• No results found

Towards virological monitoring of HIV-1 drug resistance in resource-limited settings - Thesis

N/A
N/A
Protected

Academic year: 2021

Share "Towards virological monitoring of HIV-1 drug resistance in resource-limited settings - Thesis"

Copied!
236
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

UvA-DARE is a service provided by the library of the University of Amsterdam (https://dare.uva.nl)

Towards virological monitoring of HIV-1 drug resistance in resource-limited

settings

Aitken, S.C.

Publication date

2014

Document Version

Final published version

Link to publication

Citation for published version (APA):

Aitken, S. C. (2014). Towards virological monitoring of HIV-1 drug resistance in

resource-limited settings.

General rights

It is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), other than for strictly personal, individual use, unless the work is under an open content license (like Creative Commons).

Disclaimer/Complaints regulations

If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the material inaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letter to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. You will be contacted as soon as possible.

(2)

-1 drug resistance in resource-limited settings

Susan C.

Aitken

Towards virological monitoring

of HIV-1 drug resistance

in resource-limited settings

Susan C. Aitken

(3)

HIV-1 drug resistance

in resource-limited settings

(4)

Copyright © 2013, Susan C. Aitken, The Netherlands. No part of this thesis may be reproduced, stored or transmitted in any form or by any means, without prior permis-sion of the author.

ISBN: 978-94-6108-578-8

Layout and cover design: Susan Aitken and Janna McCall-Peat Printed by: Gildeprint Drukkerijen

The studies included in this thesis were part of the Affordable Resistance Test for Africa (ART-A) program and were supported by a grant from the Netherlands Or-ganization for Scientific Research for Global Development (NWO/WOTRO), under the Netherlands African Partnership for Capacity Development and Clinical Inter-ventions against Poverty-related Diseases (NACCAP; grant W.07.05.204.00), The Hague, The Netherlands.

(5)

HIV-1 drug resistance

in resource-limited settings

ACADEMISCH PROEFSCHRIFT

ter verkrijging van de graad van doctor aan de Universiteit van Amsterdam op gezag van de Rector Magnificus

prof. dr. D.C. van den Boom

ten overstaan van een door het college voor promoties ingestelde commissie, in het openbaar te verdedigen in de Agnietenkapel

op woensdag 22 januari 2014, te 10:00 uur

door

Susan Claire Aitken

(6)

Promotores: Prof. dr. T.F. Rinke de Wit Prof. dr. W. Stevens

Co-promotores: Dr. R. Schuurman Dr. C.L. Wallis

Overige leden: Prof. dr. M.D. de Jong Prof. dr. P.R. Klatser Prof. dr. J.M.A. Lange Dr. M. Nijhuis

Prof. dr. P. Reiss Prof. dr. E.J. Wiertz

(7)

Abbreviations General Introduction

Development and evaluation of an affordable real-time qualitative assay for determining HIV-1 virological failure in plasma and dried blood spots. Journal of Clinical Microbiology (2013) 51(6):

1899-905

A pragmatic approach to HIV-1 drug resistance determination in resource-limited settings using a novel RT-only genotyping assay.

Journal of Clinical Microbiology (2013) 51(6): 1757-61

Evaluation of an affordable HIV-1 virological failure assay and an-tiretroviral drug resistance genotyping protocol. Journal of

Virolog-ical Methods (2013) 194(1-2): 300-7

Development and evaluation of an assay for HIV-1 protease and reverse transcriptase drug resistance genotyping of all major group-M subtypes. Journal of Clinical Virology (2012) 54: 21-25 Accumulation of drug resistance and loss of therapeutic options precede commonly used criteria for treatment failure in HIV-1 sub-type C infected patients. Antiviral Therapy (2012) 17(2):377-86 HIV-1 resistance testing on dried blood spots enables individual patient management in rural South African Setting. Unpublished Stability of 1 nucleic acids in dried blood spot samples for HIV-1 drug resistance genotyping. Unpublished

General discussion Summary

Nederlandse samenvatting Authors and affiliations Acknowledgements List of publications About the author

Chapter 1 Chapter 2 Chapter 3 Chapter 4 Chapter 5 Chapter 6 Chapter 7 Chapter 8 Chapter 9 Appendix 6 11 36 61 81 107 125 151 163 183 205 211 217 221 227 233

(8)

aa amino acid

AAC accelerated access initiate ABC abacavir

AIDS acquired immunodeficiency syndrome

AIGHD Amsterdam institute for global health and development AMC academic medical centre (Amsterdam)

(c)ART (combination) antiretroviral therapy ARTA affordable resistance test for Africa ARV antiretroviral (drug)

ATV atazanavir AZT zidovudine

cDNA complementary DNA

CCR5 chemokine (C-C motif) receptor 5 CBV combivir

CI confidence interval

CRF circulating recombinant form Ct cycles to threshold

CXCR4 chemokine (C-X-C motif) receptor 4 CV (%) coefficent of variance

d4T stavudine DBS dried blood spot

ddI didanosine dideoxyinosine DLV delavirdine

DNA deoxyribonucleic acid DRM drug resistance mutation DRV darunavir

EDTA ethylenediaminetetraacetic acid EFV efavirenz

EMC encephalomyocarditis virus ENF enfuvirtide

ETR etravarine

EWI early warning indicator

FDA United States food and drug administration FPV fosamprenavir calcium

FTC emtrictabine GP glycoprotein

GRADE (HIV-1) Genotypic Resistance-Algorithm Deutschland GSS genotyping sensitivity score

(9)

HAART highly active antiretroviral therapy HIV human immunodeficiency virus HIVDB HIV database (Stanford DB) HIVDR HIV drug resistance

HIVResNet global HIV drug resistance network IAS international AIDS society

IC internal control IDV indinavir

IQR inter-quartile range

JCRC joint clinical research centre LED light-emitting diode

LIC low income country LOD level of detection LPV lopinavir

LTR long terminal repeat

MEGA molecular evolutionary genetics analysis MVC maraviroc

mRNA messenger RNA n number (of samples)

NACCAP Netherlands African partnership for capacity development and clinical interventions against poverty-related diseases

NCBI national centre for biotechnology information NA nucleic acid

NFV nelfinavir

NGO non-governmental organization NGS next generation sequencing NMC Ndlovu medical centre

NNRTI non-nucleoside reverse transcriptase inhibitor NPV negative predictive value

NRTI nucleoside reverse transcriptase inhibitor NVP nevirapine

NWO/WOTRO Netherlands organisation for scientifid reaseach for global development

PASER PharmAccess African studies to evaluate resistance PBMC peripheral blood mononuclear cells

PCR polymerase chain reaction (b)PI (boosted) protease inhibitor POC point-of-care

PMTCT prevention of mother-to-child transmission of HIV PPV positive predictive value

(10)

PR protease RAL raltegravir

RDI HIV resistance response database initiative RPV rilpivirine

RLS resource-limited settings RNA ribonucleic acid

RRS resource-rich settings

RT(I) reverse transcriptase (inhibitor)

RT-PCR reverse transcriptiase polymerase chain reaction RTV ritonavir

SANBI South African national bioinformatics institute SATuRN Southern African treatent resistance network SD standard deviation

SQV saquinavir mesylate

TAM thymidine analogue mutation TDF tenofovir

TDR transmitted drug resistance TPV tipranavir

UMCU university medical centre Utrecht UNICEF United Nations children’s fund

UNAIDS joint United Nations programme on HIV/AIDS USD United States dollar

VF(A) virological failure (assay) VL (HIV) viral load

WB whole blood

(11)
(12)
(13)

C

A

H

P

T

E

R

1

(14)

S

ince the first recorded cases in 1981, human immunodeficiency virus (HIV) has spread globally, resulting in the death of over 35 million people(1). The global HIV-1 epidemic has been estimated to include 34.0

million (range 31.4-35.9 million) infected adults and children by the end of 2011. An alarming 69% (n= 23.5 million) of the total global infections are people residing in Sub-Saharan Africa. Prevalence is highest in adults aged 15-49, affecting 0.8% of the global adult population, and as high as 4.9% of adults residing in sub-Saharan Africa(2). People at greatest risk of

being infected with HIV are homosexual and heterosexual people having unprotected sex, sex workers, people who inject drugs, and children born to HIV-infected mothers(2).

HIV primarily infects CD4+ T-lymphocytes and macrophages, entering the body

via mucous membranes or through blood-blood contact. After transmission, an acute infection ensues, characterised by rapid virus replication and immune activation(3-5). The infection then stabilises to a chronic condition,

characterised by stable, continuous viral replication and a gradual decline in CD4+ T-lymphocytes(6). If not treated, the decline in CD4+ T-lymphocytes will

continue until the immune system of the infected individual is compromised, thereby making them vulnerable to opportunistic infections leading to acquired immunodeficiency syndrome (AIDS) and eventually death.

The number of people dying from AIDS-related illnesses was estimated to be 1.7 million (range 1.5-1.9 million) worldwide in 2011. Compared to 2005, this is a 24% reduction. The primary reason for this reduction is the scale-up and success of antiretroviral treatment (ART) programs, which have resulted in a decrease of AIDs related deaths and a decline in the incidence of new HIV infections. Antiretroviral (ARV) access in resource-limited settings (RLS) has also improved the health of people infected with HIV, enabling them to lead relatively normal lives, including being able to work and providing an income for themselves and their families(2).

(15)

1

Human immunodeficiency virus, HIV

HIV is an enveloped RNA virus belonging to the Family of the Retroviridae, genus lentiviridae. HIV-1 is the predominant global HIV species, with the less pathogenic HIV-2 found mostly in West Africa. HIV-1 is further divided into four groups, namely major (M), non-M/non-O (N), outlier (O) and pending the identification of further human cases (P), of which M is the primary contributor to the global HIV pandemic. Group M HIV-1 comprises of subtypes A- D, F-H, J and K, and numerous circulating recombinant forms (CRFs; currently n= 58), including common CRFs 01_AE and 02_AG(7, 8) (Figure 1). HIV-1

subtype C is responsible for the majority of global HIV-1 infections (48%), followed by subtypes A (12%) and B (11%). Evidence suggests that Central Africa is where the HIV epidemic originated and as such has the largest diversity of subtypes (Figure 1) (9-11). In sub-Saharan Africa infections are

primarily caused by HIV-1 subtype C, whilst in Europe and North America HIV-1 subtype B predominates, although subtype diversity has increased over the last decade, particularly in Europe(7).

(16)

Overview of the replicative cycle of HIV

During infection, HIV first binds to the surface of target cells by interaction of the viral surface glycoprotein 120 (gp120) with the CD4 receptor on the cell surface(12, 13). This binding causes a conformational change in gp120(14, 15), allowing for an additional interaction of gp120 with either CC chemokine

receptor 5 (CCR5)(16) or CxC chemokine receptor 4 (CXCR4)(17). This

interaction triggers the viral transmembrane subunit gp41 to interact with the target cell, eventually leading to fusion of the virus envelope with the target cell(18) and internalization of the virus particle. The virus uncoats, releasing

the genomic RNA into the cytoplasm of the cell. Once in the cytoplasm the genomic viral RNA is converted into double-stranded cDNA by the viral reverse transcriptase (RT), which is then complexed with integrase and other viral components to form the pre-integration complex(19) enabling entry

into the cells’ nucleus. In the nucleus, the integrase enzyme mitigates the integration of the double-stranded viral DNA into the host cell genome. Once integration has occurred proviral DNA will be copied into each daughter cell with every division cycle of an infected cell, thereby achieving long-term presence in the host genome. Furthermore, the proviral DNA serves as a template for transcription to produce viral mRNA and genomic HIV-1 RNA molecules. In the cytoplasm, viral mRNA is translated into viral precursor polyproteins, which are cleaved by protease (PR), a process necessary to produce infectious viral progeny(20). Cleaved viral proteins are then assembled

with RNA genomes into new virions on the cellular membranes(21), eventually

being released as virus particles (Figure 2).

Cells that have productive HIV replication generally have a short life span, either being destroyed as a result of virus replication or by the host immune response. HIV infection is not always productive, which is the case when infection stops at the point of integration and the cell enters a resting state, referred to as latently infected resting CD4+ T cells(23). Integrated proviral

DNA found in latently infected cells is referred to as archived virus, which can represent a viral population different to the currently circulating population.

(17)

1

Latently infected, resting CD4+ T cells are the largest barrier to curing HIV as they are a source for reactivation of viral replication(24-26).

Figure 2. Replication cycle of HIV (adapted from Engelman et al. 2012(22))

HIV Therapy

There are currently six classes of FDA-approved ARV drugs that target different steps of the HIV replication cycle: Nucleoside reverse transcriptase inhibitors (NRTI), non-nucleoside reverse transcriptase inhibitors (NNRTI), protease inhibitors (PI), fusion inhibitors, integrase inhibitors, and entry inhibitors (CCR5)(27). All currently available drugs for the treatment of HIV are

summarised with their introduction over time in Figure 3.

Initial treatment for HIV infection was based on the NRTI zidovudine (AZT) monotherapy. Although clinical benefit was demonstrated upon AZT treatment, the effect was rather temporal and lasted for only weeks to months(28, 29) due

(18)

Figure 3. Currently available FDA-approved antiretroviral drugs for HIV treatment,

with their introduction over time.

Nucleoside reverse transcriptase inhibitors (NRTI; Black): Zidovudine azidothymidine (AZT,

ZDV), didanosine dideoxyinosine (ddI), stavudine (d4T), lamivudine (3TC), abacavir sulphate

(ABC), enteric coated didanosine (ddI EC), tenofovir disoproxil fumerate (TDF), emtricitabine

(FTC). Non-nucleoside reverse transcriptase inhibitors (NNRTI; Blue): Nevirapine (NVP),

delavirdine (DLV), efavirenz (EFV), etravarine (ETR), rilpivirine (RPV), nevirapine extended

release (NVP-XR). Protease inhibitor (PI; Red): Saquinavir mesylate (SQV), ritonavir

(RTV), indinavir (IDV), nelfinavir mesylate (NFV), lopinavir and ritonavir (LPV/r), atazanavir

sulphate (ATV), fosamprenavir calcium (FPV), tipranavir (TPV), darunavir (DRV). Fusion

inhibitors (Green): Enfuvirtide (T-20, ENF). Entry Inhibitor (CCR5; Purple): Maraviroc (MVC).

Integrase inhibitor (Orange): Raltegravir (RAL), Elvitegravir (EVG). Cobicistat (COBI; pink)

is a cytochrome P450 3A enzyme inhibitor. Combination regimens are underlined. Generic drugs are shown in italics.

1981 1987 1991 1994 1995 1996 1997 1998 2000 2001 2003 2004 2005 2006 2007 2008 201

1

1986

First reported AIDS case First virus isolated

First FDA-approved blood test

AZT ddI d4T 3TC, SQV RTV, IDV, NVP NFV, DLV, AZT/3TC EFV, ABC LPV/r, ddI-EC, AZT/3TC/ABC TDF ENF, ATV, FTC, FPV MVC, RAL 2012

ABC/3TC, TDF/FTC, ddI generic

TPV, AZT generic DRV, TDF/FTC/EFV ETR RPV, TDF/FTC/RPV, NVP-XR EVG/FTC/TDF/COBI 1985 HIV named

First FDA-approved antiretroviral

(19)

1

to the emergence of viruses harbouring reduced susceptibility to the drug(30, 31). New drugs were developed, and in the early 1990s combination therapy

was shown to have a greater success for long term HIV treatment(32, 33). The

use of combination antiretroviral therapy (cART) was introduced in resource-rich settings (RRS) in the mid-1990s. With the introduction of cART, the number of people dying from AIDS-related illnesses substantially decreased

(34-36).

Antiretroviral Therapy Roll-out

When introduced, the cost of cART was on average USD 10 000 to 15 000 per person per year, restricting access to RRS. International lobby by HIV/ AIDS activists put pressure on governments and branded pharmaceutical companies to reduce the cost. In 2000 the International Aids Society (IAS) conference was held for the first time on African soil in Durban, South Africa. The conference highlighted the need for improved HIV treatment in RLS, and subsequently the Accelerated Access Initiate (AAI) was undertaken by the WHO, lowering the costs of cART considerably. In 2002 a total of only 300 000 infected individuals in low- and middle-income countries were receiving the necessary ART(1). The WHO “3 by 5” initiative(37) was responsible for

increasing access to cART to more than 6.6 million infected individuals in 2011(1). In 2011, a political declaration was made, the United Nations Political

Declaration on HIV and AIDS, where countries pledged to intensify efforts to eliminate HIV and AIDS(38). One of the goals of this pledge is to provide

antiretroviral therapy to 15 million people by 2015, and as a result just over 9.7 million infected individuals are currently receiving cART in low- and middle-income countries(39) (Figure 4 and 5).

The 2013 WHO guidelines on HIV treatment have greatly increased the number of people qualifying for treatment. People eligible for ART include all HIV infected adults and children older than five years of age presenting with stage 3 or 4 AIDS defining illnesses and/or with a CD4 count ≤500 (previously ≤350(41)) cells/mm3; with active TB infection; with HBV co-infection with

(20)

evidence of severe chronic liver disease; partners with HIV in serodiscordant couples to reduce HIV transmission (new); and pregnant and breastfeeding (new) women with HIV(40). For children younger than five years of age

infected with HIV, ART should be given regardless of WHO clinical stage or CD4 count. With the introduction of the new staring criteria, an additional 9.2 million people qualify for ART compared to the 2010 guidelines(41).

Figure 4. Actual and projected number of people receiving antiretroviral therapy in

low- and middle-income countries, by region, 2003-2015(39) (Source: 2013 Global

AIDS Response Progress Reporting (WHO/UNICEF/UNAIDS)).

When first-line ARV treatment is no longer effective in suppressing viral replication, in most cases due to emerging drug resistance, switching to a second-line regimen is warranted. The WHO suggested second-line regimen consists of a boosted PI, preferably atazanavir/ritonavir (ATV/r) or lopinavir/ ritonavir (LPV/r), and two NRTIs, preferably AZT and 3TC, the choice of which is dependant on an infected individuals previous first-line regimen(40).

In addition to use as a PI in second-line therapy, LPV/r and two NRTIs are also recommended in first-line treatment for HIV infected children younger

(21)

1

than three years of age(40). In general, the availability of PI-based regimens

varies largely between countries, mainly due to its much higher cost.

0 10 20 30 40 50 60 70 80 90 100 Angol a Botsw ana Burun di Cam eroon Chad Côte d 'Ivoir e Dem ocrat ic Repu blic of Cong o Ethiopi a Ghan a Keny a Lesotho Ma lawi Mozam bique Namib ia Nigeria Sout h Afric a Swa zilan d Ugand a* United Republ ic of Tanz ania Zam bia Zimbabw e

Figure 5. Antiretroviral therapy (ART) coverage among adults and children eligible for

ART in 21 African countries with a high burden of HIV infection, 2012. Blue: Southern Africa; Green: East and Central Africa; Yellow: West Africa. (Adapted from: 2013 Global AIDS Response Progress Reporting (WHO/UNICEF/UNAIDS) and 2013 UNAIDS/WHO estimates)

Current cART regimens for the treatment of HIV-1 in adults and adolescents in RLS, as recommended by WHO guidelines, are a combination of two NRTIs and one NNRTI. As this is the starting therapy, it is commonly known as first-line treatment regimen. The preferred first-line regimen is comprised of tenofovir disoproxil fumerate (TDF), lamivudine (3TC) or emtricitabine (FTC), and efavirenz (EFV)(40). Alternative regimens allow for zidovudine

(AZT) in place of TDF, and nevirapine (NVP) in place of EFV(40). For children

three years of age and up, the recommenced first-line includes EFV, 3TC, and the NRTI abacavir (ABC). Until recently, regimens in RLS often contained stavudine (d4T), however as a result of high toxicity associated with its use, d4T use is declining.

(22)

HIV Drug Resistance

The reverse transcriptase enzyme, responsible for converting viral RNA into cDNA (Figure 1), lacks a proof-reading capability resulting in, on average, one mutation per genome per replication cycle(42). A lack of proof reading

ability and frequent recombination events(43, 44), combined with the high

turn-over of virus particles, on average 109 -1011 virus particles daily(45, 46), results

in a multitude of different viruses within a population. This genetically diverse population of viruses is referred to as viral quasispecies(47), and can include

viral variants containing drug resistance mutations (DRMs)(48, 49).

Mutations conferring drug resistance are introduced in the viral genome at reverse transcription, and the proviral DNA containing these mutations is subsequently integrated into the cellular genome. Even when viral replication is suppressed by effective treatment, integrated proviral DNA is still present in cells. Virus variants produced from proviral DNA encoding resistance often have a reduced replication capacity and are out competed by wild-type virus. However, incomplete suppression of viral replication due to suboptimal drug pressure as a result of poor adherence, treatment interruption, insufficient plasma drug concentrations, or the use of inferior drugs or drug combinations, may result in a selective replicative advantage for viral variants with resistance to that drug. In such a situation, resistance virus can rapidly become the dominant virus, decreasing efficacy of the particular ART regimen and resulting in ARV treatment failure. As such, sub-optimal bioavailability of the ARV leads to the acquisition of drug resistance. In addition, individuals harbouring drug resistant virus may transmit this virus to previously uninfected individuals, referred to as transmitted drug resistance (TDR).

DRMs are specific to the drug class they affect. Reduced NRTI susceptibility is associated with specific mutations at 16 amino acid positions in RT(50).

Similarly, various mutations conferring reduced susceptibility to NNRTIs have been identified, as well as for PIs and inhibitors of the viral integrase enzyme.

(23)

1

Within and between drug classes, those mutations can have either additive, synergistic or antagonistic cross-reactivities, which makes interpretation of mutational patterns for clinical drug efficacy purposes a rather complex and specialised matter.

The WHO established a global HIV Drug Resistance Network (HIVResNet) in order to monitor the emergence of HIVDR. Countries in Africa that are currently involved include Burundi, Cameroon, Kenya, Malawi, Mozambique, Nigeria, South Africa, Swaziland, Zambia, and Zimbabwe. In the 2012 report, results of the WHO survey of acquired drug resistance obtained between 2006 and 2010 have shown that of the individuals failing therapy (VL >1000 copies/ml) at 12 months, 72% had DRMs (Figure 6). The most common mutation observed is the NRTI mutation M184V followed by several NNRTI mutations, such as K103N, Y181C and V106M(51). Due to the limited use

and later introduction of PI containing regimens, the occurrence of PI drug resistance is still low(51).

Figure 6. Prevalence of HIV drug resistance-associated mutations among people

experiencing treatment failure at 12 months. Mutations were defined using the 2009 WHO surveillance resistance mutation list. (Source: WHO Resistance Report 2012(51)).

(24)

According to the WHO resistance report for 2012, there has been an increase in TDR between 2004 and 2010 in RLS areas conducting HIVDR surveillance, either in published studies or as part of the WHO surveys(51).

Of note was the increase and prevalence of NNRTI resistance. A slightly increased prevalence of HIVDR was seen in countries with high national ART coverage(51), with the level of TDR being linked to the duration of national

roll-out programs(52, 53).

Treatment Monitoring

During the progression of HIV infection (Figure 7), an increase in viral replication generally precedes a decrease in CD4+ lymphocytes(54), which,

in the long-term, is then followed by the presentation of AIDS-defining illnesses. VL monitoring is an effective tool to monitor for the first signs of treatment failure. The role of CD4+ lymphocyte counts is to gauge the state of the immune system to determine whether there is a risk for infection by opportunistic infections.

Figure 7. A generalised graph of the progression of HIV infection. (Adapted from

(25)

1

In RRS, the standard of care for HIV treatment monitoring employs CD4+ counts and VL monitoring at entry into care and every three to six months thereafter. In addition to VL and CD4+ testing, resistance testing is often preformed prior to treatment initiation in order to ensure the most effective choice of therapy,as well as at virological failure in order to select an appropriate follow-up regimen(56).

By contrast, treatment monitoring in most RLS primarily involves clinical monitoring for stage 3 and 4 AIDS-defining illnesses and, if available, immunological monitoring using CD4+ counts(40). The inadequacies of CD4+

counts for determining treatment failure have been described on many occasions, either leading to unnecessary switching of regimens due to incorrectly diagnosed or presumed virological failure(57-61), or an accumulation

of drug resistance mutations due to extended exposure to a failing regimen(62).

Recently, the WHO added VL monitoring as a strong recommendation for diagnosis and confirmation of treatment failure in RLS(40), however its use is

limited due to costs and lack of laboratory infrastructure. Similarly, the use of HIVDR genotyping for individual patient management in RLS is limited to clinical research studies, and for pre-authorized private medical care, or at the patients own expense. Limited routine resistance testing is performed due to high cost, infrastructure requirements, and complexity of available assays.

Logistics of Treatment Monitoring

Commercial VL and HIVDR genotyping assays are performed from plasma, which require cold-chain maintenance and biohazardous containment. This limits their use in RLS to collection sites with appropriate sample processing facilities and cold-storage, which are also within easy reach of reference laboratories for subsequent testing. The use of dried blood spot samples (DBS) is becoming increasingly popular for VL monitoring and HIVDR genotyping in RLS. Numerous studies have been conducted to determine potential application of DBS in various assays, reviewed by Hamers et al. (2009)(63)

(26)

and Bertagnolio et al. (2010)(64). Although initial data suggests application is

feasible for VL and HIVDR genotyping, findings are inconsistent with respect to storage temperature and duration, and more detailed analysis on the effect of storage on sample degradation is necessary to ensure reliable results. Additionally, the sampling technique has numerous drawbacks: small sample volumes decrease analytical sensitivity, impaired nucleic acid integrity when stored in sub-optimal conditions, laborious nucleic acid isolation procedures limiting high throughput capacity, and potential interference of archived proviral DNA.

Treatment Challenges in RLS

The majority of HIV-infected individuals reside in Africa. Whilst access to treatment is increasing, there are still logistical hurdles to prevent HIVDR and maintain long-term treatment success. The largest risk factor associated with the development of HIVDR is adherence. The recent introduction of once-daily fixed dose cART greatly decreased pill burden and should improve patient adherence; however, adherence is still hindered by drug stock outs due to poor supply and procurement management, as well as limited supportive care, including VL and CD4 testing, resulting in extended exposure to failing regimens.

In addition to logistical hurdles, the vast array of subtypes found in Africa add an additional challenge to treatment and testing. Some resistance mutations are more prevalent in certain subtypes due to differences in the genetic make-up between subtypes thereby facilitating the emergence of some mutations over others. For example, studies have shown that the prevalence of K65R, conferring reduced susceptibility to d4T, ddI, ddC, 3TC, FTC, ABC, and TDF(50), is higher in nonsubtype-B clades(65-69). Similarly, a higher prevalence

of the V106M mutation over the V106A is observed in subtype C viruses, which is associated with high-level EFV resistance(70, 71). Whilst individual

patient management is currently not feasible in RLS due to costs, population-based surveillance for HIVDR mutations prevalence and subtype distribution

(27)

1

is necessary in order to make the most effective choice of ARV therapy for national guidelines.

Scope of This Thesis

Affordable Resistance Test for Africa: ARTA

Established in 2008, the ARTA project is a private-public consortium headed up by the PharmAccess Foundation, a non-profit organisation affiliated with the Center for Poverty-related Communicable Diseases of the Academic Medical Centre (AMC) in Amsterdam. The collaborators involved in the project include the University Medical Centre Utrecht (UMCU) in the Netherlands, the Centre de Recherche Public de la Santé in Luxembourg, the University of the Witwatersrand and Contract Laboratory Services in Johannesburg, South Africa, and Janssen Diagnostics BVBA (formerly Virco BVBA) in Belgium. The ARTA project aimed to develop more affordable HIV-1 treatment monitoring applications, including VL monitoring and HIVDR genotyping, which can be universally applied in RLS. The specific requirements are given in Box 1.

Box 1. Specific requirements for ARTA assays:

Simplified sample collection and transport (DBS)Optimized nucleic acid elution methodologies for DBSHIV subtype-independent assays nucleic acid amplificationPragmatic genotyping methods

Commercial VL assays provide precise measurement of viral RNA copies found in a sample, but generally require large equipment designed for high-throughput to ensure cost effectiveness, which is not always ideal for small- and medium-throughput facilities. However, in many cases exact VL quantitation is not required to determine treatment failure. Therefore, an assay that classifies a sample as either above or below a treatment success threshold could have wide application. As such, a simple virological failure (VF) screening assay that is compatible for application with DBS was

(28)

developed, validated and transferred to three laboratories in Uganda with various levels of experience on application of molecular monitoring methods

(Chapter 2).

A recent review of ARV treatment programs in sub-Saharan Africa has shown that 94% of treatment is first-line based, and effective virological suppression is achieved for approximately 80% of these patients(72). Similarly, in the 2012

WHO survey of acquired drug resistance, 76% of patients achieved virological suppression at 12-months after initiation of a standard first-line regimen(51).

However, 20-24% of patients failing cART did not have DRM, and would have had unnecessary treatment switching without access to HIVDR genotyping results. Taking into consideration that the majority of HIV infected individuals receive first-line cART, a low cost HIVDR genotyping assay targeting the region of RT harbouring all major RT inhibitor resistance mutation positions was developed and applied clinically for HIVDR determination in first-line therapy failure (Chapter 3). Further, evaluation of the application of newly

developed VL and HIVDR genotyping assays in RLS is shown to demonstrate feasibility in a South African laboratory (Chapter 4).

For more in depth studies on first- and second-line drug resistance, an alternative to commercial PR-RT based HIVDR genotyping assays is described (Chapter 5), and is integrated application in RLS-related studies is

shown (Chapters 6, and 7). Lastly, the stability of DBS samples is important

to ensure reliable results using DBS sampling, and a further understanding of the effect of storage conditions On DBS sample integrity is explored

(Chapter 8). A final discussion (Chapter 9) concludes the relevance of the

described research.

References

1. WHO/UNAIDS/UNICEF. 2011. Progress report 2011: Global HIV/AIDS

response. http://www.who.int/hiv/pub/progress_report2011/en/index.html. 2. UNAIDS. 2012. Global report: UNAIDS report in the global AIDS epidemic

(29)

1

2012. http://www.unaids.org/en/media/unaids/contentassets/documents/ epidemiology/2012/gr2012/20121120_UNAIDS_Global_Report_2012_ with_annexes_en.pdf.

3. Borrow P, Lewicki H, Hahn BH, Shaw GM, Oldstone MB. 1994.

Virus-specific CD8+ cytotoxic T-lymphocyte activity associated with control of viremia in primary human immunodeficiency virus type 1 infection. J Virol

68:6103-6110.

4. Daar ES, Moudgil T, Meyer RD, Ho DD. 1991. Transient high levels of

viremia in patients with primary human immunodeficiency virus type 1 infection. N Engl J Med 324:961-964.

5. Koup RA, Safrit JT, Cao Y, Andrews CA, McLeod G, Borkowsky W, Farthing C, Ho DD. 1994. Temporal association of cellular immune responses

with the initial control of viremia in primary human immunodeficiency virus type 1 syndrome. J Virol 68:4650-4655.

6. Dewhurst SL, da Cruz RL, Whetter L. 2000. Pathogenesis and treatment

of HIV-1 infection: recent developments (Y2K update). Front Biosci

5:D30-49.

7. Hemelaar J. 2012. The origin and diversity of the HIV-1 pandemic. Trends

Mol Med 18:182-192.

8. LANL. 2013. Los Alamos HIV Database: HIV Circulating Recombinant

Forms (CRFs). http://www.hiv.lanl.gov/content/sequence/HIV/CRFs/CRFs. html.

9. Kalish ML, Robbins KE, Pieniazek D, Schaefer A, Nzilambi N, Quinn TC, St Louis ME, Youngpairoj AS, Phillips J, Jaffe HW, Folks TM. 2004.

Recombinant viruses and early global HIV-1 epidemic. Emerg Infect Dis

10:1227-1234.

10. Rambaut A, Robertson DL, Pybus OG, Peeters M, Holmes EC. 2001.

Human immunodeficiency virus. Phylogeny and the origin of HIV-1. Nature

410:1047-1048.

11. Vidal N, Peeters M, Mulanga-Kabeya C, Nzilambi N, Robertson D, Ilunga W, Sema H, Tshimanga K, Bongo B, Delaporte E. 2000. Unprecedented

degree of human immunodeficiency virus type 1 (HIV-1) group M genetic diversity in the Democratic Republic of Congo suggests that the HIV-1 pandemic originated in Central Africa. J Virol 74:10498-10507.

(30)

the human immunodeficiency virus binds to the immunoglobulin-like domain of CD4. Nature 334:159-162.

13. Maddon PJ, Dalgleish AG, McDougal JS, Clapham PR, Weiss RA, Axel R. 1986. The T4 gene encodes the AIDS virus receptor and is expressed in

the immune system and the brain. Cell 47:333-348.

14. Trkola A, Dragic T, Arthos J, Binley JM, Olson WC, Allaway GP, Cheng-Mayer C, Robinson J, Maddon PJ, Moore JP. 1996. CD4-dependent,

antibody-sensitive interactions between HIV-1 and its co-receptor CCR-5. Nature 384:184-187.

15. Wu L, Gerard NP, Wyatt R, Choe H, Parolin C, Ruffing N, Borsetti A, Cardoso AA, Desjardin E, Newman W, Gerard C, Sodroski J. 1996.

CD4-induced interaction of primary HIV-1 gp120 glycoproteins with the chemokine receptor CCR-5. Nature 384:179-183.

16. Deng H, Liu R, Ellmeier W, Choe S, Unutmaz D, Burkhart M, Di Marzio P, Marmon S, Sutton RE, Hill CM, Davis CB, Peiper SC, Schall TJ, Littman DR, Landau NR. 1996. Identification of a major co-receptor for primary

isolates of HIV-1. Nature 381:661-666.

17. Feng Y, Broder CC, Kennedy PE, Berger EA. 1996. HIV-1 entry cofactor:

functional cDNA cloning of a seven-transmembrane, G protein-coupled receptor. Science 272:872-877.

18. Chan DC, Kim PS. 1998. HIV entry and its inhibition. Cell 93:681-684.

19. Miller MD, Farnet CM, Bushman FD. 1997. Human immunodeficiency virus

type 1 preintegration complexes: studies of organization and composition. J Virol 71:5382-5390.

20. Kohl NE, Emini EA, Schleif WA, Davis LJ, Heimbach JC, Dixon RA, Scolnick EM, Sigal IS. 1988. Active human immunodeficiency virus

protease is required for viral infectivity. Proc Natl Acad Sci U S A

85:4686-4690.

21. Nguyen DH, Hildreth JE. 2000. Evidence for budding of human

immunodeficiency virus type 1 selectively from glycolipid-enriched membrane lipid rafts. J Virol 74:3264-3272.

22. Engelman A, Cherepanov P. 2012. The structural biology of HIV-1:

mechanistic and therapeutic insights. Nat Rev Microbiol 10:279-290.

23. Garcia-Blanco MA, Cullen BR. 1991. Molecular basis of latency in

(31)

1

24. Chun TW, Stuyver L, Mizell SB, Ehler LA, Mican JA, Baseler M, Lloyd AL, Nowak MA, Fauci AS. 1997. Presence of an inducible HIV-1 latent

reservoir during highly active antiretroviral therapy. Proc Natl Acad Sci U S A

94:13193-13197.

25. Finzi D, Hermankova M, Pierson T, Carruth LM, Buck C, Chaisson RE, Quinn TC, Chadwick K, Margolick J, Brookmeyer R, Gallant J, Markowitz M, Ho DD, Richman DD, Siliciano RF. 1997. Identification of a

reservoir for HIV-1 in patients on highly active antiretroviral therapy. Science

278:1295-1300.

26. Wong JK, Hezareh M, Gunthard HF, Havlir DV, Ignacio CC, Spina CA, Richman DD. 1997. Recovery of replication-competent HIV despite

prolonged suppression of plasma viremia. Science 278:1291-1295.

27. FDA. 2013. U.S. Food and Drug Administration: Antiretroviral drugs used

in the treatment of HIV infection, http://www.fda.gov/ForConsumers/ byAudience/ForPatientAdvocates/HIVandAIDSActivities/ucm118915.htm. 28. Fischl MA, Richman DD, Hansen N, Collier AC, Carey JT, Para MF, Hardy

WD, Dolin R, Powderly WG, Allan JD, et al. 1990. The safety and efficacy

of zidovudine (AZT) in the treatment of subjects with mildly symptomatic human immunodeficiency virus type 1 (HIV) infection. A double-blind, placebo-controlled trial. The AIDS Clinical Trials Group. Ann Intern Med

112:727-737.

29. Volberding PA, Lagakos SW, Koch MA, Pettinelli C, Myers MW, Booth DK, Balfour HH, Jr., Reichman RC, Bartlett JA, Hirsch MS, et al. 1990.

Zidovudine in asymptomatic human immunodeficiency virus infection. A controlled trial in persons with fewer than 500 CD4-positive cells per cubic millimeter. The AIDS Clinical Trials Group of the National Institute of Allergy and Infectious Diseases. N Engl J Med 322:941-949.

30. Larder BA, Darby G, Richman DD. 1989. HIV with reduced sensitivity to

zidovudine (AZT) isolated during prolonged therapy. Science

243:1731-1734.

31. Larder BA, Kemp SD. 1989. Multiple mutations in HIV-1 reverse transcriptase

confer high-level resistance to zidovudine (AZT). Science 246:1155-1158.

32. Johnson VA, Hirsch MS. 1990. New developments in antiretroviral drug

therapy for human immunodeficiency virus infections. AIDS Clin Rev

(32)

33. Johnson VA, Hirsch MS. 1990. New developments in combination

chemotherapy of anti-human immunodeficiency virus drugs. Ann N Y Acad Sci 616:318-327.

34. Collier AC, Coombs RW, Schoenfeld DA, Bassett RL, Timpone J, Baruch A, Jones M, Facey K, Whitacre C, McAuliffe VJ, Friedman HM, Merigan TC, Reichman RC, Hooper C, Corey L. 1996. Treatment of

human immunodeficiency virus infection with saquinavir, zidovudine, and zalcitabine. AIDS Clinical Trials Group. N Engl J Med 334:1011-1017.

35. D’Aquila RT, Hughes MD, Johnson VA, Fischl MA, Sommadossi JP, Liou SH, Timpone J, Myers M, Basgoz N, Niu M, Hirsch MS. 1996. Nevirapine,

zidovudine, and didanosine compared with zidovudine and didanosine in patients with HIV-1 infection. A randomized, double-blind, placebo-controlled trial. National Institute of Allergy and Infectious Diseases AIDS Clinical Trials Group Protocol 241 Investigators. Ann Intern Med 124:1019-1030.

36. Palella FJ, Jr., Delaney KM, Moorman AC, Loveless MO, Fuhrer J, Satten GA, Aschman DJ, Holmberg SD. 1998. Declining morbidity and

mortality among patients with advanced human immunodeficiency virus infection. HIV Outpatient Study Investigators. N Engl J Med 338:853-860.

37. WHO/UNAIDS. 2003. Treat 3 million by 2005 initiative. http://www.who.

int/3by5/publications/documents/en/3by5StrategyMakingItHappen.pdf. 38. United Nations General Assembly 2011 Political Declaration on HIV and

AIDS: Intensidying Our Efforts to Eliminate HIV and AIDS. New York, United Nations, 2011.

39. WHO/UNAIDS/UNICEF. 2013. Global update on HIV treatment 2013:

results, impact and opportunities http://www.unaids.org/en/media/unaids/ contentassets/documents/unaidspublication/2013/20130630_treatment_ report_en.pdf.

40. WHO. 2013. Consolidated guidelines on the use of antiretroviral drugs for treating

and preventing HIV infection: recommendations fora public health approach. http://apps.who.int/iris/bitstream/10665/85321/1/9789241505727_eng.pdf. 41. WHO. 2010. Antiretroviral therapy for HIV infection in adults and adolescents:

recommendations for a public health approach 2010 revision. http:// whqlibdoc.who.int/publications/2010/9789241599764_eng.pdf.

42. Drake JW, Holland JJ. 1999. Mutation rates among RNA viruses. Proc Natl

(33)

1

43. Onafuwa-Nuga A, Telesnitsky A. 2009. The remarkable frequency of

human immunodeficiency virus type 1 genetic recombination. Microbiol Mol Biol Rev 73:451-480, Table of Contents.

44. Ramirez BC, Simon-Loriere E, Galetto R, Negroni M. 2008. Implications

of recombination for HIV diversity. Virus Res 134:64-73.

45. Ho DD, Neumann AU, Perelson AS, Chen W, Leonard JM, Markowitz M. 1995. Rapid turnover of plasma virions and CD4 lymphocytes in HIV-1

infection. Nature 373:123-126.

46. Perelson AS, Neumann AU, Markowitz M, Leonard JM, Ho DD. 1996.

HIV-1 dynamics in vivo: virion clearance rate, infected cell life-span, and viral generation time. Science 271:1582-1586.

47. Eigen M. 1993. Viral quasispecies. Sci Am 269:42-49.

48. Kearney M, Palmer S, Maldarelli F, Shao W, Polis MA, Mican J, Rock-Kress D, Margolick JB, Coffin JM, Mellors JW. 2008. Frequent

polymorphism at drug resistance sites in HIV-1 protease and reverse transcriptase. Aids 22:497-501.

49. Ribeiro RM, Bonhoeffer S, Nowak MA. 1998. The frequency of resistant

mutant virus before antiviral therapy. Aids 12:461-465.

50. Johnson VA, Calvez V, Gunthard HF, Paredes R, Pillay D, Shafer R, Wensing AM, Richman DD. 2013. Update of the drug resistance mutations

in HIV-1: March 2013. Top HIV Med 21:6-14.

51. WHO. 2012. WHO HIV drug resistance report 2012. http://apps.who.int/iris/

bitstream/10665/75183/1/9789241503938_eng.pdf.

52. Hamers RL, Wallis CL, Kityo C, Siwale M, Mandaliya K, Conradie F, Botes ME, Wellington M, Osibogun A, Sigaloff KCE, Nankya I, Schuurman R, Wit FW, Stevens W, van Vugt M, Rinke de Wit TF. 2011.

HIV-1 drug resistance among antiretroviral-naive individuals in sub-Saharan Africa after rollout of antiretroviral therapy: multicentre observational study. Lancet Infect Dis 11:750-759.

53. Price MA, Wallis CL, Lakhi S, Karita E, Kamali A, Anzala O, Sanders EJ, Bekker LG, Twesigye R, Hunter E, Kaleebu P, Kayitenkore K, Allen S, Ruzagira E, Mwangome M, Mutua G, Amornkul PN, Stevens G, Pond SL, Schaefer M, Papathanasopoulos MA, Stevens W, Gilmour J. 2011.

Transmitted HIV type 1 drug resistance among individuals with recent HIV infection in East and Southern Africa. AIDS Res Hum Retroviruses 27:5-12.

(34)

54. Deeks SG, Barbour JD, Grant RM, Martin JN. 2002. Duration and

predictors of CD4 T-cell gains in patients who continue combination therapy despite detectable plasma viremia. Aids 16:201-207.

55. Pantaleo G, Graziosi C, Fauci AS. 1993. New concepts in the

immunopathogenesis of human immunodeficiency virus infection. N Engl J Med 328:327-335.

56. 2012. 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. 1-239. Available

from: <http://www.aidsinfo.nih.gov/ContentFiles/AdultandAdolescentGL. pdf>.

57. Mee P, Fielding KL, Charalambous S, Churchyard GJ, Grant AD. 2008.

Evaluation of the WHO criteria for antiretroviral treatment failure among adults in South Africa. Aids 22:1971-1977.

58. Moore DM, Awor A, Downing R, Kaplan J, Montaner JS, Hancock J, Were W, Mermin J. 2008. CD4+ T-cell count monitoring does not accurately

identify HIV-infected adults with virologic failure receiving antiretroviral therapy. J Acquir Immune Defic Syndr 49:477-484.

59. Reynolds SJ, Nakigozi G, Newell K, Ndyanabo A, Galiwongo R, Boaz I, Quinn TC, Gray R, Wawer M, Serwadda D. 2009. Failure of immunologic

criteria to appropriately identify antiretroviral treatment failure in Uganda. Aids 23:697-700.

60. Sigaloff KC, Hamers RL, Wallis CL, Kityo C, Siwale M, Ive P, Botes ME, Mandaliya K, Wellington M, Osibogun A, Stevens WS, van Vugt M, de Wit TF. 2011. Unnecessary antiretroviral treatment switches and

accumulation of HIV resistance mutations; two arguments for viral load monitoring in Africa. J Acquir Immune Defic Syndr 58:23-31.

61. van Oosterhout JJ, Brown L, Weigel R, Kumwenda JJ, Mzinganjira D, Saukila N, Mhango B, Hartung T, Phiri S, Hosseinipour MC. 2009.

Diagnosis of antiretroviral therapy failure in Malawi: poor performance of clinical and immunological WHO criteria. Trop Med Int Health 14:856-861.

62. Gupta RK, Jordan MR, Sultan BJ, Hill A, Davis DH, Gregson J, Sawyer AW, Hamers RL, Ndembi N, Pillay D, Bertagnolio S. 2012. Global trends

in antiretroviral resistance in treatment-naive individuals with HIV after rollout of antiretroviral treatment in resource-limited settings: a global collaborative

(35)

1

study and meta-regression analysis. Lancet 380:1250-1258.

63. Hamers RL, Smit PW, Stevens W, Schuurman R, Rinke de Wit TF. 2009.

Dried fluid spots for HIV type-1 viral load and resistance genotyping: a systematic review. Antivir Ther 14:619-629.

64. Bertagnolio S, Soto-Ramirez L, Pilon R, Rodriguez R, Viveros M, Fuentes L, Harrigan PR, Mo T, Sutherland D, Sandstrom P. 2007.

HIV-1 drug resistance surveillance using dried whole blood spots. Antivir Ther

12:107-113.

65. Harrigan PR, Stone C, Griffin P, Najera I, Bloor S, Kemp S, Tisdale M, Larder B. 2000. Resistance profile of the human immunodeficiency virus

type 1 reverse transcriptase inhibitor abacavir (1592U89) after monotherapy and combination therapy. CNA2001 Investigative Group. J Infect Dis

181:912-920.

66. Hawkins CA, Chaplin B, Idoko J, Ekong E, Adewole I, Gashau W, Murphy RL, Kanki P. 2009. Clinical and genotypic findings in HIV-infected patients

with the K65R mutation failing first-line antiretroviral therapy in Nigeria. J Acquir Immune Defic Syndr 52:228-234.

67. Svarovskaia ES, Margot NA, Bae AS, Waters JM, Goodman D, Zhong L, Borroto-Esoda K, Miller MD. 2007. Low-level K65R mutation in HIV-1

reverse transcriptase of treatment-experienced patients exposed to abacavir or didanosine. J Acquir Immune Defic Syndr 46:174-180.

68. Wallis CL, Mellors JW, Venter WD, Sanne I, Stevens W. 2010. Varied

patterns of HIV-1 drug resistance on failing first-line antiretroviral therapy in South Africa. J Acquir Immune Defic Syndr 53:480-484.

69. Winters MA, Shafer RW, Jellinger RA, Mamtora G, Gingeras T, Merigan TC. 1997. Human immunodeficiency virus type 1 reverse transcriptase

genotype and drug susceptibility changes in infected individuals receiving dideoxyinosine monotherapy for 1 to 2 years. Antimicrob Agents Chemother

41:757-762.

70. Brenner B, Turner D, Oliveira M, Moisi D, Detorio M, Carobene M, Marlink RG, Schapiro J, Roger M, Wainberg MA. 2003. A V106M mutation

in HIV-1 clade C viruses exposed to efavirenz confers cross-resistance to non-nucleoside reverse transcriptase inhibitors. Aids 17:F1-5.

71. Grossman Z, Istomin V, Averbuch D, Lorber M, Risenberg K, Levi I, Chowers M, Burke M, Bar Yaacov N, Schapiro JM. 2004. Genetic

(36)

variation at NNRTI resistance-associated positions in patients infected with HIV-1 subtype C. Aids 18:909-915.

72. Barth RE, van der Loeff MF, Schuurman R, Hoepelman AI, Wensing AM. 2010. Virological follow-up of adult patients in antiretroviral treatment

programmes in sub-Saharan Africa: a systematic review. Lancet Infect Dis

(37)
(38)
(39)

C

A

H

P

T

E

R

Susan C Aitken*, Aletta Kliphuis*, Michelle Bronze, Carole L. Wallis, Cissy Kityo, Sheila Balinda, Wendy Stevens, Nicole Spieker, Tulio de Oliveira, Tobias F. Rinke de Wit and Rob Schuurman.

*These authors contributed equally

of an affordable real-time

qualitative assay for

determining HIV-1 virological

failure in plasma and dried

blood spots

(40)

Abstract

V

irological failure (VF) has been identified as the earliest, most pre-dictive determinant of HIV-1 antiretroviral treatment (ART) failure. Due to high costs and complexity of virological monitoring, VF assays are rarely performed in resource-limited settings (RLS). Rather, ART failure is determined by clinical monitoring and to a large extent immunological monitoring. This paper describes the development and evaluation of a low-cost, dried blood spot (DBS)-compatible qualitative assay to deter-mine VF, in accordance with current WHO guideline recommendations for therapy-switching in RLS. The described assay is an internally-controlled qualitative real-time PCR targeting the conserved long terminal repeat do-main of HIV-1. This assay was applied with HIV-1 subtypes A-H and further evaluated on HIV-1 clinical plasma samples from South Africa (n=191) and Tanzania (n=42). Field evaluation was performed in Uganda using local clinical plasma samples (n=176). Furthermore, assay performance was evaluated for DBS. The described assay is able to identify VF for all major HIV-1 group-M subtypes with equal specificity, and lower detection limit of 1.00E+03 copies/ml for plasma and 5.00E+03 copies/ml for DBS. Com-parative testing yielded accurate VF determination for therapy-switching in 89%-96% of samples compared to gold standards. The assay is robust and flexible, allowing for “open platform” applications and producing com-parable results to commercial assays. Assay design enables application in laboratories that can accommodate real-time PCR equipment, allowing decentralization of testing to some extent. Compatibility with DBS extends access of sampling and thus access to this test to remote settings.

(41)

2

Introduction

I

n 2010 the HIV-1 epidemic was estimated to include 34.0 million (range 31.6-35.2 million) infected adults and children across the globe. An alarm-ing 67.4% (n= 22.9 million) of the total global infections are people residalarm-ing in Sub-Saharan Africa. As a result of antiretroviral therapy (ART) scale-up initiatives, 6.65 million of infected individuals requiring treatment were receiv-ing it in Sub-Saharan Africa by the end of 2010(1). However, particularly in

re-source-limited settings (RLS), effective treatment faces challenges, which in-clude failing supply chains resulting in drug stock-outs, drug toxicity of “older generation” 1st and 2nd line drugs, failure of patient adherence, drug-drug

in-teractions, lack of qualified healthcare staff or failing adherence support, etc. As a result HIV-1 can develop drug resistance to ART, leading to virological failure (VF) and subsequently ART failure. A recent report has shown that the prevalence of pre-ART HIV-1 drug resistance in 13 sites in various countries in Sub-Saharan Africa is 5.6%, ranging from 1.1% in South Africa to 12.3% in Uganda(2). Recent scientific findings have led to the consideration of

“Treat-ment as Prevention”, which according to the most intensive “Test and Treat” scenario could ultimately increase the number of HIV patients qualifying for ART to 32 million(3). With rapidly increasing numbers of HIV patients on ART

in RLS with weak health systems, the risk of further increase of HIV-1 drug resistance is imminent.

The success of increased access to ART in RLS has largely been due to massive donor funding and important reduction of costs of selected first-line drugs. However, reduced susceptibility to these first-first-line drugs and the consequent switching to second-line would at least partly undo early ART successes and result in higher expenditures and increasing numbers of pa-tients on failing regimens with no options for effective second-line or salvage therapies(4, 5).

As per the definition of the WHO, VF is a repeated viral load ≥5.00E+03 RNA copies/ml in an individual taking ART for at least four to six months(6).

(42)

Timely detection of VF by VL testing, which is routine in industrialized coun-tries(7), is necessary to prevent accumulation of HIV drug resistance(8), or to

identify poor adherence to the treatment. However, in RLS, high costs and technical complexity limit the possibility of VL monitoring and treatment fail-ure is primarily determined using clinical monitoring for stage three and four AIDS-defining illnesses and, if available, immunological monitoring using CD4 counts(6). The inadequacies of CD4 for determining true treatment

fail-ure have been described on many occasions (9-12). The clinical-immunological

monitoring approach results in individuals being left on sub-optimal regimens for an extended period of time with the risk of accumulating drug resistance mutations or unnecessary switching to second-line therapy based on non-VL supported decisions(4). Both scenarios limit future treatment options and

increased costs associated with second-line therapy(5).

The current paper addresses the challenge of determination of VF in RLS by taking several premises into consideration that reflect the actual public health situation in these settings. First of all, the standpoint was taken that determination of an exact VL is not required to determine ART failure and therefore a less complex, and thus less expensive, assay that classifies a sample as either above or below a treatment success threshold would suf-fice. Secondly, in order to implement the WHO recommendations of task shifting and decentralization of ART to remote settings, the consequence would be that complex procedures, including drawing blood, isolation and storage of plasma, cold chain shipments to qualified labs for VL testing, should be avoided. Rather, VF should be detectable on dried blood spots (DBS), a sampling alternative that is inexpensive, easy to collect and trans-port, and has proven application for VL testing(13, 14). Thirdly, given the fact

that for accurate detection of VF a nucleic acid amplification step remains necessary and taking into consideration the realities of contamination risks in remote labs, it was decided to concentrate on a real-time PCR approach. This allows for VF determination in a closed system and with equipment that is continuously evolving, regularly reducing in price, and being adapted to

(43)

2

local circumstances through battery and solar energy applications. Finally, it was considered essential that the protocol for VF testing should be generic, “open platform”, applicable on a wide array of real-time PCR instruments in various African settings, and freely available in the public domain.

With the above assumptions in mind, the Affordable Resistance Test for Af-rica (ARTA) consortium was established, consisting of a unique combina-tion of academia, industry and non-government organizacombina-tions both in Africa and Europe. Here we report on the results of ARTA research to develop a real-time PCR assay that can be used as a screening tool to determine VF in RLS. This “virological failure assay” (VFA) can be readily applied in basic laboratories, using either plasma or DBS as the sample input. The VFA is applicable for all major HIV-1 group-M subtypes, and is specifically designed to identify VF as defined by the WHO as a VL of ≥5.00E+03 copies/ml(6). Materials and Methods

Samples

HIV-1 Subtype Reference panel

A panel of virus isolates consisting of HIV-1 subtypes A through H (Table 1) was obtained from BBI (BBI Biotech Research Laboratories Inc., Gaithers-burg, USA) for assay optimization and evaluation at the University Medical Centre in Utrecht (UMCU), The Netherlands. Serial dilutions were prepared from these stocks using HIV-1 negative human plasma. These dilutions were also used to spike HIV-1 negative whole blood for DBS preparation.

Clinical samples

Clinical plasma samples from HIV-1 infected individuals from several African sites were included for further evaluation at the UMCU, the Netherlands. Samples were selected to include several subtypes with a variety of VL in ac-cepted ranges for subsequent analysis. Samples from South Africa (n=191) were plasma samples sent for routine VL testing, performed on the

(44)

CO-BAS®AmpliPrep/COBAS®TaqMan®System v2 (Roche, Penzberg, Germa-ny), and represented HIV-1 subtype-C with a VL range of 1.30E+03-3.00E+06 (median 5.50E+04) copies/ml. Samples from Tanzania (n=42) were part of an ongoing prevention-of-mother-to-child-transmission (PMTCT) study(15),

where VL was determined using the COBAS® Amplicor HIV-1 Monitor test v1.5 (Roche). Samples included subtypes A (n=23), C (n=10), and D (n=6), and three samples with undetermined subtype, with a VL range of 6.65E+02-3.07E+05 (median 2.67E+04) copies/ml.

Table 1. HIV-1 isolated from the BBI subtype reference panel

Subtype Strain Country of Origin Accession Number

A UG275 Uganda AB485632

B BK132 Thailand AY173951

C ZB18 Zambia AB485641

D SE365 Senegal AB485648

CRF01_AE CM240 Thailand AF067154

F BZ126 Brazil AY173957

G BCFDIOUM Zaire AB485661

H BCPKITA Zaire AB485665

In addition, as part of a technology transfer program, the described assay was applied in three Joint Clinical Research Centre (JCRC) laboratory sites in Uganda, where retrospective plasma samples collected from HIV-1 posi-tive individuals as part of the PASER program were included(16). These

sam-ples represented baseline and follow-up clinical samsam-ples at yearly intervals after therapy initiation. For these samples, routine VL had been performed in Uganda using the COBAS®AmpliPrep/COBAS®TaqMan®System v2(Ro-che). A total of 176 plasma samples were included comprising of subtypes A (n=89), D (n=64), and 23 with an unknown subtype, with a VL range of 1.00E+02-1.00E+06 (median 1.00E+04) copies/ml. Twenty-five confirmed HIV-1 negative plasma samples were included for assay specificity control.

(45)

2

To investigate the application of the assay with DBS samples, DBS were prepared from EDTA collected whole blood for participants of the PASER program(16). The same blood sample was then centrifuged and the plasma

was removed for analysis. These samples are subsequently referred to as paired plasma and DBS samples. A total of 82 paired samples were tested in Uganda, with a VL range of 4.40E+01-7.18E+06 (median 2.61E+03) cop-ies/ml. DBS samples were stored at -70°C for 270-515 (median 485) days (n=31), -20°C for 45-112 (median 82) days (n=21), or room temperature for 2-192 (median 126) days (n=30).

Internal Control

An internal control (IC) was added to each clinical sample at a fixed amount of ten percent of the elution volume at the start of nucleic acid isolation. The IC comprised of the non-human RNA virus Encephalomyocarditis virus (EMC) and was prepared at the UMCU, the Netherlands, in batches of sin-gle-use aliquots and stored at -80°C until use.

VFA

Nucleic Acid Isolation, Plasma

At the UMCU, the Netherlands, viral nucleic acid (NA) isolation was per-formed using NucliSENS magnetic extraction reagents in combination with the MINIMAG (BioMérieux, Boxtel, The Netherlands). For each sample, an input of 100μl plasma was used, or two DBS of 50μl each, and 2.5μl IC. Pos-itive and negative controls were included in each run. Upon completion of the isolation procedure, purified nucleic acids were eluted in 25μl elution buffer. In Uganda, NA isolation was performed using the QIAamp RNA kit (Qia-gen GmbH, Germany), as per manufactures instructions. Input was 100μl of plasma, or two 50 μl DBS, and 5μl IC. Isolated NA was eluted in 50μl elution buffer.

(46)

Upon completion of both isolation procedures, the eluates were used imme-diately for reverse transcription (RT), and remaining nucleic acids stored at -20°C.

Nucleic Acid Isolation, DBS

At both sites, a pre-incubation step for DBS was performed. DBS samples where excised by hand using scissors, which were decontaminated between samples with 70% ethanol. For the Nuclisens method, DBS were placed in the provided 2ml lysis buffer (BioMérieux) in a 9ml tube. For the QIAamp RNA method (Qiagen GmbH), DBS were placed in 700ul of the provided Buffer AVL lysis buffer that was aliquoted in 2ml Eppendorf tubes for use. For both methods, samples were incubated at room temperature with gentle rotation for 30 minutes, after which filters were removed and NA isolated as per the plasma samples.

Reverse Transcription

Purified NA, containing both HIV-1 RNA and IC RNA, was reverse transcribed using the TaqMan® Real-time PCR system Random Hexamers RT kit (Life Technologies, Foster City, CA) according to manufactures instructions. An in-put of 10μl NA isolate was used in a final reaction volume of 25μl. Reactions were carried out according to the following conditions: 25C° for 10 minutes, 48°C for 30 minutes and 95°C for 5 minutes. Generated cDNA was used immediately for real-time PCR or stored at 4°C.

Real-time PCR

HIV-1 and IC cDNA fragments were amplified in multiplex format. A 25μl real-time PCR reaction contained 12.5μl universal TaqMan® Master Mix (Life Technologies), 10μl cDNA, 300nM primer EMC-forward, 900nM primer

EMC-reverse, 100nM probe EMC-VIC, 300nM forward primer LTR S4, a

mix-ture of 600nM HIV-LTR revere primers 3’UNI-KS-6 and 3’UNI-KS-6-AG, and 100nM MGB probe HIV-LTR-FAM (Table 2). The assay was performed at the UMCU, the Netherlands, using an Applied Biosystem 7500 Real-Time PCR

(47)

2

System (Life Technologies), and in Uganda using a MiniOpticonTM Real-Time

PCR Detection System (BioRad, Hercules, CA). Both systems included a temperature profile allowing for dUTP/UNG decontamination, namely 50°C for 2 minutes; 95°C for 10 minutes; 45 cycles of 95°C for 15 seconds and 60°C for 1 minute. In order to enable run-to-run comparison, a fixed thresh-old was established for both systems (data not shown).

Assay Controls

Positive and negative controls were included in each run. Appropriate per-formance of the run was judged based on the results of these controls. The positive control consisted of a plasma sample spiked with a fixed concentra-tion of 2.50E+04 copies/ml HIV-1, the Ct value acceptance range of which was determined for each real-time system. For this evaluation the positive control range was set at Ct 29-32. The IC was used to monitor for inhibition of each individual sample. As with the positive control, the Ct value acceptance range of the IC was determined for each real-time system. For this evalua-tion the positive control range was set at Ct 30-33. Three negative controls were included, an isolation negative that consisted of HIV-1 negative human plasma and IC, and negative RT and PCR controls which consisted of nucle-ase-free water and no IC. The result obtained for a sample was considered valid when the positive and IC controls were within their predetermined rang-es, and the negative controls were below detection.

Data Analysis

Assay Range

A five-fold serial dilution series of viral RNA for plasma and DBS for all panel subtypes (Table 1) was used to assess dynamic range, level of detection (LOD), and inter- and intra-assay reproducibility. For the ABI7500 (Life Tech-nologies) the serial dilution series ranged from 5.00E+06-3.20E+02 copies/ ml, and for the MiniOpticon (BioRad) from 1.00E+06-1.6E+03 copies/ml. Lin-earity was determined and reported as a R2 value and slope gradient.

(48)

Table 2.

Primer and probe sequences for the HIV

-1 V

irological Failure screening

Assay . Primer/Probe Sequence Function Target EMC-forward 5’-TGACCACGCCACCGC-3’ Forward primer EMC EMC-reverse 5’-T AAAGA TTTCCCTTGCCCCG-3’ Reverse primer EMC EMC-VIC 5’-TGTGAGCCAGTCGTGA TTGTGCTCC-3’ Probe EMC HIV -L TR S4 5’-AAGCCTCAA TAAAGCTTGCCTTGA-3’ Forward primer HXB2 nt520-543 3’UNI-KS-6 5’-GAGGGA TCTCT AGTT ACCAGAGTCACA-3’ Reverse primer HXB2 nt574-600 3’UNI-KS-6-AG 5’-GAGGGA TCTCT AGTT ACCAGAGTCCT A-3’ HIV -L TR-F AM 5’-T AGTGTGTGCCCGTCTG-3’ MGB probe HXB2 nt554-570

EMC: Encephalomyocarditis virus (internal control): L

TR: long terminal repeat region of HIV

-1; HXB2: HIV

Referenties

GERELATEERDE DOCUMENTEN

Zeerr graag wil ik mijn lieve moeder en, in dierbare herinnering, mijn vader, bedan- kenn voor alle mogelijkheden die zij mij hebben geboden, de liefde en toewijding waarmeee zij

Na het artsexamen in augustus 1997 werd de opleiding tot Maag-Darm- Leverartss gestart op de afdeling Inwendige Geneeskunde van het Kennemer Gasthuiss in Haarlem (opleider Dr

It is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), other than for strictly

Many healthcare professionals have relationships with industry. The increasing role of patient organizations in all aspects of patient care has also enhanced their interactions

Because children with SLI also have problems in information processing, as became clear in §4.4, this might be an extra difficulty in the RCC-task (these possibilities will

Article 48 of the GDPR provides that, unless oth- erwise authorized by EU law, a court order requiring the transfer of personal data to a location outside the European Union must

Naumov describe the rise of Nikolai Yezhov, the organizer of the Soviet Union’s Great Terror of 1937–38, leading to the arrest of more than 1.5 million people, almost half of whom

Stoppen met roken, integrale zorg, en het gebruik van medicijnen zorgen op verschillende manieren voor een hogere kwaliteit van leven van astma- en COPD-patiënten..