701
Views
12
CrossRef citations to date
0
Altmetric
Reviews

Past, present and future molecular diagnosis and characterization of human immunodeficiency virus infections

&
Pages 1-8 | Received 02 Mar 2012, Accepted 21 May 2012, Published online: 25 Jan 2019

Abstract

Substantive and significant advances have been made in the last two decades in the characterization of human immunodeficiency virus (HIV) infections using molecular techniques. These advances include the use of real-time measurements, isothermal amplification, the inclusion of internal quality assurance protocols, device miniaturization and the automation of specimen processing. The result has been a significant increase in the availability of results to a high level of accuracy and quality. Molecular assays are currently widely used for diagnostics, antiretroviral monitoring and drug resistance characterization in developed countries. Simple and cost-effective point-of-care versions are also being vigorously developed with the eventual goal of providing timely healthcare services to patients residing in remote areas and those in resource-constrained countries. In this review, we discuss the evolution of these molecular technologies, not only in the context of the virus, but also in the context of tests focused on human genomics and transcriptomics.

Keywords:

Introduction

Laboratory and clinical microbiologists perform various tests to determine the human immunodeficiency virus (HIV) infection status of a patient, evaluate the progression of disease, and monitor the effectiveness of antiretroviral therapy (ART). HIV infection can be diagnosed in five possible ways: (i) direct visualization of virions by electron microscopy; (ii) cultivation by lymphocyte culture; (iii) measurement of HIV-specific serologic responses; (iv) detection of viral antigens; and (v) detection of viral nucleic acids.Citation1,Citation2 Diagnosis can best be achieved by serologic testing but the monitoring of HIV disease progression is mostly accomplished by the quantitation of CD4 T cells and viral RNA. The detection of HIV nucleic acids in clinical specimens was first demonstrated in 1988 by amplifying HIV-1 genomic DNA from the peripheral blood mononuclear cells of HIV-1-infected individuals.Citation3 Molecular methods have since been drastically improved and, along with serologic assays, have become a routine means to characterize HIV-1 infection and disease progression.

The acute HIV infection phase, or the window period, represents the first few days after the entry of HIV into the CD4 cells of a new host. During this phase, the host has yet to build up an immune response and thus HIV viruses rapidly replicate. The number of viral RNA molecules can increase to several million copies per ml of plasma and thus can be readily detected by molecular assays, shortening the window period from 20–30 days to 10–15 days.Citation4 The viral p24 antigens encapsulating the viral RNA can also be detected but with less sensitivity. With the use of fourth generation enzyme-linked immunosorbent assays (ELISA) that detect p24 antigen and IgG and IgM antibodies, the window period between HIV entry and serologic positivity has been narrowed from approximately 1 month or more to 15–20 days.Citation5,Citation6

A CD4 T-cell count is used as the principal ART eligibility criterion for infected patients in most countries, whereas plasma viral load assays are used to monitor ART efficacy. Qualitative molecular assays have also been used to detect HIV vertical transmission from infected pregnant mothers to infants. HIV-1 drug resistance tests, which include genotyping assays for detecting nucleotide mutations in viral genomes, and phenotype assays for detecting viral replication in the presence of antiretroviral drugs, have become routine in the management of patients receiving ART in most developed countries. Furthermore, an assessment of host gene polymorphisms and immune responses has developed in the clinic for the better assessment and monitoring of HIV-1 infections. To facilitate the linkage of testing and subsequent patient care and treatment and in order to prevent loss of follow-up, molecular testing that can be used at point-of-care (POC) locations has been under vigorous development and may find utility in both diagnosis and ART monitoring in the near future (). This review summarizes the development and the potential utility of all molecular assays currently used to diagnose and characterize HIV infections.

Table 1 Current molecular methods for laboratory diagnosis and characterization of HIV infections

HIV qualitative nucleic acid assays

The qualitative detection of HIV nucleic acids is used in three major areas: the identification of acute infection, assurance of blood safety, and in early infant diagnosis. A revised testing algorithm employing qualitative nucleic acid testing was recently proposed to address the shortcomings of western blot analysis to confirm HIV infection.Citation7 The APTIMA HIV-1 RNA Qualitative Assay and the Procleix HIV-1/HCV assay (Gen-Probe, San Diego, CA, USA) are both Food and Drug Administration (FDA)-approved for blood-donor screening to exclude blood from donors with acute HIV infection.Citation8 These two assays use transcription-mediated amplification technology.Citation9 The persistence of maternal antibodies directed against HIV in exposed infants up to 18 months of age prevents the use of antibody-based assays for the early diagnosis of HIV infection. Infected infants have a high morbidity and mortality in the first 2 years of life; thus, it is important to promptly establish the infection status of the exposed infant in order to employ appropriate ART sufficiently early.Citation10,Citation11 Qualitative nucleic acid assays for the detection of HIV pro-viral DNA,Citation12 viral RNACitation13,Citation14 and total nucleic acidsCitation13,Citation15 have become the methods of choice for diagnosis in infants born to HIV-1-infected mothers.Citation12,Citation16

HIV RNA viral load assays

HIV-1 infection results in lifelong viral persistence. In chronically infected patients, the HIV RNA viral load in the plasma together with CD4 T-cell numbers are the two routine laboratory markers used to guide ART initiation, monitor treatment effectiveness, determine clinical progressionCitation17,Citation18 and determine treatment regimens.Citation19 An HIV RNA level below the detection limit is indicative of excellent drug intake adherence by patients and ART efficacy.Citation17 HIV-1 viral load determination is typically performed with HIV RNA amplification by reverse transcription-polymerase chain reaction (RT-PCR), nucleic acid sequence-based amplification (NASBA) or branched chain DNA tests.Citation20 Typically, viral load falls by at least 10-fold 1 month post-ART, and below the detection limit by 4–6 months post-ART, usually to less than 50–75 copies/ml.Citation19

An ideal viral load assay should have a broad dynamic detection range and be able to quantitate HIV-1 Group M subtypes A–G and N and O with equal efficiency. While HIV-1 subtype B predominates in Western countries, non-B subtypes predominate in resource-poor parts of the world.Citation21 The ability of a test to detect a broad range of these genetically diverse viruses with comparable sensitivity is therefore important to global HIV patient care. In the United States, five commercial plasma RNA assays from four companies are FDA-approved for HIV-1 monitoring in infected patients. Although these assays are not licensed for use in HIV screening or for confirmatory HIV tests, they have often been used in this context.Citation13,Citation20,Citation22

Plasma is the main specimen for HIV-1 viral load testing. Non-plasma specimens, such as peripheral blood mononuclear cells, saliva, cerebrospinal fluid, seminal plasma, dried plasma and dried blood spots (DBS), can also be readily tested.Citation16,Citation23,Citation24 Plasma from whole blood collected by plasma preparation tubes should be separated from red blood cells within 6 h and transferred to a new vessel prior to freezing and transportation.Citation25,Citation26 Compared with HIV DNA, viral RNA is relatively unstable, thus, it is recommended that plasma is quickly separated and stored in a −20 °C or −70 °C freezer prior to testing. Recently, DBS specimens have drawn increased attention because of their usefulness in remote rural areasCitation27 where there is difficulty in plasma transportation and also concerns as to biosafety. HIV when present in dried blood is not considered infectious. When protected from RNA degradation in conditions of high humidity, DBS can serve as a viable approach for preservation and transportation. DBS-based viral load assays have been developed for several main diagnostic platforms including the Roche Cobas, Abbott m2000 and bioMerieux NucliSens platforms.Citation16,Citation23

The Cobas Amplicor HIV-1 Monitor Assay (Roche Diagnostics, Indianapolis, IN, USA) is based on back-to-back RT and PCR.Citation28,Citation29 There are two basic assay platforms: (i) the Amplicor HIV-1 Monitor assay, a manual test performed in 96-microwell plates; and (ii) the Cobas AmpliPrep/Cobas TaqMan HIV-1 assay with full automation of nucleic acid preparation and real-time RT-PCR amplification. The latter was the first FDA-approved HIV viral load test in 1996 with a low detection limit of 400 RNA copies/ml using 140 µl of plasma. Amplicor Ultra-Sensitive test is a variation using 1 ml of plasma in order to improve the detection limit to 50 copies/ml. The new fully automated version of the COBAS AmpliPrep/COBAS TaqMan HIV-1 system (version 1.0) has a detection limit that ranges from 50 to 1 000 000 copies/ml.Citation30 The version 1.0 system targets the HIV-1 gag gene and can quantify all group M and N viruses and many circulating recombinant forms.Citation20,Citation30,Citation31 The newer 2.0 version test targets the gag and conserved LTR regions of the viral genome and extends subtype coverage to Group O.Citation20,Citation32,Citation33

The branched chain DNA-based VERSANT HIV-1 RNA 3.0 Assay (Siemens Healthcare Diagnostics, Tarrytown, NY, USA) incorporates a unique signal amplification technology and provides good reproducibility at the lower end of the detection range.Citation20,Citation28,Citation31 This assay is also less affected by the presence of inhibitory substances and product carryover contamination problems associated other methods. The disadvantages of branched chain DNA technique include the requirement for larger plasma volumes.Citation20,Citation31

The NucliSens HIV-1 RNA QT assay (bioMerieux, Inc., Durham, NC, USA) incorporates three key technologies: (i) silica-based nucleic acid extraction; (ii) NASBA; and (iii) electrochemiluminescence detection.Citation29 The NASBA technology is a sensitive and rapid amplification method that does not require a thermocycler and heat-stable enzymes. This assay can be used for measuring viral loads in other body fluids, because the RNA extraction procedure consistently generates RNA products that are free of interfering substances.Citation28,Citation30 The current second generation assay is reported to quantify subtypes A and G less reliably than other subtypes.Citation20,Citation31

The real-time HIV-1 assay in the m2000 system (Abbott Molecular, Des Plaines, IL, USA) consists of two components: the m2000sp component that allows automatic nucleic acid extraction and loading of sample and reagent, and the m2000rt component that performs nucleic acid amplification and detectionCitation30,Citation31 using a single partially double-stranded linear DNA probe.Citation34 This assay offers a broad dynamic detection range and improved sequence variation tolerance. The manual operation is kept to a minimum and the amplification and detection steps are both carried out in a closed system in order to greatly reduce the risk of carryover contamination. The system can detect a wide coverage of HIV-1 including groups M, circulating recombinant forms, and groups N and O viruses.Citation20,Citation31

Performance may vary significantly between HIV-1 viral load assay platforms depending on the HIV-1 subtypes present in the sample.Citation35 A Canadian study compared the RealTime HIV-1 m2000rt, COBAS TaqMan 48 v1.0, Easy Q v1.2 and Versant v3.0 assays for the determination of HIV-1 viral loads in a cohort of patients with diverse HIV subtype infections.Citation20 Discordant viral load results were obtained in 26.4% of samples due to missing values, false negatives, and a significant under-quantification, the extent of which varied according to HIV-1 subtype. False-negative results occurred for all platforms including 2.0% for m2000rt, 2.5% for COBAS, 6.0% for Easy Q and 2.0% for Versant. Easy Q and Versant had the highest rates of under-quantification by ≥1.0 log(10) copies/ml, mainly for HIV non-B subtypes.Citation20 HIV viral diversity in the population being tested must be considered in selecting the most appropriate detection platform and, once selected, the platform must be used consistently.Citation32 The development of quantitative HIV-1 viral load tests with increasing sensitivity has recently been reviewed.Citation36 In contrast, the development of HIV-2 specific tests has not been accomplished. The small number of HIV-2 cases restricted to West Africa provides little incentive for commercial manufacturers to develop assays specific for HIV-2.

HIV-1 genotypic drug resistance testing

The ultimate goal of highly active antiretroviral therapy is to completely suppress viral replication, the emergence of drug resistant virus mutants and clinical progression.Citation17,Citation37 Antiretrovial drug resistance is defined as the ability of HIV-1 to multiply in the presence of antiretroviral drugs. In the clinical setting, if a viral load fails to fall adequately, or if it rebounds to 1000 copies/ml or higher, tests for drug resistance are recommended.Citation38 Two types of laboratory methods are available to determine drug resistance. Phenotypic drug resistance method directly measures viral replication capability in a recombinant viral form in the presence of antiretroviral drugs, whereas genotypic drug resistance methods detect viral genome mutations known to confer a decreased drug sensitivity. Both methods are commercially available. Phenotypic assay is much more expensive and is usually reserved for patients with prior viral resistance. The clinical utility of HIV drug resistance testing has been evaluated in a number of prospective randomized clinical trials.Citation39,Citation40,Citation41

Genotypic drug resistance testing has been recommended by the International AIDS Society-USA panel as an important tool to guide therapy changes, the overall direction of therapy, and more recently, the initiation of therapy.Citation39,Citation42,Citation43 There are two currently FDA approved commercial drug resistance assays: (i) the TruGene HIV-1 Genotyping Kit and OpenGene DNA Sequencing System (Siemens Healthcare Diagnostics, Tarrytown, NY, USA); and (ii) the ViroSeq HIV-1 Genotyping System (Abbott Molecular). Both systems generate reverse transcriptase and protease gene sequences following RT-PCR amplification. One major difference between the two systems is the sequencing chemistry. ViroSeq uses a four-dye termination system, while the TrueGene uses a dye primer system. In addition, six sequencing reactions are needed to analyze one patient for ViroSeq, whereas 12 are needed for TruGene. The ViroSeq system requires an additional purification step to remove dye terminators.Citation44 While the TrueGene system is limited in throughput, it provides an interpretative report containing information as to the drug resistance mutations present in the sample.Citation45 Both systems work well for the HIV-1 B subtype circulating in North America. One recent study used the ViroSeq HIV-1 genotyping system to determine drug resistance on a panel of diverse HIV-1 group M isolates circulating in Cameroon.Citation46 The data and others performed on user-developed assays revealed that the performance of genotyping assays can be greatly influenced by the sequence variations of non-B HIV-1 strains that predominate in Africa.Citation46,Citation47

The two above-mentioned assays are mostly useful for B-subtype viruses. They detect mutations in the reverse transcriptase and protease genes. They do not detect mutations associated with fusion, integrase and C-C chemokin receptor type 5 (CCR5) inhibitors. Furthermore, the proportion of drug resistance variants have to be at least 25% of the patient's total viral population in the patient.Citation42 Mutant viruses existing at a level lower than 25% would escape detection. These viruses can lead to subsequent treatment failure.Citation48 Two other genotyping formats, the HIV PRT GeneChip assay (Affymetrix, Santa Clara, CA, USA) and the HIV-1 RT Line Probe Assay (Innogenetics, Ghent, Belgium), have shown to detect rapidly any drug resistance. Allele-specific PCR,Citation49,Citation50 single-genome sequencingCitation48,Citation49,Citation51 and next generation sequencing (NGS)Citation49,Citation52,Citation53 have also been reported to increase the detection sensitivity for minor mutants. However, due to the innate error rate of reaction enzymes, it has been reported that NGS had limited sensitivity to about 0.5% and therefore may have less utility among those treatment-experienced patients with a persistent viremia. The majority of low-frequency drug resistance-related mutations detected using NGS are most likely due to errors inherent in the NGS method or as a consequence of error-prone HIV-1 replication,Citation54 thus clinicians prescribing ART should interpret such findings with considerable caution.

HIV phenotypic antiretroviral susceptibility testing

Phenotype assays measure the ability of HIV-1 to grow in the presence of various concentrations of antiretroviral agents. Phenotyping uses clinical cutoffs associated with the treatment outcome data and estimates the net effect of multiple mutations directly.Citation55 Phenotype assays are considered as molecular methods, since these assays require the generation of recombinant viruses using sophisticated molecular techniques. HIV-1 RNA from a patient's plasma is extracted and protease and reverse transcriptase genes are amplified by RT-PCR. The amplified gene fragments are then inserted into HIV-1 vectors in order to generate a replication competent recombinant. The replication of the new resultant HIV-1 recombinant is measured by a reporter gene system.Citation56,Citation57 Data from these assays are expressed as either 50% or 90% inhibitory concentrations (IC50 or IC90), values familiar to clinicians and thus easily understood. Due to the fact that the test is labor-intensive and technically complicated, the testing turn-around time can be as long as 6 weeks. As with genotype testing, phenotype assays can only detect mutant variants that comprise more than 25% of the viral population in any individual patient. Currently, two HIV-1 phenotyping assays are available: the AntiVirogram assay at VIRCO Lab (Bridgewater, NJ, USA)Citation56 and the PhenoSense HIV assay at MonoGram Biosciences (South San Francisco, CA, USA).Citation57 More data are needed to demonstrate the clinical advantage of phenotyping over genotyping.Citation58 A virtual phenotyping procedure has also been described that provides an estimation of the phenotype by averaging viruses with similar genotypes.Citation59

As integrase and entry/fusion inhibitors become available for ART, HIV-1 genotyping assays have been extended to cover drug resistance mutations related to these inhibitors.Citation60 Since inhibition by Enfuvirtide, an entry/fusion inhibitor, blocks HIV-1 entry to an immune cell, genotyping alone cannot provide sufficient information as to resistance. The PhenoSense Entry assay (MonoGram) has been developed to assess resistance to entry inhibitors.Citation61 Maraviroc, a CCR5 inhibitor drug, is only effective against viruses that use CCR5 as a co-receptor for entry.Citation62,Citation63 Prior to Maraviroc administration, a tropism assay must be performed to determine whether the virus is CCR5-tropic.Citation64,Citation65 Two commercial tropism assays are available at the Monogram Biosciences and Pathway DiagnosticsCitation63,Citation66 ().

POC testing

POC testing can be performed at home, in the workplace, at pharmacies and physician's offices, outpatient clinics, emergency rooms, and at the bedside of patients. The need of POC testing has been driven by the clinical need to make a rapid, evidence-based, decision as to the most appropriate therapeutic regimen at or near the site of patient care.Citation67 This form of diagnosis has also been available for several years for other infectious diseases such as group A Streptococcus detection in throat swab. Numerous products are commercially available for POC diagnosis of viral, bacterial and parasitic infections.Citation68 Considerable efforts have been spent in developing POC devices including HIV antibody, nucleic acid detection, CD4 T-cell quantitation and viral load for the use in resource-limited settings in order to facilitate patient identification and delivery of care.Citation69

Currently, there are no FDA-approved molecular assays that can be used in a POC setting. Some simple nucleic acid detection devices have the potential for HIV nucleic acid amplification and detection. A SAMBA system (Diagnostics for the Real World, Cambridge, UK) performs HIV detection through an isothermal nucleic acid amplification method in an integrated cartridge combined with a small benchtop instrument.Citation70 A helicase-dependent amplification was developed to separate DNA strands at 37 °C rather than the typical 95–97 °C used in PCR.Citation71,Citation72 This modification greatly simplifies the enzymology involved in the amplification process whilst retaining the advantage common to all isothermal amplification techniques. Another helicase-dependent amplification-based IsoAmp HIV-1 assay (BioHelix Corp., Beverly, MA, USA) has been developed using a small containment device. It targets the HIV-1 gag for amplification and uses an embedded vertical-flow DNA detection strip to detect amplicons.Citation73 Similar to the serologic rapid tests, this vertical-flow DNA detection strip has an internal control line to validate the proper performance of the reactions and has a test line to detect the amplicons. The preliminary limit of detection of the IsoAmp HIV assay is 50 copies of the HIV-1 Armored RNA (Assuragen, Austin, TX, USA) introduced into the IsoAmp HIV reaction.Citation74

Molecular diagnostic methods can be subdivided into the three steps nucleic acid extraction, amplification and detection components.Citation75 A rapid POC extraction of HIV-1 proviral DNA from whole blood and its detection using real-time PCR was recently reported.Citation76 Simple and inexpensive molecular assays based on dipstick and zipper technology have also been described.Citation77,Citation78 The Cepheid GeneXpert System (Sunnyvale, CA, USA) is a single-use sample processing cartridge system with an integrated multicolor real-time PCR capacity.Citation79 Microarrays have also been incorporated with nucleic acid probes and peptides to detect and quantify HIV-1.Citation80,Citation81 Miniaturized PCR devices have been reported for microbial agent detection and identification. Integration of microfluidics and lensless imaging for POC testing has also been reported.Citation82 With the incorporation of micro/nano fabrications/crystals (e.g., quantum dots), microfluidics and array based systems, the development of more feasible immunological and molecular tests for HIV POC testing in resource-limited settings is expected.Citation83

There is a crucial need for low-cost, simple and accurate HIV-1 viral load monitoring technologies in resource-limited settings, particularly when first- and second-line treatment regimens are scaled up.Citation84,Citation85,Citation86 The Liat HIV Quant Assay (IQuum, Marlborough, MA, USA) is comprised of two components, the Liat Analyzer and the Liat Tube. This method can yield automated sample-to-result in a near-patient setting within 1.5 h. It can detect HIV-1 group M (subtypes A–H), group O, and HIV-2 with a detection linearity of 6 logs and a limit of detection of 57 copies/ml.Citation87 In contrast to the use of nucleic acid amplification, the ExaVir Load assay (Cavidi, Sweden) measures viral reverse transcriptase activity using simple equipment and a modified ELISA procedure. This method can be performed in a relatively simple laboratory environment. The measured viral reverse transcriptase activity correlates with plasma RNA levels.Citation88,Citation89,Citation90 In addition to the simple ExaVir Load start-up equipment, only that ELISA equipment which commonly found in an HIV laboratory is required for performing the tests. The integrated equipment used in the current Version 3 makes the procedure less time-consuming, more efficient and even easier to handle. This viral reverse transcriptase measurement system has been extended to HIV phenotypic susceptibility testing. This new ExaVir drug assay has been reported to work well for efavirenz but not for nevirapine.Citation90

Host genome polymorphism testing

Enhanced by the success of human genome programs, diagnostic virologists envisioned the utilization of genetics beyond HIV-1 genomes to help manage HIV infections.Citation91,Citation92 Chronic and persistent HIV-1 infections can be viewed as ‘horizontally acquired’ genetic diseases. An HIV pathogen and its host can be viewed and treated as an integrated biological system.Citation92 There is ample evidence to indicate that the outcome of HIV infections is greatly influenced by the host's genetic background of the host.Citation92,Citation93 Polymorphisms in AIDS restriction genes, e.g., CCR5, CCR2 and C-X-C chemokine receptor type 4, influence the risk of HIV infection and the rate of AIDS progression. Pine et al.Citation94 revealed that polymorphisms in Toll-like receptors 4 and 9 influenced the level of viral load in a sero-incident cohort of HIV-1-infected individuals. A recent study of high-risk South African black women indicated that the risk of acquiring HIV infection was 3-fold greater in those with the trait of Duffy-null-associated low neutrophil counts when compared with all other study participants.Citation95 Because of the high prevalence of this trait among persons of African ancestry, it may contribute to the dynamics of the HIV epidemic in Africa.Citation95

Hence, the detection of host polymorphisms in the HIV diagnostic field can help identify those who are at risk of rapid disease progression and thus the need to initiate early treatment. Allele frequencies and relative hazard values of CCR5-Δ32, CCR2 64I, CCR5 P1, interleukin (IL)-10 5'A, human leukocyte antigen (HLA)-B*35 and HLA homozygosity were determined to generate a composite relative hazard of progression to AIDS.Citation96 Possession of a chemokine (C-C motif) ligand 3-like 1 (CCL3L1) copy number lower than the population average is associated with markedly enhanced HIV/AIDS susceptibility; which is enhanced in individuals with the CCR5-Δ32 genotype.Citation97 Retaining CCR5-Δ32, CCR2 64I, CCR5 59029AA, CCL3 495TT, stromal cell-derived factor-1 (SDF1) 3'A, progressive multifocal leukoencephalopathy (PML) –225TT, peptidylprolyl isomerase A (PPIA) 1650G, and tumor susceptibility gene 101 (TSG101) –183C genetic variants may translate into lengthening or shortening the time from 500 CD4 T cells/µl to <200 CD4 T cells/µl by up to 2.8 years.Citation98 Genetic polymorphic profiles in cytochrome P450s and transporters facilitate optimal chemotherapy for HIV infections. An association between cytochrome P450 2D6 (CYP2D6) genetic variants and plasma levels of efavirenz and nevirapine in treatment-naive individuals with HIV infection was observed. Patients carrying a loss-of-function CYP2D6 allele had higher median plasma levels of both drugs compared to those without the mutation.Citation99 Polymorphisms in CYP2B6 correlate with high efavirenz concentrations in the plasma and the central nervous system.Citation100,Citation101 A recent study identified more than 300 genome-wide significant single-nucleotide polymorphisms within the major histocompatibility complex, and the data implied that HLA–viral peptide interaction was a major factor in the modulation of HIV infections.Citation102

Microarray has become a powerful technique to screen genes for multiple polymorphisms on hundreds of samples.Citation81 With recent technological advances, it is now possible to genotype over one million polymorphisms at high throughput by using Illumina or Affymetrix system.Citation103,Citation104 While increasing HIV resistance and disease progression-related host gene polymorphisms, simple and user-friendly techniques for the detection of such known mutations will soon be adapted and correctly interpreted by those responsible for a diagnosis. Currently available techniques include allele-specific nucleotide amplification,Citation105,Citation106 single nucleotide primer extension,Citation107 and oligonucleotide ligation assay.Citation108,Citation109 Real-time PCR assays based on TaqMan hydrolysis probes play an important role as a pathogen sequence-specific probe to confirm the authenticity of the detection target. These methods are very robust but less cost-effective for large-scaled studies.Citation110,Citation111 DNA sequencing remains the gold standard and is enhanced by recent high-throughput processing and deep production scaling.Citation112 It is now considered the most powerful procedure for polymorphism detection.

Host response and transcriptome analysis

In addition to measuring the levels of CD4 T cells, other host responses can be used for monitoring therapy efficacies and side effects in HIV-infected patients receiving ART. T-cell-receptor-chain rearrangement excision circles (TREC) are episomal double-stranded DNA circles, which are generated during T-cell maturation in the thymus.Citation113 TREC are stable and persist in newly matured T cells but are replication-deficient. After the entry of TREC-bearing T cells into the peripheral blood circulation, with time TREC molecules are progressively diluted out during T-cell mitosis. Quantification of TREC present in naive T cells is thus a measure of thymic function.Citation114 Although thymic function declines with age, substantial output is still maintained into late adulthood. HIV infection leads to a decrease in thymic function that can be measured in the peripheral blood and lymphoid tissues. In most treated adults, there is a rapid and sustained increase in thymic output, indicating that the adult thymus can contribute to immune reconstitution following ART.Citation114,Citation115 Thus, in addition to CD4 and viral load, TREC has been considered as another biomarker to monitor treatment efficacy, as well as to evaluate clinical disease progression.Citation115,Citation116

The mitochondrial toxicity of antiretroviral drugs, particularly that of the nucleoside reverse transcriptase inhibitor, has been postulated to be responsible for the pathogenesis of many secondary treatment side effects, including hyperlactatemia.Citation117,Citation118 During ART, clinically symptomatic mitochondrial dysfunction has been associated with mitochondrial DNA depletion. A real-time PCR was developed to determine a mitochondrial DNA versus nuclear DNA ratio as a biomarker of nucleoside reverse transcriptase inhibitor toxicity.Citation119 The observed increase in mitochondrial DNA and RNA contents during the first year of treatment may represent a restorative trend resulting from suppression of HIV-1 infection, independent of the treatment used. The mitochondrial DNA and RNA contents in individual cell subtypes, rather than in peripheral leukocytes, may be better markers of toxicity and worthy of further investigation.Citation120 Other assays, which include mitochondrial RNA quantification by real-time PCRCitation121 and mitochondrial protein synthesis by western blot immunoanalysis,Citation122 have been described to measure mitochondrial toxicity-related functional changes. Recently, a flow cytometric assay was developed to gauge mitochondrial function. Flow cytometry of a mitochondrial DNA-encoded mitochondrial protein together with a nuclear DNA-encoded mitochondrial protein was optimized and validated, allowing for the simultaneous detection of mitochondrial DNA and nuclear DNA encoded proteins at the single-cell level; this approach offers a method whereby mitochondrial function can be monitored.Citation123

New microarray techniques including those from Affymetrix (Santa Clara, CA, USA), have recently allowed host transcriptome analyses in individuals infected with HIV-1.Citation81,Citation124 A comprehensive review of the 34 studies involving HIV-1 and microarrays from 2000 to 2006 concluded that these studies yielded important data on HIV-1-mediated effects on gene expression and provided new insights into the intricate interactions occurring during infection.Citation125 Several recent studies have demonstrated progress in expanding the pool of target genes and understanding the functional correlates of gene modulation to HIV-1 pathogenesis in vivo.Citation91,Citation126 It is predicted that such host transcriptome profiles will be used for the assessment of disease progression and prognosis. The precision of transcriptome analyses will be greatly improved through the added resolution of the RNA-Seq approach using deep-sequencing technologies for transcriptome profiling.Citation127,Citation128

Concluding remarks

The development and application of molecular techniques has initiated a revolution in the diagnosis and monitoring of many infectious diseases. Molecular techniques have quickly become the mainstay for laboratory diagnosis and assessment of HIV-1 infections. Qualitative molecular assays have been used as the test of choice to diagnose perinatal and acute HIV-1 infections. HIV-1 plasma viral load assays are routinely used in combination with CD4 T-cell counts in order to determine when to initiate therapy and when a treatment regimen is failing. HIV-1 antiretroviral susceptibility tests, which include a genotyping assay to detect mutations known to confer resistance together with phenotyping measuring recombinant viral replication in the presence of antiretroviral drugs, have both become routine in the management of HIV patients undergoing antiretroviral treatment. POC or near-the-patient molecular assays are being developed with the potential to make rapid, evidence-based therapeutic intervention at or near the site of patient care. The initiation of alternative tools for improved diagnosis and the monitoring of HIV infections have led to a better understanding of the association between host gene polymorphism and viral immune responses.

Notes

Abbreviation: ART, antiretroviral therapy; HARRT, highly active antiretroviral therapy; HIV, human immunodeficiency virus; POC, point of care.

A, molecular assay is widely used for clinical diagnosis and/or monitoring of HIV-1 infections; B, molecular assay is useful under certain circumstances or for the diagnosis of particular populations; C, molecular assay is seldom useful for general diagnostic purposes but may be useful in specific researches and trials; D, molecular assay is not available or not used for laboratory diagnosis and monitoring.

  • Branson BM.State of the art for diagnosis of HIV infection. Clin Infect Dis2007;45( Suppl 4): S221–S225.
  • Tang YW, Persing DH.Diagnostic microbiology. In: Schaechter M (ed.) Encyclopedia of microbiology. 3rd ed. Oxford: Elsevier Press, 2009: 308–320.
  • Ou CY, Kwok S, Mitchell SW et al.DNA amplification for direct detection of HIV-1 in DNA of peripheral blood mononuclear cells. Science1988;239( 4837): 295–297.
  • Branson BM, Stekler JD.Detection of acute HIV infection: we can't close the window. J Infect Dis2012;205: 521–524.
  • Bentsen C, McLaughlin L, Mitchell E et al.Performance evaluation of the Bio-Rad Laboratories GS HIV Combo Ag/Ab EIA, a 4th generation HIV assay for the simultaneous detection of HIV p24 antigen and antibodies to HIV-1 (groups M and O) and HIV-2 in human serum or plasma. J Clin Virol2011;52( Suppl 1): S57–S61.
  • Chavez P, Wesolowski L, Patel P, Delaney K, Owen SM.Evaluation of the performance of the Abbott ARCHITECT HIV Ag/Ab Combo Assay. J Clin Virol2011;52( Suppl 1): S51–S55.
  • Branson BM.The future of HIV testing. J Acquir Immune Defic Syndr2010;55( Suppl 2): S102–S105.
  • Fiebig EW, Wright DJ, Rawal BD et al.Dynamics of HIV viremia and antibody seroconversion in plasma donors: implications for diagnosis and staging of primary HIV infection. AIDS2003;17: 1871–1879.
  • Giachetti C, Linnen JM, Kolk DP et al.Highly sensitive multiplex assay for detection of human immunodeficiency virus type 1 and hepatitis C virus RNA. J Clin Microbiol2002;40: 2408–2419.
  • Resino S, Resino R, Maria Bellon J et al.Clinical outcomes improve with highly active antiretroviral therapy in vertically HIV type-1-infected children. Clin Infect Dis2006;43: 243–252.
  • van Rossum AM, Fraaij PL, de Groot R.Efficacy of highly active antiretroviral therapy in HIV-1 infected children. Lancet Infect Dis2002;2: 93–102.
  • Sherman GG, Cooper PA, Coovadia AH et al.Polymerase chain reaction for diagnosis of human immunodeficiency virus infection in infancy in low resource settings. Pediatr Infect Dis J2005;24: 993–997.
  • Rouet F, Montcho C, Rouzioux C et al.Early diagnosis of paediatric HIV-1 infection among African breast-fed children using a quantitative plasma HIV RNA assay. AIDS2001;15: 1849–1856.
  • Stevens WS, Noble L, Berrie L, Sarang S, Scott LE.Ultra-high-throughput, automated nucleic acid detection of human immunodeficiency virus (HIV) for infant infection diagnosis using the Gen-Probe Aptima HIV-1 screening assay. J Clin Microbiol2009;47: 2465–2469.
  • Owen SM, Yang C, Spira T et al.Alternative algorithms for human immunodeficiency virus infection diagnosis using tests that are licensed in the United States. J Clin Microbiol2008;46: 1588–1595.
  • Okonji JA, Basavaraju SV, Mwangi J et al.Comparison of HIV-1 detection in plasma specimens and dried blood spots using the Roche COBAS Ampliscreen HIV-1 test in Kisumu, Kenya. J Virol Methods2012;179: 21–25.
  • Ford N, Nachega JB, Engel ME, Mills EJ.Directly observed antiretroviral therapy: a systematic review and meta-analysis of randomised clinical trials. Lancet2009;374: 2064–2071.
  • Mellors JW, Munoz A, Giorgi JV et al.Plasma viral load and CD4+ lymphocytes as prognostic markers of HIV-1 infection. Ann Intern Med1997;126: 946–954.
  • Kitchen CM, Kitchen SG, Dubin JA, Gottlieb MS.Initial virological and immunologic response to highly active antiretroviral therapy predicts long-term clinical outcome. Clin Infect Dis2001;33: 466–472.
  • Church D, Gregson D, Lloyd T et al.Comparison of the RealTime HIV-1, COBAS TaqMan 48 v1.0, Easy Q v1.2, and Versant v3.0 assays for determination of HIV-1 viral loads in a cohort of Canadian patients with diverse HIV subtype infections. J Clin Microbiol2011;49: 118–124.
  • Barth RE, van der Loeff MF, Schuurman R, Hoepelman AI, Wensing AM.Virological follow-up of adult patients in antiretroviral treatment programmes in sub-Saharan Africa: a systematic review. Lancet Infect Dis2010;10: 155–166.
  • Stevens W, Erasmus L, Moloi M, Taleng T, Sarang S.Performance of a novel human immunodeficiency virus (HIV) type 1 total nucleic acid-based real-time PCR assay using whole blood and dried blood spots for diagnosis of HIV in infants. J Clin Microbiol2008;46: 3941–3945.
  • Garrido C, Zahonero N, Corral A, Arredondo M, Soriano V, de Mendoza C.Correlation between human immunodeficiency virus type 1 (HIV-1) RNA measurements obtained with dried blood spots and those obtained with plasma by use of Nuclisens EasyQ HIV-1 and Abbott RealTime HIV load tests. J Clin Microbiol2009;47: 1031–1036.
  • Tang YW, Huong JT, Lloyd RM Jr, Spearman P, Haas DW.Comparison of human immunodeficiency virus type 1 RNA sequence heterogeneity in cerebrospinal fluid and plasma. J Clin Microbiol2000;38: 4637–4639.
  • Rebeiro PF, Kheshti A, Bebawy SS et al.Increased detectability of plasma HIV-1 RNA after introduction of a new assay and altered specimen-processing procedures. Clin Infect Dis2008;47: 1354–1357.
  • Salimnia H, Moore EC, Crane LR, Macarthur RD, Fairfax MR.Discordance between viral loads determined by Roche COBAS AMPLICOR human immunodeficiency virus type 1 monitor (version 1.5). Standard and ultrasensitive assays caused by freezing patient plasma in centrifuged becton-dickinson vacutainer brand plasma preparation tubes. J Clin Microbiol2005;43: 4635–4639.
  • Zhang Q, Wang L, Jiang Y et al.Early infant human immunodeficiency virus type 1 detection suitable for resource-limited settings with multiple circulating subtypes by use of nested three-monoplex DNA PCR and dried blood spots. J Clin Microbiol2008;46: 721–726.
  • Lin HJ, Pedneault L, Hollinger FB.Intra-assay performance characteristics of five assays for quantification of human immunodeficiency virus type 1 RNA in plasma. J Clin Microbiol1998;36: 835–839.
  • Revets H, Marissens D, de Wit S et al.Comparative evaluation of NASBA HIV-1 RNA QT, AMPLICOR-HIV monitor, and QUANTIPLEX HIV RNA assay, three methods for quantification of human immunodeficiency virus type 1 RNA in plasma. J Clin Microbiol1996;34: 1058–1064.
  • Scott LE, Noble LD, Moloi J, Erasmus L, Venter WD, Stevens W.Evaluation of the Abbott m2000 RealTime human immunodeficiency virus type 1 (HIV-1) assay for HIV load monitoring in South Africa compared to the Roche Cobas AmpliPrep-Cobas Amplicor, Roche Cobas AmpliPrep-Cobas TaqMan HIV-1, and BioMerieux NucliSENS EasyQ HIV-1 assays. J Clin Microbiol2009;47: 2209–2217.
  • Swanson P, Huang S, Abravaya K et al.Evaluation of performance across the dynamic range of the Abbott RealTime HIV-1 assay as compared to VERSANT HIV-1 RNA 3.0 and AMPLICOR HIV-1 MONITOR v1.5 using serial dilutions of 39 group M and O viruses. J Virol Methods2007;141: 49–57.
  • Bourlet T, Signori-Schmuck A, Roche L et al.HIV-1 load comparison using four commercial real-time assays. J Clin Microbiol2011;49: 292–297.
  • Sire JM, Vray M, Merzouk M et al.Comparative RNA quantification of HIV-1 group M and non-M with the Roche Cobas AmpliPrep/Cobas TaqMan HIV-1 v2.0 and Abbott Real-Time HIV-1 PCR assays. J Acquir Immune Defic Syndr2011;56: 239–243.
  • Huang S, Salituro J, Tang N et al.Thermodynamically modulated partially double-stranded linear DNA probe design for homogeneous real-time PCR. Nucleic Acids Res2007;35: e101.
  • Luft LM, Gill MJ, Church DL.HIV-1 viral diversity and its implications for viral load testing: review of current platforms. Int J Infect Dis2011;15: e661–e670.
  • Cobb BR, Vaks JE, Do T, Vilchez RA.Evolution in the sensitivity of quantitative HIV-1 viral load tests. J Clin Virol2011;52( Suppl 1): S77–S82.
  • Wood E, Yip B, Hogg RS et al.Full suppression of viral load is needed to achieve an optimal CD4 cell count response among patients on triple drug antiretroviral therapy. AIDS2000;14: 1955–1960.
  • Shafer RW.Genotypic testing for human immunodeficiency virus type 1 drug resistance. Clin Microbiol Rev2002;15: 247–277.
  • Baxter JD, Mayers DL, Wentworth DN et al.A randomized study of antiretroviral management based on plasma genotypic antiretroviral resistance testing in patients failing therapy. CPCRA 046 Study Team for the Terry Beirn Community Programs for Clinical Research on AIDS. AIDS2000;14: F83–F93.
  • Cunningham S, Ank B, Lewis D et al.Performance of the applied biosystems ViroSeq human immunodeficiency virus type 1 (HIV-1) genotyping system for sequence-based analysis of HIV-1 in pediatric plasma samples. J Clin Microbiol2001;39: 1254–1257.
  • Durant J, Clevenbergh P, Halfon P et al.Drug-resistance genotyping in HIV-1 therapy: the VIRADAPT randomised controlled trial. Lancet1999;353: 2195–2199.
  • Dunn DT, Coughlin K, Cane PA.Genotypic resistance testing in routine clinical care. Curr Opin HIV AIDS2011;6: 251–257.
  • Hammer SM, Eron JJ Jr, Reiss P et al.Antiretroviral treatment of adult HIV infection: 2008 recommendations of the International AIDS Society-USA panel. JAMA2008;300: 555–570.
  • Sturmer M, Berger A, Preiser W.HIV-1 genotyping: comparison of two commercially available assays. Expert Rev Mol Diagn2004;4: 281–291.
  • Erali M, Page S, Reimer LG, Hillyard DR.Human immunodeficiency virus type 1 drug resistance testing: a comparison of three sequence-based methods. J Clin Microbiol2001;39: 2157–2165.
  • Aghokeng AF, Mpoudi-Ngole E, Chia JE, Edoul EM, Delaporte E, Peeters M.High failure rate of the ViroSeq HIV-1 genotyping system for drug resistance testing in Cameroon, a country with broad HIV-1 genetic diversity. J Clin Microbiol2011;49: 1635–1641.
  • Yang C, McNulty A, Diallo K et al.Development and application of a broadly sensitive dried-blood-spot-based genotyping assay for global surveillance of HIV-1 drug resistance. J Clin Microbiol2010;48: 3158–3164.
  • Palmer S, Kearney M, Maldarelli F et al.Multiple, linked human immunodeficiency virus type 1 drug resistance mutations in treatment-experienced patients are missed by standard genotype analysis. J Clin Microbiol2005;43: 406–413.
  • Halvas EK, Aldrovandi GM, Balfe P et al.Blinded, multicenter comparison of methods to detect a drug-resistant mutant of human immunodeficiency virus type 1 at low frequency. J Clin Microbiol2006;44: 2612–2614.
  • Toni TA, Brenner BG, Asahchop EL, Ntemgwa M, Moisi D, Wainberg MA.Development of an allele-specific PCR for detection of the K65R resistance mutation in patients infected with subtype C human immunodeficiency virus type 1. Antimicrob Agents Chemother2010;54: 907–911.
  • van Laethem K, de Munter P, Schrooten Y et al.No response to first-line tenofovir+lamivudine+efavirenz despite optimization according to baseline resistance testing: impact of resistant minority variants on efficacy of low genetic barrier drugs. J Clin Virol2007;39: 43–47.
  • Hedskog C, Mild M, Jernberg J et al.Dynamics of HIV-1 quasispecies during antiviral treatment dissected using ultra-deep pyrosequencing. PLoS One2010;5: e11345.
  • Simen BB, Simons JF, Hullsiek KH et al.Low-abundance drug-resistant viral variants in chronically HIV-infected, antiretroviral treatment-naive patients significantly impact treatment outcomes. J Infect Dis2009;199: 693–701.
  • Gianella S, Delport W, Pacold ME et al.Detection of minority resistance during early HIV-1 infection: natural variation and spurious detection rather than transmission and evolution of multiple viral variants. J Virol2011;85: 8359–8367.
  • Call SA, Saag MS, Westfall AO et al.Phenotypic drug susceptibility testing predicts long-term virologic suppression better than treatment history in patients with human immunodeficiency virus infection. J Infect Dis2001;183: 401–408.
  • Hertogs K, de Bethune MP, Miller V et al.A rapid method for simultaneous detection of phenotypic resistance to inhibitors of protease and reverse transcriptase in recombinant human immunodeficiency virus type 1 isolates from patients treated with antiretroviral drugs. Antimicrob Agents Chemother1998;42: 269–276.
  • Petropoulos CJ, Parkin NT, Limoli KL et al.A novel phenotypic drug susceptibility assay for human immunodeficiency virus type 1. Antimicrob Agents Chemother2000;44: 920–928.
  • Munoz M, Carmona R, Perez-Alvarez L et al.Analysis of discrepancies in the interpretation of antiretroviral drug resistance results in HIV-1 infected patients of Basque Country, Spain. J Clin Virol2005;33: 224–229.
  • Perez-Elias MJ, Garcia-Arota I, Munoz V et al.Phenotype or virtual phenotype for choosing antiretroviral therapy after failure: a prospective, randomized study. Antivir Ther2003;8: 577–584.
  • Cooper DA, Steigbigel RT, Gatell JM et al.Subgroup and resistance analyses of raltegravir for resistant HIV-1 infection. N Engl J Med2008;359: 355–365.
  • Veazey RS, Ketas TA, Klasse PJ et al.Tropism-independent protection of macaques against vaginal transmission of three SHIVs by the HIV-1 fusion inhibitor T-1249. Proc Natl Acad Sci USA2008;105: 10531–10536.
  • Westby M, Lewis M, Whitcomb J et al.Emergence of CXCR4-using human immunodeficiency virus type 1 (HIV-1) variants in a minority of HIV-1-infected patients following treatment with the CCR5 antagonist maraviroc is from a pretreatment CXCR4-using virus reservoir. J Virol2006;80: 4909–4920.
  • Recordon-Pinson P, Soulie C, Flandre P et al.Evaluation of the genotypic prediction of HIV-1 coreceptor use versus a phenotypic assay and correlation with the virological response to maraviroc: the ANRS GenoTropism study. Antimicrob Agents Chemother2010;54: 3335–3340.
  • Low AJ, Swenson LC, Harrigan PR.HIV coreceptor phenotyping in the clinical setting. AIDS Rev2008;10: 143–151.
  • MacArthur RD, Novak RM.Reviews of anti-infective agents: maraviroc: the first of a new class of antiretroviral agents. Clin Infect Dis2008;47: 236–241.
  • Wilkin TJ, Goetz MB, Leduc R et al.Reanalysis of coreceptor tropism in HIV-1-infected adults using a phenotypic assay with enhanced sensitivity. Clin Infect Dis2011;52: 925–928.
  • Lee-Lewandrowski E, Lewandrowski K.Perspectives on cost and outcomes for point-of-care testing. Clin Lab Med2009;29: 479–489.
  • Blyth CC, Booy R, Dwyer DE.Point of care testing: diagnosis outside the virology laboratory. Methods Mol Biol2011;665: 415–433.
  • Campbell S, Fedoriw Y.HIV testing near the patient: changing the face of HIV testing. Clin Lab Med2009;29: 491–501.
  • Lee HH, Dineva MA, Chua YL, Ritchie AV, Ushiro-Lumb I, Wisniewski CA.Simple amplification-based assay: a nucleic acid-based point-of-care platform for HIV-1 testing. J Infect Dis2010;201( Suppl 1): S65–S72.
  • An L, Tang W, Ranalli TA, Kim HJ, Wytiaz J, Kong H.Characterization of a thermostable UvrD helicase and its participation in helicase-dependent amplification. J Biol Chem2005;280: 28952–28958.
  • Vincent M, Xu Y, Kong H.Helicase-dependent isothermal DNA amplification. EMBO Rep2004;5: 795–800.
  • Goldmeyer J, Li H, McCormac M et al.Identification of Staphylococcus aureus and determination of methicillin resistance directly from positive blood cultures by isothermal amplification and a disposable detection device. J Clin Microbiol2008;46: 1534–1536.
  • Tang W, Chow WH, Li Y, Kong H, Tang YW, Lemieux B.Nucleic acid assay system for tier II labs and moderately complex clinics to detect HIV in low-resource settings. J Infect Dis2010;201( Suppl): S46–S51.
  • Tang YW, Procop GW, Persing DH.Molecular diagnostics of infectious diseases. Clin Chem1997;43: 2021–2038.
  • Jangam SR, Yamada DH, McFall SM, Kelso DM.Rapid, point-of-care extraction of human immunodeficiency virus type 1 proviral DNA from whole blood for detection by real-time PCR. J Clin Microbiol2009;47: 2363–2368.
  • Guttikonda S, Wang W, Suresh M.Molecular zipper assays: a simple homosandwich with the sensitivity of PCR. J Pharm Pharm Sci2004;7: 7–16.
  • Panhotra BR, Hassan ZU, Joshi CS, Bahrani A.Visual detection of multiple viral amplicons by dipstick assay: its application in screening of blood donors a welcome tool for the limited resource settings. J Clin Microbiol2005;43: 6218.
  • Raja S, Ching J, Xi L et al.Technology for automated, rapid, and quantitative PCR or reverse transcription-PCR clinical testing. Clin Chem2005;51: 882–890.
  • Hsia CC, Chizhikov VE, Yang AX et al.Microarray multiplex assay for the simultaneous detection and discrimination of hepatitis B, hepatitis C, and human immunodeficiency type-1 viruses in human blood samples. Biochem Biophys Res Commun2007;356: 1017–1023.
  • Miller MB, Tang YW.Basic concepts of microarrays and potential applications in clinical microbiology. Clin Microbiol Rev2009;22: 611–633.
  • Lee SH, Kim SW, Kang JY, Ahn CH.A polymer lab-on-a-chip for reverse transcription (RT)-PCR based point-of-care clinical diagnostics. Lab Chip2008;8: 2121–2127.
  • Sista R, Hua Z, Thwar P et al.Development of a digital microfluidic platform for point of care testing. Lab Chip2008;8: 2091–2104.
  • Rouet F, Rouzioux C.The measurement of HIV-1 viral load in resource-limited settings: how and where? Clin Lab2007;53: 135–148.
  • Calmy A, Ford N, Hirschel B et al.HIV viral load monitoring in resource-limited regions: optional or necessary? Clin Infect Dis2007;44: 128–134.
  • Fiscus SA, Cheng B, Crowe SM et al.HIV-1 viral load assays for resource-limited settings. PLoS Med2006;3: e417.
  • Tanriverdi S, Chen L, Chen S.A rapid and automated sample-to-result HIV load test for near-patient application. J Infect Dis2010;201( Suppl 1): S52–S58.
  • Greengrass V, Lohman B, Morris L et al.Assessment of the low-cost Cavidi ExaVir Load assay for monitoring HIV viral load in pediatric and adult patients. J Acquir Immune Defic Syndr2009;52: 387–390.
  • Lambert JS, Harris DR, Stiehm ER et al.Performance characteristics of HIV-1 culture and HIV-1 DNA and RNA amplification assays for early diagnosis of perinatal HIV-1 infection. J Acquir Immune Defic Syndr2003;34: 512–519.
  • Napravnik S, Cachafeiro A, Stewart P, Eron JJ Jr, Fiscus SA.HIV-1 viral load and phenotypic antiretroviral drug resistance assays based on reverse transcriptase activity in comparison to amplification based HIV-1 RNA and genotypic assays. J Clin Virol2010;47: 18–22.
  • Fellay J, Shianna KV, Telenti A, Goldstein DB.Host genetics and HIV-1: the final phase? PLoS Pathog2010;6: e1001033.
  • Vannberg FO, Chapman SJ, Hill AV.Human genetic susceptibility to intracellular pathogens. Immunol Rev2011;240: 105–116.
  • Telenti A, Goldstein DB.Genomics meets HIV-1. Nat Rev Microbiol2006;4: 865–873.
  • Pine SO, McElrath MJ, Bochud PY.Polymorphisms in Toll-like receptor 4 and Toll-like receptor 9 influence viral load in a seroincident cohort of HIV-1-infected individuals. AIDS2009;23: 2387–2395.
  • Ramsuran V, Kulkarni H, He W et al.Duffy-null-associated low neutrophil counts influence HIV-1 susceptibility in high-risk South African black women. Clin Infect Dis2011;52: 1248–1256.
  • Carrington M, O'Brien SJ.The influence of HLA genotype on AIDS. Annu Rev Med2003;54: 535–551.
  • Gonzalez E, Kulkarni H, Bolivar H et al.The influence of CCL3L1 gene-containing segmental duplications on HIV-1/AIDS susceptibility. Science2005;307: 1434–1440.
  • Bleiber G, May M, Martinez R et al.Use of a combined ex vivo/in vivo population approach for screening of human genes involved in the human immunodeficiency virus type 1 life cycle for variants influencing disease progression. J Virol2005;79: 12674–12680.
  • Fellay J, Marzolini C, Meaden ER et al.Response to antiretroviral treatment in HIV-1-infected individuals with allelic variants of the multidrug resistance transporter 1: a pharmacogenetics study. Lancet2002;359: 30–36.
  • Haas DW, Smeaton LM, Shafer RW et al.Pharmacogenetics of long-term responses to antiretroviral regimens containing Efavirenz and/or Nelfinavir: an Adult Aids Clinical Trials Group Study. J Infect Dis2005;192: 1931–1942.
  • Tsuchiya K, Gatanaga H, Tachikawa N et al.Homozygous CYP2B6 *6 (Q172H and K262R) correlates with high plasma efavirenz concentrations in HIV-1 patients treated with standard efavirenz-containing regimens. Biochem Biophys Res Commun2004;319: 1322–1326.
  • Pereyra F, Jia X, McLaren PJ et al.The major genetic determinants of HIV-1 control affect HLA class I peptide presentation. Science2010;330: 1551–1557.
  • Kerr JR, Kaushik N, Fear D, Baldwin DA, Nuwaysir EF, Adcock IM.Single-nucleotide polymorphisms associated with symptomatic infection and differential human gene expression in healthy seropositive persons each implicate the cytoskeleton, integrin signaling, and oncosuppression in the pathogenesis of human parvovirus B19 infection. J Infect Dis2005;192: 276–286.
  • Pavy N, Pelgas B, Beauseigle S et al.Enhancing genetic mapping of complex genomes through the design of highly-multiplexed SNP arrays: application to the large and unsequenced genomes of white spruce and black spruce. BMC Genomics2008;9: 21.
  • Chehab FF, Kan YW.Detection of specific DNA sequences by fluorescence amplification: a color complementation assay. Proc Natl Acad Sci USA1989;86: 9178–9182.
  • Saiki RK, Walsh PS, Levenson CH, Erlich HA.Genetic analysis of amplified DNA with immobilized sequence-specific oligonucleotide probes. Proc Natl Acad Sci USA1989;86: 6230–6234.
  • Kornher JS, Livak KJ.Mutation detection using nucleotide analogs that alter electrophoretic mobility. Nuc Acids Res1989;17: 7779–7784.
  • Alves AM, Carr FJ.Dot blot detection of point mutations with adjacently hybridising synthetic oligonucleotide probes. Nuc Acids Res1988;16: 8723.
  • Landegren U, Kaiser R, Sanders J, Hood L.A ligase-mediated gene detection technique. Science1988;241: 1077–1080.
  • Kwok PY.Approaches to allele frequency determination. Pharmacogenomics2000;1: 231–235.
  • Shi MM.Technologies for individual genotyping: detection of genetic polymorphisms in drug targets and disease genes. Am J Pharmacogenomics2002;2: 197–205.
  • Margulies M, Egholm M, Altman WE et al.Genome sequencing in microfabricated high-density picolitre reactors. Nature2005;437: 376–380.
  • Douek DC, McFarland RD, Keiser PH et al.Changes in thymic function with age and during the treatment of HIV infection. Nature1998;396: 690–695.
  • Hatzakis A, Touloumi G, Karanicolas R et al.Effect of recent thymic emigrants on progression of HIV-1 disease. Lancet2000;355: 599–604.
  • Ribeiro RM, de Boer RJ.The contribution of the thymus to the recovery of peripheral naive T-cell numbers during antiretroviral treatment for HIV infection. J Acquir Immune Defic Syndr2008;49: 1–8.
  • Dion ML, Bordi R, Zeidan J et al.Slow disease progression and robust therapy-mediated CD4+ T-cell recovery are associated with efficient thymopoiesis during HIV-1 infection. Blood2007;109: 2912–2920.
  • Haugaard SB, Andersen O, Pedersen SB et al.Depleted skeletal muscle mitochondrial DNA, hyperlactatemia, and decreased oxidative capacity in HIV-infected patients on highly active antiretroviral therapy. J Med Virol2005;77: 29–38.
  • Swartz MN.Mitochondrial toxicity—new adverse drug effects. N Engl J Med1995;333: 1146–1148.
  • Cote HC, Brumme ZL, Craib KJ et al.Changes in mitochondrial DNA as a marker of nucleoside toxicity in HIV-infected patients. N Engl J Med2002;346: 811–820.
  • Casula M, Weverling GJ, Wit FW et al.Mitochondrial DNA and RNA increase in peripheral blood mononuclear cells from HIV-1-infected patients randomized to receive stavudine-containing or stavudine-sparing combination therapy. J Infect Dis2005;192: 1794–1800.
  • Garrabou G, Moren C, Gallego-Escuredo JM et al.Genetic and functional mitochondrial assessment of HIV-infected patients developing HAART-related hyperlactatemia. J Acquir Immune Defic Syndr2009;52: 443–451.
  • Miro O, Lopez S, Rodriguez de la Concepcion M et al.Upregulatory mechanisms compensate for mitochondrial DNA depletion in asymptomatic individuals receiving stavudine plus didanosine. J Acquir Immune Defic Syndr2004;37: 1550–1555.
  • Lin CH, Sloan DD, Dang CH et al.Assessment of mitochondrial toxicity by analysis of mitochondrial protein expression in mononuclear cells. Cytometry B Clin Cytom2009;76: 181–190.
  • Wang Z, Trillo-Pazos G, Kim SY et al.Effects of human immunodeficiency virus type 1 on astrocyte gene expression and function: potential role in neuropathogenesis. J Neurovirol2004;10 ( Suppl 1): 25–32.
  • Giri MS, Nebozhyn M, Showe L, Montaner LJ.Microarray data on gene modulation by HIV-1 in immune cells: 2000–2006. J Leukoc Biol2006;80: 1031–1043.
  • Rotger M, Dalmau J, Rauch A et al.Comparative transcriptomics of extreme phenotypes of human HIV-1 infection and SIV infection in sooty mangabey and rhesus macaque. J Clin Invest2011;121: 2391–2400.
  • Wang Z, Gerstein M, Snyder M.RNA-Seq: a revolutionary tool for transcriptomics. Nat Rev Genet2009;10: 57–63.
  • Cirulli ET, Singh A, Shianna KV et al.Screening the human exome: a comparison of whole genome and whole transcriptome sequencing. Genome Biol2011;11: R57.