Skip to main content

Recent advances and future perspectives in the therapeutics of prostate cancer

Abstract

Prostate cancer (PC) is one of the most common cancers in males and the fifth leading reason of death. Age, ethnicity, family history, and genetic defects are major factors that determine the aggressiveness and lethality of PC. The African population is at the highest risk of developing high-grade PC. It can be challenging to distinguish between low-risk and high-risk patients due to the slow progression of PC. Prostate-specific antigen (PSA) is a revolutionary discovery for the identification of PC. However, it has led to an increase in over diagnosis and over treatment of PC in the past few decades. Even if modifications are made to the standard PSA testing, the specificity has not been found to be significant. Our understanding of PC genetics and proteomics has improved due to advances in different fields. New serum, urine, and tissue biomarkers, such as PC antigen 3 (PCA3), have led to various new diagnostic tests, such as the prostate health index, 4K score, and PCA3. These tests significantly reduce the number of unnecessary and repeat biopsies performed. Chemotherapy, radiotherapy, and prostatectomy are standard treatment options. However, newer novel hormone therapy drugs with a better response have been identified. Androgen deprivation and hormonal therapy are evolving as new and better options for managing hormone-sensitive and castration-resistant PC. This review aimed to highlight and discuss epidemiology, various risk factors, and developments in PC diagnosis and treatment regimens.

Introduction

Cancer is characterized by uncontrolled proliferation, in which the cell loses its regulated division, differentiation, and apoptosis. Cancer is a global burden and a leading cause of mortality and decreasing life expectancy worldwide [1]. According to the International Agency for Research on Cancer, nearly 19.3 million new cancer cases and approximately 10 million cancer-related deaths are expected by 2020. The five most commonly diagnosed cancers are female breast cancer (11.7%), lung cancer (11.4%), colorectal cancer (10%), prostate cancer (PC; 7.3%), and stomach cancer (5.6%) [2].

PC is a non-cutaneous cancer most commonly seen in males after 50 years of age, affecting nearly 1.6 million individuals with more than 3,00,000 deaths worldwide [3]. Reports showed that it is the second most commonly identified cancer in males and the fifth major cause of cancer-related death [4]. In India, PC was one of the most frequently detected cancers in 2020, with 41,532 new cases, accounting for 5.7% of the total cancer cases in men, with one in 125 men at risk of being diagnosed with PC [5]. The higher incidence rate of PC in the last few decades has been mainly due to increased prostate-specific antigen (PSA) screening, leading to a decrease in mortality among diagnosed cases [6,7,8,9,10]. Early diagnosis and improvements in treatment strategies are major factors in the decline in mortality rates [11]. The prostate is a walnut-sized gland present in the male pelvis, which secretes seminal fluid and releases an alkaline solution that helps sperm to survive in the acidic environment of the vagina and helps in nourishing and transporting sperm [12]. Some of the different types of PC found in men include adenocarcinomas, squamous cell carcinomas, transitional cell carcinomas, neuroendocrine tumors, and prostate sarcomas. The most frequent type of PC is adenocarcinoma (90–95%). Age is the most common risk factor for the development of PC, as the incidence rate increases in those over 50 years old. Other associated risk factors includes race and ethnicity, diet, obesity, family history, and smoking [14].

The clinical symptoms of PC depend on the cancer stage, that is, whether the PC is early or advanced. The most commonly observed symptoms include urinary tract signs and symptoms such as painful and poor urinary stream, frequent urination, erectile dysfunction, painful ejaculation, and hematuria [15]. Metastasis of PC to the vertebrae can lead to Pott’s disease, with chronic back and hip pain reported in patients. Furthermore, urinary incontinence has been observed after radical prostatectomy in the early stages of PC [16,17,18]. For the early detection of PC, PSA biomarker screening has been recommended for the men aged 55–69 years, with a digital rectal examination (DRE) performed in patients with a high PSA level [19]. A systemic prostate biopsy can then be conducted for the final analysis of adenocarcinoma using transrectal ultrasound (TRUS) or transperineal biopsy or multiparametric magnetic resonance imaging (mpMRI) or targeted MRI-ultrasound fusion biopsy [20,21,22]. The Gleason grading system is used to grade the tumor, which helps the patient choose the correct therapeutic options [23]. A new diagnostic approach uses mp-MRI before the biopsy, which helps detect PC in biopsy-naive patients [24]. Non-invasive diagnostic tools, such as liquid biopsy, can also be used for detection of PC [25]. These diagnostic techniques are mainly adopted by physicians for tumor detection.

The primary methods for curing PC are pharmaceutical and surgical treatment. In recent years, inhibition of the androgen signaling pathway has emerged as a major therapeutic approach against tumors, where androgen levels are reduced by using hormones. Androgen deprivation therapy (ADT) is the name given to this treatment [26]. Hormone therapy such as ADT is highly effective in the treatment of metastatic hormone-sensitive PC (mHSPC), which leads to form metastatic castration-resistant PC (mCRPC) [27]. The approved drugs used for ADT are abiraterone acetate and enzalutamide. The drugs used in chemotherapy include docetaxel, cabazitaxel, mitoxantrone, and radium-223, which are radioisotopes used for cancer treatment [28]. The drugs got approval from United States Food and Drug Administration (US FDA) for the medication of PC has been listed in Table 1. The timeline for the development of these drugs and their respective approval year has been presented in Fig. 1. Surgical treatments are very successful in treating localized PC [29]. However, survivor of PC can have an adverse impact on the quality of life (QoL) of the survivor [30]. The most likely observed problem is depression [31] and many experiences of physical and sexual dysfunction [32].

Table 1 Different classes of drugs presently used to cure PC
Fig. 1
figure 1

Timeline demonstrating evolution in the treatment regimen for PC

This review intends to provide an overview of PC, considering aspects such as the risk factors, clinical representations, different methods of diagnosis, treatment, and management, and the QoL of patients with PC. As PC has recently emerged as a global burden, extensive research and studies are required to better understand this disease so that novel diagnostic and therapeutic approaches can be identified to reduce mortality and improve patient QoL.

Epidemiology

PC has always been the most common malignancy in men in the past few decades. In 2020, there were 1.41 million new PC cases globally, accounting for 7.3% of all cancer cases [2]. In 2021, there are 248,530 new PC cases reported and a sum of 34,130 deaths [45]. Furthermore, by 2040, these trends are estimated to increase to 2.43 million new cases and 740,000 deaths worldwide [46]. The Caribbean, Western and Northern Europe, North America, Australia, New Zealand, and Southern Africa had the highest incidence rates. At the same time, Northern Africa and Asia had the lowest incidence rates [2]. A major contributor to these varying PC incidence rates is variations in diagnostic practice. With the introduction of PSA screening in 1990s, a remarkable rise in PC incidence rates was noted in the United States, Australia, and Canada [3]. According to an investigation, one in every eight men has a risk of developing PC [45], with increasing age. For men below 50 years of age, the probability of developing PC has been reported to be low in comparison to those aged 50–59 and 60–69 years [47].

Approximately one in every 41 men dies because of PC [45]. The mortality trends did not show as much variation as the incidence rates. The highest rate of mortality are observed in regions such as sub-Saharan Africa, the Caribbean, and Micronesia/Polynesia [2]. In contrast, low mortality has been observed in South Central and Eastern Asia [3]. Currently, there are more than 3.2 million survivors of PC in the United States [48]. There has been a remarkable decline in PC incidence rates in the last decade. Due to concerns regarding the over diagnosis and over treatment of PC.

Major risk factors

Major risk factors associated with the incidence of PC includes the aging, race, family history, and genetic factors. Besides these the lifestyle factors such as diet, obesity, and smoking has also been reported to be associated with the prevalence of PC [49].

Age

PC is commonly diagnosed in men above 60 years of age. The United States Preventative Services Task Force (USPSTF) issued Grade ‘D’ recommendations for discouraging PSA use for men above 75 years of age in 2008 and all men in 2012. Subsequently, PSA screening was reduced by 25–30%, resulting in a significant decline in incidence rate of PC in the United States. In 2018, the USPSTF issued a new recommendation, stating periodic PSA screening for men aged 55–69 years. In contrast, PSA screening is discouraged in aged men above 70 years [50]. The median age of diagnosis has been reported to be 67 years, with more than 30% of deaths reported in those aged 75–84 years [48].

Race and ethnicity

Racial disparity has been observed in PC. African black men have a higher incidence rate than white men [51]. A report from the United States of America showed that African Americans have 58% more incidence and 144% more mortality than white men of European ancestry. In contrast, Hispanics have been reported to 14% lower incidence and 17% lower mortality than white men [52]. The reason for this disparity in PC is socioeconomic conditions and biological factors [53]. African American men have chromosome 8q24, which has been reported to be associated with an increased risk of PC. They also have tumor suppressor genes, such as EphB2, which increase cancer risk [54, 55].

Family history and genetic factors

Family history plays a role in the development of PC. The risk of developing PC is higher in men who have a first-degree relative with PC. Furthermore, the risk is higher if the relative is a brother [56]. Genetic factors contribute to nearly 40% of the risk of developing PC. Mutations in BRCA1 and BRCA2 increase the risk of PC. A mutation in the BRCA2 gene confers 8.6 times increased risk of PC in men aged < 65 years [57, 58]. Other rare mutations in PC include HOXB13, NBS1, and CHEK2 [59].

Lifestyle factors

Major lifestyle factors that influence the development of PC include diet, obesity, and smoking.

Diet and obesity

Diet plays a vital role in the incidence of PC, and there are certain food products that have a higher risk for PC, such as saturated animal fat, red meat, and dairy proteins. However, some dietary products decrease the development of cancer, such as soya, green tea, tomatoes, and lycopene [4, 60]. According to a survey, a sedentary lifestyle is related to increased PSA levels [61]. Some meta-analyses have reported a clear association between obesity and an enhanced incidence of PC independent of body mass index increases [62]. Obesity has been reported to be associated with a high risk of aggressive PC. The involvement of certain biological mechanisms, such as the development of insulin resistance due to physical inactivity, sex steroid hormones, and changes in metabolism, supports an association between obesity and PC [63].

Smoking

Smoking has been associated with the incidence of all types of cancers. Several reports suggested that smoking is associated with a greater number of mortalities due to PC. It also depends on the number of cigarettes smoked per day [64]. A California report stated that the rate of smoking is declining by 3.5% per year, resulting in a 2.5% decrease in PC death, suggesting that smoking reduction is beneficial for the decline in cancer mortality [65].

Novel diagnostic approaches

The traditionally used PSA test for the screening and diagnosis of PC has various limitations, including its low specificity. These challenges have led to over diagnosis and over treatment of low-risk PC patients. Thus, new diagnostic methods that are cost-efficient and non-invasive are needed to differentiate between aggressive and slow PC and to decrease the performance of excessive biopsies [66].

In the last decade, an increasing knowledge of the genetics and molecular biology of PC has led to the identification of several biomarkers that overcome the existing limitations of PSA. These newer biomarker-based methods with more specificity have been shown to be significantly better. Thus, these biomarker-based methods can help reduce the overtreatment and overdiagnosis of PC [66]. These biomarkers are used at various stages of decision making, including screening, after a positive biopsy for risk stratification, after a negative biopsy to determine whether to consider a repeat biopsy, and to monitor prognosis after treatment or in those suspected of recurrence to determine whether additional treatment is required. Despite these potential benefits of biomarkers, most are not currently used in clinical practice because there is a lack of clinically significant support validating their utility and benefits [67]. Nevertheless, we will now discuss some biomarker-based diagnostic tests that have demonstrated clinical significance.

Novel biomarker-based diagnostic approaches

Biomarkers are used for various reasons, such as screening, diagnosis, risk stratification, and prognosis. A concise overview of various biomarker-based tests used in the diagnosis of PC can be seen in Table 2.

Table 2 Clinically significant different biomarkers for the diagnosis of PC

Serum-based biomarkers

Prostate health index (PHI)

The PHI test is a serum-based analysis, developed by Beckman Coulter Inc. and the National Cancer Institute-Early Detection Research Network. The biomarkers included in the PHI are [-2] proPSA, free PSA (fPSA), and total PSA (tPSA). These individual values are then subjected to the formula [-2] proPSA/fPSA × √tPSA to generate a score that can differentiate between benign and malignant PCs, decreasing the performance of unnecessary biopsies [68]. Following this equation, high-risk patients are those with increased tPSA and [-2] proPSA levels and decreased fPSA levels. Thus, owing to the high risk of aggressive PC, patients with high PHI scores are suggested to undergo less invasive biopsies [76]. In 2012, the US FDA authorized the commercial use of PHI for patients > 50 years of age with PSA levels of 4–10 ng/mL and negative DRE reports [68].

Catalona et al. conducted a multicenter trial in the United States on the clinical significance of PHI in 892 men considered for biopsy with total PSA between 2 and 10 ng/mL and normal DRE. Their results indicated that PHI had significantly higher specificity with 80–95% sensitivity than PSA and %fPSA alone. The AUC values of PHI were 0.70, whereas PSA and % fPSA have AUCs of 0.53 and 0.65, respectively [77].

Another study of two cohort groups, including 561 and 395 men with no prior biopsy, demonstrated that PHI could predict the possibility of high-grade PC (AUC -0.81 and 0.78). PHI had a specificity of 36% when the sensitivity settings were at 95%, which is comparatively high with the specificity of fPSA and tPSA, i.e., 19.4% and 17.2%, respectively. A PHI threshold value of ≥ 24 was optimum at 95% sensitivity, with a 36% reduction in unnecessary biopsies and a few missed biopsies [78]. White et al. performed a study including two groups, with 506 men undergoing the PHI test and 683 men as controls. In this study, 73% of patient management was influenced by the PHI score, where ≥ 36 was considered the threshold. A notable reduction in unnecessary biopsies was observed among men that underwent PHI tests compared to controls (36.4% vs. 60.3%) [79].

4K score

The 4K score is a blood-based test, developed by OPKO Health Miami, FL, USA. Patients with abnormal PSA or DRE levels undergo a 4K score test to decide whether an initial biopsy is needed and patients for whom repeat biopsy is being considered. The possible candidates for this test are men with a family history of PC, but any man above 35 years of age, curious about his risk, can have this test [76].

The 4K score utilizes four levels of kallikreins- fPSA, iPSA, tPSA, and human kallikrein 2 (hK2) along with the patient data, such as DRE, age, and initial biopsy outcomes in a specific algorithm, providing results in the form of percentages from 0 to 100%. This percentage forecasts the probability of significant high-grade PC before biopsy. The 4K score can differentiate between patients with aggressive and slow PC. The patients with a Gleason score > 7 from those with a Gleason score < 7 have been considered as an aggressive in nature. The test can evaluate the threat of distant metastasis within 20 years of performing the test [69].

To validate the clinical utility and significance of 4K scores, Parekh et al. performed a trial in the United States that included 1021 men considered for biopsy, where 23% of men were found to have a Gleason score ≥ 7 PC. The 4K score demonstrated better accuracy than the Prostate Cancer Prevention Trial Risk Calculator 2.0 (PCPT-RC). The overall reduction in biopsies was reported to range between 30 and 58% depending on different thresholds, with only 1.3–4.7% cases of late diagnosis. When using a ≥ 6% probability of having a Gleason score ≥ 7 as the threshold for performing a biopsy, there would be a 30% reduction in biopsies with only 1.3% delayed cases. Similarly, when ≥ 9% and ≥ 15% is the threshold, the reduction in biopsies is 43% and 58%, with 2.4% and 4.7% delayed cases, respectively [80].

A case–control study of multi-ethnic groups involving 1,667 PC incidents and 691 controls with PSA levels of ≥ 2 ng/mL was conducted. The multi-ethnic groups included were native Hawaiians, White men, Latinos, African Americans, and Japanese. The outcomes demonstrated that among all ethnic groups, the 4K score has a higher ability to differentiate overall and aggressive PC compared to tPSA and tPSA + fPSA. Thus, the 4K score appears to be a better alternative in a multi-ethnic population, providing evidence of its more comprehensive utility [81]. Various studies have found that the 4K score test decreases the cost while simultaneously improving the quality of patient care [82, 83]. A comparative study of the PHI and 4K score showed that both increased discrimination while predicting high-grade PC and PC [84].

Urine-based biomarkers

Prostate cancer antigen 3 (PCA3)

The PCA3 gene codes for long non-coding ribonucleic acid (RNA), previously known as DD3. In more than 90% of PC cases, the PCA3 gene is 60–100 folds overexpressed compared to the normal tissues [85]. The PCA3 test was made commercially available by Hologic Inc. It is a non-invasive method and utilizes the amount of PCA3 and mRNA of PSA found in urine collected after DRE [70]. The levels of PCA3 and mRNA of PSA were quantified using quantitative real-time polymerase chain reaction (qPCR). This information was substituted in the formula PCA3 mRNA/ mRNA of PSA × 1000, generating a PCA3 score that predicts the probability of PC, decreasing the performance of unnecessary biopsies. A score of PCA3 (≥ 25) suggests a high probability of PC, whereas a score of < 25 is considered a low probability of PC [70]. Various studies have suggested a different threshold for PCA3 scores. Thus, the threshold ranges between 25 and 35 accordingly, and the reduction in unnecessary biopsies also changes from 37% to 77.1% [72]. The optimum threshold value for PCA3 remains contentious.

Merola et al. have performed a clinical study involving 407 men to assess the accuracy of PCA3 compared to total PSA and f/t PSA. They demonstrated that PCA3 surpassed the f/t PSA performance. When the cutoff score was 35, PCA3 was found to have better sensitivity (94.9%) and specificity (60.1%) compared to a cutoff score of 20 [85]. According to a meta-analysis of nine studies, PCA3 has an AUC of 0.734, with a significant sensitivity and specificity by 69% and 65%, respectively. This study showed that a cutoff score of 35 had better clinical accuracy and applicability than other values [86].

Exo-Dx (Prostate IntelliScore) (EPI)

Exo-Dx (Prostate IntelliScore) is a urine-based test. Exosomes are double-layered small vesicles that contain various cellular proteins produced by cells. This test measures exosome expression in urine by quantifying ERG and mRNA of PCA3 normalized to the SAM pointed domain containing ETS transcription factor (SPDEF) [87]. The results from this test are expressed as an EPI score ranging from 0 to 100 [71]. This test measures exosome gene expression in urine and does not require prostatic massage or pre-DRE for sample collection [87]. Based on the guidelines of National Comprehensive Cancer Network, this test can be useful in distinguishing the low-grade PC from high-grade PC in patients of more than 50 years of age with PSA levels of 2–10 ng/mL [87]. In a clinical trial study conducted on 503 men with an average age of 64 years and PSA level of 5.4 ng/mL has reported a reduction in total number of biopsies by 20%, unnecessary biopsy by 26%, and the number of missed biopsies by 7% with a negative predictive value (NPV) of 89% [88].

SelectMDx

SelectMDx is a urine-based test developed by MDx Health, Inc. Usually, a urine sample is taken after DRE. This quantifies the mRNA levels of HOXC6 and DLX1, which are considered as a biomarker gene [89]. Various clinical data, the PSA levels, DRE results, PSA density, and history of PC were also considered while calculating the score [72]. In a study with two cohorts of 519 and 386 patients, respectively, SelectMDx demonstrated its accuracy in predicting PC with the Gleason score of 7, an AUC of 0.86, and a negative predictive threshold of 98%. They also suggested that this test could reduce unnecessary biopsies by 53% and overall biopsies by 42% [90]. A study done by Haese et al. in European countries included 1955 men with PSA levels less than 10 ng/mg and tested their urine samples. At a sensitivity of 47%, SelectMDx had a specificity of 47% and an AUC score of 0.85. The NPV was 95%. The AUC values of this study were compared with those of PCPT-RC, which had an AUC of 0.76. Thus, the test outperformed the PCPT-RC results [91]. A study found that the clinical use of SelectMDx is cost-efficient in European countries [92].

Mi-Prostate Score (MiPS)

The MiPS was discovered by the University of Michigan, Michigan Labs. Urine samples were collected after a DRE or prostatic massage. MiPS measures the mRNA expression levels of the TMPRSS-ERG fusion gene, PCA3, and tPSA [93, 94]. This test is considered for patients undergoing initial biopsy, and the results predict the possibility of high-grade PC [74]. Some studies have found that the combination of PCA3 and TMPRSS2-ERG enhances diagnostic capabilities [95].

Sanda et al. conducted a multicenter study to analyze the clinical applicability of the MiPS. In a validation group of 561 men with an average age of 62 years, they found that MiPS enhanced the specificity from 17 to 33%, distinguishing high-grade PC with a sensitivity of 93%. They also observed a 42% reduction in unnecessary biopsies using the MiPS test before biopsy [96]. A validation study by Tomlins et al. demonstrated that MiPS outperforms PSA alone in diagnosis. When detecting PC, the AUC value of MiPS was 0.751, whereas the PSA AUC value was 0.585. Similarly, while detecting clinically significant high-grade PC, MiPS and PSA have AUC values of 0.772 and 0.651, respectively [97].

TMPRSS2-ERG fusion gene test

TMPRSS2-ERG (androgen-related transmembrane protease serine 2) and ERG (ETS-related gene) are present on chromosome 21. In 2005, TMPRSS2-ERG was found to be fused in 40–80% of PC cases. The TMPRSS2-ERG test gives a score that can predict the possibility of high-grade PC. This score is calculated using the formula: (TMPRSS2-ERG mRNA/PSA mRNA) × 100,000 [73]. TMPRSS2-ERG often combines with PCA3 to improve its predictability. The predictive value of this test is still under investigation [98].

Tissue-based biomarkers

ConfirmMDx

Initially, there were 20–30% chances of false-negative reports in the histological test of prostate biopsies. Thus, patients at potential risk of PC needed to undergo repeat biopsies, affecting low-risk patients. Thus, to avoid unnecessary biopsies in these patients, ConfirmMDx was designed. ConfirmMDx is a biopsy-based test that requires tissue samples to provide results. MDx Health Inc. developed this test. This epigenetic test utilizes methylation-specific PCR to evaluate the DNA hypermethylation of APC, GSTP1, and RASSF1. This test can histologically distinguish between normal and cancerous cells. Patients with negative ConfirmMDx results were found to have a < 5% lower probability of a rebiopsy, which is ten times decrease in the initial rates [75].

A multicentre study in the United States which involved 350 men who underwent for the repeated biopsy after a previous negative biopsy underwent the ConfirmMDx test within 24 months of the biopsy. The results showed an NPV of 88% and confirmed its predictive value in multivariate analysis. Therefore, unnecessary repeat biopsies can be avoided [99]. The MALTOC trial included 498 men with negative biopsies who underwent the ConfirmMDx test within 30 months of the previous biopsy. This trial showed an NPV of 90%, and the multivariate analysis showed its significance in predicting outcomes [100]. In a study done by Yonover et al. in the United States, 605 men with an average age of 64 years, average PSA of 6.8 ng/mL, and a negative biopsy report underwent the ConfirmMDx test within ten months of the previous biopsy. They found that the test significantly impacted clinical decision making in rebiopsy settings [101].

Emerging biomarkers for the diagnosis and prognosis of PC

Various potential molecular biomarkers for PC diagnosis and prognosis are still developing. Some of them are under investigation such as circulating tumor cells (CTCs), PTEN, androgen receptor variants, long non-coding RNAs such as HOX transcript antisense intergenic RNA [102, 103], SChLAP1 and MaLAT-1, and several miRNAs such as miRNA-141 and miRNA-301a. However, most of these biomarkers are yet to be approved for clinical use [104]. Feng et al. observed that spindle and kinetochore-associated complex subunit 3 (SKA3) is highly upregulated in PC cells compared to normal cells. Further, PC patients with higher expression of SKA3 are associated with an increased risk of rapid progression to metastasis [105].

Biomarkers of liquid biopsy: new edge technology for PC patients

Liquid biopsy is a non-invasive tool used for the diagnosis and management of PC patients. It is a more advanced approach for early detection and monitoring of PC. It utilizes blood, urine, and other body fluids as samples and facilitates different targeted therapies, and probability of resistance to therapies [106]. It analyses samples in real-time identifying and enumerating circulation tumor cells, cell-free DNA (cfDNA), circulating tumor DNA/RNA (ctDNA/RNA), and extracellular vesicles [107].

CTCs are cells shredded from primary or a metastatic tumor mass, circulating in the blood. CTCs are epithelial cell adhesion/activating molecule (EpCAM)-positive cells, which are used as a biomarker for the identification and enumeration of PC [108]. EpCAM is a transmembrane glycoprotein, its expression is upregulated in cancer cells. In cancer cells, it is responsible for cell adhesion, proliferation, angiogenesis, stemness, chemotherapy resistance, and epithelial to mesenchymal transition. Thus, EpCAM is a diagnostic and prognostic biomarker as well as a potential target for more precision therapy [108, 109]. The EpCAM-dependent CTC test has been the only US FDA approved technology to be used for clinical application [110]. CTCs of more than 5/7.5 ml of blood are considered unfavourable with shorter PFI and overall survival (OS) [107]. EpCAM independent CTC capture methods are also being developed such as epithelial immunospot (EPISPOT). It is an antibody-based method for quantification of live CTCs by detection of CTCs which are capable of secreting proteins such as cathepsin D, MUC1 and CK19 [111, 112]. For PC cells, PSA and FGF2 are target proteins for identification [113]. Another CTC biomarker under clinical investigation is AR-V7 [114].

Multiparametric magnetic resonance imaging (mpMRI)

mpMRI has enhanced the diagnosis, reducing unnecessary biopsies and improvised risk stratification system for prostate cancer patients [115]. mpMRI is performed when a person shows abnormal screening. If lesions (PI-RADS ≥ 3) appeared in MRI, then patient is recommended for targeted biopsy or systematic biopsy [115]. In 2019, the lesions of a clinically significant PC ranges from PI-RADS 1 to PI-RADS 5. Various lesions are graded as PI-RADS 3 or 4 characterised by mild to high chances of converting then into tumour in future [116].

Therapeutic strategies

PC is one of the leading cancers reported worldwide, and despite various advances in medical science. The treatment of PC still needs further improvement. Major therapeutic approaches for PC include surgical treatment, radiotherapy, chemotherapy, and hormone therapies [117]. The correct approach for the treatment of PC depends on whether we are trying to cure the disease or control certain symptoms. It also depends on an individual’s risk of death from other causes and the life expectancy. In nearly 80–90% of PC cases, increased androgen activity is detected in the initial stage of the disease. Hence, inhibition of the AR and reduction in androgen levels are the cornerstones of ADT. Therefore, ADT remained the first line of treatment for men with PC. ADT is variable as 20–30% of patients show tumor recurrence and become castration resistance, so a metastatic hormone naive tumor becomes mCRPC [118]. Men with localized PC have three treatment options: close monitoring, surgical treatment, and radiotherapy. Patients of metastatic PC has been reported to be treated with chemo-hormonal therapy such as docetaxel novel hormone therapy and cell-based cancer immunotherapy [119]. There is no specific sequential order of therapy for patients with PC because of rapid changes in treatment options and the approval of new drugs. We will now discuss some of the therapeutic options widely used by physicians to treat patients with PC.

Chemotherapy

Drugs approved for therapy include docetaxel, cabazitaxel, mitoxantrone, and bicalutamide (first-generation antiandrogens).

Docetaxel

Docetaxel is a taxane-based chemotherapeutic drug used to treat PC. It shows anticancer activity by inhibiting microtubule assembly during mitosis and interphase, leading to cell death (Fig. 2), and is thought to have some anti-androgenic properties [120]. Following several phase trials, this chemotherapeutic drug was the first found to increase OS of patients with PC. A multi-arm cohort study conducted on 593 patients, with the administration of ADT and docetaxel (75 mg/m2) after every three weeks and prednisone (10 mg/day) has showed a significant improvement in the OS compared with 1184 patients administered ADT alone. Therefore, this drug has an OS benefit when combined with another hormonal drug and steroids [121]. Docetaxel is usually administered intravenously once every three weeks for ten cycles. However, reducing the drug dose depends on the patient’s tolerability. Furthermore, like other chemotherapy drugs, it is also associated with certain side-effects, such as cytopenia, nausea, vomiting, and neutropenic sepsis [122].

Fig. 2
figure 2

Mechanism of action of Taxanes: a Taxane derivatives bind to β tubulin which leads to microtubule stabilization, this inhibits the proper assembly of microtubules and inhibits G2-M transition and apoptosis. b Taxanes are able to inhibit AR activity by FOXO1 mediated inhibition of AR transcriptional activity. c Taxanes inhibit antiapoptotic proteins (BCL2, and BCL-XL) and promotes activation of proapoptotic proteins (BAX, and BAK) leads to the release of cytochrome c that activates intrinsic apoptotic pathway, which leads to cell death (AR: Androgen Receptor and ARE: Androgen Receptor Element)

Cabazitaxel

Cabazitaxel is a US FDA approved semisynthetic compound. This compound is being used as a second line of therapeutic after docetaxel in patients with PC. This chemotherapeutic drug also has the same mode of action as docetaxel as it inhibits microtubule assembly (Fig. 2) [123]. It can overcome taxane resistance and shows anticancer activity in patients with post-docetaxel treatment and docetaxel-resistant cancers [124]. Cabazitaxel is administered via an intravenous infusion once every three weeks, and the standard dose is 25 mg/m2 fixed after studying various trials [125]. Detailed mechanism of action of taxanes like Docetaxel and Cabazitaxel is demonstrated in Fig. 2.

Mitoxantrone

Mitoxantrone is a synthetic compound, used as a second-line chemotherapeutic drug for the treatment of PC. Mitoxantrone causes immunogenic cell death in PC cells by activating eukaryotic initiation factor 2 [126]. Retrospective analysis of data from various phase 3 trials of mitoxantrone revealed symptomatic improvement without any survival benefits in some patients, as well as adverse impacts, such as fatigue, shortness of breath, and pancytopenia [127].

Novel hormone therapies

Novel hormone therapy, also known as androgen suppression therapy, is used to suppress the levels of androgen by targeting the androgen signaling pathway [128]. Overexpression of androgen is responsible for the progress of both mHSPC and mCRPC [129]. Some drugs that have regulatory approval include abiraterone and enzalutamide [130,131,132].

Abiraterone

Abiraterone acetate is an irreversible, selective inhibitor of cytochrome p450 17A1 (CYP17), which blocks the production of androgen in men with mCRPC [133,134,135]. Abiraterone, has been shown to provide a survival benefit and therefore approved for the treatment of mCRPC [136,137,138] by targeting androgen pathway (Fig. 3) [139]. Figure 3 shows precise mode of action of Abiraterone acetate and Enzalutamide as androgen suppression therapy. A random study conducted on 1,917 patients showed that abiraterone, ADT, and prednisone showed higher OS [140]. Abiraterone is orally administered at a dosage of 1000 mg/day and a low dose of prednisone has been reported to show fluid retention, hypertension, and hypokalemia with specific side effects such as increased levels of mineralocorticoids induced by the block of CYP17 [141].

Fig. 3
figure 3

Mechanism of action of Abiraterone acetate and Enzalutamide. Abiraterone acetate inhibits the synthesis of androgen by blocking the action of 17α-hydroxylase and C 17, 20-lyase on CYP-17, leading to the inhibition in binding of testosterone to AR. Enzalutamide binds with AR and inhibits the binding of testosterone to AR. It also inhibits nuclear translocation of AR (AA: Abiraterone acetate, E: Enzalutamide, ARE: Androgen Receptor Element, AR: Androgen Receptor, T: testosterone, DHEA: Dehydroepiandrosterone)

Enzalutamide

Enzalutamide is a second-generation antiandrogen therapeutic approved for the medication of CRPC. It is highly effective in men with non-metastatic CRPC and reduces the probability of metastasis and death by 71% [142]. Various phase 3 trials have shown that enzalutamide has anticancer activity and improves OS before and after chemotherapy. Enzalutamide is prescribed to be administered orally once at a dose of 160 mg/day. The most commonly observed side effects are gastrointestinal problems, fatigue, and hot flushes [143].

Radiotherapy

Radiation therapy is generally used to cure the early stages of PC by treating locally advanced tumors and reducing the risk of metastasis. This therapy has been slowly evolving. Indeed, various radiotherapy techniques have been developed, such as image-guided radiotherapy, stereotactic ablative body radiotherapy, intensity-modulated radiotherapy, and volumetric modulated arc therapy brachytherapy. Despite significant improvements in radiotherapy, many PC patients still suffer from the recurrence of the disease, due to the low specificity of therapy. Increasing the radiation dose to achieve adequate anti-cancer effects can cause damage to the healthy tissues and result in adverse impacts. New techniques, such as proton and carbon ion therapy, maximize their effect on tumor cells while minimizing their effect on surrounding healthy cells [144]. Nearly 10–40% of patients show tumor recurrence after having radical prostatectomy; for them, salvage radiation therapy is very effective and allows control of diseases in 60–70% of cases [145]. Radiation therapy can have certain radio-induced side effects in patients, such as radiation proctitis. The correct positioning of patients and set up verifications can help to prevent and/or reduce the risk of radiation proctitis [146]. Radical prostatectomy remains the first treatment option for localized PC and reduces the risk of mortality. It provides better survival chances than radiotherapy in patients with a primary tumor [147]. Major prostatectomies include open prostatectomy, laparoscopic prostatectomy, and TURP.

Radium-223 dichloride, also called alpha radio, is a radiopharmaceutical that emits alpha particles that selectively target bone metastases [148]. Radium-223 is specifically used to treat patients with mCRPC with bone metastases to induce irreversible double-strand breaks in DNA, leading to tumor cell death [149]. It is usually administered through intravenous infusion for four weeks with six cycles. The most commonly reported side effects include bone pain, fatigue, gastrointestinal disturbances, hematological toxicity, thrombocytopenia, and leukopenia, which affect the adjacent bone marrow [150].

Phototherapy

Phototherapy (PT) uses the materials which can absorb the electromagnetic energy and transform it into thermal energy to induce apoptosis in cancer cells [151]. This approach eliminates the risk of infection at the time of surgery and get rid of the side effects of chemotherapy. Near-infrared (800–1350 nm) light is primarily employed for the PT. The materials which have the ability to transform near-infrared radiation into temperature has been used for the PT and are recognized as the photothermal agents [151,152,153]. PT has been blended with other therapeutic regimens such as immunotherapy or chemotherapy for enhancing the therapeutic impact against tumors [154]. Photodynamic therapy (PDT) is also an approach for the treatment of cancer which promotes the generation of reactive oxygen species in cancer cells leading to the induction of apoptosis in cancer cells [155, 156]. Polymer-coated metal nanostructures have been used for the PDT. It has also been employed for improving the efficacy of radiotherapy in suppressing the liver cancer [157]. Silver-gold hollow nano shells with mesoporous silica nanoparticle have been utilized for the PDT in the experimental PC cells [158]. In another study, a hybrid nanoparticles of Au-alendronate were made for combined PDT-chemotherapy of PC [159]. The PDT not only enhanced the impact of chemotherapy in suppressing PC, but also decreased the dose of the chemotherapy needed for the treatment of PC [160,161,162].

Immunotherapy

Immunotherapy (IT) has moved the therapy concept of different tumors/cancers in clinical scenarios [163, 164]. However, the clear impact in the suppression of PC has not been reported yet [165]. PC remained as a tumor with predominantly immunosuppressive components, like regulatory T cells (Tregs) and transforming growth factor-β (TGF-β). Although tumor cells has been reported to express certain marker antigens, such as PSMA and PSA [166]. There are several immunotherapies reported to optimize the available treatment strategies.

Anticancer agents are designed to join with high selectivity of monoclonal antibodies to form antibody–drug conjugates such as Sacituzumab govitecan, trastuzumab deruxtecan targeting Trop2, and HER2. It has the capability to directly carry cytotoxic drugs to tumor [167]. The treatment of PC with immunomodulatory drugs includes both active as well as passive approaches. The active approach includes vaccines which are designed with the intent to stimulate an adaptative immune response through presentation of an antigen [165]. The passive approach includes the administration of highly specific monoclonal antibodies for tumor-associated antigens (TAAs) and tumor-specific antigens (TSAs). The efficacy of anticancer vaccines of PC can be analyzed using its specific biological markers or features, which include an early diagnosis of disease recurrences, slow growth, and a series of TAAs (PSA, and PSMA), TSAs [168], prostate stem cell antigen (PSCA), prostate acid phosphatase (PAP), PCA-3 antigen, six-transmembrane epithelial antigens of prostate (STEAP), and mucin-1 [169]. It has reported that vaccines can be used along with the other therapeutics such as second-generation hormonal treatments (docetaxel, and radiotherapy) [170]. Vaccine-based treatments can be categorized into two groups such as cell-based and viral vector-based vaccines [168, 171, 172].

Chimeric antigen receptor (CAR) T-cells is a recently advancing approach of immunotherapy for the treatment of solid tumors where antibody fragments are used along with T cells specific against TSAs [173, 174]. CAR T-cell therapy have been reported to show a remarkable success in B cell hematological malignancy [175,176,177,178]. There are several newly emerging TAAs under investigation such as immune checkpoint B7-H3 (CD276), Mucin-1, IL-6 receptor (CD126), Lewis-y antigen, STEAP-1 [179]. Major challenges faced by this therapy are manufacturing of CAR T-cells, direct attack on normal tissues that share expression of the TAAs called On-Target Off-Tumor toxicity, and cytokine toxicity [180,181,182]. IT has emerged as a potential treatment regimen for mCRPC. It can significantly improve progression-free survival and overall response rate especially in immune checkpoint inhibitors treatment. However, they cannot improve the OS [183].

Cell-based vaccines

Sipuleucel-T

Sipuleucel-T is a vaccine of autologous dendritic cells that unleashes an immune response against PAP antigen. This is an autologous active immunotherapeutic agent which has been reported to improve the survival of patients with mildly symptomatic PC [184]. It was the first US FDA approved therapeutic vaccine for cancer. It evokes patients’ immune systems to identify and combat cancer [185]. Detailed workflow of Sipuleucel-T immunotherapy is demonstrated in Fig. 4. The recommended dose for treatment is an intravenous infusion of three complete doses at an interval of two weeks. The most common after-effects include bleeding, bruising, pyrexia, fatigue, nausea, and headache [186]. This drug is not widely used because of its high production cost.

Fig. 4
figure 4

Mechanism of action of Sipuleucel-T. Sipuleucel-T is an autologous cellular immunological agent, here blood cells from prostate cancer patients are taken and processed through leukapheresis then density gradient centrifugation of leukocytes is done to get monocytes, monocytes are fused with fusion protein (PAP and GM-CSF) then it is culture for 36–44 h, infused monocyte is intravenously administered back to the patient. Infused monocyte having GM-CSF activates the APC that led to prostate tumor cell lysis by CD8 T cell. (GM-CSF-Granulocyte macrophage colony-stimulating factor, PAP-Prostatic acid phosphatase, APC- Antigen presenting cells)

G-VAX

G-VAX is a granulocyte–macrophage colony-stimulating factor (GM-CSF) gene-transfected tumor cell vaccine. It has been genetically modified for the expression of GM-CSF with the intention of increasing the differentiation and growth of dendritic cells [187]. This approach has the advantage of stimulate various TAAs without any pairing of HLA [188]. Though initial outcomes were promising, the successive results were negative against docetaxel from the phase III trials.

Viral vector-based vaccines

Viral vector-based vaccines include the vectors obtained from oncolytic viruses. Such vectors can infect tumor cells and stimulate their death by antigen-presenting cells (APCs). Therefore, the APCs can produce TAAs which are responsible for the response of T cell [171]. A recombinant vaccine of Poxvirus which contains a PSA transgene with an HLA-A2 epitope, has been altered to enhance the immunogenicity of co-stimulatory molecules. The costimulatory molecules have been reported to be a B7-1 (CD80), an intercellular adhesion molecule 1 (ICAM-1 or CD54) and lymphocyte function-associated antigen-3 (LFA-3 or CD58) [189, 190]. The results published related to the PROSTVAC-VF have not shown a clear clinical benefit in treatment of PC patients [191].

Immune checkpoint inhibitors

Tumor microenvironment (TME) is composed of various components which includes tumor cells, immune cells like myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), tumor-associated dendritic cells (tDCs), and adoptive immune cells like the regulatory T cells (Tregs), extracellular matrix, stromal cells, vessels, soluble factors and physical properties [192]. However, the immunosuppressive microenvironment is composed of cellular and soluble components that promotes tumor progression and favors immune escape [192, 193]. Immune checkpoint inhibitors (ICIs) are referred as the monoclonal antibodies which are designed to target different receptors found in immune response [194, 195]. The most clinically proven ICIs are directed against PD-L1, PD-1, and CTLA4 [195]. PD-1 is a T cell transmembrane protein which has been reported to interact with its ligand (PD-L1) in the tumor cells [196]. PD-1 and PD-L1 are located on chromosome 9p24.1, and plays vital role in maintaining immune homeostasis [197], inside TME the activity of PD-1and PD L1 is seized by cancerous cells to escape immune surveillance [198]. Activation of T-cells are damaged due to interaction of overexpressed PD-L1 in cancer cell with PD-1 on tumor-infiltrating lymphocytes (TILs) which effects the TCR-signalling cascade by phosphorylating SHP-2 [199]. Transcriptional activation of PD-L1 is regulated by various transcription factors like MYC, STAT3, NF-κB, AP1, and HIF-1. Modification processes like ubiquitination, glycosylation, phosphorylation can affect the stability of PD-L1 protein in cancer cells so regulating the expression of PD-L1 protein [200]. High expression level of PD-1/PD-L1 has been reported in the PC cases, however, its role in response to ICIs remained debatable [201]. Inactivating the mutations in cyclin-dependent kinase-12 (CDK-12) have been reported to be directly related with an increased sensitivity towards immunotherapy. However, there are very few clinical data available to support the use of ICIs [202]. Only the presence of insufficient mismatch repair has been recognized by the clinical guidelines as a transversal agnostic for anti-PD1 therapy [203]. The synergistic effect of anti-D-1/PD-L1 and anti-CTLA-4 has been reported in renal cancer and melanoma [204, 205]. It has been shown that the expression of androgen receptor splice variant 7 (AR-V7) cause alterations in genes involved in DNA repair that made them more susceptible to ICIs [206]. In another study, the ipilimumab and nivolumab had synergistically effective in the cancer patients with the expression of AR-V7 [207]. CTLA-4 receptor has been reported to be present in the membrane of T lymphocytes. The stimulation of CTLA-4 receptor accelerates the inhibition of the function of T lymphocyte [208]. Ipilimumab has been shown to function as an anti-CTLA-4 agent and demonstrated a positive result for the treatment of PC in several studies [209]. The increase of myeloid-derived suppressor cells has been reported to be associated with the resistance to treatment [210]. The application of anti-PD-1/PD-L1 is limited to clinical trials in mCRPC [211]. Preclinical studies have shown that the over expression of PD-L1 were produced by the medication with Poly (ADP-ribose) polymerase (PARP) inhibitors [212]. Moreover, it has been reported that the sensitivity of NK cells can be enhanced by the treatment of Olaparib in PC [213]. In tumor microenvironment, the interaction between the immune response and angiogenesis pathway has been shown to favor the generation of an immunosuppressive state. The therapy with antiangiogenic agents has immunomodulatory effects which enable the response to ICIs [214].

Gene therapy

Various strategies for gene therapy (GT) have been established through novel and advanced drug delivery systems. GT has shown significant potential for the cure of tumors in PC patients. Several types of GT are used to treat PC including suicide GT (SGT), tumor-suppressor GT (TSGT), anti-oncogene therapy (AOT), and immunomodulatory GT (IGT).

The concept of SGT is based on the killing of cancer cells by introducing a therapeutic gene in the cancer cell. After entry into the cancer cells, these SGTs have been reported to express and kill the cells without correcting the malignant mutations. It has also been shown that these SGTs have not affected the normal cells. SGT has been mainly divided into two major categories such as enzyme-based GT in cells. The enzyme-based SGT has been shown to suppress the proliferation of tumor [215]. In another study, Lee et al. (2020) have used the double SGT for the efficient treatment of PC using gemcitabine conjugated adenovirus [216].

TSGT has been achieved by introducing a wild-type gene into PC cells to suppress the proliferation of tumor [217]. The genes which have been usually studied for TSGT include p53, p21, and retinoblastoma [218, 219]. Successful GT has been achieved when all tumor cells remain transduced by the tumor-suppressor genes [220, 221]. The protein responsible for the tumor-suppression (p14ARF) has been used to regulate the activity of AR and modulate the level of p14ARF in prostate [218]. In another report, an Arv7-mediated CRPC has been created utilizing an active AR splice variant to inhibit the tumor [222]. Moreover, miR-21 has also been exploited to inhibit the proliferation of PC by targeting the tumor suppressor gene PTEN [223].

The capacity of immune system to detect and kill tumor cells is very low in humans. The immune system has been reported to be weakened due to the deficient expression of MHC antigens, which consequently lowers the T cells activation [224]. In this context, various immunomodulatory gene therapies have been established for the treatment of PC by using the gene vaccines [225]. Another type of immunomodulatory GT has been recognized as the intratumoral injection of cytokine genes using vectors [226].

Anti-oncogenes are made to target the specific tumor RNAs leading to the inhibition of tumor growth and proliferation. This therapeutic methodology has been used for the safe transgene delivery devoid of damaging normal cells and preventing the lysis of viral cells [227]. The cycle of cell lysis progresses until all the cancer cells are eradicated and make sure that the tumor is completely cured [228]. Adenovirus early region-1 is the most commonly used viral vector which has been reported to act as a good transgene carrier [229]. Due to the outstanding delivery of gene product, efforts has been made to conjugate with other therapeutic genes which exhibited strong promise for the treatment of PC [230].

Nanotherapies

The use of nanotechnology has been expanded the modern scope for the treatment of diseases, and their diagnosis [231,232,233]. These nanocarriers have the potential to eradicate the cancer by targeted delivery of drugs and genes. There are several nanocarrier systems which have been used for enhancing drug delivery with higher biocompatibility of [234,235,236]. Major drug delivery systems in tumor treatment are polymeric spheres, liposomes, dendrimer, carbon nanotubes, mesoporous silica nanoparticles, virosomes, extracellular vesicles [237]. Apart from these therapeutic agents, the nanocarriers are being rapidly developed for the detection of tumor markers [238]. The aptamers have been reported to lack immunogenic toxicity and are easily synthesized [239, 240]. Self-assembled polymeric nanoparticles have been synthesized using the PLGA, and PEG and functionalized with Wy5a aptamer for the suppression of PC aggression [241]. Furthermore, these nanostructures loaded with doxorubicin has substantially eradicated the PC and delayed the growth of tumor in xenograft model [242]. The modification of nanoparticles with aptamers have been shown to increase the internalization into the PC cells [243]. In addition, hyaluronic acid-modified nanoparticles carrying epigallocatechin-3-gallate have been reported to significantly decrease the rate of proliferation of PC [244].

Ribonucleic acid interference (RNAi) has been utilized for silencing of the target gene. The application of nanocarriers for targeted delivery of RNAi has been recommended for enhancing the internalization of RNAi [245,246,247]. In another study, the siRNA-loaded with gold nanoparticles has been reported to penetrate the PSMA-over expressed PC cells [248]. The mesoporous silica nanoparticles has also been used to improve the gene silencing potential of siRNA in the tumor cells [249]. PLGA based nanocarriers have been made for androgen receptor-shRNA delivery for the suppression of PC [250]. The tumor cells has been reported to show increased sensitivity towards cisplatin by down-regulating the Lcn2 gene [251]. Sorrentino et al. have reported that interleukim-30 deletion using CRISPR/Cas9 reduces PC growth and elongates progression-free survival via upregulating SOCS3 and inhibiting the expression of IGF1 and CXCL5 [252]. Various nanocarriers have also been established for the delivery of anthracyclines in PC cells [253]. In addition, the polypeptide based nanocarriers loaded with doxorubicin have been shown to raise the oxidative damage, which eventually inhibited the metastasis of PC in mouse [254]. Several nanocarriers have been made to deliver the platinum-based agents to PC cells [255,256,257].

Clinical studies

Clinical symptoms observed in PC may differ according to the level of the cancer. In other words, it depends on whether the lobe of prostate gland is affected and metastasized to other portions of body or not. In the case of locally advanced PC, cancer cells break out of the prostate gland, affecting nearby organs. Simultaneously, metastatic PC occurs when cancer metastasizes to the bones and lymph nodes. Patients with early PC are generally asymptomatic. Localized PC shows lower urinary tract symptoms (LUTS) in benign prostatic hyperplasia. Clinical manifestations of locally advanced PC include erectile dysfunction, painful ejaculation, sexual dysfunction [31, 32], hematuria, haematospermia, fatigue, low appetite, weight loss, nausea, vomiting, chronic bone pain in the pelvis, vertebrae, ribs, and hips. PSA testing and DRE enable the diagnosis of PC at early stages [258]. AR signaling have been reported to play a crucial role in initiation and the progression of PC [259]. Generally, localized PC is controlled by radical prostatectomy or radiation therapy with or without ADT [260]. In recent decades, substantial development has been made in the treatment of CRPC, such as abiraterone, apalutamide, enzalutamide, and darolutamide [261,262,263,264,265]. The new clinically approved agents by US FDA for diagnosis and treatment of PC are summarized in Table 3.

Table 3 US FDA approved therapeutic agents for the clinical use in the treatment of PC

Metastasis to bone has been successfully treated using the bisphosphonates, radium 223, and receptor activator of NFκ-B ligand inhibitor denosumab [121, 148, 266,267,268,269]. Several PARPi (rucaparib, olaparib, and talazoparib) has been evaluated in clinical trials for mCRPC [270,271,272,273]. PARP is responsible for repair of DNA damage [274]. Moreover, the early clinical studies focused to target ICIs, such as CTLA4, PD1 or PD-L1 have been evaluated [207, 275,276,277,278]. The PSMAs are greatly expressed in the cell membranes of PC [279]. Thus, PSMA targeting small molecules have been evaluated for their impact on PC cells in several clinical investigations [280,281,282,283,284,285,286,287,288,289,290]. Single-agent medication with the PI3K/AKT/mTOR inhibitors or in combination with inhibitors of AR signaling have also been studied in several studies [291,292,293,294,295,296,297]. The therapeutic agents targeting other signaling pathways, such as wingless-type protein signaling, CDK, p53, vascular endothelial growth factor, endothelin A receptor, receptor tyrosine kinases, epidermal growth factor receptor, fibroblast growth factor receptor, proto-oncogene tyrosine-protein kinase Src, transforming growth factor beta, and mitogen-activated protein kinase, have also entered clinical trials [298,299,300,301,302,303,304,305,306,307,308,309]. In recent years the several advancements have been done for the treatment of the PC. Despite these advances, current options for the treatment of PC have many limitations. Therefore, more specific treatment and targeted approaches are required for novel a better therapeutic possibility.

Recommendations and guidelines

According to the European Association of Urology (EUA) 2020 guidelines, men with a PSA level of < 10 ng/mL are considered low-risk, PSA 10–20 ng/mL as intermediate-risk, and PSA > 20 ng/mL as high-risk [310]. The European Society of Medical Oncology recommends that risk calculation and mp-MRI be performed before a biopsy is performed. They also suggested performing transperineal biopsies instead of transrectal biopsies [311]. According to the EUA, there is not currently enough data to promote the use of ConfirmMDx for rebiopsy. Thus, due to the lack of evidence regarding the clinical utility of ConfirmMDx, its routine use is not recommended [312].

Conclusions and future perspectives

PC has become a global burden because of the increasing number of patients and deaths. It is the second most commonly diagnosed cancer worldwide. Despite such a high prevalence of PC, its mortality rate is less because of PSA screening. In addition to recent screening techniques like DRE, ultrasound, and mp-MRI, liquid biopsy is an emerging diagnostic tool. The correct combination of chemotherapy drugs, such as docetaxel cabazitaxel, with ADT drugs, such as enzalutamide abiraterone, has improved OS in patients with mCRPC. Advancements in treatment strategies by analyzing cancer prognosis and patient preferences have helped lower mortality and increase the QoL of PC survivors. There appear to be a number of future therapies, such as the DNA repair pathway, platinum-based chemotherapy, and PARP inhibition. Further studies and phase trials are needed to develop therapies with fewer side effects. The use of radiotherapy and prostatectomy during an early stage of PC affects QoL of survivors. Therefore, improved therapeutic technologies will help to minimize the side effects of treatment.

Availability of data and materials

The material supporting the conclusion of this review has been included within the article.

References

  1. Bray F, Laversanne M, Weiderpass E, Soerjomataram I. The ever-increasing importance of cancer as a leading cause of premature death worldwide. Cancer. 2021;127(16):3029–30.

    PubMed  Google Scholar 

  2. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA-Cancer J Clin. 2021;71(3):209–49.

    PubMed  Google Scholar 

  3. Pernar CH, Ebot EM, Wilson KM, Mucci LA. The epidemiology of prostate cancer. Cold Spring Harb Perspect Med. 2018;8(12): a030361.

    PubMed  PubMed Central  Google Scholar 

  4. Rawla P. Epidemiology of prostate cancer. World J Oncol. 2019;10(2):63–89.

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Mathur P, Sathishkumar K, Chaturvedi M, Das P, Sudarshan KL, Santhappan S, Nallasamy V, John A, Narasimhan S, Roselind FS. Cancer Statistics, 2020: Report From National Cancer Registry Programme. India JCO Glob Oncol. 2020;2020(6):1063–75.

    Google Scholar 

  6. Pinsky PF, Miller E, Prorok P, Grubb R, Crawford ED, Andriole G. Extended follow-up for prostate cancer incidence and mortality among participants in the Prostate, Lung, Colorectal and Ovarian randomized cancer screening trial. BJU Int. 2019;123(5):854–60.

    PubMed  Google Scholar 

  7. Liu X, Zhang Y, Duan H, Yang L, Sheng C, Fan Z, Liu Y, Gao Y, Wang X, Zhang Q, et al. Risk-stratified multi-round PSA screening for prostate cancer integrating the screening reference level and subgroup-specific progression indicators. Eur J MedRes. 2023;28(1):257.

    CAS  Google Scholar 

  8. Cuzick J. The importance of long-term follow up of participants in clinical trials. Br J Cancer. 2023;128(3):432–8.

    PubMed  Google Scholar 

  9. Zeigler-Johnson C, McDonald AC, Pinheiro P, Lynch S, Taioli E, Joshi S, Alpert N, Baudin J, Joachim C, Deloumeaux J, et al. Trends in prostate cancer incidence among Black men in the Caribbean and the United States. Prostate. 2023;83(12):1207–16.

    PubMed  Google Scholar 

  10. Brawley OW. Trends in prostate cancer in the United States. J Natl Cancer Inst Monogr. 2012;2012(45):152–6.

    PubMed  PubMed Central  Google Scholar 

  11. Smith-Palmer J, Takizawa C, Valentine W. Literature review of the burden of prostate cancer in Germany, France, the United Kingdom and Canada. BMC Urol. 2019;19(1):19.

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Jain MA, Sapra A: Cancer Prostate Screening. In StatPearls. Treasure Island (FL): StatPearls Publishing. Copyright © 2021, StatPearls Publishing LLC.; 2021.

  13. Kaler J, Hussain A, Haque A, Naveed H, Patel S. A comprehensive review of pharmaceutical and surgical interventions of prostate cancer. Cureus. 2020;12(11):e11617–e11617.

    PubMed  PubMed Central  Google Scholar 

  14. Gann PH. Risk factors for prostate cancer. Rev Urol. 2002;4(5):S3–10.

    PubMed  PubMed Central  Google Scholar 

  15. Merriel SWD, Funston G, Hamilton W. Prostate cancer in primary care. Adv Ther. 2018;35(9):1285–94.

    PubMed  PubMed Central  Google Scholar 

  16. Kohada Y, Hieda K, Miyamoto S, Tasaka R, Asami A, Akiyama K, Sakamoto Y, Kirishima F, Saito K, Fukushima T, et al. Retrospective evaluation of the improvement in the urinary status-related quality of life after robot-assisted radical prostatectomy. Int J Urol. 2023. https://doi.org/10.1111/iju.15251.

    Article  PubMed  Google Scholar 

  17. Pantwalawalkar J, Chandankar S, Tade R, Khan Z, Shaikh M, Powar T, Patil P, Sugandhi V, Nangare S. Graphene quantum dot based ultrasensitive probe for biosensing of prostate cancer biomarkers: current updates and future challenges. Adv Nat Sci: Nanosci Nanotechnol. 2022;13(1): 013001.

    Google Scholar 

  18. Haglind E, Carlsson S, Stranne J, Wallerstedt A, Wilderäng U, Thorsteinsdottir T, Lagerkvist M, Damber J-E, Bjartell A, Hugosson J, et al. Urinary incontinence and erectile dysfunction after robotic versus open radical prostatectomy: a prospective, controlled, nonrandomised trial. Eur Urol. 2015;68(2):216–25.

    PubMed  Google Scholar 

  19. Force UPST. Screening for prostate cancer: US preventive services task force recommendation statement. JAMA. 2018;319(18):1901–13.

    Google Scholar 

  20. Tu X, Liu Z, Chang T, Qiu S, Xu H, Bao Y, Yang L, Wei Q. Transperineal magnetic resonance imaging-targeted biopsy may perform better than transrectal route in the detection of clinically significant prostate cancer: systematic review and meta-analysis. Clin Genitourin Cancer. 2019;17(5):e860–70.

    PubMed  Google Scholar 

  21. Pirola GM, Castellani D, Orecchia L, Giulioni C, Gubbiotti M, Rubilotta E, Maggi M, Teoh JYC, Gauhar V, Naselli A. Transperineal US-MRI fusion-guided biopsy for the detection of clinical significant prostate cancer: a systematic review and meta-analysis comparing cognitive and software-assisted technique. Cancers. 2023;15(13):3443.

    PubMed  PubMed Central  Google Scholar 

  22. Xie J, Jin C, Liu M, Sun K, Jin Z, Ding Z, Gong X. MRI/transrectal ultrasound fusion-guided targeted biopsy and transrectal ultrasound-guided systematic biopsy for diagnosis of prostate cancer: a systematic review and meta-analysis. Front Oncol. 2022;12: 880336.

    PubMed  PubMed Central  Google Scholar 

  23. Gordetsky J, Epstein J. Grading of prostatic adenocarcinoma: current state and prognostic implications. Diagn Pathol. 2016;11(1):25.

    PubMed  PubMed Central  Google Scholar 

  24. Rouvière O, Puech P, Renard-Penna R, Claudon M, Roy C, Mège-Lechevallier F, Decaussin-Petrucci M, Dubreuil-Chambardel M, Magaud L, Remontet L, et al. Use of prostate systematic and targeted biopsy on the basis of multiparametric MRI in biopsy-naive patients (MRI-FIRST): a prospective, multicentre, paired diagnostic study. Lancet Oncol. 2019;20(1):100–9.

    PubMed  Google Scholar 

  25. Puche-Sanz I, Rodríguez-Martínez A, Garrido-Navas MC, Robles-Fernández I, Vázquez-Alonso F, Álvarez Cubero MJ, Lorente-Acosta JA, Serrano-Fernández MJ, Cózar-Olmo JM. Liquid biopsy and prostate cancer. Current evidence applied to clinical practice. Actas Urol Esp (Engl Ed). 2020;44(3):139–47.

    CAS  PubMed  Google Scholar 

  26. Saranyutanon S, Srivastava SK, Pai S, Singh S, Singh AP. Therapies targeted to androgen receptor signaling axis in prostate cancer: progress, challenges, and hope. Cancers (Basel). 2020;12(1):51.

    CAS  Google Scholar 

  27. Ng K, Smith S, Shamash J. Metastatic Hormone-Sensitive Prostate Cancer (mHSPC): advances and treatment strategies in the first-line setting. Oncol Ther. 2020;8(2):209–30.

    PubMed  PubMed Central  Google Scholar 

  28. Sumanasuriya S, De Bono J. Treatment of advanced prostate cancer-a review of current therapies and future promise. Cold Spring Harb Perspect Med. 2018;8(6):35.

    Google Scholar 

  29. Bill-Axelson A, Holmberg L, Garmo H, Taari K, Busch C, Nordling S, Häggman M, Andersson S-O, Andrén O, Steineck G, et al. Radical prostatectomy or watchful waiting in prostate cancer — 29-year follow-up. N Engl J Med. 2018;379(24):2319–29.

    PubMed  Google Scholar 

  30. Groarke A, Curtis R, Skelton J, Groarke JM. Quality of life and adjustment in men with prostate cancer: Interplay of stress, threat and resilience. PLoS ONE. 2020;15(9): e0239469.

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Yabroff KR, Zhao J, Chen MH, Hoque J, Arias G, Han X, Zheng Z. Financial hardship and psychosocial well-being and quality of life among prostate cancer survivors in the United States. Urol Oncol: Semin Orig Invest. 2023;8:56.

    Google Scholar 

  32. Korfage IJ, Hak T, de Koning HJ, Essink-Bot M-L. Patients’ perceptions of the side-effects of prostate cancer treatment—a qualitative interview study. Soc Sci Med. 2006;63(4):911–9.

    PubMed  Google Scholar 

  33. Ho MY, Mackey JR. Presentation and management of docetaxel-related adverse effects in patients with breast cancer. Cancer Manage Res. 2014;6:253–9.

    Google Scholar 

  34. Baker J, Ajani J, Scotté F, Winther D, Martin M, Aapro MS, von Minckwitz G. Docetaxel-related side effects and their management. Eur J Oncol Nurs. 2008;12(3):253–68.

    PubMed  Google Scholar 

  35. Sperlich C, Saad F. Optimal management of patients receiving cabazitaxel-based chemotherapy. Can Urol Assoc J. 2013;7(1):S18-24.

    PubMed  PubMed Central  Google Scholar 

  36. Mellado B, Jimenez N, Marin-Aguilera M, Reig O. Diving into cabazitaxel’s mode of action: more than a taxane for the treatment of castration-resistant prostate cancer patients. Clin Genitourin Cancer. 2016;14(4):265–70.

    PubMed  Google Scholar 

  37. Fox EJ. Mechanism of action of mitoxantrone. Neurology. 2004;63(12 Suppl 6):S15-18.

    CAS  PubMed  Google Scholar 

  38. Cochrane ZR. Uses and risks of mitoxantrone. https://www.medicalnewstodaycom/articles/322211. Accessed 15 Jan 2019.

  39. Li R, Evaul K, Sharma KK, Chang KH, Yoshimoto J, Liu J, Auchus RJ, Sharifi N. Abiraterone inhibits 3β-hydroxysteroid dehydrogenase: a rationale for increasing drug exposure in castration-resistant prostate cancer. Clin Cancer Res. 2012;18(13):3571–9.

    CAS  PubMed  Google Scholar 

  40. Bedussi F, Galli D, Fragni M, Valcamonico F, Rossini E, Dalla Volta A, Vezzoli S, Roca E, Ferrari V, Lazzari B, et al. Amiloride is effective in the management of abiraterone-induced mineralocorticoid excess syndrome without interfering with its antineoplastic activity. Pharmacol. 2017;100(5–6):261–8.

    CAS  Google Scholar 

  41. Merseburger AS, Haas GP, von Klot CA. An update on enzalutamide in the treatment of prostate cancer. Ther Adv Urol. 2015;7(1):9–21.

    PubMed  PubMed Central  Google Scholar 

  42. Schalken J, Fitzpatrick JM. Enzalutamide: targeting the androgen signalling pathway in metastatic castration-resistant prostate cancer. BJU Int. 2016;117(2):215–25.

    CAS  PubMed  Google Scholar 

  43. Cha T-L, Wu TT-L, Vogelzang NJ, Huang C-Y, Huang S-P, Lin C-C, Ou Y-C, Pang S-T, Shen DH-Y, Wu W-J, Chang WY-H. Optimal usage of radium-223 in metastatic castration-resistant prostate cancer. J Formosan Med Assoc. 2017;116(11):825–36.

    CAS  PubMed  Google Scholar 

  44. Wei XX, Fong L, Small EJ. Prostate cancer immunotherapy with sipuleucel-T: current standards and future directions. Expert Rev Vaccines. 2015;14(12):1529–41.

    CAS  PubMed  Google Scholar 

  45. Key statistics of prostate cancer. https://www.cancer.org/cancer/prostate-cancer/about/key-statistics.html.

  46. Culp MB, Soerjomataram I, Efstathiou JA, Bray F, Jemal A. Recent global patterns in prostate cancer incidence and mortality rates. Eur Urol. 2020;77(1):38–52.

    PubMed  Google Scholar 

  47. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer Statistics, 2021. CA Cancer J Clin. 2021;71(1):7–33.

    PubMed  Google Scholar 

  48. Cancer Stat Facts: Prostate Cancer. 2021. https://seer.cancer.gov/statfacts/html/prost.html

  49. Cuzick J, Thorat MA, Andriole G, Brawley OW, Brown PH, Culig Z, Eeles RA, Ford LG, Hamdy FC, Holmberg L, et al. Prevention and early detection of prostate cancer. Lancet Oncol. 2014;15(11):e484-492.

    PubMed  PubMed Central  Google Scholar 

  50. Grossman DC, Curry SJ, Owens DK, Bibbins-Domingo K, Caughey AB, Davidson KW, Doubeni CA, Ebell M, Epling JW Jr, Kemper AR, et al. Screening for prostate cancer: US preventive services task force recommendation statement. JAMA. 2018;319(18):1901–13.

    PubMed  Google Scholar 

  51. Chowdhury-Paulino IM, Ericsson C, Vince R Jr, Spratt DE, George DJ, Mucci LA. Racial disparities in prostate cancer among black men: epidemiology and outcomes. Prostate Cancer Prostatic Dis. 2022;25(3):397–402.

    PubMed  Google Scholar 

  52. Merrill RM, Sloan A. Risk-adjusted incidence rates for prostate cancer in the United States. Prostate. 2012;72(2):181–5.

    PubMed  Google Scholar 

  53. Wu I, Modlin CS. Disparities in prostate cancer in African American men: what primary care physicians can do. Cleve Clin J Med. 2012;79(5):313–20.

    PubMed  Google Scholar 

  54. Okobia MN, Zmuda JM, Ferrell RE, Patrick AL, Bunker CH. Chromosome 8q24 variants are associated with prostate cancer risk in a high risk population of African ancestry. Prostate. 2011;71(10):1054–63.

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Robbins CM, Hooker S, Kittles RA, Carpten JD. EphB2 SNPs and sporadic prostate cancer risk in African American men. PLoS ONE. 2011;6(5): e19494.

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Kiciński M, Vangronsveld J, Nawrot TS. An epidemiological reappraisal of the familial aggregation of prostate cancer: a meta-analysis. PLoS ONE. 2011;6(10): e27130.

    PubMed  PubMed Central  Google Scholar 

  57. Junejo NN, AlKhateeb SS. BRCA2 gene mutation and prostate cancer risk. Comprehensive review and update. Saudi Med J. 2020;41(1):9–17.

    PubMed  PubMed Central  Google Scholar 

  58. Castro E, Goh C, Olmos D, Saunders E, Leongamornlert D, Tymrakiewicz M, Mahmud N, Dadaev T, Govindasami K, Guy M, et al. Germline BRCA mutations are associated with higher risk of nodal involvement, distant metastasis, and poor survival outcomes in prostate cancer. J Clin Oncol. 2013;31(14):1748–57.

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Goh CL, Eeles RA. Germline genetic variants associated with prostate cancer and potential relevance to clinical practice. Recent Results Cancer Res. 2014;202:9–26.

    CAS  PubMed  Google Scholar 

  60. Discacciati A, Wolk A. Lifestyle and dietary factors in prostate cancer prevention. Recent Results Cancer Res. 2014;202:27–37.

    CAS  PubMed  Google Scholar 

  61. Loprinzi PD, Kohli M. Effect of physical activity and sedentary behavior on serum prostate-specific antigen concentrations: results from the National Health and Nutrition Examination Survey (NHANES), 2003–2006. Mayo Clin Proc. 2013;88(1):11–21.

    CAS  PubMed  Google Scholar 

  62. Allott EH, Masko EM, Freedland SJ. Obesity and prostate cancer: weighing the evidence. Eur Urol. 2013;63(5):800–9.

    CAS  PubMed  Google Scholar 

  63. Cao Y, Giovannucci E. Obesity and prostate cancer. Recent Results Cancer Res. 2016;208:137–53.

    CAS  PubMed  Google Scholar 

  64. De Nunzio C, Andriole GL, Thompson IM Jr, Freedland SJ. Smoking and prostate cancer: a systematic review. Eur Urol Focus. 2015;1(1):28–38.

    PubMed  Google Scholar 

  65. Jones MR, Joshu CE, Kanarek N, Navas-Acien A, Richardson KA, Platz EA. Cigarette smoking and prostate cancer mortality in four US States, 1999–2010. Prev Chronic Dis. 2016;13:E51.

    PubMed  PubMed Central  Google Scholar 

  66. Salciccia S, Capriotti AL, Laganà A, Fais S, Logozzi M, De Berardinis E, Busetto GM, Di Pierro GB, Ricciuti GP, Del Giudice F, et al. Biomarkers in prostate cancer diagnosis: from current knowledge to the role of metabolomics and exosomes. Int J Mol Sci. 2021;22:9.

    Google Scholar 

  67. Olleik G, Kassouf W, Aprikian A, Hu J, Vanhuyse M, Cury F, Peacock S, Bonnevier E, Palenius E, Dragomir A. Evaluation of new tests and interventions for prostate cancer management: a systematic review. J Natl Compr Canc Netw. 2018;16(11):1340–51.

    PubMed  Google Scholar 

  68. Saini S. PSA and beyond: alternative prostate cancer biomarkers. Cell Oncol. 2016;39(2):97–106.

    CAS  Google Scholar 

  69. Punnen S, Pavan N, Parekh DJ. Finding the wolf in sheep’s clothing: The 4Kscore is a novel blood test that can accurately identify the risk of aggressive prostate cancer. Rev Urol. 2015;17(1):3–13.

    PubMed  PubMed Central  Google Scholar 

  70. Meehan J, Gray M, Martínez-Pérez C, Kay C, McLaren D, Turnbull AK. Tissue- and liquid-based biomarkers in prostate cancer precision medicine. J Pers Med. 2021;11(7):664.

    PubMed  PubMed Central  Google Scholar 

  71. Chang EK, Gadzinski AJ, Nyame YA. Blood and urine biomarkers in prostate cancer: are we ready for reflex testing in men with an elevated prostate-specific antigen? Asian J Urol. 2021;8(4):343–53.

    PubMed  PubMed Central  Google Scholar 

  72. Lomas DJ, Ahmed HU. All change in the prostate cancer diagnostic pathway. Nat Rev Clin Oncol. 2020;17(6):372–81.

    PubMed  Google Scholar 

  73. Hatakeyama S, Yoneyama T, Tobisawa Y, Ohyama C. Recent progress and perspectives on prostate cancer biomarkers. Int J Clin Oncol. 2017;22(2):214–21.

    CAS  PubMed  Google Scholar 

  74. Cucchiara V, Cooperberg MR, Dall’Era M, Lin DW, Montorsi F, Schalken JA, Evans CP. Genomic markers in prostate cancer decision making. Eur Urol. 2018;73(4):572–82.

    PubMed  Google Scholar 

  75. Wojno KJ, Costa FJ, Cornell RJ, Small JD, Pasin E, Van Criekinge W, Bigley JW, Van Neste L. Reduced rate of repeated prostate biopsies observed in confirmMDx clinical utility field study. Am Health Drug Benefits. 2014;7(3):129–34.

    PubMed  PubMed Central  Google Scholar 

  76. Matuszczak M, Schalken JA, Salagierski M. Prostate cancer liquid biopsy biomarkers’ clinical utility in diagnosis and prognosis. Cancers (Basel). 2021;13:13.

    Google Scholar 

  77. Loeb S, Catalona WJ. The Prostate Health Index: a new test for the detection of prostate cancer. Ther Adv Urol. 2014;6(2):74–7.

    PubMed  PubMed Central  Google Scholar 

  78. de la Calle C, Patil D, Wei JT, Scherr DS, Sokoll L, Chan DW, Siddiqui J, Mosquera JM, Rubin MA, Sanda MG. Multicenter evaluation of the prostate health index to detect aggressive prostate cancer in biopsy Naïve Men. J Urol. 2015;194(1):65–72.

    PubMed  PubMed Central  Google Scholar 

  79. White J, Shenoy BV, Tutrone RF, Karsh LI, Saltzstein DR, Harmon WJ, Broyles DL, Roddy TE, Lofaro LR, Paoli CJ, et al. Clinical utility of the Prostate Health Index (phi) for biopsy decision management in a large group urology practice setting. Prostate Cancer Prostatic Dis. 2018;21(1):78–84.

    PubMed  Google Scholar 

  80. Parekh DJ, Punnen S, Sjoberg DD, Asroff SW, Bailen JL, Cochran JS, Concepcion R, David RD, Deck KB, Dumbadze I, et al. A multi-institutional prospective trial in the USA confirms that the 4Kscore accurately identifies men with high-grade prostate cancer. Eur Urol. 2015;68(3):464–70.

    PubMed  Google Scholar 

  81. Darst BF, Chou A, Wan P, Pooler L, Sheng X, Vertosick EA, Conti DV, Wilkens LR, Le Marchand L, Vickers AJ, et al. The Four-Kallikrein panel is effective in identifying aggressive prostate cancer in a multiethnic population. Cancer Epidemiol Biomarkers Prev. 2020;29(7):1381–8.

    PubMed  PubMed Central  Google Scholar 

  82. Voigt JD, Dong Y, Linder V, Zappala S. Use of the 4Kscore test to predict the risk of aggressive prostate cancer prior to prostate biopsy: Overall cost savings and improved quality of care to the us healthcare system. Rev Urol. 2017;19(1):1–10.

    PubMed  PubMed Central  Google Scholar 

  83. Voigt JD, Zappala SM, Vaughan ED, Wein AJ. The Kallikrein Panel for prostate cancer screening: its economic impact. Prostate. 2014;74(3):250–9.

    PubMed  Google Scholar 

  84. Nordström T, Vickers A, Assel M, Lilja H, Grönberg H, Eklund M. Comparison between the four-kallikrein panel and prostate health index for predicting prostate cancer. Eur Urol. 2015;68(1):139–46.

    PubMed  Google Scholar 

  85. Merola R, Tomao L, Antenucci A, Sperduti I, Sentinelli S, Masi S, Mandoj C, Orlandi G, Papalia R, Guaglianone S, et al. PCA3 in prostate cancer and tumor aggressiveness detection on 407 high-risk patients: a National Cancer Institute experience. J Exp Clin Cancer Res. 2015;34(1):15.

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Rodríguez SVM, García-Perdomo HA. Diagnostic accuracy of prostate cancer antigen 3 (PCA3) prior to first prostate biopsy: A systematic review and meta-analysis. Can Urol Assoc J. 2020;14(5):E214-e219.

    PubMed  Google Scholar 

  87. Tutrone R, Donovan MJ, Torkler P, Tadigotla V, McLain T, Noerholm M, Skog J, McKiernan J. Clinical utility of the exosome based ExoDx Prostate(IntelliScore) EPI test in men presenting for initial Biopsy with a PSA 2–10 ng/mL. Prostate Cancer Prostatic Dis. 2020;23(4):607–14.

    CAS  PubMed  PubMed Central  Google Scholar 

  88. McKiernan J, Donovan MJ, Margolis E, Partin A, Carter B, Brown G, Torkler P, Noerholm M, Skog J, Shore N, et al. A prospective adaptive utility trial to validate performance of a novel urine exosome gene expression assay to predict high-grade prostate cancer in patients with prostate-specific antigen 2–10ng/ml at initial biopsy. Eur Urol. 2018;74(6):731–8.

    CAS  PubMed  Google Scholar 

  89. Hendriks RJ, van Leest MMG, Israël B, Hannink G, YantiSetiasti A, Cornel EB, Hulsbergen-vandeKaa CA, Klaver OS, Sedelaar JPM, Van Criekinge W, et al. Clinical use of the SelectMDx urinary-biomarker test with or without mpMRI in prostate cancer diagnosis: a prospective, multicenter study in biopsy-naïve men. Prostate Cancer Prostatic Dis. 2021;24(4):1110–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Van Neste L, Hendriks RJ, Dijkstra S, Trooskens G, Cornel EB, Jannink SA, de Jong H, Hessels D, Smit FP, Melchers WJ, et al. Detection of high-grade prostate cancer using a urinary molecular biomarker-based risk score. Eur Urol. 2016;70(5):740–8.

    PubMed  Google Scholar 

  91. Haese A, Trooskens G, Steyaert S, Hessels D, Brawer M, Vlaeminck-Guillem V, Ruffion A, Tilki D, Schalken J, Groskopf J, Van Criekinge W. Multicenter optimization and validation of a 2-gene mRNA urine test for detection of clinically significant prostate cancer before initial prostate biopsy. J Urol. 2019;202(2):256–63.

    PubMed  Google Scholar 

  92. Govers TM, Hessels D, Vlaeminck-Guillem V, Schmitz-Dräger BJ, Stief CG, Martinez-Ballesteros C, Ferro M, Borque-Fernando A, Rubio-Briones J, Sedelaar JPM, et al. Cost-effectiveness of SelectMDx for prostate cancer in four European countries: a comparative modeling study. Prostate Cancer Prostatic Dis. 2019;22(1):101–9.

    CAS  PubMed  Google Scholar 

  93. Eng SE, Basasie B, Lam A, John Semmes O, Troyer DA, Clarke GD, Sunnapwar AG, Leach RJ, Johnson-Pais TL, Sokoll LJ, et al. Prospective comparison of restriction spectrum imaging and non-invasive biomarkers to predict upgrading on active surveillance prostate biopsy. Prostate Cancer Prostatic Dis. 2022. https://doi.org/10.1038/s41391-022-00591-w.

    Article  PubMed  Google Scholar 

  94. Henning GM, Andriole GL, Kim EH. Liquid biomarkers for early detection of prostate cancer and summary of available data for their use in African-American men. Prostate Cancer Prostatic Dis. 2022;25(2):180–6.

    CAS  PubMed  Google Scholar 

  95. Young A, Palanisamy N, Siddiqui J, Wood DP, Wei JT, Chinnaiyan AM, Kunju LP, Tomlins SA. Correlation of urine TMPRSS2:ERG and PCA3 to ERG+ and total prostate cancer burden. Am J Clin Pathol. 2012;138(5):685–96.

    PubMed  Google Scholar 

  96. Sanda MG, Dunn RL, Michalski J, Sandler HM, Northouse L, Hembroff L, Lin X, Greenfield TK, Litwin MS, Saigal CS, et al. Quality of life and satisfaction with outcome among prostate-cancer survivors. N Engl J Med. 2008;358(12):1250–61.

    CAS  PubMed  Google Scholar 

  97. Tomlins SA, Day JR, Lonigro RJ, Hovelson DH, Siddiqui J, Kunju LP, Dunn RL, Meyer S, Hodge P, Groskopf J, et al. Urine TMPRSS2:ERG Plus PCA3 for individualized prostate cancer risk assessment. Eur Urol. 2016;70(1):45–53.

    CAS  PubMed  Google Scholar 

  98. Raja N, Russell CM, George AK. Urinary markers aiding in the detection and risk stratification of prostate cancer. Transl Androl Urol. 2018;7(Suppl 4):S436-s442.

    PubMed  PubMed Central  Google Scholar 

  99. Partin AW, Van Neste L, Klein EA, Marks LS, Gee JR, Troyer DA, Rieger-Christ K, Jones JS, Magi-Galluzzi C, Mangold LA, et al. Clinical validation of an epigenetic assay to predict negative histopathological results in repeat prostate biopsies. J Urol. 2014;192(4):1081–7.

    PubMed  PubMed Central  Google Scholar 

  100. Stewart GD, Van Neste L, Delvenne P, Delrée P, Delga A, McNeill SA, O’Donnell M, Clark J, Van Criekinge W, Bigley J, Harrison DJ. Clinical utility of an epigenetic assay to detect occult prostate cancer in histopathologically negative biopsies: results of the MATLOC study. J Urol. 2013;189(3):1110–6.

    PubMed  Google Scholar 

  101. Yonover P, Steyaert S, Cohen JJ, Ruiz C, Grafczynska K, Garcia E, DeHart J, Brawer M, Groskopf J, Criekinge WV. Clinical utility study of confirms mdx for prostate cancer in a community urology practice. J Clin Oncol. 2019;37(7):94–94.

    Google Scholar 

  102. Raju GSR, Pavitra E, Bandaru SS, Varaprasad GL, Nagaraju GP, Malla RR, Huh YS, Han Y-K. HOTAIR: a potential metastatic, drug-resistant and prognostic regulator of breast cancer. Mol Cancer. 2023;22(1):65.

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Li T, Liu N, Gao Y, Quan Z, Hao Y, Yu C, Li L, Yuan M, Niu L, Luo C, Wu X. Long noncoding RNA HOTAIR regulates the invasion and metastasis of prostate cancer by targeting hepaCAM. Br J Cancer. 2021;124(1):247–58.

    CAS  PubMed  Google Scholar 

  104. Alarcón-Zendejas AP, Scavuzzo A, Jiménez-Ríos MA, Álvarez-Gómez RM, Montiel-Manríquez R, Castro-Hernández C, Jiménez-Dávila MA, Pérez-Montiel D, González-Barrios R, Jiménez-Trejo F, et al. The promising role of new molecular biomarkers in prostate cancer: from coding and non-coding genes to artificial intelligence approaches. Prostate Cancer Prostatic Dis. 2022;25(3):431–43.

    PubMed  PubMed Central  Google Scholar 

  105. Feng D, Zhu W, Shi X, Xiong Q, Li D, Wei W, Han P, Wei Q, Yang L. Spindle and kinetochore-associated complex subunit 3 could serve as a prognostic biomarker for prostate cancer. Exp Hematol Oncol. 2022;11(1):76.

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Ionescu F, Zhang J, Wang L. Clinical applications of liquid biopsy in prostate cancer: from screening to predictive biomarker. Cancers (Basel). 2022;14(7):1728.

    CAS  PubMed  Google Scholar 

  107. Nikanjam M, Kato S, Kurzrock R. Liquid biopsy: current technology and clinical applications. J Hematol Oncol. 2022;15(1):131.

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Baeuerle PA, Gires O. EpCAM (CD326) finding its role in cancer. Br J Cancer. 2007;96(3):417–23.

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Liu Y, Wang Y, Sun S, Chen Z, Xiang S, Ding Z, Huang Z, Zhang B. Understanding the versatile roles and applications of EpCAM in cancers: from bench to bedside. Exp Hematol Oncol. 2022;11(1):97.

    PubMed  PubMed Central  Google Scholar 

  110. Zainfeld D, Goldkorn A. Liquid biopsy in prostate cancer: circulating tumor cells and beyond. Cancer Treat Res. 2018;175:87–104.

    PubMed  PubMed Central  Google Scholar 

  111. Magbanua MJM, Sosa EV, Scott JH, Simko J, Collins C, Pinkel D, Ryan CJ, Park JW. Isolation and genomic analysis of circulating tumor cells from castration resistant metastatic prostate cancer. BMC Cancer. 2012;12(1):78.

    PubMed  PubMed Central  Google Scholar 

  112. Magbanua MJM, Sosa EV, Roy R, Eisenbud LE, Scott JH, Olshen A, Pinkel D, Rugo HS, Park JW. Genomic profiling of isolated circulating tumor cells from metastatic breast cancer patients. Cancer Res. 2013;73(1):30–40.

    CAS  PubMed  Google Scholar 

  113. Cieślikowski WA, Antczak A, Nowicki M, Zabel M, Budna-Tukan J. Clinical relevance of circulating tumor cells in prostate cancer management. Biomedicines. 2021;9(9):45.

    Google Scholar 

  114. Hirano H, Nagata M, Nagaya N, Nakamura S, Ashizawa T, Lu Y, Kawano H, Kitamura K, Sakamoto Y, Fujita K, et al. Bone scan index (BSI) scoring by using bone scintigraphy and circulating tumor cells (CTCs): predictive factors for enzalutamide effectiveness in patients with castration-resistant prostate cancer and bone metastases. Sci Rep. 2023;13(1):8704.

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Khoo A, Liu LY, Sadun TY, Salmasi A, Pooli A, Felker E, Houlahan KE, Ignatchenko V, Raman SS, Sisk AE Jr, et al. Prostate cancer multiparametric magnetic resonance imaging visibility is a tumor-intrinsic phenomena. J Hematol Oncol. 2022;15(1):48.

    PubMed  PubMed Central  Google Scholar 

  116. Midiri F, Vernuccio F, Purpura P, Alongi P, Bartolotta TV. Multiparametric MRI and radiomics in prostate cancer: a review of the current literature. Diagnostics (Basel, Switzerland). 2021;11(10):34991.

    Google Scholar 

  117. Chen FZ, Zhao XK. Prostate cancer: current treatment and prevention strategies. Iran Red Crescent Med J. 2013;15(4):279–84.

    PubMed  PubMed Central  Google Scholar 

  118. Aurilio G, Cimadamore A, Mazzucchelli R, Lopez-Beltran A, Verri E, Scarpelli M, Massari F, Cheng L, Santoni M, Montironi R. Androgen receptor signaling pathway in prostate cancer: from genetics to clinical applications. Cells. 2020;9(12):2653.

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Litwin MS, Tan HJ. The diagnosis and treatment of prostate cancer: a review. JAMA. 2017;317(24):2532–42.

    PubMed  Google Scholar 

  120. Azarenko O, Smiyun G, Mah J, Wilson L, Jordan MA. Antiproliferative mechanism of action of the novel taxane cabazitaxel as compared with the parent compound docetaxel in MCF7 breast cancer cells. Mol Cancer Ther. 2014;13(8):2092–103.

    CAS  PubMed  Google Scholar 

  121. James ND, Sydes MR, Clarke NW, Mason MD, Dearnaley DP, Spears MR, Ritchie AW, Parker CC, Russell JM, Attard G, et al. Addition of docetaxel, zoledronic acid, or both to first-line long-term hormone therapy in prostate cancer (STAMPEDE): survival results from an adaptive, multiarm, multistage, platform randomised controlled trial. Lancet. 2016;387(10024):1163–77.

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Seruga B, Tannock IF. Chemotherapy-based treatment for castration-resistant prostate cancer. J Clin Oncol. 2011;29(27):3686–94.

    CAS  PubMed  Google Scholar 

  123. Vrignaud P, Sémiond D, Lejeune P, Bouchard H, Calvet L, Combeau C, Riou J-F, Commerçon A, Lavelle F, Bissery M-C. Preclinical antitumor activity of cabazitaxel, a semisynthetic taxane active in taxane-resistant tumors. Clin Cancer Res. 2013;19(11):2973–83.

    CAS  PubMed  Google Scholar 

  124. Rouyer M, Oudard S, Joly F, Fizazi K, Tubach F, Jove J, Lacueille C, Lamarque S, Guiard E, Balestra A, et al. Overall and progression-free survival with cabazitaxel in metastatic castration-resistant prostate cancer in routine clinical practice: the FUJI cohort. Br J Cancer. 2019;121(12):1001–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Parente P, Ng S, Parnis F, Guminski A, Gurney H. Cabazitaxel in patients with metastatic castration-resistant prostate cancer: safety and quality of life data from the Australian early access program. Asia Pac J Clin Oncol. 2017;13(6):391–9.

    PubMed  Google Scholar 

  126. Li C, Sun H, Wei W, Liu Q, Wang Y, Zhang Y, Lian F, Liu F, Li C, Ying K, et al. Mitoxantrone triggers immunogenic prostate cancer cell death via p53-dependent PERK expression. Cell Oncol. 2020;43(6):1099–116.

    CAS  Google Scholar 

  127. Green AK, Corty RW, Wood WA, Meeneghan M, Reeder-Hayes KE, Basch E, Milowsky MI, Dusetzina SB. Comparative effectiveness of mitoxantrone plus prednisone versus prednisone alone in metastatic castrate-resistant prostate cancer after docetaxel failure. Oncologist. 2015;20(5):516–22.

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Devos G, Devlies W, De Meerleer G, Baldewijns M, Gevaert T, Moris L, Milonas D, Van Poppel H, Berghen C, Everaerts W, et al. Neoadjuvant hormonal therapy before radical prostatectomy in high-risk prostate cancer. Nat Rev Urol. 2021;18(12):739–62.

    CAS  PubMed  Google Scholar 

  129. Nuvola G, Santoni M, Rizzo M, Rosellini M, Mollica V, Rizzo A, Marchetti A, Battelli N, Massari F. Adapting to hormone-therapy resistance for adopting the right therapeutic strategy in advanced prostate cancer. Expert Rev Anticancer Ther. 2023;23(6):593–600.

    CAS  PubMed  Google Scholar 

  130. Attard G, Murphy L, Clarke NW, Sachdeva A, Jones C, Hoyle A, Cross W, Jones RJ, Parker CC, Gillessen S, et al. Abiraterone acetate plus prednisolone with or without enzalutamide for patients with metastatic prostate cancer starting androgen deprivation therapy: final results from two randomised phase 3 trials of the STAMPEDE platform protocol. Lancet Oncol. 2023;24(5):443–56.

    CAS  PubMed  Google Scholar 

  131. Fizazi K, Foulon S, Carles J, Roubaud G, McDermott R, Fléchon A, Tombal B, Supiot S, Berthold D, Ronchin P, et al. Abiraterone plus prednisone added to androgen deprivation therapy and docetaxel in de novo metastatic castration-sensitive prostate cancer (PEACE-1): a multicentre, open-label, randomised, phase 3 study with a 2 × 2 factorial design. Lancet. 2022;399(10336):1695–707.

    CAS  PubMed  Google Scholar 

  132. Di Stefano RF, Tucci M, Turco F, Samuelly A, Bungaro M, Pisano C, Vignani F, Gallicchio M, Scagliotti GV, Di Maio M, Buttigliero C. Prognostic role of the duration of response to androgen deprivation therapy in patients with metastatic castration resistant prostate cancer treated with enzalutamide or abiraterone acetate. Prostate Cancer Prostatic Dis. 2021;24(3):812–25.

    PubMed  Google Scholar 

  133. Yin L, Hu Q. CYP17 inhibitors—abiraterone, C17,20-lyase inhibitors and multi-targeting agents. Nat Rev Urol. 2014;11(1):32–42.

    CAS  PubMed  Google Scholar 

  134. Padmakar Darne C, Velaparthi U, Saulnier M, Frennesson D, Liu P, Huang A, Tokarski J, Fura A, Spires T, Newitt J, et al. The discovery of BMS-737 as a potent, CYP17 lyase-selective inhibitor for the treatment of castration-resistant prostate cancer. Bioorg Med Chem Lett. 2022;75: 128951.

    CAS  PubMed  Google Scholar 

  135. Wróbel TM, Jørgensen FS, Pandey AV, Grudzińska A, Sharma K, Yakubu J, Björkling F. Non-steroidal CYP17A1 Inhibitors: Discovery and Assessment. J Med Chem. 2023;66(10):6542–66.

    PubMed  PubMed Central  Google Scholar 

  136. Hatano K, Nonomura N. Systemic therapies for metastatic castration-resistant prostate cancer: an updated review. World J Mens Health. 2023;41:89.

    Google Scholar 

  137. de Bono JS, Logothetis CJ, Molina A, Fizazi K, North S, Chu L, Chi KN, Jones RJ, Goodman OB, Saad F, et al. Abiraterone and increased survival in metastatic prostate cancer. N Engl J Med. 2011;364(21):1995–2005.

    PubMed  PubMed Central  Google Scholar 

  138. Hall ME, Padgett WJ, Klaassen Z, Magee DE, Luckenbaugh AN, Laviana AA, Satkunasivam R, Schaffer K, Wallis CJD. Association between RCT methodology and disease indication with mineralocorticoid-related toxicity for patients receiving abiraterone acetate for advanced prostate cancer: A meta-analysis of RCTs. Clin Genitourin Cancer. 2023. https://doi.org/10.1016/j.clgc.2023.04.007.

    Article  PubMed  Google Scholar 

  139. Pezaro CJ, Mukherji D, De Bono JS. Abiraterone acetate: redefining hormone treatment for advanced prostate cancer. Drug Discovery Today. 2012;17(5):221–6.

    CAS  PubMed  Google Scholar 

  140. James ND, de Bono JS, Spears MR, Clarke NW, Mason MD, Dearnaley DP, Ritchie AWS, Amos CL, Gilson C, Jones RJ, et al. Abiraterone for prostate cancer not previously treated with hormone therapy. N Engl J Med. 2017;377(4):338–51.

    CAS  PubMed  PubMed Central  Google Scholar 

  141. Lorente D, Omlin A, Ferraldeschi R, Pezaro C, Perez R, Mateo J, Altavilla A, Zafeiriou Z, Tunariu N, Parker C, et al. Tumour responses following a steroid switch from prednisone to dexamethasone in castration-resistant prostate cancer patients progressing on abiraterone. Br J Cancer. 2014;111(12):2248–53.

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Hussain M, Fizazi K, Saad F, Rathenborg P, Shore N, Ferreira U, Ivashchenko P, Demirhan E, Modelska K, Phung D, et al. Enzalutamide in men with nonmetastatic, castration-resistant prostate cancer. N Engl J Med. 2018;378(26):2465–74.

    CAS  PubMed  PubMed Central  Google Scholar 

  143. Hoffman-Censits J, Kelly WK. Enzalutamide: a novel antiandrogen for patients with castrate-resistant prostate cancer. Clin Cancer Res. 2013;19(6):1335–9.

    CAS  PubMed  Google Scholar 

  144. Vanneste BGL, Van Limbergen EJ, van Lin EN, van Roermund JGH, Lambin P. Prostate cancer radiation therapy: what do clinicians have to know? BioMed Res Int. 2016;2016:6829875.

    PubMed  PubMed Central  Google Scholar 

  145. Guérif S, Latorzeff I, Lagrange JL, Hennequin C, Supiot S, Garcia A, François P, Soulié M, Richaud P, Salomon L. Postoperative radiotherapy of prostate cancer. Cancer Radiother. 2014;18(5–6):517–23.

    PubMed  Google Scholar 

  146. Ferini G, Pergolizzi S. A ten-year-long update on radiation proctitis among prostate cancer patients treated with curative external beam radiotherapy. In Vivo. 2021;35(3):1379–91.

    PubMed  PubMed Central  Google Scholar 

  147. Guo X, Xia H, Su X, Hou H, Zhong Q, Wang J. Comparing the survival outcomes of radical prostatectomy versus radiotherapy for patients with de novo metastasis prostate cancer: a population-based study. Front Oncol. 2021;11: 797462.

    PubMed  PubMed Central  Google Scholar 

  148. Parker C, Nilsson S, Heinrich D, Helle SI, O’Sullivan JM, Fosså SD, Chodacki A, Wiechno P, Logue J, Seke M, et al. Alpha emitter radium-223 and survival in metastatic prostate cancer. N Engl J Med. 2013;369(3):213–23.

    CAS  PubMed  Google Scholar 

  149. Deshayes E, Roumiguie M, Thibault C, Beuzeboc P, Cachin F, Hennequin C, Huglo D, Rozet F, Kassab-Chahmi D, Rebillard X, Houédé N. Radium 223 dichloride for prostate cancer treatment. Drug Des Devel Ther. 2017;11:2643–51.

    CAS  PubMed  PubMed Central  Google Scholar 

  150. Wissing MD, van Leeuwen FW, van der Pluijm G, Gelderblom H. Radium-223 chloride: extending life in prostate cancer patients by treating bone metastases. Clin Cancer Res. 2013;19(21):5822–7.

    CAS  PubMed  Google Scholar 

  151. Shang T, Yu X, Han S, Yang B. Nanomedicine-based tumor photothermal therapy synergized immunotherapy. Biomater Sci. 2020;8(19):5241–59.

    CAS  PubMed  Google Scholar 

  152. Gu C, Li C, Zhang J, Li X, Wang L, Ju Y, Liu Y, Xu Y. Ultra-effective near-infrared Photothermal therapy for the prostate cancer Nursing care through novel intended and surface tailored photo-responsive Ga-Au@MPS nanovesicles. J Photochem Photobiol, B. 2020;202: 111685.

    CAS  PubMed  Google Scholar 

  153. Guo X, Cao B, Wang C, Lu S, Hu X. In vivo photothermal inhibition of methicillin-resistant Staphylococcus aureus infection by in situ templated formulation of pathogen-targeting phototheranostics. Nanoscale. 2020;12(14):7651–9.

    CAS  PubMed  Google Scholar 

  154. Deng X, Shao Z, Zhao Y. Solutions to the drawbacks of photothermal and photodynamic cancer therapy. Adv Sci. 2021;8(3):2002504.

    CAS  Google Scholar 

  155. Ding D, Zhong H, Liang R, Lan T, Zhu X, Huang S, Wang Y, Shao J, Shuai X, Wei B. Multifunctional nanodrug mediates synergistic photodynamic therapy and MDSCs-targeting immunotherapy of colon cancer. Adv Sci. 2021;8(14):2100712.

    CAS  Google Scholar 

  156. Zheng Y, Li Z, Yang Y, Shi H, Chen H, Gao Y. A nanosensitizer self-assembled from oleanolic acid and chlorin e6 for synergistic chemo/sono-photodynamic cancer therapy. Phytomedicine. 2021;93: 153788.

    CAS  PubMed  Google Scholar 

  157. Liu T, Song Y, Huang Z, Pu X, Wang Y, Yin G, Gou L, Weng J, Meng X. Photothermal photodynamic therapy and enhanced radiotherapy of targeting copolymer-coated liquid metal nanoparticles on liver cancer. Colloids Surf, B. 2021;207: 112023.

    CAS  Google Scholar 

  158. Poudel BK, Soe ZC, Ruttala HB, Gupta B, Ramasamy T, Thapa RK, Gautam M, Ou W, Nguyen HT, Jeong J-H, et al. In situ fabrication of mesoporous silica-coated silver-gold hollow nanoshell for remotely controllable chemo-photothermal therapy via phase-change molecule as gatekeepers. Int J Pharm. 2018;548(1):92–103.

    CAS  PubMed  Google Scholar 

  159. Plan Sangnier A, Aufaure R, Motte L, Wilhelm C, Guenin E, Lalatonne Y. Hybrid Au@alendronate nanoparticles as dual chemo-photothermal agent for combined cancer treatment. Beilstein J Nanotechnol. 2018;9:2947–52.

    PubMed  PubMed Central  Google Scholar 

  160. Weersink R, Chaudhary S, Mayo K, He J, Wilson B. Shape-based reconstruction for transrectal diffuse optical tomography monitoring of photothermal focal therapy of prostate cancer: simulation studies. J Biomed Opt. 2017;22(4): 045004.

    Google Scholar 

  161. Li S, Shi M, Zhao J, Zhang L, Huang Y, Zhao S. A highly sensitive capillary electrophoresis immunoassay strategy based on dual-labeled gold nanoparticles enhancing chemiluminescence for the detection of prostate-specific antigen. Electrophoresis. 2017;38(13–14):1780–7.

    CAS  PubMed  Google Scholar 

  162. Poudel K, Thapa RK, Gautam M, Ou W, Soe ZC, Gupta B, Ruttala HB, Thuy HN, Dai PC, Jeong J-H, et al. Multifaceted NIR-responsive polymer-peptide-enveloped drug-loaded copper sulfide nanoplatform for chemo-phototherapy against highly tumorigenic prostate cancer. Nanomedicine. 2019;21: 102042.

    CAS  PubMed  Google Scholar 

  163. Rhea LP, Mendez-Marti S, Kim D, Aragon-Ching JB. Role of immunotherapy in bladder cancer. Cancer Treat Res Commun. 2021;26: 100296.

    PubMed  Google Scholar 

  164. Braun DA, Bakouny Z, Hirsch L, Flippot R, Van Allen EM, Wu CJ, Choueiri TK. Beyond conventional immune-checkpoint inhibition—novel immunotherapies for renal cell carcinoma. Nat Rev Clin Oncol. 2021;18(4):199–214.

    CAS  PubMed  PubMed Central  Google Scholar 

  165. Bansal D, Reimers MA, Knoche EM, Pachynski RK. Immunotherapy and immunotherapy combinations in metastatic castration-resistant prostate cancer. Cancers (Basel). 2021;13(2):89.

    Google Scholar 

  166. Galon J, Bruni D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat Rev Drug Discovery. 2019;18(3):197–218.

    CAS  PubMed  Google Scholar 

  167. Criscitiello C, Morganti S, Curigliano G. Antibody–drug conjugates in solid tumors: a look into novel targets. J Hematol Oncol. 2021;14(1):20.

    CAS  PubMed  PubMed Central  Google Scholar 

  168. Liu J, Fu M, Wang M, Wan D, Wei Y, Wei X. Cancer vaccines as promising immuno-therapeutics: platforms and current progress. J Hematol Oncol. 2022;15(1):28.

    PubMed  PubMed Central  Google Scholar 

  169. Prokhnevska N, Emerson DA, Kissick HT, Redmond WL. Immunological complexity of the prostate cancer microenvironment influences the response to immunotherapy. Adv Exp Med Biol. 2019;1210:121–47.

    CAS  PubMed  Google Scholar 

  170. Uhlman MA, Bing MT, Lubaroff DM. Prostate cancer vaccines in combination with additional treatment modalities. Immunol Res. 2014;59(1):236–42.

    CAS  PubMed  Google Scholar 

  171. Adamaki M, Zoumpourlis V. Immunotherapy as a precision medicine tool for the treatment of prostate cancer. Cancers. 2021;13(2):173.

    CAS  PubMed  PubMed Central  Google Scholar 

  172. Yu J, Sun H, Cao W, Song Y, Jiang Z. Research progress on dendritic cell vaccines in cancer immunotherapy. Exp Hematol Oncol. 2022;11(1):3.

    CAS  PubMed  PubMed Central  Google Scholar 

  173. Li J, Li W, Huang K, Zhang Y, Kupfer G, Zhao Q. Chimeric antigen receptor T cell (CAR-T) immunotherapy for solid tumors: lessons learned and strategies for moving forward. J Hematol Oncol. 2018;11(1):22.

    CAS  PubMed  PubMed Central  Google Scholar 

  174. Sterner RC, Sterner RM. CAR-T cell therapy: current limitations and potential strategies. Blood Cancer J. 2021;11(4):69.

    PubMed  PubMed Central  Google Scholar 

  175. Dagar G, Gupta A, Masoodi T, Nisar S, Merhi M, Hashem S, Chauhan R, Dagar M, Mirza S, Bagga P, et al. Harnessing the potential of CAR-T cell therapy: progress, challenges, and future directions in hematological and solid tumor treatments. J Transl Med. 2023;21(1):449.

    PubMed  PubMed Central  Google Scholar 

  176. Yan X, Chen D, Wang Y, Guo Y, Tong C, Wei J, Zhang Y, Wu Z, Han W. Identification of NOXA as a pivotal regulator of resistance to CAR T-cell therapy in B-cell malignancies. Signal Transduction Targeted Ther. 2022;7(1):98.

    CAS  Google Scholar 

  177. Hu Y, Li J, Ni F, Yang Z, Gui X, Bao Z, Zhao H, Wei G, Wang Y, Zhang M, et al. CAR-T cell therapy-related cytokine release syndrome and therapeutic response is modulated by the gut microbiome in hematologic malignancies. Nat Commun. 2022;13(1):5313.

    CAS  PubMed  PubMed Central  Google Scholar 

  178. Edeline J, Houot R, Marabelle A, Alcantara M. CAR-T cells and BiTEs in solid tumors: challenges and perspectives. J Hematol Oncol. 2021;14(1):65.

    PubMed  PubMed Central  Google Scholar 

  179. Perera MPJ, Thomas PB, Risbridger GP, Taylor R, Azad A, Hofman MS, Williams ED, Vela I. Chimeric antigen receptor T-cell therapy in metastatic castrate-resistant prostate cancer. Cancers (Basel). 2022;14(3):503.

    CAS  PubMed  Google Scholar 

  180. Yu S, Yi M, Qin S, Wu K. Next generation chimeric antigen receptor T cells: safety strategies to overcome toxicity. Mol Cancer. 2019;18(1):125.

    PubMed  PubMed Central  Google Scholar 

  181. Yang Y, Yang H, Alcaina Y, Puc J, Birt A, Vedvyas Y, Gallagher M, Alla S, Riascos MC, McCloskey JE, et al. Inducible expression of interleukin-12 augments the efficacy of affinity-tuned chimeric antigen receptors in murine solid tumor models. Nat Commun. 2023;14(1):2068.

    CAS  PubMed  PubMed Central  Google Scholar 

  182. Kheyrolahzadeh K, Tohidkia MR, Tarighatnia A, Shahabi P, Nader ND, Aghanejad A. Theranostic chimeric antigen receptor (CAR)-T cells: Insight into recent trends and challenges in solid tumors. Life Sci. 2023;328: 121917.

    CAS  PubMed  Google Scholar 

  183. Zhang A, Tong D. Immunotherapy in patients with metastatic castration-resistant prostate cancer: a meta-analysis of data from 7 phase III studies and 3 phase II studies. Exp Hematol Oncol. 2022;11(1):63.

    CAS  PubMed  PubMed Central  Google Scholar 

  184. Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, Redfern CH, Ferrari AC, Dreicer R, Sims RB, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363(5):411–22.

    CAS  PubMed  Google Scholar 

  185. Cheever MA, Higano CS. PROVENGE (Sipuleucel-T) in prostate cancer: the first FDA-approved therapeutic cancer vaccine. Clin Cancer Res. 2011;17(11):3520–6.

    PubMed  Google Scholar 

  186. Small EJ, Lance RS, Redfern CH, Millard FE, Gardner TA, Karsh LI, Dawson NA, McCoy C, Stubbs A, DeVries T, et al. A randomized phase II trial of sipuleucel-T with concurrent or sequential abiraterone acetate (AA) plus prednisone (P) in metastatic castrate-resistant prostate cancer (mCRPC). Clin Cancer Res. 2013;31(15):5047–5047.

    Google Scholar 

  187. Warren TL, Weiner GJ. Uses of granulocyte-macrophage colony-stimulating factor in vaccine development. Curr Opin Hematol. 2000;7(3):168–73.

    CAS  PubMed  Google Scholar 

  188. Simmons AD, Li B, Gonzalez-Edick M, Lin C, Moskalenko M, Du T, Creson J, VanRoey MJ, Jooss K. GM-CSF-secreting cancer immunotherapies: preclinical analysis of the mechanism of action. Cancer Immunol Immunother. 2007;56(10):1653–65.

    CAS  PubMed  Google Scholar 

  189. Handa S, Hans B, Goel S, Bashorun HO, Dovey Z, Tewari A. Immunotherapy in prostate cancer: current state and future perspectives. Ther Adv Urol. 2020;12:1756287220951404.

    CAS  PubMed  PubMed Central  Google Scholar 

  190. Gulley JL, Madan RA, Tsang KY, Jochems C, Marté JL, Farsaci B, Tucker JA, Hodge JW, Liewehr DJ, Steinberg SM, et al. Immune impact induced by PROSTVAC (PSA-TRICOM), a therapeutic vaccine for prostate cancer. Cancer Immunol Res. 2014;2(2):133–41.

    CAS  PubMed  Google Scholar 

  191. Gulley JL, Borre M, Vogelzang NJ, Ng S, Agarwal N, Parker CC, Pook DW, Rathenborg P, Flaig TW, Carles J, et al. Phase III trial of PROSTVAC in asymptomatic or minimally symptomatic metastatic castration-resistant prostate cancer. J Clin Oncol. 2019;37(13):1051–61.

    CAS  PubMed  PubMed Central  Google Scholar 

  192. Fu T, Dai L-J, Wu S-Y, Xiao Y, Ma D, Jiang Y-Z, Shao Z-M. Spatial architecture of the immune microenvironment orchestrates tumor immunity and therapeutic response. J Hematol Oncol. 2021;14(1):98.

    PubMed  PubMed Central  Google Scholar 

  193. Jia Q, Wang A, Yuan Y, Zhu B, Long H. Heterogeneity of the tumor immune microenvironment and its clinical relevance. Exp Hematol Oncol. 2022;11(1):24.

    CAS  PubMed  PubMed Central  Google Scholar 

  194. Granier C, De Guillebon E, Blanc C, Roussel H, Badoual C, Colin E, Saldmann A, Gey A, Oudard S, Tartour E. Mechanisms of action and rationale for the use of checkpoint inhibitors in cancer. ESMO Open. 2017;2(2): e000213.

    PubMed  PubMed Central  Google Scholar 

  195. Darvin P, Toor SM, Sasidharan Nair V, Elkord E. Immune checkpoint inhibitors: recent progress and potential biomarkers. Exp Mol Med. 2018;50(12):1–11.

    PubMed  Google Scholar 

  196. Francisco LM, Salinas VH, Brown KE, Vanguri VK, Freeman GJ, Kuchroo VK, Sharpe AH. PD-L1 regulates the development, maintenance, and function of induced regulatory T cells. J Exp Med. 2009;206(13):3015–29.

    CAS  PubMed  PubMed Central  Google Scholar 

  197. Yi M, Zheng X, Niu M, Zhu S, Ge H, Wu K. Combination strategies with PD-1/PD-L1 blockade: current advances and future directions. Mol Cancer. 2022;21(1):28.

    PubMed  PubMed Central  Google Scholar 

  198. Marin-Acevedo JA, Kimbrough EO, Lou Y. Next generation of immune checkpoint inhibitors and beyond. J Hematol Oncol. 2021;14(1):45.

    CAS  PubMed  PubMed Central  Google Scholar 

  199. Ju X, Zhang H, Zhou Z, Wang Q. Regulation of PD-L1 expression in cancer and clinical implications in immunotherapy. Am J Cancer Res. 2020;10(1):1–11.

    CAS  PubMed  PubMed Central  Google Scholar 

  200. Yi M, Niu M, Xu L, Luo S, Wu K. Regulation of PD-L1 expression in the tumor microenvironment. J Hematol Oncol. 2021;14(1):10.

    CAS  PubMed  PubMed Central  Google Scholar 

  201. Massari F, Ciccarese C, Caliò A, Munari E, Cima L, Porcaro AB, Novella G, Artibani W, Sava T, Eccher A, et al. Magnitude of PD-1, PD-L1 and T lymphocyte expression on tissue from castration-resistant prostate adenocarcinoma: an exploratory analysis. Target Oncol. 2016;11(3):345–51.

    PubMed  Google Scholar 

  202. Antonarakis ES, Isaacsson Velho P, Fu W, Wang H, Agarwal N, Sacristan Santos V, Maughan BL, Pili R, Adra N, Sternberg CN, et al. CDK12-altered prostate cancer: clinical features and therapeutic outcomes to standard systemic therapies, poly (ADP-Ribose) polymerase inhibitors, and PD-1 inhibitors. JCO Precis Oncol. 2020;4:370–81.

    PubMed  Google Scholar 

  203. Schaeffer E, Srinivas S, Antonarakis ES, Armstrong AJ, Bekelman JE, Cheng H, Amico AV, Davis BJ, Desai N, Dorff T, et al. NCCN guidelines insights: prostate cancer, version 1.2021. J Natl Compr Canc Netw. 2021;19(2):134–43.

    CAS  PubMed  Google Scholar 

  204. Wolchok JD, Chiarion-Sileni V, Gonzalez R, Rutkowski P, Grob J-J, Cowey CL, Lao CD, Wagstaff J, Schadendorf D, Ferrucci PF, et al. Overall survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med. 2017;377(14):1345–56.

    CAS  PubMed  PubMed Central  Google Scholar 

  205. Motzer RJ, Tannir NM, McDermott DF, Arén Frontera O, Melichar B, Choueiri TK, Plimack ER, Barthélémy P, Porta C, George S, et al. Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma. N Engl J Med. 2018;378(14):1277–90.

    CAS  PubMed  PubMed Central  Google Scholar 

  206. Joshi H, Pinski JK. Association of ARV7 expression with molecular and clinical characteristics in prostate cancer. J Clin Oncol. 2016;34(2):109–109.

    Google Scholar 

  207. Sharma P, Pachynski RK, Narayan V, Fléchon A, Gravis G, Galsky MD, Mahammedi H, Patnaik A, Subudhi SK, Ciprotti M, et al. Nivolumab plus ipilimumab for metastatic castration-resistant prostate cancer: preliminary analysis of patients in the CheckMate 650 Trial. Cancer Cell. 2020;38(4):489-499.e483.

    CAS  PubMed  Google Scholar 

  208. Chen L. Co-inhibitory molecules of the B7-CD28 family in the control of T-cell immunity. Nat Rev Immunol. 2004;4(5):336–47.

    CAS  PubMed  Google Scholar 

  209. Finkelstein SE, Salenius S, Mantz CA, Shore ND, Fernandez EB, Shulman J, Myslicki FA, Agassi AM, Rotterman Y, DeVries T, Sims R. Combining immunotherapy and radiation for prostate cancer. Clin Genitourin Cancer. 2015;13(1):1–9.

    PubMed  Google Scholar 

  210. Zhang S, Ma X, Zhu C, Liu L, Wang G, Yuan X. The role of myeloid-derived suppressor cells in patients with solid tumors: a meta-analysis. PLoS ONE. 2016;11(10): e0164514.

    PubMed  PubMed Central  Google Scholar 

  211. Fakhrejahani F, Madan RA, Dahut WL, Karzai F, Cordes LM, Schlom J, Gulley JL. Avelumab in metastatic castration-resistant prostate cancer (mCRPC). J Clin Oncol. 2017;35(6):159–159.

    Google Scholar 

  212. Jiao S, Xia W, Yamaguchi H, Wei Y, Chen M-K, Hsu J-M, Hsu JL, Yu W-H, Du Y, Lee H-H, et al. PARP inhibitor upregulates PD-L1 expression and enhances cancer-associated immunosuppression. Clin Cancer Res. 2017;23(14):3711–20.

    CAS  PubMed  PubMed Central  Google Scholar 

  213. Fenerty KE, Padget M, Wolfson B, Gameiro SR, Su Z, Lee JH, Rabizadeh S, Soon-Shiong P, Hodge JW. Immunotherapy utilizing the combination of natural killer- and antibody dependent cellular cytotoxicity (ADCC)-mediating agents with poly (ADP-ribose) polymerase (PARP) inhibition. J Immunother Cancer. 2018;6(1):133.

    PubMed  PubMed Central  Google Scholar 

  214. Tripathi M, Nandana S, Billet S, Cavassani KA, Mishra R, Chung LWK, Posadas EM, Bhowmick NA. Modulation of cabozantinib efficacy by the prostate tumor microenvironment. Oncotarget. 2017;8(50):87891–902.

    PubMed  PubMed Central  Google Scholar 

  215. van der Linden RR, Haagmans BL, Mongiat-Artus P, van Doornum GJ, Kraaij R, Kadmon D, Aguilar-Cordova E, Osterhaus AD, van der Kwast TH, Bangma CH. Virus specific immune responses after human neoadjuvant adenovirus-mediated suicide gene therapy for prostate cancer. Eur Urol. 2005;48(1):153–61.

    PubMed  Google Scholar 

  216. Lee JC, Shin DW, Park H, Kim J, Youn Y, Kim JH, Kim J, Hwang JH. Tolerability and safety of EUS-injected adenovirus-mediated double-suicide gene therapy with chemotherapy in locally advanced pancreatic cancer: a phase 1 trial. Gastrointest Endosc. 2020;92(5):1044-1052.e1041.

    PubMed  Google Scholar 

  217. Zhang Y, Wang Y, Meng L, Huang Q, Zhu Y, Cui W, Cheng Y, Liu R. Targeted micelles with chemotherapeutics and gene drugs to inhibit the G1/S and G2/M mitotic cycle of prostate cancer. J Nanobiotechnol. 2021;19(1):17.

    CAS  Google Scholar 

  218. Siddiqui S, Libertini SJ, Lucas CA, Lombard AP, Baek HB, Nakagawa RM, Nishida KS, Steele TM, Melgoza FU, Borowsky AD, et al. The p14ARF tumor suppressor restrains androgen receptor activity and prevents apoptosis in prostate cancer cells. Cancer Lett. 2020;483:12–21.

    CAS  PubMed  PubMed Central  Google Scholar 

  219. Khadem H, Kebriaei H, Veisi Z. Inactivation of tumor suppressor genes and cancer therapy: an evolutionary game theory approach. Math Biosci. 2017;288:84–93.

    CAS  PubMed  Google Scholar 

  220. Ghafouri-Fard S, Taheri M. Maternally expressed gene 3 (MEG3): a tumor suppressor long non coding RNA. Biomed Pharmacother. 2019;118: 109129.

    CAS  PubMed  Google Scholar 

  221. Cozar JM, Robles-Fernandez I, Rodriguez-Martinez A, Puche-Sanz I, Vazquez-Alonso F, Lorente JA, Martinez-Gonzalez LJ, Alvarez-Cubero MJ. The role of miRNAs as biomarkers in prostate cancer. Mutat Res, Rev Mutat Res. 2019;781:165–74.

    CAS  PubMed  Google Scholar 

  222. Cato L, de Tribolet-Hardy J, Lee I, Rottenberg JT, Coleman I, Melchers D, Houtman R, Xiao T, Li W, Uo T, et al. ARv7 represses tumor-suppressor genes in castration-resistant prostate cancer. Cancer Cell. 2019;35(3):401-413.e406.

    CAS  PubMed  PubMed Central  Google Scholar 

  223. Yang Y, Guo J-X, Shao Z-Q. miR-21 targets and inhibits tumor suppressor gene PTEN to promote prostate cancer cell proliferation and invasion: an experimental study. Asian Pac J Trop Med. 2017;10(1):87–91.

    CAS  PubMed  Google Scholar 

  224. Qian L, Shen Y, Xie J, Meng Z. Immunomodulatory effects of ablation therapy on tumors: potentials for combination with immunotherapy. Biochim Biophys Acta Rev Cancer. 2020;1874(1): 188385.

    CAS  PubMed  Google Scholar 

  225. Hutmacher C, Neri D. Antibody-cytokine fusion proteins: biopharmaceuticals with immunomodulatory properties for cancer therapy. Adv Drug Deliv Rev. 2019;141:67–91.

    CAS  PubMed  Google Scholar 

  226. Claps M, Mennitto A, Guadalupi V, Sepe P, Stellato M, Zattarin E, Gillessen SS, Sternberg CN, Berruti A, De Braud FGM, et al. Immune-checkpoint inhibitors and metastatic prostate cancer therapy: learning by making mistakes. Cancer Treat Rev. 2020;88: 102057.

    CAS  PubMed  Google Scholar 

  227. Leber MF, Neault S, Jirovec E, Barkley R, Said A, Bell JC, Ungerechts G. Engineering and combining oncolytic measles virus for cancer therapy. Cytokine Growth Factor Rev. 2020;56:39–48.

    CAS  PubMed  PubMed Central  Google Scholar 

  228. Bayne RS, Puckett S, Rodrigues LU, Cramer SD, Lee J, Furdui CM, Chou JW, Miller LD, Ornelles DA, Lyles DS. MAP3K7 and CHD1 are novel mediators of resistance to oncolytic vesicular stomatitis virus in prostate cancer cells. Mol Ther Oncolytics. 2020;17:496–507.

    CAS  PubMed  PubMed Central  Google Scholar 

  229. Kemp V, Lamfers MLM, van der Pluijm G, van den Hoogen BG, Hoeben RC. Developing oncolytic viruses for clinical use: a consortium approach. Cytokine Growth Factor Rev. 2020;56:133–40.

    CAS  PubMed  Google Scholar 

  230. Lu Y. Transcriptionally regulated, prostate-targeted gene therapy for prostate cancer. Adv Drug Deliv Rev. 2009;61(7–8):572–88.

    CAS  PubMed  Google Scholar 

  231. Zare EN, Zheng X, Makvandi P, Gheybi H, Sartorius R, Yiu CKY, Adeli M, Wu A, Zarrabi A, Varma RS, Tay FR. Nonspherical metal-based nanoarchitectures: synthesis and impact of size, shape, and composition on their biological activity. Small. 2021;17(17):2007073.

    CAS  Google Scholar 

  232. Sharifi E, Bigham A, Yousefiasl S, Trovato M, Ghomi M, Esmaeili Y, Samadi P, Zarrabi A, Ashrafizadeh M, Sharifi S, et al. Mesoporous bioactive glasses in cancer diagnosis and therapy: stimuli-responsive, toxicity, immunogenicity, and clinical translation. Adv Sci. 2022;9(2):2102678.

    CAS  Google Scholar 

  233. Ghahremanloo A, Zare EN, Salimi F, Makvandi P. Electroconductive and photoactive poly(phenylenediamine)s with antioxidant and antimicrobial activities for potential photothermal therapy. New J Chem. 2022;46(13):6255–66.

    CAS  Google Scholar 

  234. Tabasi H, Babaei M, Abnous K, Taghdisi SM, Saljooghi AS, Ramezani M, Alibolandi M. Metal–polymer-coordinated complexes as potential nanovehicles for drug delivery. J Nanostruct Chem. 2021;11(4):501–26.

    CAS  Google Scholar 

  235. Tavakoli N, Divsalar A, Haertlé T, Sawyer L, Saboury AA, Muronetz V. Milk protein-based nanodelivery systems for the cancer treatment. J Nanostruct Chem. 2021;11(4):483–500.

    CAS  Google Scholar 

  236. Ashrafizadeh M, Mirzaei S, Gholami MH, Hashemi F, Zabolian A, Raei M, Hushmandi K, Zarrabi A, Voelcker NH, Aref AR, et al. Hyaluronic acid-based nanoplatforms for Doxorubicin: A review of stimuli-responsive carriers, co-delivery and resistance suppression. Carbohydr Polym. 2021;272: 118491.

    CAS  PubMed  Google Scholar 

  237. Tian H, Zhang T, Qin S, Huang Z, Zhou L, Shi J, Nice EC, Xie N, Huang C, Shen Z. Enhancing the therapeutic efficacy of nanoparticles for cancer treatment using versatile targeted strategies. J Hematol Oncol. 2022;15(1):132.

    PubMed  PubMed Central  Google Scholar 

  238. Gao X, Li L, Cai X, Huang Q, Xiao J, Cheng Y. Targeting nanoparticles for diagnosis and therapy of bone tumors: opportunities and challenges. Biomaterials. 2021;265: 120404.

    CAS  PubMed  Google Scholar 

  239. Nooranian S, Mohammadinejad A, Mohajeri T, Aleyaghoob G, Kazemi OR. Biosensors based on aptamer-conjugated gold nanoparticles: a review. Biotechnol Appl Biochem. 2022;69(4):1517–34.

    CAS  PubMed  Google Scholar 

  240. Martin JA, Smith JE, Warren M, Chávez JL, Hagen JA, Kelley-Loughnane N. A method for selecting structure-switching aptamers applied to a colorimetric gold nanoparticle assay. J Visualized Exp. 2015;96: e52545.

    Google Scholar 

  241. Fang Y, Lin S, Yang F, Situ J, Lin S, Luo Y. Aptamer-conjugated multifunctional polymeric nanoparticles as cancer-targeted, MRI-ultrasensitive drug delivery systems for treatment of castration-resistant prostate cancer. BioMed Res Int. 2020;2020:9186583.

    PubMed  PubMed Central  Google Scholar 

  242. Yu MK, Kim D, Lee I-H, So J-S, Jeong YY, Jon S. Image-guided prostate cancer therapy using aptamer-functionalized thermally cross-linked superparamagnetic iron oxide nanoparticles. Small. 2011;7(15):2241–9.

    CAS  PubMed  Google Scholar 

  243. Chen Y, Deng Y, Zhu C, Xiang C. Anti prostate cancer therapy: aptamer-functionalized, curcumin and cabazitaxel co-delivered, tumor targeted lipid-polymer hybrid nanoparticles. Biomed Pharmacother. 2020;127: 110181.

    CAS  PubMed  Google Scholar 

  244. Huang W-Y, Lin J-N, Hsieh J-T, Chou S-C, Lai C-H, Yun E-J, Lo UG, Pong R-C, Lin J-H, Lin Y-H. Nanoparticle targeting CD44-positive cancer cells for site-specific drug delivery in prostate cancer therapy. ACS Appl Mater Interfaces. 2016;8(45):30722–34.

    CAS  PubMed  Google Scholar 

  245. Mirzaei S, Mahabady MK, Zabolian A, Abbaspour A, Fallahzadeh P, Noori M, Hashemi F, Hushmandi K, Daneshi S, Kumar AP, et al. Small interfering RNA (siRNA) to target genes and molecular pathways in glioblastoma therapy: Current status with an emphasis on delivery systems. Life Sci. 2021;275: 119368.

    CAS  PubMed  Google Scholar 

  246. Ashrafizadeh M, Zarrabi A, Hushmandi K, Hashemi F, Rahmani Moghadam E, Raei M, Kalantari M, Tavakol S, Mohammadinejad R, Najafi M, et al. Progress in natural compounds/siRNA co-delivery employing nanovehicles for cancer therapy. ACS Comb Sci. 2020;22(12):669–700.

    CAS  PubMed  PubMed Central  Google Scholar 

  247. Aghamiri S, Raee P, Shahmohamadnejad S, Shabani S, Ghorbani J, Sameni M, Ebrahimi MT. Recent advances in siRNA delivery systems for prostate cancer therapy. Curr Pharm Biotechnol. 2022;23(4):579–93.

    CAS  PubMed  Google Scholar 

  248. Rahme K, Guo J, Holmes JD. Bioconjugated Gold Nanoparticles Enhance siRNA Delivery in Prostate Cancer Cells. In: Dinesh Kumar L, editor. RNA Interference and Cancer Therapy: Methods and Protocols. New York: Springer; 2019. p. 291–301.

    Google Scholar 

  249. Chen J, Yang Y, Xu D, Li J, Wu S, Jiang Y, Wang C, Yang Z, Zhao L. Mesoporous silica nanoparticles combined with AKR1C3 siRNA inhibited the growth of castration-resistant prostate cancer by suppressing androgen synthesis in vitro and in vivo. Biochem Biophys Res Commun. 2021;540:83–9.

    CAS  PubMed  Google Scholar 

  250. Yang J, Xie S-X, Huang Y, Ling M, Liu J, Ran Y, Wang Y, Thrasher JB, Berkland C, Li B. Prostate-targeted biodegradable nanoparticles loaded with androgen receptor silencing constructs eradicate xenograft tumors in mice. Nanomedicine. 2012;7(9):1297–309.

    CAS  PubMed  Google Scholar 

  251. Rahimi S, Roushandeh AM, Ebrahimi A, Samadani AA, Kuwahara Y, Roudkenar MH. CRISPR/Cas9-mediated knockout of Lcn2 effectively enhanced CDDP-induced apoptosis and reduced cell migration capacity of PC3 cells. Life Sci. 2019;231: 116586.

    CAS  PubMed  Google Scholar 

  252. Sorrentino C, D’Antonio L, Ciummo SL, Fieni C, Landuzzi L, Ruzzi F, Vespa S, Lanuti P, Lotti LV, Lollini PL, Di Carlo E. CRISPR/Cas9-mediated deletion of Interleukin-30 suppresses IGF1 and CXCL5 and boosts SOCS3 reducing prostate cancer growth and mortality. J Hematol Oncol. 2022;15(1):145.

    CAS  PubMed  PubMed Central  Google Scholar 

  253. Friedhuber AM, Chandolu V, Manchun S, Donkor O, Sriamornsak P, Dass CR. Nucleotropic doxorubicin nanoparticles decrease cancer cell viability, destroy mitochondria, induce autophagy and enhance tumour necrosis. J Pharm Pharmacol. 2015;67(1):68–77.

    CAS  PubMed  Google Scholar 

  254. Wang G, Zhang D, Yang S, Wang Y, Tang Z, Fu X. Co-Administration of genistein with doxorubicin-loaded polypeptide nanoparticles weakens the metastasis of malignant prostate cancer by amplifying oxidative damage. Biomater Sci. 2018;6(4):827–35.

    CAS  PubMed  Google Scholar 

  255. Liang S, Han L, Mu W, Jiang D, Hou T, Yin X, Pang X, Yang R, Liu Y, Zhang N. Carboplatin-loaded SMNDs to reduce GSH-mediated platinum resistance for prostate cancer therapy. J Mater Chem B. 2018;6(43):7004–14.

    CAS  PubMed  Google Scholar 

  256. Zhang C, Zhao X, Guo H. Synergic highly effective photothermal-chemotherapy with platinum prodrug linked melanin-like nanoparticles. Artif Cells Nanomed Biotechnol. 2018;46(sup2):356–63.

    CAS  PubMed  Google Scholar 

  257. Kumar A, Huo S, Zhang X, Liu J, Tan A, Li S, Jin S, Xue X, Zhao Y, Ji T, et al. Neuropilin-1-targeted gold nanoparticles enhance therapeutic efficacy of platinum(IV) drug for prostate cancer treatment. ACS Nano. 2014;8(5):4205–20.

    CAS  PubMed  Google Scholar 

  258. Halpern JA, Oromendia C, Shoag JE, Mittal S, Cosiano MF, Ballman KV, Vickers AJ, Hu JC. Use of digital rectal examination as an adjunct to prostate specific antigen in the detection of clinically significant prostate cancer. J Urol. 2018;199(4):947–53.

    PubMed  Google Scholar 

  259. Lonergan PE, Tindall DJ. Androgen receptor signaling in prostate cancer development and progression. J Carcinog. 2011;10:20.

    CAS  PubMed  PubMed Central  Google Scholar 

  260. Tannock IF, de Wit R, Berry WR, Horti J, Pluzanska A, Chi KN, Oudard S, Théodore C, James ND, Turesson I, et al. Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. N Engl J Med. 2004;351(15):1502–12.

    CAS  PubMed  Google Scholar 

  261. Ryan CJ, Smith MR, de Bono JS, Molina A, Logothetis CJ, de Souza P, Fizazi K, Mainwaring P, Piulats JM, Ng S, et al. Abiraterone in metastatic prostate cancer without previous chemotherapy. N Engl J Med. 2012;368(2):138–48.

    PubMed  PubMed Central  Google Scholar 

  262. Scher HI, Fizazi K, Saad F, Taplin M-E, Sternberg CN, Miller K, de Wit R, Mulders P, Chi KN, Shore ND, et al. Increased survival with enzalutamide in prostate cancer after chemotherapy. N Engl J Med. 2012;367(13):1187–97.

    CAS  PubMed  Google Scholar 

  263. Beer TM, Armstrong AJ, Rathkopf DE, Loriot Y, Sternberg CN, Higano CS, Iversen P, Bhattacharya S, Carles J, Chowdhury S, et al. Enzalutamide in metastatic prostate cancer before chemotherapy. N Engl J Med. 2014;371(5):424–33.

    PubMed  PubMed Central  Google Scholar 

  264. Smith MR, Saad F, Chowdhury S, Oudard S, Hadaschik BA, Graff JN, Olmos D, Mainwaring PN, Lee JY, Uemura H, et al. Apalutamide treatment and metastasis-free survival in prostate cancer. N Engl J Med. 2018;378(15):1408–18.

    CAS  PubMed  Google Scholar 

  265. Fizazi K, Shore N, Tammela TL, Ulys A, Vjaters E, Polyakov S, Jievaltas M, Luz M, Alekseev B, Kuss I, et al. Darolutamide in nonmetastatic, castration-resistant prostate cancer. N Engl J Med. 2019;380(13):1235–46.

    CAS  PubMed  Google Scholar 

  266. Saad F, Carles J, Gillessen S, Heidenreich A, Heinrich D, Gratt J, Lévy J, Miller K, Nilsson S, Petrenciuc O, et al. Radium-223 and concomitant therapies in patients with metastatic castration-resistant prostate cancer: an international, early access, open-label, single-arm phase 3b trial. Lancet Oncol. 2016;17(9):1306–16.

    CAS  PubMed  Google Scholar 

  267. Vale CL, Burdett S, Rydzewska LHM, Albiges L, Clarke NW, Fisher D, Fizazi K, Gravis G, James ND, Mason MD, et al. Addition of docetaxel or bisphosphonates to standard of care in men with localised or metastatic, hormone-sensitive prostate cancer: a systematic review and meta-analyses of aggregate data. Lancet Oncol. 2016;17(2):243–56.

    CAS  PubMed  PubMed Central  Google Scholar 

  268. Smith MR, Egerdie B, Hernández Toriz N, Feldman R, Tammela TL, Saad F, Heracek J, Szwedowski M, Ke C, Kupic A, et al. Denosumab in men receiving androgen-deprivation therapy for prostate cancer. N Engl J Med. 2009;361(8):745–55.

    CAS  PubMed  PubMed Central  Google Scholar 

  269. Fizazi K, Carducci M, Smith M, Damião R, Brown J, Karsh L, Milecki P, Shore N, Rader M, Wang H, et al. Denosumab versus zoledronic acid for treatment of bone metastases in men with castration-resistant prostate cancer: a randomised, double-blind study. Lancet. 2011;377(9768):813–22.

    CAS  PubMed  PubMed Central  Google Scholar 

  270. de Bono J, Mateo J, Fizazi K, Saad F, Shore N, Sandhu S, Chi KN, Sartor O, Agarwal N, Olmos D, et al. Olaparib for metastatic castration-resistant prostate cancer. N Engl J Med. 2020;382(22):2091–102.

    PubMed  Google Scholar 

  271. Hussain M, Mateo J, Fizazi K, Saad F, Shore N, Sandhu S, Chi KN, Sartor O, Agarwal N, Olmos D, et al. Survival with olaparib in metastatic castration-resistant prostate cancer. N Engl J Med. 2020;383(24):2345–57.

    CAS  PubMed  Google Scholar 

  272. Abida W, Patnaik A, Campbell D, Shapiro J, Bryce AH, McDermott R, Sautois B, Vogelzang NJ, Bambury RM, Voog E, et al. Rucaparib in men with metastatic castration-resistant prostate cancer harboring a BRCA1 or BRCA2 gene alteration. J Clin Oncol. 2020;38(32):3763–72.

    CAS  PubMed  PubMed Central  Google Scholar 

  273. de Bono JS, Mehra N, Scagliotti GV, Castro E, Dorff T, Stirling A, Stenzl A, Fleming MT, Higano CS, Saad F, et al. Talazoparib monotherapy in metastatic castration-resistant prostate cancer with DNA repair alterations (TALAPRO-1): an open-label, phase 2 trial. Lancet Oncol. 2021;22(9):1250–64.

    PubMed  Google Scholar 

  274. Teyssonneau D, Margot H, Cabart M, Anonnay M, Sargos P, Vuong N-S, Soubeyran I, Sevenet N, Roubaud G. Prostate cancer and PARP inhibitors: progress and challenges. J Hematol Oncol. 2021;14(1):51.

    CAS  PubMed  PubMed Central  Google Scholar 

  275. Beer TM, Kwon ED, Drake CG, Fizazi K, Logothetis C, Gravis G, Ganju V, Polikoff J, Saad F, Humanski P, et al. Randomized, double-blind, phase III trial of ipilimumab versus placebo in asymptomatic or minimally symptomatic patients with metastatic chemotherapy-naive castration-resistant prostate cancer. J Clin Oncol. 2017;35(1):40–7.

    CAS  PubMed  Google Scholar 

  276. Shenderov E, Boudadi K, Fu W, Wang H, Sullivan R, Jordan A, Dowling D, Harb R, Schonhoft J, Jendrisak A, et al. Nivolumab plus ipilimumab, with or without enzalutamide, in AR-V7-expressing metastatic castration-resistant prostate cancer: A phase-2 nonrandomized clinical trial. Prostate. 2021;81(6):326–38.

    CAS  PubMed  PubMed Central  Google Scholar 

  277. Karzai F, VanderWeele D, Madan RA, Owens H, Cordes LM, Hankin A, Couvillon A, Nichols E, Bilusic M, Beshiri ML, et al. Activity of durvalumab plus olaparib in metastatic castration-resistant prostate cancer in men with and without DNA damage repair mutations. J Immunother Cancer. 2018;6(1):141.

    PubMed  PubMed Central  Google Scholar 

  278. Kwan EM, Spain L, Anton A, Gan CL, Garrett L, Chang D, Liow E, Bennett C, Zheng T, Yu J, et al. Avelumab combined with stereotactic ablative body radiotherapy in metastatic castration-resistant prostate cancer: the phase 2 ICE-PAC Clinical Trial. Eur Urol. 2022;81(3):253–62.

    CAS  PubMed  Google Scholar 

  279. Perner S, Hofer MD, Kim R, Shah RB, Li H, Möller P, Hautmann RE, Gschwend JE, Kuefer R, Rubin MA. Prostate-specific membrane antigen expression as a predictor of prostate cancer progression. Hum Pathol. 2007;38(5):696–701.

    CAS  PubMed  Google Scholar 

  280. Schmidkonz C, Götz TI, Atzinger A, Ritt P, Prante O, Kuwert T, Bäuerle T, Goebell P, Cordes M. 99mTc-MIP-1404 SPECT/CT for assessment of whole-body tumor burden and treatment response in patients with biochemical recurrence of prostate cancer. Clin Nucl Med. 2020;45(8):e349–57.

    PubMed  Google Scholar 

  281. Hofman MS, Violet J, Hicks RJ, Ferdinandus J, Thang SP, Akhurst T, Iravani A, Kong G, Ravi Kumar A, Murphy DG, et al. [177 Lu]-PSMA-617 radionuclide treatment in patients with metastatic castration-resistant prostate cancer (LuPSMA trial): a single-centre, single-arm, phase 2 study. Lancet Oncol. 2018;19(6):825–33.

    CAS  PubMed  Google Scholar 

  282. Rahbar K, Ahmadzadehfar H, Kratochwil C, Haberkorn U, Schäfers M, Essler M, Baum RP, Kulkarni HR, Schmidt M, Drzezga A, et al. German multicenter study investigating 177Lu-PSMA-617 radioligand therapy in advanced prostate cancer patients. J Nucl Med. 2017;58(1):85–90.

    CAS  PubMed  Google Scholar 

  283. Baum RP, Kulkarni HR, Schuchardt C, Singh A, Wirtz M, Wiessalla S, Schottelius M, Mueller D, Klette I, Wester HJ. 177Lu-labeled prostate-specific membrane antigen radioligand therapy of metastatic castration-resistant prostate cancer: safety and efficacy. J Nucl Med. 2016;57(7):1006–13.

    CAS  PubMed  Google Scholar 

  284. Hofman MS, Lawrentschuk N, Francis RJ, Tang C, Vela I, Thomas P, Rutherford N, Martin JM, Frydenberg M, Shakher R, et al. Prostate-specific membrane antigen PET-CT in patients with high-risk prostate cancer before curative-intent surgery or radiotherapy (proPSMA): a prospective, randomised, multicentre study. Lancet. 2020;395(10231):1208–16.

    CAS  PubMed  Google Scholar 

  285. Autio KA, Dreicer R, Anderson J, Garcia JA, Alva A, Hart LL, Milowsky MI, Posadas EM, Ryan CJ, Graf RP, et al. Safety and efficacy of BIND-014, a docetaxel nanoparticle targeting prostate-specific membrane antigen for patients with metastatic castration-resistant prostate cancer: a phase 2 clinical trial. JAMA Oncol. 2018;4(10):1344–51.

    PubMed  PubMed Central  Google Scholar 

  286. Thang SP, Lam WWC, Tong AKT, Allen JC Jr, Ler ASL, Tay YS, Somanesan S, Kanesvaran R, Wong ASC, Ng DCE. Clinical outcomes of 177lutetium-prostate-specific membrane antigen therapy in advanced prostate cancer-a prospective pilot study in an Asian population. Nucl Med Commun. 2020;41(7):618–28.

    CAS  PubMed  Google Scholar 

  287. Kloss CC, Lee J, Zhang A, Chen F, Melenhorst JJ, Lacey SF, Maus MV, Fraietta JA, Zhao Y, June CH. Dominant-negative TGF-β receptor enhances PSMA-targeted human CAR T cell proliferation and augments prostate cancer eradication. Mol Ther. 2018;26(7):1855–66.

    CAS  PubMed  PubMed Central  Google Scholar 

  288. Calais J, Ceci F, Eiber M, Hope TA, Hofman MS, Rischpler C, Bach-Gansmo T, Nanni C, Savir-Baruch B, Elashoff D, et al. (18)F-fluciclovine PET-CT and (68)Ga-PSMA-11 PET-CT in patients with early biochemical recurrence after prostatectomy: a prospective, single-centre, single-arm, comparative imaging trial. Lancet Oncol. 2019;20(9):1286–94.

    CAS  PubMed  PubMed Central  Google Scholar 

  289. Violet J, Sandhu S, Iravani A, Ferdinandus J, Thang SP, Kong G, Kumar AR, Akhurst T, Pattison DA, Beaulieu A, et al. Long-term follow-up and outcomes of retreatment in an expanded 50-patient single-center phase ii prospective trial of (177)Lu-PSMA-617 theranostics in metastatic castration-resistant prostate cancer. J Nucl Med. 2020;61(6):857–65.

    PubMed  PubMed Central  Google Scholar 

  290. Tagawa ST, Vallabhajosula S, Christos PJ, Jhanwar YS, Batra JS, Lam L, Osborne J, Beltran H, Molina AM, Goldsmith SJ, et al. Phase 1/2 study of fractionated dose lutetium-177-labeled anti-prostate-specific membrane antigen monoclonal antibody J591 ((177) Lu-J591) for metastatic castration-resistant prostate cancer. Cancer. 2019;125(15):2561–9.

    CAS  PubMed  Google Scholar 

  291. Armstrong AJ, Shen T, Halabi S, Kemeny G, Bitting RL, Kartcheske P, Embree E, Morris K, Winters C, Jaffe T, et al. A phase II trial of temsirolimus in men with castration-resistant metastatic prostate cancer. Clin Genitourin Cancer. 2013;11(4):397–406.

    PubMed  Google Scholar 

  292. Graham L, Banda K, Torres A, Carver BS, Chen Y, Pisano K, Shelkey G, Curley T, Scher HI, Lotan TL, et al. A phase II study of the dual mTOR inhibitor MLN0128 in patients with metastatic castration resistant prostate cancer. Invest New Drugs. 2018;36(3):458–67.

    CAS  PubMed  PubMed Central  Google Scholar 

  293. Chow H, Ghosh PM, deVere WR, Evans CP, Dall’Era MA, Yap SA, Li Y, Beckett LA, Lara PN Jr, Pan CX. A phase 2 clinical trial of everolimus plus bicalutamide for castration-resistant prostate cancer. Cancer. 2016;122(12):1897–904.

    CAS  PubMed  Google Scholar 

  294. de Bono JS, De Giorgi U, Rodrigues DN, Massard C, Bracarda S, Font A, Arranz Arija JA, Shih KC, Radavoi GD, Xu N, et al. Randomized phase II study evaluating Akt blockade with ipatasertib, in combination with abiraterone, in patients with metastatic prostate cancer with and without PTEN Loss. Clin Cancer Res. 2019;25(3):928–36.

    PubMed  Google Scholar 

  295. Massard C, Chi KN, Castellano D, de Bono J, Gravis G, Dirix L, Machiels JP, Mita A, Mellado B, Turri S, et al. Phase Ib dose-finding study of abiraterone acetate plus buparlisib (BKM120) or dactolisib (BEZ235) in patients with castration-resistant prostate cancer. Eur J Cancer. 2017;76:36–44.

    CAS  PubMed  Google Scholar 

  296. Wei XX, Hsieh AC, Kim W, Friedlander T, Lin AM, Louttit M, Ryan CJ. A Phase I study of abiraterone acetate combined with BEZ235, a Dual PI3K/mTOR inhibitor. Metastatic Castration Resistant Prostate Cancer Oncologist. 2017;22(5):503-e543.

    CAS  PubMed  Google Scholar 

  297. Narayan V, Vapiwala N, Mick R, Subramanian P, Christodouleas JP, Bekelman JE, Deville C, Rajendran R, Haas NB. Phase 1 trial of everolimus and radiation therapy for salvage treatment of biochemical recurrence in prostate cancer patients following prostatectomy. Int J Radiat Oncol, Biol, Phys. 2017;97(2):355–61.

    CAS  PubMed  Google Scholar 

  298. Kase AM, Copland Iii JA, Tan W. Novel therapeutic strategies for CDK4/6 inhibitors in metastatic castrate-resistant prostate cancer. Onco Targets Ther. 2020;13:10499–513.

    CAS  PubMed  PubMed Central  Google Scholar 

  299. Kumari S, Sharma V, Tiwari R, Maurya JP, Subudhi BB, Senapati D. Therapeutic potential of p53 reactivation in prostate cancer: Strategies and opportunities. Eur J Pharmacol. 2022;919: 174807.

    CAS  PubMed  Google Scholar 

  300. Murillo-Garzón V, Kypta R. WNT signalling in prostate cancer. Nat Rev Urol. 2017;14(11):683–96.

    PubMed  Google Scholar 

  301. Melegh Z, Oltean S. Targeting angiogenesis in prostate cancer. Int J Mol Sci. 2019;20(11):34.

    Google Scholar 

  302. Fizazi K, Higano CS, Nelson JB, Gleave M, Miller K, Morris T, Nathan FE, McIntosh S, Pemberton K, Moul JW. Phase III, randomized, placebo-controlled study of docetaxel in combination with zibotentan in patients with metastatic castration-resistant prostate cancer. J Clin Oncol. 2013;31(14):1740–7.

    CAS  PubMed  Google Scholar 

  303. Corn PG, Wang F, McKeehan WL, Navone N. Targeting fibroblast growth factor pathways in prostate cancer. Clin Cancer Res. 2013;19(21):5856–66.

    CAS  PubMed  PubMed Central  Google Scholar 

  304. Lin S-R, Yeh H-L, Liu Y-N. Interplay of epidermal growth factor receptor and signal transducer and activator of transcription 3 in prostate cancer: beyond androgen receptor transactivation. Cancers. 2021;13(14):3452.

    CAS  PubMed  PubMed Central  Google Scholar 

  305. Chau V, Madan RA, Aragon-Ching JB. Protein kinase inhibitors for the treatment of prostate cancer. Expert Opin Pharmacother. 2021;22(14):1889–99.

    CAS  PubMed  Google Scholar 

  306. Odero-Marah V, Hawsawi O, Henderson V, Sweeney J. Epithelial-Mesenchymal Transition (EMT) and prostate cancer. Adv Exp Med Biol. 2018;1095:101–10.

    CAS  PubMed  Google Scholar 

  307. Varkaris A, Katsiampoura AD, Araujo JC, Gallick GE, Corn PG. Src signaling pathways in prostate cancer. Cancer Metastasis Rev. 2014;33(2–3):595–606.

    CAS  PubMed  PubMed Central  Google Scholar 

  308. Ahel J, Hudorovic N, Vicic-Hudorovic V, Nikles H. Tgf-Beta in the natural history of prostate cancer. Acta Clin Croat. 2019;58(1):128–38.

    PubMed  PubMed Central  Google Scholar 

  309. Nickols NG, Nazarian R, Zhao SG, Tan V, Uzunangelov V, Xia Z, Baertsch R, Neeman E, Gao AC, Thomas GV, et al. MEK-ERK signaling is a therapeutic target in metastatic castration resistant prostate cancer. Prostate Cancer Prostatic Dis. 2019;22(4):531–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  310. Mottet N, van den Bergh RCN, Briers E, Van den Broeck T, Cumberbatch MG, De Santis M, Fanti S, Fossati N, Gandaglia G, Gillessen S, et al. EAU-EANM-ESTRO-ESUR-SIOG Guidelines on Prostate Cancer-2020 Update. Part 1: screening, diagnosis, and local treatment with curative intent. Eur Urol. 2021;79(2):243–62.

    CAS  PubMed  Google Scholar 

  311. Parker C, Castro E, Fizazi K, Heidenreich A, Ost P, Procopio G, Tombal B, Gillessen S. Prostate cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2020;31(9):1119–34.

    CAS  PubMed  Google Scholar 

  312. Mottet N, Bellmunt J, Bolla M, Briers E, Cumberbatch MG, De Santis M, Fossati N, Gross T, Henry AM, Joniau S, et al. EAU-ESTRO-SIOG Guidelines on Prostate Cancer. Part 1: screening, diagnosis, and local treatment with curative intent. Eur Urol. 2017;71(4):618–29.

    PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by Basic Science Research Program through the National Research Foundation of Korea (NRF) Grant funded by the Korea government (MSIT) (2021R1A5A6002853 and 2022M3J7A1062940). This work was also supported by a Grant from the Korea Environmental Industry and Technology Institute (KEITI), funded by the Ministry of Environment (MOE) of the Republic of Korea (RE202201777).

Author information

Authors and Affiliations

Authors

Contributions

All authors conceptualized the review topic. GLVP, VKG, KP, and BLVKS conducted the literature search and wrote the original draft. EK and MBT visualized the description with figures. PM, and HKV read and suggested the modifications. GSRR, BLVKS and YSH reviewed and edited the manuscript. All authors have read and agreed to the submitted version of the manuscript.

Corresponding authors

Correspondence to Ganji Seeta Rama Raju, LVKS Bhaskar or Yun Suk Huh.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests:

The authors declare that they have no potential conflicts of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Varaprasad, G.L., Gupta, V.K., Prasad, K. et al. Recent advances and future perspectives in the therapeutics of prostate cancer. Exp Hematol Oncol 12, 80 (2023). https://doi.org/10.1186/s40164-023-00444-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40164-023-00444-9

Keywords