Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2010 Jan 2.
Published in final edited form as: AIDS. 2009 Jan 2;23(1):71–82. doi: 10.1097/QAD.0b013e32831cc129

Interruption of antiretroviral therapy is associated with increased plasma cystatin C

A Mocroft 1, C Wyatt 2, L Szczech 3, J Neuhaus 4, W El-Sadr 5, R Tracy 6, L Kuller 7, M Shlipak 8, B Angus 9,10, H Klinker 11, M Ross 2, for the INSIGHT SMART study group
PMCID: PMC2761385  NIHMSID: NIHMS130496  PMID: 19050388

Abstract

Background

Cystatin C has been proposed as an alternative marker of renal function. We sought to determine if participants randomized to episodic use of antiretroviral therapy guided by CD4+ count (drug conservation; DC) had altered cystatin C levels compared to those randomised to continuous antiretroviral therapy (viral suppression; VS) in the Strategies for Management of Antiretroviral Therapy Trial, and to identify factors associated with increased cystatin C.

Methods

Cystatin C was measured in plasma collected at randomization, 1, 2, 4, 8 and 12 months after randomization in a random sample of 249 and 250 participants in the DC and VS groups respectively. Logistic regression was used to model the odds of ≥ 0.15 mg/dl increase in cystatin C (1 standard deviation [SD]) in the first month after randomisation, adjusting for demographic and clinical characteristics.

Results

At randomisation, mean (SD) cystatin C level was 0.99 (0.26 mg/dl) and 1.01 (0.28 mg/dl) in the DC and VS arms respectively (p=0.29). In the first month after randomisation, 21.8% and 10.6% had ≥0.15 mg/dl increase in cystatin C in the DC and VS arm respectively (p=0.0008). The difference in cystatin C between the treatment arms was maintained through 1 year after randomisation. After adjustment, participants in the VS arm had significantly reduced odds of ≥0.15 mg/dl increase in cystatin C in the first month (OR 0.42; 95% CI 0.23–0.74, p=0.0023).

Conclusions

These results demonstrate that interruption of antiretroviral therapy is associated with an increase in cystatin C, which may reflect worsened renal function.

Introduction

The introduction of effective antiretroviral therapy (ART) for the treatment of HIV-1 infection has led to a dramatic decline in mortality and morbidity1, 2. As patients survive longer, renal disease has become an important contributing factor to morbidity and mortality3. While ART has been demonstrated to reduce the incidence and severity of HIV-associated nephropathy (HIVAN) 4-6, specific antiretrovirals may be associated with nephrotoxicity, or with increased rates of hypertension and diabetes, which in turn may increase the risk of kidney disease7, 8. Several studies have considered the impact of ART on serum creatinine, proteinuria, estimated glomerular filtration rate (GFR), and progression of kidney disease9-12. Cystatin C, a non-glycosylated basic protein secreted by all nucleated cells and freely filtered by the glomerulus, has been considered as a prognostic marker for mortality and morbidity13 and also proposed as an alternative marker of GFR14. Serum cystatin C levels correlate well with GFR in patients without HIV 15-17, and may be more sensitive than serum creatinine in the detection of early renal insufficiency18, 19. Serum cystatin C levels are not affected by muscle mass, which may be decreased in patients with advanced HIV disease20. Further, serum cystatin C levels have been correlated with renal function in HIV-infected patients and have been noted to be higher in HIV-1 infected patients compared to uninfected individuals21, 22. To our knowledge changes in cystatin C associated with starting or stopping ART have not been previously described.

The Strategies for Management of Antiretroviral Therapy (SMART) trial was a randomized controlled clinical trial which compared the episodic use of ART (drug conservation, or DC arm) guided by CD4+ cell counts with continuous therapy aimed at virologic suppression (VS arm)23. The study demonstrated that the DC strategy was associated with inferior outcomes with regards to development of opportunistic disease or death and the composite endpoint of cardiovascular, hepatic and renal disease23; an increased risk of fatal or non-fatal renal disease was described in the DC arm compared to the VS arm. In another study from SMART24, plasma biomarkers of inflammation (IL-6, high sensitivity C-reactive protein [hsCRP], amyloid A, amyloid P), coagulation (D-dimer, prothrombin fragments 1+2), and lipids (total cholesterol, HDL, LDL, triglycerides) were measured to determine whether they were associated with all cause mortality and cardiovascular disease.

The aims of the current study were to describe changes in plasma cystatin C in SMART participants included in the previous biomarker substudy, and to investigate the factors associated with increase in cystatin C levels after randomisation. Since many of the previously measured biomarkers are also associated with renal disease, we sought to determine whether these biomarkers were associated with cystatin C.

SMART Study Participants and Treatment Protocol

Between January 2002 and January 2006, 5,472 HIV-infected participants were enrolled in SMART by 318 sites in 33 countries. Participants were eligible if they had a CD4+ count >350 cells/mm3 and were willing to initiate, modify or stop ART as per study guidelines23. SMART participants were randomized to one of two ART strategies. For the VS group, ART was used in an uninterrupted manner with the goal of maximal suppression of HIV replication. The experimental drug conservation (DC) strategy entailed episodic use of ART for periods defined by CD4+ count thresholds. ART was stopped (or deferred) until the CD4+ count dropped to <250 cells/mm3, at which time ART was to be (re-)initiated and continued until the CD4+ count rose to >350 cells/mm3. Upon confirmation that the CD4+ count was >350 cells/mm3, ART was to be stopped and resumed again when the CD4+ count was <250 cells/mm3. During periods of ART use, the goal was to achieve maximal viral suppression. As previously reported, on January 11, 2006, investigators and participants were notified of a safety risk in the DC group, enrollment was stopped, and participants in the DC group were advised to restart ART23.

Specimens Collected for Biomarker Substudy

In SMART, follow-up study visits occurred at month 1, month 2, then every 2 months for the first year, and every 4 months thereafter. 5151 participants (94%) had blood stored for future use. Plasma specimens were collected using EDTA (lavender top) tubes, aliquoted, and shipped frozen to a central repository. Only specimens obtained prior to January 11, 2006 were used for the biomarker analyses described in this paper. Plasma specimens collected at randomisation and one month after randomisation were identified for participants who consented to specimen storage for future research. A random sample of 250 participants without a pre-existing history of cardiovascular disease from each treatment group was chosen from 1286 participants with available samples. One participant in the DC group who did not have an adequate specimen at randomisation was subsequently excluded.

Measurement of Cystatin-C and inflammatory markers

Cystatin-C was measured on plasma samples that were stored at −70°C. A BNII nephelometer (Dade Behring Inc., Deerfield, Ill) that utilized a particle-enhanced immunonephelometric assay (N Latex Cystatin-C) was used. The assay range is 0.195 to 7.330 mg/dl. The reference range for young, healthy individuals is 0.53 to 0.92 mg/dl. The inflammatory markers were measured by the Laboratory for Clinical Biochemistry Research at the University of Vermont.

Estimated glomerular filtration rate (eGFR) was calculated using the Modification of Diet in Renal Disease (MDRD) formula25 and the Cockcroft-Gault formula26

Statistical methods

Descriptive statistics were used to demonstrate the change in cystatin C in the first year after randomisation in the DC versus VS arm. Pearson correlation coefficients were used to determine the correlation between (a) values of cystatin C and markers of inflammation, coagulation and lipids at randomisation, (b) marker values at randomisation and change in cystatin C between randomisation and month 1, (c) marker values and cystatin C at month 1 and (d) changes in marker and cystatin C values between randomisation to month 1. Logistic regression, using forward selection with p>0.2 inclusion criteria, was used to model the odds of ≥1 SD increase in cystatin C, 0.15 mg/dl, in the first month after randomisation. Variables included in multivariate models were gender, age, race, mode of HIV infection, hepatitis B and C status, prior AIDS, HIV-RNA ≤ 400 copies/ml at randomisation, current and prior smoking status, diabetes, treatment with blood pressure or lipid lowering therapy, previous antiretroviral experience, ART status at enrolment, and exposure to protease inhibitors (PIs), non-nucleoside reverse transcriptase inhibitors (NNRTIs), or nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs). Randomisation values for age, weight, body mass index, CD4+ count, HIV-RNA levels, total cholesterol, HDL, LDL, and triglycerides, as well as nadir CD4+ count were also included.

All analyses were performed using SAS version 9.1 (Statistical analysis Software, Cary, NC USA). All tests of significance were 2-sided.

Results

Characteristics at randomisation of participants assigned to the DC and VS arm in this substudy of SMART were well balanced with no significant differences (p>0.1 for all comparisons) (Table 1). At randomisation, 73.1% and 75.2% of DC and VS participants, respectively, were on ART. At randomisation, mean (SD) cystatin C level was 0.99 (0.26 mg/dl) and 1.01 (0.28 mg/dl) in the DC and VS arms, respectively (p=0.29). 90 participants in the DC arm (36.3%) had a cystatin C level > 1.0 mg/dl, compared to 101 participants in the VS arm (41.2%, p=0.26, chi-squared test).

Table 1.

Characteristics at randomisation in the subset of participants in SMART with measured cystatin-C

Characteristics of participants from SMART with measured cystatin-C: Comparison of DC and VS arms; all factors measured at randomisation

Drug conservation
N=249
Virologic suppression
N=250
N % N %
Gender Female 72 29.9 74 29.6

Race Black 131 52.6 121 48.4
White 86 34.5 93 37.2
Other 32 12.9 26 14.4

Mode of Same sex 110 44.2 113 45.2
Infection1 Opposite sex 132 53.0 132 52.8
IDU 29 11.7 25 10.0
Blood 13 5.2 13 5.2
Other 15 6.0 17 6.8

Hepatitis B positive 11 4.4 8 3.2
C positive 50 20.1 44 17.6

Prior AIDS 71 28.5 68 27.2

HIV RNA ≤400 142 57.0 136 54.4

Cardiovascular Smoker 103 41.1 111 44.4
Disease risk Diabetes 22 8.8 24 9.6
Factors Prior cardiovascular disease 0 0 0 0
Blood pressure treatment 60 24.1 76 30.4
Lipid lowering treatment 41 16.5 45 18.0

Antiretroviral Naïve 16 6.4 15 6.0
History Protease-inhibitor exposed 170 68.3 158 62.8
Non-nucleoside exposed 158 63.5 159 63.2

ART at On antiretrovirals 182 73.1 188 75.2
Randomisation2 Protease inhibitor 77 42.3 89 47.3
Non-nucleoside 99 54.4 90 47.9
Protease inhibitor+non-nucleoside 11 6.0 13 6.9
Drug conservation Virologic suppression
Median IQR Median IQR

Age (years) 44 38-52 44 38-50
CD4 (cells/mm3) 532 432-720 550 430-743
CD4 nadir (cells/mm3) 250 133-350 267 167-386
HIV RNA viral load Log10 copies/ml 2.60 1.70-4.00 2.60 1.88-3.75
Total cholesterol (mg/dl) 181 156-208 183 157-213
HDL cholesterol (mg/dl) 40 32-47 41 31-50
LDL cholesterol (mg/dl) 103 85-126 107 86-131
Triglycerides (mg/dl) 153 93-246 157 106-258
Cystatin-C (mg/dl) 0.94 0.83-1.09 0.97 0.84-1.11
1

Patients can be classified as belonging to more than one risk group

2

% of those on antiretrovirals at randomisation

The difference in cystatin C levels between the DC and VS arms, together with the proportion of participants having a ≥0.15 mg/dl rise in cystatin C in the DC and VS arms is shown in Figure 1. Six participants with cystatin C levels > 5 mg/dl at randomisation were excluded from this analysis (1 DC, 5 VS); inclusion of these participants did not alter our findings (data not shown). In the first month after randomisation, there was a small rise in cystatin C (mean 0.05, SD 0.18 mg/dl) in the DC arm and a small decrease in cystatin C in the VS arm (mean −0.02, SD 0.17 mg/dl; p<0.0001 for difference). The proportion of participants who experienced a ≥0.15 mg/dl increase in cystatin C was significantly higher in the DC arm (21.8%) compared to the VS arm (10.6%; p=0.0008, chi-squared test). Cystatin C remained higher in the DC arm than the VS arm from month 1 through month 12 (Figure 1). Highly consistent results were seen censoring the analysis at the date of restarting ART in the DC arm (data not shown). There were no significant differences between the treatment arms in the proportion of participants with a cystatin-C level of > 1.0 mg/dl at months 1, 4, 8 and 12 (p>0.1, chi-squared test).

Figure 1.

Figure 1

Cystatin C after randomisation in SMART

Other markers of kidney function were measured in a subset of participants at randomisation, month 4 and month 12 of follow-up. There was no significant difference in serum creatinine or MDRD eGFR between participants in the DC and VS arms at any time point (Table 2), or when comparing participants from different races within the DC and VS arm (data not shown). In addition, there was no change in weight or albumin from randomisation to month 4 or month 12, or from month 4 to month 12 in either treatment arm (p>0.25, all comparisons). Among all participants, there was a significant correlation between cystatin C and MDRD eGFR at months 0, 4 and 12 (correlation coefficients −0.41, −0.22, −0.35, p<0.0001, p=0.092, p<0.0001 respectively) and between cystatin C and serum creatinine at months 0, 4 and 12 (correlation coefficients 0.234, 0.308, 0.665, p<0.0001 for all). Consistent results were seen between the DC and VS arms when GFR was estimated using the CG equation (data not shown).

Table 2.

Markers of renal function

DC VS p-value1
Month N with data Mean ± SD N with data Mean ± SD
Estimated GFR2 0 141 97.6 ± 23.2 135 93.4 ± 24.0 0.15
4 107 98.7 ± 20.8 108 95.3 ± 23.2 0.92
12 82 96.4 ± 21.5 91 94.8 ± 25.4 0.90

Creatinine 0 141 0.96 ± 0.24 135 1.05 ± 0.76 0.19
4 107 0.93 ± 0.19 108 0.97 ± 0.23 0.63
12 82 0.95 ± 0.20 91 1.00 ± 0.35 0.59

Albumin 0 133 4.14 ± 0.43 118 4.17 ± 0.41 0.52
4 107 4.01 ± 0.44 94 4.01 ± 0.62 0.17
12 73 4.09 ± 0.46 81 4.10 ± 0.49 0.33

Cystatin C 0 248 0.99 ± 0.26 245 1.01 ± 0.28 0.31
4 205 0.86 ± 0.23 202 0.84 ± 0.31 0.026
12 150 0.91 ± 0.32 133 0.91 ± 0.44 0.081
1

Adjusted for values at randomisation

2

GFR estimated by MDRD equation. Results were similar when GFR was estimated by Cockcroft-Gault.

Figure 2 shows the unadjusted odds ratio of > 1 SD increase in cystatin C comparing the VS arm to the DC arm within subgroups of patients. For example, in patients with a CD4 count of ≤500/mm3 at randomisation, 5.2% of participants in the VS arm had ≥ 0.15mg/dl increase in cystatin C compared to 22.2% in the DC arm (odds ratio [OR] 0.19; 95% confidence interval [CI] 0.07 – 0.53, p=0.0015). The corresponding proportions in participants with a CD4 > 500cells/mm3 at randomisation were 14.1% and 21.5% (OR 0.60, 95% CI 0.33 – 1.10, p=0.098, p-value for interaction=0.059). The odds ratio of > 1 SD increase in cystatin C comparing the VS arm to the DC arms was similar in patients who were on or off antiretrovirals at baseline and in patients with undetectable plasma HIV-RNA (<400 copies/ml) compared to those with plasma HIV-RNA ≥ 400 copies/ml.

Figure 2.

Figure 2

Odds ratio of >1 SD (0.15 mg/dl) increase in cystatin C by 1 month after randomisation to SMART

In a multivariate model adjusted for treatment arm, HIV exposure route, exposure to blood-pressure lowering medication and previous treatment to protease inhibitors, participants in the VS arm had less than half the odds of ≥0.15 mg/dl increase in cystatin C in the first month after randomisation to SMART compared to participants in the DC arm (adjusted OR 0.42; 95% CI 0.23 – 0.74, p=0.0023). There was a significantly higher odds of increased cystatin C in participants treated at randomisation with blood pressure lowering medications (adjusted OR 1.91; 95% CI 1.06 – 3.44, p=0.038) and those who had been treated previously with protease inhibitors (adjusted OR 2.17; 95%CI 1.07 – 4.40, p=0.030). Participants infected with HIV through intravenous drug use had marginally significantly increased odds of an increase in cystatin C (adjusted OR 2.07; 95% CI 0.91 – 4.71, p=0.077). There was no significant association between race and increase in cystatin C in univariate (p=0.57) or multivariate analyses (p=0.43). In addition, there was no relationship between exposure to specific NRTIs and change in cystatin C, although the power for this analysis was limited. These analyses were repeated modelling the change in cystatin C rather than the proportion with a ≥0.15 mg/dl increase with highly consistent results (data not shown). Multivariate analyses were also performed separately for the DC and VS arms. There was no qualitative difference in the association of variables with change in cystatin C at month 1 between the treatment groups (data not shown). In the subset of patients with cystatin C levels measured at 12 months after randomisation (n=283; 150 DC versus 133 VS)), participants in the VS arm had lower odds of a ≥0.15 mg/dl increase in cystatin C at 12 months after adjustment for the same factors as shown in table 3 (OR 0.38, 95% CI 0.13 – 1.10, p=0.075), although this was of marginal significance.

Table 3.

Pearson correlation coefficients for (a) marker values and cystatin C at randomisation; (b) marker values at randomisation and change in cystatin C between randomisation and month 1; (c) marker values at 1 month after randomisation and change in cystatin C between randomisation and month 1 and (d) change in marker values between randomisation and month 1 and change in cystatin C between randomisation and month 1

Marker (a) marker values and
cystatin C at randomisation
(b) marker values at
randomisation and change in
cystatin C (randomisation to
month 1)
(c) marker values 1 month
after randomisation and
change in cystatin C
(randomisation to month 1)
(d) change in marker
values (randomisation to
month 1) and change in
cystatin C (randomisation to
month 1)
N Correlation p N Correlation P N Correlation p N Correlation p-value
CD4 (cells/mm3) 493 −0.035 0.43 493 0.090 0.045 488 −0.060 0.18 488 −0.125 0.0058
Log HIV-RNA (copies/ml) 493 0.116 0.0099 493 −0.128 0.0044 487 0.143 0.0015 487 0.277 <0.0001
HDL (mg/dl) 492 −0.220 <0.0001 492 0.029 0.52 491 −0.240 <0.0001 490 −0.196 <0.0001
LDL (mg/dl) 493 −0.040 0.37 493 −0.032 0.47 490 0.014 0.76 490 −0.016 0.72
Total Cholesterol (mg/dl) 493 0.029 0.51 493 0.029 0.51 492 0.063 0.16 492 −0.093 0.040
Log triglycerides (mg/dl) 493 0.181 <0.0001 493 0.057 0.21 492 0.213 <0.0001 492 −0.081 0.073
Log D-dimer (μg/mL) 490 0.124 0.0061 490 −0.105 0.020 493 0.168 0.0002 490 0.254 <0.0001
Log prothrombin fragment 1+2 (pmol/L) 483 −0.014 0.76 483 −0.064 0.16 489 −0.005 0.91 479 0.079 0.083
Log Amyloid A (mg/L) 493 0.046 0.31 493 −0.005 0.92 493 0.072 0.11 493 0.148 0.0010
Log hsCRP (μg/mL) 493 0.085 0.089 493 −0.034 0.45 493 0.133 0.0030 493 0.192 <0.0001
Log Amyloid P (μg/mL) 491 0.017 0.70 491 −0.029 0.52 492 0.075 0.099 490 0.064 0.16
Log IL-6 492 0.255 <0.0001 492 −0.033 0.46 491 0.285 <0.0001 490 0.121 0.0072

The lowest and highest quintiles of cystatin C at randomization were 0.81 and 1.14 respectively; the correlation between cystatin C and serum creatinine at randomization in the lowest quintile was 0.27 (n=57 observations, p=0.045) and in the highest quintile was 0.71 (n=57 observations, p<0.0001). Additional correlations between cystatin C, inflammatory, coagulation and lipid biomarkers are shown in Table 3. The increase in cystatin C at 1 month after randomisation was correlated with an increase in hsCRP, IL-6, amyloid A, and D-dimer, and a decrease in HDL and total cholesterol between randomisation and month 1. Biomarker values at randomization were added to the logistic regression model described above. After adjustment, none of the biomarkers were independently associated with a ≥0.15 mg/dl increase in cystatin C at 1 month after randomisation. Further analyses detected no correlation between randomisation values of cystatin C and HIV-RNA in the DC arm (Table 3, column (a); correlation coefficient 0.037, p=0.57), but there was a weak correlation in the VS arm (correlation coefficient 0.195, p=0.0021). The correlation coefficients were similar in the DC and VS arms when considering the correlation between HIV-RNA at randomisation and change in cystatin C over the first month (Table 3, column (b)), and change in HIV-RNA versus change in cystatin C over the first month (Table 3, column (d)). In addition, there was a weak correlation between HIV-RNA at 1 month after randomisation and change in cystatin C in the DC arm (correlation coefficient −0.141, p=0.027), which was not apparent in the VS arm (correlation coefficient 0.061, p=0.034).

Discussion

The increased risk of kidney disease demonstrated in the DC arm of SMART23 motivated the current study of changes in plasma cystatin C, a proposed early marker of subclinical kidney damage. In the subset of SMART participants evaluated in this study, those who discontinued or deferred ART (DC arm) had a rapid small, but significant, increase in plasma cystatin C compared to participants who were treated with continuous ART (VS arm), and were significantly more likely to have cystatin C levels higher than the level at randomisation. The difference in cystatin C levels between the DC and VS arms of SMART were maintained through 1 year of follow-up.

Plasma cystatin C levels are determined by the net balance of secretion of cystatin C into plasma and clearance by glomerular filtration27. The increased cystatin C levels found in the DC arm compared to the VS arm must therefore reflect either a reduction in GFR in the DC group that was not detected by changes in serum creatinine or eGFR, and/or increased secretion of cystatin C from cells into the plasma. The latter explanation seems unlikely as research has shown that cystatin C levels do not increase in the setting of acute stress28. Systemic levels of cystatin C have been shown to be independent of several characteristics, including age29, gender29, 30 and muscle mass31. There are conflicting data in the literature regarding whether systemic inflammation can influence cystatin C levels independent of renal function. Knight et al. reported that cystatin C levels were independently associated with C reactive protein (CRP) levels in a cohort of healthy subjects, nearly all of whom were Caucasians with normal renal function32. However, another study by Singh et al., which included a more racially diverse population with a higher prevalence of chronic kidney disease, found that the association of cystatin C with inflammatory markers was not independent of GFR33. The latter study suggested that impaired renal function is associated with inflammation and that levels of cystatin C are reflective of decreased GFR even in the presence of systemic inflammation. In this study, there was a strong correlation between randomisation levels of cystatin C and serum creatinine in the upper quintile of cystatin C at randomisation. One explanation for this observation is that at higher levels, cystatin C and creatinine are well correlated and reflect impaired renal function, whereas at lower levels, cystatin C and/or creatinine are more prone to the influence of other factors such as inflammation and muscle mass, respectively. Although we identified weak correlations between levels of inflammatory markers and cystatin C, there was no independent association between hsCRP or IL-6 and change in cystatin C after adjustment for other variables. We therefore hypothesise that the increase in cystatin C observed in the DC arm of SMART upon interruption of ART reflected systemic inflammation that was concurrent with and may have contributed to renal injury.

We also found that an increase in cystatin C from randomisation to month 1 correlated with the value of D-dimers and HDL levels at 1 month after randomisation and with the change in these markers from randomisation to month 1. Increased D-dimers and decreased HDL are associated with increased risk of cardiovascular disease in the general population and can be induced by systemic inflammation34. D-dimers and HDL are also more likely to be elevated and decreased respectively in participants with chronic kidney disease, possibly contributing to the increased risk of cardiovascular mortality in that population35, 36.

There are several mechanisms by which interruption or deferral of ART may have resulted in an increase in cystatin C. While it is possible that the increase in cystatin C could reflect systemic inflammation rather than a decrease in renal function, we hypothesize that the inflammation was concurrent with and may have contributed to renal injury. Renal glomerular and tubular epithelial cells are susceptible to infection by HIV, and infected cells have been detected in the kidney, even in persons receiving ART with undetectable plasma HIV-RNA levels 37,38. HIV infection of human renal tubular epithelial cells induces apoptosis and production of inflammatory mediators39-42. Studies have demonstrated that ART can induce marked improvement in functional and histological abnormalities in some patients with HIV-associated nephropathy after a reduction in viral load38, 43, 44. Participants in the DC arm of SMART who were previously taking ART had a marked increase in plasma HIV-RNA levels after discontinuation or deferral of ART following randomisation. Moreover, participants in the DC arm were more likely to have an increase in cystatin C if they had an HIV-RNA level ≤400 copies/ml at randomisation before ART discontinuation. It is therefore plausible that withdrawal of ART resulted in increased HIV replication in renal epithelial cells, resulting in local inflammation and renal injury.

Our study has several limitations. Although there was a statistically significant difference in cystatin C levels between the DC and VS arms at month 1 after randomisation, the magnitude of the difference was small, and the clinical significance and relationship of this difference with renal disease are unclear. Since serum creatinine measurements were only available on a subset of participants, we cannot exclude the possibility that the lack of difference in creatinine and creatinine-based estimates of GFR between DC and VS arms was a result of ascertainment bias. Finally, since we did not use a “gold standard” to measure GFR (such as iothalamate or inulin clearance), we cannot be certain that the rise in cystatin C reflects a decline in GFR. However, cystatin C has been shown to be a better marker of GFR than serum creatinine in several disease settings, and creatinine itself has never been validated as a marker of GFR in HIV-infected patients using a “gold standard”. Prospective, well-designed studies are therefore urgently needed to explore the accuracy of cystatin C and creatinine as markers of GFR in patients with HIV infection.

In summary, in this randomised comparison, an increase in cystatin C levels was demonstrated after stopping ART in the DC arm of SMART. Cystatin C levels in the DC arm were consistently higher than the levels seen in the VS arm, and these increases were associated with increased production of inflammatory and coagulation-related biomarkers. These data demonstrate that discontinuation of ART was associated with an increase in cystatin C, which may be attributable to activation of inflammatory mediators. The reasons for this increase and the association with other biomarkers of inflammation and kidney function and with clinical renal disease require further investigation.

Acknowledgements

We would like to acknowledge the SMART participants, the SMART study team (see N Engl J Med, 2006:355:2294-2295 for list of investigators), and the INSIGHT Executive Committee. We would also like to thank Jim Neaton for his input, overview of the SMART study and contribution to this project.

Funding

Support provided by: NIAID, NIH grants U01AI042170, U01A1068641 and U01AI46362.

Clinical Trials.gov identifier: NCT00027352.

Appendix

Strategies for Management of Antiretroviral Therapy (SMART) Study Group

Investigators in the SMART Study Group

SMART was initiated by the Terry Beirn Community Programs for Clinical Research on AIDS (CPCRA) and implemented in collaboration with international coordinating centers in Copenhagen (Copenhagen HIV Programme), London (Medical Research Council, Clinical Trials Unit), Sydney (National Centre in HIV Epidemiology and Clinical Research) and Washington (CPCRA). Participating staff are listed below.

Copenhagen International Coordinating Center: JD Lundgren, KB Jensen, DC Gey, L Borup, M Pearson, PO Jansson, BG Jensen, J Tverland, H Juncker-Benzon, Z Fox, AN Phillips.

London International Coordinating Center: JH Darbyshire, AG Babiker, AJ Palfreeman, SL Fleck, W Dodds, E King, B Cordwell, F van Hooff, Y Collaco-Moraes.

Sydney International Coordinating Center: DA Cooper, S Emery, FM Drummond, SA Connor, CS Satchell, S Gunn, S Oka, MA Delfino, K Merlin, C McGinley.

Washington International Coordinating Center: F Gordin, E Finley, D Dietz, C Chesson, M Vjecha, B Standridge.

INSIGHT Network Coordinating Center: JD Neaton, G Bartsch, A DuChene, M George, B Grund, M Harrison, E Krum, G Larson, C Miller, R Nelson, J Neuhaus, MP Roediger, T Schultz.

ECG Reading Center: R Prineas, C Campbell, Z-M Zhang.

Endpoint Review Committee: G Perez (co-chair), A Lifson (co-chair), D Duprez, J Hoy, C Lahart, D Perlman, R Price, R Prineas, F Rhame, J Sampson, J Worley.

NIAID Data and Safety Monitoring Board: M Rein (chair), R DerSimonian (executive secretary), BA Brody, ES Daar, NN Dubler, TR Fleming, DJ Freeman, JP Kahn, KM Kim, G Medoff, JF Modlin, R Moellering Jr, BE Murray, B Pick, ML Robb, DO Scharfstein, J Sugarman, A Tsiatis, C Tuazon, L Zoloth.

NIH, NIAID: K Klingman, S Lehrman.

SMART Clinical Site Investigators by Country (SMART enrollment)

Argentina (147): J Lazovski, WH Belloso, MH Losso, JA Benetucci, S Aquilia, V Bittar, EP Bogdanowicz, PE Cahn, AD Casir, I Cassetti, JM Contarelli, JA Corral, A Crinejo, L Daciuk, DO David, G Guaragna, MT Ishida, A Krolewiecki, HE Laplume, MB Lasala, L Lourtau, SH Lupo, A Maranzana, F Masciottra, M Michaan, L Ruggieri, E Salazar, M Sánchez, C Somenzini.

Australia (170): JF Hoy, GD Rogers, AM Allworth, JStC Anderson, J Armishaw, K Barnes, A Carr, A Chiam, JCP Chuah, MC Curry, RL Dever, WA Donohue, NC Doong, DE Dwyer, J Dyer, B Eu, VW Ferguson, MAH French, RJ Garsia, J Gold, JH Hudson, S Jeganathan, P Konecny, J Leung, CL McCormack, M McMurchie, N Medland, RJ Moore, MB Moussa, D Orth, M Piper, T Read, JJ Roney, N Roth, DR Shaw, J Silvers, DJ Smith, AC Street, RJ Vale, NA Wendt, H Wood, DW Youds, J Zillman.

Austria (16): A Rieger, V Tozeau, A Aichelburg, N Vetter.

Belgium (95): N Clumeck, S Dewit, A de Roo, K Kabeya, P Leonard, L Lynen, M Moutschen, E O'Doherty.

Brazil (292): LC Pereira Jr, TNL Souza, M Schechter, R Zajdenverg, MMTB Almeida, F Araujo, F Bahia, C Brites, MM Caseiro, J Casseb, A Etzel, GG Falco, ECJ Filho, SR Flint, CR Gonzales, JVR Madruga, LN Passos, T Reuter, LC Sidi, ALC Toscano.

Canada (102): D Zarowny, E Cherban, J Cohen, B Conway, C Dufour, M Ellis, A Foster, D Haase, H Haldane, M Houde, C Kato, M Klein, B Lessard, A Martel, C Martel, N McFarland, E Paradis, A Piche, R Sandre, W Schlech, S Schmidt, F Smaill, B Thompson, S Trottier, S Vezina, S Walmsley.

Chile (49): MJ Wolff Reyes, R Northland..

Denmark (19): L Ostergaard, C Pedersen, H Nielsen, L Hergens, IR Loftheim, KB Jensen.

Estonia (5): M Raukas, K Zilmer.

Finland (21): J Justinen, M Ristola.

France (272): PM Girard, R Landman, S Abel, S Abgrall, K Amat, L Auperin, R Barruet, A Benalycherif, N Benammar, M Bensalem, M Bentata, JM Besnier, M Blanc, O Bouchaud, A Cabié, P Chavannet, JM Chennebault, S Dargere, X de la Tribonniere, T Debord, N Decaux, J Delgado, M Dupon, J Durant, V Frixon-Marin, C Genet, L Gérard, J Gilquin, B Hoen, V Jeantils, H Kouadio, P Leclercq, CP Michon, P Nau, J Pacanowski, C Piketty, I Poizot-Martin, I Raymond, D Salmon, JL Schmit, MA Serini, A Simon, S Tassi, F Touam, R Verdon, P Weinbreck, L Weiss, Y Yazdanpanah, P Yeni.

Germany (215): G Fätkenheuer, S Staszewski, F Bergmann, S Bitsch, JR Bogner, N Brockmeyer, S Esser, FD Goebel, M Hartmann, H Klinker, C Lehmann, T Lennemann, A Plettenberg, A Potthof, J Rockstroh, B Ross, A Stoehr, JC Wasmuth, K Wiedemeyer, R Winzer.

Greece(95): A Hatzakis, G Touloumi, A Antoniadou, GL Daikos, A Dimitrakaki, P Gargalianos-Kakolyris, A Karafoulidou, A Katsambas, O Katsarou, AN Kontos, T Kordossis, MK Lazanas, P Panagopoulos, G Panos, V Paparizos, V Papastamopoulos, G Petrikkos, A Skoutelis, N Tsogas, G Xylomenos.

Ireland (2): CJ Bergin, B Mooka.

Israel (13): S Pollack, MG Mamorksy, N Agmon-Levin, R Karplus, E Kedem, S Maayan, E Shahar, Z Sthoeger, D Turner, I Yust.

Italy (88): G Tambussi, V Rusconi, C Abeli, M Bechi, A Biglino, L Butini, G Carosi, S Casari, A Corpolongo, M De Gioanni, M Di Pietro, G D'Offizi, R Esposito, F Mazzotta, M Montroni, G Nardini, S Nozza, T Quirino, E Raise.

Japan (15): M Honda, M Ishisaka.

Lithuania (4): S Caplinskas, V Uzdaviniene.

Luxembourg (3): JC Schmit, T Staub.

Morocco (42): H Himmich, K Marhoum El Filali.

New Zealand (7): GD Mills, T Blackmore, JA Masters, J Morgan, A Pithie.

Norway (17): J Brunn, V Ormasssen.

Peru (57): A La Rosa, O Guerra, M Espichan, L Gutierrez, F Mendo, R Salazar.

Poland (54): B Knytz, A Horban, E Bakowska, M Beniowski, J Gasiorowski, J Kwiatkowski.

Portugal (73): F Antunes, RS Castro, M Doroana, A Horta, K Mansinho, AC Miranda, IV Pinto, E Valadas, J Vera.

Russia (17): A Rakhmanova, E Vinogradova, A Yakovlev, N Zakharova.

South Africa (26): R Wood, C Orrel.

Spain (100): J Gatell, JA Arnaiz, R Carrillo, B Clotet, D Dalmau, A González, Q Jordano, A Jou, H Knobel, M Larrousse, R Mata, JS Moreno, E Oretaga, JN Pena, F Pulido, R Rubio, J Sanz, P Viciana.

Switzerland (91): B Hirschel, R Spycher, M Battegay, E Bernasconi, S Bottone, M Cavassini, A Christen, C Franc, HJ Furrer, A Gayet-Ageron, D Genné, S Hochstrasser, L Magenta, C Moens, N Müller, R Nüesch.

Thailand (159): P Phanuphak, K Ruxrungtham, W Pumpradit, P Chetchotisakd, S Dangthongdee, S Kiertiburanakul, V Klinbuayaem, P Mootsikapun, S Nonenoy, B Piyavong, W Prasithsirikul, P Raksakulkarn.

United Kingdom (214): BG Gazzard, JG Ainsworth, J Anderson, BJ Angus, TJ Barber, MG Brook, DR Chadwick, M Chikohora, DR Churchill, D Cornforth, PJ Easterbrook, PA Fox, R Fox, PA Gomez, MM Gompels, GM Harris, S Herman, AGA Jackson, SPR Jebakumar, MA Johnson, GR Kinghorn, KA Kuldanek, N Larbalestier, C Leen, M Lumsden, T Maher, J Mantell, R Maw, S McKernan, L McLean, S Morris, L Muromba, CM Orkin, AJ Palfreeman, BS Peters, TEA Peto, SD Portsmouth, S Rajamanoharan, A Ronan, A Schwenk, MA Slinn, CJ Stroud, RC Thomas, MH Wansbrough-Jones, HJ Whiles, E Williams, IG Williams, M Youle.

United States (2989): DI Abrams, EA Acosta, S Adams, A Adamski, L Andrews, D Antoniskis, DR Aragon, R Arduino, R Artz, J Bailowitz, BJ Barnett, C Baroni, M Barron, JD Baxter, D Beers, M Beilke, D Bemenderfer, A Bernard, CL Besch, MT Bessesen, JT Bethel, S Blue, JD Blum, S Boarden, RK Bolan, JB Borgman, I Brar, BK Braxton, UF Bredeek, R Brennan, DE Britt, J Brockelman, S Brown, V Bruzzese, D Bulgin-Coleman, DE Bullock, V Cafaro, B Campbell, S Caras, J Carroll, KK Casey, F Chiang, G Childress, RB Cindrich, C Clark, M Climo, C Cohen, J Coley, DV Condoluci, R Contreras, J Corser, J Cozzolino, LR Crane, L Daley, D Dandridge, V D'Antuono, JG Darcourt Rizo Patron, JA DeHovitz, E DeJesus, J DesJardin, M Diaz-Linares, C Dietrich, P Dodson, E Dolce, K Elliott, D Erickson, M Estes, LL Faber, J Falbo, MJ Farrough, CF Farthing, P Ferrell-Gonzalez, H Flynn, C Frank, M Frank, KF Freeman, N French, G Friedland, N Fujita, L Gahagan, K Genther, I Gilson, MB Goetz, E Goodwin, F Graziano, CK Guity, P Gulick, ER Gunderson, CM Hale, K Hannah, H Henderson, K Hennessey, WK Henry, DT Higgins, SL Hodder, HW Horowitz, M Howe-Pittman, J Hubbard, R Hudson, H Hunter, C Hutelmyer, MT Insignares, L Jackson, L Jenny, M John, DL Johnson, G Johnson, J Johnson, L Johnson, J Kaatz, J Kaczmarski, S Kagan, C Kantor, T Kempner, K Kieckhaus, N Kimmel, BM Klaus, N Klimas, JR Koeppe, J Koirala, J Kopka, JR Kostman, MJ Kozal, A Kumar, A Labriola, H Lampiris, C Lamprecht, KM Lattanzi, J Lee, J Leggett, C Long, A Loquere, K Loveless, CJ Lucasti, R Luskin-Hawk, M MacVeigh, LH Makohon, S Mannheimer, NP Markowitz, C Marks, N Martinez, C Martorell, E McFeaters, B McGee, DM McIntyre, J McKee, E McManus, LG Melecio, D Melton, S Mercado, E Merrifield, JA Mieras, M Mogyoros, FM Moran, K Murphy, D Mushatt, S Mutic, I Nadeem, R Nahass, D Nixon, S O'Brien, A Ognjan, M O'Hearn, K O'Keefe, PC Okhuysen, E Oldfield, D Olson, R Orenstein, R Ortiz, J Osterberger, W Owen, F Parpart, V Pastore-Lange, S Paul, A Pavlatos, DD Pearce, R Pelz, G Perez, S Peterson, G Pierone Jr, D Pitrak, SL Powers, HC Pujet, JW Raaum, J Ravishankar, J Reeder, N Regevik, NA Reilly, C Reyelt, J Riddell IV, D Rimland, ML Robinson, AE Rodriguez, MC Rodriguez-Barradas, V Rodriguez Derouen, R Roland, C Rosmarin, WL Rossen, JR Rouff, JH Sampson, M Sands, C Savini, S Schrader, MM Schulte, C Scott, R Scott, H Seedhom, M Sension, A Sheble-Hall, A Sheridan, J Shuter, LN Slater, R Slotten, D Slowinski, M Smith, S Snap, C Somboonwit, DM States, M Stewart, G Stringer, J Sullivan, KK Summers, K Swanson, IB Sweeton, S Szabo, EM Tedaldi, EE Telzak, Z Temesgen, D Thomas, MA Thompson, S Thompson, C Ting Hong Bong, C Tobin, J Uy, A Vaccaro, LM Vasco, I Vecino, GK Verlinghieri, F Visnegarwala, BH Wade, V Watson, SE Weis, JA Weise, S Weissman, AM Wilkin, L Williams, JH Witter, L Wojtusic, TJ Wright, V Yeh, B Young, C Zeana, J Zeh.

Uruguay (3): E Savio, M Vacarezza.

Footnotes

Conflict of Interest

None reported

Reference List

  • 1.Mocroft A, Vella S, Benfield TL, et al. Changing patterns of mortality across Europe in patients infected with HIV-1. EuroSIDA Study Group. Lancet. 1998 November 28;352(9142):1725–30. doi: 10.1016/s0140-6736(98)03201-2. [DOI] [PubMed] [Google Scholar]
  • 2.Palella FJ, Jr., Delaney KM, Moorman AC, et al. Declining morbidity and mortality among patients with advanced human immunodeficiency virus infection. HIV Outpatient Study Investigators. N Engl J Med. 1998 March 26;338(13):853–60. doi: 10.1056/NEJM199803263381301. [DOI] [PubMed] [Google Scholar]
  • 3.Gupta SK, Eustace JA, Winston JA, et al. Guidelines for the management of chronic kidney disease in HIV-infected patients: recommendations of the HIV Medicine Association of the Infectious Diseases Society of America. Clin Infect Dis. 2005 June 1;40(11):1559–85. doi: 10.1086/430257. [DOI] [PubMed] [Google Scholar]
  • 4.Wali RK, Drachenberg CI, Papadimitriou JC, Keay S, Ramos E. HIV-1-associated nephropathy and response to highly-active antiretroviral therapy. Lancet. 1998 September 5;352(9130):783–4. doi: 10.1016/S0140-6736(98)24037-2. [DOI] [PubMed] [Google Scholar]
  • 5.Szczech LA, Edwards LJ, Sanders LL, et al. Protease inhibitors are associated with a slowed progression of HIV-related renal diseases. Clin Nephrol. 2002 May;57(5):336–41. doi: 10.5414/cnp57336. [DOI] [PubMed] [Google Scholar]
  • 6.Lucas GM, Eustace JA, Sozio S, Mentari EK, Appiah KA, Moore RD. Highly active antiretroviral therapy and the incidence of HIV-1-associated nephropathy: a 12-year cohort study. AIDS. 2004 February 20;18(3):541–6. doi: 10.1097/00002030-200402200-00022. [DOI] [PubMed] [Google Scholar]
  • 7.Friis-Moller N, Weber R, Reiss P, et al. Cardiovascular disease risk factors in HIV patients--association with antiretroviral therapy. Results from the DAD study. AIDS. 2003 May 23;17(8):1179–93. doi: 10.1097/01.aids.0000060358.78202.c1. [DOI] [PubMed] [Google Scholar]
  • 8.Wand H, Calmy A, Carey DL, et al. Metabolic syndrome, cardiovascular disease and type 2 diabetes mellitus after initiation of antiretroviral therapy in HIV infection. AIDS. 2007 November 30;21(18):2445–53. doi: 10.1097/QAD.0b013e3282efad32. [DOI] [PubMed] [Google Scholar]
  • 9.Gupta SK, Parker RA, Robbins GK, Dube MP. The effects of highly active antiretroviral therapy on albuminuria in HIV-infected persons: results from a randomized trial. Nephrol Dial Transplant. 2005 October;20(10):2237–42. doi: 10.1093/ndt/gfi053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Lucas GM, Mehta SH, Atta MG, et al. End-stage renal disease and chronic kidney disease in a cohort of African-American HIV-infected and at-risk HIV-seronegative participants followed between 1988 and 2004. AIDS. 2007 November 30;21(18):2435–43. doi: 10.1097/QAD.0b013e32827038ad. [DOI] [PubMed] [Google Scholar]
  • 11.Szczech LA, Grunfeld C, Scherzer R, et al. Microalbuminuria in HIV infection. AIDS. 2007 May 11;21(8):1003–9. doi: 10.1097/QAD.0b013e3280d3587f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Kalayjian RC, Franceschini N, Gupta SK, et al. Suppression of HIV-1 replication by antiretroviral therapy improves renal function in persons with low CD4 cell counts and chronic kidney disease. AIDS. 2008 February 19;22(4):481–7. doi: 10.1097/QAD.0b013e3282f4706d. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Shlipak MG, Sarnak MJ, Katz R, et al. Cystatin C and the risk of death and cardiovascular events among elderly persons. N Engl J Med. 2005 May 19;352(20):2049–60. doi: 10.1056/NEJMoa043161. [DOI] [PubMed] [Google Scholar]
  • 14.Madero M, Sarnak MJ, Stevens LA. Serum cystatin C as a marker of glomerular filtration rate. Curr Opin Nephrol Hypertens. 2006 November;15(6):610–6. doi: 10.1097/01.mnh.0000247505.71915.05. [DOI] [PubMed] [Google Scholar]
  • 15.Grubb A, Bjork J, Lindstrom V, Sterner G, Bondesson P, Nyman U. A cystatin C-based formula without anthropometric variables estimates glomerular filtration rate better than creatinine clearance using the Cockcroft-Gault formula. Scand J Clin Lab Invest. 2005;65(2):153–62. doi: 10.1080/00365510510013596. [DOI] [PubMed] [Google Scholar]
  • 16.Herget-Rosenthal S, Marggraf G, Husing J, et al. Early detection of acute renal failure by serum cystatin C. Kidney Int. 2004 September;66(3):1115–22. doi: 10.1111/j.1523-1755.2004.00861.x. [DOI] [PubMed] [Google Scholar]
  • 17.Dharnidharka VR, Kwon C, Stevens G. Serum cystatin C is superior to serum creatinine as a marker of kidney function: a meta-analysis. Am J Kidney Dis. 2002 August;40(2):221–6. doi: 10.1053/ajkd.2002.34487. [DOI] [PubMed] [Google Scholar]
  • 18.Massey D. Commentary: clinical diagnostic use of cystatin C. J Clin Lab Anal. 2004;18(1):55–60. doi: 10.1002/jcla.10098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Coll E, Botey A, Alvarez L, et al. Serum cystatin C as a new marker for noninvasive estimation of glomerular filtration rate and as a marker for early renal impairment. Am J Kidney Dis. 2000 July;36(1):29–34. doi: 10.1053/ajkd.2000.8237. [DOI] [PubMed] [Google Scholar]
  • 20.Dudgeon WD, Phillips KD, Carson JA, Brewer RB, Durstine JL, Hand GA. Counteracting muscle wasting in HIV-infected individuals. HIV Med. 2006 July;7(5):299–310. doi: 10.1111/j.1468-1293.2006.00380.x. [DOI] [PubMed] [Google Scholar]
  • 21.Jaroszewicz J, Wiercinska-Drapalo A, Lapinski TW, Prokopowicz D, Rogalska M, Parfieniuk A. Does HAART improve renal function? An association between serum cystatin C concentration, HIV viral load and HAART duration. Antivir Ther. 2006;11(5):641–5. [PubMed] [Google Scholar]
  • 22.Odden MC, Scherzer R, Bacchetti P, et al. Cystatin C level as a marker of kidney function in human immunodeficiency virus infection: the FRAM study. Arch Intern Med. 2007 November 12;167(20):2213–9. doi: 10.1001/archinte.167.20.2213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.El-Sadr WM, Lundgren JD, Neaton JD, et al. CD4+ count-guided interruption of antiretroviral treatment. N Engl J Med. 2006 November 30;355(22):2283–96. doi: 10.1056/NEJMoa062360. [DOI] [PubMed] [Google Scholar]
  • 24.Kuller L, the SMART STudy Group Elevated levels of Intereleukin-6 and D-Dimer are associated with an increased risk of death in patients with HIV. 2008 [Google Scholar]
  • 25.Levey AS, Bosch JP, Lewis JB, Greene T, Rogers N, Roth D. A more accurate method to estimate glomerular filtration rate from serum creatinine: a new prediction equation. Modification of Diet in Renal Disease Study Group. Ann Intern Med. 1999 March 16;130(6):461–70. doi: 10.7326/0003-4819-130-6-199903160-00002. [DOI] [PubMed] [Google Scholar]
  • 26.Cockcroft DW, Gault MH. Prediction of creatinine clearance from serum creatinine. Nephron. 1976;16(1):31–41. doi: 10.1159/000180580. [DOI] [PubMed] [Google Scholar]
  • 27.Tenstad O, Roald AB, Grubb A, Aukland K. Renal handling of radiolabelled human cystatin C in the rat. Scand J Clin Lab Invest. 1996 August;56(5):409–14. doi: 10.3109/00365519609088795. [DOI] [PubMed] [Google Scholar]
  • 28.Shlipak MG, Ix JH, Bibbins-Domingo K, Lin F, Whooley MA. Biomarkers to predict recurrent cardiovascular disease: the Heart and Soul Study. Am J Med. 2008 January;121(1):50–7. doi: 10.1016/j.amjmed.2007.06.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Finney H, Newman DJ, Thakkar H, Fell JM, Price CP. Reference ranges for plasma cystatin C and creatinine measurements in premature infants, neonates, and older children. Arch Dis Child. 2000 January;82(1):71–5. doi: 10.1136/adc.82.1.71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Finney H, Newman DJ, Price CP. Adult reference ranges for serum cystatin C, creatinine and predicted creatinine clearance. Ann Clin Biochem. 2000 January;37(Pt 1):49–59. doi: 10.1258/0004563001901524. [DOI] [PubMed] [Google Scholar]
  • 31.Vinge E, Lindergard B, Nilsson-Ehle P, Grubb A. Relationships among serum cystatin C, serum creatinine, lean tissue mass and glomerular filtration rate in healthy adults. Scand J Clin Lab Invest. 1999 December;59(8):587–92. doi: 10.1080/00365519950185076. [DOI] [PubMed] [Google Scholar]
  • 32.Knight EL, Verhave JC, Spiegelman D, et al. Factors influencing serum cystatin C levels other than renal function and the impact on renal function measurement. Kidney Int. 2004 April;65(4):1416–21. doi: 10.1111/j.1523-1755.2004.00517.x. [DOI] [PubMed] [Google Scholar]
  • 33.Singh D, Whooley MA, Ix JH, Ali S, Shlipak MG. Association of cystatin C and estimated GFR with inflammatory biomarkers: the Heart and Soul Study. Nephrol Dial Transplant. 2007 April;22(4):1087–92. doi: 10.1093/ndt/gfl744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Zakai NA, Katz R, Jenny NS, et al. Inflammation and hemostasis biomarkers and cardiovascular risk in the elderly: the Cardiovascular Health Study. J Thromb Haemost. 2007 June;5(6):1128–35. doi: 10.1111/j.1538-7836.2007.02528.x. [DOI] [PubMed] [Google Scholar]
  • 35.Vaziri ND, Moradi H. Mechanisms of dyslipidemia of chronic renal failure. Hemodial Int. 2006 January;10(1):1–7. doi: 10.1111/j.1542-4758.2006.01168.x. [DOI] [PubMed] [Google Scholar]
  • 36.Shlipak MG, Fried LF, Crump C, et al. Elevations of inflammatory and procoagulant biomarkers in elderly persons with renal insufficiency. Circulation. 2003 January 7;107(1):87–92. doi: 10.1161/01.cir.0000042700.48769.59. [DOI] [PubMed] [Google Scholar]
  • 37.Bruggeman LA, Ross MD, Tanji N, et al. Renal epithelium is a previously unrecognized site of HIV-1 infection. J Am Soc Nephrol. 2000 November;11(11):2079–87. doi: 10.1681/ASN.V11112079. [DOI] [PubMed] [Google Scholar]
  • 38.Winston JA, Bruggeman LA, Ross MD, et al. Nephropathy and establishment of a renal reservoir of HIV type 1 during primary infection. N Engl J Med. 2001 June 28;344(26):1979–84. doi: 10.1056/NEJM200106283442604. [DOI] [PubMed] [Google Scholar]
  • 39.Conaldi PG, Biancone L, Bottelli A, et al. HIV-1 kills renal tubular epithelial cells in vitro by triggering an apoptotic pathway involving caspase activation and Fas upregulation. J Clin Invest. 1998 December 15;102(12):2041–9. doi: 10.1172/JCI3480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Ray PE, Liu XH, Henry D, et al. Infection of human primary renal epithelial cells with HIV-1 from children with HIV-associated nephropathy. Kidney Int. 1998 May;53(5):1217–29. doi: 10.1046/j.1523-1755.1998.00900.x. [DOI] [PubMed] [Google Scholar]
  • 41.Ross MJ, Fan C, Ross MD, et al. HIV-1 infection initiates an inflammatory cascade in human renal tubular epithelial cells. J Acquir Immune Defic Syndr. 2006 May;42(1):1–11. doi: 10.1097/01.qai.0000218353.60099.4f. [DOI] [PubMed] [Google Scholar]
  • 42.Ross MJ, Wosnitzer MS, Ross MD, et al. Role of ubiquitin-like protein FAT10 in epithelial apoptosis in renal disease. J Am Soc Nephrol. 2006 April;17(4):996–1004. doi: 10.1681/ASN.2005070692. [DOI] [PubMed] [Google Scholar]
  • 43.Kirchner JT. Resolution of renal failure after initiation of HAART: 3 cases and a discussion of the literature. AIDS Read. 2002 March;12(3):103–2. [PubMed] [Google Scholar]
  • 44.Scheurer D. Rapid reversal of renal failure after initiation of HAART: a case report. AIDS Read. 2004 August;14(8):443–7. [PubMed] [Google Scholar]

RESOURCES