Übersicht
 

Publications at google scholar

1-10
11-20
21-30
31-40
41-50
51-60
61-70
71-80
81-90
91-100
101-110
111-120
121-130
131-140
141-150
151-160
161-170
171-180
181-190
191-200
201-210
211-220
221-230
231-240
241-250
251-260
261-270
271-280

 

 

 

Publikationen / Publications


Google scholar :: ORCID :: Web of Science :: Freidok+ :: Scopus :: ChemRxiv
   

1. Synthese von Retinoiden mit Aryltetrahydroanthracenstruktur.
W. Hanefeld, M. Jung; Liebigs Ann. Chem. (1994) 59-64. Link

2. Selektive Synthese ortho-disubstituierter Tetrahydroanthracene mit Retinoidstruktur.
W. Hanefeld, M. Jung; Liebigs Ann. Chem. (1994) 331-336. Link

3. Synthese eines Retinoids mit Acetylaryltetrahydroanthracenstruktur und neuartige Bindungsspaltung eines ortho-Acetyltriflates.
W. Hanefeld, M. Jung; Pharmazie 49 (1994), 18-20.

4. Synthesis of Pentacyclic Retinoids.
W. Hanefeld, M. Jung; Tetrahedron 50 (1994), 2459-2468. Link

5. Dynamic Kinetic Resolution of Racemic α-Halo Acids via a Chiral Imidazolidinone Auxiliary.
J. A. O'Meara, M. Jung, T. Durst; Tetrahedron Lett. 36 (1995), 2559-2562. Link

6. Bioorganometallic Chemistry - Synthesis and Anti-Tumor-Activity of Cobalt Carbonyl Complexes.
M. Jung, D. E. Kerr, P. D. Senter; Arch. Pharm. Pharm. Med. Chem. 330 (1997), 173-176. Link

7. Analogues of Trichostatin A and Trapoxin B as Histone Deacetylase Inhibitors.
M. Jung, K. Hoffmann, G. Brosch, P. Loidl; Bioorg. Med. Chem. Lett. 7 (1997), 1655-1658. Link

8. Synthesis and Biological Activity of β-Ionone-derived Alcohols for Cancer Chemoprevention.
M. Jung, H. Mo, C. E. Elson; Anticancer Res. 18 (1998), 189-192.

9. Enantioselective Synthesis of β-Dibenzylamino Alcohols via a Dynamic Kinetic Resolution of α-Halo Acids.
J. A. O'Meara, N. Gardee, M. Jung, R. N. Ben, T. Durst; J. Org. Chem. 63 (1998), 3117-3119. Link

10. A Non-Isotopic Assay for Histone Deacetylase Activity.
K. Hoffmann, G. Brosch, P. Loidl, M. Jung; Nucleic Acids Res. 27 (1999), 2057-2058. Link

<

11. Inhibitors of histone deacetylase suppress the growth of MCF-7 breast cancer cells.
K. Schmidt, R. Gust, M. Jung, Arch. Pharm. Pharm. Med. Chem. 332 (1999), 353-357. Link

12. Amide Analogues of Trichostatin A as Inhibitors of Histone Deacetylase and Inducers of Terminal Cell Differentiation.
M. Jung, G. Brosch, D. Kölle, H. Scherf, C. Gerhäuser, P. Loidl; J. Med. Chem. 42 (1999), 4669-4679. Link

13. Synthesis and Activity of Fluorinated Derivatives of Sulindac Sulphide and Sulindac Sulphone.
M. Jung, A. F. Wahl, W. Neupert, G. Geisslinger, P. D. Senter; Pharm. Pharmacol. Commun. 6 (2000), 217-221. Link

14. Farnesyl Anthranilate Suppresses the Growth, in vitro and in vivo, of Murine B16 Melanoma.
H. Mo, D. Tatman, M. Jung, C. E. Elson; Cancer Lett. 157 (2000), 145-153. Link

15. First Non-Radioactive Assay for In-vitro Screening of Histone Deacetylase Inhibitors.
K. Hoffmann, G. Brosch, P. Loidl, M. Jung; Pharmazie 55 (2000), 601-606.

16. Acetylenehexacarbonyldicobalt complexes, a Novel Class of Antitumor Drugs.
K. Schmidt, M. Jung, R. Keilitz, B. Schnurr, R. Gust; Inorg. Chim. Acta 306 (2000), 6-16. Link

17. Hemmstoffe der Histon-Deacetylase als neue Wirkstoffe für Prävention und Therapie von Krebserkrankungen.
M. Jung; Pharm. Unserer Zeit 29 (2000), 385-388. (Review) Link

18. Fluorescence-labeled Octapeptides as Substrates for Histone Deacetylase.
K. Hoffmann, R. M. Söll, A. G. Beck-Sickinger, M. Jung; Bioconjugate Chem. 12 (2001), 51-55. Link

19. Improvement and Validation of the Fluorescence-based Histone Deacetylase Assay using an Internal Standard.
K. Hoffmann, B. Heltweg, M. Jung; Arch. Pharm. Pharm. Med. Chem. 334 (2001), 248-252. Link

20. Inhibitors of Histone Deacetylase as New Anticancer Agents.
M. Jung; Curr. Med. Chem. 8 (2001), 1505-1511. (Review) Link

<

21. Antibody Directed Enzyme Prodrug Therapy (ADEPT) and related approaches for anticancer therapy.
M. Jung; Mini Rev. Med. Chem. 1 (2001), 399-407 (Review). Link

22. Acyclic Isoprenoid Ether Derivatives as Chemotherapeutics.
Wisconsin Alumni Research Foundation: C. E. Elson, M. Jung, H. Mo; WO 0205856 (24.01.2002). Link

23. Inhibitors of Human Histone Deacetylase: Synthesis, Enzyme and Cellular Activity of Straight Chain Hydroxamates.
S. W. Remiszewski, L. C. Sambuccetti, P. Atadja, K. W. Bair, W. D. Cornell, M. A. Green, K. L. Howell, M. Jung, P. Kwon, N. Trogani, H. Walker; J. Med. Chem. 45 (2002), 753-757. Link

24. A Microplate Reader-Based Nonisotopic Histone Deacetylase Activity Assay.
B. Heltweg, M. Jung; Anal. Biochem. 302 (2002), 175-183. Link

25. Histone Deacetylases.
M. Jung; in: Targets for Cancer Chemotherapy: Transcription Factors and Other Nuclear Proteins (Eds.: N. B. La Thangue, L. R. Bandara) (2002) 123-144. (Book Chapter)

26. Structure-Activity Relationships on Phenylalanine Containing Inhibitors of Histone Deacetylase: In-Vitro Enzyme Inhibition, Induction of Differentiation and Inhibition of Proliferation in Friend Leukemic Cells.
S. Wittich, H. Scherf, C. Xie, G. Brosch, P. Loidl, C. Gerhäuser, M. Jung; J. Med. Chem. 45 (2002), 3296-3309. Link

27. Comparative Action of Cobalt Carbonyl Complexes on Cancer Cells using Human Tumor Xenografts.
T. Roth, C. Eckert, H.-H. Fiebig, M. Jung; Anticancer Res. 22 (2002), 2281-2284.

28. Eosin Y as an internal standard for a plate reader based quantitation of a histone deacetylase substrate.
B. Heltweg, M. Jung; Arch. Pharm. Pharm. Med. Chem. 335 (2002), 296-300. Link

29. A homogeneous non-isotopic histone deacetylase activity assay.
B. Heltweg, M. Jung; J. Biomol. Screen. 8 (2003), 89-95. Link

30. Hydroxamide derivatives of short-chain fatty acids are potent inducers of fetal hemoglobin production.
E. Skarpidi, H. Cao, B. Heltweg, B. F. White, R. L. Marhenke, M. Jung, G. Stamatoyannopoulos; Exp. Hematol. 31 (2003), 197-203. Link

<

31. Trichostatin A and structurally related histone deacetylase inhibitors induce 5-LO promoter activity in transient reporter gene assays.
N. Klan, S. Seuter, N. Schnur, M. Jung, D. Steinhilber; Biol. Chem. 384 (2003), 777-785. Link

32. Nonisotopic substrate for assaying both human zinc and NAD+-dependent histone deacetylases.
B. Heltweg, F. Dequiedt, E. Verdin, M. Jung; Anal. Biochem. 319 (2002), 42-48. Link

33. Homogeneous assays for histone deacetylase activity.
M. Jung; Exp. Opin. Ther. Patents 13 (2003), 935. (Review) Link

34. Methods for the use of inhibitors of histone deacetylase as synergistic agents in cancer therapy.
M. Jung, M. Jung, A. Dritschilo; WO 2004064727 (05.08.2004) Link

35. Farnesyl-O-acetylhydroquinone and geranyl-O-acetylhydroquinone suppress the proliferation of murine B16 melanoma cells, human prostate and colon adenocarcinoma cells, human lung carcinoma cells, and human leukemia cells.
J. A. McAnally, M. Jung, H. Mo; Cancer Lett. 202 (2003), 181-192. Link

36. Identification of selective inhibitos of NAD+-dependent deacetylases using phenotypic screening in yeast.
M. Hirao, J. Posakony, M. Nelson, H. Hruby, M. Jung, J. A. Simon, A. Bedalov; J. Biol. Chem. 278 (2003), 52773-52783. Link

37. Induction of human γ-globin gene expression by histone deacetylase inhibitors.
H. Cao, G. Stamatoyannopoulos, M. Jung; Blood 103 (2004), 701-709. Link

38. Enhancement of radiation sensitivity of human squamous carcinoma cells by histone deacetylase inhibitors.
Y. Zhang, M. Jung, A. Dritschilo, M. Jung; Radiat. Res. 161 (2004), 667-674. Link

39. Subtype selective substrates for histone deacetylases.
B. Heltweg, F. Dequiedt, B. L. Marshall, C. Brauch, M. Yoshida, N. Nishino, E. Verdin, M. Jung; J. Med. Chem. 47 (2004), 5235-5243. Link

40. SIRT1 regulates HIV transcription via Tat deacetylation.
S. Pagans, A. Pedal, B. J. North, K. Kaehlcke, B. L. Marshall, A. Dorr, C. Hetzer-Egger, P. Henklein, R. Frye, M. W. McBurney, H. Hruby, M. Jung, E. Verdin, M. Ott; PLoS Biology; 3 (2005) e41. Link (open access)

<

41. Effect of inhibitors of histone deacetylase on the induction of cell differentiation in murine and human erythroleukemia cell lines.
S. Wittich, H. Scherf, C. Xie, B. Heltweg, F. Dequiedt, E. Verdin, C. Gerhäuser, M. Jung; Anti-Cancer Drugs 16 (2005), 635-643. Link

42. In vitro assays for the determination of histone deacetylase activity.
B. Heltweg, J. Trapp, M. Jung; Methods 36 (2005), 332-337. Link

43. Chromatin modifications as targets for new anticancer drugs.
S. Schäfer, M. Jung; Arch. Pharm. Chem. Life Sci. 338 (2005), 347-357. (Review) Link

44. Hydroxamide derivatives of short chain fatty acids have erythropoietic activity and induce γ gene expression in vivo.
H. Cao, M. Jung, G. Stamatoyannopoulos; Exp. Hematol. 33 (2005), 1443-1449. Link

45. Software-supported quantification of angiogenesis in an in vitro culture system. Examples of application in studies of basic research, biocompatibility and drug discovery.
K. Peters, H. Schmidt, G. Kamp, R. E. Unger, B. Hoffmann, F. Stenzel, G. Ziegler, M. Jung, S. Stumpf, C. J. Kirkpatrick; in: Trends in Angiogenesis Research (Ed. R. V. Zubar) (2005) 103-123. (Book Chapter) Link

46. Adenosine mimics as inhibitors of NAD+-dependent histone deacetylases - from kinase to sirtuin inhibitors.
J. Trapp, A. Jochum, R. Meier, L. Saunders, B. Marshall, E. Verdin, P. Goekjian, C. Kunick, W. Sippl, M. Jung; J. Med. Chem. 49 (2006) 7307-7316. Link

47. The role of NAD+ Dependent Histone Deacetylases (sirtuins) in Ageing.
J. Trapp; Curr. Drug Targets 7 (2006), 1553-1560. (Review) Link

48. Inhibitors of histone deacetylases as potential therapeutic tools for high-risk embryonal tumors of the nervous system of childhood.
S. E. Furchert, C. Lanvers-Kaminsky, H. Jürgens, M. Jung, A. Loidl, M. C. Frühwald; Int. J. Cancer 120 (2007), 1787-1794. Link

49. Target-based approach to inhibitors of histone arginine methyltransferases.
A. Spannhoff, R. Heinke, I. Bauer, P. Trojer, E. Metzger, R. Gust, R. Schüle, G. Brosch, W. Sippl, M. Jung; J. Med. Chem. 50 (2007), 2319-2325. Link

50. Determination of drug-serum protein interactions via fluorescence polarization measurements.
U. Mathias, M. Jung; Anal. Bioanal. Chem. 388 (2007), 1147-1156. Link

<

51. A novel arginine methyltransferase inhibitor with cellular activity.
A. Spannhoff, R. Machmur, R. Heinke, P. Trojer, I. Bauer, G. Brosch, R. Schüle, W. Hanefeld, W. Sippl, M. Jung; Bioorg. Med. Chem. Lett. 17 (2007), 4150-4153. Link

52. Structure activity studies on suramin analogues as inhibitors of NAD+-dependent histone deacetylases (sirtuins).
J. Trapp, R. Meier, D. Hongwiset, M. U. Kassack, W. Sippl, M. Jung; ChemMedChem. 2 (2007), 1419-1431. Link

53. Characterization of novel inhibitors of histone acetyltransferases.
E. D. Eliseeva, V. Valkov, M. Jung, M. Jung; Mol. Cancer Ther. 6 (2007), 2391-2398. Link

54. Development of Albumin-binding Camptothecin Prodrugs Using a Peptide Positional Scanning Library.
B. Schmid, A. Warnecke, I. Fichtner, M. Jung, F. Kratz; Bioconjugate Chem. 18 (2007), 1786-1799. Link

55. R306465 is a novel potent inhibitor of class I Histone Deacetylases (HDACs) with broad-spectrum antitumoral acivity against solid and haematological malignancies.
J. Arts, P. Angibaud, A. Mariën, W. Floren, B. Janssens, J. van Dun, L. Janssen, T. Geerts, R. W. Tuman, D. L. Johnson, L. Andries, M. Jung, M. Janicot, K. van Emelen; Br. J. Cancer 97 (2007), 1344-1353. Link

56. In Vivo Application of Histone Deacetylase Inhibitor Trichostatin-A Impairs Murine Male Meiosis.
I. Fenic, H. M. Hossain, V. Sonnack, S. Tchatalbachev, F. Thierer, J. Trapp, K. Failing, K. S. Edler, M. Bergmann, M. Jung, T. Chakraborty, K. Steger; J. Androl. 29 (2008), 172-185 Link

57. Phenylalanine-containing hydroxamic acids as selective inhibitors of class IIb histone deacetylases (HDACs).
S. Schäfer, L. Saunders, E. Eliseeva, A. Velena, A. Strasser, M. Jung, A. Schwienhorst, A. Strassser, A. Dickmanns, R. Ficner, E. Verdin, M. Jung; Bioorg. Med. Chem. 16 (2008), 2011-2033. Link

58. Biphenylalkylacetylhydroquinone ethers suppress the proliferation of murine B16 melanoma cells.
N. Fernandes, M. Jung, A. Daoud, H. Mo; Anticancer Res. 28 (2008), 1005-1012. Link

59. Structure activity studies on splitomicin derivatives as sirtuin inhibitors and computational prediction of binding mode.
R. C. Neugebauer, U. Uchiechowska, R. Meier, H. Hruby, V. Valkov, E. Verdin, W. Sippl, M. Jung; J. Med. Chem. 51 (2008) 1203-1213. Link

60. Inhibitors of NAD+ Dependent Histone Deacetylases (Sirtuins).
M. Jung, R. C. Neugebauer, W. Sippl; Curr. Pharm. Des. 14 (2008) 562-573. Link

<

61. Destabilization of ERBB2 transcripts by targeting 3' UTR mRNA associated HuR and histone deacetylase-6 (HDAC6).
G. K. Scott, C. Marx, C. E. Berger, L. R. Saunders, E. Verdin, S. Schäfer, M. Jung, C. C. Benz; Mol. Cancer Res. 6 (2008), 1250-1258. Link

62. Targeting Epigenetic Mechanisms: Potential of Natural Products in Cancer Chemoprevention.
A.-T. Hauser, M. Jung; Planta Med. 74 (2008), 1593-1601. Link

63. Thiobarbiturates as Sirtuin Inhibitors: Virtual Screening, Free Energy Calculations and Biological Testing.
U. Uciechowska, J. Schemies, R. C. Neugebauer, E.-M. Huda, M. L. Schmitt, R. Meier, E. Verdin, M. Jung, W. Sippl; ChemMedChem. 3 (2008) 1965-1976. Link

64. Pyridylalanine-containing hydroxamic acids as selective HDAC6 inhibitors.
S. Schäfer, L. Saunders, S. Schlimme, V. Valkov, J. M. Wagner, F. Kratz, W. Sippl, E. Verdin, M. Jung; ChemMedChem. 4 (2009) 283-290. Link

65. Cancer treatment of the future: inhibitors of histone methyltransferases.
A. Spannhoff, W. Sippl, M. Jung; Int. J. Biochem. Cell Biol. 41 (2009), 4-11. (Review) Link

66. Histone deacetylase inhibitor, Trichostatin A, affects gene expression patterns during morphogenesis of chick limb buds in vivo.
W. Zhao, F. Dai, A. Bonafede, S. Schäfer, M. Jung, F. Yusuf, A. J. Gamel, J. Wang, B. Brand-Saberi; Cells Tissues Organs 190 (2009) 121-134. Link

67. Virtual Screening and Biological Characterisation of Novel Histone Arginine Methyltransferase PRMT1 Inhibitors.
R. Heinke, A. Spannhoff, R. Meier, P. Trojer, I. Bauer, M. Jung, W. Sippl; ChemMedChem. 4 (2009), 69-77. Link

68. Assays for Histone Deacetylases.
A.-T. Hauser, M. Jung, M. Jung; Curr. Top. Med. Chem. 9 (2009), 227-234. (Review) Link

69. Suberoylanilide hydroxamic acid - a potential epigenetic therapeutic agent for lung fibrosis?
Z. Wang, C. Chen, S. N. Finger, S. d/o Kwajah M. M, M. Jung, H. Schwarz, N. Swanson, R. R. Lareu, M. Raghunath; Eur. Resp. J. 34 (2009) 145-155. Link

70. Histone deacetylase inhibitors that target tubulin.
J. Schemies, W. Sippl, M. Jung; Cancer Lett. 280 (2009) 222-232. (Review) Link

<

71. The ergot alkaloid gene cluster: functional analyses and evolutionary aspects.
N. Lorenz, T. Haarmann, S. Pažoutová, M. Jung, P. Tudzynski; Phytochemistry 70 (2009) 1822-1832. Link

72. Structural and Synthetic Investigations of Tanikolide Dimer, a SIRT2 Selective Inhibitor, and Tanikolide seco-Acid from the Madagascar Marine Cyanobacterium Lyngbya majuscula.
M. Gutiérrez, E. H. Andrianasolo, W. K. Shin, D. E. Goeger, A. Yokochi, J. Schemies, M. Jung, D. France, S. Cornell-Kennon, E. Lee, W. H. Gerwick; J. Org. Chem. 74 (2009) 5267-5275. Link

73. Determination of the antifungal agent posaconazole in human serum by HPLC with parallel column-switching technique.
W. Neubauer, A. König, R. Bolek, R. Trittler, M. Engelhardt, M. Jung, K. Kümmerer; J. Chromatogr. B 877 (2009) 2493-2498. Link

74. The Emerging Therapeutic Potential of Histone Methyltransferase and Demethylase Inhibitors.
A. Spannhoff, A.-T. Hauser, R. Heinke, W. Sippl, M. Jung; ChemMedChem. 4 (2009) 1568-1582. (Review) Link

75. NAD+-dependent histone deacetylases (sirtuins) as new drug targets.
J. Schemies, U. Uciechowska, W. Sippl, M. Jung; Med. Res. Rev. 30 (2010) 861-889. (Review) Link

76. Novel 3-Arylidene-indolin-2-ones as inhibitors of NAD+-dependent histone deacetylases (sirtuins).
K. Huber, J. Schemies, U. Uciechowska, J. M. Wagner, T. Rumpf, F. Lewrick, R. Süss, W. Sippl, M Jung, F. Bracher; J. Med. Chem. 53 (2010) 1383-1386. Link

77. Arginine methylation of the B cell antigen receptor promotes differentiation.
S. Infantino, B. Benz, T. Waldmann, M. Jung, R. Schneider, M. Reth; J. Exp. Med. 207 (2010) 711-719. Link

78. Malyngolide Dimer, a Bioactive Symmetric Cyclodepside from the Panamanian Marine Cyanobacterium Lyngbya majuscula.
M. Gutiérrez, K. Tidgewell, T. L. Capson, N. Engene, A. Almanza, J. Schemies, M. Jung, W. H. Gerwick; J. Nat. Prod. 73 (2010) 709-711. Link

79. Identification of Tri- and Tetracyclic Pyrimidindiones as Novel Sirtuin Inhibitors.
D. Rotili, A. Mai, D. Tarantino, V. Carafa, E. Lara, S. Meade, G. Botta, A. Nebbioso, J. Schemies, M. Jung, A. G. Kazantsev, M. Esteller, M. F. Fraga, L. Altucci; ChemMedChem. 5 (2010) 674-677. Link

80. Therapeutic drug monitoring of posaconazole in hematological patients - experience with a new HPLC based method.
W. Neubauer, M. Engelhardt, A. König, S. Hieke, M. Jung, H. Berz, K. Kümmerer; Antimicrobial Agents Chemother. 54 (2010) 4029-4032. Link

<

81. Schiff Bases Derived from p-Aminobenzyl Alcohol as Trigger Groups for pH-Dependent Prodrug Activation.
I. A. Müller, F. Kratz, M. Jung, A. Warnecke; Tetrahedron Lett. 51 (2010) 4371-4374. Link

82. Diminished Sensitivity of Chronic Lymphocytic Leukemia Cells to ABT-737 and ABT-263 Due to Albumin Binding in Blood.
M. Vogler, S. D. Furdas, M. Jung, M. J. S. Dyer, G. M Cohen; Clin. Cancer Res. 16 (2010) 4217-4225. Link

83. Targeting epigenetic modifiers: Inhibitors of histone methyltransferases.
E.-M. Bissinger, R. Heinke, W. Sippl, M. Jung; Med. Chem. Commun. 1 (2010) 114-124. (Review) Link

84. Inhibitors to understand molecular mechanisms of NAD+-dependent deacetylases (sirtuins).
M. R. Lawson, U. Uciechowska, J. Schemies, T. Rumpf, M. Jung, W. Sippl; Biochem. Biophys. Acta 1799 (2010) 726-739. (Review) Link

85. No taste for methyl: methylation sensitive proteolysis in drug screening.
E.-M. Bissinger, M. Jung; Chem. Biol. 17 (2010) 677-678. (Invited commentary) Link

86. From cosubstrate similarity to inhibitor diversity - inhibitors of ADP-ribosyltransferases from kinase inhibitor screening.
B. Maurer, U. Mathias, P. Papatheodorou, S. Shekfeh, J. Orth, T. Jank, C. Schwan, W. Sippl, K. Aktories, M. Jung; Mol. BioSyst. 7 (2011) 799-808. Link

87. Histone deacetylase (HDAC) inhibitors in recent clinical trials for cancer therapy.
J. M. Wagner, B. Hackansson, M. Lübbert, M. Jung; Clin. Epigenet. 1 (2010) 117-136. (Review) Link (open access)

88. Synthesis and biological activity of splitomicin analogs targeted at human NAD+-dependent histone deacetylases (sirtuins).
M. Freitag, J. Schemies, T. Larsen, K. El Gaghlab, F. Schulz, T. Rumpf, M. Jung, A. Link; Bioorg. Med. Chem. 19 (2011) 3669-3677. Link

89. Synthesis and biological testing of novel pyridoisothiazolones as histone acetyltransferase inhibitors.
S. D. Furdas, S. Shekfeh, E.-M. Bissinger, J. M. Wagner, S. Schlimme, V. Valkov, M. Hendzel, M. Jung, W. Sippl; Bioorg. Med. Chem. 19 (2011) 3678-3689. Link

90. Acyl Derivatives of p-Aminosulfonamides and Dapsone as new Inhibitors of the Arginine Methyltransferase hPRMT1.
E.-M. Bissinger, R. Heinke, A. Spannhoff, A. Eberlin, E. Metzger, V. Cura, P. Hassenboehler, J. Cavarelli, R. Schüle, M. T. Bedford, W. Sippl, M. Jung; Bioorg. Med. Chem. 19 (2011) 3717-3731. Link

<

91. Carbamate prodrug concept for hydroxamate HDAC inhibitors.
S. Schlimme, A.-T. Hauser, V. Carafa, R. Heinke, D. A. Stolfa, S. Cellamare, A. Carotti, L. Altucci, M. Jung, W. Sippl; ChemMedChem. 6 (2011) 1193-1198. Link

92. Two-Step Synthesis of Novel, Bioactive Derivatives of the Ubiquitous Cofactor Nicotinamide Adenine Dinucleotide (NAD).
T. Pesnot, J. Kempter, J. Schemies, G. Pergolizzi, U. Uciechowska, T. Rumpf, W. Sippl, M. Jung, G. K. Wagner; J. Med. Chem. 54 (2011) 3492-3499. Link

93. Computer- and structure-based lead design for epigenetic targets.
R. Heinke, L. Carlino, S. Kannan, M. Jung, W. Sippl; Bioorg. Med. Chem. 19 (2011) 3605-3615. (Review) Link

94. Epigenetik - Lesezeichen im Buch des Lebens.
J. M. Wagner, M. Jung; q&more 1 (2011) 6-11. (Review)

95. Epigenetic Drug Discovery Special Issue.
W. Sippl, M. Jung; Bioorg. Med. Chem. 19 (2011) 3603-3604. (Editorial) Link

96. Chemical probes: Sharpen your epigenetic tools.
A.-T. Hauser, M. Jung; Nat. Chem. Biol. 7 (2011) 499-500. (Invited Commentary) Link

97. A Bioluminogenic HDAC Activity Assay - Validation and Screening.
F. Halley, J. Reinshagen, M. Wolf, B. Ellinger, A. L. Niles, N. J. Evans, T. Kirkland, J. M. Wagner, M. Jung, P. Gribbon, S. Gul; J. Biomol. Screen. 16 (2011) 1227-1235. Link

98. Chemische Epigenetik - Lesezeichen im Buch des Lebens.
K. Keller, M. Jung; Nachr. Chem. 59 (2011) 822-827. (Review) Link

99. Steroid boost for sirtuin research.
T. Rumpf, M. Jung; Aging 3 (2011) 818. (Invited Commentary) Link

100. Oxidierte Cytosin-Derivate - der Schlüssel zur aktiven DNA-Demethylierung?
K. I. Ladwein, M. Jung; Angew. Chem. 123 (2011) 12347-12349. Link
Oxidized Cytosine Metabolites Offer a Fresh Perspective for Active DNA Demethylation
K. I. Ladwein, M. Jung; Angew. Chem. Int. Ed. 50 (2011) 12143-12145. Link

<

101. Small Molecule Inhibitors of Histone Acetyltransferases as Epigenetic Tools and Drug Candidates.
S. D. Furdas, S. Kannan, W. Sippl, M. Jung; Arch. Pharm. Chem. Life Sci. 345 (2012) 7-21. (Review) Link

102. Screening Assays for Epigenetic Targets using native Histones as Substrates.
A.-T. Hauser, E.-M. Bissinger, E. Metzger, A. Repenning, U.-M. Bauer, A. Mai, R.Schüle, M. Jung; J. Biomol. Screen. 17 (2012) 18-26. Link

103. Inhibition of bromodomain-mediated protein-protein interactions as a novel therapeutical strategy.
S. D. Furdas, L. Carlino, W. Sippl, M. Jung; Med. Chem. Commun. 3 (2012) 123-134. (Review) Link

104. Binding free energy calculations and biological testing of novel thiobarbiturates as inhibitors of the human NAD+ dependent histone deacetylase Sirt2.
U. Uciechowska, J. Schemies, M. Scharfe, M. Lawson, K. Wichapong, M. Jung, W. Sippl; Med. Chem. Commun. 3 (2012) 167-173. Link

105. Rhodanine carboxylic acids as novel inhibitors of histone acetyltransferases.
S. D. Furdas, S. Shekfeh, S. Kannan,W. Sippl, M. Jung; Med. Chem. Commun. 3 (2012) 305-311. Link

106. The discovery of novel and highly selective 10,11-dihydro-5H-dibenz[b,f]azepine SIRT2 inhibitors.
P. Di Fruscia, K.-K. Ho, S. Laohasinnarong, M. Khongkow, S. H. B. Kroll, S. A. Islam, M. J. E. Sternberg, K. Schmidtkunz, M. Jung, E. W.-F. Lam, M. J. Fuchter; Med. Chem. Commun. 3 (2012) 373-378. Link

107. Overexpression of transcription factor NF-E2: a novel murine model of myelo-proliferative disorders.
K. B. Kaufmann, A. Gründer, T. Hadlich, J. Wehrle, M. Gothwal, R. Bogeska, J. M. Wagner, M. Jung, T. Günther, R. Schüle, A. Rambaldi, H. L. Pahl; J. Exp. Med. 209 (2012) 35-50. Link

108. Impairment of prostate cancer cell growth by a selective and reversible LSD1 inhibitor.
D. Willmann, S. Lim, S. Wetzel, E. Metzger, A. Jandausch, W. Wilk, M. Jung, I. Forne, A. Imhof, A. Janzer, J. Kirfel, H. Waldmann, R. Schüle, R. Buettner; Int. J. Cancer 131 (2012) 2704-2709. Link

109. Lestaurtinib Inhibits Histone Phosphorylation and Androgen-dependent Gene Expression in Prostate Cancer Cells.
J. Köhler, G. Erlenkamp, A. Eberlin, T. Rumpf, I. Slynko, E. Metzger, R. Schüle, W. Sippl, M. Jung; PLoS ONE 7 (2012) e34973. Link (open access)

110. HDAC6 as a target for antileukemic drugs in acute myeloid leukemia.
B. Hackanson, L. Rimmele, M. Benkißer, M. Abdelkarim, M. Fliegauf, M. Jung, M. Lübbert; Leukemia Res. 36 (2012) 1055-1062. Link

<

111. Design, synthesis and biological evaluation of 2-aminobenzanilide derivatives as potent and selective HDAC inhibitors.
D. A. Stolfa, A. Stefanachi, J. M. Gajer (née Wagner), A. Nebbioso, L. Altucci, S. Cellamare, M. Jung, A. Carotti; ChemMedChem. 7 (2012) 1256-1266. Link

112. New lysine methyltransferase drug targets in cancer.
T. Wagner, M. Jung; Nat. Biotechnol. 30 (2012) 622-623. (Review) Link

113. Use of systemic antifungals in daily clinical practice in the haematology and oncology setting: results of a prospective observational analysis.
B. Metzke, W. C. Neubauer, S. Hieke, M. Jung, R. Wäsch, M. Engelhardt; Pharmacoepidemiol. Drug Saf. 21 (2012) 953-963. Link

114. High-throughput 1,536-Well Fluorescence Polarization Assays for α1-Acid Glycoprotein and Human Serum Albumin Binding.
A. Yasgar, S. D. Furdas, D. J. Maloney, A. Jadhav, M. Jung, A. Simeonov; PLoS ONE 7 (2012) e45594. Link (open access)

115. Benzodeazaoxaflavins as Potent Sirtuin Inhibitors with Antiproliferative Properties in Cancer Stem Cells.
D. Rotili, D. Tarantino, V. Carafa, C. Paolini, J. Schemies, M. Jung, G. Botta, S. Di Maro, E. Novellino, C. Steinkuhler, R. De Maria, P. Gallinari, L. Altucci, A. Mai; J. Med. Chem. 55 (2012) 8193-8197. Link

116. The role of histone demethylases in cancer therapy.
I. Hoffmann, M. Roatsch, M. L. Schmitt, L. Carlino, M. Pippel, W. Sippl, M. Jung; Mol. Oncol. 6 (2012) 683-703. (Review) Link

117. Inhibitors of the NAD+-Dependent Protein Desuccinylase and Demalonylase Sirt5.
B. Maurer, T. Rumpf, M. Scharfe, D. A. Stolfa, M. L. Schmitt, W. He, E. Verdin, W. Sippl, M. Jung; ACS Med. Chem. Lett. 3 (2012) 1050-1053. Link

118. Current trends in epigenetic drug discovery.
D. A. Stolfa, O. Einsle, W. Sippl, M. Jung; Future Med. Chem. 4 (2012) 2029-2037. (Review) Link

119. Nonradioactive In Vitro Assays for Histone Deacetylases.
A.-T. Hauser, J. M. Gajer (née Wagner), M. Jung; Meth. Mol. Biol. 981 (2013) 211-227. (Review) Link

120. The histone deacetylase inhibitor SAHA acts in synergism with fenretinide and doxorubicin to control growth of rhabdoid tumor cells.
K. Kerl, D. Ries, R. Unland, C. Borchert, N. Moreno, M. Hasselblatt, H. Jürgens, M. Kool, D. Görlich, M. Eveslage, M. Jung, M. Meisterernst, M. Frühwald; BMC Cancer 13 (2013) 286. Link

<

121. Posaconazole Therapeutic Drug Monitoring in the Real-life Setting: A Single-Center Experience and Review of the Literature.
B. N. Gross, G. Ihorst, M. Jung, R. Wäsch, M. Engelhardt; Pharmacotherapy 33 (2013) 1117-1125. Link

122. The nephronophthisis gene product NPHP2/Inversin interacts with Aurora A and interferes with HDAC6-mediated cilia disassembly.
M. Mergen, C. Engel, B. Müller, M. Follo, T. Schäfer, M. Jung, G. Walz; Nephrol. Dial. Transplant. 28 (2013) 2744-2753. Link

123. Structural Basis for the Inhibition of HDAC8, a Key Epigenetic Player in the Blood Fluke Schistosoma mansoni.
M. Marek, S. Kannan, A.-T. Hauser, M. Moraes Mourão, S. Caby, V. Cura, D. A. Stolfa, K. Schmidtkunz, J. Lancelot, J.-P. Renaud, G. Oliveira, W. Sippl, M. Jung, J. Cavarelli, R. J. Pierce, C. Romier; PLoS Pathog. 9 (2013) e1003645. Link (open access)

124. Schistosoma mansoni sirtuins: characterization and potential as chemotherapeutic targets.
J. Lancelot, S. Caby, F. Dubois-Abdesselem, M. Vanderstraete, J. Trolet, G. Oliveira, F. Bracher, M. Jung, R. J. Pierce; PLoS Negl. Trop. Dis. 7 (2013) e2428. Link (open access)

125. Non-peptidic propargylamines as inhibitors of lysine specific demethylase 1 (LSD1) with cellular activity.
M. L. Schmitt, A.-T. Hauser, L. Carlino, M. Pippel, J. Schulz-Fincke, E. Metzger, D. Willmann, T. Yiu, M. Barton, R. Schüle, W. Sippl, M. Jung; J. Med. Chem. 56 (2013) 7334-7342. Link

126. 4-Acyl Pyrroles Mimic Acetylated Lysines in Histone Code Reading.
X. Lucas, D. Wohlwend, M. Hügle, K. Schmidtkunz, S. Gerhardt, R. Schüle, M. Jung, O. Einsle, and S. Günther; Angew. Chem. Int. Ed. 52 (2013) 14055-14059. Link

127. Histone Deacetylase (HDAC) Inhibitors in Recent Clinical Trials for Cancer Therapy.
K. Keller, M. Jung; in: Epigenetic Therapy of Cancer (Eds.: M. Lübbert, P. A. Jones) (2014) 227-255. (Book Chapter). Link

128. Total Synthesis of (18S)- and (18R)-Homolargazole by Rhodium-Catalyzed Hydrocarboxylation.
C. Schotes, D. Ostrovskyi, J. Senger, K. Schmidtkunz, M. Jung, B. Breit; Chem. Eur. J. 20 (2014) 2164-2168. Link

129. Virtual screening of PRK1 inhibitors: ensemble docking, rescoring using binding free energy calculation and QSAR model development.
I. Slynko, M. Scharfe, T. Rumpf, J. Eib, E. Metzger, R. Schüle, M. Jung, W. Sippl; J. Chem. Inf. Model. 54 (2014) 138-150. Link

130. Mind the Methyl: Methyllysine Binding Proteins in Epigenetic Regulation.
T. Wagner, D. Robaa, W. Sippl, M. Jung; ChemMedChem. 9 (2014) 466-483. (Review) Link

<

131. BAG3 induction is required to mitigate proteotoxicity via selective autophagy following inhibition of constitutive protein degradation pathways.
F. Rapino, M. Jung, S. Fulda; Oncogene 33 (2014) 1713-1724. Link

132. Heterogeneous antibody based activity assay for lysine specific demethylase 1 (LSD1) on a histone peptide substrate.
M. L. Schmitt, K. I. Ladwein, L. Carlino, J. Schulz-Fincke, D. Willmann, E. Metzger, P. Schilcher, A. Imhof, R. Schüle, W. Sippl, M. Jung; J. Biomol. Screen. 19 (2014) 973-978. Link

133. Limited efficacy of specific HDAC inhibition in urothelial cancer cells.
L. Rosik, G. Niegisch, U. Fischer, M. Jung, W. A. Schulz, M. J. Hoffmann; Cancer Biol. Ther. 15 (2014) 742-757. Link

134. Molecular basis for the anti-parasitic activity of a mercaptoacetamide derivative that inhibits histone deacetylase 8 (HDAC8) from the human pathogen Schistosoma mansoni.
D. A. Stolfa, M. Marek, J. Lancelot, A.-T. Hauser, A. Walter, E. Leproult, J. Melesina, T. Rumpf, J.-M. Wurtz, J. Cavarelli, W. Sippl, R. J. Pierce, C. Romier, M. Jung; J. Mol. Biol. 426 (2014) 3442-3453. Link (open access)

135. Epigenetic Changes Modulate Schistosome Egg Formation and Are a Novel Target for Reducing Transmission of Schistosomiasis.
V. C. Carneiro, I. C. de Abreu da Silva, E. J. Lopes Torres, S. Caby, J. Lancelot, M. Vanderstraete, S. D. Furdas, M. Jung, R. J. Pierce, M. R. Fantappié; PLoS Pathog. 10 (2014) e1004116. Link (open access)

136. Pyrido- and benzisothiazolones as inhibitors of histone acetyltransferases (HATs).
S. D. Furdas, I. Hoffmann, D. Robaa, B. Herquel, W. Malinka, P. Swiatek, A. Akhtar, W. Sippl, M. Jung; Med. Chem. Commun. 5 (2014) 1856-1862. Link (open access)

137. Hydroxamates of para-Aminobenzoic acid as selective inhibitors of HDAC8.
U. Kulandaivelu, L. M. Chilakamari, S. S. Jadav, T. R. Rao, K. N. Jayaveera, B. Shireesha, A.-T. Hauser, J. Senger, M. Marek, C. Romier, M. Jung, V. Jayaprakash; Bioorg. Chem. 57 (2014) 116-120. Link

138. Chromo-pharmacophores: Photochromic diarylmaleimide inhibitors for sirtuins.
C. Falenczyk, M. Schiedel, B. Karaman, T. Rumpf, N. Kuzmanovic, M. Grøtli, W. Sippl, M. Jung, B. König; Chem. Sci. 5 (2014) 4794-4799. Link

139. Discovery of Inhibitors of Schistosoma Mansoni HDAC8 by Combining Homology Modeling, Virtual Screening and in Vitro Validation.
S. Kannan, J. Melesina, A.-T. Hauser, A. Chakrabarti, T. Heimburg, K. Schmidtkunz, A. Walter, M. Marek, R. J. Pierce, C. Romier, M. Jung, W. Sippl; J. Chem. Inf. Model. 54 (2014) 3005-3019. Link

140. Inhibition of p300 suppresses growth of breast cancer. Role of p300 subcellular localization.
M. E. Fermento, N. A. Gandini, D. G. Salomón, M. J. Ferronato, C. A. Vitale, J. Arévalo, A. López Romero, M. Nuñez, M. Jung, M. M. Facchinetti, A. C. Curino; Exper. Mol. Pathol. 97 (2014) 411-424. Link

<

141. PRK1/PKN1 controls migration and metastasis of androgen independent prostate cancer cells.
C. A. Jilg, A. Ketscher, E. Metzger, B. Hummel, D. Willmann, V. Rüsseler, V. Drendel, A. Imhof, M. Jung, H. Franz, S. Hölz, M. Krönig, J. M. Müller, R. Schüle; OncoTarget 5 (2014) 12646-12664. Link (open access)

142. Fluorescence-Based Screening Assays for the NAD+-Dependent Histone Deacetylase smSirt2 from Schistosoma mansoni.
M. Schiedel, M. Marek, J. Lancelot, B. Karaman, I. Almlöf, J. Schultz, W. Sippl, R. J. Pierce, C. Romier, M. Jung; J. Biomol. Screen. 20 (2015) 112-121. Link (open access)

143. The Discovery of a Highly Selective 5,6,7,8-Tetrahydrobenzo[4,5]thieno[2,3-d]pyrimidin-4(3H)-one SIRT2 Inhibitor that is Neuroprotective in an in vitro Parkinson's Disease Model.
P. Di Fruscia, E. Zacharioudakis, C. Liu, S. Moniot, S. Laohasinnarong, M. Khongkow, I. F. Harrison, K. Koltsida, C. R. Reynolds, K. Schmidtkunz, M. Jung, K. L. Chapman, C. Steegborn, D. T. Dexter, M. J. E. Sternberg, E. W.-F. Lam, M. J. Fuchter; ChemMedChem. 10 (2015) 69-82. Link

144. Histone acetyltransferase inhibitors block neuroblastoma cell growth in vivo.
J. M. Gajer, S. D. Furdas, A. Gründer, M. Gothwal, U. Heinicke, K. Keller, F. Colland, S. Fulda, H. L. Pahl, I. Fichtner, W. Sippl, M. Jung; Oncogenesis 4 (2015) e137. Link (open access)

145. Selective Sirt2 inhibition by ligand-induced rearrangement of the active site.
T. Rumpf, M. Schiedel, B. Karaman, C. Roessler, B. J. North, A. Lehotzky, J. Oláh, K. I. Ladwein, K. Schmidtkunz, M. Gajer, M. Pannek, C. Steegborn, D. A. Sinclair, S. Gerhardt, J. Ovádi, M. Schutkowski, W. Sippl, O. Einsle, M. Jung; Nat. Commun. 6 (2015) 6263. Link (open access)

146. NIK is required for NF-κB-mediated induction of BAG3 upon inhibition of constitutive protein degradation pathways.
F. Rapino, B. A. Abhari, M. Jung, S. Fulda; Cell Death Dis. 6 (2015) e1692. Link (open access)

147. Probing Chiral Recognition of Enzyme Active Sites with Octahedral Iridium(III) Propeller Complexes.
P. Göbel, F. Ritterbusch, M. Helms, M. Bischof, K. Harms, M. Jung, E. Meggers; Eur. J. Inorg. Chem. 2015 (2015) 1654-1659. Link

148. The histone code reader SPIN1 controls RET signaling in liposarcoma.
H. Franz, H. Greschik, D. Willmann, L. Ozretić, C. A. Jilg, E. Wardelmann, M. Jung, R. Buettner, R. Schüle; OncoTarget 6 (2015) 4773-4789. Link (open access)

149. Schistosome sirtuins as drug targets.
J. Lancelot, A. Cabezas-Cruz, S. Caby, M. Marek, J. Schultz, C. Romier, W. Sippl, M. Jung, R. J. Pierce; Future Med. Chem. 7 (2015) 765-782. (Review) Link (open access)

150. Drugging the schistosome zinc-dependent HDACs: current progress and future perspectives.
M. Marek, G. Oliveira, R. J. Pierce, M. Jung, W. Sippl, C. Romier; Future Med. Chem. 7 (2015) 783-800. (Review) Link (open access)

<

151. HDAC8, a multifaceted target for pharmacological intervention.
A. Chakrabarti, I. Oehme, O. Witt, G. Oliveira, W. Sippl, C. Romier, R. J. Pierce, M. Jung; Trends Pharmacol. Sci. 36 (2015) 481-492. (Review) Link (open access)

152. Discovery of Histone Demethylase Inhibitors.
A.-T. Hauser, M. Roatsch, J. Schulz-Fincke, D. Robaa, W. Sippl, M. Jung; in: Epigenetic Technological Applications (Ed.: Y. G. Zheng) (2015) 397-424. (Book Chapter) Link

153. Tetrazolylhydrazides as Selective Fragment-Like Inhibitors of the JumonjiC-Domain-Containing Histone Demethylase KDM4A.
N. Rüger, M. Roatsch, T. Emmrich, H. Franz, R. Schüle, M. Jung, A. Link; ChemMedChem 10 (2015) 1875-1883. Link

154. Seeding for sirtuins: microseed matrix seeding to obtain crystals of human Sirt3 and Sirt2 suitable for soaking.
T. Rumpf, S. Gerhardt, O. Einsle, M. Jung; Acta Crystallogr., Sect. F: Struct. Biol. Commun. 71 (2015) 1498-1510. Link (open access)

155. Structure-Based Development of an Affinity Probe for Sirtuin 2.
M. Schiedel, T. Rumpf, B. Karaman, A. Lehotzky, S. Gerhardt, J. Ovádi, W. Sippl, O. Einsle, M. Jung; Angew. Chem. Int. Ed. 55 (2016) 2252-2256. Link

156. Assembly of methylated KDM1A and CHD1 drives androgen receptor-dependent transcription and translocation.
E. Metzger, D. Willmann, J. McMillan, I. Forne, P. Metzger, S. Gerhardt, K. Petroll, A. von Maessenhausen, S. Urban, A.-K. Schott, A. Espejo, A. Eberlin, D. Wohlwend, K. M. Schüle, M. Schleicher, S. Perner, M. T. Bedford, M. Jung, J. Dengjel, R. Flaig, A. Imhof, O. Einsle, R. Schüle; Nat. Struct. Mol. Biol. 23 (2016) 132-139. Link

157. Synthesis and Biological Investigation of Oxazole Hydroxamates as Highly Selective Histone Deacetylase 6 (HDAC6) Inhibitors.
J. Senger, J. Melesina, M. Marek, C. Romier, I. Oehme, O. Witt, W. Sippl, M. Jung; J. Med. Chem. 59 (2016) 1545-1555. Link

158. Substituted 2-(2-aminopyrimidin-4-yl)pyridine-4-carboxylates as potent inhibitors of JumonjiC domain-containing histone demethylases.
M. Roatsch, D. Robaa, M. Pippel, J. E. Nettleship, Y. Reddivari, L. E. Bird, I. Hoffmann, H. Franz, R. J. Owens, R. Schüle, R. Flaig, W. Sippl, M. Jung; Future Med. Chem. 8 (2016) 1553-1571. Link (open access)

159. Epigenetic Readers Interpreting the Lysine Methylome - Biological Roles and Drug Discovery.
T. Wagner, D. Robaa, W. Sippl, M. Jung; in: Drug Discovery in Cancer Epigenetics (Eds.: G. Egger, P. Arimondo) (2016) 273-304. (Book Chapter) Link

160. Structure-Based Design and Computational Studies of Sirtuin Inhibitors.
B. Karaman, M. Jung, W. Sippl; in: Epi-Informatics: Discovery and Development of Small Molecule Epigenetic Drugs and Probes (Ed.: J. L. Medina-Franco) (2016) 297-325. (Book Chapter) Link

<

161. KATching-Up on Small Molecule Modulators of Lysine Acetyltransferases.
R. P. Simon, D. Robaa, Z. Alhalabi, W. Sippl, M. Jung; J. Med. Chem. 59 (2016) 1249-1270. (Review) Link

162. Aminothiazoles as Potent and Selective Sirt2 Inhibitors: A Structure-Activity Relationship Study.
M. Schiedel, T. Rumpf, B. Karaman, A. Lehotzky, J. Oláh, S. Gerhardt, J. Ovádi, W. Sippl, O. Einsle, M. Jung; J. Med. Chem. 59 (2016) 1599-1612. Link

163. A Continuous, Fluorogenic Sirtuin 2 Deacylase Assay: Substrate Screening and Inhibitor Evaluation.
I. Galleano, M. Schiedel, M. Jung, A. S. Madsen, C. A. Olsen; J. Med. Chem. 59 (2016) 1021-1031. Link

164. Identification of a small-molecule ligand of the epigenetic reader protein Spindlin1 via a versatile screening platform.
T. Wagner, H. Greschik, T. Burgahn, K. Schmidtkunz, A.-K. Schott, J. McMillan, L. Baranauskienė, Y. Xiong, O. Fedorov, J. Jin, U. Oppermann, D. Matulis, R. Schüle, M. Jung; Nucleic Acids Res. 44 (2016) e88. Link (open access)

165. Structure-Based Design and Synthesis of Novel Inhibitors Targeting HDAC8 from Schistosoma mansoni for the Treatment of Schistosomiasis.
T. Heimburg, A. Chakrabarti, J. Lancelot, M. Marek, J. Melesina, A.-T. Hauser, T. B. Shaik, S. Duclaud, D. Robaa, F. Erdmann, M. Schmidt, C. Romier, R. J. Pierce, M. Jung, W. Sippl; J. Med. Chem. 59 (2016) 2423-2435. Link (open access)

166. A quinone mediator drives oxidations catalysed by alcohol dehydrogenase-containing cell lysates.
J. Haas, M. A. Schätzle, S. M. Husain, J. Schulz-Fincke, M. Jung, W. Hummel, M. Müller, S. Lüdeke; Chem. Commun. 52 (2016) 5198-5201. Link

167. Targeting histone methyltransferases and demethylases in clinical trials for cancer therapy.
L. Morera, M. Lübbert, M. Jung; Clin. Epigenet. 8 (2016), 57. (Review) Link (open access)

168. Preparation data of the bromodomains BRD3(1), BRD3(2), BRD4(1), and BRPF1B and crystallization of BRD4(1)-inhibitor complexes.
M. Hügle, X. Lucas, G. Weitzel, D. Ostrovskyi, B. Breit, S. Gerhardt, K. Schmidtkunz, M.Jung, R. Schüle, O. Einsle, S. Günther, D.Wohlwend; Data in Brief 7 (2016) 1370-1374. Link (open access)

169. Phenylpyrrole-based HDAC inhibitors: synthesis, molecular modeling and biological studies.
M. Brindisi, C. Cavella, S. Brogi, A. Nebbioso, J. Senger, S. Maramai, A. Ciotta, C. Iside, S. Butini, S. Lamponi, E. Novellino, L. Altucci, M. Jung, G. Campiani, S. Gemma; Future Med. Chem. 8 (2016) 1573-1587. Link

170. 4-Biphenylalanine- and 3-Phenyltyrosine-Derived Hydroxamic Acids as Inhibitors of the JumonjiC-Domain-Containing Histone Demethylase KDM4A.
L. Morera, M. Roatsch, M. C. D. Fürst, I. Hoffmann, J. Senger, M. Hau, H. Franz, R. Schüle, M. R. Heinrich, M. Jung; ChemMedChem. 11 (2016) 2063-2083. Link

<

171. Targeting histone deacetylase 8 as a therapeutic approach to cancer and neurodegenerative diseases.
A. Chakrabarti, J. Melesina, F. R. Kolbinger, I. Oehme, J. Senger, O.Witt, W. Sippl, M. Jung; Future Med. Chem. 8 (2016) 1609-1634. (Review) Link (open access)

172. Tofacitinib and analogs as inhibitors of the histone kinase PRK1 (PKN1).
D. Ostrovskyi, T. Rumpf, J. Eib, A. Lumbroso, I. Slynko, S. Klaeger, S. Heinzlmeier, M. Forster, M. Gehringer, E. Pfaffenrot, S. M. Bauer, K. Schmidtkunz, S. Wenzler, E. Metzger, B. Kuster, S. Laufer, R. Schüle, W. Sippl, B. Breit, M. Jung; Future Med. Chem. 8 (2016) 1537-1551. Link

173. Identification of Highly Potent Protein Kinase C-Related Kinase 1 Inhibitors by Virtual Screening, Binding Free Energy Rescoring, and in vitro Testing.
I. Slynko, K. Schmidtkunz, T. Rumpf, S. Klaeger, S. Heinzlmeir, A. Najar, E. Metzger, B. Kuster, R. Schüle, M. Jung, W. Sippl; ChemMedChem. 11 (2016) 2084-2094. Link

174. Identification and Structure-Activity Relationship Studies of Small-Molecule Inhibitors of the Methyllysine Reader Protein Spindlin1.
D. Robaa, T. Wagner, C. Luise, L. Carlino, J. McMillan, R. Flaig, R. Schüle, M. Jung, W. Sippl; ChemMedChem. 11 (2016) 2327-2338. Link

175. Metabolic Response to XD14 Treatment in Human Breast Cancer Cell Line MCF-7.
D. Pan, M. Kather, L. Willmann, M. Schlimpert, C. Bauer, S. Lagies, K. Schmidtkunz, S. U. Eisenhardt, M. Jung, S. Günther, B. Kammerer; Int. J. Mol. Sci. 17 (2016) 1772. Link (open access)

176. Epigenetic therapy approaches in non-small cell lung cancer: Update and perspectives.
I. Schiffmann, G. Greve, M. Jung, M. Lübbert; Epigenetics 11 (2016) 858-870. (Review) Link

177. The Current State of NAD+-Dependent Histone Deacetylases (Sirtuins) as Novel Therapeutic Targets.
M. Schiedel, D. Robaa, T. Rumpf, W. Sippl, M. Jung; Med. Res. Rev. 38 (2018) 147-200 (Review) Link (reading possible for non-subscribers)

178. Paving the way for dose banding of chemotherapy: an analytical approach.
H. Reinhardt, R. Trittler, S. G. Eggleton, S. Wöhrl, T. Epting, M. Buck, S. Kaiser, D. Jonas, J. Duyster, M. Jung, M. J. Hug, M. Engelhardt; J. Natl. Compr. Canc. Netw. 15 (2017) 484-493. Link

179. Cellular analysis of the action of epigenetic drugs and probes.
M. Hau, F. Zenk, A. Ganesan, N. Iovino, M. Jung; Epigenetics 12 (2017) 308-322. (Review) Link

180. Synthesis and biological evaluation of 8-hydroxy-2,7-naphthyridin-2-ium salts as novel inhibitors of acetylcholinesterase (AChE) and butyrylcholinesterase (BChE).
M. Schiedel, A. Fallarero, C. Luise, W. Sippl, P. Vuorela, M. Jung; Med. Chem. Commun. 8 (2017) 465-470. Link

<

181. Chemically induced degradation of sirtuin 2 (Sirt2) by a proteolysis targeting chimera (PROTAC) based on sirtuin rearranging ligands (SirReals).
M. Schiedel, D. Herp, S. Hammelmann, S. Swyter, A. Lehotzky, D. Robaa, J. Oláh, J. Ovádi, W. Sippl, M. Jung; J. Med. Chem. 61 (2018) 482-491. Link

182. Three-component aminoalkylations yielding dihydronaphthoxazine-based sirtuin inhibitors: scaffold modification and exploration of space for polar side-chains.
S. Vojacek, K. Beese, Z. Al Halabi, S. Swyter, A. Bodtke, C. Schulzke, M. Jung, W. Sippl, A. Link; Arch. Pharm. Chem. Life Sci. 350 (2017) e1700097. Link

183. Photochromic histone deacetylase inhibitors based on dithienylethenes and fulgimides.
D. Wutz, D. Gluhacevic, A. Chakrabarti, K. Schmidtkunz, F. Erdmann, C. Romier, W. Sippl, M. Jung, B. König; Org. Biomol. Chem. 15 (2017) 4882-4896. Link

184. Isophthalic acid-based HDAC inhibitors as potent inhibitors of HDAC8 from Schistosoma mansoni.
K. Stenzel, A. Chakrabarti, J. Melesina, F. K. Hansen, J. Lancelot, S. Herkenhöhner, B. Lungerich, M. Marek, C. Romier, R. J. Pierce, W. Sippl, M. Jung, T. Kurz; Arch. Pharm. Chem. Life Sci. 350 (2017) e1700096. Link

185. Design, multicomponent synthesis and anticancer activity of a focused histone deacetylase (HDAC) inhibitor library with peptoid-based cap groups.
V. Krieger, A. Hamacher, C. G. W. Gertzen, J. Senger, M. Marek, C. Romier, T. Kurz, M. Jung, H. Gohlke, M. U. Kassack, F. K. Hansen; J. Med. Chem. 60 (2017) 5493-5506. Link

186. Alkoxyurea-based Histone Deacetylase Inhibitors Increase Cisplatin Potency in Chemoresistant Cancer Cell Lines.
K. Stenzel, A. Hamacher, F. Hansen, C. Gertzen, J. Senger, V. Marquardt, L. Marek, M. Marek, M. Remke, C. Romier, M. Jung, H. Gohlke, M. Kassack, T. Kurz; J. Med. Chem. 60 (2017) 5334-5348. Link

187. Synthesis of highly substituted 3-arylideneindolin-2-ones.
D. N. Ong, S. Dittrich, S. Swyter, M. Jung, F. Bracher; Tetrahedron 73 (2017) 5668-5679. Link

188. The histone code reader Spin1 controls skeletal muscle development.
H. Greschik, D. Duteil, N. Messaddeq, D. Willmann, L. Arrigoni, M. Sum, M. Jung, D. Metzger, T. Manke, T. Guenther, R. Schüle; Cell Death Dis. 8 (2017) e3173. Link

189. Beyond the BET family: targeting CBP/p300 with 4-acyl pyrroles.
M. Hügle, X. Lucas, D. Ostrovskyi, P. Regenass, S. Gerhardt, O. Einsle, M. Hau, M. Jung, B. Breit, S. Günther, D. Wohlwend; Angew. Chem. Int. Ed. 56 (2017) 12476-12480. Link

190. Chromatin remodeling system p300-HDAC2-Sin3A is involved in arginine starvation-induced HIF-1a degradation at the ASS1 promoter for ASS1 derepression.
W.-B. Tsai, Y. Long, J. T. Chang, N. Savaraj, L. G. Feun, M. Jung, H. H. W. Chen, M. T. Kuo; Sci. Rep. 7 (2017) 10814. Link

<

191. New chemical tools for probing activity and inhibition of the NAD+ dependent lysine deacylase sirtuin 2.
S. Swyter, M. Schiedel, D. Monaldi, S. Szunyogh, A. Lehotzky, T. Rumpf, J. Ovádi, W. Sippl, M. Jung; Phil. Trans. R. Soc. B 373 (2018) 20170083. Link

192. Structure-activity relationship of propargylamine-based HDAC inhibitors.
M. Wünsch, J. Senger, P. Schultheisz, S. Schwartzbich, K. Schmidtkunz, C. Michalek, M. Klaß, S. Goskowitz, P. Borchert, L. Praetorius, W. Sippl, M. Jung, N. Sewald; ChemMedChem. 12 (2017) 2044. Link

193. Modulation of microtubule acetylation by the interplay of TPPP/p25, Sirt2 and new anticancer agents with anti-Sirt2 potency.
A. Szabó, J. Oláh, S. Szunyogh, A. Lehotzky, T. Szénási, M. Csaplár, M. Schiedel, P. Löw, M. Jung, J. Ovádi; Sci. Rep. 7 (2017) 17070. Link

194. Structure-based design and biological characterization of selective HDAC8 inhibitors with anti-neuroblastoma activity.
T. Heimburg, F. R. Kolbinger, P. Zeyen, E. Ghazy, D. Herp, K. Schmidtkunz, J. Melesina, D. Robaa, F. Erdmann, M. Schmidt, C. Romier, O. Witt, I. Oehme, M. Jung, W. Sippl; J. Med. Chem. 60 (2017) 10188-10204. Link

195. Structure-activity studies on N-substituted tranylcypromine derivatives lead to selective inhibitors of lysine specific demethylase 1 (LSD1) and potent inducers of leukemic cell differentiation.
J. Schulz-Fincke, M. Hau, J. Barth, D. Robaa, D. Willmann, A. Kürner, J. Haas, G. Greve, T. Haydn, S. Fulda, M. Lübbert, S. Lüdeke, T. Berg, W. Sippl, R. Schüle, M. Jung; Eur. J. Med. Chem. 144 (2018) 52-67. Link

196. Carbamates as prodrugs and new warhead for HDAC inhibition.
K. King, A.-T. Hauser, J. Melesina, W. Sippl, M. Jung; Molecules 23 (2018) 321. Link

197. Identification of Bichalcones as Sirtuin Inhibitors by Virtual Screening and in Vitro Testing.
B. Karaman, Z. Alhalabi, S. Swyter, S. O. Mihigo, K. Andrae-Marobela, M. Jung, W. Sippl, F. Ntie-Kang; Molecules 23 (2018) 416. Link

198. A kinome-wide RNAi screen identifies ALK as a target to sensitize neuroblastoma cells for HDAC8-inhibitor treatment.
J. Shen, S. Najafi, S. Stäble, J. Fabian, E. Koeneke, F. R. Kolbinger, J. Wrobel, B. Meder, M. Distel T. Heimburg, W. Sippl, M. Jung, H. Peterziel, D. Kranz, M. Boutros, F. Westermann, O. Witt, I. Oehme; Cell Death Diff. 25 (2018) 2053-2070. Link

199. Reactivation of endogenous retroviral elements via treatment with DNMT- and HDAC-inhibitors.
M. Daskalakis, D. Brocks, Y.-H. Sheng, M. D. Saiful Islam, A. Ressnerova, Y. Assenov, T. Milde, I. Oehme, O. Witt, A. Goyal, A. Kuehn, M. Hartmann, D. Weichenhan, M. Jung, C. Plass; Cell Cycle 17 (2018) 811-822 Link

200. Anti-aging: Auf dem Weg zum "Jungbrunnen" ?
J. M. Gajer, M. Jung; Pharm. Ztg. 163 (2018) 418-425. Link

<

201. A novel class of Schistosoma mansoni histone deacetylase 8 (HDAC8) inhibitors identified by structure-based virtual screening and in vitro testing.
C. V. Simoben, D. Robaa, A. Chakrabarti, K. Schmidtkunz, M. Marek, J. Lancelot, S. Kannan, J. Melesina, T. J. Shaik, R. Pierce, C. Romier, M. Jung, W. Sippl; Molecules 23 (2018) 566. Link

202. Isoform-selective HDAC1/6/8 inhibitors with an imidazo-ketopiperazine cap containing stereochemical diversity.
B. Lecointre, R. Narozny, M. T. Borello, J. Senger, A. Chakrabarti, M. Jung, M. Marek, C. Romier, J. Melesina, W. Sippl, L. Bischoff, A. Ganesan; Phil. Trans. R. Soc. B 373 (2018) 20170364. Link

203. Epigenetic small molecule modulators of histone and DNA methylation.
A.-T. Hauser, D. Robaa, M. Jung; Curr. Opin. Chem. Biol. 45 (2018) 73-85. Link (open access)

204. Safety and efficacy of vorinostat, bortezomib, doxorubicin and dexamethasone in a phase I/II study for relapsed or refractory multiple myeloma (VERUMM study: vorinostat in elderly, relapsed and unfit multiple myeloma)
J. Waldschmidt, A. Keller, G. Ihorst, O. Grishina, S. Müller, D. Wider, A. V. Frey, K. King, R. P. Simon, A. M. May, P. Tassone, J. Duyster, M. Jung, R. Wäsch, M. Engelhardt; Haematologica 103 (2018) e473-e479. Link

205. Synthesis, crystallization studies and in vitro characterization of novel cinnamic acid derivatives as SmHDAC8 inhibitors for the treatment of Schistosomiasis.
T. Bayer, A. Chakrabarti, J. Lancelot, T. B. Shaik, K. Hausmann, J. Melesina, K. Schmidtkunz, M. Marek, F. Erdmann, M. Schmidt, D. Robaa, C. Romier, R. J. Pierce, M. Jung, W. Sippl; ChemMedChem. 13 (2018) 1517. Link

206. Opening the selectivity pocket in the human lysine deacetylase Sirtuin2 - new opportunities, new questions.
D. Robaa, D. Monaldi, N. Wössner, N. Kudo, T. Rumpf, M. Schiedel, M. Yoshida, M. Jung; Chem. Record. 18 (2018) 1-8. Link

207. Photochromic Indolyl Fulgimides as Chromo-Pharmacophores Targeting Sirtuins.
N. A. Simeth, L.-M. Altmann, N. Wössner, E. Bauer, M. Jung, B. König; J. Org. Chem. 83 (2018) 7919-7927. Link

208. The HDAC6/8/10 inhibitor TH34 induces DNA damage-mediated cell death in human high-grade neuroblastoma cell lines.
F. R. Kolbinger, E. Koeneke, J. Ridinger, T. Heimburg, M. Müller, T. Bayer, W. Sippl, M. Jung, N. Gunkel, A. K. Miller, F. Westermann, O. Witt, I. Oehme; Arch. Tox. 92 (2018) 2649-2664. Link

209. Novel spiroindoline HDAC inhibitors: Synthesis, molecular modelling and biological studies.
M. Brindisi, J. Senger, C. Cavella, A. Grillo, G. Chemi, S. Gemma, D. M. Cucinella, S. Lamponi, F. Sarno, C. Iside, A. Nebbioso, E. Novellino, T. B. Shaik, C. Romier, D. Herp, M. Jung, S. Butini, G. Campiani, L. Altucci, S. Brogi; Eur. J. Med. Chem. 157 (2018) 127-138. Link

210. The involvement of the mitochondrial amidoxime reducing component (mARC) in the reductive metabolism of hydroxamic acids.
C. Ginsel, B. Plitzko, D. Froriep, D. A. Stolfa, M. Jung, C. Kubitza, A. Scheidig, A. Havemeyer, B. Clement; Drug. Metab. Dispos. 46 (2018) 1396-1402. Link

<

211. Characterization of histone deacetylase 8 (HDAC8) selective inhibition reveals specific active site structural and functional determinants.
M. Marek, T. Shaik, T. Heimburg, A. Chakrabarti, J. Lancelot, E. Ramos-Morales, C. Da Veiga, D. Kalinin, J. Melesina, D. Robaa, K. Schmidtkunz, T. Suzuki, R. Holl, E. Ennifar, R. J. Pierce, M. Jung, W. Sippl, C. Romier; J. Med. Chem. 61 (2018) 10000-10016. Link

212. Avoiding chemotherapy prescribing errors: analysis and innovative strategies.
H. Reinhardt, P. Otte, M. Szymaniak-Vits, S. Ajayi, A. G. Eggleton, M. Ruch, J. Duyster, M. Jung, M. J. Hug, M. Engelhardt; Cancer 125 (2019) 1536-1546. Link

213. Cofactor Analogues as Active Site Probes in Lysine Acetyltransferases.
R. P. Simon, T. Rumpf, V. Linkuvienė, D. Matulis, A. Akhtar, M. Jung; J. Med. Chem. 62 (2019) 2582-2597. Link
Link to preprint at ChemRxiv (open access)

214. LSD1 inhibition by tranylcypromine derivatives interferes with GFI1-mediated repression of PU.1 target genes and induces differentiation in AML.
J. Barth, K. Abou-El-Ardat, D. Dalic, N. Kurrle, A.-M. Scheder, S. Mohr, J. Schüttle, L. Vassen, G. Greve, J. Schulz-Fincke, M. Schmitt, M. Tosic, E. Metzger, G. Bug, C. Khandanpour, S. Wagner, M. Lübbert, M. Jung, H. Serve, R. Schüle, T. Berg; Leukemia 33 (2019) 1411-1426. Link

215. Covalent inhibition of histone deacetylase 8 by 3,4-dihydro-2H-pyrimido [1,2-c][1,3]benzothiazin-6-imine.
M. Muth, N. Jänsch, A. Kopranovic, A. Krämer, N. Wössner, M. Jung, F. Kirschhöfer, G. Brenner-Weiß, F. Meyer-Almes; Biochim. Biophys. Acta, Gen. Subj. 1863 (2019) 577-585. Link

216. Structure-metabolism relationships of valine- and tert-leucine-derived synthetic cannabinoid receptor agonists: a systematic comparison of the in vitro phase I metabolism using pooled human liver microsomes and high-resolution mass spectrometry.
F. Franz, H. Jechle, M. Wilde, V. Angerer, L. M. Huppertz, M. Longworth, M. Kassiou, M. Jung, V. Auwärter; Forensic Toxicol. 37 (2019) 316-329. Link

217. Synthesis and biological investigation of phenothiazine-based benzhydroxamic acids as selective histone deacetylase 6 (HDAC6) inhibitors.
K. Vögerl, N. Ong, J. Senger, D. Herp, K. Schmidtkunz, M. Marek, M. Müller, K. Bartel, T. B. Shaik, N. Porter, D. Robaa, D. W. Christianson, C. Romier, W. Sippl, M. Jung, F. Bracher; J. Med. Chem. 62 (2019) 1138-1166. Link

218. Novel sustainable-by-design HDAC inhibitors for the treatment of Alzheimer's disease.
L. A. Soares Romeiro, J. L. da Costa Nunes, C. de Oliveira Miranda, G. Simões Heyn Roth Cardoso, A. Souza de Oliveira, A. Gandini, T. Kobrlova, O. Soukup, M. Rossi, J. Senger, M. Jung, S. Gervasoni, G. Vistoli, S. Petralla, F. Massenzio, B. Monti, M. L. Bolognesi; ACS Med. Chem. Lett. 10 (2019) 671-676. Link

219. A Chemical Probe For Tudor Domain Protein Spin1 to Investigate Chromatin Functions.
V. Fagan, C. Johansson, C. Gileadi, O.P. Monteiro, J.E. Dunford, R. Nibhani, M. Philpott, J. Malzahn, G. Wells, R. Farham, A. Cribbs, N. Halidi, F. Li, I. Chau, H. Greschik, S. Velupillai, A.Allali-Hassani, J. Bennett, T. Christott, C. Giroud, A.M. Lewis, K. Huber, N. Athanasou, H. Greschik, M. Jung, R. Schüle, M. Vedadi, C. Arrowsmith, Y. Xiong, J. Jin, O. Fedorov, G. Farnie, P. Brennan, U. Oppermann; J. Med. Chem. 62 (2019) 9008-9025. Link
Link to preprint at ChemRxiv (open access)

220. The Clinically Used Iron Chelator Deferasirox is an Inhibitor of Epigenetic JumonjiC Domain-Containing Histone Demethylases.
M. Roatsch, I. Hoffmann, M.I. Abboud, R.L. Hancock, H. Tarhonskaya, K.-F. Hsu, S.E. Wilkins, T.-. lan . Yeh, K. Lippl, K. Serrer, I. Moneke, T.D. Ahrens, D. Robaa, S. Wenzler, N.P.F. Barthes, H. Franz, W. Sippl, S. Lassmann, S. Diederichs, E. Schleicher, C.J. Schofield, A. Kawamura, R. Schüle, M. Jung; ACS Chem. Biol. 14(2019)1737-1750. Link
Link to preprint at ChemRxiv (open access)

<

221. Structure-Reactivity Relationships on Substrates and Inhibitors of the Lysine Deacylase Sirtuin 2 from Schistosoma Mansoni (SmSirt2).
D. Monaldi, D. Rotili, J. Lancelot, M. Marek, N. Wössner, A. Lucidi, D. Tomaselli, E. Ramos-Morales, C. Romier, R.J. Pierce, A. Mai, M. Jung; J. Med. Chem. 62 (2019) 8733-8759. Link
Link to preprint at ChemRxiv (open access)

222. KMT9 mono-methylates histone H4 Lysine 12 and controls proliferation of prostate cancer cells.
E. Metzger, S. Wang, S. Urban, D. Willmann, A. Schmidt, A. Offermann, A. Allen, M. Sum, N. Obier, F. Cottard, S. Ulferts, B.-T. Preca, B. Herrmann, J. Maurer, H. Greschik, V. Hornung, O. Einsle, S. Perner, A. Imhof, M. Jung, R. Schüle;
Nat. Struct. Mol. Biol. 26 (2019) 361-371. Link (open access)

223. Discovery of a Potent and Selective Fragment-like Inhibitor of Methyllysine Reader Protein Spindlin 1 (SPIN1).
Y. Xiong, H. Greschik, C. Johansson, L. Seifert, J. Bacher, K. Park, N. Babault, M.L. Martini, V. Fagan, F. Li, I. Chau, T. Christott, D. Dilworth, D. Barsyte-Lovejoy, M. Vedadi, C.H. Arrowsmith, P.E. Brennan, O. Fedorov, M. Jung, G. Farnie, J. Liu, U.C.T. Oppermann, R. Schüle, J. Jin;
J. Med. Chem. 62 (2019) 8996-9007. Link

224. Azologization of a hetero-stilbene based c-RAF kinase inhibitor: Towards the design of photoswitchable sirtuin inhibitors.
C.W. Grathwol, N. Wössner, S. Swyter, A.C. Smith, E. Tapavicza, R.K. Hofstetter, A. Bodtke, M. Jung, A. Link; Beilstein J.Org.Chem. 15 (2019) 2170-2183. Link (open access)

225. Tetrahydroindoles as multipurpose screening compounds and novel sirtuin inhibitors.
S. Vojacek, L. Schulig, N. Wössner, N. Geist, W. Langel, M. Jung, D. Schade, A Link; ChemMedChem 14 (2019) 853-864. Link

226. Structure-based screening of tetrazolylhydrazide inhibitors vs. KDM4 histone demethylases.
P.H. Malecki, N. Rüger, M. Roatsch, O. Krylova, A. Link, M. Jung, U. Heinemann, M.S. Weiss; ChemMedChem 14(21) (2019) 1828-1839. Link

227. Nitroreductase-mediated release of inhibitors of Lysine-Specific Demethylase 1 (LSD1) from prodrugs in transfected acute myeloid leukaemia cells.
E.-M. Herrlinger, M. Hau, D. M. Redhaber, G Greve, D. Willmann, S. Steimle, M. Müller, M. Lübbert, C. C. Miething, R Schüle, M. Jung; ChemBioChem (2020) accepted. Link to preprint at ChemRxiv (open access)

228. Screening and phenotypical characterization of Schistosoma mansoni histone deacetylase 8 (SmHDAC8) inhibitors as multi-stage antischistosomal agents.
F. Saccoccia, M. Brindisi, R. Gimmelli, N. Relitti, A. Alessandra; A. P. Saraswati, C. Cavella, S. Brogi, G. Chemi, S. Butini, G. Papoff, J. Senger, D. Herp, M. Jung, G. Campiani, S. Gemma, G. Ruberti;
ACS Infect. Dis. 6 (2019) 100-113. Link

229. Validation of slow off-kinetics of Sirtuin Rearranging Ligands (SirReals) by means of the label-free electrically switchable nanolever technology.
M. Schiedel, H.Daub, A. Itzen, M.Jung; ChemBioChem (2019) accepted. Link (open access)
Link to preprint at ChemRxiv (open access)

230. The lifespan increasing drug nordihydroguaiaretic acid inhibits p300 in vitro and in vivo and activates autophagy.
T.Tezil, M.Chamoli, C.P. Ng, R.P. Simon, V. Butler, M. Jung, J. Andersen, A. Kao, E. Verdin; NPJ Aging Mech. Dis. 5 (2019) 7. Link

<

231. Exploring structural determinants of inhibitor affinity and selectivity in complexes with Histone Deacetylase 6.
J. D. Osko, N. J. Porter, P. A. N. Reddy, Y.-C. Xiao, J. Rokka, M. Jung, J. M. Hooker, J. M. Salvino, D. W. Christianson; J. Med. Chem. 63 (2019) 295-308. Link

232. Triazole-based smHDAC8 Inhibitors: Synthesis and Biological Evaluation.
D. V. Kalinin, T. B. Shaik, S. K. Jana, M. Pfafenrot, A. Chakrabarti, J. Melesina, W. Sippl, C. Romier, M. Jung, R. Holl; ChemMedChem 15(7) (2019) 571-584. Link (open access)

233. Chemical genetics screen identifies epigenetic mechanisms involved in dopaminergic and noradrenergic neurogenesis in zebrafish.
M. Westphal, P. Sant, M.Jung, A.-T. Hauser, W. Driever; Front. Genet. 11 (2020) 80. Link (open access)

234. Epigenetic PROTACs
A. Vogelmann, D. Robaa, W. Sippl, M. Jung; Curr. Opin. Chem. Biol. 57 (2020) 8-16. (Review) Link

235. Si1th (Sirtuin 1 Inhibiting Thiocyanates) – a new class of selective Sirt1 inhibitors.
N. Wössner, Z. Alhalabi, J. Gonzalez Nieto, S. Swyter, J. Gan, K. Schmidtkunz, L. Zhang, H. Ovaa, O. Einsle, A. Vaquero, W. Sippl, M. Jung; Front. Oncol. 10 (2020) 657. Link

236. Design, synthesis, and biological evaluation of dual targeting inhibitors of histone deacetylase 6/8 and bromodomain BRPF1.
E. Ghazy; P. Zeyen; D. Herp; M. Hügle; K. Schmidtkunz; F. Erdmann; D. Robaa; M. Schmidt; E. R Morales; C. Romier; S. Günther; M. Jung, W. Sippl; Eur. J. Med. Chem. 200 (2020) 112338. Link

237. Activation of Sirt2 inhibitors employing photoswitchable geometry and aqueous solubility.
C. W. Grathwol, N. Wössner, S. Behnisch-Cornwell, L. Schulig, M. Jung; ChemMedChem 15 (15) (2020) 1480–1489. Link

238. HaloTag-Targeted Sirtuin Rearranging Ligand (SirReal) for the Development of Proteolysis Targeting Chimeras (PROTACs) Against the Lysine Deacetylase Sirtuin 2 (Sirt2).
Matthias Schiedel, Attila Lehotzky, Sándor Szunyogh, Judit Oláh, Sören Hammelmann, Nathalie Wössner,Dina Robaa, Oliver Einsle, Wolfgang Sippl, Judit Ovádi, Manfred Jung; ChemBioChem (2020). Link

239. Epigenetics meets GPCR – Inhibition of histone H3 methyltransferase (G9a) and histamine H3 receptor for Prader-Willi Syndrome.
D. Reiner, L. Seifert, C. Deck, R. Schüle, M. Jung, H. Stark; Sci. Rep. 10 (2020) 13558. Link

240. Syntheses of Thailandepsin B Pseudo-Natural Products: Access to new highly potent HDAC Inhibitors via Late Stage Modification.
J. Brosowsky, M. Lutterbeck, A. Liebich, M. Keller, D. Herp, A. Vogelmann, M. Jung, B. Breit; Chem. Eur. J. 26 (2020). Link

<

241. Metabolite patterns in human myeloid hematopoiesis result from lineage-dependent active metabolic pathways.
L. Kaiser, H. Weinschrott, I. Quint, M. Blaess, R. Csuk, M. Jung, M. Kohl, H.-P. Deigner; Int. J. Mol. Sci. 21 (17) (2020) 6092. Link

242. Identification of the subtype-selective Sirt5 inhibitor balsalazide through systematic SAR analysis and rationalization via theoretical investigations.
C. Glas, J. C. B. Dietschreit, N. Wössner, L. Urban, E. Ghazy, W. Sippl, M. Jung, C. Ochsenfeld, F. Bracher; Eur. J. Med. Chem. 206 (2020) 112676. Link

243. Spiroindoline-capped selective HDAC6 inhibitors: Design, Synthesis, Structural Analysis and Biological Evaluation.
A. P. Saraswati, N. Relitti, M. Brindisi, J. D. Osko, G. Chemi, S. Federico, A. Grillo, S. Brogi, N. H. McCabe, R. C. Turkington, O. Ibrahim, J. O'Sullivan, S. Lamponi, M. Ghanim, V. P. Kelly, D. Zisterer, R. Amet, P. Hannon, F. Vanni, C. Ulivieri, D. Herp, A. Di Costanzo, F. Saccoccia, G. Ruberti, M. Jung, L. Altucci, S. Gemma, S. Butini, D. Christianson, G. Campiani; ACS Med. Chem. Lett. 11(11) (2020) 2268-2276. Link

244. 4-Acyl pyrroles as dual BET-BRD7/9 bromodomain inhibitors address BETi insensitive human cancer cell lines.
M. Hügle, P. Regenass, R. Warstat, M. Hau,X. Lucas, D. Wohlwend, O. Einsle, M. Jung, B. Breit, S. Günther; J. Med. Chem. (2020) accepted. Link

245. Effects of environmental conditions on nephron number: Modelling maternal diabetes and epigenetic regulation in renal development.
L. Fuhrmann, S. Lindner, A.-T. Hauser, C. Höse, O. Kretz, C. D. Cohen, M. T. Lindenmeyer, W. Sippl, M. Jung, T. B. Huber, N. Wanner; Int. J. Mol. Sci. (2021) accepted. Link

246. Structure-Based Design, Docking and Binding Free Energy Calculations of A366 Derivatives as Spindlin1 Inhibitors.
Chiara Luise, Dina Robaa, Pierre Regenass, David Maurer, Dmytro Ostrovskyi, Ludwig Seifert, Johannes Bacher, Teresa Burghan, Tobias Wagner, Johannes Seitz, Holger Greschik, Kwang-Su Park, Yan Xiong, Jian Jin, Roland Schüle, Bernhard Breit, Manfred Jung, Wolfgang Sippl; Int. J. Mol. Sci. 22(11) (2021) 5910 . Link

247. Hypoxia-activated KDAC inhibitor: taking a breath from untargeted therapy.
M. Staudt, M. Jung; Cell Chem. Biol. 28 (2021) 1255-1257.

248. The potential for histone deacetylase (HDAC) inhibitors as cestocidal drugs.
Vaca HR, Celentano AM, Toscanini MA, Heimburg T, Ghazy E, Zeyen P, Hauser AT, Oliveira G, Elissondo MC, Jung M, Sippl W, Camicia F, Rosenzvit MC; PLoS Negl Trop Dis. 15 (2021) e0009226. Link

249. Novel quinolone-based potent and selective HDAC6 inhibitors: synthesis, molecular modelling studies and biological investigation.
N. Relitti, A. Prasanth Saraswati, G. Chemi, M. Brindisi, S. Brogi, D. Herp, K. Schmidtkunz, F. Saccoccia, G. Ruberti, F. Vanni, F. Sarno, L. Altucci, S. Lamponi, M. Jung, S. Gemma, S. Butini, G. Campiani; Eur. J. Med. Chem. 212 (2021) 112998. Link

250. Harnessing the role of HDAC6 in idiopathic pulmonary fibrosis: Design, synthesis, structural analysis, and biological evaluation of potent inhibitors.
G. Campiani, C. Cavella, J. D. Osko, M. Brindisi, N. Relitti, S. Brogi, A. P. Saraswati, S. Federico, G. Chemi, S. Maramai, G. Carullo, B. Jaeger, A. Carleo, R. Benedetti, F. Sarno, S. Lamponi, P. Rottoli, E. Bargagli, C. Bertucci, D. Tedesco, D. Herp, J. Senger, G. Ruberti, F. Saccoccia, S. Saponara, B. Gorelli, M. Valoti, B. Kennedy, H. Sundaramurthi, S. Butini, M. Jung, K. M. Roach, L. Altucci, P. Bradding, D. W. Christianson, S. Gemma, A. Prasse; J. Med. Chem. 64 (2021) 9960-9988. Link

<

251. Lineage-selective disturbance of human hematopoietic stem cell differentiation by the commonly used plasticizer di-2-ethylhexyl phthalate via reactive oxygen species: fatty acid oxidation makes the difference.
L. Kaiser, I. Quint, R. Csuk, M. Jung, H.-P. Deigner; Cells (2021) accepted.

252. Kappa but not delta or mu opioid receptors form homodimers at low membrane densities.
K. Cechova, C. Lan, N. P. F. Barthes, M. Jung, M. Ulbrich; Cell Mol. Life Sci. (2021). Link

253. Development of a NanoBRET assay to validate dual inhibitors of Sirt2-mediated lysine deacetylation and defatty-acylation that block prostate cancer cell migration.
A. Vogelmann, M. Schiedel, N. Wössner, A. Merz, D. Herp, S. Hammelmann, A. Colcerasa, G. Komaniecki, J. Y. Hong, M. Sum, E. Metzger, E. Neuwirt, L. Zhang, O. Einsle, O. Groß, R. Schüle, H. Lin, W. Sippl, M. Jung; RSC Chem. Biol. (2021). Link to preprint

254. Ex vivo propagation in a novel 3D high-throughput co-culture system for multiple myeloma.
J. M. Waldschmidt, S. J. Fruttiger, D. Wider, J. Jung, A. R. Thomsen, T. N. Hartmann, J. Duyster, M. J. Hug, K. A. Azab, M. Jung, R. Wäsch, M. Engelhardt; J. Cancer Res. Clin. Oncol. (2022). Link

255. Frontiers in Medicinal Chemistry 2022 Goes Virtual.
E. Diamanti, M. Méndez Pérez, T. Ross, C. A. Kuttruff, J. Lefranc, F.-M. Klingler, F. von Nussbaum, M. Jung, M. Gehringer; ChemMedChem.. (2022). Link

256. Identification of HDAC10 Inhibitors that Modulate Autophagy-Related Proteins in Transformed Cells.
P. Zeyen, Y. Zeyn, D. Herp, F. Mahmoudi, T. Z. Yesiloglu, F. Erdmann, M. Schmidt, D. Robaa, C. Romier, J. Ridinger, D. W. Christianson, I. Oehme, M. Jung, O. H. Krämer, W. Sippl; Eur. J. Med. Chem. (2022). Link

257. Azetidin-2-one-based Small Molecules as dual hHDAC6/HDAC8 Inhibitors: Investigation of their Mechanism of Action and Impact of Dual Inhibition Profile on Cell Viability.
G. Campiani, S. Federico, T. Khan, A. Fontana, S. Brogi, R. Benedetti, F. Sarno, G. Carullo, A. Pezzotta, A. P. Saraswati, E. Passaro. L. Pozzetti, A. Papa, N. Relitti, S. Gemma, S. Butini, A. Pistocchi, A. Ramunno, F. Vincenzi, K. Varani, V. Tatangelo, L. Patrussi, C. T. Baldari, S. Saponara, B. Gorelli, S. Lamponi, M. Valoti, F. Saccoccia, M. Giannaccari, G. Ruberti, D. Herp, M. Jung, L. Altucci; Eur. J. Med. Chem. (2022). Link

258. First fluorescent acetylspermidine deacetylation assay for HDAC10 identifies selective inhibitors with cellular target engagement.
D. Herp, J. Ridinger, D. Robaa, S. A. Shinsky. K. Schmidtkunz, T. Z. Yesiloglu, T. Bayer, R. R. Steimbach, C. J. Herbst-Gervasoni, A. Merz, C. Romier, P. Sehr, N. Gunkel, A. K. Miller, D. W. Christianson, I. Oehme, W. Sippl, M. Jung; ChemBioChem. (2022). Link

259. HDAC6 inhibitor JS28 prevents pathological gene expression in cardiac myocytes.
V. Ngo, B. K. Fleischmann, M. Jung, L. Hein, A. Lother; J. Am. Heart Assoc. (2022). Link

260. Defined stereoisomers of 2''-amino NAD+ and their activity against human sirtuins and a bacterial (ADP-ribosyl) transferase.
S. Zähringer, T. Rumpf, J. Melesina, A. E. Lang, K. Aktories, W. Sippl, M. Jung, G. K. Wagner; Bioorg. Med. Chem. (2022). Link

<

261. Design, Synthesis and Biological Characterization of Histone Deacetylase 8 (HDAC8) Proteolysis Targeting Chimeras (PROTACs) with Anti-Neuroblastoma Activity.
S. Darwish, E. Ghazy, T. Heimburg, D. Herp, J. Ridinger, D. Robaa, K. Schmidtkunz, M. Schmidt, C. Romier, M. Jung, I. Oehme, W. Sippl; Eur. J. Med. Chem., Preprint in ChemRxiv . (2022). Link

262. Defined stereoisomers of 2''-amino NAD+ and their activity against human sirtuins and a bacterial (ADP-ribosyl) transferase.
S. Zähringer, T. Rumpf, J. Melesina, A. E. Lang, K. Aktories, W. Sippl, M. Jung, G. K. Wagner; Bioorg. Med. Chem. (2022). Link

263. Development of hetero-triaryls as a new chemotype for subtype-selective and potent Sirt5 inhibition.
C. Glas, E. Naydenova, S. Lechner, N. Wössner, L. Yang, J. Dietschreit, H. Sun, M. Jung, B Küster, C. Ochsenfeld, F. Bracher; Eur. J. Med. Chem. (2022). Link

264. Phenotypic Screening of histone deacetylase (HDAC) inhibitors against Schistosoma mansoni.
M. Hassan, A. Sedighi, O. Olaoye, C. Haberli, A. Merz, E. Ramos-Morales, E. de Araujo, C. Romier, M. Jung, J. Keiser, P. Gunning; ChemMedChem. (2022). Link

265. Chemically Diverse S. mansoni HDAC8 Inhibitors Reduced Viability in Worm Larval and Adult Stages.
B. Noce, E. Di Bello, C. Zwergel, R. Fioravanti, S. Valente, D. Rotili, A. Masotti, G. Fianco, D. Trisciuoglio, A. Chakrabarti, C. Romier, D. Robaa, W. Sippl, M. Jung, C. Häberli, J. Keiser, A. Mai; ChemMedChem. (2022). Link

266. Comparison of cellular target engagement methods for the tubulin deacetylases Sirt2 and HDAC6: NanoBRET, CETSA, tubulin acetylation, and PROTACs.
. A. Vogelmann, M. Jung, F. K. H. Hansen, M. Schiedel; ACS Pharmacol. Transl. Sci. (2022). Link

267. A novel pan-selective bromodomain inhibitor for epigenetic drug design.
M. Hügle, R. Warstat, M. Pervaiz, P. Regenass, M. Amann, K. Schmidtkunz, O. Einsle, M. Jung, B. Breit, S. Günther; Eur. J. Med. Chem. (2023). Link

268. Generation and in vitro Assessment of HDAC6 Proteolysis Targeting Chimera (PROTACs).
S. Darwish, T. Heimburg, J. Ridinger, D. Herp, M. Schmidt, C. Romier, M. Jung, I. Oehme, W. Sippl; Methods Mol. Biol (2023). Link

269. Photoreductive ?-aminoalkylation with amino acids affords functionalized ? amino-ketones for nucleoside mimics.
S. O. Klein, A. A. Baniahmad, M. Jung; ChemComm. (2023). Link

270. Melatonin-Vorinostat Hybrid Ligands Show Higher HDAC and Cancer Cell Growth Inhibition than Vorinostat.
Y. Y. Helmi, N. Papenkordt, A. K. El-Hady, N. Labani, F. Gbahou, M. Abdel-Halim, R. Jockers, D. P. Zlotos*, M. Jung; Arch. Pharm. (2023). Link

<

271. Continuous Fluorescent Sirtuin Activity Assay Based on Fatty Acylated Lysines.
M. Zessin, M. Meleshin, S. Hilscher, C. Schiene-Fischer, C. Barinka, M. Jung, M. Schutkowski ; Int. J. Mol. Sci. (2023). Link

272. Total synthesis and biological evaluation of histone deacetylase inhibitor WF-3161.
M. Kohr, N. Papenkordt, M. Jung, U. Kazmaier ; Org. Biomol. Chem. (2023). Link

273. Development of first-in-class dual Sirt2/HDAC6 inhibitors as molecular tools for dual inhibition of tubulin deacetylation.
L. Sinatra, A. Vogelmann, F. Friedrich, M. A. Tararina, E. Neuwirt, A. Colcerasa, P. König, L. Toy, T. Z. Yesiloglu, S. Hielscher, L. Gaitzsch, N. Papenkordt, S. Zhai, L. Zhang, C. Romier, O. Einsle, W. Sippl, O. Groß, G. Bendas, D. W. Christianson, F. Hansen, M. Jung, M. Schiedel. ; J. Med. Chem. (2023). Link

274. Quinoline-based thiazolyl-hydrazones target cancer cells through autophagy inhibition .
V. Curcic, M. Olszewski, N. Maciejewska, A. Višnjevac, T. Srdic-Rajic, V. Dobricic, A. T. García-Sosa, S. B. Kokanov, J. B. Araškov, R. Silvestri, R. Schüle, M. Jung, M. Nikolic, N. R. Filipovic ; Arch. Pharm. (2024). Link

275. Design and synthesis of bioreductive prodrugs of class I histone deacetylase inhibitors and their biological evaluation in virally transfected acute myeloid leukemia cells .
M. Abdelsalam, M. Zmyslia, K. Schmidtkunz, A. Vecchio, M. Zessin, M. Schutkowski, M. Jung, C. Jessen-Trefzer, W. Sippl ; Arch. Pharm. (2024). Link

276. Structure-guided design of a selective inhibitor of the histone methyltransferase KMT9 with cellular activity.
S. Wang, S. O. Klein, S. Urban, M. Staudt, N. P. F. Barthes, D. Willmann, J. Bacher, M. Sum, H. Bauer, L. Peng, G. A. Rennar, C. Gratzke, K. M. Schüle, L. Zhang, O. Einsle, H. Greschik, C. MacLeod, C. G. Thomson, M. Jung, E. Metzger, R. Schüle ; Nat. Commun. (2024). Link

277. Potent hydroxamate-derived compounds arrest endodyogeny of Toxoplasma gondii tachyzoites.
C. Araujo-Silva, K. Vögerl, F. Breu, M. Jung, A. Oliveira Costa, W. De Souza, F. Bracher, E. Martins-Duarte, R. Vommaro ; Exper. Parasitol. (2024). Link

   
 

 

 

 
     
© 2007-2017 by ijou.de/sign Impressum