Libro-Abstractsescorial 4 9 2 PDF
Libro-Abstractsescorial 4 9 2 PDF
Libro-Abstractsescorial 4 9 2 PDF
Comité Científico
Patrocinadores
Sesión de mañana Moderadores: Dra. M.T. García López, Dra. M.L. Jimeno
Dr. D. Andreu, Dra. R. Herranz
13:00-15:00 Almuerzo
13:00-15:00 Almuerzo
14:00-16:00 Almuerzo
HORST KESSLER
i
Technische Universität München
iiiii
ȱ
10
11
ROB M. J. LISKAMP
i
Utrecht Institute for Pharmaceutical
Sciences
Rob Liskamp studied chemistry and did his Ph.D. Bio-organic Chemistry at
the University of Nijmegen, The Netherlands (1982) working with Harry
Ottenheijm. Post-doctoral research (1983-1986) was carried out in the
group of I. Bernard Weinstein in The Institute of Cancer Research and in the
group of W. Clark Still, Department of Chemistry, Columbia University, New
York. In 1986 he moved to the University of Leiden. In 1991 he was a
visiting professor at the University of California in Los Angeles in the group
of François Diederich. In 1994 he became associate professor at Utrecht
University and in 1996 professor of molecular medicinal chemistry. He is
head of the group medicinal chemistry and chemical biology
(www.pharm.uu.nl/medchem). He has a joint appointment at the
departments of Pharmaceutical Sciences and Chemistry.
He is a Member of the executive board of the Netherlands Proteomic
Centre, member of the international advisory board of the European Journal
of Organic Chemistry and the editorial advisory boards of ChemBioChem, a
European Journal of Chemical Biology, and QSAR & Combinatorial
Science.
His research interests include biologically active modified peptides and
peptidomimetics, dendrimers, peptide folding, molecular recognition,
synthetic receptors, protein mimics including synthetic antibodies and
vaccines.
His scientific contributions so far are among others ca. 200 publications,
book chapters and patents.
iiiii
ȱ
12
13
MANUEL GUZMÁN
i
Universidad Complutense de Madrid
Manuel Guzmán was born in Madrid (1963) and took his BSc (1986) and
PhD (1990) in Biology from Madrid Complutense University. Subsequently
he performed his postdoctoral research at the University of Utrecht (The
Nertherlands) and the Hannah Research Institute (Ayr, UK). He is presently
Professor of Biochemistry and Molecular Biology at Madrid Complutense
University. His PhD and postdoctoral research focused on the regulation of
liver fatty acid metabolism. During the last ten years he has been mostly
involved in the study of the molecular mechanisms of cannabinoid action,
with especial emphasis on how cannabinoids modulate neural cell
proliferation, differentiation and survival, and how cannabinoids induce
cancer-cell death and exert anti-tumour effects. These studies have allowed
the characterization of new processes and signalling pathways coupled to
cannabinoid receptors, and overall support the notion that cannabinoids
impact very basic events underlying the control of cell fate.
iiiii
ȱ
16
9
' -Tetrahydrocannabinol (THC) and other cannabinoids inhibit the growth of
glioma cells and other types of cancer cells both in vitro and in animal
models of cancer. Cannabinoid anti-tumour activity is dependent on the
engagement of cannabinoid CB receptors and the modulation of key cell
signalling pathways. This reduces in turn cancer cell survival by at least two
mechanisms: induction of apoptosis and inhibition of angiogenesis.
Remarkably, this anti-proliferative effect seems to be highly selective for
cancer cells, supporting the notion that cannabinoid receptors regulate cell
survival/death pathways differently in tumour and non-tumour cells.
Recently we have identified a new route that mediates cannabinoid-induced
apoptosis of cancer cells via the sphingolipid ceramide and the endoplasmic
reticulum stress response, and have conducted a pilot clinical trial in which
patients with recurrent glioblastoma multiforme were administered THC
intra-tumourally. The fair safety profile of THC found in that study, together
with its growth-inhibiting action on cancer cells, may set the basis for future
trials aimed at evaluating the potential anti-tumour activity of cannabinoids.
17
TERESA CARLOMAGNO
i
Max Planck Institute
Teresa Carlomagno has obtained her Ph.D. from the University of Naples
Federico II in 1997 with a thesis on “NMR Multidimensional Techniques for
a Structural Analysis in Solution of Biologically Relevant Molecules”. From
1997 to 1999 she has been a post-doctoral fellow in the group of Prof. C.
Griesinger at the University of Frankfurt, while from 2000 to 2001 she has
worked as a research assistant at the Scripps Research Institute, California,
with Prof. James Williamson. Since 2002 she is the head of the group of
“Liquid-state NMR” at the Max Planck Institute for Biophysical Chemistry in
Göttingen, Germany. She investigates the structural basis of intermolecular
interactions by nuclear magnetic resonance in solution. Her research
focuses on the study of complexes between small organic molecules and
both proteins and RNAs, on large RNA/protein complexes and on catalytic
RNAs. Additionally, her group develops novel NMR techniques for structural
investigation of biomolecules. She has about 40 publications in peer
reviewed journals and has recently obtained the “Habilitation“ from the
University of Hannover, where she holds lectures. At the end of the year she
will move to the EMBL in Heidelberg, where she will hold the position of
group leader in biological NMR spectroscopy.
iiiii
ȱ
18
19
iiiii
ȱ
20
NUEVOS ANTIMALÁRICOS
i
Federico Gómez de las Heras
GlaxoSmithKline. Enfermedades de Países en Desarrollo.
ȱ
21
CARLOS GARCÍA-ECHEVERRÍA
i
Novartis
ȱ
iiiii
22
The phosphatidylinositol 3-kinases (PI3Ks) are widely expressed lipid kinases that
phosphorylate phosphoinosites at the D-3 position of the inositol ring. These
enzymes function as key signal transducers downstream of cell-surface receptor
tyrosine kinases. The eight members of the PI3K family are grouped into three
classes based on their primary amino acid sequence, in vitro substrate specificity,
structure and mode of regulation. Class I PI3Ks -PI3KD, E, J and G- catalyzed the
formation of phosphatidylinositol-3,4,5-triphosphate, PtdIns(3,4,5)P3 -also referred
to as PIP3-, a process that is reverted by the action of a phosphatase,
phosphatase and tensin homologue deleted on chromosome 10 (PTEN). Genetic
aberrations within class I PI3Ks are common in human cancer due to amplification,
overexpression or somatic missense mutations in the PI3KCA gene. Other
biological alterations can also affect the correct regulation of PIP3 signal
transducers. In this context, loss of the PTEN protein or function has been found in
a large fraction of advanced human cancers.
Downstream of PI3K is the 3-phosphoinosite-dependent protein kinase-1 (PDK1).
The attractiveness of PDK1 as a potential anticancer target is linked to its ability to
control the activity of a diverse set of AGC kinase members, in particular the three
PKB isoforms. Full activation of PKB requires phosphorylation at two sites, one
within the activation loop (e.g., Thr-308 for PKBD) and one within the C-terminus
(e.g., Ser-473 for PKBD). Phosphorylation of the critical and conserved threonine
residue in the activation loop of the three PKB isoforms is carried out by PDK1 at
the plasma membrane.
Whatever the mechanism, the prevalence of PI3K/PKB signaling abnormalities in
human cancers and its potential biological effects (e.g., competitive growth
advantage, evasion from apoptosis and therapy resistance) has suggested the
potential use of PI3K/PKB pathway modulators as novel targeted therapeutic
agents in oncology. Following this strategy, a number of compounds have
demonstrated antitumour activity in preclinical and clinical settings by targeting
directly or indirectly the different components of this pathway. This oral
presentation will detail our structure-based design efforts to identify modulators of
PDK1 and PI3K, and will highlight the evolution of our targeted therapeutic
strategies for the PI3K/PKB survival pathway. To this end, we have identified a
clinical candidate -NVP-BEZ235-, which exhibits potent antiproliferative activity
against a broad panel of tumour cell lines by specifically blocking the biological
function of PI3K signaling components (e.g., IC50 = 10 ± 1 nM, p-PKB inhibition in
U87MG cells). The activity of this PI3K inhibitor in cellular settings translates well in
in vivo models of human cancer. Thus, the compound was well tolerated and
displayed disease statis or tumour regression when administered orally -25 to 50
mg/kg/day-, and enhanced the efficacy of other anticancer agents when used in in
vivo combination studies. Ex-vivo PK/PD analyses of tumour tissues showed a
time-dependent correlation between compound concentration and PI3K/PKB
pathway inhibition. Contrary to other modulators of PI3K/PKB pathway, and upon
comparison with the mean glucose level of the control animals, no elevated blood
glucose levels were observed in the treated animals after in vivo efficacy
experiments. Phase I clinical studies with NVP-BEZ235 in cancer patients started
at the end of 2006.
23
JAVIER BOTET
i
Centro de Investigación del Cáncer
Salamanca
Licenciado
cenciado en Biología por la Universidad de Salamanca, realizó su
doctorado en el departamento de Microbiología y Genética de dicha
universidad, bajo la dirección del Dr. José Luis Revuelta. Durante su tesis
doctoral ha empleado la colecciones de mutantes deficientes en cada uno
de los genes de la levadura Saccharomyces cerevisiae para determinar el
mecanismo de acción de fármacos. Su perfil investigador ha estado
enfocado en la planificación y desarrollo de escrutinios químico-genómicos
a gran escala, empleando las herramientas genómicas disponibles en la
levadura, con objeto de profundizar en el mecanismo de acción de
compuestos con actividades antitumorales, antifúngicas, oxidantes y
genotóxicas. Su especialización se completó durante una estancia en The
Scripps Research Institute y en el Genomics Institute of the Novartis
Research Foundation (San Diego, California), concretamente en el
laboratorio de la Dra. Elizabeth Winzeler, considerada una de las científicas
pioneras en el trabajo con las colecciones de cepas deletantes de la
levadura. Desde hace un año, se incorporó al grupo del Dr. Sergio Moreno
en el Instituto de Biología Molecular y Celular del Cáncer de Salamanca,
donde mediante el mismo tipo de aproximaciones genómicas masivas,
trabaja en colaboración con la empresa PharmaMar, en la identificación de
las dianas moleculares y en el estudio del mecanismo de acción de varios
compuestos antitumorales de nueva generación que se unen al ADN.
ȱ
iiiii
24
A big challenge in drug discovery is to uncover the cellular targets and the
precise mechanism of action of bioactive molecules. The yeast
Saccharomyces cerevisiae is an excellent eukaryotic model system to study
basic cellular processes and human diseases because many genes have
been conserved through out evolution from yeast to humans. This easily
genetically tractable organism is at the forefront of large-scale functional
genomic and proteomic approaches. Many of these new technologies are
readily applicable to drug target identification and drug mechanism of action
studies.
25
iiiii
ȱ
26
27
ANA MARTÍNEZ
i
Neuropharma
iiiii
ȱ
28
1
Kingston DG, Newman DJ. Natural products as drug leads: An old process or the new hope for drug
discovery?. Idrugs 2005, 8:990-992.
2
Newman DJ, Cragg GM. Marine natural products and related compounds in clinical and advanced
preclinical trials. J. Nat. Prod. 2004, 67:1216-1238.
3
Alonso D, Castro A, Martinez A. Marine compounds for the therapeutic treatment of neurological
disorders. Expert Opin. Ther. Patents 2005, 15:1377-1386.
4
Martinez A. Marine-derived drugs in neurology. Current Opin. Investig. Drugs 2007, 8:525-530.
29
AURELIO ORJALES
i
Faes Farma
Información
ormación personal
Nombre y dirección: Aurelio Orjales
Dirección laboral actual: FAES FARMA, S.A.
Máximo Aguirre, 14 – 48940 Leioa (Vizcaya) – España
Teléfono +34 94 481 83 00. Correo electrónico: aorjales@faes.es
EXPEDIENTE ACADEMICO
Títulos universitarios
1. Licenciado en Ciencias Químicas, Universidad de Santiago de Compostela, en
marzo de 1969.
2. Doctor en Ciencias Químicas, Universidad de Santiago de Compostela, en
marzo de 1972.
EMPLEOS
1. Técnico de Síntesis Química, Departamento de Investigación de FAES, S.A.,
desde 1972 hasta 1977.
2. Jefe del Departamento de Investigación de FAES, S.A. desde 1977 hasta
1981.
3. Director de Investigación de FAES FARMA, S.A. (anteriormente FAES, S.A.),
desde 1981 hasta la fecha.
PUBLICACIONES
a. Trabajos científicos. Un total de 52 trabajos científicos originales han sido
publicados en revistas internacionales reconocidas, cubriendo
fundamentalmente aspectos de la Química Médica y Orgánica, Farmacología,
Farmacocinética y Desarrollo farmacéutico.
b. Comunicaciones a Congresos y Simposia. Más de 100 comunicaciones a
congresos y simposia científicos, cubriendo fundamentalmente aspectos de la
Química Médica y Orgánica, Farmacología, Farmacocinética y Desarrollo
farmacéutico.
c. Patentes. Más de 65 patentes sobre síntesis química y productos
farmacéuticos.
OTROS MÉRITOS
Congresos y Simposia: Asistente a numerosos congresos y simposia nacionales e
internacionales de Química, Biofarmacia, Farmacología y otros.
Comisiones: Vocal de comisiones gestoras de diversos Planes concertados de
investigación. Ponente en la “Revisión del Plan Nacional de I+D”. Madrid, 1995.
Tesis doctorales: Miembro de diversos tribunales de tesis doctorales en las Universidades
del País Vasco, Santiago de Compostela, Alcalá de Henares, Granada, Madrid y Navarra.
iiiii ȱ
30
CENTURY
i
Aurelio Orjales
FAES FARMA, S. A., Máximo Aguirre 14, 48940 Leioa (Vizcaya),
España
ȱ
31
ANNA GRANDAS
i
Universidad de Barcelona
Anna Grandas graduated in Chemistry and got her PhD degree at the
University of Barcelona. In 1989 she was appointed Profesora Titular at the
Depatment of Organic Chemistry of the University of Barcelona, and has
recently attained the full professorship degree. During the PhD thesis and
the first postdoctoral period in Barcelona she worked in peptide chemistry.
After a postdoctoral stay in Prof. Marvin H. Caruthers’ laboratory at the
University of Colorado at Boulder (1988), she directed her research interest
towards nucleic acids. In the field of nucleic acids chemistry her main
contributions have been related to the development of methodology for the
preparation of nucleic acids analogs, in particular of different types of
peptide-oligonucleotide hybrids.
iiiii
ȱ
32
This presentation will cover some of the work carried out in our group to
develop methodology for the preparation of oligonucleotide analogs. In
particular, the synthesis of peptide-oligonucleotide conjugates using the
Diels-Alder cycloaddition in water, and the formation of cross-links between
platinated oligonucleotides and complementary chains will be described.
1
V. Marchán, S. Ortega, D. Pulido, E. Pedroso, A. Grandas. Nucleic Acids Res. 2006, 34,
e24.
2
B. Algueró, J. López de la Osa, C. González, E. Pedroso, V. Marchán, A. Grandas.
Angew. Chem. Int. Ed., 2006, 45, 8194-8197.
33
ANDRÉ LUXEN
i
Université de Liège
iiiii
ȱ
34
35
BERNAT VIDAL
i
Almirall-Prodesfarma
Datos personales:
Nombre y apellidos: Bernat Vidal Juan
Fecha y lugar de nacimiento: 11/08/1968, Porreres (Mallorca)
Licenciado en Farmacia: Facultad de Farmacia, Universidad de Barcelona
(1991)
Doctorado en Farmacia: Facultad de Farmacia, Universidad de Barcelona
(1991-1996)
Supervisores: Prof. Joan Bosch y Prof. M.L. Bennasar.
Título: “Biomimetic synthesis of indole alkaloids. First total synthesis of
alkaloids of the ervitsine-ervatamine group”
Experiencia postdoctoral: Dpt of Chemistry, Stanford University (1997)
Supervisor: Prof. Barry M. Trost.
Título del trabajo: “Novel ruthenium catalyzed reactions and development of
new chiral bidentate ligands for the ruthenium atom”.
Experiencia profesional: Laboratorios Almirall S.A.
1998-2002: investigador, Departamento de Química Médica.
2002-actualidad: jefe de sección (Dept. de Química Médica)
Artículos y patentes:
Autor en 18 publicaciones científicas en diferentes revistas de química
orgánica y de química médica y en más de 9 patentes de invención.
Conferencias impartidas en congresos:
- Conferencia invitada: "New Xanthine-Based PDE5 Inhibitors"; XVIIth
International Symposium on Medicinal Chemistry (2002), Barcelona (Spain).
- Conferencia invitada: "Combinatorial Chemistry in Industry"; 6th
International Conference on Pharmaceutical Sciences (2001), Barcelona
(Spain).
Premios:
• Premio Extraordinario de Doctorado (1997)
• Premio Extraordinario Licenciatura (1993)
ȱ
iiiii
36
i
Bernat Vidal
Almirall-Prodesfarma
ȱ
H H
N N N N
O N O NH
N N
O
N N N N
LAS38096 LAS100268
37
PAOLO MASCAGNI
i
Italfármaco
iiiii
ȱ
38
39
ROSARIO GONZÁLEZ
i
Lilly
ȱ
iiiii
40
1. (a) Clark, C. M.; Karlawish, J. H. T. Annals of Internal Medicine 2003, 138, 400. (b)
Zlokovic, B. V. Adv. Drug Del. Rev. 2002, 54, 1533. 2. (a) Golde, T. E. J. Clinical Invest.
2003, 111, 11. (b) Wolfe, M. S. J. Med. Chem. 2001, 44, 2039. 3. (a) Durham, T.B.;
Shepherd, T.A. Curr. Opin. Drug Discov. Devel. 2006, 9, 776 (b) Guo, T.; Hobbs, D.W.
Curr. Med. Chem. 2006, 13, 1811. (c) Thompson, L. A.; Bronson, J. J.; Zusi, F. C. Curr.
Pharm. Design, 2005, 11, 3383. (d) Cumming, J. N.; Iserloh, U.; Kennedy, M. Curr. Opin.
Drug Discov. Devel. 2004, 7, 536. (e) Citron, M. Trends Pharmacol. Sci. 2004, 25, 92. (f)
John, V.; Beck, J. P.; Bienkowski, M. J.; Sinha, S.; Heinrikson, R. L. J. Med. Chem. 2003,
46, 4625. (g) Vassar, R. Adv. Drug. Del. Rev. 2002, 54, 1589. (h) Ghosh, A. K.; Hong, L.;
Tang, J. Curr. Med. Chem. 2002, 9, 1135.
41
DIDIER ROGNAN
i
CNRS - Université Louis
Pasteur Strasbourg
iiiii
ȱ
42
References
1. Müller, P., Lena, G. Bezzine, S. Boilard, E. Lambeau, G. Guichard G. and Rognan D.
(2006) In silico-guided target identification of a scaffold-focused library: 1,3,5-
Triazepane-2,6-diones as novel phospholipase A2 inhibitors. J. Med. Chem., 49, 6768-
6778. 2. Schalon, C., Surgand, J.S., Kellenberger, E. And Rognan, D. A fuzzy and
simple method for comparing druggable protein-ligand binding sites. J. Mol. Biol.,
submitted.
43
LEONARDO PARDO
i
Universidad Autónoma de Barcelona
iiiii
ȱ
44
1
Hopkins, A. L.; Groom, C. R. Nat. Rev. Drug Discov. 2002, 1, 727. 2Smit, M. J.; Vischer, H. F.;
3
Bakker, R. A. et al. Annu. Rev. Pharmacol. Toxicol. 2007, 47, 53. Palczewski, K.; Kumasaka, T.;
4
Hori, T. et al. Science 2000, 289, 739. Surgand, J. S.; Rodrigo, J.; Kellenberger, E.; Rognan, D.
Proteins 2006, 62, 509. 5Ruprecht, J. J.; Mielke, T.; Vogel, R.; Villa, C.; Schertler, G. F. EMBO J.
6
2004, 23, 3609. Jongejan, A.; Bruysters, M.; Ballesteros, J. A.; Haaksma, E. et al. Nat. Chem. Biol.
2005, 1, 98. 7Lopez-Rodriguez, M. L.; Morcillo, M. J.; Fernandez, E. et al. J. Med. Chem. 2005, 48,
8
2548. Deupi, X.; Dolker, N.; Lopez-Rodriguez, M.L. et al. Curr. Top. Med. Chem. 2007, 7, 999.
9
Pardo, L.; Deupi, X.; Dolker, N. et al. Chembiochem. 2007, 8, 19. 10Urizar, E.; Claeysen, S.; Deupi,
X.; Govaerts, C. et al. J. Biol. Chem. 2005, 280, 17135.
45
GREGORIO ASENSIO
i
Universidad de Valencia
iiiii
46
i
Gregorio Asensio
Universidad de Valencia. E-mail: gregorio.asensio@uv.es
ȱ
47
Ib Screening
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32
D239
s-MGL
S122
New m-MGL
MGL Activity FAAH
BV-2
Acknowledgments: This work has been supported by MCYT (Predoctoral grant to J.A.C.,
SAF-2004/07103-C02-01) and CAM (S-SAL-249-2006)
1
Dinh, T. P. et al. Proc. Natl. Acad. Sci. USA 2002, 99, 10819. 2 Di Marzo, V. et al. Curr. Opin.
3
Lipidol. 2007, 18, 129. Ghafouri, N. et al. Br. J. Pharmacol. 2004, 143: 774.
50
The development of new drugs against cancer belongs among the priorities of the
development of science and fundamental research. Because it is difficult to
discover novel agents that selectively kill tumour cells or inhibit their proliferation
without general toxicity, the use of traditional cancer chemotherapy is still very
limited. A series of pyrimidine benzo-fused seven-membered O,N-acetals were
designed and synthesized.1,2 Later on, the pyrimidine base was substituted for the
purine moiety with the objective of increasing both the lipophilicity and the
structural diversity of the target molecules.3 Here we report the design, synthesis
and biological evaluation of a series of (6’-substituted)-7 or 9-(2,3-dihydro-5H-1,4-
benzodioxepin-3-yl)-7H-or 9H-purines.
O R = cyclohexylmethyloxy, benzyloxy,
N R
H phenyloxy, allyloxy, phenylthio, benzylthio,
N 4-phenylthio, 2,4-dichlorobenzylthio, anilino
O N
N groups
The compounds have been obtained via condensation reaction between the
seven-membered acetal and the corresponding purine derivative using tin(IV)
chloride, TCS and HMDS in dry acetonitrile at 45 ºC for 72 h. It produced the N-9’
and the N-7’ cyclic alkylated purine regioisomers, which were separated by flash
chromatography. When the reaction was carried out within 5 min by microwave
irradiation at 130 ºC only the formation of the N-9’ isomer was observed. The
anticarcinogenic potential of the target molecules is reported against the MCF-7
cancer cell line. The most active compound presents an IC50 = 5,04 ± 1,68 PM
against the MCF-7 human breast cancer cell line. These results provide promising
information for further development of potent antiproliferative agents. At present,
studies are being carried out to determine the mechanism of action at the
molecular level of the most active compounds.
---------------------------------------------
1
Saniger, E.; Campos, J.; Entrena, A.; Marchal, J. A.; Súarez, I.; Aránega, A.; Choquesillo, D.; Niclós,
J.; Gallo, M. A.; Espinosa, A. Tetrahedron, 2003, 59, 5457.
2
Marchal, J. A.; Núñez, M. C.; Suárez, I.; Díaz-Gavilán, M.; Gómez-Vidal, J. A.; Boulaiz, H.;
Rodríguez-Serrano, F.; Aránega, A.; Gallo, M. A.; Espinosa, A.; Campos, J. M. Breast Cancer
Research and Treatment, 2007, 101, 000.
3
Núñez, M. C.; Rodríguez-Serrano, F.; Marchal, J. A.; Caba, O.; Aránega, A.; Gallo, M. A.; Espinosa,
A.; Campos, J. M. Tetrahedron, 2007, 63, 183.
51
a
Departamento de Química Orgánica, Facultade de Farmacia, U.S.C, Campus Sur, E-15782,
b
Santiago de Compostela. Departamento de Química Fundamental, Facultade de Química,
U.D.C., Campus da Zapateira,15071, A Coruña. cRega Institute for Medical Research,
Katholieke Universiteit Leuven, Minderbroedersstraat, B-3000 Leuven, Belgium
(±)-3 I (±)-5 I 4
Los derivados tipo (±)-3 han sido sometidos a ensayos in vitro de actividad antiviral
frente a diversos virus de ADN y ARN.
1
2
52
Kosugi, Y.; Saito, Y.; Mori, S.; Watanabe, J. et al. Antiviral Chem. Chemother., 1994, 5, 366.
Balzarini, J.; Lee, C.-K.; Herdewijn, P.; De Clercq, E. J. Biol. Chem., 1991, 266, 21509.
3
Joubert, N.; Schinazi, R. F.; Agrofoglio, L. A. Tetrahedron, 2005, 61, 11744.
4
Cho, J. H.; Bernard, D. L.; Sidwell, R. W.; Kern, E. R.; Chu, C. K. J. Med. Chem., 2006, 49, 1140.
a
Instituto de Química Médica (C.S.I.C.), Juan de la Cierva 3, E-28006 Madrid, Spain ;
b
Karolinska Institute, S-141 86 Huddinge/Stockholm, Sweden cDepartamento de
Farmacología, Universidad de Alcalá, E-28871 Alcalá de Henares, Madrid, Spain.; dRega
Institute for Medical Research, Katholieke Universiteit Leuven, Minderbroedersstraat 10, B-
3000, Leuven, Belgium
1
Pérez-Pérez, M. J.; Hernández, A. I.; Priego, E. M; Rodríguez-Barrios, F.; Gago F.; Camarasa, M.
J.; Balzarini, J. Curr. Top. Med. Chem. 2005, 5, 1205-1220.
2
Hernández, A. I., Familiar, O.; Negri, A.; Rodríguez-Barrios, F.; Gago, F.; Karlsson, A.; Camarasa,
M. J.; Balzarini. J.; Pérez-Pérez, M. J. J. Med. Chem. 2006, 49, 7766-7773.
53
1
Meyer, A.; Auernheimer, J.; Modlinger, A.; Kessler, H. Curr. Pharm. Des. 2006, 12, 2723.
2
Haubner R.; Finsinger D.; Kessler H. Angew. Chem. Int. Ed. Engl. 1997, 36, 1374.
3
Schumann F.; Müller, A.; Koksch, M.; Müller, G.; Sewald N. J. Am. Chem. Soc. 2000, 122, 12009.
54
R
O HOOC
O O O O
H
FLUOROFORO N H H
N N NH N N
H N N N CO
O H H H OH
OH
O O
COOH
55
a
Dpto. Química Orgánica I, Facultad de Ciencias Químicas. UPV/EHU. PºManuel de
Lardizabal, 3. 20018 San Sebastián. bIkerchem, S.L. Avenida de Tolosa, 72, 4ª Planta.
20018 San Sebastián.
Williamson
Henry
O
O2N
cicloadición [3+2] O
por etapas N
R OH
N H
H O
síntesis acoplamiento
de iminas peptídico
1
(a) Langley, R. R; Carlisle, R.; Ma, L.; Specian, R. D; Gerritsen, M. E.; Granger, D. N.
Microcirculation 2001, 335 (b) Tomita, Y.; Saito, K.;Oite, T.; Shimizu, S.; Sato, S. Int. J. Cancer 1995,
60, 753. (c) Papadimitriou, M. N.; Menter, D. G.; Konstantinopoulos, K.; Novpñdpm, G. L.; McIntire, L.
V. Clin. Exp. Metatasis 1999, 17, 669.
56
The ß2 adrenergic receptor (ß2AR) is an excellent model system for studying the
mechanism of Class A GPCRs activation, as it possesses a wealth of structurally
related ligands with functionally diverse properties and a well characterized agonist
binding site. Using an array of biophysical and pharmacologic approaches, we
have shown that agonist binding is translated into a series of specific
conformational changes, related to the stabilization of different active states of the
receptor, through a multistep sequential process that can be dissected into its
component parts1,2.
By putting together these results with sequence analyses and mechanistic
hypothesis derived for other GPCRs, and using 3D
models of the receptor as structural templates, we
aim to propose a specific signal transduction
pathway for the ß2AR. This pathway is formed by a
network of interactions (arrows in the accompanying
figure) extending from the ligand-binding pocket to
the cytoplasmic region of the receptor, related to G
protein binding and activation. The mechanism of
activation progresses through a linking core of
residues highly conserved in Class A GPCRs, which
includes residues of the rotamer toggle switch in
TM6, the (S/N)xxxNPxxY motif in TM7 and the DRY
motif in TM3. This pathway attempts to describe an
early stage in the activation process, when side
chain relocations have not yet been translated into
major structural changes. As this mechanism
involves residues highly conserved within Class A
GPCRs, it is expected to be shared by other members of this family.
So far, we have been able to directly detect conformational changes in the ß2AR
related to the triggering of three of the switches forming part of this proposed
activation pathway. Specifically, we have detected a change in the cytoplasmic side
of TM6 related with the trigger of the rotamer toggle switch in the same helix, the
breaking of an ionic interaction between TM3 and TM6, and a structural
rearrangement of the cytoplasmic side of TM5 relative to TM6.
We are starting to put together some of the pieces of the puzzle in the mechanism
of activation of Class A GPCRs. Although some of the pieces are still missing, we
are starting to see an emerging picture. A better understanding of the complex
process of agonist binding and activation may prove valuable for structure-based
drug discovery efforts and facilitate the design of more effective and selective
pharmaceuticals.
57
The Skaggs Institute for Chemical Biology and Departments of Chemistry and Cell Biology,
The Scripps Research Institute, 10550 North Torrey Pines Rd, La Jolla, 92037 CA, USA
succeeded in the identification of several known (200 mM), 4h, rt, PBS, H2N
OCHN ( )3
( )3
pH = 7.4 OCHN
( )3
such a platform should be feasible and will MudPIT analysis and digestion
be
Probe
Probe
Figure 1
proteins.
Acknowledgments: This work has been supported by NIH (grant DA015197) and by a
postdoctoral fellowship from MEC and the Fulbright Scholar Program (S.O.G.).
1 2 3
Aletta, J. M. et al. Trends Biol. Sci. 1998, 23, 89. Jessani, N. et al. Nat. Methods 2005, 2, 691. Salisbury, C. M.
et al. Proc. Natl. Acad. Sci. USA 2007, 104, 1171.
58
The Chembiobank project is a joint initiative between the Parc Científic Barcelona
(PCB) which, in addition to the chemical and logistics aspects of the project, will
also handle its coordination; the Universidad de Santiago de Compostela (USC)
which will develop the screening assays for the library compounds; and GRIB
(IMIM-UPF), located at the Parc de Recerca Biomédica de Barcelona, which will
carry out the virtual screening for these compounds. The implementation of the
project will require collaboration agreements with interested academic institutions.
This project is parallel and will be coordinated with initiatives with similar objectives
being developed in several European countries, in order to build eventually a
European annotated academic molecular library.
59
IDENTIFICATION OF NEUROPEPTIDE-PROCESSING
PROTEASES BY ACTIVITY-BASED PROTEOMICS
a
Disseny, síntesi i estructura de pèptids i proteïnes, Institut de Recerca Biomèdica, Parc
b
Científic de Barcelona, Josep Samitier 1-5, 08028, Barcelona. Departament de química
orgànica, Universitat de Barcelona, Martí i Franquès 1-11, 08028, Barcelona.
Neuropeptides are produced from precursor proteins by selective cleavage at specific sites.
Classical biosynthetic cleavage occurs at basic residues and a relatively small number of
12
peptidases are responsible for processing the majority of neuropeptides . However, with
the discovery and characterization of new neuropeptides, a new non-classical pathway has
been described with cleavage occurring at Tryptophane, Leucine and other amino acids.
Neuropeptide-processing peptidases involved in this new non-classical pathway are
completely unknown but essential for correct processing of certain neuropeptides.
Therefore, our aim is to identify proteases involved in the non-classical neuropeptide
processing pathway using activity based proteomics. Some substrate-inspired peptides with
a phosphonate functional group were designed and synthesized to identify serine proteases
that may be involved in the non-classical neuropeptide-processing pathway. The analyisis
of the proteome was done by in-gel analysis and MudPIT nLC-MS/MS analysis. The
synthesised peptides were labeled either with a rhodamine or a biotin tag by click
chemistry. To validate the methodology some phosphonate inhibitors for the well-known
Prolyl oligopeptidase were synthesized to identify this protein among a complex mixture of
proteins (mouse brain homogenate). Finally, two mature neuropeptide-based molecules
containing Tryptophane and Leucine in their C-terminus were synthesized to identify
unknown proteases from mouse pituitary.
Acknowledgements: We acknowledge Prof. Benjamin Cravatt for his advice and support.
1
FRICKER, L. D. American Association of Pharmaceutical Scientists (2005), 7, E449-E453.
2
CHE et al. Procedings of the National Academy of Science (2001), 98, 9971-76-
60
On this basis, the conformation of 4 and 5, their analogues 6 and 7, without the
cyclic acetal moiety, and the fluorinated compounds 8-10 has been studied. The
conformation has been derived by using an approximation which combines
molecular mechanics and dynamics methods with NMR experimental data.2,3
Finally, the inhibition activity of compounds 4, 5, 8 and 9 has been determined, and
this activity has been related with the observed conformation.
HO OH HO OH HO OH
Man
Man O Man O O
HO H
HO HO Man
OH OH OH F
OH O O O O
O
HO OH OH 4 5 8,9
O O
O X
NaO OH HO OH HO OH
OH HO Man
O Man O O
2 X=O F
HO HO Man HO
3 X = CH2 OH F
OH OH OH OH
6 7 10
This work has been supported by BQU2003 O3550 and a FPI fellowship.
1. Asensio, J. L.; Cañada, Mootoo, D. R.; Jiménez-Barbero, J. Chem. Eur. J. 2000, 6, 1035-1041
2. Jiménez-Barbero, J. Peters, T. NMR spectroscopy of glycoconjugates, 2002,Wiley-VCH
3. Jiménez-Barbero, J. et al., Chem. Eur. J. 2002, 8, 4597-612. Org. Biomol. Chem. 2003, 1, 785-92.
4. H.-J. Gabius, H.-C. Siebert, J. Jiménez-Barbero, H. Rüdiger, ChemBioChem, 2004; 5: 740-764
61
a
Departamento de Química. Facultad de Farmacia, Universidad San Pablo CEU,
Urbanización Montepríncipe, 28668- Madrid. b Departamento de Nutrición y Metabolismo,
Instituto del Frío, CSIC, Ciudad Universitaria, 28040-Madrid.
We are currently working on the design and synthesis of new series of non
steroidal compounds as potential SERMs, following a synthetic strategy based on
a photochemical electrocyclic reaction. Thus, compounds 1-4 have been
synthesized as promising scaffolds for the future development of new SERMs.
The affinity of these ligands for
ERD and ERE has been measured
in an in vitro coactivator
recruitment functional assay.3
A computational study of the
mode of binding of 1-4 with both
estrogen receptors has been
carried out in order to understand
the activity and receptor selectivity
found for these compounds.
1
Shao, W. and Brown, M,. Breast Cancer Research, 2004, 6(1), 39-52.
2
a) Hsieh, R. W.; Rajan, S. S.; Sharma, S. K.; Guo, Y.; DeSombre, E. R.; Mrksich, M.; Greene, G.L.
The Journal of Biological Chemistry 2006, 281, 17909-17919. b) Manas, E. S. et al. Journal of the
American Chemical Society 2004, 126, (46), 15106 -15119.
3
EnBio Estrogen Receptor (Alpha, Beta)/Coactivator Ligand Assay System (Cosmo Bio Co. LTD)
62
a
Dpto. Química Orgánica I. Facultad de Ciencias Químicas. UPV/EHU. Pº Manuel de
b
Lardizabal, 3. 20018 San Sebastián. Ikerchem, S.L. Avenida de Tolosa, 72, 4ª Planta.
c
20018 San Sebastián. Dominion-Pharmakine, S. L. Parque Tecnológico de Bizkaia,
d
Edificio 801, 1ª Planta. 48160 Derio, Bizkaia. Dpto. Biología Celular e Histología, Facultad
de Medicina y Odontología, UPV-EHU. 48940 Leioa, Bizkaia
BnO
O2N
O
R H
R N () O
OCH3 BnO N n
N O2N H H2 O
O n = 3,5
R
O
N BnO
H2 O O2N
OBn O
R H
O2N N () O
N n
O
O n = 1,4
NH3
1
Wang, H.H.; McIntosh, A.R.; Hasinoff, B.B.; MacNeil, B.; Rector, E.; Nance, D. M.; Orr, F.W. Eur. J.
Cancer 2002, 38, 1261.
2
(a) Soede, R.D.; Zeelenberg, I.S.; Wijnands, Y.M.; Kamp, M.; Roos, E. J. Immunol. 2001, 166, 4293.
F.P. Cossio, L. Mendoza, A.Zubia, M.Valcárcel, Y.I. Vara, M.S. Solaun, J:J López, N. Gallot, E. San
Sebastián, F. Vidal-Vanaclocha. Novel inhibitors of the LFA-1/ICAM-1 interactions, and uses thereof.
PCT/ EP2006/ 055948
64
Ia
Zoster Virus (VZV)
I Ib
OH named CF1743 Ib.
linker = Val, Ala, Leu, Phe
The synthesis of the
proposed conjugates and their ability to act as efficient substrates of DPPIV/CD26
enzyme will be described. Oral bioavailability and aqueous solubility studies of
conjugate Ib will also be reported.
1
García-Aparicio, C.; Bonache, M. C.; De Meester, I.; San-Félix, A.; Balzarini, J.; Camarasa, M. J.;
Velázquez, S. J. Med. Chem. 2006, 49, 5339.
65
a
Dpt. of Biological Organic Chemistry, IIQAB-CSIC. J. Girona, 18. 08034 Barcelona, Spain.
b
Dpt of Molecular and Cellular Biology, IBMB-CSIC. J. Girona, 18. 08034 Barcelona, Spain.
c
Bioinformatics Unit, Centro de Biología Molecular Severo Ochoa, UAM-CSIC.
Cantoblanco, 28049 Madrid, Spain.
R1 O O
N R2
R3 O N O
N N NH2
N NH2 R1 N
O
R2 O O R3 O
I II
Acknowledgements: This work was supported by grants from the Spanish Ministry of
Science and Education (MEC) (2005-00995/BQU) and CSIC grant (PIF 200580F0202). A
predoctoral fellowship from CSIC (I3P program) is also acknowledged.
1
Hofmann RM, Pickart CM. J Biol Chem 2001, 276, 27936-27943.
66
a
Departamento de Química Orgánica, ICMA, Universidad de Zaragoza-CSIC, Zaragoza.
b
Departamento de Ciencias Experimentales y de la Salud, Universidad Pompeu Fabra,
Barcelona. cCentro de Investigaciones Biológicas, CSIC, Madrid. dInstituto de Química
Física Rocasolano, CSIC, Madrid.
67
a b
Instituto de Química Médica (CSIC), Juan de la Cierva 3, 28006 Madrid. Rega Institute for
Medical Research, K.U. Leuven, Minderbroedersstraat 10, B-3000 Leuven, Belgium
Acknowledgements: MEC and the European Commission are acknowledged for financial
support
1
Sluis-Cremer, N.; Dmitrienko, G. I.; Balzarini, J.; Camarasa, M. J.; Parniak, M. A. Biochemistry
2000, 39, 1427-1433.
2
Rodríguez Barrios, F.; Pérez, C.; Lobatón, E.; Velázquez, S.; Chamorro, C.; San-Félix, A.; Pérez-
Pérez, M. J.; Camarasa, M. J.; Pelemans, H.; Balzarini, J.; Gago, F. J. Med. Chem. 2001, 44, 1853-
1865.
68
O O O
O O OH O O
H H OH
N N OH OH
HO N N OH O N
N
H Sta H O N O N
O O
Azy H
1
Nakao, Y.; Fujita, M.; Warabi, K.; Matsunaga, S.; Fusetani, N., J. Am. Chem. Soc., 2000, 122,
10462-10463
2
Schaschke, N. Bioorganic and Medicinal Chemistry Letters, 2004, 14, 855-857
3
Piró, J.; Forns, P.; Blanchet, J.; Bonin, M.; Micouin, L.; Diez, A. Tetrahedron : Asym., 2002, 13, 995-
1004.
69
70
a
Instituto de Química Médica (CSIC), Juan de la Cierva 3, 28006 Madrid. bCentro de
Biología Molecular “Severo Ochoa” (CSIC), Universidad Autónoma de Madrid,
Cantoblanco, 28049 Madrid
Riboviruses are pathogens causing human diseases, ranging from the common
cold to exotic illnesses i.e. haemorrhagic fever, and epidemic diseases i.e. AIDS,
Hepatitis C, SARS, etc. The development of broadly effective therapeutics has
been hampered by the tremendous diversity of riboviruses, as well as by their
ability to rapidly adapt and acquire resistance to treatments. Riboviruses exhibited
an extremely high mutation frequency. Maintaining such a high mutation frequency,
however, is dangerous for the virus. 1
A recently reported new antiviral strategy called “lethal mutagenesis” attemps to
exploit the high mutation frequency of riboviruses by increasing the mutation rate
even further and driving the virus population into “error catastrophe” (lethal
accumulation of errors). This new strategy was validated with ribavirin (a
mutagen).2 This suggests that RNA virus mutagens may represent a promising
new class of antiviral drugs.
We describe here the synthesis and biological studies of potential mutagenic
ribonucleosides that may be incorporated into
O
the viral genome during replication and, by
R NH N mispairing, induce lethal mutagenesis. These
N COR'
ribonucleosides bear universal bases with
N O N
ambiguous hydrogen bonding properties
HO (hydrogen bonding interactions occur but with
O
different patterns depending on the
configuration of the molecule). We have
OH OH
documented various degrees of inhibition of the
R = H, CH3, F, Cl, Br, I; R' = OCH3, NH2 replication of foot-and-mouth disease virus,
encephalomyocarditis virus and lymphocytic
choriomeningitis virus in BHK-21 cells by several base and ribonucleoside
analogues. The inhibitory activities cannot be accounted for by the toxicity of the
drugs on BHK-21 cells. We are currently carrying out experiments to identify the
steps in the life cycle of these viruses that may be affected by the drugs.
1
Graci, J. D.; Cameron, C. E. Antiviral Chem. Chemother. 2004, 15, 1.
2
Crotty, S.; Cameron, C. E.; Andino, Proc. Natl. Acad. Sci. USA. 2001, 98, 6895.
71
a
Combinatorial Chemistry Unit, Barcelona Science Park, University of Barcelona, 08028
Barcelona, Spain, amolero@pcb.ub.es
b
Department of Biochemistry and Molecular Biology, Molecular Neurobiology Unit,
University of Barcelona, 08028 Barcelona, Spain
c
IRB Barcelona, Barcelona Science Park, University of Barcelona, 08028 Barcelona, Spain
The increasing number of publications that describe multiple ligands may suggest
an on-going re-evaluation of the ‘one-disease-one-drug’ paradigm that has
dominated the pharmaceutical industry for the past few decades. For some CNS
disorders, it has been recognised that a balanced modulation of several targets
can provide a superior therapeutic effect and a better side effect profile compared
to the action of a selective ligand in a single receptor.1 A relevant example is
Parkinson’s disease (PD), where patients receiving traditional treatment based on
single dopamine interactions, obtain only partial or transient benefits at best.2
However, balanced modulation of dopamine and adenosine receptors showed
promising efficacy and fewer side effects than single-target treatments.3 Our group
has recently reported peptide-heterocycle hybrids as a good starting point for the
development of dual ligands at adenosine and dopamine receptors.4 In the present
work, we report the synthesis and biological evaluation of novel compounds with
the capacity to interact with both adenosine and dopamine receptors. The
combination of the heterocycle indolo[2,3-a]quinolizidine with several tripeptide
libraries (1 and 2) has resulted in heterocycle-peptide hybrids (Figure 1) with dual
activity at both receptors. Binding studies, pharmacology and structure-activity
relationships of these new molecules will be discussed.
O O
N R1 N
N N
N H H N H H
H NH O H H O
O N
R3 R2
R2 HN O
1 NH2 2 R1 HN
NH O
O
O
NH2
R3
1
Morphy, R.; Kay, C.; Rankovic, Z. Drug Discovery Today 2004, 9, 641-651.
2
Mercuri, N.B.; Bernardi, G. Trends in Pharmacological Sciences 2005, 26, 341-344.
3
Kanda, T.; Jackson, M.J.; Smith, L.A.; Pearce, R.K.B.; Nakamura, J.; Kase, H.; Kuwana, Y.; Jenner,
P. Experimental Neurology 2000, 162, 321-327.
4
Vendrell, M.; Angulo, E.; Casadó, V.; Lluis, C.; Franco, R.; Albericio, F.; Royo, M. Journal of
Medicinal Chemistry, in press.
72
a
Instituto de Química Médica (CSIC), Juan de la Cierva 3, 28006 Madrid. bDepartamento
de Química Fundamental Facultade de Ciencias, Universidade da Coruña, 15071 La
Coruña
TsO
O
NH2
O
O
S O
O O 1
O O O
O N N N
O O O
O O O O O O
O
S nO S nN S O
n = 1,2 n = 1,2
O O O O H3C
O
2 3 4
1
Gruner, S. A. W.; Locardi, E.; Lohof, E.; Kessler, H. Chem. Rev. 2002, 102, 491.
2
Cordeiro, A; Quesada, E.; Bonache, M. C.;Velázquez, S.; Camarasa, M. J y San-Félix, A. J. Org.
Chem., 2006, 71, 7224.
73
a b
Ikerchem, S.L. Avenida de Tolosa, 72, 4ª Planta. 20018 San Sebastián. Dpto. Química
Orgánica I. Facultad de Ciencias Químicas, UPV-EHU. Pº Manuel de Lardizabal, 3. 20018
c
San Sebastián. Dominion-Pharmakine, S. L. Parque Tecnológico de Bizkaia, Edificio 801,
d
1ª Planta. 48160 Derio, Bizkaia. Dpto. Biología Celular e Histología, Facultad de Medicina
y Odontología, UPV-EHU. 48940 Leioa, BIzkaia.
(OR)n
(RO)n N Y
H
(OR)n n,m=1,3
(RO)n R=H, Me
X=H, NO2
Y=CO2Me, CO2H, H
(OR)n
(RO)n
N
H
Figura 1.
1
Baur, J. A.; Sinclair, D. A. Nat. Rev. Drug. Discov. 2006, 5, 493.
2
Jang, M.; Cai, L.; Udenai, G. O.; Slowing, K. V.; Thomas, C. F.; Beecher, C. W. W.; Fong, H. H. S.;
Farnsaworth, N. R.; Kinghorn, A. D.; Menta, R. G.; Moon, R. C.; Pezzuto, J. M. Science 1997, 275,
218.
74
R
identification of the
O
N REPORTER N
H TAG H
to convert these O
OH
compounds into
HO
O 5
O REPORTER
probes of high
TAG O
REPORTER
TAG
towards CB1.
1
Karanian, D.A.; Bahr, B.A. Curr. Mol. Med. 2006, 6, 677.
2
Prescher, J.A.; Bertozzi, C.R. Nat. Chem. Biol. 2005, 1, 13.
75
The approach presented herein represents the first contribution to the rational
design of agents acting at the recently identified 5-HT6 serotonin receptor.
1
Glennon, R. A. J. Med. Chem. 2003, 46, 2795-2812.
2
López-Rodríguez, M. L.; Benhamú, B.; de la Fuente, T.; Sanz, A.; Pardo, L.; Campillo, M., J. Med.
Chem., 2005, 48(13), 4216-4219.
3
López-Rodríguez, M. L.; Benhamú, B.; Martín-Fontecha, M.; de la Fuente, T. European Patent in
preparation.
76
UCM-5600
HBA I PI
The influence of the different pharmacophoric elements has been analyzed using
computational simulation studies that have determined the molecular details of
ligand-receptor interaction. The models are consistent with the binding data and
have revealed that a hydrogen bond between the compound and Ser6.55 is the
key for 5-HT7/5-HT1A selectivity in this family of ligands. In particular, ligand UCM-
3307 (HYD1 = B; spacer = (CH2)4; HYD2 + HYD3 = 1,2,3,4-tetrahydroisoquinoline)
exhibits high 5-HT7R affinity and selectivity over 5-HT1AR (5-HT7: Ki = 23 nM; 5-
HT1A: Ki = 219 nM), and is being characterized for functional behaviour. The
hypothesis proposed herein for 5-HT7/5-HT1A selectivity represents a rational
approach that should help in the development of new specific 5-HT7R ligands.
This work was supported by Ministerio de Ciencia y Tecnología (SAF2004-07103-C02-
01) and Comunidad Autónoma de Madrid (SAL-0249/2006). R. A. Medina is also grateful to
Ministerio de Educación y Ciencia for a F.P.U. predoctoral grant.
1
(a) Vanhoenacker, P.; Haegeman, G.; Leysen, J.E. Trends Pharmacol. Sci., 2000, 21, 70. (b)
López-Rodríguez, M.L.; Benhamú, B.; Morcillo, M.J.; Porras, E.; Lavandera, J.L.; Pardo, L. Curr. Med.
Chem. - CNSA, 2004, 4, 203. (c) Hedlund, P.B.; Sutcliffe, J.G. Trends Pharmacol. Sci., 2004, 25, 481.
2
López-Rodríguez, M.L.; Porras, E.; Morcillo, M.J.; Benhamú, B.; Soto, J.L.; Lavandera, J.L.; Ramos,
J.A.; Olivella, M.; Campillo, M.; Pardo, L. J. Med. Chem., 2003, 46, 5638.
77
a
Instituto de Química Física Rocasolano-CSIC, Serrano 119, 28006 Madrid.
b
Instituto de Química Médica-CSIC, Juan de la Cierva, 3, 28006 Madrid.
VEGF-A81-91: MRIKPHQGQHI
MRCKPHQGCHI
S S
Vammin69-80: MRVNPRTQSSKM
MRCNPRTQSCKM
S S
1
Pan, B.; Li, B.; Russell, S.J.; Tom, J.Y.; Cochran, A.G.; Fairbrother, W.J. J. Mol. Biol. 2002, 316,
769-787.
2
Keyt, B.A.; Nguyen, H.V.; Berleau, L.T.; Duarte, C.M.; Park, J.; Chen, H.; Ferrara, N. J. Biol. Chem.
1996, 271, 5638-5646.
78
Luis Berradea, Silvia Pérez Silanesa, Paula Egeaa, Ignacio Aldanaa, Laura
Giuratob, Salvatore Guccioneb, Lise Roman Moltzauc, Finn Olav Levyc,
Gemma Molinaroc, Ferdinando Nicolettic and Antonio Mongea
a
Unidad de Investigación y Desarrollo de Medicamentos. Centro de Investigación en
Farmacobiología Aplicada (CIFA). Universidad de Navarra 31008, Pamplona. Spain.
b
Molecular Modelling Laboratory. Dipartamento di Scienze Farmaceutiche University of
c
Catania. Viale Andrea Doria 6, I-95125, Catania. Italy. Department of Pharmacology.
d
University of Oslo, P.O. Box 1057 Blindern, 0316 Oslo, Norway. I.R.C.C.S. Instituto
Neurologico Mediterraneo Neuromed. Localitá Camerelle, 86077 Pozzilli (IS) Italy.
Results of receptor binding studies performed on rat brain tissue with these
new compounds will be presented along with molecular modelling studies
combining state of art softwares and homology modelling with the additional aim to
investigate the 5-HT7 vs. 5-HT1A binging.
1
Morphy R. and Rankovic Z. J. Med. Chem. 2005, 48, 6523-6543.
2
Hedlund P. B. and Sutcliffe J. G. Trends in Pharmacol. Sci. 2004, 25, 9, 481-486.
3
Pérez S. et al., Pharmazie 2004, 59, 499-501
79
a
Instituto de Química Médica (CSIC),Juan de la Cierva 3, 28006 Madrid, Spain. b Centro
de Química Orgánica Manuel Lora-Tamayo (CSIC), Juan de la Cierva 3, 28006 Madrid,
Spain
1
Casanova, E.; Hernández, A.I.; Priego, E.M.; Liekens, S.; Camarasa, M. J.; Balzarini, J.; Pérez-
Pérez, M. J. J. Med. Chem. 2006, 49, 5562-5570.
2
Lichtenthaler F. W.; Kitahara K.; Riess W. Nucleic Acids Res. 1978, 4, s115-s118.
3
Luzzio F. A.; Menes, M. E. A. J. Org. Chem. 1994, 59, 7267-7272.
4
Gates, K. S.; Nooner, T.; Dutta, S. Chem Res. Toxicol., 2004, 17, 839-856.
80
a
Instituto de Química Médica (CSIC), Juan de la Cierva 3, E-28006 Madrid, Spain.
b
Departamento de Farmacología, Universidad de Alcalá, E-28871 Alcalá de Henares,
Madrid, Spain. cRega Institute for Medical Research, Katholieke Universiteit Leuven,
Minderbroedersstraat 10, B-3000, Leuven, Belgium.
1
Pérez-Pérez, M. J.; Priego, E. M.; Hernández, A. I.; Camarasa, M. J.; Balzarini, J.; Liekens, S. Mini
Rev. Med. Chem. 2005, 5, 1113-1123.
2
Liekens, S.; Hernández, A. I.; Ribatti, D.; De Clercq, E.; Camarasa, M. J.; Pérez-Pérez, M. J.;
Balzarini, J. J. Biol. Chem. 2004, 279, 29598-29605.
3
El Omari, K.; Bronckaers, A.; Liekens, S.; Pérez-Pérez, M. J.; Balzarini, J.; Stammers, D. K.
Biochem. J. 2006, 399, 199-204.
4
http://lorentz.immstr.pasteur.fr/nomad-ref.php
5
http://www.scripps.edu/pub/olson-web/dock/autodock
81
HN NBn O
N NBn
O R1 OTBDMS
R1 OP
S piperazinas y cetopiperazinas
N p-Tol
H2 N NHCbz
R1
R 1 CO 2CH 3
diaminas vecinales
Agradecimientos: DGI MEC por los proyectos BQU2003-0292, CTQ2005-04632/BQU y
CTQ2006-04522/BQU). JANSSEN-CILAG por la financiación adicional de nuestro grupo.
CSIC, CM y MEC por las becas de NL, MU, EC, MA, AF, AC, IO, IC.
1
(a) Viso, A.; Fernández de la Pradilla, R.; Flores, A. Tetrahedron Letters 2006, 47, 8911-8915. (b)
Fernández de la Pradilla, R.; Castellanos, A.; Fernández, J.; Lorenzo, M.; Manzano, P.; Méndez, P.;
Priego, J.; Viso, A. J. Org. Chem. 2006, 71, 1569-1575.
82
O O
+
R7 N
R
+
R6 N
O
1
The synthesis of these compounds was carried out by a base-catalyzed
Claisen-Schmidt condensation. The conditions of temperature used by other
authors2 should be modified, establishing a required temperature of –10ºC in order
to obtain the desired compounds.
Synthesized compounds were evaluated at the National Cancer Institute (NCI,
Bethesda, USA) against 60 human tumor cell lines at a single dose of 100 µM.
Some of the compounds, which exhibit significant growth inhibition, are being
evaluated against the 60 cell panel at five concentration levels.
Acknowledgements: This work has been carried out thanks to the financial support of the
FIS project (1051005, October 2005) and we want to express our gratitude to the National
Cancer Institute (NCI, Bethesda, USA) for the evaluation of the anticancer activity. A.
Burguete. was awarded a PhD scholarship supported by the “Gobierno de Navarra”.
Zarranz, B.; Jaso, A.; Aldana, I.; Monge, A. Bioorg. Med. Chem. 2004, 12, 3711-3721
2
Nielsen, S. F.; Boesen, T.; Larsen, M.; Schonning, K.; Kromann, H. Bioorg. Med. Chem. 2004, 12,
3047-3054.
83
a
Instituto de Química Médica (C.S.I.C.), Juan de la Cierva 3, E-28006 Madrid, Spain ;
b
Institut Pasteur, Unité de Chimie Organique, CNRS URA 2128, 28, Rue du Dr. Roux,
75724 Paris Cedex, France ; cINSERM, U554, Université Montpellier 1 et 2, CNRS,
UMR5048, Centre de Biochimie Structurale, 34090-Montpellier, France
1
Pochet, S.; Dugue, L.; Douguet, D.; Labesse, G.; Munier-Lehmann, H. ChemBioChem 2002, 3, 108
2
Munier-Lehmann, H.; Chaffotte, A.; Pochet, S.; Labesse, G. Protein Sci. 2001, 10, 1195.
84
a
Departamento de Farmacología, Universidad de Alcalá, E-28871 Alcalá de Henares,
Madrid, Spain. bInstituto de Química Médica (C.S.I.C.), Juan de la Cierva 3, E-28006
Madrid, Spain. cRega Institute for Medical Research, Katholieke Universiteit Leuven,
Minderbroedersstraat 10, B-3000, Leuven, Belgium. dInstitut Pasteur, Unité de Chimie
Organique, CNRS URA 2128, 28, Rue du Dr. Roux, 75724 Paris Cedex, France.
1
Pochet, S.; Dugue, L.; Douguet, D.; Labesse, G.; Munier-Lehmann, H.ChemBioChem 2002, 3, 108-
110.
2
Munier-Lehmann, H.; Chaffotte, A.; Pochet, S.; Labesse, G. Protein Sci. 2001, 10, 1195-1205
3
Pérez-Pérez, M. J.; Hernández, A. I.; Priego, E. M; Rodríguez-Barrios, F.; Gago F.; Camarasa, M.
J.; Balzarini, J. Curr. Top. Med. Chem. 2005 5, 1205-1220.
4
Mammoth program, URL: http://ub.cbm.uam.es/mammoth/pair/index3.php
85
S 6 R2 = H; R6 = Cl 12 R2 = H; R6 = Allyloxy
N N
7 R2 = H; R6 = Br 13 R2 = H; R6 = OBn
8 R2 = H; R6 = SMe 14 R2 = H; R6 = SBn
O
N R6 9 R2 = H; R6 = OPh 15 R2 = H; R6 = OCH2C6H11
N 10 R2 = H; R6 = SPh 16 R2 = Cl; R6 = Cl
R2 11 R2 = H; R6 = NHPh
The anticarcinogenic potential of the target molecules is reported against the MCF-
7 human breast cancer cell line.
--------------------------------------------
1
Saniger, E.; Campos, J.; Entrena, A.; Marchal, J. A.; Súarez, I.; Aránega, A.; Choquesillo, D.;
Niclós, J.; Gallo, M. A.; Espinosa, A. Tetrahedron, 2003, 59, 5457.
2
Marchal, J. A.; Núñez, M. C.; Suárez, I.; Díaz-Gavilán, M.; Gómez-Vidal, J. A.; Boulaiz, H.;
Rodríguez-Serrano, F.; Aránega, A.; Gallo, M. A.; Espinosa, A.; Campos, J. M. Breast Cancer
Research and Treatment, 2007, 101, 000.
3
Núñez, M. C.; Rodríguez-Serrano, F.; Marchal, J. A.; Caba, O.; Aránega, A.; Gallo, M. A.;
Espinosa, A.; Campos, J. M. Tetrahedron, 2007, 63, 183.
86
R N N R
n n
N + +
R2 ' O Me O N R2' N
+ O N N
N N N + + R R
N N N –
2A
A– R R1 R2 R1 R2 R1 R2 R R 2A
–
87
a
Laboratori de Química Orgànica, Facultat de Farmàcia, Universitat de Barcelona, Avda.
Joan XXIII s/n, 08028 Barcelona, Spain. bDepartamento de Química Física y Analítica,
Facultat de Química, Universidad de Oviedo, 33006 Oviedo, Spain. cLaboratorios del Dr.
Esteve, S.A., Avda. Mare de Déu de Montserrat, 221, 08041 Barcelona, Spain.
N R6' N
X X X X
SMe SO2Me SMe
F
1X=O 2X=O 3X=O R6' = H 5X=O
6 X = N-OH 4X=O R6' = Me 9 X = N-OH
7 X = N-OH R6' = H
8 X = N-OH R6' = Me 6 9
Oximes 6, 7 and 9 were examined in the solid state by X-ray crystallography,
providing evidence of H-bonding networks: in particular, the relevance of crystal
packing controlled by H-bond interactions based on homomeric intermolecular
oxime···oxime interactions for 6 and oxime···N(pyridyl) interactions for 7 and 9.
Moreover, cooperative O-H···N(py) and CH/π hydrogen bonds were disclosed in
crystallines 7 and 9.
Acknowledgments:This research was supported by Laboratorios Dr. Esteve S.A.,
Barcelona, Spain, through project FBG2001/2002-301362, Fundació Bosch i Gimpera-
Universitat de Barcelona, Spain. N.M. is grateful to Laboratorios Dr. Esteve S.A. for a
post-doctoral fellowship. Thanks are also due to the DGI-MEC (CTQ2006-1182/BQU) and
the AGAUR, Grup de Recerca Consolidat 2005SGR00158.
88
a
Laboratori de Química Orgànica, Facultat de Farmàcia, Universitat de Barcelona, Av. Joan
XXIII s/n, 08028-Barcelona, Spain. bDepartament de Cristal·lografia, Mineralogia i Dipòsits
Minerals, Universitat de Barcelona, C. Martí i Franquès s/n, 08028-Barcelona, Spain.
R R R R
+ + Br
N N N N
2Br
Pd
N N N N
Br
Pd(OAc)2
O O
O O O
O
O O
2a R= nBu 1a R= nBu
2b R= iPr 1b R= iPr
1a
The Suzuki-Miyaura reaction was used to study their activity as catalysts, when
prepared either in situ or from a well-defined complex.
89
Imidazolium salts have been studied for more than 40 years as models of
carbon acids, and for their applications as ionic liquids and N-heterocyclic carbene
(NHC) precursors. Stable NHCs may be formed by deprotonation of the C(2)–H of
the imidazolium cations, evidence of which is H/D exchange of the proton in D2O
solution. Recently, the kinetic acidity of imidazolium cations, including H/D
exchange rates, carbon-proton acidity, and carbene precursor stability has been of
great interest to physical and organic R' X– k' R' X–
chemists, as the formation of NHCs is often a
N CD3OD N
crucial step in catalytic organic and
+ H + D
organometallic reactions.
N N
The deprotonation of imidazolium salts to
R R' R
N-heterocyclic carbenes is often a decisive
step in modern catalytic reactions. Because N
protonation of the NHC is fast compared to
the formation of the NHC, the H/D exchange N
rates of the C(2)–H give an indirect estimate R
for the reaction rate of its formation.
Therefore, we studied the H/D exchange of the C(2)–H of several bis-(imidazolium)
dications in methanol-d4.
R'
R5 '
R 6' R4 '
n R N N R
n
R2 ' + – +
N N N N 2X
+ + R5 R5 N N
+ + R R
–
N 2X N N N
R R R4 R3 R3 R4
2X–
R'
The influence of the counterion, concentration and presence of D2O has been
studied. The observed exchange rates might give a rational for the suitability of the
imidazolium salts as precursors of N-heterocyclic carbenes.
Acknoledgment. This research was supported by the Dirección General de Investigación
(Ministerio de Educación y Ciencia) project CTQ2006-1182/BQU. Thanks are also due to
the AGAUR, 2005SGR00158 (Generalitat de Catalunya). S. R. thanks the AGAUR
(Generalitat de Catalunya) for a F.P.I. fellowship.
90
a
Unidad en Investigación y Desarrollo de Medicamentos. Centro de Investigación en
Farmacobiología Aplicada (CIFA). University of Navarra. C/Irunlarrea, s/n, 31080
Pamplona. Spain.
O N
N N
H
R1
R
1
R= 4-F, 2-OMe, 4-OMe, 3-CN
R1= H, 3-hydroxy-3-methyl-2-butanone
As part of a general project with the main target of finding new MCH-R1
antagonists, we have synthesized and evaluated for in vitro binding 1R receptor of
the MCH a new series of biphenylmethyl urea derivatives (structure 1) in order to
develop SAR. Prior hit-to lead efforts resulted in the identification of compounds
with good affinity. We have also carried out binding assays with the hERG channel
to evaluate the toxicity of the new compounds.
Acknowledgments: We wish to thank the Gobierno de Navarra for the grants given to
Javier Ceras and Nuria Cirauqui.
1
Marsh, D.J.; Weingarth, K.T.; Novi, D.E.; Chen, H.Y.; Trumbauer, M.E.; Chen, A.S.; Guan, X.M.;
Jiang, M.M.; Feng, Y.; Camacho, R.E.; Shen, Z.; Frazier, E.G.; Yu, H.; Metzger, J.M.; Kuca, S.J.;
Shearman, L.P.; Gopal-Truter, S.; Macneil, D.J.; Strack, A.M.; MacIntyre, D.E.; Vand der Ploeg, L.H.;
Quian, S. Proc. Natl. Acad. Sci. U.S.A. 2002, 99, 3240.
2
Chen, Y.; Hu, C.; Hsu, C.K.; Zhang, Q.; Bi, C.; Asnicar, M.; Hsiung, H.M.; Fox, N.; Slieker, L.J.;
Yang, D.D.; Heiman, M.L.; Shi, Y. Endocrinology. 2002, 143, 2469.
3
Kowalsky, T.J.; McBriar, M.D. Expert. Opin. Investig. Drugs. 2004, 13, 1113.
91
a
Unidad en Investigación y Desarrollo de Medicamentos. Centro de Investigación en
Farmacobiología Aplicada (CIFA). University of Navarra. C/Irunlarrea, s/n, 31080
Pamplona. Spain.
O O R2
S R2 R1 Amide A (CH2)n N
linker
R1 N N basic
H R3
1 2
Acknowledgements: We wish to thank the Gobierno de Navarra for the grants given to
Nuria Cirauqui and Javier Ceras.
1
Shimada, M.; Tritos, N. A.; Lowell, B. B.; Flier, J. S.;Maratos-Flier, E. Nature 1998, 396, 670.
2
Ludwig, D.S.; Tritos, N. A.; Mastaitis, J. W.; Kulkarni, R.; Kokkotou, E.; Elmquist, J.; Lowell B, Flier,
JS, Maratos-Flier EJ. J. Clin. Investig. 2001, 107, 379-386.
3
Takekawa, S.; Asami, A.; Ishihara, Y.; Terauchi, J.; Kato, K.; Shimomura, Y.; Mori, M.; Murakoshi,
H.; Kato, K.; Suzuki, N.; Nishimura, O.; Fujino, M. Eur. J. Pharm. 2002, 438, 129.
92
a
Unidad en Investigación y Desarrollo de Medicamentos. Centro de Investigación en
Farmacobiología Aplicada (CIFA). University of Navarra. C/Irunlarrea, s/n, 31080
Pamplona. Spain.
Obesity is a worldwide epidemic. The lack of efficacy of the available obesity drugs
makes this disease one of the most attractive therapeutic targets. Melanin-
concentrating hormone (MCH), a cyclic 19-amino acid neuropeptide, is believed to
play a critical role as a regulator of feeding behaviour and energy balance based
on several lines of evidence. Interestingly, mice overexpressing the MCH gene
appear to give an hyperphagic and obese phenotype and display insulin-
resistance, whereas transgenic.1 MCH-R1 knockout mice are lean, resistant to diet-
induced obesity and show hyperactivity.2 Consequently, selective MCH-R1
antagonism could provide a viable treatment for obesity.
As a continuation of our research in this field and based on previous assays with
an substituted biphenylmethylurea core, we have synthesized and evaluated a
novel series of urea derivatives (structure 1) with the aim of improving the MCH-R1
in vitro activity maintaining hERG affinity.3
O N
R1
O N N
aromatic region H
linker
N N basic amine
H H
R
1
carboxamide/urea core 1 4-OMe, 3-CN
R= 4-F, 2-OMe,
R1=isopropyl, 3-hydroxy-2,2-dimethylpropyl,
1,1-dihydroxyethylpropyl,
[1-(acethyloxymethyl)-1-methyl]ethyl,
(1,1-diacethyloxymethyl)propyl
SAR was explored, suggesting that optimal binding with the receptor was achieved
when the biphenylmethyl group and the linker were substituted on the same
nitrogen of the urea moiety. We have also carried out binding assays with the
hERG channel to evaluate the toxicity of the new compounds.
Della-Zuana, O.; Presse, F.; Ortola, C.; Duhault, J.; Nahon, J.L.; Levens, N. Int. J. Obes. 2002, 26,
1289.
2
Chen, Y.; Hu, C.; Hsu, C. K.; Zhang, Q.; Bi, C.; Asnicar, M.; Hsiung, H. M.; Fox, N.; Slieker, L. J.;
Yang, D. D.;Heiman, M. L.; Shi, Y. Endocrinology. 2002, 143, 2469.
3
Galiano, S.; Ceras, J.; Cirauqui, N; Pérez, S; Juanenea, L.; Rivera, G.; Aldana, I.; Monge, A. Bioorg.
Med. Chem. 2007, 15, 3896.
93
R
O
H3CO O H
H O
CH3
O N CH3
E R
O H N
H O H
H3C CH3
N O R1 Z
A B C
N
H3CO H 6
O Z
R
R
O
Compuesto Z R
O H
Saframicina A (1) CN NH-CO-COCH3 H O
N CH3
Saframicina S (2) OH NH-CO-COCH3 R
Saframicina B (3) H NH-CO-COCH3 HN
H
Jorumicina (4) OH O-CO-CH3 O Z
Renieramicina M (5) CN (E)- O-CO-C(CH3)=CH-CH3
7
1
Revisión: Scott, J. D.; Williams, R. M. Chem. Rev. 2002, 102, 1669.
94
Tuberculosis (TB) is one of the leading infectious causes of death in the world
and has become a growing global health problem. According to the World Health
Organization, in 2003, 8.8 million new TB cases arose, and an estimated 1.7
million deaths resulting from TB occurred. One of the major problems of TB is the
development of new active compounds against multidrug-resistant tuberculosis
strains. In this sense several quinoxaline 1,4-di-N-oxide derivatives have been
reported as in vitro anti-Mycobacterium tuberculosis agents.1,2 The quinoxaline-3-
methyl-2-ketone or 2-amide 1,4-dioxide derivatives (1-10), active in previous
assays, were selected for the determination of MIC against different single and
multiply drug resistant strains of Mycobacterium tuberculosis and non-replicating
persistent bacteria, MBC, maximum tolerated dose, oral bioavailability, in vivo
efficacy testing an potential for cross resistance with another bio-reduced drug.
Comp. R6 R7 R2
O O 1 H H CH3
+ 2 H CH3 CH3
R7 N
R2 3 H OCH3 CH3
4 H F CH3
+
R6 N CH3 5 H Cl CH3
6 CH3 CH3 CH3
O
7 H H Ph
8 H H NH-Ph
9 H H NH-PH-(o)CH3
10 H Cl NH-PH-(o)CH3
One compound, 5, with an MIC of 0.78 µg/mL was bactericidal with an MBC 2,
equally active on non-replicating persistent organisms, and active on multiply drug-
resistant clinical isolates. This compound was orally bioavailable and efficacious in
the mouse efficacy.
Acknowledgments: Antimycobacterial data was provided by the Tuberculosis
Antimicrobial Acquisition and Coordinating Facility (TAACF) through a research and
development contract with the U.S. National Institute of Allergy and Infectious Diseases
(NIAID). R. Villar is indebted to the Navarra Government for a grant.
1
Jaso, A.; Zarranz, B.; Aldana, I.; Monge, A. Eur J. Med. Chem. 2003, 38, 791.
2
Zarranz, B.; Jaso, A.; Aldana, I.; Monge, A. Bioorg. Med. Chem. 2003, 11, 2149
95
O
+ N O O
Ra N + N
Ra N
+ Cl
X DMF O
+ H
Rb N N + X
TEA Rb N N
O H O H
O O O
+ N + N
Ra N Ra N
DMF,EDCI
+ HO
X NO2 O
+ H DMAP + X NO2
Rb N N Rb N N
O H O H
1
Ortega, M. A.; Sainz, Y.; Montoya, M.; et. al. Arzneim.-Forsch. 2002, 52, 113.
2
Martínez-Crespo, F. J.; Palop, J. A.; Sainz, Y.; et al. J. Heterocyclic Chem. 1996, 33, 1671.
3
Tangallapally, R. P.; Yendapally, R.; Lee, R. E.; et al. J. Med. Chem. 2004, 47, 5276.
96
O O O O O O
+ + +
R7 N R7 N H N
O Y O O
+ + +
N CH3 R6 N H N
O O O
H W
12-18
Previous leaders 1-4
R7=H,Cl; Y=CH3,Ph 5-11
1
WHO. Weekly epidemiological record. Nº 15, 2003, 78, 121.
2
Jaso, A.; Zarranz, B.; Aldana, I.; Monge, A. J. Med. Chem. 2005, 48, 2019.
3
Jaso, A.; Zarranz, B.; Aldana, I.; Monge, A. Eur. J. Med. Chem. 2003, 38, 791.
4
Zarranz, B.; Jaso, A.; Aldana, I.; Monge, A. Bioor. Med. Chem. 2003, 11, 2149.
97
Irene Suarez del Villar,a Ana Gradillas,a Ricardo Martinez-Murillo,b Alfredo Martinez Serrano,b y
Javier Pérez-Castellsa*
a
Facultad de Farmacia, Dpto. Química, Universidad San Pablo CEU, Urb. Montepríncipe, ctra.
Boadilla km 5,300 Boadilla del Monte, 28668 Madrid. E-mail: jpercas@ceu.es.
b
Instituto Ramón y Cajal-CSIC. Avda. Doctor Arce 37, 28002 Madrid
Agradecimientos: Financiación de este proyecto por el MEC (proyecto CTQ2006-00601/BQU). ISV. agradece a la
FUSP-CEU una beca predoctoral.
98
R3 CO2 Me
1 2
R4 1: R 3 y R 4 = H
10 3
9 10b N
8
7 6 4 R1 2 : R 3 = Br, allyl, prenyl, OH, CH 3
N 5
R2 R 4 = H, Br
NH
Agradecimientos: Pharma Mar, S. A. por la evaluación antitumoral; CSIC por las becas
I3P a P. Ventosa-Andrés y a J. A. González-Vera; CICYT por la financiación (SAF2006-
01205).
1
(a) González-Vera, J. A.; García-López, M. T.; Herranz, R. Org. Lett. 2004, 6, 2641-2644. (b)
González-Vera, J. A.; García-López, M. T.; Herranz, R. J. Org. Chem. 2005, 70, 8971-8976.
99
a
Instituto de Química Médica (CSIC), Juan de la Cierva 3, 28006, Madrid, Spain. bRega
Institute for Medical Research, Katholieke Universiteit Leuven, Minderbroedersstraat 10, B-
3000 Leuven, Belgium.
O R
NH 2
CO2 R 1 CO2 R 1 N
H
N O N O N O
O O
X X O
R2 R2 CH 2Ph
1 2 3 R= Me
4 R= CH 2 Phe
CH 2 CH(CH3 )2, CH(CH 3) 2
(CH2 )4 NH 2 , (CH 2)2 CO2H
This communication deals with the synthesis and evaluation against HCMV in cell
cultures of this new family of azetidines derived from amino acids.
1
Waxman, L.; Darke, P. L. Antiviral Chemistry & Chemotherapy 2000, 11, 1-22.
2
Gerona-Navarro, G; Pérez de Vega, M.J; García-López, M.T; Andrei, G; Snoeck, R;
Balzarini, J; De Clercq, E; González-Muñiz, R. J. Med. Chem. 2005, 48, 2612-2621.
100
a
Instituto de Química Médica, CSIC, Juan de la Cierva 3, 28006 Madrid. bDpto. de Ciencias
Experimentales y de la Salud, Universidad Pompeu Fabra - Parc de Recerca Biomèdica de
Barcelona, Dr Aiguader 88, 08003 Barcelona. cInstituto de Química Física Rocasolano,
CSIC, Serrano 119, 28006 Madrid. dDept. of Pharmacology, Weill Medical College of
Cornell University, 1300 York Avenue, Box 70, Whitney Pavilion, W-506, New York, 10021.
Figure 1
This communication addresses the solid-phase synthesis, molecular modelling,
NMR studies and preliminary biological results of compounds 1-7.
____________________
1
Pattarawarapan, M. and Burgess, K. J. Med. Chem., 2003, 46, 5277.
2
Fletcher, J. M., Hugues, R. A. J. Pept. Sci. 2006, 12, 515.
3
Robinson, R. C. et al. Protein Science 1999, 8, 2589.
101
R
Cl
H2N N N
N
H 3, R = Cl
NH2 N N N
N 4, R = Ph
HO HO HO 5, R = HNCH(CH2)2
6, R = NH2
S S S 7, R = OH
1 2
1
Mahony, W. B.; Domin, B. A.; Daluge, S. M.; Zimmerman, T. P. Biochemical Pharmacology 2004,
68, 1797.
2
Arts, E. J.; Wainberg, M. A. Antimicrob. Agents Chemother. 1996, 40, 527.
3
Daluge, S. M.; Martín, M. T.; Sickles, B. R.; Livingston, D. A. Nucleosides, Nucleotides & Nucleic
Acids 2000, 19, 297.
4
Abeijón, P.; Blanco, J. M.; Fernández, F.; García, M. D.; López, C. Eur. J. Org. Chem. 2006, 759.
102
X Amination RO
N Reductive OO
RO NH2-X
n n
RO RO
O O O O
n = 0, 1
Hydrolysis
Oxidation
O
O Fragmentation RO
RO OH Alkilation
n
n RO
RO O OHCO O O
O
1
(a) Matthews, J. M.; Greco, M. N.; Hecker, L. R.; Hoekstra, W. J.; Andrade-Gordon, P.; Garavilla, L.;
Demarets, K. T.; Ericsson, E.; Gunnet, J. W.; Hageman, W.; Look, R.; Moore, J. B.; Maryanoff, B. E.
Bioorg. Med. Chem. Lett. 2003, 13, 753. (b) Andrés, J. I.; Alcázar, J.; Alonso, J. M.; Díaz, A.;
Fernández, J.; Gil, P.; Iturrino, L.; Matesanz, E.; Meert, T. F.; Megens, A.; Sipido, V. K. Bioorg. Med.
Chem. 2002, 12, 243.
2
Toyooka, N.; Nemoto, H.; Kawasaki, M.; Garrafo, H. M.; Spande, T. F; Daly, J. W. Tetrahedron
2005, 61, 5139.
103
one-pot
Z O decarboxylation Z
N addition of a N B
OH nitrogen bases
(OR)n (Y)n
Decarboxilation X
MeO O E-iodination MeO O O MeO O
addition of bases KOH
O X
N N N NH MeOH N
OH N
OTMS O
O N
X I O
N
TMSO N
X = H, Me, F, I
1
(a) Yokoyama, M.; Momotake, A. Synthesis 1999, 15411554. (b) Koþalka, P.; Pohl, R.; Rejman, D.;
Rosenberg, I. Tetrahedron 2006, 62, 57635774 y referencias citadas allí.
104
105
a
Departamento de Química Orgánica y Farmacéutica. Universidad de Navarra. Irunlarrea,
1. 31008-Pamplona
b
Oncología. Centro de Investigación Médica Aplicada (CIMA). Universidad de Navarra. Pío
XII, 53. 31008-Pamplona
1
Brinkman, M.; Reulen, R.C.; Kellen, E.; Buntinx F.; Zeegers, M.P. Eur. J. Cancer 2006, 42, 2463-
2471.
2
Li, G.X.; Hu, H.B.; Jiang, C.; Schuster, T.; Lu, J.X. International J. Cancer 2007, 120, 2034-2043.
3
Goel, A.; Fuerst, F.; Hotchkiss, E.; Boland, R.; Boland, C.R. Cancer Biol. & Ther. 2006, 5, 529-535.
106
Daniel Planoa, Esther Morenoa, María Fonta, Juan Antonio Palopa, Ignacio
Encíob , Carmen Sanmartína
a
Departamento de Química Orgánica y Farmacéutica. Universidad de Navarra. Irunlarrea,
1. 31008-Pamplona
b
Departamento de Ciencias de la Salud. Universidad Pública de Navarra. Campus de
Arrosadía s/n. 31006-Pamplona
R
O Se O
N N
R' H R'
X X
1
Font, M.; Ardaiz, E.; Cordeu, L.; Cubedo, E.; García-Foncillas, J.; Sanmartín, C.; Palop, J.A. Bioorg.
Med. Chem. 2006, 14, 1942-1948.
2
Cordeu, L.; Cubedo, E.; Bandrés, E.; Rebollo, A.; Sáenz, X.; Chozas, H.; Domínguez, M.V.;
Echeverría, M.; Mendívil, B.; Sanmartín, C.; Palop, J.A.; Font, M.; Garcia-Foncillas, J. Bioorg. Med.
Chem. 2007, 15, 1659-1669.
3
Sanmartín, C.; Font, M.; Palop, J.A. Mini-Rev. Med. Chem. 2006, 6, 639-650.
4
Letavayova, L.; Vlckova, V.; Brozmanova, J. Toxicology. 2006, 227, 1-14.
107
Cl
N
H3C OH
N
N N
N NH
Figure 1
Water is a cheap, readily available nontoxic solvent for use in chemistry. There are, however,
problems with the use of water, such as the solubility of substrates. With its high dielectric
constant, water is also a potentially useful solvent for microwave-mediated synthesis. As well as
benefiting from the rate enhancement effects found when using microwave heating, when water
is heated well above its boiling point in sealed vessels, organic substrates can become partially
soluble.2 We have prepared Losartan using this technique in most of the steps.3,4,5 The synthetic
strategy has been redesigned in relation with the conventional preparation methods, to prevent
the use of protecting groups, thus simplifying the whole procedure.
OH OH
OH
Br
CN Na2CO3, TBAB NaN3
+ N N
Pd(OAc)2, H2O ZnBr N
CN
B(OH)2 2 87% H2O 66% N
H
1 60W 200W
120 ºC 190 ºC 4
10 min 3 60 min
BrH 100ºC
60 W
40% 20min
O Cl
HO N H3C N
H N H
Cl Cl Br
N N 6 N
H O
K2CO3
NaBH4
CH3 CH3 N N
N N MeOH N N CH3CN N
N N N
N N 100ºC H
H H 60 W
50min
5
8 Losartan 7 71%
1
Carini, D. J.; Duncia, J. V.; Wang, S. M.; US 5138069, 1986.
108
2
Leadbeater, N. E., Marco, M., J. Org. Chem. 2003, 68, 888.
3
Leadbeater, N. E.; Pillsbury, S. J.; Shanahan, E.; Williams, V. A. Tetrahedron 2005, 61, 3565.
4
Domka, Z. P. ; Sharpless, K. B. Organic Letters 2002, 4(15), 2525.
5
Reid, M. C. ; Clark, J. H. ; Macquarrie D. J. Green Chem., 2006, 8, 437.
Among the many serotonin-like compounds studied, the discovery of the anti-migraine drug
sumatriptan 1 stimulated the development of other 5-HT1D receptor agonists.1 From the
chemical point of view, several of them (sumatriptan 1,2, avitriptan 2,3 and almotriptan 34) have
the common feature of possessing a sulfamoylmethyl group attached, through a methylene
bridge, to the position 5 of indole nucleus (Figure 1).
H3C
N CH3 H3C
N
N N CH3
H3C O
CH3NH
S CH3NH N
S S
O O
N O O O O
H N N
H H
Sumatriptan 1 Almotriptan 3
Avitriptan 2
Figure 1
In the present work, the compound 9 has been synthesized, being the main impurity detected in
Sumatriptan5. The method employed to obtain this compound is indicated in the scheme 1.
O
O O H2, 2atm
N,N-dimetilformamide-
CH3 dimethylacetal N 10%Pd(C) MeO
MeO MeO
Toluene N
NO2 Pirrolidine H
NO2
DMF, reflux. 97% 6
4 76% 5
Oxalyl chloride
Me2NH/H2O
77%
Me Me
N Me N Me
O
O
LiAlH4 O
HO MeO
100%
N N 7
H H
8
Me
N Me
1. HCl
Me
N Me
Me
N Me 2. HO
MeHN
N 8 S
H O Me
MeHN O
S N N Me
O O 3. NaOH 10N, pH=12-13 H
N
H
20% 9 N
Sumatriptan 1
H
Scheme 1
1
Hopkins S.J., Drugs Today, 28, 155 (1992)
2
Relevant patents: Dowle, M. D. and Coates, I. H., GB 2 124 210 (1983). Dowle, M. D. and Coates US Patent 4,
816 470 (1989). Oxford, A. W., GB 2 162 522 (1984)
3
Brodfuehrer, P.R., Chen, B.C., Sattelberg, T.R., Smith, Sr.P.R., Reddy, J.P., Stara, D.R., Quinlan, S.L., Reid, J.G.,
Thottathil, J.K. and Wang, S.J., J. Org. Chem., 62, 9192 (1997)
4
Fernandez, M.D., Puig, C., Crespo, M.I. and Moragues, J., ES Patent 2, 084, 560, (1994)
5
Skwierawska, A.and Paluszkiewicz, E., Polish J. Chem., 2003, 77, 329-332.
109
a
Departamento de Química Orgánica I. Facultad de Ciencias. Químicas. Universidad
Complutense, 28040-Madrid. bInstituto de Química Orgánica General, CSIC, Juan de la
Cierva 3, 28006-Madrid.
NHR3
OR1
R2O OR1 H H H H
R2O R1O O
CN
N N N
O PMP O PMP
R4
Scheme 1
1
See, for example: (a) Afarinkia, K.; Bahar, A. Tetrahedron:Asymmetry 2005, 16, 1239. (b) Butters, T.
D.; Dwek, R. A.; Platt, F. M. Chem. Rev. 2000, 100, 4683.
2
See, for example: (a) Alcaide, B.; Almendros; Aragoncillo, C. Chem. Rev. 2007, in press. (b)
Alcaide, B.; Almendros, P. Curr. Med. Chem. 2004, 11, 1921. (c) Alcaide, B.; Almendros, P. Synlett
2002, 381. (d) Palomo, C.; Aizpurua, J. M.; Ganboa, I.; Oiarbide, M. Synlett 2001, 1813.
110
N COOH
N COOH
H H2N COOH
H
proline E-phenylproline phenylalanine
Figure 1
Although some methods reported so far can provide optically pure final
products, our main objective is the development of a versatile approach to the
synthesis of all four possible stereoisomers of E-phenylproline, i.e. two cis
enantiomers and two trans enantiomers, in a multigram scale and enantiomerically
pure form. To this end, we have applied a strategy to the synthesis of rac-cis and
rac-trans compounds that, followed by a highly efficient resolution method using
chiral HPLC, has allowed us the isolation of all four stereoisomers of E-
phenylproline in optically pure form.
111
NH NH
HN
N COOH N COOH
H H HOOC HOOC HOOC
1
(a) Avenoza, A.; Cativiela, C.; Busto, J. H.; Fernández-Recio, M. A.; Peregrina, J. M.; Rodríguez, F.
Tetrahedron 2001, 57, 545-548; (b) Casabona, D.; Cativiela, C. Tetrahedron 2006, 62, 10000-10004;
(c) Casabona, D.; Jiménez, A. I.; Cativiela, C. Tetrahedron 2007, 63, 5056-5061.
112
a
Departamento de Química Orgánica, ICMA, Universidad de Zaragoza-CSIC, 50009
Zaragoza. bInstitut de Recerca Biomèdica, Parc Científic de Barcelona, Josep Samitier 1-5,
08028 Barcelona. cUnitat de Química Combinatòria, Parc Científic de Barcelona, Josep
Samitier 1-5, 08028 Barcelona.
R1 N N R2
O O O
P1 P2 P3
Figura 1. Estructura general de los inhibidores de POP
1
Polgar, L. Cell Mol. Life Sci. 2002, 59, 349-362.
2
Polgar, L. Curr. Med. Chem. 2002, 2, 251-257.
3
Komatsu, Y. J. Neurosci. 1996, 16, 6342-6352.
4
Wilk, S.; Orlowski, M. J. Neurochem. 1983, 41, 69-75.
113
a
Departamento de Química Orgánica, Facultade de Farmacia, Universidade de Santiago
de Compostela, Campus Sur s/n. 15782 – Santiago de Compostela (España). bCIQ,
Departamento de Química, Faculdade de Ciencias, Universidade do Porto, 4169-007 –
Porto (Portugal).
estrategias usadas para corregir los síntomas de los pacientes que sufren dichas
patologías, por lo que en los últimos años se han desarrollado nuevos inhibidores
de MAO y acetilcolinesterasa como agentes neuroprotectores. 1
La Rasagilina [N-propargil-1(R)-aminoindano, I] 2 es un nuevo fármaco de uso
en la EP tanto en monoterapia como en combinación con levodopa. Es un potente,
selectivo e irreversible inhibidor de MAOB, que previene la degradación de
B
HN n( ) NH
II
Rasagilina n = 0, 1
I
R = OR, NR'R'', CO2R', CH2OR',
CONR'R'', alquilo, arilo o heteroarilo
1
Shih, J. C. et al. Annu. Rev. Neurosci. 1999, 22, 197.
2
Youdim, M. B. et al. J. Med. Chem. 2002, 45, 5260.
114
1
Mishra, R.K., Chiu, S., Chiu, P., Mishra, C.P. Methods Find. Exp. Clin. Pharmacol. 1983, 5, 203.
2
Yamamoto, H., Murphy, L.J. J. Endocrinol. 1995, 146, 141.
3
Rodríguez-Borges, J. E., García-Mera, X., Fernández, F., Lopes, V. H.C., Magalhães, A. L. and
Cordeiro, M. N. D. S. Tetrahedron 2005, 61 (46), 10951.
115
a
Research Unit on BioActive Molecules (RUBAM); Departamento de Química Orgánica
Biológica, Instituto de Investigaciones Químicas y Ambientales de Barcelona (IIQAB-
CSIC), Jordi Girona 18-26, 08034 Barcelona, España. bDepartment of Genetics and
Developmental Biology, University of Connecticut Health Center (UCHC), 263 Farmington
Avenue, Farmington CT 06030, USA,
O
R R2
Estudios sobre el efecto de esta familia sobre P. falciparum
N mostraron que, efectivamente, varios de los compuestos probados
NH O 3 inhibían el crecimiento in vitro del parásito con valores de IC50
R1 R iguales o menores de 1 µM.
1
Hanada, K..; Palacpac, N. M.; Magistrado, P.A.; Kurokawa, K.; Rai, G.; Sakata, D.; Hara, T.; Horii,
T.; Nishijima, M.; Mitamura, T.; J. Exp. Med. 2002, 195, 23-34.
2
Pessi, G.; Kociubinski, G.; Mamoun, C.B.; Proc. Natl. Aca. Sci. USA 2004, 101, 6260-6211.
116
a
Research Unit on Bioactive Molecules (RUBAM); Departamento de Química Orgánica
Biològica, (IIQAB C.S.I.C); Jordi Girona 18-26, 08034 Barcelona
b
Universidad de Barcelona, Facultad de Farmacia, Unidad de Química Farmacéutica
(Unidad Asociada al CSIC), Avda. Juan XXIII, s/n, 08028 Barcelona, España
1
Demedts IK, et al., Respir Res. , 2006,7, 53.
2
Petrache I, et al., Nat Med., 2005, 11, 471.
3
Bielawska A, et al., J Biol Chem., 1993, 268, 26226.
4
Ogretmen B, et al., J Biol Chem., 2001, 276, 32506.
5
Jiang Q, et al., Proc. Natl Acad. Sci USA, 2004, 101, 17825.
6
Stiban J, et al. Apoptosis, 2006, 11, 773.
117
Pipecolic acid derivatives are widely used in therapeutic chemistry because of their
significant and various biological activities. In particular, 4-substituted derivatives
can act as anaesthetics, antitumorals, antibiotics, anticoagulants, NMDA receptor
agonists or antagonists, immunosuppressants and enzyme inhibitors. The design
and study of novel drugs with central nervous system activity containing 4-
substituted pipecolic acids requires the development of new methodologies to
synthesize them stereoselectively.1
In this regard, taking advantage of our previous methodology to build the piperidine
ring2 and with the aim of design new potential chemotherapeutic agents, we have
optimized the asymmetric synthesis of seven important 4-substituted D-pipecolic
acids starting from the same chiral 4-piperidone. Two selective competitive
antagonists of the NMDA receptor, diastereomeric cis- and trans-4-
phosphonomethyl-D-pipecolic acids, cis-1 (CGS-20281) and trans-1,3 have been
prepared in enantiomerically pure form. (2R,4S)- and (2R,4R)-4-hydroxypipecolic
acids4 cis-2 and trans-2, two versatile chiral precursors in the preparation of
biologically active molecules, have been diastereodivergently obtained.
In addition, we have developed three synthetic routes directed to the obtention of
conformationally constrained Į-amino acid chimeras glutamic acid/pipecolic acid
trans-3, lysine/pipecolic acid trans-4 and ornithine/pipecolic acid trans-5, all of them
in ortogonally protected form. Trans-3 is a D-glutamic acid analogue with NMDA
receptor agonist activity5 and trans-4 and trans-5 are restricted analogues of D-
lysine and D-ornithine respectively, and show interesting applications in
peptidomimetics design.6
CH2NHMoc
PO3H2 CO2CH3 NHAc
OH
* *
N CO2H N CO2H N CO2H
N CO2H N CO2H
H H Boc Boc Boc
cis-1/trans-1 cis-2/trans-2 trans-3 trans-4 trans-5
1
Kadouri-Puchot, C.; Comesse, S. Amino Acids 2005, 29, 101-130.
2
Badorrey, R.; Cativiela, C.; Díaz-de-Villegas, M. D.; Gálvez, J. A. Tetrahedron 1999, 55, 7601-7612.
3
Etayo, P.; Badorrey, R.; Díaz-de-Villegas, M. D.; Gálvez, J. A. Synlett 2006, 2799-2803.
4
Celestini, P.; Danieli, B.; Lesma, G.; Sacchetti, A.; Silvani, A.; Passarella, D.; Virdis, A. Org. Lett.
2002, 4, 1367-1370 and references therein.
5
Madsen, U.; Brehm, L.; Schaumburg, K.; Jørgensen, F. S.; Krogsgaard-Larsen, P. J. Med. Chem.
1990, 33, 374-380.
6
Goswami, R.; Moloney, M. G. Chem. Commun. 1999, 2333-2334.
118
* *
R 2* N
N H N H
N R2* N R 2*
R 2* R 2* H
R 1* R 1*
cis 1/trans 1 cis 2/trans 2 3 endo-4 exo-4
1
Badorrey, R.; Cativiela, C.; Díaz-de-Villegas, M. D.; Gálvez, J. A. Tetrahedron 1999, 55, 7601-7612.
2
Singh, S. Chem. Rev. 2000, 100, 925-1024 and references therein.
3
Iceta, R.; Mesonero, J. E.; Aramayona, J. J.; Alcalde, A. I. J. Physiol. Pharmacol. 2006, 57, 119-130.
4
Veenstra, S. J.; Hauser, K.; Scilling, W.; Betschart, C.; Ofner, S. Bioorg. Med. Chem. Lett. 1996, 6,
3029-3034.
5
Choi, K. I.; Cha, J. H.; Cho, Y. S.; Pae, A. N.; Jin, C.; Yook, J.; Cheon, H. G.; Jeong, D.; Kong, J. Y.;
Koh, H. Y. Bioorg. Med. Chem. Lett. 1999, 9, 2795-2800.
119
HO
La modificación estructural de una serie de
HO HO OH compuestos extraídos de una esponja marina,
O OH
C14H29 HO
O
Agelas mauritianus ha conducido al KRN 7000
HO O HO
OH O NH OH
OH
que ha revelado una fuerte actividad
KRN 7000 C25H51 OH en configuración D antitumoral1.
OH OH
HO X HO X
OH OH
C14H29 C14H29
HO N HO N
H H
OH NH OH OH NH OH
O O
X = H, OH R X = H, OH R
R=C7H15, C25H51 R=C7H15, C25H51
N-axial N-ecuatorial
1
T.Natori, Y.Koezuka, H. Tatsuo Tetrahedron Lett., 1993, 34, 5591.
2
T, Kawano, J. Cui, Y. Koezuka, I. Toura, Y. Kaneko, K. Motoki, H. Ueno, R. Nakagawa, H. Sato, E.
Kondo, H. Koseki, M. Taniguchi, Science, 1997, 278, 1626.
120
a
Research Unit on BioActive Molecules, Departamento de Química Orgánica Biológica,
IIQAB-CSIC, Jordi Girona 18-26, 08034 Barcelona, España. bUniversitat de Barcelona,
Facultat de Farmàcia, Unitat de Química Farmacèutica (Unitat Associada al CSIC), Avda.
Joan XXIII s/n; 08029 Barcelona, España.
OH OH OH OH OBn
1
Desnick, R. J. J. Inherit. Metab. Dis. 2004, 27(3), 385-410.
2
Serrano, P.; Llebaria, A.; Vázquez, J.; de Pablo, J.; Anglada, J. M.; Delgado, A. Chem. Eur. J. 2005,
11(15), 4465-4472.
121
OCH3 O
H
H3C R
N
Estos datos nos impulsaron a H3CO
N
OCH3 X
ampliar los estudios SAR a través OCH3 OCH3
O
O
H3C
H
R
H N
de procesos de reducción quimio y H3C
N
R Bn
N
H3CO
diastereoselectiva en derivados de H3CO
N
OCH3 Y
R'
pirazino[1,2-b]isoquinolina-1,4- OCH3 O
R = CO2iPr, Boc, CH3
diona según el esquema. R = CO2iPr, Boc, CH3
R' = OBn, OH, O-acyl, N-acyl
X = H, OH; H2; Y = O
X = H2; Y = H, CN
1
Martínez, E. J.; Corey, E. J.; Owa, T. Chem. Biol. 2001, 8, 1151-1160.
2
a) González, J. F.; de la Cuesta, E., Avendaño, C. Tetrahedron Lett. 2003, 44, 4395-4398. (b) González, J.
F.; de la Cuesta, E., Avendaño, C. Tetrahedron 2004, 60, 6319-6326. (c) González, J. F.; Salazar, L.; de la
Cuesta, E., Avendaño, C. Tetrahedron 2005, 61, 7447-7455.
3
González, J. F.; de la Cuesta, E.; Avendaño, C. Bioorg. Med. Chem. 2007, 15, 112-118.
122
a
Departamento de Química Orgánica, Facultade de Farmacia, U.S.C, Campus Sur, E-15782,
b
Santiago de Compostela. Departamento de Química Fundamental, Facultade de Química,
U.D.C., Campus da Zapateira,15071, A Coruña. cRega Institute for Medical Research,
Katholieke Universiteit Leuven, Minderbroedersstraat, B-3000 Leuven, Belgium
1
a) A. A. Krayevsky and K. A. Watanabe, “Modified Nucleosides as Anti-AIDS Drugs: Current Status and
Perspectives”; Bioinform, Moscow, 1993. b) S. Thomas, J. E. McDowall, V. Cheah, A. Bye and M. B.
Segal, “The Entry of Abacavir into the Guinea-pig Brain: Comparison with Other Reverse Transcriptase
Inhibitors”. Presented at the 12th World AIDS Conference, Geneva, 1998.
2
Balzarini, J.; Lee, C.-K.; Herdewijn, P.; De Clercq, E. J. Biol. Chem., 1991, 266, 21509.
123
The synthesis of modified amino acids and peptides has allowed to obtain new
drugs, or to increase the bioactivity, biodisponibility or hydrolytic stability of existing
ones.1a For instante, the E-amino acid Ritalin is clinically used to treat hyperactivity
in children,1b and the dipeptide bestatin is a potent inhibitor of aminopeptidase B.1c
NH2 O
Ph Ph
N N CO2H
H H
HO2C OH
Ritalin Bestatin
One-pot
O R' decarboxylation O R'
BzHN nucleophile addition BzHN
N COOH N Nu
H H
R catalyst R
1. (a) Sewald, N.; Jakubke, H.-D. Peptides: Chemistry and Biology, Wiley-VCH, Weinheim 2002. (b)
Matsumura, Y.; Kanda, Y.; Shirai, K.; Onomura, O.; Maki, T. Tetrahedron 2000, 56, 74117422. (c)
Pearson, W. H.; Hines, J. V. J. Org. Chem. 1989, 54, 42354237.
124
INTRODUCTION
S-Nitrosothiols were considered chemicaly unstable products until the room
temperature stable SNAP was obtained and reported in 19781. The unstability of
nitrosothiols in solution adds serious difficulties to its development as therapeutic
drugs. Several factors affect its decomposition in solution including the exposure to
light, temperature, pH conditions and the presence of transition metal ions.2
Nitric oxide released from nitrosothiols, is an ubiquitous signaling molecule in
mammalian biology that is involved in several physiological pathways related with
cGMP cascade, endotelium relaxation and it is a known inhibitor of platelet
aggregation process 3,4.
OBJECTIVE
Our objective was to obtain new S-nitrosothiols as potential anti-aggregating
agents with higher stability than the reference compounds SNAP and GSNO.
RESULTS
Starting from the structures of SNAP and GSNO several new compounds have
been synthesised. In general, tertiary nitrosothiols are more stable than secondary,
primary or aryl nitrosothiols1.
SNO
SNO
O
O O O
OH H
N
N
H HO N OH
H
O
NH2 O
SNAP GSNO
SNO
O
R2
R1 N
H
1a-n O
Platelet aggregation was measured in vitro in human platelet rich plasma (PRP)
using a luminometric method with ADP as aggregant agent. Effect on the
expression of IIb-IIIa platelet receptors was determined by flow cytometry using
PAC-1 and in both cases the IC50 (nM) was calculated and compared with values
obtained with the reference compounds.
CONCLUSION
Compound 1b (R1=CH3, R2=OH) with a better pharmacological profile than
reference compounds (IC50 0,31 and 0,19 nM respectively) was selected as a lead
compound for optimization.
125
a
Instituto de Química Médica (CSIC), Juan de la Cierva 3, 28006-Madrid
b
Instituto de Fermentaciones Industriales (CSIC), Juan de la Cierva 3, 28006-Madrid
Oxidative stress has been associated with the development of many chronic
and degenerative illnesses, including cancer, cardiovascular failures, and neuronal
degeneration such as Alzheimer’s disease.1 In fact, epidemiological studies have
established an inverse correlation between the occurrence of these diseases and
the intake of high amounts of fruits and vegetables that are rich in antioxidants.
In the last years, several in vitro methods have been developed for assaying
the antioxidant activity of natural and synthetic products. One of the most widely
used is the fluorometric oxygen radical absorbance capacity assay (ORAC-FL)2
that measures the capacity of the tested sample to compete against a target
molecule in the capture of free oxygen radicals. The ORAC-FL assay involves the
use of a microplate fluorescence reader, more expensive than a microplate optical
reader. For this reason, the development of methodologies that could use UV-vis
spectroscopy instead of fluorometry could be of interest in small laboratories from
public research institutions and from pharmaceutical and food industries.
Results are usually expressed with reference to a well-known antioxidant
(trolox) as ORAC units. Although the ORAC scale pursuits to correct differences
among laboratories results, a literature research reveals serious discrepancies in
the ORAC values for a given pure product. For example, for the well-known
antioxidant quercetin the following ORAC-FL values could be found in the recent
scientific literature: 10.5,3 7.28,4 and 6.50.5
In this work we adapted the ORAC assay to a conventional microplate visible
reader and proposed a general experimental protocol that provided accurate
ORAC-UV values.
1
Valko, M.; Leibfritz, D.; Moncol, J.; Cronin, M. T. D.; Mazur, M.; Telser, J. Int. J. Biochem. Cell Biol.
2007, 39, 44-48.
2
Huang, D.; Ou, B.; Hampsch-Woodill, M.; Flanagan, J.; Prior, R. L. J. Agric. Food Chem. 2002, 50,
4437-4444.
3
Dávalos, A.; Gómez-Cordovés, C.; Bartolomé, B. J. Agric. Food Chem. 2004, 52, 48-54.
4
Ou, B.; Hampsch-Woodill, M.; Prior, R. L. J. Agric. Food Chem. 2001, 49, 4619-4626.
5
Ordoudi, S. A.; Tsimidou, M. Z. J. Agric. Food Chem. 2006, 54, 9347-9356.
126
OH
HO
O O
Fig.1 Fig.2
OH
R1
R
O O
Fig.3
1
Orallo, F. Biological Effects of Cis- Versus Trans-Resveratrol, in: B. B. Aggarwal, S. Shishodia (Eds.),
Resveratrol in Health and Disease, CRC Press, Boca Raton, USA 2005, pp.577-600
2
Borges, F.; Roleira, F.; Milhazes, N.; Santana, L.; Uriarte, E. Current Medicinal Chemistry, 2005, 12, 887-
916
127
Glycomimetics are used for rational drug design. Obviously, it is necessary to take
into account the shape of the receptor binding site and the conformation and
dynamic nature of the natural ligand. In this work we have investigated, by using
NMR and molecular dynamics, the conformational behaviours of three new
glycomimetics and their consequences for binding to receptors.
The conformational analysis of ligand 1 showed an equilibrium between two
conformers where the lactam moiety shows partial mobility; the galactose instead
is present in the chair conformation. Different NMR experiments were used to
evaluate the affinity of this ligand for the protein Viscumin (VAA), a galactose-
binding: saturation transfer difference (STD) underlined finally the specific binding
using the galactose moiety in an exclusive manner.
Morphine-6-O-ȕ-D-glucuronide (M6G) 2 is a natural metabolite of morphine in
humans and animals; its properties1 and its capacity to penetrate the blood-brain
barrier have been extensively studied2; morphine-6-O-Į-D-mannose (M6M) 3 was
synthesised in the attempt to reproduce the analgesic effect of M6G and to avoid
the collateral effects of the opioids. The conformational study has been also
performed by using NMR and molecular dynamics, underlining a significant
difference between the two ligands: M6G is present as an equilibrium between a
folded and an extended conformation, whereas M6M shows only the extended
one. DOSY and NOESY experiments conducted in presence of an excess of the
surfactant sodium dodecyl sulfate (SDS) permitted to detect the conformational
behaviour in a membrane-like environment for 3 and 2.
OH
OH
HO N N
SO
H O H
OH OH
O O O
O O O OH
HO O OH HO
HN
HO OH HO OH
1 2 3
OH
O
1
Carrupt P.A.; Testa B.; Bechalany A.; El Tayar N.L; Descas P.; Perrissoud D. J. Med. Chem. 1991,
34, 1272.
2
Schwarzinger S.; Hartmann M.; Kremminger P.; Müller N. Bior. Med. Chem Lett. 2001, 11, 1455.
128
O
R 5=
1
Porcal, W. Investigación y desarrollo de nitronas como agentes moduladores del estrés oxidativo.
Tesis Doctoral. 2007. Facultad de Química, Universidad de la República.
2
Boiani, M.; Cerecetto, H.; Gerpe, A.; González, M.; Hernández, P.; Porcal, W.; Cazzulo, J.J.
rd
Screening of Lybrary-Compounds as Cruzain Inhibitors. 3 Brazilian Symposium on Medicinal
Chemistry, San Pedro, San Pablo, Brasil, Noviembre 2006.
129
procedimiento "one-pot"
i) EtOH / HCl / reflux
ii) NaOH / NaOCl
0ºC-r.t.
60%
Ph
1, E
2, Z N O
O
Figura 1 25% rendimiento global N
R2
N
1deox - 6deox O
1 - 3% (por HPLC)
Figura 2 N
1
Porcal, W.; Hernández, P.; Aguirre, G.; Boiani, L.; Boiani, M.; Merlino, A.; Ferreira, A.; Di Maio, R.;
Castro, A.; González, M.; Cerecetto, H. Bioorg. Med. Chem. 2007, 15, 2768-2781.
130
a
Laboratorio de Química Orgánica, Departamento de Química Orgánica, Facultad de
Ciencias-Facultad de Química, Universidad de la República. Iguá 4225, C.P. 11400,
Montevideo, Uruguay.
R2 NO2 NH2
R2 NHOH R2
R1 OH
R2
O O
N N
O O
Compuestos líderes N N
R1
R2
OH
N
OH
N
1
Aguirre, G.; Boiani, L.; Cerecetto, H.; Di Maio, R.; González, M.; Porcal, W.; Thomson, L.; Tórtora,
V.; Denicola, A.; Möller, M. Bioorg. Med. Chem. 2005, 13, 6324.
2
Grosa, G.; Galli, U.; Rolando, B.; Fruttero, R.; Gervasio, G.; Gasco, A. Xenobiotica, 2004, 34, 345,
131
a
Departamento de Química Orgánica, Facultad de Ciencias, Universidad de la República.
Iguá 4225, 11400 Montevideo, Uruguay. bCentro de Investigaciones en Farmacología
Aplicada (CIFA), Universidad de Navarra. Irunlarrea s/n, 31080, Pamplona, España.
1
Chen, C.; Kong, A.N.T. Free Radic. Biol. Med. 2004, 36: 1505. Hotzclaw, W.D.; Dinkova-Kostova,
A.T.; Talalay, P. Adv. Enz. Regul. 2004, 44: 335. Song, L.L.; Kosmeder J. W.; Lee, S.; Gerhäuser, C.;
Lantvit, D.; Moon, R.C.; Moriarty, R. M.; Pezzuto, J.M. Cancer Res. 1999, 59: 573.
2 rd
Presentado, en parte, en el 3 Brazilian Symposium on Medicinal Chemistry. 2006, S2-069
132
a
Neuropharma, S.A., Avda. de La Industria 52, 28760 Tres Cantos (Madrid), Spain
Glycogen synthase kinase 3 (GSK-3) is a serine-threonine protein kinase
which plays an important regulatory role in several signaling pathways of cellular
processes. Disorders in many of these regulatory pathways are involved in several
human diseases, such as Parkinson’s Disease, Alzheimer’s Disease (AD), type II
diabetes, bipolar disorders, schizophrenia, chronic inflammatory disorders or prion-
induced neurodegeneration. Therefore, the discovery of new GSK3 inhibitors could
provide new therapeutic approaches to treat these diseases1.
Focusing on the study of AD, the presence of neurofibrillary tangles in
neurons of cerebral cortex is one of the abnormalities observed in the brain of AD
patients, and hyperphosporylated tau protein seems to be a main component of
these neuronal deposits. GSK3-ȕ is thought to be one of the most relevant
enzymes involved in tau phosphorylation, hence its inhibition could play an
important role in the treatment of this disease.
The ocean is considered to be a great source of potential drugs and it is
interesting to point out the prominent role that marine invertebrates have played in
the generation of novel GSK3-ȕ inhibitors including hymenialdisine, meridianines
and indirubines isolated from sponges, ascidians and molluscs2.
As part of our research, based in the isolation of new lead compounds from
marine origin with potential for the treatment of AD, we have found that the
isopropanolic extract from the mediterranean bryozoan Myriapora truncata showed
inhibitory activity against GSK3-ȕ. Fractionation and purification of active
components from this extract, guided by in vitro enzyme inhibition assays, resulted
in the isolation and identification of Clausenlactama, previously only isolated from
terrestrial sources, the tree Clausena excavata3.
In order to confirm the structure of the isolated compound and fully explore
the potential of this kind of structures as novel inhibitors of the enzyme, a program
to produce synthetic analogues have been carried out and all the compounds have
been tested in our biological assays.
1
Maijer L et al. Trends Pharmacol. Sci. 2004;25, 471-480.
2
Martínez, A.; Castro, A.; Medina, M. Glycogen Synthase kinase 3 (GSK-3) and its inhibitors. Ed.
Wiley-Interscience 2006.
3
Shang et al. Acta Botanica Yunnanica, 1993, 15 (3), 299-302.
133
a
Instituto de Química Médica (CSIC), Juan de la Cierva 3, 28006 Madrid.
b
Unidad de Regulación Génica, CNM, Instituto de Salud Carlos III, Madrid.
c
Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer
Center, The University of North Carolina, Chapel Hill, NC 27599-7260, USA.
1
Antoni, F. A. Front. Neuroendocrinol. 2000, 21, 103-132.
2
Giembycz, M. A.; Corrigan, C. J.; Seybold, J.; Newton, R.; Barnes, P. J. J. Pharmacol. 1996, 118,
1945-1958.
3
a) Li, L.; Yee, C.; Beavo, J. A. Science, 1999, 283, 848-851. b) Nakata A.; Ogawa, K.; Sasaki, T.;
Koyama, N.; Wada, K.; Kotera, J.; Kikkawa, H.; Omori, K.; Kaminuma, O. Clin. Exp. Immunol. 2002,
128, 460-466.
134
A supervised artificial neural network model has been developed for the prediction
of inhibition of Nitric Oxide Synthase. A diverse set of chemicals was chosen in this
study and the definition of the molecules was achieved from a not supervised neural
network using a home made program named CODES®. This program codifies each
structure from a topological point of view, in a set of numerical parameters with the only
knowledge of its SMILES code and consequently of its chemical structure. Using this
methodology, two models have been obtained for the prediction of inhibition of nNOS
and iNOS.
135
a
Dept. Química Bioquímica y Biología Molecular. Universidad CEU Cardenal Herrera,
Moncada, Valencia (Spain), bDiv. Farmacia y Tecnología Farmacéutica. Universidad Miguel
Hernández. Alicante (Spain)
0,25
seleccionar aquellas estructuras que
Test Inactivos
0,15
El modelo topológico diseñado, se ha
0,10 aplicado a un grupo test, formado por 100
0,05 antibacterianos y 100 no antibacterianos,
0,00
con el fin de validar la calidad del modelo,
-10 0
F.D.
10
obteniéndose elevados porcentajes de
20
1
Coll R; Gargallo-Viola D; Tudela E; Xicota MA; Llovera S; Guinea J. Antimicrob Agents Chemother.
1996, 40(1), 274.
2
Jason A. Wiles; Qiuping Wang; Edlaine Lucien;, Akihiro Hashimoto; Yongsheng Song; Jijun Cheng;
Christopher W. Marlor; Yangsi Ou; Steven D. Podos; Jane A. Thanassi. Bioorganic & medicinal
chemistry letters. 2006, 1272
136
a
Departamento de Química, Bioquímica y Biología Molecular, Universidad CEU Cardenal
Herrera. 46113 Moncada, Valencia (Spain). bDept. Quimica–Física, Facultad de Farmacia,
Universitat de Valencia, 46100 Burjassot, Valencia, Spain. cDpto. Fisiología, Farmacología
y Toxicología. Universidad CEU Cardenal Herrera, 46113 Moncada, Valencia (Spain).
Recently, the opportunistic fungal infections have risen dramatically. One of the
biggest problems facing nowadays the antifungal therapy is the arising of drugs
resistance strains for most of the drugs currently used in clinic practice. Therefore,
it is important to find new antifungal candidate compounds, particularly new leads,
able to become the basis for developing new drugs.1
Microsporum gypseum
LogMICM.gypseum = 0.017 + 0.070 ST(-OH) – 16.916 G v5 + 0.267 G v2 + 280.42 J v5 – 6.367 J2
+ 4.932 9Ȥp
N=33; SEE=0,289; SEE(vc)=0,375; r2=0.75;
The obtained results demonstrate that the molecular topology is a very useful tool
in the prediction of microbiological properties.
Acknowledgement: Financial support of this work was by the Universidad CEU Cardenal
Herrera (PRUCHB06/11 and PRUCH06/39).
1
Georgopapadakou, N.H. Curr. Opin. Microbiol. 1998, 1, 547-557
2
Furnival, G.M. Tecnometrics, 1971, 14, 403. Hocking, R.R. Tecnometrics, 1972, 14, 967.
137
p38 MAP Kinase is currently one of the most attractive targets for pharmaceutical
industry1 as well as for the medicinal chemistry community2. A unique combination of
well established pharmacology, clinical efficacy and the opportunity to employ
structure-based drug design has converted it a highly attractive target for therapeutic
intervention. It is well-known that the p38 MAP Kinase signaling pathway plays an
important role in inflammation and other physiological processes.
We have previously reported the design and discovery of a 2-aminobenzimidazole
based series as potent p38 MAPK inhibitors3. Our initial lead compound 1, had low
nanomolar activity in both ATP competitive enzyme binding assay and in the
inhibition of TNFa release in macrophages. We developed an extensive SAR around
this lead molecule and identified new benzimidazole derivatives. As a result, we
found that the sulfonyl group in the N-3 imine nitrogen, plays a key role for the
activity. This labile moiety could be replaced by an alkyl group and led to new
inhibitors which showed good activity both in vitro and in vivo (2).
The synthesis and biological activities of these compounds will be described.
N N
F NH2 F NH2
N N N N
S O
N O N
F H F
1 2
1
Chakravarty, S. Dugar, S. Ann. Rep. Med Chem 2002, 37,177
2
Ferracioli, G. F. Curr. Opin. Anti-inflam. Inmunomod. Invest. Drugs 2000, 2, 74
3
De Dios, A.; Shih, C.; Lopez de Uralde, B.; Sanchez, C.; del Prado, M.; Cabrejas, L.M.M.; Pleite, S.;
Blanco-Urgoiti, J.; Lorite, M.J.; Nevill, C.R., Jr.; Bonjouklian, R.; York, J.; Vieth, M.; Wang, Y.; Magnus,
N.; Campbell, R.M.; Anderson, B. D.; McCann, D.J.; Giera, D.D.; Lee, P.A.; Schultz, R.M.; Li, L.C.;
Johnson, L.M.; Wolos, J.A. J. Med. Chem 2005, 48, 2270.
138
a
Departamento de Química, Bioquímica y Biología Molecular, Universidad CEU Cardenal
Herrera. 46113 Moncada, Valencia (Spain). bDpto. Fisiología, Farmacología y Toxicología.
Universidad CEU Cardenal Herrera, 46113 Moncada, Valencia (Spain). cDpto Ingeniería,
Div. Farmacia y Tecnología Farmacéutica. Universidad Miguel Hernández. Alicante (Spain)
In this work a multilinear regression analysis has been carried out in order to
look for a connectivity functions capable to accurately predict biological propertires
of a group of quinolones.1 The studied properties were: the minimum inhibitory
concentration 50 (MIC50) and 90 (MIC90) versus two microorganisms Escherichia
Coli and Streptococcus pyogenes, 2 widely used nowadays because of their broad
spectrum of activity, well tolerance profile and advantaged pharmacokinetic
properties. The structural description has been achieved through topological
indices. The results obtained clearly reveal the high efficiency of molecular
topology for the prediction of these properties. Randomization and cross-validation
by use of leave-one-out test were also performed in order to assess the stability
and the prediction ability of the connectivity functions selected.
Streptococcus pyogenes
5 v N=13; SEE=0,431; SEE(vc)=0,578;
MIC50: 213,48 + 2,46 ǒ P + 1,06 2N - 24,44 ST (-OH) 2 2
r =0.951; r (cv)=0,913
7 N=12; SEE=0,698; SEE(vc)=1,102;
MIC90: - 5,34 + 7,39 ǒvP + 12,86 '1ǒ – 3.07 V4 2 2
r =0.939; r (cv)=0,848
Escherichia coli
MIC50= 0,571 - 0,035 ST(-CH3) – 2,42 J4 – 0.021 V4 N=17; SEE=0,027; SEE(vc)=0,031;
2 2
r =0,854; r (cv)=0,764
v N=14; SEE=0,073; SEE(vc)=0,093;
MIC90= 0,228 - 0,091 ST(-Cl) – 0.144 G 4 + 6,29 J5 2 2
r =0,925; r (cv)=0,876
The obtained results demonstrate that the molecular topology is a very useful tool
in the prediction of microbiological properties.
Acknowledgement: Financial support of this work was by the Universidad CEU Cardenal
Herrera (PRUCHB06/11 and PRUCH06/39).
1
Mut-Ronda, S. et al., Bioorg. Med. Chem. Let., 2003,13, 2699.
2
Calabuig, C. et al., Int. J. Pharm., 2004, 278,111.
139
a
Dept. Química Bioquímica y Biología Molecular. Universidad CEU Cardenal Herrera,
Moncada, Valencia (Spain). bDpto Ingeniería, Div. Farmacia y Tecnología Farmacéutica.
Universidad Miguel Hernández. Alicante (Spain). cDpto. Fisiología, Farmacología y
Toxicología. Universidad CEU Cardenal Herrera, Moncada, Valencia (Spain).
The obtained results demonstrate that the molecular topology is a very useful tool
in the prediction of pharmacokinetics properties.
Acknowledgement: Financial support of this work was by the Universidad CEU Cardenal
Herrera (PRUCHB06/11 and PRUCH06/39).
1
Mut-Ronda, S. et al., Bioorg. Med. Chem. Let., 2003,13, 2699.
2
Calabuig, C. et al., Int. J. Pharm., 2004, 278,111.
140
a
Departamento Química Aplicada, Universidad Pública de Navarra, Campus Arrosadía,
31006 Pamplona, bDepartamento Ciencias de la Salud, Universidad Pública de Navarra,
H R
N
O S n
O O
BTC
1
Alonso MM, Encío I, Martínez-Merino V, Gil MJ, Migliaccio M. Oncogen,2003, 22:3759.
2
Villar R, Encío I, Migliaccio M, Gil MJ, Martínez-Merino V. Biorg.Med.Chem., 2004,12:963.
3
Presentado, en parte, en el 1st European Chemistry Congress, 2006, Budapest.
141
Ar = Variable
S
1
Orus L., Pérez-Silanes S., Oficialdegui A., Martinez J., Del Castillo J.C., Mourelle M. (2002).
Synthesis and molecular modeling of new 1-aryl-3-[4-arylpiperazin-1-yl]-1-propane derivatives with
high affinity at the serotonin transporter and at 5-HT1A receptors. J. Med. Chem. 45 (19): 4128-4139.
2
Jensen J. B., Trager W. and Doherty J. (1979). Plasmodium-Falciparum - Continuous Cultivation in
a Semi-Automated Apparatus. Exp. Parasitol. 48:36-41.
3
Deharo E., García R. N., Oporto P., Gimenez A., Sauvain M., Jullian V., Ginsburg, H. (2002). A non-
radiolabeled ferriprotoporphyrin IX biomineralization inhibition test (FBIT) for the high throughput
screening of ant malarial compounds. Exp. Parasitol. 100: 252-256.
4
Peters,W. and Robinson B.L. (1992). The chemotherapy of rodent malaria. XLVII. Studies on
pyronaridine and other Mannich base antimalarials. Ann.Trop.Med.Parasitol. 86:455-465.
142
In recent years, efforts have been focused on the development of new theoretical
approach to explain in an unified framework the structure-activity relationships of
MAO A and/or B inhibitors. In this sense, we combined the Complex Networks
Analysis with the QSAR methodology called MARCH-INSIDE to carry out an
unified analysis for a very large database of heterogeneous compounds.
Initially, a Markov model was used to calculate molecular descriptors and fit a
classification function based on dataset Principal Components derived with 94.5%
of accuracy (3222 out of 3408 inputs). Next, the values of the PCA scores were
used to calculate Complex Network of MAO inhibitors.
This combined analysis was used for the design of a novel generation of
coumarin-scaffold based MAO inhibitors.
ACTIVITY
QSAR
R
O O O
STRUCTURE
143
a
Department of Organic Chemistry, and bDepartment of Pharmacology. Faculty of
Pharmacy. University of Santiago de Compostela. 15782-Santiago de Compostela, Spain.
O R O N O
X = NH, NCH3, O
N N
R'R'N X R = H, CH3 N
F N
H
I QF4108B
Acknowledgment: We thank the MEC (Ref SAF2005-08025-C03) for the financial support.
M. B. thanks the Spanish Ministerio de Educación y Cultura for a predoctoral Fellowship.
1
Meltzer, H. Y.; Matsubara, S.; Lee, J. J. Pharm. Exp. Ther. 1989, 251, 238.
2
(a) Brea, J.; Rodrigo, J.; Carrieri, A.; Sanz, F.; Cadavid, M. I.; Enguix, M. J.; Villazón, M.; Mengod,
G.; Caro, Y.; Masaguer, C. F.; Raviña, E.; Centeno, N. B.; Carotti, A.; Loza, M. I. J. Med. Chem.
2002, 45, 54; (b) Barceló, M.; Alvarado, M.; Carro, L.; Raviña, E.; Masaguer, C. F. Chem. Biodiv.
2006, 3, 106; (c) Dezi, C.; Brea, J.; Alvarado, M.; Raviña, E.; Masaguer, C. F.; Loza, M. I.; Sanz, F.;
Pastor, M. J. Med. Chem. 2007, 50 (in press).
144
a
Department of Organic Chemistry, and bDepartment of Pharmacology. Faculty of
Pharmacy. University of Santiago de Compostela. 15782-Santiago de Compostela, Spain.
In this communication we will report the synthesis and binding affinity of two
new series D3 antagonists (I and II) based on a benzolactam scaffold, which
maintain three characteristic elements of many dopamine D3 receptor antagonist:
1) an amine moiety, 2) a spacer, usually a linear alkyl chain, and 3) a hydrophobic
residue, often connected through an amide bond. 4 These new compounds will
allow us to evaluate some of the structural requirements for high affinity and
selectivity binding at the D3 receptor.
Ar Ar
O N N
N N
N m
N m
m = 2, 3 II
n I n = 1, 2, 3 n O
Acknowledgment: We thank the Xunta de Galicia (Ref PGIDIT06 PXIB203173PR) for the
financial support.
1
a) Joyce, J. N.; Miador-Woodruff, J. H. Neuropsychopharmacology 1997, 16, 375; b) Joyce, J. N.
Pharmacol. Ther. 2001, 90, 231.
2
Joyce, J. N.; Millan, M. J. Drug Discovery Today 2005,10, 917.
3
Dubuffet, T.; Newman-Tancredi, A.; Cussac, D.; Audinot, V.; Loutz, A.; Millan, M. J.; Lavielle, G.
Bioorg. Med. Chem. Lett. 1999, 9, 2059.
4
Hacling, A. E., Stark, H. ChemBioChem 2002, 3, 946.
145
a
Department of Organic Chemistry, and bDepartment of Pharmacology. Faculty of
Pharmacy. University of Santiago de Compostela. 15782-Santiago de Compostela, Spain.
I
R1
NRR
O
Acknowledgment: We thank the Xunta de Galicia (Ref PGIDIT04 BTF203004PR) for the
financial support.
1
Raviña, E.; Masaguer, C. F.; Curr. Med. Chem.: CNS Agents 2001, 1, 43.
2
(a) Raviña, E.; Casariego, I.; Masaguer, C. F.; Fontenla, J. A.; Montenegro, G. Y.; Rivas, M. E.;
Loza, M. I.; Enguix, M. J.; Villazon, M.; Cadavid, M. I.; Demontis, G. C. J. Med. Chem. 2000, 43,
4678. (b) Brea, J.; Rodrigo, J.; Carrieri, A.; Sanz, F.; Cadavid, M. I.; Enguix, M. J.; Villazón, M.;
Mengod, G.; Caro, Y.; Masaguer, C. F.; Raviña, E.; Centeno, N. B.; Carotti, A.; Loza, M. I. J. Med.
Chem. 2002, 45, 54. (c) Dezi, C.; Brea, J.; Alvarado, M.; Raviña, E.; Masaguer, C. F.; Loza, M. I.;
Sanz, F.; Pastor, M. J. Med. Chem. 2007, 50 (in press).
146
Cancer is a leading cause of death worldwide. 1 Fatty acid synthase (FASN) has
been recently validated as a therapeutic target for the treatment of cancer, since
inhibition of this enzyme induces apoptosis of the tumour cells without producing
damages in the surrounding healthy tissue.2
In a project aimed at the development of new FASN inhibitors, we have
designed, synthesized and characterized a new series of polyphenolic derivatives I
that display high cytotoxic capacity (8 – 125 PM) in several tumour breast cancer
cell lines (SK-Br3, MCF-7 and MDA-MB-231), and do not induce cytotoxicity in
normal fibroblasts. In addition, they inhibit FASN activity confirming that their
cytotoxicity is mediated by direct inhibition of this enzyme. In particular, compound
12 has shown the best properties of cytotoxicity (IC50 = 21 PM in SK-Br3 cells) and
FASN inhibition (90%).3
R1 OBn R2 OH
OH
OBn OH
OH O O
HO OH
R1 OBn
(a) O O
Subunit (b)
+ OBn Subunit Subunit
A HO A A
O O
O R1 R2 O O
OH O O
OBn OH
O
OBn OH
R eagents: (a) DCC, D M A P , TH F; (b) H 2, Pd(O H)2 /C, EtO H , CH 2 Cl2
Reagents: (a) DCC, DMAP, THF; (b) H2, Pd(OH)2, EtOH, CH2Cl2 OH
O
R 1 = O Bn, H
R = OBn, H II
R 21= O H, H
R2 = OH, H
OH
OH
R3
Subunit
=
A
, , , , , 12
Ph
R3 R=3 H,
= H, Me, OMe,
Me, OMe, Ph
Acknowledgements: This work has been supported by MEC predoctoral (CT) and Juan de
la Cierva (TP) fellowships, and grants SAF-2004/07103-C02-01, SAF-2007/67008-C02-01
and FIS-PI04/1417.
1
World Health Organization, http://www.who.int/mediacentre/factsheets/fs297/en/. 2 Liu, X.; Shi, Y.;
3
Giranda, V. L.; Lou, Y. Mol. Cancer Ther. 2006, 5, 494. López-Rodríguez, M. L.; Benhamú, B.;
Turrado, C.; Ortega-Gutiérrez, S.; Puig, T.; Brunet, J.; Colomer, R. Patent EP07110956.
147
H3C
O
H
N
CH3
S
I CO2Et
1
S. Conde, M. P. Arce, C. Pérez. XIV Congreso SEQT, Bilbao 2005
148
Acknowledgment: We thank the NCI for providing us the 32 compounds. This research
was supported by MEC (SAF2005-02608).
1
Martínez, A.; Zudaire, E.; Portal-Núñez, S.; Guédez, L.; Libutti, S. K.; Stetler Stevensor, W. G.;
Cuttitta, F. Cancer Res. 2004, 64, 6489-6494.
2
Lucyk, S.; Taha, H.; Yamamoto, H.; Miskolzie, M.; Kotovych, G. Biopolymers 2006, 81, 295-308.
3
Martinez, A.; Vos, M.; Guedez, L.; Kaur, G.; Chen, Z.; Garayoa, M.; Pio, R.; Moody, T.; Stetler-
Stevenson, W. G.; Kleinman, H. K.; Cuttitta, F. J. Natl. Cancer Inst. 2002, 94 , 1226-1237.
149
a
Div. Farmacia y Tecnología Farmacéutica. Universidad Miguel Hernández. Alicante
b
(Spain) Dept. Química Bioquímica y Biología Molecular. Universidad CEU Cardenal
Herrera, Moncada, Valencia (Spain).
e-mail: mjduart@umh.es
1
Yap, C.W.; Li, Z.R.; Chen, Y.Z.; J Mol Graph Model, 2006, 24, 383
2
Mahmood, I. J. Pharm. Sci., 2006, 95, 1810
150
a
Div. Farmacia y Tecnología Farmacéutica. Universidad Miguel Hernández. Alicante
b
(Spain) Dept. Química Bioquímica y Biología Molecular. Universidad CEU Cardenal
Herrera, Moncada, Valencia (Spain).
1
Cercenado E, Sánchez-Carrillo C.; Alcalá L.; Bouza E. Rev. Clin. Esp., 1997, 197, 12
2
Jason A. Wiles; Yongsheng Song; Qiuping Wang; Edlaine Lucien; Akihiro Hashimoto; Jijun Cheng;
Christopher W. Marlor; Yangsi Ou; Steven D. Podos; Jane A. Thanassi. Bioorganic &medicinal
chemistry letters, 2006, 16, 1277
3
Kier L.B.; Hall L.H. J. Chem. Inf. Comput. Sci, 1997, 37, 548.
151
a
Department of Experimental and Health Sciences, Pompeu Fabra University. bMunicipal
Institute of Medical Research. a,bBarcelona Biomedical Research Park, Dr. Aiguader 88,
08003 Barcelona
Surface plasmon resonance (SPR) is one of the most powerful tools for studying
sugar-protein interactions. In this technique, one of the interacting entities (protein
or sugar) is immobilized onto a sensor chip surface, the other one is flown across
and the resulting read-out enables both quantitation and kinetic analysis of the
interaction. Of the two immobilization options, the sugar-on-chip has demonstrable
advantages. We have designed an approach whereby the sugar (1) is immobilized
via a peptide module (2) on the sensor surface1. The required glycopeptide module
(3) is prepared by chemospecific oxime ligation between the reducing end of the
sugar and an aminooxyacetic acid (Aoa) residue at the peptide N-terminus
(Scheme 1).
1
Scheme 1: Oxime ligation between an N-terminal Aoa-containing peptide and a carbohydrate ligand .
1
Villa-Perelló, M.; Gutiérrez Gallego, R.; Andreu, D. ChemBioChemJ. 2005, 6, 1831.
2
Decostaire, I.P.;Lelièvre, D.; Zhang, H.; Delmas, A.F. Tetrahedron Lett. 2006, 47, 7057.
3
Jiménez-Castells, C; G. de la Torre, B.; Gutiérrez-Gallego, R.; Andreu, D. Bioorg. Med. Chem. Lett.,
in press.
152
1
O2, hQ, MeOH H
1) O
Rosa Bengala
OSiR3 O
O Ac2O, pyr O
2)
OMe
DMAP
3) TBAF
Se han determinado los aspectos estereoquímicos del método así como sus
limitaciones en cuanto al tamaño del anillo formado.2
En esta comunicación se describe la aplicación de esta metodología a la
síntesis de los polioxaciclos I y II, precursores de un gran número de compuestos
biológicamente activos.
H H
O
n
O
O O
H nOMe
I n=1
II n = 2
1
Fall, Y.; Vidal, B.; Alonso, D.; Gómez, G. Tetrahedron Lett. 2003, 44, 4467.
2
(a) Pérez, M.; Canoa, P.; Gómez, G.; Terán, C.; Fall, Y. Tetrahedron Lett. 2004, 45, 5207.; (b)
Teijeira, M.; Suárez, P.L.; Gómez, G.; Terán, C.; Fall, Y. Tetrahedron lett. 2005, 46, 5889. (c)
Canoa, P.; pérez, M.; Covelo, B.; Gómez, G.; Fall, Y. Tetrahedron Lett. 2007, 48, 3441.
153
Los líquidos iónicos (LIs)1 están constituídos por sales que contienen al menos un
componente orgánico y presentan un punto de fusión inferior a 100 ºC. Las sales
más habituales contienen un catión 1,3-dialquilimidazolio, aunque también
abundan los cationes alquilamonio, alquilfosfonio, N-alquilpiridinio o N,N-
dialquilpirrolidinio entre otros . Generalmente, el empleo de los LIs como
disolventes aumenta la velocidad de reacción y la selectividad. Además, se
pueden reutilizar, abaratando así los costes y pueden sustituir a los disolventes
orgánicos tradicionales que son volátiles y muy contaminantes. Por otra parte, los
líquidos iónicos quirales que hicieron su aparición hace unos cinco años,
descubrieron un campo totalmente nuevo en la síntesis asimétrica de compuestos
con potencial actividad biológica.
El gran interés que los LIs han despertado en la comunidad científica
queda reflejado en el aumento exponencial de artículos aparecidos durante los
últimos diez años en revistas especializadas; menos de 50 en 1995 y más de 1500
artículos sobre LIs en el año 2004 (Fig. 1).
Figura 1
En esta comunicación presentamos los últimos resultados obtenidos por nuestro
grupo de investigación en el uso de los líquidos iónicos como disolventes,
catalizadores de reacciones o como soporte de reactivos.
Agradecimientos: Al Ministerio de Educación y Ciencia por la concesión del proyecto
CTQ2007-61788 y a la empresa Green Solutions Chemicals, S.L. por el suministro de
líquidos iónicos.
1
(a) Welton T., Chem. Review., 1999, 99, 2071. (b) Wassercheid, P.; Keim, W. Angew. Chem., Int.
Ed. 2000, 39, 3772. (c) Sheldon, R. Chem. Commun. 2001, 2399. (d) Lee, S.-g. Chem. Commun.
2006, 1049. (e) Miao, W.; Chan, T. H. Acc. Chem. Res. 2006, 39, 897.
154
X X
O AlCl3,
NH2
ClCH2CH2Cl
EtO2C CN EtO2C
reflux
+
N NH2 51-96% N N
H H
12-20 3-11
In this work we describe the synthesis and biological evaluation of the tacrine-1,4-
dihydropyridine (DHP) hybrids (3-11). These multipotent molecules are the result of the
juxtaposition of an acetylcholinesterase inhibitor (AChEI) such as tacrine (1) and a 1,4-
DHP such as nimodipine (2). Compounds 3-11 are very selective and potent AChEIs and
show an excellent neuprotective profile and a moderate Ca2+ channel blockade effect.
Consequently, these molecules are new potential drugs for the treatment of
Alzheimer’s disease.
155
156
1
a) Kolks, G.; Frihart, C. R.; Rabiniwitz H. N.; Lippard, S. J. J. Am. Chem. Soc. 1976, 98, 5720. b)
Coughlin, P. K.; Dewan, J. C.; Lippard, S. J.; Watanabe E.; J.-M. Lehn, J. Am. Chem. Soc. 1979, 101,
2652.
2
Drew, M. G. B.; Cairns, C.; Lavery A.; Nelson, S. M. J. Chem. Soc., Chem. Com. 1980, 1120.
3
Tabbi, G.; Driessen, W. L.; Reedijk, J.; Bonomo, R. P.; Veldman, N.; Spek, A. L. Inorg. Chem. 1997,
36, 1168.
4
a) Salata, C. A.; Youinou, M.-T.; Burrows, C. J. Inorg. Chem. 1991, 30, 3454. b) Weser, U.;
Schubotz, L. M.; Lengfelder, E. J. Mol. Cat., 1981, 13, 249. c) Kimura, E.; Kurogi, Y.; Shionoya M.;
Shiro, M. Inorg. Chem., 1991, 30, 4524.
157
a
Dpto Ingeniería, Div. Farmacia y Tecnología Farmacéutica. Universidad Miguel
Hernández. Alicante (Spain), bDept. Química Bioquímica y Biología Molecular. Universidad
c
CEU Cardenal Herrera, Moncada, Valencia (Spain). Dpto Química-Física. Universidad de
Valencia (Spain)
1
Duart, M.J.; García-Domenech, R.; Antón-Fos, G.M.;Gálvez,J. J. Comput. Aid. Mol. Des. 2001,15,
561
2
Duart, M.J.; García-Domenech, R; Gálvez, J.; Alemán, P.A., Martín-Algarra, RV.; Antón-Fos, G.M.
J. Med. Chem. 2006,49, 3667
3
Watanabe, K.; Nakagawa, H.; Tsurufuji, S. J. Pharmacol. Methods, 1986, 15, 255.
158
Malaria is by far the world’s most important tropical parasitic disease. Mortality,
currently estimated at over a million people per year, has risen in recent years,
probably due to increasing resistance to antimalarial medicines. 1
As a continuation of our research in 3-phenylquinoxaline 1,4-di-N-oxide2 and
with the aim of obtaining new antimalarial agents which can improve the currently
available treatments, new series of 3-(2-furyl) and 3-(2-thienyl)quinoxaline-2-
carbonitrile 1,4-di-N-oxide derivatives have been synthesized following the
classical Beirut reaction. Antiplasmodial activity was evaluated in vitro against
Plasmodium falciparum (chloroquine-sensitive, 3D7, and chloroquine-resistant, K1,
strains) by the incorporation of [3H]-hypoxanthine. Cytotoxicity was tested in KB
cells using the Alamar Blue assay.
Twelve compounds were synthesized and evaluated for antimalarial activity.
Eight of them showed a IC50 lower than 1 M against both 3D7 and K1 strains. Six
of the eight products tested for cytotoxicity demonstrated good selectivity and are
now in more advanced studies. Their potency and selectivity make them valid
leads for synthesizing new compounds that possess better activity.
O O
+ N + N
N R7 N R7 = H, F, Cl, CH3, CF3, OCH3.
+ + X X = O or S
N N
O O
Cl
Previous leader2
IC50 = 1.008 µM
bioisosteric replacement
FcB1 strain (CQ-res)
1
World Health Organization (WHO). In Guidelines for the treatment of malaria. Geneva, 2006.
2
Zarranz, B.; Jaso, A.; Aldana, I.; Monge, A.; Laurel, S.; Deharo, E.; Jullian, V.; Sauvain, M.
Arzneim.-Forsch. 2005, 55, 754-761.
159
Isoserine derivatives constitute the essential fragment in natural products of high therapeutic
value, such as taxol (an anti-cancer agent), bestatin (a dipeptide modifier of the immune
response), and the kinostatins (potent inhibitors of the HIV-1 protease). Due to the E-
aminoalcohol properties of these compounds, one procedure for their synthesis is regio- and
stereoselective opening of chiral oxiranes with amines.
Our group has been using carbohydrate derivatives as chiral templates in the stereoselective
synthesis of different compounds. Recently we have developed a method for the
stereoselective epoxidation of olefins. The aim was to obtain the chiral oxiranes on a
skeleton that could be transformed easily into modified isoserines. The olefinic chain was
joined to the sugar moiety via an acetalic bond in order to be easily separated from the chiral
inductor.
Cinnamal acetals from different carbohydrate derivatives were epoxidized with m-CPBA.
High diastereoisomeric excess were obtained. The major oxirane configuration obtained
depends on the sugar moiety used as chiral inductor in each case.
O H
H O O O H H
Ph O O
O O O
O(c-C6H11) O O
Ph O
O O
NHAc O Ph
OH H H HO O
H
(2S,3R)-2,3-Epoxy-3-phenylpropylidene derivatives
Ph
R
O
O O
Ph O
O O(c-C6H11) O
HO O
NHAc O
HO OMe
OH
(2R,3S)-2,3-Epoxy-3-phenylpropylidene derivatives
The ring-opening reaction of these oxiranes with different nitrogen nucleophiles gave
compounds (in good yields and diastereoisomeric excesses) with an D-hydroxy-E-
aminoacetal structure, and can be used as precursors of phenylisoserine analogues.
N3
Ph O
O O
O O(c-C6H11)
OH HO
Ph O NHAc
O O
O(c-C6H11)
HO
NHAc
N
Ph O
O O
O(c-C6H11)
OH HO
NHAc
We are also developing epoxidation reactions employing chiral dioxiranes generated in situ
from different sugar derivatived ketones and Oxone£. To test their abilitiy as chiral catalysts
we performed the reaction of non chiral alkenes with these ketones. Good enantiomeric
excesses were obtained. The following step is the application of this methodology on
different cinnamal acetals (sugar and hydrobenzoin derivatives). We hope that double
asymmetric induction generated improves the diastereoisomeric excesses first reported.
160
1
Boström J. J Comp-Aid Mol Des. 2002, 15, 1137-1152
161
a
Instituto de Química Médica (Consejo Superior de Investigaciones Científicas), Juan de la
Cierva, 3, 28006-Madrid, Spain
b
Instituto de Química e Biotecnologia, Universidade Federal de Alagoas, Maceió, Alagoas,
57072-970, Brazil
Mosquitoes are responsible for the spread of more diseases than any other group
of arthropods. Of particular interest is Aedes aegypti because of its role as a vector
for the arboviruses responsible for yellow fever and dengue fever, both of which
are endemic to Central and South America, Asia and Africa1.
Quinones have been extensively studied due to their biological activity (especially
antitumoral) and some anthraquinones such as emodin have displayed larvicidal
activity against three mosquito species2
In this work we have examined the larvicidal activity against Aedes aegypti of two
series of naphthoquinones: halonaphthoquinones (I), prepared in several steps
from juglone, and lapachol derivatives (II).
O O
OR
Br
OR O O
I II
R = H, Ac, Me R = H, Li, Ac
162
The development of efficient methods for the synthesis of chiral sulfoxides at the
beginning of the nineties has broadened their application to new processes of
carbon-carbon, and carbon-heteroatom processes.1 Recent interesting applications
include inter alia the utilization of sulfoxides as chiral ligands or ligands precursors
in metal catalyzed asymmetric reactions, in coordination chemistry and as Lewis
base in organocatalysis. On the other hand, due to the importance of chiral amines
which account for the 75% of the total drugs or drug candidates, their preparation
has been a standing area of interest in the last decades. Our group has recently
shown that chiral sulfoxides are excellent organocatalysts in the allylation of
hydrazones with trichloroallyl silane en route to allyl amines.2 Accordingly, excellent
enantioselectivities were obtained in the allylation of benzoyl hydrazones using
simple sulfoxides, though up to 3 equivalents of the organocatalysts were
necessary. In order to determine the mechanism of the reaction and to develop a
catalytic approximation of the process, we have recently applied as ligands a range
of sterically and electronically diverse monodentate and bidentate sulfoxides
obtained by the DAG method. Opportunely, using C2-symmetric bis-sulfoxide 2,
excellent enantioselectivities were obtained in the presence of only 1 equivalent of
the ligand.3
O O O
H S S S
H N R R R
N Cl Ligand Bz NH Fe
Bz N + Si 2
Cl
1
Cl Pri Ligands:
Pri H O O
O
S S
S p-Tol R2 NHR1
p-Tol
3 4
1
Fernández, I.; Khiar, N. Chem Rev. 2003, 103, 3651.
2
Fernández, I.; Valdivia, V.; Gori, B.; Alcudia, F.; Alvárez, E.; Khiar, N. Org. Lett. 2005, 7, 1307.
3
Fernández, I.; Valdivia, V.; Gori, B.; Pernía, M.; Khiar, N. Org. Lett. 2007, 9, 2215.
163
a
Dpto.Química Orgánica-I, Centro Joxe Mari Korta Zentroa, Universidad del País Vasco, 20018
San Sebastián, Guipúzcoa,
b
Dpto. Farmacología, Facultad de Medicina, Universidad del País Vasco, 48940 Leioa,
Vizcaya.
H H R1 R
O O H2N CO2Me
H
N N
N * O
N NH2 H
O N
*
H NH2 HO
NH O O
R2 R2
PLG 1 (R1= iBu, Bn, CH2-CH=CH2); (R2= H, Me) 2
1
Khalil, E.M.; Ojala, W.H.; Pradham, A.; Fair, V. D.; Gleason, W. B.; Mishra, R. K.; Johnson, R. L. J. Med.
Chem 1999, 42, 628-37.
2
Palomo, C.; Aizpurua, J.M.; Benito, A.; Miranda, J.I.; Fratila, R.M.; Matute, C.; Domercq, M.; Gago, F.;
Martin-Santamaría, S.; Linden, A. J. Am. Chem. Soc. 2003, 125, 16243-16260.
3
Palomo, C.; Aizpurua, J.M.; Balentova, E.; Jiménez, A.; Oyarbide, J.; Fratila, R.M.; Miranda, J.I. Org.
Lett. 2007, 9, 101-104.
164
1
Walsh. C. Nature Rev. Microbiol. 2003, 1, 65-70.
2
Jomaa et al. Science 1999, 285, 1573-1576.
165
a
Instituto de Química Orgánica General, Consejo Superior de Investigaciones Científicas C/
Juan de la Cierva 3, 28006 Madrid, Spain. bDpto. Química Física I, Facultad de Ciencias
Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain. cDpto. Química
Orgánica I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040
Madrid, Spain.
R1 O Linker SO 2
Ligand Linker 1
Probe
N(CH 3)2
OR O
N N Linker
ArSO2NH
2
Acknowledgements: to the Comunidad Autónoma de Madrid for the financial support for
the project S-SAL-0249-2006.
1
Saghatelian, A. et al. Nature Chem. Biol. 2005, 1, 130.
2
(a) López-Rodríguez M. L. et al. J. Med. Chem. 2005, 48, 2548. (b) López-Rodríguez M. L. et al. J.
Med. Chem. 2005, 48, 4216.
3
(a) Jaiswal, J. K. et al. Nature Chem. Biol. 2007, 2, 92. (b) Saghatelian, A. et al. Proc. Natl. Acad.
Sci. U.S.A. 2004, 101, 10000. (c) Salisbury, C. M. et al. Proc. Natl. Acad. Sci. U.S.A. 2007, 104,
1171.
166
CH3
O NHR2 O HO
R1-N R1-N R1-N O +
O O HO
1
Hu, E.; Lee, D. Curr. Opin. Investig. Drugs. 2003, 4, 1065.
2
Wehrwein, E. A.; Morthcott, C. A.; Loberg, R.D.; Watts, S. W. J. Pharmacol. Exp. Ther.
2004, 309, 1011.
167
O O
N R1 N
N OH
N H H N H
NH O
H H
O R3
R2 HN
1 NH2 2
O
Acknowledgements. This work was partially supported by funds from CICYT (CTQ 2005-
0315 and BQU 2003-00089) and the Fundació La Marató de TV3 Grant
Marató/2001/012710 (R.F.). M.V. thanks the MECD (Spain) for a predoctoral fellowship.
1
Morphy, R.; Kay, C.; Rankovic, Z. Drug Discovery Today 2004, 9, 641-651.
2
Kanda, T.; Jackson, M.J.; Smith, L.A.; Pearce, R.K.B.; Nakamura, J.; Kase, H.; Kuwana, Y.; Jenner,
P. Experimental Neurology 2000, 162, 321-327
3
Vendrell, M.; Angulo, E.; Casadó, V.; Lluis, C.; Franco, R.; Albericio, F.; Royo, M. Journal of
Medicinal Chemistry, 2007, 50, 3062-3069.
168
Me O
X COEt
Cl N N
H
N N
Cl N
CH2CH2 C6 H5
Cl
Figure 1
1
Vigano, Rubino and Parolaro Molecular and cellular basis of cannabinoid and opioid interactions.
Pharmacology Biochemistry and Behavior 2005, 81, 360-368.
2
Christie Opioid and cannabinoid receptors: friends with benefits or just close friends? Br J Pharmacol
2006, 148, 385-386.
3
Jagerovic, N.; Fernandez Fernandez, C.; Goya Laza, P.; Callado Hernandeo, L. F.; Meana Martinez, J. J.
Pyrazolecarboxamide derivatives, method of obtaining same and use thereof an inverse angonist/agonist
of cannabinoid CB1 and opioid μ receptor. Patent WO2007028849.
169
ĺ (b) ĺ (c)
2-carbetoxiciclopentanona (a)
Tabla I Caracterización espectroscópica
1
H-RMN G > ppm a partir de las bandas del agua (b) y (c)@
13
C-RMN ( į, ppm a partir del TMS)
Calculado Experimental
Piridina DMSO
(c) C-1 157.70 157.67 158.04
C-2 25.04 28.05 28.63
C-3 24.00 20.46 21.07
1
Yanga,C.; Edsall, R.; et al . Parasitology, 2004, 122(4), 287-292
2
Ji-Tai Li and Xiao-Liang Li. Ultrasonics Sonochemistry, 2007, 14 ( 6), 677-679
170
O
H
N
Bz OMe
OH
1
One-pot scissionalkylation or
scissionarylation O
H
N
Bz OMe
O O
H H
N N
Bz OMe Bz OMe
O
N N
O O
O O
2 3 4
43% 80% 59%
1
(a) Pace, J. L.; Yang, G. Biochem. Pharmacol. 2006, 71, 968980. (b) Bridges, R. J.; Esselinger, C.
S. Pharmacol. Therap. 2005, 107, 271-285. (b) Van Bambeke, F. Curr. Opin. Pharmacol. 2004, 4,
471478. (c) Johansen, T. N.; Greenwood, J. R.; Frydenvang, K.; Madsen, U.; Krogsgaard-Larsen, P.
Chirality, 2003, 15, 167179. (d) Beck, G. Synlett 2002, 837–850. (e) Nájera, C. Synlett 2002,
171
177
178
179
180
181
182
183
184
185
186