De-Novo Design of Cereblon (CRBN) Effectors Guided by Natural Hydrolysis Products of Thalidomide DerivativesClick to copy article linkArticle link copied!
- Christopher HeimChristopher HeimDepartment of Protein Evolution, Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076 Tübingen, GermanyMore by Christopher Heim
- Dimanthi PliatsikaDimanthi PliatsikaFaculty for Chemistry und Mineralogy, Institute of Organic Chemistry, University of Leipzig, Johannisallee 29, 04103 Leipzig, GermanyMore by Dimanthi Pliatsika
- Farnoush MousavizadehFarnoush MousavizadehFaculty for Chemistry und Mineralogy, Institute of Organic Chemistry, University of Leipzig, Johannisallee 29, 04103 Leipzig, GermanyMore by Farnoush Mousavizadeh
- Kerstin BärKerstin BärDepartment of Protein Evolution, Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076 Tübingen, GermanyMore by Kerstin Bär
- Birte Hernandez AlvarezBirte Hernandez AlvarezDepartment of Protein Evolution, Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076 Tübingen, GermanyMore by Birte Hernandez Alvarez
- Athanassios Giannis*Athanassios Giannis*E-mail: [email protected] (A.G.).Faculty for Chemistry und Mineralogy, Institute of Organic Chemistry, University of Leipzig, Johannisallee 29, 04103 Leipzig, GermanyMore by Athanassios Giannis
- Marcus D. Hartmann*Marcus D. Hartmann*E-mail: [email protected] (M.D.H.).Department of Protein Evolution, Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076 Tübingen, GermanyMore by Marcus D. Hartmann
Abstract
Targeted protein degradation via cereblon (CRBN), a substrate receptor of an E3 ubiquitin ligase complex, is an increasingly important strategy in various clinical settings, in which the substrate specificity of CRBN is altered via the binding of small-molecule effectors. To date, such effectors are derived from thalidomide and confer a broad substrate spectrum that is far from being fully characterized. Here, we employed a rational and modular approach to design novel and minimalistic CRBN effectors. In this approach, we took advantage of the binding modes of hydrolyzed metabolites of several thalidomide-derived effectors, which we elucidated via crystallography. These yielded key insights for the optimization of the minimal core binding moiety and its linkage to a chemical moiety that imparts substrate specificity. Based on this scaffold, we present a first active de-novo CRBN effector that is able to degrade the neo-substrate IKZF3 in the cell culture.
Introduction
Results and Discussion
Biophysical and Structural Characterization of IMiD Analogues and Their Hydrolysis Products
Binding Mode of CBG, a Major Hydrolysis Product of Thalidomide
Rational Design of Novel Succinimide Effectors Guided by Hydrolyzed Metabolites
Degradation of Neo-Substrates
Conclusions
Experimental Section
Chemistry
General Procedures
General Procedure A
General Procedure B
General Procedure C
General Procedure D
General Procedure E
tert-Butyl (S)-(2,5-Dioxopyrrolidin-3-yl)carbamate (2a)
2,5-Dioxopyrrolidin-3-aminium 2,2,2-Trifluoroacetate (3a)
2-(2,5-Dioxopyrrolidin-3-yl)-4-nitroisoindoline-1,3-dione (4a)
2-(2,5-Dioxopyrrolidin-3-yl)-5-nitroisoindoline-1,3-dione (4b)
2-(2,6-Dioxopiperidin-3-yl)-4-nitroisoindoline-1,3-dione (4c)
2-(2,6-Dioxopiperidin-3-yl)-5-nitroisoindoline-1,3-dione (4d)
4-Amino-2-(2,5-dioxopyrrolidin-3-yl)isoindoline-1,3-dione (5a)
5-Amino-2-(2,5-dioxopyrrolidin-3-yl)isoindoline-1,3-dione (5b)
N-(2,5-Dioxopyrrolidin-3-yl)-3-methylbut-2-enamide (7a)
N-(2,5-Dioxopyrrolidin-3-yl)-2-phenylacetamide (7b)
2,4,6-Trichloro-N-(2,5-dioxopyrrolidin-3-yl)benzamide (7c)
Benzyl (2,5-Dioxopyrrolidin-3-yl)carbamate (7d)
N-(2,5-Dioxopyrrolidin-3-yl)-3,5-dinitrobenzamide (7e)
N-(2,5-Dioxopyrrolidin-3-yl)cinnamamide (7f)
3-Chloro-N-(2,5-dioxopyrrolidin-3-yl)benzo[b]thiophene-2-carboxamide (7g)
3,4-Dichloro-N-(2,5-dioxopyrrolidin-3-yl)benzo[b]thiophene-2-carboxamide (7h)
tert-Butyl (4-((((2,5-Dioxopyrrolidin-3-yl)carbamoyl)oxy)methyl)benzyl)carbamate (10a)
tert-Butyl (4-((((2,6-Dioxopiperidin-3-yl)carbamoyl)oxy)methyl)benzyl)carbamate (10b)
(4-((((2,5-Dioxopyrrolidin-3-yl)carbamoyl)oxy)methyl)phenyl)methanaminium 2,2,2-Trifluoroacetate (11a)
(4-((((2,6-Dioxopiperidin-3-yl)carbamoyl)oxy)methyl)phenyl)methanaminium 2,2,2-Trifluoroacetate (11b)
4-((4-((((2,5-Dioxopyrrolidin-3-yl)carbamoyl)oxy)methyl)benzyl)amino)-4-oxobutanoic Acid (12a)
4-((4-((((2,6-Dioxopiperidin-3-yl)carbamoyl)oxy)methyl)benzyl)amino)-4-oxobutanoic Acid (12b)
((1s,4s)-4-((tert-Butoxycarbonyl)amino)cyclohexyl)methyl (2,5-Dioxopyrrolidin-3-yl)carbamate (15a)
((1s,4s)-4-((tert-Butoxycarbonyl)amino)cyclohexyl)methyl (2,6-Dioxopiperidin-3-yl)carbamate (15b)
((1s,4s)-4-((((2,5-Dioxopyrrolidin-3-yl)carbamoyl)oxy)methyl)cyclohexyl)methanaminium 2,2,2-Trifluoroacetate (16a)
((1s,4s)-4-((((2,6-Dioxopiperidin-3-yl)carbamoyl)oxy)methyl)cyclohexyl)methanaminium 2,2,2-Trifluoroacetate (16b)
tert-Butyl (2S)-2-((((2,6-Dioxopiperidin-3-yl)carbamoyl)oxy)methyl)pyrrolidine-1-carboxylate (19b)
(2S)-2-((((2,6-Dioxopiperidin-3-yl)carbamoyl)oxy)methyl)pyrrolidin-1-ium 2,2,2-Trifluoroacetate (20b)
Cloning, Expression and Protein Purification
FRET Assay
Crystallography
MsCI4·4a | MsCI4·4b | MsCI4·5a | MsCI4·5b | MsCI4·7a | MsCI4·7b | MsCI4·7c | MsCI4·7d | MsCI4·7f | |
---|---|---|---|---|---|---|---|---|---|
Data Collection | |||||||||
space group | P212121 | P212121 | P212121 | P212121 | P212121 | P212121 | P212121 | P21 | P212121 |
Unit Cell | |||||||||
a, b, c (Å) | 56.46, 58.82, 88.23 | 56.75, 58.82, 88.57 | 56.95, 59.58, 89.05 | 56.66, 58.71, 88.01 | 56.33, 58.68, 89.27 | 56.42, 58.80, 88.23 | 56.33, 58.68, 89.27 | 31.63, 52.39, 59.29 | 56.87, 58.55, 88.28 |
α, β, γ (deg) | 90, 90, 90 | 90, 90, 90 | 90, 90, 90 | 90, 90, 90 | 90, 90, 90 | 90, 90, 90 | 90, 90, 90 | 90.0, 95.8, 90.0 | 90, 90, 90 |
resolution range, Å | 44.1–1.55 (1.64–1.55) | 44.28–1.6 (1.7–1.6) | 44.53–1.7 (1.8–1.7) | 47.64–1.75 (1.85–1.75) | 49.03–1.8 (1.86–1.8) | 48.93–1.73 (1.84–1.73) | 49.03–1.85 (1.96–1.85) | 39.2–1.1 (1.16–1.1) | 44.14–1.65 (1.75–1.65) |
redundancy | 12.7 (11.8) | 12.9 (12.9) | 12.4 (12.8) | 12.8 (12.8) | 12.86 (12) | 12.5 (12.0) | 12.9 (13.0) | 6.3 (5.3) | 12.9 (13.2) |
completeness % | 99.8 (98.9) | 99.9 (99.4) | 99.0 (98.2) | 99.6 (97.7) | 99.6 (97.6) | 98.8 (92.4) | 99.8 (98.6) | 99.1 (95.1) | 99.9 (99.6) |
R merge % | 9.0 (98.2) | 5.3 (82.8) | 7.3 (94.6) | 6.6 (98.5) | 7.2 (89.0) | 14.2 (88.8) | 6.1 (102.8) | 5.3 (39.4) | 6.9 (81.5) |
CC (1/2) | 99.9 (85.4) | 100 (90.4) | 99.9 (87.8) | 100 (87.6) | 100 (84.6) | 99.6 (85.7) | 99.9 (78.1) | 99.8 (92.4) | 99.9 (86.6) |
I/s(I) | 15.09 (1.78) | 15.09 (1.78) | 20.22 (2.02) | 20.47 (2.27) | 22.12 (2.39) | 10.55 (1.73) | 21.46 (2.25) | 17.67 (3.64) | 20.72 (2.35) |
Refinement | |||||||||
number of reflections (total/test) | 43 370 (4254) | 39 841 (3904) | 33 803 (3354) | 30 301 (2985) | 28 122 (2771) | 30 582 (2948) | 28 122 (2771) | 77 822 (7458) | 36 182 (3558) |
no. of atoms | 2871 | 2787 | 2767 | 2709 | 2544 | 2778 | 2675 | 2082 | 2464 |
protein | 2572 | 2519 | 2504 | 2523 | 2338 | 2521 | 2468 | 1768 | 2229 |
solvent | 223 | 173 | 185 | 134 | 175 | 187 | 123 | 276 | 166 |
ligand | 76 | 95 | 78 | 52 | 31 | 70 | 84 | 38 | 69 |
R work % | 0.17 | 0.19 | 0.18 | 0.19 | 0.17 | 0.17 | 0.18 | 0.12 | 0.20 |
R free % | 0.20 | 0.22 | 0.23 | 0.23 | 0.22 | 0.23 | 0.22 | 0.15 | 0.22 |
Ligand in Chain | |||||||||
A | thalidomide | hydrolyzed 4b | thalidomide | 5b | 7a | thalidomide | 7c | thalidomide | |
B | 4a | hydrolyzed 4b | hydrolyzed 5a | 5b | 7a | 7b | 7c | co-crystal with 7d | 7f |
C | hydrolyzed 4a | hydrolyzed 4b | 5a | thalidomide | 7c | ||||
PDB ID | 6R0S | 6R0V | 6R0U | 6R11 | 6R1X | 6R12 | 6R1K | 6R1D | 6R13 |
MsCI4·11a | MsCI4·12a | MsCI4·16b | MsCI4·20a | MsCI4·20b | MsCI4·CBG | ||||
---|---|---|---|---|---|---|---|---|---|
Data Collection | |||||||||
space group | P212121 | P212121 | P212121 | P212121 | P212121 | P21 | |||
Unit Cell | |||||||||
a, b, c (Å) | 56.63, 59.59, 88.90 | 56.66, 59.02, 88.59 | 56.53, 59.44, 88.36 | 56.56, 59.23, 88.43 | 56.56, 59.3, 88.0 | 61.7, 59.1, 61.7 | |||
α, β, γ (deg) | 90, 90, 90 | 90, 90, 90 | 90, 90, 90 | 90, 90, 90 | 90, 90, 90 | 90, 105.6, 90 | |||
resolution range, Å | 44.45–1.35 (1.43–1.35) | 49.12–1.5 (1.59–1.5) | 47.62–1.35 (1.43–1.35) | 49.21–1.45 (1.50–1.45) | 49.18–1.54 (1.64–1.54) | 49.17–1.50 (1.59–1.50) | |||
redundancy | 12.7 (12.4) | 12.6 (12.6) | 12.6 (12.7) | 12.8 (12.9) | 12.6 (12.1) | 3.4 (3.1) | |||
completeness % | 99.9 (99.3) | 99.5 (97.2) | 99.8 (99.1) | 99.8 (98.5) | 99.5 (96.7) | 99.0 (97.5) | |||
R merge % | 7.4 (96.3) | 11.4 (78.6) | 7.9 (98.8) | 9.5 (89.4) | 13.1 (81.5) | 8.0 (88.0) | |||
CC (1/2) | 99.9 (83.2) | 99.8 (86.5) | 99.9 (84.0) | 99.8 (86.2) | 99.7 (83.8) | 99.7 (83.8) | |||
I/s(I) | 17.62 (1.97) | 14.29 (2.32) | 16.63 (1.85) | 14.83 (2.11) | 10.28 (1.71) | 9.70 (1.15) | |||
Refinement | |||||||||
number of reflections (total/test) | 66 738 (6544) | 48 282 (4739) | 66 083 (6516) | 53 344 (5211) | 44 026 (4112) | 68 399 (6707) | |||
no. of atoms | 2977 | 2995 | 3057 | 3074 | 2928 | 3985 | |||
protein | 2619 | 2665 | 2638 | 2634 | 2617 | 3480 | |||
solvent | 281 | 252 | 338 | 339 | 232 | 421 | |||
ligand | 77 | 78 | 81 | 101 | 79 | 84 | |||
R work % | 0.16 | 0.21 | 0.16 | 0.17 | 0.18 | 0.16 | |||
R free % | 0.19 | 0.24 | 0.20 | 0.20 | 0.21 | 0.20 | |||
Ligand in Chain | |||||||||
A | thalidomide | thalidomide | thalidomide | thalidomide | thalidomide | ||||
B | 10a | 12a | 16b | 20a | thalidomide | co-crystal with CBG | |||
C | thalidomide | thalidomide | 16b | thalidomide | 20b | ||||
PDB ID | 6R18 | 6R1C | 6R1W | 6R19 | 6R1A | 6R0Q |
Cell Culture and Western Blot
Supporting Information
The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acs.jmedchem.9b00454.
HPLC traces of tested compounds, western blot quantification and normalization IKZF3, concentration response curve for IKZF3 mAb, and Ki values of tested compounds (PDF)
Molecular formula strings (CSV)
Crystal structures have been deposited in the protein data bank (PDB) under the accession codes 6R0S (4a), 6R0V (4b), 6R0U (5a), 6R11 (5b), 6R1X (7a), 6R12 (7b), 6R1K (7c), 6R1D (7d), 6R13 (7f), 6R18 (11a), 6R1C (12a), 6R1W (16b), 6R19 (20a), and 6R1A (20b), 6R0Q (CBG). The authors will release the atomic coordinates and experimental data upon article publication.
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
We thank Andrei Lupas for continuing support, Reinhard Albrecht for assistance with crystallization and crystallographic data collection, and the staff of beamline X10SA of the Swiss Light Source (PSI, Villigen, Switzerland) for excellent technical support. This work was supported by institutional funds of the Max Planck Society.
4-DMAP | 4-dimethylaminopyridine |
AML | acute myeloid leukemia |
APP | amyloid precursor protein |
ASU | asymmetric unit |
BET | bromodomain and extraterminal domain |
CBG | α-(2-carboxybenzamido)glutarimide |
CDI | N,N-carbodiimidazole |
CK1α | casein kinase 1α |
CRBN | cereblon |
CRL4CRBN | CUL4-RBX1-DDB1-CRBN |
DIPEA | N,N-diisopropylethylamine |
Et3N | triethylamine |
EtOAc | ethyl acetate |
FBS | fetal bovine serum |
GSPT1 | eukaryotic peptide chain release factor GTP-binding subunit ERF3A |
IKZF1 | IKAROS family zinc finger 1 |
IKZF3 | zinc finger protein Aiolos |
IMiD | immunomodulatory drug |
MDS | myelodysplastic syndrome |
MM | multiple myeloma |
MsCI4 | CRBN homologue from magnetospirillum gryphiswaldense |
PROTAC | proteolysis targeting chimera |
SALL4 | sal-like protein 4 |
VHL | von-Hippel-Lindau |
References
This article references 60 other publications.
- 1Somers, G. F. Pharmacological properties of thalidomide (alpha-phthalimido glutarimide), a new sedative hypnotic drug. Br. J. Pharmacol. Chemother. 1960, 15, 111– 116, DOI: 10.1111/j.1476-5381.1960.tb01217.xGoogle Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaF3cXht1ShtLc%253D&md5=4d82cdf61a96c7f74e79fb343f40bafdPharmacological properties of thalidomide (α-phthalimidoglutarimide), a new sedative hypnotic drugSomers, G. F.British Journal of Pharmacology and Chemotherapy (1960), 15 (), 111-16CODEN: BJPCAL; ISSN:0366-0826.This new non-toxic non-barbiturate sedative was assayed on male albino rats, mice, rabbits, cats, and guinea pigs for toxicity, effect on motor activity, reflexes, narcosis, analgesis, antipyresis, autonomic system, and gastrointestinal tract. It was virtually nontoxic, and had no effect on these systems, possibly because of limited absorption. It potentiated barbiturates, alc., reserpine, chlorpromazine, and counteracted methylamphetamine and methylphenidate.
- 2Lenz, W. A short history of thalidomide embryopathy. Teratology 1988, 38, 203– 215, DOI: 10.1002/tera.1420380303Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaL1M7ks1Kluw%253D%253D&md5=65dd1712af76e247727c4cf7f528ae51A short history of thalidomide embryopathyLenz WTeratology (1988), 38 (3), 203-15 ISSN:0040-3709.There is no expanded citation for this reference.
- 3Vargesson, N. Thalidomide-induced limb defects: resolving a 50-year-old puzzle. Bioessays 2009, 31, 1327– 1336, DOI: 10.1002/bies.200900103Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXisVamuw%253D%253D&md5=ca4738ec2192397e2b61142a3eda4f3eThalidomide-induced limb defects: resolving a 50-year-old puzzleVargesson, NeilBioEssays (2009), 31 (12), 1327-1336CODEN: BIOEEJ; ISSN:0265-9247. (Wiley-Blackwell)A review. Despite the recent discovery that thalidomide causes limb defects by targeting highly angiogenic, immature blood vessels, several challenges still remain and new ones have arisen. These include understanding the drug's species specificity, detg. mol. target(s) in the endothelial cell, shedding light on the mol. basis of phocomelia and producing a form of the drug that is clin. effective without having side effects. Now that the trigger of thalidomide-induced teratogenesis has been uncovered, a framework is proposed, incorporating and uniting previous models of thalidomide action, explaining how thalidomide causes not just limb defects, but also all the other defects it induces.
- 4Johnston, R. E.; Abdalla, S. H. Thalidomide in low doses is effective for the treatment of resistant or relapsed multiple myeloma and for plasma cell leukaemia. Leuk. Lymphoma 2002, 43, 351– 354, DOI: 10.1080/10428190290006143Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XntVaqs74%253D&md5=fefa82138697984cbc04d8738cfb8bf9Thalidomide in low doses is effective for the treatment of resistant or relapsed multiple myeloma and for plasma cell leukemiaJohnston, R. E.; Abdalla, S. H.Leukemia & Lymphoma (2002), 43 (2), 351-354CODEN: LELYEA; ISSN:1042-8194. (Taylor & Francis Ltd.)Thalidomide is an effective treatment for relapsed multiple myeloma (MM), but is assocd. with a significant side effect profile at higher doses. In a recent study, only half of the enrolled patients were able to tolerate the max. dose of 800 mg/day [Singhal, S., et al. (1999) "Antitumor activity of thalidomide in refractory multiple myeloma", New Engl. J. Med. 341, 1565-1571]. Moreover, the dose-response relationship has not been defined. We report our use of low dose thalidomide in a small cohort of 12 patients-eight with relapsed or refractory MM and four with plasma cell leukemia (PCL). Five of the 12 (42%) patients had a partial response, showing a median fall in their PP/BJP of 80% (63-90%) at a median dose of 175 mg (100-300 mg) with negligible side effects. Three of four patients with PCL showed an impressive response to treatment with thalidomide as a single agent. No patient who failed to show any evidence of response at low dose (<150 mg/day) responded to higher doses. In this study, thalidomide induces a similar rate of response at a lower and better tolerated dose than previously reported and produced "best ever" responses in patients with resistant PCL.
- 5US Thalomid Label. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/020785s061lbl.pdf (accessed Dec 9, 2018).Google ScholarThere is no corresponding record for this reference.
- 6Bertolini, F.; Mingrone, W.; Alietti, A.; Ferrucci, P. F.; Cocorocchio, E.; Peccatori, F.; Cineri, S.; Mancuso, P.; Corsini, C.; Burlini, A.; Zucca, E.; Martinelli, G. Thalidomide in multiple myeloma, myelodysplastic syndromes and histiocytosis. Analysis of clinical results and of surrogate angiogenesis markers. Ann. Oncol. 2001, 12, 987– 990, DOI: 10.1023/a:1011141009812Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD3MvnvVehuw%253D%253D&md5=19268541729e2a5d8762802d62ae882cThalidomide in multiple myeloma, myelodysplastic syndromes and histiocytosis. Analysis of clinical results and of surrogate angiogenesis markersBertolini F; Mingrone W; Alietti A; Ferrucci P F; Cocorocchio E; Peccatori F; Cinieri S; Mancuso P; Corsini C; Burlini A; Zucca E; Martinelli G; Cineri SAnnals of oncology : official journal of the European Society for Medical Oncology (2001), 12 (7), 987-90 ISSN:0923-7534.BACKGROUND: Thalidomide, as a single agent, has been recently found to induce a clinical response in one third of refractory or relapsed myeloma patients. Although it has been reported that thalidomide significantly inhibits angiogenesis. it is still unclear whether its clinical effect is mediated, at least in part, by its anti-angiogenic properties. PATIENTS AND METHODS: We evaluated thalidomide as a single agent in myeloma, myelodysplastic syndromes (MDS) and histiocytosis, i.e. hematological diseases characterized by increased angiogenesis, and measured prospectively a number of surrogate angiogenesis markers. RESULTS: Clinical responses were observed in 7 of 17 myeloma and 2 of 5 MDS patients. The histiocytosis patient had a partial response. At the time of the best clinical response, plasma levels of angiogenic growth factors, vascular endothelial growth factor (VEGF) and basic-fibroblast growth factor (b-FGF), were significantly decreased, and flow cytometry indicated a decrease of activated endothelial cells in the bone marrow of responding MDS patients. CONCLUSIONS: These observations confirm thalidomide efficacy in myeloma, suggest a possible use in MDS and histiocytosis and may contribute to the prediction of clinical response and to understanding the mechanism of thalidomide's action.
- 7Kale, V.; List, A. Immunomodulatory drugs (IMiDs): a new treatment option for myelodysplastic syndromes. Curr. Pharm. Biotechnol. 2006, 7, 339– 342, DOI: 10.2174/138920106778521587Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XhtVamsLrN&md5=e1597e6b7cf4543228046753231f2348Immunomodulatory drugs (IMiDs): a new treatment option for myelodysplastic syndromesKale, Vishakha; List, Alan F.Current Pharmaceutical Biotechnology (2006), 7 (5), 339-342CODEN: CPBUBP; ISSN:1389-2010. (Bentham Science Publishers Ltd.)A review. The IMiDs represent a new proprietary class of thalidomide analogs that possess greater potency and less toxicity than the parent compd. As a group, these agents share the pharmacol. property of modulating cellular response to ligand activation, the precise biol. effect of which is cell lineage and stimulant-dependent. Lenalidomide (CC-5013; Revlimid), a second generation IMiD, has shown significant erythropoietic activity in patients with lower risk MDS that have failed or are not candidates for recombinant erythropoietin treatment. Unlike cytokine therapy, lenalidomide suppresses select MDS clones and enhances erythropoietin receptor signaling to restore erythropoiesis. Activity is greatest in patients with interstitial deletions involving chromosome 5q31.1. A multicenter phase II study reported a 76% overall transfusion response rate in transfusion-dependent patients with deletion 5q, with 67% achieving transfusion independence after a median interval of 4.6 wk of treatment. Cytogenetic responses were obsd. in 73% of patients with complete cytogenetic remission in 45% patients. Both transfusion response and cytogenetic response frequency were independent of karyotype complexity, raising excitement that this new treatment strategy might favorably alter the natural history of disease in higher risk patients with deletion 5q. Lenalidomide was approved by the U.S. Food and Drug Administration on Dec. 27, 2005, for the treatment of IPSS Low and intermediate-1 risk MDS patients with del(5q) abnormality. A phase III Intergroup trial (ECOG 2905) will test the capacity to potentiate erythropoietin response by comparing response to lenalidomide monotherapy to the combination of darbepoetin and lenalidomide in non-deletion 5q MDS patients.
- 8Wu, H.; Zhao, C.; Gu, K.; Jiao, Y.; Hao, J.; Sun, G. Thalidomide plus chemotherapy exhibit enhanced efficacy in the clinical treatment of T-cell non-Hodgkin’s lymphoma: A prospective study of 46 cases. Mol. Clin. Oncol. 2014, 2, 695– 700, DOI: 10.3892/mco.2014.307Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2sbpvFKgtA%253D%253D&md5=f173c9f716db0820c32424792125ff88Thalidomide plus chemotherapy exhibit enhanced efficacy in the clinical treatment of T-cell non-Hodgkin's lymphoma: A prospective study of 46 casesWu Hongyang; Zhao Chenchen; Gu Kangsheng; Jiao Yang; Hao Jiqing; Sun GuopingMolecular and clinical oncology (2014), 2 (5), 695-700 ISSN:2049-9450.The treatment of T-cell non-Hodgkin's lymphoma (T-NHL) remains challenging. There is currently no standard regimen for the treatment of T-NHL in the first- or second-line setting. Thalidomide was previously shown to exert antitumor effects through inhibiting angiogenesis, promoting apoptosis and immunomodulatory activity. However, all the previous studies on the treatment of lymphoma with thalidomide included patient samples of limited size. In the present study, 46 cases of eligible T-NHL patients were randomized into i) the control group (conventional combined chemotherapy, n=22) and ii) the thalidomide group (thalidomide plus combined chemotherapy, n=24). The median dose of thalidomide was 200 mg (range, 150-400 mg) every night, without reported severe side effects. The clinical response to treatment was as follows: Complete response (CR) in 12 cases, partial response (PR) in 7, stable disease (SD) in 1 and progressive disease (PD) in 4 cases in the thalidomide group; and CR in 8 cases, PR in 6, SD in 3 and PD in 5 cases in the control group. The CR rate was 50.0 and 36.4% in the thalidomide and the control groups, respectively (P<0.05). The median progression-free and overall survival were 12 and undefined months, respectively, in the thalidomide group and 6 and 17 months, respectively, in the control group. The toxicity profile was considered acceptable in both groups. Our results indicated that thalidomide plus combined chemotherapy may exhibit enhanced efficacy in the clinical treatment of T-NHL. In addition, this type of treatment may reduce the frequency of adverse gastrointestinal reactions and help alleviate fear of chemotherapy. Therefore, thalidomide plus combined chemotherapy may be a viable option for the clinical treatment of T-NHL.
- 9García-Sanz, R.; González-López, T. J.; Vázquez, L.; Hermida, G.; Graciani, I. F.; San Miguel, J. F. The combination of thalidomide, cyclophosphamide and dexamethasone is potentially useful in highly resistant Hodgkin’s lymphoma. Eur. J. Haematol. 2010, 84, 266– 270, DOI: 10.1111/j.1600-0609.2009.01375.xGoogle Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXjtlGmtL0%253D&md5=11de756fa308a935acb76c8870c33110The combination of thalidomide, cyclophosphamide and dexamethasone is potentially useful in highly resistant Hodgkin's lymphomaGarcia-Sanz, R.; Gonzalez-Lopez, T. J.; Vazquez, L.; Hermida, G.; Graciani, I. F.; San Miguel, J. F.European Journal of Haematology (2010), 84 (3), 266-270CODEN: EJHAEC; ISSN:0902-4441. (Wiley-Blackwell)Few diseases have a prognosis worse than Hodgkin's lymphoma (HL), patients relapsing after autologous or allogeneic stem cell transplantation. Here, we report two highly refractory patients with HL who successfully responded to a combination of thalidomide, cyclophosphamide and dexamethasone (ThaCyDex). Despite the use of a very large no. of different drugs (>5 different schemes) including high-dose therapy and autologous and allogeneic stem cell transplantation, both patients proved to be suffering from a highly resistant disease. Fortunately, they finally responded to the ThaCyDex combination, achieving sustained complete remission that would support the running of a trial within this setting.
- 10Kuruvilla, J.; Song, K.; Mollee, P.; Panzarella, T.; McCrae, J.; Nagy, T.; Crump, M.; Keating, A. A phase II study of thalidomide and vinblastine for palliative patients with Hodgkin’s lymphoma. Hematology 2006, 11, 25– 29, DOI: 10.1080/10245330500276592Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XitVOhu74%253D&md5=7e23a9439d19a069125052db1c1c64b7A phase II study of thalidomide and vinblastine for palliative patients with Hodgkin's lymphomaKuruvilla, John; Song, Kevin; Mollee, Peter; Panzarella, Tony; McCrae, Jan; Nagy, Tracy; Crump, Michael; Keating, ArmandHematology (Abingdon, United Kingdom) (2006), 11 (1), 25-29CODEN: HMATFL; ISSN:1024-5332. (Taylor & Francis Ltd.)Introduction: Patients with Hodgkin's Lymphoma (HL) who relapse or progress after primary therapy and subsequent high dose chemotherapy with autologous stem cell transplantation (ASCT) cannot be cured with conventional treatment. We combined thalidomide (THAL), an agent with anti-angiogenic and immunomodulatory properties, with vinblastine, which is active after ASCT, to det. the objective response rate, improvement in B symptoms and toxicity in patients with refractory HD. Methods: Patients were eligible if they HD that progressed after chemotherapy and ASCT or had declined or were ineligible for curative therapy. Treatment consisted of THAL 200 mg orally given daily. After 2 wk, VBL 6 mg IV was given weekly × 6 doses on an eight-week cycle. Response and toxicity assessment occurred following each cycle. Results: Eleven patients were enrolled, 1 progressed within 6 days of study enrollment and was subsequently treated with alternative palliative therapy and thus 11 patients are response evaluable and 10 are evaluable for toxicity. Patient characteristics: relapsed after ASCT: 7; median no. of prior chemotherapy regimens: 3 (range 1-5); median time to progression post-ASCT: 7 mo (range 2-29). Four patients had a partial response to treatment (response rate 36%); two patients had stable disease. B symptoms were present at enrollment in four patients and resolved completely on treatment in two patients. Five had disease progression within 3 mo of starting treatment. The median duration of response was 9 mo (range 0-22 mo). Toxicity was mild and limited to grade 2 neuropathy in 6 patients and grade 2 or 3 neutropenia in 4 patients. Conclusions: In this small study in chemotherapy- refractory HL, THAL and VBL demonstrated encouraging activity with some durable responses and acceptable toxicity. These results suggest that chronic low dose chemotherapy combined with less toxic immunomodulatory or anti-angiogenic drugs warrants further study.
- 11Seldin, D. C.; Choufani, E. B.; Dember, L. M.; Wiesman, J. F.; Berk, J. L.; Falk, R. H.; O’Hara, C.; Fennessey, S.; Finn, K. T.; Wright, D. G.; Skinner, M.; Sanchorawala, V. Tolerability and efficacy of thalidomide for the treatment of patients with light chain-associated (AL) amyloidosis. Clin. Lymphoma 2003, 3, 241– 246, DOI: 10.3816/clm.2003.n.005Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXjtVSjtrk%253D&md5=b15425f1d8d306ab1f9bb1375a49e40bTolerability and efficacy of thalidomide for the treatment of patients with light-chain-associated (AL) amyloidosisSeldin, David C.; Choufani, Elie B.; Dember, Laura M.; Wiesman, Janice F.; Berk, John L.; Falk, Rodney H.; O'Hara, Carl; Fennessey, Salli; Finn, Kathleen T.; Wright, Daniel G.; Skinner, Martha; Sanchorawala, VaishaliClinical Lymphoma (2003), 3 (4), 241-246CODEN: CLLYAO; ISSN:1526-9655. (Cancer Information Group)A phase I/II trial was carried out with patients with AL amyloidosis, most of whom had failed prior therapy with high-dose melphalan and autologous stem cell transplantation. This trial was designed as an individualized 6-mo dose-escalation study with re-evaluation of bone marrow plasmacytosis and serum and urine monoclonal proteins after 3 and 6 mo. Sixteen patients with a median age of 62 yr (range, 37-70 yr) were enrolled. Fourteen of the patients had renal involvement, 4 had cardiac involvement, 4 had liver involvement, and 2 had predominant soft tissue or lymph node involvement. The median max. tolerated dose was 300 mg, with fatigue and other central nervous system side effects being the major dose-limiting toxicities. Side effects not frequently reported for other patient populations included exacerbation of peripheral and pulmonary edema and worsening azotemia. In all, 50% of the patients experienced grade 3/4 toxicity, and 25% had to discontinue the drug. No complete hematol. responses were seen, but 25% of the patients had a significant redn. in Bence-Jones proteinuria. Thus, while thalidomide has activity in AL amyloidosis, it also has significant toxicity in this patient population.
- 12Baird, R.; van Zyl-Smit, R. N.; Iveson, A.; Duddy, J.; Rassam, S. M. Thalidomide is highly effective in a patient with meningeal acute myeloid leukaemia. Leuk. Lymphoma 2004, 45, 179– 181, DOI: 10.1080/1042819031000149412Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD2c7mslehtw%253D%253D&md5=31006e0dbe0fb3cf55bcd7198fc8ef2aThalidomide is highly effective in a patient with meningeal acute myeloid leukaemiaBaird Richard; van Zyl-Smit Richard Nellis; Iveson Anne; Duddy James; Rassam Saad M BLeukemia & lymphoma (2004), 45 (1), 179-81 ISSN:1042-8194.We report a case of secondary acute myeloid leukaemia (AML) following high dose therapy for diffuse large B-cell non-Hodgkin's lymphoma (NHL) who developed meningeal leukaemia. This was refractory to systemic and intrathecal chemotherapy and cranial irradiation. Thalidomide has been reported to have anti-AML activity and appears to cross the blood brain barrier (BBB). We, therefore, attempted a trial of oral Thalidomide and achieved rapid biochemical and cytological remission with a short course. The patient, however, progressed systemically and succumbed to her illness.
- 13Thomas, D. A.; Estey, E.; Giles, F. J.; Faderl, S.; Cortes, J.; Keating, M.; O’Brien, S.; Albitar, M.; Kantarjian, H. Single agent thalidomide in patients with relapsed or refractory acute myeloid leukaemia. Br. J. Haematol. 2003, 123, 436– 441, DOI: 10.1046/j.1365-2141.2003.04639.xGoogle Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXpsFChs70%253D&md5=28de5d4f924d2b35a000096ceb1f5be7Single agent thalidomide in patients with relapsed or refractory acute myeloid leukaemiaThomas, Deborah A.; Estey, Elihu; Giles, Francis J.; Faderi, Stefan; Cortes, Jorge; Keating, Michael; O'Brien, Susan; Albitar, Maher; Kantarjian, HagopBritish Journal of Haematology (2003), 123 (3), 436-441CODEN: BJHEAL; ISSN:0007-1048. (Blackwell Publishing Ltd.)Thalidomide is a putative anti-angiogenesis agent that has significant anti-tumor activity in haematol. malignancies with increased bone marrow angiogenesis, including multiple myeloma (MM) and myelodysplastic syndromes (MDS). Increased levels of the mitogen for angiogenesis, vascular endothelial growth factor (VEGF), correlate with worse survival in acute myeloid leukemia (AML). A phase II trial of thalidomide was conducted in patients with relapsed- or refractory-AML previously treated with cytarabine-contg. regimens. A total of 16 patients with refractory- or relapsed-AML were treated with thalidomide 200-800 mg orally daily (median dose 400 mg daily) for a median of 27 d (range, 3-94 d). Overall, one patient (6%) achieved complete remission (CR) lasting for 36 mo, and two patients had a transient redn. in marrow blasts from 8% and 7% to less than 5% in both cases. There was no correlation between redn. in levels of angiogenesis markers and response. Toxicities related to thalidomide were significant, and precluded dose escalation beyond 400 mg orally daily in most patients. Although there appears to be some evidence of biol. activity, single agent thalidomide is not an optimal choice of therapy for salvaging patients with relapsed- or refractory-AML. Thalidomide analogs with more potent immunomodulatory activities and more favorable toxicity profiles may offer more promise as anti-AML therapy.
- 14Hartmann, M. D.; Boichenko, I.; Coles, M.; Zanini, F.; Lupas, A. N.; Hernandez Alvarez, B. Thalidomide mimics uridine binding to an aromatic cage in cereblon. J. Struct. Biol. 2014, 188, 225– 232, DOI: 10.1016/j.jsb.2014.10.010Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhvFSnu7jI&md5=e601c6e1091deea840f9738fb30fa7bbThalidomide mimics uridine binding to an aromatic cage in cereblonHartmann, Marcus D.; Boichenko, Iuliia; Coles, Murray; Zanini, Fabio; Lupas, Andrei N.; Hernandez Alvarez, BirteJournal of Structural Biology (2014), 188 (3), 225-232CODEN: JSBIEM; ISSN:1047-8477. (Elsevier Inc.)Thalidomide and its derivs. lenalidomide and pomalidomide are important anticancer agents but can cause severe birth defects via an interaction with the protein cereblon. The ligand-binding domain of cereblon is found, with a high degree of conservation, in both bacteria and eukaryotes. Using a bacterial model system, we reveal the structural determinants of cereblon substrate recognition, based on a series of high-resoln. crystal structures. For the first time, we identify a cellular ligand that is universally present: we show that thalidomide and its derivs. mimic and compete for the binding of uridine, and validate these findings in vivo. The nature of the binding pocket, an arom. cage of three tryptophan residues, further suggests a role in the recognition of cationic ligands. Our results allow for general evaluation of pharmaceuticals for potential cereblon-dependent teratogenicity.
- 15Hartmann, M. D.; Boichenko, I.; Coles, M.; Lupas, A. N.; Hernandez Alvarez, B. Structural dynamics of the cereblon ligand binding domain. PLoS One 2015, 10, e0128342 DOI: 10.1371/journal.pone.0128342Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhslGru77F&md5=525f48f454c54c466360231d1ab3330aStructural dynamics of the cereblon ligand binding domainHartmann, Marcus D.; Boichenko, Iuliia; Coles, Murray; Lupas, Andrei N.; Alvarez, Birte HernandezPLoS One (2015), 10 (5), e0128342/1-e0128342/16CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)Cereblon, a primary target of thalidomide and its derivs., has been characterized structurally from both bacteria and animals. Esp. well studied is the thalidomide binding domain, CULT, which shows an invariable structure across different organisms and in complex with different ligands. Here, based on a series of crystal structures of a bacterial representative, we reveal the conformational flexibility and structural dynamics of this domain. In particular, we follow the unfolding of large fractions of the domain upon release of thalidomide in the cryst. state. Our results imply that a third of the domain, including the thalidomide binding pocket, only folds upon ligand binding. We further characterize the structural effect of the C-terminal truncation resulting from the mental-retardation linked R419X nonsense mutation in vitro and offer a mechanistic hypothesis for its irresponsiveness to thalidomide. At 1.2Å resoln., our data provide a view of thalidomide binding at at. resoln.
- 16Chamberlain, P. P.; Lopez-Girona, A.; Miller, K.; Carmel, G.; Pagarigan, B.; Chie-Leon, B.; Rychak, E.; Corral, L. G.; Ren, Y. J.; Wang, M.; Riley, M.; Delker, S. L.; Ito, T.; Ando, H.; Mori, T.; Hirano, Y.; Handa, H.; Hakoshima, T.; Daniel, T. O.; Cathers, B. E. Structure of the human cereblon-DDB1-lenalidomide complex reveals basis for responsiveness to thalidomide analogs. Nat. Struct. Mol. Biol. 2014, 21, 803– 809, DOI: 10.1038/nsmb.2874Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtlahurjI&md5=627fc3c8eae320059ca1cdc173bb56edStructure of the human Cereblon-DDB1-lenalidomide complex reveals basis for responsiveness to thalidomide analogsChamberlain, Philip P.; Lopez-Girona, Antonia; Miller, Karen; Carmel, Gilles; Pagarigan, Barbra; Chie-Leon, Barbara; Rychak, Emily; Corral, Laura G.; Ren, Yan J.; Wang, Maria; Riley, Mariko; Delker, Silvia L.; Ito, Takumi; Ando, Hideki; Mori, Tomoyuki; Hirano, Yoshinori; Handa, Hiroshi; Hakoshima, Toshio; Daniel, Thomas O.; Cathers, Brian E.Nature Structural & Molecular Biology (2014), 21 (9), 803-809CODEN: NSMBCU; ISSN:1545-9993. (Nature Publishing Group)The Cul4-Rbx1-DDB1-Cereblon E3 ubiquitin ligase complex is the target of thalidomide, lenalidomide and pomalidomide, therapeutically important drugs for multiple myeloma and other B-cell malignancies. These drugs directly bind Cereblon (CRBN) and promote the recruitment of substrates Ikaros (IKZF1) and Aiolos (IKZF3) to the E3 complex, thus leading to substrate ubiquitination and degrdn. Here we present the crystal structure of human CRBN bound to DDB1 and the drug lenalidomide. A hydrophobic pocket in the thalidomide-binding domain (TBD) of CRBN accommodates the glutarimide moiety of lenalidomide, whereas the isoindolinone ring is exposed to solvent. We also solved the structures of the mouse TBD in the apo state and with thalidomide or pomalidomide. Site-directed mutagenesis in lentiviral-expression myeloma models showed that key drug-binding residues are crit. for antiproliferative effects.
- 17Fischer, E. S.; Böhm, K.; Lydeard, J. R.; Yang, H.; Stadler, M. B.; Cavadini, S.; Nagel, J.; Serluca, F.; Acker, V.; Lingaraju, G. M.; Tichkule, R. B.; Schebesta, M.; Forrester, W. C.; Schirle, M.; Hassiepen, U.; Ottl, J.; Hild, M.; Beckwith, R. E. J.; Harper, J. W.; Jenkins, J. L.; Thomä, N. H. Structure of the DDB1-CRBN E3 ubiquitin ligase in complex with thalidomide. Nature 2014, 512, 49– 53, DOI: 10.1038/nature13527Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXht12ms7%252FP&md5=1c1eaa48bef87463cdd3f2b1e1bd400dStructure of the DDB1-CRBN E3 ubiquitin ligase in complex with thalidomideFischer, Eric S.; Bohm, Kerstin; Lydeard, John R.; Yang, Haidi; Stadler, Michael B.; Cavadini, Simone; Nagel, Jane; Serluca, Fabrizio; Acker, Vincent; Lingaraju, Gondichatnahalli M.; Tichkule, Ritesh B.; Schebesta, Michael; Forrester, William C.; Schirle, Markus; Hassiepen, Ulrich; Ottl, Johannes; Hild, Marc; Beckwith, Rohan E. J.; Harper, J. Wade; Jenkins, Jeremy L.; Thoma, Nicolas H.Nature (London, United Kingdom) (2014), 512 (7512), 49-53CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)In the 1950s, the drug thalidomide, administered as a sedative to pregnant women, led to the birth of thousands of children with multiple defects. Despite the teratogenicity of thalidomide and its derivs. lenalidomide and pomalidomide, these immunomodulatory drugs (IMiDs) recently emerged as effective treatments for multiple myeloma and 5q-deletion-assocd. dysplasia. IMiDs target the E3 ubiquitin ligase CUL4-RBX1-DDB1-CRBN (known as CRL4CRBN) and promote the ubiquitination of the IKAROS family transcription factors IKZF1 and IKZF3 by CRL4CRBN. Here we present crystal structures of the DDB1-CRBN complex bound to thalidomide, lenalidomide and pomalidomide. The structure establishes that CRBN is a substrate receptor within CRL4CRBN and enantioselectively binds IMiDs. Using an unbiased screen, we identified the homeobox transcription factor MEIS2 as an endogenous substrate of CRL4CRBN. Our studies suggest that IMiDs block endogenous substrates (MEIS2) from binding to CRL4CRBN while the ligase complex is recruiting IKZF1 or IKZF3 for degrdn. This dual activity implies that small mols. can modulate an E3 ubiquitin ligase and thereby upregulate or downregulate the ubiquitination of proteins.
- 18Nguyen, T. V.; Lee, J. E.; Sweredoski, M. J.; Yang, S. J.; Jeon, S. J.; Harrison, J. S.; Yim, J. H.; Lee, S. G.; Handa, H.; Kuhlman, B.; Jeong, J. S.; Reitsma, J. M.; Park, C. S.; Hess, S.; Deshaies, R. J. Glutamine triggers acetylation-dependent degradation of glutamine synthetase via the thalidomide receptor cereblon. Mol. Cell 2016, 61, 809– 820, DOI: 10.1016/j.molcel.2016.02.032Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xks1OqsL0%253D&md5=641527ec1a6c97d1800e3e393582c07fGlutamine Triggers Acetylation-Dependent Degradation of Glutamine Synthetase via the Thalidomide Receptor CereblonNguyen, T. Van; Lee, J. Eugene; Sweredoski, Michael J.; Yang, Seung-Joo; Jeon, Seung-Je; Harrison, Joseph S.; Yim, Jung-Hyuk; Lee, Sang Ghil; Handa, Hiroshi; Kuhlman, Brian; Jeong, Ji-Seon; Reitsma, Justin M.; Park, Chul-Seung; Hess, Sonja; Deshaies, Raymond J.Molecular Cell (2016), 61 (6), 809-820CODEN: MOCEFL; ISSN:1097-2765. (Elsevier Inc.)Cereblon (CRBN), a substrate receptor for the cullin-RING ubiquitin ligase 4 (CRL4) complex, is a direct protein target for thalidomide teratogenicity and antitumor activity of immunomodulatory drugs (IMiDs). Here we report that glutamine synthetase (GS) is an endogenous substrate of CRL4CRBN. Upon exposing cells to high glutamine concn., GS is acetylated at lysines 11 and 14, yielding a degron that is necessary and sufficient for binding and ubiquitylation by CRL4CRBN and degrdn. by the proteasome. Binding of acetylated degron peptides to CRBN depends on an intact thalidomide-binding pocket but is not competitive with IMiDs. These findings reveal a feedback loop involving CRL4CRBN that adjusts GS protein levels in response to glutamine and uncover a new function for lysine acetylation.
- 19Del Prete, D.; Rice, R. C.; Rajadhyaksha, A. M.; D’Adamio, L. Amyloid precursor protein (APP) may act as a substrate and a recognition unit for CRL4(CRBN) and Stub1 E3 ligases facilitating ubiquitination of proteins involved in presynaptic functions and neurodegeneration. J. Biol. Chem. 2016, 291, 17209– 17227, DOI: 10.1074/jbc.m116.733626Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhslaktLrF&md5=b4cfc53ded92c62aa868a17e28929986Amyloid precursor protein (APP) may act as a substrate and a recognition unit for CRL4CRBN and Stub1 E3 ligases facilitating ubiquitination of proteins involved in presynaptic functions and neurodegenerationDel Prete, Dolores; Rice, Richard C.; Rajadhyaksha, Anjali M.; D'Adamio, LucianoJournal of Biological Chemistry (2016), 291 (33), 17209-17227CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)The amyloid precursor protein (APP), whose mutations cause Alzheimer disease, plays an important in vivo role and facilitates transmitter release. Because the APP cytosolic region (ACR) is essential for these functions, we have characterized its brain interactome. We found that the ACR interacts with proteins that regulate the ubiquitin-proteasome system, predominantly with the E3 ubiquitin-protein ligases Stub1, which binds the N-terminus of the ACR, and CRL4CRBN, which is formed by Cul4a/b, Ddb1, and Crbn, and interacts with the C-terminus of the ACR via Crbn. APP shares essential functions with APP-like protein-2 (APLP2) but not APP-like protein-1 (APLP1). Noteworthy, APLP2, but not APLP1, interacts with Stub1 and CRL4CRBN, pointing to a functional pathway shared only by APP and APLP2. In vitro ubiquitination/ubiquitome anal. indicates that these E3 ligases are enzymically active and ubiquitinated the ACR residues Lys649/650/651/676/688. Deletion of Crbn reduces ubiquitination of Lys676 suggesting that Lys676 is physiol. ubiquitinated by CRL4CRBN. The ACR facilitated in vitro ubiquitination of presynaptic proteins that regulate exocytosis, suggesting a mechanism by which APP tunes transmitter release. Other dementia-related proteins, namely Tau and apoE, interact with and are ubiquitinated via the ACR in vitro. This, and the evidence that CRBN and CUL4B are linked to intellectual disability, prompts us to hypothesize a pathogenic mechanism, in which APP acts as a modulator of E3 ubiquitin-protein ligase(s), shared by distinct neuronal disorders. The well described accumulation of ubiquitinated protein inclusions in neurodegenerative diseases and the link between the ubiquitin-proteasome system and neurodegeneration make this concept plausible.
- 20Kronke, J.; Udeshi, N. D.; Narla, A.; Grauman, P.; Hurst, S. N.; McConkey, M.; Svinkina, T.; Heckl, D.; Comer, E.; Li, X.; Ciarlo, C.; Hartman, E.; Munshi, N.; Schenone, M.; Schreiber, S. L.; Carr, S. A.; Ebert, B. L. Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells. Science 2014, 343, 301– 305, DOI: 10.1126/science.1244851Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2c3itFajsQ%253D%253D&md5=8cae3e554399308493aa38f0d05ed610Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cellsKronke Jan; Udeshi Namrata D; Narla Anupama; Grauman Peter; Hurst Slater N; McConkey Marie; Svinkina Tanya; Heckl Dirk; Comer Eamon; Li Xiaoyu; Ciarlo Christie; Hartman Emily; Munshi Nikhil; Schenone Monica; Schreiber Stuart L; Carr Steven A; Ebert Benjamin LScience (New York, N.Y.) (2014), 343 (6168), 301-5 ISSN:.Lenalidomide is a drug with clinical efficacy in multiple myeloma and other B cell neoplasms, but its mechanism of action is unknown. Using quantitative proteomics, we found that lenalidomide causes selective ubiquitination and degradation of two lymphoid transcription factors, IKZF1 and IKZF3, by the CRBN-CRL4 ubiquitin ligase. IKZF1 and IKZF3 are essential transcription factors in multiple myeloma. A single amino acid substitution of IKZF3 conferred resistance to lenalidomide-induced degradation and rescued lenalidomide-induced inhibition of cell growth. Similarly, we found that lenalidomide-induced interleukin-2 production in T cells is due to depletion of IKZF1 and IKZF3. These findings reveal a previously unknown mechanism of action for a therapeutic agent: alteration of the activity of an E3 ubiquitin ligase, leading to selective degradation of specific targets.
- 21Krönke, J.; Fink, E. C.; Hollenbach, P. W.; MacBeth, K. J.; Hurst, S. N.; Udeshi, N. D.; Chamberlain, P. P.; Mani, D. R.; Man, H. W.; Gandhi, A. K.; Svinkina, T.; Schneider, R. K.; McConkey, M.; Järås, M.; Griffiths, E.; Wetzler, M.; Bullinger, L.; Cathers, B. E.; Carr, S. A.; Chopra, R.; Ebert, B. L. Lenalidomide induces ubiquitination and degradation of CK1alpha in del(5q) MDS. Nature 2015, 523, 183– 188, DOI: 10.1038/nature14610Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXht1entr%252FN&md5=9b20a91c13e888e6569223ffa25af2cdLenalidomide induces ubiquitination and degradation of CK1α in del(5q) MDSKronke, Jan; Fink, Emma C.; Hollenbach, Paul W.; MacBeth, Kyle J.; Hurst, Slater N.; Udeshi, Namrata D.; Chamberlain, Philip P.; Mani, D. R.; Man, Hon Wah; Gandhi, Anita K.; Svinkina, Tanya; Schneider, Rebekka K.; McConkey, Marie; Jaras, Marcus; Griffiths, Elizabeth; Wetzler, Meir; Bullinger, Lars; Cathers, Brian E.; Carr, Steven A.; Chopra, Rajesh; Ebert, Benjamin L.Nature (London, United Kingdom) (2015), 523 (7559), 183-188CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Lenalidomide is a highly effective treatment for myelodysplastic syndrome (MDS) with deletion of chromosome 5q (del(5q)). Lenalidomide induces the ubiquitination of casein kinase 1A1 (CK1α) by the E3 ubiquitin ligase CUL4-RBX1-DDB1-CRBN (known as CRL4CRBN), resulting in CK1α degrdn. CK1α is encoded by a gene within the common deleted region for del(5q) MDS and haplo-insufficient expression sensitizes cells to lenalidomide therapy, providing a mechanistic basis for the therapeutic window of lenalidomide in del(5q) MDS. The authors found that mouse cells are resistant to lenalidomide but that changing a single amino acid in mouse Crbn to the corresponding human residue enables lenalidomide-dependent degrdn. of CK1α. The authors further demonstrate that minor side chain modifications in thalidomide and a novel analog, CC-122, can modulate the spectrum of substrates targeted by CRL4CRBN. These findings have implications for the clin. activity of lenalidomide and related compds., and demonstrate the therapeutic potential of novel modulators of E3 ubiquitin ligases.
- 22Matyskiela, M. E.; Lu, G.; Ito, T.; Pagarigan, B.; Lu, C.-C.; Miller, K.; Fang, W.; Wang, N.-Y.; Nguyen, D.; Houston, J.; Carmel, G.; Tran, T.; Riley, M.; Nosaka, L. A.; Lander, G. C.; Gaidarova, S.; Xu, S.; Ruchelman, A. L.; Handa, H.; Carmichael, J.; Daniel, T. O.; Cathers, B. E.; Lopez-Girona, A.; Chamberlain, P. P. A novel cereblon modulator recruits GSPT1 to the CRL4(CRBN) ubiquitin ligase. Nature 2016, 535, 252– 257, DOI: 10.1038/nature18611Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtVCltbnI&md5=1d4e340416aa0b08fc0f123faf638c3fA novel cereblon modulator recruits GSPT1 to the CRL4CRBN ubiquitin ligaseMatyskiela, Mary E.; Lu, Gang; Ito, Takumi; Pagarigan, Barbra; Lu, Chin-Chun; Miller, Karen; Fang, Wei; Wang, Nai-Yu; Nguyen, Derek; Houston, Jack; Carmel, Gilles; Tran, Tam; Riley, Mariko; Nosaka, Lyn'Al; Lander, Gabriel C.; Gaidarova, Svetlana; Xu, Shuichan; Ruchelman, Alexander L.; Handa, Hiroshi; Carmichael, James; Daniel, Thomas O.; Cathers, Brian E.; Lopez-Girona, Antonia; Chamberlain, Philip P.Nature (London, United Kingdom) (2016), 535 (7611), 252-257CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Immunomodulatory drugs bind to cereblon (CRBN) to confer differentiated substrate specificity on the CRL4CRBN E3 ubiquitin ligase. Here we report the identification of a new cereblon modulator, CC-885, with potent anti-tumor activity. The anti-tumor activity of CC-885 is mediated through the cereblon-dependent ubiquitination and degrdn. of the translation termination factor GSPT1. Patient-derived acute myeloid leukemia tumor cells exhibit high sensitivity to CC-885, indicating the clin. potential of this mechanism. Crystallog. studies of the CRBN-DDB1-CC-885-GSPT1 complex reveal that GSPT1 binds to cereblon through a surface turn contg. a glycine residue at a key position, interacting with both CC-885 and a 'hotspot' on the cereblon surface. Although GSPT1 possesses no obvious structural, sequence or functional homol. to previously known cereblon substrates, mutational anal. and modeling indicate that the cereblon substrate Ikaros uses a similar structural feature to bind cereblon, suggesting a common motif for substrate recruitment. These findings define a structural degron underlying cereblon 'neosubstrate' selectivity, and identify an anti-tumor target rendered druggable by cereblon modulation.
- 23Matyskiela, M. E.; Couto, S.; Zheng, X.; Lu, G.; Hui, J.; Stamp, K.; Drew, C.; Ren, Y.; Wang, M.; Carpenter, A.; Lee, C.-W.; Clayton, T.; Fang, W.; Lu, C.-C.; Riley, M.; Abdubek, P.; Blease, K.; Hartke, J.; Kumar, G.; Vessey, R.; Rolfe, M.; Hamann, L. G.; Chamberlain, P. P. SALL4 mediates teratogenicity as a thalidomide-dependent cereblon substrate. Nat. Chem. Biol. 2018, 14, 981– 987, DOI: 10.1038/s41589-018-0129-xGoogle Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhs1Kgu7nM&md5=7530c3850ed7a763b30cba44dab640acSALL4 mediates teratogenicity as a thalidomide-dependent cereblon substrateMatyskiela, Mary E.; Couto, Suzana; Zheng, Xinde; Lu, Gang; Hui, Julia; Stamp, Katie; Drew, Clifton; Ren, Yan; Wang, Maria; Carpenter, Aaron; Lee, Chung-Wein; Clayton, Thomas; Fang, Wei; Lu, Chin-Chun; Riley, Mariko; Abdubek, Polat; Blease, Kate; Hartke, James; Kumar, Gondi; Vessey, Rupert; Rolfe, Mark; Hamann, Lawrence G.; Chamberlain, Philip P.Nature Chemical Biology (2018), 14 (10), 981-987CODEN: NCBABT; ISSN:1552-4450. (Nature Research)Targeted protein degrdn. via small-mol. modulation of cereblon offers vast potential for the development of new therapeutics. Cereblon-binding therapeutics carry the safety risks of thalidomide, which caused an epidemic of severe birth defects characterized by forelimb shortening or phocomelia. Here we show that thalidomide is not teratogenic in transgenic mice expressing human cereblon, indicating that binding to cereblon is not sufficient to cause birth defects. Instead, we identify SALL4 as a thalidomide-dependent cereblon neosubstrate. Human mutations in SALL4 cause Duane-radial ray, IVIC, and acro-renal-ocular syndromes with overlapping clin. presentations to thalidomide embryopathy, including phocomelia. SALL4 is degraded in rabbits but not in resistant organisms such as mice because of SALL4 sequence variations. This work expands the scope of cereblon neosubstrate activity within the formerly 'undruggable' C2H2 zinc finger family and offers a path toward safer therapeutics through an improved understanding of the mol. basis of thalidomide-induced teratogenicity.
- 24Donovan, K. A.; An, J.; Nowak, R. P.; Yuan, J. C.; Fink, E. C.; Berry, B. C.; Ebert, B. L.; Fischer, E. S., Thalidomide promotes degradation of SALL4, a transcription factor implicated in Duane Radial Ray Syndrome. Elife 2018, 7. DOI: 10.7554/elife.38430Google ScholarThere is no corresponding record for this reference.
- 25Petzold, G.; Fischer, E. S.; Thomä, N. H. Structural basis of lenalidomide-induced CK1alpha degradation by the CRL4(CRBN) ubiquitin ligase. Nature 2016, 532, 127– 130, DOI: 10.1038/nature16979Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XjtFOis78%253D&md5=320fc6ff811da335fa8e02feab2d702dStructural basis of lenalidomide-induced CK1α degradation by the CRL4CRBN ubiquitin ligasePetzold, Georg; Fischer, Eric S.; Thoma, Nicolas H.Nature (London, United Kingdom) (2016), 532 (7597), 127-130CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Thalidomide and its derivs., lenalidomide and pomalidomide, are immune modulatory drugs (IMiDs) used in the treatment of hematol. malignancies. IMiDs bind CRBN, the substrate receptor of the CUL4-RBX1-DDB1-CRBN (also known as CRL4CRBN) E3 ubiquitin ligase, and inhibit ubiquitination of endogenous CRL4CRBN substrates. Unexpectedly, IMiDs also repurpose the ligase to target new proteins for degrdn. Lenalidomide induces degrdn. of the lymphoid transcription factors Ikaros and Aiolos (also known as IKZF1 and IKZF3), and casein kinase 1α (CK1α), which contributes to its clin. efficacy in the treatment of multiple myeloma and 5q-deletion assocd. myelodysplastic syndrome (del(5q) MDS), resp. How lenalidomide alters the specificity of the ligase to degrade these proteins remains elusive. Here we present the 2.45 Å crystal structure of DDB1-CRBN bound to lenalidomide and CK1α. CRBN and lenalidomide jointly provide the binding interface for a CK1α β-hairpin-loop located in the kinase N-lobe. We show that CK1α binding to CRL4CRBN is strictly dependent on the presence of an IMiD. Binding of IKZF1 to CRBN similarly requires the compd. and both, IKZF1 and CK1α, use a related binding mode. Our study provides a mechanistic explanation for the selective efficacy of lenalidomide in del(5q) MDS therapy. We anticipate that high-affinity protein-protein interactions induced by small mols. will provide opportunities for drug development, particularly for targeted protein degrdn.
- 26Sievers, Q. L.; Petzold, G.; Bunker, R. D.; Renneville, A.; Słabicki, M.; Liddicoat, B. J.; Abdulrahman, W.; Mikkelsen, T.; Ebert, B. L.; Thomä, N. H. Defining the human C2H2 zinc finger degrome targeted by thalidomide analogs through CRBN. Science 2018, 362, eaat0572 DOI: 10.1126/science.aat0572Google ScholarThere is no corresponding record for this reference.
- 27Lai, A. C.; Crews, C. M. Induced protein degradation: an emerging drug discovery paradigm. Nat. Rev. Drug Discovery 2017, 16, 101– 114, DOI: 10.1038/nrd.2016.211Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhvFCit7rP&md5=a01ff43e9b696fe4dc03f0815158a98fInduced protein degradation: an emerging drug discovery paradigmLai, Ashton C.; Crews, Craig M.Nature Reviews Drug Discovery (2017), 16 (2), 101-114CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)Small-mol. drug discovery has traditionally focused on occupancy of a binding site that directly affects protein function, and this approach typically precludes targeting proteins that lack such amenable sites. Furthermore, high systemic drug exposures may be needed to maintain sufficient target inhibition in vivo, increasing the risk of undesirable off-target effects. Induced protein degrdn. is an alternative approach that is event-driven: upon drug binding, the target protein is tagged for elimination. Emerging technologies based on proteolysis-targeting chimaeras (PROTACs) that exploit cellular quality control machinery to selectively degrade target proteins are attracting considerable attention in the pharmaceutical industry owing to the advantages they could offer over traditional small-mol. strategies. These advantages include the potential to reduce systemic drug exposure, the ability to counteract increased target protein expression that often accompanies inhibition of protein function and the potential ability to target proteins that are not currently therapeutically tractable, such as transcription factors, scaffolding and regulatory proteins.
- 28Sakamoto, K. M.; Kim, K. B.; Kumagai, A.; Mercurio, F.; Crews, C. M.; Deshaies, R. J. Protacs: Chimeric molecules that target proteins to the Skp1–Cullin–F box complex for ubiquitination and degradation. Proc. Natl. Acad. Sci. U.S.A. 2001, 98, 8554– 8559, DOI: 10.1073/pnas.141230798Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXls1Wisbk%253D&md5=540358035222f745f7b6367b38781a21Protacs: chimeric molecules that target proteins to the Skp1-Cullin-F box complex for ubiquitination and degradationSakamoto, Kathleen M.; Kim, Kyung B.; Kumagai, Akiko; Mercurio, Frank; Crews, Craig M.; Deshaies, Raymond J.Proceedings of the National Academy of Sciences of the United States of America (2001), 98 (15), 8554-8559CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The intracellular levels of many proteins are regulated by ubiquitin-dependent proteolysis. One of the best-characterized enzymes that catalyzes the attachment of ubiquitin to proteins is a ubiquitin ligase complex, Skp1-Cullin-F box complex contg. Hrt1 (SCF). We sought to artificially target a protein to the SCF complex for ubiquitination and degrdn. To this end, we tested methionine aminopeptidase-2 (MetAP-2), which covalently binds the angiogenesis inhibitor ovalicin. A chimeric compd., protein-targeting chimeric mol. 1 (Protac-1), was synthesized to recruit MetAP-2 to SCF. One domain of Protac-1 contains the IκBα phosphopeptide that is recognized by the F-box protein β-TRCP, whereas the other domain is composed of ovalicin. We show that MetAP-2 can be tethered to SCFβ-TRCP, ubiquitinated, and degraded in a Protac-1-dependent manner. In the future, this approach may be useful for conditional inactivation of proteins, and for targeting disease-causing proteins for destruction.
- 29Hu, J.; Hu, B.; Wang, M.; Xu, F.; Miao, B.; Yang, C.-Y.; Wang, M.; Liu, Z.; Hayes, D. F.; Chinnaswamy, K.; Delproposto, J.; Stuckey, J.; Wang, S. Discovery of ERD-308 as a Highly Potent Proteolysis Targeting Chimera (PROTAC) Degrader of Estrogen Receptor (ER). J. Med. Chem. 2019, 62, 1420– 1442, DOI: 10.1021/acs.jmedchem.8b01572Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXht1Cqs7s%253D&md5=678e82dd460c065e2054ba18ea8af18aDiscovery of ERD-308 as a highly potent proteolysis targeting chimera (PROTAC) degrader of estrogen receptor (ER)Hu, Jiantao; Hu, Biao; Wang, Mingliang; Xu, Fuming; Miao, Bukeyan; Yang, Chao-Yie; Wang, Mi; Liu, Zhaomin; Hayes, Daniel F.; Chinnaswamy, Krishnapriya; Delproposto, James; Stuckey, Jeanne; Wang, ShaomengJournal of Medicinal Chemistry (2019), 62 (3), 1420-1442CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The estrogen receptor (ER) is a validated target for the treatment of estrogen receptor-pos. (ER+) breast cancer. Here, we describe the design, synthesis, and extensive structure-activity relationship (SAR) studies of small-mol. ERα degraders based on the proteolysis targeting chimeras (PROTAC) concept. Our efforts have resulted in the discovery of highly potent and effective PROTAC ER degraders, as exemplified by ERD-308 I. I achieves DC50 (concn. causing 50% of protein degrdn.) values of 0.17 and 0.43 nM in MCF-7 and T47D ER+ breast cancer cell lines, resp., and induces >95% of ER degrdn. at concns. as low as 5 nM in both cell lines. Significantly, I induces more complete ER degrdn. than fulvestrant, the only approved selective ER degrader (SERD), and is more effective in inhibition of cell proliferation than fulvestrant in MCF-7 cells. Further optimization of I may lead to a new therapy for advanced ER+ breast cancer.
- 30Han, X.; Wang, C.; Qin, C.; Xiang, W.; Fernandez-Salas, E.; Yang, C.-Y.; Wang, M.; Zhao, L.; Xu, T.; Chinnaswamy, K.; Delproposto, J.; Stuckey, J.; Wang, S. Discovery of ARD-69 as a highly potent proteolysis targeting chimera (PROTAC) degrader of androgen receptor (AR) for the treatment of prostate cancer. J. Med. Chem. 2019, 62, 941– 964, DOI: 10.1021/acs.jmedchem.8b01631Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXlvFCjtQ%253D%253D&md5=2376573fb1a1e19c517d64c70199700aDiscovery of ARD-69 as a highly potent proteolysis targeting chimera (PROTAC) degrader of androgen receptor (AR) for the treatment of prostate cancerHan, Xin; Wang, Chao; Qin, Chong; Xiang, Weiguo; Fernandez-Salas, Ester; Yang, Chao-Yie; Wang, Mi; Zhao, Lijie; Xu, Tianfeng; Chinnaswamy, Krishnapriya; Delproposto, James; Stuckey, Jeanne; Wang, ShaomengJournal of Medicinal Chemistry (2019), 62 (2), 941-964CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)We report herein the discovery of highly potent PROTAC degraders of androgen receptor (AR), as exemplified by IARD-69. I induces degrdn. of AR protein in AR-pos. prostate cancer cell lines in a dose- and time-dependent manner. I achieves DC50 values of 0.86, 0.76, and 10.4 nM in LNCaP, VCaP, and 22Rv1 AR+ prostate cancer cell lines, resp. I is capable of reducing the AR protein level by >95% in these prostate cancer cell lines and effectively suppressing AR-regulated gene expression. I potently inhibits cell growth in these AR-pos. prostate cancer cell lines and is >100 times more potent than AR antagonists. A single dose of I effectively reduces the level of AR protein in xenograft tumor tissue in mice. Further optimization of I may ultimately lead to a new therapy for AR+, castration-resistant prostate cancer.
- 31Winter, G. E.; Buckley, D. L.; Paulk, J.; Roberts, J. M.; Souza, A.; Dhe-Paganon, S.; Bradner, J. E. Phthalimide conjugation as a strategy for in vivo target protein degradation. Science 2015, 348, 1376– 1381, DOI: 10.1126/science.aab1433Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXpvFansLk%253D&md5=b0277cc95d318d926b96e7a119066e3bPhthalimide conjugation as a strategy for in vivo target protein degradationWinter, Georg E.; Buckley, Dennis L.; Paulk, Joshiawa; Roberts, Justin M.; Souza, Amanda; Dhe-Paganon, Sirano; Bradner, James E.Science (Washington, DC, United States) (2015), 348 (6241), 1376-1381CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)The development of effective pharmacol. inhibitors of multidomain scaffold proteins, notably transcription factors, is a particularly challenging problem. In part, this is because many small-mol. antagonists disrupt the activity of only one domain in the target protein. The authors devised a chem. strategy that promotes ligand-dependent target protein degrdn. using as an example the transcriptional coactivator BRD4, a protein crit. for cancer cell growth and survival. The authors appended a competitive antagonist of BET bromodomains to a phthalimide moiety to hijack the cereblon E3 ubiquitin ligase complex. The resultant compd., dBET1, induced highly selective cereblon-dependent BET protein degrdn. in vitro and in vivo and delayed leukemia progression in mice. A second series of probes resulted in selective degrdn. of the cytosolic protein FKBP12. This chem. strategy for controlling target protein stability may have implications for therapeutically targeting previously intractable proteins.
- 32Steinebach, C.; Kehm, H.; Lindner, S.; Vu, L. P.; Köpff, S.; López Mármol, Á.; Weiler, C.; Wagner, K. G.; Reichenzeller, M.; Krönke, J.; Gütschow, M. PROTAC-mediated crosstalk between E3 ligases. Chem. Commun. 2019, 55, 1821– 1824, DOI: 10.1039/c8cc09541hGoogle Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtFSmtLY%253D&md5=17ed8abc73d45081c73f1995205713cfPROTAC-mediated crosstalk between E3 ligasesSteinebach, Christian; Kehm, Hannes; Lindner, Stefanie; Vu, Lan Phuong; Koepff, Simon; Lopez Marmol, Alvaro; Weiler, Corinna; Wagner, Karl G.; Reichenzeller, Michaela; Kroenke, Jan; Guetschow, MichaelChemical Communications (Cambridge, United Kingdom) (2019), 55 (12), 1821-1824CODEN: CHCOFS; ISSN:1359-7345. (Royal Society of Chemistry)Small-mol. heterobifunctional degraders can effectively control protein levels and are useful research tools. We assembled proteolysis targeting chimeras (PROTACs) from a cereblon (CRBN) and a von-Hippel-Lindau (VHL) ligase ligand and demonstrated a PROTAC-induced heterodimerization of the two E3 ligases leading to unidirectional and efficient degrdn. of CRBN.
- 33Lu, M.; Liu, T.; Jiao, Q.; Ji, J.; Tao, M.; Liu, Y.; You, Q.; Jiang, Z. Discovery of a Keap1-dependent peptide PROTAC to knockdown Tau by ubiquitination-proteasome degradation pathway. Eur. J. Med. Chem. 2018, 146, 251– 259, DOI: 10.1016/j.ejmech.2018.01.063Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXisFehsbY%253D&md5=165e6f510e4e2024aca6c4135d2840b2Discovery of a Keap1-dependent peptide PROTAC to knockdown Tau by ubiquitination-proteasome degradation pathwayLu, Mengchen; Liu, Tian; Jiao, Qiong; Ji, Jianai; Tao, Mengmin; Liu, Yijun; You, Qidong; Jiang, ZhengyuEuropean Journal of Medicinal Chemistry (2018), 146 (), 251-259CODEN: EJMCA5; ISSN:0223-5234. (Elsevier Masson SAS)Induced protein degrdn. by PROTACs has emerged as a promising strategy to target nonenzymic proteins inside the cell. The aim of this study was to identify Keap1, a substrate adaptor protein for ubiquitin E3 ligase involved in oxidative stress regulation, as a novel candidate for PROTACs that can be applied in the degrdn. of the nonenzymic protein Tau. A peptide PROTAC by recruiting Keap1-Cul3 ubiquitin E3 ligase was developed and applied in the degrdn. of intracellular Tau. Peptide 1 showed strong in vitro binding with Keap1 and Tau. With proper cell permeability, peptide 1 was found to colocalize with cellular Keap1 and resulted in the coimmunopptn. of Tau and Keap1. The results of flow cytometry and western blotting assays showed that peptide 1 can downregulate the intracellular Tau level in both time- and concn.-dependent manner. The application of Keap1 siRNA silencing and the proteasome inhibitor MG132 confirmed that peptide 1 could promote the Keap1-dependent poly-ubiquitination and proteasome-dependent degrdn. of Tau. The results suggested that using PROTACs to recruit Keap1 to induce the degrdn. of Tau may show promising character in the treatment of neurodegenerative disease. In addn., our research demonstrated that Keap1 should be a promising E3 ligase adaptor to be used in the design of novel PROTACs.
- 34Kargbo, R. B. Treatment of Alzheimer’s by PROTAC-Tau protein degradation. ACS Med. Chem. Lett. 2019, 10, 699– 700, DOI: 10.1021/acsmedchemlett.9b00083Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXkvVSnu70%253D&md5=7d4254b26c56f6401c16e6d100ddf0bdTreatment of Alzheimer's by PROTAC-Tau Protein DegradationKargbo, Robert B.ACS Medicinal Chemistry Letters (2019), 10 (5), 699-700CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)There is no expanded citation for this reference.
- 35Chu, T.-T.; Gao, N.; Li, Q.-Q.; Chen, P.-G.; Yang, X.-F.; Chen, Y.-X.; Zhao, Y.-F.; Li, Y.-M. Specific knockdown of endogenous Tau protein by peptide-directed ubiquitin-proteasome degradation. Cell Chem. Biol. 2016, 23, 453– 461, DOI: 10.1016/j.chembiol.2016.02.016Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XntlSqtbg%253D&md5=d965c74777427dfa9b6068f4f51a0f12Specific Knockdown of Endogenous Tau Protein by Peptide-Directed Ubiquitin-Proteasome DegradationChu, Ting-Ting; Gao, Na; Li, Qian-Qian; Chen, Pu-Guang; Yang, Xi-Fei; Chen, Yong-Xiang; Zhao, Yu-Fen; Li, Yan-MeiCell Chemical Biology (2016), 23 (4), 453-461CODEN: CCBEBM; ISSN:2451-9448. (Cell Press)Tau, an important pathol. protein of Alzheimer's disease (AD), can mediate the toxicity of amyloid β (Aβ). Thus, redn. of Tau with chem. mols. may offer a novel strategy for treating AD. Here, we designed and synthesized a series of multifunctional mols. that contained Tau-recognition moieties and E3 ligase-binding moieties to enhance Tau degrdn. Among these mols., TH006 had the highest activity of inducing Tau degrdn. by increasing its poly-ubiquitination. The decrement in Tau induced by TH006 could decrease the cytotoxicity caused by Aβ. Furthermore, TH006 could regulate the Tau level in the brain of an AD mouse model. Therefore, partial redn. of Tau with such multifunctional peptides may open up a novel therapeutic strategy for AD treatment.
- 36Boichenko, I.; Bär, K.; Deiss, S.; Heim, C.; Albrecht, R.; Lupas, A. N.; Hernandez Alvarez, B.; Hartmann, M. D. Chemical ligand space of cereblon. ACS Omega 2018, 3, 11163– 11171, DOI: 10.1021/acsomega.8b00959Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhslSku7bO&md5=b0fee25453d5ec83098ac0cf28575d44Chemical Ligand Space of CereblonBoichenko, Iuliia; Baer, Kerstin; Deiss, Silvia; Heim, Christopher; Albrecht, Reinhard; Lupas, Andrei N.; Hernandez Alvarez, Birte; Hartmann, Marcus D.ACS Omega (2018), 3 (9), 11163-11171CODEN: ACSODF; ISSN:2470-1343. (American Chemical Society)The protein, cereblon, serves as a substrate receptor of a ubiquitin ligase complex that can be tuned toward different target proteins by cereblon-binding agents. This approach to targeted protein degrdn. is exploited in different clin. settings and has sparked the development of a growing no. of thalidomide derivs. Here, we probed the chem. space of cereblon binding beyond such derivs. and worked out a simple set of chem. requirements, delineating the metaclass of cereblon effectors. We report co-crystal structures of Magnetospirillum gryphiswaldense cereblon isoform 4 with a diverse set of compds., including commonly used pharmaceuticals, but we also found that already minimalistic cereblon-binding moieties might exert teratogenic effects in zebrafish. These results may guide the design of a post-thalidomide generation of therapeutic cereblon effectors, and provide a framework for the circumvention of unintended cereblon-binding by neg. design for future pharmaceuticals.
- 37Burslem, G. M.; Ottis, P.; Jaime-Figueroa, S.; Morgan, A.; Cromm, P. M.; Toure, M.; Crews, C. M. Efficient synthesis of immunomodulatory drug analogues enables exploration of structure-degradation relationships. ChemMedChem 2018, 13, 1508– 1512, DOI: 10.1002/cmdc.201800271Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXht1KntL7O&md5=74ada081313c4e01b430fb6014441e2eEfficient Synthesis of Immunomodulatory Drug Analogues Enables Exploration of Structure-Degradation RelationshipsBurslem, George M.; Ottis, Philipp; Jaime-Figueroa, Saul; Morgan, Alicia; Cromm, Philipp M.; Toure, Momar; Crews, Craig M.ChemMedChem (2018), 13 (15), 1508-1512CODEN: CHEMGX; ISSN:1860-7179. (Wiley-VCH Verlag GmbH & Co. KGaA)The immunomodulatory drugs (IMiDs) thalidomide, pomalidomide, and lenalidomide have been approved for the treatment of multiple myeloma for many years. Recently, their use as E3 ligase recruiting elements for small-mol.-induced protein degrdn. has led to a resurgence in interest in IMiD synthesis and functionalization. Traditional IMiD synthesis follows a stepwise route with multiple purifn. steps. Herein we describe a novel one-pot synthesis without purifn. that provides rapid access to a multitude of IMiD analogs. Binding studies with the IMiD target protein cereblon (CRBN) reveals a narrow structure-activity relationship with only a few compds. showing sub-micromolar binding affinity in the range of pomalidomide and lenalidomide. However, anti-proliferative activity as well as Aiolos degrdn. could be identified for two IMiD analogs. This study provides useful insight into the structure-degrdn. relationships for mols. of this type as well as a rapid and robust method for IMiD synthesis.
- 38Bartlett, J. B.; Dredge, K.; Dalgleish, A. G. The evolution of thalidomide and its IMiD derivatives as anticancer agents. Nat. Rev. Cancer 2004, 4, 314– 322, DOI: 10.1038/nrc1323Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXis1Gmt7k%253D&md5=99d2ac4c0f6d6d1dac5547e5360dd74fTimeline: The evolution of thalidomide and its IMiD derivatives as anticancer agentsBartlett, J. Blake; Dredge, Keith; Dalgleish, Angus G.Nature Reviews Cancer (2004), 4 (4), 314-322CODEN: NRCAC4; ISSN:1474-175X. (Nature Publishing Group)A review. Thalidomide was originally used to treat morning sickness, but was banned in the 1960s for causing serious congenital birth defects. Remarkably, thalidomide was subsequently discovered to have anti-inflammatory and anti-angiogenic properties, and was identified as an effective treatment for multiple myeloma. A series of immunomodulatory drugs - created by chem. modification of thalidomide - have been developed to overcome the original devastating side effects. Their powerful anticancer properties mean that these drugs are now emerging from thalidomide's shadow as useful anticancer agents.
- 39Reist, M.; Carrupt, P.-A.; Francotte, E.; Testa, B. Chiral inversion and hydrolysis of thalidomide: mechanisms and catalysis by bases and serum albumin, and chiral stability of teratogenic metabolites. Chem. Res. Toxicol. 1998, 11, 1521– 1528, DOI: 10.1021/tx9801817Google Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXntFens7s%253D&md5=1952fa00825e6791eeb8388419f3cc09Chiral Inversion and Hydrolysis of Thalidomide: Mechanisms and Catalysis by Bases and Serum Albumin, and Chiral Stability of Teratogenic MetabolitesReist, Marianne; Carrupt, Pierre-Alain; Francotte, Eric; Testa, BernardChemical Research in Toxicology (1998), 11 (12), 1521-1528CODEN: CRTOEC; ISSN:0893-228X. (American Chemical Society)The chiral inversion and hydrolysis of thalidomide and the catalysis by bases and human serum albumin were investigated by using a stereoselective HPLC assay. Chiral inversion was catalyzed by albumin, hydroxyl ions, phosphate, and amino acids. Basic amino acids (Arg and Lys) had a superior potency in catalyzing chiral inversion compared to acid and neutral ones. The chiral inversion of thalidomide is thus subject to specific and general base catalysis, and it is suggested that the ability of HSA to catalyze the reaction is due to the basic groups of the amino acids Arg and Lys and not to a single catalytic site on the macromol. The hydrolysis of thalidomide was also base-catalyzed. However, albumin had no effect on hydrolysis, and there was no difference between the catalytic potencies of acidic, neutral, and basic amino acids. This may be explained by different reaction mechanisms of the chiral inversion and hydrolysis of thalidomide. Chiral inversion is deduced to occur by electrophilic substitution involving specific and general base catalysis, whereas hydrolysis is thought to occur by nucleophilic substitution involving specific and general base as well as nucleophilic catalysis. As nucleophilic attack is sensitive to steric properties of the catalyst, steric hindrance might be the reason albumin is not able to catalyze hydrolysis. 1H NMR expts. revealed that the three teratogenic metabolites of thalidomide, in sharp contrast to the drug itself, had complete chiral stability. This leads to the speculation that, were some enantioselectivity to exist in the teratogenicity of thalidomide, it could result from fast hydrolysis to chirally stable teratogenic metabolites.
- 40Chen, T. L.; Vogelsang, G. B.; Petty, B. G.; Brundrett, R. B.; Noe, D. A.; Santos, G. W.; Colvin, O. M. Plasma pharmacokinetics and urinary excretion of thalidomide after oral dosing in healthy male volunteers. Drug Metab Dispos 1989, 17, 402– 405Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL1MXlt1KnsLY%253D&md5=e171e48357c05ea8fedbc2979293650ePlasma pharmacokinetics and urinary excretion of thalidomide after oral dosing in healthy male volunteersChen, Tian Ling; Vogelsang, Georgia B.; Petty, Brent G.; Brundrett, Robert B.; Noe, Dennis A.; Santos, George W.; Colvin, O. MichaelDrug Metabolism and Disposition (1989), 17 (4), 402-5CODEN: DMDSAI; ISSN:0090-9556.The plasma pharmacokinetics and urinary excretion of thalidomide were evaluated in healthy male volunteers receiving a single oral dose of 200 mg. Concns. of thalidomide were detd. by a new HPLC assay. Plasma concn. vs. time data were well fit by a 1-compartment model. The mean peak concn., 1.15 μg/mL, was achieved at 4.39 h. Absorption and elimination half-lives were 1.70 and 8.70 h, resp., with a lag time of 0.41 h obsd. in 6 of 8 subjects. The apparent vol. of distribution and total body clearance rate, based on assumed complete bioavailability, were 120.69 L and 10.41 L/h. The urinary excretion of thalidomide accounted for only 0.6% of the total dose administered over 24 h, and the renal clearance rate was 0.08 L/h. Thus, the major route of elimination of thalidomide is nonrenal.
- 41Schumacher, H.; Smith, R. L.; Williams, R. T. The metabolism of thalidomide: the fate of thalidomide and some of its hydrolysis products in various species. Br. J. Pharmacol. Chemother. 1965, 25, 338– 351, DOI: 10.1111/j.1476-5381.1965.tb02054.xGoogle Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaF28Xhtl2rsA%253D%253D&md5=cb686e6f49ced088a96fc32b61f2256bThe metabolism of thalidomide. The fate of thalidomide and some of its hydrolysis products in various speciesSchumacher, H.; Smith, R. L.; Williams, R. T.British Journal of Pharmacology and Chemotherapy (1965), 25 (2), 338-51CODEN: BJPCAL; ISSN:0366-0826.cf. preceding abstr. The metabolism of thalidomide in the rabbit, rat, mouse, and guinea pig was investigated. The metabolites of thalidomide present in the urine, blood, and tissues of various species dosed with the drug were characterized by comparing their chromatographic mobility and color reactions with those given by authentic samples of the compds. The rabbit urinary metabolites were isolated in cryst. form by solvent extn. and adsorption chromatography, and their identity was established by analysis, m.p. behavior, and comparison of their ir spectra with those of the authentic compds. When thalidomide is fed to rabbits, rats, mice, and guinea pigs a no. of hydrolysis products appear in the urine. These hydrolysis products are formed by the spontaneous hydrolysis of thalidomide. In addn. the urine of rabbits dosed with thalidomide contains derivs. of 3- and 4-hydroxyphthalic acid; these minor metabolites were not identified. The hydrolysis products appear to be derived by spontaneous breakdown of thalidomide in the body, although it is possible that any of the hydrolytic reactions of thalidomide may be assisted by hydrolases in the body. In rats, some breakdown of thalidomide occurs in the gut before absorption; hydrolysis products are present in the gastrointestinal tract following the oral administration of the drug. Thalidomide and some of its hydrolysis products can be detected in the plasma and brain of rats dosed orally with thalidomide.
- 42Teo, S. K.; Colburn, W. A.; Tracewell, W. G.; Kook, K. A.; Stirling, D. I.; Jaworsky, M. S.; Scheffler, M. A.; Thomas, S. D.; Laskin, O. L. Clinical pharmacokinetics of thalidomide. Clin. Pharmacokinet. 2004, 43, 311– 327, DOI: 10.2165/00003088-200443050-00004Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXktFOqtbg%253D&md5=aa199f0c21a3fdb952fccde4059b71ecClinical pharmacokinetics of thalidomideTeo, Steve K.; Colburn, Wayne A.; Tracewell, William G.; Kook, Karin A.; Stirling, David I.; Jaworsky, Markian S.; Scheffler, Michael A.; Thomas, Steve D.; Laskin, Oscar L.Clinical Pharmacokinetics (2004), 43 (5), 311-327CODEN: CPKNDH; ISSN:0312-5963. (Adis International Ltd.)A review. Thalidomide is a racemic glutamic acid deriv. approved in the US for erythema nodosum leprosum, a complication of leprosy. In addn., its use in various inflammatory and oncol. conditions is being investigated. Thalidomide interconverts between the (R)- and (S)-enantiomers in plasma, with protein binding of 55% and 65%, resp. More than 90% of the absorbed drug is excreted in the urine and feces within 48 h. Thalidomide is minimally metabolized by the liver, but is spontaneously hydrolyzed into numerous renally excreted products. After a single oral dose of thalidomide 200mg (as the US-approved capsule formulation) in healthy volunteers, absorption is slow and extensive, resulting in a peak concn. (Cmax) of 1-2 mg/L at 3-4 h after administration, absorption lag time of 30 min, total exposure (AUC∞) of 18 mg • h/L, apparent elimination half-life of 6 h and apparent systemic clearance of 10 L/h. Thalidomide pharmacokinetics are best described by a one-compartment model with first-order absorption and elimination. Because of the low soly. of the drug in the gastrointestinal tract, thalidomide exhibits absorption rate-limited pharmacokinetics (the "flip-flop" phenomenon), with its elimination rate being faster than its absorption rate. The apparent elimination half-life of 6 h therefore represents absorption, not elimination. The "true" apparent vol. of distribution was estd. to be 16L by use of the faster elimination-rate half-life. Multiple doses of thalidomide 200 mg/day over 21 days cause no change in the pharmacokinetics, with a steady-state Cmax (Cssmax) of 1.2 mg/L. Simulation of 400 and 800 mg/day also shows no accumulation, with Cssmax of 3.5 and 6.0 mg/L, resp. Multiple-dose studies in cancer patients show pharmacokinetics comparable with those in healthy populations at similar dosages. Thalidomide exhibits a dose-proportional increase in AUC at doses from 50 to 400mg. Because of the low soly. of thalidomide, Cmax is less than proportional to dose, and tmax is prolonged with increasing dose. Age, sex and smoking have no effect on the pharmacokinetics of thalidomide, and the effect of food is minimal. Thalidomide does not alter the pharmacokinetics of oral contraceptives, and is also unlikely to interact with warfarin and grapefruit juice. Since thalidomide is mainly hydrolyzed and passively excreted, its pharmacokinetics are not expected to change in patients with impaired liver or kidney function.
- 43Beckmann, R. Ueber das Verhalten von Thalidomid im Organismus. Arzneimittelforschung 1963, 13, 185– 191Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaF387mtF2mug%253D%253D&md5=211cb3a294eb7d33c0cfd689bb37d2bcC-14])BECKMANN RArzneimittel-Forschung (1963), 13 (), 185-91 ISSN:0004-4172.There is no expanded citation for this reference.
- 44Chung, F.; Lu, J.; Palmer, B. D.; Kestell, P.; Browett, P.; Baguley, B. C.; Tingle, M.; Ching, L. M. Thalidomide pharmacokinetics and metabolite formation in mice, rabbits, and multiple myeloma patients. Clin. Cancer Res. 2004, 10, 5949– 5956, DOI: 10.1158/1078-0432.ccr-04-0421Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXntlyjtLw%253D&md5=51bea71546be92322a511405e004aa80Thalidomide pharmacokinetics and metabolite formation in mice, rabbits, and multiple myeloma patientsChung, Francisco; Lu, Jun; Palmer, Brian D.; Kestell, Philip; Browett, Peter; Baguley, Bruce C.; Tingle, Malcolm; Ching, Lai-MingClinical Cancer Research (2004), 10 (17), 5949-5956CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)Thalidomide has a variety of biol. effects that vary considerably according to the species tested. The authors sought to establish whether differences in pharmacokinetics could form a basis for the species-specific effects of thalidomide. Mice and rabbits were administered thalidomide (2 mg/kg) p.o. or i.v., and plasma concns. of thalidomide were measured after drug administration using high performance liq. chromatog. Plasma samples from five multiple myeloma patients over 24 h after their first dose of thalidomide (200 mg) were similarly analyzed and all data were fitted to a one-compartment model. Metabolites of thalidomide in plasma were identified simultaneously using liq. chromatog.-mass spectrometry. Plasma concn.-time profiles for the individual patients were very similar to each other, but widely different pharmacokinetic properties were found between patients compared with those in mice or rabbits. Area under the concn. curve values for mice, rabbits, and multiple myeloma patients were 4, 8, and 81 μmol/L · hour, resp., and corresponding elimination half-lives were 0.5, 2.2, and 7.3 h, resp. Large differences were also obsd. between the metabolite profiles from the three species. Hydrolysis products were detected for all species, and the proportion of hydroxylated metabolites was higher in mice than in rabbits and undetectable in patients. Our results show major interspecies differences in the pharmacokinetics of thalidomide that are related to the altered degree of metab. The authors suggest that the interspecies differences in biol. effects of thalidomide may be attributable, at least in part, to the differences in its metab. and hence pharmacokinetics.
- 45Nakamura, T.; Noguchi, T.; Miyachi, H.; Hashimoto, Y. Hydrolyzed metabolites of thalidomide: synthesis and TNF-alpha production-inhibitory activity. Chem. Pharm. Bull. 2007, 55, 651– 654, DOI: 10.1248/cpb.55.651Google Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXls1arsLc%253D&md5=32f1066b688a9de33e9dbf5da42ec514Hydrolyzed metabolites of thalidomide: synthesis and TNF-α production-inhibitory activityNakamura, Takanori; Noguchi, Tomomi; Miyachi, Hiroyuki; Hashimoto, YuichiChemical & Pharmaceutical Bulletin (2007), 55 (4), 651-654CODEN: CPBTAL; ISSN:0009-2363. (Pharmaceutical Society of Japan)Putative hydrolyzed metabolites of thalidomide (I) were prepd. and characterized, and their inhibitory activity on tumor necrosis factor (TNF)-α prodn. in the human monocytic leukemia cell line THP-1 was evaluated. α-(2-Carboxybenzamido)glutarimide (II) was a more potent TNF-α prodn. inhibitor than I.
- 46Otogawa, K.; Ogino, Y.; Ishikawa, K.; Tanaka, M.; Shiro, M.; Osaka, T.; Asahi, T. Structural and thermal analyses of a hydrolysis compound of thalidomide. Acta Crystallogr., Sect. A: Found. Adv. 2014, 70, C113, DOI: 10.1107/s2053273314098866Google ScholarThere is no corresponding record for this reference.
- 47Krönke, J.; Hurst, S. N.; Ebert, B. L. Lenalidomide induces degradation of IKZF1 and IKZF3. OncoImmunology 2014, 3, e941742 DOI: 10.4161/21624011.2014.941742Google ScholarThere is no corresponding record for this reference.
- 48Schafer, P. H.; Ye, Y.; Wu, L.; Kosek, J.; Ringheim, G.; Yang, Z.; Liu, L.; Thomas, M.; Palmisano, M.; Chopra, R. Cereblon modulator iberdomide induces degradation of the transcription factors Ikaros and Aiolos: immunomodulation in healthy volunteers and relevance to systemic lupus erythematosus. Ann. Rheum. Dis. 2018, 77, 1516– 1523, DOI: 10.1136/annrheumdis-2017-212916Google Scholar48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXjtVektbw%253D&md5=200098adf4787ee935df528d18a5508dCereblon modulator iberdomide induces degradation of the transcription factors Ikaros and Aiolos: immunomodulation in healthy volunteers and relevance to systemic lupus erythematosusSchafer, Peter H.; Ye, Ying; Wu, Lei; Kosek, Jolanta; Ringheim, Garth; Yang, Zhihong; Liu, Liangang; Thomas, Michael; Palmisano, Maria; Chopra, RajeshAnnals of the Rheumatic Diseases (2018), 77 (10), 1516-1523CODEN: ARDIAO; ISSN:0003-4967. (BMJ Publishing Group)IKZF1 and IKZF3 (encoding transcription factors Ikaros and Aiolos) are susceptibility loci for systemic lupus erythematosus (SLE). The pharmacol. of iberdomide (CC-220), a cereblon (CRBN) modulator targeting Ikaros and Aiolos, was studied in SLE patient cells and in a phase 1 healthy volunteer study. CRBN, IKZF1 and IKZF3 gene expression was measured in peripheral blood mononuclear cells (PBMC) from patients with SLE and healthy volunteers. Ikaros and Aiolos protein levels were measured by Western blot and flow cytometry. Anti-dsDNA and anti-phospholipid autoantibodies were measured in SLE PBMC cultures treated for 7 days with iberdomide. Fiftysix healthy volunteers were randomized to a single dose of iberdomide (0.03-6mg, n=6 across seven cohorts) or placebo (n=2/cohort). CD19+ B cells, CD3+ T cells and intracellular Aiolos were measured by flow cytometry. Interleukin (IL)-2 and IL-1β prodn. was stimulated with anti-CD3 and lipopolysaccharide, resp., in an ex vivo whole blood assay. SLE patient PBMCs expressed significantly higher CRBN (1.5-fold), IKZF1 (2.1-fold) and IKZF3 (4.1- fold) mRNA levels compared with healthy volunteers. Iberdomide significantly reduced Ikaros and Aiolos protein levels in B cells, T cells and monocytes. In SLE PBMC cultures, iberdomide inhibited anti-dsDNA and anti-phospholipid autoantibody prodn. (IC50 ≈10 nM). Single doses of iberdomide (0.3-6mg) in healthy volunteers decreased intracellular Aiolos (min. mean per cent of baseline: ≈12%-28% (B cells); ≈0%-33% (T cells)), decreased abs. CD19+ B cells, increased IL-2 and decreased IL-1β prodn. ex vivo. These findings demonstrate pharmacodynamic activity of iberdomide and support its further clin. development for the treatment of SLE.
- 49Matyskiela, M. E.; Zhang, W.; Man, H.-W.; Muller, G.; Khambatta, G.; Baculi, F.; Hickman, M.; LeBrun, L.; Pagarigan, B.; Carmel, G.; Lu, C.-C.; Lu, G.; Riley, M.; Satoh, Y.; Schafer, P.; Daniel, T. O.; Carmichael, J.; Cathers, B. E.; Chamberlain, P. P. A Cereblon Modulator (CC-220) with Improved Degradation of Ikaros and Aiolos. J. Med. Chem. 2018, 61, 535– 542, DOI: 10.1021/acs.jmedchem.6b01921Google Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXmt1KqsLs%253D&md5=8358ae8563f8f2c09dbf68683e5e4008A Cereblon Modulator (CC-220) with Improved Degradation of Ikaros and AiolosMatyskiela, Mary E.; Zhang, Weihong; Man, Hon-Wah; Muller, George; Khambatta, Godrej; Baculi, Frans; Hickman, Matthew; LeBrun, Laurie; Pagarigan, Barbra; Carmel, Gilles; Lu, Chin-Chun; Lu, Gang; Riley, Mariko; Satoh, Yoshitaka; Schafer, Peter; Daniel, Thomas O.; Carmichael, James; Cathers, Brian E.; Chamberlain, Philip P.Journal of Medicinal Chemistry (2018), 61 (2), 535-542CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The drugs lenalidomide and pomalidomide bind to the protein cereblon, directing the CRL4-CRBN E3 ligase toward the transcription factors Ikaros and Aiolos to cause their ubiquitination and degrdn. Here we describe CC-220 (compd. 6), a cereblon modulator in clin. development for systemic lupus erythematosis and relapsed/refractory multiple myeloma. Compd. 6 binds cereblon with a higher affinity than lenalidomide or pomalidomide. Consistent with this, the cellular degrdn. of Ikaros and Aiolos is more potent and the extent of substrate depletion is greater. The crystal structure of cereblon in complex with DDB1 and compd. 6 reveals that the increase in potency correlates with increased contacts between compd. 6 and cereblon away from the modeled binding site for Ikaros/Aiolos. These results describe a new cereblon modulator which achieves greater substrate degrdn. via tighter binding to the cereblon E3 ligase and provides an example of the effect of E3 ligase binding affinity with relevance to other drug discovery efforts in targeted protein degrdn.
- 50Capitosti, S. M.; Hansen, T. P.; Brown, M. L. Facile synthesis of an azido-labeled thalidomide analogue. Org. Lett. 2003, 5, 2865– 2867, DOI: 10.1021/ol034906wGoogle Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXlt1Cgs70%253D&md5=fdefbdcf05d15f638e322bc9e5687ebbFacile Synthesis of an Azido-Labeled Thalidomide AnalogueCapitosti, Scott M.; Hansen, Todd P.; Brown, Milton L.Organic Letters (2003), 5 (16), 2865-2867CODEN: ORLEF7; ISSN:1523-7060. (American Chemical Society)A five-step synthesis of an azido-thalidomide analog I is presented. The sequence requires cheap and readily available starting materials and reagents, and only two steps require purifn. Addnl., I possesses activity comparable to that of thalidomide in inhibiting the proliferation of human microvascular endothelial cells, thus providing impetus for its use as a potential photoaffinity label of thalidomide.
- 51Joossens, J.; Van der Veken, P.; Lambeir, A.-M.; Augustyns, K.; Haemers, A. Development of irreversible diphenyl phosphonate inhibitors for urokinase plasminogen activator. J. Med. Chem. 2004, 47, 2411– 2413, DOI: 10.1021/jm0499209Google Scholar51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXisl2it7k%253D&md5=a5e0eb9791dc09a1ac71b6cf8415af29Development of Irreversible Diphenyl Phosphonate Inhibitors for Urokinase Plasminogen ActivatorJoossens, J.; Van der Veken, P.; Lambeir, A.-M.; Augustyns, K.; Haemers, A.Journal of Medicinal Chemistry (2004), 47 (10), 2411-2413CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The authors report the synthesis and biochem. evaluation of selective, irreversible di-Ph phosphonate inhibitors for urokinase plasminogen activator (uPA). A di-Ph phosphonate group was introduced on the substrate-like peptide Z-D-Ser-Ala-Arg, and modification of the guanidine side chain was investigated. A guanylated benzyl group appeared the most promising side chain modification. A kapp value in the 103 M-1 s-1 range for uPA was obtained, together with a selectivity index higher than 240 toward other trypsin-like proteases such as tPA, thrombin, plasmin, and FXa.
- 52Kanuma, K.; Omodera, K.; Nishiguchi, M.; Funakoshi, T.; Chaki, S.; Nagase, Y.; Iida, I.; Yamaguchi, J.-i.; Semple, G.; Tran, T.-A.; Sekiguchi, Y. Identification of 4-amino-2-cyclohexylaminoquinazolines as metabolically stable melanin-concentrating hormone receptor 1 antagonists. Bioorg. Med. Chem. 2006, 14, 3307– 3319, DOI: 10.1016/j.bmc.2005.12.052Google Scholar52https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XjtFGjtb4%253D&md5=99bcc1d1e058b4c8e5baa2efcbae7447Identification of 4-amino-2-cyclohexylaminoquinazolines as metabolically stable melanin-concentrating hormone receptor 1 antagonistsKanuma, Kosuke; Omodera, Katsunori; Nishiguchi, Mariko; Funakoshi, Takeo; Chaki, Shigeyuki; Nagase, Yasuko; Iida, Izumi; Yamaguchi, Jun-ichi; Semple, Graeme; Tran, Thuy-Anh; Sekiguchi, YoshinoriBioorganic & Medicinal Chemistry (2006), 14 (10), 3307-3319CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)The optimization of the distance between two key pharmacophore features within our first hit compds. led to the identification of a new class of potent non-peptidic antagonists for the MCH-R1, based around 4-amino-2-cyclohexylaminoquinazolines. In particular, ATC0065, N 2-[cis-4-({2-[4-Bromo-2-(trifluoromethoxy)phenyl]ethyl}amino)cyclohexyl]-N4,N4-dimethylquinazoline-2,4-diamine dihydrochloride, bound with high affinity to the MCH-R1 (IC50 value of 16 nM) and showed good metabolic stability in liver microsomes from human and rat.
- 53Nouch, R.; Cini, M.; Magre, M.; Abid, M.; Diéguez, M.; Pàmies, O.; Woodward, S.; Lewis, W. Enantioselective synthesis of 6,6-disubstituted pentafulvenes containing a chiral pendant hydroxy group. Chem.—Eur. J. 2017, 23, 17195– 17198, DOI: 10.1002/chem.201704247Google Scholar53https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhvVKmurjI&md5=ed815c390637d2d2ca7c6322180f583bEnantioselective Synthesis of 6,6-Disubstituted Pentafulvenes Containing a Chiral Pendant Hydroxy GroupNouch, Ryan; Cini, Melchior; Magre, Marc; Abid, Mohammed; Dieguez, Montserrat; Pamies, Oscar; Woodward, Simon; Lewis, WilliamChemistry - A European Journal (2017), 23 (68), 17195-17198CODEN: CEUJED; ISSN:0947-6539. (Wiley-VCH Verlag GmbH & Co. KGaA)Simple enantioselective synthesis of 6,6-disubstituted pentafulvenes bearing chiral pendant hydroxy groups are attained by cascade reactivity using com. available proline-based organocatalysts. Condensation of cyclopentadiene with the acetyl function of a 1,2-formylacetophenone, followed by cyclization of a resulting fulvene-stabilized carbanion with the formyl group, generates bicyclic chiral alcs. with initial er values up to 94:6. Exceptional enantio-enrichment of the resultant alcs. results upon crystn.-even near racemic samples spontaneously de-racemize. This enables new families of substituted cyclopentadienes that are both enantiomerically and diastereomerically pure to be rapidly attained.
- 54Cao, R.; Müller, P.; Lippard, S. J. Tripodal tris-tacn and tris-dpa platforms for assembling phosphate-templated trimetallic centers. J. Am. Chem. Soc. 2010, 132, 17366– 17369, DOI: 10.1021/ja108212vGoogle Scholar54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhsVGksbzJ&md5=26ced8d2ad3686926092f1940b96a9ccTripodal Tris-tacn and Tris-dpa Platforms for Assembling Phosphate-Templated Trimetallic CentersCao, Rui; Muller, Peter; Lippard, Stephen J.Journal of the American Chemical Society (2010), 132 (49), 17366-17369CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Multidentate tripodal ligands, N(CH2-m-C6H4-CH2tacn)3 (L1) and N(CH2-o-C6H4-CH2N(CH2py)2)3 (L2), were devised for assembling high-nuclearity metal clusters. By using the same tripodal platform with different ligand appendages, either triazacyclononanes or dipicolylamines, and functionalizing either the ortho or the meta positions on the tris(xylyl) linker arms, discrete trimetal phosphate units of relevance to phosphate-metabolizing trimetallic centers in biol. were prepd. Four such compds., [(CuIICl)3(HPO4)L1](PF6) (1), [(CuIICl)3(HAsO4)L1](PF6) (2), Na2[MnIII6MnII2(H2O)2(HPO4)6(PO4)4(L1)2] (3), and [CoII3(H2PO4)Cl2(MeCN)L2](PF6)3 (4), all contg. three metal centers bound to a central phosphate or arsenate unit bridging oxygen atoms, were synthesized and structurally characterized. These results demonstrate the propensity of this novel tripodal ligand platform, in the presence of phosphate or arsenate, to assemble {M3(EO4)} units and thus structurally mimic trimetallic active sites of proteins involved in phosphate metab. Reactivity studies reveal that the tricopper complex 1 is more efficient than monocopper analogs in catalyzing the hydrolysis of 4-nitrophenyl phosphate.
- 55Steinebach, C.; Lindner, S.; Udeshi, N. D.; Mani, D. C.; Kehm, H.; Köpff, S.; Carr, S. A.; Gütschow, M.; Krönke, J. Homo-PROTACs for the chemical knockdown of cereblon. ACS Chem. Biol. 2018, 13, 2771– 2782, DOI: 10.1021/acschembio.8b00693Google Scholar55https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsFejsLrO&md5=71bf2d53ad23c2b873f754de5144883cHomo-PROTACs for the Chemical Knockdown of CereblonSteinebach, Christian; Lindner, Stefanie; Udeshi, Namrata D.; Mani, Deepak C.; Kehm, Hannes; Koepff, Simon; Carr, Steven A.; Guetschow, Michael; Kroenke, JanACS Chemical Biology (2018), 13 (9), 2771-2782CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)The immunomodulatory drugs (IMiDs) thalidomide, lenalidomide, and pomalidomide, all approved for the treatment of multiple myeloma, induce targeted ubiquitination and degrdn. of Ikaros (IKZF1) and Aiolos (IKZF3) via the cereblon (CRBN) E3 ubiquitin ligase. IMiD-based proteolysis-targeting chimeras (PROTACs) can efficiently recruit CRBN to a protein of interest, leading to its ubiquitination and proteasomal degrdn. By linking two pomalidomide mols., we designed homobifunctional, so-called homo-PROTACs and investigated their ability to induce self-directed ubiquitination and degrdn. The homodimerized compd. I was characterized as a highly potent and efficient CRBN degrader with only minimal effects on IKZF1 and IKZF3. The cellular selectivity of I for CRBN degrdn. was confirmed at the proteome level by quant. mass spectrometry. Inactivation by compd. I did not affect proliferation of different cell lines, prevented pomalidomide-induced degrdn. of IKZF1 and IKZF3, and antagonized the effects of pomalidomide on multiple myeloma cells. Homobifunctional CRBN degraders will be useful tools for future biomedical investigations of CRBN-related signaling and may help to further elucidate the mol. mechanism of thalidomide analogs.
- 56Boichenko, I.; Deiss, S.; Bär, K.; Hartmann, M. D.; Hernandez Alvarez, B. A FRET-based assay for the identification and characterization of cereblon ligands. J. Med. Chem. 2016, 59, 770– 774, DOI: 10.1021/acs.jmedchem.5b01735Google Scholar56https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xjs1yktg%253D%253D&md5=f0400506428f439e81f579204e0bd39dA FRET-Based Assay for the Identification and Characterization of Cereblon LigandsBoichenko, Iuliia; Deiss, Silvia; Baer, Kerstin; Hartmann, Marcus D.; Hernandez Alvarez, BirteJournal of Medicinal Chemistry (2016), 59 (2), 770-774CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Cereblon serves as an ubiquitin ligase substrate receptor that can be tuned toward different target proteins by various cereblon-binding agents. This offers one of the most promising avenues for targeted protein degrdn. in cancer therapy, but cereblon binding can also mediate teratogenic effects. We present an effective assay that is suited for high-throughput screening of compd. libraries for off-target cereblon interactions but also can guide lead optimization and rational design of novel cereblon effector mols.
- 57Kabsch, W. Xds. Acta Crystallogr., Sect. D: Biol. Crystallogr. 2010, 66, 125– 132, DOI: 10.1107/s0907444909047337Google Scholar57https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhs1SisLc%253D&md5=1aa9a38aeb3ce95af4ffb7d8b8a142bdSoftware XDS for image rotation, recognition and crystal symmetry assignmentKabsch, WolfgangActa Crystallographica, Section D: Biological Crystallography (2010), 66 (2), 125-132CODEN: ABCRE6; ISSN:0907-4449. (International Union of Crystallography)The usage and control of recent modifications of the program package XDS for the processing of rotation images are described in the context of previous versions. New features include automatic detn. of spot size and reflecting range and recognition and assignment of crystal symmetry. Moreover, the limitations of earlier package versions on the no. of correction/scaling factors and the representation of pixel contents have been removed. Large program parts have been restructured for parallel processing so that the quality and completeness of collected data can be assessed soon after measurement.
- 58Vagin, A.; Teplyakov, A. Molecular replacement with MOLREP. Acta Crystallogr., Sect. D: Biol. Crystallogr. 2010, 66, 22– 25, DOI: 10.1107/s0907444909042589Google Scholar58https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXit1Kktw%253D%253D&md5=820d114719aca209994ffb0403e3b20dMolecular replacement with MOLREPVagin, Alexei; Teplyakov, AlexeiActa Crystallographica, Section D: Biological Crystallography (2010), 66 (1), 22-25CODEN: ABCRE6; ISSN:0907-4449. (International Union of Crystallography)MOLREP is an automated program for mol. replacement that utilizes a no. of original approaches to rotational and translational search and data prepn. Since the first publication describing the program, MOLREP has acquired a variety of features that include weighting of the X-ray data and search models, multi-copy search, fitting the model into electron d., structural superposition of two models and rigid-body refinement. The program can run in a fully automatic mode using optimized parameters calcd. from the input data.
- 59Emsley, P.; Lohkamp, B.; Scott, W. G.; Cowtan, K. Features and development of Coot. Acta Crystallogr., Sect. D: Biol. Crystallogr. 2010, 66, 486– 501, DOI: 10.1107/s0907444910007493Google Scholar59https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXksFKisb8%253D&md5=67262cbfc60004de5ef962d5c043c910Features and development of CootEmsley, P.; Lohkamp, B.; Scott, W. G.; Cowtan, K.Acta Crystallographica, Section D: Biological Crystallography (2010), 66 (4), 486-501CODEN: ABCRE6; ISSN:0907-4449. (International Union of Crystallography)Coot is a mol.-graphics application for model building and validation of biol. macromols. The program displays electron-d. maps and at. models and allows model manipulations such as idealization, real-space refinement, manual rotation/translation, rigid-body fitting, ligand search, solvation, mutations, rotamers and Ramachandran idealization. Furthermore, tools are provided for model validation as well as interfaces to external programs for refinement, validation and graphics. The software is designed to be easy to learn for novice users, which is achieved by ensuring that tools for common tasks are 'discoverable' through familiar user-interface elements (menus and toolbars) or by intuitive behavior (mouse controls). Recent developments have focused on providing tools for expert users, with customisable key bindings, extensions and an extensive scripting interface. The software is under rapid development, but has already achieved very widespread use within the crystallog. community. The current state of the software is presented, with a description of the facilities available and of some of the underlying methods employed.
- 60Schrodinger, LLC. The PyMOL Molecular Graphics System, Version 2.2.1.Google ScholarThere is no corresponding record for this reference.
Cited By
This article is cited by 41 publications.
- Christian Steinebach, Aleša Bricelj, Arunima Murgai, Izidor Sosič, Luca Bischof, Yuen Lam Dora Ng, Christopher Heim, Samuel Maiwald, Matic Proj, Rabea Voget, Felix Feller, Janez Košmrlj, Valeriia Sapozhnikova, Annika Schmidt, Maximilian Rudolf Zuleeg, Patricia Lemnitzer, Philipp Mertins, Finn K. Hansen, Michael Gütschow, Jan Krönke, Marcus D. Hartmann. Leveraging Ligand Affinity and Properties: Discovery of Novel Benzamide-Type Cereblon Binders for the Design of PROTACs. Journal of Medicinal Chemistry 2023, 66
(21)
, 14513-14543. https://doi.org/10.1021/acs.jmedchem.3c00851
- Marina Miñarro-Lleonar, Andrea Bertran-Mostazo, Jorge Duro, Xavier Barril, Jordi Juárez-Jiménez. Lenalidomide Stabilizes Protein–Protein Complexes by Turning Labile Intermolecular H-Bonds into Robust Interactions. Journal of Medicinal Chemistry 2023, 66
(9)
, 6037-6046. https://doi.org/10.1021/acs.jmedchem.2c01692
- Fang Bai, Ning Wang, Yinshan Bai, Xiaowei Ma, Chengzhi Gu, Bin Dai, Jianpeng Chen. NHPI-Mediated Electrochemical α-Oxygenation of Amides to Benzimides. The Journal of Organic Chemistry 2023, 88
(5)
, 2985-2998. https://doi.org/10.1021/acs.joc.2c02700
- Lisa J. Alcock, Yunchao Chang, Jamie A. Jarusiewicz, Marisa Actis, Stanley Nithianantham, Anand Mayasundari, Jaeki Min, Dylan Maxwell, Jeremy Hunt, Brandon Smart, Jun J. Yang, Gisele Nishiguchi, Marcus Fischer, Charles G. Mullighan, Zoran Rankovic. Development of Potent and Selective Janus Kinase 2/3 Directing PG–PROTACs. ACS Medicinal Chemistry Letters 2022, 13
(3)
, 475-482. https://doi.org/10.1021/acsmedchemlett.1c00650
- Aleša Bricelj, Yuen Lam Dora Ng, Dominic Ferber, Robert Kuchta, Sina Müller, Marius Monschke, Karl G. Wagner, Jan Krönke, Izidor Sosič, Michael Gütschow, Christian Steinebach. Influence of Linker Attachment Points on the Stability and Neosubstrate Degradation of Cereblon Ligands. ACS Medicinal Chemistry Letters 2021, 12
(11)
, 1733-1738. https://doi.org/10.1021/acsmedchemlett.1c00368
- Samuel Maiwald, Christopher Heim, Birte Hernandez Alvarez, Marcus D. Hartmann. Sweet and Blind Spots in E3 Ligase Ligand Space Revealed by a Thermophoresis-Based Assay. ACS Medicinal Chemistry Letters 2021, 12
(1)
, 74-81. https://doi.org/10.1021/acsmedchemlett.0c00440
- James S. Scott, Iacovos N. Michaelides, Markus Schade. Property-based optimisation of PROTACs. RSC Medicinal Chemistry 2025, 51 https://doi.org/10.1039/D4MD00769G
- Elisia Villemure, Yong Wang, Yuebiao Zhou, Alice R. Wong, Christian Nilewski. Tactics and Strategies for the Synthesis of Cereblon Ligands. Synthesis 2024, 56
(23)
, 3543-3554. https://doi.org/10.1055/s-0043-1775385
- Esther J. Han, Myungeun Jeong, Seoung Rak Lee, Erik J. Sorensen, Mohammad R. Seyedsayamdost. Hirocidins, Cytotoxic Metabolites from
Streptomyces hiroshimensis
, Induce Mitochondrion‐Mediated Apoptosis. Angewandte Chemie International Edition 2024, 63
(37)
https://doi.org/10.1002/anie.202405367
- Esther J. Han, Myungeun Jeong, Seoung Rak Lee, Erik J. Sorensen, Mohammad R. Seyedsayamdost. Hirocidins, Cytotoxic Metabolites from
Streptomyces hiroshimensis
, Induce Mitochondrion‐Mediated Apoptosis. Angewandte Chemie 2024, 136
(37)
https://doi.org/10.1002/ange.202405367
- Michael J Nutt, Scott G Stewart. Strengthening Molecular Glues: Design Strategies for Improving Thalidomide Analogs as Cereblon Effectors and Anticancer Agents. Drug Discovery Today 2024, 29
(6)
, 104010. https://doi.org/10.1016/j.drudis.2024.104010
- Saki Ichikawa, N. Connor Payne, Wenqing Xu, Chia-Fu Chang, Nandini Vallavoju, Spencer Frome, Hope A. Flaxman, Ralph Mazitschek, Christina M. Woo. The cyclimids: Degron-inspired cereblon binders for targeted protein degradation. Cell Chemical Biology 2024, 31
(6)
, 1162-1175.e10. https://doi.org/10.1016/j.chembiol.2024.01.003
- Robert Shevalev, Luca Bischof, Alexander Sapegin, Alexander Bunev, Grigor'eva Olga, Grigory Kantin, Stanislav Kalinin, Marcus D. Hartmann. Discovery and characterization of potent spiro-isoxazole-based cereblon ligands with a novel binding mode. European Journal of Medicinal Chemistry 2024, 270 , 116328. https://doi.org/10.1016/j.ejmech.2024.116328
- Yilin Chen, Haoan Xue, Jianping Jin. Applications of protein ubiquitylation and deubiquitylation in drug discovery. Journal of Biological Chemistry 2024, 21 , 107264. https://doi.org/10.1016/j.jbc.2024.107264
- Kangbo Feng, Jiong Chen, Shuangxi Gu, Haifeng Wang, Fen'er Chen. New Progress of Fully Continuous Flow Reaction Technologies in Pharmaceutical Synthesis (2019~2022). Chinese Journal of Organic Chemistry 2024, 44
(2)
, 378. https://doi.org/10.6023/cjoc202307005
- Jaeyong Cho, Yukio Horikawa, Yuki Oiwa, Kazuyoshi Hosomichi, Daisuke Yabe, Takeshi Imai. Glucokinase Variant Proteins Are Resistant to Fasting-Induced Uridine Diphosphate Glucose-Dependent Degradation in Maturity-Onset Diabetes of the Young Type 2 Patients. International Journal of Molecular Sciences 2023, 24
(21)
, 15842. https://doi.org/10.3390/ijms242115842
- Sergej Semochkin. Помалидомид в терапии рецидивирующей/рефрактерной множественной миеломы. Journal of Modern Oncology 2023, 25
(2)
, 159-167. https://doi.org/10.26442/18151434.2023.2.202255
- Dazhao Mi, Yuzhan Li, Haijun Gu, Yan Li, Yihua Chen. Current advances of small molecule E3 ligands for proteolysis-targeting chimeras design. European Journal of Medicinal Chemistry 2023, 256 , 115444. https://doi.org/10.1016/j.ejmech.2023.115444
- Scott Hollingsworth, Scott Johnson, Pouyan Khakbaz, Yilin Meng, Varnavas Mouchlis, Olivia Pierce, Vera Prytkova, Erik Vik, Dahlia Weiss, Veerabahu Shanmugasundaram. The rise of targeting chimeras (TACs): next-generation medicines that preempt cellular events. Medicinal Chemistry Research 2023, 32
(7)
, 1294-1314. https://doi.org/10.1007/s00044-023-03104-z
- Xin Han, Yi Sun. PROTACs: A novel strategy for cancer drug discovery and development. MedComm 2023, 4
(3)
https://doi.org/10.1002/mco2.290
- Robert Kuchta, Christopher Heim, Alexander Herrmann, Samuel Maiwald, Yuen Lam Dora Ng, Izidor Sosič, Tim Keuler, Jan Krönke, Michael Gütschow, Marcus D. Hartmann, Christian Steinebach. Accessing three-branched high-affinity cereblon ligands for molecular glue and protein degrader design. RSC Chemical Biology 2023, 4
(3)
, 229-234. https://doi.org/10.1039/D2CB00223J
- Jian Yan, Zhibing Zheng. Discovery of Highly Potent CRBN Ligands and Insight into Their Binding Mode through Molecular Docking and Molecular Dynamics Simulations. ChemMedChem 2023, 18
(5)
https://doi.org/10.1002/cmdc.202200573
- Christopher Heim, Anna-Katharina Spring, Sören Kirchgäßner, Dirk Schwarzer, Marcus D. Hartmann. Cereblon neo-substrate binding mimics the recognition of the cyclic imide degron. Biochemical and Biophysical Research Communications 2023, 646 , 30-35. https://doi.org/10.1016/j.bbrc.2023.01.051
- Athanassios Giannis. Thalidomid: Fluch, Segen und Triumph eines Schlafmittels. 2023, 137-150. https://doi.org/10.1007/978-3-662-67510-6_11
- Jaeyong Cho, Atsushi Miyagawa, Kazuki Yamaguchi, Wakana Abe, Yoji Tsugawa, Hatsuo Yamamura, Takeshi Imai. UDP-glucose, cereblon-dependent proinsulin degrader. Scientific Reports 2022, 12
(1)
https://doi.org/10.1038/s41598-022-18902-5
- Maria Ivanova, Julien Legros, Thomas Poisson, Philippe Jubault. A multi-step continuous flow synthesis of pomalidomide. Journal of Flow Chemistry 2022, 12
(4)
, 383-387. https://doi.org/10.1007/s41981-022-00223-3
- Mikhail Krasavin, Maria Adamchik, Andrey Bubyrev, Christopher Heim, Samuel Maiwald, Daniil Zhukovsky, Petr Zhmurov, Alexander Bunev, Marcus D. Hartmann. Synthesis of novel glutarimide ligands for the E3 ligase substrate receptor Cereblon (CRBN): Investigation of their binding mode and antiproliferative effects against myeloma cell lines. European Journal of Medicinal Chemistry 2022, 327 , 114990. https://doi.org/10.1016/j.ejmech.2022.114990
- Christopher Heim, Anna-Katharina Spring, Sören Kirchgäßner, Dirk Schwarzer, Marcus D. Hartmann. Identification and structural basis of C-terminal cyclic imides as natural degrons for cereblon. Biochemical and Biophysical Research Communications 2022, 637 , 66-72. https://doi.org/10.1016/j.bbrc.2022.11.001
- Jian Wu, Emily Chu, Barry Paul, Yubin Kang. Mechanistic Studies and a Retrospective Cohort Study: The Interaction between PPAR Agonists and Immunomodulatory Agents in Multiple Myeloma. Cancers 2022, 14
(21)
, 5272. https://doi.org/10.3390/cancers14215272
- Junichi Yamamoto, Takumi Ito, Yuki Yamaguchi, Hiroshi Handa. Discovery of CRBN as a target of thalidomide: a breakthrough for progress in the development of protein degraders. Chemical Society Reviews 2022, 51
(15)
, 6234-6250. https://doi.org/10.1039/D2CS00116K
- Jaeyong Cho, Atsushi Miyagawa, Kazuki Yamaguchi, Wakana Abe, Yoji Tsugawa, Hatsuo Yamamura, Takeshi Imai. UDP-Glucose: A Cereblon-Dependent Glucokinase Protein Degrader. International Journal of Molecular Sciences 2022, 23
(16)
, 9094. https://doi.org/10.3390/ijms23169094
- S. V. Semochkin. Mechanisms of action of immunomodulatory drugs — from teratogenicity to treatment of multiple myeloma. Russian journal of hematology and transfusiology 2022, 67
(2)
, 240-260. https://doi.org/10.35754/0234-5730-2022-67-2-240-260
- Izidor Sosič, Aleša Bricelj, Christian Steinebach. E3 ligase ligand chemistries: from building blocks to protein degraders. Chemical Society Reviews 2022, 51
(9)
, 3487-3534. https://doi.org/10.1039/D2CS00148A
- Christopher Heim, Marcus D. Hartmann. High-resolution structures of the bound effectors avadomide (CC-122) and iberdomide (CC-220) highlight advantages and limitations of the MsCI4 soaking system. Acta Crystallographica Section D Structural Biology 2022, 78
(3)
, 290-298. https://doi.org/10.1107/S2059798322000092
- Alexander Kazantsev, Mikhail Krasavin. Ligands for cereblon: 2017–2021 patent overview. Expert Opinion on Therapeutic Patents 2022, 32
(2)
, 171-190. https://doi.org/10.1080/13543776.2022.1999415
- Mikhail Krasavin, Maria Adamchik, Andrey Bubyrev, Christopher Heim, Samuel Maiwald, Daniil Zhukovsky, Petr Zhmurov, Alexander Bunev, Marcus D. Hartmann. Synthesis of Novel Glutarimide Ligands for the E3 Ligase Substrate Receptor Cereblon (CRBN): Investigation of Their Binding Mode and Antiproliferative Effects Against Myeloma Cell Lines. SSRN Electronic Journal 2022, https://doi.org/10.2139/ssrn.4191806
- Zhiwen Qi, Guliang Yang, Tao Deng, Jianmin Wang, Hao Zhou, Sergey A. Popov, Elvira E. Shults, Chengzhang Wang. Design and linkage optimization of ursane-thalidomide-based PROTACs and identification of their targeted-degradation properties to MDM2 protein. Bioorganic Chemistry 2021, 111 , 104901. https://doi.org/10.1016/j.bioorg.2021.104901
- Shelton R. Boyd, Lyra Chang, Wanderson Rezende, Idris O. Raji, Prasanna Kandel, Secondra L. Holmes, Damian W. Young. Design and Applications of Bifunctional Small Molecules in Biology. Biochimica et Biophysica Acta (BBA) - Proteins and Proteomics 2021, 1869
(1)
, 140534. https://doi.org/10.1016/j.bbapap.2020.140534
- Christopher Heim, Samuel Maiwald, Christian Steinebach, Matthew K. Collins, Jonathan Strope, Cindy H. Chau, William D. Figg, Michael Gütschow, Marcus D. Hartmann. On the correlation of cereblon binding, fluorination and antiangiogenic properties of immunomodulatory drugs. Biochemical and Biophysical Research Communications 2021, 534 , 67-72. https://doi.org/10.1016/j.bbrc.2020.11.117
- Taku Shoji, Nanami Iida, Akari Yamazaki, Yukino Ariga, Akira Ohta, Ryuta Sekiguchi, Tatsuki Nagahata, Takuya Nagasawa, Shunji Ito. Synthesis of phthalimides cross-conjugated with an azulene ring, and their structural, optical and electrochemical properties. Organic & Biomolecular Chemistry 2020, 18
(12)
, 2274-2282. https://doi.org/10.1039/D0OB00164C
- Li Zhou, Wenming Chen, Chenyang Cao, Yonghui Shi, Wenchong Ye, Jiliang Hu, LingLi Wang, Wen Zhou. Design and synthesis of α-naphthoflavone chimera derivatives able to eliminate cytochrome P450 (CYP)1B1-mediated drug resistance via targeted CYP1B1 degradation. European Journal of Medicinal Chemistry 2020, 189 , 112028. https://doi.org/10.1016/j.ejmech.2019.112028
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 60 other publications.
- 1Somers, G. F. Pharmacological properties of thalidomide (alpha-phthalimido glutarimide), a new sedative hypnotic drug. Br. J. Pharmacol. Chemother. 1960, 15, 111– 116, DOI: 10.1111/j.1476-5381.1960.tb01217.x1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaF3cXht1ShtLc%253D&md5=4d82cdf61a96c7f74e79fb343f40bafdPharmacological properties of thalidomide (α-phthalimidoglutarimide), a new sedative hypnotic drugSomers, G. F.British Journal of Pharmacology and Chemotherapy (1960), 15 (), 111-16CODEN: BJPCAL; ISSN:0366-0826.This new non-toxic non-barbiturate sedative was assayed on male albino rats, mice, rabbits, cats, and guinea pigs for toxicity, effect on motor activity, reflexes, narcosis, analgesis, antipyresis, autonomic system, and gastrointestinal tract. It was virtually nontoxic, and had no effect on these systems, possibly because of limited absorption. It potentiated barbiturates, alc., reserpine, chlorpromazine, and counteracted methylamphetamine and methylphenidate.
- 2Lenz, W. A short history of thalidomide embryopathy. Teratology 1988, 38, 203– 215, DOI: 10.1002/tera.14203803032https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaL1M7ks1Kluw%253D%253D&md5=65dd1712af76e247727c4cf7f528ae51A short history of thalidomide embryopathyLenz WTeratology (1988), 38 (3), 203-15 ISSN:0040-3709.There is no expanded citation for this reference.
- 3Vargesson, N. Thalidomide-induced limb defects: resolving a 50-year-old puzzle. Bioessays 2009, 31, 1327– 1336, DOI: 10.1002/bies.2009001033https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXisVamuw%253D%253D&md5=ca4738ec2192397e2b61142a3eda4f3eThalidomide-induced limb defects: resolving a 50-year-old puzzleVargesson, NeilBioEssays (2009), 31 (12), 1327-1336CODEN: BIOEEJ; ISSN:0265-9247. (Wiley-Blackwell)A review. Despite the recent discovery that thalidomide causes limb defects by targeting highly angiogenic, immature blood vessels, several challenges still remain and new ones have arisen. These include understanding the drug's species specificity, detg. mol. target(s) in the endothelial cell, shedding light on the mol. basis of phocomelia and producing a form of the drug that is clin. effective without having side effects. Now that the trigger of thalidomide-induced teratogenesis has been uncovered, a framework is proposed, incorporating and uniting previous models of thalidomide action, explaining how thalidomide causes not just limb defects, but also all the other defects it induces.
- 4Johnston, R. E.; Abdalla, S. H. Thalidomide in low doses is effective for the treatment of resistant or relapsed multiple myeloma and for plasma cell leukaemia. Leuk. Lymphoma 2002, 43, 351– 354, DOI: 10.1080/104281902900061434https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XntVaqs74%253D&md5=fefa82138697984cbc04d8738cfb8bf9Thalidomide in low doses is effective for the treatment of resistant or relapsed multiple myeloma and for plasma cell leukemiaJohnston, R. E.; Abdalla, S. H.Leukemia & Lymphoma (2002), 43 (2), 351-354CODEN: LELYEA; ISSN:1042-8194. (Taylor & Francis Ltd.)Thalidomide is an effective treatment for relapsed multiple myeloma (MM), but is assocd. with a significant side effect profile at higher doses. In a recent study, only half of the enrolled patients were able to tolerate the max. dose of 800 mg/day [Singhal, S., et al. (1999) "Antitumor activity of thalidomide in refractory multiple myeloma", New Engl. J. Med. 341, 1565-1571]. Moreover, the dose-response relationship has not been defined. We report our use of low dose thalidomide in a small cohort of 12 patients-eight with relapsed or refractory MM and four with plasma cell leukemia (PCL). Five of the 12 (42%) patients had a partial response, showing a median fall in their PP/BJP of 80% (63-90%) at a median dose of 175 mg (100-300 mg) with negligible side effects. Three of four patients with PCL showed an impressive response to treatment with thalidomide as a single agent. No patient who failed to show any evidence of response at low dose (<150 mg/day) responded to higher doses. In this study, thalidomide induces a similar rate of response at a lower and better tolerated dose than previously reported and produced "best ever" responses in patients with resistant PCL.
- 5US Thalomid Label. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/020785s061lbl.pdf (accessed Dec 9, 2018).There is no corresponding record for this reference.
- 6Bertolini, F.; Mingrone, W.; Alietti, A.; Ferrucci, P. F.; Cocorocchio, E.; Peccatori, F.; Cineri, S.; Mancuso, P.; Corsini, C.; Burlini, A.; Zucca, E.; Martinelli, G. Thalidomide in multiple myeloma, myelodysplastic syndromes and histiocytosis. Analysis of clinical results and of surrogate angiogenesis markers. Ann. Oncol. 2001, 12, 987– 990, DOI: 10.1023/a:10111410098126https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD3MvnvVehuw%253D%253D&md5=19268541729e2a5d8762802d62ae882cThalidomide in multiple myeloma, myelodysplastic syndromes and histiocytosis. Analysis of clinical results and of surrogate angiogenesis markersBertolini F; Mingrone W; Alietti A; Ferrucci P F; Cocorocchio E; Peccatori F; Cinieri S; Mancuso P; Corsini C; Burlini A; Zucca E; Martinelli G; Cineri SAnnals of oncology : official journal of the European Society for Medical Oncology (2001), 12 (7), 987-90 ISSN:0923-7534.BACKGROUND: Thalidomide, as a single agent, has been recently found to induce a clinical response in one third of refractory or relapsed myeloma patients. Although it has been reported that thalidomide significantly inhibits angiogenesis. it is still unclear whether its clinical effect is mediated, at least in part, by its anti-angiogenic properties. PATIENTS AND METHODS: We evaluated thalidomide as a single agent in myeloma, myelodysplastic syndromes (MDS) and histiocytosis, i.e. hematological diseases characterized by increased angiogenesis, and measured prospectively a number of surrogate angiogenesis markers. RESULTS: Clinical responses were observed in 7 of 17 myeloma and 2 of 5 MDS patients. The histiocytosis patient had a partial response. At the time of the best clinical response, plasma levels of angiogenic growth factors, vascular endothelial growth factor (VEGF) and basic-fibroblast growth factor (b-FGF), were significantly decreased, and flow cytometry indicated a decrease of activated endothelial cells in the bone marrow of responding MDS patients. CONCLUSIONS: These observations confirm thalidomide efficacy in myeloma, suggest a possible use in MDS and histiocytosis and may contribute to the prediction of clinical response and to understanding the mechanism of thalidomide's action.
- 7Kale, V.; List, A. Immunomodulatory drugs (IMiDs): a new treatment option for myelodysplastic syndromes. Curr. Pharm. Biotechnol. 2006, 7, 339– 342, DOI: 10.2174/1389201067785215877https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XhtVamsLrN&md5=e1597e6b7cf4543228046753231f2348Immunomodulatory drugs (IMiDs): a new treatment option for myelodysplastic syndromesKale, Vishakha; List, Alan F.Current Pharmaceutical Biotechnology (2006), 7 (5), 339-342CODEN: CPBUBP; ISSN:1389-2010. (Bentham Science Publishers Ltd.)A review. The IMiDs represent a new proprietary class of thalidomide analogs that possess greater potency and less toxicity than the parent compd. As a group, these agents share the pharmacol. property of modulating cellular response to ligand activation, the precise biol. effect of which is cell lineage and stimulant-dependent. Lenalidomide (CC-5013; Revlimid), a second generation IMiD, has shown significant erythropoietic activity in patients with lower risk MDS that have failed or are not candidates for recombinant erythropoietin treatment. Unlike cytokine therapy, lenalidomide suppresses select MDS clones and enhances erythropoietin receptor signaling to restore erythropoiesis. Activity is greatest in patients with interstitial deletions involving chromosome 5q31.1. A multicenter phase II study reported a 76% overall transfusion response rate in transfusion-dependent patients with deletion 5q, with 67% achieving transfusion independence after a median interval of 4.6 wk of treatment. Cytogenetic responses were obsd. in 73% of patients with complete cytogenetic remission in 45% patients. Both transfusion response and cytogenetic response frequency were independent of karyotype complexity, raising excitement that this new treatment strategy might favorably alter the natural history of disease in higher risk patients with deletion 5q. Lenalidomide was approved by the U.S. Food and Drug Administration on Dec. 27, 2005, for the treatment of IPSS Low and intermediate-1 risk MDS patients with del(5q) abnormality. A phase III Intergroup trial (ECOG 2905) will test the capacity to potentiate erythropoietin response by comparing response to lenalidomide monotherapy to the combination of darbepoetin and lenalidomide in non-deletion 5q MDS patients.
- 8Wu, H.; Zhao, C.; Gu, K.; Jiao, Y.; Hao, J.; Sun, G. Thalidomide plus chemotherapy exhibit enhanced efficacy in the clinical treatment of T-cell non-Hodgkin’s lymphoma: A prospective study of 46 cases. Mol. Clin. Oncol. 2014, 2, 695– 700, DOI: 10.3892/mco.2014.3078https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2sbpvFKgtA%253D%253D&md5=f173c9f716db0820c32424792125ff88Thalidomide plus chemotherapy exhibit enhanced efficacy in the clinical treatment of T-cell non-Hodgkin's lymphoma: A prospective study of 46 casesWu Hongyang; Zhao Chenchen; Gu Kangsheng; Jiao Yang; Hao Jiqing; Sun GuopingMolecular and clinical oncology (2014), 2 (5), 695-700 ISSN:2049-9450.The treatment of T-cell non-Hodgkin's lymphoma (T-NHL) remains challenging. There is currently no standard regimen for the treatment of T-NHL in the first- or second-line setting. Thalidomide was previously shown to exert antitumor effects through inhibiting angiogenesis, promoting apoptosis and immunomodulatory activity. However, all the previous studies on the treatment of lymphoma with thalidomide included patient samples of limited size. In the present study, 46 cases of eligible T-NHL patients were randomized into i) the control group (conventional combined chemotherapy, n=22) and ii) the thalidomide group (thalidomide plus combined chemotherapy, n=24). The median dose of thalidomide was 200 mg (range, 150-400 mg) every night, without reported severe side effects. The clinical response to treatment was as follows: Complete response (CR) in 12 cases, partial response (PR) in 7, stable disease (SD) in 1 and progressive disease (PD) in 4 cases in the thalidomide group; and CR in 8 cases, PR in 6, SD in 3 and PD in 5 cases in the control group. The CR rate was 50.0 and 36.4% in the thalidomide and the control groups, respectively (P<0.05). The median progression-free and overall survival were 12 and undefined months, respectively, in the thalidomide group and 6 and 17 months, respectively, in the control group. The toxicity profile was considered acceptable in both groups. Our results indicated that thalidomide plus combined chemotherapy may exhibit enhanced efficacy in the clinical treatment of T-NHL. In addition, this type of treatment may reduce the frequency of adverse gastrointestinal reactions and help alleviate fear of chemotherapy. Therefore, thalidomide plus combined chemotherapy may be a viable option for the clinical treatment of T-NHL.
- 9García-Sanz, R.; González-López, T. J.; Vázquez, L.; Hermida, G.; Graciani, I. F.; San Miguel, J. F. The combination of thalidomide, cyclophosphamide and dexamethasone is potentially useful in highly resistant Hodgkin’s lymphoma. Eur. J. Haematol. 2010, 84, 266– 270, DOI: 10.1111/j.1600-0609.2009.01375.x9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXjtlGmtL0%253D&md5=11de756fa308a935acb76c8870c33110The combination of thalidomide, cyclophosphamide and dexamethasone is potentially useful in highly resistant Hodgkin's lymphomaGarcia-Sanz, R.; Gonzalez-Lopez, T. J.; Vazquez, L.; Hermida, G.; Graciani, I. F.; San Miguel, J. F.European Journal of Haematology (2010), 84 (3), 266-270CODEN: EJHAEC; ISSN:0902-4441. (Wiley-Blackwell)Few diseases have a prognosis worse than Hodgkin's lymphoma (HL), patients relapsing after autologous or allogeneic stem cell transplantation. Here, we report two highly refractory patients with HL who successfully responded to a combination of thalidomide, cyclophosphamide and dexamethasone (ThaCyDex). Despite the use of a very large no. of different drugs (>5 different schemes) including high-dose therapy and autologous and allogeneic stem cell transplantation, both patients proved to be suffering from a highly resistant disease. Fortunately, they finally responded to the ThaCyDex combination, achieving sustained complete remission that would support the running of a trial within this setting.
- 10Kuruvilla, J.; Song, K.; Mollee, P.; Panzarella, T.; McCrae, J.; Nagy, T.; Crump, M.; Keating, A. A phase II study of thalidomide and vinblastine for palliative patients with Hodgkin’s lymphoma. Hematology 2006, 11, 25– 29, DOI: 10.1080/1024533050027659210https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XitVOhu74%253D&md5=7e23a9439d19a069125052db1c1c64b7A phase II study of thalidomide and vinblastine for palliative patients with Hodgkin's lymphomaKuruvilla, John; Song, Kevin; Mollee, Peter; Panzarella, Tony; McCrae, Jan; Nagy, Tracy; Crump, Michael; Keating, ArmandHematology (Abingdon, United Kingdom) (2006), 11 (1), 25-29CODEN: HMATFL; ISSN:1024-5332. (Taylor & Francis Ltd.)Introduction: Patients with Hodgkin's Lymphoma (HL) who relapse or progress after primary therapy and subsequent high dose chemotherapy with autologous stem cell transplantation (ASCT) cannot be cured with conventional treatment. We combined thalidomide (THAL), an agent with anti-angiogenic and immunomodulatory properties, with vinblastine, which is active after ASCT, to det. the objective response rate, improvement in B symptoms and toxicity in patients with refractory HD. Methods: Patients were eligible if they HD that progressed after chemotherapy and ASCT or had declined or were ineligible for curative therapy. Treatment consisted of THAL 200 mg orally given daily. After 2 wk, VBL 6 mg IV was given weekly × 6 doses on an eight-week cycle. Response and toxicity assessment occurred following each cycle. Results: Eleven patients were enrolled, 1 progressed within 6 days of study enrollment and was subsequently treated with alternative palliative therapy and thus 11 patients are response evaluable and 10 are evaluable for toxicity. Patient characteristics: relapsed after ASCT: 7; median no. of prior chemotherapy regimens: 3 (range 1-5); median time to progression post-ASCT: 7 mo (range 2-29). Four patients had a partial response to treatment (response rate 36%); two patients had stable disease. B symptoms were present at enrollment in four patients and resolved completely on treatment in two patients. Five had disease progression within 3 mo of starting treatment. The median duration of response was 9 mo (range 0-22 mo). Toxicity was mild and limited to grade 2 neuropathy in 6 patients and grade 2 or 3 neutropenia in 4 patients. Conclusions: In this small study in chemotherapy- refractory HL, THAL and VBL demonstrated encouraging activity with some durable responses and acceptable toxicity. These results suggest that chronic low dose chemotherapy combined with less toxic immunomodulatory or anti-angiogenic drugs warrants further study.
- 11Seldin, D. C.; Choufani, E. B.; Dember, L. M.; Wiesman, J. F.; Berk, J. L.; Falk, R. H.; O’Hara, C.; Fennessey, S.; Finn, K. T.; Wright, D. G.; Skinner, M.; Sanchorawala, V. Tolerability and efficacy of thalidomide for the treatment of patients with light chain-associated (AL) amyloidosis. Clin. Lymphoma 2003, 3, 241– 246, DOI: 10.3816/clm.2003.n.00511https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXjtVSjtrk%253D&md5=b15425f1d8d306ab1f9bb1375a49e40bTolerability and efficacy of thalidomide for the treatment of patients with light-chain-associated (AL) amyloidosisSeldin, David C.; Choufani, Elie B.; Dember, Laura M.; Wiesman, Janice F.; Berk, John L.; Falk, Rodney H.; O'Hara, Carl; Fennessey, Salli; Finn, Kathleen T.; Wright, Daniel G.; Skinner, Martha; Sanchorawala, VaishaliClinical Lymphoma (2003), 3 (4), 241-246CODEN: CLLYAO; ISSN:1526-9655. (Cancer Information Group)A phase I/II trial was carried out with patients with AL amyloidosis, most of whom had failed prior therapy with high-dose melphalan and autologous stem cell transplantation. This trial was designed as an individualized 6-mo dose-escalation study with re-evaluation of bone marrow plasmacytosis and serum and urine monoclonal proteins after 3 and 6 mo. Sixteen patients with a median age of 62 yr (range, 37-70 yr) were enrolled. Fourteen of the patients had renal involvement, 4 had cardiac involvement, 4 had liver involvement, and 2 had predominant soft tissue or lymph node involvement. The median max. tolerated dose was 300 mg, with fatigue and other central nervous system side effects being the major dose-limiting toxicities. Side effects not frequently reported for other patient populations included exacerbation of peripheral and pulmonary edema and worsening azotemia. In all, 50% of the patients experienced grade 3/4 toxicity, and 25% had to discontinue the drug. No complete hematol. responses were seen, but 25% of the patients had a significant redn. in Bence-Jones proteinuria. Thus, while thalidomide has activity in AL amyloidosis, it also has significant toxicity in this patient population.
- 12Baird, R.; van Zyl-Smit, R. N.; Iveson, A.; Duddy, J.; Rassam, S. M. Thalidomide is highly effective in a patient with meningeal acute myeloid leukaemia. Leuk. Lymphoma 2004, 45, 179– 181, DOI: 10.1080/104281903100014941212https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD2c7mslehtw%253D%253D&md5=31006e0dbe0fb3cf55bcd7198fc8ef2aThalidomide is highly effective in a patient with meningeal acute myeloid leukaemiaBaird Richard; van Zyl-Smit Richard Nellis; Iveson Anne; Duddy James; Rassam Saad M BLeukemia & lymphoma (2004), 45 (1), 179-81 ISSN:1042-8194.We report a case of secondary acute myeloid leukaemia (AML) following high dose therapy for diffuse large B-cell non-Hodgkin's lymphoma (NHL) who developed meningeal leukaemia. This was refractory to systemic and intrathecal chemotherapy and cranial irradiation. Thalidomide has been reported to have anti-AML activity and appears to cross the blood brain barrier (BBB). We, therefore, attempted a trial of oral Thalidomide and achieved rapid biochemical and cytological remission with a short course. The patient, however, progressed systemically and succumbed to her illness.
- 13Thomas, D. A.; Estey, E.; Giles, F. J.; Faderl, S.; Cortes, J.; Keating, M.; O’Brien, S.; Albitar, M.; Kantarjian, H. Single agent thalidomide in patients with relapsed or refractory acute myeloid leukaemia. Br. J. Haematol. 2003, 123, 436– 441, DOI: 10.1046/j.1365-2141.2003.04639.x13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXpsFChs70%253D&md5=28de5d4f924d2b35a000096ceb1f5be7Single agent thalidomide in patients with relapsed or refractory acute myeloid leukaemiaThomas, Deborah A.; Estey, Elihu; Giles, Francis J.; Faderi, Stefan; Cortes, Jorge; Keating, Michael; O'Brien, Susan; Albitar, Maher; Kantarjian, HagopBritish Journal of Haematology (2003), 123 (3), 436-441CODEN: BJHEAL; ISSN:0007-1048. (Blackwell Publishing Ltd.)Thalidomide is a putative anti-angiogenesis agent that has significant anti-tumor activity in haematol. malignancies with increased bone marrow angiogenesis, including multiple myeloma (MM) and myelodysplastic syndromes (MDS). Increased levels of the mitogen for angiogenesis, vascular endothelial growth factor (VEGF), correlate with worse survival in acute myeloid leukemia (AML). A phase II trial of thalidomide was conducted in patients with relapsed- or refractory-AML previously treated with cytarabine-contg. regimens. A total of 16 patients with refractory- or relapsed-AML were treated with thalidomide 200-800 mg orally daily (median dose 400 mg daily) for a median of 27 d (range, 3-94 d). Overall, one patient (6%) achieved complete remission (CR) lasting for 36 mo, and two patients had a transient redn. in marrow blasts from 8% and 7% to less than 5% in both cases. There was no correlation between redn. in levels of angiogenesis markers and response. Toxicities related to thalidomide were significant, and precluded dose escalation beyond 400 mg orally daily in most patients. Although there appears to be some evidence of biol. activity, single agent thalidomide is not an optimal choice of therapy for salvaging patients with relapsed- or refractory-AML. Thalidomide analogs with more potent immunomodulatory activities and more favorable toxicity profiles may offer more promise as anti-AML therapy.
- 14Hartmann, M. D.; Boichenko, I.; Coles, M.; Zanini, F.; Lupas, A. N.; Hernandez Alvarez, B. Thalidomide mimics uridine binding to an aromatic cage in cereblon. J. Struct. Biol. 2014, 188, 225– 232, DOI: 10.1016/j.jsb.2014.10.01014https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhvFSnu7jI&md5=e601c6e1091deea840f9738fb30fa7bbThalidomide mimics uridine binding to an aromatic cage in cereblonHartmann, Marcus D.; Boichenko, Iuliia; Coles, Murray; Zanini, Fabio; Lupas, Andrei N.; Hernandez Alvarez, BirteJournal of Structural Biology (2014), 188 (3), 225-232CODEN: JSBIEM; ISSN:1047-8477. (Elsevier Inc.)Thalidomide and its derivs. lenalidomide and pomalidomide are important anticancer agents but can cause severe birth defects via an interaction with the protein cereblon. The ligand-binding domain of cereblon is found, with a high degree of conservation, in both bacteria and eukaryotes. Using a bacterial model system, we reveal the structural determinants of cereblon substrate recognition, based on a series of high-resoln. crystal structures. For the first time, we identify a cellular ligand that is universally present: we show that thalidomide and its derivs. mimic and compete for the binding of uridine, and validate these findings in vivo. The nature of the binding pocket, an arom. cage of three tryptophan residues, further suggests a role in the recognition of cationic ligands. Our results allow for general evaluation of pharmaceuticals for potential cereblon-dependent teratogenicity.
- 15Hartmann, M. D.; Boichenko, I.; Coles, M.; Lupas, A. N.; Hernandez Alvarez, B. Structural dynamics of the cereblon ligand binding domain. PLoS One 2015, 10, e0128342 DOI: 10.1371/journal.pone.012834215https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhslGru77F&md5=525f48f454c54c466360231d1ab3330aStructural dynamics of the cereblon ligand binding domainHartmann, Marcus D.; Boichenko, Iuliia; Coles, Murray; Lupas, Andrei N.; Alvarez, Birte HernandezPLoS One (2015), 10 (5), e0128342/1-e0128342/16CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)Cereblon, a primary target of thalidomide and its derivs., has been characterized structurally from both bacteria and animals. Esp. well studied is the thalidomide binding domain, CULT, which shows an invariable structure across different organisms and in complex with different ligands. Here, based on a series of crystal structures of a bacterial representative, we reveal the conformational flexibility and structural dynamics of this domain. In particular, we follow the unfolding of large fractions of the domain upon release of thalidomide in the cryst. state. Our results imply that a third of the domain, including the thalidomide binding pocket, only folds upon ligand binding. We further characterize the structural effect of the C-terminal truncation resulting from the mental-retardation linked R419X nonsense mutation in vitro and offer a mechanistic hypothesis for its irresponsiveness to thalidomide. At 1.2Å resoln., our data provide a view of thalidomide binding at at. resoln.
- 16Chamberlain, P. P.; Lopez-Girona, A.; Miller, K.; Carmel, G.; Pagarigan, B.; Chie-Leon, B.; Rychak, E.; Corral, L. G.; Ren, Y. J.; Wang, M.; Riley, M.; Delker, S. L.; Ito, T.; Ando, H.; Mori, T.; Hirano, Y.; Handa, H.; Hakoshima, T.; Daniel, T. O.; Cathers, B. E. Structure of the human cereblon-DDB1-lenalidomide complex reveals basis for responsiveness to thalidomide analogs. Nat. Struct. Mol. Biol. 2014, 21, 803– 809, DOI: 10.1038/nsmb.287416https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtlahurjI&md5=627fc3c8eae320059ca1cdc173bb56edStructure of the human Cereblon-DDB1-lenalidomide complex reveals basis for responsiveness to thalidomide analogsChamberlain, Philip P.; Lopez-Girona, Antonia; Miller, Karen; Carmel, Gilles; Pagarigan, Barbra; Chie-Leon, Barbara; Rychak, Emily; Corral, Laura G.; Ren, Yan J.; Wang, Maria; Riley, Mariko; Delker, Silvia L.; Ito, Takumi; Ando, Hideki; Mori, Tomoyuki; Hirano, Yoshinori; Handa, Hiroshi; Hakoshima, Toshio; Daniel, Thomas O.; Cathers, Brian E.Nature Structural & Molecular Biology (2014), 21 (9), 803-809CODEN: NSMBCU; ISSN:1545-9993. (Nature Publishing Group)The Cul4-Rbx1-DDB1-Cereblon E3 ubiquitin ligase complex is the target of thalidomide, lenalidomide and pomalidomide, therapeutically important drugs for multiple myeloma and other B-cell malignancies. These drugs directly bind Cereblon (CRBN) and promote the recruitment of substrates Ikaros (IKZF1) and Aiolos (IKZF3) to the E3 complex, thus leading to substrate ubiquitination and degrdn. Here we present the crystal structure of human CRBN bound to DDB1 and the drug lenalidomide. A hydrophobic pocket in the thalidomide-binding domain (TBD) of CRBN accommodates the glutarimide moiety of lenalidomide, whereas the isoindolinone ring is exposed to solvent. We also solved the structures of the mouse TBD in the apo state and with thalidomide or pomalidomide. Site-directed mutagenesis in lentiviral-expression myeloma models showed that key drug-binding residues are crit. for antiproliferative effects.
- 17Fischer, E. S.; Böhm, K.; Lydeard, J. R.; Yang, H.; Stadler, M. B.; Cavadini, S.; Nagel, J.; Serluca, F.; Acker, V.; Lingaraju, G. M.; Tichkule, R. B.; Schebesta, M.; Forrester, W. C.; Schirle, M.; Hassiepen, U.; Ottl, J.; Hild, M.; Beckwith, R. E. J.; Harper, J. W.; Jenkins, J. L.; Thomä, N. H. Structure of the DDB1-CRBN E3 ubiquitin ligase in complex with thalidomide. Nature 2014, 512, 49– 53, DOI: 10.1038/nature1352717https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXht12ms7%252FP&md5=1c1eaa48bef87463cdd3f2b1e1bd400dStructure of the DDB1-CRBN E3 ubiquitin ligase in complex with thalidomideFischer, Eric S.; Bohm, Kerstin; Lydeard, John R.; Yang, Haidi; Stadler, Michael B.; Cavadini, Simone; Nagel, Jane; Serluca, Fabrizio; Acker, Vincent; Lingaraju, Gondichatnahalli M.; Tichkule, Ritesh B.; Schebesta, Michael; Forrester, William C.; Schirle, Markus; Hassiepen, Ulrich; Ottl, Johannes; Hild, Marc; Beckwith, Rohan E. J.; Harper, J. Wade; Jenkins, Jeremy L.; Thoma, Nicolas H.Nature (London, United Kingdom) (2014), 512 (7512), 49-53CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)In the 1950s, the drug thalidomide, administered as a sedative to pregnant women, led to the birth of thousands of children with multiple defects. Despite the teratogenicity of thalidomide and its derivs. lenalidomide and pomalidomide, these immunomodulatory drugs (IMiDs) recently emerged as effective treatments for multiple myeloma and 5q-deletion-assocd. dysplasia. IMiDs target the E3 ubiquitin ligase CUL4-RBX1-DDB1-CRBN (known as CRL4CRBN) and promote the ubiquitination of the IKAROS family transcription factors IKZF1 and IKZF3 by CRL4CRBN. Here we present crystal structures of the DDB1-CRBN complex bound to thalidomide, lenalidomide and pomalidomide. The structure establishes that CRBN is a substrate receptor within CRL4CRBN and enantioselectively binds IMiDs. Using an unbiased screen, we identified the homeobox transcription factor MEIS2 as an endogenous substrate of CRL4CRBN. Our studies suggest that IMiDs block endogenous substrates (MEIS2) from binding to CRL4CRBN while the ligase complex is recruiting IKZF1 or IKZF3 for degrdn. This dual activity implies that small mols. can modulate an E3 ubiquitin ligase and thereby upregulate or downregulate the ubiquitination of proteins.
- 18Nguyen, T. V.; Lee, J. E.; Sweredoski, M. J.; Yang, S. J.; Jeon, S. J.; Harrison, J. S.; Yim, J. H.; Lee, S. G.; Handa, H.; Kuhlman, B.; Jeong, J. S.; Reitsma, J. M.; Park, C. S.; Hess, S.; Deshaies, R. J. Glutamine triggers acetylation-dependent degradation of glutamine synthetase via the thalidomide receptor cereblon. Mol. Cell 2016, 61, 809– 820, DOI: 10.1016/j.molcel.2016.02.03218https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xks1OqsL0%253D&md5=641527ec1a6c97d1800e3e393582c07fGlutamine Triggers Acetylation-Dependent Degradation of Glutamine Synthetase via the Thalidomide Receptor CereblonNguyen, T. Van; Lee, J. Eugene; Sweredoski, Michael J.; Yang, Seung-Joo; Jeon, Seung-Je; Harrison, Joseph S.; Yim, Jung-Hyuk; Lee, Sang Ghil; Handa, Hiroshi; Kuhlman, Brian; Jeong, Ji-Seon; Reitsma, Justin M.; Park, Chul-Seung; Hess, Sonja; Deshaies, Raymond J.Molecular Cell (2016), 61 (6), 809-820CODEN: MOCEFL; ISSN:1097-2765. (Elsevier Inc.)Cereblon (CRBN), a substrate receptor for the cullin-RING ubiquitin ligase 4 (CRL4) complex, is a direct protein target for thalidomide teratogenicity and antitumor activity of immunomodulatory drugs (IMiDs). Here we report that glutamine synthetase (GS) is an endogenous substrate of CRL4CRBN. Upon exposing cells to high glutamine concn., GS is acetylated at lysines 11 and 14, yielding a degron that is necessary and sufficient for binding and ubiquitylation by CRL4CRBN and degrdn. by the proteasome. Binding of acetylated degron peptides to CRBN depends on an intact thalidomide-binding pocket but is not competitive with IMiDs. These findings reveal a feedback loop involving CRL4CRBN that adjusts GS protein levels in response to glutamine and uncover a new function for lysine acetylation.
- 19Del Prete, D.; Rice, R. C.; Rajadhyaksha, A. M.; D’Adamio, L. Amyloid precursor protein (APP) may act as a substrate and a recognition unit for CRL4(CRBN) and Stub1 E3 ligases facilitating ubiquitination of proteins involved in presynaptic functions and neurodegeneration. J. Biol. Chem. 2016, 291, 17209– 17227, DOI: 10.1074/jbc.m116.73362619https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhslaktLrF&md5=b4cfc53ded92c62aa868a17e28929986Amyloid precursor protein (APP) may act as a substrate and a recognition unit for CRL4CRBN and Stub1 E3 ligases facilitating ubiquitination of proteins involved in presynaptic functions and neurodegenerationDel Prete, Dolores; Rice, Richard C.; Rajadhyaksha, Anjali M.; D'Adamio, LucianoJournal of Biological Chemistry (2016), 291 (33), 17209-17227CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)The amyloid precursor protein (APP), whose mutations cause Alzheimer disease, plays an important in vivo role and facilitates transmitter release. Because the APP cytosolic region (ACR) is essential for these functions, we have characterized its brain interactome. We found that the ACR interacts with proteins that regulate the ubiquitin-proteasome system, predominantly with the E3 ubiquitin-protein ligases Stub1, which binds the N-terminus of the ACR, and CRL4CRBN, which is formed by Cul4a/b, Ddb1, and Crbn, and interacts with the C-terminus of the ACR via Crbn. APP shares essential functions with APP-like protein-2 (APLP2) but not APP-like protein-1 (APLP1). Noteworthy, APLP2, but not APLP1, interacts with Stub1 and CRL4CRBN, pointing to a functional pathway shared only by APP and APLP2. In vitro ubiquitination/ubiquitome anal. indicates that these E3 ligases are enzymically active and ubiquitinated the ACR residues Lys649/650/651/676/688. Deletion of Crbn reduces ubiquitination of Lys676 suggesting that Lys676 is physiol. ubiquitinated by CRL4CRBN. The ACR facilitated in vitro ubiquitination of presynaptic proteins that regulate exocytosis, suggesting a mechanism by which APP tunes transmitter release. Other dementia-related proteins, namely Tau and apoE, interact with and are ubiquitinated via the ACR in vitro. This, and the evidence that CRBN and CUL4B are linked to intellectual disability, prompts us to hypothesize a pathogenic mechanism, in which APP acts as a modulator of E3 ubiquitin-protein ligase(s), shared by distinct neuronal disorders. The well described accumulation of ubiquitinated protein inclusions in neurodegenerative diseases and the link between the ubiquitin-proteasome system and neurodegeneration make this concept plausible.
- 20Kronke, J.; Udeshi, N. D.; Narla, A.; Grauman, P.; Hurst, S. N.; McConkey, M.; Svinkina, T.; Heckl, D.; Comer, E.; Li, X.; Ciarlo, C.; Hartman, E.; Munshi, N.; Schenone, M.; Schreiber, S. L.; Carr, S. A.; Ebert, B. L. Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells. Science 2014, 343, 301– 305, DOI: 10.1126/science.124485120https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2c3itFajsQ%253D%253D&md5=8cae3e554399308493aa38f0d05ed610Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cellsKronke Jan; Udeshi Namrata D; Narla Anupama; Grauman Peter; Hurst Slater N; McConkey Marie; Svinkina Tanya; Heckl Dirk; Comer Eamon; Li Xiaoyu; Ciarlo Christie; Hartman Emily; Munshi Nikhil; Schenone Monica; Schreiber Stuart L; Carr Steven A; Ebert Benjamin LScience (New York, N.Y.) (2014), 343 (6168), 301-5 ISSN:.Lenalidomide is a drug with clinical efficacy in multiple myeloma and other B cell neoplasms, but its mechanism of action is unknown. Using quantitative proteomics, we found that lenalidomide causes selective ubiquitination and degradation of two lymphoid transcription factors, IKZF1 and IKZF3, by the CRBN-CRL4 ubiquitin ligase. IKZF1 and IKZF3 are essential transcription factors in multiple myeloma. A single amino acid substitution of IKZF3 conferred resistance to lenalidomide-induced degradation and rescued lenalidomide-induced inhibition of cell growth. Similarly, we found that lenalidomide-induced interleukin-2 production in T cells is due to depletion of IKZF1 and IKZF3. These findings reveal a previously unknown mechanism of action for a therapeutic agent: alteration of the activity of an E3 ubiquitin ligase, leading to selective degradation of specific targets.
- 21Krönke, J.; Fink, E. C.; Hollenbach, P. W.; MacBeth, K. J.; Hurst, S. N.; Udeshi, N. D.; Chamberlain, P. P.; Mani, D. R.; Man, H. W.; Gandhi, A. K.; Svinkina, T.; Schneider, R. K.; McConkey, M.; Järås, M.; Griffiths, E.; Wetzler, M.; Bullinger, L.; Cathers, B. E.; Carr, S. A.; Chopra, R.; Ebert, B. L. Lenalidomide induces ubiquitination and degradation of CK1alpha in del(5q) MDS. Nature 2015, 523, 183– 188, DOI: 10.1038/nature1461021https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXht1entr%252FN&md5=9b20a91c13e888e6569223ffa25af2cdLenalidomide induces ubiquitination and degradation of CK1α in del(5q) MDSKronke, Jan; Fink, Emma C.; Hollenbach, Paul W.; MacBeth, Kyle J.; Hurst, Slater N.; Udeshi, Namrata D.; Chamberlain, Philip P.; Mani, D. R.; Man, Hon Wah; Gandhi, Anita K.; Svinkina, Tanya; Schneider, Rebekka K.; McConkey, Marie; Jaras, Marcus; Griffiths, Elizabeth; Wetzler, Meir; Bullinger, Lars; Cathers, Brian E.; Carr, Steven A.; Chopra, Rajesh; Ebert, Benjamin L.Nature (London, United Kingdom) (2015), 523 (7559), 183-188CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Lenalidomide is a highly effective treatment for myelodysplastic syndrome (MDS) with deletion of chromosome 5q (del(5q)). Lenalidomide induces the ubiquitination of casein kinase 1A1 (CK1α) by the E3 ubiquitin ligase CUL4-RBX1-DDB1-CRBN (known as CRL4CRBN), resulting in CK1α degrdn. CK1α is encoded by a gene within the common deleted region for del(5q) MDS and haplo-insufficient expression sensitizes cells to lenalidomide therapy, providing a mechanistic basis for the therapeutic window of lenalidomide in del(5q) MDS. The authors found that mouse cells are resistant to lenalidomide but that changing a single amino acid in mouse Crbn to the corresponding human residue enables lenalidomide-dependent degrdn. of CK1α. The authors further demonstrate that minor side chain modifications in thalidomide and a novel analog, CC-122, can modulate the spectrum of substrates targeted by CRL4CRBN. These findings have implications for the clin. activity of lenalidomide and related compds., and demonstrate the therapeutic potential of novel modulators of E3 ubiquitin ligases.
- 22Matyskiela, M. E.; Lu, G.; Ito, T.; Pagarigan, B.; Lu, C.-C.; Miller, K.; Fang, W.; Wang, N.-Y.; Nguyen, D.; Houston, J.; Carmel, G.; Tran, T.; Riley, M.; Nosaka, L. A.; Lander, G. C.; Gaidarova, S.; Xu, S.; Ruchelman, A. L.; Handa, H.; Carmichael, J.; Daniel, T. O.; Cathers, B. E.; Lopez-Girona, A.; Chamberlain, P. P. A novel cereblon modulator recruits GSPT1 to the CRL4(CRBN) ubiquitin ligase. Nature 2016, 535, 252– 257, DOI: 10.1038/nature1861122https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtVCltbnI&md5=1d4e340416aa0b08fc0f123faf638c3fA novel cereblon modulator recruits GSPT1 to the CRL4CRBN ubiquitin ligaseMatyskiela, Mary E.; Lu, Gang; Ito, Takumi; Pagarigan, Barbra; Lu, Chin-Chun; Miller, Karen; Fang, Wei; Wang, Nai-Yu; Nguyen, Derek; Houston, Jack; Carmel, Gilles; Tran, Tam; Riley, Mariko; Nosaka, Lyn'Al; Lander, Gabriel C.; Gaidarova, Svetlana; Xu, Shuichan; Ruchelman, Alexander L.; Handa, Hiroshi; Carmichael, James; Daniel, Thomas O.; Cathers, Brian E.; Lopez-Girona, Antonia; Chamberlain, Philip P.Nature (London, United Kingdom) (2016), 535 (7611), 252-257CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Immunomodulatory drugs bind to cereblon (CRBN) to confer differentiated substrate specificity on the CRL4CRBN E3 ubiquitin ligase. Here we report the identification of a new cereblon modulator, CC-885, with potent anti-tumor activity. The anti-tumor activity of CC-885 is mediated through the cereblon-dependent ubiquitination and degrdn. of the translation termination factor GSPT1. Patient-derived acute myeloid leukemia tumor cells exhibit high sensitivity to CC-885, indicating the clin. potential of this mechanism. Crystallog. studies of the CRBN-DDB1-CC-885-GSPT1 complex reveal that GSPT1 binds to cereblon through a surface turn contg. a glycine residue at a key position, interacting with both CC-885 and a 'hotspot' on the cereblon surface. Although GSPT1 possesses no obvious structural, sequence or functional homol. to previously known cereblon substrates, mutational anal. and modeling indicate that the cereblon substrate Ikaros uses a similar structural feature to bind cereblon, suggesting a common motif for substrate recruitment. These findings define a structural degron underlying cereblon 'neosubstrate' selectivity, and identify an anti-tumor target rendered druggable by cereblon modulation.
- 23Matyskiela, M. E.; Couto, S.; Zheng, X.; Lu, G.; Hui, J.; Stamp, K.; Drew, C.; Ren, Y.; Wang, M.; Carpenter, A.; Lee, C.-W.; Clayton, T.; Fang, W.; Lu, C.-C.; Riley, M.; Abdubek, P.; Blease, K.; Hartke, J.; Kumar, G.; Vessey, R.; Rolfe, M.; Hamann, L. G.; Chamberlain, P. P. SALL4 mediates teratogenicity as a thalidomide-dependent cereblon substrate. Nat. Chem. Biol. 2018, 14, 981– 987, DOI: 10.1038/s41589-018-0129-x23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhs1Kgu7nM&md5=7530c3850ed7a763b30cba44dab640acSALL4 mediates teratogenicity as a thalidomide-dependent cereblon substrateMatyskiela, Mary E.; Couto, Suzana; Zheng, Xinde; Lu, Gang; Hui, Julia; Stamp, Katie; Drew, Clifton; Ren, Yan; Wang, Maria; Carpenter, Aaron; Lee, Chung-Wein; Clayton, Thomas; Fang, Wei; Lu, Chin-Chun; Riley, Mariko; Abdubek, Polat; Blease, Kate; Hartke, James; Kumar, Gondi; Vessey, Rupert; Rolfe, Mark; Hamann, Lawrence G.; Chamberlain, Philip P.Nature Chemical Biology (2018), 14 (10), 981-987CODEN: NCBABT; ISSN:1552-4450. (Nature Research)Targeted protein degrdn. via small-mol. modulation of cereblon offers vast potential for the development of new therapeutics. Cereblon-binding therapeutics carry the safety risks of thalidomide, which caused an epidemic of severe birth defects characterized by forelimb shortening or phocomelia. Here we show that thalidomide is not teratogenic in transgenic mice expressing human cereblon, indicating that binding to cereblon is not sufficient to cause birth defects. Instead, we identify SALL4 as a thalidomide-dependent cereblon neosubstrate. Human mutations in SALL4 cause Duane-radial ray, IVIC, and acro-renal-ocular syndromes with overlapping clin. presentations to thalidomide embryopathy, including phocomelia. SALL4 is degraded in rabbits but not in resistant organisms such as mice because of SALL4 sequence variations. This work expands the scope of cereblon neosubstrate activity within the formerly 'undruggable' C2H2 zinc finger family and offers a path toward safer therapeutics through an improved understanding of the mol. basis of thalidomide-induced teratogenicity.
- 24Donovan, K. A.; An, J.; Nowak, R. P.; Yuan, J. C.; Fink, E. C.; Berry, B. C.; Ebert, B. L.; Fischer, E. S., Thalidomide promotes degradation of SALL4, a transcription factor implicated in Duane Radial Ray Syndrome. Elife 2018, 7. DOI: 10.7554/elife.38430There is no corresponding record for this reference.
- 25Petzold, G.; Fischer, E. S.; Thomä, N. H. Structural basis of lenalidomide-induced CK1alpha degradation by the CRL4(CRBN) ubiquitin ligase. Nature 2016, 532, 127– 130, DOI: 10.1038/nature1697925https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XjtFOis78%253D&md5=320fc6ff811da335fa8e02feab2d702dStructural basis of lenalidomide-induced CK1α degradation by the CRL4CRBN ubiquitin ligasePetzold, Georg; Fischer, Eric S.; Thoma, Nicolas H.Nature (London, United Kingdom) (2016), 532 (7597), 127-130CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Thalidomide and its derivs., lenalidomide and pomalidomide, are immune modulatory drugs (IMiDs) used in the treatment of hematol. malignancies. IMiDs bind CRBN, the substrate receptor of the CUL4-RBX1-DDB1-CRBN (also known as CRL4CRBN) E3 ubiquitin ligase, and inhibit ubiquitination of endogenous CRL4CRBN substrates. Unexpectedly, IMiDs also repurpose the ligase to target new proteins for degrdn. Lenalidomide induces degrdn. of the lymphoid transcription factors Ikaros and Aiolos (also known as IKZF1 and IKZF3), and casein kinase 1α (CK1α), which contributes to its clin. efficacy in the treatment of multiple myeloma and 5q-deletion assocd. myelodysplastic syndrome (del(5q) MDS), resp. How lenalidomide alters the specificity of the ligase to degrade these proteins remains elusive. Here we present the 2.45 Å crystal structure of DDB1-CRBN bound to lenalidomide and CK1α. CRBN and lenalidomide jointly provide the binding interface for a CK1α β-hairpin-loop located in the kinase N-lobe. We show that CK1α binding to CRL4CRBN is strictly dependent on the presence of an IMiD. Binding of IKZF1 to CRBN similarly requires the compd. and both, IKZF1 and CK1α, use a related binding mode. Our study provides a mechanistic explanation for the selective efficacy of lenalidomide in del(5q) MDS therapy. We anticipate that high-affinity protein-protein interactions induced by small mols. will provide opportunities for drug development, particularly for targeted protein degrdn.
- 26Sievers, Q. L.; Petzold, G.; Bunker, R. D.; Renneville, A.; Słabicki, M.; Liddicoat, B. J.; Abdulrahman, W.; Mikkelsen, T.; Ebert, B. L.; Thomä, N. H. Defining the human C2H2 zinc finger degrome targeted by thalidomide analogs through CRBN. Science 2018, 362, eaat0572 DOI: 10.1126/science.aat0572There is no corresponding record for this reference.
- 27Lai, A. C.; Crews, C. M. Induced protein degradation: an emerging drug discovery paradigm. Nat. Rev. Drug Discovery 2017, 16, 101– 114, DOI: 10.1038/nrd.2016.21127https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhvFCit7rP&md5=a01ff43e9b696fe4dc03f0815158a98fInduced protein degradation: an emerging drug discovery paradigmLai, Ashton C.; Crews, Craig M.Nature Reviews Drug Discovery (2017), 16 (2), 101-114CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)Small-mol. drug discovery has traditionally focused on occupancy of a binding site that directly affects protein function, and this approach typically precludes targeting proteins that lack such amenable sites. Furthermore, high systemic drug exposures may be needed to maintain sufficient target inhibition in vivo, increasing the risk of undesirable off-target effects. Induced protein degrdn. is an alternative approach that is event-driven: upon drug binding, the target protein is tagged for elimination. Emerging technologies based on proteolysis-targeting chimaeras (PROTACs) that exploit cellular quality control machinery to selectively degrade target proteins are attracting considerable attention in the pharmaceutical industry owing to the advantages they could offer over traditional small-mol. strategies. These advantages include the potential to reduce systemic drug exposure, the ability to counteract increased target protein expression that often accompanies inhibition of protein function and the potential ability to target proteins that are not currently therapeutically tractable, such as transcription factors, scaffolding and regulatory proteins.
- 28Sakamoto, K. M.; Kim, K. B.; Kumagai, A.; Mercurio, F.; Crews, C. M.; Deshaies, R. J. Protacs: Chimeric molecules that target proteins to the Skp1–Cullin–F box complex for ubiquitination and degradation. Proc. Natl. Acad. Sci. U.S.A. 2001, 98, 8554– 8559, DOI: 10.1073/pnas.14123079828https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXls1Wisbk%253D&md5=540358035222f745f7b6367b38781a21Protacs: chimeric molecules that target proteins to the Skp1-Cullin-F box complex for ubiquitination and degradationSakamoto, Kathleen M.; Kim, Kyung B.; Kumagai, Akiko; Mercurio, Frank; Crews, Craig M.; Deshaies, Raymond J.Proceedings of the National Academy of Sciences of the United States of America (2001), 98 (15), 8554-8559CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The intracellular levels of many proteins are regulated by ubiquitin-dependent proteolysis. One of the best-characterized enzymes that catalyzes the attachment of ubiquitin to proteins is a ubiquitin ligase complex, Skp1-Cullin-F box complex contg. Hrt1 (SCF). We sought to artificially target a protein to the SCF complex for ubiquitination and degrdn. To this end, we tested methionine aminopeptidase-2 (MetAP-2), which covalently binds the angiogenesis inhibitor ovalicin. A chimeric compd., protein-targeting chimeric mol. 1 (Protac-1), was synthesized to recruit MetAP-2 to SCF. One domain of Protac-1 contains the IκBα phosphopeptide that is recognized by the F-box protein β-TRCP, whereas the other domain is composed of ovalicin. We show that MetAP-2 can be tethered to SCFβ-TRCP, ubiquitinated, and degraded in a Protac-1-dependent manner. In the future, this approach may be useful for conditional inactivation of proteins, and for targeting disease-causing proteins for destruction.
- 29Hu, J.; Hu, B.; Wang, M.; Xu, F.; Miao, B.; Yang, C.-Y.; Wang, M.; Liu, Z.; Hayes, D. F.; Chinnaswamy, K.; Delproposto, J.; Stuckey, J.; Wang, S. Discovery of ERD-308 as a Highly Potent Proteolysis Targeting Chimera (PROTAC) Degrader of Estrogen Receptor (ER). J. Med. Chem. 2019, 62, 1420– 1442, DOI: 10.1021/acs.jmedchem.8b0157229https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXht1Cqs7s%253D&md5=678e82dd460c065e2054ba18ea8af18aDiscovery of ERD-308 as a highly potent proteolysis targeting chimera (PROTAC) degrader of estrogen receptor (ER)Hu, Jiantao; Hu, Biao; Wang, Mingliang; Xu, Fuming; Miao, Bukeyan; Yang, Chao-Yie; Wang, Mi; Liu, Zhaomin; Hayes, Daniel F.; Chinnaswamy, Krishnapriya; Delproposto, James; Stuckey, Jeanne; Wang, ShaomengJournal of Medicinal Chemistry (2019), 62 (3), 1420-1442CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The estrogen receptor (ER) is a validated target for the treatment of estrogen receptor-pos. (ER+) breast cancer. Here, we describe the design, synthesis, and extensive structure-activity relationship (SAR) studies of small-mol. ERα degraders based on the proteolysis targeting chimeras (PROTAC) concept. Our efforts have resulted in the discovery of highly potent and effective PROTAC ER degraders, as exemplified by ERD-308 I. I achieves DC50 (concn. causing 50% of protein degrdn.) values of 0.17 and 0.43 nM in MCF-7 and T47D ER+ breast cancer cell lines, resp., and induces >95% of ER degrdn. at concns. as low as 5 nM in both cell lines. Significantly, I induces more complete ER degrdn. than fulvestrant, the only approved selective ER degrader (SERD), and is more effective in inhibition of cell proliferation than fulvestrant in MCF-7 cells. Further optimization of I may lead to a new therapy for advanced ER+ breast cancer.
- 30Han, X.; Wang, C.; Qin, C.; Xiang, W.; Fernandez-Salas, E.; Yang, C.-Y.; Wang, M.; Zhao, L.; Xu, T.; Chinnaswamy, K.; Delproposto, J.; Stuckey, J.; Wang, S. Discovery of ARD-69 as a highly potent proteolysis targeting chimera (PROTAC) degrader of androgen receptor (AR) for the treatment of prostate cancer. J. Med. Chem. 2019, 62, 941– 964, DOI: 10.1021/acs.jmedchem.8b0163130https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXlvFCjtQ%253D%253D&md5=2376573fb1a1e19c517d64c70199700aDiscovery of ARD-69 as a highly potent proteolysis targeting chimera (PROTAC) degrader of androgen receptor (AR) for the treatment of prostate cancerHan, Xin; Wang, Chao; Qin, Chong; Xiang, Weiguo; Fernandez-Salas, Ester; Yang, Chao-Yie; Wang, Mi; Zhao, Lijie; Xu, Tianfeng; Chinnaswamy, Krishnapriya; Delproposto, James; Stuckey, Jeanne; Wang, ShaomengJournal of Medicinal Chemistry (2019), 62 (2), 941-964CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)We report herein the discovery of highly potent PROTAC degraders of androgen receptor (AR), as exemplified by IARD-69. I induces degrdn. of AR protein in AR-pos. prostate cancer cell lines in a dose- and time-dependent manner. I achieves DC50 values of 0.86, 0.76, and 10.4 nM in LNCaP, VCaP, and 22Rv1 AR+ prostate cancer cell lines, resp. I is capable of reducing the AR protein level by >95% in these prostate cancer cell lines and effectively suppressing AR-regulated gene expression. I potently inhibits cell growth in these AR-pos. prostate cancer cell lines and is >100 times more potent than AR antagonists. A single dose of I effectively reduces the level of AR protein in xenograft tumor tissue in mice. Further optimization of I may ultimately lead to a new therapy for AR+, castration-resistant prostate cancer.
- 31Winter, G. E.; Buckley, D. L.; Paulk, J.; Roberts, J. M.; Souza, A.; Dhe-Paganon, S.; Bradner, J. E. Phthalimide conjugation as a strategy for in vivo target protein degradation. Science 2015, 348, 1376– 1381, DOI: 10.1126/science.aab143331https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXpvFansLk%253D&md5=b0277cc95d318d926b96e7a119066e3bPhthalimide conjugation as a strategy for in vivo target protein degradationWinter, Georg E.; Buckley, Dennis L.; Paulk, Joshiawa; Roberts, Justin M.; Souza, Amanda; Dhe-Paganon, Sirano; Bradner, James E.Science (Washington, DC, United States) (2015), 348 (6241), 1376-1381CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)The development of effective pharmacol. inhibitors of multidomain scaffold proteins, notably transcription factors, is a particularly challenging problem. In part, this is because many small-mol. antagonists disrupt the activity of only one domain in the target protein. The authors devised a chem. strategy that promotes ligand-dependent target protein degrdn. using as an example the transcriptional coactivator BRD4, a protein crit. for cancer cell growth and survival. The authors appended a competitive antagonist of BET bromodomains to a phthalimide moiety to hijack the cereblon E3 ubiquitin ligase complex. The resultant compd., dBET1, induced highly selective cereblon-dependent BET protein degrdn. in vitro and in vivo and delayed leukemia progression in mice. A second series of probes resulted in selective degrdn. of the cytosolic protein FKBP12. This chem. strategy for controlling target protein stability may have implications for therapeutically targeting previously intractable proteins.
- 32Steinebach, C.; Kehm, H.; Lindner, S.; Vu, L. P.; Köpff, S.; López Mármol, Á.; Weiler, C.; Wagner, K. G.; Reichenzeller, M.; Krönke, J.; Gütschow, M. PROTAC-mediated crosstalk between E3 ligases. Chem. Commun. 2019, 55, 1821– 1824, DOI: 10.1039/c8cc09541h32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtFSmtLY%253D&md5=17ed8abc73d45081c73f1995205713cfPROTAC-mediated crosstalk between E3 ligasesSteinebach, Christian; Kehm, Hannes; Lindner, Stefanie; Vu, Lan Phuong; Koepff, Simon; Lopez Marmol, Alvaro; Weiler, Corinna; Wagner, Karl G.; Reichenzeller, Michaela; Kroenke, Jan; Guetschow, MichaelChemical Communications (Cambridge, United Kingdom) (2019), 55 (12), 1821-1824CODEN: CHCOFS; ISSN:1359-7345. (Royal Society of Chemistry)Small-mol. heterobifunctional degraders can effectively control protein levels and are useful research tools. We assembled proteolysis targeting chimeras (PROTACs) from a cereblon (CRBN) and a von-Hippel-Lindau (VHL) ligase ligand and demonstrated a PROTAC-induced heterodimerization of the two E3 ligases leading to unidirectional and efficient degrdn. of CRBN.
- 33Lu, M.; Liu, T.; Jiao, Q.; Ji, J.; Tao, M.; Liu, Y.; You, Q.; Jiang, Z. Discovery of a Keap1-dependent peptide PROTAC to knockdown Tau by ubiquitination-proteasome degradation pathway. Eur. J. Med. Chem. 2018, 146, 251– 259, DOI: 10.1016/j.ejmech.2018.01.06333https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXisFehsbY%253D&md5=165e6f510e4e2024aca6c4135d2840b2Discovery of a Keap1-dependent peptide PROTAC to knockdown Tau by ubiquitination-proteasome degradation pathwayLu, Mengchen; Liu, Tian; Jiao, Qiong; Ji, Jianai; Tao, Mengmin; Liu, Yijun; You, Qidong; Jiang, ZhengyuEuropean Journal of Medicinal Chemistry (2018), 146 (), 251-259CODEN: EJMCA5; ISSN:0223-5234. (Elsevier Masson SAS)Induced protein degrdn. by PROTACs has emerged as a promising strategy to target nonenzymic proteins inside the cell. The aim of this study was to identify Keap1, a substrate adaptor protein for ubiquitin E3 ligase involved in oxidative stress regulation, as a novel candidate for PROTACs that can be applied in the degrdn. of the nonenzymic protein Tau. A peptide PROTAC by recruiting Keap1-Cul3 ubiquitin E3 ligase was developed and applied in the degrdn. of intracellular Tau. Peptide 1 showed strong in vitro binding with Keap1 and Tau. With proper cell permeability, peptide 1 was found to colocalize with cellular Keap1 and resulted in the coimmunopptn. of Tau and Keap1. The results of flow cytometry and western blotting assays showed that peptide 1 can downregulate the intracellular Tau level in both time- and concn.-dependent manner. The application of Keap1 siRNA silencing and the proteasome inhibitor MG132 confirmed that peptide 1 could promote the Keap1-dependent poly-ubiquitination and proteasome-dependent degrdn. of Tau. The results suggested that using PROTACs to recruit Keap1 to induce the degrdn. of Tau may show promising character in the treatment of neurodegenerative disease. In addn., our research demonstrated that Keap1 should be a promising E3 ligase adaptor to be used in the design of novel PROTACs.
- 34Kargbo, R. B. Treatment of Alzheimer’s by PROTAC-Tau protein degradation. ACS Med. Chem. Lett. 2019, 10, 699– 700, DOI: 10.1021/acsmedchemlett.9b0008334https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXkvVSnu70%253D&md5=7d4254b26c56f6401c16e6d100ddf0bdTreatment of Alzheimer's by PROTAC-Tau Protein DegradationKargbo, Robert B.ACS Medicinal Chemistry Letters (2019), 10 (5), 699-700CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)There is no expanded citation for this reference.
- 35Chu, T.-T.; Gao, N.; Li, Q.-Q.; Chen, P.-G.; Yang, X.-F.; Chen, Y.-X.; Zhao, Y.-F.; Li, Y.-M. Specific knockdown of endogenous Tau protein by peptide-directed ubiquitin-proteasome degradation. Cell Chem. Biol. 2016, 23, 453– 461, DOI: 10.1016/j.chembiol.2016.02.01635https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XntlSqtbg%253D&md5=d965c74777427dfa9b6068f4f51a0f12Specific Knockdown of Endogenous Tau Protein by Peptide-Directed Ubiquitin-Proteasome DegradationChu, Ting-Ting; Gao, Na; Li, Qian-Qian; Chen, Pu-Guang; Yang, Xi-Fei; Chen, Yong-Xiang; Zhao, Yu-Fen; Li, Yan-MeiCell Chemical Biology (2016), 23 (4), 453-461CODEN: CCBEBM; ISSN:2451-9448. (Cell Press)Tau, an important pathol. protein of Alzheimer's disease (AD), can mediate the toxicity of amyloid β (Aβ). Thus, redn. of Tau with chem. mols. may offer a novel strategy for treating AD. Here, we designed and synthesized a series of multifunctional mols. that contained Tau-recognition moieties and E3 ligase-binding moieties to enhance Tau degrdn. Among these mols., TH006 had the highest activity of inducing Tau degrdn. by increasing its poly-ubiquitination. The decrement in Tau induced by TH006 could decrease the cytotoxicity caused by Aβ. Furthermore, TH006 could regulate the Tau level in the brain of an AD mouse model. Therefore, partial redn. of Tau with such multifunctional peptides may open up a novel therapeutic strategy for AD treatment.
- 36Boichenko, I.; Bär, K.; Deiss, S.; Heim, C.; Albrecht, R.; Lupas, A. N.; Hernandez Alvarez, B.; Hartmann, M. D. Chemical ligand space of cereblon. ACS Omega 2018, 3, 11163– 11171, DOI: 10.1021/acsomega.8b0095936https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhslSku7bO&md5=b0fee25453d5ec83098ac0cf28575d44Chemical Ligand Space of CereblonBoichenko, Iuliia; Baer, Kerstin; Deiss, Silvia; Heim, Christopher; Albrecht, Reinhard; Lupas, Andrei N.; Hernandez Alvarez, Birte; Hartmann, Marcus D.ACS Omega (2018), 3 (9), 11163-11171CODEN: ACSODF; ISSN:2470-1343. (American Chemical Society)The protein, cereblon, serves as a substrate receptor of a ubiquitin ligase complex that can be tuned toward different target proteins by cereblon-binding agents. This approach to targeted protein degrdn. is exploited in different clin. settings and has sparked the development of a growing no. of thalidomide derivs. Here, we probed the chem. space of cereblon binding beyond such derivs. and worked out a simple set of chem. requirements, delineating the metaclass of cereblon effectors. We report co-crystal structures of Magnetospirillum gryphiswaldense cereblon isoform 4 with a diverse set of compds., including commonly used pharmaceuticals, but we also found that already minimalistic cereblon-binding moieties might exert teratogenic effects in zebrafish. These results may guide the design of a post-thalidomide generation of therapeutic cereblon effectors, and provide a framework for the circumvention of unintended cereblon-binding by neg. design for future pharmaceuticals.
- 37Burslem, G. M.; Ottis, P.; Jaime-Figueroa, S.; Morgan, A.; Cromm, P. M.; Toure, M.; Crews, C. M. Efficient synthesis of immunomodulatory drug analogues enables exploration of structure-degradation relationships. ChemMedChem 2018, 13, 1508– 1512, DOI: 10.1002/cmdc.20180027137https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXht1KntL7O&md5=74ada081313c4e01b430fb6014441e2eEfficient Synthesis of Immunomodulatory Drug Analogues Enables Exploration of Structure-Degradation RelationshipsBurslem, George M.; Ottis, Philipp; Jaime-Figueroa, Saul; Morgan, Alicia; Cromm, Philipp M.; Toure, Momar; Crews, Craig M.ChemMedChem (2018), 13 (15), 1508-1512CODEN: CHEMGX; ISSN:1860-7179. (Wiley-VCH Verlag GmbH & Co. KGaA)The immunomodulatory drugs (IMiDs) thalidomide, pomalidomide, and lenalidomide have been approved for the treatment of multiple myeloma for many years. Recently, their use as E3 ligase recruiting elements for small-mol.-induced protein degrdn. has led to a resurgence in interest in IMiD synthesis and functionalization. Traditional IMiD synthesis follows a stepwise route with multiple purifn. steps. Herein we describe a novel one-pot synthesis without purifn. that provides rapid access to a multitude of IMiD analogs. Binding studies with the IMiD target protein cereblon (CRBN) reveals a narrow structure-activity relationship with only a few compds. showing sub-micromolar binding affinity in the range of pomalidomide and lenalidomide. However, anti-proliferative activity as well as Aiolos degrdn. could be identified for two IMiD analogs. This study provides useful insight into the structure-degrdn. relationships for mols. of this type as well as a rapid and robust method for IMiD synthesis.
- 38Bartlett, J. B.; Dredge, K.; Dalgleish, A. G. The evolution of thalidomide and its IMiD derivatives as anticancer agents. Nat. Rev. Cancer 2004, 4, 314– 322, DOI: 10.1038/nrc132338https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXis1Gmt7k%253D&md5=99d2ac4c0f6d6d1dac5547e5360dd74fTimeline: The evolution of thalidomide and its IMiD derivatives as anticancer agentsBartlett, J. Blake; Dredge, Keith; Dalgleish, Angus G.Nature Reviews Cancer (2004), 4 (4), 314-322CODEN: NRCAC4; ISSN:1474-175X. (Nature Publishing Group)A review. Thalidomide was originally used to treat morning sickness, but was banned in the 1960s for causing serious congenital birth defects. Remarkably, thalidomide was subsequently discovered to have anti-inflammatory and anti-angiogenic properties, and was identified as an effective treatment for multiple myeloma. A series of immunomodulatory drugs - created by chem. modification of thalidomide - have been developed to overcome the original devastating side effects. Their powerful anticancer properties mean that these drugs are now emerging from thalidomide's shadow as useful anticancer agents.
- 39Reist, M.; Carrupt, P.-A.; Francotte, E.; Testa, B. Chiral inversion and hydrolysis of thalidomide: mechanisms and catalysis by bases and serum albumin, and chiral stability of teratogenic metabolites. Chem. Res. Toxicol. 1998, 11, 1521– 1528, DOI: 10.1021/tx980181739https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXntFens7s%253D&md5=1952fa00825e6791eeb8388419f3cc09Chiral Inversion and Hydrolysis of Thalidomide: Mechanisms and Catalysis by Bases and Serum Albumin, and Chiral Stability of Teratogenic MetabolitesReist, Marianne; Carrupt, Pierre-Alain; Francotte, Eric; Testa, BernardChemical Research in Toxicology (1998), 11 (12), 1521-1528CODEN: CRTOEC; ISSN:0893-228X. (American Chemical Society)The chiral inversion and hydrolysis of thalidomide and the catalysis by bases and human serum albumin were investigated by using a stereoselective HPLC assay. Chiral inversion was catalyzed by albumin, hydroxyl ions, phosphate, and amino acids. Basic amino acids (Arg and Lys) had a superior potency in catalyzing chiral inversion compared to acid and neutral ones. The chiral inversion of thalidomide is thus subject to specific and general base catalysis, and it is suggested that the ability of HSA to catalyze the reaction is due to the basic groups of the amino acids Arg and Lys and not to a single catalytic site on the macromol. The hydrolysis of thalidomide was also base-catalyzed. However, albumin had no effect on hydrolysis, and there was no difference between the catalytic potencies of acidic, neutral, and basic amino acids. This may be explained by different reaction mechanisms of the chiral inversion and hydrolysis of thalidomide. Chiral inversion is deduced to occur by electrophilic substitution involving specific and general base catalysis, whereas hydrolysis is thought to occur by nucleophilic substitution involving specific and general base as well as nucleophilic catalysis. As nucleophilic attack is sensitive to steric properties of the catalyst, steric hindrance might be the reason albumin is not able to catalyze hydrolysis. 1H NMR expts. revealed that the three teratogenic metabolites of thalidomide, in sharp contrast to the drug itself, had complete chiral stability. This leads to the speculation that, were some enantioselectivity to exist in the teratogenicity of thalidomide, it could result from fast hydrolysis to chirally stable teratogenic metabolites.
- 40Chen, T. L.; Vogelsang, G. B.; Petty, B. G.; Brundrett, R. B.; Noe, D. A.; Santos, G. W.; Colvin, O. M. Plasma pharmacokinetics and urinary excretion of thalidomide after oral dosing in healthy male volunteers. Drug Metab Dispos 1989, 17, 402– 40540https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL1MXlt1KnsLY%253D&md5=e171e48357c05ea8fedbc2979293650ePlasma pharmacokinetics and urinary excretion of thalidomide after oral dosing in healthy male volunteersChen, Tian Ling; Vogelsang, Georgia B.; Petty, Brent G.; Brundrett, Robert B.; Noe, Dennis A.; Santos, George W.; Colvin, O. MichaelDrug Metabolism and Disposition (1989), 17 (4), 402-5CODEN: DMDSAI; ISSN:0090-9556.The plasma pharmacokinetics and urinary excretion of thalidomide were evaluated in healthy male volunteers receiving a single oral dose of 200 mg. Concns. of thalidomide were detd. by a new HPLC assay. Plasma concn. vs. time data were well fit by a 1-compartment model. The mean peak concn., 1.15 μg/mL, was achieved at 4.39 h. Absorption and elimination half-lives were 1.70 and 8.70 h, resp., with a lag time of 0.41 h obsd. in 6 of 8 subjects. The apparent vol. of distribution and total body clearance rate, based on assumed complete bioavailability, were 120.69 L and 10.41 L/h. The urinary excretion of thalidomide accounted for only 0.6% of the total dose administered over 24 h, and the renal clearance rate was 0.08 L/h. Thus, the major route of elimination of thalidomide is nonrenal.
- 41Schumacher, H.; Smith, R. L.; Williams, R. T. The metabolism of thalidomide: the fate of thalidomide and some of its hydrolysis products in various species. Br. J. Pharmacol. Chemother. 1965, 25, 338– 351, DOI: 10.1111/j.1476-5381.1965.tb02054.x41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaF28Xhtl2rsA%253D%253D&md5=cb686e6f49ced088a96fc32b61f2256bThe metabolism of thalidomide. The fate of thalidomide and some of its hydrolysis products in various speciesSchumacher, H.; Smith, R. L.; Williams, R. T.British Journal of Pharmacology and Chemotherapy (1965), 25 (2), 338-51CODEN: BJPCAL; ISSN:0366-0826.cf. preceding abstr. The metabolism of thalidomide in the rabbit, rat, mouse, and guinea pig was investigated. The metabolites of thalidomide present in the urine, blood, and tissues of various species dosed with the drug were characterized by comparing their chromatographic mobility and color reactions with those given by authentic samples of the compds. The rabbit urinary metabolites were isolated in cryst. form by solvent extn. and adsorption chromatography, and their identity was established by analysis, m.p. behavior, and comparison of their ir spectra with those of the authentic compds. When thalidomide is fed to rabbits, rats, mice, and guinea pigs a no. of hydrolysis products appear in the urine. These hydrolysis products are formed by the spontaneous hydrolysis of thalidomide. In addn. the urine of rabbits dosed with thalidomide contains derivs. of 3- and 4-hydroxyphthalic acid; these minor metabolites were not identified. The hydrolysis products appear to be derived by spontaneous breakdown of thalidomide in the body, although it is possible that any of the hydrolytic reactions of thalidomide may be assisted by hydrolases in the body. In rats, some breakdown of thalidomide occurs in the gut before absorption; hydrolysis products are present in the gastrointestinal tract following the oral administration of the drug. Thalidomide and some of its hydrolysis products can be detected in the plasma and brain of rats dosed orally with thalidomide.
- 42Teo, S. K.; Colburn, W. A.; Tracewell, W. G.; Kook, K. A.; Stirling, D. I.; Jaworsky, M. S.; Scheffler, M. A.; Thomas, S. D.; Laskin, O. L. Clinical pharmacokinetics of thalidomide. Clin. Pharmacokinet. 2004, 43, 311– 327, DOI: 10.2165/00003088-200443050-0000442https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXktFOqtbg%253D&md5=aa199f0c21a3fdb952fccde4059b71ecClinical pharmacokinetics of thalidomideTeo, Steve K.; Colburn, Wayne A.; Tracewell, William G.; Kook, Karin A.; Stirling, David I.; Jaworsky, Markian S.; Scheffler, Michael A.; Thomas, Steve D.; Laskin, Oscar L.Clinical Pharmacokinetics (2004), 43 (5), 311-327CODEN: CPKNDH; ISSN:0312-5963. (Adis International Ltd.)A review. Thalidomide is a racemic glutamic acid deriv. approved in the US for erythema nodosum leprosum, a complication of leprosy. In addn., its use in various inflammatory and oncol. conditions is being investigated. Thalidomide interconverts between the (R)- and (S)-enantiomers in plasma, with protein binding of 55% and 65%, resp. More than 90% of the absorbed drug is excreted in the urine and feces within 48 h. Thalidomide is minimally metabolized by the liver, but is spontaneously hydrolyzed into numerous renally excreted products. After a single oral dose of thalidomide 200mg (as the US-approved capsule formulation) in healthy volunteers, absorption is slow and extensive, resulting in a peak concn. (Cmax) of 1-2 mg/L at 3-4 h after administration, absorption lag time of 30 min, total exposure (AUC∞) of 18 mg • h/L, apparent elimination half-life of 6 h and apparent systemic clearance of 10 L/h. Thalidomide pharmacokinetics are best described by a one-compartment model with first-order absorption and elimination. Because of the low soly. of the drug in the gastrointestinal tract, thalidomide exhibits absorption rate-limited pharmacokinetics (the "flip-flop" phenomenon), with its elimination rate being faster than its absorption rate. The apparent elimination half-life of 6 h therefore represents absorption, not elimination. The "true" apparent vol. of distribution was estd. to be 16L by use of the faster elimination-rate half-life. Multiple doses of thalidomide 200 mg/day over 21 days cause no change in the pharmacokinetics, with a steady-state Cmax (Cssmax) of 1.2 mg/L. Simulation of 400 and 800 mg/day also shows no accumulation, with Cssmax of 3.5 and 6.0 mg/L, resp. Multiple-dose studies in cancer patients show pharmacokinetics comparable with those in healthy populations at similar dosages. Thalidomide exhibits a dose-proportional increase in AUC at doses from 50 to 400mg. Because of the low soly. of thalidomide, Cmax is less than proportional to dose, and tmax is prolonged with increasing dose. Age, sex and smoking have no effect on the pharmacokinetics of thalidomide, and the effect of food is minimal. Thalidomide does not alter the pharmacokinetics of oral contraceptives, and is also unlikely to interact with warfarin and grapefruit juice. Since thalidomide is mainly hydrolyzed and passively excreted, its pharmacokinetics are not expected to change in patients with impaired liver or kidney function.
- 43Beckmann, R. Ueber das Verhalten von Thalidomid im Organismus. Arzneimittelforschung 1963, 13, 185– 19143https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaF387mtF2mug%253D%253D&md5=211cb3a294eb7d33c0cfd689bb37d2bcC-14])BECKMANN RArzneimittel-Forschung (1963), 13 (), 185-91 ISSN:0004-4172.There is no expanded citation for this reference.
- 44Chung, F.; Lu, J.; Palmer, B. D.; Kestell, P.; Browett, P.; Baguley, B. C.; Tingle, M.; Ching, L. M. Thalidomide pharmacokinetics and metabolite formation in mice, rabbits, and multiple myeloma patients. Clin. Cancer Res. 2004, 10, 5949– 5956, DOI: 10.1158/1078-0432.ccr-04-042144https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXntlyjtLw%253D&md5=51bea71546be92322a511405e004aa80Thalidomide pharmacokinetics and metabolite formation in mice, rabbits, and multiple myeloma patientsChung, Francisco; Lu, Jun; Palmer, Brian D.; Kestell, Philip; Browett, Peter; Baguley, Bruce C.; Tingle, Malcolm; Ching, Lai-MingClinical Cancer Research (2004), 10 (17), 5949-5956CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)Thalidomide has a variety of biol. effects that vary considerably according to the species tested. The authors sought to establish whether differences in pharmacokinetics could form a basis for the species-specific effects of thalidomide. Mice and rabbits were administered thalidomide (2 mg/kg) p.o. or i.v., and plasma concns. of thalidomide were measured after drug administration using high performance liq. chromatog. Plasma samples from five multiple myeloma patients over 24 h after their first dose of thalidomide (200 mg) were similarly analyzed and all data were fitted to a one-compartment model. Metabolites of thalidomide in plasma were identified simultaneously using liq. chromatog.-mass spectrometry. Plasma concn.-time profiles for the individual patients were very similar to each other, but widely different pharmacokinetic properties were found between patients compared with those in mice or rabbits. Area under the concn. curve values for mice, rabbits, and multiple myeloma patients were 4, 8, and 81 μmol/L · hour, resp., and corresponding elimination half-lives were 0.5, 2.2, and 7.3 h, resp. Large differences were also obsd. between the metabolite profiles from the three species. Hydrolysis products were detected for all species, and the proportion of hydroxylated metabolites was higher in mice than in rabbits and undetectable in patients. Our results show major interspecies differences in the pharmacokinetics of thalidomide that are related to the altered degree of metab. The authors suggest that the interspecies differences in biol. effects of thalidomide may be attributable, at least in part, to the differences in its metab. and hence pharmacokinetics.
- 45Nakamura, T.; Noguchi, T.; Miyachi, H.; Hashimoto, Y. Hydrolyzed metabolites of thalidomide: synthesis and TNF-alpha production-inhibitory activity. Chem. Pharm. Bull. 2007, 55, 651– 654, DOI: 10.1248/cpb.55.65145https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXls1arsLc%253D&md5=32f1066b688a9de33e9dbf5da42ec514Hydrolyzed metabolites of thalidomide: synthesis and TNF-α production-inhibitory activityNakamura, Takanori; Noguchi, Tomomi; Miyachi, Hiroyuki; Hashimoto, YuichiChemical & Pharmaceutical Bulletin (2007), 55 (4), 651-654CODEN: CPBTAL; ISSN:0009-2363. (Pharmaceutical Society of Japan)Putative hydrolyzed metabolites of thalidomide (I) were prepd. and characterized, and their inhibitory activity on tumor necrosis factor (TNF)-α prodn. in the human monocytic leukemia cell line THP-1 was evaluated. α-(2-Carboxybenzamido)glutarimide (II) was a more potent TNF-α prodn. inhibitor than I.
- 46Otogawa, K.; Ogino, Y.; Ishikawa, K.; Tanaka, M.; Shiro, M.; Osaka, T.; Asahi, T. Structural and thermal analyses of a hydrolysis compound of thalidomide. Acta Crystallogr., Sect. A: Found. Adv. 2014, 70, C113, DOI: 10.1107/s2053273314098866There is no corresponding record for this reference.
- 47Krönke, J.; Hurst, S. N.; Ebert, B. L. Lenalidomide induces degradation of IKZF1 and IKZF3. OncoImmunology 2014, 3, e941742 DOI: 10.4161/21624011.2014.941742There is no corresponding record for this reference.
- 48Schafer, P. H.; Ye, Y.; Wu, L.; Kosek, J.; Ringheim, G.; Yang, Z.; Liu, L.; Thomas, M.; Palmisano, M.; Chopra, R. Cereblon modulator iberdomide induces degradation of the transcription factors Ikaros and Aiolos: immunomodulation in healthy volunteers and relevance to systemic lupus erythematosus. Ann. Rheum. Dis. 2018, 77, 1516– 1523, DOI: 10.1136/annrheumdis-2017-21291648https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXjtVektbw%253D&md5=200098adf4787ee935df528d18a5508dCereblon modulator iberdomide induces degradation of the transcription factors Ikaros and Aiolos: immunomodulation in healthy volunteers and relevance to systemic lupus erythematosusSchafer, Peter H.; Ye, Ying; Wu, Lei; Kosek, Jolanta; Ringheim, Garth; Yang, Zhihong; Liu, Liangang; Thomas, Michael; Palmisano, Maria; Chopra, RajeshAnnals of the Rheumatic Diseases (2018), 77 (10), 1516-1523CODEN: ARDIAO; ISSN:0003-4967. (BMJ Publishing Group)IKZF1 and IKZF3 (encoding transcription factors Ikaros and Aiolos) are susceptibility loci for systemic lupus erythematosus (SLE). The pharmacol. of iberdomide (CC-220), a cereblon (CRBN) modulator targeting Ikaros and Aiolos, was studied in SLE patient cells and in a phase 1 healthy volunteer study. CRBN, IKZF1 and IKZF3 gene expression was measured in peripheral blood mononuclear cells (PBMC) from patients with SLE and healthy volunteers. Ikaros and Aiolos protein levels were measured by Western blot and flow cytometry. Anti-dsDNA and anti-phospholipid autoantibodies were measured in SLE PBMC cultures treated for 7 days with iberdomide. Fiftysix healthy volunteers were randomized to a single dose of iberdomide (0.03-6mg, n=6 across seven cohorts) or placebo (n=2/cohort). CD19+ B cells, CD3+ T cells and intracellular Aiolos were measured by flow cytometry. Interleukin (IL)-2 and IL-1β prodn. was stimulated with anti-CD3 and lipopolysaccharide, resp., in an ex vivo whole blood assay. SLE patient PBMCs expressed significantly higher CRBN (1.5-fold), IKZF1 (2.1-fold) and IKZF3 (4.1- fold) mRNA levels compared with healthy volunteers. Iberdomide significantly reduced Ikaros and Aiolos protein levels in B cells, T cells and monocytes. In SLE PBMC cultures, iberdomide inhibited anti-dsDNA and anti-phospholipid autoantibody prodn. (IC50 ≈10 nM). Single doses of iberdomide (0.3-6mg) in healthy volunteers decreased intracellular Aiolos (min. mean per cent of baseline: ≈12%-28% (B cells); ≈0%-33% (T cells)), decreased abs. CD19+ B cells, increased IL-2 and decreased IL-1β prodn. ex vivo. These findings demonstrate pharmacodynamic activity of iberdomide and support its further clin. development for the treatment of SLE.
- 49Matyskiela, M. E.; Zhang, W.; Man, H.-W.; Muller, G.; Khambatta, G.; Baculi, F.; Hickman, M.; LeBrun, L.; Pagarigan, B.; Carmel, G.; Lu, C.-C.; Lu, G.; Riley, M.; Satoh, Y.; Schafer, P.; Daniel, T. O.; Carmichael, J.; Cathers, B. E.; Chamberlain, P. P. A Cereblon Modulator (CC-220) with Improved Degradation of Ikaros and Aiolos. J. Med. Chem. 2018, 61, 535– 542, DOI: 10.1021/acs.jmedchem.6b0192149https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXmt1KqsLs%253D&md5=8358ae8563f8f2c09dbf68683e5e4008A Cereblon Modulator (CC-220) with Improved Degradation of Ikaros and AiolosMatyskiela, Mary E.; Zhang, Weihong; Man, Hon-Wah; Muller, George; Khambatta, Godrej; Baculi, Frans; Hickman, Matthew; LeBrun, Laurie; Pagarigan, Barbra; Carmel, Gilles; Lu, Chin-Chun; Lu, Gang; Riley, Mariko; Satoh, Yoshitaka; Schafer, Peter; Daniel, Thomas O.; Carmichael, James; Cathers, Brian E.; Chamberlain, Philip P.Journal of Medicinal Chemistry (2018), 61 (2), 535-542CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The drugs lenalidomide and pomalidomide bind to the protein cereblon, directing the CRL4-CRBN E3 ligase toward the transcription factors Ikaros and Aiolos to cause their ubiquitination and degrdn. Here we describe CC-220 (compd. 6), a cereblon modulator in clin. development for systemic lupus erythematosis and relapsed/refractory multiple myeloma. Compd. 6 binds cereblon with a higher affinity than lenalidomide or pomalidomide. Consistent with this, the cellular degrdn. of Ikaros and Aiolos is more potent and the extent of substrate depletion is greater. The crystal structure of cereblon in complex with DDB1 and compd. 6 reveals that the increase in potency correlates with increased contacts between compd. 6 and cereblon away from the modeled binding site for Ikaros/Aiolos. These results describe a new cereblon modulator which achieves greater substrate degrdn. via tighter binding to the cereblon E3 ligase and provides an example of the effect of E3 ligase binding affinity with relevance to other drug discovery efforts in targeted protein degrdn.
- 50Capitosti, S. M.; Hansen, T. P.; Brown, M. L. Facile synthesis of an azido-labeled thalidomide analogue. Org. Lett. 2003, 5, 2865– 2867, DOI: 10.1021/ol034906w50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXlt1Cgs70%253D&md5=fdefbdcf05d15f638e322bc9e5687ebbFacile Synthesis of an Azido-Labeled Thalidomide AnalogueCapitosti, Scott M.; Hansen, Todd P.; Brown, Milton L.Organic Letters (2003), 5 (16), 2865-2867CODEN: ORLEF7; ISSN:1523-7060. (American Chemical Society)A five-step synthesis of an azido-thalidomide analog I is presented. The sequence requires cheap and readily available starting materials and reagents, and only two steps require purifn. Addnl., I possesses activity comparable to that of thalidomide in inhibiting the proliferation of human microvascular endothelial cells, thus providing impetus for its use as a potential photoaffinity label of thalidomide.
- 51Joossens, J.; Van der Veken, P.; Lambeir, A.-M.; Augustyns, K.; Haemers, A. Development of irreversible diphenyl phosphonate inhibitors for urokinase plasminogen activator. J. Med. Chem. 2004, 47, 2411– 2413, DOI: 10.1021/jm049920951https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXisl2it7k%253D&md5=a5e0eb9791dc09a1ac71b6cf8415af29Development of Irreversible Diphenyl Phosphonate Inhibitors for Urokinase Plasminogen ActivatorJoossens, J.; Van der Veken, P.; Lambeir, A.-M.; Augustyns, K.; Haemers, A.Journal of Medicinal Chemistry (2004), 47 (10), 2411-2413CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The authors report the synthesis and biochem. evaluation of selective, irreversible di-Ph phosphonate inhibitors for urokinase plasminogen activator (uPA). A di-Ph phosphonate group was introduced on the substrate-like peptide Z-D-Ser-Ala-Arg, and modification of the guanidine side chain was investigated. A guanylated benzyl group appeared the most promising side chain modification. A kapp value in the 103 M-1 s-1 range for uPA was obtained, together with a selectivity index higher than 240 toward other trypsin-like proteases such as tPA, thrombin, plasmin, and FXa.
- 52Kanuma, K.; Omodera, K.; Nishiguchi, M.; Funakoshi, T.; Chaki, S.; Nagase, Y.; Iida, I.; Yamaguchi, J.-i.; Semple, G.; Tran, T.-A.; Sekiguchi, Y. Identification of 4-amino-2-cyclohexylaminoquinazolines as metabolically stable melanin-concentrating hormone receptor 1 antagonists. Bioorg. Med. Chem. 2006, 14, 3307– 3319, DOI: 10.1016/j.bmc.2005.12.05252https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XjtFGjtb4%253D&md5=99bcc1d1e058b4c8e5baa2efcbae7447Identification of 4-amino-2-cyclohexylaminoquinazolines as metabolically stable melanin-concentrating hormone receptor 1 antagonistsKanuma, Kosuke; Omodera, Katsunori; Nishiguchi, Mariko; Funakoshi, Takeo; Chaki, Shigeyuki; Nagase, Yasuko; Iida, Izumi; Yamaguchi, Jun-ichi; Semple, Graeme; Tran, Thuy-Anh; Sekiguchi, YoshinoriBioorganic & Medicinal Chemistry (2006), 14 (10), 3307-3319CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)The optimization of the distance between two key pharmacophore features within our first hit compds. led to the identification of a new class of potent non-peptidic antagonists for the MCH-R1, based around 4-amino-2-cyclohexylaminoquinazolines. In particular, ATC0065, N 2-[cis-4-({2-[4-Bromo-2-(trifluoromethoxy)phenyl]ethyl}amino)cyclohexyl]-N4,N4-dimethylquinazoline-2,4-diamine dihydrochloride, bound with high affinity to the MCH-R1 (IC50 value of 16 nM) and showed good metabolic stability in liver microsomes from human and rat.
- 53Nouch, R.; Cini, M.; Magre, M.; Abid, M.; Diéguez, M.; Pàmies, O.; Woodward, S.; Lewis, W. Enantioselective synthesis of 6,6-disubstituted pentafulvenes containing a chiral pendant hydroxy group. Chem.—Eur. J. 2017, 23, 17195– 17198, DOI: 10.1002/chem.20170424753https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhvVKmurjI&md5=ed815c390637d2d2ca7c6322180f583bEnantioselective Synthesis of 6,6-Disubstituted Pentafulvenes Containing a Chiral Pendant Hydroxy GroupNouch, Ryan; Cini, Melchior; Magre, Marc; Abid, Mohammed; Dieguez, Montserrat; Pamies, Oscar; Woodward, Simon; Lewis, WilliamChemistry - A European Journal (2017), 23 (68), 17195-17198CODEN: CEUJED; ISSN:0947-6539. (Wiley-VCH Verlag GmbH & Co. KGaA)Simple enantioselective synthesis of 6,6-disubstituted pentafulvenes bearing chiral pendant hydroxy groups are attained by cascade reactivity using com. available proline-based organocatalysts. Condensation of cyclopentadiene with the acetyl function of a 1,2-formylacetophenone, followed by cyclization of a resulting fulvene-stabilized carbanion with the formyl group, generates bicyclic chiral alcs. with initial er values up to 94:6. Exceptional enantio-enrichment of the resultant alcs. results upon crystn.-even near racemic samples spontaneously de-racemize. This enables new families of substituted cyclopentadienes that are both enantiomerically and diastereomerically pure to be rapidly attained.
- 54Cao, R.; Müller, P.; Lippard, S. J. Tripodal tris-tacn and tris-dpa platforms for assembling phosphate-templated trimetallic centers. J. Am. Chem. Soc. 2010, 132, 17366– 17369, DOI: 10.1021/ja108212v54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhsVGksbzJ&md5=26ced8d2ad3686926092f1940b96a9ccTripodal Tris-tacn and Tris-dpa Platforms for Assembling Phosphate-Templated Trimetallic CentersCao, Rui; Muller, Peter; Lippard, Stephen J.Journal of the American Chemical Society (2010), 132 (49), 17366-17369CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Multidentate tripodal ligands, N(CH2-m-C6H4-CH2tacn)3 (L1) and N(CH2-o-C6H4-CH2N(CH2py)2)3 (L2), were devised for assembling high-nuclearity metal clusters. By using the same tripodal platform with different ligand appendages, either triazacyclononanes or dipicolylamines, and functionalizing either the ortho or the meta positions on the tris(xylyl) linker arms, discrete trimetal phosphate units of relevance to phosphate-metabolizing trimetallic centers in biol. were prepd. Four such compds., [(CuIICl)3(HPO4)L1](PF6) (1), [(CuIICl)3(HAsO4)L1](PF6) (2), Na2[MnIII6MnII2(H2O)2(HPO4)6(PO4)4(L1)2] (3), and [CoII3(H2PO4)Cl2(MeCN)L2](PF6)3 (4), all contg. three metal centers bound to a central phosphate or arsenate unit bridging oxygen atoms, were synthesized and structurally characterized. These results demonstrate the propensity of this novel tripodal ligand platform, in the presence of phosphate or arsenate, to assemble {M3(EO4)} units and thus structurally mimic trimetallic active sites of proteins involved in phosphate metab. Reactivity studies reveal that the tricopper complex 1 is more efficient than monocopper analogs in catalyzing the hydrolysis of 4-nitrophenyl phosphate.
- 55Steinebach, C.; Lindner, S.; Udeshi, N. D.; Mani, D. C.; Kehm, H.; Köpff, S.; Carr, S. A.; Gütschow, M.; Krönke, J. Homo-PROTACs for the chemical knockdown of cereblon. ACS Chem. Biol. 2018, 13, 2771– 2782, DOI: 10.1021/acschembio.8b0069355https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsFejsLrO&md5=71bf2d53ad23c2b873f754de5144883cHomo-PROTACs for the Chemical Knockdown of CereblonSteinebach, Christian; Lindner, Stefanie; Udeshi, Namrata D.; Mani, Deepak C.; Kehm, Hannes; Koepff, Simon; Carr, Steven A.; Guetschow, Michael; Kroenke, JanACS Chemical Biology (2018), 13 (9), 2771-2782CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)The immunomodulatory drugs (IMiDs) thalidomide, lenalidomide, and pomalidomide, all approved for the treatment of multiple myeloma, induce targeted ubiquitination and degrdn. of Ikaros (IKZF1) and Aiolos (IKZF3) via the cereblon (CRBN) E3 ubiquitin ligase. IMiD-based proteolysis-targeting chimeras (PROTACs) can efficiently recruit CRBN to a protein of interest, leading to its ubiquitination and proteasomal degrdn. By linking two pomalidomide mols., we designed homobifunctional, so-called homo-PROTACs and investigated their ability to induce self-directed ubiquitination and degrdn. The homodimerized compd. I was characterized as a highly potent and efficient CRBN degrader with only minimal effects on IKZF1 and IKZF3. The cellular selectivity of I for CRBN degrdn. was confirmed at the proteome level by quant. mass spectrometry. Inactivation by compd. I did not affect proliferation of different cell lines, prevented pomalidomide-induced degrdn. of IKZF1 and IKZF3, and antagonized the effects of pomalidomide on multiple myeloma cells. Homobifunctional CRBN degraders will be useful tools for future biomedical investigations of CRBN-related signaling and may help to further elucidate the mol. mechanism of thalidomide analogs.
- 56Boichenko, I.; Deiss, S.; Bär, K.; Hartmann, M. D.; Hernandez Alvarez, B. A FRET-based assay for the identification and characterization of cereblon ligands. J. Med. Chem. 2016, 59, 770– 774, DOI: 10.1021/acs.jmedchem.5b0173556https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xjs1yktg%253D%253D&md5=f0400506428f439e81f579204e0bd39dA FRET-Based Assay for the Identification and Characterization of Cereblon LigandsBoichenko, Iuliia; Deiss, Silvia; Baer, Kerstin; Hartmann, Marcus D.; Hernandez Alvarez, BirteJournal of Medicinal Chemistry (2016), 59 (2), 770-774CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Cereblon serves as an ubiquitin ligase substrate receptor that can be tuned toward different target proteins by various cereblon-binding agents. This offers one of the most promising avenues for targeted protein degrdn. in cancer therapy, but cereblon binding can also mediate teratogenic effects. We present an effective assay that is suited for high-throughput screening of compd. libraries for off-target cereblon interactions but also can guide lead optimization and rational design of novel cereblon effector mols.
- 57Kabsch, W. Xds. Acta Crystallogr., Sect. D: Biol. Crystallogr. 2010, 66, 125– 132, DOI: 10.1107/s090744490904733757https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhs1SisLc%253D&md5=1aa9a38aeb3ce95af4ffb7d8b8a142bdSoftware XDS for image rotation, recognition and crystal symmetry assignmentKabsch, WolfgangActa Crystallographica, Section D: Biological Crystallography (2010), 66 (2), 125-132CODEN: ABCRE6; ISSN:0907-4449. (International Union of Crystallography)The usage and control of recent modifications of the program package XDS for the processing of rotation images are described in the context of previous versions. New features include automatic detn. of spot size and reflecting range and recognition and assignment of crystal symmetry. Moreover, the limitations of earlier package versions on the no. of correction/scaling factors and the representation of pixel contents have been removed. Large program parts have been restructured for parallel processing so that the quality and completeness of collected data can be assessed soon after measurement.
- 58Vagin, A.; Teplyakov, A. Molecular replacement with MOLREP. Acta Crystallogr., Sect. D: Biol. Crystallogr. 2010, 66, 22– 25, DOI: 10.1107/s090744490904258958https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXit1Kktw%253D%253D&md5=820d114719aca209994ffb0403e3b20dMolecular replacement with MOLREPVagin, Alexei; Teplyakov, AlexeiActa Crystallographica, Section D: Biological Crystallography (2010), 66 (1), 22-25CODEN: ABCRE6; ISSN:0907-4449. (International Union of Crystallography)MOLREP is an automated program for mol. replacement that utilizes a no. of original approaches to rotational and translational search and data prepn. Since the first publication describing the program, MOLREP has acquired a variety of features that include weighting of the X-ray data and search models, multi-copy search, fitting the model into electron d., structural superposition of two models and rigid-body refinement. The program can run in a fully automatic mode using optimized parameters calcd. from the input data.
- 59Emsley, P.; Lohkamp, B.; Scott, W. G.; Cowtan, K. Features and development of Coot. Acta Crystallogr., Sect. D: Biol. Crystallogr. 2010, 66, 486– 501, DOI: 10.1107/s090744491000749359https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXksFKisb8%253D&md5=67262cbfc60004de5ef962d5c043c910Features and development of CootEmsley, P.; Lohkamp, B.; Scott, W. G.; Cowtan, K.Acta Crystallographica, Section D: Biological Crystallography (2010), 66 (4), 486-501CODEN: ABCRE6; ISSN:0907-4449. (International Union of Crystallography)Coot is a mol.-graphics application for model building and validation of biol. macromols. The program displays electron-d. maps and at. models and allows model manipulations such as idealization, real-space refinement, manual rotation/translation, rigid-body fitting, ligand search, solvation, mutations, rotamers and Ramachandran idealization. Furthermore, tools are provided for model validation as well as interfaces to external programs for refinement, validation and graphics. The software is designed to be easy to learn for novice users, which is achieved by ensuring that tools for common tasks are 'discoverable' through familiar user-interface elements (menus and toolbars) or by intuitive behavior (mouse controls). Recent developments have focused on providing tools for expert users, with customisable key bindings, extensions and an extensive scripting interface. The software is under rapid development, but has already achieved very widespread use within the crystallog. community. The current state of the software is presented, with a description of the facilities available and of some of the underlying methods employed.
- 60Schrodinger, LLC. The PyMOL Molecular Graphics System, Version 2.2.1.There 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/acs.jmedchem.9b00454.
HPLC traces of tested compounds, western blot quantification and normalization IKZF3, concentration response curve for IKZF3 mAb, and Ki values of tested compounds (PDF)
Molecular formula strings (CSV)
Crystal structures have been deposited in the protein data bank (PDB) under the accession codes 6R0S (4a), 6R0V (4b), 6R0U (5a), 6R11 (5b), 6R1X (7a), 6R12 (7b), 6R1K (7c), 6R1D (7d), 6R13 (7f), 6R18 (11a), 6R1C (12a), 6R1W (16b), 6R19 (20a), and 6R1A (20b), 6R0Q (CBG). The authors will release the atomic coordinates and experimental data upon article publication.
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.