Non-Nucleoside Reverse Transcriptase Inhibitors: The NNRTI Boom

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 30

Antiviral Chemistry & Chemotherapy 10:285–314

Review

Non-nucleoside reverse transcriptase inhibitors: the


NNRTI boom
Ole S Pedersen and Erik B Pedersen*

Department of Chemistry, University of Southern Denmark, Odense University, DK-5230 Odense M, Denmark

*Corresponding author: Tel: +45 6550 2555; Fax: +45 6615 8780; E-mail: EBP@chem.sdu.dk

Non-nucleoside reverse transcriptase inhibitors approved NNRTI drugs and focuses on the recent
(NNRTIs) are promising drugs for the treatment of efforts being made to produce second genera-
HIV when used in combination with other anti- tion inhibitors that circumvent this resistance
HIV drugs such as nucleoside reverse transcriptase problem.
(RT) inhibitors and protease inhibitors. The first
generation of NNRTIs have, however, suffered Keywords: HIV-1; non-nucleoside reverse
from the rapid development of resistance. This transcriptase inhibitors; HEPT; nevirapine;
review discusses the properties of the FDA- delavirdine; efavirenz; trovirdine

Introduction

The HIV epidemic is still a major concern. Virtually every interest in NNRTIs we intend to review this class of com-
country in the world has seen new infections in 1998, and pounds, especially the second generation of NNRTIs, using
the epidemic is out of control in many places according to a chemical approach. This review will focus on the recent
the World Health Organization (WHO) and the Joint and most interesting published results. The NNRTIs syn-
United Nations Programme on HIV/AIDS (UNAIDS). thesized before 1996 are covered by reviews by Artico
The introduction of highly active antiretroviral therapy (1996) and Tucker et al. (1996). A review focusing on the
(HAART), which has been used mainly in North America role of NNRTIs in the therapy of HIV-1 infections has
and Western Europe, has reduced the number of deaths been published by De Clercq (1998a).
caused by AIDS. However, because new infections continue
to occur and infected people are kept alive with HAART Non-nucleoside reverse transcriptase
and other combinations of anti-HIV drugs, the number of inhibitors
people living with HIV has increased in North America
and Western Europe. Combinations of anti-HIV drugs NNRTIs are bound in a hydrophobic pocket proximal to
often contain a non-nucleoside reverse transcriptase (RT) the catalytic site of RT in HIV-1 (Tantillo et al., 1994). X-
inhibitor (NNRTI), a nucleoside RT inhibitor (NRTI) and ray crystallographic studies of NNRTIs in complex with
a protease inhibitor. This review will concentrate on the RT (Ren et al., 1995; Ding et al., 1995) have shown that the
NNRTIs, of which the first, nevirapine (Viramune, NNRTIs maintain a very similar conformational ‘butterfly-
Boehringer Ingelheim), was approved as a drug for the like’ shape and appear to function as π-electron donors to
treatment of HIV-1 infection by the US Food and Drug aromatic side-chain residues surrounding the binding
Administration (FDA) in 1996. Nevirapine was followed pocket (Kroeger et al., 1995; De Clercq, 1998b).
by delavirdine mesylate (Rescriptor, Pharmacia & Upjohn) The major problem in the development of new
and efavirenz (Sustiva, DuPont), approved for the treat- NNRTIs is the rapid emergence of resistant strains of
ment of HIV-1 infection by the FDA in 1997 and 1998, HIV-1 in cell culture and patients. In patients receiving
respectively. A further three compounds, MKC-442 monotherapy with nevirapine, drug resistance developed
(Triangle Pharmaceuticals), Calanolide A (Sarawak rapidly owing to mutations in the RT; particularly signifi-
MediChem Pharmaceuticals) and AG 1549 (Agouron cant is the Y181C mutation (Cywin et al., 1998). Cross-
Pharmaceuticals) are in clinical trials according to the resistance is also a contributing factor and
Pharmaceutical Research and Manufacturers of America pyridinone-resistant strains containing the Y181C, K103N
(PhRMA) 1998 survey report. Because of the increasing or both mutations (Nunberg et al., 1991) have been found

©1999 International Medical Press 0956-3202/99/$17.00 285


OS Pedersen & EB Pedersen

Figure 1. The structure of nevirapine mutant strains of HIV-1 have to be considered in the devel-
opment of new NNRTI drug candidates. These aspects
O include metabolic stability, clearance rates, the ability to
H3C 5H cross the blood–brain barrier and protein binding. Protein
N
binding is a complex issue; a high protein binding could
4
reduce the metabolism of the drug, the clearance rates and
11 maintain high concentration of the drug in the blood.
2 N N N However, too strong protein binding may reduce the con-
1 10
centration of the free drug available for inhibitory action.
Some of the currently most interesting subclasses of
NNRTIs are described below. Tables showing the antiviral
activity and activity against purified RT are presented. The
to confer resistance to both TIBO R82150 (Pauwels et al., presentation of each drug begins with the subclass with an
1990) and nevirapine (Merluzzi et al., 1990). It has been FDA approved drug, followed by a subclass with a drug in
shown that NNRTIs rapidly select for resistant mutant clinical trials according to the PhRMA 1998 survey report,
HIV-1 strains when selection pressure is applied by drugs and ends with a description of some additional characteris-
in vitro (Kleim et al., 1995; Nunberg et al., 1991) or in tic subclasses.
monotherapy (Cywin et al., 1998; Miller et al., 1998).
Cross-resistance has been observed between many Nevirapine
NNRTIs in development (Miller et al., 1998). Mutations Nevirapine (Viramune; Boehringer Ingelheim) (Figure 1)
commonly selected for by NNRTIs occur at amino acid was approved for the treatment of HIV in combination
positions 98 to 108, 179 to 190 and 230 to 236 (Miller et with nucleoside analogues in 1996 in the USA, and in 1998
al., 1998). Cross-resistance is one of the obstacles that has in the European Union.
to be overcome for the next generation of NNRTIs, new Nevirapine has a good potency against wild-type RT,
drugs that have a resistance profile that differs from the with a 50% inhibitory concentration of 84 nM (Hargrave
resistance profile of the already known drugs. et al., 1991), good metabolic stability, good bioavailability
Many aspects beside selectivity and activity against (Cywin et al. 1998) and crosses the blood–brain barrier eas-

Table 1. The activity of some nevirapine analogues against wild-type RT and two mutant strains of HIV-1 RT

H3C O
N

N N
N
R1
R2

IC50 (µM) HIV-1 RT*


Compound R1 R2 WT† Y181C Y188L Reference
1 Cl Et 0.08 0.21 ND‡ Proudfoot et al. (1995a)
2 N-pyrrolyl Et 0.09 0.21 ND Proudfoot et al. (1995a)
3 2-furanyl Et 0.11 0.16 ND Proudfoot et al. (1995a)
4 3-furanyl Et 0.04 0.11 ND Proudfoot et al. (1995a)
5 2-pyrrolyl Et 0.07 0.07 ND Proudfoot et al. (1995a)
6 3-pyrrolyl Et 0.033 0.050 ND Kelly et al. (1997)
7 3-pyrrolyl c-Pr 0.05 0.06 ND Proudfoot et al. (1995a)
8 4-pyrazolyl Et 0.02 0.06 ND Proudfoot et al. (1995a)
9 4-pyrazolyl c-Pr 0.06 0.05 ND Proudfoot et al. (1995a)
10 4-NH2-phenyl Et 0.04 0.12 ND Proudfoot et al. (1995a)
11 indol-3-yl Et 0.028 0.028 0.090 Kelly et al. (1997)
12 5-azaindol-3-yl Et 0.044 0.098 0.384 Kelly et al. (1997)

*Inhibitor concentration to give 50% inhibition of incorporation of [3H]dGTP into a [(poly)rC•(oligo)dG] template.
†Wild-type HIV-1RT.
‡ND, Not determined.

286 ©1999 International Medical Press


The NNRTI boom

Table 2. The activity of some nevirapine analogues against wild-type RT and two mutant strains of HIV-1 RT
H3 C O
N
R2

N N
N
R1

IC50 (µM) HIV-1 RT*


Compound R1 R2 WT† Y181C Y188L Reference
13 Cl (3-ureidophenyl)ethyl 0.06 0.08 0.23 Klunder et al. (1998)
14 Cl (3-anilinyl)ethyl 0.12 0.23 0.25 Klunder et al. (1998)
15 Cl (4-(2-aminopyridyl))ethyl 0.02 0.07 0.16 Klunder et al. (1998)
16 F (4-pyridyl)ethyl 0.05 0.08 0.25 Klunder et al. (1998)
17 I (4-pyridyl)ethyl 0.09 0.18 0.43 Klunder et al. (1998)
18 Cl (4-pyridyl)ethyl 0.08 0.12 1.85 Klunder et al. (1998)
19 Cl (phenylthio)methyl 0.02 0.01 0.03 Cywin et al. (1998)
20 Cl (phenyloxy)methyl 0.11 0.06 0.64 Cywin et al. (1998)
21 Cl (2-methyl-4-pyridinyl-oxy)methyl 0.03 0.03 0.23 Cywin et al. (1998)
22 Cl phenylethyl 0.05 0.15 0.96 Cywin et al. (1998)
23 Cl (3-(aminocarbonyl)phenyloxy)methyl 0.13 0.28 0.34 Cywin et al. (1998)
24 Cl (3-aminophenyloxy)methyl 0.05 0.10 0.37 Cywin et al. (1998)

*Inhibitor concentration to give 50% inhibition of incorporation of [3H]dGTP into a [(poly)rC•(oligo)dG] template.
†Wild-type HIV-1 RT.

ily (Glynn & Yazdanian, 1998). Like all current NNRTIs, HIV activity against both wild-type RT and a broad spec-
nevirapine selects for mutations in the RT, the most com- trum of mutant RTs (Klunder et al., 1998). The aim in the
mon resistant strain of HIV-1 being characterized by the synthesis of these derivatives has been to achieve multiple
Y181C mutation. Mutations appear rapidly in response to interaction points with the enzyme or to achieve interac-
treatment with NNRTIs administered as monotherapy, tion between the compound and some more conserved
and the focus of researchers working with nevirapine ana- residues of RT. Some of these residues include the catalyt-
logues has been to develop drug candidates that retain ic aspartic acid residues, Asp-110, Asp-185 and Asp-186.
activity against both wild-type HIV-1 and known Mutation of these residues results in inactive RT enzymes
NNRTI-resistant HIV-1 strains. (Larder et al., 1987). Further research has investigated the
The substitution pattern of the dipyridodiazepinone linker between the aryl substituent and the dipyridodi-
ring system in the search for new derivatives was changed azepine system (Cywin et al., 1998). The preferred substi-
from C-4 and N-11 to N-5 and N-11, as this appears to be tution at the 8 position was with an aryloxymethyl or an
optimum for activity against both wild-type RT and arylthiomethyl, the arylthiomethyl derivatives being the
Y181C RT (Proudfoot et al., 1995a). Molecular modelling most active compounds (Table 2). The aryloxymethyl com-
of the X-ray crystal structure of nevirapine bound to wild- pounds though, were metabolically more stable, less toxic
type RT has revealed a lipophilic cavity proximal to the C- and yet possessed good activity, and are thus the choice for
4 position, which allows for placement of an aryl group at further development rather than the thio analogues (Cywin
this position. Although the placement of an amino group et al., 1998).
in the para position of this aryl group produced derivatives Several derivatives of nevirapine with changes in the
conferring activity against the Y181C mutant enzyme, the ring system have been synthesized and tested. Some of
activity was not strong (Kelly et al., 1995). Adding an aro- these are the dipyrido[2,3-b:2′,3′-c]diazepinones
matic substituent at position 2 afforded several analogues (Proudfoot et al., 1995b), pyridazinobenzodiazepines
with activity against both wild-type RT and Y181C (Heinisch et al., 1997a; Barth et al., 1996) and pyridoben-
mutant RT (Proudfoot et al., 1995a) and some have also zodiazepines (Hargrave et al., 1991). However, none of
shown activity against the Y188L mutant RT (Kelly et al., these have shown an improved anti-HIV potency.
1997) (Table 1). Changing the position of the aromatic Dibenzoxapinones and pyridobenzoxazepinones (Tables
substituent from position 2 to position 8, and introducing 3 and 4) are analogues of nevirapine that have shown activ-
an ethyl linker, have afforded new analogues with anti- ity against HIV-1 RT in the nanomolar range (Klunder et

Antiviral Chemistry & Chemotherapy 10:6 287


OS Pedersen & EB Pedersen

Table 3. The anti-HIV-1 RT activity of some Table 4. The anti-HIV-1 RT activity of some
dibenz[b,f][1,4]oxazepin-11(10H)-ones pyrido[2,3-b][1,5]benzoxazepin-5(6H)-ones
R1 O
R1 O
N 1
9 N
7 4
2
8 3
8
R2 R3
7
O 3 R3
6 4 R2 O 2
9 N
1
Compound R1 R2 R3 IC50 (nM)*
1 CH3 7-CH3 2-NH2 30 Compound R1 R2 R3 IC50 (nM)*
2 CH2CH3 9-CH3 2-NH2 60 8 CH2CH3 8,9-(CH3)2 3-NH2 71
3 CH3 7,9-(CH3)2 2-NH2 20 9 CH(CH3)2 8,9-(CH3)2 3-NH2 45
4 CH2CH3 7,9-(CH3)2 2-NH2 43 10 CH3 7,9-(CH3)2 3-NH2 46
5 CH3 7,9-(CH3)2 H 57 11 CH2CH3 7,9-(CH3)2 3-NH2 27
6 CH3 7,9-(CH3)2 2-CN 50 12 CH2CH3 7,9-(CH3)2 H 29
7 CH3 7,9-(CH3)2 2-OH 63 13 CH2CH3 7-NO2, 9-CH3 H 31
14 CH2CH3 7-CN, 9-CH3 H 19
*Inhibitor concentration to give 50% inhibition of incorporation of
[3H]dGTP into a [(poly)rC•(oligo)dG] template. 15 CH3 7-CN, 9-CH3 H 55
Data from Klunder et al. (1992). 16 CH3 7-NO2, 9-CH3 H 23
17 CH3 7-CO2CH3, 9-CH3 H 22

al., 1992). As a class, oxazepinones are less potent HIV-RT *Inhibitor concentration to give 50% inhibition of incorporation of
[3H]dGTP into a [(poly)rC•(oligo)dG] template.
inhibitors than the diazepinones. This problem has been Data from Klunder et al. (1992).
partly solved by substituent optimization. However, solu-
bility is a problem for this class of compounds because
oxazepinones have the disadvantage of being less soluble for clinical evaluation (Romero et al., 1994). The structures
than diazepinones. Replacing a phenyl ring with a pyrimi- of delavirdine mesylate (U90152S) and atevirdine mesylate
dine ring in the dibenzoxazepinones may increase the sol- (U87201E) are shown in Figure 2.
ubility, but is often accompanied by a decrease in the Later work has led to new analogues, the (alky-
inhibitory effect on RT (Klunder et al., 1992). lamino)piperidine BHAPs (AAP-BHAP). This has pro-
duced compounds with activity against RT containing the
BHAP Y181C and P236L mutations. The proline to leucine
Bis(heteroaryl)piperazine (BHAP) RT inhibitors form a mutation at position 236 appears in RT after several HIV-
class of compounds, discovered by the Upjohn laboratories 1 passages in vitro in the presence of increased concentra-
(delavirdine, atevirdine; Pharmacia & Upjohn) (Romero et tions of atevirdine or delavirdine (Romero et al., 1996).
al., 1991). This subclass of NNRTIs contain delavirdine Some AAP-BHAPs have, in a structure–activity relation-
mesylate (rescriptor), which was given FDA approval in ship programme, demonstrated excellent activity against
April 1997 for therapeutic use against HIV-1. Atevirdine the wild-type virus (Romero et al., 1996) (Table 5).
mesylate is another compound of this class that was chosen Delavirdine has been found to block the replication of 25

Figure 2. The structures of delavirdine mesylate and atevirdine mesylate

HN
HN H3CO
H3CSO2-NH

N N
N N
N
N N
N H
H .CH3SO3H
.CH3SO3H O
O

Delavirdine mesylate Atevirdine mesylate

U90152S U87201E

DLV ATV

288 ©1999 International Medical Press


The NNRTI boom

Table 5. The in vitro inhibition of recombinant HIV-1IIIB RT mutants


R2

R3 HN

N
R4 N N N
H
O R1

IC50 (µM)*
Compound R1 R2 R3 R4 WT† P236L Y181C
Atevirdine mesylate 2.3 >60 >60
Delavirdine mesylate 0.26 18.0 8.32
Nevirapine 3.1 0.32 >60
1 mesylate Me i-Pr H H 0.23 0.74 0.80
2 Me i-Pr H (OCH2CH2)OH 0.3 0.41 0.77
3 Me i-Pr CH3SO2HN H 0.5 1.5 1.1
4 Me i-Pr CH3NHCONH H 0.18 NT‡ 1.6
5 c-Pr i-Pr H H 0.70 0.59 2.31
6 Me i-Pr H HOCH2 0.17 0.37 0.49

*IC50 (µM), concentration that inhibited the RNA-dependent DNA polymerase activity of RT by 50% using a rA.dT template:primer.
†Wild-type HIV-1 RT.
‡NT, Not tested.
Data from Romero et al. (1996).

primary HIV-1 isolates in peripheral blood lymphocytes replaced C-6 (Romero et al., 1996). Substituting an ethyl
(PBL), including variants that are highly resistant to 3′- for a methyl group on the aminopiperidine spacer
azido-3′-deoxythymidine (zidovudine) and 2′,3′-dideoxyi- enhanced the activity against Y181C and P236L mutant
nosine (didanosine), with a mean 50% effective dose (ED50) RTs (Romero et al., 1996). Some of the most active com-
of 0.066±0.137 µM (Dueweke et al., 1993). The mutations pounds in this series are presented in Table 6.
Y181C and K103N conferred some resistance to delavir- Replacing the 3-alkylamino in both BHAPs and AAP-
dine, as for many other NNRTIs. However, delavirdine has BHAPs with alkoxy groups has also been investigated
been shown to inhibit the Y181C mutant RT with the (Genin et al., 1996). Incorporation of a 3-alkoxy sub-
rA.dT template:primer at a 50% inhibitory concentration stituent has been shown to be beneficial on the metabolic
(IC50) of 8.3 µM, whereas nevirapine and L-697,661 failed stability, evaluated in the presence of hepatic microsomal
to achieve 50% inhibition at 60 µM. This, together with cytochrome P450 in vitro, thus providing analogues with
good oral bioavailability and good serum drug levels in ani- similar or greater metabolic stability than delavirdine.
mals, made the mesylate salt of delavirdine a candidate for Some of these compounds possessed good activity against
clinical trials (Dueweke et al., 1993). wild-type and P236L RT enzymes, but the activity against
These compounds did not, however, possess the desired Y181C RT was diminished (Genin et al., 1996).
pharmacokinetic profile, primarily due to high clearance
rates. This afforded a new structure–activity relationship Benzoxazinones
programme focusing on compounds with increased activi- Benzoxazinones are the third class of compounds from which
ty against mutant RT (P236L and Y181C) with the appro- a non-nucleoside inhibitor has been approved by the FDA.
priate pharmaceutical properties. These included alteration Efavirenz (DMP-266; DuPont) (Table 7) was approved in
of the 3-pyridine substituent because N-dealkylation of the September 1998 by the FDA for once-daily dosing to be used
pyridine 3-alkylamino substituent is a predominant path- in combination with other anti-HIV drugs in both adult and
way for metabolism of the BHAPs (Romero et al., 1996). paediatric patients.
Work included varying the lipophilicity and introducing Efavirenz is a very potent inhibitor against a wide range
polar water solubilizing groups. Because delavirdine also of mutant HIV-1 RTs. Its activity against some of these are
underwent hydroxylation at C-6 of the pyridine ring, this presented in Table 8. In cell culture efavirenz selects for a
position was targeted for blocking. This was done by syn- double mutation (L100I plus L103A) following 10 serial
thesizing compounds containing a halogen at C-6 and one passages at increasing concentrations (Young et al., 1995a).
compound containing a pyridazine ring where nitrogen However, no single RT substitution has yielded a mutant

Antiviral Chemistry & Chemotherapy 10:6 289


OS Pedersen & EB Pedersen

Table 6. Antiviral activities of selected compounds against resistant viruses

HN
CH3SO2HN 1. R = Et, Y = t-Bu and X = H

2. R = n-Pr, Y = t-Bu and X = H

N N N X 3. R = Et, Y = Et and X = H
N
H R 4. R = Et, Y = i-Pr and X = F
O
5. R = Et, Y = t-Bu and X = F

Y
H3C
N HN
H
N 6. Z = CO, Y = i-Pr and X = H
N
Z 7. Z = CO, Y = t-Bu and X = H
N N N X
8. Z = SO2, Y = t-Bu and X = H
N Et
H 9. Z = CO, Y = t-Bu and X = F
O
10. Z = SO2, Y = t-Bu and X = F

EC90 (µM)*
Compound HIV-1MF (P236L)† HIV-1IIIB (L100I, M230L)† HIV-1IIIB (Y181C)‡
Delavirdine mesylate >10 >10 5.2
1 0.19 0.08 0.03
2 0.32 0.11 0.03
3 0.31 0.08 0.18
4 0.3–1.0 0.11 0.14
5 0.22 0.12 ≤0.03
6 0.24 0.11 0.05–0.1
7 0.16 0.16 ~0.05–0.10
8 0.34 0.14 <0.03
9 0.16 0.14 0.13
10 0.29 0.29 0.15
*EC90, concentration of drug that inhibited p24 production in the antiviral assays by 90%.
†Delavirdine was used for the selection of BHAP-resistant MF and IIIB HIV-1 variants.
‡L-697,661 was used for the selection of the resistant IIIB HIV-1 variant.
Data from Romero et al. (1996).

Figure 3. The structure of 1-[(2-hydroxyethoxy)methyl]- virus for which the EC95 for inhibition by efavirenz has
6-(phenylthio)thymine (HEPT) and the benzyl been >1.5 µM. Most mutants are inhibited by efavirenz at
analogue MKC-442 EC95 values of 50 nM or less (Table 8). The lowest con-
centration of efavirenz that has yielded evidence of cyto-
O toxicity, both in primary cells and in T cells, is 80 µM,
O
giving a selectivity index of approximately 80000 (Young et
5
HN 3 HN al., 1995a).
1
O 6
N S O N HEPT derivatives
1-[(2-Hydroxyethoxy)methyl]-6-(phenylthio)thymine,
O O HEPT (Figure 3), is the lead compound of this class of
HO NNRTI and was described in 1989 (Miyasaka et al., 1989).
HEPT was synthesized as an acyclonucleoside and was
expected to be a member of the nucleoside class of RT
HEPT MKC-442 inhibitors, which act as competitive inhibitors by mimick-
ing the natural substrate for the enzyme (Tantillo et al.,

290 ©1999 International Medical Press


The NNRTI boom

Table 7. The activity of selected benzoxazinones against HIV-1 RT and against a strain of HIV-1 with a double
mutation in RT

X R

Cl
O

N O
H

Configuration X R IC50 (nM)* WT IC50 (nM)† A17 RT ClC95 (nM)‡


(–) Efavirenz CF3 2 85 6

(–) CF3 8.6 69 12

(+/–) CF3 25 480 6

NC

(+/–) CClF2 12 350 ND§

CH
3
(+/–) CF3 N
CH
43 1950 100
3

*Inhibitor concentration to give 50% inhibition[(poly)rC•(oligo)dG] in wild-type RT.


†Inhibitor concentration to give 50% inhibition[(poly)rC•(oligo)dG] in A17RT, which is a mutant with double mutation: K103N and Y181C.
‡Concentration of inhibitor that reduced the spread of infection by at least 95% in MT-4 cells. HIV-1 p24 accumulation was directly
correlated with virus spread.
§ND, not determined.
Data from Young et al. (1995a).

1994). However, in antiviral testing it was found only to Phase III clinical trials. The importance of the substitution
inhibit HIV-1 and not HIV-2. From this it was deduced pattern of the thiophenyl ring in the 6 position and the sub-
that HEPT was different in its mechanism of inhibition of stituent at the 5 position were among the first characteris-
HIV than the known nucleoside inhibitors zidovudine, tics to be studied (Tanaka et al., 1992a). Substituting the
stavudine and didanosine. methyl group in the 5 position of HEPT with ethyl or iso-
Studies of the structure–activity relationship of HEPT propyl gave a marked increase in potency (Tanaka et al.,
analogues have led to the synthesis of MKC-442 (I-EBU; 1992a), whereas substitution with a hydrogen atom resulted
Triangle Pharmaceuticals) (Figure 3), which has entered in loss of activity. Also, the substitution pattern of the 6-

Table 8. Inhibition of wild-type and mutant HIV-1 infection in cell culture by efavirenz*

EC95 (nM)‡ EC95 (nM)‡


Virus or amino Virus or amino
acid substitution† Efavirenz L-697,661 acid substitution† Efavirenz L-697,661
Wild-type IIIB 1.5 100 V108I 3 400
Wild-type MN 3 50 V179D 3 400
Wild-type RFII 3 50 Y181C 6 >3000.0
A98G 12 800 Y188L 1500 >3000.0
L100I 100 200 K101D+K103N 1500 >3000.0
K101E 25 800 K101D+L100I 1500 >3000.0
K103N 100 800 K103N+Y181C 400 >3000.0
V106A 12 100 L100I+K103N§ 25000 1500

*Comparative values are presented for efavirenz and the unrelated pyridinone L-697,661.
†Each mutant virus expressed the noted amino acid substitution at the indicated RT residue.
‡The EC95 was defined as the concentration of test compound that inhibited virus expression by at least 95% relative to virus expression in
untreated control cultures. Assays were performed in MT-4 human T-lymphoid cells.
§This variant was selected by passage of the HIV-1 IIIB strain in MT-4 cells in increasing concentrations of efavirenz.
Data from Young et al. (1995b).

Antiviral Chemistry & Chemotherapy 10:6 291


OS Pedersen & EB Pedersen

Table 9. Inhibition of HIV-1 replication in MT-4 cells by HEPT derivatives

R1
HN

O N R2

R3

Compound R1 R2 R3 EC50 (µM) CC50 (µM) SI


HEPT Me SPh HOCH2CH2OCH2 7.0* 740 106
E-HEPU-dM Et SPh(3,5-di-Me) HOCH2CH2OCH2 0.013* 149 11500
I-HEPU-dM i-Pr SPh(3,5-di-Me) HOCH2CH2OCH2 0.0027* 128 47400
E-HEBU-dM Et CH2Ph(3,5-di-Me) HOCH2CH2OCH2 0.013† 281 22000
I-HEBU-dM i-Pr CH2Ph(3,5-di-Me) HOCH2CH2OCH2 0.0027† 221 82000
E-BPU Et SPh PhCH2OCH2 0.0059‡ 34 5800
E-EBU-dM Et CH2Ph(3,5-di-Me) EtOCH2 0.0016† 207 130000
E-EBU Et CH2Ph EtOCH2 0.041† 245 6000
I-EBU-dM i-Pr CH2Ph(3,5-di-Me) EtOCH2 0.0006† 43 72000
MKC-442 i-Pr CH2Ph EtOCH2 0.0042† 186 44000
1 Et CH2Ph(3,5-di-Me) EtSCH2 0.004§ 68 17000
2 i-Pr CH2Ph EtSCH2 0.006§ 37 6200
3 Et CH2Ph MeSCH2 0.002§ 32 16000
4 i-Pr SePh(3-Me) EtOCH2 0.0081¶ 53 6540
5 i-Pr SePh(3,5-di-Me) EtOCH2 0.0047¶ >200 42600

Values may not be directly comparable due to differences in assay conditions.


Data from: *Tanaka et al. (1992a); †Tanaka et al. (1995); ‡Tanaka et al. (1991), two separate experiments.
The values was determined by the MTT method; §Danel et al. (1996).
EC50 was determined by the p24 antigen method and the MTT method was used for CC50.
The values are expressed as the mean of three independent determinations; ¶Kim et al. (1996).
The values were determined in CEM-SS cells and are the mean of two independent determinations in duplicate.

(phenylthio) moiety was investigated and substitution at the functionalities that are introduced into the N-1 linker
2 or 4 position was ineffective, whereas substitution at the 3 between the pyrimidyl moiety and different aromatic rings,
position with methyl, ethyl or a fluoro substituent increased for example, phenyl, 2-furyl, 2-thienyl, 2-benzofuranyl and
the potency. Introduction of a methyl group or a chlorine at so on (Pontikis et al., 1997). Some 1-substituted (ethyl-
both the 3 and 5 position was even more effective (Tanaka thio)methyl and (methylthio)methyl analogues have shown
et al., 1992a). Removal of the hydroxyl group in the (2- anti-HIV-1 activity comparable to MKC-442 and although
hydroxyethoxy)methyl side chain also improved the poten- they have higher cytotoxicity, they still have high selectivity
cy (Tanaka et al., 1992b). Changing the phenylthio moiety indices (6200 to 17000) (Danel et al., 1996). Derivatives
with benzyl analogues yielded even more potent inhibitors with a selenyl in place of sulphur in the (phenylthio) moiety
of HIV-1 with selectivity indices of up to 130000 (Tanaka have also been prepared (Kim et al., 1996) and some with
et al., 1995). After studies on pharmacokinetics and follow- activities and selectivities that are comparable to those
ing toxicology tests, MKC-442 was selected as the candi- obtained for MKC-442.
date for clinical trials for AIDS chemotherapy (Tanaka et Some HEPT derivatives are highly bound to human
al., 1995). The oral bioavailability of MKC-442 in rats was serum proteins and have been tested for antiviral activity in
18.4% and the 50% lethal dose in rats was >2000 mg/kg the presence of varying percentages of human serum (HS)
(Tanaka et al., 1995). After molecular modelling studies on in place of foetal bovine serum (FBS) (Baba et al., 1993).
HEPT derivatives (Hopkins et al., 1996), an analogue of MKC-442 (I-EBU) showed EC50 values of 0.014 µM (in
MKC-442 with naphtyl in place of phenyl was synthesized, 10% FBS), 0.018 µM (in 10% HS), 0.026 µM (in 30% HS)
but it was 10-fold less active than MKC-442 and more toxic and 0.063 µM (in 50% HS). Some 2-thio analogues of
(Danel et al., 1997). Many derivatives with variations in the HEPT have also been prepared and although the anti-HIV
N-1 substituent have been synthesized. Among these are activity has been good, they all bound with high affinity to
some with more bulky groups, such as E-BPU (Tanaka et HS proteins, some with only 0.3 to 0.7% of the total com-
al., 1991) (Table 9). Some have amines, amides and alkene pound remaining unbound to proteins (Baba et al., 1993).

292 ©1999 International Medical Press


The NNRTI boom

Figure 4. The basic structure of dihydroalkoxybenzyloxopyrimidines (DABOs) and


dihydrothioalkylbenzyloxopyrimidines (S-DABOs)

O O
5′ 5′
R1 R1
HN 3 4 5 4′
HN 3 4 5 4′
2 6 2
R2 1 3′ 6
R2 1 3′
O N 2′ S N 2′
DABOs S-DABOs

R1 : H, Me, Et or i-Pr

R2 : alkyl or cycloalkyl

Dihydroalkoxybenzyloxopyrimidines and thio best activities are achieved with bulky substituents for R2
analogues (Figure 4), such as cyclohexyl, cyclopentyl or sec-butyl
The dihydroalkoxybenzyloxopyrimidines (DABOs) and (Artico et al., 1993). A methyl substituent in the 3′ position
their thio analogues (S-DABOs) are closely related to the increases the activity, whereas an additional methyl sub-
HEPT derivatives. DABO derivatives are characterized by stituent in the 5′ position does not always increase the
an alkoxy substituent, the S-DABOs by a thioalkyl group, potency. A compound with methyl in the 5 position, 3′,5′-
at C-2 in place of the N-1 substituent in the HEPT deriv- dimethyl substituted in the phenyl moiety and a sec-butyl
atives (Figure 4). in the alkoxy group at C-2 had a selectivity of >416, and
Some 6-benzylpyrimidines were synthesized as dihy- has been one of the most active among the first DABO
drofolate reductase inhibitors and because of their structur- compounds (Massa et al., 1995).
al similarities with HEPT they were tested, and some were Replacing the side chain oxygen in the 2 position with
found to be active, against HIV-1 (Botta et al., 1992). sulphur led to the S-DABO compounds, which showed
Unlike the HEPT derivatives, the DABOs are active with increased anti-HIV-1 activity (Mai et al., 1995). Alkylation
hydrogen in the 5 position comparable with compounds at N-3 and replacing C=O with C=S in the 4 position of the
having a methyl group in the 5 position (Artico et al., pyrimidine ring led to compounds devoid of activity, where-
1993). Substituting the 5 position with an ethyl or iso- as substituting the 6-benzyl group with 1-naphtylmethyl
propyl does not increase the anti-HIV activity, however the enhanced the activity of the S-DABO compounds (Mai et
size of the C-2 alkoxy group seems to be important. The al., 1997). Some highly active anti-HIV-1 compounds with

Table 10. Anti-HIV-1 activities of selected compounds of the S-DABO class


O

R1
HN

R3 R2
S N

R1 R2 R3 IC50 (µM)* ED50 (µM)† CD50 (µM)‡ SI§


Et phenyl allyl ND 1.5¶ >100 >67
H 1-naphtyl sec-butyl ND 0.33** >300 >909
Me phenyl MeSCH2 28.8±10.0†† 3.4±1.1 >100 32±10
Et phenyl MeSCH2 12.0±2.3†† 0.8±0.2 >100 146±38
i-Pr phenyl MeSCH2 5.6±2.4†† <0.001±0.000 >100 100000±0
H 2,6-di-F-phenyl sec-butyl 0.05‡‡ 0.04 >200 5000
Me 2,6-di-F-phenyl i-propyl 0.05‡‡ 0.05 >200 4000

*Concentration required to inhibit recombinant RT by 50%.


†Concentration to prevent the spread of HIV-1 in cell culture by 50%.
‡Concentration of compound that prevented proliferation of host cells by 50%.
§Selectivity index, CD50/ED50.
¶Danel et al. (1998).
**Mai et al. (1997).
††Sudbeck et al. (1998), (rA.dT template:primer).
‡‡Mai et al. (1999), (rC.dG template:primer).

Antiviral Chemistry & Chemotherapy 10:6 293


OS Pedersen & EB Pedersen

Figure 5. The structure of 2,3-dihydro-7H-thiazolo Figure 6. The structure of AG 1549, 5-(3,5-dichloro-


[3,2-a]pyrimidin-7-ones, which can be considered a phenylthio)-4-isopropyl-1-(4-pyridyl)methyl-1H-
hybrid between S-DABOs and HEPT imidazol-2-yl-methyl carbamate

O Cl
N
2 R1 Cl
N c R1: Et, i-Pr
6
b
a 5 R2 : Ph, 3,5-Me2C6H3, 1-Naphtyl
R2
N4 O
S R3 : Me, Et, CH2CH2OH N
S
1
O
OR3 N NH2

a (methylthiomethyl)thio substituent at C-2 of the pyrimi-


dine ring (Table 10) have been designed based on the struc- selected from a set of derivatives of an initial lead com-
ture of the NNRTI binding pocket of RT (Sudbeck et al., pound (Fujiwara et al., 1998). AG 1549 is a highly substi-
1998). Molecular modelling supporting a hypothesis of tuted imidazole (Figure 6), and had an IC50 value of 0.45
beneficial π-stacking interaction between Tyr-188 of the µM in a standard RT assay with poly(rA) and oligo(dT).
NNRTI binding pocket in RT and the DABOs with 2,6- Some of the beneficial characteristics for AG 1549 are a
dihalogenation in the benzyl group, has led to highly potent higher concentration in lymph nodes than in plasma after
inhibitors of HIV-1 (Mai et al., 1999). Some activities of S- oral administration to rats and an in vitro selectivity index
DABO compounds are presented in Table 10. of 8000 (Fujiwara et al., 1998). The activities of AG 1549,
Bicyclic derivatives that can be considered as hybrids nevirapine, delavirdine and loviride were compared against
between S-DABOs and HEPT analogues have also been HIV-1 mutant clones (Table 11).
synthesized (Danel et al., 1998) (Figure 5). Mutant clones with single amino acid substitutions at
These compounds showed only moderate anti-HIV residues 100, 103, 106, 181, 188, 190, 227 and 236 showed
activity with the lowest EC50 value of 0.7 µM for the less than 10-fold reduced sensitivities compared to the
compound with R1=ethyl, R2=3,5-Me2C6H3CH2 and wild-type strain, whereas the clone with mutation L234I
R3=ethyl. Some analogues with the thiazole ring fused to was 22-fold less sensitive to AG 1549 (Fujiwara et al.,
the C-2 and N-3 bond of the S-DABO pyrimidine ring 1998) and was more sensitive to nevirapine and loviride
was also synthesized, but these were either too toxic to the than the wild-type HIV strain. The double mutant strain
host cells or devoid of antiviral activity (Danel et al., with the V106A plus Y181C mutation was still sensitive to
1998). AG 1549, whereas the double mutant strain V106A plus
F227L was only somewhat sensitive.
Imidazoles AG 1549 was tested for in vitro selection of mutant
In a screening programme seeking NNRTIs that would strains of HIV-1 and two clones were found and sequenced.
inhibit HIV-1 strains that are resistant to known NNRTIs In both case more than one amino acid substitution muta-
or to zidovudine, AG 1549 (formerly S1153; Agouron tion was present. One strain contained the mutations V106A
Pharmaceuticals and Shionogi Research Laboratories) was plus F227L and the EC50 value for the clone was 740 ng/ml

Figure 7. The structures of some TIBO derivatives

S S
S
HN HN
HN
N N
N
H H
H
CH3 CH3
CH3 N N
N Cl

Cl
TIBO R82150 TIBO R82913 TIBO R86183
(9-Cl-TIBO) (8-Cl-TIBO)

294 ©1999 International Medical Press


The NNRTI boom

Table 11. Sensitivities of molecular clones of HIV with RT gene mutations to AG 1549 (S1153) and other
anti-HIV-agents

EC50 (µM)*
Virus mutation AG 1549 Nevirapine Delavirdine Loviride
Wild-type (strain NL342) 0.0069 0.025 0.011 0.018
L100I 0.0021 0.052 >0.905 0.015
K103N 0.0069 1.13 0.832 0.399
Y106A 0.0031 >1.88 0.271 0.285
Y181C 0.0093 >1.88 >0.905 >1.42
Y188C 0.0010 >1.88 0.058 0.655
G190A 0.0075 >1.88 0.0024 0.6712
F227C 0.0053 0.33 0.017 0.16
F227L 0.0010 0.11 0.0014 0.065
L234I 0.015 0.0013 0.024 0.0013
P236L 0.0024 0.079 >0.905 0.0080
V106A+Y181C 0.0055 >1.88 >0.905 >1.42
V106A+F227L 0.266 >1.88 0.067 >1.42

*Each value represents the mean of at least three experiments, each of which was performed in duplicate.
Data from Fujiwara et al. (1998).

(1.6 µM). Another clone containing K103T, V106A plus found that a 3-methyl-2-butenyl substituent on N-6
L234I had an EC50 value of 37 ng/ml (0.08 µM) in cell cul- (Kukla et al., 1991a), a methyl substituent at position 5 and
ture using the MTT assay (Fujiwara et al., 1998). a thione in position 2, gave a very potent inhibitor of HIV-
Many NNRTIs bind to serum proteins, and AG 1549 1, particularly if the 5 position had the S-configuration
has the greatest affinity for human albumin. In the presence (R82150) (Pauwels et al., 1990) (Figure 8). Additionally, a
of 50 mg/ml of HS albumin and 1 mg/ml alpha-1-acid gly- chloro substituent in position 9 gave a compound
coprotein, AG 1549 showed a ninefold reduction in its activ- (R82913) that had a 10-fold increase in activity, but also an
ity compared with the activity without albumin. Taking this increase in cytotoxicity (Pauwels et al., 1990). Changing
into account, AG 1549 may still have a sixfold greater antivi- the chloro substituent from position 9 to position 8 afford-
ral activity than nevirapine in human plasma (Fujiwara et al., ed a compound (R86183) even more potent (Ho et al.,
1998). AG 1549 is now undergoing clinical testing. 1995). Compound R82913 has been subject to clinical
Phase I testing.
TIBO In the imidazole part of the structure it has been found
Since the TIBO class ( Janssen Research Foundation) was that replacement of the carbonyl oxygen with sulphur or
introduced as non-nucleoside inhibitors of HIV in vitro selenium causes an increase in the activity. A number of
(Pauwels et al., 1990), a lot of work concerning struc- variations in this position have been tested, including S-
ture–activity relationships has been done using this class of phenylthiourea and O-methylated urea, and all have been
compounds (Figure 7). inactive. The proton on nitrogen in position 1 appeared to
In the early work of optimizing the TIBO class, it was be necessary, especially for the urea compounds, and this
group may be involved in direct hydrogen bonding to the
Figure 8. General structure of TIBO derivatives enzyme. Replacing the nitrogen in position 3 with a carbon
resulted in loss of activity. Most changes in the imidazole
X structure resulted in less active compounds or a total loss of
X = O, S or Se activity (Kukla et al., 1991b).
HN 2
Substitutions on the diazepine ring have also been inves-
N 4
tigated (Breslin et al., 1995). The 5-mono-methyl-substi-
CH3 tuted analogues, which were the original substitutions in the
9
8
N early lead compounds, and 7-mono-methyl-substituted
7
analogues were comparable, being the most active in the
series. The 4,5,7-unsubstituted analogue and the 4-mono-
methyl-substituted analogues were less active. Substituting
larger and more bulky groups such as isopropyl and phenyl

Antiviral Chemistry & Chemotherapy 10:6 295


OS Pedersen & EB Pedersen

Table 12. Anti-HIV activities of some TIBO derivatives

HN
N

R1
N
R3 R2

Compound X R1 R2 R3 EC50(µM)* CC50(µM)† SI‡


R78305 O (+)-(S)CH3 allyl H 70§ 674 10
R82150 S (+)-(S)CH3 3,3-dimethylallyl H 0.028§ >870 >31071
R82913 S (+)-(S)CH3 3,3-dimethylallyl 9-Cl 0.0015§ 31 20667
1 S (±)-CH3 3,3-dimethylallyl H 0.097¶ ND** ND
2 Se (+)-(S)CH3 3,3-dimethylallyl 9-Cl 0.018¶ ND ND
3 S (+)-(S)CH3 3,3-dimethylallyl 8-SCH3 0.0050†† ND ND
4 S (+)-(S)CH3 3,3-dimethylallyl 8-F 0.0058†† ND ND
5 S (+)-(S)CH3 3,3-dimethylallyl 9-F 0.0250†† ND ND
R86775 S (+)-(S)CH3 3,3-dimethylallyl 8-Br 0.003‡‡ 53 18000
R84674 S (+)-(S)CH3 3,3-dimethylallyl 8-CH3 0.0014‡‡ 80 5700

*EC50 is the 50% inhibitory concentration for cytopathicity by HIV-1 in MT-4 cells.
†CC50 is the 50% cytotoxic dose in mock-infected MT-4 cells.
‡The ratio CC50/EC50 is the selectivity index (SI).
§Data from Pauwels et al. (1990).
¶Data from Kukla et al. (1991b).
**ND, not determined.
††Data from Ho et al. (1995).
‡‡Data from Pauwels et al. (1994).

produced no advantage except for compounds substituted (Ahgren et al., 1995; Cantrell et al., 1996). The lead com-
with isopropyl in position 4 and with an oxo group in posi- pound of this series was LY73497 (Figure 9). Optimization
tion 2, which were more active than the 4-methyl ana- of this lead gave N-[2-(2-pyridyl)ethyl]-N-[2-(5-bro-
logues. Some (4 plus 5, 4 plus 7, 5 plus 6, 5 plus 7, 6 plus 7 mopyridyl)]thiourea hydrochloride (LY300046:HCl) (tro-
and 7 plus 8) disubstituted or ring fused analogues were also virdine; Medivir), which has been selected for clinical trials.
prepared (Breslin et al., 1995). The 5,7-di-Me (trans) ana- Extensive structure–activity relationship studies of the
logue was slightly better than the 5-mono substituted ana- PETT compounds have been made. For this purpose the
logue, being the most promising with an EC50 value of structure of PETT is considered as a product of four parts
0.042 µM in MT-4 cells. From this, the R,R (trans) and S,S (Figure 10).
(trans) 5,7-dimethyl analogues were prepared for the 2-oxo In part 1 of the structure, the phenethyl moiety, the sub-
and 2-thio analogues. These compounds have a different stitution patterns have been investigated. Meta and partic-
structure–activity relationship to the 5-methyl analogues. ularly ortho substitution is preferred over para substitution
The 2-oxo compounds were slightly more active than the (Bell et al., 1995). Both mono substitution (Vig et al.,
thio analogues and a chloro substituent at the 9 position 1998), di and tri substitution (Bell et al., 1995) have given
resulted in a significant loss in activity. very active compounds. The characteristics of the sub-
Substitution in the aromatic part of the TIBO structure stituents have also been investigated (Bell et al., 1995).
has also been investigated (Ho et al., 1995). Substituents in Both electron donating and electron withdrawing of small
the 8 position gave a large improvement in activity compared groups like fluoro, chloro, azido and methoxy substituents
with the parent compound, whereas substituents at the 9 were comparable with good activity. The chain length of an
position tended to have little effect on activity and 10 sub- alkoxy substituent was investigated, and the ethoxy sub-
stituents decreased the activity (Ho et al., 1995) (Table 12). stituent gave the maximum activity, whereas the more ster-
ically demanding propoxy and isopropoxy substituents
PETT decreased the activity. Compounds where the phenyl group
The phenethylthiazolylthiourea (PETT) compounds were was replaced with 2-pyridyl, as in trovirdine, gave the most
derived from a systematic disassemblage of the molecular active compounds in a series with different heterocycles.
structure of the known TIBO HIV-1 RT inhibitors Active compounds have also been prepared with saturated

296 ©1999 International Medical Press


The NNRTI boom

Figure 9. The structure of trovirdine and the lead Figure 10. Phenethylthiazolethiourea (PETT),
compound LY73497 separated into four parts for discussion of
structure–activity
S N

S S N
N N
H H
S
N N
LY 73497 H H

Br
S 1 2 3 4

N N N N
H H
.HCl pounds (Figure 11).
LY 300046 HCl The activity of new NNRTIs against strains of HIV
(trovirdine) resistant to known NNRTIs is becoming more important,
and the PETT compounds show good activity against
Trovirdine has an ED50 in HIV-1-infected MT-4 cells of 0.020 µM, and
some of the known resistant strains of HIV. Some of the
an IC50 value against wild-type HIV-1 RT of 0.015 µM and IC50 values
against mutant HIV-1 RTs of 0.43 µM (Ile-100) and 2.50 µM (Cys-181) PETT derivatives showing good activity against single
(Cantrell et al., 1996). mutations (Ile-100 and Cys-181) were tested in assays
against two double mutant HIV-1 strains. Five of the test-
heterocycles such as 1-piperidinyl and 1-piperazinyl (Mao ed compounds showed ED50 values below 10 µM against
et al., 1998). one of the two double mutants (Ile-100 plus Asn-103 or
In part 2 of the molecule the ethyl linker was optimal for Ile-100 plus Cys-181) (Cantrell et al., 1996). These five
activity. Methyl substitution in the benzylic position compounds are listed in Table 13.
enhanced activity, whereas a methyl in the phenylethyl The PETT compound trovirdine has shown to readily
position diminished activity (Bell et al., 1995). A cyclo- penetrate the blood–brain barrier, as the concentration in
propyl linker with cis configuration, has been shown to be brain tissue parallelled those in plasma of male Fischer 344
an advantage for the urea-PETT analogues (Sahlberg et rats (Ahgren et al., 1995). The in vitro protein binding of
al., 1998). trovirdine was 88.7% in rat plasma and 95.5% in human
For part 3 of the molecule the N,N-unsubstituted-
thiourea was most active (Bell et al., 1995). Methyl substi-
tution on nitrogen adjacent to the phenethyl side chain Figure 11. The structure of PETT derivatives and
trovirdine with the proposed internal hydrogen
completely eliminated activity, whereas methyl substitution bond
on the nitrogen adjacent to the thiazole ring only decreased
the activity slightly. The reason for the difference in activi- S
ty for N-methyl substitution may be owing to an internal
(H,Me) R
hydrogen bond (Figure 11). N N
Compounds with a urea part in place of the thiourea
H
have also been prepared. They give lower activity but may S N
possess better pharmacological properties than the thiourea
compounds (Sahlberg et al., 1998). PETT derivatives
In part 4, the thiazole moiety, the most optimum com-
pounds have been achieved by replacing the thiazole with a
5-bromopyrid-2-yl. In the thiazole series several 4-substi- S
tuted derivatives, including small alkyl, cyano, trifluo-
romethyl and ethoxycarbonyl substituted compounds were HN N N
quite potent inhibitors (Bell et al., 1995). In a series where
H
the thiazole was replaced with diazinyl it seemed that the N
nitrogen in the 2 position was essential because the 4-pyri-
dazinyl derivate was devoid of activity (Heinisch et al.,
1997b). This seems to be in accordance with the proposed Br
internal hydrogen bond in anti-HIV active PETT com- Trovirdine

Antiviral Chemistry & Chemotherapy 10:6 297


OS Pedersen & EB Pedersen

Table 13. Five compounds tested with activity against double mutant HIV-1 RT in MT-4 cells

R3

R4 R2 R1
S

N N N
H H
R5

ED50 (µM)*
R1 R2 R3 R4 R5 WT† Ile-100 Cys-181 Ile-100/Asn-103 Ile-100/Cys-181
Cl F MeO H MeO 0.002 0.009 0.003 1.0 1.0
Br F H H EtO 0.013 0.09 0.007 1.6 1.0
Br F CN Me2N H 0.013 0.15 0.020 5.5 7.5
Br F EtO H F 0.006 0.10 0.006 10 1.2
Br CN EtO H F 0.004 0.04 0.005 >10 3.0

*Concentration of inhibitor that inhibited the infection of HIV-1IIIB in MT-4 cells, assayed with the XTT method after 5 or 6 days.
†Wild-type HIV-1 RT.
Data from Cantrell et al. (1996).

Table 14. Inhibition of HIV-1 by urea-PETT compounds

R1 R4
R2
O
R2 R4
O R1
N N N
H H
N N N
H H R3
R3

1–6 7–13

ED50 (µM)*
Compound R1 R2 R3 R4 WT WT† Clone 118‡ Clone 90§
1 H F F Cl 0.03 0.85 32 >32
2 NMe2 F F Br 0.5 NT¶ >25 >25
3 OMe F OMe Cl 0.011 0.09 17 3
4 OEt F OMe Cl 0.13 NT 14 >27
5 COMe F OMe Cl 0.2 NT >27 >27
6 OEt Cl F Br 0.07 NT 15 8
7 H F F Cl 0.01 0.01 NT NT
8 OEt Cl F Cl 0.016 0.06 0.1 NT
9 OEt Cl F CN 0.01 0.02 0.27 0.53
10 OEt F F Cl 0.01 0.03 0.75 0.27
11 OMe F OMe Cl 0.012 0.1 1.6 2.7
12 OEt F Cl Cl 0.01 0.1 0.1 0.1
13 OEt F OMe Br 0.025 0.4 NT NT
Trovirdine 0.02 5 0.8 >5
9-Chloro-TIBO 0.25 NT >22 124
Nevirapine 0.15 0.12 0.62 22
L-697,661 0.065 NT 0.85 >11

*Concentration of compound that reduced the CPE of HIV-1IIIB in MT-4 cells by 50% in an XTT assay.
†Tested in the presence of 15% human AB serum.
‡Clone 118 contains a L100I mutation.
§Clone 90 contains a Y181C mutation.
¶NT, Not tested.
Data from Sahlberg et al. (1998).

298 ©1999 International Medical Press


The NNRTI boom

Table 15. The anti-HIV activity and inhibition of HIV-1 Table 16. Pyrimidine thioethers inhibition against
RT of selected alkenyldiarylmethanes (ADAM) HIV-1 wild-type and a delavirdine resistant strain
compared to delavirdine
H3COOC COOCH3 Cl

H3CO OCH3
N

X X H2N N S R

R EC90 (µM)*
Compound R HIV-1IIIB HIV-1MF†
R X IC50* EC50† CC50‡ TI§ 1 2-naphtyl 0.22 2.76
(CH2)4CH3 Br 0.38 9.2 138 15 2 CH=CHphenyl 0.15 5.05
CH2CH2N3 Br 94 1.1 >316 >278 3 CH=CHCONMe2 0.09 0.74
CH2CH2N3 Cl 2.0 0.27 41.8 155 4 CH=CHCONEt2 0.02 0.11
(CH2)3COOCH3 Cl 0.3 0.013 31.6 2430 5 3-methylphenyl 0.11 1.76
6 3-methoxyphenyl 0.27 3.21
*Inhibitory activity (µM) versus HIV-1 RT with rC.dG as the template 7 3-bromophenyl 0.35 4.52
primer. Delavirdine‡ 0.03 53.58
†50% inhibitory concentration (µM) for CPE of HIV-1RF in CEM-SS
cells.
‡Cytotoxic concentration (µM) for mock-infected CEM cells. *Concentration of drug that inhibited p24 production by 90% in
§Therapeutic index, CC50 divided by EC50. infected MT-4 cells.
Data from Cushman et al. (1998a). †Laboratory derived delavirdine-resistant variant of HIV-1.
‡Reference compound.
Data from Nugent et al. (1998).
plasma at a concentration of 2 µg/ml. Because of its simple
chemical synthesis, excellent antiviral activity, satisfactory potent compound of this class, with an EC50 value of 13
pharmacokinetic profile and acceptable toxicity, trovirdine nM and a selectivity index of 2430 (Cushman et al.,
entered Phase I clinical trials (Ahgren et al., 1995), but was 1998a,b).
later withdrawn. Bromo- and chloro substituents are preferred over iodo-
and unsubstituted derivatives (Table 15). The length of the
Urea-PETT alkenyl substituent is also important and it appears that a
The urea-PETT compounds may have better toxicological six atom chain is optimal for anti-HIV-1 activity
and phamacokinetic properties than the PETT com- (Cushman et al., 1998a).
pounds. It was found that the urea-PETT compounds
maintain their antiviral activity in cell culture in the pres- Pyrimidine thioethers
ence of added HS much better than the thiourea com- Pyrimidine thioethers (Pharmacia & Upjohn) as inhibitors
pounds (Sahlberg et al., 1998). This point urged the of HIV-1 RT were first reported by Althaus et al. (1996).
researchers at Medivir AB to make a structure–activity This class of RT inhibitors is primarily characterised by its
research (SAR) programme in which different urea-PETT potent activity against the P236L mutant, which renders
analogues were synthesized and evaluated (Table 14). This HIV-1 resistant to delavirdine (Nugent et al., 1998). The
gave ethyl linked (1–6) and conformational restricted lead compound of this class was 6-chloro-2-benzylthio-4-
cyclopropyl analogues (7–13). The cyclopropyl compounds pyrimidinamine, and several derivatives were investigated
were in general more potent than the ethyl linked com- by Nugent et al. (1998). Exchange of the chloro moiety in
pounds, especially against mutant HIV strains. the pyrimidine ring with trifluoromethyl gave a compound
only a little less potent, whereas exchange with other elec-
Alkenyldiarylmethanes tron-withdrawing groups was not so good. In the place of
Alkenyldiarylmethanes (ADAMs) were introduced as a R (Table 16) the enzyme can accommodate a large, but flat,
new class of NNRTIs in 1995 (Cushman et al., 1995, hydrophobic group. This could be an aryl group or an
1996). This afforded the first lead compound 3′,3′′-dibromo- alkenyl group. A secondary amide attached to an alkenyl
4′,4′′-dimethoxy-5′,5′′-bis(methoxycarbonyl)-1,1-diphenyl- has significantly less activity compared to a tertiary amide.
1-heptene, with an EC50 value of 9.2 µM and a selectivity This is thought to be caused by a hydrogen bonding pro-
index of 15. Further work has lead to methyl 3′,3′′- ton, or for steric reasons, in the lipophilic pocket of the
dichloro-4′,4′′-dimethoxy-5′,5′′-bis(methoxycarbonyl)-6,6- enzyme. Substitution of the sulphur with oxygen or nitro-
diphenylhexenoate (ADAM II) as the currently most gen decreased or removed the activity. Oxidation of the sul-

Antiviral Chemistry & Chemotherapy 10:6 299


OS Pedersen & EB Pedersen

Table 17. Antiviral activity of furo[2,3-c]pyridine pyrimidine thioethers against in vitro-selected NNRTI-resistant
HIV-1 variants

S N Cl
N

CH3 N

NH2

EC90 (µM)*
DLVR MF L-697,661R IIIB DLVR IIIB R88703R IIIB
Compound IIIB (WT) (P236L)† (Y181C)† (L100I)† (Y181C)†
(–)-(S)(PNU-142721) 0.001 0.008 1.1 0.07 0.17
(+)-(R) ND‡ >0.01 >3.0 ND ND
Racemate 0.002 0.01 2.4 0.06 0.17
Delavirdine 0.05 >10 5.2 >10 1.0
*EC90 concentration of drug that inhibited p24 antigen production by 90% in infected MT-4 cells.
†Primary resistance conferring mutation at the designated codon of HIV-1 RT.
‡ND, Not determined.
Data from Wishka et al. (1998a).

phur diminished the activity, and removal of the methylene c]pyridinethiopyrimidine class of compounds. These are
linker resulted in compounds devoid of activity. Table 16 characterized by the aralkyl group and a methyl substituent
shows some of the compounds with activity against wild- at the methylene linker. This methyl substitution introduces
type and the delavirdine-resistant HIV-1 (P236L) mutant a stereocentre at the carbon in the methylene linker (Table
(Nugent et al., 1998). 17). Both the (R) and (S) enantiomers proved to be
Further study within the pyrimidine thioethers for com-
pounds with broad activity against several NNRTI-resistant
Figure 12. Structure of other bicyclic TTD analogues
variants of HIV-1 have led to the furo[2,3-
O O

Table 18. Antiviral activity of some TTDs S R1


N
O O
2′
7 S X S N O
1 N
2 3′
6 S (a)
R2
5 4N O
O O
R
N S R1
N
Compound X R EC50 (µM)* CC50 (µM)† H 3C N
QM96625 H 2-Cl-benzyl 0.1 >119 N O
QM96521 H CH2CN 0.9 502.7
R2 (b)
QM96537 H CH2CCH 1.0 376
QM96539 Cl CH2CN 0.09 340 O O
Br CH2CN 0.09 68.6
QM96639 F CH2CN 0.05 93.6 S S R1
N
Cl
*Compound concentration required to achieve 50% protection of
MT-4 cells from HIV-1IIIB induced cytopathogenecity, as determined N O
by the MTT method.
†Compound concentration dosage required to reduce the viability of R2 (c)
mock-infected cells by 50%, as determined by the MTT method. All
data represent mean values of at least two separate experiments. (a) Thieno[2,3-e][1,2,4]thiadiazine,(b) 6-methylpyrazolo[4,3-
Data from Arranz et al. (1998). c][1,2,4]thiadiazine, (c) 6-chlorothieno[3,2-e][1,2,4]thiadiazine.

300 ©1999 International Medical Press


The NNRTI boom

Figure 13. Structure of carboxanilide UC84 with the istry of these thiopyrimidines. PNU-142721, the (S)-enan-
separation into four parts marked tiomer, and the racemate were found to inhibit the enzyme
with IC50 values in the submicromolar range, whereas the
O CH3 (R)-enantiomer was inactive. In cell culture PNU-142721
O CH3
was very potent against HIV-1 wild-type and several strains
H
N of HIV-1 resistant to known NNRTIs (Table 17).
S O CH3 Another compound in this series, PNU-109886, with a
O methyl substituent in the furo[2,3-c]pyridine moiety has
Cl
a b c d displayed increased potency against virus containing the
Y181C mutation. However, it is characterized by a less
favourable pharmacokinetic profile than PNU-142721
extremely potent inhibitors of wild-type HIV-1 RT with (Wishka et al., 1998b). PNU-142721 possess high antivi-
IC50 values (rA.dT) from 20 to 85 nM, and of the mutant ral activity against a broad spectrum of HIV-1 variants and
RT P236L that is resistant to delavirdine, with IC50 values a favourable pharmacokinetic profile, including penetra-
from 22 to 25 nM (Wishka et al., 1998a). Interestingly, tion of the blood–brain barrier in the rat. The levels of
activity against Y181C RT, which is observed to occur when PNU-142721 found in the brain in an experiment with
HIV-1 cultures are under pressure from many known rats was 75% of the simultaneous plasma concentrations
NNRTIs, was found to be dependent on the stereochem- (Wishka et al., 1998a).

Figure 14. Structure of some active derivatives of UC 84

O CH3
O CH3
N
H
S O CH3

O
Cl
a b c d
H
H3C N
UC 38 O
H3C
S
H3C H O
N CH3
1 O
H3C
S
H3C H CH3
N CH2 N O
2 O CH3
H3C
S
H3C H O CH3
N
3 O
H3C
S
H3C H O
N
O
4
H3C O CH3
S
H3C H O
N
O
5
H3C O
S
CH3 H
N O CH3

6 H3C
O O CH3
S
CH3 H CH3
N O

7 H3C
O O CH3
S
CH3 H O
N CH3
8 H3C
O O CH3
S
CH3

Antiviral Chemistry & Chemotherapy 10:6 301


OS Pedersen & EB Pedersen

Table 19. Antiviral activity of oxathiin carboxanilide derivatives against wild-type and mutant HIV-1 strains in
CEM cells

EC50 (µM)*
Compound IIIB L100I V106A G138K Y181C CC50 (µM)†
UC 84 0.042 ≥112 >56 ≥56 42 25
UC 38 0.03 2.2 2.2 1.3 2.0 40
1 0.01 2 2 0.3 2.0 19
2 0.03 0.67 1.4 0.2 1.1 >317
3 0.1 1.3 1.7 0.33 1.8 20
4 0.1 1.8 2.1 1.7 1.8 36
5 0.6 2.2 1.6 1.8 1.6 14
6 0.6 0.91 1.2 0.2 1.8 30
7 0.9 2.1 1.2 2 2.0 30
8 0.9 1.9 1.0 0.35 1.5 2.3
Nevirapine 0.03 0.1 8.6 8.6 0.37 >75
TIBO R82913 0.03 0.9 6.2 2 5.3 >62

*Compound concentration required to reduce HIV-1-induced giant cell formation in CEM cell cultures by 50%.
†Compound concentration required to reduce viability of the host cells by 50%.
Data from Balzarini et al. (1995).

TTD resulted in less active compounds. Systems that have been


1,1,3-Trioxo-2H,4H-thieno[3,4-e][1,2,4]thiadiazine (T- investigated are thieno[2,3-e], 6-methylpyrazolo[3,4-e]
TD) is a new class of NNRTI (Witvrouw et al., 1998). The and 6-chlorothieno[3,2-e] fused TTD analogues (Arranz
basic requirements for the structure of TTDs is N-4 alkyl et al., 1998) (Figure 12).
and N-2 benzyl groups (Table 18). Two compounds with a
2-picolyl in place of the N-2 benzyl were less active, where- Carboxanilides and thiocarboxanilides
as a 3-picolyl derivative and a 2-picolyl derivative with a Oxathiin carboxanilide, UC 84 (NSC 615985), was origi-
benzyl group at N-4 was more potent than the benzyl ana- nally synthesized as a potential fungicide by chemists at the
logues (Arranz et al., 1998). Halogens have been substitut- Uniroyal Chemical Company. In a screening programme
ed at the N-2 benzyl moiety, with optimum results by the National Cancer Institute, UC 84 exhibited anti-
achieved by one ortho- or metha substituent because a 3- HIV activity. UC 84 completely protected CEM-SS cells
fluorobenzyl derivative was more potent than the 3,5- infected with HIV-1 from the cytopathic effect of HIV
difluorobenzyl analogue (Witvrouw et al., 1998). The best with an EC50 value ranging from 0.1 µM to 1.0 µM,
results concerning the N-4 alkyl substituent have so far depending on the infectivity condition (Bader et al., 1991).
been with cyanomethyl, propargyl and benzyl. Substituents UC 84 served as a lead (Balzarini et al., 1995) and consists
of methyl, ethyl, propyl, allyl and cyanoethyl at N-4 pro- of an oxathiin moiety (part a), a carboxamide group (part b)
duce less active compounds (Arranz et al., 1998; Witvrouw and a 2-chlorobenzoic acid (part c) that is esterified with
et al., 1998). an isopropyl group (part d) (Figure 13).
Other bicyclic systems, where changes in the thiophene A series of derivatives was synthesized (Balzarini et al.,
ring of the thieno[3,4-e][1,2,4]thiadiazine have been tried, 1995) with the following changes: part a, the oxathiin part,

Figure 15. Structure and substituents of UC 10, UC 82, UC 781 and UC 040

R= N and Z = O (UC 10)


O

Cl
S

N R R= and Z = S (UC 82)


H O
and Z = O (UC 781)
Z
CH3

R= and Z = S (UC 040)


O

302 ©1999 International Medical Press


The NNRTI boom

Table 20. Anti-HIV activities of UC 10, UC 82, UC 781 thus be very interesting in a more protective manner as an
and UC 040 agent in retrovirucidal formulations.
Compound EC50 (µM)* CC50 (µM)† TI‡
TDA (thiadiazole derivatives)
UC 10 0.008 20 2500
The basic structure of the TDA derivatives is presented in
UC 82 0.008 16 2000
Table 22. A derivative (RD3-2356) with a 2-chloro sub-
UC 781 0.008 >500 62500
UC 040 0.016 10 625
stituent in the phenyl moiety enhanced the activity com-
pared with a derivative containing an unsubstituted phenyl
*Concentration required to inhibit the spread of HIV-1 by 50% moiety (RD3-2105) (Hanasaki et al., 1995). A 4-chloro
(XTT method), for paticular cell lines and virus isolates; see
Buckheit et al. (1997).
substituent has the opposite effect, producing compounds
†50% cytotoxic concentration. with decreased or lacking activity (Ijichi et al., 1996).
‡Therapeutic index. Investigating the R2 and R3 alkyl substituents revealed
Data from Buckheit et al. (1997).
that the most potent congener of the TDA derivatives is 4-
(2,6-dichlorophenyl)-1,2,5-thiadiazol-3-yl N-methyl-N-
was replaced by various alkoxy groups; part b was modified propylcarbamate (RD4-2024), whereas the
from C=O to C=S, which in most cases increased the activ- monoalkylcarbamate derivative, RD3-2380, was without
ity; part c was mostly unmodified; and part d was modified inhibitory effect (Ijichi et al., 1996). The easy synthesis of
with a variety of substituents. The eight most interesting TDA derivatives made them potential drug candidates for
compounds in this series, UC 38 being the most potent, are HIV-1 infection (Hanasaki et al., 1995).
presented in Figure 14, and test results against HIV wild- Sequence analysis of mutants resistant to TDA deriv-
type and four mutant strains are presented in Table 19. atives has revealed a point mutation at position 181
Further work with the thio analogues of this series have (Tyr→ Cys) (Ijichi et al., 1996). When anti-HIV-1
led to three new highly active compounds related to UC 10 assays were carried out in the presence of 50% HS to
(Figure 15 and Table 20), with the compound UC 781 as a evaluate TDA under more physiological conditions, the
very potent inhibitor (Buckheit et al., 1997). inhibitory effects on HIV-1 replication showed a
UC 781 shows excellent inhibitory activity against sev- 10–200-fold decrease. The degree of reduction depends
eral mutant HIV-1 strains, including the L100I, V106A, on the lipophility, more lipophilic leading to less
E138K and Y181C mutations (Table 21). In a test it has inhibitory effect. The TDA derivatives were bound to
selected for the mutations Y181C, V108I and K101E in serum protein by more than 90% under these conditions
order of appearance (Buckheit et al., 1997). UC 781 was (Ijichi et al., 1996).
shown to be a rapid tight-binding inhibitor of HIV-1 RT
(Barnard et al., 1997). In addition, UC 781 has been found Diarylsulphones
to readily penetrate isolated HIV-1 particles, and treatment The basic structure of diarylsulphones consists of two sub-
of isolated HIV-1 virions with UC 781 result in rapid inac- stituted phenyl groups linked by a sulphone group. In a
tivation of the virus (Borkow et al., 1997). UC 781 may screening of 54 diarylsulphones and related derivatives, 2-

Table 21. Activities of UC compounds against viruses resistant to HIV-1-specific inhibition

EC50 (µM)*
Inhibitor to which isolate
is resistant (mutation) UC 10 UC 040 UC 781 UC 82 Nevirapine
HIV-1IIIB (control) 0.2 0.2 0.01 0.008 0.01
Oxathiin caboxanilide (L100I) 2.0 >2 0.5 0.3 0.1
UC 10-Costatolide (K103N) 7.3 >2 0.9 1.0 ND†
Thiazolobenzimidazole (V108I) 1.2 0.9 0.1 0.06 0.3
TIBO (A98G/V108I) 1.1 1.2 0.3 0.2 0.6
Calanolide A (T139I) 0.3 0.4 0.04 0.03 0.01
Diphenylsulphone (Y181C) 1.1 >2 0.9 0.2 5.9
Pyridionone (Y181C/L103N) >100 >2 >2 >2 >38
Costatolide (Y188H) 1.6 1.0 0.1 0.07 ND
HEPT (P236L) 0.2 0.4 0.02 0.02 0.02

*Concentration required to inhibit the spread of HIV-1IIIB in CEM-SS cells by 50% (XTT method).
†ND, Not determined.
Data from Buckheit et al. (1997).

Antiviral Chemistry & Chemotherapy 10:6 303


OS Pedersen & EB Pedersen

Table 22. Structure-activity relationsship of the TDA derivatives


R2

N R3
O
R1
O

N N
S

Compound R1 R2 R3 EC50 (µM)* CC50 (µM)†


RD3-2105 – Me Me 29 210
RD3-2356 2-Cl Me Me 0.44 162
RD3-2107 4-Cl Me Me 353 353
RD3-2236 2,3-dichloro Me Me 0.94 127
RD3-2219 2,4-dichloro Me Me 8.3 77
RD3-2218 3,4-dichloro Me Me 315 315
RD3-2233 2,5-dichloro Me Me 0.37 111
RD3-2220 2,6-dichloro Me Me 0.20 190
RD4-2025 2,6-dichloro Me Et 0.037 29
RD4-2024 2,6-dichloro Me Pr 0.013 28
RD3-2102 2,6-dichloro Me Bu 0.020 23
RD4-2031 2,6-dichloro Me Hex 0.12 19
RD3-2222 2,6-dichloro C5H10 0.96 221
RD3-2101 2,6-dichloro Et Et 0.25 314
RD4-2023 2,6-dichloro Et Bu 0.33 44
RD3-2380 2,6-dichloro H Bu 139 139

*50% Effective concentration, required to inhibit HIV-1-induced CPE by 50% in MT-4 cells (MTT method).
†50% Cytotoxic concentration, required to reduce cell viability by 50% in mock-infected MT-4 cells.
Data from Ijichi et al., (1996).

nitrophenyl phenyl sulphone (NPPS) was identified as a 1996). Several of the most active compounds in a series
RT inhibitor of the non-nucleoside class (McMahon et al., studied by Buckheit et al. (1996) had a chloro and a methy-
1993). The most active compounds are those with an lamine on the ring not containing the nitro group (Table
ortho-nitro group (McMahon et al., 1993; Buckheit et al., 23). Compounds where methyl replaced chloro were only

Figure 16. The structure of calanolide A and some anti-HIV active analogues

O O O O

O O O O O O O O O O O O

OH OH OH OAc

(+) Calanolide A (+) Calanolide B (–) Calanolide B 12-Acetoxycalanolide

ED50 0.1 µM ED50 0.4 µM (Costatolide) ED50 2.7 µM


IC50 20 µM IC50 15 µM ED50 0.3 µM IC50 13 µM

IC50 5.8 µM

EC50 is the concentration of inhibitor needed to prevent the spread of HIV by 50%. IC50 is the concentration of the inhibitor which reduce the
viability of host cells by 50%.

304 ©1999 International Medical Press


The NNRTI boom

Table 23. Antiviral HIV-1 activity and RT inhibition of some diarylsulphones

R1 O O NO2

R2 S

R3

R4

Compound (NSC) R1 R2 R3 R4 EC50 (µM)* IC50 (µM)†


665527 -NH(CH2)3- Cl H 0.3 >200
667948 -NH(CH2)3- Me H 0.5 33
667951 NHMe H H Me 0.4 18.4
667952 NHMe H H Cl 0.08 61
671291 H Me NHMe H 0.2 32.7
671292 H Cl NHMe H 0.1 >200

*Concentration of compound required to inhibit the spread of HIV-1 in CEM-SS cells by 50% (XTT method).
†Concentration of compound required to inhibit HIV-1 RT by 50% using a rC.dG template primer.
Data from Buckheit et al. (1996).

slightly less active (Buckheit et al., 1996) and reduction of from Sarawak in Malaysia, showed anti-HIV activity in an
the sulphone linker also reduces the antiviral activity initial test (Kashman et al., 1992). From this extract some
(McMahon et al., 1993). Compound NSC 667952 was the coumarins were isolated and identified. The most active of
most active in the series, retaining activity against the drug- these compounds, in descending order, were (+)calanolide
resistant virus isolate and having a sensitivity profile against A, (+)calanolide B and the ester derivative 12-acetoxy-
the L100I mutation (Buckheit et al., 1996). The diarylsul- calanolide (Sarawak MediChem Pharmaceuticals) (Figure
phones have recently been investigated as HIV-1 integrase 16). In a search for calanolide A in other and more abun-
inhibitors. Some have been found to be interesting enough dant sources, several species of callophyllum were tested
for further investigation (Neamati et al., 1997). (Fuller et al., 1994). In the latex from C. teysmannii miq.
var. inophylloide, a compound identified as the (–)calanolide
Calanolide A B or costatolide was isolated and tested. This costatolide,
A 1:1 CH2Cl2:MeOH extract of fruits and twigs from though a known compound not previously identified as
Callophyllum lanigerum var. austrocoriaceum, a rainforest tree having antiviral activity, showed anti-HIV activity compa-

Figure 17. The structure of the two pyridinones L-697,639 and L-697,661
Cl
H3 C

N
N H
H N
N O
O Cl
CH3
N O
N O H
H

L-697, 639 L-697, 661

HIV-1 RT HIV-1IIIB*

IC50† CIC95‡

L-697,639 20 nM 25–50 nM

L-697,661 19 nM 25–50 nM

*Assays using MT-4 cell culture preinfected with HIV-1IIIB at a low multiplicity.
†Concentration that caused 50% inhibition of HIV RT.
‡Concentration that inhibited the spread of HIV-1 infection by ≥95%.
Data from Goldman et al. (1991).

Antiviral Chemistry & Chemotherapy 10:6 305


OS Pedersen & EB Pedersen

Table 24. Anti-HIV-1 and anti-HIV-1 RT activity of Pyridinones


some pyridyl- and phenyl analogues of the 2-
The pyridinones L-697,639 and L-697,661 (Merck
pyridinone class
Research Laboratories; Figure 17) were reported in 1991
3′
MeO X (Goldman et al., 1991). Compound L-697,661, with 95%
4′
clearance concentration (ClC95) values in the nanomolar
H R
N 5′ range, has been clinically tested and good antiviral activity
Y was observed. However, antiviral activity was of short dura-
6′
tion owing to rapid onset of viral resistance (Hoffman et al.,
N O
H 1992). When new drug candidates are tested in HIV
antiviral and RT enzymatic assays they are often compared
X Y R IC50 (nM)* ClC95 (nM)† to L-697,661.
N CH 4′-Me, 5′-Et 19 13 Structural modifications on the 2-pyridone moiety have
CH CH 4′,5′-Me2 8 13 not been successful (Hoffman et al., 1992; Saari et al.,
N CH 4′,5′-Me2 9 50 1992). These include N-methylation, O-methylation, dele-
CH N 4′,5′-Me2 70 100
tion of one or both alkyl groups and substituting 5-ethyl
CH CH 265 400
with 5-methyl. Some structure–activity studies have also
N CH 680 800
been done replacing the benzoxazole by a pyridyl or a
*Concentration of compound that inhibit HIV-1 RT by 50% using phenyl (Wai et al., 1993) (Table 24). It was found that
rC.dG template primer.
†Concentration of compound that inhibits the spread of HIV-1 maximum enzyme inhibition was obtained with com-
infection in MT-4 cells, by ≥95% using a p24 core antigen ELISA pounds containing a 2′-methoxy group on either the
assay.
Data from Wai et al. (1993). phenyl or pyridyl ring. In the phenyl subclass several sub-
stituents were tried in the ortho position. Ethoxy- and
rable to (+)calanolide A (Fuller et al., 1994). nitro substituents were comparable with the methoxy sub-
A lot of structure–activity studies concerning (+)calano- stituent and some substituents (CN, F, Cl, MeS, Me) gave
lide A and (–)calanolide B have been performed (Galinis et only modestly active compounds. Substituents such as CF3,
al., 1996; Zembower et al., 1997) and despite increased NH2 and MeOCH2 were detrimental to activity. There is
knowledge of the structure–activity relationship for this no significant difference between the 2-methoxyphenyl
class of NNRTI, no new compounds more potent than the and 2-methoxy-3-pyridyl derivatives. In the pyridyl sub-
lead compounds have been found. Calanolide A is present- class the position of the pyridine nitrogen was found to be
ly in clinical testing. important, with the position of nitrogen at 3′ as the most

Table 25. Inhibition of HIV-1 RT and viral infection spread by selected 2-pyridinone analogues together with
the rates of acid hydrolysis
N
X
R

O
N O
H

Compound X R IC50 (nM)* ClC95 (nM) T1/2 (min)‡


L-697,661 NH 4,7-Cl2 20±4 50–100 17±4
1 CH2 H 23±1 50–100 218±18
2 CH2 4,7-Cl2 14±3 50 29±1
3 NH H 210±20 400–800 35±7
4 O H 191±30 ND¶ 12±0.2
5 O 4,7-Cl2 88±16 200 11±0.2
6 S 4,7-Cl2 11±1 25–50 34±7

*The concentration that produced 50% inhibition of RT using a rC.dG template primer, and stated as the mean of at least three determina-
tions ±SEM.
†The cell culture inhibitor concentration required to inhibit by > 95% the spread of HIV-1 in susceptible cell culture, stated as a range of
values if multiple determinations were made. An assay using MT-4 cells and HIV-1IIIB strain was used.
‡Determination were carried out in 90% ethanol, 10% 1 M HCl to ensure compound solubility and to maintain a final HCl concentration of
0.1 M.
¶ND, not determined.
Data from Hoffman et al. (1993).

306 ©1999 International Medical Press


The NNRTI boom

Table 26. Anti-HIV-1 RT activity and cytotoxicity of some α-APA analogues


R2

O NH2
Cl

N
H
R1
Cl

Compound Isomer R1 R2 EC50 (nM)* CC50 (µM)† SI‡


R 15345 (±) OCH3 H 610 15 25
R 18893 (±) NO2 H 88 80 909
R 87231 (+) NO2 H 1700 82 48
R 87232 (–) NO2 H 33 66 2000
R 88703 (±) C(O)CH3 H 26 130 5000
R 88976 (+) C(O)CH3 H 6500 120 18
R 88977 (–) C(O)CH3 H 19 52 2737
R 89439 (±) C(O)CH3 CH3 13 710 54615
R 90384 (+) C(O)CH3 CH3 2100 270 129
R 90385 (–) C(O)CH3 CH3 5 420 84000
L-697,661 22 320 14545
AZT 0.5 3.5 7000

*Concentration required for prevention of spread of HIV-1 in cell culture by 50% (MTT procedure).
†Fifty percent toxicity concentration in MT-4 cells (MTT procedure).
‡Selectivity index is the ratio of CC50 to EC50.
Data from Pauwels et al. (1993).

Figure 18. Other NNRTIs

H3C NH2
O
N N
N
O Cl S

Cl N O O
S O O
CH3 O
O O
1 2 3

NH2

H3C F

Cl N
S F
N N
O O N
N
O O
F F
F
S F
4 5 6

N
O
N N
H3C H
NH N
O
Cl S SiO
O O O
SiO N O
O
O
NH2
N NH2
H O OSi
NH2 S
O OSi
S
O O
O O
7 8 9

Antiviral Chemistry & Chemotherapy 10:6 307


OS Pedersen & EB Pedersen

Figure 19. Other NNRTIs

H H
N S N S

CH3 CH3
H3C S
N CH3 O N H

O O S
O O
N
H2C CH3 H 3C CH3
O
10 11 12

S-2720 HBY 097 Thiazoloisoindol-5-one, C10H7 derivative

MeO

MeO
N CN
MeO
O

MeO OMe
Cl

OMe
13 14

U-78036

favourable. Small lipophilic groups at 4′- and 5′-positions ysis than the other derivatives. Compounds with an ether,
are required for optimum activity and analogues with larg- or a thioether linker were also prepared and tested (Table
er alkyl groups are less potent. Methyl substitution at the 25). The ether linker decreased the activity, whereas the
3′- and 6′-positions was detrimental. In this series of com- analogue of L-697,661 with a thioether linker was more
pounds 3-[[(5-ethyl-2-methoxy-6-methyl-3-pyridyl)- inhibitory to HIV-1 RT than L-697,661 and had an
methyl]amino]-5-ethyl-6-methylpyridin-2(1H)-one (first improved stability in an acid environment. It was not as
entry in Table 24) was selected for clinical evaluation due stable as the unsubstituted counterpart with an ethyl link-
to the best oral availability in rats and monkeys. er (Compound 1 in Table 25).
A structure–activity relationship study of 2-pyridinones The early candidate of the pyridinones, L-697,661, was
with a hydrolytically more stable ethyl linker in place of the very tightly bound (99.6%) to human plasma and com-
amino-methylene linker was made (Hoffman et al., 1993) pound 1 was found to be 4% unbound to human plasma
although the original lead had shown less potential with an protein, which suggested a 10-fold higher free drug level in
ethyl linker. As observed in earlier studies, a change in the vivo. Compound 1 has been studied clinically in HIV-pos-
length or a reduction in the flexibility decreased the itive patients. Resistance to compound 1 has been observed
inhibitory potency. The effect of thiocarbonyl modification in cell culture experiments with virus from clinical isolates
of the pyridinones was minimal in enzyme inhibition, but obtained from patients who developed resistance to L-
they were less effective in the prevention of virus spread in 697,661 (Hoffman et al., 1993).
cell culture, which could reflect poorer cell penetration of
these pyridinethione analogues. Many of the compounds α-APA (α-anilinophenylacetamide)
with a benzoxazole as in L-697,661 and an ethylene linker A screening programme of 2000 compounds belonging to
were highly active, particularly those unsubstituted, substi- different classes led to the discovery of the lead α-APA
tuted in the 4 or 7 position or disubstituted in the 4 and 7 derivative R 15345 with an EC50 of 0.6 µM in MT-4 cells
position. The number of potent candidates was reduced by (Table 26). Through evaluation of structurally related
investigating the susceptibility of hydrolysis in acidic envi- compounds, R 18893 with a nitro group in place of the
ronments. This revealed one compound, the unsubstituted methoxy substituent in the ortho position of the aniline
analogue 1 in Table 25, 10-fold more stable to acid hydrol- moiety was identified as a more potent inhibitor, with an

308 ©1999 International Medical Press


The NNRTI boom

EC50 value of 88 nM in MT-4 cells (Pauwels et al., 1993). nanomolar range and a selectivity index of 104 (Kleim et
At this stage it appeared as the antiviral activity was stere- al., 1993). HBY 097, another quinoxaline, was withdrawn
ospecific as the (–) isomer was 50-fold more potent (EC50 after finishing Phase IIa clinical testing. HBY 097 showed
33 nM) compared with the (+) isomer (EC50 1700 nM). activity against HIV-1 in PBL (mean of 41 clinical iso-
Further optimization was made by replacing the nitro lates) with an EC50 of 0.002 µM and with a selectivity
substituent with an acetyl group, R88703 and by adding a index of 12500 (Kleim et al., 1995). Strains of HIV-1 gen-
methyl substituent in the para position of the acetyl group erated under high selective pressure of HBY 097 revealed
on the aniline moiety. This led to the most potent a G190E mutation which is characteristic for the quinox-
inhibitor of this series, 90385, with a selectivity index of alines (Kleim et al., 1995). RT with glutamic acid in posi-
80000 (Pauwels et al., 1993). The compound R 89439 (SI tion 190 has shown a dramatic decrease in enzymatic
54,615) was further evaluated against some mutant activity compared to the HIV-1MN RT (wild-type) (Kleim
strains of HIV-RT and RT mutants containing a Y181C et al., 1993). Compound 12 [(R)-9b-(1-naphtyl)-2,3-
or Y188C mutation showed resistance to α-APA deriva- dihydrothiazolo[2,3-a]isoindol-5(9bH)-one] is a member
tives, whereas RT containing a I100L mutation was still of the thiazoloisoindol-5-ones, which has shown an EC50
sensitive to R 89439. A relatively easy chemical synthesis value of 0.046 µM in MT-2 cells, but the compound was
of R 89439, good oral availability and favourable pharma- only moderately orally available in rats (Mertens et al.,
cokinetics made R 89439 (loviride) a valid candidate 1993). Also, the quinoline U-78036 (13) has shown activ-
(Pauwels et al. 1993). The α-APA project and further ity against HIV-1 RT (Althaus et al., 1993). A tetrahy-
development of loviride has been stopped by Janssen dronaphtalene lignan derivative (14) has shown a 50%
Pharmaceuticals. effective inhibition concentration in antiviral assays of
0.15 to 0.8 µM and a selectivity index of 70–400 (Hiroto
Other compounds et al., 1997).
A lot of compounds and classes of compounds have been
found to have activity against HIV-1 RT have been syn- Future perspectives for NNRTIs
thesized. Some of these are presented in Figures 18 and 19.
Compound 1 was the most active of the 5-H-pyrrolo[1,2- NNRTIs appear to be very promising therapies in the
b][1,2,5]benzothiadiazepines (PBTDs) with an EC50 value treatment of HIV, when used in combination with other
of 0.5 M and a selectivity index of >600 (Artico et al., anti-HIV drugs such as nucleoside RT inhibitors and pro-
1996). Compound 2, a pyrrolobenzoxazepinone with an tease inhibitors. NNRTIs have the advantage that they are
IC50 value in an enzymatic assay (rC.dG) of 250 nM com- able to cross the blood–brain barrier and in this way can
pared to nevirapine (IC50 500 nM), was the most potent have an antiviral effect against HIV-1 infections in reser-
NNRTI in a series by Campiani et al. (1996). Further work voirs that are out of reach for many other anti-HIV drugs,
with sulphones and NPPS as a lead afforded the pyrryl aryl including most of the nucleoside RT inhibitors.
sulphones (PAS). Two of these compounds, 3 and 4, had The first generation of NNRTIs, nevirapine and
similar anti-HIV-1 activities with an EC50 of 0.14 µM in delavirdine, have suffered from a rapid development of
MT-4 cells and a selectivity index of >2140. This is a high- resistance. To overcome this problem, the NNRTIs have
er EC50 value and selectivity index than for zidovudine been used in high doses and in combination with two or
(Silvestri et al., 1997). three other anti-HIV drugs in HAART. The problem of
With TBZ (5) (Chimirri et al., 1997) as the lead struc- resistant strains of HIV may be reduced with the second
ture, a 2-aryl-substituted benzimidazole 6 with an EC50 generation of NNRTIs because RT will need two or more
value of 0.20 Μ in an antiviral assay was synthesized (Roth mutations for HIV-1 to be resistant to these drugs. Also,
et al., 1997). the uptake of the second generation of NNRTIs in the
A sulphone compound (7) was found to inhibit viral blood and cells may be increased and thereby increase the
spread in MT-4 cells with a ClC95 of 3 nM (Williams et al., concentration that can effect the inhibition of RT. The per-
1993). spectives of the new generation of NNRTI inhibitors are
Derivatives of TSAO-T (8) are also inhibitors of HIV-1 thus promising with an expected slower development of
RT and are unique in that they interfere at the interface resistance combined with their ability to cross the
between the p51 and p66 subunits of RT. One 1,2,3-triazole- blood–brain barrier.
TSAO analogue, compound 9, is reported to have a selectiv-
ity index of 1470 in MT-4 cells (Velázquez et al., 1998). References
Quinoxaline derivatives have also been an important
class of NNRTIs. S-2720 has shown activity in a cell Ahgren C, Backro K, Bell FW, Cantrell AS, Clemens M, Colacino
spread assay with a 50% effective concentration in the JM, Deeter JB, Engelhardt JA, Hogberg M, Jaskunas SR, Johansson

Antiviral Chemistry & Chemotherapy 10:6 309


OS Pedersen & EB Pedersen

NG, Jordan CL, Kasher JS, Kinnick MD, Lind P, Lopez C, HIV-1 reverse transcriptase inhibitors. 1. Synthesis and basic
Morin JM Jr, Muesing MA, Noreen R, Oberg B, Paget CJ, structure–activity relationship of PETT analogs. Journal of
Palkowitz JA, Parrish CA, Pranc P, Rippy MK, Rydergard C, Medicinal Chemistry 38:4929–4936.
Sahlberg C, Swanson S, Ternansky RJ, Unge T, Vasileff RT, Vrang Borkow G, Barnard J, Nguyen TM, Belmonte A, Wainberg MA &
L, West SJ, Zhang H & Zhou X-X (1995) The PETT series, a Parniak MA (1997) Chemical barriers to human immunodeficien-
new class of potent nonnucleoside inhibitors of human immunod- cy virus type 1 (HIV-1) infection: retrovirucidal activity of UC781,
eficiency virus type 1 reverse transcriptase. Antimicrobial Agents and a thiocarboxanilide nonnucleoside inhibitor for HIV-1 reverse
Chemotherapy 39:1329–1335. transcriptase. Journal of Virology 71:3023–3030.
Althaus IW, Gonzales AJ, Chou JJ, Romero DL, Deibel MR, Chou Botta M, Artico M, Massa S, Gambacorta A, Marongiu ME, Pani
KC, Kezdy FJ, Resnick L, Busso ME & So AG (1993) The A & La Colla P (1992) Synthesis, antimicrobial and antiviral
quinoline U-78036 is a potent inhibitor of HIV-1 reverse tran- activities of isotrimethoprim and some related derivatives.
scriptase. Journal of Biological Chemistry 268:14875–14880. European Journal of Medicinal Chemistry 27:251–257.
Althaus IW, Chou K-C, Lemay RJ, Franks KM, Deibel MR, Kezdy Breslin HJ, Kukla MJ, Ludovici DW, Mohrbacher R, Ho W,
FJ, Resnick L, Busso ME, So AG, Downey KM, Romero DL, Miranda M, Rodgers JD, Hitchens TK, Leo G, Gauthier DA, Ho
Thomas RC, Aristoff PA, Tarpley WG & Reusser F (1996) The CY, Scott MK, De Clercq E, Pauwels R, Andries K, Janssen
benzylthio-pyrimidine U-31,355, a potent inhibitor of HIV-1 MAC & Janssen PAJ (1995) Synthesis and anti-HIV-1 activity of
reverse transcriptase. Biochemical Pharmacology 51:743–750. 4,5,6,7-tetrahydro-5-methylimidazo[4,5,1-jk][1,4]benzodiazepin-
Arranz E, Díaz JA, Ingate ST, Witrouw M, Pannecouque C, 2(1H)-one (TIBO) derivatives. 3. Journal of Medicinal Chemistry
Balzarini J, De Clercq E & Vega S (1998) Novel 1,1,3-trioxo- 38:771–793.
2H,4H-thieno[3,4-e][1,2,4]thiadiazine derivatives as non-nucleo- Buckheit RW Jr, Fliakas-Boltz V, Russell JD, Snow M, Pallansch
side reverse transcriptase inhibitors that inhibit human LA, Yang SS, Bader JP, Khan TN & Zanger M (1996) A diaryl-
immunodeficiency virus type 1 replication. Journal of Medicinal sulphone non-nucleoside reverse transcriptase inhibitor with a
Chemistry 41:4109–4117. unique sensitivity profile to drug resistant virus isolates. Antiviral
Artico M (1996) Non-nucleoside anti-HIV-1 reverse transcriptase Chemistry & Chemotherapy 7:243–252.
inhibitors (NNRTIs): a chemical survey from lead compounds to Buckheit RW Jr, Snow MJ, Fliakas-Boltz V, Kinjerski TL, Russell
selected drugs for clinical trials. Il Farmaco 51:305–331. JD, Pallansch LA, Brouwer WG & Yang SS (1997) Highly potent
Artico M, Massa S, Mai A, Marongiu ME, Piras G, Tramontino E oxathiin carboxanilide derivatives with efficacy against nonnucleo-
& La Colla P (1993) 3,4-Dihydro-2-alkoxy-6-benzyl-4-oxopy- side reverse transcriptase inhibitor-resistant human immunodefi-
rimidines (DABOs): a new class of specific inhibitors of human ciency virus isolates. Antimicrobial Agents and Chemotherapy
immunodeficiency virus Type 1. Antiviral Chemistry & 41:831–837.
Chemotherapy 4:361–368. Campiani G, Nacci V, Fiorini I, De Filippis MP, Garofalo A, Greco
Artico M, Silvestri R, Pagnozzi E, Stefancich G, Massa S, Loi AG, G, Novellino E, Altamura S & Di Renzo L (1996)
Putzolu M, Corrias S, Spiga MG & La Colla P (1996) 5-H- Pyrrolobenzothiazepinones and pyrrolobenzoxazepinones: novel
Pyrrolo[1,2-b][1,2,5]benzothiadiazepines (PBTDs): a novel class and specific non-nucleoside HIV-1 reverse transcriptase inhibitors
of non-nucleoside reverse transcriptase inhibitors. Bioorganic and with antiviral activity. Journal of Medicinal Chemistry
Medicinal Chemistry Letters 4:837–850. 39:2672–2680.

Baba M, Yuasa S, Niwa T, Yamamoto M, Yabuuchi S, Takashima H, Cantrell AS, Engelhardt P, Högberg M, Jaskunas SR, Johansson
Ubasawa M, Tanaka H, Miyasaka T, Walker RT, Balzarini J, De NG, Jordan CL, Kangasmetsä J, Kinnick MD, Lind P, Morin JM
Clercq E & Shigeta S (1993). Effect of human serum on the in Jr, Muesing MA, Noreén R, Öberg B, Pranc P, Sahlberg C,
vitro anti-HIV-1 activity of 1-[(2Hydroxyethoxy)methyl]-6- Ternansky RJ, Vasileff RT, Vrang L, West SJ & Zhang H (1996)
(phenylthio)thymine (HEPT) derivatives as related to their Phenethylthiazolylthiourea (PETT) compounds as a new class of
lipophilicity and serum protein binding. Biochemical Pharmacology HIV-1 reverse transcriptase inhibitors. 2. Synthesis and further
45:2507–2512. structure-activity relationship studies of PETT analogs. Journal of
Medicinal Chemistry 39: 4261–4274.
Bader JP, McMahon JB, Schultz RJ, Narayanan VL, Pierce JB,
Harrison WA, Weislow OS, Midelfort CF, Stinson SF & Boyd Chimirri A, Grasso S, Molica C, Monforte A-M, Monforte P,
MR (1991) Oxathiin carboxanilide, a potent inhibitor of human Zappala M, Bruno G, Nicolo F, Witvrouw M, Jonckeere H,
immunodeficiency virus reproduction. Proceedings of the National Balzarini J & De Clercq E (1997) Structural features and anti-
Academy of Sciences, USA 88:6740–6744. human immunodeficiency virus (HIV) activity of the isomers of 1-
(2′,6′-difluorophenyl)-1H,3H-thiazolo[3,4-a]benzimidazole, a
Balzarini J, Brouwer WG, Felauer EE, De Clercq E & Karlsson A potent non-nucleoside HIV-1 reverse transcriptase inhibitor.
(1995) Activity of various thiocarboxanilide derivatives against Antiviral Chemistry & Chemotherapy 8:363–370.
wild-type and several mutant human immunodeficiency virus type
1 strains. Antiviral Research 27:219–236. Cushman M, Golebiewski M, Buckheit RW Jr, Graham L & Rice
WG (1995) Synthesis and biological evaluation of an alkenyl-
Barnard J, Borkow G & Parniak MA (1997) The thiocarboxanilide diarylmethane (ADAM) which acts as a novel non-nucleoside
nonnucleoside UC781 is a tight-binding inhibitor of HIV-1 HIV-1 recerse transcriptase inhibitor. Bioorganic & Medicinal
reverse transcriptase. Biochemistry 36:7786–7792. Chemistry Letters 5:2713–2716.
Barth B, Dierich M, Heinisch G, Jenny V, Matuszczak B, Mereiter Cushman M, Golebiewski WM, Graham L, Turpin JA, Rice WG,
K, Planer R, Schöpf I, Stoiber H, Traugott T & Aufschnaiter PV Fliakas-Boltz V & Buckheit RW Jr (1996) Synthesis and biologi-
(1996) Pyridazino[3,4-b][1,5]benzoxazepin-5(6H)-ones: synthesis cal evaluation of certain alkenyldiarylmethanes as anti-HIV-1
and biological evaluation. Antiviral Chemistry & Chemotherapy agents which act as non-nucleoside reverse transcriptase inhibitors.
7:300–312. Journal of Medicinal Chemistry 39:3217–3227.
Bell FW, Cantrell AS, Högberg M, Jaskunas SR, Johansson NG, Cushman M, Casimiro-Garcia A, Hejchman E, Ruell JA, Huang
Jordan CL, Kinnick MD, Lind P, Morin JM Jr, Noréen R, Öberg M, Schaeffer CA, Williamson K, Rice WG & Buckheit RW Jr
B, Palkowitz JA, Parrish CA, Pranc P, Sahlberg C, Ternansky RJ, (1998a) New alkenyldiarylmethanes with enhanced potencies as
Vasileff RT, Vrang L, West SJ, Zhang H & Zhou X-X (1995) anti-HIV agents which act as non-nucleoside reverse transcriptase
Phenethylthiazolethiourea (PETT) compounds, a new class of inhibitors. Journal of Medicinal Chemistry 41:2076–2089.

310 ©1999 International Medical Press


The NNRTI boom

Cushman M, Casimiro-Garcia A, Williamson K & Rice WG tives: specifiic human immunodeficiency virus type 1 reverse tran-
(1998b) Synthesis of a non-nucleoside reverse transcriptase scriptase inhibitors with antiviral activity. Proceedings of the
inhibitor in the alkenyldiarylmethane (ADAM) series with opti- National Academy of Sciences, USA 88:6863–6867.
mized potency and therapeutic index. Bioorganic and Medicinal Hanasaki Y, Watanabe H, Katsuura K, Takayama H, Shirakawa S,
Chemistry Letters 8:195–198. Yamaguchi K, Sakai S-I, Ijichi K, Fujiwara M, Konno K, Yokota
Cywin CL, Klunder JM, Hoermann M, Brickwood JR, David E, T, Shigeta S & Baba M (1995) Thiadiazole derivatives: highly
Grob PM, Schwartz R, Pauletti D, Barringer KJ, Shih C-K, Sorge potent and specific HIV-1 reverse transcriptase inhibitors. Journal
CL, Erickson DA, Joseph DP & Hattox SE (1998) Novel nonnu- of Medicinal Chemistry 38:2038–2040.
cleoside inhibitors of HIV-1 reverse transcriptase. 8. 8- Hargrave KD, Proudfoot JR, Grozinger KG, Cullen E, Kapadia SR,
Aryloxymethyl- and 8-arylthiomethyldipyridodiazepinones. Patel UR, Fuchs VU, Mauldin SC, Vitous J, Behnke ML, Klunder
Journal of Medicinal Chemistry 41:2972–2984. JM, Pal K, Skiles JW, McNeil DW, Rose JM, Chow GC, Skoog
Danel K, Larsen E, Pedersen EB, Vestergaard BF & Nielsen C MT, Wu JC, Schmidt G, Engel WW, Eberlein WG, Saboe TD,
(1996) Synthesis and potent anti-HIV-1 activity of novel 6-benzy- Campbell SJ, Rosenthal AS & Adams J (1991) Novel non-nucleo-
luracil analogs of 1-[(2-hydroxyethoxy)methyl]-6- side inhibitors of HIV-1 reverse transcriptase. 1. Tricyclic pyri-
(phenylthio)thymine. Journal of Medicinal Chemistry dobenzo- and dipyridodiazepinones. Journal of Medicinal Chemistry
39:2427–2431. 34:2231–2241.
Danel K, Nielsen C & Pedersen EB (1997) Anti-HIV active naphtyl Heinisch G, Huber E, Matuszczak B, Maurer A & Prillinger U
analogues of HEPT and DABO. Acta Chemica Scandinavica (1997a) Synthesis of pyridazino[3,4-b][1,5]benzodiazepin-5-ones
51:426–430. and their biological evaluation as non-nucleoside HIV reverse
transcriptase inhibitors. Archive der Pharmacie 330:29–34.
Danel K, Pedersen EB & Nielsen C (1998) Synthesis and anti-HIV-
1 activity of novel 2,3-dihydro-7H-thiazolo[3,2-a]pyrimidin-7- Heinisch G, Matuszczak B, Pachler S & Rakowitz D (1997b) The
ones. Journal of Medicinal Chemistry 41:191–198. inhibitory activity of diazinyl-substituted thiourea derivatives on
human immunodeficiency virus type 1 reverse transcriptase.
De Clercq E (1998a) The role of non-nucleoside reverse transcrip-
Antiviral Chemistry & Chemotherapy 8:443–446.
tase inhibitors (NNRTIs) in the therapy of HIV-1 infection.
Antiviral Research 38:153–179. Hiroto H, Fujihashi T, Sakata T, Kaji A & Kaji H (1997)
Tetrahydronaphtalenelignan compounds as potent anti-HIV type
De Clercq E (1998b) New perspectives for the treatment of HIV
1 agents. AIDS Research and Human Retroviruses 13:695–705.
infections. Collection of Czechoslovak Chemical Communications
63:449–479. Ho W, Kukla MJ, Breslin HJ, Ludovici DW, Grous PP, Diamond
CJ, Miranda M, Rodgers JD, Ho CY, De Clercq E, Pauwels R,
Ding J, Das K, Moereels H, Koymans L, Andries K, Janssen PAJ,
Andries K, Janssen MAC & Janssen PAJ (1995) Synthesis and
Hughes SH & Arnold E (1995) Structure of HIV-1 RT/TIBO R
anti-HIV-1 activity of 4,5,6,7-tetrahydro-5-methylimidazo[4,5,1-
86183 complex reveals similarity in the binding of diverse nonnu-
jk][1,4]benzodiazepin-2(1H)-one (TIBO) derivatives. 4. Journal of
cleoside inhibitors. Nature Structural Biology 2:407–415.
Medicinal Chemistry 38:794–802.
Dueweke TJ, Poppe SM, Romero DL, Swaney SM, So AG,
Hoffman JM, Wai JS, Thomas CM, Levin RB, Goldman ME &
Downey KM, Althaus IW, Reusser F, Busso M, Resnick L,
O’Brien JA (1992) Synthesis and evaluation of 2-pyridinone deriv-
Mayers DL, Lane J, Aristoff PA, Thomas RC & Tarpley WG
atives as specific HIV-1 reverse transcriptase inhibitors. 1.
(1993) U-90152, a potent inhibitor of human immunodeficiency
Phtalimidoalkyl and -alkylamino analogues. Journal of Medicinal
virus type 1 replication. Antimicrobial Agents and Chemotherapy
Chemistry 35:3784–3791.
37:1127–1131.
Hoffman JM, Smith AM, Rooney CS, Fisher TE, Wai JS, Thomas
Fujiwara T, Sato A, El-Farrash M, Miki S, Abe K, Isaka Y, Kodama
CM, Bamberger DL, Barnes JL, Williams TM, Jones JH, Olson
M, Wu Y, Chen BL, Harada H, Sugimoto H, Hatanaka M &
BD, O’Brien JA, Goldman ME, Nunberg JH, Quintero JC,
Hinuma Y (1998) S-1153 inhibits replication of known drug-
Schleif WA, Emini EA & Anderson PS (1993) Synthesis and
resistant strains of human immunodeficiency virus type 1.
evaluation of 2-pyridinone derivatives as specific reverse transcrip-
Antimicrobial Agents and Chemotherapy 42:1340–1345.
tase inhibitors. 4. 3-[2-(Benzoxazol-2-yl)ethyl]-5-ethyl-6-
Fuller RW, Bokesch HR, Gustafson KR, McKee TC, Cardellina JH methylpyridin-2(1H)-one and analogues. Journal of Medicinal
II, McMahon JB, Cragg GM, Soejarto DD & Boyd MR (1994) Chemistry 36:953–966.
HIV-inhibitory coumarins from latex of the tropical rainforest tree
Hopkins AL, Ren J, Esnouf RM, Willcox BE, Jones EY, Ross C,
Calophyllum teysmannii var. Inophylloide. Bioorganic & Medicinal
Miyasaka T, Walker RT, Tanaka H, Stammers DK & Stuart DI
Chemistry Letters 4:1961–1964.
(1996). Complexes of HIV-1 reverse transcriptase with inhibitors
Galinis DL, Fuller WT, McKee TC, Cardellina JH II, Gulakowski of the HEPT series reveal conformational changes to the design of
RJ, McMahon JB & Boyd MR (1996) Structure–activity modifi- potent non-nucleoside inhibitors. Journal of Medicinal Chemistry
cations of the HIV-1 inhibitors (+)-calanolide A and (–)-calano- 39:1589–1600.
lide B. Journal of Medicinal Chemistry 39:4507–4510.
Ijichi K, Fujiwara M, Nagano H, Matsumoto Y, Hanasaki Y, Ide T,
Genin MJ, Poel TJ, Yagi Y, Biles C, Althaus I, Keiser BJ, Kopta LA, Katsuura K, Takayama H, Shirakawa S, Aimi N, Shigeta S, Konno
Friis JM, Reusser F, Adams WJ, Olmsted RA, Voorman RL, K, Matsushima M, Yokota T & Baba M (1996) Anti-HIV-1
Thomas RC & Romero DL (1996) Synthesis and bioactivity of activity of thiadiazole derivatives: structure–activity relationship,
novel bis(heteroaryl)piperazine (BHAP) reverse transcriptase reverse transcriptase inhibition, and lipophilicity. Antiviral Research
inhibitors: structure–activity relationship and increased metabolic 31:87–94.
stability of novel substituted pyridine analogs. Journal of Medicinal
Kashman Y, Gustafson KR, Fuller RW, Cardellina II JH, McMahon
Chemistry 39:5267–5275.
JB, Currens MJ, Buckheit RW Jr, Hughes SH, Cragg GM &
Glynn SL & Yazdanian M (1998) In vitro blood–brain barrier per- Boyd MR (1992) The calanolides, a novel HIV-inhibitory class of
meability of nevirapine compared to other HIV antiretroviral coumarin derivatives from the tropical rainforest tree, Calophyllum
agents. Journal of Pharmaceutical Sciences 87:306–310. lanigerum. Journal of Medicinal Chemistry 35:2735–2743.
Goldman ME, Nunberg JH, O’Brien JA, Quintero JC, Schleif WA, Kelly TA, Proudfoot JR, McNeil DW, Patel UR, David E, Hargrave
Freund KF, Gaul SL, Saari WS, Wai JS, Hoffman JM, Anderson KD, Grob PM, Cardozo M, Agarwal A & Adams J (1995) Novel
PS, Hupe DJ, Emini EA & Stern AM (1991) Pyridinone deriva- non-nucleoside inhibitors of human immunodeficiency virus type

Antiviral Chemistry & Chemotherapy 10:6 311


OS Pedersen & EB Pedersen

1 reverse transcriptase. 5. 4-Substituted and 2,4-disubstituted Mai A, Artico M, Sbardella G, Quartarone S, Massa S, Loi AG, De
analogs of nevirapine. Journal of Medicinal Chemistry Montis A, Scintu F, Putzolu M & La Colla P (1997)
38:4839–4847. Dihydro(alkythio)(naphtylmethyl)oxopyrimidines: novel non-
Kelly TA, McNeil DW, Rose JM, David E, Shih C-K & Grob PM nucleoside reverse transcriptase inhibitors of the S-DABO series.
(1997) Novel non-nucleoside inhibitors of human immunodefi- Journal of Medicinal Chemistry 40:1447–1454.
ciency virus type 1 reverse transcriptase. 6. 2-Indol-3-yl- and 2- Mai A, Artico M, Sbardella G, Massa S, Novellino E, Greco G, Loi
azaindol-3-yl-dipyridodiazepinones. Journal of Medicinal Chemistry AG, Tramontano E, Marongiu ME & La Colla P (1999). 5-
40:2430–2433. Alkyl-2-(alkylthio)-6-(2,6-dihalophenylmethyl)-3,4-dihydropy-
Kim D-K, Kim Y-W, Gam J, Kim G, Lim J, Lee N, Kim H-T & rimidin-4(3H)-ones: novel potent and selective
Kim KH (1996) Synthesis and anti-HIV-1 activity of a series of dihydro-alkoxy-benzyl-oxopyrimidine derivatives. Journal of
1-(alkoxymethyl)-5-alkyl-6-(arylselenyl)uracils and -2-thiouracils. Medicinal Chemistry 42:619–627.
Journal of Heterocyclic Chemistry 33:1275–1283. Mao C, Vig R, Venkatachalam TK, Sudbeck EA & Uckun FM
Kleim J-P, Bender R, Billhardt U-M, Meichsner C, Riess G, Rîsner (1998) Structure-based design of N-[2-(1-piperidinylethyl)]-N′-
M, Winkler I & Paessens A (1993) Activity of a novel quinoxaline [2-(5-bromopyridyl)]-thiourea and N-[2-(1-piperazinylethyl)]-N′-
derivative against human imunnodeficiency virus type 1 reverse [2-(5-bromopyridyl)]-thiourea as potent non-nucleoside inhibitors
transcriptase and viral replication. Antimicrobial Agents and of HIV-1 reverse transcriptase. Bioorganic & Medicinal Chemistry
Chemotherapy 37:1659–1664. Letters 8:2213–2218.

Kleim J-P, Bender R, Kirsch R, Meichsner C, Paessens A, Rösner Massa S, Mai A, Artico M, Sbardella G, Tramontano E, Loi AG,
M, Rübsamen-Waigmann H, Kaiser R, Wichers M, Schneweis Scano P & La Colla P (1995) Synthesis and antiviral activity of
KE, Winkler I & Reiss G (1995) Preclinical evaluation of HBY new 3,4-dihydro-2-alkoxy-6-benzyl-4-oxopyrimidines (DABOs),
097, a new nonnucleoside reverse transcriptase inhibitor of human specific inhibitors of human immunodeficiency virus type 1.
immunodeficiency virus type 1 replication. Antimicrobial Agents Antiviral Chemistry & Chemotherapy 6:1–8.
and Chemotherapy 39:2253–2257. Merluzzi VJ, Hargrave KD, Labadia M, Grozinger K, Skoog M, Wu
Klunder JM, Hargrave KD, West M, Cullen E, Pal K, Behnke M, JC, Shih C-K, Eckner K, Hattox S, Adams J, Rosenthal AS,
Kapadia SR, McNeil DW, Wu JC, Chow GC & Adams J (1992) Faanes R, Eckner RJ, Koup RA & Sullivan JL (1990) Inhibition
Novel non-nucleoside inhibitors of HIV-1 reverse transcriptase. 2. of HIV-1 replication by a nonnucleoside reverse transcriptase
Tricyclic pyridobenzoxazepinones and dibenzoxazepinones. Journal inhibitor. Science 250:1411–1413.
of Medicinal Chemistry 35:1887–1897. Mertens A, Zilch H, König B, Schäfer W, Poll T, Kampe W, Seidel
Klunder JM, Hoermann M, Cywin CL, David E, Brickwood JR, H, Leiser U & Leinert H (1993) Selective non-nucleoside HIV-1
Schwartz R, Barringer KJ, Pauletti D, Shih C-K, Erickson DA, reverse transcriptase inhibitors. New 2,3-dihydrothiazolo[2,3-
Sorge CL, Joseph DP, Hattox SE, Adams J & Grob PM (1998) a]isoindol-5(9bH)-ones and related compounds with anti-HIV-1
Novel nonnucleoside inhibitor of HIV-1 reverse transcriptase. 7. activity. Journal of Medicinal Chemistry 36:2526–2536.
8-Arylethyldipyridodiazepinones as potenet broad-spectrum Miller V, de Bethune M-P, Kober A, Stürmer M, Hertogs K,
inhibitors of wild-type and mutant enzymes. Journal of Medicinal Pauwels R, Stoffels P & Staszewski S (1998) Patterns of resistance
Chemistry 41:2960–2971. and cross-resistance to human immunodeficiency virus type 1
Kroeger MB, Rouzer CA, Taneyhill LA, Smith NA, Hughes SH, reverse transcriptase inhibitors in patients treated with the nonnu-
Boyer PL, Janssen PAJ, Moereels H, Koymans L, Arnold E, Ding cleoside reverse transcriptase inhibitor loviride. Antimicrobial
J, Das K, Zhang W, Michejda CJ & Smith RH Jr (1995) Agents and Chemotherapy 42:3123–3129.
Molecular modelling studies of HIV-1 reverse transcriptase non- Miyasaka T, Tanaka H, Baba M, Hayakawa H, Walker RT, Balzarini
nucleoside inhibitors: total energy of complexation as a predictor J & De Clercq E (1989) A novel lead for specific anti-HIV-1
of drug placement and activity. Protein Science 4:2203–2222. agents: 1-[(2hydroxyethoxy)methyl]-6-(phenylthio)thymine.
Kukla MJ, Breslin HJ, Pauwels R, Fedde CL, Miranda M, Scott Journal of Medicinal Chemistry 32:2507–2509.
MK, Sherrill RG, Raeymaekers A, Van Gelder J, Andries K, Neamati N, Mazumder A, Zhao H, Sunder S, Burke TR Jr, Schultz
Janssen MAC, De Clercq E & Janssen PAJ (1991a) Synthesis and RJ & Pommier Y (1997) Diarylsulfones, a novel class of human
anti-HIV-1 activity of 4,5,6,7-tetrahydro-5-methylimidazo[4,5,1- immunodeficiency virus type 1 integrase inhibitors. Antimicrobial
jk][1,4]benzodiazepin-2(1H)-one (TIBO) derivatives. Journal of Agents and Chemotherapy 41:385–393.
Medicinal Chemistry 34:746–751.
Nugent AR, Schlachter ST, Murphy MJ, Cleek GJ, Poel TJ, Wishka
Kukla M J, Breslin HJ, Diamond CJ, Grous PP, Ho CY, Miranda DG, Graber DR, Yagi Y, Keiser BJ, Olmsted RA, Kopta LA,
M, Rodgers JD, Sherrill RG, De Clercq E, Pauwels R, Andries K, Swaney SM, Poppe SM, Morris J, Tarpley WG & Thomas RC
Moens LJ, Janssen MAC & Janssen PAJ (1991b) Synthesis and (1998) Pyrimidine thioethers: a novel class of HIV-1 reverse tran-
anti-HIV-1 activity of 4,5,6,7-tetrahydro-5-methylimidazo[4,5,1- scriptase inhibitors with activity against BHAP-resistant HIV.
jk][1,4]benzodiazepin-2(1H)-one (TIBO) derivatives. 2. Journal of Journal of Medicinal Chemistry 41:3793–3803.
Medicinal Chemistry 34:3187–3197.
Nunberg JH, Schleif WA, Boots EJ, O’Brien JA, Quintero JC,
Larder BA, Purifoy DJM, Powell KL & Darby G (1987). Site-spe- Hoffman JM, Emini EA & Goldman ME (1991) Viral resistance
cific mutagenesis of AIDS virus reverse transcriptase. Nature to human immunodeficiency virus type 1-specific pyridinone
327:716–717. reverse transcriptase inhibitors. Journal of Virology 65:4887–4892.
McMahon JB, Gulakowski RJ, Weislow OS, Shultz RJ, Narayanan Pauwels R, Andries K, Desmyter J, Schols D, Kukla MJ, Breslin HJ,
VL, Clanton DJ, Pedemonte R, Wassmundt FW, Buckheit RW Jr, Raeymaeckers A, Van Gelder J, Woestenborghs R, Heykants J,
Decker WD, White EL, Bader JP & Boyd MR (1993) Schellekens K, Janssen ACM, De Clercq E & Janssen PAJ (1990)
Diarylsulphones, a new chemical class of nonnucleoside antiviral Potent and selective inhibition of HIV-1 replication in vitro by a
inhibitors of human immunodeficiency virus type 1 reverse tran- novel series of TIBO derivatives. Nature 343:470–474.
scriptase. Antimicrobial Agents and Chemotherapy 37:754–760.
Pauwels R, Andries K, Debyser Z, Van Daele P, Schols D, Stoffels P,
Mai A, Artico M, Sbardella G, Massa S, Loi AG, Tramontano E, De Vreese K, Woestenborghs R, Vandamme A-M, Janssen CGM,
Scano P & La Colla P (1995) Synthesis and anti-HIV-1 activity Anne J, Cauwenbergh G, Desmyter J, Heykants J, Janssen MAC,
of thio analogues of dihydroalkoxybenzyloxopyrimidines. Journal of De Clercq E & Janssen PAJ (1993) Potent and highly selective
Medicinal Chemistry 38:3258–3263. human immunodeficiency virus type 1 (HIV-1) inhibition by a

312 ©1999 International Medical Press


The NNRTI boom

series of α-anilinophenylacetamide derivatives targeted at HIV-1 Sahlberg C, Noreen R, Engelhardt P, Högberg M, Kangasmetsä J,
reverse transcriptase. Proceedings of the National Academy of Sciences, Vrang L & Zhang H (1998) Synthesis and anti-HIV activities of
USA 90:1711–1715. urea-PETT analogs belonging to a new class of potent non-nucle-
Pauwels R, Andries K, Debyser Z, Kukla MJ, Schols D, Breslin HJ, oside HIV-1 reverse transcriptase inhibitors. Bioorganic and
Woestenborghs R, Desmyter J, Janssen MAC, De Clercq E & Medicinal Chemistry Letters 8:1511–1516.
Janssen PAJ (1994) New tetrahydroimidazo[4,5,1-jk][1,4]-benzo- Silvestri R, Artico M, Massa S, Stefancich G, Congeddu E, Putzolu
diazepin-2(1H)-one and thione derivates are potent inhibitors of M & La Colla P (1997). Sulfone derivatives with anti-HIV activi-
human immunodeficiency virus type 1 replication and are syner- ty. Il Farmaco 52:323–329.
gistic with 2′,3′-dideoxynucleoside analogs. Antimicrobial Agents
Sudbeck EA, Mao C, Vig R, Venkatachalam TK, Tuel-Ahlgren L &
and Chemotherapy 38:2863–2870.
Uckun FM (1998) Structure-based design of novel dihydroalkoxy-
Pontikis R, Benhida R, Aubertin A-M, Grierson DS & Monneret C benzyloxopyrimidine derivatives as potent nonnucleoside
(1997) Synthesis and anti-HIV activity of novel N-1 side chain- inhibitors of the human immunodeficiency virus reverse transcrip-
modified analogs of 1-[(2-hydroxyethoxy)methyl]-6- tase. Antimicrobial Agents and Chemotherapy 42: 3225–3233.
(phenylthio)thymine (HEPT). Journal of Medicinal Chemistry
Tanaka H, Baba M, Saito S, Miyasaka T, Takashima H, Sekiya K,
40:1845–1854.
Ubasawa M, Nitta I, Walker RT, Nakashima H & De Clercq E
Proudfoot JR, Hargrave KD, Kapadia SR, Patel UR, Grozinger KG, (1991) Specific anti-HIV-1 ‘acyclonucleosides’ which cannot be
McNeil DW, Cullen E, Cardozo M, Tong L, Kelly TA, Rose J, phosphorylated: synthesis of some deoxy analogues of 1-[(2-
David E, Mauldin SC, Fuchs VU, Vitous J, Hoermann M, hydroxyethoxy)methyl]-6-(phenylthio)thymine. Journal of
Klunder JM, Raghaven P, Skiles JW, Mui P, Richman DD, Medicinal Chemistry 34:1508–1511.
Sullivan JL, Shih C-K, Grob PM & Adams J (1995a) Novel non-
Tanaka H, Takashima H, Ubasawa M, Sekiya K, Nitta I, Baba M,
nucleoside inhibitors of human immunodeficiency virus type 1
Shigeta S, Walker RT, De Clercq E & Miyasaka T (1992a)
(HIV-1) reverse transcriptase. 4. 2-Substituted dipyridodi-
Structure–activity relationships of 1-[(2-hydroxyethoxy)methyl]-6-
azepinones as potent inhibitors of both wild-type and cysteine-181
(phenylthio)thymine analogues: effect of substitutions at the C-6
HIV-1 reverse transcriptase enzymes. Journal of Medicinal
phenyl ring and at the C-5 position on anti-HIV-1 activity.
Chemistry 38:4830–4838.
Journal of Medicinal Chemistry 35:337–345.
Proudfoot JR, Patel UR, Kapadia SR & Hargrave KD (1995b)
Tanaka H, Takashima H, Ubasawa H, Sekiya K, Nitta I, Baba M,
Novel non-nucleoside inhibitors of HIV-1 reverse transcriptase. 3.
Shigeta S, Walker RT, De Clercq E & Miyasaka T (1992b)
Dipyrido[2,3-b:2′,3′-e]diazepinones. Journal of Medicinal
Synthesis and antiviral activity of deoxy analogs of 1-[(2-hydrox-
Chemistry 38:1406–1410.
yethoxy)methyl]-6-(phenylthio)thymine (HEPT) as potent and
Ren J, Esnouf R, Hopkins A, Ross C, Jones Y, Stammers D & selective anti-HIV-1 agents. Journal of Medicinal Chemistry
Stuart D (1995) The structure of HIV-1 reverse transcriptase 35:4713–4719.
complexed with 9-chloro-TIBO: lessons for inhibitor design.
Tanaka H, Takashima H, Ubasawa M, Sekiya K, Inouye N, Baba M,
Structure 3:915–926.
Shigeta S, Walker RT, De Clercq E & Miyasaka T (1995)
Romero DL, Busso M, Tan C-K, Reusser F, Palmer JR, Poppe SM, Synthesis and antiviral activity of 6-benzyl analogs of 1-[(2-
Aristoff PA, Downey KM, So AG, Resnick L & Tarpley WG hydroxyethoxy)methyl]-6-(phenylthio)thymine (HEPT) as potent
(1991) Nonnucleoside reverse transcriptase inhibitors that potently and selective anti-HIV-1 agents. Journal of Medicinal Chemistry
and specifically block human immunodeficiency virus type 1 repli- 38:2860–2865.
cation. Proceedings of the National Academy of Sciences, USA Tantillo C, Ding J, Jacobo-Molina A, Nanni RG, Boyer PL, Hughes
88:8806–8810. SH, Pauwels R, Andries K, Janssen PAJ & Arnold E (1994)
Romero DL, Morge RA, Biles C, Berrios-Pena N, May PD, Palmer Locations of anti-AIDS drug binding sites and resistance muta-
JR, Johnson PD, Smith HW, Busso M, Tan C-K, Voorman RL, tions in the three-dimensional structure of HIV-1 reverse tran-
Reusser F, Althaus IW, Downey KM, So AG, Resnick L, Tarpley scriptase. Journal of Molecular Biology 243:369–387.
WG & Aristoff PA (1994) Discovery, synthesis, and bioactivity of Tucker TJ, Lumma WC & Culberson JC (1996) Development of
bis(heteroaryl)piperazines. 1. A novel class of non-nucleoside nonnucleoside HIV reverse transcriptase inhibitors. Methods in
HIV-1 reverse transcriptase inhibitors. Journal of Medicinal Enzymology 275:440–472.
Chemistry 37:999–1014.
Velázquez S, Alvarez R, Perez C, Gago F, De Clercq E, Balzarini J
Romero DL, Olmsted RA, Poel TJ, Morge RA, Biles C, Keiser BJ, & Camarasa M-J (1998) Regiospecific synthesis and anti-human
Kopta LA, Friis JM, Hosley JD, Stefanski KJ, Wishka DG, Evans immunodeficiency virus activity of novel 5-substituted N-alkylcar-
DB, Morris J, Stehle RG, Sharma SK, Yagi Y, Voorman RL, bamoyl and N,N-dialkyl carbamoyl 1,2,3-triazole-TSAO ana-
Adams WJ, Tarpley WG & Thomas RC (1996) Targeting delavir- logues. Antiviral Chemistry & Chemotherapy 9:481–489.
dine/atevirdine resistant HIV-1: identification of Vig R, Mao C, Venkatachalam TK, Tuel-Ahlgren L, Sudbeck EA &
(alkylamino)piperidine-containing bis(heteroaryl)piperazines as Uckun FM (1998) Rational design and synthesis of phenethyl-5-
broad spectrum HIV-1 reverse transcriptase inhibitors. Journal of bromopyridyl thiourea derivatives as potent non-nucleoside
Medicinal Chemistry 39:3769–3789. inhibitors of HIV reverse transcriptase. Bioorganic & Medicinal
Roth T, Morningstar ML, Boyer PL, Hughes SH, Buckheit RW Jr Chemistry 6:1789–1797.
& Michejda CJ (1997) Synthesis and biological activity of novel Wai JS, Williams TM, Bamberger DL, Fisher TE, Hoffman JM,
nonnucleoside inhibitors of HIV-1 reverse transcriptase. 2-Aryl- Hudcosky RJ, MacTough SC, Rooney CS, Saari WS, Thomas
substituted benzimidazoles. Journal of Medicinal Chemistry CM, Goldman ME, O’Brien JA, Emini EA, Nunberg JH,
40:4199–4207. Quintero JC, Schleif WA & Anderson PS (1993) Synthesis and
Saari WS, Wai JS, Fisher TE, Thomas CM, Hoffman JM, Rooney evaluation of 2-pyridinone derivatives as specific HIV-1 reverse
CS, Smith AM, Jones JH, Bamberger DL, Goldman ME, transcriptase Inhibitors. 3. Pyridyl and phenyl analogs of 3-
O’Brien JA, Nunberg JH, Quintero JC, Schleif WA, Emini EA & aminopyridin-2(1H)-one. Journal of Medicinal Chemistry
Anderson PS (1992) Synthesis and evaluation of 2-pyridinone 36:249–255.
derivatives as specific HIV-1-reverse transcriptase inhibitors. 2. Williams TM, Ciccarone TM, MacTough SC, Rooney CS, Balani
Analogs of 3-aminopyridin-2(1H)-one. Journal of Medicinal SK, Condra JH, Emini EA, Goldman ME, Greenlee WJ,
Chemistry 35:3792–3802. Kauffman LR, O’Brien JA, Sardana VV, Schleif WA, Theoharides

Antiviral Chemistry & Chemotherapy 10:6 313


OS Pedersen & EB Pedersen

AD & Anderson PS (1993) 5-Chloro-3-(phenylsulfonyl)indole-2- (1998) 1,1,3-Trioxo-2H,4H-thieno[3,4-e][1,2,4]thiadiazine


carboxamide: a novel, non-nucleoside inhibitor of HIV-1 reverse (TTD) derivatives: a new class of nonnucleoside human immun-
transcriptase. Journal of Medicinal Chemistry 36:1291–1294. odeficiency virus type 1 (HIV-1) reverse transcriptase inhibitors
with anti-HIV-1 activity. Antimicrobial Agents and Chemotherapy
Wishka DG, Graber DR, Kopta LA, Olmsted RA, Friis JM, Hosley
42:618–623.
JD, Adams WJ, Seest EP, Castle TM, Dolak LA, Keiser BJ, Yagi
Y, Jeganathan A, Schlachter ST, Murphy MJ, Cleek GJ, Nugent Young SD, Britcher SF, Tran LO, Payne LS, Lumma WC, Lyle TA,
RA, Poppe SM, Swaney SM, Han F, Watt W, White WL, Poel T- Huff JR, Anderson PS, Olsen DB, Carroll SS, Pettibone DJ,
J, Thomas RC, Voorman RL, Stefanski KJ, Stehle RG, Tarpley O’Brien JA, Ball RG, Balani SK, Lin JH, Chen I-W, Schleif WA,
WG & Morris J (1998a) (–)-6-Chloro-2-[(1-furo[2,3-c]pyridin- Sardana VV, Long WJ, Byrnes VW & Emini EA (1995a) L-
5-yl-ethyl)thio]-4-pyrimidinamine, PNU-142721, a new broad 743,726 (DMP-266): a novel, highly potent nonnucleoside
spectrum HIV-1 non-nucleoside reverse transcriptase inhibitor. inhibitor of the human immunodeficiency virus type 1 reverse
Journal of Medicinal Chemistry 41:1357–1360. transcriptase. Antimicrobial Agents and Chemotherapy
39:2602–2605.
Wishka DG, Graber DR, Seest EP, Dolak AD, Han F, Watt W &
Morris J (1998b) Stereoselective synthesis of furo[2,3-c]pyridine Young SD, Britcher SF, Payne LS, Tran LO & Lumma WC (1995b)
pyrimidine thioethers, a new class of potent HIV-1 non-nucleo- WO95/20389.
side reverse transcriptase inhibitors. Journal of Organic Chemistry Zembower DE, Liao S, Flavin MT, Xu Z-Q, Stup TL, Buckheit
63:7851–7859. RW Jr, Khilevich A, Mar AA & Sheinkman AK (1997) Structural
Witvrouw M, Arranz ME, Pannecouque C, Declerq R, Jonckheere analogues of the calanolide anti-HIV agents. Modification of the
H, Schmit J-C, Vandamme A-M, Diaz JA, Ingate ST, Desmyter J, trans-10,11-dimethyldihydropyran-12-ol ring (ring C). Journal of
Esnouf R, Van Meervelt L, Vega S, Balzarini J & De Clercq E Medicinal Chemistry 40:1005–1017.

Received 26 March 1999; accepted 20 July 1999

314 ©1999 International Medical Press

You might also like