This site is intended for healthcare professionals
Prostate Cancer
DNA Repair in Prostate Cancer

Prostate Cancer

Last updated:10th Nov 2020
Published:10th Aug 2020

Epidemiology

Globally, prostate cancer is estimated to be the second most common cancer in males and the fifth most common cause of male deaths1

Prevalence, incidence and mortality

Prostate cancer is estimated to be the second most common cancer in males globally, estimated at 1,276,106 new cases in 2018, and the fifth most common cause of male cancer-related deaths, estimated at 358,989 deaths1. There are considerable differences in incidence rates (189.1/100,000 in Guadeloupe and 1.0/100,000 in Bhutan at the extremes); however, incidence rates do not correlate with mortality rates, which varies less1,2. Both genetic and environmental factors impact on prostate cancer incidence, including rate of diagnosis, differences in screening and access to healthcare2.

The estimates for Europe for the same period show that prostate cancer is the most common male cancer, forming an estimated 21.8% of all new male cancers diagnosed in 2018 (Figure 1)3. This places prostate cancer as the third most common cause of cancer mortality in men in Europe at 10% of male cancer deaths, following lung (24.8%) and colorectum cancers (12.0%) (Figure 1)3.

Figure 1. Estimated incidence and mortality rates for the top 5 cancers in males across European countries in 2018. Adapted from Ferlay et al., 2018

Figure 1. Estimated incidence and mortality rates for the top 5 cancers in males across European countries in 2018. Adapted from Ferlay et al., 20183.

The 2018 age-standardised incidence rates of prostate cancer vary by more than 5-fold (37–189.3 per 100,000) across European countries (Figure 2)3.

Figure 2. Estimated 2018 age-standardised incidence and mortality rates (per 100,000) for prostate cancer by area and country in Europe. Adapted from Ferlay et al., 2018

Figure 2. Estimated 2018 age-standardised incidence and mortality rates (per 100,000) for prostate cancer by area and country in Europe. Adapted from Ferlay et al., 20183.

The highest incidence rates are in Northern European countries and the lowest in Central and Eastern European countries. The fold difference is an improvement on the greatest (7.7-fold) difference recorded in 2012 and likely reflects improvements in prostate cancer screening programs in Central and Eastern European countries4. In comparison with incidence, mortality rates vary less across Europe (3.5-fold difference)3. These figures likely reflect variability in national health system policies (for example, organised screening for prostate cancer), differences in risk factors between countries and regions, and disparities in the delivery of cancer management measures. The variation in incidence and mortality is much lower for the EU-5 countries (United Kingdom, France, Germany, Spain and Italy) (Figure 3)3.

Figure 3. Estimated number of new cases and deaths for prostate cancer in 2018 for the EU5 countries. Adapted from Ferlay et al, 2018

Figure 3. Estimated number of new cases and deaths for prostate cancer in 2018 for the EU5 countries. Adapted from Ferlay et al, 20183.

Prostate cancer survival

Overall prostate cancer survival has been steadily increasing over the past 50 years (Figure 4)5. This increase in survival is partly explained by early detection and treatment, however other factors are likely to be involved6.Figure 4. Age-standardised one, five– and ten–year net survival in England and Wales between 1971 and 2011. Adapted from CRUK Prostate Cancer Statistics

Figure 4. Age-standardised one, five and tenyear net survival in England and Wales between 1971 and 2011. Adapted from CRUK Prostate Cancer Statistics5.

Clinical outcome is dependent on many factors, including clinical stage at diagnosis. If detected at an early stage (Stages I–II), prostate cancer patients are likely to live beyond 5 years from diagnosis (Figure 5)5. Stage III prostate cancer patients have an approximately 93% chance of surviving beyond 5 years, whilst Stage IV patients who have incurable metastatic disease have only a 30% five-year relative survival based on data from 2002–20065.

Figure 5. Five-year relative survival (%) by prostate cancer stage in adults aged 15–99, 2002–2006 data from the National Cancer Registration Service, East Office. Adapted from CRUK Prostate Cancer Statistics

Figure 5. Five–year relative survival (%) by prostate cancer stage in adults aged 15–99, 2002–2006 data from the National Cancer Registration Service, East Office. Adapted from CRUK Prostate Cancer Statistics5.

Risk factors

The risk of prostate cancer is higher in older men, men of African-American descent and men with a family history of cancer6

Unmodifiable risks of prostate cancer include increasing age, race and a family history of prostate and other cancers, whereas modifiable risks factors include obesity and smoking6.

Age

Prostate cancer risk increases with age, with incidence rates peaking in men who are in their seventies (Figure 6)5.  

Figure 6. Average number of new cases per year and age-specific incidence rates per 100,000 males, UK. Adapted from CRUK Prostate Cancer Statistics

Figure 6. Average number of new cases per year and age-specific incidence rates per 100,000 males, UK. Adapted from CRUK Prostate Cancer Statistics5.

Race

Differences in prostate cancer incidence and mortality exist between racial and ethnic groups. In the USA, African-American men are nearly twice as likely to be diagnosed with prostate cancer compared to Caucasian men, and three times as likely compared to Asian men6. African-American men are also more likely to die from the disease than Caucasian and Asian men6,7. The reason for the difference in incidence and mortality rates between racial and ethnic groups is not clear; however, this may be impacted by differences in access to care, stage at diagnosis and genetic predisposition2,8,9.

Family risk

Genome-wide association studies have identified more than 180 genetic risk loci, indicating that predisposition genes may also impact on prostate cancer risk6. Indeed, studies in twins found a high heritability rate of up to 57%10,11. Family association studies also found that men who have a brother or a father diagnosed with prostate cancer have a two to three-fold increased risk of developing and dying of the disease themselves12,13. If both a brother and a father have been diagnosed, the risk is further increased14. Prostate cancer patients who have a first-degree family history of any cancer are also more likely to develop and die from secondary primary cancers such as lung and colorectal cancers15.

Men who have a family history of hereditary breast and ovarian cancer (HBOC) syndrome that is caused by germline mutations in homologous recombination repair (HRR) genes such as BRCA1 and BRCA2 are at an increased risk of prostate cancer16–18. Initial findings of the IMPACT study that followed up male subjects who had germline mutations in BRCA1 or BRCA2 found that BRCA2 carriers had a higher prostate cancer incidence rate per 1000 patient years compared to noncarriers (19.4 vs 12.0; P=0.03)19. This study also found that BRCA2 carriers were diagnosed at a younger age and were more likely to have clinically significant disease19. The role for BRCA1 remains unclear, and more data is needed to understand it’s role in prostate cancer risk19.

To find out more about the cellular role for BRCA2 in DNA damage repair, its prevalence in prostate cancer, and its impact on clinical practice, please visit the DNA repair section.

Find out more on the techniques for molecular testing of BRCA2 in prostate cancer here.

Modifiable risks of prostate cancer

Obesity has been associated with increased mortality and recurrence of prostate cancer in multiple studies, even when normalising to the prostate cancer stage and PSA readings at diagnosis6. Smoking is also suggested to increase the risk of death from prostate cancer, advanced stage disease and poorly differentiated cancer6. Smokers were found to have an approximately 60% increased risk of prostate cancer mortality following adjustment of potential confounding factors compared to men who never smoked6.

Detection and Classification

Based on data from 2002–2006, stage IV prostate cancer has a 5–year survival rate of approximately 30% compared to early stage disease which has a 5–year survival rate of 95–100%5. Prostate cancer staging is therefore key to ensuring the best chance of survival

Major international guidelines do not currently recommend routine screening for prostate cancer20. The diagnosis of prostate cancer therefore typically follows a patient requesting examination or following investigation for clinical symptoms. Local symptoms for prostate cancer start arising once the cancer is large enough to affect the urethra21. Subsequently, a lot of the symptoms involve problems with urinating such as frequent urination, difficulty in urinating and weak flow22. Patients may also experience blood in semen, erectile dysfunction and discomfort in the pelvic area22. Metastatic prostate cancer further present symptoms according to the organ affected and symptoms can include bone pain, a loss of appetite, and unexplained weight loss21.

Detection

A patient is assessed to determine if they have prostate cancer using detection techniques that typically involve23:

  • Digital rectal exam (DRE)
  • Prostate-specific antigen (PSA) levels in blood
  • Imaging tests
  • Biopsy

Digital rectal exam (DRE)

Digital rectal exam (DRE) is a partial physical exam used to screen for prostate cancer, and measure prostate size for the purposes of grading. Since the prostate can be felt through the rectal wall, DRE involves the insertion of a finger into the rectum to feel the prostate. Prostate cancers are mostly located in the peripheral zone of the prostate and can be detected by DRE when the volume is ≥0.2 mL23.

Prostate-specific antigen (PSA)

The most common type of prostate cancer is acinar adenocarcinoma which arises from glandular epithelial cells that line the prostate24. These cells secrete prostate-specific antigen (PSA), high blood levels of which may indicate the presence of a prostate cancer25. Some measurements for PSA include23:

  • PSA level is measurable in blood samples as ng/mL blood
  • PSA density is the level of serum PSA divided by the TRUS-determined prostate volume
  • PSA velocity describes the change in PSA levels over time
  • PSA doubling time is the time it takes the PSA level to double

Major international guidelines do not recommend routine PSA screening due to the high false positive rate20. Additionally, poorly differentiated prostate cancers and rarer types of prostate cancer that do not secrete PSA may be missed24. These include ductal adenocarcinomas and some neuroendocrine and basal cell prostate cancers that most often present normal levels of PSA24.

To overcome the challenges of PSA sensitivity and cancer specificity, other measurements and biomarkers of prostate cancer are being explored to be used in conjunction with PSA testing.

Imaging

Imaging is recommended to some patients to stage the disease and guide treatment planning26. Imaging is also used to inform on metastatic relapse, to monitor response to therapy, assess for disease progression and to investigate new symptoms potentially associated with the disease27.

Imaging techniques used to assess prostate cancer include transrectal ultrasound (TRUS) imaging, computed tomography (CT), magnetic resonance imaging (MRI), radionucleotide bone scan, and positron emission tomography (PET) in combination with CT or MRI (Figure 7)26

Figure 7. Summary of imaging techniques used for prostate cancer detection, staging and assessing treatment response

Figure 7. Summary of imaging techniques used for prostate cancer detection, staging and assessing treatment response27–34.

Ultrasound imaging: Ultrasound imaging is used to guide the retrieval of primary prostate cancer samples, known as transrectal ultrasound guided (TRUS) biopsy35. TRUS is carried out if a patient displays a high PSA level and abnormal DRE findings. Typically, ten to twelve cores are taken which are used to measure Gleason scores35. Since biopsy retrieval is based on estimation, high-grade cancers that require urgent treatment are often missed28. New techniques to distinguish between low and high-grade tumours are currently being explored.

Magnetic resonance imaging (MRI): An MRI scan uses magnetism and radio waves to create images from multiple angles to show soft tissue clearly36. Multi-parametric MRI (mpMRI) is a more recent technique which provides more information than standard MRI by combining multiple images26,30. Alongside tissue anatomy, it provides information on prostate volume, cellularity and vascularity30. Whilst not yet routine, recent studies show that mpMRI may help in stratifying patients for TRUS biopsy and reduce overdiagnosis of clinically insignificant disease, whilst improving the detection of clinically significant cancer30.

Computed tomography (CT): A CT scan uses X-rays to create cross-sectional images of the body. CT scans are used primarily for staging at base-line to assess for any suspected locally advanced or metastatic disease29. CT scans are also used for planning radiation delivery and assessing treatment response in advanced disease34.

Positron emission tomography (PET): PET scans are typically used to assess metastatic disease for staging and more recently, for treatment planning in patients who require salvage therapy31,37. PET is often carried out together with a CT scan to provide more detailed images (PET/CT scan)27,32. PET can identify specific cells using labelled radiotracers that are attached to a biologically active molecule32. Common radiotracers include 11C and 18F choline and more recently, 68Ga- or 18F-labelled prostate specific membrane antigen (PMSA) that has higher sensitivity, specificity and target-to-background ratio32,38. PSMA-PET/CT has a high positive detection rate since PSMA is over-expressed in 95% of primary prostate cancers; however, its expression is not limited to prostate tissue and can be overexpressed in other tumours31,38. PSMA PET/CT is highly sensitive and can detect prostate cancers, even when PSA levels are less than 1ng/mL31,37. This sensitivity has proven to be useful in planning salvage radiotherapy treatments for cancers with biochemical recurrence31,37.

Bone scan: More than 80% of patients with metastatic castration resistant prostate cancer (mCRPC) present with bone metastasis that is associated with greater morbidity27. A standard bone scan involves a planar whole body imager33. A radiotracer is used to mark regions of bone deposition, indicative of bone metastasis33.

Biopsy

A biopsy confirms diagnosis, gives more detail on the type of cancer and contributes to risk scoring23. Prostate cancer clinical staging informs on the extent of disease progression and is used alongside risk assessment to determine the best management option for the patient23. Using imaging results and biopsy samples, staging involves23,39:

  • Gleason scoring of the primary tumour
  • Tumour, node, metastasis (TNM) scoring

Prostate cancer staging

Prostate cancer can be broadly described as localised, locally advanced or advanced prostate cancer.

  • Localised prostate cancer is sometimes referred to as ‘early’ or ‘organ-confined’ since it describes prostate cancer that hasn’t spread from inside the prostate to other parts of the body40. It is often slow-growing and poses a low risk of spreading
  • Locally advanced prostate cancer, describes prostate cancer that has breached the prostate capsule and potentially invaded local structures including seminal vesicles, pelvic lymph nodes, bladder and rectum41
  • Advanced prostate cancer, or metastatic prostate cancer has spread to other parts of the body via the blood or lymphatic system42

The International Society of Urologic Pathology (2005) (ISUP) Gleason grade that is based on histological analysis of biopsy samples is used together with tumour, node, metastasis (TNM) scores to stage prostate cancer23.   

ISUP Gleason grade

The International Society of Urologic Pathology (2005) Gleason grade is based on patterns from core biopsy and operative specimens of the primary tumour23. Gleason patterns of the prostate gland are graded 1–5 (Figure 8). The Gleason score, which ranges from 2–10, is the sum of the most common pattern and the second most common pattern, or is doubled if only one pattern is present23. If there are more than 2 patterns, the score is the sum of the most common pattern, and that for the highest grade, irrespective of its extent (Table 1)23.

Figure 8. Gleason patterns and associated grades for glandular structures of the prostate. Adapted from NCI SEER training module

Figure 8. Gleason patterns and associated grades for glandular structures of the prostate. Adapted from NCI SEER training module43.

Table 1. Gleason score descriptions. Adapted from Cancer Research UK44.

Table 1. Gleason score descriptions. Adapted from Cancer Research UK

The Gleason scores are further sub-divided into International Society of Urological Pathology (ISUP) Gleason grades (also referred to as Gleason groups) (Table 2), that are used together with tumour node metastasis (TNM) scores and PSA levels for tumour staging23.

Table 2. International Society of Urological Pathology (ISUP) 2014 Gleason grade. Adapted from the EAU-ESTRO-ESUR-SIOG Guidelines on Prostate Cancer, 202023.

Table 2. International Society of Urological Pathology (ISUP) 2014 Gleason grade. Adapted from the EAU-ESTRO-ESUR-SIOG Guidelines on Prostate Cancer, 2020

TNM scores

The tumour, node and metastasis (TNM) score describes tumour locality (Table 3)23. The locations are primary tumour site (T), lymph nodes (N) and distant sites (M). Prostate cancer spreads most frequently to the internal and external iliac and obturator lymph nodes38. Distant metastatic sites are most frequently lymph nodes, bone, then the lungs and liver38.

Table 3. Tumour Node Metastasis (TNM) classification (2017). Adapted from the EAU-ESTRO-ESUR-SIOG Guidelines on Prostate Cancer, 202023.

Table 3. Tumour Node Metastasis (TNM) classification (2017). Adapted from the EAU-ESTRO-ESUR-SIOG Guidelines on Prostate Cancer, 2020

Stage Groups

The TNM classification is used alongside PSA levels and ISUP Grade/Gleason group to determine the prostate cancer stage group (Table 4)23.

Stage I describes a tumour that is slow growing, cannot be felt and occupies one side of the prostate or less45. Stage II describes a tumour that is small and found only in the prostate but has an increasing risk of growing and spreading45. Stage III describes a tumour that is locally advanced, growing and likely to spread whereas Stage IV describes a tumour that has spread beyond the prostate45.

There is increasing evidence that patients with oligometastatic disease (presence of 3–5 metastatic sites) respond better to some metastatic-directed therapies46. Staging of low versus high volume disease using sensitive imaging techniques may help to identify these patients who may be managed differently in the future46.

Table 4. Parameters for the stages of prostate cancer. Adapted from the American Cancer Society47.

Table 4. Parameters for the stages of prostate cancer. Adapted from the American Cancer Society

Treatment Planning

Treatment planning is informed by prostate cancer stage and risk group, as well as the patients health status and life expectancy23. Risk assessments can involve23,26:

  • Risk stratification that involves assigning the patient to a risk group
  • Nomograms
  • Molecular testing

Risk stratification

Risk stratification is used to describe the likelihood of a patient’s prostate cancer progressing to an advanced stage23. An understanding of the risk of developing advanced disease helps to guide treatment decisions at an earlier stage. The European Association of Urology prostate cancer risk stratification group guide considers the patients TNM score, Gleason score/ISUP Gleason grade and PSA levels (Table 5)23.

Table 5. Risk group stratification criteria. Adapted from the EAU-ESTRO-ESUR-SIOG Guidelines on Prostate Cancer, 2020; Parker et al., 202023,48.

Table 5. Risk group stratification criteria. Adapted from the EAU-ESTRO-ESUR-SIOG Guidelines on Prostate Cancer, 2018; Parker et al., 2015

Nomograms

Nomograms are powerful tools that use data from large studies to provide either predictive or prognostic information based on relevant patient-specific variables26. The Partin tables, the first of its kind, uses cancer stage, Gleason score and PSA levels to establish the probability that a patient with localised disease will progress to each pathologic stage26. There are nomograms that predict outcome for various treatment options, including for active surveillance, radical prostatectomy or external beam radiotherapy, and those that assess prognostic probabilities such as biochemical progression-free survival (PFS)26.

Testing for familial risk

Pathogenic alteration of homologous recombination repair (HRR) and mismatch repair (MMR) genes have been associated to an increased risk of prostate cancer, and can lead to hereditary breast and ovarian cancer syndrome (HBOC) and Lynch syndrome, respectively16–18,49–51. These are cancer predisposing syndromes that impact on prostate cancer risk, risk of prostate cancer patients developing secondary cancers, as well as overall family risk of cancer16–18,49–51.

The V2.2020 NCCN Guidelines® for Prostate Cancer recommends germline testing for DNA repair gene alterations in patients with high-risk, very-high-risk, regional or metastatic prostate cancer (Table 6)26. The V2.2020 NCCN Guidelines for Prostate Cancer also recommends germline testing for low and intermediate risk patients with a positive family history, Ashkenazi Jewish ancestry or intraductal/cribriform histology26. The DNA repair genes assessed should include at a minimum MLH1, MSH2, MSH6, and PMS2 (mismatch repair genes) and BRCA2, BRCA1, ATM, PALB2, and CHEK2 (homologous recombination repair genes)26.

Table 6: The V2.2020 NCCN Guidelines for Prostate Cancer recommendations on germline testing, and molecular and biomarker tumour testing in prostate cancer. Adapted with permissions from V2.2020 NCCN Guidelines for Prostate Cancer26.

Table 6: The V2.2020 NCCN Guidelines for Prostate Cancer recommendations on germline testing, and molecular and biomarker tumour testing in prostate cancer. Adapted with permissions from V2.2020 NCCN Guidelines for Prostate Cancer

Dr Alexander Wyatt, who leads a research group on prostate cancer genomics and diagnostics at the Vancouver Prostate Centre discusses how molecular diagnostics are now being applied in prostate cancer.

The ESMO Clinical Practice Guidelines for Prostate Cancer (2020) also recommends germline testing for BRCA2 and other DNA damage response (DDR) genes associated with predisposition syndromes in prostate cancer patients who have a family history of cancer48. It also suggests to consider germline testing for these genes in patients with metastatic prostate cancer48. The EAU guidelines (2020) do not currently provide any recommendations on molecular testing for DNA repair gene alterations in prostate cancer; however, the EAU guidelines recommend offering early PSA testing to men who have an elevated risk of prostate cancer, including men >40 years old who carry BRCA2 mutations23.

Testing for prognostic biomarkers

There is a need for prognostic biomarkers to support treatment planning and to mitigate the risk of unnecessary and costly radical treatment for early disease52. Recent developments have led to the production of diagnostic testing kits that assess collective molecular signatures associated with progression to an advanced disease state52. Whilst not routinely used in the clinic, diagnostic testing kits are a major point of interest with emerging evidence that testing increases active surveillance and reduces the number of men referred for immediate treatment52. The V2.2020 NCCN Guidelines for Prostate Cancer suggest to consider tumour multigene molecular testing in patients with low, intermediate and high-risk prostate cancer, who have a life expectancy of 10 years or more (Table 6)26. Indeed, an understanding of personal risk of aggressive disease may influence the patient choosing treatment instead of active surveillance.

Testing for predictive biomarkers

There is growing evidence that alterations to DNA repair genes may serve as predictive biomarkers of response to investigational treatments53. The V2.2020 NCCN Guidelines for Prostate Cancer therefore recommend tumour testing for HHR gene mutations in patients with metastatic prostate cancer, and suggest to consider tumour testing for MSI or defective MMR in these patients26. Tumour testing for HRR gene mutations, and MSI or defective MMR should also be considered in patients with regional disease26. The ESMO Clinical Practice Guidelines for Prostate Cancer (2020) suggest to consider tumour testing for homologous recombination repair genes and mismatch repair defects in patients with metastatic castration-resistant prostate cancer (mCRPC)48.

Visit the section on molecular diagnostics to learn more about germline and tumour testing for DNA repair gene alterations in prostate cancer.

The DNA repair section delves deep into the prevalence and the role for DNA repair genes in prostate cancer.

Management options

Multiple treatment options are available for both localised and metastatic prostate cancer disease states. Prostate cancer management involves a multidisciplinary approach to ensure the best treatment and outcome for the patient.

Multidisciplinary approach

The number and variety of treatment options for men with prostate cancer can be overwhelming for both the patient and his family. Decision making can be further complicated by the potential for poor outcomes and treatment regret54.

Good communication between patients and the various members of the clinical team can help empower patients to commit to treatment plans55

The European Association of Urology (2020) guidelines state that management decisions for prostate cancer should be made after all treatments have been discussed by a multidisciplinary team (Figure 9) and after the balance of benefits and side effects of each therapy modality has been considered by the patient with regard to their own individual circumstances23.Figure 9. Examples of members involved in the multi-disciplinary approach for prostate cancer patient management. Adapted from Murphy et al., 2019

Figure 9. Examples of members involved in the multi-disciplinary approach for prostate cancer patient management. Adapted from Murphy et al., 201923,56.

The specialists involved in the management of a patient with prostate cancer can include urologists, radiotherapists, oncologists, radiologists, pathologists, surgeons, clinical nurse specialists and psychologists23,54,56. A multidisciplinary approach to cancer care has been associated with a decrease in time from diagnosis to treatment, shorter time to completion of pre-treatment consultations and improved adherence to guidelines supported by the literature56,57.

Treatment options

Treatment options for prostate cancer vary based on disease severity and risk stratification23. Table 7 gives a brief summary of treatment options available for the EAU risk groups and advanced disease that is based on disease severity and risk group23. For exhaustive information on treatment guidelines, please refer to the European Association of Urologists prostate cancer guidelines (2020)23.

Table 7. Summary of treatment options available to prostate cancer patients based on European Association of Urology (EAU) risk groups. Adapted from the EAU-ESTRO-ESUR-SIOG Guidelines on Prostate Cancer, 202023.

Table 7. Summary of treatment options available to prostate cancer patients based on European Association of Urology (EAU) risk groups. Adapted from the EAU-ESTRO-ESUR-SIOG Guidelines on Prostate Cancer, 2020

Deferred Treatment

Active surveillance and watchful waiting are conservative management strategies that aim to avoid unnecessary treatment that may lead to genitourinary adverse events (AEs) and decreased quality of life (QoL)23.

Active surveillance aims to optimise the timing of curative intent treatment options in patients who have a life expectancy of greater than 10 years and includes an active decision not to treat the patient immediately23. The patient remains under close surveillance using assessment markers (for example, using DRE and PSA levels), and treatment is prompted by predefined thresholds indicative of progressive disease23.

Watchful waiting refers to symptom-guided palliative treatment that aims to minimise treatment-related toxicity23. Watchful waiting can be considered as an option for treating patients with a limited life expectancy (< 10 years) regardless of prostate cancer stage23.

Radical prostatectomy

Radical prostatectomy involves the removal of the entire prostate gland along with a margin of surrounding tissue58. The procedure may be accompanied by bilateral pelvic lymph node dissection23. Three radical prostatectomy approaches are currently used23:

  • open radical prostatectomy (RP)
  • laparoscopic radical prostatectomy (LRP)
  • robotic assisted laparoscopic prostatectomy (RALP).

Extended pelvic lymph node dissection (eLND) involves the removal of the lymph nodes closely associated with the prostate (overlying the external iliac artery and vein, within the obturator fossa located cranially and caudally to the obturator nerve, and nodes medial and lateral to the internal iliac artery) and provides important information for staging and prognosis23.

Urinary incontinence and erectile dysfunction are extremely common following radical prostatectomy59. Although generally transient, these can be long-term and risk increases significantly with increasing age59.

Radiotherapy

Radiotherapy is an important and valid alternative to surgery alone for curative intent therapy23. External beam radiotherapy (EBRT) and brachytherapy (internal radiotherapy) are the two treatment modalities commonly used as monotherapy or in combination with each other or androgen deprivation (hormonal therapy)23,60.

External beam radiation therapy (EBRT) is directed at the prostate gland from outside the body to target cancer cells60. EBRT may be used as a curative intent treatment in early stage disease, or to relieve symptoms in advanced disease60. The use of three-dimensional conformal radiotherapy (3D-CRT) and intensity-modulated radiation therapy (IMRT) have improved the quality and precision of EBRT and allows for dose escalation60. 3D-CRT uses imaging technologies to create a three-dimensional image of the tumour and nearby tissues and organs that is used for treatment planning to deliver concentrated radiation doses directly to the tumour whilst sparing healthy tissue61. IMRT uses multiple small radiation beams of varying intensities to precisely radiate the tumour60. The goal of IMRT is to bend the radiation dose to avoid or reduce exposure of healthy tissue and limit the side effects of treatment23,60.

Brachytherapy (also known as interstitial radiation) is an internal radiotherapy, which involves inserting radioactive ‘seeds’ close to the tumour cells60,62. Either permanent or temporary radioactive source implantation can be used in brachytherapy for patients with prostate cancer62.

Common side effects of EBRT and brachytherapy include bowel problems, urinary problems and erectile dysfunction60. Unlike with prostatectomy, erectile dysfunction and urinary issues may develop several years after radiation therapy60.

Less common options for clinically localised prostate cancer

Cryotherapy and high-intensity focused ultrasound (HIFU) have been developed as minimally invasive procedures aiming to provide equivalent treatment compared to radical prostatectomy, EBRT and brachytherapy, but with reduced toxicity23.

Hormonal therapies

Normal prostate cells are dependent on androgens such as testosterone to stimulate growth and tumour growth is similarly dependent on androgens63. If tumour cells are deprived of androgenic stimulation, they undergo apoptosis (programmed cell death)64. Hormone therapy for prostate cancer therefore aims to suppress androgen activity and is also referred to as androgen suppression therapy65. Therapies with differing modes of action may also be combined to achieve a complete (maximal or total) androgen blockade (CAB)23.

Androgen suppression can be achieved by65:

  • lowering testicular androgen levels
  • lowering androgen levels from the adrenal glands
  • stopping androgens from working
Lowering testicular androgen levels

Androgen deprivation therapy (ADT) aims to lower testicular androgen levels65. Surgical castration remains the primary treatment modality for androgen deprivation, against which all other treatments are rated23. It leads to a considerable decline in testosterone to castrate levels defined by testosterone levels of less than 20 ng/dL23.

Androgen deprivation can also be achieved through ADT drugs, including luteinizing hormone-releasing hormone (LHRH) agonists and antagonists (Table 8)65.

Table 8. Mode of the action for prostate cancer hormone therapies18,65.

Table 8. Mode of the action for prostate cancer hormone therapies

LHRH, also known as gonadotropin-releasing hormone (GnRH), is secreted by the hypothalamus and binds to its receptor in the pituitary to stimulate the release of luteinising hormone (LH) and follicle stimulating hormone (FSH)66. These hormones trigger the release of androgens (sex-steroids) to promote growth and development of both normal and tumour cells66. LHRH antagonists induce a state of sex steroid deprivation by competitive blockade of pituitary LHRH receptors, whereas LHRH agonists achieve a similar effect by down regulating the pituitary receptors for LHRH67. This reduces the levels of FSH and LH, and subsequently the levels of sex steroids.

Lowering androgen levels from the adrenal glands

Adrenal glands and prostate cancer cells can also produce androgens65. Drugs such as ketoconazole and abiraterone can block the formation of androgens made in this way65.

Abiraterone acetate is a new hormonal agent that aims to lower androgen levels produced by the adrenal glands65. New hormonal agents (NHA) target the androgen axis and were originally developed to treat patients who no longer respond to androgen deprivation therapy (Table 9)23.

Table 9. Mode of action of generic new hormonal agents (NHA) that target the androgen axis for prostate cancer treatment63,65,68,69.

Table 9. Mode of action of generic new hormonal agents (NHA) that target the androgen axis for prostate cancer treatment

Abiraterone is a CYP17 inhibitor that significantly decreases the intracellular testosterone level by suppressing CYP17-mediated androgen biosynthesis63. Abiraterone acetate must be used together with the corticosteroid prednisone/prednisolone and is indicated for the treatment of metastastic prostate cancer (mCRPC) and metastatic high-risk hormone sensitive prostate cancer (mHSPC)70.

Stopping androgens from working

Anti-androgen drugs such as flutamide, bicalutamide and nilutamide bind to androgen receptors on prostate cancer cells, thereby preventing androgen binding and signalling for growth65.

Enzalutamide is a NHA that is an AR signalling inhibitor with a high affinity for the androgen receptor (AR)63. It blocks androgen binding to AR, inhibits AR translocation to the nucleus, and impairs AR binding to DNA63. Enzalutamide is indicated for the treatment of high-risk nonmetastatic castration-resistant prostate cancer (nmCRPC) and metastatic castration-resistant prostate cancer (mCRPC)71.

Both apalutamide and darolutamide are more recently approved NHAs now indicated for nmCRPC72–75. Apalutamide in combination with ADT is additionally indicated for the treatment of metastatic hormone-sensitive prostate cancer (mHSPC)72,74. Both apalutamide and darolutamide bind to the ligand binding domain of AR to prevent its nuclear translocation, thereby preventing DNA binding and AR-mediated transcription68,69.

Chemotherapy

Cytotoxic chemotherapy agents such as docetaxel or cabazitaxel can delay progression and improve survival for patients with advanced prostate cancer76. The mode of action for both docetaxel and cabazitaxel are as anti-mitotic agents77,78. Docetaxel is indicated for mHSPC and mCRPC. Cabazitaxel is indicated as a therapy for mCRPC patients previously exposed to docetaxel77,78.

Radium-223

Radium-223 (Ra-223) is a bone-seeking alpha particle–emitting radiopharmaceutical that mimics calcium uptake at osteoblastic lesions where it leads to DNA damage and tumour death79. Ra-223 can be considered as a treatment option for patients with CRPC and symptomatic bone metastases23. This targeted treatment also possesses a short tissue-penetration range that altogether allows for a low toxicity profile80.

Bone protection therapies

The most common site for metastatic spread in prostate cancer is bone81,82. Complications associated with this include bone pain, hypercalcaemia, fractures and spinal cord compression81,82. Complications may be compounded further by issues with bone mineral density (BMD) due to long term use of ADT83

Bisphosphonates such as zoledronic acid are often used to increase bone mineral density81. They do this by preventing bone breakdown by osteoclasts and can encourage osteoblast-mediated growth of new bone tissue81. Zoledronic acid is also used to prevent complications from bone metastases and to treat hypercalcaemia associated with cancer84. Denosumab is a human monoclonal antibody that is indicated for the prevention of skeletal related events in patients with advances malignancies involving bone83,85. Osteoclast formation, activity and survival is driven by RANKL (receptor activator of nuclear factor κB ligand)23. Denosumab inhibits RANKL, thereby reducing osteoclast formation and bone deterioration23.

Palliative care

Advanced prostate cancer can be a debilitating disease. A multidisciplinary approach is important for the optimal management of symptoms impacting on quality of life including pain, constipation, anorexia, nausea, fatigue and depression23. Palliative treatments such as external beam radiation therapy (EBRT), palliative surgery and bone protection therapies may be used alone or as adjuncts to systemic therapies for advanced disease23.

Clinical trials

The introduction of abiraterone and enzalutamide for treating metastatic CRPC signalled a paradigm shift in the management of advanced prostate cancer. Recent clinical trials on these new hormonal agents have furthermore led to approvals for abiraterone and apalutamide in mHSPC, and for enzalutamide, apalutamide and darolutamide in nonmetastatic castration resistant disease (nmCRPC)70–72,75. Despite these advances, metastatic prostate cancer remains incurable86. To this end, new sequences for existing treatments are being explored in clinical trials, as well as novel therapeutic compounds that may be suitable for patients who understand the risks involved in partaking in clinical trials. 

Treatment pathways

The treatment journey for patients with prostate cancer is complex and dependent on the clinical state of the disease at diagnosis and how it progresses over time87. Multiple clinical states have been defined in prostate cancer based on the status of the primary tumour, presence or absence of detectable metastases, prior and current treatment, and serum testosterone levels (non-castrate/castrate)88.

Broadly, prostate cancer disease states include88:

  • Localised prostate cancer
  • Locally advanced prostate cancer
  • Biochemically recurrent disease
  • Nonmetastatic castration-resistant prostate cancer (nmCRPC)
  • Metastatic hormone sensitive prostate cancer (mHSPC)
  • Metastatic castration-resistant prostate cancer (mCRPC)

Metastatic prostate cancer is incurable and represents the unmet need in prostate cancer care86. The approval of multiple life-extending treatments over the past decade has transformed the treatment landscape and ongoing clinical trials indicate that this change is set to continue87

The evolving prostate cancer treatment landscape

Androgen receptor (AR) signalling is the primary driver of prostate cancer progression and androgen deprivation therapy (ADT) has been the backbone of care63. ADT aims to lower testosterone to castration levels of <20 ng/dL which can be achieved either through surgery or using hormone therapies23. Prostate cancers that respond to ADT are described as hormone sensitive prostate cancer (HSPC), whereas those that progress despite castrate levels of testosterone are known as castration-resistant prostate cancers (CRPC)88.

Figure 10. Alterations in androgen signalling associated with progression to castration resistant prostate cancer (CRPC). Adapted from Attard et al., 2016

Figure 10. Alterations in androgen signalling associated with progression to castration resistant prostate cancer (CRPC). Adapted from Attard et al., 201689. The key targets for the new hormonal agents are highlighted. AR, androgen receptor; ARE, androgen receptor element; CRPC, castration-resistant prostate cancer; CYP17A1, cytochrome P450 17A1; GR, growth hormone receptor; PI3K, phosphoinositide 3-kinase; PSA, prostate specific antigen; TMPRSS2-ERG, transmembrane protease, serine 2-ETS-related gene fusion.

The onset of castration resistance is complex and can involve multiple mechanisms (Figure 10). High intracellular androgen levels have been observed in CRPC cells compared to androgen sensitive cells, as well as an overexpression of the androgen receptor23,90. This observation led to the development of new hormonal agents (NHA; for example, abiraterone and enzalutamide) that target the androgen axis, thereby providing a treatment option for patients with castration resistant disease23. For instance, abiraterone inhibits the CYP17 enzyme that is necessary for de novo androgen synthesis whereas enzalutamide has three-fold inhibitor activity; it inhibits androgen binding to the androgen receptor, androgen entry into the nucleus and androgen receptor binding to DNA63. Both abiraterone and enzalutamide were initially approved for treating mCRPC patients; however, increasing clinical trial evidence has led to approvals for their use earlier on in the patient’s treatment journey (Figure 11)70,71. Two recently approved NHAs, apalutamide and darolutamide, are now indicated for nmCRPC69,72,73,91. Apalutamide is also EMA and FDA approved for the treatment of mHSPC72,74. Alternative treatments that do not target the androgen axis have also been integrated into clinical practice. They include docetaxel and cabazitaxel which are both anti-mitotic taxane chemotherapy agents77,78.

Please refer to the European Association of Urology (EAU) and European Society for Medical Oncology (ESMO) guidelines on prostate cancer for comprehensive details on treatment options23,48.  

Figure 11. Prostate cancer treatment journey based on changing disease states. Existing standard of care treatments according to ESMO and EAU guidelines

Figure 11. Prostate cancer treatment journey based on changing disease states70–73,77,78,92,93. Existing standard of care treatments according to ESMO and EAU guidelines23,48. *Approved for use post-docetaxel treatment. †Indicated for patients with symptomatic bone metastases and no known visceral metastases, in progression after at least two prior lines of systemic therapy for mCRPC. ADT, androgen deprivation therapy; mHSPC, metastatic hormone-sensitive prostate cancer; mCRPC, metastatic castration-resistant prostate cancer; nmCRPC, nonmetastatic castration-resistant prostate cancer; PSA, prostate-specific antigen.

Metastatic castration-resistant prostate cancer

Metastatic castration-resistant prostate cancer (mCRPC) is currently incurable, with some improvement in overall survival since the introduction of systemic drugs compared to non-systemic treatment (2.8 years versus 2.2 years)94. Currently, approved treatments for mCRPC include abiraterone, enzalutamide, docetaxel, cabazitaxel and radium-22323,48,70,71,77,78,93

First-line clinical trials

The key clinical trial studies that led to the approvals for NHAs and taxanes in the first-line treatment of mCRPC are highlighted in Table 1095–98.

Table 10. Key first-line clinical trials for metastatic castration-resistant prostate cancer (mCRPC)95–98.

Table 10. Key first-line clinical trials for metastatic castration-resistant prostate cancer (mCRPC)

The TAX 327 clinical trial found that docetaxel in the first-line setting led to a modest increase in overall survival with adverse events including fatigue, nausea, vomiting, alopecia and diarrhoea95. NHA in the first-line setting with either enzalutamide or abiraterone led to improvements in overall survival96,97. Common adverse events for abiraterone were fatigue, arthralgia, peripheral oedema and hepatotoxicity whereas fatigue, back pain, arthralgia and hypertension were reported for enzalutamide96,97.

Second-line clinical trials

In the key second-line clinical trials post-docetaxel treatment for mCRPC, improvements in overall survival were observed for mCRPC patients treated with abiraterone, enzalutamide, cabazitaxel and radium-223 (Table 11)99–102. The most common Grade 3–4 adverse events observed for abiraterone were fatigue and anaemia99. For enzalutamide, the most common Grade 3-4 adverse events observed included fatigue and diarrhoea, and for cabazitaxel, this was neutropenia and diarrhoea100,101.

Table 11: Key second-line clinical trials post-docetaxel treatment for metastatic castration-resistant prostate cancer (mCRPC)99–102.

Table 11: Key second-line clinical trials post-docetaxel treatment for metastatic castration-resistant prostate cancer (mCRPC)Sequencing therapies for mCRPC

Currently, docetaxel, abiraterone and enzalutamide are used to treat mCRPC patients, both as first- and second-line therapies (Figure 11). There is currently no clear evidence for, or guidance on, the optimal treatment sequence for mCRPC. Alternate sequential treatment for enzalutamide and abiraterone shows limited additional benefit as indicated by modest differences in overall PSA responses, though abiraterone followed by enzalutamide may be slightly better103,104. In the Phase III CARD trial, cabazitaxel was compared with NHA treatment (abiraterone or enzalutamide) in mCRPC patients previously treated with docetaxel who progressed ≤12 months while receiving the alternative NHA105. This trial found that these patients had longer progression-free and overall survival when treated with cabazitaxel than when treated with the alternative NHA105

A key issue that complicates sequencing of treatments is the possibility of intra cross-resistance (resistance shared between agents in the same class of drug) and inter cross-resistance (between agents in different classes)106. For the latter, there is some evidence that docetaxel may have reduced efficacy when given following NHA treatments; however, more evidence is needed to better understand cross-resistance103.

Nonmetastatic castration-resistant prostate cancer (nmCRPC)

It is customary practice to continue using androgen deprivation therapy (ADT) to treat nmCRPC, despite the onset of castration resistance107. Three recent Phase III double-blind studies (SPARTAN, PROSPER and ARAMIS) investigated the use of new hormonal agents in nmCRPC (Table 12)69,91,108. In these studies, ADT alone was compared to ADT plus apalutamide (SPARTAN), enzalutamide (PROSPER) or darolutamide (ARAMIS). Altogether the findings indicate that all three NHAs significantly increased metastasis-free survival (MFS) and median overall survival (final endpoint not yet reached in ARAMIS) with tolerable side effects69,91,108–111. These findings have led to FDA and EMA approvals for apalutamide, enzalutamide and daroluatmide in nmCRPC71–75,112.

Table 12. Key clinical trials for nonmetastatic castration-resistant prostate cancer (nmCRPC)69,91,108–111.

Table 12. Key clinical trials for nonmetastatic castration-resistant prostate cancer (nmCRPC)

Metastatic hormone sensitive prostate cancer (mHSPC)

Most mHSPC patients respond to the standard of care therapy, ADT; however, the majority will progress to mCRPC after 2–3 years113. Progression to metastatic disease most commonly follows biochemical recurrence; however, diagnosis of de novo mHSPC is also increasing, which is partly due to improved imaging and reduced PSA screening114. To date, there have been multiple clinical trials to assess agents used in the mCRPC setting for mHSPC (Table 13). This has led to recent approvals of alternative treatments for mHSPC, including docetaxel, abiraterone and apalutamide70,72,78. Of note, de novo mHSPC patients tend to have a worse outcome compared to mHSPC patients who progress from localised or locally advanced disease115.

Table 13: Key clinical trials for metastatic hormone sensitive prostate cancer (mHSPC)116–121.

Table 13: Key clinical trials for metastatic hormone sensitive prostate cancer (mHSPC)

Docetaxel

Three clinical trials have compared docetaxel plus ADT to ADT alone in mHSPC patients (Table 13)116,117,120. The GETUG-AFU-15 study identified no significant difference in overall survival when docetaxel was added to ADT treatment compared to ADT alone116. However, the multi-stage STAMPEDE trial identified some benefit of docetaxel addition to ADT treatment for all patients, whereas the later Phase III study (CHAARTED) identified a benefit only in mHSPC patients who presented with high volume disease (51.2 months versus 34.4 months)117,120. A follow-up report on STAMPEDE showed no difference based on metastatic burden as defined by the CHAARTED study122. A possible explanation for the difference in outcome between the two studies relate to differences in patient selection. Approximately 95% of patients enrolled on to the STAMPEDE trial had de novo mHSPC, versus only 25% of patients enrolled on the CHAARTED trial122. Based on these findings of the clinical trials, docetaxel is now approved for mHSPC78. Adverse events were found to be consistently higher in patients receiving docetaxel compared to those treated with ADT throughout the studies116,117,120.

New Hormonal Agents (NHAs)

In the STAMPEDE Phase III clinical trial, abiraterone in combination with prednisolone and ADT had a 3-year survival of 83% compared to 76% for mHSPC patients treated with ADT119. In the LATITUDE study, the likelihood of survival significantly improved by 38% when abiraterone and prednisone were added to ADT versus ADT alone118. Based on these findings, abiraterone with prednisone is now approved for mHSPC70.

Apalutamide has also shown significant increases in overall survival and radiographic progression compared to ADT treatment alone for mHSPC in a Phase III double-blind clinical trial (Table 13)121. These findings have led to recent FDA and EMA approvals for apalutamide for mHSPC71,72.

References

  1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424.
  2. Rawla P. Epidemiology of Prostate Cancer. World J Oncol. 2019;10(2):63–89.
  3. Ferlay J, Colombet M, Soerjomataram I, Dyba T, Randi G, Bettio M, et al. Cancer incidence and mortality patterns in Europe: Estimates for 40 countries and 25 major cancers in 2018. European Journal of Cancer. 2018;103:356–387.
  4. Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JWW, Comber H, et al. Cancer incidence and mortality patterns in Europe: Estimates for 40 countries in 2012. Eur J Cancer. 2013;49(6):1374–1403.
  5. Prostate cancer survival statistics | Cancer Research UK. https://www.cancerresearchuk.org/health-professional/cancer-statistics/statistics-by-cancer-type/prostate-cancer. Accessed 14 June 2020.
  6. Pernar CH, Ebot EM, Wilson KM, Mucci LA. The Epidemiology of Prostate Cancer. Cold Spring Harbor perspectives in medicine. 2018;8(12). doi:10.1101/cshperspect.a030361.
  7. Cancer Statistics Review, 1975-2013 - Previous Version - SEER Cancer Statistics Review. https://seer.cancer.gov/archive/csr/1975_2013/. Accessed 14 June 2020.
  8. Taksler GB, Keating NL, Cutler DM. Explaining racial differences in prostate cancer mortality. Cancer. 2012;118(17):4280–4289.
  9. Haiman CA, Chen GK, Blot WJ, Strom SS, Berndt SI, Kittles RA, et al. Characterizing genetic risk at known prostate cancer susceptibility loci in African Americans. PLoS Genet. 2011;7(5):e1001387.
  10. Lichtenstein P, Holm N V., Verkasalo PK, Iliadou A, Kaprio J, Koskenvuo M, et al. Environmental and heritable factors in the causation of cancer: Analyses of cohorts of twins from Sweden, Denmark, and Finland. N Engl J Med. 2000;343(2):78–85.
  11. Mucci LA, Hjelmborg JB, Harris JR, Czene K, Havelick DJ, Scheike T, et al. Familial risk and heritability of cancer among twins in nordic countries. JAMA - J Am Med Assoc. 2016;315(1):68–76.
  12. Hemminki K, Czene K. Attributable risks of familial cancer from the family-cancer database. Cancer Epidemiol Biomarkers Prev. 2002;11(12):1638–1644.
  13. Brandt A, Sundquist J, Hemminki K. Risk for incident and fatal prostate cancer in men with a family history of any incident and fatal cancer. Ann Oncol. 2012;23(1):251–256.
  14. Bratt O, Drevin L, Akre O, Garmo H, Stattin P. Family History and Probability of Prostate Cancer, Differentiated by Risk Category: A Nationwide Population-Based Study. J Natl Cancer Inst. 2016;108(10). doi:10.1093/jnci/djw110.
  15. Chattopadhyay S, Hemminki O, Försti A, Sundquist K, Sundquist J, Hemminki K. Impact of family history of cancer on risk and mortality of second cancers in patients with prostate cancer. Prostate Cancer Prostatic Dis. 2019;22(1):143–149.
  16. Moran A, O’Hara C, Khan S, Shack L, Woodward E, Maher ER, et al. Risk of cancer other than breast or ovarian in individuals with BRCA1 and BRCA2 mutations. Fam Cancer. 2012;11(2):235–242.
  17. Mersch J, Jackson MA, Park M, Nebgen D, Peterson SK, Singletary C, et al. Cancers associated with BRCA1 and BRCA2 mutations other than breast and ovarian. Cancer. 2015;121(2):269–75.
  18. NCI Dictionaries - National Cancer Institute. https://www.cancer.gov/publications/dictionaries. Accessed 14 June 2020.
  19. Page EC, Bancroft EK, Brook MN, Assel M, Hassan Al Battat M, Thomas S, et al. Interim Results from the IMPACT Study: Evidence for Prostate-specific Antigen Screening in BRCA2 Mutation Carriers. Eur Urol. 2019;76(6):831–842.
  20. Tikkinen KAO, Dahm P, Lytvyn L, Heen AF, Vernooij RWM, Siemieniuk RAC, et al. Prostate cancer screening with prostate-specific antigen (PSA) test: A clinical practice guideline. BMJ. 2018;362. doi:10.1136/bmj.k3581.
  21. Prostate cancer - Symptoms - NHS. https://www.nhs.uk/conditions/prostate-cancer/symptoms/. Accessed 14 June 2020.
  22. Prostate Cancer: Symptoms and Signs | Cancer.Net. https://www.cancer.net/cancer-types/prostate-cancer/symptoms-and-signs. Accessed 14 June 2020.
  23. Mottet N, Cornford P, van den Bergh R, Briers E, De Santis M, Fanti S, et al. EAU-ESTRO-ESUR-SIOG Guidelines on Prostate Cancer. 2020 https://uroweb.org/guideline/prostate-cancer/. Accessed 14 June 2020.
  24. Rare prostate cancers | Prostate Cancer UK. https://prostatecanceruk.org/prostate-information/further-help/rare-prostate-cancer. Accessed 14 June 2020.
  25. The PSA test | Prostate Cancer UK. https://prostatecanceruk.org/prostate-information/prostate-tests/psa-test. Accessed 14 June 2020.
  26. Referenced with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for Prostate Cancer V.2.2020. © National Comprehensive Cancer Network, Inc. 2020. All rights reserved. Accessed [14-06-2020]. To view the most recent and complete version of the guideline, go online to NCCN.org. NCCN makes no warranties of any kind whatsoever regarding their content, use or application and disclaims any responsibility for their application or use in any way.
  27. Padhani AR, Lecouvet FE, Tunariu N, Koh DM, De Keyzer F, Collins DJ, et al. Rationale for Modernising Imaging in Advanced Prostate Cancer. European Urology Focus. 2017;3(2–3):223–239.
  28. Descotes JL. Diagnosis of prostate cancer. Asian Journal of Urology. 2019;6(2):129–136.
  29. Tests to Diagnose and Stage Prostate Cancer. https://www.cancer.org/cancer/prostate-cancer/detection-diagnosis-staging/how-diagnosed.html. Accessed 14 June 2020.
  30. Ahmed HU, El-Shater Bosaily A, Brown LC, Gabe R, Kaplan R, Parmar MK, et al. Diagnostic accuracy of multi-parametric MRI and TRUS biopsy in prostate cancer (PROMIS): a paired validating confirmatory study. Lancet. 2017;389(10071):815–822.
  31. Treglia G, Pereira Mestre R, Ferrari M, Bosetti DG, Pascale M, Oikonomou E, et al. Radiolabelled choline versus PSMA PET/CT in prostate cancer restaging: a meta-analysis. Am J Nucl Med Mol Imaging. 2019;9(2):127–139.
  32. Evans JD, Jethwa KR, Ost P, Williams S, Kwon ED, Lowe VJ, et al. Prostate cancer–specific PET radiotracers: A review on the clinical utility in recurrent disease. Pract Radiat Oncol. 2018;8(1):28–39.
  33. Ulmert D, Solnes L, Thorek DL. Contemporary approaches for imaging skeletal metastasis. Bone Res. 2015;3:15024.
  34. Planning radiotherapy treatment for advanced prostate cancer | Cancer Research UK. https://www.cancerresearchuk.org/about-cancer/prostate-cancer/advanced-cancer/advanced-treatment/advanced-radiotherapy/external-radiotherapy-treatment. Accessed 14 June 2020.
  35. Harvey CJ, Pilcher J, Richenberg J, Patel U, Frauscher F. Applications of transrectal ultrasound in prostate cancer. Br J Radiol. 2012;85 Spec No 1:S3-17.
  36. MRI and multiparametric MRI (mpMRI) scan | Prostate cancer | Cancer Research UK. https://www.cancerresearchuk.org/about-cancer/prostate-cancer/getting-diagnosed/tests/mri-scan. Accessed 14 June 2020.
  37. Moghul M, Somani B, Lane T, Vasdev N, Chaplin B, Peedell C, et al. Detection rates of recurrent prostate cancer: 68Gallium (Ga)-labelled prostate-specific membrane antigen versus choline PET/CT scans. A systematic review. Ther Adv Urol.11:1756287218815793.
  38. Barbosa FG, Queiroz MA, Nunes RF, Viana PCC, Marin JFG, Cerri GG, et al. Revisiting Prostate Cancer Recurrence with PSMA PET: Atlas of Typical and Atypical Patterns of Spread. RadioGraphics. 2019;39(1):186–212.
  39. Stages, types and grades | Prostate cancer | Cancer Research UK. https://www.cancerresearchuk.org/about-cancer/prostate-cancer/stages. Accessed 14 June 2020.
  40. Localised prostate cancer | Prostate Cancer UK. https://prostatecanceruk.org/prostate-information/just-diagnosed/localised-prostate-cancer. Accessed 14 June 2020.
  41. Locally advanced prostate cancer | Prostate Cancer UK. https://prostatecanceruk.org/prostate-information/just-diagnosed/locally-advanced-prostate-cancer. Accessed 14 June 2020.
  42. Advanced prostate cancer | Prostate Cancer UK. https://prostatecanceruk.org/prostate-information/just-diagnosed/advanced-prostate-cancer. Accessed 14 June 2020.
  43. Morphology & Grade | SEER Training. https://training.seer.cancer.gov/prostate/abstract-code-stage/morphology.html. Accessed 14 June 2020.
  44. The Gleason score and Grade Groups for prostate cancer | Cancer Research UK. https://www.cancerresearchuk.org/about-cancer/prostate-cancer/stages/grades. Accessed 14 June 2020.
  45. Prostate Cancer: Stages and Grades | Cancer.Net. https://www.cancer.net/cancer-types/prostate-cancer/stages-and-grades. Accessed 14 June 2020.
  46. Stelcer E, Konkol M, Głȩboka A, Suchorska WM. Liquid biopsy in oligometastatic prostate cancer—A biologist’s point of view. Frontiers in Oncology. 2019;9(AUG):775.
  47. Prostate Cancer Stages and Other Ways to Assess Risk. https://www.cancer.org/cancer/prostate-cancer/detection-diagnosis-staging/staging.html. Accessed 14 June 2020.
  48. Parker C, Castro E, Fizazi K, Heidenreich A, Ost P, Procopio G, et al. Journal Pre-proof Prostate Cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2020. doi:10.1016/j.annonc.2020.06.011.
  49. Latham A, Srinivasan P, Kemel Y, Shia J, Bandlamudi C, Mandelker D, et al. Microsatellite Instability Is Associated With the Presence of Lynch Syndrome Pan-Cancer. J Clin Oncol. 2019;37(4):286–295.
  50. Haraldsdottir S, Hampel H, Wei L, Wu C, Frankel W, Bekaii-Saab T, et al. Prostate cancer incidence in males with Lynch syndrome. Genet Med. 2014;16(7):553–557.
  51. Ryan S, Jenkins MA, Win AK. Risk of prostate cancer in Lynch syndrome: a systematic review and meta-analysis. Cancer Epidemiol Biomarkers Prev. 2014;23(3):437–49.
  52. Hu JC, Tosoian JJ, Qi J, Kaye D, Johnson A, Linsell S, et al. Clinical Utility of Gene Expression Classifiers in Men With Newly Diagnosed Prostate Cancer. JCO Precis Oncol. 2018;(2):1–15.
  53. Athie A, Arce-Gallego S, Gonzalez M, Morales-Barrera R, Suarez C, Casals Galobart T, et al. Targeting DNA Repair Defects for Precision Medicine in Prostate Cancer. Curr Oncol Rep. 2019;21(5):42.
  54. Gomella LG, Lin J, Hoffman-Censits J, Dugan P, Guiles F, Lallas CD, et al. Enhancing Prostate Cancer Care Through the Multidisciplinary Clinic Approach: A 15-Year Experience. J Oncol Pract. 2010;6(6):e5–e10.
  55. Selby P, Popescu R, Lawler M, Butcher H, Costa A. The Value and Future Developments of Multidisciplinary Team Cancer Care. Am Soc Clin Oncol Educ B. 2019;(39):332–340.
  56. Murphy DG, Hofman MS, Azad A, Violet J, Hicks RJ, Lawrentschuk N. Going nuclear: it is time to embed the nuclear medicine physician in the prostate cancer multidisciplinary team. BJU Int. 2019. doi:10.1111/bju.14814.
  57. Sciarra A, Gentile V, Panebianco V. Multidisciplinary management of Prostate Cancer: how and why. Am J Clin Exp Urol. 2013;1(1):12–7.
  58. Radical prostatectomy: MedlinePlus Medical Encyclopedia. https://medlineplus.gov/ency/article/007300.htm. Accessed 14 June 2020.
  59. Prabhu V, Lee T, McClintock TR, Lepor H. Short-, Intermediate-, and Long-term Quality of Life Outcomes Following Radical Prostatectomy for Clinically Localized Prostate Cancer. Rev Urol. 2013;15(4):161–77.
  60. Radiation Therapy for Prostate Cancer. https://www.cancer.org/cancer/prostate-cancer/treating/radiation-therapy.html. Accessed 14 June 2020.
  61. 3D CRT (Three-Dimensional Conformal Radiation Therapy) | Moffitt. https://moffitt.org/treatments/radiation-therapy/3d-crt-three-dimensional-conformal-radiation-therapy/. Accessed 14 June 2020.
  62. Koukourakis G, Kelekis N, Armonis V, Kouloulias V. Brachytherapy for prostate cancer: A systematic review. Adv Urol. 2009. doi:10.1155/2009/327945.
  63. Schalken J, Fitzpatrick JM. Enzalutamide: targeting the androgen signalling pathway in metastatic castration-resistant prostate cancer. BJU Int. 2016;117(2):215–225.
  64. McKenzie S, Kyprianou N. Apoptosis evasion: the role of survival pathways in prostate cancer progression and therapeutic resistance. J Cell Biochem. 2006;97(1):18–32.
  65. Hormone Therapy for Prostate Cancer. https://www.cancer.org/cancer/prostate-cancer/treating/hormone-therapy.html. Accessed 14 June 2020.
  66. Marques P, Skorupskaite K, George JT, Anderson RA. Physiology of GNRH and Gonadotropin Secretion. 2000. MDText.com, Inc. http://www.ncbi.nlm.nih.gov/pubmed/25905297. Accessed 14 June 2020.
  67. Tolkach Y, Joniau S, Van Poppel H. Luteinizing hormone-releasing hormone (LHRH) receptor agonists vs antagonists: A matter of the receptors? BJU International. 2013;111(7):1021–1030.
  68. Fizazi K, Smith MR, Tombal B. Clinical Development of Darolutamide: A Novel Androgen Receptor Antagonist for the Treatment of Prostate Cancer. Clinical Genitourinary Cancer. 2018;16(5):332–340.
  69. Smith MR, Saad F, Chowdhury S, Oudard S, Hadaschik BA, Graff JN, et al. Apalutamide treatment and metastasis-free survival in prostate cancer. N Engl J Med. 2018;378(15):1408–1418.
  70. Abiraterone acetate ZYTIGA: Summary of Product Characteristics (SmPC) - (emc). https://www.medicines.org.uk/emc/product/2381. Accessed 14 June 2020.
  71. Enzalutamide Xtandi - Summary of Product Characteristics (SmPC) - (emc). https://www.medicines.org.uk/emc/product/10318/smpc. Accessed 14 June 2020.
  72. Apalutamide Erleada- Summary of Product Characteristics (SmPC) - (emc). https://www.medicines.org.uk/emc/product/9832#. Accessed 14 June 2020.
  73. Darolutamide NUBEQA- Summary of product characteristics - FDA. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/212099Orig1s000lbl.pdf. Accessed 14 June 2020.
  74. Apalutamide, Erleada; Highlights of prescribing information (FDA). https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/210951s001lbl.pdf. Accessed 14 June 2020.
  75. Darolutamide NUBEQA - Summary of Product Characteristics (SmPC) - (emc). https://www.medicines.org.uk/emc/product/11324/smpc. Accessed 14 June 2020.
  76. Chemotherapy | Prostate Cancer UK. https://prostatecanceruk.org/prostate-information/our-publications/publications/chemotherapy. Accessed 14 June 2020.
  77. Cabazitaxel Jevtana- Summary of Product Characteristics (SmPC) - (emc). https://www.medicines.org.uk/emc/product/4541. Accessed 14 June 2020.
  78. Docetaxel Accord concentrate for solution for infusion - Summary of Product Characteristics (SmPC) - (emc). https://www.medicines.org.uk/emc/product/2464/smpc. Accessed 14 June 2020.
  79. Costa RP, Cardile D, Murabito A, Tripoli V, Verderame F. Ra223 in Bone Metastases with Osteolytic Activity. World J Nucl Med. 2018;17(2):116–119.
  80. Costa RP, Bordonaro S, Cappuccio F, Tripoli V, Murabito A, Licari M, et al. Hematologic toxicity of radium-223 in elderly patients with metastatic Castration Resistant Prostate Cancer: a real-life experience. Prostate Int. 2019;7(1):25–29.
  81. Preventing and treating Prostate Cancer Spread to Bones. https://www.cancer.org/cancer/prostate-cancer/treating/treating-pain.html. Accessed 14 June 2020.
  82. Blum R SD, Fleishman SB. The Multidisciplinary Approach to Bone Metastases. Cancer Network. 2003. https://www.cancernetwork.com/bone-metastases/multidisciplinary-approach-bone-metastases. Accessed 14 June 2020.
  83. Smith MR, Egerdie B, Toriz NH, Feldman R, Tammela TLJ, Saad F, et al. Denosumab in Men Receiving Androgen-Deprivation Therapy for Prostate Cancer. N Engl J Med. 2009;361(8):745–755.
  84. Zoledronic Acid Hospira - Summary of Product Characteristics (SmPC) - (EMC). https://www.medicines.org.uk/emc/product/9507/smpc. Accessed 14 June 2020.
  85. Denosumab Xgeva Summary of Product Characteristics (SmPC) - (emc). https://www.medicines.org.uk/emc/product/4675/smpc. Accessed 14 June 2020.
  86. Albala DM. Imaging and treatment recommendations in patients with castrate-resistant prostate cancer. Rev Urol. 2017;19(3):200–202.
  87. Caffo O, Maines F, Kinspergher S, Veccia A, Messina C. Sequencing strategies in the new treatment landscape of prostate cancer. Future Oncology. 2019;15(25):2967–2982.
  88. Scher HI, Solo K, Valant J, Todd MB, Mehra M. Prevalence of prostate cancer clinical states and mortality in the United States: Estimates using a dynamic progression model. PLoS One. 2015;10(10). doi:10.1371/journal.pone.0139440.
  89. Attard G, Parker C, Eeles RA, Schröder F, Tomlins SA, Tannock I, et al. Prostate cancer. The Lancet. 2016;387(10013):70–82.
  90. Montgomery RB, Mostaghel EA, Vessella R, Hess DL, Kalhorn TF, Higano CS, et al. Maintenance of intratumoral androgens in metastatic prostate cancer: a mechanism for castration-resistant tumor growth. Cancer Res. 2008;68(11):4447–54.
  91. Fizazi K, Shore N, Tammela TL, Ulys A, Vjaters E, Polyakov S, et al. Darolutamide in Nonmetastatic, Castration-Resistant Prostate Cancer. N Engl J Med. 2019;380(13):1235–1246.
  92. Crawford ED, Petrylak D, Sartor O. Navigating the evolving therapeutic landscape in advanced prostate cancer. Urol Oncol. 2017;35S:S1–S13.
  93. Radium-223 - Xofigo - Summary of Product Characteristics (SmPC) - (emc). https://www.medicines.org.uk/emc/product/5204/smpc. Accessed 14 June 2020.
  94. Francini E, Gray KP, Shaw GK, Evan CP, Hamid AA, Perry CE, et al. Impact of new systemic therapies on overall survival of patients with metastatic castration-resistant prostate cancer in a hospital-based registry. Prostate Cancer Prostatic Dis. 2019;22(3):420–427.
  95. Tannock IF, De Wit R, Berry WR, Horti J, Pluzanska A, Chi KN, et al. Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. N Engl J Med. 2004;351(15):1502–1512.
  96. Ryan CJ, Smith MR, De Bono JS, Molina A, Logothetis CJ, De Souza P, et al. Abiraterone in metastatic prostate cancer without previous chemotherapy. N Engl J Med. 2013;368(2):138–148.
  97. Beer TM, Armstrong AJ, Rathkopf DE, Loriot Y, Sternberg CN, Higano CS, et al. Enzalutamide in metastatic prostate cancer before chemotherapy. N Engl J Med. 2014;371(5):424–433.
  98. Ryan CJ, Smith MR, Fizazi K, Saad F, Mulders PFA, Sternberg CN, et al. Abiraterone acetate plus prednisone versus placebo plus prednisone in chemotherapy-naive men with metastatic castration-resistant prostate cancer (COU-AA-302): Final overall survival analysis of a randomised, double-blind, placebo-controlled phase 3 study. Lancet Oncol. 2015;16(2):152–160.
  99. Fizazi K, Scher HI, Molina A, Logothetis CJ, Chi KN, Jones RJ, et al. Abiraterone acetate for treatment of metastatic castration-resistant prostate cancer: Final overall survival analysis of the COU-AA-301 randomised, double-blind, placebo-controlled phase 3 study. Lancet Oncol. 2012;13(10):983–992.
  100. Scher HI, Fizazi K, Saad F, Taplin ME, Sternberg CN, Miller K, et al. Increased survival with enzalutamide in prostate cancer after chemotherapy. N Engl J Med. 2012;367(13):1187–1197.
  101. De Bono JS, Oudard S, Ozguroglu M, Hansen S, MacHiels JP, Kocak I, et al. Prednisone plus cabazitaxel or mitoxantrone for metastatic castration-resistant prostate cancer progressing after docetaxel treatment: A randomised open-label trial. Lancet. 2010;376(9747):1147–1154.
  102. Nilsson S, Franzén L, Parker C, Tyrrell C, Blom R, Tennvall J, et al. Two-year survival follow-up of the randomized, double-blind, placebo-controlled phase II study of radium-223 chloride in patients with castration-resistant prostate cancer and bone metastases. Clin Genitourin Cancer. 2013;11(1):20–6.
  103. Linder S, van der Poel HG, Bergman AM, Zwart W, Prekovic S. Enzalutamide therapy for advanced prostate cancer: efficacy, resistance and beyond. Endocr Relat Cancer. 2018;26(1):R31–R52.
  104. Khalaf DJ, Annala M, Taavitsainen S, Finch DL, Oja C, Vergidis J, et al. Optimal sequencing of enzalutamide and abiraterone acetate plus prednisone in metastatic castration-resistant prostate cancer: a multicentre, randomised, open-label, phase 2, crossover trial. Lancet Oncol. 2019;20(12):1730–1739.
  105. de Wit R, de Bono J, Sternberg CN, Fizazi K, Tombal B, Wülfing C, et al. Cabazitaxel versus Abiraterone or Enzalutamide in Metastatic Prostate Cancer. N Engl J Med. 2019. doi:10.1056/nejmoa1911206.
  106. Lombard AP, Liu L, Cucchiara V, Liu C, Armstrong CM, Zhao R, et al. Intra versus Inter Cross-resistance Determines Treatment Sequence between Taxane and AR-Targeting Therapies in Advanced Prostate Cancer. Mol Cancer Ther. 2018;17(10):2197–2205.
  107. El-Amm J, Aragon-Ching JB. The Current Landscape of Treatment in Non-Metastatic Castration-Resistant Prostate Cancer. Clinical Medicine Insights: Oncology. 2019;13. doi:10.1177/1179554919833927.
  108. Hussain M, Fizazi K, Saad F, Rathenborg P, Shore N, Ferreira U, et al. Enzalutamide in men with nonmetastatic, castration-resistant prostate cancer. N Engl J Med. 2018;378(26):2465–2474.
  109. Sternberg CN, Fizazi K, Saad F, Shore ND, De Giorgi U, Penson DF, et al. Enzalutamide and Survival in Nonmetastatic, Castration-Resistant Prostate Cancer. N Engl J Med. 2020;NEJMoa2003892.
  110. Small E, Saad F, Chowdhury S, Oudard S, Hadaschik B, Graff J, et al. Final survival results from SPARTAN, a phase III study of apalutamide (APA) versus placebo (PBO) in patients (pts) with nonmetastatic castration-resistant prostate cancer (nmCRPC). ASCO Virtual Sci Program; May 29, 2020 Abstr 5516. 2020.
  111. Fizazi K, Shore ND, Tammela T, Ulys A, Vjaters E, Polyakov S, et al. Overall survival (OS) results of phase III ARAMIS study of darolutamide (DARO) added to androgen deprivation therapy (ADT) for nonmetastatic castration-resistant prostate cancer (nmCRPC). J Clin Oncol. 2020;38(15_suppl):5514–5514.
  112. Enzalutamide, Xtandi; Highlights of prescribing information (FDA). 2019 www.fda.gov/medwatch. Accessed 14 June 2020.
  113. Karantanos T, Corn PG, Thompson TC. Prostate cancer progression after androgen deprivation therapy: Mechanisms of castrate resistance and novel therapeutic approaches. Oncogene. 2013;32(49):5501–5511.
  114. Herlemann A, Washington SL, Cooperberg MR. Health Care Delivery for Metastatic Hormone-sensitive Prostate Cancer Across the Globe. European Urology Focus. 2019;5(2):155–158.
  115. Francini E, Gray KP, Xie W, Shaw GK, Valença L, Bernard B, et al. Time of metastatic disease presentation and volume of disease are prognostic for metastatic hormone sensitive prostate cancer (mHSPC). Prostate. 2018;78(12):889–895.
  116. Gravis G, Fizazi K, Joly F, Oudard S, Priou F, Esterni B, et al. Androgen-deprivation therapy alone or with docetaxel in non-castrate metastatic prostate cancer (GETUG-AFU 15): A randomised, Open-label, Phase 3 trial. Lancet Oncol. 2013;14(2):149–158.
  117. James ND, Sydes MR, Clarke NW, Mason MD, Dearnaley DP, Spears MR, 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.
  118. Fizazi K, Tran NP, Fein L, Matsubara N, Rodriguez-Antolin A, Alekseev BY, et al. Abiraterone plus prednisone in metastatic, castration-sensitive prostate cancer. N Engl J Med. 2017;377(4):352–360.
  119. James ND, de Bono JS, Spears MR, Clarke NW, Mason MD, Dearnaley DP, et al. Abiraterone for Prostate Cancer Not Previously Treated with Hormone Therapy. N Engl J Med. 2017;377(4):338–351.
  120. Kyriakopoulos CE, Chen Y-H, Carducci MA, Liu G, Jarrard DF, Hahn NM, et al. Chemohormonal Therapy in Metastatic Hormone-Sensitive Prostate Cancer: Long-Term Survival Analysis of the Randomized Phase III E3805 CHAARTED Trial. J Clin Oncol. 2018;36(11):1080–1087.
  121. Chi KN, Agarwal N, Bjartell A, Chung BH, Pereira de Santana Gomes AJ, Given R, et al. Apalutamide for Metastatic, Castration-Sensitive Prostate Cancer. N Engl J Med. 2019;381(1):13–24.
  122. Clarke NW, Ali A, Ingleby FC, Hoyle A, Amos CL, Attard G, et al. Addition of docetaxel to hormonal therapy in low- and high-burden metastatic hormone sensitive prostate cancer: long-term survival results from the STAMPEDE trial. Ann Oncol. 2019;30(12):1992–2003.