1
|
Agwu AO, Egwu CO, Okorocha AE, Enyanwuma I, Amadi CC, Okpokoro E, Akpabio FP, Aguwa CO, Onwu D, Nwokoro O. The Efficacy of Cotrimoxazole for the Prevention of Pneumocystis jirovecii Pneumonia Among HIV-Exposed and Infected Children: A Systematic Review. EPIDEMIOLOGIA 2025; 6:8. [PMID: 39982260 PMCID: PMC11843829 DOI: 10.3390/epidemiologia6010008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND HIV-related opportunistic infections like Pneumocystis jirovecii Pneumonia (PCP) remain a major contributor to child morbidity and mortality globally. PCP accounts for over 60% of AIDS in the first year of life and is responsible for a third of AIDS in children globally. Cotrimoxazole prophylaxis, which is an intervention directed towards tackling this burden, has not attained remarkable coverage despite advocacy towards scale-up. This work was therefore aimed at evaluating the efficacy of cotrimoxazole in the prevention of PCP among children exposed to and infected with HIV by carrying out a systematic review. METHODS Key scientific databases were searched for primary studies not older than 15 years old without language restrictions. Randomized Control Trials (RCTs) and Cohorts comparing the effectiveness of cotrimoxazole versus placebo in the prevention of PCP among children (<17 years) exposed to and infected with HIV were selected. Studies with a duration of follow-up not less than 3 months long were included. A meta-analysis was conducted on RevMan 5.3 statistical application software following data extraction, and the data quality and risk of bias were also assessed. Exactly Ten (10) studies were selected and analyzed. FINDINGS It was observed that cotrimoxazole had beneficial effects in terms of a reduction in mortality among HIV-exposed and infected children, as 72 fewer children in 1000 (based on an absolute 95% CI) will die as a result of cotrimoxazole compared to a placebo. Cotrimoxazole also significantly reduces hospital admissions (p-value of 0.008). The adverse events associated with cotrimoxazole are comparable to a placebo when co-administered with ARTS (p = 0.90), which did not impact adherence. CONCLUSION The benefits of cotrimoxazole prophylaxis far outweigh its risks. Therefore, scaling up the intervention is recommended as a prophylactic for wider coverage, especially in resource-limited settings.
Collapse
Affiliation(s)
- Anthony O. Agwu
- Department of Public Health, University of Chester, Chester CH1 4BJ, UK
| | - Chinedu Ogbonnia Egwu
- Medical Biochemistry Department, College of Medicine, Alex-Ekwueme Federal University Ndufu-Alike, P.M.B. 1010, Ikwo 482131, Nigeria;
| | - Albert Egwu Okorocha
- Physiology Department, College of Medicine, Alex-Ekwueme Federal University Ndufu-Alike, P.M.B. 1010, Ikwo 482131, Nigeria;
| | - Ifeanyi Enyanwuma
- Surgery Department, Alex Ekwueme Federal University Teaching Hospital, Abakaliki 480282, Nigeria; (I.E.); (O.N.)
| | - Cyril C. Amadi
- Medical Biochemistry Department, College of Medicine, Alex-Ekwueme Federal University Ndufu-Alike, P.M.B. 1010, Ikwo 482131, Nigeria;
| | - Evaezi Okpokoro
- International Research Center of Excellence, Institute of Human Virology, Abuja 900246, Nigeria;
| | | | | | - Donatus Onwu
- Orthopedics Department, Alex Ekwueme Federal University Teaching Hospital, Abakaliki 480282, Nigeria;
| | - Onyedikachi Nwokoro
- Surgery Department, Alex Ekwueme Federal University Teaching Hospital, Abakaliki 480282, Nigeria; (I.E.); (O.N.)
| |
Collapse
|
2
|
Begnel ER, Ojee E, Adhiambo J, Mabele E, Wandika B, Ogweno V, Lim ES, Gantt S, Kinuthia J, Lehman DA, Slyker J, Wamalwa D. The Linda Kizazi study: a comparison of morbidity and mortality from birth to 2 years between children who are HIV-unexposed and HIV-exposed, uninfected in the era of universal antiretroviral therapy. BMJ Glob Health 2025; 10:e015841. [PMID: 39828431 PMCID: PMC11748780 DOI: 10.1136/bmjgh-2024-015841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 12/19/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Historically, children who are HIV-exposed, uninfected (CHEU) have been found to have greater morbidity and mortality than children who are HIV-unexposed, uninfected (CHUU). To assess whether this difference persists in the era of universal antiretroviral therapy (ART), we conducted a cohort study to compare the risk of acute diarrhoea, respiratory tract infections (RTI), malaria, hospitalisation, and all-cause mortality between Kenyan CHEU and CHUU from birth to 2 years. METHODS From December 2018 to March 2020 at Mathare North Health Centre in Nairobi, we recruited pregnant women living with HIV on ART for ≥6 months and pregnant women without HIV from the same community. We followed the mother-infant pairs for 2 years post partum and collected data on symptoms of illness, clinical visits and diagnoses, and infant feeding every 3 months; a self-selected subset of participants also received weekly data collection for up to 1 year. We compared the risk of each outcome between CHEU versus CHUU using HRs from Andersen-Gill (recurrent morbidity outcomes) and Cox proportional hazards (mortality) regression models adjusted for maternal age, marital status and education level. RESULTS Among 187 mother-infant pairs with postpartum data, 86 (46%) infants were CHEU and 101 (54%) were CHUU. All initiated breastfeeding, and 88% of CHEU and 57% of CHUU were exclusively breastfed (EBF) for ≥6 months. There was no significant difference in risk of diarrhoea (HR=0.79, 95% CI 0.52 to 1.22), malaria (HR=0.44, 95% CI 0.16 to 1.21), hospitalisation (HR=1.11, 95% CI 0.30 to 4.14), or mortality (HR=1.87, 95% CI 0.17 to 20.5). However, CHEU had lower risk of any RTI (HR=0.60, 95% CI 0.44 to 0.82) and pneumonia (HR=0.29, 95% CI 0.091 to 0.89). CONCLUSIONS CHEU born to women on effective long-term ART experienced similar overall morbidity and mortality as CHUU. However, CHEU had substantially lower risk of pneumonia and other RTI, possibly due to longer EBF in this group.
Collapse
Affiliation(s)
- Emily R Begnel
- Global Health, Epidemiology, University of Washington, Seattle, Washington, USA
| | - Ednah Ojee
- Paediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Judy Adhiambo
- Paediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Eliza Mabele
- Paediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Brenda Wandika
- Paediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Vincent Ogweno
- Paediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Efrem S Lim
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
| | - Soren Gantt
- Microbiologie, Infectiologie et Immunologie, Universite de Montreal Centre de Recherche du CHU St-Justine, Montreal, Quebec, Canada
| | - John Kinuthia
- Department of Research and Programs, Kenyatta National Hospital, Nairobi, Kenya
- Global Health, University of Washington, Seattle, Washington, USA
| | - Dara A Lehman
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Jennifer Slyker
- Global Health, Epidemiology, University of Washington, Seattle, Washington, USA
| | - Dalton Wamalwa
- Paediatrics and Child Health, University of Nairobi, Nairobi, Kenya
- Global Health, University of Washington, Seattle, Washington, USA
| |
Collapse
|
3
|
Frosch AEP, Musiime V, Staley C, Conroy AL, Rutebarika D, Ategeka G, Cusick SE. Safety and efficacy of iron supplementation with 3 months of daily ferrous sulphate in children living with HIV and mild-to-moderate anaemia in Uganda: a double-blind, randomised, placebo-controlled trial. Lancet HIV 2024; 11:e727-e735. [PMID: 39401504 PMCID: PMC12001907 DOI: 10.1016/s2352-3018(24)00238-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 11/01/2024]
Abstract
BACKGROUND Iron deficiency is the most common nutritional deficiency in the world, but iron supplementation can increase risk of opportunistic infections, especially in children living with HIV. We aimed to assess the effect of supplemental iron on haemoglobin concentration in children living with HIV and mild-to-moderate anaemia in Uganda. METHODS We did a double-blind, randomised, placebo-controlled trial of iron supplementation in children aged 6 months to 12 years living with HIV at two sites (ie, Kampala and Fort Portal, Uganda). Inclusion criteria were confirmed diagnosis of HIV and stable treatment with antiretroviral therapy for at least 6 months. Exclusion criteria were already taking iron supplementation, acute illness, current opportunistic infection, fever, known sickle cell disease, severe undernutrition, or any chronic illness requiring medical attention. Children were randomly assigned (1:1) via simple randomisation to an 84-day course of either ferrous sulphate or identical placebo tablets once per day. Randomisation codes were computer-generated and stratified by age (ie, 6-23 months or 24 months and older) by the Toronto Institute of Pharmaceutical Technology, the tablet manufacturer. Participants and all individuals giving the interventions, assessing outcomes, and analysing data were masked to group assignment. Children aged 6-23 months received tablets of 12·5 mg ferrous sulphate or identical placebo; children aged 24 months or older received tablets of 30·0 mg ferrous sulphate or identical placebo. Caregivers were instructed to give the supplement after a meal, preferably after an evening meal. The primary outcome was mean haemoglobin concentration at day 84. All analyses were intention to treat. This trial is registered at ClinicalTrials.gov (NCT03596996). FINDINGS Between May 5, 2018, and Nov 6, 2019, 973 children living with HIV were screened, of whom 200 (20%) met all inclusion criteria and were enrolled. 102 (51%) were randomly assigned to receive iron and 98 (49%) to receive placebo. In the iron group, 57 (56%) of 102 children were male and 45 (44%) were female. In the placebo group, 44 (45%) of 98 children were male and 54 (55%) were female. Iron supplementation was associated with improvement in haemoglobin in unadjusted analysis (p=0·029), but not adjusted analysis (p=0·10), and with improvement in ferritin and hepcidin in both adjusted (p=0·0046; p=0·0079) and unadjusted (p<0·0001; p<0·0001) analyses at day 84. There were four hospital admissions, all for children in the iron group; none were fatal: two children were admitted to hospital with pneumonia, one with severe malaria, and one with hepatitis. Frequency of admissions was not significantly different between groups (p=0·12). INTERPRETATION Iron could have haematological benefit and improve iron status in children living with HIV in Uganda. Future studies powered for morbidity outcomes with longer follow-up are needed, as are those that evaluate the effects of iron supplementation on neurocognitive outcomes. FUNDING Minnesota Masonic Charities, the Department of Pediatrics at the University of Minnesota, the Hennepin Healthcare Research Institute, and the US National Institutes of Health.
Collapse
Affiliation(s)
- Anne E P Frosch
- Department of Medicine, School of Medicine, University of Minnesota, Minneapolis, MN, USA; Hennepin Healthcare Research Institute, Minneapolis, MN, USA
| | - Victor Musiime
- Joint Clinical Research Centre, Kampala, Uganda; Department of Paediatrics and Child Health, College of Health Sciences, School of Medicine, Makerere University, Kampala, Uganda
| | - Christopher Staley
- Department of Surgery, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Andrea L Conroy
- Department of Pediatrics, School of Medicine, Indiana University, Indianapolis, IN, USA
| | | | | | - Sarah E Cusick
- Division of Epidemiology and Clinical Research, Department of Pediatrics, School of Medicine, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
4
|
Samsami M, Parsaeian F, Haghbin Toutounchi A, Khoshnoudi H, Tahmasbi H. The impact of cotrimoxazole in idiopathic granulomatous mastitis treatment. Int J Surg Case Rep 2024; 121:109959. [PMID: 38945013 PMCID: PMC11261417 DOI: 10.1016/j.ijscr.2024.109959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/22/2024] [Accepted: 06/25/2024] [Indexed: 07/02/2024] Open
Abstract
INTRODUCTION AND IMPORTANCE Idiopathic granulomatous mastitis (IGM) is a benign inflammatory breast disease, commonly presented with a sensitive breast lump and developing scars. Currently, there is no definitive treatment for IGM but Antibiotics, steroids, immunosuppressive drugs or a surgical treatments are the usual options. This case series aimed to evaluate the effectiveness of cotrimoxazole in treatment of IGM as there is no clinical consensus on the best and most widely acknowledged therapeutic management for IGM. CASE PRESENTATION All IGM patients were treated by Cotrimoxazole (800 mg BD for one week), and they were assessed at a month, 3 months, and 6 months after that. The primary outcome was an improvement in presenting complaints and symptoms such as palpable mass, bulging, pain, erythema and hypersensitivity of breast skin, breast discharge and fluctuation. The secondary outcome was the refractory rate within 6 months. Number of 20 patients were included. At the baseline, participants exhibited various symptoms such as bulging, pain and erythema (100 %), breast discharge (80 %), and fluctuation (30 %). After the intervention, there was a significant decrease in the prevalence of symptoms over the study period. The prevalence of bulging and pain, erythema, discharge, and fluctuation symptoms were decreasedto 5 %, 0 %, and 0 %, respectively. The refractory rate of IGM within six months of cotrimoxazole treatment was estimated 30 %. CLINICAL DISCUSSION In this study, the treatment approach did not involve corticosteroids and invasive procedures and the recurrence rate of IGM within the six months was lower than in similar studies that employed steroids alone or any more invasive treatments. Additionally, our study showed a high healing rate with resolution of inflammation, pain, discharge, and fluctuation. These results suggest that cotrimoxazole may be a more favorable option than high-dose corticosteroids and a comparable alternative to low-dose corticosteroids regarding recurrence rates. CONCLUSION Cotrimoxazole may be an effective treatment option for idiopathic granulomatous mastitis. However, further research is needed on different treatment options.
Collapse
Affiliation(s)
- Majid Samsami
- Department of General Surgery, Imam Hossein medical and educational center, Shahid Beheshti University of medical sciences, Tehran, Iran.
| | - Fatemeh Parsaeian
- Department of General Surgery, Imam Hossein medical and educational center, Shahid Beheshti University of medical sciences, Tehran, Iran.
| | - Alireza Haghbin Toutounchi
- Department of General Surgery, Imam Hossein medical and educational center, Shahid Beheshti University of medical sciences, Tehran, Iran.
| | - Hojatolah Khoshnoudi
- Department of General Surgery, Imam Hossein medical and educational center, Shahid Beheshti University of medical sciences, Tehran, Iran.
| | - Hamed Tahmasbi
- Department of General Surgery, Imam Hossein medical and educational center, Shahid Beheshti University of medical sciences, Tehran, Iran.
| |
Collapse
|
5
|
Omololu A, Onukak A, Effiong M, Oke O, Isa SE, Habib AG. Hospitalization and mortality outcomes among adult persons living with HIV in a tertiary hospital in South-western Nigeria: A cross-sectional study. PLOS GLOBAL PUBLIC HEALTH 2024; 4:e0003487. [PMID: 38990938 PMCID: PMC11238994 DOI: 10.1371/journal.pgph.0003487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 06/21/2024] [Indexed: 07/13/2024]
Abstract
HIV infection continues to be a major public health issue, with significant morbidity and mortality especially in resource poor areas. Infection with HIV results in an increased risk of opportunistic infections and other complications, which may lead to hospital admission and death. Morbidity and mortality patterns among hospitalized persons living with HIV (PLHIV) have been well documented in high income countries, but there is paucity of such data in Nigeria. We investigated the reasons for hospitalization and predictors of death among adult PLHIV at the Federal Medical Center (FMC) Abeokuta, Nigeria. This was a hospital based cross-sectional study carried out over a 15-month period between January 2018 and March 2019. All consenting hospitalized adult PLHIV who met the inclusion criteria were enrolled into the study. Causes of hospitalization and death were obtained and analyzed. Over the study period, 193 hospitalizations of PLHIV were studied. Although a number of clinical syndromes were documented, Sepsis and Tuberculosis were the commonest causes of hospitalization and mortality. Mortality rate was 37(19.2%) for outcomes on day 30, with anaemia [OR 3.00 (95% C.I: 1.04-8.67)], poor adherence with Cotrimoxazole [OR 4.07 (95% C.I: 1.79-9.28)], poor adherence with cART [OR 13.40 (95% C.I: 3.92-45.44)], and a longer duration of fever [OR 3.34 (95% C.I: 1.10-9.99)] being predictors of mortality. Part of the study's limitation was resource-constraint of some of the indigent patient which affected their ability to access some diagnostic investigations and get optimal care thereby impacting on their outcome. Despite the upscaling of cART, opportunistic infections and sepsis remain common causes of hospitalization and death in adult PLHIV. More attention should therefore be placed on early diagnosis, prevention of immunosuppression and sepsis through timely administration and adherence to cART and other prophylactic measures.
Collapse
Affiliation(s)
- Ayanfe Omololu
- Department of Internal Medicine, Federal Medical Centre, Abeokuta, Nigeria
| | - Asukwo Onukak
- Department of Internal Medicine, University of Uyo, Uyo, Nigeria
| | - Mfon Effiong
- Akwa Ibom State Hospital Management Board, Uyo, Nigeria
| | - Olaide Oke
- Department of Internal Medicine, Federal Medical Centre, Abeokuta, Nigeria
| | - Samson E. Isa
- Department of Medicine, University of Jos, Jos, Nigeria
| | - Abdulrazaq G. Habib
- Department of Medicine, College of Health Sciences, Bayero University, Kano, Nigeria
| |
Collapse
|
6
|
Yuliwulandari R, Prayuni K, Viyati K, Mahasirimongkol S, Wichukchinda N. Frequencies of HLA-B alleles in Indonesian Malay Ethnic. Heliyon 2024; 10:e26713. [PMID: 38439829 PMCID: PMC10909668 DOI: 10.1016/j.heliyon.2024.e26713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 02/06/2024] [Accepted: 02/19/2024] [Indexed: 03/06/2024] Open
Abstract
Background The HLA-B alleles have been used as a marker to predict drug-induced adverse reactions and as a major contributor to hypersensitivity reactions. We examined the feasibility of HLA-B alleles as pharmacogenomic markers of drug-induced hypersensitivity in an Indonesian Malay Ethnic. Methods Fifty-eight Indonesian individuals of Malay ethnicity were enrolled in this study. HLA-B alleles were determined using reverse sequence-specific oligonucleotide probe coupled with xMAP technology. Results HLA-B*15:02 (15.52%), HLA-B*35:05 (9.48%), and HLA-B*07:05 (7.76%) were frequent alleles in the Indonesian Malay ethnic populations. We discovered at least eight pharmacogenomics markers of drug-induced hypersensitivity: HLA-B*15:02, HLA-B*15:21, HLA-B*13:01, HLA-B*35:05, HLA-B*38:02, HLA-B*51:01, HLA-B*57:01, and HLA-B*58:01. HLA-B*15:02 was in the same serotype group with HLA-B*15:21, which is a B-75 serotype associated with genetic predisposition for carbamazepine-induced Stevens-Johnson syndrome and toxic epidermal necrolysis. The Indonesian population, represented by Malay, Javanese, and Sundanese ethnicities, was similar to South East Asian, Han Chinese, and Taiwanese populations based on HLA-B*15:02 frequency as the most common allele found in Malay ethnics. Conclusion We provided valuable information on the frequency of drug hypersensitivity-associated HLA-B alleles in Indonesian Malay ethnic population, which can improve treatment safety.
Collapse
Affiliation(s)
- Rika Yuliwulandari
- Department of Pharmacology, Faculty of Medicine, Universitas Pembangunan Nasional Veteran Jawa Timur, Jalan Rungkut Madya No. 1, Surabaya, 60294, Indonesia
| | - Kinasih Prayuni
- Genetic Research Center, YARSI Research Institute, YARSI University, Jakarta Pusat, Jl. Letjen Suprapto, Cempaka Putih, 10510, Indonesia
| | - Kencono Viyati
- Genetic Research Center, YARSI Research Institute, YARSI University, Jakarta Pusat, Jl. Letjen Suprapto, Cempaka Putih, 10510, Indonesia
- Department of Histology, Faculty of Medicine, YARSI University, Jakarta Pusat, Jl. Letjen Suprapto, Cempaka Putih, 10510, Indonesia
| | | | - Nuanjun Wichukchinda
- Department of Medical Sciences, Ministry of Public Health, Tivanond Road, Nonthaburi, 11000, Thailand
| |
Collapse
|
7
|
Ghosh T, Barman D, Show K, Lo R, Manna D, Ghosh T, Maiti DK. N-Heterocyclic Carbene-Catalyzed Facile Synthesis of Phthalidyl Sulfonohydrazones: Density Functional Theory Mechanistic Insights and Docking Interactions. ACS OMEGA 2024; 9:11510-11522. [PMID: 38496936 PMCID: PMC10938401 DOI: 10.1021/acsomega.3c08529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 01/26/2024] [Accepted: 02/13/2024] [Indexed: 03/19/2024]
Abstract
N-heterocyclic carbene catalysis reaction protocol is disclosed for the synthesis of phthalidyl sulfonohydrazones. A broad range of N-tosyl hydrazones react effectively with phthalaldehyde derivatives under open-air conditions, enabling the formation of a new C-N bond via an oxidative path. The reaction proceeds under mild reaction conditions with broad substrate scopes, wide functional group tolerance, and good to excellent yields. The mechanistic pathway is studied successfully using control experiments, competitive reactions, ESI-MS spectral analyses of the reaction mixture, and computational study by density functional theory. The potential use of one of the phthalidyl sulfonohydrazone derivatives as the inhibitor of β-ketoacyl acyl carrier protein synthase I of Escherichia coli is investigated using molecular docking.
Collapse
Affiliation(s)
- Tanmoy Ghosh
- Department
of Chemistry, University of Calcutta, Kolkata 700009, India
| | - Debabrata Barman
- Department
of Chemistry, University of Calcutta, Kolkata 700009, India
| | - Krishanu Show
- Department
of Chemistry, Malda College, Malda, West Bengal 732101, India
| | - Rabindranath Lo
- Institute
of Organic Chemistry and Biochemistry, Czech
Academy of Sciences, v.v.i., Flemingovo nám. 2, Prague 6, Praha 16610, Czech Republic
| | - Debashree Manna
- Institute
of Organic Chemistry and Biochemistry, Czech
Academy of Sciences, v.v.i., Flemingovo nám. 2, Prague 6, Praha 16610, Czech Republic
- Department
of Applied Chemistry, Maulana Abul Kalam
Azad University of Technology, Haringhata, West Bengal 741249, India
| | - Tapas Ghosh
- Department
of Chemistry, Jadavpur University, Kolkata 700032, India
| | - Dilip K. Maiti
- Department
of Chemistry, University of Calcutta, Kolkata 700009, India
| |
Collapse
|
8
|
Mathur S, Smuk M, Evans C, Wedderburn CJ, Gibb DM, Penazzato M, Prendergast AJ. Estimating the impact of alternative programmatic cotrimoxazole strategies on mortality among children born to mothers with HIV: A modelling study. PLoS Med 2024; 21:e1004334. [PMID: 38377150 PMCID: PMC10914273 DOI: 10.1371/journal.pmed.1004334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 03/05/2024] [Accepted: 01/10/2024] [Indexed: 02/22/2024] Open
Abstract
BACKGROUND World Health Organization (WHO) guidelines recommend cotrimoxazole prophylaxis for children who are HIV-exposed until infection is excluded and vertical transmission risk has ended. While cotrimoxazole has benefits for children with HIV, there is no mortality benefit for children who are HIV-exposed but uninfected, prompting a review of global guidelines. Here, we model the potential impact of alternative cotrimoxazole strategies on mortality in children who are HIV-exposed. METHODS AND FINDINGS Using a deterministic compartmental model, we estimated mortality in children who are HIV-exposed from 6 weeks to 2 years of age in 4 high-burden countries: Côte d'Ivoire, Mozambique, Uganda, and Zimbabwe. Vertical transmission rates, testing rates, and antiretroviral therapy (ART) uptake were derived from UNAIDS data, trial evidence, and meta-analyses. We explored 6 programmatic strategies: maintaining current recommendations; shorter cotrimoxazole provision for 3, 6, 9, or 12 months; and starting cotrimoxazole only for children diagnosed with HIV. Modelled alternatives to the current strategy increased mortality to varying degrees; countries with high vertical transmission had the greatest mortality. Compared to current recommendations, starting cotrimoxazole only after a positive HIV test had the greatest predicted increase in mortality: Mozambique (961 excess annual deaths; excess mortality 339 per 100,000 HIV-exposed children; risk ratio (RR) 1.06), Uganda (491; 221; RR 1.04), Zimbabwe (352; 260; RR 1.05), and Côte d'Ivoire (125; 322; RR 1.06). Similar effects were observed for 3-, 6-, 9-, and 12-month strategies. Increased mortality persisted but was attenuated when modelling lower cotrimoxazole uptake, smaller mortality benefits, higher testing coverage, and lower vertical transmission rates. The study is limited by uncertain estimates of cotrimoxazole coverage in programmatic settings; an inability to model increases in mortality arising from antimicrobial resistance due to limited surveillance data in sub-Saharan Africa; and lack of a formal health economic analysis. CONCLUSIONS Changing current guidelines from universal cotrimoxazole provision for children who are HIV-exposed increased predicted mortality across the 4 modelled high-burden countries, depending on test-to-treat cascade coverage and vertical transmission rates. These findings can help inform policymaker deliberations on cotrimoxazole strategies, recognising that the risks and benefits differ across settings.
Collapse
Affiliation(s)
- Shrey Mathur
- Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Melanie Smuk
- Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Ceri Evans
- Blizard Institute, Queen Mary University of London, London, United Kingdom
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| | - Catherine J. Wedderburn
- Medical Research Council Clinical Trials Unit at University College London, London, United Kingdom
- Department of Paediatrics and Child Health and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Diana M. Gibb
- Medical Research Council Clinical Trials Unit at University College London, London, United Kingdom
| | - Martina Penazzato
- Department of Research for Health, Science Division, World Health Organization, Geneva, Switzerland
| | - Andrew J. Prendergast
- Blizard Institute, Queen Mary University of London, London, United Kingdom
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| |
Collapse
|
9
|
Wedderburn CJ, Evans C, Slogrove AL, Rehman AM, Gibb DM, Prendergast AJ, Penazzato M. Co-trimoxazole prophylaxis for children who are HIV-exposed and uninfected: a systematic review. J Int AIDS Soc 2023; 26:e26079. [PMID: 37292018 PMCID: PMC10251133 DOI: 10.1002/jia2.26079] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 03/08/2023] [Indexed: 06/10/2023] Open
Abstract
INTRODUCTION Co-trimoxazole prophylaxis is recommended for children born to women with HIV to protect those who acquire HIV from opportunistic infections, severe bacterial infections and malaria. With scale-up of maternal antiretroviral therapy, most children remain HIV-exposed uninfected (HEU) and the benefits of universal co-trimoxazole are uncertain. We assessed the effect of co-trimoxazole on mortality and morbidity of children who are HEU. METHODS We performed a systematic review (PROSPERO number: CRD42021215059). We systematically searched MEDLINE, Embase, Cochrane CENTRAL, Global Health, CINAHL Plus, Africa-Wide Information, SciELO and WHO Global Index Medicus for peer-reviewed articles from inception to 4th January 2022 without limits. Ongoing randomized controlled trials (RCTs) were identified through registries. We included RCTs reporting mortality or morbidity in children who are HEU receiving co-trimoxazole versus no prophylaxis/placebo. The risk of bias was assessed using the Cochrane 2.0 tool. Data were summarized using narrative synthesis and findings were stratified by malaria endemicity. RESULTS We screened 1257 records and included seven reports from four RCTs. Two trials from Botswana and South Africa of 4067 children who are HEU found no difference in mortality or infectious morbidity in children randomized to co-trimoxazole prophylaxis started at 2-6 weeks of age compared to those randomized to placebo or no treatment, although event rates were low. Sub-studies found that antimicrobial resistance was higher in infants receiving co-trimoxazole. Two trials in Uganda investigating prolonged co-trimoxazole after breastfeeding cessation showed protection against malaria but no other morbidity or mortality differences. All trials had some concerns or a high risk of bias, which limited the certainty of evidence. DISCUSSION Studies show no clinical benefit of co-trimoxazole prophylaxis in children who are HEU, except to prevent malaria. Potential harms were identified for co-trimoxazole prophylaxis leading to antimicrobial resistance. The trials in non-malarial regions were conducted in populations with low mortality potentially reducing generalizability to other settings. CONCLUSIONS In low-mortality settings with few HIV transmissions and well-performing early infant diagnosis and treatment programmes, universal co-trimoxazole may not be required.
Collapse
Affiliation(s)
- Catherine J. Wedderburn
- Department of Paediatrics and Child Health and Neuroscience InstituteUniversity of Cape TownCape TownSouth Africa
- Medical Research Council Clinical Trials Unit at University College LondonLondonUK
- Department of Clinical ResearchLondon School of Hygiene & Tropical MedicineLondonUK
| | - Ceri Evans
- Blizard InstituteQueen Mary University of LondonLondonUK
- Zvitambo Institute for Maternal and Child Health ResearchHarareZimbabwe
- Department of Clinical InfectionMicrobiology and ImmunologyUniversity of LiverpoolLiverpoolUK
| | - Amy L. Slogrove
- Department of Paediatrics and Child HealthFaculty of Medicine & Health SciencesStellenbosch UniversityWorcesterSouth Africa
| | - Andrea M. Rehman
- MRC International Statistics & Epidemiology GroupDepartment of Infectious Disease EpidemiologyLondon School of Hygiene & Tropical MedicineLondonUK
| | - Diana M. Gibb
- Medical Research Council Clinical Trials Unit at University College LondonLondonUK
| | - Andrew J. Prendergast
- Blizard InstituteQueen Mary University of LondonLondonUK
- Zvitambo Institute for Maternal and Child Health ResearchHarareZimbabwe
| | - Martina Penazzato
- Department of Global HIVHepatitis and Sexually Transmitted Infections ProgrammesWorld Health OrganizationGenevaSwitzerland
| |
Collapse
|
10
|
Haindongo EH, Ndakolo D, Hedimbi M, Vainio O, Hakanen A, Vuopio J. Antimicrobial resistance prevalence of Escherichia coli and Staphylococcus aureus amongst bacteremic patients in Africa: a systematic review. J Glob Antimicrob Resist 2023; 32:35-43. [PMID: 36526264 DOI: 10.1016/j.jgar.2022.11.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVES Antimicrobial resistance (AMR) is a global concern among infectious diseases. Bloodstream infections can potentially become life-threatening if they become untreatable with conventional antimicrobials. This review aims to provide an understanding of the AMR prevalence and trends of common bacteremic pathogens, namely Escherichia coli and Staphylococcus aureus in the World Health Organization (WHO) Africa region. METHODS PubMed and Google Scholar were searched using relevant keywords for published human studies (excluding case reports and reviews) reporting bacteremic AMR data on the pathogens of interest between 2008 and 2019. Two reviewers independently screened the articles against a pre-defined eligibility criterion. Data extraction and analysis were achieved with different platforms: Covidence, Excel, R version 3.6.3, and QGIS v3.4.5. The pooled prevalence, 95% confidence intervals, and I2 index (a measure of heterogeneity) were calculated for the various pathogen-antibiotic combinations. RESULTS Five hundred sixty-two papers were retrieved, with 27 papers included in the final analysis. Only 23.4% (11/47) of member states of the WHO African region had reports on AMR in bacteremia. The Clinical and Laboratory Standards Institute (CLSI) (78.5%) was the most common standard used in the region. For E. coli, the pooled resistance was: cefotaxime (42%), imipenem (4%), meropenem (0%), and colistin (0%). For S. aureus, the calculated pooled resistance was cloxacillin (34%), oxacillin (12%), and vancomycin (0%). There was a high degree of variation across studies (I2 > 90%). CONCLUSION The pooled resistance rates indicate a concerning degree of methicillin-resistant and Extended Spectrum-ß-lactamase-producing pathogens. The paucity of AMR data also presents challenges for a comprehensive understanding of the situation in the region. Continent-wide and standardized surveillance efforts therefore need strengthening.
Collapse
Affiliation(s)
- Erastus Hanganeni Haindongo
- School of Medicine, Faculty of Health Sciences and Veterinary Medicine, University of Namibia, Windhoek, Namibia; Institute of Biomedicine, University of Turku, Turku, Finland.
| | - Diana Ndakolo
- School of Pharmacy, Faculty of Health Sciences, University of Namibia, Windhoek, Namibia; Pharmaceutical Services, Ministry of Health and Social Services, Namibia
| | - Marius Hedimbi
- School of Medicine, Faculty of Health Sciences and Veterinary Medicine, University of Namibia, Windhoek, Namibia; Graduate School of Business and Postgraduate, International University of Management, Namibia
| | - Olli Vainio
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Antti Hakanen
- Institute of Biomedicine, University of Turku, Turku, Finland; Clinical Microbiology Laboratory, Turku University Hospital, Turku, Finland
| | - Jaana Vuopio
- Institute of Biomedicine, University of Turku, Turku, Finland; Clinical Microbiology Laboratory, Turku University Hospital, Turku, Finland
| |
Collapse
|
11
|
Jacobo-Vargas TB, Báez-Saldaña R, Cruz-Hervert LP, Fortoul TI, Ahumada-Topete VH, Rodríguez-Ganén O, Vega-Barrientos RS. Trimethoprim-Sulfamethoxazole-associated early neutropenia in Mexican adults living with HIV: A cohort study. PLoS One 2023; 18:e0285541. [PMID: 37167312 PMCID: PMC10174569 DOI: 10.1371/journal.pone.0285541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 04/25/2023] [Indexed: 05/13/2023] Open
Abstract
INTRODUCTION Trimethoprim/sulfamethoxazole (TMP/SMX) is the antimicrobial of first choice in the treatment and prophylaxis of Pneumocystis jirovecii pneumonia (PCP) in immunocompromised patients, particularly in people living with human immunodeficiency virus (HIV). TMP/SMX use entails different adverse effects, and its association with early neutropenia is minimally documented. This study aimed to identify the risk of early neutropenia associated with TMP/SMX use in adults living with HIV in Mexico. METHODS A prospective cohort study was conducted in TMP/SMX-naïve adults living with HIV admitted to a third-level hospital between August 2019 and March 2020. Socio-demographic, clinical, and laboratory data were collected. According to patients' diagnostic, if they required treatment or prophylaxis against PCP, medical staff decided to prescribe TMP/SMX, as it is the first-line treatment. The risk of TMP/SMX induced early neutropenia, as well as associated factors were analyzed through a bivariate model and a multivariate Poisson regression model. The strength of association was measured by incidence rate ratio (IRR) with 95% confidence interval. RESULTS 57 patients were enrolled in the study, of whom 40 patients were in the TMP/SMX treatment-group for treatment or prophylaxis of PCP (204.8 person-years of observation, median 26.5 days) and 17 patients were in the non-treatment group because they did not need the drug for treatment or prophylaxis of PCP (87.0 person-years of observation, median 21 days). The incidence rate of early neutropenia in the TMP/SMX-treatment group versus non-treatment group was 7.81 and 1.15 cases per 100 person-years, respectively. After adjusting for stage 3 of HIV infection and neutrophil count <1,500 cells/mm3 at hospital admission, the current use of TMP/SMX was not associated with an increase in the incidence rate ratio of early neutropenia (adjusted IRR: 3.46; 95% CI: 0.25-47.55; p = 0.352). CONCLUSIONS The current use of TMP/SMX in Mexican adults living with HIV was not associated with an increase in the incidence rate ratio of early neutropenia.
Collapse
Affiliation(s)
- Thalia Berenice Jacobo-Vargas
- Pharmacist in Pharmacology Research Unit and Hospital Pharmacy Department, National Institute of Respiratory Diseases, Mexico City, Mexico
- Programa de Maestría y Doctorado en Ciencias Médicas, Odontológicas y de la Salud, National Autonomous University of Mexico, Mexico City, Mexico
| | - Renata Báez-Saldaña
- Hospitalization, National Institute of Respiratory Diseases, Mexico City, Mexico
| | - Luis Pablo Cruz-Hervert
- Professor of Orthodontic Department in the Division of Postgraduate Studies and Research, Faculty of Dentistry, National Autonomous University of Mexico, Mexico City, Mexico
- Program on Epidemiological and Emerging Risks, National Autonomous University of Mexico, Mexico City, Mexico
| | | | - Victor Hugo Ahumada-Topete
- Head of the Unit of Hospital Epidemiology and Infectious Diseases, National Institute of Respiratory Diseases, Mexico City, Mexico
| | - Odalis Rodríguez-Ganén
- Head of the Hospital Pharmacy Department, National Institute of Respiratory Diseases, Mexico City, Mexico
| | | |
Collapse
|
12
|
Chen J, Chen R, Wei W, Qin F, Chen X, He J, Zhang H, Wang G, Shi M, Qin T, Liao Y, Wu Y, Lu B, Tao X, Ye L, Liang H, Jiang J. Effect and Mechanism of Cotrimoxazole Against Talaromyces marneffei in vitro. Mycopathologia 2022; 187:579-593. [DOI: 10.1007/s11046-022-00673-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 09/18/2022] [Indexed: 11/21/2022]
|
13
|
Nakkam N, Saksit N, Konyoung P, Amornpinyo W, Khunarkornsiri U, Purimart D, Pattanacheewapull O, Naewla T, Wattanachai P, Khaeso K, Chumworathayi P, Tassaneeyakul W. Associations of HLA and drug-metabolizing enzyme genes in co-trimoxazole-induced severe cutaneous adverse reactions. Drug Metab Pharmacokinet 2022; 47:100480. [DOI: 10.1016/j.dmpk.2022.100480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 10/07/2022] [Accepted: 10/13/2022] [Indexed: 11/03/2022]
|
14
|
Daniels B, Kuhn L, Spooner E, Mulol H, Goga A, Feucht U, Essack SY, Coutsoudis A. Cotrimoxazole guidelines for infants who are HIV-exposed but uninfected: a call for a public health and ethics approach to the evidence. THE LANCET GLOBAL HEALTH 2022; 10:e1198-e1203. [DOI: 10.1016/s2214-109x(22)00120-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 03/04/2022] [Accepted: 03/11/2022] [Indexed: 10/17/2022] Open
|
15
|
Microbiological Characterisation of Community-Acquired Urinary Tract Infections in Bagamoyo, Tanzania: A Prospective Study. Trop Med Infect Dis 2022; 7:tropicalmed7060100. [PMID: 35736979 PMCID: PMC9228238 DOI: 10.3390/tropicalmed7060100] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/06/2022] [Accepted: 06/08/2022] [Indexed: 02/05/2023] Open
Abstract
Urinary tract infections (UTIs) are among the most common infections in sub-Saharan Africa, but microbiological data to guide treatment decisions are limited. Hence, we investigated the bacterial aetiology and corresponding antimicrobial susceptibility patterns in outpatients with UTIs in Bagamoyo, Tanzania. Urine samples from symptomatic individuals were subjected to microbiological examinations for bacterial species identification using conventional methods and disc diffusion-based resistance testing. Subsequently, urine samples were transferred to Germany for confirmatory diagnostics using matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry and automated resistance testing. Overall, 104 out of 270 (38.5%) individuals had a positive urine culture and 119 putative pathogens were identified. The most frequently detected bacteria were Escherichia coli (23%), Klebsiella spp. (7%), Enterobacter cloacae complex (3%) and Staphylococcus aureus (2%). E. coli isolates showed high resistance against cotrimoxazole (76%), ampicillin (74%), piperacillin (74%) and fluoroquinolones (37%), but widespread susceptibility to meropenem (100%), fosfomycin (98%), piperacillin/tazobactam (97%) and amoxicillin/clavulanic acid (82%). The agreement between E. coli susceptibility testing results in Tanzania and Germany was ≥95%, except for piperacillin/tazobactam (89%) and ciprofloxacin (84%). Given the considerable resistance to frequently prescribed antibiotics, such as cotrimoxazole and fluoroquinolones, future research should explore the potential of oral alternatives (e.g., fosfomycin) for the treatment of UTIs in Tanzania.
Collapse
|
16
|
Do QTT, Otaki M, Otaki Y, Tushara C, Sanjeewa IW. Pharmaceutical Contaminants in Shallow Groundwater and Their Implication for Poor Sanitation Facilities in Low-Income Countries. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2022; 41:266-274. [PMID: 33978268 DOI: 10.1002/etc.5110] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/31/2021] [Accepted: 05/03/2021] [Indexed: 06/12/2023]
Abstract
Rudimentary on-site sanitation systems (OSSs) are extensively used in low-income countries despite the risk of groundwater contamination. The present study investigated the potential impact of community soak pits on sandy shallow aquifers in a rural area in Sri Lanka. Thirty-two groundwater samples were collected and measured for the traditional indicators fecal bacteria Escherichia coli, total coliforms, nitrate, and chloride; 7 pharmaceuticals were added as wastewater indicators. Analysis showed that the local groundwater is infected by fecal bacteria, but it was unable to determine whether the source of fecal bacteria is the OSSs. Similarly, nitrate and chloride provided unclear evidence. Conversely, detection of 3 pharmaceuticals at trace levels in groundwater evidenced wastewater impact from OSSs. Caffeine was detected in 89% of the sample at a maximum concentration 7.9 ng/L, indicating fecal pollution. Carbamazepine was detected in 42% of the samples with a concentration of up to 6.9 ng/L, whereas sulfamethoxazole was detected in only 2 samples. The presence of carbamazepine and sulfamethoxazole was also consistent with recorded drug use of the residents. Escherichia coli showed a moderate positive correlation with caffeine concentration (Kendall's τ = 0.38, p = 0.017), indicating concurring short-lifetime fecal bacteria and labile wastewater organic compounds. Nitrate showed a significant correlation with carbamazepine concentration (τ = 0.39, p = 0.016). Fecal bacteria and nitrate can be used in screening for micropollutants in domestic wells impacted by OSSs. The present case study emphasizes the potential impact of poor sanitation on groundwater resources and the necessity of sanitation improvement in achieving Sustainable Development Goals in developing countries. Environ Toxicol Chem 2022;41:266-274. © 2021 SETAC.
Collapse
Affiliation(s)
- Quyen Thi Thuy Do
- Department of Human Centered Engineering, Ochanomizu University, Tokyo, Japan
| | - Masahiro Otaki
- Faculty of Core Research, Natural Science Division, Ochanomizu University, Tokyo, Japan
| | - Yurina Otaki
- Graduate School of Social Sciences, Hitotsubashi University, Tokyo, Japan
| | - Chaminda Tushara
- Faculty of Engineering, University of Ruhuna, Hapugala, Galle, Sri Lanka
| | | |
Collapse
|
17
|
Safari D, Putri HFM, Bimantari A, Paramaiswari WT, Tafroji W, Khoeri MM, Salsabila K. Genetic characterization of co-trimoxazole non-susceptible Streptococcus pneumoniae isolates from Indonesia. Access Microbiol 2021; 3:000271. [PMID: 34816091 PMCID: PMC8604174 DOI: 10.1099/acmi.0.000271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 08/24/2021] [Indexed: 11/18/2022] Open
Abstract
We investigated the genetic variation of folA and folP genes encoding dihydropteroate synthase (DHPS) and dihydrofolate reductase (DHFR) enzymes amongst trimethoprim/sulfamethoxazole (co-trimoxazole) non-susceptible Streptococcus pneumoniae isolated from the Indonesian population. Archived S. pneumoniae isolates were screened for the presence and analysis of folA and folP genes using the polymerase chain reaction sequencing method. We found that 80 % of co-trimoxazole non-susceptible isolates (n=30/39) showed a 6 bp insertion in the sulphonamide-binding site of DHPS. The Asp-92-Ala and Ile-100-Leu substitutions were more common on DHFR (42 %; 22/53) followed by the Asp-92–Ala, Glu-94–Asp and Ile-100–Leu substitutions (32 %; 17/53). The combination of the Ile-100–Leu substitution at the DHFR region and the 6 bp insertion was the most dominant combination among isolates having both folA and folP genes.
Collapse
Affiliation(s)
- Dodi Safari
- Eijkman Institute for Molecular Biology, Jakarta, Indonesia
- *Correspondence: Dodi Safari,
| | - Hanifah Fajri Maharani Putri
- Eijkman Institute for Molecular Biology, Jakarta, Indonesia
- Biology Program, Surya University, Tangerang, Indonesia
| | | | | | - Wisnu Tafroji
- Eijkman Institute for Molecular Biology, Jakarta, Indonesia
| | | | | |
Collapse
|
18
|
Abongomera G, Koller M, Musaazi J, Lamorde M, Kaelin M, Tasimwa HB, Eberhard N, Hongler J, Haller S, Kambugu A, Castelnuovo B, Fehr J. Spectrum of antibiotic resistance in UTI caused by Escherichia coli among HIV-infected patients in Uganda: a cross-sectional study. BMC Infect Dis 2021; 21:1179. [PMID: 34814849 PMCID: PMC8609806 DOI: 10.1186/s12879-021-06865-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 11/12/2021] [Indexed: 11/28/2022] Open
Abstract
Background Antimicrobial drug resistance is one of the top ten threats to global health according to the World Health Organization. Urinary tract infections (UTIs) are among the most common bacterial infections and main reason for antibiotic prescription. The incidence of UTIs appears to be high among people living with HIV. We sought to determine the most common UTI pathogens among HIV infected patients and evaluate their susceptibility towards antibiotics. Methods We performed a cross-sectional study among HIV-infected patients aged ≥ 18 years presenting at an HIV care specialized clinic with symptoms suggestive of a urethritis. Urine cultures were subjected to antibiotic susceptibility testing according to Clinical Laboratory Standards Institute. The data was analyzed using STATA, we performed Pearson’s Chi-square and Fisher’s exact tests to compare differences between proportions. Results Out of the 200 patients, 123 (62%) were female. The median age was 41.9 years (IQR 34.7–49.3). Only 32 (16%) urine cultures showed bacterial growth. Escherichia coli was the most commonly isolated uropathogen (72%), followed by Klebsiella pneumoniae (9%). E. coli was completely resistant to cotrimoxazole and ampicillin; resistance to ciprofloxacin and ceftriaxone was 44% and 35% respectively; 9% to gentamicin; no resistance detected to nitrofurantoin and imipenem. Conclusions Our findings are congruent with the Uganda national clinical guidelines which recommends nitrofurantoin as the first line antibiotic for uncomplicated UTI. Significant ciprofloxacin and ceftriaxone resistance was detected. In the era of emerging antibiotic resistance, understanding the local susceptibilities among sub-populations such as HIV infected patients is crucial. Further investigation is needed to address reasons for the low bacterial growth rate observed in the urine cultures. Supplementary Information The online version contains supplementary material available at 10.1186/s12879-021-06865-3.
Collapse
Affiliation(s)
- George Abongomera
- Department of Public and Global Health, Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Hirschengraben 84, CH 8001, Zurich, Switzerland.
| | - Maurice Koller
- Department of Public and Global Health, Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Hirschengraben 84, CH 8001, Zurich, Switzerland
| | - Joseph Musaazi
- Makerere University Infectious Diseases Institute, Kampala, Uganda
| | - Mohammed Lamorde
- Makerere University Infectious Diseases Institute, Kampala, Uganda
| | - Marisa Kaelin
- University Hospital Zurich, Infectious Diseases and Hospital Epidemiology, Zürich, Switzerland
| | | | - Nadia Eberhard
- University Hospital Zurich, Infectious Diseases and Hospital Epidemiology, Zürich, Switzerland
| | - Jan Hongler
- Department of Public and Global Health, Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Hirschengraben 84, CH 8001, Zurich, Switzerland
| | - Sabine Haller
- Department of Public and Global Health, Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Hirschengraben 84, CH 8001, Zurich, Switzerland
| | - Andrew Kambugu
- Makerere University Infectious Diseases Institute, Kampala, Uganda
| | | | - Jan Fehr
- Epidemiology, Biostatistics and Prevention Institute, University of Zurich and University Hospital Zurich, Infectious Diseases and Hospital Epidemiology, Zürich, Switzerland
| |
Collapse
|
19
|
Peeters D, van Geloven N, Visser LE, Bogaert D, van Rossum AMC, Driessen GJA, Verhagen LM. Study protocol for a randomised controlled trial evaluating the clinical effect of antibiotic prophylaxis in children with recurrent respiratory tract infections: the Approach study. BMJ Open 2021; 11:e044505. [PMID: 34326043 PMCID: PMC8323378 DOI: 10.1136/bmjopen-2020-044505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
INTRODUCTION Respiratory tract infections (RTIs) affect children all over the world and are associated with significant morbidity and mortality. In particular, recurrent RTIs cause a high burden of disease and lead to frequent doctor visits. Children with recurrent RTIs generally have no significant alterations or deficits in systemic immunity. In an attempt to treat the assumed bacterial component involved, they are often treated with prolonged courses of prophylactic antibiotics taken on a daily basis. Despite its common use, there is no evidence that this is beneficial. Studies assessing the clinical effectiveness of antibiotic prophylaxis as well as potential adverse effects and antibiotic resistance development, are therefore urgently needed. METHODS AND ANALYSIS We present a protocol for a randomised double-blind placebo-controlled trial comparing co-trimoxazole with placebo treatment in children with recurrent RTIs. A total of 158 children (aged 6 months-10 years) with recurrent RTIs without significant comorbidity will be enrolled from a minimum of 10 Dutch hospitals. One group receives co-trimoxazole 18 mg/kg two times per day (36 mg/kg/day) and the other group receives a placebo two times per day for a period of 3 months. The main objective is to determine whether antibiotic prophylaxis is more effective than placebo to prevent/reduce respiratory symptoms in children with recurrent RTIs. Respiratory symptoms will be scored by parents on a daily basis in both study arms by the use of a mobile phone application. Our primary outcome will be the number of days with at least two respiratory symptoms during the treatment. ETHICS AND DISSEMINATION Ethics approval was obtained from the Medical Ethics Research Committee Zuidwest Holland/LDD. A manuscript with the study results will be submitted to a peer-reviewed journal. All participants will be informed about the study results. The results of the study will inform clinical guidelines regarding the prophylactic treatment of children with recurrent RTIs. TRIAL REGISTRATION NUMBER NL7044.
Collapse
Affiliation(s)
- Daphne Peeters
- Department of Paediatrics, Haga Hospital, Juliana Childrens Hospital, Den Haag, Zuid-Holland, The Netherlands
| | - Nan van Geloven
- Department of Biomedical Data Sciences, Medical Statistics, Leiden University Medical Center, Leiden, The Netherlands
| | - Loes E Visser
- Department of Hospital Pharmacy, Haga Teaching Hospital, Den Haag, The Netherlands
- Department of Hospital Pharmacy and Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Debby Bogaert
- Center for Inflammation Research, Queen Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Department of Paediatric Infectious Diseases and Immunology, University Medical Center Utrecht, Wilhelmina Children's Hospital, Utrecht, The Netherlands
| | | | - Gertjan J A Driessen
- Department of Paediatrics, Haga Hospital, Juliana Childrens Hospital, Den Haag, Zuid-Holland, The Netherlands
- Department of Paediatrics, Maastricht UMC+, Maastricht, The Netherlands
| | - Lilly M Verhagen
- Department of Paediatric Infectious Diseases and Immunology, University Medical Center Utrecht, Wilhelmina Children's Hospital, Utrecht, The Netherlands
- Section Paediatric Infectious Diseases, Laboratory of Medical Immunology, Radboud Center for Infectious Diseases, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
- Department of Paediatric Infectious Diseases and Immunology, Radboud University Medical Center, Amalia Children's Hospital, Nijmegen, The Netherlands
| |
Collapse
|
20
|
Evaluation of the usefulness of intermittent preventive treatment of malaria in pregnancy with sulfadoxine-pyrimethamine in a context with increased resistance of Plasmodium falciparum in Kingasani Hospital, Kinshasa in the Democratic Republic of Congo. INFECTION GENETICS AND EVOLUTION 2021; 94:105009. [PMID: 34284138 DOI: 10.1016/j.meegid.2021.105009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 11/21/2022]
Abstract
BACKGROUND Increasing resistance of Plasmodium falciparum to sulfadoxine-pyrimethamine (SP) threatens its usefulness for intermittent preventive treatment in pregnancy (IPTp-SP). The prophylactic effects of IPTp-SP on maternal malaria and adverse pregnancy outcomes were evaluated in Kingasani Hospital, Kinshasa in the Democratic Republic of Congo (DRC). METHODS Laboring women (n = 844) and respective newborns were investigated. Blood samples collected from women were tested for malaria using rapid diagnostic test (RDT), blood smears examination, and real-time PCR. The hemoglobin level was measured by HemoCue© analyzer. A PCR-RFLP method was applied for detecting N51I, C59R, and S108N mutations on dhfr along with A437G and K540E mutations on dhps in P. falciparum positive samples. Logistic regression models assessed relationships between IPTp-SP uptake and pregnancy outcomes. RESULTS P. falciparum malaria was detected at delivery in 10.8% of women and was statistically associated with fever during the pregnancy (OR = 2.9 [1.5; 6.3]; p = 0.004) and maternal anemia (OR = 3.9 [2.4; 6.3]; p < 0.001). One out of five parasites was a quintuple mutant encoding dhfr mutations 51I, 59R, and 108 N along with dhps mutations 437G and 540E. The molecular profile of parasites (i.e., 32.6% of parasites carrying dhps K540E) was suitable with continued use of SP for IPTp. IPTp-SP uptake was not associated with reduced maternal malaria, fever reported in pregnancy, or fetal deaths (p > 0.05). Conversely, three or more doses of SP were associated with reduced maternal anemia at delivery (OR = 0.4 [0.2; 0.9]; p = 0.024), shortened gestation (OR = 0.4 [0.2; 0.8]; p = 0.009), and low-birth weights (OR = 0.2 [0.1; 0.5]; p < 0.001). CONCLUSION IPTp-SP was not associated with reduced maternal malaria in our study, but evidence was found of a prophylactic effect against adverse pregnancy outcomes. To counteract further loss of clinical effects of IPTp-SP in the study population, alternative strategies able to improve its anti-malarial efficacy such as combination of SP with partner molecules should be implemented.
Collapse
|
21
|
Brief Report: Cessation of Long-Term Cotrimoxazole Prophylaxis in HIV-Infected Children Does Not Alter the Carriage of Antimicrobial Resistance Genes. J Acquir Immune Defic Syndr 2021; 85:601-605. [PMID: 32852361 PMCID: PMC7654951 DOI: 10.1097/qai.0000000000002489] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Cotrimoxazole (CTX) is a broad-spectrum antimicrobial, combining trimethoprim and sulfamethoxazole. CTX prophylaxis reduces mortality and morbidity among people living with HIV in regions with high prevalence of bacterial infections and malaria. The Antiretroviral research for Watoto trial evaluated the effect of stopping versus continuing CTX prophylaxis in sub-Saharan Africa. METHODS In this study, 72 HIV-infected Zimbabwean children, on antiretroviral therapy, provided fecal samples at 84 and 96 weeks after randomization to continue or stop CTX. DNA was extracted for whole metagenome shotgun sequencing, with sequencing reads mapped to the Comprehensive Antibiotic Resistance Database to identify CTX and other antimicrobial resistance genes. RESULTS There were minimal differences in the carriage of CTX resistance genes between groups. The dfrA1 gene, conferring trimethoprim resistance, was significantly higher in the continue group (P = 0.039) and the tetA(P) gene conferring resistance to tetracycline was significantly higher in the stop group (P = 0.013). CTX prophylaxis has a role in shaping the resistome; however, stopping prophylaxis does not decrease resistance gene abundance. CONCLUSIONS No differences were observed in resistance gene carriage between the stop and continue groups. The previously shown multi-faceted protective effects of CTX in antiretroviral research for Watoto trial clinical outcomes are not outweighed by the risk of multi-drug resistance gene selection due to prophylaxis. These findings are reassuring, given current recommendations for long-term CTX prophylaxis among children living with HIV in sub-Saharan Africa to decrease mortality and morbidity.
Collapse
|
22
|
Noble CCA, Sturgeon JP, Bwakura-Dangarembizi M, Kelly P, Amadi B, Prendergast AJ. Postdischarge interventions for children hospitalized with severe acute malnutrition: a systematic review and meta-analysis. Am J Clin Nutr 2021; 113:574-585. [PMID: 33517377 PMCID: PMC7948836 DOI: 10.1093/ajcn/nqaa359] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 11/06/2020] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Children hospitalized with severe acute malnutrition (SAM) have poor long-term outcomes following discharge, with high rates of mortality, morbidity, and impaired neurodevelopment. There is currently minimal guidance on how to support children with SAM following discharge from inpatient treatment. OBJECTIVES This systematic review and meta-analysis aimed to examine whether postdischarge interventions can improve outcomes in children recovering from complicated SAM. METHODS Systematic searches of 4 databases were undertaken to identify studies of interventions delivered completely or partially after hospital discharge in children aged 6-59 mo, following inpatient treatment of SAM. The main outcome of interest was mortality. Random-effects meta-analysis was undertaken where ≥2 studies were sufficiently similar in intervention and outcome. RESULTS Ten studies fulfilled the inclusion criteria, recruiting 39-1781 participants in 7 countries between 1975 and 2015. Studies evaluated provision of zinc (2 studies), probiotics or synbiotics (2 studies), antibiotics (1 study), pancreatic enzymes (1 study), and psychosocial stimulation (4 studies). Six studies had unclear or high risk of bias in ≥2 domains. Compared with standard care, pancreatic enzyme supplementation reduced inpatient mortality (37.8% compared with 18.6%, P < 0.05). In meta-analysis there was some evidence that prebiotics or synbiotics reduced mortality (RR: 0.72; 95% CI: 0.51, 1.00; P = 0.049). Psychosocial stimulation reduced mortality in meta-analysis of the 2 trials reporting deaths (RR: 0.36; 95% CI: 0.15, 0.87), and improved neurodevelopmental scores in ≥1 domain in all studies. There was no evidence that zinc reduced mortality in the single study reporting deaths. Antibiotics reduced infectious morbidity but did not reduce mortality. CONCLUSIONS Several biological and psychosocial interventions show promise in improving outcomes in children following hospitalization for SAM and require further exploration in larger randomized mortality trials. This study was registered with PROSPERO as CRD42018111342 (https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=111342).
Collapse
Affiliation(s)
- Christie C A Noble
- Blizard Institute, Queen Mary University of London, London, United Kingdom
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Jonathan P Sturgeon
- Blizard Institute, Queen Mary University of London, London, United Kingdom
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Mutsa Bwakura-Dangarembizi
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- University of Zimbabwe College of Health Sciences, Harare, Zimbabwe
| | - Paul Kelly
- Blizard Institute, Queen Mary University of London, London, United Kingdom
- Tropical Gastroenterology and Nutrition Group, University of Zambia, Lusaka, Zambia
| | - Beatrice Amadi
- Tropical Gastroenterology and Nutrition Group, University of Zambia, Lusaka, Zambia
| | - Andrew J Prendergast
- Blizard Institute, Queen Mary University of London, London, United Kingdom
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| |
Collapse
|
23
|
Bourke CD, Prendergast AJ. The Anti-inflammatory Effects of Cotrimoxazole Prophylaxis for People Living With Human Immunodeficiency Virus in Sub-Saharan Africa. J Infect Dis 2021; 222:347-350. [PMID: 31714953 DOI: 10.1093/infdis/jiz495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 10/30/2019] [Indexed: 11/13/2022] Open
Affiliation(s)
- Claire D Bourke
- Blizard Institute, Queen Mary University of London, London, United Kingdom.,Zviambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Andrew J Prendergast
- Blizard Institute, Queen Mary University of London, London, United Kingdom.,Zviambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| |
Collapse
|
24
|
Dirajlal-Fargo S, El-Kamari V, Weiner L, Shan L, Sattar A, Kulkarni M, Funderburg N, Nazzinda R, Karungi C, Kityo C, Musiime V, McComsey GA. Altered Intestinal Permeability and Fungal Translocation in Ugandan Children With Human Immunodeficiency Virus. Clin Infect Dis 2021; 70:2413-2422. [PMID: 31260509 DOI: 10.1093/cid/ciz561] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 06/28/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Children with perinatally acquired human immunodeficiency virus (HIV; PHIVs) face a lifelong cumulative exposure to HIV and antiretroviral therapy (ART). The relationship between gut integrity, microbial translocation, and inflammation in PHIV is poorly understood. METHODS This is a cross-sectional study in 57 PHIVs, 59 HIV-exposed but uninfected children, and 56 HIV-unexposed and -uninfected children aged 2-10 years old in Uganda. PHIVs were on stable ART with HIV-1 RNA <400 copies/mL. We measured markers of systemic inflammation, monocyte activation, and gut integrity. Kruskal-Wallis tests were used to compare markers by group and the Spearman correlation was used to assess correlations between biomarkers. RESULTS The mean age of all participants was 7 years and 55% were girls. Among PHIVs, the mean CD4 % was 34%, 93% had a viral load ≤20 copies/mL, and 79% were on a nonnucleoside reverse transcriptase inhibitor regimen. Soluble cluster of differentiation 14 (sCD14), beta-D-glucan (BDG), and zonulin were higher in the PHIV group (P ≤ .01). Intestinal fatty acid binding protein (I-FABP) and lipopolysaccharide binding protein (LBP) did not differ between groups (P > .05). Among PHIVs who were breastfed, levels of sCD163 and interleukin 6 (IL6) were higher than levels in PHIV who were not breastfed (P < .05). Additionally, in PHIVs with a history of breastfeeding, sCD14, BDG, LBP, zonulin, and I-FABP correlated with several markers of systemic inflammation, including high-sensitivity C-reactive protein, IL6, d-dimer, and systemic tumor necrosis factor receptors I and II (P ≤ .05). CONCLUSIONS Despite viral suppression, PHIVs have evidence of altered gut permeability and fungal translocation. Intestinal damage and the resultant bacterial and fungal translocations in PHIVs may play a role in the persistent inflammation that leads to many end-organ diseases in adults.Despite viral suppression, children with perinatally acquired human immunodeficiency virus (HIV) in Uganda have evidence of alterations in intestinal permeability and fungal translocation, compared to HIV-exposed but uninfected and HIV-unexposed children, which may play a role in HIV-associated chronic inflammation.
Collapse
Affiliation(s)
- Sahera Dirajlal-Fargo
- University Hospitals Cleveland Medical Center, Columbus.,Rainbow Babies and Children's Hospital, Columbus.,Case Western Reserve University, Columbus
| | | | | | | | | | - Manjusha Kulkarni
- Ohio State University School of Health and Rehabilitation Sciences, Columbus
| | - Nicholas Funderburg
- Ohio State University School of Health and Rehabilitation Sciences, Columbus
| | | | | | - Cissy Kityo
- Joint Clinical Research Centre, Kampala, Uganda
| | - Victor Musiime
- Joint Clinical Research Centre, Kampala, Uganda.,Department of Paediatrics and Child Health, Makerere University, Kampala, Uganda
| | - Grace A McComsey
- University Hospitals Cleveland Medical Center, Columbus.,Rainbow Babies and Children's Hospital, Columbus.,Case Western Reserve University, Columbus
| |
Collapse
|
25
|
Bourke CD, Evans C. Cotrimoxazole Prophylaxis Selects for Antimicrobial Resistance in Human Immunodeficiency Virus-Exposed, Uninfected Infants. Clin Infect Dis 2020; 71:2869-2871. [PMID: 31832637 PMCID: PMC7778347 DOI: 10.1093/cid/ciz1193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 12/11/2019] [Indexed: 01/01/2023] Open
Affiliation(s)
- Claire D Bourke
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London, United Kingdom
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Ceri Evans
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London, United Kingdom
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| |
Collapse
|
26
|
Hobbs CV, Sahu T, Neal J, Conteh S, Voza T, Borkowsky W, Langhorne J, Duffy PE. Determinants of Malaria Protective Immunity in Mice Immunized with Live Sporozoites during Trimethoprim-Sulfamethoxazole Prophylaxis. Am J Trop Med Hyg 2020; 104:666-670. [PMID: 33350377 PMCID: PMC7866335 DOI: 10.4269/ajtmh.20-0749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 10/09/2020] [Indexed: 11/21/2022] Open
Abstract
HIV and malaria geographically overlap. Trimethoprim–sulfamethoxazole (TMP-SMX) is a drug widely used in HIV-exposed uninfected and infected children in malaria-endemic areas, and is known to have antimalarial effects. Further study in terms of antimalarial impact and effect on development of malaria-specific immunity is therefore essential. Using rodent malaria models, we previously showed that repeated Plasmodium exposure during TMP-SMX administration, or chemoprophylaxis vaccination (CVac), induces CD8 T-cell–dependent preerythrocytic immunity. However, humoral immune responses have been shown to be important in models of preerythrocytic immunity. Herein, we demonstrate that antibody-mediated responses contribute to protective immunity induced by CVac immune sera using TMP-SMX in models of homologous, but not heterologous, parasite species. Clinical studies must account for potential anti-Plasmodium antibody induced during TMP-SMX prophylaxis.
Collapse
Affiliation(s)
- Charlotte V Hobbs
- Department of Microbiology, Batson Children's Hospital, University of Mississippi Medical Center, Jackson, Mississippi.,Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland.,Division of Infectious Diseases, Department of Pediatrics, Batson Children's Hospital, University of Mississippi Medical Center, Jackson, Mississippi
| | - Tejram Sahu
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland.,Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Jillian Neal
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Solomon Conteh
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Tatiana Voza
- Biological Sciences Department, New York City College of Technology, CUNY, New York, New York
| | - William Borkowsky
- Division of Infectious Disease and Immunology, Department of Pediatrics, New York University School of Medicine, New York, New York
| | | | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
27
|
Smits BM, Kleine Budde I, de Vries E, Ten Berge IJM, Bredius RGM, van Deuren M, van Dissel JT, Ellerbroek PM, van der Flier M, van Hagen PM, Nieuwhof C, Rutgers B, Sanders LEAM, Simon A, Kuijpers TW, van Montfrans JM. Immunoglobulin Replacement Therapy Versus Antibiotic Prophylaxis as Treatment for Incomplete Primary Antibody Deficiency. J Clin Immunol 2020; 41:382-392. [PMID: 33206257 PMCID: PMC7858555 DOI: 10.1007/s10875-020-00841-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 08/01/2020] [Indexed: 11/18/2022]
Abstract
Background Patients with an IgG subclass deficiency (IgSD) ± specific polysaccharide antibody deficiency (SPAD) often present with recurrent infections. Previous retrospective studies have shown that prophylactic antibiotics (PA) and immunoglobulin replacement therapy (IRT) can both be effective in preventing these infections; however, this has not been confirmed in a prospective study. Objective To compare the efficacy of PA and IRT in a randomized crossover trial. Methods A total of 64 patients (55 adults and 9 children) were randomized (2:2) between two treatment arms. Treatment arm A began with 12 months of PA, and treatment arm B began with 12 months of IRT. After a 3-month bridging period with cotrimoxazole, the treatment was switched to 12 months of IRT and PA, respectively. The efficacy (measured by the incidence of infections) and proportion of related adverse events in the two arms were compared. Results The overall efficacy of the two regimens did not differ (p = 0.58, two-sided Wilcoxon signed-rank test). A smaller proportion of patients suffered a related adverse event while using PA (26.8% vs. 60.3%, p < 0.0003, chi-squared test). Patients with persistent infections while using PA suffered fewer infections per year after switching to IRT (2.63 vs. 0.64, p < 0.01). Conclusion We found comparable efficacy of IRT and PA in patients with IgSD ± SPAD. Patients with persistent infections during treatment with PA had less infections after switching to IRT. Clinical Implication Given the costs and associated side-effects of IRT, it should be reserved for patients with persistent infections despite treatment with PA. Electronic supplementary material The online version of this article (10.1007/s10875-020-00841-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bas M Smits
- Department of Pediatric Immunology and Infectious Diseases, UMC Utrecht, Lundlaan 6, 3584 EA, Utrecht, The Netherlands
| | - Ilona Kleine Budde
- Clinical Operations, Sanquin Plasma Products B.V, Amsterdam, The Netherlands
| | - Esther de Vries
- Department of Tranzo, Tilburg School of Social and Behavioral Sciences, Tilburg University, Tilburg, The Netherlands.,Department of Jeroen Bosch Academy Research, Jeroen Bosch Hospital, 's-Hertogenbosch, The Netherlands
| | - Ineke J M Ten Berge
- Department of Internal Medicine, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, The Netherlands
| | - Robbert G M Bredius
- Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Marcel van Deuren
- Department of Internal Medicine, Radboud UMC, Nijmegen, The Netherlands
| | - Jaap T van Dissel
- Department of Infectious Diseases, Leiden University Medical Centre, University of Leiden, Leiden, The Netherlands
| | - Pauline M Ellerbroek
- Division of Internal Medicine and Dermatology, Department of Infectious Diseases, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Michiel van der Flier
- Department of Pediatric Immunology and Infectious Diseases, UMC Utrecht, Lundlaan 6, 3584 EA, Utrecht, The Netherlands.,Pediatric Infectious Diseases and Immunology, Radboudumc Amalia Children's Hospital, Nijmegen, The Netherlands
| | - P Martin van Hagen
- Department of Internal Medicine/Immunology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Chris Nieuwhof
- Department of Allergology and Clinical Immunology, Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands
| | - Bram Rutgers
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Lieke E A M Sanders
- Department of Pediatric Immunology and Infectious Diseases, UMC Utrecht, Lundlaan 6, 3584 EA, Utrecht, The Netherlands
| | - Anna Simon
- Department of Internal Medicine, Radboud UMC, Nijmegen, The Netherlands
| | - Taco W Kuijpers
- Department of Paediatric Immunology and Infectious Diseases, Emma Children's Hospital, AUMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Joris M van Montfrans
- Department of Pediatric Immunology and Infectious Diseases, UMC Utrecht, Lundlaan 6, 3584 EA, Utrecht, The Netherlands.
| |
Collapse
|
28
|
Spectrometric determination of trimethoprim and sulfamethoxazole in tablets by automated sequential injection technique. MONATSHEFTE FUR CHEMIE 2020. [DOI: 10.1007/s00706-020-02631-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
29
|
Kyosiimire-Lugemwa J, Anywaine Z, Abaasa A, Levin J, Gombe B, Musinguzi K, Kaleebu P, Grosskurth H, Munderi P, Pala P. Effect of Stopping Cotrimoxazole Preventive Therapy on Microbial Translocation and Inflammatory Markers Among Human Immunodeficiency Virus-Infected Ugandan Adults on Antiretroviral Therapy: The COSTOP Trial Immunology Substudy. J Infect Dis 2020; 222:381-390. [PMID: 31714954 PMCID: PMC7336573 DOI: 10.1093/infdis/jiz494] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 10/02/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Cotrimoxazole preventive therapy (CPT) in human immunodeficiency virus (HIV) infection is a World Health Organization-recommended standard of care in resource-limited settings, but the mechanism of CPT's beneficial effects is unclear. The COSTOP trial (ISRCTN44723643) evaluated the noninferiority of discontinuing CPT in stabilized patients on antiretroviral therapy. The COSTOP immunology substudy was conducted on a subset of COSTOP participants randomized to continue CPT (n = 86) or discontinue CPT (placebo, n = 86) as daily treatment for 1 year. METHODS We evaluated whether CPT reduces microbial translocation, indicated by the presence of bacterial lipopolysaccharide (LPS) and LPS control factors such as soluble CD14 (sCD14) and endotoxin core antibody (EndoCAb immunoglobulin M [IgM]) in plasma. Intestinal barrier damage as indicated by plasma intestinal fatty acid binding protein (IFABP), T-cell activation, and the inflammatory markers C-reactive protein (CRP), interleukin 6 (IL-6), and tumor necrosis factor α (TNF-α) were also evaluated. RESULTS We found no significant change in markers of microbial translocation (LPS, IFABP, sCD14, and T-cell activation), with decreased EndoCAb IgM. There was significant increase in inflammation markers (CRP and IL-6) after stopping CPT compared to those who continued CPT. CONCLUSIONS These results add to the evidence of immunological benefits of CPT among HIV-infected populations in resource-limited settings. However, no evidence of reducing microbial translocation was observed.
Collapse
Affiliation(s)
- Jacqueline Kyosiimire-Lugemwa
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda,Correspondence: J. Kyosiimire-Lugemwa, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, c/o Uganda Virus Research Institute, PO Box 49, Plot 51–59 Nakiwogo Road, Entebbe, Uganda ()
| | - Zacchaeus Anywaine
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Andrew Abaasa
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Jonathan Levin
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda,School of Public Health, University of the Witwatersrand, Johannesburg, South Africa
| | - Ben Gombe
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Kenneth Musinguzi
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Pontiano Kaleebu
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Heiner Grosskurth
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda,Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Paula Munderi
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda,International Association of Providers of AIDS Care, Washington, District of Columbia, USA
| | - Pietro Pala
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| |
Collapse
|
30
|
Sukasem C, Pratoomwun J, Satapornpong P, Klaewsongkram J, Rerkpattanapipat T, Rerknimitr P, Lertpichitkul P, Puangpetch A, Nakkam N, Konyoung P, Khunarkornsiri U, Disphanurat W, Srisuttiyakorn C, Pattanacheewapull O, Kanjanawart S, Kongpan T, Chumworathayi P, Saksit N, Bruminhent J, Tassaneeyakul W, Chantratita W, Pirmohamed M. Genetic Association of Co-Trimoxazole-Induced Severe Cutaneous Adverse Reactions Is Phenotype-Specific: HLA Class I Genotypes and Haplotypes. Clin Pharmacol Ther 2020; 108:1078-1089. [PMID: 32452529 DOI: 10.1002/cpt.1915] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 05/15/2020] [Indexed: 12/17/2022]
Abstract
Co-trimoxazole (CTX) causes various forms of severe cutaneous adverse reactions (SCARs). This case-control study was conducted to investigate the involvement between genetic variants of human leukocyte antigen (HLA) and CYP2C9 in CTX-induced SCARs, including Stevens-Johnson syndrome (SJS)/toxic epidermal necrolysis (TEN) and drug reaction with eosinophilia and systemic symptoms (DRESS) in Thai patients. Thirty cases of CTX-induced SCARs were enrolled and compared with 91 CTX-tolerant controls and 150 people from the general Thai population. Cases comprised 18 SJS/TEN and 12 DRESS patients. This study demonstrated that genetic association of CTX-induced SCARs was phenotype-specific. HLA-B*15:02 and HLA-C*08:01 alleles were significantly associated with CTX-induced SJS/TEN, whereas the HLA-B*13:01 allele was significantly associated with CTX-induced DRESS. In addition, a significant higher frequency of HLA-A*11:01-B*15:02 and HLA-B*13:01-C*03:04 haplotypes were detected in the group of CTX-induced Stevens-Johnson syndrome/toxic epidermal necrolysis (SJS/TEN) and DRESS cases, respectively. Genetic association of CTX-induced SCARs is phenotype-specific. Interestingly, these association was observed only in HIV-infected patients but not in non-HIV-infected patients.
Collapse
Affiliation(s)
- Chonlaphat Sukasem
- Division of Pharmacogenomics and Personalized Medicine, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.,Laboratory for Pharmacogenomics, Somdech Phra Debaratana Medical Center (SDMC), Ramathibodi Hospital, Bangkok, Thailand.,The Thai Severe Cutaneous Adverse Drug Reaction (THAI-SCAR) Research Group
| | - Jirawat Pratoomwun
- Division of Pharmacogenomics and Personalized Medicine, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.,Laboratory for Pharmacogenomics, Somdech Phra Debaratana Medical Center (SDMC), Ramathibodi Hospital, Bangkok, Thailand
| | - Patompong Satapornpong
- Division of Pharmacogenomics and Personalized Medicine, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.,Laboratory for Pharmacogenomics, Somdech Phra Debaratana Medical Center (SDMC), Ramathibodi Hospital, Bangkok, Thailand.,Division of General Pharmacy Practice, Department of Pharmaceutical Care, Faculty of Pharmacy, Rangsit University, Pathum Thani, Thailand
| | - Jettanong Klaewsongkram
- The Thai Severe Cutaneous Adverse Drug Reaction (THAI-SCAR) Research Group.,Division of Allergy and Clinical Immunology, Department of Medicine, Faculty of Medicine, Skin and Allergy Research Unit, Chulalongkorn University, Bangkok, Thailand.,King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Ticha Rerkpattanapipat
- The Thai Severe Cutaneous Adverse Drug Reaction (THAI-SCAR) Research Group.,Division of Allergy Immunology and Rheumatology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Pawinee Rerknimitr
- The Thai Severe Cutaneous Adverse Drug Reaction (THAI-SCAR) Research Group.,Division of Dermatology, Department of Medicine, Faculty of Medicine, Skin and Allergy Research Unit, Chulalongkorn University, Bangkok, Thailand
| | - Pattamon Lertpichitkul
- The Thai Severe Cutaneous Adverse Drug Reaction (THAI-SCAR) Research Group.,Division of Dermatology, Department of Medicine, Faculty of Medicine, Skin and Allergy Research Unit, Chulalongkorn University, Bangkok, Thailand
| | - Apichaya Puangpetch
- Division of Pharmacogenomics and Personalized Medicine, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.,Laboratory for Pharmacogenomics, Somdech Phra Debaratana Medical Center (SDMC), Ramathibodi Hospital, Bangkok, Thailand
| | - Nontaya Nakkam
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | | | | | - Wareeporn Disphanurat
- The Thai Severe Cutaneous Adverse Drug Reaction (THAI-SCAR) Research Group.,Division of Dermatology, Department of Medicine, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Chutika Srisuttiyakorn
- The Thai Severe Cutaneous Adverse Drug Reaction (THAI-SCAR) Research Group.,Division of Dermatology, Department of Medicine, Phramongkutklao Hospital, Phramongkutklao College of Medicine, Bangkok, Thailand
| | | | | | - Thachanan Kongpan
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Thailand
| | - Pansu Chumworathayi
- Pharmacy Unit, Srinagarind Hospital, Faculty of Medicine, Khon Kaen University, Thailand
| | - Niwat Saksit
- School of Pharmaceutical Sciences, University of Phayao, Phayao, Thailand
| | - Jackrapong Bruminhent
- Division of Infectious Diseases, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | | | - Wasun Chantratita
- Genomic Medicine Center, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Munir Pirmohamed
- Department of Molecular and Clinical Pharmacology, The Royal Liverpool, Broadgreen University Hospitals NHS Trust, MRC Centre for Drug Safety Science, Liverpool Health Partners, University of Liverpool, Liverpool, UK
| |
Collapse
|
31
|
Gough EK, Bourke CD, Berejena C, Shonhai A, Bwakura-Dangarembizi M, Prendergast AJ, Manges AR. Strain-level analysis of gut-resident pro-inflammatory viridans group Streptococci suppressed by long-term cotrimoxazole prophylaxis among HIV-positive children in Zimbabwe. Gut Microbes 2020; 11:1104-1115. [PMID: 32024435 PMCID: PMC7524282 DOI: 10.1080/19490976.2020.1717299] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Antimicrobials have become a mainstay of healthcare in the past century due to their activity against pathogens. More recently, it has become clear that they can also affect health via their impact on the microbiota and inflammation. This may explain some of their clinical benefits despite global increases in antimicrobial resistance (AMR) and reduced antimicrobial effectiveness. We showed in a randomized controlled trial of stopping versus continuing cotrimoxazole prophylaxis among HIV-positive Zimbabwean children taking antiretroviral therapy (ART), that continuation of cotrimoxazole persistently suppressed gut-resident viridans group streptococcal species (VGS) that were associated with intestinal inflammation. In this addendum, we provide a broader overview of how antibiotics can shape the microbiota and use high read-depth whole metagenome sequencing data from our published study to investigate whether (i) the impact of cotrimoxazole on gut VGS and (ii) VGS associated inflammation, is attributable to strain-level variability. We focus on S. salivarius, the VGS species that was most prevalent in the cohort and for which there was sufficient genome coverage to differentiate strains. We demonstrate that suppression of S. salivarius by cotrimoxazole is not strain specific, nor did stool concentration of the pro-inflammatory mediator myeloperoxidase vary by S. salivarius strain. We also show that gut-resident S. salivarius strains present in this study population are distinct from common oral strains. This is the first analysis of how cotrimoxazole prophylaxis used according to international treatment guidelines for children living with HIV influences the gut microbiome at the strain-level. We also provide a detailed review of the literature on the mechanisms by which suppression of VGS may act synergistically with cotrimoxazole's anti-inflammatory effects to reduce gut inflammation. A greater understanding of the sub-clinical effects of antibiotics offers new insights into their responsible clinical use.
Collapse
Affiliation(s)
- Ethan K. Gough
- Department of International Health, Division of Human Nutrition, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA,CONTACT Ethan K. Gough Department of International Health, Division of Human Nutrition, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Claire D. Bourke
- Blizard Institute, Queen Mary University of London, London, UK,Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Chipo Berejena
- College of Health Sciences, University of Zimbabwe, Harare, Zimbabwe
| | - Annie Shonhai
- College of Health Sciences, University of Zimbabwe, Harare, Zimbabwe
| | | | - Andrew J. Prendergast
- Blizard Institute, Queen Mary University of London, London, UK,Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe,MRC Clinical Trials Unit at University College London, London, UK
| | - Amee R. Manges
- School of Population and Public Health, University of British Columbia, Vancouver, Canada
| |
Collapse
|
32
|
Oyedeji OA. Malaria in a 2-Month-Old HIV-Exposed Nigerian Infant: Challenges of Care. J Int Assoc Provid AIDS Care 2020; 18:2325958219849052. [PMID: 31117862 PMCID: PMC6748458 DOI: 10.1177/2325958219849052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background: Reports on malaria and HIV coinfections in exposed infants from tropical countries are
scarce. Results: The case of a 2-month-old HIV-exposed Nigerian infant who presented with intermittent
fever at a Nigerian tertiary hospital is reported. The rarity of the case and the
challenges associated with making the diagnosis informed our decision to report the
case. Conclusion: Diagnosing malaria in HIV-exposed infants in early infancy requires a high index of
suspicion, good knowledge of the clinical presentation, and appropriate microbiological
investigations for sepsis and malaria. Further studies need to be conducted on the
association between malaria and HIV exposure.
Collapse
|
33
|
Maitland K, Olupot-Olupot P, Kiguli S, Chagaluka G, Alaroker F, Opoka RO, Mpoya A, Walsh K, Engoru C, Nteziyaremye J, Mallewa M, Kennedy N, Nakuya M, Namayanja C, Kayaga J, Nabawanuka E, Sennyondo T, Aromut D, Kumwenda F, Musika CW, Thomason MJ, Bates I, von Hensbroek MB, Evans JA, Uyoga S, Williams TN, Frost G, George EC, Gibb DM, Walker AS. Co-trimoxazole or multivitamin multimineral supplement for post-discharge outcomes after severe anaemia in African children: a randomised controlled trial. LANCET GLOBAL HEALTH 2020; 7:e1435-e1447. [PMID: 31537373 PMCID: PMC7024999 DOI: 10.1016/s2214-109x(19)30345-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 07/07/2019] [Accepted: 07/18/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Severe anaemia is a leading cause of paediatric admission to hospital in Africa; post-discharge outcomes remain poor, with high 6-month mortality (8%) and re-admission (17%). We aimed to investigate post-discharge interventions that might improve outcomes. METHODS Within the two-stratum, open-label, multicentre, factorial randomised TRACT trial, children aged 2 months to 12 years with severe anaemia, defined as haemoglobin of less than 6 g/dL, at admission to hospital (three in Uganda, one in Malawi) were randomly assigned, using sequentially numbered envelopes linked to a second non-sequentially numbered set of allocations stratified by centre and severity, to enhanced nutritional supplementation with iron and folate-containing multivitamin multimineral supplements versus iron and folate alone at treatment doses (usual care), and to co-trimoxazole versus no co-trimoxazole. All interventions were administered orally and were given for 3 months after discharge from hospital. Separately reported randomisations investigated transfusion management. The primary outcome was 180-day mortality. All analyses were done in the intention-to-treat population; follow-up was 180 days. This trial is registered with the International Standard Randomised Controlled Trial registry, ISRCTN84086586, and follow-up is complete. FINDINGS From Sept 17, 2014, to May 15, 2017, 3983 eligible children were randomly assigned to treatment, and followed up for 180 days. 164 (4%) were lost to follow-up. 1901 (95%) of 1997 assigned multivitamin multimineral supplement, 1911 (96%) of 1986 assigned iron and folate, and 1922 (96%) of 1994 assigned co-trimoxazole started treatment. By day 180, 166 (8%) children in the multivitamin multimineral supplement group versus 169 (9%) children in the iron and folate group had died (hazard ratio [HR] 0·97, 95% CI 0·79-1·21; p=0·81) and 172 (9%) who received co-trimoxazole versus 163 (8%) who did not receive co-trimoxazole had died (HR 1·07, 95% CI 0·86-1·32; p=0·56). We found no evidence of interactions between these randomisations or with transfusion randomisations (p>0·2). By day 180, 489 (24%) children in the multivitamin multimineral supplement group versus 509 (26%) children in the iron and folate group (HR 0·95, 95% CI 0·84-1·07; p=0·40), and 500 (25%) children in the co-trimoxazole group versus 498 (25%) children in the no co-trimoxazole group (1·01, 0·89-1·15; p=0·85) had had one or more serious adverse events. Most serious adverse events were re-admissions, occurring in 692 (17%) children (175 [4%] with at least two re-admissions). INTERPRETATION Neither enhanced supplementation with multivitamin multimineral supplement versus iron and folate treatment or co-trimoxazole prophylaxis improved 6-month survival. High rates of hospital re-admission suggest that novel interventions are urgently required for severe anaemia, given the burden it places on overstretched health services in Africa. FUNDING Medical Research Council and Department for International Development.
Collapse
Affiliation(s)
- Kathryn Maitland
- Department of Medicine, Imperial College London, London, UK; Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya.
| | - Peter Olupot-Olupot
- Busitema University Faculty of Health Sciences, Mbale Campus and Mbale Regional Referral Hospital Mbale, Mbale, Uganda
| | - Sarah Kiguli
- Department of Paediatrics, Makerere University and Mulago Hospital, Kampala, Uganda
| | - George Chagaluka
- College of Medicine, and Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
| | | | - Robert O Opoka
- Department of Paediatrics, Makerere University and Mulago Hospital, Kampala, Uganda
| | - Ayub Mpoya
- Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Kevin Walsh
- Nutrition Research Section, Imperial College London, London, UK
| | | | - Julius Nteziyaremye
- Busitema University Faculty of Health Sciences, Mbale Campus and Mbale Regional Referral Hospital Mbale, Mbale, Uganda
| | - Machpherson Mallewa
- College of Medicine, and Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
| | - Neil Kennedy
- College of Medicine, and Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi; School of Medicine, Dentistry and Biomedical Science, Queen's University, Belfast, UK
| | | | - Cate Namayanja
- Busitema University Faculty of Health Sciences, Mbale Campus and Mbale Regional Referral Hospital Mbale, Mbale, Uganda
| | - Julianne Kayaga
- Department of Paediatrics, Makerere University and Mulago Hospital, Kampala, Uganda
| | - Eva Nabawanuka
- Department of Paediatrics, Makerere University and Mulago Hospital, Kampala, Uganda
| | - Tonny Sennyondo
- Busitema University Faculty of Health Sciences, Mbale Campus and Mbale Regional Referral Hospital Mbale, Mbale, Uganda
| | - Denis Aromut
- Soroti Regional Referral Hospital, Soroti, Uganda
| | - Felistas Kumwenda
- College of Medicine, and Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
| | | | | | - Imelda Bates
- Liverpool School of Tropical Medicine and Hygiene, Liverpool, UK
| | | | - Jennifer A Evans
- Department of Paediatrics, University Hospital of Wales, Cardiff, UK
| | - Sophie Uyoga
- Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Thomas N Williams
- Department of Medicine, Imperial College London, London, UK; Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Gary Frost
- Nutrition Research Section, Imperial College London, London, UK
| | - Elizabeth C George
- Medical Research Council Clinical Trials Unit at University College London
| | - Diana M Gibb
- Medical Research Council Clinical Trials Unit at University College London
| | - A Sarah Walker
- Medical Research Council Clinical Trials Unit at University College London
| | | |
Collapse
|
34
|
Bourke CD, Gough EK, Pimundu G, Shonhai A, Berejena C, Terry L, Baumard L, Choudhry N, Karmali Y, Bwakura-Dangarembizi M, Musiime V, Lutaakome J, Kekitiinwa A, Mutasa K, Szubert AJ, Spyer MJ, Deayton JR, Glass M, Geum HM, Pardieu C, Gibb DM, Klein N, Edens TJ, Walker AS, Manges AR, Prendergast AJ. Cotrimoxazole reduces systemic inflammation in HIV infection by altering the gut microbiome and immune activation. Sci Transl Med 2020; 11:11/486/eaav0537. [PMID: 30944164 DOI: 10.1126/scitranslmed.aav0537] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 11/21/2018] [Accepted: 02/14/2019] [Indexed: 12/21/2022]
Abstract
Long-term cotrimoxazole prophylaxis reduces mortality and morbidity in HIV infection, but the mechanisms underlying these clinical benefits are unclear. Here, we investigate the impact of cotrimoxazole on systemic inflammation, an independent driver of HIV mortality. In HIV-positive Ugandan and Zimbabwean children receiving antiretroviral therapy, we show that plasma inflammatory markers were lower after randomization to continue (n = 144) versus stop (n = 149) cotrimoxazole. This was not explained by clinical illness, HIV progression, or nutritional status. Because subclinical enteropathogen carriage and enteropathy can drive systemic inflammation, we explored cotrimoxazole effects on the gut microbiome and intestinal inflammatory biomarkers. Although global microbiome composition was unchanged, viridans group Streptococci and streptococcal mevalonate pathway enzymes were lower among children continuing (n = 36) versus stopping (n = 36) cotrimoxazole. These changes were associated with lower fecal myeloperoxidase. To isolate direct effects of cotrimoxazole on immune activation from antibiotic effects, we established in vitro models of systemic and intestinal inflammation. In vitro cotrimoxazole had modest but consistent inhibitory effects on proinflammatory cytokine production by blood leukocytes from HIV-positive (n = 16) and HIV-negative (n = 8) UK adults and reduced IL-8 production by gut epithelial cell lines. Collectively we demonstrate that cotrimoxazole reduces systemic and intestinal inflammation both indirectly via antibiotic effects on the microbiome and directly by blunting immune and epithelial cell activation. Synergy between these pathways may explain the clinical benefits of cotrimoxazole despite high antimicrobial resistance, providing further rationale for extending coverage among people living with HIV in sub-Saharan Africa.
Collapse
Affiliation(s)
- Claire D Bourke
- Blizard Institute, Queen Mary University of London, London E1 2AT, UK.
| | - Ethan K Gough
- School of Population and Public Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | | | - Annie Shonhai
- College of Health Sciences, University of Zimbabwe, Harare, Zimbabwe
| | - Chipo Berejena
- College of Health Sciences, University of Zimbabwe, Harare, Zimbabwe
| | - Louise Terry
- Royal London Hospital, Barts Health NHS Trust, London E1 1BB, UK
| | - Lucas Baumard
- Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| | - Naheed Choudhry
- Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| | - Yusuf Karmali
- Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| | | | - Victor Musiime
- Joint Clinical Research Centre, Kampala, Uganda.,College of Health Sciences, Department of Paediatrics and Child Health, Makerere University, Kampala, Uganda
| | - Joseph Lutaakome
- Uganda Virus Research Institute/MRC Uganda Research Unit on AIDS, Entebbe, Uganda
| | - Adeodata Kekitiinwa
- Baylor College of Medicine Children's Foundation-Uganda, Mulago Hospital, Kampala, Uganda
| | - Kuda Mutasa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | | | - Moira J Spyer
- MRC Clinical Trials Unit at University College London, London WC1V 6LJ, UK
| | - Jane R Deayton
- Blizard Institute, Queen Mary University of London, London E1 2AT, UK.,Royal London Hospital, Barts Health NHS Trust, London E1 1BB, UK
| | - Magdalena Glass
- School of Population and Public Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Hyun Min Geum
- School of Population and Public Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Claire Pardieu
- Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| | - Diana M Gibb
- MRC Clinical Trials Unit at University College London, London WC1V 6LJ, UK
| | - Nigel Klein
- UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
| | - Thaddeus J Edens
- Devil's Staircase Consulting, West Vancouver, British Columbia V7T 1V7, Canada
| | - A Sarah Walker
- MRC Clinical Trials Unit at University College London, London WC1V 6LJ, UK
| | - Amee R Manges
- School of Population and Public Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Andrew J Prendergast
- Blizard Institute, Queen Mary University of London, London E1 2AT, UK.,Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe.,MRC Clinical Trials Unit at University College London, London WC1V 6LJ, UK
| |
Collapse
|
35
|
Musimbi ZD, Rono MK, Otieno JR, Kibinge N, Ochola-Oyier LI, de Villiers EP, Nduati EW. Peripheral blood mononuclear cell transcriptomes reveal an over-representation of down-regulated genes associated with immunity in HIV-exposed uninfected infants. Sci Rep 2019; 9:18124. [PMID: 31792230 PMCID: PMC6889308 DOI: 10.1038/s41598-019-54083-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 11/08/2019] [Indexed: 12/18/2022] Open
Abstract
HIV-exposed uninfected (HEU) infants are disproportionately at a higher risk of morbidity and mortality, as compared to HIV-unexposed uninfected (HUU) infants. Here, we used transcriptional profiling of peripheral blood mononuclear cells to determine immunological signatures of in utero HIV exposure. We identified 262 differentially expressed genes (DEGs) in HEU compared to HUU infants. Weighted gene co-expression network analysis (WGCNA) identified six modules that had significant associations with clinical traits. Functional enrichment analysis on both DEGs and the six significantly associated modules revealed an enrichment of G-protein coupled receptors and the immune system, specifically affecting neutrophil function and antibacterial responses. Additionally, malaria pathogenicity genes (thrombospondin 1-(THBS 1), interleukin 6 (IL6), and arginine decarboxylase 2 (ADC2)) were down-regulated. Of interest, the down-regulated immunity genes were positively correlated to the expression of epigenetic factors of the histone family and high-mobility group protein B2 (HMGB2), suggesting their role in the dysregulation of the HEU transcriptional landscape. Overall, we show that genes primarily associated with neutrophil mediated immunity were repressed in the HEU infants. Our results suggest that this could be a contributing factor to the increased susceptibility to bacterial infections associated with higher morbidity and mortality commonly reported in HEU infants.
Collapse
Affiliation(s)
- Zaneta D Musimbi
- Center of Biotechnology and Bioinformatics, Chiromo Campus, University of Nairobi, Nairobi, Kenya.
| | - Martin K Rono
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya.
- Pwani University Biotechnology Research Centre, Pwani University, Kilifi, Kenya.
| | | | | | - Lynette Isabella Ochola-Oyier
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Pwani University Biotechnology Research Centre, Pwani University, Kilifi, Kenya
| | - Etienne Pierre de Villiers
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | - Eunice W Nduati
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Pwani University Biotechnology Research Centre, Pwani University, Kilifi, Kenya
| |
Collapse
|
36
|
Maloupazoa Siawaya AC, Kuissi Kamgaing E, Minto'o Rogombe S, Obiang T, Moungoyi Massala E, Magossou Mbadinga MJV, Leboueny M, Mvoundza Ndjindji O, Mveang-Nzoghe A, Ondo JP, Mintsa Ndong A, N Essone P, Agnandji ST, Kaba M, Ategbo S, Djoba Siawaya JF. HIV-exposed uninfected compared with unexposed infants show the presence of leucocytes, lower lactoferrin levels and antimicrobial-resistant micro-organisms in the stool. Paediatr Int Child Health 2019; 39:249-258. [PMID: 30762489 DOI: 10.1080/20469047.2019.1577063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Background: HIV-exposed uninfected (HEU)-infants have been shown to be particularly vulnerable to infections. In this population, disturbance of the gut micro-environment might increase their susceptibility to enteric diseases and even favour the translocation of bacteria in the bloodstream. Methods: The gastro-intestinal micro-environment was explored in 22 HEU infants and 16 HIV-unexposed (HU) infants aged 6-24 weeks. Faecal leucocytes, firmicutes (gram-positive bacteria) and gracilicutes (gram-negative bacteria) were assessed by cytology. Faecal lactoferrin and sIgA were measured by ELISA. The spectrum of micro-organisms in infants' stool was analysed by culturing. Results: HEU infants were 14 times more likely to have leucocytes in their stool than HU infants (p < 0.005). The lactoferrin level was significantly lower in HEU infants than in HU infants (p = 0.02). Potentially pathogenic bacteria such as Escherichia coli were more prevalent in HEU than in HU infants (64% vs 23.5%). Also, E. coli strains resistant to key antibiotics including co-trimoxazole, β-lactam (cephalosporins included) and tetraclines were observed in some HEU infants. Conclusion: HEU infants are more likely to present an inflamed digestive tract as highlighted by the presence of leucocytes. In addition, there is a real risk of colonisation of HEU infants' microbiota by resistant micro-organisms.
Collapse
Affiliation(s)
- A C Maloupazoa Siawaya
- Laboratoire National de Santé Publique, Unités de Recherche et de Diagnostics Spécialisés , Libreville , Gabon.,Service Laboratoire, Centre Hospitalier Universitaire Mère-Enfant Fondation Jeanne Ebori , Libreville , Gabon
| | - E Kuissi Kamgaing
- Département de Pédiatrie, Université des Sciences de la Santé , Libreville , Gabon
| | - S Minto'o Rogombe
- Département de Pédiatrie, Université des Sciences de la Santé , Libreville , Gabon
| | - T Obiang
- Département de Pédiatrie, Université des Sciences de la Santé , Libreville , Gabon
| | - E Moungoyi Massala
- Département de Chimie, Faculté des Sciences, Université des Sciences et Techniques de Masuku , Franceville , Gabon
| | - M J V Magossou Mbadinga
- Laboratoire National de Santé Publique, Unités de Recherche et de Diagnostics Spécialisés , Libreville , Gabon.,Service Laboratoire, Centre Hospitalier Universitaire Mère-Enfant Fondation Jeanne Ebori , Libreville , Gabon
| | - M Leboueny
- Laboratoire National de Santé Publique, Unités de Recherche et de Diagnostics Spécialisés , Libreville , Gabon.,Service Laboratoire, Centre Hospitalier Universitaire Mère-Enfant Fondation Jeanne Ebori , Libreville , Gabon
| | - O Mvoundza Ndjindji
- Laboratoire National de Santé Publique, Unités de Recherche et de Diagnostics Spécialisés , Libreville , Gabon.,Service Laboratoire, Centre Hospitalier Universitaire Mère-Enfant Fondation Jeanne Ebori , Libreville , Gabon
| | - A Mveang-Nzoghe
- Laboratoire National de Santé Publique, Unités de Recherche et de Diagnostics Spécialisés , Libreville , Gabon.,Service Laboratoire, Centre Hospitalier Universitaire Mère-Enfant Fondation Jeanne Ebori , Libreville , Gabon
| | - J P Ondo
- Département de Chimie, Faculté des Sciences, Université des Sciences et Techniques de Masuku , Franceville , Gabon
| | - A Mintsa Ndong
- Laboratoire National de Santé Publique, Unité de Virologie , Libreville , Gabon
| | - P N Essone
- Laboratoire National de Santé Publique, Unités de Recherche et de Diagnostics Spécialisés , Libreville , Gabon.,Centre de Recherches Médicales de Lambaréné , Lambaréné , Gabon.,Institut für Tropenmedizin, Universitätsklinikum Tübingen , Tübingen , Germany
| | - S T Agnandji
- Centre de Recherches Médicales de Lambaréné , Lambaréné , Gabon.,Institut für Tropenmedizin, Universitätsklinikum Tübingen , Tübingen , Germany
| | - M Kaba
- Division of Medical Microbiology, University of Cape Town , Cape Town , South Africa
| | - S Ategbo
- Département de Pédiatrie, Université des Sciences de la Santé , Libreville , Gabon
| | - J F Djoba Siawaya
- Laboratoire National de Santé Publique, Unités de Recherche et de Diagnostics Spécialisés , Libreville , Gabon.,Service Laboratoire, Centre Hospitalier Universitaire Mère-Enfant Fondation Jeanne Ebori , Libreville , Gabon
| |
Collapse
|
37
|
Prendergast AJ, Walson JL. Seeking interventions to reduce post-discharge mortality among children in sub-Saharan Africa. Lancet Glob Health 2019; 7:e1306-e1307. [PMID: 31537352 DOI: 10.1016/s2214-109x(19)30373-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 08/27/2019] [Indexed: 01/24/2023]
Affiliation(s)
- Andrew J Prendergast
- Blizard Institute, Queen Mary University of London, London E1 2AT, UK; Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe.
| | - Judd L Walson
- University of Washington, Seattle, WA, USA; Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
| |
Collapse
|
38
|
Frigati L, Archary M, Rabie H, Penazzato M, Ford N. Priorities for Decreasing Morbidity and Mortality in Children With Advanced HIV Disease. Clin Infect Dis 2019. [PMID: 29514237 PMCID: PMC5850631 DOI: 10.1093/cid/ciy013] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Early mortality and morbidity remain high in children initiating antiretroviral therapy (ART), especially in sub-Saharan Africa. Many children still present with advanced human immunodeficiency virus (HIV) disease. Tuberculosis, pneumonia, and severe bacterial infections are the main causes of hospital admission in HIV-infected children. In contrast to adults with advanced HIV disease, cryptococcal disease is not common in childhood, although there is a peak in infancy and adolescence. Interventions such as TB screening in symptomatic children, and isoniazid and cotrimoxazole prophylaxis should be implemented. There is evidence suggesting that rapid initiation (within 1 week) of ART in children with severe malnutrition or those with advanced HIV disease admitted to hospital is not beneficial and should be delayed until their condition has been stabilized. Research informing the prevention of severe bacterial infections, the management of pediatric immune reconstitution inflammatory syndrome, and other potential strategies to decrease morbidity and mortality in HIV-infected children are urgently needed.
Collapse
Affiliation(s)
- Lisa Frigati
- Department of Paediatrics and Child Health, Tygerberg Hospital and Stellenbosch University, Cape Town
| | - Moherdran Archary
- University of KwaZulu Natal, Nelson R Mandela School of Medicine, Berea, South Africa
| | - Helena Rabie
- Department of Paediatrics and Child Health, Tygerberg Hospital and Stellenbosch University, Cape Town
| | | | - Nathan Ford
- HIV Department, World Health Organization, Geneva, Switzerland
| |
Collapse
|
39
|
Ocular manifestations in patients with Stevens-Johnson syndrome in Malawi-review of the literature illustrated by clinical cases. Graefes Arch Clin Exp Ophthalmol 2019; 257:2343-2348. [PMID: 31352606 DOI: 10.1007/s00417-019-04421-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 07/07/2019] [Accepted: 07/10/2019] [Indexed: 12/19/2022] Open
Abstract
PURPOSE In Sub-Saharan Africa, manifestations of Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN) are commonly seen in human immunodeficiency virus-infected patients receiving nevirapine-based antiretroviral therapy and/or cotrimoxazole. These patients often face severe ocular complications that lead to moderate to severe visual impairment or blindness. METHODS Review of the current literature, illustrated by retrospective hospital-based case series: Eight patients at Lions Sight First Eye Hospital, Blantyre, Malawi with severe ocular complications like severe cicatrizing conjunctivitis with symblephara, corneal punctate erosions, corneal vascularization, and corneal ulceration are illustrated after the diagnosis of SJS/TEN. RESULTS Light perception was reported in six (12 eyes) of them; two patients (4 eyes) had moderate visual impairment (6/36 and 6/18). In one patient, eye problems started after therapy with cotrimoxazole; in seven after therapy, with antiretroviral therapy. CONCLUSION SJS/TEN in Sub Saharan Africa correlates significantly with moderate visual impairment up to blindness. Early recognition of eye complications and involvement of ophthalmologists in the acute stage, early treatment with local steroids, and close monitoring for up to 6 months after the acute phase are crucial. Severe ocular complications seem to be more severe in dark skin phototype.
Collapse
|
40
|
van Eijk AM, Larsen DA, Kayentao K, Koshy G, Slaughter DEC, Roper C, Okell LC, Desai M, Gutman J, Khairallah C, Rogerson SJ, Hopkins Sibley C, Meshnick SR, Taylor SM, Ter Kuile FO. Effect of Plasmodium falciparum sulfadoxine-pyrimethamine resistance on the effectiveness of intermittent preventive therapy for malaria in pregnancy in Africa: a systematic review and meta-analysis. THE LANCET. INFECTIOUS DISEASES 2019; 19:546-556. [PMID: 30922818 DOI: 10.1016/s1473-3099(18)30732-1] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/30/2018] [Accepted: 11/20/2018] [Indexed: 01/05/2023]
Abstract
BACKGROUND Resistance of Plasmodium falciparum to sulfadoxine-pyrimethamine threatens the antimalarial effectiveness of intermittent preventive treatment during pregnancy (IPTp) in sub-Saharan Africa. We aimed to assess the associations between markers of sulfadoxine-pyrimethamine resistance in P falciparum and the effectiveness of sulfadoxine-pyrimethamine IPTp for malaria-associated outcomes. METHODS For this systematic review and meta-analysis, we searched databases (from Jan 1, 1990 to March 1, 2018) for clinical studies (aggregated data) or surveys (individual participant data) that reported data on low birthweight (primary outcome) and malaria by sulfadoxine-pyrimethamine IPTp dose, and for studies that reported on molecular markers of sulfadoxine-pyrimethamine resistance. Studies that involved only HIV-infected women or combined interventions were excluded. We did a random-effects meta-analysis (clinical studies) or multivariate log-binomial regression (surveys) to obtain summarised dose-response data (relative risk reduction [RRR]) and multivariate meta-regression to explore the modifying effects of sulfadoxine-pyrimethamine resistance (as indicated by Ala437Gly, Lys540Glu, and Ala581Gly substitutions in the dhps gene). This study is registered with PROSPERO, number 42016035540. FINDINGS Of 1097 records screened, 57 studies were included in the aggregated-data meta-analysis (including 59 457 births). The RRR for low birthweight declined with increasing prevalence of dhps Lys540Glu (ptrend=0·0060) but not Ala437Gly (ptrend=0·35). The RRR was 7% (95% CI 0 to 13) in areas of high resistance to sulfadoxine-pyrimethamine (Lys540Glu ≥90% in east and southern Africa; n=11), 21% (14 to 29) in moderate-resistance areas (Ala437Gly ≥90% [central and west Africa], or Lys540Glu ≥30% to <90% [east and southern Africa]; n=16), and 27% (21 to 33) in low-resistance areas (Ala437Gly <90% [central and west Africa], or Lys540Glu <30% [east and southern Africa]; n=30; ptrend=0·0054 [univariate], I2=69·5%). The overall RRR in all resistance strata was 21% (17 to 25). In the analysis of individual participant data from 13 surveys (42 394 births), sulfadoxine-pyrimethamine IPTp was associated with reduced prevalence of low birthweight in areas with a Lys540Glu prevalence of more than 90% and Ala581Gly prevalence of less than 10% (RRR 10% [7 to 12]), but not in those with an Ala581Gly prevalence of 10% or higher (pooled Ala581Gly prevalence 37% [range 29 to 46]; RRR 0·5% [-16 to 14]; 2326 births). INTERPRETATION The effectiveness of sulfadoxine-pyrimethamine IPTp is reduced in areas with high resistance to sulfadoxine-pyrimethamine among P falciparum parasites, but remains associated with reductions in low birthweight even in areas where dhps Lys540Glu prevalence exceeds 90% but where the sextuple-mutant parasite (harbouring the additional dhps Ala581Gly mutation) is uncommon. Therapeutic alternatives to sulfadoxine-pyrimethamine IPTp are needed in areas where the prevalence of the sextuple-mutant parasite exceeds 37%. FUNDING US Centers for Disease Control and Prevention, the Malaria in Pregnancy Consortium (funded through a grant from the Bill & Melinda Gates Foundation to the Liverpool School of Tropical Medicine), Worldwide Antimalarial Resistance Network, European and Developing Countries Clinical Trials Partnership.
Collapse
Affiliation(s)
- Anna Maria van Eijk
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - David A Larsen
- Department of Public Health, Food Studies and Nutrition, Syracuse University, Syracuse, NY, USA
| | - Kassoum Kayentao
- Malaria Research and Training Centre, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine, Pharmacy, and Dentistry, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali
| | - Gibby Koshy
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | | | - Cally Roper
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Lucy C Okell
- MRC Centre for Outbreak Analysis and Modelling, Department of Infectious Disease Epidemiology, Imperial College London, London, UK
| | - Meghna Desai
- Malaria Branch, US Centers for Diseases Control and Prevention, Atlanta, GA, USA
| | - Julie Gutman
- Malaria Branch, US Centers for Diseases Control and Prevention, Atlanta, GA, USA
| | - Carole Khairallah
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Stephen J Rogerson
- Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | - Carol Hopkins Sibley
- Department of Genome Sciences, University of Washington, Seattle, WA, USA; WorldWide Antimalarial Resistance Network, University of Oxford, Oxford, UK
| | - Steven R Meshnick
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Steve M Taylor
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA; Division of Infectious Diseases and Duke Global Health Institute, Duke University Medical Center, Durham, NC, USA
| | - Feiko O Ter Kuile
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK.
| |
Collapse
|
41
|
Manenzhe RI, Moodley C, Abdulgader SM, Robberts FJL, Zar HJ, Nicol MP, Dube FS. Nasopharyngeal Carriage of Antimicrobial-Resistant Pneumococci in an Intensively Sampled South African Birth Cohort. Front Microbiol 2019; 10:610. [PMID: 30972052 PMCID: PMC6446970 DOI: 10.3389/fmicb.2019.00610] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Accepted: 03/11/2019] [Indexed: 01/22/2023] Open
Abstract
Introduction: Nasopharyngeal (NP) colonization by Streptococcus pneumoniae (pneumococcus) precedes the development of respiratory tract infection. Colonization by antimicrobial-resistant pneumococci, especially in infants, is a major public health concern. We longitudinally investigated antimicrobial-resistance amongst pneumococci colonizing the nasopharynx of South African infants immunized with the 13-valent pneumococcal conjugate vaccine (PCV13). Methods: NP swabs were collected every second week from birth through the first year of life from 137 infants. Pneumococci were identified and serotyped using conventional microbiological techniques, and their antibiotic susceptibility profiles determined by disk diffusion and E-test. Results: All infants were immunized with 3 doses of PCV13. 1520 pneumococci (760 non-repeat) isolates were recovered from 137 infants; including non-typeable (n = 99), PCV13 (n = 133) and non-PCV13 serotypes (n = 528). The prevalence of penicillin, erythromycin, and cotrimoxazole non-susceptibility was 19% (95% CI 17-22%) (3% fully resistant), 18% (95% CI 15-21%) (14% fully resistant), and 45% (95% CI 42-49%) (36% fully resistant), respectively. The predominant penicillin-non-susceptible serotypes included 19A, 19F, 15B/15C, 15A, and 21, while susceptible serotypes included 23A, 34, and 17A. Multidrug-resistance (MDR) was observed in 9% (95% CI 7-11%) of the isolates. PCV13 serotypes were more likely to be non-susceptible, compared to non-PCV13 serotypes, to penicillin (26% vs. 16%, p = 0.007), erythromycin (23% vs. 15%, p = 0.027) and cotrimoxazole (62% vs. 41%, p < 0.001). Non-susceptibility to penicillin, erythromycin, and cotrimoxazole remained relatively constant through the first year of life (X 2 test for trend: p = 0.184, p = 0.171, and p = 0.572, respectively). Overall, penicillin or erythromycin-non-susceptible pneumococci were carried for a shorter duration than susceptible pneumococci [penicillin (mean days, 18 vs. 21, p = 0.013) and erythromycin (mean days, 18 vs. 21, p = 0.035)]. Within individual infants carrying the same serotype longitudinally, changes in antibiotic susceptibility were observed over time in 45% (61/137) of infants and these changes were predominantly for penicillin (76%, 79/104). Conclusion: Prevalence of NP carriage with antibiotic-non-susceptible pneumococci was relatively constant throughout the first year of life. PCV13 serotypes were more commonly non-susceptible to penicillin, erythromycin, and cotrimoxazole. Penicillin or erythromycin-non-susceptible pneumococci were carried for a shorter duration than penicillin or erythromycin-susceptible pneumococci.
Collapse
Affiliation(s)
- Rendani I. Manenzhe
- Division of Medical Microbiology, Faculty of Health Science, University of Cape Town, Cape Town, South Africa
| | - Clinton Moodley
- Division of Medical Microbiology, Faculty of Health Science, University of Cape Town, Cape Town, South Africa
- National Health Laboratory Service, Groote Schuur Hospital, Cape Town, South Africa
| | - Shima M. Abdulgader
- Division of Medical Microbiology, Faculty of Health Science, University of Cape Town, Cape Town, South Africa
| | - F. J. Lourens Robberts
- Division of Medical Microbiology, Faculty of Health Science, University of Cape Town, Cape Town, South Africa
| | - Heather J. Zar
- Department of Paediatrics and Child Health, Red Cross War Memorial Children’s Hospital and SA-MRC Unit on Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Mark P. Nicol
- Division of Medical Microbiology, Faculty of Health Science, University of Cape Town, Cape Town, South Africa
- National Health Laboratory Service, Groote Schuur Hospital, Cape Town, South Africa
| | - Felix S. Dube
- Division of Medical Microbiology, Faculty of Health Science, University of Cape Town, Cape Town, South Africa
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
42
|
Juma DW, Muiruri P, Yuhas K, John-Stewart G, Ottichilo R, Waitumbi J, Singa B, Polyak C, Kamau E. The prevalence and antifolate drug resistance profiles of Plasmodium falciparum in study participants randomized to discontinue or continue cotrimoxazole prophylaxis. PLoS Negl Trop Dis 2019; 13:e0007223. [PMID: 30897090 PMCID: PMC6445470 DOI: 10.1371/journal.pntd.0007223] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 04/02/2019] [Accepted: 02/06/2019] [Indexed: 11/19/2022] Open
Abstract
Objective Cotrimoxazole prevents opportunistic infections including falciparum malaria in HIV-infected individuals but there are concerns of cross-resistance to other antifolate drugs such as sulphadoxine-pyrimethamine (SP). In this study, we investigated the prevalence of antifolate-resistance mutations in Plasmodium falciparum that are associated with SP resistance in HIV-infected individuals on antiretroviral treatment randomized to discontinue (STOP-CTX), or continue (CTX) cotrimoxazole in Western Kenya. Design Samples were obtained from an unblinded, non-inferiority randomized controlled trial where participants were recruited on a rolling basis for the first six months of the study, then followed-up for 12 months with samples collected at enrollment, quarterly, and during sick visits. Method Plasmodium DNA was extracted from blood specimens. Initial screening to determine the presence of Plasmodium spp. was performed by quantitative reverse transcriptase real-time PCR, followed by genotyping for the presence of SP-resistance associated mutations by Sanger sequencing. Results The prevalence of mutant haplotypes associated with SP-resistant parasites in pfdhfr (51I/59R/108N) and pfdhps (437G/540E) genes were significantly higher (P = 0.0006 and P = 0.027, respectively) in STOP-CTX compared to CTX arm. The prevalence of quintuple haplotype (51I/59R/108N/437G/540E) was 51.8% in STOP-CTX vs. 6.3% (P = 0.0007) in CTX arm. There was a steady increase in mutant haplotypes in both genes in STOP-CTX arm overtime through the study period, reaching statistical significance (P < 0.0001). Conclusion The frequencies of mutations in pfdhfr and pfdhps genes were higher in STOP-CTX arm compared to CTX arm, suggesting cotrimoxazole effectively controls and selects against SP-resistant parasites. Trial registration ClinicalTrials.gov NCT01425073 Cotrimoxazole, an antifolate, is a fixed-dose trimethoprim-sulfamethoxazole used to prevent opportunistic infections including malaria in HIV-infected individuals. There are concerns that widespread use of cotrimoxazole for prophylaxis may result in selection of P. falciparum parasites with cross-resistance to other antifolate drugs such as sulphadoxine-pyrimethamine (SP), which is used as intermittent preventive treatment of malaria in pregnancy (IPTp) and in infants (IPTi) in Africa. This sub-study used samples from a clinical trial in which HIV-infected individuals on antiretroviral treatment were randomized to discontinue (STOP-CTX) or continue (CTX) cotrimoxazole prophylaxis for 12 months. The sub-study was designed to assess whether taking cotrimoxazole increased the risk of selecting for parasites with SP-resistant mutations in HIV-infected individuals. Samples were genotyped by sequencing to assess the prevalence of mutations associated with SP-resistance. We found there was no risk of selecting for parasites with SP-resistance mutations while on cotrimoxazole. In fact, the opposite was true; cotrimoxazole controlled parasites carrying SP-resistance mutations as evident by the gradual increase in the prevalence of parasites with mutant alleles in the STOP-CTX arm and not in the CTX-arm. We concluded that cotrimoxazole remains effective in controlling malaria infection despite of the high prevalence of SP-resistant parasites, and its use does not select for SP mutations.
Collapse
Affiliation(s)
- Dennis W. Juma
- Department of Emerging and Infectious Diseases (DEID), United States Army Medical Research Directorate-Africa (USAMRD-A), Kenya Medical Research Institute (KEMRI) Kisumu, Kenya
| | - Peninah Muiruri
- Department of Emerging and Infectious Diseases (DEID), United States Army Medical Research Directorate-Africa (USAMRD-A), Kenya Medical Research Institute (KEMRI) Kisumu, Kenya
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | - Krista Yuhas
- Department of Global Health, University of Washington, Seattle, Washington United States of America
| | - Grace John-Stewart
- Departments of Global Health, Medicine, Epidemiology, and Pediatrics, University of Washington, Seattle, Washington, United States of America
| | - Ronald Ottichilo
- Department of Emerging and Infectious Diseases (DEID), United States Army Medical Research Directorate-Africa (USAMRD-A), Kenya Medical Research Institute (KEMRI) Kisumu, Kenya
| | - John Waitumbi
- Department of Emerging and Infectious Diseases (DEID), United States Army Medical Research Directorate-Africa (USAMRD-A), Kenya Medical Research Institute (KEMRI) Kisumu, Kenya
| | - Benson Singa
- Centre for Clinical Research, KEMRI, Nairobi, Kenya
| | - Christina Polyak
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States of America
| | - Edwin Kamau
- Department of Emerging and Infectious Diseases (DEID), United States Army Medical Research Directorate-Africa (USAMRD-A), Kenya Medical Research Institute (KEMRI) Kisumu, Kenya
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
- * E-mail:
| |
Collapse
|
43
|
Ewing AC, Davis NL, Kayira D, Hosseinipour MC, van der Horst C, Jamieson DJ, Kourtis AP. Prescription of Antibacterial Drugs for HIV-Exposed, Uninfected Infants, Malawi, 2004-2010. Emerg Infect Dis 2018; 25. [PMID: 30561313 PMCID: PMC6302572 DOI: 10.3201/eid2501.180782] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Antimicrobial drug resistance is a serious health hazard driven by overuse. Administration of antimicrobial drugs to HIV-exposed, uninfected infants, a population that is growing and at high risk for infection, is poorly studied. We therefore analyzed factors associated with antibacterial drug administration to HIV-exposed, uninfected infants during their first year of life. Our study population was 2,152 HIV-exposed, uninfected infants enrolled in the Breastfeeding, Antiretrovirals and Nutrition study in Lilongwe, Malawi, during 2004-2010. All infants were breastfed through 28 weeks of age. Antibacterial drugs were prescribed frequently (to 80% of infants), and most (67%) of the 5,329 prescriptions were for respiratory indications. Most commonly prescribed were penicillins (43%) and sulfonamides (23%). Factors associated with lower hazard for antibacterial drug prescription included receipt of cotrimoxazole preventive therapy, receipt of antiretroviral drugs, and increased age. Thus, cotrimoxazole preventive therapy may lead to fewer prescriptions for antibacterial drugs for these infants.
Collapse
|
44
|
Williams PCM, Berkley JA. Guidelines for the treatment of severe acute malnutrition: a systematic review of the evidence for antimicrobial therapy. Paediatr Int Child Health 2018; 38:S32-S49. [PMID: 29790840 PMCID: PMC5972636 DOI: 10.1080/20469047.2017.1409453] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 11/04/2017] [Indexed: 12/31/2022]
Abstract
Background Severe acute malnutrition (SAM) affects nearly 20 million children worldwide and is responsible for up to 1 million deaths per year in children under the age of 5 years. Current WHO guidelines recommend oral amoxicillin for children with uncomplicated malnutrition and parenteral benzylpenicillin and gentamicin for those with complicated malnutrition. Because of cost pressures and increasing antimicrobial resistance, the administration of empirical antibiotics for children with SAM has recently been debated. Methods A systematic review of the current published literature was undertaken to assess the efficacy, safety, cost-effectiveness and pharmacokinetics of antimicrobial treatment of children with SAM in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses. Results The initial search found 712 papers, eight of which met the inclusion criteria. Quality assessment of the studies was performed as per the Grading of Recommendations Assessment, Development and Evaluation guidelines. International guidelines and clinical data registries were also reviewed which identified inconsistencies in current first- and second-line therapies and dosing regimens. Conclusion Current evidence supports the continued use of broad-spectrum oral amoxicillin for treating children with uncomplicated SAM as outpatients. There is no strong evidence to justify changing the current parenteral therapy guidelines for children admitted with complicated SAM, although they should be clarified to harmonise the dosage regimen of amoxicillin for the treatment of SAM to 40 mg/kg twice daily, and to continue parenteral antimicrobials beyond 2 days if indicated by the clinical condition.
Collapse
Affiliation(s)
| | - James A. Berkley
- Kenya Medical Research Institute (KEMRI)/Wellcome Trust Research Programme, Kilifi, Kenya
- The Childhood Acute Illness and Nutrition Network (CHAIN), Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
45
|
Achieving sustainable development goals for HIV/AIDS in the Republic of the Congo - Progress, obstacles and challenges in HIV/AIDS health services. Int J Infect Dis 2018; 77:107-112. [PMID: 30342250 DOI: 10.1016/j.ijid.2018.10.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 10/11/2018] [Accepted: 10/11/2018] [Indexed: 11/22/2022] Open
Abstract
The HIV epidemic continues to be a major global public health issue. Since 2012, there has been a paucity of information from the Republic of the Congo on HIV incidence and prevalence rates, national HIV programme effectiveness, highly active antiretroviral therapy (HAART) rollout, patient adherence to treatment, operational and basic science research studies on HIV/AIDS, and donor funding and its impact on the country. A review of the existing literature on HIV in the Republic of the Congo was conducted, focused on prevalence trends, effectiveness of the current national HIV programme, HAART rollout, patient adherence to antiretrovirals (ARVs), resistance to ARVs, the cost of treatment, and operational issues affecting HIV/AIDS programmes in the country. In light of the findings, several important priority areas for scaling-up HIV/AIDS services, programmatic and research activities in the Republic of the Congo are highlighted.
Collapse
|
46
|
Boettiger DC, Law MG, Sohn AH, Davies MA, Wools-Kaloustian K, Leroy V, Yotebieng M, Vinikoor M, Vreeman R, Amorissani-Folquet M, Edmonds A, Fatti G, Batte J, Renner L, Adedimeji A, Kariminia A. Temporal Trends in Co-trimoxazole Use Among Children on Antiretroviral Therapy and the Impact of Co-trimoxazole on Mortality Rates in Children Without Severe Immunodeficiency. J Pediatric Infect Dis Soc 2018; 8:450-460. [PMID: 30215763 PMCID: PMC6831936 DOI: 10.1093/jpids/piy087] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 08/10/2018] [Indexed: 11/15/2022]
Abstract
BACKGROUND Co-trimoxazole is recommended for all children with human immunodeficiency virus. In this analysis, we evaluate trends in pediatric co-trimoxazole use and survival on co-trimoxazole in children using antiretroviral therapy (ART). METHODS We used data collected between January 1, 2006, and March 31, 2016, from the International Epidemiology Databases to Evaluate AIDS. Logistic regression was used to evaluate factors associated with using co-trimoxazole at ART initiation. Competing risk regression was used to assess factors associated with death. RESULTS A total of 54113 children were included in this study. The prevalence of co-trimoxazole use at ART initiation increased from 66.5% in 2006 to a peak of 85.6% in 2010 and then declined to 48.5% in 2015-2016. A similar trend was observed among children who started ART with severe immunodeficiency. After adjusting for year of ART initiation, younger age (odds ratio [OR], 1.18 for <1 vs 1 to <5 years of age [95% confidence interval (CI), 1.09-1.28]), lower height-for-age z score (OR, 1.15 for less than -3 vs greater than -2 [95% CI, 1.08-1.22]), anemia (OR, 1.08 [95% CI, 1.02-1.15]), severe immunodeficiency (OR, 1.25 [95% CI, 1.18-1.32]), and receiving care in East Africa (OR, 8.97 vs Southern Africa [95% CI, 8.17-9.85]) were associated with a high prevalence of co-trimoxazole use. Survival did not differ according to co-trimoxazole use in children without severe immunodeficiency (hazard ratio, 1.01 for nonusers versus users [95% CI, 0.77-1.34]). CONCLUSIONS Recent declines in co-trimoxazole use may not be linked to the current shift toward early ART initiation. Randomized trial data might be needed to establish the survival benefit of co-trimoxazole in children without severe immunodeficiency.
Collapse
Affiliation(s)
- David C Boettiger
- The Kirby Institute, University of New South Wales, Sydney, Australia,Correspondence: D. C. Boettiger, Wallace Wurth Building, UNSW, Sydney, NSW 2252, Australia ()
| | - Matthew G Law
- The Kirby Institute, University of New South Wales, Sydney, Australia
| | - Annette H Sohn
- TREAT Asia/amfAR–Foundation for AIDS Research, Bangkok, Thailand
| | - Mary-Ann Davies
- Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, South Africa
| | | | - Valeriane Leroy
- INSERM, Laboratoire d’Epidémiologie et Analyses en Santé Publique (LEASP)–UMR 1027, Toulouse, France
| | | | - Michael Vinikoor
- Department of Medicine, University of North Carolina at Chapel Hill,Centre for Infectious Disease Research in Zambia, Lusaka
| | | | | | - Andrew Edmonds
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill
| | - Geoffrey Fatti
- Kheth’Impilo AIDS Free Living, Cape Town, South Africa,Division of Epidemiology and Biostatistics, Department of Global Health, Faculty of Medicine and Health Sciences, Stellenbosch University, South Africa
| | | | - Lorna Renner
- Department of Paediatrics, Korlebu Hospital, Accra, Ghana
| | - Adebola Adedimeji
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| | - Azar Kariminia
- The Kirby Institute, University of New South Wales, Sydney, Australia
| | | |
Collapse
|
47
|
Muanda FT, Sheehy O, Bérard A. Use of trimethoprim-sulfamethoxazole during pregnancy and risk of spontaneous abortion: a nested case control study. Br J Clin Pharmacol 2018; 84:1198-1205. [PMID: 29424001 PMCID: PMC5980587 DOI: 10.1111/bcp.13542] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 01/21/2018] [Accepted: 01/25/2018] [Indexed: 01/20/2023] Open
Abstract
AIMS Data available on the fetal safety of trimethoprim-sulfamethoxazole (TMP-SMX) exposure during pregnancy remains scarce and inconclusive. A previous study assessing the link between TMP-SMX exposure during pregnancy and the risk of spontaneous abortion (SA) did not control for protopathic bias and indication bias. METHODS We conducted a nested control study (n = 77 429 pregnancies including 7039 cases of SA and 70 390 controls) within the Quebec Pregnancy Cohort. For each case of SA, we selected 10 controls at the index date that were matched on gestational age and year of pregnancy. TMP-SMX exposure was defined as either having filled at least one prescription between the first day of gestation (1DG) and the index date, or as having filled a prescription before pregnancy but with a duration overlapping the 1DG (102 pregnancies exposed to TMP-SMX, including 25 cases of SA and 77 controls). RESULTS Adjusting for potential confounders, TMP-SMX exposure was associated with an increased risk of SA (AOR 2.94, 95% C 1.89-4.57, 25 exposed cases). Similar results were found after controlling for indication bias and protopathic bias. CONCLUSION Given that this drug is widely use in HIV patients to prevent opportunistic infections and malaria, there is an urgent need to identify potential data sources in Africa for analysis of early pregnancy exposure to TMP-SMX.
Collapse
Affiliation(s)
- Flory T. Muanda
- Faculty of PharmacyUniversity of Montreal2900 Edouard MontpetitMontréalQuébecCanadaH3T 1J4
- Research CenterCHU Sainte‐Justine3175, Côte‐Sainte‐CatherineMontréalQuébecCanadaH3T 1C5
| | - Odile Sheehy
- Research CenterCHU Sainte‐Justine3175, Côte‐Sainte‐CatherineMontréalQuébecCanadaH3T 1C5
| | - Anick Bérard
- Faculty of PharmacyUniversity of Montreal2900 Edouard MontpetitMontréalQuébecCanadaH3T 1J4
- Research CenterCHU Sainte‐Justine3175, Côte‐Sainte‐CatherineMontréalQuébecCanadaH3T 1C5
| |
Collapse
|
48
|
Evans C, Prendergast AJ. Co-trimoxazole for HIV-exposed uninfected infants. LANCET GLOBAL HEALTH 2018; 5:e468-e469. [PMID: 28395832 DOI: 10.1016/s2214-109x(17)30147-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 03/20/2017] [Indexed: 11/16/2022]
Affiliation(s)
- Ceri Evans
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London E1 2AT, UK; Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Andrew J Prendergast
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London E1 2AT, UK; Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe; Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
49
|
Abstract
Antibiotic therapy is not necessary for acute diarrhea in children, as rehydration is the key treatment and symptoms resolve generally without specific therapy. Searching for the etiology of gastroenteritis is not usually needed; however, it may be necessary if antimicrobial treatment is considered. The latter is left to the physician evaluation in the absence of clear indications. Antimicrobial treatment should be considered in severely sick children, in those who have chronic conditions or specific risk factors or in specific settings. Traveler’s diarrhea, prolonged diarrhea, and antibiotic-associated diarrhea may also require antibiotic therapy. Depending on the severity of symptoms or based on risk of spreading, empiric therapy may be started while awaiting the results of microbiological investigations. The choice of antibiotic depends on suspected agents, host conditions, and local epidemiology. In most cases, empiric therapy should be started while awaiting such results. Empiric therapy may be started with oral co-trimoxazole or metronidazole, but in severe cases parenteral treatment with ceftriaxone or ciprofloxacin might be considered.
Collapse
Affiliation(s)
- Eugenia Bruzzese
- Department of Translational Medical Sciences-Section of Pediatrics, University of Naples Federico II, Via S. Pansini 5, Naples, 80131, Italy
| | - Antonietta Giannattasio
- Department of Translational Medical Sciences-Section of Pediatrics, University of Naples Federico II, Via S. Pansini 5, Naples, 80131, Italy
| | - Alfredo Guarino
- Department of Translational Medical Sciences-Section of Pediatrics, University of Naples Federico II, Via S. Pansini 5, Naples, 80131, Italy
| |
Collapse
|
50
|
Yu L, Li W, Zhang M, Cui Y, Chen X, Ni J, Yu L, Shang F, Xue T. Autoinducer2 affects trimethoprim-sulfamethoxazole susceptibility in avian pathogenic Escherichia coli dependent on the folate synthesis-associate pathway. Microbiologyopen 2018; 7:e00582. [PMID: 29423970 PMCID: PMC6079169 DOI: 10.1002/mbo3.582] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 12/13/2017] [Accepted: 12/18/2017] [Indexed: 01/01/2023] Open
Abstract
Avian pathogenic Escherichia coli (APEC) causes airsacculitis, polyserositis, septicemia, and other mainly extraintestinal diseases in chickens, ducks, geese, pigeons, and other avian species, and is responsible for great economic losses in the avian industry. The autoinducer 2 (AI‐2) quorum sensing system is widely present in many species of gram‐negative and gram‐positive bacteria and has been proposed to be involved in interspecies communication. In clinical APEC strains, whether or not AI‐2 affects the expression of antibiotic‐related genes has not been reported. In this study, we have reported that exogenous AI‐2 increase the susceptibility of APEC strains to trimethoprim‐sulfamethoxazole (SXT) in a folate synthesis‐dependent pathway but not in the LsrR‐dependent manner. Our results further explained that exogenous AI‐2 can down regulate the transcription of the folate synthetase encoding genes folA and folC, and the folate synthesis‐related genes luxS, metE, and metH. Gel shift assays confirmed that LsrR, the AI‐2 receptor, did not bind to the promoters of folA and folC, suggesting that exogenous AI‐2 might influence folate metabolism by a feedback inhibition effect but not in the LsrR‐dependent pathway. This study might provide further information in the search for potential drug targets for prophylaxis of avian colibacillosis and for auxiliary antibiotics in the treatment of avian colibacillosis.
Collapse
Affiliation(s)
- Lumin Yu
- School of Life SciencesAnhui Agricultural UniversityHefeiAnhuiChina
| | - Wenchang Li
- School of Life SciencesAnhui Agricultural UniversityHefeiAnhuiChina
| | - Ming Zhang
- School of Life SciencesAnhui Agricultural UniversityHefeiAnhuiChina
| | - Yunmei Cui
- School of SciencesAnhui Agricultural UniversityHefeiAnhuiChina
| | - Xiaolin Chen
- School of Life SciencesAnhui Agricultural UniversityHefeiAnhuiChina
| | - Jingtian Ni
- School of Life SciencesAnhui Agricultural UniversityHefeiAnhuiChina
| | - Li Yu
- Department of Microbiology and ParasitologyAnhui Key Laboratory of ZoonosesAnhui Medical UniversityHefeiChina
| | - Fei Shang
- School of Life SciencesAnhui Agricultural UniversityHefeiAnhuiChina
| | - Ting Xue
- School of Life SciencesAnhui Agricultural UniversityHefeiAnhuiChina
| |
Collapse
|