Contributing Authors
Marcus Altfeld – Boston
Georg Behrens – Melbourne
Mario Ostrowski – Toronto
Andrea Rubbert – Köln
Christiane Schieferstein – Frankfurt
Reinhold E. Schmidt – Hannover
Bruce D. Walker – Boston
Eva Wolf – München
HIV Medicine 2003
www.HIVMedicine.com
Edited by
Christian Hoffmann
and
Bernd Sebastian Kamps
Flying Publisher
3
Editors
Christian Hoffmann, M.D.
University of Schleswig Holstein
Infectious Diseases Outpatient Clinic Kiel
Chemnitzstr. 33
24116 Kiel, Germany
Fax: + 49 431 1697 1273
www.HIVMedicine.com
www.SARSReference.com
Bernd Sebastian Kamps, M.D.
Flying Publisher
Rue Saulnier
75009 Paris
France
www.FlyingPublisher.com
HIV Medicine is an ever-changing field. The editors and authors of HIV
Medicine 2003 have made every effort to provide information that is accurate
and complete as of the date of publication. However, in view of the rapid
changes occurring in medical science, HIV prevention and policy, as well as the
possibility of human error, this site may contain technical inaccuracies,
typographical or other errors. Readers are advised to check the product
information currently provided by the manufacturer of each drug to be
administered to verify the recommended dose, the method and duration of
administration, and contraindications. It is the responsibility of the treating
physician who relies on experience and knowledge about the patient to
determine dosages and the best treatment for the patient. The information
contained herein is provided "as is" and without warranty of any kind. The
contributors to this site, including Flying Publisher and AmedeoGroup, disclaim
responsibility for any errors or omissions or for results obtained from the use of
information contained herein.
© 2003 by Flying Publisher – Paris, Cagliari, Wuppertal, Sevilla
Assistant Editors: Nyasha Bakare, Dianne Lydtin
Design: Attilio Baghino, www.a4w.it
ISBN: 3-924774-37-4
4
Preface
Hardly any field of medicine has ever undergone a similar
stormy development to that of the therapy of HIV infection.
Little more than 10 years passed, between the discovery of the
pathogen and the first effective treatment! However, there is
also hardly a field that is subjected to so many fast- and short-
lived trends. What today seems to be statute, is tomorrow often
already surpassed. Nevertheless, therapeutical freedom must not
be confused with freedom of choice. This book presents the
medical knowledge that is actual today: from December 2002 to
January 2003.
Because HIV medicine changes so fast, HIV Medicine 2003
will be updated every year. Additional chapters about
opportunistic infections, malignancies and hepatitis are freely
available at our Web site www.HIVMedicine.com.
Under certain conditions, the editors and the authors of this
book might agree to remove the copyright on HIV Medicine for
all languages except English and German. You could therefore
translate the content of HIV Medicine 2003 into any language
and publish it under your own name – without paying a license
fee. For more details, please see
http://hivmedicine.com/textbook/cr.htm.
Christian Hoffmann and Bernd Sebastian Kamps
Hamburg/Kiel and Paris/Cagliari, January 2003
5
Contributing Authors
Marcus Altfeld, M.D.
Partners AIDS Research Center
Massachusetts General Hospital
Bldg. 149, 13th Street, 5th floor
Charlestown, MA 02129
USA
Tel: 617-724-2461
Fax: 617-726-5411
MAltfeld@partners.org
Georg Behrens, M.D.
Immunolgy Division
Walter And Eliza Hall Institute of Medical Research
PO Royal Melbourne Hospital
Parkville, 3050, Victoria
Australia
Fax: 61-3-9347-0852
behrens@wehi.edu.au
Mario Ostrowski, M.D.
Clinical Sciences Division
University of Toronto
Medical Sciences Building, Rm 6271
1 King's College Circle
Toronto, ON M5S 1A8
Canada
Tel: 416-946-5805
FAX: 416-978-8765
E-mail: m.ostrowski@utoronto.ca
6 Contributing Authors
Andrea Rubbert, M.D.
Medizinische Klinik I
Universitätsklinik Köln
Joseph-Stelzmann Str 9
50924 Köln
Germany
Tel +49 221-478-5623
Fax: +49 221-478-6459
Christiane Schieferstein, M.D.
Medizinische Klinik II
Uniklinikum Frankfurt
Theodor-Stern-Kai 7
60590 Frankfurt am Main
Germany
Tel: +49 69-6301-0
schieferstein@wildmail.com
Reinhold E. Schmidt, M.D., Ph.D.
Abteilung Klinische Immunologie
Zentrum Innere Medizin der
Medizinischen Hochschule Hannover
Carl-Neuberg-Straße 1
30625 Hannover
Germany
Tel: +49 511-532-6656
Fax: +49 511-532-9067
Schmidt.Reinhold.Ernst@MH-Hannover.de
Contributing Authors 7
Bruce D. Walker, M.D., Ph.D.
Partners AIDS Research Center
Massachusetts General Hospital
Bldg. 149, 13th Street, 5th floor
Charlestown, MA 02129
USA
Tel: 001 617-724-8332
Fax: 001 617-726-4691
bwalker@helix.mgh.harvard.edu
Eva Wolf, Dipl. Phys. Univ., M.P.H.
MUC Research GmbH
Karlsplatz 8
80335 München
Germany
Tel: +49 89 - 558 70 30
Fax: +49 89 - 550 39 41
Contents 11
Hoffmann, Kamps, et al.
Contents
Chapter 1: Pathogenesis of HIV-1 Infection 15
Introduction 15
The Structure of HIV-1 17
The HIV Replication Cycle 21
HIV and the Immune System 31
References 41
Chapter 2: Acute HIV-1 Infection 47
Introduction 47
Signs and Symptoms 47
Diagnosis 48
Treatment 50
References 51
Chapter 3: HIV Therapy 2003 53
1. Perspective 53
2. Overview of Antiretroviral Drugs 61
3. Goals and Principles of Therapy 120
4. When to Start HAART 146
5. How to Start with HAART 163
6. When to Change HAART 194
7. How to Change HAART 199
8. Salvage Therapy 204
9. When to Stop HAART 215
10. Monitoring 230
Chapter 4: Management of Side Effects 247
Gastrointestinal Side Effects 248
CNS Disorders 249
Peripheral Polyneuropathy 250
12 Contents
HIV Medicine 2003 – www.HIVMedicine.com
Renal Problems 250
Hepatotoxicity 251
Anemia, Leukopenia 252
Allergies 253
Pancreatitis 256
Avascular Necrosis 257
Osteopenia/Osteoporosis 258
Lipodystrophy, Dyslipidemia 259
Hyperglycemia, Diabetes mellitus 259
Increased Bleeding Episodes in Hemophiliacs 260
References 260
Chapter 5: The Lipodystrophy Syndrome 263
Background 263
Clinical Manifestation 263
HAART, Lipodystrophy Syndrome and
Cardiovascular Risk 267
Pathogenesis 268
Diagnosis 272
Therapy 275
References 280
Chapter 6: HIV Resistance Testing 285
Assays for Resistance Testing 285
Background 287
Interpretation of Genotypic Resistance Profiles 289
Summary 296
References 301
Chapter 7: Drug Profiles 307
Abacavir (ABC) 307
Amprenavir (APV) 309
Contents 13
Hoffmann, Kamps, et al.
Atazanavir (AZV) 312
Combivir
®
314
Delavirdine (DLV) 314
Didanosine (ddI) 316
Efavirenz (EFV) 318
Emtricitabin (FTC) 320
Indinavir (IDV) 321
Lamivudine (3TC) 324
Lopinavir (LPV) 326
Nelfinavir (NFV) 329
Nevirapine (NVP) 330
Ritonavir (RTV) 334
Saquinavir (SQV) 337
Stavudine (d4T) 339
T-20 (Enfuvirtide) 341
Tenofovir (TDF) 343
Tipranavir 345
Trizivir
®
346
Zalcitabine (ddC) 348
Zidovudine (AZT) 349
14 Contents
HIV Medicine 2003 – www.HIVMedicine.com
Introduction 15
Hoffmann, Kamps, et al.
Chapter 1:
Pathogenesis of HIV-1 Infection
Andrea Rubbert and Mario Ostrowski
Introduction
Since the initial description of the human immunodeficiency
virus type I (HIV-1) in 1983 (1,2) and HIV-2 in 1986 (3), these
two viruses have been identified for almost 20 years as the pri-
mary cause of the acquired immunodeficiency syndrome
(AIDS). As HIV-1 is the major cause of AIDS in the world to-
day, our discussion will be primarily limited to HIV-1 infection.
Worldwide, the number of HIV-1 infected persons exceeds 40
million, the majority of whom live in the developing countries
of Asia, sub-Saharan Africa and South America.
The introduction of protease inhibitors and non-nucleoside re-
verse transcriptase inhibitors (NNRTI) to antiretroviral treat-
ment regimens in 1995 began the era of highly active antiretro-
viral therapy (HAART), and resulted in dramatic improvements
in the mortality and morbidity of HIV disease, as determined by
a decreased incidence of opportunistic infections, tumors, and
deaths. Despite all the therapeutic advantages achieved during
the last decade, including the development of highly active
antiretroviral therapy ("HAART"), once an individual has be-
come infected, eradication of the virus still remains impossible.
In addition, new problems relating to the short- and long-term
toxicity of drug treatments and the occurrence of resistance
mutations in both circulating and transmitted viruses are
emerging. In most countries in South East Asia and Africa, the
incidence and prevalence of HIV-1 infection continues to in-
crease and surpass that of Europe and North America. However,
due to the high costs of drug regimens and the lack of a
healthcare infrastructure in these developing countries, the
16 Pathogenesis of HIV-1 Infection
HIV Medicine 2003 – www.HIVMedicine.com
widespread use of HAART is currently not feasible. The further
course of the HIV-1 pandemic therefore mainly depends on
how and to what degree the developing countries with a high
HIV-1 prevalence are able to take advantage of the medical
progress achieved in Europe and North America, and whether
an effective prophylactic vaccine might become available in the
near future.
An understanding of the immunopathogenesis of HIV-1 infec-
tion is a major prerequisite for rationally improving therapeutic
strategies, developing immunotherapeutics and prophylactic
vaccines. As in other virus infections, the individual course of
HIV-1 infection depends on both host and viral factors.
The course of infection with HIV-1 in HIV-infected humans
may vary dramatically, even though the primary infections
arose from the same source (4). In some individuals with a
long-term nonprogressive HIV-1 infection (i.e. lack of decline
in CD4 counts, or chronic infection for at least seven years
without the development of AIDS), a defective virion was
identified (5). Thus, infection with a defective virus, or one
which has a poor capacity to replicate, may prolong the clinical
course of HIV-1 infection. However, in most individuals HIV-1
infection is characterized by a replication competent virus with
a high turn-over of virions produced daily. Host factors may
also deter-mine whether or not an HIV-1 infected individual
will rapidly develop clinically overt immunodeficiency or
whether this individual may belong to the group of long-term
nonprogressors, who represent about 5% of all infected patients.
The identification and characterization of host factors contrib-
uting to the course of HIV infection, including immunological
defense mechanisms and genetic factors, will be crucial for our
under-standing of the immunopathogenesis of HIV infection
and for the development of immunotherapeutic and prophylac-
tic strategies.
The Structure of HIV-1 17
Hoffmann, Kamps, et al.
The Structure of HIV-1
HIV-1 is a retrovirus and belongs to the family of lentiviruses.
Infections with lentiviruses typically show a chronic course of
disease, a long period of clinical latency, persistent viral repli-
cation and involvement of the central nervous system. Visna
infections in sheep, simian immunodeficiency virus infections
(SIV) in monkeys, or feline immunodeficiency virus infections
(FIV) in cats are typical examples of lentivirus infections.
Using electron microscopy, HIV-1 and HIV-2 resemble each
other strikingly. However, they differ with regard to the mo-
lecular weight of their proteins, as well as having differences in
their accessory genes. HIV-2 is genetically more closely related
to the SIV found in sootey mangabeys (SIVsm) rather than
HIV-1 and it is likely that it was introduced into the human
population by monkeys. Both HIV-1 and HIV-2 replicate in
CD4
+
T cells and are regarded as pathogenic in infected per-
sons although the actual immune deficiency may be less severe
in HIV-2 infected individuals.
The Morphologic Structure of HIV-1
HIV-1 viral particles have a diameter of 100 nm and are sur-
rounded by a lipoprotein membrane. Each viral particle contains
72 glycoprotein complexes which are integrated into this lipid
membrane and are each composed of trimers of an external gly-
coprotein gp120 and a transmembrane spanning protein gp41.
The bonding between gp120 and gp41 is only loose and there-
fore gp120 may be shed spontaneously within the local envi-
ronment. Glycoprotein gp120 may also be detected in the serum
(6) as well as within the lymphatic tissue of HIV-infected pa-
tients (7). During the process of budding, the virus may also
incorporate, from the membrane of the host cell into its lipo-
protein layer, different host proteins, such as HLA class I and II
proteins, or adhesion proteins, such as ICAM-1 that may facili-
tate adhesion to other target cells. The matrix protein p17 is an-
chored to the inside of the viral lipoprotein membrane. The p24
18 Pathogenesis of HIV-1 Infection
HIV Medicine 2003 – www.HIVMedicine.com
core antigen contains two copies of HIV-1 RNA. The HIV-1
RNA is part of a protein-nucleic acid complex, which is com-
posed of the nucleoprotein p7 and the reverse transcriptase p66
(RT). The viral particle contains all the enzymatic equipment
that is necessary for replication: a reverse transcriptase (RT), an
integrase p32 and a protease p11 (overview in: 8) (Fig. 1).
Figure 1: Structure of a HIV virion particle. For detailed explanations see text.
The Organization of the Viral Genome
Most replication competent retroviruses depend on three genes:
gag, pol and env : gag means "group-antigen", pol represents
"polymerase" and env is for "envelope" (overview in: 9)
(Fig. 2). The "classical" structural scheme of a retroviral ge-
nome is: 5'LTR-gag-pol-env-LTR 3'. The LTR ("long terminal
The Structure of HIV-1 19
Hoffmann, Kamps, et al.
repeat") regions represent the two end parts of the viral genome
that are connected to the cellular DNA of the host cell after in-
tegration and do not encode for any viral proteins. The gag and
env genes code for the nucleocapsid and the glycoproteins of
the viral membrane; the pol gene codes for the reverse tran-
scriptase and other enzymes. In addition, HIV-1 contains in its
9kB RNA six genes (vif, vpu, vpr, tat, rev and nef) that con-
tribute to its genetic complexity. Nef, vif, vpr and vpu were clas-
sified as accessory genes in the past, as they are not absolutely
required for replication in vitro. However, the regulation and
function of these accessory genes and their proteins have been
studied and characterized in more detail within the last years.
The accessory genes, nef, tat and rev, are all produced early in
the viral replication cycle.
Figure 2: HIV and its genes. For detailed explanations see text.
Tat and rev are regulatory proteins that accumulate within the
nucleus and bind to defined regions of the viral RNA: TAR
20 Pathogenesis of HIV-1 Infection
HIV Medicine 2003 – www.HIVMedicine.com
(transactivation-response elements), found in the LTR; and
RRE (rev response elements), found in the env gene, respec-
tively. The tat protein is a potent transcriptional activator of the
LTR promoter region and is essential for viral replication in
almost all in vitro culture systems. Cyclin T1 is a necessary
cellular cofactor for tat (10). Tat and rev stimulate the tran-
scription of proviral HIV-1-DNA into RNA, promote RNA
elongation, enhance the transportation of HIV-RNA from the
nucleus to the cytoplasm and are essential for translation. Rev is
also a nuclear export factor that is important for switching from
the early expression of regulatory proteins to the structural pro-
teins that are synthesized later.
Nef has been shown to have a number of functions. Nef may
induce downregulation of CD4 (11) and HLA class I and II
molecules (12) from the surface of HIV-1 infected cells, which
may represent an important escape mechanism for the virus to
evade an attack mediated by cytotoxic CD8
+
T cells and to
avoid recognition by CD4
+
T cells. Nef may also interfere with
T cell activation by binding to various proteins that are involved
in intracellular signal transduction pathways (overview in:13).
In SIV-infected rhesus macaques, an intact nef gene was essen-
tial for a high rate of virus production and the progression of
disease. HIV-1 with deletions in nef was identified in a cohort
of Australian long-term non-progressors (5). However, more
recent reports indicate that some of these patients are now de-
veloping signs of disease progression together with a decline of
CD4
+
T cells. Thus, although deletions of the nef gene may
slow viral replication, they cannot always prevent the develop-
ment of AIDS.
Vpr seems to be essential for viral replication in non-dividing
cells such as macrophages. Vpr may stimulate the HIV-LTR in
addition to a variety of cellular and viral promoters. More re-
cently, vpr was shown to be important for the transport of the
viral preintegration complex to the nucleus (overview in: 14)
and may arrest cells in the G2 phase of the cell cycle.
The HIV Replication Cycle 21
Hoffmann, Kamps, et al.
Vpu is important for the virus "budding" process, because mu-
tations in vpu are associated with persistence of the viral parti-
cles at the host cell surface. Vpu is also involved when CD4-
gp160 complexes are degraded within the endoplasmatic re-
ticulum and therefore allows recycling of gp160 for the forma-
tion of new virions (15).
Vif is important for intracellular transport mechanisms of viral
components. Co-localization of vif with vimentin, a protein be-
longing to the cellular cytoskeleton, was demonstrated. Virions
that are deficient in vif may still be transmitted from cell to cell,
but not from a cell free medium. Vif also seems to affect viral
morphogenesis (Overview in: 16).
The HIV Replication Cycle
HIV Entry
CD4 as a primary receptor for HIV
CD4 is a 58 kDa monomeric glycoprotein that can be detected
on the cell surface of about 60% of T-lymphocytes, of T-cell
precursors within the bone marrow and thymus, and on mono-
cytes and macrophages, eosinophils, dendritic cells and micro-
glia cells of the central nervous system. The extracellular do-
main of CD4 on T cells is composed of 370 amino acids; the
hydrophobic transmembrane domain and the cytoplasmic part
of CD4 on T cells consist of 25 and 38 amino acids, respec-
tively. Within the extracellular part of CD4, four regions D1-D4
have been characterized that represent immunoglobulin-like
domains. Residues within the V2 region of CD4 (amino acids
40-55) are important for the binding of gp120 to CD4 and this
region overlaps the part of the CD4 where its natural ligands,
HLA class II molecules, bind.
The identification of the gp120 binding site on the CD4 of
CD4
+
T cells stimulated attempts to use soluble CD4 (sCD4) to
neutralize the circulating virus in patients, with the goal being
22 Pathogenesis of HIV-1 Infection
HIV Medicine 2003 – www.HIVMedicine.com
the inhibition of viral spread. However it became evident that
even though laboratory viral isolates were easily neutralized by
sCD4, a neutralization of primary, patient-derived isolates had
not been achieved.
In contrast, sCD4 was able to induce conformational changes
within the viral envelope that promoted the infection of target
cells (18).
CD4 attaches to the T cell receptor complex (TCR) on CD4
+
T
cells and binds to the HLA class II molecules on antigen-
presenting cells. The binding of gp120 to CD4 is not only a cru-
cial step for viral entry, but also interferes with intracellular
signal transduction pathways and promotes apoptosis in CD4
+
T
cells (19).
CD4, as a primary and necessary receptor for HIV-1, HIV-2 and
SIV, was already characterized in 1984 (20, 21). However, ex-
periments using non-human cell lines transfected with human
CD4 showed that expression of human CD4 on the cell surface
of a non-human cell line was not sufficient to allow en-try of
HIV. Therefore the existence of additional human co-receptors
necessary for viral entry was postulated. On the other hand,
some laboratory HIV-1 isolates as well as some HIV-2 and SIV
isolates are able to infect human cells independently from CD4.
Interestingly, monoclonal antibodies against CD4 induced con-
formational (CD4I) epitopes to bind to the gp120 of CD4-
independent viruses. This observation suggests that the gp120
of CD4-independent viruses already exposes the regions that
are necessary for co-receptor recognition and binding and there-
fore binding to CD4 is not a prerequisite of entry for these vi-
ruses. CD4-independent viruses are easy to neutralize using the
serum of HIV-infected patients, suggesting that the immune
response selects against CD4-independent viruses (22).
Chemokine receptors as co-receptors for HIV entry
A milestone for the characterization of the early events leading
to HIV-1 entry was an observation by Cocchi and his co-
workers in 1995. CD8
+
T cells from HIV-infected patients are
The HIV Replication Cycle 23
Hoffmann, Kamps, et al.
able to suppress viral replication in co-cultures with HIV-
infected autologous or allogenic CD4
+
T cells and this is inde-
pendent from their cytotoxic activity (23). Cocchi identified the
chemokines MIP-1α, MIP-1ß and Rantes in supernatants from
CD8
+
T cells derived from HIV-infected patients, and was able
to show that these chemokines were able to suppress replication
in a dose-dependent manner of some, but not all viral isolates
tested (24). MIP-1α, MIP-1ß and Rantes are ligands for the
chemokine receptor CCR5, and a few months later several
groups were able to show that CCR5 is a necessary co-receptor
for monocytotropic (M-tropic) HIV-1 isolates (25, 26, 27). A
few weeks earlier, the chemokine receptor CXCR4 (fusin) was
described as being the co-receptor used by T-cell tropic (T-
tropic) HIV-isolates (28). Monocytotropic (M-tropic) HIV-1
isolates are classically those viruses that are most easily propa-
gated in macrophage cultures, are unable to infect T-cell lines
(i.e., immortalized T cells), but are able to easily infect primary
T cells from peripheral blood samples. Conversely, T-cell tropic
HIV-1 isolates have classically been identified as being those
that are easily propagated in T-cell lines, and grow poorly in
macrophages, but are also able to easily infect primary T cells
from peripheral blood samples. Thus, it should be noted that
both M-tropic and T-tropic HIV-1 variants can easily infect
primary human non-immortalized T cells in-vitro. Chemokines
("Chemotactic cytokines") and their receptors have been previ-
ously characterized with regard to their role in promoting the
migration ("chemotaxis") of leukocytes and their proinflamma-
tory activity.
Chemokines are proteins of 68-120 amino acids which depend
on the structure of their common cysteine motif, and which may
be subdivided into C-X-C (α-chemokines), C-C (ß-chemokines)
and C-chemokines. Chemokines typically show a high degree
of structural homology to each other and may share the recep-
tors they bind to. Chemokine receptors belong to the group of
receptors with seven transmembranic regions ("7-
24 Pathogenesis of HIV-1 Infection
HIV Medicine 2003 – www.HIVMedicine.com
transmembrane receptors"), which are intracellularly linked to
G-proteins.
SDF-1 ("stromal cell-derived factor 1") was identified as the
natural ligand of CXCR4 and is able to inhibit the entry of T-
tropic HIV-1 isolates into activated CD4
+
T cells. Rantes
("regulated upon activation T cell expressed and secreted"),
MIP-1α ("macrophage inhibitory protein") and MIP-1ß repre-
sent the natural ligands of CCR5 and are able to inhibit the en-
try of M-tropic HIV-1 isolates into T cells. A schematic model
is depicted in Figure 3: T-tropic HIV-1 isolates mainly infect
activated peripheral blood CD4
+
T cells and cell lines and use
CXCR4 for entry into the CD4
+
-positive target cell. M-tropic
isolates are able to infect CD4
+
T cells, monocytes and macro-
phages and depend on the use of CCR5 and CD4 for viral entry.
The interaction of gp120 and the cellular receptors is now un-
derstood in more detail. Gp120 primarily binds to certain epi-
topes of CD4. Binding to CD4 induces conformational changes
in gp120 that promote a more efficient interaction of the V3
loop of gp120 with its respective co-receptor. Membrane fusion
is dependent on gp120-co-receptor binding. Gp41, as the trans-
membrane part of the envelope glycoprotein gp160, is crucial
for the fusion of the viral and the host cell membrane. Similar to
influenza hemagglutinin, it was postulated that after binding of
gp120 to CD4, a conformational change is also induced in gp41
that allows gp41 to insert its hydrophobic NH
2
-terminal into the
target cell membrane. Gp41 has been compared to a "mouse
trap" and a crystallographic analysis of the ectodomanic struc-
ture of gp41 seems to confirm that hypothesis (29). The identi-
fication of crucial amino acid sequences for this process was
used to synthesize peptides that may bind to gp41 within the
domains that are critical for the induction of conformational
changes and that may inhibit membrane fusion.
The HIV Replication Cycle 25
Hoffmann, Kamps, et al.
Fig 3: Inhibition of virus entry of CCR5-utilizing (monocytotropic) and CXCR4-
utilizing (T-cell tropic) HIV isolates by the natural ligands of the chemokine co-
receptors CCR5 and CXCR4.
T20 is the first of several peptides that bind to gp41 and has
been tested in clinical trials for suppressing viral replication
(30). Currently, T20 is available as a therapeutic option for se-
lected patients. One disadvantage of T20 is that it must be taken
intramuscularly rather than as a pill.
Using transfected cell lines, besides CCR5 and CXCR4, other
chemokine receptors, such as CCR3, CCR2, CCR8, CCR9,
STRL33 ("Bonzo"), Gpr 15 ("Bob"), Gpr 1, APJ and ChemR23,
were identified and shown to be used for entry by certain HIV
isolates (31, 32). APJ may represent a relevant co-receptor
within the central nervous system. Despite this broad spectrum
of potentially available co-receptors, CCR5 and CXCR4 seem
to represent the most relevant co-receptors for HIV-1 in vivo.
26 Pathogenesis of HIV-1 Infection
HIV Medicine 2003 – www.HIVMedicine.com
The importance of CCR5 as the predominant co-receptor for M-
tropic HIV isolates is underscored by another observation. The
majority of individuals with a genetic defect of CCR5 are re-
sistant to infection with HIV-1 (33). In vitro experiments show
that lymphocytes derived from these individuals are resistant to
HIV-1 infection using M-tropic isolates but not to infection
with T-tropic isolates. Lymphocytes from these individuals do
not express CCR5 on their cell surface and genetically they
have a 32 base pair deletion of the CCR5 gene. Worldwide, a
few patients have been identified that have acquired HIV-1 in-
fection despite a homozygous deletion of the CCR5. As ex-
pected, all of them were infected with CXCR4-using HIV-1
isolates (34). In epidemiologic studies, the allelic frequency of
the CCR5 gene deletion is 10-20% among Caucasians, particu-
larly amongst those of Northern European descent. The fre-
quency of a homozygous individual is about 1% in Caucasians
(35). Studies conducted on African or Asian populations, how-
ever, do not find this 32 basepair deletion of the CCR5, sug-
gesting that this mutation arose after the separation of these
races in evolutionary history.
Individuals that are heterozygous for the 32 bp deletion of the
CCR5 show a decreased expression of CCR5 on the cell surface
and are more frequently encountered within cohorts of long-
term non-progressors compared to patients who have a rapid
progression of disease (35).
In addition to the 32bp deletion of the CCR5, other genetic
polymorphisms, with regard to the chemokine receptors
(CCR2) or their promoters (CCR5), were described. Based on
the occurrence of these polymorphisms within defined patient
cohorts, they were associated with a more rapid or a more fa-
vorable course of disease, depending on the particular polymor-
phism (36, 37).
In patients who have a rapid progression of disease (rapid drop
in CD4
+
T cell count), virus isolates that use CXCR4 as a pre-
dominant co-receptor tend to be frequently isolated from their
cells, in comparison to patients with a stable CD4
+
T cell count.
The HIV Replication Cycle 27
Hoffmann, Kamps, et al.
The expression of co-receptors on CD4
+
lymphocytes depends
on their activation level.
CXCR4 is mainly expressed on naive T cells, whereas CCR5 is
present on activated and effector/memory T cells. During the
early course of HIV-1 infection, predominantly M-tropic HIV-1
isolates are detected. Interestingly, M-tropic HIV-1 isolates are
preferentially transmitted regardless of whether or not the "do-
nor" predominantly harbors T-tropic isolates. At present, it re-
mains unclear whether this "in vivo" preference of M-tropic
HIV-1 isolates is determined by selected transportation of M-
tropic isolates by submucosally located dendritic cells or
whether the local cytokine/chemokine milieu favors the repli-
cation of M-tropic viruses. Recent intriguing studies by Cheng
Meyer et al. suggest that M-tropic HIV-1 viruses are more eas-
ily able to 'hide' from the immune system by replicating in
macrophages, in comparison to T-tropic viruses, thus giving
them a survival advantage in the infected individual.
The blockade of CCR5 therefore seems to represent a promising
target for therapeutic intervention. In vitro, monoclonal anti-
bodies to CCR5 (2D7 and others) are able to block the entry of
CCR5-using HIV isolates into CD4
+
T cells and macrophages.
Small molecule inhibitors of CCR5 have been designed and are
currently being tested in clinical trials. In vitro studies, as well
as experiments using SCID mice, however, suggest that block-
ade of CCR5-using isolates may alter their tropism towards in-
creased usage of CXCR4.
Small molecule inhibitors like T22, ALX40-4C or AMD3100
are able to inhibit CXCR4 (59, 60) and are also subject to pre-
clinical and clinical trials. Although the therapeutic use of
chemokine receptor blockers seems promising, a lot of ques-
tions still remain unanswered. Chemokine analogs such as
AOP-Rantes do not only inhibit, but also show agonistic activ-
ity and may not bind to CCR5 exclusively. Using knockout
mice it was demonstrated that the absence of CXCR4 or SDF-1
is associated with severe defects in hematopoiesis and in cere-
bellar development (61). Currently, it remains unclear whether
28 Pathogenesis of HIV-1 Infection
HIV Medicine 2003 – www.HIVMedicine.com
the blockade of CXCR4 in postnatal or adult individuals may
also affect other organ systems.
Fig 4: HIV life cycle within a CD4
+
T cell.
Postfusion Events
HIV-1 entry into quiescent T cells is comparable to HIV-1 entry
into activated T cells, but synthesis of HIV-1 DNA remains in-
complete in quiescent cells (38). The conversion of viral RNA
into proviral DNA, mediated by the viral enzyme reverse tran-
scriptase (RT), occurs in the cytoplasm of the target cell and is a
crucial step within the viral replication cycle (see Fig. 4).
Blockade of the RT by the nucleoside inhibitor, zidovudine,
was the first at-tempt to inhibit viral replication in HIV-1 in-
fected patients. To-day, numerous nucleoside, nucleotide and
non-nucleoside RT inhibitors are available for clinical use and
The HIV Replication Cycle 29
Hoffmann, Kamps, et al.
have broadened the therapeutic arsenal substantially since the
mid-eighties.
Reverse transcription occurs in multiple steps. After binding of
the tRNA primers, synthesis of proviral DNA occurs as a mi-
nus-strand polymerization starting at the PBS ("primer binding
site") up to the 5' repeat region as a short R/U5 DNA. The next
step includes degradation of RNA above the PBS by the viral
enzyme RNAase H and a "template switch" of the R/U5 DNA
with hybridization at the R sequence at the 3' RNA end. Now
the full length polymerization of proviral DNA with degrada-
tion of the tRNA is completed. Reverse transcription results in
double-stranded HIV DNA with LTR regions ("long terminal
repeats") at each end.
HIV-1 enters into quiescent T cells and reverse transcription
may result in the accumulation of proviral, non-integrating
HIV-DNA. However, cellular activation is necessary for inte-
gration of the proviral HIV DNA into the host cell genome after
transportation of the pre-integration complex into the nucleus
(38). Cellular activation may occur in vitro after stimulation
with antigens or mitogens, in vivo activation of the immune
system is observed after antigen contact or vaccination or dur-
ing an opportunistic infection. In addition, evidence is emerging
that HIV-1 gp120 itself may activate the infecting cell to en-
hance integration. Besides monocytes, macrophages and mi-
croglial cells, latently infected quiescent CD4
+
T-cells that
contain non-integrated proviral HIV-DNA represent important
long-living cellular reservoirs for HIV (39). Since natural HIV-
1 infection is characterized by continuing cycles of viral repli-
cation in activated CD4
+
T-cells, viral latency in these resting
CD4
+
T-cells likely represents an accidental phenomenon and is
not likely to be important in the pathogenesis of this disease.
This small reservoir of latent provirus in quiescent CD4
+
T-cells
gains importance, however, in individuals who are treated with
HAART, since the antivirals are unable to affect non-replicating
proviruses and thus the virus will persist in those cells and be
replication competent to supply new rounds of infection, if the
30 Pathogenesis of HIV-1 Infection
HIV Medicine 2003 – www.HIVMedicine.com
drugs are stopped. Thus, the existence of this latent reservoir
has prevented HAART from entirely eradicating the virus from
infected individuals.
Cellular transcription factors like NF-kB may also bind to the
LTR regions. After stimulation with mitogens or cytokines, NF-
kB is translocated into the nucleus where it binds to the HIV-
LTR region, thereby initiating transcription of HIV genes.
Transcription initially results in the early synthesis of regulatory
HIV-1 proteins such as tat or rev. Tat binds to the TAR site
("transactivation response element") at the beginning of the
HIV-1 RNA in the nucleus and stimulates transcription and the
formation of longer RNA transcripts. Rev activates the expres-
sion of structural and enzymatic genes and inhibits the produc-
tion of regulatory proteins, therefore promoting the formation of
mature viral particles. The proteins coded for by pol and gag
form the nucleus of the maturing HIV particle; the gene prod-
ucts coded for by env form the gp120 "spikes" of the viral en-
velope. The gp120 spikes of the envelope are synthesized as
large gp160-precursor molecules and are cleaved by the HIV-1
protease into gp120 and gp41. The gag proteins are also derived
from a large 53 kD precursor molecule, from which the HIV-
protease cleaves the p24, p17, p9 and p7 gag proteins. Cleavage
of the precursor molecules by the HIV-1 protease is necessary
for the generation of infectious viral particles, and therefore the
viral protease represents another interesting target for therapeu-
tic blockade (40). The formation of new viral particles is a
stepwise process: a new virus core is formed by HIV-1 RNA,
gag proteins and various pol enzymes and moves towards the
cell surface. The large precursor molecules are cleaved by the
HIV-1 protease, which results in the infectious viral particles
budding through the host cell membrane. During the budding
process, the virus lipid membranes may incorporate various
host cell proteins and become enriched with certain phospho-
lipids and cholesterol. In contrast to T cells, where budding oc-
curs at the cell surface and virions are released into the extra-
cellular space, the budding process in monocytes and macro-
HIV and the Immune System 31
Hoffmann, Kamps, et al.
phages results in the accumulation of virions within cellular
vacuoles.
The replication of retroviruses is error prone and is character-
ized by a high spontaneous mutation rate. On average, re-verse
transcription results in 1-10 errors per genome and per round of
replication. Mutations can lead to the formation of replication-
incompetent viral species, but mutations causing drug resistance
may also accumulate, which, provided that there is selection
pressure under certain antiretroviral drugs and incomplete sup-
pression of viral replication, may be outgrowing.
In addition, viral replication is dynamic and turns over quickly
in infected individuals at an average rate of 10
9
new virus parti-
cles being produced and subsequently cleared per day. Thus,
within any individual, because of the extensive virus replication
and mutation rates, there exists an accumulation of many
closely related virus variants within the 'population' of viruses,
referred to as a viral "quasispecies". The selection pressure on
mostly the pre-existing mutations may not only be exerted by
certain drugs, but also by components of the immune system,
such as neutralizing antibodies or cytotoxic T cells (CTL).
HIV and the Immune System
The Role of Antigen-Presenting Cells in the Patho-
genesis of HIV Infection
Dendritic cells as prototypes of antigen-presenting cells
Dendritic cells, macrophages and B cells represent the main
antigen-presenting cells of the immune system. Dendritic cells
(DC) are the most potent inducers of specific immune responses
and are considered essential for the initiation of primary anti-
gen-specific immune reactions. DC precursors migrate from the
bone marrow towards the primary lymphatic organs and into the
submucosal tissue of the gut, the genitourinary and the respira-
tory tracts. They are able to pick up and process soluble anti-
32 Pathogenesis of HIV-1 Infection
HIV Medicine 2003 – www.HIVMedicine.com
gens and migrate to the secondary lymphatic organs, where they
activate antigen-specific T cells.
DC represent a heterogenous family of cells with different
functional capacities and expression of phenotypic markers,
depending on the local microenvironment and the stage of
maturation. Immature DC have the capacity to pick up and pro-
cess foreign antigens, but do not have great T cell stimulatory
capacities. However, mature DC show a predominant immu-
nostimulatory ability. DC in tissues and Langerhans cells,
which are specialized DC in the skin and mucosal areas, repre-
sent a more immature phenotype and may take up antigen. Once
these DC have taken up the antigen they migrate to the lym-
phoid tissues where they develop a mature phenotype.
The stimulation of CD8
+
T lymphocytes and the formation of
antigen-specific cytotoxic T-cells (CTL) depend on the presen-
tation of a peptide together with MHC class I antigens. DC may
become infected with viruses, for instance influenza. Viral pro-
teins are then produced within the cytoplasm of the cell, similar
to cellular proteins, then degraded to viral peptides and translo-
cated from the cytosol into the endoplasmatic reticulum, where
they are bound to MHC class I antigens. These peptide-MHC
class I complexes migrate to the DC surface. The number of
specific antigen-MHC class I complexes is usually limited and
must eventually be recognized by rare T cell clones, up to a ra-
tio of 1:100.000 or less. The T-cell receptor (TCR) may display
only a low binding affinity (1 mM or less). The high density of
co-stimulatory molecules on the DC surface, however, enhances
the TCR-MHC:peptide interaction allowing efficient signal-ling
to occur through the T cell and resulting in proliferation (clonal
expansion) of the T cell. Virus-infected cells or tumor cells of-
ten do not express co-stimulatory molecules, and thus may not
be able to induce a clonal expansion of effector cells. This un-
derscores the importance of having a highly specialized system
of antigen-presenting cells, i.e. DC, in operation to prime T
cells to expand and proliferate initially.
HIV and the Immune System 33
Hoffmann, Kamps, et al.
The interaction of dendritic cells and B/T-cells
B- and T-lymphocytes may be regarded as the principle effector
cells of antigen-specific immune responses. However, their
function is under the control of dendritic cells. DC are able to
pick up antigens in the periphery. These antigens are processed
and expressed on the cell surface, together with co-stimulatory
molecules that initiate T cell activation. B cells may recognize
antigen after binding to the B cell receptor. Recognition of anti-
gen by T cells requires previous processing and presentation of
antigenic peptides by DC. T cells express different T cell re-
ceptors (TCR), that may bind to the peptide:MHC class I on the
surface of dendritic cells to allow activation of CD8
+
T cells, or
to the peptide:MHC class II molecules, to activate CD4
+
T cells.
The ability of DC to activate T cells also depends on the secre-
tion of stimulatory cytokines such as IL-12, which is a key cy-
tokine for the generation and activation of T
H
1 and natural killer
(NK-) cells.
Only a few DC and small amounts of antigen are sufficient to
induce a potent antigen-specific T cell response, thus demon-
strating the immunostimulatory potency of DC. The expression
of adhesion molecules and lectins, such as DC-SIGN, support
the aggregation of DC and T cells and promote the engagement
of the T cell receptor (TCR). DC-SIGN is a type C lectin that
has also been shown to bind to lentiviruses such as SIV and
HIV-1 and -2 by interaction of gp120 with carbohydrates. In
vivo, immunohistochemical studies show expression of DC-
SIGN on submucosal and intradermal DC, suggesting an impli-
cation of DC-SIGN in vertical and mucosal transmission of
HIV. The expression of DC-SIGN was shown to enhance the
transmission of HIV to T cells and allows utilization of co-
receptors if their expression is limited. Thus DC-SIGN may be
a mechanism whereby HIV-1 is taken up by DC in the mucosal
tissues. It is then transported by the DC to the lymphoid tissues,
where HIV-1 can then infect all the residing CD4
+
T cells.
34 Pathogenesis of HIV-1 Infection
HIV Medicine 2003 – www.HIVMedicine.com
Lymphatic Tissue as the Site of Viral Replication
Viral replication within the lymphatic tissue is already exten-
sive in the early stages of the disease (42,43). During the initial
phase of HIV-1 infection, there is a burst of virus into the
plasma, followed by a relative decline in viremia. During this
time, a strong HIV-1 specific cytotoxic T cell response is gen-
erated, which coincides with the early suppression of plasma
viremia in most patients. Virions are trapped by the follicular
dendritic cell (FDC) network within the lymphoid tissue.
Macrophages, and activated and quiescent CD4
+
T cells are the
main targets of infection. During the whole course of infection
with HIV-1, the lymphoid tissue represents the principle site of
HIV-1 replication. The frequency of cells containing proviral
DNA is 5-10x higher in lymphoid tissue than in circulating pe-
ripheral mononuclear cells in the blood, and the difference in
viral replication in lymphoid tissue exceeds that in the periph-
eral blood by about 10-100x. Thus, the virus mainly accumu-
lates in the lymph nodes.
After entry of HIV-1 into a quiescent CD4
+
T cell and after
completion of reverse transcription, the viral genome is repre-
sented by proviral unintegrated HIV DNA. The activation of
CD4
+
T cells is necessary for the integration of the HIV DNA
into the host cell genome and is therefore a prerequisite for the
synthesis of new virions. In this regard, the micromilieu of the
lymphoid tissue represents the optimal environment for viral
replication. The close cell-cell contact between CD4
+
T-cells
and antigen-presenting cells, the presence of infectious virions
on the surface of the FDC, and an abundant production of pro-
inflammatory cytokines such as IL-1, IL-6 or TNFα, promotes
the induction of viral replication in infected cells and augments
viral replication in cells already producing the virus. It should
be noted that both IL-1 and TNFα induce NF-kb which binds to
the HIV-1 LTR to promote proviral transcription. The impor-
tance of an antigen-induced activation of CD4
+
T cells is un-
derlined by several in vivo and in vitro studies that demonstrate
HIV and the Immune System 35
Hoffmann, Kamps, et al.
an increase of HIV-1 replication in association with a tetanus or
influenza vaccination or an infection with Mycobacterium tu-
berculosis (44). Even though the clinical benefit of vaccination
against common pathogens (e.g. influenza and tetanus) in HIV-
1 infected patients outweighs the potential risk of a temporary
increase in viral load, these studies indicate that in every situa-
tion where the immune system is activated, enhanced viral rep-
lication can also occur.
Patients undergoing HAART demonstrate a dramatic decrease
in the number of productively infected CD4
+
T cells within the
lymphoid tissue (45). However, in all patients examined so far,
there persists a pool of latently infected quiescent T cells de-
spite successful suppression of plasma viremia (39). It is these
latently infected cells which may give rise to further rounds of
viral replication, if the antiviral drugs are stopped.
During the natural course of HIV-1 disease, the number of
CD4
+
T cells slowly decreases while plasma viremia rises in
most patients. If sequential analysis of the lymphoid tissue is
performed, progression of the disease is reflected by destruction
of the lymphoid tissue architecture and a decreased viral trap-
ping. Various immunohistological studies indicate that the
paracortex of the lymph nodes represents the primary site where
HIV replication is initiated (42,43). Infection of the surrounding
CD4
+
T cells, as well as the initiation of T cell activation by
DC, contributes to the spreading of HIV-1 within the lymphoid
environment.
The HLA System and the Immune Response against
HIV
CD8
+
T cells recognize "their" antigen (peptide) in context with
HLA class I molecules on antigen-presenting cells, whereas
CD4
+
T cells require the presentation of antigenic peptides in
context with HLA class II molecules. The generation of an HIV
specific immune response is therefore dependent on the indi-
vidual HLA pattern.
36 Pathogenesis of HIV-1 Infection
HIV Medicine 2003 – www.HIVMedicine.com
Antigen-presenting cells may bind HIV peptides in different
ways within "grooves" on the HLA class I molecules. There-
fore, CD8
+
T cells can be activated in an optimal or suboptimal
way or may not be activated at all. Using large cohorts of HIV-
1 infected patients, in whom the natural course of disease (fast
versus slow progression) is known, HLA patterns were identi-
fied that were associated with a slow versus fast disease pro-
gression. These studies suggest that the HLA type could be re-
sponsible for the benign course of disease in about 40% of pa-
tients with a long-term non-progressive course of disease. Ho-
mozygosity for HLA Bw4 is regarded as being protective. Pa-
tients who display heterozygosity at the HLA class I loci are
characterized by a slower progression of immunodeficiency
than patients with homozygosity at these loci (46).
An initial study by Kaslow in 1996 demonstrated that HLA
B14, B27, B51, B57 and C8 are associated with a slow disease
progression; in contrast, the presence of HLA A23, B37 and
B49 were associated with the rapid development of immunode-
ficiency (47).
All patients with HLA B35 had developed symptoms of AIDS
after 8 years of infection.
More recent studies suggest that discordant couples with a
"mismatch" at the HLA class I have a protective effect towards
heterosexual transmission (48).
In vitro studies in HLA B57 positive patients demonstrate that
these patients display HLA B57 restricted CTL directed against
HIV-1 peptides. However it is possible that the identification of
protective HLA alleles or HLA restricted peptides in HIV-1
infected patients with a benign course of disease does not nec-
essarily indicate that the same alleles or peptides are crucial for
the design of a protective vaccine. Kaul and co-workers were
able to show that CD8
+
T cells from HIV-1 exposed but unin-
fected African women recognize different epitopes than CD8
+
T
cells from HIV-1 infected African women (49). This suggests
that the epitopes that the immune system is directed against
HIV and the Immune System 37
Hoffmann, Kamps, et al.
during a natural infection might be different from those that are
protective against infection.
HLA class II antigens are crucial for the development of an
HIV-1 specific CD4
+
T cell response. Rosenberg (1997) was the
first to show that HIV-1 infected patients with a long-term non-
progressive course of disease had HIV-1 specific CD4
+
T cells
that could proliferate against HIV-1 antigens (50). The identifi-
cation of protective or unfavorable HLA class II alleles is less
well elaborated on than the knowledge about protective HLA
class I alleles. Cohorts of vertically infected children and HIV-
infected adults demonstrate a protective effect of HLA DR13
(51).
The HIV-specific Cellular Immune Response
In comparison to HIV-1 infected patients with a rapid decline of
CD4
+
T cells, patients with a long-term non-progressive course
of disease ("LTNP" = long-term non-progressors) have HIV-1-
specific CTL precursors in high numbers and with a broad
specificity towards various HIV-1 proteins. The different ca-
pacities of certain HLA alleles to present viral particles more or
less efficiently and to induce a more or less potent immune re-
sponse may explain why certain HLA alleles are associated
with a more rapid or a slow progressive course of disease (see
above).
Individuals have been described who developed CTL "escape"
mutants after years of stable disease and the presence of a
strong CTL response. The evolution of CTL escape mutants
was associated with a rapid decline in CD4
+
T cells in these pa-
tients, indicating the protective role of CTL (52).
HIV-specific CTL responses have been detected in HIV-1 ex-
posed but uninfected individuals. Nef-specific CTL have been
identified in HIV-1 negative heterosexual partners of HIV in-
fected patients and env-specific CTL have been found in sero-
negative healthcare workers after exposure to HIV-1 containing
material (needle stick injuries) (54).
38 Pathogenesis of HIV-1 Infection
HIV Medicine 2003 – www.HIVMedicine.com
The presence of a CTL response does not only correlate with
the suppression of plasma viremia during the initial phase of
HIV infection. Patients who underwent structured therapy inter-
ruptions, especially when HAART was initiated early following
infection, demonstrated the appearance of HIV-specific CTL
during the pauses.
However, it is still unclear in most patients who exhibit a potent
temporary CTL response, why this CTL response diminishes
later on. The appearance of viral "escape" mutants might ex-
plain why previously recognized epitopes are no longer immu-
nodominant.
The nef protein may downregulate HLA class I antigens and
therefore counteract the recognition of infected cells by CTL. In
addition, the majority of infected individuals do show detect-
able CTL responses. It is unclear why they are unable to control
the virus. Interestingly, CTL from HIV-infected patients shows
a lack of perforin and an immature phenotype, even though the
ability to secrete chemokines and cytokines is not impaired. It is
possible that the CTL in most HIV-1 infected individuals, al-
though detectable, may be functionally defective, and thus un-
able to completely clear the virus. CD8
+
T cells may also be-
come HIV infected, although this was not demonstrated for
HIV-specific CD8
+
T cells. It is unclear, whether CD8
+
T cells
might temporarily express CD4 and which chemokine co-
receptors mediate infection of these CD8
+
T cells.
In addition to the cytotoxic activities directed against HIV-
infected cells, CD8
+
T cells from HIV-1 infected patients ex-
hibit a remarkable soluble HIV-1 inhibitory activity that inhibits
HIV-1 replication in autologous and allogeneic cell cultures
(55). Despite multiple efforts, the identity of this inhibitory ac-
tivity ("CAF") has not been clarified, although chemokines,
such as MIP-1α, MIP-1ß, RANTES (24), IL-16 (56), the che-
mokine MDC (57) and defensins, may account for at least some
of the inhibition.
HIV and the Immune System 39
Hoffmann, Kamps, et al.
The T
H
1/T
H
2 Immune Response
Depending on the secretion pattern of cytokines, CD4
+
T cells
may be differentiated into T
H
1 and T
H
2 cells. T
H
1 CD4
+
T cells
primarily produce interleukin-2 (IL-2) and IFNγ, which repre-
sent the cytokines that support the effector functions of the im-
mune system (CTL, NK-cells, macrophages). T
H
2 cells pre-
dominantly produce IL-4, IL-10, IL-5 and IL-6, which represent
the cytokines that favor the development of a humoral immune
response. Since T
H
1 cytokines are critical for the generation of
CTLs, an HIV-1-specific T
H
1 response is regarded as being a
protective immune response. Studies on HIV-exposed but non-
infected individuals have shown, that following in vitro stimu-
lation with HIV-1 env antigens (gp120/gp160) and peptides, T
cells from these individuals secrete IL-2 in contrast to non-
exposed control persons (58). Similar studies were undertaken
in healthcare workers after needlestick injuries and in newborns
from HIV-infected mothers. Although these observations may
indicate that a T
H
1-type immune response is potentially protec-
tive, it should be considered that similar immune responses
might also have been generated after contact with noninfectious
viral particles and therefore do not necessarily imply a means of
protection against a replication-competent virus.
HIV-1-specific Humoral Immune Responses
The association between an HIV-1 specific humoral immune
response and the course of disease is less well characterized. A
slow progression of immunodeficiency was observed in patients
with high titers of anti-p24 antibodies (63), persistence of neu-
tralizing antibodies against primary and autologous virus (64),
and lack of antibodies against certain gp120 epitopes (62).
Long-term non-progressors with HIV tend to have a broad neu-
tralizing activity towards a range of primary isolates and show
persistence of neutralizing antibodies against autologous virus.
At present, it is unclear whether the presence of neutralizing
antibodies in LTNP represents part of the protection or whether
40 Pathogenesis of HIV-1 Infection
HIV Medicine 2003 – www.HIVMedicine.com
it merely reflects the integrity of a relatively intact immune
system. Individuals that have a substantial risk for HIV-1 infec-
tion, but are considered "exposed, non-infected", by definition
represent individuals with a lack of a detectable antibody re-
sponse to HIV-1. This definition implies that a systemic hu-
moral immune response may not represent a crucial protective
mechanism. It has been shown that these individuals may dem-
onstrate a local (mucosal) IgA response against HIV-1 proteins
that are not detected by the usual antibody testing methods (65,
66). Thus, local IgA, rather than systemic IgG, may be associ-
ated with protection against HIV-1 infection. There is also some
evidence that some anti-HIV-1 antibodies can enhance the in-
fection of CD4
+
T cells.
A number of old and recent studies have shown that neutraliz-
ing antibodies do exist in HIV-1 infected individuals; however,
they seem to lag in time. That is, individuals will develop neu-
tralizing antibodies to their own viruses with time, however, by
the time these antibodies develop, the new viruses circulating in
the individual's plasma will become resistant to neutralization,
even though the older ones are now sensitive to the current an-
tibodies in the patient's serum. Thus, the antibody response ap-
pears to be hitting a 'moving' target, allowing viruses to escape
continuously. Further knowledge gained on understanding the
mechanisms of humoral escape will likely lead to potential new
therapies.
Improved knowledge and understanding of the pathophysio-
logic mechanisms during the course of HIV-1 infection have
not only contributed to the development of antiretroviral treat-
ment strategies, but have given rise to new therapeutic ap-
proaches, such as cytokine therapies, e.g., IL-2 and therapeutic
vaccination. However, the most important challenge and thus,
the demand for a better understanding of the immunopatho-
genesis of HIV-1 infection, remains the development of a pro-
tective vaccine, which is urgently needed to interrupt the epi-
demic especially in countries of the Subsahara and Southeast
Asia.
References 41
Hoffmann, Kamps, et al.
References
1. Barre-Sinoussi F, Chermann JC, Rey F, et al. Isolation of a T-
lymphotropic retrovirus from a patient at risk for AIDS. Science 1983,
220: 868-71. http://amedeo.com/lit.php?id=6189183
2. Gallo RC, Sarin PS, Gelmann EP, et al. Isolation of human T cell leuke-
mia virus in acquired immune deficiency syndrome (AIDS). Science 1983,
220: 865-7. http://amedeo.com/lit.php?id=6601823
3. Clavel F, Guetard D, Brun-Vezinet F, Chamaret S, Rey MA, Santos-
Ferreira O. Isolation of a new human retrovirus from West African pa-
tients with AIDS. Science 1986, 233: 343
4. Liu SL, Schacker T, Musey L, et al. Divergent patterns of progression to
AIDS after infection from the same source: HIV type 1 evolution and anti-
viral responses. J Virol 1997, 71: 4284-95.
http://amedeo.com/lit.php?id=9151816
5. Kirchhoff F, Greenough TC, Brettler DB, Sullivan JL, Desrosiers RC. Brief
report: Absence of intact nef sequences in a long-term survivor with non-
progressive HIV-1 infection. N Engl J Med 1995, 332: 228-32
6. Oh SY, Cruickshank WW, Raina J, et al. Identification of HIV-1 envelope
glykoprotein in the serum of AIDS and ARC patients. J Acquired Immune
Defic Syndr 1992, 5: 251.
7. Sunila I, Vaccarezza M, Pantaleo G, Fauci AS, Orenstein JM. Gp120 is
present on the plasma membrane of apoptotic CD4 cells prepared from
lymph nodes of HIV-1-infected individuals: an immunoelectron micro-
scopic study. AIDS 1997, 11: 27-32.
http://amedeo.com/lit.php?id=9110072
8. Gelderblom HR, Gentile M, Scheidler A, Özel M, Pauli G. Zur Struktur
und Funktion bei HIV. AIFO 1993, 5: 231.
9. Wong-Staal F. HIVes and their replication. In: Fundamental Virology, Ed.:
Fields BN, Knipe DM et al. Raven Press, Ltd., New York 1991.
10. Wei P, Garber ME, Fang SM, Fischer WH, Jones KA. A novel CDK9-
associated C-type cyclin interacts directly with HIV-1 Tat and mediates its
high-affinity, loop-specific binding to TAR RNA. Cell 1998, 92: 451-62.
http://amedeo.com/lit.php?id=9491887
11. Aiken C, Konner J, Landau NR, Lenburg ME, Trono D. Nef induces CD4
endocytosis: Requirement for a critical dileucine motif in the membrane-
proximal CD4 cytoplasmic do-main. Cell 1994, 76: 853-64.
http://amedeo.com/lit.php?id=8124721
12. Collins KL, Chen BK, Walker BD, Baltimore D. HIV-1 nef protein protects
infected primary cells against killing by cytotoxic T lymphocytes. Nature
1998, 391: 397-401. http://amedeo.com/lit.php?id=9450757
13. Peter F. HIV nef: The mother of all evil? Immunity, 1998, 9: 433-7.
14. Miller RH, Sarver N. HIV accessory proteins as therapeutic targets. Nat
Med 1997, 3: 389-94.
42 Pathogenesis of HIV-1 Infection
HIV Medicine 2003 – www.HIVMedicine.com
15. Bour S, Schubert U, Strebel K. The HIV type 1 vpu protein specifically
binds to the cytoplasmic domain of CD4: Implications for the mechanism
of degradation. J Virol 1995, 69: 1510-20.
http://amedeo.com/lit.php?id=7853484
16. Cullen BR. HIV-1 auxiliary proteins: making connections in a dying cell.
Cell 1998, 93: 685-92.
17. Schooley RT, Merigan TC, Gaut P, et al. Recombinant soluble CD4 ther-
apy in patients with AIDS and ARC. Ann Intern Med 1990, 112: 247-53.
http://amedeo.com/lit.php?id=2297203
18. Bour S, Geleziunas R, Wainberg MA. The HIV type 1 (HIV-1) CD4 re-
ceptor and its central role in promotion of HIV-1 infection. Microbiol Rev
1995, 59: 63-93. http://amedeo.com/lit.php?id=7708013
19. Banda NK, Bernier J, Kurahara DK, et al. Cross-linking CD4 by HIV
gp120 primes T cells for activation induced apoptosis. J Exp Med 1992,
176: 1099-106. http://amedeo.com/lit.php?id=1402655
20. Dalgleish AG, Beverley PC, Clapham PR, et al. The CD4 (T4) antigen is
an essential component of the receptor for the AIDS retrovirus. Nature
1984, 312: 763-7. http://amedeo.com/lit.php?id=6096719
21. Klatzmann D, Champagne E, Chamaret S, et al. T-lymphocyte T4 mole-
cule behaves as the receptor for human retrovirus LAV. Nature 1984,
312: 767-8. http://amedeo.com/lit.php?id=6083454
22. Edwards TG, Hoffman TL, Baribaud F, et al. Relationships between CD4
independence, neutralization sensitivity and exposure of a CD4-induced
epitope in an HIV-1 envelope protein. J Virol 2001, 75:5230-9.
http://amedeo.com/lit.php?id=11333905
23. Levy JA, Mackewicz CE, Barker E. Controlling HIV pathogenesis: the role
of thenoncytotoxic anti-HIV response of CD8+ T cells. Immunol Today
1996,17: 217-24. http://amedeo.com/lit.php?id=8991383
24. Cocchi F, DeVico AL, Garzino-Demo A, Arya S, Gallo RC, Lusso P. Iden-
tification of RANTES, MIP-1a, and MIP-1ß as the major HIV-suppressive
factors produced by CD8+ T cells. Science 1995, 270: 1811-5.
http://amedeo.com/lit.php?id=8525373
25. Deng H, Liu R, Ellmeier W, et al. Identification of a major co-receptor for
primary isolates of HIV-1. Nature 1996, 381: 661-6.
http://amedeo.com/lit.php?id=8649511
26. Doranz BJ, Rucker J, Yi Y, et al. A dual-tropic primary HIV-1 isolate that
uses fusin and the ß-chemo-kine receptors CKR-5, CKR-3, and CKR-2b
as fusion cofactors. Cell 1996, 85: 1149-58.
http://amedeo.com/lit.php?id=8674120
27. Dragic T, Litwin V, Allaway GP, et al. HIV-1 entry into CD4+ cells is me-
diated by the chemokine receptor CC-CKR-5. Nature 1996, 381: 667-73.
http://amedeo.com/lit.php?id=8649512
28. Feng Y, Broder CC, Kennedy PE, Berger EA. HIV-1 entry cofactor: func-
tional cDNA cloning of a seven-transmembrane, G protein-coupled re-
ceptor. Science 1996, 272: 872-7. http://amedeo.com/lit.php?id=8629022
References 43
Hoffmann, Kamps, et al.
29. Chan DC, Fass D, Berger JM, Kim PS. Core structure of gp41 from the
HIV envelope glycoprotein. Cell 1997, 89: 263-73.
http://amedeo.com/lit.php?id=9108481
30. Kilby JM, Hopkins S, Venetta TM, et al. Potent suppression of HIV-1
replication in humans by T-20, a peptide inhibitor of gp41-mediated virus
entry. Nat Med 1998, 4: 1302-7. http://amedeo.com/lit.php?id=9809555
31. Deng HK, Unutmaz D, Kewalramani VN, Littman DR. Expression cloning
of new receptors used by simian and human immunodeficiency viruses.
Nature 1997, 388: 296-300. http://amedeo.com/lit.php?id=9230441
32. Liao F, Alkhatib G, Peden KWC, Sharma G, Berger EA, Farber JM.
STRL-33, a novel chemokine receptor-like protein, functions as a fusion
cofactor for both macrophage-tropic and T cell line-tropic HIV-1. J Exp
Med 1997, 185: 2015-23. http://amedeo.com/lit.php?id=9166430
33. Liu R, Paxton WA, Choe S, et al. Homozygous defect in HIV-1 coreceptor
accounts for resistance of some multiply-exposed individuals to HIV-1
infection. Cell 1996, 86: 367-77. http://amedeo.com/lit.php?id=8756719
34. Biti R, Ffrench RF, Young J, Bennetts B, Stewart G, Liang T. HIV-1 infec-
tion in an individual homozygous for the CCR5 deletion allele. Nature
Med 1997, 3: 252-3.
35. Dean M, Carrington M, Winkler C, et al. Genetic restrictions of HIV-1
infection and progression to AIDS by a deletion allele of the CKR5 struc-
tural gene. Science 1996, 273: 1856-62.
http://amedeo.com/lit.php?id=8791590
36. Anzala AO, Ball TB, Rostron T, O'Brien SJ, Plummer FA, Rowland-Jones
SL. CCR2-64I allele and genotype association with delayed AIDS pro-
gression in African women. Lancet 1998, 351: 1632-3.
37. Winkler C, Modi W, Smith MW, et al. Genetic restriction of AIDS patho-
genesis by an SDF-1 chemokine gene variant. Science 1998, 279: 389-
93. http://amedeo.com/lit.php?id=9430590
38. Zack JA, Arrigo SJ, Weitsman SR, Go AS, Haislip A, Chen ISY. HIV-1
entry into quiescent primary lymphocytes: Molecular analysis reveals a
labile, latent viral structure. Cell 1990, 61: 213-22.
http://amedeo.com/lit.php?id=2331748
39. Chun TW, Carruth L, Finzi D, et al. Quantification of latent tissue reser-
voirs and total body viral load in HIV-1 infection. Nature 1997,387:183-8.
http://amedeo.com/lit.php?id=9144289
40. Kohl NE, Emini EA, Schleif WA, et al. Active HIV protease is required for
viral infectivity. Proc Natl Acad Sci USA, 1988, 85: 4686-90.
http://amedeo.com/lit.php?id=3290901
41. Geijtenbeek TB, Torensma R, van Vliet SJ, et al. Identification of DC-
SIGN, a novel dendritic cell-specific ICAM-3 receptor that supports pri-
mary immuen responses. Cell 2000, 100: 575-85.
http://amedeo.com/lit.php?id=10721994
42. Embretson J, Zupancic M, Ribas JL, et al. Massive covert infection of
helper T lymphocytes and macrophages by HIV during the incubation pe-
44 Pathogenesis of HIV-1 Infection
HIV Medicine 2003 – www.HIVMedicine.com
riod of AIDS. Nature 1993, 362: 359-62.
http://amedeo.com/lit.php?id=8096068
43. Pantaleo G, Graziosi C, Demarest JF, et al. HIV infection is active and
progressive in lymphoid tissue during the clinically latent stage of dis-
ease. Nature 1993, 362: 355-8. http://amedeo.com/lit.php?id=8455722
44. O’Brien WA, Grovit-Ferbas K, Namazi A, et al. HIV-type 1 replication can
be increased in peripheral blood of seropositive patients after influenza
vaccination. Blood 1995, 86: 1082-9.
http://amedeo.com/lit.php?id=7620162
45. Tenner-Racz K, Stellbrink HJ, van Lunzen J, et al. The unenlarged lymph
nodes of HIV-1-infected, asymptomatic patients with high CD4 T cell
counts are sites for virus replication and CD4 T cell proliferation. The im-
pact of HAART. J Exp Med 1998, 187: 949-59.
http://amedeo.com/lit.php?id=9500797
46. Carrington M, Nelson GW, Martin MP, et al. HLA and HIV-1: heterozy-
gote advantage and B*35-Cw*04 disadvantage. Science 1999: 12, 28:
1748-52. http://amedeo.com/lit.php?id=10073943
47. Kaslow RA, Carrington M, Apple R, et al. Influence of combinations of
human major histocompatibility complex genes on the course of HIV-1
infection. Nat Med 1996: 2: 405-11.
http://amedeo.com/lit.php?id=8597949
48. Lockett SF, Robertson JR, Brettle RP, et al. Mismatched human leuko-
cyte antigen alleles protect against heterosexual HIV transmission. J Acq
Imm Defic Syndr 2001: 27: 277-80.
http://amedeo.com/lit.php?id=11464148
49. Kaul R, Rowland-Jones SL, Kimani J, et al. New insights into HIV-1 spe-
cific cytotoxic T-lymphocyte responses in exposed, persistently sero-
negative Kenyan sex workers. Immunol Lett 2001, 79: 3-13.
http://amedeo.com/lit.php?id=11595284
50. Rosenberg ES, Billingsley JM, Caliendo AM, et al. Vigorous HIV-1-
specific CD4+ T cell responses associa-ted with control of viremia. Sci-
ence 1997, 278: 1447-50. http://amedeo.com/lit.php?id=9367954
51. Keet IP, Tang J, Klein MR, et al. Consistent associations of HLA class I
and II and transporter gene products with progression of HIV type 1 in-
fection in homosexual men. J Infect Dis 1999, 180: 299-309.
http://amedeo.com/lit.php?id=10395843
52. Goulder PJ, Phillips RE, Colbert RA, et al. Late escape from an immun-
dominant cytotoxic T-lymphocyte response associated with progression
to AIDS. Nat Med 1997, 3: 212-7. http://amedeo.com/lit.php?id=9018241
53. Ogg GS, Jin X, Bonhoeffer S, et al. Quantitation of HIV-1-specific cyto-
toxic T lymphocytes and plasma load of viral RNA. Science 1998, 279:
2103-6. http://amedeo.com/lit.php?id=9516110
54. Pinto LA, Sullivan J, Berzofsky JA, et al. Env-specific cytotoxic T lympho-
cyte responses in HIV seronegative health care workers occupationally
exposed to HIV contaminated body fluids. J Clin Invest 1995, 96: 867-76.
http://amedeo.com/lit.php?id=7635981
References 45
Hoffmann, Kamps, et al.
55. Walker CM, Moody DJ, Stites DP, Levy JA. CD8+ lymphocytes can con-
trol HIV infection in vitro by suppressing viral replication. Science 1986,
234: 1563-6. http://amedeo.com/lit.php?id=2431484
56. Baier M, Werner A, Bannert N, Metzner K, Kurth R. HIV suppression by
interleukin-16. Nature 1995, 378: 563.
57. Pal R, Garzino-Demo A, Markham PD, et al. Inhibition of HIV-1 infection
by the ß-chemokine MDC. Science 1997, 278: 695-8.
http://amedeo.com/lit.php?id=9381181
58. Clerici M, Giorgi JV, Chou CC, et al. Cell-mediated immune response to
HIV (HIV) type-1 in seronegative homosexual men with a recent sexual
exposure to HIV-1. J Infect Dis 1992, 165: 1012-9.
http://amedeo.com/lit.php?id=1533867
59. Murakami T, Nakajima T, Koyanagi Y, et al. A small molecule CXCR4
inhibitor that blocks T cell line-tropic HIV-1 infection. J Exp Med 1997,
186: 1389-93. http://amedeo.com/lit.php?id=9334379
60. Schols D, Struyf S, Van Damme J, et al. Inhibition of T-tropic HIV strains
by selective antagonization of the chemokine receptor CXCR4. J Exp
Med 1997, 186: 1383-8. http://amedeo.com/lit.php?id=9334378
61. Zou YR, Kottmann AH, Kuroda M, Taniuchi I, Littman DR. Function of
the chemokine receptor CXCR4 in haematopoiesis and in cerebellar de-
velopment. Nature 1998, 393: 595-9.
http://amedeo.com/lit.php?id=9634238
62. Wong MT, Warren RQ, Anderson SA, et al. Longitudinal analysis of the
humoral immune response to HIV type 1 gp160 epitopes in rapidly pro-
gressing and nonprogressing HIV-1 infected subjects. J Infect Dis 1993,
168: 1523-7. http://amedeo.com/lit.php?id=7504036
63. Hogervorst E, Jurriaans S, de Wolf F, et al. Predictors for non-and slow
progression in HIV type 1 infection: low viral RNA copy numbers in serum
and maintenance of high HIV-1 p24-specific but not V3-specific antibody
levels. J Infect Dis 1995, 171: 811-21.
http://amedeo.com/lit.php?id=7706807
64. Montefiori DC, Pantaleo G, Fink LM, et al. Neutralizing and infection-
enhancing antibody responses to HIV type 1 in long-term nonprogres-
sors. J Infect Dis 1996, 173: 60-7. http://amedeo.com/lit.php?id=8537683
65. Mazzoli S, Trabattoni D, Lo Caputo S, et al. HIV-specific mucosal and
cellular immunity in HIV-seronegative partners of HIV-seropositive indi-
viduals. Nat Med 1997, 3:1250-7. http://amedeo.com/lit.php?id=9359700
66. Saha K, Zhang J, Gupta A et al. Isolation of primary HIV-1 that target
CD8+ T lymphocytes using CD8 as a receptor. Nat Med 2001, 7: 65-72.
http://amedeo.com/lit.php?id=11135618
Introduction 47
Hoffmann, Kamps, et al.
Chapter 2: Acute HIV-1 Infection
Marcus Altfeld and Bruce D. Walker
Introduction
Acute HIV-1 infection presents in 40 – 90 % of cases as a tran-
sient symptomatic illness, associated with high levels of HIV-1
replication and an expansive virus-specific immune response.
With 14,000 new cases per day worldwide, it is an important
differential diagnosis in cases of fever of unknown origin,
maculopapular rash and lymphadenopathy.
The diagnosis of acute infection is missed in the majority of
cases, as other viral illnesses (“flu”) are often assumed to be the
cause of the symptoms, and there are no HIV-1-specific anti-
bodies detectable at this early stage of infection. The diagnosis
therefore requires a high degree of clinical suspicion, based on
clinical symptoms and history of exposure, in addition to spe-
cific laboratory tests (detection of HIV-1 RNA or p24 antigen
and negative HIV-1 antibodies) confirming the diagnosis.
An accurate early diagnosis of acute HIV-1 infection is impor-
tant, as patients may benefit from therapy at this early stage of
infection (see below), and infection of sexual partners can be
prevented.
Signs and Symptoms
After an incubation period of a few days to a few weeks, most
cases present with an acute flu-like illness. The most common
symptoms (see Table 1) are fever, maculopapular rash, oral ul-
cers, lymphadenopathy, arthralgia, pharyngitis, malaise, weight
loss, aseptic meningitis and myalgia. In a recently published
study by Hecht et al., fever (80 %) and malaise (68 %) had the
highest sensitivity for clinical diagnosis of acute HIV-1 infec-
tion, whereas loss of weight (86 %) and oral ulcers (85 %) had
48 Acute HIV-1 Infection
HIV Medicine 2003 – www.HIVMedicine.com
the highest specificity. In this study, the symptoms of fever and
rash (especially in combination), followed by oral ulcers and
pharyngitis had the highest positive predictive value for diagno-
sis of acute HIV-1 infection. In another study by Daar et al.,
fever, rash, myalgia, arthralgia and night sweats were the best
predictors for acute HIV-1 infection.
Table 1: Main symptoms of acute HIV-1 infection
Symptom Frequency Odds ratio (95% CI)
Fever 80% 5.2 (2.3-11.7)
Rash 51% 4.8 (2.4-9.8)
Oral ulcers 37% 3.1 (1.5-6.6)
Arthralgia 54% 2.6 (1.3-5.1)
Pharyngitis 44% 2.6 (1.3-5.1)
Loss of appetite 54% 2.5 (1.2-4.8)
Weight loss > 2.5 kg 32% 2.8 (1.3-6.0)
Malaise 68% 2.2 (1.1-4.5)
Myalgia 49% 2.1 (1.1-4.2)
Fever and rash 46% 8.3 (3.6-19.3)
From: Hecht FM et al. Use of laboratory tests and clinical symptoms for
identification of primary HIV infection. AIDS 2002, 16: 1119-1129
The symptomatic phase of acute HIV-1 infection lasts between
7 – 10 days, and rarely longer than 14 days. The severity and
duration of symptoms has prognostic implications, as severe
and prolonged symptoms are associated with more rapid disease
progression. The nonspecific nature of the symptoms poses a
great challenge to the clinician and underlines the importance of
a detailed history of exposure.
Diagnosis
The diagnosis of acute HIV-1 infection is based on the detec-
tion of HIV-1 replication in the absence of HIV-1 antibodies, as
these are not yet present at this early stage of infection. Differ-
ent tests are available for diagnosis of acute HIV-1 infection.
Diagnosis 49
Hoffmann, Kamps, et al.
The most sensitive tests are based on detection of plasma HIV-1
RNA.
In a recently published study, all assays for HIV-1 RNA that
were tested (branched chain DNA, PCR and GenProbe) had a
sensitivity of 100 %, but occasionally (in 2 – 5 % of cases) led
to false positive results. False positive results from these tests
are usually below 2,000 copies HIV-1 RNA per ml plasma, and
therefore far below the high titers of viral load normally seen
during acute HIV-1 infection (in our own studies on average 13
x 10
6
copies HIV-1 RNA/ml with a range of 0.25 – 95.5 x 10
6
copies HIV-1 RNA/ml). Repetition of the assay for HIV-1 RNA
from the same sample with the same test led to a negative result
in all false positive cases. Measurement of HIV-1 RNA from
duplicate samples therefore results in a sensitivity of 100 %
with 100 % specificity. In contrast, detection of p24 antigen has
a sensitivity of only 79 % with a specificity of 99.5 – 99.96 %.
The diagnosis of acute HIV-1 infection must be subsequently
confirmed with a positive HIV-1 antibody test (seroconversion)
within the following weeks.
During acute HIV-1 infection, there is frequently a marked de-
crease of CD4+ cell count, which later increases again, but usu-
ally does not normalize to the initial levels. In contrast, the
CD8
+
cell count rises initially, which may result in a
CD4
+
/CD8
+
ratio of < 1. Infectious mononucleosis is the most
important differential diagnosis. Hepatitis, influenza, toxoplas-
mosis, syphilis and side effects of medications may also be con-
sidered.
In summary, the most important step in the diagnosis of acute
HIV-1 infection is to include it in the differential diagnosis. The
clinical suspicion of an acute HIV-1 infection then merely re-
quires performance of an HIV-1 antibody test and possibly re-
peated testing of HIV-1 viral load, as shown in the algorithm in
Figure 1 (adapted from Hecht et al., AIDS 2002).
50 Acute HIV-1 Infection
HIV Medicine 2003 – www.HIVMedicine.com
Figure 1
Treatment
The goal of antiretroviral therapy during acute HIV-1 infection
is to reduce the number of infected cells, preserve HIV-1-
specific immune responses and possibly lower the viral set
point in the long term. Several studies in recent years have
shown that treatment of acute HIV-1 infection allows long-term
viral suppression, leads to preservation and even increase of
HIV-1-specific T helper cell responses and allows for the con-
servation of a very homogeneous virus population.
First studies in patients who were treated during acute HIV-1
infection and subsequently went through structured treatment
interruptions show that the HIV-1-specific immune response
References 51
Hoffmann, Kamps, et al.
could be boosted in these patients. Most patients were subse-
quently able to discontinue therapy and experienced at least
temporal control of viral replication, with viral set points re-
maining below 5,000 copies/ml for more than 3 years in some
patients. However, in a number of individuals viral load re-
bounded to higher level during longer follow-up, requiring the
initiation of therapy.
The long-term clinical benefit of early initiation of therapy has
not been demonstrated yet. It is also not known how long the
period between acute infection and initiation of therapy can be
without losing immunological, virological and clinical benefit.
In view of all these unanswered questions, patients with acute
HIV-1 infection should be treated in controlled clinical trials. If
this is not possible, the option of standard first-line treatment
should be offered and discussed. Usually, treatment continues
for at least a year, followed by structured treatment interrup-
tions within the framework of controlled studies. It is important
during counseling to clearly indicate the lack of definitive data
on clinical benefit and to address the risks of antiretroviral ther-
apy and treatment interruptions, including drug toxicity, devel-
opment of resistance, acute retroviral syndrome during viral
rebound and HIV-1 transmission and superinfection during
treatment interruptions.
References
1. Rosenberg ES Altfeld M, Poon SH, et al. Immune control of HIV-1 after
early treatment of acute infection. Nature 2000, 407:523-6.
http://amedeo.com/lit.php?id=11029005
2. Kahn JO and Walker BD. Acute HIV type 1 infection. New Eng J Med
1998, 339:33-9.
3. Altfeld M, Rosenberg ES, Shankarappa R, et al. Cellular Immune Re-
sponses and Viral Diversity in Individuals Treated during Acute and Early
HIV-1 Infection. J Exp Med 2001, 193:169-180.
http://amedeo.com/lit.php?id=11148221
4. Hecht FM, Busch MP, Rawal B, et al. Use of laboratory tests and clinical
symptoms for identification of primary HIV infection. AIDS 2002, 16:1119-
1129. http://amedeo.com/lit.php?id=12004270
52 Acute HIV-1 Infection
HIV Medicine 2003 – www.HIVMedicine.com
5. Yeni PG, et al. Antiretroviral treatment for adult HIV infection in 2002:
updated Recommendations of the International AIDS Society-USA Panel.
JAMA 2002, 288:222-235. http://jama.ama-
assn.org/issues/v288n2/ffull/jst20002.html
6. Daar E Little S, Pitt J, et al. Diagnosis of primary HIV-1 infection. Ann
Intern Med 2001, 134:25-29. http://amedeo.com/lit.php?id=11187417
7. The PRN Notebook – Special Edition February 2002: Primary HIV-1 In-
fection. (http://www.prn.org)
1. Perspective 53
Hoffmann, Kamps, et al.
Chapter 3: HIV Therapy 2003
1. Perspective
Christian Hoffmann
The development of antiretroviral therapy has been one of the
most dramatic progressions in the history of medicine. Few
other areas have been subject to such fast- and short-lived
trends. Those who have experienced the rapid developments of
the last few years have been through many ups and downs:
The early years, from 1987-1990, brought great hope and the
first modest advances using monotherapy (Volberding et al.
1990, Fischl et al. 1990). But, by the time the results of the
Concorde Study had arrived (Hamilton et al. 1992, Concorde
1994), both patients and clinicians had plunged into a depres-
sion that was to last for several years. Zidovudine was first
tested on humans in 1985, and introduced as a treatment in
March 1987 with great expectations. Initially, at least, it did not
seem to be very effective. The same was true for the nucleoside
analogs zalcitabine, didanosine and stavudine, introduced be-
tween 1991 and 1994. The lack of substantial treatment options
led to a debate that lasted for several years about which nucleo-
side analogs should be used, when, and at what dose. One such
question was: Should the alarm clock be set to go off during the
night for a sixth dose of zidovudine?
Many patients, who were infected during the early and mid-80s,
began to die. Hospices were established, as well as more and
more support groups and ambulatory nursing services. One be-
came accustomed to AIDS and its resulting death toll. There
was, however, definite progress in the field of opportunistic in-
fections (OI) – cotrimoxazole, pentamidine, ganciclovir, for-
scarnet and fluconazole saved many patients’ lives, at least in
the short-term. Some clinicians started to dream of a kind of
“mega-prophylaxis”. But the general picture was still tainted by
54 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
an overall lack of hope. Many remember the somber, almost
depressed mood of the IX
th
World AIDS Conference in Berlin,
in June 1993. Between 1989 and 1994, morbidity and mortality
rates were hardly affected.
Then, in September 1995, the preliminary results of the Euro-
pean-Australian DELTA Study (Delta 1995) and the American
ACTG 175 Study (Hammer et al. 1996) attracted attention. It
became apparent that combination therapy with two nucleoside
analogs was more effective than monotherapy. Indeed, the dif-
ferences made on the clinical endpoints (AIDS, death) were
highly significant. Both studies demonstrated that it was poten-
tially of great importance to immediately start treatment with
two nucleoside analogs, as opposed to using the drugs “sequen-
tially”.
This was by no means the final breakthrough. By this time, the
first studies with protease inhibitors (PIs), a completely new
drug class, had been ongoing for several months. PIs had been
designed in the lab using the knowledge of the molecular
structure of HIV and protease – their clinical value was initially
uncertain. Preliminary data, and many rumors, were already in
circulation. In the fall of 1995, a fierce competition started up
between three companies: Abbott, Roche and MSD. The li-
censing studies for the three PIs, ritonavir, saquinavir and indi-
navir, were pursued with a great amount of effort, clearly with
the goal of bringing the first PI onto the market. The monitors
of these studies in the different companies “lived” for weeks at
the participating clinical sites. Deep into the night, case report
files had to be perfected and thousands of queries answered. All
these efforts led to a fast track approval, between December
1995 and March 1996, for all three PIs – first saquinavir, fol-
lowed by ritonavir and indinavir – for the treatment of HIV.
Many clinicians (including the author) were not really aware at
the time of what was happening during these months. AIDS
remained ever present. Patients were still dying, as only a rela-
tively small number were participating in the PI trials – and
very few were actually adequately treated by current standards.
1. Perspective 55
Hoffmann, Kamps, et al.
Doubts remained. Hopes had already been raised too many
times in the previous years by alleged miracle cures. Early in
January 1996, other topics were more important: palliative
medicine, treatment of CMV, MAC and AIDS wasting syn-
drome, pain management, ambulatory infusion therapies, even
euthanasia.
In February 1996, during the 3
rd
Conference on Retroviruses
and Opportunistic Infections (CROI) in Washington, many
caught their breath as Bill Cameron reported the first data from
the ABT-247 Study during the latebreaker session. The audito-
rium was absolutely silent. Riveted, listeners heard that the
mere addition of ritonavir oral solution decreases the frequency
of death and AIDS from 38 % to 22 % (Cameron et al. 1998).
These were sensational results in comparison to everything else
that had been previously published!
But for many, the combination therapies that became widely
used from 1996 onwards, still came too late. Some severely ill
patients with AIDS managed to recover during these months,
but, even in 1996, many still died. Although the AIDS rate in
large centers had been cut in half between 1992 and 1996
(Brodt et al. 1997), in smaller centers roughly every fifth patient
died in this year.
However, the potential of the new drugs was slowly becoming
apparent, and the World AIDS Conference in Vancouver a few
months later, in June 1996, was like a big PI party. Even regular
news channels reported in great depth on the new “AIDS cock-
tails”. The strangely unscientific expression “highly active
antiretroviral therapy” (HAART) began to spread irreversibly.
Clinicians were only too happy to become infected by this en-
thusiasm.
By this time, David Ho, Time magazine’s “Man of the Year” in
1996, had shed light on the hitherto completely misunderstood
kinetics of HIV with his breakthrough research (Ho et al. 1995,
Perelson et al. 1996). A year earlier, Ho had already initiated
the slogan “hit hard and early”, and almost all clinicians were
56 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
now taking him by his word. With the new knowledge of the
incredibly high turnover of the virus and the relentless daily
destruction of CD4+ T cells, there was no longer any consid-
eration of a “latent phase” – and no life without antiretroviral
therapy. In many centers almost every patient was treated with
HAART. Within only three years, from 1994-1997, the propor-
tion of untreated patients in Europe decreased from 37 % to
barely 9 %, whilst the proportion of HAART patients rose from
2 % to 64 % (Kirk et al. 1998).
Things were looking good. By June 1996, the first non-
nucleoside reverse transcriptase inhibitor, nevirapine, was li-
censed, and a third drug class introduced. Nelfinavir, another
PI, had also arrived. Most patients seemed to tolerate the drugs
well. 30 pills a day? No problem, if it helps. And how it helped!
The number of AIDS cases was drastically reduced. Within
only four years, between 1994 and 1998, the incidence of AIDS
in Europe was reduced from 30.7 to 2.5 per 100 patient years –
i.e. to less than a tenth. The reduction in the incidence of several
feared OIs, particularly CMV and MAC, was even more dra-
matic. HIV ophthalmologists had to look for new areas of work.
The large OI trials, planned only a few months before, faltered
due to a lack of patients. Hospices, which had been receiving
substantial donations, had to shut down or reorientate them-
selves. The first patients began to leave the hospices, and went
back to work; ambulatory nursing services shut down. AIDS
wards were occupied by other patients.
In 1996 and 1997 some patients began to complain of an in-
creasingly fat stomach, but was this not a good sign after the
years of wasting and supplementary nutrition? Not only did the
PIs contained lactose and gelatin, but the lower viremia was
thought to use up far less energy. It was assumed that, because
patients were less depressed and generally healthier, they would
eat more. At most, it was slightly disturbing that the patients
retained thin faces. However, more and more patients also be-
gan to complain about the high pill burden.
1. Perspective 57
Hoffmann, Kamps, et al.
In June 1997, the FDA published the first warning about the
development of diabetes mellitus associated with the use of PIs
(Ault 1997). In February 1998, the CROI in Chicago finally
brought home the realization among clinicians that protease
inhibitors were perhaps not as selective as had long been be-
lieved. One poster after the next, indeed whole walls of pictures
showed fat abdomens, buffalo humps, thin legs and faces. A
new term was introduced at the beginning of 1998, which
would influence the antiretroviral therapy of the years to come:
lipodystrophy. And so the old medical wisdom was shown to
hold true even for HAART: all effective drugs have side effects.
The actual cause of lipodystrophy remained completely unclear.
Then, in early 1999, a new hypothesis emerged from the Neth-
erlands: “mitochondrial toxicity”. It has become a ubiquitous
term in HIV medicine today.
The dream of eradication (and a cure), still widely hoped for in
the beginning, eventually had to be abandoned, too. Mathemati-
cal models are evidently not suitable for predicting what will
really happen. In 1997, it was still estimated that viral suppres-
sion, with a maximum duration of three years, was necessary;
after this period, it was predicted that all infected cells would
presumably have died. Eradication was the magic word. At
every conference since then, the duration of three years has
been adjusted upwards. Nature is not so easy to predict, and
newer studies came to the sobering conclusion that HIV re-
mains detectable in latent infected cells, even after long-term
suppression. To date, nobody knows how long these latent in-
fected cells survive, and whether even a small number of them
would be sufficient for the infection to flare up again as soon as
treatment is interrupted. Finally, during the Barcelona World
AIDS Conference, experts in the field admitted to bleak pros-
pects for eradication. The most recent estimates for eradication
of these cells were approximately 50-70 years. One thing is
certain: HIV will not be curable for at least the next 10 years.
Instead of eradication, it has become more realistic to consider
the lifelong management of HIV infection as a chronic disease
58 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
in the future, similar to diabetes mellitus. This means, however,
that drugs have to be administered over many years, which
demands an enormous degree of discipline from patients. Those
who are familiar with the management of diabetes understand
the challenges that patients and clinicians have to face and how
important it will be to develop better combinations in the com-
ing years. Hardly anyone will have the discipline and ability,
both mentally and physically, to take the currently available
pills several times daily at fixed times for the next ten, twenty
or even thirty years. But, presumably, this will not be necessary.
There will be new and improved treatment regimens. Once-
daily regimens are coming; maybe even twice-weekly.
At the same time, the knowledge of the risks of antiretroviral
therapy has changed the approach of many clinicians towards
treatment over the last three years. By the year 2000, many
strict recommendations from previous years were already being
revised. “Hit HIV hard, but only when necessary” is now heard
more than “hit hard and early” (Harrington and Carpenter
2000). The simple question of “when to start?” is now being
addressed at long symposia. It is often a question that requires
great sensitivity.
Despite all the skepticism, it is important not to forget what
HAART can do. HAART can often achieve miracles! Crypto-
sporidia and Kaposi's sarcoma simply disappear; PML may
even be cured completely; secondary prophylaxis for CMV can
be stopped; and above all: patients feel significantly better, even
if some activists and AIDS counselors still do not want to admit
this.
This also means, however, that many younger clinicians in
Western countries who entered into HIV medicine at the end of
the 90s often no longer know what AIDS really means. AIDS
for them is an accident, whose damage can be fixed. They did
not experience the "stone age" of AIDS.
HIV clinicians are well advised, perhaps more than other clini-
cians, to remember the "stone age", whilst still keeping an open
1. Perspective 59
Hoffmann, Kamps, et al.
mind for new approaches. Those, who are strictly opposed to
the interruption of treatment, and insistent on particular sche-
mata of treatment, are not only overlooking the realities of
treatment, but also losing touch. Those, who do not make an
effort to broaden their knowledge several times a year at differ-
ent conferences, will not be able to provide adequate treatment
for their patients in a field that changes direction at least every
two to three years. Those, who adhere strictly to evidence-based
HIV medicine, and only treat according to guidelines, quickly
become outdated. HIV medicine is ever changing. Treatment
guidelines remain just guidelines. They are often out of date by
the time of publication. There are no laws set in stone. How-
ever, those, who confuse therapeutic freedom with random
choices and assume that data and results coming from basic re-
search can be ignored, are also missing the point. Individualized
treatment is not random treatment. In addition, it cannot be
stressed enough, that clinicians are also responsible for the
problem of bad compliance. Even if many experienced clini-
cians have come to disregard this: every patient has the right to
know why he is taking which therapy or, indeed, why it has
been omitted.
HIV remains a dangerous and cunning opponent. Patients and
clinicians must tackle it together. The following describes how
this can be done.
References
1. Ault A. FDA warns of potential protease-inhibitor link to hyperglycaemia.
Lancet 1997, 349:1819.
2. Brinkman K, Smeitink JA, Romijn JA, Reiss P. Mitochondrial toxicity in-
duced by nucleoside-analogue reverse-transcriptase inhibitors is a key
factor in the pathogenesis of antiretroviral-therapy-related lipodystrophy.
Lancet 1999, 354:1112-5. http://amedeo.com/lit.php?id=10509516
3. Brodt HR, Kamps BS, Gute P, et al. Changing incidence of AIDS-defining
illnesses in the era of antiretroviral combination therapy. AIDS 1997,
11:1731-8. http://amedeo.com/lit.php?id=9386808
4. Cameron DW, Heath-Chiozzi M, Danner S, et al. Randomised placebo-
controlled trial of ritonavir in advanced HIV-1 disease. Lancet 1998,
351:543-9. http://amedeo.com/lit.php?id=9492772
60 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
5. Concorde: MRC/ANRS randomised double-blind controlled trial of imme-
diate and deferred zidovudine in symptom-free HIV infection. Lancet
1994, 343:871-81. http://amedeo.com/lit.php?id=7908356
6. Consensus Statement on Antiretroviral Treatment for AIDS in Poor
Countries by Individual Members of the Faculty of Harvard University.
2001. http://hiv.net/link.php?id=182
7. Delta: a randomised double-blind controlled trial comparing combinations
of zidovudine plus didanosine or zalcitabine with zidovudine alone in HIV-
infected individuals. Lancet 1996, 348: 283-91.
http://amedeo.com/lit.php?id=8709686
8. Gulick RM, Mellors JW, Havlir D, et al. 3-year suppression of HIV viremia
with indinavir, zidovudine, and lamivudine. Ann Intern Med 2000, 133:35-
9. http://amedeo.com/lit.php?id=10877738
9. Hamilton JD, Hartigan PM, Simberkoff MS, et al. A controlled trial of early
versus late treatment with zidovudine in symptomatic HIV infection. N
Engl J Med 1992, 326:437-43. http://amedeo.com/lit.php?id=1346337
10. Hammer SM, Katzenstein DA, Hughes MD et al. A trial comparing nu-
cleoside monotherapy with combination therapy in HIV-infected adults
with CD4 cell counts from 200 to 500 per cubic millimeter. N Engl J Med
1996, 335:1081-90. http://amedeo.com/lit.php?id=8813038
11. Harrington M, Carpenter CC. Hit HIV-1 hard, but only when necessary.
Lancet 2000, 355:2147-52. http://amedeo.com/lit.php?id=10902643
12. Ho DD. Time to hit HIV, early and hard. N Engl J Med 1995, 333:450-1.
13. Ho DD, Neumann AU, Perelson AS, Chen W, Leonard JM, Markowitz M.
Rapid turnover of plasma virions and CD4 lymphocytes in HIV-1 infection.
Nature 1995, 373:123-6. http://amedeo.com/lit.php?id=7816094
14. Kirk O, Mocroft A, Katzenstein TL, et al. Changes in use of antiretroviral
therapy in regions of Europe over time. AIDS 1998, 12: 2031-9.
http://amedeo.com/lit.php?id=9814872
15. Mocroft A, Katlama C, Johnson AM, et al. AIDS across Europe, 1994-98:
the EuroSIDA study. Lancet 2000, 356:291-6.
http://amedeo.com/lit.php?id=11071184.
16. Perelson AS, Neumann AU, Markowitz M, Leonard JM, Ho DD. HIV-1
dynamics in vivo: virion clearance rate, infected cell life-span, and viral
generation time. Science 1996, 271:1582-6.
http://amedeo.com/lit.php?id=8599114
17. Volberding PA, Lagakos SW, Koch MA, et al. Zidovudine in asympto-
matic HIV infection. A controlled trial in persons with fewer than 500 CD4-
positive cells per cubic millimeter. N Engl J Med 1990, 322:941-9.
http://amedeo.com/lit.php?id=1969115
2. Overview of Antiretroviral Drugs 61
Hoffmann, Kamps, et al.
2. Overview of Antiretroviral Drugs
Christian Hoffmann
Table 2.1: Antiretroviral agents
Nucleoside and Nucleotide Reverse Transcriptase Inhibitors (NRTIs)
Trade name Abb. Drug Manufacturer
Combivir
®
AZT+3TC GSK
Epivir
®
3TC Lamivudine GSK
Hivid
®
ddC Zalcitabine Roche
Retrovir
®
AZT Zidovudine GSK
Trizivir
®
AZT+3TC+ABC GSK
Videx
®
ddI Didanosine BMS
Viread
®
TDF Tenofovir Gilead
Zerit
®
d4T Stavudine BMS
Ziagen
®
ABC Abacavir GSK
Non-Nucleoside Reverse Transcriptase Inhibitors (NNRTIs)
Rescriptor
®
DLV Delavirdine Pfizer
Sustiva
®
, Stocrin
®
EFV Efavirenz BMS
Viramune
®
NVP Nevirapine Boehringer
Ingelheim
Protease Inhibitors (PIs)
Agenerase
®
APV Amprenavir GSK
Crixivan
®
IDV Indinavir MSD
Fortovase
®
SQV-SGC Saquinavir soft gel Roche
Invirase
®
SQV-HGC Saquinavir hard gel Roche
Kaletra
®
LPV Lopinavir/ Ritonavir Abbott
Norvir
®
RTV Ritonavir Abbott
Viracept
®
NFV Nelfinavir Roche
Three classes of antiretroviral agents are currently available for
the treatment of HIV infection: nucleoside and nucleotide ana-
logs (NRTIs), non-nucleoside reverse transcriptase inhibitors
(NNRTIs) and protease inhibitors (PIs). Some 20 drug products
have been licensed, including formulations of both individual
and combined antiretroviral agents. The fusion inhibitor T-20
was launched in March 2003 as the prototype of a fourth drug
class. A number of other drugs and new classes of drugs are in
62 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
the pipeline and expected to be licensed in the next years. Re-
search is also focusing on immunomodulatory approaches with
vaccines or cytokines (interferons, interleukins).
The following overview will deal mainly with the individual
antiretroviral agents and their specific features and problems.
Common combinations are described in the chapter on “How to
start HAART?”.
Nucleoside Analogs (“nukes”, NRTIs)
Mechanism of action
Nucleoside analogs (slang: “nukes”) are also referred to as nu-
cleoside reverse transcriptase inhibitors. Their target is the HIV
enzyme reverse transcriptase. Acting as alternative substrates or
“false building blocks“, they compete with physiological nu-
cleosides, differing from these only by a minor modification in
the sugar (ribose) molecule. The incorporation of nucleoside
analogs aborts DNA synthesis, as phosphodiester bridges can
no longer be built to stabilize the double strand.
Nucleoside analogs are converted to the active metabolite only
after endocytosis, whereby they are phosphorylated to triphos-
phate derivatives. AZT and d4T are thymidine analogs, ddC and
3TC are cytidine analogs. A combination of AZT and d4T
would be senseless, since both drugs compete for the same
bases; this also applies to ddC and 3TC. ddI is an inosine ana-
log, which is converted to dideoxyadenosine; abacavir is a gua-
nine analog. There is a high degree of cross-resistance between
nucleoside analogs (see also the chapter on “Resistance“).
Nucleoside analogs are important components of almost all
combination regimens. They are potent inhibitors of HIV repli-
cation, and are rapidly absorbed when taken orally. However,
they can cause a wide spectrum of side effects, encompassing
myelotoxicity, lactic acidosis, polyneuropathy and pancreatitis.
Complaints include fatigue, headache and a variety of gastroin-
testinal problems such as abdominal discomfort, nausea, vom-
iting and diarrhea. Although lipodystrophy was initially linked
2. Overview of Antiretroviral Drugs 63
Hoffmann, Kamps, et al.
exclusively to treatment with protease inhibitors, numerous dis-
orders of lipid metabolism (especially lipoatrophy) are now also
attributed to nucleoside analogs (Galli et al. 2002).
Most side effects are probably related to mitochondrial toxicity,
first described in 1999 (Brinkmann et al. 1999). Mitochondrial
function also requires nucleosides. The metabolism of these
important organelles is disrupted by the incorporation of false
nucleosides, leading to mitochondrial degeneration. More re-
cent clinical and scientific data indicates that there are probably
significant differences between individual drugs with regard to
mitochondrial toxicity.
Nucleoside analogs are eliminated mainly by renal excretion
and do not interact with drugs that are metabolized by hepatic
enzymes. There is therefore little potential for interaction. How-
ever, substances such as ribavirin may decrease the intracellular
phosphorylation of AZT or d4T in vitro (Piscitelli et Galliciano
2001).
Individual agents: Special features and problems
Abacavir (Ziagen
®
) is a potent and mostly well-tolerated nu-
cleoside analog with good CNS penetration. One drawback to
the use of abacavir is the occurrence of the hypersensitivity re-
action (HSR), which is not yet fully understood. HSR occurs in
approximately 4-5% of patients, almost always (93%) within
the first six weeks of treatment. Every treating physician should
be familiar with this syndrome, which can be fatal in individual
cases, especially after rechallenge (see Management of Side
Effects). The combination of strongly worded warnings con-
tained in the package insert and the often unspecific symptoms
of the HSR poses a constant challenge to the patient-physician
relationship. Several reports were published in 2002 suggesting
that patients with HLA type B5701 may be genetically predis-
posed and at higher risk than others (Mallal et al. 2002, Heth-
erington et al. 2002). Apart from HSR, abacavir seems to have
an otherwise favorable long-term profile, especially in terms of
mitochondrial toxicity (Carr et al. 2002).
64 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
AZT – Zidovudine (Retrovir
®
) was the first antiretroviral
agent to be put on the market, in 1987. In the first few years, it
was administered in doses that were too high, which led to sig-
nificant myelotoxicity and brought the drug into somewhat of
disrepute. Even with the standard doses given today, monitoring
of blood count is obligatory. Long-term treatment almost al-
ways increases MCV. Initial gastrointestinal complaints may
present a short-term problem. AZT seems to have a more favor-
able profile with regard to long-term toxicity. Lack of neuro-
toxicity and good CNS penetration are important advantages of
this drug, which has remained the cornerstone of many HAART
regimens and transmission prophylaxis.
ddC - Zalcitabine (Hivid
®
) was investigated closely in the
double nuke studies of the early to mid-nineties. It has since
been marginalized due to the relatively frequent development of
peripheral neuropathy, the three times daily dosing requirement,
and lack of data in the HAART era. At the present time, ddC is
by far the least used nucleoside analog. Stomatitis is a side ef-
fect that is relatively specific for ddC. Although a twice daily
dose now seems possible (Moyle and Gazzard 1998), increased
competition from newer nucleoside analogs may mean that this
substance will disappear from antiretroviral therapies.
ddI – Didanosine (Videx
®
) is a nucleoside analog that has been
well investigated and shown good efficacy in numerous ran-
domized studies. The introduction of acid-resistant tablets in
2000, to replace the chewable tablets used for many years, has
done much to improve tolerability. ddI remains one of the most
important components of many HAART regimens. ddI was
shown to be more potent than AZT, even with regard to disease
progression in the ACTG 175 Study (Hammer et al. 1996), con-
firming results of an earlier study (Kahn et al. 1992). After fail-
ure with AZT, ddI is probably more effective than d4T (Havlir
et al. 2001). Gastrointestinal complaints are typical and rela-
tively frequent side effects. Pancreatitis, a less common but also
typical adverse effect, may be fatal in individual cases and is
possibly dose-related. Special caution should be given to the
2. Overview of Antiretroviral Drugs 65
Hoffmann, Kamps, et al.
combination with d4T and hydroxyurea (Havlir at al. 2001).
The advantage to the use of ddI of simple once daily dosing,
which is possible due to the long intracellular half-life, is coun-
terbalanced by the need to take the drug under fasting condi-
tions.
d4T – Stavudine (Zerit
®
) was the second thymidine analog to
be introduced after AZT. On initiation of therapy it is often
better tolerated than AZT, producing less gastrointestinal side-
effects and limited myelotoxicity. It is definitely just as effec-
tive and was for many years the most frequently prescribed
antiretroviral agent. Recently, focus on long-term toxicity rather
than efficacy has revealed that d4T seems to be associated with
more problems than other nucleoside analogs. It increases the
risk of lactic acidosis and hyperlactacidemia, especially in com-
bination with ddI or 3TC (Gerard et al. 2000, Miller at al. 2000,
Mokrzycki et al. 2000, John et al. 2001). There has also been
concern over recent reports of progressive neuromuscular
weakness. 22 of 25 patients (7 fatal cases), presenting with
symptoms similar to the Guillain-Barré syndrome and with hy-
perlactacidemia, had received d4T, 11 of these d4T+ddI (Mar-
cus et al. 2002). Lipodystrophy is probably also more frequent
with d4T. In a German cohort the risk of lipoatrophy had dou-
bled after one year of treatment (Mauss et al. 2002); in a Swiss
cohort it had tripled after two years (Bernasconi et al. 2002).
Other data, with one exception (Bogner et al. 2001), points in
the same direction (Chene et al. 2002).
Even more significant than the data from cohort studies is the
publication of the first studies showing the positive effect on
lipoatrophy of discontinuation of d4T (and replacement with
other nukes): In a randomized study from Australia, in which
111 lipoatrophic patients on stable HAART had d4T or AZT
replaced either with abacavir or not, most benefit was seen in
the d4T group (Carr et al. 2002). The effect at 24 weeks, how-
ever, was still very moderate. The increased subcutaneous fat
tissue detectable by dexa scan was not visible clinically. It may
therefore take years, as the authors concluded, for lipoatrophy
66 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
to visibly improve after discontinuation of d4T. A positive ef-
fect, albeit once again weak, has been described in two further
d4T-replacement studies (John et al. 2002, McGomsey et al.
2002). Thus, bearing resistance patterns in mind, in patients on
d4T with severe lipoatrophy, the drug should be replaced, opti-
mally with abacavir. There is, however, no assurance for reso-
lution of lipoatrophy, and, above all, great patience is required.
3TC – Lamivudine (Epivir
®
) is a very well tolerated nucleo-
side analog. This substance is frequently used, as it is a compo-
nent of both Combivir and Trizivir. Its main disadvantage is
rapid development of resistance, and a single point mutation
(M184V) is sufficient for loss of effectiveness. Since resistance
is likely to develop after only a few weeks, 3TC has practically
no effect as monotherapy. Thus, treatment with 3TC as the only
nucleoside analog component of a combination is considered
problematic. As the M184V mutation seems to impair viral fit-
ness, however, continuation of 3TC therapy following this mu-
tation may make good sense (Miller et al. 2002).
3TC is also effective against hepatitis B viruses. Once daily
dosing appears to be feasible (Sension et al. 2002). In the US,
3TC has already been approved as the first once-daily nucleo-
side analog.
Tenofovir (Viread
®
) acts as a false building block similarly to
nucleoside analogs, targeting the enzyme reverse transcriptase.
However, in addition to the pentose and nucleic base it is
monophosphorylated, and is therefore referred to as a nucleo-
tide analog. The more accurate description of the substance is
tenofovir DF (disoproxil fumarate), which is a phosphonate
from which the phosphonate component is only removed by a
serum esterase and which is activated intracellularly in two
phosphorylation steps (Robbins et al. 1998).
After the first nucleotide analog adefovir was abandoned in
HIV therapy due to weak antiviral activity and severe side ef-
fects (and is now being further developed in lower doses as the
hepatitis B medication Hepsera
®
), tenofovir has shown mark-
2. Overview of Antiretroviral Drugs 67
Hoffmann, Kamps, et al.
edly improved tolerability and would also appear to be more
potent. In the 902 Study, in which tenofovir versus placebo was
added to HAART, tenofovir decreased viral load by 0.62 log
after 48 weeks (Schooley et al. 2002). The 903 Study was a
double-blind study in which treatment-naive patients were
given tenofovir or d4T (in addition to the backbone regimen
with 3TC and efavirenz). Preliminary results showed at least
equivalent potency (Staszewski et al. 2002). Tolerability was
higher in the tenofovir group, especially with regard to polyneu-
ropathy and fat redistribution. This is consistent with in vitro
data, which shows that phosphorylated tenofovir has a low af-
finity for mitochondrial polymerases (Suo 1998).
Despite all the positive reports, long-term data on tenofovir are
not yet available. In combination with ddI, there are increased
levels of ddI, which could lead to increased toxicity (Kearney et
al. 2002); a daily dose reduction of ddI to 250 mg is probably
necessary. On the other hand, since tenofovir is eliminated re-
nally, interactions with substances metabolized in the liver are
rare. Longterm, the possibility of cumulative nephrotoxicity
needs to be clarified.
Efficacy – Which nuke backbone is best?
All classical HAART regimens contain two nucleoside analogs
as the “backbone” of treatment. For many years, numerous
studies, especially before the introduction of PIs and NNRTIs,
concentrated on the optimal combination of two nucleoside
analogs.
There are probably no great differences. Although data has been
contradictory, this is probably due to different study settings
and frequently heterogeneous patient populations. There seems
only to be consensus that ddC-containing nuke combinations
are slightly less potent. A meta-analysis of several randomized
studies has shown that AZT+ddI is more potent than AZT+ddC
(HTCG 1999). Similarly, in patients pretreated with monother-
apy, AZT+3TC was superior to AZT+ddC (Bartlett et al. 1996).
68 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
AZT+3TC or d4T+ddI?
A great deal of data is now available comparing the two most
frequent combinations – AZT+3TC and d4T+ddI. In the French
Albi Trial, d4T+ddI was clearly more effective than AZT+3TC.
However, it was later shown that d4T+ddI significantly caused
more frequent lipoatrophy (Molina et al. 1999, Chene et al.
2002), and following failure of d4T+ddI, AZT resistance was
found to be equal or more than that with AZT+3TC (Picard et
al. 2001). The combination with indinavir also showed a posi-
tive trend in favor of d4T+ddI over AZT+3TC (Eron et al.
2000).
These results, however, were not confirmed in another study
(Carr et al. 2000). Similarly, no difference in efficacy was
found between d4T+ddI, AZT+3TC and d4T+3TC, whether in
combination with nevirapine or indinavir (Foudraine et al.
1998, Squire et al. 2000, French et al. 2002).
Although ACTG 384, the ultimate large study dealing with this
issue has yet to be completed, the pendulum seems to have
swung in favor of AZT+3TC. Preliminary results, presented
recently at the World AIDS Conference in Barcelona (Robbins
et al. 2002, Shafer et al. 2002), were puzzling: AZT+3TC is
virologically superior to d4T+ddI, although only in combination
with efavirenz; a combination with nelfinavir shows no added
benefit. A plausible explanation has yet to be given.
Summary of nuke backbones
To date, the results of efficacy studies remain inconclusive and
do not provide a mandate for the choice of one particular com-
bination over another. Treatment can thus be adapted to the
particular needs of each patient.
Choice of one of the three combinations AZT+3TC, AZT+ddI
or d4T+3TC is nearly always appropriate. In view of recent
studies on lactic acidosis and lipoatrophy, the combination of
d4T+ddI should be carefully considered and monitored.
2. Overview of Antiretroviral Drugs 69
Hoffmann, Kamps, et al.
Other combinations such as AZT+ABC, d4T+ABC,
ABC+3TC, or ddI+3TC also seem acceptable, but are not as
well supported by clinical data. ddI+3TC may also produce less
favorable results than AZT+3TC or d4T+3TC, as was suggested
by the ACTG 306 Study (Kuritzkes et al. 1999).
Combinations such as AZT+d4T, ddC+3TC, d4T+ddC and
ddI+ddC should definitely be avoided. It has also been shown
that constant changing of the nuke backbone with the goal of
preventing development of resistance has no positive effect and
probably only confuses the patient (Molina et al. 1999).
Non-Nucleoside Reverse Transcriptase Inhibitors
(NNRTIs)
Mechanism of action and efficacy
As with the nucleoside analogs, the target enzyme of NNRTIs is
reverse transcriptase. NNRTIs were first described in 1990. In
contrast to the NRTIs, they are not “false” building blocks, but
rather bind directly and non-competitively to the enzyme, at a
position in close proximity to the substrate-binding site for nu-
cleosides. The resulting complex blocks the catalyst-activated
binding site of the reverse transcriptase, which can thus bind
fewer nucleosides, and polymerization is slowed down signifi-
cantly. In contrast to NRTIs, NNRTIs do not require activation
within the cell.
The three currently available NNRTIs – nevirapine, delavirdine
and efavirenz – were introduced between 1996 and 1998. Hav-
ing only limited potency as individual agents, they were ini-
tially regarded somewhat skeptically. Although studies such as
the INCAS Trial or Protocol 0021II clearly demonstrated the
superiority of triple therapy with nevirapine or delavirdine
compared to double nuke therapy (Conway et al. 2000), the
“rise” of the NNRTIs was rather hesitant, and did not receive
the media attention given to that of the PIs.
Since then, both randomized and large cohort studies have
demonstrated that NNRTIs are extremely effective in combina-
70 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
tion with nucleoside analogs. The immunological and virologi-
cal potency of NNRTIs is at least equivalent to that of PIs
(Friedl et al. 2001, Staszewski et al. 1999, Torre et al. 2001). In
contrast to PIs, however, the clinical effect has not yet been
proven, as the studies that led to licensing of NNRTIs all used
surrogate markers. Nevertheless, the simple dosage and the
overall good tolerability have led nevirapine and efavirenz to
become important components of HAART regimens, which are
often even ranked above those containing PIs. While the manu-
facturers of nevirapine and efavirenz compete for market domi-
nation, delavirdine has lost relevance (a situation which is un-
likely to change).
To date, no controlled study provides clear evidence that one
NNRTI is more potent than another. A small, randomized pilot
study from Spain demonstrated no significant differences be-
tween nevirapine and efavirenz (Nunez et al. 2002). However,
several cohort studies indicate the superiority of efavirenz. In an
Italian study, treatment failure on nevirapine was 2.08 times
more likely than on efavirenz (Cozzi-Lepri et al. 2002), and in
the Euro-SIDA study this factor was 1.75 (Phillips et al. 2001).
Such analyses should be interpreted with caution, as extremely
heterogeneous patient groups, with varying previous treatments,
were studied. This was recently underlined by the eagerly
awaited results of the 2NN Study ("The Double Non-
Nucleoside Study"). 2NN is the first large-scale randomized
trial directly comparing nevirapine and efavirenz-containing
regimens in HAART-naive patients. The trial showed that nevi-
rapine and efavirenz were comparable with respect to virologi-
cal and immunological efficacy after 48 weeks of therapy.
However, nevirapine and efavirenz have distinctive adverse
event profiles which should be considered in the choice of these
drugs (see below).
In the case of both NNRTIs, efficacy and toxicity probably cor-
relate with plasma levels (Veldkamp et al. 2001, Marzolini et al.
2001, Gonzalez et al 2002). Nevirapine and efavirenz are me-
tabolized by cytochrome P450 enzymes (Miller et al. 1997).
2. Overview of Antiretroviral Drugs 71
Hoffmann, Kamps, et al.
Nevirapine is an inductor, whereas efavirenz is both an inductor
and an inhibitor of the cytochrome P450 isoenzyme. The com-
bination of efavirenz with saquinavir or lopinavir leads to
strong interactions that require dose adjustments.
Individual agents: Special features and problems
The most significant problem with NNRTIs is resistance, with a
high risk of cross-resistance. One point mutation on position
103 (K103N) of the hydrophobic binding site is sufficient to
eliminate an entire class of drug. Point mutations may occur
very rapidly. Resistance has even been described in maternal
transmission prophylaxis, in mothers who had taken nevirapine
only once during delivery (Eshleman et al. 2002). Thus,
NNRTI-containing regimens are vulnerable – and waiting too
long to switch therapy during insufficient suppression of viral
load almost certainly leads to complete resistance.
The side effects of nevirapine and efavirenz are quite different,
and should be considered in the choice of regimen.
Nevirapine (Viramune
®
) was the first licensed NNRTI. In rare
cases, it may cause serious hepatic toxicity. To prevent allergic
reactions, the currently recommended dosage regimen is 200
mg qd for 2 weeks, followed by 200 mg bid thereafter. During
the first 8 weeks, biweekly monitoring of liver function tests is
recommended. A rash develops in 15-20 % of cases and leads
to discontinuation in 7% of patients (Miller et al. 1997). In the
case of an isolated rash or isolated elevation of transaminases
(up to five times the upper limit of normal), treatment may usu-
ally be continued. However, treatment should be discontinued
in the case of a rash with even slightly elevated transaminases
(>2-fold of norm). Patients with chronic hepatitis are probably
at a higher risk (Sulkowski et al. 2000). Similarly, there seems
to be a correlation with plasma levels (Gonzalez et al. 2002). It
is important to note that hepatic toxicity may occur even after
several months (Sulkowski et al. 2002).
In contrast, nevirapine has a good lipid profile. In the Atlantic
Study, in which nevirapine was tested against lamivudine and
72 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
indinavir, with all groups on a d4T+ddI backbone, those re-
ceiving nevirapine showed favorable lipid changes for choles-
terol and triglycerides. Astonishingly, there was an increase in
HDL (Van der Valk et al. 2001), demonstrated also in the
Spanish Lipnefa Study (Fisac et al. 2002). These effects are un-
doubtedly positive. Whether they will have clinical relevance
over time, remains to be seen.
Efavirenz (Sustiva
®
, Stocrin
®
) was the third NNRTI to be ap-
proved and the first in which it was demonstrated that NNRTIs
were at least as effective as PIs (Staszewski et al. 1999). The
long half-life allows for once-daily dosing. With the approval of
a new 600 mg capsule, dosage has been reduced to a single cap-
sule per day.
Efavirenz may cause mild CNS side effects and should there-
fore be taken in the evening. These disorders usually include
morning dizziness and somnolence; nightmares may also occur.
The side effects probably correlate with high plasma levels
(Marzolini et al. 2001). In one study, after four weeks of treat-
ment with efavirenz, 66 % of patients complained of dizziness,
48 % of abnormal dreams, 37 % of somnolence and 35 % of
insomnia. Although these symptoms seem to resolve with fur-
ther treatment (frequencies of these complaints at 24 weeks was
only 13 %, 18 %, 13 % and 7 %, respectively), patients must be
warned of these potential side effects (Fumaz et al. 2002). To
date, little is known of the effect on driving. We recommend
that efavirenz should not be prescribed to patients during ex-
amination periods, to pilots or crane operators. Patients with
impaired concentration should avoid potentially hazardous ac-
tivities such as driving or operating heavy machinery (see pack-
age insert). Efavirenz is contraindicated in pregnancy. Lipids
are not as favorably affected as with nevirapine (Hoffmann et
al. 2000), but hepatotoxicity is less frequent.
Delavirdine (Rescriptor
®
): Due to a high pill burden and the
required three times daily dosing, delavirdine is currently rarely
prescribed, although it is likely to be approximately as effective
as nevirapine and efavirenz (Wood et al. 1999, Conway 2000).
2. Overview of Antiretroviral Drugs 73
Hoffmann, Kamps, et al.
The need for prescription should be carefully considered. In
1999, an application for licensure in Europe was rejected due to
insufficient efficacy data.
Protease Inhibitors (PIs)
Mechanism of action and efficacy
The HIV protease cuts the viral gag-pol polyprotein into its
functional subunits. Inhibition of the protease, preventing pro-
teolytic splicing and maturation, leads to the release of virus
particles which are unable to infect new cells. With knowledge
of the molecular structure of the protease encoded by the virus,
the first protease inhibitors were designed in the early nineties;
these substances were modified in such a way, that they fit ex-
actly into the enzyme active site of the HIV protease (detailed
reviews: Deeks 1997, Somadossi 1999, Eron 2001).
Since 1995, protease inhibitors have revolutionized the treat-
ment of HIV infection (see also the chapter on “History”). At
least three large studies with clinical endpoints proved the effi-
cacy of indinavir, ritonavir and saquinavir (Hammer et al. 1997,
Cameron et al. 1998, Stellbrink et al. 2000). Even if in recent
years PIs have demonstrated a series of drawbacks, they remain
an essential component of HAART, especially for treatment-
experienced patients.
As with the NNRTIs, there has been intense pharmaceutical
company competition to establish which PI has superior effi-
cacy. However, comparative studies have failed to demonstrate
clear superiority of one protease inhibitor over any other.
Two exceptions have to be mentioned: the hard gel capsule
saquinavir-HGC and ritonavir. A large retrospective analysis
has shown the relative benefit of indinavir when compared to
saquinavir-HGC. There was significantly less virologic failure
in patients taking indinavir (Fätkenheuer et al. 1997). In the
Euro-SIDA cohort there was even a clinical benefit of indinavir
when compared to saquinavir hard gel capsules (Kirk et al.
2001). Saquinavir was subsequently “rescued” mainly by
74 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
boosting (see below), but also by the development of soft gel
capsules with improved resorption. A small, randomized study
showed no differences between indinavir, saquinavir soft gel
capsules, ritonavir, nelfinavir and amprenavir, when combined
with abacavir (McMahon et al. 2001). Similarly, the CHEESE
Study found no differences between saquinavir-SGC and indi-
navir (Cohen et al. 1999).
In the case of ritonavir, the main problem is poor tolerability. In
an open-label randomized trial with three groups, although no
major differences could be shown between ritonavir/saquinavir
and indinavir-containing regimens, but there was a definite un-
favorable trend for patients on ritonavir, due not to virologic
failure but to frequent discontinuation because of side effects
(Katzenstein et al. 2000).
Boosted PI regimens are presumably more effective. Lopina-
vir/r in combination with d4T/3TC was shown to reduce viral
load more effectively than nelfinavir-containing HAART. After
one year in the double-blind M98-863 Study, 67 % versus 52 %
had a viral load below 50 copies/ml (Walmsley et al. 2002).
Individual agents: Special features and problems
Apart from gastrointestinal side effects and high pill burden, all
PIs used in long-term therapy can be implicated in lipodystro-
phy and dyslipidemia (see also the chapter on “Lipodystrophy”;
review in Graham 2000). Smaller randomized studies have
shown that elevation of lipid levels is more pronounced in rito-
navir-containing regimens than with saquinavir or nelfinavir
(Roge et al. 2001, Wensing et al. 2001). In addition, there may
be significant drug interactions with ritonavir and with boosted
regimens. Sexual dysfunction has also been attributed to PIs
(Schrooten et al. 2001), although data is inconclusive (Lalle-
mand et al. 2002).
There is a high degree of cross-resistance between protease in-
hibitors, which was described even before PIs were put on the
market (Condra et al. 1995; see also the chapter on “Resis-
tance”). All PIs are inhibitors of the CYP3A4 system and inter-
2. Overview of Antiretroviral Drugs 75
Hoffmann, Kamps, et al.
act with numerous other drugs. Ritonavir is by far the strongest
inhibitor, saquinavir probably the weakest.
Amprenavir (Agenerase
®
) – As an unboosted PI, the sub-
stance is hardly acceptable today due to the high pill burden
(8 pills BID). Important side effects include gastrointestinal
disorders and, in contrast to other PIs, occasional rashes.
Whether the incidence of lipodystrophy and dyslipidemia is
reduced as compared with other PIs has yet to be proven (Noble
et al. 2000). The resistance profile of the drug is particularly
interesting, as it only partially overlaps with that of other PIs. It
is to be expected that Agenerase
®
will be removed from the
market as soon as the follow-on drug fos-amprenavir is avail-
able. This could lead to serious competition for lopinavir in the
area of salvage therapy.
Indinavir (Crixivan
®
) has been shown to be a very effective PI
in numerous studies; it is probably the most extensively tested
(Gulick et al. 1997, Hammer et al. 1997). The large amount of
data is currently the most important argument in favor of this
drug. Low protein binding (60 %) seems to allow better CNS
penetration than with other PIs (Martin et al. 1999). Whether
this is clinically significant remains to be seen.
There are, however, a number of problems associated with indi-
navir. First, it causes nephrolithiasis in approximately 5-10 % of
patients and thus requires good hydration (at least 1.5 liters
daily). Patients with a history of nephrolithiasis or renal insuffi-
ciency should therefore not receive indinavir. Secondly, in the
unboosted form, indinavir must be taken three times daily in
fasting conditions, a form of dosing that is currently unaccept-
able. Finally, the minimal inhibitory concentration is often
reached as soon as 8 hours after administration.
Unboosted twice-daily dosing is not possible. A trial using 2 x
1200 mg indinavir (3 tablets BID) in 87 patients had to be
stopped because of 36 % versus 9 % treatment failures in the
study group with twice-daily dosing (Haas et al. 2000). For this
reason, indinavir is increasingly being used in combination with
76 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
boosting doses of ritonavir. Such boosting may, however, pres-
ent some problems due to side effects (Gatell et al. 2000, Harley
et al. 2001, Shulman et al. 2002). In the MaxCmin1 Trial the
drop-out rate in the indinavir group was clearly higher than
among patients receiving saquinavir (Gerstoft et al. 2002).
There are relatively frequent mucocutaneous side effects, remi-
niscent of retinoid therapy: alopecia, dry skin and lips, ingrown
nails. Some patients may also develop asymptomatic hyperbili-
rubinemia.
Lopinavir/Ritonavir (Lopinavir/r, Kaletra
®
) is the newest PI
and the first to contain a fixed booster dose of ritonavir, which
may increase concentrations of lopinavir by more than 100-fold
(Sham et al. 1998). Lopinavir has the highest genetic barrier of
all PIs (6-8 cumulative PI resistance mutations are probably
necessary for treatment failure), and it has surprising efficacy in
salvage therapy. However, use in early treatment is controver-
sial, and to date it has also not been shown whether lopinavir is
the most effective PI for treatment-naive patients. It is probably
superior to nelfinavir (and also atazanavir), but as yet there is
no data for boosted PIs such as indinavir, saquinavir or ampre-
navir. Dyslipidemia appears to be a significant problem with
lopinavir therapy.
Nelfinavir (Viracept
®
) was the fourth PI to be put on the mar-
ket and was for a long time one of the most frequently used PIs.
Although it is also licensed for the (initially developed) dose of
3 x 3 capsules, nelfinavir may be taken twice daily in the dose
of 2 x 5 capsules. Boosting with ritonavir does not lead to sig-
nificant improvement in plasma levels.
The most frequent side effect of nelfinavir is diarrhea, which
may be quite severe. The antiretroviral potency of nelfinavir is
weaker than that of boosted PIs (Walmsley et al. 2002). In the
large Agouron 511 Study which led to licensing, 61 % of pa-
tients (with an AZT+3TC backbone) showed blood levels under
50 copies/ml at 48 weeks (Saag et al. 2001). The substance has
a good resistance profile. The D30N primary mutation for nel-
finavir reduces viral fitness (Martinez et al. 1999) and does not
2. Overview of Antiretroviral Drugs 77
Hoffmann, Kamps, et al.
influence the efficacy of other PIs. Unfortunately however,
other mutations, which in contrast can jeopardize the success of
later regimens, also occur frequently. A new formulation ena-
bling a reduction to 2 x 2 capsules daily is in development,
which may reverse the recent downward trend in sales of nelfi-
navir due to strong competition.
Ritonavir (Norvir
®
) was the first PI for which efficacy was
proven with clinical endpoints (Cameron et al. 1998). Due to its
poor tolerability (gastrointestinal complaints, perioral paresthe-
sias), ritonavir is generally no longer used as a single PI. How-
ever, when used to boost other protease inhibitors, the ritonavir
dose can be reduced to 2 x 100 mg, whereby toleration is vastly
improved. Ritonavir is a potent inhibitor of the cytochrome
P450 pathway with a great potential for interactions with other
drugs; thus, many drugs are contraindicated for concomitant
administration with ritonavir. Metabolic disorders are probably
more frequent than with other PIs. Caution should generally be
exercised in patients with impaired liver function. It is impor-
tant to inform patients that ritonavir capsules must be stored at
cool temperatures, which can become a problem during long
trips.
Saquinavir (Invirase
®
and Fortovase
®
) is the only PI which is
available in two formulations: a hard gel capsule (Invirase
®
or
saquinavir-HGC) and a soft gel capsule (Fortovase
®
or saquina-
vir-SGC). The soft gel capsules have a greatly improved bio-
availability and therefore a superior antiviral activity, which
was demonstrated in a pilot study in naive patients (Mitsuyasu
et al. 1998). However, in the era of boosting with ritonavir, this
distinction is probably less relevant (see below).
Saquinavir was the first PI to be licensed in December 1995 for
HIV therapy. Although rare serious side effects can occur, the
main adverse reactions are relatively mild gastrointestinal com-
plaints, which are more frequent with the soft gel capsules (Ku-
rowski et al. 2002). Saquinavir is otherwise well tolerated. In
the MaxCmin1 Trial, the drop-out rate was significantly lower
when compared to the indinavir group (Gerstoft et al. 2002).
78 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
Why “boost” PI regimens?
Ritonavir is a very potent inhibitor of the isoenzyme 3A4, a
subunit of the cytochrome P450 hepatic enzyme system, and
small doses of ritonavir lead to increased plasma levels (boost-
ing) of almost all PIs (Kempf et al. 1997). Indeed, nelfinavir is
the only drug for which boosting with ritonavir is not recom-
mended, as plasma levels do not rise significantly (Kurowski et
al. 2002).
The interaction between ritonavir and the other PIs simplifies
the daily regimen by reducing the number of pills to be taken
every day. Some PIs can now be used in twice-daily regimens.
Recent trials are investigating the possibility of once-daily
dosing. Boosting also aims to intensify therapy; due to the ele-
vated plasma levels, boosted indinavir or amprenavir seem to be
effective against resistant viral strains (Condra et al. 2000).
There is, however, a high degree of variability of boosted
plasma levels among individuals. Therapeutic drug monitoring
is therefore recommended (Burger et al. 2002). In addition to
achieving elevated trough levels of the boosted drug, which
prevents plasma levels from dropping below the minimal in-
hibitory concentration, ritonavir also increases peak levels,
which may lead to more side effects.
Saquinavir/ritonavir is the most-studied booster combination
regimen. Due to the low oral bioavailability of saquinavir, this
combination was tested very early on. Plasma levels of saqui-
navir can be increased 20-fold by ritonavir. Studies have shown
that the booster combination 400/400 (= 400 mg saquinavir plus
400 mg ritonavir, both twice daily) is virologically the most
effective (Cameron et al. 1999). In patients already taking
saquinavir, boosting may have only moderate effects (Smith et
al. 2001). Boosting of saquinavir in the better tolerated
1000/100 combination has recently been licensed.
When boosting saquinavir, it is worth considering using Invi-
rase
®
instead of Fortovase
®
. In a recently published study (Ku-
rowski et al. 2002), boosted levels of saquinavir were even
2. Overview of Antiretroviral Drugs 79
Hoffmann, Kamps, et al.
higher for Invirase
®
, which is also better tolerated with respect
to gastrointestinal complaints than the subsequently developed
Fortovase
®
. Interestingly, Invirase
®
is nearly twice as expensive
as Fortovase
®
– an issue that the manufacturer Hoffmann-La
Roche might well have to address in the near future, if, as can
be expected, it comes under pressure from patient advocacy
groups.
Table 2.2: Well investigated boosted PI regimens
Dose in mg Pills/day Comment
Saquinavir/
Ritonavir
2 x 1000/100 2 x 6 Officially licensed for boosting
Saquinavir/
Ritonavir
2 x 400/400 2 x 6 Good efficacy, but problematic
due to increased rate of side
effects
Indinavir/
Ritonavir
2 x 800/100 2 x 3 Higher rate of nephrolithiasis (?)
Indinavir/
Ritonavir
2 x 400/400 2 x 5 Good pharmacokinetic data
Lopinavir/
Ritonavir
2 x 400/100 2 x 3
Only fixed combination in one
capsule
Nelfinavir/
Ritonavir
Not recommended
Saquinavir/
Nelfinavir
3 x 600/750 3 x 6 Only well documented booster
combination without ritonavir, but
too many pills three times a day
Amprenavir/
Ritonavir
2 x 600/100 2 x 5 FDA approved
The combination of indinavir/ritonavir is also well investi-
gated. There is good pharmacokinetic data for the 800/100 dose
(Van Heeswijk et al. 1999). In a smaller pilot study with this
combination, however, results showed nephrolithiasis in 19/57
patients (Voigt et al. 2001). The 400/400 dose presumably in-
duces less renal side effects. The combination of indina-
vir/ritonavir seems to be associated with an increased risk of
side effects. In studies such as BEST or NICE, switching from
indinavir to indinavir/ritonavir was shown to have a slightly
80 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
higher rate of side effects and drop-outs (Gatell et al. 2000,
Harley et al. 2001, Shulman et al. 2002).
Lopinavir/ritonavir is to date the only fixed booster combina-
tion therapy available in one capsule (see above). There is good
data for amprenavir/ritonavir, especially for salvage therapy
(Condra 2000, Duval et al. 2002). The FDA approved once
daily dosing for this combination in 2002.
Boosted PIs are probably equivalent with regard to anti-HIV
potency, although only sparse clinical data is as yet available. In
the randomized MaxCmin1 Trial, efficacy of saquinavir and
indinavir was comparable. The drop-out rate in the indinavir
group was significantly higher, and was probably due to a
higher incidence of side effects (Gerstoft et al. 2002). Results
are expected to be published soon of a second trial, the
MaxCmin2 Study, in which both treatment-naive and treatment-
experienced patients were randomized to receive either saqui-
navir/ritonavir or lopinavir/r. Interim analyses have shown that
both combinations have good efficacy and exhibit no great dif-
ferences (Dragstedt et al. 2002). Publication of the final data is
expected in the third quarter of 2003.
Plasma levels even of well-boosted PIs seem to decrease with
duration of treatment. After 10 months, saquinavir levels had
dropped by 40 % in six patients (Gisolf et al. 2000). Plasma
levels must therefore be monitored for all booster combinations,
especially in patients with underlying liver disease, as the extent
of interaction is unpredictable and dose adjustments may be
required.
References: Nukes, NNRTIs, PIs
1. Bartlett JA, Benoit SL, Johnson VA, et al. Lamivudine plus zidovudine
compared with zalcitabine plus zidovudine in patients with HIV infection.
A randomized, double-blind, placebo-controlled trial. Ann Intern Med
1996, 125: 161-72. http://amedeo.com/lit.php?id=8686973
2. Bernasconi E, Boubaker K, Junghans C, et al. Abnormalities of body fat
distribution in HIV-infected persons treated with antiretroviral drugs: J Ac-
quir Immune Defic Syndr 2002, 31:50-5.
http://amedeo.com/lit.php?id=12352150
2. Overview of Antiretroviral Drugs 81
Hoffmann, Kamps, et al.
3. Bogner JR, Vielhauer V, Beckmann RA, et al. Stavudine versus zi-
dovudine and the development of lipodystrophy. J Acquir Immune Defic
Syndr 2001, 27: 237-44. http://amedeo.com/lit.php?id=11464142
4. Brinkman K, Smeitink JA, Romijn JA, Reiss P. Mitochondrial toxicity in-
duced by nucleoside-analogue reverse-transcriptase inhibitors is a key
factor in the pathogenesis of ART-related lipodystrophy. Lancet 1999,
354:1112-5. http://amedeo.com/lit.php?id=10509516
5. Burger DM, Aarnoutse RE, Hugen PW. Pros and cons of therapeutic drug
monitoring of antiretroviral agents. Curr Opin Inf Dis 2002, 15: 17-22.
6. Cameron DW, Heath-Chiozzi M, Danner S, et al. Randomised placebo-
controlled trial of ritonavir in advanced HIV-1 disease. Lancet 1998,
351:543-9. http://amedeo.com/lit.php?id=9492772
7. Cameron DW, Japour AJ, Xu Y, et al. Ritonavir and saquinavir combina-
tion therapy for the treatment of HIV infection. AIDS 1999, 13: 213-24.
http://amedeo.com/lit.php?id=10202827
8. Carr A, Chuah J, Hudson J, et al. A randomised, open-label comparison
of three HAART regimens including two nucleoside analogues and indi-
navir for previously untreated HIV-1 infection: the OzCombo1 study. AIDS
2000, 14: 1171-80. http://amedeo.com/lit.php?id=10894281
9. Carr A, Workman C, Smith DE, et al. Abacavir substitution for nucleoside
analogs in patients with HIV lipoatrophy: a randomized trial. JAMA 2002,
288:207-15. http://amedeo.com/lit.php?id=12095385
10. Chene G, Angelini E, Cotte L, et al. Role of long-term nucleoside-
analogue therapy in lipodystrophy and metabolic disorders in HIV-
infected patients. Clin Infect Dis 2002, 34: 649-57.
http://amedeo.com/lit.php?id=11810598
11. Cohen Stuart JW, Schuurman R, Burger DM, et al. Randomized trial
comparing saquinavir soft gelatin capsules versus indinavir as part of tri-
ple therapy (CHEESE study). AIDS 1999, 13: F53-8.
http://amedeo.com/lit.php?id=10357371
12. Condra JH, Petropoulos CJ, Ziermann R, et al. Drug resistance and pre-
dicted virologic responses to HIV type 1 protease inhibitor therapy. J In-
fect Dis 2000, 182: 758-65. http://amedeo.com/lit.php?id=10950769
13. Condra JH, Schleif WA, Blahy OM, et al. In vivo emergence of HIV-1
variants resistant to multiple protease inhibitors. Nature 1995, 374:569-
71. http://amedeo.com/lit.php?id=7700387
14. Conway B. Initial therapy with protease inhibitor-sparing regimens:
evaluation of nevirapine and delavirdine. Clin Infect Dis. 2000, Suppl
2:S130-4. http://amedeo.com/lit.php?id=10860897
15. Cozzi-Lepri A, Phillips AN, d'Arminio Monforte A, et al. Virologic and im-
munologic response to regimens containing nevirapine or efavirenz in
combination with 2 nucleoside analogues in the Italian Cohort Naive
Antiretrovirals (I.Co.N.A.) study. J Infect Dis 2002, 185: 1062-9.
http://amedeo.com/lit.php?id=11930316
16. Deeks SG, Grant RM, Beatty GW, et al. Activity of a ritonavir plus saqui-
navir-containing regimen in patients with virologic evidence of indinavir or
82 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
ritonavir failure. AIDS 1998, 12: F97-102.
http://amedeo.com/lit.php?id=9677159
17. Deeks SG, Smith M, Holodniy M, Kahn JO. HIV-1 protease inhibitors. A
review for clinicians. JAMA 1997, 277: 145-53.
http://amedeo.com/lit.php?id=8990341
18. Dragstedt UB, Gerstoft J, Youle M. The interim analysis of a phase IV
randomised, open-label, multicentre trial to evaluate the safety and effi-
cacy of lopinavir/ritonavir (400mg/100mg bid) versus saquinavir/ritonavir
(1000mg/100mg bid) in adult HIV-1 infected patients. Abstract 14.5, 6th
International Congress on Drug Therapy in HIV Infection 2002, Glasgow,
Scotland
19. Duval X, Lamotte C, Race E, et al. Amprenavir inhibitory quotient and
virological response in HIV-infected patients on an amprenavir-containing
salvage regimen without or with ritonavir. Antimicrob Agents Chemother
2002, 46:570-4. http://amedeo.com/lit.php?id=11796381
20. Eron JJ JR, Murphy RL, Peterson D, et al. A comparison of stavudine,
didanosine and indinavir with zidovudine, lamivudine and indinavir for the
initial treatment of HIV-1 infected individuals: selection of thymidine ana-
log regimen therapy (START II). AIDS 2000, 14: 1601-10.
http://amedeo.com/lit.php?id=10983647
21. Eron JJ JR. HIV-1 protease inhibitors. Clin Infect Dis 2000, 30 Suppl 2:
S160-70. http://amedeo.com/lit.php?id=10860901
22. Eshleman SH, Jackson JB. Nevirapine resistance after single dose pro-
phylaxis. AIDS Rev 2002, 4:59-63.
http://amedeo.com/lit.php?id=12152519
23. Fätkenheuer G, Theisen A, Rockstroh J, et al. Virological treatment fail-
ure of protease inhibitor therapy in an unselected cohort of HIV-infected
patients. AIDS 1997, 11:F113-6. http://amedeo.com/lit.php?id=9386799
24. Fichtenbaum CJ, Gerber JG, Rosenkranz SL, et al. Pharmacokinetic
interactions between protease inhibitors and statins in HIV seronegative
volunteers: ACTG Study A5047. AIDS 2002, 16:569-
77.http://amedeo.com/lit.php?id=11873000
25. Fisac C, Fumero E, Crespo M, et al. Metabolic and body composition
changes in patients switching from a PI-containing regimen to abacavir,
efavirenz or nevirapine. 12 month results of a randomized study (Li-
pnefa). Abstract ThPE7354, XIV International AIDS Conference 2002,
Barcelona, Spain.
26. Foudraine NA, de Jong JJ, Weverling J, et al. An open randomized con-
trolled trial of zidovudine plus lamivudine versus stavudine plus lami-
vudine. AIDS 1998, 12: 1513-9. http://amedeo.com/lit.php?id=9727573
27. French M, Amin J, Roth N, et al. Randomized, open-label, comparative
trial to evaluate the efficacy and safety of three antiretroviral drug combi-
nations including two nucleoside analogues and nevirapine for previously
untreated HIV-1 Infection: the OzCombo 2 study. HIV Clin Trials 2002,
3:177-85. http://amedeo.com/lit.php?id=12032876
2. Overview of Antiretroviral Drugs 83
Hoffmann, Kamps, et al.
28. Friedl AC, Ledergerber B, Flepp M, et al. Response to first protease in-
hibitor- and efavirenz-containing antiretroviral combination therapy. The
Swiss HIV Cohort Study. AIDS 2001, 15: 1793-800.
http://amedeo.com/lit.php?id=11579241
29. Fumaz CR, Tuldra A, Ferrer MJ, et al. Quality of life, emotional status,
and adherence of HIV-1-infected patients treated with efavirenz versus
PI-containing regimens. J Acquir Immune Defic Syndr 2002, 29:244-53.
http://amedeo.com/lit.php?id=11873073
30. Galli M, Ridolfo AL, Adorni F, et al. Body habitus changes and metabolic
alterations in protease inhibitor-naive HIV-1-infected patients treated with
two nucleoside reverse transcriptase inhibitors. J Acquir Immune Defic
Syndr 2002, 29: 21-31. http://amedeo.com/lit.php?id=11782586
31. Gatell JM, Lange J, Arnaiz JA, et al. A randomized study comparing con-
tinued indinavir (800 mg tid) vs switching to indinavir/ritonavir (800/100
mg bid) in HIV patients having achieved viral load suppression with indi-
navir plus 2 nucleoside analogues The BID Efficacy and Safety Trial
(BEST). Abstract WeOrB484, XIII International AIDS Conference 2000,
Durban, South Africa.
32. Gerard Y, Maulin L, Yazdanpanah Y, et al. Symptomatic hyperlactatae-
mia: an emerging complication of ART. AIDS 2000, 14:2723-30.
http://amedeo.com/lit.php?id=11125891
33. Gerstoft J, Dragstedt UB, Cahn P. Final analysis of a randomised trial to
evaluate safety and efficacy of indinavir/ritonavir versus saquina-
vir/ritonavir in adult HIV-1 infection: the MaxCmin1 trial. Abstract 2853,
ICAAC 2002, San Diego, USA.
34. Gisolf EH, van Heeswijk RP, Hoetelmans RW, Danner SA. Decreased
exposure to saquinavir in HIV-1-infected patients after long-term antiret-
roviral therapy including ritonavir and saquinavir. AIDS 2000, 14: 801-5.
http://amedeo.com/lit.php?id=10839587
35. Gonzalez de Requena D, Nunez M, Jimenez-Nacher I, Soriano V. Liver
toxicity caused by nevirapine. AIDS 2002, 16:290-1.
http://amedeo.com/lit.php?id=11807315
36. Graham NM. Metabolic disorders among HIV-infected patients treated
with protease inhibitors: a review. J Acquir Immune Defic Syndr 2000, 25
Suppl 1: S4-11. http://amedeo.com/lit.php?id=11126425
37. Gulick RM, Mellors JW, Havlir D, et al. Treatment with indinavir, zi-
dovudine, and lamivudine in adults with HIV infection and prior antiretrovi-
ral therapy. N Engl J Med 1997, 337: 734-9.
http://amedeo.com/lit.php?id=9287228
38. Hammer SM, Katzenstein DA, Hughes MD et al. A trial comparing nu-
cleoside monotherapy with combination therapy in HIV-infected adults
with CD4 cell counts from 200 to 500/ul. N Engl J Med 1996, 335:1081-
90. http://amedeo.com/lit.php?id=8813038
39. Hammer SM, Squires KE, Hughes MD, et al. A controlled trial of two
nucleoside analogues plus indinavir in persons with HIV infection and
CD4 cell counts of 200 per cubic millimeter or less. AIDS Clinical Trials
84 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
Group 320 Study Team. N Engl J Med 1997, 337:725-33.
http://amedeo.com/lit.php?id=9287227
40. Harley W, DeJeus E, Pistole M, et al. A 24-week randomized, controlled,
open-label evaluation of adherence and convenience of continuing indi-
navir versus switching to ritonavir/indinavir 400/400 mg bid. Abstract 334,
8th CROI 2001, Chicago, USA.
http://www.retroconference.org/2001/abstracts/abstracts/abstracts/334.ht
m
41. Havlir DV, Gilbert PB, Bennett K, et al. Effects of treatment intensification
with hydroxyurea in HIV-infected patients with virologic suppression.
AIDS 2001, 15: 1379-88. http://amedeo.com/lit.php?id=11504959
42. Havlir DV, Tierney C, Friedland GH, et al. In vivo antagonism with zi-
dovudine plus stavudine combination therapy. J Infect Dis 2000, 182:
321-5. http://amedeo.com/lit.php?id=10882616
43. Hetherington S, Hughes AR, Mosteller M, et al. Genetic variations in
HLA-B region and hypersensitivity reactions to abacavir. Lancet 2002,
359:1121-2. http://amedeo.com/lit.php?id=11943262
44. HIV Trialists' Collaborative Group. Zidovudine, didanosine, and zalci-
tabine in the treatment of HIV infection: meta-analyses of the randomised
evidence. Lancet 1999, 353: 2014-25.
http://amedeo.com/lit.php?id=10376616
45. Hoffmann C, Jaegel-Guedes E, Wolf E, et al. PI to Efavirenz switch effect
on lipids in HIV-positive patients. Abstract WePeB4185, XIII International
AIDS Conference 2000, Durban, South Africa.
46. John M, James I, McKinnon E, et al. A randomised, controlled, open-label
study of revision of antiretroviral regimens containing stavudine and/or a
protease inhibitor to Zidovudine/Lamivudine/Abacavir to prevent or re-
verse lipoatrophy: 48-week data. Abstract 700, 9th CROI 2002, Seattle.
http://63.126.3.84/2002/Abstract/13277.htm
47. John M, Moore CB, James IR, et al. Chronic hyperlactatemia in HIV-
infected patients taking ART. AIDS 2001, 15: 717-23.
http://amedeo.com/lit.php?id=11371686
48. Kahn JO, Lagakos SW, Richman DD, et al. A controlled trial comparing
continued zidovudine with didanosine in HIV infection. N Engl J Med
1992, 327:581-7. http://amedeo.com/lit.php?id=1353607
49. Katzenstein TL, Kirk O, Pedersen C, et al. The danish protease inhibitor
study: a randomized study comparing the virological efficacy of 3 prote-
ase inhibitor-containing regimens for the treatment of HIV type 1 infec-
tion. J Infect Dis 2000, 182: 744-50.
http://amedeo.com/lit.php?id=10950767
50. Kearney BP, Damle B, Plummer A, et al. Pharmacokinetic evaluation of
tenofovir DF and enteric-coated didanosine. Abstract 9026, XIV Interna-
tional AIDS Conference 2002, Barcelona, Spain.
51. Kempf DJ, Marsh KC, Kumar G, et al. Pharmacokinetic enhancement of
inhibitors of the HIV protease by coadministration with ritonavir. Antimi-
2. Overview of Antiretroviral Drugs 85
Hoffmann, Kamps, et al.
crob Agents Chemother 1997, 41:654-60.
http://amedeo.com/lit.php?id=9056009
52. Kirk O, Mocroft A, Pradier C, et al. Clinical outcome among HIV-infected
patients starting saquinavir hard gel compared to ritonavir or indinavir.
AIDS 2001, 15: 999-1008. http://amedeo.com/lit.php?id=11399982
53. Kuritzkes DR, Marschner I, Johnson VA, et al. Lamivudine in combination
with zidovudine, stavudine, or didanosine in patients with HIV-1 infection.
A randomized, double-blind, placebo-controlled trial. AIDS 1999, 13: 685-
94. http://amedeo.com/lit.php?id=10397563
54. Kurowski M, Kaeser B, Sawyer A, et al. Low-dose ritonavir moderately
enhances nelfinavir exposure. Clin Pharmacol Ther 2002, 72:123-32.
http://amedeo.com/lit.php?id=12189359
55. Kurowski M, Kaeser B, Sawyer A, Popescu M, Mrozikiewicz A. Low-dose
ritonavir moderately enhances nelfinavir exposure. Clin Pharmacol Ther
2002, 72:123-32. http://amedeo.com/lit.php?id=12189359
56. Kurowski M, Sternfeld T, Hill A, Moecklinghoff C. comparative pharmaco-
kinetics and short-term safety of twice daily (bid) fortovase/ritonavir and
invirase/ritonavir. Abstract 432, 9th CROI 2002, Seattle, USA.
http://63.126.3.84/2002/Abstract/13486.htm
57. Lafeuillade A, Poggi C, Djediouane A,. A pilot study of a combination of
three reverse transcriptase inhibitors in HIV-1 infection. Antivir Ther 1997,
2: 219-27. http://amedeo.com/lit.php?id=11327441
58. Lallemand F, Salhi Y, Linard F, Giami A, Rozenbaum W. Sexual dysfunc-
tion in 156 ambulatory HIV-infected men receiving HAART combinations
with and without protease inhibitors. J Acquir Immune Defic Syndr 2002,
30: 187-90. http://amedeo.com/lit.php?id=12045681
59. Mallal S, Nolan D, Witt C, et al. Association between presence of HLA-
B*5701, HLA-DR7, and HLA-DQ3 and hypersensitivity to HIV-1 reverse-
transcriptase inhibitor abacavir. Lancet 2002, 359:727-32.
http://amedeo.com/lit.php?id=11888582
60. Marcus K, Truffa M, Boxwell D, Toerner J. Recently identified adverse
events secondary to NRTI therapy in HIV-infected individuals: cases from
the FDA’s adverse event reporting system. Abstract LB14, 9th CROI
2002, Seattle, USA
http://www.retroconference.org//2002/Abstract/14036.htm
61. Martin C, Sonnerborg A, Svensson JO, Stahle L. Indinavir-based treat-
ment of HIV-1 infected patients: efficacy in the central nervous system.
AIDS 1999, 13: 1227-32. http://amedeo.com/lit.php?id=10416527
62. Martinez-Picado X, Savara A, Sutton L, D’Aquila R. Replicative fitness of
protease inhibitor-resistant mutants of HIV-1. J Virol 1999, 73: 3744-
3752.
63. Marzolini C, Telenti A, Decosterd LA, Greub G, Biollaz J, Buclin T. Efavi-
renz plasma levels can predict treatment failure and central nervous
system side effects in HIV-1-infected patients. AIDS 2001, 15: 71-5.
http://amedeo.com/lit.php?id=11192870
86 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
64. Mauss S, Corzillius M, Wolf E, et al. Risk factors for the HIV-associated
lipodystrophy syndrome in a closed cohort of patients after 3 years of
antiretroviral treatment. HIV Med 2002, 3:49-55.
http://amedeo.com/lit.php?id=12059951
65. McComsey G, Lonergan T, Fisher R, et al. Improvements in lipoatrophy
are observed after 24 weeks when stavudine is replaced by either aba-
cavir or zidovudine. Abstract 701, 9th CROI 2002, Seattle.
http://63.126.3.84/2002/Abstract/12662.htm
66. McMahon D, Lederman M, Haas DW, et al. Antiretroviral activity and
safety of abacavir in combination with selected HIV-1 protease inhibitors
in therapy-naive HIV-1-infected adults. Antivir Ther 2001, 6:105-14.
http://amedeo.com/lit.php?id=11491415
67. Miller KD, Cameron M, Wood LV, et al. Lactic acidosis and hepatic ste-
atosis associated with use of stavudine: report of four cases. Ann Intern
Med 2000, 133:192-6. http://amedeo.com/lit.php?id=10906833
68. Miller V, Stark T, Loeliger AE, Lange JM. The impact of the M184V sub-
stitution in HIV-1 reverse transcriptase on treatment response. HIV Med
2002, 3:135-45. http://amedeo.com/lit.php?id=12010361
69. Miller V, Staszewski S, Boucher CAB, Phair JP. Clinical experience with
non-nucleoside reverse transcriptase inhibitors. AIDS 1997, 11 (suppl A):
S157-164.
70. Mitsuyasu RT, Skolnik PR, Cohen SR, et al. Activity of the soft gelatin
formulation of saquinavir in combination therapy in antiretroviral-naive
patients. NV15355 Study Team. AIDS 1998,12:F103-9.
http://amedeo.com/lit.php?id=9708399
71. Mokrzycki MH, Harris C, May H, Laut J, Palmisano J. Lactic acidosis
associated with stavudine administration: a report of five cases. Clin In-
fect Dis 2000; 30: 198-200. http://amedeo.com/lit.php?id=10619755
72. Molina JM, Chene G, Ferchal F, et al. The ALBI trial: a randomized con-
trolled trial comparing stavudine plus didanosine with zidovudine plus
lamivudine and a regimen alternating both combinations in previously
untreated patients infected with HIV. J Infect Dis 1999, 180: 351-8.
http://amedeo.com/lit.php?id=10395849
73. Moyle G, Pozniak A, Opravil M, et al. The SPICE study: 48-week activity
of combinations of saquinavir soft gelatin and nelfinavir with and without
nucleoside analogues. J Acquir Immune Defic Syndr 2000, 23: 128-37.
http://amedeo.com/lit.php?id=10737427
74. Moyle GJ, Gazzard BG. Finding a role for zalcitabine in the HAART era.
Antivir Ther 1998, 3: 125-37. http://amedeo.com/lit.php?id=10682130
75. Noble S, Goa KL. Amprenavir: a review of its clinical potential in patients
with HIV infection. Drugs 2000, 60:1383-410.
http://amedeo.com/lit.php?id=11152018
76. Nunez M, Soriano V, Martin-Carbonero L, et al. SENC trial: a random-
ized, open-label study in HIV-infected naive individuals. HIV Clin Trials
2002; 3:186-94.
2. Overview of Antiretroviral Drugs 87
Hoffmann, Kamps, et al.
77. Phillips AN, Pradier C, Lazzarin A, et al. Viral load outcome of non-
nucleoside reverse transcriptase inhibitor regimens for 2203 mainly
antiretroviral-experienced patients. AIDS 2001;15:2385-95.
http://amedeo.com/lit.php?id=11740189
78. Picard V, Angelini E, Maillard A, et al. Comparison of genotypic and phe-
notypic resistance patterns of HIV type 1 isolates from patients treated
with stavudine and didanosine or zidovudine and lamivudine. J Infect Dis
2001, 184:781-4. http://amedeo.com/lit.php?id=11517441
79. Piscitelli SC, Gallicano KD. Interactions among drugs for HIV and oppor-
tunistic infections. N Engl J Med 2001, 344:984-96.
80. Raboud JM, Rae S, Vella S, et al. Meta-analysis of two randomized con-
trolled trials comparing combined zidovudine and didanosine therapy with
combined zidovudine, didanosine, and nevirapine therapy in patients with
HIV. INCAS study team. J Acquir Immune Defic Syndr 1999, 22: 260-6.
http://amedeo.com/lit.php?id=10770346
81. Robbins BL, Srinivas RV, Kim C, et al. Anti-HIV activity and cellular me-
tabolism of a potential prodrug of the acyclic nucleoside phosphonate 9-
R-(2-phosphonomethoxypropyl)adenine (PMPA),
Bis(isopropyloxymethylcarbonyl)PMPA. Antimicrob Agents Chemother
1998, 42:612-7. http://amedeo.com/lit.php?id= 9517941
82. Robbins G, Shafer R, Smeaton L, et al. Antiretroviral strategies in naive
HIV+ subjects: comparisons of sequential 3-drug regimens (ACTG 384).
Abstract LbOr20A, XIV International AIDS Conference 2002, Barcelona,
Spain.
83. Roge BT, Katzenstein TL, Gerstoft J. Comparison of P-triglyceride levels
among patients with HIV on randomized treatment with ritonavir, indinavir
or ritonavir/saquinavir. Scand J Infect Dis 2001, 33:306-11.
http://amedeo.com/lit.php?id=11345223
84. Saag MS, Tebas P, Sension M, et al. Randomized, double-blind com-
parison of two nelfinavir doses plus nucleosides in HIV-infected patients
(Agouron study 511). AIDS 2001, 15:1971-8.
http://amedeo.com/lit.php?id=11600825
85. Schooley RT, Ruane P, Myers RA, et al. Tenofovir DF in antiretroviral-
experienced patients: results from a 48-week, randomized, double-blind
study. AIDS 2002, 16:1257-63. http://amedeo.com/lit.php?id=12045491
86. Schrooten W, Colebunders R, Youle M, et al. Sexual dysfunction associ-
ated with protease inhibitor containing HAART. AIDS 2001, 15: 1019-23.
http://amedeo.com/lit.php?id=11399984
87. Sension MG, Bellos NC, Johnson J, et al. Lamivudine 300 mg QD versus
continued lamivudine 150 mg BID with stavudine and a protease inhibitor
in suppressed patients. HIV Clin Trials 2002, 3:361-70.
http://amedeo.com/lit.php?id=12407485
88. Shafer R, Robbins G, Smeaton L, et al. Antiretroviral strategies in naive
HIV+ subjects: comparison of 4-drug versus sequential 3-drug regimens
(ACTG 384). Abstract LbOr20B, XIV International AIDS Conference
2002, Barcelona, Spain.
88 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
89. Sham HL, Kempf DJ, Molla A, et al. ABT-378, a highly potent inhibitor of
the HIV protease. Antimicrob Agents Chemother 1998, 42:3218-24.
http://amedeo.com/lit.php?id=9835517
90. Shulman N, Zolopa A, Havlir D, et al. virtual inhibitory quotient predicts
response to ritonavir boosting of indinavir-based therapy in HIV-infected
patients with ongoing viremia. Antimicrob Agents Chemother 2002,
46:3907-3916. http://amedeo.com/lit.php?id=12435695
91. Smith D, Hales G, Roth N, et al. A randomized trial of nelfinavir, ritonavir,
or delavirdine in combination with saquinavir-SGC and stavudine in
treatment-experienced HIV-1-infected patients. HIV Clin Trials 2001,
2:97-107. http://amedeo.com/lit.php?id=11590517
92. Sommadossi JP. HIV protease inhibitors: pharmacologic and metabolic
distinctions. AIDS 1999, 13 Suppl 1: S29-40.
http://amedeo.com/lit.php?id=10546783
93. Squires KE, Gulick R, Tebas P, et al. A comparison of stavudine plus
lamivudine versus zidovudine plus lamivudine in combination with indina-
vir in antiretroviral naive individuals with HIV infection: selection of thymi-
dine analog regimen therapy (START I). AIDS 2000, 14: 1591-600.
http://amedeo.com/lit.php?id=10983646
94. Staszewski S, Gallant J, Pozniak AL, et al. Efficacy and saefty of tenofo-
vir disoproxil fumarate versus stavudine when used in combination with
lamivudine and efavirenz in HIV-1 infected patients naive to antiretroviral
therapy: 48-week interim results. Abstract LB17, XIV International AIDS
Conference 2002, Barcelona, Spain.
95. Staszewski S, Morales-Ramirez J, Tashima KT, et al. Efavirenz plus
zidovudine and lamivudine, efavirenz plus indinavir, and indinavir plus zi-
dovudine and lamivudine in the treatment of HIV-1 infection in adults.
Study 006 Team. N Engl J Med 1999, 341:1865-73.
http://amedeo.com/lit.php?id=10601505
96. Stellbrink HJ, Hawkins DA, Clumeck N, et al. Randomised, multicentre
phase III study of saquinavir plus zidovudine plus zalcitabine in previously
untreated or minimally pretreated hiv-infected patients. Clin Drug Invest
2000, 20:295-307.
97. Stellbrink HJ, van Lunzen J, Westby M, et al. Effects of interleukin-2 plus
highly active antiretroviral therapy on HIV-1 replication and proviral DNA
(COSMIC trial). AIDS 2002, 16:1479-87.
http://amedeo.com/lit.php?id=12131185
98. Sulkowski MS, Thomas DL, Chaisson RE, Moore RD. Hepatotoxicity
associated with antiretroviral therapy in adults infected with HIV and the
role of hepatitis C or B virus infection. JAMA 2000, 283: 74-80.
http://amedeo.com/lit.php?id=10632283
99. Sulkowski MS, Thomas DL, Mehta SH, Chaisson RE, Moore RD. Hepa-
totoxicity associated with nevirapine or efavirenz-containing antiretroviral
therapy: role of hepatitis C and B infections. Hepatology 2002, 35:182-9.
http://amedeo.com/lit.php?id=11786975
2. Overview of Antiretroviral Drugs 89
Hoffmann, Kamps, et al.
100. Suo Z, Johnson KA. Selective inhibition of HIV-1 reverse transcriptase by
an antiviral inhibitor, (R)-9-(2-Phosphonylmethoxypropyl)adenine. J Biol
Chem 1998, 273:27250-8. http://amedeo.com/lit.php?id=9765248
101. Torre D, Tambini R, Speranza F. Nevirapine or efavirenz combined with
two nucleoside reverse transcriptase inhibitors compared to HAART: a
meta-analysis of randomized clinical trials. HIV Clin Trials 2001, 2: 113-
21. http://amedeo.com/lit.php?id=11590519
102. Van der Valk M, Kastelein JJ, Murphy RL, et al. Nevirapine-containing
antiretroviral therapy in HIV-1 infected patients results in an anti-
atherogenic lipid profile. AIDS 2001, 15: 2407-14.
http://amedeo.com/lit.php?id=11740191
103. Van Heeswijk RP, Veldkamp AI, Hoetelmans RM, et al. The steady-state
plasma pharmacokinetics of indinavir alone and in combination with a low
dose of ritonavir in twice daily dosing regimens in HIV-1-infected indi-
viduals. AIDS 1999, 13: F95-9. http://amedeo.com/lit.php?id=10513637
104. Veldkamp AI, Weverling GJ, Lange JM, et al. High exposure to nevira-
pine in plasma is associated with an improved virological response in
HIV-1-infected individuals. AIDS 2001; 15: 1089-95.
http://amedeo.com/lit.php?id=11416710
105. Voigt E, Wickesberg A, Wasmuth JC, et al. First-line ritonavir/indinavir
100/800 mg twice daily plus nucleoside reverse transcriptase inhibitors in
a German multicentre study: 48-week results. HIV Med 2002, 3:277-282.
http://amedeo.com/lit.php?id=12444946
106. Walmsley S, Bernstein B, King M, et al. Lopinavir-ritonavir versus nelfina-
vir for the initial treatment of HIV infection. N Engl J Med 2002, 346:2039-
46. http://amedeo.com/lit.php?id=12087139
107. Wensing AM, Reedijk M, Richter C, Boucher CA, Borleffs JC. Replacing
ritonavir by nelfinavir or nelfinavir/saquinavir as part of HAART leads to
an improvement of triglyceride levels. AIDS 2001, 15:2191-3.
108. Wood R, Hawkins DA, Moyle G, et al. Second placebo-controlled study in
naive individuals confirms the role of delavirdine in highly active antiretro-
viral, protease-sparing treatment. Abstract 624. 6th CROI 1999, Chicago,
USA. http://www.retroconference.org/99/abstracts/624.htm
ART 2003/2004: The Horizon and Beyond
This overview focuses on drugs already well advanced in de-
velopment. In view of the large number of substances currently
being tested, it does not claim to be complete. Some products
are about to be licensed and have been available through ex-
panded access programs. At least four are likely to be approved
within the next one to two years: d4T XR, emtricitabine, fos-
90 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
amprenavir, atazanavir. The new entry-inhibitor T-20 has been
approved in the US in March 2003.
New nukes
Stavudine (Zerit
®
) will soon be available as d4T XR (extended
release) in a capsulated once-daily formulation (75 mg and 100
mg). The formulation is stable, does not accumulate and seems
to cause less polyneuropathy, possibly due to lower peak levels.
In the BMS 099 Study, d4T XR was tested double-blind (in
combination with lamivudine and efavirenz) against the stan-
dard dose of 2 x 40 mg stavudine daily. At 24 weeks, there were
no differences in efficacy (CD4+ cell count, viral load) and
safety (Pollard et al. 2002). d4T XR was approved by the FDA
on December 31, 2002.
Emtricitabine (Coviracil
®
, FTC) is a cytidine analog which
was developed by Triangle. It has a very long half-life (once-
daily dosing with 200 mg) and biochemically resembles lami-
vudine. In vitro, it was more effective than lamivudine, but this
has not been confirmed in humans (Delehanty et al. 1999). Its
advantage over lamivudine is questionable since its efficacy is
also completely impaired with the M184V point mutation.
Newer data from the FTC-301 Trial might lead to drug approval
in 2003 (Saag et al. 2002): In this double-blind, randomized
study, emtricitabine and stavudine, both in combination with
didanosine and efavirenz, were compared in 571 treatment-
naive patients. The study was stopped after a mean follow-up of
42 weeks. After this period, the probability for virologic failure
after one year, estimated by Kaplan-Meier curve, was 14 % in
the stavudine group versus 6 % in patients receiving emtrici-
tabine; the study investigators considered the difference great
enough to prematurely end the trial. Toxicity was also higher in
the stavudine group. In the Montana Study, the good tolerability
of a once-daily combination with emtricitabine, didanosine and
efavirenz seems to be confirmed (Molina et al. 2001). After the
acquisition of Triangle by Gilead, the development of a fixed
2. Overview of Antiretroviral Drugs 91
Hoffmann, Kamps, et al.
combination of emtricitabine and tenofovir in one single tablet
is planned.
DAPD (Amdoxovir) is a guanine analog developed by Trian-
gle. DAPD is converted in vivo to the highly potent DXG. It is
currently being tested in Phase I/II studies (Corbett et al. 2001).
DAPD is effective against zidovudine/lamivudine-resistant vi-
ruses, including viruses with an insertion at codon 69, which
confers multi-resistance against all nucleoside analogs. Sensi-
tivity seems reduced in the presence of mutations such as K65R
and L74V (Chong et al. 2002, Mewshaw et al. 2002). In cell
cultures the drug has synergistic effects with the fusion inhibitor
T-20, which could be useful in the future (Trembley et al.
2002). Equally pleasing is its good efficacy against hepatitis B
viruses. Reports on possible lens anomalies are less satisfying.
Although the association with DAPD is not yet certain, the
company was immediately required by the FDA to study this
issue further before continuation of trials.
A look into the lab – Experiments in cell cultures have shown
that DPC 817, a new oral cytidine analog from BMS with a
long half-life, is very effective in the presence of zi-
dovudine/lamivudine resistance mutations (Schinazi et al.
2002). The same appears to be true for ACH-126,443 (Beta-L-
Fd4C), an enantiomer of DPC 817, developed by Achillion
Pharmaceuticals. It would appear to allow once-daily dosing
and also to be effective against multi-resistant HIV strains and
against hepatitis B viruses. Phase IB studies in HIV infected
patients are being performed for both drugs. There was similar
news of BCH-13520, a drug by Shire BioChem Inc. In this
case, however, first reports of resistance mutations (Q15M and
the insertion at codon 69) have been published (Bethell et al.
2002). MIV-301 (Alovudine, FLT) is a thymidine analog,
which was initially tested in the 80s but abandoned at the time,
mainly due to myelotoxicity. MIV-301 could be celebrating a
comeback, as it seems to have excellent efficacy against nuke-
resistant viruses (Kim et al. 2001).
92 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
Out of sight, out of mind: The following drugs are currently
not being pursued, as they are either too toxic or have poor effi-
cacy:
Adefovir dipivoxil (bis POM PMEA) from Gilead Sciences
dOTC (BCH-10652) from BioChem Pharma
FddA (Beta-fluoro-ddA, Lodenosine
®
) from US Bioscience
Lobucavir from BMS
New NNRTIs
Even more than with any other drug class, the industry has the
following motto: If one cannot come up with a new drug that is
at least effective against efavirenz- and nevirapine-resistant vi-
ruses, one might as well not continue with the research. “Me-
too” drugs are not needed. In the meantime various drugs have
already been abandoned; the road to approval is especially long
and hard for NNRTIs, even though they are relatively cheap to
develop. Many of the drugs featured below will therefore not
reach market development.
TMC 125 is a new second generation NNRTI. It is effective
against both wild-type viruses and viruses with almost all of the
classical NNRTI mutations (such as K103N, Y181C). In a
Phase IIB study, in which 16 patients on stable ART mostly
with several NNRTI mutations were treated with 900 mg TMC
125 BID for 7 days, the viral load dropped by a median 0.9 log,
and in some cases up to 1.7 log (Gazzard et al. 2002, Sankatsing
et al. 2002). Even after this timepoint, viral load continued to
decrease. TMC 125 was well tolerated. The half-life is long,
and the drug is metabolized in the liver. Although first pharma-
cokinetic data indicates unfavorable interactions with PIs (espe-
cially indinavir and saquinavir), TMC 125 appears to be devel-
oping into a strong and promising drug, with a high genetic bar-
rier.
DPC 083 is a second generation NNRTI, which is also said to
be effective against NNRTI-resistant viruses. In a Phase II
study, in which the drug was tested in two doses of 100 and 200
2. Overview of Antiretroviral Drugs 93
Hoffmann, Kamps, et al.
mg qd in patients with NNRTI treatment failure (viral load >
1000 copies/ml), 4/10 patients who, based on the resistance pro-
file, received DPC 083 as the only effective drug, achieved a
viral load of < 400 copies/ml (Ruiz et al. 2002). However, the
data from this study was unsatisfactory. There was no data on
resistance mutations in these patients, and no clear dose effect
with regard to both effectiveness and side effects. In a previous
“early” Phase III study, which had compared three different
doses of DPC 083 double-blind (50 mg, 100 mg, 200 mg) in
some 100 treatment-naive patients, the effect against wild-type
viruses was comparable to that of efavirenz. The side effects
seemed to be less.
GW420867X is a quinoxaline-NNRTI from GlaxoSmithKline,
which has been shown to be quite effective in vivo in combina-
tion with zidovudine and lamivudine (Arasteh et al. 2001). It
has good CNS penetration and would probably be a candidate
for once-daily dosing (Thomas et al. 2000). As monotherapy,
GW420867X decreased viral load by 1.5 log after 8 days, and
there were no differences mong the various doses investigated
(50 mg, 100 mg, 200 mg). The side effects – neurological, gas-
trointestinal, hepatic – resemble the typical NNRTI complaints;
rash was uncommon. However, cross-resistance seems to exist
to nevirapine and efavirenz, and it is to be expected that devel-
opment will be stopped.
Capravirine (AG1549, previously S-1153) is a relatively well-
advanced NNRTI, which was initially developed by Shionogi
Pharmaceuticals (Fujiwara et al. 1998) and subsequently sold to
Agouron. Capravirine is effective in vivo even against viruses
with the K103N mutation (Wolfe et al. 2001) and was therefore
a hopeful candidate in the battle against NNRTI-resistant vi-
ruses. After animal studies in dogs showed an unusually high
rate of vasculitis after higher doses, Agouron stopped all Phase
II/III clinical trials last year. Safety evaluations have now
shown that capravirine does not cause such side effects in hu-
mans (Hawley et al. 2002), and in the meantime development
will continue. The dose will probably be set at 2 x 700 mg/day.
94 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
Emivirine (EMV, MKC-442, Coactinon) is an NNRTI which
requires twice-daily dosing and has good tolerability (Szczech
et al. 2000). The main side effects are nausea and the efavirenz-
typical dizziness. In a study of patients with relatively low
treatment experience, efficacy was good; 82 % of patients had a
viral load below 400 copies/ml at 16 weeks, on a combination
of stavudine, didanosine and emivirine (Johnson et al. 1999).
Unfortunately, the drug seems to be a “me-too” product. There
are no differences when compared to the other NNRTIs, and
significant cross-resistance (Jeffrey et al. 1999, McCreedy et al.
1999) and PI-interactions (Blum et al. 1998) exist. Further de-
velopment seems uncertain; the FDA has deemed the available
data insufficient for approval.
Out of sight, out of mind - The following NNRTIs are cur-
rently not being pursued, as they are either too toxic or have
poor efficacy:
Calanolide A from Sarawak MediChem Pharmaceu-
ticals
Atevirdine from Upjohn
Loviride from Janssen Pharmaceuticals
HBY-097 from Hoechst-Bayer
PNU142721 from Pharmacia & Upjohn
New protease inhibitors (PIs)
Fos-amprenavir (GW433908) is a calcium phosphate-ester of
amprenavir with better solubility and resorption than the parent
compound. It is generally well tolerated, and patients would
have to take either 2 x 1 or 1 x 2 pills daily when fos-
amprenavir is boosted with ritonavir. This compares favorably
with the unacceptable 8 pills bid of standard amprenavir dos-
age.
In the NEAT Study (APV30001), fos-amprenavir is currently
being compared with nelfinavir in treatment-naive patients
(Rodriguez et al. 2002). 251 patients were randomized open-
label to fos-amprenavir or nelfinavir, with a nuke backbone of
2. Overview of Antiretroviral Drugs 95
Hoffmann, Kamps, et al.
lamivudine and abacavir. Preliminary 24-week data shows that
54 % versus 40 % of patients (ITT) reached an undetectable
viral load. The difference in efficacy was more pronounced in
patients with a high viral load. There was also less diarrhea in
the fos-amprenavir group. As a potent inductor of amprenavir
metabolism, efavirenz can significantly lower plasma levels.
This does not happen when fos-amprenavir is boosted with rito-
navir, as a study recently demonstrated (Wire et al. 2002): 32
healthy volunteers received one dose of fos-amprenavir with
either 100 mg ritonavir bid, 100 mg ritonavir bid plus efavirenz,
or 200 mg ritonavir bid plus efavirenz. Overall, the outcomes
were similar. In brief: efavirenz does not change anything if
ritonavir is given in addition to fos-amprenavir. Even 100 mg
ritonavir significantly influences plasma levels; 200 mg neither
cause more side effects nor achieve further increases in plasma
levels, although lipid levels may increase slightly.
Atazanavir (Reyataz
®
) is a once-daily PI with a favorable lipid
profile (Robinson et al. 2000) and an antiviral potency which
seems comparable to that of nelfinavir (Squires et al. 2001,
Cahn et al. 2001). The 400 mg dose is currently being tested in
phase III studies (Piliero et al. 2002). Recently, data from a
large study comparing atazanavir with efavirenz (with
AZT+3TC as the nuke backbone) was presented (Squires et al.
2002). There were no differences with regard to virological re-
sponse. However, the proportion of patients with < 50 copies/ml
was very low in both groups, which is likely to be a methodo-
logical problem in this (probably too large) trial. Lipid levels
were clearly better in the atazanavir than in the efavirenz group.
The primary resistance-conferring mutation for this drug seems
to be 150L; this mutation does not interfere with sensitivity to
other PIs (including amprenavir) and may possibly even im-
prove it. Interestingly, amprenavir selects for another mutation
exactly at this codon – 150V; the drug thus seems to select on a
different basis than amprenavir (Colonno et al. 2002).
Based on the available data, atazanavir could become a good
option for initial therapy. The drug is already available in an
96 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
expanded access program and will probably be approved in
2003.
Increases in bilirubin levels seem to be a frequent problem. The
mechanism for this resembles that of the Gilbert syndrome (and
the increased levels with indinavir); there are increased levels of
indirect bilirubin due to reduced conjugation in the liver. Al-
though to date no serious hepatic disorders have been described,
liver function should be monitored.
Studies of drug interactions in healthy volunteers have shown
that rifabutin does not significantly influence atazanavir levels.
In contrast, efavirenz may lower plasma levels due to enzyme
induction, probably through induction of CYP3A4 (Preston et
al. 2002). Concomitant therapy with efavirenz will therefore
require an increased dose of atazanavir. Adding 200 mg of rito-
navir when giving efavirenz can diminish this effect, presuma-
bly also counteracting any favorable effects on lipid levels.
Tipranavir is the first non-peptide protease inhibitor and shows
good efficacy against PI-resistant viruses (Larder et al. 2000).
In a study of 41 patients previously treated with at least two PIs,
tipranavir still showed efficacy in 35 patients (Schwartz et al.
2002). Only the combination of the V82T and L33 point muta-
tions led to a reduction in sensitivity. Oral bioavailability of
tipranavir is not very good, and it always requires ritonavir
boosting (inhibition of the CYP3A4 system), as a trial in 113
HIV negative volunteers has shown. Ritonavir increases Cmax
at least 4-fold and Cmin at least 20-fold.
Mozenavir (DMP-450) is a cyclic PI with good solubility,
which was initially developed by Dupont and has now been
sold to Triangle. The required dose will probably enable manu-
facture of a single pill (Sierra-Madero 2001). A disadvantage of
the drug is its short half-life. As with indinavir, three daily
doses will probably be necessary; the resistance profile is also
similar. In a small Phase I/II study in 50 patients, virological
efficacy was comparable to indinavir. Fortunately, the longer
QT-intervals, which had occurred in dogs, were not observed.
2. Overview of Antiretroviral Drugs 97
Hoffmann, Kamps, et al.
Numerous other PIs are already in early clinical trials. Exam-
ples include the drugs TMC 114 and TMC 126 from Tibotec,
which should be effective especially against PI-resistant vi-
ruses. First Phase II trials are ongoing
.
References: New Nukes, NNRTIs, PIs
1. Arasteh K, Wood R, Muller M, et al. GW420867X administered to HIV-1-
infected patients alone and in combination with lamivudine and zi-
dovudine. HIV Clin Trials 2001, 2:307-16.
http://amedeo.com/lit.php?id=11590533
2. Bethell RC, Allard B, De Muys JM, et al. BCH-13520, a new heterosub-
stituted nucleoside analogue, is an effective inhibitor of drug-resistant
HIV-1. Abstract 386, 9th CROI 2002, Seattle, USA.
http://63.126.3.84/2002/Abstract/13340.htm
3. Blum MR, Moxham CP, Kargl DJ, et al. A pharmacokinetic interaction
evaluation of MKC-442 and nelfinavir in healthy male and female volun-
teers. Abstract 12380, 12th International AIDS Conference 1998, Ge-
neva, Switzerland.
4. Cahn P, Percival L, Phanuphak P, et al. Phase II 24-week data from
study AI424-008: Comparative results of BMS-232632, stavudine, lami-
vudine as HAART for treatment-naive HIV-infected patients. Abstract 5,
1st IAS Conference on HIV Pathogenesis and Treatment 2001, Buenos
Aires, Argentina.
5. Chong Y, Borroto-Esoda K, Furman PA, Schinazi RF, Chu CK. Molecular
mechanism of DAPD/DXG against zidovudine- and lamivudine-drug re-
sistant mutants: a molecular modelling approach. Antivir Chem Chemo-
ther 2002, 13:115-28. http://amedeo.com/lit.php?id=12238529
6. Colonno RJ, Friborg J, Rose RE, Lam E, Parkin N. Identification of amino
acid substitutions correlated with reduced atazanavir susceptibility in pa-
tients treated with atazanavir-containing regimens. Antiviral Ther 2002,
7:S4. Abstract 4.
7. Corbett AH, Rublein JC. DAPD. Curr Opin Investig Drugs 2001, 2:348-53.
http://amedeo.com/lit.php?id=11575703
8. Delehanty J, Wakeford C, Hulett L, et al. A phase I/II randomized, con-
trolled study of FTC versus 3TC in HIV-infected patients. Abstract 16, 6th
CROI 1999, Chicago, USA.
9. Fujiwara T, Sato A, el-Farrash M, et al. S-1153 inhibits replication of
known drug-resistant strains of HIV-1. Antimicrob Agents Chemother
1998, 42:1340-5. Original-Artikel
http://aac.asm.org/cgi/content/full/42/6/1340?view=full&pmid=9624472
10. Gazzard B, Pozniak A, Arasteh K, et al.TMC125, A next-generation
NNRTI, demonstrates high potency after 7 days therapy in treatment-
experienced HIV-1-infected individuals with phenotypic NNRTI resis-
tance. Abstract 4, 9th CROI 2002, Seattle, USA.
http://63.126.3.84/2002/Abstract/14022.htm
98 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
11. Hawley P, Diniz-Piraino S, Paxton W. et al. Absence of risk of vasculitis
in a hiv population taking capravirine-results of an active monitoring plan.
Abstract TuPeB4549. XIV International AIDS Conference 2002, Barce-
lona, Spain
12. Jeffrey S, Corbett J, Bacheler L. In vitro NNRTI resistance of recombinant
HIV carrying mutations observed in efavirenz treatment failures. Abstract
110, 6th CROI Chicago 1999, USA.
13. Johnson D, Sanne I, Baraldi E, et al. A phase II, open-label study to
evaluate the antiviral activity, safety, and tolerability of emivirine (EMV,
MKC-442) and stavudine + didanosine. Abstract 502, 39th ICAAC 1999,
San Francisco, USA.
14. Kim EY, Vrang L, Oberg B, Merigan TC. Anti-HIV type 1 activity of 3'-
fluoro-3'-deoxythymidine for several different multidrug-resistant mutants.
AIDS Res Hum Retroviruses 2001, 17:401-7.
http://amedeo.com/lit.php?id=11282008
15. Larder BA, Hertogs K, Bloor S, et al. Tipranavir inhibits broadly protease
inhibitor-resistant HIV-1 clinical samples. AIDS 2000, 14:1943-8.
http://amedeo.com/lit.php?id=10997398
16. McCallister S, Sabo J, Galitz L, Mayers D. An open-label steady state
investigation of the pharmacokinetics of tipranavir and ritonavir and their
effects on cytochrome P-450 (3A4) activity in normal healthy volunteers
(BI 1182.5). Abstract 434, 9th CROI 2002, Seattle, USA.
http://63.126.3.84/2002/Abstract/13434.htm
17. McCreedy B, Borroto-Esoda K, Harris J, et al. Genotypic and phenotypic
analysis of HIV-1 from patients receiving therapy containing two NRTIs in
combination with the NNRTI, emivirine (MKC-442). Abstract 1173, 39th
ICAAC 1999, San Francisco, USA.
18. Mewshaw JP, Myrick FT, Wakefield DA, et al. Dioxolane guanosine, the
active form of the prodrug diaminopurine dioxolane, is a potent inhibitor of
drug-resistant HIV-1 isolates from patients for whom standard nucleoside
therapy fails. J Acquir Immune Defic Syndr 2002, 29:11-20.
http://amedeo.com/lit.php?id=11782585
19. Molina J, Perusat S, Ferchal F, et al. Once-daily combination therapy with
emtricitabine, didanosine and efavirenz in treatment-naive HIV-infected
adults: 64-week follow-up of the ANRS 091 trial. Abstract 321, 8th CROI
2001, Chicago, USA.
20. Piliero P, Cahn C, Pantaleo G, et al. Atazanavir: A Once-Daily Protease
Inhibitor with a Superior Lipid Profile: Results of Clinical Trials Beyond
Week 48. Abstract 706, 9th CROI 2002, Seattle, USA.
http://63.126.3.84/2002/Abstract/13827.htm
21. Pollard R, Ive P, Farthing C, et al. Stavudine XR vs stavudine IR as part
of potent antiretroviral combination therapy: 24-week safety and antiviral
efficacy. Abstract 411, 9th CROI 2002, Seattle, USA.
http://63.126.3.84/2002/Abstract/13559.htm
22. Preston S, Piliero P, O'Mara E, et al. Evaluation of steady-state interac-
tion between atazanavir and efavirenz. Abstract 443, 9th CROI 2002, Se-
attle, USA. http://63.126.3.84/2002/Abstract/13543.htm
2. Overview of Antiretroviral Drugs 99
Hoffmann, Kamps, et al.
23. Robinson BS, Riccardi KA, Gong YF, et al. BMS-232632, a highly potent
HIV protease inhibitor that can be used in combination with other avail-
able antiretroviral agents. Antimicrob Agents Chemother 2000, 44:2093-
2099. Original-Artikel:
http://aac.asm.org/cgi/content/full/44/8/2093?view=full&pmid=10898681
24. Rodriguez-French A, Nadler J, and the Neat Study Team. The NEAT
Study: GW433908 efficacy and safety in anti-retroviral therapy naive
subjects, preliminary 24-week results. Abstract H-166, 42th ICAAC 2002,
San Diego, USA.
25. Ruiz N, Nusrat R, Lauenroth-Mai E, et al. Study DPC 083-203, a phase II
comparison of 100 and 200 mg once-daily DPC 083 and 2 NRTIs in pa-
tients failing a NNRTI-containing regimen. Abstract 6, 9th CROI 2002,
Seattle, USA. http://63.126.3.84/2002/Abstract/13700.htm
26. Saag M, Cahn P, Raffi F, et al. A randomized, double-blind, multicenter
comparison of emtricitabine QD to stavudine BID. Abstract LB1, 42nd
ICAAC 2002, San Diego, USA.
27. Sankatsing S, Weverling G, van't Klooster G, et al. TMC125 monotherapy
for 1 week results in a similar initial rate of decline of HIV-1 RNA as ther-
apy with a 5-drug regimen. Abstract 5, 9th CROI 2002, Seattle, USA.
http://63.126.3.84/2002/Abstract/13427.htm
28. Schinazi RF, Mellors J, Bazmi H, et al. DPC 817: a cytidine nucleoside
analog with activity against zidovudine- and lamivudine-resistant viral
variants. Antimicrob Agents Chemother 2002, 46:1394-401.
http://amedeo.com/lit.php?id=11959574
29. Schwartz R, Kazanjian P, Slater L. Resistance to tipranavir is uncommon
in a randomized trial of tipranavir/ritonavir in multiple PI-failure patients
(BI 1182.2). Abstract 562, 9th CROI 2002, Seattle, USA.
http://63.126.3.84/2002/Abstract/13757.htm
30. Sierra-Madero J. Antiviral activity, safety and phamacokinetics of moze-
navir (DMP 450), a novel cyclic urea protease inhibitor, in combination
with d4t and 3tc in treatment-naïve hiv-1 infected patients (study DMP-
102). Abstract 2, 1st IAS Conference on HIV Pathogenesis and Treat-
ment 2001, Buenos Aires, Argentina.
31. Squires K, Gatell J, Piliero P, et al. AI424-007: 48-week safety and effi-
cacy results from a phase II study of a once-daily HIV-1 protease inhibi-
tor, BMS-232632. Abstract 15, 8th CROI 2001, Chicago, USA.
32. Squires KE, Thiry A, Giordano M, for the AI424-034 International Study
Team. Atazanavir QD and efavirenz QD with fixed-dose ZDV+3TC:
Comparison of antiviral efficacy and safety through wk 24 (AI424-034).
Abstract H-1076, 42nd ICAAC 2002, San Diego, USA.
33. Szczech GM, Furman P, Painter GR, et al. Safety assessment, in vitro
and in vivo, and pharmacokinetics of emivirine, a potent and selective
nonnucleoside reverse transcriptase inhibitor of HIV type 1. Antimicrob
Agents Chemother 2000, 44:123-30.
http://amedeo.com/lit.php?id=10602732
100 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
34. Thomas S, Cass L, Prince W, Segal M. Brain and CSF entry of the novel
non-nucleoside reverse transcriptase inhibitor, GW420867X. Neuroreport
2000, 11:3811-5. http://amedeo.com/lit.php?id=11117496
35. Tremblay C, Poulain D, Hicks JL, et al. T-20 and DAPD have synergistic
in vitro anti-HIV interactions. Abstract 393, 9th CROI 2002, Seattle, USA.
http://63.126.3.84/2002/Abstract/13137.htm
36. Wire MB, Ballow C, Preston S, et al. An assessment of plasma amprena-
vir pharmacokinetics following administration of two GW433908 and rito-
navir regimens in combination with efavirenz in healthy adult subjects
(APV10010). Abstract 443, 9th CROI 2002, Seattle, USA.
http://63.126.3.84/2002/Abstract/13503.htm
37. Wolfe P, Hawley P, Boccia G, et al. Safety and efficacy of capravirine
versus placebo in HIV-infected patients failing a NNRTI-containing regi-
men: results of a phase II, double blind, placebo controlled trial. Abstract
323, 8th CROI 2001, Chicago, USA.
http://www.retroconference.org//2001/abstracts/abstracts/abstracts/323.ht
m
Entry inhibitors
There are three crucial steps for entry of HIV into the CD4+ T
cell:
binding of HIV to the CD4 receptor (“attachment” – target
of attachment inhibitors),
binding to co-receptors (target of co-receptor antagonists),
and finally
fusion of virus and cell (target of fusion inhibitors).
All three drug classes are currently summarized as entry in-
hibitors. Even if the antiviral effects of the drugs now being
tested are not overwhelming, the availability of new drugs with
different mechanisms of action could open up new possibilities
for the treatment of HIV infection.
Attachment inhibitors
BMS-806 is an “early” attachment inhibitor, which, independ-
ently of co-receptors, binds to HIV gp120 specifically and re-
versibly, and so prevents the attachment of HIV to CD4+ T-
lymphocytes (Lin et al. 2002). It has oral bioavailability with
low plasma protein binding and could probably be taken in
2. Overview of Antiretroviral Drugs 101
Hoffmann, Kamps, et al.
tablet form. Animal studies have shown good tolerability. There
is hope that there might be an additive, perhaps even synergistic
effect with other entry inhibitors. Enthusiasm about this drug
have been slightly dampened by the demonstration that differ-
ent HIV isolates have shown differences in sensitivity to BMS
806, indicating potential rapid development of resistance.
Pro-542 is a soluble antibody-like fusion protein, which also
prevents attachment of HIV to CD4+ T-lymphocytes by binding
to gp120. Phase I studies have shown good tolerability, and vi-
ral load decreased even after a single infusion (Jacobson et al.
2000). Pro-542 has already been tested in children (Shearer et
al. 2000). In the SCID mouse model, Pro-542 has shown re-
markable efficacy (Franti et al. 2002). However, the impractical
route of administration (infusion) must be improved.
Co-receptor antagonists
SCH-C is a CCR5-receptor antagonist with oral bioavailability
and a potent in vitro activity against numerous HIV isolates
(Strizki et al. 2001). In healthy volunteers, side effects such as
arrhythmias (longer QT-interval) occurred mainly at higher
doses. This problem appears to be absent with low dosage, and
the FDA has given the go-ahead for further development. A
pilot study in 12 HIV patients, who received SCH-C for 10 days
(the dose was significantly reduced to 2 x 25 mg/daily) showed
that all 10 patients had a decrease in viral load of more than 0.5
log, and 4 patients of more than 1.0 log (Reynes et al. 2002).
This effect persisted even a few days after completion of ther-
apy. However, viral escape mutants have already been de-
scribed for SCH-C, which are cross-resistant to other CCR5-
receptor antagonists (Riley et al. 2002, Xu et al. 2002). SCH-D
is claimed to be more potent and better tolerated than SCH-C,
and therefore seems to have better chances for further develop-
ment (Chen et al. 2002). However, first reports have described
development of resistance, probably via changes in the HIV env
gene.
102 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
Pro-140 is a CCR5-antagonist, which acts as a monoclonal an-
tibody (Trkola et al. 2001). In animal studies (SCID mouse
model), single doses of the drug achieved significant and dose-
related reductions in viral load. (Franti et al. 2002). Clinical
data is still lacking, and there is no information on tolerability.
AMD-3100 is a CXCR4-receptor antagonist as is T-22. Results
to date could be described as discouraging. In a complicated
study with 12 patients, who received a continuous infusion over
10 days, no reduction of viral load occurred. In addition, a vari-
ety of side effects such as thrombocytopenia, orthostasis and
arrhythmias were experienced (Hendrix et al. 2002). However,
AMD-3100 appears to be effective against CXCR4-receptor-
tropic viruses (Schols et al. 2002, van Rij et al. 2002). The one
patient who exclusively harbored such a virus population was
the only one to show a decrease in viral load by 0.87 log after
11 days, and 1.34 log after 18 days. Resistance mutations have
been described both for AMD-3100 and T-22. Whether devel-
opment of AMD-3100 will be pursued is uncertain. Follow-on
drugs, possibly for oral administration, are apparently being
studied.
Fusion inhibitors
T-20 (Enfuvirtide, Fuzeon
®
): Hardly a day goes by without
mention of T-20 in the media, and every other patient asks him-
or herself and the physician, why he or she is not yet receiving
the “new drug”. T-20 is the prototype of fusion inhibitors. It is a
relatively large peptide, comprised of 36 amino acids, and must
therefore be given by subcutaneous injection like insulin. It
binds to an intermediate structure of the HIV gp41-protein,
which appears during entry of HIV into the target cell, i.e. dur-
ing fusion.
In one of the first studies with T-20, HIV patients were given
different doses intravenously as monotherapy. There was a
dose-related effect, and with the higher dose of 2 x 100 mg per
day, median viral load was reduced by almost 2 log (Kilby et al.
1998). Because of the impracticability of twice-daily infusion,
2. Overview of Antiretroviral Drugs 103
Hoffmann, Kamps, et al.
the first study using subcutaneous application was initiated
shortly afterwards. 78 highly treatment-experienced patients
received T-20 in addition to stable HAART – either via an in-
sulin pump or twice-daily subcutaneously (Kilby et al. 2002).
Again, positive dose-related effects on viral load were shown in
both groups. However, maximal suppression was lower than
with infusions, and the maximum decrease was 1.6 log. More
importantly, the effect was short-lived; after 28 days, in most
cases viral load had returned to baseline levels. The main side
effects in this study were reactions (mostly mild) at the injec-
tion site.
Long-term evaluations have shown that the drug is also well
tolerated over longer periods of time (Lalezari et al. 2000). In
the T20-205 Study, 70 patients, mostly PI-experienced, re-
ceived 2 x 50 mg T-20 subcutaneously daily for 48 weeks. Only
few patients discontinued treatment prematurely due to side
effects. After 48 weeks, a constant effect on viral load was still
evident in at least one third of patients, but it became evident
that T-20 was of more benefit to patients who received other
new drugs for HAART at initiation of T-20. In the first Phase II
study (T20-206) for which 48-week data is available, the strat-
egy was therefore changed: 71 NNRTI-naive patients received
different doses of T-20 in addition to a new ART regimen (Lal-
ezari et al. 2002). In this study, the additional effect of T-20 was
weaker, but still present, although the study was not intended to
show differences between the individual groups. Nevertheless,
this study demonstrated that the simple addition of T-20 to an
otherwise unchanged treatment regimen would not be that bene-
ficial. Approximately two thirds of the T-20 patients in this
study had local reactions (mostly mild) at the injection site. T-
20 was generally well tolerated.
The preliminary and actually unexpected good data from the
first Phase III studies led to considerable media attention for T-
20 during the summer of 2002 (Henry et al. 2002, Clotet et al.
2002). TORO 1 (“T-20 versus optimized regimen only”, previ-
ously T20-301 Study) enrolled 491 patients in North America
104 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
and Brazil. Patients were randomized 2:1 to receive 2 x 90 mg
T-20 subcutaneously or not, on an optimized HAART regimen
(Henry et al. 2002). Almost all patients were heavily pre-treated
and harbored multiresistant viruses at entry. Again, the results
were astounding: The addition of T-20 clearly reduced viral
load compared to the optimized therapy “only”. At 24 weeks,
the reduction of viral load was 1.70 log in the T-20group, com-
pared to 0.76 log in the controls – a surprising difference of
0.93 log. In TORO 2 (T20-302), the same design was tested in
504 patients in Europe and Australia (Clotet et al. 2002). The
difference at 24 weeks was 1.43 versus 0.65 log – still a differ-
ence of 0.78 log.
Summary, evaluation and prospects of T-20: Patients with a
well-controlled viral load or who still have options with “classi-
cal” HAART would probably not require T-20 immediately.
For salvage therapy, however, the drug seems to be quite useful.
It must be stressed, however, that even in this setting wonders
cannot be achieved, the antiviral effect after one year being just
about one log. Although there is still no data from studies with
clinical endpoints, patients who currently have no other treat-
ment options might benefit clinically from this drug.
T-20 has been approved in the US in March 2003. The drug will
presumably not be available for all patients immediately, as
there are considerable logistical problems that still need to be
solved in manufacturing. According to Roche, this is one of the
most complicated drugs that the company has ever manufac-
tured: 106 steps are necessary for synthesis. This will presuma-
bly translate into high product pricing; one can thus expect that
the cost of a T-20-containing HAART regimen will be double
that of one without.
T-20 is certainly not as sensational as it has been portrayed over
the last 12 months. From a medical point of view, it is a major
breakthrough that this mechanism of action for inhibiting viral
replication actually works. A combination of different entry
inhibitors, which would hopefully act synergistically, both with
2. Overview of Antiretroviral Drugs 105
Hoffmann, Kamps, et al.
each other and with HAART, will likely inhibit HIV replication
more efficiently than traditional HAART.
T-1249 is the second fusion inhibitor to be developed and is
possibly more promising than T-20. T-1249 is a peptide that
binds to the hairpin-structure of the HIV envelope protein gp41
and consequently prevents fusion of the viral and host cell
membranes. The drug has favorable pharmacokinetics with a
once-daily dose as well as activity against T-20-resistant viruses
(Lambert et al. 1999). So far data is available from a Phase I/II
study (Eron et al. 2001, Gulick et al. 2002). 72 heavily pre-
treated HIV patients received T-1249 as monotherapy subcuta-
neously for 14 days, in doses ranging from 6.25 mg to 50 mg
per day (as a once- or twice-daily dose). A dose-related reduc-
tion of viral load was observed (maximum reduction – 1.4 log
with the 50 mg dose), with the plateau not yet reached. 40 % of
patients experienced a local reaction at the injection site; one
patient developed grade 4 neutropenia. Whether the one patient
who acquired a rash with fever had a hypersensitivity reaction
is still unclear. Selection of T-1249-resistant isolates is possible
in vitro.
Integrase inhibitors
The integrase is one of the three key enzymes encoded by the
HIV pol gene. This enzyme is involved in integration of viral
DNA into the host genome (Nair 2002). Integrase inhibitors
differ from entry inhibitors as they do not prevent entry of the
virus into the cell. Although human cells probably do not have
an integrase, the development of new and effective drugs in this
class is proving difficult, and progress is slow (Debyser et al.
2002). A good number of drugs have appeared over the last
years, only to disappear again just as quickly.
S-1360, which was developed by Shionogi/Glaxo, shows initial
promising data (Yoshinaga et al. 2002). In vitro (using an MTT
assay), S-1360 is effective against a variety of isolates, includ-
ing all NRTI- and PI-resistant mutants. There seems to be syn-
ergy with zidovudine, lamivudine, nevirapine and nelfinavir. In
106 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
animal studies (mice, rats, dogs), the drug has so far shown lit-
tle toxicity. The molecule is small, so that oral dosing is likely
to be possible. The drug was well tolerated in healthy volun-
teers (Fujiwara 2002).
Merck has also been working on integrase inhibitors. After
some difficulties, the first prototypes are now ready for testing
in clinical trials (Hazuda 2002). A new class, the naphthyridine-
7-carboxamides, shows good oral bioavailability. L-870812 and
L-870810 are currently the most promising drugs in this class.
In an animal model in SIV-infected monkeys, viral load de-
creased in 4 of 6 animals by more than one log. Phase I trials
were started based on this data.
References: Entry inhibitors, integrase inhibitors
1. Chen Z, Hu B, Huang W. HIV-1 mutants less susceptible to SCH-D, a
novel small-molecule antagonist of CCR5. Abstract 396, 9
th
CROI 2002,
Seattle, USA. http://63.126.3.84/2002/Abstract/13946.htm
2. Clotet B, Lazzarin A, Cooper D, et al. Enfuvirtide (T-20) in combination
with an optimized background (OB) regimen vs. OB alone in patients with
prior experience resistance to each of the three classes of approved
antiretrovirals in Europe and Australia. Abstract LbOr19A, XIV Interna-
tional AIDS Conference 2002, Barcelona, Spain.
3. Debyser Z, Cherepanov P, Van Maele B, et al. In search of authentic
inhibitors of HIV-1 integration. Antivir Chem Chemother 2002, 13:1-15.
http://amedeo.com/lit.php?id=12180645
4. Eron J, Merigan T, Kilby M, et al, for the T1249-101 Study Group. A 14-
day assessment of the safety, pharmacokinetics, and antiviral activity of
T-1249, a peptide inhibitor of membrane fusion. Abstract 14, 8th CROI
2001, Chicago, USA.
5. Franti M, Nagashima K, Maddon P, et al. The CCR5 co-receptor inhibitor
PRO 140 effectively controls established HIV-1 infection in vivo. Abstract
403, 9
th
CROI 2002, Seattle, USA.
http://63.126.3.84/2002/Abstract/13641.htm
6. Franti M, O’Neill T, Maddon P, et al. PRO 542 (CD4-IgG2) has a pro-
found impact on HIV-1 replication in the Hu-PBL-SCID mouse model. Ab-
stract 401, 9
th
CROI 2002, Seattle, USA.
http://63.126.3.84/2002/Abstract/13664.htm
7. Fujiwara T. Phase 1 multiple oral dose safety and pharmacokinetic study
of S-1360, an HIV integrase inhibitor with healthy volunteers. Abstract
TuPeB4431, XIV International AIDS Conference 2002, Barcelona, Spain.
8. Franti M, O’Neill T, Maddon P, et al. PRO 542 (CD4-IgG2) has a pro-
found impact on HIV-1 replication in the hu-PBL-SCID mouse model. Ab-
2. Overview of Antiretroviral Drugs 107
Hoffmann, Kamps, et al.
stract 401, 9
th
CROI 2002, Seattle, USA.
http://63.126.3.84/2002/Abstract/13664.htm
9. Gulick R, Eron J, Bartlett JA, et al. Complete analysis of T1249-101:
Safety, pharmacokinetics, and antiviral activity of T-1249, a peptide in-
hibitor of HIV membrane fusion. Abstract H-1075, 42nd ICAAC, San Di-
ego, USA.
10. Hazuda D. Integrase inhibitors. Abstract MoOrA137, XIV International
AIDS Conference 2002, Barcelona, Spain.
11. Hendrix C, Collier AC, Lederman M, et al. AMD-3100 CXCR4 receptor
blocker fails to reduce HIV viral load by > 1 log following 10-day continu-
ous infusion. Abstract 391, 9
th
CROI 2002, Seattle, USA.
http://63.126.3.84/2002/Abstract/13704.htm
12. Henry K, Lalezari J, O'Hearn M, et al. Enfuvirtide (T-20) in combination
with an optimized background (OB) regimen vs. OB alone in patients with
prior experience resistance to each of the three classes of approved
antiretrovirals in North America and Brazil. Abstract LbOr19B, XIV Inter-
national AIDS Conference 2002, Barcelona, Spain.
13. Jacobson JM, Lowy I, Fletcher CV, et al. Single-dose safety, pharmacol-
ogy, and antiviral activity of the HIV type 1 entry inhibitor PRO 542 in
HIV-infected adults. J Infect Dis 2000, 182:326-329.
http://amedeo.com/lit.php?id=10882617
14. Kilby JM, Hopkins S, Venetta TM, et al. Potent suppression of HIV-1
replication in humans by T-20, a peptide inhibitor of gp41-mediated virus
entry. Nat Med 1998, 4:1302-1307.
http://amedeo.com/lit.php?id=9809555
15. Kilby JM, Lalezari JP, Eron JJ, et al. The safety, plasma pharmacokinet-
ics, and antiviral activity of subcutaneous enfuvirtide (T-20), a peptide in-
hibitor of gp41-mediated virus fusion, in HIV-infected adults. AIDS Res
Hum Retroviruses 2002, 18:685-93.
http://amedeo.com/lit.php?id=12167274
16. Lalezari J, Cohen C, Eron J, and the T20-205 study group. Forty eight
week analysis of patients receiving T-20 as a component of multidrug
salvage therapy. Abstract LbPp116, XIII International AIDS Conference
2000, Durban, South Africa.
17. Lalezari J, DeJesus E, Northfelt D, et al. A week 48 assessment of a
randomized, controlled, open-label phase II trial (T20-206) evaluating 3
doses of T-20 in PI-experienced, NNRTI-naïve patients infected with HIV-
1. Abstract 418, 9th CROI 2002, Seattle, USA.
http://63.126.3.84/2002/Abstract/13592.htm
18. Lambert DM, Zhou J, Medinas R, et al. HIV-1 isolates from patients
treated with T-20 are sensitive to the second generation fusion inhibitor
T1249. Antiviral Ther 1999, 4 (suppl 1):8.
19. Lin PF, Guo K, Fridell R, et al. Identification and characterization of a
novel inhibitor of HIV-1 entry - II: Mechanism of Action. Abstract 10, 9th
CROI 2002, Seattle, USA. http://63.126.3.84/2002/Abstract/14126.htm
108 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
20. Lin PF, Robinson B, Gong YF, et al. Identification and characterization of
a novel inhibitor of HIV-1 entry - I: virology and resistance. Abstract 9, 9th
CROI 2002, Seattle, USA. http://63.126.3.84/2002/Abstract/14125.htm
21. Nair V. HIV integrase as a target for antiviral chemotherapy. Rev Med
Virol 2002, 12:179-93.
22. Reynes J, R. Rouzier R, Kanouni T. SCH C: safety and antiviral effects of
a CCR5 receptor antagonist in HIV-1- infected subjects. Abstract 1, 9
th
CROI 2002, Seattle, USA. http://63.126.3.84/2002/Abstract/14090.htm
23. Riley J, Wojcik L, Xu S, Strizki J. Genotypic and phenotypic analysis of in
vitro generated HIV-1 escape isolates to the CCR5 antagonist SCH-C.
Abstract 397, 9
th
CROI 2002, Seattle, USA.
http://63.126.3.84/2002/Abstract/12846.htm
24. Schols D, Claes S, De Clerq E, et al. AMD-3100, a CXCR4 antagonist,
reduced HIV viral load and X4 virus levels in humans. Abstract 2, 9
th
CROI 2002, Seattle, USA. http://63.126.3.84/2002/Abstract/13443.htm
25. Shearer W, Israel R, Starr S, et al., for the PACTG Protocol 351 Study
Team. Recombinant CD4-IgG2 in HIV type 1-infected children: phase 1/2
study. J Infect Dis 2000, 182:1774-1779.
http://amedeo.com/lit.php?id=11069253
26. Strizki JM, Xu S, Wagner NE, et al. SCH-C (SCH 351125), an orally
bioavailable, small molecule antagonist of the chemokine receptor CCR5,
is a potent inhibitor of HIV-1 infection in vitro and in vivo. Proc Natl Acad
Sci U S A 2001, 98:12718-12723. Original-Artikel:
http://www.pnas.org/cgi/content/full/98/22/12718
27. Trkola A, Ketas TJ, Nagashima KA, et al. Potent, broad-spectrum inhibi-
tion of HIV type 1 by the CCR5 monoclonal antibody PRO 140. J Virol
2001, 75:579-88. Original-Artikel:
http://jvi.asm.org/cgi/content/full/75/2/579?view=full&pmid=11134270
28. van Rij RP, Visser JA, Naarding M, et al. In vivo evolution of X4 HIV-1
variants in the natural course of infection coincides with reduced sensitiv-
ity to CXCR4 antagonists. Abstract 395, 9
th
CROI 2002, Seattle, USA.
http://63.126.3.84/2002/Abstract/12981.htm
29. Xu S, Wojcik L, Strizki J. Antagonism of the CCR5 Receptor by SCH-C
leads to elevated beta-chemokine levels and receptor expression in
chronically treated PBMC cultures. Abstract 398, 9
th
CROI 2002, Seattle,
USA. http://63.126.3.84/2002/Abstract/12848.htm
30. Yoshinaga T, Sato A, Fujishita T, Fujiwara T. S-1360: in vitro activity of a
new HIV-1 integrase inhibitor in clinical development. Abstract 8, 9
th
CROI 2002, Seattle, USA.
http://www.retroconference.org//2002/Abstract/13161.htm
2. Overview of Antiretroviral Drugs 109
Hoffmann, Kamps, et al.
Immunotherapy and its Relevance in Practice
In recent years, in addition to “conventional” ART, immuno-
logical treatment strategies have been investigated to an in-
creasing extent (reviews in: Mitsuyasu 2002, Sereti et Lane
2001). Increasing numbers of studies are being published on
approaches with interleukin-2 or hydroxyurea. All of these
therapies still lack proof of clinical benefit. Some important
approaches are nevertheless addressed briefly below.
Interleukin-2
Interleukin-2 (IL-2, aldesleukin, Proleukin
®
) is a cytokine that
is produced by activated T cells and leads to proliferation and
cytokine production by T cells, B cells and NK cells (review in:
Paredes et al. 2002). It has been employed in oncology for
years. IL-2 was already used in the early nineties, either intra-
venously or as a continuous infusion in HIV infected patients
(Wood et al. 1993). It is now usually administered subcutane-
ously.
The most important effect of IL-2 in HIV medicine is the rise in
CD4+ and CD8+ T cells, which in individual cases may be
quite impressive (Kovacs et al 1996). Several randomized
studies have consistently demonstrated significant increases in
CD4+ T cells. After administration of IL-2, CD45RO+ memory
cells initially increase, followed by naive CD45RA+ T cells.
The life span of CD4+ and CD8+ T cells may also be increased.
IL-2 is usually given in doses of 2 x 4.5 million I.E. subcutane-
ously over 5 days, in cycles 6-8 weeks apart (Davey et al. 2000,
Losso et al. 2000, Abrams et al. 2002, Lelezari et al. 2000,
Hengge et al. 1998). Daily treatment with a low dose has also
been investigated (review in: Smith 2001). After 24 to 48
weeks, increases in CD4+ cell count were 100-250 cells higher
in the IL-2 group than in the controls. Viral load was usually
unaffected by IL-2.
Unfortunately, the activation of T cells had no influence on vi-
ral reservoirs. Although the initial hope was that IL-2 could be
110 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
used to purge virus in the reservoirs and thereby “wash out”
latently infected cells from the body, (Chun et al. 1999), it is
now clear that this does not occur. In the German COSMIC
Study, 56 patients with more than 350 CD4+ T cells/µl on
HAART were randomized to receive IL-2 or placebo. Although
IL-2 led to a normalization of CD4+ cell count in significantly
more patients, IL-2 did not influence viral replication, proviral
DNA nor latently infected cells (Stellbrink et al. 1998, Stell-
brink et al. 2002).
In all larger studies, the combination of IL-2 with HAART has
so far demonstrated to be relatively safe. Nevertheless: the drug
has considerable side effects; fever, chills and sometimes severe
flu-like symptoms with myalgias are usually dose-limiting. The
side effects are the result of the IL-2-induced release of cytoki-
nes and invariably resolve 2-3 days after the last dose.
Paracetamol, rest and intake of electrolyte-rich solutions may be
helpful. The side effects, which are more severe than with inter-
feron, cannot be suppressed completely. Some researchers
question the rationale of IL-2 treatment, arguing that it might
just be the practice of lab cosmetics (“T cells ok, patient sick”).
In addition, doubts have been expressed with regard to the
quality of the immune response. Are the CD4+ T cells gener-
ated by IL-2 of the same quality as “normal” CD4+ T lympho-
cytes, and – more importantly – do the increases really prevent
AIDS? Do patients really have clinical benefit from these diffi-
cult IL-2 treatments? Little is also known about the long-term
use of IL-2 – the longest study performed to date lasted three
years (Gougeon et al. 2001).
Answers to these questions were expected from ESPRIT and
SILCAAT, the two ongoing multinational studies. Both were
intended to clarify over several years whether IL-2 has bona
fide clinical benefits. ESPRIT (http://www.espritstudy.org) is a
randomized study in which around 4,000 patients with at least
300 CD4+ T cells/µl are being treated. SILCAAT
(http://www.silcaat.com) enrolled patients with 50-299 CD4+ T
cells/µl and a viral load of < 10,000 copies/ml. 2,000 patients
2. Overview of Antiretroviral Drugs 111
Hoffmann, Kamps, et al.
were to be observed, initially for four years. After enrolment of
1,957 patients in 137 centers in 11 countries, the study was un-
fortunately stopped in October 2002, although the results of
SILCAAT (patients with low CD4+ cells!) would have been of
great importance for physicians and patients. The decision to
halt the study was probably a business decision: SILCAAT was
becoming too expensive for the manufacturer Chiron. While the
company is now trying to license the product with currently
available data (which might prove difficult), the scientific
committee under the leadership of Clifford Lane opposed the
discontinuation of the trial. Attempts are currently being made
to redirect the study into an academic investigation, so that data
from this important trial will not be lost.
The NIH-sponsored ESPRIT study will continue for the time
being.
All in all, IL-2 must still be viewed skeptically based on the
available data. In our opinion, only a few patients potentially
qualify for therapy with IL-2. These are patients with no immu-
nological response, patients whose CD4+ counts remain below
100/µl despite good viral suppression over longer periods of
time.
Hydroxyurea (Litalir
®
)
Hydroxyurea is an old chemotherapeutic agent with relatively
low toxicity, which is still being used today in hematology
(mostly in chronic myelogenous leukemia). It inhibits DNA
synthesis via the ribonucleotide reductase, and leads to an intra-
cellular shortage of deoxynucleotide triphosphates. A synergis-
tic effect on HIV replication in combination with didanosine
was demonstrated in 1994.
A randomized, double-blind study from Switzerland attracted
attention in 1998 (Rutschmann et al. 1998). The investigators
had treated 144 patients with hydroxyurea or placebo in addi-
tion to d4T+ddI. After 12 weeks, 54 % of hydroxyurea-treated
patients demonstrated a viral load below 200 copies/ml com-
pared to 28 % in the placebo group. Was this the discovery of a
112 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
new, cheaper option for HIV treatment? In the light of these
seemingly exciting results, the fact that the CD4+ T cell in-
crease in the hydroxyurea group was only 28 versus 107 cells/µl
in the placebo group had to be tolerated. Hydroxyurea was even
more in fashion after publication of the “Berlin-Patient”: a pa-
tient, who had been treated with hydroxyurea in addition to
indinavir and didanosine during acute infection, had stopped all
therapy after a few months and subsequently showed no detect-
able plasma viremia (Lisziewicz et al. 1999). Was this unex-
pected outcome due to hydroxyurea? Several smaller studies
from the US and Argentina seemed to confirm these positive
results, seen primarily in combination with didanosine (Hellin-
ger et al. 2000, Lori et al. 1999, Rodriguez et al. 2000). Many
treating physicians added the drug to ART, and even children
received hydroxyurea. Many already dreamed of a cheap com-
bination of ddI+HU for Africa.
These initial hopes subsided quite rapidly. Although the drug is
usually well tolerated, the combination with didanosine and sta-
vudine in particular seemed problematic. Data from early 2000
reported an additive effect, with a frequency of polyneuropathy
of almost 30/100 patient years (Moore et al. 2000). The ACTG
5025 Study (Havlir et al. 2001), in which hydroxyurea was
evaluated as a “stabilizer” of successful therapy (stable unde-
tectable viral load), led to the temporary demise of this drug in
HIV therapy. Three deaths on the combination of ddI+d4T
(+IDV) due to pancreatitis, all in the hydroxyurea group, were
reported. There was also a higher rate of treatment failure in
patients receiving hydroxyurea, probably due to toxicity rather
than to virological failure. The risk of pancreatitis on didano-
sine seems to be four times higher in combination with hy-
droxyurea (Moore et al. 2001). Randomized studies also failed
to show an effect in primary infection: obviously, further Ber-
lin-patients cannot simply be “reproduced”, at least not for hy-
droxyurea (Zala et al. 2002).
In October 1999, BMS received a warning from the FDA for
having too enthusiastically promoted hydroxyurea for HIV
2. Overview of Antiretroviral Drugs 113
Hoffmann, Kamps, et al.
therapy (http://hiv.net/link.php?id=164). We think that hy-
droxyurea should not be used outside clinical trials.
Interferon
The antiretroviral effect of interferon has been known for years
(Milvan 1996). The effect of 3 million I.E. daily s.c. is ca. 0.5-1
log (Haas et al. 2000). Higher dosing may pronounce the effect
further (Hatzakis et al. 2001). The antiviral effect of interferon
was initially not investigated in more depth because of the sub-
cutaneous delivery route and its side effects. There have re-
cently been indications that the drug may be useful for salvage
therapy. Pegylated interferons now allow for weekly admini-
stration, and improved efficacy with the pegylated drug is an-
ticipated in analogy to the studies in the setting of hepatitis C
infection. Schering-Plough is currently involved in trying to
license the product. However, there have been setbacks, as with
IL-2, and a pivotal multinational study in highly treatment-
experienced patients was aborted in October 2002 due to insuf-
ficient recruitment.
Other immunotherapies
The prototype of therapeutic vaccination already suffered dis-
aster years ago. Remune
®
, a therapeutic vaccine comprised of
an envelope-depleted (gp120) virus, which was developed by a
team headed by Jonas Salk, although indeed immunogenic,
does not seem to provide any clinical benefit (i.e., prolongation
of life and delay of disease progression). A large trial was inter-
rupted prematurely in May 1999 as no benefit had been demon-
strated for study participants. More than 2500 patients had
taken part for a mean of 89 weeks in this multinational study,
which was designed to evaluate the addition of Remune
®
to
HAART. In addition to the lack of clinical benefit not even ad-
vantages with respect to CD4+ cell count or viral load could be
shown (Kahn et al. 2000). The product is now probably obso-
lete, even though there have been dubious reports, mainly from
Thailand, claiming that some effect exists.
114 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
G-CSF and GM-CSF have frequently been used in HIV pa-
tients. G-CSF (granulocyte colony-stimulating factor or fil-
grastim) significantly reduces bacterial infections in HIV pa-
tients with neutropenia (Kuritzkes et al. 1998). G-CSF also sig-
nificantly improved survival in patients with CMV retinitis, al-
though the mechanisms were unclear (Davidson 2002). No ef-
fect on HIV viral load could be shown. GM-CSF (granulocyte-
macrophage colony-stimulation factor or sargramostim) showed
a slight effect on viral load in two double-blind randomized
studies (Skowron et al. 1999, Brites et al. 2000). However, such
approaches cannot be recommended outside of clinical studies.
Whether any clinical benefit exists remains unclear.
Cyclosporine A (Sandimmune
®
) – Immune activation may
lead to increased HIV replication, and an attractive treatment
hypothesis has been to suppress the immune system in an at-
tempt to slow down viral replication. This is the rationale be-
hind studies investigating the use of cyclosporine A. The drug is
normally used for prophylaxis of transplant rejection after allo-
genic organ transplantation. Between 1997 and 1999, 28 HIV
patients were recruited to receive cyclosporine A 4 mg/kg or
placebo daily for 12 weeks, with or without antiretroviral ther-
apy (two nucleoside analogs; Calabrese et al. 2002). The results
are easily summarized: cyclosporine A had no effect on CD4+
or CD8+ count, nor on expression of activation markers such as
CD38 or HLA-DR. Cyclosporine A therefore probably has no
future in the therapy of chronically infected HIV patients.
Whether, and how, cyclosporine A might improve treatment of
acute HIV infection needs to be clarified in further studies. Use
of both immunosuppressants (CsA) and immunostimulants (IL-
2) in this setting shows the clear discrepancy between scientific
knowledge and hope.
Mycophenol (Cellcept
®
) follows a concept similar to that of
hydroxyurea and cyclosporine A. Mycophenol inhibits the in-
osine monophosphate (IMP) dehydrogenase and is normally
used for prophylaxis of acute transplant rejection in patients
with allogeneic kidney, heart or liver transplantations, as well as
2. Overview of Antiretroviral Drugs 115
Hoffmann, Kamps, et al.
for some autoimmune diseases. Inhibition of lymphocyte prolif-
eration and reduction of target cells should theoretically inhibit
replication of HIV. First reports from small cohorts of patients
seem to demonstrate an effect on viral load in some cases (Mar-
golis et al. 2002, Press et al. 2002). Whether this will be con-
firmed by randomized trials seems uncertain.
Cannabinoids have no effect. A neatly designed study, in
which patients could either smoke marijuana or receive TCH
(dronabinol, Marinol
®
) or placebo in addition to HAART,
showed no effects on lymphocyte subpopulations or lympho-
cyte function after three weeks (Bredt et al. 2002).
Interleukin-12 – IL-12 stimulates T lymphocytes and NK cells
to generate a Th1-type immune response. In a randomized
Phase I study with rhIL-12 100 ng/kg 2 x/week, the drug was
well tolerated but had no effect on lymphocyte subpopulations,
antigen-specific immune response or viral load (Jacobson et al.
2002). Further development is therefore uncertain. The same
would appear to be true for interleukin-10 (Angel et al. 2000).
References
1. Abrams DI, Bebchuk JD, Denning ET, et al. Randomized, open-label
study of the impact of two doses of subcutaneous recombinant interleu-
kin-2 on viral burden in patients with HIV-1 infection and CD4+ cell
counts of >or=300/mm3: CPCRA 059. J Acquir Immune Defic Syndr
2002, 29: 221-31. http://amedeo.com/lit.php?id=11873071
2. Aladdin H, Ullum H, Katzenstein T, et al. Immunological and virological
changes in antiretroviral naive HIV infected patients randomized to G-
CSF or placebo simultaneously with initiation of HAART. Scand J Immu-
nol 2000, 51:520-5. http://amedeo.com/lit.php?id=10792845
3. Angel JB, High K, Rhame F, et al. Phase III study of granulocyte-
macrophage colony-stimulating factor in advanced HIV disease: effect on
infections, CD4 cell counts and HIV suppression. Leukine/HIV Study
Group. AIDS 2000, 14:387-95. http://amedeo.com/lit.php?id=10770541
4. Angel JB, Jacobson MA, Skolnik PR, A multicenter, randomized, double-
blind, placebo-controlled trial of recombinant human interleukin-10 in HIV-
infected subjects. AIDS 2000, 14:2503-8.
http://amedeo.com/lit.php?id=11101061
5. Bredt BM, Higuera-Alhino D, Shade SB, et al. Short-term effects of can-
nabinoids on immune phenotype and function in HIV-1-infected patients.
J Clin Pharmacol 2002, 42:82S-89S.
http://amedeo.com/lit.php?id=12412840
116 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
6. Brites C, Gilbert MJ, Pedral-Sampaio D, et al. A randomized, placebo-
controlled trial of granulocyte-macrophage colony-stimulating factor and
nucleoside analogue therapy in AIDS. J Infect Dis 2000, 182: 1531-5.
http://amedeo.com/lit.php?id=11023477
7. Calabrese LH, Lederman MM, Spritzler J, et al. Placebo-controlled trial of
Cyclosporin-A in HIV-1 disease: Implications for solid organ transplanta-
tion. J Acquir Immune Defic Syndr 2002, 29:359-362.
http://amedeo.com/lit.php?id=11917239
8. Chun TW, Engel D, Mizell SB, et al. Effect of interleukin-2 on the pool of
latently infected, resting CD4+ T cells in HIV-1-infected patients receiving
HAART. Nat Med 1999, 5:651-5. http://amedeo.com/lit.php?id=10371503
9. Davey RT JR, Murphy RL, Graziano FM, et al. Immunologic and virologic
effects of subcutaneous interleukin 2 in combination with ART: A ran-
domized controlled trial. JAMA 2000, 284: 183-9.
http://amedeo.com/lit.php?id=10889591
10. Davidson M, Min YI, Holbrook JT, et al. Use of filgrastim as adjuvant
therapy in patients with AIDS-related cytomegalovirus retinitis. AIDS
2002, 16: 757-65. http://amedeo.com/lit.php?id=11964532
11. Dybul M, Hidalgo B, Chun TW, et al. Pilot study of the effects of intermit-
tent interleukin-2 on HIV-specific immune responses in patients treated
during recently acquired HIV infection. J Infect Dis 2002; 185: 61-8.
http://amedeo.com/lit.php?id=11756982
12. Emery S, Abrams DI, Cooper DA, et al. The evaluation of subcutaneous
proleukin (interleukin-2) in a randomized international trial: rationale, de-
sign, and methods of ESPRIT. Control Clin Trials 2002; 23:198-220.
http://amedeo.com/lit.php?id=11943448
13. Gougeon ML, Rouzioux C, Liberman I, et al. Immunological and virologi-
cal effects of long term IL-2 therapy in HIV-1-infected patients. AIDS
2001, 15:1729-31. Abstract: http://amedeo.com/lit.php?id=11546950
14. Haas DW, Lavelle J, Nadler JP, et al. A randomized trial of interferon
alpha therapy for HIV type 1 infection. AIDS Res Hum Retrovir 2000,
16:183-90. http://amedeo.com/lit.php?id=10710206
15. Hatzakis A, Gargalianos P, Kiosses V, et al. Low-dose IFN-alpha mono-
therapy in treatment-naive individuals with HIV-1 infection: evidence of
potent suppression of viral replication. J Interferon Cytokine Res 2001,
21:861-9. http://amedeo.com/lit.php?id= 11710999
16. Havlir DV, Gilbert PB, Bennett K, et al. Effects of treatment intensification
with hydroxyurea in HIV-infected patients with virologic suppression.
AIDS 2001; 15: 1379-88. http://amedeo.com/lit.php?id=11504959
17. Hellinger JA, Iwane MK, Smith JJ, et al. A randomized study of the safety
and antiretroviral activity of hydroxyurea combined with didanosine in
persons infected with HIV type 1. J Infect Dis 2000, 181: 540-7.
http://amedeo.com/lit.php?id=10669337
18. Hengge UR, Goos M, Esser S, et al. Randomized, controlled phase II trial
of subcutaneous interleukin-2 in combination with HAART in HIV patients.
AIDS 1998; 12: F225-34. http://amedeo.com/lit.php?id=9863864
2. Overview of Antiretroviral Drugs 117
Hoffmann, Kamps, et al.
19. Jacobson MA, Spritzler J, Landay A, et al. A Phase I, placebo-controlled
trial of multi-dose recombinant human interleukin-12 in patients with HIV
infection. AIDS 2002, 16:1147-54.
http://amedeo.com/lit.php?id=12004273
20. Kahn JO, Cherng DW, Mayer K, Murray H, Lagakos S. Evaluation of HIV-
1 immunogen, an immunologic modifier, administered to patients infected
with HIV having 300 to 549 x 10(6)/L CD4 cell counts: A randomized
controlled trial. JAMA 2000, 284:2193-202.
http://amedeo.com/lit.php?id=11056590
21. Kovacs JA, Vogel S, Albert JM, et al. Controlled trial of interleukin-2 infu-
sions in patients infected with the HIV. N Engl J Med. 1996, 335:1350-6.
http://amedeo.com/lit.php?id=8857018
22. Kuritzkes DR, Parenti D, Ward DJ, et al. Filgrastim prevents severe neu-
tropenia and reduces infective morbidity in patients with advanced HIV
infection: results of a randomized, multicenter, controlled trial. AIDS 1998,
12:65-74. http://amedeo.com/lit.php?id=9456256
23. Lafeuillade A, Hittinger G, Chadapaud S, et al. The HYDILE trial: efficacy
and tolerance of a quadruple combination of reverse transcriptase inhibi-
tors versus the same regimen plus hydroxyurea or hydroxyurea and in-
terleukin-2 in HIV-infected patients failing PI-based combinations. HIV
Clin Trials 2002, 3:263-71. http:// amedeo.com/lit.php?id=12187499
24. Lalezari JP, Beal JA, Ruane PJ, et al. Low-dose daily subcutaneous in-
terleukin-2 in combination with HAART in HIV+ patients: a randomized
controlled trial. HIV Clin Trials 2000, 1:1-15.
http://amedeo.com/lit.php?id=11590500
25. Lisziewicz J, Rosenberg E, Lieberman J, et al. Control of HIV despite the
discontinuation of antiretroviral therapy. N Engl J Med 1999, 340:1683-4.
26. Lori F, Jessen H, Lieberman J, et al. Treatment of HIV infection with hy-
droxyurea, didanosine, and a protease inhibitor before seroconversion is
associated with normalized immune parameters and limited viral reser-
voir. J Infect Dis 1999, 180: 1827-32.
http://amedeo.com/lit.php?id=10558937
27. Lori F, Malykh A, Cara A, et al. Hydroxyurea as an inhibitor of HIV-type 1
replication. Science 1994, 266:801-5. http://
amedeo.com/lit.php?id=7973634
28. Losso MH, Belloso WH, Emery S, et al. A randomized, controlled, phase
II trial comparing escalating doses of subcutaneous interleukin-2 plus
antiretrovirals versus antiretrovirals alone in HIV-infected patients with
CD4+ cell counts /=350/mm3. J Infect Dis 2000, 181:1614-21.
http://amedeo.com/lit.php?id=10823761
29. Margolis DM, Kewn S, Coull JJ, et al. The addition of mycophenolate
mofetil to antiretroviral therapy including abacavir is associated with de-
pletion of intracellular deoxyguanosine triphosphate and a decrease in
plasma HIV-1 RNA. J Acquir Immune Defic Syndr 2002, 31:45-9.
http://amedeo.com/lit.php?id=12352149
30. Mildvan D, Bassiakos Y, Zucker ML, et al. Synergy, activity and tolerabil-
ity of zidovudine and interferon-alpha in patients with symptomatic HIV-1
118 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
infection: ACTG 068. Antivir Ther 1996; 1: 77-88.
http://amedeo.com/lit.php?id=11321183
31. Mitsuyasu R. Immune therapy: non-HAART management of HIV-infected
patients. J Infect Dis 2002, 185 (Suppl 2): S115-22.
http://amedeo.com/lit.php?id=12001032
32. Moore RD, Keruly JC, Chaisson RE. Incidence of pancreatitis in HIV-
infected patients receiving nucleoside reverse transcriptase inhibitor
drugs. AIDS 2001, 15:617-20.
33. Moore RD, Wong WM, Keruly JC, McArthur JC. Incidence of neuropathy
in HIV-infected patients on monotherapy versus those on combination
therapy with didanosine, stavudine and hydroxyurea. AIDS 2000, 14:
273-8. http://amedeo.com/lit.php?id=10716503
34. Paredes R, Lopez Benaldo de Quiros JC, Fernandez-Cruz E, Clotet B,
Lane HC. The potential role of interleukin-2 in patients with HIV infection.
AIDS Rev 2002; 4:36-40.
35. Press N, Kimel G, Harris M, et al. Case series assessing the safety of
mycophenolate as part of multidrug rescue treatment regimens. HIV Clin
Trials 2002, 3:17-20. http://amedeo.com/lit.php?id=11819181
36. Rizzardi GP, Harari A, Capiluppi B, et al. Treatment of primary HIV-1
infection with cyclosporin A coupled with HAART. J Clin Invest 2002,
109:681-688. http://amedeo.com/lit.php?id=11877476
37. Rodriguez CG, Vila J, Capurro AF,. Combination therapy with hydroxy-
urea versus without hydroxyurea as first line treatment options for antiret-
roviral-naive patients. HIV Clin Trials 2000, 1:1-8.
http://amedeo.com/lit.php?id=11590492
38. Rutschmann OT, Opravil M, Iten A, et al. A placebo-controlled trial of
didanosine plus stavudine, with and without hydroxyurea, for HIV infec-
tion. The Swiss HIV Cohort Study. AIDS 1998, 12: F71-7.
http://amedeo.com/lit.php?id=9631134
39. Rutschmann OT, Opravil M, Iten A, et al. Didanosine plus stavudine with
or without hydroxyurea in HIV-1-infected patients: 1 year follow-up. Antivir
Ther 1998, 3 (Suppl 4): 65-7. http://amedeo.com/lit.php?id=10723515
40. Sereti I, Lane HC. Immunopathogenesis of HIV: implications for immune-
based therapies. Clin Infect Dis 2001, 32: 1738-55.
http://amedeo.com/lit.php?id=11360217
41. Skowron G, Stein D, Drusano G, et al. The safety and efficacy of granu-
locyte-macrophage colony-stimulating factor (Sargramostim) added to
indinavir- or ritonavir-based antiretroviral therapy: a randomized double-
blind, placebo-controlled trial. J Infect Dis 1999, 180:1064-71.
http://amedeo.com/lit.php?id=10479132
42. Smith KA. Low-dose daily interleukin-2 immunotherapy: accelerating
immune restoration and expanding HIV-specific T-cell immunity without
toxicity. AIDS 2001, 15 Suppl 2: S28-35..
http://amedeo.com/lit.php?id=11424974
43. Stellbrink HJ, Hufert FT, Tenner-Racz K, et al. Kinetics of productive and
latent HIV infection in lymphatic tissue and peripheral blood during triple-
2. Overview of Antiretroviral Drugs 119
Hoffmann, Kamps, et al.
drug combination therapy with or without additional interleukin-2. Antivir
Ther 1998, 3: 209-14. http://amedeo.com/lit.php?id=10682140
44. Stellbrink HJ, van Lunzen J, Westby M, et al. Effects of interleukin-2 plus
highly active antiretroviral therapy on HIV-1 replication and proviral DNA
(COSMIC trial). AIDS 2002, 16:1479-87. http://
amedeo.com/lit.php?id=12131185
45. Wood R, Montoya JG, Kundu SK, Schwartz DH, Merigan TC. Safety and
efficacy of polyethylene glycol-modified interleukin-2 and zidovudine in
HIV type 1 infection: a phase I/II study. J Infect Dis 1993, 167:519-25.
http://amedeo.com/lit.php?id=8095058
46. Zala C, Salomon H, Ochoa C, et al. Higher rate of toxicity with no in-
creased efficacy when hydroxyurea is added to a regimen of stavudine
plus didanosine and nevirapine in primary HIV infection. J Acquir Immune
Defic Syndr 2002, 29: 368-73. http://amedeo.com/lit.php?id=11917241
120 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
3. Goals and Principles of Therapy
Christian Hoffmann
In the flood of monthly evaluations - including CD4+ count,
viral load, routine laboratory, genotypic and phenotypic resis-
tance testing, and drug plasma levels - the ultimate goal of
antiretroviral therapy should always be borne in mind:
To prolong the patient’s life,
while maintaining the best possible quality
of health and life.
This paradigm suggests that not only opportunistic infections
and malignancies, but also side effects of therapy, should be
prevented. Ideally, antiretroviral treatment should have little or
no influence on daily life. Even if a high CD4+ count and a low
viral load are useful therapeutic goals, the patient’s condition is
at least as significant as such laboratory results! Patients, too,
often lose focus on what really matters. The response to the
doctor’s query: "How are you?" is often accompanied by a
glance toward the CD4+ count result on the chart: "That’s what
I’d like to know from you!". It may therefore indeed be useful
to reflect upon – alone or with the patient – what one realisti-
cally wants to achieve.
Success and Failure of Treatment
Both success and failure of treatment can be evaluated with dif-
fering criteria – virological, immunological or clinical. Of these,
the earliest indicator is virological success or failure (decrease
or increase in viral load). This is followed, often a little later, by
immunological treatment success or failure (rise or fall in
CD4+ cell count). Clinical treatment failure usually becomes
apparent only much later – first the lab values deteriorate, then
3. Goals and Principles of Therapy 121
Hoffmann, Kamps, et al.
the patient! On the other hand, success of treatment may be
seen much earlier; many patients suffering from constitutional
symptoms rapidly improve on HAART. In the Swiss Cohort,
the incidence of opportunistic infections after only three months
on HAART was reduced from 15.1 to 7.7 per 100 patient years
(Ledergerber et al. 1999). For clinical treatment success, in par-
ticular in the prevention of AIDS, immunological success is
probably at least as important as virological success (Grabar et
al. 2000, Piketty et al. 2001).
Virological treatment success and failure
Virological treatment success is usually understood as a viral
load decrease to below the level of detection of 50 copies/ml.
This is based on the understanding that, the more rapid and
complete the decrease in viral load, the longer the therapeutic
effect (Kempf et al. 1998, Powderly et al. 1999, Raboud et al.
1998). In the INCAS Trial, the relative risk for treatment failure
(defined here as an increase to > 5,000 copies/ml) in patients
who had reached a viral load < 20 copies/ml was 20 times lower
than in those who had never reached a level below 400 cop-
ies/ml (Raboud et al. 1998). On HAART, viral load declines in
two phases (see also the chapter on "Monitoring"); there is an
initial very rapid decrease in the first weeks, followed by a
slower phase, in which plasma viremia is reduced only slowly.
A decrease to below the level of detection should be reached by
3-4 months; in cases of very high baseline viral load this may
take 4 or 5 months. A viral load above the level of detection
after 6 months of treatment is generally seen as failure. The
same is true if a rebound in viral load is confirmed by a second
determination after a short interval. In such cases, improve-
ments in therapy (e.g. compliance, change in the regimen)
should soon be considered.
The cut-off point of 50 copies/ml is arbitrary. It is based on the
currently available assays for measurement of viral load.
Whether 60 copies/ml are indeed worse than 30 copies/ml and
indicate a lesser success of treatment has not yet been proven At
122 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
these low levels, methodological inaccuracies must be taken
into account. A single viral load rebound ("blip") to low levels
(up to 1000 copies/ml) is often irrelevant (see below).
A viral load "below the level of detection" of 50 copies/ml
means just that – no more, no less. Numerous studies indicate
that replication and therefore development of resistance can
continue even with an undetectable virus load. 50 viral cop-
ies/ml indicate that 5 liters of blood contain 250,000 viruses; in
addition, even more actively replicating viruses are present in
the lymphatic organs. Thus, theoretically, measurable viremia,
even at very low levels, may possibly translate to a higher risk
of resistance in the long-term. Perhaps there is indeed a relevant
difference between 100 and 10 copies/ml with regard to risk for
developing resistance. But we just don’t know yet.
The good news: Morbidity and mortality may be lowered even
if the viral load is not decreased below the level of detection
(Mezzaroma et al. 1999, Deeks et al. 2000, Grabar et al. 2000).
This should be borne in mind when treating patients who have
only a limited number of treatment options. In such cases, it
may be useful to abandon viral load as a measure for success. In
patients with multiresistant viruses, virological success may not
be possible; here, stabilizing the CD4+ count should be of top
priority. Patients often remain immunologically stable for rela-
tively long periods of time, even with insufficient viral suppres-
sion.
The most important risk factors for virological treatment failure
are extensive pre-treatment with antiretroviral drugs (pre-
existing resistance mutations) and non-compliance (review:
Deeks et al. 2000). Whether viral load and CD4+ count at base-
line really play a role has not yet been proven conclusively; in
several cohorts no influence was detected (Cozzi Lepri et al.
2001, Phillips et al. 2001, Le Moing et al. 2002; see also the
chapter "When to start HAART".)
3. Goals and Principles of Therapy 123
Hoffmann, Kamps, et al.
How long does virological treatment success last?
Little is known about how long treatments remain effective.
Following the six years during which HAART has been em-
ployed, a surprisingly high number of adequately treated pa-
tients still have viral loads below the level of detection, even
after this time span. One of the few trials with a longer follow-
up period studied 336 antiretroviral-naive patients who had
reached a viral load below 50 copies/ml within 24 weeks (Phil-
lips et al. 2001). After 3.3 years, the risk of viral rebound was
relatively high at 25.3 %. More detailed analysis showed that a
large proportion of the patients experiencing viral rebound had
actually interrupted HAART. True virological failure was only
seen in 14 patients, which corresponds to a risk of 5.2 % after
3.3 years. Most importantly, the risk of virological failure de-
creased significantly with time. Thus, if treatment is not inter-
rupted, viral load may remain below the level of detection for
many years.
"Blips" – Do they mean virological failure?
Blips are transient increases in viral load. They occur in 20-40%
of patients and have been shown to be associated with a higher
level of viral replication. Blips often worry both patients and
clinicians. Strictly speaking, if one defines virological success
as < 50 copies/ml, blips signify treatment failure. However, in-
creasing data indicates that blips seem to have no consequences
in the medium-term, and do not necessarily indicate immuno-
logical or even clinical treatment failure (Havlir et al. 2001,
Moore et al. 2002, Sklar et al. 2002). This is true both for pa-
tients on first-line therapy and for highly treatment-experienced
patients. However, longer follow-up is still required to exclude
that patients with occasional blips are at more risk of develop-
ing resistance. In at least one recent analysis, the risk for treat-
ment failure after 18 months was approximately doubled
(Greub et al. 2002). Following presently available data, blips
should not necessitate an immediate change of therapy. They
can and should, however, raise the opportunity for a conversa-
124 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
tion with the patient on the subject of compliance. It should be
noted that viral load may also temporarily increase after immu-
nizations (Kolber et al. 2002).
Immunological treatment failure and success
Immunological treatment success is generally defined as an in-
crease in the CD4+ cell count. A more precise definition for
immunological treatment success does not currently exist. De-
pending on the study, increases by 50, 100 or 200 CD4+ T
cells/µl or increases to above 200 or 500 CD4+ T cells/µl are
defined as success. Failure is usually described as the absence
of an increase or decrease in the CD4+ T cell count in patients
receiving HAART.
Prediction of a rise in CD4+ count in patients on HAART is
difficult as there is significant individual variation. As with the
decrease in viral load, the increase in CD4+ count occurs in two
phases. After a first, usually rapid increase over the first three to
four months, further increases are considerably less pro-
nounced. In a prospective study involving some 1,000 patients,
CD4+ count in the first three months increased by a median of
21.2 CD4+ T cells/µl per month; in the following months the
increase was only 5.5 CD4+ T cells/µl (Le Moing et al. 2002).
There is inconclusive data as to whether lower CD4+ counts at
baseline result in a slower increase. However, normalization of
the CD4+ count (> 500 /µl) appears to be less likely and/or
takes longer, if the CD4+ count was low at initiation of therapy
(Kaufmann et al. 2002, Valdez et al. 2002). Immunological
treatment success is not necessarily linked to maximal viral
suppression; even partial suppression may lead to improvement
in the CD4+ T cell count (Kaufmann et al. 1998, Mezzaroma et
al. 1999). Neither is the level of initial viral load significant;
what seems to be crucial is that the viral load remains lower
than before treatment (Deeks et al. 2002).
3. Goals and Principles of Therapy 125
Hoffmann, Kamps, et al.
Discordant response
Failure to achieve one or two of the therapeutic goals – clinical,
immunological and virological - is referred to as a discordant
response. Some patients may have virological treatment success
without an immunological improvement, continuing to have a
very low CD4+ T cell count despite undetectable viral load
(Piketty et al. 1998, Renaud et al. 1999, Gabran et al. 2000,
Piketty et al. 2001). Conversely, HAART may be immunologi-
cally extremely effective and induce significant increases in the
CD4+ count, while the viral load remains detectable. The fre-
quencies of such discordant responses are outlined in the table
below.
Table 3.1: Prospective cohort studies, treatment response*
Response to HAART Piketty et al.
n = 150
Grabar et al.
n = 2236
Virological and immunological response 60 % 48 %
Discordant: only immunological response 19 % 19 %
Discordant: only virological response 9 % 17 %
No treatment response 12 % 16 %
* Immunological response: rise in CD4+ T cells > 100/µl after 30 months
(Piketty et al. 2001) or > 50/µl after 6 months (Grabar et al. 2000). Virological
response: continually at least 1 log below baseline or < 500 copies/ml (Piketty
et al. 2001) or < 1000 copies/ml (Grabar et al. 2000).
Immunological response is often moderate in comparison to
virological response, particularly in older patients. With in-
creasing age, the immune system becomes less capable of re-
generating, probably due to thymus degeneration (Lederman et
al. 2000). Various studies have demonstrated that the probabil-
ity of not achieving a rise in CD4+ count increases with patient
age and with progressive decrease in thymus size as detected by
CT (Goetz et al. 2001, Marimoutou et al. 2001, Piketty et al.
2001, Teixera et al. 2001, Viard et al. 2001).
126 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
Practical considerations in dealing with viral load and
CD4+ count
Viral load, since it can be directly affected, is the most im-
portant parameter in treatment monitoring.
If possible, use only one type of assay (in the same lab) –
keep in mind that there is considerable methodological
variability (up to half a log)!
Virological success should be monitored one month after
initiation or modification of HAART.
Viral load should be below 50 copies/ml after 3 months
(with high initial viral load, after 6 months at the latest) – if
it is not, check for the cause!
The greater the decrease in viral load, the more durable the
response to treatment.
Transient, low-level increases in viral load (blips) are often
of no significance – but VL should be remonitored at short
intervals (after 2-4 weeks).
The older the patient, the likelier a discordant response (low
viral load with no significant increase in CD4+ count).
In contrast to viral load, increase in CD4+ T cells, i.e. im-
munological success, is difficult to influence. CD4+ T cells
are probably more predictive of the individual risk for
AIDS.
Once CD4+ count is above 400-500/µl, controls can be per-
formed less frequently. Keep in mind that with higher
CD4+ counts, values may vary considerably from one
measurement to the next (which may cause the patient ei-
ther a false sense of euphoria or unnecessary concern).
3. Goals and Principles of Therapy 127
Hoffmann, Kamps, et al.
Clinical treatment success and failure
Clinical treatment success is dependent on virological and im-
munological therapeutic success. In individual patients, clinical
response is not always easy to assess. After all, there is no way
to show what might have occurred if treatment had not been
started. As an asymptomatic patient cannot feel any better, it
may be difficult to find good arguments to continue treatment in
the presence of side effects, which, at least temporarily, may
affect the quality of life.
Clinical success is almost always evaluated via clinical end-
points (AIDS-defining illnesses, death), although the improve-
ment on HAART in a patient with considerable constitutional
symptoms should also be seen as clinical success. With regard
to risk of disease progression, immunological response is at
least as important as virological response (Table 3.2).
Table 3.2: Risk of progression, as defined by immunological and virological
treatment response. See previous table caption for definitions. 95 % confi-
dence interval in parentheses.
Grabar et al. 2000 Piketty et al. 2001
CD4+ T cells at baseline (median) 150 73
Relative risk Relative risk
Virological and immunological re-
sponse
11
Immunological response only 1.6 (1.0-2.5) 6.5 (1.2-35.8)
Virological response only 2.0 (1.3-3.1) 9.7 (1.6-58.4)
No treatment response 3.4 (2.3-5.0) 51.0 (11.3-229.8)
The degree of virological response is also of great importance.
In the Swiss Cohort, 6.6 % of patients with a viral load con-
stantly below the level of detection suffered AIDS or died after
30 months. In contrast, AIDS or death occurred in 9.0 % of pa-
tients with viral rebound, and in 20.1 % of those whose viral
load never became undetectable (Ledergerber et al. 1999). The
importance of a complete and sustained virological treatment
response for clinical success has also been reported in other co-
horts (Salzberger et al. 1999, Thiebaud et al. 2000).
128 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
Clinical failure is usually defined as development of an AIDS-
associated condition or death. However, illness is not always
indicative of clinical treatment failure. A good example is the
immune reconstitution syndrome, where a pre-existing, sub-
clinical infection becomes apparent during the first weeks fol-
lowing initiation of antiretroviral therapy. On the other hand, if
a patient suffers serious side effects or even dies as a result of
them, this should also be seen as treatment failure.
What can be achieved today?
Every HIV clinician sees the remarkable strides made possible
by HAART reflected in his or her own patients (Table 3.3). In
many areas, the incidence of AIDS has been reduced to less
than a tenth (Mocroft et al. 2000). Today, in many Western
countries, almost all AIDS cases occur in patients who are not
being treated with antiretroviral therapy – usually because they
are unaware of their infection. The mortality rate has declined
to levels far below that of even a few years ago (Mocroft et al.
2002).
Table 3.3: Patient case (female, 41 years) demonstrating advances in treat-
ment due to HAART*
CD4+ T cells Viral load
Feb 95 AZT+ddC 23 (4 %) NA
Nov 96
AIDS: Toxoplasmosis, MAC,
Candida esophagitis
12 (1 %) 815.000
Feb 97 d4T+3TC+SQV 35 (8 %) 500
June 97 stopped HAART due to polyneu-
ropathy
July 97 AZT+3TC+IDV 17 (4 %) 141.000
Mar 98 147 (22 %) < 50
Mar 99 AZT+3TC+IDV+NVP 558 (24 %) 100
Mar 00 942 (31 %) < 50
Mar 02 1132 (33 % ) < 50
* Excellent immune reconstitution despite initially severe immunodeficiency and
several AIDS-defining illnesses. All primary/secondary prophylaxes (MAC,
Toxoplasmosis, PCP) have now been discontinued.
3. Goals and Principles of Therapy 129
Hoffmann, Kamps, et al.
Data from prospective, controlled studies on this dramatic
change is still relatively sparse, there being few randomized
trials with clinical endpoints (Hammer et al. 1997, Cameron et
al. 1998, Stellbrink et al. 2000). The results seen in these stud-
ies, which led to licensing of PIs, were also relatively modest,
due their design. In a multi-center trial, 1,090 clinically ad-
vanced patients received ritonavir liquid formulation or placebo
in addition to their ongoing treatment. Probability of AIDS and
death with a follow-up of 29 weeks was 21.9 % in the ritonavir
arm and nearly double (37.5 % ) in the placebo arm (Cameron
et al. 1998). In the SV14604 Study, the largest study of its kind
to date, involving 3,485 patients, the frequency of AIDS and
death was reduced by about 50 % in the group receiving
AZT+ddC+saquinavir hard gel, compared to the groups on dual
therapy (Stellbrink et al. 2000).
Due to the success of antiretroviral therapy, the number of
clinical endpoints that occur is fortunately now extremely low.
As a result, the duration of any contemporary study to prove
clinical benefit of one combination over another would have to
be very long. Only rarely will such investigations be undertaken
in the future (Raffi et al. 2001).
SILCAAT, a large multi-center study that had enrolled around
2,000 patients with less than 300 CD4+ T cells/µl was termi-
nated in October 2002 because the number of clinical endpoints
reached was too low to enable adequate detection of any differ-
ences.
With regard to opportunistic infections and malignancies, the
effect of HAART is equally apparent on their clinical course as
on their incidence. Illnesses such as cryptosporidiosis or PML
can be cured, while Kaposi's sarcoma can resolve completely
without specific therapy. Prophylaxis of pneumocystis, toxo-
plasma, CMV or MAC can usually be safely withdrawn. These
effects are discussed in more detail in the corresponding chap-
ters.
130 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
Table 3.4: Decline in morbidity and mortality in large cohorts
Where? (n) Patients (Period) Mortality
(/100 py)
Morbidity
(/100 py)
Palella
1998
USA (1255) < 100 CD4+ T
cells/µl
(1/94-6/97)
29.4 8.8 21.9 3.7*
Leder-
gerber
1999
Switzerland
(2410)
6 months before
versus 3 months
after HAART (9/95-
12/97)
NA
15.1 7.7
Mocroft
2000
Europe
(7331)
All (94-98) NA
30.7 2.5
Mocroft
2002
Europe
(8556)
All (94-01)
15.6 2.7
NA
* MAC, PCP, CMV. Mortality/Morbidity each per 100 py = patient years
References
1. Cameron DW, Heath-Chiozzi M, Danner S, et al. Randomised placebo-
controlled trial of ritonavir in advanced HIV-1 disease. The Advanced HIV
Disease Ritonavir Study Group. Lancet 1998, 351:543-9.
http://amedeo.com/lit.php?id=9492772
2. Cozzi Lepri A, Phillips AN, d'Arminio Monforte A, et al. When to start
HAART in chronically HIV-infected patients: evidence from the ICONA
study. AIDS 2001, 15:983-90. http://amedeo.com/lit.php?id=11399980
3. Deeks SG, Barbour JD, Grant RM, Martin JN. Duration and predictors of
CD4 T-cell gains in patients who continue combination therapy despite
detectable plasma viremia. AIDS 2002, 16: 201-7.
http://amedeo.com/lit.php?id=11807304
4. Deeks SG. Determinants of virological response to antiretroviral therapy:
implications for long-term strategies. Clin Infect Dis 2000, 30 Suppl 2:
S177-84. http://amedeo.com/lit.php?id=10860903
5. Freedberg KA, Losina E, Weinstein MC, et al. The cost effectiveness of
combination antiretroviral therapy for HIV disease. N Engl J Med 2001,
344:824-31. http://amedeo.com/lit.php?id=11248160
6. Goetz MB, Boscardin WJ, Wiley D, Alkasspooles S. Decreased recovery
of CD4 lymphocytes in older HIV-infected patients beginning HAART.
AIDS 2001, 15:1576-9. http://amedeo.com/lit.php?id=11504992
7. Grabar S, Le Moing V, Goujard C, et al. Clinical outcome of patients with
HIV-1 infection according to immunologic and virologic response after 6
months of HAART. Ann Intern Med 2000, 133: 401-10.
http://amedeo.com/lit.php?id=10975957
3. Goals and Principles of Therapy 131
Hoffmann, Kamps, et al.
8. Greub G, Cozzi-Lepri A, Ledergerber B, et al. Intermittent and sustained
low-level HIV viral rebound in patients receiving potent antiretroviral ther-
apy. AIDS 2002, 16:1967-9.
9. Hammer SM, Squires KE, Hughes MD, et al. A controlled trial of two
nucleoside analogues plus indinavir in persons with HIV infection and
CD4 cell counts of 200 per cubic millimeter or less. ACTG 320 Study
Team. N Engl J Med 1997, 337:725-33.
http://amedeo.com/lit.php?id=9287227
10. Havlir DV, Bassett R, Levitan D, et al. Prevalence and predictive value of
intermittent viremia with combination HIV therapy. JAMA 2001, 286:171-
9. http://amedeo.com/lit.php?id=11448280
11. Hosseinipur M, Cohen MS, Vernazza PL, Kashuba AD. Can antiretroviral
therapy be used to prevent sexual transmission of HIV type 1? Clin Infect
Dis 2002, 34: 1391-5. http://amedeo.com/lit.php?id=11981736
12. Kaufmann D, Pantaleo G, Sudre P, Telenti A. CD4-cell count in HIV-1-
infected individuals remaining viraemic with HAART. Swiss HIV Cohort
Study. Lancet 1998, 351:723-4.
13. Kaufmann G, Perrin L, Pantaleo G. CD4 T-lymphocyte recovery in indi-
viduals with advanced HIV-1 infection receiving potent antiretroviral ther-
apy for 4 years: The Swiss HIV Cohort Study. Abstract LB4, 9th CROI
2002, Seattle, USA.
http://www.retroconference.org//2002/Abstract/14064.htm
14. Kempf DJ, Rode RA, Xu Y, et al. The duration of viral suppression during
protease inhibitor therapy for HIV-1 infection is predicted by plasma HIV-
1 RNA at the nadir. AIDS 1998, 12: F9-14.
http://amedeo.com/lit.php?id=9543434
15. Kolber MA, Gabr AH, De La Rosa A, et al. Genotypic analysis of plasma
HIV-1 RNA after influenza vaccination of patients with previously unde-
tectable viral loads. AIDS 2002, 16: 537-42.
http://amedeo.com/lit.php?id=11872996
16. Kotler DP, Shimada T, Snow G, et al. Effect of combination antiretroviral
therapy upon rectal mucosal HIV RNA burden and mononuclear cell
apoptosis. AIDS 1998, 12:597-604.
http://amedeo.com/lit.php?id=9583599
17. Le Moing V, Chene G, Carrieri MP, et al. Predictors of virological rebound
in HIV-1-infected patients initiating a protease inhibitor-containing regi-
men. AIDS 2002, 16:21-9. http://amedeo.com/lit.php?id=11741159
18. Le Moing V, Thiebaut R, Chene G, et al. Predictors of long-term increase
in CD4(+) cell counts in HIV-infected patients receiving a protease in-
hibitor-containing antiretroviral regimen. J Infect Dis 2002, 185: 471-80.
http://amedeo.com/lit.php?id=11865399
Ledergerber B, Egger M, Erard V, et al. AIDS-related opportunistic ill-
nesses occurring after initiation of potent antiretroviral therapy: the Swiss
HIV Cohort Study. JAMA 1999, 282: 2220-6.
http://amedeo.com/lit.php?id=1060597
132 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
19. Ledergerber B, Egger M, Opravil M. et al. Clinical progression and vi-
rological failure on HAART in HIV-1 patients: a prospective cohort study.
Lancet 1999, 353: 863 –868.
20. Lederman MM, McKinnis R, Kelleher D, et al. Cellular restoration in HIV
infected persons treated with abacavir and a protease inhibitor: age in-
versely predicts naive CD4 cell count increase. AIDS 2000, 14: 2635-42.
http://amedeo.com/lit.php?id=11125881
21. Liuzzi G, Chirianni A, Bagnarelli P, et al. A combination of nucleoside
analogues and a protease inhibitor reduces HIV-1 RNA levels in semen:
implications for sexual transmission of HIV infection. Antivir Ther 1999,
4:95-9. http://amedeo.com/lit.php?id=10682154
22. Marimoutou C, Chene G, Mercie P, et al. Prognostic factors of combined
viral load and CD4+ cell count responses under triple antiretroviral ther-
apy, Aquitaine cohort, 1996-1998. J Acquir Immune Defic Syndr 2001,
27:161-7. http://amedeo.com/lit.php?id=11404538
23. Mezzaroma I, Carlesimo M, Pinter E, et al. Clinical and immunologic
response without decrease in virus load in patients with AIDS after 24
months of HAART. Clin Infect Dis 1999, 29:1423-30.
http://amedeo.com/lit.php?id=10585790
24. Mocroft A, Katlama C, Johnson AM, et al. AIDS across Europe, 1994-98:
the EuroSIDA study. Lancet 2000, 356:291-6.
http://amedeo.com/lit.php?id=11071184
25. Mocroft A, Brettle R, Kirk O, et al. Changes in the cause of death among
HIV positive subjects across Europe: results from the EuroSIDA study.
AIDS 2002, 16:1663-71. http://amedeo.com/lit.php?id= 12172088
26. Moore AL, Youle M, Lipman M, et al. Raised viral load in patients with
viral suppression on HAART: transient increase or treatment failure?
AIDS 2002, 16:615-8. http://amedeo.com/lit.php?id=11873005
27. Palella FJ JR, Delaney KM, Moorman AC, et al. Declining morbidity and
mortality among patients with advanced HIV infection. N Engl J Med
1998, 338:853-60. http://amedeo.com/lit.php?id=9516219
28. Phillips AN, Miller V, Sabin C, et al. Durability of HIV-1 viral suppression
over 3.3 years with multi-drug antiretroviral therapy in previously drug-
naive individuals. AIDS 2001, 15: 2379-84.
http://amedeo.com/lit.php?id=11740188
29. Phillips AN, Staszewski S, Weber R, et al. HIV viral load response to ART
according to the baseline CD4 cell count and viral load. JAMA 2001,
286:2560-7. http://amedeo.com/lit.php?id=11722270
30. Piketty C, Castiel P, Belec L, et al. Discrepant responses to triple combi-
nation antiretroviral therapy in advanced HIV disease. AIDS 1998,
12:745-50. http://amedeo.com/lit.php?id=9619806
31. Piketty C, Weiss L, Thomas F, et al. Long-term clinical outcome of HIV-
infected patients with discordant immunologic and virologic responses to
a protease inhibitor-containing regimen. J Infect Dis 2001, 183:1328-35.
http://amedeo.com/lit.php?id=11294663
3. Goals and Principles of Therapy 133
Hoffmann, Kamps, et al.
32. Powderly WG, Saag MS, Chapman S, et al. Predictors of optimal vi-
rological response to potent ART. AIDS 1999, 13:1873-80.
http://amedeo.com/lit.php?id=10513645
33. Raboud JM, Montaner JS, Conway B, et al. Suppression of plasma viral
load below 20 copies/ml is required to achieve a long-term response to
therapy. AIDS 1998, 12: 1619-24. http://amedeo.com/lit.php?id=9764780
34. Raboud JM, Rae S, Vella S, et al. Meta-analysis of two randomized con-
trolled trials comparing combined zidovudine and didanosine therapy with
combined zidovudine, didanosine, and nevirapine therapy in patients with
HIV. J Acquir Immune Defic Syndr 1999, 22:260-6.
http://amedeo.com/lit.php?id=10770346
35. Raffi F, Chene G, Lassalle R, et al. Progression to AIDS or death as end-
points in HIV clinical trials. HIV Clin Trials 2001, 2:330-5.
http://amedeo.com/lit.php?id=11590536
36. Renaud M, Katlama C, Mallet A, et al. Determinants of paradoxical CD4
cell reconstitution after protease inhibitor-containing antiretroviral regi-
men. AIDS 1999, 13:669-76. http://amedeo.com/lit.php?id=10397561
37. Salzberger B, Rockstroh J, Wieland U, et al. Clinical efficacy of protease
inhibitor based antiretroviral combination therapy--a prospective cohort
study. Eur J Med Res 1999, 4:449-55.
http://amedeo.com/lit.php?id=10585299
38. Sendi PP, Bucher HC, Harr T, et al. Cost effectiveness of HAART in HIV-
infected patients. Swiss HIV Cohort Study. AIDS 1999, 13:1115-22.
http://amedeo.com/lit.php?id=10397543
39. Sklar PA, Ward DJ, Baker RK, et al. Prevalence and clinical correlates of
HIV viremia ('blips') in patients with previous suppression below the limits
of quantification. AIDS 2002, 16:2035-41.
http://amedeo.com/lit.php?id=12370502
40. Stellbrink HJ, Hawkins DA, Clumeck N, et al. Randomised, multicentre
phase III study of saquinavir plus zidovudine plus zalcitabine in previously
untreated or minimally pretreated HIV-infected patients. Clin Drug Invest
2000, 20:295-307.
41. Teixeira L, Valdez H, McCune JM, et al. Poor CD4 T cell restoration after
suppression of HIV-1 replication may reflect lower thymic function. AIDS
2001, 15:1749-56. http://amedeo.com/lit.php?id=11579235
42. Thiebaut R, Morlat P, Jacqmin-Gadda H, et al. Clinical progression of
HIV-1 infection according to the viral response during the first year of
antiretroviral treatment. Groupe d'Epidemiologie du SIDA en Aquitaine
(GECSA). AIDS 2000, 14:971-8. http://amedeo.com/lit.php?id=10853978
43. Valdez H, Connick E, Smith KY, et al. Limited immune restoration after 3
years' suppression of HIV-1 replication in patients with moderately ad-
vanced disease. AIDS 2002, 16:1859-66.
http://amedeo.com/lit.php?id=12351945
44. Viard JP, Mocroft A, Chiesi A, et al. Influence of age on CD4 cell recovery
in HIV-infected patients receiving highly active antiretroviral therapy: evi-
134 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
dence from the EuroSIDA study. J Infect Dis 2001, 183: 1290-4.
http://amedeo.com/lit.php?id=11262215
3. Goals and Principles of Therapy 135
Hoffmann, Kamps, et al.
Eradication – Is it Feasible?
At this point in time, eradication of HIV in the sense of a cure is
unrealistic. Although as late as 1997, many still dreamt of
eradication, leading researchers now incline towards pessimism.
The problem lies in the pool of latently infected CD4+ T cells,
which probably comprise a lifelong reservoir. The half-life of
this reservoir is 44 months, and, according to recent estimates,
eradication could take 60-73 years (Finzi et al. 1999). Even af-
ter years of sufficient viral suppression to below 20-50 cop-
ies/ml, cellular viral transcription still takes place (Dornadula et
al. 1999, Furtado et al. 1999, Zhang et al. 1999, Sharkey et al.
2000). This holds in particular for blood cells, but also applies
to lymph nodes and sperm (Lafeuillade et al. 2001, Nunnari et
al. 2002). Different methods have been used to attempt to flush
out these latent reservoirs (IL-2, hydroxyurea, OKT), but all
have failed (Kulkosky et al. 2002, Pomerantz et al. 2002). At
the last World AIDS Conference, Bob Siliciano painted a bleak
picture of the situation (Siliciano 2002): Eradication is not pos-
sible with currently available drugs. The reservoirs cannot be
eliminated. Latently infected cells differ from uninfected cells
only in one minute detail, which is hardly detectable with pres-
ent methods and cannot be specifically targeted. Flushing out
the reservoirs or even just the complete elimination of memory
cells is either unsuccessful, too toxic, or far too dangerous.
Hopefully, future chapters can be dedicated to this issue.
Other Important Aspects of HAART
Besides the goals described above – virological, immunological
and clinical treatment success – several other aspects need to be
considered. Although not primary goals of HAART, they are
nevertheless extremely important: cost reduction; prevention;
and improving compliance – a constant challenge for every HIV
clinician.
136 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
Reduction of costs
Antiretroviral therapies are expensive. Single drugs cost be-
tween $250 and $1000 per month, depending on the drug and
the country where it is prescribed. Even within drug classes,
astonishing differences exist. In some countries, Crixivan
®
is
relatively cheap, at roughly half the price of Agenerase
®
. A
combination regimen with Trizivir
®
and Kaletra
®
adds up to at
least $2,000 per month. As a health care provider, it is therefore
important to have an idea of costs and to question the pricing
policy of some pharmaceutical companies. For example, why
does boosted Invirase
®
cost nearly twice as much as boosted
Fortovase
®
in Germany?
Despite such high costs, the positive effect of HAART remains
unquestioned. Reliable estimates assume an expenditure of
between $13,000 and $23,000 per additional QUALY (quality-
adjusted year of life; Freedberg et al. 2001) – relatively cheap in
comparison to many other therapies. HAART can avoid expen-
sive treatment of opportunistic infections, hospital and patient
care costs. In one German study, between 1997 and 2001, total
annual outgoings per patient decreased from €35,865 to
€24,482 (Stoll et al. 2002). Many patients are able to work
again, resulting in an overall economic reduction of costs
(Sendi et al. 1999).
As HAART is expensive, when treatment is being changed, ei-
ther merely to reduce pill burden or due to concern over long-
term toxicity, it is justified to ask a patient to first use up exist-
ing stocks. Privately insured patients are obviously appreciative
of this, but even patients on state health insurance should be
made aware of drug costs – not to cause guilt feelings or to
transfer insufficiencies of the health care system to the patient,
but rather to create an awareness of the value of this treatment.
Initially, only one box of tablets should be prescribed, even if,
for example, one box of Retrovir
®
prescribed at the standard
dose – 15 years after licensing! – still only lasts for 20 days.
Only by this approach can one avert a patient being left with
3. Goals and Principles of Therapy 137
Hoffmann, Kamps, et al.
many drugs should intolerance occur. Prescribing more than
three months of drugs at a time should be avoided.
Prevention
The lower the viral load, the less infectious is the patient. A
prospective study of 415 HIV-discordant couples in Uganda
showed that of 90 new infections over a period of up to 30
months, none occurred from an infected partner with a viral
load below 1500 copies/ml. The risk of infection increased with
every log of viral load by a factor of 2.45 (Quinn et al. 2000). In
a study from Thailand of 493 patients, this factor was 1.81 –
and here no case of infection was recorded below 1094 cop-
ies/ml (Tovanabutra 2002). Within limits, HAART can thus
serve as a preventive measure (Hosseinipur et al. 2002).
Most patients are interested in knowing: "Do I still need to use a
condom?". The answer is: "Yes!". Studies have shown that the
decrease of viral load in plasma and seminal fluid is roughly
parallel and that a decrease of several logs in plasma after sev-
eral months may also be seen in semen (Liuzzi et al. 1999).
Although the same seems to be true for the vaginal and anorec-
tal mucosa, individual risk remains difficult to estimate
(Lampinen et al. 2000, Cu-Uvin et al. 2000). Furthermore, viral
load levels in blood and other body fluids do not always corre-
late with one another (see also the chapter on "Monitoring").
In recent years the preventive effects of HAART seem to have
led to an increase in risk behaviour. Calculations have shown
that an increase in risk behaviour of only 10 % would offset the
effects of HAART (Blower et al. 2001, Law et al. 2001). In the
French PRIMO Cohort, so-called risk contacts of patients in-
creased from 5 % to 21 % between 1998 and 2001 (Desquilbet
et al. 2002). Small syphilis endemics among HIV infected indi-
viduals are now being reported in every major city in the USA
and Europe. Of equal concern is the increasing data on trans-
mission of multiresistant viruses.
138 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
Compliance as a goal of therapy
Compliance is the Achilles heel of antiretroviral therapy. Non-
compliance is a main, if not the most important, factor in treat-
ment failure. Insufficient plasma drug levels and partial sup-
pression of viral load are the conditions under which resistance
can develop.
Compliance is defined as consent and acceptance of a treatment
regimen by the patient. In the mid-90s a newer, more politically
correct term was adopted – "adherence". This term describes
both clinician and patient working together to achieve a treat-
ment concept acceptable for both, and emphasizes, quite cor-
rectly, that not only the patient may be responsible for treatment
failure. Adherence includes all factors that influence staying on
a regimen, in terms of "acceptability". Whichever term is used,
two facts remain:
1.
If 5 % of drugs are not taken, treatment success becomes
precarious.
2.
Clinicians usually overestimate the compliance of their pa-
tients.
"Risk patients" for non-compliance include not only individuals
with substance or alcohol abuse or those experiencing side ef-
fects. Studies on compliance have identified both patients with
depression or younger age as being particularly at risk (Murri et
al. 2001, Frank 2002). Positive factors are physician experience,
confidence of the patient in the positive effects of HAART, and
social support. Race, sex or stage of disease do not seem to be
relevant. The individual’s view of disease and health, accep-
tance of modern medicine and fear of side effects are further
considerations. However, all of these factors vary greatly, and
in the end compliance is difficult to predict in individual cases
(Lerner et al. 1998).
The importance of taking drugs regularly has been demon-
strated in numerous studies in recent years. In one study of 99
patients, in which compliance was evaluated by way of an elec-
tronic monitoring system, the rate of treatment failure was only
3. Goals and Principles of Therapy 139
Hoffmann, Kamps, et al.
22 % in patients with a level of compliance of at least 95 %
(95 % of doses taken). Failure rates in patients with 80-94 % or
< 80 % compliance were 61 % and 80 %, respectively. In this
study, 41 % of patients were misjudged by their treating clini-
cians with regard to compliance. Nurses seemed to have a better
understanding of their patients, judging incorrectly in only 30 %
of cases (Paterson et al. 2000). The importance of compliance is
also demonstrated by the successes reported in patients with
directly observed therapy (DOT). A DOT study performed in
one of Florida’s correctional facilities showed 100 % of sub-
jects with a viral load < 400 copies/ml at 48 weeks, compared
with 81 % in an unmonitored control group in the general
population (Fischl et al. 2001).
Non-compliance not only leads to virological failure. It also has
immunological consequences. In an analysis of two prospective
studies, patients with a compliance of 100 %, 80-99 % and 0-
79 % experienced reductions in viral load by 2.77, 2.33 and
0.67 log after one year. At the same time, the CD4+ cell count
rose by 179, 159 and 53, respectively (Mannheimer 2002).
Furthermore, non-compliance not only affects CD4+ count and
viral load, but also has clinical effects. In a Spanish study, pa-
tients who did not take more than 10 % of their drugs had a
four-fold increase in mortality risk (Garcia 2002). This data has
been confirmed in other studies (Maher et al. 1999, Hogg et al.
2000). Hospital stays are also less frequent in patients with high
compliance (Paterson et al. 2000). In addition, it should be con-
sidered that the risk of transmission of resistant viruses is in-
creased by non-compliant patients.
The basic mechanisms for development of resistance should be
explained to patients. One must emphasize that, in contrast with
other chronic illnesses, resistance mutations do not disappear
once they have developed. Diabetes and hypertension make ef-
fective examples: whereas these diseases may "tolerate" forget-
ting the occasional tablet, HIV is different – here even short-
term lapses can have irreversible consequences. Patients have to
be made aware of this unusual feature of HIV disease. Coop-
140 HIV Therapy 2003
HIV Medicine 2003 – www.HIVMedicine.com
eration with special treatment discussion groups offered by
various support organizations can be useful.
If compliance remains low
Despite all our efforts, some patients will not be able to im-
prove their compliance. Physicians and other health care pro-
viders are advised not to take this personally or to feel offended
should a patient not want to participate in the advances of medi-
cine. Even if it may be difficult to accept others’ views on life
and treatment, tolerance and acceptance should remain funda-
mental to the interactions of all health care providers with their
patients. Some providers, especially those who treat selective
patient populations in university settings, sometimes forget the
reality of routine medical practice. Rigidly upholding the prin-
ciples of modern medicine usually doesn’t help here, and put-
ting patients under pressure achieves even less.
The question of whether non-compliant patients should con-
tinue to be treated with antiretroviral therapy is not always easy
to address. On the one hand, there are patients who benefit even
from suboptimal therapy; on the other hand, drugs are expen-
sive and should not be prescribed too readily. When resources
are limited, available drugs should be used prudently. One
should also be beware of criminal dealings: there have recently
been several reports of patients who had deals with pharmacies
in order to receive other drugs (methadone, etc.), or even
money, in exchange for their prescriptions (black sheep are
ubiquitous!). Prescriptions have to be documented in the patient
chart. Where there are good reasons to doubt the compliance or
honesty of a patient, plasma drug levels can be determined.
3. Goals and Principles of Therapy 141
Hoffmann, Kamps, et al.
Twelve tips to improve compliance
1.
Every patient should receive a written (legible!) treatment
plan, which should be reviewed at the end of the visit. It
should include a telephone number to call in case of prob-
lems or questions.
2.
Patient and clinician should agree on the treatment plan.
The patient's concerns or critical questions should be dis-
cussed.
3.
The patient should have the impression that the treatment
regimen was not randomly chosen, but tailored to his/her
individual needs.
4.
The explanation of a new or modified treatment plan takes
time, and should not be rushed – all questions should be an-
swered.
5.
The reasons why compliance is so important should be ex-
plained. It makes sense to repeat such conversations – they
should not only take place when initiating or modifying
treatment, but should be part of routine care.
6.
Possible side effects should be explained, as well as what
can be done to alleviate them.
7.
Support groups and other types of assistance should be
utilized and offered.
8.
It is important to tell the patient to come back if any prob-
lems are encountered with HAART – it is better to solve
them together than have the patient try to deal with them
alone at home.
9.
The patient should know that the treatment regimen must be
taken in its entirety ("Last month I left out the big tablets").
10.
Prescriptions should be documented, in order to get a rough
idea of compliance. Irregularities should be addressed
openly.
11.
Especially during the early phases of therapy, the patient
should be informed of treatment success as seen by reduc-
tion of viral load and rise in CD4+ count.
12.
Treat depression!
Duesbergians