Skip to main content
Wiley Open Access Collection logoLink to Wiley Open Access Collection
. 2021 Sep 21;38(2):e3494. doi: 10.1002/dmrr.3494

Erectile dysfunction and diabetes: A melting pot of circumstances and treatments

Giuseppe Defeudis 1,, Rossella Mazzilli 2, Marta Tenuta 3, Giovanni Rossini 1, Virginia Zamponi 2, Soraya Olana 2, Antongiulio Faggiano 2, Paolo Pozzilli 1, Andrea M Isidori 3, Daniele Gianfrilli 3
PMCID: PMC9286480  PMID: 34514697

Abstract

Diabetes mellitus (DM), a chronic metabolic disease characterised by elevated levels of blood glucose, is among the most common chronic diseases. The incidence and prevalence of DM have been increasing over the years. The complications of DM represent a serious health problem. The long‐term complications include macroangiopathy, microangiopathy and neuropathy as well as sexual dysfunction (SD) in both men and women. Erectile dysfunction (ED) has been considered the most important SD in men with DM. The prevalence of ED is approximately 3.5‐fold higher in men with DM than in those without DM. Common risk factors for the development of DM and its complications include sedentary lifestyle, overweight/obesity and increased caloric consumption. Although lifestyle changes may help improve sexual function, specific treatments are often needed. This study aims to review the definition and prevalence of ED in DM, the impact of DM complications and DM treatment on ED and, finally, the current and emerging therapies for ED in patients with DM.

Keywords: diabetes, diabetes complications, diabetes treatment, erectile dysfunction, hypogonadism, PDE5 inhibitors

1. INTRODUCTION

Diabetes mellitus (DM) is one of the most common chronic diseases and is characterised by impaired carbohydrate metabolism. The incidence and prevalence of DM have increased over the years, with its complications representing a serious health problem. The long‐term complications of DM include macroangiopathy, microangiopathy and neuropathy. DM is also an important cause of sexual dysfunction (SD) in both men and women, with erectile dysfunction (ED) considered the most important SD in men with DM.

A sedentary lifestyle, overweight/obesity and increased caloric consumption have been identified as common risk factors for the development of DM and DM complications. Although lifestyle changes may help improve sexual function, specific treatments are often needed.

This review therefore focuses on determining (i) the definition and prevalence of DM and ED, (ii) the impact of DM complications and other cofactors on ED and (iii) current and emerging treatments and novel approaches for the treatment of ED in patients with DM.

2. EPIDEMIOLOGY

The incidence rate of ED is estimated at 25–30 cases per thousand person years, with approximately 20%–30% of adult men having at least 1 SD. 1 Certainly, age is an important risk factor for SD, and the prevalence of SD increases with age, ranging from 1%–10% in men aged <40 years to 50%–100% in men aged >70 years.2, 3

Men with DM are at a significantly higher risk for ED than those without DM. In line with this, Corona et al. reported a 19.4%, 15.4%, 10.4% and 21.6% prevalence of mild, mild‐to‐moderate, moderate and severe ED in men with DM, respectively. 4 The severity of ED is highly dependent on the type and duration of DM, type of treatment and comorbidities.5, 6, 7 A study conducted by Fedele et al. on a large population of men with DM showed a 26% and 37% prevalence of ED among those with type 1 DM (T1DM) and type 2 DM (T2DM), respectively. 6 Similarly, a recent meta‐analysis showed that the overall prevalence rate of ED in patients with DM was 52.5% (95% confidence interval [CI], 48.8–56.2), with prevalence rates of 37.5% and 66.3% in those with T1DM and T2DM, respectively. 8 Accordingly, patients with DM had an approximately 3.5‐fold higher prevalence of ED than those without DM.

3. THE ‘COMPLICATED ROLE’ OF COMPLICATIONS AND COFACTORS IN THE PATHOPHYSIOLOGY OF ED IN DM

ED has a very complex aetiology which can involve numerous unmodifiable and modifiable factors, which are often present at the same time and usually can influence each other. Common risk factors include, besides DM, age, dyslipidaemia, hypertension, cardiovascular disease, obesity, metabolic syndrome (MetS), lack of exercise, and smoking.9, 10, 11 Vasculogenic factors are the most common and are both due to arterial inflow or venous outflow disorders. 9 Other factors include low testosterone levels, neurogenic and iatrogenic factors (related to a medical or surgical treatment).12, 13 Moreover, also a psychological component is involved, thus complicating the clinical picture and worsening the quality of life. 14

It is, therefore, clear how many risk factors between ED and DM are in common. As hyperglycaemia is associated with increased oxidative stress, and the hyper‐production of reactive oxygen species (ROS), in DM a cascade of events occurs: reduced NO, increased prothrombotic factors such as tissue factor and plasminogen activator inhibitor‐1, an increase in endothelin‐1, with subsequent thrombosis and vasoconstriction, as well as an increase in nuclear factor kappaB and activation protein 1 with subsequent inflammation, culminating in ED. 15 In fact, the aetiology of ED in diabetic subjects could be described as a ‘diabetic erectile dysfunction wall’.

The management of DM is exceedingly complex because of its acute and long‐term complications and several comorbidities. Thus, the incidence rates of ED in patients with DM increase with the number and severity of complications and comorbidities. 16 Therefore, a complete andrological evaluation cannot overlook the complex management of DM. Similarly, the framework of patients with DM cannot be complete without careful andrological evaluation given that ED symptoms also offer a great opportunity for the early diagnosis and better management of DM complications.

The main complications and comorbidities related to ED are schematically presented in Figure 1 and are discussed below. 17

FIGURE 1.

FIGURE 1

Diabetes and erectile dysfunction: a complicated connection. The major complications and comorbidities related to diabetes (DM) that contribute to erectile dysfunction (ED) are shown in the figure. The graphic representation shows the major diabetes (DM) complications and comorbidities that contribute to ED. DM complications are associated with endothelial impairment and can be distinguished in macrovascular (cardiovascular events) and microvascular (nephropathy and neuropathy). The major diabetes comorbidities which lead to ED are hypogonadism, metabolic syndrome (obesity, hypertension and dyslipidemia), obstructive sleep apnoea and depression. The picture shows how the single pathologies are strictly interconnected: thus, increasing the incidence of ED with the number and severity of comorbidities as indicated by arrows, each individual disease can worsen DM complications and increase the incidence and number of comorbidities, thus increasing the incidence and severity of ED. Abbreviations: CV: Cardio Vascular; MetS: Metabolic Syndrome; OSAS: Obstructive Sleep Apnoea Syndrome; UTI: Urinary Tract Infection

3.1. Complications

3.1.1. Cardiovascular events

Cardiovascular disease (CVD) is the main cause of mortality and morbidity in patients with DM. 18 CVD is strictly dependent on atherosclerotic complications, which are also responsible for ED.

The direct correlation between ED and CVD has been widely confirmed in the literature.9, 19, 20, 21, 22, 23 Recent real‐world observational data have shown that patients with ED have a higher prevalence of CVD and DM than patients without ED in every age group, beginning from 30 years of age, peaking between at 60–69 years, and persisting throughout life. 24 Moreover, ED can precede the onset of coronary artery disease (CAD) by 2–5 years, providing a great opportunity for the early diagnosis of silent CAD and stroke, 25 particularly in patients aged <60 years. 26 Atherosclerotic complications affect smaller arteries, such as those in cavernous areas, before affecting the larger ones (e.g., coronary, femoral and carotid arteries). The difference between vascular and chronological age (determined from the SCORE project algorithm 27 ) can be a predictor of major adverse cardiovascular events and has been correlated with poor penile colour Doppler ultrasound parameters, even in patients referred for ED without a personal history of cardiovascular events, representing an inexpensive and safe surrogate marker of penile and systemic arterial damage.28, 29

The association between ED and CVD carries considerable importance in patients with DM. 30 Despite several algorithms enabling cardiovascular and metabolic risk stratification in patients with DM, 31 a large number of silent CAD can be overlooked. Identification of ED can therefore represent an additional tool in the evaluation of the latter category of patients, especially younger patients.32, 33 ED should be considered an effective zero‐cost predictor of CVD and requires further diagnostic investigation and aggressive risk‐decreasing therapy, especially in young men. 34

3.1.2. Nephropathy

Diabetic nephropathy, which occurs in 20%–40% of patients with DM, is dependent on increased arteriolar resistance and, therefore, glomerular pressure and hyperfiltration. A strong correlation between albuminuria and ED has been reported.35, 36 The activation of the renin‐angiotensin system increases the production of pro‐inflammatory and pro‐fibrotic molecules (endothelin‐1 and urotensin II), which further increases arteriolar resistance and simultaneously reduces nitric oxide (NO) production due to changes in the renal biosynthesis of L‐arginine (a substrate of NO) and increased production of asymmetric dimethylarginine, an endogenous inhibitor of NO synthase. 37

Erectile function is therefore common in patients with chronic kidney disease (CKD), with the severity of the former being directly proportional to that of the latter. 38 Besides endothelial and vascular damage, other mechanisms are involved in the association between ED and diabetic nephropathy, especially in the advanced stages of CKD and end‐stage renal disease, regardless of DM.39, 40 First, evidence has shown that uraemic state is associated with peripheral neuropathy (also at the penile level) and hypothalamic‐pituitary‐gonadal axis derangement, 41 which results is hypergonadotropic hypogonadism, suggestive of primary testicular damage. 42 Consequently, testosterone levels decrease significantly as CKD progresses. 43 Previous studies on testicular biopsies in patients with kidney failure have shown Leydig cell abnormalities, particularly with regard to number and morphology 44 but not hypertrophy or hyperplasia, suggesting LH resistance rather than a cytotoxic effect of uraemia. In this regard, in vitro findings revealed LH receptor blockage in uraemic serum. 45 LH levels are only modestly increased during such conditions, suggesting a reduced central response to decreased testosterone levels related to a uraemic state. Patients undergoing dialysis, however, deserve a separate discussion. In such conditions, reduction in testosterone levels may depend on histological changes in the gonads induced by substances used during dialysis or from the loss of most steroid precursors during dialysis procedures. 46 Second, SD could also depend on hyperprolactinaemia, a very common finding in end‐stage renal disease or secondary hyperparathyroidism. 47 In fact, partial inhibition of parathyroid hormone release following calcitriol administration may increase testosterone levels and improve sexual function. 48 Finally, autonomic nervous system dysfunction, secondary to uraemic toxins (urea, creatinine, parathyroid hormone, myoinositol, β2‐microglobulin and asymmetric dimethylarginine), can negatively affect erectile function. 49

3.1.3. Neuropathy

Diabetic neuropathy is another important microvascular complication. Ischaemia, Advanced glycation end products (AGE) and free radical formation can impair axonal transport and nerve conduction, causing segmental demyelination. 50 Diabetic neuropathy is also involved in ED, as reported by many large studies.49, 50, 51, 52, 53, 54

Both peripheral and autonomic neuropathy contribute to ED. Peripheral neuropathy is responsible for the impairment of sensory impulses from the penis to the reflexogenic erectile centre and motor impulses to the pelvic floor muscles, which leads to a reduction in the contractile force of the bulbocavernosus and ischiocavernosus muscles and, therefore, venous outflow from the corpora cavernosa.51, 52 In addition, as previously reported, peripheral neuropathy worsens considerably in uraemic patients with renal failure. 41 On the other hand, autonomic neuropathy is characterised by a decrease or absence of parasympathetic activity, which is necessary for relaxation of the corpora cavernosa smooth muscle. 53 Moreover, autonomic postganglionic parasympathetic nerves produce neuronal Nitric Oxide Synthase. The neuronal integrity of the cavernous nerve is therefore crucial for penile erection given that the nitric oxide secreted from the nerve terminal is responsible for initiating the erectile mechanism, which is subsequently maintained by eNOS and Akt activation, 54 indicating that diabetic neuropathy compromises both the initiation and maintenance of erection.

3.2. Comorbidities

ED and DM share common risk factors and are associated with diseases causing endothelial impairment, such as hypertension, dyslipidaemia, obesity and MetS. Other comorbidities include hypogonadism, urological disease, obstructive sleep apnoea and depression. ED can therefore be identified as an independent risk factor apart from those already mentioned in patients with DM.

3.2.1. Hypertension

Hypertension is among the most common comorbidities in patients with diabetes and one of the major risk factors for CAD, CKD and consequently, premature death.55, 56, 57, 58, 59 High blood pressure (BP) can influence erection mainly because of the resulting penile arterial dysfunction, 60 including arterial stenosis and smooth muscle tissue hypertrophy, which reduce blood flow and cause endothelial damage, thereby reducing NO production. 61

Several trials have confirmed the strong association between ED and hypertension.3, 62, 63, 64, 65, 66 Results from the SPRINT trial showed that ED was very common in middle‐aged and older men with hypertension, affecting 59.9% of the patients. The authors, however, concluded that this percentage may be higher considering that patients who already received phosphodiesterase (PDE) inhibitor treatment were also included in the study. The same study also showed that systolic BP (SBP) was inversely correlated with IIEF‐5 score, even after adjusting for several covariates. 67 This correlation was also reported in a recent study on 692 patients with T1DM, which showed that the risk of ED increased significantly for each 10‐mmHg increase in SBP above normal values, even after adjusting for age, cigarette smoking and HbA1c levels. 68 Accordingly, significant reductions in both SBP and diastolic BP (DBP) have been shown to significantly improve erectile function in patients treated for coronary heart disease. 69 This improvement was even greater after excluding patients with DM, indicating that optimal BP control may not be sufficient in patients with DM.

The relationship between hypertension and ED is difficult to establish given the coexistence of many risk factors in the majority of men with ED. In contrast with previous reports, some studies showed that high BP was not correlated with an increased risk of ED 70 and that BP control did not result in better outcomes of erectile function 71 even after 6 years of follow‐up. 72

In fact, a number of pharmacological treatments for hypertension may adversely affect sexual and erectile function, further complicating the interpretation of the results of many studies. Delineating the effects of a single drug is considerably difficult given the many interrelated factors, overlapping pathologies and multiple medications associated with ED. Evidence suggests that diuretics and β‐blockers (with the exception of nebivolol) are mainly associated with ED. A study showed that β‐blockers act both centrally (i.e., reduce testosterone levels) and peripherally (i.e., act on penile vascular smooth muscle cells and cause vasoconstriction). 73 The safety of nebivolol is instead related to its vasodilating properties depending on endothelial release of NO.74, 75, 76 Thiazide diuretics and aldosterone receptor antagonists (spironolactone and eplerenone) have also been associated with ED, the former probably due to a direct effect on penile vascular smooth muscles and the latter to its anti‐androgen effect due to competitive binding with androgen receptors.77, 78, 79 On the other hand, calcium channel blockers have been associated with a neutral effect on erectile function, and angiotensin‐converting enzyme inhibitors and mostly angiotensin receptor blockers seem to even improve it given that they block the vasoconstrictive action of angiotensin‐II.80, 81, 82, 83 Given the aforementioned data, clinicians should customise anti‐hypertensive therapies, particularly in patients with DM, who are at high risk for ED, considering angiotensin receptor blockers as their first choice, followed by angiotensin‐converting enzyme inhibitors and calcium channel blockers, with nebivolol as the preferred β‐blocker therapy when needed.

3.2.2. Dyslipidaemia

Considering the significant increase in atherogenic risk caused by dyslipidaemia, the significant contribution of vascular mechanisms to ED is unsurprising. 84 Dyslipidaemia is an independent risk factor for ED in patients with DM, 85 and patients with both DM and ED have a 2.3‐fold higher risk for dyslipidaemia.69, 86 High LDL levels, low HDL levels, 87 and hypertriglyceridemia 88 have been linked to ED 89 despite the difficulty of evaluating the isolated effects of a single lipid class. Moreover, low serum testosterone levels have also been associated with dyslipidaemia, with the most recent evidence showing that testosterone replacement therapy (TRT) can have some beneficial effects on lipid metabolism in hypogonadal patients with T2DM.90, 91

Finally, statins are usually prescribed among medications related to ED given previous findings showing an association between statin use and low testosterone levels. 92 However, recent meta‐analyses have found no evidence that statins impair erectile functions.93, 94, 95

3.2.3. Obesity and MetS

T2DM has been strongly associated with obesity, leading to the coining of the term ‘diabesity’, 96 despite the increasing prevalence of overweight and obesity among patients with T1DM. 97

The accumulation of visceral adipose tissue has been associated with MetS and cardiovascular risk. Obesity, MetS and T2DM have been associated with chronic low‐grade inflammation: visceral adipose tissue promotes insulin resistance and consequent hyperglycaemia and is able to release inflammatory cytokines (TNF‐α and IL‐6), promote the endothelial expression of chemokines (IL‐1) and adhesion molecules as well as inhibit anti‐atherogenic factors (adiponectin), representing the starting condition of the inflammatory state, oxidative stress, endothelial dysfunction and therefore CVD. 98 For these reasons, ED has been frequently associated with obesity but even more with metabolic diseases.99, 100, 101, 102 According to Lotti et al. patients with metabolically healthy obesity did not have poorer erectile functions than controls, as opposed to those with metabolically complicated obesity, 103 suggesting that metabolic risk factors represent the main cause of impairments in erectile function.

Moreover, hypogonadism represents another crucial element of this complicated vicious circle. Visceral obesity and MetS are considered among the main determinants of low testosterone levels, with low testosterone levels simultaneously being an independent risk factor for obesity, MetS and T2DM, thereby reinforcing the cycle. 104

According to current clinical evidence, ED and MetS in patients with obesity mainly depend on arterial dysfunction and reduced testosterone levels. 105 Therefore, comprehensive metabolic assessment and routine hormonal screening for male hypogonadism should be recommended in male patients with obesity with concomitant DM and ED.31, 106

Weight loss should be the first‐line therapeutic approach for improving erectile function in obese men with DM. 107 Most studies that examined the impact of lifestyle modification, diet and exercise on weight loss showed an inverse correlation between body mass index (BMI) and testosterone levels, in addition to an improvement in glycaemic control and metabolic parameters.108, 109 Bariatric surgery has been shown to be more effective than nonsurgical options for weight loss in individuals with severe obesity. 110 According to a prospective long‐term randomised controlled trial (RCT) that compared surgical and nonsurgical treatments for weight loss and their subsequent impact on sexual hormones and erectile function, the former resulted in higher testosterone levels and improved erectile function. 111

Data from many single‐centre112, 113, 114, 115 and multi‐centre116, 117 high‐quality trials and meta‐analyses 90 have highlighted the beneficial effects of TRT in patients with T2DM, obesity, MetS and ED. However, despite the encouraging results, the sole presence of obesity is not enough to start TRT according to the Endocrine Society practice guidelines for obesity. Nonetheless, TRT can be considered in individual cases who cannot achieve weight loss using conventional therapeutic approaches. 107

3.2.4. Hypogonadism

ED is one of the first symptoms of hypogonadism because testosterone modulates nearly every step of erectile function 12 Considering the moderate prevalence of hypogonadism in patients with T1DM relative to those with T2DM, 118 hyperglycaemia in the absence of insulin resistance may not be enough to cause inhibition of the axis. Nevertheless, the association among hypogonadism, obesity, MetS, insulin resistance and T2DM is widely recognised and consists of a complicated vicious circle, in which obesity is a major confounder, as previously reported. 119 Initially, patients with T2DM, obesity and MetS could present with hypogonadism, with low testosterone levels also being associated with insulin resistance, further aggravating glycaemic control, favouring the uptake of free fatty acids and proliferation of adipocytes and therefore promoting adiposity.

As reported in several cross‐sectional studies,120, 121, 122, 123 up to 40% of men with T2DM exhibit low testosterone levels, with more than 90% experiencing ED. 124 Moreover, a study reported an inverse correlation between ED severity and testosterone levels. 125 In the majority of patients with DM, low testosterone levels have been associated with inappropriately low gonadotropin levels, suggesting functional secondary hypogonadism (or late‐onset hypogonadism). This could be related to both insulin resistance 126 and the negative feedback exerted by increased oestradiol due to the aromatase activity of visceral fat. 127 Sex hormone‐binding globulin (SHBG) levels are usually low in patients with DM and are inversely correlated with HbA1c levels. Low SHBG levels have been associated with an increased risk for the development of T2DM. 128 Current guidelines suggest hormone profile dosage in only patients with DM who have symptoms and signs of testosterone deficiency,31, 106 including total testosterone, LH, SHBG and free testosterone (measured directly or calculated using total testosterone, SHBG and albumin concentrations). 129

In fact, there is no consensus regarding the beneficial effects of TRT on glycaemic control in patients with T2DM. Thus far, conflicting results have been reported, with some studies reporting beneficial effects of TRT on insulin sensitivity or HbA1c reduction116, 120, 130, 131, 132 and others not.117, 121, 133, 134

Given the lack of evidence, the Endocrine Society clinical practice guideline does not recommend TRT for improving glycaemic control. Nevertheless, it is strongly recommended to provide similar treatment for ED in hypogonadal men with and without T2DM. 106 Other beneficial effects of TRT include improvements in well‐being, muscle mass and strength and bone density. However, the 2020 American Diabetes Association guidelines still highlight the need to consider the risk of increased cardiovascular events in older men with hypogonadism receiving TRT. 31 According to the recent evidences,106, 135 however, there is no consistent evidence that testosterone supplementation increases the cardiovascular risk in hypogonadal men. However, most larger studies have included retrospective cohorts, prospective studies have been underpowered and meta‐analyses have shown no significant associations. As such, TRT cannot be established as a causative factor for cardiovascular events. 136 Nonetheless, long‐term data on the impact of untreated hypogonadism on mortality have also been lacking considering that low testosterone levels are a marker of cardiovascular risk. 137 In conclusion, current recommendations lean towards an individualised prescription of TRT after careful patient evaluation and explicit discussions on the potential risks and benefits, even among patients with DM. Furthermore, such patients should always be encouraged to undertake lifestyle modifications (reduced caloric intake, healthy diet, increased daily physical activity, smoking cessation and reduced alcohol consumption) before considering TRT. 138

3.2.5. Urologic comorbidities

Urologic comorbidities can occur in both T1DM and T2DM. The primary reason for such comorbidities is urogenital tract infections (UTI), which have higher incidence rates in patients with DM (increased urinary glucose excretion, greater adherence of bacteria to the uroepithelium, chronic neurologic bladder dysfunction and weakened immune responses). Furthermore, sodium glucose transporter 2 (SGLT2) inhibitors can slightly increase the risk of UTI, despite recent meta‐analyses suggesting no increased risk of severe bacterial infections.139, 140 Male patients with DM (both T1DM and T2DM) have a threefold higher risk for asymptomatic bacteriuria and an increased risk of UTI recurrence, 141 balanitis, balanoposthitis and male accessory gland infection. 142 While Escherichia coli has been the most frequently observed pathogen, other unusual microorganisms can be found (e.g., Klebsiella, fungal infections, gram negative rods, enterococci, group B streptococci, Pseudomonas and Proteus mirabilis). 143 As such, early diagnosis of UTI is imperative to avoid complications such as emphysematous cystitis, pyelonephritis, renal failure and bacteraemia. Balanitis or balanoposthitis, which are very frequent in patients with DM, mainly depend on Candida albicans, 144 whereas male accessory gland infection often occurs in patients with DM with autonomic neuropathy. 145

DM (T2DM more than T1DM) has also been associated with benign prostatic hyperplasia and lower urinary tract symptoms (LUTS).146, 147 Both hypoxia due to vascular damage and hyperinsulinaemia are in fact important stimuli for prostate growth.148, 149 In addition, increased HbA1c levels have been associated with further increased risk of overactive bladder urgency, urge urinary incontinence and nocturia. 150 Additionally, several studies have described a potential association between neuropathic complications and LUTS, also in T1DM.151, 152, 153 Finally, Peyronie's disease (PD), a connective tissue disorder of the penis characterised by impaired deposition of collagen and formation of fibrous plaques that may cause penile deformity with consequent pain and discomfort during erections, must be considered. 154 DM has been identified as a risk factor for PD, with prevalence rates of DM among patients with PD ranging between 10% and 43%.155, 156

In conclusion, urinary complications of DM have been associated with general discomfort and decreased quality of life, unfavourable psychological effects and possibly depression and psychiatric symptoms. 157 Recent findings have revealed that among men with DM, those with LUTS have a higher prevalence of ED than those without LUTS.158, 159

3.2.6. Obstructive sleep apnoea syndrome

Obstructive sleep apnoea syndrome (OSAS) is commonly associated with T2DM.160, 161 Considering that obesity is a common risk factor for OSAS and T2DM, this relationship is not surprising and is mutual. OSAS is a widely recognised risk factor for the development of T2DM independent of obesity and other conventional risk factors. T2DM might also be a risk factor for OSAS, despite being relatively less explored in the literature. 162 ED is one of the clinical manifestations of OSAS, with both pathologies closely linked by endothelial dysfunction.163, 164, 165 Sleep apnoea treatment, particularly continuous positive airway pressure, significantly improved the quality of life and IIEF score166, 167, 168, 169 but was less effective than PDE5 inhibitors (PDE5is) such as sildenafil.170, 171 Continuous positive airway pressure, which can improve oxygenation of both the pituitary gland and corpus cavernosum, 172 can be considered as an additional treatment to optimise therapeutic outcomes in hypogonadal men. Weight loss also plays a major role and should always be encouraged. Evidence regarding OSAS treatment for glycaemic control is still conflicting, with uncontrolled studies reporting improvement but RCTs failing to show such benefits. 162

3.2.7. Depression

Sexual function can result from not only autonomic processes but also emotional and cognitive processes. Therefore, the presence of depression is unequivocally associated with ED,173, 174 reduced libido and decreased frequency of intercourse. This association is considered bidirectional although it remains unclear whether depressive syndrome precedes ED or vice versa. 175 The incidence of depression is twofold higher in patients with DM than in patients without DM.176, 177 Moreover, the presence of ED (for all the organic reasons reported so far) can worsen a previously existing depressive state, 178 while the use of antidepressants can aggravate ED. According to a recent meta‐analysis, the risk of ED in men with DM exhibiting depressive symptoms is more than six‐fold higher than that in men with DM without depressive symptoms, with overall crude and adjusted odds ratios of 6.40 and 3.08, respectively. 5

Considering that depression in patients with DM can have a significant negative impact on self‐care, glycaemic control, health outcomes and quality of life, this diagnosis should not be underestimated. 179 As such, routine assessment of depression and ED should be part of standard DM care, and psychosexual counselling should be considered as an effective tool for men with ED and DM. 180

3.2.8. Other factors

Basic science research has expanded the knowledge on ED and identified several key molecular changes related to the pathogenesis of ED, including RhoA/Rho‐associated protein kinase (ROCK) signalling pathway, renin‐angiotensin system (RAS) and tumour necrosis factor‐alpha (TNF‐α). 181

RhoA/ROCK signalling play a role in the maintaining of the flaccid penile state and its activity in the penis is a pathogenic factor contributing to ED development. To this regard, inhibition of RhoA‐ROCK signalling seems to potentiate smooth muscle relaxation in an NO‐independent manner and could be considered a new therapeutic target for the treatment of ED.181, 182, 183

Considering angiotensin‐II (Ang II), which is the primary effector of the RAS, studies have demonstrated the presence and activity in the penis. In fact, elevated Ang II levels contribute to the development of ED both in humans and animal models 184 and might be considered as a promising therapeutic target for the treatment of ED. 185

Furthermore, clinical and experimental evidence demonstrates that cytokines, among which TNF‐α, could play an important role in ED, particularly in the context of CVD. In fact, high levels of TNF‐α were demonstrated in patients with ED. 186

Finally, endothelins are potent vasoconstrictor peptides that induce contraction of trabecular smooth muscles cells of the corpora cavernosa, mainly through transmembrane calcium flux, and calcium sensitisation by the Rho‐Rho kinase pathway. Furthermore, elevated endothelin‐1 levels are found in patients with DM and this could increase the risk of ED; alternatively, alterations in endothelin receptor sensitivity, which can occur in conditions such as DM and hypertension, may enhance vasoconstrictor processes. 187

4. EFFECTS OF ANTI‐DIABETIC DRUGS ON MALE SEXUALITY

4.1. Metformin

Several studies evaluating the effect of metformin in patients with DM and ED have mostly focussed on the analysis of the four major mechanisms of arteriogenic ED 188 : (i) endothelium‐dependent vasodilatory impairment, mediated by reduced bioavailability of nitric oxide and inducing vasoconstriction; (ii) elevation of sympathetic nerve activity, resulting in enhanced basal and myogenic tone within the corpus cavernosum; (iii) atherosclerotic luminal narrowing, which leads to reduced arterial flow to the penis 189 and (iv) hypogonadism. 190

Regarding the first aspect, small arterioles of the penis dilate in response to vasoactive substances produced by endothelial cells, with NO considered the most important molecule. 191 This mechanism is called endothelium‐dependent vasodilatation. In patients with insulin resistance, increased degradation and reduced nitric oxide synthesis can promote a state of NO deficiency. 192 The reduction in NO bioavailability induces vasoconstriction of the arteries, arterioles and sinusoids of the corpus cavernous.

Several animal studies and clinical trials have highlighted the possible positive effects of metformin on endothelium‐dependent vasodilation. Vitale et al. 193 reported a correlation between the improvement of endothelium‐dependent vasodilatation and the reduction of insulin resistance after administration of metformin (500 mg BID) for 3 months in a cohort of patients with MetS compared with that in a placebo group. Mather et al. 194 highlighted similar results after administering metformin (500 mg BID) in a cohort of patients with T2DM. In their study, a significant improvement in endothelium‐dependent vasodilatation compared with that in the placebo group was observed. Nonetheless, both studies showed significant improvements only in endothelium‐dependent vasodilatation, whereas no significant difference in the mechanism of endothelium‐independent vasodilatation induced by vasoligating agents was observed.

According to Kaya et al. metformin played a protective role against endothelial dysfunction and may be useful for treating ED in patients with insulin resistance. 195

Evidence has shown that patients with DM had a higher basal tone of the sympathetic system, as demonstrated by increased production of norepinephrine196, 197 and low frequency/high frequency ratio,198, 199 indicating relative sympathetic or parasympathetic dominance. 200 Several animal and human studies have highlighted the neuromodulatory effect of metformin. These studies, which focused on the effect on BP and heart rate parameters, suggested that metformin could attenuate excessive sympathetic nerve activity and is useful in improving erectile function.201, 202, 203 Regarding hypertension and atherosclerosis, plaque formation in the penile arteries has been known to impair blood flow and promote ED. 204 Insulin resistance has been shown to induce hypertension through various mechanisms. 205 Therefore, metformin had been hypothesised to exert positive effects on BP reduction and ED. Although this effect has been demonstrated in animal models,206, 207 no clear consensus has yet been established in human clinical trials. However, recent studies showed that metformin had no benefit on BP in patients with DM.208, 209

With regard to the reduction of testosterone levels, 190 metformin treatment for insulin resistance could be hypothesised to improve testosterone levels and consequently ED. There are limited and inconsistent data regarding the role of metformin in improving hypogonadism. In 2010, Casulari et al. reported that metformin in conjunction with lifestyle modifications had positive effects in patients with metabolic and hypogonadal syndrome. The authors concluded that treatment based on healthy diet and mild physical activity associated with metformin improved insulin sensitivity and increased total and free testosterone levels, regardless of the presence of hypogonadism. 210 A study by Hayder et al. found that metformin resulted in significantly lower testosterone levels, poorer sex drive and greater ED in patients with DM than in those receiving sulfonylurea and those without DM. These results could be explained by the inhibitory activity of metformin on cytochrome P450‐C17a, which plays an important role in the synthesis of total testosterone and in the reduction of LH levels. Conversely, sulfonylureas can inhibit 11β‐hydroxysteroid dehydrogenase type 1, reducing glucocorticoid biosynthesis and stimulating testosterone synthesis. 211

Finally, the best evidence of metformin‐induced (850 mg BID) ED improvement was shown by a double‐blind pilot study on 30 men with insulin resistance and poor response to sildenafil (100 mg). The administration of metformin resulted in improved BMI and HOMA index and IIEF‐5 scores. 212

In conclusion, metformin should be considered for men with ED and MetS or DM who show poor response to PDE‐5is.

4.2. Pioglitazone

Pioglitazone exerts its anti‐diabetic effect by increasing insulin sensitivity via the activation of a nuclear receptor, the gamma isoform of peroxisome proliferator‐activated receptor (PPARγ). Accordingly, PPARγ activation showed beneficial effects in several conditions, such as endothelial dysfunction, oxidative stress, metabolic disorders, atherosclerosis and inflammation, which are risk factors for ED.213, 214 In 2006, Kovanecz et al. conducted a study in rats with T2DM and demonstrated that pioglitazone (0.6 mg/kg) could prevent and improve veno‐occlusive dysfunction in the long term, without a significant reduction in glycaemia, thereby demonstrating its vasoprotective role through a mechanism independent from glycaemic control. 215

Studies conducted using animal models have also demonstrated the neuroprotective role of pioglitazone in rats undergoing bilateral cavernosal nerve crush injury. 216 In particular, Aliperti et al. examined the effects of pioglitazone on erectile function in a rat model undergoing prostatectomy and demonstrated that pioglitazone improved ED through the NO‐mediated pathway with a dose‐dependent mechanism. 217

The only human trial investigating the effects of pioglitazone (30 mg once daily) on men with moderate‐to‐severe ED and poor response to sildenafil was conducted by Gholamine et al. who showed that the pioglitazone group had significantly better improvement in sildenafil responsiveness and erectile function determined using IIEF‐5 (from moderate‐severe ED to mild‐moderate ED) compared with the placebo group. Furthermore, they found a correlation between improvement in ED and a significant reduction in total cholesterol levels. 218

4.3. Glibenclamide

To the best of our knowledge, only one study has investigated the effects of glibenclamide on erectile function. Ruiz Rubio et al. 219 showed that the KATP channels play a key role in the relaxation of penile resistance arteries. The blockade of the KATP channels by glibenclamide (3 M) significantly reduced the relaxation of arterial muscle cells, resulting in reduced erectile function.

4.4. SGLT‐2 inhibitors

SGLT2 inhibitors, the newest class of anti‐hyperglycaemic agents, have been shown to promote urinary excretion of glucose by blocking its reabsorption in the renal proximal tubules. 220

SGLT2 inhibitors have demonstrated unprecedented cardiorenal outcomes in large‐scale clinical trials conducted on patients with T2DM who did and did not have established CVD, reducing the incidence of major cardiovascular events, total and cardiovascular mortality and hospitalisations for heart failure, as well as slowing down the progression of kidney disease.221, 222, 223, 224, 225, 226 Despite the considerable scientific interest in this class of drugs, no scientific studies have explored the impact of SGLT2 inhibitors on sexual function.

Only one trial has analysed the possible impact of SGLT2 on T2DM‐associated ED in rats treated with empagliflozin or placebo for 4 weeks, followed by treatment with sildenafil. Erectile function was assessed by measuring intra‐cavernous pressure during electrical stimulation of the cavernous nerve. The study demonstrated that empagliflozin could significantly improve erectile function in response to electrical stimulation of the cavernous nerve and sensitivity to PDE5is. Such results could be attributable to improvements in metabolic parameters, enhanced neuronal terminal recruitment with increased NO synthesis and/or release and amelioration of NANC‐mediated relaxation, thereby attenuating diabetic neuropathy. 227

4.5. DPP4i and GLP1RA

There are limited studies on the effects of DPP4i and GLP1RA on male sexual function. Evidence from animal models suggests that DPP4i could improve erectile function in patients with T2DM, promoting vascular repair and endothelial function 228 and improving the effects of vasorelaxants mediated by vascular endothelial growth factor. 229 A randomised controlled study conducted on rats with induced DM and treated with liraglutide revealed the protective role of liraglutide on the endothelium of the corpora cavernosa due to its effect on the Akt/eNOS mechanism, with the consequent improvement of erectile function. 230

Furthermore, Giagulli et al. evaluated the effect of liraglutide in obese, hypogonadal patients with T2DM who received metformin and testosterone replacement. The group with liraglutide showed greater benefits on erectile function as well as increased testosterone levels. 231

4.6. Insulin

Insulin treatment improves erectile function in diabetes‐induced rats. Yamanaka et al. demonstrated that rats treated with insulin showed higher intra‐cavernous pressure than the control group, as well as reduced levels of pro‐apoptotic factors and increased levels of anti‐apoptotic factors. 232 Similar results were obtained by Wang in 2014 and Choi in 2015.233, 234

In 2004, Shirai et al. were the first to show that insulin treatment may restore erectile function through modulation of sex hormone receptor gene and protein expression in rats with DM, increasing the mRNA and protein expression levels of androgen receptor and oestrogen receptor‐alpha. 235

5. TREATMENT OF ED IN DIABETES

5.1. Lifestyle modifications and glycaemic control

As widely reported thus far, sexual function is the result of the integrative responses of the vascular, neurologic, endocrine and psychologic systems. As such, ED treatment requires a wholistic approach, starting from correcting modifiable risk factors, promoting lifestyle changes and improving glycaemic control. In a systemic review and meta‐analysis including 740 participants from four countries, lifestyle modifications aimed at decreasing cardiovascular risk demonstrated an improvement in sexual function. 236 Several RCTs analysing the effects of reduced caloric intake, better food quality with a Mediterranean diet and increased physical activity reported improvements in metabolic parameters, anthropometric measurements and sexual function.237, 238

A weight loss of 5% from the initial weight has been the minimum recommended weight loss percentage for clinical benefit. Greater weight loss leads to even greater benefits in terms of reducing BP; improving LDL and HDL levels and insulin resistance and reducing the number of medications to control DM, hypertension and dyslipidaemia.239, 240 As previously reported, weight loss is strongly recommended to improve erectile function considering that it can increase testosterone levels mainly by improving testicular function and reducing conversion of testosterone to β‐oestradiol via aromatase activity in the adipose tissue, 127 as well as increasing SHBG concentration as a consequence of reduced insulin levels. 241 While TRT can be considered in selected cases, 107 other strategies for weight loss include bariatric surgery 110 and some specific anti‐diabetic drug classes, such as GLP1 receptor antagonist and SGLT2 inhibitors. However, evidence regarding the impact of these drugs on testosterone levels remains unclear.242, 243

Physical exercise is strongly recommended for both patients with T1DM 244 and T2DM 245 given its contribution to weight loss, improved blood glucose control, reduced cardiovascular risk and improved well‐being and sexual function.246, 247, 248 While physical exercise may improve serum testosterone levels, high‐intensity exercise can also be counterproductive.249, 250, 251

In some cases, however, intensive lifestyle intervention failed to produce significant improvement in erectile function,238, 252 suggesting that this approach would be most likely effective in cases of mild ED. Thus, most cases may require combined therapy.

5.2. PDE5is

PDE5is have been considered the first‐line therapy for ED of any aetiology. 253 PDE5 inhibition increases cGMP levels in cavernosal tissues, leading to smooth muscle relaxation, increased arterial blood flow, venous constriction and erection. 254 Several clinical trials, however, have shown that this class of drugs had blunted efficacy in patients with DM.255, 256 The lower efficacy of PDE5is in this population can be attributed to the multifactorial nature of the disease. In patients with DM, macrovascular complications and impaired neurogenic and endothelium‐dependent relaxation of penile arteries reduce blood supply to the penile arteries. Moreover, the concomitant use of different medications to treat diabetes‐related comorbidities and the higher incidence of hypogonadism may contribute to the reduced effectiveness of PDE5is. 257 Several studies have also shown that alterations in both the structures and molecular pathways supporting erectile function lead to reduced nitric oxide levels and responsiveness, thereby decreasing cGMP concentration in penile tissues.258, 259 This often requires higher PDE5i doses or combined therapies to achieve satisfactory results. 259

While four main PDE5is are available globally (sildenafil, tadalafil, vardenafil and avanafil), other agents, such as mirodenafil, udenafil and lodenafil, are available in certain countries. 260 Structural differences among these PDE5is result in differences in pharmacokinetics and dynamic properties, which translate to different durations of action and methods of administration. 261 Most PDE5is have a half‐life ranging from 2 to 5 h, with a mean duration of action of 6–12 h and an onset of action of 30–60 min. 262 In addition to on‐demand use given their longer half‐life and duration of therapeutic effect, tadalafil and udenafil are suitable for low‐dose daily administration, 263 and this regimen has been approved for ED treatment.262, 264 Daily tadalafil and udenafil use, however, did not show a significant advantage over on‐demand regimens in a diabetic population. 265

Several clinical trials have demonstrated the effectiveness of PDE5is in improving ED in men with DM (Table 1). A recent meta‐analysis of 12 clinical trials including 3124 men with DM and six PDE5is showed that PDE5is significantly improved erectile function compared with placebo, as measured using the IIEF, SEP and GAQ questionnaires, with a good safety profile and tolerability. 266 Another systematic review and meta‐analysis found that all PDE5is showed similar effectiveness, with slightly superior response rates for sildenafil over tadalafil and vardenafil, 267 However, considering variations in the efficacy reported across the studies, superiority of one medication over the other cannot be determined. Conversely, in 2018, Liao et al. performed SUCRA and NMA analyses of data from 15 RCTs involving patients with DM, demonstrating a possible advantage of vardenafil and mirodenafil over other PDE5is in terms of efficacy and adverse effects, with an overall greater efficacy‐safety profile. 265

TABLE 1.

Clinical trials considering PDE5is in men with diabetes

PDE5i
Author Study design No of patients Disease Mean or range age Intervention Methodologies and scale Results
Price et al. 1998 Prospective, DB, PC, three‐way crossover study 21 T2DM 42–65
  • In part I single dose sildenafil 25–50 mg or placebo

  • In part II once‐daily dosing sildenafil 25–50 mg or placebo

  • For 10 days

penile plethysmography daily diary records of erectile activity and a global efficacy question Improvement in penile rigidity and erectile activity
Rendell et al. 1999 Prospective, RCT, DB, PC, flexible‐dose 268 T1 and T2DM 57 Sildenafil 25–50–100 mg or placebo IIEF 56% of sildenafil improved their erections versus placebo
Boulton et al. 2001 Prospective, PC, DB, and flexible‐dose escalation study 219 T2DM 59 Sildenafil 50 mg (25–50–100 escalation/adjusting) IIEF Improved in EF domain (mean value 10.0)
Safarinejad et al. 2004 Prospective, RCT, PC, DB 282 T2DM 46.4 Sildenafil 100 mg or placebo IIEF 59% of patients improved their IIEF
Behrend et al. 2005 Prospective, self‐ reported, flexible‐dose 33 T1 and T2DM 58.1 Sildenafil 25 to 100 mg IIEF Improved IIEF score (mean value 16,9)
Blonde et al. 2006 Retrospective analysis of pooled data from RCT PB 974 T2DM 57 Flexible‐dose sildenafil (25, 50, or 100 mg) or placebo IIEF Improvement in erections (62% vs. 18%) and in percentage of successful sexual intercourse attempts (52.6 ± 5.0 vs. 22.4 ± 5.1)
Deyoung et al. 2011 Prospective, RCT, DB, PC 24 T2DM 59.8 (plb) Daily sildenafil 50 mg IIEF‐5 Improved IIEF‐5 and FMD (2‐fold increase in the brachial artery diameter)
59.4 (50 mg) Or placebo
Utku Kirilmaz et al. 2015 Prospective, open RCT 83 T2DM 54.9 Sildenafil 100 mg in addition to lifestyle modifications and glycaemic control IIEF Improved IIEF: 5.0 points for sildenafil versus 2.5 control
Goldstein et al. 2003 Prospective, DB, PC, fixed‐dose parallel‐group 452 T1 and T2DM 57 Vardenafil 10 or 20 mg or placebo IIEF Improved IIEF (5.9/7.8 for 10/20 mg, respectively)
Ziegler et al. 2006 Prospective, RCT, PC, DB 302 T1DM 50.3 Vardenafil 5–20 mg flexible dose or placebo IIEF Improved EF (mean value 7.79)
Ishii N et al. 2006 Prospective, RCT, DB, PC, multi‐centre, parallel 778 T2DM 26–64 Vardenafil 10 or 20 mg or placebo IIEF Improved EF domain score (mean value 7 for both)
Daniele santi et al. 2016 Prospective, DB PC RCT 54 T2DM 55.8 Vardenafil 10 mg BID or placebo IIEF, FMD, testosterone level Higher IIEF and FMD scores in Vardenafil group. Total testosterone increased in hypogonadal diabetic subjects in vardenafil group
Sáenz de Tejada et al. 2002 Prospective, RCT, DB, PC 191 T2DM 55.7 Tadalafil 10–20 mg or placebo IIEF Improvement of erections in 56% and 64%, respectively, versus 25% of placebo
Fonseca et al. 2004 Prospective, RCT, DB, PC parallel group 637 T2DM 57 Tadalafil 10 mg, 20 mg, or placebo IIEF Mean improvement of 7.4 in IIEF
Buvat et al. 2006 Prospective, randomised, crossover, open‐label 762 T1 and T2DM 57 Chronic versus On‐demand of tadalafil 20 mg IIEF, SEP Mean improvement of 9 points in IIEF and 44 of SEP
Hatzichristou et al. 2008 Prospective, RCT, DB, PC 298 T2DM
  • 58 (plb)

  • 57 (2.5 mg)

  • 56 (5 mg)

Tadalafil 2.5 mg, tadalafil 5 mg or placebo IIEF, SEP, GAQ Improvements in IIEF and in mean success rates for vaginal penetration, completion of intercourse, and overall treatment satisfaction.
Mustafa Suat Bolat et al. 2018 Single centre retrospective study 63 T2DM
  • 60.5 (5 mg)

  • 60.9 (20 mg)

Tadalafil 5 mg daily or Tadalafil 20 mg on demand IIEF, IPSS, MSHQ, EHS Improvement in IIEF: mean 3/2,3 (<65 years), respectively, and 2,8 (>65 years only 5 mg group); improvement in IPSS, MSHQ and EHS
Goldstein et al. 2012 Prospective, RCT, DB, PC 390 T2DM 58 Avanafil 100/200 mg or placebo IIEF Improvement of IIEF 4.5/5.4 points, respectively
Moon et al. 2011 Prospective, RCT, DB double‐dummy, parallel‐group design multicenter study, fixed‐dose trial 174 T2DM 55 Udenafil 100 or 200 mg or placebo IIEF Improvements in the IIEF‐EFD score
Soon Hyun Park et al. 2015 Prospective, multi‐centre, randomised, open‐label, parallel‐group 161 T2DM
  • 54.4 (50 mg)

  • 53.9 (200 mg)

Udenafil 50 mg once‐daily or Udenafil 200 mg on demand IIEF, GAQ, SEP Similar improvements in IIEF, SEP and GAQ
Hyun Jun Park et al. 2010 Prospective, multicenter, DB, PC, parallel‐group, fixed‐dose RCT 112 T2DM
  • 55.5 (plb)

  • 57.3 (100 mg)

Mirodenafil 100 mg or placebo IIEF, GAQ, SEP Improvement in IIEF‐EF domain scores (9.3 ± 6.8 vs. 1.4 ± 6.1); SEP2 SEP3 and GAQ improved

Abbreviations: BP, blood pressure; CRP, c reactive protein; DB, double blind; ED, erectile dysfunction; ED, erectile dysfunction; ET endothelial; GAQ, Global Assessment Questions; IIEF, International Index of Erectile Function; IL‐6, interleukin 6; NK, not know; PC, placebo controlled; PDE5is, phosphodiesterase type 5 inhibitor; RCT, randomised controlled trial; SEP, Sexual Encounter Profile; T1DM and T2DM, Type 1 and Type 2 Diabetes Mellitus; TID, three times a day.

5.3. Sildenafil

Sildenafil was the first oral drug approved for ED treatment in 1998. 268 In addition to PDE5 inhibition, sildenafil also weakly inhibits PDE6, an enzyme present at high concentrations at the retinal rod and cone photoreceptors, potentially leading to mild impairment in colour discrimination during peak plasma levels approximately 1 h after drug ingestion. 269

A recent meta‐analysis of eight RCTs that compared the effectiveness of sildenafil at dosages varying from 25 to 100 mg to that of placebo in patients with DM who had ED reported an overall response rate of 57.7%, with a significant improvement in IIEF scores (particularly for questions 3 and 4) and a relative risk of 3.99 for answering ‘yes’ to the global efficiency question (95% CI, 2.58–6.18) in sildenafil users. 257 The most common adverse events included headaches, dyspepsia and flushing, with their intensities ranging from mild to moderate, resulting in a 2.4% discontinuation rate in the sildenafil arm.

In addition to on‐demand regimens, daily sildenafil administration has also been studied. In a double‐blind placebo‐controlled RCT, 24 men with DM were assigned to receive daily sildenafil 50 mg or placebo. After 10 weeks, their IIEF scores and endothelial function, monitored via brachial artery flow‐mediated dilatation, improved. 270 Similarly, Price et al. reported an improvement in erectile activity in 21 men receiving sildenafil 50 mg daily. 271

5.4. Vardenafil

Vardenafil, which is available in two formulations (a film‐coated tablet and an orodispersible tablet), is generally well tolerated but should be avoided in patients with prolonged QT interval or in men using antiarrhythmic drugs such as procainamide, quinidine, sotalol, or amiodarone. 272 Similar to sildenafil, vardenafil weakly inhibits PDE6 and may have similar side effects on retina photoreceptors. 273

Several studies have analysed the effectiveness of vardenafil in patients with DM. Accordingly, a 12‐week prospective, multi‐centre, double‐blind, placebo‐controlled, fixed‐dose, parallel‐group trial with 452 patients with DM reported an improvement in erections in 57% of men taking vardenafil 10 mg and 72% of men taking vardenafil 20 mg compared with 13% of those receiving placebo. 274 Moreover, an improvement of 5.9 and 7.8 points in the EF domain of the IIEF for vardenafil 10 and 20 mg had been observed, respectively, with the placebo group showing an improvement of 1.4 points. There was no correlation between HbA1c levels and vardenafil effectiveness. Adverse events, mostly headache, flushing and rhinitis, were reported as mild to moderate by 13% of the patients.

Similar results were reported by Ishii et al. 275 in an RCT involving 778 patients with DM, with a mean IIEF improvement of 7 points in the 10 and 20 mg vardenafil groups and a response rate of 63% and 57% for men allocated to the 20 and 10 mg group, respectively, with greater response rates in patients with severe ED.

Chronic vardenafil therapy also appeared to be effective and may improve hypogonadism in hypogonadal men with DM. In a 2016 placebo‐controlled RCT with 54 patients with DM, vardenafil (10 BID) resulted in higher IIEF‐15 and flow‐mediated dilatation scores. Interestingly, total testosterone levels, measured using liquid chromatography, increased to normal levels in hypogonadal patients in the vardenafil arm. 276

5.5. Tadalafil

Tadalafil has the longest half‐life among PDE5is (17.5 h), with a duration of action of 24–36 h. 277 Owing to the drug's delayed metabolism, its pharmacokinetic properties are not affected by food ingestion and gastric emptying. 278 Tadalafil is also able to inhibit PDE11, with this interaction having been thought to be responsible for some of its adverse effects, such as myalgia and back pain. 279

Tadalafil is suitable and has been approved for both low‐dose daily use (2.5–5 mg) and on‐demand use for ED treatment. Both treatments were found to be successful in the management of ED among patients with DM. In a meta‐analysis of 12 RCTs including 637 patients with DM, the use of tadalafil 20 mg resulted in a 53% response rate and a mean improvement in their IIEF score of 7.4 points, with the placebo group showing a 0.9 point improvement, independent of glycaemic control. 256 Similarly, a 12‐week placebo‐controlled RCT involving 298 patients with DM found that daily use of tadalafil 2.5 or 5 mg promoted significant improvements in both IIEF and SEP2 and SEP3 scores, with response rates of 41% and 46%, respectively. 280 The most frequent adverse events included headaches, myalgia and back pain, which were classified as mild‐to‐moderate in most cases. Interestingly, in addition to its effectiveness in ED treatment, daily use of tadalafil seemed to be effective in improving endothelial dysfunction, one of the most important pathogenetic factors in diabetic ED. 281

A recent single‐centre retrospective clinical study comparing the effects of tadalafil 5 mg daily and 20 mg OD in patients with DM revealed that both regimens promoted similar IIEF improvements in patients aged <65 years, whereas only the daily regimen facilitated significant improvements in patients aged >65 years. Both treatments also improved LUTS, with greater benefits observed in the 5‐mg group. 282

5.6. Avanafil

Avanafil, which was approved in 2012 for the management of ED, is highly selective for PDE5 versus other PDE isoenzymes such as PDE1, PDE5 and PDE11. This may result in better safety profiles in terms of lower incidences of adverse events such as visual disturbances, myalgia and back pain. 283 Although avanafil is generally well tolerated, it should not be administered to patients with severe liver damage. 284

Avanafil's effectiveness in patients with DM was evaluated in an RCT with 390 men with DM experiencing ED over a period of 12 weeks. Avanafil 100 or 200 mg resulted in significant improvements in the IIEF‐EF domain, SEP2 and SEP 3 compared with placebo. The adverse events were similar to those observed with other PDE5is. 285

5.7. Udenafil

Udenafil is a novel PDE5i that is approved in only a few countries, such as Korea, Russia and the Philippines. Based on data from clinical trials, udenafil has a half‐life of approximately 11–13 h and a duration of action of 24 h. 277 Considering its pharmacokinetic properties, it could be used both once daily and on‐demand. Studies in patients with diabetes have confirmed the efficacy of both regimens.286, 287

A 12‐week, placebo‐controlled, double‐blind, parallel‐group study involving 147 men with DM experiencing ED found that the use of udenafil 100 and 200 mg resulted in significant improvements in the IIEF‐EFD score, SEP diary and GAQ, with no significant differences between the 100‐ and 200‐mg groups. 286 The most frequent adverse events included flushing, headaches, nausea and conjunctival hyperaemia, which were reportedly mild in most cases.

Similar results were obtained in a 12‐week study that compared udenafil 50 mg once daily and udenafil 200 mg on‐demand in men with DM previously treated with idenafil 200 mg. 287 At the end of the 8‐week intervention period, both groups had similar improvements in IIEF‐EFD (10.78 points for 200 mg and 9.97 points for 50 mg daily), SEP Q2 and Q3 and GAQ. These improvements were not maintained during the 4‐week drug‐free period. Both regimens were well tolerated, with flushing and headaches recorded as the most frequent adverse events.

5.8. Mirodenafil

Mirodenafil is a second generation PDE5i with high selectivity for PDE5 over other PDE isoforms. Despite its high PDE5 selectivity, mirodenafil has a slightly greater affinity for PDE6 than sildenafil. However, no visual adverse effects have been reported in men taking mirodenafil to date. 288

Mirodenafil is an effective and well‐tolerated therapy in a broad range of patients with ED. 289 In the treatment of diabetic ED, 100 mg mirodenafil has been shown to have a positive impact on IIEF‐EF scores (9.3 ± 6.8 vs. 1.4 ± 6.1 in the placebo group), SEP2, SEP3 and GAQ, with a response rate of 61.8%. 290 The reported adverse events mostly included headaches and flushing, which were mild in intensity without serious adverse events.

5.9. PDE5is and TRT

Despite PDE5is being an effective treatment for diabetic ED, up to 50% of subjects failed to respond to this drug class. 259 Given the high prevalence of hypogonadism in patients with DM, 291 reduced testosterone levels have been speculated to contribute to the unresponsiveness of PDE5is in this subpopulation.

A recent meta‐analysis showed that TRT alone was able to significantly improve erectile function and other sexual parameters in hypogonadal men, with higher benefits for patients with more severe hypogonadism (total testosterone <8 nmol/L) and lesser benefits for those with comorbidities such as DM and obesity. 292 These findings may be due to the presence of concomitant micro‐ and macro‐vascular complications and endothelial dysfunction, in addition to the more severe testosterone deficiency noted in this population. Testosterone therapy, however, has been shown to ameliorate sexual health as well as improve insulin resistance and lipid profile in hypogonadal men with DM. 116

Nonetheless, whether testosterone replacement is able to increase the effectiveness of PDE5is remains inconclusive. A meta‐analysis of 12 RCTs evaluating the effects of testosterone therapy together with PDE5is showed a positive effect in uncontrolled studies but not in placebo‐controlled studies. 293 However, three of the five placebo‐controlled RCTs enroled mixed eugonadal/hypogonadal patients, whereas in another trial, testosterone therapy was started after a sildenafil alone run‐in period when subjects had already achieved eugonadism. 294 In another study, testosterone therapy had no effect on sexual function in subjects without testosterone deficiency at baseline. 295

Multiple studies have shown that restoring sexual activity in patients with ED using PDE5is can increase testosterone levels296, 297 and restore eugonadism. 294 Therefore, PDE5i‐only therapy may be useful for men with mild hypogonadism related to diminished sexual activity caused by anxiety or other psychological disorders. If hypogonadism persists, testosterone replacement should be started. The TADTEST study showed that adding testosterone to daily tadalafil therapy in subjects unresponsive to PDE5is was beneficial only in hypogonadal men with baseline testosterone levels of ≤3 ng/mL, suggesting the need for testosterone therapy in subjects with a higher degree of testosterone deficiency. 298

Similarly, the administration of parenteral testosterone undecanoate for 102 weeks to 29 men with DM who were unsuccessfully treated for ED with PDE5is with subnormal plasma testosterone levels (total testosterone <3.5 ng/mL) resulted in higher IIEF scores as well as improvement in MetS parameters. 299

Clinical studies have shown that PDE5i use alone can increase testosterone levels by approximately 3–6 nmol/L in healthy subjects and 2–4 nmol/L in patients with DM. 300 This is unlikely to lead to adequate levels in men with severe hypogonadism, especially when secondary to a clearly identified cause. As such, testosterone replacement is preferred in these subjects given its ability to restore sexual function alone, with the option of adding PDE5is when results are unsatisfactory.

Finally, androgen therapy can improve several diabetes‐related comorbidities other than ED; therefore, these aspects should be considered to initiate adequate treatment.

5.10. PDE5is and the cardiovascular system

Over the past decade, considerable interest has emerged regarding the benefit of PDE5is in CVD associated with T2DM. T2DM and CVD show similar alterations, such as increased levels of pro‐inflammatory factors, endothelial dysfunction and decreased nitric oxide‐mediated vasodilation. 301 Therefore, it is unsurprising that PDE5i therapy can exert cardioprotective effects in patients with DM.

Data from retrospective and observational studies have shown that PDE5is can reduce age‐related mortality in men with T2DM, 302 as well as all‐cause mortality and MI incidence and mortality. 303

Although not fully understood, the mechanisms by which PDE5is are able to ameliorate cardiovascular derangements in T2DM seem to involve improvement of endothelial dysfunction, systemic inflammation and nitric oxide bioavailability. 301

Studies have shown that treatment with sildenafil, vardenafil and tadalafil improved FMD, a measure of nitric oxide‐mediated endothelial function, 304 vascular inflammation markers and endothelin‐1 levels.305, 306 Interestingly, the cardiovascular and endothelial benefits of PDE5i seem to be independent of glycaemic control, 307 as evaluated by HbA1c levels. PDE5i treatment has also been shown to reduce the levels of pro‐inflammatory factors such as Il‐6, Il‐8, TNF‐α, CXCL‐10 and IL‐1β306, 308, 309 in both humans and animal models.

The effects of PDE5is on diabetic cardiomyopathy and related complications is another promising field that has recently been explored. Results from animal models showed that tadalafil could reverse alterations in cytoskeletal/contractile proteins in the heart, attenuate mitochondrial dysfunction,310, 311 and reduce infarct size induced by ischaemia reperfusion. 308 Vardenafil was also found to be effective in reducing myocardial hypertrophy and fibrotic remodelling, 312 decreasing apoptosis and nitro‐oxidative stress via the cGMP‐PKG pathway, whereas sildenafil‐treated mice showed improved cardiomyocyte contractility 313 due to enhanced cGMP‐PKG and phosphodiesterase 3‐dependent cAMP signalling.

PDE5i‐mediated augmentation of NOS3 activity in the heart and mitochondrial ATP‐sensitive potassium channel opening have been shown to reduce ischaemia/reperfusion injury, infarct size and the incidence of cardiomyocyte death.314, 315 Similarly, PDE5is‐mediated augmentation of the cGMP‐PKG pathway appeared to ameliorate the clinical condition of patients with heart failure with reduced ejection fraction through improvements in pulmonary circulation, cardiac remodelling and diastolic function 316 but had few effects in patients with reserved ejection fraction. 317 Other trials, however, have demonstrated increased cardiac performance,318, 319 such as left ventricle diastolic function, cardiac geometry and clinical status, in patients with T2DM.

Despite being used for ED, PDE5is clearly have favourable effects on the cardiovascular system, primarily due to the restoration of the NO‐cGMP‐PKG signalling pathway and amelioration of the pro‐inflammatory state that characterises T2DM and the consequent endothelial dysfunction and increased cardiovascular risk. Although results from preclinical and human studies have been promising, additional studies are needed to determine the role of PDE5is in cardiovascular protection in T2DM.

5.11. Other treatments for ED

For patients unresponsive to medical and lifestyle treatments, several second‐ and third‐line therapies are available, the most popular of which is alprostadil (prostaglandinE1 [PGE1]). It can be used in the form of a urethral suppository, which was first marketed as medicated urethral system for erection, via intracavernosal injections, directly into the corpora cavernosa via a small needle, or as a topical cream. PGE1 increases cAMP levels via adenylate cyclase stimulation, leading to smooth cell relaxation, vasodilation and penile erection.

Intracavernosal alprostadil has been one of the oldest and most effective treatments for ED in individuals with DM. Accordingly, studies conducted in patients with both T1DM and T2DM have showed satisfactory sexual activity and intercourse after 76.5%–99% of alprostadil injections.320, 321 The most reported adverse event was penile pain, in 24%–61% of patients, although this was tolerable in most of the patients. Priapism and fibrosis have also been found to occur after alprostadil injections, albeit rarely. 322 Interestingly, a combination of alprostadil and α‐lipoic acid, administered as an intracavernosal injection, was more effective (95.0% vs. 80.5%, p < 0.05) than tadalafil (5 mg) and was able to improve vascular endothelial function and erection hardness with a good safety profile. 323 Despite the need for injections and possible injection‐related issues (pain/difficulty), alprostadil injections have been found to be a safe and effective alternative in the long term, although a mixture of different vasoactive drugs, such as papaverine and phentolamine, instead of PGE1 alone has often been used. 324 Patients with diabetes, especially those using insulin therapy, have higher compliance with self‐injections compared to those without diabetes. 325 After years of treatment, however, they usually reduce injection frequency and may stop treatment, necessitating frequent follow‐up visits with dosage adjustments to prevent dissatisfaction and drop‐out from treatment, which occurs in up to 68% of patients in the first 3 months 326

Intraurethral alprostadil consists of a polypropylene applicator with a pellet of alprostadil at its tip. Although its overall efficacy rate is approximately 60%, 327 it causes dose‐dependent penile pain and burning in 10% of treated patients. Despite its lower efficacy compared to intracavernosal PGE1 injections, the lack of injection‐related issues and its easy administration makes it appealing for patients that dislike injectable therapies. In fact, one trial showed that intraurethral alprostadil was effective in up to 65% of patients with DM, with variable efficacy based on the severity of ED. 328

Alprostadil cream can be another promising treatment for ED. Phase 2 and 3 trials have found that alprostadil cream administered into the external urethral meatus promoted adequate erection in 74%–83% of patients,329, 330 with similarly good results in populations with diabetes and those unresponsive to PDE5is.

Among the other treatments, vacuum erection devices represent a non‐invasive alternative, which has been initially effective in approximately 90% of patients. It is a cylindrical mechanical device which is placed over the penis and pumped; consequently, it creates a negative pressure vacuum to draw blood into the penis. 331 However, these devices are often heavy and uncomfortable to use, with 64% patient discontinuation rates at 2 years 332 Trials among individuals with diabetes showed that vacuum devices are effective in up to 75% of users, 333 with improvements in psychological status, peak systolic velocity and diameter of cavernosal artery evaluated using Doppler colour flow imaging 334 after 6 months of treatment. Vacuum devices in combination with sildenafil in men with diabetes promoted better successful penetrations (73.3% vs. 46.6%) and successful intercourses (70% vs. 46.6%) at 3 months compared to sildenafil alone, 335 with higher Mean International Index of Erectile Function scores. The most reported adverse events included penile pain, numbness and bruising.

Among the surgical approaches for ED, penile prosthesis is the most attractive and effective option for patients unresponsive to medical and second line treatments. This approach represents a permanent solution for the problem.

Penile implants include malleable and inflatable devices. The malleable implant consists of two semi‐rigid rods that are placed in the corpora cavernosa, and it is bent upwards before intercourse. On the other hand, two‐piece inflatable penile prostheses involve two cylinders with a scrotal pump, which allows fluid to be transferred to the cylinder chambers when an erection is desired. 336

Despite being a substantially effective treatment for ED, with satisfaction rates greater than 95%,337, 338 men with diabetes seem to be more prone to developing prosthesis infections. 339 The risk, however, has reduced over the decades with device improvement and surgical expertise, 340 with evidence identifying short‐term glucose control (BG > 200 mg/dL), but not HbA1c levels, as the strongest risk factor for prosthesis infection. 341

Finally, penile revascularisation surgery techniques were developed in order to anastomose the inferior epigastric artery to either the dorsal artery or deep dorsal vein, to improve penile vascular inflow while reducing venous outflow; however, no standardised method is available. 258

6. CONCLUSIONS

This review underscores and confirms the importance of appropriately managing patients with DM experiencing ED, highlighting the role of comorbidities, complications and the role of anti‐diabetic drugs. In this regard, both metformin and new anti‐diabetic medications seem to play a protective role in ED pathogenesis. Finally, evidence shows that PDE5is could be considered safe and beneficial for CVD associated with T2DM.

CONFLICT OF INTEREST

All authors declare no conflict interests.

ETHICS STATEMENT

Not applicable.

AUTHOR CONTRIBUTIONS

Giuseppe Defeudis, Rossella Mazzilli, Andrea M. Isidori and Daniele Gianfrilli conceived and designed the study; Giuseppe Defeudis, Rossella Mazzilli, Marta Tenuta, Giovanni Rossini, Virginia Zamponi, Soraya Olana contributed to the acquisition of data and wrote the manuscript; Giuseppe Defeudis, Rossella Mazzilli, Antongiulio Faggiano, Paolo Pozzilli, Andrea M. Isidori and Daniele Gianfrilli critically revised the manuscript. All authors have read and approved the final manuscript.

PEER REVIEW

The peer review history for this article is available at https://publons.com/publon/10.1002/dmrr.3494.

ACKNOWLEDGEMENT

Dr Massimo Ciafrei as graphic designer.

Open Access Funding provided by Universita Campus Bio‐Medico di Roma within the CRUI–CARE Agreement.

Defeudis G, Mazzilli R, Tenuta M, et al. Erectile dysfunction and diabetes: a melting pot of circumstances and treatments. Diabetes Metab Res Rev. 2022;38(2):e3494. 10.1002/dmrr.3494

Giuseppe Defeudis and Rossella Mazzilli should be considered joint first author.

DATA AVAILABILITY STATEMENT

Not applicable.

References

  • 1. Lewis RW, Fugl‐Meyer KS, Bosch R, et al. Epidemiology/risk factors of sexual dysfunction. J Sex Med. 2004;1(1):35‐39. [DOI] [PubMed] [Google Scholar]
  • 2. Lewis RW, Fugl‐Meyer KS, Corona G, et al. Definitions/epidemiology/risk factors for sexual dysfunction. J Sex Med. 2010;7(4 Pt 2):1598‐1607. [DOI] [PubMed] [Google Scholar]
  • 3. Nicolosi A, Moreira ED, Jr , Shirai M, Tambi MIBM, Glasser DB. Epidemiology of erectile dysfunction in four countries: cross‐national study of the prevalence and correlates of erectile dysfunction. Urology. 2003;61(1):201‐206. [DOI] [PubMed] [Google Scholar]
  • 4. Corona G, Giorda CB, Cucinotta D, Guida P, Nada E, Gruppo di studio S‐D. Sexual dysfunction at the onset of type 2 diabetes: the interplay of depression, hormonal and cardiovascular factors. J Sex Med. 2014;11(8):2065‐2073. [DOI] [PubMed] [Google Scholar]
  • 5. Wang X, Yang X, Cai Y, Wang S, Weng W. High prevalence of erectile dysfunction in diabetic men with depressive symptoms: a meta‐analysis. J Sex Med. 2018;15(7):935‐941. [DOI] [PubMed] [Google Scholar]
  • 6. Fedele D, Coscelli C, Santeusanio F, et al. Erectile dysfunction in diabetic subjects in Italy. Gruppo Italiano Studio Deficit Erettile nei Diabetici. Diabetes Care. 1998;21(11):1973‐1977. [DOI] [PubMed] [Google Scholar]
  • 7. Kamenov ZA. A comprehensive review of erectile dysfunction in men with diabetes. Exp Clin Endocrinol Diabetes. 2015;123(3):141‐158. [DOI] [PubMed] [Google Scholar]
  • 8. Kouidrat Y, Pizzol D, Cosco T, et al. High prevalence of erectile dysfunction in diabetes: a systematic review and meta‐analysis of 145 studies. Diabet Med. 2017;34(9):1185‐1192. [DOI] [PubMed] [Google Scholar]
  • 9. Gandaglia G, Briganti A, Jackson G, et al. A systematic review of the association between erectile dysfunction and cardiovascular disease. Eur Urol. 2014;65(5):968‐978. [DOI] [PubMed] [Google Scholar]
  • 10. Besiroglu H, Otunctemur A, Ozbek E. The relationship between metabolic syndrome, its components, and erectile dysfunction: a systematic review and a meta‐analysis of observational studies. J Sex Med. 2015;12(6):1309‐1318. [DOI] [PubMed] [Google Scholar]
  • 11. Pizzol D, Smith L, Fontana L, et al. Associations between body mass index, waist circumference and erectile dysfunction: a systematic review and META‐analysis. Rev Endocr Metab Disord. 2020;21(4):657‐666. [DOI] [PubMed] [Google Scholar]
  • 12. Isidori AM, Buvat J, Corona G, et al. A critical analysis of the role of testosterone in erectile function: from pathophysiology to treatment‐a systematic review. Eur Urol. 2014;65(1):99‐112. [DOI] [PubMed] [Google Scholar]
  • 13. Yafi FA, Jenkins L, Albersen M, et al. Erectile dysfunction. Nat Rev Dis Primers. 2016;2:16003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Brotto L, Atallah S, Johnson‐Agbakwu C, et al. Psychological and interpersonal dimensions of sexual function and dysfunction. J Sex Med. 2016;13(4):538‐571. [DOI] [PubMed] [Google Scholar]
  • 15. Defeudis G, Gianfrilli D, Di Emidio C, et al. Erectile dysfunction and its management in patients with diabetes mellitus. Rev Endocr Metab Disord. 2015. [DOI] [PubMed] [Google Scholar]
  • 16. Feldman HA, Goldstein I, Hatzichristou DG, Krane RJ, McKinlay JB. Impotence and its medical and psychosocial correlates: results of the Massachusetts Male Aging Study. J Urol. 1994;151(1):54‐61. [DOI] [PubMed] [Google Scholar]
  • 17. Abdelhamed A, Hisasue S, Nada EA, Kassem AM, Abdel‐Kareem M, Horie S. Relation between erectile dysfunction and silent myocardial ischemia in diabetic patients: a multidetector computed tomographic coronary angiographic study. Sex Med. 2016;4(3):e127‐e134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Roger VL, Go AS, Lloyd‐Jones DM, et al. Heart disease and stroke statistics‐2012 update: a report from the American Heart Association. Circulation. 2012;125(1):e2‐e220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Montorsi P, Ravagnani PM, Galli S, et al. Association between erectile dysfunction and coronary artery disease. Role of coronary clinical presentation and extent of coronary vessels involvement: the COBRA trial. Eur Heart J. 2006;27(22):2632‐2639. [DOI] [PubMed] [Google Scholar]
  • 20. Nehra A, Jackson G, Miner M, et al. The Princeton III Consensus recommendations for the management of erectile dysfunction and cardiovascular disease. Mayo Clin Proc. 2012;87(8):766‐778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Vlachopoulos C, Jackson G, Stefanadis C, Montorsi P. Erectile dysfunction in the cardiovascular patient. Eur Heart J. 2013;34(27):2034‐2046. [DOI] [PubMed] [Google Scholar]
  • 22. Piepoli MF, Hoes AW, Agewall S, et al. European guidelines on cardiovascular disease prevention in clinical practice. Rev Esp Cardiol (Engl Ed). 2016;69(10):939. [DOI] [PubMed] [Google Scholar]
  • 23. Zhao B, Hong Z, Wei Y, Yu D, Xu J, Zhang W. Erectile dysfunction predicts cardiovascular events as an independent risk factor: a systematic review and meta‐analysis. J Sex Med. 2019;16(7):1005‐1017. [DOI] [PubMed] [Google Scholar]
  • 24. Goldstein I, Chambers R, Tang W, Stecher V, Hassan T. Real‐world observational results from a database of 48 million men in the United States: relationship of cardiovascular disease, diabetes mellitus and depression with age and erectile dysfunction. Int J Clin Pract. 2018;72(4):e13078. [DOI] [PubMed] [Google Scholar]
  • 25. Foresta C, Ferlin A, Lenzi A, Montorsi P, Italian Study Group on Cardiometabolic A. The great opportunity of the andrological patient: cardiovascular and metabolic risk assessment and prevention. Andrologia. 2017;5(3):408‐413. [DOI] [PubMed] [Google Scholar]
  • 26. Inman BA, Sauver JL, Jacobson DJ, et al. A population‐based, longitudinal study of erectile dysfunction and future coronary artery disease. Mayo Clin Proc. 2009;84(2):108‐113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Conroy RM, Pyorala K, Fitzgerald AP, et al. Estimation of ten‐year risk of fatal cardiovascular disease in Europe: the SCORE project. Eur Heart J. 2003;24(11):987‐1003. [DOI] [PubMed] [Google Scholar]
  • 28. Rastrelli G, Corona G, Mannucci E, Maggi M. Vascular and chronological age in men with erectile dysfunction: a longitudinal study. J Sex Med. 2016;13(2):200‐208. [DOI] [PubMed] [Google Scholar]
  • 29. Rastrelli G, Corona G, Mannucci E, Maggi M. Vascular and chronological age in subjects with erectile dysfunction: a cross‐sectional study. J Sex Med. 2015;12(12):2303‐2312. [DOI] [PubMed] [Google Scholar]
  • 30. Gazzaruso C, Solerte SB, Pujia A, et al. Erectile dysfunction as a predictor of cardiovascular events and death in diabetic patients with angiographically proven asymptomatic coronary artery disease: a potential protective role for statins and 5‐phosphodiesterase inhibitors. J Am Coll Cardiol. 2008;51(21):2040‐2044. [DOI] [PubMed] [Google Scholar]
  • 31. American Diabetes A. 9. Pharmacologic approaches to glycemic treatment: standards of medical care in diabetes‐2020. Diabetes Care. 2020;43(Suppl 1):S98‐S110. [DOI] [PubMed] [Google Scholar]
  • 32. Gazzaruso C, Coppola A, Pujia A, et al. Erectile dysfunction as a predictor of asymptomatic coronary artery disease in elderly men with type 2 diabetes. J Geriatr Cardiol. 2016;13(6):552‐556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Batty GD, Li Q, Czernichow S, et al. Erectile dysfunction and later cardiovascular disease in men with type 2 diabetes: prospective cohort study based on the ADVANCE (Action in Diabetes and Vascular Disease: preterax and Diamicron Modified‐Release Controlled Evaluation) trial. J Am Coll Cardiol. 2010;56(23):1908‐1913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Miner M, Nehra A, Jackson G, et al. All men with vasculogenic erectile dysfunction require a cardiovascular workup. Am J Med. 2014;127(3):174‐182. [DOI] [PubMed] [Google Scholar]
  • 35. Barassi A, Pezzilli R, Morselli‐Labate AM, et al. Evaluation of microalbuminuria in patients with erectile dysfunction. J Sex Med. 2010;7(3):1224‐1228. [DOI] [PubMed] [Google Scholar]
  • 36. Chuang YC, Chung MS, Wang PW, et al. Albuminuria is an independent risk factor of erectile dysfunction in men with type 2 diabetes. J Sex Med. 2012;9(4):1055‐1064. [DOI] [PubMed] [Google Scholar]
  • 37. Baylis C. Arginine, arginine analogs and nitric oxide production in chronic kidney disease. Nat Clin Pract Nephrol. 2006;2(4):209‐220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Costa MR, Ponciano VC, Costa TR, de Oliveira AM, Gomes CP, de Oliveira EC. Prevalence and factors associated with erectile dysfunction in patients with chronic kidney disease on conservative treatment. Int J Impot Res. 2017;29(6):219‐224. [DOI] [PubMed] [Google Scholar]
  • 39. Antonucci M, Palermo G, Recupero SM, et al. Male sexual dysfunction in patients with chronic end‐stage renal insufficiency and in renal transplant recipients. Arch Ital Urol Androl. 2016;87(4):299‐305. [DOI] [PubMed] [Google Scholar]
  • 40. Palmer BF. Outcomes associated with hypogonadism in men with chronic kidney disease. Adv Chronic Kidney Dis. 2004;11(4):342‐347. [PubMed] [Google Scholar]
  • 41. Mambelli E, Barrella M, Facchini MG, et al. The prevalence of peripheral neuropathy in hemodialysis patients. Clin Nephrol. 2012;77(6):468‐475. [DOI] [PubMed] [Google Scholar]
  • 42. Palmer BF, Clegg DJ. Gonadal dysfunction in chronic kidney disease. Rev Endocr Metab Disord. 2017;18(1):117‐130. [DOI] [PubMed] [Google Scholar]
  • 43. Fugl‐Meyer KS, Nilsson M, Hylander B, Lehtihet M. Sexual function and testosterone level in men with conservatively treated chronic kidney disease. Am J Men's Health. 2017;11(4):1069‐1076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Lim VS. Reproductive function in patients with renal insufficiency. Am J Kidney Dis. 1987;9(4):363‐367. [DOI] [PubMed] [Google Scholar]
  • 45. Suzuki E, Nishimatsu H, Oba S, Takahashi M, Homma Y. Chronic kidney disease and erectile dysfunction. World J Nephrol. 2014;3(4):220‐229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Hakim RM, Depner TA, Parker TF, 3rd . Adequacy of hemodialysis. Am J Kidney Dis. 1992;20(2):107‐123. [DOI] [PubMed] [Google Scholar]
  • 47. Sari F, Sari R, Ozdem S, Sarikaya M, Cetinkaya R. Serum prolactin and macroprolactin levels in diabetic nephropathy. Clin Nephrol. 2012;78(1):33‐39. [DOI] [PubMed] [Google Scholar]
  • 48. Massry SG, Goldstein DA, Procci WR, Kletzky OA. Impotence in patients with uremia: a possible role for parathyroid hormone. Nephron. 1977;19(6):305‐310. [DOI] [PubMed] [Google Scholar]
  • 49. Said G. Uremic neuropathy. Handb Clin Neurol. 2013;115:607‐612. [DOI] [PubMed] [Google Scholar]
  • 50. Cellek S, Cameron NE, Cotter MA, Muneer A. Pathophysiology of diabetic erectile dysfunction: potential contribution of vasa nervorum and advanced glycation endproducts. Int J Impot Res. 2013;25(1):1‐6. [DOI] [PubMed] [Google Scholar]
  • 51. Fowler CJ, Ali Z, Kirby RS, Pryor JP. The value of testing for unmyelinated fibre, sensory neuropathy in diabetic impotence. Br J Urol. 1988;61(1):63‐67. [DOI] [PubMed] [Google Scholar]
  • 52. Herman WH, Pop‐Busui R, Braffett BH, et al. Use of the Michigan neuropathy screening instrument as a measure of distal symmetrical peripheral neuropathy in type 1 diabetes: results from the diabetes control and complications trial/epidemiology of diabetes interventions and complications. Diabet Med. 2012;29(7):937‐944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Bleustein CB, Arezzo JC, Eckholdt H, Melman A. The neuropathy of erectile dysfunction. Int J Impot Res. 2002;14(6):433‐439. [DOI] [PubMed] [Google Scholar]
  • 54. Hurt KJ, Musicki B, Palese MA, et al. Akt‐dependent phosphorylation of endothelial nitric‐oxide synthase mediates penile erection. Proc Natl Acad Sci U. S. A. 2002;99(6):4061‐4066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Ferrannini E, Cushman WC. Diabetes and hypertension: the bad companions. Lancet. 2012;380(9841):601‐610. [DOI] [PubMed] [Google Scholar]
  • 56. The sixth report of the Joint National Committee on prevention, detection, evaluation, and treatment of high blood pressure. Arch Intern Med. 1997;157(21):2413‐2446. [DOI] [PubMed] [Google Scholar]
  • 57. World Health Organization. World Health Organization‐International Society of Hypertension Guidelines for the Management of Hypertension: guidelines subcommittee. J Hypertens. 1999;17(2):151‐183. [PubMed] [Google Scholar]
  • 58. Penno G, Solini A, Bonora E, et al. HbA1c variability as an independent correlate of nephropathy, but not retinopathy, in patients with type 2 diabetes: the Renal Insufficiency and Cardiovascular Events (RIACE) Italian multicenter study. Diabetes Care. 2013;36(8):2301‐2310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Bruno G, De Micheli A, Frontoni S, Monge L. Societa Italiana di Diabetologia‐Associazione Medici Diabetologi working group on the standards of care for D. Highlights from “Italian standards of care for diabetes mellitus 2009‐2010”. Nutr Metab Cardiovasc Dis. 2011;21(4):302‐314. [DOI] [PubMed] [Google Scholar]
  • 60. Jensen J, Lendorf A, Stimpel H, Frost J, Ibsen H, Rosenkilde P. The prevalence and etiology of impotence in 101 male hypertensive outpatients. Am J Hypertens. 1999;12(3):271‐275. [DOI] [PubMed] [Google Scholar]
  • 61. Viigimaa M, Vlachopoulos C, Lazaridis A, Doumas M. Management of erectile dysfunction in hypertension: tips and tricks. World J Cardiol. 2014;6(9):908‐915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Burchardt M, Burchardt T, Baer L, et al. Hypertension is associated with severe erectile dysfunction. J Urol. 2000;164(4):1188‐1191. [PubMed] [Google Scholar]
  • 63. Giuliano FA, Leriche A, Jaudinot EO, de Gendre AS. Prevalence of erectile dysfunction among 7689 patients with diabetes or hypertension, or both. Urology. 2004;64(6):1196‐1201. [DOI] [PubMed] [Google Scholar]
  • 64. Doumas M, Tsakiris A, Douma S, et al. Factors affecting the increased prevalence of erectile dysfunction in Greek hypertensive compared with normotensive subjects. J Androl. 2006;27(3):469‐477. [DOI] [PubMed] [Google Scholar]
  • 65. Johannes CB, Araujo AB, Feldman HA, Derby CA, Kleinman KP, McKinlay JB. Incidence of erectile dysfunction in men 40 to 69 years old: longitudinal results from the Massachusetts male aging study. J Urol. 2000;163(2):460‐463. [PubMed] [Google Scholar]
  • 66. Heikkila A, Kaipia A, Venermo M, Kautiainen H, Korhonen P. Relationship of blood pressure and erectile dysfunction in men without previously diagnosed hypertension. J Sex Med. 2017;14(11):1336‐1341. [DOI] [PubMed] [Google Scholar]
  • 67. Foy CG, Newman JC, Berlowitz DR, et al. Blood pressure, sexual activity, and erectile function in hypertensive men: baseline findings from the systolic blood pressure intervention trial (SPRINT). J Sex Med. 2019;16(2):235‐247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Sarma AV, Hotaling JM, de Boer IH, et al. Blood pressure, antihypertensive medication use, and risk of erectile dysfunction in men with type I diabetes. J Hypertens. 2019;37(5):1070‐1076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Kalka D, Gebala J, Rusiecki L, et al. Relation of postexercise reduction of arterial blood pressure and erectile dysfunction in patients with coronary heart disease. Am J Cardiol. 2018;122(2):229‐234. [DOI] [PubMed] [Google Scholar]
  • 70. Korhonen PE, Ettala O, Kautiainen H, Kantola I. Factors modifying the effect of blood pressure on erectile function. J Hypertens. 2015;33(5):975‐980. [DOI] [PubMed] [Google Scholar]
  • 71. Foy CG, Newman JC, Russell GB, et al. Effect of intensive vs standard blood pressure treatment upon erectile function in hypertensive men: findings from the systolic blood pressure intervention trial. J Sex Med. 2020;17(2):238‐248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Joseph P, Lonn E, Bosch J, et al. Long‐term effects of statins, blood pressure‐lowering, and both on erectile function in persons at intermediate risk for cardiovascular disease: a substudy of the heart outcomes prevention evaluation‐3 (HOPE‐3) randomized controlled trial. Can J Cardiol. 2018;34(1):38‐44. [DOI] [PubMed] [Google Scholar]
  • 73. Cordero A, Bertomeu‐Martinez V, Mazon P, et al. Erectile dysfunction in high‐risk hypertensive patients treated with beta‐blockade agents. Cardiovasc Ther. 2010;28(1):15‐22. [DOI] [PubMed] [Google Scholar]
  • 74. Brixius K, Middeke M, Lichtenthal A, Jahn E, Schwinger RH. Nitric oxide, erectile dysfunction and beta‐blocker treatment (MR NOED study): benefit of nebivolol versus metoprolol in hypertensive men. Clin Exp Pharmacol Physiol. 2007;34(4):327‐331. [DOI] [PubMed] [Google Scholar]
  • 75. Gur O, Gurkan S, Yumun G, Turker P. The comparison of the effects of nebivolol and metoprolol on erectile dysfunction in the cases with coronary artery bypass surgery. Ann Thorac Cardiovasc Surg. 2017;23(2):91‐95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Sharp RP, Gales BJ. Nebivolol versus other beta blockers in patients with hypertension and erectile dysfunction. Ther Adv Urol. 2017;9(2):59‐63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Chang SW, Fine R, Siegel D, Chesney M, Black D, Hulley SB. The impact of diuretic therapy on reported sexual function. Arch Intern Med. 1991;151(12):2402‐2408. [PubMed] [Google Scholar]
  • 78. Sica DA. Diuretic‐related side effects: development and treatment. J Clin Hypertens. 2004;6(9):532‐540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Struthers A, Krum H, Williams GH. A comparison of the aldosterone‐blocking agents eplerenone and spironolactone. Clin Cardiol. 2008;31(4):153‐158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Speel TG, Kiemeney LA, Thien T, Smits P, Meuleman EJ. Long‐term effect of inhibition of the angiotensin‐converting enzyme (ACE) on cavernosal perfusion in men with atherosclerotic erectile dysfunction: a pilot study. J Sex Med. 2005;2(2):207‐212. [DOI] [PubMed] [Google Scholar]
  • 81. Chen Y, Cui S, Lin H, et al. Losartan improves erectile dysfunction in diabetic patients: a clinical trial. Int J Impot Res. 2012;24(6):217‐220. [DOI] [PubMed] [Google Scholar]
  • 82. Bohm M, Baumhakel M, Teo K, et al. Erectile dysfunction predicts cardiovascular events in high‐risk patients receiving telmisartan, ramipril, or both: the ONgoing Telmisartan Alone and in combination with Ramipril Global Endpoint Trial/Telmisartan Randomized AssessmeNt Study in ACE iNtolerant subjects with cardiovascular Disease (ONTARGET/TRANSCEND) Trials. Circulation. 2010;121(12):1439‐1446. [DOI] [PubMed] [Google Scholar]
  • 83. Shiri R, Koskimaki J, Hakkinen J, Auvinen A, Tammela TL, Hakama M. Cardiovascular drug use and the incidence of erectile dysfunction. Int J Impot Res. 2007;19(2):208‐212. [DOI] [PubMed] [Google Scholar]
  • 84. Thorve VS, Kshirsagar AD, Vyawahare NS, Joshi VS, Ingale KG, Mohite RJ. Diabetes‐induced erectile dysfunction: epidemiology, pathophysiology and management. J Diabet Complicat. 2011;25(2):129‐136. [DOI] [PubMed] [Google Scholar]
  • 85. Ishikawa K, Sasaki H, Tanifuji S, et al. Lipid abnormality, current diabetes and age affect erectile hardness: an analysis of data from complete medical checkups carried out at a single hospital. Int J Urol. 2020;27(4):339‐343. [DOI] [PubMed] [Google Scholar]
  • 86. Azad AK, Setunge S, Selim S, et al. Dyslipidaemia as a risk factor for erectile dysfunction in type 2 diabetes mellitus patients. Diabetes Metab Syndr. 2019;13(1):748‐753. [DOI] [PubMed] [Google Scholar]
  • 87. Belba A, Cortelazzo A, Andrea G, et al. Erectile dysfunction and diabetes: association with the impairment of lipid metabolism and oxidative stress. Clin Biochem. 2016;49(1‐2):70‐78. [DOI] [PubMed] [Google Scholar]
  • 88. Corona G, Cipriani S, Rastrelli G, Sforza A, Mannucci E, Maggi M. High triglycerides predicts arteriogenic erectile dysfunction and major adverse cardiovascular events in subjects with sexual dysfunction. J Sex Med. 2016;13(9):1347‐1358. [DOI] [PubMed] [Google Scholar]
  • 89. Ponholzer A, Temml C, Rauchenwald M, Madersbacher S. Vascular risk factors and erectile dysfunction in a cohort of healthy men. Int J Impot Res. 2006;18(5):489‐493. [DOI] [PubMed] [Google Scholar]
  • 90. Corona G, Giagulli VA, Maseroli E, et al. Testosterone supplementation and body composition: results from a meta‐analysis of observational studies. J Endocrinol Invest. 2016;39(9):967‐981. [DOI] [PubMed] [Google Scholar]
  • 91. Zhang KS, Zhao MJ, An Q, et al. Effects of testosterone supplementation therapy on lipid metabolism in hypogonadal men with T2DM: a meta‐analysis of randomized controlled trials. Andrologia. 2018;6(1):37‐46. [DOI] [PubMed] [Google Scholar]
  • 92. Corona G, Boddi V, Balercia G, et al. The effect of statin therapy on testosterone levels in subjects consulting for erectile dysfunction. J Sex Med. 2010;7(4 Pt 1):1547‐1556. [DOI] [PubMed] [Google Scholar]
  • 93. Kostis JB, Dobrzynski JM. The effect of statins on erectile dysfunction: a meta‐analysis of randomized trials. J Sex Med. 2014;11(7):1626‐1635. [DOI] [PubMed] [Google Scholar]
  • 94. Cui Y, Zong H, Yan H, Zhang Y. The effect of statins on erectile dysfunction: a systematic review and meta‐analysis. J Sex Med. 2014;11(6):1367‐1375. [DOI] [PubMed] [Google Scholar]
  • 95. Elgendy AY, Elgendy IY, Mahmoud AN, et al. Statin use in men and new onset of erectile dysfunction: a systematic review and meta‐analysis. Am J Med. 2018;131(4):387‐394. [DOI] [PubMed] [Google Scholar]
  • 96. Leitner DR, Fruhbeck G, Yumuk V, et al. Obesity and type 2 diabetes: two diseases with a need for combined treatment strategies—EASO can lead the way. Obes Facts. 2017;10(5):483‐492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Prinz N, Schwandt A, Becker M, et al. Trajectories of body mass index from childhood to young adulthood among patients with type 1 diabetes‐A longitudinal group‐based modeling approach based on the DPV registry. J Pediatr. 2018;201:78‐85. [DOI] [PubMed] [Google Scholar]
  • 98. Maiorino MI, Bellastella G, Giugliano D, Esposito K. From inflammation to sexual dysfunctions: a journey through diabetes, obesity, and metabolic syndrome. J Endocrinol Invest. 2018;41(11):1249‐1258. [DOI] [PubMed] [Google Scholar]
  • 99. Han TS, Tajar A, O'Neill TW, et al. Impaired quality of life and sexual function in overweight and obese men: the European Male Ageing Study. Eur J Endocrinol. 2011;164(6):1003‐1011. [DOI] [PubMed] [Google Scholar]
  • 100. Andersson DP, Ekstrom U, Lehtihet M. Rigiscan evaluation of men with diabetes mellitus and erectile dysfunction and correlation with diabetes duration, age, BMI, lipids and HbA1c. PLoS One. 2015;10(7):e0133121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Maseroli E, Corona G, Rastrelli G, et al. Prevalence of endocrine and metabolic disorders in subjects with erectile dysfunction: a comparative study. J Sex Med. 2015;12(4):956‐965. [DOI] [PubMed] [Google Scholar]
  • 102. Chaudhary RK, Shamsi BH, Tan T, Chen HM, Xing JP. Study of the relationship between male erectile dysfunction and type 2 diabetes mellitus/metabolic syndrome and its components. J Int Med Res. 2016;44(3):735‐741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Lotti F, Rastrelli G, Maseroli E, et al. Impact of metabolically healthy obesity in patients with andrological problems. J Sex Med. 2019;16(6):821‐832. [DOI] [PubMed] [Google Scholar]
  • 104. Dimopoulou C, Goulis DG, Corona G, Maggi M. The complex association between metabolic syndrome and male hypogonadism. Metabolism. 2018;86:61‐68. [DOI] [PubMed] [Google Scholar]
  • 105. Corona G, Rastrelli G, Filippi S, Vignozzi L, Mannucci E, Maggi M. Erectile dysfunction and central obesity: an Italian perspective. Asian J Androl. 2014;16(4):581‐591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Bhasin S, Brito JP, Cunningham GR, et al. Testosterone therapy in men with hypogonadism: an endocrine society clinical practice guideline. J Clin Endocrinol Metab. 2018;103(5):1715‐1744. [DOI] [PubMed] [Google Scholar]
  • 107. Pasquali R, Casanueva F, Haluzik M, et al. European society of endocrinology clinical practice guideline: endocrine work‐up in obesity. Eur J Endocrinol. 2020;182(1):G1‐G32. [DOI] [PubMed] [Google Scholar]
  • 108. Khoo J, Piantadosi C, Duncan R, et al. Comparing effects of a low‐energy diet and a high‐protein low‐fat diet on sexual and endothelial function, urinary tract symptoms, and inflammation in obese diabetic men. J Sex Med. 2011;8(10):2868‐2875. [DOI] [PubMed] [Google Scholar]
  • 109. Giagulli VA, Castellana M, Carbone MD, et al. Weight loss more than glycemic control may improve testosterone in obese type 2 diabetes mellitus men with hypogonadism. Andrologia. 2020. [DOI] [PubMed] [Google Scholar]
  • 110. Schauer PR, Kashyap SR, Wolski K, et al. Bariatric surgery versus intensive medical therapy in obese patients with diabetes. N Engl J Med. 2012;366(17):1567‐1576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Reis LO, Favaro WJ, Barreiro GC, et al. Erectile dysfunction and hormonal imbalance in morbidly obese male is reversed after gastric bypass surgery: a prospective randomized controlled trial. Int J Androl. 2010;33(5):736‐744. [DOI] [PubMed] [Google Scholar]
  • 112. Aversa A, Bruzziches R, Francomano D, et al. Effects of testosterone undecanoate on cardiovascular risk factors and atherosclerosis in middle‐aged men with late‐onset hypogonadism and metabolic syndrome: results from a 24‐month, randomized, double‐blind, placebo‐controlled study. J Sex Med. 2010;7(10):3495‐3503. [DOI] [PubMed] [Google Scholar]
  • 113. Magnussen LV, Glintborg D, Hermann P, Hougaard DM, Hojlund K, Andersen M. Effect of testosterone on insulin sensitivity, oxidative metabolism and body composition in aging men with type 2 diabetes on metformin monotherapy. Diabetes Obes Metab. 2016;18(10):980‐989. [DOI] [PubMed] [Google Scholar]
  • 114. Hackett G, Cole N, Mulay A, Strange RC, Ramachandran S. Long‐term testosterone therapy in type 2 diabetes is associated with decreasing waist circumference and improving erectile function. World J Mens Health. 2020;38(1):68‐77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Canguven O, Talib RA, El Ansari W, Yassin DJ, Salman M, Al‐Ansari A. Testosterone therapy has positive effects on anthropometric measures, metabolic syndrome components (obesity, lipid profile, diabetes mellitus control), blood indices, liver enzymes, and prostate health indicators in elderly hypogonadal men. Andrologia. 2017;49(10). [DOI] [PubMed] [Google Scholar]
  • 116. Jones TH, Arver S, Behre HM, et al. Testosterone replacement in hypogonadal men with type 2 diabetes and/or metabolic syndrome (the TIMES2 study). Diabetes Care. 2011;34(4):828‐837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Kalinchenko SY, Tishova YA, Mskhalaya GJ, Gooren LJ, Giltay EJ, Saad F. Effects of testosterone supplementation on markers of the metabolic syndrome and inflammation in hypogonadal men with the metabolic syndrome: the double‐blinded placebo‐controlled Moscow study. Clin Endocrinol (Oxf). 2010;73(5):602‐612. [DOI] [PubMed] [Google Scholar]
  • 118. Tomar R, Dhindsa S, Chaudhuri A, Mohanty P, Garg R, Dandona P. Contrasting testosterone concentrations in type 1 and type 2 diabetes. Diabetes Care. 2006;29(5):1120‐1122. [DOI] [PubMed] [Google Scholar]
  • 119. Dhindsa S, Miller MG, McWhirter CL, et al. Testosterone concentrations in diabetic and nondiabetic obese men. Diabetes Care. 2010;33(6):1186‐1192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Kapoor D, Goodwin E, Channer KS, Jones TH. Testosterone replacement therapy improves insulin resistance, glycaemic control, visceral adiposity and hypercholesterolaemia in hypogonadal men with type 2 diabetes. Eur J Endocrinol. 2006;154(6):899‐906. [DOI] [PubMed] [Google Scholar]
  • 121. Corona G, Monami M, Rastrelli G, et al. Type 2 diabetes mellitus and testosterone: a meta‐analysis study. Int J Androl. 2011;34(6 Pt 1):528‐540. [DOI] [PubMed] [Google Scholar]
  • 122. Ding EL, Song Y, Malik VS, Liu S. Sex differences of endogenous sex hormones and risk of type 2 diabetes: a systematic review and meta‐analysis. J Am Med Assoc. 2006;295(11):1288‐1299. [DOI] [PubMed] [Google Scholar]
  • 123. Dandona P, Dhindsa S. Update: hypogonadotropic hypogonadism in type 2 diabetes and obesity. J Clin Endocrinol Metab. 2011;96(9):2643‐2651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. El Saghier EO, Shebl SE, Fawzy OA, Eltayeb IM, Bekhet LM, Gharib A. Androgen deficiency and erectile dysfunction in patients with type 2 diabetes. Clin Med Insights Endocrinol Diabetes. 2015;8:55‐62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Muraleedharan V, Marsh H, Kapoor D, Channer KS, Jones TH. Testosterone deficiency is associated with increased risk of mortality and testosterone replacement improves survival in men with type 2 diabetes. Eur J Endocrinol. 2013;169(6):725‐733. [DOI] [PubMed] [Google Scholar]
  • 126. Bruning JC, Gautam D, Burks DJ, et al. Role of brain insulin receptor in control of body weight and reproduction. Science. 2000;289(5487):2122‐2125. [DOI] [PubMed] [Google Scholar]
  • 127. Corona G, Rastrelli G, Monami M, et al. Body weight loss reverts obesity‐associated hypogonadotropic hypogonadism: a systematic review and meta‐analysis. Eur J Endocrinol. 2013;168(6):829‐843. [DOI] [PubMed] [Google Scholar]
  • 128. Wallace IR, McKinley MC, Bell PM, Hunter SJ. Sex hormone binding globulin and insulin resistance. Clin Endocrinol (Oxf). 2013;78(3):321‐329. [DOI] [PubMed] [Google Scholar]
  • 129. Defeudis G, Mazzilli R, Gianfrilli D, Lenzi A, Isidori AM. The CATCH checklist to investigate adult‐onset hypogonadism. Andrologia. 2018;6(5):665‐679. [DOI] [PubMed] [Google Scholar]
  • 130. Dhindsa S, Ghanim H, Batra M, et al. Insulin resistance and inflammation in hypogonadotropic hypogonadism and their reduction after testosterone replacement in men with type 2 diabetes. Diabetes Care. 2016;39(1):82‐91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Yeap BB, Chubb SA, Hyde Z, et al. Lower serum testosterone is independently associated with insulin resistance in non‐diabetic older men: the Health in Men Study. Eur J Endocrinol. 2009;161(4):591‐598. [DOI] [PubMed] [Google Scholar]
  • 132. Hackett G, Cole N, Saghir A, Jones P, Strange RC, Ramachandran S. Testosterone replacement therapy: improved sexual desire and erectile function in men with type 2 diabetes following a 30‐week randomized placebo‐controlled study. Andrologia. 2017;5(5):905‐913. [DOI] [PubMed] [Google Scholar]
  • 133. Grossmann M, Hoermann R, Wittert G, Yeap BB. Effects of testosterone treatment on glucose metabolism and symptoms in men with type 2 diabetes and the metabolic syndrome: a systematic review and meta‐analysis of randomized controlled clinical trials. Clin Endocrinol (Oxf). 2015;83(3):344‐351. [DOI] [PubMed] [Google Scholar]
  • 134. Gianatti EJ, Dupuis P, Hoermann R, et al. Effect of testosterone treatment on glucose metabolism in men with type 2 diabetes: a randomized controlled trial. Diabetes Care. 2014;37(8):2098‐2107. [DOI] [PubMed] [Google Scholar]
  • 135. Sesti F, Pofi R, Minnetti M, Tenuta M, Gianfrilli D, Isidori AM. Late‐onset hypogonadism: reductio ad absurdum of the cardiovascular risk‐benefit of testosterone replacement therapy. Andrologia. 2020;8(6):1614‐1627. [DOI] [PubMed] [Google Scholar]
  • 136. Seftel AD, Kathrins M, Niederberger C. Critical update of the 2010 endocrine society clinical practice guidelines for male hypogonadism: a systematic analysis. Mayo Clin Proc. 2015;90(8):1104‐1115. [DOI] [PubMed] [Google Scholar]
  • 137. Corona G, Rastrelli G, Di Pasquale G, Sforza A, Mannucci E, Maggi M. Testosterone and cardiovascular risk: meta‐analysis of interventional studies. J Sex Med. 2018;15(6):820‐838. [DOI] [PubMed] [Google Scholar]
  • 138. Dimopoulou C, Ceausu I, Depypere H, et al. EMAS position statement: testosterone replacement therapy in the aging male. Maturitas. 2016;84:94‐99. [DOI] [PubMed] [Google Scholar]
  • 139. Radholm K, Wu JH, Wong MG, et al. Effects of sodium‐glucose cotransporter‐2 inhibitors on cardiovascular disease, death and safety outcomes in type 2 diabetes—a systematic review. Diabetes Res Clin Pract. 2018;140:118‐128. [DOI] [PubMed] [Google Scholar]
  • 140. Puckrin R, Saltiel MP, Reynier P, Azoulay L, Yu OHY, Filion KB. SGLT‐2 inhibitors and the risk of infections: a systematic review and meta‐analysis of randomized controlled trials. Acta Diabetol. 2018;55(5):503‐514. [DOI] [PubMed] [Google Scholar]
  • 141. Renko M, Tapanainen P, Tossavainen P, Pokka T, Uhari M. Meta‐analysis of the significance of asymptomatic bacteriuria in diabetes. Diabetes Care. 2011;34(1):230‐235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. La Vignera S, Condorelli RA, Cannarella R, et al. Urogenital infections in patients with diabetes mellitus: beyond the conventional aspects. Int J Immunopathol Pharmacol. 2019;33:2058738419866582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. He K, Hu Y, Shi JC, Zhu YQ, Mao XM. Prevalence, risk factors and microorganisms of urinary tract infections in patients with type 2 diabetes mellitus: a retrospective study in China. Ther Clin Risk Manag. 2018;14:403‐408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Hirji I, Andersson SW, Guo Z, Hammar N, Gomez‐Caminero A. Incidence of genital infection among patients with type 2 diabetes in the UK General Practice Research Database. J Diabet Complicat. 2012;26(6):501‐505. [DOI] [PubMed] [Google Scholar]
  • 145. La Vignera S, Condorelli RA, Di Mauro M, D'Agata R, Vicari E, Calogero AE. Seminal vesicles and diabetic neuropathy: ultrasound evaluation. J Androl. 2011;32(5):478‐483. [DOI] [PubMed] [Google Scholar]
  • 146. Bang WJ, Lee JY, Koo KC, Hah YS, Lee DH, Cho KS. Is type‐2 diabetes mellitus associated with overactive bladder symptoms in men with lower urinary tract symptoms? Urology. 2014;84(3):670‐674. [DOI] [PubMed] [Google Scholar]
  • 147. Wessells H, Braffett BH, Holt SK, et al. Burden of urological complications in men and women with long‐standing type 1 diabetes in the diabetes control and complications trial/epidemiology of diabetes interventions and complications cohort. Diabetes Care. 2018;41(10):2170‐2177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Berger AP, Deibl M, Leonhartsberger N, et al. Vascular damage as a risk factor for benign prostatic hyperplasia and erectile dysfunction. BJU Int. 2005;96(7):1073‐1078. [DOI] [PubMed] [Google Scholar]
  • 149. Vikram A, Jena G, Ramarao P. Insulin‐resistance and benign prostatic hyperplasia: the connection. Eur J Pharmacol. 2010;641(2‐3):75‐81. [DOI] [PubMed] [Google Scholar]
  • 150. Chiu AF, Huang MH, Wang CC, Kuo HC. Higher glycosylated hemoglobin levels increase the risk of overactive bladder syndrome in patients with type 2 diabetes mellitus. Int J Urol. 2012;19(11):995‐1001. [DOI] [PubMed] [Google Scholar]
  • 151. Kebapci N, Yenilmez A, Efe B, Entok E, Demirustu C. Bladder dysfunction in type 2 diabetic patients. Neurourol Urodyn. 2007;26(6):814‐819. [DOI] [PubMed] [Google Scholar]
  • 152. Yamaguchi C, Sakakibara R, Uchiyama T, et al. Overactive bladder in diabetes: a peripheral or central mechanism? Neurourol Urodyn. 2007;26(6):807‐813. [DOI] [PubMed] [Google Scholar]
  • 153. Pop‐Busui R, Hotaling J, Braffett BH, et al. Cardiovascular autonomic neuropathy, erectile dysfunction and lower urinary tract symptoms in men with type 1 diabetes: findings from the DCCT/EDIC. J Urol. 2015;193(6):2045‐2051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Ralph D, Gonzalez‐Cadavid N, Mirone V, et al. The management of Peyronie's disease: evidence‐based 2010 guidelines. J Sex Med. 2010;7(7):2359‐2374. [DOI] [PubMed] [Google Scholar]
  • 155. Romero FR, Romero AW, Almeida RM, Oliveira FC, Jr , Filho RT, Jr . Prevalence and risk factors for penile lesions/anomalies in a cohort of Brazilian men >/= 40 years of age. Int Braz J Urol. 2013;39(1):55‐62. [DOI] [PubMed] [Google Scholar]
  • 156. Segundo A, Glina S. Prevalence, risk factors, and erectile dysfunction associated with peyronie's disease among men seeking urological care. Sex Med. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Jacobson AM, Braffett BH, Cleary PA, et al. Relationship of urologic complications with health‐related quality of life and perceived value of health in men and women with type 1 diabetes: the Diabetes Control and Complications Trial/Epidemiology of Interventions and Complications (DCCT/EDIC) cohort. Diabetes Care. 2015;38(10):1904‐1912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Gomes CM, Averbeck MA, Koyama M, Soler R. Association among lower urinary tract symptoms, erectile function, and sexual satisfaction: results from the Brazil LUTS study. Sex Med. 2020;8(1):45‐56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Irwin DE, Milsom I, Reilly K, et al. Overactive bladder is associated with erectile dysfunction and reduced sexual quality of life in men. J Sex Med. 2008;5(12):2904‐2910. [DOI] [PubMed] [Google Scholar]
  • 160. West SD, Nicoll DJ, Stradling JR. Prevalence of obstructive sleep apnoea in men with type 2 diabetes. Thorax. 2006;61(11):945‐950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161. Foster GD, Sanders MH, Millman R, et al. Obstructive sleep apnea among obese patients with type 2 diabetes. Diabetes Care. 2009;32(6):1017‐1019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162. Tahrani AA. Obstructive sleep apnoea in diabetes: does it matter? Diab Vasc Dis Res. 2017;14(5):454‐462. [DOI] [PubMed] [Google Scholar]
  • 163. Stannek T, Hurny C, Schoch OD, Bucher T, Munzer T. Factors affecting self‐reported sexuality in men with obstructive sleep apnea syndrome. J Sex Med. 2009;6(12):3415‐3424. [DOI] [PubMed] [Google Scholar]
  • 164. Margel D, Cohen M, Livne PM, Pillar G. Severe, but not mild, obstructive sleep apnea syndrome is associated with erectile dysfunction. Urology. 2004;63(3):545‐549. [DOI] [PubMed] [Google Scholar]
  • 165. Shin HW, Rha YC, Han DH, et al. Erectile dysfunction and disease‐specific quality of life in patients with obstructive sleep apnea. Int J Impot Res. 2008;20(6):549‐553. [DOI] [PubMed] [Google Scholar]
  • 166. Taskin U, Yigit O, Acioglu E, Aricigil M, Toktas G, Guzelhan Y. Erectile dysfunction in severe sleep apnea patients and response to CPAP. Int J Impot Res. 2010;22(2):134‐139. [DOI] [PubMed] [Google Scholar]
  • 167. Margel D, Tal R, Livne PM, Pillar G. Predictors of erectile function improvement in obstructive sleep apnea patients with long‐term CPAP treatment. Int J Impot Res. 2005;17(2):186‐190. [DOI] [PubMed] [Google Scholar]
  • 168. Khafagy AH, Khafagy AH. Treatment of obstructive sleep apnoea as a therapeutic modality for associated erectile dysfunction. Int J Clin Pract. 2012;66(12):1204‐1208. [DOI] [PubMed] [Google Scholar]
  • 169. Irer B, Celikhisar A, Celikhisar H, Bozkurt O, Demir O. Evaluation of sexual dysfunction, lower urinary tract symptoms and quality of life in men with obstructive sleep apnea syndrome and the efficacy of continuous positive airway pressure therapy. Urology. 2018;121:86‐92. [DOI] [PubMed] [Google Scholar]
  • 170. Perimenis P, Karkoulias K, Markou S, et al. Erectile dysfunction in men with obstructive sleep apnea syndrome: a randomized study of the efficacy of sildenafil and continuous positive airway pressure. Int J Impot Res. 2004;16(3):256‐260. [DOI] [PubMed] [Google Scholar]
  • 171. Pastore AL, Palleschi G, Ripoli A, et al. Severe obstructive sleep apnoea syndrome and erectile dysfunction: a prospective randomised study to compare sildenafil vs. nasal continuous positive airway pressure. Int J Clin Pract. 2014;68(8):995‐1000. [DOI] [PubMed] [Google Scholar]
  • 172. Luboshitzky R, Aviv A, Hefetz A, et al. Decreased pituitary‐gonadal secretion in men with obstructive sleep apnea. J Clin Endocrinol Metab. 2002;87(7):3394‐3398. [DOI] [PubMed] [Google Scholar]
  • 173. Chen S, Peng D, Xu X, et al. Assessment of erectile dysfunction and associated psychological distress in Chinese men with type 2 diabetes mellitus. Int J Impot Res. 2017;29(5):210‐214. [DOI] [PubMed] [Google Scholar]
  • 174. Furukawa S, Sakai T, Niiya T, et al. Depressive symptoms and prevalence of erectile dysfunction in Japanese patients with type 2 diabetes mellitus: the Dogo Study. Int J Impot Res. 2017;29(2):57‐60. [DOI] [PubMed] [Google Scholar]
  • 175. Atlantis E, Sullivan T. Bidirectional association between depression and sexual dysfunction: a systematic review and meta‐analysis. J Sex Med. 2012;9(6):1497‐1507. [DOI] [PubMed] [Google Scholar]
  • 176. Anderson RJ, Freedland KE, Clouse RE, Lustman PJ. The prevalence of comorbid depression in adults with diabetes: a meta‐analysis. Diabetes Care. 2001;24(6):1069‐1078. [DOI] [PubMed] [Google Scholar]
  • 177. Huang IS, Mazur DJ, Kahn BE, et al. Risk factors for hypogonadism in young men with erectile dysfunction. J Chin Med Assoc. 2019;82(6):477‐481. [DOI] [PubMed] [Google Scholar]
  • 178. Montejo AL, Prieto N, de Alarcon R, Casado‐Espada N, de la Iglesia J, Montejo L. Management strategies for antidepressant‐related sexual dysfunction: a clinical approach. J Clin Med. 2019;8(10). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179. Gonzalez JS, Tanenbaum ML, Commissariat PV. Psychosocial factors in medication adherence and diabetes self‐management: implications for research and practice. Am Psychol. 2016;71(7):539‐551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180. Mulhall JP, Giraldi A, Hackett G, et al. The 2018 revision to the process of care model for management of erectile dysfunction. J Sex Med. 2018;15(10):1434‐1445. [DOI] [PubMed] [Google Scholar]
  • 181. Matsui H, Sopko NA, Hannan JL, Bivalacqua TJ. Pathophysiology of erectile dysfunction. Curr Drug Targets. 2015;16(5):411‐419. [DOI] [PubMed] [Google Scholar]
  • 182. Sopko NA, Hannan JL, Bivalacqua TJ. Understanding and targeting the Rho kinase pathway in erectile dysfunction. Nat Rev Urol. 2014;11(11):622‐628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183. Zewdie KA, Ayza MA, Tesfaye BA, Wondafrash DZ, Berhe DF. A systematic review on Rho‐kinase as a potential therapeutic target for the treatment of erectile dysfunction. Res Rep Urol. 2020;12:261‐272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184. Jin LM. Angiotensin II signaling and its implication in erectile dysfunction. J Sex Med. 2009;6(Suppl 3):302‐310. [DOI] [PubMed] [Google Scholar]
  • 185. Fraga‐Silva RA, Montecucco F, Mach F, Santos RA, Stergiopulos N. Pathophysiological role of the renin‐angiotensin system on erectile dysfunction. Eur J Clin Invest. 2013;43(9):978‐985. [DOI] [PubMed] [Google Scholar]
  • 186. Carneiro FS, Webb RC, Tostes RC. Emerging role for TNF‐alpha in erectile dysfunction. J Sex Med. 2010;7(12):3823‐3834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187. Ritchie R, Sullivan M. Endothelins & erectile dysfunction. Pharmacol Res. 2011;63(6):496‐501. [DOI] [PubMed] [Google Scholar]
  • 188. Patel JP, Lee EH, Mena CI, Walker CN. Effects of metformin on endothelial health and erectile dysfunction. Transl Androl Urol. 2017;6(3):556‐565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189. Dean RC, Lue TF. Physiology of penile erection and pathophysiology of erectile dysfunction. Urol Clin North Am. 2005;32(4):379‐395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190. Pivonello R, Menafra D, Riccio E, et al. Metabolic disorders and male hypogonadotropic hypogonadism. Front Endocrinol (Lausanne). 2019;10:345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191. Durand MJ, Gutterman DD. Diversity in mechanisms of endothelium‐dependent vasodilation in health and disease. Microcirculation. 2013;20(3):239‐247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192. Lin KY, Ito A, Asagami T, et al. Impaired nitric oxide synthase pathway in diabetes mellitus: role of asymmetric dimethylarginine and dimethylarginine dimethylaminohydrolase. Circulation. 2002;106(8):987‐992. [DOI] [PubMed] [Google Scholar]
  • 193. Vitale C, Mercuro G, Cornoldi A, Fini M, Volterrani M, Rosano GM. Metformin improves endothelial function in patients with metabolic syndrome. J Intern Med. 2005;258(3):250‐256. [DOI] [PubMed] [Google Scholar]
  • 194. Mather KJ, Verma S, Anderson TJ. Improved endothelial function with metformin in type 2 diabetes mellitus. J Am Coll Cardiol. 2001;37(5):1344‐1350. [DOI] [PubMed] [Google Scholar]
  • 195. Kaya C, Uslu Z, Karaman I. Is endothelial function impaired in erectile dysfunction patients? Int J Impot Res. 2006;18(1):55‐60. [DOI] [PubMed] [Google Scholar]
  • 196. Grassi G. Role of the sympathetic nervous system in human hypertension. J Hypertens. 1998;16(12 Pt 2):1979‐1987. [DOI] [PubMed] [Google Scholar]
  • 197. Julius S, Nesbitt S. Sympathetic overactivity in hypertension. A moving target. Am J Hypertens. 1996;9(11):113S‐120S. [DOI] [PubMed] [Google Scholar]
  • 198. Lavie P, Shlitner A, Nave R. Cardiac autonomic function during sleep in psychogenic and organic erectile dysfunction. J Sleep Res. 1999;8(2):135‐142. [DOI] [PubMed] [Google Scholar]
  • 199. Bernardi L, Ricordi L, Lazzari P, et al. Impaired circadian modulation of sympathovagal activity in diabetes. A possible explanation for altered temporal onset of cardiovascular disease. Circulation. 1992;86(5):1443‐1452. [DOI] [PubMed] [Google Scholar]
  • 200. Eckberg DL. Sympathovagal balance: a critical appraisal. Circulation. 1997;96(9):3224‐3232. [DOI] [PubMed] [Google Scholar]
  • 201. Manzella D, Grella R, Esposito K, Giugliano D, Barbagallo M, Paolisso G. Blood pressure and cardiac autonomic nervous system in obese type 2 diabetic patients: effect of metformin administration. Am J Hypertens. 2004;17(3):223‐227. [DOI] [PubMed] [Google Scholar]
  • 202. Sundaresan P, Lykos D, Daher A, Diamond T, Morris R, Howes LG. Comparative effects of glibenclamide and metformin on ambulatory blood pressure and cardiovascular reactivity in NIDDM. Diabetes Care. 1997;20(5):692‐697. [DOI] [PubMed] [Google Scholar]
  • 203. Gudbjornsdottir S, Friberg P, Elam M, Attvall S, Lonnroth P, Wallin BG. The effect of metformin and insulin on sympathetic nerve activity, norepinephrine spillover and blood pressure in obese, insulin resistant, normoglycemic, hypertensive men. Blood Press. 1994;3(6):394‐403. [DOI] [PubMed] [Google Scholar]
  • 204. Meller SM, Stilp E, Walker CN, Mena‐Hurtado C. The link between vasculogenic erectile dysfunction, coronary artery disease, and peripheral artery disease: role of metabolic factors and endovascular therapy. J Invasive Cardiol. 2013;25(6):313–319. [PubMed] [Google Scholar]
  • 205. DeFronzo RA, Ferrannini E. Insulin resistance. A multifaceted syndrome responsible for NIDDM, obesity, hypertension, dyslipidemia, and atherosclerotic cardiovascular disease. Diabetes Care. 1991;14(3):173‐194. [DOI] [PubMed] [Google Scholar]
  • 206. Tsai CM, Kuo HC, Hsu CN, Huang LT, Tain YL. Metformin reduces asymmetric dimethylarginine and prevents hypertension in spontaneously hypertensive rats. Transl Res. 2014;164(6):452‐459. [DOI] [PubMed] [Google Scholar]
  • 207. Chien SJ, Lin KM, Kuo HC, et al. Two different approaches to restore renal nitric oxide and prevent hypertension in young spontaneously hypertensive rats: l‐citrulline and nitrate. Transl Res. 2014;163(1):43‐52. [DOI] [PubMed] [Google Scholar]
  • 208. Group UKPDS. UKPDS 28: a randomized trial of efficacy of early addition of metformin in sulfonylurea‐treated type 2 diabetes. Diabetes Care. 1998;21(1):87‐92. [DOI] [PubMed] [Google Scholar]
  • 209. Nagi DK, Yudkin JS. Effects of metformin on insulin resistance, risk factors for cardiovascular disease, and plasminogen activator inhibitor in NIDDM subjects. A study of two ethnic groups. Diabetes Care. 1993;16(4):621‐629. [DOI] [PubMed] [Google Scholar]
  • 210. Casulari LA, Caldas AD, Motta LDC, Lofrano‐Porto A. Effects of metformin and short‐term lifestyle modification on the improvement of male hypogonadism associated with metabolic syndrome. Minerva Endocrinol. 2010;35(3):145‐151. [PubMed] [Google Scholar]
  • 211. Al‐Kuraishy HM, Al‐Gareeb AI. Erectile dysfunction and low sex drive in men with type 2 DM: the potential role of diabetic pharmacotherapy. J Clin Diagn Res. 2016;10(12):FC21‐FC26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212. Rey‐Valzacchi GJ, Costanzo PR, Finger LA, et al. Addition of metformin to sildenafil treatment for erectile dysfunction in eugonadal nondiabetic men with insulin resistance. A prospective, randomized, double‐blind pilot study. J Androl. 2012;33(4):608‐614. [DOI] [PubMed] [Google Scholar]
  • 213. Giannini S, Serio M, Galli A. Pleiotropic effects of thiazolidinediones: taking a look beyond antidiabetic activity. J Endocrinol Invest. 2004;27(10):982‐991. [DOI] [PubMed] [Google Scholar]
  • 214. Martens FM, Visseren FL, de Koning EJ, Rabelink TJ. Short‐term pioglitazone treatment improves vascular function irrespective of metabolic changes in patients with type 2 diabetes. J Cardiovasc Pharmacol. 2005;46(6):773‐778. [DOI] [PubMed] [Google Scholar]
  • 215. Kovanecz I, Ferrini MG, Vernet D, Nolazco G, Rajfer J, Gonzalez‐Cadavid NF. Pioglitazone prevents corporal veno‐occlusive dysfunction in a rat model of type 2 diabetes mellitus. BJU Int. 2006;98(1):116‐124. [DOI] [PubMed] [Google Scholar]
  • 216. Katz EG, Moustafa AA, Heidenberg D, et al. Pioglitazone enhances survival and regeneration of pelvic ganglion neurons after cavernosal nerve injury. Urology. 2016;89:76‐82. [DOI] [PubMed] [Google Scholar]
  • 217. Aliperti LA, Lasker GF, Hagan SS, et al. Efficacy of pioglitazone on erectile function recovery in a rat model of cavernous nerve injury. Urology. 2014;84(5):1122‐1127. [DOI] [PubMed] [Google Scholar]
  • 218. Gholamine B, Shafiei M, Motevallian M, Mahmoudian M. Effects of pioglitazone on erectile dysfunction in sildenafil poor‐responders: a randomized, controlled study. J Pharm Pharm Sci. 2008;11(1):22‐31. [DOI] [PubMed] [Google Scholar]
  • 219. Ruiz Rubio JL, Hernandez M, Rivera de los Arcos L, et al. Role of ATP‐sensitive K+ channels in relaxation of penile resistance arteries. Urology. 2004;63(4):800‐805. [DOI] [PubMed] [Google Scholar]
  • 220. Inzucchi SE, Bergenstal RM, Buse JB, et al. Management of hyperglycemia in type 2 diabetes, 2015: a patient‐centered approach: update to a position statement of the American Diabetes Association and the European Association for the Study of Diabetes. Diabetes Care. 2015;38(1):140‐149. [DOI] [PubMed] [Google Scholar]
  • 221. Neal B, Perkovic V, Mahaffey KW, et al. Canagliflozin and cardiovascular and renal events in type 2 diabetes. N Engl J Med. 2017;377(21):2097‐2099. [DOI] [PubMed] [Google Scholar]
  • 222. Zinman B, Wanner C, Lachin JM, et al. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med. 2015;373(22):2117‐2128. [DOI] [PubMed] [Google Scholar]
  • 223. Verma S, Mazer CD, Fitchett D, et al. Empagliflozin reduces cardiovascular events, mortality and renal events in participants with type 2 diabetes after coronary artery bypass graft surgery: subanalysis of the EMPA‐REG OUTCOME(R) randomised trial. Diabetologia. 2018;61(8):1712‐1723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224. Fitchett D, Butler J, van de Borne P, et al. Effects of empagliflozin on risk for cardiovascular death and heart failure hospitalization across the spectrum of heart failure risk in the EMPA‐REG OUTCOME(R) trial. Eur Heart J. 2018;39(5):363‐370. [DOI] [PubMed] [Google Scholar]
  • 225. Fitchett D, Zinman B, Wanner C, et al. Heart failure outcomes with empagliflozin in patients with type 2 diabetes at high cardiovascular risk: results of the EMPA‐REG OUTCOME(R) trial. Eur Heart J. 2016;37(19):1526‐1534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226. Radholm K, Figtree G, Perkovic V, et al. Canagliflozin and heart failure in type 2 diabetes mellitus: results from the CANVAS program. Circulation. 2018;138(5):458‐468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227. Assaly R, Gorny D, Compagnie S, et al. The favorable effect of empagliflozin on erectile function in an experimental model of type 2 diabetes. J Sex Med. 2018;15(9):1224‐1234. [DOI] [PubMed] [Google Scholar]
  • 228. Huang CY, Shih CM, Tsao NW, et al. Dipeptidyl peptidase‐4 inhibitor improves neovascularization by increasing circulating endothelial progenitor cells. Br J Pharmacol. 2012;167(7):1506‐1519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229. Harmar AJ, Fahrenkrug J, Gozes I, et al. Pharmacology and functions of receptors for vasoactive intestinal peptide and pituitary adenylate cyclase‐activating polypeptide: IUPHAR review. Br J Pharmacol. 2012;166(1):4‐17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230. Yue L, Xu JL, Dong J, et al. Regulatory effect of liraglutide on the expression of eNOS in the corpus cavernosum of diabetic rats. Zhonghua Nan ke Xue. 2016;22(3):212‐218. [PubMed] [Google Scholar]
  • 231. Giagulli VA, Carbone MD, Ramunni MI, et al. Adding liraglutide to lifestyle changes, metformin and testosterone therapy boosts erectile function in diabetic obese men with overt hypogonadism. Andrologia. 2015;3(6):1094‐1103. [DOI] [PubMed] [Google Scholar]
  • 232. Yamanaka M, Shirai M, Shiina H, et al. Diabetes induced erectile dysfunction and apoptosis in penile crura are recovered by insulin treatment in rats. J Urol. 2003;170(1):291‐297. [DOI] [PubMed] [Google Scholar]
  • 233. Wang L, Tian W, Uwais Z, et al. AGE‐breaker ALT‐711 plus insulin could restore erectile function in streptozocin‐induced type 1 diabetic rats. J Sex Med. 2014;11(6):1452‐1462. [DOI] [PubMed] [Google Scholar]
  • 234. Choi WS, Kwon OS, Cho SY, Paick JS, Kim SW. Effect of chronic administration of PDE5 combined with glycemic control on erectile function in streptozotocin‐induced diabetic rats. J Sex Med. 2015;12(3):600‐610. [DOI] [PubMed] [Google Scholar]
  • 235. Shirai M, Yamanaka M, Shiina H, et al. Androgen, estrogen, and progesterone receptor gene regulation during diabetic erectile dysfunction and insulin treatment. Urology. 2004;64(6):1244‐1249. [DOI] [PubMed] [Google Scholar]
  • 236. Gupta BP, Murad MH, Clifton MM, Prokop L, Nehra A, Kopecky SL. The effect of lifestyle modification and cardiovascular risk factor reduction on erectile dysfunction: a systematic review and meta‐analysis. Arch Intern Med. 2011;171(20):1797‐1803. [DOI] [PubMed] [Google Scholar]
  • 237. Esposito K, Giugliano F, Di Palo C, et al. Effect of lifestyle changes on erectile dysfunction in obese men: a randomized controlled trial. J Am Med Assoc. 2004;291(24):2978‐2984. [DOI] [PubMed] [Google Scholar]
  • 238. Wing RR, Rosen RC, Fava JL, et al. Effects of weight loss intervention on erectile function in older men with type 2 diabetes in the Look AHEAD trial. J Sex Med. 2010;7(1 Pt 1):156‐165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239. Rothberg AE, McEwen LN, Kraftson AT, et al. Impact of weight loss on waist circumference and the components of the metabolic syndrome. BMJ Open Diabetes Res Care. 2017;5(1):e000341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240. Franz MJ, Boucher JL, Rutten‐Ramos S, VanWormer JJ. Lifestyle weight‐loss intervention outcomes in overweight and obese adults with type 2 diabetes: a systematic review and meta‐analysis of randomized clinical trials. J Acad Nutr Diet. 2015;115(9):1447‐1463. [DOI] [PubMed] [Google Scholar]
  • 241. Kim C, Dabelea D, Kalyani RR, et al. Changes in visceral adiposity, subcutaneous adiposity, and sex hormones in the diabetes prevention program. J Clin Endocrinol Metab. 2017;102(9):3381‐3389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242. Shao N, Yu XY, Yu YM, et al. Short‐term combined treatment with exenatide and metformin is superior to glimepiride combined metformin in improvement of serum testosterone levels in type 2 diabetic patients with obesity. Andrologia. 2018;50(7):e13039. [DOI] [PubMed] [Google Scholar]
  • 243. Jensterle M, Podbregar A, Goricar K, Gregoric N, Janez A. Effects of liraglutide on obesity‐associated functional hypogonadism in men. Endocr Connect. 2019;8(3):195‐202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244. Turinese I, Marinelli P, Bonini M, et al. Metabolic and cardiovascular response to exercise in patients with type 1 diabetes. J Endocrinol Invest. 2017;40(9):999‐1005. [DOI] [PubMed] [Google Scholar]
  • 245. Rejeski WJ, Ip EH, Bertoni AG, et al. Lifestyle change and mobility in obese adults with type 2 diabetes. N Engl J Med. 2012;366(13):1209‐1217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246. Same RV, Al Rifai M, Feldman DI, et al. Prognostic value of exercise capacity among men undergoing pharmacologic treatment for erectile dysfunction: the FIT Project. Clin Cardiol. 2017;40(11):1049‐1054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247. Colberg SR, Sigal RJ, Yardley JE, et al. Physical activity/exercise and diabetes: a position statement of the American Diabetes Association. Diabetes Care. 2016;39(11):2065‐2079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248. Minami H, Furukawa S, Sakai T, et al. Physical activity and prevalence of erectile dysfunction in Japanese patients with type 2 diabetes mellitus: the Dogo Study. J Diabetes Investig. 2018;9(1):193‐198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249. Cano Sokoloff N, Misra M, Ackerman KE. Exercise, training, and the hypothalamic‐pituitary‐gonadal Axis in men and women. Front Horm Res. 2016;47:27‐43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250. Hayes LD, Herbert P, Sculthorpe NF, Grace FM. Exercise training improves free testosterone in lifelong sedentary aging men. Endocr Connect. 2017;6(5):306‐310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251. Sgro P, Romanelli F, Felici F, et al. Testosterone responses to standardized short‐term sub‐maximal and maximal endurance exercises: issues on the dynamic adaptive role of the hypothalamic‐pituitary‐testicular axis. J Endocrinol Invest. 2014;37(1):13‐24. [DOI] [PubMed] [Google Scholar]
  • 252. Kirilmaz U, Guzel O, Aslan Y, Balci M, Tuncel A, Atan A. The effect of lifestyle modification and glycemic control on the efficiency of sildenafil citrate in patients with erectile dysfunction due to type‐2 diabetes mellitus. Aging Male. 2015;18(4):244‐248. [DOI] [PubMed] [Google Scholar]
  • 253. Salonia A, Bettocchi C, Boeri L, et al. European association of urology guidelines on sexual and reproductive health—2021 update: male sexual dysfunction. Eur Urol. 2021;80:(3):333–357. doi: 10.1016/j.eururo.2021.06.007 [DOI] [PubMed] [Google Scholar]
  • 254. Corbin JD. Mechanisms of action of PDE5 inhibition in erectile dysfunction. Int J Impot Res. 2004;16(Suppl 1):S4‐S7. [DOI] [PubMed] [Google Scholar]
  • 255. Cheng E. Real‐life safety and efficacy of vardenafil in the treatment of erectile dysfunction‐results from 30,010 U.S. patients. J Sex Med. 2007;4(2):432‐439. [DOI] [PubMed] [Google Scholar]
  • 256. Fonseca V, Seftel A, Denne J, Fredlund P. Impact of diabetes mellitus on the severity of erectile dysfunction and response to treatment: analysis of data from tadalafil clinical trials. Diabetologia. 2004;47(11):1914‐1923. [DOI] [PubMed] [Google Scholar]
  • 257. Shah PC, Trivedi NA. A meta‐analysis on efficacy and tolerability of sildenafil for erectile dysfunction in patients with diabetes mellitus. Indian J Sex Transm Dis AIDS. 2018;39(1):1‐6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258. Vickers MA, Satyanarayana R. Phosphodiesterase type 5 inhibitors for the treatment of erectile dysfunction in patients with diabetes mellitus. Int J Impot Res. 2002;14:(6):466–471. doi: 10.1038/sj.ijir.3900910 [DOI] [PubMed] [Google Scholar]
  • 259. Francis SH, Corbin JD. PDE5 inhibitors: targeting erectile dysfunction in diabetics. Curr Opin Pharmacol. 2011;11(6):683‐688. [DOI] [PubMed] [Google Scholar]
  • 260. Hatzimouratidis K, Salonia A, Adaikan G, et al. Pharmacotherapy for erectile dysfunction: recommendations from the fourth International consultation for sexual medicine. J Sex Med. 2016;13(4):465‐488. [DOI] [PubMed] [Google Scholar]
  • 261. Andersson KE. PDE5 inhibitors—pharmacology and clinical applications 20 years after sildenafil discovery. Br J Pharmacol. 2018;175(13):2554‐2565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262. Hatzimouratidis K, Amar E, Eardley I, et al. Guidelines on male sexual dysfunction: erectile dysfunction and premature ejaculation. Eur Urol. 2010;57(5):804‐814. [DOI] [PubMed] [Google Scholar]
  • 263. Wrishko R, Sorsaburu S, Wong D, Strawbridge A, McGill J. Safety, efficacy, and pharmacokinetic overview of low‐dose daily administration of tadalafil. J Sex Med. 2009;6(7):2039‐2048. [DOI] [PubMed] [Google Scholar]
  • 264. Zhao C, Kim SW, Yang DY, et al. Efficacy and safety of once‐daily dosing of udenafil in the treatment of erectile dysfunction: results of a multicenter, randomized, double‐blind, placebo‐controlled trial. Eur Urol. 2011;60(2):380‐387. [DOI] [PubMed] [Google Scholar]
  • 265. Liao X, Qiu S, Bao Y, Wang W, Yang L, Wei Q. Comparative efficacy and safety of phosphodiesterase type 5 inhibitors for erectile dysfunction in diabetic men: a Bayesian network meta‐analysis of randomized controlled trials. World J Urol. 2019;37(6):1061‐1074. [DOI] [PubMed] [Google Scholar]
  • 266. Li X, Zhao Q, Wang J, et al. Efficacy and safety of PDE5 inhibitors in the treatment of diabetes mellitus erectile dysfunction: protocol for a systematic review. Med (Baltimore). 2018;97(40):e12559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267. Balhara YP, Sarkar S, Gupta R. Phosphodiesterase‐5 inhibitors for erectile dysfunction in patients with diabetes mellitus: a systematic review and meta‐analysis of randomized controlled trials. Indian J Endocrinol Metab. 2015;19(4):451‐461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268. Vickers MA, Satyanarayana R. Phosphodiesterase type 5 inhibitors for the treatment of erectile dysfunction in patients with diabetes mellitus. Int J Impot Res. 2002;14(6):466‐471. [DOI] [PubMed] [Google Scholar]
  • 269. Laties AM, Zrenner E. Viagra® (sildenafil citrate) and ophthalmology. Prog Retin Eye Res. 2002;21(5):485‐506. [DOI] [PubMed] [Google Scholar]
  • 270. Deyoung L, Chung E, Kovac JR, Romano W, Brock GB. Daily use of sildenafil improves endothelial function in men with type 2 diabetes. J Androl. 2012;33(2):176‐180. [DOI] [PubMed] [Google Scholar]
  • 271. Price DE, Gingell JC, Gepi‐Attee S, Wareham K, Yates P, Boolell M. Sildenafil: study of a novel oral treatment for erectile dysfunction in diabetic men. Diabet Med. 1998;15(10):821‐825. [DOI] [PubMed] [Google Scholar]
  • 272. Borer J, Armstrong P, Food, Drug Administration C , Renal Drugs Advisory C . Proceedings of the 99th meeting of the food and drug administration cardiovascular and renal drugs advisory committee. May 29th and 30th, 2003. Circulation. 2003;107(23):e9052. [DOI] [PubMed] [Google Scholar]
  • 273. Bischoff E. Potency, selectivity, and consequences of nonselectivity of PDE inhibition. Int J Impot Res. 2004;16(Suppl 1):S11‐S14. [DOI] [PubMed] [Google Scholar]
  • 274. Goldstein I, Young JM, Fischer J, Bangerter K, Segerson T, Taylor T. Vardenafil, a new phosphodiesterase type 5 inhibitor, in the treatment of erectile dysfunction in men with diabetes: a multicenter double‐blind placebo‐controlled fixed‐dose study. Diabetes Care. 2003;26(3):777‐783. [DOI] [PubMed] [Google Scholar]
  • 275. Ishii N, Nagao K, Fujikawa K, Tachibana T, Iwamoto Y, Kamidono S. Vardenafil 20‐mg demonstrated superior efficacy to 10‐mg in Japanese men with diabetes mellitus suffering from erectile dysfunction. Int J Urol. 2006;13(8):1066‐1072. [DOI] [PubMed] [Google Scholar]
  • 276. Santi D, Granata AR, Guidi A, et al. Six months of daily treatment with vardenafil improves parameters of endothelial inflammation and of hypogonadism in male patients with type 2 diabetes and erectile dysfunction: a randomized, double‐blind, prospective trial. Eur J Endocrinol. 2016;174(4):513‐522. [DOI] [PubMed] [Google Scholar]
  • 277. Bruzziches R, Francomano D, Gareri P, Lenzi A, Aversa A. An update on pharmacological treatment of erectile dysfunction with phosphodiesterase type 5 inhibitors. Expert Opin Pharmacother. 2013;14(10). [DOI] [PubMed] [Google Scholar]
  • 278. Padma‐Nathan H. Efficacy and tolerability of tadalafil, a novel phosphodiesterase 5 inhibitor, in treatment of erectile dysfunction. Am J Cardiol. 2003;92(9A):19M‐25M. [DOI] [PubMed] [Google Scholar]
  • 279. Kayik G, Tuzun NS, Durdagi S. Investigation of PDE5/PDE6 and PDE5/PDE11 selective potent tadalafil‐like PDE5 inhibitors using combination of molecular modeling approaches, molecular fingerprint‐based virtual screening protocols and structure‐based pharmacophore development. J Enzyme Inhib Med Chem. 2017;32(1):311‐330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 280. Hatzichristou D, Gambla M, Rubio‐Aurioles E, et al. Efficacy of tadalafil once daily in men with diabetes mellitus and erectile dysfunction. Diabet Med. 2008;25(2):138‐146. [DOI] [PubMed] [Google Scholar]
  • 281. Aversa A, Fittipaldi S, Francomano D, et al. Tadalafil improves lean mass and endothelial function in nonobese men with mild ED/LUTS: in vivo and in vitro characterization. Endocrine. 2017;56(3):639‐648. [DOI] [PubMed] [Google Scholar]
  • 282. Bolat MS, Cinar O, Akdeniz E, Asci R. Low dose daily versus on‐demand high dose tadalafil in diabetic patients with erectile and ejaculatory dysfunction. Int J Impot Res. 2018;30(3):102‐107. [DOI] [PubMed] [Google Scholar]
  • 283. Wang R, Burnett AL, Heller WH, et al. Selectivity of avanafil, a PDE5 inhibitor for the treatment of erectile dysfunction: implications for clinical safety and improved tolerability. J Sex Med. 2012;9(8):2122‐2129. [DOI] [PubMed] [Google Scholar]
  • 284. New drug: Avanafil for erectile dysfunction. Aust Prescr. 2019;42(3):113–114. doi: 10.18773/austprescr.2019.043 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 285. Goldstein I, Jones LA, Belkoff LH, et al. Avanafil for the treatment of erectile dysfunction: a multicenter, randomized, double‐blind study in men with diabetes mellitus. Mayo Clin Proc. 2012;87(9):843‐852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286. Moon du G, Yang DY, Lee CH, et al. A therapeutic confirmatory study to assess the safety and efficacy of Zydena (udenafil) for the treatment of erectile dysfunction in male patients with diabetes mellitus. J Sex Med. 2011;8(7):2048‐2061. [DOI] [PubMed] [Google Scholar]
  • 287. Park SH, Park SW, Cha BY, et al. Comparison of the efficacy and safety of once‐daily dosing and on‐demand use of udenafil for type 2 diabetic patients with erectile dysfunction. Asian J Androl. 2015;17(1):143‐148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 288. Cho MC, Paick JS. A review of the efficacy and safety of mirodenafil in the management of erectile dysfunction. Ther Adv Urol. 2016;8(2):100‐117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 289. Du W, Li J, Fan N, Shang P, Wang Z, Ding H. Efficacy and safety of mirodenafil for patients with erectile dysfunction: a meta‐analysis of three multicenter, randomized, double‐blind, placebo‐controlled clinical trials. Aging Male. 2014;17(2):107‐111. [DOI] [PubMed] [Google Scholar]
  • 290. Park HJ, Choi HK, Ahn TY, et al. Efficacy and safety of oral mirodenafil in the treatment of erectile dysfunction in diabetic men in Korea: a multicenter, randomized, double‐blind, placebo‐controlled clinical trial. J Sex Med. 2010;7(8):2842‐2850. [DOI] [PubMed] [Google Scholar]
  • 291. Dhindsa S, Ghanim H, Batra M, Dandona P. Hypogonadotropic hypogonadism in men with diabesity. Diabetes Care. 2018;41(7):1516‐1525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 292. Corona G, Rastrelli G, Morgentaler A, Sforza A, Mannucci E, Maggi M. Meta‐analysis of results of testosterone therapy on sexual function based on International index of erectile function scores. Eur Urol. 2017;72(6):1000‐1011. [DOI] [PubMed] [Google Scholar]
  • 293. Corona G, Isidori AM, Buvat J, et al. Testosterone supplementation and sexual function: a meta‐analysis study. J Sex Med. 2014;11(6):1577‐1592. [DOI] [PubMed] [Google Scholar]
  • 294. Spitzer M, Basaria S, Travison TG, et al. Effect of testosterone replacement on response to sildenafil citrate in men with erectile dysfunction: a parallel, randomized trial. Ann Intern Med. 2012;157(10):681‐691. [DOI] [PubMed] [Google Scholar]
  • 295. Isidori AM, Giannetta E, Gianfrilli D, et al. Effects of testosterone on sexual function in men: results of a meta‐analysis. Clin Endocrinol (Oxf). 2005;63(4):381‐394. [DOI] [PubMed] [Google Scholar]
  • 296. Carosa E, Martini P, Brandetti F, et al. Type V phosphodiesterase inhibitor treatments for erectile dysfunction increase testosterone levels. Clin Endocrinol (Oxf). 2004;61(3):382‐386. [DOI] [PubMed] [Google Scholar]
  • 297. Balercia G, Boscaro M, Lombardo F, Carosa E, Lenzi A, Jannini EA. Sexual symptoms in endocrine diseases: psychosomatic perspectives. Psychother Psychosom. 2007;76(3):134‐140. [DOI] [PubMed] [Google Scholar]
  • 298. Buvat J, Montorsi F, Maggi M, et al. Hypogonadal men nonresponders to the PDE5 inhibitor tadalafil benefit from normalization of testosterone levels with a 1% hydroalcoholic testosterone gel in the treatment of erectile dysfunction (TADTEST study). J Sex Med. 2011;8(1):284‐293. [DOI] [PubMed] [Google Scholar]
  • 299. Garcia JA, Sanchez PE, Fraile C, Escovar P. Testosterone undecanoate improves erectile dysfunction in hypogonadal men with the metabolic syndrome refractory to treatment with phosphodiesterase type 5 inhibitors alone. Andrologia. 2011;43(5):293‐296. [DOI] [PubMed] [Google Scholar]
  • 300. Jannini EA, Isidori AM, Aversa A, Lenzi A, Althof SE. Which is first? The controversial issue of precedence in the treatment of male sexual dysfunctions. J Sex Med. 2013;10(10):2359‐2369. [DOI] [PubMed] [Google Scholar]
  • 301. Barbagallo F, Campolo F, Franceschini E, et al. PDE5 inhibitors in type 2 diabetes cardiovascular complications. Endocr. 2020;1(2):90‐101. [Google Scholar]
  • 302. Hackett G, Jones PW, Strange RC, Ramachandran S. Statin, testosterone and phosphodiesterase 5‐inhibitor treatments and age related mortality in diabetes. World J Diabetes. 2017;8(3):104‐111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 303. Anderson SG, Hutchings DC, Woodward M, et al. Phosphodiesterase type‐5 inhibitor use in type 2 diabetes is associated with a reduction in all‐cause mortality. Heart. 2016;102(21):1750‐1756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 304. Desouza C, Parulkar A, Lumpkin D, Akers D, Fonseca VA. Acute and prolonged effects of sildenafil on brachial artery flow‐mediated dilatation in type 2 diabetes. Diabetes Care. 2002;25(8):1336‐1339. [DOI] [PubMed] [Google Scholar]
  • 305. Rosano GM, Aversa A, Vitale C, Fabbri A, Fini M, Spera G. Chronic treatment with tadalafil improves endothelial function in men with increased cardiovascular risk. Eur Urol. 2005;47(2):214‐222. discussion 220‐212. [DOI] [PubMed] [Google Scholar]
  • 306. Aversa A, Vitale C, Volterrani M, et al. Chronic administration of Sildenafil improves markers of endothelial function in men with type 2 diabetes. Diabet Med. 2008;25(1):37‐44. [DOI] [PubMed] [Google Scholar]
  • 307. Poolsup N, Suksomboon N, Aung N. Effect of phosphodiesterase‐5 inhibitors on glycemic control in person with type 2 diabetes mellitus: a systematic review and meta‐analysis. J Clin Transl Endocrinol. 2016;6:50‐55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 308. Varma A, Das A, Hoke NN, Durrant DE, Salloum FN, Kukreja RC. Anti‐inflammatory and cardioprotective effects of tadalafil in diabetic mice. PLoS One. 2012;7(9):e45243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 309. Santi D, Giannetta E, Isidori AM, Vitale C, Aversa A, Simoni M. Therapy of endocrine disease. Effects of chronic use of phosphodiesterase inhibitors on endothelial markers in type 2 diabetes mellitus: a meta‐analysis. Eur J Endocrinol. 2015;172(3):R103‐R114. [DOI] [PubMed] [Google Scholar]
  • 310. Koka S, Xi L, Kukreja RC. Chronic treatment with long acting phosphodiesterase‐5 inhibitor tadalafil alters proteomic changes associated with cytoskeletal rearrangement and redox regulation in type 2 diabetic hearts. Basic Res Cardiol. 2012;107(2):249. [DOI] [PubMed] [Google Scholar]
  • 311. Koka S, Aluri HS, Xi L, Lesnefsky EJ, Kukreja RC. Chronic inhibition of phosphodiesterase 5 with tadalafil attenuates mitochondrial dysfunction in type 2 diabetic hearts: potential role of NO/SIRT1/PGC‐1alpha signaling. Am J Physiol Heart Circ Physiol. 2014;306(11):H1558‐H1568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 312. Matyas C, Nemeth BT, Olah A, et al. Prevention of the development of heart failure with preserved ejection fraction by the phosphodiesterase‐5A inhibitor vardenafil in rats with type 2 diabetes. Eur J Heart Fail. 2017;19(3):326‐336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 313. West TM, Wang Q, Deng B, et al. Phosphodiesterase 5 associates with beta2 adrenergic receptor to modulate cardiac function in type 2 diabetic hearts. J Am Heart Assoc. 2019;8(15):e012273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 314. Salloum FN, Abbate A, Das A, et al. Sildenafil (Viagra) attenuates ischemic cardiomyopathy and improves left ventricular function in mice. Am J Physiol Heart Circ Physiol. 2008;294(3):H1398‐H1406. [DOI] [PubMed] [Google Scholar]
  • 315. Das A, Xi L, Kukreja RC. Phosphodiesterase‐5 inhibitor sildenafil preconditions adult cardiac myocytes against necrosis and apoptosis. Essential role of nitric oxide signaling. J Biol Chem. 2005;280(13):12944‐12955. [DOI] [PubMed] [Google Scholar]
  • 316. Hutchings DC, Anderson SG, Caldwell JL, Trafford AW. Phosphodiesterase‐5 inhibitors and the heart: compound cardioprotection? Heart. 2018;104(15):1244‐1250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 317. Wang H, Anstrom K, Ilkayeva O, et al. Sildenafil treatment in heart failure with preserved ejection fraction: targeted metabolomic profiling in the RELAX trial. JAMA Cardiol. 2017;2(8):896‐901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 318. Guazzi M, Vicenzi M, Arena R, Guazzi MD. PDE5 inhibition with sildenafil improves left ventricular diastolic function, cardiac geometry, and clinical status in patients with stable systolic heart failure: results of a 1‐year, prospective, randomized, placebo‐controlled study. Circ Heart Fail. 2011;4(1):8‐17. [DOI] [PubMed] [Google Scholar]
  • 319. Giannetta E, Isidori AM, Galea N, et al. Chronic inhibition of cGMP phosphodiesterase 5A improves diabetic cardiomyopathy: a randomized, controlled clinical trial using magnetic resonance imaging with myocardial tagging. Circulation. 2012;125(19):2323‐2333. [DOI] [PubMed] [Google Scholar]
  • 320. Heaton JP, Lording D, Liu SN, et al. Intracavernosal alprostadil is effective for the treatment of erectile dysfunction in diabetic men. Int J Impot Res. 2001;13(6):317‐321. [DOI] [PubMed] [Google Scholar]
  • 321. Tsai YS, Lin JS, Lin YM. Safety and efficacy of alprostadil sterile powder (S. Po., CAVERJECT) in diabetic patients with erectile dysfunction. Eur Urol. 2000;38(2):177‐183. [DOI] [PubMed] [Google Scholar]
  • 322. Hatzimouratidis K, Hatzichristou DG. A comparative review of the options for treatment of erectile dysfunction: which treatment for which patient? Drugs. 2005;65(12):1621‐1650. [DOI] [PubMed] [Google Scholar]
  • 323. Zhang L, Zhang HY, Huang FC, Huang Q, Liu C, Li JR. Study on the clinical value of alprostadil combined with alpha‐lipoic acid in treatment of type 2 diabetes mellitus patients with erectile dysfunction. Eur Rev Med Pharmacol Sci. 2016;20(18):3930‐3933. [PubMed] [Google Scholar]
  • 324. Perimenis P, Konstantinopoulos A, Perimeni PP, et al. Long‐term treatment with intracavernosal injections in diabetic men with erectile dysfunction. Asian J Androl. 2006;8(2):219‐224. [DOI] [PubMed] [Google Scholar]
  • 325. Perimenis P, Gyftopoulos K, Athanasopoulos A, Barbalias G. Diabetic impotence treated by intracavernosal injections: high treatment compliance and increasing dosage of vaso‐active drugs. Eur Urol. 2001;40(4):398‐403. discussion 403. [DOI] [PubMed] [Google Scholar]
  • 326. Vardi Y, Sprecher E, Gruenwald I. Logistic regression and survival analysis of 450 impotent patients treated with injection therapy: long‐term dropout parameters. J Urol. 2000;163(2):467‐470. [PubMed] [Google Scholar]
  • 327. Aversa A, Francomano D, Lenzi A. Does testosterone supplementation increase PDE5‐inhibitor responses in difficult‐to‐treat erectile dysfunction patients? Expert Opin Pharmacother. 2015;16(5):625‐628. [DOI] [PubMed] [Google Scholar]
  • 328. Garrido Abad P, Sinues Ojas B, Martinez Blazquez L, Conde Caturla P, Fernandez Arjona M. Safety and efficacy of intraurethral alprostadil in patients with erectile dysfunction refractory to treatment using phosphodiesterase‐5 inhibitors. Actas Urol Esp. 2015;39(10):635‐640. [DOI] [PubMed] [Google Scholar]
  • 329. Rooney M, Pfister W, Mahoney M, Nelson M, Yeager J, Steidle C. Long‐term, multicenter study of the safety and efficacy of topical alprostadil cream in male patients with erectile dysfunction. J Sex Med. 2009;6(2):520‐534. [DOI] [PubMed] [Google Scholar]
  • 330. Moncada I, Cuzin B. Clinical efficacy and safety of Vitaros(c)/Virirec(c) (Alprostadil cream) for the treatment of erectile dysfunction. Urologia. 2015;82(2):84‐92. [DOI] [PubMed] [Google Scholar]
  • 331. Bosshardt RJ, Farwerk R, Sikora R, Sohn M, Jakse G. Objective measurement of the effectiveness, therapeutic success and dynamic mechanisms of the vacuum device. Br J Urol. 1995;75(6):786‐791. [DOI] [PubMed] [Google Scholar]
  • 332. Canguven O, Bailen J, Fredriksson W, Bock D, Burnett AL. Combination of vacuum erection device and PDE5 inhibitors as salvage therapy in PDE5 inhibitor nonresponders with erectile dysfunction. J Sex Med. 2009;6(9):2561‐2567. [DOI] [PubMed] [Google Scholar]
  • 333. Arauz‐Pacheco C, Basco M, Ramirez LC, Pita JM, Pruneda L, Raskin P. Treatment of diabetic impotence with a vacuum device: efficacy and effects on psychological status. Am J Med Sci. 1992;303(5):281‐284. [DOI] [PubMed] [Google Scholar]
  • 334. Pajovic B, Dimitrovski A, Fatic N, Malidzan M, Vukovic M. Vacuum erection device in treatment of organic erectile dysfunction and penile vascular differences between patients with DM type I and DM type II. Aging Male. 2017;20(1):49‐53. [DOI] [PubMed] [Google Scholar]
  • 335. Sun L, Peng FL, Yu ZL, Liu CL, Chen J. Combined sildenafil with vacuum erection device therapy in the management of diabetic men with erectile dysfunction after failure of first‐line sildenafil monotherapy. Int J Urol. 2014;21(12):1263‐1267. [DOI] [PubMed] [Google Scholar]
  • 336. Zermann DH, Kutzenberger J, Sauerwein D, Schubert J, Loeffler U. Penile prosthetic surgery in neurologically impaired patients: long‐term followup. J Urol. 2006;175(3 Pt 1):1041‐1044. discussion 1044. [DOI] [PubMed] [Google Scholar]
  • 337. Levine LA, Estrada CR, Morgentaler A. Mechanical reliability and safety of, and patient satisfaction with the ambicor inflatable penile prosthesis: results of a 2 center study. J Urol. 2001;166(3):932‐937. [PubMed] [Google Scholar]
  • 338. Montorsi F, Rigatti P, Carmignani G, et al. AMS three‐piece inflatable implants for erectile dysfunction: a long‐term multi‐institutional study in 200 consecutive patients. Eur Urol. 2000;37(1):50‐55. [DOI] [PubMed] [Google Scholar]
  • 339. Lipsky MJ, Onyeji I, Golan R, et al. Diabetes is a risk factor for inflatable penile prosthesis infection: analysis of a large statewide database. Sex Med. 2019;7(1):35‐40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 340. Christodoulidou M, Pearce I. Infection of penile prostheses in patients with diabetes mellitus. Surg Infect (Larchmt). 2016;17(1):2‐8. [DOI] [PubMed] [Google Scholar]
  • 341. Cakan M, Demirel F, Karabacak O, Yalcinkaya F, Altug U. Risk factors for penile prosthetic infection. Int Urol Nephrol. 2003;35(2):209‐213. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Not applicable.


Articles from Diabetes/Metabolism Research and Reviews are provided here courtesy of Wiley

RESOURCES