Recent strategies in the development of new human cytomegalovirus inhibitors

18
Recent Strategies in the Development of New Human Cytomegalovirus Inhibitors Ana Martinez, Ana Castro, Carmen Gil, Concepcion Perez Instituto de Quı ´mica Me ´dica (CSIC), Juan de la Cierva 3, 28006 Madrid, Spain ! Abstract: Human cytomegalovirus (HCMV) is one of the most common opportunistic infections in immunucompromised individuals, such as AIDS patients and organ transplant recipients, and is the most frequent congenital viral infection in humans. Despite a reduction of the incidence of AIDS-related opportunistic infections in patients under highly active antiretroviral treatment, attention should be paid to the HCMV risk factor in these individuals. Furthermore, HCMV may have an important role in atherosclerosis. Existing antiviral treatments for the HCMV infection suffer from poor bioavailability, toxicity, and limited effectiveness, mainly due to the development of drug resistance. Fortunately there are novel and potentially very effective new compounds undergoing pre-clinical and clinical evaluation. This review provides an overview in the last five years of new HCMV inhibitors (chemical structures, SAR and new mechanisms of action) with the aim to provide new clues for the development of the future drugs against this opportunistic virus. ß 2001 John Wiley & Sons, Inc. Med Res Rev, 21, No. 3, 227–244, 2001 Key words: HCMV inhibitors; HCMV protease; opportunistic infection; immunocompromised patients 1. INTRODUCTION Human cytomegalovirus (HCMV) is a highly species-specific DNA virus member of the beta- herpesvirinae, a subfamily of the herpesviridae. 1 Like other herpesviruses, primary infection with HCMV is followed by persistence of the virus in a latent form. The sites of latency are still largely undefined, 2 but they probably include bone marrow progenitor cells and peripheral blood monocytes. From these sites, the virus can reactivate, 3 resulting in renewed shedding of the virus, or, in immunocompromised persons, development of disease. HCMV-infected cells may become enlarged (cytomegalia) showing intranuclear inclusions, and co-survival of cells and virus is often established. Except for a mononucleosis-like illness in some persons, infection with HCMV rarely 227 Correspondence to: Dr. Ana Martinez, Instituto de Qui ¤ mica Me¤ dica (CSIC), Juan de la Cierva 3, 28006 Madrid, Spain; E-mail: [email protected] Contract grant sponsor: Fondo de Investigaciones Sanitarias; Contract grant number: FIS 98/253; Contract grant sponsor: Comuni- dad de Madrid; Contract grant number: 82/36.1/1999. Medical Research Reviews,Vol. 21, No. 3, 227^244, 2001 ß 2001 John Wiley & Sons, Inc.

Transcript of Recent strategies in the development of new human cytomegalovirus inhibitors

Recent Strategies in the Developmentof NewHuman Cytomegalovirus

Inhibitors

Ana Martinez, Ana Castro, Carmen Gil, Concepcion Perez

Instituto de QuõÂmica MeÂdica (CSIC), Juan de la Cierva 3, 28006 Madrid, Spain

!

Abstract: Human cytomegalovirus (HCMV) is one of the most common opportunistic infections

in immunucompromised individuals, such as AIDS patients and organ transplant recipients, and is

the most frequent congenital viral infection in humans. Despite a reduction of the incidence of

AIDS-related opportunistic infections in patients under highly active antiretroviral treatment,

attention should be paid to the HCMV risk factor in these individuals. Furthermore, HCMV may

have an important role in atherosclerosis. Existing antiviral treatments for the HCMV infection

suffer from poor bioavailability, toxicity, and limited effectiveness, mainly due to the development

of drug resistance. Fortunately there are novel and potentially very effective new compounds

undergoing pre-clinical and clinical evaluation. This review provides an overview in the last ®ve

years of new HCMV inhibitors (chemical structures, SAR and new mechanisms of action) with

the aim to provide new clues for the development of the future drugs against this opportunistic

virus. ß 2001 John Wiley & Sons, Inc. Med Res Rev, 21, No. 3, 227±244, 2001

Key words: HCMV inhibitors; HCMV protease; opportunistic infection; immunocompromised

patients

1 . I N T R O D U C T I O N

Human cytomegalovirus (HCMV) is a highly species-speci®c DNA virus member of the beta-

herpesvirinae, a subfamily of the herpesviridae.1 Like other herpesviruses, primary infection with

HCMV is followed by persistence of the virus in a latent form. The sites of latency are still largely

unde®ned,2 but they probably include bone marrow progenitor cells and peripheral blood

monocytes. From these sites, the virus can reactivate,3 resulting in renewed shedding of the virus, or,

in immunocompromised persons, development of disease. HCMV-infected cells may become

enlarged (cytomegalia) showing intranuclear inclusions, and co-survival of cells and virus is often

established. Except for a mononucleosis-like illness in some persons, infection with HCMV rarely

227

Correspondence to: Dr. AnaMartinez, Instituto deQuimicaMedica (CSIC), Juande la Cierva 3, 28006Madrid,Spain;E-mail: [email protected] sponsor:Fondode Investigaciones Sanitarias; Contractgrant number:FIS 98/253; Contractgrant sponsor: Comuni-daddeMadrid; Contractgrant number: 82/36.1/1999.

Medical Research Reviews,Vol. 21,No. 3, 227^244, 2001ß 2001 John Wiley & Sons, Inc.

causes disease in inmunocompetent individuals.4 However, reactivation of virus is of signi®cant

concern in the immunocompromised individual,5,6 particularly when the de®ciency affects cell-

mediated immune responses such as in HIV-infected patients. Immunosupressed individuals,

including solid organ and allogeneic bone marrow recipients,7 are also at great risk of developing

HCMV disease. This pathogen is the most common congenital viral infection in humans due to the

high prevalence of the virus in the general population, with up to 90% of the urban population

infected.8 Although it is well known that highly active antiretroviral therapy has decreased

substantially the incidence of HCMV retinitis in AIDS patients,9 this change does not indicate that

HCMV in AIDS patients has abated and is no longer a concern.10,11 On the contrary, HCMV disease

in solid organ recipients, newborns, and even in AIDS patients still accounts for considerable

morbidity, and drug resistance to the anti-HCMV compounds offers a major concern.12±14 Further-

more, HCMV may have an important role in the development of metabolic diseases, such as

atherosclerosis.15,16

Antiviral agents currently licensed for the treatment of HCMV infections include ganciclovir,17

foscarnet,18 cidofovir,19 and fomivirsen.20 All of these drugs, either directly or indirectly inhibit

viral polymerase and are able to reduce viral proliferation in patients who develop the clinical

symptoms associated with HCMV disease. However, such therapy cannot clear the virus comple-

tely. Additionally, these agents exhibit toxicity21 and require intravenous administration to obtain

therapeutic drug levels, both of which obviate their use for long-term treatment. Incomplete

suppression of viral replication during chronic antiviral treatment predisposes to the development of

viral drug resistance and consequent drug failure. Indeed, the emergence of CMV strains with

reduced susceptibility to ganciclovir, foscarnet, cidofovir and/or fomivirsen during long-term

therapy has been documented.22±24

There is renewed interest in the search for new HCMV inhibitors because of the growing

population of inmunocompromised individuals. This is mainly due to two facts: ®rstly, improve-

ments on transplantation techniques have increased the number of people living with a transplanted

organ; and secondly, the advent of new retroviral therapies has converted AIDS into a chronic

immunocompromised disease. Additionally, advances in gene therapy25,26 have stimulated the

search for new anti-HCMV inhibitors. Fortunately there are novel and potentially very effective new

compounds27,28 undergoing pre-clinical and clinical evaluation. These developments point the way

toward new therapies and also to clearer understanding of the biology of HCMV replication,

infection, and disease. This review provides an overview in the last 5 years of new HCMV inhibitors

(chemical structures and new mechanisms of action) with the aim to provide new clues for the

development of the future drugs against this opportunistic viral infection.

A. New Prodrugs of Existing Clinical Agents

Much effort has been made to overcome the poor pharmacokinetics of the approved drugs for the

treatment of HCMV infections. The bioavailability of orally administered ganciclovir in humans

ranged from 2.6 to 7.3%,29 although giving the drug with food increases absorption. Thus, the

search for a prodrug that is orally well-absorbed and then readily converted to ganciclovir (1) is of

high priority. It has been recently reported that the mono-3-(2 0-acetoxy-4 0,6 0-dimethylphenyl)-3,

3-dimethylpropanoic ester of ganciclovir (2), showed a 4-fold increase in oral bioavailability over

the parent drug in rats.30 It has been reported that 6-¯uoropurine acyclonucleosides are converted to

ganciclovir in the presence of calf intestinal mucosal adenosine deaminase.31 Recently, the

monoester (3) has shown a 15-fold higher oral gancicolovir bioavailability in rats.32

The serious irreversible nephrotoxic effects associated with cidofovir (4) treatment has been

improved in the recently described tetrahydrofuran diethyl phosphonate analogue (5). It has a

potency comparable to that of cidofovir against HCMV and a greater safety index in vivo.33 Prodrug

approaches have also been investigated to circumvent problems associated with the bioavailability

228 * MARTINEZETAL.

and permeability of phosphonoformate (PFA, foscarnet (6)). Synthetic methods have been developed

in order to incorporate the PFA moiety within biomimetics of nucleotides, carbohydrates,

and phospholipids.34 Cyclic PFA esters (7±9) are much more reactive towards hydrolysis than

their acyclic analogs. This approach holds promise when combined with a strategy for activating

the prodrug to subsequent de-esteri®cation at phosphorous. Some other pronucleotide design

approaches have been recently reviewed35 offering important tools for the in vivo delivery of

antiviral nucleotides.

B. Nucleoside and Nucleotide Derivatives as HCMV Inhibitors

Nucleoside and nucleotide analogs are the focus of current interest as antiviral and antitumor agents.

A great number of HCMV inhibitors are concentrated in this group of compounds. In general, the

new nucleotide/nucleoside analogs described are derived from the natural purine or pyrimidine

heterocycle nucleus, although derivatives of synthetic halo-benzimidazoles seem to be very

promising inhibitors.

1. Purine and Pyrimidine Nucleoside Analogs

Modi®cations of some ribofuranosyl derivatives of purine and pyrimidine related heterocycles such

as imidazoisothiazole36 (10), pyrrolopyridazinone37 (11) or pyranoic acid derivatives38 (12), have

Figure 1. Newprodrugs ofclinical agents.

NEWHUMANCYTOMEGALOVIRUS INHIBITORS * 229

been performed in order to increase anti-HCMV activity. However, the antiviral potency of these

compounds was tempered by their toxicity to uninfected cells (Fig. 2).

Anti-HCMV activity has been found for a series of carbohydrate-modi®ed nucleoside analogs

such as those comprising methylenecyclopropane function39 (13±14) or cyclohexene rings40 (15±16)

(Fig. 3). In both cases, the two enantiomeric nucleosides exhibited similar activity against several

herpesviruses. As the viral thymidine kinase (TK) plays an important role in their biological activity

(through phosphorylation of 5 0-hydroxyl mimic), these data con®rm previous observations that TK

could accommodate a con®gurational changes in the nucleoside upon binding, resulting in phos-

phoration of both pairs of enantiomers.

Natural endogenous nucleosides must be phosphorylated to the corresponding 5 0-triphosphated

to be incorporated into the DNA strand being synthesized in the cell. The ®rst phosphorylation step

leading to the formation of nucleoside 5 0-monophosphate, is commonly catalyzed by a nucleoside

kinase, such as TK, encoded by the host cell or the virus infecting the host cell. Inactive nucleoside

analogs can be activated by conversion to lipophilic pronucleotides, thus bypassing the ®rst

phosphorylation step in their activation mechanism. These pronucleotides are usually phosphate-

esther prodrugs of nucleoside 5 0-monophosphates. The phenyl phosphoranilate 17 increased over

50-fold its activity against HCMV compared to the parent spironucleoside 18, whereas the cyto-

toxicity remained low (Fig. 3).41 These results con®rm the hypothesis that phosphorylation is

indispensable for the activity of this new kind of nucleoside.

The area of oligonucleotide chemical synthesis has recently opened up a source of antiviral

agents.42 The working hypothesis developed during these studies was that the ability to form an

ordered, non-hydrogen-bonded array in solution was an important criterion for activity.43 The poly-

and oligonucleotides containing triazolo[2,3-a]purines (19±20) (Fig. 3) have been shown to be more

potent inhibitors of HCMV than ganciclovir.44 Their mechanism of action is currently under study.

More interesting are the inhibitors that exert their activity against HCMV by a novel mechanism

of action. This is the case of the novel pyrimidine and purine derivatives of L-ascorbic acid (Fig. 3).

Compound 21 exhibited a potent (yet speci®c) antiviral effect against TK� and TKÿ varicella zoster

virus and HCMV.45 The mechanism of action of these new anti-HCMV inhibitors, although different

from DNA-polymerase inhibition, actually remains unknown.

Analogs of the pyrrolo[2,3-d]pyrimidine nucleosides tyocamycin (22), sangivamycin (23), and

thiosangivamycin (24) have been shown to be potent inhibitors of HCMV (Fig. 3). The parent

nucleosides possess signi®cant activity against HCMV but are highly toxic to uninfected cells.46 In

an effort to decrease the cytotoxic effects and obtain more selective antiviral agents, a diverse class

of 7-substituted 4-aminopyrrolo[2,3-d]pyrimidines were prepared,47 including non-nucleoside

analogs as acyclic48 (25) or benzylic derivatives49 (26). Structure-activity data established that the

N-7 substituent in this family of compounds is important to modulate the toxicity against the

uninfected cells, while the 5-thioamide moiety is crucial in conferring antiviral activity against

HCMV. Phosphorylation of a side chain hydroxyl group is not required for antiviral activity, thus

indicating a different mechanism of action than current nucleoside clinical drugs. Biological studies

Figure 2. Cytotoxic HCMVribonucleoside inhibitors.

230 * MARTINEZETAL.

established that the pyrrolopyrimidines are active earlier in the replication cycle than ganciclovir,

which targets viral DNA polymerase, so the target of this new kind of HCMV inhibitor may be an

immediate-early protein.50

2. Halo-benzimidazoles

Riboside analogs derived from 2-substituted 5,6-dichlorobenzimidazole have proven to be potent

and selective inhibitors of the replication of HCMV at noncytotoxic concentrations. 2,5,6-trichloro-

1-(b-D/-ribofuranosyl)benzimidazole (TCRB) (27) and 2-bromo-5,6-dichloro-1-(b-D-ribofuranosyl)

benzimidazole (BDCRB) (28) were the ®rst lead compounds discovered (Fig. 4).51 Although these

compounds have a 5 0-OH in the riboside moiety that could be triphosphorylated to provide

inhibitors of DNA polymerase, biological evaluations of TCRB and BDCRB have established that

their antiviral action involves neither phosphorylation of the compound nor inhibition of DNA, RNA,

Figure 3. Carbocyclic, oligonucleotide, andnucleosideanalogswith HCMV inhibitoryactivity.

NEWHUMANCYTOMEGALOVIRUS INHIBITORS * 231

or protein synthesis. Rather they act by a unique mechanism which involves the inhibition of viral

DNA processing and virus assembly.52 Further studies revealed that certain benzimidazoles

ribonucleosides are less toxic to hemopoietic progenitors than ganciclovir53 which could

circumvent one of the toxic effects of this clinical drug in prolonged maintenance therapy.

Chemical modi®cations in the 5 0-position of these benzimidazole derivatives (29, 30) indicate

that the HCMV receptor for the TCRB tolerates signi®cant modi®cation in bulk and electro-

negativity in this region without adversely affecting antiviral activity,54 including the lack of the

5 0-hydroxymethyl group exempli®ed by the potent erythrofuranosyl compound (31).55 In addition

to these D-carbohydrate derivatives, several acyclic56 (32), L-carbohydrate57,58 (32, 34) and benzyl59

(35) derivatives have been synthesized and evaluated (Fig. 4). From these data, the carbohydrate

moiety (ribo-, lyxo-, arabino-, D- or L-) is necessary for anti-HCMV activity as the acyclic or

benzylic derivatives are completely devoid of anti-HCMV activity. The addition of ribose to a

polyhalogenated benzimidazole reduces toxicity and retains or increases activity against HCMV.60

However, pharmacokinetics studies of TCRB in rat and monkeys revealed that the nucleosides

are metabolized to a certain extent in vivo by glycosidic bond cleavage. The instability of 27 and 28is detrimental to their use as antiviral agents because the half-lives of the parent compounds are less

than 1 h. Several structural modi®cations have been explored to improve pharmacokinetic

properties. Some approaches, like the synthesis of C-nucleoside61 (36) or ¯uorosugar analogs62 (37)

failed, as they do not provide more potent compounds. However, carbocyclic benzimidazoles (38)

resulted in very stable, potent, and interesting inhibitors.63

Concerning the base-like heterocycle moiety, several changes have been made including

3-deaza analogs (39).64 The order for activity against HCMV among the 5,6-dihaloribonucleosides

was I�Br�Cl>>F>H compared to I>Br>Cl>F, H for cytotoxicity, establishing that the size and

electronegativity of substituents in the 5- and 6-positions are critical for activity against HCMV and

for toxicity to uninfected cells.65 Structure-activity relationships have revealed the importance of

substitution at the 2-position.66 From among these studies, 5,6-dichloro-2-isopropylamino-1-

(b-L-ribofuranosyl)benzimidazole (1263W94) (40) was chosen for clinical trials due to its potent

and selective activity in vitro and its superior pharmacokinetic properties.67 This L-ribofuranosyl

derivative inhibits HCMV DNA synthesis by a mechanism that does not require phosphorylation

and does not involve DNA polymerase.68 Compound 1263W94 is in clinical development by Glaxo

Wellcome for the treatment of HCMV infections.69

Genotypic characterization of HCMV resistant to TCRB and BDCRB has identi®ed two viral

genes, UL56 and UL89, which mutate to give drug-resistant virus.70 Thus, the proteins encoded by

these two genes are likely the target(s) TCRB and related analogs. However, little is known

regarding their overall three-dimensional structure nor of benzimidazole binding pocket(s) on

the proteins. To probe the spatial limitation of the target enzyme, a serie of tricyclic nucleosides71

(41±44) have been synthetized leading to active but very cytotoxic compounds. A three-dimen-

sional QSAR study will be desirable in the future to speci®cally delineate structure-activity

relationships.

C. Nonnucleoside HCMV Inhibitors

The clear medical need for HCMV drugs with a more desirable pro®le and/or new mechanism of

action has initiated different screening programs to discover new leads. Broad screening of the

Pharmacia and Upjohn chemical library led to identi®cation of the acyloxyamidine 45 as a

competitive inhibitor of the HCMV DNA polymerase.72 Preliminary SAR investigation charac-

terized this class by two aryl groups connected by an acyloxyamidine linker. Initial studies

in which the two aryl groups were varied revealed little tolerance for modi®cation of the left hand

2,4-dichlorophenyl group, but replacing the right hand benzothiazole ring with a disubstituted

isoxazole ring as in 46 (Fig. 5) led to a 30-fold enhancement of activity.

232 * MARTINEZETAL.

Targeting alternative viral protein functions led to the discovery of the simple, unsubstituted

indolocarbazole Arcyria¯avin A 47 (Fig. 5) as a potent, selective inhibitor of HCMV replication in

vitro.73 A series of novel symmetrical analogs have been prepared to investigate SAR, affording a

number of very active compounds, such as the 2,10-di¯uoro derivative 48, which is approximately

100-times more potent than ganciclovir in vitro. The study of their mechanism of action is under

study. The anti-HCMVactivity does not appear to be related to the inhibition of protein kinase C, the

pharmacophore of compounds 47 and 48 are related to bis-indolylphtalimides, such as

staurosporine, which are known to be potent protein kinase C (PKC) inhibitors. The anti-HCMV

mode of action of indolocarbazoles could be inhibition of UL97,74 an alternative viral kinase, or

inhibition of an induced or elevated host kinase.

The identi®cation of 1,6-naphthyridine 49 (Fig. 5) as a potent HCMV inhibitor is the result of

the screening program of BioChem Pharm. SAR investigation on this class of compounds has

de®ned the structural requirements for potency and selectivity.75 On the benzylamine side of the

molecule, a number of modi®cations can be tolerated, however, the 2 0-alkoxy benzylamines seem to

be the most promising in terms of potency and selectivity. Substitution on the C-8 of the

napththyridine ring enhances potency whereas introduction of a methyl at C-4 did not result in any

improvement. The isoquinoline 50, a deaza derivative of the lead, showed also interesting anti-

HCMV activity.76 However, during preliminary pharmacological evaluation of 49 and 50, it was

found that these compounds were not stable in mouse or monkey S9 liver preparations, indicating

that they are likely to be subjected to ®rst pass of metabolism. Additionally, in vivo studies in mice

Figure 4. Benzimidazolenucleosidesactiveagainst HCMV.

NEWHUMANCYTOMEGALOVIRUS INHIBITORS * 233

con®rmed that no parent compounds were detected 20 min after 49 or 50 were given orally. In an

effort to circumvent these metabolism issues, the tetrahydroisoquinoline 51 provided a meta-

bolically stable and orally bioavailable alternative to the 1,6 naphthyridines and isoquinolines.77

The favourable pharmacokinetic pro®le of this class of compounds will allow their future evaluation

as potential oral-anti HCMV agents with a new mechanism of action implicated on events at the

early and late stage of virus replication.78

Some imidazo[1,2-a]pyridine and pyrimidine derivatives (52, 53) proved quite promising anti-

HCMV and anti-VZV agents.79 SAR studies de®ned the nature of the thioether side chain which

must contain an aromatic cyclic ring.80 The fact that they show selective activity against these two

viruses suggests that they are preferentially metabolized by the virus infected cells and/or show

particular af®nity for virus-speci®c enzymes.

Recently the benzothiadiazine dioxide (BTD) modi®ed acyclonucleosides (54), has been

reported to exhibit marked activity against HCMV and varicella-zoster virus (VZV).81 The structure

of this compound is quite unique, not only with respect of the nature of the heterocyclic base, but

also because of the lack of the 5 0-OH mimetic group present in GCV and other current anti-CMV

drugs, which points to a different mechanism of action.82 Initial structure±activity data showed the

necessity of a double substitution in the heterocycle, while monosubstituted compounds were

completely devoid of antiviral activity.83 Simultaneously, lipophilicity in the acyclic side chain is

essential to preserve the anti-HCMV activity. These two factors were considered when preparing

second-generation analogs of BTD. The chlorophenylmethyl BTD derivatives (55, 56) proved to be

potent non-nucleoside HCMV inhibitors.84 In order to obtain further insights into the structural

requirements for the biological activity of BTD as new anti-HCMV drugs, a CoMFA model was

developed which identi®ed the most signi®cant steric and electrostatic interactions involved in anti-

HCMV activity. The model suggested that the steric component is a predominant factor in the

antiviral activity of these analogs with electrostatic factors playing a smaller yet signi®cant role.85

Figure 5. NonnucleosideHCMV inhibitors.

234 * MARTINEZETAL.

The BTD compounds do not inhibit the HCMV protease. Time-of-addition experiments performed,

revealed that the anti-HCMVaction may be targeted at an early stage of the viral replicative cycle.86

Further experiments are in progress to elucidate the mechanism of inhibition and molecular target of

action of these novel compounds.

D. HCMV Protease Inhibitors

The cytomegalovirus protease (or assemblin) is essential for proper assembly of viral capsids and, as

a consequence, viral replication.87 A great deal of attention is being paid to these enzymes as

therapeutic targets for the development of novel antiherpetic agents,88,89 specially for the highly

prevalent HCMV as the existing antiviral treatments suffer from toxicity and limited effectiveness.

All herpesvirus proteases are synthesized as a fusion with capsid assembly protein processing

two speci®c proteolytic cleavages sites.90 One of these takes place at the maturation site (M-site)

near the C-terminus of the assembly protein while the others occurs at the realease site (R-site)

separating the amino-terminal protease from the assembly protein domain. More recently, yet a third

HCMV protease autoprocessing site has been identi®ed.91 This cleavage site, called the internal-site

(I-site) is limited to simian- and human-cytomegalovirus proteases and is located within the protease

domain.

The quest for new inhibitors of HCMV protease92 has been paralleled by much activity

focusing on the structural biology of the target,93,94 which has elucidated the highly novel structure

of this enzime.95 X-ray crystallographic studies96 have revealed HCMV protease to possess a

protein fold previously unseen among serine proteases as well as a unique catalytic triad (Ser, His,

His).97 The crystal structure revealed that it is an homodimeric enzyme with two well-separated

active sites on opposite faces of the dimer.98 This biological and structural knowledge99 has been

used to rationally design both mechanism- and nonmechanism-based inhibitors of HCMV

protease.100

Development of peptidomimetic inhibitors of HCMV protease based on amino acid sequences

of the major cleavages sites of the protease has been one of the strategies employed for the design of

new inhibitors resulting in derivatives 57, 58 (Fig. 6), which showed sub-micromolar enzymatic

inhibitory activity.101 Further investigation on these compounds concluded that the HCMV protease

is an induced-®t enzyme, undergoing a conformational change upon complexation of electrophilic

ketone peptidic inhibitors such as 57.102, 103 Nevertheless cell culture assay of some members of this

series showed EC50 values in the 100 mM range probably due to the peptidic nature of the inhibitors

and poor cell penetrability. This poor activity has prompted up the design of new mechanism-based

derivatives incorporating a b-lactam ring.104 The peptidic series of monobactam inhibitors of

HCMV protease, illustrated with derivative 59, exhibited good in vitro potency (IC50 values against

HCMV protease N0 were between 0.01 and 1 mM) and were selective when tested for inhibitory

activity using a number of serine and cysteine proteases.105 Further NMR and mass spectrometry

studies demonstrated that monocyclic b-lactam derivatives inhibit HCMV protease by acting as

alternatedcomprised substrate inhibitors through the formation of an acyl enzyme intermediate.106

Moreover, some of the compounds in a smaller series bearing an aromatic ring linked by a

methlylene group at C-4 compounds 60, 61, exhibited activity in a viral replication assay which

suggested that these compounds were cell penetrable and could potentially be useful antiviral

agents.107,108

Directed mechanism-based screening, targeted to compounds that could afford a stable acyl-

enzyme adduct of the active site serine, identi®ed a novel class of serine protease inhibitors, the

spirocyclopropyl oxazolones represented by 62±64 as submicromolar inhibitors of HCMV

protease,109 and the known serine protease inhibitor class of 2-substituted benzoxazinones.110,111

A series of 6-substituted 2-aminobenzoxazinone analogs (65±67) has been prepared that show

potent inhibitory activity toward the target enzyme (IC50� 0.46±4 mM) and demonstrate antiviral

NEWHUMANCYTOMEGALOVIRUS INHIBITORS * 235

Figure 6. Inhibitors of HCMV protease.

236 * MARTINEZETAL.

activity in cell culture (IC50� 23 mM). Modi®cations of the substituent at the 6-position modulates

both enzyme selectivity and plasma stability without adversely affecting potency toward the viral

protease. Further chemical structure optimization led to the thiophene oxazinones (68, 69), depicted

in Figure 6, which are potent and selective mechanism based inhibitors of the HCMV protease with

enhanced stability relative to benzoxazinones.112 A parallel synthesis approach was utilized to

prepare a range of N-acyl analogs113 (70, 71) and further studies revealed that these potent and

selective inhibitors of HCMV act by not only acylating the catalytic serine 132 (opening of the

oxazine ring of the inhibitor) but also by alkylating cysteine 161 of the protease (Michael addition to

an enedione side chain).114 Incorporation of moieties with H-bonding potential in an attempt to

increase potency vs. the enzyme, led to the thienoxazinone derived nitroso and hydroxylamine

compounds (72, 73) which are some of the most potent, non-peptide inhibitors of HCMV protease

prepared to date.115 The inhibition of the enzyme is through 1:1 adducts at Ser 132, although the

reason for the increased potency of these last compounds has not been de®nitively established.

A high-throughput screen to identify inhibitors of HCMV protease activity was devel-

oped116,117 to enable a variety of samples types, including natural products, to be screened. During

the search of potential new anti-viral drugs a series of tripepsides, cytonic acids A (74) and B (75),

was isolated from solid-state fermentation of the fungus Cytonamea sp,118 while the naphtha-

ceneaquinone glycosides, quanolirones I (76) and II (77), were isolated from an ethyl acetate extract

of Streptomyces sp WC76535.119 Assemblin protease contains several surface accessible cysteine

sulphhydryl groups. Moreover, this protease must be in a reduced state to maintain its catalytic

activity. Thus, many reported inhibitors which contain polyphenolic groups (e.g., 74, 75) or quinone

moieties (compounds 76, 77) may inhibit assemblin via an oxidative mechanism. Additionally, the

previous HTS identi®ed benzothiopyran-S,S-dioxides (78, 79) and 1,4-naphthoquinones (80, 81) as

potent inhibitors of HCMV protease. Studies of benzothiopyrane compounds supports a reversible

mode of inhibition that does not involve oxidative inactivation of the enzyme,120 while the naphtho-

quinone derivatives 80 and 81 are irreversible protease inhibitors which covalently modify Cys

202.121 Certain examples retain activity in viral plaque assays, but have limited chemical stability

toward biologically relevant nucleophiles.

E. Vaccines

The most important rationale for development of a HCMV vaccine is prevention of congenital

HCMV disease by preventing primary maternal infection. This is complicated by the fact that

maternal infection usually passes unnoticed.

The presence of maternal antibodies before conception clearly is effective in reducing the rate

of congenital infection and preventing severe disease. It has been estimated that vaccination of

HCMV seronegative mothers would prevent 80±85% of congenital HCMV disease. Additionally,

effective vaccination strategies may also be useful in speci®c high-risk adult populations, e.g.,

seronegative transplant recipients.

In initial efforts to identify an effective vaccine, the Towne strain of HCMV has been used,

which is a laboratory-adapted, attenuated virus variant, originally isolated from a newborn with

congenital disease. Initial vaccines conferred limited ef®cacy,122 so alternative strategies with

improved protective action were investigated. Thus included the use of subunit vaccines, directed

toward speci®c antigens,123,124 or DNA vaccination. This last methodology provides an elegant

vaccination approach, in which immunogenic proteins are synthesized by host cells after intra-

cellular delivery of genes encoding these proteins. The potential advantages of DNA vaccination

include the lack of an infectious agent or live vector, the ease of production, manipulation and

storage of DNA vaccines, and potentially long-lasting responses to DNA vaccination. DNA

vaccination against cytomegalovirus125 is currently undergoing evaluation in animal models with

good results.126,127

NEWHUMANCYTOMEGALOVIRUS INHIBITORS * 237

F. Conclusions

Existing antiviral treatments for the high prevalent HCMV suffer from toxicity and limited

effectiveness. Additionally, long-term therapies needed for managing HCMV infections in

immunosupressed patients provided the background for the emergence of resistance to currently

approved antivirals agents.

From the above reported studies, it is evident that several new HCMV inhibitors with less

adverse effects or/and new mechanism of action than current clinical drugs have been discovered.

Nonnucleosidic compounds show a great potential for the future treatment of HCMV infections

because of their potent and selective HCMV inhibition, and different mechanisms of action than that

of current clinically used drugs. Several of these promising candidates are in different clinical phase

studies which will greatly enable the advent of novel HCMV therapy.

R E F E R E N C E S

1. DeJong MD, Galasso GJ, Gazzard B, Grif®ths PD, Jabs DA, Kern ER, Spector SA. Summary of the II

International Symposium on Cytomegalovirus. Antiviral Res 1998;39:141±162.

2. Soderberg-Naucler C, Fish KN, Nelson JA. Growth of human cytomegalovirus in primary macrophages.

Methods 1998;16:126±138.

3. Emery VC, Cope AV, Bowen EF, Gor D, Grif®ths PD. The dynamics of human cytomegalovirus

replication in vivo. J Exp Med 1999;190:177±182.

4. Baldanti F, Sarasini A, Furione M, Gatti M, Comolli G, Revello MG, Gerna G. Coinfection of the

immunocompromised but not the immunocompetent host by multiple human cytomegalovirus strains.

Arch Virol 1998;143:1701±1709.

5. Goldman RC, Klein LL. Problems and progress in opportunistic infections. Ann Rep Med Chem

1994;29:155±164.

6. Contreras A, Slots J. Active cytomegalovirus infection in human periodontitis. Oral Microbiol Immunol

1998;13:225±230.

7. Stocchi R, Ward KN, Fanin R, Baccarani M, Apperley JF. Management of human cytomegalovirus

infection and disease after allogeneic bone marrow transplantation. Haematologica 1999;84:71±79.

8. Trincado DE, Scott GM, White PA, Hunt C, Rasmussen L, Rawlinson WD. Human cytomegalovirus

strains associated with congenital and perinatal infections. J Med Virol 2000;61:481±487.

9. O'Sullivan CE, Drew WL, McMullen DJ, Miner R, Lee JY, Kaslow RA, Lazar JG, Saag MS. Decrease of

cytomegalovirus replication in human immunode®ciency virus infected-patients after the treatment with

higly active antiretroviral therapy. J Infect Dis 1999; 180:847±849.

10. Holland GN. New strategies for the management of AIDS-related CMV retinitis in the era of potent

antiretroviral therapy. Ocul Immunol In¯amm 1999;7:179±188.

11. Nichols WG, Boeckh M. Recent advances in the therapy and prevention of CMV infections. J Clin Virol

2000;16:25±40

12. Perez JL. Resistance to antivirals in human cytomegalovirus: mechanism and clinical signi®cance.

Microbiologia 1997;13:343±352.

13. Erice A. Resistance of human cytomegalovirus to antiviral drugs. Clin Microbiol Rev 1999;12:286±297.

14. Augustynowicz E, Dziezanowska D. Molecular analysis of the human cytomegalovirus strains isolated

from infected infants. Acta Microbiol Pol 1998;47:373±383.

15. Horvath R, Cerny J, Benedik J, Holk J, Jelinkova I, Benedik J. The possible role of human

cytomegalovirus (HCMV) in the origin of atherosclerosis. J Clin Virol 2000;16:17±24.

16. Berensci K, Endresz V, Klurfeld D, Kari L, Kritchevsky D, Gonczol E. Early atherosclerotic plaques in

the aorta following cytomegalovirus infection of mice. Cell Adhes Commun 1998;5:39±47.

17. Faulds D, Heel RC. Ganciclovir. A review of its antiviral activity, pharmacokinetic properties and

therapeutic ef®cacy in cytomegalovirus infections. Drugs 1990;39:597±638.

18. Chrisp P, Clissold SP. Foscarnet. A review of its antiviral activity, pharmacokinetic properties and

therapeutic use in immunocompromised patients with cytomegalovirus retinitis. Drugs 1991;41:104±129.

238 * MARTINEZETAL.

19. De Clercq E. Therapeutic potential of HPMPC as an antiviral drug. Rev Med Virol 1993;3:85±96.

20. DeSmet MD, Meenken CJ, Van den Horn GJ. Fomivirsen: a phosphorothioate oligonucleotide for the

treatment of CMV retinitis. Ocul Immunol In¯amm 1999;7:189±198.

21. Bedard J, May S, Lis M, Tryphonas L, Drach J, Huffman J, Sidwell R, Chan L, Bowlin T, Rando R.

Comparative study of the anti-human cytomegalovirus activities and toxicities of a tetrahydrofuran

phosphonate analogue of guanosine and cidofovir. Antimicrob Agents Chemother 1999;43:557±567.

22. Mendez JC, Sia IG, Tau KR, Espy MJ, Smith TF, Chou S, Paya CV. Novel mutation in the CMV UL97

gene associated with resistance to ganciclovir therapy. Transplantation 1999;67:755±757.

23. Abraham B, Lastere S, Reynes J, Bibollet-Ruche F, Vidal N, Segondy M. Ganciclovir resistance and

UL97 gene mutations in cytomegalovirus blood isolates from patients with AIDS treated with

ganciclovir. J Clin Virol 1999;13:141±148.

24. Mulamba GB, Hu A, Azad RF, Anderson KP, Coen DM. Human cytomegalovirus mutant with sequence-

dependent resistance to the phosphorothioate oligonucleotide fomivirsen (ISIS 2922). Antimicrob Agents

Chemother 1998;42:971±973.

25. Post LE. Gene therapy: progress, new directions and issues. Ann Rep Med Chem 1995;35:219±226.

26. Harms JS, Oliveira SC, Splitter GA. Regulation of transgene expression in genetic immunization. Braz J

Med Biol Res 1999;32:155±162.

27. Field AK. Human cytomegalovirus: challenges opportunities and new drug development. Antiviral Chem

Chemother 1999;10:219±232.

28. Castro A, Martinez A. Novel agents for the treatment of human cytomegalovirus infection. Exp Opin

Ther Patents 2000;10:165±177.

29. Spector SA, Busch DF, Follansbee S, Squires K, Lalezari JP, Jacobson MA, Connor JD, Jung D, Shadman

A, Mastre B, Buhles W, Drew WL. Pharmacokinetics, safety, and antiviral pro®les of oral ganciclovir in

persons infected with human inmunode®ciency virus: A phase I-II study. J Infect Dis 1995;171:1431±1437.

30. Dillon MP, Cai H, Maag H. Application of the ` Trimethyl Lock'' to ganciclovir, a pro-drug with

increased oral bioavailability. Bioorg Med Chem Lett 1996;6:1653±1656.

31. Kim DK, Kim HK, Chae YB. Design and synthesis of 6-¯uoropurine acyclonucleosides: potential

prodrugs of acyclovir and ganciclovir. Bioorg Med Chem Lett 1994;4:1309±1312.

32. Kim DK, Chang K, Im GJ, Kim HT, Lee N, Kim KH. Synthesis and evaluation of 2-amino-9(1,3-

dihydroxy-2-propoxymethyl)-6-¯uoropurine mono- and diesters as potential prodrugs of ganciclovir. J

Med Chem 1999;42:324±328.

33. Bedard J, May S, Lis M, Tryphonas L, Drach JC, Huffman J, Sidwell R, Chan L, Bowlin T, Rando R.

Comparative study of the anti-human cytomegalovirus activities and toxicities of a tetrahydrofuran

phosphonate analogue of guanosine and cidofovir. Antimicrobial Agents Chemother 1999;43:557±567.

34. Fergunson CG, Gorin BI, Thatcher RJ. Design of novel derivatives of phosphonoformate (foscarnet) as

prodrugs and antiviral agents. J Org Chem 2000;65:1218±1221.

35. Wagner CR, Iver VV, MacInte EJ. Pronucleotides: Toward the in vivo delivery of antiviral and anticancer

nucleotides. Med Res Rev 2000;20: 417±451.

36. Swayze EE, Drach JC, Wotring LL, Townsend LB. Synthesis, antiproliferative and antiviral activity of

imidazo[4,5-d]isothiazole nucleosides as 5:5 fused analogs of nebularine and 6-methylpurine ribo-

nucleoside. J Med Chem 1997;40:771±784.

37. Meade EA, Wotring LL, Drach JC, Townsend LB. Synthesis and antiproliferative and antiviral activity of

carbohydrate pyrrolo[2,3-c]pyridazin-7-one nucleosides. J Med Chem 1997, 40:794±801.

38. Walker JA, Liu W, Wise DS, Drach JC, Townsend LB. Synthesis and antiviral evaluation of certain novel

pyrazinoic acid c-nucleosides. J Med Chem 1998;41:1236±1241.

39. Qiu YL, Hempel A, Camerman N, Camerman A, Geiser F, Ptak RG, Beitenbach JM, Kira T, Li L,

Gullen E, Cheng YC, Drach JC, Zemlicka J. (R)-(ÿ)- and (S)-(� )-Synadenol: Synthesis, absolute

con®guration, and enantioselectivity of antiviral effect. J Med Chem 1998;41:5257±5264.

40. Wang J, Froeyen M, Hendrix C, Andrei G, Snoeck R, De Clercq E, Herdewijn. The cyclohexene ring

system as a furanose mimic: Synthesis and antiviral activity of both enantiomers of cyclohexenylguanine.

J Med Chem 2000;43:736±745.

41. Guan HP, Ksebati MB, Cheng YC, Drach JC, Kern ER, Zemlicka J. Spiropentane mimics of nucleosides:

Analogues of 2 0-deoxyadenosine and 2 0-deoxyguanosine. Synthesis of all stereoisomers, isomeric

assignment, and biological evaluation. J Org Chem 2000;65:1280±1290.

NEWHUMANCYTOMEGALOVIRUS INHIBITORS * 239

42. Broom AD, Agrawal VK, Tutonda MG, Fain HD, Buckheit RW. Unusual single-stranded poly-

ribonucleotides as potent anti-HIV agents. J Med Chem 1995;38:3253±3257.

43. Tutonda MG, Fain HD, Buckheit RW, Broom AD. The role of hydrophobic versus hydrophilic base

character in the anti-HIV activity of purine-containing polyribonucleotides. Nucleosides Nucleotides

1999,16:173±182.

44. Tutonda MG, Buckheit RW, Agrawal VK, Broom AD. Antiviral Oligo- and polyribonucleotides

containing selected triazolo[2,3-a]purines. J Med Chem 1998;41:4958±4964.

45. Raic-Malic S, Hergold-Brundic A, Hagl A, Grdisa M, Pavelic K, De Clercq E, Mintas M. Novel

pyrimidine and purine derivatives of L-ascorbic acid: Synthesis and biological evaluation. J Med Chem

1999;42:2673±2678.

46. Krawczyk SH, Reza Nassiri M, Kucera LS, Kern ER, Ptak RG, Wotring LL, Drach JC, Townsend LB.

Synthesis and antiproliferative and antiviral activity of 2 0-Deoxy-2 0-¯uoroarabinofuranosyl analogs of

the nucleoside antibiotics toyocamycin and sangivamycin. J Med Chem 1995;38:4106±4114.

47. Krawczyk SH, Renau TE, Reza Nassiri M, Westerman AC, Wotring LL, Drach JC, Townsend LB.

Synthesis and evaluation of certain thiosangivamycin analogs as potential inhibitors of cell proliferation

and human cytomegalovirus. J Med Chem 1995;38:4115±4119.

48. Renau TE, Wotring LL, Drach JC, Townsend LB. Synthesis of non-nucleoside analogs of toyocamycin,

sangivamycin, and thiosangivamycin: in¯uence of various 7-substituents on antiviral activity. J Med

Chem 1996;39:873±880.

49. Renau TE, Kennedy C, Ptak RG, Breitenbach JM, Drach JC, Townsend LB. Synthesis of non-nucleoside

analogs of toyocamycin, sangivamycin, and thiosangivamycin: the effect of certain 4- and 4,6-Substi-

tuents on the Antiviral Activity of Pyrrolo[2,3-d]pyrimidines. J Med Chem 1996;39:3470±3476.

50. Jacobson JG, Renau TE, Nassiri MR, Sweier DG, Breitenbach JM, Townsend LB, Drach JC.

Nonnucleoside pyrrolopyrimidines with a unique mechanism of action against human cytomegalovirus.

Antimicrobial Agents Chemother 1999;43:1888±1894.

51. Townsend LB, Devivar RV, Turk SR, Nassiri MR, Drach JC. Design, synthesis, and antiviral activity of

certain 2,5,6-trihalo-1-(b-D-ribofuranosyl)benzimidazoles. J Med Chem 1995;38:4098±4105.

52. Underwood MR, Harvey RJ, Stanat SC, Hemphill ML, Miller T, Drach JC, Townsend LB, Biron KK.

Inhibition of human cytomegalovirus DNA maturation by a benzimidazole ribonucleoside is mediated

through the UL89 gene product. J Virol 1998;72:717±725.

53. Nassiri MR, Emerson SG, Devivar RV, Townsed LB, Drach JC, Taichman RS. Comparison of

benzimidazole nucleosides and ganciclovir on the in vivo proliferation and colony formation of human

marrow progenitor cells. Br J Haematol 1996,93:273±279.

54. Gudmundsson KS, Drach JC, Wotring LL, Townsend LB. Synthesis and antiviral evaluation of certain

5 0-modi®ed analogs of 2,5,6-trichloro-1-(b-D-ribofuranosyl)benzimidazole. J Med Chem 1997;40:785±

793.

55. Gudmundsson KS, Tidwell J, Lippa N, Koszalka GW, van Draanen N, Ptak RG, Drach JC, Townsend LB.

Synthesis and antiviral evaluation of halogenated -D- and L-erythrofuranosylbenzimidazoles. J Med Chem

2000;43:2464±2472.

56. Saluja S, Zou R, Drach JC, Townsend LB. Structure-activity relationships among 2-substituted

5,6-dichloro-, 4,6- dichloro, and 4,5-dichloro-1-[(2-hydroxyethoxy)methyl]- and -1-[(1,3-dihydroxy-2-

propoxy)methyl]benzimidazoles. J Med Chem 1996;39:881±891.

57. Migawa MT, Girardet JL, Walker II JA, Koszalka GW, Chamberlain SD, Drach JC, Townsend LB.

Design, synthesis, and antiviral activity of a-nucleosides: D- and L-isomers of lyxofuranosyl- and (5-

deoxyfuranosyl)benzimidazoles. J Med Chem 1998;41:1242±1251.

58. Girardet JL, Drach JC, Chamberlain SD, Koszalka GW, Townsend LB. Design, synthesis and antiviral

activity of a-L-arabinofuranosyl derivatives of 2-substituted-5,6-dichlorobenzimidazoles. Nucleosides

Nucleotides 1998;17:2389±2401.

59. Porcari AR, Devivar RV, Kucera LS, Drach JC, Townsend LB. Design, synthesis, and antiviral

evaluations of 1-(substitutedbenzyl)-2-substituted-5,6-dichlorobenzimidazoles as nonnucleoside analo-

gues of 2,5,6-trichloro-1-(b-D-ribofuranosyl)benzimidazole. J Med Chem 1998;41:1252±1262.

60. Zou R, Drach JC, Townsend LB. Design, synthesis and antiviral evaluation of 2-chloro-5,6-dihalo1-b-

D-ribofuranosylbenzimidazoles as potential agents for human cytomegalovirus infections. J Med Chem

1997;40:811±818.

240 * MARTINEZETAL.

61. Townsend LB, Gudmundsson KJ, Daluge SM, Chen JJ, Zhu Z, Koszalka GW, Boyd L, Chamberlain SD,

Freeman GA, Biron KK, Drach JC. Studies designed to increase the stability and antiviral activity

(HCMV) of the active benzimidazole nucleoside, TCRB. Nucleosides Nucleotides 1999;18:509±519.

62. Gudmundsson KS, Freeman GA, Drach JC, Townsend LB. Synthesis of ¯uorosugar analogues of

2,5,6-trichloro-1-(b-D-ribofuranosyl)benzimidazole as antivirals with potentially increased glycosidic

bond stability. J Med Chem 2000;43:2473±2478.

63. Townsend LB, Drach JC, Good SS, Daluge SM, Martin MT. Therapeutic nucleosides. Patent US

1996;5:534±535.

64. Chen JJ, Wei Y, Drach JC, Townsend LB. Synthesis and antiviral evaluation of trisubstituted indole

N-nucleosides as analogues of 2,5,6-trichloro-1-(b-D-ribofuranosyl)benzimidazole (TCRB). J Med Chem

2000;43:2449±2456.

65. Zou R, Ayres KR, Drach JC, Townsend LB. Synthesis and antiviral evaluation of certain disubstituted

benzimidazole ribonucleosides. J Med Chem 1996;39:3477±3482.

66. Zou R, Drach JC, Townsend LB. Design, synthesis and antiviral evaluation of 2-substituted 4,5-dichloro-

and 4,6-dichoro-1-b-D-ribofuranosylbenzimidazoles as potential agents for Human cytomegalovirus

infections. J Med Chem 1997;40:802±810.

67. Koszalka GW, Chamberlain SD, Harvey RJ, Frick LW, Good SS, Davis ML, Smith A, Drach JC,

Townsend LB, Biron KK. Benzimidazoles for the treatment of human cytomegalovirus. Antiv Res

1996;30:A43.

68. Zacny VL, Gershburg E, Davis MG. Inhibition of Epstein-Barr virus replication by a benzimidazole L-

riboside: novel antiviral mechanism of 5,6-dichloro-2-(isopropylamino)-1-b-L-ribofuranosyl-1H-benzi-

midazole. J Virol 1999,73:7271±7277.

69. Chan JH, Chamberlain SD, Biron KK, Davis MG, Harvey RJ, Selleseth DW, Dornsife RE, Dark EH,

Frick LW, Townsend LB, Drach JC, Koszalka GW. Synthesis and evaluation of a series of 2 0-deoxy

analogues of the antiviral agent 5,6-dichloro-2-isopropylamino-1-(b-L-ribofuranosyl)-1H-benzimidazole

(1263W94). Nucleosides Nucleotides Nucleic Acids 2000;19:101±123.

70. Krosky PM, Underwood MR, Turk SR, Feng KWH, Jain RK, Ptak RG, Westerman AC, Biron KK,

Townsend LB, Drach JC. Resistance of human cytomegalovirus to benzimidazole ribonucleosides maps

to two open reading frames: UL89 and UL56. J Virol 1998;72:4721±4728.

71. Zhu Z, Lippa B, Drach JC, Townsend LB. Design, synthesis, and biological evaluation of tricyclic

nucleosides (dimensional probes) as analogues of certain antiviral polyhalogenated benzimidazole

ribonucleosides. J Med Chem 2000;43:2430±2437.

72. Tucker JA, Clayton TL, Chidester CG, Schulz MW, Harrington LE, Conrad SJ, Yagi Y, Oien NL, Yurek D,

Kuo MS. Structure±activity relationship of acyloxyamidine cytomegalovirus DNA polymerase inhibitors.

Bioorg Med Chem 2000;8:601±615.

73. Slater MJ, Cockerill S, Baxter R, Bonser RW, Gohil K, Gowrie C, Robinson E, Litter E, Parry N,

Randall R, Snowden W. Indolocarbazole: Potent, selective inhibitors of human cytomegalovirus

replication. Bioorg Med Chem 1999;7:1067±1074.

74. He Z, He YS, Kim Y, Chu L, Ohmstede C, Biron KK, Coen DM. The human cytomegalovirus UL97

protein is a protein kinase that autophosphorylates on serines and threonines. J Virol 1997;71:405±411.

75. Chan L, Jin H, Stefanac T, LavalleÂe JF, Falardeau G, Wang W, BeÂdard J, May S, Yuen L. Discovery of 1,6-

naphthyridines as a novel class of potent and selective human cytomegalovirus inhibitors. J Med Chem

1999;42:3023±3025.

76. Chan L, Jin H, Stefanac T, Wang W, LavalleÂe JF, BeÂdard JS, Yuen L. Isoquinoline-6-carboxamides as

potent and selective anti-human cytomegalovirus (HCMV) inhibitors. Bioorg Med Chem Lett 1999;9:

2583±2586.

77. Chan L, Stefanac T, Turcotte N, Hu Z, Chen Y, BeÂdard JS, May S, Jin H. Design and evaluation of

dihydroisoquinolines as potent and orally bioavailable human cytomegalovirus inhibitors. Bioorg Med

Chem Lett 2000;10:1477±1480.

78. Bedard J, May S, L'Heureux L, Stamminger T, Copsey A, Drach J, Huffman J, Chan J, Jin H, Rando RF.

Antiviral properties of a series of 1,6-naphthyridine and 7,8-dihydroisoquinoline derivatives exhibiting

potent activity against human cytomegalovirus. Antimicrob Agents Chemother 2000;44:929±937.

79. Gueif®er A, Lhassani M, Elhakmaoui A, Snoeck R, Andrei G, Chavignon O, Teulade JC, Kerbal A,

Essassi EM, Debouzzy JC, Witvrouw M, Blache Y, Balzarini J, De Clercq E, Chapat JP. Synthesis of

NEWHUMANCYTOMEGALOVIRUS INHIBITORS * 241

C-acyclo-nucleosides in the imidazo[1,2-a]pyridine and pyrimidine series as antiviral agents. J Med

Chem 1996;39:2856±2859.

80. Gueif®er A, Mavel S, Lhassani M, Elhakmaoui A, Snoeck R, Andrei G, Chavignon O, Teulade JC,

Witvrouw M, Balzarini J, De Clercq E, Chapat JP. Synthesis of imidazo[1,2-a]pyridines as antiviral

agents. J Med Chem 1998;41:5108±5112.

81. Martinez A, Esteban AI, De Clercq E. Benzothiadiazine dioxide acyclonucleosides as lead compounds

for the development of new agents against human cytomegalovirus and varicella-zoster virus infections.

Bioorg Med Chem Lett 1997;7: 1031±1032.

82. Martinez A, Esteban AI, Castro A, Gil C, Conde S, Andrei G, Snoeck R, Balzarini J, De Clercq E. Novel

potential agents for human cytomegalovirus infection: Synthesis and antiviral activity evaluation of

benzothiadiazine acyclonucleosides. J Med Chem 1999;42:1145±1150.

83. Martinez A, Castro A, Gil C, Miralpeix M, Segarra V, Domenech T, Beleta J, Palcios JM, Ryder H,

Miro X, Bonet C, Casacuberta JM, Azorin F, PinÄa B, Puigdomenech P. Benzyl derivatives of 2,1,3-benzo-

and benzothieno[3,2-a]thiadiazine 2,2-dioxides: First phosphodiesterase 7 inhibitors. J Med Chem

2000;43:683±689.

84. Martinez A, Gil C, Perez C, Castro A, Prieto C, Otero J. Chlorophenylmethyl benzothiadiazine dioxides

derivatives: Potent human cytomegalovirus inhibitors. Bioorg Med Chem Lett 1999;9:3133±3136.

85. Martinez A, Gil C, Abasolo MI, Castro A, Bruno AM, Perez C, Prieto C; Otero J. Benzothiadiazine

dioxides (BTD) dibenzyl derivatives as potent human cytomegalovirus inhibitors. Synthesis and

comparative molecular ®eld analysis. J Med Chem 2000;43:3218±3225.

86. Martinez A, Gil C, Perez C, Castro A, Andrei G, Snoeck R, Balzarini J, De Clerq E. Nonnucleoside

human cytomegalovirus inhibitors. Synthesis and antiviral evaluation of chlorophenylmethyl benzothia-

diazine dioxides (BTD) derivatives. J Med Chem 2000;43:3267±3273.

87. Gibson W, Hall MR. Assemblin, an essential herpesvirus proteinase. Drug Design Discov 1997;15:39±47.

88. Holwerda BC. Herpesvirus proteases: targets for novel antiviral drugs. Antiviral Research 1997;35:1±21.

89. Flynn DL, Abood NA, Holwerda BC. Recent advances in antiviral research: identi®cation of inhibitors of

the herpesvirus proteases. Current Opin Chem Biol 1997;1:190±196.

90. Newcomb WW, Homa FL, Thomsen DR, Booy FP, Trus BL, Steven AC, Spencer JV, Brown JC.

Assembly of the herpes simplex virus capsid: Characterization of intermediates observed during cell-free

capsid formation. J Mol Biol 1996;263.432±446.

91. Holwerda BC, Witter AJ, Duf®n KL, Smith C, Toth MW, Carr LS, Wiegand RC, Bryant ML. Activity of

two-chain recombinant human cytomegalovirus protease. J Mol Biol 1994;269:25911±25915.

92. Flynn DL, Becker DP, Dilworth VM, Highkin MK, Hippenmeyer PJ, Houseman KA, Levine LM, Li M,

Moormann AE, Rankin A, Toth MV, Villamil CI, Wittwer AJ, Holwerda BC. The herpesvirus protease:

Mechanistic studies and discovery of inhibitors of the human cytomegalovirus protease. Drug Design

Discov 1997;15:3±15.

93. Huffaker TK, Binford S, Patick AK,Pinko C, Kan CC, Zalman LS. Comparison of human

cytomegalovirus (HCMV) protease sequences among laboratory strains and seven clinical isolates.

Antiviral Res 1997;33:215±218.

94. Qian C, Lagace L, Massariol MJ, Chabot C, Yoakim C, Deziel R, Tong L. A rational approach towards

successful crystallization and crystal treatment of human cytomegalovirus protease and its inhibitor

complex. Acta Crystallogr D Biol Crystalogr 2000;56:175±180.

95. Chen P, Tsuge H, Almassy RJ, Gribskov CL, Katoh S, Vanderpool DL, Margosiak SA, Pinko C,

Matthews, Kan CC. Structure of the human cytomegalovirus protease catalytic domain reveals a novel

serine protease fold and catalytic triad. Cell 1996;86: 835±843.

96. Shieh HS, Kurumbail RG, Stevens AM, Stegeman RA, Sturman EJ, Pak JY, Wittwer AJ, Palmier MC,

Wiegand RC, Holwerda BC, Stallings WC. Three dimensional structure of human cytomegalovirus

protease. Nature 1996;383:279±282.

97. Qiu X, Culp JS, DiLella AG, Hellming B, Hoog SS, Janson CA, Smith WW, Abdel-Meguid SS. Unique

fold and active site in cytomegalovirus protease. Nature 1996;383:275±279.

98. Tong L, Qian C, Massariol MJ, Bonneau PR, Cordingley MG, Lagace. A new serine-protease fold

revealed by the crystal structure of human cytomegalovirus protease. Nature 1996;383:272±275.

99. Tong L, Qian C, Massariol MJ, dezeil R, Yoakim C, Lagace L. Conserved mode of peptidomimetic

inhibition and substrate recognition of human cytomegalovirus protease. Nat Struct Biol 1998;5:819±826.

242 * MARTINEZETAL.

100. LaPlante SR, Bonneau PR, Aubry N, Cameron DR, DeÂziel R, Grand-Maitre C, Plouffe C, Tong L,

Kawai SH. Characterization of human cytomegalovirus protease as an induced-®t serine protease and the

implications to the design of mechanism-based inhibitors. J Am Chem Soc 1999; 121:2974±2986.

101. Ogilvie W, Bailey M, Poupart MA, Abraham A, Bhavsar A, Bonneau P, Bordeleau J, Bousquet Y,

Chabot C, Duceppe JS, Fazal G, Goulet S, Grand-Maitre C, Guse I, Halmos T, Lavalle P, Leach M,

Malenfant E, O'Meara J, Plante R, Plouffe C, Poirier M, Soucy F, Yoakim C, DeÂziel R. Peptidomimetic

inhibitors of the human cytomegalovirus protease. J Med Chem 1997;40:4113±4135.

102. Bonneau PR, Grand-Maitre C, Greenwood DJ, Lagace L, LaPlante SR, Massariol M-J, Ogilvie WW,

O'Meara JA, Kawai SH. Evidence of a conformational change in the human cytomegalovirus protease

upon binding of peptidyl-activated carbonyl inhibitors. Biochemistry 1997; 36:12644±12652.

103. LaPlante SR, Aubry N, Bonneau PR, Cameron DR, Lagace L, Massariol MJ, Montpetit H, Plouffe C,

Kawai SH, Fulton BD, Chen Z, Ni F. Human cytomegalovirus protease complexes its substrate

recognition sequences in an extended peptide conformation. Biochemistry 1998;27:9793±9801.

104. Borthwick AD, Weingarten G, Haley TM, Tomaszewski M, Wang W, Hu Z, Bedard J, Jin H, Yuen L,

Mansour TS. Design and synthesis of monocyclic b-lactams as mechanism-based inhibitors of human

cytomelagovirus protease. Bioorg Med Chem Lett 1998;8:365±370.

105. DeÂziel R, Malenfant E. Inhibition of human cytomegalovirus protease No with monocyclic b-lactams.

Bioorg Med Chem Lett 1998;8:1437±1442.

106. Bonneau PR, Hasine F, Plouffe C, Malenfant E, LaPlante SR, Guse I, Ogilvie WW, Plante R,

Davidson WC, Hopkins JL, Morelock MM, Cordingley MG, DeÂziel R. Inhibition of human

cytomegalovirus protease by monocyclic b-lactam derivatives: Kinetic characterization using ¯uorescent

probe. J Am Chem Soc 1999; 121:2965±2973.

107. Yoakim C, Ogilvie WW, Cameron DR, Chabot C, Guse I, Hache B, Naud J, O'Meara JA, Plante R,

DeÂziel R. b-Lactam derivatives as inhibitors of human cytomegalovirus protease. J Med Chem 1998;41:

2882±2891.

108. Ogilvie WW, Yoakim C, Do F, Hache B, Laglace L, Haud J, O'Meara JA, DeÂziel R. Synthesis and

antiviral activity of monolactams inhibiting the human cytomegalovirus protease. Bioorg Med Chem

1999;7:1521±1531.

109. Pinto IL, West A, Debouck CM, DiLella A, Gorniak JG, O'Donnell KC, O'Shannessy DJ, Patel A,

Jarvest RL. Novel, selective mechanism-based inhibitors of herpes protease. Bioorg Med Chem Lett

1996;6:2467±2472.

110. Abood NA, Schretzman LA, Flynn DL, Houseman KA, Wittwer AJ, Dilworth VM, Hippenmeyer PJ,

Holwerda BC. Inhibition of human cytomegalovirus protease by benzoxazinones and evidence of

antiviral activity in cell culture. Bioorg Med Chem Lett 1997;7:2105±2108.

111. Jarvest RL, Parratt MJ, Debouck CM, Gorniak JG, Jennings LJ, Sera®noska HT, Strickler JE. Inhibition

of HSV-1 protease by benzoxazinones. Bioorg Med Chem Lett 1996;6:2463±2466.

112. Jarvest RL, Connor SC, Gorniak JG, Jennings LJ, Sera®nowska HT, West A. Potent selective

thienoxazinone inhibitors of herpes proteases. Bioorg Med Chem Lett 1997;7:1733±1738.

113. Jarvest RL, Pinto IL, Asham SM, Dabrowski CE, Fernandez AV, Jennings, Lavery P, Tew DG. Inhibition

of herpes proteases and antiviral activity of 2-substituted thieno[2,3-d]oxazinones. Bioorg Med Chem

Lett 1999;9:443±448.

114. Pinto IL, Javerst RL, Clarke B, Dabrowski CE, Fenwick A, Gorczyca MM, Jennings LJ, Lavery P,

Sternberg EJ, Tew DG, West A. Inhibiton of human cytomegalovirus protease by enedione derivatives of

thieno[2,3-d]oxazinones through a novel dual acylation/alkylation mechanism. Bioorg Med Chem Lett

1999;9:449±452.

115. Smith DG, Gribble AD, Haigh D, Ife RJ, Lavery P, Skett P, Slingsby BP, Stacey R, Ward RW, West A. The

inhibiton of human cytomegalovirus (hCMV) protease by hydroxylamine derivatives. Bioorg Med Chem

Lett 1999;9:3137±3142.

116. Bedard J, Barbeau D, Yuen L, Rando RF, Bowlin TL. A high throughput colorimetric cell proliferation

assay for the identi®cation of human cytomegalovirus inhibitors. Antiviral Res 1999;41:35±43.

117. Bonneau PR, Plouffe A, Wernic D, Poupart MA. Design of ¯uorogenic peptide substrates for human

cytomegalovirus protease based on structure-activity relationship studies. Anal Biochem 1998;255:59±65.

118. Guo B, Dai JR, Ng S, Huang Y, Leong C, Ong W, Carte BK. Cytonic acids A and B: Novel tripepside inhibitors of

hCMV protease from the endophytic fungus Cytonaema Species. J Med Chem 2000;63:602±604.

NEWHUMANCYTOMEGALOVIRUS INHIBITORS * 243

119. Qian-Cutrone J, Kolb JM, McBrien K, Huang S, Gustavson D, Lowe SE, Nanly SP. Quanolirones I and II,

two new human cytomegalovirus protease inhibitors produced by Streptomyces sp. WC76535. J Nat Prod

1998;61:1379±1382.

120. Dhanak D, Keenan RM, Burton G, Kaura A, Darcy MG, Shah DH, Ridgers LH, Breen A, Lavery P,

Tew DG, West A. Benzothiopyran-4-one based reversible inhibitors of the human cytomegalovirus

(HCMV) protease. Bioorg Med Chem Lett 1998;8:3677±3682.

121. Ertl P, Cooper D, Allen G, Slater MJ. 2-chloro-3-substitued-1,4-Naphthoquinone inactivators of human

cytomegalovirus protease. Bioorg Med Chem Lett 1998;9:2863±2866.

122. Plotkin SA, Higgins R, Kurtz JB, Morris P, Campbell AJ, Shope TC, Spector SA, Danker WM.

Multicenter trial of Towne strain attenuated virus vaccine in seronegative renal transplant recipients.

Transplantation 1994;58:1176±1178.

123. Park JW, Kim DJ, Kim J, Park CG, Hwang ES, Cha CY. Little role of anti-gB antibodies in neutralizing

activity of patient's sera with human cytomegalovirus infection. J Korean Med Sci. 2000;15:133±138.

124. Tackaberry ES, Dudani AK, Prior F, Tocchi M, Sardana R, Altosaar I, Ganz PR. development of

biopharmaceuticals in plant expression systems: cloning, expression and immunological reactivity of

human cytomegalovirus glycoprotein B (UL55) in seeds of transgenic tobacoo. Vaccine 1999;17:3020±

3029.

125. Hwang ES, Kwon KB, Park JW, Kim DJ, Park CG, Cha CY. Induction of neutralizing antibody against

human cytomegalovirus (HCMV) with DNA-mediated immunization of HCMV glycoprotein B in mice.

Microbiol Immunol 1999;43:307±310.

126. Gonzalez JC, Morello C, Cranmer LD, Spector DH. DNA immunization confers protection against

murine cytomegalovirus infection. J Virol 1996;70:7921±7928.

127. Morello CS, Cranmer LD, Spector DH. Suppresion of murine cytomegalovirus (MCMV) replication with

a DNA vaccine encoding MCMV M84 (a homolog of human cytomegalovirus pp65). J Virol 2000;74:

3696±3708.

Ana Martinez received her Ph.D from Complutense University of Madrid in 1987. In 1988, she began a CSIC

post-doctoral fellowship at Medicinal Chemistry Institute with Dr. P. Goya where she applied the molecular

modeling techniques to different ®elds of drug design. In 1990 she joined Medicinal Chemistry Institute as a

tenured scientist. Currently, her laboratory is interested in the design, synthesis, and evaluation of antiviral

drugs, specially those for opportunistic infections, and in the discovery of new therapeutic strategies for the

treatment of neurodegenerative disease.

Ana Castro graduated from Autonoma University of Madrid and received her Ph.D. in 1996, also from

Autonoma University, under the supervision of Dr. A. Martinez. Following 2 years as a postdoctoral fellow in

the laboratory of Professor W. Graham Richards at Oxford University, she joined the Medicinal Chemistry

Institute. Her current research interest includes rational development of bioactive molecules focused on two

different ®elds: antiviral agents and neurodegenerative disorders.

Carmen Gil received her B.S. degree in pharmacy at Complutense University of Madrid in 1995. She

completed her graduate education in medicinal chemistry under the supervision of Dr. A. Martinez at Medicinal

Chemistry Institute where She is currently a pre-doctoral fellow. She current research interests include the

synthesis of new anti-HCMV inhibitors.

Concepcin PeÂrez obtained her Ph.D from Complutense University of Madrid in 1995, investigating the

differentiation of U937 promonocytic cells by antitumor DNA topoisomerase II inhibitors. She joined the

Medicinal Chemistry Institute in 1998. Her current interests include antiviral agents and Alzheimer disease.

244 * MARTINEZETAL.