ArticlePDF Available

Primary resistance to integrase strand transfer inhibitors in Spain using ultrasensitive HIV-1 genotyping

Authors:

Abstract and Figures

Background: Transmission of resistance mutations to integrase strand transfer inhibitors (INSTIs) in HIV-infected patients may compromise the efficacy of first-line antiretroviral regimens currently recommended worldwide. Continued surveillance of transmitted drug resistance (TDR) is thus warranted. Objectives: We evaluated the rates and effects on virological outcomes of TDR in a 96 week prospective multicentre cohort study of ART-naive HIV-1-infected subjects initiating INSTI-based ART in Spain between April 2015 and December 2016. Methods: Pre-ART plasma samples were genotyped for integrase, protease and reverse transcriptase resistance using Sanger population sequencing or MiSeq™ using a ≥ 20% mutant sensitivity cut-off. Those present at 1%-19% of the virus population were considered to be low-frequency variants. Results: From a total of 214 available samples, 173 (80.8%), 210 (98.1%) and 214 (100.0%) were successfully amplified for integrase, reverse transcriptase and protease genes, respectively. Using a Sanger-like cut-off, the overall prevalence of any TDR, INSTI-, NRTI-, NNRTI- and protease inhibitor (PI)-associated mutations was 13.1%, 1.7%, 3.8%, 7.1% and 0.9%, respectively. Only three (1.7%) subjects had INSTI TDR (R263K, E138K and G163R), while minority variants with integrase TDR were detected in 9.6% of subjects. There were no virological failures during 96 weeks of follow-up in subjects harbouring TDR as majority variants. Conclusions: Transmitted INSTI resistance remains rare in Spain and, to date, is not associated with virological failure to first-line INSTI-based regimens.
Content may be subject to copyright.
Primary resistance to integrase strand transfer inhibitors in Spain
using ultrasensitive HIV-1 genotyping
M. Casadella`
1
†, J. R. Santos
2
*†, M. Noguera-Julian
1
, R. Mica´n-Rivera
3
, P. Domingo
4
, A. Antela
5
, J. Portilla
6
,
J. Sanz
7
, M. Montero-Alonso
8
, J. Navarro
9
, M. Masia´
10
, N. Valcarce-Pardeiro
11
, A. Ocampo
12
,
L. Pe´rez-Martı´nez
13
, J. Pasquau
14
, M. J. Vivancos
15
,A.Imaz
16
, P. Carmona-Oyaga
17
,L.Mu
~
noz-Medina
18
,
J. Villar-Garcı´a
19
, P. Barrufet
20
and R. Paredes
1,2
on behalf of the INSTINCT Study Group‡
1
IrsiCaixa AIDS Research Institute, Badalona, Catalonia, Spain;
2
Lluita contra la SIDA Foundation, Hospital Universitari Germans Trias i
Pujol, Badalona, Spain;
3
University Hospital La Paz, Madrid, Spain;
4
Infectious Diseases Unit, Hospital de la Santa Creu i Sant Pau,
Barcelona, Spain;
5
Infectious Diseases Unit, Santiago de Compostela Clinical University Hospital, Santiago de Compostela, Spain;
6
Hospital General Universitario de Alicante, Alicante, Spain;
7
University Hospital de La Princesa, Madrid, Spain;
8
Infectious Diseases
Unit, La Fe University and Polytechnic Hospital, Valencia, Spain;
9
Infectious Diseases Department, Hospital Universitari Vall d’Hebron,
Barcelona, Spain;
10
Infectious Diseases Unit, Elche University General Hospital, Elche, Spain;
11
Infectious Diseases Unit, Hospital
Arquitecto Marcide, Ferrol, Spain;
12
HIV Unit, Hospital A
´lvaro Cunqueiro, Vigo, Spain;
13
Infectious Diseases Area, Hospital San Pedro-
CIBIR, Logro~
no, Spain;
14
University Hospital Virgen de las Nieves, Granada, Spain;
15
Infectious Diseases Unit, Ramo´ n y Cajal Hospital,
Madrid, Spain;
16
HIV and STI Unit, Infectious Diseases Department, Bellvitge University Hospital, Bellvitge Biomedical Research
Institute (IDIBELL), Hospitalet de Llobregat, Spain;
17
Infectious Diseases Unit, Donostia University Hospital, San Sebastia´ n, Spain;
18
University Hospital San Cecilio, Granada, Spain;
19
Infectious Diseases Department, Hospital del Mar IMIM, Barcelona, Spain;
20
Infectious Diseases Unit, Mataro´ Hospital, Mataro´ , Spain
*Corresponding author. E-mail: jrsantos@flsida.org
†These authors contributed equally.
‡Members are listed in the Acknowledgements section.
Received 2 April 2020; accepted 3 July 2020
Background: Transmission of resistance mutations to integrase strand transfer inhibitors (INSTIs) in HIV-
infected patients may compromise the efficacy of first-line antiretroviral regimens currently recommended
worldwide. Continued surveillance of transmitted drug resistance (TDR) is thus warranted.
Objectives: We evaluated the rates and effects on virological outcomes of TDR in a 96 week prospective multi-
centre cohort study of ART-naive HIV-1-infected subjects initiating INSTI-based ART in Spain between April 2015
and December 2016.
Methods: Pre-ART plasma samples were genotyped for integrase, protease and reverse transcriptase resistance
using Sanger population sequencing or MiSeq
TM
using a 20% mutant sensitivity cut-off. Those present at
1%–19% of the virus population were considered to be low-frequency variants.
Results: From a total of 214 available samples, 173 (80.8%), 210 (98.1%) and 214 (100.0%) were successfully
amplified for integrase, reverse transcriptase and protease genes, respectively. Using a Sanger-like cut-off, the
overall prevalence of any TDR, INSTI-, NRTI-, NNRTI- and protease inhibitor (PI)-associated mutations was
13.1%, 1.7%, 3.8%, 7.1% and 0.9%, respectively. Only three (1.7%) subjects had INSTI TDR (R263K, E138K and
G163R), while minority variants with integrase TDR were detected in 9.6% of subjects. There were no virological
failures during 96 weeks of follow-up in subjects harbouring TDR as majority variants.
Conclusions: Transmitted INSTI resistance remains rare in Spain and, to date, is not associated with virological
failure to first-line INSTI-based regimens.
Introduction
Integrase strand transfer inhibitors (INSTIs) are the current key-
stones of ART due to their high efficacy, safety and tolerability.
13
The first-generation INSTIs, raltegravir and elvitegravir, were the
preferred drugs for ART initiation until 2017.
4,5
Dolutegravir and,
more recently, bictegravir have replaced them as the preferred
drugs for naive patients,
2,3
and are also suitable options for salvage
V
CThe Author(s) 2020. Published by Oxford University Press on behalf of the British Society for Antimicrobial Chemotherapy. All rights reserved.
For permissions, please email: journals.permissions@oup.com.
3517
J Antimicrob Chemother 2020; 75: 3517–3524
doi:10.1093/jac/dkaa349 Advance Access publication 15 September 2020
Downloaded from https://academic.oup.com/jac/article/75/12/3517/5905666 by guest on 12 April 2023
therapy, even in subjects with previous failure or some degree of
resistance to raltegravir or elvitegravir failure.
69
Virological failure (VF) to first-generation INSTI-based regimens
is often associated with selection of INSTI-resistant HIV-1, which
can be transmitted and potentially impair the clinical and virologic-
al outcomes of INSTI-based ART in drug-naive individuals.
1012
However, although anecdotal cases of INSTI resistance transmis-
sion have been reported,
13,14
the prevalence of INSTI-associated
mutations in ART-naive subjects has remained low to date.
1519
Given the increasing global use of INSTI-containing regimens,
including raltegravir, elvitegravir, dolutegravir, bictegravir and soon
cabotegravir, and the significant degree of cross-resistance among
different INSTIs, it is essential to perform continuous surveillance
of integrase transmitted drug resistance (TDR) as well as to moni-
tor its clinical impact.
In this study, we evaluated the prevalence of integrase-
associated mutations in naive patients from clinical practice start-
ing INSTI-based regimens in Spain between 2015 and 2016, and
the rate of VF in patients with transmitted INSTI mutations.
Materials and methods
Study design and data collection
The INSTINCT cohort study is a prospective, 96 week observational study
involving a cohort of995 HIV-positive adult subjects who initiated raltegra-
vir, elvitegravir or dolutegravir between 1 April 2015 and 5 October 2016 in
19 HIV care centres across Spain. Patients were included if they satisfied
the inclusion criteria for one of the following groups: (i) ART-naive, i.e. previ-
ously untreated subjects at the time of INSTI initiation; (ii) ART switch, i.e.
ART-experienced individuals initiating the new INSTI with a viral load (VL)
50 copies/mL; and (iii) ARTsalvage, i.e. ART-experienced patientsinitiating
the new INSTI with a VL >200 copies/mL, including patients who were on a
voluntary ART interruption. Data were collected according to a pre-defined
schedule on the date of raltegravir, elvitegravir or dolutegravir initiation
(baseline) and every 6 months thereafter up to 2 years, in accordance with
the study protocol. The study collected information on demographic char-
acteristics, AIDS stage at the time of INSTI initiation, HCV and HBV coinfec-
tions, HIV-1 RNA levels, CD4 cell counts, CD4 count nadir at baseline and
genotypic test results performed before raltegravir, elvitegravir and dolute-
gravir initiation(when available) and at VF.
In the current analysis, we evaluated the percentage of TDR to INSTIs
and other drug classes in ART-naive patients as well as the rates of VF in
subjects with TDR to INSTIs. VF was defined as two consecutive HIV-1 RNA
measurements 200 copies/mL after 6 months while receiving raltegravir,
elvitegravir or dolutegravir, and at least 3 monthsafter INSTI initiation.
Ethics
Prior approval was given by the Ethics Committee (EPA-15-006) of each
participating centre for the study, which was performed following the stipu-
lations of the Declaration of Helsinki (Brazil, 2013). All patients provided
their written informedconsent at study enrolment.
Virological analyses
Baseline plasma samples of subjects from 13 different Spanish hospitals
were shipped to the IrsiCaixa laboratory (Badalona, Spain) for ultrasensitive
genotyping using next-generation sequencing (NGS). Local performance of
Sanger population sequencing was allowed in cases where there were
issues of storage or shipping.
Given the current absence of a consensus list of transmitted INSTI
mutations, we considered as INSTI resistance mutations those included in
the IAS-USA 2019 list
20
or in the Stanford University HIV drug resistance
database (HIVdb, version 8.9 available at: https://hivdb.stanford.edu/)
with a scoreof >14 to at least one INSTI. For protease (PR) and reverse tran-
scriptase (RT) mutations, the WHO 2009 list was used to determine the
prevalence of primaryresistance.
21
We defined two viral population sensitivity thresholds: mutations
detected by Sanger sequencing or present at 20% of the virus population
by NGS (Sanger-like sequencing) were regarded as high-frequency variants;
those present at 1%–19% of the virus population were considered low-
frequency variants. High-frequency variants were considered in order to
evaluate the prevalence of TDR mutations in our study.
In addition, the number of ‘active’ drugs prescribed at baseline was
evaluated by means of the HIVdb Genotypic Susceptibility Score (GSS),
considering a score of 0–9 as susceptible (1 point), 10–59 as intermediate
(0.5 points) and 60 as resistant (0 points).
For NGS, amplification and sequencing steps were performed as
previously described by Inzaule et al.
22
In brief, viral RNA was extracted
from plasma samples using the QIAmp Viral RNA Mini Kit (Qiagen Inc.,
Chatsworth, CA, USA), following the manufacturer’s protocol. RNA was
thereafter retro-transcribed and amplified, generating an amplicon of the
HIV-1 pol gene by a one-step reaction using the SuperScript
TM
III One-Step
RT–PCR System with Platinum Taq DNA Polymerase (ThermoFisher
Scientific Inc., Waltham, MA, USA), followed by a nested PCR using
Platinum
TM
Taq DNA Polymerase HighFidelity (ThermoFisher Scientific Inc.).
Once amplified, DNA was purified using AMPure XP Beads (Beckman Coulter
Inc., Brea, CA, USA) and quantified in duplicate using the Quant-iT
TM
PicoGree0060045
TM
dsDNA Assay Kit (ThermoFisher Scientific Inc.). It was
then diluted and prepared for sequencing using the Nextera XT DNA
Sample Preparation Kit and Nextera XT Index Kit (Illumina, San Diego, CA,
USA). Finally, samples were pooled and sequenced with a 500 cycle MiSeq
ReagentKit v0.2 (Illumina).
MiSeq sequences (FastQ files) were downloaded from Basespace
(Illumina) and analysed using PASeq v1.4 (https://paseq.org) (IrsiCaixa,
Barcelona, Spain). Briefly, data were quality filtered using Trimmomatic
(v0.30),
23
with a Q25/5bp minimum sliding window and a 70 bp minimum
length. Non-viral contamination was filtered out using BBsplit (v35.76).
Filtered reads were then merged with PEAR (v0.9.6) aligned to reference
sequence using Bowtie2 (v2.1.0).
24
Amino acid variants were then called at
the codon level using Perl code, and these data were used to query the
Stanford HIVdb for resistance interpretation. A consensus sequence was
built using major nucleotide readoutat each position.
Results
A total of 241 naive subjects were included in the study. Of these,a
genotype was requested in 232. Reliable results by NGS were
obtained for 146 samples, whereas Sanger sequencing was locally
available for 68 subjects. In Sanger-sequenced samples, the inte-
grase gene was analysed in only 27 (39.7%), while the RT and PR
genes were evaluated in 64 (94.1%) and 68 (100.0%) samples, re-
spectively. In summary, out of a total of 214 available genotypes,
173 (80.8%), 210 (98.1%) and 214 (100.0%) contained informa-
tion for the integrase, RT and PR genes, respectively (a flow chart of
genotyped samples is detailed in Figure 1). Subtype B was the
most common HIV-1 subtype found (158/214, 73.8% of subjects).
Subjects characteristics are summarized in Table 1.
Considering only high-frequency variants, the overall rate of
TDR was 13.1% (28/214), if we only considered subjects with com-
plete genotypic information on integrase, RT and PR. Respectively,
INSTI-, NRTI-, NNRTI- and PI-associated resistance mutations
were detected in 3/173 (1.7%), 8/210 (3.8%), 15/210 (7.1%) and
2/214 (0.9%) subjects (Figure 2a). In addition, 37/146 (25.3%)
Casadella` et al.
3518
Downloaded from https://academic.oup.com/jac/article/75/12/3517/5905666 by guest on 12 April 2023
samples evaluated by NGS had low-frequency TDR mutations
(Figure 2a). The mutations found are listed inTable S1 (available as
Supplementary data at JAC Online).
INSTI resistance
Only 3/173 (1.7%) individuals had transmitted mutations with
expected impact on INSTI susceptibility, E138K, R263K and
G163R, respectively (Figure 2a). Polymorphic mutations were
found in another 5/173 (2.8%) subjects [T97A in three samples;
L74I and L74M in another two (Table S1)]. All subjects carried
only one INSTI mutation, except for one individual who simul-
taneously revealed T97A (>99% of viruses) and R263K (1.9% of
viruses) mutations.
Low-frequency INSTI-associated mutations were found in
14/146 (9.6%) subjects (Figure 2a). Only two subjects (1.4%)
with low-frequency Q148H and R263K variants, respectively,
were detected (Figure 2b). No patients with N155H or Y143R
were found. Combinations of majority and minority INSTI muta-
tions were found in three (2.1%) individuals analysed by NGS,
including G163R (2.3%) !Q95K (2.3%), E138K (2.8%) !S230R
(1.0%) and the previously described T97A (>99%) !R263K
(1.9%).
NNRTI resistance
K103N was the most frequently detected NNRTI mutation, being
found as the majority variant in 10/210 (4.8%) samples. All the
other NNRTI-associated mutations only accounted for 2.4% of
transmitted NNRTI mutations (Figure 2b).
A total of 6/146 (4.1%) samples had low-frequency NNRTI-
associated mutations, two of which had K103S and Y188C
mutations in >5%–20% of their viral population while four had
Figure 1. Flow chart of baseline samples for HIV genotypes. NGS, next-
generation sequencing; INI, integrase; RT, reverse transcriptase; PR, pro-
tease. This figure appears in colour in the online version of JAC and in
black and white in the print version of JAC.
Table 1. Patient characteristics (n= 241)
RAL (n= 11) EVG (n= 80) DTG (n= 150) Total (n= 241)
Age (years) 42.0 (28.5–57.5) 33.5 (29.0–41.5) 35.0 (30.0–43.0) 35.0 (29.0–43.0)
HIV-1 RNA (copies/mL) 78 311 (37 060–301 444) 34 669 (7617–127 837) 56063 (17 700–180 068) 51 500 (15 043–174 500)
Gender, n(%)
female 1 (9.1) 8 (10.0) 17 (11.3) 26 (10.8)
male 10 (90.9) 72 (90.0) 133 (88.7) 215 (89.2)
Mode of transmission, n(%)
MSM 4 (36.4) 60 (75.0) 103 (68.7) 167 (69.3)
MSW 4 (36.4) 15 (18.8) 31 (20.7) 50 (20.7)
IDU 1 (9.1) 3 (3.8) 6 (4.0) 10 (4.1)
other/unknown 2 (18.2) 2 (2.5) 10 (6.7) 14 (5.8)
Baseline CD4 cell count, cells/mm
3
454.0 (117.5–515.0) 415.00 (273.0–548.0) 429.0 (295.8–614.5) 429.0 (285.3–599.0)
Nadir CD4 cell count, cells/mm
3
451.0 (123.0–467.0) 374.0 (286.0–520.0) 384.5 (288.3–551.8) 384.0 (281.5–542.5)
C stage of AIDS at diagnosis, n(%) 3 (27.3) 4 (5.0) 9 (6.0) 16 (6.6)
Hepatitis coinfection, n(%)
no/unknown 0 2 (2.5) 0 2 (0.8)
HCV 0 3 (3.8) 2 (1.3) 5 (2.1)
HCV 2 (18.2) 6 (7.5) 9 (6.0) 17 (7.1)
Nucleoside pair, n(%)
TDF or TAF/FTC 5 (83.3) 50 (98) 15 (16.5) 70 (47.3)
ABC/3TC 1 (16.7) 1 (2) 76 (83.5) 78 (52.7)
RAL, raltegravir; EVG, elvitegravir; DTG, dolutegravir; MSM, men who have sex with men; MSW, men who have sex with women; IDU, intravenous drug
use; HCV, hepatitis C virus; HBV, hepatitis B virus; TDF, tenofovir disoproxil fumarate; TAF/FTC, tenofovir alafenamide plus emtricitabine; ABC/3TC, aba-
cavir plus lamivudine.
Data are presented as median (IQR) unless otherwise stated.
Primary INSTI resistance in Spain JAC
3519
Downloaded from https://academic.oup.com/jac/article/75/12/3517/5905666 by guest on 12 April 2023
other NNRTI-associated mutations in 1%–5% of their viral popula-
tion (Figure 2b).
NRTI resistance
NRTI-associated mutations were detected as majority variants
in 8/210 (3.8%) samples, and in another 8/146 (5.5%) subjects
as low-frequency variants (Figure 2b). The most frequent muta-
tion was M41L, which was detected as the majority variant in 3/
210 (1.4%) individuals. Other NRTI-associated mutations found
as majority and low-frequency variants are detailed in
Figure 2b.
PI resistance
Only 2/214 (0.9%) subjects had transmitted PI mutations
(L90M and M46L, respectively) as majority variants. Additional
low-frequency PI-resistant mutants included in the WHO 2009
surveillance mutation list were found in 10/146 (6.8%) subjects
(Figure 2b).
GSSs
Considering only subjects with the integrase gene successfully
analysed and mutations found as majority variants, virtually all
viruses showed full susceptibility to INSTI-based regimens. Only
one subject harbouring L74I (GSS = 2 points) started raltegravir !
abacavir/lamivudine (Figure 3). Of subjects undergoing dolutegra-
vir/lamivudine dual therapy, only 2/173 (1.15%) had a GSS 1.5
points (each harbouring mutations E138K and R263K in 99.8% of
their viral population, respectively).
Virological outcomes of TDR
TDR did not affect the outcomes of first-line INSTI-based ART, since
none of the subjects harbouring transmitted INSTI-, NNRTI-, NRTI-
or PI-associated mutations experienced VF during the 96 weeks
of follow-up.
Discussion
This study found a very low rate of transmitted INSTI resistance
mutations in naive patients starting INSTI-based regimens in
Figure 2. Patients with transmitted HIV mutations according to viral population. (a) Percentage of patients with transmitted HIV mutations according
to antiretroviral classes. This represents the percentage of patients with TDR detected according to the viral population and the antiretroviral class
affected. (b) Number of patients with mutations in the integrase, protease and reverse transcriptase genes. For integrase, those mutations included
in the IAS-USA 2019 list or in the Stanford University HIV drug resistance database (HIVdb, version 8.9, available at: https://hivdb.stanford.edu/) with
a score >14 for at least one INSTI were considered. For reverse transcriptase and protease, those mutations included in the WHO 2009 surveillance
mutation list were considered. WHO 2009 non-listed mutations were not represented. Percentages of patients harbouring mutations were calculated
based on the number of successfully amplified samples for integrase, reverse transcriptase and protease genes. This figure appears in colour in the
online version of JAC and in black and white in the print version of JAC.
Casadella` et al.
3520
Downloaded from https://academic.oup.com/jac/article/75/12/3517/5905666 by guest on 12 April 2023
Spain. Moreover, TDR had absolutely no impact on the effective-
ness of first-line INSTI ART, since all subjects with TDR remained
virologically suppressed at 96 weeks.
As in other Spanish and European cohorts,
15,16,18,19,25,26
the
overall rate of TDR in this study was 13.1%, with a predominance
of NNRTI mutations and very low rates of INSTI- and PI-associated
mutations. In our study, only 1.7% of subjects had INSTI-
associated mutations as majority variants, although this rate
increased to 9.6% when low-frequency variants were also
considered.
INSTI-associated low-frequency resistance mutations have been
analysed in several studies,
2731
but there is no consensus regarding
their clinical relevance. Although they may affect INSTI susceptibility
in INSTI-experienced subjects,
31
studies have failed to show a dis-
cernible impact of low-frequency INSTI mutations on virological out-
comes of first-line INSTI-based ART.
28,30,32
Concordantly with these
data, in our study, none of the subjects with transmitted INSTI- or
NRTI-associated mutations, either as high- or low-frequency var-
iants, developed VF. In fact, virtually all subjects retained an HIV-1
fully susceptible to all INSTI-based regimens.
As far as we are aware, this is the first study demonstrating that
R263K transmission is possible in clinical settings and, in this case,
it was not associated with any significant effect on virological re-
sponse. Interestingly, we found one sample with transmitted
R263K alone as the majority variant, while in another sample it
was a low-frequency variant, together with the T97A mutation as
a majority variant. To date, the mutation R263K had only been
reported in clinical samples in patients developingVF to dolutegra-
vir.
3335
This mutation confers low-level resistance to dolutegravir
and bictegravir as a single mutation, does not seem to be
associated with compensatory substitutions that might increase
levels of resistance
36,37
and has a negative impact on viral replica-
tive fitness.
37,38
In fact, some studies have hypothesized that
R263K leads HIV into an evolutionary dead-end,
39
although this
does not seem to happen in vivo.
In our study, mutation E138K was detected as a majority vari-
ant in only one case, and in combination with other INSTI minority
variants, including one subject with Q148H who did not experience
VF during follow-up. By itself, the E138K mutation does not reduce
INSTI susceptibility, but in combination with Q148 mutations it
confers cross-resistance to all INSTIs.
36,40
Q148H/R/K mutations
can be selected in patients receiving raltegravir, elvitegravir and
cabotegravir, and also have minimal effects on dolutegravir sus-
ceptibility when they are detected as a single mutation, but confer
a high-level cross-resistance to all INSTIs when other secondary
resistance substitutions are present.
40
Nevertheless, our study
suggests that Q148H does not seem tohave any impact on the ef-
ficacy of first-line INSTI-based ART when it is present as a minority
variant.
Integrase polymorphisms, and in particular E157Q, may have a
negative impact on INSTI-based regimens and have been found to
be relatively prevalent in some subpopulations of HIV-infected
patients.
4143
Nevertheless, the studies where they have been
reported have included a limited number of subjects.
4143
In our
study, we did not find viruses harbouring E157Q, although we
found other integrase mutations, such as G163R, detected as both
majority and minority variants, and minority viruses harbouring
some polymorphic and non-polymorphic mutations (T97A, L74I/
M, H51Y, V151A/L and S230R). Alone, however, they had little, if
any, effect on INSTI susceptibility.
38
The main strengths of this study are that, unlike most epi-
demiological surveillance studies performed in Europe in which the
transmitted resistance is usually evaluated by Sanger sequencing,
the analysis of TDR was mainly performed by means of NGS.
In addition, the clinical outcomes of TDR could be evaluated thanks
to the prospective follow-up of the patients. Nevertheless, our
study is subject to a number of limitations. Genotyping tests at
baseline were performed by means of NGS and Sanger population
sequencing. Therefore, the rate of minority variants and their
effects on virological outcomes may have been underestimated.
However, this reflects the observational design of our study and
the reality of HIV care centres in which Sanger sequencing is still
widely used. In addition, there is no consensus list of integrase
mutations for surveillance. Although integrase resistance
Figure 3. Susceptibility for INSTI based on regimens according to GSS in naive patients starting first-line ART and with an available genotyping test
analysed by NGS or Sanger population sequencing (n= 173). The potential susceptibility of the main INSTI-based regimens currently available was
evaluated by means of the GSS corresponding to the number of ‘active’ drugs prescribed at baseline. According to the GSS, susceptibility of regimens
was classified in: 2.5–3.0; 2; and <1.5 points. HIVdb was used to calculate the GSS, considering a score of 0–9 as susceptible (1 point), 10–59 as inter-
mediate (0.5 points) and 60 points as resistant (0 points). RAL, raltegravir; EVG, elvitegravir; DTG, dolutegravir; 3TC, lamivudine; ABC, abacavir; FTC,
emtricitabine; TDF, tenofovir disoproxil fumarate; TAF, tenofovir alafenamide; BIK, bictegravir. This figure appears in colour in the online version of JAC
and in black and white in the print version of JAC.
Primary INSTI resistance in Spain JAC
3521
Downloaded from https://academic.oup.com/jac/article/75/12/3517/5905666 by guest on 12 April 2023
information was obtained taking into account majority variants
included in validated resistance algorithms (IAS and HIVdB), there
are differences between these algorithms that could make it diffi-
cult to evaluate the rate of mutations. In particular, the inclusion
of polymorphic integrase positions in our evaluation could have led
to an overestimation of the already low rates of transmitted INSTI
resistance. This reflects the need to develop a consensus list for
the purpose of epidemiological surveillance, which would make it
possible to perform larger and more precise epidemiological stud-
ies. Automated analysis pipelines for NGS could be useful for this
purpose.
44,45
However, to date, there is no evidence that lowering
the mutant detection threshold below 15%–20% (e.g. the Sanger
sequencing equivalent) provides any clinical or public health value.
Taken together, however, our data confirm that the prevalence
of transmitted INSTI-associated mutations is still very low in naive
patients and such mutations have a minimal impact, if any, on the
susceptibility of currently available INSTIs. The low frequency of
TDR mutations conferring resistance to dolutegravir, and the more
recently available bictegravir, is concordant with the idea that
genotyping tests are not mandatory when triple INSTI-based
regimens are considered for starting first-line ART.
46
Conversely,
pre-ART resistance testing probably continues to provide value in
subjects initiating dolutegravir !lamivudine dual therapy.
47
Although transmission of INSTI mutations is rare, transmission
of M184V mutants is frequent in newly diagnosed subjects with
previous prophylaxis pre-exposition exposure.
48
In conclusion, the rate of INSTI resistance mutations in naive
subjects remains very low in Spain and thus far has not been
associated with VF. Pre-ART genotyping tests are not needed to
guide INSTI-based initiation as first-line ART, although periodic sur-
veillance of INSTI TDR is warranted.
Acknowledgements
We are grateful to Ne´stor Sanchez and Nuria Pe´rez-A
´lvarez for support
with data management and the statistical analysis, Miryam Soler for
help with coordinating and recording all data, and Michael Kennedy-
Scanlon for proofreading assistance.
Members of the INSTINCT Study Group
Jose´ R. Santos, Isabel Bravo, Anna Chamorro, Cristina Miranda (Lluita
contra la SIDA Foundation, Hospital Universitari Germans Trias i Pujol,
Badalona); Rafael Mica´n Rivera, Juan Gonza´lez (University Hospital La
Paz, Madrid); Antonio Antela (Infectious Diseases Unit, Santiago de
Compostela Clinical University Hospital); Marcos Diez, Irene Portilla,
Melissa Carreres, Livia Giner, Vicente Boix, Sergio Reus, Esperanza Merino,
Diego Torru´s, Joaquı´n Portilla (General University Hospital, Alicante);
Jesu´s Sanz, A
´ngela Gutie´rrez Liarte, Ana Go´mez Berrocal (University
Hospital de La Princesa, Madrid); Pere Domingo, Marı´a del Mar Gutie´rrez,
Marı´a Gracia Mateo, Je`ssica Mu~
noz Rodrı´guez (Infectious Diseases Unit,
Hospital de la Santa Creu i Sant Pau, Barcelona); Marta Montero-Alonso
(Infectious Diseases Unit, La Fe University and Polytechnic Hospital,
Valencia); Adria` Curran, Ariadna Torrella, Bibiana Planas, Jordi Navarro
(Infectious Diseases Department, Hospital Universitari Vall d’Hebron,
Barcelona); Mar Masia´, Sergio Padilla, Catalina Robledano, Araceli Adsuar,
Fernando Montolio, Fe´lix Gutie´rrez (Infectious Diseases Unit, Elche
University General Hospital, Elche); Nieves Valcarce Pardeiro, Hortensia
A
´lvarez, Ana Mari~
no (Infectious Diseases Unit, Hospital Arquitecto
Marcide, Ferrol); Antonio Ocampo, Alfredo Rodrı´guez, Celia Miralles (HIV
Unit, Hospital A
´lvaro Cunqueiro, Vigo); Laura Pe´rez-Martı´nez, Jose´ Ramo´n
Blanco (Infectious Diseases Area, Hospital San Pedro-CIBIR, Logro~
no);
Coral Garcı´a Vallecillos, Juan Pasquau (University Hospital Virgen de las
Nieves, Granada); Marı´a Je´ sus Pe´rez-Elı´as, Fernando Dronda, Marı´a Jesu´s
Vivancos, Santiago Moreno (Infectious Diseases Unit, Ramo´n y Cajal
Hospital, Madrid); Arkaitz Imaz, Daniel Podzamczer [HIV and STI Unit,
Infectious Diseases Department, Bellvitge University Hospital, Bellvitge
Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat];
Maialen Ibarguren, Xabier Kortajarena, Marı´a Pilar Carmona-Oyaga,
Josean A. Iribarren (Infectious Diseases Unit, Donostia University
Hospital, San Sebastia´n); Leopoldo Mu~
noz Moreno, Jose´ Herna´ndez
Quero (University Hospital San Cecilio, Granada); Judit Villar-Garcı´a,
Hernando Knobel (Infectious Diseases Service, Hospital del Mar,
Barcelona); Pilar Barrufet, Lluı´s Force (Infectious Diseases Unit, Mataro´
Hospital, Mataro´); Maria Casadella`, Roger Paredes, Marc Noguera-Julian
(IrsiCaixa AIDS Reseach Institute, Hospital Universitai Germans Trias i
Pujol, Universitat Auto`noma de Barcelona, Badalona).
Funding
This study was supported by ViiV Healthcare and, in part, by grants from
the Lluita contra la SIDA Foundation (Barcelona, Spain), the Spanish AIDS
Network ‘Red Tema´tica Cooperativa de Investigacio´n en SIDA’ [RIS,
RD16/0025/0041, NEAT (European AIDS Treatment Network)], the
Ministerio de Economı´a y Competitividad of Spain (MINECO/FEDER,
MTM2015-64465-C2-1-R) and GRBIO (Grup de Recerca en Bioestadı´stica i
Bioinforma`tica; 2017 SGR 622). The funders had no role in the study de-
sign, data collection and analysis, the decision to publish or drafting of
the manuscript.
Transparency declarations
J.R.S., A.A. and J. Pasquau have received research funding, consultancy
fees and lecture sponsorships from and have served on advisory boards
for Gilead Sciences, Janssen-Cilag, Merck Sharp & Dohme and ViiV
Healthcare. J. Portilla, J.S., R.P. and A.O. have received research funding
and consultancy fees from and have served on advisory boards for
Gilead Sciences, Merck Sharp & Dohme and ViiV Healthcare. P.D. has
received honoraria for speeches, advisory boards, slide sets and research
grants from Gilead Sciences, Merck Sharp & Dohme, ViiV Healthcare and
Janssen-Cilag. M.M. has received research funding, consultancy fees
and lecture sponsorships from and has served on advisory boards for
Gilead Sciences, Janssen-Cilag, Merck Sharp & Dohme, Pfizer and ViiV
Healthcare. M.M.-A. has received consultancy fees and lecture sponsor-
ships from and has served on advisory boards for Abbvie, Janssen-Cilag,
ViiV Healthcare and Merck Sharp & Dohme. J.N. has received honoraria
and/or speakers’ fees from Abbvie, Gilead, Janssen-Cilag, Merck Sharp &
Dohme and ViiV Healthcare outside of the submitted work. M.J.V. has
received research funding, consultancy fees and lecture sponsorships
from and has served on advisory boards for Gilead and ViiV Healthcare.
A.I. has received research funding, financial compensation for lectures
and educational activities, or has served on advisory boards for Gilead
Sciences, Janssen-Cilag, Merck Sharp & Dohme and ViiV Healthcare.
P.B. has received consultancy fees from and has served on advisory
boards for Gilead Sciences, Merck Sharp & Dohme, Janssen-Cilag and
ViiV Healthcare. M.C., N.V.-P., R.M.-R., L.M.-M., J.V.-G., P.C.-O., L.P.-M. and
M.N.-J. declare no competing interests.
Author contributions
J.R.S. and R.P. designed the INSTINCT study. M.C., J.R.S. and R.P. wrote the
manuscript, which was reviewed and approved by all the authors. M.C.
performed the laboratory and sequencing data analyses. The remaining
Casadella` et al.
3522
Downloaded from https://academic.oup.com/jac/article/75/12/3517/5905666 by guest on 12 April 2023
authors contributed clinical and Sanger sequencing data (when required)
and followed the study subjects.
Supplementary data
Table S1 is available as Supplementary data at JAC Online.
References
1European AIDS Clinical Society (EACS). Guidelines Version 9.1. October
2018. https://www.eacsociety.org/files/2018_guidelines-9.1-english.pdf.
2DHHS Panel on Antiretroviral Guidelines for Adults and Adolescents. Adults
and Adolescents Living with HIV Guidelines for the Use of Antiretroviral
Agents in Adults and Adolescents Living with HIV Developed by the
DHHS Panel on Antiretroviral Guidelines for Adults.Department of Health
and Human Services 2018. https://aidsinfo.nih.gov/contentfiles/Adultand
AdolescentGL003536.pdf.
3WHO. Consolidated Guidelines on the Use of Antiretroviral Drugs for
Treating and Preventing HIV Infection: What’s New? 2015: 1–16. https://
www.who.int/hiv/pub/arv/en/.
4DHHS Panel on Antiretroviral Guidelines for Adults and Adolescents.
Guidelines for the Use of Antiretroviral Agents in HIV-1-Infected Adults and
Adolescents Developed by the DHHS Panel on Antiretroviral Guidelines for
Adults. Department of Health and Human Services 2016. https://aidsinfo.nih.
gov/contentfiles/AdultandAdolescentGL003464.pdf.
5European AIDS Clinical Society (EACS). Guidelines Version 9.0. October
2017. https://www.eacsociety.org/files/guidelines_9.0-english.pdf.
6Eron JJ, Clotet B, Durant J et al. Safety and efficacy of dolutegravir in
treatment-experienced subjects with raltegravir-resistant HIV type 1 infec-
tion: 24-week results of the VIKING study. JInfectDis2013; 207:7408.
7Nichols G, Lazzarin A, Maggiolo F et al. Phase 3 assessment of dolutegravir
(DTG) 50 mg twice daily (BID) in HIV-1-infected subjects with raltegravir (RAL)
and/or elvitegravir (EVG) resistance in VIKING-3: week 24 results of all 183
subjects enrolled. Seventh IAS Conference on HIV Pathogenesis, Treatment
and Prevention, Kuala Lumpur, Malaysia, 2013. Poster TULBPE19.
8Tsiang M, Jones GS, Goldsmith J et al. Antiviral activity of bictegravir
(GS-9883), a novel potent HIV-1 integrase strand transfer inhibitor with
an improved resistance profile. Antimicrob Agents Chemother 2016; 60:
7086–97.
9Santoro M, Fornabaio C, Malena M et al. Susceptibility to Bictegravir in high-
ly ARV experienced patients after INSTI failure. Sixteenth Conference on
Retroviruses and Opportunistic Infections (CROI),Seattle, WA, USA, 2019.
Poster 0550.
10 Kantor R, Smeaton L, Vardhanabhuti S et al. Pretreatment HIV drug re-
sistance and HIV-1 subtype C are independently associated with virologic
failure: results from the multinational PEARLS (ACTG A5175) Clinical Trial. Clin
Infect Dis 2015; 60: 1541–9.
11 Wittkop L, Gu¨nthard HF,de WolfF et al. Effect of transmitted drug resist-
ance on virological and immunological response to initial combination anti-
retroviral therapy for HIV (EuroCoord-CHAIN joint project): a European
multicohort study. Lancet Infect Dis 2011; 11: 363–71.
12 Hamers RL, Schuurman R, Sigaloff KCE et al. Effect of pretreatment HIV-1
drug resistance on immunological, virological, and drug-resistance outcomes
of first-line antiretroviral treatment in sub-Saharan Africa: a multicentre co-
hort study. Lancet Infect Dis 2012; 12: 307–17.
13 Boyd SD, Maldarelli F, Sereti I et al. Transmitted raltegravir resistance in
an HIV-1 CRF_AG-infected patient. Antivir Ther 2011; 16: 257–61.
14 Young B, Fransen S, Greenberg KS et al. Transmission of integrase strand-
transfer inhibitor multidrug-resistant HIV-1: case report and response to
raltegravir-containing antiretroviral therapy. Antivir Ther 2011; 16:2536.
15 Santos JR, Blanco JL, Masia´Met al. Virological failure to raltegravir
in Spain: incidence, prevalence and clinical consequences. J Antimicrob
Chemother 2015; 70: 3087–95.
16 Hauser A, Hofmann A, Meixenberger K et al. Increasing proportions of
HIV-1 non-B subtypes and of NNRTI resistance between 2013 and 2016 in
Germany: results from the national molecular surveillance of new HIV-diag-
noses. PLoS One 2018; 13: 1–18.
17 Casadella` M, van Ham PM, Noguera-Julian M et al. Primary resistance to
integrase strand-transfer inhibitors in Europe. J Antimicrob Chemother 2015;
70: 2885–8.
18 Alvarez M, Casas P, de Salazar A et al. Surveillance of transmitted drug
resistance to integrase inhibitors in Spain: implications for clinical practice.
J Antimicrob Chemother 2019; 74: 1693–700.
19 Raffaelli CS, Rossetti B, Paglicci L et al. Impact of transmitted HIV-1 drug
resistance on the efficacy of first-line antiretroviral therapy with two
nucleos(t)ide reverse transcriptase inhibitors plus an integrase inhibitor or a
protease inhibitor. J Antimicrob Chemother 2018; 73: 2480–4.
20 Wensing AM, Calvez V, Ceccherini-Silberstein F et al. 2019 update of the
drug resistance mutations in HIV-1. Top Antivir Med 2019; 27: 111–21.
21 Bennett D, Camacho R, Otelea D et al. List of mutations for surveillance of
transmitted drug resistant HIV: 2009 update. Sixteenth Conference on
Retroviruses and Opportunistic Infections (CROI 2009), Montreal, Canada´,
2009. Poster 689.
22 Inzaule SC, Hamers RL, Noguera-Julian M et al. Primary resistance to
integrase strand transfer inhibitors in patients infected with diverse HIV-1
subtypes in sub-Saharan Africa. J Antimicrob Chemother 2018; 73: 1167–72.
23 Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for
Illumina sequence data. Bioinformatics 2014; 30: 2114–20.
24 Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2.
Nat Methods 2012; 9:3579.
25 Hofstra LM, Sauvageot N, Albert J et al. Transmission of HIV drug resist-
anceandthepredictedeffectoncurrentfirst-lineregimensinEurope.Clin
Infect Dis 2016; 62: 655–63.
26 Tostevin A, White E, Dunn D et al. Recent trends and patterns in HIV-1
transmitted drug resistance in the United Kingdom. HIV Med 2017; 18:
204–13.
27 Armenia D, Vandenbroucke I, Fabeni L et al. Study of genotypic and
phenotypic HIV-1 dynamics of integrase mutations during raltegravir treat-
ment: a refined analysis by ultra-deep 454 pyrosequencing. JInfectDis2012;
205: 557–67.
28 Charpentier C, Laureillard D, Piketty C et al. High frequency of integrase
Q148R minority variants in HIV-infected patients naive of integrase inhibitors.
AIDS 2010; 24: 867–73.
29 Li JZ, Chapman B, Charlebois P et al. Comparison of Illumina and 454
deep sequencing in participants failing raltegravir-based antiretroviral
therapy. PLoS One 2014; 9: e90485.
30 Liu J, Miller MD, Danovich RM et al. Analysis of low-frequency mutations
associated with drug resistance to raltegravir before antiretroviral treatment.
Antimicrob Agents Chemother 2011; 55: 1114–9.
31 Gibson RM, Weber J, Winner D et al. Contribution of human immunodefi-
ciency virus type 1 minority variants to reduced drug susceptibility in patients
on an integrase strand transfer inhibitor-based therapy. PLoS One 2014; 9:
e104512.
32 Acosta RK, Willkom M, Martin R et al. Resistance analysis of bictegravir–
emtricitabine–tenofovir alafenamide in HIV-1 treatment-naive patients
through 48 weeks. Antimicrob Agents Chemother 2019; 63:128.
33 Wijting I, Lungu C, Rijnders B et al. HIV-1 resistance dynamics in patients
failing dolutegravir maintenance monotherapy. JInfectDis2018; 218:
688–97.
34 Cardoso M, Baptista T, Diogo I et al. Two cases of dolutegravir failure with
R263K mutation. AIDS 2018; 32: 2639–40.
Primary INSTI resistance in Spain JAC
3523
Downloaded from https://academic.oup.com/jac/article/75/12/3517/5905666 by guest on 12 April 2023
35 Ahmed N, Flavell S, Ferns B et al. Development of the R263K mutation to
dolutegravir in an HIV-1 subtype D virus harboring 3 class-drug resistance.
Open Forum Infect Dis 2019; 6:35.
36 Mesple`de T, Osman N, Wares M et al. Addition of E138K to R263K in HIV
integrase increases resistance to dolutegravir, but fails to restore activity of
the HIV integrase enzyme and viral replication capacity. JAntimicrob
Chemother 2014; 69: 2733–40.
37 Mesple`deT,QuashiePK,OsmanNet al. Viral fitness cost prevents HIV-1
from evading dolutegravir drug pressure. Retrovirology 2013; 10:17.
38 Anstett K, Brenner B, Mesple`de T, Wainberg MA. HIV drug resistance
against strand transfer integrase inhibitors. Retrovirology 2017; 14:
1–16.
39 Anstett K, Mesple`de T, Oliveira M et al. Dolutegravir resistance mutation
R263K cannot coexist in combination with many classical integrase inhibitor
resistance substitutions. JVirol2015; 89: 4681–4.
40 Oliveira M, Ibanescu RI, Anstett K et al. Selective resistance profiles
emerging in patient-derived clinical isolates with cabotegravir, bictegravir,
dolutegravir, and elvitegravir. Retrovirology 2018; 15:114.
41 Ambrosioni J, Rico JA
´FC, Nicola´s D et al. E157Q integrase strand-transfer
inhibitor substitution in patients with acute/recent HIV infection. AIDS 2019;
33: 1613–17.
42 Ambrosioni J, Nicola´ s D, Manzardo C et al. Integrase strand-transfer
inhibitor polymorphic and accessory resistance substitutions in patients
with acute/recent HIV infection. J Antimicrob Chemother 2017; 72:
205–9.
43 Charpentier C, Malet I, Andre-Garnier E et al. Phenotypic analysis of HIV-1
E157Q integrase polymorphism and impact on virological outcome in
patients initiating an integrase inhibitor-based regimen. JAntimicrob
Chemother 2018; 73: 1039–44.
44 Noguera-Julian M, Edgil D, Harrigan PR et al. Next-generation human
immunodeficiency virus sequencing for patient management and drug
resistance surveillance. JInfectDis2017; 216: S829–33.
45 Casadella` M, Paredes R. Deep sequencing for HIV-1 clinical management.
Virus Res 2017; 239: 69–81.
46 HyleEP,ScottJA,SaxPet al. Clinical impact and cost-effectiveness of
genotype testing at human immunodeficiency virus diagnosis in the United
States. Clin Infect Dis 2019; 70: 1353–63.
47 Cahn P, Madero JS, Arribas JR et al. Dolutegravir plus lamivudine versus
dolutegravir plus tenofovir disoproxil fumarate and emtricitabine in
antiretroviral-naive adults with HIV-1 infection (GEMINI-1 and GEMINI-2):
week 48 results from two multicentre, double-blind, randomised, non-infer-
ior. Lancet 2019; 393: 143–55.
48 Misra K, Huang J, Daskalakis DC et al. Impact of PrEP on drug resistance
and acute HIV infection, New York City, 2015–2017. Sixteenth Conference on
Retroviruses and Opportunistic Infections (CROI), Seattle, WA, USA, 2019.
Poster 107.
Casadella` et al.
3524
Downloaded from https://academic.oup.com/jac/article/75/12/3517/5905666 by guest on 12 April 2023
... Highly active antiretroviral therapy (HAART) has proven to be efficacious in suppressing HIV replication and mitigating HIV-related morbidity and mortality [2]. Nevertheless, the wide utilization of antiretroviral drugs in clinical practice has given rise to HIV drug resistance, which significantly impacts the effectiveness of ART in both treatment-naive and experienced patients [3][4][5]. ...
... However, the growing application of INSTIs in clinical practice has resulted in the inevitable emergence of resistance. Previously published studies have reported significant INSTIs resistance mutations among HIV-1 patients in various regions, including the United States [8], Canada [9], Europe [10], and Spain [5]. Likewise, major INSTIs resistance mutations have also been identified in several locations within China, such as Shenyang [11], Henan [4], Yunnan [12], and Jiangsu [13]. ...
Article
Full-text available
In this study, we examined the occurrence of acquired and transmitted drug resistance to integrase strand transfer inhibitor (INSTI) in HIV-1 strains in Chongqing (China) for guiding for the routine testing of INSTI-associated HIV-1 genotype resistance. Plasma samples were obtained from HIV-1 patients at Chongqing Public Health Medical Center from July 2019 to August 2022. Besides, amplification, sequence, and analysis of the portion of the HIV-1 pol gene that encodes the integrase protein were implemented to identify INSTI resistance. Integrase sequence data was harvested for a comprehensive cohort of 1032 patients infected with HIV-1. This cohort consisted of 564 ART-naive patients, 465 ART-treated patients, and 3 patients with an unknown treatment history. Within the study group, we identified INSTI resistance in 21 patients (2.03%, 21/1032), including 17 ART-treated patients (3.66%, 17/465). Among the ART-treated patients, 12 were INSTI-treated (11.76%, 12/102), 5 were INSTI-naive (1.38%, 5/363), and 4 were ART-ineffective patients (0.71%, 4/564). The prevalent major resistance mutation was Q148R (0.48%, 5/1032), while the most prevalent accessory resistance mutation was E157Q (1.65%, 17/1032). In light of the above, it is recommended that the incidence of accessory genotype analysis should be considered before starting any future INSTI-based therapy, especially in patients with drug resistance to NRTIs and NNRTIs and the reduction of INSTI sensitivity should be carefully monitored and investigated. Regular monitoring for resistance should be implemented after the use of INSTIs, and, importantly, ongoing monitoring of the decreasing susceptibility to INSTIs is crucial following the initiation of treatment with INSTIs.
... In the last years, a global increase in the use of INSTIcontaining regimens is occurring. In spite of this, very low prevalences of transmitted drug resistance (TDR) to INSTIs have been reported (Inzaule et al., 2018;Casadellá et al., 2020), supporting the use of this drug class in first-line ARV treatments. However, the great increase in the use of second-generation INSTIs and cross-resistance among INSTIs advocate for a continued surveillance of TDR, as well as monitoring their clinical impact. ...
... Moreover, the most frequent INSTI resistance mutation found among these patients was the polymorphic G163K/R mutation (Tzou et al., 2020), which is only associated with a low-level resistance to RAL and EVG. The low frequency of transmitted INSTIs resistance mutations has been reported in previous surveillance studies and meta-analyses (Doyle et al., 2015;Hauser et al., 2018;Inzaule et al., 2018;Raffaelli et al., 2018;Alvarez et al., 2019;Liu et al., 2019;Casadellá et al., 2020;Mbisa et al., 2020;Tzou et al., 2020;Semengue et al., 2021). This scenario supports the use of INSTIs in first-line therapeutic regimens. ...
Article
Full-text available
Integrase strand transfer inhibitor (INSTI)-containing regimens in HIV-1-infected patients have experienced a global increase. Recently, WHO has emphasized the need to fast-track the transition to dolutegravir (DTG)-based antiretroviral (ARV) treatments. However, continued surveillance of INSTI resistance is recommended. In this study, clinical, epidemiological, and virological features associated with INSTI resistance diagnosed in Spain were analyzed. Samples collected between 2008 and 2021 from HIV-1-infected patients were analyzed in integrase, protease, and reverse transcriptase using Sanger population sequencing. ARV drug resistance was evaluated with the Stanford University HIVdb program. Among 2,696 patients, 174 (6.5%) had INSTI resistance, all of them to first-generation INSTIs, and 71 (2.6%) had also resistance to second-generation INSTIs. Of these, only 5 individuals were exposed to DTG as the only INSTI, in whom resistance development was associated with poor treatment adherence and/or resistance to other ARV classes. Of newly HIV-1-diagnosed individuals, 0.92% harbored INSTI-resistant viruses, with low prevalences maintained along time, and only one had low-level resistance to DTG. Persons who inject drugs, age over 39 years, resistance to other ARV classes, and longer time from diagnosis were associated with INSTI resistance ( p < 0.001). Non-subtype B INSTI-resistant viruses lacked the Q148H + G140S resistance pathway and showed lower INSTI resistance levels than subtype B viruses. In conclusion, INSTI resistance is uncommon and associated with long-term infections, older age and additional resistance to other ARV drug classes, and is rare in newly diagnosed HIV-1 infections. Our results also support the preferential use of DTG-containing regimens in first-line treatments, although surveillance of INSTI resistance is encouraged.
... The latest study, led by Roger Paredes and others, found that the prevalence of INSTI resistance mutations among treatment-naive individuals remains remarkably low in Spain. However, in 9.6% of the subjects, minority variants carrying integrase transmitted drug resistance (TDR) were detected, which have not yet been linked with virologic failure (VF)[17]. And their research ndings suggested that setting a very low threshold (< 1%) when using NGS may affect the speci city of the test and lead to a high misjudgment rate, incorrectly categorizing patients with viral control as being at risk of VF[18]. ...
Preprint
Full-text available
Background The rapid initiation of antiretroviral therapy has become an international trend, necessitating lifelong medication for all HIV patients. Sanger sequencing, as the gold standard for clinically detecting HIV drug resistance, often fails to detect mutations comprising less than 20% of the total viral population. With the advancement of detection technologies, HIV-1 DRMinVs have garnered increasing attention. However, there are few studies exploring the hotspots and trends in this field. Fortunately, bibliometrics, a novel approach to literature analysis, can fill this gap effectively. Methods Publications related to HIV-1 DRMinVs from 1994 to 2022 were searched on the WoSCC database. Visual knowledge maps and bibliometric analyses were generated using VOSviewers, CiteSpace, and the R package "bibliometrix." Results In total, 853 publications concerning HIV-1 DRMinVs were identified from 1994 to 2022, demonstrating a steady increase in publication output over the years. The United States, France, and the United Kingdom significantly lead in publication output. The main research institutions are Harvard University, University of Pittsburgh, Stanford University and National Cancer Institute. The Journal of Antimicrobial Chemotherapy holds the highest prominence among journals in this domain, while the Journal of Virology emerges as the most frequently co-cited journal. A total of 5687 authors have contributed to these publications. Among them, Vincent Calvez, Francesca Ceccherini-Silberstein, and John M. Coffin emerge as the most prolific authors, having published the highest number of articles. Additionally, Metzner, KJ emerges as the most frequently co-cited author. The main trends include the origins, molecular epidemiology, detection methods of DRMinVs, their impact on virological outcomes in drug-naïve patients, and novel research focuses primarily revolve around keywords such as "NGS," "ART," "VF," and "GRT." Conclusions The use of medication inevitably leads to drug resistance. For HIV-1 DRMinVs, the emergence of NGS has addressed the issue of missed detections by Sanger sequencing. However, its high cost and stringent laboratory requirements have limited its widespread application. Therefore, future research should focus on improving and refining NGS to make it simpler and more affordable, and explore when it can serve as a supplement to Sanger sequencing.
... On the other hand, the prevalence of drug resistance to PIs was lower than observed in the previous study (2.5% vs. 3.9%) [7]. As expected, drug resistance to INSTIs in naïve patients was very low (0.3%), and these findings corroborate data from the European literature, where the prevalence of TDR to INSTIs ranges from 0.2% to 1.7% [22][23][24][25], with E138K and R263K being more frequently identified, which can be selected by the first-line ART regimen currently adopted in Portugal. ...
Article
Full-text available
Background: The global scale-up of antiretroviral treatment (ART) offers significant health benefits by suppressing HIV-1 replication and increasing CD4 cell counts. However, incomplete viral suppression poses a potential threat for the emergence of drug resistance mutations (DRMs), limiting ART options, and increasing HIV transmission. Objective: We investigated the patterns of transmitted drug resistance (TDR) and acquired drug resistance (ADR) among HIV-1 patients in Portugal. Methods: Data were obtained from 1050 HIV-1 patient samples submitted for HIV drug resistance (HIVDR) testing from January 2022 to June 2023. Evaluation of DRM affecting viral susceptibility to nucleoside/tide reverse transcriptase inhibitors (NRTIs), non-nucleoside reverse transcriptase inhibitors (NNRTIs), protease inhibitors (PIs), and integrase strand transfer inhibitors (INSTIs) was performed using an NGS technology, the Vela Diagnostics Sentosa SQ HIV-1 Genotyping Assay. Results: About 71% of patients were ART naïve and 29% were experienced. Overall, 20% presented with any DRM. The prevalence of TDR and ADR was 12.6% and 41.1%, respectively. M184V, T215S, and M41L mutations for NRTI, K103N for NNRTI, and M46I/L for PIs were frequent in naïve and treated patients. E138K and R263K mutations against INSTIs were more frequent in naïve than treated patients. TDR and ADR to INSTIs were 0.3% and 7%, respectively. Patients aged 50 or over (OR: 1.81, p = 0.015), originating from Portuguese-speaking African countries (PALOPs) (OR: 1.55, p = 0.050), HIV-1 subtype G (OR: 1.78, p = 0.010), and with CD4 < 200 cells/mm³ (OR: 1.70, p = 0.043) were more likely to present with DRMs, while the males (OR: 0.63, p = 0.003) with a viral load between 4.1 to 5.0 Log10 (OR: 0.55, p = 0.003) or greater than 5.0 Log10 (OR: 0.52, p < 0.001), had lower chances of presenting with DRMs. Conclusions: We present the first evidence on TDR and ADR to INSTI regimens in followed up patients presenting for healthcare in Portugal. We observed low levels of TDR to INSTIs among ART-naïve and moderate levels in ART-exposed patients. Regimens containing PIs could be an alternative second line in patients with intermediate or high-level drug resistance, especially against second-generation INSTIs (dolutegravir, bictegravir, and cabotegravir).
... INSTI is the latest drug to be developed with high efficacy, safety, and tolerability. The first generation drugs of INSTI, namely, raltegravir (RAL) and elvitegravir (EVG), are recommended for the initiation of ARV therapy [26]. RAL is classified under INSTI with a high need for daily pill consumption and low genetic barrier, which puts patients at risk for developing resistance mutations. ...
Article
Full-text available
Although primary integrase strand transfer inhibitor resistance mutations are currently uncommon, the increasing use of integrase strand transfer inhibitor as a key component of the first, second and third-line antiretroviral regimens suggests that the prevalence of integrase drug resistance mutations will likely increase. The rise of several polymorphic mutations and natural polymorphisms also affects the level of susceptibility of human immunodeficiency virus (HIV) type-1 to integrase strand transfer inhibitor. The considerable variability among the various subtypes of human immunodeficiency virus type-1 may contribute to differences in integrase mutations associated with integrase strand transfer inhibitors. Notably, non-B subtypes of HIV type-1 (HIV-1) are the predominant cause of human immunodeficiency virus infection worldwide. The presence of diverse integrase drug resistance mutations can have significant implications on the administration of integrase strand transfer inhibitor-based antiretroviral therapy to patients with human immunodeficiency virus infection.
... The United States Food and Drug Administration (United States FDA) has approved the following five drugs for clinical use: dolutegravir (DTG), raltegravir (RAL), elvitegravir (EVG), bictegravir (BIC), and cabotegravir (CAB). Despite integrase inhibitors plays an effective role in antiretroviral action with a novel mechanism of action, resistance is inevitable (Jiang et al., 2016), and previously published studies have shown that there were important INSTIs resistance mutations in newly diagnosed HIV-1 patients in Spain, Canada, and the United States (Stekler et al., 2015;Ji et al., 2018;Casadellà et al., 2020;López et al., 2021). Major INSTIs resistance mutations have also been discovered in Yunnan (Deng et al., 2019) and Jiangsu (Yin et al., 2021) in China, and INSTIs resistance mutations were found in our neighboring provinces such as Henan (Yang et al., 2021), Beijing . ...
Article
Full-text available
Antiretroviral therapy (ART) regimens containing integrase strand transfer inhibitors (INSTIs) are the recommended treatment for human immunodeficiency virus type 1 (HIV-1)-infected patients in the most recent guidelines in China. In this study, we investigated INSTI resistance mutations in newly diagnosed therapy-naive HIV-positive patients in Baoding City, Hebei Province (China) to provide guidance for implementing routine INSTI-associated HIV-1 genotypic resistance testing. Plasma samples were collected from HIV-1-infected patients without treatment at Baoding People’s Hospital from January 2020 to December 2021. The part of HIV-1 pol gene encoding integrase was amplified, sequenced, and analyzed for INSTI resistance. Clinical data including demographic data, CD4⁺ T cell counts, HIV-RNA loads, and resistance mutations were collected. Treatment-naïve HIV-1 patients (n = 131) were enrolled. We identified ten genotypes, and the predominant genotype was CRF01_AE in 67 patients (51.15%), CRF07_ BC in 39 patients (29.77%), subtype B in 11 patients (8.40%), and other subtypes (CRF68_01B, 3.82%; CRF55_01B, 1.53%, CRF80_0107, 1.53%; URFs 1.53%; and CRF103_01B, CRF59_01B, and CRF65_cpx, 1.4% each). Four major (E138A, R263k, G140S, and S147G) and three accessory (H51Y, Q146QL, and S153F) INSTI-resistance mutations were observed (genotype CRF01_AE, three patients; genotype B, one patient; and genotype CRF07_BC, one patient), resulting in different degrees of resistance to the following five INSTIs: raltegravir, elvitegravir, dolutegravir, bictegravir, and cabotegravir. The overall resistance rate was 3.82% (5/131). All INSTI-resistant strains were cross-resistant. The primary INSTI drug resistance rate among newly diagnosed HIV-infected patients in Baoding was low, but monitoring and research on HIV INSTI resistance should be strengthened in Baoding because INSTI-based regimen prescriptions are anticipated to increase in the near future.
... However, following the wide scale-up of DTG, an increase in DTG resistance has been reported, especially in persons receiving DTG monotherapy [15,19,[23][24][25][26][27][28]. Hitherto, the prevalence of transmitted resistance to DTG resistance has been low [20][21][22][23]. Similarly, in Ethiopia, after implementing the test-andtreat strategy, an increased number of patients will be on a DTG-based regimen. ...
Article
Full-text available
Dolutegravir-based antiretroviral therapy (ART) has been scaled up in many developing countries, including Ethiopia. However, subtype-dependent polymorphic differences might influence the occurrence of HIV-drug-resistance mutations (HIVDRMs). We analyzed the prevalence of pre-treatment integrase strand transfer inhibitor (INSTI) HIVDRMs and naturally occurring poly-morphisms (NOPs) of the integrase gene, using plasma samples collected as part of the national HIVDR survey in Ethiopia in 2017. We included a total of 460 HIV-1 integrase gene sequences from INSTI-naïve (n = 373 ART-naïve and n = 87 ART-experienced) patients. No dolutegravir-associated HIVDRMs were detected, regardless of previous exposure to ART. However, we found E92G in one ART-naïve patient specimen and accessory mutations in 20/460 (4.3%) of the specimens. Moreover, among the 288 integrase amino acid positions of the subtype C, 187/288 (64.9%) were conserved (<1.0% variability). Analysis of the genetic barrier showed that the Q148H/K/R dolutegravir resistance pathway was less selected in subtype C. Docking analysis of the dolutegravir showed that protease-and reverse-transcriptase-associated HIVDRMs did not affect the native structure of the HIV-1 integrase. Our results support the implementation of a wide scale-up of dolutegravir-based regimes. However, the detection of polymorphisms contributing to INSTI warrants the continuous surveillance of INSTI resistance.
... The prevalence of TDR to the InSTI family in Spain is low, 19 although some studies have reported higher prevalence rates of InSTI polymorphic substitutions possibly capable of reducing the activity of first-generation InSTI (raltegravir, elvitegravir/cobicistat). 20,21 In our study, we identified five TN individuals carrying 97A and three TN individuals carrying 157Q detected in the baseline genotype. ...
Article
Background: The use of bictegravir/emtricitabine/tenofovir alafenamide (BIC/FTC/TAF) is mainly based on robust, pivotal clinical trials. Objectives: To provide data on clinical use of BIC/FTC/TAF in real life. Patients and methods: This was an observational, retrospective and single-centre study. We included all adult, treatment-naive (TN) and treatment-experienced (TE) people living with HIV (PLWH) starting BIC/FTC/TAF from 8 June 2018. We evaluated effectiveness [on treatment (OT), modified intention-to-treat (mITT) and intention-to-treat (ITT)], tolerability and safety in those patients who reached 6 months of follow-up (M6). Results: We included 1584 PLWH [213 TN (13%) and 1371 TE (87%)]. The median (IQR) follow-up was 16 (7-21) months, with 81% and 53% of PLWH reaching M6 and M12, respectively. By OT, mITT and ITT, HIV-RNA <50 copies/mL was 77%, 70% and 62% at M6 and 92%, 77% and 63% at M12 for TN PLWH and 94%, 89% and 83% at M6 and 93%, 85% and 78% at M12 for TE PLWH, respectively. In PLWH carrying an M184V/I substitution, OT RNA <50 copies/mL was 89.5% at M6. The median CD4 cell count increased from 329 to 511/μL in TN PLWH and from 630 to 683/μL in TE PLWH at M6. Of the total, 1148 (88%) PLWH continued on BIC/FTC/TAF at M6. The most frequent known reason for discontinuation was toxicity [42 (69%) cases]; only 7 cases were considered virological failures (0.6% of the total OT cohort at M6), with no emerging resistance substitutions. Conclusions: In real life, BIC/FTC/TAF showed high rates of virological suppression and also in PLWH carrying lamivudine/emtricitabine resistance substitutions. The tolerability and safety of BIC/FTC/TAF were good, with high persistence observed for patients on this regimen at M6.
Article
Objective: To study the prevalence of transmitted drug resistance (TDR) to INSTIs and NRTIs, and of clinically relevant resistance (CRR), in newly-diagnosed people with HIV (PWH) naïve to antiretroviral therapy (ART) in Europe. Methods: MeditRes HIV is a consortium that includes ART naïve PWH newly diagnosed in France, Greece, Italy, Portugal, and Spain during the years 2018-2021. Reverse transcriptase (RT) and Integrase (INSTI) sequences were provided by participating centers. To evaluate the prevalence of surveillance drug resistance mutations (SDRM) we used the CPR tools from Stanford HIV-website. To evaluate clinically relevant resistance (CRR), defined as any resistance level >= 3, we used the Stanford v.9.1HIVDB Algorithm. Results: We included 2705 PWH, 72% men, median age of 37 (IQR, 30-48); 43.7% infected by non-B subtypes. The prevalence of INSTI-SDRMs was 0.30% (T66I, T66A, E92Q, E138T, E138K, Y143R, S147G and R263K, all n = 1), and of NRTI-SDRMs was 5.77% (M184V n = 23, 0.85%; M184I n = 5, 0.18%; K65R/N n = 3, 0.11%; K70E n = 2, 0.07%; L74V/I n = 5, 0.18%; any TAMs n = 118, 4.36%). INSTI-CRR was 2.33% (0.15% dolutegravir/bictegravir; 2.29% raltegravir/elvitegravir), and 1.74% to first-line NRTIs (0.89% tenofovir/tenofovir alafenamide fumarate; 1.74% abacavir; 1.07% lamivudine/emtricitabine). Conclusions: We present the most recent data on TDR to integrase based first-line regimens in Europe. Given the low prevalence of CRR to second generation integrase inhibitors and to first-line NRTIs, in the years 2018-2021 it is unlikely that newly diagnosed PWH in MeditRes countries would present with baseline resistance to a first-line regimen based on second generation integrase inhibitors.
Thesis
Full-text available
Article
Full-text available
The 2019 edition of the IAS-USA drug resistance mutations list updates the Figure last published in January 2017. The mutations listed are those that have been identified by specific criteria for evidence and drugs described. The Figure is designed to assist practitioners in identifying key mutations associated with resistance to antiretroviral drugs, and therefore, in making clinical decisions regarding antiretroviral therapy.
Article
Full-text available
Background: US guidelines recommend standard genotype at HIV diagnosis ("baseline genotype") to detect transmitted drug resistance (TDR) to NNRTIs, NRTIs, and PIs. With INSTI-based regimens now recommended as first-line ART, the clinical and economic value of baseline genotypes is uncertain. Methods: We used the CEPAC model to examine the clinical impact and cost-effectiveness of Baseline Genotype compared to No Baseline Genotype for people starting ART with dolutegravir (DTG) and an NRTI pair. For people with no TDR (83.8%), baseline genotype does not alter regimen selection. Among people with transmitted NRTI resistance (NRTI-R, 5.8%), baseline genotype guides NRTI pair selection and informs subsequent ART after adverse events (DTG AE, 14%). Among people with transmitted NNRTI resistance (NNRTI-R, 7.2%), baseline genotype influences care only for people with DTG AE who move to an NNRTI-based regimen. 48-week virologic suppression varied (40%-92%), depending on TDR. Costs included $320/genotype, $3,000/month for DTG-based and darunavir/ritonavir-based regimens, and $2,500/month for rilpivirine-based regimens. Results: Compared to No Baseline Genotype, Baseline Genotype would result in <1 additional undiscounted quality-adjusted life day (QALD), cost $500 more per person, and would not be cost-effective (ICER, $420,000/quality-adjusted life year). In univariate sensitivity analysis, the clinical benefits of Baseline Genotype never exceeded 5 QALDs for all newly diagnosed people with HIV. Baseline Genotype was cost-effective at current TDR prevalence only under unlikely conditions, such as DTG-based regimens achieving ≤50% suppression of transmitted NRTI-R. Conclusions: With INSTI-based first-line regimens in the US, Baseline Genotype offers minimal clinical benefit and is not cost-effective.
Article
Full-text available
In Studies 1489 and 1490, bictegravir/emtricitabine/tenofovir alafenamide (B/F/TAF), dolutegravir/abacavir/lamivudine (DTG/ABC/3TC), or DTG+F/TAF treatment achieved high rates of virologic suppression in HIV-1 treatment-naïve participants through Week 48. Pre-existing primary drug resistance was present at 1.3% INSTI resistance (-R), 2.7% NRTI-R, 14.1% NNRTI-R, and 3.5% PI-R in the 1274 participants from these studies. These mutations did not affect treatment outcomes. Resistance analyses in 13 virologic failures found no emergent resistance to study drugs.
Article
Full-text available
Dolutegravir (DTG), a second-generation integrase strand-transfer inhibitor (INSTI), is equivalent or superior to current non-nucleotide reverse transcriptase inhibitors (NNRTIs), protease inhibitors (PIs), and first-generation INSTI-based antiretroviral regimens (ARVs). It has the potential to make big improvements in HIV control globally and within patients. This is perhaps the most “precious” HIV drug available. The integrase mutation R263K has been observed in tissue culture experiments and in patients treated with dolutegravir monotherapy in clinical trials. Globally, adherence and monitoring may be less than optimal and therefore DTG resistance more common. This is particularly important in low–middle-income countries, where patients may remain on failing regimens for longer periods of time and accumulate drug resistance. Data on this mutation in non–subtype B infections do not exist. We describe the first report of the R263K integrase mutation in a dolutegravir-exposed subtype D–infected individual with vertically acquired HIV. We have used deep sequencing of longitudinal samples to highlight the change in resistance over time while on a failing regimen. The case highlights that poorly adherent patients should not be offered dolutegravir even as part of a combination regimen and that protease inhibitors should be used preferentially.
Article
Full-text available
Background Molecular surveillance of newly diagnosed HIV-infections is important for tracking trends in circulating HIV-variants, including those with transmitted drug resistances (TDR) to sustain ART efficacy. Methods Dried serum spots (DSS) are received together with the statutory notification of a new diagnosis. 'Recent infections' (<155 days) classified by a 'recent infection test algorithm' (BED-CEIA and clinical data) are genotyped in HIV-protease (PR), reverse transcriptase (RT) and integrase (INT) to determine the HIV-1 subtype, to calculate prevalence and trends of TDR, to predict baseline susceptibility and to identify potential transmission clusters for resistant variants. Results Between January 2013 and December 2016, 1,885 recent infections were analysed regarding the PR/RT genomic region, with 43.5% of these also being subjected to the analysis of INT. The proportion of HIV-1 non-B viruses (31.3%; 591/1,885) increased from 21.6% to 36.0%, particularly the subtypes A (5.0% to 8.3%) and C (3.2% to 7.7%; all ptrends < 0.01). The subtype A increment is mainly due to transmissions within men who have sex with men (MSM) while subtype C transmissions are associated with heterosexuals and people who inject drugs. The prevalence of TDR was stable at 11.0% (208/1,885) over the study period. Resistances to nucleotide RT inhibitors (NRTI) and PR inhibitors (PI) were 4.5% and 3.2%, respectively, without identifiable trends. In contrast, resistances to non-NRTIs (NNRTI, 4.7%) doubled between 2014 and 2016 from 3.2% to 6.4% (ptrend = 0.02) mainly due to the K103N mutation (from 1.7% to 4.1%; ptrend = 0.03) predominantly detected in recently infected German MSM not linked to transmission clusters. Transmitted INSTI mutations were present in only one case (T66I) and resistance to dolutegravir was not identified at all. Reduced susceptibility to recommended first-line therapies was low with 1.0% for PIs, 1.3% for NRTIs and 0.7% for INSTIs, but high for the NNRTIs efavirence (4.9%) and rilpivirine (6.0%) due to the K103N mutation and the polymorphic mutation E138A. These trends in therapy-naïve individuals impact current first-line regimens and require awareness and vigilant surveillance.
Article
Full-text available
Background Integrase strand transfer inhibitors (INSTIs) are recommended for first-line HIV therapy based on their relatively high genetic barrier to resistance. Although raltegravir (RAL) and elvitegravir (EVG) resistance profiles are well-characterized, resistance patterns for dolutegravir (DTG), bictegravir (BIC), and cabotegravir (CAB) remain largely unknown. Here, in vitro drug selections compared the development of resistance to DTG, BIC, CAB, EVG and RAL using clinical isolates from treatment-naïve primary HIV infection (PHI) cohort participants (n = 12), and pNL4.3 recombinant strains encoding patient-derived Integrase with (n = 5) and without (n = 5) the E157Q substitution. Results Patient-derived viral isolates were serially passaged in PHA-stimulated cord blood mononuclear cells in the presence of escalating concentrations of INSTIs over the course of 36–46 weeks. Drug resistance arose more rapidly in primary clinical isolates with EVG (12/12), followed by CAB (8/12), DTG (8/12) and BIC (6/12). For pNL4.3 recombinant strains encoding patient-derived integrase, the comparative genetic barrier to resistance was RAL > EVG > CAB > DTG and BIC. The E157Q substitution in integrase delayed the advent of resistance to INSTIs. With EVG, T66I/A, E92G/V/Q, T97A or R263K (n = 16, 3, 2 and 1, respectively) arose by weeks 8–16, followed by 1–4 accessory mutations, conferring high-level resistance (> 100-fold) by week 36. With DTG and BIC, solitary R263K (n = 27), S153F/Y (n = 7) H51Y (n = 2), Q146 R (n = 3) or S147G (n = 1) mutations conferred low-level (< 3-fold) resistance at weeks 36–46. Similarly, most CAB selections (n = 18) resulted in R263K, S153Y, S147G, H51Y, or Q146L solitary mutations. However, three CAB selections resulted in Q148R/K followed by secondary mutations conferring high-level cross-resistance to all INSTIs. EVG-resistant viruses (T66I/R263K, T66I/E157Q/R263K, and S153A/R263K) retained residual susceptibility when switched to DTG, BIC or CAB, losing T66I by week 27. Two EVG-resistant variants developed resistance to DTG, BIC and CAB through the additional acquisition of E138A/Q148R and S230N, respectively. One EVG-resistant variant (T66I) acquired L74M/G140S/S147G, L74M/E138K/S147G and H51Y with DTG CAB and BIC, respectively. Conclusions Second generation INSTIs show a higher genetic barrier to resistance than EVG and RAL. The potency of CAB was lower than BIC and DTG. The development of Q148R/K with CAB can result in high-level cross-resistance to all INSTIs.
Article
Background: Integrase strand-transfer inhibitors (INSTIs) constitute at present one of the pillars of first-line ART. Objectives: To study the prevalence of and the trend in transmitted drug resistance (TDR) to INSTIs in ART-naive patients in Spain. Methods: During the period 2012-17, 1109 patients from CoRIS were analysed. The Stanford algorithm v8.7 was used to evaluate TDR and transmission of clinically relevant resistance. To describe individual mutations/polymorphisms, the most recent IAS list (for INSTIs) and the 2009 WHO list update (for the backbone NRTIs used in combination with INSTIs in first-line treatment) were used. Results: Clinically relevant resistance to the INSTI class was 0.2%: T66I, 0.1%, resistance to elvitegravir and intermediate resistance to raltegravir; and G163K, 0.1%, intermediate resistance to raltegravir and elvitegravir. No clinical resistance to dolutegravir or bictegravir was observed. The prevalence of INSTI TDR following the IAS-USA INSTI mutation list was 2.6%, with no trend towards changes in the prevalence throughout the study period. The overall prevalence of NRTI WHO mutations was 4.3%, whereas clinically relevant resistance to tenofovir, abacavir and emtricitabine/lamivudine was 1.7%, 1.9% and 0.7%, respectively. Conclusions: Given the low prevalence of clinically relevant resistance to INSTIs and first-line NRTIs in Spain, it is very unlikely that a newly diagnosed patient will present with clinical resistance to a first-line INSTI-based regimen. These patients may not benefit from INSTI and NRTI baseline resistance testing.
Article
Objectives: Integrase strand-transfer inhibitor (InSTI)-based regimens are the preferred combinations for naïve HIV-infected individuals. Polymorphic substitutions that reduce InSTIs activity have been described, with E157Q being one of the most frequently found. This study aimed to evaluate the prevalence of E157Q substitution in newly diagnosed acute/recent HIV cases and the presence of transmission clusters. Design: Prospective cohort study in patients with acute/recent HIV infection METHODS:: Genotypic drug resistance tests were performed in all consecutive patients prospectively enrolled with a documented infection of less than six months from May 2015 to May 2017. Sequences were obtained by ultra-deep sequencing. Phylogenetic inferences were performed using maximum likelihood (ML) trees constructed with Mega 6.06. Bootstrap values of 75% or greater were defined for cluster assignment. Follow-up was, at least, one year. Results: In six out of 67 consecutive patients (8.95%, 95% CI 4.17-18.19) with acute/recent HIV infection, strains carrying the E157Q InSTI substitution were detected. All cases were men-who-have-sex-with-men patients infected with subtype B strains. No other resistance substitutions were detected in these cases. Median viral load was 5.33 (IQR: 4.54-5.71) log10 copies/mL and, in all cases, the mutational viral load was high (>95%). Three cases were included in transmission clusters. Three cases responded to dolutegravir-based regimens; non-nucleoside-reverse-transcriptase-inhibitor based regimens were used for the other case (s). Conclusions: E157Q substitution, reducing raltegravir and elvitegravir activity, was frequently found in acute/recent HIV cases. All cases were infected with subtype B and some were included in clusters. Cases treated with dolutegravir-based regimens had good virological response.
Article
Background: Effective two-drug regimens could decrease long-term drug exposure and toxicity with HIV-1 antiretroviral therapy (ART). We therefore aimed to evaluate the efficacy and safety of a two-drug regimen compared with a three-drug regimen for the treatment of HIV-1 infection in ART-naive adults. Methods: We conducted two identically designed, multicentre, double-blind, randomised, non-inferiority, phase 3 trials: GEMINI-1 and GEMINI-2. Both studies were done at 192 centres in 21 countries. We included participants (≥18 years) with HIV-1 infection and a screening HIV-1 RNA of 500 000 copies per mL or less, and who were naive to ART. We randomly assigned participants (1:1) to receive a once-daily two-drug regimen of dolutegravir (50 mg) plus lamivudine (300 mg) or a once-daily three-drug regimen of dolutegravir (50 mg) plus tenofovir disoproxil fumarate (300 mg) and emtricitabine (200 mg). Both drug regimens were administered orally. We masked participants and investigators to treatment assignment: dolutegravir was administered as single-entity tablets (similar to its commercial formulation, except with a different film colour), and lamivudine tablets and tenofovir disoproxil fumarate and emtricitabine tablets were over-encapsulated to visually match each other. Primary endpoint was the proportion of participants with HIV-1 RNA of less than 50 copies per mL at week 48 in the intention-to-treat-exposed population, using the Snapshot algorithm and a non-inferiority margin of −10%. Safety analyses were done on the safety population. GEMINI-1 and GEMINI-2 are registered with ClinicalTrials.gov, numbers NCT02831673 and NCT02831764, respectively. Findings: Between July 18, 2016, and March 31, 2017, 1441 participants across both studies were randomly assigned to receive either the two-drug regimen (n=719) or three-drug regimen (n=722). At week 48 in the GEMINI-1 intention-to-treat-exposed population, 320 (90%) of 356 participants receiving the two-drug regimen and 332 (93%) of 358 receiving the three-drug regimen achieved plasma HIV-1 RNA of less than 50 copies per mL (adjusted treatment difference −2·6%, 95% CI −6·7 to 1·5); in GEMINI-2, 335 (93%) of 360 in the two-drug regimen and 337 (94%) of 359 in the three-drug regimen achieved HIV-1 RNA of less than 50 copies per mL (adjusted treatment difference −0·7%, 95% CI −4·3 to 2·9), showing non-inferiority at a −10% margin in both studies (pooled analysis: 655 [91%] of 716 in the two-drug regimen vs 669 [93%] of 717 in the three-drug regimen; adjusted treatment difference −1·7%, 95% CI −4·4 to 1·1). Numerically, more drug-related adverse events occurred with the three-drug regimen than with the two-drug regimen (169 [24%] of 717 vs 126 [18%] of 716); few participants discontinued because of adverse events (16 [2%] in the three-drug regimen and 15 [2%] in the two-drug regimen). Two deaths were reported in the two-drug regimen group of GEMINI-2, but neither was considered to be related to the study medication. Interpretation: The non-inferior efficacy and similar tolerability profile of dolutegravir plus lamivudine to a guideline-recommended three-drug regimen at 48 weeks in ART-naive adults supports its use as initial therapy for patients with HIV-1 infection. Funding: ViiV Healthcare.