The Absence of CD4 + T Cell Count Recovery Despite Receipt of Virologically Suppressive Highly...

10
328 CID 2009:48 (1 February) HIV/AIDS HIV/AIDS INVITED ARTICLE Kenneth H. Mayer, Section Editor The Absence of CD4 + T Cell Count Recovery Despite Receipt of Virologically Suppressive Highly Active Antiretroviral Therapy: Clinical Risk, Immunological Gaps, and Therapeutic Options Lidia Gazzola, Camilla Tincati, Giusi Maria Bellistrı`, Antonella d’Arminio Monforte, and Giulia Marchetti Department of Medicine, Surgery, and Dentistry, Clinic of Infectious Diseases, “San Paolo” Hospital, University of Milan, Milan, Italy Up to 30% of human immunodeficiency virus (HIV)–infected patients who are receiving long-term highly active antiretroviral therapy do not exhibit a marked increase in the CD4 + T cell count, despite achieving complete suppression of the HIV load. These patients are referred to as “immunological nonresponders.” When treating immunological nonresponders, the practicing clinician has several questions, including questions about the clinical risk associated with persistent immunodeficiency and about possible approaches to treatment that would provide clinical and immunological benefits. However, tentative answers to these questions require investigations of the mechanisms that underlie the lack of immune recovery, because only the deepest comprehension of the immunological gaps underlying functional defects will allow administration of highly targeted and efficacious treatment strategies. The aim of our review is to provide a thorough assessment of the clinical implications of a lack of increase in the CD4 + T cell count in immunological nonresponders, to examine the immunological gaps limiting recovery of the CD4 + T cell count, and to note possible therapeutic avenues, which may offer clinicians guidance regarding how to most efficaciously treat these critical patients. IMMUNOLOGICAL FAILURE AND HAART In the era of HAART, HIV/AIDS clinicians have observed a remarkable reduction in AIDS-related morbidity and mortality rates. In fact, HAART has been shown to improve survival rates among HIV-infected individuals through its ability to reduce the HIV load to undetectable levels and to increase the CD4 + lymphocyte count in peripheral blood (defined as a full re- sponse) [1, 2]. However, 15%–30% of patients have discordant responses to long-term HAART consisting of a lack of increase in the CD4 + T cell count but full suppression of HIV replication; these people are referred to as “immunological nonresponders” (INRs) [3, 4]. From a clinical standpoint, lack of CD4 + immune reconstitution during receipt of effective HAART represents an everyday issue in the clinical management of HIV/AIDS. In Received 28 May 2008; accepted 16 September 2008; electronically published 5 January 2009. Reprints or correspondence: Dr. Giulia Marchetti, Dept. of Medicine, Surgery, and Dentistry, Clinic of Infectious Diseases, “San Paolo” Hospital, University of Milan, Via A. Di Rudinı `, 8, 20142 Milan, Italy ([email protected]). Clinical Infectious Diseases 2009; 48:328–37 2009 by the Infectious Diseases Society of America. All rights reserved. 1058-4838/2009/4803-0010$15.00 DOI: 10.1086/595851 particular, clinicians question whether INRs have an increased risk of clinical progression to AIDS and whether it is possible to identify early predictive factors of immunological failure during HAART. From a therapeutic standpoint, failure of CD4 + cell recon- stitution during receipt of virologically suppressive HAART in- dicates the need for alternative treatment strategies. However, the essential premise of studies of therapeutic options in this clinical context is the deep comprehension of the pathogenetic mechanisms that underlie immunological failure. We assessed the clinical implications of a lack of a recovery in the CD4 + cell count among INRs, the immunological gaps limiting CD4 + cell count recovery, and the possible therapeutic avenues that may offer clinicians guidance regarding the most effective treat- ments for these critical patients. INVESTIGATING THE CLINICAL CORRELATES OF IMMUNOLOGICAL FAILURE DURING HAART: ARE INRS AT INCREASED RISK OF HIV/ AIDS DISEASE PROGRESSION AND DEATH? An essential premise in the assessment of clinical correlates of insufficient immunological response to long-term HAART is by guest on May 2, 2016 http://cid.oxfordjournals.org/ Downloaded from

Transcript of The Absence of CD4 + T Cell Count Recovery Despite Receipt of Virologically Suppressive Highly...

328 • CID 2009:48 (1 February) • HIV/AIDS

H I V / A I D S I N V I T E D A R T I C L EKenneth H. Mayer, Section Editor

The Absence of CD4+ T Cell Count Recovery DespiteReceipt of Virologically Suppressive Highly ActiveAntiretroviral Therapy: Clinical Risk, Immunological Gaps,and Therapeutic Options

Lidia Gazzola, Camilla Tincati, Giusi Maria Bellistrı, Antonella d’Arminio Monforte, and Giulia MarchettiDepartment of Medicine, Surgery, and Dentistry, Clinic of Infectious Diseases, “San Paolo” Hospital, University of Milan, Milan, Italy

Up to 30% of human immunodeficiency virus (HIV)–infected patients who are receiving long-term highly active antiretroviraltherapy do not exhibit a marked increase in the CD4+ T cell count, despite achieving complete suppression of the HIV load.These patients are referred to as “immunological nonresponders.” When treating immunological nonresponders, the practicingclinician has several questions, including questions about the clinical risk associated with persistent immunodeficiency andabout possible approaches to treatment that would provide clinical and immunological benefits. However, tentative answersto these questions require investigations of the mechanisms that underlie the lack of immune recovery, because only thedeepest comprehension of the immunological gaps underlying functional defects will allow administration of highly targetedand efficacious treatment strategies. The aim of our review is to provide a thorough assessment of the clinical implicationsof a lack of increase in the CD4+ T cell count in immunological nonresponders, to examine the immunological gaps limitingrecovery of the CD4+ T cell count, and to note possible therapeutic avenues, which may offer clinicians guidance regardinghow to most efficaciously treat these critical patients.

IMMUNOLOGICAL FAILURE AND HAART

In the era of HAART, HIV/AIDS clinicians have observed a

remarkable reduction in AIDS-related morbidity and mortality

rates. In fact, HAART has been shown to improve survival rates

among HIV-infected individuals through its ability to reduce

the HIV load to undetectable levels and to increase the CD4+

lymphocyte count in peripheral blood (defined as a full re-

sponse) [1, 2]. However, 15%–30% of patients have discordant

responses to long-term HAART consisting of a lack of increase

in the CD4+ T cell count but full suppression of HIV replication;

these people are referred to as “immunological nonresponders”

(INRs) [3, 4]. From a clinical standpoint, lack of CD4+ immune

reconstitution during receipt of effective HAART represents an

everyday issue in the clinical management of HIV/AIDS. In

Received 28 May 2008; accepted 16 September 2008; electronically published 5 January2009.

Reprints or correspondence: Dr. Giulia Marchetti, Dept. of Medicine, Surgery, and Dentistry,Clinic of Infectious Diseases, “San Paolo” Hospital, University of Milan, Via A. Di Rudinı, 8,20142 Milan, Italy ([email protected]).

Clinical Infectious Diseases 2009; 48:328–37� 2009 by the Infectious Diseases Society of America. All rights reserved.1058-4838/2009/4803-0010$15.00DOI: 10.1086/595851

particular, clinicians question whether INRs have an increased

risk of clinical progression to AIDS and whether it is possible

to identify early predictive factors of immunological failure

during HAART.

From a therapeutic standpoint, failure of CD4+ cell recon-

stitution during receipt of virologically suppressive HAART in-

dicates the need for alternative treatment strategies. However,

the essential premise of studies of therapeutic options in this

clinical context is the deep comprehension of the pathogenetic

mechanisms that underlie immunological failure. We assessed

the clinical implications of a lack of a recovery in the CD4+

cell count among INRs, the immunological gaps limiting CD4+

cell count recovery, and the possible therapeutic avenues that

may offer clinicians guidance regarding the most effective treat-

ments for these critical patients.

INVESTIGATING THE CLINICAL CORRELATESOF IMMUNOLOGICAL FAILURE DURINGHAART: ARE INRS AT INCREASED RISK OF HIV/AIDS DISEASE PROGRESSION AND DEATH?

An essential premise in the assessment of clinical correlates of

insufficient immunological response to long-term HAART is

by guest on May 2, 2016

http://cid.oxfordjournals.org/D

ownloaded from

HIV/AIDS • CID 2009:48 (1 February) • 329

the definition of immunological response itself. There is no

clear-cut agreement on how to assess an insufficient immune

response to HAART, with particular regard to the adequate

time to evaluation of immune response after the commence-

ment of HAART. Although an interval of 12 months may be

too premature to evaluate immune response to HAART [5], it

is our opinion that the broadly used criteria of an increase in

the CD4+ T cell count of !30% and an absolute CD4+ T cell

count �200 cells/mL during the first 6–12 months of HIV-

suppressive HAART identifies immunological nonresponse [6],

because immune response after 3–6 months of HAART is pre-

dictive of both immune reconstitution and HIV-related mor-

bidity and mortality in the long term [7, 8]. However, this

definition may identify an overly heterogeneous population and

fail to discriminate between a real long-term nonresponse and

a delayed response.

Kaufmann et al. [7] demonstrated that an incomplete im-

mune response in the short term led to an elevated risk of

Centers for Disease Control and Prevention category B and C

events in the long term, indicating an augmented risk of disease

progression in INRs. Additional observational studies have de-

fined an approximately doubled relative risk of clinical pro-

gression to AIDS and an increased risk of mortality in INRs,

compared with patients who had a complete response [9–13].

In addition, a new clinical concern was recently raised by

Gutierrez et al. [12], who showed that, despite having similar

proportions of new AIDS-defining events, INRs had an overall

higher rate of non–AIDS-related mortality, compared with pa-

tients who had a complete response, causing additional clinical

concern about the long-term risks of subclinical immunode-

ficiency. Accordingly, guidelines of the Department of Health

and Human Services (DHHS) prefer to define immunological

failure as the lack of an increase in the CD4+ T cell count to

more than 350–500 cells/mL after receipt of 4–7 years of effective

HAART [14], given recent data linking these immunological

thresholds with the risk of non-AIDS clinical events [15].

INVESTIGATION OF FACTORS ASSOCIATEDWITH IMMUNOLOGICAL FAILURE DURINGLONG-TERM HAART: WHICH FACTORSPREDICT IMMUNOLOGICAL NONRESPONSE?

Having defined an overall increased risk of HIV/AIDS disease

progression in INRs, we still need to assess whether the clinician

can depend on specific factors for early identification of which

patients are most likely to experience insufficient immune re-

covery while receiving HAART. As illustrated in table 1, several

factors have been individually associated with immunological

failure during HAART, and yet only those below have been

invariably proven to predict immunological nonresponse.

Age has been reported to have a significant impact on im-

mune recovery: the older the patient is, the more likely that

he or she will experience delayed immune reconstitution [18].

Indeed, older age has been associated with a decrease of thymic

function and other regenerative mechanisms, thus explaining

its role as an independent predictive factor of impaired immune

recovery [7, 10, 11, 16, 19]. In addition, recent data from cohort

studies suggest that immune recovery in older patients may

hide a more profound functional impairment, as evidenced by

a persistent increased risk of AIDS-related events in these pa-

tients, even after adjustment for CD4+ T cell counts [20, 21].

Another factor presumed to affect immune recovery during

HAART is concurrent viral hepatitis. In a recent meta-analysis,

Miller et al. [22] showed that increases in the CD4+ T cell count

during HAART are significantly lower in the course of hepatitis

C virus (HCV) coinfection. The biological rationale posits that

there are higher levels of T cell activation and death in HIV-

HCV–coinfected patients and that there is a possible direct

negative effect of HCV infection and replication inside lym-

phocyte subpopulations [23, 24]. However, the actual impact

of HCV infection on immune recovery in HIV-infected patients

still remains a matter of controversy, given the numerous con-

founding factors often present in the HCV-infected population,

such as a history of injection drug use, poor adherence to

therapy, and decreased access to health care [25–28]. Of these

factors, adherence to antiretroviral therapy has been proven to

be a strong independent predictor of recovery in the CD4+ T

cell count and the ultimate cause of impaired immune recovery

in injection drug users [29, 30].

With regard to immunovirological factors, nadir CD4+ T cell

count is the most common and most reliable determinant of

suboptimal immune recovery during HAART [7, 16, 17, 31,

32]. From a mechanistic standpoint, differences in CD4+ T cell

nadir are indicators of differences in immunological regulatory

functions over T cell homeostasis that might affect immune

recovery competence [33]. However, the sole quantification of

nadir CD4+ T cell count may fail to qualitatively estimate the

immunological mechanism(s) that hinder CD4+ T cell count

rescue, indicating a need for a more detailed assessment of

immunological gaps associated with nadir-driven T cell

homeostasis.

The identification of immunopathogenetic models behind

the negative impact of age, HCV coinfection, and nadir CD4+

cell count on immune recovery raises an additional clinical

question: are there immunological, virological, or genetic mark-

ers that can be directly exploited, from bench to bedside, to

predict immunological failure during HAART in at-risk pa-

tients? Although Spritzler et al. [34] demonstrate that peripheral

immunophenotype has a limited ability to predict HAART re-

sponse, only the most thorough investigation of immunovi-

rological correlates of discordant responses will allow for a

targeted immune-based treatment approach.

by guest on May 2, 2016

http://cid.oxfordjournals.org/D

ownloaded from

330

Tabl

e1.

Ove

rvie

wof

the

maj

orst

udie

sev

alua

ting

pred

ictiv

efa

ctor

sof

shor

t-ter

man

dlo

ng-te

rmim

mun

olog

ical

resp

onse

toan

tiret

rovi

ral

ther

apy

(ART

).

Ref

eren

ceYe

arS

tudy

popu

latio

nA

RT

regi

men

(s)

Defi

nitio

nof

“inc

ompl

ete

imm

unol

ogic

alre

spon

se”

No.

(%)

ofpa

tient

sS

tatis

tical

anal

ysis

Ris

kfa

ctor

sfo

und

byst

atis

tical

anal

ysis

Gra

bar

etal

.[9

]20

0022

36P

I-nai

vepa

tient

s(7

7.3%

wer

eA

RT

expe

rienc

edan

d22

.7%

wer

eA

RT

naiv

e)

2N

RTI

spl

usa

PI

Incr

ease

inth

eC

D4+

Tce

llco

unt

!50

cells

/mL

at6

mon

ths

afte

rA

RT

intr

o-du

ctio

n,w

itha

decr

ease

inth

epl

asm

aH

IVlo

ad11

log 1

0co

pies

/mL

ora

plas

ma

HIV

load

!10

00co

pies

/mL

387

(17.

3)K

rusk

al-W

allis

and

x2

test

sID

U,

high

erba

selin

eC

D4+

Tce

llco

unt,

and

low

erba

selin

epl

asm

aH

IVlo

ad

Kau

fman

net

al.

[16]

2002

95P

atie

nts

(52%

wer

eN

RTI

expe

ri-en

ced

and

48%

wer

eN

RTI

naiv

e)

2N

RTI

spl

usa

PI,

2N

RTI

spl

usan

NN

RTI

,an

NR

TIpl

us2

PIs

,an

NN

RTI

plus

aP

I,or

2N

RTI

s

Tota

lCD

4+T

cell

coun

t!50

0ce

lls/m

Lat

4ye

ars

afte

rA

RT

intr

oduc

tion,

with

plas

ma

HIV

load

!40

0co

pies

/mL

22(2

3)U

niva

riate

logi

stic

regr

essi

onLo

wer

nadi

rC

D4+

Tce

llco

unt

and

low

erba

selin

eC

D4+

Tce

llco

unt

Mul

tivar

iate

logi

stic

regr

essi

onLo

wer

nadi

rC

D4+

Tce

llco

unt

Dro

nda

etal

.[5

]20

0225

5A

RT-

naiv

epa

tient

s2

NR

TIs

plus

aP

Ior

2N

RTI

spl

usan

NN

RTI

Incr

ease

inth

eC

D4+

Tce

llco

unt

!10

0ce

lls/m

L24

mon

ths

afte

rA

RT

intr

o-du

ctio

n,w

ithpl

asm

aH

IVlo

ad!50

copi

es/m

L

42(1

6.5)

Uni

varia

telo

gist

icre

gres

sion

Low

erba

selin

epl

asm

aH

IVlo

ad,

high

erba

selin

eC

D4+

Tce

llco

unt,

and

prev

i-ou

sID

U

Mul

tivar

iate

logi

stic

regr

essi

onP

revi

ous

IDU

Flor

ence

etal

.[1

7]20

0378

0P

atie

nts

(61%

wer

eN

RTI

expe

ri-en

ced

and

39%

wer

eN

RTI

naiv

e)

NR

TIs

plus

aP

I,N

RTI

spl

usan

NN

RTI

,or

anN

NR

TIpl

usa

PI

Incr

ease

inth

eC

D4+

Tce

llco

unt

!50

cells

/mL

bym

onth

6or

!75

cells

/mL

bym

onth

12,

with

apl

asm

aH

IVlo

ad!50

0co

pies

/mL

225

(29)

Uni

varia

telo

gist

icre

gres

sion

Old

erag

e,in

terv

albe

twee

nH

IVin

fec-

tion

diag

nosi

sto

com

men

cem

ent

ofH

AA

RT,

NR

TIex

perie

nce,

low

erna

dir

CD

4+T

cell

coun

t,gr

eate

rin

crea

sein

CD

4+T

cell

coun

tw

ithpr

evio

usA

RT,

low

erba

selin

ean

dpe

akpl

asm

aH

IVlo

ads,

and

prev

ious

KS

Mul

tivar

iate

logi

stic

regr

essi

onO

lder

age,

low

erna

dir

CD

4+T

cell

coun

t,lo

wer

base

line

CD

4+T

cell

coun

t,gr

eate

rin

crea

sein

CD

4+T

cell

coun

tw

ithpr

evio

usA

RT,

and

low

erba

selin

epl

asm

aH

IVlo

ad

by guest on May 2, 2016

http://cid.oxfordjournals.org/D

ownloaded from

331

Kau

fman

net

al.

[7]

2005

293

AR

T-na

ive

patie

nts

2N

RTI

spl

usa

PI,

2N

RTI

spl

usan

NN

RTI

,an

NR

TIpl

us2

PIs

,an

NR

TIpl

usan

NN

RTI

and

aP

I,or

3N

RTI

s

Tota

lCD

4+T

cell

coun

t!50

0ce

lls/m

L5

year

saf

ter

AR

Tin

trod

uctio

n,w

itha

plas

ma

HIV

load

!10

00co

pies

/mL

105

(35.

8)U

niva

riate

logi

stic

regr

essi

onO

lder

age,

dura

tion

ofH

IVin

fect

ion,

HC

Van

tibod

ypo

sitiv

ity,

CD

Cin

fec-

tion

cate

gorie

sB

and

C,

and

low

erba

selin

eC

D4+

Tce

llco

unt

Mul

tivar

iate

logi

stic

regr

essi

onO

lder

age,

dura

tion

ofH

IVin

fect

ion,

and

low

erba

selin

eC

D4+

Tce

llco

unt

Moo

reet

al.

[10]

2005

1527

AR

T-na

ive

patie

nts

2N

RTI

spl

usa

PI

or2

NR

TIs

plus

anN

NR

TIIn

crea

sein

the

CD

4+T

cell

coun

t!50

cells

/mL

3–9

mon

ths

afte

rA

RT

intr

o-du

ctio

n,w

itha

plas

ma

HIV

load

!50

0co

pies

/mL

235

(15.

4)M

ultiv

aria

telo

gist

icre

gres

sion

Old

erag

ean

dH

AA

RT

back

bone

of3T

Cpl

usZD

V

Nic

astr

iet

al.

[11]

2005

2143

Pat

ient

s(7

4.2%

wer

eN

RTI

expe

ri-en

ced

and

25.8

%w

ere

NR

TIna

ive)

2N

RTI

spl

usa

PI

Incr

ease

inth

eC

D4+

Tce

llco

unt

�10

0ce

lls/m

L12

mon

ths

afte

rA

RT

intr

o-du

ctio

n,w

itha

plas

ma

HIV

load

!50

0co

pies

/mL

336

(15.

7)M

ultiv

aria

telo

gist

icre

gres

sion

Old

erag

e,A

RT

naiv

e,an

dba

selin

eC

D4+

Tce

llco

unt

�35

0ce

lls/m

L

Mul

tinom

iall

ogis

ticre

gres

-si

on(c

ompa

red

with

com

-pl

ete

resp

onse

)

Bas

elin

eC

D4+

Tce

llco

unt

�35

0ce

lls/

mL

Gut

ierr

ezet

al.

[12]

2008

650

AR

T-na

ive

patie

nts

2N

RTI

spl

usa

PI,

2N

RTI

spl

usan

NN

RTI

,3

NR

TIs,

oran

NN

RTI

plus

aP

I

Incr

ease

inth

eC

D4+

Tce

llco

unt

!50

cells

/mL

12m

onth

saf

ter

AR

Tin

tro-

duct

ion,

with

apl

asm

aH

IVlo

ad!50

0co

pies

/mL

108

(16.

6)U

niva

riate

logi

stic

regr

essi

onO

lder

age,

patie

ntse

x,lo

wer

base

line

plas

ma

HIV

load

,an

dhi

gher

base

line

CD

4+T

cell

coun

t

Mul

tivar

iate

logi

stic

regr

essi

onO

lder

age

and

IDU

Tan

etal

.[1

3]20

0840

4A

RT-

naiv

epa

tient

s2

NR

TIs

plus

aP

I,2

NR

TIs

plus

anN

NR

TI,

or3

NR

TIs

Incr

ease

inth

eC

D4+

Tce

llco

unt

!50

cells

/mL

3–9

mon

ths

afte

rA

RT

intr

o-du

ctio

n,w

ithan

unde

tect

-ab

lepl

asm

aH

IVlo

ad

35(8

.7)

Mul

tivar

iate

logi

stic

regr

essi

onN

ofa

ctor

s

NO

TE

.C

DC

,Cen

ters

forD

isea

seC

ontr

olan

dP

reve

ntio

n;ID

U,i

njec

tion

drug

use;

KS

,Kap

osis

arco

ma;

NN

RTI

,non

nucl

eosi

dere

vers

e-tr

ansc

ripta

sein

hibi

tor;

NR

TI,n

ucle

osid

ere

vers

e-tr

ansc

ripta

sein

hibi

tor;

PI,

prot

ease

inhi

bito

r;3T

C,

lam

ivud

ine;

ZDV,

zido

vudi

ne.

by guest on May 2, 2016

http://cid.oxfordjournals.org/D

ownloaded from

332 • CID 2009:48 (1 February) • HIV/AIDS

Figure 1. Differences in CD4+ T cell dynamics between persons who experienced a full immunological response (FRs; A) and immunologicalnonresponders (INRs; B). Compared with FRs, INRs are notable for reduced bone marrow and thymic output of naive T cells in peripheral circulation,greater HIV- and antigen-driven T cell activation, and reduced content of T regulatory cells, ultimately leading to increase cellular death by apoptosis.Despite significantly lower CD4+ T cell counts in the periphery, a similar stimulation of the IL-7 compensatory loop is displayed by INRs and FRs.However, compared with FRs, INRs are characterized by reduced IL-7R expression on different T cell populations, thus potentially limiting the compensatoryeffect of IL-7. Different T cell pools are represented: recent thymic emigrants, naive T cells, resting memory T cells, activated T cells, T regulatorycells, and hematopoietic stem cell precursors (HSCPs). The size of the boxes and the number of cells represent the relative amount of each cell pooland are not to scale. IL-7R is represented on T cell surface according to its expression, not to scale. Ag, antigen.

INVESTIGATION OF THE PATHOGENETICCORRELATES OF INRS: WHICHIMMUNOVIROLOGICAL AND GENETICMECHANISMS LIMIT IMMUNE RECOVERYDURING LONG-TERM HAART?

Failure in De Novo CD4+ T Cell Production

The pathogenesis of immunological nonresponse may be sec-

ondary to specific failure of the T cell armamentarium ma-

chinery (i.e., failure of the bone marrow to produce hemato-

poietic stem cells) or to a deficiency in thymic output (figure

1) [35].

The role of bone marrow. Decreased bone marrow pro-

genitor cell growth and abnormal stromal microenvironment

have been described in patients with HIV/AIDS [36] and have

been partly restored after the commencement of HAART [37,

38]. Recent observations support the hypothesis that the lack

of immune recovery in INRs may be due, at least in part, to

persistent bone marrow impairment, despite receipt of HAART,

that is possibly characterized by altered clonogenic capability

and stromal cell function (figure 1) [39–41].

The role of thymic output. The failure to restore circulating

CD4+ T cells during HAART may partially be caused by de-

ficiencies in thymopoietin [42–45]. Indeed, several studies have

demonstrated a trend toward smaller thymuses and lower thy-

mopoietin levels in INRs (figure 1) [35, 46–48]. Given that a

hypofunctional thymus may account for the immunological

failure in INRs, a question remains whether the thymus itself

is unable to respond to thymopoietic signals (e.g., IL-7) or

whether the latter are insufficient to drive thymic-dependent

immune reconstitution.

The role of IL-7. IL-7, which is mainly produced by thymic

and bone marrow stromal cells, is a vital cytokine for thymocyte

development for which production is up-regulated in lympho-

penic conditions [49, 50]. A relative deficiency in IL-7 pro-

duction or function may occur in INRs. Studies have found

that plasma IL-7 levels in INRs are comparable to those in

patients who had complete responses, suggesting maintenance

of the IL-7 compensatory loop [47, 48]. Marziali et al. [47]

reported a reduction in IL-7R expression on CD4+ T cells ob-

tained from INRs, compared with CD4+ T cells obtained from

persons who had complete responses, as well as a positive cor-

relation between peripheral percentage of CD4+ T cells and

those expressing IL-7R, leading to speculation about defective

IL-7R expression. Additional studies are needed to aid com-

by guest on May 2, 2016

http://cid.oxfordjournals.org/D

ownloaded from

HIV/AIDS • CID 2009:48 (1 February) • 333

prehension about the role of thymopoiesis and IL-7 signaling

in INRs [50, 51].

Excessive CD4+ T Cell Destruction

As illustrated in figure 1, INRs may also be characterized by

augmented levels of CD4+ T cell loss in the periphery [35].

CD4+ T cell hyperactivation. CD4+ T cell hyperactivation

has been demonstrated to persist even after HAART virological

suppression occurs and to have a significant effect on recovery

of the CD4+ T cell count during HAART [52, 53]. Indeed,

despite complete suppression of the HIV load, INRs maintain

CD4+ T cell hyperactivation comparable to that in patients who

have not experienced viral suppression (figure 1) [48, 54].

Although highlighting the possible pathogenetic role of en-

hanced CD4+ T cell activation in insufficient CD4+ T cell count

recovery, these findings also raise questions on the driver(s) of

persistently elevated CD4+ T cell activation.

Ongoing viral replication. Even in the context of complete

viral suppression, INRs have been shown to have higher levels

of proviral DNA in total, memory, and naive CD4+ T cells,

reinforcing the role of increased HIV antigen–driven CD4+ T

cell activation as a driving force of continuous loss of CD4+ T

cells in INRs [48]. On the one hand, despite control of plasma

viral loads, the persistence of residual low-level viral replication

at levels less than the detection limit of the most-sensitive meth-

ods, either in blood or in other compartments, maintains the

HIV DNA burden in the reservoir; on the other hand, it rep-

resents a continuous trigger of immune activation [55, 56].

Persistent antigenic stimulation. CD4+ T cell hyperacti-

vation may also be secondary to ongoing chronic inflammatory

disease. Recently, a pathogenetic model of increased translo-

cation of microbial bioproducts from the gastrointestinal lumen

has been proposed as a continuous trigger of immune activation

in patients with HIV/AIDS [57, 58]. Brenchley et al. [59] el-

egantly demonstrated that high levels of plasma lipopolysac-

charide—an indicator of microbial translocation—in HIV-in-

fected patients correlate with immune hyperactivation and are

only partially restored by HAART. We reported a consistent

trend toward higher lipopolysaccharide levels in INRs, com-

pared with levels in persons who had a complete response, that

correlated with the proportion of activated Ki67+ CD4+ and

CD8+ cells. Taken together, these data allow speculation that

microbial translocation, by perpetuating peripheral CD4+ T cell

activation, may contribute to the inefficient recovery in the

CD4+ T cell count in INRs [60]. Consistent with this finding

is the fact that HIV-driven dysfunction of CD4+ T cell ho-

meostasis in the gut mucosa, with local immune activation, has

been shown to persist after commencement of HAART [61–

64]. However, a clear-cut correlation between mucosal immune

restoration and immunological and clinical outcome during

HAART is still missing [64, 65].

Immunoregulatory mechanisms. T regulatory cells are a

specialized T cell subpopulation with the ability to down-mod-

ulate immune activation and function [66]. Indeed, Marziali

et al. [47] reported a significant reduction in T regulatory cell

count in INRs, compared with patients who had complete re-

sponses, inversely correlating with activated CD4+ T cells, al-

lowing one to hypothesize that persistently low levels of T

regulatory cells, which are unable to turn off immune activa-

tion, could partly account for immune reconstitution failure

in INRs [47].

Genetic Influence

A role of genetic polymorphisms involved in CD4+ T cell ho-

meostasis has also been postulated in dictating the magnitude

of recovery in the CD4+ T cell count during HAART. In a

recent multivariate analysis, Haas et al. [67] found a significant

association between CD4+ T cell count recovery and polymor-

phisms in genes encoding TNF-related apoptosis-induced li-

gand, TNF-a, Bcl-2–interacting molecule, and IL-15/IL-15R.

Similarly, diverse polymorphisms in chemokine or chemokine

receptors and HLA genes have been associated with the re-

sponse to HAART, although different association patterns were

demonstrated by different authors [68–70]. In conclusion, it

seems reasonable that multiple genetic variants may be involved

in the pathogenesis of immunological nonresponse vis-a-vis

complex immune phenotypes in these individuals.

INVESTIGATION OF THE THERAPEUTICOPTIONS FOR INRS: WHICH STRATEGIES MAYIMPROVE IMMUNE RECOVERY IN INRS?

Having assessed the clinical risk and the immunological defects

that may be potential targets for adjuvant approaches in INRs,

additional questions arise on the actual direction for an effective

therapeutic strategy. Should we aim at improving peripheral T

cell production and expansion or rather, should we seek im-

mune activation control, enhancing peripheral survival of CD4+

T cells?

MOLECULES THAT SUSTAIN THE CD4+ T CELLCOUNT IN THE PERIPHERY

IL-2. Thus far, several controlled studies have been conducted

to investigate the effect of IL-2 in INRs, concluding that IL-2

is safe and efficacious in inducing a rapid and significant re-

constitution of the CD4+ T cell compartment, with no signif-

icant impact on HIV load [71–73]. Most interestingly, data

showing no HIV-related events among IL-2–treated patients,

compared with recipients of HAART alone, suggest that the IL-

2–driven increases in the CD4+ T cell count may also be effective

in preserving an adequate cellular immunity [72, 74–76], even

though the ultimate evidence of IL-2 clinical impact is still being

sought [14, 77].

by guest on May 2, 2016

http://cid.oxfordjournals.org/D

ownloaded from

334 • CID 2009:48 (1 February) • HIV/AIDS

Nonetheless, major constraints to the clinical use of IL-2 in

INRs include a time-limited immune benefit in sustaining CD4+

T cell counts and inefficacy in a minor group of nonresponding

patients. Although a possible way to overcome such limitations

has recently been proposed (consisting of induction mainte-

nance strategies of long-term IL-2 treatment [78]), it is clear

that IL-2 immunotherapy alone for the treatment of INRs failed

to meet the initial expectations. Given these controversies and

possible drug-associated adverse effects in IL-2 recipients,

DHHS guidelines recommend the use of IL-2 immunotherapy

only in the context of clinical trials [14].

IL-7. Two recent trials have demonstrated a sustained dose-

dependent increase in naive and memory CD4+ and CD8+ T

cells after administration of IL-7 to HIV-infected patients [79–

81]. However, the opportunity to use IL-7 as an immune ad-

juvant in INRs is still controversial, given evidence that en-

dogenous plasma IL-7 levels are already elevated in INRs. In

our opinion, a possible way to understand this controversy is

analysis of IL-7 production and signaling in INRs that would

yield evidence on the functional status of the IL-7 axis and,

thus, to the possible benefits of its exogenous administration.

However, DHHS guidelines recommend use of IL-7 immu-

notherapy only in the context of clinical trials [14].

Modulation of T regulatory cells. Although they are an

intriguing target for immune therapy, no clear-cut consensus

has been reached on whether and how to exogenously modulate

T regulatory cell function and/or activity [82]. Thus far, data

on the effects of IL-2 and IL-7 immunotherapy on T regulatory

cells have been discordant [83, 84], indicating the importance

of investigating the role of cytokine-based approaches to this

particular T cell population.

STRATEGIES AIMED AT TURNING OFF IMMUNEHYPERACTIVATION

Antiretroviral therapy strategies. Differences in CD4+ T cell

response may exist among different drug classes. Controversial

data on a greater immunological efficacy of protease inhibitor–

containing regimens have been generated [85]. Indeed, a recent

randomized clinical trial that compared first-line regimens in-

cluding nonnucleoside reverse-transcriptase inhibitors versus

protease inhibitor reported a superior immune response to the

latter regimen, but this occurred only in the long term, and

there was doubt about its clinical significance [86]. Further-

more, data on thymidine analogue–associated mitochondrial

toxicity and oxidative stress have accumulated recently. These

findings suggest a role for thymidine analogues—including nu-

cleoside reverse-transcriptase–based regimens—in cell dys-

function [87], possibly hampering recovery of the CD4+ T cell

count.

The negative impact of low-level residual HIV levels to less

than suppressive threshold during CD4+ immune recovery in

INRs led to the hypothesis that intensification of HAART may

improve immunological responses in INRs [88–92]. The avail-

ability of new drug classes, such as fusion and integrase inhib-

itors, which provide the strongest suppression of HIV loads

and HIV DNA burden and the greatest immunological efficacy

[93, 94], makes this issue more relevant and current. Recently,

CCR5 antagonists have been suggested to have the potential to

yield the greatest CD4+ T cell count gains, given their specific

effect in the suppression of immune activation [95]. Indeed, a

very recent meta-analysis investigated the differences in in-

creases in the CD4+ T cell count among patients enrolled in

phase II/III trials of new antiretrovirals and found an enhanced

CD4+ T cell response in recipients of CCR5 antagonist–con-

taining regimens, compared with recipients of other regimens,

that was independent of suppression of the HIV load [96].

Even if the durability and clinical meaning of this finding re-

main to be determined, these data support deeper investigation

of this approach in INRs.

In addition to identification of the “best” antiretroviral strat-

egy for INRs, a major challenge for clinicians is the identifi-

cation of the “best” CD4+ T cell range for initiation of HAART.

Indeed, given the persistence of different trajectories for the

recovery of the CD4+ T cell count based on different baseline

ranks, with some individuals barely achieving a protective CD4+

T cell count of !200 cells/mL after 5 years of HAART, the impact

of commencement of HAART should be evaluated in patients

with a high risk of becoming INRs, such as older patients

[21, 97].

Immunosuppressive agents. Because there has been inter-

est in “turning off” excessive immune activation, various im-

munosuppressive agents have been investigated [98–103]. In

addition to providing positive data on the efficacious control

of immune activation, these trials have also demonstrated an

immunological benefit in terms of increases in the CD4+ T cell

count [99, 101]. Unfortunately, trials assessing the benefit of

administration of immunosuppressive drugs to INRs are still

lacking, the major concern being the potential risk of admin-

istering immunosuppressive agents to patients with a highly

depleted CD4+ T cell compartment. In our opinion, a prom-

ising approach may be the identification of targeted interven-

tions to interact with pathways of immune activation that are

specific to INRs, thus allowing for a more efficacious and less

dangerous intervention.

CONCLUSIONS AND FUTURE DIRECTIONS

The absence of a recovery in the CD4+ T cell count during

long-term, virologically suppressive HAART is an unquestion-

able source of anxiety to HIV-infected patients and their treat-

ing clinicians, given the long-term risks of disease progression

and death, underscoring the need for means of identifying early

predictive factors and treatment options. Furthermore, INRs

may be viewed as a merging point between clinical practice

and basic science: while HIV/AIDS clinicians investigate which

by guest on May 2, 2016

http://cid.oxfordjournals.org/D

ownloaded from

HIV/AIDS • CID 2009:48 (1 February) • 335

treatment most effectively sustains peripheral the CD4+ T cell

count, bench scientists, by investigating the immunovirological

pathways behind insufficient recovery of the CD4+ T cell count,

may provide tools both to forecast immunological failure and

to investigate new therapeutic strategies.

Unfortunately, even though several approaches have been

suggested and investigated, no consensus has been reached yet

on the most efficacious treatment for immunological nonre-

sponse. Thus, we are now facing a dichotomy, whereby the

ever-growing list of new-class antiretrovirals provides us with

the most potent weapons against HIV/AIDS, while we still lack

a full grasp on the most proper means of administering them

to INRs. However, even though such research has not yet pro-

vided a unique treatment protocol, research has revealed diverse

immunovirological and even genetic patterns behind impaired

CD4+ T cell count recovery, indicating the need for novel treat-

ment scenarios tailored to different INRs, possibly including

antiretrovirals and immunomodulants. Thus, with regard to

the frustrating lack of concrete therapeutic options, we strongly

advocate the continuous investigation of the correlates of the

failure of CD4+ T cell counts to recover during HAART to aid

HIV/AIDS clinicians who treat INRs today, to find the most

effective and timely therapeutic options in the future.

Acknowledgments

We thank Tiziana Formenti for providing invaluable proofreading andediting. We are particularly thankful to all the staff members of the Clinicof Infectious Diseases, “San Paolo” Hospital, University of Milan.

Financial support. Fondo Interno Ricerca Scientifica e Tecnologica2007–Universita degli Studi di Milano; Istituto Superiore di Sanita, “Na-tional research program on AIDS”; and Fondazione Romeo and EnricaInvernizzi.

Potential conflicts of interest. All authors: no conflicts.

References

1. Palella FJ Jr, Delaney KM, Moorman AC, et al. Declining morbidityand mortality among patients with advanced human immunodefi-ciency virus infection. HIV Outpatient Study Investigators. N Engl JMed 1998; 338:853–60.

2. Sepkowitz KA. Effect of HAART on natural history of AIDS-relatedopportunistic disorders. Lancet 1998; 351:228–30.

3. Autran B, Carcelain G, Li TS, et al. Restoration of the immune systemwith anti-retroviral therapy. Immunol Lett 1999; 66:207–11.

4. Piketty C, Castiel P, Belec L, et al. Discrepant responses to triplecombination antiretroviral therapy in advanced HIV disease. AIDS1998; 12:745–50.

5. Dronda F, Moreno S, Moreno A, Casado JL, Perez-Elias MJ, AntelaA. Long-term outcomes among antiretroviral-naive human immu-nodeficiency virus–infected patients with small increases in CD4+ cellcounts after successful virologic suppression. Clin Infect Dis 2002;35:1005–9.

6. Yeni PG, Hammer SM, Hirsch MS, et al. Treatment for adult HIVinfection: 2004 recommendations of the International AIDS Soci-ety–USA Panel. JAMA 2004; 292:251–65.

7. Kaufmann GR, Furrer H, Ledergerber B, et al. Characteristics, de-terminants, and clinical relevance of CD4 T cell recovery to !500cells/mL in HIV type 1–infected individuals receiving potent antiret-roviral therapy. Clin Infect Dis 2005; 41:361–72.

8. d’Arminio Monforte A, Testori V, Adorni F, et al. CD4 cell counts at

the third month of HAART may predict clinical failure. AIDS 1999;13:1669–76.

9. Grabar S, Le Moing V, Goujard C, et al. Clinical outcome of patientswith HIV-1 infection according to immunologic and virologic re-sponse after 6 months of highly active antiretroviral therapy. AnnIntern Med 2000; 133:401–10.

10. Moore DM, Hogg RS, Yip B, et al. Discordant immunologic andvirologic responses to highly active antiretroviral therapy are associ-ated with increased mortality and poor adherence to therapy. J AcquirImmune Defic Syndr 2005; 40:288–93.

11. Nicastri E, Chiesi A, Angeletti C, et al. Clinical outcome after 4 yearsfollow-up of HIV-seropositive subjects with incomplete virologic orimmunologic response to HAART. J Med Virol 2005; 76:153–60.

12. Gutierrez F, Padilla S, Masia M, et al. Patients’ characteristics andclinical implications of suboptimal CD4 T-cell gains after 1 year ofsuccessful antiretroviral therapy. Curr HIV Res 2008; 6:100–7.

13. Tan R, Westfall AO, Willig JH, et al. Clinical outcome of HIV-infectedantiretroviral-naive patients with discordant immunologic and viro-logic responses to highly active antiretroviral therapy. J Acquir Im-mune Defic Syndr 2008; 47:553–8.

14. Panel on Antiretroviral Guidelines for Adults and Adolescents. Guide-lines for the use of antiretroviral agents in HIV-1 infected adults andadolescents. Washington, DC: Department of Health and HumanServices, 2008.

15. Lau B, Gange SJ, Moore RD. Risk of non-AIDS-related mortality mayexceed risk of AIDS-related mortality among individuals enrollinginto care with CD4+ counts greater than 200 cells/mm3. J AcquirImmune Defic Syndr 2007; 44:179–87.

16. Kaufmann GR, Bloch M, Finlayson R, Zaunders J, Smith D, CooperDA. The extent of HIV-1-related immunodeficiency and age predictthe long-term CD4 T lymphocyte response to potent antiretroviraltherapy. AIDS 2002; 16:359–67.

17. Florence E, Lundgren J, Dreezen C, et al. Factors associated with areduced CD4 lymphocyte count response to HAART despite full viralsuppression in the EuroSIDA study. HIV Med 2003; 4:255–62.

18. Viard JP, Mocroft A, Chiesi A, et al. Influence of age on CD4 cellrecovery in human immunodeficiency virus–infected patients receiv-ing highly active antiretroviral therapy: evidence from the EuroSIDAstudy. J Infect Dis 2001; 183:1290–4.

19. Lederman MM, McKinnis R, Kelleher D, et al. Cellular restorationin HIV infected persons treated with abacavir and a protease inhibitor:age inversely predicts naive CD4 cell count increase. AIDS 2000; 14:2635–42.

20. Collaboration of Observational HIV Epidemiological Research Europe(COHERE) Study Group. Response to combination antiretroviraltherapy: variation by age. AIDS 2008; 22:1463–73.

21. Phillips A. Short-term risk of AIDS according to current CD4 cellcount and viral load in antiretroviral drug-naive individuals and thosetreated in the monotherapy era. AIDS 2004; 18:51–8.

22. Miller MF, Haley C, Koziel MJ, Rowley CF. Impact of hepatitis Cvirus on immune restoration in HIV-infected patients who start highlyactive antiretroviral therapy: a meta-analysis. Clin Infect Dis 2005;41:713–20.

23. Laskus T, Radkowski M, Wang LF, Vargas H, Rakela J. The presenceof active hepatitis C virus replication in lymphoid tissue in patientscoinfected with human immunodeficiency virus type 1. J Infect Dis1998; 178:1189–92.

24. Laskus T, Radkowski M, Piasek A, et al. Hepatitis C virus in lymphoidcells of patients coinfected with human immunodeficiency virus type1: evidence of active replication in monocytes/macrophages and lym-phocytes. J Infect Dis 2000; 181:442–8.

25. Greub G, Ledergerber B, Battegay M, et al. Clinical progression, sur-vival, and immune recovery during antiretroviral therapy in patientswith HIV-1 and hepatitis C virus coinfection: the Swiss HIV CohortStudy. Lancet 2000; 356:1800–5.

26. De Luca A, Bugarini R, Lepri AC, et al. Coinfection with hepatitisviruses and outcome of initial antiretroviral regimens in previouslynaive HIV-infected subjects. Arch Intern Med 2002; 162:2125–32.

by guest on May 2, 2016

http://cid.oxfordjournals.org/D

ownloaded from

336 • CID 2009:48 (1 February) • HIV/AIDS

27. Chung RT, Evans SR, Yang Y, et al. Immune recovery is associatedwith persistent rise in hepatitis C virus RNA, infrequent liver testflares, and is not impaired by hepatitis C virus in co-infected subjects.AIDS 2002; 16:1915–23.

28. Sullivan PS, Hanson DL, Teshale EH, Wotring LL, Brooks JT. Effectof hepatitis C infection on progression of HIV disease and early re-sponse to initial antiretroviral therapy. AIDS 2006; 20:1171–9.

29. Wood E, Hogg RS, Yip B, Harrigan PR, O’Shaughnessy MV, MontanerJS. The impact of adherence on CD4 cell count responses amongHIV-infected patients. J Acquir Immune Defic Syndr 2004; 35:261–8.

30. Wood E, Montaner JS, Yip B, et al. Adherence to antiretroviral therapyand CD4 T-cell count responses among HIV-infected injection drugusers. Antivir Ther 2004; 9:229–35.

31. Moore RD, Keruly JC. CD4+ cell count 6 years after commencementof highly active antiretroviral therapy in persons with sustained vi-rologic suppression. Clin Infect Dis 2007; 44:441–6.

32. Egger M, May M, Chene G, et al. Prognosis of HIV-1-infected patientsstarting highly active antiretroviral therapy: a collaborative analysisof prospective studies. Lancet 2002; 360:119–29.

33. Ferraris L, Bellistri GM, Pegorer V, et al. Untangling the immuno-logical implications of nadir on CD4+ cell recovery during suppressivehighly active antiretroviral therapy. Clin Infect Dis 2008; 46:149–50.

34. Spritzler J, Mildvan D, Russo A, et al. Can immune markers predictsubsequent discordance between immunologic and virologic re-sponses to antiretroviral therapy? Adult AIDS Clinical Trials Group.Clin Infect Dis 2003; 37:551–8.

35. Benveniste O, Flahault A, Rollot F, et al. Mechanisms involved in thelow-level regeneration of CD4+ cells in HIV-1–infected patients re-ceiving highly active antiretroviral therapy who have prolonged un-detectable plasma viral loads. J Infect Dis 2005; 191:1670–9.

36. Moses A, Nelson J, Bagby GC Jr. The influence of human immu-nodeficiency virus-1 on hematopoiesis. Blood 1998; 91:1479–95.

37. Isgro A, Aiuti A, Mezzaroma I, et al. Improvement of interleukin 2production, clonogenic capability and restoration of stromal cell func-tion in human immunodeficiency virus-type-1 patients after highlyactive antiretroviral therapy. Br J Haematol 2002; 118:864–74.

38. Isgro A, Aiuti A, Leti W, et al. Immunodysregulation of HIV diseaseat bone marrow level. Autoimmun Rev 2005; 4:486–90.

39. Aiuti F, Mezzaroma I. Failure to reconstitute CD4+ T-cells despitesuppression of HIV replication under HAART. AIDS Rev 2006; 8:88–97.

40. Isgro A, Leti W, De Santis W, et al. Altered clonogenic capability andstromal cell function characterize bone marrow of HIV-infected sub-jects with low CD4+ T cell counts despite viral suppression duringHAART. Clin Infect Dis 2008; 46:1902–10.

41. Badolato R. Immunological nonresponse to highly active antiretroviraltherapy in HIV-infected subjects: is the bone marrow impairmentcausing CD4 lymphopenia? Clin Infect Dis 2008; 46:1911–2.

42. Kolte L, Dreves AM, Ersboll AK, et al. Association between largerthymic size and higher thymic output in human immunodeficiencyvirus–infected patients receiving highly active antiretroviral therapy.J Infect Dis 2002; 185:1578–85.

43. Franco JM, Rubio A, Martinez-Moya M, et al. T-cell repopulationand thymic volume in HIV-1-infected adult patients after highly activeantiretroviral therapy. Blood 2002; 99:3702–6.

44. Fry TJ, Mackall CL. What limits immune reconstitution in HIV in-fection? Divergent tools converge on thymic function. AIDS 2001;15:1881–2.

45. Douek DC, McFarland RD, Keiser PH, et al. Changes in thymicfunction with age and during the treatment of HIV infection. Nature1998; 396:690–5.

46. Teixeira L, Valdez H, McCune J, et al. Poor CD4 T cell restorationafter suppression of HIV-1 replication may reflect lower thymic func-tion. AIDS 2001; 15:1749–56.

47. Marziali M, De Santis W, Carello R, et al. T-cell homeostasis alterationin HIV-1 infected subjects with low CD4 T-cell count despite un-detectable virus load during HAART. AIDS 2006; 20:2033–41.

48. Marchetti G, Gori A, Casabianca A, et al. Comparative analysis of T-

cell turnover and homeostatic parameters in HIV-infected patientswith discordant immune-virological responses to HAART. AIDS2006; 20:1727–36.

49. Fry T, Connick E, Falloon J, et al. A potential role for interleukin-7in T-cell homeostasis. Blood 2001; 97:2983–90.

50. Napolitano L, Grant R, Deeks S, et al. Increased production of IL-7accompanies HIV-1-mediated T-cell depletion: implications for T-cellhomeostasis. Nature Medicine 2001; 7:73–9.

51. Fry TJ, Mackall CL. The many faces of IL-7: from lymphopoiesis toperipheral T cell maintenance. J Immunol 2005; 174:6571–6.

52. Valdez H, Connick E, Smith KY, et al. Limited immune restorationafter 3 years’ suppression of HIV-1 replication in patients with mod-erately advanced disease. AIDS 2002; 16:1859–66.

53. Hunt PW, Martin JN, Sinclair E, et al. T cell activation is associatedwith lower CD4+ T cell gains in human immunodeficiency vi-rus–infected patients with sustained viral suppression during anti-retroviral therapy. J Infect Dis 2003; 187:1534–43.

54. Pitrak DL, Bolanos J, Hershow R and Novak RM. Discordant CD4T lymphocyte responses to antiretroviral therapy for HIV infectionare associated with ex-vivo rates of apoptosis. AIDS 2001; 15:1317–9.

55. Ostrowski SR, Katzenstein TL, Thim PT, Pedersen BK, Gerstoft J,Ullum H. Low-level viremia and proviral DNA impede immune re-constitution in HIV-1–infected patients receiving highly active anti-retroviral therapy. J Infect Dis 2005; 191:348–57.

56. Chun T-W, Shawn Justement J, Pandya P, et al. Relationship betweenthe size of the human immunodeficiency virus type 1 (HIV-1) res-ervoir in peripheral blood CD4+ T cells and CD4+:CD8+ T cell ratiosin aviremic HIV-1–infected individuals receiving long-term highlyactive antiretroviral therapy. J Infect Dis 2002; 185:1672–6.

57. Brenchley JM, Price DA, Douek DC. HIV disease: fallout from amucosal catastrophe? Nat Immunol 2006; 7:235–9.

58. Douek D. HIV disease progression: immune activation, microbes, anda leaky gut. Top HIV Med 2007; 15:114–7.

59. Brenchley JM, Price DA, Schacker TW, et al. Microbial translocationis a cause of systemic immune activation in chronic HIV infection.Nat Med 2006; 12:1365–71.

60. Marchetti G, Bellistri GM, Borghi E, et al. Microbial translocation isassociated with sustained failure in CD4+ T-cell reconstitution in HIV-infected patients on long-term highly active antiretroviral therapy.AIDS 2008; 22:2035–8.

61. Guadalupe M, Sankaran S, George MD, et al. Viral suppression andimmune restoration in the gastrointestinal mucosa of human im-munodeficiency virus type 1-infected patients initiating therapy dur-ing primary or chronic infection. J Virol 2006; 80:8236–47.

62. Guadalupe M, Reay E, Sankaran S, et al. Severe CD4+ T-cell depletionin gut lymphoid tissue during primary human immunodeficiencyvirus type 1 infection and substantial delay in restoration followinghighly active antiretroviral therapy. J Virol 2003; 77:11708–17.

63. Estes J, Baker JV, Brenchley JM, et al. Collagen deposition limitsimmune reconstitution in the gut. J Infect Dis 2008; 198:456–64.

64. Paiardini M, Frank I, Pandrea I, Apetrei C, Silvestri G. Mucosal im-mune dysfunction in AIDS pathogenesis. AIDS Rev 2008; 10:36–46.

65. Read SW, Sereti I. HIV infection and the gut: scarred for life? J InfectDis 2008; 198:453–5.

66. Sakaguchi S. Naturally arising Foxp3-expressing CD25+CD4+ regu-latory T cells in immunological tolerance to self and non-self. NatImmunol 2005; 6:345–52.

67. Haas DW, Geraghty DE, Andersen J, et al. Immunogenetics of CD4lymphocyte count recovery during antiretroviral therapy: an AIDSClinical Trials Group study. J Infect Dis 2006; 194:1098–107.

68. O’Brien TR, McDermott DH, Ioannidis JP, et al. Effect of chemokinereceptor gene polymorphisms on the response to potent antiretroviraltherapy. AIDS 2000; 14:821–6.

69. Puissant B, Roubinet F, Massip P, et al. Analysis of CCR5, CCR2,CX3CR1, and SDF1 polymorphisms in HIV-positive treated patients:impact on response to HAART and on peripheral T lymphocytecounts. AIDS Res Hum Retroviruses 2006; 22:153–62.

70. Rauch A, Nolan D, Furrer H, et al. HLA-Bw4 homozygosity is as-

by guest on May 2, 2016

http://cid.oxfordjournals.org/D

ownloaded from

HIV/AIDS • CID 2009:48 (1 February) • 337

sociated with an impaired CD4 T cell recovery after initiation ofantiretroviral therapy. Clin Infect Dis 2008; 46:1921–5.

71. Arno A, Ruiz L, Juan M, et al. Efficacy of low-dose subcutaneousinterleukin-2 to treat advanced human immunodeficiency virus type1 in persons with �250/mL CD4 T cells and undetectable plasma virusload. J Infect Dis 1999; 180:56–60.

72. Marchetti G, Meroni L, Varchetta S, et al. Low-dose prolonged in-termittent interleukin-2 adjuvant therapy: results of a randomizedtrial among human immunodeficiency virus–positive patients withadvanced immune impairment. J Infect Dis 2002; 186:606–16.

73. Katlama C, Carcelain G, Duvivier C, et al. Interleukin-2 acceleratesCD4 cell reconstitution in HIV-infected patients with severe immu-nosuppression despite highly active antiretroviral therapy: the ILSTIMstudy–ANRS 082. AIDS 2002; 16:2027–34.

74. Levy Y, Durier C, Krzysiek R, et al. Effects of interleukin-2 therapycombined with highly active antiretroviral therapy on immune res-toration in HIV-1 infection: a randomized controlled trial. AIDS2003; 17:343–51.

75. Sullivan A, Hardy G, Nelson M, Gotch F, Gazzard B, Imami N. In-terleukin-2 associated viral breakthroughs induce HIV-1-specific CD4T cell responses in patients on fully suppressive highly active anti-retroviral therapy. AIDS 2003; 17:628–9.

76. Blattman J, Grayson J, Wherry E, Kaech S, Smith K, Ahmed R. Ther-apeutic use of IL-2 to enhance antiviral T-cell responses in vivo.Nature Medicine 2003; 9:540–7.

77. Marchetti G, Franzetti F, Gori A. Partial immune reconstitution fol-lowing highly active antiretroviral therapy: can adjuvant interleukin-2 fill the gap? J Antimicrob Chemother 2005; 55:401–9.

78. Farel CE, Chaitt DG, Hahn BK, et al. Induction and maintenancetherapy with intermittent interleukin-2 in HIV-1 infection. Blood2004; 103:3282–6.

79. Sereti I, Aga E, Spritzler J, et al.; ACTG 5214 team. rhIL7 in HIV-1-infected subjects with CD4 T-cell count 1100 cells/mL and viral load!50,000 copies/mL: results from a randomized, placebo-controlled,double-blinded study (ACTG5214) [abstract 128]. In: Program andabstracts of the 14th Conference on Retroviruses and OpportunisticInfections (Los Angeles). Alexandria, VA: Foundation for Retrovi-rology and Human Health, 2007:112.

80. Levy Y, Weiss L, Viard JP, et al. Repeated r-hIL-7 doses improve T-cell recovery in HIV-1-infected patients enrolled in a phase i/ii mul-ticentric study [abstract 127]. In: Program and abstracts of the 14thConference on Retroviruses and Opportunistic Infections (Los An-geles). Alexandria, VA: Foundation for Retrovirology and HumanHealth, 2007:112.

81. Levy Y, Weiss L, Goujard C, et al. Sustained immunological efficacyof repeated r-hIL-7 doses in HIV-1-infected patients: long-term fol-low-up of a phase i/ii multicentric study [abstract 708]. In: Programand abstracts of the 15th Conference on Retroviruses and Oppor-tunistic Infections (Boston). Alexandria, VA: Foundation for Retro-virology and Human Health, 2008:325.

82. Chougnet CA, Shearer GM. Regulatory T cells (Treg) and HIV/AIDS:summary of the September 7–8, 2006 workshop. AIDS Res HumRetroviruses 2007; 23:945–52.

83. Sereti I, Imamichi H, Natarajan V, et al. In vivo expansion ofCD4CD45RO-CD25 T cells expressing foxP3 in IL-2-treated HIV-infected patients. J Clin Invest 2005; 115:1839–47.

84. Rosenberg SA, Sportes C, Ahmadzadeh M, et al. IL-7 administrationto humans leads to expansion of CD8+ and CD4+ cells but a relativedecrease of CD4+ T-regulatory cells. J Immunother 2006; 29:313–9.

85. MacArthur RD, Novak RM, Peng G, et al. A comparison of threehighly active antiretroviral treatment strategies consisting of non-nu-cleoside reverse transcriptase inhibitors, protease inhibitors, or bothin the presence of nucleoside reverse transcriptase inhibitors as initialtherapy (CPCRA 058 FIRST Study): a long-term randomised trial.Lancet 2006; 368:2125–35.

86. Riddler SA, Haubrich R, DiRienzo AG, et al. Class-sparing regimensfor initial treatment of HIV-1 infection. N Engl J Med 2008; 358:2095–106.

87. Caron M, Auclairt M, Vissian A, Vigouroux C, Capeau J. Contributionof mitochondrial dysfunction and oxidative stress to cellular pre-mature senescence induced by antiretroviral thymidine analogues.Antivir Ther 2008; 13:27–38.

88. Garcia F, Vidal C, Plana M, et al. Residual low-level viral replicationcould explain discrepancies between viral load and CD4+ cell responsein human immunodeficiency virus–infected patients receiving anti-retroviral therapy. Clin Infect Dis 2000; 30:392–4.

89. Giorgi JV, Majchrowicz MA, Johnson TD, Hultin P, Matud J, DetelsR. Immunologic effects of combined protease inhibitor and reversetranscriptase inhibitor therapy in previously treated chronic HIV-1infection. AIDS 1998; 12:1833–44.

90. Hejdeman B, Lenkei R, Leandersson AC, et al. Clinical and immu-nological benefits from highly active antiretroviral therapy in spite oflimited viral load reduction in HIV type 1 infection. AIDS Res HumRetroviruses 2001; 17:277–86.

91. Smith D, Zaunders J, Kauffman G, et al. Greater suppression of CD8activation with 4 vs. 3 drugs in early stages of primary HIV infection(PHI) despite similar virological decay rates [abstract 61]. In: Programand abstracts of the 1st International AIDS Society Conference onHIV Pathogenesis and Treatment (Buenos Aires, Argentina) Geneva:International AIDS Society, 2001:101.

92. Havlir DV, Strain MC, Clerici M, et al. Productive infection maintainsa dynamic steady state of residual viremia in human immunodefi-ciency virus type 1-infected persons treated with suppressive antiret-roviral therapy for five years. J Virol 2003; 77:11212–9.

93. Bourgarit A, Lascoux C, Palmer P, et al. First-line use of enfuvirtide-containing HAART regimen with dramatic clinical and immunolog-ical improvement in three cases. AIDS 2006; 20:471–3.

94. Soria A, Cavarelli M, Sala S, et al. Unexpected dramatic increase inCD4+ cell count in a patient with AIDS after enfuvirtide treatmentdespite persistent viremia and resistance mutations. J Med Virol2008; 80:937–41.

95. Watson C, Jenkinson S, Kazmierski W, Kenakin T. The CCR5 recep-tor-based mechanism of action of 873140, a potent allosteric non-competitive HIV entry inhibitor. Mol Pharmacol 2005; 67:1268–82.

96. Wilkin T, Ribaudo H, Gulick R. The relationship of CCR5 inhibitorsto CD4 cell count changes: a meta-analysis of recent clinical trials intreatment-experienced subjects [abstract 800]. In: Program and ab-stracts of the 15th Conference on Retroviruses and OpportunisticInfections (Boston). Alexandria, VA: Foundation for Retrovirologyand Human Health, 2008:362.

97. Hughes R, Sabin C, Sterne JAC,. Long-term trends in CD4 counts inpatients starting HAART: UK CHIC study [abstract P18.4/04/BPD].In: Program and abstracts of the 11th European AIDS Conference(Madrid). European AIDS Clinical Society, 2007:137.

98. Coull JJ, Turner D, Melby T, Betts MR, Lanier R, Margolis DM. Apilot study of the use of mycophenolate mofetil as a component oftherapy for multidrug-resistant HIV-1 infection. J Acquir ImmuneDefic Syndr 2001; 26:423–34.

99. Rizzardi GP, Harari A, Capiluppi B, et al. Treatment of primary HIV-1 infection with cyclosporin A coupled with highly active antiretroviraltherapy. J Clin Invest 2002; 109:681–8.

100. Lederman MM, Smeaton L, Smith KY, et al. Cyclosporin A providesno sustained immunologic benefit to persons with chronic HIV-1infection starting suppressive antiretroviral therapy: results of a ran-domized, controlled trial of the AIDS Clinical Trials Group A5138. JInfect Dis 2006; 194:1677–85.

101. Wallis RS, Kyambadde P, Johnson JL, et al. A study of the safety,immunology, virology, and microbiology of adjunctive etanercept inHIV-1-associated tuberculosis. Aids 2004; 18:257–64.

102. Gori A, Rossi M, Trabattoni D, et al. Tumor necrosis factor–a in-creased production during thalidomide treatment in patients withtuberculosis and HIV coinfection. J Infect Dis 2000; 182:639–40.

103. De Maria A. Discordant responses to HAART in HIV-1 patients: theneed to focus on intervention. Expert Rev Anti Infect Ther 2007; 5:523–7.

by guest on May 2, 2016

http://cid.oxfordjournals.org/D

ownloaded from