You are on page 1of 71

05/09/2020, 11)59 PM

emedicine.medscape.com

HIV Infection and AIDS


Updated: Aug 20, 2020
Author: Shelley A Gilroy, MD, FACP, FIDSA; Chief Editor: Michael Stuart Bronze, MD

Overview

Practice Essentials
Human immunodeficiency virus (HIV) is a blood-borne virus typically transmitted via sexual intercourse, shared
intravenous drug paraphernalia, and mother-to-child transmission (MTCT), which can occur during the birth process or
during breastfeeding. HIV disease is caused by infection with HIV-1 or HIV-2, which are retroviruses in the Retroviridae
family, Lentivirus genus. See the image below.

Electron microscopy of human immunodeficiency virus (HIV)–1 virions. Courtesy of CDC (Dr Edwin P Ewing, Jr).

Signs and symptoms

The patient with HIV may present with signs and symptoms of any of the stages of HIV infection. No physical findings are
specific to HIV infection; the physical findings are those of the presenting infection or illness. Manifestations include the
following:

Acute seroconversion manifests as a flulike illness, consisting of fever, malaise, and a generalized rash
The asymptomatic phase is generally benign

https://emedicine.medscape.com/article/211316-print Page 1 of 71
05/09/2020, 11)59 PM

Generalized lymphadenopathy is common and may be a presenting symptom


AIDS manifests as recurrent, severe, and occasionally life-threatening infections or opportunistic malignancies
HIV infection can cause some sequelae, including AIDS-associated dementia/encephalopathy and HIV wasting
syndrome (chronic diarrhea and weight loss with no identifiable cause)

The history should address risk factors for possible exposure to HIV, including the following:

Unprotected sexual intercourse, especially receptive anal intercourse


A large number of sexual partners
Previous or current sexually transmitted diseases (STDs)
Sharing of intravenous (IV) drug paraphernalia
Receipt of blood products (before 1985 in the United States)
Mucosal contact with infected blood or needle-stick injuries
Maternal HIV infection (for newborns, infants, and children)

See Clinical Presentation for more detail.

Diagnosis

HIV screening recommendations include the following:

The US Preventive Services Task Force (USPSTF) recommends that clinicians screen for HIV in all adolescents
and adults at increased risk for HIV infection, and all pregnant women [1]
The Centers for Disease Control and Prevention (CDC) recommends opt-out HIV screening for patients in all
health-care settings; persons at high risk for HIV infection should be screened at least annually [2]
The American College of Physicians (ACP) recommends that clinicians adopt routine screening for HIV and
encourage all patients to be tested [3]
Current CDC guidelines recommend testing for HIV infection with a US Food and Drug Administration (FDA)–
approved antigen/antibody immunoassay that detects HIV-1 and HIV-2 antibodies and the HIV-1 p24 antigen, with
supplemental testing following a reactive assay result to differentiate between HIV-1 and HIV-2 antibodies. If
supplemental testing for HIV-1/HIV-2 antibodies shows nonreactive or indeterminant results (or if acute HIV
infection or recent exposure is suspected or reported), an HIV-1 nucleic acid test is recommended to differentiate
acute HIV-1 infection from a false-positive test result. [4]
The World Health Organization (WHO) recommends an HIV testing strategy/algorithm whereby a combination of
rapid diagnostic tests (RDTs) and/or enzyme immunoassays (EIAs) are used to achieve at least a 99% positive
predictive value (ie, < 1 false-positive result per 100 people diagnosed with HIV infection). [5]

The CD4 T-cell count reliably reflects the current risk of acquiring opportunistic infections, as follows:

Reference range, 500-2000 cells/µL


Because CD4 counts vary, serial counts are generally a better measure of significant changes
After seroconversion, CD4 counts tend to decrease (~700/µL) and continue to decline over time
For surveillance, a CD4 count below 200/µL is considered AIDS-defining in the United States
In children younger than 5 years, the CD4 T-cell percentage is considered more important than the absolute count
(< 25% is considered to warrant therapy)
In adults with chronic hepatitis C and low absolute CD4 T-cells, the CD4 percentage may also be more useful [6]

Viral load in peripheral blood is used as a surrogate marker of viral replication rate; however, quantitative viral-load assays
should not be used as a diagnostic tool. Clinical relevance is as follows:

Rate of progression to AIDS and death is related to the viral load; patients with viral loads greater than 30,000/mL
are 18.5 times more likely to die of AIDS than those with undetectable viral loads.
With therapy, viral loads can often be suppressed to an undetectable level (< 20-75 copies/mL; optimal viral
suppression); complete inhibition of viral replication appears impossible and may be unnecessary
Successfully treated patients may demonstrate intermittent low-level viremia (eg, < 400 copies/mL), but this is not
thought to represent viral replication or to predict virologic failure (defined as a confirmed viral load of > 200

https://emedicine.medscape.com/article/211316-print Page 2 of 71
05/09/2020, 11)59 PM

copies/mL [7]

In August 2013, the FDA approved Alere Determine HIV-1/2 Ag/Ab Combo test (Orgenics, Ltd) as the first rapid HIV test
for the simultaneous detection of HIV-1 p24 antigen as well as antibodies to both HIV-1 and HIV-2 in human serum,
plasma, and venous or fingerstick whole blood specimens.[8, 9] The test does not distinguish between antibodies to HIV-1
and HIV-2, and is not intended to be used for screening of blood donors.[8, 9]

Baseline studies for other infections that are important in the initial workup of a patient with newly diagnosed HIV infection
include the following:

Purified protein derivative (PPD) skin testing for tuberculosis


Cytomegalovirus (CMV) testing
Syphilis testing
Rapid amplification testing for gonococcal and chlamydial infection
Hepatitis A, B, and C serology
Anti- Toxoplasma antibody
Ophthalmologic examination

The CDC classifies HIV infection into 3 categories, as follows[10] :

Category A: Asymptomatic HIV infection without a history of symptoms or AIDS-defining conditions


Category B: HIV infection with symptoms that are directly attributable to HIV infection (or a defect in T-cell–
mediated immunity) or that are complicated by HIV infection
Category C: HIV infection with AIDS-defining opportunistic infections

These 3 categories are further subdivided on the basis of the CD4+ T-cell count, as follows:

> 500/µL: Categories A1, B1, C1


200-400/µL: Categories A2, B2, C2
< 200/µL: Categories A3, B3, C3

See Workup for more detail.

Management

Current Department of Health and Human Services (DHHS) guidelines on the timing of initiation of antiretroviral therapy
are as follows:[11]

Antiretroviral therapy (ART) is recommended in all persons with HIV infection to reduce morbidity and mortality and
to prevent HIV transmission to others.
The Panel on Antiretroviral Guidance for Adults and Adolescents recommends initiating ART immediately (or as
soon as possible) after diagnosis to increase the uptake of ART linkage to care and to hasten and improve the rate
of viral suppression.
When initiating ART, it is important to educate patients regarding the benefits of ART and to deploy strategies to
optimize care engagement and treatment adherence.
Initiating ART is particularly important in patients with AIDS-defining conditions, patients with acute or recent HIV
infection, and pregnant patients. Delaying therapy in these subpopulations has been associated with high risks of
morbidity and mortality and HIV transmission.
Durable viral suppression improves immune function and overall quality of life, lowers the risk of both AIDS-defining
and non–AIDS-defining complications, and allows persons with HIV infection to live a lifespan approaching that of
persons without HIV infection. Two large randomized controlled trials, ART-START and TEMPRANO, demonstrated
reductions in morbidity and mortality among individuals with HIV infection who had CD4 T-lymphocyte (CD4) cell
counts of greater than 500 cells/uL and who were randomized to receive ART immediately compared with
individuals in whom ART initiation was delayed.
All persons with HIV infection should be informed that maintaining a plasma HIV RNA (viral load) of less than 200
copies/mL with ART prevents sexual transmission of HIV to their partners. Patients may recognize this concept as
“Undetectable = Untransmittable (U=U).” For persons with HIV infection who intend to rely on treatment as
prevention (TasP), providers should make an individual assessment of the person's risk tolerance, personal health,

https://emedicine.medscape.com/article/211316-print Page 3 of 71
05/09/2020, 11)59 PM

history of maintaining viral suppression with treatment, and access to healthcare services and ART, as well as other
factors that may affect their ability to maintain a high level of adherence to ART.

Highly active antiretroviral therapy (HAART) is the principal method for preventing immune deterioration. Classes of
antiretroviral agents include the following:

Nucleoside reverse transcriptase inhibitors (NRTIs)


Protease inhibitors (PIs)
Nonnucleoside reverse transcriptase inhibitors (NNRTIs)
Fusion inhibitors
CCR5 co-receptor antagonists (entry inhibitors)
HIV integrase strand transfer inhibitors
Entry inhibitors (CD4-directed post-attachment inhibitors)

Current DHHS guidelines list the below regimens as preferred in most treatment-naive patients.[11]

INSTI-based regimens are as follows:

Bictegravir/tenofovir alafenamide/emtricitabine (single-tablet regimen)


Dolutegravir/abacavir/lamivudine (single-tablet regimen) - Only for patients who are HLA-B*5701–negative and
without chronic hepatitis B virus (HBV) coinfection (In women of childbearing age, discuss the risks and benefits of
prescribing dolutegravir around the time of conception, including the low risk of neural tube defects [NTDs] and the
relative lack of information regarding the safety of using other commonly prescribed antiretrovirals [ARVs].)
Dolutegravir plus (emtricitabine or lamivudine) plus (tenofovir alafenamide or tenofovir disoproxil fumarate)
Raltegravir plus (emtricitabine or lamivudine) plus (tenofovir alafenamide or tenofovir disoproxil fumarate)
Dolutegravir plus lamivudine - Except in individuals with HIV RNA of more than 500,000 copies/mL, persons with
HBV coinfection, or patients in whom ART is to be started before the results of HIV genotypic resistance for reverse
transcriptase or HBV testing are available

The PI/r–based regimen is darunavir/ritonavir plus (tenofovir alafenamide or tenofovir disoproxil fumarate) plus
(emtricitabine or lamivudine).

HIV-2 is intrinsically resistant to NNRTIs and enfuvirtide.

To address individual patient characteristics and needs, the Panel also provides a list of Recommended Initial Regimens in
Certain Clinical Situations.[11]

Regimen selection is individualized based on the following:

Virologic efficacy
Toxicity
Pill burden
Dosing frequency
Drug-drug interaction potential
Drug resistance testing results
Comorbid conditions
Pregnancy

In particular cases, prophylaxis is indicated for specific opportunistic infections, including the following:

Pneumocystis jiroveci
Toxoplasma
Mycobacterium avium complex
Fungal and viral infections: Although prophylaxis for these infections is not routinely necessary, some recommend
fluconazole in patients with CD4 + T-cell counts under 50/µL to protect against cryptococcosis and endemic fungal
infections. Oral fluconazole is not recommended for routine primary prophylaxis against Candida infection.
Administration of ART and immune restoration are effective for preventing disease. Oral valganciclovir primary
prophylaxis for CMV infection is not recommended in patients who will be receiving or are not receiving ART. [12]

https://emedicine.medscape.com/article/211316-print Page 4 of 71
05/09/2020, 11)59 PM

Additional treatment measures include the following:

Treatment of opportunistic infections (directed at the specific pathogen)


Treatment of HIV lipodystrophy (tesamorelin)
Suppressive therapy for herpes simplex virus 2 (HSV-2) infection (acyclovir)
Treatment of HIV-associated diarrhea (crofelemer [13] )

The CDC has recommended HIV postexposure prophylaxis (PEP) and HIV pre-exposure prophylaxis (PrEP) regimens.
[12]

PEP drug regimens are as follows:

Preferred option 1: Dolutegravir plus tenofovir disoproxil fumarate/emtricitabine (TDF/FTC) (Truvada)


Preferred option 2: Raltegravir plus Truvada (TDF/FTC)
Alternative: Truvada (TDF/FTC ) plus darunavir (Prezista) plus ritonavir daily

PrEP drug regiments are as follows:

Preferred PrEP drug regimen: Truvada (TDF/FTC)


Alternative: Descovy (TAF/FTC)

See HIV Infection and AIDS Treatment & Management for more detail.

Background
Human immunodeficiency virus (HIV) is a blood-borne, sexually transmissible virus (see the image below.) The virus is
typically transmitted via sexual intercourse, shared intravenous drug paraphernalia, and mother-to-child transmission
(MTCT), which can occur during the birth process or during breastfeeding.

Electron microscopy of human immunodeficiency virus (HIV)–1 virions. Courtesy of CDC (Dr Edwin P Ewing, Jr).

https://emedicine.medscape.com/article/211316-print Page 5 of 71
05/09/2020, 11)59 PM

The most common route of infection varies from country to country and even among cities, reflecting the population in
which HIV was introduced initially and local practices. Co-infection with other viruses that share similar routes of
transmission, such as hepatitis B, hepatitis C, and human herpes virus 8 (HHV8; also known as Kaposi sarcoma herpes
virus [KSHV]), is common.

Two distinct species of HIV (HIV-1 and HIV-2) have been identified, and each is composed of multiple subtypes, or clades.
All clades of HIV-1 tend to cause similar disease, but the global distribution of the clades differs. This may have
implications on any future vaccine, as the B clade, which is predominant in the developed world (where the large
pharmaceutical companies are located), is rarely found in the developing countries that are more severely affected by the
disease.

HIV-1 probably originated from one or more cross-species transfers from chimpanzees in central Africa.[14] HIV-2 is
closely related to viruses that infect sooty mangabeys in western Africa.[15] Genetically, HIV-1 and HIV-2 are superficially
similar, but each contains unique genes and its own distinct replication process.

HIV-2 carries a slightly lower risk of transmission, and HIV-2 infection tends to progress more slowly to acquired immune
deficiency syndrome (AIDS). This may be due to a less-aggressive infection rather than a specific property of the virus
itself. Persons infected with HIV-2 tend to have a lower viral load than people with HIV-1,[16, 17] and a greater viral load is
associated with more rapid progression to AIDS in HIV-1 infections.[18, 19]

HIV-2 is rare in the developed world. Consequently, most of the research and vaccine and drug development has been
(perhaps unfairly) focused on HIV-1.

For information on HIV infection in children, see Pediatric HIV.

Initial description and early spread


In the United States, HIV disease was first described in 1981 among 2 groups, one in San Francisco and the other in New
York City. Numerous young homosexual men presented with opportunistic infections that, at the time, were typically
associated with severe immune deficiency: Pneumocystis pneumonia (PCP) and aggressive Kaposi sarcoma.[20]

HIV itself was not identified for another 2 years.[21] During that time, various other causes were considered, including
lifestyle factors, chronic drug abuse, and other infectious agents.[22] The HIV epidemic spread rapidly and silently in the
absence of testing.

However, clear clinical implications arose before society became aware of the disease; for example, prior to the
recognition of HIV, only one case of Pneumocystis pneumonia not clearly associated with immune suppression was
diagnosed in the United States between January 1976 and June 1980. In 1981 alone, 42 similar diagnoses were made,
and by December 1994, 127,626 cases of Pneumocystis pneumonia with HIV infection as the only identified cause of
immune suppression had been reported to the Centers for Disease Control and Prevention (CDC). Also, Kaposi sarcoma
is up to 30,000 times more likely to develop in persons with HIV infection than in immunocompetent persons.

The spread of HIV was retrospectively shown to follow the trucking routes across Africa from logging camps, and the
bush-meat trade combined with aggressive logging and improved transportation in the mid-20th century may have allowed
what was likely occasional cross-species transmission events to propagate across the country and, eventually, the globe.
[23]

Stigma of HIV infection

A considerable amount of stigma has been attached to HIV infection, mostly because of the virus's association with sexual
acquisition and the inference of sexual promiscuity. Consequences of this stigma have included discrimination and
reluctance to be tested for HIV infection. The stigma of HIV infection is also associated with a fear of acquiring a rapidly
fatal infection from relatively casual contact.

Such attitudes are inappropriate because HIV is poorly transmissible without sexual contact or blood contact. In addition,
the expected survival is long in patients with HIV infection who are receiving treatment. HIV is not transmitted during
casual contact and is readily inactivated by simple detergents. Much of the concern regarding HIV infection is due to the
incurability of the infection and the relentless immune decline and eventual premature death in the vast majority of infected

https://emedicine.medscape.com/article/211316-print Page 6 of 71
05/09/2020, 11)59 PM

people.

AIDS denialism

A small but vocal minority of people, including some scientists, continue to argue that HIV does not exist, or does not
cause AIDS, and that the HIV tests are unreliable or that the therapies are toxic. Such misinformation is usually based on
a lack of understanding of the scientific literature, deliberate misrepresentation, or logical fallacies based on
pseudoscientific arguments.

All of the arguments proposed by these dissenters have been addressed and rebutted in the scientific literature and public
discussion and even tested and rejected in the legal system. Nevertheless, they persist, and such views can have
extremely harmful effects on people who are exposed to HIV infection unnecessarily or who refuse treatment for their
progressing infection.

Clinicians should be aware of these issues, should be able and willing to address misinformation, and should direct their
patients to reliable sources of information.

Political denial and inaction have also likely caused considerable damage. Several governments in countries with high HIV
infection rates were slow to admit that they had an HIV epidemic, and at least one (South Africa) initially rejected that
AIDS was even a problem, then that the disease was caused by HIV infection, and, most recently, that antiretroviral
therapy was effective in treating HIV infection and preventing MTCT. Changes have now occurred but have been slow and
have cost hundreds of thousands of lives.

A regularly updated reference for addressing AIDS denial and misinformation can be found at AIDSTruth.org.

The quest for understanding of HIV

Since the discovery of HIV and its link to AIDS, great strides have been made in understanding its biology and in
developing effective treatments. The difficulty in dealing with HIV on a global scale is largely due to the fact that HIV
infection is far more common in resource-poor countries.

In the developed world, antiretroviral therapy has greatly improved prognosis and increased survival rates. Public
education programs have raised awareness such that testing and prevention of infection are more common. Both of these
approaches are difficult in countries with undereducated or underfunded populations.

A thorough discussion of the history of AIDS and the biologic link between HIV and AIDS can be found in an article entitled
" The relationship between the human immunodeficiency virus and the acquired immunodeficiency syndrome " at the
National Institute of Allergy and Infectious Diseases Web site. The document was originally written in September 1995,
prior to the advent of highly active antiretroviral therapy (HAART), which has significantly improved AIDS-free survival in
persons infected with HIV. This version was updated March 2010.

Patient confidentiality
HIV-related health information is typically considered separate from other health information and may require separate
consent to share or divulge.

Health care workers who are infected with HIV may be required to divulge their status to their employer or patients and
may be restricted in the types of procedures they can perform.

Pathophysiology
HIV produces cellular immune deficiency characterized by the depletion of helper T lymphocytes (CD4+ cells). The loss of
CD4+ cells results in the development of opportunistic infections and neoplastic processes.

https://emedicine.medscape.com/article/211316-print Page 7 of 71
05/09/2020, 11)59 PM

Virology of HIV

HIV-1 and HIV-2 are retroviruses in the Retroviridae family, Lentivirus genus. They are enveloped, diploid, single-stranded,
positive-sense RNA viruses with a DNA intermediate, which is an integrated viral genome (a provirus) that persists within
the host-cell DNA.

HIV contains 3 species-defining retroviral genes: gag, pol, and env. The gag gene encodes group-specific antigen; the
inner structural proteins. The pol gene encodes polymerase; it also contains integrase and protease (the viral enzymes)
and is produced as a C-terminal extension of the Gag protein). The env gene encodes the viral envelope—the outer
structural proteins responsible for cell-type specificity. Glycoprotein 120, the viral-envelope protein, binds to the host CD4+
molecule.

HIV-1 has 6 additional accessory genes: tat, rev, nef, vif, vpu, and vpr. HIV-2 does not have vpu but instead has the unique
gene vpx. The only other virus known to contain the vpu gene is simian immunodeficiency virus in chimpanzees (SIVcpz),
which is the simian equivalent of HIV.[14] Interestingly, chimpanzees with active HIV-1 infection are resistant to disease.
[24]

The accessory proteins of HIV-1 and HIV-2 are involved in viral replication and may play a role in the disease process.[25,
26] The outer part of the genome consists of long terminal repeats (LTRs) that contain sequences necessary for gene
transcription and splicing, viral packaging of genomic RNA, and dimerization sequences to ensure that 2 RNA genomes
are packaged. (See the image below.)

Genome layout of human immunodeficiency virus (HIV)–1 and HIV-2.

The dimerization, packaging, and gene-transcription processes are intimately linked; disruption in one process often
subsequently affects another. The LTRs exist only in the proviral DNA genome; the viral RNA genome contains only part of
each LTR, and the complete LTRs are re-created during the reverse-transcription process prior to integration into the host
DNA.

The biologic basis for AIDS

The specific details of the disease process that leads to AIDS are not fully understood despite considerable progress in
the virology of HIV and the immunology of the human host, much of which has been driven by the urge to better
understand AIDS.[27, 28, 29]

There is a specific decline in the CD4+ helper T cells, resulting in inversion of the normal CD4/CD8 T-cell ratio and
dysregulation of B-cell antibody production.[30, 31] Immune responses to certain antigens begin to decline, and the host
fails to adequately respond to opportunistic infections and normally harmless commensal organisms. Because the defect
preferentially affects cellular immunity, the infections tend to be nonbacterial (fungal, viral).

The pattern of opportunistic infections in a geographic region reflects the pathogens that are common in that area. For
example, persons with AIDS in the United States tend to present with commensal organisms such as Pneumocystis and
Candida species, homosexual men are more likely to develop Kaposi sarcoma because of co-infection with HHV8, and

https://emedicine.medscape.com/article/211316-print Page 8 of 71
05/09/2020, 11)59 PM

tuberculosis is common in developing countries.

Gut-associated lymphoid tissue (GALT) plays a role in HIV replication.[32] Although the portal of entry for HIV infection is
typically through direct blood inoculation or exposure of the virus to genital mucosal surfaces, the GI tract contains a large
amount of lymphoid tissue, making this an ideal site for HIV replication.

GALT has been shown to be a site of early viral seeding and establishment of the proviral reservoir. This reservoir
contributes to the difficulty of controlling the infection, and efforts to reduce the levels of HIV provirus through sustained
antiretroviral therapy (alone or in combination with interleukin-2 activation of resting HIV-infected T cells) have consistently
failed.[33]

A feature of HIV replication in GALT is that it is compartmentalized, even among different areas of the gut.[34]
Measurements of CD4+ T cells in GALT show relatively less reconstitution with antiretroviral therapy than that observed in
peripheral blood.[35, 36] At least one report has suggested that early treatment may result in better GALT CD4+ T-cell
recovery,[36] but clinical data generally argue against early initiation of therapy, which has not been shown to improve
long-term survival.

In addition, HIV replication can be detected even in patients with supposedly suppressed replication, as judged by plasma
viral load measurements. CD8+ killer T-cell responses to HIV occur in GALT and do not decline with antiviral therapy as
much as peripheral measurements do.[37] These findings underscore the limitations of peripheral measurements in what
is really a central viral replication.

One theory for the discrepancy between GALT and blood measurements is that ongoing viral replication in the lymphoid
tissue, and the resulting immune activation, may actually hamper efficient CD4+ T-cell replenishment.[38]

Studies of T-cell–replication kinetics have revealed that untreated HIV infection is characterized by rapid T-cell turnover
but a defect in T-cell replication from the thymus.[39, 40, 41] These changes can be reversed with effective long-term
antiviral therapy,[42, 43] suggesting that they are due to a direct effect of the virus or are a feature of the immune
response against HIV.

It is known that normal cell cycling is necessary to produce a normal cytokine profile[44] and that HIV causes cell-cycle
arrest.[45] Whether this is the exact mechanism is unresolved, however. Analysis of cytokine levels in HIV infected,
uninfected, and HAART-treated patients with HIV show that cytokines involved in T-cell homeostasis were definitely
affected, and therapy partially corrected these defects. In particular there was decreased IL-7, IL-12, IL-15 and FGF-2, and
increased TNF-alpha and IP-10.[46, 47]

Several of the HIV proteins directly affect T-cell function, either by disrupting cell cycling or down-regulating the CD4
molecule. The loss of T cells is clearly a primary issue, as the T-cell repertoire narrows in terms of which antigens the
immune system will recognize and respond to. Antiviral therapy is able to reverse these changes,[48] but the degree of
reversal is decreased if therapy is initiated very late in the infection and is further decreased when therapy is initiated when
CD4 T-cell counts are 200/µL and below.

Direct cytotoxic effects of viral replication are likely not the primary cause of CD4 T-cell loss; a significant bystander
effect[49] is likely secondary to T-cell apoptosis as part of immune hyperactivation in response to the chronic infection.
Infected cells may also be affected by the immune attack.

One interesting issue is that the co-receptor usage of the virus strains tends to change over time. The initial infection
nearly always involves a strain that uses the chemokine receptor 5 (CCR5), which is found on macrophages and dendritic
cells, as a co-receptor with CD4. People who are homozygous for deletions in the CCR5 gene (ie, CCR5-delta32) tend to
be resistant to infection,[50, 51] and those with heterozygosity for the polymorphism tend to show slower progression of
disease.[52]

Over time, the receptor usage shifts to chemokine-related receptor (CXCR4) and other related receptors found on CD4+ T
cells. These virus strains are more likely to cause cell fusion (syncytia formation). This trend is far from absolute but does
correlate in many people with disease progression.[53]

A single case report detailed a possible cure resulting from stem-cell transplantation from a CCR5-delta32 homozygous
donor (performed to treat acute myelocytic leukemia). Although this important finding is unlikely to impact routine

https://emedicine.medscape.com/article/211316-print Page 9 of 71
05/09/2020, 11)59 PM

management of HIV infection, it does suggest that reconstitution of a host immune system with a population of mutant
cells is a possible avenue of research to explore.[54]

Regardless of the cause for the disruption, a loss of thymic replacements in the face of an induced state of immune
activation and T-cell loss seems to be a key component of the mechanism by which HIV narrows the T-cell repertoire and
progresses to AIDS.[55, 56, 57]

Visible effects of HIV infection come in the form of disrupted lymph-node architecture. This disruption is temporal, and, at
one point, lymph-node biopsy was considered as a form of staging the disease.[58, 59] The disruption of the follicular
dendritic network in the lymph nodes and subsequent failure of normal antigen presentation are likely contributors to the
disease process.

HIV replicates in activated T cells (its promotor contains a nuclear factor kappa B [NF-kappa-B]–binding region, the same
protein that promotes other proteins in activated T cells and macrophages), and activated T cells migrate to the lymph
nodes. As such, much of the viral replication occurs outside of the peripheral blood, even though serum viral load is still a
useful surrogate marker of viral replication.

As mentioned above, with regards to GALT, HIV infection may be compartmentalized; specifically, areas of immune-
privilege may occur such as in the testes and central nervous system where not only will there be differences in HIV
pseudospecies but also different degrees of antiretroviral drug penetration. There is evidence that even with good
peripheral control of HIV, the virus may still be detectable in the CSF and semen of some infected patients.[60, 61]

Phases of HIV infection

Clinical HIV infection undergoes 3 distinct phases: acute seroconversion, asymptomatic infection, and AIDS. Each is
discussed below. (See the image below.)

Timeline of CD4 T-cell and viral-load changes over time in untreated human immunodeficiency virus (HIV) infection.
Courtesy of Wikipedia (based on an original from Pantaleo et al (1993)).

Acute seroconversion

Animal models show that Langerhans cells are the first cellular targets of HIV, which fuse with CD4+ lymphocytes and
spread into deeper tissues. In humans, rapid occurrence of plasma viremia with widespread dissemination of the virus is
observed 4-11 days after mucosal entrance of the virus.

There is no fixed site of integration, but the virus tends to integrate in areas of active transcription, probably because these
areas have more open chromatin and more easily accessible DNA.[62, 63] This greatly complicates eradication of the
virus by the host, as latent proviral genomes can persist without being detected by the immune system and cannot be
targeted by antivirals. See the image below.

During this phase, the infection is established and a proviral reservoir is created.[64, 65] This reservoir consists of

https://emedicine.medscape.com/article/211316-print Page 10 of 71
05/09/2020, 11)59 PM

persistently infected cells, typically macrophages, and appears to steadily release virus. Some of the viral release
replenishes the reservoir, and some goes on to produce more active infection.

The proviral reservoir, as measured by DNA polymerase chain reaction (PCR), seems to be incredibly stable. Although it
does decline with aggressive antiviral therapy, the half-life is such that eradication is not a viable expectation.

The size of the proviral reservoir correlates to the steady-state viral load and is inversely correlated to the anti-HIV CD8+
T-cell responses. Aggressive early treatment of acute infection lowers the proviral load, and treatment in newly infected
(but postseroconversion) patients yields long-term benefit.

At this point, the viral load is typically very high, and the CD4+ T-cell count drops precipitously. With the appearance of
anti-HIV antibodies and CD8+ T-cell responses, the viral load drops to a steady state and the CD4+ T-cell count returns to
levels within the reference range, although slightly lower than before infection.

Seroconversion may take a few weeks, up to several months. Symptoms during this time may include fever, flulike illness,
lymphadenopathy, and rash. These manifestations develop in approximately half of all people infected with HIV.

Asymptomatic HIV infection

At this stage in the infection, persons infected with HIV exhibit few or no signs or symptoms for a few years to a decade or
more. Viral replication is clearly ongoing during this time,[66] and the immune response against the virus is effective and
vigorous. In some patients, persistent generalized lymphadenopathy is an outward sign of infection. During this time, the
viral load, if untreated, tends to persist at a relatively steady state, but the CD4+ T-cell count steadily declines. This rate of
decline is related to, but not easily predicted by, the steady-state viral load.

Evidence now shows that therapy initiation early in the asymptomatic period is effective. However, very late initiation is
known to result in a less effective response to therapy and a lower level of immune reconstitution.

AIDS

When the immune system is damaged enough that significant opportunistic infections begin to develop, the person is
considered to have AIDS. For surveillance purposes in the United States, a CD4+ T-cell count less than 200/µL is also
used as a measure to diagnose AIDS, although some opportunistic infections develop when CD4+ T-cell counts are higher
than 200/µL, and some people with CD4 counts under 200/µL may remain relatively healthy.

Many opportunistic infections and conditions are used to mark when HIV infection has progressed to AIDS. The general
frequency of these infections and conditions varies from rare to common, but all are uncommon or mild in
immunocompetent persons. When one of these is unusually severe or frequent in a person infected with HIV and no other
causes for immune suppression can be found, AIDS can be diagnosed.[10]

Immunologic control of HIV

The primary mechanism for immunologic control of HIV appears to be CD8+ cytotoxic T-cells. T-cell responses are
correlated with the steady-state viral load and hence, the rate of progression.[67] Cellular immunity is apparently
responsible for some multiply-exposed, but uninfected individuals.[68, 69]

Although antibodies against HIV can be detected, it is clear that they are not sufficiently neutralizing to assist with
immunologic control of the infection.

The role of NK (Natural Killer) cells may be important in the initial control of HIV. Escape mutations have been detected,
implying that immunologic pressure on HIV exists from NK cells.[70]

Opportunistic infections and conditions

Even after starting therapy and with effective suppression of viral load, patients with persistently low CD4 counts remain at
high risk for opportunistic infections. In general, all patients remain at a relatively high risk for opportunistic infections and
other AIDS-related events for the first 6 months of antiretroviral therapy.[71] An observational study of 20,730 HIV patients
in Uganda found that, among patients with more than six months of follow-up after the initiation of antiretroviral therapy,

https://emedicine.medscape.com/article/211316-print Page 11 of 71
05/09/2020, 11)59 PM

the pre-therapy CD4 count was still predictive of mortality.[72]

Opportunistic infections and conditions include the following (*added in the 1993 AIDS surveillance case definition):

Candidiasis of bronchi, trachea, or lungs

Candidiasis, esophageal

Cervical cancer, invasive*

Coccidioidomycosis, disseminated or extrapulmonary

Cryptococcosis, extrapulmonary

Cryptosporidiosis, chronic intestinal (duration >1 mo)

Cytomegalovirus disease (other than liver, spleen, or nodes)

Cytomegalovirus retinitis (with vision loss)

Encephalopathy, HIV-related

Herpes simplex: chronic ulcer or ulcers (duration >1 mo) or bronchitis, pneumonitis, or esophagitis

Histoplasmosis, disseminated or extrapulmonary

Isosporiasis, chronic intestinal (duration >1 mo)

Kaposi sarcoma

Lymphoma, Burkitt (or equivalent term)

Lymphoma, immunoblastic (or equivalent term)

Lymphoma, primary, of the brain

Mycobacterium avium complex or Mycobacterium kansasii infection, disseminated or extrapulmonary

M tuberculosis infection, any site (pulmonary* or extrapulmonary)

Mycobacterium infection with other species or unidentified species, disseminated or extrapulmonary

Pneumocystis pneumonia

Pneumonia, recurrent*

Progressive multifocal leukoencephalopathy

Salmonella septicemia, recurrent

Toxoplasmosis of the brain

Wasting syndrome due to HIV infection

Although malaria is not typically considered an opportunistic infection, its incidence was found to be significantly higher
among children in Tanzania that were perinatally infected with HIV than those without HIV infection.[73] This was true for
physician-diagnosed clinical malaria, probable malaria involving laboratory testing for parasitemia as well as malaria that
was confirmed by blood smear.

There also appears to be an increased rate of anal cancer in high-risk groups (in particular, men who have sex with men).
This is unsurprising considering the link between anal cancer and human papillomavirus (HPV), and the fact that cervical

https://emedicine.medscape.com/article/211316-print Page 12 of 71
05/09/2020, 11)59 PM

cancer, also caused by HPV, is considered an AIDS-defining condition.[74]

HIV Encephalopathy is a severe condition usually seen in end-stage disease. Milder cognitive impairments may exist with
less advanced disease. For example, one study found significant deficits in cognition, planning, coordination and reaction
times in HIV-infected compared to uninfected children, effects that were more pronounced in those with higher viral loads.
[75]

Etiology
HIV disease is caused by infection with HIV-1 or HIV-2, both of which cause very similar conditions. They differ in
transmission and progression risks.

Epidemiology
United States statistics

According to the Centers for Disease Control and Prevention (CDC), from 2013-2017, the estimated rate of HIV infection
diagnosis in all 50 US states and District of Columbia decreased. The annual number of diagnoses remained stable. In
2018, the rate was 11.4 per 100,000 population,[76] and 37,832 individuals were diagnosed with HIV infection that year.
Numbers and rates of HIV infection increased in some subgroups and decreased in others. Variations in trends among
groups are expected and may result from differences in testing behaviors, targeted HIV testing initiatives, and/or changes
in the numbers of new HIV infections in some subgroups.[77]

From 2013-2017, the annual number and rate of HIV infections classified as stage 3 (AIDS) in the United States
decreased. In 2018, the rate of infections classified as stage 3 (AIDS) was 5.2 per 100,000 population. The number and
rate of deaths among persons with infection ever classified as stage 3 (AIDS) remained stable. In 2017, the rate of deaths
in persons with stage 3 (AIDS) was 3.9 per 100,000 population. Deaths among persons with stage 3 (AIDS) may be due to
any cause.[76]

US rates vary by state. See the latest CDC surveillance report for full details.

The overall figures may give a false impression that the HIV epidemic is relatively homogeneous. In fact, the HIV epidemic
is best viewed as numerous separate epidemics among distinct risk groups, although the various epidemics clearly have
some level of overlap. In any given area, the infection may be most prevalent among users of intravenous drugs who
share needles. In another, the main risk group may be men who have sex with other men. And in yet another, the main
risk group may be female sex workers.

These sub-epidemics each follow their own pattern, although there is some degree of interdependence. Early on, nearly
all cases of HIV infection detected in the Western Hemisphere were in homosexual men, but the spread of the disease to
female partners of bisexual men with HIV infection gave rise to an increased rate among heterosexual persons.

Contributing to the increased cross-prevalence were persons with hemophilia who had been infected with HIV from
contaminated factor VIII concentrate and persons who used intravenous drugs, an activity that transcends all sexual
preferences. In 2014, 70% of new HIV infections were reported in homosexual men, and infected heterosexual women
outnumber infected heterosexual men nearly two to one.[77]

One community-based study targeting areas where men who have sex with men (MSM) meet demonstrated that an
average of 44% of study participants appeared unaware of their HIV-positive status. High rates of positivity and
unawareness of positive status were associated with younger participants, men of black non-Hispanic race, and lower

https://emedicine.medscape.com/article/211316-print Page 13 of 71
05/09/2020, 11)59 PM

education levels.

Healthcare visits in the preceding year were associated with a lower rate of unawareness (37% vs 81%) but a higher rate
of HIV-positivity (21% vs 12%). Because this study targeted a high-risk group and may involve participation bias, the
overall rate of HIV infection (19%) cannot be easily extrapolated to the overall population.[78]

Mortality from HIV disease has not been among the 15 leading causes of death in the US since 1997. The age-adjusted
death rate for HIV disease peaked in 1995 at 16.3 per 100,000 population, decreased 69.9% through 1998, then further
decreased 30.2% from 1999 through 2007, to 3.7 per 100,000 population. In 2007, a total of 11,295 persons died from HIV
disease. However, HIV disease has remained among the 5 leading causes of death for specific age groups for females,
and in the black population.[79]

Adolescents and young adults

From 2013-2017, CDC HIV surveillance statistics show that rates among children (< 13 years) and persons aged 13-24
years decreased. In 2018, the highest rate (32.4 per 100,000 population) of new HIV infections in the United States were
in adults aged 25-29 years, followed by adults aged 20-24 years (27.6 per 100,000 population). Among all adults and
adolescents, males accounted for 81% of new HIV infections.[76] The highest rates per 100,000 population were 39.3 in
blacks, followed by 16.2 in Hispanics/Latinos, 12.4 in persons of multiple races, 11.8 in native Hawaiians/other Pacific
Islanders, 7.8 in American Indians/Alaskan natives, 4.9 in whites, and 4.7 in Asians. Male-to-male sexual contact
accounted for 72.1% (8800 individuals) and 3% for male-to-male sexual contact and injection drug use.[76] The
percentage of youths tested for HIV infection was 12.9% in high-school students and 34.5% in individuals aged 18-24
years. Testing was lower in males than females. More than half (59.5%) of youths with HIV infection are unaware of their
infection.[80]

International statistics

According to the Joint United Nations Programme on HIV/AIDS (UNAIDS),[81] worldwide in 2018, approximately 37.9
million people were infected with HIV. UNAIDS estimates that approximately 1.7 million people were newly infected with
HIV and that 770,000 people died of AIDS in 2018, both statistics showing a decline over time.

The vast majority of infections remain in sub-Saharan Africa, where 5.2% of the population is believed to be infected.
Between 2004 and 2006, the prevalence of HIV infection in central and eastern Asia and Eastern Europe increased by
21%. During this period, the number of new HIV infections in persons aged 15 to 64 years rose by 70% in Eastern Europe
and central Asia.

The infection rates in many developed countries remain stable, and some developing countries have achieved significant
gains in controlling and even reversing the effects of the HIV epidemic. However, this is partially due to deaths in HIV-
infected people, together with simultaneous prevention of new infections. India, for example, has used a national
prevention campaign focusing on high-risk populations that may have prevented 100,000 new HIV infections over the 5
years it has been implemented, with increasing results seen in areas with higher levels of investment.[82] These figures
together show that global HIV infection is in a state of flux.

Men who have sex with men (MSM) are still 28 times more likely than heterosexuals to contract HIV infection despite
sharp declines of infection among such populations in Western countries. The institution of pre-exposure prophylaxis
(PrEP) in Western Europe, North America, and Australia has dramatically decreased transmission rates among gay men
in those areas.[83]

The mortality rate in some countries has greatly increased. In South Africa (a country that, despite having a relatively late-
onset HIV epidemic, has developed one of the highest prevalence rates), the all-cause HIV-associated mortality rate
increased by 79% between 1997 and 2004. In women aged 25-34 years, mortality rates increased by 500% during this
period.

Swaziland has the highest overall prevalence of HIV infection (>26% of all adults based on 2007 figures).

The Ministry of Health in Zambia predicts that, without therapy and assuming current levels of prevalence, young adults
have a 50% lifetime risk of dying from AIDS.

https://emedicine.medscape.com/article/211316-print Page 14 of 71
05/09/2020, 11)59 PM

In developing nations, co-infection with HIV and tuberculosis is very common. The immunosuppressed state induced by
HIV infection contributes not only to a higher rate of tuberculosis reactivation but also to an increased disease severity, as
with many other opportunistic infections.

Further details of the global epidemic can be found in the UNAIDS Global HIV & AIDS statistics — 2019 fact sheet.

Racial, sexual, and age-related differences in incidence

In the United States, the rate of HIV infection is highest in blacks (44.3 cases per 100,000 population). The prevalence is
also high among Hispanic persons (16.4 per 100,000 population).[77] These increased rates result from socioeconomic
factors rather than genetic predisposition.

In the developed world, HIV infection is much more common in males. In 2015, males accounted for 81% of all diagnoses
of HIV infection among adults and adolescents in the United States.[77] Among heterosexuals, females are more likely to
acquire HIV infection from an infected male than a male is from an infected female, but a large proportion of infections in
males are due to homosexual contact, with or without injection drug use. Males are also more likely to acquire HIV
infection from injection drug use alone.

Males were also more likely to acquire HIV infection through contaminated blood products for treatment of hemophilia
before universal testing of the blood supply was instituted. The risk of HIV exposure from factor VIII concentrates has been
virtually eliminated by viricidal treatment of plasma-derived factor VIII concentrates, as well as the introduction of
recombinant factor VIII concentrates and the gradual elimination of albumin from the production process used for these
products.

In the developing world, HIV infection is equally common in males and females. The primary route of HIV transmission in
the developing world is heterosexual contact.

Young adults tend to be at higher risk of acquiring HIV, typically through high-risk activities such as unprotected sexual
intercourse or intravenous drug use. In 2009 in the US, the largest percentage (15% of all diagnoses) and the highest rate
(36.9 per 100,000 population) were in persons aged 20–24 years.[77]

Children may become infected by transplacental transmission or by breastfeeding. Rare cases of children infected after
sexual abuse by HIV-infected adults have also been reported.

Prognosis
The prognosis in patients with untreated HIV infection is poor, with an overall mortality rate of more than 90%. The
average time from infection to death is 8-10 years, although individual variability ranges from less than 1 year to long-term
nonprogression. Many variables have been implicated in HIV's rate of progression, including CCR5-delta32
heterozygosity, mental health,[84] concomitant drug or alcohol abuse, superinfection with another HIV strain, nutrition, and
age.

There is less evidence that treatment of HIV-2 infection slows progression, and certain antiretroviral medications
(specifically the non-nucleoside–analogue reverse-transcriptase inhibitors) are not effective against HIV-2. The HIV-1 viral-
load assays are much less reliable at quantifying HIV-2, if they work at all. HIV-2 viral load assays have been developed,
but none has been approved by the US Food and Drug Administration except as blood donor–screening tools.

Once infection has progressed to AIDS, the survival period is usually less than 2 years in untreated patients. Persons in
whom the infection does not progress long-term may not develop AIDS for 15 years or longer, although many still exhibit
laboratory evidence of CD4 T-cell decline or dysfunction.[85, 86, 87, 88]

The appropriate use of combination antiretroviral therapies and prophylaxis for opportunistic infections dramatically
improves survival and greatly decreases the risk of secondary opportunistic infections.[89, 90, 91] The risk of AIDS-

https://emedicine.medscape.com/article/211316-print Page 15 of 71
05/09/2020, 11)59 PM

associated lymphoma is not altered by antiviral therapy and, as such, has grown in prevalence among overall AIDS-
defining conditions.

Sackoff et al found that between 1999 and 2004, the HIV-related mortality rate in New York City decreased each year by
approximately 50 deaths per 10,000 people with AIDS. The rate of non–HIV-related deaths also showed a decline, more
modest but consistent, with about 7.5 fewer deaths per 10,000 people with AIDS per year.[90]

Importantly, many researchers have consistently shown that the primary risk factor for infection affects mortality. For
example, the mortality rate among intravenous drug users tends to be higher, whether related to HIV disease or non-HIV
disease.

Overall, with the increasing use of antiretroviral therapy and the introduction of better antiviral regimens, survival with HIV
infection has increased over time, although it is not yet equivalent to that in uninfected individuals. (See the image below.)

Changes in survival of people infected with HIV. As therapies have become more aggressive, they have been more
effective, although survival with HIV infection is not yet equivalent to that in uninfected people. Modified from Lohse N et
al. Survival of persons with and without HIV infection in Denmark, 1995-2005. Ann Intern Med. 2007;146(2):87-95.

In addition to the concern for new opportunistic infections, pre-existing infections can reactivate and cause significant
disease in people with AIDS. The most important example on a global scale is that of tuberculosis, as reactivated
tuberculosis can cause symptomatic disease with lower levels of reactivation.

Other important pathogens include cytomegalovirus, (which causes retinitis, pneumonitis, and colitis) and Pneumocystis
jiroveci (formerly known as Pneumocystis carinii; the causative organism in Pneumocystis pneumonia). In
immunocompetent hosts, these organisms are generally nonpathogenic, and asymptomatic infection is common (and in
the case of cytomegalovirus infection, life-long).

Antiviral medications are associated with adverse effects and thus contribute to patient morbidity and mortality rates,
especially because of the growing population of long-term survivors who are receiving combination antiviral therapy. In
particular, protease inhibitors may cause lipid-profile abnormalities.

In a study of 6,036 HIV-infected patients who had achieved suppression of HIV with antiretroviral therapy, researchers
found that the incidence of non-Hodgkin lymphoma (NHL) remained high (171 per 100,000 person-years [PY]), far
exceeding the rate of approximately 10 to 20 per 100,000 person-years reported in HIV-uninfected populations. The high
incidence of NHL was observed even in patients with nadir CD4 cell count > 200 cells/µl (140 per 100,000 PY). After
adjustment for older age, white race, male sex, HCV coinfection, and time-varying CD4 cell count, the risk of NHL risk was
higher when HIV viremia was above the limit of detection (50 copies/mL) in a dose-dependent manner.[92, 93]

https://emedicine.medscape.com/article/211316-print Page 16 of 71
05/09/2020, 11)59 PM

Patient Education
Patients with HIV infection should be counseled about the risks of infecting their sexual partners with HIV. Safer sex
practices and treatment of concurrent sexually transmitted diseases, both in the patient and in sexual partners,
considerably reduces the risk of transmission. Patients with HIV infection should be encouraged to inform their sexual
partners of their status; failure to do so has resulted in successful prosecutions in several countries. Sexual contacts
should be tested.

Some HIV-infected people actively seek out other persons with HIV infection for sex under the assumption that they are
not putting themselves or anyone else at an increased risk. However, it is clear that co-infections with multiple HIV strains
(whether the same or different clades) can and do occur, and that such events may result in a rapid deterioration of a
previously stable infection. A growing number of new infections are drug resistant upon first presentation, suggesting that
these infections were transmitted from individuals receiving therapy.

Higher viral loads in the source partner are associated with higher transmission rates; thus, because barrier contraception
is imperfect (although by far the best method to prevent sexual transmission), good control of viral load is important.

Intravenous drug users should be counseled on the risks of sharing intravenous drug paraphernalia.

For patient education information, see the Infections Center and Sexual Health Center, as well as HIV/AIDS and Rapid
Oral HIV Test.

Presentation

History
The history should be carefully taken to elicit possible exposures to human immunodeficiency virus (HIV). Risk factors
include the following:

Unprotected sexual intercourse, especially receptive anal intercourse (8-fold higher risk of transmission)

A large number of sexual partners

Prior or current sexually transmitted diseases (STDs): Gonorrhea and chlamydia infections increase the HIV
transmission risk 3-fold, syphilis raises the transmission risk 7-fold, and herpes genitalis raises the transmission risk
up to 25-fold during an outbreak

Sharing of intravenous drug paraphernalia

Receipt of blood products (before 1985 in the United States)

Mucosal contact with infected blood or needle-stick injuries

Maternal HIV infection (for newborns, infants, and children): Steps taken to reduce the risk of transmission at birth
include cesarean delivery and prenatal antiretroviral therapy in the mother and antiretroviral therapy in the newborn
immediately after birth.

The patient may present with signs and symptoms of any of the stages of HIV infection. Acute seroconversion manifests
as a flulike illness, consisting of fever, malaise, and a generalized rash. The asymptomatic phase is generally benign.
Generalized lymphadenopathy is common and may be a presenting symptom.

AIDS manifests as recurrent, severe, and occasionally life-threatening infections and/or opportunistic malignancies. The
signs and symptoms are those of the presenting illness, meaning that HIV infection should be suspected as an underlying

https://emedicine.medscape.com/article/211316-print Page 17 of 71
05/09/2020, 11)59 PM

illness when unusual infections present in apparently healthy individuals.

HIV infection itself does cause some sequelae, including AIDS-associated dementia/encephalopathy and HIV wasting
syndrome (chronic diarrhea and weight loss with no identifiable cause).

Physical Examination
No physical findings are specific to HIV infection. The physical findings are those of the presenting infection or illness.
Generalized lymphadenopathy is common. Weight loss may be apparent.

Evidence for risk factors or minor concurrent opportunistic infections (eg, herpetic lesions on the groin, widespread oral
candidiasis) may be clues to HIV infection.

DDx

Diagnostic Considerations
Human immunodeficiency virus (HIV) infection should be considered in any patient with unusual or recurrent serious
infections without another cause, especially in those with risk factors for HIV infection.

Any of the opportunistic infections or cancers associated with acquired immune deficiency syndrome (AIDS) can also
occur in the absence of HIV infection, although they usually develop in patients with some other form of immune
suppression or defect. The possibility of HIV infection must be considered on a case-by-case basis. Other causes of
immune suppression (eg, chemotherapy, immune disorders, severe combined immune deficiency [SCID], severe
malnutrition) should be considered. For example, a young adult with leukemia undergoing chemotherapy is at high risk for
many opportunistic infections.

Differential Diagnoses
Burkitt Lymphoma

Candidiasis

Coccidioidomycosis and Valley Fever

Cryptococcosis

Cryptosporidiosis

Cytomegalovirus (CMV)

Herpes Simplex

High-Grade Malignant Immunoblastic Lymphoma

Mycobacterium Avium Complex (MAC) (Mycobacterium Avium-Intracellulare [MAI])

Toxoplasmosis

https://emedicine.medscape.com/article/211316-print Page 18 of 71
05/09/2020, 11)59 PM

Workup

Workup

Approach Considerations
Screening for human immunodeficiency virus (HIV) infection is paramount, since infected individuals may remain
asymptomatic for years while the infection progresses. Serologic tests are the most important studies in the evaluation for
HIV infection.

Secondary testing that may be performed to assist with diagnosis or staging includes the following:

Viral culture

Lymph node biopsy

Proviral DNA polymerase chain reaction (PCR)

Genotyping of viral DNA/RNA

In June 2014, the Centers for Disease Control and Prevention (CDC) issued new recommendations for HIV testing in
laboratories that are aimed at reducing the time needed to diagnose HIV infection by as much as 3-4 weeks over previous
testing approaches. The new testing algorithm is performed as follows[94, 95] :

Diagnosis starts with a fourth-generation test that detects HIV in the blood earlier than antibody tests can; it
identifies the viral protein HIV-1 p24 antigen, which appears in the blood before antibodies do

If this test is positive, an immunoassay that differentiates HIV-1 from HIV-2 antibodies should be performed; results
from such assays can be obtained faster than they can from the Western blot test

In patients with positive results on the initial antigen test but with negative or indeterminate results on the antibody
differentiation assay, HIV-1 nucleic acid testing should be performed to determine whether infection is present

In August 2013, the FDA approved the Alere Determine HIV-1/2 Ag/Ab Combo test (Orgenics, Ltd), the first rapid HIV test
for the simultaneous detection of HIV-1 p24 antigen as well as antibodies to both HIV-1 and HIV-2 in human serum,
plasma, and venous or fingerstick whole blood specimens. Detection of HIV-1 antigen permits earlier detection of HIV-1
infection than is possible by testing for HIV-1 antibodies alone.[8, 9]

This rapid test can be used in outreach settings to identify HIV-infected individuals who might not be able to be tested in
traditional health care settings. The test does not distinguish between antibodies to HIV-1 and HIV-2, and is not intended
to be used for screening of blood donors.[8, 9]

Staging of HIV disease is based partially on clinical presentation, but other laboratory tests can help in deciding whether to
initiate or modify treatment.

Baseline laboratory studies for other infections (eg, tuberculosis) are important in the initial workup of a patient with newly
diagnosed HIV infection. In addition, baseline levels of factors that may be affected by antiretroviral therapy (eg, lipids)
should be measured.

Screening for HIV Infection


https://emedicine.medscape.com/article/211316-print Page 19 of 71
05/09/2020, 11)59 PM

The U.S. Preventive Services Task Force (USPSTF) strongly recommends that clinicians screen for HIV in all adolescents
and adults at increased risk for HIV infection, and all pregnant women.[1]

The American College of Obstetricians and Gynecologists recommends that all females aged 13-64 years be tested for
HIV at least once during their lifetime.[96, 97] Retesting annually or more often is recommended for those at high risk
because of injection drug use, sex with an injection drug user, sex for money or drugs, sex since their most recent HIV test
with men who have sex with men, or sex since their most recent HIV test with more than 1 person.

Guidelines issued in 2015 on HIV testing during pregnancy by the American College of Obstetricians and Gynecologists
are as follows:[98, 99]

Women should be tested for HIV during routine prenatal testing, on an opt-out basis when possible.
Women at high risk for HIV infection, including injection drug users and women with multiple sex partners during
their pregnancy, should be retested in their third trimester.
Women who have not been tested should be offered rapid screening when in labor; if the rapid test result is
positive, antiretroviral therapy should be initiated while awaiting results of a confirmatory test.
All pregnant women should be screened for HIV infection as early as possible during each pregnancy using the
opt-out approach when allowed.
Repeat HIV testing in the third trimester is recommended among women in areas with high HIV incidence or
prevalence and among women known to be at risk for acquiring HIV infection.
Women who were not tested earlier in pregnancy or whose HIV status is otherwise undocumented should be
offered rapid screening upon labor and delivery using the opt-out approach when allowed.
If a rapid HIV test result in labor is reactive, antiretroviral prophylaxis should be immediately initiated while awaiting
supplemental test results.
If the diagnosis of HIV infection is established, the woman should be linked into ongoing care with a specialist in
HIV care for comanagement.

The Centers for Disease Control and Prevention (CDC) recommends HIV screening for patients in all health-care settings,
after the patient is notified that testing will be performed unless the patient declines (opt-out screening); the CDC
recommends that persons at high risk for HIV infection be screened for HIV at least annually.[2]

Citing the benefits of early diagnosis and treatment and the failure of risk-based screening to identify a substantial
proportion of HIV-infected patients early in the disease, the American College of Physicians recommends that clinicians
adopt routine screening for HIV and encourage all patients to be tested.[3]

Screening assays

A high-sensitivity enzyme-linked immunoabsorbent assay (ELISA) should be used for screening. Most ELISAs can be
used to detect HIV-1 types M, N, and O and HIV-2.

A positive ELISA result should be followed with confirmatory testing in the form of one or more Western blot assays or
similar specific assay. Specific diagnostic criteria vary by test. Results are typically reported as positive, negative, or
indeterminate.

Testing for HIV-2 should be ensured for patients from an HIV-2 endemic area or those who have indeterminate results on
HIV-1 Western blot testing. Not all HIV tests include detection of HIV-2 or Group O. In New York City, 62 cases of HIV-2
were detected over an 8-year period, of which 40 were initially misdiagnosed as HIV-1.[100]

Early detection using combination screens may be more effective than simply using serology. The additional detection of
p24 antigen or viral RNA may detect a greater number of very recent infections before seroconversion occurs. This would
likely result in significant reductions in transmission as well as overall health costs and healthcare burden.[101]

To address the problem of confirmatory supplemental tests giving false-negative results early in the course of HIV
infection, the CDC conducted two prospective evaluations of a new HIV diagnostic algorithm. The new diagnostic
algorithm replaces the Western blot (WB) with an HIV-1/HIV-2 antibody differentiation assay as the supplemental test and
includes an RNA test to resolve reactive immunoassay (IA) with negative supplemental test results.[102]

https://emedicine.medscape.com/article/211316-print Page 20 of 71
05/09/2020, 11)59 PM

CD4+ T-cell Count


The CD4 T-cell count is a reliable indicator of the current risk of acquiring opportunistic infections. CD4 counts vary, and
serial counts are generally a better measure of any significant changes. The reference range for CD4 counts is 500-2000
cells/µL. After seroconversion, CD4 counts tend to decrease (around 700/µL on average) and continue to decline over
time. For surveillance purposes, a CD4 count under 200/µL is considered AIDS-defining in the United States owing to the
increased risk of opportunistic infections at this level. The magnitude of discordance between absolute CD4 T-cell
numbers and CD4 T-cell percentages is greatest in those with active hepatitis C virus and more advanced liver disease.
[103]

In children under five years of age, the CD4 T-cell percentage is considered more important than the absolute count. (Less
than 25% is considered worthy of starting therapy, regardless of the total CD4 count). In adults with chronic hepatitis C
and low absolute CD4 T-cells, the CD4 percentage may also be more useful, due to probable T-cell sequestration in the
liver.[6]

Viral Load
Viral load in peripheral blood is used as a surrogate marker of viral replication rate. This is a surrogate because most of
the viral replication occurs in the lymph nodes rather than in the peripheral blood.

The test is a quantitative amplification of the viral RNA using nucleic acid sequence-based amplification (NASBA), reverse-
transcription polymerase chain reaction (RT-PCR), or similar technologies. Quantitative viral-load assays should not be
used as a diagnostic tool because several false-positive misdiagnoses have been reported in the literature.

The rate of progression to AIDS and death is related to the viral load, although, on an individual level, it is poorly predictive
of the absolute rate of CD4 T-cell loss. Patients with viral loads greater than 30,000/mL are 18.5 times more likely to die of
AIDS than those with undetectable viral loads.

With therapy, viral loads can often be suppressed to an undetectable level (ie, < 20-75 copies/mL, depending on the assay
used); this is considered optimal viral suppression. At the same time, the CD4 count rises and the risk of opportunistic
infections and death is reduced. Complete inhibition of viral replication appears impossible and may be unnecessary.

Not uncommonly, successfully treated patients will demonstrate intermittent viremia, with viral loads transiently detectable
at low levels (typically, < 400 copies/mL); this appears to occur more commonly with some viral load assays than others.
Such “blips” are not thought to represent viral replication or to predict virologic failure.[7] Virologic failure is defined as a
confirmed viral load of more than 200 copies/mL; although this is a research definition, it may be useful in clinical practice.
[7]

Secondary HIV Testing


Viral culture is expensive and time-consuming and is less sensitive in patients with low viral loads. Viral culture may be
performed as part of phenotypic drug-resistance testing.

Lymph node architecture is disrupted during HIV infection. HIV DNA, RNA, and proteins may be detected with molecular

https://emedicine.medscape.com/article/211316-print Page 21 of 71
05/09/2020, 11)59 PM

techniques, and electron microscopy may reveal virions.

Proviral DNA PCR is usually performed only in newborns because conventional serologic testing is useless in these
patients (maternal antibodies may persist for 9 months or longer). Two or more negative results separated by at least one
month is considered a negative result.

Genotyping of viral DNA/RNA can guide therapy. Because patterns of mutations that lead to resistance to specific drugs or
drug classes are now well-recognized, sequencing of the viral genome allows for the selection of specific antivirals that are
more likely to elicit a response.

Baseline Studies
Baseline studies for other infections that are important in the initial workup of a patient with newly diagnosed HIV infection
include the following:

Purified protein derivative (PPD) skin testing for tuberculosis

Cytomegalovirus (CMV) testing

Syphilis testing

Rapid amplification testing for gonococcal and chlamydial infection

Hepatitis A, B, and C serology

Anti-Toxoplasma antibody

Ophthalmologic examination

A purified protein derivative skin test is placed to evaluate for tuberculosis infection. Chest radiography should be
performed in patients with a positive PPD test result.

Serology should be performed to test for CMV infection. The presence of anti-CMV IgG indicates previous exposure to
CMV. Ophthalmologic examination is used to evaluate for CMV retinitis in people with very low CD4 T-cell counts.

For syphilis screening, rapid plasma reagent (RPR) testing can be used initially, but more specific testing should be used
for follow-up, as RPR can yield false-positive results. Lumbar puncture is used to evaluate neurologic symptoms.

Rapid amplification testing is used to evaluate for gonococcal infection and chlamydia in cases of sexual HIV transmission.
Pelvic examination is performed in females (with wet mount for trichomoniasis).

Hepatitis A, B, and C serology is performed to determine the need for vaccination or treatment and to evaluate for chronic
infection. Patients infected with hepatitis C may be candidates for treatment. Genotyping and baseline liver function tests
are crucial.

Anti-Toxoplasma antibody is measured to determine whether patients have had toxoplasmosis, and thus are at risk for
reactivation of infection in the event of immunocompromise. Patients with prior Toxoplasma infection require prophylaxis if
their CD4+ T-cell counts drop below 100/µL.

Tests to establish baseline values of factors that may be affected by antiretroviral therapy include the following:

Liver function tests

Serum chemistries

https://emedicine.medscape.com/article/211316-print Page 22 of 71
05/09/2020, 11)59 PM

Blood urea nitrogen (BUN)/serum creatinine

Fasting lipid panel

Vitamin B12 and folate levels

Thyroid function studies[104]

Other tests include urinalysis to evaluate for HIV-associated nephropathy and a drug screen to effectively exclude other
metabolic and infectious etiologies.[105]

Histologic Findings
Certain histologic findings are characteristic of various features of HIV infection and AIDS. The lymph node architecture is
progressively disrupted; this can be reversed with effective antiviral therapy. Findings include hyperplasia, multinucleated
syncytia of T cells, and loss of the normal follicular dendritic network. Nucleic acid or immunohistochemical stains for viral
antigens shows virus localizing to macrophages, T cells, and dendritic cells. Electron microscopy may reveal virions or
intracellular virus within phagosomes in macrophages.

Multinucleated giant cells are a characteristic finding in patients with HIV encephalopathy. Myelin pallor and microgliosis
may also be observed.

Staging
The CDC classifies HIV infection into 3 categories, according to the presence of certain infections or diseases.[10] These
conditions may be exacerbated by the HIV infection or represent true opportunistic infections.

Category A is asymptomatic HIV infection without a history of symptoms or AIDS-defining conditions.

Category B is HIV infection with symptoms that are directly attributable to HIV infection (or a defect in T-cell–mediated
immunity) or that are complicated by HIV infection. These include, but are not limited to, the following:

Bacillary angiomatosis

Oropharyngeal candidiasis (thrush)

Vulvovaginal candidiasis, persistent or resistant

Pelvic inflammatory disease (PID)

Cervical dysplasia (moderate or severe)/cervical carcinoma in situ

Oral hairy leukoplakia

Idiopathic thrombocytopenic purpura

Constitutional symptoms, such as fever (>38.5°C) or diarrhea lasting more than 1 month

Peripheral neuropathy

https://emedicine.medscape.com/article/211316-print Page 23 of 71
05/09/2020, 11)59 PM

Herpes zoster (shingles), involving 2 or more episodes or 1 or more dermatomes

Category C is HIV infection with AIDS-defining opportunistic infections, as outlined in Pathophysiology.

These 3 categories are further subdivided based on the CD4+ T-cell count. Categories A1, B1, and C1 are characterized
by CD4+ T-cell counts greater than 500/µL. Categories A2, B2, and C2 are characterized by CD4+ T-cell counts between
200/µL and 400/µL. HIV infections in patient with CD4+ T-cell counts under 200/µL are designated as A3, B3, or C3.

Importantly, once an HIV infection has been staged into a higher clinical category, it remains in that category permanently.
In addition, the infection is classified based on the lowest CD4+ T-cell count in that patient.

For example, if a given HIV-positive patient recovers from a bout of Pneumocystis pneumonia (PCP) and the CD4+ T-cell
count improves from 50/µL to 250/µL, that patient’s HIV infection remains classified as C3. Persons with A3, B3, and C1-3
HIV infection are considered to have AIDS. This is important to recognize, as this designation is not based solely on the
previous occurrence of opportunistic infections but rather on the current risk of infection based on a reduced CD4+ T-cell
count.

Treatment

Approach Considerations
The treatment of human immunodeficiency virus (HIV) disease depends on the stage of the disease and any concomitant
opportunistic infections.[7] In general, the goal of treatment is to prevent the immune system from deteriorating to the point
that opportunistic infections become more likely. Immune reconstitution syndrome is also less likely in patients whose
immune systems are weakened to this point.

Highly active antiretroviral therapy (HAART) is the principal method for preventing immune deterioration. In addition,
prophylaxis for specific opportunistic infections is indicated in particular cases.

Successful long-term HAART results in a gradual recovery of CD4 T-cell numbers and an improvement of immune
responses and T-cell repertoire (previously lost antigen responses may be restored). The peripheral T-cell counts initially
surge after therapy is initiated, but this represents redistribution of activated T cells from the viral replication centers in the
lymph nodes rather than a true increase in total-body CD4 T-cell counts.[106, 107]

In addition to virologic response and reduced risk of opportunistic infection, there is evidence to suggest that non-AIDS-
defining illnesses, in particular psychiatric and renal disease, may also be reduced when on HAART. Although
multifactorial in nature (transmission mode and patient educational level are independent risk factors for these events)
there may also be a direct role of HIV in these events, or an indirect role mediated through the subsequent immune
dysfunction. Some non-AIDS-defining illnesses, such as liver and cardiovascular disease, are not improved by HAART.
[108]

Treatment guidelines for HIV infection are age-specific. Guidelines for pediatric populations are compiled by the Working
Group on Antiretroviral Therapy and Medical Management of HIV-Infected Children; guidelines for adults and adolescents
are compiled by the Panel on Clinical Practices for Treatment of HIV Infection. Complete treatment guidelines may be
viewed at the National Institutes of Health Web site.

For discussion of antiretroviral drugs and regimens, see Antiretroviral Therapy for HIV Infection.

Primary care interventions

The Infectious Diseases Society of America (IDSA) issued updated guidelines in November 2013 for the management of
HIV infection.[109, 110] Because of advances in management, HIV-infected patients are now having fewer complications

https://emedicine.medscape.com/article/211316-print Page 24 of 71
05/09/2020, 11)59 PM

and surviving longer; as a result, they are increasingly experiencing common health problems seen in the general
population, and these problems must be addressed. Accordingly, the updated IDSA guidelines emphasize the role of
primary care interventions, as follows:

HIV-infected individuals should undergo screening for diabetes, osteoporosis, and colon cancer as appropriate and
should be vaccinated against pneumococcal infection, influenza, varicella, and hepatitis A and B

Lipid monitoring and management of lipids and other cardiovascular risk factors should be performed

Patients with well-controlled infection should undergo blood monitoring for viral levels every 6-12 months

Women with HIV should undergo annual trichomoniasis screening, and all infected patients who may be at risk
should undergo annual screening for gonorrhea and chlamydia

HAART Studies and DHHS Guidelines


The introduction of HAART has significantly improved mortality rates. One study of nearly 7000 men with HIV infection
found that annual mortality rates decreased from 7% in 1996 to 1.3% in 2004, although the findings highlighted the fact
that non–AIDS-related illnesses were accounting for a greater proportion of deaths.[91]

These findings were repeated in another, more recent study of over 83,000 people with AIDS in the United States from
1990-2006,[111] which showed that cancers as a cause of mortality decreased overall but increased as a percentage of
deaths, with non-Hodgkin lymphoma being the most common AIDS-related cancer and lung cancer being the most
common non–AIDS-related cancer.

A National Cancer Institute study attributed increased non-AIDS-defining cancer mortality to the 4-fold expansion in the
HIV-infected population in the United States, which was largely driven by greater numbers of people aged 40 years and
older.[112]

Treatment failures are most closely related to the timing of therapy initiation (and, therefore, of timeliness of diagnosis).
CD4 counts under 200/µL and evidence for AIDS (in the form of cytomegalovirus retinitis) are strong predictors of mortality
(risk ratios of 2.7 and 1.6, respectively).[113]

Studies on the initiation of antiretroviral therapy

An analysis of a series of 18 prospective cohort studies in the United Kingdom found that deferring combination
antiretroviral therapy until patients reached a CD4 cell count of 251-350 cells/µL was associated with higher rates of AIDS
and death than starting therapy at 351-450 cells/µL. The adverse effect of deferring treatment increased with decreasing
CD4 cell count threshold. The researchers suggested than the minimum threshold for initiating treatment should be 350
CD4+ T cells/µL rather than 200/µL.[114]

This finding was echoed in a Haitian study (a resource-limited setting), where early initiation of antiretroviral therapy
significantly decreased the rates of death and incident tuberculosis. Initiating antiretroviral therapy treatment during early
phases of disease (CD4+ T-cell count between 200/µL and 350/µL) was found to increase survival in Haitians compared
with waiting until CD4+ T cells fell below 200/µL.[115]

The HIV-CAUSAL Collaboration analyzed data from the United States Veterans Health Administration and HIV clinics in
Europe to compare the results of therapy initiation at CD4 cell counts from 0.200-0.500 × 109 cells/L. The study concluded
that initiating HAART therapy at the 0.350 × 109 cells/L threshold decreased AIDS-free survival compared with initiation at
0.500 × 109 cells/L, but did not substantially increase mortality. A significant rise in mortality was seen at initiation
thresholds below 0.300 × 109 cells/L. This result differs from other studies. Because CD4 cell count at initiation is not
randomized in such observational cohort studies, confounding factors may exist.[116]

The first randomized controlled trial to investigate the question of when to initiate therapy was the NIH Comprehensive

https://emedicine.medscape.com/article/211316-print Page 25 of 71
05/09/2020, 11)59 PM

International Program of Research on AIDS (CIPRA) HT 001 clinical study. This work showed that starting antiretroviral
therapy at CD4+ T-cell counts between 200 and 350 cells/µL improves survival compared with deferring treatment until the
CD4+ T-cell count drops to less than 200 cells/µL (the standard of care at the time).

Interim analysis of CIPRA HT 001 showed that of 816 HIV-infected adults with early HIV disease, 6 of those who began
antiretroviral therapy within 2 weeks of enrollment (early treatment) died, while 23 participants in the standard-of-care
group died.[117] Among participants who began the study without tuberculosis infection, 18 individuals in the early
treatment group developed tuberculosis, while 36 people in the standard-of-care group developed tuberculosis.

These interim results were statistically significant and led to ending the trial early to offer antiretroviral therapy to all
participants in the standard-of-care group with a CD4+ T-cell count of less than 350 cells/µL.

One study has suggested that extremely early initiation of ART during this acute seroconversion period (within 2 weeks of
converting) may result in better long-term CD4 counts and steady-state viral load. Although the numbers were small,
acutely treated individuals had a mean of 0.48 log10 copies/mL lower viral load and higher CD4 counts (average, 112
cells/µL) than an untreated cohort. The effects were less pronounced and lasted for a shorter time for patients with an
“early” initiation of therapy (within 2 weeks to 6 months of seroconversion).[118]

Attempts have been made to characterize the timeframe of seroconversion, especially in patients without a clear source of
exposure that can be dated accurately. Although imperfect, algorithms based on the number of Western Blot bands and
the actual ELISA signal compared to the positive cutoff may have some utility in this case.[119]

Rapid ART Start

Randomized trials in Lesotho, Haiti, and South Africa showed significant improvements in viral load suppression at 10-12
months of treatment and retention in care with rapid initiation of therapy.[120]

In one study, individuals were randomized to early ART with simplified counseling and point-of-care CD4 cell assays or to
standard care. In the intervention group, 80% began ART within 14 days, and 71% started ART the same day of eligibility,
compared with 38% and 18%, respectively, in the control group. Virologic suppression at one year was improved in the
intervention group (85% vs 75%). San Francisco implemented a citywide rapid ART program in which newly diagnosed
persons were linked to care within 5 days of diagnosis and offered treatment on the day of the clinic visit. Of 265 newly
diagnosed persons, 97% were linked to care (30% within 5 days) and 81% started ART; time from diagnosis to HIV RNA
level below 200 copies/mL decreased by more than 50%, and time from first care visit to ART initiation decreased from 27
days to 1 day. A large HIV clinic in Atlanta implemented rapid access to ART on the day of the initial visit. Median time
from initial diagnosis to HIV-1 RNA level below 200 copies/mL decreased from 67 to 41 days; however, the program was
not sustainable because of increased patient load and inadequate funding for staffing.[120]

Starting ART on the day of diagnosis requires coordination between testing and treatment setting and access to resources
that may limit uptake.

ART initiation, including rapid start, is recommended in all infected ambulatory patients committed to starting ART (unless
the patient has symptoms that suggest an opportunistic infection for which immediate ART is contraindicated) and in those
with unclear HIV diagnosis (eg, discordant serologic or rapid test results). Treatment with nonucleoside reverse
transcriptase inhibitors (NNRTIs) is not recommended for rapid start owing to concerns about transmitted drug resistance
(eg, K103N mutation), and abacavir should not be initiated until results of testing for the HLA-B*5701 allele are available.
[120]

Therapy initiation recommendations in the United States

Two sets of guidelines are commonly used in the United States, the Department of Health and Human Services (DHHS)
and the International AIDS Society - USA Panel (IAS-USA), to determine when to treat and which treatment approach to
use. The IAS-USA guidelines are generally updated every other year, with the most recent update occurring in August
2018. DHHS updated their treatment guidelines in December 2019,[11] reflecting changes in HIV prevention and care.
While these two guidelines differ in some areas, for general practitioners, they represent similar strategies for HIV
prevention through the use of PrEP and TasP, HIV treatment with INSTI-based regimens in most patients, and guidance
for the use of alternative regimens in select patients.

https://emedicine.medscape.com/article/211316-print Page 26 of 71
05/09/2020, 11)59 PM

While previous guideline versions have recommended waiting to initiate HIV treatment, the DHHS and IAS-USA guidelines
both advocate initiation of HIV treatment as soon as possible, which some have termed "rapid" start of HIV medications.
Some centers are able to do this on the same day as diagnosis. If patients are ready to begin treatment, it should be
offered. Rapid-start protocols have been shown to improve patient retention in care, reduce the interval from diagnosis to
virologic suppression, and, in small studies, demonstrate mortality benefit. As a result, Ending the Epidemic (ETE): A Plan
for America recommends rapid treatment initiation as soon as possible in patients with newly diagnosed HIV infection.
Excellent outcome data have been gathered and presented from San Francisco, New York City, and other cities,
demonstrating the benefits of not waiting to start treatment.

Antiretroviral agents

Classes of antiretroviral agents include the following:

Nucleoside reverse transcriptase inhibitors (NRTIs)


Protease inhibitors (PIs)
Nonnucleoside reverse transcriptase inhibitors (NNRTIs)
Entry inhibitors (EI)
Integrase strand transfer inhibitors (INSTI)

Current drug regimen recommendations

The December 2019 DHHS guideline[11] lists the below regimens as "recommended for most" for treatment-naive
patients.

INSTI-based regimens are as follows:

Bictegravir/tenofovir alafenamide/emtricitabine (BIC/TAF/FTC) (single-tablet regimen)


Dolutegravir/abacavir/lamivudine (DTG/ABC/3TC) (single-tablet regimen) - Only for patients who are HLA-B*5701–
negative
Dolutegravir plus tenofovir disoproxil fumarate/emtricitabine OR tenofovir alafenamide/emtricitabine (two-tablet
regimen)
Dolutegravir/lamivudine (DTG/3TC) (single-tablet regimen) - Avoid in patients with an HIV viral load of more than
500,000 copies/mL, patients with HBV co-infection, and patients in whom HIV NRTI resistance testing results are
unavailable prior to initiation.
Raltegravir plus tenofovir disoproxil fumarate/emtricitabine OR tenofovir alafenamide/emtricitabine

Women who become pregnant while taking antiretroviral agents should contact their physician and register with the
Antiretroviral Pregnancy Registry.

Regimen selection

Antiretrovirals should be prescribed by an infectious disease specialist. Antiretroviral regimen selection is individualized,
on the basis of the following[7] :

Virologic efficacy

Toxicity

Pill burden

Dosing frequency

Drug-drug interaction potential

Drug resistance testing results

Comorbid conditions

https://emedicine.medscape.com/article/211316-print Page 27 of 71
05/09/2020, 11)59 PM

Drug resistance testing typically involves genotyping or phenotyping of resistance in the patient's viral strains. The January
2011 DHHS guidelines recommend genotypic testing to guide the choice of initial therapy in antiretroviral-naïve patients,
as well as in patients in whom first or second regimens produce a suboptimal virologic response or virologic failure.
Phenotypic testing is generally added to genotypic testing when complex drug resistance mutation patterns, especially to
protease inhibitors, are confirmed or suspected.[7]

Recent research on antiretroviral agents

A review of 2725 HIV isolates for protease inhibitor susceptibility helped delineate the specific contributions of various
resistance mutations to each currently available protease inhibitor. The study revealed that certain mutations could result
in increased susceptibility to a particular drug, and that some effects on resistance had been underestimated. The study
concluded that cross-resistance between the various protease inhibitors now and in the future may be missed without
systematic analysis of the effects of specific mutations.[121]

A study by Lennox et al in treatment-naive patients from 67 centers on 5 continents demonstrated benefits of raltegravir
(another INSTI) over efavirenz (an NNRTI) as part of combination antiretroviral therapy.[122] Participants had viral RNA
(vRNA) concentrations greater than 5000 copies/mL and demonstrated no baseline drug resistance to efavirenz, tenofovir,
or emtricitabine. They were randomly assigned to receive raltegravir 400 mg bid (n=281) or efavirenz 600 mg daily
(n=282).

The primary endpoint was a vRNA concentration of less than 50 copies/mL at week 48. In the raltegravir group, 86.1%
achieved the primary endpoint, compared with 81.9% in the efavirenz group (difference 4.2%, 95% CI, -1.9 to 10.3). The
time to viral suppression was shorter in the raltegravir group than in the efavirenz group. Significantly fewer adverse drug
reactions were reported in the raltegravir group (44.1%) than in the efavirenz group (77%).[122]

Similarly, in a randomized, phase III, noninferiority trial of raltegravir-based treatment versus efavirenz-based therapy, in
563 treatment-naïve HIV-1–infected patients, the addition of raltegravir to tenofovir/emtricitabine, compared with the
addition of efavirenz to tenofovir/emtricitabine, resulted in significantly greater vRNA suppression rates and increases in
baseline CD4 counts at week 240. In addition, significantly fewer patients in the raltegravir group experienced
neuropsychiatric and drug-related adverse events.[123]

A new INSTI, dolutegravir (Tivicay), was approved by the FDA in August 2013 for treatment of HIV-1 infection in
combination with other antiretroviral agents in adults and children aged 12 years or older who weigh at least 40 kg.
Approval was based on several studies showing evidence of virologic suppression in both treatment-naive and treatment-
experienced patients on a daily regimen of the drug.[124, 125, 126, 127] Approval of dolutegravir for the indication in
children aged 12 years or older was based on data in integrase-naïve patients.

Knowledge of resistance patterns in resource-limited areas is vital in the selection of first-line antiretroviral treatment. In a
subset of the Development of Antiretroviral Treatment in Africa (DART) trial, the virological response to zidovudine-
lamivudine plus abacavir (an NRTI) at 32 weeks was inferior to the response to zidovudine-lamivudine plus nevirapine (an
NNRTI). HIV RNA levels were lower in the nevirapine group than in the abacavir group.

The authors concluded that first-line zidovudine-lamivudine plus abacavir therapy will eventually lead to extensive
nucleoside analogue resistance and that continued research is needed to optimize first- and second-line therapies in
resource-limited settings.[128]

Approval of the ART combination product elvitegravir/cobicistat/emtricitabine/tenofovir (Stribild) was based on analyses of
48-week data from 2 randomized, double-blind, active-controlled trials in treatment-naïve, HIV-1 infected individuals
(n=1408). Results showed a single tablet regimen of Stribild met its primary objective of noninferiority compared to Atripla
(efavirenz 600 mg/emtricitabine 200 mg/tenofovir 300 mg) and to a regimen containing ritonavir-boosted atazanavir plus
Truvada (emtricitabine/tenofovir).[129, 130]

In a study of 484 HIV-infected pregnant women, 3 short-term antiretroviral strategies, initiated simultaneously with the
administration of single-dose nevirapine (sdNVP), resulted in a low rate (1.2%) of new NVP-resistance mutations. In the
study, HIV-infected pregnant women were randomized to receive sdNVP and either zidovudine/lamivudine (3TC),
tenofovir/emtricitabine (FTC), or lopinavir/ritonavir for either 7 or 21 days. According to the results, 21-day antiretroviral
regimens are better at preventing the emergence of minor NVP resistance variants compared to 7-day regimens. Of the
412 women who had primary endpoint results available, 4 of 215 in the 7-day arms had new NVP resistance (1.9%),

https://emedicine.medscape.com/article/211316-print Page 28 of 71
05/09/2020, 11)59 PM

whereas only 1 of 197 (0.5%) in the 21-day arms exhibited the same resistance.[131]

Prophylaxis for Opportunistic Infections


Prophylaxis for Pneumocystisjiroveci (a normally harmless commensal organism) is most important, as this causes a
common, preventable, serious infection. In patients with CD4 counts of less than 200/µL, prophylaxis with trimethoprim-
sulfamethoxazole (TMP-SMX; Bactrim) has been shown to prevent Pneumocystis pneumonia (PCP).

In patients whose CD4+ T-cell counts rise above 200/µL with effective therapy, PCP prophylaxis may be discontinued.
When TMP-SMX cannot be used, alternatives include dapsone (after screening for glucose-6-phosphate dehydrogenase
[G6PD] deficiency) and atovaquone or monthly nebulized pentamidine treatments.

TMP-SMX also prevents toxoplasmosis and should be administered when the CD4+ T-cell count drops to below 100/µL if
the patient is not already receiving it to prevent PCP.

CD4+ counts below 50/µL place the patient at risk for Mycobacterium avium complex infection, and weekly azithromycin or
clarithromycin is recommended as prophylaxis. In individuals with ART-induced viral suppression, the incidence and
overall mortality of M avium complex disease are low enough that primary M avium complex prophylaxis is no longer
recommended.[120]

Prophylaxis for fungal or viral infections is not routinely necessary, but some have recommended fluconazole in patients
with CD4+ T-cell counts under 50/µL to prevent candidal or cryptococcal infections and to protect against endemic fungal
infections in geographic locales of hyperendemicity for histoplasmosis or coccidioidomycosis. However, the emergence of
resistant Candida strains is a realistic concern.[120]

Oral ganciclovir is indicated for prophylaxis of cytomegalovirus infection in patients with advanced AIDS and is about 50%
effective in reducing invasive disease.[132] As with fluconazole, there are concerns about resistance, and prophylaxis
should be reserved for those with CD4+ T-cell counts under 50/µL and evidence of previous cytomegalovirus infection.

Treatment of Opportunistic Infections


Treatment of opportunistic infections is paramount and should be directed at the specific pathogen. Although effective
antiretroviral therapy reduces the risk of acquiring an opportunistic infection and reverses the effects of many opportunistic
infections (eg, Kaposi sarcoma, cytomegalovirus retinitis), aggressive treatment of life-threatening or otherwise serious
infections may necessitate a temporary stay of antiretroviral therapy to avoid drug interactions or cumulative toxicity.

With specific regard to TB, the relationship with antiretroviral therapy is complex. A large multi-national study found that
the relative risk of acquiring TB after starting HAART was approximately half that of those in whom HAART was not
started. However, there was evidence for immune reconstitution inflammatory syndrome (IRIS) in some patients co-
infected with HIV and TB in the first few months of therapy. In addition, those older than 50, or with pretreatment CD4 T-
cell counts less than 50/µL, were less likely to see the same reduction in TB incidence.[133]

Recent data support the recommendation to start ART within the first 2 weeks of initiating treatment for tuberculosis in
patients with CD4 cell counts below 50/uL and within the first 2-8 weeks in those with CD4 cell counts of 50/uL or more. In
patients with cryptococcal meningitis in high-resource settings with access to optimal antifungal therapy, frequent
monitoring, and aggressive management of intracranial pressures, ART should begin within 2 weeks of diagnosis. Careful
monitoring for immune reconstitution inflammatory syndrome is essential. In individuals diagnosed with HIV infection and
malignancy concurrently, ART should be initiated immediately. Early adverse effects of ART can be monitored and
managed while cancer staging and molecular testing are performed.[120]

https://emedicine.medscape.com/article/211316-print Page 29 of 71
05/09/2020, 11)59 PM

Treatment of HIV-Associated Lipodystrophy


HIV lipodystrophy is a syndrome of abnormal central fat accumulation and/or localized loss of fat tissue that occurs in
patients taking antiretroviral drugs. Tesamorelin (Egrifta), a growth hormone–releasing factor, was approved by the US
Food and Drug Administration in 2010 to reduce excess visceral abdominal fat in HIV-infected patients with lipodystrophy.

FDA approval of tesamorelin was based on 2 studies in which visceral adipose tissue was significantly decreased from
baseline at 26 weeks and sustained at 52 weeks.[134, 135, 136] These multicenter, randomized, double-blind, placebo-
controlled phase 3 studies consisting of a 26-week main phase and a 26-week extension phase in 816 HIV-infected
patients with excess abdominal fat associated with lipodystrophy. In phase III, randomized, double-blind studies that
assessed the effect of tesamorelin on HIV-associated abdominal fat accumulation, a reduction in adiposity correlated with
overall improved metabolic profiles of lipids and glucose.[137]

Suppressive Therapy for Herpes Simplex Virus 2 Infection


Most individuals infected with HIV-1 are also infected with herpes simplex virus type 2 (HSV-2). Suppressive therapy of
HSV-2 with acyclovir reduces plasma HIV-1 concentrations. Lingappa et al found that acyclovir reduced risk for HIV-1
disease progression by 16% compared with placebo.[138] Disease progression was defined as first occurrence of CD4+
T-cell counts dropping below 200/µL.

In this study, patients (n=3381) who were dually infected with HSV-2 and HIV-1, had CD4+ cell counts of at least 250/µL,
and were not taking antiretroviral therapy were randomized to receive either acyclovir 400 mg PO orally twice daily or
placebo. In patients with CD4+ counts of 350/µL or more, acyclovir delayed risk of CD4+ counts falling below 350/µL by
19%. The use of acyclovir to suppress HSV-2 before initiating antiretroviral therapy merits further study to determine its
effects on HIV-1 disease progression.

A second study of 440 people in Uganda showed that, in those with HIV and HSV-2 as well as HIV viral loads above
50,000/mL, there was a significant delay in the progression to AIDS-defining illnesses or CD4 T-cell decline in the
acyclovir group compared to placebo. Interestingly, no significant benefit was found for those with viral loads below
50,000/mL.[139]

Treatment of HIV-Associated Diarrhea


In December 2012, the FDA approved crofelemer for the relief of diarrhea in patients with HIV/AIDS who are undergoing
antiretroviral therapy.[13] However, before patients are treated with this drug, they should be properly tested to confirm
that the diarrhea is not caused by an infection or a gastrointestinal (GI) disease.

Deterrence and Prevention of HIV Infection

https://emedicine.medscape.com/article/211316-print Page 30 of 71
05/09/2020, 11)59 PM

On an individual level, the most effective methods for prevention of HIV infection include (1) avoidance of sexual contact
outside a monogamous relationship, (2) the use of safer sex practices for all other sexual encounters, and (3) abstinence
from nonmedical parenteral drug use.

In addition, measures can also be taken to prevent or deter HIV transmission risk from infected persons to noninfected
individuals through behavioral, biomedical, and structural interventions aimed at reducing their infectiousness and their
risk of exposing others to HIV. Such measures are detailed in the CDC's Recommendations for HIV Prevention in Adults
and Adolescents with HIV Infection in the United States.[11]

In March 2019, the CDC reported that approximately 80% of new US HIV infections are transmitted by 40% of persons
with HIV infection. In 2016, persons unaware of their HIV infection (15% of the infected population) transmitted 38% of
new HIV infections. Persons aware of their HIV infection but not receiving care (23% of the infected population)
transmitted 43% of new HIV infections. Persons with HIV infection receiving care but not virally suppressed (11% of the
infected population) transmitted 20% of new HIV infections. HIV-infected persons undergoing treatment with viral
suppression (51% of the infected population) transmitted 0% of new infections.[140]

These findings suggest that increased HIV status awareness, HIV treatment initiation and sustained treatment compliance,
effective viral suppression with ART, and prevention methods such as condom use and pre-exposure prophylaxis (PrEP)
can greatly reduce the rate of new HIV infections.

Sexual transmission

Prevention measures include the following:

Abstinence when possible

Reduction in number of sexual partners

Using barrier contraception

Treatment of concurrent sexually transmitted diseases (STDs)

Testing of self and partner for HIV infection and other STDs

Concomitant infection with other STDs (eg, gonorrhea, herpes, syphilis) is the most well-known risk factor that
predisposes to transmission of HIV. These STDs may cause mucosal ulcerations or tears or a higher concentration of
inflammatory cells in the mucosa, which are targets for HIV infection. Comprehensive testing for these should be obtained
when a sexual transmission is suspected or the source of infection is unknown, both in the patient and in sexual partners.

Certain sexual acts are more likely to lead to HIV infection than others. For example, fellatio carries the lowest risk of
transmission (with very few case reports in the literature), while receptive anal intercourse carries the highest risk (a
likelihood of approximately 1.5% per act with an infected individual).

An apparent effect of hormonal contraception on HIV transmission to and from women has been reported, with a slight but
statistically significant increase in transmission involving women on hormonal contraception. In a study of 3790
serodiscordant couples from Africa, the hazard ratios for transmission were 1.98 to, and 1.97 from women on hormonal
contraception. Although barrier contraception should be employed in instances of serodiscordance anyway, this finding
further strengthens that recommendation in those couples where the female partner is using hormonal contraception.[141]

Vertical transmission

Prevention measures include the following:

Maternal testing

Effective control of maternal infection

Prenatal antiviral therapy and treatment of mother and infant during labor, delivery, and the neonatal period

https://emedicine.medscape.com/article/211316-print Page 31 of 71
05/09/2020, 11)59 PM

Cesarean delivery

Avoidance of breastfeeding (unless local conditions make this unsafe or unfeasible)

A retrospective cohort study reviewed the records of 3,273 HIV-positive women receiving prenatal care in Malawi and
Mozambique from July 2005 to December 2009. Patients were treated with triple antiviral therapy during pregnancy until 6
months postpartum for prevention of vertical transmission. Regardless of CD4 count, ART provided a protective effect
against mortality, fetal demise, and premature birth.[142]

The prevention of mother-to-child transmission of HIV-2 is less certain than for HIV-1, from which most of the
recommendations have been derived. Transmission of HIV-2 is less frequent (perhaps 10-fold less efficient), but HIV-2 is
intrinsically resistant to the non-nucleoside RTI nevirapine, removing one option for pharmacologic prophylaxis at the time
of delivery.

In a large French cohort study, the mother-to-child transmission rate of HIV-2 infection was 0.6%.[143] Transmission was
related to poor control of HIV-2 infection in the mother or due to breastfeeding.

In the absence of definitive clinical trial data, the only definite conclusion is that effective control of maternal infection is
paramount, and other nonspecific measures (identification of infected mothers, caesarean section, avoidance of
breastfeeding) are probably effective at preventing transmission.

Empiric prophylaxis with zidovudine, as in HIV-1 infection, is probably warranted and effective but does not appear to be
evidence-based.

Blood-borne transmission

Prevention measures include the following:

Blood-product and donor screening

Avoidance of reusing needles for intravenous drug abuse (needle-exchange programs are widespread in the
developed world, but the evidence that they have had a significant effect is debatable)

Postexposure prophylaxis

The CDC has recommended basic and expanded HIV postexposure prophylaxis (PEP) regimens. For full details, see the
Updated U.S. Public Health Service guidelines for the management of occupational exposures to HIV and
recommendations for postexposure prophylaxis.

Also, see the Medscape Reference articles Antiretroviral Therapy for HIV Infection and Body Fluid Exposures.

An overview of the CDC recommendations for preferred HIV PEP regimen is as follows:

Basic PEP regimen: Raltegravir or dolutegravir plus tenofovir DF/emtricitabine FTC (Truvada)
Alternative PEP regimens: Darunavir/ritonavir plus tenofovir DF/emtricitabine FTC
Generally not recommended for PEP: Didanosine, nelfinavir, tipranavir

Nevirapine for PEP is contraindicated because of a risk of early-onset rash and severe hepatotoxicity.

Vaccination efforts

The initial hope of an effective vaccine against HIV has not been fulfilled. Aside from the virus being able to rapidly mutate
antigenic portions of key surface proteins, HIV infection progresses despite the host’s humoral and cellular immune
responses; therefore, any vaccination effect needs to surpass the normal host response to HIV.

A study from Thailand suggests a possible benefit of vaccines in heterosexuals at risk for HIV-1 transmission.[144] In the
randomized, multicenter, double-blind, placebo-controlled trial by Rerks-Ngarm et al, 16,402 healthy participants aged 18-
30 years received either 4 priming injections of recombinant canarypox vector vaccine (ALVAC-HIV [vCP1521]) plus 2

https://emedicine.medscape.com/article/211316-print Page 32 of 71
05/09/2020, 11)59 PM

booster shots of recombinant glycoprotein 120 subunit vaccine (AIDSVAX B/E) or placebo.

In the per-protocol analysis, which excluded subjects who seroconverted during the vaccination series, the vaccine
efficacy was 26.2%. In the modified-intention-to-treat analysis, which excluded subjects who had baseline HIV-1 infection,
the vaccine efficacy was 31.2%. However, the 95% confidence intervals in these analyses were extremely wide (-13.3 to
51.9 and 1.1 to 52.1, respectively), which precludes concluding that the vaccine had proven efficacy.[144]

Among study subjects who developed HIV-1 infection, viremia and CD4+ T cell counts were unchanged by vaccination.
This suggests that, if infection did occur, there was no apparent immunologic benefit from having received the vaccine.

With respect to risk behavior, a post-hoc analysis of efficacy found that the combination of the HIV vaccines, ALVAC-HIV
(vCP1521) and AIDSVAX B/E, was more effective in those who maintained lower-risk sexual behavior compared to those
that reported high or increasing-risk behavior.[145]

Preexposure prophylaxis

An innovative and controversial strategy for preventing HIV transmission is regular use of antiretroviral medications by
uninfected individuals. An updated clinical practice guideline released in May 2014 by the CDC, extends recommendations
for preexposure prophylaxis (PrEP) of HIV in high-risk patients.[146, 147] According to these guidelines, PrEP should be
considered for the following non-HIV-infected individuals:

Anyone who is in an ongoing sexual relationship with an HIV-infected partner

A gay or bisexual man who has had sex without a condom or has been diagnosed with a sexually transmitted
infection within the past 6 months and is not in a mutually monogamous relationship with a partner who recently
tested HIV-negative

A heterosexual man or woman who does not always use condoms when having sex with partners known to be at
risk for HIV and is not in a mutually monogamous relationship with a partner who recently tested HIV-negative

Anyone who, in the preceding 6 months, has injected illicit drugs and shared equipment or been in a treatment
program for injection drug use

Daily oral PrEP with the fixed-dose combination of tenofovir disoproxil fumarate (TDF) 300 mg and emtricitabine (FTC)
200 mg (Truvada) has been shown to be safe and effective in reducing the risk of sexual HIV acquisition in adults,
adolescents, and discordant couples. In addition, daily administration of emtricitabine 200 mg plus tenofovir alafenamide
(AF) 25 mg (Descovy) is approved in at-risk adults and adolescents for HIV-1 pre-exposure prophylaxis (PrEP) to reduce
the risk of HIV-1 infection from sex, excluding those who have receptive vaginal sex.[148] Daily emtricitabine/tenofovir is
one prevention option that is part of the general guidelines for HIV prevention. For more information, see Preexposure HIV
Prophylaxis.

PrEP treatment guidelines include the following recommendations:

Acute and chronic HIV infection must be excluded by symptom history and HIV testing immediately before PrEP is
prescribed; evidence level IA

Two medication regimens are approved by the FDA and recommended for PrEP with all the populations specified in
the CDC guideline is daily TDF 300 mg coformulated with FTC 200 mg (Truvada) or the fixed-dose combination of
FTC 200 mg plus TAF 25 mg (Descovy); evidence level IA

TDF alone has shown substantial efficacy and safety in trials with IDUs and heterosexually active adults and can be
considered as an alternative regimen for these populations, but not for MSM, among whom its efficacy has not
been studied; evidence level IC

Use of other ART medications for PrEP, either in place of or in addition to TDF/FTC (or TDF) or TAF/FTC is not
recommended; evidence level IIIA

Oral PrEP for coitally-timed or other noncontinuous daily use is not recommended; evidence level IIIA

https://emedicine.medscape.com/article/211316-print Page 33 of 71
05/09/2020, 11)59 PM

Interim CDC guidelines were issued in 2011 based on the multinational study called the Pre-exposure Prophylaxis
Initiative (iPrEx) trial that found that once-daily emtricitabine plus tenofovir disoproxil fumarate (FTC-TDF) provided an
additional 44% protection against HIV infection in a study population of 2499 high-risk, HIV-negative men or transgender
women who have sex with men.[149]

Over a median 1.2 years of follow up, 36 patients in the FTC-TDF group and 64 in the placebo group became infected with
HIV. All study subjects also received comprehensive prevention services that included monthly HIV testing, condom
provision, counseling, and management of other STDs.[149]

Additional studies have been completed or are ongoing in serodiscordant heterosexual couples and intravenous drug
users.[150, 151]

There remain policy considerations surrounding costs, opportunity costs, and ethical issues that must be addressed before
broad implementation in the United States.[152] Potential drawbacks include the possibility that pre-exposure prophylaxis
may encourage some recipients to practice less-safe sex; it does not address transmission of other STDs; and it could
encourage the development of drug resistance.

Compliance is essential. In studies, the level of protection varied widely depending on how consistently participants used
pre-exposure prophylaxis. Among those whose data (based on self-reports, bottles dispensed, and pill counts) indicate
use on 90% or more days, HIV risk was reduced by 73%. Among those whose adherence by the same measure was less
than 90%, HIV risk was reduced by only 21%.[153, 154]

Topical antivirals could potentially help with preventing transmission, but studies to date have failed to produce positive
results. For example, a double-blinded, randomized, controlled trial of a vaginal microbicide gel with in vitro activity against
HIV failed to show protective effects. The study involved 9385 women from South Africa, Tanzania, Uganda, and Zambia
who used a synthetic naphthalene sulphonate polymer. Infection rates per 100 person-years were similar between groups
(4.7 for 2% gel, 4.6 for 0.5% gel, and 3.9 for placebo).

IAS-USA guidelines
In July 2014, the International Antiviral Society-USA (IAS-USA) released new recommendations for HIV prevention in
adolescents and adults in clinical care settings[155, 156] in conjunction with updated recommendations on antiretroviral
treatment (ART) of adult HIV infection.[156, 157]

The IAS-USA panel suggested that combined biomedical/behavioral approaches to HIV prevention in clinical settings have
the potential to not only prevent the disease but also cause nearly all HIV-infected individuals to become noninfectious.
[155, 156] Among its key HIV-prevention recommendations for teens and adults are the following[155, 156] :

Perform HIV testing at least once for all adults and adolescents; repeat testing often for those at increased risk.

Be vigilant for potential HIV infection, and promptly perform diagnostic testing in those with suspected acute HIV
infection.

For HIV-confirmed patients, provide multimodal interventions to include prompt initiation of ART, support for
treatment adherence, individualized risk assessment and counseling, assistance with partner notification, and
periodic screening for common sexually transmitted infections.

For high-risk, HIV-uninfected patients, administer preexposure prophylaxis with daily emtricitabine/tenofovir
disoproxil fumarate and provide multimodal interventions (eg, individualized counseling on risk reduction).

Provide intravenous/injection drug users with multimodal harm reduction services such as access to needle/syringe
exchange programs, supervised injection, medically assisted therapies, and detoxification programs.

Administer prompt postexposure prophylaxis in individuals with mucosal/parenteral HIV exposure from a known
infected source.

https://emedicine.medscape.com/article/211316-print Page 34 of 71
05/09/2020, 11)59 PM

Consultations
Consultation with an infectious disease or HIV specialist should be strongly considered for all new cases of HIV infection.
Studies have clearly shown that the successful management of patients with HIV is related to the expertise and HIV
caseload of the treating physician. In particular, pediatric cases of HIV infection are handled differently; cutoffs for CD4
counts at which prophylaxis would be recommended and antiviral drug availability (on- or off-study for experimental drugs
or regimens) differ on the basis of age.

Input from an infectious disease consultant may be helpful in the management of other unrelated illnesses in patients
infected with HIV.

Long-Term Monitoring
Guidelines from the DHHS Panel on Antiretroviral Guidelines for Adults and Adolescents recommend performing the
following tests every 3 months in patients on antiretroviral therapy[7] :

Basic chemistry profile

Liver function studies

Complete blood count with differential

The basic chemistry studies should include serum sodium, potassium, bicarbonate, chloride, blood urea nitrogen (BUN),
and creatinine, and glucose (preferably fasting), plus an estimate of creatinine clearance. Fasting glucose is repeated
every 3-6 months if abnormal at the last measurement, or every 6 months if normal at the last measurement.

A fasting lipid profile is measured every 6 months if abnormal at the last measurement, or every 12 months if normal at the
last measurement.

In a clinically stable patient on an regimen whose viral load is suppressed and whose CD4+ T-cell count is well above the
threshold for opportunistic infection risk, 2011 DHHS guidelines recommend that the CD4+ T-cell count may be monitored
every 6-12 months (instead of every 3-6 months), unless there are changes in the patient’s clinical status, such as new
HIV-associated clinical symptoms or initiation of treatment with interferon, corticosteroids, or anti-neoplastic agents.[7]

Guidelines

CDC Guidelines on HIV Screening in Men Who Have Sex with Men
The following are guidelines on HIV screening among men who have sex with men (MSM)[158]

The CDC concludes that the evidence, programmatic experience, and expert opinions are insufficient to warrant
changing the current recommendation of annual screening for men who have sex with men (MSM) to more
frequent screening (every 3 or 6 months).
Providers in clinical settings should offer HIV screening at least annually to all sexually active MSM.
Clinicians can also consider the potential benefits of more frequent HIV screening (eg, every 3 or 6 months) for
some asymptomatic sexually active MSM based on their individual risk factors, local HIV epidemiology, and local
policies.
Among MSM who are prescribed preexposure prophylaxis, HIV testing every 3 months and immediate testing

https://emedicine.medscape.com/article/211316-print Page 35 of 71
05/09/2020, 11)59 PM

whenever signs and symptoms of acute HIV infection are reported is indicated.
MSM who experience a specific high-risk sexual exposure or have symptoms of recent HIV infection should seek
immediate HIV testing, and clinicians should be alert for the symptoms of acute HIV infection and provide
appropriate diagnostic testing.
Tenofovir alafenamide (TAF)/emtricitabine (FTC) is now included as an alternative for pre-exposure prophylaxis
(PrEP) in men who have sex with men (MSM).

European AIDS Clinical Society Guidelines


The European AIDS Clinical Society (EACS) has issued updated guidelines on the management of HIV infection. In this
latest version of the guidelines (version 10), a new drug-drug interaction panel has been introduced. The guidelines have
six main sections, including a general overview table of all major issues in patients living with HIV infection;
recommendations on antiretroviral treatment (ART); drug-drug interactions; and diagnosis, monitoring, and treatment of
comorbidities, coinfections, and opportunistic diseases.[159]

Antiretroviral therapy

A new recommendation in the updated guideline favors an unboosted integrase strand transfer inhibitor (INSTI) with high
genetic barrier (bictegravir [BIC] or dolutegravir [DTG]) as a third agent for therapy initiation in treatment-naïve individuals
with HIV infection.

Two nucleos(t)ide reverse transcriptase inhibitors (NRTIs) plus doravirine (DOR) has been included among the
recommended regimens.

Tenofovir disoproxil fumarate (TDF)/lamivudine (3TC) has been added as a backbone, when indicated.

DTG plus 3TC dual therapy has been upgraded as a recommended regimen.

High-genetic-barrier INSTI or protease inhibitors pharmacologically boosted with cobicistat or ritonavir (PI/b) are
recommended as initial therapy for primary HIV infection if resistance testing is unavailable.

Ritonavir- or cobicistat-boosted darunavir (DRV/b) plus rilpivirine (RPV) has been included as a dual-therapy option.

Monotherapy with PI/b is not recommended.

Initiation of ART in patients with tuberculosis (TB) and HIV coinfection who have a CD4 cell count of more than 50/µL may
be deferred up to 8 weeks after TB treatment has been started.

Tenofovir alafenamide (TAF)/emtricitabine (FTC), raltegravir (RAL) once daily, and bictegravir (BIC) have been added as
potential drugs for postexposure prophylaxis (PEP).

TAF/FTC is now included as an alternative for pre-exposure prophylaxis (PrEP) in men who have sex with men (MSM)
and transgender women.

Comorbidities

Screening for kidney disease should incorporate albumin/creatinine ratio for glomerular disease and protein/creatinine
ratio for screening for and diagnosing antiretroviral-related tubulopathy.

Lipid targets have been updated in terms of the threshold for ART modification, from 20% 10-year risk for cardiovascular
disease (CVD) to 10% 10-year risk for CVD.

Medical management of hypertension has been updated to include amended drug-sequencing suggestions and
recommended drugs.

https://emedicine.medscape.com/article/211316-print Page 36 of 71
05/09/2020, 11)59 PM

The workup of liver disease among individuals with HIV infection should now include a fourth step to include risk
stratification based on risk prediction tools and transient elastography and an updated algorithm for surveillance of varices.

Department of Health and Human Services


The U.S. Department of Health and Human Services Panel on Antiretroviral Guidelines for Adults and Adolescents has
published guidelines to help providers integrate treatment as prevention (TasP) into their clinical practice. The Panel
emphasizes the importance of screening and early diagnosis of HIV. In order for persons with HIV to benefit from early
diagnosis, the Panel recommends that ART be started immediately or as soon as possible after diagnosis to increase the
uptake of ART, decrease the time required to achieve linkage to care and virologic suppression for individual patients,
reduce the risk of HIV transmission, and improve the rate of virologic suppression among persons with HIV.[160]

The DHHS recommendations include the following:

All persons with HIV should be informed that maintaining a plasma HIV RNA (viral load) of < 200 copies/mL,
including any measurable value below this threshold value, with antiretroviral therapy (ART) prevents sexual
transmission of HIV to their partners. Patients may recognize this concept as Undetectable = Untransmittable or
U=U.
Persons with HIV who are starting ART should use another form of prevention with sexual partners (eg., condoms,
preexposure prophylaxis [PrEP] for the HIV-negative sexual partner, sexual abstinence) for at least the first 6
months of treatment and until a viral load of < 200 copies/mL has been documented. Many experts would
recommend confirming sustained suppression before assuming that there is no further risk of sexual HIV
transmission.
When the viral load is ≥200 copies/mL, additional methods are needed to prevent transmission of HIV to sexual
partners until resuppression to < 200 copies/mL has been confirmed.
Persons with HIV who intend to rely upon ART for prevention need to maintain high levels of ART adherence. They
should be informed that transmission is possible during periods of poor adherence or treatment interruption.
At each visit for HIV care, clinicians should assess adherence to ART and counsel patients regarding the
importance of ART to their own health as well as its role in preventing sexual HIV transmission.
Providers should inform patients that maintaining a viral load of < 200 copies/mL does not prevent acquisition or
transmission of other sexually transmitted infections (STIs).
Providers should also routinely screen all sexually active persons with HIV for STIs, both for their own health and to
prevent transmission of STIs to others.

The Panel has added DTG (dolutegravir) /3TC (lamivudine) to the list of Recommended Initial Regimens for Most People
with HIV, except for individuals with pretreatment HIV RNA >500,000 copies/mL; who are known to have active hepatitis B
virus (HBV) coinfection; or who will initiate ART before results of HIV genotype testing for reverse transcriptase or HBV
testing are available.

Bictegravir/TAF (tenofovir alafenamide) /FTC (emtricitabine) has been added as a treatment option for persons with acute
or recent HIV infection in cases where ART will be initiated before genotypic drug resistance testing results are available.

Medication

Medication Summary
Effective antiretroviral therapy is the most important intervention in terms of improving longevity and preventing
opportunistic infections in patients with human immunodeficiency virus (HIV) infection. Therapy should involve
combinations of drugs recommended by current guidelines. Recommendations are specific for various patient groups (eg,

https://emedicine.medscape.com/article/211316-print Page 37 of 71
05/09/2020, 11)59 PM

treatment naïve, treatment experienced, coinfection with hepatitis C).[7] Antiretroviral drug classes and agents within each
class are listed in Table 1, below (see individual medication tables for more detail).

Many of the antiretroviral drugs that have been approved for HIV-infected adults and adolescents are gaining FDA
approval for use in younger children. For more information, see Pediatric HIV Infection.

Of the antiretroviral drugs that have been approved, several are no longer being manufactured either because of the
development of improved formulations (ie, amprenavir replaced by fosamprenavir) or because of limited use (ie,
delavirdine and zalcitabine [ddC]).

Several combination products that contain tenofovir alafenamide (TAF) are now approved in the United States. TAF is a
more targeted form of tenofovir that has demonstrated high antiviral efficacy at a dose that is 10 times lower than that of
tenofovir DF, as well as an improved renal and bone safety profile.

Table 1. Antiretroviral Drug Classes and Agents (Open Table in a new window)

Abacavir (Ziagen, ABC)

Didanosine (Videx, Videx EC, ddI)

Emtricitabine (Emtriva, FTC)

Lamivudine (Epivir, 3TC)

Nucleoside reverse transcriptase inhibitors (NRTIs) Stavudine (Zerit, Zerit XR, d4T)

Tenofovir DF (Viread, TDF)

Tenofovir AF (TAF)

Zalcitabine (Hivid, ddC)*

Zidovudine (Retrovir, ZDV, AZT)

Amprenavir (Agenerase, AVP)*

Atazanavir (Reyataz , ATV)

Darunavir (Prezista, DRV)

Fosamprenavir (Lexiva, f-APV)

Indinavir (Crixivan, IDV)


Protease inhibitors (PIs)
Lopinavir and ritonavir (Kaletra, LPV/r)

Nelfinavir (Viracept, NFV)

Ritonavir (Norvir, RTV)

Saquinavir (Invirase [hard gel] capsule, SQV)

Tipranavir (Aptivus, TPV)

https://emedicine.medscape.com/article/211316-print Page 38 of 71
05/09/2020, 11)59 PM

Delavirdine (Rescriptor, DLV)

Efavirenz (Sustiva, EFV)

Etravirine (Intelence, ETR)


Non-nucleoside reverse transcriptase inhibitors (NNRTIs)
Nevirapine (Viramune, NVP)

Rilpivirine (Edurant)

Doravirine (Pifeltro, DOR)

Fusion inhibitors Enfuvirtide (Fuzeon, T-20)

Cellular chemokine receptor (CCR5) antagonists Maraviroc (Selzentry, MVC)

Raltegravir (Isentress, RAL)

Integrase inhibitors Dolutegravir (Tivicay, DTG)

Elvitegravir (Vitekta, EVG)

Entry inhibitors (CD4 post-attachment inhibitors) Ibalizumab (Trogarzo, IBA)

gp120 Attachment inhibitors Fostemsavir (Rukobia, FTR)

*No longer available on market

Combination therapy has been shown to dramatically reduce the likelihood of drug resistance (many drug-resistant
mutations are mutually exclusive) and to suppress viral replication to the point that progression to AIDS is significantly
slowed. Antiviral-resistance mutations often affect more than one drug simultaneously because of similar development
pipelines and the ultimate molecular structure of the drug, and combination choices should account for this possibility.

Ritonavir, a PI that may be used in its own right, boosts blood levels of other PIs. This permits a reduced dosage of the
coadministered drug. Various products have been formulated to include PIs combined with ritonavir.

Combination products

Numerous antiretroviral combination products are available on the market to assist patients with compliance and decrease
the daily number of tablets and capsules required (see Table 2, below).

Table 2. Antiretroviral Combination Products (Open Table in a new window)

Drug Content

https://emedicine.medscape.com/article/211316-print Page 39 of 71
05/09/2020, 11)59 PM

per Brand Adult Dose (≥40 kg)


Tablet/Capsule* Name

Darunavir 800
mg

Cobicistat 150
mg
Symtuza# 1 tab PO qd with food
Emtricitabine
200 mg

Tenofovir AF 10
mg

Bictegravir 50
mg

Emtricitabine
200 mg Biktarvy# 1 tab PO qd

Tenofovir AF 25
mg

Efavirenz 400
mg

Lamivudine
300 mg Symfi Lo# 1 tab PO qHS on an empty stomach

Tenofovir DF
300 mg

Efavirenz 600
mg

Lamivudine
300 mg Symfi# 1 tab PO qHS on an empty stomach

Tenofovir DF
300 mg

Dolutegravir 50 1 tab PO qd with a meal


mg
Note: This is a complete once-daily regimen in adults who are virologically
Juluca# suppressed (HIV-1 RNA < 50 copies/mL) on a stable ART regimen for ≥6 months
Rilpivirine 25

https://emedicine.medscape.com/article/211316-print Page 40 of 71
05/09/2020, 11)59 PM

mg with no history of treatment failure and no known substitutions associated with


resistance

Dolutegravir 50
1 tab PO qd with or without food
mg
Dovato# Note: This a complete once-daily regimen in treatment-naïve adults with no known
Lamivudine
substitutions associated with resistance to dolutegravir or lamivudine
300 mg

Elvitegravir 150
mg

Cobicistat 150
mg
Genvoya# 1 tab PO qd
Emtricitabine
200 mg

Tenofovir AF 10
mg

Elvitegravir 150
mg

Cobicistat 150
mg
Stribild# 1 tab PO qd
Emtricitabine
200 mg

Tenofovir DF
300 mg

Abacavir 600
mg
Epzicom 1 tab PO qd
Lamivudine
300 mg

Abacavir 600
mg

Dolutegravir 50
Triumeq 1 tab PO qd
mg

Lamivudine
300 mg

https://emedicine.medscape.com/article/211316-print Page 41 of 71
05/09/2020, 11)59 PM

Abacavir 300
mg

Lamivudine
Trizivir 1 tab PO bid
150 mg

Zidovudine 300
mg

Efavirenz 600
mg
1 tab PO qd on empty stomach
Emtricitabine
Atripla#
200 mg Note: May be used alone as a complete regimen or in combination with other
ARTs
Tenofovir DF
300 mg

Emtricitabine
200 mg

Rilpivirine 25
Complera# 1 tab PO qd with a meal
mg

Tenofovir DF
300 mg

Emtricitabine
200 mg

Rilpivirine 25
Odefsey# 1 tab PO qd with a meal
mg

Tenofovir AF 25
mg

Emtricitabine 1 tab PO qd
200 mg
Truvada CrCl 30-49 mL/min: 1 tab PO q48h
Tenofovir DF
300 mg CrCl < 30 mL/min: Do not administer

Emtricitabine 1 tab PO qd

https://emedicine.medscape.com/article/211316-print Page 42 of 71
05/09/2020, 11)59 PM

200 mg Descovy CrCl < 30 mL/min: Do not administer

Tenofovir AF
300 mg

Lamivudine
150 mg
Combivir 1 tab PO bid
Zidovudine 300
mg

Lamivudine
300 mg
Cimduo 1 tab PO qd
Tenofovir DF
300 mg

Doravirine 100
mg

Lamivudine
300 mg Delstrigo# 1 tab PO qd with or without food

Tenofovir DF
300 mg

*Not indicated for patients requiring dosage adjustments (eg, weight < 40 kg, renal impairment, hepatic impairment,
dose-limiting adverse effects) unless otherwise stated.

#Complete once-daily regimen

A subgroup analysis of black patients with HIV enrolled in the 48-week, open-label SPIRIT (Switching PI to Rilpivirine In-
combination with Truvada) trial showed that switching from an antiretroviral regimen consisting of a boosted PI and
ritonavir (RTV) plus 2 nucleoside/nucleotide reverse-transcriptase inhibitors (PI+RTV+2NRTIs) to a simplified once-daily,
single-tablet regimen of rilpivirine/emtricitabine/tenofovir DF (RPV/FTC/TDF) is safe and effective in this population.[161,
162]

Patients were randomized to undergo an immediate switch to RPV/FTC/TDF at baseline or to maintain their
PI+RTV+2NRTIs regimen for 24 weeks and then switch to RPV/FTC/TDF for 24 weeks (delayed switch).

At 24 weeks, a subgroup analysis of black patients in the study showed that viral suppression rates (HIV-1 RNA < 50
copies/mL) were 95% in the RPV/FTC/TDF group and 91% in the group receiving PI+RTV+2NRTIs; ie, no significant
difference existed. At 48 weeks, 89% of black patients in the immediate-switch group maintained viral suppression,
compared with 95% of those in the delayed-switch group, which again was not considered a significant difference.[161,
162]

At 48 weeks, when all patients in the study were taken into account, there was no significant difference in viral

https://emedicine.medscape.com/article/211316-print Page 43 of 71
05/09/2020, 11)59 PM

suppression between the immediate-switch (89%) and delayed-switch (92%) groups; the rates of adverse events were
similar in both groups as well.[161, 162] However, investigators noted significant improvement in lipid levels in patients
who received the single-tablet RPV/FTC/TDF regimen.[162]

The FDA has approved a once-daily, fixed-dose triple-combination pill (Triumeq) containing the antivirals dolutegravir,
abacavir, and lamivudine for the treatment of patients aged 18 years or older with HIV infection. The FDA based its
approval primarily on the results of 2 studies, in one of which, a 96-week study in treatment-naive adults, virologic
suppression occurred in 80% of patients receiving dolutegravir and abacavir/lamivudine (administered separately),
compared with 72% of patients taking a single-pill regimen of efavirenz, emtricitabine, and tenofovir (Atripla).[163]

The FDA approved the NNRTI doravirine in August 2018. Also with this new drug, a complete regimen combination
containing doravirine/lamivudine (3TC)/tenofovir DF (TDF) was also approved. Approval was based on the DRIVE-
FORWARD clinical trial (n=766). Patients who were antiretroviral-naïve were randomly assigned to once-daily treatment
with doravirine or darunavir 800 mg plus ritonavir 100 mg (DRV+r), each in combination with emtricitabine (FTC)/TDF or
abacavir (ABC)/3TC. Treatment with doravirine led to sustained viral suppression through 48 weeks, meeting its primary
endpoint of noninferiority compared with DRV+r, each in combination with FTC/TDF or ABC/3TC. At week 48, 84% of the
doravirine group and 80% of the DRV+r group had plasma HIV-1 RNA levels of less than 50 copies/mL.[164]

The doravirine/3TC/TDF combination was approved based on data from the DRIVE-AHEAD trial (n=728). Patients who
were antiretroviral-naïve were randomly assigned to once-daily treatment with doravirine/3TC/TDF or efavirenz
(EFV)/emtricitabine/tenofovir disoproxil fumarate (EFV 600 mg/FTC 200 mg/TDF 300 mg). The doravirine/3TC/TDF
combination provided sustained viral suppression through 48 weeks, meeting its primary endpoint of noninferiority
compared with EFV/FTC/TDF. At week 48, 84% of the doravirine/3TC/TDF group had plasma HIV-1 RNA levels of less
than 50 copies/mL, as did 81% of the EFV/FTC/TDF group.[165]

Antiretroviral agent, nucleoside reverse-transcriptase inhibitor

Class Summary
NRTIs agents inhibit viral replication by inhibiting viral RNA–dependent DNA polymerase.

Abacavir (Ziagen)
This NRTI interferes with HIV viral RNA–dependent DNA polymerase and inhibits viral replication. Guidelines from the
Department of Health and Human Services Panel on Clinical Practices for Treatment of HIV Infection recommends
screening for HLA-B*5701 before starting patients on a regimen that contains abacavir, to reduce the risk of
hypersensitivity reaction.

Didanosine (Videx, Videx EC)


Didanosine interferes with HIV viral RNA–dependent DNA polymerase and inhibits viral replication.

Emtricitabine (Emtriva)
This agent is a synthetic nucleoside cytosine analog classified as an NRTI. It competes with deoxycytidine-5'-triphosphate
and incorporates into viral DNA, causing chain termination.

Lamivudine (Epivir)

https://emedicine.medscape.com/article/211316-print Page 44 of 71
05/09/2020, 11)59 PM

Lamivudine is a thymidine analog that inhibits viral replication.

Stavudine (Zerit, Zerit XR)


Stavudine competes with deoxycytidine-5'-triphosphate and incorporates into viral DNA, causing chain termination.

Tenofovir disoproxil fumarate (Viread)


This agent inhibits the activity of HIV reverse transcriptase by competing with the natural substrate deoxyadenosine 5'-
triphosphate and, after incorporation into DNA, by DNA chain termination. It is administered as the prodrug bis-
isopropoxycarbonyloxymethyl ester derivative of tenofovir, which is converted, through various enzymatic processes, to
tenofovir, a nucleotide analog of adenosine 5'-monophosphate. Bioavailability is enhanced by a high-fat meal. Prolonged
intracellular distribution allows for once-daily dosing.

This drug has shown substantial efficacy and safety in PReP trials with IV drug users and heterosexually active adults.
CDC guidelines recommend tenofovir alone as an alternative regimen to emtricitabine/tenofovir for these populations, but
not for MSM, among whom its efficacy has not been studied.

Zidovudine (Retrovir)
Zidovudine is a thymidine analog that inhibits viral replication.

Antiretroviral Agent, Protease Inhibitor

Class Summary
PIs inhibit protein precursors necessary for HIV infection of uninfected cells.

Atazanavir (Reyataz)
An azapeptide HIV-1 PI, atazanavir prevents virion maturation by selectively inhibiting Gag and Gag-Pol polyproteins in
HIV-1–infected cells.

Darunavir (Prezista)
This HIV-1 PI selectively inhibits HIV-encoded Gag-Pol polyprotein cleavage in infected cells, thereby preventing mature
virus particle formation. It is indicated for HIV disease that has not responded to treatment with other antiretroviral agents.
Coadminister with low-dose ritonavir (ritonavir-boosted therapy decreases elimination and increases darunavir serum
concentration).

Darunavir is typically coadministered with other anti-HIV agents (eg, NRTIs). Food increases maximum plasma
concentration (Cmax) and area under the curve (AUC). This agent is indicated for HIV infection in antiretroviral treatment–
experienced adults (eg, those with HIV-1 strains resistant to more than one PI).

Fosamprenavir (Lexiva)

https://emedicine.medscape.com/article/211316-print Page 45 of 71
05/09/2020, 11)59 PM

Fosamprenavir is a prodrug that is converted to amprenavir by cellular phosphatases in vivo.

Lopinavir and ritonavir (Kaletra)


Lopinavir inhibits HIV protease and renders the enzyme incapable of processing polyprotein precursor, leading to
production of noninfectious immature HIV particles. Ritonavir inhibits CYP3A metabolism of lopinavir, increasing plasma
levels of lopinavir.

Nelfinavir (Viracept)
Nelfinavir inhibits HIV-1 protease, resulting in the production of an immature and noninfectious virus.

Indinavir (Crixivan)
Indinavir prevents formation of protein precursors necessary for HIV infection of uninfected cells and viral replication.

Ritonavir (Norvir)
This HIV PI is used as a part of double or triple therapy with nucleosides and other protease inhibitors.

Saquinavir (Invirase)
Saquinavir inhibits HIV protease and renders the enzyme incapable of processing polyprotein precursor, leading to
production of noninfectious immature HIV particles.

Tipranavir (Aptivus)
A nonpeptidic PI that inhibits HIV replication. Indicated for combination antiretroviral treatment in adults with HIV-1
infection who have evidence of viral replication and who are highly experienced with treatment or who have HIV-1 strains
that are resistant to multiple PIs.

Tipranavir must be coadministered with ritonavir (200 mg) to attain therapeutic levels (ie, tipranavir/ritonavir).
Administration alone, without ritonavir-boosted levels, is not effective. Genotypic or phenotypic testing and/or treatment
history should guide use.

Antiretroviral agent, non-nucleoside reverse-transcriptase inhibitor

Class Summary
NNRTIs inhibit viral replication.

Delavirdine (Rescriptor)
An NNRTI of HIV-1, delavirdine directly binds to reverse transcriptase and inhibits RNA- and DNA-dependent DNA
polymerase.

https://emedicine.medscape.com/article/211316-print Page 46 of 71
05/09/2020, 11)59 PM

Efavirenz (Sustiva)
Efavirenz is an NNRTI with activity against HIV-1. This agent binds to reverse transcriptase and blocks RNA-dependent
and DNA-dependent DNA polymerase activities, including HIV-1 replication. Efavirenz does not require intracellular
phosphorylation for antiviral activity.

Etravirine (Intelence)
Etravirine is an NNRTI of HIV-1 that binds directly to reverse transcriptase, causing catalytic site disruption. This action
blocks RNA- and DNA-dependent DNA polymerase activities. Etravirine does not inhibit human DNA polymerases alpha,
beta, or gamma.

This agent is indicated for use in combination with other antiretroviral agents for treatment-experienced HIV-infected adults
who have viral replication and HIV-1 strains resistant to NNRTIs and other antiretroviral agents. If virologic failure was
experienced with other NNRTIs, do not use etravirine in combination with NRTIs only.

Nevirapine (Viramune)
Nevirapine is an NNRTI that limits virus replication by a mechanism different from the nucleosidase inhibitors such as
zidovudine and lamivudine.

Rilpivirine (Edurant)
Rilpivirine is a NNRTI that inhibits HIV-1 replication by noncompetitive inhibition of HIV-1 reverse transcriptase. It does not
inhibit the human cellular DNA polymerases alpha, beta, and gamma.

Doravirine (Pifeltro)
Indicated in combination with other antiretroviral agents for the treatment of HIV-1 infection in adults (1) who have not
undergone antiretroviral therapy (ART) or (2) as a replacement for the current antiretroviral regimen in patients who are
who are virologically suppressed (HIV-1 RNA

Antiretroviral Agent, Integrase Inhibitor

Class Summary
This agent prevents insertion of a DNA copy of the viral genome into host cell DNA. It is indicated for HIV-1 infection
combination therapy in treatment-experienced adults with evidence of viral replication and drug-resistant HIV-1 strains.

Raltegravir (Isentress)
Raltegravir was the first available agent in the class of integrase strand transfer inhibitors.

Dolutegravir (Tivicay)
Dolutegravir is an integrase strand transfer inhibitor (INSTI) that inhibits catalytic activity of HIV-1 integrase, an HIV

https://emedicine.medscape.com/article/211316-print Page 47 of 71
05/09/2020, 11)59 PM

encoded enzyme required for viral replication. It is approved for use in children 12 years or older who weigh at least 40 kg.

Elvitegravir (Vitekta)
Integrase inhibitor that is used in combination with an HIV protease inhibitor (ie, atazanavir, lopinavir, darunavir,
fosamprenavir, or tipranavir) and coadministered with ritonavir plus other antiretroviral drug(s) as indicated for the
treatment of HIV-1 infection in antiretroviral treatment-experienced adults. It was originally approved as part of the
combination product elvitegravir/cobicistat/emtricitabine/tenofovir (Stribild).

Antiretroviral Agent, Fusion Inhibitor

Class Summary
These agents disrupt the ability of HIV to fuse with and infect healthy T cells.

Enfuvirtide (Fuzeon)
This is the first agent in the new anti-HIV class called fusion inhibitors. Indicated for use in combination with other
antiretroviral agents for HIV-1 infection in treatment-experienced patients who demonstrate evidence of HIV-1 replication
despite ongoing antiretroviral therapy.

Enfuvirtide binds to HIV gp41 surface protein, thereby, disrupting the virus's ability to fuse with and infect healthy T cells. In
clinical trials, subjects were twice as likely to achieve undetectable HIV-1 plasma levels (eg, < 40 copies/mL) when
enfuvirtide was added to antiretroviral optimized regimens than without enfuvirtide added to therapy.

Antiretroviral agent, CCR5 antagonist

Class Summary
These agents block viral entry into white blood cells via the CCR5 co-receptor.

Maraviroc (Selzentry)
Maraviroc blocks viral entry via the CCR5 co-receptor into WBCs, reduces viral load, and increases T-cell counts in
infection with CCR5-tropic HIV-1 (ie, R5 virus). Accelerated approval by the US Food and Drug Administration (FDA) was
based on 24-wk data. This agent is indicated for combination treatment with optimized background therapy in treatment-
experienced adults infected with only R5 virus who have evidence of viral replication and have HIV-1 strains resistant to
multiple antiretroviral agents.

https://emedicine.medscape.com/article/211316-print Page 48 of 71
05/09/2020, 11)59 PM

HIV, Entry Inhibitors

Class Summary
Approval of ibalizumab was based on the MB-301 phase 3 trial. MB-301 was a single-arm, 24-week study of ibalizumab
plus optimized background regimen (OBR) in treatment-experienced patients infected with multidrug-resistant HIV-1 virus.

The following study results were observed at 24 weeks:[166]

Among study participants, 43% achieved viral suppression < 50 copies/mL, and half achieved < 200 copies/mL.

While 60% of those with a baseline CD4 count of > 50 cells/µL achieved undetectable viral load, this fell to < 20% in those
with lower CD4 counts.

Among participants, 55% had at least a 1 log decrease and 48% had at least a 2 log decrease in HIV RNA; the average
reduction from baseline was 1.6 log.

The overall average CD4 cell gain was 48 cells/µL, but this differed according to baseline level; people who started with at
least 50 cells/µL saw a mean gain of about 75 cells/µL, while those with lower baseline levels gained an average of 9 cells/
µL.

Ibalizumab (Ibalizumab-uiyk, Trogarzo)


CD4-directed post-attachment inhibitor that prevents viral entry and fusion within the CD4 cell. Ibalizumab does not inhibit
HIV gp120 attachment to CD4; however, its postbinding conformational effects block the gp120-CD4 complex from
interacting with CCR5 or CXCR4 and thus prevents viral entry and fusion while preserving normal immunological function.
It is indicated for HIV-1 infection in heavily treated adults with multidrug-resistance. It is used in combination with the
patient’s current ART regimen.

Complete Regimen Combinations

Class Summary
Complete fixed-dose regimens assist with medication adherence. Two-, three-, and four-drug combination products are
available to decrease pill burden and administration frequency.

Dolutegravir/rilpivirine (Juluca)
First 2-drug, fixed-dose combination complete regimen approved for HIV infection. It is indicated for adults who are
virologically suppressed (HIV-1 RNA < 50 copies/mL) and on a stable ART regimen for ≥ 6 months with no history of
treatment failure and no known substitutions associated with resistance to dolutegravir or rilpivirine.

Dolutegravir/lamivudine (Dovato)
First 2-drug, fixed-dose, complete regimen for treatment-naïve adults with HIV-1 infection with no known substitutions
associated with resistance to dolutegravir or lamivudine. Combines integrase strand transfer inhibitor (INSTI) plus
nucleoside reverse transcriptase inhibitor (NRTI).

https://emedicine.medscape.com/article/211316-print Page 49 of 71
05/09/2020, 11)59 PM

Elvitegravir/cobicistat/emtricitabine/tenofovir AF (Genvoya)
Antiretroviral combination product that provides a complete, once-daily regimen for treatment-naïve adults and
adolescents aged ≥ 12 y, or to replace the current ART regimen in those who are virologically suppressed (HIV-1 RNA <
50 copies/mL) on a stable ART regimen for at least 6 months with no history of treatment failure. Each tablet contains an
integrase inhibitor (elvitegravir 150 mg) and 2 NRTIs (emtricitabine 200 mg and tenofovir AF 10 mg). It also contains
cobicistat, a CYP3A4 inhibitor used as a booster for elvitegravir, a CYP3A4 substrate.

Darunavir/cobicistat/emtricitabine/tenofovir AF (Symtuza)
Complete regimen indicated for treatment of HIV-1 infection in adults and adolescents weighing at least 40 kg who have
no prior antiretroviral (ART) treatment history or who are virologically suppressed (HIV-1 RNA < 50 copies/mL) on a stable
ART regimen for at least 6 months and have no known substitutions associated with resistance to darunavir or tenofovir.

Emtricitabine/rilpivirine/tenofovir AF (Odefsey)
Antiretroviral combination product that provides a complete, once-daily regimen for treatment-naïve adults and
adolescents aged ≥ 12 y, or to replace the current ART regimen in those who are virologically suppressed (HIV-1 RNA <
50 copies/mL) on a stable ART regimen for at least 6 months with no history of treatment failure. Each tablet contains 1
NNRTI (rilpivirine 25 mg) and 2 NRTIs (emtricitabine 200 mg and tenofovir AF 25 mg).

Elvitegravir/cobicistat/emtricitabine/tenofovir DF (Stribild)
Antiretroviral combination product that provides a complete, once-daily regimen for treatment-naïve adults. Contains an
integrase inhibitor and 2 NRTIs plus cobicistat, a CYP3A4 inhibitor used as a booster for elvitegravir, a CYP3A4 substrate.
Cobicistat enhances the systemic exposure of CYP3A substrates, such as elvitegravir, where bioavailability is limited and
half-life is shortened by CYP3A-dependent metabolism.

Emtricitabine/rilpivirine/tenofovir DF (Complera)
Complete regimen combination consisting of 2 NRTIs and 1 NNRTI. It is indicated for treatment of HIV-1 infection in
patients aged ≥ 12 years with no antiretroviral treatment history and with HIV-1 RNA ≤ 100,000 copies/mL at the start of
therapy, and in certain virologically-suppressed (HIV-1 RNA < 50 copies/mL) patients on a stable antiretroviral regimen at
start of therapy in order to replace their current antiretroviral treatment regimen.

Bictegravir/emtricitabine/tenofovir AF (Biktarvy)
Three-drug combination of bictegravir (BIC), an HIV-1 integrase strand transfer inhibitor (INSTI), and emtricitabine (FTC)
and tenofovir alafenamide (TAF), both HIV-1 nucleoside analog reverse transcriptase inhibitors (NRTIs). Indicated as a
complete regimen for treatment of HIV-1 infection in adults and pediatric patients weighing ≥25 kg who are ART-naïve or
replacement of current antiretroviral regimen in patients who are virologically suppressed (HIV-1 RNA

Efavirenz/lamivudine/tenofovir DF (Symfi, Symfi Lo)


Three-drug combination of a non-nucleoside reverse transcriptase inhibitor, and two nucleo(t)side reverse transcriptase
inhibitors and is indicated as a complete regimen for the treatment of human immunodeficiency virus type 1 (HIV-1)
infection in adults and children who weigh at least 35 kg (Symfi Lo) and 40 kg (Symfi).

Doravirine/lamivudine/tenofovir DF (Delstrigo)

https://emedicine.medscape.com/article/211316-print Page 50 of 71
05/09/2020, 11)59 PM

Indicated as a complete, once daily, regimen for treatment of HIV-1 infection in adults (1) who have not undergone
antiretroviral therapy (ART) or (2) as a replacement for the current antiretroviral regimen in patients who are who are
virologically suppressed (HIV-1 RNA

Antiretroviral Combinations

Class Summary
Combination products are valuable to patient care and help ensure compliance.

Abacavir, lamivudine, zidovudine (Trizivir)


Abacavir is a nucleoside reverse transcriptase inhibitor, which interferes with HIV viral RNA dependent DNA polymerase
and inhibits viral replication. Lamivudine and zidovudine are thymidine analogs that inhibit viral replication.

Abacavir/lamivudine (Epzicom)
This is a NRTI combination product. It is indicated in combination with other ART agents (eg, NNRTIs, PIs) for the
treatment of HIV-1 infection in adults and pediatric patients aged 12 years or older. It is also indicated in combination with
safer sex practices for preexposure prophylaxis (PrEP) to reduce the risk of sexually acquired HIV-1 in adults at high risk.
This indication is based on clinical trials in men who have sex with men (MSM) at high risk for HIV-1 infection and in
heterosexual serodiscordant couples. CDC guidelines include additional populations (eg, IV drug users, heterosexual
individuals at high risk).

Abacavir/dolutegravir/lamivudine (Triumeq)
Contains 2 NRTIs (abacavir 600 mg and lamivudine 300 mg) and an integrase strand inhibitor (dolutegravir 50 mg).
Dosage modification may be required when coadministered with strong CYP3A4 inducers by adding a single-entity
evening dose of dolutegravir.

Emtricitabine/tenofovir DF/efavirenz (Atripla)


NRTI and NNRTI combination product.

Lamivudine/zidovudine (Combivir)
NRTI combination product.

Emtricitabine/tenofovir DF (Truvada)
NRTI combination product. Indicated in combination with other ART agents (eg, NNRTIs, PIs) for the treatment of HIV-1
infection in adults. It is also approved for pediatric patients who weigh at least 17 kg and can swallow the tablet whole. In
addition, it is indicated in combination with safer sex practices for preexposure prophylaxis (PrEP) to reduce the risk of
sexually acquired HIV-1 in adults and adolescents at high risk. This indication is based on clinical trials in men who have
sex with men (MSM) at high risk for HIV-1 infection and in heterosexual serodiscordant couples. CDC guidelines include
additional populations (eg, IV drug users, heterosexual individuals at high risk).

https://emedicine.medscape.com/article/211316-print Page 51 of 71
05/09/2020, 11)59 PM

Emtricitabine/tenofovir AF (Descovy)
NRTI combination product. Indicated in combination with other ART agents (eg, NNRTIs, PIs) for the treatment of HIV-1
infection in adults and pediatric patients aged 12 y or older. In addition, it is indicated for HIV-1 preexposure prophylaxis
(PrEP) in at-risk adults and adolescents to reduce the risk of HIV-1 infection from sex, excluding those who have receptive
vaginal sex.

Lamivudine/tenofovir DF (Cimduo)
Combination contains 2 NRTIs (lamivudine 300 mg and tenofovir disoproxil fumarate 300 mg) and is indicated in
combination with other antiretroviral agents for the treatment of HIV-1 infection in adults and children who weigh at least
35 kg.

CYP3A4 Inhibitors

Class Summary
Boosting agents (eg, ritonavir, cobicistat) may be part of various ART drug regimens to inhibit metabolism of ART CYP3A
substrates, resulting in increased systemic exposure and efficacy.

Cobicistat (Tybost)
CYP3A inhibitor. As a single agent, it is indicated to increase systemic exposure of atazanavir or darunavir (once-daily
dosing regimen) in combination with other antiretroviral agents. It was originally approved as part of the combination
product elvitegravir/cobicistat/emtricitabine/tenofovir (Stribild).

Antibiotic, Sulfonamide Derivative

Class Summary
Therapy should cover all likely pathogens in this clinical setting.

Sulfamethoxazole and Trimethoprim (Bactrim, Bactrim DS, Septra, Septra DS,


Sulfatrim)
This combination inhibits bacterial synthesis of dihydrofolic acid by competing with paraaminobenzoic acid. This results in
inhibition of bacterial growth.

https://emedicine.medscape.com/article/211316-print Page 52 of 71
05/09/2020, 11)59 PM

Growth hormone releasing factor

Class Summary
This agent decreases visceral adipose tissue.

Tesamorelin (Egrifta)
Tesamorelin is a growth hormone–releasing factor (GRF) analog indicated for reduction of excess abdominal fat in
patients with HIV-associated lipodystrophy. This agent stimulates growth hormone production and increases serum levels
of insulin-like growth factor–1 (IGF-1). It elicits anabolic and lipolytic actions.

HIV, Attachment Inhibitors

Class Summary
HIV-1 attachment inhibitors decrease viral replication by binding directly to the glycoprotein 120 (gp120) subunit on the
surface of the virus.

Fostemsavir (Rukobia)
Prodrug of temsavir, a first-in-class glycoprotein 120 (gp120) attachment inhibitor. Binds directly to the gp120 subunit on
surface of the virus, thereby blocking HIV from attaching to host immune system CD4+ T cells and other immune cells. It is
indicated in combination with other ATV drugs for the treatment of HIV-1 infection in heavily treatment-experienced adults
with multidrug-resistant HIV-1 infection failing their current ART regimen owing to resistance, intolerance, or safety
considerations.

Questions & Answers


Overview

What is HIV and how is it transmitted?

What are the signs and symptoms of HIV infection?

What are risk factors for HIV exposure?

What are the recommendations for HIV screening?

What is the role of CD4 T-cell count in the risk assessment of opportunistic infection in HIV infection?

What is the role of viral load in peripheral blood in the management of HIV infection?

Which HIV test is FDA approved?

What tests are performed following the initial diagnosis of HIV infection?

https://emedicine.medscape.com/article/211316-print Page 53 of 71
05/09/2020, 11)59 PM

What are the three categories of HIV?

What are the DHHS guidelines for initiation of antiretroviral therapy (ART) for HIV infection?

What are the classes for antiretroviral agents used in the treatment of HIV infection?

Which regimens are preferred by the DHHS guidelines for treatment-naive patients with HIV infection?

What is the basis for regimen selection for treatment of HIV infection?

When is prophylaxis against opportunistic infections indicated in the management of HIV infection?

Which treatments may be needed in the management of HIV infection?

What are the CDC recommended HIV postexposure prophylaxis (PEP) and pre-exposure prophylaxis (PrEP) regimens?

What is HIV and how is it transmitted?

What is the most common route of HIV transmission?

How does the global distribution of HIV species vary?

What is the history of the HIV epidemic in the US?

What are the stigmas associated with HIV infection?

What common misinformation about HIV exists?

How has antiretroviral therapy (ART) affected HIV survival rates?

How is HIV-related health information linked to other health information?

What is the pathophysiology of HIV infection?

How is HIV characterized?

What are the three species-defining retroviral genes of HIV?

What are the HIV-1 accessory genes?

What are the accessory proteins of HIV?

What is the pathogenesis of AIDS?

What is the role of CD4+ helper T cells in the pathogenesis of HIV infection?

What is the global pattern for opportunistic infections in HIV infection?

What is the role of gut-associated lymphoid tissue (GALT) in the pathogenesis of HIV infection?

What is a feature of HIV replication in GALT?

How is the discrepancy in viral load between GALT and blood measurements explained?

What have the studies of T-cell–replication kinetics revealed about HIV infection?

What is the role of cell-cycling in the pathogenesis of HIV infection?

How do the HIV proteins affect T-cell function?

What is the role of direct cytotoxic effects of viral replication in the pathogenesis of HIV infection?

https://emedicine.medscape.com/article/211316-print Page 54 of 71
05/09/2020, 11)59 PM

Which individuals tend to be resistant to HIV infection?

What is the role of the chemokine-related receptor (CXCR4) in the pathogenesis of HIV infection?

What is the role of stem-cell transplantation in the management of HIV infection?

What is the relationship between T-cell loss and how HIV progresses to AIDS?

What is the role of lymph nodes in the pathogenesis of HIV infection?

How does HIV replicate?

How is HIV compartmentalized?

What are the three distinct phases of HIV pathogenesis?

What are the first cellular targets of HIV?

Where does HIV tend to integrate in the body?

What happens during acute seroconversion of HIV infection?

How long will it take for symptoms of HIV infection to appear?

What are the HIV symptoms during the acute seroconversion stage of infection?

When is AIDS diagnosed?

What is the primary mechanism for immunologic control of HIV infection?

When are patients at a higher risk for opportunistic infections and other AIDS-related events?

Which opportunistic infections and conditions are seen in patients with AIDS?

What is HIV encephalopathy?

What causes HIV infection?

What is the prevalence of HIV infection in the US?

What is the incidence of AIDS in the US?

Who is at higher risk of infection with HIV?

What group is most likely to be unaware they are HIV positive?

Among groups at high risk for HIV, what is the effect of health care visits?

What is the mortality rate of HIV infection?

How does the prevalence of HIV vary by age and race?

What is the global prevalence of HIV infection?

Which races and ethnicities in the US have the highest prevalence of HIV infection?

How does the prevalence of HIV vary by sex?

Who is at a higher risk for acquiring HIV infection?

What is the prognosis of untreated HIV infection?

https://emedicine.medscape.com/article/211316-print Page 55 of 71
05/09/2020, 11)59 PM

What is the efficacy of antiretroviral medications for treatment of HIV-2?

How long is the survival period for untreated AIDS?

What can increase survival for AIDS patients?

What is the mortality rate of AIDS in New York City?

What is the effect of pre-existing infections on people with AIDS?

Which organisms become pathogenic in patients with AIDS?

What are adverse effects of antiviral medications for AIDS?

What is the risk for non-Hodgkin lymphoma in patients with HIV infection?

What information about HIV should patients receive?

What are the risks of unprotected sex when both partners are HIV positive?

What information about HIV should IV drug users receive?

Presentation

What are the risk factors for exposure to HIV?

What are the signs and symptoms of HIV infection?

What are the signs and symptoms of AIDS?

What are the sequelae of HIV infection?

Which physical findings suggest HIV infections?

Which minor opportunistic infections suggest HIV infection?

DDX

What causes of immune suppression should be included in the differential diagnoses of HIV infection?

What are the differential diagnoses for HIV Infection and AIDS?

Workup

Why is screening for HIV infection needed?

What secondary testing is helpful in the diagnosis of HIV infection?

What are the CDC recommendations for HIV testing?

What is the Alere Determine HIV-1/2 Ag/Ab Combo test?

How is the HIV rapid test used?

What is the staging of HIV infection?

What baseline tests are performed in newly diagnosed patients with HIV infection?

What are the USPSTF recommendations for HIV screening?

What are the ACOG recommendations for HIV testing?

https://emedicine.medscape.com/article/211316-print Page 56 of 71
05/09/2020, 11)59 PM

What are the ACOG guidelines on HIV testing during pregnancy?

What are the CDC recommendations for HIV screening in high risk groups?

What are the benefits of early diagnosis of HIV infection?

What test should be used for HIV screening?

How are HIV test results reported?

When is HIV-2 testing indicated?

Which testing is most effective for early detection of HIV infection?

What is the CDC diagnostic algorithm for HIV diagnosis?

What is the role of CD4 T-cell count in the management of HIV infection?

What is the role of CD4 T-cell percentage in the management of HIV in children under five years of age?

What is the role of RT-PCR in the management of HIV infection?

What affects the rate of progression of HIV infection to AIDS?

How are HIV viral loads suppressed?

What is HIV virologic failure?

When is viral cultures for HIV indicted?

What is disrupted during HIV infection?

Which HIV test is performed only in newborns?

What can guide therapy for HIV infection?

What studies are performed in newly diagnosed patients with HIV infection?

Which tests are performed to evaluate tuberculosis infection in patients with newly diagnosed HIV infection?

Which tests is used to evaluate CMV infection in patients with newly diagnosed HIV infection?

Which tests are used to screen for syphilis in patients with newly diagnosed HIV infection?

Which STDs are evaluated in patients with newly diagnosed HIV infection?

Why is hepatitis serology performed in patients with newly diagnosed HIV infection?

What is toxoplasma antibody testing performed in newly diagnosed HIV infections?

What are the baseline values that may be affected by antiretroviral therapy (ART) for HIV infection?

What are histologic findings characteristic of HIV infection and AIDS?

What is the CDC classification of HIV infection?

What happens once an HIV infection has been staged into a higher clinical category?

Treatment

How is treatment for HIV infection determined?

https://emedicine.medscape.com/article/211316-print Page 57 of 71
05/09/2020, 11)59 PM

What is the principal treatment for HIV infection?

What are the results of successful HAART therapy for HIV infection?

Which secondary diseases may be reduced for HIV patients on HAART?

Who has released treatment guidelines for HIV infection?

What are the IDSA treatment guidelines for HIV infection?

How has the introduction of HAART affected mortality rates?

What increase in non-AIDS-defining cancer is attributed to HIV infection?

What are strong predictors of mortality for AIDS?

What are the adverse effects of delay in antiretroviral therapy (ART) for HIV infection?

At what CD4+T cell count should antiretroviral therapy (ART) for HIV be initiated?

What is the result of early initiation of antiretroviral therapy (ART) during the acute seroconversion period in HIV infection?

What are the IAS-USA and DHHS antiretroviral treatment (ART) recommendations for HIV infection?

What are the classes for antiretroviral agents for HIV infection?

What are the DHHS guidelines for treatment-naive patients with HIV infection?

How should HIV be treated during pregnancy?

What is the basis for antiretroviral regimen selection for HIV infection?

What the DHHS recommend for genotypic testing in HIV infection?

What mutations could result in increased susceptibility to a particular drug in HIV infection?

What is the impact of combination antiretroviral therapy (ART) on HIV/AIDS?

What are the benefits of and indications for rapid antiretroviral therapy (ART) start (Rapid ART Start) in the management
of HIV infection and AIDS?

What is the role of dolutegravir (Tivicay) in the treatment of HIV infection?

What is an important factor in selecting first-line antiretroviral therapy (ART) for HIV infection?

What is the role of zidovudine-lamivudine plus abacavir for the treatment of HIV infection?

What is the efficacy of elvitegravir/cobicistat/emtricitabine/tenofovir (Stribild) for the treatment of HIV infection?

What the benefits of 21-day antiretroviral regimens during pregnancy?

Why is prophylaxis for Pneumocystis jiroveci important for HIV management?

When is PCP prophylaxis discontinued in the management of HIV infection?

What is the role of TMP-SMX in the management of HIV infection?

When are HIV patients at risk for Mycobacterium avium complex infection?

When is prophylaxis for fungal or viral infections indicated in the management of HIV infection?

https://emedicine.medscape.com/article/211316-print Page 58 of 71
05/09/2020, 11)59 PM

What is the role of oral ganciclovir in the management of AIDS?

How should opportunistic infections be treated in HIV infection?

What is the relationship between TB and HIV infection?

What is HIV lipodystrophy?

What is the role of acyclovir in the treatment of HIV infection?

What is the role of crofelemer in the treatment of HIV infection?

How is HIV infection prevented?

What are sexual transmission prevention measures for HIV infection?

What is the role of STDs in the transmission of HIV?

Which sexual acts are more likely to lead to HIV infection than others?

How do oral contraceptives affect the risk for HIV transmission?

What are vertical transmission prevention measures for HIV infection?

How effective is triple antiviral therapy for HIV during pregnancy?

How is vertical transmission of HIV-2 prevented?

What is the mother-to-child transmission rate of HIV-2 in France?

What measures can be taken to prevent vertical HIV transmission?

What is the role of zidovudine in the prevention of vertical HIV transmission?

How is blood-borne transmission of HIV prevented?

What has the CDC recommended for HIV postexposure prophylaxis (PEP)?

What regimens does the CDC recommend for PEP?

What is the role of nevirapine in the prevention of HIV infection?

What HIV vaccinations are available?

What is the role of antiretroviral medications for the prevention of HIV transmission?

What is the efficacy of preexposure prophylaxis (PrEP) in the prevention of HIV infection?

What are the CDC guidelines for PrEP in HIV infection?

What is the efficacy of PrEP in preventing HIV infection?

What are the limitations in the US to broad implementation of PrEP for prevention of HIV infection?

What was the reduction in risk with compliant pre-exposure prophylaxis against HIV infection?

What is the role in topical antivirals in the prevention of HIV infection?

What does the International Antiviral Society-USA (IAS-USA) recommend for HIV prevention in adolescents and adults?

Which specialists should be consulted for all new cases of HIV infection?

https://emedicine.medscape.com/article/211316-print Page 59 of 71
05/09/2020, 11)59 PM

Which tests does the DHHS recommend be performed every 3 months in patients with HIV infection?

What is the role of basic chemistry studies in the management of HIV infection?

How often should fasting lipid profiles be measured in the management of HIV infection?

How often should T-cell counts be monitored in the management of HIV infection?

Guidelines

What are the CDC guidelines on HIV screening in men who have sex with men (MSM)?

What are the European AIDS Clinical Society guidelines on antiretroviral therapy (ART) and chronic HIV infection?

Medications

What is the role of antiretroviral therapy (ART) for HIV infection?

What are the benefits of combination therapy for the treatment of HIV infection?

Which antiretroviral combination products are used to treat HIV infection?

What have been shown to be safe and effective for treatment of black patients with HIV infection?

What is the role of Triumeq in the treatment of HIV infection?

What is the role of the NNRTI doravirine in the treatment of HIV infection?

Which medications in the drug class Antiretroviral agent, nucleoside reverse-transcriptase inhibitor are used in the
treatment of HIV Infection and AIDS?

Which medications in the drug class Antiretroviral Agent, Protease Inhibitor are used in the treatment of HIV Infection and
AIDS?

Which medications in the drug class Antiretroviral agent, non-nucleoside reverse-transcriptase inhibitor are used in the
treatment of HIV Infection and AIDS?

Which medications in the drug class Antiretroviral Agent, Integrase Inhibitor are used in the treatment of HIV Infection and
AIDS?

Which medications in the drug class Antiretroviral Agent, Fusion Inhibitor are used in the treatment of HIV Infection and
AIDS?

Which medications in the drug class Antiretroviral agent, CCR5 antagonist are used in the treatment of HIV Infection and
AIDS?

Which medications in the drug class HIV, Entry Inhibitors are used in the treatment of HIV Infection and AIDS?

Which medications in the drug class Complete Regimen Combinations are used in the treatment of HIV Infection and
AIDS?

Which medications in the drug class Antiretroviral Combinations are used in the treatment of HIV Infection and AIDS?

Which medications in the drug class CYP3A4 Inhibitors are used in the treatment of HIV Infection and AIDS?

Which medications in the drug class Antibiotic, Sulfonamide Derivative are used in the treatment of HIV Infection and
AIDS?

Which medications in the drug class Growth hormone releasing factor are used in the treatment of HIV Infection and
AIDS?

https://emedicine.medscape.com/article/211316-print Page 60 of 71
05/09/2020, 11)59 PM

Which medications in the drug class HIV, Attachment Inhibitors are used in the treatment of HIV Infection and AIDS?

Contributor Information and Disclosures

Author

Shelley A Gilroy, MD, FACP, FIDSA Associate Professor of Medicine, Infectious Disease and HIV Medicine, Albany
Medical College; Associate Chief of Staff for Education/DEO, Lead Physician for HIV Medicine, Division of Infectious
Diseases, Albany Stratton VA Medical Center

Shelley A Gilroy, MD, FACP, FIDSA is a member of the following medical societies: American College of Physicians,
American Medical Association, Infectious Diseases Society of America

Disclosure: Nothing to disclose.

Coauthor(s)

John J Faragon, PharmD, BCPS, AAHIVP Pharmacist, HIV and HCV Medicine, Division of HIV Medicine and
Department of Pharmacy, Albany Medical Center Hospital; Regional Pharmacy Director, Northeast Caribbean AIDS
Education and Training Centers, Columbia University; HIV and HCV Education Consultant, VirologyEd Consultants

Disclosure: Serve(d) as a speaker or a member of a speakers bureau for: Gilead; Janssen; Merck.

Chief Editor

Michael Stuart Bronze, MD David Ross Boyd Professor and Chairman, Department of Medicine, Stewart G Wolf
Endowed Chair in Internal Medicine, Department of Medicine, University of Oklahoma Health Science Center; Master of
the American College of Physicians; Fellow, Infectious Diseases Society of America; Fellow of the Royal College of
Physicians, London

Michael Stuart Bronze, MD is a member of the following medical societies: Alpha Omega Alpha, American College of
Physicians, American Medical Association, Association of Professors of Medicine, Infectious Diseases Society of America,
Oklahoma State Medical Association, Southern Society for Clinical Investigation

Disclosure: Nothing to disclose.

Additional Contributors

Nicholas John Bennett, MBBCh, PhD, MA(Cantab), FAAP Assistant Professor of Pediatrics, Co-Director of
Antimicrobial Stewardship, Medical Director, Division of Pediatric Infectious Diseases and Immunology, Connecticut
Children's Medical Center

Nicholas John Bennett, MBBCh, PhD, MA(Cantab), FAAP is a member of the following medical societies: Alpha Omega
Alpha, American Academy of Pediatrics

Disclosure: Received research grant from: Cubist<br/>Received income in an amount equal to or greater than $250 from:
Horizon Pharmaceuticals, Shire<br/>Medico legal consulting for: Various.

Acknowledgements

Aaron Glatt, MD Professor of Clinical Medicine, New York Medical College; President and CEO, Former Chief Medical
Officer, Departments of Medicine and Infectious Diseases, St Joseph Hospital (formerly New Island Hospital)

Aaron Glatt, MD is a member of the following medical societies: American College of Chest Physicians, American College
of Physician Executives, American College of Physicians, American College of Physicians-American Society of Internal
Medicine, American Medical Association, American Society for Microbiology, American Thoracic Society, American

https://emedicine.medscape.com/article/211316-print Page 61 of 71
05/09/2020, 11)59 PM

Venereal Disease Association, Infectious Diseases Society of America, International AIDS Society, and Society
forHealthcare Epidemiology of America

Disclosure: Nothing to disclose.

Mary L Windle, PharmD Adjunct Associate Professor, University of Nebraska Medical Center College of Pharmacy; Editor-
in-Chief, Medscape Drug Reference

Disclosure: Nothing to disclose.

References

1. US Preventive Services Task Force., Owens DK, Davidson KW, Krist AH, Barry MJ, Cabana M, et al. Screening for HIV
Infection: US Preventive Services Task Force Recommendation Statement. JAMA. 2019 Jun 18. 321 (23):2326-2336. [Medline].

2. Branson BM, Handsfield HH, Lampe MA, et al. Revised recommendations for HIV testing of adults, adolescents, and pregnant
women in health-care settings. MMWR Recomm Rep. 2006 Sep 22. 55:1-17; quiz CE1-4. [Medline].

3. Qaseem A, Snow V, Shekelle P, Hopkins R Jr, Owens DK. Screening for HIV in health care settings: a guidance statement from
the American College of Physicians and HIV Medicine Association. Ann Intern Med. 2009 Jan 20. 150(2):125-31. [Medline].

4. Centers for Disease Control and Prevention. Laboratory testing for the diagnosis of HIV infection : updated recommendations.
Centers for Disease Control and Prevention. Available at ttps://stacks.cdc.gov/view/cdc/23447. June 27, 2014; Accessed: March
5, 2020.

5. World Health Organization (WHO). HIV/AIDS. World Health Organization (WHO). Available at https://www.who.int/health-
topics/hiv-aids/#tab=tab_1.

6. Reynolds SJ, Makumbi F, Newell K, et al. Effect of daily aciclovir on HIV disease progression in individuals in Rakai, Uganda, co-
infected with HIV-1 and herpes simplex virus type 2: a randomised, double-blind placebo-controlled trial. Lancet Infect Dis. 2012
Jun. 12(6):441-8. [Medline]. [Full Text].

7. [Guideline] Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines for the use of antiretroviral agents in HIV-1-
infected adults and adolescents. Department of Health and Human Services. October 17, 2017. U.S. Department of Health and
Human Services. Available at https://aidsinfo.nih.gov/guidelines/html/1/adult-and-adolescent-arv/0. December 18, 2019;
Accessed: March 5, 2020.

8. US Food and Drug Administration. FDA approves first rapid diagnostic test to detect both HIV-1 antigen and HIV-1/2 antibodies.
US Department of Health and Human Services, US Food and Drug Administration. Available at
http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm364480.htm. Accessed: August 12, 2013.

9. Lowes R. FDA OKs First Rapid Test for HIV-1/2 Antibodies, HIV-1 Antigen. Medscape [serial online]. Available at
http://www.medscape.com/viewarticle/809183. Accessed: August 15, 2013.

10. 1993 revised classification system for HIV infection and expanded surveillance case definition for AIDS among adolescents and
adults. MMWR Recomm Rep. 1992 Dec 18. 41:1-19. [Medline].

11. [Guideline] AIDSInfo. Guidelines for the Use of Antiretroviral Agents in Adults and Adolescents with HIV. AIDSInfo. Available at
https://aidsinfo.nih.gov/contentfiles/lvguidelines/adultandadolescentgl.pdf. December 18, 2019; Accessed: January 24, 2020.

12. U.S. Department of Health and Human Services. AIDSInfo. AIDSInfo. Available at https://aidsinfo.nih.gov/.

13. Jeffrey S. FDA approves first antidiarrheal drug for HIV/AIDS. Medscape Medical News. Dec 31, 2012. [Full Text].

14. Gao F, Bailes E, Robertson DL, et al. Origin of HIV-1 in the chimpanzee Pan troglodytes troglodytes. Nature. 1999 Feb 4.
397(6718):436-41. [Medline].

15. Hirsch VM, Olmsted RA, Murphey-Corb M, Purcell RH, Johnson PR. An African primate lentivirus (SIVsm) closely related to HIV-
2. Nature. 1989 Jun 1. 339(6223):389-92. [Medline].

16. Popper SJ, Sarr AD, Gueye-Ndiaye A, Mboup S, Essex ME, Kanki PJ. Low plasma human immunodeficiency virus type 2 viral
load is independent of proviral load: low virus production in vivo. J Virol. 2000 Feb. 74(3):1554-7. [Medline]. [Full Text].

https://emedicine.medscape.com/article/211316-print Page 62 of 71
05/09/2020, 11)59 PM

17. Popper SJ, Sarr AD, Travers KU, et al. Lower human immunodeficiency virus (HIV) type 2 viral load reflects the difference in
pathogenicity of HIV-1 and HIV-2. J Infect Dis. 1999 Oct. 180(4):1116-21. [Medline].

18. Mellors JW, Munoz A, Giorgi JV, et al. Plasma viral load and CD4+ lymphocytes as prognostic markers of HIV-1 infection. Ann
Intern Med. 1997 Jun 15. 126(12):946-54. [Medline].

19. Rodríguez B, Sethi AK, Cheruvu VK, et al. Predictive value of plasma HIV RNA level on rate of CD4 T-cell decline in untreated
HIV infection. JAMA. 2006 Sep 27. 296(12):1498-506. [Medline].

20. Kaposi's sarcoma and Pneumocystis pneumonia among homosexual men--New York City and California. MMWR Morb Mortal
Wkly Rep. 1981 Jul 3. 30(25):305-8. [Medline].

21. Barre-Sinoussi F, Chermann JC, Rey F, et al. Isolation of a T-lymphotropic retrovirus from a patient at risk for acquired immune
deficiency syndrome (AIDS). Science. 1983 May 20. 220(4599):868-71. [Medline].

22. Ascher MS, Sheppard HW, Winkelstein W Jr, Vittinghoff E. Does drug use cause AIDS?. Nature. 1993 Mar 11. 362(6416):103-4.
[Medline].

23. Korber B, Muldoon M, Theiler J, et al. Timing the ancestor of the HIV-1 pandemic strains. Science. 2000 Jun 9. 288(5472):1789-
96. [Medline].

24. Koopman G, Haaksma AG, ten Velden J, Hack CE, Heeney JL. The relative resistance of HIV type 1-infected chimpanzees to
AIDS correlates with the maintenance of follicular architecture and the absence of infiltration by CD8+ cytotoxic T lymphocytes.
AIDS Res Hum Retroviruses. 1999 Mar 1. 15(4):365-73. [Medline].

25. Birch MR, Learmont JC, Dyer WB, et al. An examination of signs of disease progression in survivors of the Sydney Blood Bank
Cohort (SBBC). J Clin Virol. 2001 Oct. 22(3):263-70. [Medline].

26. Dyer WB, Geczy AF, Kent SJ, et al. Lymphoproliferative immune function in the Sydney Blood Bank Cohort, infected with natural
nef/long terminal repeat mutants, and in other long-term survivors of transfusion-acquired HIV-1 infection. AIDS. 1997 Nov.
11(13):1565-74. [Medline].

27. Pantaleo G, Graziosi C, Fauci AS. New concepts in the immunopathogenesis of human immunodeficiency virus infection. N Engl
J Med. 1993 Feb 4. 328(5):327-35. [Medline].

28. Pantaleo G, Fauci AS. New concepts in the immunopathogenesis of HIV infection. Annu Rev Immunol. 1995. 13:487-512.
[Medline].

29. Weber J. The pathogenesis of HIV-1 infection. Br Med Bull. 2001. 58:61-72. [Medline].

30. Frazer IH, Mackay IR, Crapper RM, et al. Immunological abnormalities in asymptomatic homosexual men: correlation with
antibody to HTLV-III and sequential changes over two years. Q J Med. 1986 Oct. 61(234):921-33. [Medline].

31. Schechter MT, Boyko WJ, Craib KJ, et al. Effects of long-term seropositivity to human immunodeficiency virus in a cohort of
homosexual men. AIDS. 1987 Jul. 1(2):77-82. [Medline].

32. Talal AH, Irwin CE, Dieterich DT, Yee H, Zhang L. Effect of HIV-1 infection on lymphocyte proliferation in gut-associated lymphoid
tissue. J Acquir Immune Defic Syndr. 2001 Mar 1. 26(3):208-17. [Medline].

33. Poles MA, Boscardin WJ, Elliott J, et al. Lack of decay of HIV-1 in gut-associated lymphoid tissue reservoirs in maximally
suppressed individuals. J Acquir Immune Defic Syndr. 2006 Sep. 43(1):65-8. [Medline].

34. van Marle G, Gill MJ, Kolodka D, McManus L, Grant T, Church DL. Compartmentalization of the gut viral reservoir in HIV-1
infected patients. Retrovirology. 2007 Dec 4. 4:87. [Medline]. [Full Text].

35. Talal AH, Monard S, Vesanen M, et al. Virologic and immunologic effect of antiretroviral therapy on HIV-1 in gut-associated
lymphoid tissue. J Acquir Immune Defic Syndr. 2001 Jan 1. 26(1):1-7. [Medline].

36. Guadalupe M, Reay E, Sankaran S, et al. Severe CD4+ T-cell depletion in gut lymphoid tissue during primary human
immunodeficiency virus type 1 infection and substantial delay in restoration following highly active antiretroviral therapy. J Virol.
2003 Nov. 77(21):11708-17. [Medline]. [Full Text].

37. Shacklett BL, Cox CA, Sandberg JK, Stollman NH, Jacobson MA, Nixon DF. Trafficking of human immunodeficiency virus type 1-
specific CD8+ T cells to gut-associated lymphoid tissue during chronic infection. J Virol. 2003 May. 77(10):5621-31. [Medline].

https://emedicine.medscape.com/article/211316-print Page 63 of 71
05/09/2020, 11)59 PM

[Full Text].

38. Guadalupe M, Sankaran S, George MD, et al. Viral suppression and immune restoration in the gastrointestinal mucosa of human
immunodeficiency virus type 1-infected patients initiating therapy during primary or chronic infection. J Virol. 2006 Aug.
80(16):8236-47. [Medline]. [Full Text].

39. Al-Harthi L, Marchetti G, Steffens CM, Poulin J, Sékaly R, Landay A. Detection of T cell receptor circles (TRECs) as biomarkers
for de novo T cell synthesis using a quantitative polymerase chain reaction-enzyme linked immunosorbent assay (PCR-ELISA).
J Immunol Methods. 2000 Apr 3. 237(1-2):187-97. [Medline].

40. Hellerstein M, Hanley MB, Cesar D, et al. Directly measured kinetics of circulating T lymphocytes in normal and HIV-1-infected
humans. Nat Med. 1999 Jan. 5(1):83-9. [Medline].

41. Bandera A, Ferrario G, Saresella M, et al. CD4+ T cell depletion, immune activation and increased production of regulatory T
cells in the thymus of HIV-infected individuals. PLoS One. 2010 May 24. 5(5):e10788. [Medline]. [Full Text].

42. Franco JM, Rubio A, Martínez-Moya M, et al. T-cell repopulation and thymic volume in HIV-1-infected adult patients after highly
active antiretroviral therapy. Blood. 2002 May 15. 99(10):3702-6. [Medline].

43. Mohri H, Perelson AS, Tung K, et al. Increased turnover of T lymphocytes in HIV-1 infection and its reduction by antiretroviral
therapy. J Exp Med. 2001 Nov 5. 194(9):1277-87. [Medline]. [Full Text].

44. Bird JJ, Brown DR, Mullen AC, et al. Helper T cell differentiation is controlled by the cell cycle. Immunity. 1998 Aug. 9(2):229-37.
[Medline].

45. Re F, Braaten D, Franke EK, Luban J. Human immunodeficiency virus type 1 Vpr arrests the cell cycle in G2 by inhibiting the
activation of p34cdc2-cyclin B. J Virol. 1995 Nov. 69(11):6859-64. [Medline]. [Full Text].

46. Keating SM, Golub ET, Nowicki M, et al. The effect of HIV infection and HAART on inflammatory biomarkers in a population-
based cohort of women. AIDS. 2011 Sep 24. 25(15):1823-32. [Medline]. [Full Text].

47. Landires I, Bugault F, Lambotte O, et al. HIV infection perturbs interleukin-7 signaling at the step of STAT5 nuclear relocalization.
AIDS. 2011 Sep 24. 25(15):1843-53. [Medline].

48. Lederman MM. Immune restoration and CD4+ T-cell function with antiretroviral therapies. AIDS. 2001 Feb. 15 Suppl 2:S11-5.
[Medline].

49. Finkel TH, Tudor-Williams G, Banda NK, et al. Apoptosis occurs predominantly in bystander cells and not in productively infected
cells of HIV- and SIV-infected lymph nodes. Nat Med. 1995 Feb. 1(2):129-34. [Medline].

50. 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 Aug 9. 86(3):367-77. [Medline].

51. Samson M, Libert F, Doranz BJ, et al. Resistance to HIV-1 infection in caucasian individuals bearing mutant alleles of the CCR-5
chemokine receptor gene. Nature. 1996 Aug 22. 382(6593):722-5. [Medline].

52. Poropatich K, Sullivan DJ Jr. Human immunodeficiency virus type 1 long-term non-progressors: the viral, genetic and
immunological basis for disease non-progression. J Gen Virol. 2011 Feb. 92:247-68. [Medline].

53. van der Ende ME, Schutten M, Raschdorff B, et al. CD4 T cells remain the major source of HIV-1 during end stage disease.
AIDS. 1999 Jun 18. 13(9):1015-9. [Medline].

54. Allers K, Hutter G, Hofmann J, et al. Evidence for the cure of HIV infection by CCR5?32/?32 stem cell transplantation. Blood.
2011 Mar 10. 117(10):2791-9. [Medline].

55. Kostense S, Raaphorst FM, Notermans DW, et al. Diversity of the T-cell receptor BV repertoire in HIV-1-infected patients reflects
the biphasic CD4+ T-cell repopulation kinetics during highly active antiretroviral therapy. AIDS. 1998 Dec 24. 12(18):F235-40.
[Medline].

56. McCune JM. The dynamics of CD4+ T-cell depletion in HIV disease. Nature. 2001 Apr 19. 410(6831):974-9. [Medline].

57. 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 Sep 28. 15(14):1749-56. [Medline].

https://emedicine.medscape.com/article/211316-print Page 64 of 71
05/09/2020, 11)59 PM

58. Pantaleo G, Graziosi C, Demarest JF, et al. HIV infection is active and progressive in lymphoid tissue during the clinically latent
stage of disease. Nature. 1993 Mar 25. 362(6418):355-8. [Medline].

59. Vago L, Antonacci MC, Cristina S, et al. Morphogenesis, evolution and prognostic significance of lymphatic tissue lesions in HIV
infection. Appl Pathol. 1989. 7(5):298-309. [Medline].

60. Edén A, Fuchs D, Hagberg L, et al. HIV-1 viral escape in cerebrospinal fluid of subjects on suppressive antiretroviral treatment. J
Infect Dis. 2010 Dec 15. 202(12):1819-25. [Medline]. [Full Text].

61. Karris MY, Anderson CM, Morris SR, Smith DM, Little SJ. Cost savings associated with testing of antibodies, antigens, and
nucleic acids for diagnosis of acute HIV infection. J Clin Microbiol. 2012 Jun. 50(6):1874-8. [Medline]. [Full Text].

62. Pruss D, Bushman FD, Wolffe AP. Human immunodeficiency virus integrase directs integration to sites of severe DNA distortion
within the nucleosome core. Proc Natl Acad Sci U S A. 1994 Jun 21. 91(13):5913-7. [Medline].

63. Schröder AR, Shinn P, Chen H, Berry C, Ecker JR, Bushman F. HIV-1 integration in the human genome favors active genes and
local hotspots. Cell. 2002 Aug 23. 110(4):521-9. [Medline].

64. Blankson JN, Persaud D, Siliciano RF. The challenge of viral reservoirs in HIV-1 infection. Annu Rev Med. 2002. 53:557-93.
[Medline].

65. Chun TW, Engel D, Berrey MM, Shea T, Corey L, Fauci AS. Early establishment of a pool of latently infected, resting CD4(+) T
cells during primary HIV-1 infection. Proc Natl Acad Sci U S A. 1998 Jul 21. 95(15):8869-73. [Medline].

66. Ho DD, Moudgil T, Alam M. Quantitation of human immunodeficiency virus type 1 in the blood of infected persons. N Engl J Med.
1989 Dec 14. 321(24):1621-5. [Medline].

67. Saez-Cirion A, Lacabaratz C, Lambotte O, et al. HIV controllers exhibit potent CD8 T cell capacity to suppress HIV infection ex
vivo and peculiar cytotoxic T lymphocyte activation phenotype. Proc Natl Acad Sci U S A. 2007 Apr 17. 104(16):6776-81.
[Medline]. [Full Text].

68. Kaul R, Plummer FA, Kimani J, et al. HIV-1-specific mucosal CD8+ lymphocyte responses in the cervix of HIV-1-resistant
prostitutes in Nairobi. J Immunol. 2000 Feb 1. 164(3):1602-11. [Medline].

69. Alimonti JB, Kimani J, Matu L, et al. Characterization of CD8 T-cell responses in HIV-1-exposed seronegative commercial sex
workers from Nairobi, Kenya. Immunol Cell Biol. 2006 Oct. 84(5):482-5. [Medline].

70. Alter G, Heckerman D, Schneidewind A, et al. HIV-1 adaptation to NK-cell-mediated immune pressure. Nature. 2011 Aug 3.
476(7358):96-100. [Medline]. [Full Text].

71. Zoufaly A, an der Heiden M, Kollan C, et al. Clinical outcome of HIV-infected patients with discordant virological and
immunological response to antiretroviral therapy. J Infect Dis. 2011 Feb 1. 203(3):364-71. [Medline]. [Full Text].

72. Mills EJ, Bakanda C, Birungi J, Yaya S, Ford N. The prognostic value of baseline CD4 cell count beyond 6 months of
antiretroviral therapy in HIV positive patients in Uganda. AIDS. 2012 Apr 21. [Medline].

73. Ezeamama AE, Spiegelman D, Hertzmark E, et al. HIV infection and the incidence of malaria among HIV-exposed children from
Tanzania. J Infect Dis. 2012 May 15. 205(10):1486-94. [Medline]. [Full Text].

74. Lambert-Niclot S, Tubiana R, Beaudoux C, et al. Detection of HIV-1 RNA in seminal plasma samples from treated patients with
undetectable HIV-1 RNA in blood plasma on a 2002-2011 survey. AIDS. 2012 May 15. 26(8):971-5. [Medline].

75. Scherzer R, Estrella M, Li Y, et al. Association of tenofovir exposure with kidney disease risk in HIV infection. AIDS. 2012 Apr 24.
26(7):867-75. [Medline].

76. CDC. Diagnoses of HIV Infection in the United States and Dependent Areas, 2018 (Preliminary), Volume 30. CDC. Available at
https://www.cdc.gov/hiv/pdf/library/reports/surveillance/cdc-hiv-surveillance-report-2018-vol-30.pdf. November 2019; Accessed:
March 5, 2020.

77. Centers for Disease Control and Prevention. Diagnoses of HIV Infection in the United States and Dependent Areas, 2018
(Preliminary). Available at http://www.cdc.gov/hiv/topics/surveillance/resources/reports/. December 2018; Accessed: March 5,
2020.

78. Centers for Disease Control and Prevention. Prevalence and awareness of HIV infection among men who have sex with men ---

https://emedicine.medscape.com/article/211316-print Page 65 of 71
05/09/2020, 11)59 PM

21 cities, United States, 2008. MMWR Morb Mortal Wkly Rep. 2010 Sep 24. 59(37):1201-7. [Medline].

79. Xu JQ, Kochanek KD, Murphy SL, Tejada-Vera B. Deaths: Final data for 2007. National vital statistics reports; vol 58 no 19.
Hyattsville, MD: National Center for Health Statistics. 2010. Available at
http://www.cdc.gov/NCHS/data/nvsr/nvsr58/nvsr58_19.pdf. Accessed: June 21, 2011.

80. Vital Signs: HIV Infection, Testing, and Risk Behaviors Among Youths - United States. MMWR Morb Mortal Wkly Rep. 2012 Nov
30. 61(47):971-6. [Medline]. [Full Text].

81. UNAIDS. Global AIDS update 2019 — Communities at the centre. UNAIDS. Available at
https://www.unaids.org/sites/default/files/media_asset/2019-global-AIDS-update_en.pdf. December 10, 2019; Accessed: March
5, 2020.

82. Ng M, Gakidou E, Levin-Rector A, Khera A, Murray CJ, Dandona L. Assessment of population-level effect of Avahan, an HIV-
prevention initiative in India. Lancet. 2011 Nov 5. 378(9803):1643-52. [Medline].

83. Greenhalgh H. Men With Same-Sex Partners 28 Times More Likely to Get HIV - U.N. Medscape News. Available at
https://www.medscape.com/viewarticle/899482. July 19, 2018; Accessed: July 24, 2018.

84. Creswell JD, Myers HF, Cole SW, Irwin MR. Mindfulness meditation training effects on CD4+ T lymphocytes in HIV-1 infected
adults: a small randomized controlled trial. Brain Behav Immun. 2009 Feb. 23(2):184-8. [Medline]. [Full Text].

85. Cao Y, Qin L, Zhang L, Safrit J, Ho DD. Virologic and immunologic characterization of long-term survivors of human
immunodeficiency virus type 1 infection. N Engl J Med. 1995 Jan 26. 332(4):201-8. [Medline].

86. Levy JA. HIV pathogenesis and long-term survival. AIDS. 1993 Nov. 7(11):1401-10. [Medline].

87. Pantaleo G, Menzo S, Vaccarezza M, et al. Studies in subjects with long-term nonprogressive human immunodeficiency virus
infection. N Engl J Med. 1995 Jan 26. 332(4):209-16. [Medline].

88. Paroli M, Propato A, Accapezzato D, Francavilla V, Schiaffella E, Barnaba V. The immunology of HIV-infected long-term non-
progressors--a current view. Immunol Lett. 2001 Nov 1. 79(1-2):127-9. [Medline].

89. Nesheim SR, Kapogiannis BG, Soe MM, et al. Trends in opportunistic infections in the pre- and post-highly active antiretroviral
therapy eras among HIV-infected children in the Perinatal AIDS Collaborative Transmission Study, 1986-2004. Pediatrics. 2007
Jul. 120(1):100-9. [Medline].

90. Sackoff JE, Hanna DB, Pfeiffer MR, Torian LV. Causes of death among persons with AIDS in the era of highly active antiretroviral
therapy: New York City. Ann Intern Med. 2006 Sep 19. 145(6):397-406. [Medline]. [Full Text].

91. Palella FJ Jr, Baker RK, Moorman AC, et al. Mortality in the highly active antiretroviral therapy era: changing causes of death
and disease in the HIV outpatient study. J Acquir Immune Defic Syndr. 2006 Sep. 43(1):27-34. [Medline].

92. Brooks M. Low Level HIV Viremia a Modifiable Risk Factor for Non-Hodgkin Lymphoma. Medscape Medical News. Available at
http://www.medscape.com/viewarticle/821277. Accessed: March 12, 2014.

93. Achenbach CJ, Buchanan AL, Cole SR, Hou L, Mugavero MJ, Crane HM, et al. HIV viremia and incidence of non-Hodgkin
lymphoma in patients successfully treated with antiretroviral therapy. Clin Infect Dis. 2014 Feb 12. [Medline].

94. [Guideline] Brooks M. New CDC HIV Testing Recommendations Offer Faster Diagnosis. Medscape Medical News. Jun 26 2014.
[Full Text].

95. [Guideline] CDC. Laboratory Testing for the Diagnosis of HIV Infection: Updated Recommendations. Centers for Disease Control
and Prevention. Available at http://www.cdc.gov/hiv/pdf/HIVtestingAlgorithmRecommendation-Final.pdf. Accessed: Jul 7 2014.

96. Barclay L. ACOG Guidelines Recommend Repeat HIV Screening, Prophylaxis. Medscape Medical News. Available at
http://www.medscape.com/viewarticle/824112. Accessed: May 3, 2014.

97. [Guideline] Committee Opinion No 596: Routine Human Immunodeficiency Virus Screening. Obstet Gynecol. 2014 May.
123(5):1137-1139. [Medline].

98. Skwarecki B. ACOG updates recommendations for prenatal HIV testing. Available at:. Medscape Medical News. WebMD Inc.
Available at http://www.medscape.com/viewarticle/845416. May 27, 2015;

https://emedicine.medscape.com/article/211316-print Page 66 of 71
05/09/2020, 11)59 PM

99. [Guideline] American College of Obstetricians and Gynecologists. Committee opinion no: 635: prenatal and perinatal human
immunodeficiency virus testing: expanded recommendations. Obstet Gynecol. 2015 Jun. 125 (6):1544-7. [Medline].

100. Torian LV, Eavey JJ, Punsalang AP, et al. HIV type 2 in New York City, 2000-2008. Clin Infect Dis. 2010 Dec 1. 51(11):1334-42.
[Medline].

101. Claassen CW, Diener-West M, Mehta SH, Thomas DL, Kirk GD. Discordance Between CD4+ T-Lymphocyte Counts and
Percentages in HIV-Infected Persons With Liver Fibrosis. Clin Infect Dis. 2012 Jun. 54(12):1806-13. [Medline].

102. Detection of Acute HIV Infection in Two Evaluations of a New HIV Diagnostic Testing Algorithm - United States, 2011-2013.
MMWR Morb Mortal Wkly Rep. 2013 Jun 21. 62(24):489-94. [Medline].

103. Hull MW, Rollet K, Odueyungbo A, et al. Factors associated with discordance between absolute CD4 cell count and CD4 cell
percentage in patients coinfected with HIV and hepatitis C virus. Clin Infect Dis. 2012 Jun. 54(12):1798-805. [Medline].

104. Hoffmann CJ, Brown TT. Thyroid function abnormalities in HIV-infected patients. Clin Infect Dis. 2007 Aug 15. 45(4):488-94.
[Medline].

105. Lee PL, Yiannoutsos CT, Ernst T, et al. A multi-center 1H MRS study of the AIDS dementia complex: validation and preliminary
analysis. J Magn Reson Imaging. 2003 Jun. 17(6):625-33. [Medline].

106. Bucy RP, Hockett RD, Derdeyn CA, et al. Initial increase in blood CD4(+) lymphocytes after HIV antiretroviral therapy reflects
redistribution from lymphoid tissues. J Clin Invest. 1999 May 15. 103(10):1391-8. [Medline]. [Full Text].

107. Pakker NG, Notermans DW, de Boer RJ, et al. Biphasic kinetics of peripheral blood T cells after triple combination therapy in
HIV-1 infection: a composite of redistribution and proliferation. Nat Med. 1998 Feb. 4(2):208-14. [Medline].

108. Masia M, Padilla S, Alvarez D, et al. Risk, predictors, and mortality associated with non-AIDS events in newly diagnosed HIV-
infected patients: role of antiretroviral therapy. AIDS. 2013 Jan 14. 27(2):181-9. [Medline].

109. Barclay L. HIV: guidelines stress role of primary care in management. Medscape Medical News. November 14, 2013. [Full Text].

110. Aberg JA, Gallant JE, Ghanem KG, Emmanuel P, Zingman BS, Horberg MA. Primary Care Guidelines for the Management of
Persons Infected With HIV: 2013 Update by the HIV Medicine Association of the Infectious Diseases Society of America. Clin
Infect Dis. 2013 Nov 13. [Medline].

111. Simard EP, Engels EA. Cancer as a cause of death among people with AIDS in the United States. Clin Infect Dis. 2010 Oct 15.
51(8):957-62. [Medline]. [Full Text].

112. Shiels MS, Pfeiffer RM, Gail MH, et al. Cancer burden in the HIV-infected population in the United States. J Natl Cancer Inst.
2011 May 4. 103(9):753-62. [Medline]. [Full Text].

113. Puhan MA, Van Natta ML, Palella FJ, Addessi A, Meinert C. Excess mortality in patients with AIDS in the era of highly active
antiretroviral therapy: temporal changes and risk factors. Clin Infect Dis. 2010 Oct 15. 51(8):947-56. [Medline]. [Full Text].

114. Sterne JA, May M, Costagliola D, et al. Timing of initiation of antiretroviral therapy in AIDS-free HIV-1-infected patients: a
collaborative analysis of 18 HIV cohort studies. Lancet. 2009 Apr 18. 373(9672):1352-63. [Medline]. [Full Text].

115. Severe P, Juste MA, Ambroise A, et al. Early versus standard antiretroviral therapy for HIV-infected adults in Haiti. N Engl J Med.
2010 Jul 15. 363(3):257-65. [Medline].

116. Cain LE, Logan R, Robins JM, et al. When to initiate combined antiretroviral therapy to reduce mortality and AIDS-defining
illness in HIV-infected persons in developed countries: an observational study. Ann Intern Med. 2011 Apr 19. 154(8):509-15.
[Medline].

117. US Department of Health and Human Services. National Institutes of Health. NIH News. Starting Antiretroviral Therapy Earlier
Yields Better Clinical Outcomes. June 8, 2009.

118. Hecht FM, Wang L, Collier A, et al. A multicenter observational study of the potential benefits of initiating combination
antiretroviral therapy during acute HIV infection. J Infect Dis. 2006 Sep 15. 194(6):725-33. [Medline].

119. Hecht FM, Wellman R, Busch MP, et al. Identifying the early post-HIV antibody seroconversion period. J Infect Dis. 2011 Aug 15.
204(4):526-33. [Medline]. [Full Text].

https://emedicine.medscape.com/article/211316-print Page 67 of 71
05/09/2020, 11)59 PM

120. Saag MS, Benson CA, Gandhi RT, Hoy JF, Landovitz RJ, Mugavero MJ, et al. Antiretroviral Drugs for Treatment and Prevention
of HIV Infection in Adults: 2018 Recommendations of the International Antiviral Society-USA Panel. JAMA. 2018 Jul 24. 320
(4):379-396. [Medline].

121. Rhee SY, Taylor J, Fessel WJ, et al. HIV-1 protease mutations and protease inhibitor cross-resistance. Antimicrob Agents
Chemother. 2010 Oct. 54(10):4253-61. [Medline]. [Full Text].

122. Lennox JL, DeJesus E, Lazzarin A, et al. Safety and efficacy of raltegravir-based versus efavirenz-based combination therapy in
treatment-naive patients with HIV-1 infection: a multicentre, double-blind randomised controlled trial. Lancet. 2009 Sep 5.
374(9692):796-806. [Medline].

123. Rockstroh JK, DeJesus E, Lennox JL, et al. Durable efficacy and safety of raltegravir versus efavirenz when combined with
tenofovir/emtricitabine in treatment-naive HIV-1-infected patients: final 5-year results from STARTMRK. J Acquir Immune Defic
Syndr. 2013 May 1. 63(1):77-85. [Medline].

124. Raffi F, Rachlis A, Stellbrink HJ, Hardy WD, Torti C, Orkin C, et al. Once-daily dolutegravir versus raltegravir in antiretroviral-
naive adults with HIV-1 infection: 48 week results from the randomised, double-blind, non-inferiority SPRING-2 study. Lancet.
2013 Mar 2. 381(9868):735-43. [Medline].

125. Walmsley S, Antela A, Clumeck N, et al. Dolutegravir (DTG; S/GSK1349572) + Abacavir/Lamivudine Once Daily Statistically
Superior to Tenofovir/Emtricitabine/Efavirenz: 48-Week Results - SINGLE (ING114467). Abstract presented at: 52nd Interscience
Conference on Antimicrobial Agents and Chemotherapy (ICAAC). Sept 2012. Abstract H-556b:

126. Cahn P, Pozniak AL, Mingrone H, Shuldyakov A, Brites C, Andrade-Villanueva JF, et al. Dolutegravir versus raltegravir in
antiretroviral-experienced, integrase-inhibitor-naive adults with HIV: week 48 results from the randomised, double-blind, non-
inferiority SAILING study. Lancet. 2013 Jul 2. [Medline].

127. Nichols G, Mills A, Grossberg R, et al. Antiviral Activity of Dolutegravir in Subjects With Failure on an Integrase Inhibitor–Based
Regimen: Week 24 Phase 3 Results From VIKING-3. Poster presented at: 11th International Congress on Drug Therapy in HIV
Infection. Nov 2012. Poster O232:

128. Ndembi N, Goodall RL, Dunn DT, et al. Viral rebound and emergence of drug resistance in the absence of viral load testing: a
randomized comparison between zidovudine-lamivudine plus Nevirapine and zidovudine-lamivudine plus Abacavir. J Infect Dis.
2010 Jan 1. 201(1):106-13. [Medline].

129. Sax PE, DeJesus E, Mills A, et al. Co-formulated elvitegravir, cobicistat, emtricitabine, and tenofovir versus co-formulated
efavirenz, emtricitabine, and tenofovir for initial treatment of HIV-1 infection: a randomised, double-blind, phase 3 trial, analysis of
results after 48 weeks. Lancet. 2012 Jun 30. 379(9835):2439-48. [Medline].

130. DeJesus E, Rockstroh JK, Henry K, et al. Co-formulated elvitegravir, cobicistat, emtricitabine, and tenofovir disoproxil fumarate
versus ritonavir-boosted atazanavir plus co-formulated emtricitabine and tenofovir disoproxil fumarate for initial treatment of HIV-
1 infection: a randomised, double-blind, phase 3, non-inferiority trial. Lancet. 2012 Jun 30. 379(9835):2429-38. [Medline].

131. McMahon DK, Zheng L, Hitti J, Chan ES, Halvas EK, Hong F, et al. Greater Suppression of Nevirapine Resistance With 21- vs 7-
Day Antiretroviral Regimens After Intrapartum Single-Dose Nevirapine for Prevention of Mother-to-Child Transmission of HIV.
Clin Infect Dis. 2013 Apr. 56(7):1044-51. [Medline]. [Full Text].

132. Spector SA, McKinley GF, Lalezari JP, et al. Oral ganciclovir for the prevention of cytomegalovirus disease in persons with AIDS.
Roche Cooperative Oral Ganciclovir Study Group. N Engl J Med. 1996 Jun 6. 334(23):1491-7. [Medline].

133. Impact of antiretroviral therapy on tuberculosis incidence among HIV-positive patients in high-income countries. Clin Infect Dis.
2012 May. 54(9):1364-72. [Medline].

134. Falutz J, Potvin D, Mamputu JC, et al. Effects of tesamorelin, a growth hormone-releasing factor, in HIV-infected patients with
abdominal fat accumulation: a randomized placebo-controlled trial with a safety extension. J Acquir Immune Defic Syndr. 2010
Mar 1. 53(3):311-22. [Medline].

135. Falutz J, Allas S, Blot K, Potvin D, Kotler D, Somero M, et al. Metabolic effects of a growth hormone-releasing factor in patients
with HIV. N Engl J Med. 2007 Dec 6. 357(23):2359-70. [Medline].

136. Falutz J, Mamputu JC, Potvin D, Moyle G, Soulban G, Loughrey H, et al. Effects of tesamorelin (TH9507), a growth hormone-
releasing factor analog, in human immunodeficiency virus-infected patients with excess abdominal fat: a pooled analysis of two
multicenter, double-blind placebo-controlled phase 3 trials with safety extension data. J Clin Endocrinol Metab. 2010 Sep.

https://emedicine.medscape.com/article/211316-print Page 68 of 71
05/09/2020, 11)59 PM

95(9):4291-304. [Medline].

137. Stanley TL, Falutz J, Marsolais C, et al. Reduction in visceral adiposity is associated with an improved metabolic profile in HIV-
infected patients receiving tesamorelin. Clin Infect Dis. 2012 Jun. 54(11):1642-51. [Medline]. [Full Text].

138. Lingappa JR, Baeten JM, Wald A, Hughes JP, Thomas KK, Mujugira A, et al. Daily acyclovir for HIV-1 disease progression in
people dually infected with HIV-1 and herpes simplex virus type 2: a randomised placebo-controlled trial. Lancet. 2010 Mar 6.
375(9717):824-33. [Medline]. [Full Text].

139. Ruel TD, Boivin MJ, Boal HE, et al. Neurocognitive and motor deficits in HIV-infected Ugandan children with high CD4 cell
counts. Clin Infect Dis. 2012 Apr. 54(7):1001-9. [Medline]. [Full Text].

140. Gaps in HIV Testing and Treatment Hinder Efforts to Stop New Infections. CDC. Available at
https://www.cdc.gov/media/releases/2019/p0315-gaps-hinder-hiv-testing.html. March 18, 2019; Accessed: March 20, 2019.

141. Heffron R, Donnell D, Rees H, Celum C, Mugo N, Were E, et al. Use of hormonal contraceptives and risk of HIV-1 transmission:
a prospective cohort study. Lancet Infect Dis. 2011 Oct 3. [Medline].

142. Marazzi MC, Palombi L, Nielsen-Saines K, et al. Extended antenatal use of triple antiretroviral therapy for prevention of mother-
to-child transmission of HIV-1 correlates with favorable pregnancy outcomes. AIDS. 2011 Aug 24. 25(13):1611-8. [Medline].

143. Burgard M, Jasseron C, Matheron S, et al. Mother-to-child transmission of HIV-2 infection from 1986 to 2007 in the ANRS
French Perinatal Cohort EPF-CO1. Clin Infect Dis. 2010 Oct 1. 51(7):833-43. [Medline].

144. Rerks-Ngarm S, Pitisuttithum P, Nitayaphan S, et al. Vaccination with ALVAC and AIDSVAX to prevent HIV-1 infection in
Thailand. N Engl J Med. 2009 Dec 3. 361(23):2209-20. [Medline].

145. Robb ML, Rerks-Ngarm S, Nitayaphan S, et al. Risk behaviour and time as covariates for efficacy of the HIV vaccine regimen
ALVAC-HIV (vCP1521) and AIDSVAX B/E: a post-hoc analysis of the Thai phase 3 efficacy trial RV 144. Lancet Infect Dis. 2012
Jul. 12(7):531-7. [Medline]. [Full Text].

146. Brooks M. CDC updates HIV preexposure prophylaxis guidelines. Medscape Medical News. May 14, 2014. [Full Text].

147. Preexposure Prophylaxis for the Prevention of HIV Infection in the United States – 2014 Clinical Practice Guideline. Centers for
Disease Control and Prevention. May 2014. Available at http://www.cdc.gov/hiv/pdf/PrEPguidelines2014.pdf.

148. Descovy (emtricitabine/tenofovir AF) [package insert]. Foster City, CA: Gilead Sciences. October, 2019. Available at [Full Text].

149. Grant RM, Lama JR, Anderson PL, et al. Preexposure chemoprophylaxis for HIV prevention in men who have sex with men. N
Engl J Med. 2010 Dec 30. 363(27):2587-99. [Medline]. [Full Text].

150. Cohen MS, Chen YQ, McCauley M, Gamble T, Hosseinipour MC, Kumarasamy N, et al. Prevention of HIV-1 infection with early
antiretroviral therapy. N Engl J Med. 2011 Aug 11. 365(6):493-505. [Medline]. [Full Text].

151. Choopanya K, Martin M, Suntharasam P, Sangkum U, Mock P, Leethochawalit M, et al. Antiretroviral prophylaxis for HIV
infection in injecting drug users in Bangkok, Thailand (the Bangkok Tenofovir Study): a randomized, double-blind, placebo-
controlled phase 3 trial. Lancet. 2013. 2083-90.

152. Leibowitz AA, Parker KB, Rotheram-Borus MJ. A US Policy Perspective on Oral Preexposure Prophylaxis for HIV. Am J Public
Health. 2011 Jun. 101(6):982-5. [Medline].

153. Interim guidance: preexposure prophylaxis for the prevention of HIV infection in men who have sex with men. MMWR Morb
Mortal Wkly Rep. 2011 Jan 28. 60(3):65-8. [Medline].

154. Centers for Disease Control and Prevention. Last updated February 22, 2011. HIV/AIDS - Pre-Exposure Prophylaxis (PrEP).
[Full Text].

155. [Guideline] Marrazzo JM, del Rio C, Holtgrave DR, et al, for the International Antiviral Society-USA Panel. HIV prevention in
clinical care settings: 2014 recommendations of the International Antiviral Society-USA Panel. JAMA. 2014 Jul 23-30.
312(4):390-409. [Medline]. [Full Text].

156. Smith M. Antiviral group: 'Biomedical' tx could slow HIV. MedPage Today. July 21, 2014. [Full Text].

157. [Guideline] Günthard HF, Aberg JA, Eron JJ, for the International Antiviral Society-USA Panel. Antiretroviral treatment of adult

https://emedicine.medscape.com/article/211316-print Page 69 of 71
05/09/2020, 11)59 PM

HIV infection: 2014 recommendations of the International Antiviral Society-USA Panel. JAMA. 2014 Jul 23-30. 312(4):410-25.
[Medline]. [Full Text].

158. [Guideline] DiNenno EA, Prejean J, Irwin K, Delaney KP, Bowles K, Martin T, et al. Recommendations for HIV Screening of Gay,
Bisexual, and Other Men Who Have Sex with Men - United States, 2017. MMWR Morb Mortal Wkly Rep. 2017 Aug 11. 66
(31):830-832. [Medline].

159. EACS. European AIDS Clinical Society Guidelines Version 10.0. European AIDS Clinical Society. Available at
https://www.eacsociety.org/files/2019_guidelines-10.0_final.pdf. November 2019; Accessed: December 2, 2019.

160. [Guideline] US DHHS. Guidelines for the Use of Antiretroviral Agents in Adults and Adolescents with HIV. U.S. Department of
Health and Human Services. Available at https://aidsinfo.nih.gov/guidelines/html/1/adult-and-adolescent-arv/0. December 18,
2019; Accessed: August 20, 2020.

161. Mounzer K, Palella F, Slim J, et al. SPIRIT: Simplifying to rilpivirine/emtricitabine/tenofovir Df single-tablet regimen from boosted
protease inhibitor regimen maintains HIV suppression in the black subgroup [abstract H-656]. Presented at: The 53rd
Interscience Conference onAntimicrobial Agents and Chemotherapy (ICAAC); September 11, 2013; Denver, Colorado. [Full
Text].

162. Kling J. Single-tablet HIV regimen effective. Medscape Medical News. September 19, 2013. [Full Text].

163. Tucker ME. FDA OKs New Triple-Combination Pill (Triumeq) for HIV. Medscape Medical News. Aug 22 2014. [Full Text].

164. Molina JM, Squires K, Sax PE, Cahn P, Lombaard J, DeJesus E, et al. Doravirine versus ritonavir-boosted darunavir in
antiretroviral-naive adults with HIV-1 (DRIVE-FORWARD): 48-week results of a randomised, double-blind, phase 3, non-
inferiority trial. Lancet HIV. 2018 May. 5 (5):e211-e220. [Medline].

165. Orkin C, Squires KE, Molina JM, Sax PE, Wong WW, Sussmann O, et al. Doravirine/Lamivudine/Tenofovir Disoproxil Fumarate
is Non-inferior to Efavirenz/Emtricitabine/Tenofovir Disoproxil Fumarate in Treatment-naive Adults With Human
Immunodeficiency Virus-1 Infection: Week 48 Results of the DRIVE-AHEAD Trial. Clin Infect Dis. 2018 Aug 31. [Medline].

166. Lewis S, Fessel J, Emu B, Schrader S, Kumar P, Richmond GJ, et al. Long-acting ibalizumab in patients with multi-drug resistant
HIV-1: A 24-week study. Presented at the 2017 Conference on Retroviruses and Opportunistic Infections. February 13-16, 2017.
Seattle, WA. Abstracts from the 2017 Conference on Retroviruses and Opportunistic Infections. February 13-16, 2017. Seattle,
Washington. Top Antivir Med. 2017 Apr. 25 (1s):4s-446s. [Medline]. [Full Text].

167. Joint United Nations Programme on HIV/AIDS. Global AIDS Update 2016. Available at
http://www.unaids.org/sites/default/files/media_asset/global-AIDS-update-2016_en.pdf. Accessed: September 6, 2017.

168. [Guideline] Canavan N. New HIV Treatment Guidelines to Cut Millions of Deaths. Medscape Medical News. Jul 1 2013. [Full
Text].

169. [Guideline] World Health Organization. Consolidated Guidelines on the Use of Antiretroviral Drugs for Treating and Preventing
HIV Infection. Jun 2013. [Full Text].

170. [Guideline] Keller DM. New HIV Guidelines Address Broad Range of Medical Conditions. Medscape Medical News. Oct 24 2013.
[Full Text].

171. [Guideline] EACS. European AIDS Clinical Society Guidelines, version 7.0. Oct 2013. [Full Text].

172. [Guideline] Centers for Disease Control and Prevention, Health Resources and Services Administration, National Institutes of
Health, American Academy of HIV Medicine, Association of Nurses in AIDS Care, International Association of Providers of AIDS
Care, et al. Recommendations for HIV Prevention with Adults and Adolescents with HIV in the United States, 2014: Summary for
Clinical Providers. 2014. Available at http://stacks.cdc.gov/view/cdc/26063.

173. Ruiz L, van Lunzen J, Arno A, et al. Protease inhibitor-containing regimens compared with nucleoside analogues alone in the
suppression of persistent HIV-1 replication in lymphoid tissue. AIDS. 1999 Jan 14. 13(1):F1-8. [Medline].

174. Barclay L. Men With HIV Have Higher Risk, Greater Extent of CAD. Medscape Medical News. Available at
http://www.medscape.com/viewarticle/822867. Accessed: April 7, 2014.

175. CDC. Pre-Exposure Prophylaxis (PrEP) for HIV Prevention: Promoting Safe and Effective Use in the United States. CDC.
Available at http://www.cdc.gov/nchhstp/Newsroom/PrEPforHIVFactSheet.html. Accessed: 11/29/2010.

https://emedicine.medscape.com/article/211316-print Page 70 of 71
05/09/2020, 11)59 PM

176. [Guideline] Committee Opinion No 595: Preexposure Prophylaxis for the Prevention of Human Immunodeficiency Virus. Obstet
Gynecol. 2014 May. 123(5):1133-1136. [Medline].

177. Currier JS, Stein JH. HIV and Atherosclerosis: Moving From Associations to Mechanisms and Interventions. Ann Intern Med.
2014 Apr 1. 160(7):509-10. [Medline].

178. Freiberg MS, Chang CC, Kuller LH, Skanderson M, Lowy E, Kraemer KL, et al. HIV Infection and the Risk of Acute Myocardial
Infarction. JAMA Intern Med. 2013 Mar 4. 1-9. [Medline].

179. Gilead. iPrEx Study Results: Public Statement. Gilead. [Full Text].

180. Havlir DV, Bassett R, Levitan D, et al. Prevalence and predictive value of intermittent viremia with combination hiv therapy.
JAMA. 2001 Jul 11. 286(2):171-9. [Medline].

181. Hulskotte EG, Feng HP, Xuan F, van Zutven MG, Treitel MA, Hughes EA, et al. Pharmacokinetic Interactions Between the
Hepatitis C Virus Protease Inhibitor Boceprevir and Ritonavir-Boosted HIV-1 Protease Inhibitors Atazanavir, Darunavir, and
Lopinavir. Clin Infect Dis. 2013 Mar. 56(5):718-26. [Medline].

182. Janeczko JL. HIV Patients Lose Seroprotection Before Vaccine Boosters Are Due. Medscape. Jan 24 2014. [Full Text].

183. Kerneis S, Launay O, Turbelin C, et al. Long-term immune responses to vaccination in HIV-infected patients: a systematic review
and meta-analysis. Clin Infect Dis. 2014 Jan 10. [Medline].

184. Masiá M, Padilla S, Alvarez D, et al. Risk, predictors, and mortality associated with non-AIDS events in newly diagnosed HIV-
infected patients: role of antiretroviral therapy. AIDS. 2013 Jan 14. 27(2):181-9. [Medline].

185. McCormack S, Ramjee G, Kamali A, et al. PRO2000 vaginal gel for prevention of HIV-1 infection (Microbicides Development
Programme 301): a phase 3, randomised, double-blind, parallel-group trial. Lancet. 2010 Oct 16. 376(9749):1329-37. [Medline].
[Full Text].

186. Post WS, Budoff M, Kingsley L, Palella FJ Jr, Witt MD, Li X, et al. Associations Between HIV Infection and Subclinical Coronary
Atherosclerosis. Ann Intern Med. 2014 Apr 1. 160(7):458-67. [Medline].

187. [Guideline] World Health Organization. Scaling up antiretroviral therapy in resource-limited settings: Treatment guidelines for a
public health approach: 2003 revision. World Health Organization, Geneva 2004. Available at
http://www.who.int/hiv/pub/prev_care/en/arvrevision2003en.pdf.

188. EACS Guidelines version 9.0. EACS European AIDS Clinical Society. Available at http://www.eacsociety.org/files/guidelines_9.0-
english.pdf. October 2017;

189. Djawe K, Buchacz K, Hsu L, Chen MJ, Selik RM, Rose C, et al. Mortality Risk After AIDS-Defining Opportunistic Illness Among
HIV-Infected Persons--San Francisco, 1981-2012. J Infect Dis. 2015 Nov 1. 212 (9):1366-75. [Medline].

https://emedicine.medscape.com/article/211316-print Page 71 of 71

You might also like