Erectile dysfunction associated with prostate cancer treatment and therapeutic advances: a narrative review
Review Article

Erectile dysfunction associated with prostate cancer treatment and therapeutic advances: a narrative review

Xin Zhang1, Jiangxiao Pan2, Runnan Xu3, Yijia Fu1, Bodong Lv3

1The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China; 2Hangzhou Jiuyao Medical Beauty Clinic, Hangzhou, China; 3Department of Urology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China

Contributions: (I) Conception and design: B Lv; (II) Administrative support: B Lv, X Zhang; (III) Provision of study materials or patients: X Zhang, J Pan; (IV) Collection and assembly of data: R Xu, Y Fu; (V) Data analysis and interpretation: X Zhang, Y Fu; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Bodong Lv, MD. Department of Urology, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 300 Juyuan Road, Shangcheng District, Hangzhou 310000, China. Email: lbd168@zju.edu.cn.

Background and Objective: The treatment of prostate cancer (PCa) often comes with the risk of erectile dysfunction (ED). As therapeutic technologies continue to advance, the incidence of ED and its treatment methods are also evolving. This paper aimed to provide a comprehensive analysis of the latest developments in PCa treatment, with a particular focus on its relationship with ED, and to review current innovative strategies for ED treatment.

Methods: This study conducted a literature search in databases including PubMed, Excerpta Medica, Web of Science, Scopus, and the Cochrane Library, using keywords including “prostate cancer”, “active surveillance”, “radiation therapy”, “cryotherapy”, “radical prostatectomy”, “immunotherapy”, “chemotherapy”, “androgen deprivation therapy”, “erectile dysfunction”, and “therapeutic advances”, to collect English-language literature published from 1966 to June 2024.

Key Content and Findings: Active surveillance (AS) strategies have significantly reduced the incidence of ED. Technological advancements such as radiation therapy (RT), precise delineation techniques, and improvements in cryotherapy equipment are all dedicated to reducing the risk of ED. Intraoperative nerve monitoring combined with robot-assisted radical prostatectomy (RP) plays a key role in protecting the cavernous nerves and improving postoperative erectile function (EF) recovery. The impact of immunotherapy and chemotherapy on the risk of ED still needs to be clarified with additional clinical data. Androgen deprivation therapy (ADT) is often carried out as part of a combined treatment or through novel administration modalities to reduce its side effects. Given the limitations of traditional ED treatment methods, emerging treatments such as physical energy therapy, stem cell and platelet-rich plasma (PRP) therapy, gene and targeted therapies, tissue engineering and nerve transplantation, and traditional Chinese medicine (TCM) represent novel solutions for ED treatment.

Conclusions: With the innovation of PCa treatment technologies, the incidence of ED has declined, and emerging ED treatment methods have benefited the recovery of sexual function in patients with PCa. These advances may form the basis from which further innovations in PCa treatment strategies can be developed.

Keywords: Prostate cancer treatment (PCa treatment); erectile dysfunction (ED); therapeutic advances


Submitted Sep 23, 2024. Accepted for publication Nov 12, 2024. Published online Nov 28, 2024.

doi: 10.21037/tau-24-514


Introduction

According to the World Health Organization, prostate cancer (PCa) is the most common type of cancer in 118 countries/regions (as of 2022) and the second most common malignant tumor in men worldwide (1,2). In 2020 alone, there were 1,466,680 new cases, accounting for 7.3% of the total new cancer cases, which led to 396,792 deaths, constituting 4.1% of the total cancer-related deaths. It is projected that by 2040, the number of new annual PCa cases worldwide will increase to about 2.3 million, with number of related deaths reaching 740,000 (3). The treatment of PCa not only poses a threat to health but also may lead to side effects such as erectile dysfunction (ED), even when accounting for the shared risk factors between PCa and ED, including aging, obesity, metabolic disorders, and sleep apnea (4,5). ED is defined as the inability to achieve and maintain an erection sufficient for satisfactory sexual activity, either occasionally or consistently (6). Higher residual radiation doses in the penile bulb and penile root after radiation therapy (RT) (7), damage to the neurovascular bundles of the prostate due to radical prostatectomy (RP) (8) and cryotherapy (9), and reduced testosterone levels due to androgen deprivation therapy (ADT) (10) can all lead to ED. With the advancement of diagnostic and therapeutic equipment and technology, the relationship between PCa treatment and ED is undergoing transformation. Emerging physical energy therapies (11), the rise of precision medicine (gene and targeted therapies) (12), the exploration of biotissue engineering and nerve transplantation (13), and research on traditional Chinese medicine (TCM) (14) in the treatment of ED represent new approaches for addressing ED in patients with PCa. Therefore, it is essential to systematically review the connection between PCa treatment and ED and to discuss the latest strategies for managing ED. We present this article in accordance with the Narrative Review reporting checklist (available at https://tau.amegroups.com/article/view/10.21037/tau-24-514/rc).


Methods

This study employed a systematic literature search strategy to ensure comprehensiveness and accuracy (Table 1). We established the following inclusion criteria: (I) all studies must be directly related to the treatment of PCa and its impact on ED, including the latest advancements in the treatment of ED; (II) studies must be published in peer-reviewed academic journals to ensure the quality of the research; (III) we limited the language of the studies to English to ensure our accurate understanding and analysis of the literature. Exclusion criteria included: (I) non-empirical studies (such as case reports); (II) studies with duplicate data publication; (III) those with small sample sizes that might affect the generalizability of the results. Through these criteria, we systematically screened relevant literature from databases such as PubMed, Excerpta Medica, Web of Science, Scopus, and the Cochrane Library from June 1966 to June 2024. By employing this method, we were able to conduct a narrative review of the evolution of PCa treatment methods, with particular emphasis on the incidence of ED as a complication of treatment. Furthermore, we summarized the latest treatment methods in the field of ED, providing valuable references for clinicians and researchers.

Table 1

Search strategy

Item Specification
Date of search July 2, 2024
Databases and other sources searched PubMed, Excerpta Medica, Web of Science, Scopus, Cochrane Library
Search terms used Prostate cancer, active surveillance, radiation therapy, cryotherapy, radical prostatectomy, immunotherapy, chemotherapy, androgen deprivation therapy, erectile dysfunction, therapeutic advances
Timeframe June 1966 to June 2024
Inclusion and exclusion criteria Inclusion criteria: (I) all studies must be directly related to the treatment of PCa and its impact on ED, including the latest advancements in the treatment of ED; (II) studies must be published in peer-reviewed academic journals to ensure the quality of the research; (III) we limited the language of the studies to English to ensure our accurate understanding and analysis of the literature
Exclusion criteria: (I) non-empirical studies (such as case reports, etc.); (II) studies with duplicate data publication; (III) those with small sample sizes that might affect the generalizability of the results
Selection process All the authors selected the studies together

PCa, prostate cancer; ED, erectile dysfunction.


Discussion

The impact of PCa treatment on erectile function (EF)

Active surveillance (AS) and ED

The widespread use of prostate-specific antigen (PSA) screening and improved biopsy techniques have increased the early diagnosis rate of PCa by 10% to 80% globally (15). A study has shown that the metastasis and mortality rates of early-stage, localized PCa are extremely low within 10 to 15 years (16). This indicates an urgent need to explore new treatment plans for patients with early, low-risk, localized PCa to avoid the decline in quality of life caused by overtreatment. AS is a method of assessing tumor progression by monitoring PSA via digital rectal examination and imaging, enabling the adjustment of the treatment plan in a timely manner (17). A prospective study by Newcomb et al. published in JAMA showed that 49% of patients undergoing AS had no progression or treatment within 10 years, with a metastatic cancer incidence rate of less than 2% and a cancer-related mortality rate of less than 1% (18). A multicenter study involving 5,302 patients by Bokhorst et al. also produced similar results, with a 10-year distant metastasis rate and cancer-specific mortality rate of just 0.5% and 0.1%, respectively (19). A prospective study by Hirama et al. assessing the impact of AS on 525 patients with PCa showed that the patients had a good quality of life and mental health status, with low levels of anxiety and depression (20). These results confirm that AS is an effective management strategy for early-stage, localized PCa. In developed countries, 37.9% to 74% of patients with PCa select AS as their first choice (21-23).

Compared with active treatment, AS significantly reduces the incidence of ED in patients with PCa. A cohort study by Donovan et al. showed that among patients with PCa undergoing AS, 52% at 6 months, 41% at 36 months, and 30% at 6 years were able to maintain an erection sufficient for sexual intercourse, representing superior outcomes to those of patients undergoing RP or RT at these time points (24). Alberti et al. reviewed 17 prospective clinical experiments using different patient-reported outcome tools [such as Expanded Prostate Cancer Index Composite 26 (EPIC-26)], including two randomized controlled trials (RCTs) and other prospective observational or cross-sectional studies, and found that the impact of AS on EF was minimal compared to that of RP and RT (25).

It has been reported that PSA biopsies may increase the risk of ED. A longitudinal study by Fujita et al. followed 333 patients with PCa undergoing AS, of whom 69% completed follow-up. Those who underwent 3 or more biopsies had an average decrease of 5.7 points in their Sexual Health Inventory for Men (SHIM) score, while those who underwent 0 to 2 biopsies had an average decrease of 2.3 points (26). However, the observational study by Braun et al. (27) and Pearce et al. (28) did not find a direct link between the number of biopsies and ED. The application of different prostate biopsy techniques significantly influences the risk of ED. Extensive research indicates a marked decrease in the International Index of Erectile Function-5 (IIEF-5) scores at 1, 3, and 6 months post transrectal prostate biopsy (TRPBx) (29-32). Transperineal prostate biopsy (TPB) is favored by clinicians due to its more effective sampling of the anterior and apical regions of the PCa. The largest study examining ED following TPB included 610 patients who underwent 18-core (extended TPB), 360 who underwent 28-core (saturated TPB), and 210 who underwent 32-core biopsies. The findings revealed mild ED in 15 (4.2%) of the extended TPB group, 7 (6.4%) of the saturated TPB group, and 7 (7%) of the saturated TPB group one month after biopsy, with no persistent effects observed after three months. Additionally, a study by Chong et al. found a significant decrease in IIEF-5 scores one month post-biopsy in 64 patients who underwent TPB (19.5 vs. 10.5) (P<0.001) (33). However, a single-center, observational, prospective study by García Rojo et al. noted no significant difference in IIEF-5 scores at 3- and 6-month post-biopsy compared to pre-biopsy levels following both transrectal and transperineal prostate biopsies (34). These differences may be attributed to the location of the tumor, the skill level of the clinician performing the biopsy, and the patient’s own condition. The progression of PCa can lead to an increase in prostate volume, which in turn can cause lower urinary tract symptoms (LUTS), which have been confirmed to be an independent risk factor for ED (35,36). In the future, incorporating patient preferences and quality of life assessments into the standards of AS plans can aid in comprehensively monitoring patients’ EF and effectively improving their quality of life. However, it is important to note that unhealthy lifestyle habits such as smoking, and obesity may simultaneously cause increases in PSA levels and ED, indicating that these factors should be considered when establishing AS criteria (37,38).

RT and ED

RT, including external beam radiation therapy (EBRT) and brachytherapy (BT), is an effective treatment for PCa. Before external beam RT is implemented, it is necessary to delineate the radiation target area. The resolution of early two-dimensional X-ray technology and limitations in three dimensions made it difficult to accurately locate the RT target area. In addition, previous three-dimensional conformal radiation therapy (3DCRT) relied on fixed beams and collimators to block the rays, causing damage to surrounding tissues and precipitating complications such as ED and urinary incontinence. Several prospective trials have shown that the incidence of ED after 3DCRT ranges from 7% to 72% (39-44). Subsequently, the introduction of computed tomography (CT) and magnetic resonance imaging (MRI) have significantly improved the accuracy of delineation, with CT reducing the target error by more than 30% (45) and MRI being 30% more accurate than CT (46,47).

In the 1990s, intensity-modulated radiation therapy (IMRT) gradually became the preferred strategy of clinicians. IMRT involves the adjustment the collimator from different angles to irradiate the tumor through computer control and imaging guidance, providing a higher radiation concentration and reducing the incidence of ED. Brown et al. followed up with 32 patients who underwent IMRT, and in a median follow-up time of 36.8 months, 50% of patients reported a SHIM score of less than 10 (48).

Hypofractionated RT, which rose to favor in the beginning of the 21st century, utilizes the repair ability of healthy cells and the insufficient repair of tumor cells and administers precise doses in fractions to eliminate the tumor and protect normal tissues. Stereotactic body radiation therapy (SBRT) represents a refinement of this method, enhancing the accuracy and efficiency of hypofractionated RT through three-dimensional precise positioning, further reducing the incidence of ED. In a single-center prospective study, among 184 patients with initially intact EF who underwent SBRT treatment, the functional erection preservation rate was 57% at 24 months and decreased to 45% at 60 months (49).

In recent years, image-guided radiation therapy (IGRT) has provided real-time monitoring of tumors and surrounding tissues by employing CT and MRI during treatment, allowing for the precise positioning and dynamic adjustment of treatment plans. From the IGRT substudy of the CHHiP (Conventional or Hypofractionated High-Dose Intensity-Modulated Radiotherapy for Prostate Cancer) trial, Murray et al. found that IGRT technology reduced the radiation dose to the penile bulb, reducing the risk of severe ED in patients with PCa (50).

At the beginning of the 21st century, BT became increasingly popular due to its lower incidence of ED. This method acts directly on the tumor by implanting radioactive particles or isotope catheters. Incrocci et al.’s study showed that BT with iodine-125 (I-125) or palladium-103 (Pd-103), whether used alone or in combination, yielded an ED incidence rate between 5% and 51% (51). The latest RT technology uses the “Bragg peak” effect of heavy particles, such as protons and carbon ions, which release a maximum amount of energy before penetrating and stopping at the tumor tissue, thereby effectively destroying tumor cells while minimizing the impact on surrounding healthy tissues. Ho et al. followed up with 254 patients who received proton therapy for as long as 7.1 years, finding that their EF baseline of 90% decreased to 62% after 1 year and to 67% after 5 years (52). The peak of ED occurs between 6 months and 2 years after RT (53,54).

It is currently believed that the etiology of ED caused by RT primarily revolves around the radiation-induced damage to penile vasculature, cavernous nerves, and penile tissue. In the 1980s, Goldstein et al. performed selective internal pudendal artery angiography on patients who had undergone RT, revealing obstructive vascular disease within the pelvic radiation field (55). In 1998, Zelefsky and Eid applied ultrasound scanning of cavernous tissue in patients treated with RT and discovered that arterial dysfunction is a principal cause of ED after RT. Subsequent animal studies have revealed that the reduction of nitric oxide synthase (NOS)-containing nerve fibers within the penile corpora may play a critical role in the development of ED following RT (56), a finding that could provide significant insights into understanding the pathogenesis of post-RT ED (57). Furthermore, the incidence of ED is also correlated with patient age, economic status, and the standard of local healthcare (58,59).

The development of RT technology has consistently relied upon improving the accuracy of delineation and creating more effective drug administration methods. In the future, cutting-edge imaging technology—such as use of specific biomarkers—intraoperative real-time monitoring of IGRT, and the development of safer and more effective radiation sources required for BT, will advance RT strategies and reduce the occurrence of ED.

Cryotherapy and ED

In 1966, Cooper and Hirose pioneered the first generation of modern cryotherapy using liquid nitrogen cooling probes (60). In 1968, Soanes and Hirose applied this technology to the ablation of prostate tumors, but due to the lack of real-time monitoring and protection of surrounding tissues, thus often led to complications such as urethral rectal fistula (61). Unfortunately, few studies have explored the impact of cryotherapy on EF. In 1993, Onik et al. conducted a study introducing ultrasound imaging monitoring technology into cryotherapy, and the results showed that 65% of the 14 patients with normal preoperative EF experienced ED (62).

In the 1990s, an innovative cryotherapy device leveraging the Joule-Thomson throttling refrigeration principle was developed, enabling rapid cooling for cryotherapy treatment with argon gas and controlled needle removal through heating with helium gas. Unexpectedly, related studies on the impact of the third-generation cryotherapy on EF produced highly discrepant findings. In Asterling et al.’s prospective study, 20 out of 25 patients (80%) with normal preoperative EF experienced ED during the follow-up period of 3 to 54 months after surgery (63). Zhou et al.’s meta-analysis also found that patients had a higher incidence of ED after cryotherapy (64). However, in Roberts et al.’s retrospective study, only 20.1% of the 380 patients who underwent cryotherapy reported ED after 12 months (65). This difference may be related to the patient’s age, physical condition, cancer staging, and tumor location. The mechanism of ED caused by cryotherapy is related to the cryogenic damage to the neurovascular bundle near the prostate (9). In the future, cryotherapy can employ imaging and tumor cell markers to achieve precise positioning of the treatment area, which is expected to further reduce the damage to the neurovascular bundle and more effectively preserve patients’ EF.

RP and ED

RP often leads to cavernous nerve damage due to intraoperative traction, thermal injury, postoperative local tissue inflammation, and ischemia, which in turn triggers a series of pathological changes such as apoptosis, pyroptosis, and autophagy of smooth muscle cells, ultimately resulting in the onset of ED (Figure 1).

Figure 1 Causes of erectile dysfunction after radical prostatectomy. RP, radical prostatectomy; CC, corpus cavernosum; CCSMC, corpus cavernosum smooth muscle cell; ROS, reactive oxygen species; ED, erectile dysfunction.

In 1905, Young was the first to use the perineal approach for radical perineal prostatectomy (RPP) (66). In 1945, Millin described retroperitoneal prostatectomy, laying the foundation for the later retropubic radical prostatectomy (RRP) (67). Walsh first used this approach for RRP in 1949. However, almost all patients experienced severe ED (68). Subsequently, research into RP has long sought the means to reducing RP-related ED complications.

In 1983, Walsh discovered the neurovascular bundle that runs along the posterolateral aspect of the prostate through autopsy, and then developed an RRP technique that retains these neurovascular bundles. That is, by making an incision in front of the neurovascular bundle and closely separating from the prostate, to reduce the damage to the cavernous nerves (69). Walsh et al. modified nerve-sparing surgery on 12 patients with PCa aged 44 to 68 years, and all patients were able to have erections 8 weeks after surgery, with 50% successfully achieving vaginal penetration and orgasm (69). Biopsy of the prostate specimen after the modified operation showed that the technique could more thoroughly remove the tumor due to it being performed completely within the lateral pelvic fascia (69,70). Subsequently, the nerve-sparing RP technique was widely used in the clinic, effectively improving the EF of patients after RP.

In the 1990s, significant developments were made in laparoscopic radical prostatectomy (LRP) technology (71). However, due to its high technical requirements, it was not until Binder and Kramer introduced the da Vinci robot in 2000 that robot-assisted laparoscopic radical prostatectomy (RALP) began to be widely adopted (72). Subsequently, Menon et al. optimized the RALP technique and proposed an improved retropubic approach, the Vattikuti Institute prostatectomy technique, to better protect the neurovascular bundle and lateral prostate fascia, making RALP the mainstream method of surgical treatment for PCa in the United States (73).

Data from Memorial Sloan Kettering Cancer Center showed that from 2008 to 2015, the proportion of patients undergoing RALP increased from 21% to 68%, while the proportion of patients undergoing open radical prostatectomy (ORP) and LRP decreased from 29% and 50% to 14% and 28%, respectively (74). In Moretti et al.’s meta-analysis, at 6 months after surgery, the recovery rate of EF in patients undergoing RRP, LRP, and RALP was 30%, 44%, and 54%, respectively; 18 months later, the recovery rate increased to 58%, 52%, and 67%, respectively, with the recovery rate of RALP consistently being the highest (75). Yang et al. (76) conducted a study assessing the efficacy of transvesical robot-assisted RP in treating localized PCa. They found that the approach, which involves a combined longitudinal incision through the bladder neck and prostate, not only effectively removes the tumor but also particularly preserves the anatomical structures related to sexual function and urinary control, which is especially beneficial for the recovery of EF postoperatively. In 2023, Zhang et al. proposed a modified Retzius space preservation technique (Hood technique) for the anterior approach. This method only requires limited dissection of the fat layer on the surface of the bladder’s anterior aspect in establishing a surgical operating space and maximizes the preservation of the key anatomical structures around the urethra related to urination and EF. However, there is a lack of large clinical trials conducted on the impact of this technique on EF (77). It is worth noting that there is an association between single-port or multi-port robot-assisted laparoscopic surgery and the incidence of postoperative ED, but regrettably, there are currently no clinical data reported on this matter.

Intraoperative cavernous nerve monitoring is crucial for protecting EF. Electrical nerve stimulation, as the earliest intraoperative nerve monitoring technology, is widely applied via the CaverMap device, but faces limitations in clinical application due to insufficient specificity and a high false-positive rate (78,79). Optical coherence tomography (OCT) is an imaging technology based on fiber optics, which can be integrated into surgical instruments and achieve real-time high-resolution tissue imaging through backscattered light (80,81). Experimental versions of this technology in RP are being explored. Other intraoperative nerve monitoring technologies such as optical nerve stimulation (ONS), fluorescence dye marking, confocal, and multiphoton microscopy have not been widely adopted due to their distinct limitations (82-84).

The future development of RP surgery needs to continue in the direction of reducing damage to the prostatic neurovascular bundles, such as developing more real-time and convenient intraoperative neural imaging technologies, and improving computer-assisted robotic surgery to reduce intraoperative injuries. In addition, strengthening the rehabilitation of patients’ postoperative EF is also crucial.

Immunotherapy and ED

In 2000, Hurwitz found that anti-CTLA-4 (a T-cell inhibitory receptor) treatment could reduce the grade of prostate tumors in mice, although this treatment was accompanied by prostatitis and destruction of epithelial cells (85). This finding suggests that PCa cells may express specific antigens and may be suitable for immunotherapy. A phase III trial of sipuleucel-T, a customized vaccine based on modified immune cells from the patient, was conducted in 2006 involving 127 patients from 19 clinical centers. Patients treated with sipuleucel-T had a median overall survival of 25.9 months, while patients treated with placebo had a median overall survival of 21.4 months (86). In 2010, the US Food and Drug Administration (FDA) approved sipuleucel-T for the treatment of metastatic castration-resistant prostate cancer (mCRPC), marking the beginning of immunotherapy for this disease (87). There are currently several immunotherapy drugs being studied, including CTLA-4 inhibitors, PD-1/PD-L1 inhibitors, and antibody therapies targeting prostate-specific membrane antigen (PSMA). However, their efficacy is limited, and their side effects are severe (88-90).

There are currently no reports of PCa immunotherapy causing ED. Ryder et al.’s analysis showed that among 211 melanoma patients treated with ipilimumab, 8% developed pituitary inflammation, and 6% developed hypothyroidism or thyroiditis, all of which increased the risk of ED (91). Similarly, Mahzari et al.’s study found that among 6 patients who developed pituitary inflammation after treatment with Ipilimumab, 4 (67%) had central hypogonadism (92). These cases of ED were not directly caused by immunotherapy, and the patients did not have PCa. Additional large-sample, double-blind, clinical RCTs are needed in the future to clarify the connection between PCa immunotherapy drugs and ED.

Chemotherapy and ED

Early chemotherapy for PCa relied on nonspecific cytotoxic drugs such as estramustine, which could alleviate symptoms but demonstrated limited ability in improving survival rates (93). Subsequently, it was found that the combination of anthracycline drugs such as mitoxantrone and prednisone could alleviate pain and improve the quality of life, but it also failed to significantly extend the patient’s survival period. In 2004, the US FDA approved docetaxel for the treatment of mCRPC, marking an important milestone in PCa chemotherapy (94,95). Tannock et al.’s phase III clinical trial showed that compared with mitoxantrone plus prednisone, docetaxel combined with prednisone performed better in extending the survival period of patients with advanced hormone-resistant PCa, relieving pain and reducing PSA levels (96). Unfortunately, almost all patients developed drug resistance within 6–8 months (97). To solve this problem, cabazitaxel (CAB), a second-generation semisynthetic taxane drug, was introduced for the treatment of patients with mCRPC who failed docetaxel therapy. In 2010, de Bono et al. conducted the TROPIC phase III clinical trial, which showed that cabazitaxel could effectively reduce the risk of PCa-related death, but hematological adverse events (neutropenia, leukopenia, and anemia) were also more common (98). In recent years, the combination of platinum drugs (such as carboplatin and cisplatin) and taxane drugs has shown potential. The literature review by Leal and García-Perdomo indicated that the clinical overall response rate and the PSA response rate for combination chemotherapy involving platinum and paclitaxel ranges from 23% to 78% and from 17% to 100%, respectively (99).

Kataoka et al.’s animal experiments showed that after 4 weeks of docetaxel treatment, the testosterone levels in the experimental group rats decreased; moreover, the ratio of intracranial pressure to mean arterial pressure (ICP/MAP) in the experimental group was significantly lower than that in the control group, indicating that docetaxel has the potential to induce ED (100). Unfortunately, there is currently no clear clinical evidence confirming that chemotherapy induces ED. A multicenter study involving 428 patients with PCa showed that 48% of patients experienced gonadal dysfunction when taking oral anticancer drugs (such as platinum and paclitaxel) (101). This suggests that chemotherapy drugs may indirectly affect EF by causing gonadal dysfunction. In addition, it was found that docetaxel may cause peripheral neuropathy, which may further lead to ED (102,103). Psychological problems such as anxiety and depression caused by chemotherapy, as well as side effects such as fatigue, may also degrade the overall health status of patients, indirectly leading to ED. More research directly investigating the impact of chemotherapy drugs on ED is required.

ADT and ED

In 1941, Huggins and Hodges first discovered the benefits of castration and estrogen injection for metastatic PCa, laying the foundation for ADT (104). Subsequently, gonadotropin-releasing hormone agonists (GnRH-As) became the mainstream drugs for ADT in the 1980s (105). Studies have shown that the overall survival rate of GnRH-A is comparable to that of orchiectomy (106). However, GnRH-As may initially cause a surge in testosterone, leading to disease progression. To prevent this reaction, Kuhn et al. used a combination of nilutamide and buserelin, significantly alleviating the symptoms caused by the surge in testosterone (107). In addition, gonadotropin-releasing hormone antagonists (GnRHs) represent an alternative choice for preventing the surge in testosterone and are superior to GnRH-A in terms of PSA progression and overall survival rate. However, almost all drug treatments will eventually lead to mCRPC, and thus androgen receptor antagonists are being increasingly favored in clinical treatment. The benefit of abiraterone in the treatment of castration-resistant PCa was confirmed in Ryan et al.’s multicenter, randomized, double-blind phase III clinical trial (108), which found that the median overall survival of the abiraterone combination group was 34.7 months, while the placebo group’s survival was 30.3 months; additionally, the radiographic progression-free survival was 16.5 and 8.3 months, respectively (109). Currently, androgen receptor antagonists are often used in combination with ADT, with the aim of more effectively inhibiting the growth of PCa by simultaneously reducing androgen levels and blocking its receptors. Emerging treatment strategies include biphasic androgen therapy (BAT) and rapid androgen testosterone cycling (RAC). BAT attempts to induce the periodic apoptosis of cancer cells by cycling between low androgen levels and high androgen levels. Meanwhile, RAC is a more aggressive method, involving more frequent changes in androgen levels, with the purpose of interfering with the adaptability and survival of cancer cells through rapid changes in hormone levels. However, there is currently a lack of extensive clinical trials evaluating the effectiveness of BAT and RAC in the treatment of PCa. Only Teply et al. reported that among 21 asymptomatic metastatic patients with CRPC who progressed after enzalutamide treatment, 15 had a 50% decrease in PSA levels after BAT treatment (110).

However, regardless of the improvements made to ADT, ED remains an inevitable complication. Gryzinski et al. reviewed the previous ADT literature and found that ADT increased the risk of ED by threefold (10). The reason lies for this is that testosterone is essential for maintaining normal EF, and low levels of testosterone cause vascular endothelial dysfunction and reduce penile blood flow, nitric oxide levels, and the enlargement of phosphodiesterase type 5, thereby affecting the quality and duration of erections (111). Exogenous testosterone supplementation may alleviate ED but is contraindicatory to ADT and may promote tumor development (112,113). In the future, ADT may incorporate the assessment of patients’ EF into its protocols, allowing for timely intervention in cases of ED caused by treatment.

Emerging treatment strategies for ED

Traditional treatment methods for ED include psychosocial intervention, phosphodiesterase type 5 inhibitors (PDE-5is), intracavernous injection of alprostadil, vacuum erection devices (VEDs), testosterone replacement therapy, and prosthesis implantation, but each of these methods has limitations and complications. For patients with organic lesions, the efficacy of psychosocial intervention therapy is poor. PDE-5is are only effective in 35% of ED patients, and their use in combination with nitrates or alpha-blockers can lead to hypotension (114-116). Intracavernous injection and VED are associated with poor patient compliance and a series of complications. The controversy surrounding testosterone replacement therapy is primarily centered on whether exogenous testosterone increases the risk of tumor progression or recurrence (117,118). Penile prosthesis implantation also involves complications such as mechanical failure, infection, penile perforation, and cross-infection (119,120). Therefore, more efficient and safer treatment plans urgently need to be developed.

Physical energy therapy

Physical energy, which can promote angiogenesis and reduce inflammatory responses, has been widely used in the treatment of ED (121). Extensive RCTs and prospective clinical studies have validated that low-intensity extracorporeal shock wave therapy (LI-ESWT) significantly improves the IIEF-5 scores in patients with ED (122,123). A systematic review and meta-analysis conducted by Clavijo et al., encompassing 7 studies and 602 subjects, revealed a significant finding: compared to those receiving sham therapy, males undergoing LI-ESWT demonstrated a statistically significant change in the International Index of Erectile Function-Erectile Function (IIEF-EF) scores from baseline to follow-up (124). A number of recent studies have shown that LI-ESWT can be effective in treating ED after RP and ED caused by RT for PCa. Examining the literature on patients with ED after RP and treated with LI-ESWT, Rho et al. found that the International Index of Erectile Function (IIEF) scores increased significantly after 3 or 4 months of LI-ESWT (125). In a study by Chan et al., LI-ESWT was used to treat patients with ED after RP for PCa, and the results showed that the IIEF-5 scores of patients increased from 10 to 19 (126). Lu et al.’s study demonstrated that, compared to monotherapy with tadalafil, the combination of LI-ESWT and tadalafil significantly improved patients’ IIEF-5 scores after 1 month (15.34±7.15 vs. 13.96±7.71, P=0.039) (127). In the treatment of ED after RP, low-intensity pulsed ultrasound (LIPUS) has been widely used since it is a noninvasive, low-energy method. Cui et al.’s RCT showed that LIPUS treatment for 12 weeks can significantly improve the IIEF-5 scores of patients with mild-to-moderate ED (128). According to research on animals, LIPUS can increase the amount of smooth muscle cells, stimulate the growth of Schwann cells, encourage peripheral nerve regeneration, and increase the expression of neuronal nitric oxide synthase (nNOS) in the cavernous and dorsal penile nerves, the expression of neurotrophic factor (BDNF) and vascular endothelial growth factor (VEGF), and ICP (129-133). A substantial body of research currently indicates that the combination of LIPUS with tadalafil therapy is more effective in improving EF than the use of PDE-5 inhibitors alone (134,135). Pulsed electromagnetic field stimulation therapy (PEMST) is another simple, noninvasive method. The Rainer team conducted a double-blind, placebo-controlled trial to evaluate the efficacy of PEMST on ED. Twenty patients with ED or orgasmic disorder were subjected to 10 minutes of PEMST at an intensity of 40% and a frequency of 20 Hz. The results demonstrated a significant improvement in EF following the therapy (136). Physical energy therapy methods have demonstrated significant potential in animal and clinical trials, providing effective and side effect-free options for the treatment of ED. However, current research is often constrained by small sample sizes, which limit the broad applicability of the results. The varying responses to treatment effects among different patients necessitate personalized assessments prior to therapy. There is an urgent need for large-scale clinical trials in the future to delve deeper into the impact of physical energy therapy on ED.

Penile tissue engineering and nerve transplantation therapy

An interdisciplinary field of study, tissue engineering combines biomedical engineering, cell biology, material science, and biotechnology to solve medical problems by building, repairing, or replacing damaged tissue. In the application of ED treatment, tissue engineering mainly involves the printing and implantation of artificial penile scaffolds. Kershen et al. implanted cultured human penile corpus cavernosum smooth muscle cells (CCSMCs) on degradable polyethylene glycol acid polymer scaffolds and then implanted the scaffolds into the penile corpus cavernosum, observing that the implants contributed to the formation of tissue blood vessels (137). Ji et al. inoculated purified muscle-derived stem cells (MDSCs) on decellularized penile corpus cavernosum collagen matrix (ACCM) and implanted them into the rabbit peritoneum. They found that through immunohistochemistry, scanning electron microscopy, and other means that MSCs can effectively increase the content of alpha smooth muscle actin (α-SMA) and NOS-positive cells in the penis (13). Future research will focus on optimizing the selection of cells and scaffold materials, enhancing the biocompatibility and functional integration of implants, and further conducting human trials, which may provide better therapeutic outcomes for patients with ED.

Nerve transplantation has shown significant potential in the treatment of ED after RP. Traditional treatment methods include the use of autologous nerve transplantation, such as sural nerve transplantation, which although effective, requires a second surgery and sacrifices healthy nerve tissue, bringing certain risks of complications (138). In recent years, researchers have explored the use of decellularized nerve scaffolds as an alternative, which removes the cellular components of the donor nerve, leaving only the extracellular matrix, thereby reducing immune rejection reactions (139). Although animal experiments have shown that decellularized nerve scaffolds can effectively promote nerve regeneration and restore EF, comparable to autologous nerve grafting, there is currently a lack of large-scale clinical trial data in humans to confirm the efficacy and safety of decellularized nerve scaffolds (140). Nerve transplantation also involves high surgical complexity and the need for a second surgery. Despite these challenges, nerve transplantation still points to a new direction for the treatment of ED, potentially reducing the burden on patients and improving therapeutic outcomes.

Stem cell and platelet-rich plasma (PRP) therapy

Undifferentiated or partially differentiated stem cells can self-renew and differentiate into other types of cells. Albersen et al. conducted an experiment where adipose tissue-derived stem cells (ADSCs) were injected into the corpora cavernosa of rats that had experienced cavernous nerve injury (CNI). The findings indicated several positive outcomes in the ADSC-injected group: an enhanced presence of nNOS in the dorsal penile nerves, a higher density of smooth muscle cells within the cavernous tissue, reduced apoptosis, diminished fibrosis, increased intracavernous pressure, and overall improved EF (141). Meanwhile, Sun et al. injected bone marrow mesenchymal stem cells (BM-MSCs) into the corpora cavernosa of the rats penis. The outcomes of the experiment demonstrated that BM-MSCs could upregulate the production of VEGF, nerve growth factor, and brain-derived neurotrophic factor (BDNF); protect nerve growth factor-positive nerve fibers; and raise the level of nNOS (142). Clinical trials also support the improvement of ED by stem cell therapy. Using various concentrations of BM-MNCs, Yiou et al. conducted a phase I–II clinical trial on 12 patients with ED after RP who were not responding to PDE-5i treatment. Evaluation of the results showed that patients in the treatment group had significantly improved sexual satisfaction and EF (143). Moreover, Haahr et al. reported that 8 of 17 patients who had undergone RP and were unresponsive to conventional therapy exhibited improvement in their EF after receiving an injection of ADSCs (144). Studies have shown that stem cell therapy for ED has almost no side effects, with only two clinical trials reporting minor effects at the injection site (transient redness, scrotal and penile hematoma), all of which resolved spontaneously (82,144,145). PRP is a plasma product extracted from autologous blood, rich in high concentrations of platelets and containing various cytokines, including platelet-derived growth factor (PDGF), transforming growth factor-beta (TGF-β), and vascular endothelial growth factor (VEGF). Clinical studies have shown that the level of PDGF in the serum of patients with ED is significantly elevated, while the levels of VEGF and TGF-β are approximately 1.5 times lower than in the normal population (146,147). Current literature supports that PRP treatment for ED is safe (148,149), and numerous preclinical (150) and clinical studies (151,152) have demonstrated the positive effects of PRP on EF. However, in a study by Masterson et al., after injecting PRP into the corpora cavernosa of ED patients, it was found that one month later, the average IIEF-5 score for those treated with PRP increased from 17.4 (95% confidence interval: 15.8–19.0) to 21 (17.9–24.0), while the placebo group’s scores increased from 18.6 (17.3–19.8) to 21.6 (19.1–24.1), with no significant difference observed between the two groups (153). This outcome may be related to individual differences in the severity of ED among patients, suggesting that different concentrations of PRP may be required for treatment. In the context of ED following PCa surgery, stem cell and PRP therapies still require in-depth research to determine the optimal preparation methods, dosage, and routes of administration. Furthermore, these treatment methods necessitate specific technical and equipment support, which to some extent limits their application in large-scale clinical trials.

Gene and targeted therapy

Gene therapy is a promising tool for the precision diagnosis and treatment for ED in the future. A study has shown that the injection of BDNF or VEGF can significantly restore the nNOS-positive nerve fibers in rats (154). BDNF not only promotes the formation of Schwann cell myelin sheaths but also repairs the cavernous nerves through the AKT/STAT pathway (155). Yin et al. used Ninj1 antibody (Ninj1-Ab) to treat rats with bilateral CNI. The results showed that Ninj1-Ab significantly increased the content of NOS and neurofilament in penile neurons by upregulating angiogenin-1 and downregulating angiogenin-2, inducing the proliferation of cavernous endothelial cells and the phosphorylation of Akt and endothelial NOS and reducing the apoptosis of endothelial cells. A dose of 2.5 µg/20 µL of Ninj1-Ab restored EF in 91% of rats with CNI-induced ED (CNIED) (156). RhoA signal transduction regulates smooth muscle contraction by activating Rho-associated protein kinase (ROCK), promoting the inactivation of myosin light chain phosphatase (MLCP) and inhibiting the sensitivity of smooth muscle cells to Ca2+ (157). Currently, there is controversy concerning the expression levels of ROCK1 and ROCK2 in CNIED (158,159). Sauzeau and Sopko’s respective studies indicated that Y-27632 (ROCK-1 inhibitor) can upregulate the expression of nNOS and endothelial NOS in the penis of CNIED rats, inhibit the apoptosis of nNOS-positive axons, and help protect the EF of rats (160,161). Gene therapy can precisely regulate the expression of key genes and has shown great potential in the field of ED treatment. However, its high cost and the need for personalized customization have limited the widespread adoption of gene therapy at this stage.

TCM and Chinese herbal extracts

In 1998, the TCM formula, BetterMAN, debuted in the American market, igniting a surge of interest in the use of TCM for the treatment of ED. Post-prostatectomy ED encompasses a variety of pathological pathways (162). While modern Western medications tend to focus on single-target therapies, TCM, with its multitargeted interventional approach, can more effectively address the complex etiologies of ED following PCa treatment. A substantial body of animal and clinical experiments has indicated that TCM and its extracts can treat ED (Table 2). Unfortunately, no multicenter RCTs have examined the use of TCM for ED in patients with PCa, and future large-scale, double-blinded, RCTs are necessary for validation in this context. TCM holds immense potential in the treatment of ED, and further exploration of ED treatment with TCM integrated with modern technological advancements may provide considerable benefit to patients.

Table 2

Traditional Chinese medicine and Chinese herbal extracts for the treatment of erectile dysfunction

Author Chinese medicine Experimental Results
Ye et al. (163) HongJing I (163) Rat HongJing I reduced RhoA and ROCK2 expression, increased smooth muscle content, decreased collagen area, and improved erectile function in rats with bilateral cavernous nerve injury
Gu et al. (164) YS-10 (a novel flavonoid derivative of icariside II) Rat YS-10 improved erectile function in rats with bilateral cavernous nerve injury by increasing the abundance of smooth muscle cells in penile tissue, elevating Reca-1 and endothelial NOS levels in penile endothelial cells, enhancing nNOS content in the dorsal nerve, and raising β-Catenin and cyclin D1 levels
Ye et al. (165) BHD Rat BHD improved erectile function in rats by increasing penile size and smooth muscle content, while reducing smooth muscle cell apoptosis. Fluorescent staining showed an increased α-SMA level, and Western blotting demonstrated reduced ROS, TGF-β1, type I collagen, Bax, PCa-JNK, and p-JNK expression
Xu et al. (166) Icariin Rat Icariin combined with sildenafil more effectively improved erectile function and increased nNOS content in the dorsal nerve and cGMP content in cavernous smooth muscle cells as compared to sildenafil alone in rats with bilateral cavernous nerve injury
Ye et al. (167) Salidroside Rat Salidroside improved erectile function in rats with BCNI by promoting protective autophagy, inhibiting apoptosis and fibrosis, and reducing the loss of neuronal components, endothelial cells, and smooth muscle cells in the penile corpus cavernosum
Zheng et al. (168) ICA II Rat Icariside II facilitated adipose tissue-derived stem cell differentiation into Schwann cells
Hu et al. (169) Angelica sinensis Rat An intrapelvic injection of Angelica sinensis solution could prevent rats’ cavernous nerve injury-induced reduction in NOS activity
Xiao et al. (170) Ligusticum chuanxiong Hort Rabbit Dong quai relaxed the smooth muscle of the penile corpus cavernosum by raising cAMP and cGMP levels
Wu et al. (44) EGb761 (Ginkgo biloba extract) Rat EGb761 protected the nerve fibers that express nNOS in rats
Liu et al. (171) Ginsenoside Rg3 Rat Rg3 lessened oxidative stress in the penile corpus cavernosum and halted the degeneration of penile dorsal nerve neurons
Zhao et al. (172) LBP Rat LBP could increase the amount of myelinated axons in corpus cavernosum neurons, decrease MDA levels, boost SOD, and GSH-Px expression, and improve erectile performance in rats

BHD, Buyang Huanwu decoction; ICA II, Icariside II; LBP, Lycium barbarum polysaccharides; NOS, nitric oxide synthase; nNOS, neural nitric oxide synthase; α-SMA, alpha smooth muscle actin; ROS, reactive oxygen species; TGF-β1, transforming growth factor beta 1; cGMP, cyclic guanosine monophosphate; BCNI, bilateral cavernous nerve injury; cAMP, cyclic adenosine monophosphate; MDA, malondialdehyde; SOD, superoxide dismutase.


Conclusions

With the development of medical technology, the treatment of PCa is gradually moving toward more precise and personalized plans for reducing the impact on patients’ EF. The popularization of PSA screening has improved the early diagnosis rate of PCa, making AS the preferred option for low-risk patients, as it significantly reduces the incidence of ED compared to active treatment. The optimization of equipment and technology for RT and cryotherapy, the improvement of surgical techniques, and the continuous evolution of intraoperative nerve monitoring technology have also mitigated the adverse effects on EF. Immunotherapy and chemotherapy may elevate the risk of ED, but determining their exact impact still requires additional clinical investigation. ADT therapy appears to invariably lead to ED, and further exploration of novel treatment plans is needed.

With the continuous advancement of biomedical technology, emerging treatment strategies have great potential. Physical energy therapy, such as LI-ESWT and PEMST, represent a new direction in the noninvasive treatment of ED, as it can promote angiogenesis and reduce inflammatory responses. Tissue engineering technology, especially that related to artificial penile scaffolds, is a source of optimism in the future treatment of ED. Nerve transplantation technology, through the application of autologous nerves or decellularized nerve scaffolds, has shown noteworthy potential in restoring EF after PCa surgery. Gene therapy, by precisely regulating molecular signaling pathways, may provide new targets and methods for the treatment of ED. Stem cell therapy, with its strong regenerative ability, is expected to significantly improve the nerve and tissue damage caused by surgery or other treatment methods, thereby restoring EF. PRP therapy aids in the functional recovery of patients with ED. In addition, the application of TCM in the treatment of ED has gradually received increased attention, and its unique advantage of multitargeted intervention may supplement Western therapy for patients with PCa. All emerging treatment modalities have their particular limitations, such as the need for personalized customization, high cost, and complex operation.

While this narrative review provides an in-depth analysis of the relationship between PCa treatment and ED, it acknowledges certain limitations. Specifically, our literature search may not have comprehensively covered all relevant studies, risking the omission of key literature. Furthermore, the subjectivity inherent in the review process may have influenced the selection and interpretation of the literature. Despite these limitations, we recognize the complex relationship between PCa treatment and ED, as well as the efficacy of emerging treatment methods on ED and the barriers to their large-scale clinical application. Therefore, we emphasize the development of more effective and comprehensive ED treatment plans for PCa patients through multidisciplinary collaboration and innovation, aiming to enhance their quality of life and improve their psychological well-being and intimate relationships.


Acknowledgments

Funding: This study was funded by the Zhejiang Province Traditional Chinese Medicine Prevention and Control Major Disease Research Plan (No. 2018ZY007), the National Natural Science Foundation of China (Nos. 82174376 and 82074433), and the Natural Science Foundation of Zhejiang Province (No. LGF20H270002).


Footnote

Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://tau.amegroups.com/article/view/10.21037/tau-24-514/rc

Peer Review File: Available at https://tau.amegroups.com/article/view/10.21037/tau-24-514/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tau.amegroups.com/article/view/10.21037/tau-24-514/coif). All authors report that this study was funded by the Zhejiang Province Traditional Chinese Medicine Prevention and Control Major Disease Research Plan (No. 2018ZY007), the National Natural Science Foundation of China (Nos. 82174376 and 82074433), and the Natural Science Foundation of Zhejiang Province (No. LGF20H270002). B.L. reports that he serves as the next chairman of the Urology Branch of the Chinese Association of Integrated Traditional Chinese and Western Medicine without compensation. The authors have no other conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Bergengren O, Pekala KR, Matsoukas K, et al. 2022 Update on Prostate Cancer Epidemiology and Risk Factors-A Systematic Review. Eur Urol 2023;84:191-206. [Crossref] [PubMed]
  2. Bray F, Laversanne M, Sung H, et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2024;74:229-63. [Crossref] [PubMed]
  3. Abumsimir B, Almahasneh I, Kasmi Y, et al. Prostate cancer and viral infections: epidemiological and clinical indications. Oncogenic Viruses 2023:263-72.
  4. Cantone E, Massanova M, Crocetto F, et al. The relationship between obstructive sleep apnoea and erectile dysfunction: An underdiagnosed link? A prospective cross-sectional study. Andrologia 2022;54:e14504. [Crossref] [PubMed]
  5. Pitta RM, de Lima Queiroga L, Louzada ACS, et al. What are the Main Risk Factors Associated with Erectile Dysfunction in the Elderly? A Cross-Sectional Study of 2436 Brazilian Elderly Men. Clin Interv Aging 2023;18:1047-54. [Crossref] [PubMed]
  6. Rew KT, Heidelbaugh JJ. Erectile Dysfunction. Am Fam Physician 2016;94:820-7.
  7. van der Wielen GJ, Mulhall JP, Incrocci L. Erectile dysfunction after radiotherapy for prostate cancer and radiation dose to the penile structures: a critical review. Radiother Oncol 2007;84:107-13. [Crossref] [PubMed]
  8. Latcu SC, Novacescu D, Buciu VB, et al. The Cavernous Nerve Injury Rat Model: A Pictorial Essay on Post-Radical Prostatectomy Erectile Dysfunction Research. Life (Basel) 2023;13:2337. [Crossref] [PubMed]
  9. Ranjbartehrani P, Ramirez DA, Schmidlin F, et al. Nerve Protection During Prostate Cryosurgery. Ann Biomed Eng 2023;51:538-49. [Crossref] [PubMed]
  10. Gryzinski GM, Fustok J, Raheem OA, et al. Sexual function in men undergoing androgen deprivation therapy. Androgens: Clinical Research and Therapeutics 2022;3:149-58.
  11. Pai R, Ory J, Delgado C, et al. Energy-Based Therapies for Erectile Dysfunction: Current and Future Directions. Urol Clin North Am 2021;48:603-10. [Crossref] [PubMed]
  12. Andersson KE, Christ GJ. Gene Therapy in Erectile Dysfunction: Dead or Alive? J Sex Med 2020;17:1587-9. [Crossref] [PubMed]
  13. Ji C, Min F, Liang W, et al. Construction of tissue-engineered corpus cavernosum with muscle-derived stem cells and transplantation in vivo. BJU Int 2011;107:1638-46. [Crossref] [PubMed]
  14. Leisegang K, Finelli R. Alternative medicine and herbal remedies in the treatment of erectile dysfunction: A systematic review. Arab J Urol 2021;19:323-39. [Crossref] [PubMed]
  15. Wong MC, Goggins WB, Wang HH, et al. Global Incidence and Mortality for Prostate Cancer: Analysis of Temporal Patterns and Trends in 36 Countries. Eur Urol 2016;70:862-74. [Crossref] [PubMed]
  16. Johansson JE, Andrén O, Andersson SO, et al. Natural history of early, localized prostate cancer. JAMA 2004;291:2713-9. [Crossref] [PubMed]
  17. Shill DK, Roobol MJ, Ehdaie B, et al. Active surveillance for prostate cancer. Transl Androl Urol 2021;10:2809-19. [Crossref] [PubMed]
  18. Newcomb LF, Schenk JM, Zheng Y, et al. Long-Term Outcomes in Patients Using Protocol-Directed Active Surveillance for Prostate Cancer. JAMA 2024;331:2084-93. [Crossref] [PubMed]
  19. Bokhorst LP, Valdagni R, Rannikko A, et al. A Decade of Active Surveillance in the PRIAS Study: An Update and Evaluation of the Criteria Used to Recommend a Switch to Active Treatment. Eur Urol 2016;70:954-60. [Crossref] [PubMed]
  20. Hirama H, Sugimoto M, Miyatake N, et al. Health-related quality of life in Japanese low-risk prostate cancer patients choosing active surveillance: 3-year follow-up from PRIAS-JAPAN. World J Urol 2021;39:2491-7. [Crossref] [PubMed]
  21. Cooperberg MR, Carroll PR. Trends in Management for Patients With Localized Prostate Cancer, 1990-2013. JAMA 2015;314:80-2. [Crossref] [PubMed]
  22. Loeb S, Curnyn C, Walter D, et al. Health state utilities among contemporary prostate cancer patients on active surveillance. Transl Androl Urol 2018;7:197-202. [Crossref] [PubMed]
  23. Weerakoon M, Papa N, Lawrentschuk N, et al. The current use of active surveillance in an Australian cohort of men: a pattern of care analysis from the Victorian Prostate Cancer Registry. BJU Int 2015;115:50-6. [Crossref] [PubMed]
  24. Donovan JL, Hamdy FC, Lane JA, et al. Patient-Reported Outcomes after Monitoring, Surgery, or Radiotherapy for Prostate Cancer. N Engl J Med 2016;375:1425-37. [Crossref] [PubMed]
  25. Alberti A, Nicoletti R, Castellani D, et al. Patient-reported Outcome Measures and Experience Measures After Active Surveillance Versus Radiation Therapy Versus Radical Prostatectomy for Prostate Cancer: A Systematic Review of Prospective Comparative Studies. Eur Urol Oncol 2024;S2588-9311(24)00138-X.
  26. Fujita K, Landis P, McNeil BK, et al. Serial prostate biopsies are associated with an increased risk of erectile dysfunction in men with prostate cancer on active surveillance. J Urol 2009;182:2664-9. [Crossref] [PubMed]
  27. Braun K, Ahallal Y, Sjoberg DD, et al. Effect of repeated prostate biopsies on erectile function in men on active surveillance for prostate cancer. J Urol 2014;191:744-9. [Crossref] [PubMed]
  28. Pearce SM, Wang CH, Victorson DE, et al. A Longitudinal Study of Predictors of Sexual Dysfunction in Men on Active Surveillance for Prostate Cancer. Sex Med 2015;3:156-64. [Crossref] [PubMed]
  29. Akbal C, Türker P, Tavukçu HH, et al. Erectile function in prostate cancer-free patients who underwent prostate saturation biopsy. Eur Urol 2008;53:540-4. [Crossref] [PubMed]
  30. Aktoz T, Kaplan M, Turan U, et al. 'Multimodal' approach to management of prostate biopsy pain and effects on sexual function: efficacy of levobupivacaine adjuvant to diclofenac sodium - a prospective randomized trial. Andrologia 2010;42:35-40. [Crossref] [PubMed]
  31. Murray KS, Bailey J, Zuk K, et al. A prospective study of erectile function after transrectal ultrasonography‐guided prostate biopsy. BJU international 2015;116:190-5. [Crossref] [PubMed]
  32. Tuncel A, Kirilmaz U, Nalcacioglu V, et al. The impact of transrectal prostate needle biopsy on sexuality in men and their female partners. Urology 2008;71:1128-31. [Crossref] [PubMed]
  33. Chong JJ, Van Hemelrijck M, Cahill D, et al. Serial transperineal sector prostate biopsies: impact on long-term erectile dysfunction. Ecancermedicalscience 2016;10:643. [Crossref] [PubMed]
  34. García Rojo E, García Gómez B, González Padilla DA, et al. Assessment of the influence of transrectal and transperineal prostate biopsies on erectile function: A prospective observational single-center study. Int J Urol 2019;26:1054-8. [Crossref] [PubMed]
  35. Calogero AE, Burgio G, Condorelli RA, et al. Epidemiology and risk factors of lower urinary tract symptoms/benign prostatic hyperplasia and erectile dysfunction. Aging Male 2019;22:12-9. [Crossref] [PubMed]
  36. Ozayar A, Zumrutbas AE, Yaman O. The relationship between lower urinary tract symptoms (LUTS), diagnostic indicators of benign prostatic hyperplasia (BPH), and erectile dysfunction in patients with moderate to severely symptomatic BPH. Int Urol Nephrol 2008;40:933-9. [Crossref] [PubMed]
  37. Algotar AM, Stratton SP, Ranger-Moore J, et al. Association of obesity and smoking with PSA and PSA velocity in men with prostate cancer. Am J Mens Health 2011;5:272-8. [Crossref] [PubMed]
  38. Tarantino G, Crocetto F, Vito CD, et al. Clinical factors affecting prostate-specific antigen levels in prostate cancer patients undergoing radical prostatectomy: a retrospective study. Future Sci OA 2021;7:FSO643. [Crossref] [PubMed]
  39. Beard CJ, Propert KJ, Rieker PP, et al. Complications after treatment with external-beam irradiation in early-stage prostate cancer patients: a prospective multiinstitutional outcomes study. J Clin Oncol 1997;15:223-9. [Crossref] [PubMed]
  40. Beckendorf V, Hay M, Rozan R, et al. Changes in sexual function after radiotherapy treatment of prostate cancer. Br J Urol 1996;77:118-23. [Crossref] [PubMed]
  41. Borghede G, Hedelin H. Radiotherapy of localised prostate cancer. Analysis of late treatment complications. A prospective study. Radiother Oncol 1997;43:139-46. [Crossref] [PubMed]
  42. Tendulkar RD, Commentary RE. Androgen Deprivation with Radiation Therapy Compared with Radiation Therapy Alone for Locally Advanced Prostatic Carcinoma: A Randomized Comparative Trial of the Radiation Therapy Oncology Group. Urology 2020;145:312. [Crossref] [PubMed]
  43. Turner SL, Adams K, Bull CA, et al. Sexual dysfunction after radical radiation therapy for prostate cancer: a prospective evaluation. Urology 1999;54:124-9. [Crossref] [PubMed]
  44. Wu YN, Liao CH, Chen KC, et al. Effect of Ginkgo biloba Extract (EGb-761) on Recovery of Erectile Dysfunction in Bilateral Cavernous Nerve Injury Rat Model. Urology 2015;85:1214.e7-1214.e15. [Crossref] [PubMed]
  45. Low NN, Vijayakumar S, Rosenberg I, et al. Beam's eye view based prostate treatment planning: is it useful? Int J Radiat Oncol Biol Phys 1990;19:759-68. [Crossref] [PubMed]
  46. Chang JH, Lim Joon D, Nguyen BT, et al. MRI scans significantly change target coverage decisions in radical radiotherapy for prostate cancer. J Med Imaging Radiat Oncol 2014;58:237-43. [Crossref] [PubMed]
  47. Horsley PJ, Aherne NJ, Edwards GV, et al. Planning magnetic resonance imaging for prostate cancer intensity-modulated radiation therapy: Impact on target volumes, radiotherapy dose and androgen deprivation administration. Asia Pac J Clin Oncol 2015;11:15-21. [Crossref] [PubMed]
  48. Brown MW, Brooks JP, Albert PS, et al. An analysis of erectile function after intensity modulated radiation therapy for localized prostate carcinoma. Prostate Cancer Prostatic Dis 2007;10:189-93. [Crossref] [PubMed]
  49. Zelefsky MJ, Cowen D, Fuks Z, et al. Long term tolerance of high dose three-dimensional conformal radiotherapy in patients with localized prostate carcinoma. Cancer 1999;85:2460-8. [Crossref] [PubMed]
  50. Murray J, Dean J, Mossop H, et al. Effect of dose and image guided radiation therapy (IGRT) on patient-reported sexual function in prostate radiation therapy. Int J Radiat Oncol Biol Phys 2016;96:E232.
  51. Incrocci L, Slob AK. Incidence, etiology, and therapy for erectile dysfunction after external beam radiotherapy for prostate cancer. Urology 2002;60:1-7.
  52. Ho CK, Bryant CM, Mendenhall NP, et al. Long-term outcomes following proton therapy for prostate cancer in young men with a focus on sexual health. Acta Oncol 2018;57:582-8. [Crossref] [PubMed]
  53. Incrocci L. Radiation therapy for prostate cancer and erectile (dys)function: the role of imaging. Acta Oncol 2005;44:673-8. [Crossref] [PubMed]
  54. Miller DC, Sanda MG, Dunn RL, et al. Long-term outcomes among localized prostate cancer survivors: health-related quality-of-life changes after radical prostatectomy, external radiation, and brachytherapy. J Clin Oncol 2005;23:2772-80. [Crossref] [PubMed]
  55. Goldstein I, Feldman MI, Deckers PJ, et al. Radiation-associated impotence. A clinical study of its mechanism. JAMA 1984;251:903-10. [Crossref] [PubMed]
  56. Zelefsky MJ, Eid JF. Elucidating the etiology of erectile dysfunction after definitive therapy for prostatic cancer. Int J Radiat Oncol Biol Phys 1998;40:129-33. [Crossref] [PubMed]
  57. Carrier S, Hricak H, Lee SS, et al. Radiation-induced decrease in nitric oxide synthase--containing nerves in the rat penis. Radiology 1995;195:95-9. [Crossref] [PubMed]
  58. Goldstein I, Goren A, Li VW, et al. Epidemiology Update of Erectile Dysfunction in Eight Countries with High Burden. Sex Med Rev 2020;8:48-58. [Crossref] [PubMed]
  59. Pellegrino F, Sjoberg DD, Tin AL, et al. Relationship Between Age, Comorbidity, and the Prevalence of Erectile Dysfunction. Eur Urol Focus 2023;9:162-7. [Crossref] [PubMed]
  60. Cooper IS, Hirose T. Application of cryogenic surgery to resection of parenchymal organs. N Engl J Med 1966;274:15-8. [Crossref] [PubMed]
  61. Soanes WA, Gonder MJ. Use of cryosurgery in prostatic cancer. J Urol 1968;99:793-7. [Crossref] [PubMed]
  62. Onik GM, Cohen JK, Reyes GD, et al. Transrectal ultrasound-guided percutaneous radical cryosurgical ablation of the prostate. Cancer 1993;72:1291-9. [Crossref] [PubMed]
  63. Asterling S, Greene DR. Prospective evaluation of sexual function in patients receiving cryosurgery as a primary radical treatment for localized prostate cancer. BJU Int 2009;103:788-92. [Crossref] [PubMed]
  64. Zhou JT, Fang DM, Xia S, et al. The incidence proportion of erectile dysfunction in patients treated with cryotherapy for prostate cancer: a meta-analysis. Clin Transl Oncol 2019;21:1152-8. [Crossref] [PubMed]
  65. Roberts CB, Jang TL, Shao YH, et al. Treatment profile and complications associated with cryotherapy for localized prostate cancer: a population-based study. Prostate Cancer Prostatic Dis 2011;14:313-9. [Crossref] [PubMed]
  66. Young HH. The early diagnosis and radical cure of carcinoma of the prostate. Being a study of 40 cases and presentation of a radical operation which was carried out in four cases. 1905. J Urol 2002;168:914-21. [Crossref] [PubMed]
  67. Millin T. Retropubic urinary surgery. E. & S. Livingstone; 1947.
  68. Walsh PC, Donker PJ. Impotence following radical prostatectomy: insight into etiology and prevention. J Urol 1982;128:492-7. [Crossref] [PubMed]
  69. Walsh PC, Lepor H, Eggleston JC. Radical prostatectomy with preservation of sexual function: anatomical and pathological considerations. Prostate 1983;4:473-85. [Crossref] [PubMed]
  70. Eggleston JC, Walsh PC. Radical prostatectomy with preservation of sexual function: pathological findings in the first 100 cases. J Urol 1985;134:1146-8. [Crossref] [PubMed]
  71. Guillonneau B, Vallancien G. Laparoscopic radical prostatectomy: initial experience and preliminary assessment after 65 operations. Prostate 1999;39:71-5. [Crossref] [PubMed]
  72. Binder J, Kramer W. Robotically-assisted laparoscopic radical prostatectomy. BJU Int 2001;87:408-10. [Crossref] [PubMed]
  73. Menon M, Tewari A, Peabody JO, et al. Vattikuti Institute prostatectomy, a technique of robotic radical prostatectomy for management of localized carcinoma of the prostate: experience of over 1100 cases. Urol Clin North Am 2004;31:701-17. [Crossref] [PubMed]
  74. Capogrosso P, Vertosick EA, Benfante NE, et al. Are We Improving Erectile Function Recovery After Radical Prostatectomy? Analysis of Patients Treated over the Last Decade. Eur Urol 2019;75:221-8. [Crossref] [PubMed]
  75. Moretti TBC, Magna LA, Reis LO. Erectile dysfunction criteria of 131,350 patients after open, laparoscopic, and robotic radical prostatectomy. Andrology 2024;12:1865-71. [Crossref] [PubMed]
  76. Yang Y, Wang J, Zhang D, et al. Efficacy of the transvesical approach for robotic-assisted radical prostatectomy via a bladder neck and prostate combined longitudinal incision for the treatment of localized prostate cancer. Front Surg 2022;9:1053140. [Crossref] [PubMed]
  77. Zhang H, Ning Z, Jia G, et al. Modified hood technique for single-port robot-assisted radical prostatectomy contributes to early recovery of continence. Front Surg 2023;10:1132303. [Crossref] [PubMed]
  78. Kim HL, Stoffel DS, Mhoon DA, et al. A positive caver map response poorly predicts recovery of potency after radical prostatectomy. Urology 2000;56:561-4. [Crossref] [PubMed]
  79. Klotz L, Herschorn S. Early experience with intraoperative cavernous nerve stimulation with penile tumescence monitoring to improve nerve sparing during radical prostatectomy. Urology 1998;52:537-42. [Crossref] [PubMed]
  80. Aron M, Kaouk JH, Hegarty NJ, et al. Second prize: preliminary experience with the Niris optical coherence tomography system during laparoscopic and robotic prostatectomy. J Endourol 2007;21:814-8. [Crossref] [PubMed]
  81. Fried NM, Rais-Bahrami S, Lagoda GA, et al. Imaging the cavernous nerves in the rat prostate using optical coherence tomography. Lasers Surg Med 2007;39:36-41. [Crossref] [PubMed]
  82. Boyette LB, Reardon MA, Mirelman AJ, et al. Fiberoptic imaging of cavernous nerves in vivo. J Urol 2007;178:2694-700. [Crossref] [PubMed]
  83. Fried NM, Lagoda GA, Scott NJ, et al. Noncontact stimulation of the cavernous nerves in the rat prostate using a tunable-wavelength thulium fiber laser. J Endourol 2008;22:409-13. [Crossref] [PubMed]
  84. Yadav R, Mukherjee S, Hermen M, et al. Multiphoton microscopy of prostate and periprostatic neural tissue: a promising imaging technique for improving nerve-sparing prostatectomy. J Endourol 2009;23:861-7. [Crossref] [PubMed]
  85. Hurwitz AA, Foster BA, Kwon ED, et al. Combination immunotherapy of primary prostate cancer in a transgenic mouse model using CTLA-4 blockade. Cancer Res 2000;60:2444-8.
  86. Graff JN, Chamberlain ED. Sipuleucel-T in the treatment of prostate cancer: an evidence-based review of its place in therapy. Core Evid 2015;10:1-10. [Crossref] [PubMed]
  87. Kantoff PW, Higano CS, Shore ND, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med 2010;363:411-22. [Crossref] [PubMed]
  88. Han Y, Liu D, Li L. PD-1/PD-L1 pathway: current researches in cancer. Am J Cancer Res 2020;10:727-42.
  89. Kwon ED, Drake CG, Scher HI, et al. Ipilimumab versus placebo after radiotherapy in patients with metastatic castration-resistant prostate cancer that had progressed after docetaxel chemotherapy (CA184-043): a multicentre, randomised, double-blind, phase 3 trial. Lancet Oncol 2014;15:700-12. [Crossref] [PubMed]
  90. Sadaghiani MS, Sheikhbahaei S, Werner RA, et al. A Systematic Review and Meta-analysis of the Effectiveness and Toxicities of Lutetium-177-labeled Prostate-specific Membrane Antigen-targeted Radioligand Therapy in Metastatic Castration-Resistant Prostate Cancer. Eur Urol 2021;80:82-94. [Crossref] [PubMed]
  91. Ryder M, Callahan M, Postow MA, et al. Endocrine-related adverse events following ipilimumab in patients with advanced melanoma: a comprehensive retrospective review from a single institution. Endocr Relat Cancer 2014;21:371-81. [Crossref] [PubMed]
  92. Mahzari M, Liu D, Arnaout A, et al. Immune checkpoint inhibitor therapy associated hypophysitis. Clin Med Insights Endocrinol Diabetes 2015;8:21-8. [Crossref] [PubMed]
  93. Eisenberger MA, Simon R, O'Dwyer PJ, et al. A reevaluation of nonhormonal cytotoxic chemotherapy in the treatment of prostatic carcinoma. J Clin Oncol 1985;3:827-41. [Crossref] [PubMed]
  94. Osoba D, Tannock IF, Ernst DS, et al. Health-related quality of life in men with metastatic prostate cancer treated with prednisone alone or mitoxantrone and prednisone. J Clin Oncol 1999;17:1654-63. [Crossref] [PubMed]
  95. Tannock IF, Osoba D, Stockler MR, et al. Chemotherapy with mitoxantrone plus prednisone or prednisone alone for symptomatic hormone-resistant prostate cancer: a Canadian randomized trial with palliative end points. J Clin Oncol 1996;14:1756-64. [Crossref] [PubMed]
  96. Tannock IF, de Wit R, Berry WR, et al. Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. N Engl J Med 2004;351:1502-12. [Crossref] [PubMed]
  97. Hashemi M, Zandieh MA, Talebi Y, et al. Paclitaxel and docetaxel resistance in prostate cancer: Molecular mechanisms and possible therapeutic strategies. Biomed Pharmacother 2023;160:114392. [Crossref] [PubMed]
  98. de Bono JS, Oudard S, Ozguroglu M, et al. Prednisone plus cabazitaxel or mitoxantrone for metastatic castration-resistant prostate cancer progressing after docetaxel treatment: a randomised open-label trial. Lancet 2010;376:1147-54. [Crossref] [PubMed]
  99. Leal F, García-Perdomo HA. Effectiveness of Platinum-Based Chemotherapy in Patients With Metastatic Prostate Cancer: Systematic Review and Meta-analysis. Clin Genitourin Cancer 2019;17:e627-44. [Crossref] [PubMed]
  100. Kataoka T, Sanagawa A, Suzuki J, et al. Influence of anticancer agents on sexual function: An in vivo study based on the US FDA Adverse Event Reporting System. Andrology 2022;10:166-78. [Crossref] [PubMed]
  101. Fleishman SB, Khan H, Homel P, et al. Testosterone levels and quality of life in diverse male patients with cancers unrelated to androgens. J Clin Oncol 2010;28:5054-60. [Crossref] [PubMed]
  102. Cheng HL, Molassiotis A, Leung AKT, et al. Docetaxel-Induced Peripheral Neuropathy in Breast Cancer Patients Treated with Adjuvant or Neo-Adjuvant Chemotherapy. Breast Care (Basel) 2021;16:269-75. [Crossref] [PubMed]
  103. Micheli L, Collodel G, Moretti E, et al. Redox imbalance induced by docetaxel in the neuroblastoma SH-SY5Y cells: a study of docetaxel-induced neuronal damage. Redox Rep 2021;26:18-28. [Crossref] [PubMed]
  104. Huggins C, Hodges CV. The effect of castration, of estrogen and of androgen injection on serum phosphatases in metastatic carcinoma of the prostate. Cancer Research 1941;1:293-7.
  105. Waxman J, Man A, Hendry WF, et al. Importance of early tumour exacerbation in patients treated with long acting analogues of gonadotrophin releasing hormone for advanced prostatic cancer. Br Med J (Clin Res Ed) 1985;291:1387-8. [Crossref] [PubMed]
  106. Sharifi N, Gulley JL, Dahut WL. Androgen deprivation therapy for prostate cancer. JAMA 2005;294:238-44. [Crossref] [PubMed]
  107. Kuhn JM, Billebaud T, Navratil H, et al. Prevention of the transient adverse effects of a gonadotropin-releasing hormone analogue (buserelin) in metastatic prostatic carcinoma by administration of an antiandrogen (nilutamide). N Engl J Med 1989;321:413-8. [Crossref] [PubMed]
  108. Ryan CJ, Smith MR, de Bono JS, et al. Abiraterone in metastatic prostate cancer without previous chemotherapy. N Engl J Med 2013;368:138-48. [Crossref] [PubMed]
  109. Queenan AM, Cassiday PK, Evangelista A. Pertactin-negative variants of Bordetella pertussis in the United States. N Engl J Med 2013;368:583-4. [Crossref] [PubMed]
  110. Teply BA, Wang H, Luber B, et al. Bipolar androgen therapy in men with metastatic castration-resistant prostate cancer after progression on enzalutamide: an open-label, phase 2, multicohort study. Lancet Oncol 2018;19:76-86. [Crossref] [PubMed]
  111. Castela A, Vendeira P, Costa C. Testosterone, endothelial health, and erectile function. ISRN Endocrinol 2011;2011:839149. [Crossref] [PubMed]
  112. Potosky AL, Knopf K, Clegg LX, et al. Quality-of-life outcomes after primary androgen deprivation therapy: results from the Prostate Cancer Outcomes Study. J Clin Oncol 2001;19:3750-7. [Crossref] [PubMed]
  113. Zhang XH, Morelli A, Luconi M, et al. Testosterone regulates PDE5 expression and in vivo responsiveness to tadalafil in rat corpus cavernosum. Eur Urol 2005;47:409-16; discussion 416. [Crossref] [PubMed]
  114. Fode M, Jensen CFS, Østergren PB. Sildenafil in postprostatectomy erectile dysfunction (perspective). Int J Impot Res 2019;31:61-4. [Crossref] [PubMed]
  115. Kallidonis P, Adamou C, Kotsiris D, et al. Combination Therapy with Alpha-blocker and Phosphodiesterase-5 Inhibitor for Improving Lower Urinary Tract Symptoms and Erectile Dysfunction in Comparison with Monotherapy: A Systematic Review and Meta-analysis. Eur Urol Focus 2020;6:537-58. [Crossref] [PubMed]
  116. Schwartz BG, Kloner RA. Drug interactions with phosphodiesterase-5 inhibitors used for the treatment of erectile dysfunction or pulmonary hypertension. Circulation 2010;122:88-95. [Crossref] [PubMed]
  117. Elena BW, Zachary M, Haritha P, et al. Current status of intracavernosal injection therapy in erectile dysfunction. Expert Opin Pharmacother 2023;24:925-33. [Crossref] [PubMed]
  118. Sultana A, Grice P, Vukina J, et al. Indications and characteristics of penile traction and vacuum erection devices. Nat Rev Urol 2022;19:84-100. [Crossref] [PubMed]
  119. Chung E, Van CT, Wilson I, et al. Penile prosthesis implantation for the treatment for male erectile dysfunction: clinical outcomes and lessons learnt after 955 procedures. World J Urol 2013;31:591-5. [Crossref] [PubMed]
  120. Wilson SK, Zumbe J, Henry GD, et al. Infection reduction using antibiotic-coated inflatable penile prosthesis. Urology 2007;70:337-40. [Crossref] [PubMed]
  121. Gruenwald I, Spector A, Shultz T, et al. The beginning of a new era: treatment of erectile dysfunction by use of physical energies as an alternative to pharmaceuticals. Int J Impot Res 2019;31:155-61. [Crossref] [PubMed]
  122. Kennady EH, Bryk DJ, Ali MM, et al. Low-intensity shockwave therapy improves baseline erectile function: a randomized sham-controlled crossover trial. Sex Med 2023;11:qfad053. [Crossref] [PubMed]
  123. Patel P, Katz J, Lokeshwar SD, et al. Phase II Randomized, Clinical Trial Evaluating 2 Schedules of Low-Intensity Shockwave Therapy for the Treatment of Erectile Dysfunction. Sex Med 2020;8:214-22. [Crossref] [PubMed]
  124. Clavijo RI, Kohn TP, Kohn JR, et al. Effects of Low-Intensity Extracorporeal Shockwave Therapy on Erectile Dysfunction: A Systematic Review and Meta-Analysis. J Sex Med 2017;14:27-35. [Crossref] [PubMed]
  125. Rho BY, Kim SH, Ryu JK, et al. Efficacy of Low-Intensity Extracorporeal Shock Wave Treatment in Erectile Dysfunction Following Radical Prostatectomy: A Systematic Review and Meta-Analysis. J Clin Med 2022;11:2775. [Crossref] [PubMed]
  126. Chan CW, Wong CH. Treatment of radiation-induced erectile dysfunction with low-intensity extracorporeal shock wave: A case report. Australasian Medical Journal (Online) 2017;10:901-3.
  127. Lu M, Huang Y, Chen Y, et al. (281) Low-Intensity Shockwave Therapy Combined with Tadalafil 5mg Daily has Better Efficacy for Mild and Moderate Erectile Dysfunction: The Preliminary Results of a Multicenter Clinical Study in China. The Journal of Sexual Medicine 2024;21:qdae002.
  128. Cui W, Li H, Guan R, et al. Efficacy and safety of novel low-intensity pulsed ultrasound (LIPUS) in treating mild to moderate erectile dysfunction: a multicenter, randomized, double-blind, sham-controlled clinical study. Transl Androl Urol 2019;8:307-19. [Crossref] [PubMed]
  129. Chang CJ, Hsu SH, Lin FT, et al. Low-intensity-ultrasound-accelerated nerve regeneration using cell-seeded poly(D,L-lactic acid-co-glycolic acid) conduits: an in vivo and in vitro study. J Biomed Mater Res B Appl Biomater 2005;75:99-107. [Crossref] [PubMed]
  130. Lei H, Xin H, Guan R, et al. Low-intensity Pulsed Ultrasound Improves Erectile Function in Streptozotocin-induced Type I Diabetic Rats. Urology 2015;86:1241.e11-8. [Crossref] [PubMed]
  131. Li Z, Ye K, Yin Y, et al. Low-intensity pulsed ultrasound ameliorates erectile dysfunction induced by bilateral cavernous nerve injury through enhancing Schwann cell-mediated cavernous nerve regeneration. Andrology 2023;11:1188-202. [Crossref] [PubMed]
  132. Su WS, Wu CH, Chen SF, et al. Transcranial ultrasound stimulation promotes brain-derived neurotrophic factor and reduces apoptosis in a mouse model of traumatic brain injury. Brain Stimul 2017;10:1032-41. [Crossref] [PubMed]
  133. Tsuang YH, Liao LW, Chao YH, et al. Effects of low intensity pulsed ultrasound on rat Schwann cells metabolism. Artif Organs 2011;35:373-83. [Crossref] [PubMed]
  134. Chung E, Lee J, Liu CC, et al. Clinical Practice Guideline Recommendation on the Use of Low Intensity Extracorporeal Shock Wave Therapy and Low Intensity Pulsed Ultrasound Shock Wave Therapy to Treat Erectile Dysfunction: The Asia-Pacific Society for Sexual Medicine Position Statement. World J Mens Health 2021;39:1-8. [Crossref] [PubMed]
  135. Gao QQ, Wang J, Li DS, et al. Efficacy and safety of low-intensity pulsed ultrasound (LIPUS) combined with tadalafil in the treatment of severe erectile dysfunction: a retrospective cohort study. Transl Androl Urol 2024;13:2045-54. [Crossref] [PubMed]
  136. Pelka RB, Jaenicke C, Gruenwald J. Impulse magnetic-field therapy for erectile dysfunction: a double-blind, placebo-controlled study. Adv Ther 2002;19:53-60. [Crossref] [PubMed]
  137. Kershen RT, Yoo JJ, Moreland RB, et al. Reconstitution of human corpus cavernosum smooth muscle in vitro and in vivo. Tissue Eng 2002;8:515-24. [Crossref] [PubMed]
  138. Kim ED, Scardino PT, Hampel O, et al. Interposition of sural nerve restores function of cavernous nerves resected during radical prostatectomy. J Urol 1999;161:188-92.
  139. Gulati AK. Evaluation of acellular and cellular nerve grafts in repair of rat peripheral nerve. J Neurosurg 1988;68:117-23. [Crossref] [PubMed]
  140. Connolly SS, Yoo JJ, Abouheba M, et al. Cavernous nerve regeneration using acellular nerve grafts. World J Urol 2008;26:333-9. [Crossref] [PubMed]
  141. Albersen M, Fandel TM, Lin G, et al. Injections of adipose tissue-derived stem cells and stem cell lysate improve recovery of erectile function in a rat model of cavernous nerve injury. J Sex Med 2010;7:3331-40. [Crossref] [PubMed]
  142. Sun C, Lin H, Yu W, et al. Neurotrophic effect of bone marrow mesenchymal stem cells for erectile dysfunction in diabetic rats. Int J Androl 2012;35:601-7. [Crossref] [PubMed]
  143. Yiou R, Hamidou L, Birebent B, et al. Safety of Intracavernous Bone Marrow-Mononuclear Cells for Postradical Prostatectomy Erectile Dysfunction: An Open Dose-Escalation Pilot Study. Eur Urol 2016;69:988-91. [Crossref] [PubMed]
  144. Haahr MK, Jensen CH, Toyserkani NM, et al. Safety and Potential Effect of a Single Intracavernous Injection of Autologous Adipose-Derived Regenerative Cells in Patients with Erectile Dysfunction Following Radical Prostatectomy: An Open-Label Phase I Clinical Trial. EBioMedicine 2016;5:204-10. [Crossref] [PubMed]
  145. Levy JA, Marchand M, Iorio L, et al. Determining the Feasibility of Managing Erectile Dysfunction in Humans With Placental-Derived Stem Cells. J Am Osteopath Assoc 2016;116:e1-5. [Crossref] [PubMed]
  146. Khodamoradi K, Dullea A, Golan R, et al. Growth Factor Concentration Can Vary in Platelet-Rich Plasma from Men with Erectile Dysfunction. J Sex Med 2022;19:S103. [Crossref] [PubMed]
  147. Khodamoradi K, Dullea A, Golan R, et al. Platelet Rich Plasma (PRP) Growth Factor Concentration Varies in Men With Erectile Dysfunction. J Sex Med 2022;19:1488-93. [Crossref] [PubMed]
  148. Fazekas D, Campbell K, Ledesma B, et al. Platelet-rich plasma for erectile dysfunction: a review of the current research landscape. Sex Med Rev 2023;11:369-74. [Crossref] [PubMed]
  149. Wu YN, Liao CH, Chen KC, et al. CXCL5 Cytokine Is a Major Factor in Platelet-Rich Plasma's Preservation of Erectile Function in Rats After Bilateral Cavernous Nerve Injury. J Sex Med 2021;18:698-710. [Crossref] [PubMed]
  150. Huang YC, Wu CT, Chen MF, et al. Intracavernous Injection of Autologous Platelet-Rich Plasma Ameliorates Hyperlipidemia-Associated Erectile Dysfunction in a Rat Model. Sex Med 2021;9:100317. [Crossref] [PubMed]
  151. Matz EL, Pearlman AM, Terlecki RP. Safety and feasibility of platelet rich fibrin matrix injections for treatment of common urologic conditions. Investig Clin Urol 2018;59:61-5. [Crossref] [PubMed]
  152. Ruffo A, Stanojevic N, Iacono F, et al. 529 Treating erectile dysfunction with a combination of low-intensity shock waves (LISW) and platelet-rich plasma (PRP) injections. J Sex Med 2018;15:S318-S9.
  153. Masterson TA, Molina M, Ledesma B, et al. Platelet-rich Plasma for the Treatment of Erectile Dysfunction: A Prospective, Randomized, Double-blind, Placebo-controlled Clinical Trial. J Urol 2023;210:154-61. [Crossref] [PubMed]
  154. Campbell JD, Burnett AL. Neuroprotective and Nerve Regenerative Approaches for Treatment of Erectile Dysfunction after Cavernous Nerve Injury. Int J Mol Sci 2017;18:1794. [Crossref] [PubMed]
  155. Bella AJ, Lin G, Tantiwongse K, et al. Brain-derived neurotrophic factor (BDNF) acts primarily via the JAK/STAT pathway to promote neurite growth in the major pelvic ganglion of the rat: part I. J Sex Med 2006;3:815-20. [Crossref] [PubMed]
  156. Yin GN, Kim WJ, Jin HR, et al. Nerve injury-induced protein 1 (Ninjurin-1) is a novel therapeutic target for cavernous nerve injury-induced erectile dysfunction in mice. J Sex Med 2013;10:1488-501. [Crossref] [PubMed]
  157. Ding Z, Shen X, Li Y. Association of nNOS and Rho-kinase with age-associated erectile dysfunction in Sprague-Dawley rats. Exp Ther Med 2017;13:1133-6. [Crossref] [PubMed]
  158. Song SH, Park K, Kim SW, et al. Involvement of Rho-Kinase/LIM Kinase/Cofilin Signaling Pathway in Corporal Fibrosis after Cavernous Nerve Injury in Male Rats. J Sex Med 2015;12:1522-32. [Crossref] [PubMed]
  159. Uvin P, Albersen M, Bollen I, et al. Additive effects of the Rho kinase inhibitor Y-27632 and vardenafil on relaxation of the corpus cavernosum tissue of patients with erectile dysfunction and clinical phosphodiesterase type 5 inhibitor failure. BJU Int 2017;119:325-32. [Crossref] [PubMed]
  160. Sauzeau V, Le Jeune H, Cario-Toumaniantz C, et al. Cyclic GMP-dependent protein kinase signaling pathway inhibits RhoA-induced Ca2+ sensitization of contraction in vascular smooth muscle. J Biol Chem 2000;275:21722-9. [Crossref] [PubMed]
  161. Sopko NA, Hannan JL, Bivalacqua TJ. Understanding and targeting the Rho kinase pathway in erectile dysfunction. Nat Rev Urol 2014;11:622-8. [Crossref] [PubMed]
  162. Nandipati KC, Raina R, Agarwal A, et al. Erectile dysfunction following radical retropubic prostatectomy: epidemiology, pathophysiology and pharmacological management. Drugs Aging 2006;23:101-17. [Crossref] [PubMed]
  163. Ye MY, Zhao F, Ma K, et al. Effect of HongJing I in Treating Erectile Function and Regulating RhoA Pathway in a Rat Model of Bilateral Cavernous Nerve Injury. Evid Based Complement Alternat Med 2019;2019:1083737. [Crossref] [PubMed]
  164. Gu SJ, Li M, Yuan YM, et al. A novel flavonoid derivative of icariside II improves erectile dysfunction in a rat model of cavernous nerve injury. Andrology 2021;9:1893-901. [Crossref] [PubMed]
  165. Ye MY, Zhao F, Ma K, et al. Buyang Huanwu Decoction Ameliorates Damage of Erectile Tissue and Function Following Bilateral Cavernous Nerve Injury. Chin J Integr Med 2023;29:791-800. [Crossref] [PubMed]
  166. Xu Y, Xin H, Wu Y, et al. Effect of icariin in combination with daily sildenafil on penile atrophy and erectile dysfunction in a rat model of bilateral cavernous nerves injury. Andrology 2017;5:598-605. [Crossref] [PubMed]
  167. Ye M, Zhao F, Ma K, et al. Enhanced effects of salidroside on erectile function and corpora cavernosa autophagy in a cavernous nerve injury rat model. Andrologia 2021;53:e14044. [Crossref] [PubMed]
  168. Zheng T, Zhang TB, Wang CL, et al. Icariside II Promotes the Differentiation of Adipose Tissue-Derived Stem Cells to Schwann Cells to Preserve Erectile Function after Cavernous Nerve Injury. Mol Cells 2018;41:553-61. [Crossref] [PubMed]
  169. Hu W, Hu L, Cheng B. Effect of angelica sinensis on NOS activity in penile tissues of rats after crushing injury to cavernous nerves. Zhonghua Nan Ke Xue 2001;7:29-31.
  170. Xiao HJ, Wang T, Chen J, et al. Chuanxiongzine relaxes isolated corpus cavernosum strips and raises intracavernous pressure in rabbits. Int J Impot Res 2010;22:120-6. [Crossref] [PubMed]
  171. Liu T, Peng YF, Jia C, et al. Ginsenoside Rg3 improves erectile function in streptozotocin-induced diabetic rats. J Sex Med 2015;12:611-20. [Crossref] [PubMed]
  172. Zhao Z, Yu H, Ding X, et al. The effect of Lycium barbarum polysaccharides on erectile function recovery in a rat model of cavernous nerve injury. Journal of Medicinal Plants Research 2013;6:2867-72.
Cite this article as: Zhang X, Pan J, Xu R, Fu Y, Lv B. Erectile dysfunction associated with prostate cancer treatment and therapeutic advances: a narrative review. Transl Androl Urol 2024;13(11):2625-2643. doi: 10.21037/tau-24-514

Download Citation