Communications
with CVB3 Nancy in HeLa cells (Figure 2A). Pretreatment of
cells with 36 prior to virus inoculation had no effect on virus-
induced plaque formation. In contrast, incubation of 36 with
cell-free virus resulted in strong loss of virus infectivity as
shown by high percentage of plaque reduction in Figure 2A.
Here, high concentrations of CVB3 Nancy were incubated with
or without the inhibitor in test medium for one hour at 378C.
Then the mixture was diluted 1:106 to obtain noneffective in-
hibitor concentrations and added to confluent cell monolayers
for virus adsorption.
Table 1. Impact of amino acid substitutions detected in compound 36-
resistant CVB3 Nancy variants on plaque inhibition in HeLa cells.
CVB3
Nancy
Amino acid
substitution
IC50 [mm][a]
Compd 36
Pleconaril[b]
Compd 25
wildtype
none
n.a.
n.a.
n.a.
n.a.
n.a.
n.a.
0.16Æ0.03
35.03Æ0.01
n.a.
0.25Æ0.06
3.43Æ0.97
n.a.
36-resistant
36-resistant
36-resistant
36-resistant
36-resistant
P1094S
S1190T
I1207N
I1207m
S2209C
n.a.
n.a.
n.a.
n.a.
31.53Æ7.89
32.47Æ4.59
Acute (>3 gkgÀ1) and chronic (12.5, 50 and 200 mgkgÀ1
doses, 30 days treatment in male BALB/c mice) toxicological
studies in mice showed no indications of adverse anatomical,
behavioral or physiological effects (see Table S3 in the Support-
ing Information). In vitro ADMET profiling of 36 (Table S4 in
the Supporting Information) included analysis of plasma pro-
tein binding and recovery (96.7% and 100%, respectively),
plasma stability (100%), mutagenicity (none), and hERG chan-
nel inhibition (low). Compound 36 is neither an inducer nor an
inhibitor of relevant cytochrome P450 isoforms. A 100% meta-
bolic stability of 36 in human liver microsomes was observed
(see the Supporting Information, Table S4) explaining in vitro
its efficacy after oral administration in mice. The in vitro data
were confirmed by pharmacokinetics studies in mice during
a 100 mgkgÀ1 per os administration. The in vivo results show
that 36 has druglike pharmacokinetic properties, as reported
in the Supporting Information (Table S5), and in part summar-
[a] Data represent the meanÆSD of at least three independent determi-
nations in plaque reduction assays performed in duplicate; [b] not active
(n.a.) due to naturally occurring pleconaril resistance (L1092).
Compound 36 strongly inhibited the cytopathic effect of the
pleconaril-resistant CVB3 Nancy (L1092[4c]) as well as pleconaril-
susceptible CVB3 97927 (I1092) with IC50 values of 0.16 and
1.05 mm, respectively (Figure 1A; see also the Supporting Infor-
mation).
We postulate that inhibitor binding to viral capsid proteins
leads to the prevention of virus adsorption and might also be
responsible for the observed antiviral effect during and after
adsorption. The detection of amino acid substitutions in VP1
(2P1094S, 2S1190T, 1I1207N, 1I1207M) and VP2 (2
S2209C) of high-level 36-resistant CVB3 Nancy variants sup-
ports this (Table 1). In agreement with pleconaril studies,[8]
a concentration of 1 mgmLÀ1 (2.7 mm) of 36 was used for their
isolation. In ten independently prepared pools of wild-type
CVB3 Nancy, the mean frequency of resistance to 36 was ~7
105, corresponding well with the data published for pleconar-
il.[8] Based on these phenotypic and genotypic data, we expect
that the lipophilic 36 binds inside the hydrophobic pocket of
VP1 in a similar fashion to pleconaril and vapendavir.[7b,15] The
potential binding mode of 36 as derived by automated ligand
docking with GOLD[16] corroborates this mechanism of action
(Figure 2B,C). The model suggests that the observed mutations
causing high-level resistance induce larger conformational rear-
rangements leading to the disruption of hydrophobic interac-
tions formed by the trifluorophenyl moiety of 36 (which is
postulated to correspond to the methylisoxazole moiety in ple-
conaril).
ized
here:
Cmax
:
1.25 mgmLÀ1
;
tmax
:
3 h;
AUC0Àt
:
6418.3 ngmLhÀ1; AUCoÀ1: 9598.53 ngmLhÀ1; t1/2: 3.5 h; Cmax
/
AUC0Àt: 0.2 hÀ1; MRT: 3.54 h; Cl: 10.42 mLhkgÀ1; Kel: 0.18 hÀ1
;
Vd: 52.72 mLkgÀ1.
Effective compound concentrations were reached by oral ad-
ministration of 100 mgkgÀ1 of 36 twice daily, for 7 days. When
treatment was started 3 h prior to (prophylactically) or 1 and 3
days p. i. (therapeutically), 36 exerted a strong antiviral effect
in a model of CVB3-induced chronic myocarditis in NMRI mice
(Figure 3; see also, Tables S6 and S7 in the Supporting Informa-
tion). Placebo-treated, CVB3-infected mice markedly lost body
weight (Figure 3A) and became ill (Figure 3B) on day 3 p. i.
Prophylactic and therapeutic treatment significantly improved
both study parameters (Figure 3). As a result of viral replica-
tion, pronounced inflammation as well as necrosis and fibrosis
of myocardial tissue were observed in hematoxylin-eosin and
sirius red-stained heart specimens, respectively, in placebo-
treated animals on day 28 p. i. (Figure 3C). CVB3 also com-
pletely destroyed the exocrine pancreas tissue (Figure 3C).
Compound 36 significantly decreased tissue damage as well as
inflammation in the heart and pancreas when treatment was
started 3 h before, or 1 or 3 days p. i. No therapeutic effect
was observed when treatment started 7 days p. i. (results not
shown).
A panel of 46 RV serotypes comprising viruses from species
A (n=33) and B (n=13), two EV species A, nine EV species B,
one EV species C, and two EV species D were assessed regard-
ing their sensitivity to 36 in CPE inhibition assays in HeLa
cells.[13] Compound 36 inhibited 52 of the 60 strains tested
(Figure 1B). Strong antiviral activity was detected against
ECHO11 and EV68, which are known to cause outbreaks of
aseptic meningitis and acute lower respiratory tract infections,
respectively.[14] The highest activities were measured against
high-level pleconaril-resistant viruses including RV5, RV42,
RV44, RV48, and RV69.[4a,c,10] The IC50 values for these specific
EVs ranged from 0.04 to 0.64 mm (Table S2 in the Supporting
Information).
In conclusion, pyrazolopyrimidines represent a novel class of
compounds targeting the capsid of clinical important RVs and
EVs. Notably, lead compound 36 is orally available, inhibits
a broad-spectrum of EVs and RVs, helps to overcome the re-
sistance of known capsid inhibitors, and does not exert an
effect on relevant cytochrome isoforms. The favorable pharma-
The effect of 0.5 and 1 mm of 36 on different steps of the
viral replication cycle was studied in plaque reduction assays
ChemMedChem 2015, 10, 1629 – 1634
1631 ꢀ 2015 The Authors. Published by Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim