Você está na página 1de 21

European Journal of Medicinal Chemistry 115 (2016) 463e483

Contents lists available at ScienceDirect

European Journal of Medicinal Chemistry


journal homepage: http://www.elsevier.com/locate/ejmech

Research paper

Design, synthesis and biological evaluation of pyrido[2,3-d]pyrimidin-


7-(8H)-ones as HCV inhibitors

Marta Camarasa a, Raimon Puig de la Bellacasa a, Alex L. Gonzalez a, Raül Ondon~ o a,
Roger Estrada a, Sandra Franco b, Roger Badia b, Jose Este 
 b, Miguel Angel Martínez b,
a b  I. Borrell
 , Bonaventura Clotet , Jose a, *
Jordi Teixido
a
Grup d’Enginyeria Molecular, Institut Químic de Sarria, Universitat Ramon Llull, Via Augusta 390, E-08017 Barcelona, Spain
b noma de Barcelona, 08916 Badalona, Spain
Retrovirology Laboratory IrsiCaixa, Hospital Universitari Germans Trias i Pujol, Universitat Auto

a r t i c l e i n f o a b s t r a c t

Article history: The design and selection of a combinatorial library of pyrido[2,3-d]pyrimidin-7(8H)-ones (4) has allowed
Received 28 July 2015 the synthesis of 121 compounds, using known and new synthetic methodologies, and the evaluation of
Received in revised form the inhibitory activity against hepatitis C virus (HCV) genotype 1b replicon. Among these compounds, 21
3 March 2016
{4,10} and 24{2,10} presented very high activities [EC50 ¼ 0.027 mM (CC50 ¼ 5.3 mM) and EC50 ¼ 0.034 mM
Accepted 18 March 2016
Available online 21 March 2016
(CC50 ¼ 13.5 mM), respectively] and high selectivity indexes, 196 and 397. These values are similar to the
EC50 reported for sofosbuvir (2) (0.048 mM) using a similar methodological approach and the same virus
subtype. 21{4,10} and 24{2,10} are obtained through shorter synthetic itineraries than sofosbuvir and 24
Keywords:
Hepatitis C
{2,10} is achiral contrary to sofosbuvir which presents 4 stereogenic centers. In silico studies suggest that
HCV genotype 1b replicon 21{4,10} and 24{2,10} inhibits NS5B polymerase through allosteric site binding.
Small molecule inhibitor © 2016 Elsevier Masson SAS. All rights reserved.
Pyrido[2,3-d]pyrimidin-7-(8H)-ones

1. Introduction genotypes (1e7) with several subtypes within each genotype [2],
which differ by 30e35% of the nucleotide sites over the complete
Hepatitis C is an infectious disease caused by the hepatitis C genome. For instance, subtypes 1a and 1b are found worldwide and
virus (HCV), which constitutes the leading cause of chronic liver cause 60% of hepatitis C cases.
diseases. HCV has infected an estimated 150e200 million people HCV encodes structural (E1, E2, C and p7) and nonstructural
being an important health care problem worldwide [1]. The virus proteins (NS2, NS3/4A, NS4B, NS5A, NS5B) [3]. Nonstructural pro-
persists in the liver in about 85% of those infected. teins represent the main targets for intensive anti-HCV drug dis-
HCV belongs to the genus Hepacivirus, a member of the family covery programs.
Flaviviridae. HCV is a positive-sense single-stranded RNA virus of Before the development of direct acting antivirals (DAAs),
approximately 9.6 Kb. Based on genetic differences between HCV treatment for hepatitis C was based on a combination of (pegy-
isolates, the hepatitis C virus species is classified into seven lated) interferon-alpha (PEG-IFN-a) plus ribavirin (1) for 24 or 48
weeks, depending on the HCV genotype [4]. This combination
therapy yields a sustained virologic response (SVR) in more than
40% of patients infected by genotype 1 and 4 viruses and about
Abbreviations: BOP, (Benzotriazol-1-yloxy)tris(dimethylamino)phosphonium
hexafluorophosphate; m-CPBA, 3-Chloroperbenzoic acid; DBU, 1,8-Diazabicyclo 80% of those infected by genotypes 2 and 3. Besides the limited
[5.4.0]undec-7-ene; DDQ, 2,3-Dichloro-5,6-dicyano-p-benzoquinone; PSII, Palm efficacy on HCV type 1, this combination therapy has significant
site II of NS5B polymerase; PyBOP, (Benzotriazol-1-yloxy)tripyrrolidinophospho- side effects and is poorly tolerated in many patients. The most
nium hexafluorophosphate.
common side effects that may occur after treatment with PEG-
* Corresponding author.
E-mail addresses: martacamarasan@gmail.com (M. Camarasa), raimonpc@gmail.
IFN-a are: nausea, diarrhea, fever, chills, muscle and joint pain,
com (R. Puig de la Bellacasa), alejandro.lopez@iqs.url.edu (A.L.  Gonza lez), difficulty concentrating, thyroid disorders, hair loss, insomnia, ir-
raulondonom@iqs.url.edu (R. Ondon ~ o), roger.estrada@iqs.url.edu (R. Estrada), ritability, mild to severe depression, and rarely, suicidal thoughts.
sfranco@irsicaixa.es (S. Franco), rbadia@irsicaixa.es (R. Badia), jaeste@irsicaixa.es Other serious side effects that may occur are bone marrow
(J. Este ), MMartinez@irsicaixa.es (M.A.  Martínez), jordi.teixido@iqs.url.edu
), bclotet@irsicaixa.es (B. Clotet), j.i.borrell@iqs.url.edu (J.I. Borrell).
toxicity, cardiovascular disorders, hypersensitivity disorders,
(J. Teixido

http://dx.doi.org/10.1016/j.ejmech.2016.03.055
0223-5234/© 2016 Elsevier Masson SAS. All rights reserved.
464 M. Camarasa et al. / European Journal of Medicinal Chemistry 115 (2016) 463e483

a non-nucleotide inhibitor. As starting point we consider a series of


patents of Gilead Sciences on the use of pyrido[3,2-d]pyrimidines
(3) as HCV inhibitors (see for instance [9]) due to the structural
similarity between such compounds and the 7-oxopyrido[2,3-d]
pyrimidines (4) developed by our group.
In this context, our group has a broad experience in the syn-
thesis of 4-amino-5,6-dihydropyrido[2,3-d]pyrimidin-7-(8H)-ones
(9) (Scheme 1) from a,b-unsaturated esters (5). Thus, 2-methoxy-6-
oxo-1,4,5,6-tetrahydropyridin-3-carbonitriles (7) are obtained by
reaction of an a,b-unsaturated ester (5) and malononitrile (6) in
NaOMe/MeOH [10]. Treatment of pyridones 7 with guanidine sys-
tems (8, R4 ¼ H, alkyl, aryl) affords 4-amino-pyrido[2,3-d]pyrimi-
dines (9, R4 ¼ H, alkyl, aryl) [11].
More recently, we have developed protocols for the dehydro-
Fig. 1. Ribavirin (1), sofosbuvir (2), pyrido[3,2-d]pyrimidines active against HCV (3), genation of 5,6-dihydropyrido[2,3-d]pyrimidin-7(8H)-ones 12 to
and pyrido[2,3-d]pyrimidin-7-(8H)-ones (4). pyrido[2,3-d]pyrimidin-7(8H)-ones 11 [12] and a methodology for
the synthesis of 2-arylamino substituted 4-amino-5,6-
dihydropyrido[2,3-d]pyrimidin-7(8H)-ones (9, R4 ¼ aryl) via the
endocrine disorders, pulmonary disorders, colitis, pancreatitis and corresponding 3-aryl substituted pyridopyrimidines 10, formed
ophthalmologic disorders. PEG-IFN-a can cause neuropsychiatric, upon treatment of pyridones 7 with an arylsubstituted guanidine 8
autoimmune, ischemic and infectious fatal. With respect to riba- in dioxane, which underwent the Dimroth rearrangement to the
virin, anemia is often the most frequent side effect during the desired 4-aminopyridopyrimidines (9, R4 ¼ aryl) with NaOMe/
treatment of hepatitis C. Ribavirin can also cause decreased white MeOH. The overall yields of such two-step protocol are in general
blood cells and platelets [5]. higher than the reaction between pyridones 7 and an arylsub-
In 2011, the first two DAAs (boceprevir and telaprevir) that stituted guanidine 8 (Scheme 1) [13].
targeted NS3/4A protease viral protein were introduced to the The present paper deals with the use of such methodology for
market. Added to PEG-IFN-a and ribavirin increased the SVR in the synthesis of pyrido[2,3-d]pyrimidin-7-(8H)-ones as a potential
genotype infected patients. However, several disadvantages were treatment of HCV.
reported [6]. Thus, further pursuit of this strategy as a potential
treatment was warranted. More recently, sofosbuvir (2, Sovaldi™)
2. Results and discussion
[7], a nucleotide analog, has been approved for the treatment of
HCV infection. Treatment with this new drug without PEG-IFN-a
2.1. Virtual combinatorial library of pyrido[2,3-d]pyrimidin-7-
provides higher cure rates, less side effects, and a reduction of the
(8H)-ones
duration of the therapy. Sofosbuvir inhibits the NS5B polymerase,
one of the most attractive targets of HCV because it presents a
Inhibitory activity of several privileged scaffolds has been
highly conserved region in the genome with respect to the different
intensively researched as potential anti-HCV candidates. Among
genotypes [8]. Sofosbuvir has changed the prognosis of this severe
them, Gilead Sciences reported a large series of pyrido[3,2-d]py-
disease, however the price associated with the treatment (in the
rimidines with promising activities but the main HCV target of
range 20,000e40,000 V per one year) has engendered considerable
these molecules remains unclear [9]. A chemical and structural
controversy including patent invalidity claims (Fig. 1).
similarity search of the crystallized targets, using a subset of pyrido
We consider that this scenario shows that there is still a need for
[3,2-d]pyrimidines as query, yielded similarity scores higher than
clinically effective agents for the treatment of infections caused the
0.7 for molecules targeting NS5B polymerase (see Methods for
hepatitis C virus (HCV), which may improve current therapeutic
details), thus we hypothesize that this scaffold could potentially
strategies, in particular, which may reduce unwanted side effects of
target this HCV enzyme. Nevertheless, in view of the potency of
current drugs.
pyrido[3,2-d]pyrimidines and own expertise complemented by
We decided to focus on the NS5B polymerase in order to develop
computer-assisted rational design, we chose to evaluate the activity

Scheme 1. Synthesis of 7-oxopyrido[2,3-d]pyrimidines 9, 10, and 11 from a,b-unsaturated esters 5 via pyridones 7.
M. Camarasa et al. / European Journal of Medicinal Chemistry 115 (2016) 463e483 465

of a combinatorial library of pyrido[2,3-d]pyrimidin-7-(8H)-ones


for optimization.
In order to construct our virtual combinatorial library of pyrido
[2,3-d]pyrimidin-7-(8H)-ones (4), we decided to take into account
the structural information contained in a series of patents of Gilead
Sciences on the use of pyrido[3,2-d]pyrimidines (3) as HCV in-
hibitors (WO2008/077651, WO2008/077650, WO2010/002998,
WO2008/077649, and WO2006/120252). The large number of
compounds included in such patents (800) led us to reduce them
on the basis of the values of the EC50 (effective concentration
required to inhibit 50% of replicating cells containing the HCV
replicon) and cytotoxic CC50 (concentration needed to induce cell
death in 50% of treated cells). It should be noted that these patents
do not give the exact value of the EC50 or the CC50 but only classified
compounds in three different categories for each parameter: EC50
(<0.1 mM; between 0.1 and 0.25 mM; and between 0.25 and 2 mM)
and CC50 (>25 mM; between 5 and 25 mM; and <5 mM). Conse-
quently, we decided to select molecules with EC50 < 0.1 mM and,
simultaneously, CC50 > 25 mM. This assumption drastically reduced
the number of molecules to be considered from the initial 800 to
236 from which we studied the nature of the substituents R1, R2 and
R3 present in structures 3. Thus, we analyzed the most repeated
substituents present at positions R1, R2 and R3 of these compounds
and we classified them according to frequency of occurrence from
major to minor (see supplementary information).
This protocol allowed us to select 31 potential substituents at R1,
32 at R2 and 89 at R3. The use of this set of substituents in our pyrido
[2,3-d]pyrimidin-7-(8H)-ones (4) afforded a potential virtual
combinatorial library of 31  32  89 ¼ 88,288 compounds, which Fig. 2. a,b-Unsaturated esters 5{x} used for library production.
could be even higher if we considered different substituents at R4
(H, Me, Et, CH2C6H5, ..).
Given the overall size of the potential library, a diverse selection complete in the case of PyBOP. In that way we obtained 14{2}, 14
of 200 compounds 4 was carried out based on the R1, R2 and R3 {4}, and 14{5} in 74e93% yield.
residues with higher frequencies of occurrence (including substi- Once obtained the intermediates 14{x}, we started to study the
tution at R4) and the commercial availability of the necessary substitution by different amines and alcohols of general structure
building blocks. Similarity scores of these molecules ranged be- R2-H and we realized that the direct substitution of the benzo-
tween 0.8 and 1.2 in a scale of 0e2, which implies high similarity of triazol-1-yloxy group was only possible with secondary amines
pyrido[2,3-d]pyrimidin-7-(8H)-ones compared to those forming a (such as pyrrolidine) or alcohols (in the presence of Cs2CO3). Using
complex with NS5B (see Supporting Information). The resulting this later protocol we obtained the corresponding 4-methoxy
sublibrary was synthesized and submitted to biological evaluation substituted (15{2,1} and 15{5,1}), 4-ethoxy substituted (15{2,2}),
(see Fig. 1). 4-(2-methoxyethoxy) substituted (15{5,3}), and 4-(2-
propoxyethoxy) substituted (15{5,4}) pyridopyrimidines.
2.2. Chemistry Consequently, for the synthesis of the 4-amino substituted
pyridopyrimidines 15{x,y} it was necessary to modify our approach
In order to obtain the selected sublibrary, including those pyrido and to react the starting 4-oxopyrido[2,3-d]pyrimidine 13{x} with
[2,3-d]pyrimidin-7-(8H)-ones (4) with complex disubstitution on the corresponding amine, BOP and DBU in anhydrous acetonitrile
positions C2 and C4 of the pyrimidine ring, we developed a new heated at 90  C for 2 days in a one-pot process. In that way we
synthetic strategy for the construction of the pyridopyrimidine obtained the corresponding 4-pentylamino substituted (15{2,5}
nucleus [14]. Thus, the treatment of 2-aryl-substituted acrylates 5 and 15{5,5}), 4-(2-propoxyethylamino) substituted (15{2,6} and 15
{1e10} (Fig. 2) with 2,6-diaminopyrimidine-4(3H)-one (12) with 1 {5,6}), and 4-pyrrolidino substituted (15{2,7} and 15{5,7}) pyr-
equivalent of sodium methoxide in ethylene glycol at 180  C for 3 h idopyrimidines in 40e78% yield. Nevertheless, in some cases it was
under microwave irradiation yielded the corresponding 2-amino- necessary to carry out the substitution on the corresponding ben-
6-aryl-5,6-dihydropyrido[2,3-d]pyrimidine-4,7(3H,8H)-diones (13 zotriazol-1-yloxy substituted intermediate 14{x}. Thus we obtained
{1e10}) in 68e94% yield (Scheme 2). the 4-(2-propoxyethylamino) derivative 15{4,6} and the 4-
For the introduction of an amino or alkoxy group at position benzylamino substituted compound 15{5,8}. Although we tried to
C4 of 4-oxopyrido [2,3-d]pyrimidines 13{1e10}, we activated the use these protocols for the substitution of the benzotriazol-1-yloxy
C4 carbonyl group for nucleophilic displacement upon treatment group by ammonia to introduce a 4-amino group, the reaction did
with BOP or PyBOP [15e17]. This strategy involves the synthesis not proceed in any case. Consequently, the synthesis of 2,4-
of a benzotriazol-1-yloxy substituted intermediate 14{x}, which diaminopyrido[2,3-d]pyrimidines must be carried out using the
is a good leaving group for a nucleophilic substitution with an procedures previously described by our group (see Scheme 1).
amine or alcohol to yield the corresponding 4-amino or 4-alkoxy However, we explored the reaction between the methyl 2-
derivative 15{x,y} (Scheme 2). After several trials we found that arylacrylate 5{2}, sodium methoxide and 2,6-diaminopyrimidin-
the treatment of the corresponding 13{x} with BOP and DBU in 4(3H)-one in anhydrous ethylene glycol under microwave irradia-
anhydrous acetonitrile at rt for 2 days under nitrogen atmo- tion and we obtained the 2,4-diaminopyridopyrimidine 15{2,4} in
sphere afforded the corresponding 14{x}, the reaction not being
466 M. Camarasa et al. / European Journal of Medicinal Chemistry 115 (2016) 463e483

Scheme 2. Synthetic methodologies used for the preparation of the pyrido[2,3-d]pyrimidin-7-(8H)-ones included in the tested sublibrary tested against HCV.

26% yield. derivatives 20{x} in 60e74% yield. Using such protocol we obtained
We studied several methodologies for the dehydrogenation of compounds 20{1}, 20{2}, 20{4}, 20{5}, 20{8}, and 20{11}.
5,6-dihydropyrido[2,3-d]pyrimidine-7(8H)-ones to the corre- The subsequent treatment of compounds 20{x} with the corre-
sponding aromatic compounds [12], however we have not found a sponding anilines afforded compounds 21{1,10}, 21{2,10}-21{2,13},
general procedure and the reaction conditions and the oxidizing 21{4,10}, and 21{8,10} in 67e94% yield.
agent strongly depend on the structure of the starting pyr- Similarly, the treatment of 20{2} with sodium phenoxide in 2-
idopyrimidine. In this work we used three different reaction propanol afforded the 2-phenoxy substituted pyridopyrimidine
conditions: (a) NaH in DMSO at 100  C [12]; (b) DDQ in 1,2- 21{2,14} in 72% yield.
dichloroethane at 100  C; (c) activated MnO2 in AcOH at reflux. Once obtained the 2-substituted pyridopyrimidines 21{x,y}, we
Using the adequate protocol we obtained compounds 16{2}, 16 used the same protocol described above for the introduction of a
{2,5}, 16{2,6}, 16{2,7}, 16{4,6}, 16{5,9}, 16{5,5}, and 16{5,6} in second substituent at C4. Thus, we treated compounds 21{x,y} with
50e90% yield. BOP and DBU in anhydrous acetonitrile to afford the corresponding
The aromatic compounds 16{x,y} were finally methylated on the 4-benzotriazol-1-yloxy substituted intermediate 22{x,y}. In that
lactam nitrogen using MeI in NaH/DMSO at rt to afford the methyl way we obtained 22{2,10}, 22{2,14}, and 22{11,10} in 66e88% yield.
derivatives 17{x,y}. Thus we obtained compounds 17{2,1}, 17{2,5}, Subsequently, we substituted the 4-benzotriazol-1-yloxy group
17{2,6}, 17{2,7}, and 17{5,3} in 26e89% yield. of compounds 22{x,y} by amines also following the protocol pre-
On the other hand, the synthesis of pyridopyrimidines 23{x,y,z} viously described to afford pyridopyrimidines 23{x,y,z} bearing
with a double complex substitution at C2 and C4 of the pyrimidine substituents at C2 and C4 (23{2,10,6} and 23{2,14,6} were obtained
ring also needed the development of a new synthetic strategy in 64% and 54% yield, respectively).
(Scheme 2). Thus, the reaction between 2-aryl-substituted acry- Then, compounds 23{x,y,z} were aromatized to the corre-
lates 5{1e10} (Fig. 2) and 6-amino-2-(methylthio)pyrimidin-4(3H)- sponding 24{x,y,z} using the dehydrogenation protocols described
one (18) with K2CO3 as base in 2-propanol at 170  C for 3 h under above. Thus, 24{2,10,6} and 24{2,14,6} were obtained in 78% and
microwave irradiation yielded the corresponding 2-(methylthio)- 75% yield, respectively.
6-aryl-5,6-dihydropyrido[2,3-d]pyrimidine-4,7(3H,8H)-diones (19 To complete the sublibrary, we used the protocol for the syn-
{1e5} and 19{7e11}) in 56e88% yield (Scheme 2) [14]. thesis of 4-amino-2-arylamino substituted pyridopyrimidines 9
The direct substitution of the methylthio group of compounds described in Scheme 1 [13] to obtain compounds 9{1}, 9{4}, 9{5}, 9
19{x} was possible with nucleophilic amines, such as benzylamine {6}, and 9{7} in 32e74% yield from pyridines 7{x} via intermediates
which afforded in 2-propanol under microwave irradiation the 10{x} [13,19], which were subsequently dehydrogenated to 11{1}, 11
corresponding 2-benzylamino derivatives 21{2,8} and 21{5,8} in {4}, 11{5}, 11{6}, and 11{7} in 59e95% yield [19].
53% and 36% respectively from 19{2} and 19{8}, respectively. These later compounds were alkylated on the lactam nitrogen
Similarly, the 2-(4-fluorobenzyl) amino derivative 21{2,9} in 40% with different C1eC6 linear and branched alkyl chains to afford
yield. compounds 25{x,y,z,k}. In that way we obtained compounds 25
However, our main interest was the introduction of arylamino {4,10,4,1} [19], 25{5,10,4,5}, 25{5,12,4,1}, 25{5,10,4,1}, 25{6,10,4,1}
substituents at position C2 of the pyrimidine ring. Consequently, [19], 25{1,10,4,1} [19], 25{5,10,4,6}, 25{5,10,4,3}, 25{5,10,4,7}, 25
we decided to convert the methylthio group present in compounds {7,10,4,1} [19], 25{5,10,4,2}, and 25{5,10,4,4} included in Table 1,
19{x} into a methylsulfonyl group to favor the nucleophilic among others.
displacement. According to the literature we tested several Finally, taking into account that all the members of this sub-
oxidizing agents (H2O2/AcOH, H2O2/NbC/EtOH [18], H2O2/EtOH, library present a substituent at position C4 of the pyrimidine ring
carbamide peroxide/MeOH, oxone® (potassium perox- (oxo, amino, alkylamino, alkoxy), we decided to include three
ymonosulfate)/MeOH/H2O) but the conversion was never com- unsubstituted compounds for comparison purposes. Thus, starting
plete, affording mixtures of the desired sulfone 20{x} and the from compound 26 [20] we obtained 27 and 28 in 87 and 96% yield,
corresponding sulfoxide. respectively (Fig. 3).
Finally, the treatment of compounds 19{x} with m-CPBA in DMF
at 0  C afforded the corresponding methylsulfonyl substituted
M. Camarasa et al. / European Journal of Medicinal Chemistry 115 (2016) 463e483 467

Table 1
EC50 and CC50 of pyrido[2,3-d]pyrimidines against LucUbiNeo-ET cell line and Huh-7/Lunet cell line, respectively.

Entry Code Structure EC50 (mg/ml) EC50 (mM) CC50 (mM) CC50 (mg/ml)

1 21{4,10} 0.010 0.027 5.3 1.95

2 24{2,10} 0.012 0.034 13.5 4.71

3 25{4,10,4,1} [19] 0.06 0.16 10.4 3.94

4 25{5,10,4,5} 0.1 0.22 184.9 84

5 25{5,12,4,1} [25] 0.11 0.22 3.2 1.58

6 28 0.09 0.23 0.9 0.34

7 25{5,10,4,1} [25] 0.1 0.24 0.5 0.22

8 25{6,10,4,1} [19] 0.15 0.30 0.8 0.42

9 11{1} [19] 0.16 0.49 13.8 4.54

10 25{1,10,4,1} [19] 0.30 0.90 23.5 8.07

11 29 [25] 0.46 0.90 2.6 1.32

12 25{5,10,4,6} 0.50 0.99 240.9 109.9

13 22{2,10} 0.60 1.28 12.2 5.68

14 22{11,10} 0.70 1.35 5.5 2.83

15 25{5,10,4,3} 0.62 1.4 181.1 79.8

16 16{2,6} 0.74 2.1 129.9 46.44

(continued on next page)


468 M. Camarasa et al. / European Journal of Medicinal Chemistry 115 (2016) 463e483

Table 1 (continued )

Entry Code Structure EC50 (mg/ml) EC50 (mM) CC50 (mM) CC50 (mg/ml)

17 25{5,10,4,7} 1.00 2.3 29.3 13

18 25{7,10,4,1} [19] 1.22 2.8 6.3 2.7

19 16{5,5} 1.24 3.2 11.0 4.32

20 24{2,14,6} 1.43 3.3 79.8 24.3

21 17{2,6} 1.75 4.7 54.3 20.16

22 25{5,10,6,1} 2.54 5.1 84.7 42.22

23 25{5,10,4,2} 2.70 6.3 >125

24 11{6} [19] 3.77 7.7 166 81.02

25 24{2,13} 3.55 9.4 46 17.24

26 21{5,8} 3.98 9.6 69 28.5

27 17{5,9} 4.39 10.4 50.1 21.16

28 17{2,7} 3.57 10.5 111.4 37.81

29 30 [14] 3.38 11.2 38.3 11.54

30 25{5,10,4,4} 5.41 11.9 e >125

31 23{5,10,6} 6.01 12.4 50.5 24.55

32 17{5,4} 5.6 16.7 120.2 40.4

33 11{4} [19] 6.39 17.5 2.4 0.89


M. Camarasa et al. / European Journal of Medicinal Chemistry 115 (2016) 463e483 469

Table 1 (continued )

Entry Code Structure EC50 (mg/ml) EC50 (mM) CC50 (mM) CC50 (mg/ml)

34 22{2,14} 8.72 18.6 62.7 3.40

35 17{2,5} 7.44 20.9 79.8 28.37

36 15{5,3} 8.45 22.0 65.0 24.91

[25] and 30 [14]). The 36 compounds with the lower EC50 are
included in Table 1.
The results contained in Table 1 show that there are 12 com-
pounds with an EC50 < 1 mM, 9 compounds in the range
1 < EC50 < 5 mM, and 15 compounds in the range 5 < EC50 < 22 mM.
If we focus on the first group of compounds, there are two
compounds with very low EC50 and good CC50: 21{4,10} with
EC50 ¼ 0.027 mM (CC50 ¼ 5.3 mM) and 24{2,10} with
EC50 ¼ 0.034 mM (CC50 ¼ 13.5 mM), respectively. Moreover, it is
interesting to note what their selectivity index or therapeutic index,
which is the ratio CC50/EC50 and gives an idea safety of these po-
Fig. 3. 4-Unsubstituted pyrido-[2,3-d]pyrimidines included in the sublibrary tested tential drugs, are 196 and 397.
against HCV.
Furthermore, the EC50 of 21{4,10} and 24{2,10} are in the same
range of the EC50 of the HCV NS3 protease inhibitors Inh I and Inh II
2.3. Biological evaluation of the HCV antiviral activity used as internal controls and, more importantly, are of the same
order of magnitude of the EC50 of sofosbuvir in a similar HCV 1b
Once the library of pyrido[2,3-d]pyrimidines was synthesized, replicon test (0.048 mM).
we evaluated the HCV antiviral activity and the toxicity of such In order to try to locate the target of our antiviral compounds,
compounds. Consequently, we determined the EC50 and the CC50. we carried out an HCV NS5B drug resistance assay without
For testing the EC50 (mg/mL), we used a stable cell line, concluding results. This experiment consisted of trying to isolate
LucUbiNeo-ET, that contains a subgenomic HCV genotype 1b replicons resistant to our most potent compounds propagating the
replicon, I389/NS3-3’/LucUbiNeo-ET, that express luciferase replicon at concentrations equivalent to the EC90. We obtained
constitutively [21,22]. resistant populations (EC50 between 32.7 and 63.7 mg/mL) but
To evaluate the cellular toxicity (CC50, mg/mL), we used another when we sequenced them, we did not found mutations clearly
cell line, Huh-7/Lunet [23], that originally carried a selectable associated with non-nucleotide inhibitors of the NS5B polymerase.
luciferase HCV 1b replicon and was cured by treatment with an
HCV-specific inhibitor.
2.4. Molecular docking study of 21{4,10} and 24{2,10} on NS5B
As positive controls for these assays we used two commercially
polymerase
available HCV NS3 protease inhibitors Inh I and Inh II (Fig. 4) with
EC50 ¼ 0.0608 mM (CC50 > 2.0 mM) and EC50 ¼ 0.0032 mM
As mentioned above, we hypothesized that NS5B polymerase
(CC50 > 2.0 mM), respectively [24].
should be a feasible target for substituted pyrido[2,3-d]pyrimidin-
A total of 121 compounds were evaluated, including those
7-(8H)-ones due to the high similarity scores of these molecules
described in the present paper and pyrido[2,3-d]pyrimidines
with the available NS5B-ligand X-ray complexes. NS5B is one of the
coming from other research programs within our group (such as 29
main targets for HCV replication inhibition, and several clinical
reports increased the attractiveness of using this enzyme as a
therapeutic target [26e39]. NS5B inhibitors can be classified in two
major groups: active site nucleotide/nucleoside analogues (Nucs)
and nonnucleoside inhibitors (NNIs) that bind to specific allosteric
sites on the enzyme. The latter, achieve inhibition by inducing
conformational changes into the protein. Several NS5B e inhibitor
complex structures have been characterized, which leads to an
attractive NNIs database for screening and comparison
[29e31,36,37]. These crystal structures revealed at least four
pockets as potential allosteric inhibitory sites, each of which is
characterized by specific contacts and hydrogen bonding patterns
that depend on the scaffold derivatization. Sites I and II occur in the
Fig. 4. Commercially available HCV NS3 protease inhibitors Inh I and Inh II used as thumb domain (TSI and TSII respectively), while sites III and IV can
positive controls for the assays. be found in the palm domain (also known as PSI and PSII
470 M. Camarasa et al. / European Journal of Medicinal Chemistry 115 (2016) 463e483

respectively). 21{4,10} optimize the intermolecular contacts and reduces the


In view of the promising anti-HCV activity of our molecules, we sterical hindrance of the pyrido[2,3-d]pyrimidin-7-(8H)-one sub-
rationalized its potency through in silico approaches. Despite a stituents (Fig. 6). However, the core positioning is inverted so the
wealth of structural information of NS5B complexes we could not difluoro phenyl ring points towards the NS5B active site and locates
assure the binding domain of pyrido[2,3-d]pyrimidin-7-(8H)-ones at the initiation of the primer strand. A recent structure of NS5B
by the previous similarity strategy. Thus, we proceeded with a cocrystallized with sofosbuvir diphosphate and viral RNA demon-
cross-docking protocol consisting in the systematical docking of strates the conformational adaptions of the enzyme during RNA
ligands into the different allosteric binding sites of NS5B. The ob- replication [40]. Intriguingly, superposition of the molecular
tained conformations were then refined allowing the enzyme to docking complexes and NS5B-RNA structure suggests that both 21
accommodate the small molecules through an induced-fit method. {4,10} and 24{2,10} can potentially interact with riboses of the
Our findings suggest that compounds 21{4,10} and 24{2,10} bind primer strand to disrupt polymerization (Fig. 6).
to NS5B PSII with affinities close to the reference compounds.
Indeed, a high structural similarity should imply a similar binding 3. Conclusions
mode to the active site. For instance, 24{2,10} binds to PSII repro-
ducing a similar pose to (4-((N-(5-cyclopropyl-2-(4-fluorophenyl)- Using the structural information contained in a series of patents
3-(methylcarbamoyl)benzofuran-6-yl)methylsulfonamido) of Gilead Sciences on the use of pyrido[3,2-d]pyrimidines (3) as
methyl)-2-fluorophenyl)boronic acid (PDB ID 4KE5) [37]. The py- HCV inhibitors, we designed a library of pyrido[2,3-d]pyrimidin-
rimidine core forms a cation-p stacking interaction with the 7(8H)-ones (4) including more than 89,000 compounds. After a
conserved R200 residue. At the same time, the fluorophenyl ring diversity selection, we synthesized 121 compounds using the
occupies a hydrophobic pocket of the palm cleft with an excellent synthetic methodologies contained in Scheme 2. In addition, we
superposition to the reference (Fig. 5). However, the lack of water determined the EC50 (mg/mL) using a stable cell line containing a
bridges and the lower number of hydrogen-bond donor and ac- subgenomic HCV subtype 1b replicon. Moreover, we also deter-
ceptors provided by 24{2,10} should reduce the potency of this mined the cellular toxicity (CC50, mg/mL) using a cell line that
compound. Previous reports hypothesized that potent PSII in- originally carried a selectable luciferase HCV 1b replicon and was
hibitors blocked polymerization by inactivating the b-hairpin flap cured by treatment with an HCV-specific inhibitor.
though hydrogen bond binding to Y448. This tyrosine is conserved Among these compounds, 21{4,10} and 24{2,10} presented very
through all the genotypes, located at the tip of the b-flap comprised high activities [EC50 ¼ 0.027 mM (CC50 ¼ 5.3 mM) and
of F193, P197 and M414, which can form a hydrogen-bond either EC50 ¼ 0.034 mM (CC50 ¼ 13.5 mM), respectively] and high selec-
with K200 or potential PSII inhibitors. Unfortunately, our com- tivity indexes, 196 and 397.
pound is not able to form hydrogen bonding with Y448 due to The EC50 of 21{4,10} and 24{2,10} are of the same order of
unfavorable positioning of the NHeC6H5 group. These observations magnitude of the EC50 of sofosbuvir in a similar HCV 1b replicon
are consistent with the 20-fold increase relative to EC50 of our test (0.048 mM) [41]. It is interesting to note that sofosbuvir, a
compound for the 1b replicon, if compared to the crystal structure compound containing 4 stereogenic centers, is synthesized through
ligand. a long multistep synthesis [42] while 21{4,10} presents only one
More interestingly, we found that compound 21{4,10} yields a stereogenic center and is obtained in three steps from commer-
highest affinity complex which is in accordance with the biological cially available starting materials. Similarly, the achiral 24{2,10} is
activity observed. Comparing both structures it becomes clear that obtained in four steps also from commercially available starting

Fig. 5. Representation of 24{2,10} binding to the PSII of NS5B. (A) Superposition of compound with the reference crystal structure (PDB ID 4KE5). 24{2,10} and the crystal structure
ligand are depicted as orange and green sticks respectively. (B) Positioning of 24{2,10} into the PSII pocket. (C) Relative positioning of the compound into the b-flap pocket. Non-
polar hydrogens were removed for clarity purposes in all the representations.
M. Camarasa et al. / European Journal of Medicinal Chemistry 115 (2016) 463e483 471

4. Experimental

4.1. Synthesis and characterization of compounds

4.1.1. General
All solvents and chemicals were reagent grade. Unless otherwise
mentioned, all solvents and chemicals were purchased from com-
mercial vendors (Fluka, Aldrich, ABCR and ACROS Organics) and
used without purification. Compounds 13{1e10}, 19{1e5}, and 19
{7e10} were prepared as previously described [14].1H and 13C NMR
spectra were recorded on a Varian 400-MR spectrometer that was
operating at a field strength of 400 and 100.6 MHz, respectively.
Chemical shifts were reported in parts per million (d) and coupling
constants (J) were in Hz by using, in the case of 1H NMR spectros-
copy, tetramethylsilane (TMS) as an internal standard, and in the
case of 13C NMR spectroscopy, solvent residual peak was taken as
reference: CDCl3 at 77.0 ppm, DMSO-d6 at 39.5 ppm. Standard and
peak multiplicities are designed as follows: s, singlet; d, doublet;
dd, doublet of doublets; dt, doublet of triplets t, triplet; br, broad
signal. IR spectra were recorded in a Thermo Scientific Nicolet iS10
FTIR spectrophotometer with Smart iTr. Wavenumbers (n) are
expressed in cm1. MS data (m/z (%), EI, 70 eV) were obtained by
using an Agilent Technologies 5975 spectrometer and a Hewlett
Packard HP5988A quadrupole mass spectrometer operating in
electronic ionization (EI) mode at 70 eV and at 4 kV accelerating
potential, or a Bruker Biotoff II spectrometer operating in electro-
spray ionization (ESI) mode with a Time of Flight (TOF) detector.
HRMS data were obtained by using a VG AutoSpec (Micromass
Instruments) Trisector EBE high resolution spectrometer (EI or FAB
mode), a Bruker Biotof II mass spectrometer (ESI TOF mode).
Elemental microanalyses were obtained on a EuroVector In-
struments Euro EA elemental analyzer. The melting points were
determined with a Büchi-Tottoli 530 capillary apparatus and are
uncorrected. Automatic flash chromatography was performed in an
Isco Combiflash medium pressure liquid chromatograph with
RediSep® silica gel columns (35e70 mm) using a suitable mixture of
solvents as eluent. Microwave irradiation experiments were carried
out in an Initiator™ (Biotage) microwave apparatus, operating at a
frequency of 2.45 GHz with continuous irradiation power from 0 to
Fig. 6. (A) Superposition of 24{2,10} (orange) and 21{4,10} (green) in the PSII binding
400 W. Reactions were carried out in 0.5, 2.5, 5, and 20 mL glass
site. (B) Representation of a potential interaction of 21{4,10} with RNA and a cocrys-
tallized sofosbuvir diphosphate unit. tubes, sealed with aluminum/Teflon crimp tops, which can be
exposed up to 250  C and 20 bar internal pressure. Temperature
was measured with an IR sensor on the outer surface of the process
vial. After the irradiation period, the reaction vessel was cooled
materials.
rapidly to 50  C by air jet cooling.
Although it has not been possible yet to stablish the molecular
target for these compounds, chemoinformatic studies suggest a
4.1.2. General procedure for the synthesis of 4-((1H-benzo[d][1,2,3]
high degree of similarity between 21{4,10} and 24{2,10} and PSII
triazol-1-yl)oxy)-6-aryl-3,4,5,6-tetrahydropyrido[2,3-d]pyrimidine-
inhibitors of NS5B polymerase. Further cross docking analysis
7(8H)-ones (14{x})
shows an excellent binding of our compounds with the hydro-
The corresponding 2-amino-6-aryl-5,6-dihydropyrido[2,3-d]
phobic pocket of the palm cleft, as it has been described in other
pyrimidine-4,7(3H,8H)-diones (13{x}) (0.50 mmol) [14] was sus-
structural studies with known PSII inhibitors. An optimal cation-p
pended in 13 mL of anhydrous acetonitrile. Then, BOP (0.75 mmol)
stacking of the pyrido[2,3-d]pyrimidine core of 21{4,10} and 24
and DBU (1.25 mmol) were added into the solution. The system was
{2,10}, combined with the optimal positioning of the 2,6-
stirred at room temperature for 2 days under nitrogen atmosphere.
difluorophenyl and the p-fluorophenyl substituents respectively,
After this period, 200 mL of water were added and the resulting
seems to be compatible with the interaction of these compounds
precipitate was filtered, washed with water and dried in vacuo over
with RNA riboses of the primer strand which could disrupt poly-
phosphorus pentoxide to afford the corresponding 14{x} that can
merization, in good agreement with the observed biological
be used without further purification.
activity.
Taken together, these results point out to achiral compound 24
4.1.2.1. 4-((1H-benzo[d][1,2,3]triazol-1-yl)oxy)-2-amino-6-(4-
{2,10} as a promising candidate to be further developed as drug for
fluorophenyl)-3,4,5,6-tetrahydropyrido[2,3-d]pyrimidine-7(8H)-one
the treatment of hepatitis C. Further studies should be conducted to
(14{2}). Starting from compound 13{2} to afford 14{2} in 74% yield,
validate such hypothesis.
white solid. IR (KBr), nmax (cm1): 3425, 3337, 3223, 1637, 1575,
1510, 1356, 1237, 1160, 1088, 847, 738. 1H NMR (400 MHz, DMSO-
d6): d 10.95 (s, 1H), 8.13 (dt, J ¼ 8.4, 0.9 Hz, 1H), 7.70e7.61 (m, 2H),
472 M. Camarasa et al. / European Journal of Medicinal Chemistry 115 (2016) 463e483

7.51 (ddd, J ¼ 8.1, 6.4, 1.6 Hz), 7.39 (dd, J ¼ 8.7, 5.6 Hz, 2H), 7.21 (t, 4 .1. 3 . 2 . 2 - A m i n o - 6 - ( 2 , 6 - d i ch l o ro p h e nyl ) - 4 - m e t h o x y- 5 , 6 -
J ¼ 8.9 Hz, 1H), 6.59 (s, 2H, NH2), 4.11 (dd, J ¼ 10.7, 6.9 Hz, 1H), dihydropyrido[2,3-d]pyrimidine-7(8H)-one (15{5,1}). Starting from
3.29e3.12 (m, 2H). 13C NMR (100.6 MHz, DMSO-d6): d 172.4, 166.0, compound 14{5} and methanol to afford 15{5,1} in 96% yield, white
161.8, 161.0, 143.2, 135.4, 130.8 (d, J ¼ 7.9 Hz, C12), 129.5 (C17), 128.9, solid. IR (KBr), nmax (cm1): 3438, 3330, 3222, 2951, 2891, 1688,
125.6 (C18), 120.3 (C19), 115.6 (d, J ¼ 21.1 Hz, C13), 110.0, 109.8 (C16), 1634, 1577, 1488, 1466, 1371, 1257, 1196, 774. 1H NMR (400 MHz,
86.0 (C5), 45.4 (C7), 24.5 (C6). TFA-d): d 7.37 (dd, J ¼ 15.0, 8.1 Hz, 2H), 7.24 (t, J ¼ 8.2 Hz, 1H), 5.08
(dd, J ¼ 13.3, 9.4 Hz, 1H), 4.11 (s, 3H), 3.30e3.08 (m, 2H). 13C NMR
(100.6 MHz, TFA-d): d 173.9, 170.8, 154.3, 146.6, 136.0, 134.7, 131.7,
4.1.2.2. 4-((1H-benzo[d][1,2,3]triazol-1-yl)oxy)-2-amino-6-(2,6-
130.2, 129.4, 128.4, 90.0, 55.7, 42.6, 21.0. MS (70 eV, EI): m/z
difluorophenyl)-5,6-dihydropyrido[2,3-d]pyrimidin-7(8H)-one (14
(%) ¼ 338.1 (60) [M]þ, 303.1 (100) [M-Cl]þ, 153.0 (67) [M-
{4}). Starting from compound 13{4} to afford 14{4} in 93% yield,
C6H9N4O]þ. HRMS (70 eV, EI): m/z calculated for C14H12N4O2Cl2:
white solid. IR (KBr), nmax (cm1): 3374, 3207, 2895, 1698, 1651,
338.0340, [M]þ, found: 338.0337. Anal. Calcd. (%) for
1571, 1470, 1371, 1273, 1247, 1008, 785. 1H NMR (400 MHz, DMSO-
C14H12Cl2N4O2: C, 49.58; H, 3.57; N, 16.52. Found: C, 49.41; H, 3.76;
d6): d 11.03 (s, 1H, NH), 8.13 (dt, J ¼ 8.4, 0.9 Hz, 1H, C15eCH), 7.80
N, 16.89.
(dt, J ¼ 8.3, 0.9 Hz, 1H, C18eCH), 7.64 (ddd, J ¼ 8.3, 6.9, 0.9 Hz, 1H,
C16eCH), 7.51 (ddd, J ¼ 8.4, 6.9, 1.0 Hz, 1H, C17eCH), 7.49e7.42 (m,
4.1.3.3. 2-Amino-4-ethoxy-6-(4-fluorophenyl)-5,6-dihydropyrido
1H, C14eCH), 7.16 (t, J ¼ 8.7 Hz, 2H, C13-CHx2), 6.63 (s, 2H, NH2),
[2,3-d]pyrimidin-7(8H)-one (15{2,2}). Starting from compound 14
4.51 (dd, J ¼ 13.9, 7.5 Hz, 1H, C7eCH), 3.31e3.25 (m, 1H, C6eCH),
{2} and ethanol to afford 15{2,2} in 20% yield, white solid. IR (KBr),
3.10e3.03 (m, 1H, C6eCH). 13C NMR (100.6 MHz, DMSO-d6): d 170.0,
nmax (cm1): 3408, 3338, 3220, 1686, 1632, 1575, 1511, 1434, 1227. 1H
165.5, 161.4, 160.9 (d, J ¼ 247.2 Hz, C12), 160.8 (d, J ¼ 246.8 Hz, C12),
NMR (400 MHz, DMSO-d6): d 10.45 (s, 1H, NH), 7.27 (dd, J ¼ 8.8,
160.4, 142.7, 129.9 (t, J ¼ 10.6 Hz, C14), 128.9 (C16), 128.5, 125.1
5.6 Hz, 2H, C12-CHx2), 7.14 (t, J ¼ 8.8 Hz, 2H, C13-CHx2), 6.35 (s, 2H,
(C17), 119.8 (C15), 114.7 (t, J ¼ 18.5 Hz, C11), 111.8 (dd, J ¼ 22.3,
NH2), 4.26 (qd, J ¼ 7.2, 1.3 Hz, 2H, C15eCH2), 3.86 (dd, J ¼ 10.4,
2.5 Hz, C13), 109.6 (C18), 85.1 (C5), 36.1 (C7), 22.4 (C6). Anal. Calcd.
7.2 Hz, 1H, C7eCH), 2.93e2.71 (m, 2H, C6eCH2), 1.25 (t, J ¼ 7.0 Hz,
(%) for C19H13F2N7O2: C, 55.75; H, 3.20; N, 23.95. Found: C, 56.06; H,
3H, C16eCH3). 13C NMR (100.6 MHz, DMSO-d6): d 172.1, 166.4,
3.22; N, 23.78.
162.4, 161.8, 160.0, 158.4, 135.5 (C11), 130.2 (d, J ¼ 8,5 Hz, C12), 115.1
(d, J ¼ 21.0 Hz, C12), 86.7 (C5), 61.5 (C15), 45.4 (C7), 24.8 (C6), 14.6
4.1.2.3. 4-((1H-benzo[d][1,2,3]triazol-1-yl)oxy)-2-amino-6-(2,6- (C16). MS (70 eV, EI): m/z (%) ¼ 302.1 (100) [M]þ, 287.1 (25) [M-
dichloro phenyl)-3,4,5,6-tetrahydropyrido[2,3-d]pyrimidine-7(8H)- CH3]þ, 273.1 (42) [M-C2H5]þ, 258.1 (19) [M-OC2H5]þ. HRMS (70 eV,
one (14{5}). Starting from compound 13{5} to afford 14{5} in 78% EI): m/z calculated for C15H15N4O2F: 302.1183, [M]þ, found:
yield, white solid. IR (KBr): n (cm1): 3382, 3324, 3215, 2898, 1670, 302.1179.
1648, 1571, 1367, 1241, 1089, 1037, 774, 734. 1H NMR (400 MHz,
DMSO-d6): d 11.00 (s, 1H), 8.13 (dt, J ¼ 8.4, 0.9 Hz, 1H), 7.81 (dt, 4.1.3.4. 2-Amino-6-(2,6-dichlorophenyl)-4-ethoxy-5,6-dihydropyrido
J ¼ 8.4, 1.0 Hz, 1H), 7.68e7.61 (m, 1H), 7.60e7.46 (m, 3H), 7.39 (t, [2,3-d]pyrimidin-7(8H)-one (15{5,2}). Starting from compound 14
J ¼ 8.1 Hz, 1H), 6.63 (s, 2H, NH2), 4.91 (dd, J ¼ 12.6, 9.3 Hz, 1H), {5} and ethanol to afford 15{5,2} in 31% yield, white solid. IR (KBr),
3.31e3.16 (m, 2H). 13C NMR (100.6 MHz, DMSO-d6): 170.14, 166.17, nmax (cm1): 3409, 3329, 3220, 1686, 1633, 1577, 1435, 1373, 778. 1H
161.79, 160.48, 143.17, 135.79, 135.38, 135.23, 130.39, 130.20, 129.42, NMR (400 MHz, DMSO-d6): d 10.47 (s, 1H, NH), 7.54e7.48 (m, 2H,
128.94, 128.86, 125.58, 120.22, 110.05, 84.62, 43.20, 21.98. HRMS C13-CHx2), 7.36 (t, J ¼ 8.1 Hz, 1H, C13eCH), 6.38 (s, 2H, NH2), 4.62
(70 eV, EI): m/z calculated for C19H13N7O2Cl2: 441.0508, [M]þ, (dd, J ¼ 12.8, 9.3 Hz, 1H, C7eCH), 4.26 (qd, J ¼ 7.1, 2.1 Hz, 2H,
found: 441.0507. C15eCH2), 2.88e2.79 (m, 2H, C6eCH2), 1.25 (t, J ¼ 7.0 Hz, 3H,
C16eCH3). 13C NMR (100.6 MHz, DMSO-d6): d 169.5, 166.4,
161.83,157.9, 135.3, 134.7, 129.7, 128.3, 85.1 (C5), 61.4 (C15), 43.1
4.1.3. General procedure for the synthesis of 4-alkoxy-substituted
(C7), 22.2 (C6), 14.5 (C16). HRMS (70 eV, EI): m/z calculated for
pyrido[2,3-d]pyrimidines 15{x,y}
C15H14N4O2Cl2: 352.0494, [M]þ, found: 352.0494.
A mixture of the corresponding 4-((1H-benzo[d][1,2,3]triazol-1-
yl)oxy)-6-aryl-3,4,5,6-tetrahydropyrido[2,3-d]pyrimidine-7(8H)-
4.1.3.5. 2-Amino-6-(2,6-dichlorophenyl)-4-(2-methoxyethoxy)-5,6-
one (14{x}) (0.42 mmol), Cs2CO3 (0.54 mmol) and the corre-
dihydropyrido[2,3-d]pyrimidin-7(8H)-one (15{5,3}). Starting from
sponding alcohol (9 mL) was heated a reflux overnight. The
compound 14{5} and 2-methoxyethanol to afford 15{5,3} in 76%
resulting solution was cooled down, water (100 mL) was added and
yield, white solid. IR (KBr), nmax (cm1): 3404, 3218, 2894, 1688,
the reaction crude was neutralized with 2 M HCl. The resulting solid
1633, 1576, 1434, 1372, 775. 1H NMR (400 MHz, DMSO-d6): d 10.54
was collected by filtration and washed with water, and EtOEt to
(s, 1H, NH), 7.55e7.48 (m, 2H, C-13-CHx2), 7.37 (t, J ¼ 8.1 Hz, 1H,
afford the corresponding 15{x,y}.
C14eCH), 6.44 (s, 2H, NH2), 4.64 (dd, J ¼ 12.8, 9.4 Hz, 1H, C7eCH),
4.37e4.32 (m, 2H, C15eCH2), 3.60 (t, J ¼ 4.8 Hz, 2H, C16eCH2), 3.26
4.1.3.1. 2-Amino-6-(4-fluorophenyl)-4-methoxy-5,6-dihydropyrido (s, 3H, OCH3), 2.93e2.79 (m, 2H, C6eCH2). 13C NMR (100.6 MHz,
[2,3-d]pyrimidin-7(8H)-one (15{2,1}). Starting from compound 14 DMSO-d6): d 169.8, 166.4, 161.6, 157.9, 135.3, 135.2, 134.7, 129.8
{2} and methanol to afford 15{2,1} in 39% yield, white solid. IR (KBr), (C14), 129.7 (C13), 128.3 (C13), 85.2 (C5), 70.1 (C16), 64.8 (C15), 58.2
nmax (cm1): 3381, 3227, 1686, 1658, 1633, 1575, 1511, 1464, 1384, (OCH3), 43.1 (C7), 22.2 (C6). MS (70 eV, EI): m/z (%) ¼ 382.0 (15)
1247. 1H NMR (400 MHz, DMSO-d6): d 10.49 (s, 1H, NH), 7.26 (dd, [M]þ, 351.0 (23) [M-CH3O]þ, 322.9 (33) [M-C3H6O]þ, 289.0 (100)
J ¼ 8.7, 5.7 Hz, 2H, C12-CHx2), 7.14 (t, J ¼ 8.9 Hz, 2H, C13-CHx2), 6.40 [M-C3H6OCl]þ, 205.9 (26) [C9H10N4O2]þ. HRMS (70 eV, EI): m/z
(s, 2H, NH2), 3.86 (dd, J ¼ 9.9, 6.9 Hz, 1H, C7eCH), 3.80 (s, 3H; OCH3), calculated for C16H16N4O3F4Cl2: 382.0598, [M]þ, found: 382.0599.
2.93e2.72 (m, 2H, C6eCH2). 13C NMR (100.6 MHz, DMSO-d6):
d 172.0, 166.7, 161.7, 158.3, 153.0, 135.4 (d, J ¼ 2.9 Hz, C11), 130.1 (d, 4.1.3.6. 2-Amino-6-(2,6-dichlorophenyl)-4-(2-propoxyethoxy)-5,6-
J ¼ 8.1 Hz, C12), 115.0 (d, J ¼ 21.3 Hz, C13), 86.5 (C5), 53.2 (OCH3), dihydropyrido[2,3-d]pyrimidin-7(8H)-one (15{5,9}). Starting from
45.3 (7), 24.6 (6). MS (70 eV, EI): m/z (%) ¼ 288.2 (100) [M]þ, 193.0 compound 14{5} and 2-propoxyethanol to afford 15{5,9} in 38%
(25) [M-C6H4F]þ. HRMS (70 eV, EI): m/z calculated for C14H13N4O2F: yield, white solid. IR (KBr), nmax (cm1): 3479, 3332, 3220, 2773,
288.1028, [M]þ, found: 288.1023. 1692, 1631, 1575, 1458, 1435, 1373, 777. 1H NMR (400 MHz, DMSO-
M. Camarasa et al. / European Journal of Medicinal Chemistry 115 (2016) 463e483 473

d6): d 10.51 (s, 1H, NH), 7.58e7.47 (m, 2H, C13eCH), 7.37 (t, 59.80; H, 6.04; N, 19.82.
J ¼ 8.1 Hz, 1H, C14eCH), 6.42 (s, 2H, NH2), 4.63 (dd, J ¼ 12.9, 9.2 Hz,
1H, C7eCH), 4.35e4.32 (m, 2H, C15eCH2), 3.63 (t, J ¼ 4.9 Hz, 2H, 4.1.4.3. 2-Amino-6-(4-fluorophenyl)-4-(pyrrolidin-1-yl)-5,6-
C16eCH2), 3.35 (t, J ¼ 6.6 Hz, 2H, C17eCH2), 2.93e2.79 (m, 2H, dihydropyrido[2,3-d]pyrimidin-7(8H)-one (15{2,7}). Starting from
C6eCH2), 1.46 (h, J ¼ 7.3 Hz, 2H, C18eCH2), 0.80 (t, J ¼ 7.4 Hz, 3H, compound 13{2} and pyrrolidine and following procedure A to
CH3). 13C NMR (100.6 MHz, DMSO-d6): d 169.6, 166.4, 161.8, 158.0, afford 15{2,7} in 44% yield, white solid. IR (KBr), nmax (cm1): 3441,
135.3, 135.1, 129.8, 128.3, 85.2 (C5), 71.9 (C17), 68.2 (C16), 65.0 (C15), 3329, 3227, 2963, 2873, 1695, 1639, 1608, 1540, 1510, 1448, 1340,
43.1 (C7), 22.4 (C18), 22.2 (C16), 10.4 (C19). Anal. Calcd. (%) for 1232, 840. 1H NMR (400 MHz, DMSO-d6): d 10.31 (s, 1H, NH),
C18H20Cl2N4O2: C, 52.57; H, 4.90; N, 13.62. Found: C, 52.76; H, 5.12; 7.30e7.24 (m, 2H, C12-2xCH), 7.14 (t, J ¼ 8.9 Hz, 2H, C13-2xCH), 5.89
N, 13.98. (s, 2H, NH2), 3.78 (dd, J ¼ 10.4, 6.6 Hz, 1H, C7eCH), 3.50e3.41 (m,
4H, 2xCH2), 3.16e3.02 (m,2H, C6eCH2) 1.85e1.67 (m, 4H, 2xCH2).
13
4.1.4. General procedure for the synthesis of 4-amino-substituted C NMR (100.6 MHz, DMSO-d6): d 171.6 (CO), 161.2, 161.1 (d,
pyrido [2,3-d]pyrimidines 15{x,y} J ¼ 241.2 Hz, C14) 160.7, 157.5, 135.5 (d, J ¼ 3.0 Hz, C11), 130.3 (d,
Procedure A: 4.88 mmol of the corresponding 4-oxopyrido[2,3- J ¼ 8.1 Hz, C12), 114.9 (d, J ¼ 21.0 Hz, C13), 86.3 (C5), 48.9 (C15), 45.9
d]pyrimidine (13{x}) was suspended in acetonitrile (350 mL), then (C7), 28.5 (C6), 25.0 (C16). HRMS (70 eV, EI): m/z calculated for
BOP (2.805 g, 6.34 mmol) and DBU (1.1 mL, 7.32 mmol) were added C17H18N5OF: 327.1495, [M]þ, found: 327.1496.
to the solution. The mixture was stirred at room temperature for
2 h. The corresponding amine (14.6 mmol) was added and the 4.1.4.4. 2-Amino-6-(2,6-difluorophenyl)-4-((2-propoxyethyl)amino)-
mixture was heated at 90  C for 2 days. The solvent was concen- 5,6-dihydropyrido[2,3-d]pyrimidin-7(8H)-one (15{4,6}).
trated under reduced pressure and water was added to the residue. Starting from compound 14{4} and 2-propoxyethylamine and
The solid was collected by filtration and washed with water and following procedure B to afford 15{4,6} in 60% yield, white solid. IR
EtOEt. (KBr), nmax (cm1): 3416, 3332, 3212, 2929, 2871, 1630, 1577, 1470,
Procedure B: 0.419 mmol of the corresponding benzotriazol-1- 1388, 1007, 783. 1H NMR (400 MHz, DMSO-d6): d 10.25 (s, 1H, NH),
yloxy substituted intermediate 14{x} was suspended in acetoni- 7.46e7.37 (m, 1H, C14eCH), 7.13 (t, J ¼ 8.8 Hz, 2H, C13-CHx2), 6.34
trile (20 mL) and 3 equivalents of the corresponding amine (s, 1H, NH), 5.91 (s, 2H, NH2), 4.16 (dd, J ¼ 14.0, 7.6 Hz, 1H, C7eCH),
(1.257 mmol) was added to the suspension and the mixture was 3.44e3.43 (m, 4H, C15eCH2, C16eCH2), 3.34e3.31 (m, 2H,
heated at 140  C by microwave irradiation for 5 h. Then, water was C17eCH2), 2.97e2.78 (m, 2H, C6eCH2), 1.48 (h, J ¼ 7.4 Hz, 2H,
added to the residue and the solid was collected by filtration and C18eCH2), 0.83 (t, J ¼ 7.4 Hz, 3H, C19eCH3). 13C NMR (100.6 MHz,
washed with water and EtOEt. DMSO-d6): d 170.3, 160.8 (d, J ¼ 246.8 Hz, C12), 160.7 (d,
J ¼ 246.6 Hz, C12), 161.8, 160.2, 155.6, 129.7 (t, J ¼ 10.2 Hz, C14),
4.1.4.1. 2-Amino-6-(4-fluorophenyl)-4-(pentylamino)-5,6- 115.4 (t. J ¼ 18.7 Hz, C11), 111.7 (dd, J ¼ 1.9, 22.3 Hz, C13), 83.6 (C5),
dihydropyrido [2,3-d]pyrimidin-7(8H)-one (15{2,5}). Starting from 71.7 (C17), 68.7 (C16), 40.0 (C15), 36.4 (C7), 24.0 (C6), 22.5 (C18),
compound 13{2} and pentylamine and following procedure A to 10.5 (C19). Anal. Calcd. (%) for C18H21F2N5O2: C, 57.29; H, 5.61; N,
afford 15{2,5} in 40% yield, white solid. IR (KBr), nmax (cm1): 3395, 18.56. Found: C, 57.25; H, 3.41; N, 18.74.
3333, 3219, 2930, 2869, 1694, 1630, 1578, 1510, 1469, 1376, 1214,
841. 1H NMR (400 MHz, acetone): d 7.38 (m, 2H, 2xCH), 7.11 (t, 4.1.4.5. 2-Amino-6-(2,6-dichlorophenyl)-4-(pentylamino)-5,6-
J ¼ 8.8 Hz, 2H, 2xCH), 3.94 (dd, J ¼ 10.3, 7.2 Hz, 1H, CHeC7), 3.42 dihydropyrido [2,3-d]pyrimidin-7(8H)-one (15{5,5}). Starting from
(dd, J ¼ 12.9, 6.6 Hz, 2H, CH2eC15), 2.87 (m, 2H, CH2eC6), 1.67e1.51 compound 13{5} and pyrrolidine and following procedure A to
(m, 2H, CH2eC16), 1.41e1.25 (m, 4H, 2xCH2eC17, C18), 0.88 (t, afford 15{5,5} in 62% yield, white solid. IR (KBr), nmax (cm1): 3505,
J ¼ 6.8 Hz, 3H, CH3). 13C NMR (100.6 MHz, acetone) d 174.4 (CO), 3392, 3323, 3206, 2929, 2870, 1675, 1629, 1577, 1472, 1436, 1385,
163.3, 162.9 (d, J ¼ 69.1 Hz, C14), 161.6, 156.6 (C10), 136.6 (d, 1287, 776. 1H NMR (400 MHz, DMSO-d6): d 10.57 (s, 1H, CONH), 7.53
J ¼ 3.3 Hz, C11), 131.2 (d, J ¼ 8.1 Hz, C12), 115.8 (d, J ¼ 21.3 Hz, C31), (m, 2H, C12-CHx2), 7.37 (t, J ¼ 8.1 Hz, 1H, C13eCH), 6.30 (s, 1H, NH),
85.1 (C5), 46.7 (C7), 41.5 (C15), 29.2 (C16), 29.0 (C17), 26.6 (C6), 23.2 6.06 (s, 2H, NH2), 4.64 (dd, J ¼ 13.1, 8.9 Hz, 1H, C7eCH), 3.31e3.20
(C18), 14.4 (C19). MS (70 eV, EI): m/z (%) ¼ 343.3 [M]þ, 314.2 [M- (m, 2H, C14eCH2), 2.94e2.65 (m, 2H, CH2), 1.49 (p, J ¼ 7.1 Hz, 2H,
C2H5]þ, 300.1 [M-C3H9]þ, 286.1 [M-C4H9]þ, 272.1 [M-C5H11]þ. C15eCH2), 1.27 (m, 4H, C15,C16eCH2x2), 0.85 (t, J ¼ 6.9 Hz, 3H,
CH3). 13C NMR (100.6 MHz, DMSO-d6): d 169.8, 161.8, 160.5, 154.9,
4.1.4.2. 2-Amino-6-(4-fluorophenyl)-4-((2-propoxyethyl)amino)-5,6- 135.7, 135.2, 134.8, 129.7, 128.3, 82.5 (C5), 43.1 (C7), 40.3 (C14) 28.8
dihydropyrido[2,3-d]pyrimidin-7(8H)-one (15{2,6}). Starting from (C15), 28.7 (C16), 23.2 (C6), 22.0 (C17), 14.0 (C18). MS (70 eV, EI): m/
compound 13{2} and 2-propoxyethylamine and following proce- z (%) ¼ 393.1 (69) [M]þ,351.0 (78) [M-C3H7]þ,336.0 (91) [M-
dure A to afford 15{2,6} in 63% yield, white solid. IR (KBr), nmax C4H9]þ,322.9 (54) [M-C5H11]þ, 248.1 (43) [M-C6H3Cl2]þ, 206.1 (83)
(cm1): 3391, 3226, 2932, 2874, 1689, 1629, 1575, 1511, 1477, 1226. [C9H12N5O]þ, 192.0 (69) [C8H8N5O]þ. HRMS (70 eV, EI): m/z calcu-
1
H NMR (400 MHz, CDCl3): d 11.99 (s, 1H, NH), 7.26e7.20 (m, 2H, lated for C18H21N5OCl2: 393.1123, [M]þ, found: 393.1122.
C12-CHx2), 7.04 (t, J ¼ 8.7 Hz, 2H, C13-CHx2), 4.81e4.75 (m, 1H,
NH), 3.84 (dd, J ¼ 10.2, 7.3 Hz, 1H, C7eCH), 3.64e3.61 (m, 2H, 4.1.4.6. 2-Amino-6-(2,6-dichlorophenyl)-4-((2-propoxyethyl)
C15eCH2), 3.58e3.52 (m, 2H, C16eCH2), 3.39 (t, J ¼ 6.7 Hz, 2H, amino)-5,6-dihydropyrido [2,3-d]pyrimidin-7(8H)-one (15{5,6}).
C17eCH2), 2.85e2.67 (m, 2H, C16eCH2), 1.63e1.51 (m, 2H, Starting from compound 13{5} and 2-propoxyethylamine and
C18eCH2), 0.88 (t, J ¼ 7.4 Hz, 3H, CH3). 13C NMR (100.6 MHz, CDCl3): following procedure A to afford 15{5,6} in 70% yield, white solid. IR
d 173.8 (CO), 161.5 (d, J ¼ 161.0 Hz, C14), 161.1, 155.9, 134.4 (d, (KBr), nmax (cm1): 3440, 3333, 3217, 2928, 2869, 1631, 1576, 1478,
J ¼ 3.4 Hz, C11), 129.9 (d, J ¼ 8.1 Hz, C12), 115.9 (d, J ¼ 21.2 Hz, C13), 776. 1H NMR (400 MHz, DMSO-d6): d 10.28 (s, 1H, CONH), 7.53 (ddd,
84.5 (C5), 72.9 (C17), 69.3 (C16), 46.2 (C7), 40.8 (C15), 26.3 (C6), 22.9 J ¼ 14.8, 8.3, 1.3 Hz, 2H, C13-2xCH), 7.37 (t, J ¼ 8.1 Hz, 1H, C14eCH),
(C18), 10.7 (C19). MS (70 eV, EI): m/z (%) ¼ 359.1 (24) [M]þ, 316.1 6.33 (s, 1H, NH), 5.95 (s, 2H, NH2), 4.60 (dd, J ¼ 13.1, 8.9 Hz, 1H,
(11) [M-C3H5]þ, 301.1 (17) [M-C3H8O]þ, 286.1 (100) [M-C4H9O]þ, C7eCH), 3.44 (m, 4H, C15eCH2, C16eCH2), 3.31e3.34 (m, 2H,
273.1 (80) [M-C5H12O]þ. HRMS (70 eV, EI): m/z calculated for C17eCH2) 2.92e2.64 (m, 2H, C6eCH2), 1.48 (h, J ¼ 7.2 Hz, 2H,
C18H22N5O2F: 359.1758, [M]þ, found: 359.1742. Anal. Calcd. (%) for C18eCH2), 0.84 (t, J ¼ 7.4 Hz, 3H, C19eCH3). 13C NMR (100.6 MHz,
C18H22FN5O2: C, 60.15; H, 6.17; F, 5.29; N, 19.49; O, 8.90. Found: C, DMSO-d6): d 169.5, 161.9, 155.1, 135.6, 135.1, 134.8, 129.7, 128.3,
474 M. Camarasa et al. / European Journal of Medicinal Chemistry 115 (2016) 463e483

82.62 (C5), 71.7 (C17), 68.6 (C16), 43.2 (C7), 39.5 (C15) 23.2 (C6), pyridopyrimidine 13{x} or 15{x,y} and 3.5 mmol of DDQ in 30 mL of
22.5 (C18), 10.5 (C19). HRMS (70 eV, EI): m/z calculated for 1,2-dichloroethane was heated at 100  C overnight protected from
C18H21N5O2Cl2: 409.1077, [M]þ, found: 409.1072. Anal. Calcd. (%) for moisture. The resulting solution was cooled down, water (300 mL)
C18H21Cl2N5O2: C, 52.69; H, 5.16; N, 17.07. Found: C, 52.60; H, 65.36; was added and the resulting precipitate was filtered, washed with
N, 17.24. EtOH and EtOEt and dried in vacuo over phosphorus pentoxide to
afford the corresponding 16{x,y}.
4.1.4.7. 2-Amino-6-(2,6-dichlorophenyl)-4-(pyrrolidin-1-yl)-5,6- Procedure C: A mixture of 0.5 mmol of the corresponding pyr-
dihydropyrido[2,3-d]pyrimidin-7(8H)-one (15{5,7}). Starting from idopyrimidine 13{x} or 15{x,y} and 114 mg (1.0 mmol) of activated
compound 13{5} and pyrrolidine and following procedure A to MnO2 in 8.5 mL of acetic acid was refluxed for 3 h. The resulting hot
afford 15{5,7} in 78% yield, white solid. IR (KBr), nmax (cm1): 3468, suspension was filtered and the solvent was removed in vacuo. The
3309, 3203, 2971, 2874, 1686, 1614, 1540, 1457, 1441, 1386, 1340, resulting solid was refluxed with water in order to eliminate MnO2
1286, 772. 1H NMR (400 MHz, DMSO-d6): d 10.11 (s, 1H, NH), 7.50 traces, then filtrated and dried in vacuo over phosphorus pentoxide
(dd, J ¼ 15.6, 8.4 Hz, 4H, C13-CHx2), 7.36 (t, J ¼ 8.0 Hz, 1H, C14eCH), to afford the corresponding 16{x,y}.
5.85 (s, 2H, NH2), 4.48 (dd, J ¼ 13.6, 8.0 Hz, 1H, C7eCH), 3.46 (m, 4H,
C15eCH2x2), 2.85e2.65 (m, 2H, C6eCH2), 1.76 (m, 4H, C16eCH2x2).
13 4.1.6.1. 2-Amino-6-(4-fluorophenyl)pyrido[2,3-d]pyrimidine-
C NMR (100.6 MHz, TFA-d): d 173.7, 151.8, 151.6, 134.8, 130.7, 130.6,
4,7(3H,8H)-dione (16{2}). Starting from 13{2} and following pro-
129.6, 128.5, 86.8 (C5), 51.9 (C15), 42.5 (C7), 24.4 (C16, C7). MS
cedure B to afford 16{2} in 90% yield, yellow solid. IR (KBr), nmax
(70 eV, EI): m/z (%) ¼ 377.1 (100) [M]þ, 232.1 (73) [M-C6H3Cl2]þ,
(cm1): 3323, 3096, 2922, 1633, 1505, 1393, 1231, 1159, 582. 1H NMR
218.0 (42) [M-C7H5Cl2]þ, 204.0 (59) [M-C8H7Cl2]þ, 176 (13) [M-
(400 MHz, DMSO-d6): d 11.92 (s, 1H, NHeCO), 7.84 (s, 1H, C6eCH),
C10H11Cl2]þ. HRMS (70 eV, EI): m/z calculated for C17H17N5OCl2:
7.71 (dd, J ¼ 8.9, 5.6 Hz, 2H, C12-2xCH), 7.19 (t, J ¼ 9.0 Hz, 2H, C13-
377.0801, [M]þ, found: 377.0810.
2xCH), 6.08 (s, 2H, NH2). 13C NMR (100.6 MHz, DMSO-d6) d 162.7
(CO), 162.4, 160.0, 156.2, 155.7, 134.0 (C7), 132.8 (d, J ¼ 3.0 Hz), 130.0
4.1.4.8. 2-Amino-4-(benzylamino)-6-(2,6-dichlorophenyl)-5,6-
(d, J ¼ 31.8 Hz, C12), 121.9 (C6) 114.7 (d, J ¼ 84.1 Hz C13), 95.9 (C5).
dihydropyrido[2,3-d]pyrimidin-7(8H)-one (15{5,8}). Starting from
HRMS (ESIeTOF): m/z calculated for C13H9FN4O2: 273.0775,
compound 14{5} and benzylamine and following procedure B to
[MþH]þ, found: 273.0782.
afford 15{5,8} in 60% yield, white solid. 1H NMR (400 MHz, DMSO-
d6): d 10.18 (s, 1H, NH), 7.56e7.47 (m, 3H), 7.39e7.34 (m, 2H),
7.30e7.28 (m, 2H), 7.22e7.17 (m, 1H), 6.88 (t, J ¼ 6.0 Hz, 1H, NH), 4.1.6.2. 2-Amino-6-(4-fluorophenyl)-4-(pentylamino)pyrido[2,3-d]
5.90 (s, 2H, NH2), 4.62 (dd, J ¼ 13.1, 8.9 Hz, 1H, C7eCH), 4.52 (td, pyrimidin-7(8H)-one (16{2,5}). Starting from 15{2,5} and following
J ¼ 9.2, 3.5 Hz, 2H, C15eCH2), 2.97e2.66 (m, 2H, C6eCH2). HRMS procedure C to afford 16{2,5} in 73% yield, white solid. IR (KBr), nmax
(70 eV, EI): m/z calculated for C20H17N5OCl2: 413.0810, [M]þ, found: (cm1): 3380, 3171, 2930, 2859, 1619, 1564, 1509, 1475, 1227, 837,
413.0809. 796. 1H NMR (400 MHz, DMSO-d6): d 12.11 (s, 1H, NHCO), 8.27 (s,
1H, C6eCH), 7.76 (m, 2H, C12-CHx2), 7.24 (m, 2H, C13-CHx2),
4.1.5. 2,4-Diamino-6-(4-fluorophenyl)-5,6-dihydropyrido[2,3-d] 3.47e3.30 (m, 2H, C15eCH2) 1.65e1.54 (m, 2H, C16eCH2),
pyrimidin-7(8H)-one (15{2,4}) 1.37e1.29 (m, 4H, C17eCH2, C18eCH2), 0.89 (t, J ¼ 6.8 Hz, 3H, CH3).
13
Methyl 2-(4-fluorophenyl)acrylate (5{2}) (4.00 mmol) was dis- C NMR (100.6 MHz, DMSO-d6): d 162.8, 162.5, 160.0, 159.6, 155.5,
solved in anhydrous ethylene glycol (20 mL). 2,6- 130.6, 133.1 (d, J ¼ 6.6 Hz, C11), 130.3 (d, J ¼ 7.7 Hz, C12), 121.0 (C7),
diaminopyrimidin-4(3H)-one (4.00 mmol) and sodium methoxide 114.7 (d, J ¼ 21.0 Hz, C13), 91.7 (C5), 40.4 (C15), 28.8 (C17), 28.6
(4.00 mmol) were added into the solution. The mixture was heated (C16), 22.1 (C18), 14.0 (C19). HRMS (70 eV, EI): m/z calculated for
under microwave irradiation at 180  C for 3 h. Water was added and C18H20N5OF: 341.1653, [M]þ, found: 341.1652.
the reaction crude was neutralized with a 2 M solution of HCl. The
solid was collected by filtration and washed with water, MeOH and
4.1.6.3. 2-Amino-6-(4-fluorophenyl)-4-((2-propoxyethyl)amino)pyr-
Et2O to afford 15{2,4} as a white solid in 26% yield. IR (KBr), nmax
ido[2,3-d]pyrimidin-7(8H)-one (16{2,6}). Starting from 15{2,6} and
(cm1): 3495, 3334, 3219, 3104, 2888, 1698, 1659, 1638, 1570, 1510,
following procedure C to afford 16{2,6} in 72% yield, white solid. IR
1460, 1210, 837. 1H NMR (400 MHz, DMSO-d6): d 10.72 (s, 1H, NH),
(KBr), nmax (cm1): 3380, 2177, 2961, 2873, 1621, 1566, 1509, 1481,
7.31 (dd, J ¼ 8.8, 5.6 Hz, 2H, C12-CHx2), 7.15 (t, J ¼ 8.9 Hz, 2H, C13-
1228, 838,796. 1H NMR (400 MHz, DMSO-d6): d 12.26 (s, 1H, NH),
CHx2), 6.26 (s, 2H, C2eNH2), 6.01 (s, 2H, C4eNH2), 3.88 (dd,
8.30 (s, 1H, C6eCH), 7.90 (s, 1H, NH), 7.83e7.73 (m, 2H, C12eCH2),
J ¼ 10.4, 6.9 Hz, 1H, C7eCH), 2.90e2.69 (m, 2H, C6eCH2). 13C NMR
7.23 (t, J ¼ 8.7 Hz, 3H, C13eCH2), 6.98 (s, 2H, NH2), 3.60e3.55 (m,
(100.6 MHz, DMSO-d6): d 172.6 (CO), 161.8, 161.7, 161.2 (d,
4H, C15eCH2, C16eCH2), 3.40e3.35 (m, 2H, C17eCH2), 1.57e1.45
J ¼ 241 Hz, C14) 156.3, 135.7 (d, J ¼ 3.1 Hz, C11), 130.2 (d, J ¼ 7.9 Hz,
(m, 2H, C18eCH2), 0.85 (t, J ¼ 7.4 Hz, 3H, C19eCH3). 13C NMR
C12), 115.0 (d, J ¼ 20.9 Hz, C13), 83.9 (C5), 45.5 (C7), 25.6 (C6). MS
(100.6 MHz, DMSO-d6): d 162.6, 161.1 (d, J ¼ 245.0 Hz, C14), 159.6,
(70 eV, EI): m/z (%) ¼ 273.1 (100) [M]þ. HRMS (70 eV, EI): m/z
155.6, 133.1 (d, J ¼ 1.9 Hz, C11), 132.8, 130.1 (d, J ¼ 7.9 Hz, C12), 121.1,
calculated for C13H12N5OF: 273.1026, [M]þ, found: 273.1026.
121.0, 114.5 (d, J ¼ 21.1 Hz, C13), 91.5 (C5), 71.8 (C17), 68.5 (C16),
39.5 (C15), 22.4 (C18), 10.5 (C19). HRMS (ESIeTOF): m/z calculated
4.1.6. General procedure for aromatization of pyrido[2,3-d]
for C18H21FN5O2, 358.1678, [M-H]þ, found: 358.1674.
pyrimidines 13{x} or 15{x,y}
Procedure A: A mixture of 0.5 mmol of the corresponding pyr-
idopyrimidine 13{x} or 15{x,y} and 60.0 mg (1.5 mmol) of sodium 4.1.6.4. 2-Amino-6-(4-fluorophenyl)-4-(pyrrolidin-1-yl)pyrido[2,3-d]
hydride (60% dispersion in mineral oil) in 5 mL of anhydrous DMSO pyrimidin-7(8H)-one (16{2,7}). Starting from 15{2,7} and following
was heated for 4 h at 100  C protected from moisture. The resulting procedure C to afford 16{2,7} in 82% yield, light brown solid. IR
solution was cooled down, water (300 mL) was added and it was (KBr), nmax (cm1):3459, 3311, 3181, 2970, 2874, 1635, 1606, 1530,
neutralized with AcOH. The resulting precipitate was filtered, 1511, 1452, 1452, 1224, 839. 1H NMR (400 MHz, DMSO-d6): d 11.77
washed with MeOH and EtOEt and dried in vacuo over phosphorus (s, 1H, NH), 8.10 (s, 1H, C6eCH), 7.73 (m, 2H, C12-CHx2), 7.19 (t,
pentoxide to afford the corresponding 16{x,y}. J ¼ 9.0 Hz, 2H, C13-CHx2), 6.65 (s, 2H, NH2), 3.76 (t, J ¼ 6.4 Hz, 4H,
Procedure B: A mixture of 2.7 mmol of the corresponding C15eCH2x2), 1.91 (t, J ¼ 6.5 Hz, 4H, C16eCH2x2).
M. Camarasa et al. / European Journal of Medicinal Chemistry 115 (2016) 463e483 475

4.1.6.5. 2-Amino-6-(2,6-difluorophenyl)-4-((2-propoxyethyl)amino) of DMSO, 28.0 mg (0.7 mmol) of sodium hydride (60% dispersion in
pyrido[2,3-d]pyrimidin-7(8H)-one (16{4,6}). Starting from 15{4,6} mineral oil) were added and the mixture was stirred for 1 h at room
and following procedure A to afford 16{4,6} in 60% yield, light temperature under nitrogen atmosphere. After this period,
brown solid. IR (KBr), nmax (cm1): 3387, 3188, 2931, 2858, 1627, 0.7 mmol of methyl iodide were added dropwise and then stirred
1568, 1466, 1004, 788. 1H NMR (400 MHz, DMSO-d6): d 11.93 (s, 1H, overnight at rt. The reaction was quenched by addition of 300 mL of
NH), 8.16 (s, 1H, C6eCH), 7.70 (s, 1H, NH), 7.49e7.41 (m, 1H, water and the resulting precipitate was filtered, washed with water
C14eCH), 7.20e7.12 (m, 2H, C13-CHx2), 6.81 (s, 2H, NH2), 3.50e3.57 and dried in vacuo over phosphorus pentoxide to afford the cor-
(m, 4H, C15eCH2, C16eCH2), 3.35e3.40 (m, 2H, C17eCH2), 1.49 (h, responding 17{x,y}.
J ¼ 7.4 Hz, 2H, C18eCH2), 0.84 (t, J ¼ 7.4 Hz, 3H, C19eCH3). 13C NMR
(100.6 MHz, DMSO-d6): d 163.0, 161.9, 160.4 (d, J ¼ 246.6 Hz, C12), 4.1.7.1. 2-Amino-6-(4-fluorophenyl)-4-methoxy-8-methylpyrido[2,3-
160.3 (d, J ¼ 246.7 Hz, C12), 159.6, 156.4, 136.7, 129.9 (t, J ¼ 10.3 Hz, d]pyrimidin-7(8H)-one (17{2,1}). Starting from 16{2,1} to afford 17
C14), 113.8 (t, J ¼ 20.1 Hz, C11), 111.6e111.3 (m, C13), 111.2, 91.0 (C5), {2,1} in 71% yield, white solid. IR (KBr), nmax (cm1): 3494, 3338,
71.8 (C17), 68.3 (C16), 40.2 (C15), 22.4 (C18), 10.5 (C19). HRMS 2926, 1608, 1587, 1510, 1468, 1390, 834, 796. 1H NMR (400 MHz,
(70 eV, EI): m/z calculated for C18H19N5O2F2: 375.1507, [M]þ, found: CDCl3): d 7.88 (s, 1H, C7eCH), 7.65 (dd, J ¼ 8.9, 5.5 Hz, 2H, C12-
375.1509. CHx2), 7.08 (t, J ¼ 8.8 Hz, 2H, C13-CHx2), 4.03 (s, 3H, OCH3), 3.71
(s, 3H, NCH3). 13C NMR (100.6 MHz, CDCl3) d 167.85, 163.72, 163.13,
4.1.6.6. 2-Amino-6-(2,6-dichlorophenyl)-4-(2-propoxyethoxy)pyrido 161.27, 156.81, 132.95 (C7), 130.61 (d, J ¼ 7.9 Hz,C12-CHx2), 130.45
[2,3-d]pyrimidin-7(8H)-one (16{5,9}). Starting from 15{5,9} and (C6), 125.64 (C11), 115.12 (d, J ¼ 21.3 Hz,C13-CHx2), 94.75 (C5),
following procedure B to afford 16{5,9} in 82% yield, white solid. IR 54.42 (OCH3), 28.89 (NCH3). HRMS (70 eV, EI): m/z calculated for
(KBr), nmax (cm1): 3392, 3183, 2962, 2868, 1626, 1553, 1509, 1432, C15H13N4O2F: 300.1025, [M]þ, found: 300.1023.
1336, 1252, 802. 1H NMR (400 MHz, DMSO-d6): d 12.00 (s, 1H, NH),
7.58e7.51 (m, 3H, C13-CHx2, C7eCH), 7.42 (dd, J ¼ 8.7, 7.4 Hz, 1H, 4.1.7.2. 2-Amino-6-(4-fluorophenyl)-8-methyl-4-(pentylamino)pyr-
C14eCH), 7.21 (s, 2H, NH2), 4.51e4.45 (m, 2H, C15eCH2), 3.74e3.68 ido[2,3-d]pyrimidin-7(8H)-one (17{2,5}). Starting from 16{2,5} to
(m, 2H, C16eCH2), 3.38 (t, J ¼ 6.6 Hz, 2H, C17eCH2), 1.47 (h, afford 17{2,5} in 89% yield, white solid. IR (KBr), nmax (cm1): 3345,
J ¼ 7.3 Hz, 2H, C18eCH2), 0.80 (t, J ¼ 7.4 Hz, 3H, CH3). 13C NMR 2930, 2858, 1603, 1567, 1508, 1466, 835, 799. 1H NMR (400 MHz,
(100.6 MHz, DMSO-d6): d 166.1, 162.7, 161.4, 157.4, 150.8, 135.2, CDCl3): d 7.91 (s, 1H, C6eCH), 7.52e7.46 (m, 2H, C12eCHx2), 6.94 (t,
134.5, 134.2 (C6), 130.3 (C14), 129.2, 128.0 (C13), 122.1, 91.6 (C5), J ¼ 8.4 Hz, 2H, C13eCHx2), 5.64 (t, J ¼ 5.4 Hz, 1H, NH), 5.09 (s, 2H,
71.9 (C17), 67.9 (C16), 65.7 (C15), 22.4 (C18), 10.4 (C19). HRMS NH2), 3.63 (s, 3H, CH3), 3.53e3.41 (m, 2H, C15eCH2), 1.65e1.58 (m,
(70 eV, EI): m/z calculated for C18H18N4O3Cl2: 408.0750, [M]þ, 2H, C16eCH2), 1.40e1.29 (m, 4H, C17eCH2, C18eCH2), 0.90 (t,
found: 408.0756. J ¼ 7.2 Hz, 3H, CH3). 13C NMR (100.6 MHz, CDCl3): d 163.4, 162.9,
161.3 (d, J ¼ 166.4 Hz, C14), 161.0, 155.8, 133.2 (d, J ¼ 3.2 Hz, C11),
4.1.6.7. 2-Amino-6-(2,6-dichlorophenyl)-4-(pentylamino)pyrido[2,3- 130.4 (d, J ¼ 7.8 Hz, C12), 129.4 (C6), 123.4 (C7), 114.9 (d, J ¼ 21.3 Hz,
d]pyrimidin-7(8H)-one (16{5,5}). Starting from 15{5,5} and C13), 92.8 (C5), 41.4 (C15), 29.3, 29.3, 28.78 (NeCH3), 22.5 (C18),
following procedure B to afford 16{5,5} in 51% yield, white solid. IR 14.1 (C19). MS (70 eV, EI): m/z (%) ¼ 355.2 (100) [M]þ, 312.1 (42) [M-
(KBr), nmax (cm1): 3126, 1856, 1640, 1465, 1430, 169, 780. 1H NMR C3H7]þ, 298.1 (61) [M-C4H9]þ, 285.1 (62) [M-C5H12]þ, 158.0 (40) [M-
(400 MHz, DMSO-d6): d 10.27 (s, 1H, NH), 8.89 (s, 1H, NH), 8.36 (s, C12H20FN]þ.
1H, C6eCH), 7.98 (s, 2H, NH2), 7.60 (d, J ¼ 7.8 Hz, 2H, C13-CHx2),
7.47 (dd, J ¼ 8.8, 7.4 Hz, 1H, C14eCH), 3.49 (q, J ¼ 6.8 Hz, 2H, 4.1.7.3. 2-Amino-6-(4-fluorophenyl)-8-methyl-4-((2-propoxyethyl)
C15eCH2), 1.60 (m, 2H, C16eCH2), 1.37e1.25 (m, 4H, C17eCH2, amino)pyrido[2,3-d]pyrimidin-7(8H)-one (17{2,6}). Starting from 16
C18eCH2), 0.91e0.83 (m, 3H, C19eCH3). 13C NMR (100.6 MHz, {2,6} to afford 17{2,6} in 87% yield, white solid. IR (KBr), nmax
DMSO-d6): d 169.9, 161.3, 155.7, 154.9, 135.4 (C6), 135.0, 134.7, 133.9, (cm1): 3341, 3199, 2932, 2872, 1603, 1570, 1509, 1468, 1227, 1160,
130.7 (C14), 129.7, 128.1 (C13), 86.8 (C5), 43.4 (C15), 28.5 (C17), 27.9 837, 799. 1H NMR (400 MHz, CDCl3): d 7.58 (dd, J ¼ 8.7, 5.6 Hz, 2H,
(C16), 21.8 (C18), 13.9 (C19). MS (70 eV, EI): m/z (%) ¼ 391.2 (20.6) C12eCHx2), 7.47 (s, 1H, C6eCH), 7.05 (t, J ¼ 8.8 Hz, 2H, C13eCHx2),
[M]þ, 356.3 (100) [M-Cl]þ, 326.1 (19.0) [M-C2H5Cl]þ, 300.1 (7.5) [M- 5.81 (s, 1H, NH), 5.03 (s, 2H, NH2), 3.72 (q, J ¼ 5.1 Hz, 2H, C15eCH2),
C4H9Cl]þ. HRMS (70 eV, EI): m/z calculated for C18H19N5OCl2: 3.68 (s, 3H, NCH3), 3.63 (t, J ¼ 5.0 Hz, 2H, C16eCH2), 3.44 (t,
391.0967, [M]þ, found: 391.0967. J ¼ 6.7 Hz, 2H, C17eCH2), 1.61 (dq, J ¼ 14.2, 7.2 Hz, 2H, C18eCH2),
0.93 (t, J ¼ 7.4 Hz, 3H, C19eCH3). 13C NMR (100.6 MHz, CDCl3):
4.1.6.8. 2-Amino-6-(2,6-dichlorophenyl)-4-((2-propoxyethyl)amino) d 163.59, 162.00 (d, J ¼ 162.0 Hz), 160.32, 155.98, 155.52, 133.21,
pyrido[2,3-d]pyrimidin-7(8H)-one (16{5,6}). Starting from 15{5,6} 130.50 (d, J ¼ 7.9 Hz, C12), 128.97 (C6), 124.01, 115.11 (d, J ¼ 21.0 Hz,
and following procedure B to afford 16{5,6} in 52% yield, light C13), 92.73 (C5), 73.04 (C17), 69.05 (C16), 41.06 (C15), 28.81 (NCH3),
brown solid. IR (KBr), nmax (cm1): 3128, 2870, 1640, 1453, 1430, 22.91 (C18), 10.71 (C19). MS (70 eV, EI): m/z (%) ¼ 371.2 (73) [M]þ,
1284, 796. 1H NMR (400 MHz, DMSO-d6): d 8.94 (s, 1H, NH), 8.36 (s, 312.1 (15) [M-C3H7O]þ, 298.1 (86) [M-C4H9O]þ, 285.1 (100) [M-
1H, C6eCH), 7.62e7.55 (m, 2H, C13-CHx2), 7.47 (t, J ¼ 8.7 Hz, 1H, C5H12O]þ, 158.0 (44) [M-C12H20NFO]þ. HRMS (70 eV, EI): m/z
C14eCH), 3.66 (t, J ¼ 4.8 Hz, 2H, C15eCH2), 3.58 (t, J ¼ 5.3 Hz, 2H, calculated for C19H22N5O2F: 371.1758, [M]þ, found: 371.1755.
C16eCH2), 3.36 (t, J ¼ 6.6 Hz, 2H, C17eCH2), 1.48 (h, J ¼ 7.4, 6.4 Hz,
2H, C18eCH2), 0.82 (t, J ¼ 7.4 Hz, 3H, C19eCH3). 13C NMR 4.1.7.4. 2-Amino-6-(4-fluorophenyl)-8-methyl-4-(pyrrolidin-1-yl)
(100.6 MHz, DMSO-d6): d 156.42, 155.79, 150.86, 135.99 (C6), 135.00 pyrido[2,3-d]pyrimidin-7(8H)-one (17{2,7}). Starting from 16{2,7}
(CeCl), 133.81, 130.83 (C14), 129.23, 128.15 (C13), 113.74, 101.71 (C5), to afford 17{2,7} in 59% yield, white solid. IR (KBr), nmax (cm1):
71.80 (C17), 67.68 (C16), 41.25 (C15), 22.38 (C18), 10.46 (C19). HRMS 3508, 3345, 3233, 2964, 2868, 1638, 1590, 1564, 1524, 1504, 1443,
(70 eV, EI): m/z calculated for C18H19N5O2Cl2: 407.0916, [M]þ, 833, 795. 1H NMR (400 MHz, CDCl3): d 8.01 (s, 1H, C7eCH), 7.61 (dd,
found: 407.0911. J ¼ 8.8, 5.4 Hz, 2H, C12eCHx2), 7.08 (t, J ¼ 8.8 Hz, 2H, C13eCHx2),
4.89 (s, 2H, NH2), 3.82e3.75 (m, 4H, C15eCH2), 2.03e1.96 (m, 4H,
4.1.7. General procedure for the methylation at N8 of pyrido[2,3-d] C16eCH2). 13C NMR (100.6 MHz, CDCl3): d 162.4, 162.1 (d,
pyrimidines 16{x,y} J ¼ 265.8 Hz, C14), 161.0, 160.9, 157.8, 133.8 (C11), 133.3 (C6), 130.4
To a solution of 0.7 mmol of the corresponding 16{x,y} in 20 mL (d, J ¼ 7.8 Hz, C12), 122.2 (C7), 115.1 (d, J ¼ 21.2 Hz, C13), 94.6 (C5),
476 M. Camarasa et al. / European Journal of Medicinal Chemistry 115 (2016) 463e483

51.0 (C15), 29.2 (CH3), 25.8 (C16). HRMS (70 eV, EI): m/z calculated 4.1.9. General procedure for the substitution of the methylthio
for C18H18N5OF: 339.1493, [M]þ, found: 339.1495. group of compounds 19{x} by benzylamines
The corresponding benzylamine (20 mmol) was added to the
solution of the 2-methylthio-substituted pyridopyrimidine (19{x})
4 .1. 7 . 5 . 2 - A m i n o - 6 - ( 2 , 6 - d i ch l o r o p h e n yl ) - 8 - m e t h yl - 4 - ( 2 - (2 mmol) in 14 mL of 2-propanol and the mixture was heated under
propoxyethoxy)pyrido[2,3-d]pyrimidin-7(8H)-one (17{5,9}). microwave irradiation at 170  C for 5 h. Cyclohexane was added to
Starting from 16{5,9} to afford 17{5,9} in 26% yield, white solid. IR the solution and the solid was collected by filtration and washed
(KBr), nmax (cm1): 3336, 2960, 2872, 1658, 1585, 1464, 1429, 1338, with cyclohexane, and Et2O to afford the corresponding (21{x,y}).
801. 1H NMR (400 MHz, DMSO-d6): d 7.62 (s, 1H, C6eCH), 7.54 (d,
J ¼ 8.0 Hz, 2H, CHeC13x2), 7.42 (dd, J ¼ 8.8, 7.3 Hz, 1H, CHeC14), 4.1.9.1. 2-(benzylamino)-6-(4-fluorophenyl)-5,6-dihydropyrido[2,3-
7.38 (s, 2H, NH2), 4.53e4.45 (m, 2H, C15eCH2), 3.77e3.68 (m, 2H, d]pyrimidine-4,7(3H,8H)-dione (21{2,8}). Starting from 19{2} and
C16eCH2), 3.59 (m, 2H, C17e CH2), 3.55 (s, 3H, NCH3), 1.46 (h, benzylamine to afford 21{2,8} in 53% yield, white solid. IR (KBr),
J ¼ 7.1 Hz, 2H, C18eCH2), 0.79 (t, J ¼ 7.4 Hz, 3H, C19eCH3). 13C NMR nmax (cm1): 3423, 3180, 3068, 2958, 1689, 1644, 1612, 1513, 1229,
(100.6 MHz, DMSO-d6): d 166.6, 162.4, 160.6, 157.2, 135.2, 134.8, 836, 582. 1H NMR (400 MHz, TFA-d): d 7.42e7.30 (m, 5H, CHArx5),
132.8 (C6), 130.4 (C14), 128.0 (C13), 120.6, 109.6, 92.0 (C5), 71.9 7.22 (dd, J ¼ 7.8, 5.2 Hz, 2H, CHeC12x2), 7.05 (t, J ¼ 8.5 Hz, 2H,
(C17), 67.9 (C16), 65.9 (C15), 28.2 (NCH3), 22.4 (C18), 10.4 (C19). CHeC13x2), 4.75 (s, 2H, CH2eC15), 4.05 (dd, J ¼ 11.3, 7.3 Hz, 1H,
HRMS (ESIeTOF): m/z calculated for C19H21Cl2N4O3: 223.0983, CHeC7), 3.24 (dd, J ¼ 16.8, 7.4 Hz, 1H, C6eCH2), 3.03 (dd, J ¼ 16.8,
[MþH]þ, found: 423.0985. Anal. Calcd. (%) for C19H20Cl2N4O3: C, 11.4 Hz, 1H, C6eCH2). 13C NMR (100.6 MHz, TFA-d): d 179.9, 168.9,
53.91; H, 4.76; N, 13.24. Found: C, 53.97; H, 4.94; N, 12.93. 166.5 (d, J ¼ 235.5 Hz, C14) 153.9, 150.6, 136.2, 134.7 (d, J ¼ 3.4 Hz,
C11), 134.0 (d, J ¼ 8.5 Hz, C12) 133.7 (CAr), 133.7 (CAr), 131.6 (CAr),
120.3 (d, J ¼ 22.7 Hz, C13) 96.2 (C5), 50.7 (C15), 49.7 (C7), 27.8 (C6).
4.1.7.6. 4-Amino-6-(2,6-dichlorophenyl)-8-methylpyrido[2,3-d]py- HRMS (70 eV, EI): m/z calculated for C20H17N4O2F: 324.0183, [M]þ,
rimidine-7(8H)-one (17{5,4}). Starting from 9{5,4} [25] and found: 324.0181. Anal. Calcd. (%) for C20H17FN4O2: C, 65.93; H, 4.70;
following procedure A described in 4.1.6. to afford 16{5,4} in 97% N, 15.38. Found: C, 65.53; H, 4.47; N, 15.49.
yield, white solid. IR (KBr), nmax (cm1): 3457, 3329, 3182, 2791,
1615, 1545, 1494, 1462, 1429, 1400, 1327, 1290, 1191, 936, 802, 775. 4.1.9.2. 2-(benzylamino)-6-(2,6-dichlorophenyl)-5,6-dihydropyrido
1
H NMR (400 MHz, DMSO-d6): d 12.17 (br s, 1H), 7.98 (s, 1H), 7.54 (d, [2,3-d]pyrimidine-4,7(3H,8H)-dione (21{5,8}). Starting from 19{5}
J ¼ 8.0 Hz, 2H), 7.40 (dd, J ¼ 8.6, 7.6 Hz, 1H), 7.17 (br s, 2H), 6.81 (br s, and benzylamine to afford 21{5,8} in 36% yield, white solid. IR (KBr),
2H). 13C NMR (100.6 MHz, DMSO-d6): d 163.1, 161.9, 161.5, 156.6, nmax (cm1): 3263, 3181, 3066, 2934, 1686, 1639, 1613, 1597, 1545,
136.3, 135.5, 134.8, 130.2, 128.0, 119.6, 90.5. MS (ESI-TOF): m/z 1520, 1374, 1289, 770. 1H NMR (400 MHz, DMSO-d6): d 10.57 (s, 1H,
(%) ¼ 322.0 (100) [MþH]þ, 305.0 (9) [M-NH2]þ, 286.0 (9) [M-Cl]þ, NH), 10.27 (s, 1H, NH), 7.50 (ddd, J ¼ 16.6, 8.1, 1.3 Hz, 2H,
252.1 (7) [MþHeCl2]þ. HRMS (ESI-TOF): m/z calculated for CHeC13x2), 7.40e7.31 (m, 5H, CH-Ar), 7.27 (d, J ¼ 7.0 Hz, 1H, CH-
C13H10N5OCl2: 322.0257 [MþH]þ, found: 322.0256. 16{5,4} was Ar), 6.99 (s, 1H, NH-Bz), 4.53 (dd, J ¼ 13.4, 8.9 Hz, 1H, CHeC7),
converted to 17{5,4} in 70% yield, white solid. IR (KBr), nmax (cm1): 4.47 (d, J ¼ 6.1 Hz, 2H), 2.84e2.64 (m, 2H, CH2eC6). 13C NMR
3347, 3212, 2925, 1627, 1607, 1568, 1546, 1462, 1430, 1394, 1360, (100.6 MHz, DMSO-d6): d 169.7, 161.6, 155.5, 153.7, 139.1, 135.4,
1328, 1202 802. 1H NMR (400 MHz, DMSO-d6): d 7.99 (s, 1H), 7.55 (d, 135.2, 134.7, 129.7, 129.7, 128.3, 128.3, 127.6, 127.0, 87.1 (C5), 43.4
J ¼ 7.9 Hz, 2H), 7.41 (dd, J ¼ 8.6, 7.5 Hz, 1H), 7.15 (br s, 2H), 6.67 (br s, (C15), 43.4 (C7), 22.4 (C6). MS (70 eV, EI): m/z (%) ¼ 414.0 (100),
2H), 3.50 (s, 3H). 13C NMR (100.6 MHz, DMSO-d6): d 162.8, 162.1, [M]þ, 379.1 [M-Cl]þ, 228.9 (34.0) [C12H12N4O]þ, 105.8 (62.6)
160.6, 156.7, 135.6, 135.5, 134.1, 131.0, 128.0, 118.4, 90.5, 28.0. MS [C7H8N]þ, 90.9 (100) [C7H7]þ. HRMS (70 eV, EI): m/z calculated for
(ESI-TOF): m/z (%) ¼ 336.0 (100) [MþH]þ, 286.0 (3) [MþH-CH3Cl]þ, C20H16N4O2Cl2: 414.0650, [M]þ, found: 414.0650. Anal. Calcd. (%)
266.1 (5) [MþH-Cl2]þ. HRMS (ESIeTOF): m/z calculated for for C20H16Cl2N4O2: C, 57.84; H, 3.88; N, 13.49. Found: C, 57.71; H,
C14H12Cl2N5O: 336.0413, [MþH]þ, found: 336.0424. 3.78; N, 13.47.

4.1.9.3. 2-((4-fluorobenzyl)amino)-6-(4-fluorophenyl)-5,6-
4.1.8. 6-(2,4-dichlorophenyl)-2-(methylthio)-5,6-dihydropyrido dihydropyrido[2,3-d]pyrimidine-4,7(3H,8H)-dione (21{2,9}).
[2,3-d]pyrimidine-4,7(3H,8H)-dione (19{11}) Starting from 19{2} and 4-flurobenzylamine to afford 21{2,9} in 40%
2-(2,4-dichlorophenyl)acrylate (5{11}) (4.00 mmol) was dis- yield, white solid. IR (KBr), nmax (cm1): 3424, 3248, 3181, 2931,
solved in 2-propanol (20 mL), 6-amino-2-(methylthio)pyrimidin- 1637, 1611, 1513, 1230, 834. 1H NMR (400 MHz, TFA-d): d 7.31 (dd,
4(3H)-one (52) (5.60 mmol) and potassium carbonate (4.00 mmol) J ¼ 8.6, 5.0 Hz, 2H, C17-CHx2), 7.19 (dd, J ¼ 8.7, 5.0 Hz, 2H, C12-
were added into the solution. The mixture was heated under mi- CHx2), 7.06e7.00 (m, 4H, C18-CHx2, C13-CHx2), 4.70 (s, 2H,
crowave irradiation at 170  C for 3 h. Then, the solvent was C15eCH2), 4.02 (dd, J ¼ 11.2, 7.2 Hz, 1H, C7eCH), 3.22 (dd, J ¼ 16.8,
removed under reduced pressure, water was added to the crude 7.4 Hz, 1H, C6eCH2), 3.00 (dd, J ¼ 16.8, 11.4 Hz, 1H, C6eCH2). 13C
and this was neutralized with a 2 M solution of HCl. The solid was NMR (100.6 MHz, DMSO-d6): d 179.9, 168.0 (d, J ¼ 248.8 Hz, C14),
collected by filtration and washed with water, and Et2O to afford 19 167.6 (d, J ¼ 247.7 Hz), 165.3, 154.0, 150.7, 134.7 (d, J ¼ 3.5 Hz, C11),
{11} in 80% yield, white solid. IR (KBr): n (cm1): 3404, 3131, 2868, 134.0 (d, J ¼ 8.5 Hz, C12), 133.7 (C17), 132.2 (C16), 120.5 (d,
1694, 1633, 1557, 1505, 1479, 1459, 1374, 1285, 1241, 1151, 1103, 964, J ¼ 24.5 Hz), 120.4 (d, J ¼ 22.3 Hz), 96.1 (C5), 50.0 (C15), 49.7 (C7),
823, 576. 1H NMR (400 MHz, DMSO-d6): d 10.73 (s, 1H), 7.63 (d, 27.8 (C6). HRMS (70 eV, EI): m/z calculated for C20H16N4O2F2:
J ¼ 1.9 Hz, 1H), 7.43 (dd, J ¼ 8.4, 2.0 Hz, 1H), 7.40 (d, J ¼ 8.3 Hz, 1H), 382.1241, [M]þ, found: 382.1241.
4.19 (dd, J ¼ 12.8, 7.7 Hz, 1H, C7eCH), 2.89 (dd, J ¼ 16.2, 7.7 Hz, 1H,
C6eCH2), 2.75 (dd, J ¼ 16.1, 12.8 Hz, 1H, C6eCH2), 2.50 (s, 3H, SCH3). 4.1.10. General procedure for oxidation of 2-(methylthio)-5,6-
13
C NMR (100.6 MHz, DMSO-d6): d 170.25, 161.8, 154.4, 135.9 (C12), dihydropyrido[2,3-d]pyrimidine-4,7(3H,8H)-diones (19{x}) to
134.3 (C14), 132.6(C16), 132.0 (C12), 129.0 (C13), 127.6 (C15), 95.0 methylsulfones 20{x}
(C5), 43.3 (C7), 23.7 (C6), 12.8 (SCH3). Anal. Calcd. (%) for m-CPBA (18 mmol) was suspended in 20 mL of DMF and was
C14H11Cl2N3O2S: C, 47.20; H, 3.11; N, 11.80; S, 9.00. Found: C, 47.07; added dropwise into a solution of the 2-methylthio-substituted
H, 2.97; N, 11.96; S, 9.01. pyridopyrimidine (19{x}) (6 mmol) in 20 mL of DMF a 0  C. The
M. Camarasa et al. / European Journal of Medicinal Chemistry 115 (2016) 463e483 477

mixture was stirred overnight at room temperature. Then the sol- 4.85 (dd, J ¼ 10.2, 7.9 Hz, 1H, C7eH), 3.33 (s, 3H, SCH3), 3.23e3.13
vent was removed under reduced pressure and the residue was (m, 2H, C6eCH2). 13C NMR (100.6 MHz, DMSO-d6): d 171.5, 167.1,
suspended in EtOEt and the solid was collected by filtration and 162.7, 158.8, 135.0, 133.6, 131.1, 129.0 (C16), 127.8 (C12), 126.2 (C17),
washed with water, and EtOEt to afford the corresponding 2- 125.7 (C18), 125.5 (C14, C13), 124.0 (C19), 100.6 (C5), 41.5 (C7), 39.1
methylsulfonyl-substituted pyrido[2,3-d]pyrimidine (20{x}). (SCH3), 25.0 (C6). Anal. Calcd. (%) for C18H15N3O4S: C, 58.53; H, 4.09;
N, 11.38; S, 8.68. Found: C, 58.47; H, 4.18 N, 11.57; S, 8.69.
4.1.10.1. 2-(methylsulfonyl)-6-phenyl-5,6-dihydropyrido[2,3-d]py-
rimidine-4,7(3H,8H)-dione (20{1}). Starting from 19{1} to afford 20 4 .1.10 . 6 . 6 - ( 2 , 4 - d i c h l o r o p h e n yl ) - 2 - ( m e t hyl s u l f o n yl ) - 5 , 6 -
{1} in 74% yield, white solid. IR (KBr), nmax (cm1): 3429, 3200, dihydropyrido[2,3-d]pyrimidine-4,7(3H,8H)-dione (20{11}).
3136, 3022, 2919, 2866, 1691, 1643, 1612, 1486, 1341, 1235, 1165, Starting from 19{11} to afford 20{11} in 70% yield, white solid. IR
1134, 699. 1H NMR (400 MHz, DMSO-d6): d 13.29 (s, 1H, NH), 11.32 (KBr), nmax(cm1): 3432, 3279, 3040, 2925, 2868, 1651, 1492, 1329,
(s, 1H, NH), 7.36e7.31 (m, 2H, C13-CHx2), 7.30e7.22 (m, 3H, C12- 1276, 1231, 1156, 1132, 944, 766, 572, 529. 1H NMR (400 MHz,
CHx2, C14eCH), 4.00 (dd, J ¼ 9.4, 7.3 Hz, 1H, C7eCH), 3.31 (s, 3H, DMSO-d6): d 13.34 (s, 1H, NH), 11.42 (s, 1H, NH), 7.65 (dd, J ¼ 1.5,
SCH3), 3.14e3.00 (m, 2H, C6eCH2). 13C NMR (100.6 MHz, DMSO- 0.9 Hz, 1H), 7.46 (s, 1H, C13eCH), 7.46 (d, J ¼ 0.9 Hz, 1H), 4.38 (dd,
d6): d 171.4, 167.1, 162.6, 158.6, 138.4, 128.5, 128.0, 127.1 (C12), 100.4 J ¼ 11.6, 9.3 Hz, 1H, C7eCH), 3.31 (s, 3H, SCH3), 3.07e2.81 (s, 2H,
(C5), 44.8 (C7), 39.1 (SCH3), 24.7 (C6). Anal. Calcd. (%) for C6eCH2). 13C NMR (100.6 MHz, DMSO-d6): d 169.9, 167.0, 162.7,
C14H13N3O4S: C, 52.66; H, 4.10; N, 13.16; S, 10.04. Found: C, 52.32; H, 158.6, 135.3, 134.4, 132.7, 132.1, 129.0 (C13), 127.6, 100.2 (C5), 42.6
4.16 N, 13.02; S, 10.34. (C7), 28.5 (SCH3), 23.8 (C6). Anal. Calcd. (%) for C14H11Cl2N3O4S: C,
43.31; H, 2.86; N, 10.82; S, 8.26. Found: C, 43.48; H, 2.66; N, 10.91; S,
4.1.10.2. 6-(4-fluorophenyl)-2-(methylsulfonyl)-5,6-dihydropyrido 8.11.
[2,3-d]pyrimidine-4,7(3H,8H)-dione (20{2}). Starting from 19{2} to
afford 20{2} in 60% yield, white solid. IR (KBr): n (cm1): 3393, 4.1.11. General procedure for the substitution of the methylsulfonyl
3036, 2867, 1635, 1607, 1513, 1490, 1337, 1233, 1160, 839, 767, 541. 1H group of pyridopyrimidines 20{x} by arylamines
NMR (400 MHz, DMSO-d6): d 13.28 (s, 1H, NH), 11.33 (s, 1H, NH), The corresponding arylamine (10 mmol) was added to a solution
7.30 (dd, J ¼ 8.7, 5.8 Hz, 2H, C12-CHx2), 7.17 (t, J ¼ 8.9 Hz, 2H, C13- of the 2-methylsulfonyl-substituted pyridopyrimidine (20{x})
CHx2), 4.03 (dd, J ¼ 10.2, 7.4 Hz, 1H, C7eCH), 3.31 (s, 3H, SCH3), (2 mmol) in 14 mL 2-propanol and the mixture was heated under
3.14e2.94 (m, 2H, C6eCH2). 13C NMR (100.6 MHz, DMSO-d6): microwave irradiation at 170  C for 5 h. Cyclohexane was added to
d 171.4, 167.0, 162.7, 161.3 (d, J ¼ 243.2 Hz, C14), 158.7, 134.6 (d, the solution and the solid obtained was collected by filtration and
J ¼ 3.1 Hz, C11), 130.2 (d, J ¼ 8.1 Hz, C12), 115.2 (d, J ¼ 21.3 Hz, C13), washed with cyclohexane and Et2O to afford the corresponding 21
100.4 (C5), 44.1 (C7), 29.5 (SCH3), 24.8 (C6). HRMS (70 eV, EI): m/z {x,y}
calculated for C14H12N3O4FS: 337.0533, [M]þ, found: 337.0533.
Anal. Calcd. (%) for C14H12FN3O4S: C, 49.85; H, 3.59; N, 12.46; S, 9.51. 4.1.11.1. 6-Phenyl-2-(phenylamino)-5,6-dihydropyrido[2,3-d]pyrimi-
Found: C, 49.68; H, 3.84; N, 12.70; S, 9.18. dine-4,7(3H,8H)-dione (21{1,10}). Starting from 20{1} and aniline to
afford 21{1,10} in 82% yield, white solid. IR (KBr), nmax(cm1): 3401,
4 .1.10 . 3 . 6 - ( 2 , 6 - d i fl u o r o p h e n yl ) - 2 - ( m e t h yl s u l f o n yl ) - 5 , 6 - 3058, 1615, 1654, 1462, 750, 395. 1H NMR (400 MHz, DMSO-d6):
dihydropyrido[2,3-d]pyrimidine-4,7(3H,8H)-dione (20{4}). d 10.48 (s, 2H, NHx2), 8.80 (s, 1H, NH), 7.73e7.66 (m, 2H, C16-CHx2),
Starting from 19{4} to afford 20{4} in 60% yield, white solid. IR 7.34e7.22 (m, 7H, C17-CHx2, C12-CHx2, C13-CHx2, C14eCH), 7.02
(KBr): n (cm1): 3432, 3219, 3027, 2923, 1704, 1649, 1612, 1474, (tt, J ¼ 7.3, 1.2 Hz, 1H, C8eCH), 3.82 (dd, J ¼ 8.5, 7.1 Hz, 1H, C7eCH),
1459, 1339, 1235, 1164, 1134, 1010, 783, 542. 1H NMR (400 MHz, 2.92e2.72 (m, 2H, C6eCH2). 13C NMR (100.6 MHz, DMSO-d6):
DMSO-d6): d 13.33 (s, 1H), 11.41 (s, 1H), 7.44 (tt, J ¼ 8.4, 6.6 Hz, 1H, d 172.4, 139.7, 139.1, 129.2, 128.8, 128.8, 128.5, 128.4, 127.3, 123.0
C14eCH), 7.14 (t, J ¼ 8.8 Hz, 2H, C13-CHx2), 4.46 (dd, J ¼ 14.0, 7.6 Hz, (C18), 119.8 (C16), 114.3, 90.7 (C5), 46.2 (C7), 25.0 (C6). HRMS
1H, C7eCH), 3.31 (s, 3H, SCH3), 3.10 (dd, J ¼ 16.5, 7.7 Hz, 1H, (70 eV, EI): m/z calculated for C19H16N4O2: 332.1273, [M]þ, found:
C6eCH2), 2.94e2.82 (m, 1H, C6eCH2). 13C NMR (100.6 MHz, DMSO- 332.1275.
d6): d 169.6, 167.0, 162.7, 162.0 (d, J ¼ 8.3 Hz, C12), 159.6 (d,
J ¼ 8.2 Hz, C12), 158.7, 130.0 (t, J ¼ 10.6 Hz, C11), 114.5 (t, J ¼ 18.6 Hz, 4.1.11.2. 6-(4-fluorophenyl)-2-(phenylamino)-5,6-dihydropyrido[2,3-
C14), 111.8 (dd, J ¼ 22.3, 3.1 Hz, C13), 100.0 (C5), 39.2 (SCH3), 35.3 d]pyrimidine-4,7(3H,8H)-dione (21{2,10}). Starting from 20{2} and
(C7), 23.4 (C6). Anal. Calcd. (%) for C14H11F2N3O4S: C, 47.32; H, 3.12; aniline to afford 21{2,10} in 94% yield, white solid. IR (KBr), nmax
N, 11.83; S, 9.02. Found: C, 47.14; H, 3.19 N, 11.72; S, 9.28. (cm1): 3400, 3186, 3049, 1655, 1614, 1511, 1461, 1232. 1H NMR
(400 MHz, DMSO-d6): d 10.47 (s, 2H, 2xNH), 9.25 (s, 1H, NH-Ph),
4 .1.10 . 4 . 6 - ( 2 , 6 - d i ch l o r o p h e nyl ) - 2 - ( m e t hyl s u l fo nyl ) - 5 , 6 - 7.79e7.67 (m, 2H, 2xCHPh), 7.33e7.24 (m, 4H, 2xCHPh þ C12-CHx2),
dihydropyrido[2,3-d]pyrimidine-4,7(3H,8H)-dione (20{5}). 7.15 (t, J ¼ 8.9 Hz, 2H, C13-CHx2), 7.04e6.98 (m, 1H), 3.85 (dd,
Starting from 19{5} to afford 20{5} in 98% yield, white solid. 1H NMR J ¼ 9.6, 7.0 Hz, 1H), 2.92e2.70 (m, 2H, C6eCH2). 13C NMR
(400 MHz, DMSO-d6): d 11.35 (s, 1H, NH), 7.53 (dd, J ¼ 12.3, 8.1 Hz, (100.6 MHz, DMSO-d6): d 172.0, 161.8, 161.3 (d, J ¼ 242.8 Hz, C14),
2H, C13-CHx2), 7.39 (t, J ¼ 8.1 Hz, 1H, C14eCH), 4.87 (dd, J ¼ 12.9, 155.6, 151.1, 138.8, 135.5 (d, J ¼ 3.1 Hz, C11), 130.1 (d, J ¼ 8.1 Hz, C12),
9.2 Hz, 1H, C7eCH), 3.31 (s, 3H, SCH3), 3.06 (dd, J ¼ 17.1, 9.4 Hz, 1H, 128.8, 122.6, 119.5, 115.2 (d, J ¼ 21.1 Hz, C13) 90.4 (C5), 45.1 (C7),
C6eCH2), 2.99 (dd, J ¼ 17.0, 13.5 Hz, 1H, C6eCH2). 24.7 (C6). HRMS (70 eV, EI): m/z calculated for C19H15N4O2F:
350.1179, [M]þ, found: 350.1179.
4.1.10.5. 2-(methylsulfonyl)-6-(naphthalen-1-yl)-5,6-dihydropyrido
[2,3-d]pyrimidine-4,7(3H,8H)-dione (20{8}). Starting from 19{8} to 4.1.11.3. 6-(4-fluorophenyl)-2-((4-fluorophenyl)amino)-5,6-
afford 20{8} in 68% yield, white solid. IR (KBr), nmax(cm1): 3432, dihydropyrido[2,3-d]pyrimidine-4,7(3H,8H)-dione (21{2,11}).
3211, 3022, 1690, 1641, 1614, 1487, 1345, 1225, 1167, 1136, 775. 1H Starting from 20{2} and 4-fluoroaniline to afford 21{2,11} in 67%
NMR (400 MHz, DMSO-d6): d 13.24 (s, 1H, NH), 11.44 (s, 1H, NH), yield, white solid. IR (KBr), nmax (cm1): 3409, 3171, 2958, 1645,
8.13e8.07 (m, 1H, C19eCH), 8.00e7.94 (m, 1H, C16eCH), 7.87 (d, 1603, 1512, 1242, 836. 1H NMR (400 MHz, DMSO-d6): d 10.58 (s, 1H,
J ¼ 8.2 Hz, 1H, C12eCH), 7.58e7.51 (m, 2H, C17eCH, C18eCH), 7.48 NH), 10.48 (s, 1H, NH), 8.88 (s, 1H, NH), 7.71 (dd, J ¼ 9.1, 4.9 Hz, 2H,
(t, J ¼ 7.2 Hz, 1H, C13eCH), 7.40 (dd, J ¼ 7.2, 1.2 Hz, 1H, C14eCH), C16-CHx2), 7.28 (dd, J ¼ 8.7, 5.6 Hz, 2H, C12-CHx2), 7.13 (m, 4H,
478 M. Camarasa et al. / European Journal of Medicinal Chemistry 115 (2016) 463e483

C13-CHx2, C17-CHx2), 3.84 (dd, J ¼ 9.5, 7.1 Hz, 1H, C7eCH), 2.86 1H, C23eCH), 4.64 (dd, J ¼ 9.1, 7.6 Hz, 1H, C7eCH), 3.00 (dd, J ¼ 16.1,
(dd, J ¼ 16.0, 7.1 Hz, 1H, C6eCH), 2.73 (dd, J ¼ 16.0, 9.6 Hz, 1H, 7.7 Hz, 1H, C6eCH2), 2.85 (dd, J ¼ 16.1, 9.1 Hz, 1H, C6eCH2). 13C NMR
C6eCH). 13C NMR (100.6 MHz, DMSO-d6): d 171.9, 161.2 (d, (100.6 MHz, DMSO-d6): d 172.0, 171.0, 162.9, 155.4, 138.7, 135.9,
J ¼ 242.6 Hz, C14), 157.8 (d, J ¼ 239.4 Hz, C18), 156.2, 155.4, 154.8, 133.7, 131.0, 128.8 (C22,C16), 127.5 (C14), 126.1, 125.6, 125.4 (C13),
135.4 (d, J ¼ 3.1 Hz, C11), 135.0 (C15), 130.0 (d, J ¼ 8.1 Hz, C12), 121.3 125.1 (C12), 123.9 (C19), 122.6 (C23), 119.4 (C21), 90.3 (C5), 42.5
(d, J ¼ 7.6 Hz, C16), 115.2 (d, J ¼ 17.7 Hz, C13), 115.0 (d, J ¼ 16.7 Hz, (C7), 24.8 (C6). HRMS (70 eV, EI): m/z calculated for C23H18N4O2:
C17), 90.3 (C5), 45.0 (C7), 24.6 (C6). HRMS (70 eV, EI): m/z calcu- 382.1430, [M]þ, found: 382.1429.
lated for C19H14N4O2F2: 368.1085, [M]þ, found: 368.1081.

4.1.11.4. 2-((4-bromophenyl)amino)-6-(4-fluorophenyl)-5,6- 4.1.12. 6-(4-fluorophenyl)-2-phenoxy-5,6-dihydropyrido[2,3-d]


dihydropyrido[2,3-d]pyrimidine-4,7(3H,8H)-dione (21{2,12}). pyrimidine-4,7(3H,8H)-dione (21{2,14})
Starting from 20{2} and 4-bromoaniline to afford 21{2,12} in 74% Phenol (4 mmol) was heated at 60  C in a mixture of sodium
yield, white solid. IR (KBr), nmax (cm1): 3400, 3163, 2963, 1643, methoxyde (4 mmol) in 2-propanol (20 mL) for 20 min. 20{2}
1600, 1511, 1488, 1236, 835, 795, 771. 1H NMR (400 MHz, DMSO-d6): (2 mmol) was added to the mixture and it was heated under mi-
d 10.53 (s, 1H, NHx2), 8.93 (s, 1H, NH), 7.69 (d, J ¼ 8.9 Hz, 2H, crowave irradiation at 170  C for 3 h. The solvent was eliminated
C17eCHx2), 7.43 (d, J ¼ 8.8 Hz, 2H, C16eCHx2), 7.28 (dd, J ¼ 8.7, under reduced pressure, water was added to the residue and the
5.6 Hz, 2H, C12eCHx2), 7.15 (t, J ¼ 8.9 Hz, 2H, C13eCHx2), 3.85 (dd, mixture was neutralized with HCl 2 M. The solid was collected by
J ¼ 9.6, 7.1 Hz, 1H, C7eCH), 2.91e2.69 (m, 2H, C6eCH2). 13C NMR filtration, washed with cyclohexane and Et2O to afford 21{2,14} in
(100.6 MHz, DMSO-d6): d 171.9, 161.2 (d, J ¼ 242.8 Hz, C14), 158.5, 72% as a white solid. IR (KBr), nmax (cm1): 3408, 3129, 2902, 1692,
158.2, 155.4, 138.2 (C15), 135.4 (d, J ¼ 3.1 Hz, C11), 131.4 (C17), 130.0 1632, 1577, 1513, 1469, 1337, 1220. 1H NMR (400 MHz, DMSO-d6):
(d, J ¼ 8.1 Hz, C12), 121.4 (C16), 115.1 (d, J ¼ 21.3 Hz, C13), 114.0 d 12.58 (s, 1H, NH), 10.48 (s, 1H, NH), 7.44 (dd, J ¼ 8.6, 7.3 Hz, 2H,
(C18), 90.6 (C5), 45.0 (C7), 24.6 (C6). HRMS (70 eV, EI): m/z calcu- C12eCHx2), 7.28e7.25 (m, 5H, C15-CHx2, C16eCHx2, C17eCH),
lated for C19H14N4O2FBr: 428.0284, [M]þ, found: 428.0287. 7.15 (t, J ¼ 8.9 Hz, 2H, C13eCHx2), 3.84 (dd, J ¼ 10.0, 7.1 Hz, 1H, C7),
2.88 (dd, J ¼ 16.2, 7.1 Hz, 1H, C6eCH), 2.76 (dd, J ¼ 16.2, 10.2 Hz, 1H,
4.1.11.5. 6-(4-fluorophenyl)-2-((4-methoxyphenyl)amino)-5,6- C6eCH). 13C NMR (100.6 MHz, DMSO-d6): d 171.5, 163.2, 161.2 (d,
dihydropyrido[2,3-d]pyrimidine-4,7(3H,8H)-dione (21{2,13}). J ¼ 242.7 Hz, C14), 157.6, 155.6, 151.6, 135.1 (d, J ¼ 3.1 Hz, C11), 130.1
Starting from 20{2} and 1.2 equivalents of 4-methoxyaniline to (d, J ¼ 8.9 Hz, C12), 129.8 (C16), 125.8 (C17), 121.6 (C15), 115.1 (d,
afford 21{2,13} in 79% yield, white solid. IR (KBr), nmax (cm1): 3404, J ¼ 21.3 Hz, C13), 93.7 (C5), 44.7 (C7), 24.5 (C6). HRMS (70 eV, EI): m/
3148, 2813, 1656, 1615, 1509, 1457, 1228, 844. 1H NMR (400 MHz, z calculated for C19H14N3O3F: 351.1019, [M]þ, found: 351.1033.
DMSO-d6): d 10.40 (s, 1H, NH), 8.62 (s, 1H, NH), 7.56 (d, J ¼ 9.0 Hz,
2H, C17-CHx2), 7.28 (dd, J ¼ 8.7, 5.6 Hz, 2H, C12eCHx2), 7.15 (t,
J ¼ 8.9 Hz, 2H, C13eCHx2), 6.87 (d, J ¼ 9.0 Hz, 2H, C16eCHx2), 3.83 4.1.13. 4-((1H-benzo[d] [1,2,3]triazol-1-yl)oxy)-6-(4-fluorophenyl)-
(dd, J ¼ 9.5, 7.0 Hz, 1H, C7eCH), 3.73 (s, 3H, OCH3), 2.89e2.69 (m, 2-(phenylamino)-5,6-dihydropyrido[2,3-d]pyrimidin-7(8H)-one
2H, C6eCH2). 13C NMR (100.6 MHz, DMSO-d6): d 171.9, 161.4, 161.1 (22{2,10})
(d, J ¼ 242.8 Hz, C14), 156.2, 155.1, 154.8, 135.4 (d, J ¼ 3.1 Hz, C11), Starting from compound 21{2,10} and using procedure
131.55, 130.0 (d, J ¼ 8.1 Hz, C12), 121.5 (C17), 115.0 (d, J ¼ 21.3 Hz, described in 4.1.2. to afford 22{2,10} in 88% yield, white solid. 1H
C13), 113.9 (C16), 89.8 (C5) 55.2 (OMe), 45.1 (C7), 24.6 (C6). HRMS NMR (400 MHz, DMSO-d6): d 11.19 (s, 1H, NH), 9.46 (s, 1H, NH), 8.23
(70 eV, EI): m/z calculated for C20H17N4O3F: 380.1285, [M]þ, found: (d, J ¼ 8.5 Hz, 1H), 7.76 (dt, J ¼ 8.4, 0.9 Hz, 1H), 7.69e7.64 (m, 1H),
380.1286. 7.56 (ddd, J ¼ 8.2, 6.9, 1.1 Hz, 1H), 7.43 (dd, J ¼ 8.7, 5.5 Hz, 2H,
C12eCHx2), 7.23 (t, J ¼ 8.9 Hz, 2H, C13eCHx2), 6.95e6.90 (m, 2H),
4.1.11.6. 6-(2,6-difluorophenyl)-2-(phenylamino)-5,6-dihydropyrido 6.85e6.79 (m, 2H), 6.75e6.77 (m, 1H, C18eCH), 4.19 (dd, J ¼ 11.0,
[2,3-d]pyrimidine-4,7(3H,8H)-dione (21{4,10}). Starting from 20{4} 7.0 Hz, 1H, C7eCH), 3.41e3.29 (m, 2H, C6eCH2). HRMS (70 eV, EI):
and aniline to afford 21{4,10} in 83% yield, white solid. IR (KBr), m/z calculated for C25H18N7O2F: 467.1506, [M]þ, found: 467.1508.
nmax(cm1): 3423, 3231, 3060, 2931, 1701, 1643, 1627, 1593, 1469,
1235, 1009, 786. 1H NMR (400 MHz, DMSO-d6): d 10.58 (s, 2H,
2xNH), 8.83 (s, 1H, NH), 7.74e7.68 (m, 2H, C15eCHx2), 7.46e7.40 4.1.14. 4-((1H-benzo[d] [1,2,3]triazol-1-yl)oxy)-6-(4-fluorophenyl)-
(m, 1H, C14eCH), 7.34e7.27 (m, 2H, C16eCHx2), 7.13 (t, J ¼ 8.8 Hz, 2-phenoxy-5,6-dihydropyrido[2,3-d]pyrimidin-7(8H)-one (22
2H, C13eCHx2), 7.03 (t, J ¼ 7.4 Hz, 1H, C17eCH), 4.19 (dd, J ¼ 14.1, {2,14})
7.7 Hz, 1H, C7eCH), 2.90 (dd, J ¼ 15.7, 7.8 Hz, 1H, C6eCH2), Starting from compound 21{2,14} and using procedure
2.53e2.55 (m, 1H, C6eCH2). 13C NMR (100.6 MHz, DMSO-d6): described in 4.1.2. to afford 22{2,14} in 72% yield, white solid. IR
d 170.0, 162.6, 160.8 (d, J ¼ 246.7 Hz, C12), 160.7 (d, J ¼ 246.8 Hz, (KBr), nmax (cm1): 3061, 2977, 2863, 1714, 1635, 1581, 1512, 1412,
C12), 155.5, 138.6, 129.7 (t, J ¼ 10.3 Hz, C14), 128.8 (C15), 122.7 (C17), 1364, 1219, 1099, 746. 1H NMR (400 MHz, DMSO-d6): d 11.50 (s, 1H,
119.4 (C16), 115.2 (t, J ¼ 18.6 Hz, C11), 113.9, 111.7 (d, J ¼ 22.4, 3.2 Hz, NH), 8.07 (dt, J ¼ 8.4, 0.9 Hz, 1H, C22), 7.70e7.60 (m, 2H, C19eCH,
C13), 90.1 (C5), 36.4 (C7), 23.3 (C6). HRMS (70 eV, EI): m/z calcu- C20eCH), 7.53e7.49 (m, 1H, C21eCH), 7.41 (dd, J ¼ 8.7, 5.5 Hz, 2H,
lated for C19H14N4O2F2: 368.1085, [M]þ, found: 368.1087. C12eCHx2), 7.22 (t, J ¼ 8.9 Hz, 2H, C13eCHx2), 7.11e7.00 (m, 3H,
C18eCH, C17eCHx2), 6.84e6.78 (m, 2H, C16eCHx2), 4.20 (dd,
4.1.11.7. 6-(Naphthalen-1-yl)-2-(phenylamino)-5,6-dihydropyrido J ¼ 11.5, 7.1 Hz, 1H, C7eCH), 3.44e3.34 (m, 2H, C6eCH2). 13C NMR
[2,3-d]pyrimidine-4,7(3H,8H)-dione (21{8,10}). Starting from 20{8} (100.6 MHz, DMSO-d6): d 171.5, 165.9, 162.2, 162.1, 161.4 (d,
and aniline to afford 21{8,10} in 67% yield, white solid. IR (KBr), J ¼ 243.5 Hz, C14), 151.6, 142.5, 134.3 (d, J ¼ 3.1 Hz, C11), 130.5 (d,
nmax(cm1): 3430, 3167, 3054, 1644, 1595, 1579, 1540, 1495, 1235, J ¼ 8.2 Hz, C12), 129.1 (C20), 129.1 (C17), 128.3, 125.1 (C21), 125.0
777. 1H NMR (400 MHz, DMSO-d6): d 10.64 (s, 1H, NH), 8.86 (s, 1H, (C18), 120.8 (C16), 119.9 (C22), 115.2 (d, J ¼ 21.3 Hz, C13), 109.2
NH), 8.09 (d, J ¼ 7.7 Hz, 1H, C19eCH), 7.98e7.93 (m, 1H, C16eCH), (C19), 92.9 (C5), 44.3 (C7), 24.0 (C6). HRMS (70 eV, EI): m/z calcu-
7.85 (d, J ¼ 8.1 Hz, 1H, C14eCH), 7.74 (d, J ¼ 7.7 Hz, 2H, C21eCHx2), lated for C25H17N6O3F: 468.1346, [M]þ, found: 468.1337. Anal. Calcd.
7.60e7.50 (m, 3H, C17eCH, C18eCH), 7.46 (dd, J ¼ 8.2, 7.1 Hz, 1H, (%) for C25H17FN6O3: C, 64.10; H, 3.66; N, 17.94. Found: C, 64.06; H,
C13eCH), 7.41e7.29 (m, 3H, C12eCH, C22eCHx2), 7.04 (t, J ¼ 7.3 Hz, 3.62; N, 18.04.
M. Camarasa et al. / European Journal of Medicinal Chemistry 115 (2016) 463e483 479

4.1.15. 4-((1H-benzo[d][1,2,3]triazol-1-yl)oxy)-6-(2,4- ultrasounds and mechanical stirring for 1 h at room temperature


dichlorophenyl)-2-(phenylamino)-5,6-dihydropyrido[2,3-d] and then heated at 80  C for 1 h. The resulting dispersion is diluted
pyrimidin-7(8H)-one (22{11,10}) with water (300 mL), filtered and washed with water, ethanol and
Starting from compound 21{11,10} and using procedure diethyl ether to afford 144 mg (0.36 mmol, yield 72%) of 6-(2,6-
described in 4.1.2. to afford 22{11,10} in 66% yield, white solid. IR dichlorophenyl)-2-(phenylamino)-5,8-dihydropyrido[2,3-d]pyrim-
(KBr): n (cm1): 3418, 3274, 3143, 2983, 1687, 1652, 1581, 1555, idine-4,7(3H,6H)-dione 21{5,10} as a white solid. IR (KBr), nmax
1461, 1327, 1252, 1088, 1064, 1043, 901, 814, 741, 603, 507. 1H NMR (cm1): 3395, 3222, 3106, 2898, 1732, 1645, 1621, 1598, 1563, 1510,
(400 MHz, DMSO-d6): d 11.27 (s, 1H,), 9.48 (s, 1H), 8.22 (d, J ¼ 8.4 Hz, 1497, 1455, 1436, 1367, 1330, 1234, 758. 1H NMR (400 MHz, DMSO-
1H), 7.81 (dt, J ¼ 8.4, 0.9 Hz, 1H), 7.70 (d, J ¼ 2.2 Hz, 1H, C13eCH), d6): d 10.55 (br s, 1H), 10.52 (br s, 1H), 8.80 (br s, 1H), 7.72 (dd, J ¼ 8.7,
7.66 (ddd, J ¼ 8.3, 6.9, 0.9 Hz, 1H), 7.58 (d, J ¼ 8.4 Hz, 1H), 7.56 (ddd, 1.0 Hz, 2H), 7.58e7.46 (m, 2H), 7.37 (t, J ¼ 8.1 Hz, 1H), 7.33e7.23 (m,
J ¼ 8.1, 6.9, 1.0 Hz, 1H), 7.51 (dd, J ¼ 8.4, 2.2 Hz, 1H), 7.03e6.78 (m, 2H), 7.03 (tt, J ¼ 7.4, 1.0 Hz, 1H), 4.61 (dd, J ¼ 13.4, 9.0 Hz, 1H), 2.87
4H), 6.78e6.72 (m, 1H, C18eCH), 4.55 (dd, J ¼ 11.6, 9.0 Hz, 1H, (dd, J ¼ 15.9, 9.0 Hz, 1H), 2.74 (dd, J ¼ 15.9, 13.5 Hz, 1H). 13C NMR
C7eCH), 3.35 (d, J ¼ 8.9 Hz, 2H, C6eCH2). 13C NMR (100.6 MHz, (100.6 MHz, DMSO-d6): d 169.7, 161.1, 155.0, 151.0, 138.7, 135.5,
DMSO-d6): d 170.4, 165.0, 160.3, 157.2, 142.9, 139.3, 135.4, 134.6, 135.2, 134.7, 129.8, 129.8, 128.8, 128.3, 122.5, 119.2, 89.1, 43.2, 22.5.
132.8, 132.2, 129.2, 129.0, 128.6, 127.9, 127.6, 125.2, 121.6, 119.9, MS (ESI-TOF): m/z (%) ¼ 401.1 (100) [MþH]þ, 365.1 (3) [M-Cl]þ.
118.5, 109.5, 87.9, 43.3. HRMS (ESI-TOF): m/z calculated for C19H15N4O2Cl2: 401.0567
[MþH]þ, found: 401.0563. 401.2 mg (1.0 mmol) of 6-(2,6-
4.1.16. 6-(4-fluorophenyl)-2-(phenylamino)-4-((2-propoxyethyl) dichlorophenyl)-2-(phenylamino)-5,8-dihydropyrido[2,3-d]pyrim-
amino)-5,6-dihydropyrido[2,3-d]pyrimidin-7(8H)-one (23{2,10,6}) idine-4,7(3H,6H)-dione 21{5,10} were suspended in acetonitrile
Starting from 22{2,10} using procedure B described in 4.1.4. and (75 mL), then BOP (574.6 g, 1.30 mmol) and DBU (225 mL,
2-propoxyethylamine to afford 23{2,10,6} in 64% yield, white solid. 1.50 mmol) were added to the solution. The mixture was stirred at
1
H NMR (400 MHz, DMSO-d6): d 10.28 (s, 1H, CONH), 8.82 (s, 1H, room temperature for 2 days. The solvent was removed in vacuo
NHPh), 7.81 (d, J ¼ 7.7 Hz, 2H, C16eCHx2), 7.32 (dd, J ¼ 8.7, 5.6 Hz, and water was added. The resulting solid was filtered and dried
2H, C12eCHx2), 7.22e7.12 (m, 4H, C17eCHx2, C13eCHx2), 6.85 (t, under reduced pressure over phosphorus pentoxide. Without
J ¼ 7.3 Hz, 1H, C18eCH), 6.70 (s, 1H, C4eNH), 3.92 (dd, J ¼ 10.5, further purification, 259.2 mg (0.5 mmol) of 22{5,10} and 182 mL
6.9 Hz, 1H, C7eCH), 3.54e3.51 (m, 4H, C19eCH2, C20eCH2), (1.5 mmol) of 2-propoxyethylamine were solved in 10 mL of
3.37e3.29 (m, 2H, C21eCH2), 2.99e2.71 (m, 2H, C6eCH2), 1.48 (p, acetonitrile and heated at 140  C for 5 h. The solvent was concen-
J ¼ 7.0 Hz, 2H, C22eCH2), 0.84 (t, J ¼ 7.4 Hz, 3H, CH3). 13C NMR trated under reduced pressure and water was added to the residue.
(100.6 MHz, DMSO-d6): d 171.7, 161.2 (d, J ¼ 242.8 Hz, C14), 160.0, The solid was collected by filtration and washed with water and
158.0, 155.5, 141.4, 135.7 (d, J ¼ 3.1 Hz, C11), 130.3 (d, J ¼ 7.9 Hz, C12), EtOEt to afford 6-(2,6-dichlorophenyl)-2-(phenylamino)-4-((2-
128.2 (C17), 120.3 (C18), 118.4 (C16), 115.0 (d, J ¼ 21.3 Hz, C13), 86.1 propoxyethyl)amino)-5,6-dihydropyrido[2,3-d]pyrimidine-7(8H)-
(C5), 71.8 (C21), 68.7 (C20), 45.3 (C7), 41.4 (19), 25.6 (C6), 22.5 (C22), one (23{5,10,6}) in 78% yield as a yellow solid. IR (KBr), nmax (cm1):
10.5(C23). HRMS (70 eV, EI): m/z calculated for C24H26N5O2F: 3441, 3286, 3207, 3132, 2960, 2923, 2871, 1683, 1637, 1600, 1581,
435.2071, [M]þ, found: 435.2070. 1553, 1523, 1499, 1441, 1375, 1349, 1289, 1248, 1110, 1087, 985, 829,
781, 754, 693. 1H NMR (400 MHz, DMSO-d6) d 10.31 (br s, 1H), 8.82
4.1.17. 6-(4-fluorophenyl)-2-phenoxy-4-((2-propoxyethyl)amino)- (br s, 1H), 7.83 (dd, J ¼ 8.7, 1.0 Hz, 2H), 7.54 (m, 2H), 7.39 (t,
5,6-dihydropyrido[2,3-d]pyrimidin-7(8H)-one (23{2,14,6}) J ¼ 8.1 Hz, 1H), 7.19 (dd, J ¼ 8.5, 7.4 Hz, 2H), 6.85 (br t, J ¼ 7.3 Hz, 1H),
Starting from 22{2,14} using procedure B described in 4.1.4. and 6.60 (s, 1H), 4.68 (dd, J ¼ 13.1, 8.9 Hz, 1H), 3.58e3.48 (m, 4H), 3.35 (t,
2-propoxyethylamine to afford 23{2,14,6} in 54% yield, white solid. J ¼ 6.6 Hz, 2H), 2.99e2.90 (m, 1H), 2.77 (dd, J ¼ 15.7, 13.2 Hz, 1H),
IR (KBr), nmax (cm1): 3466, 3412, 2961, 2333, 2856, 1700, 1623, 1.49 (hex, J ¼ 7.3 Hz, 2H), 0.84 (t, J ¼ 7.4 Hz, 3H). 13C NMR
1587, 1510, 1407, 1348, 1214. 1H NMR (400 MHz, DMSO-d6): d 8.01 (s, (100.6 MHz, DMSO-d6): d 169.2, 160.1, 158.1, 155.1, 141.34, 135.6,
1H, NH), 7.35 (t, J ¼ 7.7 Hz, 2H, C12eCHx2), 7.23e7.12 (m, 5H, 135.2, 134.8, 129.8, 129.7, 128.3, 128.2, 120.3, 118.4, 84.6, 71.7, 68.6,
C16eCHx2, C17eCHx2, C18eCH), 7.01 (t, J ¼ 8.6 Hz, 2H, C13-CHx2), 43.1, 40.3, 23.4, 22.5, 10.5. MS (ESI-TOF): m/z (%) ¼ 486.1 (100)
5.14 (s, 1H, NH), 3.83 (dd, J ¼ 9.7, 7.2 Hz, 1H, C7eCH), 3.61e3.44 (m, [MþH]þ, 426.1 (11) [M-OPr]þ HRMS (ESI-TOF): m/z calculated for
4H, C19eCH2, C20eCH2), 3.35 (t, J ¼ 6.7 Hz, 2H, C21eCH2), C24H26N5O2Cl2: 486.1458, [MþH]þ, found: 486.1456. Anal. Calcd.
2.94e2.68 (m, 2H, C6eCH2), 1.54 (h, J ¼ 7.1 Hz, 2H, C22eCH2), 0.87 (%) for C24H25Cl2N5O2: C, 59.26; H, 5.18; N, 14.40. Found: C, 59.20; H,
(t, J ¼ 7.4 Hz, 3H, C23eCH2). 13C NMR (100.6 MHz, DMSO-d6): 5.29; N, 14.48.
d 170.8, 163.9, 162.3 (d, J ¼ 246.6 Hz, C14), 161.4, 156.2, 153.1, 133.8
(d, J ¼ 3.3 Hz, C11), 129.8 (d, J ¼ 8.1 Hz, C12) 129.2 (C17), 125.0 (C18), 4.1.19. Aromatization of pyrido[2,3-d]pyrimidines 21{x,y} or 23
122.0 (C16), 115.9 (d, J ¼ 21.5 Hz, C13), 88.8 (C5), 72.8 (C21), 68.9 {x,y,z}
(C20), 45.9 (C7), 41.1 (C19), 26.0 (C6), 22.8 (C22), 10.6 (C23). HRMS 4.1.19.1. 6-(4-fluorophenyl)-2-(phenylamino)-4-((2-propoxyethyl)
(70 eV, EI): m/z calculated for C24H25N4O3F: 436.1911, [M]þ, found: amino)pyrido[2,3-d]pyrimidin-7(8H)-one (24{2,10,6}). Starting from
436.1913. Anal. Calcd. (%) for C24H25N4O3F: C, 66.04; H, 5.77; N, 23{2,10,6} and following procedure A described in 4.1.6. to afford 24
12.84. Found: C, 66.09; H, 6.08; N, 12.56. {2,10,6} in 78% yield, white solid. IR (KBr), nmax (cm1): 3445, 3059,
2928, 1636, 1603, 1570, 1499, 1451, 1310, 1228. 1H NMR (400 MHz,
4.1.18. 6-(2,6-dichlorophenyl)-2-(phenylamino)-4-((2- DMSO-d6): d 11.80 (s, 1H, NH), 9.31 (s, 1H, NH), 8.32 (s, 1H, C6),
propoxyethyl)amino)-5,6-dihydropyrido[2,3-d]pyrimidine-7(8H)- 7.90e7.85 (m, 2H, C16eCHx2), 7.80 (dd, J ¼ 8.8, 5.7 Hz, 2H,
one (23{5,10,6}) C12eCHx2), 7.27e7.22 (m, 4H, C13eCHx2, C17-CHx2), 6.96e6.91
A dispersion of 9{5,10} [13] (200.1 mg, 0.5 mmol) in acetic acid (m, 1H, C18), 3.67e3.57 (m, 4H, C19eCH2, C20eCH2), 3.38 (t,
(10 mL) is treated with sodium nitrite (173 mg, 2.51 mmol) for 2 h at J ¼ 6.7 Hz, 2H, C21eCH2), 1.51 (h, J ¼ 7.2 Hz, 2H, C22eCH2), 0.84 (t,
room temperature. Then, the mixture is diluted with water J ¼ 7.4 Hz, 3H, C23eCH3). 13C NMR (100.6 MHz, DMSO-d6): d 162.4,
(100 mL), filtered and the resulting cake intensively washed with 161.3 (d, J ¼ 244.6 Hz, C14), 158.9, 157.5, 155.0, 140.6 (C15), 133.0 (d,
water. This cake is recovered by solubilization with ethanol, which J ¼ 7.9 Hz, C11), 132.2 (C6), 130.2 (d, J ¼ 7.7 Hx, C12), 128.3 (C17),
can be removed by vacuum distillation. The resulting slurry is 122.8 (C7), 121.4 (C18), 119.4 (C16), 114.6 (d, J ¼ 21.4 Hz, C13), 92.4
dispersed in 1 M aqueous hydrochloric acid 1 M (30 mL) by (C5), 71.9 (C21), 68.4 (C20), 41.4 (C19), 22.4 (C22), 10.5 (C23). HRMS
480 M. Camarasa et al. / European Journal of Medicinal Chemistry 115 (2016) 463e483

(70 eV, EI): m/z calculated for C24H24N5O2F: 433.1914, [M]þ, found: (%) for C24H23Cl2N5O2: C, 59.51; H, 4.79; N, 14.46. Found: C, 59.85;
433.1927. Anal. Calcd. (%) for C24H24FN5O2: C, 66.50; H, 5.58; N, H, 5.63; N, 14.26.
16.16. Found: C, 66.53; H, 5.86; N, 16.25.
4.1.20. General procedure for the alkylation at N8 of pyrido[2,3-d]
4.1.19.2. 6-(4-fluorophenyl)-2-phenoxy-4-((2-propoxyethyl)amino)
pyrimidines 24{x,y,z}
pyrido[2,3-d]pyrimidin-7(8H)-one (24{2,14,6}). Starting from 23
To a solution of (0.7 mmol) of the corresponding pyrido[2,3-d]
{2,14,6} and following procedure A described in 4.1.6. to afford 24
pyrimidine 24{x,y,z} in 10 mL of anhydrous DMSO, 28.0 mg
{2,14,6} in 75% yield, white solid. IR (KBr), nmax (cm1): 3313, 2933,
(0.7 mmol) of sodium hydride (NaH) (60% dispersion in mineral oil)
2873, 1648, 1612, 1591, 1571, 1510, 1419, 1401, 1264, 1207. 1H NMR
were added and the mixture was stirred for 1 h at room tempera-
(400 MHz, DMSO-d6): d 9.08 (s, 1H, NH), 7.64e7.58 (m, 3H,
ture under nitrogen atmosphere. After this period, (0.7 mmol) of
C12eCHx2, C6eCH), 7.43e7.37 (m, 2H, C17eCHx2), 7.25e7.17 (m,
the corresponding alkyl halide were added dropwise and then
3H, C18eCHx2, C16eCH), 7.07 (t, J ¼ 8.7 Hz, 2H, C13-CHx2), 6.08 (s,
stirred overnight at room temperature. The reaction was quenched
1H, NH), 3.69 (q, J ¼ 5.1 Hz, 2H, C19eCH2), 3.58 (t, J ¼ 5.0 Hz, 2H,
by addition of 300 mL of water and the resulting precipitate was
C20eCH2), 3.42 (t, J ¼ 6.7 Hz, 2H, C21eCH2), 1.64e1.58 (m, 2H,
filtered, washed with water and dried in vacuo over phosphorus
C22.CH2), 0.93 (t, J ¼ 7.4 Hz, 3H, C23eCH3). 13C NMR (100.6 MHz,
pentoxide to afford the corresponding pyrido[2,3-d]pyrimidine 25
DMSO-d6): d 164.8, 162.7 (d, J ¼ 248.1 Hz, C14), 162.4, 160.7, 155.2,
{x,y,z,k}.
152.8, 131.7 (d, J ¼ 3.3 Hz, C11), 130.6, 130.4 (d, J ¼ 8.1 Hz, C12), 129.3
(C17), 127.9, 125.4 (C18), 122.1 (C16), 115.3 (d, J ¼ 21.4 Hz, C13), 94.5
(C5), 72.9 (C21), 68.7 (C20), 41.3 (C19), 22.7 (C22), 10.6 (C23). HRMS 4.1.20.1. 4-Amino-6-(2,6-dichlorophenyl)-8-ethyl-2-(phenylamino)
(70 eV, EI): m/z calculated for C24H23N4O3F: 434.1754, [M]þ, found: pyrido[2,3-d]pyrimidine-7(8H)-one (25{5,10,4,2}). Starting from 24
434.1758. Anal. Calcd. (%) for C24H23N4O3F: C, 66.35; H, 5.34; N, {5,10,4} and following the general methodology described above to
12.90. Found: C, 66.57; H, 5.31; N, 12.51. afford 25{5,10,4,2} in 88% yield, brownish solid. IR (KBr), nmax
(cm1): 3386, 2974, 2929, 1670, 1614, 1570, 1517, 1497, 1472, 1446,
4.1.19.3. 6-(4-fluorophenyl)-2-(phenylamino)pyrido[2,3-d]pyrimi- 1401, 1337, 1279, 1238, 1185, 1030, 832, 803, 772, 753. 1H NMR
dine-4,7(3H,8H)-dione (24{2,10}). Starting from 21{2,10} and (400 MHz, DMSO-d6) d 9.41 (br s, 1H), 8.09 (s, 1H), 7.83 (dd, J ¼ 8.7,
following procedure A described in 4.1.6. to afford 24{2,10} in 78% 1.0 Hz, 2H), 7.60e7.55 (m, 2H), 7.44 (dd, J ¼ 8.7, 7.5 Hz, 1H), 7.38 (br
yield, white solid. IR (KBr), nmax (cm1): 3391, 2924, 2850, 1611, s, 2H), 7.29 (dd, J ¼ 8.6, 7.4 Hz, 2H), 6.97 (tt, J ¼ 7.3, 1.1 Hz, 1H), 4.35
1554, 1497, 1442, 1226. 1H NMR (400 MHz, DMSO-d6): d 12.18 (s, 1H, (q, J ¼ 6.9 Hz, 2H), 1.24 (t, J ¼ 7.0 Hz, 3H). 13C NMR (100.6 MHz,
NH), 10.81 (s, 1H, NH), 9.12 (s, 1H, NH), 7.89 (s, 1H, C6eCH), DMSO-d6) d 161.7, 160.0, 159.2, 155.5, 140.4, 135.3, 135.3, 134.0,
7.78e7.72 (m, 4H, C12eCHx2, C16eCHx2), 7.35 (dd, J ¼ 8.6, 7.3 Hz, 130.2, 128.4, 128.0, 121.6, 120.4, 119.5, 91.6, 36.0, 13.0. MS (70 eV, EI):
2H, C17eCHx2), 7.21 (t, J ¼ 8.9 Hz, 2H, C13eCHx2), 7.14e7.08 (m, m/z (%) ¼ 425.1 (1) [M]þ, 396.1 (1) [M-NEt]þ, 390.1 (2) [M-Cl]þ, 43.1
1H, C18eCH). 13C NMR (100.6 MHz, DMSO-d6): d 162.4, 161.4 (d, (100) [NEt]þ. HRMS (70 eV, EI): m/z calculated for C21H17N5OCl2:
J ¼ 244.6 Hz, C14), 159.7, 155.1, 151.0, 137.9, 133.5 (C7), 132.6 (d, 425.0810, [M]þ, found: 425.0806.
J ¼ 3.1 Hz, C11), 130.1 (d, J ¼ 8.1 Hz, C12), 128.9 (C17), 123.7 (C18),
123.5, 120.1 (C16), 114.8 (d, J ¼ 21.2 Hz, C13), 97.2 (C5). HRMS (70 eV,
4.1.20.2. 4-Amino-6-(2,6-dichlorophenyl)-2-(phenylamino)-8-
EI): m/z calculated for C19H13N4O2F: 348.1023, [M]þ, found:
propylpyrido[2,3-d]pyrimidine-7(8H)-one (25{5,10,4,3}).
348.1022.
Starting from 24{5,10,4} and following the general methodology
described above to afford 25{5,10,4,3} in 86% yield, brownish solid.
4.1.19.4. 6-(4-fluorophenyl)-2-((4-methoxyphenyl)amino)pyrido[2,3-
IR (KBr), nmax (cm1): 3336, 3141, 2958, 2927, 2872, 1677, 1625,
d]pyrimidine-4,7(3H,8H)-dione (24{2,13}). Starting from 21{2,13}
1574, 1518, 1473, 1447, 1339, 1236, 1185, 1031, 802, 754, 694. 1H NMR
and following procedure A described in 4.1.6. to afford 24{2,13} in
(400 MHz, DMSO-d6) d 9.39 (br s, 1H), 8.09 (s, 1H), 7.82 (dd, J ¼ 8.7,
70% yield as a white solid.1H NMR (400 MHz, DMSO-d6): d 12.07 (s,
1.0 Hz, 2H), 7.61e7.54 (m, 2H), 7.43 (dd, J ¼ 8.7, 7.5 Hz, 1H), 7.35 (br s,
1H, NH), 10.74 (s, 1H, NH), 8.96 (s, 1H, NH), 7.87 (s, 1H, C6eCH), 7.74
2H), 7.31e7.23 (m, 2H), 6.97 (tt, J ¼ 7.3, 1.0 Hz, 1H), 4.31e4.21 (m,
(dd, J ¼ 8.9, 5.6 Hz, 2H, C12eCHx2), 7.62 (d, J ¼ 9.0 Hz, 2H,
2H), 1.76e1.64 (hex, J ¼ 7.4 Hz, 2H), 0.94 (t, J ¼ 7.5 Hz, 3H). 13C NMR
C16eCHx2), 7.20 (t, J ¼ 8.9 Hz, 2H, C13eCHx2), 6.92 (d, J ¼ 9.0 Hz,
(100.6 MHz, DMSO-d6) d 161.6, 160.2, 159.1, 155.6, 140.4, 135.4,
2H, C15eCHx2), 3.76 (s, 3H, OCH3). 13C NMR (100.6 MHz, DMSO-
135.3, 133.9, 130.2, 128.3, 128.0, 121.7, 120.4, 119.5, 91.6, 42.4, 20.8,
d6): d 162.5, 160.1, 159.8, 155.8, 155.3, 151.3, 133.9, 133.6 (C6), 132.7
11.3. HRMS (ESI-TOF): m/z calculated for C22H20N5OCl2: 440.1039,
(d, J ¼ 2.9 Hz, C11), 130.7, 130.1 (d, J ¼ 8.0 Hz, C12), 123.3, 122.4
[MþH]þ, found: 440.1046.
(C16), 114.8 (d, J ¼ 21.2 Hz, C13), 114.1 (C17), 97.0 (C5), 55.3 (C6).

4 .1.19 . 5 . 6 - ( 2 , 6 - d i ch l o ro p h e n yl ) - 2 - ( p h e n yl a m i n o ) - 4 - ( ( 2 - 4.1.20.3. 4-Amino-8-butyl-6-(2,6-dichlorophenyl)-2-(phenylamino)


propoxyethyl)amino)pyrido[2,3-d]pyrimidin-7(8H)-one (24{5,10,6}). pyrido[2,3-d]pyrimidine-7(8H)-one (25{5,10,4,4}). Starting from 24
Starting from 23{5,10,6} and following procedure A described in {5,10,4} and following the general methodology described above to
4.1.6. to afford 24{5,10,6} in 65% yield, brownish solid. IR (KBr), nmax afford 25{5,10,4,4} in 78% yield, brownish solid. IR (KBr), nmax
(cm1): 3209, 3060, 2929, 1614, 1596, 1573, 1499, 1450, 1304, 1263, (cm1): 3346, 3150, 2959, 2927, 2859, 1626, 1573, 1518, 1497, 1471,
1191, 1108, 989, 903, 798. 1H NMR (400 MHz, DMSO-d6) d 11.85 (br s, 1445, 1339, 1234, 1202, 1031, 803, 772, 755, 692. 1H NMR (400 MHz,
1H), 9.34 (br s, 1H), 8.09 (s, 1H), 7.91e7.82 (m, 3H), 7.60e7.53 (m, DMSO-d6) d 9.39 (br s, 1H), 8.09 (s, 1H), 7.82 (dd, J ¼ 8.7, 1.1 Hz, 2H),
2H), 7.44 (dd, J ¼ 8.6, 7.6 Hz, 1H), 7.26 (t, J ¼ 7.4 Hz, 2H), 6.95 (t, 7.57 (m, 2H), 7.43 (dd, J ¼ 8.7, 7.4 Hz, 1H), 7.34 (br s, 2H), 7.30e7.24
J ¼ 7.3 Hz, 1H), 3.67e3.54 (m, 4H), 3.35 (t, J ¼ 6.7 Hz, 2H), 1.49 (h, (m, 2H), 6.97 (tt, J ¼ 7.3, 1.1 Hz, 1H), 4.33e4.23 (m, 2H), 1.66 (quint,
J ¼ 7.3 Hz, 2H), 0.83 (t, J ¼ 7.4 Hz, 3H). 13C NMR (100.6 MHz, DMSO- J ¼ 8.0 Hz, 2H), 1.39 (hex, J ¼ 7.5 Hz, 2H), 0.93 (t, J ¼ 7.4 Hz, 3H). 13C
d6): d 161.1, 159.4, 159.2, 155.9, 149.4, 140.5, 135.4, 135.0, 130.3, NMR (100.6 MHz, DMSO-d6) d 161.7, 160.3, 159.2, 155.7, 140.4, 135.4,
128.3, 128.0, 121.5, 119.5, 109.6, 91.6, 71.8, 68.2, 40.5, 22.4, 10.5. MS 135.4, 134.0, 130.4, 128.5, 128.1, 121.9, 120.6, 119.6, 91.8, 40.9, 29.7,
(ESIeTOF): m/z (%) ¼ 484.1 (75) [MþH]þ, 448.1 (100) [M-Cl]þ, 426.1 20.0, 13.9. MS (70 eV, EI): m/z (%) ¼ 453.1 (1) [M]þ, 418.1 (1) [M-Cl]þ,
(54) [MþH-OPr]þ. HRMS (ESI-TOF): m/z calculated for 396.1 (2) [M-But]þ, 57.0 (100) [But]þ. HRMS (70 eV, EI): m/z
C24H24N5O2Cl2: 484.1302, [MþH]þ, found: 484.1293. Anal. Calcd. calculated for C23H21N5OCl2: 453.1123, [M]þ, found: 453.1123.
M. Camarasa et al. / European Journal of Medicinal Chemistry 115 (2016) 463e483 481

4.1.20.4. 4-Amino-6-(2,6-dichlorophenyl)-8-isobutyl-2-(phenyl- N, 13.79. MS (ESI-TOF): m/z (%) ¼ 498.1 (100) [MþH]þ, 438.1 (13)
amino)pyrido[2,3-d]pyrimidine-7(8H)-one (25{5,10,4,5}). [M-OC3H7]þ. HRMS (ESI-TOF): m/z calculated for C25H26N5O2Cl2:
Starting from 24{5,10,4} and following the general methodology 498.1458, [MþH]þ, found: 498.1460.
described above to afford 25{5,10,4,5} in 83% yield, brownish solid.
IR (KBr), nmax (cm1): 3333, 3199, 2958, 2926, 2869, 1630, 1571, 4.2. Biology
1522, 1497, 1468, 1446, 1340, 1307, 1234, 1186, 1091, 1027, 799, 778,
753, 691. 1H NMR (400 MHz, DMSO-d6) d 9.38 (br s, 1H), 8.09 (s, 1H), 4.2.1. Cell lines and cell culture
7.82 (dd, J ¼ 8.7, 1.1 Hz, 2H), 7.60e7.55 (m, 2H), 7.44 (dd, J ¼ 8.7, To analyze the antiviral activity of the compounds, we used a
7.5 Hz, 1H), 7.34 (br s, 2H), 7.32e7.23 (m, 2H), 6.98 (tt, J ¼ 7.3, 1.1 Hz, stable cell line, LucUbiNeo-ET, that contains a subgenomic HCV
1H), 4.18 (d, J ¼ 7.4 Hz, 2H), 2.29 (hept, J ¼ 7.0 Hz, 1H), 0.90 (d, genotype 1b replicon, I389/NS3-3’/LucUbiNeo-ET, that express
J ¼ 6.7 Hz, 6H). 13C NMR (100.6 MHz, DMSO-d6) d 161.7, 160.6, 159.0, luciferase constitutively [21,22]. To evaluate the cellular toxicity, we
155.9, 140.4, 135.5, 135.4, 133.9, 130.3, 128.4, 128.1, 121.8, 120.6, use other cell line, Huh-7/Lunet [23], that originally carried a
119.6, 91.7, 47.4, 26.9, 20.0. MS (70 eV, EI): m/z (%) ¼ 453.1 (1) [M]þ, selectable luciferase HCV 1b replicon and was cured by treatment
418.3 (1) [M-Cl]þ, 396.1 (4) [M-isoBut]þ, 57.0 (100) [isoBut]þ. HRMS with an HCV-specific inhibitor. These cells are characterized by
(70 eV, EI): m/z calculated for C23H21N5OCl2: 453.1123, [M]þ, found: high permissiveness for HCV RNA replication. LucUbiNeo-ET cell
453.1128. line was cultured at 37  C in a humidified atmosphere with 5% CO2
in Dulbecco's modified Eagle medium (D-MEM) supplemented
4.1.20.5. 4-Amino-6-(2,6-dichlorophenyl)-8-(2-methoxyethyl)-2- with high glucose concentration (4.5 g/l) (Invitrogen), 10% (vol/vol)
(phenylamino)pyrido[2,3-d]pyrimidine-7(8H)-one (25{5,10,4,6}). heat-inactivated fetal bovine serum (Hyclone, Cultek), 2 mM L-
Starting from 24{5,10,4} and following the general methodology Glutamine (SigmaeAldrich), 1 mM sodium pyruvate (Sigma-
described above to afford 25{5,10,4,6} in 83% yield, brownish solid. aldrich), 1% MEM NEEA (SigmaeAldrich), and 0.5 mg/mL of
IR (KBr), nmax (cm1): 3340, 2925, 1632, 1574, 1523, 1497, 1468, 1447, Geneticin (G418) (Gibco, Invitrogen). Cured or Huh-7/Lunet cell line
1341, 1306, 1235, 1196, 1117, 1028, 801, 771, 754, 692. 1H NMR was maintained at the same conditions as described above without
(400 MHz, DMSO-d6) d 9.42 (br s, 1H), 8.10 (s, 1H), 7.82 (dd, J ¼ 8.7, the addition of Geneticin.
1.0 Hz, 2H), 7.61e7.55 (m, 2H), 7.44 (dd, J ¼ 8.7, 7.5 Hz, 1H), 7.40 (br
s, 2H), 7.31e7.25 (m, 2H), 6.97 (tt, J ¼ 7.3, 1.1 Hz, 1H), 4.50 (t, 4.2.2. Evaluation of the in vitro antiviral activity (EC50) and
J ¼ 6.6 Hz, 2H), 3.61 (t, J ¼ 6.6 Hz, 2H), 3.27 (s, 3H). 13C NMR cytotoxicity assays (CC50)
(100.6 MHz, DMSO-d6) d 161.8, 160.3, 159.1, 155.9, 140.3, 135.4, Antiviral activity was carried out in white-clear bottom 96-well
135.3, 134.3, 130.4, 128.5, 128.1, 121.9, 120.3, 119.7, 91.7, 68.5, 58.2, plates, and cytotoxicity assays were performed in clear 96-well
40.2. MS (ESI-TOF): m/z (%) ¼ 456.1 (51) [MþH]þ, 424.1 (100) [M- plates. Cells were seeded at a density of 104/well in 100ul D-MEM
OMe]þ. HRMS (ESI-TOF): m/z calculated for C22H20N5O2Cl2: without selection of antibiotics. After 24 h, nine fivefold serial
456.0989, [MþH]þ, found: 456.0988. dilution of compounds, with 3 replicates at each dilution, were
prepared in D-MEM and added to the appropriate wells, yielding
4.1.20.6. 4-Amino-6-(2,6-dichlorophenyl)-8-(2-hydroxyethyl)-2- concentrations of 125 to 0,00032 ug/ml in a final volume of 100 ul
(phenylamino)pyrido[2,3-d]pyrimidine-7(8H)-one (25{5,10,4,7}). of D-MEM. Following 2 days of incubation, we determined the
Starting from 24{5,10,4} and following the general methodology antiviral activity (EC50) by quantifying the luciferase with the
described above with an extra purification consisting of a chro- Luciferase Bright Glo assay (Promega). Briefly, LucUbiNeo-ET cells
matographic separation in silica and CH2Cl2/MeOH as solvent to were harvested with the addition of 100 ul of luciferase per well,
afford 25{5,10,4,7} in 34% yield, brownish solid. IR (KBr), nmax and after 2 min of shaking, the light was read in a Luminometer
(cm1): 3418, 2924, 1634, 1599, 1568, 1525, 1497, 1469, 1447, 1341, Fluoroskan Ascent FL. Toxicity (CC50) was analyzed in the Lunet/
1306, 1235, 1201, 1057, 1015, 802, 772, 691. 1H NMR (400 MHz, HuH7 cells by the addition of 10 ul of MTT (3-(4,5-dimethylthiazol-
DMSO-d6) d 9.40 (br s, 1H), 8.09 (s, 1H), 7.84 (dd, J ¼ 8.7, 1.0 Hz, 2H), 2-yl)-2,5-diphenyltetrazolium bromide) per well. After 4 h of in-
7.60e7.55 (m, 2H), 7.44 (dd, J ¼ 8.7, 7.5 Hz, 1H), 7.38 (br s, 2H), 7.28 cubation at 37  C in a humidified atmosphere with 5% CO2, the
(m, 2H), 6.96 (tt, J ¼ 7.3, 1.1 Hz, 1H), 4.82 (br t, J ¼ 5.3 Hz, 1H), 4.42 (t, amount of formazan produced was quantified spectrophotometri-
J ¼ 7.1 Hz, 2H), 3.64 (q, J ¼ 6.5 Hz, 2H). 13C NMR (100.6 MHz, DMSO- cally at 550/620 nm [43]. The EC50 and CC50 values were calculated
d6) d 161.8, 160.4, 159.2, 156.0, 140.4, 135.4, 135.3, 134.2, 130.4, 128.6, for each compound. These assays were done in duplicate and the
128.1, 121.8, 120.4, 119.6, 91.7, 58.0, 42.8. MS (ESI-TOF): m/z results shown in Table 1 represent the mean value of these
(%) ¼ 442.1 (100) [MþH]þ, 424.1 (81) [M-OH]þ. HRMS (ESI-TOF): m/ duplicates.
z calculated for C21H18N5O2Cl2: 442.0832, [MþH]þ, found:
442.0839. 4.3. Computational studies

4.1.20.7. 6-(2,6-dichlorophenyl)-8-methyl-2-(phenylamino)-4-((2- Chemical Similarity Searching. Chemical similarity was per-


propoxyethyl)amino)pyrido[2,3-d]pyrimidine-7(8H)-one (25 formed using Omega 2.3.2 [44] and ROCS 3.0 (Rapid Overlay of
{5,10,6,1}). Starting from 24{5,10,6} and following the general Chemical Structures) [45] software from the OpenEye Scientific
methodology described above to afford 25{5,10,6,1} in 96% yield, Software. Using the RCSB Protein Data Bank, NS5B X-ray structures
brownish solid. IR (KBr), nmax (cm1): 3338, 2930, 2872, 1642, 1577, cocrystallized with a ligand were retrieved and classified according
1519, 1498, 1474, 1446, 1341, 1316, 1229, 1184, 1103, 799, 753, 692. 1H to the binding site (sites I to IV). For each ligand, 100 conformations
NMR (400 MHz, DMSO-d6) d 9.52 (br s, 1H), 8.14 (s, 1H), 7.93 (br s, were generated using Omega and compared to our in-house com-
1H), 7.83 (d, J ¼ 7.6 Hz, 2H), 7.58 (m, 2H), 7.44 (dd, J ¼ 8.7, 7.5 Hz, pound database. Three-dimensional shape comparisons (both
1H), 7.30 (t, J ¼ 7.4 Hz, 2H), 6.98 (t, J ¼ 7.3 Hz, 1H), 3.70e3.54 (m, shape Tanimoto coefficients and color scores) were performed with
7H), 3.37 (t, J ¼ 6.6 Hz, 2H), 1.49 (hex, J ¼ 7.4 Hz, 2H), 0.83 (t, ROCS. The alignment overlap was calculated using the color force
J ¼ 7.4 Hz, 3H). 13C NMR (100.6 MHz, DMSO-d6) d 160.8, 160.3, 159.2, field implicit Mills-Dean [46]. The Tanimoto Combo score corre-
156.2, 140.7, 135.8, 135.8, 133.8, 130.7, 128.9, 128.5, 122.2, 120.4, sponds to the sum of the shape Tanimoto coefficient and the color
120.0, 92.5, 72.2, 68.6, 41.1, 28.9, 22.9, 10.9. Anal. Calcd. (%) for score. Molecules with a Tanimoto Combo score higher than 0.7
C25H25Cl2N5O2: C, 60.25; H, 5.06; N, 14.05. Found: C, 60.50; H, 4.84; were considered as similar while scores higher than 0.95 were
482 M. Camarasa et al. / European Journal of Medicinal Chemistry 115 (2016) 463e483

considered as highly similar. [3,2-d]pyrimdines Useful for Treating Viral Infections, WO2008/077651
(Gilead Sciences, INC.) (24/12/2007).
Cross docking methodology. Complexes PDB ID 2WCX [31], , R. Pascual, M. Mora, J. Pujol
[10] B. Martínez-Teipel, J. Teixido a, T. Fujimoto,
2WHO [30], 3BSC [36], 3H98 [29], 4KE5 [37] and 4NLD [32] were J.I. Borrell, E.L. Michelotti, 2-Methoxy-6-oxo-1,4,5,6-tetrahydropyridine-3-
selected based on the previous chemical similarity results and used carbonitriles: versatile starting materials for the synthesis of libraries with
as target structures for our modeling studies. Water molecules diverse heterocyclic scaffolds, J. Comb. Chem. 7 (2005) 436e448 and the
references therein.
were deleted and the protein structures were prepared with [11] P. Victory, R. Nomen, O. Colomina, M. Garriga, A. Crespo, New synthesis of
Schro€dinger Protein Preparation Wizard [47]. This workflow en- pyrido[2,3-d]pyrimidines. 1. Reaction of 6-alkoxy-5-cyano-3,4-dihydro-2-
sures chemical corrected and optimized protein structures for pyridones with guanidine and cyanamide, Heterocycles 23 (1985)
1135e1141.
further analysis. Hydrogen atom were added and bond order and [12] I. Perez-Pi, X. Berzosa, I. Galve, J. Teixido, J.I. Borrell, Dehydrogenation of 5,6-
charges were assigned. A small number of minimization steps were dihydropyrido[3,2-d]pyrimidin-7(8H)-ones: a convenient last step for a syn-
applied in order to relieve steric clashes using the OPLS 2005 force thesis of pyrido[2,3-d]pyrimidin-7(8H)-ones, Heterocycles 82 (2010)
581e591.
field. [13] I. Galve, R. Puig de la Bellacasa, D. Sanchez-Garcia, X. Batllori, J. Teixido ,
The ligand database used during the cross docking protocol J.I. Borrell, Synthesis of 2-arylamino substituted 5,6-dihydropyrido[2,3-d]py-
consist of our in house database as well as the ligands cocrystallized rimidine-7(8H)-ones from arylguanidines, Mol. Diver. 16 (2012) 639e649.
[14] M. Camarasa, C. Barnils, R. Puig De La Bellacasa, J. Teixido , J.I. Borrell, A new
in the aforementioned NS5B complexes. Compounds were sub- and practical method for the synthesis of 6-aryl-5,6-dihydropyrido[2, 3-d]
mitted to the LigPrep utility which takes into account ionization pyrimidine-4,7(3H,8H)-diones, Mol. Divers. 17 (2013) 525e536.
states, tautomers, stereochemistries and ring conformations at a pH [15] Z. Wan, S. Wacharasindhu, E. Binnun, T. Mansour, An efficient direct amina-
tion of cyclic amides and cyclic ureas, Org. Lett. 8 (11) (2006) 2425e2428.
range close to 7.
[16] S.M. Devine, P.J. Scammells, Synthesis and utility of 2-Halo-O6-(benzotriazol-
Cross docking methodology was performed using Glide [48] 1-yl)-functionalized purine nucleosides, Eur. J. Org. Chem. (6) (2011)
from the Schro €dinger Suite. All docking calculations were run in 1092e1098.
extra precision mode and the grid was centered on the crystallo- [17] M.K. Lakshman, J. Frank, A simple method for C-6 modification of guanine
nucleosides, Org. Biomol. Chem. 7 (14) (2009) 2933e2940.
graphic position of the ligand. Further docking refinement was [18] M. Kirihara, A. Itou, T. Noguchi, J. Yamamoto, Tantalum carbide or niobium
achieved by an induced fit (IF) protocol and ligands were ranked carbide catalyzed oxidation of sulfides with hydrogen peroxide: highly effi-
according to the IF docking score. All figures were prepared using cient and chemoselective syntheses of sulfoxides and sulfones, Synlett 10
(2010) 1557e1561.
PyMOL [49]. [19]  , M. Camarasa, R. Puig de la Bellacasa, M.A. Martínez, S.
J.I. Borrell, J. Teixido
Franco, R. Badia, B. Clotet, J. Este , Pyrido[2,3-d]pyrimidin-7(8H)-one De-
Acknowledgment rivatives and Uses Thereof, EP14382046.2 (IQS and IRSICaixa) (7/02/2014).
[20] X. Berzosa, X. Bellatriu, J. Teixido , J.I. Borrell, An unusual michael addition of
3,3-dimethoxypropanenitrile to 2-Aryl acrylates: a convenient route to 4-
Financial support by the Spanish Ministerio de Economia y unsubstituted 5,6-dihydropyrido[2,3-d]pyrimidines, J. Org. Chem. 75 (2010)
Competividad project SAF2010-C21617-C02 is greatly acknowl- 487e490.
[21] N. Krieger, V. Lohmann, R. Bartenschlager, Enhancement of hepatitis C virus
edged. M. Camarasa wants to thank IQS for a scholarship. R. Puig de RNA replication by cell culture-adaptive mutations, J. Virol. 75 (10) (2001)
la Bellacasa wants to thank Generalitat de Catalunya for a grant 4614e4624.
within the Talent empresa (TEM) 2009 program (2009 TEM 00128). [22] M. Frese, K. Barth, A. Kaul, V. Lohmann, V. Schwarzle, R. Bartenschlager,
Hepatitis C virus RNA replication is resistant to tumour necrosis factor-^I±,
J. Gen. Virol. 84 (5) (2003) 1253e1259.
Appendix A. Supplementary data [23] P. Friebe, J. Boudet, J. Simorre, R. Bartenschlager, Kissing-loop interaction in
the 3' end of the hepatitis C virus genome essential for RNA replication,
J. Virol. 79 (1) (2005) 380e392.
Supplementary data related to this article can be found at http:// [24] N.J. Liverton, M.K. Holloway, J.A. McCauley, M.T. Rudd, J.W. Butcher,
dx.doi.org/10.1016/j.ejmech.2016.03.055. S.S. Carroll, J. DiMuzio, C. Fandozzi, K.F. Gilbert, S.S. Mao, C.J. McIntyre,
Taken together, comparison of crystallographic data with our K.T. Nguyen, J.J. Romano, M. Stahlhut, B.L. Wan, D.B. Olsen, J.P. Vacca, Mo-
lecular modeling based approach to potent P2-P4 macrocyclic inhibitors of
best candidates 21{4,10} and 24{2,10} suggest that substituted hepatitis C NS3/4A protease, J. Am. Chem. Soc. 130 (14) (2008) 4607e4609.
pyrido[2,3-d]pyrimidin-7-(8H)-ones are a feasible target for NS5B [25] R. Puig de la Bellacasa, G. Roue , P. Balsas, P. Pe rez-Gala
n, J. Teixido , D. Colomer,
and can potentially disrupt the RNA processing channel. J.I. Borrell, 4-Amino-2-Arylamino-6-(2,6-dichlorophenyl)-pyrido[2,3-d]pyr-
imidin-7-(8H)-ones as BCR kinase inhibitors for B lymphoid malignancies,
Eur. J. Med. Chem. 86 (2014) 664e675.
References [26] M.H. Powdrill, J.A. Bernatchez, M. Go €tte, Inhibitors of the hepatitis C virus
RNA-dependent RNA polymerase NS5B, Viruses 2 (2010) 2169e2195.
[1] C.W. Shepard, L. Finelli, M.J. Alter, Global epidemiology of hepatitis C virus [27] S. Chinnaswamy, H. Cai, C. Kao, An update on small molecule inhibitors of the
infection, Lancet Infect. Dis. 5 (9) (2005) 558e567. HCV NS5B polymerase: effects on RNA synthesis in vitro and in cultured cells,
[2] P. Simmonds, E.C. Holmes, T.A. Cha, S.W. Chan, F. McOmish, B. Irvine, E. Beall, and potential resistance in viral, Virus Adapt. Treat. (2010) 73e89.
P.L. Yap, J. Kolberg, M.S. Urdea, Classification of hepatitis C virus into six major [28] S.E. Lazerwith, W. Lew, J. Zhang, P. Morganelli, Q. Liu, E. Canales, M.O. Clarke,
genotypes and a series of subtypes by phylogenetic analysis of the NS-5 re- E. Doerffler, D. Byun, M. Mertzman, H. Ye, L. Chong, L. Xu, T. Appleby, X. Chen,
gion, J. Gen. Virol. 74 (Pt 11) (1993) 2391e2399. M. Fenaux, A. Hashash, S.A. Leavitt, E. Mabery, M. Matles, J.W. Mwangi, Y. Tian,
[3] C.W. Kim, K.M. Chang, Hepatitis C virus: virology and life cycle, Clin. Mol. Y.J. Lee, J. Zhang, C. Zhu, B.P. Murray, W.J. Watkins, Discovery of GS-9669, a
Hepatol. 19 (2013) 17e25. thumb site II non-nucleoside inhibitor of NS5B for the treatment of genotype
[4] A.U. Neumann, N.P. Lam, H. Dahari, D.R. Gretch, T.E. Wiley, T.J. Layden, 1 chronic hepatitis C infection, J. Med. Chem. 57 (2014) 1893e1901.
A.S. Perelson, Hepatitis C viral dynamics in vivo and the antiviral efficacy of [29] G. Wang, H. Lei, X. Wang, D. Das, J. Hong, C.H. Mackinnon, T.S. Coulter,
interferon-alpha therapy, Science 282 (5386) (1998) 103e107. C.A. Montalbetti, R. Mears, X. Gai, S.E. Bailey, D. Ruhrmund, L. Hooi, S. Misialek,
[5] M.P. Manns, H. Wedemeyer, M. Cornberg, Treating viral hepatitis C: efficacy, P.T. Rajagopalan, R.K. Cheng, J.J. Barker, B. Felicetti, D.L. Scho € nfeld,
side effects, and complications, Gut 55 (9) (2006) 1350e1359. A. Stoycheva, B.O. Buckman, K. Kossen, S.D. Seiwert, L. Beigelman, HCV NS5B
[6] E. Cholongitas, G.V. Papatheodoridis, Sofosbuvir: a novel oral agent for chronic polymerase inhibitors 2: synthesis and in vitro activity of (1,1-dioxo-2H-
hepatitis C, Ann. Gastroenterol. 27 (4) (2014) 331e337. [1,2,4]benzothiadiazin-3-yl)azolo[1,5-a]pyridine and azolo[1,5-a]pyrimidine
[7] B. Maasoumy, K. Port, A.A. Markova, B.C. Serrano, M. Rogalska-Taranta, derivatives, Bioorg. Med. Chem. Lett. 19 (2009) 4480e4483.
L. Sollik, C. Mix, J. Kirschner, M.P. Manns, H. Wedemeyer, M. Cornberg, Eligi- [30] J.M. Ontoria, E.H. Rydberg, S. Di Marco, L. Tomei, B. Attenni, S. Malancona,
bility and safety of triple therapy for hepatitis C: lessons learned from the first J.I. Martin Hernando, N. Gennari, U. Koch, F. Narjes, M. Rowley, V. Summa,
experience in a real world setting, PLoS One 8 (2013) e55285. S.S. Carroll, D.B. Olsen, R. De Francesco, S. Altamura, G. Migliaccio, A. Carfì,
[8] a) C. Zhao, Y. Wang, S. Ma, Recent advances on the synthesis of hepatitis C Identification and biological evaluation of a series of 1H-benzo[de]isoquino-
virus NS5B RNA-dependent RNA-polymerase inhibitors, Eur. J. Med. Chem. line-1,3(2H)-diones as hepatitis C virus NS5B polymerase inhibitors, J. Med.
102 (2015) 188e214; Chem. 52 (2009) 5217e5227.
b) G. Jin, S. Lee, M. Choi, S. Son, G.W. Kim, J.W. Oh, C. Lee, K. Lee, Chemical [31] J.I. Martin Hernando, J.M. Ontoria, S. Malancona, B. Attenni, F. Fiore, F. Bonelli,
genetics-based discovery of indole derivatives as HCV NS5B polymerase in- U. Koch, S. Di Marco, S. Colarusso, S. Ponzi, N. Gennari, S.E. Vignetti, M. Del
hibitors, Eur. J. Med. Chem. 75 (2014) 413e425. Rosario Rico Ferreira, J. Habermann, M. Rowley, F. Narjes, Optimization of
[9] S.S. Bondy, Ch.-H. Chou, W.J. Watkins, L.S. Chong, J.R. Zhang, R. Mishra, Pyrido thienopyrrole-based finger-loop inhibitors of the hepatitis C virus NS5B
M. Camarasa et al. / European Journal of Medicinal Chemistry 115 (2016) 463e483 483

polymerase, ChemMedChem. 4 (2009) 1695e1713. RNA-dependent RNA polymerase, J. Med. Chem. 57 (2014) 1902e1913.
[32] R.G. Gentles, M. Ding, J.A. Bender, C.P. Bergstrom, K. Grant-Young, [38] G. Burton, T.W. Ku, T.J. Carr, T. Kiesow, R.T. Sarisky, J. Lin-Goerke,
P. Hewawasam, T. Hudyma, S. Martin, A. Nickel, A. Regueiro-Ren, Y. Tu, G.A. Hofmann, M.J. Slater, D. Haigh, D. Dhanak, V.K. Johnson, N.R. Parry,
Z. Yang, K.S. Yeung, X. Zheng, S. Chao, J.H. Sun, B.R. Beno, D.M. Camac, P. Thommes, Studies on acyl pyrrolidine inhibitors of HCV RNA-dependent
C.H. Chang, M. Gao, P.E. Morin, S. Sheriff, J. Tredup, J. Wan, M.R. Witmer, RNA polymerase to identify a molecule with replicon antiviral activity, Bio-
D. Xie, U. Hanumegowda, J. Knipe, K. Mosure, K.S. Santone, D.D. Parker, org. Med. Chem. Lett. 17 (2007) 1930e1933.
X. Zhuo, J. Lemm, M. Liu, L. Pelosi, K. Rigat, S. Voss, Y. Wang, Y.K. Wang, [39] E. De Clercq, The design of drugs for HIV and HCV, Nat. Rev. Drug Discov. 6
R.J. Colonno, M. Gao, S.B. Roberts, Q. Gao, A. Ng, N.A. Meanwell, J.F. Kadow, (2007) 1001e1018.
Discovery and preclinical characterization of the cyclo- [40] T.C. Appleby, J.K. Perry, E. Murakami, O. Barauskas, J. Feng, A. Cho, D. Fox,
propylindolobenzazepine BMS-791325, a potent allosteric inhibitor of the D.R. Wetmore, M.E. McGrath, A.S. Ray, M.J. Sofia, S. Swaminathan,
hepatitis C virus NS5B polymerase, J. Med. Chem. 57 (2014) 1855e1879. T.E. Edwards, Viral replication. Structural basis for RNA replication by the
[33] G. Manfroni, R. Cannalire, M.L. Barreca, N. Kaushik-Basu, P. Leyssen, hepatitis C virus polymerase, Science 347 (6223) (2015) 771e775.
J. Winquist, N. Iraci, D. Manvar, J. Paeshuyse, R. Guhamazumder, A. Basu, [41] A.M. Lam, C. Espiritu, S. Bansal, H.M. Micolochick Steuer, C. Niu, V. Zennou,
S. Sabatini, O. Tabarrini, U.H. Danielson, J. Neyts, V. Cecchetti, The versatile M. Keilman, Y. Zhu, S. Lan, M.J. Otto, P.A. Furman, Genotype and subtype
nature of the 6-aminoquinolone scaffold: identification of submicromolar profiling of PSI-7977 as a nucleotide inhibitor of hepatitis C virus, Antimicrob.
hepatitis C virus NS5B inhibitors, J. Med. Chem. 57 (5) (2014) 1952e1963. Agents Chemother. 56 (6) (2012) 3359e3368.
[34] M. Fenaux, S. Eng, S.A. Leavitt, Y.-J. Lee, E.M. Mabery, Y. Tian, D. Byun, [42] B.S. Ross, M.J. Sofia, G.R. Pamulapati, S. Rachakonda, H. Zhang, B. Chun, P.
E. Canales, M.O. Clarke, E. Doerffler, S.E. Lazerwith, W. Lew, Q. Liu, Wang, N-[(2'R)-2'-deoxy-2'-fluoro-2'-methyl-p-phenyl-5'-uridylyl]-L-
M. Mertzman, P. Morganelli, L. Xu, H. Ye, J. Zhang, M. Matles, B.P. Murray, alanine-1-methylethylester and Process for its Production, WO2010135569
J. Mwangi, J. Zhang, A. Hashash, S.H. Krawczyk, A.M. Bidgood, T.C. Appleby, (Pharmaset Inc.) (25/11/2010).
W.J. Watkins, Preclinical characterization of GS-9669, a thumb site II inhibitor [43] C. Pannecouque, D. Daelemans, E. De Clercq, Tetrazolium-based colorimetric
of the hepatitis C virus NS5B polymerase, Antimicrob. Agents Chemother. 57 assay for the detection of HIV replication inhibitors: revisited 20 years later,
(2013) 804e810. Nat. Protoc. 3 (3) (2008) 427e434.
[35] S.E. Boyce, N. Tirunagari, A. Niedziela-Majka, J. Perry, M. Wong, E. Kan, [44] P.C.D. Hawkins, A.G. Skillman, G.L. Warren, B.A. Ellingson, M.T. Stahl,
L. Lagpacan, O. Barauskas, M. Hung, M. Fenaux, T. Appleby, W.J. Watkins, Conformer generation with OMEGA: algorithm and validation using high
U. Schmitz, R. Sakowicz, Structural and regulatory elements of HCV NS5B quality structures from the protein databank and Cambridge structural
polymerase e b-loop and C-terminal tail e are required for activity of allo- database, J. Chem. Inf. Model. 50 (4) (2010) 572e584.
steric thumb site II inhibitors, PLoS One 9 (1) (2014) e84808. [45] P.C.D. Hawkins, A.G. Skillman, A. Nicholls, Comparison of shape-matching and
[36] M. V Sergeeva, Y. Zhou, D.M. Bartkowski, T.G. Nolan, D.A. Norris, E. Okamoto, docking as virtual screening tools, J. Med. Chem. 50 (1) (2007) 74e82.
L. Kirkovsky, R. Kamran, L.A. Lebrun, M. Tsan, R. Patel, A.M. Shah, M. Lardy, [46] J.E. Mills, P.M. Dean, Three-dimensional hydrogen-bond geometry and prob-
A. Gobbi, L.S. Li, J. Zhao, T. Bertolini, N. Stankovic, Z. Sun, D.E. Murphy, ability information from a crystal survey, J. Comput. Aided Mol. Des. 10 (1996)
S.E. Webber, P.S. Dragovich, Novel HCV NS5B polymerase inhibitors derived 607e622.
from 4-(1’,1’-dioxo-1’,4’-dihydro-1'lambda6-benzo[1',2',4']thiadiazin-3'-yl)- [47] G.M. Sastry, M. Adzhigirey, T. Day, R. Annabhimoju, W. Sherman, Protein and
5-hydroxy-2H-pyridazin-3-ones: part 4. Optimization of DMPK properties, ligand preparation: parameters, protocols, and influence on virtual screening
Bioorg. Med. Chem. Lett. 18 (2008) 3421e3426. enrichments, J. Comput. Aided Mol. Des. 27 (3) (2013) 221e234.
[37] A. Maynard, R.M. Crosby, B. Ellis, R. Hamatake, Z. Hong, B.A. Johns, K.M. Kahler, [48] R.A. Friesner, J.L. Banks, R.B. Murphy, T.A. Halgren, J.J. Klicic, D.T. Mainz,
C. Koble, A. Leivers, M.R. Leivers, A. Mathis, A.J. Peat, J.J. Pouliot, C.D. Roberts, M.P. Repasky, E.H. Knoll, D.E. Shaw, M. Shelley, J.K. Perry, P. Francis,
V. Samano, R.M. Schmidt, G.K. Smith, A. Spaltenstein, E.L. Stewart, P.S. Shenkin, Glide: a new approach for rapid, accurate docking and scoring. 1.
P. Thommes, E.M. Turner, C. Voitenleitner, J.T. Walker, G. Waitt, Method and assessment of docking accuracy,, J. Med. Chem. 47 (2004)
J. Weatherhead, K. Weaver, S. Williams, L. Wright, Z.Z. Xiong, D. Haigh, 1739e1749.
J.B. Shotwell, Discovery of a potent boronic acid derived inhibitor of the HCV [49] The PyMOL Molecular Graphics System, Version 1.2r3pre, Schro € dinger, LLC

Você também pode gostar