Sie sind auf Seite 1von 72

HIV-1 NNRTIs: Structural Diversity,

Pharmacophore Similarity, and


Implications for Drug Design

Peng Zhan,1 Xuwang Chen,1 Dongyue Li,1 Zengjun Fang,1 Erik De Clercq,2
and Xinyong Liu1
1
Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, Jinan,
Shandong, P.R. China
2
Rega Institute for Medical Research, Katholieke Universiteit Leuven, Leuven, Belgium

Published online 26 April 2011 in Wiley Online Library (wileyonlinelibrary.com).


DOI 10.1002/med.20241
.

Abstract: Nonnucleoside reverse transcriptase inhibitors (NNRTIs) nowadays represent very potent and
most promising anti-AIDS agents that specically target the HIV-1 reverse transcriptase (RT). However,
the effectiveness of NNRTI drugs can be hampered by rapid emergence of drug-resistant viruses and severe
side effects upon long-term use. Therefore, there is an urgent need to develop novel, highly potent NNRTIs
with broad spectrum antiviral activity and improved pharmacokinetic properties, and more efcient
strategies that facilitate and shorten the drug discovery process would be extremely benecial. Fortunately,
the structural diversity of NNRTIs provided a wide space for novel lead discovery, and the pharmacophore
similarity of NNRTIs gave valuable hints for lead discovery and optimization. More importantly, with the
continued efforts in the development of computational tools and increased crystallographic information on
RT/NNRTI complexes, structure-based approaches using a combination of traditional medicinal chem-
istry, structural biology, and computational chemistry are being used increasingly in the design of NNRTIs.
First, this review covers two decades of research and development for various NNRTI families based on
their chemical scaffolds, and then describes the structural similarity of NNRTIs. We have attempted to
assemble a comprehensive overview of the general approaches in NNRTI lead discovery and optimization
reported in the literature during the last decade. The successful applications of medicinal chemistry stra-
tegies, crystallography, and computational tools for designing novel NNRTIs are highlighted. Future
directions for research are also outlined. & 2011 Wiley Periodicals, Inc. Med. Res. Rev., 33, No. S1, E1E72, 2013

Key words: HIV-1; RT; NNRTIs; drug resistance; drug design; medicinal chemistry; crystallography;
computational chemistry
Contract grant sponsor: National Natural Science Foundation of China (NSFC); Contract grant numbers: 30873133; 30772629;
30371686; Contract grant sponsor: Key Project of NSFC for International Cooperation; Contract grant number: 30910103908; Con-
tract grant sponsor: Research Fund for the Doctoral Program of Higher Education of China; Contract grant number: 070422083;
Contract grant sponsor: Independent Innovation Foundation of Shandong University (IIFSDU); Contract grant number:
2010GN044; Contract grant sponsor: Shangdong Postdoctoral Innovation Science Research Special Program; Contract grant number:
201002023; Contract grant sponsor: China Postdoctoral Science Foundation; Contract grant number: 20100481282.
Correspondence to: Xinyong Liu, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University,
44,West Culture Road, 250012, Jinan, Shandong, P.R. China, E-mail: xinyongl@sdu.edu.cn

Medicinal Research Reviews, 33, No. S1, E1E72, 2013


& 2011 Wiley Periodicals, Inc.
2E2 K K ZHAN
ZHAN
ETAL.
ETAL.

1. INTRODUCTION

Human immunodeciency virus type 1 (HIV-1) reverse transcriptase (RT) is a primary target
for antiretroviral chemotherapy. HIV-1 nonnucleoside reverse transcriptase inhibitors
(NNRTIs) are important in the drug combination therapies (namely, highly active anti-
retroviral therapy) currently used to treat HIV infection and AIDS due to their unique
antiviral activity, high specicity, and low toxicity.13 Nevertheless, drug resistance is still the
main reason of failure for their anti-HIV infection efcacy. Three NNRTIs clinically used
(efavirenz, nevirapine, and delavirdine) could effectively inhibit proliferation of the wild-type
(WT) HIV, but they are less effective against clinically important RT mutant viruses, such as
Y188C, Y181C, K103N, and L100I, which are involved in high-level resistance to most
current NNRTIs.2 Although the newly approved etravirine (Fig. 1) shows improved potency
against many drug resistance mutations, it lacks the convenience of once-daily dosing and is
also involved in cutaneous and hypersensitivity.4 In addition, efavirenz, as the most pre-
scribed NNRTI, is associated with side effects including dysfunction of central nervous
system and teratogenicity.5 Therefore, there is compelling need for the discovery of next
generation NNRTIs that possess an improved safety property with the convenience of once-
daily administration and high activity against drug-resistant mutant viruses.6
NNRTIs are targeted at a specic and allosteric binding site (an especially exible pocket
formed upon binding the inhibitor) situated about 10 A from the polymerase catalytic site
within the HIV-1 RT. One of the main issues in computational NNRTIs drug design is
incorporation of the inherent exibility of the NNRTIs binding pocket (NNIBP). This is
especially crucial in ligand binding process, when induced t can lead to structural re-
arrangement of RT. Although current computational methods (such as docking) deal with
exible ligands, it is very challenging to manage receptor exibility.79 To date, the advances
in the medicinal chemistry of HIV-1 NNRTIs have relied mostly on ligand-based design.
The exibility of the binding pocket in RT resulted in the structural and chemical diversity
of NNRTIs, providing a wide opportunity for novel scaffold discovery. Although they are
structurally quite diverse, all NNRTIs bind in the NNIBP in a similar conformation and in
identical fashion, providing valuable information on NNRTI lead discovery and optimization.
Furthermore, with the continued efforts in the development of computational tools and in-
creased structural information on RT, coordinated multidisciplinary effort involving tradi-
tional medicinal chemistry (SAR analysis, bioisosteric replacement, molecular hybridization,
scaffold hopping, multi-target/multivalent drug design, etc.), structural biology (crystal-
lography), and computational chemistry (molecular modeling) have proven to be powerful
strategies to handle exibility of the NNIBP and to identify novel NNRTIs with high antiviral
activity against WT and mutant viruses, and improved pharmacokinetic proles (Fig. 2).

Me
CN CN
Me
HN
N
N Me Me
N
N O F3C O N NH
N H Cl
Me S N N O
N
O Br
N N O
H O N O
Me CH3SO3H H NH2
H

1, Nevirapine 2, Delavirdine 3, Efavirenz 4, Etravirine/TMC-125

Figure 1. Chemical structures of NNRTIs approved by the FDA for HIV-1 treatment.

Medicinal Research Reviews DOI 10.1002/med


HIV-1 NNRTIs K E3

RT
Mutations in NNIBP Flexibility of NNIBP

NNRTIs
Pharmacophore
Drug Resisitance Structural Diversity Similarity

Necessity Feasibility Inspiration

NNRTIs Leads Discovery and Optimization

Medicinal Chemistry Crystallography Computational Chemistry

Bioisosterism Principle The Explanation of QSAR Molecular Docking


RT/NNRTI Interaction
Database Searching
Molecular Hybridization Concept
The Identification of Pharmacophore Modeling
Scaffold Hopping Tolerate Region in NNRTI
De Novo Methodologies
Prodrug Approach
Structural Basis Free Energy Perturbation
Multiple Ligands Design
QM/MM calculations
Multivalency Theory Strategies Tools

Multidisciplinary Coordination
Figure 2. The paradigm for NNRTI lead discovery and optimization. [Color figures can be viewed in the online issue, which is
available at wileyonlinelibrary.com.]

In this review article, we will rst describe the structural diversity and the pharmaco-
phore similarity of NNRTIs, as well as the implications for drug design. Then, the following
sections will emphasize recent research approaches in NNRTIs lead discovery and mod-
ications. Especially, the application of the medicinal chemistry strategies in the structural
modications will be discussed with the aid of illustrative examples.

2. STRUCTURAL DIVERSITY OF NNRTIS

Currently, more than 50 structurally diverse classes of compounds have been identied as
genuine NNRTIs (Table I), which specically suppress HIV-1 replication and are targeted at
the NNIBP.1017 These compounds can be divided into two classes: the rst generation
NNRTIs (exemplied by HEPT and TIBO), originally obtained by fortuitous discovery
(HEPT) or targeted screening, and showed a dramatic decrease of activity against single
point mutations in the NNIBP, and the second generation NNRTIs, discovered as a result of
comprehensive strategies involving computational chemistry (molecular modeling), struc-
ture-based rational drug design and synthesis, and biological and pharmacokinetic assays.
Generally, second generation NNRTIs tend to be more active against WT and broad-
spectrum HIV-1 drug-resistant strains than the rst generation compounds.
Based on the types of the chemical scaffolds, the structural classes of NNRTIs can
be devided into: multicyclic scaffolds, benzo-fused heterocyclic scaffolds, six-membered
Medicinal Research Reviews DOI 10.1002/med
4E4 K K ZHAN
ZHAN
ETAL.
ETAL.

Table I. Overview of the Structural Classes of NNRTIs

Chemical General Representative


scaffolds NNRTIs families abbreviation compounds References
18a,b
Multicyclic Tetrahydromidazo [4,5,1-jk][1,4] TIBO 5a, Tivirapine
scaffolds benzodizepin-2(1H)-one
19
Dipyridodiazepinone 1, Nevirapine
20
Indolyldipyridodiazepinone 6
21a,b
Thiazolobenzimidazole TBZ 7, NSC 625487
Thiazolo-iso-indolinone 8, BM 151.0836 22
23
Pyrrolobenzodiazepinone 9
24
Imidazodipyridodiazepine 10, UK-129,485
25
Arylpyrido(thio)diazepine 11, MEN 10979
26a,b
Pyrrolobenzoxazepinone 12
27
Indolobenzothiazepine 13
28
Imidazopyridazine 14
29
Trioxothienothiadiazine TTD 15, QM96521
30
Imidazoquinazoline 16
31
Pyrido[1,2a]indole 17, BCH-4989
32
Tricyclic benzothiophene 18, NSC-380292
33a,b
Coumarin 19, Calanolide A
33bd
20a, DCK
34
5H-Pyrrolo[1,2-b] PBTD 21
[1,2,5]benzothiadiazepine
35
Pyranoquinazolinones 22
36
Dihydro-1H-pyrido[3,2-b]indole 23, VRX-329747
37a,b
Benzo-fused Bisheteroarylpiperazine BHAP 2, Delavirdine
38
heterocycles Quinoxaline 24a, HBY 097
39a,b
Indole carboxamide 25, L-737,126
40
Benzothiadiazine 26,
41
Quinazolinone 27a
42
Benzoxazinone 3, Efavirenz
43
Benzothiadiazepine 28
44
Oxindoles 29a
45a
Quinolones 30
45b
31
45c
32
46
Benzimidazolones 33a
47
1-(2,6-Diuorobenzyl)-2-(2, BPBIs 34
6-diuorophenyl)benzimidazoles
48
Six-membered Diaryltriazine DATA 35
49a,b
heterocycles Diarylpyrimidine DAPY 4a, TMC125
50
core Diarylpyrazinone 36
51
Diarylpyridine 37
52
Diarylaniline 38
53
1-[(2-Hydroxyethoxy)methyl]-6- HEPT 39a, HEPT
(phenylthio)thymine derivatives
54
Alkoxy(arylthio)uracil 39b
55a,b
Dihydroalkoxybenzyloxypyrimidine DABO 40ac
56ac
Pyridinone 41a, L-697,661
57a,b
Benzylthiopyrimidine 42a, U-31355
(Pyrimidinethioethers)
58
Furopyridinylthiopyrimidinamine 42d, PNU-142721
59ac
Diarylamines (HetNHPhU) 43

Medicinal Research Reviews DOI 10.1002/med


HIV-1 NNRTIs
HIV-1 NNRTIs K K 5
E5

Table I. Continued

Chemical General Representative


scaffolds NNRTIs families abbreviation compounds References
60
Five-membered Thiadiazolyl dialkylcarbamate TDA 44, RD-4-2024
61a,b
heterocycles Highly substituted pyrrole 45a, 45b
62ac
core Azoles 46
63ac
Imidazole 47, S-1153
64
1,5-Diphenylpyrazole 48, PNU-32945
65
Diarylthiazolidinone 49a
66a,b
N-Aryl pyrrolidinone 50
67ah
Sulfanyltriazole/tetrazole 51a, VRX-480773
51b, RDEA806
68a,b
Thiazolidenebenzenesulfonamide 52ac
(Thio)amide a-Anilinophenylacetamido a-APA 53, Loviride 69

linker (a-APA, R89439)


70
containing Imidoyl thiourea ITU 54
71
scaffolds Thiocarboxanilide 55, UC-781
72ac
Phenethylthiazolylthiourea PETT 56a, Trovirdine
(LY 300046)
73
Quinoxalinylethylpyridylthiourea QXPT 56b, 6-FQXPT
74ad
Urea-PETT analogues 56c, MSC-204;
56d, MSC-372
(PETT-5)
75
O-Phthalimidoethyl- TC 57ac
N-arylthiocarbamate
76
Diphenyl Alkenyldiarylmethane ADAM 58a
77
scaffolds Tetrahydronaphthalene lignan 59a
78
Sulfonylbenzonitrile 60
79
Indazole 61a, 61b
80
Benzophenone 62a
81
Diaryl ether 63a
82
1-[2-(Diphenylmethoxy)ethyl]-2- DAMNI 64
methyl-5-nitroimidazole
83
Others Hexahydroxybiphenyl derivatives 65a, 65b
20 ,50 -Bis-O-(tert-butyldimethylsilyl)- TSAO 66, TSAO-m3T 84ac

30 -spiro-500 -(400 -amino-100 ,200 -


oxathiole-200 ,20 -
dioxide)pyrimidine

heterocyclic core scaffolds, ve-membered heterocyclic core scaffolds, (thio)amide linker


containing scaffolds, diphenyl scaffold, and so on. As shown in Table I, the prototype
leads or the promising candidates are selected as representatives in each class of NNRTIs
(Figs. 39).
As is well known, the current anti-AIDS drug research priorities are looking actively for
new drugs from the original leads and complex prescription for management of HIV infection
effectively, as well as against various mutant viral strains. In addition to the four approved
NNRTIs, a few more candidates are in clinical stage. Ideally, in order to be considered a bona
de candidate drug, a compound of interest must meet all the following criteria.10d (1) High
level of activity against key mutants (i.e. K103N and Y181C) resistant to other NNRTIs
without allowing breakthrough; (2) excellent oral bioavailability, and prolonged duration
Medicinal Research Reviews DOI 10.1002/med
6E6 K K ZHAN
ZHAN
ETAL.
ETAL.

Me H
Me N Me F
N
Me N N
N N N N
F
Cl N S
N
S
N N
N H O O
H Me Me N

5a, Tivirapine (R 86183) 1, (Nevirapine, BI-RG-587) 6 7, NSC 625487

Me Me Me N
N
Me N
O MeO N N N
S
N S

N N N
N N S
H Me Me
O
O
8, BM +51.0836 9 10, UK-129,485 11, MEN 10979

O N
Me N
O S N
O
Cl N
N NH
N

O N O
H Me O
12 13 14

O O N Me Me
Cl
S N
N O
S N
7b
N O O OMe
N O 2a
O
O HO S
CN
MeO
15, QM96521 16 17, BCH-4989 18, NSC-380292

O
Me O
N
Me Me Cl S N
O

N
O O
Me CF3
Cl
O O O 21 NH
O 2N
O O O
Me OH Me N O
H
Me Me O Me
Me 22
19, (+)-Calanolide A O O O
Me O O N
Me O
CN
Me O
N
Me O H
20a, DCK 23, VRX-329747

Figure 3. Chemical structures of NNRTIs with multicyclic scaffolds.

of potency; (3) signicantly decreased toxicity; and (4) easy of synthesis and formulation.
Obviously, the more requirements a selected drug meets, the more success it will have in the
clinical setting and in the global market as the miracle drug for the treatment of AIDS.
Medicinal Research Reviews DOI 10.1002/med
HIV-1 NNRTIs
HIV-1 NNRTIs K K 7
E7
Me
Me
Me Me
HN
N O O
N
O O
O MeO N S S
H O
SMe Cl NH N
Me S N N
O N S N S
N O H N O H
CH SO H H
H
2, Delavirdine (U-90152) 24a, HBY 097 25, L-737,126 26, NSC 287474

OEt
Me
O
N O O Me
FC
Cl Cl Cl S N Br
NH O
Me O
N N N N
H H H O H
27a 3, Efavirenz 28 29a

Me

Me

R Me Me
Cl CO R O S O
X N O S
O Me
Cl R MeO OH Cl N
N O
H O
R = (CH ) CHCH , nPent N
N O O
R = Et, Me, Allyl H H
30 31 32 33a

F F
F

F F F F
N N
Cl N F
O N Me N
N F F
H Me
O O
33b 34a 34b

Figure 4. Chemical structures of NNRTIs with benzo-fused heterocyclic scaffolds.

3. COMMON FEATURES OF NNRTIs

NNRTIs, although belonging to a wide range of structurally diverse scaffolds, contain many
ubiquitous fragments in their structures and possess a common pharmacophore model,
including an aromatic ring able to participate in p-p stacking interactions, amide or thioamide
moieties capable of hydrogen bonding, and one or more hydrocarbon-rich domain that
participate in hydrophobic interactions. In addition, the interactions with RT show similar
binding mode which is considered reminiscent of a buttery with one body and two
hydrophobic wings.

A. Ubiquitous Fragments (Motifs) in NNRTI Scaffolds


The existence of ubiquitous motifs in NNRTIs is very similar with the concept of privileged
structures85,86 in medicinal chemistry. For instance, para-substituted aniline motif exists in
multicyclic-typed NNRTIs (i.e. tivirapine) and many second generation NNRTIs with
Medicinal Research Reviews DOI 10.1002/med
8E8 K K ZHAN
ZHAN
ETAL.
ETAL.

N N N N
N N N N N
Me

Me Me Me Me
Me Me O N NH O N NH Me Me Me Me
N NH O N NH O NH
N
Br Me N O
N N
NH Me NH ON NH
35 4a, TMC 125 36 37 38

O O Me O
Cl
Me R'
HN HN Me HN
R'' R N
O N S O N S X N H
N
HO O Me O
Cl
O Me
40a, DABO (X = O) Me N O
Me Me 40b, S-DABO (X = S) H
39a, HEPT 39b 40c, N-DABO (X = N) 41a, L-697,661
Me
Cl Cl
Me
N N
Me H O
N
HN N S HN N S R
Het
O N N
42d, PNU-142721
H 43
42a, U-31355 (S)-(-)-enantiomer

Figure 5. Chemical structures of NNRTIs with six-membered heterocycles core scaffolds.

benzo-fused heterocyclic scaffolds (i.e. efavirenz, HBY 097), and (thio)amide linker con-
taining scaffolds (i.e. UC-781, trovirdine) (Fig. 10A). A diphenyl, substituted thiourea
functional group, a (thio)acetamide linker, and a nitrile-containing aromatic (not shown), are
also ubiquitous motifs in many NNRTI families (Fig. 10BD). Structural similarity is
remarkable among the NNRTIs with different chemical scaffolds (Fig. 10EG). Otherwise,
the usage of three different drug design strategies, bioisosteric principle, de novo design
approach, and pharmacophore-based virtual screening, resulted in the same quinolone
scaffold NNRTIs 3032, respectively, illustrating the structural similarity of NNRTIs.
Based on the fragment-based ligand discovery strategy,87 the ubiquitous motifs derived
from databases of known NNRTIs with high potency against WT and drug-resistant variants
of HIV-1 RT, high oral bioavailability and favorable pharmacokinetics, can be used as basic
fragments for the generation of privileged scaffolds libraries that are capable of providing
high-quality NNRTI hits. In fact, the computational tools of understanding bioactive mo-
lecular similarity has been employed in the colonization of the existing chemistry space for
each molecular scaffold and in association with de novo drug design45b,59a and classical
medicinal chemistry concepts (such as molecular hybridization) has assisted the rational
design of new drug-candidate prototypes against WT and key mutant viruses. The method
can be easy to put in place, and is fast enough to be iteratively applied to different sources of
drug-like molecules.

B. Molecular Modeling (From Buttery to Horseshoe)


On the basis of molecular modeling and X-ray crystallography investigations on nevirapine
(1), TIBO (5b) and 8b, Schafer et al. proposed a three-dimensional (3D) model describing the
Medicinal Research Reviews DOI 10.1002/med
HIV-1 NNRTIs
HIV-1 NNRTIs K K 9
E9
Me
Me
Me Me
N N
S N
OMe O
N O S
O Me O
N NH
Me O
Cl Cl
N
Me OMe Me N O
H
O O

44, RD4-2024 45a 45b

Cl Cl

N Cl
Me
R R
S N N
NH Cl
N
O NH O N
Me N N
O S
X N
Me Me CN O
X = CH, N
46 47, Capravirine (AG1549) 48, PNU-32945 49a

O NH
N N Cl
H O Me
N N S
N Br S R S
N
O N Me
O
R
MeO CN Me Me
O
52a: R = Cl. EC = 1.7 nM, SI > 15000
51a, VRX-480773, (R = SO NH ) 52b: R = Br. EC = 2.1 nM, SI > 12000
50 51b, RDEA806, (R = COOH) 52c: R = CN. EC = 1.8 nM, SI = 6100

Figure 6. Chemical structures of NNRTIs with five-membered heterocycles core scaffolds.


N
Me
N
Cl Me O Me

H O
N O Br
Cl Cl HN
Cl N
H Me S
Cl N NH
HN O N
H
Me NH S N S
H
53, Loviride (-APA, R89439) 54 55, UC-781 56a, Trovirdine (LY 300046)

Me Me
O O

N O
OH
O N
N O
X
F F F O

Br HN Br HN NC HN
N N N N S
S O O H
N N N
H H H 57a, TC-1, X = Cl, EC = 40 nM
56b, 6-FQXPT 56c, MSC-204 56d,MSC-372 57b, TC-2, X = Br, EC = 30 nM
(Active metabolite of PETT-4) (PETT-5) 57c, TC-3, X = I, EC = 20 nM

Figure 7. Chemical structures of NNRTIs with (thio)amide linker containing scaffolds.

Medicinal Research Reviews DOI 10.1002/med


10
E10 KK ZHAN
ZHANETAL.
ETAL.

COOMe COOMe MeO Br NC CN


MeO OMe S
MeO F
O
Br Br OMe O S Me O

CN
MeO OMe R
OMe NH N NH

58 a 59a 60 61a: R = Me, RT IC = 50 nM


MeO 61b: R = Et, RT IC = 25 nM
O

H Cl CN Me
N
O O O
C N
O N

A B O O NH
N ON
MeO
Me N Me Cl
62a 63a 64, RS1478

Figure 8. Chemical structures of NNRTIs with diphenyl scaffolds.

OMe O
Br Me
O HN

O COOMe O N

O COOMe Me2(t-Bu)SiO O
H 2N
O R
O OSi(t-Bu)Me2
OMe S
O
O
65a: R = H
65b: R = Br 66, TSAO-m3T

Figure 9. Chemical structures of NNRTIs with other scaffolds.

structural elements that were critical determinants for anti-HIV-1 activity (Fig. 11)88: a
central lipophilic domain (body) to which are attached two hydrophobic (normally a
benzene ring and an extended p system) moieties (wings), as in a buttery-like
orientation. Moreover, an additional lipophilic region, such as a carbonyl or thiocarbonyl
group, should be adjacent to the benzene ring. Other NNRTIs, such as a-APA, DABO, TBZ,
and TTD, are examples of agents conformationally related to nevirapine and TIBO. Taking
account the buttery-like conformation as determinant for anti-HIV-1 activity, many
novel families of NNRTIs were initially designed, such as 1-[2-(diarylmethoxy)ethyl]-2-me-
thyl-5-nitroimidazoles (DAMNIs).
Another buttery-like model was derived utilizing eight well-known NNRTIs, i.e. ne-
virapine (1), delavirdine (2), efavirenz (3), indole carboxamide (25), benzothiadiazine-1-oxide
(26), thiocarboxanilide (44), loviride (53), and trovirdine (56a).89 As shown in Figure 12, the
3D-pharmacophoric distance model proposed may be considered reminiscent of a butter-
y with a hydrophilic center (body: amide or thioamide groups) and two hydrophobic
outskirts (wings), one of which is usually substituted by a halogen atom.
Medicinal Research Reviews DOI 10.1002/med
HIV-1 NNRTIs K E11

A C NH
N
R
Br N
HN
N O (S) Cl N S
H
N N S
H
PETT (LY 300046) U-31355

B
Me Me
R R
N O
X Me S
X = C, O, SO N
Cl N
S N S
H
N
H
R 86183 NSC 287474

D Me Me N O NH
Cl
Me
Me N
N N
S S N
Cl

N N N MeO
S S
Me O
O O
8, BM +51.0836 11, MEN 10979 49a 50

N N Cl
H H
N N
Br N S O O
C
O O
SO NH
A B
MeO
Me N Me
51a, VRX-480773 62a

Me
E Me

Me Me Me
O N
Me O O
O S Me
Cl N Cl N Cl Me
O S
N N N S
H H H
R 86183 55, UC-781
33

F O
Me CN
Me NC

R R
F
Me Me N
O NH
N NH
HN NH
X N
N N
X = CH, N O
35 46 56d, MSC-372

Me
G Me
Me Br
Me
Me Me
N O
O O S O
O S
S Me O N O S Me O
O O S Me
Cl NH
NH MeO OH Cl N
CN
N O
N Me N
O H O N
H O H
O NH
25, L-737,126 45b 32 33a 60

Figure 10. Some ubiquitous fragments (motifs) and structural similarity in different NNRTI scaffolds: (A) para-substituted
aniline motif; (B) diphenyl scaffold; (C) substituted thiourea; (D) (thio)acetamide. [Color figures can be viewed in the online issue,
which is available at wileyonlinelibrary.com.]

Medicinal Research Reviews DOI 10.1002/med


E12 K ZHAN ETAL.

Me
Me

Me
N
N N N
Lipophilic
S region
Benzene ring Extended -system
N or aromatic ring
N N
H O S
Me 4.5--5A
N
H O CH
O 108--115
(Thio) carbonyl Methyl or other group
1, Nevirapine 5b, TIBO 8b or sulphonyl Linked at -system

Figure 11. The common three-dimensional model derived from the comparison of 1, 5b, and 8b (see text). [Color figures can
be viewed in the online issue, which is available at wileyonlinelibrary.com.]

It must be pointed out that these two buttery-like molecular models were derived from
ligand-based pharmaphore modeling. As a wealth of crystal structures of RT-NNRTI
complexes were reported,9092 the buttery-like conformation of NNRTIs with different
structural classes has been conrmed by crystallographic analysis. For instance, the rst-
generation NNRTI nevirapine and the recently approved etravirine can be superimposed in a
two-wing binding mode (compare the superposition gure of nevirapine with etravirine).12
Comparing the conformation of ten NNRTIs (nevirapine, delavirdine, efavirenz, a-APA,
BM121.1326, 9-Cl-TIBO, PETT-2, UC-781, MKC-442, and capravirine (S-1153)) when
bound with RT also disclosed a high degree of inhibitor overlap causing a buttery-like
conguration separated by a linker (body), where planes of the wings are separated by
1201 (compare two orthogonal views of ten NNRTIs in NNIBP).93
The RT-bound conformation of the ITU/DATA/DAPY NNRTIs resembles a U or
horseshoe, in contrast to the above-mentioned buttery-like shape.49b
There are important differences in the conformations of these two models (buttery
and horseshoe) and specic positioning within the NNIBP. For instance, the horseshoe
model could adapt to the plasticity and changes of the NNIBP better, which appear to be
critical for potency against WT and a wide range of drug-resistant mutant HIV-1 RTs.

C. 3D Pharmacophore Model of NNRTIs


Based on the crystallographic structures of RT-NNRTI complexes and computational stu-
dies, it has been demonstrated that the buttery-like or horseshoe NNRTIs also share
the common binding mode with NNIBP (the key interactions involved in binding sum-
marized in Table II)94,95 and 3D pharmacophore model (illustrated in Fig. 14),96,97 which is
the spatial arrangement of key chemical features: hydrophobic domain, hydrogen bond
acceptor, and donor. The hydrophobic domain lls the hydrophobic subpocket consisted of
the residues Y181, Y188, F227, and W229 (Fig. 13). The hydrogen bond acceptor and donor
form a key hydrogen bond with the backbone imino and carbonyl groups of K101 (or K103)
residue (directly or via a structural water molecule). The derived pharmacophore model can
facilitate insight into the detailed interactions between NNRTIs and RT, and to design the
next generaction of NNRTIs.
As is well known, perceiving a pharmacophore is the rst essential step towards un-
derstanding the receptorligand interaction. Other similar pharmacophore models for
NNRTIs were also obtained.45c,46,98100 As the pharmacophore-based virtual screening has
evolved into one of the well-established computational tools in rational drug design, this
technology could be used for obtaining new potentially active NNRTIs, lead optimization, as
well as combinatorial library focusing (will be described in detail in the following section).
Medicinal Research Reviews DOI 10.1002/med
HIV-1 NNRTIs K E13

Me
Me
HN
N
N
N
O O
N N H FC S
Cl O
Me S N N O Cl NH
O
N N N O
H O O N O
Me CH SO H H H
H

1, Nevirapine 2, Delavirdine 3, Ef avirenz 25, L-737,126

Me
Me N
N Cl Me O
S N
O O H
S N
O N
N Br HN
N
Cl Cl Cl S
N S HN O
H N
H
Me
26, NSC 287474 44, RD4-2024 53, Loviride 56a, Trovirdine

Molecular mechanics calculations

Geometrical optimization

Uniform sampling of low energy conformers

Mapping the essential structural components

Hydrophilic site

A
4.2 - 6.7 A 9.12 - 9.44 A

114-119

B C
4.25 - 7.6 A
Lipophilic site Lipophilic site

Figure 12. Schematic representation of the pharmacophoric distance map derived from eight well-known NNRTIs.89 In general,
NNRTIs consistoftwo hydrophobic moieties (wing I and wing II) connected by a polar central body. [Color figures can be viewed inthe
online issue, which is available at wileyonlinelibrary.com.]

D. General Types of NNIBP


There are subtle differences in the conformation of the NNIBP which mainly rely on the
NNRTI itself. The size and volume of the NNIBP are remarkably affected by the re-
positioning of b10 strand (residues 232234) and b11 strand (residues 239241) and the
moving of the P236 hairpin loop, as veried by crystallographic studies.101 Consequently,
it is commonly accepted that various NNRTI pockets can be divided into two discrete
binding modes. The rst mode is a small NNRTI (such as nevirapine)-type binding pocket
(PDB code 1VRT) in which there is a main chain hydrogen bond between the carbonyl

Medicinal Research Reviews DOI 10.1002/med


E14 K ZHAN ETAL.

Table II. The Key Interactions Involved in Binding to HIV RT and Denition of Pharmacophore
Points94,95

Pharmacophore
points Groups Interaction

Hydrogen bond OH, SH, NH Donor to K103 (1EP4, 1KLM)


donors or K101
Hydrogen bond (Thio)carbonyl, ester, sulfone O in ether, Acceptor to K103 (1EP4, 1KLM)
acceptors Pyridine N atom, N, O in aromatic or K101
5/6-membered rings
Hydrophobic Aliphatic or aromatic rings, Double and Hydrophobic interaction with Y188,
domains triple CC bonds, CF3, Aliphatic chains Y181, W229, F227, L100, L234,
V106, Y318

W229 Wing I Wing II


CN CN

Y188 HN

OH Me Me
Y181 Hydrophobic domain O N N
OH Me H
Cl N
Me Br
NH2
N
TMC125 (4)
Me N N
H Hydrogen bond donor
9-Cl-TIBO (5b)
S
Hydrogen bond acceptor O
H
N
K101

Figure 13. Positioning of NNRTI binding into NNIBP by mapping of the pharmacophore points (exemplified by 9-Cl-TIBO
and TMC125).96,97 [Color figures can be viewed in the online issue, which is available at wileyonlinelibrary.com.]

oxygen of P236 and the nitrogen of K103. The second mode is a larger NNRTI [such as
delavirdine (PDB code 1KLM) or 1-(2-hydroxyethoxymethyl)-6-(phenylthio) thymine
(HEPT)]-type NNIBP in which there is no hydrogen bond between P236 and K103.101,102
These bulkier NNRTIs bind with the RT in an expanded volume relative to smaller in-
hibitors and contain members that extend from the common NNIBP toward a exible
protein/solvent interface (Fig. 14), providing a valuable clue for drug design (will be
described in detail in the following section).103

4. THE NNRTI LEAD DISCOVERY APPROACHES

Lead discovery is one of the most considerable components in rational drug design, and
represents a major research area of medicinal chemistry today. There are two basic
Medicinal Research Reviews DOI 10.1002/med
HIV-1 NNRTIs K E15

Figure 14. Positioning of delavirdine in the NNIBP (PBD code:1KLM).101 [Color figures can be viewed in the online issue, which
is available at wileyonlinelibrary.com.]

approaches to obtain original leads, namely, de novo design and database screening.
Although de novo design is conceptually more attractive, high-throughput screening (HTS) of
compound collections is still the predominant approach in drug lead exploration, including
NNRTI lead discovery.

A. HTS
In the pharmaceutical industry, database HTS is frequently applied at the beginning of a
drug discovery program to detect novel hits.104,105 The combinatorial chemistry strategy
together with the HTS approach has permitted the discovery and optimization of novel anti-
HIV lead compounds.106 Some novel and diverse NNRTI leads discovered through HTS
using the cell-based HIV replication or enzymatic assay (presented in Fig. 15), such as
benzophenone (62),80 sulfanyltriazole (51c and 51d),67b,c sulfanyltetrazole (51e and 51f),67d
oxindole (29),44 tricyclic benzothiophene (18),32 N-aryl pyrrolidinone (50),66a and thiazoli-
denebenzenesulfonamide (52d and 52e),68a are of great value for the discovery of the next
generation NNRTI candidates.

B. Natural Products
Nature has always provided a wealth of drugs for various disease.107 Natural products with
promising anti-HIV properties have been revealed.108110 Over the past decade, signicant
progress has been made in the investigation of the medicinal plants as novel NNRTIs. These
plant-derived naturally occurring compounds belong to a broad range of diverse structural
families, e.g. avonoids, coumarins, terpenes, tannins, lignans, alkaloids, polysaccharides,
and naphtha(anthrax)quinones.107110 Most of them can serve as leads for developing novel
RT inhibitors that may be developed into anti-HIV candidates. This section described
representative examples of the successful use of chemical modication or molecular simpli-
cation strategy in the discovery of new leads from bioactive natural products.
Medicinal Research Reviews DOI 10.1002/med
E16 K ZHAN ETAL.

H N N N N Br
N H H
O O N N
Me N S Me N S
O
O O
Me

MeO
62a Me N Me 51c 51d
Me Me

OEt
N N Cl N N NO N
H H O
N N O
N N 7b
N S N S
Br OMe
Me Me O O 2a
O O
S
N
H MeO
51e 51f 29 18, NSC-380292
Me
IC (WT RT Pol) = 5 nM 2aS,7bS- and 2aR,7bR enantiomers
IC (WT RT Pol) = 5 nM IC (K103N) = 20 nM EC = 75 nM
EC = 1.24 M, SI > 800
IC (K103N) = 15 nM IC (K103N/Y181C) = 849 nM

O NH

N
Me O Me O
S N S N
Me S NO S NO
MeO O
Me N O Me N
O Me Me
Me Cl
50 52d 52e
EC = 125 nM EC = 8 5 nM EC = 48 nM

Figure 15. Examples of representative NNRTIs discovered from HTS.

OH
HO
O
OMe
HO
O O Br

OH
O O O COOMe
HO O
OH O COOMe
OH OH
O
HO O
O R
OH
HO O
OMe
Hexahydroxybiphenyl
O
HO 65a: R = H, EC 50 = 0.52 ug/mL, SI > 190
67, Ellagitannin punicalin 65b: R = Br, EC 50 = 0.23 ug/mL, SI > 480
OH

Figure 16. The discovery of hexahydroxybiphenyl NNRTIs.

As early as in 1990s, the hexahydroxybiphenyl compounds 65a and 65b (Fig. 16), derived
from tannins ellagitannin punicalin (67), were identied as a unique NNRTI family.83,111
With biologically active lignans, phenylpropanoid dimer compounds of plant origin, as a
starting point, synthetic tetrahydronaphthalene (THN) lignan derivatives 59a and 59b have
demonstrated potent anti-HIV activity (Fig. 17).77
Naturally occurring Calanolides112ad and Inophyllums (mainly isolated from
Calophyllum lanigerum33a and the Malaysian tree, Calophyllum inophyllum,113 respectively),
Medicinal Research Reviews DOI 10.1002/med
HIV-1 NNRTIs K E17

H H
MeO O
S S
MeO O
H H
OMe OMe

MeO OMe OMe


OMe OBz
59a 59b
EC50 = 0.15 M, SI = 161.6 EC 50 = 1.09 M, SI > 769

Figure 17. Lignans-based NNRTIs.

Me Me Me Me Me Me Me Me Me Me Me Me Me Me Me

O O O O O

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

Me OH Me OH Me OH Me OH Me OH
Me Me Me Me Me

Me Me Me Me Me Me Me
Me
O O Me O O

O O O O O O O O O O O O
Y
Cl
O Me X Me OH Me O
Me Me Me

Figure 18. The structures of Calanolides and Inophyllums. [Color figures can be viewed in the online issue, which is available
at wileyonlinelibrary.com.]

as well as their structural analogs,114 represent novel coumarin-based NNRTIs (Fig. 18). In
particular, (1)-calanolide A is unique in inhibiting HIV-1 Y181C isolates.112b The (1)-cala-
nolide A also displays additive to synergistic anti-HIV activity with other antiviral agents to
suppress mutant strains. Nevertheless, its low potency against HIV-1 is likely to account for the
limitation in clinical trials although it was proved to be well tolerated in phase Ia/Ib trials.112c,d
Thus, many structural modications on (1)-calanolide A with the aim of improving
inhibitory potency have been performed. Very recently, 10-chloromethyl-11-demethyl-12-
oxo-calanolide A, 19f, was reported, which shows druggable properties with higher oral
bioavailability (32.7% in the rat), tolerable toxicity upon a single oral dose administration in
mice, and interesting resistance proles (especially the feature of high inhibitory potency
against the WT and Y181C mutant HIV-1 at an EC50 5 7.4 and 0.46 nM, respectively).115
Taking the natural product, suksdorn (68) (Fig. 19), which was isolated from the fruit
of Lomatium suksdori,116 as a lead compound, further modications led to the discovery of
3,4-di-O-(S)-camphanoyl-(1)-cis-khellactone (20a, DCK) and its analogs,117130 such as
Medicinal Research Reviews DOI 10.1002/med
E18 K ZHAN ETAL.

R2
R1

O O O
Me
O O O Me O Me
Me Me
O O
Me O Me Me O O 20a, R1 = R 2 = H (DCK)
O O Me 20b, R1 = H, R 2 = Me
O
O Me 20c, R1 = CH 2OH, R2 = Me (HMDCK)
Me O
Me 20d, R1 = CH 2CN, R2 = Me
Me O
68, Suksdorfin

Figure 19. The novel suksdorfin derivatives as potent NNRTIs.

4-methyl-DCK (20b), 3-hydroxymethyl-4-methyl-DCK (20c, HMDCK), and (30 R,40 R)-3-


cyanomethyl-4-methyl-DCK (20d), which belong to a novel NNRTI family (Fig. 19). The
structural optimization of DCKs is still a research focus in the current anti-HIV drug
discovery eld.131
Especially, DCK derivative 20d, which contains a cyano group, not only exhibited
promising potency against WT HIV-1 in H-9 lymphocytes assay but also showed improved
drug resistance proles. It also has mediocre oral bioavailability, mediocre cell permeability,
and low systemic clearance.130 Its design was based upon the computational studies,132
coupled with the fact that a cyano group demonstrates good metabolic stability under most
conditions. This cyano group is also a good H-bond acceptor and can favorably interact with
key residues on the NNIBP, which are determinant requisites for the afnity of several
NNRTIs.133 These results suggest that natural products combined with rational drug design-
based modications and analogs synthesis could provide promising lead molecules for the
development as clinical trial candidates.
In summary, structural modication (simplication) of a natural product has afforded a
practical way in theory to nd novel NNRTI leads with promising antiviral potency and less
toxicity. However, identifying novel NNRTIs by random screening of natural product
libraries and then optimizing them by systematic chemical modications are highly time- and
resource consuming. Therefore, faster and more efcient approaches that facilitate and
shorten the novel NNRTI discovery process would be extremely benecial.

C. Virtual Screening
Virtual screening is becoming a major source in the discovery of hit- and lead-com-
pounds.134136 Several approaches of virtual screening, such as molecular docking and
pharmacophore-based searching algorithms, are gaining acceptance and are applied to dis-
cover more potent NNRTIs. Numerous related software tools have been developed. In this
section, we will describe several examples of different virtual screening approaches for
NNRTI lead discovery.

1. Docking-Based Virtual Screening


Docking-based virtual screening is a widely used computational tool in hit identication and
lead optimization, which dock small molecules into the binding sites of macromolecular
targets and score the targetligand binding afnity.137
Sangma et al. used a docking and neural network combined approach to screen active
compounds targeting HIV-1 RT and PR from the Thai medicinal plants database.138 The
Medicinal Research Reviews DOI 10.1002/med
HIV-1 NNRTIs K E19

results show that this combined approach allows to execute the successive screening and to
minimize the analyzing step from the docking and scoring procedure.
In a recent report, virtual screening of the Maybridge library of 70,000 compounds was
carried out using a similarity lter, docking, and molecular mechanics-generalized Born/
surface area postprocessing to search for novel NNRTIs. Although known NNRTIs were
retrieved, purchase and evaluation of top-scoring representative compounds from the library
failed to obtain any active HIV inhibitors.62a
Hierarchical lters, from simple structural and PK lead lters (e.g. molecular weight,
number of rotatable bonds, and number of hydrogen bond acceptors/donors) to high-
throughput rigid docking were applied to seek the National Cancer Institute (NCI) database
for HIV-1 RT hits. Finally, in vitro assay, a novel molecule NIC 14129 (69), was identied as
the most efcient hit for HIV-1 RT inhibition (Fig. 20).139
To discover NNRTIs that are effective against both WT virus and Y181C mutations,
docking-based virtual screening using standard-precision scoring and the more computa-
tional intensive extra-precision scoring with the Glide program was carried out using three
RT proteins (PDB entries 1RT4, 2BE2, and 1JLA) and more than 2,000,000 commercially
available compounds. The structures 1RT4 and 2BE2 are for WT RT with different con-
formations of Y181, while the protein 1JLA contains the Y181C mutant. Among the
purchased compounds, 70 shows moderate inhibitory activity against both the WT and
Y181C-mutant HIV-1 in low micromolar level, while 71 has 7.5 mM activity against the

O CN
Me H N S
N N
N Me
4 NH O
O N
O N N
O
Me HO NH N
69, NIC 14129
IC = 18.9 M Me O N O
H
Nevirapine IC = 4.20 M 70 71 72

S N Br
O H
N N S
S
MeO HN
O N HN
N
H
73 S
EC = 374 nM O
MeO HN
SI > 446 OMe 74a Me
HN
EC = 3.0 M
Me
O
S
Cl OMe Me
Me HN 74c
Me Me HN EC = 168 nM
O S SI > 345
N
O O
MeO OH Cl 74b Me
EC = 2.6 M
O

32

H
S N

NH
O
S
O
O
75, NAPETA

Figure 20. The structures of NNRTIs obtained using virtual screening.

Medicinal Research Reviews DOI 10.1002/med


E20 K ZHAN ETAL.

Y181C mutant, and 72 is a 4.8 mM NNRTI toward the WT virus. This research illustrates a
viable protocol to uncover anti-HIV agents with improved resistance properties.140
The virtual screening by docking a large compounds library (Leadquest 3) into two RTs
(PDB entries 1FK9 and 1DTQ) led to initial identication of several potential compounds
(i.e. 73, 74a, and 74b) with effects against the RT-associated DNA polymerase activities and
the HIV-1 pseudovirus infection in susceptible cells. Moreover, combining functional groups
of two structurally related inhibitors 74a and 74b resulted in a more potent inhibitor 74c that
inhibited the RNA-dependent DNA polymerase activity of recombinant HIV-1 RT with an
IC50 value of about 510 nM. This compound also suppressed the infection of HIV-1 pseu-
dovirus in human lymphocytes with an EC50 value of about 168 nM.141 It should be noticed
that these structures have high similarity to the known PETT family.
Through docking-based virtual screening of a large library of 50,000 compounds, N-f2-[4-
(aminosulfonyl)phenyl]ethylg-2-(2-thienyl)acetamide (NAPETA, 75) was identied as novel
HIV-1 RT inhibitor, which interfered with the formation of the RT-DNA complex. This
mechanism is different from that of the classical NNRTIs used for treating HIV infection.142
2. Pharmacophore-Based 3D Database Searching
Once a bona de drug against a known target has been identied, computational approaches,
such as 3D pharmacophore-based database searching, can play a pivotal role in the discovery
of novel leads with different chemical scaffolds. The large number of successful applications
of 3D pharmacophore-based searching in medicinal chemistry clearly demonstrate its utility
in the modern drug discovery paradigm.143,144
As early as 1998, a 3D stereoelectronic pharmacophore derived from a 3D-QSAR
investigation was reported as the foundation of the development of a two-phase data-mining
methodology to seek novel NNRTI leads.145
In 2007, a 3D structure-based pharmacophore model for DAPY NNRTIs was used to
screen large chemical databases (CAP Complete 2004 and Derwent-WDI2005). The obtained
hits were further ltered by taking into account the tness score and by using Lipinskis rule
of ve in addition to molecular docking studies (Glide program). Finally, six compounds
were selected for assaying of their inhibitory potency against HIV-1 RT. In particular,
compound 32, which belongs to the quinolin-2(1H)-one scaffold, showed an IC50 value
(200 nM) comparable to that of nevirapine (180 nM).45c
It must be pointed out that pharmacophore-based virtual screening by itself may not
always guarantee a successful identication of potent inhibitors against HIV-1 RT and may
therefore require additional complementary approaches. The hierarchical multiple-lter
database searching strategy combined many cheminformatic tools is an attractive strategy in
drug lead exploration. Two efcient approaches by using hierarchical database screenings are
presented in Figure 21.99,146
Although the applicability of virtual screening methodology has been well established, it
seems that there are still no successful projects of NNRTIs drug discovery in which virtual
screening has been the pivotal contributor.

D. De Novo Design of NNRTIs


De novo drug design is an active area of structure-based rational design approach, which
relies only on prior knowledge of the 3D structure of target to design entirely new lead
compounds with desired properties.147 Two categories can be divided for de novo design:
atom-based and fragment-based approaches. The fragment-based method shows more
attractive as a virtual structure can be easily constructed from combinatorial building blocks.
Since the discovery of NNRTIs, crystal structures of WT and mutated RT have been
used extensively in the de novo design of novel NNRTIs.148 However, the considerable
Medicinal Research Reviews DOI 10.1002/med
HIV-1 NNRTIs K E21

Figure 21. Flowchart representation of the hierarchical multiple-filter database searching strategy. (A) Hierarchical database
screening combined sequentially a pharmacophore model, multiple-conformation rigid docking, solvation docking, and mole-
cular mechanics-Poisson--Boltzmann/surface area (MM-PB/SA) sequentially.99 (B) Virtual screening by pre-filter pharmacophore
hypotheses and docking.146 [Color figures can be viewed in the online issue, which is available at wileyonlinelibrary.com.]

Four RT crystal structures: WT, Y181C, K103N, L100I + K103N

Search in each structures for fragments that bind to NNRTIs binding site

Identified fragments that appears in all structures

Build a molucule by linking cross reacting fragments

Minimize and dock final molecule to each structure separately

Synthesize and test various derivatives based on the designed pharmacophore

I O
76

Figure 22. Outline of the design process of new NNRTIs.149 [Color figures can be viewed in the online issue, which is available
at wileyonlinelibrary.com.]

conformational exibility of RT has complicated the traditional structure-based de novo drug


design approach for the discovery of novel NNRTIs, because no one would ever predict the
formation of the allosteric binding pocket just looking for the unbound structure.
In 2007, a successful de novo drug discovery of NNRTI was reported (Fig. 22).149 First,
the authors searched small fragments capable of interacting with each one of four RT pro-
teins (WT and Y181C, K103N, L100I1K103N mutations), using the Ludi module (Cerius2
LUDI user guide; San Diego, CA, 2003), a program widely used to dock small molecular
Medicinal Research Reviews DOI 10.1002/med
E22 K ZHAN ETAL.

fragments in target binding sites for the de novo design approach. Then, these fragments were
combined to build a core scaffold. Out of 27 synthesized compounds, four had low micro-
mole activity against the DNA polymerase activity of RT (IC50 value o10 mM). The most
potent compound 76 shows high RT inhibitory activity with (IC50 value: about 3.5 mM),
meanwhile, its potency against clinically relevant drug resistance mutants was more ef-
ciently than that of nevirapine.
Factors important for tight binding of inhibitors and resilience to mutations were elu-
cidated based on the detailed analysis of a broad range of crystal structures of RT in
complexing with various NNRTIs together with data on drug-resistant HIV-1 mutations.
These information were used to the structure-based (fragment-based methods) discovery of a
novel series of quinolone NNRTIs 31ad. Several of them retain high activity against the
Y181C and L100I mutated RT (Table III). Crystal structure analysis conrms the predicted
binding modes.45b
The ligand-growing program BOMB (Biochemical and Organic Model Builder,
Jorgensen, W. L. BOMB, v 2.5; Yale University: New Haven, CT, 2004.) is used to construct
molecules by adding layers of substituents to a scaffold that is isolated or that has been
placed in a binding site. De novo design of NNRTIs with the program BOMB based on
diarylamines (HetNHPhU) scaffold resulted in the identication of compound 77
(Het 5 2-thiazolyl and U 5 dimethylallyloxy) as a promising original hit (Fig. 23). As
described below, this compound was further optimized to multiple highly potent NNRTIs.59a

Table III. Anti-RT (WT and Mutant) Activity (IC50, mM) and Antiviral Activity (EC50, mM) of
Quinolone Derivatives

Cl X
Cl R 31a: R = Pr, X = O
31b: R = i-Pr, X = O
N O 31c: R = Pr, X = S
H
N O 31d: R = Pr, X = SO
Popular fragment H

Nevirapine-resistant
Compd RTWT RTY181C RTL100I HIV-1IIIB HIV-1

31a 0.71 1.13 1.36 0.035 0.607


31b 0.63 2.50 1.96 0.051 0.710
31c 0.24 0.86 1.61 0.100 0.368
31d 0.38 1.26 2.38 0.242 2.210
Nevirapine 0.30 4100 4100

Me
U
Me O

BOMB
S
Het Ph
N N N
H H
Het-NH-Ph-U 77: EC50 = 10,000 nM

Figure 23. The discovery of diarylamine NNRTIs using BOMB. [Color figures can be viewed in the online issue, which is
available at wileyonlinelibrary.com.]

Medicinal Research Reviews DOI 10.1002/med


HIV-1 NNRTIs K E23

Because the Gordian knot of most de novo design methods may be synthetic in-
accessibility, an advance de novo design program SYNOPSIS providing a synthesis route for
each generated molecule has been developed.150 This novel program has been successfully
applied to design several NNRTIs showing HIV inhibitory activity in vitro.150

5. OPTIMIZATION STRATEGIES OF NNRTI LEADS

In the NNRTI modication process, the combination of traditional medicinal chemistry,


computational chemistry, and crystallographic studies led to the identication of numerous
promising candidates, which were active against both WT and drug-resistant variants of RT.

A. Crystallographic Studies and Implication for Modification of NNRTIS


Nowadays, the course of drug development for the treatment of AIDS is being revolutionized
by high-resolution crystallographic structures of keyproteins in the HIV-l life cycle. Espe-
cially, crystal structures of WT and drug-resistant mutants of HIV-1 RT have been used
extensively in the design of novel NNRTIs. The structural information reveals the important
interaction mode of inhibitor, indicating the essential aspects determining their binding af-
nity and generates new ideas about opportunities for improving drug efcacy.151
1. The Identication of Tolerant Region in NNRTIS or NNIBP
The identication of the tolerated region in the lead compounds using the structure biology
information of HIV-1 RT/NNRTI complexes is a premise that allows rational optimization.
In view of the plasticity of NNIBP, a composite binding pocket model, integrating all
available crystallographic information on the NNIBP of HIV-1 RT,152154 was used to
rationally design potent inhibitors with improved proles against drug-resistance mutants of
HIV-1 RT. This composite binding pocket was successfully demonstrated as a powerful
tool to handle exibility of the NNIBP and to identify specic domains (the potentially
usable space) for structural optimization of the inhibitors. Based on the obtained results,
several novel, highly potent NNRTIs with broad-spectrum antiviral activity were identi-
ed.155159
On the basis of the crystallographic studies on binding of larger NNRTIs, the P236
hairpin loop in RT is closer to the apo conformation, forming a more open pocket; this
represented a tolerant region for introducing additional groups to the binding NNRTIs
(described in the earlier section).160
Additionally, two relatively inexible residues at the pocket entrance, V179 and L100,
manage the openness of the entrance and form the fortress of the inhibitor-unbinding channel,
which therefore was regarded as another attracting tolerant region and could be exploited by
diverse groups, giving an additional contribution to the generation of the allosteric NNIBP.161
For instance, the 5,6-positions in the central pyrimidine ring of DAPY NNRTIs162,163 and the
5-position in the pyrimidone ring of HEPT NNRTIs164 are well accommodated in a small
cavity consisting of V179 side chain, respectively, which has been validated by crystallographic
studies (Fig. 24) and further chemical modications (Figs. 25 and 26).
In summary, the plasticity of NNIBP resulted in the discovery and/or validation of the
tolerated region in NNRTIs or NNIBP using crystallographic studies, which provides a
broad space for the discovery of new generations of NNRTIs. It seems possible to exploit the
tolerated region of NNIBP to gain specic proteinligand interactions to accommodate
substantial modications of the NNRTI molecule which improve its pharmacokinetic
properties or to construct multi-target ligand165,166 by incorporating another bioactive
moiety without a signicant deprivation of binding afnity.
Medicinal Research Reviews DOI 10.1002/med
E24 K ZHAN ETAL.

Figure 24. TMC125 (A) and TNK-6123 (compound 39e) (B) in the NNIBP (PDB code for TMC125 and TNK-6123: 3MEC,
1C1C).151,164 [Color figures can be viewed in the online issue, which is available at wileyonlinelibrary.com.]

CN CN CN CN CN CN Me CN Me CN

Me Me Me Me Me Me Me Me Me Me
O N NH O N NH O N NH O N NH O N NH

N N N N N
Br HN N
NH NH N NH
HO
V179 V179 V179 V179 V179
4d 4e 4f 4g
4c
EC < 10 nM EC < 10 nM EC < 10 nM EC < 10 nM (WT)
(WT, L100I+K103N) (WT, L100I+K103N) (WT, L100I+K103N) 10 nM < EC < 100 nM
(100I+K103N)

Figure 25. Diverse bicyclic heterocycle DAPYanalogs.163 [Color figures can be viewed in the online issue, which is available at
wileyonlinelibrary.com.]

O V179 O Me V179 O Me V179 O Me V179


Me
HN HN Me HN Me HN Me

O N S O N O N O N S

O O O O

HO
Me Me
39a, HEPT 39c, TNK-651 39d, MKC-442 39e, TNK-6123

Figure 26. HEPT and the second generation pyrimidine diones. [Color figures can be viewed in the online issue, which is
available at wileyonlinelibrary.com.]

2. Structural Biology Explanation of NNRTIs With Resilience to Mutations


We gained a thorough understanding of the structural biology of HIV-1 RT/NNRTI com-
plexes and the data on drug-resistance mutations, inherent conformational exibility, and
positional adaptability of NNRTIs,72c,167169 forming extensive hydrogen bonding170 or
Medicinal Research Reviews DOI 10.1002/med
HIV-1 NNRTIs K E25

other key interactions171 with the main chain. This has allowed us to target highly conserved
residues in the HIV-1 RT167,172 and to exploit unconventional mechanisms for NNRTI-
mediated inhibition of RT.173175 These are important factors for the design of inhibitors
with improved resilience to mutations.
Therefore, only using these information as a staring point would the application of the
following described medicinal chemistry strategies be highly effective.

B. Medicinal Chemistry Strategies in the Modification of NNRTIs


In this section, successful applications of medicinal chemistry strategies (bioisosteric re-
placement, molecular hybridization, scaffold hopping, design of multiple ligands, and mul-
tivalent drug design) for NNRTI modications are reviewed.

1. Bioisosterism Principle: Me-Too


Bioisosteric replacement is an excellent tool for lead optimization to produce the desired
potency, selectivity, and the required ADME proles for a marketable drug.176ac The use of
bioisosteres in NNRTI lead optimization is illustrated by some recent examples from the
literature.
Case 1. Diarylazine-based NNRTIs
Taking a-anilinophenylacetamido (a-APA) as the original lead compound,69a further
optimization carried out by Janssen and his colleagues led to the discovery of the imino-
thiourea (ITU) analogs (i.e. 54, IC50(LAI) 5 3 nM)70 to the diaryltriazine (DATA) analogs (i.e.
35, IC50(LAI) 5 0.3 nM)48 and the diarylpyrimidine (DAPY) analogs (i.e. 4c, TMC120),
successively.49 The rst DAPY compound, dapivirine (TMC120), is being pursued for its
microbicidal potential. The second DAPY compound etravirine (TMC125) (Fig. 27),177
which has been recently approved (Intelences) for clinical use, and the third DAPY com-
pound (R278474) (35) corresponds to rilpivirine (TMC278) (Fig. 27),178 which is expected to
be approved soon for clinical use. Thus, the structural modications of the DAPY series are
still a research hotspot.179182 Recently, using the isosteric replacement strategy, pyr-
azinone,50 diarylpyridine,51 and diarylaniline compounds52 were discoveried as novel scaf-
folds of potent NNRTIs, active against both WT and drug-resistant HIV-1 strains.
Especially, the diarylpyridine 37 showed low nanomolar EC50 values and high SI values
(410,000) against HIV-1 IIIB, NL4-3, and RTMDR1.51 The diarylaniline 38 inhibited WT
HIV-1 strains with low nanomolar EC50 values of 0.0030.032 mM and inhibited several
drug-resistant strains with EC50 values in 0.0050.604 mM range.52 In addition, the pyridine
or substituted aniline ring replacement offers a more convenient and shorter synthetic route,
using inexpensive commercial reagents, compared to the synthesis of DAPY derivatives
TMC125 and TMC278.
Case 2. Efavirenz analogs
Efavirenz (DMP 266, L-743,726, SustivaTM) is one of the four approved NNRTI drugs
for the treatment of HIV-1 infection, which belongs to the series of benzoxazinones, struc-
turally related to the rst subset of NNRTIs developed by Merck and based on a quina-
zolinone skeleton. The development of quinazolinone derivatives started from the lead
L-608,788 (78), unstable under screening conditions (Fig. 28).41 Researchers have continued
to develop additional potent NNRTIs with other ring systems, structurally related to efa-
virenz, such as dihydroquinazolinone skeleton (DPC 961, DPC 963, DPC 082, and DPC
083),183187 cis-3-alkylbenzoxazepinone skeleton (79a, 79b),188 and benzothiadiazine skeleton
(80).189 They showed antiviral activity in the nanomolar range. Based on the structural
features of efavirenz (3), HBY-097 (24), and oxindole 29b, a quinolone scaffold (30b) was
obtained as a good surrogate of efavirenz.45a
Medicinal Research Reviews DOI 10.1002/med
E26 K ZHAN ETAL.

N N N N
Me Me

Cl Cl Cl Cl Me Me Me Me
H H
N NH N NH N NH HN N NH

NH S NH N N N N
54 N
35 4c, TMC 120
(1998)

N N N N N N N N

Me Me Me Me Me Me Me Me

O N NH O N NH O N NH O NH

N NH
Br Me N O NH ON

NH Me
36 37 38
4a, TMC 125 Pyrazinones (2005) Diarylpyridine (2009) Diarylaniline (2010)
(1999)
IC (LAI) = 6 nM EC = 1.4 nM; SI = 22,700
IC (K103N + Y181C) = 63 nM (HIV-1 ,MT-2 cell)
EC = 0.68 nM; SI = 13,206
N (HIV-1 , TZM-b1 cell)
EC = 0.96 nM; SI = 9,354
N (HIV-1 ,TZM-b1 cell)

Me Me
HN N NH

4b, TMC 278


(2001)

Figure 27. The chemical evolution of diarylazine-based NNRTIs (Arrows represent development timelines). [Color figures can
be viewed in the online issue, which is available at wileyonlinelibrary.com.]

Case 3. Capravirineanalogs
Imidazole compound capravirine (47, formerly known as AG1549 and S-1153) displayed
potency at subnanomolar concentrations in vitro against a broad-spectrum of HIV-1 strains
(EC50 values: 0.710.3 nM).63ac Unfortunately, capravirines development was discontinued
due to poor results in clinical trials and complex interactions with other anti-HIV ther-
apies.63ac In the continued modication of capravirine, novel arylthio isopropyl pyr-
idinylmethylpyrrolemethanol (AThP) derivatives were found to be active to block the
replication of HIV-1 in infected cells in the concentration range of 0.00853 mM.190ab Com-
pounds 81a and 81b were the most two active derivatives in cell-based assays with similar
inhibitory effects and SI to that of capravirine (EC50 5 8, 7, and 3 nM; SI 5 6,250, 16,357, and
7,000, respectively).190ab Compound 81b retained impressive activities against clinically
important drug-resistant RT carrying K103N, Y181I, and L100I mutations.190b 5-Aryloxy
imidazole 82, achieved by removing a metabolically vulnerable pyridine ring and replacing a
vulnerable sulfur atom with oxygen, illustrates the impressive overall prole against both WT
RT and the clinically relevant mutations K103N and Y181C and remarkable improvement in
potency and metabolic stability over capravirine.191,192 Recently, a new series of S-1153 analogs
Medicinal Research Reviews DOI 10.1002/med
HIV-1 NNRTIs K E27

OEt H

Cl Cl Cl OEt
NMe NMe NH H

O
N S N O N O Cl
H H H
27a O
78, L-608,788 27b
IC = 12 nM N
H
29b
Me Me

O O
FC FC FC
Cl MeO N
NH NH O
X X SMe

N O N O N O
H H H N O
H
27c, DPC 961 (X = 6-Cl) 27e, DPC 083 (X = 6-Cl) 3, Efavirenz
24, HBY-097
27d, DPC 963 (X = 5,6-diF) 27f, DPC 082 (X = 5,6-diF)

Me

Efavirenz: EC = 38 nM Me R O
DPC961: EC = 32 nM
DPC083: EC = 23 nM Cl Br
NH
OEt
FC FC S O
N N O
Cl O Cl O H O H
Me Me 80 30b
EC = 182 nM
N N
H O H O
79a 79b
EC = 82 nM EC = 46 nM

Figure 28. The discovery and development of Efavirenz. [Color figures can be viewed in the online issue, which is available at
wileyonlinelibrary.com.]

were studied, in which the imidazole ring is replaced by a pyrazole moiety. Several of these
derivatives (such as compound 83a) displayed the excellent broad spectrum anti-HIV acti-
vity.193a,b Especially, compound 83b (UK-453,061, lersivirine) was selected for further clinical
evaluation due to its very impresive potency against an interesting panel of key HIV-1 mutants,
safety, pharmacokinetic, and pharmaceutical proles (Fig. 29).193ce
Case 4. Sulfanyltriazole/tetrazolederivatives
Sulfanyltriazole/tetrazole derivatives were newly emerging HIV-1 NNRTIs with low
nanomolar intrinsic potency against the RT and anti-HIV activity cell-based assay.67ag
Currently, a number of compounds derived from this triazole/tetrazole scaffold are being
considered for further clinical trials for the treatment of HIV infection.67h Molecular mod-
eling indicated that the triazole/tetrazole moiety stays in the center of the NNIBP, anchoring
the substituents in the scaffold into the optimal space for interactions with NNIBP, which are
in agreement with the present insights of SAR and provide valuable avenues for the future
optimization of novel analogs as promising candidates for the treatment of AIDS.67a Based
on these analyses, additional and potentially more potent scaffolds have been designed and
synthesized independently in our194 and other195 laboratories (Fig. 30). Some derivatives
demonstrated high potency in inhibiting HIV-1 proliferation at nanomolar concentrations.
Case 5. Diaryletherderivatives
Taking advantage of the solvent exposed region (in the RT/solvent interface) of NNRTIs
as a means to incorporate solubilizing groups and to further optimize physico-chemical
properties and pharmacokinetics led to the discovery of novel structurally diverse diaryl ether
derivatives, such as indazole 63a,81 pyrazolopyridine 93 (MK-4965),196 pyrazolopyridazine
Medicinal Research Reviews DOI 10.1002/med
E28 K ZHAN ETAL.

Pyrrole core O
Me Me
Me Me
HN
HO O
N
S S S
O O N N N
Cl Cl
NH

Cl Me Cl
N N N

47, Capravirine 81a 81b

Designed to improve
metabolic stability Me Pyrazole core
Me Me Me

N N N
O O
N N
N
HO HO HO
CN Me Cl CN
Me Me
NC Cl NC
82 83a 83b, UK-453,061 (lersivirine)
EC =10 nM
EC =33 nM (WT) EC = 4 nM (WT)
CC >100 M

Figure 29. Optimization strategy for capravirine. [Color figures can be viewed in the online issue, which is available at wi-
leyonlinelibrary.com.]

94,197a pyridazinone 95,197b triazolinone 96,197c 97, 98,197d imidazolinone 99, 100, benzo-
[d]imidazolinone 101, imidazopyridinone 102, 103, imidazo[4,5-c]pyridazine 104, pyridone
105, pyrimidones 106, 107,197d pyrazole 108,198 and oxadiazole 109,199 which were regarded
as classic bioisosteres (Fig. 31). These analogs have high levels of activity against HIV-1
bearing WT strain and a panel of key mutations, and some of them show excellent oral
bioavailability and favorable pharmacokinetics.
Case 6. Modications of other NNRTIs based on bioisosterismprinciple
1-[2-(Diarylmethoxy)ethyl]-2-methyl-5-nitroimidazoles (DAMNIs) is an unique and
highly potent family of HIV-1 NNRTIs.82,200 Replacement of one phenyl group of lead
compound RS1408 (64) with heterocycles, such as 3-pyridinyl (110) or 2-thienyl (111), led to
novel DAMNIs with increased potency (Fig. 32). In HIV-1 WT cell-based assay, compounds
110 and 111 were found to be 2.5 and 6.7 times more active than compound 64. Compound
111 (IC50 5 8.25 mM) was also found more active than efavirenz (IC50 5 25 mM) against the
K103N mutant RT, suggesting for this compound a potential addition in efavirenz-based
anti-HIV regimens.201
A bioisosterism principle-based alteration, focusing on the replacement of the benzylthio
group of pyrimidine thioether NNRTI 42a,57a by different heteroaromatic systems, led to the
identication of PNU-142721 (42) as a clinical candidate (Fig. 33).57b,c,58
(7)-6-Ethyl-6-phenylpyrrolo[2,1-d][1,5]benzoxazepin-7(6H)-one (12) is the prototype of
pyrrolobenzoxazepinone NNRTs.26a Structural modications based on the bioisosterism
principle, dictated by docking studies, prompted the discovery of pyrrolopyridooxazepinones
(PPOs) analogs 112ac featuring a meta-substituted phenyl or a 2-thienyl ring at C-6 and a
pyridine system in place of the fused-benzene ring in the lead compound with a signicantly
improved pharmacological properties, in terms of efcacy, broad spectrum, and low cyto-
toxicity (Fig. 34). Compared with the lead 12 and nevirapine, PPOs 112ac displayed higher
inhibitory activity against WT RT and several key clinical RT mutations: L100I, K103N,
Medicinal Research Reviews DOI 10.1002/med
HIV-1 NNRTIs K E29

N S X N X N Se X
H H H
N N R N N N
S N S S
R2 O R2 O R2 O
Y Y Y

84 85
R4 86
R4 R4

Novel scaffolds discovered in our lab

Bioisosterism Principle

N N N N Br
H H
N N
Me N S Me N S
O O
Me
51c 51d
EC50= 2.053 M (WT) EC50 = 0.1 M (WT)
EC50= 1.3 M (K103N/Y181C)
Me

N N Cl N N NO2
H H
N N N N
N S N S
Me Me O O

51e 51f
IC50 = 5 nM (WT) IC50 = 9.5 nM (WT)
Me IC50 = 766 nM (K103N/Y181C)

Bioisosterism Principle

N X X N X
H H H
N N N N Me N N
N S N S S
R2 O R2 O R2 O
Y Y Y

87 88 89
R4 R4 R4

S X O X HN N X
H H H
N N N N Me N
S S S
R2 O R2 O R2 O
Y Y Y

90 91 92

R4 R4 R4

Novel scaffolds discovered in other labs

Figure 30. The structures of sulfanyltriazole/tetrazole derivatives. [Color figures can be viewed in the online issue, which is
available at wileyonlinelibrary.com.]

Medicinal Research Reviews DOI 10.1002/med


E30 K ZHAN ETAL.

F
NC O O NC O O NC O N
NH
Cl N Cl N Cl
Cl NH Cl NH Cl N
63a 93 94 N
N
HN EC (WT): 1 nM
EC (wt): 4.7 nM EC (WT): 1.35 nM EC (K103N/Y181C): 5 nM
EC (K103N/Y181C): 141 nM EC (K103N/Y181C): 84 nM
F F F
O
NC O N NC O N NC O
NH N
NH NH
N N
Cl O R Me Br Me
R 95 Me Cl 96 O O
Cl
R= Cl, Br, Me, Et, c-Pro 97
R = F, OMe, CN
IC (WT) = 7 nM, IC (K103N/Y181C) = 5 nM
EC (WT): 1 nM EC (WT): 2-11 nM
EC (K103N/Y181C): 4-40 nM EC (WT) = 3 nM, EC (K103N/Y181C) = 28 nM
EC (K103N/Y181C): 20-40 nM

F 98a: F
O O
NC O IC (WT) = 45 nM, IC (K103N/Y181C) = 13 nM NC O
N N
NH EC (WT) = 14 nM, EC (K103N/Y181C) = 55 nM NH

Br N 98b: Br
R
IC (WT) = 55 nM, IC (K103N/Y181C) = 12 nM Cl O
Cl 98a, R = H 99
98b, R = Me EC (WT) = 11 nM, EC (K103N/Y181C) > 100 nM
IC (WT) = 9 nM, IC (K103N/Y181C) = 11 nM
EC (WT) = 4 nM, EC (K103N/Y181C) > 100 nM

F F
O F
NC O NC O N
N NC O
NH N
N NH
Br Me Br NH
O Br O
Cl O Cl 101
100 Cl 102

IC (WT) = 6 nM, IC (K103N/Y181C) = 7 nM IC (WT) = 13 nM, IC (K103N/Y181C) = 8 nM IC (WT) = 5 nM, IC (K103N/Y181C) = 4 nM


EC (WT) = 4 nM, EC (K103N/Y181C) = 11 nM EC (WT) = 5 nM, EC (K103N/Y181C) = 46 nM EC (WT) = 0.4 nM, EC (K103N/Y181C) = 6 nM

F F F
N
NC O NC O NC O
N N N N

NH NH Br O N
Br O Br O H
Cl 103 Cl 104 Cl 105

IC (WT) = 6 nM, IC (K103N/Y181C) = 5 nM IC (WT) = 5 nM, IC (K103N/Y181C) = 4 nM IC (WT) = 19 nM, IC (K103N/Y181C) = 18 nM


EC (WT) = 3 nM, EC (K103N/Y181C) = 20 nM EC (WT) = 4 nM, EC (K103N/Y181C) = 38 nM EC (WT) = 25 nM, EC (K103N/Y181C) > 100 nM

F F
107a:
NC O Me NC O R IC (WT) = 4 nM, IC (K103N/Y181C) = 4 nM
N N
EC (WT) = 1 nM, EC (K103N/Y181C) = 19 nM
Br O N O Br O N O 107b:
H H
Cl 106 Cl
107a, R = H IC (WT) = 3 nM, IC (K103N/Y181C) = 3 nM
IC (WT) = 6 nM, 107b, R = Me
EC (WT) = 1 nM, EC (K103N/Y181C) = 11 nM
107c, R = Cl
EC (WT) = 5 nM, EC (K103N/Y181C) = 66 nM 107c:
IC (WT) = 4 nM, IC (K103N/Y181C) = 4 nM
N
EC (WT) = 2 nM, EC (K103N/Y181C) = 10 nM

F
NC O O NH
NC O O R2
N
N N
Cl Cl R
Cl Cl
108 109

IC (WT) = 4.5 nM, IC (L100I) = 21.9 nM IC (WT) < 10 nM, IC (K103N) < 10 nM,
IC (K103N) = 19.7 nM IC (V106A) < 10 nM,IC (Y181C) < 10 nM

Figure 31. Diaryl ether-based antivirals. [Color figures can be viewed inthe online issue, which is available at wileyonlinelibrary.com.]

Medicinal Research Reviews DOI 10.1002/med


HIV-1 NNRTIs K E31

Me Me Me
O O O
N N N N N N

O 2N O 2N S O 2N
N
64, RS1478 110 111

EC50 = 200 nM, SI >500 EC50 = 80 nM, SI >1250 EC50 = 30 nM, SI >3333

Figure 32. The strategy to the optimization of DAMNIs. [Color figures can be viewed in the online issue, which is available at
wileyonlinelibrary.com.]

Cl Cl Cl

N N N
Me H
N
H2N N S H 2N N S R H 2N N S

42a 42b: R = CONMe 2, EC90 = 90 nM (WT) 42d, PNU-142721 O


42c: R = CONEt 2, EC90 = 20 nM (WT) (S)-(-)-enantiomer
Delavirdine: EC90 = 30 nM (WT) IC50 = 20 nM (WT)
IC50 = 22 nM (P236L)

Figure 33. The strategy to the development of PNU-142721. [Color figures can be viewed in the online issue, which is available
at wileyonlinelibrary.com.]

N N N N N N N

O O O O
Me Me Me Me
O O O O

S
Me
12 112a 112b 112c
CEM-SS cells, EC50 = 470 nM, SI = 11 EC50 = 69 nM, SI = 56 EC50 = 150 nM, SI >13 EC50 = 27 nM, SI >75
C8166 cells, EC50 = 800 nM, SI = 12.5 EC50 = 54 nM, SI = 111 EC50 = 1200 nM, SI = 16.6 EC50 = 270 nM, SI = 26.7

Figure 34. Structural modification of pyrrolobenzoxazepinone NNRTIs. [Color figures can be viewed in the online issue, which
is available at wileyonlinelibrary.com.]

Table IV. Inhibition of WT HIV-1 and Several Key RT Mutations

Ki (mM)

Compd WT L100I K103N V106A Y181I Y188L

12 0.19 0.75 7.7 3.9 410 410


112a 0.022 0.04 0.3 0.07 4 1.5
112b 0.093 0.09 0.045 0.09 0.43 0.475
112c 0.021 0.044 0.03 0.4 1.2 1.5
Nevirapine 0.4 9 7 10 36 18

V106A, Y181I, and Y188L (Table IV). The antiviral activity and the synergistic antiviral
properties of 112a and 112b with AZT suggest a potential therapeutic usefulness in combi-
nation with NRTIs, against clinically signicant NNRTI-resistant HIV-1 strains.26b
Medicinal Research Reviews DOI 10.1002/med
E32 K ZHAN ETAL.

Table V. Anti-HIV Activities and Metabolic Stability of Cosalane Analogues

EC50 (mM) CC50 (mM)

HIV-1RF HIV-1IIIB IC50 CEM-SS MT-4 t1/2


Compd (CEM-SS cells) (MT-4 cells) (mM) cells cells (min) References
a b a a b b 202,203a
58b 0.013 0.6 0.3 31.6 160 5.8
204,205b
58c ND 1.8 ND ND 4224 55.3
206
58d 2.7 ND ND 16.3 ND 22.1
207
58e 0.7 0.24 0.67 2.9 12.4 3,641
207
58f 0.36 0.42 0.39 2.8 6.4 331
208
58g 0.05 0.14 0.47 5.7 7.0 864
209
58h 0.04 0.02 0.91 0.5 1.09 3.46
209
58i 0.6 41.1 0.63 1.2 41.1 51.4
210
58j 0.03 0.09 0.02 5.1 16.86 1.3
a represents that the date is from reference 202 or 203; b represents that the date is from reference 204 or 205.

The potential clinical utility of the alkenyldiarylmethanes (ADAMs) NNRTIs could be


affected by the metabolic instability of ester moieties that are prone to be hydrolyzed by
human nonspecic esterases.202,203 The replacement of labile esters with some bioisosters,
such as thioesters,204 various heterocycles,205209 and nitriles,206 led to the successful devel-
opment of several sub-micromolar NNRTIs that exhibited increased metabolic stability in
rat plasma relative to their parent analogs (Table V). Especially, ADAM 58e demonstrated
improved metabolic stability in rat plasma (t1/2 5 61 hr) along with the potency to inhibit
HIV-1 RT and the replication of HIV-1IIIB and HIV-1RF strains in cell assay at sub-
micromolar concentrations.207 Although the rat plasma half-lives of benzoxazolone deriva-
tive 58j was not optimized when compared to the prototype compounds, it was identied as
one of the highly potent compounds, which inhibited the replication of both HIV-1IIIB and
HIV-1RF with EC50 values of 90 and 30 nM, respectively, and inhibited HIV-1 RT with an
IC50 value of 20 nM (Fig. 35).210
In summary, a large number of exemplications show that the bioisosterism principle is
one of the most widely used medicinal chemistry strategies in NNRTI modications.
However, like any tool used in modern drug discovery, the bioisosterism principle has lim-
itations that require researchers with experience, insight, and creativity to use it intelligently
in the solution of the practical problems encountered in drug discovery.

2. Molecular Hybridization Concept: Me-Better


Molecular hybridization is a novel concept in drug design and optimization based on the
combination of basic structural elements (the pharmacophores) of different bioactive sub-
stances to obtain a new hybrid entity with improved afnity and potency, when compared to
the prototypes.211,212 The conceptual approach illustrated by many examples of NNRTI
hybrids, such as capravirineefavirenz hybrid 61,79 HBYefavirenz hybrid 113,213 pyr-
idinoneefavirenz hybrid 114,214 pyridinoneHEPT hybrid 115,215ad and pyridinoneDAPY
hybrid 116,216a,b promises to be broadly useful in the search for new chemical entities and can
contribute greatly to improve the speed and overall efciency of drug optimization.211,212
Figure 36 shows the recent adcances in the discovery of novel NNRTI platforms based on the
rational hybridization of two structurally distinct leads.
The discovery of aryl phosphinate-indoles 117a (IDX-899) and 117b was the result of
coordinated medicinal chemistry principles involving bioisosterism principle and molecular
hybridization (Fig. 37).217 In the structure of RTK103N/Y181C-117b complex, the newly
Medicinal Research Reviews DOI 10.1002/med
HIV-1 NNRTIs K E33
OMe
Me Me
Me Me N F
S O S O Me
O OMe
O OMe
MeO OMe Me N
OMe O
N Me
O O
Cl Cl Me

N
N O
58c O N
58d O 58e
Me
MeO O

MeO O MeO O MeO O S O


Me Me Me
O OMe MeO OMe OMe
O
N N
Me Cl Cl Me
MeO MeO

N
N O N
58f O 58b 58g N
MeO O
Me Me

MeO O MeO O O OMe


Me Me Me
OMe OMe O OMe
O O
N N O
Me N Me
Me
MeO MeO Me

O OMe N 58j MeO O


58i
58h
O
O

Figure 35. The strategy to improve the metabolic stability of ADAMs. [Color figures can be viewed in the online issue, which is
available at wileyonlinelibrary.com.]

introduced 3-substituent CH= =CHCN of the phenyl points directly to the highly conserved
W229. This must have contributed to its high potency. Because of its favorable safety prole
and predictable pharmacokinetics,217b a phase II clinical evaluation of IDX-899 has been
initiated recently.217c
As one of the most valuable structural modication tools useful for the discovery of new
ligands, the molecular hybridization approach has been successfully exploited across
different NNRTIs families and may be an effective strategy in the discovery of the next
generation NNRTI candidates.

3. Scaffold Hopping (Chemotype Switching): From One Hit to Another Attractive Series
Scaffold hopping has been recently reviewed and dened.218ae In chemoinformatics,
searching for compounds with structural diversity and common biological activity is entitled
scaffold hopping. On account of the structural similarity of NNRTIs families, scaffold
hopping or chemotype switching via dismantlement and simplication of known NNRTIs is
signicant since it can be used to produce alternative scaffolds with improved efciency and
Medicinal Research Reviews DOI 10.1002/med
E34 K ZHAN ETAL.

CN
Capravirine-efavirenz hybrid HBY-efavirenz hybrid

Me Me
NC O R
F O O Me

N
F N Et O O
N
H
61a: R=Me, RT IC = 50nM N
61b: R=Et, RT IC = 25nM N O CF
H
24b, HBY1293/GW867 N O
H
IC = 45 nM;
IC = 4.8 nM
Cl
113

FC
Cl
Cl S O
Me

N N O
Me H FC
N
3, Efavirenz Me O
N
O Me
Me N O
HN R H
O 114: IC = 32 nM
47, Capravirine N Pyridinone-efavirenz hybrid
X
Me O Me
R

Me N O
H
41a, L697,661: X = NH, R = Cl
41b, L-696,229: X = CH , R = H X Me
R
Me

Me N O 115
H
S
Pyridinone-HEPT hybrid
Me
O N a: R = COOEt, X = S. EC = 3 nM, SI > 3333
b: R = NO , X = S. EC = 6 nM, SI > 1666
O N O c: R = NH , X = CO. EC = 6 nM, SI > 1666
H d: R = NHCHO, X = CH . EC = 3 nM, SI > 3333
OH e: R = N(Me) , X = CH . EC = 0.2 nM, SI > 5000
39a, HEPT
f: R = I, X = O. EC = 1.3 nM, SI = 9000 (LAI);
EC = 3 nM (K103N); EC = 20 nM (Y181C)

a: R = -CN, X = C(=O), R = R = Me R
EC (nM) = 2 (LAI), 6 (K103N),
40 (Y181C), 398 (Y188L) 116 R
SI = 6310 (LAI)
b: R = -C=C-CN, X = C(=O), R = R = Me X
EC (nM) = 1 (LAI), 2 (K103N),
N
16 (Y181C), 158 (Y188L) Me R Me Me
SI = 10,000 (LAI) R
c: R = -C=C-CN, X = CH , R = Me, R = Et NH
Me N O N
EC (nM) = 1 (LAI), 2 (K103N), H X
32 (Y181C), 251 (Y188L) N
SI = 10,000 (LAI)
d: R = -C=C-CN, X = CH , R = Me, R = -(CH ) OMe N N Y
H
EC (nM) = 1 (LAI), 20 (K103N),
40 (Y181C), 316 (Y188L) 4a, TMC125: R = -CN, X = Br, Y = NH
Pyridinone-DAPY hybrid 4b, TMC278: R = -C=C-CN, X = Y = H
SI = 10,000 (LAI)

Figure 36. Representative cases of hybrid NNRTIs. [Color figures can be viewed in the online issue, which is available at
wileyonlinelibrary.com.]

unexpected side effects. In this section, the current use of scaffold hopping in the NNRTI
optimization is reviewed based on the representative examples of scaffold hopping in the
literature.
Medicinal Research Reviews DOI 10.1002/med
HIV-1 NNRTIs K E35

N Me
N

O O
S X MeO
O P
Cl NH2 Cl NH2
Me Me
HN N NH
N O N O
H H
N 117a, X= H
25, L-737,126 4b, TMC 278 117b, X = F

W229 Y188
Me
N
F227

V106 O
L234 F MeO
P
Cl NH2
H235 O L100
N O
P236 Y318 H
H
O N
N103
K101

Figure 37. Strategy for phosphoindole NNRTI discovery and schematic structure of RTK103N/Y181C with117b.217a,b [Color figures
can be viewed in the online issue, which is available at wileyonlinelibrary.com.]

DAMNI derivative RS1478 (64), an open-analog derived from thiazoloisoindolone 8b by


disruption of two nitrogen linkages of the thiazolidine ring, was identied as a potential
NNRTI candidate for anti-HIV-1 assays. Imidazole was chosen as a terminal ring because of
its presence in the structures of many tricyclic NNRTIs.82
LY300046-HCl (56a), a phenethylthiazolylthiourea (PETT) analog, obtained from the
dismantling of the rigid tricyclic nucleus of the tetrahydroimidazobenzodiazepinthione
(TIBO) derivative (Fig. 38), was more inhibitory to the Y188, Y181, and L100 mutations of
HIV-1 RT than was the parent compound 9-Cl-TIBO. Besides the minimal pharmacophoric
moieties of the TIBO structure necessary for antiviral activity, the presence of an additional
torsional freedom degree in LY300046 relative to earlier rigid TIBO analogs also probably
contributes to the excellent resistance proles of LY300046.72c
By dismantling the rigid tricyclic nucleus of the thiazolobenzimidazole (TBZ) prototype
lead, 2,3-diaryl-1,3-thiazolidin-4-one (49a),65 1,3-dihydro-2H-benzimidazol-2-one (33b),98
and 1-(2,6-diuorobenzyl)-2-(2,6-diuorophenyl)benzimidazole (34a)47ac were developed
as new NNRTIs, which proved to be more active than TBZs in inhibiting HIV-1
proliferation. These compounds can be envisaged as open models of TBZs since they keep
the basic pharmacophoric elements of the TBZs necessary for the HIV-1 RT inhibitory
activity.
The above mentioned and some other typical examples167169 indicated that the con-
formational exibility and positional adaptability of an NNRTI can contribute to the
inhibitor retaining efcacy against a variety of drug-resistant HIV-1 strains. It is worth
Medicinal Research Reviews DOI 10.1002/med
E36 K ZHAN ETAL.

A Me B S S
O HN HN
Disconnect N N
S N Disconnect HN

ON Me CH
N
Cl N Cl HN
O Me Me
64, RS1478
8b 5b, 9-Cl-TIBO (R82913)
Me Me
EC = 200 nM, SI >500
Isolation of potential
pharmacophore

S
C F F S
HN HN
2 HN HN
F Disconnect: 2 F
S F N
N
N N
EC = 1100 nM, HCl
N Br
1 SI = 45 N
7, NSC 625487 F 56a, LY300046 hydrochloride
Disconnect: 2 Me 34a
Disconnect: 1

F
Cl Cl
F
Cl N
S
Me N N O
N
O 33b H
49a EC = 240 nM, SI > 1766

Figure 38. The discovery of DAMNIs (A),82 PETTs (B),72c 2,3-diaryl-1,3-thiazolidin-4-one (49a),65 1,3-dihydro-2H-benzimida-
zol-2-one (33b)98 and1-(2,6-difluorobenzyl)-2-(2,6-difluorophenyl)benzimidazole (34a)47a--c (C) by dismantling of the rigid tricyclic
nucleus of the prototype leads. [Color figures can be viewed in the online issue, which is available at wileyonlinelibrary.com.]

highlighting that, in certain NNRTIs, reduced conformational exibility could also improve
inhibitory potency against HIV and RT enzyme together with selectivity, provided that the
conformationally restricted analogs with at least one conformation, among a few en-
ergetically allowed conformations, could be recognized by the binding site.219
Analysis of the bound conformation of tetra(tri)azole-based NNRTIs via molecular
modeling and NMR permitted the scaffold hopping from azoles to novel tertiary thio-
carbamate-based NNRTIs. Although these compounds did not provide the ideal improve-
ment in metabolic stability, they represent a novel, potent family of NNRTIs with a broad
spectrum of antiviral activity.67e,195f Analysis of the SARs of the thiocarbamate-based
NNRTIs contributed to the design of novel tetrahydroquinoline derivatives as potent
NNRTIs with nanomolar intrinsic activity against the WT and key mutant RTs and potent
anti-HIV activity in infected cells. In addition, the sulfur methylene linker was replaced with
a cis-cyclopropyl ring. A modeling study demonstrated that the conformation of a cis-
cyclopropyl amide could mimic the thiocarbamate.220
Also, the SAR conclusion, crystallography, and molecular modeling of tetra(tri)azole
and benzophenone-based NNRTIs permitted the scaffold hopping to a novel series of diaryl
ether NNRTIs which have excellent potency against WT and key mutant viruses (Fig. 39).221
Further systematic optimizations of the lead structure 121 resulted in the discovery of
compound 63a, which is the prototype of a potent and novel NNRTI family. In Figure 40, as
hypothesized, the two pyrrolo nitrogen atoms of the indazole moiety could be used as a
surrogate for the amide group, which appear to form two direct hydrogen bond interactions
with the carbonyl and imino of the backbone of K103.81
Following up on the aurintricarboxylic acid (ATA) lead, Mark Cushman and his cow-
orkers then described an ATA derivative cosalane (Fig. 41), in which one salicylic acid
Medicinal Research Reviews DOI 10.1002/med
HIV-1 NNRTIs K E37

O Cl O Cl
H H
Me N N
N S N S
Me Me O Cl O
SO2NH2 SO2NH2
118a
119
IC50(wt): 8 nM IC50(wt): 1 nM
IC50(K103N): 9 nM IC50(K103N): 1 nM
Me IC50(Y181C): 80 nM Cl IC50(Y181C): 4 nM

O Cl
H
N O Cl
Me N S H
N
Cl O N cis
Cl O
118b 120 SO2CH3
CH2COOH
IC50(wt): 34 nM IC50(wt): 18 nM
IC50(K103N/Y181C): 35 nM IC50(K103N): 18 nM
Me Me
Me Cl IC50(Y181C): 99 nM
Tertiary thiocarbamate-based NNRTIs
Tetrahydroquinoline-based NNRTIs

Scaffold hopping

Me
N N Cl H
H N
X N O O
N S
R O
R6 R2 O SO2NH2
SO2NH2

X = N, CH
R1 Cl
R4 Tetr(tri)azole-based NNRTIs
Benzophenone-based NNRTIs

Scaffold hopping

Cl
Y181/Y188/W229 H Y181/Y188/W229 C
N
CN O Scaffold hopping CN O
O N NH
SO2NH2
A B
O
Cl O H Cl O H
N N
Cl Solvent Cl Solvent
K103 K103
121 63a
V179 V179
IC50(wt): 0.14 nM IC50(wt): 1.35 nM
IC50(K103N): 0.21 nM IC50(K103N): 1.12 nM EC50(wt): 4.7 nM
IC50(Y181C): 0.28 nM IC50(Y181C): 2.62 nM EC50(K103N/Y181C): 141 nM
Diaryl ether-based NNRTIs Diaryl ether/pyrazole-based NNRTIs

Figure 39. Scaffold hopping paradigm for tetr(tri)azole-based NNRTIs and proposed binding mode for compounds 121
and 63a.67e, 195f, 220 [Color figures can be viewed in the online issue, which is available at wileyonlinelibrary.com.]

moiety is replaced by one cholestane group. Alkenyldiarylmethanes (ADAMs), structurally


related to cosalane, represent an addition to the group of NNRTIs.222 The crystallographic
studies revealed that ADAMs are highly hydrophobic and the shape of the NNIBP is unique
among other disclosed NNRTI-RT crystal structures.223
Medicinal Research Reviews DOI 10.1002/med
E38 K ZHAN ETAL.

Figure 40. The X-ray crystal structure of compound 63a (blue) in the WT-NNIBP (2.7 resolution, PDB code is 3C6U). (Hydrogen
bonds are shown as broken lines).81 [Color figures can be viewed in the online issue, which is available at wileyonlinelibrary.com.]
OH OH OH OH
Cl Me
O O
Me
OH Me Scaffold hopping
OH OH
Me
O Me
O O

HO O HO
Cl
122, Aurintricarboxylic acid 123, Cosalane

MeO O MeO O MeS O


Me
MeO OMe O OMe
N
Cl Cl Me
MeO

OMe OMe
Br O N
O 58b 58g
Scaffold hopping N
MeO O
OMe Me

O O
MeO O OMe
Me Me Me
MeO OMe
O O OMe
Br 124 O
N
Me N Me
EC = 7.1 5.0 M
(HIV-1 /CEM-SS cells) MeO Me

58h O OMe 58j MeO O

Figure 41. Historical synopsis of the discovery and development of the ADAM-type NNRTIs. [Color figures can be viewed in
the online issue, which is available at wileyonlinelibrary.com.]

Medicinal Research Reviews DOI 10.1002/med


HIV-1 NNRTIs K E39

4. The Usage of the Tolerant Region in the Modication of NNRTIs


(a) To improve pharmacokinetic properties
Interestingly, certain moiety in many NNRTI families sits between V106 and P236, and
points toward the solvent-exposed region. Consequently, this feature should be benecial for
the modulation of the physicochemical properties of the NNRTIs.
GW678248 (62b), a novel benzophenone NNRTI, potently inhibits WT and mutant RTs
in enzyme inhibition assays, with an IC50 between 0.8 and 6.8 nM. An N-propionyl sulfo-
namide derivative GW695634 (62c), designed as amide prodrug of GW678248, displayed
improved solubility and bioavailability in clinical trials (Fig. 43).224
The N-2 position of pyridazinone 95c, a potent NNRTI that has limited aqueous so-
lubility, was derivatized into a set of hydroxymethyl esters and carbonates as well as one
phosphate. These derivatives served as prodrugs to effectively deliver 95c to rat plasma upon
oral treatment at 50 mg/kg. Increases of 4.3- to 8.6-fold in 24-hr exposure of 95c (over that of
prototype) were observed, while the prodrugs and the hydroxymethyl derivative 95c-1 were
undetectable.225
An N-pyridinyl pyrimidinedione (KRV-2110, 39f) was previously identied as a potent
NNRTI (Fig. 42).226a However, pharmacokinetics in three animal models (rat, dog, and
monkey) demonstrated that once-daily dosage in humans was unlikely. Endeavor to improve
the suboptimal pharmacokinetic prole of 39f led to the discovery of compounds 39g and
39h, which represent the promising compounds in this series with similar antiviral potency as
inhibitor 39f and improved pharmacokinetics that may support once-daily dosage.226b,c
In the tetrazolyl series, it has been demonstrated that the introduction of substituents
(i.e. alkynyl fragment) at the para position of the anilide in 51f led to substantial improve-
ment in the overall physicochemical proles of the molecule while keeping excellent potency
against the K103N/Y181C double mutant RT. As shown in Figure 42D, extensive SAR and
pharmacokinetic evaluation resulted in the discovery of candidate 51i with favorable oral
bioavailability and PK properties in rats.67f

(b) Multiple ligands design strategy


Designed multiple ligands (DML), an emerging and appealing drug discovery strategy, using
a single chemical entity to inhibit multitargets, should be effective in improving patient
compliance, reducing problems of dosing complexity, drugdrug interactions and toxicities,
as well as diminishing the likelihood of virusdrug resistance.166 The exploration of DML
strategy should be valuable in anti-HIV drug discovery.166
Apparently, the key to rational DML strategy would be to identify a tolerant region in
the drug target. Crystallographic studies have shown that the phenyl group in the N-1
substituent of the HEPT type of NNRTIs and the methylsulfonamide group at the C-5
position of delavirdine are situated in an open area (the solvent-exposed region) controlled by
the P236 loop where structural alternations could be tolerated. Based on these general
knowledge and the DML strategy,165,166 several series of RT/IN (integrase) dual inhibitors
were designed and synthesized via incorporation of an IN pharmacophore element to this
tolerant region of a known potent NNRTI (Fig. 43), and many inhibitors demonstrate
activity against RT at low to submicromolar range, and against HIV at nanomolar range
(compounds 126a and 126b), which also conrms that the introduction of a second phar-
macophore to these NNRTIs does not seriously impair their binding with RT. In addition,
moderate anti-IN activity was also observed.227ad
Undoubtedly, the growing efforts in recent years to discover multitarget agents resulting
from the rational combination of pharmacophoric moieties of different known lead com-
pounds will bring a new perspective for the treatment of AIDS.
Medicinal Research Reviews DOI 10.1002/med
E40 K ZHAN ETAL.

A Me
Me
H H
N N
O O O O
H
NC O NH2 NC O N
S S Me
O O O O
O

Cl Cl Cl Cl

62b, GW678248 62c, GW695634


B
F F
NC O N NC O N
NH N O

Cl O Cl O
CN 95c Me CN 95c-1 ~ 95c-9 Me

O O O O
OH P NH2
H ONa N
ONa HCl HCl
O

95c-1 95c-2 95c-3 95c-4 95c-5

O O O O O
H
NH2 N NMe2 N
O O
HCl HCl
HCl HCl
95c-6 95c-7 95c-8 95c-9

C O Me O Me O Me

HN Me HN Me HN Me
O O O
O N O N O N
H2N

N N
Me CN Me CN Me CN
F F
39f, KRV-2110 39g 39h

D
N N Cl N N Cl
H H
N N N N H
S S N
N N NH
51g: R =
Cl O Cl O Me Me O
H R OH
51h: R = N
51f H
Me Me O
EC (WT) = 82 nM O
EC (K103N/Y181C) = 505 nM
t = 4 min (RLM) 51g: EC = 1.1 nM(WT); 8.1 nM(K103N/Y181C)
51i: R = OH
t = 109 min (RLM)
51h: EC = 1.6 nM(WT); 13 nM(K103N/Y181C) Me Me
t = 79 min (RLM) O O O
51i: EC = 7.1 nM(WT); 58 nM(K103N/Y181C)
S
t = 90 min (RLM) N Me
51j: R =
51j: EC = 4.2 nM(WT); 27 nM(K103N/Y181C) H
Me Me
t = 82 min (RLM)

Figure 42. Successful cases of NNRTIs modifications in the tolerant region to improve pharmacokinetic profiles. [Color figures
can be viewed in the online issue, which is available at wileyonlinelibrary.com.]

Medicinal Research Reviews DOI 10.1002/med


HIV-1 NNRTIs K E41

O Me
Me
HN

O N
RT IC = 0.016 M O RT IC >100 M
O IN IC > 100 M IN IC =0.093 M
OH
HIV-1 EC = 0.016 M HIV-1 EC =0.16 M
O OH
RT inhibitor SI> 610 IN inhibitor 125 SI> 61
39e,TNK651

O O Me Me
Me Me

HN HN

O N O N Me
Me

O O O O

126a OH 126b OH
O OH O OH

RT IC = 0.024 M RT IC = 0.028 M
IN IC = 4.4 M IN IC = 14 M
HIV-1 EC = 0.0097 M HIV-1 EC = 0.014 M
RT/IN dual inhibitor
SI> 1000 SI> 710

Figure 43. Discovery of RT/IN dual inhibitors 126a and 126b against RTand IN, combining RT inhibitor 39e with IN inhibitor 125.
[Color figures can be viewed in the online issue, which is available at wileyonlinelibrary.com.]

(c) Multivalency drug design strategy (to form additional proteinligand interactions)
By the classical concept for drug design, a favorable inhibitor should efciently enter and
maximally occupy the binding site, thus interacting effectively with the residues around the
binding pocket.228 Therefore, the larger the NNRTI, the stronger is the interaction with
residues around the NNIBP, provided that the NNRTI can enter efciently inside the
NNIBP. This is partly supported by docking studies that the overall shape and chemical
structure of larger second generation NNRTIs enables them to occupy more space in the
NNIBP than the smaller and less active NNRTIs such as nevirapine,229 and thereby form
additional proteinligand interactions, especially the strong interactions of the drug with the
conserved regions or polypeptide backbone in the NNIBP, which is regarded as a primary
strategy to improve the potency of NNRTIs against resistant mutations.
This is consistent with the concept of multivalency, that using one entity to bind
multiple targets or binding additional sites in one target, simultaneously, could result in a
signicantly improved efciency.230,231
For example, several novel 8-substituted nevirapine-based analogs (127ah) display ex-
cellent broad antiviral activity against a panel of prevalent RT mutants and excellent
pharmacokinetic proles (Fig. 44).232235 Especially, BILR 355 BS (127h) was once advanced
into Phase II clinical trials in 2005.236 X-ray crystallographic study reveals that while the
dipyridodiazepinone core of BILR 355 BS and its analogs bind in an overall similar con-
formation to that of nevirapine, it has a greater ability to accommodate its orientation in the
NNIBP of the mutants. Additionally, the extended C-8 substituent off the dipyr-
idodiazepinone core likely make additional favorable binding contacts with RT (including
Medicinal Research Reviews DOI 10.1002/med
E42 K ZHAN ETAL.

Me Me Me Me
O O O
N N N

8 N Cl N Cl Cl
N N S N N
N N N N
Me Me
Me
127a 127b 127c

Me Me Me
O H O O
N N N

Me
N Cl N Cl N Cl
N N N
N S N O N O N
Me Me Me

Me 127d 127e 127f


N N
O O

Me Me
O O
N N

N N N N
Me O N O N
Me Me

127g 127h
N
HO O
O

Figure 44. 8-Substituted nevirapine-based NNRTIs. [Color figures can be viewed in the online issue, which is available at
wileyonlinelibrary.com.]

Me Me

O Me O Me

I I
Me S
O
Me N O Me N O
H H

1152f 115g, R221239

Figure 45. Pyridinone NNRTI115 g (R221239) and its contacts with RT (PDB code: 2BE2). [Color figures can be viewed in the
online issue, which is available at wileyonlinelibrary.com.]

P236 and K103 backbone) that may stabilize binding of these 8-substituted analogs even in
the presence of NNRTI-resistance mutations.236
Compared with pyridinone derivative 115f,208d R221239 (115g) contains a exible linker
and a connected furan ring which permits close contacts with V106, F227, and P236. These
additional interactions appear to enhance the inhibitory activity of R221239 against the
HIV-1 strains carrying the V106A, Y188L, and F227C mutations (Fig. 45).171
Compared with benzimidazole 34a, its analog 34b is apparently able to make signicant
interactions with the RT backbone via additional hydrogen bonds (Fig. 46), which are unlikely
to be disrupted by side chain mutations and contribute signicantly to the compacted binding
for the inhibitor, and eventually, signicantly improve the resistance prole of this inhibitor.47ac
Medicinal Research Reviews DOI 10.1002/med
HIV-1 NNRTIs K E43

F F

F F F F
N N

N N
F F
Me
O
O

34a 34b
IC50 = 0.2 M,EC50 = 0.44 M(WT) EC50 (M) = 0.062 (WT); 0.068 (L100I);
SI > 100 0.025 (K101E); 0.027 (K103N); 0.013 (V108I);
2.32 (Y181C); 0.033 (Y188C); 4.5 (V106A)

Figure 46. Benzimidazole-based NNRTIs.47a--c [Color figures can be viewed in the online issue, which is available at
wileyonlinelibrary.com.]

O Me

HN Me
O
O N
H 2N

N
Me CN
F
39f, KRV- 2110

Figure 47. Compound 39f bound in the NNRTI binding pocket of HIV-1RT (PDB code 3LAK, 2.3 resolution).218b [Color
figures can be viewed in the online issue, which is available at wileyonlinelibrary.com.]

As mentioned above, one difference in the binding manner of different NNRTIs are the
hydrogen bond interactions with the protein backbone. For instance, diarylpyrimidine (DAPY)
NNRTIs could make a hydrogen bond from the imino (connected to pyrimidine) to the
backbone carbonyl of K101, while the benzophenone family has a hydrogen bond interaction
between its amide carbonyl and the imino of the K103 amide. Interestingly, some promising
NNRTIs are capable to make double hydrogen bond interactions with K101 and K103.
Antiviral proles revealed that HEPT derivative 39f exhibited potent activity against
Y181C and K103N mutant strains. Detailed crystallographic analysis of 39f/HIV RT complex
structure indicated that an additional hydrogen bond was formed between the amino of the
aminouoropyridyl moiety and the backbone carbonyl of the K103 besides the hydrogen bond
between the imino of the pyrimidinedione and the backbone carbonyl of K101 (Fig. 47).226b
Compared with etravirine, piperidine-linked aminopyrimidine derivatives 128a and 128b
possess favorable potency against WT RT as well as several important mutant strains
(including the K103N/Y181C and Y188L mutants) (Table VI). Crystal structure analysis of
this series compounds showed that, besides a hydrogen bond from the aminopyrimidine
imino to the backbone carbonyl of K101, the piperidine nitrogen could probably make an
additional hydrogen bond with the K103 backbone (via a bridging water molecule).237a,b
Medicinal Research Reviews DOI 10.1002/med
E44 K ZHAN ETAL.

Table VI. Activity of 128a, 128b, and Etravirine Against WT RT as well as Several Important Mutant
Strains
SO2CH3 CONH2
N N

Cl

Me Me F Me
O N NH O N NH

N N
Br Br
128a 128b

WT K103N/Y181C K103N/L100I Y188L G190A V106A


Compds (nM) (nM) (nM) (nM) (nM) (nM)

128a237a 6.1 7.3 5.9 2.8 1.8 2.0


128b237b 6 5 ND 29 ND ND
Etravirine237a 2.1 9.2 9.5 3.1 1.1 2.0

O Me
N
O HN Me

N O

Cl
N
129
238

238
Figure 48. Cocrystal structure of 129 inthe NNIBP (3FFI). [Color figures can be viewed inthe online issue, which is available at
wileyonlinelibrary.com.]

Similarly, pyridone inhibitor 129, which was found to strongly inhibit the WT and
NNRTI-resistant K103N and Y181C mutant HIV RT, could not only interact with the
carbonyl group of K101 through a strong traditional hydrogen bond but also engage in
additional hydrogen bond interaction with the backbone amide of K103 (Fig. 48).238
In comparison with that of compound 33b, the improved potency of compounds 33a and
101 might be owing to the electronic characteristics of the sulfonyl moiety and the phenyl
group carrying a cyano group, respectively (Fig. 49). Molecular modeling indicated that the
sulfonyl linker in compound 33a was able to form tight interactions with residues Y181,
Y188, V179, and V106, while the methylene linker of compound 33b forms contacts only
with residues Y188 and V179.46
Medicinal Research Reviews DOI 10.1002/med
HIV-1 NNRTIs K E45

Cl

Me NC Br

F Me O

O
F S F
Cl Cl O
N N N
O O O
N N N
H H H

33b 33a 101


EC50 = 240 nM, SI > 1766 IC50 = 5 nM, EC 50 = 2 nM IC50 (WT) = 13 nM, IC 50 (K103N/Y181C) = 8 nM
SI = 17,846 EC50 (WT) = 5 nM, EC 50 (K103N/Y181C) = 46 nM

Figure 49. Benzimidazolone-based NNRTIs.46,197d [Color figures can be viewed in the online issue, which is available at
wileyonlinelibrary.com.]

K101
V179
C
O O K103
B
V106 Me
HN
Y318 R P95
D Me
S 2 N
F227 R R Y181
F F
P225 MeO Y188
A
P236 40d W229

F227
L100 L234

EC50: 0.07 nM (WT); 36 nM (K103N); 13 nM (Y181C); 1.5 nM (Y188L);


CC50: 14640 nM

Figure 50. Interactions of a C2-arylalkyl S-DABO (40d) within the NNIBP.239b [Color figures can be viewed in the online issue,
which is available at wileyonlinelibrary.com.]

It seems likely that extended phenyl ring in compound 101 (Fig. 49) could (i) occupy an
hydrophobic space near the top of the NNIBP, thus creating additional intermolecular contacts
with the adjoining residues and (ii) increase the hydrophobic interaction with the highly conserved
W229 at the roof of NNIBP, thereby decreasing the dependence on binding with Y181.197d
The structural modications of S-DABO NNRTIs were aimed at exploring the SAR of
the C2-functionalization in pyrimidine core,239a,b leading to the discovery of a potent in-
hibitor 40d having picomolar activity against WT RT and nanomolar activity against many
key mutant strains. The introduction of an arylalkyl group in C2 signicantly increased the
antiviral activity due to protable hydrophobic interaction with a large pocket (zone D) of
the allosteric pocket. Especially, the cyclopropyl group, as a bulky lipophilic substituent,
probably formed additional interactions with the hydrophobic residues of zone D, giving an
additional contribution to the afnity with the allosteric site (Fig. 50).239b
When introducing modications in the indolylarylsulfone (IAS) type of NNRTIs,240,241
new potent candidates 25cg were obtained by coupling several kinds of amino acids to the
2-carboxamide of the indole core (Fig. 51). In human T-lymphocyte (CEM) cell assay, the
Medicinal Research Reviews DOI 10.1002/med
E46 K ZHAN ETAL.

Me Me
NH O
Me Me 25c: R = 25f: R =
S
Me O NH
O
O O NH
S S
O O 25d: R =
Cl NH Cl HN R O
O 25g: R =
S
NH NH
N O N O Me O
H H 25e: R =
25b 25c-g O

Figure 51. The structures of novel indolylarylsulfones bearing natural and unnatural amino acids.241

IASs could inhibit the HIV-1 replication at low/subnanomolar concentrations and with weak
cytotoxicity. The antiviral potency against the K103N, Y181C, and L100I mutant strains in
CEM cells was comparable to that of EFV. With the aim to investigate the conceivable
binding mode of the new IASs, the highly potent compound 25e (IC50 5 26 nM; EC50 5
0.70 nM) was selected for docking studies into the NNIBP of WT RT (PDB code 2RF2) and
L100I RT (PDB codes 1S1T, 2OPQ). From docking 25e into the NNIBP were the newly
formed interactions of the N-(3-amino-3-oxopropyl)carboxamide moiety with the residues
K101 and E138 at the bottom of the NNIBP, which seemed to be particularly important for
the anti-HIV activity. Mutation of leucine to isoleucine did not affect the binding mode.241
In addition, the highly potent pyrazolo[3,4-c]pyridazine derivatives discovered by
structure-based optimization of diaryl ether NNRTIs could bind RT in an expanded volume
relative to most other analogs in the diaryl ether family, and probably engage in additional
interactions with RT.197a
Besides the above described NNRTIs, several series of divalent RT inhibitors by
tethering one molecule of NRTI to an NNRTI structure via a exible linker were designed
and synthesized to occupy the distinct but proximal catalytic site and NNIBP simulta-
neously. However, the results of their anti-HIV assay were not satisfactory.242

5. The Usefulness of Stereochemistry in Overcoming Drug Resistance


Molecular modeling studies pointed to the asymmetric geometry of the NNIBP, and
experimental data proved that the regiochemistry and stereochemistry of NNRTIs can in-
uence their anti-HIV activity substantially.243ad
The introduction of different stereocenters in important pharmacophoric sites of pro-
mising NNRTIs scaffold could allow the emerge of a collection of enantiopure drugs with
improved efcacy and potential usefulness in managing drug-induced mutations. Typically,
the chiral cyclopropane ring-containing oxindole 29,44 quinolones 30,45a S-DABO 40d,239
urea-PETT analogs 56c, 56d,74a,b 56e,74c,d and tetrahydroquinoline 120220 exhibited nano-
molar activity against several clinically relevant mutants (Fig. 52).

C. Computational Chemistry Approaches for NNRTIs Optimization


In the past 15 years, with the signicantly increased computer speed and program efciency,
the role of computational chemistry in drug design has expanded exponentially. Recently, the
applicability of computational chemistry and the computer-aided drug design (CADD)
techniques on NNRTIs has been intensively reviewed by Hannongbua244,245 and Jorgensen
et al.246 Pharmacophore modeling, database searching, and de novo methodologies have been
described in the above sections. Thus, the aim of this section is to summarize the applications
of computational chemistry to the structural modication of HIV-1 NNRTIs. The following
applications are emphasized, including quantitative structure-activity relationship (QSAR),
Medicinal Research Reviews DOI 10.1002/med
HIV-1 NNRTIs K E47
OEt
O
O
R Me
Br Cl CO R HN

O Me
S N
N N O
H H F F
MeO
29a R = (CH ) CHCH , nPent
40d
R = Et, Me, Allyl
30

Me Me
O O
F
OH
O O Cl
H
F N
N cis
F F Cl O
Br NC SO Me
HN HN O HN
N N N
O O O
N O 120
N N
H H H Cl
NH
HN
56c, MSC-204 56d, MSC-372 56e

Figure 52. NNRTIs containing a chiral cyclopropane ring. [Color figures can be viewed in the online issue, which is available
at wileyonlinelibrary.com.]

molecular docking, free energy perturbation (FEP)-guided lead optimization, and sub-
structural molecular fragments (SMF) method.
Many applications have been reported on the use of two- and three-dimensional QSAR
(2D/3D-QSAR) studies to understand the NNRTI-RT interactions and help in the design of
more effective analogs.247256 The 3D-QSAR model based on the docked conformation
reveals an excellent capability to predict the activity and provides valuable information on
possible improvement in the ligand structure for increasing potency of the inhibitors.
Combination of structure-based docking simulation with ligand-based QSAR will enhance
the likelihood for nding novel lead compounds. Moreover, the particular interaction energy
trend calculated from quantum chemical calculations (QCC) of the NNRTIs and individual
residues in the NNIBP and QM/MM methodology (such as ONIOM method our own
N-layered integrated molecular orbital and molecular mechanics) should increase the un-
derstanding of NNRTI-RT interaction mechanism as well as providing some insights into
drug resistance, which can be used as a promising descriptor identifying a key structural
element for QSAR study.244,245
To rationalize the most relevant SARs of potent NNRTIs and to investigate the
orientation and estimated binding energy of NNRTIs in the NNIBP, molecular docking
simulations were usually performed. The docking results gave an insight into the pharma-
cophoric structural requirements for efcient RT inhibition of related NNRTIs, providing an
informative guideline in designing new derivatives. To manage the conformational exibility
of RT, the inclusion of structural variability in a docking study and the concept of ligand-
induced t by cross-docking approach (the process of docking each ligand into the binding
site of a number of different ligandreceptor complexes) have been taken into account.96,257
Dock molecular mechanics-generalized born/surface area (MM-GB/SA-ADME) is a useful
tool to predict the afnity of NNRTIs with the RT and further screen for promising
candidate drugs though CADD.258,259
FEP-guided lead optimization is regarded as a valuable approach for molecular design
including drug discovery.260 Efcient optimization of an inactive 2-anilinyl-5-benzyloxadiazole
Medicinal Research Reviews DOI 10.1002/med
48
E48 KK ZHAN
ZHANETAL.
ETAL.

A
Me Me CN
Me
FEP-Guided
Optimization
X X
X
O NH O
Y NH

N N N
Cl
46a, inactive Cl 46e, X= Cl, Y = H, EC 50= 22 nM
O NH 46f, X= F, Y = H, EC 50= 13 nM
46g, X= F,Y = Cl, EC 50= 6 nM
N N
46b, X= Cl, Y= H. EC 50= 820 nM
46c, X= Y= Cl. EC 50= 310 nM
46d, X= CN, Y= H. EC 50= 130 nM

B
Me Me Me

Me Me Me

O FEP-Guided O FEP-Guided O
Optimization Cl Optimization R2
S N N

N N N N R1 N N
H H H
77: EC50 = 10,000 nM 43a: EC50 = 200 nM 43b: R1 = MeO, R 2 = Cl. EC 50 = 10 nM
FEP-Guided 43c: R1 = MeO, R 2 = CN. EC 50 = 2 nM
Optimization 43d: R1 = MeNH, R 2 = CN. EC 50 = 5 nM

Me Me Me

Me Me Me

O O O Y O
Cl N Cl CN
X N N

N N N N X N N
H H H
130a: X = CH, EC 50 = 6 nM, SI = 4167 131: EC50 = 19 nM, SI = 1053 43e: X = MeS, Y = H. EC 50 = 5 nM
130b: X = N, EC 50 = 5 nM, SI = 3400 43f: X = MeO, Y = NH 2. EC50 = 9 nM

Figure 53. The application of free energy perturbation (FEP)-guided lead optimization.

46 (false-positive molecule in virtual screening) and the less potent thiazole 77 (generated by
de novo design) has been guided by FEP calculations to provide potent anilinylbenzyloxa-
(dia)zole-based NNRTIs62ad and diarylamine-based NNRTIs,59ac,261,262 respectively
(Fig. 53). More importantly, achieving simultaneous efcacy for the WT RT and a panel of
commonly observed mutants can be highly automated by running FEP calculations in parallel
for a given designed NNRTI with all known, common mutant forms of the RT.
The SMF method was also applied for computer-aided design of new NNRTIs poten-
tially possessing high anti-HIV activities, such as TIBO and HEPT derivatives.263
The innovative computational approach GRID-Based Pharmacophore Model
(GBPM) was believed to have great value in the identication of conserved regions of the
HIV-1 RT, to be targeted for the development of novel therapeutic agents.264
Medicinal Research Reviews DOI 10.1002/med
HIV-1NNRTIs
HIV-1 NNRTIs KK 49
E49

Totally, as more structural information has become available and a wide range of
computational approaches have proven to have reasonable predictive value, CADD should
play increasingly important roles in designing novel NNRTIs to defeat the nagging resistance
issue and handling the exibility of the target site (NNIBP).

6. SUMMARY AND PERSPECTIVES

In recent years, in spite of the rapid growth of HIV-1 RT 3D-structural information, the
difculty in structure-based de novo design of NNRTI scaffolds and docking-based virtual
screening lies in the following two aspects: (i) The exibility of NNIBP, formed by con-
formational changes in the RT on binding of the NNRTI ligand; (ii) The NNRTI-resistant
mutations situated in and around the NNIBP. Therefore, as reviewed above, structure-based
and ligand-based combined drug design methodology was carried out to facilitate both drug
lead generation and lead optimization. Quite a few cases illustrated the benets for NNRTIs
design of closely coupled traditional medicinal chemistry, structural biology, computational
chemistry methodology, and several other disciplines.265,266
In the NNRTI lead discovery process, compared to the structure-based de novo design
and computer-aided in silico screening, large chemical libraries, combinatorial chemistry,
HTS, and naturally occurring products still serve as approaches or sources of new active
NNRTI leads to be further developed as anti-AIDS drug candidates. Yet, such screenings
and the subsequent optimization of the inhibitors by systematic chemical modications are
highly time- and resource consuming.
In the NNRTIs modication process, the crystallographic studies, providing a basis for
understanding the interactions or tolerant region between the bound NNRTIs and the sur-
rounding amino acid residues, contributed to the improved resilience or afnity of the opti-
mized compounds. Great progress has been made in the development and application of
medicinal chemistry strategies, such as bioisosterism, molecular hybridization, scaffold hop-
ping, and multiple/multivalent ligand design strategy. The coordination of computational
chemistry and crystallography is playing an increasingly important role in attempting to
understand and defeat virusdrug resistance and the target site exibility problem.246 For
instance, cross-docking experiments on the WT and mutated RTs were conducted to consider the
enzyme exibility as an inevitable problem for structure-based drug design studies and to gain
insight into the mode of action of new NNRTIs active against both WT and resistant strains.
The fundamental goal of medicinal chemists is to make drug discovery more efcient by
reducing the number of compounds that need to be synthesized and assayed; the ultimate
goal is to obtain novel anti-HIV drugs with high levels of potency against WT and key
mutant HIV strains without allowing breakthrough, excellent oral bioavailability, and
overall pharmacokinetics. It is clear that only multidisciplinary coordination could help to
achieve these goals.

REFERENCES

1. Tronchet JM, Seman M. Nonnucleoside inhibitors of HIV-1 reverse transcriptase: From the
biology of reverse transcription to molecular design. Curr Top Med Chem 2003;3:14961511.
2. Martins S, Ramos MJ, Fernandes PA. The current status of the NNRTI family of antiretrovirals
used in the HAART regime against HIV infection. Curr Med Chem 2008;15:10831095.
3. Zhou Z, Lin X, Madura JD. HIV-1 RT nonnucleoside inhibitors and their interaction with RT for
antiviral drug development. Infect Disord Drug Targets 2006;6:391413.

Medicinal Research Reviews DOI 10.1002/med


50
E50 KK ZHAN
ZHANETAL.
ETAL.

4. (a) Seminari E, Castagna A, Lazzarin A. Etravirine for the treatment of HIV infection. Expert Rev
Anti Infect Ther 2008;6:427433. (b) Daar ES. Emerging resistance proles of newly approved
antiretroviral drugs. Top HIV Med 2008;16:110116.
5. Arendt G, de Nocker D, von Giesen HJ, Nolting T. Neuropsychiatric side effects of efavirenz
therapy. Expert Opin Drug Saf 2007;6:147154. (b) Fumaz CR, Munoz-Moreno JA, Molto J,
Negredo E, Ferrer MJ, Sirera G, Perez-Alvarez N, Gomez G, Burger D, Clotet B. Long-term
neuropsychiatric disorders on efavirenz-based approaches: Quality of life, psychologic issues, and
adherence. J Acquir Immune Dec Syndr 2005;38:560565. (c) SUSTIVAs (efavirenz) capsules
and tablets: Package insert, Bristol-Myers Squibb Co, Princeton, NJ, 2008. Package inserts.
Bms/com/pi/pi_sustiva.pdf.
6. (a) Boone LR. Next-generation HIV-1 non-nucleoside reverse transcriptase inhibitors. Curr Opin
Investig Drugs 2006;7:128135. (b) Sweeney ZK, Klumpp K. Improving non-nucleoside reverse
transcriptase inhibitors for rst-line treatment of HIV infection: The development pipeline and
recent clinical data. Curr Opin Drug Discov Dev 2008;11:458470.
7. Alberts IL, Todorov NP, Dean PM. Receptor exibility in de novo ligand design and docking.
J Med Chem 2005;48:65856596.
8. Cozzini P, Kellogg GE, Spyrakis F, Abraham DJ, Costantino G, Emerson A, Fanelli F,
Gohlke H, Kuhn LA, Morris GM, Orozco M, Pertinhez TA, Rizzi M, Sotriffer CA. Target
exibility: An emerging consideration in drug discovery and design. J Med Chem 2008;51:
62376255.
9. B-Rao C, Subramanian J, Sharma SD. Managing protein exibility in docking and its
applications. Drug Discov Today 2009;14:394400.
10. (a) De Clercq E. The role of non-nucleoside reverse transcriptase inhibitors (NNRTIs) in the
therapy of HIV-1 infection. Antiviral Res 1998;38:153179. (b) De Clercq E. Perspectives of
non-nucleoside reverse transcriptase inhibitors (NNRTIs) in the therapy of HIV-1 infection.
Farmaco 1999;54:2645. (c) De Clercq E. Non-nucleoside reverse transcriptase inhibitors
(NNRTIs): Past, present, and future. Chem Biodivers 2004;1:4464. (d) De Clercq E. Another
ten stories in antiviral drug discovery (part C): Old and new antivirals, strategies, and
perspectives. Med Res Rev 2009;29:611645.
11. Campiani G, Ramunno A, Maga G, Nacci V, Fattorusso C, Catalanotti B, Morelli E,
Novellino E. Non-nucleoside HIV-1 reverse transcriptase (RT) inhibitors: Past, present, and
future perspectives. Curr Pharm Des 2002;8:615657.
12. Pauwels R. New non-nucleoside reverse transcriptase inhibitors (NNRTIs) in development for the
treatment of HIV infections. Curr Opin Pharmacol 2004;4:437446.
13. Basavapathruni A, Anderson KS. Developing novel nonnucleoside HIV-1 reverse transcriptase
inhibitors: Beyond the buttery. Curr Pharm Des 2006;12:18571865.
14. Balzarini J. Current status of the non-nucleoside reverse transcriptase inhibitors of human
immunodeciency virus type 1. Curr Top Med Chem 2004;4:921944.
15. Zhan P, Liu X, Li Z. Recent advances in the discovery and development of novel HIV-1 NNRTI
platforms: 20062008 update. Curr Med Chem 2009;16:28762889.
16. de Bethune MP. Non-nucleoside reverse transcriptase inhibitors (NNRTIs), their discovery,
development, and use in the treatment of HIV-1 infection: A review of the last 20 years
(19892009). Antiviral Res 2010;85:7590.
17. Prajapati DG, Ramajayam R, Yadav MR, Giridhar R. The search for potent, small molecule
NNRTIs: A review. Bioorg Med Chem 2009;17:57445762.
18. (a) Pauwels R, Andries K, Desmyter J, Schols D, Kukla MJ, Breslin HJ, Raeymaeckers A, Van
Gelder J, Woestenborghs R, Heykants J, Schellekens K, Janssen Marcel AC, De Clercq E, Janssen
Paul AJ. Potent and selective inhibition of HIV-1 replication in vitro by a novel series of TIBO
derivatives. Nature 1990;343:470474. (b) Pauwels R, Andries K, Debyser Z, Kukla M, Schols D,
Desmyter J, De Clercq E, Janssen PA. TIBO derivatives: A new class of highly potent and specic
inhibitors of HIV-1 replication. Biochem Soc Trans 1992;20:509512.

Medicinal Research Reviews DOI 10.1002/med


HIV-1NNRTIs
HIV-1 NNRTIs KK 51
E51

19. Kelly TA, McNeil DW, Rose JM, David E, Shih CK, Grob PM. Novel non-nucleoside inhibitors
of human immunodeciency virus type 1 reverse transcriptase. 6. 2-Indol-3-yl- and 2-azaindol-3-
yl-dipyridodiazepinones. J Med Chem 1997;40:24302433.
20. Merluzzi VJ, Hargrave KD, Labadia M, Grozinger K, Skoog M, Wu JC, Shih CK, Eckner K,
Hattox S, Adams J, Rosehthal AS, Faanes R, Eckner RJ, Koup RA, Sullivan JL. Inhibition of
HIV-1 replication by a nonnucleoside reverse transcriptase inhibitor. Science 1990;250:14111413.
21. (a) Chimirri A, Grasso S, Monforte AM, Monforte P, Zappala M. Anti-HIV agents II. Synthesis
and in vitro anti-HIV activity of novel 1H,3H-thiazolo[3,4-a]benzimidazoles. Farmaco 1991;46:
925933. (b) Buckheit RW, Jr, Hollingshead MG, Germany-Decker J, White EL, McMahon JB,
Allen LB, Ross LJ, Decker WD, Westbrook L, Shannon WM, Weislow O, Bader JP, Boyd MR.
Thiazolobenzimidazole: Biological and biochemical anti-retroviral activity of a new nonnucleoside
reverse transcriptase inhibitor. Antiviral Res 1993;21:247265.
22. Maass G, Immendoerfer U, Koenig B, Leser U, Mueller B, Goody R, Pfaff E. Viral resistance to
the thiazolo-iso-indolinones, a new class of nonnucleoside inhibitors of human immunodeciency
virus type 1 reverse transcriptase. Antimicrob Agents Chemother 1993;37:26122617.
23. De Lucca GV, Otto MJ. Synthesis and anti-HIV activity of pyrrolo[1,2-d]-(1,4)-benzodiazepin-6-
ones. Bioorg Med Chem Lett 1992;2:16391644.
24. Terrett NK, Bojanic D, Merson JR, Stephenson PT. Imidazo[20 -30 -:6,5]dipyrido[3,2-b:20 ,30 -e]-1,4-
diazepines: Non-nucleoside HIV-1 reverse transcriptase inhibitors with greater enzyme afnity
than nevirapine. Bioorg Med Chem Lett 1992;2:17451750.
25. Bellarosa D, Antonelli G, Bambacioni F, Giannotti D, Viti G, Nannicini R, Giachetti A,
Dianzani F, Witvrouw M, Pauwels R, Desmyter J, De Clercq E. New arylpyrido-diazepine and
-thiodiazepine derivatives are potent and highly selective HIV-1 inhibitors targeted at the reverse
transcriptase. Antiviral Res 1996;30:109124.
26. (a) Campiani G, Nacci V, Fiorini I, De Filippis MP, Garofalo A, Greco G, Novellino E,
Altamura S, Di Renzo L. Pyrrolobenzothiazepinones and pyrrolobenzoxazepinones: Novel and
specic non-nucleoside HIV-1 reverse transcriptase inhibitors with antiviral activity. J Med Chem
1996;39:26722680. (b) Campiani G, Morelli E, Fabbrini M, Nacci V, Greco G, Novellino E,
Ramunno A, Maga G, Spadari S, Caliendo G, Bergamini A, Faggioli E, Uccella I, Bolacchi F,
Marini S, Coletta M, Nacca A, Caccia S. Pyrrolobenzoxazepinone derivatives as non-nucleoside
HIV-1 RT inhibitors: Further structure-activity relationship studies and identication of more
potent broad-spectrum HIV-1 RT inhibitors with antiviral activity. J Med Chem 1999;42:
44624470.
27. Silvestri R, Artico M, Bruno B, Massa S, Novellino E, Greco G, Marongiu ME, Pani A,
De Montis A, La Colla P. Synthesis and biological evaluation of 5H-indolo [3,2-b][1,5]benzothia-
zepine derivatives, designed as conformationally constrained analogues of the human immuno-
deciency virus type 1 reverse transcriptase inhibitor L-737,126. Antiviral Chem Chemother 1998;
9:139148.
28. Livermore DGH, Bethell RC, Cammack N, Hancock AP, Hann MM, Green DVS, Lamont RB,
Noble SA, Orr DC, Payne JJ, Ramsay MVJ, Shingler AH, Smith C, Storer R, Williamson C,
Willson T. Synthesis and anti-HIV-1 activity of a series of imidazo[1,5-b]pyridazines. J Med Chem
1993;36:37843794.
29. Witvrouw M, Arranz ME, Pannecouque C, Declercq R, Jonckheere H, Schmit JC,
Vandamme AM, Diaz JA, Ingate ST, Desmyter J, Esnouf R, Van Meervelt L, Vega S,
Balzarini J, De Clercq E. 1,1,3-Trioxo-2H,4H-thieno[3,4-e][1,2,4]thiadiazine (TTD) derivatives: A
new class of nonnucleoside human immunodeciency virus type 1 (HIV-1) reverse transcriptase
inhibitors (NNRTIs) with anti-HIV-1 activity. Antimicrob Agents Chemother 1998:42:618623.
30. Krikorian D, Parushev S, Tarpanov V, Mechkarova P, Mikhova B, Botta M, Corelli F, Maga G,
Spadari S. Synthesis and biological evaluation of imidazo[1,2-d][1,4]benzodiazepines and related
compounds as potential anti-HIV-1 agents. Med Chem Res 1997:7;546556.
31. Taylor DL, Ahmed PS, Tyms AS, Bedard J, Duchaine J, Falardeau G, Lavallee JF, Hamel M,
Rando RF, Bowlin T. A pyrido [1,2a] indole derivative identied as a novel non-nucleoside reverse

Medicinal Research Reviews DOI 10.1002/med


52
E52 KK ZHAN
ZHANETAL.
ETAL.

transcriptase inhibitor of HIV-1. Abstract 11th Int. Conf. Antiviral Res., San Diego, CA, April
510, 1998, Antiviral Res 1998:37:A53(abstract no. 44).
32. Krajewski K, Zhang Y, Parrish D, Deschamps J, Roller PP, Pathak VK. New HIV-1 reverse
transcriptase inhibitors based on a tricyclic benzothiophene scaffold: Synthesis, resolution, and
inhibitory activity. Bioorg Med Chem Lett 2006;16:30343038.
33. (a) Kashman Y, Gustafson KR, Fuller RW, Cardellina JH, 2nd, McMahon JB, Currens MJ,
Buckheit RW, Jr, Hughes SH, Cragg GM, Boyd MR. The calanolides, a novel HIV-inhibitory
class of coumarin derivatives from the tropical rainforest tree, Calophyllum lanigerum. J Med
Chem 1992;35:27352743. (b) Kostova I. Coumarins as inhibitors of HIV reverse transcriptase.
Curr HIV Res 2006;4:347363. (c) Yu D, Suzuki M, Xie L, Morris-Natschke SL, Lee KH. Recent
progress in the development of coumarin derivatives as potent anti-HIV agents. Med Res Rev
2003;23:322345. (d) Kostova I, Raleva S, Genova P, Argirova R. Structure-activity relationships
of synthetic coumarins as HIV-1 inhibitors. Bioinorg Chem Appl 2006:68274.
34. Artico M, Silvestri R, Pagnozzi E, Stefancich G, Massa S, Loi AG, Putzolu M, Corrias S,
Spiga MG, La Colla P. 5H-pyrrolo[1,2-b] [1,2,5]benzothiadiazepines (PBTDs): A novel class of
non-nucleoside reverse transcriptase inhibitors. Bioorg Med Chem 1996;4:837850.
35. Corbett JW, Pan S, Markwalder JA, Cordova BC, Klabe RM, Garber S, Rodgers JD, Erickson-
Viitanen SK. 3,3a-Dihydropyrano[4,3,2-de]quinazolin-2(1H)-ones are potent non-nucleoside
reverse transcriptase inhibitors. Bioorg Med Chem Lett 2001;11:211214.
36. Zhang Z, Walker M, Xu W, Shim JH, Girardet JL, Hamatake RK, Hong Z. Novel nonnucleoside
inhibitors that select nucleoside inhibitor resistance mutations in human immunodeciency virus
type 1 reverse transcriptase. Antimicrob Agents Chemother 2006;50:27722781.
37. (a) Dueweke TJ, Kezdy FJ, Waszak GA, Deibel MR, Jr, Tarpley WG. The binding of a novel
bisheteroarylpiperazine mediates inhibition of human immunodeciency virus type 1 reverse
transcriptase. J Biol Chem 1992;267:2730. (b) Dueweke TJ, Poppe SM, Romero DL,
Swaney SM, So AG, Downey KM, Althaus IW, Reusser F, Busso M, Resnick L, Mayers DL,
Lane J, Aristoff PA, Thomas RC, Gary Tarpley W. U-90152, a potent inhibitor of human
immunodeciency virus type 1 replication. Antimicrob Agents Chemother 1993;37:11271131.
38. Kleim JP, Bender R, Billhardt UM, Meichsner C, Riess G, Rosner M, Winkler I, Paessens A.
Activity of a novel quinoxaline derivative against human immunodeciency virus type 1 reverse
transcriptase and viral replication. Antimicrob Agents Chemother 1993;37:16591664.
39. (a) Williams TM, Ciccarone TM, MacTough SC, Rooney CS, Balani SK, Condra JH, Emini EA,
Goldman ME, Greenlee WJ, Kauffman LR, OBrien JA, Sardana VV, Schleif WA,
Theoharides AD, Anderson PS. 5-Chloro-3-(phenylsulfonyl)indole-2-carboxamide: A novel,
non-nucleoside inhibitor of HIV-1 reverse transcriptase. J Med Chem 1993;36:12911294.
(b) Silvestri R, Artico M. Indolyl aryl sulfones (IASs): Development of highly potent NNRTIs
active against wt-HIV-1 and clinically relevant drug resistant mutants. Curr Pharm Des 2005;11:
37793806.
40. Buckheit RW, Jr, Fliakas-Boltz V, Decker WD, Roberson JL, Pyle CA, White EL, Bowdon BJ,
McMahon JB, Boyd MR, Bader JP, Nickell DG, Barth H, Antonucci TK. Biological and
biochemical anti-HIV activity of the benzothiadiazine class of nonnucleoside reverse transcriptase
inhibitors. Antiviral Res 1994;25:4356.
41. Tucker TJ, Lyle TA, Wiscount CM, Britcher SF, Young SD, Sanders WM, Lumma WC,
Goldman ME, OBrien JA, Ball RG, Homnick CF, Schleif WA, Emini EA, Huff JR,
Anderson PS. Synthesis of a series of 4-(arylethynyl)-6-chloro-4-cyclopropyl-3,4-dihydroquina-
zolin-2(1H)-ones as novel non-nucleoside HIV-1 reverse transcriptase inhibitors. J Med Chem
1994;37:24372444.
42. Young SD, Britcher SF, Tran LO, Payne LS, Lumma WC, Lyle TA, Huff JR, Anderson PS,
Olsen DB, Carroll SS, Pettibone DJ, OBrien JA, Ball RG, Balani SK, Lin JH, Chen I-WU,
SChleif WA, Sardana VV, Long WJ, Byrnes VW, Emini EA. L-743, 726 (DMP-266): A novel,
highly potent nonnucleoside inhibitor of the human immunodeciency virus type 1 reverse
transcriptase. Antimicrob Agents Chemother 1995;39:26022605.

Medicinal Research Reviews DOI 10.1002/med


HIV-1
HIV-1NNRTIs
NNRTIs KK 53
E53

43. Di Santo R, Costi R, Artico M, Massa S, Marongiu ME, Loi AG, De Montis A, La Colla P.
1,2,5-Benzothiadiazepine and pyrrolo[2,1-d][1,2,5]benzothiadiazepine derivatives with specic
anti-human immunodeciency virus type 1 activity. Antiviral Chem Chemother 1998;9:
127137.
44. Jiang T, Kuhen KL, Wolff K, Yin H, Bieza K, Caldwell J, Bursulaya B, Wu TY, He Y. Design,
synthesis and biological evaluations of novel oxindoles as HIV-1 non-nucleoside reverse
transcriptase inhibitors. Part I. Bioorg Med Chem Lett 2006;16:21052108.
45. (a) Ellis D, Kuhen KL, Anaclerio B, Wu B, Wolff K, Yin H, Bursulaya B, Caldwell J,
Karanewsky D, He Y. Design, synthesis, and biological evaluations of novel quinolones as HIV-1
non-nucleoside reverse transcriptase inhibitors. Bioorg Med Chem Lett 2006;16:42464251.
(b) Hopkins AL, Ren J, Milton J, Hazen RJ, Chan JH, Stuart DI, Stammers DK. Design of
non-nucleoside inhibitors of HIV-1 reverse transcriptase with improved drug resistance properties.
1. J Med Chem 2004;47:59125922. (c) Barreca ML, De Luca L, Iraci N, Rao A, Ferro S,
Maga G, Chimirri A. Structure-based pharmacophore identication of new chemical scaffolds as
non-nucleoside reverse transcriptase inhibitors. J Chem Inf Model 2007;47:557562.
46. Barreca ML, Rao A, De Luca L, Iraci N, Monforte AM, Maga G, De Clercq E, Pannecouque C,
Balzarini J, Chimirri A. Discovery of novel benzimidazolones as potent non-nucleoside reverse
transcriptase inhibitors active against wild-type and mutant HIV-1 strains. Bioorg Med Chem Lett
2007;17:19561960.
47. Kroeger Smith MB, Hose BM, Hawkins A, Lipchock J, Farnsworth DW, Rizzo RC, Tirado-
Rives J, Arnold E, Zhang W, Hughes SH, Jorgensen WL, Michejda CJ, Smith RH, Jr. Molecular
modeling calculations of HIV-1 reverse transcriptase nonnucleoside inhibitors: Correlation of
binding energy with biological activity for novel 2-aryl-substituted benzimidazole analogues.
J Med Chem 2003;46:19401947. (b) Morningstar ML, Roth T, Farnsworth DW, Smith MK,
Watson K, Buckheit RW, Jr, Das K, Zhang W, Arnold E, Julias JG, Hughes SH, Michejda CJ.
Synthesis, biological activity, and crystal structure of potent nonnucleoside inhibitors of HIV-1
reverse transcriptase that retain activity against mutant forms of the enzyme. J Med Chem
2007;50:40034015. (c) Substituted benzimidazoles as non-nucleoside inhibitors of reverse
transcriptase. (WO 9837072 A1 19980827)
48. Ludovici DW, Kavash RW, Kukla MJ, Ho CY, Ye H, De Corte BL, Andries K, de Bethune MP,
Azijn H, Pauwels R, Moereels HE, Heeres J, Koymans LM, de Jonge MR, Van Aken KJ,
Daeyaert FF, Lewi PJ, Das K, Arnold E, Janssen PA. Evolution of anti-HIV drug candidates.
Part 2: Diaryltriazine (DATA) analogues. Bioorg Med Chem Lett 2001;11:22292234.
49. (a) Ludovici DW, De Corte BL, Kukla MJ, Ye H, Ho CY, Lichtenstein MA, Kavash RW,
Andries K, de Bethune MP, Azijn H, Pauwels R, Lewi PJ, Heeres J, Koymans LM, de Jonge MR,
Van Aken KJ, Daeyaert FF, Das K, Arnold E, Janssen PA. Evolution of anti-HIV drug
candidates. Part 3: Diarylpyrimidine (DAPY) analogues. Bioorg Med Chem Lett 2001;11:
22352239. (b) Das K, Clark AD, Jr, Lewi PJ, Heeres J, De Jonge MR, Koymans LM,
Vinkers HM, Daeyaert F, Ludovici DW, Kukla MJ, De Corte B, Kavash RW, Ho CY, Ye H,
Lichtenstein MA, Andries K, Pauwels R, De Bethune MP, Boyer PL, Clark P, Hughes SH,
Janssen PA, Arnold E. Roles of conformational and positional adaptability in structure-based
design of TMC125-R165335 (etravirine) and related non-nucleoside reverse transcriptase
inhibitors that are highly potent and effective against wild-type and drug-resistant HIV-1
variants. J Med Chem 2004;47:25502560.
50. Heeres J, de Jonge MR, Koymans LM, Daeyaert FF, Vinkers M, Van Aken KJ, Arnold E, Das K,
Kilonda A, Hoornaert GJ, Compernolle F, Cegla M, Azzam RA, Andries K, de Bethune MP,
Azijn H, Pauwels R, Lewi PJ, Janssen PA. Design, synthesis, and SAR of a novel pyrazinone
series with non-nucleoside HIV-1 reverse transcriptase inhibitory activity. J Med Chem 2005;48:
19101918.
51. Tian X, Qin B, Lu H, Lai W, Jiang S, Lee KH, Chen CH, Xie L. Discovery of diarylpyridine
derivatives as novel non-nucleoside HIV-1 reverse transcriptase inhibitors. Bioorg Med Chem Lett
2009;19:54825485.

Medicinal Research Reviews DOI 10.1002/med


54
E54 KK ZHAN
ZHANETAL.
ETAL.

52. Qin B, Jiang X, Lu H, Tian X, Barbault F, Huang L, Qian K, Chen CH, Huang R, Jiang S,
Lee KH, Xie L. Diarylaniline derivatives as a distinct class of HIV-1 non-nucleoside reverse
transcriptase inhibitors. J Med Chem 2010;53:49064916.
53. Baba M, Tanaka H, De Clercq E, Pauwels R, Balzarini J, Schols D, Nakashima H, Perno CF,
Walker RT, Miyasaka T. Highly specic inhibition of human immunodeciency virus type 1
by a novel 6-substituted acyclouridine derivative. Biochem Biophys Res Commun 1989;165:
13751381.
54. Kim DK, Gam J, Kim YW, Lim J, Kim HT, Kim KH. Synthesis and anti-HIV-1 activity of a
series of 1-alkoxy-5-alkyl-6-(arylthio)uracils. J Med Chem 1997;40:23632373.
55. (a) Artico M, Massa S, Mai A, Marongiu ME, Piras G, Tramontano E, La Colla P. 3,4-Dihydro-
2-alkoxy-6-benzyl-4-oxopyrimidines (DABOs): A new class of specic inhibitors of human
immunodeciency virus type 1. Antiviral Chem Chemother 1993;4:361368. (b) Artico M.
Selected non-nucleoside reverse transcriptase inhibitors (NNRTIs): The DABOs family. Drugs
Fut 2002;27:159175.
56. (a) Goldman ME, Nunberg JH, OBrien JA, Quintero JC, Schleif WA, Freund KF, Gaul SL,
Saari WS, Wai JS, Hoffman JM, Anderson PS, Hupe DJ, Emini EA, Stern AM. Pyridinone
derivatives: Specic human immunodeciency virus type 1 reverse transcriptase inhibitors with
antiviral activity. Proc Natl Acad Sci USA 1991;88:68636867. (b) Saari WS, Hoffman JM,
Wai JS, Fisher TE, Rooney CS, Smith AM, Thomas CM, Goldman ME, OBrien JA,
Nunberg JH, Quintero JC, Schleif WA, Emini EA, Stern AM, Anderson PS. 2-Pyridinone
derivatives: A new class of nonnucleoside, HIV-1-specic reverse transcriptase inhibitors. J Med
Chem 1991;34:29222925. (c) Medina-Franco JL, Martnez-Mayorga K, Juarez-Gordiano C,
Castillo R. Pyridin-2(1H)-ones: A promising class of HIV-1 non-nucleoside reverse transcriptase
inhibitors. Chem Med Chem 2007;2:11411147.
57. Althaus IW, Chou KC, Lemay RJ, Franks KM, Deibel MR, Kezdy FJ, Resnick L, Busso ME,
So AG, Downey KM, Romero DL, Thomas RC, Aristoff PA, Tarpley WG, Reusser F. The
benzylthio-pyrimidine U-31,355, a potent inhibitor of HIV-1 reverse transcriptase. Biochem
Pharmacol 1996;51:743750. (b) Nugent RA, Schlachter ST, Murphy MJ, Cleek GJ, Poel TJ,
Wishka DG, Graber DR, Yagi Y, Keiser BJ, Olmsted RA, Kopta LA, Swaney SM, Poppe SM,
Morris J, Tarpley WG, Thomas RC. Pyrimidine thioethers: a novel class of HIV-1 reverse
transcriptase inhibitors with activity against BHAP-resistant HIV. J Med Chem 1998;41:
37933803. (c) Nugent RA, Wishka DG, Cleek GJ, Graber DR, Schalachter ST, Murphy MJ,
Morris J, Thomas RC. Preparation of 2-pyrimidino alkyl ethers and thioethers as inhibitors of
viral reverse transcriptase. WO/9635678.
58. Wishka DG, Graber DR, Kopta LA, Olmsted RA, Friis JM, Hosley JD, Adams WJ, Seest EP,
Castle TM, Dolak LA, Keiser BJ, Yagi Y, Jeganathan A, Schlachter ST, Murphy MJ, Cleek GJ,
Nugent RA, Poppe SM, Swaney SM, Han F, Watt W, White WL, Poel TJ, Thomas RC,
Voorman RL, Stefanski KJ, Stehle RG, Tarpley WG, Morris J. (-)-6-Chloro-2-[(1-furo[2,3-
c]pyridin-5-ylethyl)thio]-4-pyrimidinamine, PNU-142721, a new broad spectrum HIV-1 non-
nucleoside reverse transcriptase inhibitor. J Med Chem 1998;41:13571360.
59. (a) Jorgensen WL, Ruiz-Caro J, Tirado-Rives J, Basavapathruni A, Anderson KS, Hamilton AD.
Computer-aided design of non-nucleoside inhibitors of HIV-1 reverse transcriptase. Bioorg Med
Chem Lett 2006;16:663667. (b) Ruiz-Caro J, Basavapathruni A, Kim JT, Bailey CM, Wang L,
Anderson KS, Hamilton AD, Jorgensen WL. Optimization of diarylamines as non-nucleoside
inhibitors of HIV-1 reverse transcriptase. Bioorg Med Chem Lett 2006;16:668671.
(c) Thakur VV, Kim JT, Hamilton AD, Bailey CM, Domaoal RA, Wang L, Anderson KS,
Jorgensen WL. Optimization of pyrimidinyl- and triazinyl-amines as non-nucleoside inhibitors of
HIV-1 reverse transcriptase. Bioorg Med Chem Lett 2006;16:56645667.
60. Ijichi K, Fujiwara M, Hanasaki Y, Watanabe H, Katsuura K, Takayama H, Shirakawa S,
Sakai SI, Shigeta S, Konno K, Yokota T, Baba M. Potent and specic inhibition of human
immunodeciency virus type 1 replication by 4-(2,6-dichlorophenyl)-1,2,5-thiadiazol-3-Y1 N,N-
dialkylcarbamate derivatives. Antimicrob Agents Chemother 1995;39:23372340.

Medicinal Research Reviews DOI 10.1002/med


HIV-1
HIV-1NNRTIs
NNRTIs KK 55
E55

61. Antonucci T, Warmus JS, Hodges JC, Nickell DG. Characterization of the antiviral activity of
highly substituted pyrroles: A novel class of non-nucleoside HIV-1 reverse transcriptase inhibitor.
Antiviral Chem Chemother 1995;6:98108. (b) Williams TM, Zhang XF, Obligado VE,
Poehnelt RA. Preparation of pyrrole carboxamides as non-nucleoside reverse transcriptase
inhibitors. WO 2008054605 A2.
62. (a) Barreiro G, Guimaraes CR, Tubert-Brohman I, Lyons TM, Tirado-Rives J, Jorgensen WL.
Search for non-nucleoside inhibitors of HIV-1 reverse transcriptase using chemical similarity,
molecular docking, and MM-GB/SA scoring. J Chem Inf Model 2007;47:24162428.
(b) Barreiro G, Kim JT, Guimaraes CR, Bailey CM, Domaoal RA, Wang L, Anderson KS,
Jorgensen WL. From docking false-positive to active anti-HIV agent. J Med Chem 2007;50:
53245329. (c) Zeevaart JG, Wang L, Thakur VV, Leung CS, Tirado-Rives J, Bailey CM,
Domaoal RA, Anderson KS, Jorgensen WL. Optimization of azoles as anti-human immunode-
ciency virus agents guided by free-energy calculations. J Am Chem Soc 2008;130:94929499.
(d) Leung CS, Zeevaart JG, Domaoal RA, Bollini M, Thakur VV, Spasov KA, Anderson KS,
Jorgensen WL. Eastern extension of azoles as non-nucleoside inhibitors of HIV-1 reverse
transcriptase; cyano group alternatives. Bioorg Med Chem Lett 2010;20:24852488.
63. (a) Fujiwara T, Sato A, el-Farrash M, Miki S, Abe K, Isaka Y, Kodama M, Wu Y, Chen LB,
Harada H, Sugimoto H, Hatanaka M, Hinuma Y. S-1153 inhibits replication of known drug-
resistant strains of human immunodeciency virus type 1. Antimicrob Agents Chemother 1998;42:
13401345. (b) Gewurz BE, Jacobs M, Proper JA, Dahl TA, Fujiwara T, Dezube BJ. Capravirine,
a nonnucleoside reverse-transcriptase inhibitor in patients infected with HIV-1: A phase 1 study.
J Infect Dis 2004;190:19571961. (c) Sato A, Hammond J, Alexander TN, Graham JP, Binford S,
Sugita K, Sugimoto H, Fujiwara T, Patick AK. In vitro selection of mutations in human
immunodeciency virus type 1 reverse transcriptase that confer resistance to capravirine, a novel
nonnucleoside reverse transcriptase inhibitor. Antiviral Res 2006;70:6674.
64. Genin MJ, Biles C, Keiser BJ, Poppe SM, Swaney SM, Tarpley WG, Yagi Y, Romero DL.
Novel 1,5-diphenylpyrazole nonnucleoside HIV-1 reverse transcriptase inhibitors with enhanced
activity versus the delavirdine-resistant P236L mutant: Lead identication and SAR of 3- and
4-substituted derivatives. J Med Chem 2000;43:10341040.
65. Barreca ML, Chimirri A, De Luca L, Monforte AM, Monforte P, Rao A, Zappala M, Balzarini J,
De Clercq E, Pannecouque C, Witvrouw M. Discovery of 2,3-diaryl-1,3-thiazolidin-4-ones as
potent anti-HIV-1 agents. Bioorg Med Chem Lett 2001;11:17931796.
66. Wu B, Kuhen K, Ngoc Nguyen T, Ellis D, Anaclerio B, He X, Yang K, Karanewsky D, Yin H,
Wolff K, Bieza K, Caldwell J, He Y. Synthesis and evaluation of N-aryl pyrrolidinones as novel
anti-HIV-1 agents. Part 1. Bioorg Med Chem Lett 2006;16:34303433. (b) Wu B, Nguyen TN,
Ellis DA, He X, Anaclerio BM, Yang K, Chol H-S, Wang Z, Marsilje T, He Y. Phenyl-substituted
pyrrolidones. 2006; WO2006081544.
67. (a) Zhan P, Li Z, Liu X, De Clercq E. Sulfanyltriazole/tetrazoles: A promising class of HIV-1
NNRTIs. Mini Rev Med Chem 2009;9:10141023. (b) Wang Z, Wu B, Kuhen KL, Bursulaya B,
Nguyen TN, Nguyen DG, He Y. Synthesis and biological evaluations of sulfanyltriazoles as novel
HIV-1 non-nucleoside reverse transcriptase inhibitors. Bioorg Med Chem Lett 2006;16:41744177.
(c) De La Rosa M, Kim HW, Gunic E, Jenket C, Boyle U, Koh YH, Korboukh I, Allan M,
Zhang W, Chen H, Xu W, Nilar S, Yao N, Hamatake R, Lang SA, Hong Z, Zhang Z,
Girardet JL. Tri-substituted triazoles as potent non-nucleoside inhibitors of the HIV-1 reverse
transcriptase. Bioorg Med Chem Lett 2006;16:44444449. (d) Muraglia E, Kinzel OD, Laufer R,
Miller MD, Moyer G, Munshi V, Orvieto F, Palumbi MC, Pescatore G, Rowley M, Williams PD,
Summa V. Tetrazole thioacetanilides: Potent non-nucleoside inhibitors of WT HIV reverse
transcriptase and its K103N mutant. Bioorg Med Chem Lett 2006;16:27482752. (e) OMeara JA,
Jakalian A, LaPlante S, Bonneau PR, Coulombe R, Faucher AM, Guse I, Landry S, Racine J,
Simoneau B, Thavonekham B, Yoakim C. Scaffold hopping in the rational design of novel HIV-1
non-nucleoside reverse transcriptase inhibitors. Bioorg Med Chem Lett 2007;17:33623366.
(f) Gagnon A, Amad MH, Bonneau PR, Coulombe R, DeRoy PL, Doyon L, Duan J, Garneau M,
Guse I, Jakalian A, Jolicoeur E, Landry S, Malenfant E, Simoneau B, Yoakim C. Thiotetrazole

Medicinal Research Reviews DOI 10.1002/med


56
E56 KK ZHAN
ZHANETAL.
ETAL.

alkynylacetanilides as potent and bioavailable non-nucleoside inhibitors of the HIV-1 wild type
and K103N/Y181C double mutant reverse transcriptases. Bioorg Med Chem Lett 2007;17:
44374441. (g) Zhang Z, Xu W, Koh YH, Shim JH, Girardet JL, Yeh LT, Hamatake RK,
Hong Z. A novel nonnucleoside analogue that inhibits human immunodeciency virus type 1
isolates resistant to current nonnucleoside reverse transcriptase inhibitors. Antimicrob Agents
Chemother 2007;51:429437. (h) Moyle G, Bofto M, Stoehr A, Rieger A, Shen Z, Manhard K,
Sheedy B, Hingorani V, Raney A, Nguyen M, Nguyen T, Ong V, Yeh LT, Quart B. Phase 2a
randomized controlled trial of short-term activity, safety, and pharmacokinetics of a novel
nonnucleoside reverse transcriptase inhibitor, RDEA806, in HIV-1-positive, antiretroviral-naive
subjects. Antimicrob Agents Chemother 2010;54:31703178.
68. (a) Masuda N, Yamamoto O, Fujii M, Ohgami T, Fujiyasu J, Kontani T, Moritomo A, Orita M,
Kurihara H, Koga H, Nakahara H, Kageyama S, Ohta M, Inoue H, Hatta T, Suzuki H, Sudo K,
Shimizu Y, Kodama E, Matsuoka M, Fujiwara M, Yokota T, Shigeta S, Baba M. Studies of
nonnucleoside HIV-1 reverse transcriptase inhibitors. Part 1: Design and synthesis of
thiazolidenebenzenesulfonamides. Bioorg Med Chem 2004;12:61716182. (b) Masuda N,
Yamamoto O, Fujii M, Ohgami T, Fujiyasu J, Kontani T, Moritomo A, Orita M, Kurihara H,
Koga H, Kageyama S, Ohta M, Inoue H, Hatta T, Shintani M, Suzuki H, Sudo K, Shimizu Y,
Kodama E, Matsuoka M, Fujiwara M, Yokota T, Shigeta S, Baba M. Studies of non-nucleoside
HIV-1 reverse transcriptase inhibitors. Part 2: Synthesis and structure-activity relationships of
2-cyano and 2-hydroxy thiazolidenebenzenesulfonamide derivatives. Bioorg Med Chem 2005;13:
949961.
69. Pauwels R, Andries K, Debyser Z, Van Daele P, Schols D, Stoffels P, De Vreese K,
Woestenborghs R, Vandamme AM, Janssen CG, Anne J, Cauwenbergh G, Desmyter J,
Heykants J, Janssen MA, De Clercq E, Janssen PA. Potent and highly selective human
immunodeciency virus type 1 (HIV-1) inhibition by a series of alpha-anilinophenylacetamide
derivatives targeted at HIV-1 reverse transcriptase. Proc Natl Acad Sci USA 1993;90:17111715.
70. Ludovici DW, Kukla MJ, Grous PG, Krishnan S, Andries K, de Bethune MP, Azijn H,
Pauwels R, De Clercq E, Arnold E, Janssen PA. Evolution of anti-HIV drug candidates. Part 1:
From alpha-anilinophenylacetamide (alpha-APA) to imidoyl thiourea (ITU). Bioorg Med Chem
Lett 2001;11:22252228.
71. Balzarini J, Brouwer WG, Felauer EE, De Clercq E, Karlsson A. Activity of various
thiocarboxanilide derivatives against wild-type and several mutant human immunodeciency
virus type 1 strains. Antiviral Res 1995;27:219236.
72. (a) Bell FW, Cantrell AS, Hogberg M, Jaskunas SR, Johansson NG, Jordan CL, Kinnick MD,
Lind P, Morin JM, Jr, Noreen R, Oberg B, Palkowitz JA, Parrish CA, Pranc P, Sahlberg C,
Ternansky RJ, Vasileff RT, Vrang L, West SJ, Zhang H, Zhou XX. Phenethylthiazolethiourea
(PETT) compounds, a new class of HIV-1 reverse transcriptase inhibitors. 1. Synthesis and basic
structure-activity relationship studies of PETT analogs. J Med Chem 1995;38:49294936.
(b) Cantrell AS, Engelhardt P, Hogberg M, Jaskunas SR, Johansson NG, Jordan CL,
Kangasmetsa J, Kinnick MD, Lind P, Morin JM, Jr, Muesing MA, Noreen R, Oberg B,
Pranc P, Sahlberg C, Ternansky RJ, Vasileff RT, Vrang L, West SJ, Zhang H. Phenethylthia-
zolylthiourea (PETT) compounds as a new class of HIV-1 reverse transcriptase inhibitors. 2.
Synthesis and further structure-activity relationship studies of PETT analogs. J Med Chem 1996;
39:42614274. (c) Ahgren C, Backro K, Bell FW, Cantrell AS, Clemens M, Colacino JM,
Deeter JB, Engelhardt JA, Hogberg M, Jaskunas SR, Johansson NG, Jordan CL, Kasher JS,
Kinnick MD, Lind P, Lopez C, Morin JM, Jr, Muesing MA, Noreen R, Oberg B, Paget CJ,
Palkowitz JA, Parrish CA, Pranc P, Rippy MK, Rydergard C, Sahlberg C, Swanson S,
Ternansky RJ, Unge T, Vasileff RT, Vrang L, West SJ, Zhang H, Zhou XX. The PETT series, a
new class of potent nonnucleoside inhibitors of human immunodeciency virus type 1 reverse
transcriptase. Antimicrob Agents Chemother 1995;39:13291335.
73. Campiani G, Aiello F, Fabbrini M, Morelli E, Ramunno A, Armaroli S, Nacci V, Garofalo A,
Greco G, Novellino E, Maga G, Spadari S, Bergamini A, Ventura L, Bongiovanni B, Capozzi M,
Bolacchi F, Marini S, Coletta M, Guiso G, Caccia S. Quinoxalinylethylpyridylthioureas (QXPTs)

Medicinal Research Reviews DOI 10.1002/med


HIV-1NNRTIs
HIV-1 NNRTIs KK 57
E57

as potent non-nucleoside HIV-1 reverse transcriptase (RT) inhibitors. Further SAR studies and
identication of a novel orally bioavailable hydrazine-based antiviral agent. J Med Chem 2001;44:
305315.
74. (a) Sahlberg C, Noreen R, Engelhardt P, Hogberg M, Kangasmetsa J, Vrang L, Zhang H.
Synthesis and anti-HIV activities of urea-PETT analogs belonging to a new class of potent non-
nucleoside HIV-1 reverse transcriptase inhibitors. Bioorg Med Chem Lett 1998;8:15111516.
(b) Hogberg M, Sahlberg C, Engelhardt P, Noreen R, Kangasmetsa J, Johansson NG, Oberg B,
Vrang L, Zhang H, Sahlberg BL, Unge T, Lovgren S, Fridborg K, Backbro K. Urea-PETT
compounds as a new class of HIV-1 reverse transcriptase inhibitors. 3. Synthesis and further
structure-activity relationship studies of PETT analogues. J Med Chem 1999;42:41504160.
(c) Dmitry A, Christian S, Stefan L, Christer S. Preparation of pyridinyl(thio)urea derivatives of
cyclopropachromenes, cyclopropaindenes, cyclopropanaphthalenes, and benzocyclopropacyclo-
heptenes as non-nucleoside reverse transcriptase inhibitors. WO 2004021969 A2. (d) Sund C,
Roue N. Non-nucleotide reverse transcriptase inhibitors. WO 2005066131 A1.
75. Cesarini S, Spallarossa A, Ranise A, Fossa P, La Colla P, Sanna G, Collu G, Loddo R.
Thiocarbamates as non-nucleoside HIV-1 reverse transcriptase inhibitors. Part 1: Parallel
synthesis, molecular modelling and structure-activity relationship studies on O-[2-(hetero)aryl-
ethyl]-N-phenylthiocarbamates. Bioorg Med Chem 2008;16:41604172.
76. (a) Cushman M, Golebiewski WM, Graham L, Turpin JA, Rice WG, Fliakas-Boltz V,
Buckheit RW, Jr. Synthesis and biological evaluation of certain alkenyldiarylmethanes as anti-
HIV-1 agents which act as non-nucleoside reverse transcriptase inhibitors. J Med Chem 1996;39:
32173227. (b) Zhan P, Li Z, Liu X. Cosalane and its analogues: A unique class of anti-HIV
agents. Mini Rev Med Chem 2010;10:966976.
77. Hara H, Fujihashi T, Sakata T, Kaji A, Kaji H. Tetrahydronaphthalene lignan compounds as
potent anti-HIV type 1 agents. AIDS Res Hum Retroviruses 1997;13:695705.
78. Chan JH, Hong JS, Hunter RN, 3rd, Orr GF, Cowan JR, Sherman DB, Sparks SM, Reitter BE,
Andrews CW, 3rd, Hazen RJ, St Clair M, Boone LR, Ferris RG, Creech KL, Roberts GB,
Short SA, Weaver K, Ott RJ, Ren J, Hopkins A, Stuart DI, Stammers DK. 2-Amino-6-
arylsulfonylbenzonitriles as non-nucleoside reverse transcriptase inhibitors of HIV-1. J Med Chem
2001;44:18661882.
79. Jones LH, Allan G, Barba O, Burt C, Corbau R, Dupont T, Knochel T, Irving S, Middleton DS,
Mowbray CE, Perros M, Ringrose H, Swain NA, Webster R, Westby M, Phillips C. Novel
indazole non-nucleoside reverse transcriptase inhibitors using molecular hybridization based on
crystallographic overlays. J Med Chem 2009;52:12191223.
80. Wyatt PG, Bethell RC, Cammack N, Charon D, Dodic N, Dumaitre B, Evans DN, Green DV,
Hopewell PL, Humber DC, Lamont RB, Orr DC, Plested SJ, Ryan DM, Sollis SL, Storer R,
Weingartenten GG. Benzophenone derivatives: A novel series of potent and selective inhibitors of
human immunodeciency virus type 1 reverse transcriptase. J Med Chem 1995;38:16571665.
81. Tucker TJ, Saggar S, Sisko JT, Tynebor RM, Williams TM, Felock PJ, Flynn JA, Lai MT,
Liang Y, McGaughey G, Liu M, Miller M, Moyer G, Munshi V, Perlow-Poehnelt R, Prasad S,
Sanchez R, Torrent M, Vacca JP, Wan BL, Yan Y. The design and synthesis of diaryl ether second
generation HIV-1 non-nucleoside reverse transcriptase inhibitors (NNRTIs) with enhanced
potency versus key clinical mutations. Bioorg Med Chem Lett 2008;18:29592966.
82. Silvestri R, Artico M, Massa S, Marceddu T, De Montis F, La Colla P. 1-[2-(Diphenylmethoxy)-
ethyl]-2-methyl-5-nitroimidazole: A potent lead for the design of novel NNRTIs. Bioorg Med
Chem Lett 2000;10:253256.
83. Xie L, Xie JX, Kashiwada Y, Cosentino LM, Liu SH, Pai RB, Cheng YC, Lee KH. Anti-AIDS
(acquired immune deciency syndrome) agents. 17. New brominated hexahydroxybiphenyl
derivatives as potent anti-HIV agents. J Med Chem 1995;38:30033008.
84. (a) Balzarini J, Perez-Perez MJ, San-Felix A, Schols D, Perno CF, Vandamme AM, Camarasa MJ, De
Clercq E. 20 ,50 -Bis-O-(tert-butyldimethylsilyl)-30 -spiro-500 -(400 -amino-100 ,200 -oxathiole-200 ,20 -dioxide)-
pyrimidine (TSAO) nucleoside analogues: Highlyselective inhibitors of human immunodeciency

Medicinal Research Reviews DOI 10.1002/med


58
E58 KK ZHAN
ZHANETAL.
ETAL.

virus type 1 that are targeted at the viral reverse transcriptase. Proc Natl Acad Sci USA 1992;89:
43924396. (b) Camarasa MJ, San-Felix A, Velazquez S, Perez-Perez MJ, Gago F, Balzarini J.TSAO
compounds: The comprehensive story of a unique family of HIV-1 specic inhibitors of reverse
transcriptase. Curr Top Med Chem 2004;4:945963. (c) Camarasa MJ, Velazquez S, San-Felix A,
Perez-Perez MJ, Bonache MC, De Castro S. TSAO derivatives, inhibitors of HIV-1 reverse
transcriptase dimerization: Recent progress. Curr Pharm Des 2006;12:18951907.
85. Duarte CD, Barreiro EJ, Fraga CA. Privileged structures: A useful concept for the rational design
of new lead drug candidates. Mini Rev Med Chem 2007;7:11081119.
86. DeSimone RW, Currie KS, Mitchell SA, Darrow JW, Pippin DA. Privileged structures:
Applications in drug discovery. Comb Chem High Throughput Screen 2004;7:473494.
87. Fischer M, Hubbard RE. Fragment-based ligand discovery. Mol Interv 2009;9:2230.
88. Schafer W, Friebe WG, Leinert H, Mertens A, Poll T, von der Saal W, Zilch H, Nuber B,
Ziegler ML. Non-nucleoside inhibitors of HIV-1 reverse transcriptase: Molecular modeling and
X-ray structure investigations. J Med Chem 1993;36:726732.
89. (a) Sriram D, Bal TR, Yogeeswari P. Aminopyrimidinimino isatin analogues: Design and
synthesis of novel non- nucleoside HIV-1 reverse transcriptase inhibitors with broad-spectrum
anti-microbial properties. Med Chem 2005;1:277285. (b) Sriram D, Bal TR, Yogeeswari P.
Aminopyrimidinimino isatin analogues: Design of novel non- nucleoside HIV-1 reverse
transcriptase inhibitors with broad-spectrum chemotherapeutic properties. J Pharm Pharm Sci
2005;8:565577. (c) Sriram D, Bal TR, Yogeeswari P. Newer aminopyrimidinimino isatin
analogues as non-nucleoside HIV-1 reverse transcriptase inhibitors for HIV and other
opportunistic infections of AIDS: Design, synthesis and biological evaluation. Farmaco 2005;
60:377384. (d) Sriram D, Bal TR, Yogeeswari P. Design, synthesis and biological evaluation of
novel non-nucleoside HIV-1 reverse transcriptase inhibitors with broad-spectrum chemother-
apeutic properties. Bioorg Med Chem 2004;12:58655873.
90. Ren J, Esnouf R, Garman E, Somers D, Ross C, Kirby I, Keeling J, Darby G, Jones Y, Stuart D,
Stammers D. High resolution structures of HIV-1 RT from four RT-inhibitor complexes. Nat
Struct Biol 1995;2:293302.
91. Ding J, Das K, Moereels H, Koymans L, Andries K, Janssen PA, Hughes SH, Arnold E. Structure
of HIV-1 RT/TIBO R 86183 complex reveals similarity in the binding of diverse nonnucleoside
inhibitors. Nat Struct Biol 1995;2:407415.
92. Ren J, Esnouf R, Hopkins A, Ross C, Jones Y, Stammers D, Stuart D. The structure of HIV-1 reverse
transcriptase complexed with 9-chloro-TIBO: Lessons for inhibitor design. Structure 1995;3:915926.
93. Ren J, Stammers DK. Structural basis for drug resistance mechanisms for non-nucleoside
inhibitors of HIV reverse transcriptase. Virus Res 2008;134:157170.
94. Patel Y, Gillet VJ, Bravi G, Leach AR. A comparison of the pharmacophore identication
programs: Catalyst, DISCO and GASP. J Comput Aided Mol Des 2002;16:653681.
95. Iwase K, Hirono S. Estimation of active conformations of drugs by a new molecular superposing
procedure. J Comput Aided Mol Des 1999;13:499512.
96. Daeyaert F, de Jonge M, Heeres J, Koymans L, Lewi P, Vinkers MH, Janssen PA. A
pharmacophore docking algorithm and its application to the cross-docking of 18 HIV-NNRTIs
in their binding pockets. Proteins 2004;54:526533.
97. Daszykowski M, Walczak B, Xu QS, Daeyaert F, de Jonge MR, Heeres J, Koymans LM, Lewi PJ,
Vinkers HM, Janssen PA, Massart DL. Classication and regression treesStudies of HIV
reverse transcriptase inhibitors. J Chem Inf Comput Sci 2004;44:716726.
98. Barreca ML, Rao A, De Luca L, Zappala M, Monforte AM, Maga G, Pannecouque C,
Balzarini J, De Clercq E, Chimirri A, Monforte P. Computational strategies in discovering novel
non-nucleoside inhibitors of HIV-1 RT. J Med Chem 2005;48:34333437.
99. Wang J, Kang X, Kuntz ID, Kollman PA. Hierarchical database screenings for HIV-1 reverse
transcriptase using a pharmacophore model, rigid docking, solvation docking, and MM-PB/SA.
J Med Chem 2005;48:24322444.

Medicinal Research Reviews DOI 10.1002/med


HIV-1NNRTIs
HIV-1 NNRTIs KK 59
E59

100. Zhang Z, Zheng M, Du L, Shen J, Luo X, Zhu W, Jiang H. Towards discovering dual functional
inhibitors against both wild type and K103N mutant HIV-1 reverse transcriptases: Molecular
docking and QSAR studies on 4,1-benzoxazepinone analogues. J Comput Aided Mol Des 2006;
20:281293.
101. Esnouf RM, Ren J, Hopkins AL, Ross CK, Jones EY, Stammers DK, Stuart DI. Unique features
in the structure of the complex between HIV-1 reverse transcriptase and the bis(heteroaryl)piper-
azine (BHAP) U-90152 explain resistance mutations for this nonnucleoside inhibitor. Proc Natl
Acad Sci USA 1997;94:39843989.
102. Sluis-Cremer N, Temiz NA, Bahar I. Conformational changes in HIV-1 reverse transcriptase
induced by nonnucleoside reverse transcriptase inhibitor binding. Curr HIV Res 2004;2:323332.
103. Zhan P, Liu X, Li Z, Pannecouque C, De Clercq E. Design strategies of novel NNRTIs to
overcome drug resistance. Curr Med Chem 2009;16:39033917.
104. Fox S, Farr-Jones S, Sopchak L, Boggs A, Comley J. High-throughput screening: Searching for
higher productivity. J Biomol Screen 2004;9:354358.
105. Posner BA. High-throughput screening-driven lead discovery: Meeting the challenges of nding
new therapeutics. Curr Opin Drug Discov Dev 2005;8:487494.
106. Pettersson S, Clotet-Codina I, Este JA, Borrell JI, Teixido J. Recent advances in combinatorial
chemistry applied to development of anti-HIV drugs. Mini Rev Med Chem 2006;6:91108.
107. Lee KH. Current developments in the discovery and design of new drug candidates from plant
natural product leads. J Nat Prod 2004;67:273283.
108. De Clercq E. Current lead natural products for the chemotherapy of human immunodeciency
virus (HIV) infection. Med Res Rev 2000;20:323349.
109. Tziveleka LA, Vagias C, Roussis V. Natural products with anti-HIV activity from marine
organisms. Curr Top Med Chem 2003;3:15121535.
110. Matthee G, Wright AD, Konig GM. HIV reverse transcriptase inhibitors of natural origin. Planta
Med 1999;65:493506.
111. Nonaka G, Nishioka I, Nishizawa M, Yamagishi T, Kashiwada Y, Dutschman GE, Bodner AJ,
Kilkuskie RE, Cheng YC, Lee KH. Anti-AIDS agents, 2: Inhibitory effects of tannins on
HIV reverse transcriptase and HIV replication in H9 lymphocyte cells. J Nat Prod 1990;53:
587595.
112. (a) Buckheit RW, Jr, White EL, Fliakas-Boltz V, Russell J, Stup TL, Kinjerski TL, Osterling MC,
Weigand A, Bader JP. Unique anti-human immunodeciency virus activities of the nonnucleoside
reverse transcriptase inhibitors calanolide A, costatolide, and dihydrocostatolide. Antimicrob
Agents Chemother 1999;43:18271834. (b) Buckheit RW, Jr, Russell JD, Xu ZQ, Flavin M.
Anti-HIV-1 activity of calanolides used in combination with other mechanistically diverse
inhibitors of HIV-1 replication. Antivir Chem Chemother 2000;11:321327. (c) Creagh T,
Ruckle JL, Tolbert DT, Giltner J, Eiznhamer DA, Dutta B, Flavin MT, Xu ZQ. Safety and
pharmacokinetics of single doses of (1)-calanolide a, a novel, naturally occurring nonnucleoside
reverse transcriptase inhibitor, in healthy, human immunodeciency virus-negative human
subjects. Antimicrob Agents Chemother 2001;45:13791386. (d) Eiznhamer DA, Creagh T,
Ruckle JL, Tolbert DT, Giltner J, Dutta B, Flavin MT, Jenta T, Xu ZQ. Safety and
pharmacokinetic prole of multiple escalating doses of (1)-calanolide A, a naturally occurring
nonnucleoside reverse transcriptase inhibitor, in healthy HIV-negative volunteers. HIV Clin Trials
2002;3:435450.
113. Patil AD, Freyer AJ, Eggleston DS, Haltiwanger RC, Bean MF, Taylor PB, Caranfa MJ,
Breen AL, Bartus HR, Johnson RK, Hertzberg RP, Westley JW. The inophyllums, novel
inhibitors of HIV-1 reverse transcriptase isolated from the Malaysian tree, Calophyllum
inophyllum Linn. J Med Chem 1993;36:41314138.
114. Zembower DE, Liao S, Flavin MT, Xu ZQ, Stup TL, Buckheit RW, Jr, Khilevich A, Mar AA,
Sheinkman AK. Structural analogues of the calanolide anti-HIV agents. Modication of the
trans-10,11-dimethyldihydropyran-12-ol ring (ring C). J Med Chem 1997;40:10051017.

Medicinal Research Reviews DOI 10.1002/med


60
E60 KK ZHAN
ZHANETAL.
ETAL.

115. Xue H, Lu X, Zheng P, Liu L, Han C, Hu J, Liu Z, Ma T, Li Y, Wang L, Chen Z, Liu G. Highly
suppressing wild-type HIV-1 and Y181C mutant HIV-1 strains by 10-chloromethyl-11-demethyl-
12-oxo-calanolide A with druggable prole. J Med Chem 2010;53:13971401.
116. Lee TT, Kashiwada Y, Huang L, Snider J, Cosentino M, Lee KH. Suksdorn: An anti-HIV
principle from Lomatium suksdori, its structure-activity correlation with related coumarins, and
synergistic effects with anti-AIDS nucleosides. Bioorg Med Chem 1994;2:10511056.
117. Huang L, Kashiwada Y, Cosentino LM, Fan S, Chen CH, McPhail AT, Fujioka T, Mihashi K,
Lee KH. Anti-AIDS agents. 15. Synthesis and anti-HIV activity of dihydroseselins and related
analogs. J Med Chem 1994;37:39473955.
118. Huang L, Yuan X, Yu D, Lee KH, Chen CH. Mechanism of action and resistant prole of anti-
HIV-1 coumarin derivatives. Virology 2005;332:623628.
119. Xie L, Takeuchi Y, Cosentino LM, Lee KH. Anti-AIDS agents. 33. Synthesis and anti-HIV
activity of mono-methyl substituted 30 ,40 -di-O-()-camphanoyl-(1)-cis-khellactone (DCK) analogues.
Bioorg Med Chem Lett 1998;8:21512156.
120. Yang ZY, Xia Y, Xia P, Cosentino LM, Lee KH. Anti-AIDS agents. 31. Synthesis and anti-HIV
activity of 4-substituted 30 ,40 -di-O-()-camphanoyl-(1)-cis-khellactone (DCK) thiolactone
analogs. Bioorg Med Chem Lett 1998;8:14831486.
121. Xie L, Takeuchi Y, Cosentino LM, Lee KH. Anti-AIDS agents. 37. Synthesis and structure-
activity relationships of (30 R,40 R)-(1)-cis-khellactone derivatives as novel potent anti-HIV agents.
J Med Chem 1999;42:26622672.
122. Yang ZY, Xia Y, Xia P, Brossi A, Cosentino LM, Lee KH. Anti-AIDS agents part 41: Synthesis
and anti-HIV activity of 30 ,40 -di-o-()-camphanoyl-(1)-cis-khellactone (DCK) lactam analogues.
Bioorg Med Chem Lett 2000;10:10031005.
123. Xie L, Takeuchi Y, Cosentino LM, McPhail AT, Lee KH. Anti-AIDS agents. 42. Synthesis and
anti-HIV activity of disubstituted (30 R,40 R)-30 ,40 -di-O-(S)-camphanoyl-(1)-cis-khellactone analo-
gues. J Med Chem 2001;44:664671.
124. Xie L, Yu D, Wild C, Allaway G, Turpin J, Smith PC, Lee KH. Anti-AIDS agents. 52. Synthesis
and anti-HIV activity of hydroxymethyl (30 R,40 R)-30 ,40 -di-O-(S)-camphanoyl-(1)-cis-khellactone
derivatives. J Med Chem 2004;47:756760.
125. Xie L, Zhao CH, Zhou T, Chen HF, Fan BT, Chen XH, Ma JZ, Li JY, Bao ZY, Lo Z, Yu D,
Lee KH. Molecular modeling, design, synthesis, and biological evaluation of novel 30 ,40 -
dicamphanoyl-(1)-cis-khellactone (DCK) analogs as potent anti-HIV agents. Bioorg Med Chem
2005;13:64356449.
126. Chen Y, Zhang Q, Zhang B, Xia P, Xia Y, Yang ZY, Kilgore N, Wild C, Morris-Natschke SL,
Lee KH. Anti-AIDS agents. Part 56: Synthesis and anti-HIV activity of 7-thia-di-O-()-
camphanoyl-(1)-cis-khellactone (7-thia-DCK) analogs. Bioorg Med Chem 2004;12:63836387.
127. Zhang Q, Chen Y, Xia P, Xia Y, Yang ZY, Yu D, Morris-Natschke SL, Lee KH. Anti-AIDS
agents. Part 62: Anti-HIV activity of 20 -substituted 4-methyl-30 ,40 -di-O-()-camphanoyl-(1)-cis-
khellactone (4-methyl DCK) analogs. Bioorg Med Chem Lett 2004;14:58555857.
128. Yu D, Brossi A, Kilgore N, Wild C, Allaway G, Lee KH. Anti-HIV agents. Part 55: 30 R,40 R-
Di-(O)-()-camphanoyl-20 ,20 -dimethyldihydropyrano[2,3-f]chromone (DCP), a novel anti-HIV
agent. Bioorg Med Chem Lett 2003;13:15751576.
129. Yu D, Chen CH, Brossi A, Lee KH. Anti-AIDS agents. 60. Substituted 30 R,40 R-di-O-()-
camphanoyl-20 ,20 -dimethyldihydropyrano[2,3-f]chromone (DCP) analogues as potent anti-HIV
agents. J Med Chem 2004;47:40724082.
130. Xie L, Guo HF, Lu H, Zhuang XM, Zhang AM, Wu G, Ruan JX, Zhou T, Yu D, Qian K,
Lee KH, Jiang S. Development and preclinical studies of broad-spectrum anti-HIV agent
(30 R,40 R)-3-cyanomethyl-4-methyl-30 ,40 -di-O-(S)-camphanoyl-(1)-cis-khellactone (3-cyanomethyl-4-
methyl-DCK). J Med Chem 2008;51:76897696.
131. Xu SQ, Yan X, Chen Y, Xia P, Qian K, Yu D, Xia Y, Yang ZY, Morris-Natschke SL, Lee KH.
Anti-AIDS agents 84. Synthesis and anti-human immunodeciency virus (HIV) activity of

Medicinal Research Reviews DOI 10.1002/med


HIV-1NNRTIs
HIV-1 NNRTIs KK 61
E61

20 -monomethyl-4-methyl- and 10 -thia-4-methyl-(30 R,40 R)-30 ,40 -di- O-(S)-camphanoyl-(1)-cis-


khellactone (DCK) analogs. Bioorg Med Chem 2010. DOI: 10.1016/j.bmc.2010.08.031.
132. Chen HF, Fan BT, Zhao CY, Xie L, Zhao CH, Zhou T, Lee KH, Allaway G. Computational
studies and drug design for HIV-1 reverse transcriptase inhibitors of 30 ,40 -di-O-(S)-camphanoyl-
(1)-cis-khellactone (DCK) analogs. J Comput Aided Mol Des 2005;19:243258.
133. Fleming FF, Yao L, Ravikumar PC, Funk L, Shook BC. Nitrile-Containing Pharmaceuticals:
Efcacious Roles of the Nitrile Pharmacophore. J Med Chem 2010. DOI: 10.1021/jm100762r.
134. Schneider G. Virtual screening: An endless staircase? Nat Rev Drug Discov 2010;9:273276.
135. McInnes C. Virtual screening strategies in drug discovery. Curr Opin Chem Biol 2007;11:
494502.
136. Shoichet BK. Virtual screening of chemical libraries. Nature 2004;432:862865.
137. Kitchen DB, Decornez H, Furr JR, Bajorath J. Docking and scoring in virtual screening for drug
discovery: Methods and applications. Nat Rev Drug Discov 2004;3:935949.
138. Sangma C, Chuakheaw D, Jongkon N, Saenbandit K, Nunrium P, Uthayopas P, Hannongbua S.
Virtual screening for anti-HIV-1 RT and anti-HIV-1 PR inhibitors from the Thai medicinal plants
database: A combined docking with neural networks approach. Comb Chem High Throughput
Screen 2005;8:417429.
139. Bustanji Y, Al-Masri IM, Qasem A, Al-Bakri AG, Taha MO. In silico screening for
non-nucleoside HIV-1 reverse transcriptase inhibitors using physicochemical lters and
high-throughput docking followed by in vitro evaluation. Chem Biol Drug Des 2009;74:
258265.
140. Nichols SE, Domaoal RA, Thakur VV, Tirado-Rives J, Anderson KS, Jorgensen WL. Discovery
of wild-type and Y181C mutant non-nucleoside HIV-1 reverse transcriptase inhibitors using
virtual screening with multiple protein structures. J Chem Inf Model 2009;49:12721279.
141. Herschhorn A, Hizi A. Virtual screening, identication, and biochemical characterization of novel
inhibitors of the reverse transcriptase of human immunodeciency virus type-1. J Med Chem 2008;
51:57025713.
142. Herschhorn A, Oz-Gleenberg I, Hizi A. Mechanism of inhibition of HIV-1 reverse transcriptase
by the novel broad-range DNA polymerase inhibitor N-{2-[4-(aminosulfonyl)phenyl]ethyl}-2-(2-
thienyl)acetamide. Biochemistry 2008;47:490502.
143. (a) Kurogi Y, Guner OF. Pharmacophore modeling and three-dimensional database searching for
drug design using catalyst. Curr Med Chem 2001;8:10351055. (b) Guner O, Clement O,
Kurogi Y. Pharmacophore modeling and three dimensional database searching for drug design
using catalyst: Recent advances. Curr Med Chem 2004;11:29913005.
144. (a) van Drie JH. Pharmacophore discoveryLessons learned. Curr Pharm Des 2003;9:16491664.
(b) Langer T, Krovat EM. Chemical feature-based pharmacophores and virtual library screening
for discovery of new leads. Curr Opin Drug Discov Devel 2003;6:370376.
145. Gussio R, Pattabiraman N, Kellogg GE, Zaharevitz DW. Use of 3D QSAR methodology for data
mining the National Cancer Institute Repository of Small Molecules: Application to HIV-1
reverse transcriptase inhibition. Methods 1998;14:255263.
146. Leitao A, Andricopulo AD, Montanari CA. In silico screening of HIV-1 non-nucleoside reverse
transcriptase and protease inhibitors. Eur J Med Chem 2008;43:14121422.
147. Schneider G, Fechner U. Computer-based de novo design of drug-like molecules. Nat Rev Drug
Discov 2005;4:649663.
148. Bauman JD, Das K, Ho WC, Baweja M, Himmel DM, Clark AD, Jr, Oren DA, Boyer PL,
Hughes SH, Shatkin AJ, Arnold E. Crystal engineering of HIV-1 reverse transcriptase for
structure-based drug design. Nucleic Acids Res 2008;36:50835092.
149. Herschhorn A, Lerman L, Weitman M, Gleenberg IO, Nudelman A, Hizi A. De novo
parallel design, synthesis and evaluation of inhibitors against the reverse transcriptase of
human immunodeciency virus type-1 and drug-resistant variants. J Med Chem 2007;50:
23702384.

Medicinal Research Reviews DOI 10.1002/med


62
E62 KK ZHAN
ZHANETAL.
ETAL.

150. Vinkers HM, de Jonge MR, Daeyaert FF, Heeres J, Koymans LM, van Lenthe JH, Lewi PJ,
Timmerman H, Van Aken K, Janssen PA. SYNOPSIS: SYNthesize and OPtimize System in
Silico. J Med Chem 2003;46:27652773.
151. Lansdon EB, Brendza KM, Hung M, Wang R, Mukund S, Jin D, Birkus G, Kutty N, Liu X.
Crystal structures of HIV-1 reverse transcriptase with Etravirine (TMC125) and Rilpivirine
(TMC278): Implications for drug design. J Med Chem 2010;53:42954299.
152. Grifth R, Luu TT, Garner J, Keller PA. Combining structure-based drug design and
pharmacophores. J Mol Graph Model 2005;23:439446.
153. DCruz OJ, Uckun FM. Novel tight binding PETT, HEPT and DABO-based non-nucleoside
inhibitors of HIV-1 reverse transcriptase. J Enzyme Inhib Med Chem 2006;21:329350.
154. DCruz OJ, Uckun FM. Novel broad-spectrum thiourea non-nucleoside inhibitors for the
prevention of mucosal HIV transmission. Curr HIV Res 2006;4:329345.
155. Vig R, Mao C, Venkatachalam TK, Tuel-Ahlgren L, Sudbeck EA, Uckun FM. Rational design
and synthesis of phenethyl-5-bromopyridyl thiourea derivatives as potent non-nucleoside
inhibitors of HIV reserve transcriptase. Bioorg Med Chem 1998;6:17891797.
156. Mao C, Vig R, Venkatachalam TK, Sudbeck EA, Uckun FM. Structure-based design of N-[2-(1-
piperidinylethyl)]-N0 -[2-(5-bromopyridyl)]-thiourea and N-[2-(1-piperazinylethyl)]-N0 -[2-(5-bro-
mopyridyl)]-thiourea as potent non-nucleoside inhibitors of HIV-1 reverse transcriptase. Bioorg
Med Chem Lett 1998;8:22132218.
157. Vig R, Mao C, Venkatachalam TK, Tuel-Ahlgren L, Sudbeck EA, Uckun FM. Rational design
and synthesis of phenethyl-5-bromopyridyl thiourea derivatives as potent non-nucleoside
inhibitors of HIV reserve transcriptase. Bioorg Med Chem 1998;6:17891797.
158. Mao C, Sudbeck EA, Venkatachalam TK, Uckun FM. Structure-based drug design of non-
nucleoside inhibitors for wild-type and drug-resistant HIV reverse transcriptase. Biochem
Pharmacol 2000;60:12511265.
159. Vig R, Mao C. Nonnucleoside inhibitors of reverse transcriptase, composite binding pocket
and methods for use thereof. WO9947501A, 1999; US6380190B, 2002; US2005153995A, 2005.
160. Hopkins AL, Ren J, Esnouf RM, Willcox BE, Jones EY, Ross C, Miyasaka T, Walker RT,
Tanaka H, Stammers DK, Stuart DI. Complexes of HIV-1 reverse transcriptase with inhibitors of
the HEPT series reveal conformational changes relevant to the design of potent non-nucleoside
inhibitors. J Med Chem 1996;39:15891600.
161. Shen L, Shen J, Luo X, Cheng F, Xu Y, Chen K, Arnold E, Ding J, Jiang H. Steered
molecular dynamics simulation on the binding of NNRTI to HIV-1 RT. Biophys J 2003;84:
35473563.
162. Heeres J, Lewi PJ. The medicinal chemistry of the DATA and DAPY series of HIV-1
non-nucleoside reverse transcriptase inhibitors (NNRTIS). Adv Antiviral Drug Des 2007;5:
213242.
163. Girardet JL, Koh Y H, Shaw S, Kim HW. Diaryl-purine, azapurines and -deazapurines as non-
nucleoside reverse transcriptase inhibitors for treatment of HIV. Patent. WO2006122003, 2006.
164. Hopkins AL, Ren J, Tanaka H, Baba M, Okamato M, Stuart DI, Stammers DK. Design of
MKC-442 (emivirine) analogues with improved activity against drug-resistant HIV mutants.
J Med Chem 1999;42:45004505.
165. (a) Morphy R, Rankovic Z. Designed multiple ligands. An emerging drug discovery paradigm.
J Med Chem 2005;48:65236543. (b) Morphy R, Kay C, Rankovic Z. From magic bullets to
designed multiple ligands. Drug Discov Today 2004;9:641651. (c) Morphy R, Rankovic Z. The
physicochemical challenges of designing multiple ligands. J Med Chem 2006;49:49614970.
(d) Morphy R, Rankovic Z. Fragments, network biology and designing multiple ligands. Drug
Discov Today 2007;12:156160. (e) Morphy R, Rankovic Z. Designing multiple ligands
Medicinal chemistry strategies and challenges. Curr Pharm Des 2009;15:587600.
166. Zhan P, Liu X. Designed multiple ligands: An emerging anti-HIV drug discovery paradigm. Curr
Pharm Des 2009;15:18931917.

Medicinal Research Reviews DOI 10.1002/med


HIV-1NNRTIs
HIV-1 NNRTIs KK 63
E63

167. Das K, Bauman JD, Clark AD, Jr, Frenkel YV, Lewi PJ, Shatkin AJ, Hughes SH,
Arnold E. High-resolution structures of HIV-1 reverse transcriptase/TMC278 complexes:
Strategic exibility explains potency against resistance mutations. Proc Natl Acad Sci USA
2008;105:14661471.
168. Das K, Saraanos SG, Clark AD, Jr, Boyer PL, Hughes SH, Arnold E. Crystal structures of
clinically relevant Lys103Asn/Tyr181Cys double mutant HIV-1 reverse transcriptase in complexes
with ATP and non-nucleoside inhibitor HBY 097. J Mol Biol 2007;365:7789.
169. Hsiou Y, Das K, Ding J, Clark AD, Jr, Kleim JP, Rosner M, Winkler I, Riess G, Hughes SH,
Arnold E. Structures of Tyr188Leu mutant and wild-type HIV-1 reverse transcriptase complexed
with the non-nucleoside inhibitor HBY 097: Inhibitor exibility is a useful design feature for
reducing drug resistance. J Mol Biol 1998;284:313323.
170. Ren J, Nichols C, Bird LE, Fujiwara T, Sugimoto H, Stuart DI, Stammers DK. Binding of the
second generation non-nucleoside inhibitor S-1153 to HIV-1 reverse transcriptase involves
extensive main chain hydrogen bonding. J Biol Chem 2000;275:1431614320.
171. Himmel DM, Das K, Clark AD, Jr, Hughes SH, Benjahad A, Oumouch S, Guillemont J,
Coupa S, Poncelet A, Csoka I, Meyer C, Andries K, Nguyen CH, Grierson DS, Arnold E. Crystal
structures for HIV-1 reverse transcriptase in complexes with three pyridinone derivatives: A new
class of non-nucleoside inhibitors effective against a broad range of drug-resistant strains. J Med
Chem 2005;48:75827591.
172. Pelemans H, Esnouf R, De Clercq E, Balzarini J. Mutational analysis of trp-229 of human
immunodeciency virus type 1 reverse transcriptase (RT) identies this amino acid residue as a
prime target for the rational design of new non-nucleoside RT inhibitors. Mol Pharmacol 2000;57:
954960.
173. Ren J, Stammers DK. HIV reverse transcriptase structures: Designing new inhibitors and
understanding mechanisms of drug resistance. Trends Pharmacol Sci 2005;26:47.
174. Pata JD, Stirtan WG, Goldstein SW, Steitz TA. Structure of HIV-1 reverse transcriptase bound to
an inhibitor active against mutant reverse transcriptases resistant to other nonnucleoside
inhibitors. Proc Natl Acad Sci USA 2004;101:1054810553.
175. Himmel DM, Saraanos SG, Dharmasena S, Hossain MM, McCoy-Simandle K, Ilina T,
Clark AD, Jr, Knight JL, Julias JG, Clark PK, Krogh-Jespersen K, Levy RM, Hughes SH,
Parniak MA, Arnold E. HIV-1 reverse transcriptase structure with RNase H inhibitor dihydroxy
benzoyl naphthyl hydrazone bound at a novel site. ACS Chem Biol 2006;1:702712.
176. (a) Patani GA, LaVoie EJ. Bioisosterism: A rational approach in drug design. Chem Rev 1996;96:
31473176. (b) Olesen PH. The use of bioisosteric groups in lead optimization. Curr Opin Drug
Discov Dev 2001;4:471478. (c) Lima LM, Barreiro EJ. Bioisosterism: A useful strategy for
molecular modication and drug design. Curr Med Chem 2005;12:2349.
177. Andries K, Azijn H, Thielemans T, Ludovici D, Kukla M, Heeres J, Janssen P, De Corte B,
Vingerhoets J, Pauwels R, de Bethune MP. TMC125, a novel next-generation nonnucleoside
reverse transcriptase inhibitor active against nonnucleoside reverse transcriptase inhibitor-
resistant human immunodeciency virus type 1. Antimicrob Agents Chemother 2004;48:
46804686.
178. Mordant C, Schmitt B, Pasquier E, Demestre C, Queguiner L, Masungi C, Peeters A,
Smeulders L, Bettens E, Hertogs K, Heeres J, Lewi P, Guillemont J. Synthesis of novel
diarylpyrimidine analogues of TMC278 and their antiviral activity against HIV-1 wild-type and
mutant strains. Eur J Med Chem 2007;42:567579.
179. Guillemont J, Pasquier E, Palandjian P, Vernier D, Gaurrand S, Lewi PJ, Heeres J, de Jonge MR,
Koymans LM, Daeyaert FF, Vinkers MH, Arnold E, Das K, Pauwels R, Andries K, de
Bethune MP, Bettens E, Hertogs K, Wigerinck P, Timmerman P, Janssen PA. Synthesis of novel
diarylpyrimidine analogues and their antiviral activity against human immunodeciency virus
type 1. J Med Chem 2005;48:20722079.
180. Thakur A, Thakur M, Bharadwaj A, Thakur S. SAR and QSAR studies: Modelling of new DAPY
derivatives. Eur J Med Chem 2008;43:471477.

Medicinal Research Reviews DOI 10.1002/med


64
E64 KK ZHAN
ZHANETAL.
ETAL.

181. Feng XQ, Liang YH, Zeng ZS, Chen FE, Balzarini J, Pannecouque C, De Clercq E. Structural
modications of DAPY analogues with potent anti-HIV-1 activity. Chem Med Chem 2009;4:
219224.
182. Liang YH, Feng XQ, Zeng ZS, Chen FE, Balzarini J, Pannecouque C, De Clercq E. Design,
synthesis, and SAR of naphthyl-substituted Diarylpyrimidines as non-nucleoside inhibitors of
HIV-1 reverse transcriptase. ChemMedChem 2009;4:15371545.
183. Corbett JW, Ko SS, Rodgers JD, Gearhart LA, Magnus NA, Bacheler LT, Diamond S, Jeffrey S,
Klabe RM, Cordova BC, Garber S, Logue K, Trainor GL, Anderson PS, Erickson-Viitanen SK.
Inhibition of clinically relevant mutant variants of HIV-1 by quinazolinone non-nucleoside reverse
transcriptase inhibitors. J Med Chem 2000;43:20192030.
184. Corbett JW, Ko SS, Rodgers JD, Jeffrey S, Bacheler LT, Klabe RM, Diamond S, Lai CM,
Rabel SR, Saye JA, Adams SP, Trainor GL, Anderson PS, Erickson-Viitanen SK. Expanded-
spectrum nonnucleoside reverse transcriptase inhibitors inhibit clinically relevant mutant variants
of human immunodeciency virus type 1. Antimicrob Agents Chemother 1999;43:28932897.
185. Corbett JW, Gearhart LA, Ko SS, Rodgers JD, Cordova BC, Klabe RM, Erickson-Viitanen SK.
Novel 2,2-dioxide-4,4-disubstituted-1,3-H-2,1,3-benzothiadiazines as non-nucleoside reverse
transcriptase inhibitors. Bioorg Med Chem Lett 2000;10:193195.
186. Patel M, Ko SS, McHugh RJ, Jr, Markwalder JA, Srivastava AS, Cordova BC, Klabe RM,
Erickson-Viitanen S, Trainor GL, Seitz SP. Synthesis and evaluation of analogs of Efavirenz
(SUSTIVA) as HIV-1 reverse transcriptase inhibitors. Bioorg Med Chem Lett 1999;9:28052810.
187. Patel M, McHugh RJ, Jr, Cordova BC, Klabe RM, Bacheler LT, Erickson-Viitanen S,
Rodgers JD. Synthesis and evaluation of novel quinolinones as HIV-1 reverse transcriptase
inhibitors. Bioorg Med Chem Lett 2001;11:19431945.
188. Cocuzza AJ, Chidester DR, Cordova BC, Klabe RM, Jeffrey S, Diamond S, Weigelt CA, Ko SS,
Bacheler LT, Erickson-Viitanen SK, Rodgers JD. 4,1-Benzoxazepinone analogues of efavirenz
(Sustiva) as HIV-1 reverse transcriptase inhibitors. Bioorg Med Chem Lett 2001;11:13891392.
189. Corbett JW, Gearhart LA, Ko SS, Rodgers JD, Cordova BC, Klabe RM, Erickson-Viitanen SK.
Novel 2,2-dioxide-4,4-disubstituted-1,3-H-2,1,3-benzothiadiazines as non-nucleoside reverse transcriptase
inhibitors. Bioorg Med Chem Lett 2000;10:193195.
190. (a) Di Santo R, Costi R, Artico M, Miele G, Lavecchia A, Novellino E, Bergamini A, Cancio R,
Maga G. Arylthiopyrrole (AThP) derivatives as non-nucleoside HIV-1 reverse transcriptase
inhibitors: Synthesis, structure-activity relationships, and docking studies (part 1). ChemMed-
Chem 2006;1:13671378. (b) Lavecchia A, Costi R, Artico M, Miele G, Novellino E, Bergamini A,
Crespan E, Maga G, Di Santo R. Arylthiopyrrole (AThP) derivatives as non-nucleoside HIV-1
reverse transcriptase inhibitors: Synthesis, structure-activity relationships, and docking studies
(part 2). Chem Med Chem 2006;1:13791390.
191. Jones LH, Dupont T, Mowbray CE, Newman SD. A concise and selective synthesis of novel
5-aryloxyimidazole NNRTIs. Org Lett 2006;8:17251727.
192. Jones LH, Allan G, Corbau R, Hay D, Middleton DS, Mowbray CE, Newman SD, Perros M,
Randall A, Vuong H, Webster R, Westby M, Williams D. Optimization of 5-aryloxyimidazole
non-nucleoside reverse transcriptase inhibitors. Chem Med Chem 2008;3:17561762.
193. (a) Mowbray CE, Burt C, Corbau R, Perros M, Tran I, Stupple PA, Webster R, Wood A.
Pyrazole NNRTIs 1: Design and initial optimisation of a novel template. Bioorg Med Chem Lett
2009;19:55995602. (b) Mowbray CE, Corbau R, Hawes M, Jones LH, Mills JE, Perros M,
Selby MD, Stupple PA, Webster R, Wood A. Pyrazole NNRTIs 3: Optimisation of
physicochemical properties. Bioorg Med Chem Lett 2009;19:56035606. (c) Mowbray CE,
Burt C, Corbau R, Gayton S, Hawes M, Perros M, Tran I, Price DA, Quinton FJ, Selby MD,
Stupple PA, Webster R, Wood A. Pyrazole NNRTIs 4: Selection of UK-453,061 (lersivirine) as a
development candidate. Bioorg Med Chem Lett 2009;19:58575860. (d) Fatkenheuer G,
Staszewski S, Plettenburg A, Hackman F, Layton G, McFadyen L, Davis J, Jenkins TM.
Activity, pharmacokinetics and safety of lersivirine (UK-453,061), a next-generation nonnucleo-
side reverse transcriptase inhibitor, during 7-day monotherapy in HIV-1-infected patients. AIDS

Medicinal Research Reviews DOI 10.1002/med


HIV-1
HIV-1NNRTIs
NNRTIs KK 65
E65

2009;23:21152122. (e) Corbau R, Mori J, Phillips C, Fishburn L, Martin A, Mowbray C,


Panton W, Smith-Burchnell C, Thornberry A, Ringrose H, Knochel T, Irving S, Westby M,
Wood A, Perros M. Lersivirine: A Non-Nucleoside Reverse Transcriptase Inhibitor with Activity
against Drug-Resistant Human Immunodeciency Virus-1. Antimicrob Agents Chemother 2010
July 26 [Epub ahead of print].
194. (a) Zhan P, Liu X, Cao Y, Wang Y, Pannecouque C, De Clercq E. 1,2,3-Thiadiazole
thioacetanilides as a novel class of potent HIV-1 non-nucleoside reverse transcriptase inhibitors.
Bioorg Med Chem Lett 2008;18:53685371. (b) Zhan P, Liu X, Li Z, Fang Z, Li Z, Wang D,
Pannecouque C, Clercq ED. Novel 1,2,3-thiadiazole derivatives as HIV-1 NNRTIs with improved
potency: Synthesis and preliminary SAR studies. Bioorg Med Chem 2009;17:59205927.
(c) Zhan P, Liu X, Fang Z, Li Z, Pannecouque C, De Clercq E. Synthesis and anti-HIV activity
evaluation of 2-(4-(naphthalen-2-yl)-1,2,3-thiadiazol-5-ylthio)-N-acetamides as novel non-nucleo-
side HIV-1 reverse transcriptase inhibitors. Eur J Med Chem 2009;44:46484653. (d) Zhan P,
Liu X, Li Z, Fang Z, Pannecouque C, De Clercq E. Synthesis and biological evaluation of a new
series of 2-(4-(3,4-dichlorophenyl)-1,2,3-thiadiazol-5-ylthio)acetamides as HIV-1 inhibitors. Chem
Biodivers 2010;7:17171727. (e) Zhan P, Liu X, Zhu J, Fang Z, Li Z, Pannecouque C, Clercq ED.
Synthesis and biological evaluation of imidazole thioacetanilides as novel non-nucleoside HIV-1
reverse transcriptase inhibitors. Bioorg Med Chem 2009;17:57755781. (f) Zhan P, Liu X, Fang Z,
Pannecouque C, De Clercq E. 1,2,3-Selenadiazole thioacetanilides: Synthesis and anti-HIV
activity evaluation. Bioorg Med Chem 2009;17:63746379.
195. (a) Girardet JL, Zhang Z, Hamatake R, de la Rosa Hernandez MA, Gunic E, Hong Z, Kim H,
Koh YH, Nilar S, Shaw S, Yao N. Non-nucleoside reverse transcriptase inhibitors. Patent WO
2004030611, 2004. (b) Simoneau B, Thavonekham B, Landry S, OMeara J, Yoakim C,
Faucher AM. Non-nucleoside reverse transcriptase inhibitors. Patent WO 2004050643, 2004.
(c) Shaw-Reid CA, Miller MD, Hazuda DJ, Ferrer M, Sur SM, Summa V, Lyle TA, Kinzel O,
Pescatore G, Muraglia E, Orvieto F, Williams PD. HIV reverse transcriptase inhibitors. Patent
WO 2005115147, 2005. (d) Girardet JL, Koh YH, Hernandez M, De la Rosa, Gunic E, Kim HW,
Hong W. S-Triazolyl a-mercaptoacetanildes as inhibitors of HIV reverse transcriptase. Patent WO
2006026356, 2006. (e) Simoneau B, Thavonekham B, Landry S, OMeara J, Yoakim C,
Faucher AM. Non nucleoside reverse transcriptase inhibitors. Patent WO 2005118575, 2005.
(f) Koch U, Kinzel O, Muraglia E, Summa V. HIV reverse transcriptase inhibitors. Patent WO
2006037468, 2006. (g) Gagnon A, Landry S, Coulombe R, Jakalian A, Guse I, Thavonekham B,
Bonneau PR, Yoakim C, Simoneau B. Investigation on the role of the tetrazole in the binding of
thiotetrazolylacetanilides with HIV-1 wild type and K103N/Y181C double mutant reverse
transcriptases. Bioorg Med Chem Lett 2009;19:11991205.
196. Tucker TJ, Sisko JT, Tynebor RM, Williams TM, Felock PJ, Flynn JA, Lai MT, Liang Y,
McGaughey G, Liu M, Miller M, Moyer G, Munshi V, Perlow-Poehnelt R, Prasad S, Reid JC,
Sanchez R, Torrent M, Vacca JP, Wan BL, Yan Y. Discovery of 3-{5-[(6-amino-1H-pyrazolo[3,4-
b]pyridine-3-yl)methoxy]-2-chlorophenoxy}-5-chlorobenzonitrile (MK-4965): A potent, orally
bioavailable HIV-1 non-nucleoside reverse transcriptase inhibitor with improved potency against
key mutant viruses. J Med Chem 2008;51:65036511.
197. (a) Sweeney ZK, Harris SF, Arora SF, Javanbakht H, Li Y, Fretland J, Davidson JP,
Billedeau JR, Gleason SK, Hirschfeld D, Kennedy-Smith JJ, Mirzadegan T, Roetz R, Smith M,
Sperry S, Suh JM, Wu J, Tsing S, Villasenor AG, Paul A, Su G, Heilek G, Hang JQ, Zhou AS,
Jernelius JA, Zhang FJ, Klumpp K. Design of annulated pyrazoles as inhibitors of HIV-1 reverse
transcriptase. J Med Chem 2008;51:74497458. (b) Sweeney ZK, Dunn JP, Li Y, Heilek G,
Dunten P, Elworthy TR, Han X, Harris SF, Hirschfeld DR, Hogg JH, Huber W, Kaiser AC,
Kertesz DJ, Kim W, Mirzadegan T, Roepel MG, Saito YD, Silva TM, Swallow S, Tracy JL,
Villasenor A, Vora H, Zhou AS, Klumpp K. Discovery and optimization of pyridazinone
non-nucleoside inhibitors of HIV-1 reverse transcriptase. Bioorg Med Chem Lett 2008;18:
43524354. (c) Sweeney ZK, Acharya S, Briggs A, Dunn JP, Elworthy TR, Fretland J,
Giannetti AM, Heilek G, Li Y, Kaiser AC, Martin M, Saito YD, Smith M, Suh JM, Swallow S,
Wu J, Hang JQ, Zhou AS, Klumpp K. Discovery of triazolinone non-nucleoside inhibitors of HIV

Medicinal Research Reviews DOI 10.1002/med


66
E66 KK ZHAN
ZHANETAL.
ETAL.

reverse transcriptase. Bioorg Med Chem Lett 2008;18:43484351. (d) Sweeney ZK,
Kennedy-Smith JJ, Wu J, Arora N, Billedeau JR, Davidson JP, Fretland J, Hang JQ,
Heilek GM, Harris SF, Hirschfeld D, Inbar P, Javanbakht H, Jernelius JA, Jin Q, Li Y,
Liang W, Roetz R, Sarma K, Smith M, Stefanidis D, Su G, Suh JM, Villasenor AG, Welch M,
Zhang FJ, Klumpp K. Diphenyl ether non-nucleoside reverse transcriptase inhibitors with
excellent potency against resistant mutant viruses and promising pharmacokinetic properties.
Chem Med Chem 2009;4:8899.
198. Su DS, Lim JJ, Tinney E, Tucker TJ, Saggar S, Sisko JT, Wan BL, Young MB, Anderson KD,
Rudd D, Munshi V, Bahnck C, Felock PJ, Lu M, Lai MT, Touch S, Moyer G, Distefano DJ,
Flynn JA, Liang Y, Sanchez R, Perlow-Poehnelt R, Miller M, Vacca JP, Williams TM,
Anthony NJ. Biaryl ethers as potent allosteric inhibitors of reverse transcriptase and its key
mutant viruses: Aryl substituted pyrazole as a surrogate for the pyrazolopyridine motif. Bioorg
Med Chem Lett 2010;20:43284332.
199. Aquino CJ, Dickson H. Chemical compounds. WO2008157330.
200. Silvestri R, Artico M, De Martino G, Ragno R, Massa S, Loddo R, Murgioni C, Loi AG, La
Colla P, Pani A. Synthesis, biological evaluation, and binding mode of novel 1-[2-(diarylmethoxy)-
ethyl]-2-methyl-5-nitroimidazoles targeted at the HIV-1 reverse transcriptase. J Med Chem 2002;
45:15671576.
201. De Martino G, La Regina G, Di Pasquali A, Ragno R, Bergamini A, Ciaprini C, Sinistro A,
Maga G, Crespan E, Artico M, Silvestri R. Novel 1-[2-(diarylmethoxy)ethyl]-2-methyl-5-
nitroimidazoles as HIV-1 non-nucleoside reverse transcriptase inhibitors. A structure-activity
relationship investigation. J Med Chem 2005;48:43784388.
202. Cushman M, Casimiro-Garcia A, Hejchman E, Ruell JA, Huang M, Schaeffer CA, Williamson K,
Rice WG, Buckheit RW, Jr. New alkenyldiarylmethanes with enhanced potencies as anti-HIV
agents which act as non-nucleoside reverse transcriptase inhibitors. J Med Chem 1998;41:20762089.
203. Cushman M, Casimiro-Garcia A, Williamson K, Rice WG. Synthesis of a non-nucleoside reverse
transcriptase inhibitor in the alkenyldiarylmethane (ADAM) series with optimized potency and
therapeutic index. Bioorg Med Chem Lett 1998;8:195198.
204. Silvestri MA, Nagarajan M, De Clercq E, Pannecouque C, Cushman M. Design, synthesis, anti-
HIV activities, and metabolic stabilities of alkenyldiarylmethane (ADAM) non-nucleoside reverse
transcriptase inhibitors. J Med Chem 2004;47:31493162.
205. Deng BL, Hartman TL, Buckheit RW, Jr, Pannecouque C, De Clercq E, Fanwick PE,
Cushman M. Synthesis, anti-HIV activity, and metabolic stability of new alkenyldiarylmethane
HIV-1 non-nucleoside reverse transcriptase inhibitors. J Med Chem 2005;48:61406155.
206. Deng BL, Hartman TL, Buckheit RW, Jr, Pannecouque C, De Clercq E, Cushman M.
Replacement of the metabolically labile methyl esters in the alkenyldiarylmethane series of non-
nucleoside reverse transcriptase inhibitors with isoxazolone, isoxazole, oxazolone, or cyano
substituents. J Med Chem 2006;49:53165323.
207. Sakamoto T, Cullen MD, Hartman TL, Watson KM, Buckheit RW, Pannecouque C, De
Clercq E, Cushman M. Synthesis and anti-HIV activity of new metabolically stable
alkenyldiarylmethane non-nucleoside reverse transcriptase inhibitors incorporating N-methoxy
imidoyl halide and 1,2,4-oxadiazole systems. J Med Chem 2007;50:33143321.
208. Cullen MD, Deng BL, Hartman TL, Watson KM, Buckheit RW, Jr, Pannecouque C, Clercq ED,
Cushman M. Synthesis and biological evaluation of alkenyldiarylmethane HIV-1 non-nucleoside
reverse transcriptase inhibitors that possess increased hydrolytic stability. J Med Chem 2007;50:
48544867.
209. Deng BL, Zhao Y, Hartman TL, Watson K, Buckheit RW, Jr, Pannecouque C, De Clercq E,
Cushman M. Synthesis of alkenyldiarylmethanes (ADAMs) containing benzo[d]isoxazole and
oxazolidin-2-one rings, a new series of potent non-nucleoside HIV-1 reverse transcriptase
inhibitors. Eur J Med Chem 2009;44:12101214.
210. Deng BL, Cullen MD, Zhou Z, Hartman TL, Buckheit RW, Jr, Pannecouque C, De Clercq E,
Fanwick PE, Cushman M. Synthesis and anti-HIV activity of new alkenyldiarylmethane (ADAM)

Medicinal Research Reviews DOI 10.1002/med


HIV-1NNRTIs
HIV-1 NNRTIs KK 67
E67

non-nucleoside reverse transcriptase inhibitors (NNRTIs) incorporating benzoxazolone and


benzisoxazole rings. Bioorg Med Chem 2006;14:23662374.
211. Viegas-Junior C, Danuello A, da Silva Bolzani V, Barreiro EJ, Fraga CA. Molecular
hybridization: A useful tool in the design of new drug prototypes. Curr Med Chem 2007;14:
18291852.
212. Terasaka T, Kinoshita T, Kuno M, Nakanishi I. A highly potent non-nucleoside adenosine
deaminase inhibitor: Efcient drug discovery by intentional lead hybridization. J Am Chem Soc
2004;126:3435.
213. Corbett JW, Kresge KJ, Pan S, Cordova BC, Klabe RM, Rodgers JD, Erickson-Viitanen SK.
Triuoromethyl-containing 3-alkoxymethyl- and 3-aryloxymethyl-2-pyridinones are potent
inhibitors of HIV-1 non-nucleoside reverse transcriptase. Bioorg Med Chem Lett 2001;11:
309312.
214. Patel M, McHugh RJ, Jr, Cordova BC, Klabe RM, Erickson-Viitanen S, Trainor GL,
Rodgers JD. Synthesis and evaluation of quinoxalinones as HIV-1 reverse transcriptase inhibitors.
Bioorg Med Chem Lett 2000;10:17291731.
215. (a) Dolle V, Fan E, Nguyen CH, Aubertin AM, Kirn A, Andreola ML, Jamieson G, Tarrago-
Litvak L, Bisagni E. A new series of pyridinone derivatives as potent non-nucleoside human
immunodeciency virus type 1 specic reverse transcriptase inhibitors. J Med Chem 1995;38:
46794686. (b) Dolle V, Aubertin AM, Ludwig O, Chi HN, Bisagni E, Legraverend M. Synthesis
of new non-nucleoside inhibitors of HIV-1. Bioorg Med Chem Lett 1996;6:173178. (c) Dolle V,
Nguyen CH, Legraverend M, Aubertin AM, Kirn A, Andreola ML, Ventura M,
Tarrago-Litvak L, Bisagni E. Synthesis and antiviral activity of 4-benzyl pyridinone derivatives
as potent and selective non-nucleoside human immunodeciency virus type 1 reverse transcriptase
inhibitors. J Med Chem 2000;43:39493962. (d) Benjahad A, Guillemont J, Andries K,
Nguyen CH, Grierson DS. 3-iodo-4-phenoxypyridinones (IOPYs), a new family of highly potent
non-nucleoside inhibitors of HIV-1 reverse transcriptase. Bioorg Med Chem Lett 2003;13:
43094312.
216. (a) Benjahad A, Courte K, Guillemont J, Mabire D, Coupa S, Poncelet A, Csoka I, Andries K,
Pauwels R, de Bethune MP, Monneret C, Bisagni E, Nguyen CH, Grierson DS. 4-Benzyl- and
4-benzoyl-3-dimethylaminopyridin-2(1H)-ones, a new family of potent anti-HIV agents: Optimiza-
tion and in vitro evaluation against clinically important HIV mutant strains. J Med Chem 2004;47:
55015514. (b) Benjahad A, Croisy M, Monneret C, Bisagni E, Mabire D, Coupa S, Poncelet A,
Csoka I, Guillemont J, Meyer C, Andries K, Pauwels R, de Bethune MP, Himmel DM, Das K,
Arnold E, Nguyen CH, Grierson DS. 4-Benzyl and 4-benzoyl-3-dimethylaminopyridin-2(1H)-ones:
In vitro evaluation of new C-3-amino-substituted and C-5,6-alkyl-substituted analogues against
clinically important HIV mutant strains. J Med Chem 2005;48:19481964.
217. (a) Storer R, Alexandre FR, Dousson C, Moussa AM, Bridges E. Enantiomerically pure
phosphoindoles as HIV inhibitors. PCT Int. Appl. WO2008042240, 2008. (b) Zhou XJ,
Pietropaolo K, Damphousse D, Belanger B, Chen J, Sullivan-Bolyai J, Mayers D. Single-dose
escalation and multiple-dose safety, tolerability, and pharmacokinetics of IDX899, a candidate
human immunodeciency virus type 1 nonnucleoside reverse transcriptase inhibitor, in healthy
subjects. Antimicrob Agents Chemother 2009;53:17391746. (c) Klibanov OM, Kaczor RL. IDX-
899, an aryl phosphinate-indole non-nucleoside reverse transcriptase inhibitor for the potential
treatment of HIV infection. Curr Opin Investig Drugs 2010;11:237245.
218. (a) Brown N, Jacoby E. On scaffolds and hopping in medicinal chemistry. Mini Rev Med Chem
2006;6:12171229. (b) Tsunoyama K, Amini A, Sternberg MJ, Muggleton SH. Scaffold hopping
in drug discovery using inductive logic programming. J Chem Inf Model 2008;48:949957.
(c) Zhao H. Scaffold selection and scaffold hopping in lead generation: A medicinal chemistry
perspective. Drug Discov Today 2007;12:149155. (d) Mauser H, Guba W. Recent developments
in de novo design and scaffold hopping. Curr Opin Drug Discov Devel 2008;11:365374.
(e) Renner S, Schneider G. Scaffold-hopping potential of ligand-based similarity concepts. Chem
Med Chem 2006;1:181185.

Medicinal Research Reviews DOI 10.1002/med


68
E68 KK ZHAN
ZHANETAL.
ETAL.

219. Mai A, Sbardella G, Artico M, Ragno R, Massa S, Novellino E, Greco G, Lavecchia A, Musiu C,
La Colla M, Murgioni C, La Colla P, Loddo R. Structure-based design, synthesis, and biological
evaluation of conformationally restricted novel 2-alkylthio-6-[1-(2,6-diuorophenyl)alkyl]-3,4-
dihydro-5-alkylpyrimidin-4(3H)-ones as non-nucleoside inhibitors of HIV-1 reverse transcriptase.
J Med Chem 2001;44:25442554.
220. Su DS, Lim JJ, Tinney E, Wan BL, Young MB, Anderson KD, Rudd D, Munshi V, Bahnck C,
Felock PJ, Lu M, Lai MT, Touch S, Moyer G, Distefano DJ, Flynn JA, Liang Y, Sanchez R,
Prasad S, Yan Y, Perlow-Poehnelt R, Torrent M, Miller M, Vacca JP, Williams TM, Anthony NJ.
Substituted tetrahydroquinolines as potent allosteric inhibitors of reverse transcriptase and its key
mutants. Bioorg Med Chem Lett 2009;19:51195123.
221. http://www.natap.org/2007/ResisWksp/ResisWksp_42.htm
222. De Clercq E. The next ten stories on antiviral drug discovery (part E): Advents, advances, and
adventures. Med Res Rev 2011;31:118160.
223. Cullen MD, Ho WC, Bauman JD, Das K, Arnold E, Hartman TL, Watson KM, Buckheit RW,
Pannecouque C, De Clercq E, Cushman M. Crystallographic study of a novel subnanomolar
inhibitor provides insight on the binding interactions of alkenyldiarylmethanes with human
immunodeciency virus-1 reverse transcriptase. J Med Chem 2009;52:64676473.
224. Ferris RG, Hazen RJ, Roberts GB, St Clair MH, Chan JH, Romines KR, Freeman GA,
Tidwell JH, Schaller LT, Cowan JR, Short SA, Weaver KL, Selleseth DW, Moniri KR,
Boone LR. Antiviral activity of GW678248, a novel benzophenone nonnucleoside reverse
transcriptase inhibitor. Antimicrob Agents Chemother 2005;49:40464051.
225. Elworthy TR, Dunn JP, Hogg JH, Lam G, Saito YD, Silva TM, Stefanidis D,
Woroniecki W, Zhornisky E, Zhou AS, Klumpp K. Orally bioavailable prodrugs of a BCS
class 2 molecule, an inhibitor of HIV-1 reverse transcriptase. Bioorg Med Chem Lett 2008;18:
63446347.
226. (a) Guo H, Kim CH, Kim HS, Lee CK, Lee IY, Mitchell ML, Son JC, Xu L. Novel HIV reverse
transcriptase inhibitors. WO 2008016522A2. (b) Mitchell ML, Son JC, Lee IY, Lee CK, Kim HS,
Wang J, Hayes J, Wang M, Paul A, Lansdon EB, Chen JM, Eisenberg G, Geleziunas R, Xu L,
Kim CU. N1-Heterocyclic pyrimidinediones as non-nucleoside inhibitors of HIV-1 reverse
transcriptase. Bioorg Med Chem Lett 2010;20:15851588. (c) Mitchell ML, Son JC, Guo H,
Im YA, Cho EJ, Wang J, Hayes J, Wang M, Paul A, Lansdon EB, Chen JM, Graupe D,
Rhodes G, He GX, Geleziunas R, Xu L, Kim CU. N1-Alkyl pyrimidinediones as non-nucleoside
inhibitors of HIV-1 reverse transcriptase. Bioorg Med Chem Lett 2010;20:15891592.
227. (a) Wang Z, Bennett EM, Wilson DJ, Salomon C, Vince R. Rationally designed dual inhibitors of
HIV reverse transcriptase and integrase. J Med Chem 2007;50:34163419. (b) Wang Z, Vince R.
Synthesis of pyrimidine and quinolone conjugates as a scaffold for dual inhibitors of HIV reverse
transcriptase and integrase. Bioorg Med Chem Lett 2008;18:12931296. (c) Wang Z, Vince R.
Design and synthesis of dual inhibitors of HIV reverse transcriptase and integrase: Introducing a
diketoacid functionality into delavirdine. Bioorg Med Chem 2008;16:35873595. (d) Wang Z,
Tang J, Salomon CE, Dreis CD, Vince R. Pharmacophore and structure-activity relationships of
integrase inhibition within a dual inhibitor scaffold of HIV reverse transcriptase and integrase.
Bioorg Med Chem 2010;18:42024211.
228. Zhou Z, Madrid M, Evanseck JD, Madura JD. Effect of a bound non-nucleoside RT inhibitor on
the dynamics of wild-type and mutant HIV-1 reverse transcriptase. J Am Chem Soc 2005;127:
1725317260.
229. Grob PM, Wu JC, Cohen KA, Ingraham RH, Shih CK, Hargrave KD, McTague TL,
Merluzzi VJ. Nonnucleoside inhibitors of HIV-1 reverse transcriptase: Nevirapine as a prototype
drug. AIDS Res Hum Retroviruses 1992;8:145152.
230. Vance D, Shah M, Joshi A, Kane RS. Polyvalency: A promising strategy for drug design.
Biotechnol Bioeng 2008;101:429434.
231. Choi SK. Synthetic multivalent molecules: concepts and biomedical applications. New York:
Wiley; 2004. 418p.

Medicinal Research Reviews DOI 10.1002/med


HIV-1
HIV-1NNRTIs
NNRTIs KK 69
E69

232. Klunder JM, Hoermann M, Cywin CL, David E, Brickwood JR, Schwartz R, Barringer KJ,
Pauletti D, Shih CK, Erickson DA, Sorge CL, Joseph DP, Hattox SE, Adams J, Grob PM. Novel
nonnucleoside inhibitors of HIV-1 reverse transcriptase. 7. 8-Arylethyldipyridodiazepinones as
potent broad-spectrum inhibitors of wild-type and mutant enzymes. J Med Chem 1998;41:
29602971.
233. Cywin CL, Klunder JM, Hoermann M, Brickwood JR, David E, Grob PM, Schwartz R,
Pauletti D, Barringer KJ, Shih CK, Sorge CL, Erickson DA, Joseph DP, Hattox SE. Novel
nonnucleoside inhibitors of HIV-1 reverse transcriptase. 8. 8-Aryloxymethyl- and 8-arylthio-
methyldipyridodiazepinones. J Med Chem 1998;41:29722984.
234. Yoakim C, Bonneau PR, Deziel R, Doyon L, Duan J, Guse I, Landry S, Malenfant E, Naud J,
Ogilvie WW, OMeara JA, Plante R, Simoneau B, Thavonekham B, Bos M, Cordingley MG.
Novel nevirapine-like inhibitors with improved activity against NNRTI-resistant HIV:
8-Heteroarylthiomethyldipyridodiazepinone derivatives. Bioorg Med Chem Lett 2004;14:739742.
235. OMeara JA, Yoakim C, Bonneau PR, Bos M, Cordingley MG, Deziel R, Doyon L, Duan J,
Garneau M, Guse I, Landry S, Malenfant E, Naud J, Ogilvie WW, Thavonekham B, Simoneau B.
Novel 8-substituted dipyridodiazepinone inhibitors with a broad-spectrum of activity against
HIV-1 strains resistant to non-nucleoside reverse transcriptase inhibitors. J Med Chem 2005;48:
55805588.
236. Coulombe R, Fink D, Landry S, Lessard IAD, McCollum R, Naud J, OMeara J, Simoneau B,
Yoakim C, Bonneau PR. Crystallographic study with BILR 355 BS, A novel nonnucleoside
reverse transcriptase inhibitor (NNRTI) with a broad anti HIV-1 prole. 3rd International AIDS
Society Conference on HIV Pathogenesis and Treatment. Rio de Janeiro, July 2427, 2005.
237. (a) Kertesz DJ, Brotherton-Pleiss C, Yang M, Wang Z, Lin X, Qiu Z, Hirschfeld DR, Gleason S,
Mirzadegan T, Dunten PW, Harris SF, Villasenor AG, Hang JQ, Heilek GM, Klumpp K.
Discovery of piperidin-4-yl-aminopyrimidines as HIV-1 reverse transcriptase inhibitors. N-Benzyl
derivatives with broad potency against resistant mutant viruses. Bioorg Med Chem Lett 2010;20:
42154218. (b) Tang G, Kertesz DJ, Yang M, Lin X, Wang Z, Li W, Qiu Z, Chen J, Mei J,
Chen L, Mirzadegan T, Harris SF, Villasenor AG, Fretland J, Fitch WL, Hang JQ, Heilek G,
Klumpp K. Exploration of piperidin-4-yl-aminopyrimidines as HIV-1 reverse transcriptase
inhibitors. N-Phenyl derivatives with broad potency against resistant mutant viruses. Bioorg Med
Chem Lett 2010. DOI: 10.1016/j.bmcl.2010.08.068.
238. Kennedy-Smith JJ, Arora N, Roland Billedeau J, Fretland J, Hang JQ, Heilek GM, Harris SF,
Hirschfeld D, Javanbakht H, Li Y, Liang W, Roetz R, Smith M, Su G, Suh JM, Villasenor AG,
Wu J, Yasuda D, Klumpp K, Sweeney ZK. Synthesis and biological activity of new pyridone
diaryl ether non-nucleoside inhibitors of HIV-1 reverse transcriptase. Med Chem Commun 2010;1:
7983.
239. (a) Nawrozkij MB, Rotili D, Tarantino D, Botta G, Eremiychuk AS, Musmuca I, Ragno R,
Samuele A, Zanoli S, Armand-Ugon M, Clotet-Codina I, Novakov IA, Orlinson BS, Maga G,
Este JA, Artico M, Mai A. 5-Alkyl-6-benzyl-2-(2-oxo-2-phenylethylsulfanyl)pyrimidin-4(3H)-
ones, a series of anti-HIV-1 agents of the dihydro-alkoxy-benzyl-oxopyrimidine family with
peculiar structure-activity relationship prole. J Med Chem 2008;51:46414652. (b) Radi M,
Maga G, Alongi M, Angeli L, Samuele A, Zanoli S, Bellucci L, Ta A, Casaluce G, Giorgi G,
Armand-Ugon M, Gonzalez E, Este JA, Baltzinger M, Bec G, Dumas P, Ennifar E, Botta M.
Discovery of chiral cyclopropyl dihydro-alkylthio-benzyl-oxopyrimidine (S-DABO) derivatives as
potent HIV-1 reverse transcriptase inhibitors with high activity against clinically relevant mutants.
J Med Chem 2009;52:840851.
240. Zhao Z, Wolkenberg SE, Lu M, Munshi V, Moyer G, Feng M, Carella AV, Ecto LT,
Gabryelski LJ, Lai MT, Prasad SG, Yan Y, McGaughey GB, Miller MD, Lindsley CW,
Hartman GD, Vacca JP, Williams TM. Novel indole-3-sulfonamides as potent HIV non-
nucleoside reverse transcriptase inhibitors (NNRTIs). Bioorg Med Chem Lett 2008;18:554559.
241. Piscitelli F, Coluccia A, Brancale A, La Regina G, Sansone A, Giordano C, Balzarini J, Maga G,
Zanoli S, Samuele A, Cirilli R, La Torre F, Lavecchia A, Novellino E, Silvestri R.

Medicinal Research Reviews DOI 10.1002/med


70
E70 KK ZHAN
ZHANETAL.
ETAL.

Indolylarylsulfones bearing natural and unnatural amino acids. Discovery of potent inhibitors of
HIV-1 non-nucleoside wild type and resistant mutant strains reverse transcriptase and coxsackie
B4 virus. J Med Chem 2009;52:19221934.
242. Pontikis R, Dolle V, Guillaumel J, Dechaux E, Note R, Nguyen CH, Legraverend M, Bisagni E,
Aubertin AM, Grierson DS, Monneret C. Synthesis and evaluation of AZT-HEPT, AZT-
pyridinone, and ddC-HEPT conjugates as inhibitors of HIV reverse transcriptase. J Med
Chem 2000;43:19271939.
243. (a) Venkatachalam TK, Sudbeck EA, Mao C, Uckun FM. Stereochemistry of halopyridyl and
thiazolyl thiourea compounds is a major determinant of their potency as nonnucleoside inhibitors of
HIV-1 reverse transcriptase. Bioorg Med Chem Lett 2000;10:20712074. (b) Venkatachalam TK,
Mao C, Uckun FM. Stereochemistry as a major determinant of the anti-HIV activity of chiral
naphthyl thiourea compounds. Antivir Chem Chemother 2001;12:213221. (c) Venkatachalam TK,
Mao C, Uckun FM. Effect of stereochemistry on the anti-HIV activity of chiral thiourea
compounds. Bioorg Med Chem 2004;12:42754284. (d) Venkatachalam TK, Mao C, Uckun FM.
Effect of stereo and regiochemistry towards wild and multidrug resistant HIV-1 virus: Viral potency
of chiral PETT derivatives. Biochem Pharmacol 2004;67:19331946.
244. Hannongbua S. Structural information and drugenzyme interaction of the non-nucleoside
reverse transcriptase inhibitors based on computational chemistry approaches. Top Heterocycl
Chem 2006;4:5584.
245. Pornpan P, Punkvang A, Saparpakorn P, Wolschann P, Hannongbua S. Recent advances in
NNRTI design: Computer-aided molecular design approaches. Current Computer-Aided Drug
Design 2009;5:174199.
246. Smith MB, Smith RH, Jr, Jorgensen WL. Assault on resistance: The use of computational
chemistry in the development of anti-HIV drugs. Curr Pharm Des 2006;12:18431856.
247. Debnath AK. Application of 3D-QSAR techniques in anti-HIV-1 drug design-an overview. Curr
Pharm Des 2005;11:30913110.
248. Hannongbua S, Lawtrakul L, Sotriffer CA, Rode BM. Comparative molecular eld analysis of
HIV-1 reverse transcriptase inhibitors in the class if 1[(2-hydroxyethoxy)-methyl]-6-(phenylthio)-
thymine. Quant Struct-Act Rel 1996;15:389394.
249. Kireev DB, Chretien JR, Grierson DS, Monneret C. A 3D QSAR study of a series of HEPT
analogues: The inuence of conformational mobility on HIV-1 reverse transcriptase inhibition.
J Med Chem 1997;40:42574264.
250. Hannongbua S, Nivesanond K, Lawtrakul L, Pungpo P, Wolschann P. 3D-quantitative structure-
activity relationships of HEPT derivatives as HIV-1 reverse transcriptase inhibitors based on Ab
initio calculations. J Chem Inf Comput Sci 2001;41:848855.
251. Hannongbua S, Pungpo P, Limtrakul J, Wolschann P. Quantitative structure-activity relation-
ships and comparative molecular eld analysis of TIBO derivatised HIV-1 reverse transcriptase
inhibitors. J Comput Aid Mol Des 1999;13:563577.
252. Chen HF, Yao XJ, Li Q, Yuan SG, Panaye A, Doucet JP, Fan BT. Comparative study of non
nucleoside inhibitors with HIV-1 reverse transcriptase based on 3D-QSAR and docking. SAR
QSAR Environ Res 2003;14:455474.
253. Gussio R, Pattabiraman N, Zaharevitz DW, Kellogg GE, Topol IA, Rice WG, Schaeffer CA,
Erickson JW, Burt SK. All-atom models for the non-nucleoside binding site of HIV-1 reverse
transcriptase complexed with inhibitors: A 3D QSAR approach. J Med Chem 1996;39:16451650.
254. Pungpo P, Hannongbua S. Three-dimensional quantitative structure-activity relationships study
on HIV-1 reverse transcriptase inhibitors in the class of dipyridodiazepinone derivatives using
comparative molecular eld analysis. J Mol Graph Model 2000;18:581590,601.
255. Filipponi E, Cruciani G, Tabarrini O, Cecchetti V, Fravolini A. QSAR study and VolSurf
characterization of anti-HIV quinolone library. J Comput Aid Mol Des 2001;15;203217.
256. Samee W, Ungwitayatom J, Matayatsuk C, Pimthon J. 3D-QSAR studies on phthalimide
derivatives as HIV-1 reverse transcriptase inhibitors. ScienceAsia 2004;30:8188.

Medicinal Research Reviews DOI 10.1002/med


HIV-1
HIV-1NNRTIs
NNRTIs KK 71
E71

257. Ragno R, Frasca S, Manetti F, Brizzi A, Massa S. HIV-reverse transcriptase inhibition: Inclusion
of ligand-induced t by cross-docking studies. J Med Chem 2005;48:200212.
258. Sengupta D, Verma D, Naik PK. Docking mode of delvardine and its analogues into the p66
domain of HIV-1 reverse transcriptase: Screening using molecular mechanics-generalized born/
surface area and absorption, distribution, metabolism and excretion properties. J Biosci 2007;32:
13071316.
259. Sengupta D, Verma D, Naik PK. Docking-MM-GB/SA and ADME screening of HIV-1 NNRTI
inhibitor: Nevirapine and its analogues. In Silico Biol 2008;8:275289.
260. Park H, Lee S. Free energy perturbation approach to the critical assessment of selective
cyclooxygenase-2 inhibitors. J Comput Aided Mol Des 2005;19:1731.
261. Kim JT, Hamilton AD, Bailey CM, Domaoal RA, Wang L, Anderson KS, Jorgensen WL.
FEP-guided selection of bicyclic heterocycles in lead optimization for non-nucleoside inhibitors of
HIV-1 reverse transcriptase. J Am Chem Soc 2006;128:1537215373.
262. Jorgensen WL. Efcient drug lead discovery and optimization. Acc Chem Res 2009;42:724733.
263. Varnek A, Solovev VP. In silico design of potential anti-HIV actives using fragment
descriptors. Comb Chem High Throughput Screen 2005;8:403416.
264. Alcaro S, Artese A, Ceccherini-Silberstein F, Chiarella V, Dimonte S, Ortuso F, Perno CF.
Computational analysis of human immunodeciency virus (HIV) Type-1 reverse transcriptase
crystallographic models based on signicant conserved residues found in highly active
antiretroviral therapy (HAART)-treated patients. Curr Med Chem 2010;17:290308.
265. Janssen PA, Lewi PJ, Arnold E, Daeyaert F, de Jonge M, Heeres J, Koymans L, Vinkers M,
Guillemont J, Pasquier E, Kukla M, Ludovici D, Andries K, de Bethune MP, Pauwels R, Das K,
Clark AD, Jr, Frenkel YV, Hughes SH, Medaer B, De Knaep F, Bohets H, De Clerck F,
Lampo A, Williams P, Stoffels P. In search of a novel anti-HIV drug: Multidisciplinary
coordination in the discovery of 4-[[4-[[4-[(1E)-2-cyanoethenyl]-2,6-dimethylphenyl]amino]-2-
pyrimidinyl]amino]benzonitrile (R278474, rilpivirine). J Med Chem 2005;48:19011909.
266. De Corte BL. From 4,5,6,7-tetrahydro-5-methylimidazo[4,5,1-jk](1,4)benzodiazepin-2(1H)-one
(TIBO) to etravirine (TMC125): Fifteen years of research on non-nucleoside inhibitors of HIV-1
reverse transcriptase. J Med Chem 2005;48:16891696.

Dr. Peng Zhan was born in 1983 in Jinan, Shandong province, China. He obtained his B.S.
degree from Shandong University, China, in 2005. Then, he earned his M.S. degree and Ph.D. in
medicinal chemistry from Shandong University under the supervision of Prof. Xinyong Liu in
2008 and 2010, respectively. He is now working as a young researcher in the lab of Prof.
Xinyong Liu. His research area involves design and synthesis of novel HIV-1 non-nucleoside
reverse transcriptase inhibitors.

Xuwang Chen was born in 1987 in Zoucheng, Shandong province, China. He graduated from
Shandong University and obtained his B.S. degree in 2009. At the same year he was
recommended without examination to study for his Ph.D. in the Institute of Medicinal
Chemistry, School of Pharmaceutical Sciences, Shandong University.

Dongyue Li was born in 1984 in Guyuan, Ningxia province, China. In 2007, he graduated from
the School of Pharmaceutical Sciences in Shandong University. He is currently studying for
master degree in the Department of Medicinal Chemistry of the School of Pharmaceutical
Sciences in Shandong University.

Medicinal Research Reviews DOI 10.1002/med


72
E72 KK ZHAN
ZHANETAL.
ETAL.

Zengjun Fang received his M.S. degree from Shandong University in 2007. His Ph.D. project,
which was under the supervision of Prof. Xinyong Liu, was on the design and synthesis of novel
HIV-1 non-nucleoside reverse transcriptase inhibitors as potential antiviral therapeutics.

Erik De Clercq, M.D., Ph.D. has been the Chairman of the Department of Microbiology and
Immunology of the Medical School at the Katholieke Universiteit Leuven (K.U.Leuven) as well
as Chairman of the Board of the Rega Institute for Medical Research (until September 2006).
He is currently the President of the Rega Foundation, a member of the Belgian (Flemish) Royal
Academy of Medicine, a member of the Academia Europaea, and Fellow of the American
Association for the Advancement of Science. He has also been the titular of the Prof. P. De
Somer Chair for Microbiology at the K.U.Leuven. Professor De Clercq received in 1996 the
Hoechst Marion Roussel (now called Aventis) award (American Society for Microbiology),
and in 2000 the Maisin Prize for Biomedical Sciences (National Science Foundation, Belgium)
for his pioneering efforts in the eld of antiviral research. He is an honorary doctor of several
Universities [i.e. Ghent (Belgium), Athens (Greece), Ferrara (Italy), Shandong (Jinan,
China), Charles University (Prague, Czech Republic), and Jihoceska University (Ceske
Budejovice, Czech Republic)]. In 2008 he was elected European Inventor of the Year (Life time
achievement award). His scientic interests are in the antiviral chemotherapy eld, and, in
particular, the development of new antiviral agents for various viral infections, including herpes
simplex virus (HSV), varicella-zoster virus (VZV), cytomegalovirus (CMV), human
immunodeciency virus (HIV), hepatitis B virus (HBV), human papilloma virus (HPV),
and hepatitis C virus (HCV). He has (co)-discovered a number of antiviral drugs, currently
used in the treatment of HSV infections (valaciclovir, Valtrexs, Zelitrexs), VZV infections
(brivudin, Zostexs, Briviracs, Zerpexs), CMV infections (cidofovir, Vistides), HBV
infections (adefovir dipivoxil, Hepseras), and HIV infections (AIDS) (tenofovir disoproxil
fumarate, marketed as Vireads, and, in combination with emtricitabine, as Truvadas, and, in
combination with both emtricitabine and efavirenz, as Atriplas). Vireads has also recently
been approved for the treatment of HBV infections (chronic hepatitis B).

Prof. Dr. Xinyong Liu was born in 1963 in Qingdao, Shandong province, China. He received his
B.S. and M.S. degrees from School of Pharmaceutical Sciences, Shandong University, in 1984
and in 1991, respectively. From 1997 to 1999 he worked at Instituto de Quimica Medica
(CSIC) in Spain as a senior visiting scholar. He obtained his Ph.D. from Shandong University
in 2004. He is currently a distinguished professor, a designated Ph.D. advisor, Director of the
Institute of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University.
His research work is partly engaged in rational drug design, synthesis, and bio-evaluation of a
variety of molecules targeted at the specic enzymes and receptors. At present time, his research
interests are mainly focused on the design and synthesis of novel anti-HIV agents based on the
mechanism of drugs action and computer-assisted drug design, such as HIV-1 (non)nucleoside
reverse transcriptase inhibitors, HIV-1 transactivation inhibitors based on HIV-1 Tat-TAR
interaction, and Rev-RRE interaction in the viral transcription step. His second ongoing
program is total synthesis and structural modications of some natural products from Chinese
Traditional Medicine active in cerebro- and cardio-vascular biology. He has contributed to
about 150 scientic publications and patents as well as many monographs.

Medicinal Research Reviews DOI 10.1002/med

Das könnte Ihnen auch gefallen