Abstract
Semaglutide, a glucagon-like peptide-1 receptor agonist (GLP-1RA), has demonstrated substantial efficacy in managing type 2 diabetes mellitus (T2DM). It provides glycemic control, promotes weight loss, and offers cardiovascular protection. Evidence also supports its role in diabetic kidney disease (DKD), a leading global cause of end-stage renal disease. DKD arises from a multifactorial interaction involving hyperglycemia, hypertension, and inflammation, which leads to cumulative nephron loss. Beyond glycemic control, semaglutide’s mechanisms of action target metabolic and hemodynamic pathways that contribute to renal damage. This review evaluates the preclinical and clinical evidence of semaglutide’s role in preventing DKD, focusing on its renal effects and the mechanistic basis for renoprotection. We also position semaglutide within the broader DKD therapeutic landscape by reviewing clinical trial findings, translational studies, real-world evidence, and its effectiveness compared to other drug classes. The expanded actions of semaglutide make it a promising agent in patients with T2DM and DKD and encourage further mechanistic research and long-term evaluation.
1. Introduction
Diabetic kidney disease (DKD) is a microvascular complication of diabetes mellitus. It is currently the leading cause of chronic kidney disease (CKD) and end-stage renal disease (ESRD) globally. As the number of type 2 diabetes mellitus (T2DM) cases is rising, the prevalence of DKD is also increasing. DKD is primarily linked to morbidity, mortality, and rising healthcare costs [1,2], and has been observed to develop in about 20–50% of individuals with T2DM. The features of DKD are persistent albuminuria, a reduced glomerular filtration rate (GFR), and an increased cardiovascular risk [3,4]. Despite advances in glycemic control, renin–angiotensin system blockade, and blood pressure and lipid management, the residual risks of renal and cardiovascular issues remain high [5,6].
Pharmacological treatment for T2DM has changed significantly in the past decade. New drugs now provide additional glucose-lowering benefits. Among these, glucagon-like peptide-1 receptor agonists (GLP-1RAs) are highly prioritized as they provide cardiovascular protection, weight reduction, and anti-inflammatory effects [7,8,9]. Once-weekly administration of GLP-1RA semaglutide more effectively reduces glycated hemoglobin (HbA1c) levels, body weight, and cardiovascular events than traditional agents [9,10]. Notably, new evidence suggests that it may provide renoprotective effects [11]. Semaglutide stands out among GLP-1 receptor agonists for its advanced pharmacologic profile and consistent renal benefits. Its molecular design exhibits enhanced albumin binding and structural modifications that prolong its plasma half-life, enabling once-weekly subcutaneous dosing, while the oral formulation is taken once daily [12,13]. This extended exposure sustains receptor activation and improves glycemic and extra-glycemic efficacy compared with other agents. For example, liraglutide requires daily injection, while dulaglutide, though administered weekly, leads to smaller reductions in HbA1c and body weight [13,14]. Data from the SUSTAIN-6 trial demonstrate that semaglutide exerts renoprotective effects, predominantly through the attenuation of new-onset macroalbuminuria [15]. By contrast, the renal outcomes observed after liraglutide (LEADER trial) and dulaglutide (REWIND trial) administration were favorable but of comparatively lower magnitude [16,17]. Collectively, these findings suggest that semaglutide’s molecular modifications translate into clinically meaningful renal protection beyond glycemic control, distinguishing it within the GLP-1RA class. This review aimed to assess the literature on semaglutide in DKD. We summarize its pharmacological properties, actions, and recent clinical and mechanistic evidence, evaluating its promise as a disease-modifying therapy for diabetic nephropathy.
2. Semaglutide
Semaglutide is a weekly administered glucagon-like peptide-1 receptor agonist (GLP-1RA) used in the treatment of type 2 diabetes mellitus (T2DM) as well as obesity. Semaglutide is 94% homologous with endogenous GLP-1 but contains the necessary modifications to offer an enhanced stability and half-life [18]. Particularly, substitution at position 8 in alanine with α-aminoisobutyric acid confers resistance to degradation by dipeptidyl peptidase-4 (DPP-4), and acylation with C18 fatty acid enables albumin binding and systemic exposure over prolonged periods [13,19]. These structural changes allow for once-weekly dosing, a regimen that has been demonstrated to greatly increase drug persistence and adherence, thereby reducing patient treatment burden and potentially maximizing the therapeutic outcomes in real-world clinical practice [20]. The pharmacological effects of semaglutide are mediated by GLP-1 receptor activation on pancreatic β-cells, resulting in increased glucose-dependent insulin secretion and suppression of glucagon release. Semaglutide decreases gastric emptying and enhances satiety via central hypothalamic mechanisms, leading to significant weight loss [21,22]. Importantly, these effects are not accompanied by a high risk of hypoglycemia [23]. In SUSTAIN 5 and SUSTAIN 7 clinical trials, semaglutide consistently decreased HbA1c by 1.1% to 1.8% and body weight by 4–6 kg in diverse populations [14,24]. In SUSTAIN-6, a historic cardiovascular outcomes trial, semaglutide reduced major adverse cardiovascular events (MACEs) by 26% relative to a placebo [9]. Semaglutide had kidney-protective effects in patients with T2DM, which were more pronounced in those with baseline chronic kidney disease [11]. Semaglutide has a similar safety profile to other members of the GLP-1RA class, the most common adverse events being gastrointestinal effects such as nausea, vomiting, and diarrhea, particularly on dose escalation. These findings are mirrored in randomized controlled trials (RCTs) and further corroborated by post-marketing pharmacovigilance experience [25,26]. Available data from clinical trials and post-marketing experience have not demonstrated a consistent, causal association between exposure to semaglutide and the development of acute pancreatitis or medullary thyroid carcinoma (MTC) in humans. However, in rodents, it causes thyroid C-cell tumors; therefore, this contraindicates its use in patients with a personal or family history of MTC or Multiple Endocrine Neoplasia syndrome type 2 (MEN2) [27,28]. When used alone or in combination with metformin, semaglutide has an inherently low hypoglycemia risk, but this increases when used with insulin or with insulin secretagogues such as sulfonylureas [23]. Diabetic retinopathy complications are a rare but serious problem, as observed in SUSTAIN-6, though subsequent analysis suggests that this is more a consequence of too-rapid glycemic correction than a drug effect per se [29,30].
3. Diabetic Kidney Disease
Diabetic kidney disease (DKD), or diabetic nephropathy, is a chronic disease of both type 1 and type 2 diabetes mellitus, where the latter is the most significant contributor due to its global prevalence. DKD is responsible for about 30–50% of all cases of end-stage renal disease (ESRD) in developed nations. It is also associated with very high cardiovascular morbidity and mortality [2,31]. Despite advancements in managing diabetes, DKD is continuing to increase in prevalence, particularly among older adults and obese individuals [32]. Pathologically, DKD is characterized by initial glomerular hyperfiltration, followed by microalbuminuria, macroalbuminuria, and finally a reduced estimated glomerular filtration rate (eGFR). Histologically, it is characterized by mesangial matrix expansion, glomerular basement membrane thickening, loss of podocytes, arteriolar hyalinosis, and interstitial fibrosis [33,34]. These alterations are induced by chronic hyperglycemia, oxidative stress, activation of the renin–angiotensin–aldosterone system (RAAS), and pro-inflammatory and profibrotic pathway upregulation, especially through transforming growth factor-β (TGF-β) and nuclear factor-kappa B (NF-κB). Hyperinsulinemia, insulin resistance, dyslipidemia, and sodium retention also contribute [35,36]. Clinically, a diagnosis of DKD is most commonly established based on persistent albuminuria (albumin-to-creatinine ratio > 30 mg/g), a declining eGFR (<60 mL/min/1.73 m2), or both, excluding other renal disease etiologies [37]. Nevertheless, 56.6% of T2DM patients with DKD may have a decreased eGFR without albuminuria, an indication of the heterogeneity of disease phenotypes [38].
4. Semaglutide’s Effect on Diabetic Kidney Disease
4.1. Mechanistic Basis for Renoprotection
Experimental and translational data suggest that GLP-1 receptor agonists modulate multiple biological processes implicated in the pathogenesis of DKD. These include oxidative stress, inflammation, fibrosis, and endothelial dysfunction. Chronic hyperglycemia is the main determinant of the metabolic and molecular disturbances that form the basis of diabetic nephropathy. It leads to overgeneration of reactive oxygen species (ROS), and consequently oxidative stress, producing an environment that damages glomerular and tubular cell function [39]. This oxidative environment also interacts with inflammatory pathways, creating a feed-forward loop that exacerbates renal injury. In combination with this redox stress, inflammatory signaling is triggered. Clinical and experimental evidence has particularly underlined the role of pro-inflammatory cytokines, including tumor necrosis factor-α (TNF-α), interleukin-1 (IL-1), interleukin-6 (IL-6), and interleukin-18 (IL-18), which exacerbate structural and functional renal injury [40]. Therefore, oxidative stress and inflammation appear to be concomitant mechanisms, where metabolic stress preconditions renal tissue for damage and cytokine-mediated cascades facilitate disease progression. In a preclinical model of renal damage related to obesity, administration of semaglutide significantly reduced TNF-α, IL-6, and IL-1β levels, with increased activity of antioxidant enzymes such as superoxide dismutase (SOD) and reduced renal malondialdehyde (MDA) [41]. This suggests that semaglutide could have a protective effect against oxidative stress and inflammation in DKD. Building on these oxidative and inflammatory mechanisms, recent research has also explored the cellular and molecular pathways through which semaglutide may exert renoprotective effects. Recent research has highlighted mechanisms in which semaglutide can exhibit its renoprotective effects in DKD. One line of evidence implicates β-Klotho (KLB) as a key mediator. This involves the cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA)/phosphorylation and cAMP-response element-binding protein (CREB) signaling pathways. Semaglutide increases KLB expression and activates AMPK and thus reprograms ferroptosis-related metabolic pathways such as iron regulation, fatty acid synthesis, and antioxidant response to lipid peroxidation. The result is suppression of ferroptosis with secondary reductions in renal inflammation and fibrosis [42]. Another study demonstrated that in a model of hypertensive-accelerated DKD in uninephrectomized db/db mice with renin overexpression, treatment with semaglutide significantly improved kidney injury, including glomerulosclerosis, with a simultaneous significant improvement in hyperglycemia, hypertension, and albuminuria. It also improved the podocyte filtration slit density and reduced the levels of urine and renal kidney injury molecule-1 (KIM-1), which is a marker for kidney injury, as well as suppressing inflammatory and fibrogenic gene expression [43]. Findings support that semaglutide could have the potential to manage DKD with its actions of glycemic control and direct cellular protection, modulation of ferroptosis, and improvement of hemodynamic and inflammatory stressors.
Extending from these intracellular and metabolic effects, semaglutide may also influence intercellular communication pathways that contribute to DKD progression. Moreover, recent evidence indicates that endothelial-derived extracellular vesicles (EVs) contribute to macrophage activation and upregulation of pro-inflammatory cytokines in diabetic tissues, implicating EV-mediated inflammatory signaling in the propagation of chronic inflammation. This identifies EVs as critical intermediaries linking endothelial dysfunction with immune activation, representing an emerging mechanistic insight in diabetes and its complications [44].
The results of McLean et al. show that semaglutide decreases atherosclerosis in mice, regardless of GLP-1 receptor expression on Tie2+ endothelial cells and hematopoietic cells, suggesting that the vascular protective effects of semaglutide are to mediated a great extent by positive systemic metabolic effects, e.g., reductions in glucose, lipid toxicity, and inflammation, but not via endothelial GLP-1R signaling. However, the same study identified Tie2+ GLP-1R signaling as important for anti-inflammatory and antifibrotic action in the liver, which correlates with tissue-specific variances [45]. This is consistent with the hypothesis that semaglutide could indirectly improve endothelial dysfunction and albuminuria in DKD through systemic metabolic and anti-inflammatory actions, but not through direct endothelial receptor interactions. Finally, linking these systemic effects to renal hemodynamics further illustrates semaglutide’s multifaceted renoprotective profile. A study conducted by Farah et al. showed that GLP-1 receptor signaling plays a crucial role in the regulation of renal sodium handling, primarily by phosphorylating and inhibiting the proximal tubule Na+/H+ exchanger NHE3 via cAMP/PKA-dependent pathways, thereby increasing natriuresis and diuresis [46]. While Farah et al. studied endogenous GLP-1 rather than semaglutide specifically, these mechanisms provide a probable explanation for how GLP-1RA-like semaglutide may influence renal hemodynamics, reduce intraglomerular pressure, and provide an additive renoprotective effect, particularly in the hyperfiltration setting, a characteristic of DKD. These mechanisms are summarized schematically in Figure 1, which illustrates the major cellular and molecular pathways implicated in DKD that are targeted by semaglutide. A concise mapping of these pathways, linking preclinical and clinical evidence, is provided in Table 1.
Figure 1.
Proposed mechanisms of semaglutide-mediated renoprotection. Abbreviation: EV, endothelial-derived extracellular vesicle.
Table 1.
Mechanistic pathways of semaglutide-mediated renoprotection and supporting preclinical and clinical evidence.
4.2. Clinical Trial Evidence
Data for the renoprotective actions of semaglutide in humans have been extracted from several randomized clinical trials and post hoc analyses (Table 2). Although not originally designed to assess the renal effects of semaglutide as part of the primary endpoints, except for FLOW trial, the prespecified analyses have yielded important information. In SUSTAIN-6, a 3297-patient randomized double-blind trial of type 2 diabetics at high cardiovascular risk, semaglutide reduced the risk of new or worsening nephropathy by 36% (HR 0.64, 95% CI 0.46–0.88). This effect was explained in large part by a 46% reduction in new-onset persistent macroalbuminuria (HR 0.54, 95% CI 0.37–0.77), but there were also nonsignificant trends towards a lower decline in eGFR and fewer cases of serum creatinine doublings [9]. A pooled analysis of the SUSTAIN 1–7 programs confirmed these findings, showing attenuated eGFR declines across diverse patient populations without new safety concerns [47].
Oral semaglutide has also demonstrated renal safety and potential renoprotection. In PIONEER-5, involving patients with T2DM and moderate CKD, oral semaglutide was effective, well tolerated, and associated with stabilization of kidney function, including mild reductions in albuminuria [48]. In PIONEER-6, designed primarily as a cardiovascular outcomes trial, renal endpoints were exploratory. Nonetheless, a post hoc analysis revealed 36% fewer nephropathy events (HR 0.64, 95% CI 0.46–0.88) and a 46% lower risk of new-onset macroalbuminuria (HR 0.54, 95% CI 0.37–0.77) compared with a placebo, paralleling the pattern seen with injectable semaglutide [15]. Collectively, these findings support a class-wide renoprotective effect of GLP-1 receptor agonists rather than formulation-specific effects.
A systematic review and meta-analysis of seven GLP-1RA cardiovascular outcome trials by Sattar et al., 2021 [49] showed a consistent 21% reduction in composite kidney outcomes with GLP-1RA use, largely driven by decreases in macroalbuminuria. Semaglutide, however, appeared to show a quantitatively greater effect than other agents, including liraglutide and dulaglutide, although the mechanism for this difference has not been firmly established. Overall, while evidence from clinical trials regarding the effects of semaglutide in the kidneys is compelling, it is not yet exhaustive. The FLOW trial should provide definitive clarity on semaglutide’s role as a disease-modifying therapy in DKD.
Larger, long-term renal outcome studies were previously required to confirm these preliminary observations. To surmount these limitations, the FLOW trial (NCT03819153), the first renal-dedicated outcomes trial for a GLP-1 receptor agonist, has now reported its primary results [50,51]. This global, randomized, double-blind, placebo-controlled trial evaluated once-weekly subcutaneous semaglutide (1.0 mg) versus placebo in 3533 patients with T2DM and CKD (eGFR 25–75 mL/min/1.73 m2 and UACR 100–5000 mg/g). A 24% reduction in the primary composite outcome of sustained ≥ 50% reduction in eGFR, ESKD, or death from renal or cardiovascular causes (HR 0.76; 95% CI 0.66–0.88) was observed during a median follow-up of 3.4 years [51]. The risk of kidney failure alone was reduced by 24%, and the risk of major adverse cardiovascular events by 18%, with consistent safety findings compared with the placebo. As the first stand-alone renal outcomes trial for a GLP-1 receptor agonist with hard renal endpoints, FLOW establishes robust evidence that semaglutide slows kidney disease progression in patients with T2DM and CKD, beyond its glucose-lowering effect. These findings align with previous meta-analyses and pooled analyses supporting the renoprotective actions of GLP-1 receptor agonists.
Table 2.
Summary of clinical trials and post hoc analyses evaluating renal outcomes with semaglutide in patients with T2DM and DKA.
Table 2.
Summary of clinical trials and post hoc analyses evaluating renal outcomes with semaglutide in patients with T2DM and DKA.
| Trial | Study Design | Population | Intervention | Duration | Renal Baseline Characteristics | Primary Renal Outcome | Key Findings | Reference |
|---|---|---|---|---|---|---|---|---|
| SUSTAIN-6 | Randomized, double-blind, placebo-controlled | T2DM with high CV risk | Semaglutide 0.5–1.0 mg weekly vs. placebo | Median 2.1 years | Mean eGFR: 75.6 mL/min/1.73 m2; UACR 34 mg/g | Combination of new/worsening nephropathy (≥30% eGFR decline, ESKD, or death from renal causes) | 36% reduction in nephropathy events (HR 0.64, 95% CI 0.46–0.88); 46% lower risk of macroalbuminuria (HR 0.54, 95% CI 0.37–0.77); slower eGFR decline vs. placebo | [9] |
| SUSTAIN 1–7 pooled analysis | A pooled post hoc analysis of the SUSTAIN 1–7 randomized controlled trials | T2DM patients | Semaglutide vs. placebo or active comparators, depending on the specific SUSTAIN trial | Varies by trial | Varies by trial | Changes in eGFR, reductions in urinary albumin-to-creatinine ratio (UACR), and the occurrence of kidney-related adverse events | Initial eGFR dip followed by stabilization, resulting in a net annual slope improvement of ≈0.59 mL/min/1.73 m2 versus comparators; significant reductions in UACR; no new renal safety signals observed | [47] |
| PIONEER-5 | Randomized, double-blind, placebo-controlled phase 3a | T2DM with moderate renal impairment (eGFR 30–59 mL/min/1.73 m2) | Oral semaglutide (14 mg daily) vs. placebo | A 26-week randomized treatment period and a follow-up period of 5 weeks | eGFR 30–59 mL/min/1.73 m2 (moderate CKD) | Exploratory renal safety and albuminuria change (not dedicated renal composite) | eGFR remained generally stable in both groups (no slope provided); UACR decreased numerically in the semaglutide group (magnitude not reported, no formal statistical test); no new renal safety issues were identified | [48] |
| PIONEER-6 | Randomized, double-blind, placebo-controlled CVOT | T2DM with high CV risk (eGFR ≥ 30 mL/min/1.73 m2) | Oral semaglutide (14 mg daily) vs. placebo | Median follow-up = 1.3 years | Mean baseline eGFR = 74 mL/min/1.73 m2 (estimated from pooled data) | Exploratory renal safety outcomes were assessed, but there was no prespecified primary renal endpoint | Exploratory renal outcomes: HRs for persistent eGFR decline <1.0 overall (NS); in patients with baseline eGFR 30–<60, semaglutide significantly reduced the risk of 30% eGFR decline (p = 0.03); no major renal safety concerns; no statistically different interactions between treatment and CKD subgroup were observed | [15] |
| Post hoc analysis of SUSTAIN-6 and PIONEER-6 | Pooled post hoc analysis of two cardiovascular outcome trials (SUSTAIN-6 and PIONEER-6) | Patients with T2DM at high cardiovascular risk (n = 6480; semaglutide = 3239; placebo = 3241) | Semaglutide (s.c. and oral) vs. placebo | Median follow-up = 2.1 years (SUSTAIN-6) + 1.3 years (PIONEER-6) | eGFR ≥ 30 mL/min/1.73 m2; subgroup with eGFR 30–<60 mL/min/1.73 m2 analyzed separately; baseline UACR available for SUSTAIN-6 only | Annual eGFR slope; time to persistent eGFR decline (≥30%, ≥40%, ≥50%, ≥57%) | Annual eGFR decline was slower by 0.59 mL/min/1.73 m2/year (95% CI 0.29–0.89) with semaglutide vs. placebo. In patients with baseline eGFR 30–<60, the difference was 1.06 mL/min/1.73 m2/year (95% CI 0.45–1.67). HRs for time to persistent eGFR decline were consistently <1.0, favoring semaglutide, though not all reached statistical significance | [52] |
| Meta-analysis of GLP-1 receptor agonists | Systematic review and meta-analysis of 8 major cardiovascular outcome trials (n = 60,080) | Patients with T2DM; broad cardiovascular risk spectrum | GLP-1 RAs (including liraglutide, semaglutide, dulaglutide, albiglutide, exenatide, lixisenatide) vs. placebo | Median trial duration 1.3–5.4 years | Baseline mean eGFR ~78 mL/min/1.73 m2; ~23% with eGFR < 60 mL/min/1.73 m2 | Combined kidney outcomes (new-onset macroalbuminuria, sustained ≥30–40% decline in eGFR, kidney replacement therapy, or renal death) | GLP-1 receptor agonists reduced the combined kidney outcome by 21% (HR 0.79; 95% CI 0.73–0.87; p < 0.0001), driven largely by a 26% lower risk of new-onset macroalbuminuria (HR 0.74; 95% CI 0.68–0.81). Risk reduction for hard renal endpoints (≥40% eGFR decline, doubling of serum creatinine, end-stage kidney disease, or renal death) was nominal (HR 0.86; 95% CI 0.72–1.02). Effects were consistent across baseline eGFR subgroups | [49] |
| FLOW | Randomized, double-blind, placebo-controlled, event-driven kidney-outcomes trial | 3533 adults with T2DM and CKD (eGFR ~47 mL/min/1.73 m2; median UACR ~568 mg/g) | Semaglutide 1.0 mg weekly vs. placebo (plus standard of care) | Median ~3.4 years | Mean baseline eGFR = 47 mL/min/1.73 m2; median UACR = 568 mg/g | Composite of kidney failure, sustained ≥50% eGFR decline, kidney/cardiovascular death | 24% reduction in primary composite kidney outcome (HR 0.76; 95% CI 0.66–0.88). Annual eGFR decline slowed by ~1.16 mL/min/1.73 m2/yr in the semaglutide group | [50,51] |
4.3. Translational Studies and Real-World Evidence
Although randomized controlled trials (RCTs) are still the gold standard to assess drug efficacy, they are usually limited by narrow inclusion criteria, limited follow-up times, and a lack of external validity. Translational studies and real-world evidence (RWE) can potentially close these gaps through an assessment of therapy performance in more general populations as well as through the creation of mechanisms in human tissue. For semaglutide, aggregate real-world and translational evidence has started to substantiate its renal protective role.
In actual clinical practice, semaglutide treatment of patients with T2DM and CKD has been linked to reduced progression of kidney deterioration and statistically significant decreases in albuminuria. In a Spanish multicenter cohort of n = 156 patients, a substantial amelioration in the annual slope of eGFR from −3.29 to −0.79 mL/min/1.73 m2/year was documented, despite a follow-up duration of at least two years and the sample size of the study [53]. Another observational trial in 122 CKD patients demonstrated a mean 51% decrease in the UACR in macroalbuminuria subjects with a sustained eGFR over a period of 12 months.
Similarly, a multicenter real-life observational study of T2DM 2 and CKD (mostly stage 2 and 3) patients treated with subcutaneous semaglutide revealed that albuminuria was significantly reduced, especially among those with macroalbuminuria at baseline, and the eGFR was unchanged over 12 months of follow-up [54]. However, the majority of mechanistic and clinical world evidence comes from small, unblinded, or observational trials that did not have solid endpoints like ESKD or doubling of serum creatinine. However, they do offer considerable biologic plausibility and demonstrate that semaglutide’s kidney-protective effects are not a consequence of enhanced glycemia or weight loss. The combination of RCT data, observational data, and mechanistic research at the tissue level supports the hypothesis that semaglutide has multifactorial renal protective action on DKD. Table 3 summarizes the key distinctions between randomized controlled trials and real-world observational studies in semaglutide research.
Table 3.
Comparison of randomized controlled trials (RCTs) and real-world observational studies (RWE) evaluating semaglutide in T2DM and DKD.
4.4. Comparative Effectiveness with Other Drug Classes
The conventional therapy for DKD has been centered on intense glycemic control; reducing blood pressure, particularly with RAAS inhibitors; reducing lipids; and lifestyle modifications. All these treatments, while useful in the sense of retarding progression, are not yet able to eliminate the risk of ESRD or cardiovascular events. Large trials have substantiated this: tight glucose control has been observed to lead to a small reduction in nephropathy in the ADVANCE trial, but has a low impact on macrovascular events [55], and there was no clear renal benefit of strict glucose reduction in the ACCORD trial and it was indeed related to increased mortality in some groups [56].
Recent therapeutic advances have significantly changed the landscape of treatment. Sodium-glucose cotransporter-2 (SGLT2) inhibitors canagliflozin and dapagliflozin showed robust renoprotective effects in large cardiovascular and renal outcomes trials [57,58]. Similarly, nonsteroidal mineralocorticoid receptor antagonist finerenone led to decreases in the risk of CKD progression and cardiovascular events [59]. However, significant residual risks still exist, e.g., finerenone-induced hyperkalemia, emphasizing the need for adjunctive agents with complementary mechanisms of action. GLP-1 receptor agonists are also promising drugs in the context of lowering the rates of DKD development and progression by virtue of their anti-inflammatory, antifibrotic, and natriuretic properties, in addition to their established metabolic and cardiovascular benefits. Of these, liraglutide has shown early evidence of renal benefits in outcome trials [16], and mechanistic and translational studies also support the kidney benefits of the GLP-1RA class [60,61]. Semaglutide is increasingly mentioned as having potential renal effects. While the evidence is less extensive than for liraglutide, injectable and oral formulations have yielded promising findings. Semaglutide should be considered in the context of other renoprotective medications, both to put its efficacy into perspective and to assess any additive or complementary value it may have.
There are two other drug classes already well known for their effects of slowing the progression of DKD: SGLT2 inhibitors and MRAs. SGLT2 inhibitors such as canagliflozin, dapagliflozin, and empagliflozin have demonstrated robust and consistent renal benefits in studies like CREDENCE, DAPA-CKD, and EMPA-KIDNEY [57,58,62]. These drugs reduce glomerular hyperfiltration via afferent arteriolar vasoconstriction caused by tubuloglomerular feedback. They also lower intraglomerular pressure, albuminuria, systemic blood pressure, and renal hypoxia. In the DAPA-CKD study, dapagliflozin reduced the composite risk of a sustained ≥50% eGFR reduction, ESKD, and renal or cardiovascular death by 39% compared with the placebo, even in non-diabetic individuals [58]. For semaglutide, in contrast, the renal benefits are mostly linked to decreases in albuminuria, with less evidence (so far) for direct eGFR protection. Semaglutide also has more generalized systemic effects, including dramatic weight loss, appetite regulation, and anti-inflammatory effects. While SGLT2 inhibitors have modest effects on weight and glycemia, GLP-1RAs have shown more pronounced effects on both weight and glycemia, particularly in those with obesity or suboptimal glycemic control, highlighting their potential as a comprehensive treatment option in DKD [26]. A nonsteroidal selective MRA, finerenone, also demonstrated renal and cardiovascular advantages in patients with DKD, as seen in the FIDELIO-DKD and FIGARO-DKD studies [59,63]. Finerenone has antifibrotic and anti-inflammatory effects through mineralocorticoid receptor antagonism, leading to an average reduction of about 18% in the development of ESKD or a persistent decrease in eGFR. Nevertheless, it is associated with an elevated risk of hyperkalemia and requires more intense monitoring. Semaglutide, on the other hand, is not reportedly linked to electrolyte imbalance and is safer in that regard.
Studies have explored the co-administration of GLP-1RA and SGLT2 inhibitors in T2DM, presenting evidence of their synergistic effects for enhanced metabolic, cardiovascular, and renal benefits, as well as additive effects on weight and blood pressure without worsening their safety profiles [64,65]. This could be interrupted by SGLT2 inhibitors’ action on renal hemodynamics and natriuresis and GLP-1Ras’ action on oxidative stress and systemic inflammation. This is supported by the American Diabetes Association (ADA) and Kidney Disease Improving Global Outcomes (KDIGO) [66,67]. However, direct comparisons between semaglutide and other renoprotective medications remain limited. The FLOW trial will be the first to establish whether semaglutide can match or exceed SGLT2 inhibitors’ or MRAs’ kidney benefits. Until then, it needs to be considered as a dual therapy in DKD, rather than an alternative therapy, particularly in those with obesity, significant cardiovascular risk, or intolerance to other medications. Table 4 summarizes the comparative mechanisms of action, renal outcomes, and safety profiles of semaglutide, SGLT2 inhibitors, and nonsteroidal MRAs.
Table 4.
Comparative summary of semaglutide, SGLT2 inhibitors, and MRAs in DKD.
4.5. Future Directions
Despite emerging data, there are substantial gaps in the knowledge to be addressed prior to supporting the use of semaglutide as a foundation therapy for diabetic kidney disease. At the forefront of these is the need for conclusive evidence on long-term renal outcomes. While declines in albuminuria are promising surrogate endpoints [68], the FLOW trial (NCT03819153) has provided a definition of the impact of semaglutide on the long-term trends in eGFR, ESKD rate, and renal mortality [50].
The efficacy and tolerability in advanced CKD (eGFR < 30 mL/min/1.73 m2) are not well investigated. Patients with ESRD are often excluded or underrepresented, but are most likely to require efficacious renoprotective therapy. Well-considered investigations of tolerability, dosing regimens, and the impacts on volume status in this cohort are needed.
The identification and validation of response biomarkers to semaglutide—comprising urinary kidney injury molecule-1 (KIM-1), neutrophil gelatinase-associated lipocalin (NGAL), and transforming growth factor-beta (TGF-β) levels, multi-omics, and imaging—would allow for precision medicine approaches to identify patients who would gain the maximum benefit and could be used to follow response to treatment and determine semaglutide’s mechanisms. Therefore, research needs to be conducted to investigate semaglutide’s renoprotective mechanism of action in humans, specifically for its anti-inflammatory, antifibrotic, and hemodynamic action. In addition, long-term renal biopsy studies, along with new imaging technologies, would also provide information at the tissue level. Moreover, the optimal combination and timing of semaglutide with SGLT2 inhibitors and MRA need to be determined in subsequent randomized controlled trials. Whether combination therapy’s benefits are additive or synergistic, or whether it simply causes side effects, would be useful to know in practice. Lastly, real-world evidence from diverse populations, many of which are comorbid and ethnically diverse, will supplement trial data and guide generalizability. In sum, semaglutide is highly promising for DKD management, but its definitive place, optimum utilization, and maximum patient benefits have yet to be confirmed through high-quality intensive research. Table 5 summarizes the current knowledge gaps and suggested areas for future investigation.
Table 5.
Knowledge gaps and research priorities for semaglutide in DKD.
5. Conclusions
DKD is a serious clinical issue that has high morbidity and mortality rates and is a large burden on healthcare systems, even with improvements in overall glycemic and blood pressure control. Semaglutide has shown promise as an agent not only for glycemic control and weight reduction, but also cardiovascular and renal protection. Preclinical evidence suggests that semaglutide has effects beyond the primary pathways of DKD pathogenesis, including effects on inflammation, oxidative stress, fibrosis, and endothelial dysfunction. In the clinic, cardiovascular outcome trials have demonstrated significant decreases in new or worsening nephropathy, driven primarily by decreased albuminuria.
Although the FLOW trial provides definitive evidence of semaglutide’s potential to retard kidney function decline and prevent ESKD, evidence in the literature indicates that it could be a valuable adjunct or alternative to current renoprotective agents such as SGLT2 inhibitors and mineralocorticoid receptor antagonists. Semaglutide’s pleiotropic effects, beneficial safety profile, and potential for combination therapy make it a candidate for managing combined diabetes and kidney disease. However, critical gaps in knowledge remain, such as its effectiveness in various stages of CKD, the best combination strategies, and its mechanisms of action in humans. Biomarker-based strategies and precision medicine have the potential to further optimize future applications. Overall, semaglutide targets metabolic, inflammatory, and fibrotic pathways in DKD and could be a promising therapeutic agent to change patient outcomes and reduce the worldwide burden of this disease.
Funding
This research was funded by Prince Sattam bin Abdulaziz University, project number (PSAU/2025/R/1447).
Data Availability Statement
No new data were created or analyzed in this study.
Acknowledgments
This study is supported via funding from Prince Sattam bin Abdulaziz University, project number (PSAU/2025/R/1447).
Conflicts of Interest
The author declares no conflicts of interest.
References
- Afkarian, M.; Sachs, M.C.; Kestenbaum, B.; Hirsch, I.B.; Tuttle, K.R.; Himmelfarb, J.; De Boer, I.H. Kidney disease and increased mortality risk in type 2 diabetes. J. Am. Soc. Nephrol. 2013, 24, 302–308. [Google Scholar] [CrossRef]
- Alicic, R.Z.; Rooney, M.T.; Tuttle, K.R. Diabetic kidney disease: Challenges, progress, and possibilities. Clin. J. Am. Soc. Nephrol. 2017, 12, 2032–2045. [Google Scholar] [CrossRef]
- Hoogeveen, E.K. The Epidemiology of Diabetic Kidney Disease. Kidney Dial. 2022, 2, 433–442. [Google Scholar] [CrossRef]
- Retnakaran, R.; Cull, C.A.; Thorne, K.I.; Adler, A.I.; Holman, R.R. Risk factors for renal dysfunction in type 2 diabetes: U.K. Prospective Diabetes Study 74. Diabetes 2006, 55, 1832–1839. [Google Scholar] [CrossRef] [PubMed]
- Chaudhuri, A.; Ghanim, H.; Arora, P. Improving the residual risk of renal and cardiovascular outcomes in diabetic kidney disease: A review of pathophysiology, mechanisms, and evidence from recent trials. Diabetes Obes. Metab. 2022, 24, 365–376. [Google Scholar] [CrossRef] [PubMed]
- Giugliano, D.; Maiorino, M.I.; Bellastella, G.; Esposito, K. The residual cardiorenal risk in type 2 diabetes. Cardiovasc. Diabetol. 2021, 20, 36. [Google Scholar] [CrossRef]
- Drucker, D.J. Mechanisms of Action and Therapeutic Application of Glucagon-like Peptide-1. Cell Metab. 2018, 27, 740–756. [Google Scholar] [CrossRef]
- Drucker, D.J.; Habener, J.F.; Holst, J.J. Discovery, characterization, and clinical development of the glucagon-like peptides. J. Clin. Investig. 2017, 127, 4217–4227. [Google Scholar] [CrossRef]
- Marso, S.P.; Bain, S.C.; Consoli, A.; Eliaschewitz, F.G.; Jódar, E.; Leiter, L.A.; Lingvay, I.; Rosenstock, J.; Seufert, J.; Warren, M.L.; et al. Semaglutide and Cardiovascular Outcomes in Patients with Type 2 Diabetes. N. Engl. J. Med. 2016, 375, 1834–1844. [Google Scholar] [CrossRef]
- Ahmann, A.; Capehorn, M.; Charpentier, G.; Dotta, F.; Henkel, E.; Lingvay, I.; Holst, A.G.; Annett, M.; Aroda, V. Efficacy and safety of once-weekly semaglutide vs exenatide ER after 56 Weeks in subjects with type 2 diabetes (SUSTAIN 3). Diabetes Res. Clin. Pract. 2016, 120, S51. [Google Scholar] [CrossRef]
- Shaman, A.M.; Bain, S.C.; Bakris, G.L.; Buse, J.B.; Idorn, T.; Mahaffey, K.W.; Mann, J.F.; Nauck, M.A.; Rasmussen, S.; Rossing, P.; et al. Effect of the Glucagon-Like Peptide-1 Receptor Agonists Semaglutide and Liraglutide on Kidney Outcomes in Patients With Type 2 Diabetes: Pooled Analysis of SUSTAIN 6 and LEADER. Circulation 2022, 145, 575–585. [Google Scholar] [CrossRef]
- Lau, J.; Bloch, P.; Schäffer, L.; Pettersson, I.; Spetzler, J.; Kofoed, J.; Madsen, K.; Knudsen, L.B.; McGuire, J.; Steensgaard, D.B.; et al. Discovery of the Once-Weekly Glucagon-Like Peptide-1 (GLP-1) Analogue Semaglutide. J. Med. Chem. 2015, 58, 7370–7380. [Google Scholar] [CrossRef]
- Knudsen, L.B.; Lau, J. The discovery and development of liraglutide and semaglutide. Front. Endocrinol. 2019, 10, 155. [Google Scholar] [CrossRef] [PubMed]
- Pratley, R.E.; Aroda, V.R.; Lingvay, I.; Lüdemann, J.; Andreassen, C.; Navarria, A.; Viljoen, A. Semaglutide versus dulaglutide once weekly in patients with type 2 diabetes (SUSTAIN 7): A randomised, open-label, phase 3b trial. Lancet Diabetes Endocrinol. 2018, 6, 275–286. [Google Scholar] [CrossRef] [PubMed]
- Husain, M.; Birkenfeld, A.L.; Donsmark, M.; Dungan, K.; Eliaschewitz, F.G.; Franco, D.R.; Jeppesen, O.K.; Lingvay, I.; Mosenzon, O.; Pedersen, S.D.; et al. Oral Semaglutide and Cardiovascular Outcomes in Patients with Type 2 Diabetes. N. Engl. J. Med. 2019, 381, 841–851. [Google Scholar] [CrossRef] [PubMed]
- Mann, J.F.; Ørsted, D.D.; Brown-Frandsen, K.; Marso, S.P.; Poulter, N.R.; Rasmussen, S.; Tornøe, K.; Zinman, B.; Buse, J.B. Liraglutide and Renal Outcomes in Type 2 Diabetes. N. Engl. J. Med. 2017, 377, 839–848. [Google Scholar] [CrossRef]
- Gerstein, H.C.; Colhoun, H.M.; Dagenais, G.R.; Diaz, R.; Lakshmanan, M.; Pais, P.; Probstfield, J.; Riesmeyer, J.S.; Riddle, M.C.; Rydén, L.; et al. Dulaglutide and cardiovascular outcomes in type 2 diabetes (REWIND): A double-blind, randomised placebo-controlled trial. Lancet 2019, 394, 121–130. [Google Scholar] [CrossRef]
- Jensen, L.; Helleberg, H.; Roffel, A.; van Lier, J.J.; Bjørnsdottir, I.; Pedersen, P.J.; Rowe, E.; Karsbøl, J.D.; Pedersen, M.L. Absorption, metabolism and excretion of the GLP-1 analogue semaglutide in humans and nonclinical species. Eur. J. Pharm. Sci. 2017, 104, 31–41. [Google Scholar] [CrossRef]
- Deacon, C.F.; Knudsen, L.B.; Madsen, K.; Wiberg, F.C.; Jacobsen, O.; Holst, J.J. Dipeptidyl peptidase IV resistant analogues of glucagon-like peptide-1 which have extended metabolic stability and improved biological activity. Diabetologia 1998, 41, 271–278. [Google Scholar] [CrossRef]
- Polonsky, W.H.; Arora, R.; Faurby, M.; Fernandes, J.; Liebl, A. Higher Rates of Persistence and Adherence in Patients with Type 2 Diabetes Initiating Once-Weekly vs Daily Injectable Glucagon-Like Peptide-1 Receptor Agonists in US Clinical Practice (STAY Study). Diabetes Ther. 2022, 13, 175–187. [Google Scholar] [CrossRef]
- Andersen, A.; Lund, A.; Knop, F.K.; Vilsbøll, T. Glucagon-like peptide 1 in health and disease. Nat. Rev. Endocrinol. 2018, 14, 390–403. [Google Scholar] [CrossRef]
- Gabery, S.; Salinas, C.G.; Paulsen, S.J.; Ahnfelt-Rønne, J.; Alanentalo, T.; Baquero, A.F.; Buckley, S.T.; Farkas, E.; Fekete, C.; Frederiksen, K.S.; et al. Semaglutide lowers body weight in rodents via distributed neural pathways. JCI Insight 2020, 5, e133429. [Google Scholar] [CrossRef] [PubMed]
- Smits, M.M.; Van Raalte, D.H. Safety of Semaglutide. Front. Endocrinol. 2021, 12, 645563. [Google Scholar] [CrossRef] [PubMed]
- Rodbard, H.W.; Lingvay, I.; Reed, J.; de la Rosa, R.; Rose, L.; Sugimoto, D.; Araki, E.; Chu, P.-L.; Wijayasinghe, N.; Norwood, P. Semaglutide Added to Basal Insulin in Type 2 Diabetes (SUSTAIN 5): A Randomized, Controlled Trial. J. Clin. Endocrinol. Metab. 2018, 103, 2291–2301. [Google Scholar] [CrossRef] [PubMed]
- Shu, Y.; He, X.; Wu, P.; Liu, Y.; Ding, Y.; Zhang, Q. Gastrointestinal adverse events associated with semaglutide: A pharmacovigilance study based on FDA adverse event reporting system. Front. Public Health 2022, 10, 996179. [Google Scholar] [CrossRef]
- Wilding, J.P.H.; Batterham, R.L.; Calanna, S.; Davies, M.; Van Gaal, L.F.; Lingvay, I.; McGowan, B.M.; Rosenstock, J.; Tran, M.T.; Wadden, T.A.; et al. Once-Weekly Semaglutide in Adults with Overweight or Obesity. N. Engl. J. Med. 2021, 384, 989–1002. [Google Scholar] [CrossRef]
- FDA. OZEMPIC (Semaglutide) Injection, for Subcutaneous Use Initial U.S. Approval: 2017 [Internet]. FDA-Approved Drugs—Drugs@FDA. 2017; p. 44. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/209637s003lbl.pdf (accessed on 1 January 2020).
- Ayoub, M.; Chela, H.; Amin, N.; Hunter, R.; Anwar, J.; Tahan, V.; Daglilar, E. Pancreatitis Risk Associated with GLP-1 Receptor Agonists, Considered as a Single Class, in a Comorbidity-Free Subgroup of Type 2 Diabetes Patients in the United States: A Propensity Score-Matched Analysis. J. Clin. Med. 2025, 14, 944. [Google Scholar] [CrossRef]
- Sharma, A.; Parachuri, N.; Kumar, N.; Saboo, B.; Tripathi, H.N.; Kuppermann, B.D.; Bandello, F. Semaglutide and the risk of diabetic retinopathy—Current perspective. Eye 2022, 36, 10–11. [Google Scholar] [CrossRef]
- Wang, F.; Mao, Y.; Wang, H.; Liu, Y.; Huang, P. Semaglutide and Diabetic Retinopathy Risk in Patients with Type 2 Diabetes Mellitus: A Meta-Analysis of Randomized Controlled Trials. Clin. Drug Investig. 2022, 42, 17–28. [Google Scholar] [CrossRef]
- Thomas, M.C.; Brownlee, M.; Susztak, K.; Sharma, K.; Jandeleit-Dahm, K.A.M.; Zoungas, S.; Rossing, P.; Groop, P.-H.; Cooper, M.E. Diabetic kidney disease. Nat. Rev. Dis. Primers 2015, 1, 15018. [Google Scholar] [CrossRef]
- Afkarian, M.; Zelnick, L.R.; Hall, Y.N.; Heagerty, P.J.; Tuttle, K.; Weiss, N.S.; De Boer, I.H. Clinical manifestations of kidney disease among US adults with diabetes, 1988–2014. JAMA—J. Am. Med. Assoc. 2016, 316, 602–610. [Google Scholar] [CrossRef]
- Fioretto, P.; Mauer, M. Histopathology of Diabetic Nephropathy. Semin. Nephrol. 2007, 27, 195–207. [Google Scholar] [CrossRef] [PubMed]
- Fioretto, P.; Steffes, M.W.; Mauer, M. Glomerular structure in nonproteinuric IDDM patients with various levels of albuminuria. Diabetes 1994, 43, 1358–1364. [Google Scholar] [CrossRef] [PubMed]
- Kanwar, Y.S.; Sun, L.; Xie, P.; Liu, F.Y.; Chen, S. A glimpse of various pathogenetic mechanisms of diabetic nephropathy. Annu. Rev. Pathol. Mech. Dis. 2011, 6, 395–423. [Google Scholar] [CrossRef] [PubMed]
- An, Z.; Qin, J.; Bo, W.; Li, H.; Jiang, L.; Li, X.; Jiang, J. Prognostic Value of Serum Interleukin-6, NF- κ B plus MCP-1 Assay in Patients with Diabetic Nephropathy. Dis. Markers 2022, 2022, 4428484. [Google Scholar] [CrossRef]
- Levin, A.; Stevens, P.E.; Bilous, R.W.; Coresh, J.; De Francisco, A.L.M.; De Jong, P.E.; Griffith, K.E.; Hemmelgarn, B.R.; Iseki, K.; Lamb, E.J.; et al. Kidney disease: Improving global outcomes (KDIGO) CKD work group. KDIGO 2012 clinical practice guideline for the evaluation and management of chronic kidney disease. Kidney Int. Suppl. 2013, 3, 1–150. [Google Scholar] [CrossRef]
- Penno, G.; Solini, A.; Bonora, E.; Fondelli, C.; Orsi, E.; Zerbini, G.; Trevisan, R.; Vedovato, M.; Gruden, G.; Cavalot, F.; et al. Clinical significance of nonalbuminuric renal impairment in type 2 diabetes. J. Hypertens. 2011, 29, 1802–1809. [Google Scholar] [CrossRef]
- Forbes, J.M.; Cooper, M.E. Mechanisms of diabetic complications. Physiol. Rev. 2013, 93, 137–188. [Google Scholar] [CrossRef]
- Navarro-González, J.F.; Mora-Fernández, C. The role of inflammatory cytokines in diabetic nephropathy. J. Am. Soc. Nephrol. 2008, 19, 433–442. [Google Scholar] [CrossRef]
- Chen, X.; Chen, S.; Ren, Q.; Niu, S.; Pan, X.; Yue, L.; Li, Z.; Zhu, R.; Jia, Z.; Chen, X.; et al. Metabolomics Provides Insights into Renoprotective Effects of Semaglutide in Obese Mice. Drug Des. Dev. Ther. 2022, 16, 3893–3913. [Google Scholar] [CrossRef]
- Tian, S.; Zhou, S.; Wu, W.; Lin, Y.; Wang, T.; Sun, H.; A-Ni-Wan, A.; Li, Y.; Wang, C.; Li, X.; et al. GLP-1 Receptor Agonists Alleviate Diabetic Kidney Injury via β-Klotho-Mediated Ferroptosis Inhibition. Adv. Sci. 2025, 12, e2409781. [Google Scholar] [CrossRef]
- Dalbøge, L.S.; Christensen, M.; Madsen, M.R.; Secher, T.; Endlich, N.; Drenic’, V.; Manresa-Arraut, A.; Hansen, H.H.; Rune, I.; Fink, L.N.; et al. Nephroprotective Effects of Semaglutide as Mono- and Combination Treatment with Lisinopril in a Mouse Model of Hypertension-Accelerated Diabetic Kidney Disease. Biomedicines 2022, 10, 1661. [Google Scholar] [CrossRef]
- Shan, S.; Alanazi, A.H.; Han, Y.; Zhang, D.; Liu, Y.; Narayanan, S.P.; Somanath, P.R. Pro-Inflammatory Characteristics of Extracellular Vesicles in the Vitreous of Type 2 Diabetic Patients. Biomedicines 2024, 12, 2053. [Google Scholar] [CrossRef]
- McLean, B.A.; Wong, C.K.; Kaur, K.D.; Seeley, R.J.; Drucker, D.J. Differential importance of endothelial and hematopoietic cell GLP-1Rs for cardiometabolic versus hepatic actions of semaglutide. JCI Insight 2021, 6, e153732. [Google Scholar] [CrossRef]
- Farah, L.X.S.; Valentini, V.; Pessoa, T.D.; Malnic, G.; McDonough, A.A.; Girardi, A.C.C. The physiological role of glucagon-like peptide-1 in the regulation of renal function. Am. J. Physiol. Physiol. 2015, 310, F123–F127. [Google Scholar] [CrossRef]
- Mann, J.F.E.; Hansen, T.; Idorn, T.; A Leiter, L.; Marso, S.P.; Rossing, P.; Seufert, J.; Tadayon, S.; Vilsbøll, T. Effects of once-weekly subcutaneous semaglutide on kidney function and safety in patients with type 2 diabetes: A post-hoc analysis of the SUSTAIN 1–7 randomised controlled trials. Lancet Diabetes Endocrinol. 2020, 8, 880–893. [Google Scholar] [CrossRef]
- Mosenzon, O.; Blicher, T.M.; Rosenlund, S.; Eriksson, J.W.; Heller, S.; Hels, O.H.; Pratley, R.; Sathyapalan, T.; Desouza, C.; Abramof, R.; et al. Efficacy and safety of oral semaglutide in patients with type 2 diabetes and moderate renal impairment (PIONEER 5): A placebo-controlled, randomised, phase 3a trial. Lancet Diabetes Endocrinol. 2019, 7, 515–527. [Google Scholar] [CrossRef]
- Sattar, N.; Lee, M.M.Y.; Kristensen, S.L.; Branch, K.R.H.; Del Prato, S.; Khurmi, N.S.; Lam, C.S.P.; Lopes, R.D.; McMurray, J.J.V.; Pratley, R.E.; et al. Cardiovascular, mortality, and kidney outcomes with GLP-1 receptor agonists in patients with type 2 diabetes: A systematic review and meta-analysis of randomised trials. Lancet Diabetes Endocrinol. 2021, 9, 653–662. [Google Scholar] [CrossRef]
- Rossing, P.; Baeres, F.M.M.; Bakris, G.; Bosch-Traberg, H.; Gislum, M.; Gough, S.C.L.; Idorn, T.; Lawson, J.; Mahaffey, K.W.; E Mann, J.F.; et al. The rationale, design and baseline data of FLOW, a kidney outcomes trial with once-weekly semaglutide in people with type 2 diabetes and chronic kidney disease. Nephrol. Dial. Transplant. 2023, 38, 2041–2051. [Google Scholar] [CrossRef]
- Perkovic, V.; Tuttle, K.R.; Rossing, P.; Mahaffey, K.W.; Mann, J.F.; Bakris, G.; Baeres, F.M.; Idorn, T.; Bosch-Traberg, H.; Lausvig, N.L.; et al. Effects of Semaglutide on Chronic Kidney Disease in Patients with Type 2 Diabetes. N. Engl. J. Med. 2024, 391, 109–121. [Google Scholar] [CrossRef]
- Tuttle, K.R.; Bosch-Traberg, H.; Cherney, D.Z.; Hadjadj, S.; Lawson, J.; Mosenzon, O.; Rasmussen, S.; Bain, S.C. Post hoc analysis of SUSTAIN 6 and PIONEER 6 trials suggests that people with type 2 diabetes at high cardiovascular risk treated with semaglutide experience more stable kidney function compared with placebo. Kidney Int. 2023, 103, 772–781. [Google Scholar] [CrossRef]
- Luna, E.; Álvarez, Á.; Rodriguez-Sabiñón, J.; Villa, J.; Giraldo, T.; Martín, M.V.; Vázquez, E.; Fernández, N.; Ruiz, B.; Garcia-Pino, G.; et al. Long-Term Effect of Semaglutide on the Glomerular Filtration Rate Slope in High-Risk Patients with Diabetic Nephropathy: Analysis in Real-World Clinical Practice. Pharmaceutics 2025, 17, 943. [Google Scholar] [CrossRef]
- Bueno, B.A.; Soler, M.J.; Perez-Belmonte, L.; Millan, A.J.; Ruiz, F.R.; de Lucas, M.D.G. Semaglutide in type 2 diabetes with chronic kidney disease at high risk progression-real-world clinical practice. Clin. Kidney J. 2022, 15, 1593–1600. [Google Scholar] [CrossRef]
- ADVANCE Collaborative Group. Intensive Blood Glucose Control and Vascular Outcomes in Patients with Type 2 Diabetes. N. Engl. J. Med. 2008, 358, 2560–2572. [Google Scholar] [CrossRef] [PubMed]
- ACCORD Study Group. Effects of Intensive Glucose Lowering in Type 2 Diabetes. N. Engl. J. Med. 2008, 8, 2545–2559. [Google Scholar] [CrossRef]
- Perkovic, V.; Jardine, M.J.; Neal, B.; Bompoint, S.; Heerspink, H.J.L.; Charytan, D.M.; Edwards, R.; Agarwal, R.; Bakris, G.; Bull, S.; et al. Canagliflozin and Renal Outcomes in Type 2 Diabetes and Nephropathy. N. Engl. J. Med. 2019, 380, 2295–2306. [Google Scholar] [CrossRef] [PubMed]
- Heerspink, H.J.L.; Stefánsson, B.V.; Correa-Rotter, R.; Chertow, G.M.; Greene, T.; Hou, F.-F.; Mann, J.F.E.; McMurray, J.J.V.; Lindberg, M.; Rossing, P.; et al. Dapagliflozin in Patients with Chronic Kidney Disease. N. Engl. J. Med. 2020, 383, 1436–1446. [Google Scholar] [CrossRef]
- Bakris, G.L.; Agarwal, R.; Anker, S.D.; Pitt, B.; Ruilope, L.M.; Rossing, P.; Kolkhof, P.; Nowack, C.; Schloemer, P.; Joseph, A.; et al. Effect of Finerenone on Chronic Kidney Disease Outcomes in Type 2 Diabetes. N. Engl. J. Med. 2020, 383, 2219–2229. [Google Scholar] [CrossRef]
- Muskiet, M.H.A.; Tonneijck, L.; Smits, M.M.; Van Baar, M.J.B.; Kramer, M.H.H.; Hoorn, E.J.; Joles, J.A.; Van Raalte, D.H. GLP-1 and the kidney: From physiology to pharmacology and outcomes in diabetes. Nat. Rev. Nephrol. 2017, 13, 605–628. [Google Scholar] [CrossRef]
- Campbell, J.E.; Drucker, D.J. Pharmacology, physiology, and mechanisms of incretin hormone action. Cell Metab. 2013, 17, 819–837. [Google Scholar] [CrossRef]
- EMPA-Kidney Collaborative Group. Empagliflozin in Patients with Chronic Kidney Disease. N. Engl. J. Med. 2023, 388, 117–127. [Google Scholar] [CrossRef] [PubMed]
- Pitt, B.; Filippatos, G.; Agarwal, R.; Anker, S.D.; Bakris, G.L.; Rossing, P.; Joseph, A.; Kolkhof, P.; Nowack, C.; Schloemer, P.; et al. Cardiovascular Events with Finerenone in Kidney Disease and Type 2 Diabetes. N. Engl. J. Med. 2021, 385, 2252–2263. [Google Scholar] [CrossRef]
- DeFronzo, R.A. Combination therapy with GLP-1 receptor agonist and SGLT2 inhibitor. Diabetes Obes. Metab. 2017, 19, 1353–1362. [Google Scholar] [CrossRef]
- Gourdy, P.; Darmon, P.; Dievart, F.; Halimi, J.M.; Guerci, B. Combining glucagon-like peptide-1 receptor agonists (GLP-1RAs) and sodium-glucose cotransporter-2 inhibitors (SGLT2is) in patients with type 2 diabetes mellitus (T2DM). Cardiovasc. Diabetol. 2023, 22, 79. [Google Scholar] [CrossRef]
- American Diabetes Association Professional Practice Committee. Cardiovascular Disease and Risk Management: Standards of Medical Care in Diabetes—2022. Diabetes Care 2022, 45 (Suppl. S1), S144–S174. [Google Scholar] [CrossRef]
- Rossing, P. KDIGO 2020 Clinical Practice Guideline for Diabetes Management in Chronic Kidney Disease. Kidney Int. Suppl. 2020, 98, S1–S115. [Google Scholar] [CrossRef]
- Heerspink, H.J.L.; Greene, T.; Tighiouart, H.; Gansevoort, R.T.; Coresh, J.; Simon, A.L.; Chan, T.M.; Hou, F.F.; Locatelli, F.; Praga, M.; et al. Change in albuminuria as a surrogate endpoint for progression of kidney disease: A meta-analysis of treatment effects in randomised clinical trials. Lancet Diabetes Endocrinol. 2019, 7, 128–139. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the author. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).