You are on page 1of 10

ARTICLES

New helicase-primase inhibitors as drug candidates for the


treatment of herpes simplex disease

GERALD KLEYMANN, RÜDIGER FISCHER, ULRICH A.K. BETZ, MARTIN HENDRIX,


WOLFGANG BENDER, UDO SCHNEIDER, GABRIELE HANDKE, PETER ECKENBERG, GUY HEWLETT,
VENIAMIN PEVZNER, JUDITH BAUMEISTER, OLAF WEBER, KERSTIN HENNINGER, JÖRG KELDENICH,
© 2002 Nature Publishing Group http://medicine.nature.com

AXEL JENSEN, JÖRG KOLB, UTE BACH, ANDREAS POPP, JUTTA MÄΒEN, ISABELLE FRAPPA,
DIETER HAEBICH, OSWALD LOCKHOFF & HELGA RÜBSAMEN-WAIGMANN

Bayer AG, Pharma Research, Wuppertal, Germany


Correspondence should be addressed to G.K.; email: gerald.kleymann@freenet.de

The vast majority of the world population is infected with at least one member of the human
herpesvirus family. Herpes simplex virus (HSV) infections are the cause of cold sores and genital
herpes as well as life-threatening or sight-impairing disease mainly in immunocompromized pa-
tients, pregnant women and newborns. Since the milestone development in the late 1970s of
acyclovir (Zovirax), a nucleosidic inhibitor of the herpes DNA polymerase, no new non-nucleo-
sidic anti-herpes drugs have been introduced. Here we report new inhibitors of the HSV heli-
case-primase with potent in vitro anti-herpes activity, a novel mechanism of action, a low
resistance rate and superior efficacy against HSV in animal models. BAY 57-1293
(N-[5-(aminosulfonyl)-4-methyl-1,3-thiazol-2-yl]-N-methyl-2-[4-(2-pyridinyl)phenyl]acetamide),
a well-tolerated member of this class of compounds, significantly reduces time to healing, pre-
vents rebound of disease after cessation of treatment and, most importantly, reduces frequency
and severity of recurrent disease. Thus, this class of drugs has significant potential for the treat-
ment of HSV disease in humans, including those resistant to current medications.

Eight human herpesviruses (HHV1 to HHV8) have been identi- of cross-resistance to nucleosides were the key criteria in the
fied so far1. One key feature of these viruses is their ability to re- search for the next generation of drugs directed against novel
main latent in their host after primary infection and to targets of herpesviruses.
reactivate intermittently from a pool of latently infected cells
upon diverse internal and external stimuli. The HHV genomes Identification and optimization of antiviral compounds
(dsDNA ≈ 125–230 kb in size) code for more than 50 genes that We have used a new fluorometric high-throughput screening
are essential for viral replication in vitro and/or in vivo and many (HTS) assay to identify active and tolerable substances in com-
viral gene products are potential targets for antiviral therapy. pound libraries that inhibit any target essential for viral replica-
High-sequence homologies have been identified in HHV genes tion in cell culture5,6. Efficacy of a test sample is measured by
encoding the helicase-primase enzyme complex, which is es- assessing the viability and proliferation of surviving cells in a
sential for DNA and viral replication2. herpes simplex virus (HSV)-infected cell culture with a fluores-
Acyclovir (trade name Zovirax)3, developed in the late 1970s, cent dye at the time when cell viability in the virus (no-drug)
was the first specific and selective antiviral drug. Newer nucleo- control approaches zero. Approximately 4.2 × 105 compounds
sidic drugs4 that are chemically related to acyclovir, such as pen- were tested at a concentration of 10 µM and several compound
ciclovir or their pro-drugs valacyclovir (Valtrex) and classes with antiviral activity were discovered. Starting with the
famcyclovir (Famvir), were launched in the mid 1990s. identified compound BAY 38-9489 (ref. 5), several thousand
However, nucleosides are pro-drugs that have to be phosphory- analogs and new compounds were synthesized during consecu-
lated by the viral thymidine kinase (TK) and subsequently by tive optimization cycles of synthesis, biological profiling and
cellular kinases in order to inhibit the viral DNA polymerase. If modeling techniques to optimize the thiazole urea BAY 38-9489
the virus does not express a functional TK (for example, a resis- to the lead structure BAY 44-5138 (ref. 5). BAY 44-5138 showed
tant HHV1 strain or TK– viruses such as human cy- first in vivo activity, and finally BAY 57-1293 (ref. 6) was opti-
tomegalovirus) or if the DNA polymerase does not have the mized and selected for pre-clinical development (Tables 1 and
optimal primary structure, the potency of the drug diminishes, 2). The structure–activity relationship (SAR) and selected exam-
the selectivity index is significantly smaller, higher doses have ples that illustrate the progress during the optimization phase
to be administered and adverse effects are more likely to be as- are summarized in Table 1 and 2 and briefly outlined below.
sociated with treatment. Until now, nucleosides were the only
compound class available for systemic treatment of herpes dis- SAR of the novel herpes inhibitors
ease and resistance was developing in the growing number of Replacement of the morpholino group in BAY 44-5138 by a
immunocompromized patients. Thus, potent anti-herpes activ- phenyl ring and the observation that BAY 44-5138 is demethy-
ity, efficacy (especially upon delayed treatment), safety and lack lated in vivo led to BAY 51-3295, the first compound that was

392 NATURE MEDICINE • VOLUME 8 • NUMBER 4 • APRIL 2002


ARTICLES

Fig. 1 Antiviral effect of inhibitors in vitro. Dose-dependent anti-herpes ac-


tivity of the screening hit BAY 38-9489 (IC50 = 0.5 µM) and development
candidate BAY 57-1293 (IC50 = 20 nM) compared with acyclovir (IC50 = 1
µM) determined with the fluorescence-based cytopathogenicity assay5,6. The
quotient of relative fluorescence units of an infected well in the presence of
test sample and the uninfected cell control is shown in percentage.
Representative results of 10 experiments are shown. The s.d. for a given con-
centration is generally between 2–8% and at concentrations approaching
the IC50 values, less than 25%. , acyclovir, HSV-1 F; , BAY 38-9489 HSV-1
F; , BAY 57-1293 HSV-1 F; , BAY 57-1293 HSV-1 clinical isolate; , BAY
57-1293 HSV-2 clinical isolate.
© 2002 Nature Publishing Group http://medicine.nature.com

methyl-2-[4-(2-pyridinyl)phenyl]acetamide with the sum for-


mula C18H18N4O3S2 and a molar mass of 402.5 g/mol. The com-
pound is a stable white powder at room temperature with a
more potent than valacyclovir in vivo. Exchanging the urea moi- melting point of 193 °C (decomposition).
ety with an amide group (BAY 54-6322) and introduction of the
primary sulfonamide resulted in BAY 56-1655, a remarkably po- Biological and medicinal profile of BAY 57-1293
tent compound in vitro (50% inhibitory concentration (IC50) < The concentration at which a drug reduced cytopathic effects
0.01 µM (HSV-1 F and HSV-2 G) and in vivo (50% effective dose (CPE) of viral replication (IC50) or cell viability (Tox50) in cell
(ED50) = 0.5 mg/kg (HSV-1 Walki) and 1.1 mg/kg (HSV-2 MS)). culture by 50% as well as the selectivity index (SI = Tox50/IC50), a
However, solubility of the compounds BAY 51-3295 and BAY 56- measure of tolerability in vitro, are listed in Table 1. The first
1655 was too low for simple intravenous formulation. The 2- identified compound, BAY 38-9489, was tolerable over a broad
pyridyl substituent in para-position of the phenyl ring in BAY concentration range and at least as active as acyclovir in vitro
57-1293 solved this problem and this compound was selected for (Fig. 1). The IC50 of the optimized compound BAY 57-1293 was
in-depth characterization of its biophysical properties as well as determined to be 0.012 ± 0.004 µM (median, sextuplicate exper-
its pharmacokinetic, pharmacodynamic and safety profiles. iment). The results of the fluorescence-based assay5,6 are compa-
rable with those obtained from conventional cytopathogenicity
Physical and chemical properties of BAY 57-1293 or plaque reduction assays7–9. The inhibitory dose (ID50), in-
The systematic name of BAY 57-1293 within the IUPAC nomen- hibitory or effective concentration (IC50 or EC50) of the plaque
clature is N-[5-(aminosulfonyl)-4-methyl-1,3-thiazol-2-yl]-N- reduction assay or the cytopathogenicity assay denotes the drug

Table 1 Pharmacological profile of the helicase primase inhibitors

In vitro viral replication assay


Acyclovir-resistant HSV-1 F Sequencing ATPase inhibition*
Compound Structural HSV-1 F HSV-1 F HSV-2 G HSV-2 G HSV-1 F mutant Mutants Resistance Results (at 1 mM ATP)
formula IC50 [µM] SI IC50 [µM] SI IC50 [µM] IC50 [µM] Index (RI) mutation(s)# IC50 [nM]
Cl
O

BAY 38-9489 O
N O
N
S
S N
O
0.5 500 0.7 500 0.5
N
N

UL5 M355V
O

BAY 44-5138 O
N O
N
S
S N
O
0.75 350 1.5 167 0.75 >250 >333 UL5 V662I 700
N
N
UL52 A897T

O
BAY 51-3295 O N S N
O
0.001 2.5x105 0.005 25000 0.001 >250 >250000 25
S
N N

BAY 54-6322 O N
O 0.0005 1 × 105 0.05 5000 0.0005 >250 >500000 UL5 K356N 120
S N
N S O >1 >2000 UL52 A897T

>8 >400 UL5 G352V


O
BAY 57-1293 N O

N
N

S
S N
O
0.02 2500 0.02 3000 0.02 >0.1 >5 UL5 M355T 30
>3 >150 UL5 K356Q

Acyclovir 1 250 4 125 125 1 on all no inhibition


BAY mutants <100000

Valtrex Val-acyclovir see Acyclovir see Acyclovir


IC50 (inhibitory concentration) and SI (selective index) values of HSV (wild-type and mutants) were determined at a m.o.i. of 0.0025 on Vero cells. *, The UL5 and UL52 genes of
wild-type HSV-1F were cloned into a Baculovirus expression system and kinetic analysis of the enzyme complex UL5-UL52-6xHis, purified from doubly infected Sf9 cells, revealed
the following kinetic constants with Km (ATP) = 500 µM, Kcat = Vmax/E0 = 362 min–1, and IC50 = 30 nM in the case of BAY 57-1293. Neither the spontaneous hydrolysis of ATP (back-
ground) nor the weak DNA-independent ATPase activity of the enzyme complex is altered in the presence of the thiazolyl amides such as BAY 57-1293 or BAY 56-165516. #, A re-
sistant virus containing 3 mutations was selected in the presence of compound BAY 44-5138, whereas single mutations were observed in the genome of 2 viruses resistant to BAY
54-6322 and 3 viruses resistant to BAY 57-1293.

NATURE MEDICINE • VOLUME 8 • NUMBER 4 • APRIL 2002 393


ARTICLES

concentration that reduces the number of viral plaques or viral-


Table 2 Antiviral activity of compounds in vivo (10 mice per group)
induced cytopathogenicity (CPE, lysis or morphological
changes of infected cells) by 50% as compared with the un- Compound Lethal challenge model
treated control (infected cell culture). The IC50 of the new fluo- ED50 [mg/kg], p.o., 3× daily
rescence-based cytopathogenicity assay stands for the drug HSV-1 Walki HSV-2 MS
concentrations that block the decay of the esterase activity of BAY 38-9489 49 (i.p.)
an infected cell culture in the presence of compound by 50% at BAY 44-5138 60 weak
the time when the esterase activity of the untreated control (in- BAY 51-3295 8 11
BAY 54-6322 1 3
fected cell layer) approaches zero (signal of control cells divided
BAY 57-1293 0.5 0.5
by signal of the virus control (infected cells) > 10). The fluores-
© 2002 Nature Publishing Group http://medicine.nature.com

Acyclovir 22 16
cence signal (esterase activity) corresponds with the number of Valacyclovir 17 14
vital or viable cells in the test well; figuratively speaking, the
p.o., per os; i.p., intraperitoneal.
IC50 is the drug concentration at which the microscopic picture
of an infected cell culture in the presence of compound shows
50% CPE. The IC50 values of the plaque reduction assay (IC50 Mechanism of action
acyclovir = 0.4–0.75 µM; IC50 BAY 57-1293 = 0.01–0.02 µM) and To elucidate the mechanism of action of the inhibitors pre-
the conventional cytopathogenicity assay (IC50 acyclovir = sented here, we analyzed the influence of compounds on indi-
0.5–1.5 µM; IC50 BAY 57-1293 = 0.01–0.03 µM) were determined vidual steps of the replication cycle at several time points after
for comparison on Vero cells infected with HSV-1 F. infection. In the presence of BAY 57-1293, the virus still infected
BAY 57-1293 is nearly two orders of magnitude more potent permissive cells and expression of viral immediate early genes
than acyclovir in vitro and the superiority was even more promi- was initiated. However, PCR, Southern-blot and in situ hy-
nent when the viral load was increased (BAY 57-1293 IC50 = bridization analyses demonstrated complete inhibition of viral
12 nM, 20 nM and 50 nM; acyclovir IC50 = 1 µM, 3 µM and DNA synthesis. In agreement with these findings, electron mi-
10–50 µM at a multiplicity of infection (m.o.i.) of 0.0025, 0.02 croscopy showed a total absence of DNA-containing C-capsids,
and 0.2, respectively). A minor increase in IC50 values at higher which normally bud from infected cells as enveloped infectious
viral loads was observed for all thiazolyl compounds listed in viruses. Reverse transcriptase (RT)-PCR experiments revealed
Table 1. BAY 57-1293 was also active against porcine (IC50 = 5 that only immediate early genes are expressed normally in the
µM) and bovine (IC50 = 0.12 µM) herpes strains; however, only presence of inhibitor, whereas early and late gene expression is
weak activity was observed against varicella-zoster virus or reduced, suppressed or not initiated (Fig. 2). In parallel to this
human cytomegalovirus. Inhibition of viral replication in vitro line of experiments, 10 drug-resistant HSV mutants were se-
was completely reversible upon removal of the drug whereas lected in the presence of 0.5–2.0 µM inhibitor. The frequency at
the delayed addition of inhibitor up to 6–10 hours post-infec- which mutant HSV-1 F viruses appeared in vitro (0.5–4.5 × 10–6) is
tion (p.i.) left the IC50 values nearly unchanged. Similar results at least one order of magnitude lower compared with acyclovir
upon delayed addition of inhibitor have been reported for acy- (1 × 10–3–1 × 10–4)10. When cells were transfected with a variety of
clovir, which indicates that both the thiazolyl and nucleoside PCR fragments obtained from drug-resistant viruses and subse-
compounds block viral replication during β gene expression, al- quently infected with wild-type virus in the presence of in-
beit as a result of different mechanisms. BAY 57-1293 was active hibitor, viral plaques were detected only when cells were
at an IC50 of 10–30 nM against all clinical isolates of HSV-1 transfected with a mutant UL5 or UL52 gene or a mix of mutant
(from 35 patients) and HSV-2 (from 19 patients) tested so far UL5 or UL52 and wild-type UL5, UL8 and UL52 genes, but not
with near identical IC50 values to those seen with HSV-1 F and when cells were transfected with a wild-type UL5, UL8 or UL52
HSV-2 G (Fig. 1 and Table 1) as well as HSV-1 Walki (IC50 = 20 gene (data not shown). From these complementation experi-
nM) or HSV-2 MS (IC50 = 15 nM). The same antiviral activity was ments, we concluded that the helicase-primase complex (UL5
also observed on permissive cell lines of epithelial (human em- and UL52 gene products) is the target of the thiazole ureas and
bryonic lung fibroblasts and normal human diploid fibroblasts) their corresponding amides. Furthermore, sequencing of the
and neuronal origin (NT-2, HSV-1 F, IC50 = 20 nM) when viral complete genome of a virus resistant to BAY 54-6322 revealed a
titers were determined on the respective cell lines and cell cul- single mutation (UL5, K356N). Sequencing of partial genomes
ture conditions were comparable. Finally, BAY 57-1293 was ac- and individual genes of nine other mutants localized all resis-
tive against acyclovir resistant mutants (Table 1), which display tance-conferring mutations to the UL5 and UL52 genes. The ob-
mutations in the TK and/or pol genes10, with nearly identical served mutations and resistance indices were irrespective of the
IC50 values observed for HSV-1 F or clinical isolates (Fig. 1). permissive cell type and are summarized in Table 1.

a b Fig. 2 Compounds inhibit early and late


gene expression. a–f, Agarose gel elec-
trophoresis of RT-PCR fragments prepared
from virus (HSV-1 F) infected cell cultures
c d (Vero cells) in the presence of compound BAY
57-1293 (a, c, e; 2 µM) shows inhibition of β
(c versus d; UL8, 12 p.i.) and γ (e versus f; UL13,
16 p.i.) gene expression. Identical results were
obtained with BAY 44-5138 (25 µM). α gene
e f expression is not altered in the presence of in-
hibitor (a versus b; UL54, 2 p.i.).

394 NATURE MEDICINE • VOLUME 8 • NUMBER 4 • APRIL 2002


ARTICLES

Fig. 3 Potency and efficacy of compounds in HSV-infected mice. a and b,


a Superior potency and efficacy of BAY 57-1293 compared with the nucleo-
side analog valacyclovir (Valtrex) in the murine lethal challenge12,13 (a) and
zosteriform spread model14 (b). a, Comparison of BAY 57-1293 with valacy-
clovir in the murine lethal challenge model. 10 anesthetized BALB/cABom
female mice per group were intranasally infected with a lethal dose of HSV-
2 MS and treated per os with BAY 57-1293 or valacyclovir once daily from
d0 to d4 post infection. Infected mice were inspected daily and a survival
curve was recorded. The comparison of survival curves was performed
using the generalized Wilcoxon test. All treatment groups yielded statisti-
cally significant different survival curves. , placebo; , valacyclovir 60
© 2002 Nature Publishing Group http://medicine.nature.com

mg/kg; , BAY 57-1293 7 mg/kg. b, Comparison of BAY 57-1293 with


valacyclovir in the zosteriform spread model14. The scarified skin of
C3H/TifBom-hr female mice was infected with 1 × 106 p.f.u. HSV-2G and
the disease score (mean of 10 animals per group; see Methods) was
b recorded daily. Delayed treatment (15 & 60 mg/kg BAY 57-1293 and 60 &
240 mg/kg valacyclovir, 0.5% Tylose/PBS, 3× daily, per os) commenced on
d3 post infection and continued until d7. Sum of disease scores were com-
pared between treatment groups using one-way ANOVA. In order to ac-
count for multiple testing, resulting P-values were adjusted according to
Bonferroni–Holm. Treatment with 15 mg/kg BAY 57-1293 was more effec-
tive compared with treatment with 240 mg/kg valacyclovir (P < 0.011).
, placebo; , valacyclovir 60 mg/kg; , valacyclovir 240 mg/kg; , BAY
57-1293 15 mg/kg; , BAY 57-1293 60 mg/kg.

rent disease was measured in terms of a disease score (Fig. 4e).


BAY 57-1293 not only completely suppressed clinical symp-
toms of primary disease (Fig. 4a–c) but also suppressed viral
shedding by 3 orders of magnitude to nearly undetectable levels
The mutations in the UL5 gene of HSV-1 were mainly found in vaginal swabs (mean plaque-forming units (p.f.u.) per ml log
between nucleotides 1045 to 1077 corresponding to amino 0.6 ± 0.6 for BAY 57-1293-treated animals in comparison to log
acids 349 to 359 (the α-helical stretch HEFGNLMKVLE), which 3.7 ± 0.8 for placebo-treated animals). Treatment of acute dis-
is also the region of highest homology among the UL5 se- ease also prevented recurrent disease symptoms in the follow-
quences of the herpesviridae and just adjacent to the conserved up period (Fig. 4e). Delayed initiation of therapy after
motif IV of the six protein domains required for helicase activ- appearance of vesicles showed that BAY 57-1293 was clearly
ity11. Mutant virus K356N replicated with near wild-type growth more efficacious than valacyclovir in reducing time to healing
rate and titers comparable to wild-type virus were obtained, (Fig. 4d) in guinea pigs.
whereas other mutants such as M355T were strongly impaired
with respect to growth rate and titers. In addition to the com- Pharmacokinetic and safety profile of BAY 57-1293
plementation analysis and the sequencing results (Table 1), the When female BALB/c mice were treated with a single dose of
mechanism of action of BAY 57-1293 and congeners was also 1 mg/kg BAY 57-1293 per os as suspension in 0.5% tylose, a
confirmed by direct inhibition of the ATPase activity of the viral maximal concentration of 1.8 mg/l (4.4 µM) was reached 1 hour
helicase-primase enzyme complex in a dose-dependent manner after administration. Due to the slow elimination of BAY 57-
(Table 1). 1293 (t1/2 = 6.0 h), plasma concentrations above 100 µg/ml (0.25
µM) were detected up to 24 hours after dosing. Similar favorable
Superior anti-herpes activity of BAY 57-1293 in vivo pharmacokinetics with a high bioavailability (> 60%) were also
We used a murine model of disseminated herpes infection observed in rats and dogs.
(lethal challenge model)12,13 to assess the antiviral activity of the Exploratory toxicology and safety pharmacology studies did
compounds in vivo. Potential drug candidates were tested in es- not reveal any safety relevant findings at 30, 100 and 300 mg/kg
calating doses and the ED50 was determined. The results for se- BAY 57-1293 (once daily per os) in a 4-week chronic toxicity
lected compounds are summarized in Table 2. After oral (per os) study in dogs. However, administration of the same dose to rats
administration BAY 57-1293 was at least 10 times more potent for 4 weeks resulted in a dose-dependent transitional hyperplasia
than valacyclovir (Table 2), even when applied once daily (Fig. of the urinary bladder epithelium. The N-[5-(aminosulfonyl)-4-
3a). Superior efficacy and potency of BAY 57-1293 was also methyl-1,3-thiazol-2-yl]-N-methylacetamide moiety of BAY 57-
demonstrated in a model of cutaneous HSV infection (Fig. 3b; 1293 resembles the structure of the diuretic drug Diamox
zosteriform spread model)14 when oral treatment was initiated (acetazolamide). Primary sulfonamides can inhibit rat, dog and
after onset of disease and herpes vesicles were already apparent. human carbonic anhydrase enzymes and are known to cause a
In particular, after treatment was stopped, the rebound of dis- species-specific hyperplasia in bladder epithelium of rodents,
ease was more effectively controlled in BAY 57-1293-treated ani- but not in other animals or humans. In contrast to acetazo-
mals. lamide, which has an IC50 of approximately 30 nM, BAY 57-1293
The guinea pig model of intravaginal HSV-2 infection resem- inhibits the carbonic anhydrase with an IC50 in the range of
bles the clinical situation in humans15. BAY 57-1293 was found 2–5 µM in the standard carbonic anhydrase–catalyzed CO2 hy-
superior to valacyclovir in this model when acute treatment dration test system (data not shown). Due to the high exposure,
(Fig. 4a–c), delayed treatment (Fig. 4d) or suppression of recur- which reached 100–300 µM in rats, it is likely that the hyperpla-

NATURE MEDICINE • VOLUME 8 • NUMBER 4 • APRIL 2002 395


ARTICLES

a b c d
© 2002 Nature Publishing Group http://medicine.nature.com

Fig. 4 Potency and efficacy of compounds in HSV-2-infected guinea pigs. Suppression of acute dis-
ease symptoms (photographed at d6 p.i.) of 10 vaginally infected (2.5 × 105 p.f.u. HSV-2 strain MS) fe-
male guinea pigs per group. a–c, Animals treated with BAY 57-1293 (20 mg/kg, disease score 0) (a), e
valacyclovir (150 mg/kg, disease score 2) (b) and placebo (disease score 4) (c). Redness, swelling and
herpetic vesicles are observed only in placebo- and valacyclovir-treated animals. d, Time to healing
after late treatment of vaginally HSV-2 infected guinea pigs15 with BAY 57-1293 () in comparison
with valacyclovir () and placebo (). Delayed treatment with BAY 57-1293 (20 mg/kg 2× daily per os,
treatment day 4–14) abrogates progression of disease symptoms (mean of 10 animals per group) of
HSV-2 infected guinea pigs within 1 d of treatment and healing is observed subsequently, whereas a
7.5−fold higher dose of valacyclovir (150 mg/kg 2× daily) shows marginal therapeutic efficacy com-
pared with placebo. Time to healing in the BAY 57-1293 group was significantly shorter than in the
valacyclovir-treated group. e, Influence of acute treatment on the recurrence rate of HSV-2 infections in
the guinea pig model15. BAY 57-1293 reduces recurrent HSV-2 disease almost completely when vagi-
nally HSV-2 infected guinea pigs were treated 2× daily per os with 30 mg/kg from d0–d4 (acute treat-
ment; treatment start 6 h post infection). In this experiment valacyclovir (3× daily with 100 mg/kg;
gray line) had no effect compared with placebo (dashed line), whereas BAY 57-1293 treated animals
(2× daily 30 mg/kg; black line) show only very few recurrent episodes. Graph shows cumulative disease
score over time of recurrent genital herpes in the guinea pig model (mean of 10 animals per group).

sia observed in rats is in fact due to the carboanhydrase inhibi- cation. The new selective and specific mechanism of action
tion by BAY 57-1293. meets this requirement and because it differs from the mecha-
nism of nucleosidic drugs it also satisfies the strong medical
Discussion need to combat nucleoside drug resistance, especially in im-
The best strategy for control of disease caused by pathogens munocompromised patients.
such as HSV would obviously be prevention. Rising incidence The remarkable potency of the new compound class may be
data show that it is difficult to avoid new infections due to the explained by the pattern of resistance-conferring mutations in
insufficient awareness of the problem within the population the genes encoding UL5 and or UL52. It can be concluded that
and the high prevalence and contagious nature of the virus. In the thiazolyl-amides bind to the helicase and primase subunits
view of the significant challenge associated with the develop- simultaneously thereby mimicking combination therapy with a
ment of a prophylactic vaccine that can block HSV infections single drug. Although all selected resistant viruses displayed
of mucosal surfaces and the limited use of antibodies, thera- mutations in the viral helicase and/or primase subunits we can-
peutic proteins and immunostimulants, treatment with an not rule out that a further target such as UL8 may be identified,
orally available, small-molecule antiviral drug amenable to thereby increasing the incidence of resistant viruses under se-
large-scale production is a proven option to treat HSV infec- lection pressure. However, the lower resistance rate in vitro com-
tions3,4,7. However, although the nucleosidic drugs on the mar- pared with acyclovir, which targets the viral thymidine kinase
ket represented a major breakthrough, their efficacy is limited, and DNA-polymerase, argues against this possibility.
particularly when they are administered late after onset of dis- The physicochemical properties of BAY 57-1293 permit the
ease. development of topical, oral and intravenous formulations to
The preclinical pharmacological profile of the new helicase- suppress or treat primary and recurrent HSV diseases such as
primase inhibitor BAY 57-1293 is superior to the current stan- herpes labialis, herpes genitalis, ocular herpes infections, en-
dard HSV treatment represented by Zovirax, Valtrex and Famvir cephalitis and disseminated or visceral HSV infections. The po-
with respect to all parameters of efficacy. BAY 57-1293 is not tency of BAY 57-1293 in the lethal challenge model and the
only more potent in vitro and in vivo by at least one order of efficacy regarding the reduction of the recurrence rate after
magnitude, it is especially efficacious when treatment is de- treatment of a primary infection are encouraging for the initia-
layed or the viral load is increased, features essential for treat- tion of trials in humans, but one has to be cautious in predict-
ment of herpes encephalitis and disseminated disease. ing the clinical benefit in humans from animal studies due to
Importantly, recurrent disease and asymptomatic virus shed- different parameters among species such as metabolism and
ding are nearly completely suppressed by the helicase-primase pharmacodynamics.
inhibitors, which should decrease person-to-person transmis- Nevertheless, it seems likely that BAY 57-1293 will further re-
sion. As some of the disease syndromes caused by HSV are com- duce mortality and morbidity for life-threatening HSV disease as
mon, relatively benign and self-limiting, the therapeutic index well as decrease the establishment of latency and subsequent re-
of an anti-herpes drug must be extremely high for broad appli- lapse. Clinical trials will now have to show whether the in vitro

396 NATURE MEDICINE • VOLUME 8 • NUMBER 4 • APRIL 2002


ARTICLES

activity, the efficacy in animal models and the favorable phar- 5 mM MgCl2, 0.2–5.0 mM ATP, 100 µg BSA per ml, 10% glycerol, 1 mM
macokinetic and safety profiles open the way to a novel once DTT (DL-dithiothreitol) and incubated for 60 min at 37 °C. The released in-
daily, non-nucleoside treatment of diseases caused by HSV. organic phosphate was detected colorimetrically as described16.
DNA dependent ATPase activity was calculated from the net absorbance
change in the presence and absence of inhibition.
Methods
Animals, cells and viruses. BALB/cABom female mice (weight, 19 g; age, Generation and sequencing of resistant viral mutants. Naturally occur-
7 wk) and C3H/TifBom-hr female mice (age, 7 wk) were purchased from ring, resistant viral mutants were selected in the presence of 1 µM BAY
M&B A/S (Ry, Denmark) whereas female Hartley guinea pigs were pur- 57-1293 or of at least 100 times the IC50 concentration of the compounds
chased from Charles River Laboratories (Wilmington, Massachusetts) or listed in Table 1 using the fluorescence based viral replication assay. 1 ×
Harlan & Winkelmann (Borchen, Germany). Cells (Vero (African green 104 Vero cells were seeded in 96-well MTPs as described above and incu-
© 2002 Nature Publishing Group http://medicine.nature.com

monkey) kidney cells), human embryonic lung fibroblasts, normal human bated overnight. Compound was added to the final concentration and
diploid fibroblasts and viruses were obtained from ATCC (American Type the cells were infected with 1,000 p.f.i. (m.o.i. = 0.1) per well. Mutants
Culture Collection, Manassas, Virginia) or DSMZ (Deutsche Sammlung (viral plaques) were identified with a microscope after 3–5 d incubation
von Mikroorganismen und Zellkulturen, Braunschweig, Germany) and cul- or by storing replica samples of 10 µl of each well and analyzing the
tivated according to standard procedures8. Animals were housed accord- 20–40 MTP with the fluorescence dye fluorescein diacetate as described
ing to German animal protection laws, guidelines and approval. above. If a resistant virus is present in an individual well, the fluorescence
read-out decreases at least by a factor of 3 as compared with mutant-free
Fluorescence based cytopathogenicity assay. 1 × 104 Vero cells per well of wells. Mutant-positive supernatants or stored samples were used to pro-
a microtiter plate (MTP) were infected with 25 p.f.u. of herpes simplex virus duce stocks, the titer was determined, the DNA prepared8 and sequenced
(HSV-1 F or clinical isolates of HSV-1 or 2; multiplicity of infection (m.o.i. = (Custom sequencing, Qiagen).
0.0025)) in a total volume of 200 µl media (M199 medium supplemented
with 5% FCS, 2 mM glutamine, 100 IU/ml penicillin and 100 µg/ml strepto- Murine lethal challenge model. 50 µl virus suspension (HSV-1 Walki or
mycin) in the presence or absence of drug and incubated for 5 d at 37 °C, HSV2MS) in ice-cold PBS (∼5 × 104 p.f.u.) was applied to the nares of
5% CO2. The wells of the MTP were washed with PBS (200 µl) and then filled lightly ether-anesthetized BALB/cABom female mice12,13, resulting in a
with 200 µl PBS containing 10 µg/ml fluorescein diacetate. After a 45-min mortality of 90–100% after 7–10 d. For oral treatment compounds were
incubation at room temperature, fluorescence was measured at 485 nm ex- suspended in 0.5% Tylose (MH 4000 P2, Clariant, Muttenz, Switzerland)
citation and 538 nm emission wavelengths. IC50 values were determined by in PBS via ultrasonication. The suspension was subsequently administered
a nonlinear plot of antiviral activity as a function of drug concentration5,6. via oral gavage in a total volume of 200 µl/dose twice daily at the day of
infection and 3× daily (7:00 AM, 2:00 PM and 7:00 PM) from d1 to d4 p.i.
Agarose gel electrophoresis of RT-PCR fragments prepared from virus Infected animals were inspected daily for signs of disease (encephalitis,
infected cell cultures. 5 × 106 Vero cells were seeded in tissue culture paralysis) and moribund animals were killed.
flasks and incubated over night at 37 °C, 5% CO2. The cells were infected
(m.o.i. = 1) with HSV-1 F in the presence and absence of BAY 57-1293 (2 Murine zosteriform spread model. C3H/TifBom-hr female mice were
µM). Identical results were obtained with BAY 44-5138 (25 µM). anesthetized by ether and the lateral side of the body was scratched 10
The RNA of infected cells was purified at 2 h (UL54), 12 h (UL8) and 16 h times in a crossed-hatch pattern with a 27-gauge needle. 10 µl virus sus-
(UL13) after infection (Qiagen, Hilden, Germany) RNA purification (RNeasy pension (1 × 106 p.f.u. HSV-2G) was inoculated on the scarified area and
kit; 40 µl elution) and quantified (absorption at 260 nm). The RNA (2 µg) rubbed using the pipette tip14. Mice were inspected daily and disease
was reverse transcribed with a specific primer (2 pmol; UL54, 5′ severity was determined using a scoring system: 0, no signs of infection
AAACAGGGAGTTGCAATAAA 3′; UL8, 5′ GGCAAACAGAAACGACATCT 3′; visible; 1, vesicle formation; 2, slight zoster spread; 3, large patches of
UL13, 5′ CGACAGCGCGTGCCGCGCGC 3′) into cDNA according to the zoster formed; 4, confluent zoster band; 5, hind limb paralysis; 6, death.
Superscript II protocol (Invitrogen, Karlsruhe, Germany). Aliquots (2 µl) of Moribund animals were killed.
the reverse transcription reaction were amplified by PCR. A 300-bp frag-
ment of the HSV UL54 gene, a 350-bp fragment of the UL8 gene and a 600- Guinea pig model of genital herpes. Female Hartley guinea pigs were
bp fragment of the UL13 gene were amplified in 30 cycles (UL54 and UL8: 3 infected intravaginally15 with 2.5 × 105 p.f.u. HSV-2 strain MS. Depending
min, 94 °C hot start; 1 min, 94 °C denaturation; 1 min, 55 °C annealing; on the therapy regimen examined, treatment started 6 h p.i. and contin-
1 min, 72 °C polymerization. UL13: 3 min, 94 °C hot start; 1 min, 94 °C de- ued for 4 more days (‘very early episodic therapy’), or started at d4 p.i.
naturation; 1 min, 60 °C annealing; 1 min, 72 °C polymerization.) by PCR and lasted for 10 d (‘delayed episodic therapy’) or started at d20 p.i. and
(Taq-Polymerase, Stratagene, Amsterdam Zuidoost, The Netherlands), in a continued for 20 d (‘suppression therapy’). Animals were examined daily
100-µl reaction volume with the following oligonucleotides: UL54, 5′ GCC for herpes lesions and the lesion severity was scored on a 0–4 scale.
TGT GCG GCC TGG ACG AA 3′ and 5′ AAT ATT TGC CGT GCA CGT AC 3′; During d1–d4, p.i. samples of vaginal secretion were obtained using cot-
UL8, 5′ CGCCTGCGACCGCCTTATCT 3′ and 5′ TGTCGTCAAAGGGATA- ton tipped swabs. Individual swabs were placed in tubes with 1 ml
CACA 3′; UL13, 5′ CGATCGCCCGGGGGCAGTTT 3′ and 5′ ACGGGTTGGT- Dulbecco’s MEM and processed as described15. Virus titers were deter-
GTGACACAGG 3′; 0.1 nmol each. 8-µl aliquots of cycle 20–30 (lanes mined using standard plaque test procedures8.
2–12) of the PCR were resolved on a 2% agarose gel (Invitrogen) accord-
ing to the manufacturer’s instructions. Antiviral compounds used in animal models. BAY 57-1293 was synthe-
sized at Bayer AG, Central Research, Leverkusen and micronized using a
Cloning and preparation of the helicase-primase enzyme. HSV-1 heli- conventional air-jet mill. Acyclovir was purchased as Zovirax injection flasks
case-primase heterodimer was produced in doubly infected Sf9 (Spodoptera (GlaxoSmithKline), valacyclovir as Valtrex film tablets (GlaxoSmithKline).
frugiperda) cells using recombinant baculoviruses expressing the UL5 and
the UL52-6xHis genes of the helicase-primase subunits. The genes were am- Acknowledgments
plified by PCR from HSV-1 F (American Type Culture Collection ATCC VR- We thank J. Blümel for clinical field isolates (UB1-31, UB33-47 & UB49-56) of
733) and cloned into the baculovirus expression system (UL5, pFASTBAC1; HSV-1 and HSV-2; P. Spear and L. Kwan for analyzing herpesvirus entry into
UL52, pFASTHTb) according to the Instruction Manual BAC-TO-BAC cells; P. Schaeffer for ICP-4 plasmids; S. Weller for recombinant baculoviruses;
Baculovirus (Expression Systems, Invitrogen, Karlsruhe, Germany). The het-
H.R. Hehnen for material and lab space to carry out the animal virus
erodimeric enzyme was purified by IMAC-Chromatography as described16.
experiments (equine, porcine and bovine herpes strains).; M. Becka for statisti-
ATPase Assay. Purified heterodimeric helicase-primase complex (200–400 cal analysis of data; and K. Ostertag-Palm, M. Hucke, C. Stamm, E. Clemente,
ng) was incubated with 1 µg DNA (Sigma D3287 or D8681) in 20 mM M. Werth, J. Wann, I. Hulsmann, S. Schaab, S. Vogel, S. Veldhoen, D. Ganzer,
HEPES (pH 7.6; 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid), G. Köppe, J. Hotho, J. Leske, U. Zuther, H. Schoop, O. Augustin, M. Peters, J.

NATURE MEDICINE • VOLUME 8 • NUMBER 4 • APRIL 2002 397


ARTICLES

Dornieden, H. Blum, E. Lindner, G. Heckmann, E. Carrozzo, B. Schulz, H. New Jersey, 1998).


9. Crumpacker, C.S., Schnipper, L.E., Zaia, J.A. & Levin M.J. Growth inhibition by
Küper, U. Reimann, M. Heidtmann, D. Höpker, A. Haas, A. Rudek, J. Daheim,
acycloguanosine of herpesviruses isolated from human infections. Antimicrob.
J. Verlage, B. Poschmann, M. Heine and P. Hartmann for technical assistance. Agents Chemother. 15, 642–645 (1979).
10. Collins, P. & Darby G. Laboratory studies of herpes simplex virus strains resistant
to acyclovir. Rev. Med. Virol. 1, 19–28 (1991).
RECEIVED 2 JULY 2001; ACCEPTED 13 FEBRUARY 2002 11. Hall, M.C. & Matson, S.W. Helicase motifs: the engine that powers DNA unwind-
ing. Mol. Microbiol. 34, 867–877 (1999).
1. Knipe, D.M. & Howley, P.M. Fields Virology, 4th edn. 2399–2509 (Lippincott 12. Field, H.J., Anderson, J.R. & Efstathiou, S. A quantitative study of the effects of
Williams Wilkins, Philadelphia, 2001). several different nucleoside analogues on established herpes encephalitis in mice.
2. Boehmer, P.E. & Lehmann I.R. Herpes simplex virus replication. Ann. Rev. J. Gen. Virol. 65, 707–719 (1984).
Biochem. 66, 347–384 (1997). 13. de Clercq, E. & Luczac, M. Intranasal challenge of mice with herpes simplex virus:
3. Schaeffer, H.J. et al. 9-(2-Hydroxyethoxymethyl)guanine activity against viruses an experimental model for the evaluation of the efficacy of antiviral drugs. J. Inf.
© 2002 Nature Publishing Group http://medicine.nature.com

of the herpes group. Nature 272, 583–585 (1978). Dis. 133, A226–A236 (1976).
4. Keating, M.R. Antiviral agents for non-human immunodeficiency virus infection. 14. Simmons, A. & Nash, A.A. Zosteriform spread of herpes simplex virus as a model
Mayo Clin. Proc. 74, 1266–1283 (1999). of recrudescence and its use to investigate the role of immune cells in prevention
5. Fischer, R. et al. Thiazole urea derivatives and their utilization as antiviral agents. of recurrent disease. J. Virol. 52, 816–821 (1984).
Patent application WO-00/53591-A, PCT/EP01498 EP1161423 (2000). 15. Kern, E.R. & Glasgow L. Treatment of genital herpes simplex virus infections in
6. Fischer, R. et al. Thiazole amide derivatives and their utilization as antiviral guinea pigs. in Herpesvirus, Proceedings of a Burroughs Wellcome–UCLA Symposium
agents. Patent application WO-01/47904, PCT/EP01498 EP1161423 (2001). (ed. Rapp, F.) 617–636 (Alan R. Liss Inc., New York, 1984).
7. O’Brien, J.J. & Campoli-Richards, D.M. Acyclovir—an updated review of its an- 16. Spector, F.C., Liang, L., Giordano, H., Sivaraja, M. & Peterson, M.G. Inhibition of
tiviral activity, pharmacokinetic properties and therapeutic efficacy. Drugs 37, herpes simplex virus replication by a 2-amino thiazole via interactions with the
233–309 (1989). helicase component of the UL5-UL8-UL52 complex. J. Virol. 74, 6979–6987
8. Brown, S.M. & MacLean, A.R. Herpes Simplex Virus Protocols (Humana, Totowa, (1998).

398 NATURE MEDICINE • VOLUME 8 • NUMBER 4 • APRIL 2002


NEWS & VIEWS

New anti-HSV therapeutics target the helicase–primase


complex
The only targets for clinical treatment of herpes simplex virus infections have been the viral enzymes thymidine
kinase and DNA polymerase. Now, animal experiments show the healing benefits of new antiherpes drugs that act
on the viral helicase–primase complex and appear superior to the standard treatment, acyclovir.
© 2002 Nature Publishing Group http://medicine.nature.com

H erpes simplex virus types 1 and 2 (HSV-


1 and -2) are responsible for the quiet
pandemics of oral and genital herpes that
CLYDE S. CRUMPACKER &
PRISCILLA A. SCHAFFER
phate. Acyclovir triphosphate acts as a
competitive inhibitor of HSV DNA poly-
merase and a terminator of viral DNA-
have plagued humanity for centuries. In chain elongation (Fig. 1)6. The specificity of
the United States acyclovir also reflects
alone, 22% of the the fact that it in-
population is in- a Accessory protein hibits cellular DNA
fected with HSV-2, UL42 polymerase weakly in
DNA polymerase
and 1.6 × 106 new 3'
comparison to its in-
genital herpes infec- hibition of viral DNA
Single-stranded DNA-binding
tions are predicted to Leading strand polymerase. To date,
protein ICP8
occur each year. viral TK and DNA
Newborn infants and polymerase have
immunocompro- been the only molec-
mised patients suffer 8 ular targets success-
the greatest risk of 52 fully exploited for the
5
life-threatening HSV development of clini-
infections. HSV is Helicase–primase complex cally useful anti-HSV
(UL5,8,52) 3'
also the leading cause 3'
drugs. For the
of infectious blind- Lagging strand
neonate, the patient
ness in the industrial- with herpes en-
ized world. In the b c cephalitis and the im-
absence of an effec- munocompromised
tive vaccine to pre- patient, acyclovir is a
vent primary and life-saving therapy.
recurrent HSV infec- For those with genital
tions, the antiviral herpes, acyclovir has
drug acyclovir and its greatly improved the
derivatives have been quality of life.
Renee Lucas

the only effective and TZP/U Acyclovir triphosphate Despite the avail-
safe oral therapies for ability of acyclovir
treatment of HSV in- therapy, life-threat-
Fig. 1 Mechanism of DNA synthesis inhibition by acyclovir and the new drugs (modified
fections since acy- from Boehmer and Lehman11). a, HSV DNA synthesis. The helicase–primase complex, consist- ening infections with
clovir was initially ing of viral proteins UL5, -8 and -52, unwinds HSV DNA at the replication fork and primes both HSV such as herpes
reported as the first spe- lagging-strand (wavy green lines) and leading-strand (not shown) DNA synthesis. The single- encephalitis continue
cific antiviral drug in stranded DNA binding protein, ICP8, binds to single-stranded template DNA (straight blue to be associated with
1977 (ref. 1). Now, two lines). HSV DNA polymerase and its accessory protein, UL42, promote leading- and lagging- significant morbidity
papers in this issue of strand DNA synthesis (wavy blue lines). The arrows indicate the direction of movement of the and mortality. In im-
Nature Medicine de- DNA replication proteins. b, New drugs target the helicase–primase complex. The TZP/Us munocompromised
(amino-thiazolylphenyl-containing compounds and thiazole urea derivatives; red band) en-
scribe two distinct but patients, HSV infec-
hance binding of the UL5 and UL52 subunits of the helicase–primase complex to both leading-
structurally related and lagging-strand DNA resulting in inhibition of helicase activity, primase activity and viral tion can disseminate
classes of drugs2,3 that DNA synthesis. c, Acyclovir. Acyclovir triphosphate (orange wedge) binds to HSV DNA poly- to involve liver, brain
show great promise merase, inhibiting polymerase activity and synthesis of both leading and lagging DNA strands. and lungs, and acy-
as novel anti-HSV clovir has been the
agents. The drugs tar- primary treatment
get the HSV helicase–primase complex ing that of acyclovir. regimen in this group. However, resistance
(Fig. 1), a molecular target distinct from The viral specificity of acyclovir results, to acyclovir has posed a significant prob-
the viral DNA polymerase and thymidine in part, from the fact that HSV TK initiates lem, and in HIV infected patients, 6% of
kinase (TK) targeted by acyclovir and its the activation of the drug. TK monophos- HSV isolates are resistant to acyclovir. The
derivatives4,5 (Fig. 1). In animal models of phorylates acyclovir and cellular enzymes only treatment alternative for acyclovir- re-
HSV infection, the new drugs appear to subsequently add two additional phos- sistant HSV has been the pyrophosphate
have therapeutic value rivaling or exceed- phate groups to yield acyclovir triphos- analog, phosphonoformic acid (Foscarnet),

NATURE MEDICINE • VOLUME 8 • NUMBER 4 • APRIL 2002 327


NEWS & VIEWS

a reversible inhibitor of HSV DNA poly- the helicase–primase inhibitors were gen- the latent state. Consequently, continua-
merase which is more toxic than acyclovir erally superior. The claim by Kleymann et tion of the HSV pandemic is anticipated
and must be administered intravenously7. al. that the thiazole urea derivative BAY and efforts to develop vaccines and novel
Despite its unrivaled safety record and effi- 57-1293 reduces the severity and fre- therapies aimed at eliminating latent
cacy in the treatment of primary and recur- quency of recurrences is intriguing but virus are still warranted.
rent HSV disease, acyclovir has limited oral sparse data in support of this conclusion
1. Elion, G.B. et al. Selectivity of action of an antiher-
bioavailability, must be given early in the will necessitate additional experimenta- pectic agent 9-(hydroxyethoxymethyl) guanine.
disease to have a significant effect and the tion. The finding that the amino-thia- Proc. Natl. Acad. Sci. USA 74, 5716–5720 (1977).
2. Crute, J.J. et al. Herpes simplex virus helicase–pri-
active form of the drug, acyclovir triphos- zolylphenyl-containing drug B1LS-179-BS mase inhibitors are active in animal models of
phate, possesses a short intracellular half- was effective even when treatment was human disease. Nature Med. 8, 386–391 (2002).
© 2002 Nature Publishing Group http://medicine.nature.com

life. For these reasons, new therapies are delayed by 65 hours may represent an im- 3. Kleymann, G. et al. New helicase–primase in-
hibitors as drug candidates for the treatment of
clearly needed. portant advance over nucleoside analogs, herpes simplex disease. Nature Med. 8, 392–398
The two new classes of drugs, amino- which require early application for suc- (2002).
thiazolylphenyl-containing drugs2 and cess. The evidence that resistance to heli- 4. Coen, D. & Schaffer, P. Two distinct loci confer re-
sistance to acycloguanosine in herpes simplex virus
thiazole urea derivatives3, have a different case–primase inhibitors occurs less type 1. Proc. Natl. Acad. Sci. USA 77, 2265–2269
target in the HSV DNA replication frequently than resistance to acyclovir is (1980).
5. Schnipper, L.E. & Crumpacker, C.S. Resistance of
process, the helicase–primase complex promising, but must be confirmed in the herpes simplex virus to acycloguanosine: The role
(Fig. 1). This complex consists of three clinic. Both classes of drugs inhibit HSV-1 of viral thymidine kinase and DNA polymerase loci.
viral proteins, the products of the HSV and -2 (but not varicella zoster virus or cy- Proc. Natl. Acad. Sci. USA 77, 2270–2273 (1980).
6. Furman, P.A. et al. Inhibition of herpes simplex
UL5, -8, and -52 genes, which act together tomegalovirus), both are effective against virus induced DNA polymerase activity and viral
to unwind double-stranded viral DNA acyclovir-resistant mutants and both ap- DNA replication by 9-(2-hydroxyehoxymethyl)
and generate primers for DNA synthesis pear to be well tolerated in animals. guanine and its triphosphate. J. Virol. 32, 72–77
(1979).
by the viral DNA polymerase8,9. Mutations Carefully controlled clinical trials in 7. Safrin, S. et al. Treatment of acyclovir-resistant mu-
in virus isolates that are resistant to the humans are clearly the next step for both cocutaneous herpes simplex infection in patients
with the acquired immunodeficiency syndrome. A
new drugs map to the UL5 and UL52 classes of compounds. It will be interest- randomized multicenter study of foscarnet vs. vi-
genes, demonstrating that both drugs tar- ing to see how these new drugs compare darabrine. New Engl. J. Med. 325, 551–555 (1991)
get the helicase–primase complex. At the with acyclovir, valacyclovir and famcy- 8. Crute, J.J. & Lehman, I.R. Herpes simplex virus heli-
case primase. Physical and catalytic properties. J.
molecular level, the amino-thia- clovir with regard to efficacy and reduc- Biol. Chem. 266, 4484–4488 (1991).
zolylphenyl-containing drugs appear to tion of recurrences. The remarkable lack 9. Zhu, L.A. & Weller, S.K. The UL5 gene of herpes
act by enhancing the affinity of the heli- of toxicity and side effects observed with simplex virus type 1: Isolation of a lac Z insertion
mutant and association of the UL5 gene product
case–primase complex for DNA, which in- acyclovir in humans poses a strong chal- with other members of the helicase-primase com-
hibits the enzymatic activities of the lenge for any new class of antiherpes plex. J. Virol. 66, 1458–1468 (1992).
10. Spector, F.C., Liang, L., Giordano, H., Sivaraja, M.
complex2 (Fig. 1). The anti-HSV activity of drugs. Such safety features are essential if & Peterson, M.G. Inhibition of herpes simplex virus
an amino-thiazole compound has been anti-HSV therapy is to be carried out for replication by a 2-amino-thiazole via interactions
reported previously, but the compound extended periods in healthy patients. with the helicase component of the UL5-UL8-UL52
complex. J. Virol. 72, 6979–6987 (1998).
described was less potent in vitro and no The clinical implications for treatment 11. Boehmer, P.E. & Lehman, I.R. Herpes simplex virus
data concerning efficacy in animals or with these new classes of orally available DNA replication. Annu. Rev. Biochem. 66, 347–384
comparisons with acyclovir were pre- drugs are enormous. If helicase–primase (1997).

sented10. drugs inhibit HSV infections more effec-


Both newly described drugs are active tively than nucleoside analogs and ex- Departments of Medicine and Microbiology and
orally and both exhibit impressive effects hibit safety parameters comparable with Molecular Genetics
on the healing of HSV-1 and HSV-2 in cu- acyclovir, they would represent a major Harvard Medical School and
taneously infected mice, and HSV-2 in advance in controlling HSV infections. Division of Infectious Diseases
vaginally infected guinea pigs. When Despite the potential promise of these Beth Israel Deaconess Medical Center
compared with acyclovir and valacyclovir new drugs, however, neither they nor the Boston, Massachusetts
in the same experiments, the results with nucleoside analogs can eliminate virus in Email: pschaffe@caregroup.harvard.edu

Polygenic or Pollyanna?
A study suggesting that many genes, each conferring a small risk, account for most cases of breast cancer could have
important implications for population-based prevention programs—provided that the genes can be identified.

Nature Genetics by Pharoah et al.1 suggests


A bout 3% of all primary breast cancer
is due to mutations in BRCA1 or
BRCA2, which confer a high risk of de-
WILLIAM D. FOULKES that there is no BRCA3 as such. Instead,
many susceptibility alleles, each causing
veloping the disease. However, most reasonable expectation in the research a small (perhaps very small) augmenta-
analyses of large data sets suggest that community that at least one more highly tion of a woman’s breast-cancer risk,
other genes must contribute to the famil- penetrant susceptibility gene would be would be responsible for familial cluster-
ial clustering of breast cancer. Following identified, but no such gene has been ing of breast cancer that essentially mim-
the discovery of BRCA1 and BRCA2 (in found despite intensive searching by sev- ics the presence of a single allele when a
1994 and 1995, respectively), there was a eral groups. An article in the May issue of critical number of them segregate to-

328 NATURE MEDICINE • VOLUME 8 • NUMBER 4 • APRIL 2002

You might also like