Skip to main content
Erschienen in: BMC Infectious Diseases 1/2019

Open Access 01.12.2019 | Research article

Schistosomiasis and HIV-1 viral load in HIV-infected outpatients with immunological failure in Tanzania: a case-control study

Erschienen in: BMC Infectious Diseases | Ausgabe 1/2019

Abstract

Background

Schistosoma sp. infection has been shown to interact with HIV-1 by modifying susceptibility to the virus and impacting AIDS outcome, but very little is known about the potential impact of Schistosoma sp. infection on the efficiency of antiretroviral treatment (ART) in HIV-1 infected individuals. One study suggested increased immunological failure in patients infected with schistosomes compared to those uninfected. To our knowledge, no report exists on the virological response to ART in schistosome-infected individuals. In addition, viral load in HIV-1 infected individuals changes over the course of the HIV infection. This study assessed the impact of HIV-1/Schistosoma sp. co-infections on viral load in people with immunological failure on ART, taking into account the duration of HIV-1 infection.

Methods

We enrolled HIV-1 infected Tanzanian adults over 18 years of age who had used first line ART for more than 6 months and were identified to have immunological failure by the WHO criteria (50% drop from peak CD4 count, or CD4 count equal to or below baseline after 6 months of ART, or CD4 count below 100cells/mm3 after 1 year of ART). Patients were also tested for schistosome infection by microscopy for ova in urine and stool and by circulating anodic antigen (CAA) levels in serum. The duration of HIV-1 infection was calculated using baseline CD4+ T-cell (CD4) counts determined at enrollment. Univariable and multivariable analyses were conducted to compare viral loads in schistosome infected and uninfected patients.

Results

A total of 188 patients were enrolled. After univariable analysis, female sex, lower peak CD4 counts, lower current CD4 counts, anemia, and shorter time infected with HIV-1 were all significantly associated with higher viral load. Schistosome infection was not associated with viral load even after adjusting for sex, current CD4 counts and duration of HIV-1 infection.

Conclusions

The current study of HIV-infected patients with immunological failure on ART suggests that once ART is introduced, ART is the dominant driver of viral load and schistosome infection may no longer have an impact.
Abkürzungen
95%CI
95% confidence interval
ART
Antiretrovial therapy
BMC
Bugando Medical Centre
CAA
Schistosome circulating anodic antigen
CD4
CD4+ T-cells
IQR
Interquartile range
N
Total number of observations
Th
T-helper
UCP-LF
Luminescent up-converting phosphor technology in combination with a lateral flow-based platform
WHO
World Health Organization

Background

Schistosoma sp. and HIV are co-endemic globally and an estimated 6 million individuals are co-infected worldwide [1, 2]. Schistosoma sp. infection has been shown to increase susceptibility to HIV and to impact HIV/AIDS outcomes [38]. It has been hypothesized that schistosome-induced down-regulation of T helper (Th)-1 immune responses may permit increased viral replication, and that T regulatory cells may also play an important role in the observed interactions.
Availability of antiretroviral therapy (ART) is increasing, with 61% of adults infected with HIV being on antiretroviral treatment in sub-Saharan Africa [9]. Yet very little is known about the impact of Schistosoma sp. infection on the effectiveness of ART. One study from Tanzania showed schistosome infection to be significantly associated with immunological failure and poorer CD4+ T-cells (CD4) count gain with ART [10]. However that study did not investigate whether the immunological failure was associated with concomitant increase of viral load (virological failure). Results from human studies of schistosome infection and HIV-1 RNA viral loads are variable, with some showing lower and some showing higher viral replication during schistosome co-infection [3, 1118].
Our group has recently demonstrated that people with schistosome infections have higher HIV-1 viral load set-points shortly following HIV acquisition, yet that in chronic HIV-schistosome co-infection, viral loads are lower than in those with HIV infection alone [19]. To our knowledge, no report exists on the virological response to ART in schistosome-infected individuals. Furthermore, controlling for duration of HIV-1 infection appears to be a critical factor to consider in studies of HIV-1 and schistosome co-infection, since HIV-1 viral load differs over the course of infection [19, 20].
In the current study, we aimed to assess the impact of HIV-1/Schistosoma sp. co-infections on viral load in HIV-1 infected people with immunological failure on ART, taking into account the duration of HIV-1 infection.

Methods

Study participants and enrollment

This study was conducted in HIV outpatient clinics at Bugando Medical Centre (BMC) and Sekou Toure Hospital in Mwanza. The participants were HIV-1 infected adults over 18 years of age, who had used first line ART for more than 6 months and were identified to have immunological failure by the World Health Organization (WHO) criteria. Patients on stavudine-containing regimens were excluded due to this drug’s known association with treatment failure [21, 22]. Eligible patients provided urine and stool samples for schistosomiasis testing by microscopy as well as serum for quantitation of schistosome circulating anodic antigen (CAA). Plasma was also collected for viral load measurement. Additional information was extracted from the HIV clinic database and the patient’s chart.

Laboratory methods

Tests for schistosome infections were performed at the National Institute of Medical Research in Mwanza, Tanzania. Microscopic examinations were performed on 10 mL of urine (for S. haematobium) by the filtration technique and on stool (for S. mansoni) following the Kato Katz method. CAA testing was performed as previously described using a luminescent up-converting phosphor technology in combination with a lateral flow-based platform (UCP-LF) [23, 24]. A 30 pg/mL cutoff threshold was used. Schistosome infection was defined as having either a positive microscopy or CAA test.
Plasma viral load was quantified using the AmpliPrep/COBAS® TaqMan® HIV-1 Test (Roche Molecular Systems Inc., Pleasanton, California, USA) machine at the BMC clinical laboratory, with a lower limit of detection of 20 copies/mL. Virological failure was defined as a viral load above 1000 copies/mL.

Statistical analysis

We estimated that we needed to enroll 190 patients to provide 80% power to detect the primary outcome, a predicted prevalence of virological treatment failure of 45% in those with schistosome infection compared to 25% in those without schistosomes, assuming that 30% of the study population would be infected with schistosomes [10]. Data was double entered, verified and cleaned using Microsoft Excel 2013 and analysis was performed using STATA version 13. Chi-square tests and t-tests were used to compare presence of demographic and clinical factors in those with versus without schistosome infection. Univariable and multivariable linear regressions were used to determine factors associated with log10 of the viral load, including schistosome infection. All variables significantly associated with the outcome in the univariable analysis were included in the stepwise multivariable analysis. We pre-specified that we would include schistosome status and duration of HIV-1 infection in the model.
We calculated the time delay between HIV infection and start of ART using the normal CD4 decay per calendar year in drug naïve individuals as previously described [19], assuming that the upper reference values of CD4 counts in healthy Tanzanians are 1278.9 cells/μL for men and 1406. 11 cells/μL for women [25]. In addition, we used the overall CD4 decay per calendar year of 34.5 cells/ μL per year that had been reported from Rakai, Uganda, which has a similar HIV-1 clade composition to Mwanza, Tanzania [2629].
We modeled decay by the square-root function [28], which meant we subtracted 5.87 cells0.5/μL0.5 from 35.76 cells0.5/μL0.5 for men and 37.51 cells0.5/μL0.5 for women per calendar year until the square root of CD4 count found at time of start of ART was reached. The time period for this to happen was considered to be the estimated period between HIV-1 acquisition and start of ART. The time between start of ART and date of viral load testing was then added to this variable to obtain the total duration of HIV-1 infection. This led to an estimated median time from seroconversion to ART initiation of 4.4[3. 11–5.1] years, which is similar to what was found using HIV surveillance data [Colombe et al., manuscript in revision].

Definitions

Immunological failure was defined as either a confirmed 50% drop from peak CD4 count (if known) during current treatment or as a CD4 count equal to or below baseline after 6-months of ART, or as a CD4 count below 100cells/mm3 after 1 year of ART as per WHO guidelines [30]. Poor ART adherence was defined as missing 3 doses on 2 or more days of ART within the last 3 months according to Tanzanian national guidelines [31]. This measure of adherence is routinely recorded at every monthly visit by nurses and doctors at the clinic [31].

Results

From August to December 2014, we screened 237 outpatients who were diagnosed with immunological treatment failure while using first-line ART. Of these, 31 were using a stavudine-containing regimen, 10 had been on ART for less than 6 months, 6 did not want to participate in the study, and viral load testing failed on 5 patients. Thus we enrolled and analyzed data from a total of 188 patients.
The median age was 41.0 [36.0–46.5] years and 116 (61.7%) of participants were female. The majority of participants resided in urban areas (152, 80.9%), were employed and selling food (89, 47. 3%), and had completed only primary education (118, 62. 11%). 63/87 (72%) of anemic patients were female (p = 0.005). 79/188 (42.0%) of patients had > 1000 viral copies/mL. Among those with viral loads < 1000 copies/mL, 94 (86.2%) were virologically suppressed with viral loads < 20 copies/mL. 85/173 (49.1%) had either a microscopy positive test or a positive CAA test for Schistosoma sp. (Table 1). More specifically, 19/170 (11.18%) had a stool microscopy positive test, no one had a urine microscopy positive test and 82/188 (43.62%) had a positive CAA test. Sixteen people were positive both by CAA and microscopy test.
Table 1
Demographic characteristics of 188 HIV-1 outpatients with immunological treatment failure on ART
Variable
Viral load > 1000 copies/mL
N (%) or Median [IQR]
N = 79
Viral load < = 1000 copies/mL
N (%) or Median [IQR]
N = 109
p-value
Female
53/79 (67.1%)
63/109 (57.8%)
0.20
Age in years
40 [34–46]
42 [37–48]
0.034
Level of education
  
0.44
 Informal
47/79 (59.5%)
71/109 (65.1%)
 
 Any primary schooling
21/79 (26.6%)
23/109 (21.1%)
 
 Any secondary schooling
3/79 (3.8%)
1/109 (0.9%)
 
 Any university
8/79 (10.1%)
14/109 (12.8%)
 
Time on ART
  
0.25
 6 m–1y
2/79 (2.5%)
1/109 (0.9%)
 
 1–2y
6/79 (7.6%)
18/109 (16.5%)
 
 2–3y
14/79 (17.7%)
16/109 (14.7%)
 
 >3y
57/79 (72.2%)
74/109 (67.9%)
 
ART Regimen
  
0.77
 AZT/3TC/NVP
35/79 (44.3%)
49/109 (45.0%)
 
 AZT/3TC/EFV
10/79 (12.7%)
19/109 (17.4%)
 
 TDF/3TC/EFV
22/79 (27.8%)
28/109 (25.7%)
 
 TDF/3TC/EFV
12/79 (15.2%)
13/109 (11.9%)
 
History of change ART Regimen
34/79 (43.0%)
40/109 (36.7%)
0.38
Peak CD4 (cells/μL)
358 [202–506]
450 [317–630]
0.0027
Current CD4 (cells/μL)
104 [47–179]
192 [125–239]
< 0.001
History of stopping ART
19/79 (24.1%)
16/109 (14.7%)
0.10
Use of herbal/alternative medicine
32/79 (40.5%)
54/109 (49.5%)
0.22
Other comorbidities
  
0.38
 None
73/79 (92.4%)
94/109 (86.2%)
 
 Hypertension
4/79 (5.1%)
8/109 (7.3%)
 
 Others (Diabetes, heart failure, asthma etc.)
2/79 (2.5%)
7/109 (6.4%)
 
Body mass index in kg/m2
21.5 [18.9–25.4]
22 [19.9–24.0]
0.65
Anemia (Hemoglobin< 12 g/dl)
50/79 (6.3%)
37/109 (33.9%)
< 0.001
Schistosome positivity
36/71 (50.7%)
49/102 (48.0%)
0.73
Ln of CAA (pg/ml)
3.05 [2.1–5.2]
2.8 [2.0–6.1]
0.65
Years infected with HIV-1
8.9 [6.6–11.1]
9.9 [7.7–11.4]
0.068
After univariable analysis, female sex, lower peak CD4 counts, lower current CD4 counts, anemia, and shorter time infected with HIV-1 were all significantly associated with higher log10 of viral load. Schistosome infection was not associated with log10 of viral load.
After multivariable analysis, the best fit model included sex and current CD4 counts. We kept duration of HIV-1 infection in the model. Results of both the univariable and multivariable analyses are presented in Table 2. Neither schistosome infection nor duration of HIV-1 infection was associated with log10 of viral load. The interaction between sex and schistosome status and its impact on log10 of viral load was not statistically significant.
Table 2
Results of univariable and multivariable analyses of factors associated with log10 of viral load
Variable
Univariable analysis
Multivariable analysis
Slope coefficient [95%CI]
p-value
Slope coefficient [95%CI]
p-value
Female
0.6 [0.07;1.2]
0.029
0.6 [0.05;1.2]
0.034
Age (as a continuous variable)
− 0.02 [− 0.05;0.006]
0.11
  
Level of education
 Informal
Ref
   
 Any primary schooling
0.2 [− 0.5;0.9]
0.56
  
 Any secondary schooling
−0.08 [− 1.8;1.6]
0.92
  
 Any university
0.03 [− 0.98;1.04]
0.95
  
Time on ART
 6 m-1y
Ref
   
 1–2y
−1.6 [−4.0;0.8]
0.20
  
 2–3y
−1.0 [− 3.4;1.4]
0.40
  
 >3y
− 0.7 [− 3.0;1.6]
0.53
  
History of change ART Regimen
0.2 [− 0.4;0.8]
0.42
  
Highest/Peak CD4 (cells/μL)
 < 200
Ref
   
 200–500
−0.8 [− 1.6;-0.05]
0.037
  
 500–1000
− 1.1 [− 1.9;-0.3]
0.011
  
 > 1000
−3.0 [− 6.2;0.2]
0.068
  
Current CD4 (cells/μL)
 < 100
Ref
 
Ref
 
 100–200
− 0.5 [− 1.2;0.08]
0.090
−0.5 [− 1.1;0.2]
0.15
 200–300
−1.4 [−2.2;-0.7]
< 0.001
-1.3 [− 2.1;-0.5]
0.001
 > 300
-2.8 [−5.0;-0.6]
0.014
-3.0 [− 5.2;-0.8]
0.007
History of stopping ART
−0.05 [− 0.7;0.6]
0.89
  
Use of herbal/alternative medicine
−0.1 [− 0.7;0.4]
0.61
  
Other comorbidities
 None
Ref
   
 Hypertension
−0.7 [− 1.9;0.6]
0.30
  
 Others (Diabetes, heart failure, asthma etc.)
−0.7 [− 2.2;0.8]
0.34
  
Anemia
0.8 [0.3–1.4]
0.005
  
Schistosome positivity
0.15 [− 0.5;0.8]
0.63
0.1 [−0.4;0.7]
0.68
Ln of CAA (in ln pg/mL)
0.03 [−0.04;0.1]
0.42
  
Years infected with HIV-1
0.1 [0.01;0.2]
0.027
0.06 [−0.06;0.2]
0.32
When looking at whether the median log10 of viral load between schistosome infected and non-infected patients changed over time, we found no significant differences (Fig. 1).

Discussion

We found no association between schistosome infection and viral loads among patients with immunological failure, including after adjustment for time infected with HIV-1. This suggests that observed effects of schistosome infection on HIV-1 viral load occur in the absence of ART and that, once ART commences, effective ART is the dominant driver of viral load. In addition, our finding suggests that the increased immunological failure found by Efraim et al. might be due to an effect of Schistosoma sp. on CD4 counts, but not on viral loads, in people on ART [10].
Our findings, adjusting for duration of HIV-1 infection, differ from those in ART-naïve patients in the same clinic, where schistosome-infection was associated with lower viral loads after adjustment for duration of HIV infection [19]. It is likely that the effect of ART, which typically decreases the viral load by several log10 fold copies/mL, is so strong that it would mask any effect of schistosome infection on viral load, which we estimated in our prior study to cause a difference of 0.7 log10 copies/mL in ART-naïve patients. Viral load dynamics of patients on ART appear to be vastly different from those of ART naïve patients, as suggested by the fact that baseline viral loads do not predict viral load rebounds on ART in previously ART-naïve patients [32].
Expectedly, higher peak and current CD4 counts were associated with lower viral load. This finding is consistent with several other studies in sub-Saharan Africa [33, 34]. Similarly the association between anemia and higher viral load has been shown before [35]. The significant association of female sex with higher viral load as observed in our study contradicted what is usually reported [33, 36, 37]. This might be due to the fact that women were significantly more likely to have anemia, which itself is associated with higher viral load and became insignificant when included in the multivariable analysis.
These results need to be considered in light of some limitations. The study included only people with immunological failure, implying that our cohort has overall low CD4 count and thus high viral loads. In addition, as immunological failure is also associated with specific host and viral genetics [38, 39], our overall findings may not directly apply to each individual patient on ART. Finally the sample size was calculated for a statistical analysis based on a binary outcome rather than a continuous one. By using viral load as a continuous variable, we lose some power. Nonetheless, the conclusions when using a binary outcome remain the same (Unadjusted Odds Ratio = 1.11[0.61–2.04], p = 0.73; Odds Ratio adjusted for CD4 counts and anemia = 0.99[0.50–1.95], p = 0.98).

Conclusions

In conclusion, our study suggests that schistosome infection does not impact viral load in HIV-1 infected patients immunologically failing on ART, likely due to the major effects of ART on viral load once ART is initiated.

Acknowledgements

We would like to thank Theresia Gabriel, Zulkhani Msuya, Shibide Mondea, Leonard Muhoja, Veronica Tungaraza and Revocatus Baraka (Bugando CTC) for helping with data collection. We want to thank Jim Todd (London School of Tropical Hygiene and Medicine) for his input on the calculation of the duration of HIV-1 infection.

Funding

This work was funded by the Catholic University for Health and Allied Sciences and the Wurzburg Foundation (all funding was awarded to P.M.). The Funding Body had no role in the design of the study and collection, analysis, and interpretation of data nor in writing the manuscript.

Availability of data and materials

The data that support the findings of this study are available from Revocatus Baraka (revocatusbaraka8​9@gmail.​com) but restrictions apply to the availability of these data, which were used under license for the current study, and so are not publicly available. De-identified data are however available from the authors upon reasonable request and with permission of Revocatus Baraka (revocatusbaraka8​9@gmail.​com).
All participants were recruited after providing written informed consent. Clearance was obtained from the joint CUHAS/BMC Research Ethics Committee [CREC/037/2014]. All results were made available immediately to clinicians and recorded in the patient’s medical record. All patients with schistosome infection received praziquantel 40 mg/kg in one dose on one single day.
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open Access This 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.
Literatur
2.
Zurück zum Zitat Mbah MLN, Poolman EM, Drain PK, Coffee MP, van der Werf MJ, Galvani AP. HIV and Schistosoma haematobium prevalences correlate in sub-Saharan Africa. Tropical Med Int Health. 2013;18(10):1174–9.CrossRef Mbah MLN, Poolman EM, Drain PK, Coffee MP, van der Werf MJ, Galvani AP. HIV and Schistosoma haematobium prevalences correlate in sub-Saharan Africa. Tropical Med Int Health. 2013;18(10):1174–9.CrossRef
3.
Zurück zum Zitat Downs JA, Dupnik KM, van Dam GJ, Urassa M, Lutonja P, Kornelis D, et al. Effects of schistosomiasis on susceptibility to HIV-1 infection and HIV-1 viral load at HIV-1 seroconversion: a nested case-control study. PLoS Negl Trop Dis. 2017;11(9):e0005968.CrossRef Downs JA, Dupnik KM, van Dam GJ, Urassa M, Lutonja P, Kornelis D, et al. Effects of schistosomiasis on susceptibility to HIV-1 infection and HIV-1 viral load at HIV-1 seroconversion: a nested case-control study. PLoS Negl Trop Dis. 2017;11(9):e0005968.CrossRef
4.
Zurück zum Zitat Kjetland EF, Ndhlovu PD, Gomo E, Mduluza T, Midzi N, Gwanzura L, et al. Association between genital schistosomiasis and HIV in rural Zimbabwean women. AIDS. 2006;20:593–600.CrossRef Kjetland EF, Ndhlovu PD, Gomo E, Mduluza T, Midzi N, Gwanzura L, et al. Association between genital schistosomiasis and HIV in rural Zimbabwean women. AIDS. 2006;20:593–600.CrossRef
5.
Zurück zum Zitat Downs JA, van Dam GJ, Changalucha JM, Corstjens PLAM, Peck RN, de Dood CJ, et al. Association of schistosomiasis and HIV infection in Tanzania. Am J Trop Med Hyg. 2012;87:868–73.CrossRef Downs JA, van Dam GJ, Changalucha JM, Corstjens PLAM, Peck RN, de Dood CJ, et al. Association of schistosomiasis and HIV infection in Tanzania. Am J Trop Med Hyg. 2012;87:868–73.CrossRef
6.
Zurück zum Zitat Brodish PH, Singh K. Association between schistosoma haematobium exposure and human immunodeficiency virus infection among females in Mozambique. Am J Trop Med Hyg. 2016;94(5):1040–4.CrossRef Brodish PH, Singh K. Association between schistosoma haematobium exposure and human immunodeficiency virus infection among females in Mozambique. Am J Trop Med Hyg. 2016;94(5):1040–4.CrossRef
7.
Zurück zum Zitat Downs JA, de Dood CJ, Dee HE, McGeehan M, Khan H, Marenga A, et al. Schistosomiasis and human immunodeficiency virus in men in Tanzania. Am J Trop Med Hyg. 2017;96(4):856–62.PubMedPubMedCentral Downs JA, de Dood CJ, Dee HE, McGeehan M, Khan H, Marenga A, et al. Schistosomiasis and human immunodeficiency virus in men in Tanzania. Am J Trop Med Hyg. 2017;96(4):856–62.PubMedPubMedCentral
10.
Zurück zum Zitat Efraim L, Peck RN, Kalluvya SE, Kabangila R, Mazigo HD, Mpondo B, et al. Schistosomiasis and impaired response to antiretroviral therapy among HIV-infected patients in Tanzania. J Acquir Immune Defic Syndr. 2013;62(5):e153–6.CrossRef Efraim L, Peck RN, Kalluvya SE, Kabangila R, Mazigo HD, Mpondo B, et al. Schistosomiasis and impaired response to antiretroviral therapy among HIV-infected patients in Tanzania. J Acquir Immune Defic Syndr. 2013;62(5):e153–6.CrossRef
11.
Zurück zum Zitat Lawn SD, Karanja DM, Mwinzia P, Andove J, Colley DG, Folks TM, et al. The effect of treatment of schistosomiasis on blood plasma HIV-1 RNA concentration in coinfected individuals. AIDS. 2000;14(16):2437–43.CrossRef Lawn SD, Karanja DM, Mwinzia P, Andove J, Colley DG, Folks TM, et al. The effect of treatment of schistosomiasis on blood plasma HIV-1 RNA concentration in coinfected individuals. AIDS. 2000;14(16):2437–43.CrossRef
12.
Zurück zum Zitat Elliott AM, Mawa PA, Joseph S, Namujju PB, Kizza M, Nakiyingi JS, et al. Associations between helminth infection and CD4+ T cell count, viral load and cytokine responses in HIV-1-infected Ugandan adults. Trans R Soc Trop Med Hyg. 2003;97(1):103–8.CrossRef Elliott AM, Mawa PA, Joseph S, Namujju PB, Kizza M, Nakiyingi JS, et al. Associations between helminth infection and CD4+ T cell count, viral load and cytokine responses in HIV-1-infected Ugandan adults. Trans R Soc Trop Med Hyg. 2003;97(1):103–8.CrossRef
13.
Zurück zum Zitat Brown M, Kizza M, Watera C, Quigley MA, Rowland S, Hughes P, et al. Helminth infection is not associated with faster progression of HIV disease in coinfected adults in Uganda. J Infect Dis. 2004;190(10):1869–79.CrossRef Brown M, Kizza M, Watera C, Quigley MA, Rowland S, Hughes P, et al. Helminth infection is not associated with faster progression of HIV disease in coinfected adults in Uganda. J Infect Dis. 2004;190(10):1869–79.CrossRef
14.
Zurück zum Zitat Brown M, Mawa PA, Joseph S, Bukusuba J, Watera C, Whitworth JA, et al. Treatment of Schistosoma mansoni infection increases helminth-specific type 2 cytokine responses and HIV-1 loads in coinfected Ugandan adults. J Infect Dis. 2005;191(10):1648–57.CrossRef Brown M, Mawa PA, Joseph S, Bukusuba J, Watera C, Whitworth JA, et al. Treatment of Schistosoma mansoni infection increases helminth-specific type 2 cytokine responses and HIV-1 loads in coinfected Ugandan adults. J Infect Dis. 2005;191(10):1648–57.CrossRef
15.
Zurück zum Zitat Kallestrup P, Zinyama R, Gomo E, Butterworth AE, Mudenge B, van Dam GJ, et al. Schistosomiasis and HIV-1 infection in rural Zimbabwe: effect of treatment of schistosomiasis on CD4 cell count and plasma HIV-1 RNA load. J Infect Dis. 2005;192(11):1956–61.CrossRef Kallestrup P, Zinyama R, Gomo E, Butterworth AE, Mudenge B, van Dam GJ, et al. Schistosomiasis and HIV-1 infection in rural Zimbabwe: effect of treatment of schistosomiasis on CD4 cell count and plasma HIV-1 RNA load. J Infect Dis. 2005;192(11):1956–61.CrossRef
17.
Zurück zum Zitat Walson J, Singa B, Sangaré L, Naulikha J, Piper B, Richardson B, et al. Empiric deworming to delay HIV disease progression in adults with HIV who are ineligible for initiation of antiretroviral treatment (the HEAT study): a multi-site, randomised trial. Lancet Infect Dis. 2012;12(12):925–32.CrossRef Walson J, Singa B, Sangaré L, Naulikha J, Piper B, Richardson B, et al. Empiric deworming to delay HIV disease progression in adults with HIV who are ineligible for initiation of antiretroviral treatment (the HEAT study): a multi-site, randomised trial. Lancet Infect Dis. 2012;12(12):925–32.CrossRef
19.
Zurück zum Zitat Colombe S, Corstjens PLAM, de Dood CJ, Miyaye M, Magawa RG, Mngara J, et al. HIV-1 viral loads are not elevated in individuals co-infected with Schistosoma spp. after adjustment for duration of HIV-1 infection. Front Immunol. 2018;9:2005.CrossRef Colombe S, Corstjens PLAM, de Dood CJ, Miyaye M, Magawa RG, Mngara J, et al. HIV-1 viral loads are not elevated in individuals co-infected with Schistosoma spp. after adjustment for duration of HIV-1 infection. Front Immunol. 2018;9:2005.CrossRef
20.
Zurück zum Zitat Walson JL, John-Stewart G. Treatment of helminth co-infection in individuals with HIV-1: a systematic review of the literature. PLoS Negl Trop Dis. 2007;1(3):e102.CrossRef Walson JL, John-Stewart G. Treatment of helminth co-infection in individuals with HIV-1: a systematic review of the literature. PLoS Negl Trop Dis. 2007;1(3):e102.CrossRef
21.
Zurück zum Zitat Menezes CN, Maskew M, Sanne I, Crowther NJ, Raal FJ. A longitudinal study of stavudine-associated toxicities in a large cohort of South African HIV infected subjects. BMC Infect Dis. 2011;11:244.CrossRef Menezes CN, Maskew M, Sanne I, Crowther NJ, Raal FJ. A longitudinal study of stavudine-associated toxicities in a large cohort of South African HIV infected subjects. BMC Infect Dis. 2011;11:244.CrossRef
22.
Zurück zum Zitat van Griensven J, De Naeyer L, Mushi T, Ubarijoro S, Gashumba D, Gazille C, et al. High prevalence of lipoatrophy among patients on stavudine-containing first-line antiretroviral therapy regimens in Rwanda. Trans R Soc Trop Med Hyg. 2007;101(8):793–8.CrossRef van Griensven J, De Naeyer L, Mushi T, Ubarijoro S, Gashumba D, Gazille C, et al. High prevalence of lipoatrophy among patients on stavudine-containing first-line antiretroviral therapy regimens in Rwanda. Trans R Soc Trop Med Hyg. 2007;101(8):793–8.CrossRef
24.
Zurück zum Zitat van Dam GJ, de Dood CJ, Lewis M, Deelder AM, van Lieshout L, Tanke HJ, et al. A robust dry reagent lateral flow assay for diagnosis of active schistosomiasis by detection of Schistosoma circulating anodic antigen. Exp Parasitol. 2013;135(2):274–82.CrossRef van Dam GJ, de Dood CJ, Lewis M, Deelder AM, van Lieshout L, Tanke HJ, et al. A robust dry reagent lateral flow assay for diagnosis of active schistosomiasis by detection of Schistosoma circulating anodic antigen. Exp Parasitol. 2013;135(2):274–82.CrossRef
25.
Zurück zum Zitat Ngowi BJ, Mfinanga SG, Bruun JN, Morkve O. Immunohaematological reference values in human immunodeficiency virus-negative adolescent and adults in rural northern Tanzania. BMC Inf Dis. 2009;9:1.CrossRef Ngowi BJ, Mfinanga SG, Bruun JN, Morkve O. Immunohaematological reference values in human immunodeficiency virus-negative adolescent and adults in rural northern Tanzania. BMC Inf Dis. 2009;9:1.CrossRef
27.
Zurück zum Zitat Kasang C, Kalluvya S, Majinge C, Stich A, Bodem J, Kongola G, et al. HIV drug resistance (HIVDR) in antiretroviral therapy-naive patients in Tanzania not eligible for WHO threshold HIVDR survey is dramatically high. PLoS One. 2011;6(8):e23091.CrossRef Kasang C, Kalluvya S, Majinge C, Stich A, Bodem J, Kongola G, et al. HIV drug resistance (HIVDR) in antiretroviral therapy-naive patients in Tanzania not eligible for WHO threshold HIVDR survey is dramatically high. PLoS One. 2011;6(8):e23091.CrossRef
28.
Zurück zum Zitat Kiwanuka N, Robb M, Laeyendecker O, Kigozi G, Wabwire-Mangen F, Makumbi FE, et al. HIV-1 viral subtype differences in the rate of CD4+ T-cell decline among HIV seroincident antiretroviral naive persons in Rakai district, Uganda. J Acquir Immune Defic Syndr. 2010;54(2):180–4.PubMedPubMedCentral Kiwanuka N, Robb M, Laeyendecker O, Kigozi G, Wabwire-Mangen F, Makumbi FE, et al. HIV-1 viral subtype differences in the rate of CD4+ T-cell decline among HIV seroincident antiretroviral naive persons in Rakai district, Uganda. J Acquir Immune Defic Syndr. 2010;54(2):180–4.PubMedPubMedCentral
29.
Zurück zum Zitat Ndembi N, Lyagoba F, Nanteza B, Kushemererwa G, Serwanga J, Katongole-Mbidde E. Transmitted antiretroviral drug resistance surveillance among newly HIV type 1-diagnosed women attending an antenatal clinic in Entebbe, Uganda. AIDS Res Hum Retrovir. 2008;24(6):889–95.CrossRef Ndembi N, Lyagoba F, Nanteza B, Kushemererwa G, Serwanga J, Katongole-Mbidde E. Transmitted antiretroviral drug resistance surveillance among newly HIV type 1-diagnosed women attending an antenatal clinic in Entebbe, Uganda. AIDS Res Hum Retrovir. 2008;24(6):889–95.CrossRef
32.
Zurück zum Zitat Phillips AN, Staszewski S, Weber R, Kirk O, Francioli P, Miller V, et al. HIV viral load response to antiretroviral therapy according to the baseline CD4 cell count and viral load. JAMA. 2001;286(20):2560–7.CrossRef Phillips AN, Staszewski S, Weber R, Kirk O, Francioli P, Miller V, et al. HIV viral load response to antiretroviral therapy according to the baseline CD4 cell count and viral load. JAMA. 2001;286(20):2560–7.CrossRef
33.
Zurück zum Zitat Marconi VC, Wu B, Hampton J, Ordóñez CE, Johnson BA, Singh D, et al. Early warning indicators for first-line virologic failure independent of adherence measures in a south African urban clinic. AIDS Patient Care STDs. 2013;27(12):657–68.CrossRef Marconi VC, Wu B, Hampton J, Ordóñez CE, Johnson BA, Singh D, et al. Early warning indicators for first-line virologic failure independent of adherence measures in a south African urban clinic. AIDS Patient Care STDs. 2013;27(12):657–68.CrossRef
34.
Zurück zum Zitat Evans DH, Maskew M, Fox M, McNamara L, MacPhail P, Mathews C, et al. CD4 criteria improves the sensitivity of a clinical algorithm developed to identify viral failure in HIV-positive patients on first-line antiretroviral therapy. J Int AIDS Soc. 2012;15:122–3. Evans DH, Maskew M, Fox M, McNamara L, MacPhail P, Mathews C, et al. CD4 criteria improves the sensitivity of a clinical algorithm developed to identify viral failure in HIV-positive patients on first-line antiretroviral therapy. J Int AIDS Soc. 2012;15:122–3.
36.
Zurück zum Zitat Chalamilla G, Hawkins C, Okuma J, Spiegelman D, Aveika A, Christian B, et al. Mortality and treatment failure among HIV-infected adults in Dar Es Salaam, Tanzania. J Int Assoc Physicians AIDS Care (Chic). 2012;11(5):296–304.CrossRef Chalamilla G, Hawkins C, Okuma J, Spiegelman D, Aveika A, Christian B, et al. Mortality and treatment failure among HIV-infected adults in Dar Es Salaam, Tanzania. J Int Assoc Physicians AIDS Care (Chic). 2012;11(5):296–304.CrossRef
37.
Zurück zum Zitat Penot P, Héma A, Bado G, Kaboré F, Soré I, Sombié D, et al. The vulnerability of men to virologic failure during antiretroviral therapy in a public routine clinic in Burkina Faso. J Int AIDS Soc. 2014;17:1–7.CrossRef Penot P, Héma A, Bado G, Kaboré F, Soré I, Sombié D, et al. The vulnerability of men to virologic failure during antiretroviral therapy in a public routine clinic in Burkina Faso. J Int AIDS Soc. 2014;17:1–7.CrossRef
Metadaten
Titel
Schistosomiasis and HIV-1 viral load in HIV-infected outpatients with immunological failure in Tanzania: a case-control study
Publikationsdatum
01.12.2019
Erschienen in
BMC Infectious Diseases / Ausgabe 1/2019
Elektronische ISSN: 1471-2334
DOI
https://doi.org/10.1186/s12879-019-3876-8

Weitere Artikel der Ausgabe 1/2019

BMC Infectious Diseases 1/2019 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Therapiestart mit Blutdrucksenkern erhöht Frakturrisiko

25.04.2024 Hypertonie Nachrichten

Beginnen ältere Männer im Pflegeheim eine Antihypertensiva-Therapie, dann ist die Frakturrate in den folgenden 30 Tagen mehr als verdoppelt. Besonders häufig stürzen Demenzkranke und Männer, die erstmals Blutdrucksenker nehmen. Dafür spricht eine Analyse unter US-Veteranen.

Update Innere Medizin

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