Вы находитесь на странице: 1из 6

ANTIMICROBIAL AGENTS AND CHEMOTHERAPY, Mar. 2001, p. 743–748 Vol. 45, No.

3
0066-4804/01/$04.00⫹0 DOI: 10.1128/AAC.45.3.743–748.2001
Copyright © 2001, American Society for Microbiology. All Rights Reserved.

Cyclopentane Neuraminidase Inhibitors with Potent In Vitro


Anti-Influenza Virus Activities
DONALD F. SMEE,* JOHN H. HUFFMAN, ANN C. MORRISON, DALE L. BARNARD,
AND ROBERT W. SIDWELL

Institute for Antiviral Research, Utah State University, Logan, Utah 84322-5600
Received 12 July 2000/Returned for modification 11 October 2000/Accepted 30 November 2000

A novel series of cyclopentane derivatives have been found to exhibit potent and selective inhibitory effects
on influenza virus neuraminidase. These compounds, designated RWJ-270201, BCX-1827, BCX-1898, and BCX-
1923, were tested in parallel with zanamivir and oseltamivir carboxylate against a spectrum of influenza A
(H1N1, H3N2, and H5N1) and influenza B viruses in MDCK cells. Inhibition of viral cytopathic effect ascer-
tained visually and by neutral red dye uptake was used, with 50% effective (virus-inhibitory) concentrations
(EC50) determined. Against the H1N1 viruses A/Bayern/07/95, A/Beijing/262/95, A/PR/8/34, and A/Texas/36/91,
EC50s (determined by neutral red assay) of the novel compounds were <1.5 ␮M. Twelve strains of H3N2 and
two strains of avian H5N1 viruses were inhibited at <0.3 ␮M. Influenza B/Beijing/184/93 and B/Harbin/07/94
viruses were inhibited at <0.2 ␮M, with three other B virus strains inhibited at 0.8 to 8 ␮M. The novel in-
hibitors were comparable in potency to (or slightly more potent than) zanamivir and oseltamivir carboxylate.

Downloaded from aac.asm.org by on January 12, 2009


No cytotoxicity was seen with the compounds at concentrations of <1 mM in cell proliferation assays. The
antiviral activity of RWJ-270201, chosen for clinical development, was studied in greater detail. Its potency and
that of oseltamivir carboxylate decreased with increasing multiplicity of virus infection. Time-of-addition
studies indicated that treatment with either compound needed to begin 0 to 12 h after virus exposure for op-
timal activity. Exposure of cells to RWJ-270201 caused most of the virus to remain cell associated, with ex-
tracellular virus decreasing in a concentration-dependent manner. This is consistent with its effect as a neur-
aminidase inhibitor. RWJ-270201 shows promise in the treatment of human influenza virus infections.

Influenza has continued to be a significant public health structure (26) have led to the identification of new inhibitors.
concern, with annual epidemics responsible for serious mor- A series of cyclopentane derivatives was found to cause potent
bidity and mortality (1, 13). Much attention has conse- and selective inhibition of influenza virus neuraminidase (2).
quently been given to the development of antiviral drugs for The chemical structures of the more potent antiviral com-
the treatment of this disease. Amantadine and rimantadine pounds (Fig. 1) have features in common with both zanamivir
both have been approved for prophylaxis of influenza A vi- and oseltamivir carboxylate but differ in having a five-mem-
rus infection (6). Ribavirin was shown to be effective against bered-ring structure. In this report the activities of these novel
experimental influenza virus infections in mice (9), and was compounds in vitro against various strains of influenza virus
studied in humans by small-particle aerosol delivery against are presented. Compound RWJ-270201 was evaluated in great-
severe influenza virus infections (11). However, it was not er detail in secondary assays, since it has been selected for
effective enough to receive drug approval. As early as 1976 clinical development.
Palese and Compans (19) reported an inhibitor of influenza
virus neuraminidase. This research was largely ignored for MATERIALS AND METHODS
many years, and it was not until recently that the search for Compounds. RWJ-270201, BCX-1827, BCX-1898, BCX-1923, zanamivir, and
more potent neuraminidase inhibitors has intensified. From oseltamivir carboxylate were synthesized at BioCryst Pharmaceuticals (Birming-
these investigations, zanamivir (GG167) and oseltamivir carbox- ham, Ala.). Ribavirin was obtained from ICN Pharmaceuticals (Costa Mesa,
Calif).
ylate (GS4071) emerged; these compounds were found to be Viruses. The following viruses were provided by H. Regnery of the Influ-
highly active against both influenza A and B viruses (10, 27). enza Branch of the Centers for Disease Control and Prevention (Atlanta, Ga.):
Zanamivir, a topical agent approved for clinical use, is effective A/Texas/36/91 (H1N1), A/Bayern/07/95 (H1N1), A/Beijing/262/95 (H1N1), A/
prophylactically and therapeutically for the treatment of influ- Washington/05/96 (H3N2), A/Johannesburg/33/94 (H3N2), A/Sydney/05/97
(H3N2), A/Shangdong/09/93 (H3N2), A/Beijing/32/92 (H3N2), B/Beijing/184/93,
enza (16, 17). Oseltamivir, the orally active prodrug form of
B/Panama/45/90, and B/Harbin/07/94. A/NWS/33 (H1N1) was provided by K.
oseltamivir carboxylate (22), is also clinically approved and has Cochran of the University of Michigan (Ann Arbor). A/PR/8/34 (H1N1) was
been found to be effective for both prophylaxis and treatment obtained from F. Schabel, Jr., Southern Research Institute (Birmingham, Ala.).
of influenza in humans (7, 18). A/Victoria/3/75 (H3N2), A/Port Chalmers/1/73 (H3N2), B/Hong Kong/5/72, and
Structure-activity analyses with the purified influenza virus B/Lee/40 were obtained from the American Type Culture Collection (Manassas,
Va.). A/Los Angeles/2/87 (H3N2) and A/Washington/897/80 (H3N2) were from
neuraminidase enzyme and knowledge of its three-dimensional Program Resources, Inc. (Rockville, Md.). A/X-31 (H3N2), a reassortment virus
containing hemagglutinin and neuraminidase genes from A/Aichi/2/68 (H3N2)
and the remainder of the genes from A/PR/8/34 (H1N1), was obtained from
* Corresponding author. Mailing address: Institute for Antiviral Re- E. Kilbourne, Mount Sinai School of Medicine, New York Medical College, City
search, Department of Animal, Dairy and Veterinary Sciences, Utah University of New York (New York, N.Y.). A/Port Chalmers/1/73r (H3N2), an
State University, 5600 Old Main Hill, Logan, UT 84322-5600. Phone: amantadine-resistant virus, was prepared from the wild-type virus by serial pas-
(435) 797-2897. Fax: (435) 797-3959. E-mail: dsmee@cc.usu.edu. sage in the presence of the drug in this laboratory. A/Virginia/2/88r (H3N2), a

743
744 SMEE ET AL. ANTIMICROB. AGENTS CHEMOTHER.

Downloaded from aac.asm.org by on January 12, 2009


FIG. 1. Chemical structures of cyclopentane derivatives, zanamivir, and oseltamivir carboxylate.

clinically isolated amantadine-resistant virus, was provided by F. Hayden, Uni- determined by the method of Finter (5), using a computerized EL-309 micro-
versity of Virginia School of Medicine (Charlottesville). A/Duck/MN/1525/81 plate autoreader (Bio-Tek Instruments, Winooski, Vt.). Antiviral activity was
(H5N1) and A/Gull/PA/4175/83 (H5N1) were obtained from R. Webster of the expressed as the 50% effective (virus-inhibitory) concentration (EC50) deter-
St. Jude Children’s Research Hospital (Memphis, Tenn.). All viruses were pas- mined by plotting compound concentration versus percent inhibition on semi-
saged in cells to prepare pools for use in these experiments. logarithmic graph paper. Although the CPE and NR methods were both used for
Cells and media. Madin-Darby canine kidney (MDCK) cells were grown in calculating EC50 against all of the influenza virus strains, for brevity only data
antibiotic-free minimum essential medium with nonessential amino acids (Gibco, obtained from the NR assays are reported. In general, the EC50 determined by
Long Island, N.Y.) containing 5% fetal bovine serum (HyClone Laboratories, NR assay were two- to fourfold higher than those obtained by the CPE method.
Logan, Utah) and 0.1% NaHCO3. Test medium consisted of minimum essential Cytotoxicity of compounds was assessed in parallel with the antiviral determi-
medium 0.18% NaHCO3, 10 U of trypsin per ml, 1 ␮g of EDTA per ml, and 50 nations in the same microplates, except in the absence of virus. From these
␮g of gentamicin/ml. results, 50% cytotoxic end points (50% cell-inhibitory concentrations [IC50s])
Cell culture assays. Three methods were used to assay antiviral activity in were determined. Later, the compounds were assayed for toxicity in actively
vitro: inhibition of virus-induced cytopathic effect (CPE) determined by visual proliferating MDCK cells. This was done by seeding 96-well microplates with 2 ⫻
(microscopic) examination of the cells, increase in neutral red (NR) dye uptake 104 cells per well. Compounds were diluted in medium containing 5% fetal
into cells, and virus yield reduction. In the CPE inhibition method, which was bovine serum and then were placed into the wells following cell attachment.
reported previously by Sidwell and Huffman (21), seven concentrations of test After 3 days, the percent inhibition of cell proliferation was assessed by NR assay
drug were evaluated against each virus in 96-well flat-bottomed microplates. The as described above.
compounds were added 5 to 10 min prior to virus, which was used at a concen- Virus yield reduction assays were performed by a method which separated and
tration of approximately 50 cell culture 50% infections doses per well. This virus quantified extracellular (supernatant) from cell-associated virus. These tests,
challenge dose equated to a multiplicity of infection (MOI) of approximately using A/Texas/36/91 (H1N1), A/Sydney/05/97 (H3N2), and B/Beijing/184/93 vi-
0.001 infectious particle per cell. The tests were read after incubation at 37°C for ruses, were initiated in 24-well plates of MDCK cells infected at a virus MOI of
72 h. In the NR uptake assay, dye (0.34% concentration in medium) was added 0.001. A visual determination of viral CPE was made after 72 h of incubation,
to the same set of plates used to obtain the visual scores. After 2 h, the color when cell destruction in untreated cultures was maximum, at which time the
intensity of the dye absorbed by and subsequently eluted from the cells was extracellular medium was removed and placed in test tubes. The plates were
VOL. 45, 2001 CYCLOPENTANE INHIBITORS OF INFLUENZA VIRUSES 745

TABLE 1. Activities of cyclopentane derivatives, zanamivir, and oseltamivir carboxylate on influenza virus replication
in MDCK cells as determined by NR assay
Oseltamivir
RWJ-270201 BCX-1827 BCX-1898 BCX-1923 Zanamivir
Virus carboxylate

EC50a(␮M) SIb EC50 SI EC50 SI EC50 SI EC50 SI EC50 SI

H1N1
A/Bayern/07/95 1.0 ⬎1,000 0.64 ⬎1,560 0.36 ⬎2,770 0.72 ⬎1,390 3.4 ⬎290 2.7 ⬎370
A/Beijing/262/95 0.36 ⬎2,770 0.56 ⬎1,780 0.18 ⬎5,550 0.37 ⬎2,700 2.6 ⬎380 2.4 ⬎410
c
A/NWS/33 21 ⬎47 19 ⬎52 21 ⬎47 23 ⬎43 ⬎100 — ⬎100 —
A/PR/8/34 1.5 ⬎660 1.0 ⬎1,000 0.7 ⬎1,420 0.7 ⬎1,420 0.42 ⬎2,380 0.22 ⬎4,540
A/Texas/36/91 0.09 ⬎11,110 0.10 ⬎10,000 0.06 ⬎16,660 0.11 ⬎9,090 0.22 ⬎4,540 0.17 ⬎5,880

H3N2
A/Beijing/32/92 0.07 ⬎14,280 0.14 ⬎7,140 0.05 ⬎20,000 0.05 ⬎20,000 0.65 ⬎1,530 0.23 ⬎4,340
A/Johannesburg/33/94 0.16 ⬎6,200 0.1 ⬎10,000 0.09 ⬎11,110 0.05 ⬎20,000 0.62 ⬎1,610 0.50 ⬎2,000
A/Los Angeles/2/87 0.07 ⬎14,280 0.07 ⬎14,280 0.04 ⬎25,000 0.06 ⬎16,660 0.18 ⬎5,550 0.38 ⬎2,630
A/Port Chalmers/1/73 0.07 ⬎14,280 0.02 ⬎50,000 0.06 ⬎16,660 0.03 ⬎33,330 0.15 ⬎6,660 0.07 ⬎14,280
A/Port Chalmers/1/73r 0.08 ⬎12,500 0.10 ⬎10,000 0.1 ⬎1,800 0.1 ⬎10,000 0.15 ⬎6,660 0.2 ⬎5,000
A/Shangdong/09/93 0.17 ⬎5,880 0.18 ⬎5,550 0.23 ⬎4,340 0.28 ⬎3,570 0.50 ⬎2,000 0.32 ⬎3,120
A/Sydney/05/97 0.19 ⬎5,260 0.13 ⬎7,690 0.10 ⬎1,000 0.22 ⬎4,540 0.45 ⬎2,220 0.31 ⬎3,220
A/Victoria/3/75 0.10 ⬎10,000 0.06 ⬎16,660 0.07 ⬎14,280 0.06 ⬎16,660 0.41 ⬎2,440 0.06 ⬎16,660
A/Virginia/2/88r ⬍0.01 ⬎100,000 ⬍0.01 ⬎100,000 0.03 ⬎33,330 0.05 ⬎20,000 0.02 ⬎50,000 ⬍0.01 ⬎100,000
A/Washington/897/80 ⬍0.01 ⬎100,000 ⬍0.01 ⬎100,000 ⬍0.01 ⬎100,000 ⬍0.01 ⬎100,000 ⬍0.01 ⬎100,000 ⬍0.01 ⬎100,000
A/Washington/05/96 0.02 ⬎50,000 0.01 ⬎100,000 ⬍0.01 ⬎100,000 0.01 ⬎100,000 0.2 ⬎50,000 0.08 ⬎12,500
A/X-31 0.01 ⬎100,000 ⬍0.01 ⬎100,000 ⬍0.01 ⬎100,000 ⬍0.01 ⬎100,000 0.4 ⬎2,500 0.12 ⬎8,330

Downloaded from aac.asm.org by on January 12, 2009


H5N1
A/Duck/MN/1525/81 0.01 ⬎100,000 0.022 ⬎45,450 0.02 ⬎50,000 0.01 ⬎100,000 0.20 ⬎5,000 0.22 ⬎4,540
A/Gull/PA/4175/83 0.02 ⬎50,000 0.03 ⬎33,330 0.02 ⬎50,000 0.02 ⬎50,000 0.22 ⬎4,540 0.26 ⬎3,840

B
Beijing/184/93 0.06 ⬎16,660 0.09 ⬎11,110 0.02 ⬎50,000 0.02 ⬎50,000 0.03 ⬎33,330 0.11 ⬎9,090
Harbin/07/94 0.12 ⬎8,330 0.16 ⬎6,250 0.06 ⬎16,660 0.06 ⬎16,660 0.20 ⬎5,000 0.26 ⬎3,840
Hong Kong/5/72 2.2 ⬎450 2.4 ⬎410 2.0 ⬎500 1.8 ⬎550 1.0 ⬎1,000 2.5 ⬎400
Lee/40 3.2 ⬎310 8.0 ⬎125 8.0 ⬎125 1.7 ⬎580 0.6 ⬎1,660 3.0 ⬎330
Panama/45/90 2.3 ⬎430 2.8 ⬎350 0.8 ⬎1,250 1.6 ⬎620 1.3 ⬎770 1.5 ⬎660
a
Results are means from two or three independent assays. Assay variability ranged from 20 to 50%.
b
Determined by dividing the IC50 (which was ⬎1,000 ␮M for all of these compounds) by the EC50.
c
—, the SI cannot be calculated, since the highest concentration tested (100 ␮M) was less than the IC50.

refed fresh medium. The tubes containing extracellular virus and cell debris were influenza A (H5N1), and influenza B viruses in MDCK cell
centrifuged at 3,200 ⫻ g for 5 min, and most of the supernatant fluid was culture by the NR method (Table 1). Of the influenza A
removed and transferred to unused tubes. This procedure was carefully done to
avoid collecting any of the cell pellets. The remainder of the medium from each
(H1N1) viruses, the A/Texas/36/91 strain was the most sensi-
tube was discarded, and the resulting pellets were recombined with the fresh tive to inhibition by the compounds, with EC50s ranging from
medium (and adhered cells) from wells where they originated. Eight wells were 0.06 to 0.22 ␮M. The A/Bayern/07/95, A/Beijing/262/95, and
used per concentration of compound. Samples from the eight wells were paired, A/PR/8/34 viruses were sensitive to inhibition in the 0.18 to 3.4
yielding a total of four samples for titration. The plates of cells and tubes of
extracellular virus were frozen and thawed, sonicated for 30 s each, and then
␮M range. Activities against the A/NWS/33 virus were up to an
assayed for virus titer. Titrations were conducted by adding the serially diluted order of magnitude less, at 19 to ⬎100 ␮M. Overall, zanamivir
samples to four wells each (0.1 ml/well) in 96-well plates of MDCK cells. After and oseltamivir carboxylate were slightly less potent (usually
2 h of virus adsorption, the medium was replaced with fresh medium to remove threefold or less) than the cyclopentane derivatives against the
residual compound present in the original samples. The plates were checked for
virus-induced CPE on days 3 and 6. Quantitation of virus yield titers was by the
H1N1 viruses.
end point method of Reed and Muench (20), and the titers were expressed as Twelve influenza A (H3N2) strains were inhibited by the
log10 50% cell culture infectious doses per 0.1 ml of medium assayed. cyclopentane derivatives at ⬍0.3 ␮M. The activities of zana-
Effects of MOI and delay of treatment initiation on antiviral activity. Exper- mivir and oseltamivir carboxylate were similar against these
iments using CPE inhibition and confirmed by NR uptake were done to ascertain
the effects of various viral challenge doses on antiviral, potency using MOIs of
viruses, with 50% inhibition at 0.65 ␮M or less. The A/Wash-
0.00018, 0.0009, 0.0045, and 0.0225. To examine the influence of delay of treat- ington/897/80, A/Washington/05/96, and A/X-31 strains were
ment initiation on antiviral activity. MOI of approximately 0.001 was used. uniformly more sensitive to inhibition by the cyclopentane
Compounds were added to the cells at 24 h pre-virus exposure and then rinsed inhibitors than were the other viruses.
off, added at 5 min pre-virus exposure (time zero), or added at 2, 4, 6, 8, 12, or 24 h
post-virus exposure. EC50 were calculated from the NR assay results as described
Because of the recent emergence of an influenza A (H5N1)
above. Influenza A/Sydney/05/97 (H3N2) virus was used for these studies. virus from chickens that was transmitted to humans and re-
sulted in lethal consequences (25), the neuraminidase inhibi-
RESULTS tors were evaluated against two strains of influenza A (H5N1)
virus. Both the A/duck/MN/1525/81 and A/gull/PA/4175/83 vi-
Antiviral activities against influenza A and B virus strains. ruses were markedly inhibited by the cyclopentane derivatives
Six neuraminidase inhibitors were evaluated for activity against at 0.01 to 0.03 ␮M. Zanamivir and oseltamivir carboxylate
several strains of influenza A (H1N1), influenza A (H3N2), were 10-fold less potent (0.2 to 0.26 ␮M) than the cyclopen-
746 SMEE ET AL. ANTIMICROB. AGENTS CHEMOTHER.

TABLE 2. Effect of time of treatment initiation on the concentrations were required to inhibit virus-induced cytopa-
anti-influenza A/Sydney/05/97 (H3N2) virus activities thology. These studies were done with a low input MOI, indi-
of RWJ-270201, oseltamivir carboxylate,
and ribavirin in MDCK cells cating that early treatments were necessary to suppress or
contain the later rounds of virus replication.
Time of treatment EC50a (␮M)
relative to
Effect of virus MOI. Certain compounds which inhibit virus
virus infection
RWJ-270201
Oseltamivir
Ribavirin
in cell cultures infected at low MOI, often are less active (or
(h) carboxylate
even inactive) at higher virus-to-cell ratios. To explore this
⫺24–0b 27 ⫾ 4 100 ⫾ 27 490 ⫾ 102 possibility with these compounds, antiviral activity was deter-
0–72 0.01 ⫾ 0.006 0.02 ⫾ 0.003 8⫾2 mined over a range of infecting MOIs differing five-fold from
2–72 0.03 ⫾ 0.006 0.02 ⫾ 0.004 20 ⫾ 6
each other (Table 3). RWJ-270201 and oseltamivir carboxylate
4–72 0.02 ⫾ 0.004 0.04 ⫾ 0.014 9⫾2
6–72 0.06 ⫾ 0.02 0.02 ⫾ 0.003 22 ⫾ 4 were most potent when virus infections were initiated at low
8–72 0.02 ⫾ 0.004 0.03 ⫾ 0.004 26 ⫾ 6 MOIs, and activities decreased with increasing viral challenge
12–72 0.2 ⫾ 0.03 0.06 ⫾ 0.006 53 ⫾ 8 dose. In contrast, the efficacy of ribavirin was not influenced by
24–72 65 ⫾ 36 ⬎100 530 ⫾ 120
increasing the MOI. This phenomenon has been previously
a
Determined by NR assay. Results are means and standard deviations from reported for ribavirin against other viruses (24).
four replicates.
b
The inhibitor was removed and cells were rinsed twice prior to infection. Virus yield reduction studies. Because neuraminidase is in-
volved in the efficient release of mature viruses from cells, mu-
tant viruses lacking neuraminidase activity aggregate and re-
tane derivatives but were still highly active inhibitors of these main at the cell surface (12, 14). Treatment with a neuraminidase
viruses. inhibitor should produce the same effect. To demonstrate this,

Downloaded from aac.asm.org by on January 12, 2009


The influenza B/Beijing/262/95 and B/Harbin 07/94 strain three influenza viruses were exposed to RWJ-270201 for 3
were sensitive to inhibition by these compounds (EC50s rang- days, followed by assay of extracellular and cell-associated vi-
ing from 0.02 to 0.26 ␮M). However, three other strains (Hong rus yields (Fig. 2). Ribavirin was evaluated in parallel, it being
Kong/5/72, Panama/45/90, and Lee/40) of influenza B virus an influenza virus inhibitor with different modes of action
were inhibited at 30- to 400-fold-higher concentrations (0.6 to (4, 28) unrelated to inhibition of viral neuraminidase. The
8 ␮M). Zanamivir and oseltamivir carboxylate were as potent A/Texas/36/91 (H1N1), A/Sydney/05/97 (H3N2), and B/Beijing/
as the cyclopentane derivatives against the influenza B virus 184/93 viruses showed a similar pattern, in that more virus was
strains. found extracellularly than cell associated in untreated cultures.
Antiviral selectivities. None of the compounds exhibited RWJ-270201 blocked the production of extracellular virus in a
cytotoxicity in MDCK cells as determined by visual and NR dose-dependent manner. As expected, large amounts of cell-
assay methods at concentrations of up to 1,000 ␮M. In addi- associated virus were present. EC90s for A/Texas, A/Sydney,
tion, actively dividing cells were not inhibited in their growth at
and B/Beijing were 0.15, 0.1, and 10 ␮M, respectively. These
a 1,000 ␮M concentration of each compound. By dividing this
values approximated the EC50s obtained against these viruses
concentration (1,000 ␮M) by the EC50s, selectivity indices (SIs)
from the NR assays (Table 1). In contrast, cell-associated virus
were obtained, and these are reported in Table 1. For highly
yields were not reduced at ⬎10, ⬎10, and ⬎100 ␮M, respec-
sensitive viruses such as influenza A/Washington/897/80 (H3N2),
A/Virginia/2/88r (H3N2), and A/duck/MN/1525/81 (H5N1), SIs tively, against the three viruses. Ribavirin caused dose-depen-
were ⬎100,000. SIs against most other viruses were less (⬎125 dent inhibition of both extracellular and cell-associated virus
to ⬎50,000) but were still high compared to those of most yields. The EC90s of ribavirin against extracellular A/Texas,
nucleoside-type antiviral agents. Antiviral selectivities were A/Sydney, and B/Beijing viruses were 45, 32, and 5.5 ␮M,
least against the A/NWS/33 (H1N1) virus (⬎43 to ⬎52 or less) respectively. EC90s of this compound against cell-associated
due to lower potencies. virus yields were 60, 32, and 12 ␮M, respectively. This is con-
Time-of-addition studies. Compounds have a certain span of sistent with virus yield reduction results using ribavirin against
time when they are most active; this is related to the life cycle other types of viruses (8).
of the virus and the step in the cycle where the compounds act.
The neuraminidase inhibitors RWJ-270201 and oseltamivir
carboxylate were compared with ribavirin (a nucleoside ana-
log) for inhibition of influenza A/Sydney/05/97 (H3N2) virus
when added at a different times relative to virus infection TABLE 3. Effect of virus MOI on the anti-influenza
A/Sydney/05/97 (H3N2) virus activities of RWJ-270201,
(Table 2). Treatment of cells starting at the time of infection oseltamivir carboxylate, and ribavirin in MDCK cells
required a ⬍0.01 ␮M concentration of the neuraminidase in-
hibitors and 5.7 ␮M ribavirin for inhibition of viral CPE. Treat- EC50a (␮M)
MOI
ment of cells prior to infection required much higher concen- RWJ-270201 Oseltamivir carboxylate Ribavirin
trations of the compounds, indicating a lack of persistence
0.00018 0.01 ⫾ 0.006 0.01 ⫾ 0.003 20 ⫾ 2
following their removal from the culture medium. Treatments 0.0009 0.1 ⫾ 0.02 0.07 ⫾ 0.05 22 ⫾ 2
with the three compounds could begin at any time from 2 to 12 0.0045 1.0 ⫾ 0.2 0.9 ⫾ 0.2 24 ⫾ 3
h post-virus infection and still require about the same concen- 0.0225 ⬎10 ⬎10 24 ⫾ 4
tration to achieve 50% inhibition as was effective for the 0-h a
Determined by NR assay. Results are means and standard deviations from
time point of treatment initiation. However, by 24 h, higher four replicates.
VOL. 45, 2001 CYCLOPENTANE INHIBITORS OF INFLUENZA VIRUSES 747

Downloaded from aac.asm.org by on January 12, 2009


FIG. 2. Cell-associated (F) and extracellular (■) virus yields produced in the presence of RWJ-270201 and ribavirin. (A and B) Influenza
A/Texas/36/91 (H1N1) virus; (C and D) influenza A/Sydney/05/97 (H3N2) virus; (E and F) influenza B/Beijing/184/93 virus. Data points represent
means from four samples ⫾ standard deviations.

DISCUSSION the influenza A/NWS/33 (H1N1) virus, which we found to be


relatively insensitive to inhibition in vitro compared to other
The results of this study show that the cyclopentane deriva- viral strains, is highly sensitive to treatment with zanamivir and
tives were active against a large number of influenza A and B oseltamivir in mice (22).
virus strains in cell culture at nontoxic concentrations. The po- High levels of cell-associated virus were produced in the
tencies of these compounds were similar to or slightly greater
presence of RWJ-270201. Yet, considerably less infectious vi-
than those of zanamivir and oseltamivir carboxylate. Only the
rus was detected in the extracellular medium relative to virus
A/NWS/33 (H1N1) virus was clearly less sensitive to inhibition
yields from inhibitor-free cultures. It was shown with neur-
by these compounds. Viruses with greater resistance to in-
aminidase-deficient influenza virus mutants that the viruses are
hibition are in the process of being prepared by cell culture
passage in the presence of RWJ-270201. These viruses will not fully released from cells but remain clumped at the cell
be examined to determine whether the insensitivity is due to surface (12, 14). An analogous situation should occur with
a resistant neuraminidase protein, altered hemagglutinin or wild-type virus replication in the presence of a neuraminidase
both. Resistance to neuraminidase inhibitors has been the sub- inhibitor. Clumping is caused because viruses have picked up
ject of considerable investigation, as recently reviewed (15). cell antigens that attract other viruses, resulting in their aggre-
Some of the viruses resistant in vitro are inhibited in mice and gation. The function of neuraminidase is to cleave off these
ferrets by these types of compounds (3), indicating dependence cellular residues so that the virus particles are not attracted to
on the enzyme for spread and disease progression in vivo. Even one another.
748 SMEE ET AL. ANTIMICROB. AGENTS CHEMOTHER.

In cell culture studies, the potencies of RWJ-270201 and 1973. Suppression by 1-␤-D-ribofuranosyl-1,2,4-triazole-3-carboxamide (Vi-
razole, ICN 1229) of influenza virus-induced infections in mice. Antimicrob.
oseltamivir carboxylate were dependent upon the time of ini-
Agents Chemother. 3:517–522.
tiation of treatment and the virus MOI. In order to prevent 10. Kim, C. U., W. Lew, M. A. Williams, H. Liu, L. Zhang, S. Swaminathan, N.
neuraminidase activity leading to inefficient virus release (or to Bischofberger, M. S. Chen, D. B. Mendel, C. Y. Tai, W. G. Laver, and R. C.
Stevens. 1997. Influenza neuraminidase inhibitors possessing a novel hydro-
cause virus aggregation as described above), treatments with phobic interaction in the enzyme active site: design, synthesis, and structural
the compound needed to be initiated within 12 h of infection. analysis of carbocyclic sialic acid analogue with potent anti-influenza activity.
Increasing the MOI increased the number of cells initially J. Am. Chem. Soc. 119:681–690.
11. Knight, V., S. Z. Wilson, J. M. Quarles, S. E. Greggs, H. W. McClung, B. K.
infected. These cells were not spared by treatment, and high Waters, R. W. Cameron, J. M. Zerwas, and R. B. Couch. 1981. Ribavirin
levels of cell-associated virus were produced. Treatment would small-particle aerosol treatment of influenza. Lancet ii:945–949.
protect neighboring cells during the secondary infection. Thus, 12. Liu, C., M. C. Eichelberger, R. W. Compans, and G. M. Air. 1995. Influenza
type A virus neuraminidase does not play a role in viral entry, replication,
under low-MOI conditions, a larger number of cells were not assembly, or budding. J. Virol. 69:1099–1106.
initially infected and would have escaped later infection by 13. Lui, K. J., and A. P. Kendal. 1987. Impact of influenza epidemics on mor-
antiviral treatment. These results suggest that patients would tality in the United States from October 1972 to May 1985. Am. J. Public
Health 77:712–716.
most benefit by treatment early in the course of influenza 14. Luo, G., J. Chung, and P. Palese. 1993. Alterations of the stalk of the
illness prior to developing high respiratory tract virus titers. influenza virus neuraminidase: deletions and insertions. Virus Res. 29:141–
RWJ-270201, which has been selected for clinical develop- 153.
15. McKimm-Breschkin, J. L. 2000. Resistance of influenza viruses to neuramin-
ment, may prove to be effective in humans based upon recent idase inhibitors—a review. Antiviral Res 47:1–17.
results of animal studies. Our experiments with this compound, 16. Monto, A. S., D. M. Fleming, D. Henry, R. de Groot, M. Makela, T. Klein, M.
which are being published separately (23), indicate efficacy in Elliott, O. N. Keene, and C. Y. Man. 1999. Efficacy and safety of the neur-
aminidase inhibitor zanamivir in the treatment of influenza A and B virus
treating influenza virus infections in mice when administered infections. J. Infect. Dis. 180:254–261.
orally. RJW-270201 is well tolerated in mice, as are the Food 17. Monto, A. S., D. P. Robinson, M. L. Herlocher, J. M. Hinson Jr., M. J.

Downloaded from aac.asm.org by on January 12, 2009


Elliott, and A. Crisp. Zanamivir in the prevention of influenza among
and Drug Administration-approved neuraminidase inhibitors
healthy adults: a randomized controlled trial. JAMA 282:31–35.
zanamivir and oseltamivir (22). RWJ-270201 appears to be a 18. Nicholson, K. G., F. Y. Aoki, A. D. Osterhaus, S. Trottier, O. Carewicz, C. H.
viable candidate for the treatment of influenza infections in Mercier, A. Rode, N. Kinnersley, and P. Ward. 2000. Efficacy and safety of
oseltamivir in treatment of acute influenza: a randomised controlled trial.
humans. Lancet 355:1845–1850.
19. Palese, P., and R. W. Compans. 1976. Inhibition of influenza virus replication
in tissue culture by 2-deoxy-2,3-dehydro-N-trifluoroacetylneuraminic acid
ACKNOWLEDGMENTS
(FANA): mechanism of action. J. Gen. Virol. 33:159–163.
This work was supported by contract N01-AI-85348 from the Virol- 20. Reed, L. J., and M. Muench. 1938. A simple method of estimating fifty
ogy Branch, National Institute of Allergy and Infectious Diseases, percent end points. Am. J. Hyg. 27:493–498.
National Institutes of Health, and by a grant from The R. W. Johnson 21. Sidwell, R. W., and J. H. Huffman. 1971. Use of disposable micro tissue
culture plates for antiviral and interferon induction studies. Appl. Microbiol.
Pharmaceutical Research Institute.
22:797–801.
22. Sidwell, R. W., J. H. Huffman, D. L. Barnard, K. W. Bailey, M.-H. Wong, A.
REFERENCES Morrison, T. Syndergaard, and C. U. Kim. 1998. Inhibition of influenza virus
1. Anonymous. 1999. Update: influenza activity—worldwide, May–September infections in mice by GS4104, an orally effective influenza virus neuramini-
1999. Morb. Mortal. Wkly. Rep. 48:883–886. dase inhibitor. Antiviral Res. 37:107–120.
2. Babu, Y. S., P. Chand, S. Bantia, P. Kotian, A. Dehghani, Y. El-Kattan, T. H. 23. Sidwell, R. W., D. F. Smee, J. H. Huffman, D. L. Barnard, K. W. Bailey, J. D.
Lin, T. L. Hutchison, A. J. Elliott, C. D. Parker, S. L. Ananth, L. L. Horn, Morrey, and Y. S. Babu. 2001. In vivo influenza virus-inhibitory effects of
G. W. Laver, and J. A. Montgomery. 2000. BCX-1812 (RWJ-270201): dis- the cyclopentane neuraminidase inhibitor RWJ-270201. Antimicrob. Agents
covery of a novel, highly potent, orally active, and selective influenza neur- Chemother. 45:749–757.
aminidase inhibitor through structure-based drug design. J. Med Chem. 24. Smee, D. F., R. W. Sidwell, B. B. Barnett, and R. S. Spendlove. 1981.
43:3482–3486. Inhibition of rotaviruses by selected antiviral substances, p. 123–138. In S. D.
3. Blick, T. J., A. Sahasrabudhe, M. McDonald, I. J. Owens, P. J. Morley, R. J. Acres, S. D. Forman, and H. Fast (ed.), Proceedings of the Third Interna-
Fenton, and J. L. McKimm-Breschkin. 1998. The interaction of neuramin- tional Symposium of Neonatal Diarrhea. Veterinary Infectious Disease Or-
idase and hemagglutinin mutations in influenza virus in resistance to 4-gua- ganization, Saskatoon, Saskatchewan, Canada.
nidino-Neu5Ac2en. Virology 246:95–103. 25. Subbarao, K., A. Klimov, J. Katz, H. Regnery, W. Lim, H. Hall, M. Perdue,
4. Eriksson, B., E. Helgstrand, N. G. Johansson, A. Larsson, A. Misiorny, J. O. D. Swayne, C. Bender, J. Huang, M. Hemphill, T. Rowe, M. Shaw, X. Xu, K.
Noren, L. Philipson, K. Stenberg, G. Stening, S. Stridh, and B. Oberg. 1977. Fukuda, and N. Cox. 1998. Characterization of an avian influenza A (H5N1)
Inhibition of influenza virus ribonucleic acid polymerase by ribavirin triphos- virus isolated from a child with a fatal respiratory illness. Science 279:393–
phate. Antimicrob. Agents Chemother. 11:946–951. 396.
5. Finter, N. B. 1969. Dye uptake methods for assessing viral cytopathogenicity 26. Von Itzstein, M., W.-Y. Wu, G. B. Kok, M. S. Pegg, J. C. Cyason, B. Jin, T. V.
and their application to interferon assays. J. Gen. Virol. 5:419–427. Phan, M. L. Smythe, H. F. White, S. W. Oliver, P. M. Colman, J. N.
6. Hayden, F. G. 1997. Antivirals for pandemic influenza. J. Infect. Dis 176 Varghese, D. M. Ryan, J. M. Woods, R. C. Bethell, V. J. Hotham, J. M.
(Suppl. 1):S56–S61. Cameron, and C. R. Penn. 1993. Rational design of potent sialidase-based
7. Hayden, F. G., J. J. Treanor, R. S. Fritz, M. Lobo, R. F. Betts, M. Miller, N. inhibitors of influenza virus replication. Nature 363:418–423.
Kinnersley, R. G. Mills, P. Ward, and S. E. Straus. 1999. Use of the oral 27. Woods, J. M., R. C. Bethell, J. A. V. Coates, N. Healy, S. A. Hiscox, B. A.
neuraminidase inhibitor oseltamivir in experimental human influenza: ran- Pearson, D. M. Ryan, J. Ticehurst, J. Tilling, S. M. Walcott, and C. R. Penn.
domized controlled trials for prevention and treatment. JAMA 282:1240– 1993. 4-Guanidino-2,4-dideoxy-2,3-dehydro-N-acetylneuraminic acid is a
1246. highly effective inhibitor both of the sialidase (neuraminidase) and of growth
8. Huffman, J. H., R. W. Sidwell, G. P. Khare, J. T. Witkowski, L. B. Allen, and of a wide range of influenza A and B viruses in vitro. Antimicrob. Agents
R. K. Robins. 1973. In vitro effect of 1-␤-D-ribofuranosyl-1,2,4-triazole-3- Chemother. 37:1473–1479.
carboxamide (Virazole, ICN 1229) on deoxyribonucleic acid and ribonucleic 28. Wray, S. K., B. E. Gilbert, M. W. Noall, and V. Knight. 1985. Mode of action
acid viruses. Antimicrob. Agents Chemother. 3:235–241. of ribavirin: effect of nucleotide pool alterations on influenza virus ribonu-
9. Khare, G. P., R. W. Sidwell, J. T. Witkowski, L. N. Simon, and R. K. Robins. cleoprotein synthesis. Antiviral Res. 5:29–37.

Вам также может понравиться