ArticlePDF AvailableLiterature Review

Abstract and Figures

Population aging has become a global phenomenon, and HIV infection among older individuals is also increasing. Because age can affect the progression of HIV infection, we aimed to evaluate the present knowledge on HIV infection in older patients. Literature review of the last 20 years. Older HIV-infected patients have lower CD4(+) T cell counts, higher viral load and are more frequently symptomatic at diagnosis. The infection progresses more rapidly, with higher morbidity and lethality rates. However, older patients are more compliant to antiretroviral treatment; they experience a better virologic response, and treatment represents a positive clinical impact. Aging affects the complex interaction between HIV infection and the immune system. Both conditions contribute to the dysfunction of immune cells, including a decrease in the phagocytes' microbicidal capability, natural killer cells' cytolytic function, expression of toll-like receptors and production of interleukin-12. Chronic immune activation responsible for the depletion of CD4(+) and CD8(+) T cells in HIV infection appears to worsen with senescence. Older patients also exhibit a less robust humoral response, with the production of less avid and specific antibodies. Both HIV and aging contribute to immune dysfunction, morbidity and mortality. However, highly active antiretroviral therapy (HAART) is beneficial for older patients, and treatment of older patients should not be discouraged.
Content may be subject to copyright.
This article appeared in a journal published by Elsevier. The attached
copy is furnished to the author for internal non-commercial research
and education use, including for instruction at the authors institution
and sharing with colleagues.
Other uses, including reproduction and distribution, or selling or
licensing copies, or posting to personal, institutional or third party
websites are prohibited.
In most cases authors are permitted to post their version of the
article (e.g. in Word or Tex form) to their personal website or
institutional repository. Authors requiring further information
regarding Elsevier’s archiving and manuscript policies are
encouraged to visit:
http://www.elsevier.com/copyright
Author's personal copy
Ageing Research Reviews 10 (2011) 163–172
Contents lists available at ScienceDirect
Ageing Research Reviews
journal homepage: www.elsevier.com/locate/arr
Review
Aging and HIV infection
Vivian Iida Avelino-Silvaa,b,, Yeh-Li Hoa,b, Thiago Junqueira Avelino-Silvac, Sigrid De Sousa Santosa,b
aInfectious and Parasitic Diseases Division, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo Avenida
Dr. Eneas de Carvalho Aguiar 255, 4Andar do Instituto Central, Sala 4028 05403-000–Sao Paulo, SP, Brazil
bInfectious and Parasitic Diseases Department, Faculdade de Medicina da Universidade de Sao Paulo Avenida
Dr. Eneas de Carvalho Aguiar 470, 2Andar do Instituto de Medicina Tropical I 05403-000–Sao Paulo, SP, Brazil
cGeriatrics Division, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo Avenida
Dr. Eneas de Carvalho Aguiar 255, 8Andar do Predio dos Ambulatorios, Bloco 8 05403-000–Sao Paulo, SP, Brazil
article info
Article history:
Received 8 June 2010
Received in revised form 12 October 2010
Accepted 15 October 2010
Keywords:
Aging
Immunosenescence
HIV
abstract
Introduction: Population aging has become a global phenomenon, and HIV infection among older indi-
viduals is also increasing. Because age can affect the progression of HIV infection, we aimed to evaluate
the present knowledge on HIV infection in older patients.
Methods: Literature review of the last 20 years.
Results: Older HIV-infected patients have lower CD4+T cell counts, higher viral load and are more fre-
quently symptomatic at diagnosis. The infection progresses more rapidly, with higher morbidity and
lethality rates. However, older patients are more compliant to antiretroviral treatment; they experience
a better virologic response, and treatment represents a positive clinical impact. Aging affects the complex
interaction between HIV infection and the immune system. Both conditions contribute to the dysfunc-
tion of immune cells, including a decrease in the phagocytes’ microbicidal capability, natural killer cells’
cytolytic function, expression of toll-like receptors and production of interleukin-12. Chronic immune
activation responsible for the depletion of CD4+and CD8+T cells in HIV infection appears to worsen with
senescence. Older patients also exhibit a less robust humoral response, with the production of less avid
and specific antibodies.
Conclusion: Both HIV and aging contribute to immune dysfunction, morbidity and mortality. However,
highly active antiretroviral therapy (HAART) is beneficial for older patients, and treatment of older
patients should not be discouraged.
© 2010 Elsevier B.V. All rights reserved.
1. Introduction
Population aging has been observed in countries around the
world, including developing countries. On an individual level, aging
is associated with complex modifications of the immune system
that result in increased susceptibility to infectious, autoimmune,
and neoplastic diseases (Pawelec, 1999) in addition to a decreased
response to active immunization (Haynes, 2005).
In parallel, human immunodeficiency virus (HIV) infection has
been spreading among older individuals through the decades. In
Brazil there has been an absolute and relative increase in AIDS cases
in this population (Brasil, 1980), mainly due to heterosexual trans-
mission, and the aged population is greatly vulnerable to this mode
of transmission (Stall and Catania, 1994).
Corresponding author at: Rua Frei Caneca, 557, Cerqueira Cesar, São Paulo, zip
code 01307-001, SP, Brazil. Tel.: +55 11 3120 5290.
E-mail address: viviansilva87@gmail.com (V.I. Avelino-Silva).
The course of HIV infection can differ between older and younger
people. Thus, it is fundamental to study the specific characteristics
of HIV infection in older patients.
2. Methods
We performed a review of the literature from the last 20 years
using the electronic databases MEDLINE, LILACS, EMBASE, and
SCIELO. The search terms “aged,” “elderly,” “aging,” and “HIV”
were combined with the term “immunology” using the “AND”
Boolean operator. The search terms “aged,” “elderly,” and “aging”
were combined with the term “HIV” using the “AND” Boolean
operator. We excluded case reports and articles related to the
diagnosis or treatment of opportunistic infections or comorbidi-
ties. In addition, articles and other data sources were found from
bibliographic references and from informal channels of com-
munication, such as personal correspondences and unpublished
events.
1568-1637/$ see front matter © 2010 Elsevier B.V. All rights reserved.
doi:10.1016/j.arr.2010.10.004
Author's personal copy
164 V.I. Avelino-Silva et al. / Ageing Research Reviews 10 (2011) 163–172
3. Results
3.1. Definitions
While aging is a continuous and dynamic process, World Health
Organization and most clinical and geriatric references define the
chronological age of 60 years as a cutoff of “elderly” or older popu-
lation (World Health Organization, 2010). However, the Centers for
Disease Control and Prevention defines older HIV people as those
ages 50 and over, originally because 50 years was considered the
upper age range for HIV infection (Linsk, 2000). Accordingly, most
articles referred in the text follow the 50 years cutoff for HIV older
patients and 60 years cutoff for general geriatric population.
3.2. Clinical aspects
Aside from any of the effects of HIV infection, aging is also asso-
ciated with an increased susceptibility to infectious complications
such as pulmonary and urinary tract infections (Gavazzi and Krause,
2002), which occur at increased frequency and clinical severity, and
are more often associated with comorbidities (Kovaiou et al., 2007).
Age-related degenerative processes and comorbidities affect pri-
mary defense barriers such as dermal integrity, coughing reflexes,
and mucociliary clearance (Castle, 2000).
The clinical presentation of infections also differs between older
and younger people. Older patients are usually less symptomatic
and may have atypical symptoms such as anorexia, confusion,
asthenia or fatigue. Fever may be very low or even nonexistent in
up to 50% of older patients with documented infections (Gavazzi
and Krause, 2002).
Older people are at higher risk for hospitalization and death
due to influenza infections. Although they offer protective benefits,
influenza vaccinations are less effective in the elderly compared
to younger adults (Prevention, 1999; Webster, 2000). Aging has
also been observed to be associated with a lower response to the
pneumococcal vaccine (Musher et al., 1986), a lower reactivity to
intradermal tests (Carvalho, 1970), and a greater susceptibility to
neoplastic diseases (Gavazzi and Krause, 2002).
Older HIV-infected patients present with lower CD4+T cell
counts (Phillips et al., 1991; Operskalski et al., 1995), higher viral
load and are more frequently symptomatic at diagnosis (O’Brien
et al., 1996; Ferro and Salit, 1992; Ena et al., 1998). The diagno-
sis of HIV infection is frequently late in patients over 50 years
old. The vulnerability of this group is often neglected by health
professionals who assume that older patients are neither sexually
active nor users of intravenous drugs. In addition, symptoms such
as weight loss, fatigue, and visual and cognitive impairments are
repeatedly assumed to be part of the normal aging process (Sanders
et al., 2008). Older patients themselves are less likely to suspect
HIV infection. Up to 81.3% of HIV-infected patients over the age of
60 never suspected they had an infection prior to testing (Castro,
2007; Babiker et al., 2001). Furthermore, the course of infection is
more rapidly progressive, with higher morbidity and lethality rates
probably as result of immunosenescence, comorbidities, and later
diagnosis (Ena et al., 1998; Soriano et al., 1998; Carre et al., 1994;
Skiest et al., 1996; Butt et al., 2001).
3.3. Morphofunctional changes of immune organs
Aging and HIV infection both contribute to peripheral blood
cytopenia of one or more lineages. The bone marrow hematopoietic
compartment becomes gradually smaller with age, and it is substi-
tuted by adipose tissue (Compston, 2002). This structural change
is probably associated with hormonal modifications, such as the
decline in human growth hormone production (Lamberts et al.,
1997; French et al., 2002). In addition, HIV infects bone marrow
accessory cells, causing defective stromal function and alteration of
the hematopoietic growth factor network (Isgro et al., 2005; Alexaki
and Wigdahl, 2008); older HIV patients frequently present with
dysplasia affecting the myeloid, erythroid and platelet precursor
cells (Tripathi et al., 2005).
Old mice have an increased number of bone marrow
macrophages with impaired ability to generate or release tumor
necrose factor (TNF)(Wang et al., 1995). Among all the stromal
cells, the macrophage is the most important cell type that is produc-
tively infected by HIV-1, and macrophages express viral antigens
both in vivo and in vitro (Isgro et al., 2005).
Age diminishes interleukin-7 production by bone marrow cells
(Kang et al., 2004; Tsuboi et al., 2004). This cytokine is apparently
involved in thymocyte proliferation, differentiation, the matura-
tion of T and B lymphocytes and the restoration of peripheral T-cell
counts in HIV-infected patients (Beq et al., 2004).
The thymus is the primary organ for T-cell maturation, and
with age, the thymus undergoes an important volume reduction,
an increase in the proportion of epithelial tissue and perivascu-
lar component, and a decrease in new lymphocytes production
(Steinmann, 1986; Aw et al., 2007). Thymic atrophy is associated
with a shift from a stimulatory to a suppressive cytokine pattern
(Gruver et al., 2007) and can be reversed by growth hormone (GH)
replacement (French et al., 2002). In older HIV-infected individuals,
thymic involution results in a lower production of naïve cells and
can affect CD4+T cell reconstitution during antiretroviral therapy
(Douek et al., 1998; Casau, 2005); GH replacement can enlarge the
thymic volume and increase naïve CD4+T cell counts (Napolitano
et al., 2002).
As suggested by Effros et al. (2008), age-related changes in
gut-associated lymphoid tissue (GALT) are likely to occur, since
gastrointestinal immunity against some intestinal pathogens is
reduced in the elderly. GALT is also profoundly affected by HIV
infection. HIV replicates most intensely in GALT, with rapid and
massive depletion of lamina propria CD4+T cells during acute
infection. Immune reconstitution of the gastrointestinal tract after
HAART is poor and occurs at a much lower rate than in the periph-
eral blood (Brenchley and Douek, 2008).
3.4. Innate immunity
Macrophages and neutrophils appear to lose their microbici-
dal capability with age. Their weakened respiratory burst results in
decreased production of reactive nitrogen and oxygen intermedi-
ates (Plackett et al., 2004; Gomez et al., 2005) as well as impaired
phagocytic function (Butcher et al., 2001).
Similarly, HIV-1-infected patients have a significant decrease in
macrophage phagocytic and oxidative capability, apparently medi-
ated by the Nef protein and related to CD4+T cells depression (Torre
et al., 2002; Noursadeghi et al., 2006). HIV glycoproteins gp120 and
gp41 also have inhibitory activity on monocyte chemotaxis and
activation by chemokines (Noursadeghi et al., 2006). An expected
consequence of the interaction of older age and HIV infection would
be an additional degeneration in macrophagic function. In addition,
macrophages exert an important role in HIV mucosal infection as
they act as a cellular reservoir for viruses and enable HIV penetra-
tion and persistence in the central nervous system through a Trojan
horse mechanism (Crowe et al., 2003; Orenstein, 2001). It is con-
ceivable that aged mucosa facilitates HIV infection due in part to a
macrophagic dysfunction.
In older patients, bacterial products, cytokines and inflamma-
tory mediators such as lipopolysaccharide, interleukin 2 (IL-2),
granulocyte macrophage colony-stimulating factor (GM-CSF) and
granulocyte colony-stimulating factor (G-CSF) are less able to pre-
vent apoptosis of aged neutrophils following stimulation. In the
young, inflammatory mediators are able to prevent apoptosis. This
Author's personal copy
V.I. Avelino-Silva et al. / Ageing Research Reviews 10 (2011) 163–172 165
response would guarantee a continued involvement of neutrophils
in controlling microbes’ proliferation, while other cells traffic to the
site of injury. Bacterial products, cytokines and inflammatory medi-
ators as lipopolysaccharide (LPS), interleukin-2, GM-CSF, and G-CSF
are less able to prevent apoptosis of aged neutrophils following
stimulation (Plackett et al., 2004).
Aging is associated with a decrease in toll-like receptors expres-
sion and function in monocytes and macrophages; in particular,
these cells have decreased expression of TLR1 and TLR4 and conse-
quently decreased production of TNF-and interleukin 6 (IL-6).
This defect in cytokine expression may be responsible for the
absence of the systemic signals of infection in older patients (Panda
et al., 2009; Renshaw et al., 2002). Experimental evidence has sug-
gested that HIV-1 infection also reduces monocyte expression of
TLR4 (Noursadeghi et al., 2006), thus increasing risk of delayed
diagnosis of infectious illnesses due to lack of specific symptoms.
The production of prostaglandin E2 (PGE2) is enhanced in aged
macrophages (Wu et al., 2001); PGE2 induces interleukin-10 (IL-
10) production, inhibits major histocompatibility complex class II
(MHC-II) expression by dendritic cells, and up-regulates T-helper
cell type 2 (TH2) cytokines (Plackett et al., 2004). Despite the associ-
ation between HIV, cytokine profile, and disease progression is still
controversial, researchers have observed an over-representation
of TH2 responses against viral capsid proteins in subjects with
chronic-progressive HIV-1 infection (Chevalier et al., 2009). Pos-
sible consequences of TH2 cytokines up-regulation include an
exacerbation in allergic reactions and an antagonistic effect on TH1
responses, reducing microbicidal activity of phagocytes and elimi-
nation intracellular microbes.
Dendritic cells (DC) play a critical role in HIV mucosal transmis-
sion and dissemination (Wu and KewalRamani, 2006), and recently
they have been implicated as HIV-1 long-term reservoirs (Coleman
and Wu, 2009). Simultaneously, however, DC are potent stimula-
tors of immune responses and have been used in prophylactic and
therapeutic approaches for HIV infection (Lu et al., 2004). Aging
is associated with a reduced capacity of these cells to phagocy-
tose antigens, produce interleukin-12, and migrate in response
to chemokines (Agrawal et al., 2007). Furthermore, older patients
experience a progressive decline of follicular myeloid DCs (Della
Bella et al., 2007). In older HIV patients, several paradoxical impli-
cations could derive from this decline in dendritic cells function
with age, including a less effective HIV transmission, dissemination
and long-term maintenance, as well as a decrease in the control of
HIV infection.
Natural killer (NK) cells are innate cytotoxic lymphocytes that
do not express T-cell antigen receptors. They play an important
role in the defense against malignancies and viral infections. NK
cells contribute to resistance of certain individuals to HIV infec-
tion, and to immune response in the chronic infection (Fauci et al.,
2005). Although the number of NK cells increases with age, they
are more likely to have a mature phenotype (CD56dim), a dimin-
ished lytic efficiency, and impaired production of cytokines (IFN-,
IL-2) and chemokines (MIP-1a, RANTES, and interleukin 8) (Panda
et al., 2009; Renshaw et al., 2002). HIV infection further affects
the NK cell compartment and induces a decreased proportion of
immature:mature subsets (CD56dim/CD56bright ), lower expression
of natural cytotoxicity receptors and reduced cytolytic potential
(Mantegani et al., 2009). Therefore, elderly individuals could be
more susceptible to HIV induced NK cell dysfunctions.
3.5. Cell-mediated adaptive immunity
In healthy individuals the T cell population shows exceptional
homeostatic control regarding numbers and proportions of the two
major functional subsets of T lymphocytes CD8+T cells, whose
principal function is to kill virus-infected cells, and CD4+T cells,
which are critical for activating other immune cells (Vrisekoop
et al., 2009).
In HIV-infected individuals, the depletion and dysfunction of
CD4+T cells are responsible for the majority of AIDS complications.
CD4+T cell loss results from a complex interplay between the virus
and the immune system, and it seems to be a biphasic phenomenon,
with an initial massive depletion of CD4+T cells in mucosal tis-
sues during acute HIV infection (Veazey et al., 1998; Kewenig et al.,
1999), followed by a slow decline of the remaining CD4+T cells
during chronic infection. Direct viral cytopathicity alone cannot
explain the course of the disease, particularly during the chronic
phase of infection, since only 0.01–1% of CD4+T cells are infected
in both the peripheral blood and lymph nodes (Chun et al., 1997;
Haase, 1999; Haase et al., 1996; Anderson et al., 1998; Douek et al.,
2002). Another reasonable explanation for CD4+T cells depletion is
that HIV is likely to either directly or indirectly induce a chronic
immune activation that disrupts T cell homeostasis (Vrisekoop
et al., 2009; Douek et al., 2003). Indeed, the degree of chronic acti-
vation is a good predictor of disease progression (Simmonds et al.,
1991; Leng et al., 2001; Roussanov et al., 2000; Giorgi et al., 1999).
Two causal models for this immune activation have been pro-
posed. The first is a homeostatic T cell imbalance due to a chronic
activation of innate immunity through plasmacytoid dendritic
cells activation (Mandl et al., 2008; Meier et al., 2009) and to a
damage in the mucosal surfaces resulting in translocation of pro-
inflammatory microbial products from the intestinal lumen into the
circulation (Brenchley et al., 2006). Alternatively, the second model
purposes a chronic immune activation due to CD4+T cells deple-
tion, either a general decline in CD4+T cells number or a decline
in particular subsets of effector CD4+T cells or T regulatory lym-
phocytes (TREG) (Brenchley et al., 2008; Fazekas de St Groth and
Landay, 2008). Immune activation induced by HIV infection gen-
erates new available target cells for viral replication, resulting in
a positive feedback loop with further CD4+T cells destruction and
immune activation (Douek et al., 2003; Grossman et al., 2002).
Age also induces changes in the number, proportion and func-
tion of lymphocytes. Naïve lymphocytes of elderly mice have a
lower capacity of activation, cytokine production and differenti-
ation to Th1 and Th2 subpopulations (Aw et al., 2007; Haynes et al.,
2003; Haynes et al., 2005; Haynes and Maue, 2009). The aging
process is associated with a reduction in the naïve lymphocyte pop-
ulation, lower expression of CD27 and CD28 and a decrease in T-cell
receptor (TCR) diversity (Haynes and Maue, 2009; Pawelec et al.,
2009; Derhovanessian et al., 2009). Naïve lymphocyte survival is
influenced by cytokines such as interleukin 7 (IL-7). Accordingly,
the elderly have decreased serum IL-7 levels (Kang et al., 2004),
but among centenarians, who represent a living model of survival
accomplishment, plasma levels of IL-7 are relatively high (Nasi
et al., 2006), suggesting an influence on longevity.
Thymic involution with aging seems to result in the inability
to replace CD4+T cells depleted by HIV infection (Casau, 2005;
Douek et al., 1998, 2003). HIV infection is also associated with a
decrease in the replicative ability of T lymphocyte precursor cells
(Effros et al., 1996). The continuous recruitment and turnover of
naïve T cells, required to maintain the memory pool, eventually
becomes unsustainable (Hazenberg et al., 2000) leading to exhaus-
tion of some T-lymphocytes clones. Moreover, telomeric length in
CD4+and CD8+T cells has been shown to be significantly shorter
in HIV-infected patients (Bestilny et al., 2000).
The number of memory lymphocytes increase with age, but
these cells exhibit a decreased capacity for activation, signaling
and proliferation in aged mouse models (Miller et al., 1997; Nel
and Slaughter, 2002). Memory cells have a diminished ability to
respond to primary antigen when challenges occur at advanced
ages (Haynes et al., 2003; Haynes et al., 2005; Miller et al., 1997;
Kapasi et al., 2002). Several steps in TCR signal transduction are
Author's personal copy
166 V.I. Avelino-Silva et al. / Ageing Research Reviews 10 (2011) 163–172
declined, possibly due to changes in lipid raft composition in the
surface membrane of senescent T cells (nel). CD4 T cells from young
(2- to 4-month-old) and aged (14- to 16-month-old) mice have
been tested regarding proliferation and effector cytokine produc-
tion, and sticking differences have been demonstrated both ex vivo
and in vivo, with decreased proliferation in response to antigen
stimulation, in IL-2 production by Th1 and IL-4 and IL-5 production
by Th2 memory cells, and in cognate helper function.
HIV infection decreases the CD8+T cells population by apoptosis
or direct cytotoxicity. In addition, HIV perturbs CD8+T cell cyto-
toxic function by impairing perforin production and expression of
Fas-ligand (FasL). The interaction between FasL and Fas molecules
induces the apoptosis of infected target cells (Saha et al., 2001;
Piazza et al., 2002).
Aging results in a progressive decrease in CD28 expression
on CD8+T cells. CD28 is a co-receptor essential to elicit strong
responses to antigenic challenge; CD28 signal transduction acti-
vates IL-2 gene transcription, enhances IL-2 receptor expression,
modulates T cell migration and homing, and, notably, enhances
telomerase activity (Effros, 2004). Consequently, CD28-negative T
lymphocytes produce lower levels of IL-2 and IL-2r, have shorter
telomeres and thus, lower activating, signaling and proliferative
potential (Effros and Pawelec, 1997; Plunkett et al., 2007).
Progressive loss of CD28 expression on CD8+T cells with aging
is apparently related to cytomegalovirus (CMV) infection (Pawelec
et al., 2009; Hadrup et al., 2006; Pawelec et al., 2004; Akbar and
Fletcher, 2005). As a permanent infection, CMV requires a persis-
tent immune control which would lead to a marked expansion of
CD8+CD28CMV-specific cells (Pawelec et al., 2004). Accordingly,
CMV-seropositivity has been included in a cluster of age-related
changes in immune parameters, named “immune risk phenotype”
(IRP), which was found to predict mortality in a Swedish longitudi-
nal study (Wikby et al., 1994). Other immune parameters described
in the IRP are poor T-cell proliferative responses to mitogens, low
numbers of B cells and inverted CD4:CD8 ratio (Hadrup et al.,
2006; Wikby et al., 1998). The clonal expansion of CD8+CD28
T lymphocytes additionally decreases the response to vaccines
(Saurwein-Teissl et al., 2002).
It is possible that HIV infection induces an immunologic effect
similar to that of CMV infection through a chronic immune activa-
tion (Moanna et al., 2005). A shift in the CD4:CD8 ratio occurs in
advanced HIV infection due to the fall in CD4+T cell counts and the
severe decrease in CD28 expression (Effros, 2000). Thus, it could be
expected that HIV-infected elderly may experience an accentuation
of the deleterious effects already characteristic of the aged immune
risk profile. When higher proportions of CD8+CD28T lymphocytes
were detected at early stages of infection, disease progressed faster
(Cao et al., 2009a).
It is also reasonable to imagine that CMV infection and its
immunological consequences could negatively impact the immu-
nity to HIV by leading to a premature aging of T cells. However,
this association needs to be further investigated. Despite its poten-
tial harmful effects, the maintenance of an adequate immune
response against CMV is essential in preventing its reactivation
during advanced stages of AIDS.
Older patients have a higher concentration of cholesterol in
their cell membranes, which leads to a decrease in membrane flu-
idity and consequent impairment in lymphocyte capacity of lipid
raft internalization. This may weaken lymphocyte signaling and
further contribute to the inefficiency of acquired cellular immune
responses in the elderly (Larbi et al., 2006). Changes in cholesterol
metabolism are also very frequent in HIV-infected/AIDS patients,
especially those under HAART, and elevated cholesterol levels
in this population may be related to a lower cellular membrane
fluidity and lymphocyte signaling impairment (Dube, 2003). For
an aged HIV-infected individual, it is possible to hypothesize this
overlap membrane fluidity impairment could further decrease
lymphocyte signaling.
3.6. Humoral adaptive immunity
Older individuals experience both quantitative and qualitative
modifications in their humoral immune response (Weksler and
Szabo, 2000). Production of B lymphocytes in the bone marrow
is reduced in the elderly as a result of lower levels of precursor
cells and extrinsic factors, such as bone marrow stem cells and
IL-7 (Szabo et al., 1998). HIV infection seems to exert an opposite
effect as it promotes B-cell activation by increasing the produc-
tion of several cytokines and growth factors, such as interferon-,
tumor necrosis factor, interleukin-6, interleukin-10, CD40 ligand,
B-cell-activating factor, and IL-7 (Moir and Fauci, 2009).
Despite the decreased production of B lymphocytes by the bone
marrow, B cell number is strictly regulated and remains constant
with age. This is probably due to peripheral regeneration and
improved cell survival. Nevertheless, following drug-induced lym-
phopenia, aged mice regenerate a more restricted repertoire of B
lymphocyte than younger mice, probably due to a decreased bone
marrow production (Weksler and Szabo, 2000).
Peripheral B lymphocytes may be classified by their expression
of IgD and CD27 markers. The B cell subset that is IgDand CD27
is significantly increased among older patients. These cells do not
act as antigen presenting cells, nor do they express significant lev-
els of the CD40 molecule necessary to interact with T lymphocytes
(Caruso et al., 2009). HIV also drives the expansion of some human
B-cell subpopulations, such as immature transitional B cells, acti-
vated mature B cells, and exhausted B cells that are CD27(Moir
and Fauci, 2009).
Older mice and humans both have elevated production of
polyreactive antibodies that are less avid and specific (Weksler and
Szabo, 2000; Ben-Yehuda et al., 1998). The elderly population, espe-
cially those who suffer from chronic diseases, has increased levels
of autoantibodies, a mortality risk factor in this group (Weksler
and Szabo, 2000; Hallgren et al., 1973). Additionally, HIV infec-
tion is characterized by B-cell hyperactivation and poorly inducible
antibody responses. B-cell hyperactivation is manifested by hyper-
gammaglobulinemia, polyclonal B-cell activation, increased cell
turnover, expression of activation markers, and differentiation of
B cells to plasmablasts, with increased production of autoantibod-
ies and an increase in the frequency of B-cell malignancies (Moir
and Fauci, 2009).
A large number of studies have recently demonstrated that a
variety of vaccines are less efficient in elderly persons, includ-
ing influenza vaccine, pneumococcal polysaccharide vaccine and
hepatitis A vaccine. These impaired responses are correlated to a
decrease in several innate and adaptive immune mechanisms with
age, such as a reduced phagocytosis, processing and presentation
of antigens, a decline in naive T cell counts and an accumulation
of highly differentiated effector T cells with impaired stimulatory
and replicative capacities (Weinberger et al., 2008). All these age-
related impairments could lead to a decreased uptake of antigen at
the site of injection, diminished activation and stimulation of adap-
tive immune cells and reduced responses to neoantigens (Weinberg
et al., 2008).
HIV infection is also associated with a reduced efficacy of vac-
cines and lower titers of vaccine-elicited antibodies. One of the
abnormalities probably linked to poor vaccine response in HIV
infection is a defect in class switching, particularly pronounced
in patients with CD4+T cell counts of less than 200 cells/mm3
(Doria-Rose and Connors, 2009). Regardless of the immunologic
reconstitution after HAART, patients with lower pretreatment CD4+
T cell count nadir have a weaker response to vaccination (Lange
et al., 2003).
Author's personal copy
V.I. Avelino-Silva et al. / Ageing Research Reviews 10 (2011) 163–172 167
3.7. Immunosenescence and immune response imbalance
Autoreactive immature lymphocyte clones are usually produced
by the lymphoid organs, and one of the main immune tolerance
mechanisms resides in suppression by TREG. In particular, the TREG
subset characterized as CD4+CD25+Foxp3+cells has been shown to
exert immune modulation in experimental and observational mod-
els (Belkaid and Rouse, 2005; Dejaco et al., 2006; Rouse and Suvas,
2007). These cells seem to balance the immune response between
two poles, one overstimulated with the occurrence of autoimmune
diseases, and one suppressed pole with opportunistic recurrent
infections (Maloy et al., 2003; Sugimoto et al., 2003; Boyer et al.,
2004).
High TREG counts might contribute to immune response sup-
pression in the elderly, leading to a lower response to vaccines
and a higher susceptibility to infections and cancer (Miller et al.,
1997). Theoretically, an elevation in TREG lymphocyte counts
could also damage the CD4+T cell responses to HIV infection
in older patients (Miller et al., 1997). Moreover, a recent study
found TREG lymphocyte expansion in HIV-infected patients and
established its association with disease progression (Cao et al.,
2009b).
Immunosenescence implies much more than just a decline in
immune function and is actually an imbalance between suppres-
sion and exacerbation of a chronic inflammatory status (Licastro
et al., 2005). For instance, peripheral mononuclear cells from older
individuals stimulated in vitro produce more proinflammatory
cytokines than cells from younger people (Effros, 2004; Fagiolo
et al., 1993). This might also happen in vivo in older patients suffer-
ing from inflammatory injuries (Bruunsgaard et al., 2001).
Therefore, aging results in chronic immune activation associated
with several clinical conditions, such as bone reabsorption, frailty
and sarcopenia, metabolic syndrome, osteoarthritis, atheroscle-
rosis and cardiovascular diseases, Parkinson’s and Alzheimer’s
diseases, and cancer (Castle, 2000; Licastro et al., 2005). This state
of chronic immune activation is sometimes defined as “autotoxic-
ity” rather than autoimmunity in which there is a direct immune
aggression against “self” antigens (McGeer and McGeer, 2004).
Aside from immunosuppression with polyclonal B-cell acti-
vation, chronic immune activation is a characteristic feature
of progressive HIV disease; including increased T-cell turnover,
increased frequencies of activated T cells, and increased serum lev-
els of proinflammatory cytokines and chemokines (Douek et al.,
2003). This inflammatory state may be elicited by circulating
microbial products possibly derived from the gastrointestinal tract
(Brenchley et al., 2006), and it can be manifested by the exac-
erbation of inflammatory rheumatic and dermatologic conditions
(Cuellar and Espinoza, 2000; Dlova and Mosam, 2006).
Fig. 1 illustrates the main changes in the immune organs and
in innate, cell-mediated, and humoral immunity expected in older
HIV-infected patients.
3.8. Treatment response in HIV-infected older patients
Several authors have studied the immunologic and virologic
response to antiretroviral therapy in older HIV-infected patients
and found conflicting results (Table 1).
Some studies have established that older patients who undergo
HAART present a lower elevation in CD4+T cell counts and a
decreased production of naïve lymphocytes (Goetz et al., 2001;
Grabar et al., 2004; Manfredi and Chiodo, 2000; Manfredi et al.,
2003; Cohen Stuart et al., 2002; Lederman et al., 2000; Teixeira
et al., 2001; Kalayjian et al., 2005; Yamashita et al., 2001). A few
authors have described an inverse relationship between age and
immunologic HAART response (Viard et al., 2001; Florence et al.,
2003). In contrast, other studies have not observed such relation-
ships (Grimes et al., 2002; Knobel et al., 2001; Tumbarello et al.,
2004; Nogueras et al., 2006).
Although the immunologic response to HAART in older patients
is controversial, there is no doubt regarding a better virologic
response (Paredes et al., 2000; Wellons et al., 2002) and a signif-
icant reduction in the mortality among them (Perez and Moore,
2003). Better compliance and lower treatment abandonment might
explain these results (Hinkin et al., 2004). Additionally, advanced
age may be associated with lower viral load independent of
antiretroviral use (Goodkin et al., 2004).
The higher prevalence of comorbidities and polypharmacy in
HIV-infected older individuals can influence the occurrence of
adverse effects of antiretroviral drugs but do not seem to adversely
affect treatment (Shah et al., 2002; Adeyemi et al., 2003). HAART
discontinuation because of adverse effects occurs at earlier stages
of treatment in patients over 50 years of age. Treatment discontin-
uation as the result of neuropsychiatric and hematologic adverse
effects were more frequent among older patients than younger
patients (Cuzin et al., 2007).
4. Discussion
Aging is one of the current characteristics of the HIV epidemic.
It is not only a consequence of effective treatment and care of peo-
ple living with HIV/AIDS but also an effect of increasing sexual
transmission of HIV among older individuals (Myers, 2009).
Clinically, older patients with HIV infection are especially prone
to infectious and neoplastic complications and death (Ena et al.,
1998; Soriano et al., 1998; Carre et al., 1994; Skiest et al., 1996;
Butt et al., 2001), and several symptoms of HIV infection can be
confounded by the natural processes of aging (Sanders et al., 2008).
However, this population has better treatment compliance com-
pared to young people despite more frequent comorbidities and
polypharmacy (Hinkin et al., 2004; Shah et al., 2002; Adeyemi
et al., 2003). Therefore, the interest in detecting HIV infection in the
elderly is essential for appropriate early diagnosis and treatment
and to prevent serious complications.
The senescence process affects the complex interaction between
HIV infection and the immune system. Sufficient knowledge of
these changes is essential to improve the care of this growing pop-
ulation.
Both HIV infection and aging contribute to changes and dysfunc-
tion in the bone marrow, thymus and GALT, particularly in terms of
the number and function of immune and stromal cells (Compston,
2002; Isgro et al., 2005; Alexaki and Wigdahl, 2008; Tripathi et al.,
2005; Steinmann, 1986; Aw et al., 2007; Gruver et al., 2007; Effros
et al., 2008; Brenchley and Douek, 2008). Some of these alterations
may be related to the decreased production of growth hormone
and interleukin-7 (Lamberts et al., 1997; French et al., 2002; Kang
et al., 2004; Tsuboi et al., 2004; Beq et al., 2004; Napolitano et al.,
2002). The knowledge about pathophysiology of HIV infection in
the elderly will be beneficial for the development and use of some
immune-based therapies (Deeks, 2009).
Innate immunity is equally affected by HIV infection and senes-
cence; there is a decline in the macrophages’ and neutrophils’
microbicidal capability, monocytes’ expression of toll-like recep-
tors, dendritic cell production of interleukin-12 and in natural killer
cell function, in addition to an increase in neutrophils’ susceptibil-
ity to apoptosis (Plackett et al., 2004; Gomez et al., 2005; Butcher
et al., 2001; Torre et al., 2002; Noursadeghi et al., 2006; Panda
et al., 2009; Renshaw et al., 2002; Agrawal et al., 2007; Della Bella
et al., 2007; Mantegani et al., 2009). Some experimental and clinical
studies have tried to enhance the innate immunological response
to both HIV infection and aging with several different substances.
Zinc supplementation showed beneficial effects in older individu-
Author's personal copy
168 V.I. Avelino-Silva et al. / Ageing Research Reviews 10 (2011) 163–172
Table 1
Antiretroviral therapy response in older HIV-infected individuals.
Author, year Number of patients Design Follow-up (months) HAART response Observation
V. I.
Lederman et al., 2000 71 Prospective cohort 12 NE Naïve CD4+T cell increase inversely related to age following dual
therapy
Manfredi and Chiodo, 2000 105 Retrospective cohort 12 = Worse immunological and similar virological response over age 55
following first line HAART
Paredes et al., 2000 1469 Prospective cohort 9–20 NE Older patients more likely to achieve virological success
Goetz et al., 2001 80 Retrospective cohort Mean 16.7 NE Among virological responders, each decade of age decreased CD4+T
cell count response by 35 cells
Knobel et al., 2001 699 Prospective cohort 24 = = Trend towards better adherence, virological and immunological
response over 60
Teixeira et al., 2001 22 Retrospective case-control 12 NE Poor CD4+T cell responders were older than good responders
Viard et al., 2001 1956 Prospective cohort 12–31 NE Inverse relation between age and CD4+T cell increase
Yamashita et al., 2001 397 Prospective cohort 30–60 NE Worse short-term (6 months) CD4+T cell increase with older age
Grimes et al., 2002 104 Retrospective cohort 24–102 = = No differences in AIDS-related mortality, CD4+T cell counts or viral
load
Cohen Stuart et al., 2002 45 Prospective cohort 12 NE Naive T-cell recovery rate inversely related with age
Wellons et al., 2002 303 Retrospective cohort Mean 29 = Better virological response and similar immunological response
Florence et al., 2003 225 Transversal Not applied NE Unsatisfactory CD4+T cell response increases with age despite good
virological response
Manfredi et al., 2003 57 Retrospective cohort 12 = Lower immunological response in patients aged 65 and older,
compared to those aged 55–65
Perez and Moore, 2003 797 Retrospective cohort 36.5 NE NE Older patients have a greater survival benefit with HAART
Goodkin et al., 2004 135 Transversal Not applied NE Lower plasma viral load with older age independent of antiretroviral
use
Grabar et al., 2004 3015 Prospective cohort Mean 31.5 ↑↓Slower CD4+T cell response and higher risk of clinical progression
despite better virologic response over age 50
Tumbarello et al., 2004 243 Prospective cohort 6–78 = = No differences in virological or immunological responses
Kalayjian et al., 2005 80 Prospective cohort 12 = Smaller increases in naive T cells and similar virological responses over
age 45
Nogueras et al., 2006 455 Prospective cohort 3–66 = Similiar virological response, lower CD4+T-cell response and faster
progression to AIDS in older patients.
V., virological responde to HAART; I., immunological response to HAART; NE, not evaluated; , higher response; , lower response; =, similar response.
Author's personal copy
V.I. Avelino-Silva et al. / Ageing Research Reviews 10 (2011) 163–172 169
Fig. 1. Expected immune consequences of HIV infection in the elderly.
als, but there were contradictory results regarding its effects on HIV
infection (Prasad, 2009). Melatonin, a neurohormone secreted by
the pineal gland, has been shown to have a potentially therapeutic
value in older patients (Cardinali et al., 2008).
CD4+and CD8+T cells depletion and dysfunction caused by
chronic immune activation in HIV infection seems to worsen with
senescence (Vrisekoop et al., 2009; Douek et al., 2003; Haynes and
Maue, 2009; Licastro et al., 2005). Aging may theoretically decrease
the CD4+T cell response to HAART (Goetz et al., 2001; Grabar et al.,
2004; Manfredi and Chiodo, 2000; Manfredi et al., 2003; Cohen
Stuart et al., 2002; Lederman et al., 2000; Teixeira et al., 2001;
Kalayjian et al., 2005; Yamashita et al., 2001; Viard et al., 2001;
Florence et al., 2003). Therefore, it is necessary to emphasize the
importance of early diagnosis and treatment in older HIV-infected
patients.
Older individuals exhibit deficient humoral immune responses,
including a greater proportion of exhausted B cells and produc-
tion of less avid and specific antibodies (Weksler and Szabo, 2000;
Lange et al., 2003). Although HIV infection stimulates B-lymphocyte
growth and activation, there is a polyclonal activation with poorly
inducible antibody responses (Moir and Fauci, 2009). Consequently,
HIV infection and aging equally worsen humoral immunity, pre-
disposing an individual to infections and to a reduced efficacy of
vaccines.
5. Conclusion
In conclusion, HIV infection and aging undoubtedly contribute
equally to a complex immune dysfunction and to increased mor-
bidity and mortality in older HIV-infected individuals. Several
studies are currently trying to better understand the immune con-
sequences of the coexistence of these two clinical conditions and
how to manage them. However, evidence does favor the benefi-
cial use of HAART for older patients, which results in a virologic,
immunologic, and clinical response and a reduction of mortality.
One should, therefore, not use age as a discouraging factor in treat-
ing HIV-infected patients but as a stimulus to pursue early diagnosis
and thus, obtain the best possible treatment responses and clinical
outcomes.
References
Pawelec, G., 1999. Immunosenescence: impact in the young as well as the old? Mech.
Ageing Dev. 108 (1), 1–7.
Haynes, L., 2005. The effect of aging on cognate function and development of immune
memory. Curr. Opin. Immunol. 17 (5), 476–479.
Brasil. Ministério da Saúde. Departamento de informática do SUS. Casos de Aids
notificados no Brasil desde 1980. Available from: http://w3.datasus.gov.br/
datasus/datasus.php.
Stall, R., Catania, J., 1994. AIDS risk behaviors among late middle-aged and elderly
Americans. The National AIDS Behavioral Surveys. Arch Int. Med. 154 (1), 57–63.
Definition of An Older or Elderly Person. World Health Organization. 2010
06/10/2010. Available from: http://www.who.int/healthinfo/survey/
ageingdefnolder/en/index.html.
Linsk, N.L., 2000. HIV among older adults: age-specific issues in prevention and
treatment. AIDS Read. 10 (7), 430–440.
Gavazzi, G., Krause, K.H., 2002. Ageing and infection. Lancet Infect. Dis. 2 (11),
659–666.
Kovaiou, R.D., Herndler-Brandstetter, D., Grubeck-Loebenstein, B., 2007. Age-related
changes in immunity: implications for vaccination in the elderly. Expert Rev.
Mol. Med. 9 (3), 1–17.
Castle, S.C., 2000. Clinical relevance of age-related immune dysfunction. Clin. Infect.
Dis. 31 (2), 578–585.
Prevention and Control of Influenza: Recommendations of the Advisory Committee
on Immunization Practices (ACIP). MMWR Recomm Rep, 1999. 48(RR-4), pp.
1–28.
Webster, R.G., 2000. Immunity to influenza in the elderly. Vaccine 18 (16),
1686–1689.
Author's personal copy
170 V.I. Avelino-Silva et al. / Ageing Research Reviews 10 (2011) 163–172
Musher, D.M., et al., 1986. Natural and vaccine-related immunity to Streptococcus
pneumoniae. J. Infect. Dis. 154 (2), 245–256.
Carvalho, J., 1970. Fecundidade e Mortalidade no Brasil, 1960/1970. 1978, Belo Hor-
izonte: CEDEPLAR/UFMG.
Phillips, A.N., et al., 1991. More rapid progression to AIDS in older HIV-infected
people: the role of CD4+T-cell counts. J. Acquir. Immune Defic. Syndr. 4 (10),
970–975.
Operskalski, E.A., et al., Transfusion Safety Study Group, 1995. Human immunod-
eficiency virus type 1 infection: relationship of risk group and age to rate of
progression to AIDS. J. Infect. Dis. 172 (3), 648–655.
O’Brien, T.R., et al., 1996. Serum HIV-1 RNA levels and time to development of AIDS
in the Multicenter Hemophilia Cohort Study. JAMA 276 (2), 105–110.
Ferro, S., Salit, I.E., 1992. HIV infection in patients over 55 years of age. J. Acquir.
Immune Defic. Syndr. 5 (4), 348–353.
Ena, J., et al., 1998. Clinical presentation of HIV infection in patients aged 50 years
or older. J. Infect. 37 (3), 213–216.
Sanders, G.D., et al., 2008. Cost-effectiveness of HIV screening in patients older than
55 years of age. Ann. Int. Med. 148 (12), 889–903.
Castro, M.P., 2007. O viver com HIV/aids na perspectiva de pessoas idosas atendidas
em ambulatório especializado da cidade de São Paulo in Faculdade de Medicina.
2007, Universidade de São Paulo: São Paulo.
Babiker, A.G., et al., 2001. Age as a determinant of survival in HIV infection. J. Clin.
Epidemiol. 54 (Suppl. 1), S16–21.
Soriano, V., et al., 1998. The decline in CD4+T lymphocytes as a function of the
duration of HIV infection, age at seroconversion, and viral load. J. Infect. 36 (3),
307–311.
Carre, N., et al., SEROCO Study Group, 1994. Effect of age and exposure group on the
onset of AIDS in heterosexual and homosexual HIV-infected patients. Aids 8 (6),
797–802.
Skiest, D.J., et al., 1996. The importance of comorbidity in HIV-infected patients over
55: a retrospective case-control study. Am. J. Med. 101 (6), 605–611.
Butt, A.A., et al., 2001. Human immunodeficiency virus infection in elderly patients.
South Med. J. 94 (4), 397–400.
Compston, J.E., 2002. Bone marrow and bone: a functional unit. J. Endocrinol. 173
(3), 387–394.
Lamberts, S.W., van den Beld, A.W., van der Lely, A.J., 1997. The endocrinology of
aging. Science 278 (5337), 419–424.
French, R.A., et al., 2002. Age-associated loss of bone marrow hematopoietic cells is
reversed by GH and accompanies thymic reconstitution. Endocrinology 143 (2),
690–699.
Isgro, A., et al., 2005. Immunodysregulation of HIV disease at bone marrow level.
Autoimmun. Rev. 4 (8), 486–490.
Alexaki, A., Wigdahl, B., 2008. HIV-1 infection of bone marrow hematopoietic pro-
genitor cells and their role in trafficking and viral dissemination. PLoS Pathog. 4
(12), pe1000215.
Tripathi, A.K., et al., 2005. Bone marrow abnormalities in HIV disease. J. Assoc. Physi-
cians India 53, 705–710.
Wang, C.Q., et al., 1995. Effect of age on marrow macrophage number and function.
Aging (Milano) 7 (5), 379–384.
Kang, I., et al., 2004. Age-associated change in the frequency of memory CD4+T cells
impairs long term CD4+T cell responses to influenza vaccine. J. Immunol. 173
(1), 673–681.
Tsuboi, I., et al., 2004. Senescent B lymphopoiesis is balanced in suppressive home-
ostasis: decrease in interleukin-7 and transforming growth factor-beta levels in
stromal cells of senescence-accelerated mice. Exp. Biol. Med. (Maywood) 229
(6), 494–502.
Beq, S., Delfraissy, J.F., Theze, J., 2004. Interleukin-7 (IL-7): immune function, involve-
ment in the pathogenesis of HIV infection and therapeutic potential. Eur.
Cytokine Netw. 15 (4), 279–289.
Steinmann, G.G., 1986. Changes in the human thymus during aging. Curr. Top Pathol.
75, 43–88.
Aw, D., Silva, A.B., Palmer, D.B., 2007. Immunosenescence: emerging challenges for
an ageing population. Immunology 120 (4), 435–446.
Gruver, A.L., Hudson, L.L., Sempowski, G.D., 2007. Immunosenescence of ageing. J.
Pathol. 211 (2), 144–156.
Douek, D.C., et al., 1998. Changes in thymic function with age and during the treat-
ment of HIV infection. Nature 396 (6712), 690–695.
Casau, N.C., 2005. Perspective on HIV infection and aging: emerging research on the
horizon. Clin. Infect. Dis. 41 (6), 855–863.
Napolitano, L.A., et al., 2002. Increased thymic mass and circulating naive CD4 T cells
in HIV-1-infected adults treated with growth hormone. Aids 16 (8), 1103–1111.
Effros, R.B., et al., 2008. Aging and infectious diseases: workshop on HIV infection
and aging: what is known and future research directions. Clin. Infect. Dis. 47 (4),
542–553.
Brenchley, J.M., Douek, D.C., 2008. HIV infection and the gastrointestinal immune
system. Mucosal Immunol. 1 (1), 23–30.
Plackett, T.P., et al., 2004. Aging and innate immune cells. J. Leukoc. Biol. 76 (2),
291–299.
Gomez, C.R., Boehmer, E.D., Kovacs, E.J., 2005. The aging innate immune system.
Curr. Opin. Immunol. 17 (5), 457–462.
Butcher, S.K., et al., 2001. Senescence in innate immune responses: reduced neu-
trophil phagocytic capacity and CD16 expression in elderly humans. J. Leukoc.
Biol. 70 (6), 881–886.
Torre, D., et al., 2002. Impaired macrophage phagocytosis of apoptotic neutrophils
in patients with human immunodeficiency virus type 1 infection. Clin. Diagn.
Lab. Immunol. 9 (5), 983–986.
Noursadeghi, M., Katz, D.R., Miller, R.F., 2006. HIV-1 infection of mononuclear phago-
cytic cells: the case for bacterial innate immune deficiency in AIDS. Lancet Infect.
Dis. 6 (12), 794–804.
Crowe, S., Zhu, T., Muller, W.A., 2003. The contribution of monocyte infection and
trafficking to viral persistence, and maintenance of the viral reservoir in HIV
infection. J. Leukoc. Biol. 74 (5), 635–641.
Orenstein, J.M., 2001. The macrophage in HIV infection. Immunobiology 204 (5),
598–602.
Panda, A., et al., 2009. Human innate immunosenescence: causes and consequences
for immunity in old age. Trends Immunol. 30 (7), 325–333.
Renshaw, M., et al., 2002. Cutting edge: impaired toll-like receptor expression and
function in aging. J. Immunol. 169 (9), 4697–4701.
Wu, D., Hayek, M.G., Meydani, S., 2001. Vitamin E and macrophage cyclooxygenase
regulation in the aged. J. Nutr. 131 (2), S382–S388.
Chevalier, M.P.A., Jolin, J.S., Addo, M., Kwon, D.S., Toth, I., Walker, B., Streeck, H., 2009.
Skewed HIV-1-specific CD4+Th2 helper cell contribution in progressive HIV-1
infection. Retrovirology 6 (S3), 260.
Wu, L., KewalRamani, V.N., 2006. Dendritic–cell interactions with HIV: infection and
viral dissemination. Nat. Rev. Immunol. 6 (11), 859–868.
Coleman, C.M., Wu, L., 2009. HIV interactions with monocytes and dendritic cells:
viral latency and reservoirs. Retrovirology 6, 51.
Lu, W., et al., 2004. Therapeutic dendritic–cell vaccine for chronic HIV-1 infection.
Nat. Med. 10 (12), 1359–1365.
Agrawal, A., et al., 2007. Altered innate immune functioning of dendritic cells
in elderly humans: a role of phosphoinositide 3-kinase-signaling pathway. J.
Immunol. 178 (11), 6912–6922.
Della Bella, S., et al., 2007. Peripheral blood dendritic cells and monocytes are dif-
ferently regulated in the elderly. Clin. Immunol. 122 (2), 220–228.
Fauci, A.S., Mavilio, D., Kottilil, S., 2005. NK cells in HIV infection: paradigm for
protection or targets for ambush. Nat. Rev. Immunol. 5 (11), 835–843.
Mantegani, P., et al., 2009. Perturbation of the natural killer cell compartment dur-
ing primary human immunodeficiency virus 1 infection primarily involving the
CD56(bright) subset. Immunology.
Vrisekoop, N., Mandl, J.N., Germain, R.N., 2009. Life and death as a T lymphocyte:
from immune protection to HIV pathogenesis. J. Biol. 8 (10), 91.
Veazey, R.S., et al., 1998. Gastrointestinal tract as a major site of CD4+T cell depletion
and viral replication in SIV infection. Science 280 (5362), 427–431.
Kewenig, S., et al., 1999. Rapid mucosal CD4(+) T-cell depletion and enteropathy
in simian immunodeficiency virus-infected rhesus macaques. Gastroenterology
116 (5), 1115–1123.
Chun, T.W., et al., 1997. Quantification of latent tissue reservoirs and total body viral
load in HIV-1 infection. Nature 387 (6629), 183–188.
Haase, A.T., 1999. Population biology of HIV-1 infection: viral and CD4+T cell
demographics and dynamics in lymphatic tissues. Annu. Rev. Immunol. 17,
625–656.
Haase, A.T., et al., 1996. Quantitative image analysis of HIV-1 infection in lymphoid
tissue. Science 274 (5289), 985–989.
Anderson, R.W., Ascher, M.S., Sheppard, H.W., 1998. Direct HIV cytopathicity cannot
account for CD4 decline in AIDS in the presence of homeostasis: a worst-
case dynamic analysis. J. Acquir. Immune Defic. Syndr. Hum. Retrovirol. 17 (3),
245–252.
Douek, D.C., et al., 2002. HIV preferentially infects HIV-specific CD4+T cells. Nature
417 (6884), 95–98.
Douek, D.C., Picker, L.J., Koup, R.A., 2003. T cell dynamics in HIV-1 infection. Annu.
Rev. Immunol. 21, 265–304.
Simmonds, P., et al., 1991. Determinants of HIV disease progression: six-year lon-
gitudinal study in the Edinburgh haemophilia/HIV cohort. Lancet 338 (8776),
1159–1163.
Leng, Q., et al., 2001. Immune activation correlates better than HIV plasma viral load
with CD4 T-cell decline during HIV infection. J. Acquir. Immune Defic. Syndr. 27
(4), 389–397.
Roussanov, B.V., Taylor, J.M., Giorgi, J.V., 2000. Calculation and use of an HIV-1 disease
progression score. AIDS 14 (17), 2715–2722.
Giorgi, J.V., et al., 1999. Shorter survival in advanced human immunodeficiency virus
type 1 infection is more closely associated with T lymphocyte activation than
with plasma virus burden or virus chemokine coreceptor usage. J. Infect. Dis.
179 (4), 859–870.
Mandl, J.N., et al., 2008. Divergent TLR7 and TLR9 signaling and type I interferon pro-
duction distinguish pathogenic and nonpathogenic AIDS virus infections. Nat.
Med. 14 (10), 1077–1087.
Meier, A., et al., 2009. Sex differences in the Toll-like receptor-mediated response of
plasmacytoid dendritic cells to HIV-1. Nat. Med. 15 (8), 955–959.
Brenchley, J.M., et al., 2006. Microbial translocation is a cause of systemic immune
activation in chronic HIV infection. Nat. Med. 12 (12), 1365–1371.
Brenchley, J.M., et al., 2008. Differential Th17 CD4 T-cell depletion in pathogenic and
nonpathogenic lentiviral infections. Blood 112 (7), 2826–2835.
Fazekas de St Groth, B., Landay, A.L., 2008. Regulatory T cells in HIV infection:
pathogenic or protective participants in the immune response? AIDS 22 (6),
671–683.
Grossman, Z., et al., 2002. CD4+T-cell depletion in HIV infection: are we closer to
understanding the cause? Nat. Med. 8 (4), 319–323.
Haynes, L., et al., 2003. CD4 T cell memory derived from young naive cells functions
well into old age, but memory generated from aged naive cells functions poorly.
Proc. Natl. Acad. Sci. U.S.A. 100 (25), 15053–15058.
Haynes, L., et al., 2005. Newly generated CD4 T cells in aged animals do not exhibit
age-related defects in response to antigen. J. Exp. Med. 201 (6), 845–851.
Author's personal copy
V.I. Avelino-Silva et al. / Ageing Research Reviews 10 (2011) 163–172 171
Haynes, L., Maue, A.C., 2009. Effects of aging on T cell function. Curr. Opin. Immunol.
21 (4), 414–417.
Pawelec, G., et al., 2009. Cytomegalovirus and human immunosenescence. Rev. Med.
Virol. 19 (1), 47–56.
Derhovanessian, E., Larbi, A., Pawelec, G., 2009. Biomarkers of human immunose-
nescence: impact of Cytomegalovirus infection. Curr. Opin. Immunol. 21 (4),
440–445.
Nasi, M., et al., 2006. Thymic output and functionality of the IL-7/IL-7 receptor system
in centenarians: implications for the neolymphogenesis at the limit of human
life. Aging Cell 5 (2), 167–175.
Effros, R.B., et al., 1996. Shortened telomeres in the expanded CD28–CD8+cell subset
in HIV disease implicate replicative senescence in HIV pathogenesis. Aids 10 (8),
F17–22.
Hazenberg, M.D., et al., 2000. T cell depletion in HIV-1 infection: how CD4+T cells
go out of stock. Nat. Immunol. 1 (4), 285–289.
Bestilny, L.J., et al., 2000. Accelerated replicative senescence of the peripheral
immune system induced by HIV infection. Aids 14 (7), 771–780.
Miller, R.A., et al., 1997. Early activation defects in T lymphocytes from aged mice.
Immunol. Rev. 160, 79–90.
Nel, A.E., Slaughter, N., 2002. T-cell activation through the antigen receptor. Part 2:
role of signaling cascades in T-cell differentiation, anergy, immune senescence,
and development of immunotherapy. J. Allergy Clin. Immunol. 109 (6), 901–915.
Kapasi, Z.F., et al., 2002. Defective generation but normal maintenance of memory T
cells in old mice. Eur. J. Immunol. 32 (6), 1567–1573.
Saha, K., Zhang, J., Zerhouni, B., 2001. Evidence of productively infected CD8+T cells
in patients with AIDS: implications for HIV-1 pathogenesis. J. Acquir. Immune
Defic. Syndr. 26 (3), 199–207.
Piazza, P., Fan, Z., Rinaldo Jr., C.R., 2002. CD8+T-cell immunity to HIV infection. Clin.
Lab. Med. 22 (3), 773–797.
Effros, R.B., 2004. T cell replicative senescence: pleiotropic effects on human aging.
Ann. NY Acad. Sci. 1019, 123–126.
Effros, R.B., Pawelec, G., 1997. Replicative senescence of T cells: does the Hayflick
Limit lead to immune exhaustion? Immunol. Today 18 (9), 450–454.
Plunkett, F.J., et al., 2007. The loss of telomerase activity in highly differentiated
CD8 + CD28–CD27T cells is associated with decreased Akt (Ser473) phospho-
rylation. J. Immunol. 178 (12), 7710–7719.
Hadrup, S.R., et al., 2006. Longitudinal studies of clonally expanded CD8 T cells reveal
a repertoire shrinkage predicting mortality and an increased number of dysfunc-
tional cytomegalovirus-specific T cells in the very elderly. J. Immunol. 176 (4),
2645–2653.
Pawelec, G., et al., 2004. Is immunosenescence infectious? Trends Immunol. 25 (8),
406–410.
Akbar, A.N., Fletcher, J.M., 2005. Memory T cell homeostasis and senescence during
aging. Curr. Opin. Immunol. 17 (5), 480–485.
Wikby, A., et al., 1994. Age-related changes in immune parameters in a very old
population of Swedish people: a longitudinal study. Exp. Gerontol. 29 (5),
531–541.
Wikby, A., et al., 1998. Changes in CD8 and CD4 lymphocyte subsets, T cell prolif-
eration responses and non-survival in the very old: the Swedish longitudinal
OCTO-immune study. Mech. Ageing Dev. 102 (2–3), 187–198.
Saurwein-Teissl, M., et al., 2002. Lack of antibody production following immuniza-
tion in old age: association with CD8(+)CD28) T cell clonal expansions and an
imbalance in the production of Th1 and Th2 cytokines. J. Immunol. 168 (11),
5893–5899.
Moanna, A., et al., 2005. CD4+T-cell depletion in HIV infection: killed by friendly
fire? Curr. HIV/AIDS Rep. 2 (1), 16–23.
Effros, R.B., 2000. Costimulatory mechanisms in the elderly. Vaccine 18 (16),
1661–1665.
Cao, W., et al., 2009a. Premature aging of T cells is associated with faster HIV-1
disease progression. J. Acquir. Immune Defic. Syndr. 50 (2), 137–147.
Larbi, A., et al., 2006. Differential role of lipid rafts in the functions of CD4+and CD8+
human T lymphocytes with aging. Cell Signal. 18 (7), 1017–1030.
Dube, M.P., et al., 2003. Guidelines for the evaluation and management of dys-
lipidemia in human immunodeficiency virus (HIV)-infected adults receiving
antiretroviral therapy: recommendations of the HIV Medical Association of the
Infectious Disease Society of America and the Adult AIDS Clinical Trials Group.
Clin. Infect. Dis. 37 (5), 613–627.
Weksler, M.E., Szabo, P., 2000. The effect of age on the B-cell repertoire. J. Clin.
Immunol. 20 (4), 240–249.
Szabo, P., et al., 1998. Maturation of B cell precursors is impaired in thymic-deprived
nude and old mice. J. Immunol. 161 (5), 2248–2253.
Moir, S., Fauci, A.S., 2009. B cells in HIV infection and disease. Nat. Rev. Immunol. 9
(4), 235–245.
Caruso, C., et al., 2009. Mechanisms of immunosenescence. Immun. Ageing 6, 10.
Ben-Yehuda, A., et al., 1998. Increased VH 11 and VH Q52 gene use by splenic B cells
in old mice associated with oligoclonal expansions of CD5+B cells. Mech. Ageing
Dev. 103 (2), 111–121.
Hallgren, H.M., et al., 1973. Lymphocyte phytohemagglutinin responsiveness,
immunoglobulins and autoantibodies in aging humans. J. Immunol. 111 (4),
1101–1107.
Weinberger, B., et al., 2008. Biology of immune responses to vaccines in elderly
persons. Clin. Infect. Dis. 46 (7), 1078–1084.
Weinberg, A., et al., 2008. In vitro cell-mediated immune responses of
human immunodeficiency virus-infected and -uninfected individuals to whole
cytomegalovirus antigens and their subunits. Clin. Vaccine Immunol. 15 (9),
1398–1409.
Doria-Rose, N.A., Connors, M., 2009. Antibody-secreting B cells in HIV infection. Curr.
Opin. HIV AIDS 4 (5), 426–430.
Lange, C.G., et al., 2003. Nadir CD4+T-cell count and numbers of CD28+ CD4+T-cells
predict functional responses to immunizations in chronic HIV-1 infection. Aids
17 (14), 2015–2023.
Belkaid, Y., Rouse, B.T., 2005. Natural regulatory T cells in infectious disease. Nat.
Immunol. 6 (4), 353–360.
Dejaco, C., Duftner, C., Schirmer, M., 2006. Are regulatory T-cells linked with aging?
Exp. Gerontol. 41 (4), 339–345.
Rouse, B.T., Suvas, S., 2007. Regulatory T cells and immunity to pathogens. Expert
Opin. Biol. Ther. 7 (9), 1301–1309.
Maloy, K.J., et al., 2003. CD4+CD25+T(R) cells suppress innate immune pathology
through cytokine-dependent mechanisms. J. Exp. Med. 197 (1), 111–119.
Sugimoto, K., et al., 2003. Suppression of HCV-specific T cells without differential
hierarchy demonstrated ex vivo in persistent HCV infection. Hepatology 38 (6),
1437–1448.
Boyer, O., et al., 2004. CD4+CD25+regulatory T-cell deficiency in patients with
hepatitis C-mixed cryoglobulinemia vasculitis. Blood 103 (9), 3428–3430.
Cao, W., et al., 2009b. Regulatory T cell expansion and immune activation during
untreated HIV type 1 infection are associated with disease progression. AIDS
Res. Hum. Retroviruses 25 (2), 183–191.
Licastro, F., et al., 2005. Innate immunity and inflammation in ageing: a key for
understanding age-related diseases. Immun. Ageing 2, 8.
Fagiolo, U., et al., 1993. Increased cytokine production in mononuclear cells of
healthy elderly people. Eur. J. Immunol. 23 (9), 2375–2378.
Bruunsgaard, H., Pedersen, M., Pedersen, B.K., 2001. Aging and proinflammatory
cytokines. Curr. Opin. Hematol. 8 (3), 131–136.
McGeer, P.L., McGeer, E.G., 2004. Inflammation and the degenerative diseases of
aging. Ann. NY Acad. Sci. 1035, 104–116.
Cuellar, M.L., Espinoza, L.R., 2000. Rheumatic manifestations of HIV-AIDS. Baillieres
Best Pract. Res. Clin. Rheumatol. 14 (3), 579–593.
Dlova, N.C., Mosam, A., 2006. Inflammatory noninfectious dermatoses of HIV. Der-
matol. Clin. 24 (4), 439–448, vi.
Goetz, M.B., et al., 2001. Decreased recovery of CD4 lymphocytes in older HIV-
infected patients beginning highly active antiretroviral therapy. Aids 15 (12),
1576–1579.
Grabar, S., et al., 2004. Immunologic and clinical responses to highly active antiretro-
viral therapy over 50 years of age. Results from the French Hospital Database on
HIV. Aids 18 (15), 2029–2038.
Manfredi, R., Chiodo, F., 2000. A case-control study of virological and immunologi-
cal effects of highly active antiretroviral therapy in HIV-infected patients with
advanced age. Aids 14 (10), 1475–1477.
Manfredi, R., et al., 2003. Antiretroviral treatment and advanced age: epidemiologic,
laboratory, and clinical features in the elderly. J. Acquir. Immune Defic. Syndr.
33 (1), 112–114.
Cohen Stuart, J., et al., 2002. Reconstitution of naive T cells during antiretroviral
treatment of HIV-infected adults is dependent on age. Aids 16 (17), 2263–2266.
Lederman, M.M., et al., 2000. Cellular restoration in HIV infected persons treated
with abacavir and a protease inhibitor: age inversely predicts naive CD4 cell
count increase. Aids 14 (17), 2635–2642.
Teixeira, L., et al., 2001. Poor CD4 T cell restoration after suppression of HIV-1 repli-
cation may reflect lower thymic function. Aids 15 (14), 1749–1756.
Kalayjian, R.C., et al., 2005. Distinct mechanisms of T cell reconstitution can be iden-
tified by estimating thymic volume in adult HIV-1 disease. J. Infect. Dis. 192 (9),
1577–1587.
Yamashita, T.E., et al., 2001. Immunologic and virologic response to highly active
antiretroviral therapy in the Multicenter AIDS Cohort Study. Aids 15 (6),
735–746.
Viard, J.P., et al., 2001. Influence of age on CD4 cell recovery in human immunod-
eficiency virus-infected patients receiving highly active antiretroviral therapy:
evidence from the EuroSIDA study. J. Infect. Dis. 183 (8), 1290–1294.
Florence, E., et al., 2003. Factors associated with a reduced CD4 lymphocyte count
response to HAART despite full viral suppression in the EuroSIDA study. HIV
Med. 4 (3), 255–262.
Grimes, R.M., et al., 2002. Clinical experience with human immunodeficiency virus-
infected older patients in the era of effective antiretroviral therapy. Clin. Infect.
Dis. 34 (11), 1530–1533.
Knobel, H., et al., 2001. Response to highly active antiretroviral therapy in HIV-
infected patients aged 60 years or older after 24 months follow-up. Aids 15
(12), 1591–1593.
Tumbarello, M., et al., 2004. Older age does not influence CD4 cell recovery in HIV-1
infected patients receiving highly active antiretroviral therapy. BMC Infect. Dis.
4, 46.
Nogueras, M., et al., 2006. Epidemiological and clinical features, response to HAART,
and survival in HIV-infected patients diagnosed at the age of 50 or more. BMC
Infect. Dis. 6, 159.
Paredes, R., et al., 2000. Predictors of virological success and ensuing failure in HIV-
positive patients starting highly active antiretroviral therapy in Europe: results
from the EuroSIDA study. Arch Int. Med. 160 (8), 1123–1132.
Wellons, M.F., et al., 2002. HIV infection: treatment outcomes in older and younger
adults. J. Am. Geriatr. Soc. 50 (4), 603–607.
Perez, J.L., Moore, R.D., 2003. Greater effect of highly active antiretroviral therapy
on survival in people aged > or =50 years compared with younger people in an
urban observational cohort. Clin. Infect. Dis. 36 (2), 212–218.
Hinkin, C.H., et al., 2004. Medication adherence in HIV-infected adults: effect of
patient age, cognitive status, and substance abuse. Aids 18 (Suppl. 1), S19–25.
Author's personal copy
172 V.I. Avelino-Silva et al. / Ageing Research Reviews 10 (2011) 163–172
Goodkin, K., et al., 2004. Older age and plasma viral load in HIV-1 infection. Aids 18
(Suppl. 1), S87–98.
Shah, S.S., et al., 2002. Comorbid conditions, treatment, and health maintenance in
older persons with human immunodeficiency virus infection in New York City.
Clin. Infect. Dis. 35 (10), 1238–1243.
Adeyemi, O.M., et al., 2003. HIV infection in older patients. Clin. Infect. Dis. 36 (10),
1347.
Cuzin, L., et al., 2007. Immunologic and clinical responses to highly active antiretro-
viral therapy in patients with HIV infection aged >50 years. Clin. Infect. Dis. 45
(5), 654–657.
Myers, J.D., 2009. Growing old with HIV: the AIDS epidemic and an aging population.
JAAPA 22 (1), 20–24.
Deeks, S.G., 2009. Immune dysfunction, inflammation, and accelerated aging in
patients on antiretroviral therapy. Top. HIV Med. 17 (4), 118–123.
Prasad, A.S., 2009. Zinc: role in immunity, oxidative stress and chronic inflammation.
Curr. Opin. Clin. Nutr. Metab. Care 12 (6), 646–652.
Cardinali, D.P., et al., 2008. Melatonin and the immune system in aging. Neuroim-
munomodulation 15 (4–6), 272–278.
... For Latino women over 50 years if age, increased risk is associated with aspects of aging and culture [3]. Age presents additional population-specific risk reasons contributing to poor utilization of diagnostic, treatment and prevention services [8,9]. With over half of the 1.1 million PLWH in the US age 50 years of age and over, and rates for older Latinos five times higher than their non-Hispanic White counterparts, sexual risk behaviors of older Latino women must be explored to inform targeted prevention efforts [3,8]. ...
... Studies that attempt to use theoretical frameworks for behavioral predicators of HIV risk in women have focused on the younger women of adolescence and reproductive age [10,11]. Yet, considerable measurement errors exists to capture demographic characteristics, beliefs and attitudes in studies of younger women, where a variety of other reasons contribute to risk behavior (e.g., pregnancy) [9,11]. This results in the inability to quantify and relate observed effects to women midlife and older [5]. ...
Article
Full-text available
Background: The feminization and ethnic diversification of HIV infection, has resulted in a call for gender- and culture-specific prevention strategies for at-risk groups including Latinos in the United States. The steadily changing demographic profile of the AIDS epidemic challenges prevention strategies to remain relevant and up-to-date, particularly in populations of women midlife and older where an understanding of risk remains under explored. As the CDC requests country-specific HIV risk profiles for Latino communities in the US, understanding the socio-economic, behavioral and personal risk reasons of HIV risk for older Dominican women is critical for prevention. Methods: We conducted focus group discussions informed by the Theory of Gender and Power (TGP). The three constructs of the TGP: 1) Affective influences/social norms; 2) Gender-specific norms and. 3) Power and Authority guided the thematic analysis and identified themes that described the socio-cultural and contextual reasons that that contribute to perceptions of HIV risk. Results: Sixty Dominican American women ages 57-73 participated in our focus group discussions. Sexual Division of Labour: 1) Economic Dependence; 2) Financial Need and 3) Education and Empowerment. Sexual Division of Power: 4) HIV Risk and 5) Relationship Dynamics. Cathexis: Affective Influences/Social Norms: 6) HIV/AIDS Knowledge and 7) Prevention and Testing. Importantly, participants were concerned about partner fidelity when visiting the Dominican Republic, as the country accounts for the second highest HIV rates in the Caribbean. Conclusions: Our results confirm previous findings about perceptions of HIV risk and provide additional insight into aging-related aspects of HIV risk for Latino women midlife and older.
... 23 HIV has been associated with accelerated ageing. 24 This phenomenon is thought to be partly due to chronic inflammation and immune activation caused by HIV infection. These factors contribute to increased rates of cardiovascular disease, osteoporosis, kidney disease, liver disease, and certain cancers in people with HIV. 25 HIV can also have significant effects on brain health, including neurocognitive impairment, peripheral neuro pathy, and Alzheimer's disease. ...
... Accordingly, chronic HCV infected individuals are frequently afflicted with one or more morbidities, such as liver cirrhosis, HIV infection, and/or age-associated immune impairment. Compared to one disease entity, the combination of two or more morbidities in one individual results in a greater magnitude of immunological impairment and, therefore, warrants more careful consideration when designing therapeutic strategies for this population [10][11][12][13][14]. ...
Article
Full-text available
Chronic hepatitis C virus (HCV) infection is associated with naïve CD4+ T cell lymphopenia and long-standing/persistent elevation of cellular and soluble immune activation parameters, the latter heightened in the setting of HIV co-infection. The underlying mechanisms are not completely understood. However, we recently reported that accelerated peripheral cell death may contribute to naïve CD4+ T cell loss and that mechanistic relationships between monocyte activation, T cell activation, and soluble inflammatory mediators may also contribute. Chronic HCV infection can be cured by direct-acting anti-viral (DAA) therapy, and success is defined as sustained virological response (SVR, undetectable HCV RNA (ribonucleic acid) at 12 weeks after DAA treatment completion). However, there is no general consensus on the short-term and long-term immunological outcomes of DAA therapy. Here, we consolidate previous reports on the partial normalization of naïve CD4+ lymphopenia and T cell immune activation and the apparent irreversibility of monocyte activation following DAA therapy in HCV infected and HCV/HIV co-infected individuals. Further, advanced age and cirrhosis are associated with delayed or abrogation of immune reconstitution after DAA therapy, an indication that non-viral factors also likely contribute to host immune dysregulation in HCV infection.
... The typical clinical presentation of an older HIV patients display a more sluggish humoral immunity, a lower CD4 (+) T cell counts, higher viral load, more symptoms at diagnosis, and a tendency for rapid progression with more co-morbid conditions, indicating that the complex aging process that synergistically plays part between HIV infections, immune system, among elderly with HIV infection [40][41][42][43][44][45]. The pharmacological management of HARRTS regiments in older HIV infected patients continue to requires the careful risk assessment for class selection, timing and potential drug-drug interactions as we continue our search on ways enhancing tolerability and individualization of therapeutic treatment options with d4T+3TC+, NNRTI, and AZT+3TC+NNRTI therapeutics [46][47][48][49][50][51][52][53]. Finally, the treatment and care of often frail with a less robust immune system older HIV-infected patients remain compounded by a synergism between aging, HIV infection and ongoing immunodeficiency that contribute to age-associated end-organ dysfunction and non-AIDSdefining co-morbidities (i.e.dyslipidemia, metabolic syndrome, osteoporosis, and neurocognitive disorder) therefore, making more longitudinal investigational research in aging a priority for developing in age sensitive age-specific guidelines in order to reduce burden of chronic care the resource challenged health care settings [54][55][56][57][58][59][60][61][62][63][64]. ...
Preprint
Full-text available
Abstract The movement disorders are chronic neurodegenerative group of diseases such as; Parkinson’s disease, Huntington’s disease, tremor, and dystonia which are increasingly becoming more prevalent. The establishing of their classification, accurate diagnosis and clinical management can be a difficult and lengthy process in part due to their complex natural history and heterogeneous presentations. Unfortunately, the under recognition and under diagnosed movement disorders are also common partly as a result of the clinicians inadequate understanding of disease phenomenology and phonotypical variability and challenges related inability to differentiate functional and secondary movement disorders from organic movement disorders. In addition to challenges of clinicians knowledge and attitudes and the lack of coordination and obstacles in the dynamics of care pathways that link the primary care services with the higher specialists care can significant have adverse impacts on of quality of the chronic neurological care delivery needed among the vulnerable patient population. A unique group of conditions that are commonly encountered by the community healthcare providers are the secondary movement disorders or the mimickers of movement disorders that include the drug-induced and the systemic or metabolically-induced movement disorders which present both as a diagnostic and management challenge and opportunity for the general practitioners and community neurologist alike. Keywords: Movement disorders; Chronic neurological care; Clinicians knowledge and attitudes; Healthcare delivery primary care; Secondary movement disorders; Drug and metabolically -induced movement disorders
... HIV treatment has lengthened life, and dramatically decreased susceptibility to opportunistic infections. 1,2 Immune restoration with highly active antiretroviral therapy (HAART) 3,4 has contributed tremendously to these outcomes. However, long-term survivorship of people living with HIV (PLWH) is characterized by greater numbers of comorbidities, not simply explained by the decline in AIDS-related mortality. ...
... However, immune recovery is highly variable despite consistent viral suppression, and up to 30% of people living with HIV (PLH) under suppressive treatment fail to reach CD4 + T cell counts >500/µL [2] Determinants of low immune recovery are still incompletely understood. A number of factors have been associated with this condition, such as advanced age [3,4], higher pre-treatment HIV viral load (HIV-VL) [2,5] , lower pretreatment CD4 + T cell counts [5][6][7] , thymus and bone marrow malfunction [8] , persistent immune activation and increased circulating lipopolysaccharide levels [9][10][11] . ...
Article
Background: HIV infection leads to depletion of intestinal CD4+ T cells, mucosal barrier dysfunction, increased gut permeability and microbial translocation even among patients on suppressive ART. Previous studies suggest probiotics may help restore intestinal function. Methods: In this double-blind, placebo-controlled pilot study, we enrolled HIV-infected patients on suppressive ART with poor CD4+ recovery to address the effect of daily oral use of Lactobacillus casei Shirota (LcS) on CD4+ T cell count and CD4/CD8 ratio at 6 and 12 weeks after treatment initiation; immune activation and intestinal microbiome composition were addressed as secondary outcomes. Results: From Jan/2015 to Jul/2016, 48 patients were randomized (1:1) to active intervention or placebo. Groups had comparable demographic and clinical characteristics; only CD4+ T cell nadir was statistically different between groups. All participants were virologically-suppressed under ART. At week 6, the increment in CD4+ T cell count was 17 cells/mm3 (interquartile range [IQR] -33 to 74) in the active intervention arm and 4 cells/mm3 (IQR -43 to 51) in the placebo arm (p = 0.291); at week 12, the change in CD4+ T cell count was 8 cells/mm3 (IQR -30 to 70) in the active arm and 10 cells/mm3 (IQR -50 to 33) among participants allocated to placebo (p = 0.495). Median change in CD4/CD8 ratio at week 6 compared to baseline was 0 (IQR -0.04 to 0.05) in the active intervention arm and -0.01 in the placebo arm (IQR -0.06 to 0.03; p = 0.671). At week 12, the change in CD4/CD8 ratio was higher in the active product group compared to placebo (respectively 0.07 and 0.01), but this difference failed to reach statistical significance (p = 0.171). We found no significant effects of LcS on immune activation markers, CD4+ and CD8+ subpopulations, sCD14 levels or NK cells at week 12. Finally, we found no statistically significant differences between groups in the change of enteric microbiome at week 12. Conclusions: In this pilot study, we found no statistically significant effect of LcS probiotic on CD4+ T cell counts, CD4/CD8 ratio, immune activation or intestinal microbiome among HIV-infected patients on suppressive ART with poor CD4+ recovery.
... There is strong immune and HIV basic science rationale to the parallels between how aging and chronic-treated HIV infection affects the immune system [36,37,113,114]. In chronic PLHIV, premature aging is probably caused through an integrated pathway of aetiologies converging towards chronic immune activation [115], which is worse as a function of HIV-related immune compromise. ...
Article
Full-text available
Purpose The purpose of this review is to provide an overview of established risk factors for all-type dementia and results of interventions on dementia modifiable risk factors, all with relevance to aging people living with HIV (PLHIV). Methods Narrative literature review. Results Our review identifies a high prevalence of risk factors for dementia in the global HIV population that is entering dementia age range (60 +), in relation to both traditional and HIV-specific risk factors. This includes age (HIV-related premature aging and possibly HIV-related accelerated brain aging and cerebrovascular injury), HIV-related and non-HIV-related cardiovascular diseases burden with related-vascular brain damage, HIV-associated neurocognitive disorders, high mental health burden, low educational/socio-economic status, historical immune compromise, and persistent immune activation with consequent augmented immune senescence. Our review highlights that the results of interventions on all-type dementia modifiable factors show discrepancies between positive observational study results and inconclusive clinical trials. The main reasons for such discrepancies relate to the preventative framework that complex interventions’ trials have difficulty to emulate and the suboptimal measurement of cognitive change. Multi-domain intervention trials are now advocated to concomitantly tackle complex age-related comorbid profiles. Conclusions The burden of dementia risk in aging PLHIV is higher than that in the general population, particularly in the most vulnerable clusters. Epidemiological studies are urgently needed to provide accurate estimates. Lessons from interventions trials in all-type dementia on modifiable factors need to be carefully considered for enhancing trials’ potential in aging PLHIV. A comprehensive and preventative neurogeriatric healthcare response linked with HIV communities and dementia associations should be urgently put in place.
Chapter
In den Ländern des globalen Nordens ist in den letzten zwei Jahrzehnten eine Entwicklung in den Blick geraten, die lange undenkbar erschien: Aufgrund der medizinischen Normalisierung der HIV-Infektion als gut behandelbare, chronische Erkrankung ist das durchschnittliche Lebensalter der Menschen mit HIV und Aids deutlich gestiegen, und der Anteil älterer Menschen unter den HIV-Infizierten wird immer größer. Der Beitrag gibt einen Überblick über den Forschungsstand zum Altern mit HIV/AIDS und zur HIV-bezogenen Stigmatisierung und stellt ausgewählte quantitative und qualitative Ergeb- nisse der Studie 50plusHIV vor. Die Studie hat erstmals für den deutschen Kontext die psychosozialen Herausforderungen des Älterwerdens von HIV- Positiven untersucht und eine empirisch fundierte und genaue Beschreibungder Lebenswirklichkeit von älteren Menschen mit HIV und Aids vorgelegt. Ein besonderer Fokus des Beitrags liegt auf den nach wie vor weitreichenden Erfahrungen und den tiefgreifenden Folgen HIV-bezogener Stigmatisierung. Aufgezeigt werden daraus resultierende Implikationen für die gesundheitliche und pflegerische Versorgung, für Selbsthilfe und Politik und für Menschen mit HIV und Aids selbst.
Chapter
Full-text available
In den Ländern des globalen Nordens ist in den letzten zwei Jahrzehnten eine Entwicklung in den Blick geraten, die lange undenkbar erschien: Aufgrund der medizinischen Normalisierung der HIV-Infektion als gut behandelbare, chronische Erkrankung ist das durchschnittliche Lebensalter der Menschen mit HIV und Aids deutlich gestiegen, und der Anteil älterer Menschen unter den HIV-Infizierten wird immer größer. Der Beitrag gibt einen Überblick über den Forschungsstand zum Altern mit HIV/AIDS und zur HIV-bezogenen Stigmatisierung und stellt ausgewählte quantitative und qualitative Ergebnisse der Studie 50plusHIV vor. Die Studie hat erstmals für den deutschen Kontext die psychosozialen Herausforderungen des Älterwerdens von HIV-Positiven untersucht und eine empirisch fundierte und genaue Beschreibung der Lebenswirklichkeit von älteren Menschen mit HIV und Aids vorgelegt. Ein besonderer Fokus des Beitrags liegt auf den nach wie vor weitreichenden Erfahrungen und den tiefgreifenden Folgen HIV-bezogener Stigmatisierung. Aufgezeigt werden daraus resultierende Implikationen für die gesundheitliche und pflegerische Versorgung, für Selbsthilfe und Politik und für Menschen mit HIV und Aids selbst.
Preprint
Full-text available
HIV long-term survivorship is characterized by higher rates of comorbidities compared to uninfected groups. Aging with HIV involves complex interactions of factors (e.g., Individual Characteristics, Infections) that result in a 20% increase in comorbidity risk. With over half of the 1.1 million people living with HIV in the US age 50 and over, the need exists to further understand this interplay and differences in aging-related outcomes. Electronic health record data was analyzed for HIV infected (N=208) and uninfected (N=208) adult inpatients, propensity score matched by age and gender. Diagnostic codes were extracted that comprise the factors of Individual Characteristics, High Risk Behaviors, Chronic Conditions, Mental Health Conditions and Infections. Identified codes were assessed for their contributions to medical resource utilization, based on Charlson Comorbidity scores. Significant contributors to high Charslon scores for HIV infected patients were age (β=0.116; [95% CI 0.077, 0.155]) and admission frequency (β=0.159; [95% CI 0.114, 0.205]) in addition to the comorbidities of acute kidney failure (β=3.27; [95% CI 1.76, 4.78]), hypertension (β= -1.77; [95% CI -2.99, -0.551]). Significant contributors for HIV uninfected patients were age (β=0.110; [95% CI 0.087, 0.133]), length of hospital stay (β=0.006; [95% CI 0.003, 0.009]), acute kidney failure (β=1.556; [95% CI 0.611, 2.50]), heart failure (β= 1.713; [95% CI 0.717, 2.71]), and diabetes mellitus II (β= 1.385; [95% CI 0.634, 2.14]). Our findings enhance the understanding of the contributions to medical resource utilization based on HIV status and can inform intervention efficacy for improved HIV aging outcomes.
Article
Full-text available
Background Predictors of virological response to highly active antiretroviral therapy (HAART) have never been systematically evaluated in a large continental multicenter cohort of unselected human immunodeficiency virus (HIV)–infected people.Objective To determine the factors related to achieving and maintaining undetectable plasma HIV-1 RNA levels among HIV-1–infected patients first starting protease inhibitor– or nonnucleoside retrotranscriptase inhibitor–containing HAART in Europe.Design Prospective multicenter cohort study.Setting Fifty-two clinical centers in 17 European countries included in the EuroSIDA Study Group, from August 1996 to April 1999.Patients A total of 1469 HIV-positive patients first starting HAART recruited from an unselected cohort of more than 7300 HIV-positive patients.Main Outcome Measure Detection of factors related to virological success after first starting HAART (baseline) and ensuing failure by standard survival techniques, including Kaplan-Meier techniques and Cox proportional hazards models. All analyses were intention to treat.Results Most patients (80%) achieved plasma HIV-1 RNA levels of less than 500 copies/mL during follow-up (60.4% at 6 months from the onset of HAART). Patients with higher baseline HIV-1 RNA levels (relative hazard [RH], 0.76 per log higher; 95% confidence interval [CI], 0.69-0.84; P<.001) and those taking saquinavir mesylate hard gel as a single protease inhibitor (RH, 0.62; 95% CI, 0.47-0.82; P<.001) were less likely to reach undetectable HIV-1 RNA levels. Conversely, higher CD4+ lymphocyte counts (RH per 50% higher, 1.09; 95% CI, 1.02-1.16; P=.008) and the initiation of 3 or more new antiretroviral drugs (RH, 1.29; 95% CI, 1.03-1.61; P=.02) were independent predictors of higher success. Once success was achieved, HIV-1 RNA levels rebounded in more than one third of all patients during follow-up (24% at 6 months). Antiretroviral-naive patients (RH, 0.50; 95% CI, 0.29-0.87; P=.01), older patients (RH, 0.86 per year older; 95% CI, 0.75-0.99; P=.04), and those starting a protease inhibitor other than saquinavir hard gel (RH, 0.66; 95% CI, 0.44-0.98; P=.04) were at decreased hazard for virological failure. Higher baseline HIV-1 RNA level (RH, 1.18 per log higher; 95% CI, 0.99-1.40; P=.06) and a longer time to achieve virological success (RH per 12 months, 1.53; 95% CI, 0.99-2.38; P=.06) were marginally significant predictors of a decreased hazard of ensuing virological failure.Conclusions HAART is associated with a favorable virological response if started when the baseline HIV-1 RNA level is low, if at least 2 new nucleoside retrotranscriptase inhibitors are added, and if standard doses of saquinavir hard gel capsule are avoided as a single protease inhibitor. Older patients are more likely to achieve virological success. Thereafter, the higher durability of virological response is predicted by an antiretroviral-naive status and by the use of specific regimens. Lower baseline HIV-1 RNA levels and rapid maximal viral suppression seem to be other important factors in the durability of virological response. Figures in this Article AN APPROPRIATE comprehension of the factors that affect the virological response to antiretroviral treatments is warranted to improve the clinical treatment of human immunodeficiency virus (HIV)–infected patients. It is important, as well, to expand the rational basis for the accurate design of effective therapies and to reduce the duration and complexity of clinical trials. Formerly, the natural history of HIV infection was invariably unidirectional, progressively leading to acquired immunodeficiency syndrome (AIDS) and death, and the efficacy of therapy was determined by its ability to delay this fatal progression. Therefore, predictors of clinical progression have been extensively studied and precisely determined in HIV-infected patients.1- 8 Today, the clinical prognosis of HIV infection has radically changed because of the widespread use of highly active antiretroviral therapy (HAART), including protease inhibitors (PIs).9- 11 Partly because these studies link plasma HIV-1 RNA levels with risk of clinical progression, the positivist goal of antiretroviral therapy is now to reduce and maintain HIV-1 RNA levels below the lowest detectable.12 However, predictors of short-term virological response to treatments should be considered apart from predictors of clinical progression. Different pathogenic mechanisms probably determine viral dynamic responses to treatments and clinical disease progression. Low baseline CD4+ T-cell counts undoubtedly determine the risk of death in HIV-infected people.13 But do they affect the initial decay of plasma HIV-1 RNA levels after the start of an intense triple therapy containing PIs or nonnucleoside reverse transcriptase inhibitors (NNRTIs)? In addition, it is uncertain whether short-term plasma HIV-1 RNA responses reflect the long-term clinical prognosis of HIV-infected patients.14 Results of recent studies15 have shown that the minimum HIV-1 RNA levels achievable are required to obtain durable virological responses. Durability of virological response should be understood as the major goal to improve the clinical prognosis of patients. However, discrepant virological and immunologic responses to antiretroviral regimens16- 17 indicate that not only plasma HIV-1 RNA level plays a role in this clinical prognostic improvement. Because an appropriate definition for treatment success and failure abides for consensus,16 authors can usually define failure of therapy in terms of lack of "sufficient" suppression of viral replication. It is important to assess the impact of HAART in unselected HIV-infected patients because the efficacy of therapy in daily clinical practice clearly differs from the high rates of success seen in clinical trials.18 The main purpose of the present study is to properly define predictors of virological success in a large European cohort of 1469 unselected HIV-positive patients who start a HAART approach for the first time and predictors of eventual failure once success has been accomplished. This is the first report assessing this issue in a large prospective multinational multicenter study that includes all major risk groups.
Article
Full-text available
Over the last years, the mean age of subjects with HIV infection and AIDS is increasing. Moreover, some epidemiological and clinical differences between younger and older HIV-infected individuals have been observed. However, since introduction of HAART therapy, there are controversial results regarding their response to HAART. The aim of the present study is to evaluate epidemiological and clinical features, response to HAART, and survival in elderly HIV-infected patients with regard to younger HIV-infected patients. A prospective cohort study (1998-2003) was performed on patients from Sabadell Hospital, in Northeast of Spain. The cohort includes newly attended HIV-infected patients since January 1, 1998. For the purpose of this analysis, data was censured at December 31, 2003. Taking into account age at time of diagnosis, it was considered 36 HIV-positive people aged 50 years or more (Group 1, G1) and 419 HIV-positive people aged 13-40 years (Group 2, G2). Epidemiological, clinical, biological and therapy data are recorded. Statistical analysis was performed using Chi-squared test and Fisher exact test, Mann-Whitney U test, Kaplan-Meier, Log Rank test, and Two-Way ANOVA from random factors. G1 showed higher proportion of men than G2. The most common risk factors in G1 were heterosexual transmission (P = 0.01) and having sex with men or women (P < 0.001). G1 and G2 show parallel profiles through the time regarding immunological response (P = 0.989) and virological response (P = 0.074). However, older people showed lower CD4 cell counts at first clinic visit (P < 0.001) and, eventually, they did not achieve the same counts as G2. G1 presented faster progression to AIDS (P < 0.001) and shorter survival (P < 0.001). Older patients have different epidemiological features. Their immunological and virological responses are good. However, older patients do not achieve the same CD4 cell counts likely due to they have lower counts at first clinic visit. Thus, it is essential physicians know older HIV-infected patients features to consider the possibility of HIV infection in these patients with the aim of treatment would not be delayed.
Article
There has recently been a resurgence of interest in the gastrointestinal pathology observed in patients infected with HIV. The gastrointestinal tract is a major site of HIV replication, which results in massive depletion of lamina propria CD4 T cells during acute infection. Highly active antiretroviral therapy leads to incomplete suppression of viral replication and substantially delayed and only partial restoration of gastrointestinal CD4 T cells. The gastrointestinal pathology associated with HIV infection comprises significant enteropathy with increased levels of inflammation and decreased levels of mucosal repair and regeneration. Assessment of gut mucosal immune system has provided novel directions for therapeutic interventions that modify the consequences of acute HIV infection.
Article
Deterioration of the thymus gland during aging is accompanied by a reduction in plasma GH. Here we report gross and microscopic results from 24-month-old Wistar-Furth rats treated with rat GH derived from syngeneic GH3 cells or with recombinant human GH. Histological evaluation of aged rats treated with either rat or human GH displayed clear morphologic evidence of thymic regeneration, reconstitution of hematopoietic cells in the bone marrow, and multiorgan extramedullary hematopoiesis. Quantitative evaluation of formalin-fixed, hematoxylin and eosin-stained sections of bone marrow from aged rats revealed at least a 50% reduction in the number hematopoietic bone marrow cells, compared with that of young 3-month-old rats. This age-associated decline in bone marrow leukocytes, as well as the increase in bone marrow adipocytes, was significantly reversed by in vivo treatment with GH. Restoration of bone marrow cellularity was caused primarily by erythrocytic and granulocytic cells, but all cell lineages were represented and their proportions were similar to those in aged control rats. On a per-cell basis, GH treatment in vivo significantly increased the number of in vitro myeloid colony forming units in both bone marrow and spleen. Morphological evidence of enhanced extramedullary hematopoiesis was observed in the spleen, liver, and adrenal glands from animals treated with GH. These results confirm that GH prevents thymic aging. Furthermore, these data significantly extend earlier findings by establishing that GH dramatically promotes reconstitution of another primary hematopoietic tissue by reversing the accumulation of bone marrow adipocytes and by restoring the number of bone marrow myeloid cells of both the erythrocytic and granulocytic lineages.
Article
Hepatitis C virus (HCV) has a high propensity for persistence. To better define the immunologic determinants of HCV clearance and persistence, we examined the circulating HCV-specific T-cell frequency, repertoire, and cytokine phenotype ex vivo in 24 HCV seropositive subjects (12 chronic, 12 recovered), using 361 overlapping peptides in 36 antigenic pools that span the entire HCV core, NS3-NS5. Consistent with T-cell-mediated control of HCV, the overall HCV-specific type-1 T-cell response was significantly greater in average frequency (0.24% vs. 0.04% circulating lymphocytes, P = .001) and scope (14/36 vs. 4/36 pools, P = .002) among the recovered than the chronic subjects, and the T-cell response correlated inversely with HCV titer among the chronic subjects (R = −0.51, P = .049). Although highly antigenic regions were identified throughout the HCV genome, there was no apparent difference in the overall HCV-specific T-cell repertoire or type-1/type-2 cytokine profile relative to outcome. Notably, HCV persistence was associated with a reversible CD4-mediated suppression of HCV-specific CD8 T cells and with higher frequency of CD4+CD25+ regulatory T cells (7.3% chronic vs. 2.5% recovered, P = .002) that could directly suppress HCV-specific type-1 CD8 T cells ex vivo. In conclusion, we found that HCV persistence is associated with a global quantitative and functional suppression of HCV-specific T cells but not differential antigenic hierarchy or cytokine phenotype relative to HCV clearance. The high frequency of CD4+CD25+ regulatory T cells and their suppression of HCV-specific CD8 T cells ex vivo suggests a novel role for regulatory T cells in HCV persistence. (Hepatology 2003;38:1437-1448.)
Article
Background: Although HIV infection is more prevalent in people younger than age 45 years, a substantial number of infections occur in older persons. Recent guidelines recommend HIV screening in patients age 13 to 64 years. The cost-effectiveness of HIV screening in patients age 55 to 75 years is uncertain. Objective: To examine the costs and benefits of HIV screening in patients age 55 to 75 years. Design: Markov model. Data Sources: Derived from the literature. Target Population: Patients age 55 to 75 years with unknown HIV status. Time Horizon: Lifetime. Perspective: Societal. Intervention: HIV screening program for patients age 55 to 75 years compared with current practice. Outcome Measures: Life-years, quality-adjusted life-years (QALYs), costs, and incremental cost-effectiveness. Results of Base-Case Analysis: For a 65-year-old patient, HIV screening using traditional counseling costs $55 440 per QALY compared with current practice when the prevalence of HIV was 0.5% and the patient did not have a sexual partner at risk. In sexually active patients, the incremental cost-effectiveness ratio was $30020 per QALY. At a prevalence of 0.1%, HIV screening cost less than $60000 per QALY for patients younger than age 75 years with a partner at risk if less costly streamlined counseling is used. Results of Sensitivity Analysis: Cost-effectiveness of HIV screening depended on HIV prevalence, age of the patient, counseling costs, and whether the patient was sexually active. Sensitivity analyses with other variables did not change the results substantially. Limitations: The effects of age on the toxicity and efficacy of highly active antiretroviral therapy and death from AIDS were uncertain. Sensitivity analyses exploring these variables did not qualitatively affect the results. Conclusion: If the tested population has an HIV prevalence of 0.1 % or greater, HIV screening in persons from age 55 to 75 years reaches conventional levels of cost-effectiveness when counseling is streamlined and if the screened patient has a partner at risk. Screening patients with advanced age for HIV is economically attractive in many circumstances.
Article
Objective. —To determine if the long-term incidence of the acquired immunodeficiency syndrome (AIDS) is related to human immunodeficiency virus type 1 (HIV-1) RNA levels measured early in HIV-1 infection.Design. —Epidemiologic cohort study.Setting. —Five hemophilia treatment centers in the United States.Subjects. —A total of 165 subjects with hemophilia and HIV-1 infection (age at HIV-1 seroconversion, 1-66 years) followed from 1979 to 1995.Methods. —The HIV-1 RNA level was measured by polymerase chain reaction over a range of 200 to 1 million or more HIV-1 RNA copies/mL in archived serum specimens collected 12 to 36 months (median, 27 months) after the estimated date of HIV-1 seroconversion. Kaplan-Meier methods were used to examine the risk of AIDS and proportional hazards models were used to estimate relative hazards.Results. —The HIV-1 RNA values were similar in subjects younger than 17 years at seroconversion (median, 5214 copies/mL) and those 18 to 34 years old (median, 4693 copies/mL), but higher in those 35 years or older (median, 12 069 copies/mL) (P=.02 compared with each younger group). At 10 years after seroconversion, the proportions of subjects with AIDS were 72% among subjects with 100 000 or more HIV-1 RNA copies/mL measured 12 to 36 months after HIV-1 seroconversion (n=9), 52% among subjects with 10 000 to 99 999 copies/mL (n=55), 22% among subjects with 1000 to 9999 copies/mL (n=82), and 0% among subjects with fewer than 1000 copies/mL (n=19) (P<.001). The age-adjusted relative hazard for AIDS for subjects with 10 000 or more copies/mL was 14.3 (95% confidence interval, 1.9-105.6) compared with subjects with fewer than 1000 copies/mL.Conclusions. —The HIV-1 RNA level during early chronic HIV-1 infection is a strong, age-independent predictor of clinical outcome; low levels define persons with a high probability of long-term AIDS-free survival.
Article
Objective: To analyse the influence of age at seroconversion and sexual exposure group on the progression of HIV disease. Design: This multicentre prospective cohort study involved 443 subjects whose date of HIV infection was known to within +/-1 year. Individuals whose sexual behaviour was exclusively heterosexual after HIV infection constituted the heterosexual group (n = 131). AIDS-free survival was compared with that of men (n = 312) infected through homosexual sex and who continued homosexual activity after HIV infection. They constituted the homosexual group. Methods: The end-point was the onset of an AIDS-defining illness listed in the 1987 revised Centers for Disease Control and Prevention (CDC) criteria. Using the Kaplan-Meier method, AIDS-free survival curves were plotted for three age categories ( < 20, 20-39, >40 years). A Cox model was used to quantify the effect of age and to assess the influence of exposure group on AIDS onset after adjustment for age. Because of the high incidence of Kaposi's sarcoma (KS) among homosexual men, a disease that can be an early AIDS-defining illness, multivariate analysis was performed with and without consideration of the occurrence of KS. Results: Patients aged >=40 years at seroconversion progressed more rapidly to AIDS than younger patients (P < 0.006). When age was fitted as a continuous variable and adjusted for exposure group, the relative risk of developing AIDS by any time after seroconversion was 1.34 for a 10-year increase difference [P= 0.03; 95% confidence interval (Cl), 1.03-1.77]. After adjustment for age, the relative risk of developing AIDS (CDC criteria) was 2.42 (P = 0.008; 95% Cl, 1.18-4.97) among the homosexual men (AIDS cases, n = 56). All cases of KS (n = 19) involved the homosexual group. Excluding KS as a first manifestation of AIDS, homosexual or bisexual subjects had a risk of AIDS of 1.92 (P =0.07; 95% Cl, 0.92-4.03) compared with heterosexual subjects. Conclusions: The risk of AIDS increases with age at seroconversion. The more rapid progression towards AIDS in the homosexual group than in the heterosexual group persisted after adjustment for age. Further studies are required to determine the possible role of repeated exposure to HIV or other pathogens acquired sexually. (C) Lippincott-Raven Publishers.