International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Prostate cancer (PCa) is a major public health burden in men: In 2022, there were ~1.47 million new prostate cancer cases globally, making it the second most common cancer in men, and it accounted for ~4.1% of all male cancer mortalities worldwide. In the United States alone, ~313,780 new cases and 35,770 mortalities from prostate cancer are projected in 2025 (1,2). Early and accurate diagnosis is pivotal to improving clinical outcomes, yet the disease often progresses silently in its initial stages. Conventional screening approaches-such as serum prostate-specific antigen (PSA) testing and digital rectal examination-have long served as cornerstones of early detection (2). However, their limited specificity and inability to distinguish indolent from aggressive disease frequently result in overdiagnosis, overtreatment and associated morbidity (3). These shortcomings highlight a need for novel biomarkers that enable more precise disease characterization, risk stratification and individualized treatment planning (4).
Recent advances in genomics, transcriptomics and metabolomics have revolutionized biomarker discovery, unveiling new molecular signatures that bridge laboratory research and clinical application (5). Genetic alterations such as mutations in BRCA1/2, HOXB13 and PTEN, as well as TMPRSS2-ERG gene fusions, have been associated with PCa initiation, progression and therapeutic resistance (6,7). These genetic insights not only enhance the understanding of tumor biology but also aid in identifying high-risk patients, predicting treatment response and guiding precision interventions such as PARP inhibitor therapy (8).
Parallel to these genomic developments, RNA-based biomarkers-including prostate cancer antigen (PCA) 3, microRNAs (miRNAs; such as miR-141 and miR-375) and long non-coding RNAs (lncRNAs)-have emerged as promising tools for non-invasive diagnosis (9). Detection of these RNA molecules in urine, blood or exosomal (exo) fractions notably improves diagnostic accuracy when used alongside PSA testing, thereby minimizing unnecessary biopsies (10). The advent of liquid biopsy, encompassing circulating tumor DNA (ctDNA) and exo RNA analysis, further enhances real-time disease monitoring. This approach enables dynamic assessment of tumor burden, therapeutic response and the emergence of resistance-associated mutations in key genes such as androgen receptor (AR) and BRCA1/2 (11,12). In addition to genomic and transcriptomic markers, metabolic biomarkers provide valuable insights into PCa pathophysiology. Metabolites such as citrate, polyamines and sarcosine reflect the distinct metabolic reprogramming that occurs during malignant transformation (13). Their quantification in biofluids-including serum, urine and prostatic secretions-offers complementary diagnostic information and helps differentiate benign prostatic hyperplasia from malignant disease (14).
Collectively, these discoveries mark a paradigm shift in PCa management. The integration of genetic, RNA and metabolic biomarkers into diagnostic workflows has the potential to transform routine screening, prognostic evaluation and therapeutic decision-making. The present review synthesizes current evidence on emerging biomarkers in PCa, emphasizing their translational relevance, diagnostic performance and clinical feasibility. The present review also discusses persistent challenges, such as biomarker validation, inter-population variability and the harmonization of multi-omics data-and explores future directions, including artificial intelligence (AI)-assisted liquid biopsy interpretation and multi-omics integration for advancing precision oncology (Fig. 1) (15).
PSA, encoded by the KLK3 gene, is a glycoprotein predominantly secreted by the epithelial cells of the prostate gland (16). While its primary physiological role is the liquefaction of semen, its high organ specificity has established it as a cornerstone biomarker in the screening and diagnosis of prostate diseases (17). In the serum, PSA exists primarily in three molecular forms: Free PSA (f-PSA), which constitutes 10-30% of the total PSA (t-PSA); PSA complexed with α1-antichymotrypsin and PSA complexed with α2-macroglobulin. The latter two are collectively termed complexed PSA (18).
f/t PSA provides key diagnostic information beyond t-PSA levels alone (19). In patients with PCa, the proportion of f-PSA is markedly lower compared with those with benign conditions such as benign prostatic hyperplasia. The f/t-PSA ratio is calculated as follows (20): f-PSA/t-PSA=free PSA concentration (ng/ml)/total PSA concentration (ng/ml).
An f/t PSA ratio <0.15 is associated with an increased risk of malignancy and is a key factor in recommending prostate biopsy, thereby improving diagnostic specificity (21,22). This metric is particularly valuable within the diagnostic 'gray zone' of t-PSA (4-10 ng/ml) (23). A ratio >0.25 typically suggests a benign etiology, whereas a ratio <0.15 warrants further invasive assessment (24,25). Notably, when t-PSA >10 ng/ml, biopsy is generally indicated regardless of the f/t PSA ratio (26). A large multicenter study demonstrated that incorporating the f/t PSA ratio into clinical decision-making reduced misclassification by ~43%, underscoring its utility in enhancing diagnostic accuracy (27).
Emerging strategies seek to integrate the f/t PSA ratio with other clinical parameters, including patient age, family history and racial background, to develop multivariate predictive models (28,29). These refined algorithms promise to further personalize PCa risk stratification and support early intervention strategies (30).
PSAD, defined as the ratio of serum PSA concentration to prostate volume (PSAD=t-PSA/prostate volume), refines diagnostic precision by accounting for gland size (31). Prostate volume is typically quantified via transrectal ultrasound (TRUS) or, with increasing frequency, MRI. A PSAD value >0.15 ng/ml/cm3 is consistently associated with a heightened risk of PCa (32,33). This normalization mitigates the confounding effect of benign gland enlargement, thereby reducing false-positive results (34).
Longitudinal cohort studies have confirmed that an elevated PSAD is a robust predictor of PCa incidence and progression (35,36). Recent evidence indicates that MRI-based PSAD calculations, benefiting from superior soft-tissue resolution, outperform TRUS-derived values (37). Consequently, MRI-calculated PSAD exhibits enhanced diagnostic sensitivity and specificity in discriminating malignant from benign lesions, supporting its integration into modern diagnostic pathways (38).
PSAV measures the dynamic change in PSA levels over time, calculated as the absolute change in PSA per year [PSAV=(PSA2-PSA1)/ΔTime in years] (39). A velocity >0.75 ng/ml per year is considered clinically considerable and suspicious for underlying malignancy (40). PSAV is particularly useful for monitoring individuals with borderline PSA levels and for assessing disease progression in patients' post-treatment (41). A rapid rise in PSA is often associated with aggressive tumor biology and a worse prognosis (42).
The clinical utility of PSAV is being augmented by AI and machine learning (43). Emerging models integrate PSAV with a multitude of clinical, imaging and genomic parameters to achieve more accurate predictions of disease trajectory, thereby aiding in personalized surveillance and therapeutic decision-making (44,45).
PHI is an advanced blood test that combines three distinct forms of PSA: f-PSA, t-PSA and a truncated form of kallikrein (KLK)-related protein 4 (k-PSA, also known as hK4). It is calculated using the following formula (46): PHI=(f-PSA)/(t-PSA) x t-PSA √(t-PSA) x k-PSA.
The resulting score stratifies patients according to their risk of harboring clinically considerable PCa, with higher values indicating greater risk (47). The established clinical thresholds guide management as follows: i) A PHI <15 suggests a low likelihood of malignancy, ii) values between 15 and 35 fall into an indeterminate range where adjunctive evaluation with multiparametric MRI (mpMRI) is recommended, iii) a PHI >35 signifies high risk and typically warrants biopsy (48,49). PHI demonstrates superior diagnostic accuracy compared with t-PSA or f/t PSA alone, particularly within the t-PSA gray zone and has been shown to reduce unnecessary biopsies by ~20% (50,51). Prospective validation studies confirm that PHI effectively identifies aggressive PCa, improving patient selection for invasive procedures (52,53).
The integration of PHI with mpMRI represents a powerful diagnostic synergy. This combined model enhances detection confidence for clinically considerable cancer while minimizing overtreatment (54). Furthermore, ongoing research into ethnicity-specific PHI thresholds highlights the importance of personalized diagnostic standards to optimize accuracy across diverse populations (55,56).
KLK family comprises 15 secreted serine proteases (KLK1-KLK15) clustered on chromosome 19q13.4. These enzymes share structural similarity, characterized by a conserved catalytic triad of histidine, aspartic acid and serine residues (57-59). KLKs participate in diverse physiological processes, including skin desquamation, neural plasticity and inflammation (60-62). For instance, KLK1 cleaves kininogen to generate bradykinin, thereby modulating vascular tone, permeability and inflammatory signaling (63). KLK5 and KLK7 regulate keratinocyte differentiation and desquamation, with aberrant activity implicated in psoriasis and atopic dermatitis (64,65).
Among all KLKs, KLK3 (a PSA) is the best-established biomarker for PCa (66). Nevertheless, increasing evidence highlights that other family members, particularly KLK2, act synergistically with KLK3 to promote prostate tumorigenesis and reshape the tumor microenvironment (67,68). Dysregulation of the KLK proteolytic network facilitates cancer progression by enhancing tumor invasion, angiogenesis and metastasis (69).
Transcriptomic analyses demonstrate distinct expression patterns of multiple KLKs in prostate carcinoma compared with benign tissue, underscoring their combined diagnostic and prognostic potential (70,71). KLK2, sharing ~78% sequence identity with KLK3, is also AR-regulated and activates pro-PSA by cleaving its pro-peptide (72). Due to its highly restricted expression profile, KLK2 has emerged as an attractive therapeutic target (73). Several innovative platforms are currently in development, including: i) Radioligand therapies delivering cytotoxic payloads selectively to KLK2-expressing cells (74), ii) bispecific T-cell engagers redirecting cytotoxic lymphocytes toward KLK2-positive tumor cells (75); and iii) chimeric antigen receptor T-cell therapies engineered to recognize KLK2 on the cancer cell surface (76). Promising clinical candidates from companies such as Johnson & Johnson and EpimAb Biotherapeutics have advanced into Phase I trials, representing an encouraging step toward precision immunotherapy in PCa (77,78).
Nucleic acid biomarkers, encompassing DNA- and RNA-based alterations, have emerged as powerful tools for the diagnosis, prognostic stratification and treatment selection of PCa (79). Their analysis, particularly through non-invasive liquid biopsies, offers a real-time snapshot of tumor genetics and dynamics (80).
Mutations in the BRCA1 and, more importantly, BRCA2 genes are associated with hereditary PCa (81). Carriers face a 2-3-fold increased lifetime risk of developing the disease and are more likely to present with aggressive tumor phenotypes (82). Genetic testing for these mutations is key for identifying high-risk individuals, guiding genetic counseling and implementing intensified screening protocols (such as more frequent PSA monitoring or MRI) for early detection (83).
HOXB13 gene, located on chromosome 17q21.3, encodes a transcription factor key for prostate development and homeostasis (84,85). The G84E germline mutation (c.250G>A) is an autosomal dominant risk allele, found in 5-10% of familial PCa cases, with a higher prevalence in European and North American populations (86-89). Carriers have a 2-3-fold increased PCa risk, a cumulative risk of <42% by age 70 and are associated with higher-grade tumors, increased postoperative recurrence and worse outcomes, particularly in metastatic castration-resistant prostate cancer (mCRPC) (90-93).
Somatic mutations in key tumor suppressor genes drive PCa progression. PTEN loss, occurring in 30-40% of localized cancer types and more frequently in mCRPC, hyperactivates the PI3K-AKT pathway, promoting tumor invasiveness and resistance to androgen deprivation therapy (94-96). TP53 mutations, rare in localized disease but present in 30-40% of mCRPC, induce genomic instability and chemoresistance (97-99). ATM mutations, involved in DNA damage repair, are found in 10-15% of mCRPC cases and influence sensitivity to PARP inhibitors and platinum-based chemotherapy (100-102). Co-mutations, such as PTEN/TP53 or ATM/BRCA2, are common in advanced disease and associate with aggressive clinical behavior and distinct therapeutic vulnerabilities (103-106).
TMPRSS2 gene, located on chromosome 21q22.3, encodes a transmembrane serine protease under AR regulation (107). It is predominantly expressed in prostate epithelial cells, where it contributes to extracellular matrix remodeling and signal transduction (108). The ERG gene, situated nearby on 21q22.2, belongs to the ETS transcription factor family and is normally expressed in hematopoietic cells but not in healthy prostate tissue (109).
In PCa, intrachromosomal rearrangements such as inversions or translocations within the 21q22 region can fuse the TMPRSS2 promoter with the ERG coding sequence, creating the TMPRSS2-ERG fusion gene (110). This fusion places ERG expression under AR control, leading to aberrant ERG overexpression in prostate epithelial cells and promoting tumorigenesis (111).
The TMPRSS2-ERG fusion occurs in 40-60% of sporadic prostate cancer types, representing the most frequent gene rearrangement in PCa (112). Its prevalence increases to 50-70% in familial and early-onset (≤55 years) cases (113). Fusion-positive tumors are often associated with higher Gleason scores (≥7), elevated PSA levels and increased risk of lymph node and bone metastases, as well as castration-resistant progression (114,115). However, the independent prognostic value of TMPRSS2-ERG for overall survival remains controversial, as it may depend on co-occurring genomic alterations (116).
Mechanistically, ERG activation regulates multiple downstream targets, including matrix metalloproteinases and vascular endothelial growth factor, thereby enhancing cell proliferation, invasion and angiogenesis (117). ERG can also repress DNA repair genes such as BRCA1, leading to impaired repair capacity and increased genomic instability (118). Notably, PTEN deletions co-occur in 30-40% of TMPRSS2-ERG-positive tumors, acting synergistically to drive cancer progression and enzalutamide resistance (119). By contrast, SPOP-mutated prostate cancer types exhibit a markedly lower TMPRSS2-ERG fusion rate (<10%), indicating mutually exclusive oncogenic pathways among PCa molecular subtypes (120).
Recent genomic studies have expanded understanding of TMPRSS2-ERG-associated oncogenesis (121,122). Large-scale genome-wide association studies have identified additional susceptibility loci that may interact with known genes such as BRCA1/2, broadening the scope of genetic risk assessment in PCa (123). Furthermore, emerging domestic studies highlight that the TMPRSS2-ERG fusion can regulate PCa cell behavior through modulation of non-coding RNAs, suggesting novel avenues for targeted therapy and improved understanding of PCa pathogenesis (124,125) (Table I).
PCA3 is a lncRNA highly overexpressed (10-100 fold) in PCa tissue (126,127). Its level in urine collected after digital rectal examination is quantified as a PCA3 score, which aids in diagnosis, especially within the PSA 'gray zone' (4-10 ng/ml) (128,129). The PCA3 score is calculated using the formula: PCA3 score=(PCA3 copy number/PSA copy number) × 100. A score >35 suggests a high probability of cancer, helping to reduce unnecessary biopsies while maintaining high sensitivity (130). Specificity can be affected by prostatitis (131).
Expression of ERG mRNA, a product of the TMPRSS2-ERG fusion, and SPINK1 mRNA, often overexpressed in fusion-negative types of cancer, provides complementary diagnostic and subtyping information (132-134). miRNAs such as miR-141 and miR-375 in blood or urine show promise as non-invasive biomarkers (135,136). Panels combining multiple RNA markers (such as PCA3, ERG and miRNAs) have demonstrated superior diagnostic accuracy over single-marker tests (137,138). Furthermore, specific miRNA expression patterns are implicated in modulating the tumor immune microenvironment, suggesting potential for guiding immunotherapy strategies (139,140).
ctDNA, released into the bloodstream by tumor cells, provides a non-invasive means to assess tumor genetics and heterogeneity (141-143). Unlike prostate needle biopsies, which are invasive and subject to sampling bias due to tumor heterogeneity, ctDNA testing provides a non-invasive method to detect genetic alterations throughout the body, offering a more comprehensive outlook of tumor characteristics (144,145). This makes ctDNA testing particularly valuable for monitoring patients with castration-resistant prostate cancer or those with inaccessible metastatic lesions (146,147).
In mCRPC, ctDNA analysis enables real-time monitoring of AR mutations (such as T878A and H875Y) that confer resistance to abiraterone and enzalutamide, allowing for dynamic treatment adjustments (148). Of all patients with mCRPC, ~20% harbor mutations in DNA repair genes (such as BRCA1/2 and ATM) detectable in ctDNA, which predict responsiveness to PARP inhibitors (149). ctDNA testing offers a non-invasive method to detect these mutations, helping physicians identify candidates for PARP inhibitor-targeted therapy (150). Monitoring the abundance of drug-resistant mutations in ctDNA, such as AR mutations, can provide early indications of tumor progression, enabling timely adjustments to treatment regimens (151). ctDNA dynamics also serve as a sensitive marker for minimal residual disease and early recurrence after radical prostatectomy (152,153). Emerging applications include using ctDNA methylation profiles to predict responses to immunotherapy (154,155), with ongoing clinical trials validating its role in precision oncology (Fig. 2) (156).
Exosomes are extracellular vesicles (30-150 nm) that carry nucleic acids, including RNAs, from their cell of origin, reflecting the physiological and pathological state (157,158). In cancer, the RNA profiles in exosomes undergo specific alterations that reflect tissue damage or malignant transformation (159,160). For instance, miR-122 in exosomes from liver cancer cells has been identified as a diagnostic marker for liver cancer, while Aβ-related mRNA levels in exosomes from cerebrospinal fluid can indicate Alzheimer's disease (161).
In PCa, exo RNA holds considerable diagnostic and therapeutic potential (162). RNA secreted by PCa cells may contribute to tumor development, metastasis and progression (163). Certain miRNAs, such as miR-141 and miR-375, show altered expression in urinary exosomes of patients with PCa, which could serve as novel non-invasive biomarkers for early PCa detection (164).
Beyond diagnostics, exosomes are being engineered as therapeutic vehicles for the targeted delivery of siRNA or miRNA to tumor cells, showing efficacy in preclinical models (165). For example exosomes loaded with anti-miR-21 have been used to inhibit metastasis in breast cancer and similar strategies are under investigation for PCa (166,167). Additionally, blocking the release of metastasis-promoting miRNAs (such as miR-10b) in exosomes from tumor cells has been shown to inhibit cancer cell metastasis in lung cancer models, offering a potential strategy for preventing PCa metastasis by interfering with exo RNA function (Table II) (168).
Moreover, the regulation of exo lncRNA also presents novel avenues for interfering with cancer proliferation and invasion (169). In preclinical studies, some researchers have successfully developed exosome carriers specifically targeting PCa cells and loaded them with anti-cancer RNA molecules (170-172). These carriers have demonstrated promising effects in inhibiting tumor growth and metastasis in animal models, providing new hope for targeted PCa therapies (172). Similarly, some studies have analyzed exo RNA profiles at different stages of PCa, revealing that certain RNA characteristics can accurately determine disease stage and prognosis, which can guide clinicians in formulating more precise treatment plans (Fig. 3) (172,173).
Citric acid and polyamines have attracted increasing attention as metabolic biomarkers for PCa diagnosis and management (173). Citric acid, abundantly present in prostatic fluid, blood and tissues, serves as an indicator of prostate metabolism (174). Reduced citric acid levels are typically associated with malignant transformation, offering potential diagnostic utility in differentiating benign from malignant lesions (175). The use of magnetic resonance spectroscopy (MRS) enables non-invasive quantification of citric acid, providing valuable metabolic information for clinical decision-making (176). However, its diagnostic sensitivity remains limited when used alone and combination with other biomarkers such as PSA improves accuracy and specificity (177).
Polyamines, such as putrescine, spermidine and spermine-also show association with tumor aggressiveness and metastatic potential (178,179). Elevated polyamine concentrations in serum or urine may indicate enhanced tumor invasiveness and poor prognosis (180). Monitoring polyamine profiles could therefore assist in predicting disease progression and evaluating treatment responses. Nevertheless, benign conditions such as inflammation can modestly elevate polyamine levels, limiting diagnostic specificity (181).
From a therapeutic perspective, abnormal metabolism of citric acid and polyamines represents a promising target for intervention (182). Strategies aiming to restore mitochondrial metabolism or inhibit ornithine decarboxylase, a key enzyme in polyamine synthesis, have demonstrated potential in enhancing endocrine therapy and overcoming drug resistance (182,183). Recent metabolomics-based studies have identified urinary derivatives such as N1-acetylputrescine, which, when analyzed together with citric acid metabolites, notably improve early screening sensitivity (184,185). Integration of MRS-derived citric acid signals and serum polyamine levels with machine learning algorithms has further enhanced diagnostic precision, particularly for small lesions (186,187). Cross-ethnic studies have revealed distinct metabolic profiles of citric acid and polyamines, suggesting that population-specific diagnostic thresholds may improve biomarker accuracy (188,189). Moreover, emerging evidence indicates that gut microbial metabolites can modulate prostate metabolism by influencing citric acid and polyamine pathways, offering new insights into prostate carcinogenesis and potential microbiota-targeted therapies (189).
Basal cell markers p63 and 34βE12 are indispensable tools in PCa histopathology (190). In normal glands, basal cells form a continuous layer expressing nuclear p63 and cytoplasmic 34βE12 (191-193). Their absence is a hallmark of adenocarcinoma, reflecting the replacement of normal basal epithelium by malignant cells (194,195).
Compared with p63, which is nuclear and occasionally affected by tissue fixation, 34βE12 provides stable cytoplasmic staining, allowing reliable detection even in suboptimally preserved specimens (196,197). During prostate biopsy evaluation, the concurrent loss of both markers indicates carcinoma, with diagnostic sensitivity >95% and specificity >90% (198-200). When basal cells appear partially positive, additional markers such as PSA or α-methylacyl-CoA racemase are used to confirm malignancy (201,202).
Recent advances combine p63 and 34βE12 immunostaining with AI-assisted pathology, markedly improving interpretive accuracy and reducing observer variability (203,204). Single-cell sequencing and organoid models have further elucidated the dynamic expression of these markers across tumor grades, associating their loss with malignancy progression, recurrence risk and treatment outcomes (205-207).
DNA methylation represents one of the earliest and most consistent molecular alterations in PCa. Among the numerous genes identified, GSTP1 promoter methylation is the most extensively validated (208). GSTP1 encodes glutathione S-transferase, an enzyme involved in detoxification and oxidative defense. Hypermethylation-induced silencing of GSTP1 occurs in >90% of PCa cases but rarely in benign tissues, making it a highly specific diagnostic indicator (209). Combined detection of GSTP1 methylation with urinary PCA3 enhances diagnostic specificity >90% (210).
Other genes such as APC, RASSF1A, PTEN and CDH1 also demonstrate marked clinical relevance (211). APC promoter methylation is observed in 50-70% of cases and is associated with tumor progression and lymph node metastasis (212). RASSF1A methylation frequently co-occurs with GSTP1 and helps detect high-grade tumors with greater accuracy (213). PTEN methylation activates the PI3K/AKT pathway and contributes to castration resistance (214), while CDH1 and SFRP2 methylation associate with increased invasiveness and disease progression (Table III) (215).
Genome-wide methylation profiling has recently identified novel PCa-related epigenetic signatures with potential diagnostic and prognostic applications (216). Combining methylation markers with genetic and RNA-based assays, particularly in liquid biopsy formats, offers a more comprehensive and non-invasive diagnostic strategy (217). Studies also indicate that non-coding RNAs may regulate methylation marker expression, adding another layer of complexity to prostate tumorigenesis (218,219). These findings highlight the promise of methylation-based diagnostics and their integration into precision oncology (Fig. 4) (219).
The convergence of genetic, transcriptomic and metabolic biomarkers has fundamentally transformed PCa research and clinical management. Genetic alterations-such as BRCA1/2, HOXB13 and TMPRSS2-ERG fusions-enable precise risk stratification and therapeutic guidance (220). RNA-based biomarkers, including PCA3, ERG mRNA and miRNA panels, have established robust, non-invasive tools for early detection and disease monitoring (221). Metabolic biomarkers such as citric acid and polyamines complement these findings by reflecting tumor aggressiveness and metabolic reprogramming (222). Epigenetic and liquid biopsy markers, particularly DNA methylation (such as GSTP1 and APC) and circulating nucleic acids (ctDNA, exo RNA), are emerging as cornerstones of precision diagnostics (223). Their integration into clinical workflows enables dynamic disease monitoring and individualized therapy optimization (224).
Future research should focus on multi-omics integration-combining genomic, transcriptomic, methylomic and metabolomic data to construct comprehensive biomarker networks (225). Incorporating AI and machine learning will further enhance diagnostic accuracy and predict therapeutic outcomes (226). Additionally, investigating population-specific biomarker variations and tumor microenvironment interactions will be vital for developing equitable, personalized care strategies (227). Ultimately, the continued refinement and clinical validation of these emerging biomarkers will usher in a new era of PCa precision medicine, enabling earlier detection, tailored therapy and improved survival and quality of life for patients worldwide (Fig. 5) (228).
Not applicable.
YH, JM and XL contributed to the writing of the original draft and the preparation of figures. YH contributed to the data analysis. JM and XL provided writing review and editing, as well as supervision. All authors read and approved the final manuscript. Data authentication not applicable.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
The authors would like to express their sincere gratitude to Mr. Zhenjun Xi and Mr. Li Wang from Chongqing Medical and Pharmaceutical College, Chongqing, China for their invaluable contributions to the manuscript; their meticulous efforts ensured the accuracy and reliability of the research findings of the present study.
The authors declare that financial support was received for the research, authorship and publication of this article. This work was funded by 2023 Chongqing Medical Scientific Research project (Joint project of Chongqing Health Commission and Science and Technology Bureau; grant no. 2023GGXM006), 2024 Scientific Research project of Chongqing Medical and Pharmaceutical College (grant no. ygzrc2024101), Chongqing Education Commission Natural Science Foundation (grant no. KJQN202402821; grant no. KJQN202502819) and 2024 Chongqing Medical and Pharmaceutical College Innovation Research Group Project (grant no. ygz2024401), Chongqing Science and Health Joint Medical Research Project (grant no. 2024SQKWLHMS051), Natural Science Foundation of Chongqing (grant no. CSTB2025NSCO-GPX1116) and Technological Innovation Project of Shapingba District, Chongqing (grant no. 2025016, 2025017 and 2025031), Scientific Research Project of AnShun University, Guizhou Province [grant no. asxybsjj (202307) and asxykypt (202402)] and Natural Science Research Project of Guizhou Provincial Department of Education [Qian Jiao He KY (2020) grant no. 063] respectively.
|
Kratze TB, Mazzitelli N, Star J, Dahut WL, Jemal A and Siegel RL: Prostate cancer statistics, 2025. CA Cancer J Clin. 75:485–497. 2025. | |
|
Braga R, Morais S, Pacheco-Figueiredo L, Araujo N and Lunet N: Prostate cancer screening: Knowledge, attitudes, and practices by medical doctors in portugal. J Cancer Educ. Jun 19–2025.Epub ahead of print. View Article : Google Scholar : PubMed/NCBI | |
|
Takashima Y, Yoshii K, Tanaka M and Tashiro K: Ubiquitin-proteasome pathway-linked gene signatures as prognostic indicators in prostate cancer. Anticancer Res. 45:1825–1841. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Elgehama AM, Yang Q, He Z, Ruegg L, You S and Yang W: Identification of a RIPK2-regulated gene signature as a candidate biomarker for RIPK2 activity and prognosis in prostate cancer. bioRxiv [Preprint]. 2025.04.30.651490. 2025.PubMed/NCBI | |
|
Mellor J, Hunter E and Akoulitchev A: Paradigm lost. Cancers (Basel). 17:21872025. View Article : Google Scholar : PubMed/NCBI | |
|
Wilkosz J, Sobieraj DW, Kaluzewski T, Kaczmarek J, Szwalski J, Bednarek M, Morel A, Kasprzyk Z, Kepczynski L, Salamunia J, et al: Personalized high-resolution genetic diagnostics of prostate adenocarcinoma guided by multiparametric magnetic resonance imaging: Results of a pilot study. Int J Mol Sci. 26:56482025. View Article : Google Scholar : PubMed/NCBI | |
|
Wang X, Waldman L, Silberman Y, Wang M, Tackey C, Hanna L, Vesprini D, Emmenegger U, Eisen A and Smoragiewicz M: Mainstream model of genetic testing for prostate cancer at a large tertiary cancer centre. Clin Genitourin Cancer. 22:1020522024. View Article : Google Scholar : PubMed/NCBI | |
|
Gonzalez-Peramato P, Alvarez-Maestro M, Heredia-Soto V, Mendiola Sabio M, Linares E, Serrano A, Alvarez-Ossorio JL, Lopez Alcina E, Prieto L, Vazquez Alonso F, et al: Comparing prostatype P-score and traditional risk models for predicting prostate cancer outcomes in Spain. Actas Urol Esp (Engl Ed). 49:5017882025.PubMed/NCBI | |
|
Stella M, Russo GI, Leonardi R, Carco D, Gattuso G, Falzone L, Ferrara C, Caponnetto A, Battaglia R, Libra M, et al: Extracellular RNAs from whole urine to distinguish prostate cancer from benign prostatic hyperplasia. Int J Mol Sci. 25:100792024. View Article : Google Scholar : PubMed/NCBI | |
|
Pattou M, Neuzillet Y, Ghoneim T, Bosset PO, Herve JM, Vanalderwerelt V, Bohin D, Lugagne PM, Soorojebally Y and Lebret T: PSA density correlates to pathology T stage and ISUP grade: Insights from a cohort of 3568 radical prostatectomy cases. World J Urol. 43:4452025. View Article : Google Scholar : PubMed/NCBI | |
|
Alahdal M, Perera RA, Moschovas MC, Patel V and Perera RJ: Current advances of liquid biopsies in prostate cancer: Molecular biomarkers. Mol Ther Oncolytics. 30:27–38. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Traphagen NA, Wheeler E, Li R, Lu F, Ahmed B, Tewari AK, Balk SP, Nelson PS, Corey E, Long H, et al: Lack of synergy between AR targeted therapies and PARP inhibitors in homologous recombination-proficient prostate cancer. bioRxiv [Preprint]. 2025.06.02.657429.2025. | |
|
Castellnou P, Gomez-Martinez M, Gomez-Vallejo V, Baz Z, Lopez-Gallego F, Rondon-Lorefice I, Zabala-Letona A, Poot AJ, Mendizabal I, Carracedo A, et al: Unravelling the role of L- and D-alanine in prostate cancer: A positron emission tomography study in a genetic mouse model. Nucl Med Biol. 148-149:1090482025. View Article : Google Scholar | |
|
Coradduzza D, Sanna A, Di Lorenzo B, Congiargiu A, Marra S, Cossu M, Tedde A, De Miglio MR, Zinellu A, Mangoni AA, et al: Associations between plasma and urinary heavy metal concentrations and the risk of prostate cancer. Sci Rep. 15:142742025. View Article : Google Scholar : PubMed/NCBI | |
|
Mitura P, Paja W, Mlynarczyk G, Kowalski R, Bar K and Depciuch J: Urine-based Raman markers for prostate cancer diagnosis: A machine learning approach using fingerprint and lipid spectral region. Spectrochim Acta A Mol Biomol Spectrosc. 344(Pt 1): 1266612026. View Article : Google Scholar | |
|
Sharma D, Mitra D, Bansal VK and Singh KV: Study of the correlation between multi-parametric MRI (MP-MRI) prostate findings and transrectal ultrasound (TRUS)-guided prostate biopsy results in patients with raised serum PSA. Cureus. 17:e862162025.PubMed/NCBI | |
|
Huang S, Hart JC, Smith JF, Bench S, Yepes LR, Griscom B and Clark-Langone KM: Tissue of origin characterization of cell free DNA in seminal plasma: Implications for new liquid biopsies. PLoS One. 20:e03177122025. View Article : Google Scholar : PubMed/NCBI | |
|
Mo H, Hu C, Li P, Miao Z, Zhang H, Shen G and Sun J: PSA proportion of index lesion: a novel index for predicting prostate pathology outcomes in biopsy-naive patients with PSA < 10 ng/ml and PI-RADS 3 lesions. World J Urol. 43:4442025. View Article : Google Scholar | |
|
Wang X, He Z, He X, Zeng R, He H, Zhang K and Xu Y: High-efficiency detection of Total-PSA and Free-PSA in whole blood by microfluidic chip integrated with electromagnetic co-preprocessing. ACS Sens. 10:3878–3887. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Arcay Ozturk A, Erkilic M, Bural GG, Aydin F and Boz A: Physiological biodistribution on Ga68-PSMA PET/CT and the factors effecting biodistribution. Ann Nucl Med. 38:894–903. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Schulze A, Christoph F, Sachs M, Schroeder J, Stephan C, Schostak M and Koenig F: Use of the prostate health index and density in 3 outpatient centers to avoid unnecessary prostate biopsies. Urol Int. 104:181–186. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Y, Chang Y, Song L, Ren J, Yin L, Cao W, Xu W and Ren S: Robotic prostatectomy for large-volume prostates in prostate cancer: A retrospective analysis of 50 cases (>100 ml). BMC Cancer. 25:7012025. View Article : Google Scholar | |
|
Zheng B, Mo F, Shi X, Li W, Shen Q, Zhang L, Liao Z, Fan C, Liu Y, Zhong J, et al: An automatic deep-radiomics framework for prostate cancer diagnosis and stratification in patients with serum prostate-specific antigen of 4.0-10.0 ng/mL: A multicenter retrospective study. Acad Radiol. 32:2709–2722. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Qiu L, Tian T, Ma Z, Sun J, Zhao Y, Yang L, Ji Y, Yi L, Yan L, Xu C and Li D: Predictive value of the pretreatment serum sialic acid/total protein ratio for bone metastases in newly diagnosed prostate cancer patients: development of a nomogram model. Gland Surg. 14:1066–1078. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Rastogi S, Sato N, Lee S, Lee MJ, McKinney Y, Lindenberg L, Mena E, Thomas A, Shrestha RL and Choyke PL: Circulating tumor cells are detectable and independent of PSA and PSMA-PET metrics in localized high-risk and biochemically recurrent prostate cancer. medRxiv [Preprint]. 2025.07.09.25331014. 2025.PubMed/NCBI | |
|
Fan YP, Zhang YT, Zhang G, Ma L, Lv Y, Li J, Luan Y, Zhang YX, Chen YT, Ren HY, et al: Proteomic profiling of urinary large extracellular vesicles for the diagnosis of prostate cancer. Anal Chem. 97:17368–17379. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Chen D, Ma Y, Yang A, Hu L, Zhou H, Xu J, Chen S, Nie D, Feng W, Cai H, et al: Dual-Enhanced SERS satellite immuno-nanocomplex for multiple PSA-Mediated PHI assay toward clinical prostate cancer screening. Adv Sci (Weinh). 12:e24117472025. View Article : Google Scholar | |
|
Yao W, Wu J, Kong Y, Xu F, Zhou Y, Sun Q, Gao Q, Cai Z, Yang C and Huang Y: Associations of systemic immune-inflammation index with high risk for prostate cancer in middle-aged and older US males: A population-based study. Immun Inflamm Dis. 12:e13272024. View Article : Google Scholar : PubMed/NCBI | |
|
Hochma E, Firer MA and Minnes R: Near-infrared and sono-enhanced photodynamic therapy of prostate cancer cells using phyto-second harmonic generation nanoconjugates. Polymers (Basel). 17:18312025. View Article : Google Scholar : PubMed/NCBI | |
|
Luo B, Zhou J, Zhan X, Ying B, Lan F and Wu Y: Smartphone-based free-to-total prostate specific antigen ratio detection system using a colorimetric reaction integrated with proximity-induced bio-barcode and CRISPR/Cas12a assay. Small. 20:e23102122024. View Article : Google Scholar : PubMed/NCBI | |
|
Serkan Y, Caner E, Serkan A, Bulent K, Yasin V, Adem A and Omer Y: Significance of inflammation markers to predict curative treatment for prostate cancer patients on active surveillance. J Clin Lab Anal. 39:e700592025. View Article : Google Scholar : PubMed/NCBI | |
|
Stojadinovic M, Jurisevic N, Stojadinovic M and Jankovic S: Predictive clinical characteristics for adverse pathological outcomes in intermediate- and high-risk prostate cancer during biopsy. Int Urol Nephrol. 57:4077–4086. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Yorioka MAW, Murta CB, Leite KRM, Cardili L, de Mello ES, de Carvalho Fazoli AJ, Cordeiro MD, Coelho RF, Viana PCC, Kohama CSN, et al: ERG and PTEN role on active surveillance for low-risk prostate cancer in the multiparametric MRI Era. Prostate. 85:364–373. 2025. View Article : Google Scholar | |
|
Weng Y and Ji B: Serum levels of prostate specific antigen, free PSA, (-2)proPSA, fPSA/tPSA ratio, prostate health index, and glycosylation patterns of free PSA in patients with benign prostatic hyperplasia pharmacotherapy: Limitations and future directions. Prostate. 85:1248–1249. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Encarnacion Navarro JA, Morillo Macias V, Borras Calbo M, De la Fuente Munoz I, Lozano Martinez A, Garcia Martinez V, Fernandez Fornos L, Guijarro Roche M, Amr Rey O and Garcia Gomez R: Multicenter real-world study: 432 patients with apalutamide in metastatic hormone-sensitive prostate cancer. Curr Oncol. 32:1192025. View Article : Google Scholar : PubMed/NCBI | |
|
Gjelsvik YM, Myklebust TA, Fosså SD, Haug ES, Kvåle R, Ursin G and Johannesen TB: A nationwide, longitudinal collection of patient-reported outcomes from prostate cancer patients and controls. Qual Life Res. 34:2689–2700. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Zeng Y, Liao H, Jiang G, Wu G and Zhong J: Diagnostic value of 18F-prostate-specific membrane antigen-1007 PET/MRI versus 18F-prostate-specific membrane antigen-1007 PET/computed tomography for biochemical recurrence of prostate cancer after radical prostatectomy. Nucl Med Commun. 46:1052–1060. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Williams N, Remmers S, Jay A, Santoro K and O'Callaghan M: External validation of MRI-based prostate cancer risk calculators in an Australian cohort. ANZ J Surg. 95:1906–1911. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Van der Eecken H, De Cock D, Roussel E, Giesen A, Vansevenant B, Goeman L, Quackels T and Joniau S: Nutritional supplement with fermented soy in patients under active surveillance for low-risk or intermediate-risk prostate cancer: Results from the PRAEMUNE trial. Cancers (Basel). 16:36342024. View Article : Google Scholar : PubMed/NCBI | |
|
Guo P, A G, Chen T, Guo Y, Tang Y, Pan J, Wang B, Gong R, Chen G and Huang S: A hematological and inflammatory marker-based model for prostate carcinoma diagnosis. Am J Cancer Res. 15:2551–2563. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Cirulli GO, Davis M, Stephens A, Chiarelli G, Finati M, Chase M, Tinsley S, Arora S, Sood A, Lughezzani G, et al: Midlife baseline prostate-specific antigen, velocity, and doubling time association with lethal prostate cancer and mortality. Cancer. 131:e355632025. View Article : Google Scholar | |
|
Kobayashi M, Kijima T, Yashi M and Kamai T: Prostate-specific antigen kinetics contributes to decision making for biopsy referral: the predictive implication for PSA retest in patients with elevated PSA levels. Prostate Int. 11:27–33. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Javaeed A, Ghauri SK, Ibrahim A and Doheim MF: Prostate-specific antigen velocity in diagnosis and prognosis of prostate cancer - a systematic review. Oncol Rev. 14:4492020. View Article : Google Scholar : PubMed/NCBI | |
|
Ono K, Hiyoshi Y, Ono A, Ouchi M, Kosumi K, Eto K, Ida S, Iwatsuki M, Baba Y, Miyamoto Y, et al: Locally advanced rectal cancer in a young adult affected with dyskeratosis congenita (Zinsser-Cole-Engman syndrome): A case report. Surg Case Rep. 10:2062024. View Article : Google Scholar : PubMed/NCBI | |
|
Dai F, He Y, Duan J, Lin K, Lv Q, Zhao Z, Zou Y, Jiang J, Zheng Z and Qiu X: Global trends in the use of artificial intelligence for urological tumor histopathology: A 20-year bibliometric analysis. Digit Health. 11:205520762513488342025. View Article : Google Scholar : PubMed/NCBI | |
|
Aktas S, Yucetas U, Yucetas E, Ates HA, Gene C and Erkan E: The efficacy of prostate health index (PHI) in predicting pathological progression in low-risk localized prostate cancer cases under active surveillance protocol. World J Urol. 43:4312025. View Article : Google Scholar : PubMed/NCBI | |
|
Assam KD, Pierce JM, Galloway LA and Tosoian JJ: Blood- and urine-based biomarkers for the detection of clinically significant prostate cancer: A contemporary review. Curr Opin Urol. 35:590–596. 2025. View Article : Google Scholar | |
|
Gupta VK, Cortese BD and Talwar R: Cost-effectiveness of serum, urine, and tissue-based prostate cancer biomarkers. Curr Opin Urol. 35:412–417. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Jeon SS, Song W, Kang M, Sung HH, Jeon HG, Jeong BC, Seo SI and Chung JH: Diagnostic value of prostate health index in patients with no index lesion on mpMRI or negative previous combined biopsy. Investig Clin Urol. 66:124–129. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Jirasko M, Vitak R, Pecen L, Pinkeova A, Tkac J, Bertok T, Bergman N and Kucera R: Serum levels of prostate specific antigen, free PSA, (-2)proPSA, fPSA/tPSA ratio, prostate health index, and glycosylation patterns of free PSA in patients with benign prostatic hyperplasia pharmacotherapy. Prostate. 85:65–72. 2025. View Article : Google Scholar | |
|
Vakili S, Beheshti I, Barzegar Behrooz A, Los MJ, Vitorino R and Ghavami S: Transforming prostate cancer care: Innovations in diagnosis, treatment, and future directions. Int J Mol Sci. 26:53862025. View Article : Google Scholar : PubMed/NCBI | |
|
Britton CJ, Andrews JR, Arafa A, Kim Y, Latuche LR, Schulte PJ, Joshi VB, Ahmed ME, Jeffrey Karnes R and Lucien F: Prostate extracellular vesicles and prognostic biomarkers of clinically significant prostate cancer: A prospective single-institution pilot study. Prostate. 85:594–602. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Lee IT, Hou CM, Vo TTT, Hong JH, Huang CY, Wang YL, Chen YL and Chiang CH: Optimizing prostate cancer care: Clinical utility of the prostate health index. Prostate. 85:1357–1368. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Chung Y and Hong SK: Evaluating prostate cancer diagnostic methods: The role and relevance of digital rectal examination in modern era. Investig Clin Urol. 66:181–187. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Fasulo V, Chiarelli G, Garofano G, Ripamonti CB, Barile M, Bianchi P, Morenghi E, Benetti A, Aljoulani M and Finocchiaro A: Impact of prostate cancer screening in European ancestry un-affected men with germline DNA repair pathogenic variants. BJUI Compass. 6:e4242025. View Article : Google Scholar : PubMed/NCBI | |
|
Chang CH, Yu PH, Hsieh PF, Hong JH, Chiang CH, Cheng HM, Wu HC, Huang CY and Lin TP: Prostate health index density aids the diagnosis of prostate cancer detected using magnetic resonance imaging targeted prostate biopsy in Taiwanese multicenter study. J Chin Med Assoc. 87:678–685. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Stuardo-Parada A, Lopez-Munoz R, Villarroel-Espindola F, Figueroa CD and Ehrenfeld P: Minireview: Functional roles of tissue kallikrein, kinins, and kallikrein-related peptidases in lung cancer. Med Oncol. 40:2242023. View Article : Google Scholar : PubMed/NCBI | |
|
Kelam N, Ogorevc M, Gotovac I, Kuzmic Prusac I, Vukojevic K, Saraga-Babic M and Mardesic S: Analysis of kallikrein 6, Acetyl-α-tubulin, and aquaporin 1 and 2 expression patterns during normal human nephrogenesis and in congenital anomalies of the kidney and urinary tract (CAKUT). Genes (Basel). 16:4992025. View Article : Google Scholar | |
|
Sasiadek L, Bielecka E, Falkowski K, Kulczycka M, Bereta G, Maksylewicz A, Zubrzycka N, Dobosz E, Koziel J, Drukala J, et al: Human tissue kallikrein 14 induces the expression of IL-6, IL-8, and CXCL1 in skin fibroblasts through protease-activated receptor 1 signaling. FEBS J. 292(21): 5659–5675. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Giuffre M, Lista AD, Paulson N and Masuoka L: A phase 1C, open label, single ascending dose study to evaluate the safety, tolerability, and pharmacokinetics of DM199 administered intravenously with a polyvinyl chloride bag in adult healthy subjects and adults recently taking angiotensin-converting enzyme inhibitors. Clin Pharmacol Drug Dev. 14:452–460. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Singh SP, Dhanasekara CS, Melkus MW, Bose C, Khan SY, Sardela de Miranda F, Mahecha MF, Gukhool PJ, Tonk SS, Jun SR, et al: Relevance of cellular homeostasis-related gene expression signatures in distinct molecular subtypes of breast cancer. Biomedicines. 13:10582025. View Article : Google Scholar : PubMed/NCBI | |
|
Kumar N, Kole L and Singh AK: Precision assay for detection and quantification of anti-prostate specific antigen antibodies using indirect ELISA. Infect Disord Drug Targets. May 15–2025.Epub ahead of print. View Article : Google Scholar | |
|
Boumali R, David E, Chaaya N, Lucas M, Ait Amiri S, Lefort V, Nina-Diogo A, Salmain M, Petropoulos I, Corce V, et al: Deferasirox derivatives as inhibitors of kallikrein-related peptidases associated to neurodegenerative diseases. ChemMedChem. 20:e2025001872025. View Article : Google Scholar : PubMed/NCBI | |
|
Koistinen H, Kovanen RM, Hollenberg MD, Dufour A, Radisky ES, Stenman UH, Batra J, Clements J, Hooper JD, Diamandis E, et al: The roles of proteases in prostate cancer. IUBMB Life. 75:493–513. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Angel MR, Loeffler D, Charifa A, Sinit RB, Amery T, Cengiz B, Beer TM and Thomas GV: TMPRSS2 expression in lung tissue of prostatic adenocarcinoma patients: A pathologic perspective on androgen deprivation therapy. medRxiv [Preprint]. 2025.05.03.25326931. 2025.PubMed/NCBI | |
|
Srinivasan S, Kryza T, Batra J and Clements J: Remodelling of the tumour microenvironment by the kallikrein-related peptidases. Nat Rev Cancer. 22:223–238. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Stein MN, Vinceneux A, Robbrecht D, Doger B, Autio KA, Schweizer MT, Calvo E, Medina L, Van Dongen M, Deville JL, et al: Real-world effectiveness of darolutamide in metastatic castration-resistant prostate cancer. J Clin Oncol. 43:2515–2526. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao J, Sun Y, Ren L, Huang S and Zhang J: Antagonism of androgen receptor signaling by aloe-emodin. Food Chem Toxicol. 181:1140922023. View Article : Google Scholar : PubMed/NCBI | |
|
Boyukozer FB, Tanoglu EG, Ozen M, Ittmann M and Aslan ES: Kallikrein gene family as biomarkers for recurrent prostate cancer. Croat Med J. 61:450–456. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Wenta T, Nastaly P, Lipinska B and Manninen A: Remodeling of the extracellular matrix by serine proteases as a prerequisite for cancer initiation and progression. Matrix Biol. 134:197–219. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Matin F, Jeet V, Srinivasan S, Cristino AS, Panchadsaram J, Clements JA and Batra J; Australian Prostate Cancer BioResource: MicroRNA-3162-5p-mediated crosstalk between kallikrein family members including prostate-specific antigen in prostate cancer. Clin Chem. 65:771–780. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Nakayama H, Sekine Y, Oka D, Miyazawa Y, Arai S, Koike H, Matsui H, Shibata Y and Suzuki K: Combination therapy with novel androgen receptor antagonists and statin for castration-resistant prostate cancer. Prostate. 82:314–322. 2022. View Article : Google Scholar | |
|
Karan D, Dubey S, Gunewardena S, Iczkowski KA, Singh M, Liu P, Poletti A, Choo YM, Chen HZ and Hamann MT: Manzamine A reduces androgen receptor transcription and synthesis by blocking E2F8-DNA interactions and effectively inhibits prostate tumor growth in mice. Mol Oncol. 18:1966–1979. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Nordstrand A, Bovinder Ylitalo E, Thysell E, Jernberg E, Crnalic S, Widmark A, Bergh A, Lerner UH and Wikstrom P: Bone cell activity in clinical prostate cancer bone metastasis and its inverse relation to tumor cell androgen receptor activity. Int J Mol Sci. 19:12232018. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang J and Chadha JS: Developmental therapeutics in metastatic prostate cancer: New targets and new strategies. Cancers (Basel). 16:30982024. View Article : Google Scholar : PubMed/NCBI | |
|
Isaacs JT, Brennen WN, Christensen SB and Denmeade SR: Mipsagargin: The beginning-not the end-of thapsigargin prodrug-based cancer therapeutics. Molecules. 26:74692021. View Article : Google Scholar : PubMed/NCBI | |
|
Ma Z, Shi B, Wei Z, Guo W, Hu L, Li Y, Wu X, Gong S, Wu D and Wu C: Abstract 3520: EM1031, a novel KLK2 x CD3 bispecific T-cell engager with highly effective efficacy in preclinical models. Cancer Res. 85(8_Supplement_l): S35202025. View Article : Google Scholar | |
|
Shen F, Smith R, McDevitt T, Menard K, Tian S, Chu G, Chaudhary R, McCann J, Oyer H, Wang SC, et al: Human kallikrein 2: A novel lineage-specific surface target in prostate cancer. Clin Cancer Res. 31:4543–4556. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Vietri MT, Della Pepa C, Caliendo G, Mignano A, Albanese L, Zitiello M, Stilo M and Molinari AM: Expanding the genomic landscape of HBOC and cancer risk among mutation carriers. Int J Mol Sci. 26:59282025. View Article : Google Scholar : PubMed/NCBI | |
|
Nyberg T, Frost D, Barrowdale D, Evans DG, Bancroft E, Adlard J, Ahmed M, Barwell J, Brady AF, Brewer C, et al: Prostate cancer risks for male BRCA1 and BRCA2 mutation carriers: A prospective cohort study. Eur Urol. 77:24–35. 2020. View Article : Google Scholar : | |
|
Yatrano S, Pepe P, Pepe L, Vella N, Alario C, Chiaranda A, Taranto C, Scillieri R, Mauceri C and Fraggetta F: BRCA mutations and prostate cancer: should urologist improve daily clinical practice? Arch Ital Urol Androl. 97:136352025. | |
|
Fizazi K, Azad AA, Matsubara N, Carles J, Fay AP, De Giorgi U, Young Joung J, Fong PCC, Voog E, Jones RJ, et al: Talazoparib plus enzalutamide in men with HRR-deficient metastatic castration-resistant prostate cancer: Final overall survival results from the randomised, placebo-controlled, phase 3 TALAPRO-2 trial. Lancet. 406:461–474. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Amini AE, Hunter AE, Almashad A, Feng AJ, Patel ND, O'Dea MR, McCormick SR, Rodgers LH and Salari K: Magnetic resonance imaging-based prostate cancer screening in carriers of pathogenic germline mutations: Interim results from the initial screening round of the prostate cancer genetic risk evaluation and screening study. Eur Urol Oncol. 7:1358–1366. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Li X and Mao J: Research progress on the role of lipoxygenase and its inhibitors in prostate cancer. Future Oncol. 20:3549–3568. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Santos-Pereira M, Pereira SC, Matos B, Fardilha M, Oliveira PF and Alves MG: Increased susceptibility to prostate cancer biomarkers in the offspring of male mouse progenitors with lifelong or early life exposure to high-fat diet. Eur J Nutr. 64:2122025. View Article : Google Scholar : PubMed/NCBI | |
|
Biyikoglu M, Tanriverdi R, Bozlu M, Send S, Fidanci SB, Tamer L and Akbay E: Evaluation of homeobox protein B13 (HOXB13) gene G84E mutation in patients with prostate cancer. World J Urol. 42:4762024. View Article : Google Scholar : PubMed/NCBI | |
|
Sulaiman KM and Hama Salih RM: Study of HOXB13 gene variants in prostate cancer patients. Cureus. 16:e725132024.PubMed/NCBI | |
|
Barashi NS, Li T, Angappulige DH, Zhang B, O'Gorman H, Nottingham CU, Shetty AS, Ippolito JE, Andriole GL, Mahajan NP, et al: Symptomatic benign prostatic hyperplasia with suppressed epigenetic regulator HOXB13 shows a lower incidence of prostate cancer development. Cancers (Basel). 16:2132024. View Article : Google Scholar : PubMed/NCBI | |
|
de Angelis M, Siech C, Jannello LMI, Di Bello F, Penaranda NR, Scilipoti P, Goyal JA, Tian Z, Longo N, de Cobelli O, et al: Does race/ethnicity affect secondary bladder and rectal cancer rates after prostate cancer radiation? Results from a population-based study. J Racial Ethn Health Disparities. Jul 11–2025.Epub ahead of print. View Article : Google Scholar | |
|
Wei J, Shi Z, Na R, Wang CH, Resurreccion WK, Zheng SL, Hulick PJ, Cooney KA, Helfand BT, Isaacs WB and Xu J: Germline HOXB13 G84E mutation carriers and risk to twenty common types of cancer: Results from the UK Biobank. Br J Cancer. 123:1356–1359. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Chartier J, Chansavang A, Jouinot A, Hamzaoui N, Srikaran A, Moliere D, Huillard O, Thibault C, Tiako M, Sibony M, et al: Predisposition to prostate cancer and clinical implications in a real-life cohort. Eur J Hum Genet. 33:1163–1172. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Fraser M, Livingstone J, Wrana JL, Finelli A, He HH, van der Kwast T, Zlotta AR, Bristow RG and Boutros PC: Somatic driver mutation prevalence in 1844 prostate cancers identifies ZNRF3 loss as a predictor of metastatic relapse. Nat Commun. 12:62482021. View Article : Google Scholar : PubMed/NCBI | |
|
Poon DM, Yuan J, Wong OL, Yang B, Chiu ST, Cheung KY, Chiu G and Yu SK: Stereotactic total ablative radiotherapy with MR-LINAC for synchronous oligometastatic prostate cancer. Front Oncol. 15:16076102025. View Article : Google Scholar : PubMed/NCBI | |
|
Garcia de Herreros M, Jimenez N, Padrosa J, Aversa C, Ferrer-Mileo L, Garcia-Esteve S, Rodriguez-Carunchio L, Trias I, Fernandez-Manas L, Marin-Aguilera M, et al: Clinical and transcriptomic characterization of metastatic hormone-sensitive prostate cancer patients with low PTEN expression. Int J Mol Sci. 26:62442025. View Article : Google Scholar : PubMed/NCBI | |
|
Einspieler H, Ofner H, Ozenil M, Spielvogel CP, Langrate IK, Hassler MR, Nics L, Bamminger K, Baltzer PAT, Shariat SF, et al: Does PARP1 up-regulation correlate with PSMA expression in patients with metastatic castration-resistant prostate cancer studied with ((18)F)PARPi and ((68)Ga) PSMA PET/CT? Eur J Nucl Med Mol Imaging. July 19–2025.Epub ahead of print. View Article : Google Scholar | |
|
Handa N, Li EV, Michael J, Proudfoot JA, Weiner AB, Alam R, Alshalalfa M, Hao Y, Hakansson A, Zhao X, et al: Prevalence of potential candidates for targeted therapies according to treatment-related transcriptomic signatures among 140 548 patients with nonmetastatic prostate cancer. Eur Urol Oncol. 8:1050–1058. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Yin L, Yang F, Wang W, Zhang L, Cao Z, Shi H, Pan K, Wu L, Xiao H and Xing N: PSMA-Targeted nanoparticles with PI3K/mTOR dual inhibitor downregulate P-Glycoprotein and inactivate myeloid-derived suppressor cells for enhanced chemotherapy and immunotherapy in prostate cancer. Adv Mater. 37:e24153222025. View Article : Google Scholar : PubMed/NCBI | |
|
Miyachi S, Sasaki T, Kato M, Higashi S, Masui S, Nishikawa K, Fujiwara T, Kitajima T, Hashizume Y, Uchida K, et al: Comprehensive genomic profiling tests for Japanese patients with metastatic castration-resistant prostate cancer: A single-institution experience. Hinyokika Kiyo. 71:171–179. 2025.In Japanese. PubMed/NCBI | |
|
Giantini-Larsen A, Ramos AD, Martin D, Panageas KS, Kostrzewa CE, Abou-Mrad Z, Schmitt A, Bromberg JF, Safonov A, Rudin CM, et al: Integration of next generation sequencing data to inform survival prediction of patients with spine metastasis. Cancers (Basel). 17:22182025. View Article : Google Scholar : PubMed/NCBI | |
|
Kldiashvili E, Abiatari I, Kekelia E, Iordanishvili S, Metreveli T and Dumbadze E: SOX2, PIWI proteins, and MALAT1 - plasma-based emerging biomarkers for cancer detection and monitoring. PLoS One. 20:e03285572025. View Article : Google Scholar : PubMed/NCBI | |
|
Yazaki S, Pei X, Powell S, Khan A, Setton J and Riaz N: AlphaMissense for identifying pathogenic missense mutations in DNA damage repair genes in cancer. JCO Precis Oncol. 9:e24009082025. View Article : Google Scholar : PubMed/NCBI | |
|
Hommerding M, Hommerding O, Bernhardt M, Kreft T, Sanders C, Tischler V, Basitta P, Pelusi N, Wulf AL, Ohlmann CH, et al: Real-world data on the prevalence of BRCA 1/2 and HRR gene mutations in patients with primary and metastatic castration resistant prostate cancer. World J Urol. 42:4912024. View Article : Google Scholar | |
|
Kwon A and Joung JY: Precision targeting in metastatic prostate cancer: molecular insights to therapeutic frontiers. Biomolecules. 15:6252025. View Article : Google Scholar : PubMed/NCBI | |
|
Naisam S, Mohan A, Sivakumar GS, Gopalakrishnan S and Sreekumar N: Discovering potential therapeutic agents for lupus nephritis: Insights from in silico research. Medinformatics. 2:226–240. 2025. | |
|
Norgaard M, Rusan M, Kondrup K, Sorensen EMG, Weiss S, Bjerre MT, Fredsoe J, Vang S, Jensen JB, De Laere B, et al: Deep targeted sequencing of circulating tumor DNA to inform treatment in patients with metastatic castration-resistant prostate cancer. J Exp Clin Cancer Res. 44:1202025. View Article : Google Scholar : PubMed/NCBI | |
|
Dhabhai B, Dhakar R, Ranga V, Surolia P, Menon AM, Sharma NK, Dakal TC and Lohar D: Integrated bioinformatics approach showed linagliptin as potential drug for prevention of cardiac arrest and cancer. Medinformatics. 1:131–141. 2023. View Article : Google Scholar | |
|
Yoon CE, Kang S, Rhew SA, Kwon HJ, Shin D, Moon HW, Kim MY and Lee JY: Genetic alterations of prostate cancer: In localized and metastatic prostate cancer. BMC Urol. 25:1662025. View Article : Google Scholar : PubMed/NCBI | |
|
Wierbilowicz K, Yang CS, Almaghasilah A, Wesolowski PA, Pracht P, Dworak NM, Masur J, Wijngaarden S, Filippov DV, Wales DJ, et al: Parp7 generates an ADP-ribosyl degron that controls negative feedback of androgen signaling. EMBO J. 44:4720–4744. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Fragkoulis C, Glykas I, Tzelves L, Stamatakos PV, Papadopoulos G, Stathouros G, Dellis A, Ntoumas K, Kostopoulou A, Deliveliotis C and Papatsoris A: Clinical impact of ERG and PTEN status in prostate cancer patients underwent radical prostatectomy. Arch Ital Urol Androl. 94:390–395. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Lorenzoni M, De Felice D, Beccaceci G, Di Donato G, Foletto V, Genovesi S, Bertossi A, Cambuli F, Lorenzin F, Savino A, et al: ETS-related gene (ERG) undermines genome stability in mouse prostate progenitors via Gsk3β dependent Nkx3.1 degradation. Cancer Lett. 534:2156122022. View Article : Google Scholar | |
|
Ionescu CA, Cozaru GC, Aschie M, Leopa N, Cimpineanu B, Voinea F, Matei E, Mitroi A, Deacu M, Iorga I and Pundiche M: Toward personalized surgery in advanced prostate cancer: Stratification by PTEN, AR-V7, TP53, TMPRSS2-ERG, and ERBB2 genetic alterations. Chirurgia (Bucur). 120:265–274. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Check MH, Ernst SE and Sfanos KS: Use of droplet digital PCR for consistent detection of TMPRSS2:ERG gene fusion transcripts initiated in vitro. Prostate. 85:1282–1289. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Raspin K, O'Malley DE, Marthick JR, Donovan S, Malley RC, Banks A, Redwig F, Skala M, Dickinson JL and FitzGerald LM: Analysis of a large prostate cancer family identifies novel and recurrent gene fusion events providing evidence for inherited predisposition. Prostate. 82:540–550. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Xie J, Xiang J, Shen Y and Shao S: Mechanistic insights into the tools for intracellular protein delivery. Chem Bio Eng. 2:132–155. 2024. View Article : Google Scholar | |
|
Dematteis A, Miszczyk M, Cormio A, Matsukawa A, Gontero P and Shariat SF: The role of radiotherapy in pelvic nodal recurrence following definitive treatment for prostate cancer. Curr Opin Urol. 35:574–582. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Feng EM, Vo-Phamhi J, Subramanian AN, Dias M, Foye A, Vinson J, Hong JC, Freedland SJ, Alumkal JJ, Beltran H, et al: Racial variation in the advanced prostate cancer genome. Prostate Cancer Prostatic Dis. 28:902–907. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Hadjimichael AC, Foukas AF, Savvidou OD, Mavrogenis AF, Psyrri AK and Papagelopoulos PJ: The anti-neoplastic effect of doxycycline in osteosarcoma as a metalloproteinase (MMP) inhibitor: A systematic review. Clin Sarcoma Res. 10:72020. View Article : Google Scholar : PubMed/NCBI | |
|
Saad F, Armstrong AJ, Shore N, George DJ, Oya M, Sugimoto M, McKay RR, Hussain M and Clarke NW: Olaparib monotherapy or in combination with abiraterone for the treatment of patients with metastatic castration-resistant prostate cancer (mCRPC) and a BRCA mutation. Target Oncol. 20:445–466. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
McGrath CB, Shreves AH, Shanahan MR, Guard HE, Nhliziyo MV, Pernar CH, Penney KL, Lotan TL, Fiorentino M, Mucci LA and Stopsack KH: Etiology of prostate cancer with the TMPRSS2:ERG fusion: A systematic review of risk factors. Int J Cancer. 156:1898–1908. 2025. View Article : Google Scholar : | |
|
Zhang H, Kong L, Li J, Liu Z, Zhao Y, Lv X, Wu L, Chai L, You H, Jin J, et al: SPOP mutations increase PARP inhibitor sensitivity via CK2/PIAS1/SPOP axis in prostate cancer. JCI Insight. 10:e1868712025. View Article : Google Scholar : PubMed/NCBI | |
|
Ding T, Li X, Zhang L, Wei Z, Xiong C, Wang H, Hao X and Zeng X: Comparison of androgen receptor mutation detection between plasma extracellular vesicle DNA and cell-free DNA and its relationship to prostate cancer prognosis. Ann Med. 56:24267702024. View Article : Google Scholar : PubMed/NCBI | |
|
Bugoye FC, Torrorey-Sawe R, Biegon R, Dharsee N, Mafumiko F, Kibona H, Aboud S, Patel K and Mining S: Exploring therapeutic applications of PTEN, TMPRSS2:ERG fusion, and tumour molecular subtypes in prostate cancer management. Front Oncol. 15:15212042025. View Article : Google Scholar : PubMed/NCBI | |
|
Urabe F and Takemura K: Evaluating the genetic landscape of prostate cancer: New insights from BRCA1/2, ATM and CDK12 mutations. BMJ Oncol. 4:e0007172025. View Article : Google Scholar : PubMed/NCBI | |
|
Sekar A, Chatterjee R, Selvadurai BR, Pal LR, Verma A, Nataraj NB, Garcia DD, Giri S, Genna A, Karatekin F, et al: TMPRSS2-ERG confers resistance to antiandrogens: Mechanism and therapeutic implications. bioRxiv [Preprint]. 2025.01.17.633582. 2025. | |
|
Qi L, Li X, Liu Z, Zhang P and Liu L: A depth-wise separable residual neural network for PCDH8 status prediction in thyroid cancer pathological images. Intell Oncol. 1:290–298. 2025. View Article : Google Scholar | |
|
Cheng B, Luo T, Wu Y, Hu J, Yang C, Wu J, Luo Y, Shangguan W, Li W, Yang L, et al: Urinary exosomal FAM153C-RPL19 chimeric RNA as a diagnostic and prognostic biomarker for prostate cancer in Chinese patients. Cancer Lett. 631:2179382025. View Article : Google Scholar : PubMed/NCBI | |
|
Mugoni V, Ciani Y, Nardella C and Demichelis F: Circulating RNAs in prostate cancer patients. Cancer Lett. 524:57–69. 2022. View Article : Google Scholar | |
|
Zvirble M, Vaicekauskaite I, Survila Z, Bosas P, Dobrovolskiene N, Mlynska A, Sabaliauskaite R and Pasukoniene V: Liquid-based diagnostic panels for prostate cancer: The synergistic role of soluble PD-L1, PD-1, and mRNA biomarkers. Int J Mol Sci. 26:7042025. View Article : Google Scholar : PubMed/NCBI | |
|
Crocetto F, Musone M, Chianese S, Conforti P, Digitale Selvaggio G, Caputo VF, Falabella R, Del Giudice F, Giulioni C, Cafarelli A, et al: Blood and urine-based biomarkers in prostate cancer: Current advances, clinical applications, and future directions. J Liq Biopsy. 9:1003052025. View Article : Google Scholar : PubMed/NCBI | |
|
Cengiz T, Yurtbay A, Muslu O, Aydin Simsek S, Ozbalci AB, Coskun HS, Baris YS and Dabak N: Evaluation of the diagnostic accuracy of percutaneous core needle biopsy in bone and soft tissue tumors. Acta Chir Orthop Traumatol Cech. 91:376–384. 2024. View Article : Google Scholar | |
|
Aferin U, Bahtiyar N, Onaran I and Ozkara H: Are elevated mitochondrial DNA fragments in prostatic inflammation a potential biomarker for prostate cancer? Arch Ital Urol Androl. 95:116102023.PubMed/NCBI | |
|
Valentini V, Santi R, Silvestri V, Saieva C, Roviello G, Amorosi A, Comperat E, Ottini L and Nesi G: CD44 methylation levels in androgen-deprived prostate cancer: A putative epigenetic modulator of tumor progression. Int J Mol Sci. 26:25162025. View Article : Google Scholar : PubMed/NCBI | |
|
Su XA, Stopsack KH, Schmidt DR, Ma D, Li Z, Scheet PA, Penney KL, Lotan TL, Abida W, DeArment EG, et al: RAD21 promotes oncogenesis and lethal progression of prostate cancer. Proc Natl Acad Sci USA. 121:e24055431212024. View Article : Google Scholar : PubMed/NCBI | |
|
Montoya Perez I, Jambor I, Pahikkala T, Airola A, Merisaari H, Saunavaara J, Alinezhad S, Vaananen RM, Tallgren T, Verho J, et al: Prostate cancer risk stratification in men with a clinical suspicion of prostate cancer using a unique biparametric mri and expression of 11 genes in apparently benign tissue: Evaluation using machine-learning techniques. J Magn Reson Imaging. 51:1540–1553. 2020. View Article : Google Scholar | |
|
Gan L, Li W, Chen Q, Cheng L, Zhang F, Zhong H, Lu Y, Zheng L and Qian B: MicroRNA-145 in urologic tumors: Biological roles, regulatory networks, and clinical translation. Front Pharmacol. 16:16096462025. View Article : Google Scholar : PubMed/NCBI | |
|
Rafiq A and Kanavarioti A: The potential and limitations of the MinION/Yenos Platform for miRNA-Enabled early cancer detection. Int J Mol Sci. 26:38222025. View Article : Google Scholar : PubMed/NCBI | |
|
Davey M, Benzina S, Savoie M, Breault G, Ghosh A and Ouellette RG: Affinity vesicles captured urinary extracellular provide mRNA and miRNA biomarkers for improved accuracy of prostate cancer detection: A pilot study. Int J Mol Sci. 21:83302020. View Article : Google Scholar | |
|
Fuso GA, Bianciardi G, Mei R, Brusa I, Emiliani S, Fortunati E and Nanni C: Radiotracing the future: Non-FDG radiotracers nuclear medicine. Semin Nucl Med. 55:648–663. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Cicatiello AG, Musone M, Imperatore S, Giulioni C, La Rocca R, Cafarelli A, Del Giudice F, Dentice M and Crocetto F: Circulating miRNAs in genitourinary cancer: Pioneering advances in early detection and diagnosis. J Liq Biopsy. 8:1002962025. View Article : Google Scholar : PubMed/NCBI | |
|
Tong G, Jing X, Yang Y, Wang X, Lu J, Hu J, Wang Y, Jumuddin FA, Zhang W and Lv Y: Screening and validation of key genes involved in castration-resistant prostate cancer based on transcriptomics sequencing. Sci Rep. 15:256482025. View Article : Google Scholar : PubMed/NCBI | |
|
Muniz M, Childs DS, Andrews J, Mahmoud AM, Park S, Sartor O, Kase AM, Riaz IB, Stish BJ, Chaudhuri AA, et al: Deferral of systemic therapy in patients with oligorecurrent prostate cancer treated with metastasis-directed radiotherapy. Ann Transl Med. 13:292025. View Article : Google Scholar : PubMed/NCBI | |
|
Kwan EM, Ng SWS, Tolmeijer SH, Emmett L, Sandhu S, Buteau JP, Iravani A, Joshua AM, Francis RJ, Subhash V, et al: Lutetium-177-PSMA-617 or cabazitaxel in metastatic prostate cancer: Circulating tumor DNA analysis of the randomized phase 2 TheraP trial. Nat Med. 31:2722–2736. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Shiota M, Matsubara N, Kato T, Eto M, Osawa T, Abe T, Shinohara N, Nishimoto K, Yasumizu Y, Tanaka N, et al: Prediction of undetectable circulating tumor DNA by comprehensive genomic profiling assay in metastatic prostate cancer: The SCRUM-Japan MONSTAR SCREEN project. World J Urol. 42:5262024. View Article : Google Scholar : PubMed/NCBI | |
|
Peng N, Chen D, Yang M, Zeng C and Li W: PARP inhibitors for HRR-deficient metastatic castration-resistant prostate cancer: Mechanisms and clinical strategies. Eur J Pharm Sci. 212:1071932025. View Article : Google Scholar : PubMed/NCBI | |
|
Liang A, Gulati S, Huang Q, Dowst H, Lim A, Zarrin-Khameh N, Godoy G, Noor AB, Castro P, Scheurer ME, et al: Real-world effectiveness of darolutamide in metastatic castration-resistant prostate cancer. Endocr Relat Cancer. 32:e2401882025. View Article : Google Scholar : PubMed/NCBI | |
|
Sweeney CJ, Petry R, Xu C, Childress M, He J, Fabrizio D, Gjoerup O, Morley S, Catlett T, Assaf ZJ, et al: Circulating Tumor DNA assessment for treatment monitoring adds value to PSA in metastatic castration-resistant prostate cancer. Clin Cancer Res. 30:4115–4122. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Dariane C and Timsit MO: DNA-Damage-Repair gene alterations in genitourinary malignancies. Eur Surg Res. 63:155–164. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Okuda Y, Kato T, Ishizuya Y, Hayashi T, Yamamoto Y, Hatano K, Kawashima A, Murai J and Nonomura N: PARP Inhibitors in Genitourinary Cancer: A New Paradigm Beyond Prostate Cancer. Int J Urol. 32:1091–1101. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Virga A, Urbini M, Polano M, Petracci E, Tedaldi G, Gurioli G, Marisi G, Angeli D, Ambrosini-Spaltro A, De Luca G, et al: Mutational and low-coverage whole genome sequencing identifies actionable DNA repair alterations in prostate cancer plasma DNA. Sci Rep. 15:212962025. View Article : Google Scholar : PubMed/NCBI | |
|
Slootbeek PHJ, Tolmeijer SH, Mehra N and Schalken JA: Therapeutic biomarkers in metastatic castration-resistant prostate cancer: does the state matter? Crit Rev Clin Lab Sci. 61:178–204. 2024. View Article : Google Scholar | |
|
Zhang X, Zhou F, Lu T, Zhang S, Wei X, Qiu X, Xu L, Guo H and Zhuang J: Neoadjuvant darolutamide plus androgen deprivation therapy for high-risk and locally advanced prostate cancer: A multicenter, open-label, single-arm, phase II trial. World J Urol. 43:582025. View Article : Google Scholar : PubMed/NCBI | |
|
Wong J, Tian Y, Patel MS, Avasthi K, Hanson C, Larsen M, Ampaw E, Fadlullah MZH, Finklestein J, Tan AC, et al: Plasma cell-free DNA methylation-based prognosis in metastatic castrate-resistant prostate cancer. Res Sq [Preprint] rs.3.rs-6331572. 2025. | |
|
Crumbaker M, Goldstein LD, Murray DH, Tao J, Pathmanandavel S, Boulter N, Ratnayake L, Joshua AM, Kummerfeld S and Emmett L: Circulating tumour DNA biomarkers associated with outcomes in metastatic prostate cancer treated with Lutetium-177-PSMA-617. Eur Urol Open Sci. 57:30–36. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Fei X, Du X, Gong Y, Liu J, Fan L, Wang J, Wang Y, Zhu Y, Pan J, Dong B and Xue W: Early plasma circulating tumor DNA as a potential biomarker of disease recurrence in non-metastatic prostate cancer. Cancer Res Treat. 55:969–977. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Earland N, Chen K, Semenkovich NP, Chauhan PS, Zevallos JP and Chaudhuri AA: Emerging roles of circulating tumor DNA for increased precision and personalization in radiation oncology. Semin Radiat Oncol. 33:262–278. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Lianidou E: Detection and relevance of epigenetic markers on ctDNA: Recent advances and future outlook. Mol Oncol. 15:1683–1700. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Huang YX, Wang HY, Zhang YH, Lin YF, Qiao XQ and Hu LH: Bibliometric analysis of exosomes in the biomarker research field. Se Pu. 43:498–507. 2025.PubMed/NCBI | |
|
Aseervatham J: Dynamic role of exosome microRNAs in cancer cell signaling and their emerging role as noninvasive biomarkers. Biology (Basel). 12:7102023.PubMed/NCBI | |
|
Trujillo B, Wu A, Wetterskog D and Attard G: Blood-based liquid biopsies for prostate cancer: clinical opportunities and challenges. Br J Cancer. 127:1394–1402. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Pudova E, Kobelyatskaya A, Katunina I, Snezhkina A, Nyushko K, Fedorova M, Pavlov V, Bulavkina E, Dalina A, Tkachev S, et al: Docetaxel resistance in castration-resistant prostate cancer: Transcriptomic determinants and the effect of inhibiting Wnt/β-catenin signaling by XAV939. Int J Mol Sci. 23:128372022. View Article : Google Scholar | |
|
Chen Z, Fu S, Shan Y, He Z, Gu J, Wu H, Lin J, Huang Y, Wang H, Lu Y and Ding M: Circ_0001047 inhibits prostate cancer progression and enhances abiraterone sensitivity via miR-122-5p/FKBP5/PHLPPl/AKT axis in vitro. Discov Oncol. 15:5692024. View Article : Google Scholar | |
|
Song X, Li T, Zhou W, Feng C, Zhou Z, Chen Y, Li D, Chen L, Zhao J, Zhang Y and Han B: CAF-derived exosomal miR-196b-5p after androgen deprivation therapy promotes epithelial-mesenchymal transition in prostate cancer cells through HOXC8/NF-KB signaling pathway. Biol Direct. 20:802025. View Article : Google Scholar | |
|
Pecoraro G, Leone I, Nuzzo S, Negueruela S, Smaldone G and Buono L: Co-modulation of a circular form of PCDH11Y during neuroendocrine differentiation of prostate cancer. Front Oncol. 15:15024052025. View Article : Google Scholar : PubMed/NCBI | |
|
Julio MK: Elevating prostate cancer diagnostics through extracellular vesicle miRNAs. Gene. 1496622025. View Article : Google Scholar | |
|
Qin X, Niu R, Tan Y, Huang Y, Ren W, Zhou W, Wu H, Zhang J, Xu M, Zhou X, et al: Exosomal PSM-E inhibits macrophage M2 polarization to suppress prostate cancer metastasis through the RACK1 signaling axis. Biomark Res. 12:1382024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang J, Chen B, Gan C, Sun H, Zhang J and Feng L: A comprehensive review of small interfering RNAs (siRNAs): Mechanism, therapeutic targets, and delivery strategies for cancer therapy. Int J Nanomedicine. 18:7605–7635. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Kren BT, Henzler CM, Ahmed K and Trembley JH: Impact of protein kinase CK2 downregulation and inhibition on oncomir clusters 17 ~ 92 and 106b ~ 25 in prostate, breast, and head and neck cancers. Mol Med. 30:1752024. View Article : Google Scholar | |
|
Shukla KK, Choudhary GR, Sankanagoudar S, Misra S, Vishnoi JR, Pareek P, Pilla KK, Pandey SN and Sharma P: Deregulation of miR-10b and miR-21 correlate with cancer stem cells expansion through the apoptotic pathway in prostate cancer. Asian Pac J Cancer Prev. 24:2105–2119. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Qiu J, Wu J, Zhou N, Lai C, Huang X, Liu C, Yuan X and Xu K: An EcDNA gene-based risk model and functional verification of a key ec-lncRNA AC016394.2 for prostate cancer. Cancer Cell Int. 25:2402025. View Article : Google Scholar : PubMed/NCBI | |
|
Gupta P, Dutta S, Kumar P, Kaushik M, Ashique S and Bhowmick M: Targeted management: Unlocking the crucial role of PROTACs in cancer treatment. Curr Drug Discov Technol. Feb 26–2025.Epub ahead of print. View Article : Google Scholar | |
|
Miao G, Shang Z and Wang X, Zhang J, Xu M, He P, Zhong Q, Zhao X, Tan G and Wang X: Concurrent inhibition of tumor growth and metastasis by a lipidated nanophotosensitizer tracing and disabling tumor extracellular vesicles. Nat Cancer. 6:1438–1457. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Wang C, Feng Y, Rong X, Yan J, Lv B, Jiang H, Duan L and Jiang J: Mesenchymal stromal cell exosomes for drug delivery of prostate cancer treatments: A review. Stem Cell Res Ther. 16:182025. View Article : Google Scholar : PubMed/NCBI | |
|
Bellomo G, Marcocci F, Bianchini D, Mezzenga E, D'Errico V, Menghi E, Zannoli R and Sarnelli A: MR spectroscopy in prostate cancer: New algorithms to optimize metabolite quantification. PLoS One. 11:e01657302016. View Article : Google Scholar : PubMed/NCBI | |
|
Gungoren M, Koyuncu I, Kilinc E and Baysal Z: Docetaxel-Loaded, Dipeptide-Functionalized Y-Fe(2)O(3) magnetic nanoparticles: Synthesis, characterization, and targeting to prostate cancer. Chem Biodivers. 22:e2024032452025. View Article : Google Scholar | |
|
Galey L, Olanrewaju A, Nabi H, Paquette JS, Pouliot F and Audet-Walsh E: PSA, an outdated biomarker for prostate cancer: In search of a more specific biomarker, citrate takes the spotlight. J Steroid Biochem Mol Biol. 243:1065882024. View Article : Google Scholar : PubMed/NCBI | |
|
Jupin M, van Heijster FHA and Heerschap A: Metabolite interactions in prostatic fluid mimics assessed by (1)H NMR. MAGMA. 35:683–694. 2022. View Article : Google Scholar | |
|
Ma T, Xie W, Xu Z, Gao W, Zhou J, Wang Y and Chan FL: Estrogen-related receptor alpha (ERRalpha) controls the sternness and cellular energetics of prostate cancer cells via its direct regulation of citrate metabolism and zinc transportation. Cell Death Dis. 16:1542025. View Article : Google Scholar | |
|
Feng D, Zhang J, Niu H, Zheng X, Jia M, Lu Q, Wang J, Guo W, Sun Q, Yuan H and Lou H: Spermidine inactivates proteasome activity and enhances ferroptosis in prostate cancer. Acta Pharm Sin B. 15:2095–2113. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Li QZ, Zuo ZW, Zhou ZR and Ji Y: Poly amine homeostasis-based strategies for cancer: The role of combination regimens. Eur J Pharmacol. 910:1744562021. View Article : Google Scholar | |
|
Tamura R, Chen J, De Jaeger M, Morris JF, Scott DA, Vangheluwe P and Looger LL: Genetically encoded fluorescent sensors for visualizing polyamine levels uptake, and distribution. bioRxiv [Preprint]. 2024.08.21.609037. 2024. | |
|
Hunt RL, Oh J, Jain A, Kuo TH, Berardi D, Jian W, Song D, Wu Q, Goodman AL, Palm NW, et al: Discovery of a widespread polyamine-low-molecular-weight thiol hybrid pathway in clostridioides difficile. ACS Infect Dis. 11:2246–2264. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Zong Y, Li H, Liao P, Chen L, Pan Y, Zheng Y, Zhang C, Liu D, Zheng M and Gao J: Mitochondrial dysfunction: Mechanisms and advances in therapy. Signal Transduct Target Ther. 9:1242024. View Article : Google Scholar : PubMed/NCBI | |
|
Kumar R, Jonnatan S, Sanin DE, Vakkala V, Kadam A, Kumar S, Dalrymple SL, Zhao L, Foley J, Holbert CE, et al: Androgen receptor drives polyamine synthesis creating a vulnerability for prostate cancer. medRxiv [Preprint]. 2024.12.12.24318845. 2024.PubMed/NCBI | |
|
Danilova EY, Maslova AO, Stavrianidi AN, Nosyrev AE, Maltseva LD and Morozova OL: CKD urine metabolomics: Modern concepts and approaches. Pathophysiology. 30:443–466. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu D, Dai J, Jia J, Kanagaraj T, Rajalakshmi K, Muthusamy S, Geng L and Yuan G: Biogenic synthesis of N-doped carbon dots from S. cumini seeds for prostate cancer biomarker citrate detection, its live cancer cell imaging. Spectrochim Acta A Mol Biomol Spectrosc. 329:1255682025. View Article : Google Scholar | |
|
Jimenez Gutierrez GE, Borbolla Jimenez FV, Munoz LG, Tapia Guerrero YS, Murillo Melo NM, Cristobal-Luna JM, Leyva Garcia N, Cordero-Martinez J and Magana JJ: The molecular role of polyamines in age-related diseases: An update. Int J Mol Sci. 24:164692023. View Article : Google Scholar : PubMed/NCBI | |
|
Lista S, Gonzalez-Dominguez R, Lopez-Ortiz S, Gonzalez-Dominguez A, Menendez H, Martin-Hernandez J, Lucia A, Emanuele E, Centonze D, Imbimbo BP, et al: Integrative metabolomics science in Alzheimer's disease: Relevance and future perspectives. Ageing Res Rev. 89:1019872023. View Article : Google Scholar : PubMed/NCBI | |
|
Sena LA: Polyamine metabolism in prostate cancer. Curr Opin Oncol. 37:223–232. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Hu P, Yuan M, Guo B, Lin J, Yan S, Huang H, Chen JL, Wang S and Ma Y: Citric acid promotes immune function by modulating the intestinal barrier. Int J Mol Sci. 25:12392024. View Article : Google Scholar : PubMed/NCBI | |
|
Wu WF, Song XY, Warner M, Imamov O, Antonson P and Gustafsson JA: The role of estrogen receptor beta in maintaining basal cells and modulating the immune environment in the prostate. Proc Natl Acad Sci USA. 122:e25057971222025. View Article : Google Scholar | |
|
Ramatlho P, Rugemalila MM, Tawe L, Pain D, Choga OT, Ndlovu AK, Koobotse MO, Lal P, Rebbeck TR, Paganotti GM, et al: The role of immunohistochemistry for AMACR/p504s and p63 in distinguishing prostate cancer from benign prostate tissue samples in botswana. Res Rep Urol. 17:1–10. 2025.PubMed/NCBI | |
|
Spieker AJ, Gordetsky JB, Maris AS, Dehan LM, Denney JE, Arnold Egloff SA, Scarpato K, Barocas DA and Giannico GA: PTEN expression and morphological patterns in prostatic adenocarcinoma. Histopathology. 79:1061–1071. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang HZ, Wang SY, Guo YN and Zhao M: Isolated intraductal carcinoma of the prostate: A clinicopathological and molecular analysis. Zhonghua Bing Li Xue Za Zhi. 53:803–808. 2024.In Chinese. PubMed/NCBI | |
|
Martini C, Logan JM, Sorvina A, Prabhakaran S, Ung BSY, Johnson IRD, Hickey SM, Brooks RD; kConFab Consortium; Caruso MC, et al: Distinct patterns of biomarker expression for atypical intraductal proliferations in prostate cancer. Virchows Arch. 485:723–728. 2024. View Article : Google Scholar : | |
|
Akhoundova D, Fischer S, Triscott J, Lehner M, Thienger P, Maletti S, Jacquet M, Lubis DSH, Bubendorf L, Jochum W and Rubin MA: Rare histologic transformation of a CTNNB1 (β-catenin) mutated prostate cancer with aggressive clinical course. Diagn Pathol. 19:832024. View Article : Google Scholar | |
|
Ayala Soriano C, Benitez Barzaga M, Chhina A, Jain M and Nava VE: Hepatoid prostatic carcinoma with adrenal metastasis and novel genetic alterations. Diagn Cytopathol. 50:E310–E314. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Ruschoff JH, Stratton S, Roberts E, Clark S, Sebastiao N, Fankhauser CD, Eberli D, Moch H, Wild PJ and Rupp NJ: A novel 5x multiplex immunohistochemical staining reveals PSMA as a helpful marker in prostate cancer with low p504s expression. Pathol Res Pract. 228:1536672021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou L, Xu J, Wang S, Yang X, Li C, Zhou J, Zhang P, Xu H and Wang C: Papillary renal neoplasm with reverse polarity: A clinicopathologic study of 7 cases. Int J Surg Pathol. 28:728–734. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Nolan E, Lindeman GJ and Visvader JE: Deciphering breast cancer: From biology to the clinic. Cell. 186:1708–1728. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Chenlo M, Aliyev E, Rodrigues JS, Vieiro-Balo P, Blanco Freire MN, Cameselle-Teijeiro JM and Alvarez CV: Sequential Colocalization of ERa, PR, and AR hormone receptors using confocal microscopy enables new insights into normal breast and prostate tissue and cancers. Cancers (Basel). 12:35912020. View Article : Google Scholar : PubMed/NCBI | |
|
Comperat E, Renard-Penna R, Ondet V, Cancel-Tassin G and Cussenot O: Diagnosis of prostate cancer in one day: The benefits of cytology in tumour detection. Cytopathology. 32:211–216. 2021. View Article : Google Scholar | |
|
Bee J, Zhou X, Tipirneni Y, Chen S, Qi J, Iczkowski KA, Dall'Era M and Marcu L: Rapid discrimination of high-grade prostate cancer using label-free fluorescence lifetime measurements. medRxiv [Preprint]. 2025.05.23.25328257. 2025. | |
|
Riddi VL, Harahap EU, Sitinjak D and Nirmawati R: Ductal adenocarcinoma of the prostate presenting with bladder invasion and bone metastasis: A case report. Cureus. 17:e860002025.PubMed/NCBI | |
|
Salvi M, Manini C, Lopez JI, Fenoglio D and Molinari F: Deep learning approach for accurate prostate cancer identification and stratification using combined immunostaining of cytokeratin, p63, and racemase. Comput Med Imaging Graph. 109:1022882023. View Article : Google Scholar : PubMed/NCBI | |
|
Bolis M, Bossi D, Vallerga A, Ceserani V, Cavalli M, Impellizzieri D, Di Rito L, Zoni E, Mosole S, Elia AR, et al: Dynamic prostate cancer transcriptome analysis delineates the trajectory to disease progression. Nat Commun. 12:70332021. View Article : Google Scholar : PubMed/NCBI | |
|
Conte MI, Fuentes-Trillo A and Dominguez Conde C: Opportunities and tradeoffs in single-cell transcriptomic technologies. Trends Genet. 40:83–93. 2024. View Article : Google Scholar | |
|
Zhou L, Zhang C, Zhang Y and Shi C: Application of organoid models in prostate cancer research. Front Oncol. 11:7364312021. View Article : Google Scholar : PubMed/NCBI | |
|
Mharrach I, Aqerrout M, Tadlaoui KA, Laraqui A, Ghazzaly AE and Ennaji MM: Methylation status of GSTP1 and APC promoter genes as potential biomarkers for early detection and tumor aggressiveness in prostate cancer: Insights from a Moroccan cohort. Mol Biol Rep. 52:7112025. View Article : Google Scholar : PubMed/NCBI | |
|
Aloke C, Onisuru OO and Achilonu I: Glutathione S-transferase: A versatile and dynamic enzyme. Biochem Biophys Res Commun. 734:1507742024. View Article : Google Scholar : PubMed/NCBI | |
|
Gupta H, Inoue H, Nakai Y, Nakayama M, Jones T, Hicks JL, Kumar B, Gurel M, Nelson WG, De Marzo AM and Yegnasubramanian S: Progressive spreading of DNA methylation in the GSTP1 promoter CpG island across transitions from precursors to invasive prostate cancer. Cancer Prev Res (Phila). 16:449–460. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Ji R, Zhang L, Xing Y, Zhu Y, Mao K, Wang M, Xuan K, Yang Y, Lo R, Luo B and Lu Z: Clinical significance of urine-based automated detection of GSTP1 methylation for the diagnosis of suspected prostate cancer patients. Transl Androl Urol. 14:1204–1213. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Cussenot O, Timms KM, Perrot E, Blanchet P, Brureau L, Solimeno C, Fromont G, Comperat E and Cancel-Tassin G: Tumour-based mutational profiles predict visceral metastasis outcome and early death in prostate cancer patients. Eur Urol Oncol. 7:597–604. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Turan G, Turan M, Basbinar Y and Ellidokuz H: Prostate cancer in Turkiye: Trend analysis of incidence and mortality rates. Semin Oncol. 52:1523912025. View Article : Google Scholar | |
|
Lam D, Clark S, Stirzaker C and Pidsley R: Advances in prognostic methylation biomarkers for prostate cancer. Cancers (Basel). 12:29932020. View Article : Google Scholar : PubMed/NCBI | |
|
Eismann L, von Walter P, Jung A, Chaloupka M, Rodler S, Westhofen T, Buchner A, Stief CG, Stadler T and Schlenker B: Methylation status of various gene loci in localized prostate cancer: Novel biomarkers for diagnostics and biochemical recurrence. Urol Oncol. 41:325 el–325 e8. 2023. View Article : Google Scholar | |
|
Deng Y, Cai B and Wang X: A fully automated quantitative analysis method based on deep learning algorithms for immunohistochemical staining expression intensities. Intell Oncol. 1:256–264. 2025. View Article : Google Scholar | |
|
Friedemann M, Horn F, Gutewort K, Tautz L, Jandeck C, Bechmann N, Sukocheva O, Wirth MP, Fuessel S and Menschikowski M: Increased sensitivity of detection of RASSF1A and GSTP1 DNA fragments in serum of prostate cancer patients: Optimisation of diagnostics using OBBPA-ddPCR. Cancers (Basel). 13:44592021. View Article : Google Scholar : PubMed/NCBI | |
|
Tufail M: PTEN-mediated resistance in cancer: From foundation to future therapies. Toxicol Rep. 14:1019872025. View Article : Google Scholar : PubMed/NCBI | |
|
Bucker L and Lehmann U: CDH1 (E-cadherin) gene methylation in human breast cancer: Critical appraisal of a long and twisted story. Cancers (Basel). 14:43772022. View Article : Google Scholar : PubMed/NCBI | |
|
Corso G, Magnoni F, Dal Molin M, Marino E, Nicosia L, Pesapane F, Noonan DM and Albini A: Second-hit CDH1 gene mechanisms in hereditary diffuse gastric and lobular breast cancer syndrome: Frequency and impact on tumorigenesis. Hum Mol Genet. 34:1345–1352. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang W, Zhang K, Ma Y, Song Y, Qi T, Xiong G, Zhang Y, Kan C, Zhang J, Han F and Sun X: Secreted frizzled-related proteins: A promising therapeutic target for cancer therapy through Wnt signaling inhibition. Biomed Pharmacother. 166:1153442023. View Article : Google Scholar : PubMed/NCBI | |
|
Fiu C, Tang H, Hu N and Fi T: Methylomics and cancer: The current state of methylation profiling and marker development for clinical care. Cancer Cell Int. 23:2422023. View Article : Google Scholar | |
|
Zhou Y, Tao L, Qiu J, Xu J, Yang X, Zhang Y, Tian X, Guan X, Cen X and Zhao Y: Tumor biomarkers for diagnosis, prognosis and targeted therapy. Signal Transduct Target Ther. 9:1322024. View Article : Google Scholar : PubMed/NCBI | |
|
Ibrahim J, Peeters M, Van Camp G and Op de Beeck K: Methylation biomarkers for early cancer detection and diagnosis: Current and future perspectives. Eur J Cancer. 178:91–113. 2023. View Article : Google Scholar | |
|
Bao Y, Wang X, Zeng B, Shi Y, Huang Y, Huang Y, Shang S, Shan F and Ma F: Research progress of liquid biopsy based on DNA methylation in tumor diagnosis and Treatment. Biomolecules. 14:16342024. View Article : Google Scholar | |
|
Wan JCM, Sasieni P and Rosenfeld N: Promises and pitfalls of multi-cancer early detection using liquid biopsy tests. Nat Rev Clin Oncol. 22:566–580. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Fi X and Fi X, Wang F, Hou Y, Fiu Y, Mao J, Zhang F and Fi X: Advancing Traditional Chinese medicine research through network pharmacology: Strategies for target identification, mechanism elucidation and innovative therapeutic applications. Am J Chin Med. 53:2021–2042. 2025. View Article : Google Scholar | |
|
Maliye R, Babu S, Harris AE, Patke R, Ntekim AI, Rutland CS, James V, Mongan NP, Madhusudan S, Jeyapalan JN and Semenova MM: Evaluation of human papillomavirus as a risk factor in prostate cancer pathogenesis. Discov Oncol. 16:13752025. View Article : Google Scholar : PubMed/NCBI |