396
Views
3
CrossRef citations to date
0
Altmetric
Reviews

Proteasome inhibitor patents (2010 – present)

, , &

Bibliography

  • Rubin DM, Finley D. The proteasome: a protein-degrading organelle? Curr Biol 1995;5:854-4
  • Baumeister W, Walz J, Proteolysis C. The proteasome: compartmentalizing protease. Cell 1998;92:367-80
  • Groll M, Ditzel L, Lowe J, et al. Structure of 20S proteasome from yeast at 2.4 A resolution. Nature 1997;386(6624):463-71
  • Elsasser S, Gali RR, Schwickart M, et al. Proteasome subunit Rpn1 binds ubiquitin-like protein domains. Nat Cell Biol 2002;4:725-30
  • Pickart CM, Cohen RE. Proteasomes and their kin: proteases in the machine age. Nat Rev Mol Cell Biol 2004;5:177-87
  • Bose S, Stratford F, Broadfoot K, et al. Phosphorylation of 20S proteasome alpha subunit C8 (alpha7) stabilizes the 26S proteasome and plays a role in the regulation of proteasome complexes by gamma-interferon. Biochem J 2004;378:177-84
  • Borissenko L, Groll M. 20S proteasome and its inhibitors: crystallographic knowledge for drug development. Chem Inform 2007;38:687-717
  • Tsvetkov L, Nanjundan M, Domino M, et al. The ubiquitin-proteasome system and assays to determine responses to inhibitors. Expert Opin Drug Discov 2010;5:1221-36
  • Kisselev AF, van der Linden WA, Overkleeft HS. Proteasome inhibitors: an expanding army attacking a unique target. Chem Biol 2012;19:99-115
  • Kloetzel PM, Ossendorp F. Proteasome and peptidase function in MHC-class-I-mediated antigen presentation. Curr Opin Immunol 2004;16:76-81
  • Murata S, Takahama Y, Tanaka K. Thymoproteasome: probable role in generating positively selecting peptides. Curr Opin Immunol 2008;20:192-6
  • An W, Hwang S, Trepel J, et al. Protease inhibitor-induced apoptosis: accumulation of wt p53, p21WAF1/CIP1, and induction of apoptosis are independent markers of proteasome inhibition. Leukemia 2000;14:1276
  • Rajkumar SV, Richardson PG, Hideshima T, et al. Proteasome inhibition as a novel therapeutic target in human cancer. J Clin Oncol 2005;23:630-9
  • Almond J, Cohen G. The proteasome: a novel target for cancer chemotherapy. Leukemia 2002;16:433
  • Paul S. Dysfunction of the ubiquitin–proteasome system in multiple disease conditions: therapeutic approaches. Bioessays 2008;30:1172-84
  • Willis MS, Townley-Tilson WD, Kang EY, et al. Sent to destroy: the ubiquitin proteasome system regulates cell signaling and protein quality control in cardiovascular development and disease. Circ Res 2010;106:463-78
  • Yu X, Kem DC. Proteasome inhibition during myocardial infarction. Cardiovasc Res 2010;85:312-20
  • Flechner SM, Fatica R, Askar M, et al. The role of proteasome inhibition with bortezomib in the treatment of antibody-mediated rejection after kidney-only or kidney-combined organ transplantation. Transplantation 2010;90:1486-92
  • Pevzner Y, Metcalf R, Kantor M, et al. Recent advances in proteasome inhibitor discovery. Expert Opin Drug Discov 2013;8:537-68
  • Kisselev AF, Goldberg AL. Proteasome inhibitors: from research tools to drug candidates. Chem Biol 2001;8:739-58
  • Adams J. Potential for proteasome inhibition in the treatment of cancer. Drug Discov Today 2003;8(7):307-15
  • Zhu Y, Zhao X, Zhu X, et al. Design, synthesis, biological evaluation, and structure- activity relationship (SAR) discussion of dipeptidyl boronate proteasome inhibitors, part I: comprehensive understanding of the SAR of alpha-amino acid boronates. J Med Chem 2009;52:4192-9
  • Mujtaba T, Dou Q. Advances in the understanding of mechanisms and therapeutic use of bortezomib. Discov Med 2011;12:471-80
  • Shah MH, Young D, Kindler HL, et al. Phase II study of the proteasome inhibitor bortezomib (PS-341) in patients with metastatic neuroendocrine tumors. Clin Cancer Res 2004;10:6111-18
  • Kondagunta GV, Drucker B, Schwartz L, et al. Phase II trial of bortezomib for patients with advanced renal cell carcinoma. J Clin Oncol 2004;22:3720-5
  • Markovic SN, Geyer SM, Dawkins F, et al. A phase II study of bortezomib in the treatment of metastatic malignant melanoma. Cancer 2005;103:2584-9
  • Li R, Cui J, Zhu Y, et al. Tripetide boronic acid or boronic ester, preparative method and use thereof. Google Patents. US13379142; 2010
  • Bernardini R, Bernareggi A, Cassara PG, et al. Proteasome inhibitors and methods of using the same. US8058262; 2011
  • Olhava EJ, Danca MD. Proteasome inhibitors. US13209511; 2011
  • Shenk KD, Paralati F, Bennett MK. Compounds for immunoproteasome inhibition. US0072422; 2013
  • Soppimath K, Pejaver S, Patel KR, et al. Stable Bortezomib Formulations. US0322763; 2012
  • Fostier K, De Becker A, Schots R. Carfilzomib: a novel treatment in relapsed and refractory multiple myeloma. Oncol Targets Ther 2012;5:237
  • Kuhn DJ, Chen Q, Voorhees PM, et al. Potent activity of carfilzomib, a novel, irreversible inhibitor of the ubiquitin-proteasome pathway, against preclinical models of multiple myeloma. Blood 2007;110:3281-90
  • Demo SD, Kirk CJ, Aujay MA, et al. Antitumor activity of PR-171, a novel irreversible inhibitor of the proteasome. Cancer Res 2007;67:6383-91
  • Kirk CJ, Jiang J. Use of Peptide Epoxyketones for Metastasis Suppression. US0035295; 2013
  • Shenk KD, Parlati F, Zhou HJ, et al. Compounds for enzyme inhibition. US8080576; 2011
  • Smyth MS, Laidig GJ. Compounds for proteasome enzyme inhibition. US8198270; 2012
  • Smyth MS, Laidig GJ. Compounds for enzyme inhibition. US8207125; 2012
  • Smyth MS, Laidig GJ. Compounds for enzyme inhibition. US8207127; 2012
  • Kisselev A, Filippov DV, Overkleeft H. Inhibitors of the trypsin-like site of the proteasome and methods of use thereof. US8455431; 2013
  • US Food and Drug Administration. Approved Drugs - Carfilzomib. 2012. Available from: http://www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm312945.htm
  • Guo S, Lu J, Subramanian A, et al. Microarray-assisted pathway analysis identifies mitogen-activated protein kinase signaling as a mediator of resistance to the green tea polyphenol epigallocatechin 3-gallate in her-2/neu–overexpressing breast cancer cells. Cancer Res 2006;66:5322-9
  • Sun CL, Yuan JM, Koh WP, et al. Green tea, black tea and breast cancer risk: a meta-analysis of epidemiological studies. Carcinogenesis 2006;27:1310-15
  • Sartippour MR, Heber D, Ma J, et al. Green tea and its catechins inhibit breast cancer xenografts. Nutr Cancer 2001;40:149-56
  • Landau JM, Wang ZY, Yang GY, et al. Inhibition of spontaneous formation of lung tumors and rhabdomyosarcomas in A/J mice by black and green tea. Carcinogenesis 1998;19:501-7
  • Horie N, Hirabayashi N, Takahashi Y, et al. Synergistic effect ofgreen tea catechins on cell growth and apoptosis induction in gastric carcinoma cells. Biol Pharm Bull 2005;28:574-9
  • Bettuzzi S, Brausi M, Rizzi F, et al. Chemoprevention of human prostate cancer by oral administration of green tea catechins in volunteers with high-grade prostate intraepithelial neoplasia: a preliminary report from a one-year proof-of-principle study. Cancer Res 2006;66:1234-40
  • Kemberling J, Hampton JA, Keck RW, et al. Inhibition of bladder tumor growth by the green tea derivative epigallocatechin-3-gallate. J Urol 2003;170:773-6
  • Smith DM, Daniel KG, Wang Z, et al. Docking studies and model development of tea polyphenol proteasome inhibitors: applications to rational drug design. Proteins 2003;54:58-70
  • Nam S, Smith DM, Dou QP. Ester bond-containing tea polyphenols potently inhibit proteasome activity in vitro and in vivo. J Biol Chem 2001;276:13322-30
  • Bonfili L, Cecarini V, Amici M, et al. Natural polyphenols as proteasome modulators and their role as anti-cancer compounds. FEBS J 2008;275:5512-26
  • Chan TH, Dou QP. Polyphenol compounds for inhibiting proteasome and uses thereof. US12968759; 2010
  • Chan TH, Lam WH, Chow LMC, et al. (-)-epigallocatechin gallate derivatives for inhibiting proteasome. US8193377; 2012
  • Dou QP, Chan TH, Smith DM. Polyphenol proteasome inhibitors, synthesis, and methods of use. US13271058; 2011
  • Milacic V, Chen D, Ronconi L, et al. A novel anticancer gold(III) dithiocarbamate compound inhibits the activity of a purified 20S proteasome and 26S proteasome in human breast cancer cell cultures and xenografts. Cancer Res 2006;66:10478-86
  • Milacic V, Dou QP. The tumor proteasome as a novel target for gold (III) complexes: implications for breast cancer therapy. Coord Chem Rev 2009;253:1649-60
  • Cvek B, Milacic V, Taraba J, et al. Ni (II), Cu (II), and Zn (II) diethyldithiocarbamate complexes show various activities against the proteasome in breast cancer cells. J Med Chem 2008;51:6256-8
  • Frezza M, Hindo SS, Tomco D, et al. Comparative activities of nickel (II) and zinc (II) complexes of asymmetric [NN O] ligands as 26S proteasome inhibitors. Inorg Chem 2009;48:5928-37
  • Daniel KG, Gupta P, Harbach RH, et al. Organic copper complexes as a new class of proteasome inhibitors and apoptosis inducers in human cancer cells. Biochem Pharmacol 2004;67:1139-51
  • Daniel KG, Chen D, Orlu S, et al. Clioquinol and pyrrolidine dithiocarbamate complex with copper to form proteasome inhibitors and apoptosis inducers in human breast cancer cells. Breast Cancer Res 2005;7:R897-908
  • Milacic V, Jiao P, Zhang B, et al. Novel 8-hydroxylquinoline analogs induce copper-dependent proteasome inhibition and cell death in human breast cancer cells. Int J Oncol 2009;35:1481-91
  • Hindo SS, Frezza M, Tomco D, et al. Metals in anticancer therapy: copper (II) complexes as inhibitors of the 20S proteasome. Eur J Med Chem 2009;44:4353-61
  • Zuo J, Bi C, Fan Y, et al. Cellular and computational studies of proteasome inhibition and apoptosis induction in human cancer cells by amino acid schiff base-copper complexes. J Inorg Biochem 2013;118:83-93
  • Hollingshead RGW. Studies on derivatives of 8-hydroxyquinoline (oxine). The sensitivity and selectivity of 5-nitroso-oxine and 2-methyl-5-nitroso-oxine towards the group IIIB metals. Anal Chim Acta 1955;12:401-7
  • Schimmer AD. Use of 5AHQ and bortezomib for the treatment of hematological diseases. US13203878; 2010
  • Sheshbaradaran H. Combination therapy with A proteasome inhibitor and a gallium complex. US0096068; 2013
  • Feling RH, Buchanan GO, Mincer TJ, et al. Salinosporamide A: a highly cytotoxic proteasome inhibitor from a novel microbial source, a marine bacterium of the new genus Salinospora. Angew Chem Int Ed 2003;42:355-7
  • Fenical W, Jensen PR, Palladino MA, et al. Discovery and development of the anticancer agent salinosporamide A (NPI-0052). Bioorg Med Chem 2009;17:2175-80
  • Lam KS, Lloyd GK, Neuteboom STC, et al. From natural product to clinical trials: NPI-0052 (salinosporamide A), a marine actinomycete-derived anticancer agent. Chapter 12. Nat Prod Chem Drug Discov 2010;355-73
  • Macherla VRR, Potts BC, Manam RR, et al. Proteasome inhibitors. US8227503; 2012
  • Ling T, Macherla VRR, Potts BC, et al. Total synthesis of salinosporamide A and analogs thereof. US8067616; 2011
  • Fenical WH, Jensen PR, Mincer TJ, et al. Salinosporamides and methods for use thereof. US8217072; 2012
  • Chauhan D, Singh A, Brahmandam M, et al. Combination of proteasome inhibitors bortezomib and NPI-0052 trigger in vivo synergistic cytotoxicity in multiple myeloma. Blood 2008;111:1654-64
  • Ruiz S, Krupnik Y, Keating M, et al. The proteasome inhibitor NPI-0052 is a more effective inducer of apoptosis than bortezomib in lymphocytes from patients with chronic lymphocytic leukemia. Mol Cancer Ther 2006;5:1836-43
  • Miller CP, Ban K, Dujka ME, et al. NPI-0052, a novel proteasome inhibitor, induces caspase-8 and ROS-dependent apoptosis alone and in combination with HDAC inhibitors in leukemia cells. Blood 2007;110:267-77
  • Miller CP, Rudra S, Keating MJ, et al. Caspase-8 dependent histone acetylation by a novel proteasome inhibitor, NPI-0052: a mechanism for synergy in leukemia cells. Blood 2009;113:4289-99
  • Sallam AA, Ramasahayam S, Meyer SA, et al. Design, synthesis, and biological evaluation of dibromotyrosine analogues inspired by marine natural products as inhibitors of human prostate cancer proliferation, invasion, and migration. Bioorg Med Chem 2010;18:7446-57
  • Mudit M, Khanfar M, Muralidharan A, et al. Discovery, design, and synthesis of anti-metastatic lead phenylmethylene hydantoins inspired by marine natural products. Bioorg Med Chem 2009;17:1731-8
  • Lawrence HR, Sebti SM, Ozcan S, et al. Proteasome chymotrypsin-like inhibition using PI-1833 analogs. WO129564; 2012
  • Lawrence H, Ge Y, Sebti S, et al. Proteasome inhibitors having chymotrypsin-like activity. WO102286; 2010
  • Bachovchin W, Lai HS, Poplawski S, et al. FAP-activated proteasome inhibitors for treating solid tumors. WO033396; 2013
  • Scanlan MJ, Raj B, Calvo B, et al. Molecular cloning of fibroblast activation protein alpha, a member of the serine protease family selectively expressed in stromal fibroblasts of epithelial cancers. Proc Natl Acad Sci 1994;91:5657-61
  • Ravasz E, Somera AL, Mongru DA, et al. Hierarchical organization of modularaity in metabolic networks. Science 2002;297:1551-5
  • Jeong H, Tombor B, Albert R, et al. The large-scale organization of metabolic networks. Nature 2000;407:651-4
  • Barabasi AL, Oltvai ZN. Network biology: understanding the cell's functional organization. Nat Rev Genet 2004;5:101-13
  • Ma H, Zeng AP. Reconstruction of metabolic networks from genome data and analysis of their global structure for various organisms. Bioinformatics 2003;19:270-7
  • Taylor KB. Enzyme kinetics and mechanisms. 2002; Kluwer Academic Publishers, Norwell: MA, USA
  • Gottesman MM. Mechanisms of cancer drug resistance. Annu Rev Med 2002;53:615-27
  • Dean M, Fojo T, Bates S. Annual review of medicine. Tumour stem cells and drug resistance. Nat Rev Cancer 2005;5:275-84
  • Fertig G, Friess T, Klein C, et al. Combination therapy of a type II anti-CD20 antibody with a proteasome inhibitor. US0219549; 2012
  • Forster C. Means and methods for treating ischemic conditions. US0245078; 2012
  • Piva R, Ruggeri B, Williams M, et al. CEP-18770: a novel, orally active proteasome inhibitor with a tumor-selective pharmacologic profile competitive with bortezomib. Blood 2008;111:2765-75
  • Gallerani E, Zucchetti M, Brunelli D, et al. A first in human phase I study of the proteasome inhibitor CEP-18770 in patients with advanced solid tumours and multiple myeloma. Eur J Cancer 2013;49(2):290-6
  • Ruggeri B, Seavey M. Proteasome inhibitor delanzomib for use in the treatment of lupus. WO119056; 2012
  • Li R, Yang N, Zhang L, et al. Inhibition of Jak/STAT signaling ameliorates mice experimental nephrotic syndrome. Am J Nephrol 2007;27:580-9
  • Nikrad M, Johnson T, Puthalalath H, et al. The proteasome inhibitor bortezomib sensitizes cells to killing by death receptor ligand TRAIL via BH3-only proteins Bik and Bim. Mol Cancer Ther 2005;4:443-9
  • Williams S, Pettaway C, Song R, et al. Differential effects of the proteasome inhibitor bortezomib on apoptosis and angiogenesis in human prostate tumor xenografts. Mol Cancer Ther 2003;2:835-43
  • Broyl A, Corthals SL, Jongen JL, et al. Mechanisms of peripheral neuropathy associated with bortezomib and vincristine in patients with newly diagnosed multiple myeloma: a prospective analysis of data from the HOVON-65/GMMG-HD4 trial. Lancet Oncol 2010;11:1057-65
  • Nawrocki ST, Carew JS, Pino MS, et al. Bortezomib sensitizes pancreatic cancer cells to endoplasmic reticulum stress-mediated apoptosis. Cancer Res 2005;65:11658-66
  • Lonial S, Waller EK, Richardson PG, et al. Risk factors and kinetics of thrombocytopenia associated with bortezomib for relapsed, refractory multiple myeloma. Blood 2005;106:3777-84
  • Zangari M, Yaccoby S, Cavallo F, et al. Response to bortezomib and activation of osteoblasts in multiple myeloma. Clin Lymphoma Myeloma 2006;7:109-14
  • Takeda K, Mori A, Yanagida M. Identification of genes affecting the toxicity of anti-cancer drug bortezomib by genome-wide screening in S. pombe. PloS One 2011;6:e22021
  • McConkey DJ. A novel role for a familiar protein in apoptosis induced by proteasome inhibition. Cancer Cell 2008;14:1-2
  • Shoshan MC, Linder S. Target specificity and off-target effects as determinants of cancer drug efficacy. Expert Opin Drug Metab Toxicol 2008;4(3):273-80
  • Dick LR, Fleming PE. Building on bortezomib: second-generation proteasome inhibitors as anti-cancer therapy. Drug Discov Today 2010;15:243-9
  • Arastu-Kapur S, Anderl JL, Kraus M, et al. Nonproteasomal targets of the proteasome inhibitors bortezomib and carfilzomib: a link to clinical adverse events. Clin Cancer Res 2011;17:2734-43
  • Chen S, Blank JL, Peters T, et al. Genome-wide siRNA screen for modulators of cell death induced by proteasome inhibitor bortezomib. Cancer Res 2010;70:4318-26
  • Nickeleit I, Zender S, Sasse F, et al. Argyrin A reveals a critical role for the tumor suppressor protein p27 in mediating antitumor activities in response to proteasome inhibition. Cancer Cell 2008;14:23-35

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.