Most recently published

  • Photo-Adrenalines": b2-Adrenergic receptor agonists as molecular probes for the study of spatiotemporal adrenergic signaling

  • Bridging the binding sites 2.0: Photoswitchable dualsteric ligands for the cannabinoid 2 receptor

  • Visible light photoswitchable benzimidazole azo-arenes as β-arrestin2-biased selective cannabinoid 2 receptor agonists

List of publications

  • “Flavonoid-phenolic acid hybrids are potent inhibitors of ferroptosis via attenuation of mitochondrial impairment” M. Günther, S. Dabare, J. Fuchs, S. Gunesch, J. Hofmann, M. Decker, C. Culmsee*, Antioxidants 2024, in press

  • “Photo-Adrenalines": b2-Adrenergic receptor agonists as molecular probes for the study of spatiotemporal adrenergic signaling” A. Sink, H. Gerwe, H. Hübner, V. Boivin-Jahns, J. Fender, K. Lorenz, P. Gmeiner, M. Decker*, Chem. Eur. J. 2024, e202303506

  • a)“Three-photon infrared stimulation of endogenous neuroreceptors in vivo“ R. Sortino, M. Cunquero, G. Castro-Olvera, R. Gelabert, M. Moreno, F. Riefolo, C. Matera, N. Fernàndez-Castillo, L. Agnetta, M. Decker, J. M. Lluch, J.Hernando, P. Loza-Alvarez*, P. Gorostiza*, Angew. Chem. Int. Ed. 2023, 62(51), e202311181

    b) “Three-photon infrared stimulation of endogenous neuroreceptors in vivo“ R. Sortino, M. Cunquero, G. Castro-Olvera, R. Gelabert, M. Moreno, F. Riefolo, C. Matera, N. Fernàndez-Castillo, L. Agnetta, M. Decker, J. M. Lluch, J.Hernando, P. Loza-Alvarez*, P. Gorostiza*, Angew. Chem. 2023, 135(51), e202311181

  • “Bridging the binding sites 2.0: Photoswitchable dualsteric ligands for the cannabinoid 2 receptor” S. A. M. Steinmüller, A. Tutov, J. N. Hislop, M. Decker*, ACS Chem. Neurosci. 2023, 14(20), 3737–3744

  • “Bridging the binding sites: dualsteric ligands for the cannabinoid 2 receptor (CB2R)” A. Tutov, S. A. M. Steinmüller, Y. A. Ramírez, J. A. Jack, D. A. Rodríguez-Soacha, C. A. Sotriffer, J. N. Hislop, M. Decker*, Adv. Therap. 2023, 6(4), 2200260

  • a) “Visible light photoswitchable benzimidazole azo-arenes as β-arrestin2-biased selective cannabinoid 2 receptor agonists” S. A. M. Steinmüller, J. Fender, M. H. Deventer, A. Tutov, K. Lorenz, C. P. Stove, J. N. Hislop, M. Decker*, Angew. Chem. Int. Ed. 2023, 62(49), e202306176 – selected for frontispiece picture

    b) “Mit sichtbarem Licht schaltbare Benzimidazol-Azoarene wirken als b-Arrestin-funktionell-selektive Cannabinoid-Rezeptor 2 Agonisten” S. A. M. Steinmüller, J. Fender, M. H. Deventer, A. Tutov, K. Lorenz, C. P. Stove, J. N. Hislop, M. Decker*, Angew. Chem. 2023, 135(49), e202306176 – selected for frontispiece picture

  • “Dual-acting small molecules: Subtype-selective cannabinoid receptor 2 agonist / butyrylcholinesterase inhibitor hybrids show neuroprotection in an Alzheimer's disease mouse model“ P. Spatz, S. A. M. Steinmüller, A. Tutov, E. Poeta, A. Morilleau, A. Carles, M. H. Deventer, J. Hofmann, C. P. Stove, B. Monti, T. Maurice*, M. Decker*, J. Med. Chem. 2023, 66(9), 6414-6435

  • ”Rationalizing binding modes of radiotracers targeting norepinephrine transporter after initial translational studies” A. Tutov, X. Chen, S. Mühlig, R. A. Werner, S. Mühlig. T. Zimmermann, N. Nose, K. Koshino, C. Lapa, M. Decker*, T. Higuchi*, Pharmaceutics 2023, 15(2),690

  • "Amber light control of peptide secondary structure by a perfluoroaromatic azobenzene photoswitch” E. Cataldi, M. Raschig, M. Gutmann, P. T. Geppert, M. Ruopp, M. Schock, H. Gerwe, R. Bertermann, L. Meinel, M. Finze, A. Nowak-Król, M. Decker, T. Lühmann*, ChemBioChem 2023, 24(5), e2002200570

  • „Photoswitchable pseudo-irreversible butyrylcholineserase inhibitors allow optical control of inhibition in vitro and enable restoration of cognition in an Alzheimer mouse model upon irradiation”
    M. Scheiner, A. Sink, M. Hoffmann, C. Vrigneau, E. Endres, A. Carles, C. Sotriffer, T. Maurice*, M. Decker*, J. Am. Chem. Soc. 2022, 144(7), 3279-3284

  • “Synthesis and biological evaluation of flavonoid-cinnamic acid amide hybrids with distinct activity in phenotypic screening for neurodegeneration lead to a compound with pronounced activity in a mouse model of Alzheimer’s disease” J. Hofmann, P. Spatz, R. Walther, M. Gutmann, T. Maurice, M. Decker*, Chem. Eur. J. 2022, in press

  • „The structure of cyclodecatriene collinolactone, its biosynthesis and semisynthetic analogues: Effects of monoastral phenotype and protection from intracellular oxidative stress”
    J. C. Schmid, K. Frey, M. Scheiner, J. F. Guerrero Garzón, L. Hoffmann, J.-N. Fricke, M Schuppe, H.-J. Schiewe, A. Zeeck, T. Weber, I. Usón, R. Kemkemer, M. Decker, S. Grond* Angew. Chem. Int. Ed. 2021, in press

  • „From virtual screening hits targeting a cryptic pocket in BACE-1 to a nontoxic brain permeable multitarget anti-Alzheimer lead with disease-modifying and cognition-enhancing effects”
    C. Pont, T. Ginex, C. Griñán-Ferré, M. Scheiner, A. Mattellone, N. Martínez, E. M. Arce, Y. Soriano-Fernández, M. Naldi, A. De Simone, M. Barenys, J. Gómez-Catalán, B. Pérez, R. Sabate, V. Andrisano, M. I. Loza, J. Brea, M.Bartolini, M. L. Bolognesi, M. Decker, M. Pallàs, F. J. Luque,* D. Muñoz-Torrero* Eur. J. Med. Chem. 2021, 225, 1137792

  • „Selective pseudo-irreversible butyrylcholinesterase inhibitors transferring antioxidant moieties to the enzyme show pronounced in vivo efficacy"
    M. Scheiner, M. Hoffmann, F. He, E. Poeta, B. Monti, A. Chatonnet, T. Maurice, M. Decker*, J. Med. Chem. 2021, 64(13), 9302–9320

  • „Photo-rimonabant: Synthesis and biological evaluation of novel photoswitchable molecules derived from rimonabant lead to a highly selective and nanomolar “cis-on” CB1R antagonist”
    D. A. Rodriguez-Soacha, J. Fender, Y. A. Ramírez, J. A. Collado, E. Muñoz, R. Maitra, C. Sotriffer, K. Lorenz, M. Decker*, ACS Chem. Neurosci. 2021, 12(9), 1632–1647

  • „Melatonin- and ferulic acid-based dual-cap selective HDAC6 inhibitors exhibit pronounced immunomodulatory effects in vitro and neuroprotective effects in a pharmacological Alzheimer’s disease mouse model”
    F. He, C. J. Chou, M. Scheiner, E. Poeta, N. Y. Chen, S. Gunesch, M. Hoffmann, C. Sotriffer, B. Monti, T. Maurice*, M. Decker*, J. Med. Chem. 2021, 64(7), 3794–381

  •  „Synthesis and initial characterization of a selective, pseudoirreversible inhibitor of human butyrylcholinesterase as PET tracers”
    C. Gentzsch, M. Hoffmann, Y. Ohshima, N. Nose, X. Chen, T. Higuchi*, M. Decker*, ChemMedChem 2021, 16(9), 1427-1437 - Hot Topic Special Collection “Fluorine Chemistry”

  •  „Synthetic azobenzene-based bioisosters of curcumin exhibit pronounced activity against amyloid aggregation, intracellular oxidative stress and neuroinflammation”
    J. Hofmann, T. Ginex, A. Espargaró, M. Scheiner, S. Gunesch, C. Stigloher, R. Sabaté, J. Luque,* M. Decker*, Chem. Eur. J. 2021, 27(19), 6015-6027

  •  „Synthesis, in vitro and in vivo characterization of a reversible, selective 18F-labeled radiotracer for human butyrylcholinesterase”
    C. Gentzsch, X. Chen, P. Spatz, U. Košak, D. Knez, S. Gobec, T. Higuchi*, M. Decker*, Mol. Imaging Biol. 2021, in press

  •  „Photopharmacology on acetylcholinesterase - Novel photoswitchable inhibitors with improved pharmacological profiles"
    A. Sink, M. Scheiner, P. Spatz, E. Endres, M. Decker*, ChemPhotoChem 2020, in press - Special Collection „Photopharmacology

  • „Studies into the flavonoid sterubin: total synthesis, enantioseparation, enantiopurity in nature, neuroprotectivity in vitro and in vivo
    J. Hofmann, S. Saef, M. Scheiner, M. Hoffmann, S. Oertler, A. Appelt-Menzel, P. Maher, T. Maurice, G. Bringmann*, M. Decker* Chem. Eur. J. 202026, 7299 – 7308

  • „7-O-Esters of taxifolin with pronounced overadditive effects in neuroprotection, anti-neuroinflammation and short-term memory”
    S. Gunesch, M. Hoffmann, C. Kiermeier, W. Fischer, A. Pinto, T. Maurice, P. Maher*, M. Decker* Redox Biol. 2020, 29, 101378

  • „Initial evaluation of AF78: A rationally designed fluorine-18-labelled PET radiotracer targeting norepinephrine transporter"
    X. Chen, A. Fritz, R. A. Werner, N. Nose; Y. Yagi, H. Kimura S. P. Rowe, K. Kazuhiro, M. Decker*, T. Higuchi* Mol. Imaging Biol. 2020, 22, 602-611

  • „Investigation into active state k opioid receptor homodimerization under physiological conditions via Single Molecule Microscopy using antagonistic fluorescent probes”
    A. Drakopoulos, Z. Köszegi, Y. Lanoiselée, H. Hübner, P. Gmeiner, D. Calebiro*, M. Decker* J. Med. Chem. 2020, 63(7), 3596-3609

  • „N-1,2,3-Triazole-isatin derivatives for cholinesterase and b-amyloid aggregation inhibition: a comprehensive bioassay study”
    C. S. Marques, Ó. López, D. Bagetta, E. P. Carreiro, S. Petrallae, M. Bartolini, M. Hoffmann, S. Alcaro, B. Monti, M. L. Bolognesi, M. Decker, J. G. Fernández-Bolaños A. J. Burke* Bioorg. Chem. 2020, 98, 103753

  • „Selective and wash-resistant fluorescent ligands allow single-molecule imaging of m-opioid receptor dimerization”
    C. Gentzsch, K. Seier, A. Drakopoulos, M.-L. Jobin, Y. Lanoiselée, D. Maurel, R. Sounier, H. Hübner, P. Gmeiner, S. Granier, D. Calebiro*, M. Decker* Angew. Chem. 2020, 132(15), 6014–6020 and Angew. Chem. Int. Ed. 2020, 59(15), 5958–5964
    “highly important” paper and selected for inside back cover picture

  • „7-O-Esters of taxifolin with pronounced overadditive effects in neuroprotection, anti-neuroinflammation and short-term memory”
    S. Gunesch, M. Hoffmann, C. Kiermeier, W. Fischer, A. Pinto, T. Maurice, P. Maher*, M. Decker* Redox Biol. 2020, 29, 101378

  • „Tunable duration of action of highly selective BChE inhibitors by chemical modification of transferable carbamate moieties lead to pronounced neuroprotective effect in an Alzheimer´s disease mouse model”
    M. Hoffmann, C. Stiller, E. Endres, M. Scheiner, S. Gunesch, C. Sotriffer, T. Maurice, M. Decker* J. Med. Chem. 2019, 62(20), 9116-9140

  • „Initial evaluation of AF78: A rationally designed fluorine-18-labelled PET radiotracer targeting norepinephrine transporter"
    X. Chen, A. Fritz, R. A. Werner, N. Nose; Y. Yagi, H. Kimura S. P. Rowe, K. Kazuhiro, M. Decker*, T. Higuchi* Mol. Imaging Biol. 2019, DOI: 10.1007/s11307-019-01407-5

  • „Dual-acting cholinesterase-human cannabinoid receptor 2 ligands show pronounced neuroprotection in vitro, and overadditive and disease-modifying neuroprotective effects in vivo”
    M. Scheiner, D. Dolles, S. Gunesch, M. Hoffmann, M. Nabissi, M. Oliviero, M. Naldi, M. Bartolini, S. Petralla, E. Poeta, B. Monti, C. Falkeis, M. Vieth, H. Hübner, P. Gmeiner, R. Maitra, T. Maurice, M. Decker* J. Med. Chem. 2019, 62(20), 9078-9102

  • „(R)-Tonkafuranone and related compounds – improved synthesis, stereochemical purity in nature, and bioactivities of the pure enantiomers”
    M. Hoffmann, J.-P. Mufusama Koy Sita, C. Kleider, S. Gunesch, B. Rothmund, L. Lehmann, G. Bringmann*, M. Decker* Flavour Frag. J. 2019, 34(5), 329-338

  • „Novel BQCA and TBPB derived M1 receptor hybrid-ligands: orthosteric carbachol differentially regulates partial agonism”
    S. Schramm, L. Agnetta, M. Bermudez, H. Gerwe, T. Littmann, M. Irmen, J. Holze, G. Wolber, C. Tränkle, M. Decker* ChemMedChem 2019, 14(14), 1349-1358

  • "Fluorination of photoswitchable muscarinic agonists tunes receptor pharmacology and photochromic properties”
    L. Agnetta, M. Bermudez, F. Riefolo, C. Matera, E. Claro, R. Messerer, T. Littmann, G. Wolber, U. Holzgrabe, M. DeckerJ. Med. Chem. 201962(6), 3009–3020 

  • "Control of cardiac function in vivo with a light-regulated drug"
    F. Riefolo, C. Matera, A. Garrido-Charles, A. Gomila, L. Agnetta, E. Claro, R. Masgrau, U. Holzgrabe, M. Batlle, M. Decker, E. Guasch, P. Gorostiza* J. Am. Chem. Soc. 2019141(18), 7628-7636 

  • "18F‐Labeled Derivatives of Irbesartan for Angiotensin II Receptor PET Imaging"
    M. Hoffmann,  X. Chen, M. Hirano, K. Arimitsu, H. Kimura, T. Higuchi, M. Decker, Chem Med Chem 2018, 13, 2546 –2557 

  • „Novel 18F-Labeled PET Imaging Agent FV45 Targeting the Renin–Angiotensin System"
    X. Chen, M. Hirano, R. A. Werner, M. Decker, T. Higuchi ACS Omega  2018, 3 (9), 10460-10470 

  • „Photopharmacology in Alzheimer's Disease"
    D. R. Soacha, M. DeckerAdv Ther 2018, 1800037 

  • „Regioselective synthesis of 7-O-esters of the flavonolignan silibinin and SARs lead to compounds with overadditive neuroprotective effects"
    S. Schramm, G. Huang, S. Gunesch, F. Lang, J. Roa, P. Högger, R. Sabate, P. Maher, M. Decker Eur. J. Med. Chem. 2018, 146, 93-107 

  • „The first photochromic affinity switch for the human cannabinoid receptor 2”
    D. Dolles, A. Strasser, H.-J. Wittmann, O. Marinelli, M. Nabissi, R. G. Pertwee, M. DeckerAdv. Therap. 2018, in press

  • „Cytotoxic properties of the alkaloid rutaecarpine and its oligocyclic derivatives and chemical modifications to enhance water-solubility”
    G. Huang, A. Drakopoulos, M. Saedtler, H. Zou, L. Meinel, J. Heilmann, M. Decker*Bioorg. Med. Chem. Lett.2017, 27(21), 4937-4941

  • „Evaluation of HepaRG cells for the assessment of indirect drug-induced hepatotoxicity using INH as a model substance”
    A. Mann, T. Pelz, K. Rennert, A. Mosig, M. Decker, A. Lupp* Hum. Cell 2017, 30(4), 267-278

  • „Novel bipharmacophoric inhibitors of the cholinesterases with affinity to the muscarinic receptors”
    R. Messerer, C. Dallanoce, C. Matera, S. Wehle, L. Flammini, B. Chirinda, A. Bock, C. Tränkle, E. Barocelli, M. Decker, C. Sotriffer, M. De Amici, U. Holzgrabe* Med. Chem. Commun. 2017, 8, 1346-1359

  • (a) „A photoswitchable dualsteric ligand controlling receptor efficacy”
    L. Agnetta, M. Kauk, M. C. A. Canizal, R. Messerer, U. Holzgrabe*, C. Hoffmann*, M. Decker* Angew. Chem. Int. Ed. 2017, 56(25), 7282-7287

    (b) „Ein photoschaltbarer Ligand zur Regulierung der Rezeptoraktivierung“
    L. Agnetta, M. Kauk, M. C. A. Canizal, R. Messerer, U. Holzgrabe*, C. Hoffmann*, M. Decker* Angew. Chem. 2017, 129(25), 7388-7393

  • „Experimental and theoretical investigation into the stability of cyclic aminals”
    E. Sawatzky, A. Drakopoulos, M. Rölz, C. Sotriffer, B. Engels, M. Decker* Beilstein J. Org. Chem. 2016, 12, 2280-2292

  • „The dual-acting AChE inhibitor and H3 receptor antagonist UW-MD-72 reverses amnesia induced by scopolamine or dizocilpine in passive avoidance paradigm in rats“
    B. Sadek*, N. Khan, F. H. Darras, S. Pockes, M. Decker Physiol. Behav. 2016, 165, 383-391

  • „A novel way to directly radiolabel human butyrylcholinesterase for PET through irreversible transfer of the radiolabeled moiety”
    E. Sawatzky, E. Al-Momani, R. Kobayashi, S. Samnick, T. Higuchi, M. Decker* ChemMedChem 2016, 11(14), 1540-1550

  • „Investigation into reactivity and stability of the pentacyclic alkaloid dehydroevodiamine and the benz-analog thereof”
    S. Wehle, A. Espargaró, R. Sabate, M. Decker*Tetrahedron2016, 72(20), 2535-2543       - selected for cover picture

  • „Discovery of highly selective and nanomolar carbamate-based butyrylcholinesterase inhibitors by rational investigation into their inhibition mode”
    E. Sawatzky, S. Wehle, B. Kling, J. Wendrich, G. Bringmann, C. Sotriffer, J. Heilmann, M. Decker*J. Med. Chem.2016, 59(5), 2067-2082

  • „Synthesis and biological assessment of racemic benzochromenopyrimidinimines as potent antioxidant, cholinesterase and Aβ1-42 self-induced aggregation inhibitors for Alzheimer’s disease therapy”
    Y. Dgachi, L. Ismaili,* D. Knez, M. Benchekroun, H. Martin Szałaj, S. Wehle, O. M. Bautista-Aguilera, V. Luzet, A. Bonnet, B. Malawska, S. Gobec, M. Chioua, M. Decker, F. Chabchoub*, J. Marco-Contelles* ChemMedChem 2016, 11(12), 1318-1327 – Special Issue: Polypharmacology & Multitarget Drugs

  • „Unconventional application of the Mitsunobu reaction: selective flavonolignan dehydration yielding hydnocarpins”
    G. Huang, S. Schramm, J. Heilmann, D. Biedermann, V. Křen, M. Decker*BeilsteinJ. Org. Chem. 2016, 12, 662-669

  • „Aminobenzimidazole and structural isomers as templates for dual-acting butyrylcholinesterase inhibitors and hCB2 ligands to combat neurodegenerative disorders“
    D. Dolles, M. Nimczick, M. Scheiner, J. Ramler, P. Stadtmüller, E. Sawatzky, A. Drakopoulos, C. Sotriffer, J. Wittmann, A. Strasser, M. Decker*ChemMedChem2016, 11(12), 1270-1283 – Special Issue: Polypharmacology & Multitarget Drugs; selected for cover picture

  • "The dual-acting H3 receptor antagonist and AChE inhibitor UW-MD-71 dose-dependently enhances memory retrieval and reverses dizocilpine-induced memory impairment in rats”
    N. Khan, A. Saad, S. M. Nurulain, F. H. Darras, M. Decker, B. Sadek* Behav. Brain Res. 2016, 297, 155-164

  • „Novel tacrine-grafted Ugi adducts as multipotent anti-Alzheimer drugs: A synthetic renewal in tacrine-ferulic acid hybrids”
    M. Benchekroun, M. Bartolini, J. Egea, A. Romero, E. Soriano,  M. Pudlo, V. Luzet, V. Andrisano, M.-L. Jimeno, M. G. López, S. Wehle, T. Gharbi, B. Refouvelet, L. de Andrés, C. Herrera-Arozamena, B. Monto, M. L. Bolognesi, M. I. Rodríguez-Franco, M. Decker, J. Marco-Contelles*, L. Ismaili* ChemMedChem 2015, 10(3), 523-539 – Editor’s pick 03/2015: Natural products

  • „Rational design of partial agonists for the muscarinic M1 acetylcholine receptor”
    X. Chen, J. Klöckner, J. Holze, C. Zimmermann, W. K. Seemann, R. Schrage, A. Bock, K. Mohr, C. Tränkle*, U. Holzgrabe*, M. Decker*J. Med. Chem.2015, 58(2), 560-576

  • „Acetylcholinesterase inhibitors with photoswitchable inhibition of b-amyloid aggregation”
    X. Chen, S. Wehle, N. Kuzmanovic, B. Merget, U. Holzgrabe, B. König, C. A. Sotriffer, M. Decker*ACS Chem. Neurosci.2014, 5(5), 377-389

  • „Synthesis and biological evaluation of bivalent cannabinoid receptor ligands based on hCB2R selective benzimidazole agonists reveal unexpected intrinsic properties”
    M. Nimczick, D. Pemp, F. H. Darras, X. Chen, J. Heilmann, M. Decker*Bioorg. Med. Chem. 2014, 22(15), 3938-3946

  • „Design, synthesis and biological evaluation of novel uni- and bivalent ligands for the cannabinoid receptor 1 with variation of spacer length and structure”
    G. Huang, D. Pemp, P. Stadtmüller, M. Nimczick, J. Heilmann, M. Decker* Bioorg. Med. Chem. Lett. 2014, 24(17), 4209-4214

  • „A simple heterocyclic fusion reaction and its application for expeditious syntheses of rutaecarpine and its analogs”
    G. Huang, D. Roos, P. Stadtmüller, M. Decker* Tetrahedron Lett. 2014, 55(26), 3607-3609

  • „Amine substitution of quinazolinones leads to selective nanomolar AChE inhibitors with "inverted" binding mode”
    F. H. Darras, S. Wehle, G. Huang, C. A. Sotriffer, M. Decker* Bioorg. Med. Chem. 2014, 22(17), 4867-4881

  • „Identification of a neuroprotective and selective butyrylcholinesterase inhibitor derived from the natural alkaloid evodiamine”
    G. Huang, B. Kling, F. H. Darras, J. Heilmann, M. Decker* Eur. J. Med. Chem.2014, 81, 15-21

  • „Synthesis, biological evaluation, and computational studies of tri- and tetracyclic nitrogen-bridgehead compounds as potent dual-acting AChE inhibitors and hH3 receptor antagonists”
    F. H. Darras, S. Pockes, G. Huang, S. Wehle, A. Strasser, H.-J. Wittmann, M. Nimczick, C. A. Sotriffer, M. Decker*ACS Chem. Neurosci.2014, 5(3), 225-242

  • „Cyclic acyl guanidines bearing carbamate structures allow potent and dirigible inhibition of either acetyl- or butyrylcholinesterase”
    F. H. Darras, B. Kling, E. Sawatzky, J. Heilmann, M. Decker*Bioorg. Med. Chem.2014, 22(17), 5020-5034

  • „Investigation into the selective debenzylation and ring cleavage of quinazoline-based heterocycles“
    E. Sawatzky, J. Bukowcan, M. Decker* Tetrahedron Lett. 2014, 55(18), 2973-2976

  • „Flavonoids, flavonoid metabolites, and phenolic acids inhibit oxidative stress in the neuronal cell line HT-22 monitored by ECIS and MTT assay: a comparative study”
    B. Kling, D. Bücherl, P. Palatzky, F.-M. Matysik, M. Decker, J. Wegener, J. Heilmann*J. Nat. Prod. 2014, 77(3), 446–454

  • In vitro stability and metabolism of a tacrine-silibinin codrug“
    K. Zenger, X. Chen, M. Decker, B. Kraus* J. Pharm. Pharmacol.2013, 65(12), 1765-1772 – Special Issue: Natural products for Alzheimer's disease therapy: Basic and application

  • „Tacrine-silibinin co-drug shows neuro- and hepatoprotective effects in vitro and procognitive and hepatoprotective effects in vivo
    X. Chen, K. Zenger, A. Lupp, B. Kling, J. Heilmann, C. Fleck*, B. Kraus, M. Decker*J. Med. Chem.2012, 55(11), 5231-5242

  • „Neuroprotective tri- and tetracyclic BChE inhibitors releasing reversible inhibitors upon carbamate transfer”
    F. H. Darras, B. Kling, J. Heilmann, M. Decker*ACS Med. Chem. Lett.2012, 3(11), 914-919 – Special issue: Alzheimer’s Disease

  • Mycobacterium tuberculosis and cholinesterase inhibitors from Voacanga globosa
    P. G. Macabeo,* W. S. Vidar, X. Chen, M. Decker, J. Heilmann, B. Wan, S. G. Franzblau, E. V. Galvez, M. A. M. Aguinaldo, G. A. Cordell Eur. J. Med. Chem.2011, 46(7), 3118-3123

  • „Probing the mid-gorge of cholinesterases with spacer-modified bivalent quinazolinimines leads to highly potent and selective butyrylcholinesterase inhibitors"
    X. Chen, I. G. Tikhonova, M. Decker* Bioorg. Med. Chem.2011, 19(3), 1222-1235

  • „Hybrid molecules from xanomeline and tacrine: Enhanced tacrine actions on cholinesterases and muscarinic M1 receptors”
    L. Fang, S. Jumpertz, D. Appenroth, C. Fleck, K. Mohr, C. Tränkle,* M. Decker* J. Med. Chem. 2010, 53(5), 2094-2103

  • „Cardioprotective effect of NO-metoprolol in murine Coxsackievirus B3-induced myocarditis”
    Glück,* K. Dahlke, R. Zell, A. Krumbholz, M. Decker, J. Lehmann, P. Wutzler J. Med. Virol. 2010, 82(12), 2043-2052

  • „Synthesis and opioid receptor binding affinities of 2-substituted and 3-amino-morphinans: ligands for mu, kappa and delta opioid receptors”
    M. Decker, Y.-G. Si, B. I. Knapp, J. M. Bidlack, J. L. Neumeyer* J. Med. Chem.2010, 53(1), 402-418

  • „Bivalent 5,8,9,13b-tetrahydro-6H-isochinolino-[1,2-a]isoquinolines and -isoquinolinium salts: Novel heterocyclic templates for butyrylcholinesterase inhibitors”
    M. Schulze, O. Siol, M. Decker, J. Lehmann* Bioorg. Med. Chem. Lett.2010, 20(9), 2946-2949

  • „Investigation into the in vivo effects of five newly synthesized tacrine / ferulic acid and ß-carboline derivatives on scopolamine-induced cognitive impairments in rats using radial maze experiments”
    Fleck,* D. Appenroth, L. Fang, Y. Schott, J. Lehmann, M. DeckerDrug Res. 2010, 60(6), 299-307

  • „Tacrine-NO and tacrine-ferulic acid hybrid molecules as new anti-Alzheimer agents: Hepatotoxicity and influence on the cytochrome P450 system in comparison to tacrine”
    Lupp, D. Appenroth, L. Fang, M. Decker, J. Lehmann, C. Fleck* Drug Res. 2010, 60(5), 229-237

  • „Univalent and bivalent ligands of butorphan: Characteristics of the linking chain determine the affinity and potency of such ligands"
    M. Decker, B. Fulton, B. Zhang, B. I. Knapp, J. M. Bidlack, J. L. Neumeyer* J. Med. Chem.2009, 52(23), 7389-7396 – Centennial Issue

  • „NO-donating tacrine hybrid compounds improve scopolamine-induced cognition impairment and show less hepatotoxicity”
    L. Fang, D. Appenroth, M. Decker, M. Kiehntopf, A. Lupp, S. Peng, C. Fleck, Y. Zhang,* J. Lehmann* J. Med. Chem. 2008, 51(24), 7666-7669

  • „A new way of data interpretation for cognition tests in rats used to characterize six cholinesterase inhibitors with heterocyclic nitrogen-bridgehead structure intended for Alzheimer therapy”
    Fleck,* D. Appenroth, M. Decker, J. Lehmann Drug Res. 2008, 58(11), 543-550

  • „Design and synthesis of tacrine-ferulic acid hybrids as multi-potent anti-Alzheimer drug candidates”
    L. Fang, B. Kraus, J. Lehmann, J. Heilmann, Y. Zhang,* M. Decker* Bioorg. Med. Chem. Lett. 2008, 18(9), 2905-2909

  • „Design, synthesis and pharmacological evaluation of hybrid molecules out of quinazolinimines and lipoic acid lead to highly potent and selective butyrylcholinesterase inhibitors with antioxidant properties”
    M. Decker,* B. Kraus, J. Heilmann Bioorg. Med. Chem. 2008, 16(8), 4252-4261

  • „Synthesis and biological evaluation of NO-donor-tacrine hybrids as hepatoprotective anti-Alzheimer drug candidates"
    L. Fang, D. Appenroth, M. Decker, M. Kiehntopf, C. Roegler, T. Deufel, C. Fleck, S. Peng, Y. Zhang,* J. Lehmann* J. Med. Chem.2008, 51(4), 713–716

  • „In vivo-investigations on the cholinesterase-inhibiting effects of tricyclic quinazolinimines: Scopolamine-induced cognitive impairments in rats are attenuated at low dosage and reinforced at higher dosage”
    Appenroth, M. Decker, J. Lehmann, C. Traenkle, K. Mohr, C. Fleck*Pflug. Arch. Eur. J. Phy. 2008, 455(5), 895-901

  • „6-Aryl-4-oxohexanoic acids: Synthesis, effects on eicosanoid biosynthesis, and anti-inflammatory in vivo-activities”
    K. Abouzid,* P. Frohberg, J. Lehmann, M. Decker* Med. Chem.2007, 3, 433-438

  • „6-Hydroxy- and 6-methoxy-b-carbolines as acetyl- and butyrylcholinesterase inhibitors“
    Y. Schott, M. Decker,* H. Rommelspacher, J. Lehmann* Bioorg. Med. Chem. Lett.2006, 16(22), 5840-5843

  • „Homobivalent quinazolinimines as novel nanomolar inhibitors of cholinesterases with dirigible selectivity toward butyrylcholinesterase”
    M. Decker* J. Med. Chem. 2006, 49(18), 5411-5413

  • „Alzheimer-like plaque formation by human macrophages is reduced by fibrillation inhibitors and lovastatin”
    G. P. Gellermann, K. Ullrich, A. Tannert, C. Unger, G. Habicht, S. R. N. Sauter, P. Hortschansky, U. Horn, U. Möllmann, M. Decker, J. Lehmann, M. Fändrich* J. Mol. Biol. 2006, 360, 251-257

  • „Synthesis and reactivity of dibenz[d,g]azecin-14(5H)-ones”
    P. Mohr, J. Lehmann, M. Decker* Heterocycles2006, 68(5), 879-884

  • „Novel tricyclic quinazolinimines and related tetracyclic nitrogen bridgehead compounds as cholinesterase inhibitors with selectivity towards butyrylcholinesterase”
    M. Decker,* F. Krauth, J. Lehmann Bioorg. Med. Chem.2006, 14(6), 1966-1977

  • „Dopamine/serotonin receptor ligands. Part XII: SAR studies on hexahydro-dibenz[d,g]azecines lead to 4-chloro-7-methyl-5,6,7,8,9,14-hexahydrodibenz[d,g]azecin-3-ol, the first picomolar D5-selective dopamine receptor antagonist”
    P. Mohr, M. Decker, C. Enzensperger, J. Lehmann* J. Med. Chem. 2006, 49(6), 2110-2116

  • „Dopamine/serotonin receptor ligands. Part XI. LE300: New results on its ability to antagonize the discriminative stimulus effects of cocaine”
    M. Decker, J. Lehmann* Pharmazie2006, 61(3), 248-250

  • „Dopamine/serotonin receptor ligands. Part X: SAR studies on azecine type dopamine receptor ligands by functional screening at human cloned D1, D2L and D5 receptors with a microplate reader based calcium assay lead to a novel potent D1/D5 selective antagonist”
    B. Höfgen, M. Decker, P. Mohr, A. M. Schramm, S. A. F. Rostom, H. El-Subbagh, P. M. Schweikert, D. R. Rudolf, M. U. Kassack, J. Lehmann* J. Med. Chem. 2006, 49(2), 760-769

  • „Novel inhibitors of acetyl- and butyrylcholinesterase derived from the alkaloids dehydroevodiamine and rutaecarpine”
    M. Decker* Eur. J. Med. Chem.2005, 40(3), 305-313

  • „Synthesis and vasorelaxant properties of hybrid molecules out of NO-donors and the b-receptor blocking drug propranolol”
    M. Decker,* A. König, E. Glusa, J. Lehmann Bioorg. Med. Chem. Lett.2004, 14(19), 4995-4997

  • „Dopamine/serotonin receptor ligands. Part IX: Oxygen containing midsized heterocyclic ring-systems and non-rigidised analogs – a step towards dopamine D5 receptor selectivity”
    T. W. Wittig, M. Decker, J. Lehmann* J. Med. Chem.2004, 47(17), 4155-4158

  • „Investigation into the mechanism of lactamization of lactones yielding in a novel route to biologically active tryptamine derivatives”
    M. Decker, T. T. H. Nguyen, J. Lehmann* Tetrahedron2004, 60(21), 4567-78

  • „Pharmacological studies on nitro-naproxen (naproxen-2-nitroxyethylester)”
    N. K. Jain, C. S. Patil, R. E. Kartasasmita, M. Decker, J. Lehmann, S. K. Kulkarni* Drug Dev. Res.2004, 61, 66-78

  • „Synthese von 6-Nitratoalkoxy-Analoga des Naproxens als Kandidaten für neuartige NO-freisetzende, nichtsteroidale Entzündungshemmer aus 6-Bromalkoxy-Analoga des Naproxen-methylesters“
    R. E. Kartasasmita,* M. Decker, J. Lehmann* Acta Pharmaceutica Indonesia2004, 29(3), 110-116

  • „Dopamine/serotonin receptor ligands. Part VIII: The dopamine receptor antagonist LE300 - Modelled and X-ray structure plus further pharmacological characterization, including serotonin receptor binding, biogenic amine transporter testing and in vivo testings”
    M. Decker, K.-J. Schleifer, M. Nieger, J. Lehmann* Eur. J. Med. Chem.2004, 39(6), 481-489

  • „Dopamine/serotonin receptor ligands. Part VII: Novel 3-substituted 5-phenyl-1,2,3,4,5,6-hexahydro-azepino-[4,5-b]-indoles as ligands for the dopamine receptors”
    M. Decker, J. Lehmann* Arch. Pharm. Chem. Life Sci. 2003, 366(10), 466-476

  • „Synthesis of 6-bromalkoxy analogs of naproxen methylesters as the first step in the preparation of novel NO-releasing naproxen-derivatives
    R. E. Kartasasmita,* M. Decker, J. Lehmann* Acta Pharmaceutica Indonesia2003, 28(4), 62-70

  • „Pharmacological characterization of the benz[d]indolo[2,3-g]azecine LE300, a novel type of a nanomolar dopamine receptor antagonist”
    M. U. Kassack,* B. Höfgen, M. Decker, N. Eckstein, J. Lehmann Naunyn Schmiedeberg`s Arch. Pharmacol.2002, 366(6), 543-550

  • „Dopamine/serotonin receptor ligands. Part IV: Synthesis and pharmacology of novel 3-benzazecines and 3-benzazonines as potential 5-HT2A and dopamine receptor ligands”
    H. El-Subbagh, T. Wittig, M. Decker, S. Elz, M. Nieger, J. Lehmann* Arch. Pharm. Chem. Life Sci. 2002, 355(9), 443-448

  • „Dopamine/serotonin receptor ligands. Part III: Synthesis and biological activities of 7,7’-alkylene-bis-6,7,8,9,14,15-hexahydro-5H-benz[d]indolo[2,3-g]azecines – Application of the bivalent ligand approach to a novel type of dopamine receptor antagonist”
    H. Abadi, S. Lankow, B. Höfgen, M. Decker, M. U. Kassack, J. Lehmann* Arch. Pharm. Chem. Life Sci. 2002, 335(8), 367-373

  • „Indoles. Part XIV: 4-Phenylpyrido[4,3-b]-Indoles and 5-phenylazepino[4,5-b]-indoles by cyclisation of N-indolylalkyl-ephedrine derivatives – Scope and stereoselectivity”
    M. Decker, R. Faust, M. Wedig, M. Nieger, U. Holzgrabe, J. Lehmann* Heterocycles2001, 55(8), 1455-1466

  • „Enantioseparation of linear and cyclic chiral bis(phenethyl)amines by means of cyclodextrin-modified capillary electrophoresis”
    M. Wedig, M. Thunhorst, S. Laug, M. Decker, J. Lehmann, U. Holzgrabe* Fresenius J. Anal. Chem.2001, 371(2), 212-217