Skip to main content
Erschienen in: BMC Infectious Diseases 1/2015

Open Access 01.12.2015 | Research article

Outcome of patients on second line antiretroviral therapy under programmatic condition in India

verfasst von: Jaya Chakravarty, Shyam Sundar, Ankita Chourasia, Pallav Narayan Singh, Swarali Kurle, Srikanth P. Tripathy, Devidas N Chaturbhuj, Madhukar Rai, Amit Kumar Agarwal, Rabindra Nath Mishra, Ramesh S. Paranjape

Erschienen in: BMC Infectious Diseases | Ausgabe 1/2015

Abstract

Background

The National AIDS Control Organization of India has been providing free second line antiretroviral therapy (ART) since 2008. This observational study reports the survival and virologic suppression of patients on second-line ART under programmatic condition and type of mutations acquired by those failing therapy.

Methods

170 patients initiated on second-line therapy between 2008 and 2012 were followed up till 2013. Viral Load (VL) was repeated at 6 months for all patients and at 12 months for those with VL >400 copies/ml at 6 months. Adequate virological response was defined as plasma HIV-1 VL <400 copies/ml and virological failure was defined as VL >1000 copies/ml. Genotyping was done in 16 patients with virological failure.

Results

Out of 170 patients, 110 (64.7 %) were alive and on therapy and 35 (20.5 %) expired. In the first year the occurrence of death was 13.7 /100 person years while between1 and 5 year it was 3.88 /100 person years. In the first year, duration of immunological failure >12 months, weight <45 kg, WHO clinical stage 3 and 4 and WHO criteria CD4 count less than pretherapy baseline [hazard ratio HR 4.2. 15.8, 11.9 & 4.1 respectively] and beyond first year poor first and second line adherence and first line CD4 count < 200/μL [HR 5.2,15.8, 3.3 respectively] had high risk of death.
119/152 (78.2 %) had adequate virological response and 27/152 (17.7 %) had virological failure. High viral load at baseline and poor second line adherence (Odds Ratio 3.4 & 2.8 respectively) had increased risk of virological failure. Among those genotyped, 50 % had major Protease Inhibitor mutation (M46I commonest) however 87.5 % were still susceptible to darunavir.

Conclusions

Second line therapy has shown high early mortality but good virological suppression under programmatic conditions.
Hinweise
Jaya Chakravarty and Shyam Sundar contributed equally to this work.

Competing interests

None of the authors have any conflict of interest.

Authors’ contribution

JC & SS contributed towards conception, design, data collection, interpretation of data and drafting the manuscript. PNS, MR. AKA, RNM contributed towards data collection, interpretation of data and drafting the manuscript. AC, DNC, SK, SPT,RSP contributed towards data collection, genotyping and drafting the manuscript. All authors read and approved the manuscript.
Abkürzungen
ART
antiretroviral therapy
ATV/r
atazanavir/ ritonavir
bPI
boosted Protease Inhibitors
DRV
darunavir
ESS
etravirine susceptibility scores
GSS
genotype sensitivity scores
HIVDR
human immunodeficiency virus drug resistance
LFU
lost to follow up
LPV/r
lopinavir/ritonavir
NACO
National AIDS control organization
NNRTI
nonnucleoside reverse transcriptase inhibitors
NRTI
nucleoside reverse transcriptase inhibitors
PLHIV
people living with HIV,VL-viral load
RLS
resource limited settings
TAM
thymidine analogue mutations
TR out
transfer out

Background

Of the 4.8 million people living with HIV (PLHIV) in Asia, nearly half (49 %) are in India. There has been a massive scale-up of antiretroviral therapy (ART) services in India since the National AIDS Control Organization (NACO) launched the ART centres providing free antiretroviral drugs in 2004. As of September 2014, there were 453 fully functional ART centres across the country providing free first line ART to 810,339 PLHIVs [1]. With the increase in number of patients on first line therapy it is envisioned that a proportion of patients will experience treatment failure and need second line ART regimens over time. In 2008, NACO piloted a national strategy for the provision of free second-line ART in India and these drugs are being provided at 10 Centres of Excellence and 16 ART Plus Centre. As on September 2014, 10,223 patients were receiving second line drugs in the National program [1].
Similar to other resource-limited settings (RLS), access to routine viral load testing to monitor therapy or genotyping before start of second line is not available in the program. Patients suspected of treatment failure on first line regimens based on WHO defined immunologic or clinical criteria are subjected to viral load testing, and only those with definitive virologic failure qualify for switch to second-line ART. Like other RLS the only second line ART available consists of a boosted protease inhibitor (bPI) with two nucleoside reverse transcriptase inhibitors (NRTIs).
Amongst the RLS, reports of efficacy and survival of patients on second line regimens are available mostly from Africa [24]. Although the second line ART program has been launched by NACO for six years studies to evaluate the survival and efficacy of the prevailing second line regimen are lacking in the country. A recent study from India has shown an excellent early outcome of second line treatment [5]. In the near future, NACO plans to consolidate the first and second line treatment and launch third line ART [6]. Therefore, there is an urgent need to evaluate the efficacy of the second line therapy and to know the mutations acquired by those failing therapy for the selection of an appropriate third line regimen. This study was done to report the survival and virologic suppression and their predictors in patients receiving Protease Inhibitor (PI) based second-line ART under programmatic condition and to assess the mutations acquired by those failing second line therapy.

Methods

Study site

This observational study was conducted at the ART Centre of the Centre of Excellence (COE), of Banaras Hindu University (BHU). The ART centre is one of the largest in this region with 18,746 PLHIV registered in HIV care and 4530 currently on first line ART. It is also a referral centre for evaluation of patients suspected of first line failure from 14 ART centres.

Study population

The study was approved by the Ethics Committee of the Institute of Medical Sciences, Banaras Hindu University. Data of all patients >15 years of age who were started on second line therapy due to failure of first line ART at COE BHU since the beginning of the program in December 2008 to December 2012 were included in this study. Written informed consent from patients or their guardians for children were taken for HIV drug resistance genotyping.
The first line regimen recommended by NACO and received by patients was Zidovudine + Lamivudine + Nevirapine if hemoglobin was >9gm/dl or Stavudine + Lamivudine + Nevirapine if hemoglobin <9gm/dl. Efavirenz was substituted for Nevirapine in those taking antitubercular drugs and those with Nevirapine toxicity. Patients on first line therapy were eligible for evaluation for second-line ART if they had been receiving ART for at least 6 months, and had demonstrated treatment adherence of >95 %, and had WHO clinical or immunological failure as per NACO guidelines. Viral load estimation was done in these patients and those with HIV RNA >10,000 copies/mL were considered as first line failure and started on second line therapy [7]. In May 2011, the cut off level of viral load for starting ART was reduced to >5000 copies/ml by NACO [8].
All patients with first line failure between 2008 and April 2011 were given a uniform second-line ART regimen provided by NACO comprising of tenofovir (TDF) + lamivudine (3TC) + zidovudine (AZT) + lopinavir/ritonavir (LPV/r). Zidovudine was excluded from the regimen in patients with significant anemia (hemoglobin less than 9 g/dL) or previous history of zidovudine induced anemia. After May 2011, the second line ART provided by NACO was tenofovir with lamivudine and Atazanavir/ritonavir (ATV/r).All new patients and those who were previously on lopinavir based regimen were shifted to this regimen [8]. In patients with concomitant tuberculosis rifampicin was replaced by rifabutin without any change in the ART regimen.
Patients were followed up monthly and CD4 count (FACS Calibur Becton Dickinson Biosciences) was done 6 monthly for all patient. During each visit, patients were counselled for adherence and evaluated for drug toxicity, clinical improvement and opportunistic infections. Patient’s weight, clinical stage, functional status, drug toxicity, adherence to ART medication, presence of opportunistic infection, any change in therapy were documented. Viral load (VL) was repeated by COBAS TaqMan HIV-1 assay in all patients at 6 months, if it was <400copies/ml at 6 months it was not repeated further as per National guidelines. Patients with VL >400 copies/ml at 6 months, adherence was reinforced and VL was repeated at 12 months. Adherence was calculated on the basis of pill count at every visit by the formula: Number of pills actually taken by a patient for a particular time period/Number of pills prescribed for this time period × 100. For analysis we compared those with >95 % adherence at every visit with those with <95 % at any visit. At the end of the month patients were labelled as ‘on treatment’ if they picked up their drugs, ‘missed’ if they did not pick up drugs for the month, dead if they expired and ‘transferred out’ if they were transferred out to another ART centre. Those patients who did not come for 3 consecutive months were labelled as lost to follow up (LFU) at the end of fourth month as per NACO guidelines.

HIV drug resistance (HIVDR) genotyping

The HIVDR genotyping was performed in patients with viral load >1000 copies/ml on two occasions after 6 months of second line therapy. Plasma samples stored at −70 °C were used for detection of HIVDR mutations by in-house assay described in a previous study [9]. RNA extraction from plasma samples were performed using the NucliSENSEasyMAGTM (Biomerieux, Durham, NC) automated nucleic acid extraction system according to the manufacturer’s recommendations (NucliSENSEasyMAG user manual, v1.1; BioMe’rieux, Boxtel, Netherlands). The RobusT one step RT-PCR kit (FinnzymesOy, Finland) was used to amplify the complete protease and RT region of the pol gene. RT-PCR was performed as described previously using 2021 F and 4521R primers [10]. The nested PCR was performed using the inner primer pair2135F and 3338R to get an amplified fragment of 2336 bp. A Gene Amp PCR system 9700 thermal cycler (Applied Biosystem, CA, USA) was used for all PCRs. DNA sequencing was performed on 3100 DNA genetic analyzer (Applied Biosystem, CA, USA) using a set of six specific primers (2135 F, 2493 F, 3012 F, 2557R, 3117R, 3338R). The raw sequence data from ABI 3100 genetic analyzer was assembled, aligned and edited with the SeqScape v2.0 software (Applied Biosystem, CA, USA). The quality of the sequences was assessed using the SQUAT software. The HIVDR mutational analysis was performed using the online “HIVdb Program: Sequence Analysis” program from Stanford University [11].
Genotype sensitivity scores (GSS) for the prescribed regimen at the time of second line failure were calculated based on the five Stanford HIVdb resistance categories: susceptible or complete activity, potential low-level resistance or good activity, low-level resistance or partial activity, intermediate-level resistance or scarce activity, high-level resistance or no activity corresponding with scores of 1.00, 0.75, 0.50, 0.25, and 0.00, respectively [12, 13]. Etravirine susceptibility scores (ESS) were calculated using the scoring system described by Vingerhoets et al. [14] and recommended by the IAS-USA.

Statistical analysis

Data was extracted and was analysed using SPSS version 16.0. The data was presented as mean ± standard deviation (SD) for continuous variables and frequency with their respective percentages for categorical variables. Patient characteristics was described in terms of median with their inter quartile range (IQR) for skewed continuous data. For categorical data Chi-square test and Fischer Exact test was used and for continuous data Student’s t test and Mann Whitney U test was used.
For efficacy analysis, virological response at one year of therapy was observed. Adequate virological response was defined as plasma HIV-1 VL <400 copies/ml, VL between 400 and 1000 copies /ml was defined as on-going viremia and VL >1000 copies/ml was used to define virological failure or treatment failure at one year. Efficacy data was analyzed by an intention-to-treat (ITT) i.e. among all patients included in the study and on-treatment (OT) approach i.e. among those patients who at least had a VL at six month. VL at six month was included for analysis for those patients eligible for twelve month viral load but it could not be done due to death or LFU and for those who had adequate virological suppression at 6 months.
To assess the predictors of treatment failure patients with VL >1000 copies/ml were compared with those with VL < 400copies/ml. To define predictors of treatment failure, univariate analyses were performed with the following determinants: age, gender, first-line ART regimen (zidovudine versus stavudine), weight, CD4 count, clinical stage at the start of first and second-line treatment, presence of tuberculosis, adherence, duration of treatment of first and second-line therapy. A multivariate analysis was performed by using the binary logistic regression model, including all variables that were associated with the outcome (P < 0.05) in univariate analysis. To compare the efficacy of LPV/r and ATV/r based second line regimen, virological suppression was assessed at one year by comparing patients started on LPV/r based second line treatment between 2008 and April 2010 and a VL report before change of therapy to ATV/r with those newly started on ATV/r based regimen after May 2011with 6 or 12 month VL.
In survival analysis, patients started on second line therapy between 2008 and 2012 were included. Status of these patient was assessed in December 2013, so that all subjects had the potential to complete a minimum of 1 year of follow-up. The main outcome variable was death and the time of its occurrence during the follow up period. The data was treated as censored when either patients were lost-to follow up or transferred-out to other ART centres. The predictor variables used in the analysis were duration of immunological failure, WHO criteria for immunological failure, weight, WHO clinical stage, CD4 count, presence of tuberculosis, adherence at the start of both first and second line treatment and age, sex, viral load at the beginning of second line treatment. Variables that were statistically significant (p value <0.05) in the bivariate analysis were subsequently considered for multivariate analysis (Cox proportional hazard model) to assess the relationship between these variables and mortality.

Results

Two hundred two patients were started on second line ART between December 2008 and December 2012, out of which 7 were already on therapy from outside and 25 were transferred out to other centre before six month viral load thus only 170 patients were included in the analysis.
Baseline characteristics of patient started on second line therapy are given in Table 1. 82.9 % were males, median duration of second line therapy was 22.50 months (IQR-14-40.25). 33 (19.4 %) patients had tuberculosis during second line treatment and adherence during second line was > 95 % in 132 (77.6 %) patients. All patients received TDF and 3TC as a part of second line regimen. Among PI, 12 patients received only LPV/r (as they had expired before change to ATV/r), 79 received both LPV/r and ATV/r and 79 received only ATV/r based PI regimen. Median CD4 count at the start of second line therapy was 78.50/μL (IQR 49.75–121.25), at 12 months 273/μL (IQR 182–357), at 24 month 319/μL (IQR 221–452), at 36 month 315/μL (IQR 209–467), at 48 month 285/μL (IQR 242–473) and at 60 month 343 /μL (IQR 228–504).
Table 1
Baseline characteristics of patients on second line (2nd Line) ART
Variable
Number (n = 170)
Age(years)
Mean ± SD
36.78 ± 7.30
Median (IQR)
35.0 (32.0-42.0)
Sex
Male n (%)
141 (82.9)
Female n (%)
29 (17.1)
Duration of Immunological failure (months)
Mean ± SD
17.30 ± 15.71
 
Median (IQR)a
12 (6.0-23.0)
Weight(Kg) at the start of 2nd line ART
Mean ± SD
48.34 ± 9.07
Median (IQR)a
49.0 (41–55)
Clinical Stage at the start of 2nd line ART
I + II n (%)
118 (69.4)
III + IV n (%)
52 (30.6)
CD4 count at the start of 2nd line ART (/μL)
Mean ± SD
94.36 ± 72.98
Median (IQR)a
78.50 (49.75-121.25)
Viralload baseline (copies/ml)
Mean ± SD
490000.00 ± 1150000.00
Median (IQR)a
1770000 (67375–3540000)
Last NRTI regimen
Stavudine n (%)
81 (47.6)
Zidovudine n (%)
89 (52.4)
Total Duration of 2nd line ART (months)
Mean ± SD
26.65 ± 16.76
Median (IQR)a
22.50 (14–40.25)
Tuberculosis during 1st line ART
Yes n (%)
89 (52.4)
No n (%)
81 (47.6)
Tuberculosis during 2nd line ART
Yes n (%)
33 (19.4)
No n (%)
137 (80.6)
1st line ART Adherence
Adherence (<80-95 % ) n (%)
46 (27.1)
Adherence (>95 %) n (%)
124 (72.9)
2nd line ART Adherence
Adherence (<80 %-95 %) n (%)
38 (22.4)
Adherence (>95 %) n (%)
132 (77.6)
aInter quartile range

Survival

At the end of follow up, out of the 170 patients started on second line therapy, 35 (20.6 %) had expired, 5 (2.9 %) were lost to follow up, 20 (11.8 %) were transferred out to other ART centres and 110 (64.7 %) were alive and on ART.
Out of the 35 patients who died, 21 (60 %) expired within 12 months of starting second line. Among them 10 had tuberculosis, one each had pneumonia, oesophageal candidiasis and diarrhea and no cause was documented in 8 patients. Among these 21 patients, viral load at 6 month was not done in 17 patients due to death, 1 had virological failure, 2 patients had adequate virological suppression and 1 had ongoing viremia.
14 patients expired after 1 year of second line. Among them 2 had tuberculosis, one each had anal carcinoma, carcinoma penis, chronic renal failure and HIV encephalopathy. Among these 14 patients 6 had virological failure, 1 had ongoing viremia, rest had adequate virological suppression. The risk of death in the first year of therapy was 13.7/100 person years while between 1 and 5 year it was 3.88/100 person years.
In the first year duration of immunological failure >12 months [HR 4.218; 95 % CI: 1.146-15.519], presence of WHO immunological criteria CD4 count less than pretherapy (at the start of first line) baseline [HR 4.111; 95 % CI: 1.050-16.096]; weight <45 kg [HR 15.777; 95 % CI: 1.734 -143.535],WHO clinical stage 3 and 4 [HR 11.871; 95 % CI: 2.695-52.296] had higher risk of death. Beyond first year, patients with CD4 count less than pretherapy baseline [HR 3.328; 95 % CI: 1.031-10.742], poor adherence during first line [HR 5.226; 95 % CI: 1.587-17.210] as well as during second line therapy [HR 15.838; 95 % CI: 4.274-58.685] had higher risk of death (Table 2).
Table 2
Univariate and multivariate analysis of survived and expired patients within 1 year and beyond 1 year
 
Within 1 year
1-5 year
Characteristics
Uni-variate
p- value
Multi-variate
p- value
Uni-variate
p-value
Multi-variate
p-value
HR (95 % CI)
HR (95 % CI)
HR (95 % CI)
HR (95 % CI)
Age (years)
 >40
1.718 (0.712-4.145)
0.229
  
2.508 (0.879-7.154)
0.086
  
 ≤40
1
   
1
   
Sex
 Male
2.059 (0.480-8.839)
0.331
  
0.508 (0.158-1.630)
0.255
  
 Female
1
   
1
   
WHO Stage 1st line
 III + IV
2.616 (1.111-6.162)
0.028
0.710 (0.202-2.499)
0.594
1.956 (0.677-5.652)
0.215
  
 I + II
1
 
1
 
1
   
Tuberculosis 1st line
 Yes
1.218 (0.513-2.891)
0.654
  
1.001 (0.346-2.894)
0.998
  
 No
1
   
1
   
Adherence 1st line
 <95 %
1.115 (0.433-2.874)
0.822
  
5.673 (1.778-18.094)
0.003
5.226 (1.587-17.210)
0.007
 >95 %
1
   
1
 
1
 
Weight (Kg) 1st line
 ≤45
0.867 (0.350-2.149)
0.759
  
2.407 (0.826-7.013)
0.107
  
 >45
1
   
1
   
CD4 count 1st line (/μL)
 <200
4.989 (0.670-37.178)
0.117
  
28.304 (0.085-924.0)
0.259
  
 >200
1
   
1
   
Duration of Immunological failure (months)
 >12
3.262 (1.182-11.123)
0.024
4.218 (1.146-15.519)
0.030
2.684 (0.824-8.745)
0.101
  
 < 12
1
 
1
 
1
   
Clinical failure
 Yes
2.721 (1.098-6.743)
0.031
1.056 (0.240-4.645)
0.943
0.640 (0.143-2.869)
0.560)
  
 No
1
 
1
 
1
   
CD4 count less than baseline
 Yes
3.579 (1.204-10.638)
0.022
4.111 (1.050-16.096)
0.042
2.120 (0.709-6.343)
0.179
3.328 (1.031-10.742)
0.044
 No
1
 
1
 
1
 
1
 
50 % fall from peak CD4 count
 Yes
0.448 (0.174-1.154)
0.096
  
25.70 (0.038-1719.0)
0.328
  
 No
1
   
1
   
CD4 count < 100/μL
 Yes
2.100 (0.815-5.413)
0.125
  
3.038 (0.847-10.895)
0.088
  
 No
1
   
1
   
CD4 count 2nd line (/μL)
 <200
0.644 (0.150-2.765)
0.554
  
22.856 (0.003-1692.0)
0.491
  
 >200
1
   
1
   
WHO Stage 2nd line
 III + IV
15.854 (4.666-53.872)
<0.001
11.871 (2.695-52.296)
0.001
1.446 (0.484-4.318)
0.509
  
 I + II
1
 
1
 
1
   
Weight (Kg) 2nd line
 ≤45
6.491 (1.512-27.877)
0.012
15.777 (1.734-143.535)
0.014
1.089 (0.363-3.269)
0.879
  
 >45
1
 
1
 
1
   
Viral Load 2nd Line (copies/ml)
 >177000
2.042 (0.824-5.061)
0.123
  
0.790 (0.274-2.279)
0.662
  
 ≤177000
1
   
1
   
Tuberculosis 2nd line
 Yes
4.219 (1.791-9.939)
0.001
2.336 (0.752-7.256)
0.142
0.854 (0.190-3.837)
0.837
  
 No
1
 
1
 
1
   
Adherence 2nd line
 <95 %
1.483 (0.575-3.821)
0.415
  
14.682 (4.093-52.660)
<0.001
15.838 (4.274-58.685)
<0.001
 >95 %
1
   
1
 
1
 

Efficacy

At 6 months viral load was done in 152 patients as 17 had expired and 1 was LFU. 102 patients had a viral load of <400 copies/ml, 12 had viral load between 400 and 1000 copies/ml and 38 had viral load >1000 copies/ml. At 12 months viral load was repeated in only 41 patients out of the 50 patients whose VL was >400 copies/ml at six month, as 5 expired and 4 were LFU. Among these 41 patients, 17 had VL <400 copies/ml, 5 had VL between 400 and 1000 copies/ml and 19 had viral load >1000 copies/ml.
Overall, out of 170 patients who were started on second line, 119/170 i.e. 70 % by ITT and 119/152 i.e. 78.2 % by OT had adequate virological response, 6 (3.5 % ITT, 3.9 % OT) had ongoing viremia and 27 (15.8 % ITT, 17.7 % OT) had virological failure at one year. Viral load ≥177,000 copies/ml at baseline (OR 3.402, 95 % CI: 1.272-9.097) and <95 % adherence during second line treatment (OR 2.788, 95 % CI: 1.044-7.445) was significantly associated with second line failure (Table 3). On comparing the efficacy of LPV/r vs ATV/r, the two PIs used as second line, 85.2 % (46/54) had virological suppression with LPV/r while 69 % (49/71) had virological suppression with ATV/r at one year.
Table 3
Univariate and multivariate analysis between virological Failure and adequate virological response
Characteristics
Uni-variate
p-value
Multi-variate
p-value
 
Odds ratio (95 % CI)
 
Odds ratio (95 % CI)
 
Age (years)
 >40
1.249 (0.496-3.147)
0.637
  
 ≤40
1
   
Sex
 Male
0.423 (0.161-1.112)
0.081
  
 Female
1
   
WHO Stage 1st line
 III + IV
1.038 (0.400-2.698)
0.938
  
 I + II
1
   
Tuberculosis 1st line
 Yes
2.327 (0.968-2.327)
0.059
  
 No
1
   
Adherence 1st line
 <95 %
2.600 (1.090-6.201)
0.031
1.918 (0.741-4.962)
0.180
 >95 %
1
 
1
 
Weight (Kg) 1st line
 ≤45
1.215 (0.517-2.854)
0.655
  
 >45
1
   
CD4 count 1st line (/μL)
 <200
3.618 (1.455-8.998)
0.006
2.788 (1.045-7.439)
0.041
 >200
1
 
1
 
Duration of Immunological failure (months)
 >12
0.542 (0.229-1.283)
0.164
  
 <12
1
   
Clinical failure
 Yes
5.253 (0.673-40.976)
0.114
  
 No
1
   
CD4 count less than baseline
 Yes
2.335 (0.949-5.749)
0.065
  
 No
1
   
50 % fall from peak CD4 count
 Yes
1.092 (0.339-3.519)
0.882
  
 No
1
   
CD4 count < 100/μL
 Yes
0.688 (0.297-1.593)
0.382
  
 No
1
   
CD4 count 2nd line (/μL)
 <200
3.275 (0.856-12.537)
0.083
  
 >200
1
   
WHO Stage 2nd line
 III + IV
1.287 (0.477-3.477)
0.618
  
 I + II
1
   
Weight (Kg) 2nd line
 ≤45
1.143 (0.492-2.653)
0.756
  
 >45
1
   
Viral Load 2nd Line (copies/ml)
 >177000
3.558 (1.399-9.050)
0.008
3.402 (1.272-9.097)
0.015
 ≤177000
1
 
1
 
Tuberculosis 2nd line
 Yes
1.364 (0.455-4.090)
0.580
  
 No
1
   
Adherence 2nd line
 <95 %
2.870 (1.176-7.004)
0.021
2.788 (1.044-7.445)
0.041
 >95 %
1
 
1
 

HIVDR genotyping

Out of the 27 patients who failed second line therapy 3 expired and 2 were LFU and sample for genotyping could not be collected in 3 patients. Out of the 19 samples available, genotyping data of only 16 patients could be included in the study as sample (SL) 7, 17 and 18 did not amplify. All samples were HIV 1 virus subtype C. 50 % (8/16) had major PI mutation, 62.5 % (10/16) had minor PI mutation, 81.25 % (13/16) had NRTI mutation and 93.75 % (15/16) had NNRTI (non nucleoside reverse transcriptase) mutation (Table 4). Median GSS was 1.0 (0–1.81).
Table 4
Mutations acquired by patients failing second line treatment
Patient ID
Gene Bank
Accession No.
Duration of second line treatment (months)
Adherence during 2nd line treatment
Subtype
Major
PI mutation
Minor PI mutation
NRTI mutation
NNRTI mutation
GSS score
Etravirine sensitivity score
SL-1
KJ933454
46
>95 %
C
M46I, I47A, I50IL, I84V
A71V
M41L, D67N, K70R, L74IL, M184V, T215Y, K219Q
K103N, K238KN
0
0
SL-2
KJ933455
13
>95 %
C
NONE
NONE
M41L, D67N, M184V, T215F, K219W
Y181V, G190A
1.25
4
SL-3
KJ933456
23
>95 %
C
M46I, N88S
L24I, Q58E, A71V
M41L, K65R, K70T, M184V
A98G, K101E, G190A
0
3
SL-4
KJ933457
9
>95 %
C
NONE
NONE
M184V
K101E, G190A
2
2
SL-5
KJ933458
20
>95 %
C
M46I
L23IL
D67N, V75M, M184V
Y181C
1.25
2.5
SL-6
KJ933459
24
>95 %
C
None
K20I, L90LW
None
V108IV, Y181C, H221Y
3
2.5
SL-9
KJ933461
25
<95 %
C
M46I, I50L, V82A
L33FL, A71AV, G73S, N83DN
M41L, D67N, V75M, M184V, L210W, T215Y, K219N
A98G, K103N
0
1
SL-11
KJ933463
9
>95 %
C
M46I, N88NS
K20IT, E35DEG, A71AT, T74S
M41L, M184V, L210W, T215Y, K219KN
K101H, Y181CFIS, G190A
0
7.5
SL-12
KJ933464
14
>95 %
C
NONE
K20KT, G73S
M41L, D67E, V75M, M184V, L210W, T215Y
V90I, V108I, Y181C
0.5
3.5
SL-13
KJ933465
10
>95 %
A1C
I84IV
L23I, A71V
M41L, D67N, L74I, V75M, M184V, L210W, T215Y, K219N
A98G, K103N, G190
0
2
SL-14
KJ933466
9
<95 %
C
None
None
None
None
3
0
SL-15
KJ933467
20
>95 %
C
NONE
NONE
M41L, D67N, V75M, M184V, T215Y, K219N
V90I, K103N
1.25
1
SL-16
KJ933468
20
>95 %
C
NONE
T74S
M41L, K70R, L74I, M184V, T215F, K219W
V106M, V108I, Y181C, F227L
1.25
2.5
SL-19
KJ933469
12
<95 %
C
NONE
NONE
NONE
K101E
3
1
SL-20
KJ933470
36
>95 %
C
M46I
NONE
M41L, D67N, T69D, V75M, M184V, L210W, T215Y
A98G, K101EK, V108I, V179IT, G190A, F227L
0.75
4
SL-21
KJ933471
18
<95 %
C
N88S
L10F, K20T, Q58E
D67N, T69D, K70R, M184V, T215F, K219Q
K101E, Y181C
0.25
3.5
Among PI mutations, M46I (n = 5) was the commonest mutation, followed by N88S (n = 3), I50L (n = 2), I84V (n = 2), V 82A (n = 1). 6 patients with >12 months of second line therapy as compared to 2 patients with <12 months therapy had at least one major PI mutation. Fourteen (87.5 %) patients were susceptible to darunavir (DRV) while two were associated with low level resistance to DRV. 6 (37.5 %) patients were associated with high resistance, 3 (18.75 %) had low level resistance and 7 (43.75 %) were still susceptible to atazanavir (ATV).
M184V was the commonest NRTI mutation and was present in 13 out of 16 patients. 62.5 % (n = 10) patients had ≥3 TAM (Thymidine analogue mutations). 81.25 % (n = 13) had more than 1 NNRTI mutation.Y181C and G190A was the most common NNRTI mutation present in 37.5 % (n = 6) patient each. ESS were ≥ 2.5 in 56.25 % (9/16) patients.

Discussion

This study reports the outcome of patients receiving second line antiretroviral therapy under the National AIDS control programme of India. Retention in care at the end of follow up was similar to a study from rural South Africa where routine virological monitoring is done. While in our study mortality was higher, lost to follow up was much lesser than this cohort from South Africa [14]. Early mortality was high in our study but was comparable to a study from Malawi where WHO defined immunological and clinical failure criteria were used to detect first line failure similar to our study [2]. Risk factor for early death (within one year) in our study was duration of immunological >12 months, presence of WHO immunological criteria, CD4 count less than pretherapy, weight <45 kg, WHO clinical stage III &IV, all of which suggests that patients failing first line for a long time and those in poor clinical condition at the start of second line were at increased risk of death. Similar findings were observed in the study from Malawi where clinical failure at baseline and body mass index <18.5 were risk factors for death [2].
These finding suggests that delay in detecting first line failure may be the main reason for this high early mortality in our study. Studies have shown that WHO immunological failure criteria have low sensitivity for detecting virological failure [15, 16]. Moreover, the cut off viral load level for starting second line in our study was much higher (5000–10,000 copies/ml) as compared to other studies [2, 14]. A recent study from Africa has shown that delayed switch of antiretroviral therapy after virological failure is associated with increased mortality [17]. Decreasing the cut–off level for viral load for starting second line to 1000 copies/ml and ensuring early referral through education of medical officers are some of the initiatives taken by NACO which will decrease the mortality among second line patients in future. However, increasing the accessibility of viral load testing as recommended by the recent WHO guidelines would definitely go a long way in improving the second line program.
Efficacy analysis showed that 119 (70 % by ITT, 78.2 % OT) had adequate virological response at one year. Similar findings were observed in studies from other resource limited settings [4, 14], while a study from India showed slightly better outcome [5]. A recent large trial from Africa has shown adequate virological suppression in 86 % of patients at 96 weeks with the WHO recommended second line regimen of NRTI and boosted PI [18]. High early mortality, higher viral load cut off for starting therapy in our study could be the reasons for the difference in virological response from this study.
Poor adherence during second line therapy was an important risk factor for virological failure in our study similar to other studies [2, 3, 19]. Poor adherence was also a major risk for death beyond first year in our study. The fact that 50 % (7/14) of our patients who died after one year of therapy did not have adequate virological suppression, suggests that poor adherence might have led to virological failure and death.
Interestingly, 17/50 (34.0 %) of our patients who had ongoing viremia or virological failure at 6 months adequately suppressed their viral load at 12 months. Similar findings were observed in a study where 62 % patients suspected of second-line ART failure, responded to enhanced adherence support and had a two-log decrease in their level of HIV on subsequent VL testing [20]. These findings further endorses the WHO guidelines which recommends that patients failing virologically be subject to an adherence support intervention, after which a second viral load test should be performed prior to deciding on a regimen change. High viral load at baseline as a risk factor for virological failure was also recently observed in the results of TREAT Asia HIV observational database [21].We observed that virological suppression was better in those with LPV/r as compared to ATV/r, however, as this analysis included only a small subset of patients further studies are needed to come to any conclusions.
This is the first study to report the mutations acquired by second line failure patients in the National program. Median GSS was very low (1.0) in our study. 62.5 % patients had ≥3 TAM which they must have acquired during first line therapy and suggests that patients were failing on first line therapy long before detection. It also implies that they were relatively on PI monotherapy and at risk of developing PI mutations. 93.75 % of the patients in our study had NNRTI mutation, which may be due to re-emergence of archived mutations from first-line NNRTI regimens. Etravirine susceptibility scores (ESS) was ≥ 2.5 in 56.25 % patients, which is a concern as best virologic suppression are seen when the ESS score is less than or equal to 2.0 and this drug is also a part of the WHO recommended third line regimen. 50 % of our second line failure patients had major PI mutation which is much higher than previous studies [22, 23] but is similar to a recent study from Nigeria and a tertiary care centre in India [24, 25]. Patients with longer duration of therapy had more chances of acquiring PI mutation in our study, similar to the study from Nigeria [24]. Studies have shown that several Gag substrate mutations can cause drug resistance mutations that confer PI resistance in the absence of protease mutations [2628]. This could explain the fact that only 50 % of the patients had PI mutation while all of them were failing second line. M46I was the commonest PI mutation observed in our study which reduces susceptibility to all PI except darunavir [11]. Similar findings were observed in other studies from India, [25, 29]. As M46I mutation has also been observed in few PI naïve patients we could have slightly overestimated the selection of this mutation in our study as genotyping was not performed before the start of second line therapy [30, 31]. N88S was the second most common PI mutation in our study which is associated with resistance to ATV/r and nelfinavir. Only two patients had I84V mutation which confers broad spectrum resistance to all PIs and low level resistance to DRV. Thus, most patients were still susceptible to DRV the recommended PI for third line regimen by WHO.
The major limitations of our study was the inability to do viral load testing for all patients at 12 months and the fact that genotyping was not done at the start of second line therapy.

Conclusion

In this observational study there was high early mortality but good long term outcome as well as virological suppression in patients starting second line therapy under programmatic conditions in India. This early mortality can be circumvented by introducing routine virological monitoring in the program which will help in early detection of patients with failure. Virological response in our cohort was similar to other resource poor settings. Although 50 % of our second line failure patients had major PI mutation most were still susceptible to darunavir. Thus, darunavir in combination with integrase inhibitor which has not been used in the program remains a good option as a third line ART for the National program.

Acknowledgement

We gratefully acknowledge the funding support provided by COE, NACO (M-18017/72/ 2007/NACO (GF Rd6), India and Indian Council of Medical Research (ICMR), New Delhi for sponsoring the HIVDR genotyping under the HIV drug resistance database project (File No. AMR/28/2011-ECD-I).
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Competing interests

None of the authors have any conflict of interest.

Authors’ contribution

JC & SS contributed towards conception, design, data collection, interpretation of data and drafting the manuscript. PNS, MR. AKA, RNM contributed towards data collection, interpretation of data and drafting the manuscript. AC, DNC, SK, SPT,RSP contributed towards data collection, genotyping and drafting the manuscript. All authors read and approved the manuscript.
Literatur
1.
Zurück zum Zitat NACO: Annual report 2014–15. Department of AIDS Control, Ministry of Health and Family Welfare, Government of India. NACO: Annual report 2014–15. Department of AIDS Control, Ministry of Health and Family Welfare, Government of India.
2.
Zurück zum Zitat Hosseinipour MC, Kumwenda JJ, Weigel R, Brown LB, Mzinganjira D, Mhango B, et al. Second-line treatment in the Malawi antiretroviral programme: high early mortality, but good outcomes in survivors, despite extensive drug resistance at baseline. HIV Med. 2010;11(8):510–8.PubMed Hosseinipour MC, Kumwenda JJ, Weigel R, Brown LB, Mzinganjira D, Mhango B, et al. Second-line treatment in the Malawi antiretroviral programme: high early mortality, but good outcomes in survivors, despite extensive drug resistance at baseline. HIV Med. 2010;11(8):510–8.PubMed
3.
Zurück zum Zitat Fox MP, Ive P, Long L, Maskew M, Sanne I. High rates of survival, immune reconstitution, and virologic suppression on second-line antiretroviral therapy in South Africa. J Acquir Immune Defic Syndr. 2010;53(4):500–6.CrossRefPubMed Fox MP, Ive P, Long L, Maskew M, Sanne I. High rates of survival, immune reconstitution, and virologic suppression on second-line antiretroviral therapy in South Africa. J Acquir Immune Defic Syndr. 2010;53(4):500–6.CrossRefPubMed
4.
Zurück zum Zitat Ajose O, Mookerjee S, Mills EJ, Boulle A, Ford N. Treatment outcomes of patients on second-line antiretroviral therapy in resource-limited settings: a systematic review and meta-analysis. AIDS. 2012;26(8):929–38.CrossRefPubMed Ajose O, Mookerjee S, Mills EJ, Boulle A, Ford N. Treatment outcomes of patients on second-line antiretroviral therapy in resource-limited settings: a systematic review and meta-analysis. AIDS. 2012;26(8):929–38.CrossRefPubMed
5.
Zurück zum Zitat Patel D, Desai M, Shah AN, Dikshit RK. Early outcome of second line antiretroviral therapy in treatment-experienced human immunodeficiency virus positive patients. Perspect Clin Res. 2013;4(4):215–20.PubMedCentralCrossRefPubMed Patel D, Desai M, Shah AN, Dikshit RK. Early outcome of second line antiretroviral therapy in treatment-experienced human immunodeficiency virus positive patients. Perspect Clin Res. 2013;4(4):215–20.PubMedCentralCrossRefPubMed
6.
Zurück zum Zitat National AIDS Control program phase IV (2012–2017). Strategy document. Department of AIDS Control. Ministry of Health and Family Welfare, Government of India. National AIDS Control program phase IV (2012–2017). Strategy document. Department of AIDS Control. Ministry of Health and Family Welfare, Government of India.
7.
Zurück zum Zitat WHO: World Health Organization. Antiretroviral Therapy for HIV Infection in Adults and Adolescents in Resource-Limited Settings. Geneva: Towards Universal Access; 2006. WHO: World Health Organization. Antiretroviral Therapy for HIV Infection in Adults and Adolescents in Resource-Limited Settings. Geneva: Towards Universal Access; 2006.
8.
Zurück zum Zitat NACO. National AIDS Control Organisation. New Delhi, India: Ministry of Health and Family Welfare Government of India. National Guidelines on Second-line ART for adults and adolescents; 2011. NACO. National AIDS Control Organisation. New Delhi, India: Ministry of Health and Family Welfare Government of India. National Guidelines on Second-line ART for adults and adolescents; 2011.
9.
Zurück zum Zitat Chaturbhuj DN, Nirmalkar AP, Paranjape RS, Tripathy SP. Evaluation of a cost effective in-house method for HIV-1 drug resistance genotyping using plasma samples. PLoS One. 2014;9(2), e87441.PubMedCentralCrossRefPubMed Chaturbhuj DN, Nirmalkar AP, Paranjape RS, Tripathy SP. Evaluation of a cost effective in-house method for HIV-1 drug resistance genotyping using plasma samples. PLoS One. 2014;9(2), e87441.PubMedCentralCrossRefPubMed
10.
Zurück zum Zitat Chaturbhuj DN, Deshmukh PS, Hingankar NK, Siddhaarth K, Deshpande SN, Sen S, et al. Evaluations of an in-house drug resistance method for HIV-1 drug resistance using ViroSeq 2.0 genotyping system as a gold standard. J Virol Methods. 2013;189(1):87–92.CrossRefPubMed Chaturbhuj DN, Deshmukh PS, Hingankar NK, Siddhaarth K, Deshpande SN, Sen S, et al. Evaluations of an in-house drug resistance method for HIV-1 drug resistance using ViroSeq 2.0 genotyping system as a gold standard. J Virol Methods. 2013;189(1):87–92.CrossRefPubMed
12.
Zurück zum Zitat Zazzi M, Prosperi M, Vicenti I, Di Giambenedetto S, Callegaro A, Bruzzone B, et al. Rules-based HIV-1 genotypic resistance interpretation systems predict 8 week and 24 week virological antiretroviral treatment outcome and benefit from drug potency weighting. J Antimicrob Chemother. 2009;64(3):616–24.CrossRefPubMed Zazzi M, Prosperi M, Vicenti I, Di Giambenedetto S, Callegaro A, Bruzzone B, et al. Rules-based HIV-1 genotypic resistance interpretation systems predict 8 week and 24 week virological antiretroviral treatment outcome and benefit from drug potency weighting. J Antimicrob Chemother. 2009;64(3):616–24.CrossRefPubMed
13.
Zurück zum Zitat Vingerhoets J, Tambuyzer L, Azijn H, Hoogstoel A, Nijs S, Peeters M, et al. Resistance profile of etravirine: combined analysis of baseline genotypic and phenotypic data from the randomized, controlled Phase III clinical studies. AIDS. 2010;24(4):503–14.CrossRefPubMed Vingerhoets J, Tambuyzer L, Azijn H, Hoogstoel A, Nijs S, Peeters M, et al. Resistance profile of etravirine: combined analysis of baseline genotypic and phenotypic data from the randomized, controlled Phase III clinical studies. AIDS. 2010;24(4):503–14.CrossRefPubMed
14.
Zurück zum Zitat Schoffelen AF, Wensing AM, Tempelman HA, Geelen SP, Hoepelman AI, Barth RE. Sustained virological response on second-line antiretroviral therapy following virological failure in HIV-infected patients in rural South Africa. PLoS One. 2013;8(3):e58526.PubMedCentralCrossRefPubMed Schoffelen AF, Wensing AM, Tempelman HA, Geelen SP, Hoepelman AI, Barth RE. Sustained virological response on second-line antiretroviral therapy following virological failure in HIV-infected patients in rural South Africa. PLoS One. 2013;8(3):e58526.PubMedCentralCrossRefPubMed
15.
Zurück zum Zitat Vallabhaneni S, Chandy S, Heylen E, Ekstrand ML. Evaluation of WHO immunologic criteria for treatment failure: implications for detection of virologic failure, evolution of drug resistance and choice of second-line therapy in India. J Int AIDS Soc. 2013;16:18449.PubMedCentralCrossRefPubMed Vallabhaneni S, Chandy S, Heylen E, Ekstrand ML. Evaluation of WHO immunologic criteria for treatment failure: implications for detection of virologic failure, evolution of drug resistance and choice of second-line therapy in India. J Int AIDS Soc. 2013;16:18449.PubMedCentralCrossRefPubMed
16.
Zurück zum Zitat Rawizza HE, Chaplin B, Meloni ST, Eisen G, Rao T, Sankale JL, et al. Immunologic criteria are poor predictors of virologic outcome: implications for HIV treatment monitoring in resource-limited settings. Clin Infect Dis. 2011;53(12):1283–90.PubMedCentralCrossRefPubMed Rawizza HE, Chaplin B, Meloni ST, Eisen G, Rao T, Sankale JL, et al. Immunologic criteria are poor predictors of virologic outcome: implications for HIV treatment monitoring in resource-limited settings. Clin Infect Dis. 2011;53(12):1283–90.PubMedCentralCrossRefPubMed
17.
Zurück zum Zitat Petersen ML, Tran L, Geng EH, Reynolds SJ, Kambugu A, Wood R, et al. Delayed switch of antiretroviral therapy after virologic failure associated with elevated mortality among HIV-infected adults in Africa. AIDS. 2014;28(14):2097–107.PubMedCentralCrossRefPubMed Petersen ML, Tran L, Geng EH, Reynolds SJ, Kambugu A, Wood R, et al. Delayed switch of antiretroviral therapy after virologic failure associated with elevated mortality among HIV-infected adults in Africa. AIDS. 2014;28(14):2097–107.PubMedCentralCrossRefPubMed
18.
Zurück zum Zitat Paton NI, Kityo C, Hoppe A, Reid A, Kambugu A, Lugemwa A, et al. Assessment of second-line antiretroviral regimens for HIV therapy in Africa. N Engl J Med. 2014;371(3):234–47.CrossRefPubMed Paton NI, Kityo C, Hoppe A, Reid A, Kambugu A, Lugemwa A, et al. Assessment of second-line antiretroviral regimens for HIV therapy in Africa. N Engl J Med. 2014;371(3):234–47.CrossRefPubMed
19.
Zurück zum Zitat Pujades-Rodriguez M, Balkan S, Arnould L, Brinkhof MA, Calmy A. Treatment failure and mortality factors in patients receiving second-line HIV therapy in resource-limited countries. JAMA. 2010;304(3):303–12.CrossRefPubMed Pujades-Rodriguez M, Balkan S, Arnould L, Brinkhof MA, Calmy A. Treatment failure and mortality factors in patients receiving second-line HIV therapy in resource-limited countries. JAMA. 2010;304(3):303–12.CrossRefPubMed
20.
Zurück zum Zitat Khan S, Das M, Andries A, Deshpande A, Mansoor H, Saranchuk P, et al. Second-line failure and first experience with third-line antiretroviral therapy in Mumbai. India Glob Health Action. 2014;7:24861.PubMed Khan S, Das M, Andries A, Deshpande A, Mansoor H, Saranchuk P, et al. Second-line failure and first experience with third-line antiretroviral therapy in Mumbai. India Glob Health Action. 2014;7:24861.PubMed
21.
Zurück zum Zitat Boettiger DC, Nguyen VK, Durier N, Bui HV, Heng Sim BL, Azwa I, et al. Efficacy of second-line antiretroviral therapy among people living with HIV/AIDS in Asia: results from the TREAT Asia HIV observational database. J Acquir Immune Defic Syndr. 2015;68(2):186–95.CrossRefPubMed Boettiger DC, Nguyen VK, Durier N, Bui HV, Heng Sim BL, Azwa I, et al. Efficacy of second-line antiretroviral therapy among people living with HIV/AIDS in Asia: results from the TREAT Asia HIV observational database. J Acquir Immune Defic Syndr. 2015;68(2):186–95.CrossRefPubMed
22.
Zurück zum Zitat El-Khatib Z, Ekstrom AM, Ledwaba J, Mohapi L, Laher F, Karstaedt A, et al. Viremia and drug resistance among HIV-1 patients on antiretroviral treatment: a cross-sectional study in Soweto, South Africa. AIDS. 2010;24(11):1679–87.PubMedCentralCrossRefPubMed El-Khatib Z, Ekstrom AM, Ledwaba J, Mohapi L, Laher F, Karstaedt A, et al. Viremia and drug resistance among HIV-1 patients on antiretroviral treatment: a cross-sectional study in Soweto, South Africa. AIDS. 2010;24(11):1679–87.PubMedCentralCrossRefPubMed
23.
Zurück zum Zitat van Zyl GU, van Mens TE, McIlleron H, Zeier M, Nachega JB, Decloedt E, et al. Low lopinavir plasma or hair concentrations explain second-line protease inhibitor failures in a resource-limited setting. J Acquir Immune Defic Syndr. 2011;56(4):333–9.PubMedCentralCrossRefPubMed van Zyl GU, van Mens TE, McIlleron H, Zeier M, Nachega JB, Decloedt E, et al. Low lopinavir plasma or hair concentrations explain second-line protease inhibitor failures in a resource-limited setting. J Acquir Immune Defic Syndr. 2011;56(4):333–9.PubMedCentralCrossRefPubMed
24.
Zurück zum Zitat Rawizza HE, Chaplin B, Meloni ST, Darin KM, Olaitan O, Scarsi KK, et al. Accumulation of protease mutations among patients failing second-line antiretroviral therapy and response to salvage therapy in Nigeria. PLoS One. 2013;8(9):e73582.PubMedCentralCrossRefPubMed Rawizza HE, Chaplin B, Meloni ST, Darin KM, Olaitan O, Scarsi KK, et al. Accumulation of protease mutations among patients failing second-line antiretroviral therapy and response to salvage therapy in Nigeria. PLoS One. 2013;8(9):e73582.PubMedCentralCrossRefPubMed
25.
Zurück zum Zitat Saravanan S, Vidya M, Balakrishnan P, Kantor R, Solomon SS, Katzenstein D, et al. Viremia and HIV-1 drug resistance mutations among patients receiving second-line highly active antiretroviral therapy in Chennai, Southern India. Clin Infect Dis. 2012;54(7):995–1000.PubMedCentralCrossRefPubMed Saravanan S, Vidya M, Balakrishnan P, Kantor R, Solomon SS, Katzenstein D, et al. Viremia and HIV-1 drug resistance mutations among patients receiving second-line highly active antiretroviral therapy in Chennai, Southern India. Clin Infect Dis. 2012;54(7):995–1000.PubMedCentralCrossRefPubMed
26.
Zurück zum Zitat Nijhuis M, van Maarseveen NM, Lastere S, Schipper P, Coakley E, Glass B, et al. A novel substrate-based HIV-1 protease inhibitor drug resistance mechanism. PLoS Med. 2007;4(1):e36.PubMedCentralCrossRefPubMed Nijhuis M, van Maarseveen NM, Lastere S, Schipper P, Coakley E, Glass B, et al. A novel substrate-based HIV-1 protease inhibitor drug resistance mechanism. PLoS Med. 2007;4(1):e36.PubMedCentralCrossRefPubMed
27.
Zurück zum Zitat Gupta RK, Kohli A, McCormick AL, Towers GJ, Pillay D, Parry CM. Full-length HIV-1 Gag determines protease inhibitor susceptibility within in vitro assays. AIDS. 2010;24(11):1651–5.PubMedCentralCrossRefPubMed Gupta RK, Kohli A, McCormick AL, Towers GJ, Pillay D, Parry CM. Full-length HIV-1 Gag determines protease inhibitor susceptibility within in vitro assays. AIDS. 2010;24(11):1651–5.PubMedCentralCrossRefPubMed
28.
29.
Zurück zum Zitat Gupta A, Saple DG, Nadkarni G, Shah B, Vaidya S, Hingankar N, et al. One-, two-, and three-class resistance among HIV-infected patients on antiretroviral therapy in private care clinics: Mumbai, India. AIDS Res Hum Retroviruses. 2010;26(1):25–31.PubMedCentralCrossRefPubMed Gupta A, Saple DG, Nadkarni G, Shah B, Vaidya S, Hingankar N, et al. One-, two-, and three-class resistance among HIV-infected patients on antiretroviral therapy in private care clinics: Mumbai, India. AIDS Res Hum Retroviruses. 2010;26(1):25–31.PubMedCentralCrossRefPubMed
30.
Zurück zum Zitat Nwobegahay JM, Bessong PO, Masebe TM, Mavhandu LG, Iweriebor BC, Selabe G. Prevalence of antiretroviral drug resistance mutations and HIV-I subtypes among newly-diagnosed drug-naive persons visiting a voluntary testing and counselling centre in northeastern South Africa. J Health Popul Nutr. 2011;29(4):303–9.PubMedCentralCrossRefPubMed Nwobegahay JM, Bessong PO, Masebe TM, Mavhandu LG, Iweriebor BC, Selabe G. Prevalence of antiretroviral drug resistance mutations and HIV-I subtypes among newly-diagnosed drug-naive persons visiting a voluntary testing and counselling centre in northeastern South Africa. J Health Popul Nutr. 2011;29(4):303–9.PubMedCentralCrossRefPubMed
31.
Zurück zum Zitat Balakrishnan P, Kumarasamy N, Kantor R, Solomon S, Vidya S, Mayer KH, et al. HIV type 1 genotypic variation in an antiretroviral treatment-naive population in southern India. AIDS Res Hum Retroviruses. 2005;21(4):301–5.CrossRefPubMed Balakrishnan P, Kumarasamy N, Kantor R, Solomon S, Vidya S, Mayer KH, et al. HIV type 1 genotypic variation in an antiretroviral treatment-naive population in southern India. AIDS Res Hum Retroviruses. 2005;21(4):301–5.CrossRefPubMed
Metadaten
Titel
Outcome of patients on second line antiretroviral therapy under programmatic condition in India
verfasst von
Jaya Chakravarty
Shyam Sundar
Ankita Chourasia
Pallav Narayan Singh
Swarali Kurle
Srikanth P. Tripathy
Devidas N Chaturbhuj
Madhukar Rai
Amit Kumar Agarwal
Rabindra Nath Mishra
Ramesh S. Paranjape
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
BMC Infectious Diseases / Ausgabe 1/2015
Elektronische ISSN: 1471-2334
DOI
https://doi.org/10.1186/s12879-015-1270-8

Weitere Artikel der Ausgabe 1/2015

BMC Infectious Diseases 1/2015 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Echinokokkose medikamentös behandeln oder operieren?

06.05.2024 DCK 2024 Kongressbericht

Die Therapie von Echinokokkosen sollte immer in spezialisierten Zentren erfolgen. Eine symptomlose Echinokokkose kann – egal ob von Hunde- oder Fuchsbandwurm ausgelöst – konservativ erfolgen. Wenn eine Op. nötig ist, kann es sinnvoll sein, vorher Zysten zu leeren und zu desinfizieren. 

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

Proximale Humerusfraktur: Auch 100-Jährige operieren?

01.05.2024 DCK 2024 Kongressbericht

Mit dem demographischen Wandel versorgt auch die Chirurgie immer mehr betagte Menschen. Von Entwicklungen wie Fast-Track können auch ältere Menschen profitieren und bei proximaler Humerusfraktur können selbst manche 100-Jährige noch sicher operiert werden.

Die „Zehn Gebote“ des Endokarditis-Managements

30.04.2024 Endokarditis Leitlinie kompakt

Worauf kommt es beim Management von Personen mit infektiöser Endokarditis an? Eine Kardiologin und ein Kardiologe fassen die zehn wichtigsten Punkte der neuen ESC-Leitlinie zusammen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.