Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Dec 3.
Published in final edited form as: Nat Rev Urol. 2021 Aug 27;18(12):707–724. doi: 10.1038/s41585-021-00500-1

Proteomic discovery of non-invasive biomarkers of localized prostate cancer using mass spectrometry

Amanda Khoo 1,2,14, Lydia Y Liu 1,2,3,4,5,14, Julius O Nyalwidhe 6,7, O John Semmes 6,7, Danny Vesprini 8,9, Michelle R Downes 10, Paul C Boutros 1,3,4,5,11,12,13,, Stanley K Liu 1,8,9,, Thomas Kislinger 1,2,
PMCID: PMC8639658  NIHMSID: NIHMS1749612  PMID: 34453155

Abstract

Prostate cancer is the second most frequently diagnosed non-skin cancer in men worldwide. Patient outcomes are remarkably heterogeneous and the best existing clinical prognostic tools such as International Society of Urological Pathology Grade Group, pretreatment serum PSA concentration and T-category, do not accurately predict disease outcome for individual patients. Thus, patients newly diagnosed with prostate cancer are often overtreated or undertreated, reducing quality of life and increasing disease-specific mortality. Biomarkers that can improve the risk stratification of these patients are, therefore, urgently needed. The ideal biomarker in this setting will be non-invasive and affordable, enabling longitudinal evaluation of disease status. Prostatic secretions, urine and blood can be sources of biomarker discovery, validation and clinical implementation, and mass spectrometry can be used to detect and quantify proteins in these fluids. Protein biomarkers currently in use for diagnosis, prognosis and relapse-monitoring of localized prostate cancer in fluids remain centred around PSA and its variants, and opportunities exist for clinically validating novel and complimentary candidate protein biomarkers and deploying them into the clinic.


Prostate cancer is the second most diagnosed non-skin cancer in men worldwide1, and will account for ~26% of non-skin cancer diagnoses and ~11% of cancer-related deaths in men in the United States in 2021 (REF2). Many prostate cancers are indolent in nature, but others are aggressive and are either diagnosed when already metastasized or rapidly spread beyond the prostate, leading to morbidity and potential prostate cancer-specific mortality3. Distinguishing indolent from aggressive disease to enable personalized treatment regimens is, therefore, a key challenge in prostate cancer research4.

Screening, diagnosis and prognosis of localized prostate cancer remain heavily reliant on clinical features such as serum PSA level, clinical T category and clinical International Society of Urological Pathology Grade Group (ISUP GG) that do not have optimal validity (sensitivity and specificity) for non-indolent disease57. For example, both upgrading (~20–30%) and downgrading (~10%) could occur between needle biopsy and radical prostatectomy around the commonly used boundary between clinically insignificant (ISUP GG = 1) and clinically significant disease (ISUP GG ≥2)8,9. Thus, the best existing risk-stratification schemes, such as the National Comprehensive Cancer Network and European Association of Urology risk groups systems, still lead to overtreatment and under-treatment for individual patients10. Men with low-risk disease managed non-curatively have a 8.9% cumulative prostate cancer-specific mortality at 15 years (compared with a 49.5% risk of dying from other causes)11, whereas patients with actively managed radiorecurrent, high-risk prostate cancer experience rapid metastases and have a 30% chance of dying from their prostate cancer12. Current management pathways are in need of complementary biomarkers that accurately reflect prostate cancer aggressiveness at screening and diagnosis, and improve risk stratification13,14. Similarly, biomarkers that indicate clinically meaningful progression and can be used for frequent longitudinal monitoring are needed to support follow-up monitoring during active surveillance (AS) or after local definitive treatment (FIG. 1).

Fig. 1 |. The role of biomarkers in prostate cancer management.

Fig. 1 |

Common tests and treatment modalities in the management of patients with prostate cancer and the applicability of molecular markers at each stage. At clinical diagnosis and staging, molecular biomarkers can help to inform whether initial or repeat biopsy is required and to improve knowledge in instances in which PSA is elevated but multiparametric magnetic resonance imaging (mpMRI) and transrectal ultrasonography (TRUS)-guided biopsy are negative. During active surveillance, fluid-based biomarkers can be used more frequently than TRUS-guided biopsy and mpMRI, therefore, enabling clinicians to rapidly identify patients whose disease has progressed. Biomarkers could be used in conjunction with PSA for monitoring patients following definitive local treatment, and could direct patients with probable clinically significant biochemical recurrence (BCR) for further imaging. At the advanced-disease stage, fluid-based biomarkers can be used for predicting a patient’s response to therapy, for which no metric exists. In addition, biomarkers can be used to detect low-volume metastases and early signs of androgen deprivation therapy failure. DRE, digital rectal examination; PET, positron emission tomography.

Identifying molecular biomarkers that can complement existing clinical tools for diagnosis, prognosis and disease monitoring has been a major component of prostate cancer research since the early 2000s. Fluids that are in contact with and/or are in close proximity to the prostate — prostate-associated fluids — have always held promise as sources of molecular biomarkers. Protein abundance can be measured accurately and precisely in a targeted manner in fluids, making proteins a promising molecule for biomarker discovery in prostate-associated fluids1516. With increasing interest in personalized medicine and efforts to profile tumour proteomes through The Human Protein Atlas, The Cancer Genome Atlas and the Clinical Proteomic Tumour Analysis Consortium, great opportunities exist for prostate cancer protein-based biomarker discovery and validation17,18.

Protein abundance.

A measure of the number of proteins in a sample, estimated from direct measurements of peptide abundance.

Mass spectrometry.

(MS). An analytical technique that separates ions by mass:charge ratio in a mass analyser.

Proteomics.

The large-scale study of proteomes, which are sets of proteins produced in a biological context such as by a cell, tissue or organism.

Mass spectrometry-based proteomics can be used to detect and quantify thousands of proteins as well as their post-translational modifications19. Mass spectrometry is a biophysical technique that enables the structural analysis of various biomolecules in the form of gas phase ions, resulting in their detection and quantification. In this Review, we outline mass spectrometry (MS) technologies that enable the systematic discovery and targeted validation of protein-based biomarkers in prostate-associated fluids. We discuss clinically relevant biospecimens for protein-based biomarker discovery and highlight candidate protein-based biomarkers applicable at each step of the clinical decision-making pathway. We also discuss the challenges of clinical translation and application of these biomarkers and present potential solutions.

Need for biomarkers in disease management

Diagnostic, prognostic and predictive biomarkers enable patient-specific management of prostate cancer. Serum PSA quantification, digital rectal examination (DRE) and systematic transrectal ultrasonography (TRUS)-guided biopsies form the cornerstone of prostate cancer management13,14, but they have been increasingly supplemented by molecular assays, germline testing and multiparametric magnetic resonance imaging (mpMRI), which provide complementary information2022. However, novel biomarkers are still needed at every step of prostate cancer management to improve personalized patient care.

Some of the limitations in the clinical management pathways for prostate cancer can be attributed to the weaknesses in the key clinical assays used. For example, no single value of PSA is able to achieve simultaneously high sensitivity and specificity, as elevated PSA levels can indicate non-cancer states such as BPH or prostatitis23. Similarly, DRE has both low sensitivity and specificity (<60% for both) in the primary-care setting5. TRUS-guided biopsies are prone to undersampling, especially when tumour volume is low and prostate volume is high (with a false-negative rate of up to 30%7), and miss prostate cancer in the anterior zone, which can account for up to 20% of prostate cancers24. In addition to pain and discomfort, biopsies are also associated with a small but substantial risk of potentially life-threatening urosepsis25. Switching to a transperineal approach could reduce the risk of infection, although it is still associated with pain and discomfort26.

Many fluid-based molecular biomarkers, such as the Prostate Health Index, 4Kscore, Progensa, MIPs, SelectMDx, ExoDX Prostate, Apifiny and Proclarix, have been developed to support prediction of aggressive disease upon biopsy or repeat biopsy21. These tests are typically approved for limited use in the diagnostic setting, but no consensus exists on which biomarker should be used in which scenario27. Approved tissue-based molecular biomarkers, including ProMark, OncotypeDX, Decipher and Prolaris, have received positive recommendations for use in patients with very-low-risk or low-risk treatment-naive disease for deciding between AS or definitive therapy, but no randomized control trials have studied their utility, comparative effectiveness or economic efficiency, limiting their uptake14. Additionally, needle biopsies are needed to obtain tissue-based biomarkers, which are associated with morbidity and are limited by the relatively low volume and spatially heterogeneous disease in the prostate, resulting in potential sampling bias28.

mpMRI has emerged as a promising tool to improve detection of clinically significant tumours (negative predictive value 63–98%)29,30. When combined with systematic biopsy, mpMRI-targeted biopsy also enables diagnosis of more prostate cancer and more clinically significant disease than either method alone29,30. However, up to 35% of clinically significant tumours in any lesion remain invisible to mpMRI, including high-grade (Gleason pattern 4 and 5) tumours31,32. Sequential mpMRI in AS can be used to indicate the need for repeat biopsy or potentially replace biopsy completely, but no conclusive evidence has shown an association between mpMRI progression and pathological upgrading33,34. Furthermore, interobserver variability, reader experience, limited availability of 3-T instruments in non-academic settings, and the cost associated with mpMRI can hinder the timely and frequent adoption of mpMRI in prostate cancer management35,36.

The current goal of prostate cancer biomarker discovery studies is to develop assays that provide complementary information to existing modalities at each step of disease management. Prostate cancer is highly heterogenous between and within patients; thus, combining biomarkers with existing modalities has the most potential for achieving personalized management in diagnosis, prognosis and treatment prediction. To support prostate cancer screening and diagnosis, a biomarker needs to be inexpensive, reproducible and non-invasive to enable its longitudinal application in large populations. Importantly, a screening and diagnostic biomarker should be developed to detect only clinically significant disease when used alone or in combination with mpMRI after suspicious PSA and/or DRE analysis. The biomarker should have a high negative predictive value, so that negative results in patients with benign or clinically insignificant disease will enable them to confidently avoid biopsies. Ideally, such a diagnostic pathway would remove the need for biopsy as a diagnostic tool and reserve it for the use of tissue-based risk stratification for patients requiring active management. At the least, a diagnostic biomarker should provide information to distinguish between BPH and prostate cancer when combined with PSA, meaning that it should be most informative in the PSA range of 4–10 ng/ml, in which the outcome is most uncertain37

A diagnostic biomarker that is able to predict aggressive disease can also be used for risk stratification in men diagnosed with prostate cancer. The key biomarker of interest in this setting is one that has high sensitivity for the prediction of disease progression in order to encourage men with very-low to favourable-intermediate-risk disease and negative results to select and remain on AS to avoid overtreatment. As prostate cancer is spatially heterogeneous and influenced by microenvironmental factors such as hypoxia38,39, a biomarker that reflects the presence of aggressive disease anywhere in the prostate and indicates unfavourable tumour microenvironmental conditions (such as a protein biomarker that is elevated in patients with hypoxic tumours) could provide holistic information for prognosis. In order for the same prognostic biomarker to be used for monitoring during AS, it should be non-invasive for ease of frequent sampling. A highly standardized assay with results that can be transferred across health-care providers is also desirable for the long-term surveillance of these patients. The ideal biomarker should also indicate meaningful disease progression so that re-biopsy is only performed to confirm upgrading in these patients.

Serum PSA testing for monitoring of biochemical recurrence (BCR) remains the basis of follow-up monitoring after local treatment13,14. The ideal biomarker for monitoring disease progression after definitive local therapy shares many of the same characteristics desirable in a biomarker for monitoring patients on AS. Its primary goal would be to complement PSA in the determination of clinically relevant BCR, as the definition of BCR is based on PSA but not all BCR results in morbidity or increased mortality40. Ideally, the biomarker would also provide additional prognostic and predictive information upon disease progression. Biomarkers for monitoring after local treatment could provide a low-cost tool that is complementary to imaging-based assessments, such as mpMRI, bone scans, abdominopelvic CT and PET, and help the early detection of recurrent disease. Studies to discover such a biomarker will require multidisciplinary collaboration and the collection of a sizable matching cohort of biospecimens and imaging results from patients with extensive follow-up data.

Easily accessible fluids that can be collected using routine, standardized protocols can provide the basis of biomarker assays that need to be performed non-invasively and serially. Serum-based and urine-based biomarkers are especially attractive in the setting of prostate cancer, as they could easily be collected and assayed alongside PSA. Fluids might also be more likely to fully capture diverse information that reflects intratumoural, microenvironmental and systemic conditions than tissue biopsies, which are limited to small regions of prostate tissue. Urine is particularly appealing for its potential to be collected outside of medical facilities during times of interrupted health-care availability. Fluid-based molecular biomarkers can also be measured with highly standardizable and reproducible targeted MS assays that can be transferred between clinical laboratories41.

Fluid-based protein biomarkers are particularly attractive for several reasons. First, proteins are more stable in fluids than RNA or DNA15,16. Second, processes associated with cancer such as angiogenesis, cell adhesion and migration are mediated by proteins at the plasma membrane, which are shed into prostate-associated fluids and can be detected at high concentrations42. Third, secreted and cell-surface proteins are involved in crosstalk with the tumour microenvironment and are differentially expressed in tumour cells43,44. Fourth, fluid-based protein biomarkers can also capture the spatial heterogeneity of the prostate compared with proteins obtained from tissue biopsies, which are limited by spatial sampling. Finally, prostate cancer typically has a ‘quiet’ genome, with low rates of most types of mutations45, but a high level of subclonal diversity38,46, which considerably limits the utility of single-gene mutational biomarkers and means that biomarker panels are needed to fully and accurately capture this heterogeneity47. Secreted proteins are the cumulative result of all levels of information and regulation through the central dogma and, therefore, have the potential to capture dysregulation beyond somatic mutations. Multiple protein biomarkers can also be developed into multiplexed protein panels or combined with other molecular biomarkers from the same fluid to fully capture disease heterogeneity.

Despite the promises offered by fluid-based protein biomarkers, few studies have succeeded in identifying candidates with biomarker potential. A big challenge impeding prostate cancer candidate biomarker discovery studies from yielding meaningful outcomes is the limited information provided by the small discovery cohorts, which hinders the assessment of measurement variability and limits potential candidates to those with large effect sizes. In addition, few studies share the same biomarker candidates owing to a lack of standardized methods for sample collection, sample preparation, data acquisition and bioinformatics analysis. Many studies identify statistically significant biomarker candidates, typically by testing whether an area under the receiver operating characteristic curve (AUC) is significantly different from 0.5. However, these study premises inherently lead to the discovery of molecules that could never be used as biomarkers as they are either clinically irrelevant or aimed at being proxies for existing modalities. For example, urine biomarkers that can distinguish men with prostate cancer from those without are not clinically needed, as all men suspected to have prostate cancer would either be symptomatic or have abnormal PSA and/or DRE. Similarly, a biomarker that performs marginally better than PSA (albeit significant) for the detection of clinically significant disease has no clinical utility, as clinical paradigms could not be changed overnight and whether the biomarker could reliably compliment PSA in distinguishing clinically insignificant and significant disease is a more important question. In addition, studies that do not develop a standardizable validation assay for candidates are difficult to interpret, as how the initial discoveries are dependent on the protocol used is unclear. Finally, the lack of independent validation cohorts reduces confidence in any biomarker candidate, and validation cohorts that are underpowered and do not have relevant clinical annotations available also mean that, ultimately, assessing whether candidates add value to existing modalities is impossible. Furthermore, several key questions remain unanswered in biomarker development and validation for prostate cancer. To what degree current molecular and biomarker studies underestimate the true molecular heterogeneity of prostate as a result of insufficient or biased spatial sampling is unclear38,48,49. Additionally, how the short-term and long-term temporal dynamics of biomarkers affect their validity and clinical utility is uncertain22,50. The influence of germline polymorphisms and race on biomarkers also remains underinvestigated51,52. Easily accessible fluids in combination with standardized targeted proteomics assays have great promise for analysing the large cohorts required to answer these questions and identify biomarkers that overcome these challenges.

Proteomic technologies

MS can be used at various stages of the biomarker discovery process to identify and validate protein biomarker candidates (FIG. 2). Commonly used MS-based proteomics technologies include shotgun proteomics, native peptidomics, imaging mass cytometry (IMC) for biomarker discovery and targeted MS for biomarker validation.

Shotgun proteomics.

An untargeted workflow for identifying and quantifying proteins by mass spectrometry via proteolytic digestion of proteins into peptides.

Dynamic range.

In mass spectrometry, this term is the range of protein abundances in a sample.

Precursor ion.

Intact ions that later dissociate into smaller fragment ions.

Fragment ion.

An ion that is the product of fragmentation. For peptides, fragment ions are produced from fragmentation at the peptide backbone.

Data-dependent acquisition.

(DDA). A mass-spectrometry acquisition method in which the top N most intense peptides are selected for fragmentation.

Data-independent acquisition.

(DIA). A mass-spectrometry acquisition method in which all peptides within a given mass window (such as 15–50 m/z) are selected for fragmentation. Peptides in a selected mass range are fragmented using sequential windows.

Fig. 2 |. Biomarker discovery to translation into the clinic.

Fig. 2 |

Mass spectrometry-based assays can be used at several points in biomarker discovery and translation. Once a clinical need has been determined and clinical samples are collected or preclinical models are established, samples can be characterized by shotgun proteomics or imaging mass cytometry (IMC) at the biomarker discovery stage to identify protein biomarkers of interest. Following proteolytic digestion of proteins from the sample of interest, peptides are separated by liquid chromatography, then ionized through electrospray ionization (ESI), and analysed using mass spectrometry53. Data-dependent acquisition (DDA) or data-independent acquisition (DIA) scan modes can be used for analysis. In DDA-MS, the top N most abundant peptide precursor ions in each scan are sequentially selected and isolated within a narrow mass window (typically 1–2 m/z) for fragmentation. The resulting MS2 spectrum is matched to in silico spectra via database searching for peptide identification. In DIA-MS, all peptide precursor ions in a predefined mass window (typically 10–50 m/z) are isolated for fragmentation, yielding complex MS2 spectra. MS2 spectra are matched to peptides via deconvolution using a spectral library as a reference. IMC can also be used in the discovery stage to characterize tumour spatial heterogeneity at subcellular resolution to distinguish between cell populations of interest. Cells are labelled with metal-conjugated antibodies and laser ablation coupled to inductively coupled plasma (ICP)-MS-based detection is used to quantify reporter ion intensity in each region of the tissue. Statistical analysis is used to select candidates for validation by targeted MS, and for building multipeptide biomarker panels from targeted MS results. A robust statistical analysis workflow requires feature selection, model selection, as well as model hyper-parameter tuning and training to be performed solely on the discovery cohort, after which the model is frozen (that is, the parameters and hyperparameters can no longer be adjusted). The frozen model can then be evaluated in unseen validation cohorts to gauge its performance and generalizability. Targeted MS can be used to validate biomarker candidates in a larger cohort. Sample preparation for targeted proteomics is similar to shotgun proteomics, with the addition of stable isotope-labelled ‘heavy’ peptides synthesized for peptides that best represent the protein candidate of interest. These peptides are placed on an inclusion list, and only peptides on this list are fragmented and analysed, resulting in a lower limit of detection and higher reproducibility in quantification than shotgun proteomics. Parallel reaction monitoring (PRM) can be used to quantify all the fragment ions generated from each peptide of interest, whereas selected reaction monitoring or multiple reaction monitoring (SRM/MRM) can be used to quantify select fragment ions (typically 3) from each peptide. Once these assays are developed and standardized, they can be used as-is in future clinical trials and in the clinic.

Shotgun proteomics.

In the biomarker discovery stage, shotgun proteomics53, also called bottom-up proteomics, is often used to detect and quantify proteins from biological samples. In shotgun proteomics, a mixture of proteins are digested using proteases to obtain peptides before analysis. Trypsin and Lys-C are most commonly used for digestion because they yield peptides with positively charged C-termini that are amenable to ionization and are, therefore, detectable by MS54. Optionally, sample complexity can be further reduced by fractionating samples using off-line orthogonal methods, such as high pH-reversed phase or strong cation exchange chromatography55,56. Off-line fractionation increases proteome coverage (number of detected proteins)56,57, but it requires increased instrument time and input material (>100 μg of protein), reducing feasibility with many clinical samples. Thus, this technique is not often performed in biomarker discovery studies using clinical samples. Then, in order to increase the dynamic range of observed peptide concentrations and increase detection of low-abundance ions, peptides are separated using nanolitre flow-rate liquid chromatography (LC) columns coupled directly (termed ‘in-line’) to the mass spectrometer (called LC–MS). C18 resins that separate peptides based on hydrophobicity are most commonly used for separation, although other modes of separation, such as capillary electrophoresis (CE), which separate peptides based on their electrophoretic mobility, can also be used58. As peptides elute from the chromatography column, they are ionized by electrospray ionization, through protonation of tryptic peptides by mild acids in the mobile phases. In addition to the separation methods described above, ions can be further separated by ion mobility before mass acquisition59,60, in which peptide ions are separated by their mass-to-charge ratio (m/z) in mass analysers53.

The mass spectrometer records two independent but related spectra: the MS1 spectrum, which records the intensity and m/z of intact peptide ions (termed precursor ion), and the fragment ion mass spectrum (termed MS2 or MS/MS spectrum), which is generated through fragmentation of peptides present in the MS1 spectrum. These peptides are fragmented along the peptide backbone through collision with inert gas molecules such as helium, nitrogen or argon61, producing smaller fragment ions. The MS2 spectrum records the intensity and m/z of fragmented peptide ions. To determine the identity of a peptide, both the MS1 and matching MS2 spectra are required. The most common strategy is database searching, in which experimental fragment ion spectra are matched to theoretical spectra generated by in silico digestion of a reference proteome53. To control for the error rates associated with spectral matching in complex proteomes, experimental fragment ion spectra are also matched against ‘dummy sequences’ generated through inversion or scrambling of known protein sequences (such as the target-decoy approach)62,63. Matched spectra are carried forward for reporting and protein inference (that is, the process of matching detected peptide sequences to available protein accession, which is similar to aligning sequence reads to genomic coordinates). Relative peptide abundance can be quantified from the MS1 spectrum, usually by integrating the area under the curve to the ion abundance over elution (the retention time) of the peptide. This approach is commonly referred to as label-free quantification (LFQ), as no stable isotope standards or chemical labelling strategies are employed64. Peptide quantity can also be derived by label-based quantification using isobaric labels, such as tandem mass tags (TMT) and others (for example iTRAQ or EASI-tag). Isobaric labelling enables the multiplexed analyses of up to 16 samples with potentially increased quantitative precision compared with LFQ65,66, but it has decreased quantitative accuracy and peptide identification rates67,68.

Two independent MS scan modes are commonly used in shotgun proteomics — data-dependent acquisition (DDA) and data-independent acquisition (DIA)69,70. In DDA, the top N most abundant ions from each MS1 scan are selected for fragmentation, in which N is selected based on the instrument’s scan speed and sample complexity. Each cycle consists of one MS1 scan, followed by N fragment ion (MS2) scans. The main drawback of DDA is that selecting peptides for fragmentation is semi-stochastic; thus, lowabundance peptides might not be reproducibly detected and quantified in biological replicates. Nevertheless, protein quantification by DDA is robust47. In DIA, all peptides in successive 10–50-m/z windows are selected for fragmentation19. Owing to the higher rates of co-fragmentation than DDA, which results in mixed spectra, spectral matching to theoretical spectra is not commonly used for peptide inference. Instead, experimental data from DIA are matched to spectral libraries that consist of previously recorded MS2 spectra, usually generated from pooled or highly fractionated samples previously acquired in DDA mode. The spectral library is used to deconvolve the mixed fragment ion spectra from multiple peptides71. Although peptide detection and quantification in each DIA run is limited to peptides present in the spectral library, data acquisition is continuous and, therefore, low-abundance peptides can be quantified more reproducibly than in DDA71.

Performing shotgun proteomics on specific subsets of the proteome, such as post-translationally modified (PTM) proteins, or extracellular vesicle (EV) proteomes, can complement biomarker discovery by highlighting proteins involved in specific tumour pathways. For example, enriching the sample for EVs by ultracentrifugation or filtration can increase detection of EV proteins, which include low-abundance proteins such as signalling molecules72,73. Enriching for PTM proteins can also improve the detection of specific subsets of the proteome. The two most common PTM-enrichment workflows are phosphoproteomics and N-glycoproteomics. Enriching for phosphoproteins from cells and tissues using immobilized metal affinity chromatography or titanium dioxide magnetic beads can help to identify biomarkers associated with aberrant signalling pathways74. For example, phosphoproteomics on prostate cancer tissues from patients with metastatic castration-resistant prostate cancer (mCRPC) revealed actionable therapeutic pathways such as cell-cycle regulation, nuclear receptor, PI3K–AKT–mTOR and sternness pathways, as well as associated kinases that are targetable with currently available clinical inhibitors75. N-glycoproteomics can enrich for cell surface proteins, as the majority of proteins at the cell membrane are N-glycosylated76. Applying these subset-specific enrichment workflows to biomarker discovery is currently limited by the large amount of input material required for enrichment, as PTM-modified peptides are relatively low in abundance compared with their unmodified forms77,78. Historically, phosphoproteomics and N-glycoproteomics workflows required a minimum of 500–1,000 μg of protein as input material79,80, which might be challenging to obtain from clinical samples. Developments in sample preparation and improvements in MS technology for low-input proteomics have reduced the required input material by up to 20-fold while maintaining a similar number of detected peptides, reproducibility and signal80,81. These methods use magnetic bead capture, robotic automation or spin tips to capture peptides within a small volume, therefore, reducing non-specific sample losses while enabling the use of harsh detergents for cell lysis8083. These low-input methods are promising, but are not yet widely used in clinical proteomics owing to their novelty, and have not been applied to prostate cancer-associated fluids.

Native peptidomics.

The native peptidome, consisting of low-molecular-weight proteins and cleaved peptides, can also be analysed using LC-MS/MS or capillary electrophoresis MS (CE-MS), an analogous technique to LC-MS84. In native peptidomics, peptides are isolated from fluids by filtration through a low-molecular-weight cut-off filter, then separated by charge and size before MS.

Selected reaction monitoring/multiple reaction monitoring.

(SRM/MRM). A targeted mass spectrometry method that sequentially isolates and records pre-selected fragment ion masses from a peptide.

Triple-quadrupole mass spectrometer.

A tandem mass spectrometer consisting of two quadrupole mass analysers arranged sequentially for mass isolation with an additional quadrupole in the middle that is used for collision-induced dissociation.

Parallel reaction monitoring.

(PRM). A targeted mass spectrometry method that isolates and records all fragment ion masses from a peptide.

Orbitrap mass analyser.

An ion-trap mass analyser that detects m/z signals by oscillating ions around a cylindrical electrode with tapered ends.

Imaging mass cytometry.

An emerging technology for mapping the spatial distribution of proteins on a tissue section at single-cell resolution is IMC85. In this technique, sectioned tissues are stained with antibodies complexed to heavy metal reporter ions, and laser ablation coupled with MS-based detection is used to quantify reporter ion intensity in each region of the tissue85. In addition, images generated from IMC can be co-registered onto gross pathology, such as haematoxylin and eosin-stained tissue or other small molecule stains (for example, pimonidazole staining for hypoxia). IMC has not yet been applied to prostate tissues, as the technology is very novel, having only been applied to breast cancer tissue in 2020 (REF85). In breast cancer, this technique has been shown to be able to distinguish single tumour cells and stromal cells, and characterize tissue organization and heterogeneity85. IMC has the potential to enable the assessment of proteins specific to adverse prostate cancer histologies (such as intraductal carcinoma and Gleason pattern 4 and 5) while retaining spatial information and provide potential biomarker candidates that could be quantified in prostate-associated fluids.

Targeted proteomics.

Protein biomarker candidates in fluid can be validated using western blot and enzyme-linked immunosorbent assays (ELISAs)86. Western blot has low throughput, as each experiment is limited by the number of wells in a gel; thus, ELISA is most commonly used for protein biomarker validation owing to its higher throughput than western blot. Nevertheless, several caveats to using antibody-based assays for validation of protein biomarkers exist. First, they are limited to proteins for which high-quality antibodies are available87,88. In addition, ELISA development is expensive (costing $100,000–$1,000,000 per assay) and inefficient (1–2 years for development)89. Thus, only a small number (in the order of tens) of high-priority candidates can move on to the validation stage, limiting the number of potential candidate biomarkers that can be validated.

To overcome the limitations of using antibody-based validation of candidate biomarkers, targeted MS can be used in a high-throughput and cost-effective manner. Hundreds of candidates can be multiplexed and evaluated in a single analysis90 at a cost of <$5,000 and ~6 months for assay development89,9193. Targeted MS assays, such as selected reaction monitoring/multiple reaction monitoring (SRM/MRM) for triple-quadrupole mass spectrometers, and parallel reaction monitoring (PRM) for orbitrap mass analysers, quantify fragment ions of selected proteotypic peptides from each candidate protein. A proteotypic peptide is a peptide that is unique to a protein or protein isoform and produces a good MS response94. Proteotypic peptides that best represent proteins of interest from the discovery stage are selected for measurement. To ensure that quantification is reproducible across different laboratories, stable isotope-labelled peptides at known concentrations are synthesized for every candidate peptide and used as spike-in standards. When an assay has been validated on an instrument, it can be transferred across sites and clinical laboratories94.

In summary, shotgun proteomics using LC-MS/MS is most suited to protein biomarker discovery in medium-complexity to high-complexity samples such as tissues, cell lysates and patient fluids. It provides a broad overview of protein abundance in a biological sample for comparisons between different clinical groups. As no standard of known quantity is spiked into each sample, quantification is relative53. Of the acquisition methods used for shotgun proteomics, DDA is the best established53, and DDA acquisition can be multiplexed by labelling samples with isobaric labels65,66. DIA is gaining in popularity as it enables continuous data acquisition for improved scalability and robust quantification19. However, like DDA, DIA is most suited to the biomarker discovery stage, as quantification is relative19. Similar to LC-MS/MS, CE-MS is also most suited to the biomarker discovery stage, such as for native peptidomics, especially for studying PTM-modified peptides like phosphopeptide positional isomers that cannot be separated by LC-MS95, as well as for profiling samples with low protein input96. However, as CE-MS instruments are not as readily available as LC-MS instruments, its potential for identifying peptides in fluids has not been fully explored.

When candidate peptides have been selected from the discovery stage, targeted MS proteomics assays (such as SRM or PRM) are most suited to validating targets, as they have lower limits of detection and better quantitative accuracy than shotgun proteomics methods when used in combination with stable isotope-labelled synthetic peptides at known concentrations97. For clinical implementation, once a clinical laboratory has a LC-MS/MS system with SRM or PRM functionality, targeted proteomics assays can be easily implemented and transferred.

For both shotgun and targeted proteomics, spatial molecular information is lost. IMC can complement shotgun proteomics analyses and provide information about the spatial organization of proteins and protein post-translational modifications within the tissue, as well as reveal protein heterogeneity within tumour regions. This tool is best used to study the tumour microenvironment and the immune landscape of the tumour85. Although these complementary MS technologies provide a basis for improved protein biomarker discovery and validation, careful sample selection is still required to improve protein detection.

Sample selection for MS

Technological advances in MS instrumentation and sample preparation mean that 2,100 proteins can now be detected in 100 μl of urine98 and 450 proteins in 10 μl of platelet-free plasma in a single MS run99. This ability presents an opportunity to discover and validate protein biomarkers in various sample types (FIG. 3) and patient cohorts, enabling improved analysis of intertumoural and intratumoural heterogeneity47. One challenge of shotgun proteomics is that the dynamic range of peptide detection in a single scan is limited to approximately four orders of magnitude100. For example, for matrices that contain proteins at vastly different concentrations, a deep proteome might be difficult to obtain, as high-abundance proteins are sampled more frequently than low-abundance proteins101. High-abundance ions that co-elute with low-abundance ions can also reduce the ionization efficiency (that is, reduce the signal:noise ratio) of low-abundance ions, termed ion suppression102. Because proteins that are leaked or secreted by tissue and/or are involved in signalling are present at low concentrations in fluids in general101, careful sample selection is required to help to increase the detection of prostate cancer-derived proteins (TABLE 1).

Fig. 3 |. Clinically relevant fluids in prostate cancer.

Fig. 3 |

a | Prostatic fluid can be obtained through vigorous prostate massage, termed expressed prostatic secretions (EPS). EPS can be collected after digital rectal examination (DRE) in voided first-catch urine, termed post-DRE urine. These prostatic fluids are the most interesting for biomarker discovery as they contain cells from the prostate as well as secreted proteins and extracellular vesicle (EV) proteins released from both nonmalignant and tumour cells. As the prostate is well vascularized, secreted proteins can also be found in blood. b | EVs are formed either by direct budding from the cell membrane or by fusion of intraluminal vesicles containing exosomes with the cell membrane. EVs also have a lipid bilayer that protects proteins from degradation by extracellular proteases. Secreted proteins can also be detected in fluids.

Table 1 |.

Clinically relevant fluid biospecimens for biomarker discovery

Sample Advantages Disadvantages Ideal use
Blood Non-invasive collection; contains proteins secreted and shed from prostate epithelial cells, and lysed cells High dynamic range, detection of low-abundance proteins is challenging; prostate-derived proteins are diluted Detection; longitudinal monitoring (AS, treatment response)
EPS Highly enriched in prostate-derived proteins Collection is challenging; not routinely collected in the clinic Biomarker discovery
Post-DRE urine Enriched in prostate-derived molecules; non-invasive collection; increased peptide stability compared with blood Contains non-prostate-derived proteins (from the kidneys and the bladder); protein concentration varies between individuals Detection; longitudinal monitoring (AS, treatment response)
Extracellular vesicles Found in serum, post-DRE urine, prostatic secretions; deep coverage of the prostate proteome; enables multi-omics analysis Isolation is challenging, no gold-standard method exists, tedious, and requires large volumes of fluid Biomarker discovery

AS, active surveillance; DRE, digital rectal examination; EPS, expressed prostatic secretions.

Blood.

Owing to its ease of collection, blood is applicable to every stage of prostate cancer clinical management and is ideal for longitudinal monitoring. Analysis of blood is able to provide information about systemic disease; thus, this fluid is especially suitable for relapse monitoring after definitive treatment. Blood is best suited to validating potential biomarkers using targeted MS assays that have a low limit of detection, but is less suited to MS-based biomarker discovery as the dynamic range of blood proteins spans 10 orders of magnitude (from 5 pg/ml to 50 mg/ml)101. In addition, tumour-derived proteins are present at lower concentrations in the circulation than in fluids proximal to the prostate103 and are, therefore, difficult to detect using shotgun proteomics. Immunodepletion can be used to remove high-abundance proteins before MS analysis to reduce the range of protein concentrations104. However, some caveats of immunodepletion include potential non-specific binding of non-target and carrier proteins, varying efficiencies of depletion for high-abundance proteins (ranging from 30% to 99%) and reduced throughput of sample preparation104.

Prostatic fluid.

Prostatic fluid is an attractive source for biomarker discovery as most of its proteome is prostate-derived and largely devoid of proteins from the seminal vesicles and testes105. Prostatic fluid is protein rich, containing proteins that are secreted or shed by epithelial cells and released into the lumen of the prostatic ducts, which empty into the urethra105. The abundance of prostate-enriched proteins in prostatic fluid differs between disease states, for example, PSA, PAP and TGM4 are differentially expressed in the prostatic fluid of patients with organ-confined disease compared with those with extracapsular disease106. Prostatic fluid can be collected by ejaculation as a component of seminal plasma107 or expressed directly from the prostate following prostate massage and collected through the urethra, termed expressed prostatic secretions (EPS)108. EPS is not routinely collected in the clinic owing to patient discomfort109, but can be collected before radical prostatectomy while the patient is under anaesthesia108. This setting for sample collection is ideal as it causes no additional discomfort to the patient, and presurgical and postsurgical clinical information (such as ISUP GG) are available to guide biomarker discovery for future validation in fluids that are more accessible than EPS.

Urine.

Urine can be collected routinely in the clinic and used in both diagnostic and prognostic settings. This fluid is ideal for longitudinal monitoring during AS, and can be applied to longitudinal post-treatment monitoring of patients who are undergoing radiotherapy. Urine-derived peptides are more stable than those from blood, as most protein degradation by proteases would have been completed in the bladder before collection16. The primary limitation of using urine for biomarker discovery as well as clinical assays is the variability of the urine proteome, which can be affected by hydration status110. Various different methods for normalizing protein abundance in urine have been suggested to account for hydration status, time of collection, and other conditions that can affect protein abundance, such as proteinuria. These methods include normalizing by fluid volume, total protein concentration, peptide concentration, urine osmolarity and urinary creatinine levels111. However, no consensus exists on the best normalization method. By contrast, the proteome of post-DRE urine, defined as first-catch urine following prostatic massage via DRE, is more stable than neat urine (urine collected without a DRE) as it contains prostate-derived proteins from EPS along with the kidney-derived and bladder-derived proteins generally found in urine112,113. This increased stability is evident in the increased abundance of prostate-derived proteins and RNAs (such as TGM4, KLK3 and HOXB13) in post-DRE urine compared with neat urine112,113 and, therefore, post-DRE urine can be considered a more accessible form of EPS than neat EPS. As post-DRE urine contains many prostate-derived proteins and can be easily collected in the clinic, it is an ideal fluid for biomarker discovery and for the validation of candidates previously discovered in EPS or tissue114. Post-DRE urine is already in clinical use for the detection of the FDA-approved biomarker PCA3, a long non-coding RNA115.

Extracellular vesicles.

EVs are membrane-bound particles containing proteins, nucleic acids and lipids, which are shed from most cell types via various mechanisms116, and can be readily found in bodily fluids such as blood and urine116. EVs are released into the extracellular space either by direct budding from the cell membrane or by fusion of intraluminal vesicles containing exosomes with the cell membrane116. EVs also have a lipid bilayer that protects proteins from degradation by extracellular proteases116. In cancer, host cells communicate with neighbouring cells by releasing EVs to promote inflammation, cell migration and proliferation117. EVs are well-suited to biomarker discovery as they can be readily obtained from fluids; thus, enriching for EVs by ultracentrifugation, filtration or size-exclusion chromatography in prostatic secretions and urine might improve the detection of secreted prostate-derived proteins by reducing the amounts of high abundance kidney-derived or blood-derived soluble proteins present in the fluid72,113. The most abundant proteins in the urine-derived EV proteome of patients with prostate cancer and benign prostatic conditions consist of prostate-enriched proteins (for example, PAP, PSA, PSMA and TGM4) that are derived from the prostate and EV-enriched proteins (for example, CD9, CD63 and CD81) that are common in EVs in general72,118. Kidney-derived proteins (such as uromodulin) and blood-derived proteins (such as albumin and apolipoproteins) remain highly abundant in urine EVs, but their concentration is reduced compared with urine itself72,118. A considerable challenge in using EVs in the clinic is the lack of a standardized protocol for EV isolation. Ultracentrifugation is the most commonly used method for isolating EVs, but the use of different centrifugation speeds, rotors, centrifugation time and chemical treatment can make comparison of results challenging119. The feasibility of isolating EVs via ultracentrifugation from large clinical cohorts is an additional challenge of EV-based biomarker discovery.

The unique properties and caveats of each sample type should be carefully considered for each stage of biomarker discovery and validation. Blood and urine are most suited for longitudinal monitoring owing to their ease of collection, but are not the most ideal for biomarker discovery because of their high dynamic range that reduces the depth of protein coverage101. Conversely, prostatic fluid and EVs are highly enriched in prostate-derived proteins and are, therefore, most suited to biomarker discovery in which a deep proteome is needed, but less suited to validation owing to the challenges in sample collection and processing109.

Non-invasive protein biomarker candidates

Novel biomarker candidates are emerging from studies that have employed proteomics techniques on prostate cancer tissue and relevant fluids. Over the past 10 years, studies that have overcome challenges in non-invasive protein biomarker discovery have been able to identify and validate biomarker candidates (TABLE 2). These biomarkers readily lend themselves to targeted assay validation and have the potential to be translated into the clinic to improve prostate cancer diagnosis, prognosis and prediction algorithms.

Table 2 |.

Promising prostate cancer biomarkers identified using mass spectrometry

Biospecimen Biomarker Assay Validation Status Ref.
Diagnosis and decision to biopsy
Post-DRE urine EVs TGM4 TMA, IHC Prostate cancer (n = 136) versus tumour-adjacent nonmalignant tissue (n = 98): AUC = 0.81, 95% CI = 0.74–0.88, P < 0.001 Needs independent validation in post-DRE urine 122
Urine β2M, PGA3, MUC3 protein abundance, tPSA Western blot Prostate cancer (n = 90) versus BPH (n = 83): AUC = 0.812, 95% CI = 0.740–0.885, P < 0.001 Needs further validation 123
Serum Proclarix (Proteomedix): CTSD, THSB, %fPSA, tPSA, age ELISA Patients within the PSA ‘grey zone’ (tPSA 2–10 ng/ml, normal DRE, prostate volume ≥35 ml by TRUS); ISUP GG 1 versus ISUP GG ≥2: sensitivity 90%, specificity 43% (95% CI = 39–46%, P < 0.001, nno PCa = 546, nISUP GG 1 = 239, nISUP GG ≥2 = 170) CE-marked in vitro diagnostic (Europe) 127,128,181183
Post-DRE urine Peptides from the proteins IDHC, SERA, IGJ, EF2 and KCRB SRM-MS BPH (n = 48) versus prostate cancer (n = 90): AUC = 0.77, 95% CI = 0.68–0.87, P < 0.05 Needs further validation in larger cohort 114
Prognosis and active surveillance monitoring
Post-DRE urine EVs FABP5 SRM-MS ISUP GG 1 (n = 5) versus ISUP GG ≥2 (n = 13): AUC = 0.86, 95% CI = 0.71–1.00, P = 0.002 Outperforms tPSA but needs validation in larger post-DRE urine cohort 130
Post-DRE urine Peptides from the proteins 6PGL, SERA, GELS, PEDF, PARK7, 1433 S and RINI SRM-MS Extraprostatic (n = 29) versus organ-confined disease (n = 61): AUC = 0.74, 95% CI = 0.62–0.85, P < 0.05 Needs further validation in larger post-DRE cohort with clinical follow-up data 114
Post-DRE urine EVs TGM4 TMA, IHC ISUP GG 1–2 (n = 50) versus ISUP GG ≥ 3 (n = 86) disease: AUC = 0.82, 95% CI = 0.69–0.91, P < 0.001
BCR (n = 64) versus no BCR (n = 72): AUC = 0.80, 95% CI = 0.69–0.91, P < 0.001
Needs further validation in post-DRE urine with clinical follow-up data 122

%fPSA, percentage free-to-total serum PSA as measured by ELISA; AUC, area under the ROC (receiver operating characteristic) curve; BCR, biochemical recurrence, BPH, benign prostatic hyperplasia; CI, confidence interval; DRE, digital rectal examination; ELISA, enzyme-linked immunosorbent assay; IHC, immunohistochemistry; ISUP GG, International Society of Urological Pathology Grade Group; MS, mass spectrometry; OR, odds ratio; SRM-MS, selected reaction monitoring mass spectrometry; TMA, tissue microarray; tPSA: total serum PSA as measured by ELISA; TRUS, transrectal ultrasonongraphy.

Diagnosis and decision to biopsy.

Protein-based diagnostic biomarkers for prostate cancer currently only include the classic prostate cancer biomarkers: immunoassay-based quantification of serum level total PSA, free PSA and [−2]pro-PSA120. Many studies on prostate cancer biomarkers focus on identifying proteins and protein panels that can distinguish prostate cancer from benign conditions (such as BPH) in liquid biopsy assays, with the aim of sparing patients with BPH from biopsy sampling and overdiagnosis.

A targeted approach is sometimes employed in diagnostic biomarker discovery studies when analysing fluid samples to focus only on proteins that are known to have elevated abundance in prostate tissues. TGM4 is a protein that is highly enriched in prostate tissues compared with non-prostate tissues, not predicted to be actively secreted to the blood121, but can be detected in neat urine, post-DRE urine and EVs isolated from urine112,122. In one study investigating post-DRE urine EVs using SRM-MS, TGM4 was found to be elevated in benign disease (BPH and high-grade prostatic intraepithelial neoplasia (HGPIN)) compared with prostate cancer (prostate cancer:benign ratio = 0.60, benign n = 54, prostate cancer n = 53)122. In the same study, the diagnostic value of TGM4 was validated in an independent tissue microarray cohort including 98 tumour-adjacent nonmalignant and 136 prostate cancer samples (from a total of 165 patients) using IHC. Consistent with what was observed in urine EVs, TGM4 was shown to have elevated abundance in adjacent nonmalignant tissues compared with prostate cancer tissues122. The study results further demonstrated that TGM4 abundance in prostate cancer tissue was higher in ISUP GG 1–2 (n = 50) prostate cancer than in ISUP GG 3–5 prostate cancer (n = 86) and the IHC score achieved an AUC of 0.82 (95% CI = 0.71–0.92, P < 0.001) for distinguishing between these two groups. TGM4 was also prognostic for BCR in this tissue microarray cohort as its abundance in prostate cancer tissue was higher in patients without BCR compared with those with BCR (nBCR= 64, nno BCR = 72; AUC = 0.80, 95% CI = 0.69–0.91, P < 0.001). The validation of TGM4 in tissue cannot demonstrate its potential as a fluid biomarker and the ideal validation of TGM4 could have been carried out in post-DRE urine using targeted MS, but this study design should be a model for future projects in which maximization of the applicability of potential biomarkers in the clinic is desired. By using clinical cohorts that enable the characterization of both diagnostic and prognostic abilities of biomarker candidates, studies could be steps ahead in narrowing biomarker candidates for validation, as only diagnostic biomarkers that distinguish clinically significant disease are of interest in the clinic.

The number of proteins that are highly enriched in prostate tissues compared with non-prostate tissues is limited; thus, studies commonly characterize prostate-associated fluid proteomes using shotgun proteomics. This study design enables identification of proteins that can be detected in fluids and are differentially expressed in patients with benign prostatic conditions compared with those with prostate cancer. In one study, DDA-MS was used to characterize neat urine, and three proteins (p2M, PGA3 and MUC3) were identified as potentially able to distinguish prostate cancer from BPH when used individually or in combination123. The discovery cohort with proteomics characterization is of very limited size (nBpH = 4 nprostate cancer = 4) but the three proteins were validated in a larger, non-independent cohort of 173 neat urine samples (83 patients with BPH and 90 patients with prostate cancer) by western blot. The combined three-protein biomarker achieved AUC of 0.71 (95% CI = 0.63–0.79, P < 0.001), which did not outperform PSA in the same cohort (AUC = 0.73, 95% CI = 0.65–0.81, P < 0.001). However, when the three-protein biomarker was applied in combination with PSA categories (0–4, 4.1–10 and >10 ng/ml), the multimodal biomarker performed significantly better than any alone (AUC = 0.81, 95% CI = 0.74–0.89, P < 0.001). Ideally, the proteins would have been validated using a more quantitative and reproducible assay than western blot, such as ELISA or targeted MS, but the results of this study demonstrate the possibility of finding biomarkers that are complementary to PSA.

The potential of a multimodal combination biomarker discovery approach has also been demonstrated in a study comparing serum and post-DRE urine from patients with BPH, HGPIN, localized prostate cancer or metastatic prostate cancer using iTRAQ-labelled DDA-MS124. Of the 193 patients included in the study as part of a consecutive series, 166 had blood and post-DRE urine collected, of which 10 patients from each group were randomly selected as the discovery cohort. After detection and quantification, protein biomarker candidates that were significantly differentially expressed between the prostate cancer groups and the benign disease groups were further selected based on criteria including potential oncogenic activity, poor characterization in the literature, whether the protein is secreted and antibody availability. Two serum proteins (PF4V1 and TAGLN2) and one post-DRE urine protein (CRISP3) were selected for validation using ELISA for serum samples and western blot for post-DRE urine samples from 126 patients from the rest of the cohort (38 BPH, 22 HGPIN and 66 prostate cancer). TAGLN2 did not show differences in abundance between the patient groups in the validation cohort, but serum PF4V1 (AUC = 0.80, 95% CI = 0.71–0.88, P < 0.05) and post-DRE urine CRISP3 (AUC = 0.87, 95% CI = 0.80–0.94, P < 0.05) were both validated as independent diagnostic biomarkers distinguishing BPH from prostate cancer (local and metastatic combined), as well as potential biomarkers distinguishing HGPIN from prostate cancer (AUCPF4V1 = 0.756, ACUCRISP3 = 0.912, confidence interval and P value not provided). Serum PF4V1 abundance was higher in patients with BPH or HGPIN than in those with prostate cancer and was not significantly different within the benign and prostate cancer groups. Urine CRISP3 showed the opposite trend, in which its abundance was higher in the prostate cancer groups than in the benign groups (with no difference within groups). The multimodal biomarker combining serum PF4V1, urine CRISP3 and PSA was significantly superior to any one alone for distinguishing BPH from prostate cancer (AUC = 0.94, 95% CI = 0.90–0.98, P< 0.05), although results on the combination of only PF4V1 and CRISP3 were not reported. CRISP3 is an interesting protein in the context of prostate cancer because high CRISP3 protein abundance in tissue is associated with advanced tumour stage, high ISUP GG, positive surgical margins, and early BCR125. In addition, CRISP3 expression was increased in CRPC compared with benign prostate tissue, and high levels of CRISP3 is more common in prostate tumours that are ERG-positive and have PTEN loss compared with those that do not126. However, PF4V1 has only been reported as a tumour suppressor and was not previously associated with prostate cancer124.

Glycoproteomics.

The large-scale study of the glycoproteome, the set of glycosylated proteins, by selective enrichment of N-glycosylated or O-glycosylated peptides.

The results of these studies have shown biomarker candidates in various fluids that can distinguish prostate cancer from benign prostatic disease, profiled using targeted or shotgun proteomics and validated using western blot or ELISA. However, they fail to address how the biomarker performs in asymptomatic men with elevated PSA (4–10 ng/ml) and negative DRE, for whom the dilemma of whether to biopsy is the greatest. Quantification of glycoproteins THBS1 and CTSD using ELISA in serum combined with %fPSA has been shown in a cohort of men with PSA levels of 2–10 ng/ml to discriminate between patients with a positive biopsy and those with a negative biopsy (AUC = 0.86 95% CI = 0.82–0.91, P < 0.001)127. The glycoproteins THBS1 and CTSD were previously identified as potential biomarker candidates for prostate cancer diagnosis using DDA-MS glycoproteomics characterization of PTEN-null mouse model tumour and serum128. The three biomarkers, THBS1, CTSD and %fPSA, were combined into a single multimodal biomarker using multivariable logistic regression, and trained on a training set of 237 patients, 130 of whom were diagnosed with prostate cancer on biopsy. The multimodal biomarker outperformed %fPSA alone (AUC = 0.64, 95% CI = 0.57–0.71, P < 0.001) in the validation set of 237 men, 108 of whom had a biopsy that was positive for prostate cancer, and had 62% specificity for prostate cancer at 90% sensitivity compared with a specificity of 23% at 90% sensitivity for %fPSA alone. Furthermore, the three biomarkers were also used to develop multimodal biomarkers to distinguish between insignificant (ISUP GG 1) from clinically significant (ISUP GG ≥2) prostate cancer (clinical ISUP GG: AUC = 0.73, 95% CI 0.65–0.81, nISUP GG 1 = 54, nISUP GG ≥ 2 = 54; pathological ISUP GG: AUC = 0.83, 95% CI 0.77–0.88, nISUP GG 1 = 22, nISUP GG ≥ 2 = 86). The strength of this study was that the patient cohort was restricted to men with total serum PSA levels between 2 and 10 ng/ml, negative DRE, and an enlarged prostate (determined using TRUS). These results have led to the successful translation of this biomarker, named Proclarix (Proteomedix), which is now CE marked127. A study to determine if this biomarker can identify men who can safely avoid an upfront mpMRI, or men who can avoid biopsy when mpMRI is indeterminate, is also ongoing128.

In summary, the most promising biomarkers for prostate cancer diagnosis and decision to biopsy will emerge from studies that consider their performance in combination with PSA and actively characterize their ability to detect clinically significant disease. A multimodal biomarker discovery approach also has great potential to deliver a biomarker with the desired clinical specificity and sensitivity, but needs to be accompanied by sizable validation cohorts to prove their generalizability.

Prognosis of localized prostate cancer.

A variety of MS-based approaches have been used in biomarker discovery studies for localized prostate cancer prognosis and risk stratification. These studies typically use risk groups or clinical ISUP GG as a proxy for non-aggressive and aggressive disease, in which clinical ISUP GG 2 or more indicates clinically significant disease that is likely to be aggressive.

An example of a biomarker discovery workflow in which biospecimens were profiled using shotgun proteomics and selected candidates were validated using targeted proteomics in independent cohorts is a study in which FABP5 in post-DRE urine-derived EVs was discovered and validated129. In this study, FABP5 was found to be elevated in patients with intermediate-grade or high-grade prostate cancer (ISUP GG ≥2) compared with those with low-grade (ISUP GG 1) disease (P < 0.001) and predicted the presence of ISUP GG ≥2 tumours in treatment-naive patients (AUC = 0.86, 95% CI = 0.71–1.00, P = 0.002), outperforming serum total PSA (AUC = 0.511, 95% CI = 0.28–0.76, P = 0.87)130. In addition, FABP5 was enriched in urine-derived EVs from patients with prostate cancer compared with those with a negative biopsy. However, the discovery and validation cohort sample sizes are small, as only 18 samples were used for discovery and 29 for validation (11 healthy participants, 5 men with ISUP GG 1 disease, 7 ISUP men with GG 2–3 disease, and 6 men with ISUP GG 4–5 disease). Further validation in a larger independent cohort of patients similarly spanning the entire risk spectrum of prostate cancer is required. As EV-based protein biomarkers are challenging to apply routinely in the clinic owing to the low-throughput EV isolation workflows131, validation of FABP5 in post-DRE urine would be ideal, as FABP5 can also be detected in post-DRE urine112.

In one study, DDA-MS was used to analyse EPS for biomarker discovery and candidate biomarkers were then validated in post-DRE urine by SRM-MS106, making use of the strengths of both fluids — EPS as a rich source of prostate-derived secreted proteins, and urine as an easily accessible fluid suitable for clinical implementation112. Overall, 133 proteins in EPS were identified that were differentially abundant in extraprostatic (stage pT3) and organ-confined (stage ≤pT2) disease, from which 147 peptides that are unique to and represent the proteins of interest were suitable for verification by targeted MS (SRM) in post-DRE urine114. The performance of these SRM assays was evaluated in two independent sets of post-DRE urine114. Individually, 24 diagnostic peptides could distinguish between cancer and non-cancer groups (P < 0.05) and 14 prognostic peptides could distinguish between extracapsular and organ-confined groups (P < 0.10)114. Peptide triaging combined with machine learning then enabled the development of multi-peptide biomarker panels for prognosis (to distinguish organ-confined from extraprostatic disease) and also diagnosis114. A biomarker panel, consisting of peptides from the proteins 6PGL, SERA, GELS, PEDF, PARK7, 1433 S and RINI, was able to distinguish between extraprostatic (n = 29) and organ-confined (n = 61) disease (AUC = 0.74, 95% CI = 0.62–0.85, P < 0.05), substantially outperforming serum PSA (AUC 0.66, confidence interval and P value not shown). A second multipeptide biomarker panel, consisting of peptides from the proteins IDHC, SERA, IGJ, EF2 and KCRB, could distinguish men with prostate cancer (n = 90) from healthy individuals (n = 48) (AUC = 0.77, 95% CI = 0.68–0.87, P< 0.05), also outperforming serum PSA (AUC 0.67). These results demonstrate the potential of combining computationally guided proteomics with clinically annotated patient cohorts for the discovery of non-invasive biomarkers.

MS-based proteomics can be used to detect differences in the fluid proteome of patients with localized disease. However, most studies characterize small patient cohorts at the discovery stage (n < 20), limiting biomarker discovery to large protein changes, such as those between low-risk and intermediate-risk or high-risk groups130, or between organ-confined and extraprostatic disease106. Ideally, clinical follow-up monitoring of these cohorts should be extended to observe BCR after definitive treatment or even prostate cancer-specific survival, so that biomarker candidates can be selected based on an end point instead of patient risk group at diagnosis.

Monitoring and prediction after definitive treatment.

Few studies have identified biomarkers for the clinical management of localized prostate cancer after therapy. The lack of studies is probably caused by difficulties in establishing cohorts with a sufficient number of patients. Thus, biomarker discovery studies in this area are particularly limited by sample size and a lack of validation cohorts, and a range of clinical questions using diverse patient groups are also asked.

In one study, the serum of patients with prostate cancer with or without bone metastases (all patients were androgen deprivation therapy (ADT)-naive) was profiled using DDA-MS with iTRAQ-labelled samples with the aim of finding upregulated proteins in bone-metastatic prostate cancer132. The discovery cohort included 30 patients with bone metastases and 30 patients without, and each group was pooled for shotgun proteomics. In an independent cohort of patients with metastatic prostate cancer, non-metastatic prostate cancer or BPH (n = 50 each), CD59 was shown to be significantly elevated in the serum of bone metastases patients using ELISA (meanbone metastasis = 58 ± 17 ng/ml meannon-metastatic = 29 ± 6 ng/ml meanBPH = 16 ± 5 ng/ml P < 0.05). The biomarker potential of CD59 was not directly assessed, but a future study could validate this protein along with other candidates for the prediction of metastases after definitive treatment or early detection of bone metastasis during post-treatment monitoring. The best cohort of patients for this validation would be those with high-risk localized prostate cancer, ideally with serial blood collection after definitive treatment and long-term follow-up data.

In one study, 15 paired serum proteomes were collected from patients with hormone-sensitive prostate cancer before and 4 months after ADT initiation, and, in addition, serum proteomes from patients with early (<2 years) or late (>3 years) ADT failure (n = 10 in each arm)133. From DDA-MS with iTRAQ-labelled samples, 47 proteins were identified on the basis of both differential abundance in 4-month post-ADT-initiation sera compared with ADT-failure sera and differential abundance between early and late ADT-failure sera (both at false discovery rate <0.2). Similarly, in another study, serum EVs from 36 patients with metastatic prostate cancer (untreated men n = 8, men with ADT-sensitive disease n = 8 and men with mCRPC n = 20) were specifically profiled using TMT-labelled DDA-MS to find potential novel therapeutic targets in mCRPC134. Levels of ACTN4 were found to be elevated in serum EVs from patients with mCRPC compared with those from patients with hormone-sensitive disease (fold change = 1.4, false discovery rate <0.01). These proteins are interesting, but independent validation was not undertaken for the detected proteins in either study. In addition, whether any of the candidates could be biomarkers that predict future early versus late castration resistance before or shortly after the initiation of ADT is a crucial clinical question. Answering this question would instead require a cohort of patients treated with ADT with at least 3 years of follow-up data, with serum collected before and ideally at multiple time points during therapy to see whether there are proteins that could indicate resistance.

Biomarker discovery studies for monitoring and prediction after definitive treatment are scarce despite the great clinical need in this area. More clinical cohorts and clinically relevant models that enable biomarker discovery in this area are urgently needed, and biomarker studies should continue to monitor their prospective patient populations so that fluid proteomes can be reanalysed in the future when outcome data are available.

Challenges in biomarker translation

The crucial challenge in translating fluid-based biomarkers into clinical use is the lack of large retrospective patient cohorts with biobanked fluids and matched clinical data (such as clinical and/or pathological ISUP GG, serum PSA level and mpMRI). Most biomarker discovery and validation have occurred in small cohorts, especially for non-routine fluids such as EPS and post-DRE urine. Most patient cohorts with fluid sample collection are prospective and lack long-term follow-up data to explore biomarker candidate associations with disease relapse after local therapy in time-to-event analyses. Thus, biomarker identification strategies are limited to approximating strong predictors of outcome, such as ISUP GG and clinical risk group, rather than directly identifying independent predictors of disease metastasis and patient survival. As a result, many clinical questions remain unanswered owing to the lack of fluid samples from appropriate patient cohorts. For example, to determine which men would most benefit from needle biopsy, control samples from men that have elevated PSA and negative DRE but are confirmed to not have prostate cancer by needle biopsy or long-term follow-up monitoring are needed. Matched mpMRI data for these men are needed to account for the role that mpMRI has in diagnosis and to facilitate discovery of biomarkers that predict which men would most benefit from biopsy following a negative mpMRI result. Similarly, for predicting which men would most benefit from AS, cohorts with upgrading data are needed, especially those that employ new clinical protocols incorporating mpMRI and molecular biomarkers in addition to standard-of-care pathways.

Patient-derived xenografts.

(PDX). Tumours grown from the implantation of a patient’s tumour cells into immunodeficient or immunocompromised mice.

A substantial technical challenge for clinical translation of protein biomarkers is the time and cost required to develop targeted MS assays for multipeptide biomarkers. For targeted MS assays, stable isotope-labelled standards (synthetic peptides carrying a ‘heavy’ stable isotope) can be used to systematically identify the best responding peptides and to generate analytical figures of merit, such as linearity, lower limit of detection and lower limit of quantification135. The sequence composition, solubility and length of the peptide, as well as the complexity of the biological sample matrix, can all affect quantification accuracy; thus, rigorous assessment of each peptide during assay development is required in order to select the best performing peptides135. Another technical challenge involves reducing sample loss in the sample preparation stage. Using robotic liquid handlers and miniaturizing reaction vessels might improve the peptide yield from low-input samples that are typically available in the clinical setting82,136. Optimizing chromatographic performance by using etched array columns with lower flow rates can also reduce the amount of input material needed for shotgun proteomics analysis by 100-fold137.

Beyond technical challenges in protein biomarker discovery and validation, the regulatory challenges in biomarker translation are substantial. Assay development and trial design need to meet the requirements for regulatory approval processes120,138,139. After regulatory clearance, the clinical uptake of biomarkers is also not guaranteed, especially in prostate cancer for which multiple commercially available molecular biomarkers target the same patient population and report outcomes in overlapping end points. Differing recommendations between authoritative bodies and the lack of a single go-to option suggest that comparative studies are needed to determine the optimal application of molecular assays and how they should be combined with mpMRI and mpMRI-targeted biopsies in diagnosis and monitoring settings14,27. Test availability, insurance coverage, guidelines for clinicians and education of the public are all factors that require additional investigation to guide strategies to improve biomarker uptake27.

Future directions

Currently, immunoassays, usually in the form of immunohistochemistry panels, are the prevailing technology for protein biomarker assessment in the clinic given their low cost (approximately US $15–20 per marker)140 and easy implementation in clinical laboratories. When considering biomarker candidates, they need to be amenable to translation into an assay format that enables ease of clinical implementation. In the future, targeted MS could be used as a platform for validating biomarker panels of up to 300 candidates in one analysis and transferred across laboratories. As contemporary pathology reports now contain prognostic and predictive factors in addition to diagnostic information, incorporation of novel proteomic biomarkers would provide complementary tools for patient management algorithms.

When patient fluids are not readily available, patient tissue and preclinical model systems such as prostate cancer cell lines, patient-derived xenografts (PDXs), organoids and transgenic or genetically engineered mouse models are an important resource for the identification and validation of new biomarker candidates141143. Tissue is the biospecimen that is most frequently used for biomarker discovery, disease classification and disease staging as it closely reflects tumour biology and structural changes can be readily visualized using histopathology. Tissue proteomic profiles can be linked to histopathological and imaging features, and other -omics data, such as genomic, transcriptomic, epigenomic and metabolomic information, to improve study of tumour heterogeneity and prioritize biologically relevant pathways or proteins38,47,144,145. Proteins detected in prostate tissues are also more likely to be related to prostate biology than those only detected in prostate-associated fluids. Computational tools can help to select proteins that are secreted, contained in EVs or localized to cell membranes to prioritize candidates for validation in fluids using a variety of different algorithmic approaches121,146148.

Established prostate cancer cell lines have the advantages of being well characterized, and their ease of growth has made them especially useful tools for studying therapy resistance when the availability of patient-derived tissue is limited149151. Use of prostate cancer cell lines has facilitated proteomic studies to investigate the biology of prostate cancer therapy resistance and metastatic potential152,153. However, these prostate cancer cell lines are limited in number and they do not fully represent the clinical spectrum of disease as most were derived frommetastatic tumours. For example, DU145, PC3 and 22Rv1 were developed from castration-resistant metastases, whereas LNCaP cells and VCaP cells were developed from hormone-naive metastases154. Additionally, extensive passaging causes genetic drift, which can result in phenotypic differences between cells used across experiments and laboratories155 and their growth in 2D culture means that their representation of the original tumours is limited. These limitations have been partly addressed by the derivation of primary PDXs in immunodeficient mice, which account for non-immune microenvironmental effects and maintain the genomic and histological features of the original tumour. However, their low engraftment rate, slow growth rate and lack of consistent growth in vivo, hinders their adoption for predictive assays156. In addition, PDXs lose tumour-specific genomic features and subclonal complexity during serial passaging157. Organoids are in vitro, 3D cellular systems that might provide a reasonable compromise between cell lines and PDXs, as they form quickly, maintain the mutational landscape and histological features of their original cancer, and are amenable for therapy prediction assays158,159.

The development of prostate cancer mouse models enables the study of cancer progression throughout all stages from the precancerous state to localized cancer and ultimately to metastatic disease. The transgenic probasin-SV40 T antigen (TRAMP) model provides a model of prostate cancer progression that includes metastatic spread to lymph nodes, bones and lungs160162. The probasin PTEN-null mouse models represent prostate cancer progression from localized to metastatic disease; however, the tumours display androgen-independent growth, in contrast to the TRAMP model128,163,164. Use of label-free LC-MS shotgun proteomics for the characterization of TRAMP model prostate glands enabled the identification of increased abundance of PDGFβ in TRAMP-derived prostate tumours relative to wild type littermates165. Furthermore, proteomic (via iTRAQ-labelled DDA-MS) and transcriptomic (via microarray) profiling of the PTEN-null model demonstrated upregulation of immunity and inflammatory pathways in PTEN-null mice prostate tumours166. Thus, the use of mouse models of prostate cancer has demonstrated early promise in identifying novel protein biomarkers, although a potential limitation in these models is their inability to fully encompass the heterogeneity of human prostate cancer167. Performing sufficiently powered studies using preclinical models is challenging, but these investigations can be a starting point for directing biomarker discovery towards biologically relevant pathways, and building confidence in existing biomarker candidates168.

To date, effectively, almost all existing molecular cancer biomarkers use a single type of information: DNA mutations, RNA abundance, protein abundance or some other information. Because each of these types of molecular information can be dysregulated in cancer in myriad ways, it might be useful to create single biomarkers that merge these different types of information. These so-called multimodal biomarkers are wide-spread in clinical practice, in which clinical assays are often integrated with pathological and radiological data using clinical risk-stratification tools (such as National Comprehensive Cancer Network guidelines) or nomograms13,14. Creating integrated molecular markers might have several advantages. First, they might stabilize risk predictions by reducing technical and biological variance by pooling predictive information across multiple data types. Considerable evidence suggests that pooling predictors reduces variance, even in proof-of-concept studies as simple as incorporating both continuous and discrete RNA abundance in predictive modelling, which has been shown to outperform models that use only continuous or discrete data45,169171. Second, they might increase the ability of the biomarker to distinguish signal from noise by pooling the strengths and alleviating the shortcomings of each individual data type (that is relying on information from one data type in contexts where it is most reliable). Many examples of this possibility exist. For example, in prostate cancer, strong evidence shows that both genomic instability172,173 and tumour evolutionary complexity46 are accurate biomarkers of disease aggression, and both are more accurately detected from the genome than from the transcriptome or proteome174,175; in contrast, the tumour microenvironment does change the prostate cancer genome176, but inferring the state of the tumour microenvironment through the transcriptome or proteome is easier177. Thus, one can imagine a multimodal biomarker measuring both the genome and gene expression (either or both of the transcriptome and proteome) to integrate information from genomic instability, evolutionary complexity and the tumour microenvironment for prostate cancer prognosis. Finally, the lack of correlation between molecular data types might in itself be a useful biomarker, as the lack of correlation could be an indication of additional dysregulation in the cancer signalling network, although to our knowledge, no studies have directly and systematically assessed the biomarker potential of these47,178180. Detailed studies of patient prostate-associated fluids with analysis of features such as matched germline DNA sequencing from blood and fluid EV cargo will be needed to quantify whether these benefits exist and outweigh the financial and implementation-complexity costs of multi-modal molecular biomarkers.

Conclusions

No fluid-based proteomics assay is currently available to supplement the clinical management of prostate cancer. Prostate-associated fluids are suitable for proteomic characterization and are ideal for non-invasive and low-cost targeted proteomics assays. Current challenges for biomarker discovery include limited discovery cohort sizes, suboptimal sample preparation and MS techniques, and lack of appropriate patient cohorts for validation. Existing performance metrics for different assays should not be compared given the variability in size and clinical features of test cohorts. With an increasing number of groups working on this problem, and increased sample collection and clinical annotation efforts, favourable candidates will probably emerge and be clinically validated, providing complementary biomarkers to existing clinical assessments for the diagnosis, risk-stratification and treatment monitoring of prostate cancer.

Key points.

  • Standard-of-care clinical tools for the management of localized prostate cancer result in substantial overdiagnosis and overtreatment.

  • Fluid-based protein biomarkers have the potential to complement clinical decision-making.

  • Advances in mass spectrometry, such as increased scan speeds and mass resolution, have enabled the systematic discovery and validation of protein biomarkers in prostate-associated fluids.

  • Appropriate sample selection for biomarker discovery and validation can improve detection of prostate-derived proteins in fluids.

Acknowledgements

This work was partially funded by the National Cancer Institute Early Detection Research Network (1U01CA214194–01), a Prostate Cancer Canada Discovery Grant (400398) and a Canadian Cancer Society Impact Grant (705649). A.K. was supported by an Ontario Graduate Scholarship and a Paul STARITA Graduate Student Fellowship. L.Y.L. was supported by a CIHR Vanier Award. This work was supported by the NIH/NCI under award number P30CA016042.

Footnotes

Competing interests

The authors declare no competing interests.

Peer review information

Nature Reviews Urology thanks K. Rodland, P. Guedes de Pinho and other anonymous reviewer(s) for their contribution to the peer review of this work.

References

  • 1.Sung H et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 71, 209–249 (2021). [DOI] [PubMed] [Google Scholar]
  • 2.Siegel RL, Miller KD & Jemal A Cancer statistics, 2020. CA Cancer J. Clin. 70, 7–30 (2020). [DOI] [PubMed] [Google Scholar]
  • 3.Musunuru HB et al. Active surveillance for intermediate risk prostate cancer: survival outcomes in the sunnybrook experience. J. Urol. 196, 1651–1658 (2016). [DOI] [PubMed] [Google Scholar]
  • 4.Ku S-YY, Gleave ME & Beltran H Towards precision oncology in advanced prostate cancer. Nat. Rev. Urol. 16, 645–654 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Naji L et al. Digital rectal examination for prostate cancer screening in primary care: a systematic review and meta-analysis. Ann. Fam. Med. 16, 149–154 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Thompson IM et al. Operating characteristics of prostate-specific antigen in men with an initial PSA level of 3.0 ng/mL or lower. J. Am. Med. Assoc. 294, 66–70 (2005). [DOI] [PubMed] [Google Scholar]
  • 7.Serefoglu EC et al. How reliable is 12-core prostate biopsy procedure in the detection of prostate cancer? J. Can. Urol. Assoc. 7, E293–E298 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Freedland SJ et al. Upgrading and downgrading of prostate needle biopsy specimens: risk factors and clinical implications. Urology 69, 495–499 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Epstein JI, Feng Z, Trock BJ & Pierorazio PM Upgrading and downgrading of prostate cancer from biopsy to radical prostatectomy: incidence and predictive factors using the modified Gleason grading system and factoring in tertiary grades. Eur. Urol. 61, 1019–1024 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Loeb S et al. Overdiagnosis and overtreatment of prostate cancer. Eur. Urol. 65, 1046–1055 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Rider JR et al. Long-term outcomes among noncuratively treated men according to prostate cancer risk category in a nationwide, population-based study. Eur. Urol. 63, 88–96 (2013). [DOI] [PubMed] [Google Scholar]
  • 12.Philipson RG et al. Patterns of clinical progression in radiorecurrent high-risk prostate cancer. Eur. Urol. 10.1016/j.eururo.2021.04.035 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Mottet N et al. EAU-ESTRO-SIOG guidelines on prostate cancer. Part I: screening, diagnosis, and local treatment with curative intent. Eur. Urol. 71, 618–629 (2017). [DOI] [PubMed] [Google Scholar]
  • 14.Mohler JL et al. Prostate cancer, version 2.2019. JNCCN 17, 479–505 (2019). [DOI] [PubMed] [Google Scholar]
  • 15.Zimmerman LJ, Li M, Yarbrough WG, Slebos RJC & Liebler DC Global stability of plasma proteomes for mass spectrometry-based analyses. Mol. Cell. Proteom. 11, M111.014340 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Decramer S et al. Urine in clinical proteomics. Mol. Cell. Proteom. 7, 1850–1862 (2008). [DOI] [PubMed] [Google Scholar]
  • 17.Uhlén M et al. Tissue-based map of the human proteome. Science 347, 1260419 (2015). [DOI] [PubMed] [Google Scholar]
  • 18.Ellis MJ et al. Connecting genomic alterations to cancer biology with proteomics: the NCI clinical proteomic tumor analysis consortium. Cancer Discov. 3, 1108–1112 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Aebersold R & Mann M Mass-spectrometric exploration of proteome structure and function. Nature 537, 347–355 (2016). [DOI] [PubMed] [Google Scholar]
  • 20.Stabile A et al. Multiparametric MRI for prostate cancer diagnosis: current status and future directions. Nat. Rev. Urol. 17, 41–61 (2019). [DOI] [PubMed] [Google Scholar]
  • 21.Koo KM, Mainwaring PN, Tomlins SA & Trau M Merging new-age biomarkers and nanodiagnostics for precision prostate cancer management. Nat. Rev. Urol. 16, 302–317 (2019). [DOI] [PubMed] [Google Scholar]
  • 22.Jeon J et al. Temporal stability and prognostic biomarker potential of the prostate cancer urine miRNA transcriptome. J. Natl Cancer Inst. 112, 247–255 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Balk SP, Ko YJ & Bubley GJ Biology of prostate-specific antigen. J. Clin. Oncol. 21, 383–391 (2003). [DOI] [PubMed] [Google Scholar]
  • 24.Hossack T et al. Location and pathological characteristics of cancers in radical prostatectomy specimens identified by transperineal biopsy compared to transrectal biopsy. J. Urol. 188, 781–785 (2012). [DOI] [PubMed] [Google Scholar]
  • 25.Loeb S et al. Systematic review of complications of prostate biopsy. Eur. Urol. 64, 876–892 (2013). [DOI] [PubMed] [Google Scholar]
  • 26.Stefanova V et al. Transperineal prostate biopsies using local anesthesia: experience with 1,287 patients. prostate cancer detection rate, complications and patient tolerability. J. Urol. 201, 1121–1125 (2019). [DOI] [PubMed] [Google Scholar]
  • 27.Lomas DJ & Ahmed HU All change in the prostate cancer diagnostic pathway. Nat. Rev Clin. Oncol. 17, 372–381 (2020). [DOI] [PubMed] [Google Scholar]
  • 28.Capitanio U et al. Biopsy core number represents one of foremost predictors of clinically significant gleason sum upgrading in patients with low-risk prostate cancer. Urology 73, 1087–1091 (2009). [DOI] [PubMed] [Google Scholar]
  • 29.Fütterer JJ et al. Can clinically significant prostate cancer be detected with multiparametric magnetic resonance imaging? A systematic review of the literature. Eur Urol. 68, 1045–1053 (2015). [DOI] [PubMed] [Google Scholar]
  • 30.Ahdoot M et al. MRI-targeted, systematic, and combined biopsy for prostate cancer diagnosis. N. Engl. J. Med. 382, 917–928 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Johnson DC et al. Detection of individual prostate cancer foci via multiparametric magnetic resonance imaging. Eur Urol. 75, 712–720 (2019). [DOI] [PubMed] [Google Scholar]
  • 32.Filson CP et al. Prostate cancer detection with magnetic resonance-ultrasound fusion biopsy: the role of systematic and targeted biopsies. Cancer 122, 884–892 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Schoots IG et al. Magnetic resonance imaging in active surveillance of prostate cancer: a systematic review. Eur Urol. 67, 627–636 (2015). [DOI] [PubMed] [Google Scholar]
  • 34.Hsiang W et al. Outcomes of serial multiparametric magnetic resonance imaging and subsequent biopsy in men with low-risk prostate cancer managed with active surveillance. Eur. Urol. Focus. 7, 47–54 (2019). [DOI] [PubMed] [Google Scholar]
  • 35.Padhani AR, Haider MA, Villers A & Barentsz JO Multiparametric magnetic resonance imaging for prostate cancer detection: what we see and what we miss. Eur Urol. 75, 721–722 (2019). [DOI] [PubMed] [Google Scholar]
  • 36.Sonn GA et al. Prostate magnetic resonance imaging interpretation varies substantially across radiologists. Eur Urol. Focus. 5, 592–599 (2019). [DOI] [PubMed] [Google Scholar]
  • 37.Noguchi M, Stamey TA, McNeal JE & Yemoto CM Relationship between systematic biopsies and histological features of 222 radical prostatectomy specimens: lack of prediction of tumor significance for men with nonpalpable prostate cancer. J. Urol. 166, 104–110 (2001). [PubMed] [Google Scholar]
  • 38.Boutros PC et al. Spatial genomic heterogeneity within localized, multifocal prostate cancer. Nat. Genet. 47, 736–745 (2015). [DOI] [PubMed] [Google Scholar]
  • 39.Chua MLK et al. A prostate cancer “nimbosus”: genomic instability and SChLAP1 dysregulation underpin aggression of intraductal and cribriform subpathologies. Eur. Urol. 72, 665–674 (2017). [DOI] [PubMed] [Google Scholar]
  • 40.Boorjian SA et al. Long-term risk of clinical progression after biochemical recurrence following radical prostatectomy: the impact of time from surgery to recurrence. Eur. Urol. 59, 893–899 (2011). [DOI] [PubMed] [Google Scholar]
  • 41.Addona TA et al. Multi-site assessment of the precision and reproducibility of multiple reaction monitoring-based measurements of proteins in plasma. Nat. Biotechnol. 27, 633–641 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Leth-Larsen R, Lund RR & Ditzel HJ Plasma membrane proteomics and its application in clinical cancer biomarker discovery. Mol. Cell. Proteom. 9, 1369–1382 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Petricoin EF, Belluco C, Araujo RP & Liotta LA The blood peptidome: a higher dimension of information content for cancer biomarker discovery. Nat. Rev. Cancer 6, 961–967 (2006). [DOI] [PubMed] [Google Scholar]
  • 44.Niu Y-N & Xia S-J Stroma-epithelium crosstalk in prostate cancer. Asian J. Androl. 11, 28–35 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Fraser M et al. Genomic hallmarks of localized, non-indolent prostate cancer. Nature 541, 359–364 (2017). [DOI] [PubMed] [Google Scholar]
  • 46.Espiritu SMG et al. The evolutionary landscape of localized prostate cancers drives clinical aggression. Cell 173, 1003–1013.e15 (2018). [DOI] [PubMed] [Google Scholar]
  • 47.Sinha A et al. The proteogenomic landscape of curable prostate cancer. Cancer Cell 35, 414–427.e6 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Lindberg J, Kristiansen A, Wiklund P, Grönberg H & Egevad L Tracking the origin of metastatic prostate cancer. Eur. Urol. 67, 819–822 (2015). [DOI] [PubMed] [Google Scholar]
  • 49.Shipitsin M et al. Identification of proteomic biomarkers predicting prostate cancer aggressiveness and lethality despite biopsy-sampling error. Br. J. Cancer 111, 1201–1212 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Bruderer R et al. Analysis of 1508 plasma samples by capillary-flow data-independent acquisition profiles proteomics of weight loss and maintenance. Mol. Cell. Proteom. 18, 1242–1254 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Halabi S et al. Overall survival of black and white men with metastatic castration-resistant prostate cancer treated with docetaxel. J. Clin. Oncol. 37, 403–410 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Taylor RA et al. Germline BRCA2 mutations drive prostate cancers with distinct evolutionary trajectories. Nat. Commun. 8, 13671 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Steen H & Mann M The ABC’s (and XYZ’s) of peptide sequencing. Nat. Rev. Mol. Cell Biol. 5, 699–711 (2004). [DOI] [PubMed] [Google Scholar]
  • 54.Laskay ÜA, Lobas AA, Srzentić K, Gorshkov MV & Tsybin YO Proteome digestion specificity analysis for rational design of extended bottom-up and middle-down proteomics experiments. J. Proteome Res. 12, 5558–5569 (2013). [DOI] [PubMed] [Google Scholar]
  • 55.Batth TS, Francavilla C & Olsen JV Off-line high-pH reversed-phase fractionation for in-depth phosphoproteomics. J. Proteome Res. 13, 6176–6186 (2014). [DOI] [PubMed] [Google Scholar]
  • 56.Wolters DA, Washburn MP & Yates JR An automated multidimensional protein identification technology for shotgun proteomics. Anal. Chem. 73, 5683–5690 (2001). [DOI] [PubMed] [Google Scholar]
  • 57.Batth TS & Olsen JV Offline high pH reversed-phase peptide fractionation for deep phosphoproteome coverage. in Methods in Molecular Biology Vol. 1355 179–192 (Humana Press Inc., 2016). [DOI] [PubMed] [Google Scholar]
  • 58.Gomes FP & Yates JR Recent trends of capillary electrophoresis-mass spectrometry in proteomics research. Mass. Spectrom. Rev. 38, 445–460 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Vasilopoulou CG et al. Trapped ion mobility spectrometry and PASEF enable in-depth lipidomics from minimal sample amounts. Nat. Commun. 11, 331 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Bekker-Jensen DB et al. A compact quadrupole-orbitrap mass spectrometer with FAIMS interface improves proteome coverage in short LC gradients. Mol. Cell. Proteom. 19, 716–729 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Mitchell Wells J & McLuckey SA Collision-induced dissociation (CID) of peptides and proteins. Methods Enzymol. 402, 148–185 (2005). [DOI] [PubMed] [Google Scholar]
  • 62.Kislinger T et al. PRISM, a generic large scale proteomic investigation strategy for mammals. Mol. Cell. Proteom. 2, 96–106 (2003). [DOI] [PubMed] [Google Scholar]
  • 63.Elias JE, Haas W, Faherty BK & Gygi SP Comparative evaluation of mass spectrometry platforms used in large-scale proteomics investigations. Nat. Methods 2, 667–675 (2005). [DOI] [PubMed] [Google Scholar]
  • 64.Wong JWH & Cagney G An overview of label-free quantitation methods in proteomics by mass spectrometry. Methods Mol. Biol. 604, 273–283 (2010). [DOI] [PubMed] [Google Scholar]
  • 65.Thompson A et al. Tandem mass tags: A novel quantification strategy for comparative analysis of complex protein mixtures by MS/MS. Anal. Chem. 75, 1895–1904 (2003). [DOI] [PubMed] [Google Scholar]
  • 66.Hogrebe A et al. Benchmarking common quantification strategies for large-scale phosphoproteomics. Nat. Commun. 9, 1045 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Altelaar AFM et al. Benchmarking stable isotope labeling based quantitative proteomics. J. Proteom. 88, 14–26 (2013). [DOI] [PubMed] [Google Scholar]
  • 68.Li Z et al. Systematic comparison of label-free, metabolic labeling, and isobaric chemical labeling for quantitative proteomics on LTQ orbitrap velos. J. Proteome Res. 11, 1582–1590 (2012). [DOI] [PubMed] [Google Scholar]
  • 69.Venable JD, Dong MQ, Wohlschlegel J, Dillin A & Yates JR Automated approach for quantitative analysis of complex peptide mixtures from tandem mass spectra. Nat. Methods 1, 39–45 (2004). [DOI] [PubMed] [Google Scholar]
  • 70.Xuan Y et al. Standardization and harmonization of distributed multi-center proteotype analysis supporting precision medicine studies. Nat. Commun. 11, 5248 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Röst HL et al. OpenSWATH enables automated, targeted analysis of data-independent acquisition MS data. Nat. Biotechnol. 32, 219–223 (2014). [DOI] [PubMed] [Google Scholar]
  • 72.Principe S et al. In-depth proteomic analyses of exosomes isolated from expressed prostatic secretions in urine. Proteomics 13, 1667–1671 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Rontogianni S et al. Proteomic profiling of extracellular vesicles allows for human breast cancer subtyping. Commun. Biol. 2, 325 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Yang W, Freeman MR & Kyprianou N Personalization of prostate cancer therapy through phosphoproteomics. Nat. Rev. Urol. 15, 483–497 (2018). [DOI] [PubMed] [Google Scholar]
  • 75.Drake JM et al. Phosphoproteome integration reveals patient-specific networks in prostate cancer. Cell 166, 1041–1054 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Gahmberg CG & Tolvanen M Why mammalian cell surface proteins are glycoproteins. Trends Biochem. Sci. 21, 308–311 (1996). [PubMed] [Google Scholar]
  • 77.Leitner A Enrichment strategies in phosphoproteomics. in Methods in Molecular Biology Vol. 1355 105–121 (Humana Press Inc., 2016). [DOI] [PubMed] [Google Scholar]
  • 78.Riley NM, Bertozzi CR & Pitteri SJ A pragmatic guide to enrichment strategies for mass spectrometry-based glycoproteomics. Mol. Cell. Proteom. 20, 100029 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Mertins P et al. Reproducible workflow for multiplexed deep-scale proteome and phosphoproteome analysis of tumor tissues by liquid chromatography-mass spectrometry. Nat. Protoc. 13, 1632–1661 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Huang P, Li H, Gao W, Cai Z & Tian R A fully integrated spintip-based approach for sensitive and quantitative profiling of region-resolved in vivo brain glycoproteome. Anal. Chem. 91, 9181–9189 (2019). [DOI] [PubMed] [Google Scholar]
  • 81.Leutert M, Rodriguez-Mias RA, Fukuda NK & Villen J R2-P2 rapid-robotic phosphoproteomics enables multidimensional cell signaling studies. Mol. Syst. Biol. 15, e9021 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Zhu Y et al. Nanodroplet processing platform for deep and quantitative proteome profiling of 10–100 mammalian cells. Nat. Commun. 9, 882 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Moggridge S, Sorensen PH, Morin GB & Hughes CS Extending the compatibility of the SP3 paramagnetic bead processing approach for proteomics. J. Proteome Res. 17, 1730–1740 (2018). [DOI] [PubMed] [Google Scholar]
  • 84.Frantzi M, Latosinska A, Merseburger AS & Mischak H Recent progress in urinary proteome analysis for prostate cancer diagnosis and management. Expert Rev. Mol. Diagn. 15, 1539–1554 (2015). [DOI] [PubMed] [Google Scholar]
  • 85.Jackson HW et al. The single-cell pathology landscape of breast cancer. Nature 578, 615–620 (2020). [DOI] [PubMed] [Google Scholar]
  • 86.Rifai N, Gillette MA & Carr SA Protein biomarker discovery and validation: the long and uncertain path to clinical utility. Nat. Biotechnol. 24, 971–983 (2006). [DOI] [PubMed] [Google Scholar]
  • 87.Hoofnagle AN & Wener MH The fundamental flaws of immunoassays and potential solutions using tandem mass spectrometry. J. Immunol. Methods 347, 3–11 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Joshi A & Mayr M In aptamers they trust: caveats of the SOMAscan biomarker discovery Platform from SomaLogic. Circulation 138, 2482–2485 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Wang P, Whiteaker JR & Paulovich AG The evolving role of mass spectrometry in cancer biomarker discovery. Cancer Biol. Ther. 8, 1083–1094 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Lange V, Picotti P, Domon B & Aebersold R Selected reaction monitoring for quantitative proteomics: a tutorial. Mol. Syst. Biol. 4, 222 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Carr SA & Anderson L Protein quantitation through targeted mass spectrometry: the way out of biomarker purgatory? Clin. Chem. 54, 1749–1752 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Schiess R, Wollscheid B & Aebersold R Targeted proteomic strategy for clinical biomarker discovery. Mol. Oncol. 3, 33–44 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Anderson NL et al. A human proteome detection and quantitation project. Mol. Cell. Proteom. 8, 883–886 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Mallick P et al. Computational prediction of proteotypic peptides for quantitative proteomics. Nat. Biotechnol. 25, 125–131 (2007). [DOI] [PubMed] [Google Scholar]
  • 95.Faserl K, Sarg B, Maurer V & Lindner HH Exploiting charge differences for the analysis of challenging post-translational modifications by capillary electrophoresis-mass spectrometry. J. Chromatogr. A 1498, 215–223 (2017). [DOI] [PubMed] [Google Scholar]
  • 96.Lombard-Banek C, Choi SB & Nemes P Single-cell proteomics in complex tissues using microprobe capillary electrophoresis mass spectrometry. in Methods in Enzymology Vol. 628 263–292 (Academic Press Inc., 2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Carr SA et al. Targeted peptide measurements in biology and medicine: best practices for mass spectrometry-based assay development using a fit-for-purpose approach. Mol. Cell. Proteom. 13, 907–917 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Virreira Winter S et al. Urinary proteome profiling for stratifying patients with familial Parkinson’s disease. EMBO Mol Med. 13, e13257 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Geyer PE et al. Plasma proteome profiling to detect and avoid sample-related biases in biomarker studies. EMBO Mol. Med. 11, e10427 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Makarov A, Denisov E, Lange O & Horning S Dynamic range of mass accuracy in LTQ orbitrap hybrid mass spectrometer. J. Am. Soc. Mass. Spectrom. 17, 977–982 (2006). [DOI] [PubMed] [Google Scholar]
  • 101.Geyer PE, Holdt LM, Teupser D & Mann M Revisiting biomarker discovery by plasma proteomics. Mol. Syst. Biol. 13, 942 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Annesley TM Ion suppression in mass spectrometry. Clin. Chem. 49, 1041–1044 (2003). [DOI] [PubMed] [Google Scholar]
  • 103.Hanash SM, Pitteri SJ & Faca VM Mining the plasma proteome for cancer biomarkers. Nature 452, 571–579 (2008). [DOI] [PubMed] [Google Scholar]
  • 104.Tu C et al. Depletion of abundant plasma proteins and limitations of plasma proteomics. J. Proteome Res. 9, 4982–4991 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Drake RR et al. In-depth proteomic analyses of direct expressed prostatic secretions. J. Proteome Res. 9, 2109–2116 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Kim Y et al. Identification of differentially expressed proteins in direct expressed prostatic secretions of men with organ-confined versus extracapsular prostate cancer. Mol. Cell. Proteom. 11, 1870–1884 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Drabovich AP, Saraon P, Jarvi K & Diamandis EP Seminal plasma as a diagnostic fluid for male reproductive system disorders. Nat. Rev. Urol. 11, 278–288 (2014). [DOI] [PubMed] [Google Scholar]
  • 108.Drake RR et al. Clinical collection and protein properties of expressed prostatic secretions as a source for biomarkers of prostatic disease. J. Proteom. 72, 907–917 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.McNaughton Collins M, Fowler FJ, Elliott DB, Albertsen PC & Barry MJ Diagnosing and treating chronic prostatitis: do urologists use the four-glass test? Urology 55, 403–407 (2000). [DOI] [PubMed] [Google Scholar]
  • 110.Theodorescu D et al. Discovery and validation of urinary biomarkers for prostate cancer. Proteomics Clin. Appl. 2, 556–570 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Harpole M, Davis J & Espina V Current state of the art for enhancing urine biomarker discovery. Expert Rev. Proteom. 13, 609–626 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Principe S et al. Identification of prostate-enriched proteins by in-depth proteomic analyses of expressed prostatic secretions in urine. J. Proteome Res. 11, 2386–2396 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Pellegrini KL et al. Detection of prostate cancer-specific transcripts in extracellular vesicles isolated from post-DRE urine. Prostate 77, 990–999 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Kim Y et al. Targeted proteomics identifies liquid-biopsy signatures for extracapsular prostate cancer. Nat. Commun. 7, 11906 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Bussemakers MJ et al. DD3: a new prostate-specific gene, highly overexpressed in prostate cancer 1. Cancer Res. 59, 5975–5979 (1999). [PubMed] [Google Scholar]
  • 116.van Niel G, D’Angelo G & Raposo G Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 19, 213–228 (2018). [DOI] [PubMed] [Google Scholar]
  • 117.Becker A et al. Extracellular vesicles in cancer: cell-to-cell mediators of metastasis. Cancer Cell 30, 836–848 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Overbye A et al. Identification of prostate cancer biomarkers in urinary exosomes. Oncotarget 6, 30357–30376 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Merchant ML, Rood IM, Deegens JKJ & Klein JB Isolation and characterization of urinary extracellular vesicles: implications for biomarker discovery. Nat. Rev. Nephrol. 13, 731–749 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Füzéry AK, Levin J, Chan MM & Chan DW Translation of proteomic biomarkers into FDA approved cancer diagnostics: Issues and challenges. Clin. Proteom. 10, 13 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Uhlén M et al. The human secretome. Sci. Signal. 12, eaaz0274 (2019). [DOI] [PubMed] [Google Scholar]
  • 122.Sequeiros T et al. Targeted proteomics in urinary extracellular vesicles identifies biomarkers for diagnosis and prognosis of prostate cancer. Oncotarget 8, 4960–4976 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Jedinak A et al. Novel non-invasive biomarkers that distinguish between benign prostate hyperplasia and prostate cancer. BMC Cancer 15, 259 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Zhang M et al. Combined serum and EPS-urine proteomic analysis using iTRAQ technology for discovery of potential prostate cancer biomarkers. Discov. Med. 22, 281–295 (2016). [PubMed] [Google Scholar]
  • 125.Grupp K et al. Cysteine-rich secretory protein 3 overexpression is linked to a subset of PTEN-deleted ERG fusion-positive prostate cancers with early biochemical recurrence. Mod. Pathol. 26, 733–742 (2013). [DOI] [PubMed] [Google Scholar]
  • 126.Al Bashir S et al. Cysteine-rich secretory protein 3 (CRISP3), ERG and PTEN define a molecular subtype of prostate cancer with implication to patients’ prognosis. J. Hematol. Oncol. 7, 21 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Macagno A et al. Analytical performance of thrombospondin-1 and cathepsin D immunoassays part of a novel CE-IVD marked test as an aid in the diagnosis of prostate cancer. PLoS One 15, e0233442 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Cima I et al. Cancer genetics-guided discovery of serum biomarker signatures for diagnosis and prognosis of prostate cancer. Proc. Natl Acad. Sci. USA 108, 3342–3347 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Pye H et al. Evaluation of Proclarix, a prostate cancer risk score, used together with magnetic resonance imaging for the diagnosis of clinically significant prostate cancer. J. Clin. Oncol. 38, 278–278 (2020). [Google Scholar]
  • 130.Fujita K et al. Proteomic analysis of urinary extracellular vesicles from high Gleason score prostate cancer. Sci. Rep. 7, 1–9 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Gandham S et al. Technologies and standardization in research on extracellular vesicles. Trends Biotechnol. 38, 1066–1098 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Yan B et al. iTRAQ-based comparative serum proteomic analysis of prostate cancer patients with or without bone metastasis. J. Cancer 10, 4165–4177 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Kohli M et al. Serum proteomics on the basis of discovery of predictive biomarkers of response to androgen deprivation therapy in advanced prostate cancer. Clin. Genitourin. Cancer 17, 248–253.e7 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Ishizuya Y et al. The role of actinin-4 (ACTN4) in exosomes as a potential novel therapeutic target in castration-resistant prostate cancer. Biochem. Biophys. Res. Commun. 523, 588–594 (2020). [DOI] [PubMed] [Google Scholar]
  • 135.Abbatiello SE, Mani DR, Keshishian H & Carr SA Automated detection of inaccurate and imprecise transitions in peptide quantification by multiple reaction monitoring mass spectrometry. Clin. Chem. 56, 291–305 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Müller T et al. Automated sample preparation with SP 3 for low-input clinical proteomics. Mol. Syst. Biol. 16, e911 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Stadlmann J et al. Improved sensitivity in low-input proteomics using micropillar array-based chromatography. Anal. Chem. 91, 14203–14207 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Zhang Z & Chan DW The road from discovery to clinical diagnostics: lessons learned from the first FDA-cleared in vitro diagnostic multivariate index assay of proteomic. Biomarkers. 19, 2995–2999 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Kraus VB Biomarkers as drug development tools: discovery, validation, qualification and use. Nat. Rev. Rheumatol. 14, 354–362 (2018). [DOI] [PubMed] [Google Scholar]
  • 140.Raab SS The cost-effectiveness of immunohistochemistry. Arch. Pathol. Lab. Med. 124, 1185–1191 (2000). [DOI] [PubMed] [Google Scholar]
  • 141.Drake JM et al. Metastatic castration-resistant prostate cancer reveals intrapatient similarity and interpatient heterogeneity of therapeutic kinase targets. Proc. Natl Acad. Sci. USA 110, E4762–E4769 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Myers JS et al. Proteomic characterization of paired non-malignant and malignant African-American prostate epithelial cell lines distinguishes them by structural proteins. BMC Cancer 17, 480 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Jarnuczak AF et al. An integrated landscape of protein expression in human cancer. Sci. Data 8, 115 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Kishan AU et al. Transcriptomic heterogeneity of gleason grade group 5 prostate cancer. Eur. Urol. 78, 327–332 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Guo T et al. Multi-region proteome analysis quantifies spatial heterogeneity of prostate tissue biomarkers. Life Sci. Alliance 1, e201800042 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Keerthikumar S et al. ExoCarta: a web-based compendium of exosomal cargo. J. Mol. Biol. 428, 688–692 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Käll L, Krogh A & Sonnhammer ELL Advantages of combined transmembrane topology and signal peptide prediction — the Phobius web server. Nucleic Acids Res. 35, W429–W432 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Nielsen H Predicting secretory proteins with signalP in Methods in Molecular Biology Vol. 1611 59–73 (Humana Press Inc., 2017). [DOI] [PubMed] [Google Scholar]
  • 149.Namekawa T, Ikeda K, Horie-Inoue K & Inoue S Application of prostate cancer models for preclinical study: advantages and limitations of cell lines, patient-derived xenografts, and three-dimensional culture of patient-derived cell. Cells 8, 74 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Huang X et al. miRNA-95 mediates radioresistance in tumors by targeting the sphingolipid phosphatase SGPP1. Cancer Res. 73, 6972–6986 (2013). [DOI] [PubMed] [Google Scholar]
  • 151.Ghiam AF et al. Long non-coding RNA urothelial carcinoma associated 1 (UCA1) mediates radiation response in prostate cancer. Oncotarget 8, 4668–4689 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Höti N, Shah P, Hu Y, Yang S & Zhang H Proteomics analyses of prostate cancer cells reveal cellular pathways associated with androgen resistance. Proteomics 17, 10.1002/pmic.201600228 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Katsogiannou M et al. Integrative proteomic and phosphoproteomic profiling of prostate cell lines. PLoS One 14, e0224148 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Cunningham D & You Z In vitro and in vivo model systems used in prostate cancer research. J. Biol. Methods 2, e17 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Ben-David U et al. Genetic and transcriptional evolution alters cancer cell line drug response. Nature 560, 325–330 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Davies AH, Wang Y & Zoubeidi A Patient-derived xenografts: a platform for accelerating translational research in prostate cancer. Mol. Cell. Endocrinol. 462, 17–24 (2018). [DOI] [PubMed] [Google Scholar]
  • 157.Ben-David U et al. Patient-derived xenografts undergo mouse-specific tumor evolution. Nat. Genet. 49, 1567–1575 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Gao D et al. Organoid cultures derived from patients with advanced prostate cancer. Cell 159, 176–187 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Clevers H Modeling development and disease with organoids. Cell 165, 1586–1597 (2016). [DOI] [PubMed] [Google Scholar]
  • 160.Gingrich JR & Greenberg NM A transgenic mouse prostate cancer model. Toxicol. Pathol. 24, 502–504 (1996). [DOI] [PubMed] [Google Scholar]
  • 161.Gingrich JR et al. Metastatic prostate cancer in a transgenic mouse. Cancer Res. 56, 4096–102 (1996). [PubMed] [Google Scholar]
  • 162.Gelman IH How the tramp model revolutionized the study of prostate cancer progression. Cancer Res. 76, 6137–6139 (2016). [DOI] [PubMed] [Google Scholar]
  • 163.Pencik J et al. STAT3 regulated ARF expression suppresses prostate cancer metastasis. Nat. Commun. 6, 7736 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Wang S et al. Prostate-specific deletion of the murine Pten tumor suppressor gene leads to metastatic prostate cancer. Cancer Cell 4, 209–221 (2003). [DOI] [PubMed] [Google Scholar]
  • 165.Zhang Y et al. Quantitative proteomics of TRAMP mice combined with bioinformatics analysis reveals that PDGF-B regulatory network plays a key role in prostate cancer progression. J. Proteome Res. 17, 2401–2411 (2018). [DOI] [PubMed] [Google Scholar]
  • 166.Zhang J et al. Proteomic and transcriptomic profiling of Pten gene-knockout mouse model of prostate cancer. Prostate 80, 588–605 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Hensleym PJ & Kyprianou N Modeling prostate cancer in mice: review limitations and opportunities. J. Androl. 33, 133–144 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Terp MG & Ditzel HJ Application of proteomics in the study of rodent models of cancer. Proteom. Clin. Appl. 8, 640–652 (2014). [DOI] [PubMed] [Google Scholar]
  • 169.Gong IY, Fox NS, Huang V & Boutros PC Prediction of early breast cancer patient survival using ensembles of hypoxia signatures. PLoS One 13, e0204123 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Bayani J et al. Molecular stratification of early breast cancer identifies drug targets to drive stratified medicine. NPJ Breast Cancer 3, 3 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Bhandari V & Boutros PC Comparing continuous and discrete analyses of breast cancer survival information. Genomics 108, 78–83 (2016). [DOI] [PubMed] [Google Scholar]
  • 172.Lalonde E et al. Tumour genomic and microenvironmental heterogeneity for integrated prediction of 5-year biochemical recurrence of prostate cancer: a retrospective cohort study. Lancet Oncol. 15, 1521–1532 (2014). [DOI] [PubMed] [Google Scholar]
  • 173.Lalonde E et al. Translating a prognostic DNA genomic classifier into the clinic: retrospective validation in 563 localized prostate tumors. Eur. Urol. 72, 22–31 (2017). [DOI] [PubMed] [Google Scholar]
  • 174.Andor N, Maley CC & Ji HP Genomic instability in cancer: teetering on the limit of tolerance. Cancer Res. 77, 2179–2185 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Tarabichi M et al. A practical guide to cancer subclonal reconstruction from DNA sequencing. Nat. Methods 18, 144–155 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Bhandari V et al. Molecular landmarks of tumor hypoxia across cancer types. Nat. Genet. 51, 308–318 (2019). [DOI] [PubMed] [Google Scholar]
  • 177.Bhandari V, Li CH, Bristow RG & Boutros PC Divergent mutational processes distinguish hypoxic and normoxic tumours. Nat. Commun. 11, 737 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Hopkins JF et al. Mitochondrial mutations drive prostate cancer aggression. Nat. Commun. 8, 656 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Haider S et al. Pathway-based subnetworks enable cross-disease biomarker discovery. Nat. Commun. 9, 4746 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Fox NS, Haider S, Harris AL & Boutros PC Landscape of transcriptomic interactions between breast cancer and its microenvironment. Nat. Commun. 10, 3116 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Endt K et al. Development and clinical testing of individual immunoassays for the quantification of serum glycoproteins to diagnose prostate cancer. PLoS ONE 12, e0181557 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Steuber T et al. Thrombospondin 1 and cathepsin D improve prostate cancer diagnosis by avoiding potentially unnecessary prostate biopsies. BJU Int. 123, 826–833 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Klocker H et al. Development and validation of a novel multivariate risk score to guide biopsy decision for the diagnosis of clinically significant prostate cancer. BJUI Compass 1, 15–20 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES