Skip to main content
Erschienen in: Virology Journal 1/2023

Open Access 01.12.2023 | Review

Trends in HIV-1 pretreatment drug resistance and HIV-1 variant dynamics among antiretroviral therapy-naive Ethiopians from 2003 to 2018: a pooled sequence analysis

verfasst von: Mulugeta Kiros, Sirak Biset, Birhane Gebremariam, Gebrehiwet Tesfay Yalew, Woldaregay Erku Abegaz, Alene Geteneh

Erschienen in: Virology Journal | Ausgabe 1/2023

Abstract

Background

Ethiopia is among the highly HIV-affected countries, with reported 12,000 and 12,000 AIDS-related deaths and incidents as per reports from 2021. Although the country has made a promising progress in antiretroviral therapy, recent studies have indicated that pretreatment drug resistance (PDR) is alarmingly increasing, which has become a challenge for the effectiveness of HIV treatment. Epidemiologic data on PDR is necessary to help establish ART regimens with good efficacy. Thus, this systematic review aimed to determine the trend analysis of PDR among ART-naïve individuals along with HIV variant dynamics in Ethiopia.

Method

HIV-1 pol sequences from studies conducted between 2003 and 2018 among ART-naïve Ethiopian individuals were retrieved from GenBank and analyzed for the presence of PDR mutations (PDRM) along with the analysis of HIV-1 variant dynamics. The Calibrated Population Resistance (CPR) tool Version 8.1 and the REGA HIV-1 Subtyping Tool Version 3 were used to determine the PDRM and HIV-1 genetic diversity, respectively.

Result

We identified nine studies and analyzed 1070 retrieved HIV-1 pol sequences in this systematic review. The pooled prevalence of PDR was 4.8% (51/1070), including 1.4% (15/1070), 2.8% (30/1070), and 0.8% (9/1070) for nucleoside reverse transcriptase inhibitor (NRTI), non-NRTI (NNRTI), and protease inhibitor (PI) resistance, respectively. NRTI and NNRTI concurrent PDRM were observed among 0.2% (2/799) of the analyzed sequences. The overall PDR prevalence has been increasing over the years. Though the prevalence of the NNRTI, NRTI, and PI PDR also increased over the years, the NNRTI increment was more pronounced than the others, reaching 7.84% in 2018 from 2.19% in 2003. The majority (97%; 1038/1070) of the genetic diversity was HIV-1 subtype C virus, followed by subtype C’ (2%; 20/1038) and other subtypes (1%; 10/1038).

Conclusions

According to this systematic review, the overall pooled prevalence of PDR is low. Despite the low prevalence, there has been an increasing trend of PDR over the years, which implies the need for routine surveillance of PDRMs along with preventive measures. Hence, this supports the recently endorsed transition of ART regimens from NNRTI to integrase strand transfer inhibitor-based regimens recommended by the WHO. In addition, this finding underscores the need for routine baseline genotypic drug resistance testing for all newly diagnosed HIV-infected patients before initiating treatment to halt the upward trend of PDR.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12985-023-02205-w.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
AIDS
Acquired immunodeficiency syndrome
ART
Antiretroviral treatment
ATV
Atazanavir
AZT
Zidovudine
D4T
Stavudine
EFV
Efavirenz
ETR
Etravirine
FTC
Emtricitabine
HIV
Human immunodeficiency virus
LPV
Lopinavir
NFV
Nelfinavir
NNRTI
Non-nucleoside reverse-transcriptase inhibitor
NRTI
Nucleoside reverse-transcriptase inhibitor
NVP
Nevirapine
PDR
Pretreatment drug resistance
PDRM
Pretreatment drug resistance mutation
PI
Protease inhibitor
PLWH
People living with HIV
PR
Protease
RPV
Rilpivirine
RT
Reverse-transcriptase
SQV
Saquinavir
3TC
Lamivudine
WHO
World health organization

Introduction

Although decades have passed since the start of the pandemic, the Human Immunodeficiency Virus (HIV) remains a major public health threat globally. The COVID-19 pandemic exacerbated the challenge of HIV infection due to overstretched healthcare delivery, competition for resources (including financial and healthcare-providing professionals), and the synergistic effect of co-infection [1]. According to the UNAIDS 2022 report, approximately 38.4 million people were living with HIV (PLWH) worldwide during the year 2021. Among these, the eastern and southern African regions bear the highest burden of the illness, covering 54% (20.6 million) of the global estimate. Similarly, being situated in this region, Ethiopia is among the most highly affected countries, with 12,000 AIDS-related deaths and 12,000 new HIV infections in the same year [2].
Anti-retroviral therapy (ART) has been scaled up globally from 7.8 million in 2010 to 28.7 million people in December 2021, resulting in a reduction of the burden associated with the pandemic [2]. While this is an outstanding progress in terms of scale-up, the development of pretreatment HIV-1 drug resistance (PDR), which comes in parallel with ART, is becoming a challenge for ART effectiveness [3]. ART scale-up in Sub-Saharan Africa, where drug resistance testing is not routinely available, is also similarly associated with an increasing prevalence of PDR [4, 5].
In Ethiopia, the publicly funded ART program was started in 2005, and the number of people using ART has now scaled up from 58,405 in the beginning to around 436,000 in 2017 [69]. Despite this, however, recent reports show that the PDR is increasing [10]. Therefore, epidemiologic data on PDR is crucial to helping establish optimal empiric ART regimens, especially in countries like Ethiopia, where treatment options are limited and HIV drug resistance testing is absent. To the best of our knowledge, no review study shows the trend of PDR among ART-naive HIV-positive individuals in Ethiopia. Hence, this review has examined the trend of PDR among ART-naïve HIV-positive individuals along with the HIV-1 variant dynamics over the years in Ethiopia.

Methods

Sequence data source, selection criteria, and search strategy

The online databases Google Scholar and PubMed were searched for PDR in Ethiopia to identify relevant literature published until March 30, 2023. The search for the literature was done using the following search terms: [(“human immunodeficiency virus” OR HIV OR AIDS OR “acquired immunodeficiency syndrome”) AND (“antiretroviral therapy” OR HAART OR ART) AND (“drug resistance” OR naïve OR "pregnant women" OR antenatal OR subtype OR surveillance) AND (Ethiopia)]. In addition, the International AIDS Society (IAS) Conference, and Conference on Retroviruses and Opportunistic Infections (CROI) databases were also searched for additional articles. Upon duplicate removal, all retrieved publications were reviewed to determine which studies contained data regarding PDR in Ethiopia. All PDR studies that involved ART-naïve HIV-infected individuals aged ≥ 14 years old in Ethiopia were included in this review. We excluded studies conducted in children, those that have incomplete sequences (did not contain complete PR (99 codons) and partial RT (the first 230 codons), and those studies that were done among ART-experienced individuals (Fig. 1).

Sequence analysis

Sequences of the included studies were downloaded from GenBank if they were publicly available and, if they were not found online, study authors were contacted for the respective sequences. Where several sequences were found from the same research participant (for instance, in cohort studies), we only included the sequence from the earliest time point. Accordingly, we included 1070 sequences (from the total of 1226 retrieved sequences) that have full protease and partial RT. Before sequence analysis, quality control of the pooled sequences from these studies was done using the Calibrated Population Resistance (CPR) tool version 8.1, available in the Stanford HIVDB database (http://​cpr.​stanford.​edu/​cpr.​cgi, (accessed on 02 April 2023)). Sequences (156 out of the total 1226 retrieved sequences) with poor quality based on the CPR quality tool were excluded from the analysis.

Genotypic drug resistance, phylogenetic and statistical analysis

Genotypic PDR mutations (PDRMs) were determined using CPR tool version 8.1. (accessed on 05 April 2023) and were characterized according to the sampling year of the retrieved sequence data. We did not include sex and age information in the datasets or perform correlations between PDRMs and sociodemographic factors because most sequence annotations retrieved did not include this information. The HIV-1 subtype determination was done using the REGA HIV subtyping tool version 3.46 (Leuven University, Leuven, Belgium; https://​www.​genomedetective.​com/​app/​typingtool/​hiv, accessed on 04 April 2023). The sequence alignment and maximum likelihood phylogenetic tree construction was made using the Geneious Prime® 2023.0.4 Software (https://​www.​geneious.​com) and was consecutively visualized by Interactive Tree Of Life (iTOL) version 6 (https://​itol.​embl.​de/​). PDR level classification (low: < 5%, moderate: 5–15%, or high: > 15%) was made based on the WHO threshold survey protocol [11].

Results

We first identified 823 research articles and excluded 788 of the studies based on title and abstract review. The rest 25 full-text articles were further reviewed based on the eligibility criteria, which led to the selection of 12 articles for analysis, three of which were removed due to the unavailability of sequence data (Fig. 1).

Prevalence and drug resistance mutation pattern of HIV

The pooled prevalence of PDR was 4.8% (51/1070). Thirty (2.8%; 30/1070) of the sequences harbored NNRTI PDRM, which is the highest proposition, followed by NRTI (1.4%; 15/1070) and PI PDRM (0.8%; 9/1070). NRTI and NNRTI concurrent PDRM were observed among two individuals (0.2%; 2/1070) of the analyzed sequences (Table 1).
Table 1
Proportion of sequences with PDRMs
Resistance category
No. analyzed
No. containing PDRM
%
Sequences with any PDRM
1070
51
4.8
PR sequences with any PI PDRM
1070
9
0.8
RT sequences with any NRTI PDRM
1070
15
1.4
RT sequences with any NNRTI PDRM
1070
30
2.8
RT sequences with any NRTI + any NNRTI PDRM
1070
2
0.2
PDRM pretreatment drug-resistance mutation, PR protease, PI protease inhibitor, RT reverse-transcriptase, NRTI nucleotide reverse-transcriptase inhibitor, NNRTI non-nucleotide reverse-transcriptase inhibitor
The K103N NNRTI-associated mutation was the most prevalent observed in this review (17.6%; 9/51), followed by G190A (13.7%; 7/51), K101E (7.8%; 4/51), Y188C (7.8%; 4/51), and G190S (5.9%; 3/51). Concerning NRTI mutations, the most frequent mutations were: K219Q (9.8%; 5/51), L210W (7.8%; 4/51), T215S (5.9%; 3/51), and T215FIS (3.9%; 2/51). Among the PI-associated major mutations, the I85V mutation was the most prevalent mutation observed in this review (5.9%; 3/51), followed by M46I and F53L, which were detected in two individuals each (3.9%; 2/51) (Fig. 2). Among the individuals who harbored any PDRMs, four harbored more than one NNRTI-associated mutation, while two harbored more than one NRTI-related mutation simultaneously. The remaining 45 individuals only harbored a single PDRM at a time. For an extensive review of all identified PDRMs, see Additional file 1: Table S1.

Magnitude and trend of PDR over time

To assess the trend of PDR over time, we pooled sequences based on the sampling year. Accordingly, we were able to observe an increment in PDR prevalence from low in 2003 (3.3%) to moderate (5.2%) in 2008–2012. This increase continued and reached 9.8% in 2018, which shows an overall increment in PDR prevalence from 2003 to 2018 (Fig. 3). The NNRTI PDRM was the most prevalent mutation observed over the years relative to the NRTI and PI PDRMs. The trend of NNRTI, NRTI, and PI PDRMs has increased over the years, reaching a peak prevalence in 2018; 7.8%, 1.96%, and 1.96%, respectively (Fig. 4).

HIV variant dynamics and transmission cluster characteristics

The REGA HIV subtyping tool indicated that most of the sequences analyzed (97%; 1038/1070) were HIV-1 subtype C virus followed by subtype C’ (2%; 20/1070), and others (1%; 10/1070) (see Fig. 5). Regarding the geographical distribution of the variants in all study areas across the nation, the HIV-1 subtype C virus remains the major clade, accounting for more than 97% of the total sequences. The majority (80%) of the HIV-1 subtype C-like clade was mainly observed in the northwest of the country, while the other subtypes were dispersed across the nation.
To depict transmission clusters, the 1070 sequences, along with other additional reference sequences (A-K and CRFs; Table S2 in Additional file 2) from the Los Alamos HIV sequence database (https://​www.​hiv.​lanl.​gov/​) were aligned and a maximum likelihood (ML) phylogenetic tree was constructed using Geneious Prime® 2023.0.4 Software (https://​www.​geneious.​com). The neighbor-joining method with 1000 bootstrap replicates under Kimura’s 2-parameter correction was employed. Accordingly, we identified 50 transmission clusters in total (Fig. 6). Those transmission clusters that have a bootstrap value ≥ 90% only are shown in the tree below.

Discussion

Currently, PDR has become a challenge to the success of ART, which makes HIV prevention very difficult. According to the WHO's recent report, PDR prevalence is increasing globally [12]. The same is true for low and middle-income countries, which have shown a substantial increment recently [4]. Therefore, determining PDR at the population level is crucial for optimizing patient and population-level treatment outcomes that will consequently help achieve the third WHO/UNAIDS 90–90–90 target. This review provided the first nationally representative description of the PDR trend along with the HIV variant dynamics over the years.
The pooled prevalence of PDR among ART-naïve individuals in Ethiopia in this systematic review was 4.8%, which is low as per the WHO HIV drug resistance classification [11]. This finding is in line with a similar study from China [13, 14], reports from Tanzania [15], and Kenya [16]. It is, however, lower than the rate of PDR reported from South Africa [17], Cameroon [18], Namibia [19], Brazil [20], Italy [21], and England [22].
The NNRTI-associated PDRMs were the most frequently observed mutations in this review, which corroborates observations from different studies across the globe [2328]. This finding supports the recent transition of the ART regimen in Ethiopia from NNRTI to WHO-recommended integrase strand transfer inhibitor-based regimens. The K103N mutation, which is a non-polymorphic mutation that confers a high-level resistance against efavirenz (EFV) and nevirapine (NVP) [29, 30], was found in a high proportion (17.6%; 9/51) from the NNRTI associated PDRMs. This is similar to reports from Malawi [26], Israel [23], Brazil [20], Tanzania [15], Namibia [19], and Italy [21]. The G190A (13.7%; 7/51) mutation, which is equally selected in persons receiving NVP and EFV [31, 32] was also a common NNRTI-associated mutation, followed by the K101E mutation (7.8%; 4/51), Y188C (7.8%; 4/51) and G190S (5.9%; 3/51) (Fig. 2). The K101E and Y188C mutations are associated with reduced susceptibility to NVP, Etravirine (ETR), and Rilpivirine (RPV) [3335], while Y188C and G190S reduce susceptibility to NVP and EFV [33, 3537].
Among the NRTI-associated mutations, K219Q and L210W mutations which are selected by the thymidine analogs zidovudine (AZT) and Stavudine (d4T), were reported in high frequencies (9.8% (5/51) and 7.8% (4/51)), respectively. In addition to these mutations, others like T215S (5.9%; 3/51) and T215FIS (3.9%; 2/51) were also observed in this review, although in relatively lower frequencies. T215S/I mutation is associated with an increased risk of virologic failure associated with AZT or d4T usage, while T215F confers reduced susceptibility to all currently approved NRTIs except emtricitabine (FTC) and lamivudine (3TC) [38]. It is known that AZT and d4T are commonly used in the country similar to other resource-limited settings; hence, the presence of the mentioned mutations underscores the need for routine genotypic resistance testing.
Protease inhibitor (PI) mutations are usually observed at lower frequencies in different studies [2328, 39]. Likewise, the frequency of PI mutations is low (0.8%) in this review. The mutations observed are I85V (5.9%; 3/51), which was the most prevalent PI mutation, followed by M46I (3.9%; 2/51) and F53L (3.9%; 2/51). The I85V is a non-polymorphic mutation selected by Nelfinavir (NFV) and Atazanavir (ATV) [40, 41] while the M46I and F53L are associated with reduced susceptibility to ATV and Lopinavir (LPV) [42, 43]. M46I, a non-active site mutation in HIV-1 protease, has been clinically associated with saquinavir (SQV) resistance in HIV patients [44]. Altogether, the mentioned mutation profile in this review indicates that regimens containing NFV, ATV, and LPV are more likely to be effective relative to their NNRTI and/or NRTI counterparts.
Concerning the PDR prevalence per year, the overall PDR prevalence was observed to increase from 3.3% in 2003 to 9.8% in 2018 (Fig. 3). The accumulation of the PDRMs over time might have led to the upward trend of PDR over the years. In line with this finding, a similar increment has also been reported in other studies from Mozambique [45] and South Africa [17]. In addition, the NNRTI-associated PDRMs also increased over time (from 2.19% in 2003 to 7.84% in 2018) (Fig. 4), which is once again similar to observations from other studies around the world [2328, 39].
The current study confirmed the HIV-1 subtype C virus as the predominant clade in Ethiopia, similar to other eastern and southern African countries like South Africa [46], Mozambique [45], Malawi [26], and Botswana [47]. This class accounts for 97% of the pandemic in the country. Despite being low, the influx of other variants, including HIV-1 subtype C-like (2%, 20/1070), recombinant of C and A1 (0.38%, 4/1070), HIV-1 subtype A (A1) (0.38%, 4/1070), HIV-1 subtype D (0.09%, 1/1070), HIV-1 CRF 02_AG (0.09%, 1/1070), and recombinant of D and A1 (0.09%, 1/1070), has been observed (Fig. 5). The occurrence of these other variants indicates the possible introduction of other HIV-1 subtypes from neighboring countries. Therefore, preventive measures should always be there to prevent the possibility of further introduction of new subtypes. The 50 transmission clusters found in the sequences analyzed suggest that many HIV-positive individuals engage in risky sexual behavior, emphasizing the necessity of a thorough understanding of the transmission dynamics of HIV-1 subtype C in the population. An increased public health intervention program aimed at these people is also necessary to stop such potentially risky behaviors that will negatively affect HIV transmission. The current review has some limitations. Firstly, all data used in the analysis were retrieved from the literature, and hence we were unable to analyze the correlation of drug resistance with demographic and clinical characteristics, as this information was not available for the majority of sequences. Secondly, in the current review, the studies were mainly from the capital city and from the northern part of the country, and hence some regions might have been underrepresented (due to absence of study), which might lower the representativeness of the study.

Conclusions

In this systematic review, we presented temporal trends of HIV-1 PDR among ART-naïve individuals along with HIV-1 variant dynamics in Ethiopia. Despite the decade-long pandemic, the HIV variant dynamics remained the same, with HIV-1 subtype C still accounting for the majority of the clade (> 97%). Our analysis showed that the overall pooled prevalence of PDR is borderline low (4.8%) 15 years after the rollout of ART in Ethiopia. However, the trend has been seen to be increasing over the years, which is concerning and indicates the need for routine surveillance of PDRMs along with consecutive preventive measures in the country. In addition, the observed increasing trend in NNRTI PDR supports the recent transition of the ART regimen from NNRTI to WHO-recommended integrase strand transfer inhibitor-based regimens.

Acknowledgements

We would like to thank the Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, and Aksum University staff for their unreserved support during this manuscript preparation.

Declarations

Not applicable.
All authors have approved the manuscript for publication.

Competing interests

All the authors declare that they have no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat UNAIDS. UNAIDS Global AIDS Update. 2022. UNAIDS. UNAIDS Global AIDS Update. 2022.
3.
Zurück zum Zitat Bertagnolio S, Hermans L, Jordan MR, Avila-Rios S, Iwuji C, Derache A, et al. Clinical impact of pretreatment human immunodeficiency virus drug resistance in people initiating nonnucleoside reverse transcriptase inhibitor-containing antiretroviral therapy: a systematic review and meta-analysis. J Infect Dis. 2020;224:377–88.PubMedCentralCrossRef Bertagnolio S, Hermans L, Jordan MR, Avila-Rios S, Iwuji C, Derache A, et al. Clinical impact of pretreatment human immunodeficiency virus drug resistance in people initiating nonnucleoside reverse transcriptase inhibitor-containing antiretroviral therapy: a systematic review and meta-analysis. J Infect Dis. 2020;224:377–88.PubMedCentralCrossRef
4.
Zurück zum Zitat Gupta RK, Gregson J, Parkin N, Haile-Selassie H, Tanuri A, Andrade Forero L, et al. HIV-1 drug resistance before initiation or re-initiation of first-line antiretroviral therapy in low-income and middle-income countries: a systematic review and meta-regression analysis. Lancet Infect Dis. 2018;18(3):346–55.PubMedPubMedCentralCrossRef Gupta RK, Gregson J, Parkin N, Haile-Selassie H, Tanuri A, Andrade Forero L, et al. HIV-1 drug resistance before initiation or re-initiation of first-line antiretroviral therapy in low-income and middle-income countries: a systematic review and meta-regression analysis. Lancet Infect Dis. 2018;18(3):346–55.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Boerma RS, Boender TS, Sigaloff KC, de Wit TFR, van Hensbroek MB, Ndembi N, et al. High levels of pre-treatment HIV drug resistance and treatment failure in Nigerian children. J Int AIDS Soc. 2016;19(1):21140.PubMedPubMedCentralCrossRef Boerma RS, Boender TS, Sigaloff KC, de Wit TFR, van Hensbroek MB, Ndembi N, et al. High levels of pre-treatment HIV drug resistance and treatment failure in Nigerian children. J Int AIDS Soc. 2016;19(1):21140.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Kloos H, Assefa Y, Adugna A, Mulatu MS, Mariam DH. Utilization of antiretroviral treatment in Ethiopia between February and December 2006: spatial, temporal, and demographic patterns. Int J Health Geogr. 2007;6:45.PubMedPubMedCentralCrossRef Kloos H, Assefa Y, Adugna A, Mulatu MS, Mariam DH. Utilization of antiretroviral treatment in Ethiopia between February and December 2006: spatial, temporal, and demographic patterns. Int J Health Geogr. 2007;6:45.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Federal Ministry of Health (FMOH). National guidelines for comprehensive HIV prevention, care and treatment. 2018. Federal Ministry of Health (FMOH). National guidelines for comprehensive HIV prevention, care and treatment. 2018.
8.
Zurück zum Zitat Fekade D, Weldegebreal T, Teklu AM, Damen M, Abdella S, Baraki N, et al. Predictors of survival among adult Ethiopian patients in the national ART program at Seven University Teaching Hospitals: a prospective cohort study. Ethiop J Health Sci. 2017;27(1):63–71.PubMedPubMedCentralCrossRef Fekade D, Weldegebreal T, Teklu AM, Damen M, Abdella S, Baraki N, et al. Predictors of survival among adult Ethiopian patients in the national ART program at Seven University Teaching Hospitals: a prospective cohort study. Ethiop J Health Sci. 2017;27(1):63–71.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Arimide DA, Abebe A, Kebede Y, Adugna F, Tilahun T, Kassa D, et al. HIV-genetic diversity and drug resistance transmission clusters in Gondar, Northern Ethiopia, 2003–2013. PLoS ONE. 2018;13(10):e0205446.PubMedPubMedCentralCrossRef Arimide DA, Abebe A, Kebede Y, Adugna F, Tilahun T, Kassa D, et al. HIV-genetic diversity and drug resistance transmission clusters in Gondar, Northern Ethiopia, 2003–2013. PLoS ONE. 2018;13(10):e0205446.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Kiros M, Alemayehu DH, Geberekidan E, Mihret A, Maier M, Abegaz WE, et al. Increased HIV-1 pretreatment drug resistance with consistent clade homogeneity among ART-naive HIV-1 infected individuals in Ethiopia. Retrovirology. 2020;17(1):1–10.CrossRef Kiros M, Alemayehu DH, Geberekidan E, Mihret A, Maier M, Abegaz WE, et al. Increased HIV-1 pretreatment drug resistance with consistent clade homogeneity among ART-naive HIV-1 infected individuals in Ethiopia. Retrovirology. 2020;17(1):1–10.CrossRef
11.
Zurück zum Zitat Bennett DE, Myatt M, Bertagnolio S, Sutherland D, Gilks CF. Recommendations for surveillance of transmitted HIV drug resistance in countries scaling up antiretroviral treatment. Antivir Ther. 2008;13:25.PubMedCrossRef Bennett DE, Myatt M, Bertagnolio S, Sutherland D, Gilks CF. Recommendations for surveillance of transmitted HIV drug resistance in countries scaling up antiretroviral treatment. Antivir Ther. 2008;13:25.PubMedCrossRef
12.
Zurück zum Zitat Organization WH. HIV drug resistance report 2021. 2021. Organization WH. HIV drug resistance report 2021. 2021.
13.
Zurück zum Zitat Chen H, Zhou Y, Hao J, Zhang J, Hu J, Song C, et al. Effects of pretreatment HIV drug resistance on the virological response of HIV-infected patients after 3-year antiretroviral therapy. Zhonghua Liuxingbingxue Zazhi. 2022;43(11):1778–83.PubMed Chen H, Zhou Y, Hao J, Zhang J, Hu J, Song C, et al. Effects of pretreatment HIV drug resistance on the virological response of HIV-infected patients after 3-year antiretroviral therapy. Zhonghua Liuxingbingxue Zazhi. 2022;43(11):1778–83.PubMed
14.
Zurück zum Zitat Zuo L, Liu K, Liu H, Hu Y, Zhang Z, Qin J, et al. Trend of HIV-1 drug resistance in China: a systematic review and meta-analysis of data accumulated over 17 years (2001–2017). EClinicalMedicine. 2020;18:100238.PubMedPubMedCentralCrossRef Zuo L, Liu K, Liu H, Hu Y, Zhang Z, Qin J, et al. Trend of HIV-1 drug resistance in China: a systematic review and meta-analysis of data accumulated over 17 years (2001–2017). EClinicalMedicine. 2020;18:100238.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Rudovick L, Brauner JM, Englert J, Seemann C, Plugaru K, Kidenya BR, et al. Prevalence of pretreatment HIV drug resistance in Mwanza, Tanzania. J Antimicrob Chemother. 2018;73(12):3476–81.PubMed Rudovick L, Brauner JM, Englert J, Seemann C, Plugaru K, Kidenya BR, et al. Prevalence of pretreatment HIV drug resistance in Mwanza, Tanzania. J Antimicrob Chemother. 2018;73(12):3476–81.PubMed
16.
Zurück zum Zitat Beck I, Levine M, McGrath C, Bii S, Milne R, Kingoo J. Pre-treatment HIV-drug resistance associated with virologic outcome of first-line NNRTI-antiretroviral therapy: a cohort study in Kenya. EClin Med. 2020;14(18):100239. Beck I, Levine M, McGrath C, Bii S, Milne R, Kingoo J. Pre-treatment HIV-drug resistance associated with virologic outcome of first-line NNRTI-antiretroviral therapy: a cohort study in Kenya. EClin Med. 2020;14(18):100239.
17.
Zurück zum Zitat Chimukangara B, Lessells RJ, Rhee S-Y, Giandhari J, Kharsany AB, Naidoo K, et al. Trends in pretreatment HIV-1 drug resistance in antiretroviral therapy-naive adults in South Africa, 2000–2016: a pooled sequence analysis. EClinical Med. 2019;9:26–34.CrossRef Chimukangara B, Lessells RJ, Rhee S-Y, Giandhari J, Kharsany AB, Naidoo K, et al. Trends in pretreatment HIV-1 drug resistance in antiretroviral therapy-naive adults in South Africa, 2000–2016: a pooled sequence analysis. EClinical Med. 2019;9:26–34.CrossRef
18.
Zurück zum Zitat Fokam J, Takou D, Teto G, Nforbih SE, Kome OP, Santoro MM, et al. Pre-treatment drug resistance and HIV-1 genetic diversity in the rural and urban settings of Northwest-Cameroon. PLoS ONE. 2020;15(7):e0235958.PubMedPubMedCentralCrossRef Fokam J, Takou D, Teto G, Nforbih SE, Kome OP, Santoro MM, et al. Pre-treatment drug resistance and HIV-1 genetic diversity in the rural and urban settings of Northwest-Cameroon. PLoS ONE. 2020;15(7):e0235958.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Taffa N, Roscoe C, Sawadogo S, De Klerk M, Baughman AL, Wolkon A, et al. Pretreatment HIV drug resistance among adults initiating ART in Namibia. J Antimicrob Chemother. 2018;73(11):3137–42.PubMedCrossRef Taffa N, Roscoe C, Sawadogo S, De Klerk M, Baughman AL, Wolkon A, et al. Pretreatment HIV drug resistance among adults initiating ART in Namibia. J Antimicrob Chemother. 2018;73(11):3137–42.PubMedCrossRef
20.
Zurück zum Zitat Bahls LD, Canezin PH, Reiche EMV, Fernandez JCC, Dias JRC, Meneguetti VAF, et al. Moderate prevalence of HIV-1 transmitted drug resistance mutations in southern Brazil. AIDS Res Ther. 2019;16(1):4.PubMedPubMedCentralCrossRef Bahls LD, Canezin PH, Reiche EMV, Fernandez JCC, Dias JRC, Meneguetti VAF, et al. Moderate prevalence of HIV-1 transmitted drug resistance mutations in southern Brazil. AIDS Res Ther. 2019;16(1):4.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Rossetti B, Di Giambenedetto S, Torti C, Postorino M, Punzi G, Saladini F, et al. Evolution of transmitted HIV-1 drug resistance and viral subtypes circulation in Italy from 2006 to 2016. HIV Med. 2018;19(9):619–28.PubMedCrossRef Rossetti B, Di Giambenedetto S, Torti C, Postorino M, Punzi G, Saladini F, et al. Evolution of transmitted HIV-1 drug resistance and viral subtypes circulation in Italy from 2006 to 2016. HIV Med. 2018;19(9):619–28.PubMedCrossRef
22.
Zurück zum Zitat Tostevin A, White E, Dunn D, Croxford S, Delpech V, Williams I, et al. Recent trends and patterns in HIV-1 transmitted drug resistance in the United Kingdom. HIV Med. 2017;18(3):204–13.PubMedCrossRef Tostevin A, White E, Dunn D, Croxford S, Delpech V, Williams I, et al. Recent trends and patterns in HIV-1 transmitted drug resistance in the United Kingdom. HIV Med. 2017;18(3):204–13.PubMedCrossRef
23.
Zurück zum Zitat Wagner T, Zuckerman NS, Halperin T, Chemtob D, Levy I, Elbirt D, et al. Epidemiology and transmitted HIV-1 drug resistance among treatment-naïve individuals in Israel, 2010–2018. Viruses. 2021;14(1):71.PubMedPubMedCentralCrossRef Wagner T, Zuckerman NS, Halperin T, Chemtob D, Levy I, Elbirt D, et al. Epidemiology and transmitted HIV-1 drug resistance among treatment-naïve individuals in Israel, 2010–2018. Viruses. 2021;14(1):71.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Pang X, Tang K, He Q, Huang J, Fang N, Zhou X, et al. HIV drug resistance and HIV transmission risk factors among newly diagnosed individuals in Southwest China. BMC Infect Dis. 2021;21(1):160.PubMedPubMedCentralCrossRef Pang X, Tang K, He Q, Huang J, Fang N, Zhou X, et al. HIV drug resistance and HIV transmission risk factors among newly diagnosed individuals in Southwest China. BMC Infect Dis. 2021;21(1):160.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Lan Y, Li L, He X, Hu F, Deng X, Cai W, et al. Transmitted drug resistance and transmission clusters among HIV-1 treatment-naïve patients in Guangdong, China: a cross-sectional study. Virol J. 2021;18(1):181.PubMedPubMedCentralCrossRef Lan Y, Li L, He X, Hu F, Deng X, Cai W, et al. Transmitted drug resistance and transmission clusters among HIV-1 treatment-naïve patients in Guangdong, China: a cross-sectional study. Virol J. 2021;18(1):181.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Neuhann F, de Forest A, Heger E, Nhlema A, Scheller C, Kaiser R, et al. Pretreatment resistance mutations and treatment outcomes in adults living with HIV-1: a cohort study in urban Malawi. AIDS Res Ther. 2020;17(1):1–7.CrossRef Neuhann F, de Forest A, Heger E, Nhlema A, Scheller C, Kaiser R, et al. Pretreatment resistance mutations and treatment outcomes in adults living with HIV-1: a cohort study in urban Malawi. AIDS Res Ther. 2020;17(1):1–7.CrossRef
27.
Zurück zum Zitat Matías-Florentino M, Chaillon A, Ávila-Ríos S, Mehta SR, Paz-Juárez HE, Becerril-Rodríguez MA, et al. Pretreatment HIV drug resistance spread within transmission clusters in Mexico City. J Antimicrob Chemother. 2020;75(3):656–67.PubMedCrossRef Matías-Florentino M, Chaillon A, Ávila-Ríos S, Mehta SR, Paz-Juárez HE, Becerril-Rodríguez MA, et al. Pretreatment HIV drug resistance spread within transmission clusters in Mexico City. J Antimicrob Chemother. 2020;75(3):656–67.PubMedCrossRef
28.
Zurück zum Zitat Sebastião CS, Neto Z, de Jesus CS, Mirandela M, Jandondo D, Couto-Fernandez JC, et al. Genetic diversity and drug resistance of HIV-1 among infected pregnant women newly diagnosed in Luanda, Angola. PLoS ONE. 2019;14(11):e0225251.PubMedPubMedCentralCrossRef Sebastião CS, Neto Z, de Jesus CS, Mirandela M, Jandondo D, Couto-Fernandez JC, et al. Genetic diversity and drug resistance of HIV-1 among infected pregnant women newly diagnosed in Luanda, Angola. PLoS ONE. 2019;14(11):e0225251.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Margot N, Lu B, Cheng A, Miller M, Team S. Resistance development over 144 weeks in treatment-naive patients receiving tenofovir disoproxil fumarate or stavudine with lamivudine and efavirenz in Study 903. HIV Med. 2006;7(7):442–50.CrossRef Margot N, Lu B, Cheng A, Miller M, Team S. Resistance development over 144 weeks in treatment-naive patients receiving tenofovir disoproxil fumarate or stavudine with lamivudine and efavirenz in Study 903. HIV Med. 2006;7(7):442–50.CrossRef
30.
Zurück zum Zitat Rhee S-Y, Varghese V, Holmes SP, Van Zyl GU, Steegen K, Boyd MA, et al. Mutational correlates of virological failure in individuals receiving a WHO-recommended tenofovir-containing first-line regimen: an international collaboration. EBioMed. 2017;18:225–35.CrossRef Rhee S-Y, Varghese V, Holmes SP, Van Zyl GU, Steegen K, Boyd MA, et al. Mutational correlates of virological failure in individuals receiving a WHO-recommended tenofovir-containing first-line regimen: an international collaboration. EBioMed. 2017;18:225–35.CrossRef
31.
Zurück zum Zitat Gulick RM, Ribaudo HJ, Shikuma CM, Lustgarten S, Squires KE, Meyer WA III, et al. Triple-nucleoside regimens versus efavirenz-containing regimens for the initial treatment of HIV-1 infection. N Engl J Med. 2004;350(18):1850–61.PubMedCrossRef Gulick RM, Ribaudo HJ, Shikuma CM, Lustgarten S, Squires KE, Meyer WA III, et al. Triple-nucleoside regimens versus efavirenz-containing regimens for the initial treatment of HIV-1 infection. N Engl J Med. 2004;350(18):1850–61.PubMedCrossRef
32.
Zurück zum Zitat Bacheler L, Jeffrey S, Hanna G, D’Aquila R, Wallace L, Logue K, et al. Genotypic correlates of phenotypic resistance to efavirenz in virus isolates from patients failing nonnucleoside reverse transcriptase inhibitor therapy. J Virol. 2001;75(11):4999–5008.PubMedPubMedCentralCrossRef Bacheler L, Jeffrey S, Hanna G, D’Aquila R, Wallace L, Logue K, et al. Genotypic correlates of phenotypic resistance to efavirenz in virus isolates from patients failing nonnucleoside reverse transcriptase inhibitor therapy. J Virol. 2001;75(11):4999–5008.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Melikian GL, Rhee S-Y, Varghese V, Porter D, White K, Taylor J, et al. Non-nucleoside reverse transcriptase inhibitor (NNRTI) cross-resistance: implications for preclinical evaluation of novel NNRTIs and clinical genotypic resistance testing. J Antimicrob Chemother. 2014;69(1):12–20.PubMedCrossRef Melikian GL, Rhee S-Y, Varghese V, Porter D, White K, Taylor J, et al. Non-nucleoside reverse transcriptase inhibitor (NNRTI) cross-resistance: implications for preclinical evaluation of novel NNRTIs and clinical genotypic resistance testing. J Antimicrob Chemother. 2014;69(1):12–20.PubMedCrossRef
34.
Zurück zum Zitat Rimsky L, Vingerhoets J, Van Eygen V, Eron J, Clotet B, Hoogstoel A, et al. Genotypic and phenotypic characterization of HIV-1 isolates obtained from patients on rilpivirine therapy experiencing virologic failure in the phase 3 ECHO and THRIVE studies: 48-week analysis. JAIDS J Acquir Immune Defic Syndr. 2012;59(1):39–46.PubMedCrossRef Rimsky L, Vingerhoets J, Van Eygen V, Eron J, Clotet B, Hoogstoel A, et al. Genotypic and phenotypic characterization of HIV-1 isolates obtained from patients on rilpivirine therapy experiencing virologic failure in the phase 3 ECHO and THRIVE studies: 48-week analysis. JAIDS J Acquir Immune Defic Syndr. 2012;59(1):39–46.PubMedCrossRef
35.
Zurück zum Zitat Lai M-T, Feng M, Falgueyret J-P, Tawa P, Witmer M, DiStefano D, et al. In vitro characterization of MK-1439, a novel HIV-1 nonnucleoside reverse transcriptase inhibitor. Antimicrob Agents Chemother. 2014;58(3):1652–63.PubMedPubMedCentralCrossRef Lai M-T, Feng M, Falgueyret J-P, Tawa P, Witmer M, DiStefano D, et al. In vitro characterization of MK-1439, a novel HIV-1 nonnucleoside reverse transcriptase inhibitor. Antimicrob Agents Chemother. 2014;58(3):1652–63.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Rhee S-Y, Liu T, Ravela J, Gonzales MJ, Shafer RW. Distribution of human immunodeficiency virus type 1 protease and reverse transcriptase mutation patterns in 4,183 persons undergoing genotypic resistance testing. Antimicrob Agents Chemother. 2004;48(8):3122–6.PubMedPubMedCentralCrossRef Rhee S-Y, Liu T, Ravela J, Gonzales MJ, Shafer RW. Distribution of human immunodeficiency virus type 1 protease and reverse transcriptase mutation patterns in 4,183 persons undergoing genotypic resistance testing. Antimicrob Agents Chemother. 2004;48(8):3122–6.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Huang W, Gamarnik A, Limoli K, Petropoulos CJ, Whitcomb JM. Amino acid substitutions at position 190 of human immunodeficiency virus type 1 reverse transcriptase increase susceptibility to delavirdine and impair virus replication. J Virol. 2003;77(2):1512–23.PubMedPubMedCentralCrossRef Huang W, Gamarnik A, Limoli K, Petropoulos CJ, Whitcomb JM. Amino acid substitutions at position 190 of human immunodeficiency virus type 1 reverse transcriptase increase susceptibility to delavirdine and impair virus replication. J Virol. 2003;77(2):1512–23.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Wensing A, Calvez V, Ceccherini-Silberstein F, Charpentier C, Günthard H, Paredes R, et al. 2022 update of the drug resistance mutations in HIV-1. Top Antivir Med. 2023;30(4):559–74. Wensing A, Calvez V, Ceccherini-Silberstein F, Charpentier C, Günthard H, Paredes R, et al. 2022 update of the drug resistance mutations in HIV-1. Top Antivir Med. 2023;30(4):559–74.
39.
Zurück zum Zitat Appah A, Beelen CJ, Kirkby D, Dong W, Shahid A, Foley B, et al. Molecular epidemiology of HIV-1 in Ghana: subtype distribution, drug resistance and coreceptor usage. Viruses. 2022;15(1):128.PubMedPubMedCentralCrossRef Appah A, Beelen CJ, Kirkby D, Dong W, Shahid A, Foley B, et al. Molecular epidemiology of HIV-1 in Ghana: subtype distribution, drug resistance and coreceptor usage. Viruses. 2022;15(1):128.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat de Carvalho Lima EN, Lima RSA, Piqueira JRC, Sucupira MC, Camargo M, Galinskas J, et al. Evidence of genomic information and structural restrictions of HIV-1 PR and RT gene regions from individuals experiencing antiretroviral virologic failure. Infect Genet Evol. 2020;78:104134.PubMedCrossRef de Carvalho Lima EN, Lima RSA, Piqueira JRC, Sucupira MC, Camargo M, Galinskas J, et al. Evidence of genomic information and structural restrictions of HIV-1 PR and RT gene regions from individuals experiencing antiretroviral virologic failure. Infect Genet Evol. 2020;78:104134.PubMedCrossRef
41.
Zurück zum Zitat Rhee S-Y, Boehm M, Tarasova O, Di Teodoro G, Abecasis AB, Sönnerborg A, et al. Spectrum of atazanavir-selected protease inhibitor-resistance mutations. Pathogens. 2022;11(5):546.PubMedPubMedCentralCrossRef Rhee S-Y, Boehm M, Tarasova O, Di Teodoro G, Abecasis AB, Sönnerborg A, et al. Spectrum of atazanavir-selected protease inhibitor-resistance mutations. Pathogens. 2022;11(5):546.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Vermeiren H, Van Craenenbroeck E, Alen P, Bacheler L, Picchio G, Lecocq P, et al. Prediction of HIV-1 drug susceptibility phenotype from the viral genotype using linear regression modeling. J Virol Methods. 2007;145(1):47–55.PubMedCrossRef Vermeiren H, Van Craenenbroeck E, Alen P, Bacheler L, Picchio G, Lecocq P, et al. Prediction of HIV-1 drug susceptibility phenotype from the viral genotype using linear regression modeling. J Virol Methods. 2007;145(1):47–55.PubMedCrossRef
43.
Zurück zum Zitat Rhee S-Y, Taylor J, Fessel WJ, Kaufman D, Towner W, Troia P, et al. HIV-1 protease mutations and protease inhibitor cross-resistance. Antimicrob Agents Chemother. 2010;54(10):4253–61.PubMedPubMedCentralCrossRef Rhee S-Y, Taylor J, Fessel WJ, Kaufman D, Towner W, Troia P, et al. HIV-1 protease mutations and protease inhibitor cross-resistance. Antimicrob Agents Chemother. 2010;54(10):4253–61.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Rana N, Singh AK, Shuaib M, Gupta S, Habiballah MM, Alkhanani MF, et al. Drug resistance mechanism of M46I-mutation-induced saquinavir resistance in HIV-1 protease using molecular dynamics simulation and binding energy calculation. Viruses. 2022;14(4):697.PubMedPubMedCentralCrossRef Rana N, Singh AK, Shuaib M, Gupta S, Habiballah MM, Alkhanani MF, et al. Drug resistance mechanism of M46I-mutation-induced saquinavir resistance in HIV-1 protease using molecular dynamics simulation and binding energy calculation. Viruses. 2022;14(4):697.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Ismael N, Wilkinson E, Mahumane I, Gemusse H, Giandhari J, Bauhofer A, et al. Molecular epidemiology and trends in HIV-1 transmitted drug resistance in mozambique 1999–2018. Viruses. 2022;14(9):1992.PubMedPubMedCentralCrossRef Ismael N, Wilkinson E, Mahumane I, Gemusse H, Giandhari J, Bauhofer A, et al. Molecular epidemiology and trends in HIV-1 transmitted drug resistance in mozambique 1999–2018. Viruses. 2022;14(9):1992.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Steegen K, Carmona S, Bronze M, Papathanasopoulos MA, van Zyl G, Goedhals D, et al. Moderate levels of pre-treatment HIV-1 antiretroviral drug resistance detected in the first south African national survey. PLoS ONE. 2016;11(12):e0166305.PubMedPubMedCentralCrossRef Steegen K, Carmona S, Bronze M, Papathanasopoulos MA, van Zyl G, Goedhals D, et al. Moderate levels of pre-treatment HIV-1 antiretroviral drug resistance detected in the first south African national survey. PLoS ONE. 2016;11(12):e0166305.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Seatla KK, Maruapula D, Choga WT, Ntsipe T, Mathiba N, Mogwele M, et al. HIV-1 Subtype C drug resistance mutations in heavily treated patients failing integrase strand transfer inhibitor-based regimens in Botswana. Viruses. 2021;13(4):594.PubMedPubMedCentralCrossRef Seatla KK, Maruapula D, Choga WT, Ntsipe T, Mathiba N, Mogwele M, et al. HIV-1 Subtype C drug resistance mutations in heavily treated patients failing integrase strand transfer inhibitor-based regimens in Botswana. Viruses. 2021;13(4):594.PubMedPubMedCentralCrossRef
Metadaten
Titel
Trends in HIV-1 pretreatment drug resistance and HIV-1 variant dynamics among antiretroviral therapy-naive Ethiopians from 2003 to 2018: a pooled sequence analysis
verfasst von
Mulugeta Kiros
Sirak Biset
Birhane Gebremariam
Gebrehiwet Tesfay Yalew
Woldaregay Erku Abegaz
Alene Geteneh
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
Virology Journal / Ausgabe 1/2023
Elektronische ISSN: 1743-422X
DOI
https://doi.org/10.1186/s12985-023-02205-w

Weitere Artikel der Ausgabe 1/2023

Virology Journal 1/2023 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Perioperative Checkpointhemmer-Therapie verbessert NSCLC-Prognose

28.05.2024 NSCLC Nachrichten

Eine perioperative Therapie mit Nivolumab reduziert das Risiko für Rezidive und Todesfälle bei operablem NSCLC im Vergleich zu einer alleinigen neoadjuvanten Chemotherapie um über 40%. Darauf deuten die Resultate der Phase-3-Studie CheckMate 77T.

Positiver FIT: Die Ursache liegt nicht immer im Dickdarm

27.05.2024 Blut im Stuhl Nachrichten

Immunchemischer Stuhltest positiv, Koloskopie negativ – in solchen Fällen kann die Blutungsquelle auch weiter proximal sitzen. Ein Forschungsteam hat nachgesehen, wie häufig und in welchen Lokalisationen das der Fall ist.

GLP-1-Agonisten können Fortschreiten diabetischer Retinopathie begünstigen

24.05.2024 Diabetische Retinopathie Nachrichten

Möglicherweise hängt es von der Art der Diabetesmedikamente ab, wie hoch das Risiko der Betroffenen ist, dass sich sehkraftgefährdende Komplikationen verschlimmern.

Update Innere Medizin

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