throbber
CMLS, Cell. Mol. Life Sci. 57 (2000) 1408–1422
`1420-682X:00:101408-15 $ 1.50(cid:27)0.20:0
`© Birkha¨user Verlag, Basel, 2000
`
`Review
`
`Molecular mechanisms of HIV-1 resistance to nucleoside
`reverse transcriptase inhibitors (NRTIs)
`N. Sluis-Cremer, D. Arion and M. A. Parniak*
`
`Lady Davis Institute for Medical Research and McGill University AIDS Centre, Sir Mortimer B. Davis-Jewish
`General Hospital, 3755 Cote Ste-Catherine Road, Montreal, Quebec H3T 1E2 (Canada), Fax (cid:27)1514 340 7502,
`e-mail: mparniak@ldi.jgh.mcgill.ca
`
`Received 16 December 1999; received after revision 3 April 2000; accepted 3 April 2000
`
`Abstract. Nucleoside reverse transcriptase inhibitors
`typic mechanisms have been identified or proposed to
`such as 3%-azido-3%-deoxythymidine, 2%,3%-
`(NRTIs),
`account for HIV-1 RT resistance to NRTIs. These
`dideoxyinosine and 2%,3%-dideoxy-3%-thiacytidine, are ef-
`mechanisms include alterations of RT discrimination
`fective inhibitors of human immunodeficiency type 1
`between NRTIs and the analogous dNTP (direct effects
`(HIV-1)
`replication. NRTIs
`are deoxynucleoside
`on NRTI binding and:or incorporation), alterations in
`triphosphate analogs, but lack a free 3%-hydroxyl group. RT-template:primer interactions, which may influence
`Once NRTIs are incorporated into the nascent viral
`subsequent NRTI incorporation, and enhanced removal
`of the chain-terminating residue from the 3% end of the
`DNA, in reactions catalyzed by HIV-1 reverse tran-
`primer. These different resistance phenotypes seem to
`scriptase (RT), further viral DNA synthesis is effectively
`terminated. NRTIs should therefore represent the ideal
`correlate with different sets of mutations in RT. This
`review discusses the relationship between HIV-1 drug
`antiviral agent. Unfortunately, HIV-1 inevitably devel-
`resistance genotype and phenotype, in relation to our
`ops resistance to these inhibitors, and this resistance
`correlates with mutations in RT. To date, three pheno-
`current knowledge of HIV-1 RT structure.
`
`Key words. Human immunodeficiency virus type 1; reverse transcriptase; nucleoside reverse transcriptase in-
`hibitors; DNA polymerization; chain termination; antiviral drug resistance; phosphorolysis; pyrophosphorolysis.
`
`Introduction
`
`Retroviruses such as the human immunodeficiency virus
`(HIV) are RNA viruses that replicate through a double-
`strand DNA intermediate. This novel viral replication
`cycle requires that retroviruses carry a specific enzyme,
`reverse transcriptase (RT), since there are no cellular
`enzymes that can convert single-strand RNA into dou-
`ble-strand DNA. RT is a DNA polymerase that can
`copy both DNA templates (like cellular enzymes) and
`RNA templates (unlike cellular enzymes). HIV RT dif-
`
`* Corresponding author.
`
`fers from cellular DNA polymerases in two additional
`respects. First, HIV RT readily utilizes many chemically
`altered analogs of the normal deoxynucleoside triphos-
`phate (dNTP) DNA polymerase substrates. Second,
`HIV RT lacks a formal %proofreading% activity. These
`characteristics are important from a pharmaceutical
`focus, and direct the use of nucleoside analog inhibitors
`as anti-HIV pharmaceuticals. As of January 2000, six of
`the current FDA-approved anti-HIV drugs are nu-
`cleoside reverse transcriptase inhibitors (NRTIs) [1].
`NRTIs are analogs of the normal dNTP substrates of
`DNA polymerases, with important modifications (fig.
`1). The 2%,3%-dideoxynucleosides such as ddC and ddI
`
`Columbia Ex. 2080
`Illumina, Inc. v. The Trustees
`of Columbia University in the
`City of New York
`IPR2020-00988, -01065,
`-01177, -01125, -01323
`
`

`

`CMLS, Cell. Mol. Life Sci. 57 (2000) 1408–1422
`1420-682X:00:101408-15 $ 1.50(cid:27)0.20:0
`© Birkha¨user Verlag, Basel, 2000
`
`Review
`
`Molecular mechanisms of HIV-1 resistance to nucleoside
`reverse transcriptase inhibitors (NRTIs)
`N. Sluis-Cremer, D. Arion and M. A. Parniak*
`
`Lady Davis Institute for Medical Research and McGill University AIDS Centre, Sir Mortimer B. Davis-Jewish
`General Hospital, 3755 Cote Ste-Catherine Road, Montreal, Quebec H3T 1E2 (Canada), Fax (cid:27)1514 340 7502,
`e-mail: mparniak@ldi.jgh.mcgill.ca
`
`Received 16 December 1999; received after revision 3 April 2000; accepted 3 April 2000
`
`Abstract. Nucleoside reverse transcriptase inhibitors
`typic mechanisms have been identified or proposed to
`such as 3%-azido-3%-deoxythymidine, 2%,3%-
`(NRTIs),
`account for HIV-1 RT resistance to NRTIs. These
`dideoxyinosine and 2%,3%-dideoxy-3%-thiacytidine, are ef-
`mechanisms include alterations of RT discrimination
`fective inhibitors of human immunodeficiency type 1
`between NRTIs and the analogous dNTP (direct effects
`(HIV-1)
`replication. NRTIs
`are deoxynucleoside
`on NRTI binding and:or incorporation), alterations in
`triphosphate analogs, but lack a free 3%-hydroxyl group. RT-template:primer interactions, which may influence
`Once NRTIs are incorporated into the nascent viral
`subsequent NRTI incorporation, and enhanced removal
`of the chain-terminating residue from the 3% end of the
`DNA, in reactions catalyzed by HIV-1 reverse tran-
`primer. These different resistance phenotypes seem to
`scriptase (RT), further viral DNA synthesis is effectively
`terminated. NRTIs should therefore represent the ideal
`correlate with different sets of mutations in RT. This
`review discusses the relationship between HIV-1 drug
`antiviral agent. Unfortunately, HIV-1 inevitably devel-
`resistance genotype and phenotype, in relation to our
`ops resistance to these inhibitors, and this resistance
`correlates with mutations in RT. To date, three pheno-
`current knowledge of HIV-1 RT structure.
`
`Key words. Human immunodeficiency virus type 1; reverse transcriptase; nucleoside reverse transcriptase in-
`hibitors; DNA polymerization; chain termination; antiviral drug resistance; phosphorolysis; pyrophosphorolysis.
`
`Introduction
`
`Retroviruses such as the human immunodeficiency virus
`(HIV) are RNA viruses that replicate through a double-
`strand DNA intermediate. This novel viral replication
`cycle requires that retroviruses carry a specific enzyme,
`reverse transcriptase (RT), since there are no cellular
`enzymes that can convert single-strand RNA into dou-
`ble-strand DNA. RT is a DNA polymerase that can
`copy both DNA templates (like cellular enzymes) and
`RNA templates (unlike cellular enzymes). HIV RT dif-
`
`* Corresponding author.
`
`fers from cellular DNA polymerases in two additional
`respects. First, HIV RT readily utilizes many chemically
`altered analogs of the normal deoxynucleoside triphos-
`phate (dNTP) DNA polymerase substrates. Second,
`HIV RT lacks a formal %proofreading% activity. These
`characteristics are important from a pharmaceutical
`focus, and direct the use of nucleoside analog inhibitors
`as anti-HIV pharmaceuticals. As of January 2000, six of
`the current FDA-approved anti-HIV drugs are nu-
`cleoside reverse transcriptase inhibitors (NRTIs) [1].
`NRTIs are analogs of the normal dNTP substrates of
`DNA polymerases, with important modifications (fig.
`1). The 2%,3%-dideoxynucleosides such as ddC and ddI
`
`

`

`CMLS, Cell. Mol. Life Sci. Vol. 57, 2000
`
`Review Article
`
`1409
`
`lack a 3%-OH on the sugar, whereas other analogs such
`as 3%-azido-3%-deoxythymidine (AZT) have the 3%-OH
`replaced by other functional groups that do not allow
`primer extension. NRTIs require intracellular metabolic
`transformation for antiviral activity, namely conversion
`to the triphosphate, a process catalyzed by cellular
`kinases [2].
`After conversion to the active triphosphate, NRTIs
`must compete with the natural dNTPs both for recogni-
`tion by RT as a substrate (binding) and for incorpora-
`tion into the nascent viral DNA chain (catalysis).
`NRTIs thus inhibit RT-catalyzed proviral DNA synthe-
`sis by two mechanisms [3]. First, they are competitive
`inhibitors for binding and:or catalytic incorporation
`with respect to the analogous dNTP substrate. Second,
`they terminate further viral DNA synthesis, due to lack
`of a 3%-OH group. Chain termination is the principal
`mechanism of NRTI antiviral action.
`NRTIs should be the ‘ideal’ anti-HIV therapeutics.
`Each HIV virion carries only two copies of genomic
`RNA. There are about 20,000 nucleotide incorporation
`
`events catalyzed by RT during the synthesis of complete
`viral DNA, thus providing about 5000 chances for
`chain termination by any given NRTI. Since HIV-1 RT
`lacks a formal proofreading activity (i.e. some formal
`mechanism to identify and excise inappropriate nucle-
`otide incorporation), a single NRTI incorporation event
`should suffice to quell viral DNA synthesis. In reality,
`NRTIs are less potent inhibitors of HIV replication
`than might be expected; reasons for this will be dis-
`cussed later. In addition, although NRTI therapy is
`initially quite effective in reducing viral load in HIV-1-
`infected individuals,
`the viral burden inevitably re-
`bounds despite
`continued therapy, due
`to the
`appearance of drug-resistant strains of HIV. Numerous
`mutations in HIV-1 RT have been identified in NRTI-
`resistant HIV strains (table 1) [4, 5].
`The simplest mechanism for resistance would be one of
`discrimination, i.e. some mechanism for RT to exclude
`the NRTI, while retaining the ability to recognize the
`analogous natural dNTP substrate. However, this dis-
`crimination is actually somewhat of a problem, since in
`
`Figure 1. Structures of current clinically-used nucleoside reverse transcriptase inhibitors (NRTI). AZT (1), ddI (2), ddC (3), d4T (4),
`3TC (5), abacavir (6).
`
`

`

`1410
`
`N. Sluis-Cremer et al.
`
`HIV resistance to NRTI
`
`Table 1. Mutations in HIV-1 RT correlated with resistance to NRTI.
`
`ddN
`
`RT residue
`
`41
`
`65
`
`67
`
`69
`
`70
`
`74
`
`75
`
`151*
`
`184
`
`210
`
`215
`
`219
`
`AZT†
`ddC
`3TC
`ddI (ddA)
`d4T
`Abacavir
`
`M41L
`
`D67N
`
`K70R
`
`K70E
`
`T69D
`
`68¡¡70‡
`
`K65R
`K65R
`K65R
`
`K65R
`
`V75T
`
`V75T
`V75T
`
`L74V
`
`L74V
`
`M184V
`M184V
`
`M184V
`
`L210W T215Y:F
`
`K219Q
`
`* The mutation Q151M appears in patients with resistance to multiple ddN.
`† High-level AZT resistance requires the presence of two or more mutations.
`‡ Two-amino acid insertion mutants are not specific for d4T resistance, but were first identified in patients with d4T resistance. These
`insertion mutations are found in multidrug resistant HIV-1, generally over a background of AZT-resistance mutations.
`
`Table 2. Resistance phenotypes associated with NRTI-resistance mutations in HIV-1 RT.
`
`Resistance conferred
`
`NRTI-resistance phenotype
`
`Mutation
`
`K65R
`T69-S-S:G-K70*
`
`L74V
`V75T
`E89G
`Q151M
`M184I:V
`
`ddC, ddI, 3TC, PMEA
`multidrug resistance
`
`ddI, ddC
`ddC, d4T
`ddG
`multidrug resistance
`3TC
`
`discrimination
`uncertain (may be combination of discrimination and
`phosphorolysis)
`T:P repositioning
`uncertain
`T:P repositioning
`discrimination
`discrimination (also negative effect on phosphorolysis);T:P
`repositioning may also play a role
`phosphorolysis
`phosphorolysis†
`uncertain
`
`D67N:K70R:T215F(Y):Q219K
`M41L:T215Y
`L210W
`
`AZT
`AZT
`AZT
`
`* AZT-resistance mutations are required in addition to insertion mutations to provide multi-drug resistance.
`† D. Arion, N. Sluis-Cremer, M. A. Parniak, (unpublished).
`
`many cases this requires that RT must selectively ignore
`a structurally less rich compound (NRTI, lacking the
`3%-OH) in favor of the structurally more complex dNTP
`analog. In addition, our understanding of the molecular
`aspects of NRTI resistance has been complicated by the
`complex patterns of mutations required for resistance to
`some NRTIs such as AZT [6].
`To date, three mechanisms have been proposed to ac-
`count for the molecular basis of the NRTI resistance
`phenotype. These mechanisms apply to different stages
`of NRTI inhibition, and include (i) Selective alterations
`in NRTI binding and:or incorporation (i.e. discrimina-
`tion), (ii) template:primer (T:P) repositioning, which
`then influences NRTI incorporation and (iii) phospho-
`rolytic removal of an incorporated chain-terminating
`NRTI residue from the 3%-end of the nascent viral
`DNA. Correlations of these phenotypes with specific
`mutations in RT and the NRTIs affected are summa-
`rized in table 2.
`
`HIV-1 RT structure and function
`
`The HIV-1 RT gene encodes a 66-kDa protein; how-
`ever, the presumed biologically relevant form of HIV-1
`RT is a heterodimer comprising of subunits of 66 and
`51 kDa (termed p66 and p51) [7]. The p51 subunit is
`produced during viral assembly and maturation via
`HIV-1 protease-mediated cleavage of the C-terminal
`domain of a p66 subunit. The structure of the HIV-1
`RT heterodimer is illustrated in fig. 2.
`The overall shape of the p66 subunit has been likened to
`that of a ‘right hand’ [8], with the major subdomains of
`the polymerase domain of p66 appropriately termed
`fingers (residues 1–85, 118–155), palm (86–117, 156–
`237) and thumb (238–318) (fig. 2). The DNA poly-
`merase catalytic aspartate residues (D110, D185, D186)
`are in the palm subdomain. In addition, the p66 has two
`additional major subdomains, the ‘connection’ (residues
`319–426) and the C-terminal ribonuclease H (RNase
`
`

`

`CMLS, Cell. Mol. Life Sci. Vol. 57, 2000
`
`Review Article
`
`1411
`
`H) (427–565) domains. The latter subdomain is missing
`in the p51 subunit. Whereas the overall folding of the
`subdomains is similar in both p66 and p51 subunits, the
`spatial arrangement of the subdomains differs markedly
`[8, 9]. The p66 subunit adopts an ‘open’, catalytically
`competent conformation that can ‘grasp’ a nucleic acid
`template, whereas the p51 subunit is in a ‘closed’ confor-
`mation. The p51 subunit is considered to play a largely
`structural role, although it may also be important in
`interacting with the transfer RNA (tRNA)Lys3 primer
`used for the initiation of HIV-1 DNA synthesis [8].
`Mutations associated with NRTI resistance occur pri-
`marily in the fingers and the palm subdomains of RT
`(table 1, fig. 2). Because of the nature of HIV-1 RT
`
`heterodimer formation, NRTI resistance mutations ob-
`viously occur in both p66 and p51 subunits. However,
`only those in the p66 subunit are generally considered to
`have phenotypic consequences. Whereas one report has
`hypothesized that mutations in the p51 subunit may also
`contribute to the resistance phenotype [10], there are no
`biochemical data to support this conjecture.
`The conversion of HIV-1 genomic RNA into double-
`strand viral DNA is a complex process, yet all chemical
`steps are catalyzed by RT. This requires RT to be
`multifunctional, with two types of DNA polymerase
`activity, RNA-dependent DNA polymerase (RDDP) to
`synthesize a DNA strand copy of the viral genomic
`RNA template and DNA-dependent DNA polymerase
`
`Figure 2. Structure of the HIV-1 RT heterodimer showing locations of residues mutated in NRTI resistance. The crystal coordinates
`used to generate this figure are Brookhaven Protein Data Bank (PDB) 2HMI [19].
`
`

`

`1412
`
`N. Sluis-Cremer et al.
`
`HIV resistance to NRTI
`
`Figure 3. Reaction mechanism for RT-catalyzed DNA synthesis, showing the multiple mechanistic forms of RT involved, and the
`relationship between forward reaction DNA synthesis and reverse reaction pyrophosphorolysis.
`
`(DDDP) to complete the synthesis of double-strand
`viral DNA. As well, RT possesses an intrinsic RNase H
`activity, to degrade the genomic RNA component of
`
`the DNA:RNA duplex intermediate formed during RT
`RDDP synthesis. NRTIs are directed against RT
`RDDP and DDDP.
`
`

`

`CMLS, Cell. Mol. Life Sci. Vol. 57, 2000
`
`Review Article
`
`1413
`
`HIV-1 RT DNA synthesis follows an ordered ‘bi bi’
`mechanism [11–14] involving several RT mechanistic
`species. Free RT first binds the template:primer (T:P) to
`form a tight RT-T:Pn binary complex (fig. 3, step 1).
`This is followed by the binding of dNTP, to form the
`RT-T:Pn-dNTP ternary complex (step 2, fig. 3). Binding
`of dNTP appears to be a two-stage process [15]. The
`initial interaction may be nonselective, with each of the
`four dNTPs binding with relatively similar affinities.
`The second stage is more selective, and involves the
`correct positioning of only that dNTP complementary
`to the template base. The selectivity involved in posi-
`tioning and incorporating the correct nucleotide is pri-
`marily controlled by the free energy of base pairing with
`the complementary template nucleotide base. Since NR-
`TIs and the analogous dNTPs have identical base struc-
`tures, in most cases RT has little opportunity to readily
`effect discrimination between natural substrates and
`inhibitors at this stage.
`Formation of the initial RT-T:Pn-dNTP induces a rate-
`limiting change in protein conformation to form a very
`tight ternary complex, RT*-T:Pn-dNTP (step 3, fig. 3).
`The formation of this ternary complex allows the criti-
`cal transition state to be reached, enabling nucleophilic
`attack by the 3%-OH primer terminus on the a-phos-
`phate of the bound dNTP. This results in phosphodi-
`ester-bond formation and extension of the viral DNA
`strand by one nucleotide, along with formation of the
`RT-T:Pn(cid:27) 1-PPi ternary pyrophosphate complex (step
`4). The PPi product then dissociates, leaving the RT-T:
`Pn(cid:27) 1 binary complex, which is kinetically indistinguish-
`able from the RT-T:Pn binary complex. RT can then
`either dissociate from the newly elongated T:Pn(cid:27) 1 (dis-
`tributive mode of polymerization) or translocate along
`the bound template, bind the next complementary
`dNTP and continue DNA synthesis (processive mode of
`polymerization). The key kinetic steps that define the
`incorporation of dNTP (and the analogous NRTIs) are
`the nucleotide binding event (step 2), the rate-limiting
`conformational change (step 3) and the efficiency of
`incorporation (step 4).
`Crystal structures for three of the RT mechanistic forms
`have been solved. These include substrate-free enzyme
`(RT) [16, 17], the RT-T:P binary complex [18, 19] and
`an RT*-T:P-dNTP ternary complex [20]. The ‘missing’
`structures in terms of the RT reaction pathway include
`the RT-T:P-PPi product ternary complex. Also missing
`is that complex resulting from the initial binding of the
`dNTP (RT-T:P-dNTP). However, such a structure has
`been solved for another DNA polymerase [21]. These
`structural data have provided considerable insight into
`the conformational changes associated with DNA poly-
`merization, the molecular structure of the RT active
`site, and the relative position of certain critical residues
`during catalysis, all of which are important for under-
`
`standing the molecular basis of HIV-1 RT resistance to
`NRTIs.
`Comparison of the structures of free RT with the RT-T:
`P binary complex shows that interaction of RT with the
`nucleic acid induces a conformational rotation of the
`p66 thumb subdomain [18, 19]. Comparison of the
`structures of the RT-T:P binary complex with the RT*-
`T:P-dNTP ternary complex reveals that dNTP binding
`induces further conformational changes in RT [20]. In
`particular, parts of the fingers subdomain rotate in-
`wards toward the palm subdomain and the polymerase
`active site, effectively ‘clamping’ the dNTP into the
`active site region. Two fingers residues make important
`contacts with the dNTP substrate. The o-amino of K65
`and the guanidinium group of R72 interact with the g-
`and a-phosphates of the bound dNTP substrate, respec-
`tively (fig. 4). As seen in tables 1 and 2, the K65R
`mutation arises in HIV-1 resistance to a number of
`different NRTIs. The dNTP triphosphate moeity also
`interacts with the main-chain amide-NH of residues
`D113 and A114, and with the Mg2(cid:27) divalent metal ion
`cofactor bound to the polymerase active site aspartates
`(D110, D185, D186).
`The divalent metal ions facilitate the nucleophilic attack
`by the 3%-OH of the primer terminus on the a-phospho-
`rous of the incoming dNTP, and stabilize the transition
`state [22]. Only two of the active site aspartates (D110,
`D185) are coordinated to Mg2(cid:27) in the HIV-1 RT
`ternary complex [20]. The third aspartate (D186) may
`be involved in positioning the primer terminus by inter-
`acting with the 3%-OH of the primer terminal nucleotide
`(this functional group is absent in the ternary complex).
`The sugar 3%-OH of the dNTP projects into a small
`pocket formed by the side chains of D113, A114, Y115,
`F116 and Q151. Interestingly, the 3%-OH of the nucle-
`otide forms a hydrogen bond with the main-chain -NH
`of Y115. Since all NRTIs are missing the 3%-OH, this
`interaction may be of significance in understanding the
`kinetic efficiency of NRTI incorporation into the grow-
`ing viral DNA chain.
`
`Mechanisms of HIV-1 resistance to NRTIs
`
`Discrimination-resistance due to impaired NRTI binding
`and:or incorporation
`A large number of mutations in HIV-1 RT have been
`identified in resistance to NRTIs (tables 1 and 2). In a
`few cases, such as the AZT resistance and certain multi-
`drug resistance phenotypes, multiple mutations are
`needed to observe high phenotypic resistance. For the
`most part, however, single point mutations suffice to
`confer resistance to individual NRTI.
`Point mutations such as K65R, T69D, Q151M and
`M184V:I lead to alterations in affinity of RT for spe-
`
`

`

`1414
`
`N. Sluis-Cremer et al.
`
`HIV resistance to NRTI
`
`cific NRTIs with little or no change in affinity for the
`corresponding dNTP substrate. These mutations are
`located in, or close to, the dNTP substrate binding site
`[20], and may therefore affect the initial binding and:or
`the subsequent positioning of the NRTI for catalysis in
`a manner such that the mutant RT is able to discrimi-
`nate between the NRTI and the analogous dNTP sub-
`strate. As mentioned previously, discrimination between
`NRTIs and analogous dNTPs requires that RT must
`selectively ignore the structurally less rich NRTI in
`favor of the structurally more complex dNTP analog. It
`is important to note that wild-type RT is generally less
`efficient at catalyzing the incorporation of bound
`ddNTP into the nascent DNA compared with the corre-
`sponding dNTP substrate [13, 23]. Two examples will
`
`be used to illustrate how RT is able to carry out this
`discrimination.
`K65R. The first example concerns the K65R mutation.
`As seen in table 1, this mutation confers some level of
`resistance to a variety of NRTIs, but is primarily recog-
`nized for viral cross-resistance to 3TC, ddC and ddI
`(ddA), as well as to PMEA [24–27]. In vitro studies
`with purified recombinant RT show that the K65R
`HIV-1 RT exhibits an eight-fold decrease in the affinity
`for ddCTP and ddATP, compared with only a twofold
`decrease in KM for dCTP and dATP [24, 28]. K65 is in
`the highly flexible b3– b4 loop in the fingers domain of
`the p66 subunit of RT, and the o-amino of K65 inter-
`acts with the g-phosphate of the bound dNTP substrate
`[20]. Molecular modeling studies indicate that the K65R
`
`Figure 4. View of the dNTP binding pocket in the RT*-T:P-dNTP ternary complex. The figure was created on Sybyl 6.5 using PDB
`coordinates 1RTD [20]. The RT residues shown are those that interact with the dNTP substrate (K65, R72, G112, D113, A114, Y115,
`Q151), the primer 3%-terminal nucleotide (Y183, M184), the complementary template nucleotide (L74, V75) and the catalytic aspartate
`triad (D110, D185, D186) with coordinated metals.
`
`

`

`CMLS, Cell. Mol. Life Sci. Vol. 57, 2000
`
`Review Article
`
`1415
`
`substitution induces packing rearrangements of the nu-
`cleotide substrate, leading to subtle differences in orien-
`tation of the phosphate backbone [28]. This leads to
`displacement of the base component of the nucleotide,
`which may hinder the positioning of the base for hydro-
`gen bonding to the complementary template base. But
`why is this effect more pronounced for the NRTI than
`the analogous dNTP? As previously discussed, the 3%-
`OH of the dNTP substrate makes significant interac-
`tions with the ‘3%-OH’ binding pocket in RT, and in
`particular to residue Q151. These interactions may facil-
`itate proper positioning of the dNTP base to allow base
`pairing with the template. NRTI lack a 3%-OH, and are
`therefore unable to contact the ‘3%-OH’ pocket on the
`enzyme. The absence of this positioning interaction
`makes it more difficult for the NRTI base to hydrogen-
`bond to the template, and the RT-bound NRTI is
`therefore more likely to adopt an orientation in the
`polymerase active site that is unfavorable for catalysis.
`Thus, the absence of the 3%-OH positioning interaction
`in NRTI binding, coupled with the subtle displacement
`of the NRTI base arising from the K65R substitution,
`can account for the selective decrease in RT affinity for
`ddCTP and ddATP relative to the dNTP substrates.
`The slightly decreased affinity of the K65R mutant for
`dATP and dCTP further increases the already existing
`bias of RT against NRTI (in this case ddCTP and
`ddATP) incorporation into the nascent viral DNA,
`resulting in a readily observable resistance phenotype.
`The K65R mutant RT possesses an increased processiv-
`ity in DNA synthesis relative to wild-type enzyme [29].
`This may compensate for the modest loss of affinity for
`dCTP and dATP exhibited by the mutant enzyme,
`thereby enabling normal replication kinetics.
`It is intriguing that the K65R mutation appears to
`confer selective resistance to some, but not all, NRTIs.
`For example, we demonstrated that whereas the K65R
`mutation provides cross-resistance to 3TC, ddC and ddI
`(ddA), the mutant is not resistant to either ddG or ddT.
`While the precise mechanism for this selective resistance
`is not yet known, a clue is obtained from crystal studies
`of the dNTP complexes with another DNA polymerase,
`the Klenow fragment of Thermus aquaticus DNA poly-
`merase I [30]. The binding of all the dNTP is due in part
`to interaction of the g- and a-phosphates of the dNTP
`with two basic amino acid residues (analogous to K65
`and R72 of HIV-1 RT). However, the base components
`of the dNTPs occupy different positions in the bound
`structures. The base rings of dCTP and dATP occupy
`similar positions in the bound structure, and have few
`contacts with protein residues. The base rings of dGTP
`and dTTP also occupy similar regions of space as well,
`but these differ from the regions occupied by the bases
`of dCTP and dATP. The base components of dGTP
`and dTTP make significant additional contacts with
`
`surrounding enzyme residues, which may stabilize the
`positioning of these dNTPs for incorporation. We pro-
`pose that a similar situation may exist for RT. The
`selective resistance to ddCTP and ddATP conferred by
`the K65R mutation results from (i) the loss of the
`3%-OH pocket interaction, and (ii) the lack of additional
`base-stabilizing contacts with surrounding RT residues.
`This results in an unacceptable flexibility in ddCTP and
`ddATP binding such that correct positioning for cata-
`lytic incorporation is impaired. In contrast, the flexibil-
`ity of ddGTP and ddTTP binding is reduced due to
`additional contacts of the base ring with RT residues.
`These NRTIs are more readily positioned for incorpo-
`ration into the growing DNA chain.
`We have recently compared the properties of recombi-
`nant RTs with a variety of amino acid substitutions at
`K65 [28]. Whereas the K65R mutation confers resis-
`tance only to specific NRTIs, other mutations such as
`K65A and K65Q result in broad resistance to all NR-
`TIs (i.e. multidrug resistance). So why does HIV not
`generate these mutations? These mutant RTs have
`severe impairments in the incorporation of the natural
`dNTP substrates as well. Our modeling experiments [28]
`suggest that only enzymes with K or R at position 65
`are able to make significant contacts with the g-phos-
`phate of the dNTP. The loss of this contact in the other
`position 65 mutants prevents the proper positioning of
`the dNTP for catalytic incorporation. Thus, HIV-1 is
`severely curtailed in the choice of mutations at this
`position.
`M184V (and M184I). The second example of discrimi-
`nation as a resistance phenotype concerns the point
`mutation M184V (sometimes M184I) that confers high-
`level resistance to 3TC [31, 32]. M184 is located in the
`YMDD motif, which is highly conserved among retro-
`viral RTs. In HIV-1 RT, this motif contains two of the
`three catalytic aspartates of the DNA polymerase active
`site [16–20]. The M184V RT is about 140-fold less
`efficient in incorporating 3TCTP into the nascent viral
`DNA compared with the wild-type enzyme [33]. This
`decreased incorporation efficiency results from steric
`hindrance that leads to decreased binding and:or inap-
`propriate positioning of the 3TCTP for catalysis. In the
`RT*-T:P-dNTP ternary complex of wild-type RT [20],
`the side chain of M184 contacts the sugar and the base
`of the 3% nucleotide in the primer. Molecular modeling
`experiments [20, 34] show that substitution of M184
`with an amino acid with a b-branched side chain
`(isoleucine or valine) results in an additional contact
`with the dNTP sugar moiety (fig. 5). The side chain of
`a b-branched amino acid at position 184 makes inap-
`propriate contact with the sulfur of the sugar oxathi-
`olane ring of 3TCTP, preventing proper positioning of
`3TCTP for catalysis, consistent with the data of Feng
`and Anderson [33]. The analogous substrate dCTP does
`
`

`

`1416
`
`N. Sluis-Cremer et al.
`
`HIV resistance to NRTI
`
`not have a sulfur in its sugar ring, and no steric hin-
`drance of dCTP binding is noted. In contrast, Krebs et
`al. [23], while demonstrating that the M184V mutant RT
`shows a markedly reduced efficiency of incorporation of
`3TCMP into viral DNA, found no differences in binding
`affinity of the wild-type and M184V mutant enzymes for
`3TCTP. These observations differ from those of Feng
`and Anderson, and may support the T:P repositioning
`model (see below). Thus, the precise mechanism by
`which the M184V:I mutations confer resistance to 3TC
`is still not completely certain. However, we feel that
`steric hindrance preventing correct positioning of
`3TCTP in the RT active site likely plays a major role in
`this resistance mechanism.
`
`Template:primer repositioning may, however, play a
`role in the decreased DNA synthesis processivity associ-
`ated with the L74V mutation for ddI resistance [36].
`However, this altered processivity does not necessarily
`contribute to the resistance phenotype. Whereas L74
`interacts with the template nucleotide that is base-
`paired to the incoming dNTP molecule, it is also proxi-
`mal
`to Q151 and R72,
`two residues
`that make
`significant contacts with the bound dNTP [20]. Thus the
`L74V mutation may alter ddNTP binding and:or catal-
`ysis by altering the packing rearrangements of ddNTP
`through its
`interactions with the aforementioned
`residues.
`
`Template:primer repositioning
`Boyer et al. [35] showed that wild-type RT was inhibited
`by ddITP when the template extension was four nucle-
`otides or more beyond the 3% end of the primer, but was
`resistant to ddITP when the template extension was
`three nucleotides or fewer [35]. In contrast, ddITP resis-
`tance shown by the L74V and E89G mutant enzymes
`was independent of template extension length. The RT-
`T:P binary complex crystal structure [18] suggested that
`residues 74 and 89 contacted the DNA T:P; the recently
`solved RT*-T:P-dNTP ternary complex [20] shows that
`L74 interacts with the template nucleotide that is base-
`paired to the incoming dNTP. Boyer et al. [35] therefore
`proposed that the L74V and E89G mutations (and
`perhaps others) confer resistance to NRTI as a result of
`repositioning of the T:P in the RT-T:P complex, which
`in turn alters the ability of the enzyme to select for, or
`reject an incoming dNTP. While there is no structural
`evidence for T:P repositioning in the L74V or E89G
`mutants, comparison of the M184I RT-T:P binary com-
`plex with the wild type RT-T:P binary complex reveals
`some alterations in the position of the T:P in the M184I
`mutant RT structure [34]. As discussed above, the data
`of Krebs et al. [23] provide circumstantial support for
`the T:P repositioning model in 3TC resistance. Nonethe-
`less, the relevance of T:P repositioning in phenotypic
`resistance to NRTI is uncertain. How could this reposi-
`tioning selectively alter incorporation of ddNTP, with-
`out also dramatically affecting incorporation of the
`corresponding dNTP substrate? In addition, it is not
`certain whether the shift in T:P position noted in the
`M184I RT-T:P binary complex would be retained in the
`subsequent RT-T:P-dNTP ternary complex. Finally, as
`discussed above, the phenotype for 3TC resistance is
`satisfactorily explained by the steric conflict between the
`oxathiolane ring of 3TCTP and the side chain of the
`branched b-amino acid. Therefore, there does not ap-
`pear to be a need to invoke T:P repositioning to explain
`this resistance.
`
`Phosphorolytic removal of incorporated
`chain-terminating NRTI—the novel mechanism of AZT
`resistance
`The efforts of investigators to elucidate the biochemical
`phenotype for HIV-1 resistance to AZT was compli-
`cated by the absence of any measurable effect on inhibi-
`tion of RT by AZTTP in vitro, despite the more than
`200-fold resistance to AZT demonstrated in cell culture
`[37, 38]. AZT resistance correlates with multiple muta-
`tions in RT, including M41L, D67N, K70R, L210W,
`T215Y:F and Q219K [39–42] (table 1). The extent of
`AZT resistanc

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket