullenger,et al.  Aptamers as Therapeutics. , 2017, Annual review of pharmacology and toxicology.

[75]  J. Rossi,et al.  Aptamers as targeted therapeutics: current potential and challenges , 2016, Nature Reviews Drug Discovery.

[76]  G. Shan,et al.  The isolation of an RNA aptamer targeting to p53 protein with single amino acid mutation , 2015, Proceedings of the National Academy of Sciences.

[77]  K. Morris,et al.  Cell-specific RNA aptamer against human CCR5 specifically targets HIV-1 susceptible cells and inhibits HIV-1 infectivity. , 2015, Chemistry & biology.

[78]  J. Shankaranarayanan,et al.  Aptamer-based therapeutics of the past, present and future: from the perspective of eye-related diseases. , 2014, Drug discovery today.

[79]  L. Garraway,et al.  A phase II trial of AS1411 (a novel nucleolin-targeted DNA aptamer) in metastatic renal cell carcinoma , 2014, Investigational New Drugs.

[80]  Katharina Berg,et al.  Stabilized Interleukin-6 receptor binding RNA aptamers , 2014, RNA biology.

[81]  Y. Yarden,et al.  Aptamer to ErbB-2/HER2 enhances degradation of the target and inhibits tumorigenic growth , 2013, Proceedings of the National Academy of Sciences.

[82]  K. Ikebukuro,et al.  Selection of DNA aptamers that recognize α-synuclein oligomers using a competitive screening method. , 2012, Analytical chemistry.

[83]  Xiaojian Zhang,et al.  Development of Hypoxia-activated PROTAC Exerting More Potent Effect in Tumor Hypoxia than in Normoxia , 2021, Chemical Communications.

[84]  Haoxing Xu,et al.  Lysosomal physiology. , 2015, Annual review of physiology.