Tachedjian et al.indd Drug Target Insights 2007: 2 159–182 159 REVIEW Correspondence: Gilda Tachedjian, Ph.D., Molecular Interactions Group, The Macfarlane Burnet Institute for Medical Research and Public Health, GPO Box 2284, Melbourne, Victoria, 3001, Australia. Tel: 61 3 9282 2256; Fax: 61 3 9282 2100; Email: gildat@burnet.edu.au Please note that this article may not be used for commercial purposes. For further information please refer to the copyright statement at http://www.la-press.com/copyright.htm Targeting Human Immunodefi ciency Virus Type 1 Assembly, Maturation and Budding Johanna Wapling1,2, Seema Srivastava1, Miranda Shehu-Xhilaga3,4 and Gilda Tachedjian1,2,3 1Molecular Interactions Group, Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, 3004, Australia. 2Department of Microbiology, Monash University, Clayton, Victoria 3168, Australia. 3Department of Medicine, Monash University, Prahran, Victoria 3181, Australia. 4Infectious Diseases Unit, Alfred Hospital, Prahran, Victoria 3181, Australia. Abstract: The targets for licensed drugs used for the treatment of human immunodefi ciency virus type 1 (HIV-1) are confi ned to the viral reverse transcriptase (RT), protease (PR), and the gp41 transmembrane protein (TM). While currently approved drugs are effective in controlling HIV-1 infections, new drug targets and agents are needed due to the eventual emergence of drug resistant strains and drug toxicity. Our increased understanding of the virus life-cycle and how the virus interacts with the host cell has unveiled novel mechanisms for blocking HIV-1 replication. This review focuses on inhibitors that target the late stages of virus replication including the synthesis and traffi cking of the viral polyproteins, viral assembly, maturation and budding. Novel approaches to blocking the oligomerization of viral enzymes and the interactions between viral proteins and host cell factors, including their feasibility as drug targets, are discussed. Keywords: HIV-1, antiretroviral drugs, drug targets, assembly, maturation, budding, protease dimerization, reverse transcriptase dimerization. Introduction HIV-1 is a major public health problem affecting an estimated 40 million individuals worldwide (www. unaids.org). Although it has been over 20 years since HIV-1 was identifi ed as the etiologic cause of acquired immune defi ciency syndrome (AIDS) an effective vaccine is not available. Thus, apart from public health measures that aim at HIV-1 prevention, the only effective strategy for controlling HIV-1 infections and lowering HIV-1 transmission is the use of antiretroviral drugs either for the treatment or prevention of infections. Current antiretroviral drugs belong to four classes, the nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs), nonnucleoside reverse transcriptase inhibitors (NNRTIs), protease (PR) inhibitors (PI) and fusion inhibitors (Vivet-Boudou et al. 2006; De Clercq, 1998; Abdel-Rahman et al. 2002; Manfredi and Sabbatani, 2006). NRTIs and NNRTIs are respectively, competitive and allosteric inhibitors of the HIV-1 reverse transcriptase (RT) and act early in the viral life-cycle by blocking the conversion of the viral RNA genome into a double stranded proviral DNA precursor (Shehu-Xhilaga et al. 2005). Fuzeon (enfuvirtide or T20) is a peptide that also acts early in the virus life-cycle by preventing viral entry through interaction with the gp41 transmembrane protein (Shehu-Xhilaga et al. 2005). In contrast, PIs inhibit the late stage of virus replication by blocking the specifi c cleavage of Gag and Gag-Pol polyproteins to mature structural proteins and enzymes (Shehu-Xhilaga et al. 2005). Early antiretroviral regimens consisted of one or two RTIs, which were delivered as sequen- tial monotherapy and led to treatment failure (Piacenti, 2006). The advent of combination therapy, or highly active antiretroviral therapy (HAART) since 1996 has been responsible for a dramatic decrease in AIDS mortality (Palella et al. 1998). Current HAART regimens generally comprise three antiretroviral drugs, usually two NRTIs and either a PI or an NNRTI (Yeni et al. 2002). While an armoury of agents is available for the treatment of HIV-1 patients, new drugs and drug targets need to be identifi ed due to drug toxicity and the eventual emergence of drug resistant strains to current antiretroviral inhibitors (Clavel and Hance, 2004). Moreover, resistance to one drug normally results 160 Wapling et al Drug Target Insights 2007: 2 in cross-resistance to inhibitors of the same class, rendering a large number of agents to limited clinical use (Clavel and Hance, 2004). Therefore, the development or availability of new drugs such as Fuzeon, the HIV-1 integrase inhibitor ralte- gravir (MK-0518) (Grinsztejn et al. 2007) and the CCR5 antagonist maraviroc (Stephenson, 2007) that remain active against drug resistant virus is essential for the continuing success of HAART (Yeni, 2006). The increased understanding of how HIV-1 reproduces and interacts with the host cell machinery has resulted in the identifi cation of potential drug targets, which can be exploited for the development of new classes of inhibitors. Here we describe strategies and agents that block the late stages of HIV-1 replication including the synthesis and traffi cking of viral polyproteins, viral assembly, maturation and budding. Novel approaches to blocking the oligomerization of viral enzymes and the interactions between viral proteins and host cell factors are discussed including their feasibility as drug targets. While peptidomimetic PIs act at the late stage of HIV-1 replication to block viral maturation, this review will deal with agents that inhibit HIV-1 PR by novel mechanisms that are distinct to these transition state mimetics that are competi- tive inhibitors of the HIV-1 PR. Late Stages of the HIV-1 Life Cycle Following virus attachment, fusion and uncoating the single stranded positive sense RNA genome of HIV-1 is reverse transcribed by the viral RT into a proviral DNA precursor in a reverse tran- scription complex (RTC) containing viral and possibly host cell factors (Fig. 1). The RTC matures into a preintegration complex (PIC) and traffi cs to the nucleus where the viral cDNA is inserted into the host cell chromosome by the HIV-1 integrase (IN) (Telesnitsky A. and Goff, 1997). The processes from entry up to and including integration are defi ned as the early steps in the viral life cycle. The late stage of virus replication begins with transcription of the viral mRNAs from the integrated provirus (Fig.1). Singly and multiply spliced mRNAs encode the HIV-1 envelope proteins and regula- tory/accessory proteins, respectively (Rabson and Graves, 1997). Pr55gag (Gag) and Pr160gag-pol (Gag-Pol) polyproteins are translated from unspliced mRNAs (Swanstrom, 1997). Formation of two types of polyproteins from the same unspliced mRNA is mediated by a ribosomal frameshifting mechanism that brings the pol sequence in the same reading frame as gag. Perturbation of ribosomal frameshifting leads to changes in the Gag and Gag-Pol ratio that is detrimental to virus assembly, morphogenesis and release (Swanstrom, 1997). Gag encodes the viral structural proteins matrix (MA), capsid (CA), nucleocapsid (NC), p6 and two spacer peptides, p1 and p2. Gag-Pol also encodes MA, CA and NC in addition to the three viral enzymes, PR, RT and IN. After trans- lation, Gag and Gag-Pol are targeted to the host cell plasma membrane, a process that is depen- dent on the myristoylation of the N-terminus of Gag (Fig. 1) (Swanstrom, 1997). Inhibition of myristoylation disrupts the proper targeting of Gag and Gag-Pol to the plasma membrane (Swanstrom, 1997). Gag-Gag, Gag/Gag-Pol and Gag-RNA interac- tions are also essential for the proper assembly and maturation of infectious virions. Gag and Gag-Pol assemble at the plasma membrane along with viral envelope glycoproteins gp120 and gp41 to form immature viral particles (Fig. 1). Gag is necessary and suffi cient for virus particle forma- tion (Freed, 1998; Swanstrom, 1997). The viral genomic RNA is also packaged into virions through interactions with the NC of Gag and a psi packaging signal in the genome (Swanstrom, 1997). As the newly assembled virions bud from the cell it is believed that Gag-Pol polyproteins oligomerize in order to activate the HIV-1 PR by forming an active PR homodimer. This results in the sequential cleavage of Gag and Gag-Pol into the mature structural proteins and enzymes (Kaplan et al. 1994; Pettit et al. 1998). Agents that bind to domains in Gag or Gag-Pol and modulate their oligomerization are likely to have a negative effect on virus assembly, maturation and budding (Fig. 1). Agents that interfere with HIV-1 PR mediated cleavage of Gag and Gag-Pol result in the production of immature viral particles that are non-infectious (Kohl et al. 1988). Virus particle budding and egress is mediated by interactions of viral proteins such as the p6 late domain with components of the endosomal sorting machinery. Ion channels formed by viral protein U (Vpu) also facilitate viral particle egress from the host cell. Below we describe in more detail the 161 Targeting the late stages of HIV-1 replication Drug Target Insights 2007: 2 specifi c processes required for viral assembly, maturation and budding and agents that have been described that block these steps. Inhibitors of Gag and Gag-Pol Expression: Targeting Ribosomal Frameshifting HIV-1 Gag and Gag-Pol polyproteins are encoded by overlapping open reading frames on the same unspliced mRNA. During translation Gag-Pol is synthesized by a -1 ribosomal frameshifting mechanism that occurs at a frequency of 5 to 10% of Gag translation events (Jacks et al. 1988b). Similar frameshifting mechanisms are also used by other retroviruses including Rous sarcoma virus and Mouse mammary tumor virus in order to regulate expression of Gag-Pol (Jacks and Varmus, 1985; Jacks et al. 1987; Jacks et al. 1988a). The HIV-1 frameshift site is a heptanucleotide AU-rich sequence (UUUUUUA) found at the 3′ end of the NC coding sequence and is conserved amongst HIV-1 isolates. This slippery sequence and a downstream RNA stem loop structure stall the ribosome during the synthesis of Gag, allowing the ribosome to slip Figure 1. Overview of the HIV-1 life-cycle. Early events in virus replication include attachment, fusion and uncoating of the virus followed by reverse transcription in the cytoplasm of the cell, nuclear import of the preintegration complex and integration of the proviral DNA precur- sor into the host cell chromosome. Late events begin with transcription of unspliced and spliced RNA from the provirus and export of the mRNAs to the cytoplasm, resulting in the translation of Gag, Gag-Pol, Env and the accessory and regulatory proteins of HIV-1. Regulation of Gag-Pol synthesis is mediated by a ribosomal frameshifting mechanism from unspliced mRNA that also expresses Gag. Myristoylation of Gag is necessary for traffi cking of Gag and Gag-Pol to the site of viral assembly. Assembly is driven by interactions between Gag-Gag, Gag/Gag-Pol, Gag-RNA. Viral budding and egress involves host cell factors. During or shortly after budding the HIV-1 PR cleaves the Gag and Gag-Pol polyproteins resulting in a mature and infectious viral particle. Late Event Targets Frameshifting Myristoylation & Trafficking Assembly Budding & Egress Maturation Early Events Late Events Recognition & Attachment Fusion Uncoating & Reverse Transcription Nuclear Import mRNA export Translation Myristoylation & Trafficking Maturation Integration Transcription & Splicing Assembly Budding & Egress RTCPIC Gag/Gag-Pol ratio Myristoylation Gag/Gag interactions Gag/Gag-Pol interactions Gag/RNA interactions VPU p6 PTAT motif PR dimerization RT dimerization IN dimerization Gag cleavage 162 Wapling et al Drug Target Insights 2007: 2 back one nucleotide and enable synthesis of the Gag-Pol fusion protein (Jacks et al. 1988b). This sequence, the stem-loop structure and its stability and adjacent interacting sequences are believed to be the key components of the frameshifting signal (Jacks et al. 1988b; Kollmus et al. 1994; Hill et al. 2005). Details of a recently reported NMR structure and an analysis of current HIV-1 frameshifting models have recently been reviewed (Brierley and Dos Ramos, 2006). Studies demonstrate that perturbation of the Gag/Gag-Pol ratio result in major defects in virus replication, suggesting that interfering with ribo- somal frameshifting represents a viable drug target. Alteration of the Gag/Gag-Pol ratio, by engineering vectors with gag and pol genes in the same open reading frame, results in major defects in assembly and budding (Karacostas et al. 1993; Park and Morrow, 1991). The block in virus assembly is partially overcome by inhibition of the HIV-1 PR, suggesting that increased HIV-1 PR activity is responsible for the defect (Karacostas et al. 1993). A later study, in which the impact of decreasing the ratio of Gag/Gag-Pol on virion production was determined by co-transfection of plasmids expressing Gag and Gag-Pol alone demonstrate that the maintenance of this ratio is not only important for HIV-1 replication but also for virion RNA dimer formation and stability (Shehu-Xhilaga et al. 2001a). Furthermore, a decrease in Gag-Pol translation results in major defects in virus maturation and HIV-1 infectivity (Dulude et al. 2006). The small molecule, 1,4- bis-[N-(3-N,N-dimethylpropyl)amidino]benzene tetrahydrochloride (RG501, Table 1), is thought to enhance ribosomal frameshifting of HIV-1 by binding to the RNA stem loop structure of the ribosomal frameshifting signal resulting in increased ribosomal pausing (Hung et al. 1998). The imbalance in the resulting Gag/Gag-Pol ratio is associated with inhibition of acute and chronic HIV-1 infection in CCRF-CEM cells and peripheral blood mononuclear cells. Targeting Gag and Gag-Pol Traffi cking During the late phase of the viral life cycle, Gag polyproteins are targeted to the plasma membrane, where they are believed to colocalise to lipid raft microdomains for assembly into immature virions (Morikawa et al. 1996; Bryant and Ratner, 1990; Bouamr et al. 2003; Ding et al. 2003; Holm et al. 2003; Tang et al. 2004). Membrane targeting of Gag is mediated by the N-terminal myristoyl group in concert with conserved basic amino acids at the N-terminus of the MA domain of Gag (Bryant and Ratner, 1990; Facke et al. 1993; Ono and Freed, 1999; Ono et al. 2000). Myristic acid is a saturated 14-carbon fatty acid, post transationally attached to the N-terminal glycine of both Gag and Gag-Pol (Veronese et al. 1988). Myristoylation of Gag but not Gag-Pol is critical for targeting these polypro- teins to the plasma membrane (Park and Morrow, 1992; Smith et al. 1993). Mutations that interfere with Gag myristoylation inhibit viral budding and misdirect virion assembly to the cytosolic fraction (Gottlinger et al. 1989; Bryant and Ratner, 1990). However, complete inhibition of Gag myris- toylation is necessary to block HIV-1 budding (Morikawa et al. 1996). Myristoylation is a two-step process involving activation of myristate to myristoyl-CoA by acyl- CoA synthetase and transfer of the myristoyl moiety from the myristoyl-CoA substrate to the N-terminal glycine of Gag by the enzyme N-myristoyltransferase (NMT) (Morikawa et al. 1996; Veronese et al. 1988). This pathway has been utilized to deliver alternate myristoylation substrates that perturb viral assembly. The myristic acid analogue 12-azidododeca- noic acid is a potent inhibitor of HIV-1 produc- tion in acute and chronically infected T-cell lines, exhibiting a maximum inhibitory effect between 10–50 µM at noncytotoxic concentrations, however the mechanism of action is not defi ned (Devadas et al. 1992). Another analogue, 4-oxatetra-decanoic acid, reduces HIV-1 replication in a T-cell line at 18 µM (Langner et al. 1 9 9 2 ) . Heteroatom-substituted analogs of myristic acid such as 12-methoxydodecanoate (13-oxamyristate or 13-OxaMyr), 5-octyloxypen- tanoate (6-oxamyristate or 6-OxaMyr), 11-ethyl- thioundecanoic acid and 12-thioethyldodecanoic acid act as alternate substrates for Gag myris- toylation (Bryant et al. 1989; Bryant et al. 1991; Parang et al. 1997) and can prevent membrane binding of the modifi ed Gag proteins (Bryant et al. 1989; Bryant et al. 1991). Of the heteroatom substi- tuted analogs, 13-OxaMyr is the most potent inhibitor. 13-OxaMyr is added to Gag with an effi cacy similar to that of myristate and alters viral polyprotein processing, which is suggested to be a consequence of inhibiting Gag and 163 Targeting the late stages of HIV-1 replication Drug Target Insights 2007: 2 Table 1. Inhibitors of the late stages of HIV-1 replication. Inhibitor Description Reference Ribosomal frameshifting 1,4-bis-[N-(3-N,N Small molecule (Hung et al. 1998). dimethylpropyl)amidino]benzene tetrahydrochloride (RG501) Myristoylation and Traffi cking 12-azidododecanoic acid Myristic acid analogue (Devadas et al. 1992) 4-oxatetra-decanoic acid (Langner et al. 1992) 12-methoxydodecanoate Heteroatom-substituted (Bryant et al. 1989) 5-ocytl-oxypentanoate myristic acid analogues (Bryant et al. 1991) 11-ethylthioundecanoic acid (Parang et al. 1997) 12-thioethyldodecanoic acid 5-cis-tetradecenoic acid Unsaturated 14-Carbon (Lindwasser and Resh, 2002). (physeteric acid) fatty acids 5-cis,8-cis-tetradecenoic acid (goshuyic acid) Assembly—Gag/Gag Interactions CAP-1 Small molecule (Tang et al. 2003) PAATLEEMMTA CA derived peptide (Niedrig et al. 1994). GPG-NH2 CA derived tripeptide amide (Hoglund et al. 2002) CAI Peptide (Sticht et al. 2005) (Ternois et al. 2005) Maturation—Gag processing 3-0-(3′-3′-dimethylsuccinyl)-betulinic Small molecule (Li et al. 2003) acid (Zhou et al. 2004) (PA-457/bevirimat) (Sakalian et al. 2006) electrophilic disulfi de-substituted NC Zn fi nger inhibitor (Rice et al. 1995) benzamides (DIBAs) (Turpin et al. 1996) 1,2-dithiane-4,5-diol,1,1-dioxide NC Zn fi nger inhibitor (Rice et al. 1997) (NSC 624151) S-acyl 2-mercaptobenzamide thioester NC Zn fi nger inhibitor (Schito et al. 2006). (SAMT) Maturation—PR dimerisation Ac-TLNF-OH PR C-terminal tetrapeptide (Zhang et al. 1991) Pal-YDL-OH Modifi ed PR C-terminal (Schramm et al. 1999) Pal-YD-(biphenylalaine)-OH lipopeptides Pal-YDT-OH Apam(2)-YD-thyroxine-OH (Dumond et al. 2003) PQITL(GGG)CTLNF Glycine linked PR interface (Babe et al. 1992) tetra-peptides HO-FNLTS-NH-(CH2)n-N-PQITLW-OH Alkyl linked PR interface (Zutshi et al. 1997) peptides (Ulysse and Chmielewski, 1998) (Zutshi and Chmielewski, 2000) (Continued) 164 Wapling et al Drug Target Insights 2007: 2 Molecular tongs Scaffold constrained PR (Bouras et al. 1999) interface peptides (Breccia et al. 2003) (Merabet et al. 2004) (Hwang and Chmielewski, 2005) (Bannwarth et al. 2006) β-sheet peptide/peptidomimetic PR interface derived (Song et al. 2001) peptidomimetics PQITL-RKKRRQRRRPPQV-SFNF- PR C-terminal fusion (Davis et al. 2006) C/ATLN (P27/P27A) peptide BocFψ[CH2NH]FEF-NH-CH2-CO- Linked PR C-terminal (Uhlikova et al. 1996) TLNF-OH tetrapeptide—active site (Skalova et al. 2003) inhibitor Pentaester 13e Didemnaketal A analogue (Fan et al. 1998) Ursolic Acid Triterpene (Quere et al. 1996) NHGRNLLTQI (S8) PR LES peptide (Broglia et al. 2005) (Broglia et al. 2006) IVQVDAEG (p51) Random peptide (Park and Raines, 2000). Vpr-(spacer)-TLNF-OH Vpr, PR C-terminal fusion peptide (Cartas et al. 2001). Maturation—RT dimerisation [2′,5′-bis-O-(tert-butyldimethylsilyl)-β- Small molecule (Sluis-Cremer et al. 2000) D-ribofuranosyl]-3′-spiro-5″-(4″-amino- (Rodriguez-Barrios et al. 1′,2″-oxathiole-2″,2″-dioxide) 2001) thymine (TSAO-T) N-(4-tert-Butylbenzoyl)-2-hydroxy-1- Small molecule (Arion et al. 2002) naphthaldehyde hydrazone) (BBNH) (Sluis-Cremer and Tachedjian, 2002) (Himmel et al.) KETWETWWTE (Pep-7) RT connection subdomain peptide (Morris et al. 1999) (Depollier et al. 2005) TLMALELKGKLLLAGLAPSAFLPLSFP Designed peptide targeting (Campbell et al. 2002) EGL (TLMA2993) RT connection subdomain (Hosokawa et al. 2004) Maturation—IN Dimerisation INI 1 Host cell factor (Yung et al. 2001) (Sorin et al. 2006) (Ariumi et al. 2006) (Kalpana et al. 1994) Budding and Egress 5-(N,N-hexamethylene)amiloride (HMA) Amiloride analogue (Ewart et al. 2002) 5-(N,N-dimethyl)amiloride (DMA) (Ewart et al. 2004) Vpu Binding protein (UBP) Host cell factor (Callahan et al. 1998) (Handley et al. 2001) (Harila et al. 2006) (Neil et al. 2006) TSG101 Host cell factor (Garrus et al. 2001) (Demirov et al. 2002) (Goila-Gaur et al. 2003) zLLL/MG-132 Lactocystin Proteosome inhibitor (Schubert et al. 2000) (Ott et al. 2003) Abbreviations: Ac: acetylation; Pam: palmitoyl; Apam: 2-aminopalmitic acid Table 1. (Continued) Inhibitor Description Reference 165 Targeting the late stages of HIV-1 replication Drug Target Insights 2007: 2 Gag-Pol traffi cking (Bryant et al. 1991). In an acutely infected T-cell line 13-OxaMyr reduces HIV-1 replication in the 40–80 µM range (Bryant et al. 1989). 13-OxaMyr also inhibits viral production in chronically infected H9/IIIB cells, which is consistent for an inhibitor that targets the late stage of HIV-1 replication (Bryant et al. 1991). 13-OxaMyr exhibits a synergistic anti- HIV-1 effect with AZT suggesting its potential for use in combination therapy (Bryant et al. 1991). The therapeutic effi cacy of 13-OxaMyr can be further enhanced by conjugation with glycerophospholipid L-∝-phosphatidylethanol- amine (Pidgeon et al. 1993). The selectivity of these heteroatom analogs for the target protein is dependent on the position of the substituted heteroatom, thus they can be exploited as a therapeutic antiretroviral strategy. Nevertheless, heteroatom-substituted myristic acid analogs are still expected to adversely affect a substantial range of cellular processes that depend on protein N-myristoylation (Lindwasser and Resh, 2002). An alternative strategy for targeting Gag myris- toylation is the exogenous treatment of cells with unsaturated 14-carbon fatty acids including 5-cis- tetradecenoic acid (14:1n-9, physeteric acid) and 5-cis,8-cis-tetradecadienoic acid (14:2n-6, goshuyic acid) (Lindwasser and Resh, 2002). As lipid rafts have preference for saturated fatty acids, treatment with unsaturated analogs interferes with membrane targeting of Gag and consequentially viral assembly and production (Lindwasser and Resh, 2002). These inhibitors also interfere with certain Src- kinase mediated cellular pathways, although they appear to have no effect on cell proliferation (Campbell and Vogt, 1995). It is suggested that direct dietary intake of physeteric acid and goshuyic acid could be a useful therapeutic strategy for the treatment of HIV-1 infections. However, the effect of long term intake of these unsaturated fatty acids and their effect on N-myristoylated signaling proteins such as Src, G-proteins, Arf and heteroge- neously N-acylated retinal proteins needs to be assessed (Lindwasser and Resh, 2002). Recent studies also indicate a role for phospha- tidylinositide 4,5-bisphosphate [PI(4,5)P2] in regulating Gag localization (Ono et al. 2004). In HIV-1, binding of PI(4,5)P2 to the MA domain in Gag activates the “myristyol switch” and also acts as the point of membrane attachment (Saad et al. 2006). The binding site of PI(4,5)P2 on MA is highly conserved amongst HIV-1 strains and therefore represents an attractive antiviral target (Shkriabai et al. 2006; Saad et al. 2006). Targeting HIV-1 CA CA plays an important role in the HIV-1 life-cycle by promoting Gag-Gag interactions during virion maturation. The N- and C-terminal domains of this protein serve distinct functions. As shown by mutational analysis, the N terminal domain of CA (N-CA), otherwise known as the NTD, is respon- sible for maintaining the proper conformation of CA during the assembly process (Worthylake et al. 1999; Li et al. 2000). The C-terminal domain of CA (C-CA) or the CTD, is critical for Gag-Gag interactions during assembly and maturation (Gamble et al. 1996; Gamble et al. 1997) and described mutations in this region have major consequences on virion maturation and infectivity (von Schwedler et al. 2003; Ganser-Pornillos et al. 2004). The NMR structure of CA has demonstrated that the protein consists mainly of seven α-helices, two β-hairpins and a loop structure (Momany et al. 1996; Gitti et al. 1996). Five of the α-helices form a coiled-coiled structure while one of the β-hairpins is located on the surface of the N-terminal domain of the protein (Momany et al. 1996). The second β-hairpin is predicted to be formed after cleavage by the HIV-1 PR (Tang et al. 2002). Cleavage of CA from its neighbouring proteins is necessary for core condensation and conical capsid shell formation (Vogt, 1996; Wiegers et al. 1998). Compounds that bind to these regions would be expected to disrupt proper CA shell formation and virion infectivity making CA an important and attractive target for the development of antiretro- viral agents. A proof of concept study, demonstrating the potential of inhibiting CA-CA interactions as an antiretroviral target has been published (Tang et al. 2003). Computational high throughput screening of a small molecule library and NMR analysis for binding specifi city resulted in the identifi cation of CAP-1 and CAP-2 which bind to an apical site on the NTD of both immature and mature CA (Tang et al. 2003). While CAP-2, is toxic to U1 cells, CAP-1 reduces viral infectivity by 95% at 100 µM. The released virions lack cone shaped cores and resemble viral particles that have been observed in HIV-1 expressing mutations that disrupt CA-CA interactions (Dorfman et al. 1994; Reicin et al. 1996; von Schwedler et al. 2003; 166 Wapling et al Drug Target Insights 2007: 2 Lanman et al. 2003). Despite aberrant viral morphology CAP-1 does not affect viral particle release or proteolytic processing (Tang et al. 2003). CAP-1 and CAP-2 bind to a common site within the NTD thus preventing CA-CA interac- tions and proper Gag assembly. Peptides derived from HIV-1 CA have also been described to affect viral morphogenesis by interfering with capsid formation (Niedrig et al. 1994). The synthetic peptide, PAATLEEMMTA, inhibits HIV-1 replication in cell culture assays at 20–200 µg/ml and results in the production of immature and aberrant viral particles (Niedrig et al. 1994). Tripeptide amides derived from the carboxyl terminus of CA inhibit HIV-1 replication, with the three most potent peptides interacting with CA as demonstrated by capillary electrophoresis analysis (Hoglund et al. 2002). Glycyl-prolyl-glycine-amide (GPG-NH2) inter- feres with the formation of HIV-1 particles with a normal conical core structure (Hoglund et al. 2002). G-NH2 is an active metabolite of GPG-NH2 indicating that the latter acts as a pro-drug (Andersson et al. 2005). However, the development of HIV-1 resistance to either G-NH2 or GPG-NH2 has been elusive suggesting that the peptides mediate their effects through a host cell or other factor (Andersson et al. 2004). CAI, a small peptide selected by phage display screening, acts as an inhibitor of the assembly of immature Gag in vitro (Sticht et al. 2005; Ternois et al. 2005). CAI binds to the C-terminus of CA (Kd ~ 800 µM), thus preventing the necessary conformational changes in CA that lead to the formation of mature cores (Sticht et al. 2005). The structure of CAI complexed with CA has revealed that the CAI binding region is a highly conserved hydrophobic pocket within the C terminus of CA where the peptide forms an extra α-helix, which binds to the four α-helices of CA (Ternois et al. 2005). The resulting protein- peptide complex is therefore a fi ve α-helix bundle with reduced CA-CA dimerization contacts that destabilizes the dimer interface. Binding of CAI to the C-CA not only affects the assembly of the immature capsid particles but also reduces the amount of correctly assembled mature capsids in vitro, thus acting as a promising two-step inhibitor (Sticht et al. 2005). The C-terminal domain of Gag in the context of Gag-Pol is essential for its interaction with Gag and its incorporation into the virion (Srinivasakumar et al. 1995; Chiu et al. 2002; Chien et al. 2006). This sequence includes a highly conserved “major homology region” (MHR) in the CA domain of Gag and the adjacent CA-SP1 (Srinivasakumar et al. 1995; Chien et al. 2006). These sequences are also critical for HIV-1 Gag assembly as they drive Gag oligomerization. However, the magni- tude of the virion incorporation defect of Gag-Pol MHR deletion mutants varies between different studies making the value of targeting this region of Gag-Pol unclear with respect to inhibition of the late stages of HIV-1 replication (Mammano et al. 1994; Srinivasakumar et al. 1995; Chiu et al. 2002; Chien et al. 2006). Sequences involved in Gag and Gag-Pol interactions are assumed to be similar to those involved in Gag-Gag interactions. However, virions generated in the presence of CAP-1 are unlikely to affect Gag/Gag-Pol interactions as defects in proteolytic processing in the virus or virion associated RT activity were not observed (Tang et al. 2003). The proline rich region of p6 has also been implicated in the packaging of cleaved Pol proteins into virions, which is suggested to be mediated by host cell proteins (Dettenhofer and Yu, 1999; Cen et al. 2004). Identifying the host cell factor implicated in the virion incorporation of cleaved Pol will be necessary for establishing this process as a viable drug target. Targeting HIV-1 NC The HIV-1 NC (NCp7) contains two highly conserved zinc fi nger motifs C-X2-C-X4-H-X4-C (X, any amino acid). The zinc fi ngers of NC are critical in the early and late stages of HIV-1 replication with mutations in the zinc chelating amino acids resulting in formation of non- infectious virus (Aldovini and Young, 1990). The zinc fi ngers of NCp7 are required for initi- ation, elongation and effi cient template switching d u r i n g r e v e r s e t r a n s c r i p t i o n ( R o d r i g u e z - Rodriguez et al. 1995; Tanchou et al. 1995). NCp7 is also involved in HIV-1 genomic RNA dimerization, IN cleavage activity and coats the viral RNA genome protecting it from nucleases (Lapadat-Tapolsky et al. 1993). Given the critical role of NCp7 zinc fi ngers in HIV-1 replication it is not surprising that agents that covalently modify the zinc chelating residues of NCp7 have been described as inhibitors of HIV-1 replication (Rice et al. 1995). The electrophilic 167 Targeting the late stages of HIV-1 replication Drug Target Insights 2007: 2 disulfi de-substituted benzamides (DIBAs) inactivate cell free virus and inhibit the early and late stages of HIV-1 replication by interfering with reverse transcription and viral particle maturation (Rice et al. 1995; Turpin et al. 1996). In the U1 cell line treatment with DIBAs results in the inhibition of virus particle release, processing of Gag, and the production of virions with reduced infectivity (Turpin et al. 1996). The defect in viral particle release and maturation was attributed to the formation of intermolecular cross-linkages between the zinc fi ngers of adjacent Gag mole- cules, thereby preventing effi cient cleavage by the HIV-1 PR (Turpin et al. 1996). T h e n o n - d i s s o c i a b l e t e t h e r e d d i t h i a n e compound 1,2-dithiane-4,5-diol,1,1-dioxide, (NSC 624151) also mediates similar defects in Gag processing (Rice et al. 1997). Although cellular proteins also contain zinc fingers, these inhibitors appear to preferentially target retro- viral zinc fingers. This may be explained by the inaccessibility of these inhibitors to the appro- priate cellular compartments where zinc finger containing cellular proteins are located. The in vivo anti-HIV-1 activity of zinc finger inhibitors has been demonstrated in a transgenic murine model where infectious HIV-1 is induced from an integrated provirus (Schito et al. 2003). A recent study in a nonhuman primate model demonstrated a reduction in the levels of SIV/ DeltaB670 in peripheral blood mononuclear cells during therapy with the zinc finger inhib- itor, S-acyl 2-mercaptobenzamide thioester (SAMT), although there was no effect on viral load (Schito et al. 2006). Further studies are in progress to optimise the bioavailability and pharmacokinetics of this promising inhibitor. Targeting HIV-1 PR Much of our understanding of how the PR domain in Gag-Pol is activated and the processing cascade of Gag and Gag-Pol are due to the contributions of Kaplan and colleagues (Kaplan et al. 1994; Pettit et al. 2005). Strict regulation of PR function is critical for effi cient production of mature viral particles. Premature activation, partial inhibition, or over-expression of HIV-1 PR leads to major defects in viral assembly and the production of non-infectious viral particles (Krausslich, 1991; Kaplan et al. 1993; Karacostas et al. 1993). Hence novel inhibitors designed to prevent or perturb PR dimerization could potentially inhibit the mature PR homodimer and the immature Gag-Pol embedded PR. Targeting the PR Dimer Interface with Interface Peptides HIV-1 PR is a homodimeric aspartyl protease formed by the symmetrical association of two 99 amino acid subunits. The crystal structure reveals a compact, predominantly β-strand structure with a short α-helix region near the C terminus (Wlodawer et al. 1989). Dimerization of the PR monomers generates both the substrate-binding pocket and the catalytic centre and is essential for PR activity (Cheng et al. 1990). The PR dimer has a dissociation constant of 50 nM and Gibbs free energy of dimer stabilisation of 10 kcal/mol (25 oC, pH 3.4). Nearly 75% of the binding energy is contributed by the four-stranded β-sheet formed by the N- and C-termini (Todd et al. 1998).The four-stranded β-sheet comprising the N- and C-termini from each PR monomer represents an attractive drug target for the following reasons: 1. It is the major stabilising region of the active dimer, 2. The region is relatively free of known PR resis- tance mutations, 3. The sequence is highly conserved in most HIV-1 and HIV-2 isolates and 4. It provides a unique target minimising potential toxicity issues for eukaryotic aspartyl proteases (Gustchina and Weber, 1991). A standard methodology for analysing potential PR inhibitors that prevent PR dimerization (disso- ciative inhibition) or target and bind to the PR active site (competitive inhibition) has been described (Zhang et al. 1991). An example of a dissociative inhibitor is the C-terminal tetrapeptide, Ac-T-L-N-F, which exhibits activity in the micro- molar range (Ki 45 µM) (Zhang et al. 1991). Other studies have also shown the capacity of N- and C-terminal peptides, or ‘interface’ peptides, to bind to PR monomers and thus prevent PR dimerization and activity (Babe et al. 1991; Franciskovich et al. 1993; Schramm et al. 1991; Schramm et al. 1996). The identifi cation of these lead peptides provides proof of concept that targeting the PR β-sheet region constitutes a viable strategy for the develop- ment of novel inhibitors of HIV-1 PR. The potency of C-terminal tetrapeptides are increased by truncation to a core tripeptide, amino acid modifi cation, and the addition of a linear hydrophobic moiety such as palmitoyl to the amino 168 Wapling et al Drug Target Insights 2007: 2 HIV-1 PR monmer N-terminal C-terminal peptide peptide scaffold ‘molecular tong’ terminus of the peptide. The lipid moiety is thought to increase the dissociative activity of the peptides by directing it to the hydrophobic PR interface (Schramm et al. 1999). However, despite their capacity to inhibit PR activity at low nanomolar concentrations, these lipopeptides are poorly soluble and susceptible to protein degradation. Further modifi cations have been made to the lipo- peptides by making them less peptide-like (Cafl isch et al. 2000) and by modifying the lipid moiety to increase their solubility while retaining potency (Dumond et al. 2003). Cross-linking interfacial peptides represent another strategy, with the aim to increase the affi nity of the peptides by presenting them in a conformation similar to a PR monomer. The fi rst interface tetrapeptides tethered with a glycine linker display greater potency (PF1, IC50= 40 µM) compared to free tetrapeptides (IC50 ≥ 150 µM) (Babe et al. 1992). This approach has evolved to linking peptides with fl exible alkyl tethers (Zutshi and Chmielewski, 2000) and semirigid alkyl based tethers (Ulysse and Chmielewski, 1998), which increase the distance between the peptides to approximate that of the PR termini in the dimer (~10 Å). The conformational freedom of these linked peptides was addressed by the use of pyridinediol and naphthalene based molecu- larly constrained scaffolds (Bouras et al. 1999; Song et al. 2001; Merabet et al. 2004; Bannwarth et al. 2006). Known as ‘molecular tongs’, these compounds are designed to position the interface peptides to clamp the termini of a PR monomer (Fig. 2). These studies have culminated in a set of optimised tongs with symmetrical peptidomimetic sequences based on an optimised PR C-terminal sequence. The tongs inhibit the activity of HIV-1 PR that are either sensitive or resistant to PIs with Ki values from 0.4–4.8 µM in cell free assays (Bannwarth et al. 2006). Other variations on the theme of interface peptides include combining the advantages of lipopeptides and molecular tongs. Interface peptides have been linked to lipophilic groups by a rigid bicyclic guanidinium scaffold (Breccia et al. 2003). The most potent compound demonstrates PR inhibitory activity similar to tethered peptides and molecular tongs. Cross-linked interfacial peptides have been designed to irreversibly inhibit HIV-1 PR by formation of a disulfi de bond between the peptide and the conserved PR residues C-95 and C-67, and demonstrate a Ki in the low micro- molar range (Zutshi and Chmielewski, 2000). The C-terminal tetrapeptide has also been tethered to a peptidic PR active site inhibitor, combining both dissociative and competitive methods of inhibition in one molecule (Uhlikova et al. 1996). Random peptides that are dissociative inhibitors of HIV-1 PR have been described. The bacterio- phage lambda repressor protein was utilised to develop a powerful two-hybrid PR dimerization assay. From a library of 5 × 108 random peptides, 300 were identifi ed as potential PR dimerization inhibitors. The most potent peptide identifi ed, p52, was a pure dissociative inhibitor with low Ki of 780 nM (Park and Raines, 2000). Ultimately, one of the major hurdles in devel- oping peptidic inhibitors is to obtain a biologically stable compound that can be delivered inside the cell. One mechanism to achieve this is to fuse the peptide to amino acid sequences that promote either encapsidation into viral particles or entry into the host cell. Virus protein R (Vpr) is a HIV-1 accessory protein packaged into virions by its trans association with the Gag p6 motif. Both viral and cellular proteins have been successfully delivered into viral particles as Vpr fusion proteins (Wu et al. 1997). Inhibition of HIV-1 replication has been reported by the fusion of Vpr to viral PR recognition sequences (Serio et al. 2000). Expression of the PR C-terminal tetrapep- tide as a Vpr fusion [Vpr-(spacer)-T-L-N-F-OH] attenuates HIV-1 replication in chronically infected cells and in single-round replication assays (Cartas et al. 2001). Most recently, inhibition of HIV-1 replication has been demonstrated by delivering PR interface peptides as a fusion peptide utilising the HIV-1 Tat derived cell permeable protein transduction domain Figure 2. A molecular tong bound to the C-terminus of the HIV-1 PR monomer. 169 Targeting the late stages of HIV-1 replication Drug Target Insights 2007: 2 (Davis et al. 2006). Peptides P27/A are PR dimerization inhibitors that inhibit the activity of wild-type and drug resistant PR in cell free assays with IC50 values in the 0.28–0.58 µM range. These peptides are successfully delivered into chronically HIV-1 infected cells and reduce viral particle production. This was observed by a reduction in p24, rather than inhibition of Gag processing which suggests that the peptide may interact with the Gag-Pol embedded PR and disrupt the ordered processing of Gag-Pol leading a decrease in viral particle production (Davis et al. 2006). PR Folding Inhibitors Local elementary structures (LES) are comprised of strongly interacting, highly conserved amino acids that are usually hydrophobic. These amino acids are suggested to direct the folding of a protein into its native conformation. Short peptides corre- sponding to or mimicking the LES are hypothesised to act as folding inhibitors, preventing the protein achieving its native conformation (Broglia et al. 2005). A peptide has been identifi ed from a LES in the HIV-1 PR (peptide S8, amino acids 83–93) that inhibits PR activity with a Ki of 2.58 µM and results in disorganisation of the PR secondary structure by reducing the β-sheet content from 30% to 14% (Broglia et al. 2005; Broglia et al. 2006). Current efforts are directed towards developing a shorter less hydrophobic peptide or mimetic based on the S8 lead peptide. Catalytically Inactive PR Subunits as Dominant Negative Inhibitors of PR activity Catalytically inactive PR monomers act in a dominant negative fashion to inhibit wild-type HIV-1 PR by forming inactive heterodimers in recombinant protein assays (Babe and Craik, 1991). When virus expressing a PR active site mutation is co-transfected with wild-type HIV-1, both viral replication and virus infectivity are reduced (Babe et al. 1995). Computer modelling has been used to successfully design an optimised dominant-negative PR expressing D25K, G49W and I50W (KWW) (McPhee et al. 1996), which also reduces viral replication and infectivity (Junker et al. 1996). Biochemical studies on recom- binant dominant negative PRs confi rm that the mechanism of action is by formation of inactive heterodimers (Rozzelle et al. 2000). Interestingly, the mutant PRs cannot homodimerize and they fold only when expressed with wild-type PR. PR heterodimers are also more stable that the wild-type homodimer (Rozzelle et al. 2000). Hence inactive PR heterodimers form the dominant species. Such a dominant negative strategy for the inhibition of HIV-1 PR would require in vivo delivery by a gene- therapy system, the therapeutic use of which is unlikely in the near future. Non-Peptide Inhibitors of HIV-1 PR A screen of a crude extract from the marine organism magenta ascidian didemnum identifi ed two didemnaketals, A (a bicyclic ketal) and B (a linear heptaprenoid), that inhibit HIV-1 PR activity with IC50 values of 2 µM and 10 µM, respectively (Potts et al. 1991). These compounds are unsuitable drug candidates, but have given rise to a novel class of pentaesters, the most potent of which is a dissociative inhibitor of PR with a Ki of 2.1 µM (Fan et al. 1998). A novel class of PR dimerization inhibitors were identifi ed by searching the Cambridge structural database for pharmacophores that mimic the action of previously identified inhibitory interface peptides (Quere et al. 1996). Several triterpene structures were identifi ed, of which ursolic acid acts as a dissociative inhibitor of PR with an IC50 of 2 µM. It has been suggested that triterpene could provide another basic scaffold for building more effective peptidomimetics. Interestingly, another member of the triterpene family, PA-457, acts as a novel inhibitor of HIV-1 maturation which is discussed later in this review. A β-sheet mimetic was tested for its ability to inhibit PR homodimer- ization by perturbation of β-sheet formation (Song et al. 2001). The β-sheet mimetic had a relatively high IC50 of 30 µM and the method of inhibition appears to be complex, however the structure provides a non-peptidic lead compound for PR inhibitors. Targeting Gag Processing The rate and the specifi city of Gag cleavage by the HIV-1 PR is dependent on the amino acid composition of the different cleavage sites recognized by the viral PR (Swanstrom, 1997). Based on the order of proteolysis by HIV-1 PR, these sites are classified as primary (p2/NC), secondary (MA/CA and p1/p6) or tertiary (CA/p2 170 Wapling et al Drug Target Insights 2007: 2 or CA/SP2) cleavage sites. The lack of processing of any of these sites by PR results in the formation of aberrant particles (Swanstrom, 1997). In particular, inhibition of cleavage at the CA/p2 site has severe consequences for core formation, stability and virion infectivity (Pettit et al. 1994; Wiegers et al. 1998; Pettit et al. 1998; Shehu- Xhilaga et al. 2001b). The α-helical structure that stretches between the C-terminus of CA and the N-terminus of SP1 is critical for virion assembly and p2 function (Accola et al. 1998). Clearly, these PR cleavage sites are potential targets for antiretroviral drug design. In this regard, a compound that interferes with viral maturation by blocking CA/p2 cleavage has been identifi ed. 3-O-(3′,3′-Dimethysuccinyl) betulinic acid (PA-457 or bevirimat) potently inhibits HIV-1 maturation and infectivity (Li et al. 2003; Zhou et al. 2004). PA-457 specifi cally blocks the cleavage of CA/p2 in cell based (Li et al. 2003) and in cell free assays (Zhou et al. 2005; Sakalian et al. 2006), thus inhibiting core condensation and virion matura- tion. Inhibition of Gag processing at the CA/p2 junction results in the generation of the uncleaved p25 product in transfected cells at 0.1 µg/ml of PA-457 (Li et al. 2003). Consistent with the proposed mechanism, PA-457 resistant HIV-1 selected in long term cultures in the presence of betulinic acid contain mutations in the regions that fl ank the P-P’ scissile bond (Adamson et al. 2006; Zhou et al. 2006). These mutated sites in Gag are recognized by the viral PR during proteolysis. In addition, other single amino acid substitutions have been identifi ed that confer resistance to PA-457 and are exclusively located either at the C terminus of CA or within the fi rst three amino acids of the p2 spacer peptide (Adamson et al. 2006). Interestingly, they all conferred resistance independently and were located within the boundaries of the CA/p2 proteins, a region well known to promote Gag multimerization (Adamson et al. 2006). These data suggest that there is more than one mechanism by which these mutants have acquired resistance to PA-457. PA-457 has successfully undergone Phase 1 and 2 clinical trials and is currently in a Phase 2b trial to test the effi cacy of different doses of PA-457 in combination with approved HIV-1 inhibitors as part of an optimised regimen in patients failing therapy due to the emergence of drug resistant virus. Targeting the RT Domain in Gag-Pol Like other HIV-1 enzymes, RT subunits must oligomerize to form an active enzyme. The biologically relevant form that is present in the virion is an asymmetric heterodimer comprised of the p66 (66 kDa) and the p51 (51 kDa) subunits (Jacobo-Molina et al. 1993; Kohlstaedt et al. 1992). The RT heterodimer is extremely stable and has an extensive protein surface area (4800 Å2) that is buried upon subunit dimerization. Ther- modynamic measurements of the association between the p66 and p51 RT subunits have esti- mated Gibbs free energy of dimer stabilization of approximately 10–12 kcal/mol–1, corresponding to a dissociation constant of approximately 3 × 10–7 M (Venezia et al. 2006). For an extensive review on HIV-1 RT dimerization see Srivastava et al. 2006. Regions both upstream and downstream of the PR region in Gag-Pol have been investigated for effects on PR activation (Bukovsky and Gottlinger, 1996; Partin et al. 1991; Louis et al. 1999). Large deletions within or C-terminal truncations of RT in the context of Gag-Pol result in an increase in virion associated Gag processing intermediates, suggesting a defect in PR activity (Cherry et al. 1998; Liao and Wang, 2004; Quillent et al. 1996). These studies suggest that modulating RT dimer- ization in the context of Gag-Pol may have a negative impact on PR activation and HIV-1 maturation. The importance of the RT region in Gag-Pol for both RT maturation and viral particle produc- tion has been demonstrated by the study of RT point mutations that prevent RT heterodimeriza- tion and p66 homodimerization. Mutations at W401, a component of the highly conserved tryptophan repeat motif in the connection subdo- main, blocks RT dimerization in vitro (Tachedjian et al. 2003; Tachedjian et al. 2005b). When expressed in HIV-1 it manifests as defects in reverse transcription, aberrant processing of RT, and low levels of infectivity (Wapling et al. 2005). The L234A mutation, located in the RT primer grip region, prevents RT dimerization and decreases Gag-Pol stability (Tachedjian et al. 2000; Yu et al. 1998). This mutation reduces PR incorporation into virions, increases the accumu- lation of Gag processing intermediates, and results in the production of non-infectious virus particles (Tachedjian et al. 2000; Yu et al. 1998). These examples demonstrate the potential for 171 Targeting the late stages of HIV-1 replication Drug Target Insights 2007: 2 targeting the Gag-Pol embedded RT for blocking HIV-1 maturation. Inhibitors that Modulate HIV-1 RT Subunit Interaction Apart from classical NNRTIs, there exists a class of unconventional NNRTIs that bind to HIV-1 RT and inhibit enzyme activity by decreasing the overall stability of the heterodimer without dissociating the complex (Sluis-Cremer et al. 2000; Sluis-Cremer and Tachedjian, 2002; Sluis-Cremer et al. 2006; Camarasa et al. 2006). The TSAO-T derivatives ([2′,5′-bis-O- (tert-butyldimethylsilyl)-β-D-ribofuranosyl]-3′- spiro-5″-(4″-amino-1″,2″-oxathiole-2″,2″-dioxide) thymine) destabilise both RT heterodimers and p66 homodimers by inducing changes at the RT dimer interface (Sluis-Cremer et al. 2000; Rodriguez- Barrios et al. 2001). The putative binding site is at the RT dimer interface and overlaps in part with the NNRTI binding pocket (Rodriguez-Barrios et al. 2001). Resistance mutations to the TSAO drugs are readily generated in cell culture indicating that these drugs are specific inhibitors of the HIV-1 RT (Balzarini et al. 1993). While TSAO represent the fi rst class of small molecules that destabilize the RT heterodimer, preclinical studies demonstrate that the pharmacological profile of TSAO inhibitors is unfavourable for further clinical development (Camarasa et al. 2006). The N-acylhydrazone derivative N-(4-tert- Butylbenzoyl)-2-hydroxy-1-naphthaldehyde hydrazone (BBNH) binds to both the DNA poly- merase and RNase H domains of RT and inhibits both enzymatic activities of the RT (Arion et al. 2002). Similar to TSAO, BBNH prevents RT activity through destabilizing, but not dissociating the subunits. BBNH derivatives that bind to the DNA polymerase domain alone are suffi cient to induce dimer destabilization (Sluis-Cremer and Tachedjian, 2002). The recently resolved structure of HIV-1 RT bound to a BBNH derivative has confi rmed that the binding site is in close proximity to, but distinct from both the polymerase active site and NNIBP. It is thought that BBNH destabi- lizes the RT heterodimer by inducing changes in the primer-grip motif, which is an important region for RT dimer stability (Himmel et al. 2006; Srivastava et al. 2006). By targeting the RT dimerization interface, it is possible that unconventional NNRTIs may have an affect on the late stage of virus replication. In particular, the TSAO drugs that destabilize the p66 homodimer, may also perturb the process of RT maturation to the heterodimer, and arguably even target the RT domain in Gag-Pol and interfere with PR activation. However, these possible effects have not been described. Further elucidation of the impact of nonclassical NNRTIs on RT maturation, and the mechanism of destabilization would be advantageous for designing more potent inhibitors of both RT function and RT maturation. Classical NNRTIs as RT Inhibitors Acting at the Late Stage of Viral Replication Interestingly, several classical NNRTIs have been shown to confer a concentration dependant increase in RT heterodimer formation, corre- sponding with a loss of RT polymerase function (Tachedjian et al. 2001; Venezia et al. 2006). Efavi- renz (EFV) is a strong enhancer of RT dimeriza- tion, and also enhances the formation of p66 and p51 homodimers (Tachedjian et al. 2005a). The exact mechanism for increasing RT subunit inter- actions is unknown but it is suspected that the binding of EFV to the RT mediates conformational changes in the p66 subunit that promotes interac- tion with p51 (Tachedjian et al. 2001). EFV has also demonstrated the capacity to enhance the homodimerization of p66 in vitro, and a 90kDa model Pol protein in an inducible bacterial expres- sion system (Sluis-Cremer et al. 2004; Tachedjian et al. 2005a). It has recently been demonstrated that NNRTIs enhance Gag-Pol dimerization, resulting in premature PR activation and a decrease in viral particle release (Figueiredo et al. 2006). In HIV-1 transfected cells, EFV, TMC120 and TMC125 increased Gag and Gag-Pol processing, and caused up to 45% decrease in viral particle production. Similar effects were not observed for NNRTIs that do not signifi cantly enhance p66 homodimerization and NRTIs (Figueiredo et al. 2006). Hence, NNRTIs that are potent enhancers of RT dimerization also affect the late stage of viral replication, which represents a novel inhibitory mechanism for these drugs. However, the concen- trations required to mediate this effect are two to three orders of magnitude higher then concentra- tions that block RT function. This is likely due to reduced binding affi nity of the NNRTIs to the proposed target which is the RT domain of Gag-Pol. Strategies to identify drugs that are more potent 172 Wapling et al Drug Target Insights 2007: 2 inhibitors of this late stage in the viral life-cycle could be identifi ed by screening for molecules that enhance Gag-Pol dimerization. Such a screen could be enhanced by incorporating mutations in the RT that are known to confer decreased susceptibility to current NNRTIs in order to select for drugs that have the potential to block NNRTI resistant strains of HIV-1. Peptide Based Inhibitors of RT dimerization Two strategies have been utilised to generate peptides designed to target the RT dimer interface in order to block HIV-1 RT function, including peptides corre- sponding to regions that are known to have an impor- tant role in RT dimerization (Debyser and De Clercq, 1996; Depollier et al. 2005; Divita et al. 1994; Morris et al. 1999b; Morris et al. 1999a). The most successful of these peptides, Pep-7, corresponds to RT residues 395–404, derived from the highly conserved trypto- phan repeat motif (W398–W414) (Depollier et al. 2005). Pep-7 interacts with p51, and destablizes both the RT heterodimer and the p66 homodimer. Similar to unconventional NNRTIs, Pep-7 is unable to induce RT dissociation (Depollier et al. 2005). Pep-7 based peptides are potent suppressors of HIV-1 replication at noncytotoxic concentrations (Morris et al. 1999b). The method of inhibition in HIV-1 infected cells has not been elucidated. However, given that Pep-7 cannot induce RT subunit dissociation, it has been suggested that it acts at the late stage of virus replication by preventing the formation of an active RT heterodimer (Morris et al. 1999b). Rational strategies utilising the available RT structures to direct the design and manufacture of mimetic peptides targeting subunit interaction is a recent development (Campbell et al. 2002; Hosokawa et al. 2004). These studies have led to the synthesis of a peptide, TLMA2993, which also targets the RT connection subdomain. TLMA2993 inhibits RT activity at micromolar concentrations (Campbell et al. 2002). Cells stably transfected with this peptide are protected from HIV-1 infection in a concentration dependant manner due to inhibition of reverse transcription, as observed by a decrease in HIV-1 DNA (Hosokawa et al. 2004). Targeting the IN Domain in Gag-Pol IN catalyses the insertion of viral DNA into the host chromosome and thus inhibits an early crucial step in the virus life cycle. IN is also implicated in reverse transcription, nuclear import of the pre-integration complex, viral assembly and budding (Engelman et al. 1995; Hehl et al. 2004). Despite the numerous roles of IN in HIV-1 replica- tion, new approaches for inhibiting viral replication have focused on targeting the catalytic activity of IN that is required for proviral DNA integration. Two IN inhibitors, MK-0518 and GS-9137 (JTK-303) have entered clinical trials (Cotelle, 2006; Makhija, 2006) and have shown effi cacy in Phase III clinical trials (Stephenson, 2007). Since IN is expressed as part of Gag-Pol, agents that bind to the IN domain in this polypro- tein are likely to impact on the late stages of replication. Consistent with this notion, mutations in IN have been reported to effect virion formation (Shin et al. 1994). Truncations of IN at the C-terminus of Gag-Pol result in aberrant virion core structures, with a reduction in the overall levels of cell-associated viral Gag, suggesting a defect in Gag-Pol processing (Engelman et al. 1995; Bukovsky and Gottlinger, 1996). IN requires oligomerization for activity. Therefore, inhibitors of IN function that mediate their effects through negating IN subunit interactions are also likely to interfere with viral assembly. This would be manifested by interfering with Gag-Pol/Gag-Pol interactions leading to subsequent effects on HIV-1 PR activation (Muriaux et al. 2004). In this regard peptide inhibitors of IN dimerization have been reported, however their effects on the late stages of the virus life-cycle remains to be determined (Maroun et al. 2001; Zhao et al. 2003). Certain host cell factors are incorporated into the virion by interaction with the IN domain of Gag-Pol. A cellular factor that has been implicated in affecting the late stage of the virus life cycle is integrase interactor 1 (INI1). INI1 was identifi ed in a yeast two-hybrid screen for host cell proteins interacting with HIV-1 IN (Yung et al. 2001;Yung et al. 2004; Kalpana et al. 1994). INI1 mutants that abrogate interaction with IN or cells defi cient in INI1 exhibit a substantial reduction in viral produc- tion (Yung et al. 2001). INI1 affects several steps during HIV-1 replication (Ariumi et al. 2006; Sorin et al. 2006; Yung et al. 2001) and is also packaged into HIV-1 particles (Kalpana et al. 1994). A 110- amino-acid fragment of INI1 (S6) with a minimal IN-interaction domain inhibits viral production (Yung et al. 2001; Yung et al. 2004). The inhibitory effect of S6 on HIV-1 production is mediated by 173 Targeting the late stages of HIV-1 replication Drug Target Insights 2007: 2 binding of the ectopically expressed S6 to the Gag-Pol embedded IN. Furthermore, stable expression of a transdominant S6 mutant inhibits infection in T-cells. S6 represents a potential lead for the development of inhibitors of the late stage of HIV-1 replication (Yung et al. 2001). Proteosome Inhibitors Intracellular degradation of misfolded, damaged or unwanted proteins is mediated by the proteo- some, which is a multisubunit proteolytic complex of 26S (Schubert et al. 2000). Proteins are tagged for proteolytic destruction by the covalent attach- ment of a chain of ubiquitin polypeptides on lysine residues of the protein (Schubert et al. 2000). Proteosome inhibitors inhibit the late stages of the HIV-1 life-cycle by interfering with viral particle release and maturation (Schubert et al. 2000). Decreased budding has also been demonstrated for retroviruses expressing the PPPY- or PTAP containing late domains but not those that use the YPDL type late domain (Schubert et al. 2000; Ott et al. 2003). The effect is not dependent on the viral particle assembly site (i.e. cytoplasm or plasma membrane) or on monoubiquitination of Gag (Ott et al. 2003). In addition to a decrease in viral particle release (4-fold), virions released from cells treated with proteosome inhibitors have approxi- mately a 10-fold decrease in infectivity (Schubert et al. 2000). The impact of proteosome inhibitors is dependent on an active HIV-1 PR and the pres- ence of the p6 late domain but is independent of Vpu function. Inhibition of HIV-1 maturation and budding is observed with reversible (zLLL also known as MG-132) and irreversible (lactocystin) proteosome inhibitors (Schubert et al. 2000). Proteosome inhibitors also interfere with the activity of the HIV-1 viral infectivity factor (Vif) on the antiviral function of apolipoprotein B mRNA- editing enzyme catalytic polypeptide-like 3G (APOBEC3G) in virus producing cells (Stopak et al. 2003; Sheehy et al. 2003; Mehle et al. 2004; Yu et al. 2003). Wild-type viruses expressing Vif are able to prevent incorporation of APOBEC3G into the virion by promoting its degradation in the cytoplasm of the producer cell. Inhibition of APOBEC3G incor- poration in the virus prevents hypermutation in newly synthesized viral DNA following infection of target cells due to C to U modifi cations during minus stand DNA synthesis mediated by APOBEC3G. Proteosome inhibitors interfere with Vif dependent degradation of APOBEC3G suggesting that these inhibitors can impede the mechanisms used by the virus to evade the innate defences of the host cell (Stopak et al. 2003; Sheehy et al. 2003; Mehle et al. 2004; Yu et al. 2003). Targeting an essential cellular process like the proteosome is anticipated to be cytotoxic and not well tolerated in vivo. Nevertheless, the highly specifi c proteosome inhibitor epoxomicin, which also inhibits HIV-1 maturation, is well tolerated in mice (Meng et al. 1999). The proteosome inhibitor, PS-341, is approved as a last resort treatment of multiple myelomas and is associated with adverse effects (Kane et al. 2006). The use of proteosome inhibitors in HIV-1 infected individuals needs to be considered in the context of the potential risk benefi t and the net effect on inhibition of HIV-1 replication as proteosome inhibitors also enhance the early step of the virus life-cycle by preventing degradation of the reverse transcription complex mediated by TRIM5α (Schwartz et al. 1998; Wu et al. 2006; Wei et al. 2005). Targeting HIV-1 Egress Mediated by Vpu The HIV-1 accessory protein, Vpu is a 16 kDa type 1 integral membrane protein that is indispensable for viral pathogenesis (Li et al. 2005). Vpu plays two distinctive roles in the viral life-cycle that include down regulating host cell CD4 receptors (Willey et al. 1992) and enhancing viral particle release from the cell surface, the latter associated with its ion channel forming properties (Schubert et al. 1996a). Vpu is unique to HIV-1/SIVcpz viruses (Binette and Cohen, 2004). Interestingly the two closely related retroviruses HIV-2 and SIV, which lack Vpu, are less pathogenic (Bour and Strebel, 2003). The role of Vpu in the viral budding process is coupled to its ion channel forming properties, which is predicted to be a pentameric structure composed of fi ve transmembrane domains (Grice et al. 1997). The Vpu ion channel is thought to function by altering the electric potential at the plasma membrane or alternatively by overcoming host restriction factors for viral release (Neil et al. 2006). The HIV-1 Vpu is a member of viral ion channel proteins called viroporins and is structur- ally similar to the M2 ion channel protein of infl uenza (Gonzalez and Carrasco, 2003; Hout et al. 2006). An interesting feature of Vpu is its role in 174 Wapling et al Drug Target Insights 2007: 2 viral particle release from nondividing cells such as macrophages (Deora and Ratner, 2001). In this regard the rate of host cell proliferation is a deter- mining factor for Vpu mediated viral particle release (Deora and Ratner, 2001). Analogues of amiloride (a sodium channel blocker) inhibit Vpu ion channel activity. The amiloride analogues, 5-(N,N-hexamethylene) amiloride (HMA) and 5-(N,N-dimethyl)amiloride (DMA) inhibit Vpu mediated virus budding and viral replication in macrophages (Ewart et al. 2002; Ewart et al. 2004). The inhibitory effects are observed in the absence of cytotoxicity. Both analogs exhibit strong inhibition of HIV-1 replica- tion as measured by viral p24 levels in culture supernatants (Ewart et al. 2004). HMA at 4 µM suppresses viral p24 in culture supernatants to unde- tectable levels for more than 10 days in culture (Ewart et al. 2004). While amiloride analogues demonstrate activity in macrophages, they fail to inhibit HIV-1 replication in T-cells (Ewart et al. 2002). Nevertheless, Vpu ion channel inhibitors have the potential for use in combination therapy, targeting viral reservoirs and drug-resistant variants. An amiloride derivative, BIT225, is currently being pursued for drug development (Biotron Limited, Sydney, NSW, Australia). BIT225 represents a promising antiretroviral therapeutic although the evaluation of its in vivo effi cacy will present a challenge since inhibition of HIV-1 replication appears to be restricted to nonproliferating cells. Recent studies also suggest that rimantadine, an ion channel blocker of infl uenza A viruses, can be a useful lead compound for designing Vpu inhibitors. Rimantadine and amantadine belong to class of polycyclic amines that are active against the M2 ion channel of infl uenza A but not against HIV-1 Vpu (Hout et al. 2006). Studies indicate that mutating histidine at residue 19 to alanine results in a rimantadine sensitive Vpu ion channel demon- strating the potential of this class of inhibitor as HIV-1 ion channel blockers (Hout et al. 2006). Vpu is also implicated to interact with certain host cell restriction factors that interfere with viral particle egress. The host cell protein, Vpu-binding protein (UBP), is suggested to be a negative factor for virus assembly (Callahan et al. 1998; Bour and Strebel, 2003). UBP is a 34-kDa protein that exhibits competitive binding with Vpu and Gag (Callahan et al. 1998; Handley et al. 2001). Overexpression of UBP has been reported to significantly suppress viral particle release suggesting that UBP is a negative factor that requires displacement by Gag or Vpu (Callahan et al. 1998). It is suggested that Vpu mediates its effect on viral egress by facilitating membrane targeting of Gag precursors (Handley et al. 2001; Harila et al. 2006; Neil et al. 2006). Supporting this notion, Vpu-defective particles appear in internal membrane-bound compartments suggesting a Gag targeting defect (Klimkait et al. 1990). Another host cell restriction factor implicated in viral release is an acid-sensitive potassium channel-forming protein, TASK-1, which is down regulated during viral infection (Hsu et al. 2004). Due to the structural homology of TASK-1 and HIV-1 Vpu, TASK-1 has been suggested to form hetero-oligomers with Vpu which interferes with both TASK-1 mediated conductance and Vpu ion channel function (Hsu et al. 2004). Further delinea- tion of the how these host cell factors interact with Vpu is required in order to design small molecule inhibitors that inhibit viral particle release. Targeting HIV-1 Egress Mediated by the p6 Late Domain In the recent years, major advances have been made in our understanding of how HIV-1 and other retroviruses are released from infected cells. In the early 1990s it was reported that mutations in the p6 late domain inhibit virion particle release (Gottlinger et al. 1991). Moreover, a PTAP sequence, encompassing amino acids 7 to 10 of p6, is critical for virus particle production (Huang et al. 1995). The PTAP motif binds specifi cally to the host cell protein, tumor suppressor gene 101 (TSG101), resulting in the recruitment of components of the endosomal sorting complex required for transport-I (ESCRT-I) (VerPlank et al. 2001; Martin-Serrano et al. 2001; Garrus et al. 2001; Demirov et al. 2002). Deletion of the PTAP motif results in approximately 80% reduction in HIV-1 particle release. Similarly, overexpression and silencing of TSG101 abolish viral egress (Garrus et al. 2001; Demirov et al. 2002; Goila-Gaur et al. 2003). NMR structure analysis of a 14 amino acid peptide derived from p6 encompassing the PTAP motif complexed with the UEV domain of TSG101 has revealed that the PTAP motif binds to a groove in TSG101. This binding creates two main pockets: the “A-P” pocket through contact of amino acids 7–10 and the “P” pocket through the binding of amino acids 9–10 of the peptide (Pornillos et al. 175 Targeting the late stages of HIV-1 replication Drug Target Insights 2007: 2 2002a; Pornillos et al. 2002b). Interruption of this viral host protein-protein interaction with compounds that bind to this pocket in TSG101 and compete with Gag would potentially inhibit viral particle release and the rate of cell-cell HIV-1 transmission (Bieniasz, 2006). So far there has been one report that describes the synthesis and selection of small molecules with up to fi ve fold better binding capacity than a peptide that contains the sequence of the wild type PTAP motif in the L domain of HIV-1 (Liu et al. 2006). In this study, the authors describe an approach previously employed to obtain peptoids in which a key proline residue is substituted with glycine at the N-terminus of the parent Pro-rich sequence in order to improve binding specifi city to Src homology 3 (SH3) domains (Nguyen et al. 2000). Similarly, the proline rich PTAP domain in p6 was considered a good candidate for the N-substituted glycine residue approach. In this study, the Tsg101 binding affi nity (Kd) of the 9-mer wild-type PTAP containing peptide was 50 µM (Liu et al. 2006). Binding constants for the highest affi nity peptoid- hydrazones (designated 11q and 11p) were 17.5 and 9.8 µM, respectively. The highest affi nity peptoid hydrazone was found to be the n-butyl-containing 11p peptide, with a fi ve-fold increase in binding affi nity compared to the wild-type 9-mer PTAP peptide. The capacity of these peptides to compete with HIV-1 Gag for TSG101 binding and their effect on HIV-1 egress in vitro and in vivo remains to be determined. Although disruption of viral-host cell interac- tions is a very attractive approach to abolish virion release and infectivity, interfering with the host cell machinery could have major consequences for the host (Bieniasz, 2006). TSG101 is a multifunctional protein and plays a critical role in cell proliferation as shown by studies conducted in TSG101 defi cient mice (Ruland et al. 2001). TSG101 is involved in cellular transcription and plays a central role in endosomal sorting of cargo protein that is destined for degradation by the proteasome. TSG101 is recruited to the endosomal sorting pathway by a specifi c interaction with the host cell protein, hepa- tocyte growth factor-regulated tyrosine kinase substrate (Hrs), which binds to TSG101 through a PSAP motif (Lu et al. 2003). Disruption of this interaction inhibits delivery of epidermal growth factor receptor (EGFR) to the late endosomes. In recruiting TSG101, it is believed that HIV-1 mimics Hrs in order to enter the endosomal sorting pathway and negotiate its release from the infected cell (Pornillos et al. 2003). Thus, targeting TSG101 would result in the accumulation of proteins at the plasma membrane and the disruption of protein sorting within the infected cell. Specifi c inhibition of HIV-1 budding by targeting the late domain binding site on TSG101 will require preferential inhibition of p6 binding compared to Hrs. Conclusion Considerable progress has been made in under- standing the late steps of the viral life-cycle leading to the production of infectious viral particles. Many of these processes rely on protein:protein interac- tions either between viral proteins or viral proteins and host cell factors. The interactions between the host and viral proteins have either a role in facili- tating virus replication, as is observed for the viral p6 and Tsg101, or are necessary to overcome nega- tive effects of the host on virus replication, as mediated by Vif. HIV-1 also relies on posttransla- tional modifi cations mediated by the host cell machinery in order for viral polyproteins to be traffi cked to the appropriate compartment of the cell for viral assembly and budding. Arguably, one of the most effective drugs used to treat HIV-1 infected individuals are the HIV-1 PR inhibitors that block viral maturation. This underscores the effectiveness of targeting the late stage of virus replication. Nevertheless, while some of the strategies described in this review inhibit virus replication in cell culture assays, they require improvements in potency, specifi city and delivery before being viable for use in the clinic. Acknowledgements J.W. is supported by an Australian Postgraduate Award, S.S. and M. S-X are supported by funding from the Australian Centre for HIV and Hepatitis Virology Research and G.T. is supported by funding from NHMRC R.D. Wright Biomedical Career Development grant 235102 and NHMRC Project Grant 381703. Johanna Wapling and Seema Srivastava made equal contribution to the review. References Abdel-Rahman, H.M., Al-Karamany, G.S., El-Koussi, N.A., Youssef, A.F. and Kiso, Y. 2002. HIV protease inhibitors: peptidomimetic drugs and future perspectives. Curr. Med. Chem., 9:1905–1922. Accola, M.A., Hoglund, S. and Gottlinger, H.G. 1998. A putative alpha- helical structure which overlaps the capsid-p2 boundary in the human immunodefi ciency virus type 1 Gag precursor is crucial for viral particle assembly. J. Virol., 72:2072–2078. 176 Wapling et al Drug Target Insights 2007: 2 Adamson, C.S., Ablan, S.D., Boeras, I., Goila-Gaur, R., Soheilian, F., Nagashima, K., Li, F., Salzwedel, K., Sakalian, M., Wild, C.T. and Freed, E.O. 2006. In Vitro Resistance to the Human Immunodefi - ciency Virus Type 1 Maturation Inhibitor PA-457 (Bevirimat). J. Virol., 80:10957–10971. Aldovini, A. and Young, R.A. 1990. Mutations of RNA and protein sequences involved in human immunodeficiency virus type 1 packaging result in production of noninfectious virus. J. Virol., 64:1920–1926. Andersson, E., Horal, P., Jejcic, A., Hoglund, S., Balzarini, J., Vahlne, A. and Svennerholm, B. 2005. Glycine-amide is an active metabolite of the antiretroviral tripeptide glycyl-prolyl-glycine-amide. Antimicrob. Agents Chemother., 49:40–44. Andersson, E., Horal, P., Vahlne, A. and Svennerholm, B. 2004. No cross- resistance or selection of HIV-1 resistant mutants in vitro to the antiretro- viral tripeptide glycyl-prolyl-glycine-amide. Antiviral Res., 61:119–124. Arion, D., Sluis-Cremer, N., Min, K.L., Abram, M.E., Fletcher, R.S. and Parniak, M.A. 2002. Mutational analysis of Tyr-501 of HIV-1 reverse transcriptase. Effects on ribonuclease H activity and inhibition of this activity by N-acylhydrazones. J. Biol. Chem., 277:1370–1374. Ariumi, Y., Serhan, F., Turelli, P., Telenti, A. and Trono, D. 2006. The inte- grase interactor 1 (INI1) proteins facilitate Tat-mediated human immunodefi ciency virus type 1 transcription. Retrovirology, 3:47. Babe, L.M. and Craik, C.S. 1991. Time dependent heterodimer formation leads to inhibition of HIV protease activity. Adv. Exp. Med. Biol., 306:543–547. Babe, L.M., Pichuantes, S. and Craik, C.S. 1991. Inhibition of HIV protease activity by heterodimer formation. Biochemistry, 30:106–111. Babe, L.M., Rose, J. and Craik, C.S. 1992. Synthetic “interface” peptides alter dimeric assembly of the HIV 1 and 2 proteases. Protein Sci., 1:1244–1253. Babe, L.M., Rose, J. and Craik, C.S. 1995. Trans-dominant inhibitory human immunodefi ciency virus type 1 protease monomers prevent protease activation and virion maturation. Proc. Natl. Acad. Sci. U.S.A., 92:10069–10073. Balzarini, J., Karlsson, A., Vandamme, A.M., Perez-Perez, M.J., Zhang, H., Vrang, L., Oberg, B., Backbro, K., Unge, T., San-Felix, A. et al. 1993. Human immunodefi ciency virus type 1 (HIV-1) strains selected for resistance against the HIV-1-specific [2′,5′-bis-O-(tert- butyldimethylsilyl)-3′-spiro-5″-(4″-amino-1″,2″-oxathiole- 2″,2″-dioxide)]-beta-D-pentofurano syl (TSAO) nucleoside analogues retain sensitivity to HIV-1-specifi c nonnucleoside inhibitors. Proc. Natl. Acad. Sci. U.S.A., 90:6952–6956. Bannwarth, L., Kessler, A., Pethe, S., Collinet, B., Merabet, N., Boggetto, N., Sicsic, S., Reboud-Ravaux, M. and Ongeri, S. 2006. Molecular tongs containing amino acid mimetic fragments: new inhibitors of wild- type and mutated HIV-1 protease dimerization. J. Med. Chem., 49:4657–4664. Bieniasz, P.D. 2006. Late budding domains and host proteins in enveloped virus release. Virology, 344:55–63. Binette, J. and Cohen, E.A. 2004. Recent advances in the understanding of HIV-1 Vpu accessory protein functions. Curr. Drug. Targets Immune Endocr. Metabol. Disord., 4:297–307. Bouamr, F., Scarlata, S. and Carter, C. 2003. Role of myristylation in HIV-1 Gag assembly. Biochemistry, 42:6408–6417. Bour, S. and Strebel, K. 2003. The HIV-1 Vpu protein: a multifunctional enhancer of viral particle release. Microbes Infect., 5:1029–1039. Bouras, A., Boggetto, N., Benatalah, Z., De Rosny, E., Sicsic, S. and Reboud- Ravaux, M. 1999. Design, synthesis, and evaluation of conformation- ally constrained tongs, new inhibitors of HIV-1 protease dimerization. J. Med. Chem., 42:957–962. Breccia, P., Boggetto, N., Perez-Fernandez, R., Van Gool, M., Takahashi, M., Rene, L., Prados, P., Badet, B., Reboud-Ravaux, M. and De Mendoza, J. 2003. Dimerization inhibitors of HIV-1 protease based on a bicyclic guanidinium subunit. J. Med. Chem., 46:5196–5207. Brierley, I. and Dos Ramos, F.J. 2006. Programmed ribosomal frameshift- ing in HIV-1 and the SARS-CoV. Virus Res., 119:29–42. Broglia, R.A., Provasi, D., Vasile, F., Ottolina, G., Longhi, R. and Tiana, G. 2006. A folding inhibitor of the HIV-1 protease. Proteins, 62:928–933. Broglia, R.A., Tiana, G., Sutto, L., Provasi, D. and Simona, F. 2005. Design of HIV-1-PR inhibitors that do not create resistance: blocking the folding of single monomers. Protein Sci., 14:2668–2681. Bryant, M. Ratner, L. 1990. Myristoylation-dependent replication and as- sembly of human immunodefi ciency virus 1. Proc. Natl. Acad. Sci. U.S.A, 87:523–527. Bryant, M.L., Heuckeroth, R.O., Kimata, J.T., Ratner, L. and Gordon, J.I. 1989. Replication of human immunodefi ciency virus 1 and Moloney murine leukemia virus is inhibited by different heteroatom-containing analogs of myristic acid. Proc. Natl. Acad Sci. U.S.A., 86:8655– 8659. Bryant, M.L., Ratner, L., Duronio, R.J., Kishore, N.S., Devadas, B., Adams, S.P. and Gordon, J.I. 1991. Incorporation of 12-methoxydodecanoate into the human immunodefi ciency virus 1 gag polyprotein precursor inhibits its proteolytic processing and virus production in a chroni- cally infected human lymphoid cell line. Proc. Natl. Acad. Sci. U.S.A., 88:2055–2059. Bukovsky, A. and Gottlinger, H. 1996. Lack of integrase can markedly affect human immunodefi ciency virus type 1 particle production in the presence of an active viral protease. J. Virol., 70:6820–6825. Cafl isch, A., Schramm, H.J. and Karplus, M. 2000. Design of dimerization inhibitors of HIV-1 aspartic proteinase: a computer-based combina- torial approach. J. Comput. Aided Mol. Des., 14:161–179. Callahan, M.A., Handley, M.A., Lee, Y.H., Talbot, K.J., Harper, J.W. and Panganiban, A.T. 1998. Functional interaction of human immunode- fi ciency virus type 1 Vpu and Gag with a novel member of the tet- ratricopeptide repeat protein family. J. Virol., 72:5189–5197. Camarasa, M.J., Velazquez, S., San-Felix, A., Perez-Perez, M.J., Bonache, M.C. and De Castro, S. 2006. TSAO derivatives, inhibitors of HIV-1 reverse transcriptase dimerization: recent progress. Curr. Pharm. Des., 12:1895–1907. Campbell, S. and Vogt, V.M. 1995. Self-assembly in vitro of purifi ed CA-NC proteins from Rous sarcoma virus and human immunodefi ciency virus type 1. J. Virol., 69:6487–6497. Campbell, W., Kleiman, L., Barany, L., Li, Z., Khorchid, A., Fujita, E., Okada, N. and Okada, H. 2002. A novel genetic algorithm for design- ing mimetic peptides that interfere with the function of a target molecule. Microbiol. Immunol., 46:211–215. Cartas, M., Singh, S.P., Serio, D., Rizvi, T.A., Kalyanaraman, V.S., Goldsmith, C.S., Zaki, S.R., Weber, I.T. and Srinivasan, A. 2001. Intravirion display of a peptide corresponding to the dimer structure of protease attenuates HIV-1 replication. DNA Cell Biol., 20:797–805. Cen, S., Niu, M., Saadatmand, J., Guo, F., Huang, Y., Nabel, G.J. and Kleiman, L. 2004. Incorporation of pol into human immunodefi - ciency virus type 1 Gag virus-like particles occurs independently of the upstream Gag domain in Gag-pol. J. Virol., 78:1042–1049. Cheng, Y.S., Yin, F.H., Foundling, S., Blomstrom, D. and Kettner, C.A. 1990. Stability and activity of human immunodefi ciency virus pro- tease: comparison of the natural dimer with a homologous, single- chain tethered dimer. Proc. Natl. Acad. Sci. U.S.A., 87:9660–9664. Cherry, E., Liang, C., Rong, L., Quan, Y., Inouye, P., Li, X., Morin, N., Kotler, M. and Wainberg, M.A. 1998. Characterization of human immunodefi ciency virus type-1 (HIV-1) particles that express prote- ase-reverse transcriptase fusion proteins. J. Mol. Biol., 284: 43–56. Chien, A.I., Liao, W.H., Yang, D.M. and Wang, C.T. 2006. A domain di- rectly C-terminal to the major homology region of human immuno- defi ciency t-ype 1 capsid protein plays a crucial role in directing both virus assembly and incorporation of Gag-Pol. Virology, 348:84–95. Chiu, H.C., Yao, S.Y. and Wang, C.T. 2002. Coding sequences upstream of the human immunodefi ciency virus type 1 reverse transcriptase domain in Gag-Pol are not essential for incorporation of the Pr160(gag-pol) into virus particles. J. Virol., 76:3221–3231. Clavel, F. and Hance, A.J. 2004. HIV drug resistance. N. Engl. J. Med., 350: 1023–1035. 177 Targeting the late stages of HIV-1 replication Drug Target Insights 2007: 2 Cotelle, P. 2006. Patented HIV-1 integrase inhibitors (1998–2005). Recent Patents on Anti-Infective Drug Discovery, 1:1–15. Davis, D.A., Brown, C.A., Singer, K.E., Wang, V., Kaufman, J., Stahl, S.J., Wingfi eld, P., Maeda, K., Harada, S., Yoshimura, K., et al. 2006. Inhibition of HIV-1 replication by a peptide dimerization inhibitor of HIV-1 protease. Antiviral Res., 72:89–99. De Clercq, E. 1998. The role of non-nucleoside reverse transcriptase inhibitors (NNRTIs) in the therapy of HIV-1 infection. Antiviral Res., 38:153–179. Debyser, Z. and De Clercq, E. 1996. Chemical crosslinking of the subunits of HIV-1 reverse transcriptase. Protein Sci., 5:278–286. Demirov, D.G., Ono, A., Orenstein, J.M. and Freed, E.O. 2002. Overexpres- sion of the N-terminal domain of TSG101 inhibits HIV-1 budding by blocking late domain function. Proc. Natl. Acad. Sci. U.S.A., 99: 955–960. Deora, A. and Ratner, L. 2001. Viral protein U (Vpu)-mediated enhancement of human immunodefi ciency virus type 1 particle release depends on the rate of cellular proliferation. J. Virol., 75:6714–6718. Depollier, J., Hourdou, M.L., Aldrian-Herrada, G., Rothwell, P., Restle, T. and Divita, G. 2005. Insight into the mechanism of a peptide inhibi- tor of HIV reverse transcriptase dimerization. Biochemistry, 44: 1909–1918. Dettenhofer, M. and Yu, X.F. 1999. Proline residues in human immunode- fi ciency virus type 1 p6(Gag) exert a cell type-dependent effect on viral replication and virion incorporation of Pol proteins. J. Virol., 73:4696–4704. Devadas, B., Lu, T., Katoh, A., Kishore, N.S., Wade, A.C., Mehta, P.P., Rudnick, D.A., Bryant, M.L., Adams, S.P., Li, Q. et al. 1992. Substrate specifi city of Saccharomyces cerevisiae myristoyl-CoA: protein N-myristoyltransferase. Analysis of fatty acid analogs containing carbonyl groups, nitrogen heteroatoms, and nitrogen heterocycles in an in vitro enzyme assay and subsequent identifi cation of inhibitors of human immunodefi ciency virus I replication. J. Biol. Chem., 267: 7224–7239. Ding, L., Derdowski, A., Wang, J.J. and Spearman, P. 2003. Independent segregation of human immunodefi ciency virus type 1 Gag protein complexes and lipid rafts. J. Virol., 77:1916–1926. Divita, G., Restle, T., Goody, R.S., Chermann, J.C. and Baillon, J.G. 1994. Inhibition of human immunodefi ciency virus type 1 reverse transcrip- tase dimerization using synthetic peptides derived from the connec- tion domain. J. Biol. Chem., 269:13080–13083. Dorfman, T., Bukovsky, A., Ohagen, A., Hoglund, S. and Gottlinger, H.G. 1994. Functional domains of the capsid protein of human immuno- defi ciency virus type 1. J. Virol., 68:8180–8187. Dulude, D., Berchiche, Y.A., Gendron, K., Brakier-Gingras, L. and Heveker, N. 2006. Decreasing the frameshift effi ciency translates into an equivalent reduction of the replication of the human immuno- defi ciency virus type 1. Virology, 345:127–136. Dumond, J., Boggetto, N., Schramm, H.J., Schramm, W., Takahashi, M. and Reboud-Ravaux, M. 2003. Thyroxine-derivatives of lipopeptides: bifunctional dimerization inhibitors of human immunodefi ciency virus-1 protease. Biochem. Pharmacol., 65:1097–1102. Engelman, A., Englund, G., Orenstein, J.M., Martin, M.A. and Craigie, R. 1995. Multiple effects of mutations in human immunodefi ciency virus type 1 integrase on viral replication. J. Virol., 69:2729–2736. Ewart, G.D., Mills, K., Cox, G.B. and Gage, P.W. 2002. Amiloride deriva- tives block ion channel activity and enhancement of virus-like par- ticle budding caused by HIV-1 protein Vpu. Eur. Biophys. J., 31: 26–35. Ewart, G.D., Nasr, N., Naif, H., Cox, G.B., Cunningham, A.L. and Gage, P.W. 2004. Potential new anti-human immunodefi ciency virus type 1 compounds depress virus replication in cultured human macro- phages. Antimicrob. Agents Chemother., 48:2325–2330. Facke, M., Janetzko, A., Shoeman, R.L. and Krausslich, H.G. 1993. A large deletion in the matrix domain of the human immunodefi ciency virus gag gene redirects virus particle assembly from the plasma membrane to the endoplasmic reticulum. J. Virol., 67:4972–4980. Fan, X., Flentke, G.F. and Rich, D.H. 1998. Inhibition of HIV-1 protease by a subunit of didemnaketal A. Journal of the American Chemical Society, 120:8893–8894. Figueiredo, A., Moore, K.L., Mak, J., Sluis-Cremer, N., De Bethune, M.P. and Tachedjian, G. 2006. Potent Nonnucleoside Reverse Transcriptase Inhibitors Target HIV-1 Gag-Pol. PLoS Pathog., 2: e119. Franciskovich, J., Houseman, K., Meuller, R. and Chmielewski, J. 1993. A systematic evaluation of the inhibition of HIV-1 protease by its C- and N-terminal peptides. Bioorg. Med. Chem. Lett., 765–768. Freed, E.O. 1998. HIV-1 gag proteins: diverse functions in the virus life cycle. Virology, 251:1–15. Gamble, T.R., Vajdos, F.F., Yoo, S., Worthylake, D.K., Houseweart, M., Sundquist, W.I. and Hill, C.P. 1996. Crystal structure of human cyclophilin A bound to the amino-terminal domain of HIV-1 capsid. Cell, 87:1285–1294. Gamble, T.R., Yoo, S., Vajdos, F.F., Von Schwedler, U.K., Worthylake, D.K., Wang, H., Mccutcheon, J.P., Sundquist, W.I. and Hill, C.P. 1997. Structure of the carboxyl-terminal dimerization domain of the HIV- 1 capsid protein. Science, 278:849–853. Ganser-Pornillos, B.K., Von Schwedler, U.K., Stray, K.M., Aiken, C. and Sundquist, W.I. 2004. Assembly properties of the human immuno- defi ciency virus type 1 CA protein. J. Virol., 78:2545–2552. Garrus, J.E., Von Schwedler, U.K., Pornillos, O.W., Morham, S.G., Zavitz, K.H., Wang, H.E., Wettstein, D.A., Stray, K.M., Cote, M., Rich, R.L., Myszka, D.G. and Sundquist, W.I. 2001. Tsg101 and the vacuolar protein sorting pathway are essential for HIV-1 budding. Cell, 107: 55–65. Gitti, R.K., Lee, B.M., Walker, J., Summers, M.F., Yoo, S. and Sundquist, W.I. 1996. Structure of the amino-terminal core domain of the HIV- 1 capsid protein. Science, 273:231–235. Goila-Gaur, R., Demirov, D.G., Orenstein, J.M., Ono, A. and Freed, E.O. 2003. Defects in human immunodefi ciency virus budding and endo- somal sorting induced by TSG101 overexpression. J. Virol., 77: 6507–6519. Gonzalez, M.E. and Carrasco, L. 2003. Viroporins. FEBS Lett., 552:28–34. Gottlinger, H.G., Dorfman, T., Sodroski, J.G. and Haseltine, W.A. 1991. Effect of mutations affecting the p6 gag protein on human immuno- defi ciency virus particle release. Proc. Natl. Acad. Sci. U.S.A., 88: 3195–3199. Gottlinger, H.G., Sodroski, J.G. and Haseltine, W.A. 1989. Role of capsid precursor processing and myristoylation in morphogenesis and in- fectivity of human immunodefi ciency virus type 1. Proc. Natl. Acad. Sci. U.S.A., 86:5781–5785. Grice, A.L., Kerr, I.D. and Sansom, M.S. 1997. Ion channels formed by HIV-1 Vpu: a modelling and simulation study. FEBS Lett., 405: 299–304. Grinsztejn, B., Nguyen, B.Y., Katlama, C., Gatell, J.M., Lazzarin, A., Vittecoq, D., Gonzalez, C.J., Chen, J., Harvey, C.M. and Isaacs, R.D. 2007. Safety and effi cacy of the HIV-1 integrase inhibitor raltegravir (MK-0518) in treatment-experienced patients with multidrug-resistant virus: a phase II randomised controlled trial. Lancet, 369:1261–1269. Gustchina, A. and Weber, I.T. 1991. Comparative analysis of the sequences and structures of HIV-1 and HIV-2 proteases. Proteins, 10:325– 339. Handley, M.A., Paddock, S., Dall, A. and Panganiban, A.T. 2001. Associa- tion of Vpu-binding protein with microtubules and Vpu-dependent redistribution of HIV-1 Gag protein. Virology, 291:198–207. Harila, K., Prior, I., Sjoberg, M., Salminen, A., Hinkula, J. and Suoma- lainen, M. 2006. Vpu and Tsg101 regulate intracellular targeting of the human immunodefi ciency virus type 1 core protein precursor Pr55gag. J. Virol., 80:3765–3772. Hehl, E.A., Joshi, P., Kalpana, G.V. and Prasad, V.R. 2004. Interaction between human immunodefi ciency virus type 1 reverse transcriptase and integrase proteins. J. Virol., 78:5056–5067. Hill, M., Tachedjian, G. and Mak, J. 2005. The packaging and maturation of the HIV-1 Pol proteins. Curr. HIV. Res., 3:73–85. 178 Wapling et al Drug Target Insights 2007: 2 Himmel, D.M., Sarafi anos, S.G., Dharmasena, S., Hossain, M.M., Mccoy- Simandle, K., Ilina, T., Clark, A.D., Jr., Knight, J.L., Julias, J.G., Clark, P.K. et al. 2006. HIV-1 reverse transcriptase structure with RNase H inhibitor dihydroxy benzoyl naphthyl hydrazone bound at a novel site. ACS Chem. Biol., 1:702–712. Hoglund, S., Su, J., Reneby, S.S., Vegvari, A., Hjerten, S., Sintorn, I.M., Foster, H., Wu, Y.P., Nystrom, I. and Vahlne, A. 2002. Tripeptide interference with human immunodefi ciency virus type 1 morphogen- esis. Antimicrob Agents Chemother., 46:3597–3605. Holm, K., Weclewicz, K., Hewson, R. and Suomalainen, M. 2003. Human immunodefi ciency virus type 1 assembly and lipid rafts: Pr55(gag) associates with membrane domains that are largely resistant to Brij98 but sensitive to Triton X-100. J. Virol., 77:4805–4817. Hosokawa, M., Imai, M., Okada, H. and Okada, N. 2004. Inhibition of HIV-1 infection in cells expressing an artifi cial complementary peptide. Biochem. Biophys. Res. Commun., 324:236–240. Hout, D.R., Gomez, L.M., Pacyniak, E., Miller, J.M., Hill, M.S. and Stephens, E.B. 2006. A single amino acid substitution within the transmembrane domain of the human immunodefi ciency virus type 1 Vpu protein renders simian-human immunodefi ciency virus (SHIV(KU-1bMC33)) susceptible to rimantadine. Virology, 348:449–461. Hsu, K., Seharaseyon, J., Dong, P., Bour, S. and Marban, E. 2004. Mutual functional destruction of HIV-1 Vpu and host TASK-1 channel. Mol. Cell, 14:259–267. Huang, M., Orenstein, J.M., Martin, M.A. and Freed, E.O. 1995. p6Gag is required for particle production from full-length human immunodefi ciency virus type 1 molecular clones expressing protease. J. Virol., 69:6810–6818. Hung, M., Patel, P., Davis, S. and Green, S.R. 1998. Importance of ribo- somal frameshifting for human immunodefi ciency virus type 1 par- ticle assembly and replication. J. Virol., 72:4819–4824. Jacks, T., Madhani, H.D., Masiarz, F.R. and Varmus, H.E. 1988a. Signals for ribosomal frameshifting in the Rous sarcoma virus gag-pol region. Cell, 55:447–458. Jacks, T., Power, M.D., Masiarz, F.R., Luciw, P.A., Barr, P.J. and Varmus, H.E. 1988b. Characterization of ribosomal frameshifting in HIV-1 gag-pol expression. Nature, 331:280–283. Jacks, T., Townsley, K., Varmus, and H.E. Majors, J. 1987. Two effi cient ribosomal frameshifting events are required for synthesis of mouse mammary tumor virus gag-related polyproteins. Proc. Natl. Acad. Sci. U.S.A., 84:4298–4302. Jacks, T. and Varmus, H.E. 1985. Expression of the Rous sarcoma virus pol gene by ribosomal frameshifting. Science, 230:1237–1242. Jacobo-Molina, A., Ding, J., Nanni, R.G., Clark, A.D., Jr., Lu, X., Tantillo, C., Williams, R.L., Kamer, G., Ferris, A.L., Clark, P. et al. 1993. Crystal structure of human immunodefi ciency virus type 1 reverse transcrip- tase complexed with double-stranded DNA at 3.0 A resolution shows bent DNA. Proc. Natl. Acad. Sci. U.S.A., 90:6320–6324. Junker, U., Escaich, S., Plavec, I., Baker, J., Mcphee, F., Rose, J.R., Craik, C.S. and Bohnlein, E. 1996. Intracellular expression of human immunodefi ciency virus type 1 (HIV-1) protease variants inhibits replication of wild-type and protease inhibitor-resistant HIV-1 strains in human T-cell lines. J. Virol., 70:7765–7772. Kalpana, G.V., Marmon, S., Wang, W., Crabtree, G.R. and Goff, S.P. 1994. Binding and stimulation of HIV-1 integrase by a human homolog of yeast transcription factor SNF5. Science, 266:2002–2006. Kane, R.C., Farrell, A.T., Sridhara, R. and Pazdur, R. 2006. United States Food and Drug Administration approval summary: bortezomib for the treatment of progressive multiple myeloma after one prior therapy. Clin. Cancer Res., 12:2955–2960. Kaplan, A.H., Manchester, M. and Swanstrom, R. 1994. The activity of the protease of human immunodefi ciency virus type 1 is initiated at the mem- brane of infected cells before the release of viral proteins and is required for release to occur with maximum effi ciency. J. Virol., 68:6782–6786. Kaplan, A.H., Zack, J.A., Knigge, M., Paul, D.A., Kempf, D.J., Norbeck, D.W. and Swanstrom, R. 1993. Partial inhibition of the human immunodefi ciency virus type 1 protease results in aberrant virus assembly and the formation of noninfectious particles. J. Virol., 67: 4050–4055. Karacostas, V., Wolffe, E.J., Nagashima, K., Gonda, M.A. and Moss, B. 1993. Overexpression of the HIV-1 gag-pol polyprotein results in intracellular activation of HIV-1 protease and inhibition of assembly and budding of virus-like particles. Virology, 193:661–671. Klimkait, T., Strebel, K., Hoggan, M.D., Martin, M.A. and Orenstein, J.M. 1990. The human immunodefi ciency virus type 1-specifi c protein vpu is required for effi cient virus maturation and release. J. Virol., 64:621–629. Kohl, N.E., Emini, E.A., Schleif, W.A., Davis, L.J., Heimbach, J.C., Dixon, R.A., Scolnick, E.M. and Sigal, I.S. 1988. Active human immuno- defi ciency virus protease is required for viral infectivity. Proc. Natl. Acad. Sci. U.S.A., 85:4686–4690. Kohlstaedt, L.A., Wang, J., Friedman, J.M., Rice, P.A. and Steitz, T.A. 1992. Crystal structure at 3.5 A resolution of HIV-1 reverse transcriptase complexed with an inhibitor. Science, 256:1783–1790. Kollmus, H., Honigman, A., Panet, A. and Hauser, H. 1994. The sequences of and distance between two cis-acting signals determine the effi - ciency of ribosomal frameshifting in human immunodefi ciency virus type 1 and human T-cell leukemia virus type II in vivo. J. Virol., 68:6087–6091. Krausslich, H.G. 1991. Human immunodefi ciency virus proteinase dimer as component of the viral polyprotein prevents particle assembly and viral infectivity. Proc. Natl. Acad. Sci. U.S.A., 88:3213–3217. Langner, C.A., Lodge, J.K., Travis, S.J., Caldwell, J.E., Lu, T., Li, Q., Bryant, M.L., Devadas, B., Gokel, G.W., Kobayashi, G.S. et al. 1992. 4-oxatetradecanoic acid is fungicidal for Cryptococcus neoformans and inhibits replication of human immunodefi ciency virus I. J. Biol. Chem., 267:17159–17169. Lanman, J., Lam, T.T., Barnes, S., Sakalian, M., Emmett, M.R., Marshall, A.G. and Prevelige, P.E., Jr. 2003. Identifi cation of novel interactions in HIV-1 capsid protein assembly by high-resolution mass spectrom- etry. J. Mol. Biol., 325:759–772. Lapadat-Tapolsky, M., De Rocquigny, H., Van Gent, D., Roques, B., Plasterk, R. and Darlix, J.L. 1993. Interactions between HIV-1 nucleocapsid protein and viral DNA may have important functions in the viral life cycle. Nucleic Acids Res., 21:831–839. Li, F., Goila-Gaur, R., Salzwedel, K., Kilgore, N.R., Reddick, M., Matallana, C., Castillo, A., Zoumplis, D., Martin, D.E., Orenstein, J.M., Allaway, G.P., Freed, E.O. and Wild, C.T. 2003. PA-457: a potent HIV inhibitor that disrupts core condensation by targeting a late step in Gag processing. Proc. Natl. Acad. Sci. U.S.A., 100:13555–13560. Li, L., Li, H.S., Pauza, C.D., Bukrinsky, M. and Zhao, R.Y. 2005. Roles of HIV-1 auxiliary proteins in viral pathogenesis and host-pathogen interactions. Cell Res., 15:923–934. Li, S., Hill, C.P., Sundquist, W.I. and Finch, J.T. 2000. Image reconstruc- tions of helical assemblies of the HIV-1 CA protein. Nature, 407:409–413. Liao, W.H. and Wang, C.T. 2004. Characterization of human immunodefi - ciency virus type 1 Pr160 gag-pol mutants with truncations down- stream of the protease domain. Virology, 329:180–188. Lindwasser, O.W. and Resh, M.D. 2002. Myristoylation as a target for inhibiting HIV assembly: unsaturated fatty acids block viral budding. Proc. Natl. Acad. Sci. U.S.A., 99:13037–13042. Liu, F., Stephen, A.G., Adamson, C.S., Gousset, K., Aman, M.J., Freed, E.O., Fisher, R.J. and Burke, T.R., Jr. 2006. Hydrazone- and hydrazide- containing N-substituted glycines as peptoid surrogates for expedited library synthesis: Application to the preparation of Tsg101-directed HIV-1 budding antagonists. Org. Lett., 8:5165–5168. Louis, J.M., Clore, G.M. and Gronenborn, A.M. 1999. Autoprocessing of HIV-1 protease is tightly coupled to protein folding. Nat. Struct. Biol., 6:868–875. Lu, Q., Hope, L.W., Brasch, M., Reinhard, C. and Cohen, S.N. 2003. TSG101 interaction with HRS mediates endosomal traffi cking and receptor down-regulation. Proc. Natl. Acad. Sci. U.S.A, 100:7626–7631. Makhija, M.T. 2006. Designing HIV integrase inhibitors--shooting the last arrow. Curr. Med. Chem., 13:2429–2441. Mammano, F., Ohagen, A., Hoglund, S. and Gottlinger, H.G. 1994. Role of the major homology region of human immunodefi ciency virus type 1 in virion morphogenesis. J. Virol., 68:4927–4936. 179 Targeting the late stages of HIV-1 replication Drug Target Insights 2007: 2 Manfredi, R. and Sabbatani, S. 2006. A novel antiretroviral class (fusion inhibitors) in the management of HIV infection. Present features and future perspectives of enfuvirtide (T-20). Curr. Med. Chem., 13: 2369–2384. Maroun, R.G., Gayet, S., Benleulmi, M.S., Porumb, H., Zargarian, L., Merad, H., Leh, H., Mouscadet, J.F., Troalen, F. and Fermandjian, S. 2001. Peptide inhibitors of HIV-1 integrase dissociate the enzyme oligo- mers. Biochemistry, 40:13840–13848. Martin-Serrano, J., Zang, T. and Bieniasz, P.D. 2001. HIV-1 and Ebola virus encode small peptide motifs that recruit Tsg101 to sites of particle assembly to facilitate egress. Nat. Med., 7:1313–1319. Mcphee, F., Good, A.C., Kuntz, I.D. and Craik, C.S. 1996. Engineering human immunodefi ciency virus 1 protease heterodimers as macro- molecular inhibitors of viral maturation. Proc. Natl. Acad. Sci. U.S.A., 93:11477–11481. Mehle, A., Strack, B., Ancuta, P., Zhang, C., McPike, M. and Gabuzda, D. 2004. Vif overcomes the innate antiviral activity of APOBEC3G by promoting its degradation in the ubiquitin-proteasome pathway. J. Biol. Chem., 279:7792–7798. Meng, L., Mohan, R., Kwok, B.H., Elofsson, M., Sin, N. and Crews, C.M. 1999. Epoxomicin, a potent and selective proteasome inhibitor, ex- hibits in vivo antiinfl ammatory activity. Proc. Natl. Acad. Sci. U.S.A., 96:10403–10408. Merabet, N., Dumond, J., Collinet, B., Van Baelinghem, L., Boggetto, N., Ongeri, S., Ressad, F., Reboud-Ravaux, M. and Sicsic, S. 2004. New constrained “molecular tongs” designed to dissociate HIV-1 protease dimer. J. Med. Chem., 47:6392–6400. Momany, C., Kovari, L.C., Prongay, A.J., Keller, W., Gitti, R.K., Lee, B.M., Gorbalenya, A.E., Tong, L., Mcclure, J., Ehrlich, L.S., Summers, M.F., Carter, C. and Rossmann, M.G. 1996. Crystal structure of di- meric HIV-1 capsid protein. Nat. Struct. Biol., 3:763–770. Morikawa, Y., Hinata, S., Tomoda, H., Goto, T., Nakai, M., Aizawa, C., Tanaka, H. and Omura, S. 1996. Complete inhibition of human im- munodefi ciency virus Gag myristoylation is necessary for inhibition of particle budding. J. Biol. Chem., 271:2868–2873. Morris, M.C., Berducou, C., Mery, J., Heitz, F. and Divita, G. 1999a. The thumb domain of the p51-subunit is essential for activation of HIV reverse transcriptase. Biochemistry, 38:15097–15103. Morris, M.C., Robert-Hebmann, V., Chaloin, L., Mery, J., Heitz, F., Devaux, C., Goody, R.S. and Divita, G. 1999b. A new potent HIV-1 reverse transcriptase inhibitor. A synthetic peptide derived from the interface subunit domains. J. Biol. Chem., 274:24941–24946. Muriaux, D., Darlix, J.L. and Cimarelli, A. 2004. Targeting the assembly of the human immunodefi ciency virus type I. Curr. Pharm. Des., 10:3725–3739. Neil, S.J., Eastman, S.W., Jouvenet, N. and Bieniasz, P.D. 2006. HIV-1 Vpu promotes release and prevents endocytosis of nascent retrovirus particles from the plasma membrane. PLoS Pathog., 2:e39. Nguyen, J.T., Porter, M., Amoui, M., Miller, W.T., Zuckermann, R.N. and Lim, W.A. 2000. Improving SH3 domain ligand selectivity using a non-natural scaffold. Chem. Biol., 7:463–473. Niedrig, M., Gelderblom, H.R., Pauli, G., Marz, J., Bickhard, H., Wolf, H. and Modrow, S. 1994. Inhibition of infectious human immunodefi - ciency virus type 1 particle formation by Gag protein-derived peptides. J. Gen. Virol., 75:1469–1474. Ono, A., Ablan, S.D., Lockett, S.J., Nagashima, K. and Freed, E.O. 2004. Phosphatidylinositol (4,5) bisphosphate regulates HIV-1 Gag targeting to the plasma membrane. Proc. Natl. Acad. Sci. U.S.A., 101:14889–14894. Ono, A. and Freed, E.O. 1999. Binding of human immunodefi ciency virus type 1 Gag to membrane: role of the matrix amino terminus. J. Virol., 73:4136–4144. Ono, A., Orenstein, J.M. and Freed, E.O. 2000. Role of the Gag matrix domain in targeting human immunodefi ciency virus type 1 assembly. J. Virol., 74:2855–2866. Ott, D.E., Coren, L.V., Sowder, R.C., Adams, J. and Schubert, U. 2003. Retroviruses have differing requirements for proteasome function in the budding process. J. Virol., 77:3384–3393. Palella, F.J., Jr., Delaney, K.M., Moorman, A.C., Loveless, M.O., Fuhrer, J., Satten, G.A., Aschman, D.J. and Holmberg, S.D. 1998. Declining morbidity and mortality among patients with advanced human immunodefi ciency virus infection. HIV outpatient study investigators. N. Engl. J. Med., 338:853–860. Parang, K., Wiebe, L.I., Knaus, E.E., Huang, J.S., Tyrrell, D.L. and Csizmadia, F. 1997. In vitro antiviral activities of myristic acid analogs against human immunodefi ciency and hepatitis B viruses. Antiviral Res., 34:75–90. Park, J. and Morrow, C.D. 1991. Overexpression of the gag-pol precursor from human immunodefi ciency virus type 1 proviral genomes results in effi cient proteolytic processing in the absence of virion production. J. Virol., 65:5111–5117. Park, J. and Morrow, C.D. 1992. The nonmyristylated Pr160gag-pol poly- protein of human immunodefi ciency virus type 1 interacts with Pr55gag and is incorporated into virus like particles. J. Virol., 66: 6304–6313. Park, S.H. and Raines, R.T. 2000. Genetic selection for dissociative inhibitors of designated protein-protein interactions. Nat. Biotechnol., 18:847–851. Partin, K., Zybarth, G., Ehrlich, L., Decrombrugghe, M., Wimmer, E. and Carter, C. 1991. Deletion of sequences upstream of the proteinase improves the proteolytic processing of human immunodefi ciency virus type 1. Proc. Natl. Acad. Sci. U.S.A., 88:4776–4780. Pettit, S.C., Lindquist, J.N., Kaplan, A.H. and Swanstrom, R. 2005. Process- ing sites in the human immunodefi ciency virus type 1 (HIV-1) Gag-Pro-Pol precursor are cleaved by the viral protease at different rates. Retrovirology, 2:66. Pettit, S.C., Moody, M.D., Wehbie, R.S., Kaplan, A.H., Nantermet, P.V., Klein, C.A. and Swanstrom, R. 1994. The p2 domain of human immunodefi ciency virus type 1 Gag regulates sequential proteolytic processing and is required to produce fully infectious virions. J. Virol., 68:8017–8027. Pettit, S.C., Sheng, N., Tritch, R., Erickson-Viitanen, S. and Swanstrom, R. 1998. The regulation of sequential processing of HIV-1 Gag by the viral protease. Adv. Exp. Med. Biol., 436:15–25. Piacenti, F.J. 2006. An update and review of antiretroviral therapy. Pharmacotherapy, 26:1111–1133. Pidgeon, C., Markovich, R.J., Liu, M.D., Holzer, T.J., Novak, R.M. and Keyer, K.A. 1993. Antiviral phospholipids. Anti-HIV drugs conju- gated to the glycerobackbone of phospholipids. J. Biol. Chem., 268: 7773–7778. Pornillos, O., Alam, S.L., Davis, D.R. and Sundquist, W.I. 2002a. Structure of the Tsg101 UEV domain in complex with the PTAP motif of the HIV-1 p6 protein. Nat. Struct. Biol., 9:812–817. Pornillos, O., Alam, S.L., Rich, R.L., Myszka, D.G., Davis, D.R. and Sun- dquist, W.I. 2002b. Structure and functional interactions of the Tsg101 UEV domain. EMBO J., 21:2397–2406. Pornillos, O., Higginson, D.S., Stray, K.M., Fisher, R.D., Garrus, J.E., Payne, M., He, G.P., Wang, H.E., Morham, S.G. and Sundquist, W.I. 2003. HIV Gag mimics the Tsg101-recruiting activity of the human Hrs protein. J. Cell Biol., 162:425–434. Potts, B.C.M., Faulkner, D.J., Chan, J.A., Simolike, G.C., Offen, P., Hemling, M.E. and Francis, T.E. 1991. Didemnaketal A and B, HIV-1 Protease Inhibitors from the Accidian Didemnum sp. Journal of the American Chemical Society, 113:6321–6322. Quere, L., Wenger, T. and Schramm, H.J. 1996. Triterpenes as potential dimerization inhibitors of HIV-1 protease. Biochem. Biophys. Res. Commun., 227:484–488. Quillent, C., Borman, A.M., Paulous, S., Dauguet, C. and Clavel, F. 1996. Extensive regions of pol are required for effi cient human immuno- defi ciency virus polyprotein processing and particle maturation. Virology, 219:29–36. Rabson, A.B. and Graves, B.J. 1997. Synthesis and processing of viral RNA. In Coffi n, J.M., Hughes, S. H. & Varmus, H.E. ed. Retroviruses. New York, Cold Spring Harbor Laboratory Press. Reicin, A.S., Ohagen, A., Yin, L., Hoglund, S. and Goff, S.P. 1996. The role of Gag in human immunodefi ciency virus type 1 virion morphogen- esis and early steps of the viral life cycle. J. Virol., 70:8645–8652. 180 Wapling et al Drug Target Insights 2007: 2 Rice, W.G., Baker, D.C., Schaeffer, C.A., Graham, L., Bu, M., Terpening, S., Clanton, D., Schultz, R., Bader, J.P., Buckheit, R.W., Jr., Field, L., Singh, P.K. and Turpin, J.A. 1997. Inhibition of multiple phases of human immunodefi ciency virus type 1 replication by a dithiane compound that attacks the conserved zinc fingers of retroviral nucleocapsid proteins. Antimicrob. Agents Chemother., 41:419–426. Rice, W.G., Supko, J.G., Malspeis, L., Buckheit, R.W., Jr., Clanton, D., Bu, M., Graham, L., Schaeffer, C.A., Turpin, J.A., Domagala, J., Gogliotti, R., Bader, J.P., Halliday, S.M., Coren, L., Sowder, R.C., Arthur, L.O. and Henderson, L.E. 1995. Inhibitors of HIV nucleocapsid protein zinc fingers as candidates for the treatment of AIDS. Science, 270:1194–1197. Rodriguez-Barrios, F., Perez, C., Lobaton, E., Velazquez, S., Chamorro, C., San-Felix, A., Perez-Perez, M.J., Camarasa, M.J., Pelemans, H., Balzarini, J. and Gago, F. 2001. Identifi cation of a putative binding site for [2′,5′-bis-O-(tert-butyldimethylsilyl)-beta-D-ribofuranosyl]- 3′-spiro-5″-(4″-amino-1″,2″-oxathiole-2″,2″-dioxide)thymine (TSAO) derivatives at the p51-p66 interface of HIV-1 reverse tran- scriptase. J. Med. Chem., 44:1853–1865. Rodriguez-Rodriguez, L., Tsuchihashi, Z., Fuentes, G.M., Bambara, R.A. and Fay, P.J. 1995. Infl uence of human immunodefi ciency virus nucleocapsid protein on synthesis and strand transfer by the reverse transcriptase in vitro. J. Biol. Chem., 270:15005–15011. Rozzelle, J.E., Dauber, D.S., Todd, S., Kelley, R. and Craik, C.S. 2000. Macromolecular inhibitors of HIV-1 protease. Characterization of designed heterodimers. J. Biol. Chem., 275:7080–7086. Ruland, J., Sirard, C., Elia, A., Macpherson, D., Wakeham, A., Li, L., De La Pompa, J.L., Cohen, S.N. and Mak, T.W. 2001. p53 accumulation, defective cell proliferation, and early embryonic lethality in mice lacking tsg101. Proc. Natl. Acad. Sci. U.S.A., 98:1859–1864. Saad, J.S., Miller, J., Tai, J., Kim, A., Ghanam, R.H. and Summers, M.F. 2006. Structural basis for targeting HIV-1 Gag proteins to the plasma membrane for virus assembly. Proc. Natl. Acad. Sci. U.S.A., 103:11364–11369. Sakalian, M., Mcmurtrey, C.P., Deeg, F.J., Maloy, C.W., Li, F., Wild, C.T. and Salzwedel, K. 2006. 3-O-(3′,3′-dimethysuccinyl) betulinic acid inhibits maturation of the human immunodefi ciency virus type 1 Gag precursor assembled in vitro. J. Virol., 80:5716–5722. Schito, M.L., Goel, A., Song, Y., Inman, J.K., Fattah, R.J., Rice, W.G., Turpin, J.A., Sher, A. and Appella, E. 2003. In vivo antiviral activity of novel human immunodefi ciency virus type 1 nucleocapsid p7 zinc fi nger inhibitors in a transgenic murine model. AIDS Res. Hum. Retroviruses, 19:91–101. Schito, M.L., Soloff, A.C., Slovitz, D., Trichel, A., Inman, J.K., Appella, E., Turpin, J.A. and Barratt-Boyes, S.M. 2006. Preclinical evaluation of a zinc fi nger inhibitor targeting lentivirus nucleocapsid protein in SIV-infected monkeys. Curr. HIV Res., 4:379–386. Schramm, H.J., Boetzel, J., Buttner, J., Fritsche, E., Gohring, W., Jaeger, E., Konig, S., Thumfart, O., Wenger, T., Nagel, N.E. and Schramm, W. 1996. The inhibition of human immunodefi ciency virus prote- ases by ‘interface peptides’. Antiviral Res., 30: 155-170. Schramm, H.J., De Rosny, E., Reboud-Ravaux, M., Buttner, J., Dick, A. and Schramm, W. 1999. Lipopeptides as dimerization inhibitors of HIV-1 protease. Biol. Chem., 380:593–596. Schramm, H.J., Nakashima, H., Schramm, W., Wakayama, H. and Yama- moto, N. 1991. HIV-1 reproduction is inhibited by peptides derived from the N- and C-termini of HIV-1 protease. Biochem. Biophys. Res. Commun., 179:847–851. Schubert, U., Ferrer-Montiel, A.V., Oblatt-Montal, M., Henklein, P., Strebel, K. and Montal, M. 1996a. Identifi cation of an ion channel activity of the Vpu transmembrane domain and its involvement in the regulation of virus release from HIV-1-infected cells. FEBS Lett, 398:12–18. Schubert, U., Ott, D.E., Chertova, E.N., Welker, R., Tessmer, U., Prin- ciotta, M.F., Bennink, J.R., Krausslich, H.G. and Yewdell, J.W. 2000. Proteasome inhibition interferes with gag polyprotein processing, release, and maturation of HIV-1 and HIV-2. Proc. Natl. Acad. Sci. U.S.A., 97:13057–13062. Schwartz, O., Marechal, V., Friguet, B., Arenzana-Seisdedos, F. and Heard, J.M. 1998. Antiviral activity of the proteasome on incoming human immunodefi ciency virus type 1. J. Virol., 72: 3845–3850. Serio, D., Singh, S.P., Cartas, M.A., Weber, I.T., Harrison, R.W., Louis, J.M. and Srinivasan, A. 2000. Antiviral agent based on the non-structural protein targeting the maturation process of HIV-1: expression and susceptibility of chimeric Vpr as a substrate for cleavage by HIV-1 protease. Protein. Eng., 13:431–436. Sheehy, A.M., Gaddis, N.C. and Malim, M.H. 2003. The antiretroviral enzyme APOBEC3G is degraded by the proteasome in response to HIV-1 Vif. Nat. Med., 9:1404–1407. Shehu-Xhilaga, M., Crowe, S.M. and Mak, J. 2001a. Maintenance of the Gag/Gag-Pol ratio is important for human immunodefi ciency virus type 1 RNA dimerization and viral infectivity. J. Virol., 75:1834–1841. Shehu-Xhilaga, M., Kraeusslich, H.G., Pettit, S., Swanstrom, R., Lee, J.Y., Marshall, J.A., Crowe, S.M. and Mak, J. 2001b. Proteolytic process- ing of the p2/nucleocapsid cleavage site is critical for human immunodefi ciency virus type 1 RNA dimer maturation. J. Virol., 75: 9156–9164. Shehu-Xhilaga, M., Tachedjian, G., Crowe, S.M. and Kedzierska, K. 2005. Antiretroviral compounds: mechanisms underlying failure of HAART to eradicate HIV-1. Curr. Med. Chem., 12:1705–1719. Shin, C.G., Taddeo, B., Haseltine, W.A. and Farnet, C.M. 1994. Genetic analysis of the human immunodefi ciency virus type 1 integrase protein. J. Virol., 68:1633–1642. Shkriabai, N., Datta, S.A., Zhao, Z., Hess, S., Rein, and A. Kvaratskhelia, M. 2006. Interactions of HIV-1 Gag with assembly cofactors. Biochemistry, 45:4077–4083. Sluis-Cremer, N., Arion, D., Abram, M.E. and Parniak, M.A. 2004. Proteo- lytic processing of an HIV-1 pol polyprotein precursor: insights into the mechanism of reverse transcriptase p66/p51 heterodimer forma- tion. Int. J. Biochem. Cell. Biol., 36:1836–1847. Sluis-Cremer, N., Dmitrienko, G.I., Balzarini, J., Camarasa, M.J. and Parniak, M.A. 2000. Human immunodefi ciency virus type 1 reverse transcrip- tase dimer destabilization by 1-[Spiro[4″-amino-2″,2″-dioxo-1″,2″ - oxathiole-5″,3′-[2′, 5′-bis-O-(tert-butyldimethylsilyl)-beta-D-ribofu- ranosyl]]]-3-ethylthy mine. Biochemistry, 39:1427–1433. -Cremer, N., Hamamouch, N., San Felix, A., Velazquez, S., Balzarini, J. and Camarasa, M.J. 2006. Structure-activity relationships of [2′,5′- bis-O-(tert-butyldimethylsilyl)-beta-D-ribofuranosyl]- 3′-spiro-5″- (4″-amino-1″,2″-oxathiole-2″,2″-dioxide)thymine derivatives as inhibitors of HIV-1 reverse transcriptase dimerization. J. Med. Chem., 49:4834–4841. Sluis-Cremer, N. and Tachedjian, G. 2002. Modulation of the oligomeric structures of HIV-1 retroviral enzymes by synthetic peptides and small molecules. Eur. J. Biochem., 269:5103–5111. Smith, A.J., Srinivasakumar, N., Hammarskjold, M.L. and Rekosh, D. 1993. Requirements for incorporation of Pr160gag-pol from human immunodefi ciency virus type 1 into virus-like particles. J. Virol., 67: 2266–2275. Song, M., Rajesh, S., Hayashi, Y. and Kiso, Y. 2001. Design and synthesis of new inhibitors of HIV-1 protease dimerization with conformation- ally constrained templates. Bioorg. Med. Chem. Lett., 11:2465– 2468. Sorin, M., Yung, E., Wu, X. and Kalpana, G.V. 2006. HIV-1 replication in cell lines harboring INI1/hSNF5 mutations. Retrovirology, 3:56. Srinivasakumar, N., Hammarskjold, M.L. and Rekosh, D. 1995. Character- ization of deletion mutations in the capsid region of human immunodefi ciency virus type 1 that affect particle formation and Gag-Pol precursor incorporation. J. Virol., 69:6106–6114. Srivastava, S., Sluis-Cremer, N. and Tachedjian, G. 2006. Dimerization of human immunodefi ciency virus type 1 reverse transcriptase as an antiviral target. Curr. Pharm. Des., 12:1879–1894. Stephenson, J. 2007. Researchers buoyed by novel HIV drugs: will expand drug arsenal against resistant virus. JAMA, 297:1535–1536. Sticht, J., Humbert, M., Findlow, S., Bodem, J., Muller, B., Dietrich, U., Werner, J. and Krausslich, H.G. 2005. A peptide inhibitor of HIV-1 assembly in vitro. Nat. Struct. Mol. Biol., 12:671–677. 181 Targeting the late stages of HIV-1 replication Drug Target Insights 2007: 2 Stopak, K., De Noronha, C., Yonemoto, W. and Greene, W.C. 2003. HIV-1 Vif blocks the antiviral activity of APOBEC3G by impairing both its translation and intracellular stability. Mol. Cell., 12:591–601. Swanstrom, R., and J.W. Wills. 1997. Synthesis, assembly, and processing of viral proteins. In Coffi n, J. M., Hughes, S. H. and Varmus H. E. (ed.), Retroviruses. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., p. 263–334. Tachedjian, G., Aronson, H.E., De Los Santos, M., Seehra, J., Mccoy, J.M. and Goff, S.P. 2003. Role of residues in the tryptophan repeat motif for HIV-1 reverse transcriptase dimerization. J. Mol. Biol., 326:381– 396. Tachedjian, G., Aronson, H.E. and Goff, S.P. 2000. Analysis of mutations and suppressors affecting interactions between the subunits of the HIV type 1 reverse transcriptase. Proc. Natl. Acad. Sci. U.S.A., 97: 6334–6339. Tachedjian, G., Moore, K.L., Goff, S.P. and Sluis-Cremer, N. 2005a. Efa- virenz enhances the proteolytic processing of an HIV-1 pol polypro- tein precursor and reverse transcriptase homodimer formation. FEBS Lett., 579:379–384. Tachedjian, G., Orlova, M., Sarafi anos, S.G., Arnold, E. and Goff, S.P. 2001. Nonnucleoside reverse transcriptase inhibitors are chemical enhanc- ers of dimerization of the HIV type 1 reverse transcriptase. Proc. Natl. Acad. Sci. U.S.A., 98:7188–7193. Tachedjian, G., Radzio, J. and Sluis-Cremer, N. 2005b. Relationship between enzyme activity and dimeric structure of recombinant HIV-1 reverse transcriptase. Proteins, 60:5–13. Tanchou, V., Gabus, C., Rogemond, V. and Darlix, J.L. 1995. Formation of stable and functional HIV-1 nucleoprotein complexes in vitro. J. Mol. Biol., 252:563–571. Tang, C., Loeliger, E., Kinde, I., Kyere, S., Mayo, K., Barklis, E., Sun, Y., Huang, M. and Summers, M.F. 2003. Antiviral inhibition of the HIV-1 capsid protein. J. Mol. Biol., 327:1013–1020. Tang, C., Loeliger, E., Luncsford, P., Kinde, I., Beckett, D. and Summers, M.F. 2004. Entropic switch regulates myristate exposure in the HIV-1 matrix protein. Proc. Natl. Acad. Sci. U.S.A., 101:517–522. Tang, C., Ndassa, Y. and Summers, M.F. 2002. Structure of the N-terminal 283-residue fragment of the immature HIV-1 Gag polyprotein. Nat. Struct. Biol., 9:537–543. Telesnitsky A. and Goff, S. 1997. Reverse transcriptase and the generation of retroviral DNA. In Coffi n, J.M., Hughes, S. H. and Varmus, H.E. (ed.), Retroviruses. Cold Spring Harbor Laboratory Press, New York. Ternois, F., Sticht, J., Duquerroy, S., Krausslich, H.G. and Rey, F.A. 2005. The HIV-1 capsid protein C-terminal domain in complex with a virus assembly inhibitor. Nat. Struct. Mol. Biol., 12:678–682. Todd, M.J., Semo, N. and Freire, E. 1998. The structural stability of the HIV-1 protease. J. Mol. Biol., 283:475–488. Turpin, J.A., Terpening, S.J., Schaeffer, C.A., Yu, G., Glover, C.J., Felsted, R.L., Sausville, E.A. and Rice, W.G. 1996. Inhibitors of human immunodefi ciency virus type 1 zinc fi ngers prevent normal process- ing of gag precursors and result in the release of noninfectious virus particles. J. Virol., 70:6180–6189. Uhlikova, T., Konvalinka, J., Pichova, I., Soucek, M., Krausslich, H.G. and Vondrasek, J. 1996. A modular approach to HIV-1 proteinase inhibitor design. Biochem. Biophys. Res. Commun., 222:38–43. Ulysse, L.G. and Chmielewski, J. 1998. Restricting the fl exibility of cross- linked, interfacial peptide inhibitors of HIV-1 protease. Bioorg. Med. Chem. Lett., 8:3281–3286. Venezia, C.F., Howard, K.J., Ignatov, M.E., Holladay, L.A. and Barkley, M.D. 2006. Effects of efavirenz binding on the subunit equilibria of HIV-1 reverse transcriptase. Biochemistry, 45:2779–2789. Veronese, F.D., Copeland, T.D., Oroszlan, S., Gallo, R.C. and Sarngadharan, M.G. 1988. Biochemical and immunological analysis of human im- munodefi ciency virus gag gene products p17 and p24. J. Virol., 62: 795–801. Verplank, L., Bouamr, F., Lagrassa, T.J., Agresta, B., Kikonyogo, A., Leis, J. and Carter, C.A. 2001. Tsg101, a homologue of ubiquitin-conjugating (E2) enzymes, binds the L domain in HIV type 1 Pr55(Gag). Proc. Natl. Acad. Sci., U.S.A., 98: 7724–7729. Vivet-Boudou, V., Didierjean, J., Isel, C. and Marquet, R. 2006. Nucleoside and nucleotide inhibitors of HIV-1 replication. Cell. Mol. Life. Sci., 63:163–186. Vogt, V.M. 1996. Proteolytic processing and particle maturation. Curr. Top. Microbiol. Immunol., 214:95–131. Von Schwedler, U.K., Stray, K.M., Garrus, J.E. and Sundquist, W.I. 2003. Functional surfaces of the human immunodefi ciency virus type 1 capsid protein. J. Virol., 77:5439–5450. Wapling, J., Moore, K.L., Sonza, S., Mak, J. and Tachedjian, G. 2005. Mutations that abrogate human immunodefi ciency virus type 1 reverse transcriptase dimerization affect maturation of the reverse transcrip- tase heterodimer. J. Virol., 79:10247–10257. Wei, B.L., Denton, P.W., O’neill, E., Luo, T., Foster, J.L. and Garcia, J.V. 2005. Inhibition of lysosome and proteasome function enhances human immunodeficiency virus type 1 infection. J. Virol., 79: 5705–5712. Wiegers, K., Rutter, G., Kottler, H., Tessmer, U., Hohenberg, H. and Krausslich, H.G. 1998. Sequential steps in human immunodefi - ciency virus particle maturation revealed by alterations of individu- al Gag polyprotein cleavage sites. J. Virol., 72:2846–2854. Willey, R.L., Maldarelli, F., Martin, M.A. and Strebel, K. 1992. Human immunodefi ciency virus type 1 Vpu protein induces rapid degradation of CD4. J. Virol, 66:7193–7200. Wlodawer, A., Miller, M., Jaskolski, M., Sathyanarayana, B.K., Baldwin, E., Weber, I.T., Selk, L.M., Clawson, L., Schneider, J. and Kent, S.B. 1989. Conserved folding in retroviral proteases: crystal structure of a synthetic HIV-1 protease. Science, 245:616–621. Worthylake, D.K., Wang, H., Yoo, S., Sundquist, W.I. and Hill, C.P. 1999. Structures of the HIV-1 capsid protein dimerization domain at 2.6 A resolution. Acta. Crystallogr. D. Biol. Crystallogr., 55:85–92. Wu, X., Anderson, J.L., Campbell, E.M., Joseph, A.M. and Hope, T.J. 2006. Proteasome inhibitors uncouple rhesus TRIM5alpha restriction of HIV-1 reverse transcription and infection. Proc. Natl. Acad. Sci. U.S.A., 103:7465–7470. Wu, X., Liu, H., Xiao, H., Conway, J.A., Hunter, E. and Kappes, J.C. 1997. Functional RT and IN incorporated into HIV-1 particles indepen- dently of the Gag/Pol precursor protein. EMBO J., 16:5113–5122. Yeni, P. 2006. Update on HAART in HIV. J. Hepatol., 44:S100–103. Yeni, P.G., Hammer, S.M., Carpenter, C.C., Cooper, D.A., Fischl, M.A., Gatell, J.M., Gazzard, B.G., Hirsch, M.S., Jacobsen, D.M., Katzen- stein, D.A., et al. 2002. Antiretroviral treatment for adult HIV infec- tion in 2002: updated recommendations of the International AIDS Society-USA Panel. JAMA, 288:222–235. Yu, Q., Ottmann, M., Pechoux, C., Le Grice, S. and Darlix, J.L. 1998. Mutations in the primer grip of human immunodefi ciency virus type 1 reverse transcriptase impair proviral DNA synthesis and virion maturation. J. Virol., 72:7676–7680. Yu, X., Yu, Y., Liu, B., Luo, K., Kong, W., Mao, P. and Yu, X.F. 2003. Induc- tion of APOBEC3G ubiquitination and degradation by an HIV-1 Vif-Cul5-SCF complex. Science, 302:1056–1060. Yung, E., Sorin, M., Pal, A., Craig, E., Morozov, A., Delattre, O., Kappes, J., Ott, D. and Kalpana, G.V. 2001. Inhibition of HIV-1 virion produc- tion by a transdominant mutant of integrase interactor 1. Nat. Med., 7:920–926. Yung, E., Sorin, M., Wang, E.J., Perumal, S., Ott, D. and Kalpana, G.V. 2004. Specifi city of interaction of INI1/hSNF5 with retroviral inte- grases and its functional signifi cance. J. Virol., 78:2222–2231. Zhang, Z.Y., Poorman, R.A., Maggiora, L.L., Heinrikson, R.L. and Kezdy, F.J. 1991. Dissociative inhibition of dimeric enzymes. Kinetic char- acterization of the inhibition of HIV-1 protease by its COOH-termi- nal tetrapeptide. J. Biol. Chem., 266:15591–15594. Zhao, L., O’reilly, M.K., Shultz, M.D. and Chmielewski, J. 2003. Interfacial peptide inhibitors of HIV-1 integrase activity and dimerization. Bioorg. Med. Chem. Lett., 13:1175-1177. Zhou, J., Chen, C.H. and Aiken, C. 2004. The sequence of the CA-SP1 junction accounts for the differential sensitivity of HIV-1 and SIV to the small molecule maturation inhibitor 3-O-{3′,3′-dimethylsucci- nyl}-betulinic acid. Retrovirology, 1:15. 182 Wapling et al Drug Target Insights 2007: 2 Zhou, J., Chen, C.H. and Aiken, C. 2006. HIV-1 Resistance to the Small Molecule Maturation Inhibitor 3-O-{3′,3′-dimethylsuccinyl}-betu- linic acid is conferred by a variety of single amino acid substitutions at the CA-SP1 cleavage site in Gag. J. Virol., 80:12095–12101. Zhou, J., Huang, L., Hachey, D.L., Chen, C.H. and Aiken, C. 2005. Inhibi- tion of HIV-1 maturation via drug association with the viral Gag protein in immature HIV-1 particles. J. Biol. Chem., 280:42149– 42155. Zutshi, R. and Chmielewski, J. 2000. Targeting the dimerization interface for irreversible inhibition of HIV-1 protease. Bioorg. Med. Chem. Lett., 10:1901–1903. << /ASCII85EncodePages false /AllowTransparency false /AutoPositionEPSFiles true /AutoRotatePages /None /Binding /Left /CalGrayProfile (Dot Gain 20%) /CalRGBProfile (sRGB IEC61966-2.1) /CalCMYKProfile (U.S. Web Coated \050SWOP\051 v2) /sRGBProfile (sRGB IEC61966-2.1) /CannotEmbedFontPolicy /Error /CompatibilityLevel 1.4 /CompressObjects /Tags /CompressPages true /ConvertImagesToIndexed true /PassThroughJPEGImages true /CreateJDFFile false /CreateJobTicket false /DefaultRenderingIntent /Default /DetectBlends true /ColorConversionStrategy /LeaveColorUnchanged /DoThumbnails false /EmbedAllFonts true /EmbedJobOptions true /DSCReportingLevel 0 /EmitDSCWarnings false /EndPage -1 /ImageMemory 1048576 /LockDistillerParams false /MaxSubsetPct 100 /Optimize true /OPM 1 /ParseDSCComments true /ParseDSCCommentsForDocInfo true /PreserveCopyPage true /PreserveEPSInfo true /PreserveHalftoneInfo false /PreserveOPIComments false /PreserveOverprintSettings true /StartPage 1 /SubsetFonts true /TransferFunctionInfo /Apply /UCRandBGInfo /Preserve /UsePrologue false /ColorSettingsFile () /AlwaysEmbed [ true ] /NeverEmbed [ true ] /AntiAliasColorImages false /DownsampleColorImages true /ColorImageDownsampleType /Bicubic /ColorImageResolution 300 /ColorImageDepth -1 /ColorImageDownsampleThreshold 1.50000 /EncodeColorImages true /ColorImageFilter /DCTEncode /AutoFilterColorImages true /ColorImageAutoFilterStrategy /JPEG /ColorACSImageDict << /QFactor 0.15 /HSamples [1 1 1 1] /VSamples [1 1 1 1] >> /ColorImageDict << /QFactor 0.15 /HSamples [1 1 1 1] /VSamples [1 1 1 1] >> /JPEG2000ColorACSImageDict << /TileWidth 256 /TileHeight 256 /Quality 30 >> /JPEG2000ColorImageDict << /TileWidth 256 /TileHeight 256 /Quality 30 >> /AntiAliasGrayImages false /DownsampleGrayImages true /GrayImageDownsampleType /Bicubic /GrayImageResolution 300 /GrayImageDepth -1 /GrayImageDownsampleThreshold 1.50000 /EncodeGrayImages true /GrayImageFilter /DCTEncode /AutoFilterGrayImages true /GrayImageAutoFilterStrategy /JPEG /GrayACSImageDict << /QFactor 0.15 /HSamples [1 1 1 1] /VSamples [1 1 1 1] >> /GrayImageDict << /QFactor 0.15 /HSamples [1 1 1 1] /VSamples [1 1 1 1] >> /JPEG2000GrayACSImageDict << /TileWidth 256 /TileHeight 256 /Quality 30 >> /JPEG2000GrayImageDict << /TileWidth 256 /TileHeight 256 /Quality 30 >> /AntiAliasMonoImages false /DownsampleMonoImages true /MonoImageDownsampleType /Bicubic /MonoImageResolution 1200 /MonoImageDepth -1 /MonoImageDownsampleThreshold 1.50000 /EncodeMonoImages true /MonoImageFilter /CCITTFaxEncode /MonoImageDict << /K -1 >> /AllowPSXObjects false /PDFX1aCheck false /PDFX3Check false /PDFXCompliantPDFOnly false /PDFXNoTrimBoxError true /PDFXTrimBoxToMediaBoxOffset [ 0.00000 0.00000 0.00000 0.00000 ] /PDFXSetBleedBoxToMediaBox true /PDFXBleedBoxToTrimBoxOffset [ 0.00000 0.00000 0.00000 0.00000 ] /PDFXOutputIntentProfile () /PDFXOutputCondition () /PDFXRegistryName (http://www.color.org) /PDFXTrapped /Unknown /Description << /JPN <FEFF3053306e8a2d5b9a306f30019ad889e350cf5ea6753b50cf3092542b308030d730ea30d730ec30b9537052377528306e00200050004400460020658766f830924f5c62103059308b3068304d306b4f7f75283057307e305930023053306e8a2d5b9a30674f5c62103057305f00200050004400460020658766f8306f0020004100630072006f0062006100740020304a30883073002000520065006100640065007200200035002e003000204ee5964d30678868793a3067304d307e305930023053306e8a2d5b9a306b306f30d530a930f330c8306e57cb30818fbc307f304c5fc59808306730593002> /FRA <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> /DEU <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> /PTB <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> /DAN <FEFF004200720075006700200064006900730073006500200069006e0064007300740069006c006c0069006e006700650072002000740069006c0020006100740020006f0070007200650074007400650020005000440046002d0064006f006b0075006d0065006e0074006500720020006d006500640020006800f8006a006500720065002000620069006c006c00650064006f0070006c00f80073006e0069006e0067002000740069006c0020007000720065002d00700072006500730073002d007500640073006b007200690076006e0069006e0067002000690020006800f8006a0020006b00760061006c0069007400650074002e0020005000440046002d0064006f006b0075006d0065006e007400650072006e00650020006b0061006e002000e50062006e006500730020006d006500640020004100630072006f0062006100740020006f0067002000520065006100640065007200200035002e00300020006f00670020006e0079006500720065002e00200044006900730073006500200069006e0064007300740069006c006c0069006e0067006500720020006b007200e600760065007200200069006e0074006500670072006500720069006e006700200061006600200073006b007200690066007400740079007000650072002e> /NLD <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> /ESP <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> /SUO <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> /ITA <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> /NOR <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> /SVE <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> /ENU <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> >> >> setdistillerparams << /HWResolution [2400 2400] /PageSize [612.000 792.000] >> setpagedevice