You are on page 1of 16

2/17/2021 The impact of resistance on viral fitness and its clinical implications - Antiretroviral Resistance in Clinical Practice - NCBI

al Practice - NCBI Bookshelf

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Geretti AM, editor. Antiretroviral Resistance in Clinical Practice. London: Mediscript; 2006.

Chapter 12 The impact of resistance on viral fitness and its clinical


implications
Authors

Andrea De Luca.

Introduction
The primary goal of antiretroviral treatment is to durably suppress viral replication in order to promote immune
reconstitution and reduce AIDS-related morbidity and mortality. In HIV-1-infected patients with a history of treatment
failure, the selection of drug-resistant and multidrug-resistant virus may not allow complete suppression of viral
replication. In these patients, immunological stability is often observed despite ongoing viral replication, when drug-
resistant virus with lower fitness is selected by antiretroviral drug pressure [1]. In fact, when HIV-1 develops resistance
to antiretroviral drugs through the acquisition of mutations in the pol gene, it often pays a price in terms of reduced
replication capacity and reduced pathogenicity [2,3]. The deleterious effects on viral fitness associated with the
acquisition of drug resistance presumably result from mutation-induced structural changes in the binding of the natural
substrate and in the catalytic activity of the reverse transcriptase (RT) and protease [4,5]. However, the degree of
impairment of viral replication appears to vary widely among viral strains that are resistant to antiretroviral agents (see
Figure 1) [6,7]. Additional data suggest also that viral fitness varies among clinical isolates from drug-naive
individuals, reflecting natural genetic polymorphisms present in each viral strain [8].

Evidence to date clearly indicates that several drug-resistance mutations are associated with reduced ability of HIV-1 to
replicate. More recent data also suggest that, whereas drug susceptibility remains the most important determinant of
treatment response, reduced viral fitness can be exploited to derive a clinical benefit, especially in settings in which
patients have limited therapeutic alternatives.

Definition of HIV-1 fitness


Fitness refers to the ability of an organism to adapt and reproduce in a defined environment. Alterations in any of the
viral genes that play a key role in the replication cycle, as well as in the evasion of HIV-1 from the immune response or
from antiretroviral drug pressure, can affect viral fitness.

Methods to determine HIV-1 fitness


A number of methods have been developed to measure viral fitness in vitro, ex vivo or in vivo. These can be
categorised as follows:

Growth competition assays


These consist of competitive culture assays in which two viral variants are mixed and, after serial passage, the
proportions of the competing viruses are carefully measured over time using a variety of genotypic or phenotypic
techniques (e.g. recombinant marker virus assay and heteroduplex tracking assay) [9–11]. For example, a wild-type and
a drug-resistant mutant are cultured together at a defined wild-type:mutant ratio in the MT-2 cell line or in stimulated
peripheral blood mononuclear cells (PBMCs), and the relative amount of the drug-resistance mutation is measured
after several passages [12,13]. Competition experiments are currently the `gold standard' to measure viral fitness
because they estimate the replicative abilities of two viral strains under identical conditions. However, these assays are
labour intensive and take weeks to perform.

Assays of replication kinetics

https://www.ncbi.nlm.nih.gov/books/NBK2244/?report=printable 1/16
2/17/2021 The impact of resistance on viral fitness and its clinical implications - Antiretroviral Resistance in Clinical Practice - NCBI Bookshelf

These assays quantify the level of HIV-1 replication in parallel cultures [9,14]. Following infection of either cell lines
(e.g. MT-2 and MT-4) or PBMCs, the total replication capacity of each virus is determined by quantifying the
production of p24 antigen or the RT activity in cell culture supernatants at several time points [13,15,16]. The method
is relatively simple but suffers from lack of sensitivity in determining subtle differences in replication kinetics between
viruses.

Single-cycle replication assays


A fixed amount of recombinant virus is used to infect indicator cell lines that release an indicator enzyme upon
infection. To measure the infectious titre, the enzymatic activity in the supernatant is quantified after a single viral
replication cycle using an adequate substrate in a colorimetric assay. The ratio of mutant to wild-type infectivity
obtained from parallel assays is then calculated to obtain a fitness-related value in the absence or in the presence of
drugs at different concentrations [17]. This assay can be used both for measuring phenotypic drug susceptibility
(Phenoscript, Viralliance, France) and for measuring viral fitness in the absence of drugs.

Another single-cycle replication assay uses luciferase as the reporter gene and is a modification of an assay for the
determination of antiretroviral phenotypic susceptibility (PhenoSense; Monogram Biosciences, San Francisco, CA,
USA) [2,18]. The replication capacity is expressed as a percentage of a reference wild-type recombinant virus. The
assay can measure replication capacity in the absence or the presence of varying concentrations of drugs [19]. This
`replication capacity' assay does not measure viral fitness directly, but rather measures a component of viral fitness.
Because the assay is run using a recombinant vector containing a portion of the patient's virus, including the 3′ end of
gag, all of protease and most of RT, not all of the possible determinants of viral fitness are captured [20,21].

Indirect methods to estimate viral fitness


In addition to the aforementioned in vitro systems, viral fitness can be indirectly estimated in vivo by the relative
outgrowth of specific viral populations using calculations derived from standard population genetics theory [22]. For
example, Frost and colleagues calculated the relative fitness of M184V and M184I in vivo by using the relative rates of
outgrowth of these lamivudine (3TC)-resistant mutants during monotherapy with 3TC derived from the observed
frequency of resistant mutants over time and a mathematical model. The same can be done by calculating the time
needed for specific mutants to disappear after antiretroviral treatment interruption [23,24]. Under conditions that allow
ongoing replication of virus in the presence of drug pressure, wild-type virus acquired at the time of infection is
replaced as the dominant quasispecies by resistant variants of lower relative fitness. The dominance of the resistant
variant persists for as long as drug pressure continues because, in the presence of drugs, the resistant variant is the fitter
virus. However, once drug pressure is removed, the fitter wild-type virus re-emerges and eventually outgrows the
resistant variant. The time required for the wild-type variant to re-establish dominance is a function of a number of
variables, including the fitness difference between the two viruses and the size of the archived reservoir of wild-type
virus. For example, M184V, which is associated with 3TC resistance and has a signficant impact on viral fitness,
rapidly disappears from the dominant quasispecies after specific drug pressure is removed [24].

Genotypic drug resistance and viral fitness


Assessments of viral fitness using different techniques indicate an association between the accumulation of resistance
mutations and diminished fitness. In vitro kinetic and competition studies, as well as in vivo data, have established a
role for several resistance-associated mutations in reducing viral fitness. Table 1 shows the fitness effects associated
with the individual drug-resistance mutations cited by the International AIDS Society (IAS)-USA 2005 panel [25]. The
fitness effect of a combination of mutations was deliberately omitted from Table 1 in order to give preference to
findings on the fitness impact of individual mutations. One should consider several limitations when interpreting the
fitness data. Fitness values for a given mutation may vary depending on the presence of other drug-resistance mutations
or viral charactersistics, as well as on experimental conditions. In addition to defects in RT and in protease, other
elements influencing the fitness of a viral clone include the cell system used for the assay and the type of assay
employed (see above).

https://www.ncbi.nlm.nih.gov/books/NBK2244/?report=printable 2/16
2/17/2021 The impact of resistance on viral fitness and its clinical implications - Antiretroviral Resistance in Clinical Practice - NCBI Bookshelf

Nucleoside reverse transcriptase inhibitor-resistant mutations


Several nucleoside reverse transcriptase inhibitor (NRTI)-resistant mutations are associated with reduced viral fitness
(Table 1). The 3TC/emtricitabine resistance-associated mutations at codon 184 are clearly associated with reduced
fitness. Other mutations linked to reduced fitness are the thymidine analogue mutations (TAMs), especially the TAM
pattern 1 mutations (M41L, L210W and T215Y), the K65R and L74V mutations that emerge with the newer non-
thymidine NRTI regimens, and the multi-nucleoside resistance pattern associated with insertions at codon 69. However,
the TAM pattern 2 mutations (particularly D67N, K70R and K219Q/E, as T215F is less well characterised), which
usually confer less cross-resistance to NRTIs than the TAM pattern 1 mutations, do not differ significantly in fitness
from wild-type. In addition, the 215 `revertant' mutations T215D/S do not appear to reduce viral fitness and show a
better fitness than T215Y in the absence of drug pressure. Finally, the Q151M multi-NRTI resistance pattern does not
seem to affect viral fitness significantly.

Non-nucleoside reverse transcriptase inhibitor-resistant mutations


Resistance mutations selected by available non-nucleoside reverse transcriptase inhibitors (NNRTIs) generally have a
modest impact on viral fitness (Table 2). These in vitro data are confirmed by the in vivo finding of a slow
disappearance of these mutations from the dominant quasispecies after interruption of NNRTI-based therapy. However,
differences between mutations can be observed within this class. The two most common resistant mutants, K103N and
Y181C, show a fitness that is very close to that of wild-type virus. Less-frequent mutations, such as V106A, Y188C
and G190S, are associated with lower viral fitness (for more details, see Table 2 and references therein).

Protease inhibitor-resistant mutations


The impact on fitness of individual resistance-associated mutations in the protease region is difficult to explore because
viral mutants often contain multiple changes at both major and minor resistance sites. For example, growth competition
experiments have shown that strains with the mutation D30N, which is associated with resistance to nelfinavir, are
outcompeted by wild-type strains, whereas mutants with L90M, usually selected by saquinavir, are less fit than wild -
type but usually outgrow D30N mutants [13]. Combinations of drug-selected mutations in protease have variable
effects on viral fitness [9,19]. Table 3 shows results from studies to investigate the impact of single mutations.
Substantial reductions in viral fitness have been observed in clinical isolates and/or molecular clones containing the
mutations D30N, G48V, I50V and V82A/T. In addition, M46I/L, I54V, I84V, N88D/S and L90M are also associated
with significant reductions in fitness.

Glycoprotein gp41 resistance mutations


Preliminary observations have shown that enfuvirtide-resistance mutations in the first heptad repeat (HR-1) coding
region of the gp41 gene are associated with reduced viral fitness (see Table 3). In vitro data are confirmed by the in
vivo observation of prompt disappearance of the mutant virus from the dominant quasispecies after drug
discontinuation. The mechanism thought to be responsible for the reduced fitness is a reduction in viral infectivity as a
result of delayed and reduced efficiency in the fusion process [26].

Other mutations associated with reduced viral fitness


A number of other mutations in RT are associated with reduced fitness. These include rare resistance mutations not
included in the IAS-USA list (such as the deletion at codon 67 or V179D), including amino acid substitutions not
conferring drug resistance, which occurs at codons involved in drug resistance (such as Q151L/K and K70E) and
mutations at sites not known to be associated with resistance (E89K, L92I, E138X and S156A). In the protease,
insertions at codons 17, 35 and 95 have been found to confer reduced viral fitness [27]. Polymorphisms at codons 14
and 43, and the R41N mutation have been associated with low replication capacity using the Phenosense assay [28].

Compensatory mutations

https://www.ncbi.nlm.nih.gov/books/NBK2244/?report=printable 3/16
2/17/2021 The impact of resistance on viral fitness and its clinical implications - Antiretroviral Resistance in Clinical Practice - NCBI Bookshelf

A number of mutations have been found to restore viral fitness of drug-resistant mutants. In the RT, S68G [29] and
M230I [30] have both been associated with recovery of viral fitness in the presence of other mutations that impair
fitness. Most studies, however, have focused on the mechanisms leading to increased fitness during therapy with
protease inhibitors. It has been found that several mutations in the protease gene, including substitutions at codons 10I,
63P, 71V and 77I [15,31], compensate for the fitness loss associated with major protease-resistant mutations.

Other determinants of viral fitness


Viral replication capacity is also influenced by polymorphisms located outside the therapy target-coding regions. The
envelope (env) gene, which is linked to multiple viral characteristics such as entry into the host cell, transmission, co-
receptor usage and tropism, plays a major role in fitness of wild-type HIV-1 [32,33]. In particular, Marozsan and
coauthors have reported a higher avidity to the cell receptors CD4 and CCR5 mediated by the gp120 coding region of
the env gene in isolates with increased fitness. This was associated with an increased binding/fusion, as well as
decreased sensitivity to entry inhibitors. By producing chimeric env viruses with common HIV-1 genetic backbones,
Marozsan et al. have confirmed that the gp120 region of the HIV-1 env gene alone was sufficient to explain the fitness
of the entire primary HIV-1 strain from which it was derived. This finding implies that, in wild-type isolates, viral
entry, rather than other replication steps, is the major determinant of viral fitness. How much the variability of env will
determine the fitness of drug-resistant HIV-1 strains remains to be established.

Changes in gag also play a role in restoring viral fitness. Mutations at cleavage sites in gag (between p7 and p1)
increase the processing of the gag polyprotein [34,35], whereas mutations in other regions of gag render the cleavage
sites more accessible to the viral protease [36,37] and mutations of the frameshift signal lead to increased protease
synthesis [38]. Additional changes associated with increased fitness occur at the polypeptide region adjacent to the
protease coding region [39] and in the primer-binding-site loop of the 5′-end of the non-coding region [40].

Viral fitness and therapeutic response: potential clinical applications

Viral fitness as a predictor of clinical outcomes

The potential benefit of measuring HIV-1 fitness for predicting clinical outcome is under investigation both in drug-
naive and in drug-experienced patients failing therapy.

The Hemophilia Growth and Development Study [41] enrolled 207 HIV-1-infected haemophilia patients between 6 and
19 years of age in 1989 and 1990. Among these, 128 were assessed every 6 months during 1997 using the Phenosense
replication capacity assay. The baseline replication capacity correlated with the baseline plasma HIV-1 RNA load
(r=0.189; P =0.03), was inversely related to the CD4 count (r = −0.197; P =0.03), and independently predicted the rate
of decline in CD4 count and progression to AIDS.

Barbour et al. [42] studied 191 drug-naive subjects who had recently been diagnosed with HIV-1 infection and found
that the viral replication capacity, measured using the Phenosense assay, was associated with CD4 count at study entry
and over time, both before and after treatment initiation. The significant association between replication capacity and
CD4 count persisted after adjustment for drug resistance.

In a study of patients with low-level virological failure (viral load, <5000 copies/ml) on protease inhibitor-based
therapy, a significantly greater replication capacity (Phenosense) was found among patients with declining CD4 counts
(mean replication capacity, 22%) than among patients with stable or increasing CD4 counts (mean replication capacity,
12%; P =0.04) [43]. Despite the limited size of the study, the data were consistent with prior observations suggesting
an inverse relationship between replication capacity and CD4 count [2].

In an analysis [44] of 207 patients from the CCTG 575 trial [45], the baseline replication capacity (Phenosense) was
the strongest predictor of an increase in CD4 count from pre-study nadir to study baseline in multivariate models (P
=0.0004). A similar association with replication capacity was noted for the change in CD4 count from nadir to months
6 and 12 of the study (P <0.002). A decrease in replication capacity of 1 log10 unit at baseline led to an average rise in
CD4 count from nadir of 82 cells/μl. Furthermore, among 97 patients with virological failure at month 6 of the study,
https://www.ncbi.nlm.nih.gov/books/NBK2244/?report=printable 4/16
2/17/2021 The impact of resistance on viral fitness and its clinical implications - Antiretroviral Resistance in Clinical Practice - NCBI Bookshelf

the median change in viral load from baseline to month 6 was −0.54 log10 copies/ml in patients with a replication
capacity below 35% (n =49) as compared with +0.08 log10 copies/ml in patients with baseline replication capacity
values greater than 35% (n =48; P=0.0003) [44].

In a longitudinal observational study of patients maintained on failing protease inhibitor-based regimens for a median
period of 26 months, gradual increases in plasma viral load and phenotypic resistance to protease inhibitors were
observed, whereas the replication capacity (Phenosense) remained stable and the CD4 count increased gradually [21].
Emergence of compensatory mutations occurred relatively slowly over time and did not lead to a recovery of
replication capacity to the levels observed with wild-type virus.

De Luca et al. analysed 139 patients from the Argenta trial with an extended follow-up of 36 months. Patients had
experienced a median of two previous highly active antiretroviral therapy (HAART) regimens and received a median
of 18 months of HAART [7]. The median baseline replication capacity, as measured by the Phenosense assay, was 59%
(see Figure 1) and correlated positively with the number of phenotypically active drugs (r =0.32; P <0.001). Overall,
replication capacity did not predict treatment outcome. However, in a subset of 85 patients who remained viraemic
(viral load >500 copies/ml at each time point), a higher replication capacity predicted less-profound changes in viral
load at 3 months. In 25 patients with a phenotypic susceptibility score (PSS) of 3 for the first salvage regimen (i.e. a
first salvage regimen containing three drugs to which the patients' virus was shown to be fully susceptible), a higher
replication capacity predicted less pronounced viral load responses at 3 months. In persistently viraemic patients, after
adjusting for PSS, a higher baseline replication capacity was an independent predictor of lower gains in CD4 count at
months 3, 9, 12 and 24. The conclusion of this work was that, in patients in whom viral suppression cannot be
achieved, replication capacity can be a useful tool, after resistance testing, to guide treatment decisions.

Viral fitness and treatment interruption studies


Several studies have exploited the strategy of partial treatment interruptions in patients with virological failure to
identify the residual antiviral activity of different components of a regimen. In one study [46], patients interrupting
treatment with protease inhibitors and continuing NRTI therapy showed a stable viral load. The pre-existing protease-
resistance mutations decreased and replication capacity, as well as viral load, increased only after long-term
observation. This means that, in the presence of NRTI drugs, significant antiviral activity persisted that kept viral
replication partially under control, selecting for a viral isolate with reduced fitness for a significant period of time. The
HIV-1 RNA levels also remained stable in subjects interrupting NNRTI treatment, indicating lack of residual antiviral
activity of available NNRTIs after the emergence of resistance, consistent with the lack of a significant impact on viral
fitness of common NNRTI-associated resistance mutations. In contrast, all subjects who interrupted NRTI treatment
exhibited immediate increases in viral load with loss of NRTI-resistance mutations. Therefore, NRTIs often exert
continued antiviral activity in the setting of drug-resistance mutations, and both NRTIs and protease inhibitors can
select for drug-resistance mutations that reduce viral fitness [46]. In another study [47], patients with the M184V
mutation were randomly assigned to discontinue therapy or continue monotherapy with 3TC (the drug that selects for
M184V). At 48 weeks, patients assigned to 3TC continued to carry M184V and had a less pronounced viral load
increase and CD4 cell loss compared with those interrupting therapy, who had a concurrent rapid loss of M184V and
steeper decrease in CD4 count. Patients in the 3TC arm also maintained other drug-resistance mutations as well as
significantly reduced viral fitness as compared to the control arm. This implies that, despite high-level resistance, 3TC
continues to exert clinically significant antiviral activity in the presence of virus of lower fitness with M184V and other
resistance mutations selected by previous drug treatments.

Although data regarding the clinical utility of fitness measurements are still preliminary, certain consistent findings
have emerged from the studies reported to date. In patients failing antiretroviral therapy, as well as in untreated, acutely
or chronically infected patients, detection of virus with lower fitness is associated with higher CD4 counts. Lower
fitness is also associated with lower viral load during treatment failure. In addition, in patients with virological failure,
low viral fitness is associated with stable or increasing CD4 counts, despite increasing viral load. Low viral fitness and
stable CD4 counts appear to be maintained over several years of follow-up.

https://www.ncbi.nlm.nih.gov/books/NBK2244/?report=printable 5/16
2/17/2021 The impact of resistance on viral fitness and its clinical implications - Antiretroviral Resistance in Clinical Practice - NCBI Bookshelf

Conclusions
The fitness, or replication capacity, of HIV-1 is an intrinsic viral characteristic that has multiple viral determinants and
several virological and clinical correlates. Although reduced viral fitness is typically associated with the emergence of
resistance mutations in the targets of antiretroviral therapy, wild-type strains of HIV-1 can also demonstrate reduced
fitness. Despite numerous studies, the genotypic correlates of viral fitness remain to be fully characterised. The fitness
cost associated with the development of antiretroviral resistance is probably responsible for the sustained
immunological benefit that is often observed in patients with failure of HAART and virological breakthrough. Further
studies are required to define strategies to exploit optimally these fitness effects of drug resistance in order to at least
partially prolong the effect of treatment.

Recommendations for clinical practice

Virological suppression remains the primary goal of treatment. This goal should be pursued in all cases, where
clinically feasible, by selecting active drugs for salvage therapy using a resistance assay.

In multidrug-experienced patients with limited or no drug options available, one management option might be to
continue the failing therapy if the virus has low replication capacity/fitness.

Patients failing therapy with a virus of high replication capacity/fitness may benefit from treatment change, even
if there are few available options, in order to attempt to select for a virus with lower fitness.

The Phenosense replication capacity assay is the only assay currently available for clinical practice.
Nevertheless, no study has been carried out to show whether using this assay confers more benefit than using
surrogate markers of fitness, such as viral load and/or CD4 count, to guide therapy management in clinical
practice. Notwithstanding, the fact that the replication capacity assay is able to predict the subsequent viral load
and CD4 count, implies that it is an early marker that could be used to anticipate and more favourably drive
treatment outcome.

In the absence of a viral fitness assay in a patient with a low CD4 count who has failed multidrug treatment, it
seems reasonable to select the treatment that, with acceptable tolerability, will contain the highest possible
number of active drugs, together with drugs that tend to select for mutations associated with reduced viral fitness.

References
1. Deeks SG, Hoh R, Neilands TB. et al. Sustained CD4+ T cell response after virologic failure of protease inhibitor-
based regimens in patients with human immunodeficiency virus infection. J Infect Dis. 2000;181:946–953.
[PubMed: 10720517]
2. Deeks SG, Wrin T, Liegler T. et al. Virologic and immunologic consequences of discontinuing combination
antiretroviral drug therapy in HIV infected patients with detectable viremia. N Engl J Med. 2001;344:472–480.
[PubMed: 11172188]
3. Prado JG, Parkin NT, Clotet B. et al. HIV type 1 fitness evolution in antiretroviral-experienced patients with
sustained CD4+ T cell counts but persistent virologic failure. Clin Infect Dis. 2005;41:729–737. [PubMed:
16080097]
4. Dauber DS, Ziermann R, Parkin N. et al. Altered substrate specificity of drug-resistant human immunodeficiency
virus type 1 protease. J Virol. 2002;76:1359–1368. [PMC free article: PMC135855] [PubMed: 11773410]
5. Malim MH, Emerman M. HIV-1 sequence variation: drift, shift and attenuation. Cell. 2001;104:469–472.
[PubMed: 11239404]
6. Cingolani A, Antinori A, Rizzo MG. et al. Usefulness of monitoring HIV drug resistance and adherence in
individuals failing highly active antiretroviral therapy: a randomized study (ARGENTA). AIDS. 2002;16:369–379.
[PubMed: 11834948]
7. De Luca A, Bates M, Di Giambenedetto S et al. HIV-1 replication capacity (RC) in HAART-failing patients
predicts virologic and immunologic responses when accounting for viral susceptibility to the salvage regimen:

https://www.ncbi.nlm.nih.gov/books/NBK2244/?report=printable 6/16
2/17/2021 The impact of resistance on viral fitness and its clinical implications - Antiretroviral Resistance in Clinical Practice - NCBI Bookshelf

results from the Argenta trial. 10th Conference on Retroviruses and Opportunistic Infections, Boston, 2005, Abstr.
692.
8. Wrin T, Gamarnik AV, Whitehurst N. et al. Natural variation of replication capacity measurements in drug-
naïve/susceptible HIV-1. Antivir Ther. 2001;6(suppl 1):20.
9. Resch W, Ziermann R, Parkin N. et al. Nelfinavir-resistant, amprenavir-hypersusceptible strains of human
immunodeficiency virus type 1 carrying an N88S mutation in protease have reduced infectivity, reduced replication
capacity, and reduced fitness and process the Gag polyprotein precursor aberrantly. J Virol. 2002;76:8659–8666.
[PMC free article: PMC136408] [PubMed: 12163585]
10. Lu J, Kuritzkes DR. A novel recombinant marker virus assay for comparing the relative fitness of hiv-1 reverse
transcriptase variants. J Acquir Immune Defic Syndr. 2001;27:7–13. [PubMed: 11404514]
11. Quinones-Mateu ME, Ball SC, Marozsan AJ. et al. A dual infection/competition assay shows a correlation
between ex vivo human immunodeficiency virus type 1 fitness and disease progression. J Virol. 2000;74:9222–
9233. [PMC free article: PMC102121] [PubMed: 10982369]
12. Kosalaraksa P, Kavlick MF, Maroun V. et al. Comparative fitness of multi-dideoxynucleoside-resistant human
immunodeficiency virus type 1 (HIV-1) in an in vitro competitive HIV-1 replication assay. J Virol. 1999;73:5356–
5363. [PMC free article: PMC112591] [PubMed: 10364282]
13. Martinez-Picado J, Savara AV, Sutton L, D'Aquila RT. Replicative fitness of protease inhibitor-resistant mutants
of human immunodeficiency virus type 1. J Virol. 1999;73:3744–3752. [PMC free article: PMC104151]
[PubMed: 10196268]
14. Clavel F, Race E, Mammano F. HIV drug resistance and viral fitness. Adv Pharmacol. 2000;49:41–99. [PubMed:
11013760]
15. Markowitz M, Mo H, Kempf DJ. et al. Selection and analysis of human immunodeficiency virus type 1 variants
with increased resistance to ABT-538, a novel protease inhibitor. J Virol. 1995;69:701–706. [PMC free article:
PMC188631] [PubMed: 7815532]
16. Nicastri E, Sarmati L, d'Ettorre G. et al. Replication capacity, biological phenotype, and drug resistance of HIV
strains isolated from patients failing antiretroviral therapy. J Med Virol. 2003;69:1–6. [PubMed: 12436471]
17. Mammano F, Trouplin V, Zennou V. et al. Retracing the evolutionary pathways of human immunodeficiency virus
type 1 resistance to protease inhibitors: virus fitness in the absence and in the presence of drug. J Virol.
2000;74:8524–8531. [PMC free article: PMC116364] [PubMed: 10954553]
18. Petropoulos CJ, Parkin NT, Limoli K. et al. A novel phenotypic drug susceptibility assay for HIV-1. Antimicrob
Agents Chemother. 2000;44:920–928. [PMC free article: PMC89793] [PubMed: 10722492]
19. Prado JG, Wrin T, Beauchaine J. et al. Amprenavir-resistant HIV-1 exhibits lopinavir cross-resistance and reduced
replication capacity. AIDS. 2002;16:1009–1017. [PubMed: 11953467]
20. Bates M, Wrin T, Huang W. et al. Practical applications of viral fitness in clinical practice. Curr Opin Infect Dis.
2003;16:11–18. [PubMed: 12821824]
21. Barbour JD, Wrin T, Grant RM. et al. Evolution of phenotypic drug susceptibility and viral replication capacity
during long-term virologic failure of protease inhibitor therapy in human immunodeficiency virus-infected adults.
J Virol. 2002;76:11104–11112. [PMC free article: PMC136622] [PubMed: 12368352]
22. Frost SD, Nijhuis M, Schuurman R. et al. Evolution of lamivudine resistance in human immunodeficiency virus
type 1-infected individuals: the relative roles of drift and selection. J Virol. 2000;74:6262–6268. [PMC free
article: PMC112131] [PubMed: 10864635]
23. Joly V, Descamps D, Peytavin G. et al. Evolution of human immunodeficiency virus type 1 (HIV-1) resistance
mutations in nonnucleoside reverse transcriptase inhibitors (NNRTIs) in HIV-1-infected patients switched to
antiretroviral therapy without NNRTIs. Antimicrob Agents Chemother. 2004;48:172–175. [PMC free article:
PMC310183] [PubMed: 14693536]
24. Zaccarelli M, Perno CF, Forbici F. et al. Using a database of HIV patients undergoing genotypic resistance test
after HAART failure to understand the dynamics of M184V mutation. Antivir Ther. 2003;8:51–56. [PubMed:
12713064]
25. Johnson VA, Brun-Vezinet F, Clotet B. et al. Update of the drug resistance mutations in HIV-1: fall 2005. Top
HIV Med. 2005;13:125–131. [PubMed: 16304457]

https://www.ncbi.nlm.nih.gov/books/NBK2244/?report=printable 7/16
2/17/2021 The impact of resistance on viral fitness and its clinical implications - Antiretroviral Resistance in Clinical Practice - NCBI Bookshelf

26. Reeves JD, Lee FH, Miamidian JL. et al. Enfuvirtide resistance mutations: impact on human immunodeficiency virus
envelope function, entry inhibitor sensitivity, and virus neutralization. J Virol. 2005;79:4991–4999. [PMC free article:
PMC1069568] [PubMed: 15795284]
27. Kim EY, Winters MA, Kagan RM. et al. Functional correlates of insertion mutations in the protease gene of
human immunodeficiency virus type 1 isolates from patients. J Virol. 2001;75:11227–11233. [PMC free article:
PMC114703] [PubMed: 11602763]
28. Bates M, Chappey C, Bates S et al. Influence of mutations in protease and the C-terminal end of Gag on
replication capacity in drug sensitive clade B HIV-1. 43rd Interscience Conference on Antimicrobial Agents and
Chemotherapy, Chicago, 2003, Abstr. H-812.
29. Garcia-Lerma JG, Gerrish PJ, Wright AC. et al. Evidence of a role for the Q151L mutation and the viral
background in development of multiple dideoxynucleoside-resistant human immunodeficiency virus type 1. J
Virol. 2000;74:9339–9346. [PMC free article: PMC112361] [PubMed: 11000201]
30. Matsumi S, Kosalaraksa P, Tsang H. et al. Pathways for the emergence of multi-dideoxynucleoside-resistant HIV-
1 variants. AIDS. 2003;17:1127–1137. [PubMed: 12819513]
31. Nijhuis M, Schuurman R, de Jong D. et al. Increased fitness of drug resistant HIV-1 protease as a result of
acquisition of compensatory mutations during suboptimal therapy. AIDS. 1999;13:2349–2359. [PubMed:
10597776]
32. Rangel HR, Weber J, Chakraborty B. et al. Role of the human immunodeficiency virus type 1 envelope gene in
viral fitness. J Virol. 2003;77:9069–9073. [PMC free article: PMC167250] [PubMed: 12885922]
33. Marozsan AJ, Moore DM, Lobritz MA. et al. Differences in the fitness of two diverse wild-type human
immunodeficiency virus type 1 isolates are related to the efficiency of cell binding and entry. J Virol.
2005;79:7121–7134. [PMC free article: PMC1112120] [PubMed: 15890952]
34. Zhang YM, Imamichi H, Imamichi T. et al. Drug resistance during indinavir therapy is caused by mutations in the
protease gene and in its Gag substrate cleavage sites. J Virol. 1997;71:6662–6670. [PMC free article:
PMC191944] [PubMed: 9261388]
35. Pettit SC, Henderson GJ, Schiffer CA, Swanstrom R. Replacement of the P1 amino acid of human
immunodeficiency virus type 1 Gag processing sites can inhibit or enhance the rate of cleavage by the viral
protease. J Virol. 2002;76:10226–10233. [PMC free article: PMC136535] [PubMed: 12239298]
36. Gatanaga H, Suzuki Y, Tsang H. et al. Amino acid substitutions in Gag protein at non-cleavage sites are
indispensable for the development of a high multitude of HIV-1 resistance against protease inhibitors. J Biol
Chem. 2002;277:5952–5961. [PubMed: 11741936]
37. Myint L, Matsuda M, Matsuda Z. et al. Gag non-cleavage site mutations contribute to full recovery of viral fitness
in protease inhibitor-resistant human immunodeficiency virus type 1. Antimicrob Agents Chemother.
2004;48:444–452. [PMC free article: PMC321554] [PubMed: 14742193]
38. Doyon L, Payant C, Brakier-Gingras L, Lamarre D. Novel Gag-Pol frameshift site in human immunodeficiency
virus type 1 variants resistant to protease inhibitors. J Virol. 1998;72:6146–6150. [PMC free article: PMC110421]
[PubMed: 9621079]
39. Dautin N, Karimova G, Ladant D. Human immunodeficiency virus (HIV) type 1 transframe protein can restore
activity to a dimerization-deficient HIV protease variant. J Virol. 2003;77:8216–8226. [PMC free article:
PMC165233] [PubMed: 12857890]
40. Yuste E, Borderia AV, Domingo E. et al. Few mutations in the 5' leader region mediate fitness recovery of
debilitated human immunodeficiency type 1 viruses. J Virol. 2005;79:5421–5427. [PMC free article:
PMC1082768] [PubMed: 15827156]
41. Daar ES, Kesler KL, Wrin T. et al. Hemophilia Growth and Development Study. HIV-1 pol replication capacity
predicts disease progression. AIDS. 2005;19:871–877. [PubMed: 15905667]
42. Barbour JD, Hecht FM, Wrin T. et al. Higher CD4+ T cell counts associated with low viral pol replication
capacity among treatment-naive adults in early HIV-1 infection. J Infect Dis. 2004;190:251–256. [PubMed:
15216458]
43. Sufka SA, Ferrari G, Gryszowka VE. et al. Prolonged CD4+ cell/virus load discordance during treatment with
protease inhibitor-based highly active antiretroviral therapy: immune response and viral control. J Infect Dis.

https://www.ncbi.nlm.nih.gov/books/NBK2244/?report=printable 8/16
2/17/2021 The impact of resistance on viral fitness and its clinical implications - Antiretroviral Resistance in Clinical Practice - NCBI Bookshelf

2003;187:1027–1037. [PubMed: 12660916]


44. Haubrich R, Wrin T, Hellmann N. et al. Replication capacity (RC) as a predictor of immunologic and virologic
benefit despite virologic failure of an antiretroviral regimen. Antivir Ther. 2002;7(suppl 1):S133.
45. Haubrich RH, Kemper CA, Hellmann NS. et al. A randomized, prospective study of phenotype susceptibility
testing versus standard of care to manage antiretroviral therapy: CCTG 575. AIDS. 2005;19:295–302. [PubMed:
15718840]
46. Deeks SG, Barbour JD, Martin JN. et al. Interruption of treatment with individual therapeutic drug classes in
adults with multidrug-resistant HIV-1 infection. J Infect Dis. 2005;192:1537–1544. [PubMed: 16206068]
47. Castagna A, Danise A, Menzo S et al. E-184V study. Lamivudine monotherapy vs treatment interruption in
failing HIV-1 infected subjects, harbouring the M184V mutation: 48-week final results. 3rd IAS Conference on
HIV Pathogenesis and Treatment, Rio de Janeiro, 2005, Abstr. WeFb0204.
48. Harrigan PR, Kinghorn I, Bloor S. et al. Significance of amino acid variation at human immunodeficiency virus
type 1 reverse transcriptase residue 210 for zidovudine susceptibility. J Virol. 1996;70:5930–5934. [PMC free
article: PMC190612] [PubMed: 8709214]
49. Harrigan PR, Bloor S, Larder BA. Relative replicative fitness of zidovudine-resistant human immunodeficiency
virus type 1 isolates in vitro. J Virol. 1998;72:3773–3778. [PMC free article: PMC109599] [PubMed: 9557659]
50. Sharma PL, Crumpacker CS. Attenuated replication of human immunodeficiency virus type 1 with a didanosine-
selected reverse transcriptase mutation. J Virol. 1997;71:8846–8851. [PMC free article: PMC192351] [PubMed:
9343245]
51. de Jong MD, Veenstra J, Stilianakis NI. et al. Host-parasite dynamics and outgrowth of virus containing a single
K70R amino acid change in reverse transcriptase are responsible for the loss of human immunodeficiency virus
type 1 RNA load suppression by zidovudine. Proc Natl Acad Sci U S A. 1996;93:5501–5506. [PMC free article:
PMC39275] [PubMed: 8643604]
52. Garcia-Lerma JG, MacInnes H, Bennett D. et al. Transmitted human immunodeficiency virus type 1 carrying the
D67N or K219Q/E mutation evolves rapidly to zidovudine resistance in vitro and shows a high replicative fitness
in the presence of zidovudine. J Virol. 2004;78:7545–7552. [PMC free article: PMC434071] [PubMed: 15220429]
53. de Ronde A, van Dooren M, van Der Hoek L. et al. Establishment of new transmissible and drug-sensitive human
immunodeficiency virus type 1 wild types due to transmission of nucleoside analogue-resistant virus. J Virol.
2001;75:595–602. [PMC free article: PMC113955] [PubMed: 11134272]
54. Goudsmit J, de Ronde A, de Rooij E, de Boer R. Broad spectrum of in vivo fitness of human immunodeficiency
virus type 1 subpopulations differing at reverse transcriptase codons 41 and 215. J Virol. 1997;71:4479–4484.
[PMC free article: PMC191667] [PubMed: 9151839]
55. Yerly S, Rakik A, De Loes SK. et al. Switch to unusual amino acids at codon 215 of the human immunodeficiency
virus type 1 reverse transcriptase gene in seroconvertors infected with zidovudine-resistant variants. J Virol.
1998;72:3520–3523. [PMC free article: PMC109570] [PubMed: 9557630]
56. Harrigan PR, Stone C, Griffin P. et al. Resistance profile of the human immunodeficiency virus type 1 reverse
transcriptase inhibitor abacavir (1592U89) after monotherapy and combination therapy. CNA2001 Investigative
Group. J Infect Dis. 2000;181:912–920. [PubMed: 10720512]
57. Miller M, White KL, Petropoulos C and Parkin NT. Decreased replication capacity of HIV-1 clinical isolates
containing K65R or M184V RT mutations. 10th Conference on Retroviruses and Opportunistic Infections, 2003,
Boston, Abstr. 616.
58. Deval J, Navarro JM, Selmi B. et al. A loss of viral replicative capacity correlates with altered DNA
polymerization kinetics by the human immunodeficiency virus reverse transcriptase bearing the K65R and L74V
dideoxynucleoside resistance substitutions. J Biol Chem. 2004;279:25489–25496. [PubMed: 15044478]
59. Sharma PL, Crumpacker CS. Decreased processivity of human immunodeficiency virus type 1 reverse
transcriptase (RT) containing didanosine-selected mutation Leu74Val: a comparative analysis of RT variants
Leu74Val and lamivudine-selected Met184V. J Virol. 1999;73:8448–8456. [PMC free article: PMC112864]
[PubMed: 10482597]
60. Weber J, Chakraborty B, Weberova J. et al. Diminished replicative fitness of primary human immunodeficiency
virus type 1 isolates harboring the K65R mutation. J Clin Microbiol. 2005;43:1395–1400. [PMC free article:

https://www.ncbi.nlm.nih.gov/books/NBK2244/?report=printable 9/16
2/17/2021 The impact of resistance on viral fitness and its clinical implications - Antiretroviral Resistance in Clinical Practice - NCBI Bookshelf

PMC1081293] [PubMed: 15750116]


61. White KL, Margot NA, Wrin T. et al. Molecular mechanisms of resistance to human immunodeficiency virus type
1 with reverse transcriptase mutations K65R and K65R+M184V and their effects on enzyme function and viral
replication capacity. Antimicrob Agents Chemother. 2002;46:3437–3446. [PMC free article: PMC128721]
[PubMed: 12384348]
62. Quinones-Mateu ME, Tadele M, Parera M. et al. Insertions in the reverse transcriptase increase both drug
resistance and viral fitness in a human immunodeficiency virus type 1 isolate harboring the multi-nucleoside
reverse transcriptase inhibitor resistance 69 insertion complex mutation. J Virol. 2002;76:10546–10552. [PMC
free article: PMC136580] [PubMed: 12239335]
63. Prado JG, Franco S, Matamoros T. et al. Relative replication fitness of multi-nucleoside analogue-resistant HIV-1
strains bearing a dipeptide insertion in the fingers subdomain of the reverse transcriptase and mutations at codons
67 and 215. Virology. 2004;326:103–112. [PubMed: 15262499]
64. Maeda Y, Venzon DJ, Mitsuya H. Altered drug sensitivity, fitness, and evolution of human immunodeficiency
virus type 1 with pol gene mutations conferring multi-dideoxynucleoside resistance. J Infect Dis. 1998;177:1207–
1213. [PubMed: 9593005]
65. Iversen AK, Shafer RW, Wehrly K. et al. Multidrug-resistant human immunodeficiency virus type 1 strains
resulting from combination antiretroviral therapy. J Virol. 1996;70:1086–1090. [PMC free article: PMC189915]
[PubMed: 8551567]
66. Rayner MM, Cordova B, Jackson DA. Population dynamics studies of wild-type and drug-resistant mutant HIV in
mixed infections. Virology. 1997;236:85–94. [PubMed: 9299620]
67. Imamichi T, Berg SC, Imamichi H. et al. Relative replication fitness of a high-level. J Virol. 2000;74:10958–
10964. [PMC free article: PMC113175] [PubMed: 11069990]
68. Collins JA, Thompson MG, Paintsil E. et al. Competitive fitness of nevirapine-resistant human immunodeficiency
virus type 1 mutants. J Virol. 2004;78:603–611. [PMC free article: PMC368761] [PubMed: 14694092]
69. Archer RH, Dykes C, Gerondelis P. et al. Mutants of human immunodeficiency virus type 1 (HIV-1) reverse
transcriptase resistant to nonnucleoside reverse transcriptase inhibitors demonstrate altered rates of RNase H
cleavage that correlate with HIV-1 replication fitness in cell culture. J Virol. 2000;74:8390–8401. [PMC free
article: PMC116350] [PubMed: 10954539]
70. Huang W, Wrin T, Gamarnik AV. et al. RT mutations that confer NNRTI resistance may also impair replication
capacity. Antivir Ther. 2002;7(suppl 1):S79.
71. Iglesias-Ussel MD, Casado C, Yuste E. et al. In vitro analysis of human immunodeficiency virus type 1 resistance
to nevirapine and fitness determination of resistant variants. J Gen Virol. 2002;83:93–101. [PubMed: 11752705]
72. Antinori A, Liuzzi G, Cingolani A. et al. Drug-resistant mutants of HIV-1 in patients exhibiting increasing CD4
cell count despite virological failure of highly active antiretroviral therapy. AIDS. 2001;15:2325–2327. [PubMed:
11698709]
73. Huang W, Gamarnik A, Limoli K. et al. Amino acid substitutions at position 190 of human immunodeficiency
virus type 1 reverse transcriptase increase susceptibility to delavirdine and impair virus replication. J Virol.
2003;77:1512–1523. [PMC free article: PMC140843] [PubMed: 12502865]
74. Gerondelis P, Archer RH, Palaniappan C. et al. The P236L delavirdine-resistant human immunodeficiency virus
type 1 mutant is replication defective and demonstrates alterations in both RNA 5′-end- and DNA 3′-end-directed
RNase H activities. J Virol. 1999;73:5803–5813. [PMC free article: PMC112641] [PubMed: 10364332]
75. Menzo S, Monachetti A, Balotta C. et al. Processivity and drug-dependence of HIV-1 protease: determinants of
viral fitness in variants resistant to protease inhibitors. AIDS. 2003;17:663–671. [PubMed: 12646788]
76. Devereux HL, Emery VC, Johnson MA. et al. Replicative fitness in vivo of HIV-1 variants with multiple drug
resistance-associated mutations. J Med Virol. 2001;65:218–224. [PubMed: 11536226]
77. Markowitz M, Mo H, Kempf DJ. et al. Selection and analysis of human immunodeficiency virus type 1 variants
with increased resistance to ABT-538, a novel protease inhibitor. J Virol. 1995;69:701–706. [PMC free article:
PMC188631] [PubMed: 7815532]
78. Maguire MF, Guinea R, Griffin P. et al. Changes in human immunodeficiency virus type 1 Gag at positions L449
and P453 are linked to I50V protease mutants in vivo and cause reduction of sensitivity to amprenavir and

https://www.ncbi.nlm.nih.gov/books/NBK2244/?report=printable 10/16
2/17/2021 The impact of resistance on viral fitness and its clinical implications - Antiretroviral Resistance in Clinical Practice - NCBI Bookshelf

improved viral fitness in vitro. J Virol. 2002;76:7398–7406. [PMC free article: PMC136352] [PubMed:
12097552]
79. Croteau G, Doyon L, Thibeault D. et al. Impaired fitness of human immunodeficiency virus type 1 variants with
high-level resistance to protease inhibitors. J Virol. 1997;71:1089–1096. [PMC free article: PMC191160]
[PubMed: 8995629]
80. Smidt ML, Potts KE, Tucker SP. et al. A mutation in human immunodeficiency virus type 1 protease at position
88, located outside the active site, confers resistance to the hydroxyethylurea inhibitor SC-55389A. Antimicrob
Agents Chemother. 1997;41:515–522. [PMC free article: PMC163743] [PubMed: 9055985]
81. Maschera B, Darby G, Palu G. et al. Human immunodeficiency virus. Mutations in the viral protease that confer
resistance to saquinavir increase the dissociation rate constant of the protease-saquinavir complex. J Biol Chem.
1996;271:33231–33235. [PubMed: 8969180]
82. Lu J, Sista P, Giguel F. et al. Relative replicative fitness of human immunodeficiency virus type 1 mutants
resistant to enfuvirtide (T-20). J Virol. 2004;78:4628–4637. [PMC free article: PMC387671] [PubMed: 15078945]

https://www.ncbi.nlm.nih.gov/books/NBK2244/?report=printable 11/16
2/17/2021 The impact of resistance on viral fitness and its clinical implications - Antiretroviral Resistance in Clinical Practice - NCBI Bookshelf

Figures

Figure 1
: Distribution of replication capacity of 139 HIV-1 isolates from baseline plasma samples of patients failing highly
active antiretroviral therapy enrolled in the Argenta trial [6,7].

https://www.ncbi.nlm.nih.gov/books/NBK2244/?report=printable 12/16
2/17/2021 The impact of resistance on viral fitness and its clinical implications - Antiretroviral Resistance in Clinical Practice - NCBI Bookshelf

Tables

Table 1 Influence of nucleoside reverse transcriptase inhibitor resistance-associated mutations


(IAS-USA 2005 list) on viral fitness measures

NRTI- Fitness compared


resistance Fitness compared with other Type of evidence and Presence of
mutations with wild-type mutants technique drugs Reference
K70R 97% >> T215Y, M41L, In vitro, growth competition No [48–50]
L210W
K70R – M41L, T215Y> In vivo ZDV [51]
D67N >WT In vitro, growth competition ZDV [52]
K219Q or E >WT In vitro, growth competition ZDV [52]
T215Y 85% >M41L, L210W In vitro, growth competition No [48–50]
M41L 80% >L210W In vitro, growth competition No [48–50]
T215D or S 100% >T215Y T215D In vitro, growth competition No [53,54]
>T215S
T215C or D – >T215Y T215C In vivo No [55]
>T215S
L210W 21% M41L > L210W In vitro, growth competition ZDV [48]
M184I WT>> M184V>> In vitro, growth competition No [56]
M184V WT>>; 57% >> M184I In vitro, growth competition. Ex No [56,57]
vivo, single-replication cycle
assay
M184V 10% >M184I In vivo, population dynamics Yes [22]
L74V 89–94% K70R, M184V>; In vitro, growth competition No [58,59]
K65R>
L74V 100% = M184V In vitro, growth competition ddI [59]
K65R From 55%–57% to >K65R + L74V Ex vivo: single-replication cycle No [57,58,60,
68%–90%; with assay
M184V 30%
Insertions WT>> – In vitro, growth competition Yes, in the [62,63]
codon 69 context of
TAMS
Q151M > WT in the presence – In vitro, growth competition No [12,64,65]
complex of F77L

ddI, didanosine; IAS-USA, International AIDS Society-USA; NRTI, nucleoside reverse transcriptase inhibitor; TAM, thymidine analogue
mutation; WT, wild-type; ZDV, zidovudine; >>, much greater than.

https://www.ncbi.nlm.nih.gov/books/NBK2244/?report=printable 13/16
2/17/2021 The impact of resistance on viral fitness and its clinical implications - Antiretroviral Resistance in Clinical Practice - NCBI Bookshelf

Table 2 Influence of non-nucleoside reverse transcriptase inhibitor resistance-associated


mutations (IAS-USA 2005 list) on viral fitness measures

Fitness
NNRTI- compared
resistance with wild- Fitness compared with Presence
mutations type other mutants Type of evidence and technique of drugs Reference
L100I > WT In vitro, growth competition No [66]
K103N 100% In vitro, growth competition; in vivo, No [16,23,67,
treatment interruption (persistence) 68]
V106A WT> (approx K103N, Y181C, G190A In vitro, growth competition and No [68–71]
15%) and Y188C>; =G190A (in single-replication cycle assay
other example)
Y181C WT = or > > G190A, Y188C; V106A; In vitro, growth competition; in vivo, Both [23,68,
K103N> worse CD4 recovery; persistence after 71,72]
treatment interruption
Y181I = WT In vitro, single-replication cycle assay; No [23,70]
persistence after treatment
interruption
Y188L/H = WT In vitro, single-replication cycle assay No [70]
Y188C WT = or > K103N, Y181C, G190A, In vitro, growth competition and No [68,70]
V106A/Y181C> single replication cycle assay
G190A 83% K103N, Y181C> and In vitro, growth competition; in vivo: No [23,68,71,
>Y188C persistence after treatment 73]
interruption
G190S 21% In vitro, growth competition No [73]
P225H 100% In vitro, single-replication cycle assay No [70]
M230L WT> In vitro, single-replication cycle assay No [70]
P236L 35% K103N> Ex vivo, growth competition; single- No [70,74]
replication cycle assay

IAS-USA, International AIDS Society-USA; NNRTI, non-nucleoside reverse transcriptase inhibitor; WT, wild-type; >>, much greater than.

https://www.ncbi.nlm.nih.gov/books/NBK2244/?report=printable 14/16
2/17/2021 The impact of resistance on viral fitness and its clinical implications - Antiretroviral Resistance in Clinical Practice - NCBI Bookshelf

Table 3 Influence of drug resistance-associated mutations in the protease and gp41 coding regions
(IAS-USA 2005 list) on viral fitness measures

Fitness
compared Fitness compared with other
with wild- mutants: compensatory effect of Presence
type other mutations Type of evidence and technique of drugs Reference
Mutations in protease
I84V WT> or ~90% L10I partially compensates In vitro, growth competition and No [66,75,76]
replication kinetics; in vivo,
treatment interruption
V82A WT>; 79% L10I partially compensates In vitro, processivity; in vivo, No [75,76]
treatment interruption
V82F = WT = L63P, A71V, I84V In vitro, replication kinetics No [77]
V82T WT>> M46I, L63P>> In vitro, growth competition No [17,31]
L90M WT>> L10I and 63P compensatory In vitro and ex vivo, replication No [13,75]
kinetics; ex vivo single-replication
cycle assay
I50V WT>> In vitro, single-replication cycle No [19,78]
assay
N88S WT> L63P and V77I partially In vitro, growth competition No [9]
compensatory
N88D ~90% In vivo, treatment interruption No [76]
D30N 88% or WT>> L90M>> In vivo, treatment interruption; in No [13,76]
vitro replication kinetics
V32I = WT I84V/A further reduced fitness In vitro, growth competition, No [79]
protease catalytic activity
M46I 79% In vivo, treatment interruption No [76]
or L
I54V ~90% In vivo, treatment interruption No [76]
G73S ~90% In vivo, treatment interruption No [76]
L10F WT> In vitro, growth competition or No [19,80]
replication kinetics
G48V WT>> > L90M In vitro, protease activity No [81]
Mutations in the gp41 env
G36D WT > With V38M In vitro, growth competition No [82]
or S
V38M WT > In vitro, growth competition No [82]
N42T WT > In vitro, growth competition No [82]

IAS-USA, International AIDS Society-USA; WT, wild-type; >>, much greater than.

Copyright © 2006, Mediscript.


https://www.ncbi.nlm.nih.gov/books/NBK2244/?report=printable 15/16
2/17/2021 The impact of resistance on viral fitness and its clinical implications - Antiretroviral Resistance in Clinical Practice - NCBI Bookshelf
Bookshelf ID: NBK2244 PMID: 21249769

https://www.ncbi.nlm.nih.gov/books/NBK2244/?report=printable 16/16

You might also like