Skip to main content
Clinical Kidney Journal logoLink to Clinical Kidney Journal
. 2022 May 19;15(12):2200–2213. doi: 10.1093/ckj/sfac143

Repurposing drugs for highly prevalent diseases: pentoxifylline, an old drug and a new opportunity for diabetic kidney disease

Javier Donate-Correa 1,2,3, María Dolores Sanchez-Niño 4, Ainhoa González-Luis 5,6, Carla Ferri 7,8, Alberto Martín-Olivera 9,10, Ernesto Martín-Núñez 11,12, Beatriz Fernandez-Fernandez 13,14, Víctor G Tagua 15, Carmen Mora-Fernández 16,17,18,b, Alberto Ortiz 19,20,b, Juan F Navarro-González 21,22,23,24,25,
PMCID: PMC9664582  PMID: 36381364

ABSTRACT

Diabetic kidney disease is one of the most frequent complications in patients with diabetes and constitutes a major cause of end-stage kidney disease. The prevalence of diabetic kidney disease continues to increase as a result of the growing epidemic of diabetes and obesity. Therefore, there is mounting urgency to design and optimize novel strategies and drugs that delay the progression of this pathology and contain this trend. The new approaches should go beyond the current therapy focussed on the control of traditional risk factors such as hyperglycaemia and hypertension. In this scenario, drug repurposing constitutes an economic and feasible approach based on the discovery of useful activities for old drugs. Pentoxifylline is a nonselective phosphodiesterase inhibitor currently indicated for peripheral artery disease. Clinical trials and meta-analyses have shown renoprotection secondary to anti-inflammatory and antifibrotic effects in diabetic patients treated with this old known drug, which makes pentoxifylline a candidate for repurposing in diabetic kidney disease.

Keywords: diabetes, diabetic kidney disease, pentoxifylline, repurposing

DIABETIC KIDNEY DISEASE, AN INCREASING PROBLEM

Diabetes mellitus (DM) is a world epidemic that affects ˃425 million people according to the International Diabetes Federation [1]. Recent estimates from this organization predict a prevalence of ˃630 million people with DM by the year 2045 [1]. One of the most relevant complications of DM is diabetic kidney disease (DKD) which occurs in ˃40% of diabetic patients, with no difference between patients with type 1 or type 2 DM [2–4]. Metabolic and haemodynamic insults drive the pathophysiology of DKD causing the deterioration of kidney functions. Until recently, chronic kidney disease (CKD) derived from DM was diagnosed as diabetic nephropathy, which begins with microalbuminuria, followed by a gradual decline in kidney function and overt macroalbuminuria. However, the report of patients with DM and impaired renal function without albuminuria has led to the concept of DKD. DKD is defined as CKD with diabetes being partially involved in the pathogenesis of kidney disease, encompassing the concept of classical diabetic nephropathy [5–8]. Despite advances in therapeutics, healthcare structures and overall population health, DKD is the single most common cause of end-stage kidney disease (ESKD) [9, 10]. Patients with DKD present 20–40 times higher cardiovascular morbidity and mortality rates than patients with DM without kidney impairment; in fact, most patients with DKD die from cardiovascular disease before they start renal replacement therapy.

As a consequence of the ever-growing epidemic of diabetes and obesity, the absolute number of people with ESKD continues to rise [11]. This situation has made the prevention and treatment of DKD a global challenge and a threat to human health and mortality, with a significant social and economic burden [12, 13]. At present, there are no specific therapeutic strategies for DKD, which makes finding new approaches a formidable challenge for the scientific community, since simple control of risk factors is insufficient to cope with disease progression. In search for new therapies, researchers have explored several drug-repurposing opportunities [14].

The pathogenesis of DKD comprises metabolic (hyperglycaemia, dyslipidaemia) and haemodynamic (glomerular hypertension) perturbations which, together, cause mesangial expansion, impairment of endothelial cell function and loss of podocytes in the glomerulus and interstitial fibrosis in the tubular compartment [15–17]. However, the full pathogenesis of the disease remains to be understood, and specific therapeutic targets have not been determined. Current practice guidelines are focussed on halting or delaying the progression of DKD through nonspecific multidisciplinary therapeutic approaches based on an adequate metabolic control and in the control of blood pressure with the renin-angiotensin system (RAS) blockade as a cornerstone therapy [18, 19]. Although this approach improves the systemic blood pressure as well as intraglomerular pressure, a key driver of albuminuria and CKD progression and also decreases kidney inflammation and fibrosis [20, 21], it does not generally halt the progression to ESKD. Moreover, the combination of RAS blockers such as angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin receptor blockers (ARBs) did not improve monotherapy results and is associated with adverse events including hyperkalaemia, acute kidney injury and hypotension [22–25]. Importantly, sodium-glucose cotransporter 2 inhibitors (SGLT2i) have been recently added to these multidisciplinary treatments, as drugs of choice for DKD treatment [26]. Although the evidence demonstrates renoprotection with the use of SGLT2i on top of RAS blockade, patients with DM continue to suffer from kidney disease and a high percentage of them progress to ESKD. Therefore, there is a need to evaluate new strategies to improve kidney function, delay the progression of the disease and eventually improve kidney survival. These new therapeutic approaches become even more necessary if we consider that recent trials designed to find effective renoprotection in DM patients have failed or were prematurely stopped because of safety concerns; i.e. ruboxistaurin and sulodexide failed to show clear-cut renoprotection in patients with type 2 DM and clinical trials with avosentan and bardoxolone methyl were prematurely terminated because of serious safety concerns [25, 27–31]. The efforts are focussed on targeting key mechanisms involved in the onset and progression of DKD including hyperglycaemia, oxidative stress [32], inflammation [33] and fibrosis [34].

The drug pentoxifylline is a methyl-xanthine derivative and a nonselective phosphodiesterase inhibitor with anti-inflammatory, antiproliferative and antifibrotic actions currently indicated for peripheral artery disease. Clinical trials and meta-analyses have shown renoprotection secondary to anti-inflammatory and antifibrotic effects in diabetic patients treated with pentoxifylline when added to RAS blockade, making pentoxifylline a potential candidate for repurposing in DKD [35].

EMERGING THERAPIES AND POTENTIAL REPURPOSED DRUGS IN DKD

In recent years, promising nephroprotective therapeutic strategies have arisen with the use of new antidiabetic drugs on top of RAS blockade. As discussed above, the current main pharmacological agents in DKD are RAS blockers and SGLT2i. SGLT2i are anti-hyperglycaemic agents that block glucose reabsorption by SGLT2 channels at proximal tubules, thereby stimulating glucosuria and decreasing blood glucose levels in an insulin-independent fashion [36]. But, beyond glycaemic control, secondary outcome analyses in cardiovascular safety randomized controlled trials (RCTs) in type 2 DM patients have shown improved kidney outcomes in patients with CKD [26, 37, 38]. As a result of this evidence, recent consensus documents have placed SGLT2i as antidiabetic drugs of choice on top of RAS blockade for type 2 DM patients with evidence of kidney disease [39, 40]. Despite this success, renal decline still continues in many individuals with diabetes and incident or worsening nephropathy occurs in 12.7% of individuals treated with empagliflozin [37] and new treatments are needed.

The unexpected nephroprotective success of SGLT2i in DKD has not been replicated and a large number of drugs, even with added RAS blockade, have failed [41]. New drug candidates include the groups of steroidal and nonsteroidal mineralocorticoid receptor antagonists (MRA). MRAs exert antihypertensive actions by suppressing the action of aldosterone, the end product of RAS, and has been reported to decrease proteinuria [42–47]. Two groups of anti‐diabetic drugs that could present nephroprotective effects, possibly independently of the glycaemic control, are the glucagon‐like peptide‐1 (GLP‐1) receptor agonists (GLP‐1RA) and the dipeptidyl peptidase‐4 (DPP‐4) inhibitors [48, 49]. These incretin-based drugs decrease albuminuria in DKD patients, but controversy persists over their potential to slow the rate of estimated glomerular filtration rate (eGFR) decline [50–59].

The effectiveness of inhibiting advanced glycation end product (AGE) accumulation has been also conducted. AGE accumulation in kidney samples correlates with DKD progression and, at present, the administration of AGE inhibitors in DKD patients is the focus of clinical and basic research, with controversial results in a decrease of proteinuria and in the progression of GFR decline [60–63].

With the exception of SGLT2i and finerenone, there have been no new therapies for the treatment of nephropathy in type 2 DM since the approval of irbesartan and losartan by the Food and Drug Administration (FDA) ˃ 15 years ago. There is a desperate need to identify treatments for DKD, and several large-scale trials in people with DKD have been conducted and failed [24, 25, 29, 30]. In this sense, together with new antidiabetic drugs, drug repurposing is an alternative to de novo drug discovery, to find promising candidates to treat DKD. Drug repurposing offers multiple advantages, such as an accelerated and inexpensive drug development process. This approach decreases development risks, since the safety of the compound, which is one of the main reasons for high attrition rates, is already well established [35, 64, 65].

The strategy of drug repurposing has been widely employed in recent times during the coronavirus disease 2019 (COVID-19) pandemic, witnessing the evaluation and use of several existing molecules for their therapeutic potential against coronaviruses including hydroxychloroquine, remdesivir, ivermectin, lopinavir/ritonavir, baricitinib, dexamethasone and others [66]. Well-known examples of drug repositioning include thalidomide, which was used to prevent morning sickness and posteriorly repositioned for the treatment of multiple myeloma [67]; minoxidil and finasteride, initially approved for the treatment of hypertension and benign prostate hyperplasia, respectively, were repurposed for the treatment of male pattern baldness.

Methyl bardoxolone is a semi-synthetic triterpenoid with anti-inflammatory effects [68]. Methyl bardoxolone, initially studied for the prevention and treatment of cancer, was repurposed for other diseases with an inflammatory component including DKD following the observation of decreased serum creatinine in cancer patients [69, 70]. These promising results led to the Bardoxolone Methyl Evaluation in Patients with Chronic Kidney Disease and Type 2 Diabetes Mellitus: The Occurrence of Renal Events (BEACON NCT01351675) phase III clinical trial [30], which included 2185 participants with type 2 DM. Although this trial was terminated due to serious adverse events originating from high rates of heart failure-related hospitalizations and deaths in patients treated with bardoxolone, post hoc analyses showed that the increase in heart failure events was most likely caused by fluid overload in the first 4 weeks after randomization [71]. Moreover, elevated baseline B-type natriuretic peptide (BNP) levels (>200 pg/mL) and a history of hospitalization were identified as the only risk factors for heart failure. Patients without these two risk factors showed the same incidence of heart failure in the bardoxolone methyl and in the placebo groups (2%) [72]. The Phase 2 Study of Bardoxolone Methyl in Patients with Chronic Kidney Disease and Type 2 Diabetes (TSUBAKI, NCT02316821) [73] for the treatment of CKD in Japanese patients without these clinical characteristics, again indicated an increase in the measured GFR in patients treated with methyl-bardoxolone without cases of death or heart failure in any participant.

Other anti-inflammatory agents repurposed for DKD include CCX-140 and bariticinib, both originally developed for rheumatoid arthritis. CCX140-B is an inhibitor of C-C chemokine receptor type 2 (CCR2) that decreases macrophage migration and activation that was repurposed for DKD after the results of a phase II RCT showing kidney-protective effects in patients with type 2 DM when administered on top of standard medication [74]. Administration of baricitinib, which selectively inhibits Janus kinase 1 and 2 (JAK1 and JAK2), has been recently tested in a phase II RCT including 129 DKD patients, finding a decrease in albuminuria [75].

Endothelin A is a vasoactive peptide that exerts vasoconstrictive actions of glomerular afferent and efferent arterioles, crucial determinants of glomerular haemodynamics, which leads to a decrease in GFR [76] and also generates kidney injury via inflammation, endothelial injury, podocyte disruption and fibrosis. Endothelin A receptor antagonists were first evaluated in men with metastatic hormone-refractory prostate cancer [77] and are currently approved for the treatment of pulmonary arterial hypertension [78]. The endothelin A receptor antagonists atrasentan decreases proteinuria in experimental kidney disease [79], which has led to clinical testing in DKD [80–82]. In DKD, atrasentan decreased blood pressure and albuminuria when added to stable RAS blockade, but was associated with fluid overload and heart failure exacerbation [83].

Finally, pentoxifylline has recently been added to this group of potentially repurposed kidney protective drugs based on its anti-inflammatory and antiproteinuric effects. Pentoxifylline is currently indicated for peripheral artery disease, but open-label trials have shown beneficial results in DKD and also in nonspecific CKD and chronic allograft nephropathy. Along with the decrease in albuminuria and inflammation, the deceleration in the GFR decline rate and the preservation of the anti-ageing factor Klotho are the most important findings in DKD patients treated with pentoxifylline [84–86].

PENTOXIFYLLINE IN DKD

An old-new friend

Pentoxifylline [3,7‐dimethyl‐1‐(5‐oxohexyl)‐3,7‐dihydro‐1H‐purine‐2,6‐dione] is a promising anti-inflammatory methyl‐xanthine derivative with haemorheological actions. Pentoxifylline was approved by the United States FDA for the treatment of intermittent claudication resulting from peripheral vascular disease ˃30 years ago [87–89]. This drug decreases blood viscosity, erythrocyte aggregation, erythrocyte rigidity and platelet aggregation. The improvement in red blood cell flexibility and deformability leads to an improved blood flow [89, 90]. The pharmacological properties of pentoxifylline have been frequently revisited, and recent evidence indicates other possible beneficial effects of this old drug [91]. Thus, the repurposing of pentoxifylline has been suggested for treating brain ischaemia, non-alcoholic fatty liver diseases and preserving skeletal muscle function [90].

The haemorheological properties and its potential to decrease intraglomerular pressure led to an early interest in pentoxifylline as a therapeutic agent in kidney disease. In 1982, Blagosklonnaia et al. [92] presented the first clinical evidence of kidney protective effects of pentoxifylline. Diabetic patients treated with 300 mg/day of pentoxifylline for 3 weeks improved eGFR and decreased proteinuria. The possible application of pentoxifylline for kidney protection in DKD was recently renewed as studies showed pentoxifylline anti-inflammatory, anti-proliferative and anti-fibrotic effects [93, 94] (Table 1).

Table 1.

Main clinical and experimental studies on the use of pentoxifylline in DKD

Ref. Type of study Type of intervention Population Treatment Main effects of PTF
Clinical studies
 [84] RCT PTF versus untreated DM patients, n = 169
Albuminuria >30 mg/24 h,
eGFR 60–15 mL/min/1.73 m2
1200 mg/day, 2 years. Background RAS blockade Improvements in GFR decay and in proteinuria. Decrease of urinary levels of TNFα
 [95] Open-label RCT PTF versus untreated DM patients, n = 24
Albuminuria >300 mg/24 h; creatinine clearance <35 mL/min
400 mg/day, 6 months Decrease in proteinuria and TNFα levels
 [96] Open-label RCT PTF versus captopril DM patients, n = 39
Albuminuria >300 mg/24 h; eGFR >60 mL/min/1.73 m2
1200 mg/day, 8 weeks Both PTF and captopril decreased proteinuria
 [97] Open-label RCT PTF versus captopril DM patients, n = 130
UAE 20–200 μg/min
1200 mg/day, 6 months Both PTF and captopril decreased proteinuria
 [98] Open-label RCT PTF versus untreated DM patients, n = 61
Albuminuria >300 mg/24 h;
eGFR >90 mL/min/1.73 m2
1200 mg/day,
4 months. Background RAS blockade
Decrease in serum and urinary TNFα levels. TNFα changes related to UAE
 [99] Double-blind RCT PTF versus placebo DM patients, n = 40
UAE 20–200 μg/min
1200 mg/day,
4 months
Decrease in urinary levels of high and low molecular weight proteins
 [100] Double-blind RCT PTF versus placebo Patients with GN, n = 18
proteinuria >500 mg/24 h, mean eGFR 71.2 ± 30.6 mL/min/1.73 m2
800–1200 mg/day,
6 months. Background RAS blockade
Decrease of proteinuria without affecting GFR
 [101] Open-label, controlled trial PTF versus untreated Diabetic GE patients, n = 14
Proteinuria >1.5 g/24 h;
Cr clearance >15 mL/min
400–800 mg/day, 1 year. Background RAS blockade PTF did not decrease proteinuria or improved renal function
 [102] Double-blind RCT PTF versus placebo CKD patients, n = 40
Mean eGFR 29.5 ± 10.1 mL/min/1.73 m2
Proteinuria >1 g/24 h
800 mg/day, 1 year
Background RAS blockade
PTF stabilized GFR. No decrease of proteinuria
 [103] RCT PTF versus untreated CKD patients, n = 91
albuminuria >300 mg/24 h, eGFR <60 mL/min/1.73 m2
800 mg/day, 1 year. Background RAS blockade PTF stabilized GFR. No decrease of proteinuria. Decrease in TNFα, fibrinogen and hsCRP
 [104] RCT PTF versus untreated CKD patients, n = 56
Proteinuria >500 mg/g of Cr; eGFR 10–60 mL/min/1.73 m2
400–800 mg/day, 1 year. Background RAS blockade Decrease of proteinuria and stabilization of GFR. Decrease in TNFα and MCP1 levels
 [105] Prospective trial All in PTF Patients with GN; non-diabetic, n = 17. Spot proteinuria >1.5 g/g Cr; eGFR 24–115 mL/min/1.73 m2 800 mg/day, 6 months Decrease in spot and 24 h proteinuria (g/g Cr) and in MCP-1 levels
 [106] Single-centre retrospective study PTF versus untreated CKD patients, n = 661
Mean proteinuria 1102 mg/g of Cr, eGFR <45 mL/min/1.73 m2
400–800 mg/day,
1 year. Background RAS blockade
Better renal outcome in patients with higher proteinuria
 [107] Prospective trial All in PTF CAN patients, n = 17
UAE 20–200 μg/min, mean eGFR 38 ± 8 mL/min/1.73 m2
1200 mg/day, 6 months Decrease of proteinuria at 3rd month and improved graft survival. Decrease in CD4 + cells bearing TNFα and IL10
 [85] RCT post-hoc analysis PTF versus untreated DM patients, n = 166
Albuminuria >30 mg/24 h,
eGFR 60–15 mL/min/1.73 m2
1200 mg/day,
2 years. Background RAS blockade
Increments in serum and urine Klotho. Changes in TNFα associated with changes of urinary Klotho
Ref. Experimental model Treatment conc. Main effects of PTF
Experimental studies
 [108] RAW 264.7 macrophages 100 µg/mL, 1 h before the addition of LPS Inhibition of endotoxin-induced TNFα synthesis
 [109] Rat model of crescentic GN Intravenous 0.1 g/kg/day Suppression of progressive renal injury through inhibition of renal TNFα, ICAM-1, RANTES, MCP-1 and OPN
 [110] Streptozotocin-induced diabetic rat model Intraperitoneal 25 mg/kg/day Decrease in renal TNFα and IL6 and amelioration of renal hypertrophy and sodium retention
 [111] Alloxan-induced diabetic rat model Oral 25, 50 or 100 mg/kg/day Decrease in renal TNFα and IL6
 [85] Renal tubular cells 4, 40, 200, 400 or 800 µg/mL, 1 h before the addition of albumin Upregulation and prevention of albuminuria-induced downregulation of expression of Klotho

RCT, randomized controlled trial; PTF, pentoxifylline; DM, diabetes mellitus; CKD, chronic kidney disease; RAS, renin-angiotensin aldosterone system; TNF, tumour necrosis factor; GE, glomeruloesclerosis; GN, glomerulonephritis; GFR, glomerular filtration rate; hsCRP, high sensitivity C-reactive protein; MCP1, monocyte chemoattractant protein 1; ICAM-1, intercellular adhesion molecule-1; OPN, osteopontin; CAN, chronic allograft nephropathy; UAE, urinary albumin excretion.

A series of five open-label clinical trials conducted between 1999 and 2006 focussed on the potential kidney protective effects of pentoxifylline in DKD (Table 1). First, Navarro et al. [95] reported a 42.2 and 59.3% decrease in serum tumour necrosis factor α (TNFα) and proteinuria levels, respectively, in a small group of patients with DM and advanced CKD under pentoxifylline treatment (400 mg/day; 6 months) when compared with a control group. Posteriorly, two open-label RCTs conducted by Aminorroaya et al. [96] and Rodríguez-Morán et al. [97] also reported a decrease in proteinuria in non-hypertensive type 2 DM patients with microalbuminuria comparable with those achieved with ACEI treatment (captopril) after the administration of 400 mg pentoxifylline three times a day (t.i.d.) for 2 (40% in pentoxifylline-group and 38.5% in captopril-group) and 6 months (77.2% in pentoxifylline-group and 76.6% in captopril-group), respectively. In a subsequent randomized, open-label trial, Navarro et al. [98] found an additive percentage decrease in proteinuria of 11.2% in those ARB-treated DM patients who also received 1200 mg/day pentoxifylline for 4 months; i.e. patients receiving pentoxifylline. Pentoxifylline treatment also decreased both serum and urinary levels of TNFα, without significant variations in patients exclusively under therapy with ARB. The antiproteinuric effect of pentoxifylline correlated with a decrease in urinary TNFα levels [98]. Finally, a subsequent RCT by Rodríguez-Morán et al. [99] newly reported a decrease in the levels of both high and low molecular weight urinary protein excretion (73.8 and 86.4% decrease, respectively) in non-hypertensive microalbuminuric type 2 DM patients treated with 400 mg pentoxifylline (t.i.d. for 16 weeks) not receiving ACEi or ARB therapy. An RCT published by Badri et al. [100] showed a 56% decrease in proteinuria in a small group of non-diabetic patients with glomerulonephritis with add-on pentoxifylline therapy to the background RAS blockade without affecting eGFR. Other clinical trials with different study designs, drug dosages and follow-up periods, also examined the kidney protective effects of pentoxifylline with generally inconclusive results. An open-label controlled clinical trial conducted by Diskin et al. [101] did not find any additive antiproteinuric effect of pentoxifylline in diabetic glomerulosclerosis patients with a background of ACEI and ARB therapy after 1 year of follow-up. Important concerns of this study are its non-randomized design, the small number of participants (14 patients) and the use of dual RAS blockade, which has important safety concerns [25, 112]. In a double-blind RCT, Perkins et al. [102] also found no differences in proteinuria in 40 DKD patients with mild to moderate CKD after 1 year of add-on pentoxifylline therapy to RAS blockade. However, they observed deceleration in renal function decline in the group treated with pentoxifylline when compared with the control group, with a mean difference between groups of 6.0 mL/min/1.73 m2, and argued that the proteinuria may not always constitute an optimal surrogate outcome parameter in these studies.

To date, the most important RCT evaluating the kidney protective effects of pentoxifylline in DKD is the Pentoxifylline for Renoprotection in Diabetic Nephropathy (PREDIAN) study, published in 2015 by Navarro Gonzalez et al. [84]. The study comprised 169 type 2 DM patients with CKD stages 3 and 4 and residual albuminuria despite RAS blockade. After 2 years of follow-up, patients randomized to the active group (1200 mg/day of pentoxifylline on top of RAS blockade) presented a decrease in the rate of progression of kidney disease, with an eGFR mean difference between groups of 4.3 mL/min/1.73 m2, accompanied by a 14.9% decrease in proteinuria (increased by 5.7% in the control group). The deceleration in the decline of GFR in the pentoxifylline arm began at month 6 and reached statistical significance after 1 year, suggesting that the therapeutic benefit may only be observed in the long term. Moreover, urine TNFα presented a 10.6% decrease in the pentoxifylline group, with no changes in the control group.

At present, the identification of the central role of inflammation in the development and progression of CKD and its possible therapeutic targets constitutes an important field of research for nephrologists. The anti-inflammatory actions elicited by pentoxifylline have been related to antialbuminuric effects [93, 113–118]. In this regard, an antiproteinuric or kidney function preservation effect of pentoxifylline has also been found in non-diabetic subjects. Goicoechea et al. [103] reported stabilization of kidney function and a significant decrease in markers of inflammation, such as TNFα, fibrinogen and high sensitivity C-reactive protein (CRP; a 45.5, 11.1 and 57.4% decrease, respectively) in patients with stage 3 CKD or higher who received pentoxifylline therapy when compared with those exclusively on RAS blockade. Proteinuria did not decrease in the pentoxifylline group, although there was a drop-out and incomplete follow-up rate. Lin et al. [104] found that pentoxifylline on top of ARB background therapy stabilized GFR and decreased proteinuria (−23.9%) in macroalbuminuric CKD stage 3 patients after 1 year of follow-up as compared with ARB monotherapy, for whom proteinuria increased 13.8%. Moreover, pentoxifylline decreased urinary levels of TNFα and monocyte chemoattractant protein 1 (MCP-1) (TNFα: 42.8% versus 18.8% and MCP-1: −28.9% versus 6.2%, for pentoxifylline and control groups, respectively). A decrease in both parameters was directly related to the change in proteinuria in the pentoxifylline group. Chen et al. [105] reported that 800 mg/day pentoxifylline for 6 months decreased proteinuria in 17 patients with primary glomerular diseases [36.5% and 33.9% decrease in spot and 24 h proteinuria (g/g Cr)]. This decrease was associated with a decline in urinary mean percentage decrease of 46% in MCP-1 urinary excretion levels. In a larger study, Chen et al. published a retrospective analysis of a study comprising 661 patients with CKD stages 3–5 treated with pentoxifylline [106]. Again, pentoxifylline on top of RAS blockade had kidney protective effects in the subset of patients with higher levels of proteinuria. A trial conducted by Shu et al. [107] reported a 19.6% decrease in proteinuria in third month and improved graft survival by the end of the study in non-diabetic renal transplant recipients with chronic allograft nephropathy and microalbuminuria treated with pentoxifylline for at least 6 months.

Two recent meta-analyses reported the effects of pentoxifylline alone or in combination with other treatments in the decrease in proteinuria and in the preservation of kidney function in patients with diabetic or non-diabetic CKD. In the first meta-analysis, Leporini et al. [119] concluded that pentoxifylline was effective in decreasing proteinuria compared with control, a benefit that was more evident in patients with type 1 DM, higher proteinuria at baseline and early renal impairment. They also found an improvement in renal function (eGFR/creatinine clearance) in the long-term and in patients with more advanced CKD. In the second meta-analysis, Liu et al. [120] concluded that pentoxifylline in combination with RAS blockade decreases proteinuria and slows down the decline of renal function in patients with CKD stages 3–5.

Finally, an analysis of a nationwide administrative dataset of advanced CKD patients identifying two propensity score-matched cohorts (pentoxifylline users and nonusers) reported that the pentoxifylline group was protected from ESKD [121] This was the first evidence of the ability of pentoxifylline in decreasing the risk of ESKD even in patients with advanced CKD.

Mechanisms of kidney protection by pentoxifylline

Pentoxifylline is a methyl-xanthine derivative with several effects including the non-selective inhibition of phosphodiesterases (PDEs). The balance of intracellular cyclic adenosine-3,5-monophosphate (cAMP), an important intracellular second signalling messenger, is mainly dependent on the activity of two enzymes: adenylyl cyclase, which plays a major role in cAMP synthesis and PDEs, which hydrolyze cAMP [122, 123]. Therefore, the inhibition of PDEs by pentoxifylline prevents the degradation of cAMP (Fig. 1). High cAMP levels in turn promote protein kinase A (PKA) activation leading to the phosphorylation of diverse effectors followed by inhibition of signalling pathways involved in proteinuria and renal fibrosis [124–126].

FIGURE 1:

FIGURE 1:

Mechanism proposed to explain the anti-inflammatory effects of pentoxifylline. The inhibition of PDE exerted by pentoxifylline increases cAMP levels activating PKA. Active PKA would inhibit ubiquitination that drives IkB to proteasome degradation preventing thus p50/p65 translocation and the expression of inflammatory cytokines and other genes. Dashed lines, inhibition; PTF, pentoxifylline; PDE, phosphodiesterase; ATP, adenosine triphosphate; AC, adenylate cyclase; cAMP, cyclic adenosine-3,5-monophosphate; IkB, inhibitor of kappa B; TNF, tumour necrosis factor α; IL, interleukin; IFN, interferon; ICAM1, intercellular adhesion molecule 1; VCAM1, vascular cell adhesion molecule 1; CRP, C-reactive protein.

PDEs have emerged as promising targets for pharmacological intervention against CKD progression [127–129]. Mammalian cells have 11 gene families (PDE1–PDE11) and each family encompasses 1–4 distinct genes, giving ˃20 genes in mammals encoding ˃60 different PDE isoforms. In vitro, pentoxifylline inhibits PDE3 and/or PDE4 isozymes through a PKA-dependent pathway [124, 126, 130]. Importantly, PDE3 and PDE4 isozymes are mainly expressed in monocytes and neutrophils [131–133], which makes them a therapeutic target in many inflammatory diseases, including asthma, chronic obstructive pulmonary disease, inflammatory bowel disease, psoriasis, nervous system inflammation and rheumatoid arthritis [133]. Pentoxifylline presents anti-inflammatory properties mediated by the inhibition of PDEs that supports its potential application in the kidney protection of the patient with DM [113–118]. In experimental models, pentoxifylline modulates signalling pathways or components triggered by inflammatory cytokines. In vitro, pentoxifylline inhibits endotoxin-induced TNFα synthesis in RAW 264.7 macrophages [108]. Pentoxifylline also inhibited endotoxin-induced TNFα production both in the serum of mice and in cultured adherent peritoneal exudate cells [93]. In a rat model of crescentic glomerulonephritis, pentoxifylline exerts anti-inflammatory and immunomodulatory actions through the inhibition of renal TNFα, ICAM-1, RANTES, MCP-1 and OPN, thereby suppressing progressive renal injury [109]. Similarly, pentoxifylline decreases the renal expression of pro-inflammatory cytokines including TNFα and IL6 in streptozotocin- or alloxan-induced diabetic rat models [110, 111], ameliorating renal hypertrophy and sodium retention [110]. Clinical trials evaluating the anti-inflammatory properties of this drug in non-diabetic patients report considerable modulating effects on the production of inflammatory cytokines and adhesion molecules in patients with coronary artery disease and atherosclerosis [134, 135]. Similarly, pentoxifylline decreased TNFα and interferon-gamma T-cell expression in ESKD patients [136].

As discussed above, most RCTs evaluating the renal effects of pentoxifylline in patients with DM have shown kidney protection, evidenced by the decrease in proteinuria and, in some cases, the improvement or preservation of GFR (Fig. 2). Importantly, some RCTs also observed a significant decrease in inflammatory parameters. The antiproteinuric effect of pentoxifylline has been associated with a significant decrease in TNFα levels [95, 99]. Similarly, clinical trials conducted in CKD patients with stage 3 or higher reported stabilization of renal function and decreased circulating levels of TNFα, fibrinogen and CRP after treatment with pentoxifylline [118] and a decrease in proteinuria and urinary levels of TNFα and MCP1 after 1 year on add‐on pentoxifylline to ARB background therapy [104]. The PREDIAN trial [84] evaluated the kidney-protective effects of pentoxifylline in DKD patients under RAS blockade. After 2 years, patients on pentoxifylline presented a decrease in the progression of renal disease that was accompanied by a decrease in proteinuria and urinary levels of TNFα. Two meta‐analyses also pointed to the decrease of proinflammatory cytokines production as the most likely explanation for this antiproteinuric effect in DKD patients [137] and concluded that pentoxifylline additively decreases proteinuria and TNFα in DKD patients receiving RAS inhibitors [138].

FIGURE 2:

FIGURE 2:

Proposed beneficial effects of pentoxifylline on kidney function in DKD patients.

An unexpected beneficial effect of pentoxifylline in DKD patients could be the stimulation of factors that promote kidney health [139]. The protein Klotho is an important regulator of mineral metabolism mainly expressed in kidney tubular epithelial cells and, to a lesser extent, in parathyroid glands, choroid plexus of the brain, vascular tissue and peripheral blood cells [140, 141]. Two forms of Klotho can be found: a single-pass transmembrane protein and a soluble form generated from proteolytic cleavage of the extracellular domain of the membrane-bound form [142]. Soluble Klotho is found in the cerebrospinal fluid, urine and blood, and declines in CKD patients with the progression of the disease. Klotho has anti-ageing and kidney-protective effects. In fact, specific epigenetic prevention of Klotho downregulation prevented acute kidney injury in mice [143]. Interestingly, patients with type 2 DM also have lower soluble Klotho levels [144, 145] and kidney Klotho is decreased in biopsies from patients with early stages of DKD [146]. Taken together, these data point to the potential utility of Klotho as an early biomarker of renal impairment in type 2 DM patients and to Klotho downregulation as a driver of DKD progression [147]. Moreover, Klotho has been inversely related to inflammation. Pro-inflammatory cytokines like TNFα and TWEAK (tumor necrosis factor-like weak inducer of apoptosis) inhibit renal Klotho expression in an NF‐κB‐mediated manner in vivo and in vitro [148–151]. Clinical translation of this observation is supported by a post‐hoc analysis of the PREDIAN trial [84]. Administration of pentoxifylline to type 2 DM patients with CKD stages 3 and 4 decreased serum and urinary TNFα and increased serum and urinary Klotho levels [85]. Even though the precise mechanisms are unknown, a feasible hypothesis is that the stimulation of Klotho production by pentoxifylline may result from its anti-inflammatory properties, although in cultured tubular cells, pentoxifylline directly prevented the albuminuria-induced downregulation of Klotho expression [85, 152]. Moreover, pentoxifylline increased tubular cell Klotho above baseline levels, suggesting that promoting kidney expression may be one of the mechanisms of kidney protection by pentoxifylline. Moreover, a recently published experimental study, confirmed the positive effect of therapeutic doses of pentoxifylline (10 μg/mL) on Klotho expression in RAW 264.7 cells, showing that this up-regulation is not limited to kidney cells [153].

CONCLUSIONS AND FUTURE PERSPECTIVES

The global burden of diabetes is predicted to increase dramatically in the coming decades in parallel with the rising of obesity. One of the most important complications of diabetes is DKD, which substantially increases cardiovascular morbidity and mortality, determining a considerable impairment in the quality of life. Indeed, CKD is set to become the fifth global cause of death by 2040 [154]. In a recent large, retrospective cohort study including 65000 adults with type 2 DM and CKD, a high proportion (10%–17%) of patients presented disease progression over a median follow-up of only 2 years [153]. Previous studies also indicated that DM is a more frequent cause of accelerated progression from CKD to ESKD compared with other predictors including proteinuria, heart failure, anaemia and elevated systolic blood pressure [155].

Even with the widespread use of SGLT2i and GLP-1 receptor agonists, a substantial residual risk of DKD progression remains. Therefore, there is a need to find new therapeutic targets and strategies. Pentoxifylline constitutes a potential repurposed drug for the treatment of DKD. Although the repurposing of pentoxifylline constitutes a promising opportunity in providing a low-cost access to a feasible therapy in DKD, various challenges remain. A recent study reported an increased risk of major bleeding events in CKD patients on pentoxifylline treatment [156]. This population carries a higher risk of bleeding due to the presence of platelet dysfunction and anaemia, especially in patients with albuminuria [157, 158].

As pointed out by Leporini et al. [119] in a systematic review and meta-analysis aimed at evaluating the benefits of pentoxifylline on renal outcomes in CKD patients, whether this drug may be useful for retarding disease progression remains partly unanswered for definite reasons. One of them is the heterogeneity of the studies with respect to sample sizes, doses of pentoxifylline, poblation in the study (CKD stage and aetiology, proteinuria levels) and control groups and concomitant RAS blocking treatments. Most RCTs were of low methodological quality and carried out with small sample sizes and only provided short-term data for surrogate kidney function endpoints such as eGFR or serum creatinine, proteinuria and albuminuria.

Future studies with longer follow-up, larger sample size and including hard clinical outcomes are needed. To our knowledge, at present, there are two phase IV clinical trials evaluating the effects of pentoxifylline in the progression of DKD patients. The Veterans Affairs (VA) PTXRx (NCT03625648) is a double-blind, placebo-controlled, multicentre RCT designed to evaluate the utility of pentoxifylline, when added to usual care, in the decrease in the incidence of ESRD or death in patients with type 2 diabetes with DKD [159]. The enrolment of patients for this study (which is estimated in 2510 participants) began in 2019 and the primary completion date will be at the beginning of 2028. The second clinical trial in course, PENFOSIDINE (Pentoxifylline Effect in Patients With Diabetic Nephropathy, NCT03664414), began in 2018. PENFOSIDINE included 196 patients with DKD and the objectives included evaluating the antioxidant, anti-inflammatory and antifibrotic effects of receiving pentoxifylline (400 mg/three times a day) for a period of 2 years. The results of these trials will shed light on the long-term effects of pentoxifylline on various markers of inflammation, oxidative stress and fibrosis, on surrogate markers of renal function such as a decrease in proteinuria and changes in eGFR and on hard endpoints such as ESRD and death.

In any case, an important problem that faces the repurposing of many drugs, including pentoxifylline, is the absence of exclusivity for the industry. Since pentoxifylline was approved for intermittent claudication, no studies were conducted about its utility for other diseases prior to the approval or before the expiration of the patent. This implies that funding for future research is highly compromised even though current research may demonstrate the efficacy of pentoxifylline in other diseases. Pharmaceutical companies find it less lucrative redirecting resources towards repurposing programmes given the lack of standardized regulations ensuring market protection. Moreover, even after marketing approval, the off-label and unlicensed prescription of a generic version of this old drug leaves little or no space for profit for the industry. Therefore, pivotal RCTs in search of a DKD indication for pentoxifylline should be funded by government-supported trial programmes.

In addition, a successful new indication of the repurposed drug requires extensive knowledge of the pathogenesis of the target disease, as well as the postulated mechanism of action of the drug. Present data point to the ability of pentoxifylline to slow CKD progression when macroalbuminuria is present, even in advanced stages of DKD with maximized RAS blocker therapy. However, although there is evidence for an anti-inflammatory effect, the precise mechanism for the beneficial effect is not known and may also involve increasing Klotho production. In fact, the pathogenic pathways for DKD are poorly understood, as exemplified by the poor therapeutic toolkit and low-grade inflammation is just one of the interrelated contributing mechanisms along with hyperglycaemia, altered lipid metabolism, RAS hyperactivation and increased sympathetic activity. In any case, an in-depth study of the hidden therapeutic potential of pentoxifylline and its repurposing for the DKD new indication offers a tremendous hope to decrease residual kidney risk and prevent the growth of the DKD pandemic.

Contributor Information

Javier Donate-Correa, Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain; GEENDIAB (Grupo Español para el estudio de la Nefropatía Diabética), Sociedad Española de Nefrología, Santander, Spain; RICORS2040 (RD21/0005/0013), Instituto de Salud Carlos III, Madrid, Spain.

María Dolores Sanchez-Niño, Departamento de Nefrología e Hipertensión, IIS-Fundación Jiménez Díaz y Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.

Ainhoa González-Luis, Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain; Escuela de doctorado, Universidad de La Laguna, La Laguna, Spain.

Carla Ferri, Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain; Escuela de doctorado, Universidad de La Laguna, La Laguna, Spain.

Alberto Martín-Olivera, Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain; Escuela de doctorado, Universidad de La Laguna, La Laguna, Spain.

Ernesto Martín-Núñez, Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain; RICORS2040 (RD21/0005/0013), Instituto de Salud Carlos III, Madrid, Spain.

Beatriz Fernandez-Fernandez, Departamento de Nefrología e Hipertensión, IIS-Fundación Jiménez Díaz y Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain; RICORS2040 (RD21/0005/0001), Instituto de Salud Carlos III, Madrid, Spain.

Víctor G Tagua, Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain.

Carmen Mora-Fernández, Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain; GEENDIAB (Grupo Español para el estudio de la Nefropatía Diabética), Sociedad Española de Nefrología, Santander, Spain; RICORS2040 (RD21/0005/0013), Instituto de Salud Carlos III, Madrid, Spain.

Alberto Ortiz, Departamento de Nefrología e Hipertensión, IIS-Fundación Jiménez Díaz y Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain; RICORS2040 (RD21/0005/0001), Instituto de Salud Carlos III, Madrid, Spain.

Juan F Navarro-González, Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain; GEENDIAB (Grupo Español para el estudio de la Nefropatía Diabética), Sociedad Española de Nefrología, Santander, Spain; RICORS2040 (RD21/0005/0013), Instituto de Salud Carlos III, Madrid, Spain; Servicio de Nefrología, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain; Instituto de Tecnologías Biomédicas, Universidad de La Laguna, Santa Cruz de Tenerife, Spain.

Funding

This work was supported by Instituto de Salud Carlos III (ISCIII): PI07/0870, PI15/00298, PI16/02057, PI16/00024, PI19/00035, PI20/00744, PI21/00251, PI21/01037, PI19/00815, DTS18/00032 ISCIII-RETIC REDinREN RD16/0009, and RICORS2040. We acknowledge cofunding by Fondo Europeo de Desarrollo Regional [FEDER], Unión Europea [‘Una forma de hacer Europa’]. J.D.-C. is the recipient of a contract from Miguel Servet Program (CP20/00122) of the ISCIII. A.G.-L. and C.F. are recipients of contracts from Agencia Canaria de Investigación Innovación y Sociedad de la Información del Gobierno de Canarias (ACIISI) (TESIS2021010045 and TESIS2018010110). E.M.-N. is funded by a research contract from REDinREN-ISCIII (RD16/0009/0022). V.G.-T. is supported by Cabildo de Tenerife, TF Innova, Fondo de Desarrollo de Canarias (FDCAN) and Marco Estratégico de Desarrollo Insular (MEDI), in the Agustín de Betancourt programme.

CONFLICT OF INTEREST STATEMENT

A.O. has received grants from Sanofi and consultancy or speaker fees or travel support from Advicciene, Astellas, AstraZeneca, Amicus, Amgen, Fresenius Medical Care, GSK, Bayer, Sanofi-Genzyme, Menarini, Kyowa Kirin, Alexion, Idorsia, Chiesi, Otsuka, Novo-Nordisk and Vifor Fresenius Medical Care Renal Pharma, and is Director of the Catedra Mundipharma-UAM of diabetic kidney disease and the Catedra AstraZeneca-UAM of chronic kidney disease and electrolytes. A.O. is the past Editor-in-Chief of CKJ. B.F.-F. reports speaker fees or travel support from AbbVie, AstraZeneca, Boehringer Ingelheim, Esteve, Menarini, Mundipharma, Novartis and Novo Nordisk. J.F.N.-G. has served as a consultant and has received speaker fees or travel support from AbbVie, Amgen, AstraZeneca, Boehringer Ingelheim, Esteve, Eli Lilly, MSD, Mundipharma, Novartis, NovoNordisk, Sanofi-Genzyme, Servier, Shire and Vifor Pharma. The other authors declare no conflict of interest.

REFERENCES

  • 1. International Diabetes Federation . IDF Diabetes Atlas. 8th ed.International Diabetes Federation. 2017 [Google Scholar]
  • 2. Atkins RC, Zimmet P. Diabetic kidney disease: act now or pay later. Kidney Int 2010; 77: 375–377 [DOI] [PubMed] [Google Scholar]
  • 3. Hovind P, Rossing P, Tarnow Let al. Progression of diabetic nephropathy. Kidney Int 2001; 59: 702–709 [DOI] [PubMed] [Google Scholar]
  • 4. Thomas MC, Cooper ME, Zimmet P. Changing epidemiology of type 2 diabetes mellitus and associated chronic kidney disease. Nat Rev Nephrol 2016, 12: 73–81 [DOI] [PubMed] [Google Scholar]
  • 5. Yamazaki T, Mimura I, Tanaka Tet al. Treatment of diabetic kidney disease: current and future. Diabetes Metab J 2021; 45: 11–26 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Yokoyama H, Sone H, Oishi Met al. Prevalence of albuminuria and renal insufficiency and associated clinical factors in type 2 diabetes: the Japan Diabetes Clinical Data Management Study (JDDM15). Nephrol Dial Transplant 2009; 24: 1212–1219 [DOI] [PubMed] [Google Scholar]
  • 7. Afkarian M, Zelnick LR, Hall YNet al. Clinical manifestations of kidney disease among us adults with diabetes, 1988-2014. JAMA 2016; 316: 602–610 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Gnudi L, Gentile G, Ruggenenti P.. Oxford Textbook of Clinical Nephrology. 4th ed.Oxford University Press. Oxford: 2016; 1199–1247 [Google Scholar]
  • 9. Ritz E, Rychlík I, Locatelli Fet al. End-stage renal failure in type 2 diabetes: a medical catastrophe of worldwide dimensions. Am J Kidney Dis 1999; 34: 795–808 [DOI] [PubMed] [Google Scholar]
  • 10. Atkins RC. The epidemiology of chronic kidney disease. Kidney Int 2005; 67: S14–S18 [DOI] [PubMed] [Google Scholar]
  • 11. Gregg EW, Li Y, Wang Jet al. Changes in diabetes-related complications in the United States, 1990-2010. N Engl J Med 2014; 370: 1514–1523 [DOI] [PubMed] [Google Scholar]
  • 12. Jha V, Garcia-Garcia G, Iseki Ket al. Chronic kidney disease: global dimension and perspectives. Lancet 2013; 382: 260–272 [DOI] [PubMed] [Google Scholar]
  • 13. Cooper ME. Diabetes: treating diabetic nephropathy-still an unresolved issue. Nat Rev Endocrinol 2012; 8: 515–516 [DOI] [PubMed] [Google Scholar]
  • 14. Cooper ME. Interaction of metabolic and haemodynamic factors in mediating experimental diabetic nephropathy. Diabetologia 2001; 44: 1957–1972 [DOI] [PubMed] [Google Scholar]
  • 15. Gnudi L, Thomas SM, Viberti G. Mechanical forces in diabetic kidney disease: a trigger for impaired glucose metabolism. J Am Soc Nephrol 2007; 18: 2226–2232 [DOI] [PubMed] [Google Scholar]
  • 16. Brownlee M. The pathobiology of diabetic complications: a unifying mechanism. Diabetes 2005; 54: 1615–1625 [DOI] [PubMed] [Google Scholar]
  • 17. National Kidney Foundation . KDOQI Clinical Practice Guideline for Diabetes and CKD: 2012 Update. Am J Kidney Dis 2012; 60: 850–886 [DOI] [PubMed] [Google Scholar]
  • 18. Sanz AB, Ramos AM, Soler MJet al. Advances in understanding the role of angiotensin-regulated proteins in kidney diseases. Expert Rev Proteomics 2019; 16: 77–92 [DOI] [PubMed] [Google Scholar]
  • 19. Thomson HJ, Ekinci EI, Radcliffe NJet al. Elevated baseline glomerular filtration rate (GFR) is independently associated with a more rapid decline in renal function of patients with type 1 diabetes. J Diabetes Complications 2016; 30: 256–261 [DOI] [PubMed] [Google Scholar]
  • 20. Mezzano SA, Ruiz-Ortega M, Egido J. Angiotensin II and renal fibrosis. Hypertension 2001; 38: 635–638 [DOI] [PubMed] [Google Scholar]
  • 21. Makani H, Bangalore S, Desouza KAet al. Efficacy and safety of dual blockade of the renin-angiotensin system: meta-analysis of randomised trials. BMJ 2013; 346: F360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Mann JF, Schmieder RE, McQueen Met al. Renal outcomes with telmisartan, ramipril, or both, in people at high vascular risk (The ONTARGET study): a multicentre, randomised, double-blind, controlled trial. Lancet 2008; 372: 547–553 [DOI] [PubMed] [Google Scholar]
  • 23. Esteras R, Perez-Gomez MV, Rodriguez-Osorio Let al. Combination use of medicines from two classes of renin-angiotensin system blocking agents: risk of hyperkalemia, hypotension, and impaired renal function. Ther Adv Drug Saf 2015; 6: 166–176 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Parving HH, Brenner BM, McMurray JJet al. Cardiorenal end points in a trial of aliskiren for type 2 diabetes. N Engl J Med 2012; 367: 2204–2213 [DOI] [PubMed] [Google Scholar]
  • 25. Fried LF, Emanuele N, Zhang JHet al. Combined angiotensin inhibition for the treatment of diabetic nephropathy. N Engl J Med 2013; 369: 1892–1903 [DOI] [PubMed] [Google Scholar]
  • 26. Perkovic V, Jardine MJ, Neal Bet al. Canagliflozin and renal outcomes in type 2 diabetes and nephropathy. N Engl J Med 2019; 380: 2295–2306 [DOI] [PubMed] [Google Scholar]
  • 27. Packham DK, Wolfe R, Reutens ATet al. Sulodexide fails to demonstrate renoprotection in overt type 2 diabetic nephropathy. J Am Soc Nephrol 2012; 23: 123–130 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Lewis EJ, Greene T, Spitalewiz Set al. Pyridorin in type 2 diabetic nephropathy. J Am Soc Nephrol 2012; 23: 131–136 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Pfeffer MA, Burdmann EA, Chen CYet al. A trial of darbepoetin alfa in type 2 diabetes and chronic kidney disease. N Engl J Med 2009; 361: 2019–2032 [DOI] [PubMed] [Google Scholar]
  • 30. de Zeeuw D, Akizawa T, Audhya Pet al. Bardoxolone methyl in type 2 diabetes and stage 4 chronic kidney disease. N Engl J Med 2013; 369: 2492–2503 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Mann JF, Green D, Jamerson Ket al. Avosentan for overt diabetic nephropathy. J Am Soc Nephrol 2010; 21: 527–535 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Singh DK, Winocour P, Farrington K. Oxidative stress in early diabetic nephropathy: fueling the fire. Nat Rev Endocrinol 2011; 7: 176–184 [DOI] [PubMed] [Google Scholar]
  • 33. Navarro-Gonzalez JF, Mora-Fernández C, Muros-de-Fuentes Met al. Inflammatory molecules and pathways in the pathogenesis of diabetic nephropathy. Nat Rev Nephrol 2011; 7: 327–340 [DOI] [PubMed] [Google Scholar]
  • 34. Zhang Y, Jin D, Kang Xet al. Signaling pathways involved in diabetic renal fibrosis. Front Cell Dev Biol 2021; 9: 696542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Panchapakesan U, Pollock C. Drug repurposing in kidney disease. Kidney Int 2018; 94: 40–48 [DOI] [PubMed] [Google Scholar]
  • 36. Thomas MC, Cherney DZI. The actions of SGLT2 inhibitors on metabolism, renal function and blood pressure. Diabetologia 2018; 61: 2098–2107 [DOI] [PubMed] [Google Scholar]
  • 37. Wanner C, Inzucchi SE, Lachin JMet al. Empagliflozin and progression of kidney disease in type 2 diabetes. N Engl J Med 2016; 375: 323–334 [DOI] [PubMed] [Google Scholar]
  • 38. Perkovic V, de Zeeuw D, Mahaffey KWet al. Canagliflozin and renal outcomes in type 2 diabetes: results from the CANVAS Program randomized clinical trials. Lancet Diabetes Endocrinol 2018; 6: 691–704 [DOI] [PubMed] [Google Scholar]
  • 39. Sarafidis P, Ferro CJ, Morales Eet al. SGLT-2 inhibitors and GLP-1 receptor agonists for nephroprotection and cardioprotection in patients with diabetes mellitus and chronic kidney disease. A consensus statement by the EURECA-m and the DIABESITY working groups of the ERA-EDTA. Nephrol Dial Transplant 2019; 34: 208–230 [DOI] [PubMed] [Google Scholar]
  • 40. Davies MJ, D'Alessio DA, Fradkin Jet al. Management of hyperglycemia in type 2 diabetes, 2018. A consensus report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetes Care 2018; 41: 2669–2701 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Perez-Gomez MV, Sanchez-Niño MD, Sanz ABet al. Targeting inflammation in diabetic kidney disease: early clinical trials. Expert Opin Investig Drugs 2016; 25: 1045–1058 [DOI] [PubMed] [Google Scholar]
  • 42. Hou J, Xiong W, Cao Let al. Spironolactone add-on for preventing or slowing the progression of diabetic nephropathy: a meta-analysis. Clin Ther 2015; 37: 2086–2103 [DOI] [PubMed] [Google Scholar]
  • 43. Williams GH, Burgess E, Kolloch REet al. Efficacy of eplerenone versus enalapril as monotherapy in systemic hypertension. Am J Cardiol 2004; 93: 990–996 [DOI] [PubMed] [Google Scholar]
  • 44. Bakris GL, Agarwal R, Chan JCet al. Effect of finerenone on albuminuria in patients with diabetic nephropathy: a randomized clinical trial. JAMA 2015; 314: 884–894 [DOI] [PubMed] [Google Scholar]
  • 45. Wan N, Rahman A, Nishiyama A. Esaxerenone, a novel nonsteroidal mineralocorticoid receptor blocker (MRB) in hypertension and chronic kidney disease. J Hum Hypertens 2021; 35: 148–156 [DOI] [PubMed] [Google Scholar]
  • 46. Pitt B, Filippatos G, Agarwal Ret al. Cardiovascular events with finerenone in kidney disease and type 2 diabetes. N Engl J Med 2021; 385: 2252–2263 [DOI] [PubMed] [Google Scholar]
  • 47. Bakris GL, Agarwal R, Anker SDet al. Effect of finerenone on chronic kidney disease outcomes in type 2 diabetes. N Engl J Med 2020; 383: 2219–2229 [DOI] [PubMed] [Google Scholar]
  • 48. Hattori S. Sitagliptin reduces albuminuria in patients with type 2 diabetes. Endocr J 2011; 58: 69–73 [DOI] [PubMed] [Google Scholar]
  • 49. Sakata K, Hayakawa M, Yano Yet al. Efficacy of alogliptin, a dipeptidyl peptidase-4 inhibitor, on glucose parameters, the activity of the advanced glycation end product (AGE)-Receptor for AGE (RAGE) axis and albuminuria in Japanese type 2 diabetes. Diabetes Metab Res Rev 2013; 29: 624–663 [DOI] [PubMed] [Google Scholar]
  • 50. Marso SP, Daniels GH, Brown-Frandsen Ket al. Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med 2016; 375: 311–322 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Mann JFE, Orsted DD, Brown-Frandsen Ket al. Liraglutide and renal outcomes in type 2 diabetes. N Engl J Med 2017; 377: 839–848 [DOI] [PubMed] [Google Scholar]
  • 52. Marso SP, Bain SC, Consoli Aet al. Semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med 2016; 375: 1834–1844 [DOI] [PubMed] [Google Scholar]
  • 53. Palmer SC, Tendal B, Mustafa RAet al. Sodium-glucose cotransporter protein-2 (SGLT-2) inhibitors and Glucagon-like peptide-1 (GLP-1) receptor agonists for type 2 diabetes: systematic review and network meta-analysis of randomised controlled trials. BMJ 2021; 372: m4573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Sattar N, Lee MMY, Kristensen SLet 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 [DOI] [PubMed] [Google Scholar]
  • 55. Pfeffer MA, Claggett B, Diaz Ret al. Lixisenatide in patients with type 2 diabetes and acute coronary syndrome. N Engl J Med 2015; 373: 2247–2257 [DOI] [PubMed] [Google Scholar]
  • 56. Tuttle KR, Lakshmanan MC, Rayner Bet al. Dulaglutide versus insulin glargine in patients with type 2 diabetes and moderate-to-severe chronic kidney disease (AWARD-7): a multicentre, open-label, randomised trial. Lancet Diabetes Endocrinol 2018; 6: 605–617 [DOI] [PubMed] [Google Scholar]
  • 57. Rosenstock J, Perkovic V, Johansen OEet al. Effect of linagliptin vs placebo on major cardiovascular events in adults with type 2 diabetes and high cardiovascular and renal risk: the CARMELINA randomized clinical trial. JAMA 2019; 321: 69–79 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Groop PH, Cooper ME, Perkovic Vet al. Linagliptin and its effects on hyperglycaemia and albuminuria in patients with type 2 diabetes and renal dysfunction: the randomized MARLINA-T2D trial. Diabetes Obesity Metab 2017; 19: 1610–1619 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Mosenzon O, Leibowitz G, Bhatt DLet al. Effect of saxagliptin on renal outcomes in the SAVOR-TIMI 53 trial. Diabetes Care 2017; 40: 69–76 [DOI] [PubMed] [Google Scholar]
  • 60. Williams ME, Bolton WK, Khalifah RGet al. Effects of pyridoxamine in combined phase 2 studies of patients with type 1 and type 2 diabetes and overt nephropathy. Am J Nephrol 2007; 27: 605–614 [DOI] [PubMed] [Google Scholar]
  • 61. Rabbani N, Alam SS, Riaz Set al. High-dose thiamine therapy for patients with type 2 diabetes and microalbuminuria: a randomised, double-blind placebo-controlled pilot study. Diabetologia 2009; 52: 208–212 [DOI] [PubMed] [Google Scholar]
  • 62. Bolton WK, Cattran DC, Williams MEet al. Randomized trial of an inhibitor of formation of advanced glycation end products in diabetic nephropathy. Am J Nephrol 2004; 24: 32–40 [DOI] [PubMed] [Google Scholar]
  • 63. Alkhalaf A, Klooster A, van Oeveren Wet al. A double-blind, randomized, placebo-controlled clinical trial on benfotiamine treatment in patients with diabetic nephropathy. Diabetes Care 2010; 33: 1598–1601 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Sleigh S, Barton CL. Repurposing strategies for therapeutics. Pharm Med 2010; 24: 151–159 [Google Scholar]
  • 65. Breckenridge A, Jacob R. Overcoming the legal and regulatory barriers to drug repurposing. Nat Rev Drug Discov 2019; 18: 1–2 [DOI] [PubMed] [Google Scholar]
  • 66. Parvathaneni V, Gupta V. Utilizing drug repurposing against COVID-19–efficacy, limitations, and challenges. Life Sci 2020; 259: 118275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Moehler MT, Hillengass J, Glasmacher Aet al. Thalidomide in multiple myeloma. Curr Pharm Biotechnol 2006; 7: 431–440 [DOI] [PubMed] [Google Scholar]
  • 68. Dinkova-Kostova AT, Liby KT, Stephenson KKet al. Extremely potent triterpenoid inducers of the phase 2 response: correlations of protection against oxidant and inflammatory stress. Proc Natl Acad Sci USA 2005; 102: 4584–4589 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Pergola PE, Krauth M, Huff JWet al. Effect of bardoxolone methyl on kidney function in patients with T2D and stage 3b-4 CKD. Am J Nephrol 2011; 33: 469–476 [DOI] [PubMed] [Google Scholar]
  • 70. Pergola PE, Raskin P, Toto RDet al. Bardoxolone methyl and kidney function in CKD with type 2 diabetes. N Engl J Med 2011; 365: 327–336 [DOI] [PubMed] [Google Scholar]
  • 71. Chin MP, Reisman SA, Bakris GLet al. Mechanisms contributing to adverse cardiovascular events in patients with type 2 diabetes mellitus and stage 4 chronic kidney disease treated with bardoxolone methyl. Am J Nephrol 2014; 39: 499–508 [DOI] [PubMed] [Google Scholar]
  • 72. Chin MP, Wrolstad D, Bakris GLet al. Risk factors for heart failure in patients with type 2 diabetes mellitus and stage 4 chronic kidney disease treated with bardoxolone methyl. J Card Fail 2014; 20: 953–958 [DOI] [PubMed] [Google Scholar]
  • 73. Nangaku M, Kanda H, Takama Het al. Randomized clinical trial on the effect of bardoxolone methyl on GFR in diabetic kidney disease patients (TSUBAKI study). Kidney Int Rep 2020; 5: 879–890 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. de Zeeuw D, Bekker P, Henkel Eet al. The effect of CCR2 inhibitor CCX140-B on residual albuminuria in patients with type 2 diabetes and nephropathy: a randomized trial. Lancet Diabetes Endocrinol 2015; 3: 687–696 [DOI] [PubMed] [Google Scholar]
  • 75. Tuttle KR, Brosius FC, Adler SGet al. JAK1/JAK2 inhibition by baricitinib in diabetic kidney disease: results from a phase 2 randomized controlled clinical trial. Nephrol Dial Transplant 2018; 33: 1950–1959 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Kohan DE, Barton M. Endothelin and endothelin antagonists in chronic kidney disease. Kidney Int 2014; 86: 896–904 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Carducci MA, Padley RJ, Breul Jet al. Effect of endothelin-A receptor blockade with atrasentan on tumor progression in men with hormone-refractory prostate cancer: a randomized, phase II, placebo-controlled trial. J Clin Oncol 2003; 21: 679–689 [DOI] [PubMed] [Google Scholar]
  • 78. Weber MA, Black H, Bakris Get al. A selective endothelin-receptor antagonist to reduce blood pressure in patients with treatment-resistant hypertension: a randomised, double-blind, placebo-controlled trial. Lancet 2009; 374: 1423–1431 [DOI] [PubMed] [Google Scholar]
  • 79. Kassab S, Miller MT, Novak Jet al. Endothelin-A receptor antagonism attenuates the hypertension and renal injury in dahl salt-sensitive rats. Hypertension 1998; 31: 397–402 [DOI] [PubMed] [Google Scholar]
  • 80. de Zeeuw D, Coll B, Andress Det al. The endothelin antagonist atrasentan lowers residual albuminuria in patients with type 2 diabetic nephropathy. J Am Soc Nephrol 2014; 25: 1083–1093 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Kohan DE, Pritchett Y, Molitch Met al. Addition of atrasentan to renin-angiotensin system blockade reduces albuminuria in diabetic nephropathy. J Am Soc Nephrol 2011; 22: 763–772 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Heerspink HJL, Parving HH, Andress DLet al. Atrasentan and renal events in patients with type 2 diabetes and chronic kidney disease (SONAR): a double-blind, randomized, placebo-controlled trial. Lancet 2019; 393: 1937–1947 [DOI] [PubMed] [Google Scholar]
  • 83. Andress DL, Coll B, Pritchett Yet al. Clinical efficacy of the selective endothelin a receptor antagonist, atrasentan, in patients with diabetes and chronic kidney disease (CKD). Life Sci 2012; 91: 739–742 [DOI] [PubMed] [Google Scholar]
  • 84. Navarro-González JF, Mora-Fernández C, Muros-de-Fuentes Met al. Effect of pentoxifylline on renal function and urinary albumin excretion in patients with diabetic kidney disease: the PREDIAN trial. J Am Soc Nephrol 2015; 26: 220–229 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Navarro-González JF, Sánchez-Niño MD, Donate-Correa Jet al. Effects of pentoxifylline on soluble klotho concentrations and renal tubular cell expression in diabetic kidney disease. Diabetes Care 2018; 41: 1817–1820 [DOI] [PubMed] [Google Scholar]
  • 86. Donate-Correa J, Tagua VG, Ferri Cet al. Pentoxifylline for renal protection in diabetic kidney disease. A model of old drugs for new horizons. J Clin Med 2019; 8: 287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. US Food & Drug Administration . Drugs-FDA: FDA Approved Drug Products. http://www.accessdata.fda.gov (15 October 2021, date last accessed) [Google Scholar]
  • 88. De Sanctis MT, Cesarone MR, Belcaro Get al. Treatment of intermittent claudication with pentoxifylline: a 12 month, randomized trial–walking distance and microcirculation. Angiology 2002; 53: 7–12 [PubMed] [Google Scholar]
  • 89. Aviado DM, Porter JM.. Pentoxifylline: a new drug for the treatment of intermittent claudication. Mechanism of action, pharmacokinetics, clinical efficacy and adverse effects. Pharmacotherapy 1984; 4: 297–307 [DOI] [PubMed] [Google Scholar]
  • 90. Aviado DM, Dettelbach HR. Pharmacology of pentoxifylline, a hemorheologic agent for the treatment of intermittent claudication. Angiology 1984; 35: 407–417 [DOI] [PubMed] [Google Scholar]
  • 91. Bell DSH. Are the protean effects of pentoxifylline in the therapy of diabetes and its complications still relevant? Diabetes Ther 2021; 12: 3025–3035 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Blagosklonnaia IAV, Mamedov R, Kozlov VVet al. Effect of trental on indices kidney function in diabetes mellitus. Probl Endokrinol 1982; 28: 3–8 [PubMed] [Google Scholar]
  • 93. Doherty GM, Jensen JC, Alexander HRet al. Pentoxifylline suppression of tumor necrosis factor gene transcription. Surgery 1991; 110: 192–198 [PubMed] [Google Scholar]
  • 94. Wen WX, Lee SY, Siang Ret al. Repurposing pentoxifylline for the treatment of fibrosis: an overview. Adv Ther 2017; 34: 1245–1269 [DOI] [PubMed] [Google Scholar]
  • 95. Navarro JF, Mora C, Rivero Aet al. Urinary protein excretion and serum tumor necrosis factor in diabetic patients with advanced renal failure: effects of pentoxifylline administration. Am J Kidney Dis 1999; 33: 458–463 [DOI] [PubMed] [Google Scholar]
  • 96. Aminorroaya A, Janghorbani M, Rezvanian Het al. Comparison of the effect of pentoxifylline and captopril on proteinuria in patients with type 2 diabetes mellitus. Nephron Clin Pract 2005; 99: c73–c77 [DOI] [PubMed] [Google Scholar]
  • 97. Rodríguez-Morán M, Guerrero-Romero F.. Pentoxifylline is as effective as captopril in the reduction of microalbuminuria in non-hypertensive type 2 diabetic patients-A randomized, equivalent trial. Clin Nephrol 2005; 64: 91–97 [DOI] [PubMed] [Google Scholar]
  • 98. Navarro JF, Mora C, Muros Met al. Additive antiproteinuric effect of pentoxifylline in patients with type 2 diabetes under angiotensin II receptor blockade: a short-term, randomized, controlled trial. J Am Soc Nephrol 2005; 16: 2119–2126 [DOI] [PubMed] [Google Scholar]
  • 99. Rodríguez-Morán M, González-González G, Bermúdez-Barba MVet al. Effects of pentoxifylline on the urinary protein excretion profile of type 2 diabetic patients with microproteinuria: a double-blind, placebo-controlled randomized trial. Clin Nephrol 2006; 66: 3–10 [DOI] [PubMed] [Google Scholar]
  • 100. Badri S, Dashti-Khavidaki S, Ahmadi Fet al. Effect of add-on pentoxifylline on proteinuria in membranous glomerulonephritis: a 6-month placebo-controlled trial. Clin Drug Investig 2013; 33: 215–222 [DOI] [PubMed] [Google Scholar]
  • 101. Diskin CJ, Stokes TJ, Dansby LMet al. Will the addition of pentoxifylline reduce proteinuria in patients with diabetic glomerulosclerosis refractory to maximal doses of both an angiotensin converting enzyme inhibitor and an angiotensin receptor blocker? J Nephrol 2007; 20: 410–416 [PubMed] [Google Scholar]
  • 102. Perkins RM, Aboudara MC, Uy ALet al. Effect of pentoxifylline on GFR decline in CKD: a pilot, double-blind, randomized, placebo-controlled trial. Am J Kidney Dis 2009; 53: 606–616 [DOI] [PubMed] [Google Scholar]
  • 103. Goicoechea M, García de Vinuesa S, Quiroga Bet al. Effects of pentoxifylline on inflammatory parameters in chronic kidney disease patients: a ran-domized trial. J Nephrol 2012; 25: 969–975 [DOI] [PubMed] [Google Scholar]
  • 104. Lin SL, Chen YM, Chiang WCet al. Effect of pentoxifylline in addition to losartan on proteinuria and GFR in CKD: a 12 month randomized trial. Am J Kidney Dis 2008; 52: 464–474 [DOI] [PubMed] [Google Scholar]
  • 105. Chen YM, Lin SL, Chiang WCet al. Pentoxifylline ameliorates proteinuria through suppression of renal monocyte chemoattractant protein-1 in patients with proteinuric primary glomerular diseases. Kidney Int 2006; 69: 1410–1415 [DOI] [PubMed] [Google Scholar]
  • 106. Chen P, Lai T, Chen Pet al. Renoprotective effect of combining pentoxifylline with angiotensin-converting enzyme inhibitor or angiotensin II receptor blocker in advanced chronic kidney disease. J Formos Med Assoc 2015; 113: 219–226 [DOI] [PubMed] [Google Scholar]
  • 107. Shu KH, Wu MJ, Chen CHet al. Effect of pentoxifylline on graft function of renal transplant recipients complicated with chronic allograft nephropathy. Clin Nephrol 2007; 67: 157–163 [DOI] [PubMed] [Google Scholar]
  • 108. Han J, Thompson P, Beutler B. Dexamethasone and pentoxifylline inhibit endotoxin-induced cachectin/tumor necrosis factor synthesis at separate points in the signaling pathway. J Exp Med 1990; 172: 391–394 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Chen YM, Ng YY, Lin SLet al. Pentoxifylline suppresses renal tumour necrosis factor-alpha and ameliorates experimental crescentic glomerulonephritis in rats. Nephrol Dial Transplant 2004; 19: 1106–1115 [DOI] [PubMed] [Google Scholar]
  • 110. DiPetrillo K, Gesek FA. Pentoxifylline ameliorates renal tumor necrosis factor expression, sodium retention, and renal hypertrophy in diabetic rats. Am J Nephrol 2004; 24: 352–359 [DOI] [PubMed] [Google Scholar]
  • 111. Garcia FA, Rebouças JF, Balbino TQet al. Pentoxifylline reduces the inflammatory process in diabetic rats: relationship with decreases of pro-inflammatory cytokines and inducible nitric oxide synthase. J Inflamm (Lond) 2015; 12: 33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Gentile G, Remuzzi G, Ruggenenti P. Dual renin-angiotensin system blockade for nephroprotection: still under scrutiny. Nephron 2015; 129: 39–41 [DOI] [PubMed] [Google Scholar]
  • 113. Voisin L, Breuillé D, Ruot Bet al. Cytokine modulation by PX differently affects specific acute phase proteins during sepsis in rats. Am J Physiol Content 1998; 275: R1412–R1419 [DOI] [PubMed] [Google Scholar]
  • 114. Strutz F, Heeg M, Kochsiek Tet al. Effects of pentoxifylline, pentifylline and gamma-interferon on proliferation, differentiation, and matrix synthesis of human renal fibroblasts. Nephrol Dial Transplant 2000; 15: 1535–1546 [DOI] [PubMed] [Google Scholar]
  • 115. Abdel-Salam O. The anti-inflammatory effects of the phosphodiesterase inhibitor pentoxifylline in the rat. Pharmacol Res 2003; 47: 331–340 [DOI] [PubMed] [Google Scholar]
  • 116. Dávila-Esqueda ME, Martínez-Morales F. Pentoxifylline diminishes the oxidative damage to renal tissue induced by streptozotocin in the rat. Exp Diabesity Res 2004; 5: 245–251 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Navarro-González JF, Mora-Fernández C. The role of inflammatory cytokines in diabetic nephropathy. J Am Soc Nephrol 2008; 19: 433–442 [DOI] [PubMed] [Google Scholar]
  • 118. Donate-Correa J, Martín-Núñez E, Muros-de-Fuentes Met al. Inflammatory cytokines in diabetic nephropathy. J Diabetes Res 2015; 2015: 948417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Leporini C, Pisano A, Russo Eet al. Effect of pentoxifylline on renal outcomes in chronic kidney disease patients: a systematic review and meta-analysis. Pharmacol Res 2016; 107: 315–332 [DOI] [PubMed] [Google Scholar]
  • 120. Liu D, Wang LN, Li HXet al. Pentoxifylline plus ACEIs/ARBs for proteinuria and kidney function in chronic kidney disease: a meta-analysis. J Int Med Res 2017; 45: 383–398 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Wu PC, Wu CJ, Lin CJet al. Pentoxifylline decreases dialysis risk in patients with advanced chronic kidney disease. Clin Pharmacol Ther 2015; 98: 442–449 [DOI] [PubMed] [Google Scholar]
  • 122. Lugnier C. Cyclic nucleotide phosphodiesterase (PDE) superfamily: a new target for the development of specific therapeutic agents. Pharmacol Ther 2006; 109: 366–398 [DOI] [PubMed] [Google Scholar]
  • 123. Cheng J, Grande JP. Cyclic nucleotide phosphodiesterase (PDE) inhibitors: novel therapeutic agents for progressive renal disease. Exp Biol Med (Maywood) 2007; 232: 38–51 [PubMed] [Google Scholar]
  • 124. Lin SL, Chen RH, Chen YMet al. Pentoxifylline inhibits platelet-derived growth factor-stimulated cyclin D1 expression in mesangial cells by blocking Akt membrane translocation. Mol Pharmacol 2003; 64: 811–822 [DOI] [PubMed] [Google Scholar]
  • 125. Lin SL, Chen RH, Chen YMet al. Pentoxifylline attenuates tubulointerstitial fibrosis by blocking Smad3/4-activated transcription and profibrogenic effects of connective tissue growth factor. J Am Soc Nephrol 2005; 16: 2702–2713 [DOI] [PubMed] [Google Scholar]
  • 126. Chen YM, Chiang WC, Lin SLet al. Dual regulation of tumor necrosis fac-tor-alpha-induced CCL2/monocyte chemoattractant protein-1 expression in vascular smooth muscle cells by nuclear factor-kappaB and activator protein-1: modulation by type III phosphodiesterase inhibition. J Pharmacol Exp Ther 2004; 309: 978–986 [DOI] [PubMed] [Google Scholar]
  • 127. Ward A, Clissold S. Pentoxifylline: a review of its pharmacodynamics and pharmacokinetic properties and its therapeutic efficacy. Drugs 1987; 34: 50–97 [DOI] [PubMed] [Google Scholar]
  • 128. Lee SY, Kim SI, Choi ME. Therapeutic targets for treating fibrotic kidney diseases. Transl Res 2015; 165: 512–530 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Toth-Manikowski S, Atta MG. Diabetic kidney disease: pathophysiology and therapeutic targets. J Diabetes Res 2015; 2015: 697010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Chen YM, Wu KD, Tsai TJet al. Pentoxifylline inhibits PDGF-induced proliferation of and TGF-beta-stimulated collagen synthesis by vascular smooth muscle cells. J Mol Cell Cardiol 1999; 31: 773–783 [DOI] [PubMed] [Google Scholar]
  • 131. Tenor H, Schudt C, Hatzelmann A.. Analysis of PDE isoenzyme profiles in cells and tissues by pharmacological methods. In phosphodiesterase inhibitors. Eur Respir J 1995; 8: 1179–1183 [DOI] [PubMed] [Google Scholar]
  • 132. Wang P, Wu P, Ohleth KMet al. Phosphodiesterase 4B2 is the predominant phosphodiesterase species and undergoes differential regulation of gene expression in human monocytes and neutrophils. Mol Pharmacol 1999; 56: 170–174 [DOI] [PubMed] [Google Scholar]
  • 133. Li H, Zuo J, Tang W. Phosphodiesterase-4 inhibitors for the treatment of inflammatory diseases. Front Pharmacol 2018; 9: 1048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Fernandes JL, De Oliveira RTD, Mamoni RLet al. Pentoxifylline reduces pro-inflammatory and increases anti-inflammatory activity in patients with coronary artery disease—A randomized placebo-controlled study. Atherosclerosis 2008; 196: 434–442 [DOI] [PubMed] [Google Scholar]
  • 135. Mohammadpour AH, Falsoleiman H, Shamsara Jet al. Pentoxifylline decreases serum level of adhesion molecules in atherosclerosis patients. Iran Biomed J 2014; 18: 23–27 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Cooper A. Pentoxifylline improves hemoglobin levels in patients with erythropoietin-resistant anemia in renal failure. J Am Soc Nephrol 2004; 15: 1877–1882 [DOI] [PubMed] [Google Scholar]
  • 137. McCormick BB, Sydor A, Akbari Aet al. The effect of pentoxifylline on proteinuria in diabetic kidney disease: a meta-analysis. Am J Kidney Dis 2008; 52: 454–463 [DOI] [PubMed] [Google Scholar]
  • 138. Tian ML, Shen Y, Sun ZLet al. Efficacy and safety of combining pentoxifylline with angiotensin converting enzyme inhibitor or angiotensin II receptor blocker in diabetic nephropathy: a meta-analysis. Int Urol Nephrol 2015; 47: 815–822 [DOI] [PubMed] [Google Scholar]
  • 139. Ruiz-Andres O, Sanchez-Niño MD, Moreno JAet al. Downregulation of kidney protective factors by inflammation: role of transcription factors and epigenetic mechanisms. Am J Physiol Renal Physiol 2016; 311: F1329–F1340 [DOI] [PubMed] [Google Scholar]
  • 140. Kuro-o M, Matsumura Y, Aizawa Het al. Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature 1997; 390: 45–51 [DOI] [PubMed] [Google Scholar]
  • 141. Donate-Correa J, Mora-Fernández C, Martínez-Sanz Ret al. Expression of FGF23/KLOTHO system in human vascular tissue. Int J Cardiol 2013; 165: 179–183 [DOI] [PubMed] [Google Scholar]
  • 142. Mencke R, Harms G, Moser Jet al. Human alternative Klotho mRNA is a nonsense-mediated mRNA decay target inefficiently spliced in renal disease. JCI Insight 2017; 2: e94375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Liao HK, Hatanaka F, Araoka Tet al. In vivo target gene activation via CRISPR/Cas9-mediated trans-epigenetic modulation. Cell 2017; 171: 1495–1507 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Liu JJ, Liu S, Morgenthaler NGet al. Association of plasma soluble α-klotho with pro-endothelin-1 in patients with type 2 diabetes. Atherosclerosis 2014; 233: 415–418 [DOI] [PubMed] [Google Scholar]
  • 145. Wu C, Wang Q, Lv Cet al. The changes of serum sKlotho and NGAL levels and their correlation in type 2 diabetes mellitus patients with different stages of urinary albumin. Diabetes Res Clin Pr 2014; 106: 343–350 [DOI] [PubMed] [Google Scholar]
  • 146. Asai O, Nakatani K, Tanaka Tet al. Decreased renal α-Klotho expression in early diabetic nephropathy in humans and mice and its possible role in urinary calcium excretion. Kidney Int 2012; 81: 539–547 [DOI] [PubMed] [Google Scholar]
  • 147. Kim SS, Song SH, Kim IJet al. Decreased plasma α-Klotho predict progression of nephropathy with type 2 diabetic patients. J Diabetes Complicat 2016; 30: 887–892 [DOI] [PubMed] [Google Scholar]
  • 148. Moreno JA, Izquierdo MC, Sanchez-Niño MDet al. The inflammatory cytokines TWEAK and TNFα reduce renal klotho expression through NF-κB J. Am Soc Nephrol 2011; 22: 1315–1325 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Zhao Y, Banerjee S, Dey Net al. Klotho depletion contributes to increased inflammation in kidney of the db/db mouse model of diabetes via RelA (serine)536 phosphorylation. Diabetes 2011; 60: 1907–1916 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Martín-Núñez E, Donate-Correa J, Ferri Cet al. Association between serum levels of Klotho and inflammatory cytokines in cardiovascular disease: a case-control study. Aging (Albany NY) 2020; 12: 1952–1964 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Kovesdy CP, Isaman D, Petruski-Ivleva Net al. Chronic kidney disease progression among patients with type 2 diabetes identified in US administrative claims: a population cohort study. Clin Kidney J 2020; 14: 1657–1664 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Fernández-Fernández B, Valiño-Rivas L, Sánchez-Niño MDet al. Albuminuria downregulation of the anti-aging factor klotho: the missing link potentially explaining the association of pathological albuminuria with premature death. Adv Ther 2020; 37: 62–72 [DOI] [PubMed] [Google Scholar]
  • 153. Seo MH, Kim DW, Kim YSet al. Pentoxifylline-induced protein expression change in RAW 264.7 cells as determined by immunoprecipitation-based high performance liquid chromatography. PLoS One 2022; 17: e0261797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Ortiz A, Asociación Información Enfermedades Renales Genéticas (AIRG-E), European Kidney Patients' Federation (EKPF) et al.RICORS2040: the need for collaborative research in chronic kidney disease. Clin Kidney J 2021; 15: 372–387 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Go AS, Yang J, Tan TCet al. Contemporary rates and predictors of fast progression of chronic kidney disease in adults with and without diabetes mellitus. BMC Nephrol 2018; 19: 146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Fang JH, Chen YC, Ho CHet al. The risk of major bleeding event in patients with chronic kidney disease on pentoxifylline treatment. Sci Rep 2021; 11: 13521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Ocak G, Rookmaaker MB, Algra Aet al. Chronic kidney disease and bleeding risk in patients at high cardiovascular risk: a cohort study. J Thromb Haemost 2018; 16: 65–73 [DOI] [PubMed] [Google Scholar]
  • 158. Acedillo RR, Shah M, Devereaux PJet al. The risk of perioperative bleeding in patients with chronic kidney disease: a systematic review and meta-analysis. Ann Surg 2013; 258: 901–913 [DOI] [PubMed] [Google Scholar]
  • 159. Leehey DJ, Carlson K, Reda DJet al. Pentoxifylline in diabetic kidney disease (VA PTXRx): protocol for a pragmatic randomised controlled trial. BMJ Open 2021; 11: e053019. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Clinical Kidney Journal are provided here courtesy of Oxford University Press

RESOURCES