349
Views
6
CrossRef citations to date
0
Altmetric
Review

Tackling prion diseases: a review of the patent landscape

ORCID Icon & ORCID Icon
Pages 1097-1115 | Received 17 Mar 2021, Accepted 15 Jun 2021, Published online: 21 Jul 2021

References

  • Prusiner SB. Shattuck lecture--neurodegenerative diseases and prions. N Engl J Med. 2001 May;344(20):1516–1526.
  • Ma J, Wang F. Prion disease and the ‘protein-only hypothesis. Essays Biochem. 2014;56:181–191.
  • Colby DW, Prusiner SB. Prions. Cold Spring Harb Perspect Biol. 2011 Jan;3(1):a006833.
  • Atkinson CJ, Zhang K, Munn AL, et al. Prion protein scrapie and the normal cellular prion protein. Prion. 2016;10(1):63–82.
  • Brown K, Mastrianni JA. The prion diseases. J Geriatr Psychiatry Neurol. 2010 Dec;23(4):277–298.
  • Prusiner SB, Safar JG Method of preparing cow brain homogenate. WO0022438A1 (2000).
  • Tee BL, Longoria Ibarrola EM, Geschwind MD. Prion diseases. Neurol Clin. 2018 11;36(4):865–897. .
  • Bruce ME, Will RG, Ironside JW, et al. Transmissions to mice indicate that ‘new variant’ CJD is caused by the BSE agent. Nature. 1997 Oct;389(6650):498–501.
  • Will RG, Ironside JW, Zeidler M, et al. A new variant of Creutzfeldt-Jakob disease in the UK. Lancet. 1996 Apr;347(9006):921–925.
  • Comoy EE, Mikol J, Luccantoni-Freire S, et al. Transmission of scrapie prions to primate after an extended silent incubation period. Sci Rep. 2015 Jun;5(1):11573. .
  • Nemani SK, Myskiw JL, Lamoureux L, et al. Exposure risk of chronic wasting disease in humans. Viruses. 2020 12; 12(12): 1454.
  • Morales R, Abid K, Soto C. The prion strain phenomenon: molecular basis and unprecedented features. Biochim Biophys Acta. 2007 Jun;1772(6):681–691.
  • Morales R. Prion strains in mammals: different conformations leading to disease. PLoS Pathog. 2017 07;13(7):e1006323.
  • Colini Baldeschi A, Vanni S, Zattoni M, et al. Novel regulators of PrP. Expert Opin Ther Targets. 2020 08;24(8):759–776.
  • Bosque PJ, Prusiner SB. Cultured cell sublines highly susceptible to prion infection. J Virol. 2000 May;74(9):4377–4386.
  • Vilette D. Cell models of prion infection.Vet Res. 2008 [2008 Jul-Aug];39(4):10. .
  • Grassmann A, Wolf H, Hofmann J, et al. Cellular aspects of prion replication in vitro. Viruses. 2013 Jan;5(1):374–405. .
  • Brandner S, Jaunmuktane Z. Prion disease: experimental models and reality. Acta Neuropathol. 2017 Feb;133(2):197–222.
  • Moreno JA, Telling GC. Insights into mechanisms of transmission and pathogenesis from transgenic mouse models of prion diseases. Methods Mol Biol. 2017;1658:219–252.
  • Saborio GP, Permanne B, Soto C. Sensitive detection of pathological prion protein by cyclic amplification of protein misfolding. Nature. 2001 Jun;411(6839):810–813.
  • Castilla J, Saa P, Soto C Ultrasensitive detection of prions by automated protein misfolding cyclic amplification. WO2006113915A2 (2006).
  • Atarashi R, Wilham JM, Christensen L, et al. Simplified ultrasensitive prion detection by recombinant PrP conversion with shaking. Nat Methods. 2008 Mar;5(3):211–212.
  • Orru CD, Caughey B, Kuhn F, et al. Methods for amplification and detection of prions. WO2012099884A1 (2012).
  • Lowe DM, Corbett PT, Murray-Rust P, et al. Chemical name to structure: OPSIN, an open source solution. J Chem Inf Model. 2011 Mar;51(3):739–753.
  • Prusiner SB, McKinley MP, Bowman KA, et al. Scrapie prions aggregate to form amyloid-like birefringent rods. Cell. 1983 Dec;35(2):349–358.
  • Caughey B, Race R, Treatment of diseases associated with amyloid formation by using Congo Red. WO9401116A1 (1994).
  • Caughey B, Race RE. Potent inhibition of scrapie-associated PrP accumulation by congo red. J Neurochem. 1992 Aug;59(2):768–771.
  • Ingrosso L, Ladogana A, Pocchiari M. Congo red prolongs the incubation period in scrapie-infected hamsters. J Virol. 1995 Jan;69(1):506–508.
  • Klunk WE, Debnath ML, Pettegrew JW. Development of small molecule probes for the beta-amyloid protein of Alzheimer’s disease. Neurobiol Aging. 1994 Nov-Dec;15(6):691–698.
  • Gray LE, Ostby JS. The effects of prenatal administration of azo dyes on testicular development in the mouse: a structure activity profile of dyes derived from benzidine, dimethylbenzidine, or dimethoxybenzidine. Fundam Appl Toxicol. 1993 Feb;20(2):177–183.
  • Talaska G. Aromatic amines and human urinary bladder cancer: exposure sources and epidemiology. J Environ Sci Health C Environ Carcinog Ecotoxicol Rev. 2003 May;21(1):29–43.
  • Gilbert I, Sellarajah S . Compounds. WO2005085189A2 (2005).
  • Roikhel VM, Fokina GI, Pogodina VV. Influence of aminasine on experimental scrapie in mice. Acta Virol. 1984 Jul;28(4):321–324.
  • Doh-Ura K, Iwaki T, Caughey B. Lysosomotropic agents and cysteine protease inhibitors inhibit scrapie-associated prion protein accumulation. J Virol. 2000 May;74(10):4894–4897.
  • Korth C, May BC, Cohen FE, et al. Acridine and phenothiazine derivatives as pharmacotherapeutics for prion disease. Proc Natl Acad Sci U S A. 2001 Aug;98(17):9836–9841.
  • Prusiner SB, Korth C, May BCH Optically active compounds clearing malformed proteins. WO02096431A1 (2002).
  • Post K, Pitschke M, Schäfer O, et al. Rapid acquisition of beta-sheet structure in the prion protein prior to multimer formation. Biol Chem. 1998 Nov;379(11):1307–1317. .
  • Ferguson LR, Denny WA. Genotoxicity of non-covalent interactions: DNA intercalators. Mutat Res. 2007 Oct;623(1–2):14–23.
  • May BC, Fafarman AT, Hong SB, et al. Potent inhibition of scrapie prion replication in cultured cells by bis-acridines. Proc Natl Acad Sci U S A. 2003 Mar;100(6):3416–3421. .
  • Prusiner SB, Korth C, May BCH Cyclic bis-compounds clearing malformed proteins. US2004229898A1 (2004).
  • Barret A, Tagliavini F, Forloni G, et al. Evaluation of quinacrine treatment for prion diseases. J Virol. 2003 Aug;77(15):8462–8469.
  • Weissmann C, Aguzzi A. Approaches to therapy of prion diseases. Annu Rev Med. 2005;56(1):321–344.
  • Collins SJ, Lewis V, Brazier M, et al. Quinacrine does not prolong survival in a murine Creutzfeldt-Jakob disease model. Ann Neurol. 2002 Oct;52(4):503–506. .
  • Klingenstein R, Löber S, Kujala P, et al. Tricyclic antidepressants, quinacrine and a novel, synthetic chimera thereof clear prions by destabilizing detergent-resistant membrane compartments. J Neurochem. 2006 Aug;98(3):748–759.
  • Korth C, Klingenstein R, Loeber S, et al. 9-amino-acridine derivatives and their use for eliminating misfolded proteins. WO2006092395A1 (2006).
  • Murakami-Kubo I, Doh-Ura K, Ishikawa K, et al. Quinoline derivatives are therapeutic candidates for transmissible spongiform encephalopathies. J Virol. 2004 Feb;78(3):1281–1288.
  • Peters W. The evolution of tafenoquine--antimalarial for a new millennium? J R Soc Med. 1999 Jul;92(7):345–352.
  • Chiang PK, Levy DE, Guo M, et al. Methods and compositions for treating diseases associated with pathogenic proteins. US2007179123A1 (2007).
  • Scott MR, Köhler R, Foster D, et al. Chimeric prion protein expression in cultured cells and transgenic mice. Protein Sci. 1992 Aug;1(8):986–997.
  • Supattapone S, Nguyen HO, Cohen FE, et al. Elimination of prions by branched polyamines and implications for therapeutics. Proc Natl Acad Sci U S A. 1999 Dec;96(25):14529–14534. .
  • Tang MX, Redemann CT, Szoka FC. In vitro gene delivery by degraded polyamidoamine dendrimers. Bioconjug Chem. 1996 Nov-Dec;7(6):703–714. . 1996.
  • Taraboulos A, Raeber AJ, Borchelt DR, et al. Synthesis and trafficking of prion proteins in cultured cells. Mol Biol Cell. 1992 Aug;3(8):851–863.
  • Borchelt DR, Taraboulos A, Prusiner SB. Evidence for synthesis of scrapie prion proteins in the endocytic pathway. J Biol Chem. 1992 Aug;267(23):16188–16199.
  • Caughey B, Raymond GJ, Ernst D, et al. N-terminal truncation of the scrapie-associated form of PrP by lysosomal protease(s): implications regarding the site of conversion of PrP to the protease-resistant state. J Virol. 1991 Dec;65(12):6597–6603.
  • Prusiner SB, Supattapone S, Scott MR Food additives which affect conformationally altered proteins. US6331296B1 (2001).
  • Cashman NR, Caughey B. Prion diseases--close to effective therapy? Nat Rev Drug Discov. 2004 Oct;3(10):874–884.
  • Chakroun N, Prigent S, Dreiss CA, et al. The oligomerization properties of prion protein are restricted to the H2H3 domain. FASEB J. 2010 Sep;24(9):3222–3231.
  • Xu Z, Deslys J-P RH Compositions for use in the treatment or diagnosis of prion diseases. WO2011151798A1 (2011).
  • Wong C, Xiong LW, Horiuchi M, et al. Sulfated glycans and elevated temperature stimulate PrP(Sc)-dependent cell-free formation of protease-resistant prion protein. EMBO J. 2001 Feb;20(3):377–386.
  • Uzawa H, Horiuchi M, Nishida Y, et al. Prion Growth Inhibitor. WO2005077382A1 (2005).
  • Vieira TC, Cordeiro Y, Caughey B, et al. Heparin binding confers prion stability and impairs its aggregation. FASEB J. 2014 Jun;28(6):2667–2676.
  • Minetti P, Penco S, Naggi A, et al. Low molecular weight heparin derivatives having neuroprotective activity. WO2009007224A1 (2009).
  • Horonchik L, Tzaban S, Ben-Zaken O, et al. Heparan sulfate is a cellular receptor for purified infectious prions. J Biol Chem. 2005 Apr;280(17):17062–17067.
  • Shyng SL, Lehmann S, Moulder KL, et al. Sulfated glycans stimulate endocytosis of the cellular isoform of the prion protein, PrPC, in cultured cells. J Biol Chem. 1995 Dec;270(50):30221–30229.
  • Caughey B, Raymond GJ. Sulfated polyanion inhibition of scrapie-associated PrP accumulation in cultured cells. J Virol. 1993 Feb;67(2):643–650.
  • Domb AJ, Katz E, Yudovin-Farber I, et al. Polysaccharide-oligoamine conjugates as anti-amyloid and anti-viral agents. WO2006090365A2 (2006).
  • Camilleri P, Haskins NJ, Howlett DR. beta-Cyclodextrin interacts with the Alzheimer amyloid beta-A4 peptide. FEBS Lett. 1994 Mar;341(2–3):256–258.
  • Zidovetzki R, Levitan I. Use of cyclodextrins to manipulate plasma membrane cholesterol content: evidence, misconceptions and control strategies. Biochim Biophys Acta. 2007 Jun;1768(6):1311–1324.
  • McMahon H, Prior M Compositions comprising oligosaccharides for treating prion disease. WO2007137808A1 (2007).
  • Tiwari G, Tiwari R, Rai AK. Cyclodextrins in delivery systems: applications. J Pharm Bioallied Sci. 2010 Apr;2(2):72–79.
  • Sedel F, Haik S, Bizat N Method for treating prion diseases. WO2020025598A1 (2020).
  • Bach S, Talarek N, Andrieu T, et al. Isolation of drugs active against mammalian prions using a yeast-based screening assay. Nat Biotechnol. 2003 Sep;21(9):1075–1081.
  • Walker BR, Deitch MW, Schneider BE, et al. Long-term therapy of hypertension with guanabenz. Clin Ther. 1981;4(3):217–228.
  • Blondel M, Bach S, Vilette D, et al. Use of chlorine guanabenz derivatives for treating prion-based diseases. WO2008041134A2 (2008).
  • Charvériat M, Reboul M, Wang Q, et al. New inhibitors of prion replication that target the amyloid precursor. J Gen Virol. 2009 May;90(5):1294–1301.
  • Reboul M, Mouthon F, Deslys J-P, et al. Novel derivative of erythromycin for the treatment and diagnosis of prion disease. WO2010084201A1 (2010).
  • Bieschke J, Giese A, Schulz-Schaeffer W, et al. Ultrasensitive detection of pathological prion protein aggregates by dual-color scanning for intensely fluorescent targets. Proc Natl Acad Sci U S A. 2000 May;97(10):5468–5473.
  • Winklhofer KF, Tatzelt J. Cationic lipopolyamines induce degradation of PrPSc in scrapie-infected mouse neuroblastoma cells. Biol Chem. 2000 May-Jun;381(5–6):463–469.
  • Bertsch UWE, Giese A, Kretzschmar H, et al. Systematic identification of new anti-prion drugs by high-throughput screening based on Scanning for Intensely Fluorescent Targets (SIFT). WO2005116640A2 (2005).
  • Giese A, Bertsch UWE, Kretzschmar H, et al. New drug for inhibiting aggregation of proteins involved in diseases linked to protein aggregation and/or neurodegenerative diseases. WO2010000372A2 (2010).
  • Massignan T, Sangiovanni V, Biggi S, et al. A small-molecule inhibitor of prion replication and mutant prion protein toxicity. ChemMedChem. 2017 08; 12(16): 1286–1292.
  • Altieri A, Spiridonov EA, Sivtzev SI, et al. Generation, optimization and characterization of novel anti-prion compounds. Bioorg Med Chem. 2020 11; 28(21): 115717.
  • Westergard L, Christensen HM, Harris DA. The cellular prion protein (PrP(C)): its physiological function and role in disease. Biochim Biophys Acta. 2007 Jun;1772(6):629–644.
  • Biasini E, Turnbaugh JA, Unterberger U, et al. Prion protein at the crossroads of physiology and disease. Trends Neurosci. 2012 Feb;35(2):92–103.
  • Apetri AC, Surewicz WK. Kinetic intermediate in the folding of human prion protein. J Biol Chem. 2002 Nov;277(47):44589–44592.
  • Perrier V, Wallace AC, Kaneko K, et al. Mimicking dominant negative inhibition of prion replication through structure-based drug design. Proc Natl Acad Sci U S A. 2000 May;97(11):6073–6078.
  • Kuwata K, Li H, Yamada H, et al. Locally disordered conformer of the hamster prion protein: a crucial intermediate to PrPSc? Biochemistry. 2002 Oct;41(41):12277–12283.
  • Kuwata K, Nishida N, Katamine S, Inhibitor against specific structure formation of amyloid precursor. JP2005120002A (2005).
  • Kuwata K, Nishida N, Matsumoto T, et al. Hot spots in prion protein for pathogenic conversion. Proc Natl Acad Sci U S A. 2007 Jul;104(29):11921–11926.
  • Kuwata K, Kimura T, Muto J Prion protein structure transformation inhibitor and utilization of same. WO2010131717A1 (2010).
  • Kuwata K, Maleate of anti-prion compound, method for producing same, and pharmaceutical composition thereof. WO2015119111A1 (2015).
  • Yamaguchi K, Kamatari YO, Ono F, et al. A designer molecular chaperone against transmissible spongiform encephalopathy slows disease progression in mice and macaques. Nat Biomed Eng. 2019 03; 3(3): 206–219.
  • Ishibashi D, Nakagaki T, Ishikawa T, et al. Structure-based drug discovery for prion disease using a novel binding simulation. EBioMedicine. 2016 Jul;9:238–249.
  • Ishibashi D, Nishida N, Mizuta S, et al. Therapeutic agent for prion diseases. WO2020059841A1 (2020).
  • Biasini E, Harris DA, Beeler A, et al. Prion protein ligands as therapeutic agents for neurodegenerative disorders. WO2014025785A2 (2014).
  • Hachiya NS, Imagawa M, Kaneko K. The possible role of protein X, a putative auxiliary factor in pathological prion replication, in regulating a physiological endoproteolytic cleavage of cellular prion protein. Med Hypotheses. 2007;68(3):670–673.
  • Tamgüney G, Giles K, Glidden DV, et al. Genes contributing to prion pathogenesis. J Gen Virol. 2008 Jul;89(7):1777–1788.
  • Stein-Gerlach M, Salassidis K, Bacher G, et al. Pyridylpyrimidine derivatives as effective compounds against prion infections and prion diseases. US2006217404A1 (2006).
  • Kim KE, Park S, Cheon S, et al. Novel application of radotinib for the treatment of solid tumors via natural killer cell activation. J Immunol Res. 2018;2018:9580561.
  • Kim DY, Shin JS, Cho DJ, et al. Pharmaceutical composition for prevention or treatment of prion disease. WO2020009522A1 (2020).
  • Barbisin M, Vanni S, Schmadicke AC, et al. Gene expression profiling of brains from bovine spongiform encephalopathy (BSE)-infected cynomolgus macaques. BMC Genomics. 2014 Jun 5;15(1):434.
  • Vanni S, Moda F, Zattoni M, et al. Differential overexpression of SERPINA3 in human prion diseases. Sci Rep. 2017 Nov 15;7(1):15637.
  • Legname G, Vanni S, Carloni P, et al. Serpin inhibitors for the treatment of prion and prion-like diseases. WO2019211265A1 (2019).
  • Bu G. The roles of receptor-associated protein (RAP) as a molecular chaperone for members of the LDL receptor family. Int Rev Cytol. 2001;209:79–116.
  • Kerr ML, Gasperini R, Gibbs ME, et al. Inhibition of Abeta aggregation and neurotoxicity by the 39-kDa receptor-associated protein. J Neurochem. 2010 Mar;112(5):1199–1209.
  • Cervenakova L, Yakovleva O Inhibition of prion protein propagation by Receptor Associated Protein (RAP), its derivatives, mimetics and syntentic peptides. WO2010134970A1 (2010).
  • Björkhem I, Meaney S. Brain cholesterol: long secret life behind a barrier. Arterioscler Thromb Vasc Biol. 2004 May;24(5):806–815.
  • Russell DW, Halford RW, Ramirez DM, et al. Cholesterol 24-hydroxylase: an enzyme of cholesterol turnover in the brain. Annu Rev Biochem. 2009;78(1):1017–1040.
  • Gilch S, Karri S Treatment of a prion disease. WO2018209445A1 (2018).
  • Altmeppen HC, Puig B, Dohler F, et al. Proteolytic processing of the prion protein in health and disease. Am J Neurodegener Dis. 2012;1(1):15–31.
  • Chen SG, Teplow DB, Parchi P, et al. Truncated forms of the human prion protein in normal brain and in prion diseases. J Biol Chem. 1995 Aug;270(32):19173–19180.
  • GC T, Rp G, Calpains as targets for inhibition of prion propagation. US2006234971A1 (2006).
  • Llorens F, Thüne K, Sikorska B, et al. Altered Ca. Acta Neuropathol Commun. 2017 04; 5(1): 35.
  • Heiseke A, Aguib Y, Schatzl HM. Autophagy, prion infection and their mutual interactions. Curr Issues Mol Biol. 2010;12(2):87–97.
  • Booth L, Roberts JL, Ecroyd H, et al. AR-12 inhibits chaperone proteins preventing virus replication and the accumulation of toxic misfolded proteins. J Clin Cell Immunol. 2016 Oct;7(5). https://doi.org/10.4172/2155-9899.1000454.
  • Schaetzl HM, Abdulrahman B, Gilch S, et al. Compositions and methods for reducing prion levels. WO2017151687A1 (2017).
  • Chen J, Lu M, Niu R New use of natural plant polyphenol and medicament and/or health care product containing natural plant polyphenol. WO02092110A1 (2002).
  • Corbi G, Conti V, Komici K, et al. Phenolic plant extracts induce sirt1 activity and increase antioxidant levels in the rabbit’s heart and liver. Oxid Med Cell Longev. 2018;2018:2731289.
  • Park SY, Seo JS, Jeong JK, et al. Resveratrol effective in treating and preventing prion diseases. WO2011105825A2 (2011).
  • Jeong JK, Moon MH, Lee YJ, et al. Autophagy induced by the class III histone deacetylase Sirt1 prevents prion peptide neurotoxicity. Neurobiol Aging. 2013 Jan;34(1):146–156.
  • Berteau O, Mulloy B. Sulfated fucans, fresh perspectives: structures, functions, and biological properties of sulfated fucans and an overview of enzymes active toward this class of polysaccharide. Glycobiology. 2003 Jun;13(6):29R–40R.
  • Doh-Ura K, Kuge T, Uomoto M, et al. Prophylactic effect of dietary seaweed Fucoidan against enteral prion infection. Antimicrob Agents Chemother. 2007 Jun;51(6):2274–2277.
  • Dohura K Agent for preventing development of prion disease, food additive and feed additive comprising the same. WO2006090815A1 (2006).
  • Legname G, Leita L, Sequi P Humic substances and therapeutic uses thereof. WO2011007320A1 (2011).
  • Teruya K, Oguma A, Nishizawa K, et al. A single subcutaneous injection of cellulose ethers administered long before infection confers sustained protection against prion diseases in rodents. PLoS Pathog. 2016 Dec;12(12):e1006045.
  • Dohura K Pharmaceutical composition for conformational disease. WO2007123187A1 (2007).
  • Frangione B, Wisniewski T, Sigurdsson EM Synthetic immunogenic but non-deposit-forming polypeptides and peptides homologous to amyloid beta, prion protein, amylin, alpha-synuclein, or polyglutamine repeats for induction of an immune response thereto. WO03045128A2 (2003).
  • De Silva BS, Egodage KL, Wilson GS. Purified protein derivative (PPD) as an immunogen carrier elicits high antigen specificity to haptens. Bioconjug Chem. 1999 May-Jun;10(3):496–501.
  • Raven NDH, Sutton JM, Murdoch H Treatment of TSE infection. WO03080665A2 (2003).
  • Sigurdsson EM, Chabalgoity JA, Goni FR, et al. Mucosal immunization to prevent prion infection. US2007059807A1 (2005).
  • Bachmann M, Maurer P, Pellicioli E, et al. Prion protein carrier-conjugates. WO03059386A2 (2003).
  • Wisniewski HM, Kascsak RJ, Rubenstein R, et al. Hybrid cell lines producing monoclonal antibodies dircted against scrapie-associated fibril proteins. US4806627A (1989).
  • Korth C, Stierli B, Streit P, et al. Prion (PrPSc)-specific epitope defined by a monoclonal antibody. Nature. 1997 Nov;390(6655):74–77. .
  • Paramithiotis E, Pinard M, Lawton T, et al. A prion protein epitope selective for the pathologically misfolded conformation. Nat Med. 2003 Jul;9(7):893–899.
  • Marciniuk K, Määttänen P, Taschuk R, et al. Development of a multivalent, PrP(Sc)-specific prion vaccine through rational optimization of three disease-specific epitopes. Vaccine. 2014 Apr;32(17):1988–1997.
  • Tayebi M Antibodies specific for misfolded proteins and methods for their production. WO2010015834A1 (2010).
  • Schaetzl H Prion protein dimers useful for vaccination. US2002168377A1 (2002).
  • Aguzzi A, Genoud N, Raeber A, Soluble hybrid prion proteins and their use in the diagnosis, prevention and treatment of transmissible spongiform encephalopathies. WO2004081052A1. (2004).
  • Büeler H, Aguzzi A, Sailer A, et al. Mice devoid of PrP are resistant to scrapie. Cell. 1993 Jul;73(7):1339–1347.
  • Sethi S, Lipford G, Wagner H, et al. Postexposure prophylaxis against prion disease with a stimulator of innate immunity. Lancet. 2002 Jul;360(9328):229–230.
  • Sigurdsson EM, Brown DR, Daniels M, et al. Immunization delays the onset of prion disease in mice. Am J Pathol. 2002 Jul;161(1):13–17.
  • Collinge J, Hawke S. Prion Inhibition. WO2004050120A2. (2004).
  • Liu T, Zwingman T, Li R, et al. Differential expression of cellular prion protein in mouse brain as detected with multiple anti-PrP monoclonal antibodies. Brain Res. 2001 Mar;896(1–2):118–129.
  • Williamson RA, Peretz D, Pinilla C, et al. Mapping the prion protein using recombinant antibodies. J Virol. 1998 Nov;72(11):9413–9418.
  • Enari M, Flechsig E, Weissmann C. Scrapie prion protein accumulation by scrapie-infected neuroblastoma cells abrogated by exposure to a prion protein antibody. Proc Natl Acad Sci U S A. 2001 Jul;98(16):9295–9299.
  • Heppner FL, Musahl C, Arrighi I, et al. Prevention of scrapie pathogenesis by transgenic expression of anti-prion protein antibodies. Science. 2001 Oct;294(5540):178–182.
  • Prusiner SB, Peretz D, Williamson RA, et al. Antibodies abolish prion propagation and promote clearance of infectivity. WO02087502A2 (2002).
  • Perrier V, Solassol J, Crozet C, et al. Anti-PrP antibodies block PrPSc replication in prion-infected cell cultures by accelerating PrPC degradation. J Neurochem. 2004 Apr;89(2):454–463.
  • Korth CDR, Stitz L, Petsch B, et al. Anti-prion protein antibody fragment. WO2009153029A1 (2009).
  • Soto C, Sigurdsson EM, Morelli L, et al. Beta-sheet breaker peptides inhibit fibrillogenesis in a rat brain model of amyloidosis: implications for Alzheimer’s therapy. Nat Med. 1998 Jul;4(7):822–826.
  • Soto C, Kascsak RJ, Saborío GP, et al. Reversion of prion protein conformational changes by synthetic beta-sheet breaker peptides. Lancet. 2000 Jan;355(9199):192–197.
  • Adessi C, Halazy S, Saborio G, et al. Prion inhibiting peptides and derivatives thereof. WO03050139A2 (2003).
  • Lundberg P, Magzoub M, Lindberg M, et al. Cell membrane translocation of the N-terminal (1–28) part of the prion protein. Biochem Biophys Res Commun. 2002 Nov;299(1):85–90.
  • Magzoub M, Sandgren S, Lundberg P, et al. N-terminal peptides from unprocessed prion proteins enter cells by macropinocytosis. Biochem Biophys Res Commun. 2006 Sep;348(2):379–385.
  • Löfgren K, Wahlström A, Lundberg P, et al. Antiprion properties of prion protein-derived cell-penetrating peptides. FASEB J. 2008 Jul;22(7):2177–2184.
  • Adenot M, Merida P, Lahana R. Applications of a blood-brain barrier technology platform to predict CNS penetration of various chemotherapeutic agents. 2. Cationic peptide vectors for brain delivery. Chemotherapy. 2007;53(1):73–76.
  • Loefgren K, Wahlstroem A, Lundberg P, et al. Prion protein derived cell penetrating peptides and their uses. WO2009041902A1. (2009).
  • Lim YB, Ryou C Intracellular pH-responsive fusion peptide and pharmaceutical composition for reducing abnormal prion protein aggregation or misfolding. US10232012B2 (2019).
  • Pastrana Gonzalez MA Use of polymerised protein particles generated from prion protein for the treatment of prion diseases. WO2014170521A1 (2014).
  • Keefe AD, Pai S, Ellington A. Aptamers as therapeutics. Nat Rev Drug Discov. 2010 07;9(7):537–550.
  • Dj K, Jg S, Legname G, et al. Thioaptamer interactions with prion proteins: sequence-specific and non-specific binding sites. J Mol Biol. 2007 Jun;369(4):1001–1014.
  • James WS Ligands specific for an isoform of the prion protein. WO0188123A1 (2001).
  • Proske D Prion protein-specific aptamers. EP1333090A1 (2003).
  • Mashima T, Nishikawa F, Kamatari YO, et al. Anti-prion activity of an RNA aptamer and its structural basis. Nucleic Acids Res. 2013 Jan;41(2):1355–1362.
  • Mashima T, Lee JH, Kamatari YO, et al. Development and structural determination of an anti-PrP. Sci Rep. 2020 03; 10(1): 4934.
  • Amato J, Mashima T, Kamatari YO, et al. Improved anti-prion nucleic acid aptamers by incorporation of chemical modifications. Nucleic Acid Ther. 2020 12; 30(6): 414–421.
  • Hannon GJ. RNA interference. Nature. 2002 Jul;418(6894):244–251.
  • Brummelkamp TR, Bernards R, Agami R. A system for stable expression of short interfering RNAs in mammalian cells. Science. 2002 Apr;296(5567):550–553.
  • Tilly G, Chapuis J, Vilette D, et al. Efficient and specific down-regulation of prion protein expression by RNAi. Biochem Biophys Res Commun. 2003 Jun;305(3):548–551.
  • Sakaguchi S, Taira K Method of inhibiting prion protein. US2005053583A1 (2005).
  • Lehmann S, Relano-Gines A, Resina S, et al. Systemic delivery of siRNA down regulates brain prion protein and ameliorates neuropathology in prion disorder. PLoS One. 2014;9(2):e88797.
  • Pulford B, Reim N, Bell A, et al. Liposome-siRNA-peptide complexes cross the blood-brain barrier and significantly decrease PrP on neuronal cells and PrP in infected cell cultures. PLoS One. 2010 Jun;5(6):e11085.
  • Bender H, Noyes N, Annis JL, et al. PrPC knockdown by liposome-siRNA-peptide complexes (LSPCs) prolongs survival and normal behavior of prion-infected mice immunotolerant to treatment. PLoS One. 2019;14(7):e0219995. .
  • Leavitt BR, Tabrizi SJ. Antisense oligonucleotides for neurodegeneration. Science. 2020 Mar;367(6485):1428–1429.
  • Bennett CF, Hung G Modulation of prion expression. WO2010019270A1 (2010).
  • Nazor Friberg K, Hung G, Wancewicz E, et al. Intracerebral infusion of antisense oligonucleotides into prion-infected mice. Mol Ther Nucleic Acids. 2012 Feb;1:e9.
  • Raymond GJ, Zhao HT, Race B, et al. Antisense oligonucleotides extend survival of prion-infected mice. JCI Insight. 2019 7; 5:https://doi.org/10.1172/jci.insight.131175.
  • Minikel EV, Zhao HT, Le J, et al. Prion protein lowering is a disease-modifying therapy across prion disease stages, strains and endpoints. Nucleic Acids Res. 2020 11; 48(19): 10615–10631.
  • Jucker M, Walker LC. Self-propagation of pathogenic protein aggregates in neurodegenerative diseases. Nature. 2013 Sep;501(7465):45–51.
  • Ayers JI, Paras NA, Prusiner SB. Expanding spectrum of prion diseases. Emerg Top Life Sci. 2020 09;4(2):155–167.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.