• Nenhum resultado encontrado

Antiretroviral drug therapy alters the profile of human immunodeficiency virus type 1-specific T-cell responses and shifts the immunodominant cytotoxic T-lymphocyte response from gag to pol

N/A
N/A
Protected

Academic year: 2017

Share "Antiretroviral drug therapy alters the profile of human immunodeficiency virus type 1-specific T-cell responses and shifts the immunodominant cytotoxic T-lymphocyte response from gag to pol"

Copied!
6
0
0

Texto

(1)

0022-538X/07/$08.00⫹0 doi:10.1128/JVI.00779-07

Copyright © 2007, American Society for Microbiology. All Rights Reserved.

Antiretroviral Drug Therapy Alters the Profile of Human Immunodeficiency

Virus Type 1-Specific T-Cell Responses and Shifts the

Immunodominant Cytotoxic T-Lymphocyte Response

from Gag to Pol

A. C. Karlsson,

1,2

* J. M. Chapman,

3

B. D. Heiken,

1

R. Hoh,

4

E. G. Kallas,

5

J. N. Martin,

4

F. M. Hecht,

3

S. G. Deeks,

3

† and D. F. Nixon

3

Gladstone Institute of Virology and Immunology, University of California, San Francisco, California 941411; The Swedish Institute for

Infectious Disease Control and Karolinska Institutet, 171 82 Solna, Sweden2; Division of Experimental Medicine, Department of

Medicine, University of California, San Francisco,3and Positive Health Program, San Francisco General Hospital,4

San Francisco, California 94110; and Federal University of Sa˜o Paulo, Sao Paulo, Brazil5

Received 11 April 2007/Accepted 20 July 2007

Antiretroviral drug therapy and cytotoxic T lymphocytes (CTL) both exert selective pressures on human immunodeficiency virus type 1, which influence viral evolution. Compared to chronically infected, antiretro-viral-untreated patients, most chronically infected, treated patients with detectable viremia lack a cellular immune response against the Gag 77–85(SL9) epitope but show a new immunodominant response against an epitope in protease PR 76–84. Hence, mutations induced by antiretroviral therapy likely alter the profile of epitopes presented to T cells and thus the direction of the response. The consequences of dual pressures from treatment and CTL need to be considered in monitoring of drug therapy.

The course of human immunodeficiency virus type 1 (HIV-1) infection is characterized by the development of an intricate CD8⫹ cytotoxic T-lymphocyte (CTL) response

di-rected against an evolving viral genome (24). CTL pressure can lead to early escape from a dominant epitope, as shown in the simian immunodeficiency virus model, where pressure on Tat leads to rapid escape and a change in dominance to recogni-tion of the Gag CM9 epitope (3). A different pattern of chang-ing epitopic usage over time is illustrated by responses to the immunodominant HLA-A2-restricted Gag 77–85 (77SLYNTV

ATL85[SL9]) epitope in p17 Gag. SL9 is recognized by the

majority of chronically infected individuals but not by most acutely infected individuals (4, 10, 16, 18, 21, 24, 37). This temporal relationship for CTL recognition of virus proteins/ epitopes is also supported by observations that the HIV-1-Nef protein is recognized before other proteins (2, 5, 9, 28) and, in mouse models, by constantly evolving CTL immunodominance (7, 42–44).

In addition to CTL, therapy with antiretroviral drugs also exerts significant pressures on the viral genome. This treat-ment-associated pressure is largely direct and can lead to the rapid accumulation of drug-resistance associated mutations. Among treated patients with drug-resistant HIV, the steady-state viral load is often lower than the pretreated levels. This partial viral suppression is due in part to residual activity of antiretroviral drugs and reduced replicative capacity of the

drug-resistant variants (6, 13). We and others, however, have argued that drug treatment may exert pressure indirectly via the immune system (1, 12, 23, 36, 38). This interaction can occur via a number of non-mutually exclusive mechanisms, including treatment-mediated selection of new mutations that act as novel epitopes (23, 31–33, 39–41) and/or treatment-mediated decreases in replicative capacity (“fitness”), which can lead to a reduction in the ability of HIV-1 to destroy antigen-specific T cells (6, 11, 13, 20).

Although several studies suggest that treatment can modify viral evolution via its effect on T-cell immunity, no study has comprehensively studied the impact of treatment on the hier-archy of immunodominant responses. To address this issue, we conducted a detailed study of the interactions between viral sequence changes and the specificity of the cellular immune responses. We focused our analysis on well-described restricted epitopes, including SL9, and studied HLA-A2-positive treated and untreated subjects during primary and chronic HIV-1 infection.

A total of 38 HLA-A2-positive subjects were identified from the UCSF cohorts of primary infection (OPTIONS) (19, 24) and from the SCOPE cohort of chronically infected subjects (14) and were divided into three groups. The three groups were designated as follows. Group 1 contained subjects with acute infection and who were antiretroviral untreated (n⫽8; median viral load of 4.4 logs, median CD4 cell count of 541 cells/mm3

and median estimated duration of infection 8 weeks), group 2 contained subjects with chronic infection and who were antiretroviral untreated (n⫽10; median viral load of 4.4 logs, median CD4 cell count of 322 cells/mm3

), and group 3 contained subjects with chronic infection and who were pro-tease inhibitor treated with detectable viremia (n⫽20; median viral load of 3.9 logs and median CD4 cell count of 287 cells/ mm3

). The protease inhibitor-treated subjects all had devel-* Corresponding author. Mailing address: Karolinska Institutet and

The Swedish Institute for Infectious Disease Control (SMI), Depart-ment of Virology, Immunology, and Vaccinology, 171 82 Solna, Swe-den. Phone: 46 8 457 26 19. Fax: 46 8 33 72 72. E-mail: annika.karlsson @smi.ki.se.

† S.G.D. and D.F.N. made equal contributions to this study.

Published ahead of print on 1 August 2007.

(2)

oped a number of resistance-associated mutations against both reverse transcriptase and protease inhibitors (23). Viral se-quence data were generated using population-based sequenc-ing from extracted viral RNA in plasma [Dynabeads Oligo(dT)25; Invitrogen Corporation, Dynal Biotech, Oslo,

Norway] and viral DNA from peripheral blood mononuclear cells (QIAamp DNA blood kit; QIAGEN, Valencia, CA), as indicated in Table 1. Sequences obtained from the same indi-vidual originating from viral DNA and RNA always aligned together and showed high sequence identity with each other. When available, sequences obtained from viral RNA in plasma were used in the sequence analysis. The study was approved by the UCSF Institutional Review Board, and all subjects pro-vided written informed consent.

We first analyzed CTL responses targeting SL9 in a group of 20 chronically HIV-1-infected, antiretroviral-treated subjects with detectable viremia. Surprisingly, we found that these individuals generally lacked a response against this epitope. Using the intracellular cytokine flow cytometry (ICS) assay (23, 24), only 3 of 20 patients had a detectable CTL response measured by the production of interferon gamma (IFN-␥) against the SL9 epitope. Prior studies have repeatedly shown responses to the SL9 epitope in the majority of chronically infected, antiretroviral-untreated subjects (4, 18, 21, 22, 37), although these responses were not always im-munodominant (8).

To evaluate if the lack of T-cell responses to SL9 were influenced by treatment exposure and sequence changes, we performed additional studies assessing Gag-, Env-, Nef-, and Pol-specific responses from 10 antiretroviral-treated and 10 antiretroviral-untreated subjects with chronic infection (Table 1). Using the sequence information obtained by

population-based sequences of the Gag p17, protease, reverse transcrip-tase (partial), and Nef regions, we manufactured peptides cor-responding to a panel of 20 epitopes and measured HIV-1-specific IFN-␥and tumor necrosis factor alpha (TNF-␣) CTL responses using the ICS assay corresponding to autologous and consensus B sequences (Fig. 1) (24). As has been observed by others (4, 18, 21, 22, 37), the majority of chronically infected, TABLE 1. Patient characteristics and treatment history of chronically untreated and treatment-experienced HIV-1-infected subjects

Patient (copies/ml)Viral load (cells/mmCD4 count3) Treatmenta

Origin of viral sequences

Gag Pol Nef

Chronically infected, untreated

1028 8,626 411 Last meds in 1992; no PIs RNA/DNA RNA RNA

1029 2,062 373 Never RNA RNA RNA

1030 201,304 258 Never RNA/DNA RNA RNA

1034 34,004 365 Never RNA RNA RNA

1038 27,846 178 Never RNA/DNA RNA RNA

1051 65,276 238 Last meds in 1998; no PIs RNA RNA RNA

1057 8,638 216 last meds in 1999; no PIs RNA RNA RNA

1058 63,932 585 Never RNA/DNA RNA RNA

1074 9,946 688 Never RNA RNA RNA

1079 25,655 438 Never RNA RNA RNA

Chronically infected, treated (viremic)

3002 6,655 195 16 yr; PIs for 5 yr RNA RNA RNA

3007 8,712 288 14.5 yr; PIs for 5.5 yr RNA/DNA RNA RNA

3011 7,585 125 6.75 yr; PIs for 4 yr RNA/DNA RNA RNA/DNA

3040 21,487 237 10 yr; PIs for 10 yr RNA/DNA RNA DNA

3042 278 160 3.75 yr; PIs for 3 yr RNA/DNA RNA RNA/DNA

3057 20,760 514 10 yr; PIs for 6 yr RNA/DNA RNA RNA/DNA

3109 13,731 296 13 yr; PIs for 6 yr DNA RNA/DNA RNA/DNA

3151 28,610 184 4.5 yr; PIs for 2 yr RNA/DNA RNA/DNA RNA/DNA

3153 5,630 454b 13 yr; PIs for 4 yr RNA/DNA RNA/DNA RNA/DNA

3156 175,000 891 1.5 yr; PIs for 1 yr RNA/DNA RNA/DNA RNA/DNA

ameds, medications; PIs, protease inhibitors. bEstimated.

FIG. 1. Antiviral treatment alters the distribution of epitope rec-ognition. Most chronically infected, antiretroviral-treated patients have gained a response targeting an epitope, which is under strong protease inhibitor-mediated drug pressure (PR 77–85). In contrast to the untreated chronically infected subjects, the treated individuals generally lack responses against the SL9 epitope. A sample was con-sidered positive when the responses were at least two times the exper-imental background and above 0.05% IFN-␥- and TNF-␣-positive CD8⫹

CD4⫺

CD3⫹

(3)

untreated subjects had measurable CTL responses targeting the autologous variant of the SL9 epitope (seven of nine sub-jects tested) (Table 2). This is in contrast to the chronically infected, treated individuals, of whom only 2 of 6 had a mea-surable CTL response against the autologous SL9 epitope (P⫽

0.14; Fisher’s exact test for chronically infected, treated versus untreated) and 2 of 10 had a response against the wild-type SL9 epitope (P ⫽ 0.02). Although the treated individuals lacked strong SL9-specific responses, they exhibited much stronger responses directed against an epitope in protease spanning two amino acid positions associated with resistance to protease inhibitor treatment (PR76-84) (Table 3). Seven out of nine antiretroviral-treated, chronically infected patients tested had a response targeting their autologous variant of the PR76-84 epitope compared to only 1 of 10 chronically infected, untreated subjects (treated versus untreated,P⬍0.01; Fisher’s exact test) (Fig. 1).

As expected, an extensive number of HIV-1 Pol mutations were present in and around the protease epitope in the chron-ically infected, treated individuals (all subjects were receiving a protease inhibitor-based regimen [Table 3]). Most of these mutations were found in the flanking regions, and six of seven patients who had developed the resistance-associated mutation L90M showed reactivity against the PR 76-84 epitope. Since

the L90M mutation was predicted to introduce an additional protease cleavage site at position L89 in Pol (NetChop 3.0), it may have influenced processing and presentation of the epitope.

We next considered what impact this apparent lack of HLA-A2-restricted Gag-specific (SL9) responses in favor of Pol-specific (PR 76-84) responses in treated subjects was having on viral evolution. It should be emphasized that mutations within the targeted epitope previously have been shown to lead to a decay of CTL against the wild-type epitope sequence in chronic infection (22). If, as suggested by our data, protease inhibitor treatment leads to a shift in immunodominant CTL responses from established epitopes (e.g., SL9) to new epitopes (e.g., PR 76-84), then presumably this lack of CTL response may facilitate reversion of CTL-induced escape mu-tations associated with a fitness cost (15, 17, 27). This would be particularly true for SL9 given evidence that mutations in the Gag region apparently reduce viral fitness (15, 21, 24, 27, 30, 35). In support of this hypothesis, as shown in Table 2, protease inhibitor-treated patients exhibited less sequence variation from the consensus in the SL9 epitope, as compared to un-treated, chronically infected individuals. Also, compared to the untreated subjects, the treated subjects had a lower prevalence of the phenylalanine (79F) mutation in position 3 of the SL9 TABLE 2. Treatment alters viral evolution and influence recognition of the SL9 epitopea

Patient no. and sequence type SL9 sequence Frequency (%) HLA-A2 (nM)Affinity to b

CD8⫹T-cell response

(% IFN-␥⫺producing cells)

Wild type Variantc

Reference sequence 77SLYNTVATL85

Chronically infected, untreated

1028 --F--I-V- 100 102 ⫺d

1029 --F--- 70 144 ⫺ 0.286

--F--I---e 30 88 ⫺ 0.451

1030 ---I-V- 90 102 0.184 0.074

---V-e 10 172 0.184 0.104

1034 ---I--- 100 99 ⫺ ⫺

1038 ---I-V- 80 102 ⫺ NAf

---V-e 20 172 ⫺ NA

1051 --F-AI-V- 100 33 ⫺ 0.336

1057 ---I-V- 100 102 0.087 0.078

1058 -I--LI--- 100 151 ⫺ 0.106

1074 --F--I-V- 100 102 0.170 0.265

1079 ---I--- 100 99 0.156 0.123

Chronically infected, treated (viremic)

3002 ---V- 100 172 ⫺ NA

3007 --- 100 162 1.560 NVTg

3011 ---V- 100 172 ⫺ ⫺

3040 ---I-V- 100 102 ⫺ ⫺

3042 --- 100 162 0.356 NVT

3057 ---V- 100 172 ⫺ ⫺

3109 ---L--- 100 140 ⫺ NA

3151 ---V- 100 172 ⫺ NA

3153 --- 100 162 ⫺ NVT

3156 ---I-V- 100 102 ⫺ NA

aPositions 77 to 85 in HXB2 protease.

bAffinity to HLA-A2 as predicted by NetMHC 3.0 (http://www.cbs.dtu.dk/services/NetMHC/). cAutologous viral variant of the SL9 epitope tested.

d

⫺, negative.

eMinor variant.

(4)

epitope (P⫽0.08; Fisher’s exact test) and a lower prevalence of isoleucine (82I) or leucine (82L) in position 6 (P ⫽0.02; Fisher’s exact test). This loss of sequence diversity may reflect fitness-associated reversion back to wild type in the absence of any residual CTL activity. It is also possible that treatment-mediated changes in protease may have led to reduced Gag polypeptide processing (and reduced fitness) and ultimately to the emergence of compensatory changes in Gag (29, 34). These results suggest that treatment-induced sequence varia-tions can alter immunodominance. The cross-sectional nature of our study, however, prohibits us from excluding the possi-bility that the lack of SL9-specific responses in our treated patients may have existed prior to treatment (and indeed may have reflected a poor immunologic response leading to the need to start therapy). A longitudinal study in which patients are studied before treatment and after long-term virologic failure is needed to more fully address these issues; however, such a study may not be feasible given that a large number of treatment-naı¨ve patients would be needed to be studied as they failed several sequential regimens over a period of several years.

To further evaluate how the viral sequence may be changing at different stages of the infection and under the influence of antiviral therapy, we used viral epidemiology signature pattern

analysis (25) to compare HIV-1 sequences obtained from our chronically infected subjects with those observed in eight HLA-A2-positive untreated subjects at primary infection (OP177, OP428, OP474, OP488, OP506, OP539, OP581, and OP583, as described in reference 24). Compared to sequences from untreated individuals with primary infections, the chron-ically infected, untreated individuals showed differences at four amino acid positions within Gag (K30R, V82I, T84V, and E102D). Positions 82 and 84 fall within the SL9 epitope, and position 102 is flanking a highly immunogenic region. In con-trast, compared to sequences from primary infection, se-quences from the chronically infected, treated subjects showed differences at only two positions (84 and 102). We next used the Shannon entropy score (26) to quantify variations in the protein sequence alignments between the different groups. Once again, the numbers of sites in the sequences showing a significant difference were greater between the chronically in-fected, untreated and treated subjects (nine positions) than between the chronically infected, treated subjects and un-treated subjects with primary infections (three positions) (data not shown). Our sequence data therefore indicate that HIV-1 Gag sequences from chronically infected, treated individuals are more similar to untreated subjects with primary infection than chronically infected, untreated subjects. Although we TABLE 3. Antiretroviral treatment alters viral evolution within the HIV-1 protease region that may influence processing and

recognition of the PR 76–84 epitope

Patient no. and sequence type Sequence of PR 76–84a Affinity to

HLA-A2 (nM)b

CD8⫹T-cell responses

(% IFN-␥-producing cells)

Wild type Variantc

Reference sequence 64IEICGHKAIGTVLVGPTPVNIIGRNLLTQI93

Chronically infected, untreated

1028 ---- - - ---- 4,762 ⫺d NVTe

1029 Z---Z---- - - ---- 4,762 ⫺ NVT

1030 ---V----- - - ----L 4,762 ⫺ NVT

1034 ---- - - ---- 4,762 ⫺ NVT

1038 ---- - - ----L 4,762 ⫺ NVT

1051 ---- - - ---- 4,762 0.116 NVT

1057 ---- - - ---- 4,762 ⫺ NVT

1058 ---E---- I- - - ---- 2,101 ⫺ ⫺

1074 ---Q---- - - ---- 4,762 ⫺ NVT

1079 ---- - - ---- 4,762 ⫺ NVT

Chronically infected, treated (viremic)

3002 ---I----- - - N- ----M--- 16,091 ⫺ ⫺

3007 --F----VVZ--- - - S- ----K- 12,353 ⫺ NAf

3011 V---VLS--- - - ----M--- 4,762 0.505 NVT

3040 ---LTS--- - - A- ----M--L 5,931 0.306 0.619

3042 V----Y---- I- - - ---- 2,101 ⫺ ⫺

3057 ---TZ---- - - ----S-Z--L 4,762 0.055 NVT

3109 ---Z-T--S-- I- - - - A- ----MZ-L 2,778 0.427 0.467

3151 ---ITZ--- - - V----VM-K- 2,247 0.247 0.232

3153 ---T--- - - Z- - - - V---M--L 2,247 1.972 0.559

3156 --Z----Z----- - - ----ZL 4,762 0.308 NVT

aThe sequence from positions 64 to 93 in HXB2 protease is shown. Predicted protease cleavage sites are underlined in the reference sequence. The portion of each

sequence corresponding to the PR 76–84 epitope (76LVGPTPVNI84) is shown in boldface. The letter Z represents viral polymorphism at the nucleic acid level giving rise to two different amino acid variants, including the wild-type variant.

bAffinity to HLA-A2 as predicted by NetMHC 3.0. cAutologous viral variant of the PR76–84 epitope tested. d

⫺, negative.

(5)

have not followed the subjects longitudinally, it is possible that the potential reversions to a more consensus B-like viral se-quence may be facilitated by the lack of Gag-specific CTL responses against the wild-type sequence.

In summary, we found an altered distribution of HIV-1 specific CTL epitope recognition in antiretroviral drug-treated subjects. Compared to chronically infected, untreated patients, most chronically infected, treated patients had lost a cellular immune response against the SL9 epitope but gained a new immunodominant response against an epitope in PR 76-84. We thus show that mutations induced by antiretroviral therapy alter the profile of epitopes presented to T cells and thus the direction of the response. The consequences of dual pressures from drugs and CTL need to be considered in monitoring of drug therapy, and as drug therapy becomes more widespread, transmission of dual drug and immune-shaped virus popula-tions will become more prevalent.

The results also open the possibility of manipulating drug-resistant HIV-1 by targeted CTL boosting or using antiretro-viral drugs to drive CTL escape reversion. Considering the important role of the cellular immune responses in durably controlling drug-resistant HIV-1 (14), we suggest that novel therapeutic approaches, including epitope-specific vaccina-tions, could be used to either prohibit the development of specific resistance-associated mutations or mobilize a response against the mutant virus.

Nucleotide sequence accession number.The sequences ob-tained in this study have been submitted to GenBank and were given accession no. EU011832 to EU011921.

This work was supported in part by grants from the NIAID (AI052745, AI055273, and AI44595), the National Institutes of Health UCSF/Gladstone Institute of Virology & Immunology Center for AIDS Research (P30 MH59037 and P30 AI27763), the Center for AIDS Prevention Studies (P30 MH62246), the General Clinical Re-search Center at San Francisco General Hospital (5-MO1-RR00083-37), the AIDS Biology Program of the UCSF ARI, Fogarty grant D43 TW00003, and the Swedish Agency for International Development Cooperation-Sida (2005-001756 and 2006-018).

REFERENCES

1.Alatrakchi, N., C. Duvivier, D. Costagliola, A. Samri, A. G. Marcelin, G. Kamkamidze, M. Astriti, R. Agher, V. Calvez, B. Autran, and C. Katlama. 2005. Persistent low viral load on antiretroviral therapy is associated with T cell-mediated control of HIV replication. AIDS19:25–33.

2.Ali, A., R. Lubong, H. Ng, D. G. Brooks, J. A. Zack, and O. O. Yang.2004. Impacts of epitope expression kinetics and class I downregulation on the antiviral activity of human immunodeficiency virus type 1-specific cytotoxic T lymphocytes. J. Virol.78:561–567.

3.Allen, T. M., D. H. O’Connor, P. Jing, J. L. Dzuris, B. R. Mothe, T. U. Vogel, E. Dunphy, M. E. Liebl, C. Emerson, N. Wilson, K. J. Kunstman, X. Wang, D. B. Allison, A. L. Hughes, R. C. Desrosiers, J. D. Altman, S. M. Wolinsky, A. Sette, and D. I. Watkins.2000. Tat-specific cytotoxic T lymphocytes select for SIV escape variants during resolution of primary viraemia. Nature407: 386–390.

4.Altfeld, M., T. M. Allen, E. T. Kalife, N. Frahm, M. M. Addo, B. R. Mothe, A. Rathod, L. L. Reyor, J. Harlow, X. G. Yu, B. Perkins, L. K. Robinson, J. Sidney, G. Alter, M. Lichterfeld, A. Sette, E. S. Rosenberg, P. J. R. Goulder, C. Brander, and B. D. Walker.2005. The majority of currently circulating human immunodeficiency virus type 1 clade B viruses fail to prime cytotoxic T-lymphocyte responses against an otherwise immunodominant HLA-A2-restricted epitope: implications for vaccine design. J. Virol.79:5000–5005. 5.Bansal, A., E. Gough, S. Sabbaj, D. Ritter, K. Yusim, G. Sfakianos, G.

Aldrovandi, R. A. Kaslow, C. M. Wilson, M. J. Mulligan, J. M. Kilby, and P. A. Goepfert.2005. CD8 T-cell responses in early HIV-1 infection are skewed towards high entropy peptides. AIDS19:241–250.

6.Barbour, J. D., T. Wrin, R. M. Grant, J. N. Martin, M. R. Segal, C. J. Petropoulos, and S. G. Deeks.2002. Evolution of phenotypic drug suscepti-bility and viral replication capacity during long-term virologic failure of

protease inhibitor therapy in human immunodeficiency virus-infected adults. J. Virol.76:11104–11112.

7.Bergmann, C. C., J. D. Altman, D. Hinton, and S. A. Stohlman.1999. Inverted immunodominance and impaired cytolytic function of CD8⫹T cells during viral persistence in the central nervous system. J. Immunol. 163:3379–3387.

8.Betts, M. R., J. P. Casazza, B. A. Patterson, S. Waldrop, W. Trigona, T.-M. Fu, F. Kern, L. J. Picker, and R. A. Koup.2000. Putative immunodominant human immunodeficiency virus-specific CD8⫹T-cell responses cannot be

predicted by major histocompatibility complex class I haplotype. J. Virol. 74:9144–9151.

9.Cao, J., J. McNevin, S. Holte, L. Fink, L. Corey, and M. J. McElrath.2003. Comprehensive analysis of human immunodeficiency virus type 1 (HIV-1)-specific gamma interferon-secreting CD8⫹T cells in primary HIV-1

infec-tion. J. Virol.77:6867–6878.

10.Dalod, M., M. Dupuis, J. C. Deschemin, C. Goujard, C. Deveau, L. Meyer, N. Ngo, C. Rouzioux, J. G. Guillet, J. F. Delfraissy, M. Sinet, and A. Venet. 1999. Weak anti-HIV CD8(⫹) T-cell effector activity in HIV primary infec-tion. J. Clin. Investig.104:1431–1439.

11.Deeks, S. G., R. Hoh, T. B. Neilands, T. Liegler, F. Aweeka, C. J. Petropoulos, R. M. Grant, and J. N. Martin.2005. Interruption of treatment with indi-vidual therapeutic drug classes in adults with multidrug-resistant HIV-1 infection. J. Infect. Dis.192:1537–1544.

12.Deeks, S. G., J. N. Martin, E. Sinclair, J. Harris, T. B. Neilands, H. T. Maecker, E. Hagos, T. Wrin, C. J. Petropoulos, B. Bredt, and J. M. McCune. 2004. Strong cell-mediated immune responses are associated with the main-tenance of low-level viremia in antiretroviral-treated individuals with drug-resistant human immunodeficiency virus type 1. J. Infect. Dis.189:312–321. 13.Deeks, S. G., T. Wrin, T. Liegler, R. Hoh, M. Hayden, J. D. Barbour, N. S. Hellmann, C. J. Petropoulos, J. M. McCune, M. K. Hellerstein, and R. M. Grant. 2001. Virologic and immunologic consequences of discontinuing combination antiretroviral-drug therapy in HIV-infected patients with de-tectable viremia. N. Engl. J. Med.344:472–480.

14.Emu, B., E. Sinclair, D. Favre, W. J. Moretto, P. Hsue, R. Hoh, J. N. Martin, D. F. Nixon, J. M. McCune, and S. G. Deeks.2005. Phenotypic, functional, and kinetic parameters associated with apparent T-cell control of human immunodeficiency virus replication in individuals with and without antiret-roviral treatment. J. Virol.79:14169–14178.

15.Fernandez, C. S., I. Stratov, R. De Rose, K. Walsh, C. J. Dale, M. Z. Smith, M. B. Agy, S.-L. Hu, K. Krebs, D. I. Watkins, D. H. O’Connor, M. P. Davenport, and S. J. Kent.2005. Rapid viral escape at an immunodominant simian-human immunodeficiency virus cytotoxic T-lymphocyte epitope ex-acts a dramatic fitness cost. J. Virol.79:5721–5731.

16.Ferrari, G., W. Neal, J. Ottinger, A. M. Jones, B. H. Edwards, P. Goepfert, M. R. Betts, R. A. Koup, S. Buchbinder, M. J. McElrath, J. Tartaglia, and K. J. Weinhold.2004. Absence of immunodominant anti-Gag p17 (SL9) responses among Gag CTL-positive, HIV-uninfected vaccine recipients ex-pressing the HLA-A*0201 allele. J. Immunol.173:2126–2133.

17.Friedrich, T. C., E. J. Dodds, L. J. Yant, L. Vojnov, R. Rudersdorf, C. Cullen, D. T. Evans, R. C. Desrosiers, B. R. Mothe, J. Sidney, A. Sette, K. Kunstman, S. Wolinsky, M. Piatak, J. Lifson, A. L. Hughes, N. Wilson, D. H. O’Connor, and D. I. Watkins.2004. Reversion of CTL escape-variant immunodeficiency viruses in vivo. Nat. Med.10:275–281.

18.Goulder, P. J., M. A. Altfeld, E. S. Rosenberg, T. Nguyen, Y. Tang, R. L. Eldridge, M. M. Addo, S. He, J. S. Mukherjee, M. N. Phillips, M. Bunce, S. A. Kalams, R. P. Sekaly, B. D. Walker, and C. Brander.2001. Substantial differences in specificity of HIV-specific cytotoxic T cells in acute and chronic HIV infection. J. Exp. Med.193:181–194.

19.Hecht, F. M., M. P. Busch, B. Rawal, M. Webb, E. Rosenberg, M. Swanson, M. Chesney, J. Anderson, J. Levy, and J. O. Kahn.2002. Use of laboratory tests and clinical symptoms for identification of primary HIV infection. AIDS16:1119–1129.

20.Iversen, A. K. N., R. W. Shafer, K. Wehrly, M. A. Winters, J. I. Mullins, B. Chesebro, and T. C. Merigan.1996. Multidrug-resistant human immunode-ficiency virus type 1 strains resulting from combination antiretroviral ther-apy. J. Virol.70:1086–1090.

21.Iversen, A. K. N., G. Stewart-Jones, G. H. Learn, N. Christie, C. Sylvester-Hviid, A. E. Armitage, R. Kaul, T. Beattie, J. K. Lee, Y. Li, P. Chotiyarnwong, T. Dong, X. Xu, M. A. Luscher, K. Macdonald, H. Ullum, B. Klarlund-Pedersen, P. Skinhoj, L. Fugger, S. Buus, J. I. Mullins, E. Y. Jones, P. A. van der Merwe, and A. J. McMichael.2006. Conflicting selective forces affect T cell receptor contacts in an immunodominant human immunodeficiency virus epitope. Nat. Immunol.7:179–189.

22.Jamieson, B. D., O. O. Yang, L. Hultin, M. A. Hausner, P. Hultin, J. Matud, K. Kunstman, S. Killian, J. Altman, K. Kommander, B. Korber, J. Giorgi, and S. Wolinsky.2003. Epitope escape mutation and decay of human im-munodeficiency virus type 1-specific CTL responses. J. Immunol.171:5372– 5379.

(6)

antiretro-viral therapy and cell-mediated immune response on the human immuno-deficiency virus type 1 protease gene. J. Virol.77:6743–6752.

24.Karlsson, A. C., A. K. Iversen, J. M. Chapman, T. de Oliviera, G. Spotts, A. J. McMichael, M. P. Davenport, F. M. Hecht, and D. F. Nixon.2007. Sequential broadening of CTL responses in early HIV-1 infection is associ-ated with viral escape. PLoS ONE2:e225.

25.Korber, B., and G. Myers.1992. Signature pattern analysis: a method for assessing viral sequence relatedness. AIDS Res. Hum. Retrovir. 8:1549– 1560.

26.Korber, B. T. M., K. J. Kunstman, B. K. Patterson, M. Furtado, M. M. McEvilly, R. Levy, and S. M. Wolinsky.1994. Genetic differences between blood- and brain-derived viral sequences from human immunodeficiency virus type 1-infected patients: evidence of conserved elements in the V3 region of the envelope protein of brain-derived sequences. J. Virol.68:7467– 7481.

27.Leslie, A. J., K. J. Pfafferott, P. Chetty, R. Draenert, M. M. Addo, M. Feeney, Y. Tang, E. C. Holmes, T. Allen, J. G. Prado, M. Altfeld, C. Brander, C. Dixon, D. Ramduth, P. Jeena, S. A. Thomas, A. S. John, T. A. Roach, B. Kupfer, G. Luzzi, A. Edwards, G. Taylor, H. Lyall, G. Tudor-Williams, V. Novelli, J. Martinez-Picado, P. Kiepiela, B. D. Walker, and P. J. Goulder. 2004. HIV evolution: CTL escape mutation and reversion after transmission. Nat. Med.10:282–289.

28.Lichterfeld, M., X. G. Yu, D. Cohen, M. M. Addo, J. Malenfant, B. Perkins, E. Pae, M. N. Johnston, D. Strick, T. M. Allen, E. S. Rosenberg, B. Korber, B. D. Walker, and M. Altfeld.2004. HIV-1 Nef is preferentially recognized by CD8 T cells in primary HIV-1 infection despite a relatively high degree of genetic diversity. AIDS18:1383–1392.

29.Mammano, F., C. Petit, and F. Clavel.1998. Resistance-associated loss of viral fitness in human immunodeficiency virus type 1: phenotypic analysis of protease andgagcoevolution in protease inhibitor-treated patients. J. Virol. 72:7632–7637.

30.Martinez-Picado, J., J. G. Prado, E. E. Fry, K. Pfafferott, A. Leslie, S. Chetty, C. Thobakgale, I. Honeyborne, H. Crawford, P. Matthews, T. Pillay, C. Rousseau, J. I. Mullins, C. Brander, B. D. Walker, D. I. Stuart, P. Kiepiela, and P. Goulder. 2006. Fitness cost of escape mutations in p24 Gag in association with control of human immunodeficiency virus type 1. J. Virol. 80:3617–3623.

31.Mason, R. D., M. I. Bowmer, C. M. Howley, M. Gallant, J. C. Myers, and M. D. Grant.2004. Antiretroviral drug resistance mutations sustain or en-hance CTL recognition of common HIV-1 Pol epitopes. J. Immunol.172: 7212–7219.

32.Mason, R. D., and M. D. Grant.2005. A therapy-related point mutation changes the HLA restriction of an HIV-1 Pol epitope from A2 to B57 and enhances its recognition. AIDS19:981–984.

33.Mueller, S. M., B. Schaetz, K. Eismann, S. Bergmann, M. Bauerle, M. Schmitt-Haendle, H. Walter, B. Schmidt, K. Korn, H. Sticht, B. Spriewald, E. G. Harrer, and T. Harrer.2007. Dual selection pressure by drugs and

HLA class I-restricted immune responses on human immunodeficiency virus type 1 protease. J. Virol.81:2887–2898.

34.Myint, L., M. Matsuda, Z. Matsuda, Y. Yokomaku, T. Chiba, A. Okano, K. Yamada, and W. Sugiura.2004. Gag non-cleavage site mutations contribute to full recovery of viral fitness in protease inhibitor-resistant human immu-nodeficiency virus type 1. Antimicrob. Agents Chemother.48:444–452. 35.Nietfield, W., M. Bauer, M. Fevrier, R. Maier, B. Holzwarth, R. Frank, B.

Maier, Y. Riviere, and A. Meyerhans.1995. Sequence constraints and rec-ognition by CTL of an HLA-B27-restricted HIV-1 gag epitope. J. Immunol. 154:2189–2197.

36.Nixon, D. F., S. G. Deeks, B. L. Shacklett, and A. C. Karlsson.2005. Mul-tidrug-resistant, dual-tropic HIV-1 and rapid progression. Lancet365:1924– 1925. (Letter.)

37.Ogg, G. S., X. Jin, S. Bonhoeffer, P. R. Dunbar, M. A. Nowak, S. Monard, J. P. Segal, Y. Cao, S. L. Rowland-Jones, V. Cerundolo, A. Hurley, M. Markowitz, D. D. Ho, D. F. Nixon, and A. J. McMichael.1998. Quantitation of HIV-1-specific cytotoxic T lymphocytes and plasma load of viral RNA. Science279:2103–2106.

38.Price, D. A., G. Scullard, A. Oxenius, R. Braganza, S. A. Beddows, S. Kazmi, J. R. Clarke, G. E. Johnson, J. N. Weber, and R. E. Phillips.2003. Discor-dant outcomes following failure of antiretroviral therapy are associated with substantial differences in human immunodeficiency virus-specific cellular immunity. J. Virol.77:6041–6049.

39.Samri, A., G. Haas, J. Duntze, J.-M. Bouley, V. Calvez, C. Katlama, and B. Autran.2000. Immunogenicity of mutations induced by nucleoside reverse transcriptase inhibitors for human immunodeficiency virus type 1-specific cytotoxic T cells. J. Virol.74:9306–9312.

40.Schmitt, M., E. Harrer, A. Goldwich, M. Bauerle, I. Graedner, J. R. Kalden, and T. Harrer.2000. Specific recognition of lamivudine-resistant HIV-1 by cytotoxic T lymphocytes. AIDS14:653–658.

41.Stratov, I., C. J. Dale, S. Chea, J. McCluskey, and S. J. Kent.2005. Induction of T-cell immunity to antiretroviral drug-resistant human immunodeficiency virus type 1. J. Virol.79:7728–7737.

42.van der Most, R. G., K. Murali-Krishna, J. G. Lanier, E. J. Wherry, M. T. Puglielli, J. N. Blattman, A. Sette, and R. Ahmed.2003. Changing immu-nodominance patterns in antiviral CD8 T-cell responses after loss of epitope presentation or chronic antigenic stimulation. Virology315:93–102. 43.van der Most, R. G., A. Sette, C. Oseroff, J. Alexander, K. Murali-Krishna,

L. L. Lau, S. Southwood, J. Sidney, R. W. Chesnut, M. Matloubian, and R. Ahmed.1996. Analysis of cytotoxic T cell responses to dominant and sub-dominant epitopes during acute and chronic lymphocytic choriomeningitis virus infection. J. Immunol.157:5543–5554.

Referências

Documentos relacionados

This log must identify the roles of any sub-investigator and the person(s) who will be delegated other study- related tasks; such as CRF/EDC entry. Any changes to

a) 30 dias consecutivos de gozo obrigatório, os quais são gozados imediatamente após o nascimento; b) 30 dias de gozo facultativo, seguidos ou interpolados, gozados após o

didático e resolva as ​listas de exercícios (disponíveis no ​Classroom​) referentes às obras de Carlos Drummond de Andrade, João Guimarães Rosa, Machado de Assis,

active antiretroviral therapy are at higher risk of cardiovascular diseases than human immunodeficiency virus infected patients not receiving highly active antiretroviral therapy,

The probability of attending school four our group of interest in this region increased by 6.5 percentage points after the expansion of the Bolsa Família program in 2007 and

A  crescente  urbanização  brasileira  caracteriza‐se,  especialmente  nas  últimas  décadas,  pelo  crescimento  descontrolado  da  população  em  conjunto  com 

If the access to medical care is more limited in developing countries or if there are interactions between birth weight and infant mortality, the estimates derived