You are on page 1of 7

Antiretroviral therapy with the integrase inhibitor

raltegravir alters decay kinetics of HIV, significantly


reducing the second phase
John M. Murraya,b, Sean Emerya, Anthony D. Kellehera, Matthew Lawa,
Joshua Chenc, Daria J. Hazudac, Bach-Yen T. Nguyenc,
Hedy Tepplerc and David A. Coopera

Objective: Raltegravir (MK-0518) belongs to the new class of HIV integrase inhibitors.
To date, there have been no reports investigating the potential for differential effects on
Downloaded from http://journals.lww.com/aidsonline by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8K2+Ya6H515kE= on 08/30/2021

viral dynamics with integrase inhibitors relative to current antiretroviral drugs.


Methods: Patients in this phase II study (P004) were antiretroviral treatment naive.
Part 1 of this study compared monotherapy with raltegravir (100 mg, 200 mg, 400 mg, or
600 mg twice daily) with placebo over 10 days. In part 2, patients were enrolled for
48 weeks of combination therapy, with randomization to one of the four dosages of
raltegravir or to efavirenz, in addition to tenofovir and lamivudine. Mathematical
models were used to investigate processes underlying viral dynamics.
Results: From day 15 through to day 57, individuals in the raltegravir arm were
significantly more likely to have HIV RNA < 50 copies/ml (P  0.047). Plasma viral
loads were 70% lower at initiation of second-phase decay for individuals taking
raltegravir than for those taking efavirenz (P < 0.0001). This challenges the current
hypothesis that second-phase virus originates from infected long-lived cells, as an
integrase inhibitor should not impact on viral production from this cell population.
Mathematical modeling supported two hypotheses as consistent with these obser-
vations: (i) that second-phase virus arises from cells newly infected by long-lived
infected cells and (2) that it arises from activation of latently infected cells with full-
length unintegrated HIV DNA.
Conclusions: These observations challenge the current understanding of HIV-1 turn-
over and compartmentalization. They also indicate the promise of this new integrase
inhibitor raltegravir. ß 2007 Wolters Kluwer Health | Lippincott Williams & Wilkins

AIDS 2007, 21:2315–2321

Keywords: antiviral therapy, HIV-1, integrase inhibitors, mathematical models,


raltegravir, viral load

Introduction requires integration of HIV DNA, an integrase inhibitor


prevents an infectious virion that has progressed through
HIV integrase inhibitors are a new class of antiretroviral reverse transcription from successfully infecting the cell.
drugs that target a crucial process in the life cycle of HIV This class of drugs has a mode of action that is independent
[1–3]. They act by blocking incorporation of the proviral of reverse transcriptase and protease inhibitors and has the
HIV DNA into the host cell DNA. As productive infection capacity to assist in control of HIV replication.

From the aNational Centre in HIV Epidemiology and Clinical Research, the bSchool of Mathematics and Statistics, University of
New South Wales, Sydney, Australia, and the cMerck Research Laboratories, North Wales, Pennsylvania, USA.
Correspondence to Dr J. Murray, School of Mathematics and Statistics, University of New South Wales, Sydney, NSW 2052,
Australia.
E-mail: J.Murray@unsw.edu.au
Received: 14 June 2007; revised: 10 August 2007; accepted: 13 August 2007.

ISSN 0269-9370 Q 2007 Wolters Kluwer Health | Lippincott Williams & Wilkins 2315
Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
2316 AIDS 2007, Vol 21 No 17

Ten-day monotherapy trials investigating dosing levels HIV RNA of  5000 copies/ml and a CD4 cell count
and tolerability of integrase inhibitors have been reported of  100 cells/ml. HIV RNA was assessed by the
[1,3], and preliminary results from phase II studies suggest AMPLICOR HIV-1 Monitor Standard Assay version
that this new class of drugs can be effective in controlling 1.5 (limit of detection, 400 copies/ml; Roche Molecular
HIV replication even in individuals infected with Systems, Branchburg, New Jersey, USA); those samples
multiclass drug-resistant virus [4]. To date, however, below the limit of detection were reassessed by the
there have been no reports investigating the potential for AMPLICOR HIV-1 Monitor UltraSensitive Assay version
differential effects on viral dynamics with integrase 1.5 (limit of detection, 50 copies/ml).
inhibitors relative to current antiretroviral drugs.
The mathematical models are described in the supple-
Classically, the reduction in HIV replication under mentary text. Statistical comparisons for continuously
antiretroviral therapy (ART) is characterized by a biphasic distributed variables between groups were performed with
decay pattern of HIV RNA in blood. The first phase the Wilcoxon rank sum test, while comparisons for
exhibits a mean half-life of between 0.9 and 1.6 days and proportions of patients above or below detection limits
occurs over the first 7 to 10 days [5–7]. The second phase were performed using Fisher’s exact test. Nominal P values
has a mean half-life of 14 days and successful therapy < 0.05 were considered significant and all tests were two
reduces plasma HIV RNA below standard limits of sided. In this exploratory analysis, no adjustments were
detection [6]. Analysis of HIV RNA < 50 copies/ml made to P values for multiple testing. Mathematical
suggests that HIV RNA stabilizes at low but detectable modeling and statistical analyses were conducted within
levels in plasma, with 50% of patients who have been Matlab, version 7, The MathWorks.
taking ART for at least 6 months having HIV RNA
> 2.5 copies/ml [8].

It is generally believed that virus observed during the Results


first-phase decay arises from productively infected CD4
T cells, and that this phase of viral decay tracks their death Patients in this study were naive for ART, had baseline
[9,10]. The origins of HIV RNA during the second phase plasma HIV RNA of  5000 copies/ml and CD4 T cell
are not clearly defined. It has been hypothesized the virus counts  100 cells/ml. This phase II dose ranging study of
may arise from long-lived infected cells [6], from raltegravir was conducted in two parts. Part 1 compared
dissociation of virus from follicular dendritic cells [11] monotherapy with raltegravir against placebo over 10 days
or from productively infected CD4 T cells that are not [3]. Patients entering part 1 were randomized to one of four
subject to clearance by the host immune responses given different dosages of raltegravir (100, 200, 400 and 600 mg),
the waning levels of HIV antigen [12]. each taken twice daily, or matched placebo. In Part 1 there
were approximately eight patients in each of the
The addition of an integrase inhibitor such as raltegravir five groups. At the end of Part 1 therapy was stopped
(formerly known as MK-0518) to antiretroviral drugs before commencement of Part 2. In Part 2, an additional
from other classes may be expected to provide at least an 150 patients were enrolled for 48 weeks of combination
additive effect on inhibition of HIV replication. therapy, with randomization to one of the four dosages of
However, it would not be expected to interfere with raltegravir or to efavirenz (600 mg every night), in addition
production of progeny virus from the above sources since to tenofovir (300 mg once a day) and lamivudine (300 mg
the cells that are hypothesized to be responsible for the once a day) for all individuals.
observed second-phase virus are already infected or, in
the case of dissociation from dendritic cells, these virions First-phase decay with raltegravir monotherapy
have already been produced. Monotherapy with raltegravir over 10 days produced
extensive monophasic decay for all dosage groups, with a
To investigate these matters, viral dynamics were analyzed median 2.2 log10 copies/ml decrease. Linear regression
from treatment-naive patients enrolled in a phase II dose- analysis determined no significant differences in the first-
ranging study of raltegravir over 10 days of monotherapy phase half-lives between the dosage groups (P  0.18).
and then in comparison over 48 weeks with efavirenz, Mean half-lives were 1.3, 1.1, 1.3, and 1.2 days for the 100,
each in combination with two other antiretroviral drugs. 200, 400 and 600 mg raltegravir groups, respectively,
with an overall mean of 1.2 days (SD, 0.26). The first-phase
half-lives were consistent with the ranges previously
observed [5–7].
Methods
Combination therapy with raltegravir
Patients were naive to ARTor had received less than 7 days Under combination therapy, each of the raltegravir
total ART. Patients were at least 18 years of age with a treatment groups achieved undetectable HIV RNA
positive HIV enzyme-linked immunosorbent test, had (<50 copies/ml), faster than the efavirenz group. From

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Raltegravir reduces second-phase virus Murray et al. 2317

Table 1. Numbers of individuals in each treatment group with HIV RNA/ml < 50 copies/ml.

Raltegravir (mg) twice daily


a
Efavirenz (600 mg every night) b
Time (days) 100 (n ¼ 39) 200 (n ¼ 40) 400 (n ¼ 41) 600 (n ¼ 40) (n ¼ 38) P value

1 0 0 0 0 0 1
15 12 12 19 12 4  0.047
29 25 22 31 25 8  0.003
57 33 34 37 28 14  0.006
85 35 34 36 35 26 0.03  P 0.10
112 29 31 35 36 27 0.02  P 1.0
168 31 34 36 38 33 0.30  P 1.0
223 28 31 34 33 31 0.40  P 1.0
280 29 29 34 34 32 0.30  P 1.0
335 24 27 31 29 29 0.20  P 1.0
a
Mean time over all patients.
b
Probability that the efavirenz treatment group was significantly different to each of the raltegravir treatment groups.

the second (day 15 of therapy, on average) to the fourth second-phase half-life for raltegravir individuals was
time point (day 57), patients in the raltegravir groups 15.5 days [interquartile range (IQR), 14], whereas it was
were more likely to be below the level of HIV detection 18.3 days (IQR, 10) for those on efavirenz, but these half-
(Table 1). Since first-phase monotherapy decay rates were lives were not significantly different (P ¼ 0.2). The slightly
indistinguishable for each of the dosage groups, and all numerically faster second-phase dynamics for the ralte-
raltegravir dosages achieved undetectable RNA at similar gravir group may be because some of these individuals had
rates with combination therapy, subsequent analysis was a first-phase decline that was prolonged beyond day 15.
performed on the combined raltegravir patients. Plasma This appears likely given the extended first-phase decline
HIV RNA was significantly lower for the combined exhibited under monotherapy. Including only those
raltegravir group until day 168 (Fig. 1). From day 1 to day individuals on raltegravir who had detectable HIV
15, median HIV RNA fell from 58 350 to 98 copies/ml RNA at days 15 and 29 (n ¼ 42), gave a median
for the combined raltegravir patients, while those in the second-phase half-life of 19.6 days, which also was not
efavirenz group decreased from 70 200 to 537 copies/ml. significantly different to the efavirenz group (P ¼ 0.6).

Second-phase half-lives were calculated using linear Although the rate of decline during the second phase did
regression on HIV RNA log10 copies/ml from the second not differ between the raltegravir and efavirenz regimens,
time point (day 15 on average), until the first point where the point at which this decline initiated did differ.
HIV RNA fell below the 50 copies/ml limit. The median The significantly lower viral levels in plasma from day 15

(a) (c)
HIV RNA (log10 copies/ml)

6 6
HIV RNA (log10 copies/ml)

5 5

4 4

3 3

2 2

1 1
0 50 100 150 200 250 300 350 0 50 100
Days
(b)
HIV RNA (log10 copies/ml)

1
0 50 100 150 200 250 300 350
Days

Fig. 1. Combination therapy starting at day 1 for patients taking all dosages of (a) raltegravir (160 patients at day 1 to 121 at final
time) or (b) efavirenz (36 patients at day 1 to 30 at final time). At each time point, the central horizontal line depicts the median,
with the box extending over the interquartile range. Outliers, more than 1.5 times the interquartile range away from the box
extremities, are depicted as ‘þ’. (c) Median HIV RNA for raltegravir (squares) and efavirenz (diamonds) arms.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
2318 AIDS 2007, Vol 21 No 17

onwards for the raltegravir group meant that the first Sanctuary
phase of viral decay was more extensive, thereby reducing site
the viral level at which the second phase commenced
(Fig. 1). Estimation of the size of the viral compartment IS
contributing to second-phase viral decay was calculated
by extrapolating the regression line of this second phase
back to baseline. The ratio of this baseline second-phase M

Ra
lte
value to baseline HIV RNA (‘M ratio’), has been listed for

gr
3
six individuals in Perelson et al. [6] (Table 1, pM0/cV0).

av
ir
In that study combination therapy with nelfinavir,
zidovudine and lamividine resulted in M ratios with a D
1
mean of 0.04 and median of 0.03. The lower the M ratio Ralteg
ravir
V
the more extensive was first phase decline. In the present
study, M ratios for the efavirenz group were lower but not
statistically different to values reported in the literature,

ir
with the higher literature values perhaps arising from a

av
2 I

gr
slightly different method where a viral load curve was

lte
Ra
fitted to the data in that study [6] (P ¼ 0.08). The M ratio
was 70% lower for individuals taking raltegravir than
efavirenz (median 0.0042 versus 0.014; P < 0.0001),
implying that raltegravir reduces viral production from Fig. 2. A cartoon representation of the effects of raltegravir
the second-phase source by this amount over and above in the three models shown as numbered circles. Long-lived
standard regimens. infected cells are denoted with M, latently infected cells with
unintegrated virus as D, productively infected cells as I, and
The decrease in the second-phase viral production with the productively infected cells in a sanctuary site as IS, with virus
addition of raltegravir necessitates rethinking current as V. HIV DNA is denoted by a solid red bar in each cell.
theories of the source of virus responsible for the second
phase. Current hypotheses are that second-phase viral load infected cells (M). These latter cells decay at the slow rate
arises through production by long-lived infected cells [6], observed in this phase, thereby providing this same
release from follicular dendritic cells [11] or decreased dynamic to newly infected cells and second-phase virus
cytotoxic lymphocyte response [12]. None of these (Fig. 3a,b). The integrase inhibitor provides an additional
theories can explain the effects produced by raltegravir, effect to reverse transcriptase and protease inhibitors in
if it is acting as a pure integrase inhibitor, since they suggest their inhibition of new productive infection from long-
that second-phase viral load originates from sources that an lived infected cells, decreasing second-phase viral levels
integrase inhibitor would not affect: previously infected more (solid lines Fig. 3a,b) than with efavirenz (dashed
cells or virus already produced but bound to dendritic cells. lines).

Hypotheses for second-phase virus The results achieved with the integrase inhibitor in model
If an integrase inhibitor is to influence viral levels in the 1 could theoretically be achieved through more potent
second phase, the virus at that stage must arise from (i) cells reverse transcriptase or protease inhibitors. By comparison,
that are either newly infected and where the integrase model 2 simulates a process of infection that is not
inhibitor provides an additional benefit above that of susceptible to traditional drugs, where virus from the
reverse transcriptase and protease inhibitors, (ii) cells that second phase arises from latently infected cells that contain
have been previously infected but where the proviral DNA proviral DNA in a full-length unintegrated form, which
has yet to be integrated into the host cell genome, or (iii) a has been estimated to form the vast majority of latent
sanctuary site where the integrase inhibitor has improved infection [13]. In this model, second-phase virus originates
penetration. To investigate the viability of these hypotheses from latently infected cells with unintegrated viral DNA
as explanations of the reduced second phase under an (D) that are activated and converted to productively
integrase inhibitor, three mathematical models were infected cells (I). Increased potency of traditional drugs will
developed and applied to the median data derived from reduce the replenishment of the latent pool D but will not
the monotherapy and combination trials with raltegravir stop its conversion to productive infection. Model 2 also
and efavirenz. Model 1 simulated the first hypothesis provides a consistent explanation for the effect achieved by
above, model 2 simulated the second, and model 3 the integrase inhibitor (Fig. 3c,d).
simulated the third (supplementary text). A cartoon of the
effects of raltegravir in these models is displayed in Fig. 2. The third model, where the differential effects of the
integrase inhibitor are achieved through better penetration
In model 1, new productive infection (I), which generates of a sanctuary site, was less successful in duplicating the data
virus (V) in the second phase, originates from long-lived (Fig. 3e,f). Although it achieved lower second-phase viral

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Raltegravir reduces second-phase virus Murray et al. 2319

(a) (c) (e)


5 5 5
10 10 10

HIV RNA (copies/ml)

0 0 0
10 10 10
0 20 40 60 0 20 40 60 0 20 40 60
Days Days Days
(b) (d) (f)
0 0 0
10 10 10
Cell count (cells/µl)

0 20 40 60 0 20 40 60 0 20 40 60
Days Days Days

Fig. 3. Simulations for model 1 (a,b), model 2 (c,d) and model 3 (e,f). (a,c,e) Simulations versus median HIV RNA data from part 1
and part 2 with raltegravir (squares) and part 2 data for the efavirenz arm (diamonds). (b,d,f) Simulations of underlying cell types.
Simulations with an integrase inhibitor are depicted as solid lines and those without an integrase inhibitor are depicted as dashed
lines. Some cell population simulations show no difference whether an integrase inhibitor is included or not, and for these cases
the solid line overlays the dashed line. (b) Model 1 simulations for long-lived infected cells M (green lines) and productively
infected cells I (blue lines). (d) Model 2 simulations for long-lived infected cells M (green lines), productively infected cells I
(blue lines), and latently infected cells D (red lines). (f) Model 3 simulations for productively infected cells in a sanctuary site
IS (green lines) and productively infected cells I in a site susceptible to conventional antiretroviral therapy (blue lines).

load, the slope of this second phase was significantly faster raltegravir arm with those on the efavirenz arm,
for the integrase inhibitor (solid line Fig. 3e) than for the raltegravir achieved a 70% reduction of second-phase
efavirenz arm (dashed line Fig. 3e). This was at odds with viral levels.
the results described above, where there was no significant
difference. The second-phase virus in this model traffics The effect of raltegravir on the second phase of viral decay
from the sanctuary site, where it is produced by was unexpected given current hypotheses for the processes
productively infected cells. In order to achieve this underlying this phase [6,11,12] and the mechanism by
differential effect for the integrase inhibitor, the efavirenz which the drug is thought to work. We proposed three
arm must have extremely low efficacy, 6%, in order for the hypotheses to explain the observed effects and compared
additional efficacy provided by the integrase inhibitor to simulations of mathematical models for these hypotheses to
significantly perturb the decay rate of productively infected median data for ART with or without an integrase
cells in the sanctuary site (green solid line versus green inhibitor. Models 1 and 2 successfully reproduced the
dashed line Fig. 3f). clinical data (Fig. 3).

The effect of adding an integrase inhibitor to model 1 is to


provide an additional effect to that of the reverse
Discussion transcriptase inhibitors (and any protease inhibitors that
act in the model through decreasing infectivity), thereby
Raltegravir significantly extended the first phase of viral decreasing new infection from virus associated with long-
decay and reduced the plasma HIV RNA level at which lived infected cells. Hence under these assumptions, the
the second phase commenced (Fig. 1). It enabled integrase inhibitor does not target new infection mech-
individuals to suppress HIV RNA to below detection anisms but reduces the success of transmission from long-
limits significantly faster than current regimens (Table 1). lived infected cells to productively infected cells. Model 2,
The 50 copies/ml assay limit did not allow investigation of where the second phase is assumed to arise from activation
any effects this may have on subsequent viral levels. By of latently infected cells that have HIV DNA in a
comparing M ratios for individuals enrolled on the predominantly unintegrated form, provides a mechanism

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
2320 AIDS 2007, Vol 21 No 17

that is completely resistant to any additional benefit from culture conditions, as infection of resting cells can fail
improvements in reverse transcriptase inhibition. For this during reverse transcription [19,20] or through the
second model, the integrase inhibitor provides a protective lability of the full-length unintegrated HIV DNA
mechanism that cannot be replaced by reverse transcriptase transcript [20], or it can lead to stable infection that is
or protease inhibitors, no matter their potency, since for transcriptionally inactive without cellular activation [21].
reverse transcriptase and protease inhibitors new virions The in-vivo situation under ART is equally unclear. Total
will be produced, although these will be mostly HIV DNA exceeds integrated HIV DNA in resting CD4
noninfectious in the presence of protease inhibitors [14]. T cells by 100-fold, yet does not seem to contribute
Under each of these models, there may be an increase in significantly to replication competent virus [13]. The
aborted HIV DNA integration and a resulting rise in presence of unintegrated HIV DNA under ART,
extrachromosomal viral DNA in circular forms containing however, indicates either that there is substantial ongoing
one or two long terminal repeats (2LTR circles), as infection, if this molecule is labile, or that it is long lived.
observed in vitro [2]. In those experiments, cell viability was If it is the latter, then integrase inhibitors can provide
not affected, and clinical trials with raltegravir have not an effective block in new productive infection upon
shown toxicities higher than with current antiretroviral activation of this latent component, as hypothesized in
drugs [3,4]. Therefore, any resulting increases in model 2. Regardless, integrated HIV DNA must undergo
unintegrated HIV DNA would not be expected to be some replenishment since it decays slowly under ART but
detrimental to these cells. Although not conclusive, the rebounds within weeks upon cessation of therapy [13]. It
simulations of model 3 suggest that second-phase virus does is difficult to reconcile these very different dynamics if
not originate from a sanctuary site where raltegravir is more there is no compensatory loss in this compartment, and
successful in penetrating than standard antiretroviral drugs. hence continual replacement. Detailed analysis of
integrated and unintegrated HIV DNA in resting CD4
Availability of protease inhibitors in the 1990s and their T cells, in the presence and absence of an integrase
combination with reverse transcriptase inhibitors evoked inhibitor, would provide valuable information on this
hopes that this new potent ART would at last allow matter.
clearance of HIV infection. Estimates were made on the
length of treatment needed before this could be achieved
[6]. Although plasma levels of HIV RNA fall below
detection limits within several months of ART, it was
soon realized that latent infection provided a lower bound Acknowledgements
on the rate at which HIV infection could be removed
from the body. This component is established early in We thank the patients and investigators who participated
infection [15] and consists of both unintegrated and in this study.
integrated HIV DNA even under ART [13]. The high
levels under ART of unintegrated HIV DNA in resting Sponsorship: The National Centre in HIV Epidemiology
and Clinical Research is funded by the Australian
CD4 T cells [13], and proviral sequence evolution in Commonwealth Department of Health and Ageing and
individuals with undetectable HIV RNA [16], suggest is affiliated with the Faculty of Medicine at the
that there is persistent viral replication even in the University of New South Wales. AK is supported by a
presence of seemingly successful ART. Regardless of Practitioner Fellowship from the NHMRC.
whether ongoing infection or the slow turnover of resting
CD4 T cells is primarily responsible, the latent pool Note: The P004 study is registered at ClinicalTrials.gov
decreases slowly under standard ART, with a half-life of under identifier NCT00100048.
anywhere between 6 months [16,17] and 44 months [18].
This leads to estimates of between 8 years [16,18] and Note: Supplementary material can be accessed online.
more than 60 years for clearance of infection. The rate of
decrease is dependent on residual viral replication [17], References
and hence drugs like raltegravir that increase the
1. DeJesus E, Berger D, Markowitz M, Cohen C, Hawkins T, Ruane
effectiveness of ART, especially for viral components P, et al. Antiviral activity, pharmacokinetics, and dose response
such as unintegrated HIV DNA that are immune to of the HIV-1 integrase inhibitor GS-9137 (JTK-303) in
current drugs, may improve this rate of decay. treatment-naive and treatment-experienced patients. J Acquir
Immune Defic Syndr 2006; 43:1–5.
2. Hazuda DJ, Felock P, Witmer M, Wolfe A, Stillmock K, Grobler
There is considerable uncertainty as to the prevalence of JA, et al. Inhibitors of strand transfer that prevent integration
unintegrated HIV DNA in resting CD4 T cells during and inhibit HIV-1 replication in cells. Science 2000; 287:646–
650.
ART, and the impact this component makes to renewing 3. Markowitz M, Morales-Ramirez JO, Nguyen BY, Kovacs CM,
the integrated pool. In-vitro experiments have established Steigbigel RT, Cooper DA, et al. Antiretroviral Activity,
that HIV can enter resting CD4 T cells with high pharmacokinetics, and tolerability of MK-0518, a novel inhi-
bitor of HIV-1 integrase, dosed as monotherapy for 10 days in
efficiency, but that productive infection does not ensue. treatment-naive HIV-1-infected individuals. J Acquir Immune
The cause of this failure seems dependent on the cell Defic Syndr 2006; 43:509–515.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Raltegravir reduces second-phase virus Murray et al. 2321

4. Grinsztejn B, Nguyen BY, Katlama C, Gatell JM, Lazzarin A, 13. Chun TW, Stuyver L, Mizell SB, Ehler LA, Mican JA, Baseler M,
Vittecoq D, et al. Safety and efficacy of the HIV-1 integrase et al. Presence of an inducible HIV-1 latent reservoir during
inhibitor raltegravir (MK-0518) in treatment-experienced highly active antiretroviral therapy. Proc Natl Acad Sci USA
patients with multidrug-resistant virus: a phase II randomised 1997; 94:13193–13197.
controlled trial. Lancet 2007; 369:1261–1269. 14. Ashorn P, McQuade TJ, Thaisrivongs S, Tomasselli AG, Tarpley
5. Plipat N, Ruan PK, Fenton T, Yogev R. Rapid Human immu- WG, Moss B. An inhibitor of the protease blocks maturation of
nodeficiency virus decay in highly active antiretroviral therapy human and simian immunodeficiency viruses and spread of
(HAART)-experienced children after starting mega-HAART. infection. Proc Natl Acad Sci USA 1990; 87:7472–7476.
J Virol 2004; 78:11272–11275. 15. Chun TW, Engel D, Berrey MM, Shea T, Corey L, Fauci AS. Early
6. Perelson AS, Essunger P, Cao Y, Vesanen M, Hurley A, Saksela establishment of a pool of latently infected, resting CD4(R)
K, et al. Decay characteristics of HIV-1-infected compartments T cells during primary HIV-1 infection. Proc Natl Acad Sci USA
during combination therapy. Nature 1997; 387:188–191. 1998; 95:8869–8873.
7. Kuritzkes DR, Ribaudo HJ, Squires KE, Koletar SL, Santana J, 16. Zhang L, Ramratnam B, Tenner-Racz K, He Y, Vesanen M,
Riddler SA, et al. Plasma HIV-1 RNA dynamics in antiretroviral- Lewin S, et al. Quantifying residual HIV-1 replication in
naive subjects receiving either triple-nucleoside or efavirenz- patients receiving combination antiretroviral therapy. N Engl
containing regimens: ACTG A5166s. J Infect Dis 2007; J Med 1999; 340:1605–1613.
195:1169–1176. 17. Ramratnam B, Mittler JE, Zhang L, Boden D, Hurley A, Fang F,
8. Palmisano L, Giuliano M, Nicastri E, Pirillo MF, Andreotti M,
et al. The decay of the latent reservoir of replication-competent
Galluzzo CM, et al. Residual viraemia in subjects with chronic HIV-1 is inversely correlated with the extent of residual viral
HIV infection and viral load < 50 copies/ml: the impact of replication during prolonged antiretroviral therapy. Nat Med
highly active antiretroviral therapy. AIDS 2005; 19:1843– 2000; 6:82–85.
1847.
9. Ho DD, Neumann AU, Perelson AS, Chen W, Leonard JM, 18. Finzi D, Blankson J, Siliciano JD, Margolick JB, Chadwick K,
Markowitz M. Rapid turnover of plasma virions and CD4 Pierson T, et al. Latent infection of CD4R T cells provides a
lymphocytes in HIV-1 infection. Nature 1995; 373:123–126. mechanism for lifelong persistence of HIV-1, even in patients
10. Wei X, Ghosh SK, Taylor ME, Johnson VA, Emini EA, Deutsch P, on effective combination therapy. Nat Med 1999; 5:512–517.
et al. Viral dynamics in human immunodeficiency virus type 1 19. Zack JA. The role of the cell cycle in HIV-1 infection. Adv Exp
infection. Nature 1995; 373:117–122. Med Biol 1995; 374:27–31.
11. Hlavacek WS, Stilianakis NI, Notermans DW, Danner SA, 20. Zhou Y, Zhang H, Siliciano JD, Siliciano RF. Kinetics of human
Perelson AS. Influence of follicular dendritic cells on decay immunodeficiency virus type 1 decay following entry into
of HIV during antiretroviral therapy. Proc Natl Acad Sci USA resting CD4R T cells. J Virol 2005; 79:2199–2210.
2000; 97:10966–10971. 21. Spina CA, Guatelli JC, Richman DD. Establishment of a stable,
12. Arnaout RA, Nowak MA, Wodarz D. HIV-1 dynamics revisited: inducible form of human immunodeficiency virus type 1 DNA
biphasic decay by cytotoxic T lymphocyte killing? Proc R Soc in quiescent CD4 lymphocytes in vitro. J Virol 1995; 69:2977–
Lond, Ser B Biol Sci 2000; 267:1347–1354. 2988.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

You might also like