Skip to main content

Telmisartan versus EnalapRil In heart failure with redUced ejection fraction patients with Moderately impaired kidney Functions; randomized controlled trial: “TRIUMF trial

Abstract

Background

When heart failure with reduced ejection fraction (HFrEF) and chronic kidney disease (CKD) co-exist, Renin angiotensin-aldosterone system inhibitors (RAASi) are often underutilized for the fear of worsening renal function (WRF). Telmisartan is a RAASi characteristic for a favorable renal profile, although data on its utility in HFrEF is limited. This study aimed to compare efficacy and tolerability of Telmisartan versus Enalapril in patients with HFrEF and CKD.

Results

This study randomized 107 patients with HFrEF and CKD to either Telmisartan (10–80 mg) or Enalapril (5–40 mg) daily. The achieved RAASi dose, dose reductions (DR) or dis-continuation (DC), death/Heart failure rehospitalization (HFH), NYHA class and 6MWT were compared at 3- and 6-months. At 3- and 6-months, 93.5% versus 68.6% and 95.2% versus 72.9% were maintaining ≥ 50% of the target dose in the Telmisartan- versus Enalapril-group, respectively. Despite the higher achieved dose by 3- and 6-months, Telmisartan versus Enalapril was associated with less WRF (6.4% vs. 22.9%, p = 0.022 and 7.3% vs. 13.6%, p = 0.28) and fewer episodes of DR-DC (31.9% vs. 55.1%, p = 0.018 and 35.7% vs. 56.5%, p = 0.041), respectively. By the end of the study, there were 5 deaths in each group, yet, HFH occurred in 34.1% versus 55.3%, p = 0.035, and NYHA class changed by − 1 [− 2, 0] versus 0 [− 1, 1], p = 0.017 in Telmisartan- versus Enalapril patients, respectively. Within-group results showed improvement in 6MWT in Telmisartan-, and increase in diuretic requirements in Enalapril-group.

Conclusions

In patients with HFrEF and CKD, Telmisartan was better tolerated to uptitrate, caused less WRF, less HFH and showed better functional improvement compared to Enalapril.

Clinical trial registration This study was prospectively registered on clinicaltrials.gov, with registration number (NCT04736329).

Background

Heart failure (HF) represents a global public health burden. It affects an estimated population of 64 million people worldwide and is associated with significant morbidity and mortality [1, 2]. The HF syndrome results in decreased cardiac output with subsequent activation of neuro-hormonal compensatory mechanisms, mainly the Renin Angiotensin Aldosterone System (RAAS) and the Sympathetic Nervous System (SNS) [3]. Despite initial usefulness, uncontrolled RAAS activation enhances several deleterious effects including renal sodium reabsorption, fluid retention, worsening congestion, arteriolar vasoconstriction, catecholamines release with overstimulation of the SNS, adverse cardiac remodeling and myocardial fibrosis, all ending into significant worsening of HF prognosis [3, 4]. Hence, RAAS inhibitors (RAASi) are fundamental in the medical management of HF with reduced ejection fraction (HFrEF) [1, 2].

Worsening renal function (WRF) and hyperkalemia are frequent drivers for under prescribing RAASi or failing to reach the target doses. This issue is particularly relevant when HFrEF patients have concomitant chronic kidney disease (CKD) [5]. HF and CKD share several common risk factors and therefore commonly co-exist, with one of them begetting, complicating and exacerbating the other. It is estimated that ≥ 50% of chronic HF patients had estimated Glomerular Filtration Rate (eGFR) of < 60 ml/min/1.73 m2, conversely, cardiovascular disease (CVD) remains the leading cause of death in CKD patients [6, 7]. Nevertheless, for the fear of WRF and hyperkalemia, a large proportion of HFrEF patients with CKD are deprived from the cardioprotective and the renoprotective long-term benefits of RAASi [5].

Telmisartan is an angiotensin receptor blocker (ARB), characterized by being primarily excreted via hepatic rather than renal pathway. Provisional experiences with Telmisartan from prior studies suggest better tolerability compared to other RAASi in the management of CVD in CKD patients [8,9,10].

On these grounds, this study was designed to evaluate the efficacy, safety and tolerability of Telmisartan in HFrEF patients with concomitant moderate renal dysfunction, compared to the standard Angiotensin converting enzyme inhibitor (ACEi) “Enalapril,” in a randomized controlled fashion.

Methods

This was a prospective, open-label, randomized, controlled, trial that was conducted in Cairo University hospitals during the period from February 2021 to October 2022. The study protocol was approved by the research ethics committee, Faculty of Medicine, Cairo University (MS-352-2020).

The study recruited 107 HFrEF patients with concomitant CKD who met the eligibility criteria after providing a written informed consent. Inclusion criteria comprised: (1) chronic HF with New York Heart Association (NYHA) class II, III or IV, with established diagnosis of HFrEF for ≥ 6 months; (2) CKD with established diagnosis for ≥ 3 months with entry eGFR of 40–60 ml/min/1.73 m2; (3) inability to sustain regular Angiotensin Receptor Neprilysin Inhibitor (ARNI) therapy for financial or medical causes; and (4) age between 18-and-80 years. Exclusion criteria included: (1) clinical contraindications to ACEi therapy (i.e., angioneurotic edema on previous exposure or significant bilateral renal artery stenosis); or (2) refusal to participate in the study or withdrawal of the consent at any stage.

In consistence with the clinical guidelines and the universal definition of heart failure, HFrEF was defined as the clinical syndrome of heart failure with stable left ventricular ejection fraction (LVEF) < 40% assessed via transthoracic echocardiography (TTE) by Bi-plane Simpson’s technique [1, 2, 11]. Moderate renal impairment was defined as stable eGFR at the study entry in the range of ≥ 40 to < 60 ml/min/1.73 m2. Estimated GFR was calculated by the Modification of Diet in Renal Disease (MDRD-4) equation with consideration of patients’ age, sex, weight and serum creatinine [12].

Groups allocation

Eligible HFrEF patients were randomized into two equal groups using web-based randomization table and closed envelopes system. Group 1 received Telmisartan (between 10 mg o.d. and − 40 mg b.i.d) as tolerated, while group 2 received Enalapril (between 2.5 mg b.i.d and 20 mg b.i.d) as tolerated. Patients who were already receiving regular RAASi agents (ACEi or ARB) were switched to a starting RAASi dose that is (at least) equivalent to their original agent/dose. Conveniently, total daily doses of 40 mg Enalapril, 80 mg Telmisartan, 10 mg Ramipril or 160 mg Valsartan were considered equivalent to 100% of the recommended target dose, and fractions of these doses were equated accordingly.

Patients who were not on any ACEi or ARBs were started on the smallest dose of the study drug then uptitrated gradually to the recommended or the maximum tolerated dose. Worth mentioning that ability to sustain stable ARNI therapy was re-evaluated during recruitment, and if presumed possible, was considered an exclusion criterion to recruitment. Also, for lack of solid evidence about the maximum Telmisartan dose in HFrEF; in patients with blood pressure (BP) persistently > 140/90 mmHg, it was allowed to increase Telmisartan dose to a maximum of 80 mg b.i.d if not limited by hyperkalemia and/or WRF, while still considered as 100% of the target RAASi dose.

Upon recruitment

All the study cohort had their basic medical therapy revised. Apart from RAASi, HFrEF therapies with proven survival benefit (namely, Beta-blockers (BB), Mineralocorticoid antagonist (MRA) and Sodium glucose cotransporter type 2 inhibitors (SGLT2i)) were tabulated indicating prevalence of use and the used agent and dose. For standardization, HFrEF therapies were also expressed as percent from the guidelines recommended target dose, (i.e., Bisoprolol 10 mg, Nebivolol 10 mg, Metoprolol 200 mg or Carvedilol 50 mg represented 100% of the recommended BB daily dose). Similarly, oral loop diuretics were tabulated indicating agent and dose. A standardized loop diuretic dose was calculated in Frusemide equivalent dose (i.e., counting Bumetanide 1 mg, or Torsemide 20 mg equivalent to Frusemide 40 mg).

Study work-up

Baseline clinical, laboratory and echocardiographic data were collected upon patients’ recruitment, then formally repeated at 3-months and 6-months of follow-up. HF-related symptoms were represented in NYHA classification [1]. NYHA class shifts through the various check-points of the study (baseline, at 3- and 6-months) were represented as +ve or −ve scores for deterioration or improvement, respectively, (i.e., improvement from NYHA III to NYHA I is represented as “− 2”, while deterioration from NYHA II to NYHA III is represented as “+ 1”). 

Effort tolerance was assessed semi-quantitatively by performing a six-minute walk test (6MWT) according to the American Thoracic Society instructions [13], utilizing the 25-m department corridor.

Periodic clinical assessment included evaluation of blood pressure (BP) sitting and 3-min after standing, heart rate, signs suggestive of systemic and/or pulmonary congestion. Orthostatic hypotension was reported when there was a drop between sitting and 3-min standing of > 20 or > 10 mmHg in systolic or diastolic BP measurements, respectively [14]. Orthostatic hypotension was considered as a red flag, mandating consideration of RAASi dose reduction (DR) or transient discontinuation (DC) if other causes were excluded.

Besides clinical evaluation, periodic assessments entailed examination of basic laboratory parameters, particularly serum urea and creatinine, serum electrolytes (including sodium, potassium and magnesium). Detailed TTE assessment of left ventricular (LV) dimensions and function was performed at baseline and 6-months according to American society of echocardiography and the European association of cardiovascular imaging consensus for cardiac chamber quantification [15].

Heart failure rehospitalization (HFH) was defined as clinical decompensation of previously stabilized HF judged to indicate intravenous HF therapies. Rehospitalization for reasons other than decompensated HF, was tabulated but not counted in the study end-points. During hospitalizations, kidney function and serum electrolytes' levels were assessed daily or more frequently if clinically indicated, while for out-patients, these were performed with the scheduled clinical visits every 10–15 days.

Management of HFrEF therapies through-out the study

As per study protocol, both groups were equally prescribed all other guidelines directed medical therapy (GDMT) for HFrEF, and doses were periodically evaluated if uptitration was considered safely possible. The ability to maintain guidelines-directed doses of foundational HFrEF therapies (specifically BB and MRA) were reported. Also, clinically-judged doses of loop diuretics (expressed in Frusemide dose equivalent) were updated on every clinical visit.

Concerning RAASi, patients were evaluated periodically to uptitrate the study drug if feasible. Conversely, any mandated DR or transient DC through the study period were meticulously reported as well as their primary reason (i.e., intolerable rise in serum creatinine, profound hyperkalemia, symptomatic or orthostatic hypotension, or signs of hemodynamic instability). The composite of DR-DC events was evaluated as a surrogate to drug tolerance.

Dose management of the randomization drug (Telmisartan vs. Enalapril) with respect to the changes in kidney functions followed the expert opinion statement of the French Society of Cardiology [16]. After meticulous patient assessment, including volume status, BP and potential drug-drug interaction to identify and correct any reversible causes, it was acceptable to presume the same RAASi dose if serum creatinine increased by less than 50% of the baseline value, while the dose was reduced by half (i.e., DR) when the rise was 50-to 100% of the baseline value. However, transient DC was triggered when serum creatinine increasesd by ≥ 100% from baseline, till it wanes off [16]. Another indication for DR or DC was significant hyperkalemia (defined as > 5.5 mEq/L) on 2 subsequent days, as per guidelines’ recommendations [1, 2].

Worsening renal function

Because of lack of standardized definition, WRF had been identified in previous studies when there is absolute increase in serum creatinine from prior assay by ≥ 0.3 mg/d [17, 18], or ≥ 0.5 mg/dL [19, 20], or when there is a relative drop by ≥ 20% in eGFR from baseline [21,22,23]. Data strongly support that in patients with pre-existing CKD, the definition respecting baseline eGFR and considering a relative decline by ≥ 20% as a diagnostic threshold is better correlated with clinical outcomes [21, 22, 24]. Nevertheless, the investigators resorted to evaluate WRF by all the three validated definitions and express them in the results.

The study objectives were to compare Telmisartan versus Enalapril in regard to the following key components:

  1. (a)

    Efficacy

    1. (1)

      Reduction in the composite of all-cause death and HFH,

    2. (2)

      Improvement in NYHA class and 6MWT from baseline;

  2. (b)

    Safety and tolerability

    1. (1)

      Ability to safely maintain > 50% of the recommended RAASi dose,

    2. (2)

      Rates of study-drug DR-DC, as surrogate for drug tolerance,

    3. (3)

      Prevalence of tolerating other foundational HFrEF therapies (mainly MRAs and BB).

Statistical analysis

Data were verified, coded then anonymized by the investigators before it was sent to the statisticians. Statistical analysis was conducted using IBM- Statistical Package for the Social Sciences for windows, version 24.0 (IBM-SPSS Inc., Chicago, IL, USA). Descriptive statistics were represented as means ± standard deviations (SD) or medians and interquartile ranges [IQR] for continuous variables, while as frequencies and percentages for categorical variables. For categorical variables, Chi square or Fisher’s Exact tests were used to compare frequencies and McNemar’s test for repeated measures. While for continuous variables, independent t-test analysis was carried out to compare the means of normally distributed data, while Mann–Whitney was alternatively used in cases on non-normally distributed data. For continuous variables with more than two categories or repeated measures; repeated measure ANOVA (RM-ANOVA) test was calculated to test the mean differences of the data that follow normal distribution and had repeated measures (between groups, within groups and overall difference). A P-value was considered significant when it is less than 0.05.

Results

This prospective randomized controlled trial recruited 107 eligible HFrEF patients with concomitant moderate CKD as per study protocol. Through the 6-months study period, 12 patients were lost from follow-up and 10 patients died. Deceased patients were calculated in the composite clinical end-point of all-cause death and HFH, but were subtracted from the analyses of clinical- (i.e., NYHA class, 6MWT), laboratory- (i.e., eGFR, WRF) or medications data (study-drug dose, DR-DC) next to their death dates. Flow chart for study recruitment process is demonstrated in Fig. 1.

Fig. 1
figure 1

Flow chart for the study recruitment process

Baseline data

Mean age of the study group was 59 ± 9.9 years. Males represented almost 2/3 of the recruited cohort. Baseline features were comparable across the groups except for baseline eGFR which had a small but a significant difference in favor of the Enalapril- over the Telmisartan group.

During recruitment, there were no significant differences between the 2 groups in the standardized doses of diuretics, BB, MRA or SGLT2i. Similarly, the starting doses of Telmisartan and Enalapril were comparable, with median [IQR] of 50% [50–50] of the RAASi target dose in both groups.

Detailed baseline clinical, laboratory and echocardiographic features of the whole study group, and comparison between those randomized to Telmisartan versus Enalapril are demonstrated in Table 1.

Table 1 Baseline clinical, laboratory and echocardiographic parameters of the study groups

By the 3-months check-point

Through the first 3 months of the study, maintaining ≥ 50% of the target RAASi dose in the Telmisartan-compared to the Enalapril-group was achieved in 93.5% versus 68.6%, respectively, (p = 0.002). The median [IQR] of the utilized dose in the Telmisartan versus Enalapril represented 100% [100–100] versus 50% [25–100] of the target dose, respectively (p < 0.001). Additionally, the odds ratio (OR) and 95% confidence interval (CI) for Telmisartan- compared to Enalapril-allocated patients to maintain ≥ 50% of the target dose was 1.31 [1.09–1.57].

Despite the higher achieved RAASi dose, Telmisartan- compared to Enalapril-group had significantly fewer episodes of DR (10.6% vs. 32.7%, p = 0.008) and the composite of DR-DC episodes (31.9% vs. 55.1%, p = 0.018). There were no differences in serum creatinine, serum potassium or eGFR between the 2 groups. WRFGFR (defined as ≥ 20% drop of the eGFR from the prior assessment) occurred significantly less in the Telmisartan- than Enalapril-group (6.4% vs. 22.9%, p = 0.022).

Maintaining BB and MRA therapies at 3-months was nearly similar between the Telmisartan versus the Enalapril group, suggesting acceptable tolerability of other HFrEF foundational therapies. The standardized doses of loop diuretics were numerically higher in the Enalapril- compared to the Telmisartan group, (109 ± 126 vs. 125 ± 113 mg of Frusemide equivalents, p = 0.362).

Concerning the clinical endpoints at the 3-months evaluation, rates of all-cause death, of HFH and of the composite of all-cause death/HFH were comparable between the Telmisartan and the Enalapril, reaching 2 (4%) versus 2 (4%), 9 (18.4%) versus 14 (28%) and 11 (22.4%) versus 14 (28%), respectively.

The median change in NYHA class (expressed as + ve or -ve units for deterioration or improvement, respectively) was comparable. The remainder of clinical data from baseline-to-the 3-months assessment are detailed in Table 2.

Table 2 Data of baseline-to-3-months period

Between 3 and 6 months

By the 6-months evaluation, 95.2% and 72.9% of the Telmisartan- and the Enalapril-patients were maintaining ≥ 50% of the RAASi target dose, p = 0.004, with median [IQR] of the target dose of 100% [100–100] versus 100% [25–100], respectively, (p < 0.001). The OR for Telmisartan- compared to Enalapril-allocated patients to maintain ≥ 50% of the target dose by 6-months was 1.31 [95% CI 1.1–1.6].

DR events occurred numerically fewer in the Telmisartan- compared to the Enalapril group in 23.8% versus 39.1%, respectively, p = 0.094. However, the composite of DR-DC was significantly less in the Telmisartan- compared to the Enalapril-group, occurring in 35.7% versus 56.5%, respectively, p = 0.041. Despite the higher achieved RAASi dose, Telmisartan was associated with almost half the rate of WRFGFR compared to the Enalapril group (7.3% vs. 13.6%), yet without achieving statistical significance.

Rates of new HFH events between 3-to-6 months tended to be fewer in the Telmisartan- compared to Enalapril-group, reaching 7 (15.9%) versus 15 (31.9%), p = 0.061. There were 3 new deaths in each group through that period. The composite of all-cause death/HFH showed a trend for fewer events in the Telmisartan compared to Enalapril, with a rate of 8 (18.2%) versus 16 (34%), p = 0.069. The total (through the whole 6-months period) rate of all-cause death was comparable (5 cases in each group), while HFH and composite of death/HFH were significantly less in Telmisartan- compared to Enalapril-patients with rates of 15 (34.1%) versus 26 (55.3%), p = 0.035 and 18 (36.7%) versus 28 (57.1%), p = 0.034, respectively.

By the end of the study period, Telmisartan-group showed significantly better NYHA class shifts (changes from baseline) compared to Enalapril, with a median change of − 1 [− 2, 0] versus 0 [− 1, 1], p = 0.017. The distribution of NYHA classes was non-significant between the 2 groups at 6-months, however, the within-group changes (comparing baseline-to-the end of the study period) exhibited a favorable re-distribution only in the Telmisartan that seemed mainly derived from the noticeable reduction in NYHA class IV prevalence, [changing from 14.8 to 7.3% vs. 19 to 25% in the Telmisartan vs. the Enalapril-group, respectively].

Similarly, despite the non-significant between-groups difference in the 6MWT, the within-group changes (by repeated measures ANOVA) pointed to a statistically significant improvement in the Telmisartan group but not in the Enalapril group. Furthermore, the standardized doses of loop diuretics showed nearly stable requirements from baseline to the end of the study in the Telmisartan group contrasted to an obvious progressive rise in the Enalapril group through successive check-points that demonstrated significant within-group differences.

The details of the 6-month data are shown in Tables 3 and 4, and Figs. 2 and 3.

Table 3 Data by the 6-months assessment
Table 4 Between-groups and within-group differences in NYHA class, 6MWT and required maintenance loop diuretic doses at baseline and by the end of the study
Fig. 2
figure 2

A Distribution of NYHA class across the Telmisartan- and Enalapril groups at baseline and by the end of the study. B Mann–Whitney testing of the NYHA class shifts through the study period (baseline -to-6 months) expressed as − 1 for each class improvement (i.e., IV-to-III) and + 1 for each class deterioration (i.e., II-to-III)

Fig. 3
figure 3

A Scores of 6MWT for the 2 groups at baseline and by the end of the study with demonstration of the within-group p value. B Daily requirements of the standardized loop diuretics dose from baseline to the end of the study demonstrating changes in dose requirements and the p values of within-group differences between baseline-to-6 months and 3-to-6 months

Discussion

In this randomized controlled study selectively recruiting 107 patients with HFrEF and concomitant CKD who were unable to sustain ARNI therapy, Telmisartan compared to Enalapril was better tolerated, achieved higher percentages of the target dose, was associated with fewer episodes of mandated DR and/or DC, while reducing rates of worsening renal functions. The improved RAAS inhibition in the Telmisartan arm (better tolerability and sustainability), was translated to more favorable changes in the NYHA class and 6MWT and reduction in rehospitalization, compared to the Enalapril-allocated controls.

RAASi are cornerstone in HFrEF management with proven symptoms and survival benefits, hence, are granted class I recommendation in most practice guidelines [1, 2]. In the PARADIGM-HF trial, the ARNI (Sacubitril/Valsartan) showed substantial reduction in the composite of death/rehospitalization when compared to the prototype ACEi, the Enalapril [25]. Since then, ARNI had become the preferred RAASi agent. However, for the significant high monthly cost, sustained ARNI use is underutilized among the indicated cohorts in low-to-mid-income countries, particularly when not fully insured [26]. Moreover, in the PARADIGM-HF trial, after 10,513 initially entered the study, around 20% were filtered out through the roll-in phase mainly because of drug intolerance, ending in 8442 proceeding to the randomization phase [25]. Hence, at least for these 2 main limitations, it is very clear that even in the era of ARNI, the role of conventional and readily affordable RAASi (ACEi/ARBs) in HFrEF management is not over yet.

One of the main identified barriers to RAASi in HFrEF management is the consequent impairment of renal functions, specially in patients with pre-existing renal dysfunction [5]. Acknowledging the intimate overlap of risk factors and injurious agents/conditions for CVD and CKD, one can appreciate the high prevalence of their co-existence, and that one of them can cause and/or exacerbate the other, while the two of them begets worsening of each other [6, 27]. It is well established that CVD is the leading cause of death in CKD patients, while conversely, an eGFR < 60 ml/m/1.73 m2 was prevalent in nearly 60% of HF hospitalizations [7].

Importantly, full benefits of RAASi (including ACEi/ARBs/ARNI) on both CV- and renal-outcomes are conditioned by sustaining the doses identified by clinical trials and recommended by guidelines. This belief is supported by multiple trials and registries where HF patients limited to low-dose RAASi demonstrated significantly poorer outcomes including all-cause mortality, all cause hospitalization, CV hospitalization as well as renal outcomes compared to those receiving higher doses [28,29,30]. In the large CHAMP-HF registry, < 30% of the indicated cohort could achieve the RAASi target dose (including ACEi/ARB/ARNI), while patients sustaining ≥ 50% of the target dose showed significantly better NYHA class and less HFH compared to those receiving lower doses [5].

Hence, from the grounds of registries and daily clinical practice, a large cohort of HFrEF with co-existing CKD are denied the substantial clinical benefit of RAASi. For the fear of WRF, these patients are either completely deprived from-, or at the best, are prescribed low-dose of RAASi [31]. Thereby, a RAASi agent that can be better tolerated in HFrEF with concomitant CKD remains to be a lingering unmet need.

Unlike most ACEi/ARBs that are dependent on renal clearance, Telmisartan is an ARB unique for a predominant hepatic clearance, reducing the risk of drug accumulation or toxicity in patients with CKD [32]. The ONTARGET and TRANSCEND studies demonstrated that Telmisartan is an appealing RAASi in the management of hypertension, CVD and diabetes with end-organ damage, particularly in ACEi intolerant groups [33, 34].

Additionally, Telmisartan was postulated to provide further renal benefits independent form its characteristic predominant hepatic clearance. In a dedicated analysis of the > 25,000 participants in the ONTARGET study, there was significant progressive reduction in proteinuria observed in the Telmisartan arm, with improved renal outcomes by the end of the study [34]. The mechanism of such renoprotective effect is not fully understood, though is suggested to be related to the agonistic effects of Telmisartan on the Peroxisome Proliferator Activated Receptor-Gamma (PPAR-γ) pathway [35].

Although the evidence on Telmisartan in HFrEF remains limited, in the few available studies Telmisartan was found to be at least as good as other “standard” RAASi agents in HF. In a study that had randomized 120 NYHA III patients to receive either Telmisartan or other standard ACEi, besides other GDMT, both arms showed equivalent improvement in LVEF and serum brain natriuretic peptide (BNP) levels after 1-year of therapy [36]. In another double-blinded randomized study recruiting 378 HFrEF patients on stable therapy, both Telmisartan and Enalapril arms showed comparable improvements in effort tolerance, with a numerical reduction in rates of cough complaint in the Telmisartan-arm that did not achieve statistical significance [37]. Notably, these studies were in non-selected HFrEF patients who were able to receive guidelines-recommended doses of RAASi agents, while dedicated studies in the subgroup of HFrEF and concomitant CKD seemed lacking.

In the present study, we have selectively recruited HFrEF patients with concomitant moderate renal impairment who cannot sustain regular ARNI therapy. Moderate renal impairment was defined as entry eGFR of 40–60 ml/min/1.73 m2. Baseline features were comparable between the 2 groups except for a small but significant difference in the eGFR in favor of Enalapril-group. Through the 6-months study period, Telmisartan was significantly better tolerated than Enalapril. By the 3- and 6-months check-points, the median utilized dose in the Telmisartan-group achieved 100% [100–100] and 100% [100–100] of the target dose compared to 50% [25–100] and 100% [25–100] in the Enalapril-group, respectively. Better tolerability was well demonstrated by the proportion of patients sustaining ≥ 50% of the target dose, which was 93.5% versus 68.6% by the 3-months, and 95.2% versus 72.9% by the 6-months check-points, for the Telmisartan versus Enalapril, respectively. A cut-off of ≥ 50% of the target dose was considered after it had shown in the CHAMP-HF registry to be associated with significantly better mid- and long-term clinical benefits compared to lower doses [5].

WRF seemed to lack standardization of a diagnostic threshold, with some definitions utilizing an absolute increase in serum creatinine while others considering a relative reduction in the eGFR. Among the most utilized definitions in HF studies was an absolute increase of ≥ 0.3 mg/dL in serum creatinine level within 5 days, compared to the baseline value [17, 18]. Another definition used alternatively the cut-off of absolute increase by ≥ 0.5 mg/dL as the diagnostic threshold [19, 20]. A third definition was later proposed and proved to be a better prognosticator for patients with already pre-existing renal impairment, and instead of an absolute change in serum creatinine, it identified WRF as a drop by ≥ 20% in eGFR from baseline [21,22,23]. The selective recruitment of patients with renal dysfunction would arguably make the WRF definition relying on a relative drop from the baseline eGFR (WRFGFR) more meaningful [23, 38].

In this study, despite the higher achieved RAASi dose in the Telmisartan-group, the rate of WRFGFR, was significantly less compared to the Enalapril-group. Moreover, the rates of DR and DC were less in the Telmisartan- compared to Enalapril-group, supporting a better tolerability- and safety-profiles of Telmisartan in this selective HFrEF cohort.

Concerning clinical outcomes through the study period, death events were comparable, however, rates of HFH and of the composite of death/HFH were significantly less in the Telmisartan compared to Enalapril. Indeed, the distribution of NYHA classes between the 2 groups did not achieve statistical significance, however, the within-group distribution at 6-months compared to baseline demonstrated a significant change (improvement) in Telmisartan- but not in Enalapril-group. More interestingly, NYHA class shifts t in the Telmisartan versus Enalapril demonstrated significant improvement by the 6-months- compared to baseline-evaluation, with a median change of − 1 [− 2, 0] versus 0 [− 1, 1], p = 0.017. Effort tolerability represented by 6MWT showed similar results to NYHA distribution, with a non-significant between-groups difference, but a significant improvement from baseline to 6-months in the Telmisartan- while not in the Enalapril-group.

Another supportive observation for the favorable clinical outcome was the stable loop diuretic dose in the Telmisartan- in contrast to the progressively increasing requirements in the Enalapril-group. Higher loop diuretic dose was found to be an independent predictor for worse outcomes and increased mortality as demonstrated in a large study recruiting 1354 HFrEF patients, showing adjusted HR of death of 4.0 (95% CI 1.9–8.4) for those requiring > 160 mg compared to those requiring 0-to-40 mg daily equivalent diuretic dose [39].

In fact, there is paucity of RCT-derived evidences on safety and efficacy of RAASi use in patients with advanced kidney disease. According to a recently published state-of-the-art review [40], only one RCT was available evaluating RAASi therapy in HFrEF patients with end-stage kidney disease (ESKD). In that single study, the investigators chose Telmisartan for that selected cohort, and concluded that Telmisartan compared to placebo reduced both; all-cause- and cardiovascular-mortality with HR of 0.51 [0.32–0.82] and 0.42 [0.38–0.61], respectively, with similar rates of DC of the study drug through the ≥ 3-years of the study, supporting an excellent tolerability of Telmisartan in CKD patients [41].

Another important evidence for the utility of Telmisartan in patients with poor tolerability to ACEi comes from a larger (n = 5962) study selectively recruiting patients with cardiovascular disease or diabetes with end-organ damage who are identified as completely ACEi intolerants [8]. These patients were randomized to Telmisartan 80 mg/day versus placebo. After median follow-up of 56 months, compared to placebo, Telmisartan reduced the composite of CV death, myocardial infarction, and stroke with OR 0.87, [95% CI 0.76–1.0] with unadjusted p value = 0.048. Worth mentioning that in that ACEi intolerant selected cohort, DC was comparable in the Telmisartan and placebo groups and had occurred in 21.6% versus 23.8%, respectively [8].

Although evidences are still insufficient to qualify Telmisartan as one of the standard RAASi in HFrEF unselected cohorts, it seems that selectively in HFrEF and CKD patients Telmisartan is better tolerated, leads to fewer WRF, shows less frequent drug interruptions, and achieves higher RAASi doses. Such superior and consistent inhibition of RAAS proved to be paralleled by greater clinical benefits that what can be achieved by low-doses of frequently interrupted other standard ACEi/ARBs.

Limitations

This randomized study has some limitations to be disclosed. The relatively small sample size (107), and the short period of follow-up might have led to underestimation of the endpoints. Also, having an open-label design would allow for potential interpretation bias of the NYHA class. Unfortunately, the logistic setup of the recruiting center could not allow for a double-blinded design, nevertheless, the investigators added the 6MWT as an objective assessment tool to offset such concern. Being self-funded, the available resources did not permit to perform baseline and repeated testing of NT-pro-BNP and albumin/creatinine ratio (ACR) for all patients, while they could have added a critical quantitative prognostic tool to assess study drug performance. A larger, double-blinded, multi-center study, designed with a longer follow-up and entailing BNP, ACR and LV strain assessment in baseline and by the study end is strongly recommended to confirm the findings of this study

Conclusions

In HFrEF patients with concomitant moderate CKD, Telmisartan showed better tolerability-, sustainability- and safety-profiles compared to the conventional RAASi, Enalapril. Because of ability to sustain higher RAASi dose with less frequent WRF and DR/DC, Telmisartan improved NYHA class changes, reduced HFH and improved effort tolerance by 6-months compared to Enalapril.

Availability of data and materials

Data can be provided (anonymized) upon reasonable request sent to the corresponding author.

Abbreviations

6MWT:

Six-minute walk test

ACEi:

Angiotensin converting enzyme inhibitor

ARB:

Angiotensin receptor blocker

ARNI:

Angiotensin receptor neprilysin inhibitor

b.i.d:

Twice daily

BB:

Beta blocker

BNP:

Brain Natriuretic peptide

BP:

Blood pressure

CI:

Confidence interval

CKD:

Chronic kidney disease

CV:

Cardiovascular

CVD:

Cardio-vascular disease

DC:

Dis-continuation

DR:

Dose reduction

eGFR:

Estimated glomerular filtration rate

ESKD:

End-stage kidney disease

GDMT:

Guidelines directed medical therapy

HF:

Heart failure

HFH:

Heart failure re-hospitalization

HFrEF:

Heart failure with reduced ejection fraction

IQR:

Inter-quartile range, denoting the 25th and 75th percentiles

LV:

Left ventricle

LVEF:

Left ventricular ejection fraction

MDRD:

Modification of Diet in Renal Disease

mg:

Milligrams

MRA:

Mineralocorticoid antagonist

NYHA:

New York Heart Association

o.d:

Once daily

OR:

Odds ratio

PPAR-g:

Peroxisome Proliferator Activated Receptor-Gamma

RAAS:

Renin angiotensin aldosterone system

RAASi:

Renin angiotensin aldosterone system inhibitor(s)

SGLT2i:

Sodium Glucose co-Transporter type-2 inhibitor

TTE:

Transthoracic echocardiography

WRF:

Worsening renal function

References

  1. Heidenreich PA, Bozkurt B, Aguilar D, Allen LA, Byun JJ, Colvin MM, Deswal A, Drazner MH, Dunlay SM, Evers LR, Fang JC, Fedson SE, Fonarow GC, Hayek SS, Hernandez AF, Khazanie P, Kittleson MM, Lee CS, Link MS, Milano CA, Nnacheta LC, Sandhu AT, Stevenson LW, Vardeny O, Vest AR, Yancy CW (2022) AHA/ACC/HFSA guideline for the management of heart failure: a report of the American College of Cardiology/American Heart Association Joint Committee on clinical practice guidelines. Circulation 145:895–1032

    Google Scholar 

  2. McDonagh TA, Metra M, Adamo M, Gardner RS, Baumbach A, Böhm M, Burri H, Butler J, Celutkiene J, Chioncel O, Cleland JGF, Coats AJS, Crespo-Leiro MG, Farmakis D, Gilard M, Heymans S (2021) 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure. Eur Heart J 42(36):3599–3726. https://doi.org/10.1093/eurheartj/ehab368/6358045

    Article  CAS  PubMed  Google Scholar 

  3. Schwinger RHG (2021) Pathophysiology of heart failure. Cardiovasc Diagn Ther 11(1):263–276

    Article  PubMed  PubMed Central  Google Scholar 

  4. Oliveros E, Oni ET, Shahzad A, Kluger AY, Lo KB, Rangaswami J, McCullough PA (2020) Benefits and risks of continuing angiotensin-converting enzyme inhibitors, angiotensin II receptor antagonists, and mineralocorticoid receptor antagonists during hospitalizations for acute heart failure. Cardiorenal Med 10(2):69–84

    Article  CAS  PubMed  Google Scholar 

  5. Greene SJ, Butler J, Albert NM, DeVore AD, Sharma PP, Duffy CI, Hill CL, McCague K, Mi X, Patterson JH, Spertus JA, Thomas L, Williams FB, Hernandez AF, Fonarow GC (2018) Medical therapy for heart failure with reduced ejection fraction: the CHAMP-HF registry. J Am Coll Cardiol 72(4):351–366

    Article  PubMed  Google Scholar 

  6. Costanzo MR (2020) The cardiorenal syndrome in heart failure. Heart Fail Clin 16(1):81–97. https://doi.org/10.1016/j.hfc.2019.08.010

    Article  PubMed  Google Scholar 

  7. Rangaswami J, Bhalla V, Blair JEA, Chang TI, Costa S, Lentine KL, Lerma EV, Mezue K, Molitch M, Mullens W, Ronco C, Tang WHW, Mccullough PA (2019) Cardiorenal syndrome: classification, pathophysiology, diagnosis, and treatment strategies: a scientific statement from the American Heart Association. Circulation 139:840–878

    Article  Google Scholar 

  8. Telmisartan Randomised AssessmeNt Study in ACE iNtolerant subjects with cardiovascular Disease (TRANSCEND) Investigators, Yusuf S, Teo K, Anderson C, Pogue J, Dyal L, Copland I, Schumacher H, Dagenais G, Sleight P (2008) Effects of the angiotensin-receptor blocker telmisartan on cardiovascular events in high-risk patients intolerant to angiotensin-converting enzyme inhibitors: a randomised controlled trial. Lancet 372(9644):1174–1183

  9. Sent U, Gössl R, Elliott J, Syme HM, Zimmering T (2015) Comparison of efficacy of long-term oral treatment with Telmisartan and Benazepril in cats with Chronic Kidney Disease. J Vet Intern Med 29(6):1479–1487

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Hou Y, Zhang F, Liu Z, Su S, Wu X, Wang Z (2017) Effect of Telmisartan and Enalapril on ventricular remodeling and kidney prognosis of patients with coronary artery disease complicated with diabetic nephropathy. Exp Ther Med 13(1):131–134. https://doi.org/10.3892/etm.2016.3933

    Article  CAS  PubMed  Google Scholar 

  11. Bozkurt B, Coats AJS, Tsutsui H, Abdelhamid CM, Adamopoulos S, Albert N, Anker SD, Atherton J, Böhm M, Butler J, Drazner MH, Michael Felker G, Filippatos G, Fiuzat M, Fonarow GC, Gomez-Mesa JE, Heidenreich P, Imamura T, Jankowska EA, Januzzi J, Khazanie P, Kinugawa K, Lam CSP, Matsue Y, Metra M, Ohtani T, Francesco Piepoli M, Ponikowski P, Rosano GMC, Sakata Y, Seferović P, Starling RC, Teerlink JR, Vardeny O, Yamamoto K, Yancy C, Zhang J, Zieroth S (2021) Universal definition and classification of heart failure: a report of the Heart Failure Society of America, Heart Failure Association of the European Society of Cardiology, Japanese Heart Failure Society and Writing Committee of the Universal Definition of heart failure. Eur J Heart Fail 23(3):352–380. https://doi.org/10.1002/ejhf.2115

    Article  PubMed  Google Scholar 

  12. Levey AS, Coresh J, Greene T, Stevens LA, Zhang Y, Hendriksen S, Kusek JW, van Lente F (2006) Using standardized serum creatinine values in the modification of diet in renal disease study equation for estimating glomerular filtration rate. Ann Intern Med 145(4):247–254. https://doi.org/10.7326/0003-4819-145-4-200608150-00004

    Article  CAS  PubMed  Google Scholar 

  13. Issues S, Test MW, Equipment R, Preparation P (2002) American Thoracic Society ATS statement: guidelines for the six-minute walk test. 166:111–117

  14. Biaggioni I, Kaufmann H (2014) Orthostatic hypotension. Encycl Neurolog Sci 698–700

  15. Lang RM, Badano LP, Mor-Avi V, Afilalo J, Armstrong A, Ernande L, Flachskampf FA, Foster E, Goldstein SA, Kuznetsova T, Lancellotti P, Muraru D, Picard MH, Rietzschel ER, Rudski L, Spencer KT, Tsang W, Voigt JU (2015) Recommendations for cardiac chamber quantification by echocardiography in adults: an update from the American society of echocardiography and the European association of cardiovascular imaging. Eur Heart J Cardiovasc Imaging 16(3):233–271

    Article  PubMed  Google Scholar 

  16. Mewton N, Girerd N, Boffa JJ, Courivaud C, Isnard R, Juillard L, Lamblin N, Legrand M, Logeart D, Mariat C, Meune E, Sabouret P, Sebbag L, Rossignol P (2020) Practical management of worsening renal function in outpatients with heart failure and reduced ejection fraction: statement from a panel of multidisciplinary experts and the Heart Failure Working Group of the French Society of Cardiology. Arch Cardiovasc Dis 113(10):660–670

    Article  PubMed  Google Scholar 

  17. Metra M, Nodari S, Parrinello G, Bordonali T, Bugatti S, Danesi R, Fontanella B, Lombardi C, Milani P, Verzura G, Cotter G, Dittrich H, Massie BM, Dei CL (2008) Worsening renal function in patients hospitalised for acute heart failure: clinical implications and prognostic significance. Eur J Heart Fail 10(2):188–195

    Article  PubMed  Google Scholar 

  18. Breidthardt T, Socrates T, Noveanu M, Klima T, Heinisch C, Reichlin T, Potocki M, Nowak A, Tschung C, Arenja N, Bingisser R, Mueller C (2011) Effect and clinical prediction of worsening renal function in acute decompensated heart failure. Am J Cardiol 107(5):730–735

    Article  PubMed  Google Scholar 

  19. Chittineni H, Miyawaki N, Gulipelli S, Fishbane S (2007) Risk for acute renal failure in patients hospitalized for decompensated congestive heart failure. Am J Nephrol 27(1):55–62

    Article  PubMed  Google Scholar 

  20. Iglesias JI, DePalma L, Hom D, Antoniotti M, Ayoub S, Levine JS (2008) Predictors of mortality in adult patients with congestive heart failure receiving nesiritide–retrospective analysis showing a potential adverse interaction between nesiritide and acute renal dysfunction. Nephrol Dial Transplant 23(1):144–153

    Article  CAS  PubMed  Google Scholar 

  21. Testani JM, McCauley BD, Chen J, Shumski M, Shannon RP (2010) Worsening renal function defined as an absolute increase in serum creatinine is a biased metric for the study of cardio-renal interactions. Cardiology 116(3):206–212

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Ather S, Bavishi C, McCauley MD, Dhaliwal A, Deswal A, Johnson S, Chan W, Aguilar D, Pritchett AM, Ramasubbu K, Wehrens XHT, Bozkurt B (2013) Worsening renal function is not associated with response to treatment in acute heart failure. Int J Cardiol 167(5):1912–1917

    Article  PubMed  Google Scholar 

  23. Shirakabe A, Hata N, Kobayashi N, Okazaki H, Matsushita M, Shibata Y, Nishigoori S, Uchiyama S, Asai K, Shimizu W (2018) Worsening renal function definition is insufficient for evaluating acute renal failure in acute heart failure. ESC Heart Fail 5(3):322–331

    Article  PubMed  PubMed Central  Google Scholar 

  24. Damman K, Valente MAE, Voors AA, O’Connor CM, Van Veldhuisen DJ, Hillege HL (2014) Renal impairment, worsening renal function, and outcome in patients with heart failure: an updated meta-analysis. Eur Heart J 35(7):455–469

    Article  PubMed  Google Scholar 

  25. McMurray JJV, Packer M, Desai AS, Gong J, Lefkowitz MP, Rizkala AR, Rouleau JL, Shi VC, Solomon SD, Swedberg K, Zile MR (2014) Angiotensin–Neprilysin inhibition versus Enalapril in heart failure. N Engl J Med 371(11):993–1004. https://doi.org/10.1056/NEJMoa1409077

    Article  CAS  PubMed  Google Scholar 

  26. Abdelhamid M, Kandil H, Hassanin M, Shaheen S, Sobhy M, ElEtreby A, Hasan-Ali H, Mahfouz H, Nasr G, Shawky I, Emil S, ElSetiha M, Hasssan M, Sadek Y, Karim MA, Asham A, Ghaleb M, Samir A, Shokry K (2022) Egyptian expert opinion for the use of sodium-glucose cotransporter-2 inhibitors in patients with heart failure with reduced ejection fraction. ESC Heart Fail 9(2):800–811. https://doi.org/10.1002/ehf2.13811

    Article  PubMed  PubMed Central  Google Scholar 

  27. Yogasundaram H, Chappell MC, Braam B, Oudit GY (2019) Cardiorenal syndrome and heart failure—challenges and opportunities. Can J Cardiol 35(9):1208–1219. https://doi.org/10.1016/j.cjca.2019.04.002

    Article  PubMed  Google Scholar 

  28. Packer M, Poole-Wilson PA, Armstrong PW, Cleland JGF, Horowitz JD, Massie BM, Rydén L, Thygesen K, Uretsky BF (1999) Comparative effects of low and high doses of the angiotensin-converting enzyme inhibitor, lisinopril, on morbidity and mortality in chronic heart failure. Circulation 100(23):2312–2318. https://doi.org/10.1161/01.CIR.100.23.2312

    Article  CAS  PubMed  Google Scholar 

  29. Konstam MA, Neaton JD, Dickstein K, Drexler H, Komajda M, Martinez FA, Riegger GAJ, Malbecq W, Smith RD, Guptha S, Poole-Wilson PA, HEAAL Investigators (2009) Effects of high-dose versus low-dose losartan on clinical outcomes in patients with heart failure (HEAAL study): a randomised, double-blind trial. Lancet 374(9704):1840–1848

  30. Basi S, Lewis J (2006) Microalbuminuria as a target to improve cardiovascular and renal outcomes. Am J Kidney Dis 47(6):927–946

    Article  CAS  PubMed  Google Scholar 

  31. Mentz RJ, Bakris GL, Waeber B, McMurray JJV, Gheorghiade M, Ruilope LM, Maggioni AP, Swedberg K, Piña IL, Fiuzat M, O’Connor CM, Zannad F, Pitt B (2013) The past, present and future of renin–angiotensin aldosterone system inhibition. Int J Cardiol 167(5):1677–1687

    Article  PubMed  Google Scholar 

  32. Bakheit AHH, Abd-Elgalil AA, Mustafa B, Haque A, Wani TA (2015) Telmisartan. Profiles Drug Subst Excip Relat Methodol 40:371–429

    Article  CAS  PubMed  Google Scholar 

  33. Amarenco P, Bogousslavsky J, Callahan A III Goldstein LB, Hennerici M, Rudolph AE, Sillesen H, Simunovic L, Szarek M, Welch KMA, Zivin JA, SP by AR in CL (SPARCL) I, Raymond E, Pisano E, Gatsonis C, Boineau R, Domanski M, Troutman C, Anderson J, Johnson G, Mcnulty SE, Clapp-channing N, Davidson-ray LD, Fraulo ES, Fishbein DP, Luceri RM, Teo KK, Pogue J, Dyal L, Copland I, Health P, Sciences HHHH, Schumacher H, Ingelheim B, Dagenais G, Kingdom U, Anderson C, Sciences HHHH (2008) Telmisartan, ramipril, or both in patients at high risk for vascular events. N Engl J Med 358(15):1547–1559. https://doi.org/10.1056/NEJMoa0801317

  34. Mann JF, Schmieder RE, McQueen M, Dyal L, Schumacher H, Pogue J, Wang X, Maggioni A, Budaj A, Chaithiraphan S, Dickstein K, Keltai M, Metsärinne K, Oto A, Parkhomenko A, Piegas LS, Svendsen TL, Teo KK, Yusuf S (2008) Renal outcomes with telmisartan, ramipril, or both, in people at high vascular risk (the ONTARGET study): a multicentre, randomised, double-blind, controlled trial. Lancet 372(9638):547–553

    Article  CAS  PubMed  Google Scholar 

  35. Kusunoki H, Taniyama Y, Azuma J, Iekushi K, Sanada F, Otsu R, Iwabayashi M, Okayama K, Rakugi H, Morishita R (2012) Telmisartan exerts renoprotective actions via peroxisome proliferator-activated receptor-γ/hepatocyte growth factor pathway independent of angiotensin II type 1 receptor blockade. Hypertension 59(2):308–316

    Article  CAS  PubMed  Google Scholar 

  36. Hu B, Zhou Y, Shi D, Zhao Y (2010) e0405 Effect of telmisartan on cardiac function and brain natriuretic peptide in patients with chronic heart failure. Heart 96(Suppl 3):A125–A125. https://doi.org/10.1136/hrt.2010.208967.405

    Article  Google Scholar 

  37. Dunselman PH (2001) Replacement of Angiotensin Converting Enzyme Inhibition (REPLACE) Investigators. Effects of the replacement of the angiotensin converting enzyme inhibitor enalapril by the angiotensin II receptor blocker telmisartan in patients with congestive heart failure. The replacement of angiotensin converting enzyme inhibition (REPLACE) investigators. Int J Cardiol 77(2–3):131–8; discussion 139–40

  38. Metra M, Davison B, Bettari L, Sun H, Edwards C, Lazzarini V, Piovanelli B, Carubelli V, Bugatti S, Lombardi C, Cotter G, Dei CL (2012) Is worsening renal function an ominous prognostic sign in patients with acute heart failure? The role of congestion and its interaction with renal function. Circ Heart Fail 5(1):54–62. https://doi.org/10.1161/CIRCHEARTFAILURE.111.963413

    Article  PubMed  Google Scholar 

  39. Eshaghian S, Horwich TB, Fonarow GC (2006) Relation of loop diuretic dose to mortality in advanced heart failure. Am J Cardiol 97(12):1759–1764

    Article  CAS  PubMed  Google Scholar 

  40. Khan MS, Ahmed A, Greene SJ, Fiuzat M, Kittleson MM, Butler J, Bakris GL, Fonarow GC (2022) Managing heart failure in patients on dialysis: state-of-the-art review. J Card Fail

  41. Cice G, Di Benedetto A, D’Isa S, D’Andrea A, Marcelli D, Gatti E, Calabr R (2010) Effects of telmisartan added to angiotensin-converting enzyme inhibitors on mortality and morbidity in hemodialysis patients with chronic heart failure: A double-blind, placebo-controlled trial. J Am Coll Cardiol 56(21):1701–1708. https://doi.org/10.1016/j.jacc.2010.03.105

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not available.

Funding

Authors received no funds. This study was conducted in- and supported by Cairo University Hospitals.

Author information

Authors and Affiliations

Authors

Contributions

All authors well contributed in the conceptual design, writing and revision of the manuscript and approved the final version. Order is according to authors contribution.

Corresponding author

Correspondence to Ahmad Samir.

Ethics declarations

Ethics approval and consent to participate

The study protocol and the informed consent form were revised and approved by the research ethics committee, Faculty of Medicine, Cairo University under registration number (MS-352-2020). [Research ethics committee reference: + 0201201730249; kasralainyrec@gmail.com] Written informed consents were acquired from all study participants.

Consent for publication

Patient informed consent form included approval to publish anonymized clinical data.

Competing interests

None of the authors have competing or conflicting interests that are directly or indirectly related to the work submitted for this publication.

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 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/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Samir, A., Aboel-Naga, S., Shehata, A. et al. Telmisartan versus EnalapRil In heart failure with redUced ejection fraction patients with Moderately impaired kidney Functions; randomized controlled trial: “TRIUMF trial”. Egypt Heart J 75, 68 (2023). https://doi.org/10.1186/s43044-023-00398-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43044-023-00398-7

Keywords