SlideShare a Scribd company logo
1 of 1
Download to read offline
TEMPLATE DESIGN © 2007
www.PosterPresentations.com
The efficient synthesis of α-fluoromethylhistidine di-hydrochloride, its interaction with HDC,
and the structure-based identification of novel HDC inhibitors
Kelly Considine, Dr. Karl Grozinger, and Dr. Joseph Audie
Sacred Heart University 5151 Park Ave. Fairfield, CT 06825
Abstract
Introduction
Results and Discussion (Continued)
Future Work
References
Methods (Continued)
Acknowledgements
Conclusion
Histidine decarboxylase (HDC) is an enzyme that converts histidine to
histamine. Inhibition of HDC has many medical applications including tumor
suppression, ulcer therapy, circadian rhythm modulation, and allergy
relief. Moreover, HDC inhibitors are needed to study histidine metabolism. α-
fluoromethylhistidine di-hydrochloride (α-FMH) is an extremely potent HDC
inhibitor. Unfortunately, α-FMH is commercially unavailable. Here we report
on the novel, inexpensive, and efficient synthesis of α-FMH, opening the door
to commercially available α-FMH. To study the irreversible binding of α-FMH,
we carried out covalent docking studies on HDC homology models. In an effort
to identify novel inhibitors of HDC, we will perform virtual screening using the
same homology models. To the best of our knowledge, this will be the first
virtual screen applied to HDC. The present study makes an important
contribution to the science of HDC inhibition and suggests new avenues for
studying and therapeutically modulating HDC.
α-fluoromethylhistidine di-hydrochloride (C7H10FN3O2·2HCl ) is one of
the most active histidine decarboxylase inhibitors, but it is not commercially
available (19). Histidine decarboxylase converts histidine to histamine (25).
Histamine: Histamine acts as a neurotransmitter in memory, appetite,
circadian rhythm regulation, intracellular communication, and is a modulator
of cell growth (23). Histamine is involved in allergies, gastric acid secretions,
smooth muscle contraction, vasodilation, inflammatory diseases, bone loss,
tumor progression, learning deficiencies, and epilepsy (17, 18).
Histidine decarboxylase inhibitors: α-FMH has pharmacological actions due to
enzyme inhibition and potential therapeutic uses (19). It can control certain
tumors that have a high histamine level, act as a therapy for ulcers and allergy,
increase slow-wave sleep, decrease motor-excitation, decrease learning and
memory potential, treat inflammatory diseases, neurological and
neuroendocrine diseases, osteoporosis, and neoplasias, and prevent certain
inflammatory responses, peptic ulcers, and schizophrenia (17, 19, 21, 22).
Chemical bonds in α-FMH: Replacement of hydrogen by fluorine should not
have a major consequence on steric hinderance. Fluorine is the most
electronegative element making a polar bond with carbon creating a
hydrophobic molecule that more easily crosses the lipid membranes, in order
to act on the appropriate enzyme.
Ring opening of aziridines: Aziridines have a ring strain of 26.7 kcal/mol which
shifts the reactivity to ring opening, however, when inactivated they are not
highly reactive towards nuclear attack (7). It is predicted that aziridine 1,
scheme 1, will demonstrate ring opening at the least hindered position.
α-fluoromethylhisitidine synthesis: Various patents describe the preparation
of α-FMH, but they are not suitable to scale-up because they are low yielding
and not cost effective. One reagent used to fluorinate amino acids was HF
containing BF3 or SbF5 (12, 13). Other patents describe the fluorination of α-
amino alkanoic esters to yield α-FMH (14, 15). A synthesis was reported using
Seebach’s chemistry of self-regeneration of stereocenters. L-histidine methyl
ester was converted to 1,3-imidazolidinones which was reacted with CH2ClF, a
green house gas (no longer available), to give α-FMH (10).
Molecular docking: There is no definitive active site defined in the literature,
so this will allow us to predict a binding site to use in virtual screening studies.
Kelly Considine acknowledges the financial support from Sacred Heart
University in Fairfield, CT and Boehriniger-Ingelheim Parmaceutical Inc. for the
donation of aziridine (6).
Karl Grozinger acknowledges the valuable support from the Chemical
Development Department at Boehringer-Ingelheim Pharmaceutical Inc. (Victor
Fuchs, Matt Hrapchak, and Jon Lorenz).
Any future research on the synthesis would be to identify a scheme
that allows for aziridine ring opening while preventing dimerization. In
addition, a synthesis that proceeds without racmeization would be ideal.
A goal of the docking study is to put the covalent bond in between α-FMH and
the appropriate serine residue and to further perform monte carlo
minimizations.
After finding the appropriate model with the lowest energy, or possibly
several predicted models, the next step in this project is to use virtual
screening to suggest better small molecule inhibitors for histidine
decarboxylase as potential drug leads. Also, docking of histidine methyl ester,
another known inhibitor, would help to determine the best inhibition.
Furthermore, an enzyme assay would be beneficial in order to
determine the enzymatic inhibition.
The results here indicate that the synthesis produced a moderate yield
of α-FMH at 57%. It was determined that the best yield was achieved when
using 10 equivalents of HF/pyridine to the 1 equivalent of methyl 2-aziridinyl-
3-(N-triphenylmethyl-4-imidazolyl) propionate, but 5 equivalents is sufficient.
This was a novel synthesis that was cost effective and produced a greater yield
than other patents and procedures. NMR, IR, mass spectroscopy, melting
point, and the positive optical rotation all verify that α-FMH was successfully
synthesized.
With future research into the modeling and docking study, we will be
able to predict binding activity and suggest reasonable active site residues. We
will also be able to suggest the residue involved in covalent bonding with α-
FMH. This active site will allow us to examine the potential for other possible
inhibitors using a virtual screening.
1. Altschul, S. F., T. L. Madden, et al. (1997). "Gapped BLAST and PSI-BLAST: a new generation of protein database
search programs." Nucleic Acids Research 25(17): 3389-3402.
2. Arnold, K., L. Bordoli, et al. (2006). "The SWISS-MODEL workspace: a web-based environment for protein
structure homology modelling." Bioinformatics 22(2): 195-201.
3. Bates, P. A., L. A. Kelley, et al. (2001). "Enhancement of Protien Modelling by Human Intervention in Applying the
Automatic Programs 3D-JIGSAW and 3D-PSSM." Proteins: Structure, Function and Genetics Suppl 5: 39-46.
4. Bates, P. A. and M. J. E. Sternberg (1999). "Model Building by Comparison at CASP3: Using Expert Knowledge and
Computer Automation." Proteins: Structure, Function and Genetics Suppl 3: 47-54.
5. Bikadi, Z. and E. Hazai (2009). "Application of the PM6 semi-empirical method to modeling proteins enhances
docking accuracy of AutoDock." Journal of Chemical Information and Modeling 1: 15.
6. Boehringer-Ingelheim Pharmaceutical Inc. donated aziridine 1 to SHU
7. Bowling, N. (2002). Stereospecific Routes to Aziridines, University of Wisconsin-Madison.
8. Combet, C., M. Jambon, et al. (2002). "Geno3D: automatic comparative molecular modelling of protein."
Bioinformatics 18: 213-214.
9. Contreras-Moreira, B. and P. A. Bates (2002). "Domain Fishing: a first step in protein comparative modelling."
Bioinformatics 18: 1141-1142.
10. Grozinger, K. G., R. W. Kriwacki, et al. (1993). "A novel stereoselective route to (S)-(+)-α-(fluoromethyl)histidine: α-
halomethylation of (2R,4S)-3-benzoyl-2-(1,1-dimethylethyl)-1-methyl-4-[(N-tritylimidazol-4-yl)methyl]-1,3-
imidazolidin-5-one. Synthesis and proton NMR spectroscopy." Journal of Organic Chemistry 58(3): 709-713.
11. Kiefer, F., K. Arnold, et al. (2009). "The SWISS-MODEL Repository and associate resources." Nucleic Acids Research
37(Database issue): D387-D392.
12. Kollonitsch, J. and S. Marburg (1981). Fluorination process, Merck & Co., Inc.
13. Kollonitsch, J., S. Marburg, et al. (1975). "Selective Fluorination of Hydroxy Amines and Hydroxy Amino Acids with
Sulfur Tetrafluoride in Liquid Hydrogen Fluoride." Journal of Organic Chemistry 40(25): 3808-3809.
14. Kollonitsch, J. and A. A. Patchett (1982). Fluorinated amino acids. United States, Merck & Co., Inc. .
15. Kollonitsch, J., L. M. Perkins, et al. (1982). Acid addition salts of N-trityl-α-fluoromethylhistidine enantiomer
derivatives, Merck & Co., Inc. .
16. Lambert, C., N. Leonard, et al. (2002). "EsyPred3D: Prediction of proteins 3D structures." Bioinformatics 18(9):
1250-1256.
17. Moya-Garcia, A. A., M. Á. Medina, et al. (2005). "Mammalian histidine decarboxylase: from structure to function."
BioEssays 27(1): 57-63.
18. Moya-García, A. A., J. Ruiz-Pernía, et al. (2008). "Analysis of the decarboxylation step in mammalian histidine
decarboxylase: a computational study." Journal of Biological Chemistry 283: 12393-12401.
19. NCBI. (2005). "alpha-fluoromethylhistidine – Compound Summary (CID: 107653)." PubChem Compound, from
http://pubchem.ncbi.nlm.nih.gov/summary/summary.cgi?cid=107653&loc=ec_rcs.
20. Pieper, U., N. Eswar, et al. (2004). "MODBASE, a database of annotated comparative protein structure models, and
associated resources." Nucleic Acids Research 32(Database issue): D217-D222.
21. Puebla, L. and E. Arilla (1995). "α-Fluoromethylhistidine influences somatostatin content, binding and inhibition of
adenylyl cyclase activity in the rat frontoparietal cortex." Regulatory Peptides 59(1): 111-120.
22. Sakai, N., E. Sakurai, et al. (1995). "Long-term depletion of brain histamine induced by α-fluoromethylhistidine
increases feeding-associated locomotor activity in mice with a modulation of brain amino acid levels." Behavioural
Brain Research 72(1-2): 83-88.
23. Sigma-RBI. (2008). "Histamine Synthesis and Metabolism." Sigma-RBI eHandbook, from
http://www.sigmaaldrich.com/etc/medialib/docs/Sigma/RBI_Handbook/sg_ls_cs_rbibook_histsynth.Par.0001.File.
tmp/sg_ls_cs_rbibook_histsynth.pdf
24. Summa, C. M. and M. Levitt (2007). "Near-native structure refinement using in vacuo energy minimization."
Proceedings of the National Academy of Sciences of the United States of America 104(9): 3177-3182.
25. Tahanejad, F. S., H. Naderi-Manesh, et al. (2000). "Homology-based molecular modelling of PLP-dependent
histidine decarboxylase from Morganella morganii." European Journal of Medicinal Chemistry 35: 567-576.
Methods
The synthesis followed 3 chemical steps that are outlined in scheme 1.
These 3 steps comprise a novel and cost effective method for preparation of α-
FMH from aziridine 1 (6), scheme 1. The last step, 4 to 5, is an effort to isolate
the free amino acid without the di-hydrochloride salt.
1. 0.03 moles methyl 2- aziridinyl-3-(N-triphenylmethyl-4-imidazolyl)
propionate 1 (6) was fluorinated with 0.37 moles HF/Pyridine (70% : 30%)
in toluene at 70°C and stirring to fluorinate and remove the trityl group to
give crude 3. Note: 10 equivalents had the best yield, but it was later
determined that 5 equivalents is sufficient.
2. Calcium carbonate was added to the two phase system and vacuum
filtration was used to isolate calcium fluoride.
3. The crude reaction product, methyl ester 3, was hydrolyzed with 6 N HCl to
form α-FMH 4.
4. Crude α -FMH di-hydrochloride was purified by a cation exchange column
using 0.5-6.0 N HCL.
5. Fractions were collected and concentrated to dryness.
Scheme 1. Novel synthesis of α-FMH
Purification:
Filtrate collected from the 6.0 N HCl elution was re-chromatographed using HCl
concentrations over 0.5-6.0 N. The Pauly’s test was used to test for histidine.
Identificiation:
Melting point, FT-IR, carbon and proton NMR’s (run in DMSO-d6 and D2O),
optical roation, and mass spectroscopy (run at Boehringer-Ingelheim for the
free amino acid monomer of α-FMH) were used for identification.
Sequence alignment:
A BLASTP (1) database search was performed on the 662 amino acid sequence
of Homo sapiens HDC.
Homology model building:
Swiss-Model (11) had the best homology model (previously built) using DOPA
decarboxylase as a model after minimization when compared to ModBase (20),
3D-Jigsaw Comparative Modeling (3, 4, 9), ESyPred3D Web Server 1.0 (16), and
Geno3D (8).
Energy minimization and model refinement:
All models were optimized and refined using Summa Lab’s Protein Refinement
Server (24), and then submitted to the Swiss-Model server (2) for structural
quality checks and energy evaluations.
Inhibitor docking:
α-fluoromethylhistidine was docked into HDC using DockingServer (5).
Results and Discussion
The fractions from 2.0 N HCl yielded 3.3 g of α-FMH from the first
column and the fraction from 3.0 N HCl resulted in 0.7 grams of the dimer or
trimer as indicated by a lower Rf value than the monomer from 2.0 N HCl and
later confirmed with NMR. The product from 2.0 N HCl was further crystallized
with ether and ethanol to give 0.37 grams of crystals. The fraction retrieved
with 6.0 N HCl was rechromatographed on a second column. The fraction from
2.0 N HCl was 1.2 grams and 1.3 grams was retrieved from the fraction of 3.0 N
HCl. The fraction retrieved from 6.0 N HCl had a negative Pauly’s test.
A total of 4.5 g was retrieved (3.3 grams and 1.2 grams of α-FMH
crystals) giving a yield of 57%. Some material was lost during chromatography
and due to the harsh conditions of 100°C and pH 1. These crystals had a
melting point of 194-215°C compared to the literature melting point of 217-
219°C (15).
The optical rotation in H2O at 22°C and 546 nm was +0.09° compared to
a literature rotation of +17.3° in CF3COOH/H2O (15).
Figure 1. FT-IR spectrum of the product isolated with 2 N HCl from column 1
Figure 2 shows the NMR spectra of our crystals obtained from elution
with 2.0 N HCl in column 1 in DMSO-d6. The chemical shifts at 4.80 and 4.92
ppm demonstrate fluorine spin-spin coupling. Each are split into a doublet due
to the long range coupling with the methylene group adjacent to the imidazole
ring.
A B
Figure 2. A- 1H-NMR (DMSO-d6) of α-FMH and B- 13C-NMR (DMSO) of α-FMH
Mass spectroscopy of the free amino acid monomer was run at
Boehringer-Ingelheim and the observed mass was 188.0845, seen in figure 3,
which matches the true molecular weight of α-FMH.
Figure 3. Mass spectra of α-FMH, free amino acid monomer
Therefore, all tests identified the product as the desired α-FMH.
Process development:
Ring-opening of aziridine 1 with KF in DMF, with a catalyst
(tetrabutylammonium hexafluorophosphate 98%) and without a catalyst and
under various temperatures and times resulted in no reaction.
Docking study:
The inhibitor, α-FMH, was docked into the histidine decarboxylase from
Swiss-Model which had a dFire of -706.81 and a QMean of -78.1822.
It was hypothesized that the fluorine of the ligand would interact with a
serine residue in the active site, most likely serine 354, and that the phosphoryl
group would interact with lysine 305. The best docked binding modes agreed
with these predictions. As can be seen in table 1, the dockings numbered 1
and 2 do not show interactions of the phosphoryl group with the lysine
residue, but instead were exposed to the solvent.
Table 1. Docking Models
Serine 354 (pink) in figure 4 A is where the covalent bond is suspected
to form. As can be seen in figure 4 B, the fluorine of the ligand is 3.46 Å away
from the oxygen of the serine so it is within an appropriate distance for a
covalent bond to form.
A B
Figure 4. A – Histidine decarboxylase ribbon diagram (Pink = serine 354), B -
Docking of α-FMH-PLP into histidine decarboxylase active site (white =
phosphoryl, royal blue = α-FMH, green = fluorine, light blue = serine 354, 3.46
= distance between fluorine and serine)
Number Estimated Free
Energy of
Binding
(kcal/mol)
Total
Intermolecular
Energy
(kcal/mol)
Interactions Of
Interest
Distance (Å)
1 -2.50 -5.12 F-Ser 354 3.46
2 -1.54 -3.97 F-Ser 151
F-Ser 196
2.80
3.19
3 +1.03 -3.40 F-Ser 354
P-Lys 305
3.09
3.88
4 +2.48 -0.80 F-Ser 151
F-Ser 196
P-Lys 305
3.35
3.11
3.51
N
N
H
NH2
O
CH3
O
F
N
N
NH
O
O
CH3
N
N
O
O
CH3
F
NH2
HF/pyridine
(S)-alpha Fluoromethylhistidine x 2HCl
HCl aqueous
N
N
H
NH2
OHO
F
HCl aqueous
N
N
H
NH2
OHO
F
NH4OH
(S)-alpha Fluoromethylhistidine
1
2
3 4
5
Fluorination
Remove Trityl
Protecting Group
Ester Hydrolysis

More Related Content

What's hot

71st ICREA Colloquium - Intrinsically disordered proteins (IDPs) the challeng...
71st ICREA Colloquium - Intrinsically disordered proteins (IDPs) the challeng...71st ICREA Colloquium - Intrinsically disordered proteins (IDPs) the challeng...
71st ICREA Colloquium - Intrinsically disordered proteins (IDPs) the challeng...Mayi Suárez
 
Novel Hybrid Molecules of Quinazoline Chalcone Derivatives: Synthesis and Stu...
Novel Hybrid Molecules of Quinazoline Chalcone Derivatives: Synthesis and Stu...Novel Hybrid Molecules of Quinazoline Chalcone Derivatives: Synthesis and Stu...
Novel Hybrid Molecules of Quinazoline Chalcone Derivatives: Synthesis and Stu...Ratnakaram Venkata Nadh
 
Synthesis and characterization of new benzotriazole derivatives for possible ...
Synthesis and characterization of new benzotriazole derivatives for possible ...Synthesis and characterization of new benzotriazole derivatives for possible ...
Synthesis and characterization of new benzotriazole derivatives for possible ...SriramNagarajan18
 
Recent Structure Activity Relationship Studies of 1,4-Benzodiazepines
Recent Structure Activity Relationship Studies of 1,4-BenzodiazepinesRecent Structure Activity Relationship Studies of 1,4-Benzodiazepines
Recent Structure Activity Relationship Studies of 1,4-Benzodiazepinespeertechzpublication
 
Eckenhoff et al 2001
Eckenhoff et al 2001Eckenhoff et al 2001
Eckenhoff et al 2001Kin Chan
 
Scaffold hopping in drug development ppt
Scaffold hopping in drug development  pptScaffold hopping in drug development  ppt
Scaffold hopping in drug development pptAkliluSamuel3
 
Prediction of disorder in protein structure (amit singh)
Prediction of disorder in protein structure (amit singh)Prediction of disorder in protein structure (amit singh)
Prediction of disorder in protein structure (amit singh)Amit Singh
 
Determining stable ligand orientation
Determining stable ligand orientationDetermining stable ligand orientation
Determining stable ligand orientationijaia
 
Effect of glycyrrhizic acid on protein binding of diltiazem,
Effect of glycyrrhizic acid on protein binding of diltiazem,Effect of glycyrrhizic acid on protein binding of diltiazem,
Effect of glycyrrhizic acid on protein binding of diltiazem,Grace Ginz Sinusinga
 
Synthesis and Catalytic Application of Chiral 1,1′-Bi-2-naphtholand Biphenant...
Synthesis and Catalytic Application of Chiral 1,1′-Bi-2-naphtholand Biphenant...Synthesis and Catalytic Application of Chiral 1,1′-Bi-2-naphtholand Biphenant...
Synthesis and Catalytic Application of Chiral 1,1′-Bi-2-naphtholand Biphenant...DrMAdamSayah
 
In-vitro Interaction of αB-Crystallin on Serum Amyloid A and Serum Amyloid A ...
In-vitro Interaction of αB-Crystallin on Serum Amyloid A and Serum Amyloid A ...In-vitro Interaction of αB-Crystallin on Serum Amyloid A and Serum Amyloid A ...
In-vitro Interaction of αB-Crystallin on Serum Amyloid A and Serum Amyloid A ...theijes
 
In silico discovery of histone methyltranferase 1
In silico discovery of histone methyltranferase 1In silico discovery of histone methyltranferase 1
In silico discovery of histone methyltranferase 1juancarlosrise
 
Stable Drug Designing by Minimizing Drug Protein Interaction Energy Using PSO
Stable Drug Designing by Minimizing Drug Protein Interaction Energy Using PSO Stable Drug Designing by Minimizing Drug Protein Interaction Energy Using PSO
Stable Drug Designing by Minimizing Drug Protein Interaction Energy Using PSO csandit
 
Peptide Ligation Honorus Project Report Mikolaj Zabrocki
Peptide Ligation Honorus Project Report Mikolaj ZabrockiPeptide Ligation Honorus Project Report Mikolaj Zabrocki
Peptide Ligation Honorus Project Report Mikolaj ZabrockiMikolaj Zabrocki
 

What's hot (20)

Sekhon 12
Sekhon 12Sekhon 12
Sekhon 12
 
71st ICREA Colloquium - Intrinsically disordered proteins (IDPs) the challeng...
71st ICREA Colloquium - Intrinsically disordered proteins (IDPs) the challeng...71st ICREA Colloquium - Intrinsically disordered proteins (IDPs) the challeng...
71st ICREA Colloquium - Intrinsically disordered proteins (IDPs) the challeng...
 
Novel Hybrid Molecules of Quinazoline Chalcone Derivatives: Synthesis and Stu...
Novel Hybrid Molecules of Quinazoline Chalcone Derivatives: Synthesis and Stu...Novel Hybrid Molecules of Quinazoline Chalcone Derivatives: Synthesis and Stu...
Novel Hybrid Molecules of Quinazoline Chalcone Derivatives: Synthesis and Stu...
 
Synthesis and characterization of new benzotriazole derivatives for possible ...
Synthesis and characterization of new benzotriazole derivatives for possible ...Synthesis and characterization of new benzotriazole derivatives for possible ...
Synthesis and characterization of new benzotriazole derivatives for possible ...
 
Recent Structure Activity Relationship Studies of 1,4-Benzodiazepines
Recent Structure Activity Relationship Studies of 1,4-BenzodiazepinesRecent Structure Activity Relationship Studies of 1,4-Benzodiazepines
Recent Structure Activity Relationship Studies of 1,4-Benzodiazepines
 
Eckenhoff et al 2001
Eckenhoff et al 2001Eckenhoff et al 2001
Eckenhoff et al 2001
 
Scaffold hopping in drug development ppt
Scaffold hopping in drug development  pptScaffold hopping in drug development  ppt
Scaffold hopping in drug development ppt
 
Prediction of disorder in protein structure (amit singh)
Prediction of disorder in protein structure (amit singh)Prediction of disorder in protein structure (amit singh)
Prediction of disorder in protein structure (amit singh)
 
Determining stable ligand orientation
Determining stable ligand orientationDetermining stable ligand orientation
Determining stable ligand orientation
 
Effect of glycyrrhizic acid on protein binding of diltiazem,
Effect of glycyrrhizic acid on protein binding of diltiazem,Effect of glycyrrhizic acid on protein binding of diltiazem,
Effect of glycyrrhizic acid on protein binding of diltiazem,
 
Synthesis and Catalytic Application of Chiral 1,1′-Bi-2-naphtholand Biphenant...
Synthesis and Catalytic Application of Chiral 1,1′-Bi-2-naphtholand Biphenant...Synthesis and Catalytic Application of Chiral 1,1′-Bi-2-naphtholand Biphenant...
Synthesis and Catalytic Application of Chiral 1,1′-Bi-2-naphtholand Biphenant...
 
In-vitro Interaction of αB-Crystallin on Serum Amyloid A and Serum Amyloid A ...
In-vitro Interaction of αB-Crystallin on Serum Amyloid A and Serum Amyloid A ...In-vitro Interaction of αB-Crystallin on Serum Amyloid A and Serum Amyloid A ...
In-vitro Interaction of αB-Crystallin on Serum Amyloid A and Serum Amyloid A ...
 
c4ra02698e
c4ra02698ec4ra02698e
c4ra02698e
 
MI-4
MI-4MI-4
MI-4
 
LTA4H FBDD 2009 JMC
LTA4H FBDD 2009 JMCLTA4H FBDD 2009 JMC
LTA4H FBDD 2009 JMC
 
In silico discovery of histone methyltranferase 1
In silico discovery of histone methyltranferase 1In silico discovery of histone methyltranferase 1
In silico discovery of histone methyltranferase 1
 
Stable Drug Designing by Minimizing Drug Protein Interaction Energy Using PSO
Stable Drug Designing by Minimizing Drug Protein Interaction Energy Using PSO Stable Drug Designing by Minimizing Drug Protein Interaction Energy Using PSO
Stable Drug Designing by Minimizing Drug Protein Interaction Energy Using PSO
 
Peptide Ligation Honorus Project Report Mikolaj Zabrocki
Peptide Ligation Honorus Project Report Mikolaj ZabrockiPeptide Ligation Honorus Project Report Mikolaj Zabrocki
Peptide Ligation Honorus Project Report Mikolaj Zabrocki
 
P02 ab sar
P02 ab sarP02 ab sar
P02 ab sar
 
Paper
PaperPaper
Paper
 

Similar to a-FMH Poster

MOLECULAR DOCKING AND RELATED DRUG DESIGN ACHIEVEMENTS
MOLECULAR DOCKING AND RELATED DRUG DESIGN ACHIEVEMENTS MOLECULAR DOCKING AND RELATED DRUG DESIGN ACHIEVEMENTS
MOLECULAR DOCKING AND RELATED DRUG DESIGN ACHIEVEMENTS santosh Kumbhar
 
SF and PE CTR-IN 2016 Poster_FInal
SF and PE CTR-IN 2016 Poster_FInalSF and PE CTR-IN 2016 Poster_FInal
SF and PE CTR-IN 2016 Poster_FInalSteve Flynn
 
Perspective on QSAR modeling of transport
Perspective on QSAR modeling of transportPerspective on QSAR modeling of transport
Perspective on QSAR modeling of transportSean Ekins
 
Homology modeling, docking and comparative study of the selectivity of some h...
Homology modeling, docking and comparative study of the selectivity of some h...Homology modeling, docking and comparative study of the selectivity of some h...
Homology modeling, docking and comparative study of the selectivity of some h...Alexander Decker
 
Pharmacology Of Pain Essay
Pharmacology Of Pain EssayPharmacology Of Pain Essay
Pharmacology Of Pain EssayLeslie Lee
 
Accelrys UGM slides 2011
Accelrys UGM slides 2011Accelrys UGM slides 2011
Accelrys UGM slides 2011Sean Ekins
 
Systems Pharmacology as a tool for future therapy development: a feasibility ...
Systems Pharmacology as a tool for future therapy development: a feasibility ...Systems Pharmacology as a tool for future therapy development: a feasibility ...
Systems Pharmacology as a tool for future therapy development: a feasibility ...Guide to PHARMACOLOGY
 
17064-9fQK1439202911-
17064-9fQK1439202911-17064-9fQK1439202911-
17064-9fQK1439202911-rajkutty1983
 
Epigenetic modulators - review - BMCL digest
Epigenetic modulators - review - BMCL digestEpigenetic modulators - review - BMCL digest
Epigenetic modulators - review - BMCL digestBoobalan Pachaiyappan
 
Bacteria Induced Cryptic Meroterpenoid Pathway in Pathogenic Aspergillus fumi...
Bacteria Induced Cryptic Meroterpenoid Pathway in Pathogenic Aspergillus fumi...Bacteria Induced Cryptic Meroterpenoid Pathway in Pathogenic Aspergillus fumi...
Bacteria Induced Cryptic Meroterpenoid Pathway in Pathogenic Aspergillus fumi...Debanjan Chatterjee
 
Network Pharmacology Tri-Con 022212
Network Pharmacology Tri-Con 022212Network Pharmacology Tri-Con 022212
Network Pharmacology Tri-Con 022212Philip Bourne
 
69. synthesis, ex vivo and in silico studies of 3 cyano-2-pyridone derivative...
69. synthesis, ex vivo and in silico studies of 3 cyano-2-pyridone derivative...69. synthesis, ex vivo and in silico studies of 3 cyano-2-pyridone derivative...
69. synthesis, ex vivo and in silico studies of 3 cyano-2-pyridone derivative...Fernando Hernandez Borja
 
Xia G. PYT article. J Med Chem. Mar 4 2011
Xia G. PYT article. J Med Chem. Mar 4 2011Xia G. PYT article. J Med Chem. Mar 4 2011
Xia G. PYT article. J Med Chem. Mar 4 2011nemata
 

Similar to a-FMH Poster (20)

MOLECULAR DOCKING AND RELATED DRUG DESIGN ACHIEVEMENTS
MOLECULAR DOCKING AND RELATED DRUG DESIGN ACHIEVEMENTS MOLECULAR DOCKING AND RELATED DRUG DESIGN ACHIEVEMENTS
MOLECULAR DOCKING AND RELATED DRUG DESIGN ACHIEVEMENTS
 
published
publishedpublished
published
 
SF and PE CTR-IN 2016 Poster_FInal
SF and PE CTR-IN 2016 Poster_FInalSF and PE CTR-IN 2016 Poster_FInal
SF and PE CTR-IN 2016 Poster_FInal
 
Perspective on QSAR modeling of transport
Perspective on QSAR modeling of transportPerspective on QSAR modeling of transport
Perspective on QSAR modeling of transport
 
LSD1 - bmc-paper
LSD1 - bmc-paperLSD1 - bmc-paper
LSD1 - bmc-paper
 
Homology modeling, docking and comparative study of the selectivity of some h...
Homology modeling, docking and comparative study of the selectivity of some h...Homology modeling, docking and comparative study of the selectivity of some h...
Homology modeling, docking and comparative study of the selectivity of some h...
 
BCSRCv1.3
BCSRCv1.3BCSRCv1.3
BCSRCv1.3
 
Pharmacology Of Pain Essay
Pharmacology Of Pain EssayPharmacology Of Pain Essay
Pharmacology Of Pain Essay
 
Accelrys UGM slides 2011
Accelrys UGM slides 2011Accelrys UGM slides 2011
Accelrys UGM slides 2011
 
Molecular docking
Molecular dockingMolecular docking
Molecular docking
 
Systems Pharmacology as a tool for future therapy development: a feasibility ...
Systems Pharmacology as a tool for future therapy development: a feasibility ...Systems Pharmacology as a tool for future therapy development: a feasibility ...
Systems Pharmacology as a tool for future therapy development: a feasibility ...
 
Peptidomimetics
PeptidomimeticsPeptidomimetics
Peptidomimetics
 
2012 JMC Amidoximes HiRes
2012 JMC Amidoximes HiRes2012 JMC Amidoximes HiRes
2012 JMC Amidoximes HiRes
 
INVENTI-LAVANYA
INVENTI-LAVANYAINVENTI-LAVANYA
INVENTI-LAVANYA
 
17064-9fQK1439202911-
17064-9fQK1439202911-17064-9fQK1439202911-
17064-9fQK1439202911-
 
Epigenetic modulators - review - BMCL digest
Epigenetic modulators - review - BMCL digestEpigenetic modulators - review - BMCL digest
Epigenetic modulators - review - BMCL digest
 
Bacteria Induced Cryptic Meroterpenoid Pathway in Pathogenic Aspergillus fumi...
Bacteria Induced Cryptic Meroterpenoid Pathway in Pathogenic Aspergillus fumi...Bacteria Induced Cryptic Meroterpenoid Pathway in Pathogenic Aspergillus fumi...
Bacteria Induced Cryptic Meroterpenoid Pathway in Pathogenic Aspergillus fumi...
 
Network Pharmacology Tri-Con 022212
Network Pharmacology Tri-Con 022212Network Pharmacology Tri-Con 022212
Network Pharmacology Tri-Con 022212
 
69. synthesis, ex vivo and in silico studies of 3 cyano-2-pyridone derivative...
69. synthesis, ex vivo and in silico studies of 3 cyano-2-pyridone derivative...69. synthesis, ex vivo and in silico studies of 3 cyano-2-pyridone derivative...
69. synthesis, ex vivo and in silico studies of 3 cyano-2-pyridone derivative...
 
Xia G. PYT article. J Med Chem. Mar 4 2011
Xia G. PYT article. J Med Chem. Mar 4 2011Xia G. PYT article. J Med Chem. Mar 4 2011
Xia G. PYT article. J Med Chem. Mar 4 2011
 

a-FMH Poster

  • 1. TEMPLATE DESIGN © 2007 www.PosterPresentations.com The efficient synthesis of α-fluoromethylhistidine di-hydrochloride, its interaction with HDC, and the structure-based identification of novel HDC inhibitors Kelly Considine, Dr. Karl Grozinger, and Dr. Joseph Audie Sacred Heart University 5151 Park Ave. Fairfield, CT 06825 Abstract Introduction Results and Discussion (Continued) Future Work References Methods (Continued) Acknowledgements Conclusion Histidine decarboxylase (HDC) is an enzyme that converts histidine to histamine. Inhibition of HDC has many medical applications including tumor suppression, ulcer therapy, circadian rhythm modulation, and allergy relief. Moreover, HDC inhibitors are needed to study histidine metabolism. α- fluoromethylhistidine di-hydrochloride (α-FMH) is an extremely potent HDC inhibitor. Unfortunately, α-FMH is commercially unavailable. Here we report on the novel, inexpensive, and efficient synthesis of α-FMH, opening the door to commercially available α-FMH. To study the irreversible binding of α-FMH, we carried out covalent docking studies on HDC homology models. In an effort to identify novel inhibitors of HDC, we will perform virtual screening using the same homology models. To the best of our knowledge, this will be the first virtual screen applied to HDC. The present study makes an important contribution to the science of HDC inhibition and suggests new avenues for studying and therapeutically modulating HDC. α-fluoromethylhistidine di-hydrochloride (C7H10FN3O2·2HCl ) is one of the most active histidine decarboxylase inhibitors, but it is not commercially available (19). Histidine decarboxylase converts histidine to histamine (25). Histamine: Histamine acts as a neurotransmitter in memory, appetite, circadian rhythm regulation, intracellular communication, and is a modulator of cell growth (23). Histamine is involved in allergies, gastric acid secretions, smooth muscle contraction, vasodilation, inflammatory diseases, bone loss, tumor progression, learning deficiencies, and epilepsy (17, 18). Histidine decarboxylase inhibitors: α-FMH has pharmacological actions due to enzyme inhibition and potential therapeutic uses (19). It can control certain tumors that have a high histamine level, act as a therapy for ulcers and allergy, increase slow-wave sleep, decrease motor-excitation, decrease learning and memory potential, treat inflammatory diseases, neurological and neuroendocrine diseases, osteoporosis, and neoplasias, and prevent certain inflammatory responses, peptic ulcers, and schizophrenia (17, 19, 21, 22). Chemical bonds in α-FMH: Replacement of hydrogen by fluorine should not have a major consequence on steric hinderance. Fluorine is the most electronegative element making a polar bond with carbon creating a hydrophobic molecule that more easily crosses the lipid membranes, in order to act on the appropriate enzyme. Ring opening of aziridines: Aziridines have a ring strain of 26.7 kcal/mol which shifts the reactivity to ring opening, however, when inactivated they are not highly reactive towards nuclear attack (7). It is predicted that aziridine 1, scheme 1, will demonstrate ring opening at the least hindered position. α-fluoromethylhisitidine synthesis: Various patents describe the preparation of α-FMH, but they are not suitable to scale-up because they are low yielding and not cost effective. One reagent used to fluorinate amino acids was HF containing BF3 or SbF5 (12, 13). Other patents describe the fluorination of α- amino alkanoic esters to yield α-FMH (14, 15). A synthesis was reported using Seebach’s chemistry of self-regeneration of stereocenters. L-histidine methyl ester was converted to 1,3-imidazolidinones which was reacted with CH2ClF, a green house gas (no longer available), to give α-FMH (10). Molecular docking: There is no definitive active site defined in the literature, so this will allow us to predict a binding site to use in virtual screening studies. Kelly Considine acknowledges the financial support from Sacred Heart University in Fairfield, CT and Boehriniger-Ingelheim Parmaceutical Inc. for the donation of aziridine (6). Karl Grozinger acknowledges the valuable support from the Chemical Development Department at Boehringer-Ingelheim Pharmaceutical Inc. (Victor Fuchs, Matt Hrapchak, and Jon Lorenz). Any future research on the synthesis would be to identify a scheme that allows for aziridine ring opening while preventing dimerization. In addition, a synthesis that proceeds without racmeization would be ideal. A goal of the docking study is to put the covalent bond in between α-FMH and the appropriate serine residue and to further perform monte carlo minimizations. After finding the appropriate model with the lowest energy, or possibly several predicted models, the next step in this project is to use virtual screening to suggest better small molecule inhibitors for histidine decarboxylase as potential drug leads. Also, docking of histidine methyl ester, another known inhibitor, would help to determine the best inhibition. Furthermore, an enzyme assay would be beneficial in order to determine the enzymatic inhibition. The results here indicate that the synthesis produced a moderate yield of α-FMH at 57%. It was determined that the best yield was achieved when using 10 equivalents of HF/pyridine to the 1 equivalent of methyl 2-aziridinyl- 3-(N-triphenylmethyl-4-imidazolyl) propionate, but 5 equivalents is sufficient. This was a novel synthesis that was cost effective and produced a greater yield than other patents and procedures. NMR, IR, mass spectroscopy, melting point, and the positive optical rotation all verify that α-FMH was successfully synthesized. With future research into the modeling and docking study, we will be able to predict binding activity and suggest reasonable active site residues. We will also be able to suggest the residue involved in covalent bonding with α- FMH. This active site will allow us to examine the potential for other possible inhibitors using a virtual screening. 1. Altschul, S. F., T. L. Madden, et al. (1997). "Gapped BLAST and PSI-BLAST: a new generation of protein database search programs." Nucleic Acids Research 25(17): 3389-3402. 2. Arnold, K., L. Bordoli, et al. (2006). "The SWISS-MODEL workspace: a web-based environment for protein structure homology modelling." Bioinformatics 22(2): 195-201. 3. Bates, P. A., L. A. Kelley, et al. (2001). "Enhancement of Protien Modelling by Human Intervention in Applying the Automatic Programs 3D-JIGSAW and 3D-PSSM." Proteins: Structure, Function and Genetics Suppl 5: 39-46. 4. Bates, P. A. and M. J. E. Sternberg (1999). "Model Building by Comparison at CASP3: Using Expert Knowledge and Computer Automation." Proteins: Structure, Function and Genetics Suppl 3: 47-54. 5. Bikadi, Z. and E. Hazai (2009). "Application of the PM6 semi-empirical method to modeling proteins enhances docking accuracy of AutoDock." Journal of Chemical Information and Modeling 1: 15. 6. Boehringer-Ingelheim Pharmaceutical Inc. donated aziridine 1 to SHU 7. Bowling, N. (2002). Stereospecific Routes to Aziridines, University of Wisconsin-Madison. 8. Combet, C., M. Jambon, et al. (2002). "Geno3D: automatic comparative molecular modelling of protein." Bioinformatics 18: 213-214. 9. Contreras-Moreira, B. and P. A. Bates (2002). "Domain Fishing: a first step in protein comparative modelling." Bioinformatics 18: 1141-1142. 10. Grozinger, K. G., R. W. Kriwacki, et al. (1993). "A novel stereoselective route to (S)-(+)-α-(fluoromethyl)histidine: α- halomethylation of (2R,4S)-3-benzoyl-2-(1,1-dimethylethyl)-1-methyl-4-[(N-tritylimidazol-4-yl)methyl]-1,3- imidazolidin-5-one. Synthesis and proton NMR spectroscopy." Journal of Organic Chemistry 58(3): 709-713. 11. Kiefer, F., K. Arnold, et al. (2009). "The SWISS-MODEL Repository and associate resources." Nucleic Acids Research 37(Database issue): D387-D392. 12. Kollonitsch, J. and S. Marburg (1981). Fluorination process, Merck & Co., Inc. 13. Kollonitsch, J., S. Marburg, et al. (1975). "Selective Fluorination of Hydroxy Amines and Hydroxy Amino Acids with Sulfur Tetrafluoride in Liquid Hydrogen Fluoride." Journal of Organic Chemistry 40(25): 3808-3809. 14. Kollonitsch, J. and A. A. Patchett (1982). Fluorinated amino acids. United States, Merck & Co., Inc. . 15. Kollonitsch, J., L. M. Perkins, et al. (1982). Acid addition salts of N-trityl-α-fluoromethylhistidine enantiomer derivatives, Merck & Co., Inc. . 16. Lambert, C., N. Leonard, et al. (2002). "EsyPred3D: Prediction of proteins 3D structures." Bioinformatics 18(9): 1250-1256. 17. Moya-Garcia, A. A., M. Á. Medina, et al. (2005). "Mammalian histidine decarboxylase: from structure to function." BioEssays 27(1): 57-63. 18. Moya-García, A. A., J. Ruiz-Pernía, et al. (2008). "Analysis of the decarboxylation step in mammalian histidine decarboxylase: a computational study." Journal of Biological Chemistry 283: 12393-12401. 19. NCBI. (2005). "alpha-fluoromethylhistidine – Compound Summary (CID: 107653)." PubChem Compound, from http://pubchem.ncbi.nlm.nih.gov/summary/summary.cgi?cid=107653&loc=ec_rcs. 20. Pieper, U., N. Eswar, et al. (2004). "MODBASE, a database of annotated comparative protein structure models, and associated resources." Nucleic Acids Research 32(Database issue): D217-D222. 21. Puebla, L. and E. Arilla (1995). "α-Fluoromethylhistidine influences somatostatin content, binding and inhibition of adenylyl cyclase activity in the rat frontoparietal cortex." Regulatory Peptides 59(1): 111-120. 22. Sakai, N., E. Sakurai, et al. (1995). "Long-term depletion of brain histamine induced by α-fluoromethylhistidine increases feeding-associated locomotor activity in mice with a modulation of brain amino acid levels." Behavioural Brain Research 72(1-2): 83-88. 23. Sigma-RBI. (2008). "Histamine Synthesis and Metabolism." Sigma-RBI eHandbook, from http://www.sigmaaldrich.com/etc/medialib/docs/Sigma/RBI_Handbook/sg_ls_cs_rbibook_histsynth.Par.0001.File. tmp/sg_ls_cs_rbibook_histsynth.pdf 24. Summa, C. M. and M. Levitt (2007). "Near-native structure refinement using in vacuo energy minimization." Proceedings of the National Academy of Sciences of the United States of America 104(9): 3177-3182. 25. Tahanejad, F. S., H. Naderi-Manesh, et al. (2000). "Homology-based molecular modelling of PLP-dependent histidine decarboxylase from Morganella morganii." European Journal of Medicinal Chemistry 35: 567-576. Methods The synthesis followed 3 chemical steps that are outlined in scheme 1. These 3 steps comprise a novel and cost effective method for preparation of α- FMH from aziridine 1 (6), scheme 1. The last step, 4 to 5, is an effort to isolate the free amino acid without the di-hydrochloride salt. 1. 0.03 moles methyl 2- aziridinyl-3-(N-triphenylmethyl-4-imidazolyl) propionate 1 (6) was fluorinated with 0.37 moles HF/Pyridine (70% : 30%) in toluene at 70°C and stirring to fluorinate and remove the trityl group to give crude 3. Note: 10 equivalents had the best yield, but it was later determined that 5 equivalents is sufficient. 2. Calcium carbonate was added to the two phase system and vacuum filtration was used to isolate calcium fluoride. 3. The crude reaction product, methyl ester 3, was hydrolyzed with 6 N HCl to form α-FMH 4. 4. Crude α -FMH di-hydrochloride was purified by a cation exchange column using 0.5-6.0 N HCL. 5. Fractions were collected and concentrated to dryness. Scheme 1. Novel synthesis of α-FMH Purification: Filtrate collected from the 6.0 N HCl elution was re-chromatographed using HCl concentrations over 0.5-6.0 N. The Pauly’s test was used to test for histidine. Identificiation: Melting point, FT-IR, carbon and proton NMR’s (run in DMSO-d6 and D2O), optical roation, and mass spectroscopy (run at Boehringer-Ingelheim for the free amino acid monomer of α-FMH) were used for identification. Sequence alignment: A BLASTP (1) database search was performed on the 662 amino acid sequence of Homo sapiens HDC. Homology model building: Swiss-Model (11) had the best homology model (previously built) using DOPA decarboxylase as a model after minimization when compared to ModBase (20), 3D-Jigsaw Comparative Modeling (3, 4, 9), ESyPred3D Web Server 1.0 (16), and Geno3D (8). Energy minimization and model refinement: All models were optimized and refined using Summa Lab’s Protein Refinement Server (24), and then submitted to the Swiss-Model server (2) for structural quality checks and energy evaluations. Inhibitor docking: α-fluoromethylhistidine was docked into HDC using DockingServer (5). Results and Discussion The fractions from 2.0 N HCl yielded 3.3 g of α-FMH from the first column and the fraction from 3.0 N HCl resulted in 0.7 grams of the dimer or trimer as indicated by a lower Rf value than the monomer from 2.0 N HCl and later confirmed with NMR. The product from 2.0 N HCl was further crystallized with ether and ethanol to give 0.37 grams of crystals. The fraction retrieved with 6.0 N HCl was rechromatographed on a second column. The fraction from 2.0 N HCl was 1.2 grams and 1.3 grams was retrieved from the fraction of 3.0 N HCl. The fraction retrieved from 6.0 N HCl had a negative Pauly’s test. A total of 4.5 g was retrieved (3.3 grams and 1.2 grams of α-FMH crystals) giving a yield of 57%. Some material was lost during chromatography and due to the harsh conditions of 100°C and pH 1. These crystals had a melting point of 194-215°C compared to the literature melting point of 217- 219°C (15). The optical rotation in H2O at 22°C and 546 nm was +0.09° compared to a literature rotation of +17.3° in CF3COOH/H2O (15). Figure 1. FT-IR spectrum of the product isolated with 2 N HCl from column 1 Figure 2 shows the NMR spectra of our crystals obtained from elution with 2.0 N HCl in column 1 in DMSO-d6. The chemical shifts at 4.80 and 4.92 ppm demonstrate fluorine spin-spin coupling. Each are split into a doublet due to the long range coupling with the methylene group adjacent to the imidazole ring. A B Figure 2. A- 1H-NMR (DMSO-d6) of α-FMH and B- 13C-NMR (DMSO) of α-FMH Mass spectroscopy of the free amino acid monomer was run at Boehringer-Ingelheim and the observed mass was 188.0845, seen in figure 3, which matches the true molecular weight of α-FMH. Figure 3. Mass spectra of α-FMH, free amino acid monomer Therefore, all tests identified the product as the desired α-FMH. Process development: Ring-opening of aziridine 1 with KF in DMF, with a catalyst (tetrabutylammonium hexafluorophosphate 98%) and without a catalyst and under various temperatures and times resulted in no reaction. Docking study: The inhibitor, α-FMH, was docked into the histidine decarboxylase from Swiss-Model which had a dFire of -706.81 and a QMean of -78.1822. It was hypothesized that the fluorine of the ligand would interact with a serine residue in the active site, most likely serine 354, and that the phosphoryl group would interact with lysine 305. The best docked binding modes agreed with these predictions. As can be seen in table 1, the dockings numbered 1 and 2 do not show interactions of the phosphoryl group with the lysine residue, but instead were exposed to the solvent. Table 1. Docking Models Serine 354 (pink) in figure 4 A is where the covalent bond is suspected to form. As can be seen in figure 4 B, the fluorine of the ligand is 3.46 Å away from the oxygen of the serine so it is within an appropriate distance for a covalent bond to form. A B Figure 4. A – Histidine decarboxylase ribbon diagram (Pink = serine 354), B - Docking of α-FMH-PLP into histidine decarboxylase active site (white = phosphoryl, royal blue = α-FMH, green = fluorine, light blue = serine 354, 3.46 = distance between fluorine and serine) Number Estimated Free Energy of Binding (kcal/mol) Total Intermolecular Energy (kcal/mol) Interactions Of Interest Distance (Å) 1 -2.50 -5.12 F-Ser 354 3.46 2 -1.54 -3.97 F-Ser 151 F-Ser 196 2.80 3.19 3 +1.03 -3.40 F-Ser 354 P-Lys 305 3.09 3.88 4 +2.48 -0.80 F-Ser 151 F-Ser 196 P-Lys 305 3.35 3.11 3.51 N N H NH2 O CH3 O F N N NH O O CH3 N N O O CH3 F NH2 HF/pyridine (S)-alpha Fluoromethylhistidine x 2HCl HCl aqueous N N H NH2 OHO F HCl aqueous N N H NH2 OHO F NH4OH (S)-alpha Fluoromethylhistidine 1 2 3 4 5 Fluorination Remove Trityl Protecting Group Ester Hydrolysis