Skip to main content

Immediate versus deferred antiretroviral therapy in HIV-infected patients presenting with acute AIDS-defining events (toxoplasmosis, Pneumocystis jirovecii-pneumonia): a prospective, randomized, open-label multicenter study (IDEAL-study)

Abstract

Background

To evaluate clinical outcomes after either immediate or deferred initiation of antiretroviral therapy in HIV-1-infected patients, presenting late with pneumocystis pneumonia (PCP) or toxoplasma encephalitis (TE).

Methods

Phase IV, multicenter, prospective, randomized open-label clinical trial. Patients were randomized into an immediate therapy arm (starting antiretroviral therapy (ART) within 7 days after initiation of OI treatment) versus a deferred arm (starting ART after completing the OI-therapy). All patients were followed for 24 weeks. The rates of clinical progression (death, new or relapsing opportunistic infections (OI) and other grade 4 clinical endpoints) were compared, using a combined primary endpoint. Secondary endpoints were hospitalization rates after completion of OI treatment, incidence of immune reconstitution inflammatory syndrome (IRIS), virologic and immunological outcome, adherence to proteinase-inhibitor based antiretroviral therapy (ART) protocol and quality of life.

Results

61 patients (11 patients suffering TE, 50 with PCP) were enrolled. No differences between the two therapy groups in all examined primary and secondary endpoints could be identified: immunological and virologic outcome was similar in both groups, there was no significant difference in the incidence of IRIS (11 and 10 cases), furthermore 9 events (combined endpoint of death, new/relapsing OI and grade 4 events) occurred in each group.

Conclusions

In summary, this study supports the notion that immediate initiation of ART with a ritonavir-boosted proteinase-inhibitor and two nucleoside reverse transcriptase inhibitors is safe and has no negative effects on incidence of disease progression or IRIS, nor on immunological and virologic outcomes or on quality of life.

Introduction

Among the approximately 36.7 million people infected with the human immunodeficiency virus (HIV) worldwide [1], the population of patients presenting late, at advanced disease stages, is still a significant and challenging medical issue. Current evidence points to high rates of late presenters across Europe—between 15 and 63.6% of all new HIV cases [2,3,4,5,6,7,8,9,10]. Moreover, the proportion of late presenters is increasing or at best stagnant in many countries, despite several attempts to encourage earlier diagnosis [5, 11,12,13,14,15].

Late diagnosis of HIV infection often only follows after patients present with opportunistic infections (OI) requiring immediate treatment, a situation which poses numerous management dilemmas and risks due to overlapping drug toxicities, potential pharmacokinetic interactions with antiretroviral drugs and the risk of developing immune reconstitution inflammatory syndrome (IRIS) [14, 16,17,18,19,20,21]. There has been much debate about the optimal timing of antiretroviral therapy (ART) initiation in ART naïve patients presenting with an AIDS-defining event although clinical trials are lacking. Some recent studies suggest that a delay of initiation may be associated with poorer outcomes and more rapid HIV disease progression [14, 19, 22,23,24,25,26,27]. However, there is conflicting evidence in favor for a deferred initiation of ART in patients diagnosed with cerebral tuberculosis and cryptococcal meningitis [28, 29].

Late presenting patients with OIs are underrepresented or excluded in most ART therapy-studies and a limited number of studies address the optimal timing of ART initiation during the course of treatment of an OI. An active AIDS-defining illness is almost always among the exclusion criteria. This situation has resulted in a striking lack of evidence concerning the antiretroviral treatment of such patients. Consequently, the best antiretroviral regimen and the best timing for starting antiretroviral therapy in treatment-naive patients with advanced infection have not yet been established [14] and guidelines for specific antiretroviral treatment for late-presenting patients are lacking [22].

The objective of the current study was to compare the rates of clinical progression in early versus deferred initiation of ART in two study groups with treatment naïve patients, presenting with pneumocystis pneumonia (PCP) and toxoplasmic encephalitis (TE). The ART consisted of atazanavir/ritonavir (ATV/r) and tenofovir-disoproxil fumarate (TDF)/emtricitabine (FTC).

Methods

Trial design

This study (NCT 01417949, clinicaltrials.gov/ct2/show/NCT01417949, registered August 16th 2011) was planned and conducted as a phase IV, multicenter, prospective, randomized open-label clinical trial. A biostatistician was involved in planning of the study. Recruitment was conducted in 16 specialized infectious diseases care clinics in Germany, recruitment period was September 2011 until May 2015. All feasible patients in these centers were screened for inclusion.

Subjects

All patients gave written informed consent. Inclusion criteria were defined as HIV-1-infected adults and either ART-naïve or patients with no antiretroviral therapy for at least 6 months prior to screening and no evidence for prior virologic failure due to resistance against nucleoside reverse transcriptase inhibitors (NRTIs) and protease inhibitors (PIs), who have developed either pneumocystis pneumonia (PCP) or toxoplasma encephalitis (TE). Exclusion criteria included renal failure (defined as CrCl < 60 mL/min), patients who were unable to initiate ART or with current contraindications against ATV/r, AIDS-defining events other than PCP or TE, pregnancy or women of childbearing potential wanting to become pregnant. After statistical calculations we planned 210 patients to include into the study to gain statistically significant evidence. The sample size calculation was carried out with Pass 2008 (Power Analysis and Sample Size Software; NCSS, LLC. Kaysville, Utah, USA). The underlying assumption was a reduction of the primary event rate from 25 to 10% with a statistical power of 80% and a type I error of 5% within a balanced study design. In addition, a maximum dropout rate of 7.5% is assumed for the 194 necessary patients. This results in the final total number of 210 subjects.

Study centers

All patients were recruited in an in-hospital setting. All study centers were HIV-care experienced infectious-diseases units of hospitals with associated outpatient care centers in urban settings.

Study medication and schedule

Antiretroviral treatment consisted of Ritonavir boosted Atazanavir (ATV/r) plus 2 nucleosidic non reverse transcriptase inhibitors (NRTIs). The immediate treatment group (IT) started ATV/r + 2 NRTIs immediately after HIV/AIDS diagnosis (as soon as possible but no later than 7 days after initiation of OI treatment). The deferred treatment arm (DT) started ART after completion of OI treatment, which was achieved for PCP at the earliest at day 21, for TE at the earliest at day 28, but no later than 6 weeks after randomization. Patients were followed up for a total of 24 weeks. Bristol-Myers Squibb provided the study sites with Norvir® and Reyataz® for this clinical trial. Randomization was performed by the principal investigator within the electronic case report form (eCRF). After entering the information that the patient had given written informed consent the decision when to start ART (“immediate” or “deferred”) was provided by the e-CRF system. The day of randomization was considered to be baseline in both arms. Follow up period was over 24 weeks (with visits every 4 weeks). The in-patient care not directly related to the study procedures was up to the specific study site (for example the choice of treatment of the OI or the time of discharging the patients). In other words, the decision to discharge a patient was made on clinical grounds. There was a documentation of the hospitalization days (see next section).

Primary and secondary endpoints

The primary endpoint was defined as clinical progression [death, all new or relapsing OI, grade 4 (G4) clinical endpoint] within 24 weeks after inclusion in the study. For G4 events, standardized toxicity grading tables were used (https://rsc.niaid.nih.gov/sites/default/files/table-for-grading-severity-of-adult-pediatric-adverse-events.pdf). For abnormalities not found in the toxicity tables, a grade 4 event was defined as potentially life-threatening (extreme limitation in activity, significant assistance required; significant medical intervention/therapy required, hospitalization or hospice care probable). Patients who dropped out of study observation before the end of week 12 were counted as clinical progression (intention-to-treat analysis). Secondary endpoints were defined as incidence of immune reconstitution inflammatory syndrome (IRIS) within 24 weeks after inclusion, virological outcome (proportion of patients achieving a viral load below 400 or 50 copies/mL at 24 weeks, changes of the antiretroviral regimen (for lack of efficacy or toxicity), parameters for quality of life (SF 36 score at inclusion and at 24 weeks) and adherence of ART (pill count) and the number of hospitalization days.

Statistical considerations

Originally intended statistical method (log-likelihood Chi-square test for primary, Wilcoxon t- and Chi-square test for secondary endpoints) could not be applied every time due to low inclusion of patients (see results). In those cases, Chi-square was replaced by Fisher’s exact test. All analyses were performed using SAS 9.4 (SAS Institute 2016, Cary, NC). All tests were fully functional despite the small study populations.

Ethics statement

All participating sites had local ethical approval due to national laws. All subjects provided written informed consent.

Results

We screened 67 patients overall, 6 patients couldn’t be included (either no informed consent could be obtained due to psychiatric impairment or due to comorbidities and comedication, 4× TE, 2× PCP with severe disease/sepsis). 31 patients were randomized to the immediate arm and 30 patients to the deferred arm, of whom 29/31 and 24/30 completed the follow up period of 24 weeks. Mean age, gender- and race-distribution were comparable in both groups (see Tables 1 and 2 and Fig. 1). Body mass index, blood pressure, pulse- and respiratory rate at baseline and at last follow up (not shown in the tables) showed no significant differences. 11 patients with toxoplasmosis and 50 patients with PCP were included. There were no significant differences in adherence to treatment or treatment interruptions (< 10% of patients had interruptions in treatment). All patients received ATV/r + TDF/FTC (no other NRTI-backbone was chosen in the included study population). In May 2015, the study was terminated due to low recruitment (decision was made during a prolongation period by the principal investigator and the sponsor together).

Table 1 Demographics, clinical baseline characteristics of the study populations
Table 2 Demographics, clinical characteristics and outcome (week 24) of the study populations
Fig. 1
figure 1

Study subject enrollment and discontinuations

Clinical progression (primary endpoint)

No significant differences in clinical progression between groups were detected: 9 of 31 patients in the immediate and 9 of 30 patients in the deferred group (p-value: 0.9340) had all new or relapsing OI or grade 4 events diagnosed. A new or relapsing opportunistic infection occurred in 4 of 31 patients in the immediate arm and in 1 of 30 patients in the deferred arm. In total, seven (7) grade 4 adverse events occurred throughout the observation period: in 3 of 31 immediate group patients and in 4 of 30 deferred group patients. Serious adverse events were reported for either 2 out of 31 immediate group patients and 6 out of 30 deferred group patients dropped-out.

Of the 61 randomized patients, 8 of 11 patients with TE and 10 of 50 patients PCP had an event, however, these were similarly distributed between the IT and DT groups (see Table 2).

Adverse events

Overall, the patients experienced 274 adverse events (AE). 242 (88%) were graded as grade 1 or 2 adverse events. Most prevalent AE were: allergy to antibiotics (12), herpes infection (8× herpes simples, 3× herpes zoster), mucosal candida infection (8), nausea (7), fever (7), respiratory tract infections (bronchitis, pneumonia) (5), cytomegalovirus (CMV) infection (5), diarrhea (5), Kaposi sarcoma (3).

The distribution of grade 1 and 2 adverse events was relatively equal between the study arms. Although not statistically significant, grade 3 and 4 events appeared to be more common in the DT group (see Table 2).

24 AE (8.76%) were assessed as related to study treatment (18 grade 1, 5 grade 2 and 1 grade 4). The dose of study drug was not changed for grade 1 and 2 AEs however, drug treatment was discontinued after the grade 4 AE (the patient was in the DT group). No serious adverse drug reaction (SADR) or suspected unexpected serious adverse reaction (SUSAR) was recorded in either treatment group. The drop-out rate after treatment initiation was 2/31 (6.45%) in the IT group and 6/30 (20%) in the DT group.

In total, 18 SAEs occurred during the clinical trial. Two of them, occurring within the same patient, were reported as fatal (a 35-year-old man in the deferred treatment group, was diagnosed with a T-cell lymphoma and then later died due to septic multiorgan failure).

Secondary endpoints

No significant difference or trends were detectable amongst secondary endpoints. This includes hospitalization days, IRIS incidence, immunological and virologic outcome, the proportion of patients with changes in the antiretroviral regimen or the SF36-scores (Table 2).

Discussion

This trial was conducted to gain more evidence regarding the ideal timing of initiation of ART in late presenting HIV positive patients who present with either PCP oder TE.

The major strength of this study is that it provides data for the important question when to start ART in a very vulnerable group of HIV-infected patients with two common opportunistic infections.

The protease-inhibitor (PI) atazanavir/ritonavir (ATV/r) was chosen as core agent, recommended in several both national and international guidelines at the time of study planning and initiation 2010–2011 (WHO, EACS, CDC) [1, 30, 31]. ATV/r was chosen as a once daily (at this time) state of the art regimen, as it has a higher genetic barrier compared to e.g. efavirenz and causes a rapid decline in viral load [32]. Retrospectively seen, this choice probably contributed to the low recruitment, as possible drug–drug interactions.

In general, the study medication was well tolerated, most adverse events were mild. There were no unsuspected adverse events related to the study medication. Only few adverse events were severe adverse events and only few drop-outs occurred. The total number of these events (drop-out, AE grade 4, SAE) were equally low in both treatment groups, considering the severity of the underlying condition.

A review of the available literature regarding timing of ART in HIV positive patients with opportunistic infections was performed. To our knowledge only one randomized clinical trial, performed by the AIDS Clinical Trial Group (ACTG 5164) seems similar and comparable to our study population and setting [22]. In this trial a total of 282 subjects (39 sites within the United states and Johannesburg, South Africa) were randomized to initiate ART immediately or after OI treatment. The study provided lopinavir/ritonavir [LPV/r], emtricitabine [FTC], tenofovir disoproxil fumarate [TDF], and stavudine [d4T]. However, any antiretroviral agent approved by the FDA for the initial treatment of HIV was allowed. A boosted PI with two NRTIs was used in 89% of subjects in the immediate arm and 85% of subjects in the deferred arm while NNRTI-based regimens with two nRTIs were used in 11% and 16%, respectively.

There was no statistically significant difference in the primary endpoint and both arms achieved similar CD4 T-cell counts and viral load declines by week 24. Importantly, the immediate arm had fewer deaths/AIDS progressions (p = 0.035), longer time to death/AIDS progression (stratified HR = 0.53, p = 0.02), and shorter time to achieving an increase in CD4 T-cell counts to > 100/μL (11.8 versus 4.2 weeks). The authors concluded that, although there was no significant difference between immediate and deferred ART in the primary endpoint (clinical and virologic response), immediate ART reduces death/AIDS progression over 48 weeks. However, there are major differences in the study settings to our trial because different antiretroviral regimens have been used and heterogeneous opportunistic infections were allowed. The initiation of ART in the immediate arm was 12 days, compared to 45 days in the deferred arm. To get better comparability, we’ve chosen a defined ART regimen with boosted Atazanavir as the third agent. This was an appropriate, frequently recommended and used regimen at time of study planning 2010–2011 [30, 33]. We focused on the two most frequently occurring OI in high income countries, which is another strength of this study.

This study shows no evidence that an immediate antiretroviral combination treatment with ATV/r and 2 NRTIs (TDF/FTC) in patients with an acute AIDS-defining opportunistic infection (toxoplasmosis or pneumocystis pneumonia) has any positive or negative effect on the overall outcome of patients with advanced HIV-related immunosuppression (CDC class C). The primary endpoint ‘clinical progression’ (including death, all new or relapsing OI, and other G4 clinical endpoints within 24 weeks) occurred equally in both treatment arms. Secondary endpoints as hospitalization days after completion of OI treatment, incidence of IRIS, virologic outcome at week 24 were also distributed equally in each treatment group. There were also no significant differences in self-reported bodily or psychological well-being using the SF36 body score and SF36 psychological score questionnaire between the two treatment groups.

Other trials with different study populations, mostly in resource poor countries and with different OI (like tuberculosis, cryptococcal meningitis) showed worse outcomes, if ART was initiated sooner, compared to deferred initiation [26, 28, 29].

The overall message regarding those OI (tuberculosis and cryptococcosis) was, not to defer ART at low CD4 cell counts, unless patients are suffering from neurologic affection, especially cryptococcal meningitis [34,35,36,37].

However, some further limitations deserve to be mentioned. Due to the small size of the two treatment groups, minor differences in primary and secondary endpoints of incidence rates of SAE might not have been detectable.

Another limitation is the fact, that nowadays a majority of patients is treated with an integrase inhibitor as the core agent in first-line ART. Data from the IDEAL trial probably cannot be applied for this treatment selection, because the incidence of treatment-associated immunological reconstitution might be higher with integrase inhibitors (INI). Therefore, it would be necessary to run another trial using INI as the core agent.

Furthermore, epidemiology, diagnostics and therapy in both PCP and TE are evolving, possibly leading to limitations in transferring the results to current populations [38, 39].

Despite a multicenter approach with much effort in 16 German HIV care experienced study centers only 61 of the planned 210 patients could get enrolled into the study. For the most part, in our opinion this was caused by the severity of the underlying condition (e.g. neurological alterations due cerebral toxoplasmosis, patients in critical conditions on intensive care units (e.g. patients couldn’t give informed consent in the short time period, which was mandatory for inclusion into the study). Additionally, difficulties occurred due to multiple potential drug interactions due to supportive drugs. E.g. ATV/r and proton pump inhibitors, often used in critical care patients, couldn’t be coadministered. These difficulties should be considered in the planning of potential future early treatment evaluation trials with novel integrase-based HIV regimens, that have fewer side effects.

Conclusion

In summary, this study supports the hypothesis that immediate initiation of antiretroviral therapy with a ritonavir-boosted proteinase-inhibitor and a combination of two nucleoside reverse transcriptase inhibitors in treating the above mentioned OI is save and has no negative effects on incidence of disease progression or immune reconstitution inflammatory syndrome, nor on immunological and virologic outcome or on quality of life.

We have to emphasize the lack of both, other studies in this important field of HIV medicine and long-term observations. For long term analysis, monitoring the study population after several years is planned. Further investigations should investigate other OI and use other, modern ART-regimens.

Availability of data and material

Full data is available.

References

  1. World health organization (WHO). Fact sheet about HIV/AIDS, update November 2017. http://www.who.int/mediacentre/factsheets/fs360/en/. Accessed 06 Jan 2018.

  2. Adler A, Mounier-Jack S, Coker RJ. Late diagnosis of HIV in Europe: definitional and public health challenges. AIDS Care. 2009;21(3):284–93.

    Article  CAS  Google Scholar 

  3. Borghi V, Girardi E, Bellelli S, Angeletti C, Mussini C, Porter K, Esposito R. Late presenters in an HIV surveillance system in Italy during the period 1992-2006. J Acquir Immune Defic Syndr. 2008;49(3):282–6.

    Article  Google Scholar 

  4. Castilla J, Sobrino P, de la Fuente L, Noguer I, Guerra L, Parras F. Late diagnosis of HIV infection in the era of highly active antiretroviral therapy: consequences for AIDS incidence. AIDS. 2002;16:1945–51.

    Article  Google Scholar 

  5. Girardi E, Aloisi MS, Arici C, Pezzotti P, Serraino D, Balzano R, Vigevani G, Alberici F, Ursitti M, D’Alessandro M, d’Arminio Monforte A, Ippolito G, for the IcoNA Behavioural Epidemiology Study Group. Delayed presentation and late testing for HIV: demographic and behavioural risk factors in a multicenter study in Italy. J Acquir Immune Defic Syndr. 2004;36(4):951–9.

    Article  Google Scholar 

  6. Schäfer G, Kreuels B, Schmiedel S, Hertling S, Hüfner A, Degen O, van Lunzen J, Schulze zur Wiesch J. High proportion of HIV late presenters at an academic tertiary care center in northern Germany confirms the results of several cohorts in Germany. Infection. 2016;44:347–52.

    Article  Google Scholar 

  7. Wolbers M, Bucher HC, Furrer H, Rickenbach M, Cavassini M, Weber R, Schmid P, Bernasconi E, Hirschel B, Battegay M, Swiss HIV Cohort Study. Delayed diagnosis of HIV infection and late initiation of antiretroviral therapy in the Swiss HIV Cohort Study. HIV Med. 2008;9(6):397–405.

    Article  CAS  Google Scholar 

  8. Zoufaly A, An der Heiden M, Marcus U, Hoffmann C, Stellbrink H, Voll L, van Lunzen J, Hamouda O, ClinSurv Study Group. Late presentation for HIV diagnosis and care in Germany. HIV Med. 2012;12(3):172–81.

    Article  Google Scholar 

  9. Brännström J, Akerlund B, Arneborn M, Blaxhult A, Giesecke J. Patients unaware of their HIV infection until AIDS diagnosis in Sweden 1996–2002—a remaining problem in the highly active antiretroviral therapy era. Int J STD AIDS. 2005;16(10):702–6.

    Article  Google Scholar 

  10. Mussini C, Manzardo C, Johnson M, et al. Patients presenting with AIDS in the HAART era: a collaborative cohort analysis. AIDS. 2008;22:2461–9.

    Article  Google Scholar 

  11. Girardi E, Sabin CA, Monforte AD. Late diagnosis of HIV infection: epidemiological features, consequences and strategies to encourage earlier testing. J Acquir Immune Defic Syndr. 2007;46(Suppl 1):S3–8.

    Article  Google Scholar 

  12. Stöhr W, Dunn D, Porter K, Hill T, Gazzard B, Walsh J, Gilson R, Easterbrook P, Fisher M, Johnson M, Delpech V, Phillips A, Sabin C, UK CHIC Study. CD4 cell count and initiation of antiretroviral therapy: trends in seven UK centres, 1997–2003. HIV Med. 2007;8(3):135–41.

    Article  Google Scholar 

  13. Antinori A, Coenen T, Costagiola D, European Late Presenter Consensus Working Group, et al. Late presentation of HIV infection: a consensus definition. HIV Med. 2011;12(1):61–4.

    Article  CAS  Google Scholar 

  14. Manzardo C, Zaccarelli M, Agüero F, Antinori A, Miró JM. Optimal timing and best antiretroviral regimen in treatment-naïve HIV-infected individuals with advanced disease. J Acquir Immune Defic Syndr. 2007;46(Suppl 1):S9–18.

    Article  CAS  Google Scholar 

  15. Deen L, Cowan S, Wejse C, Petersen JH, Norredam M, ESCMID Study Group for Infections in Travellers and Migrants. Refugees and family-reunified immigrants have a high incidence of HIV diagnosis and late presentation compared with Danish born: a nationwide register-based cohort study. Infection. 2018;46(5):659–67.

    Article  Google Scholar 

  16. Elston JWT, Thacker H. Immune reconstitution inflammatory syndrome. Int J STD AIDS. 2009;20:221–4.

    Article  CAS  Google Scholar 

  17. Hocking JS, Rodger AJ, Rhodes DG, Crofts N. Late presentation of HIV infection associated with prolonged survival following AIDS diagnosis: characteristics of individuals. Int J STD AIDS. 2000;11:503–8.

    Article  CAS  Google Scholar 

  18. International Network for the Study of HIV-associated IRIS (INSHI). Definition IRIS. http://www.inshi.umn.edu/definitions/General_IRIS/home.html.

  19. Grant PM, Komarow L, Andersen J, et al. Risk factor analyses for immune reconstitution inflammatory syndrome in a randomized study of early vs. deferred ART during an opportunistic infection. PLoS ONE. 2010;5(7):e11416.

    Article  Google Scholar 

  20. Murdoch DM, Suchard MS, Venter WDF, Mhlangu P, Ottinger JS, Feldman C. Polychromatic immunophenotypic characterization of T cell profiles among HIV-infected patients experiencing immune reconstitution inflammatory syndrome (IRIS). AIDS Res Ther. 2009;6:16.

    Article  Google Scholar 

  21. Roberston J, Meier M, Wall J, Ying J, Fichtenbaum CJ. Immune reconstitution syndrome in HIV: validating a case definition and identifying clinical predictors in persons initiating antiretroviral therapy. Clin Infect Dis. 2006;42:1639–46.

    Article  Google Scholar 

  22. Zolopa A, Andersen J, Powderly W, et al. Early antiretroviral therapy reduces AIDS progression/death in individuals with acute opportunistic infections: a multicenter randomized strategy trial. PLoS ONE. 2009;46(Suppl):S9–18.

    Google Scholar 

  23. Battegay M, Fehr J, Flückiger U, Elzi L. Antiretroviral therapy of late presenters with advanced HIV disease. J Antimicrob Chemother. 2008;62(1):41–4.

    Article  CAS  Google Scholar 

  24. Manzardo C, Esteve A, Ortega N, et al. Optimal timing for initiation of highly active antiretroviral therapy in treatment-naïve HIV-I-infected individuals presenting with AIDS-defining diseases: the experience of the PISCIS Cohort. Clin Microb Inf. 2013;19:646–53.

    Article  CAS  Google Scholar 

  25. Lawn SD, Török ME, Wood R. Optimal time to start antiretroviral therapy during HIV-associated opportunistic infections. Curr Opin Infect Dis. 2011;24(1):34–42.

    Article  Google Scholar 

  26. Abdool Karim SS, Naidoo K, Grobler A, et al. Timing of initiation of antiretroviral drugs during tuberculosis therapy. N Engl J Med. 2010;362(8):697–706.

    Article  CAS  Google Scholar 

  27. The HIV-CAUSAL-Collaboration, Lodi S, del Amo J, et al. Opportunistic infections and AIDS malignancies early after initiation combination antiretroviral therapy in high-income countries. AIDS. 2014;28:2461–73.

    PubMed Central  Google Scholar 

  28. Boulware DR, Meya DB, Muzoora C, et al. Timing of antiretroviral therapy after diagnosis of cryptococcal meningitis. N Engl J Med. 2014;370:2487–98.

    Article  Google Scholar 

  29. Njei B, Kongnyuy EJ, Kumar S, et al. Optimal timing for antiretroviral therapy initiation in patients with HIV infection and concurrent cryptococcal meningitis. Cochrane Database Syst Rev. 2013. https://doi.org/10.1002/14651858.CD009012.pub2.

    Article  PubMed  Google Scholar 

  30. European AIDS Clinical Society (EACS). Guidelines for the clinical management and treatment of HIV-infected adults in Europe. 2011. http://www.eacsociety.org/files/2011_eacsguidelines-v6.0-english_oct.pdf.

  31. Services, DHHS: US Department of Health and Human. Guidelines for the clinical management and treatment of HIV-infected adults in Europe. 2011. https://aidsinfo.nih.gov/contentiles/AdultandAdolescentGL002111.pdf.

  32. Molina JM, Andrade-Villanueva J, et al. Once-daily atazanavir/ritonavir compared with twice-daily lopinavir/ritonavir, each in combination with tenofovir and emtricitabine, for management of antiretroviral naive HIV-1-infected patients: 96 week efficacy and safety results from the CASTLE study. J AIDS. 2010;53:323–32.

    CAS  Google Scholar 

  33. Malan DR, Krantz E, David N, Wirtz V, Hammond J, McGrath D. Efficacy and safety of atazanavir, with or without ritonavir, as part of once-daily highly active antiretroviral therapy regimens in antiretroviral-naive patients. J AIDS. 2008;47:161–7.

    CAS  Google Scholar 

  34. Havlir DV, Kendall MA, Ive P, et al. Timing of antiretroviral therapy for HIV-1 infection and tuberculosis. N Engl J Med. 2011;365:1482–91.

    Article  CAS  Google Scholar 

  35. Blanc F-X, Sok T, Laureillard D, et al. Earlier versus later start of antiretroviral therapy in HIV-infected adults with tuberculosis. N Engl J Med. 2011;365:1471–81.

    Article  CAS  Google Scholar 

  36. Török ME, Farrar JJ. When to start antiretroviral therapy in HIV-associated tuberculosis. N Engl J Med. 2011;365:1538–40.

    Article  Google Scholar 

  37. Eshun-Wilson I, Okwen MP, Richardson M, Bicanic T. Early versus delayed antiretroviral treatment in HIV-positive people with cryptococcal meningitis. Cochrane Database Syst Rev. 2018. https://doi.org/10.1002/14651858.CD009012.pub3.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Rostami A, Karanis P, Fallahi S. Advances in serological, imaging techniques and molecular diagnosis of Toxoplasma gondii infection. Infection. 2018;46(3):303–15.

    Article  CAS  Google Scholar 

  39. Salzer HJF, Schäfer G, Hoenigl M, Günther G, Hoffmann C, Kalsdorf B, Alanio A, Lange C. Clinical, diagnostic, and treatment disparities between HIV-infected and non-HIV-infected immunocompromised patients with Pneumocystis jirovecii pneumonia. Respiration. 2018;96:52–65.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This study was investigator initiated (University Hospital Hamburg-Eppendorf, Infectious Diseases Unit). The study was financially supported with a total of 660,000 Euro by Bristol-Myers-Squibb (BMS), BMS sponsored also Atazanvir/Ritonvir (Reyataz) as part of the study medication.

Funding

The study wasn’t funded, see “Acknowledgements”.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

GS and SS compiled the manuscript, EF was responsible for statistical analysis, JvL, SS, GS and CH developed the study protocol. All other authors contributed to the analysis and interpretation of data for the work, made important revisions. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Guido Schäfer.

Ethics declarations

Ethics approval and consent to participate

All participating sites had local ethical approval according to national laws. All subjects provided written informed consent.

Consent for publication

All mentioned authors gave their consent for publication.

Competing interests

Johannes R. Bogner received honorary for advisory boards and lecturing from AbbVie, ViiV, MSD, Gilead, Janssen, BMS, Hexal. Christian Hoffmann has received honoraria for lecturing, advisory boards and travelling grants and research grants from AbbVie, Bristol Myers-Squibb, Boehringer-Ingelheim, Gilead Sciences, HEXAL, Hormosan, Janssen-Cilag, MSD, Teratech and ViiV Healthcare. Gerd Faetkenheuer received lecture fees, honorary for advisory boards, and travel and research grants: Bristol Myers Squibb, Gilead, Merck Sharp & Dohme. Jan van Lunzen has received honoraria for lecturing, advisory boards and travelling grants from AbbVie, Bristol Myers-Squibb, Boehringer-Ingelheim, Gilead Sciences, HEXAL, Janssen-Cilag, MSD, Teratech and ViiV Healthcare. Christoph Stephan received financial support, related to scientific projects by MSD, BMS, AbbVie and received honorary for lectures or scientific advisory boards from: Abbott, BMS, ViiV, Gilead, Janssen, Roche. Juergen Rockstroh has received honoraria for consulting or speaking at educational events from Abbvie, BMS, Gilead, Merck, Janssen and ViiV. Olaf Degen received honoraria for lecturing, advisory boards and travelling grants from ViiV, GSK, Gilead, BMS, Janssen-Cilag. Matthias Stoll received honoraria as advisor, lecturer or funding of studies by Abbott, Abbvie, Boehringer-Ingelheim, Bristol-Myers-Squibb, Gilead, Glaxo-Smith-Kline, Hexal, Hormosan, Janssen-Cilag, Merck, Sharp & Dohme, Novartis, Pfizer, Roche, Tibotec, ViiV, Viro-Pharmaceuticals. All other authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Schäfer, G., Hoffmann, C., Arasteh, K. et al. Immediate versus deferred antiretroviral therapy in HIV-infected patients presenting with acute AIDS-defining events (toxoplasmosis, Pneumocystis jirovecii-pneumonia): a prospective, randomized, open-label multicenter study (IDEAL-study). AIDS Res Ther 16, 34 (2019). https://doi.org/10.1186/s12981-019-0250-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12981-019-0250-2

Keywords