Current Understanding of the Heterogeneity in Prostate Cancer and Renal Cell Carcinoma

A special issue of Journal of Clinical Medicine (ISSN 2077-0383). This special issue belongs to the section "Oncology".

Deadline for manuscript submissions: closed (20 September 2023) | Viewed by 5650

Special Issue Editors


E-Mail Website
Guest Editor
Department of Urology, West China School of Medicine/West China Hospital of Sichuan University, Chengdu, China
Interests: prostate cancer; renal cell carcinoma; immunotherapy; synthetic lethal therapy; cancer pathology; tumor biomarker
Department of Urology, West China School of Medicine/West China Hospital of Sichuan University, Chengdu, China
Interests: urological tumors; oncogenomics; genomic integrity; DNA repair in tumors; homologous recombination deficiency

Special Issue Information

Dear Colleagues,

Prostate cancer and renal cell carcinoma are genomically and phenotypically heterogeneous urological tumors. Thus, they require a multidisciplinary approach to address the diagnosis and therapy-related clinical hurdles in the comprehensive management of these diseases.

Molecularly, recent years have seen many exciting new data in the genomic, transcriptional, and epigenomic perspectives of these two malignancies. These findings have revolutionized patients' diagnostic procedures, treatment, and drug development as well. With the help of next-generation sequencing and following functional experiments, obtaining a more detailed understanding of specific genomic aberrations is becoming more reachable, and we are taking steps forward toward personalized therapy. Additionally, clinically relevant data from a histopathological view will significantly improve our understanding of tumor heterogeneity.

Therefore, this Special Issue aims to cover recent findings on the heterogeneity in these two cancers, including sequencing-based studies, computational and bioinformatic approaches, clinical observations (especially from a pathological and imaging view), and also mechanistic studies to decipher novel significant players in cancer networks.

Prof. Dr. Hao Zeng
Dr. Sha Zhu
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Journal of Clinical Medicine is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2600 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • prostate cancer
  • renal cell carcinoma
  • heterogeneity
  • oncogenomics
  • personalized therapy
  • next-generation sequencing

Published Papers (4 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Editorial

Jump to: Research

6 pages, 207 KiB  
Editorial
Our Current Understanding of the Heterogeneity in Prostate Cancer and Renal Cell Carcinoma
by Sha Zhu, Junru Chen and Hao Zeng
J. Clin. Med. 2023, 12(4), 1526; https://doi.org/10.3390/jcm12041526 - 15 Feb 2023
Cited by 2 | Viewed by 1108
Abstract
Urological cancer is a collective term for cancers of the bladder, kidney, ureter, penis, prostate, and testicles. Last year, more than 444,000 people were diagnosed with urinary cancers in the United States. In this review, we talk about the complexity of prostate and [...] Read more.
Urological cancer is a collective term for cancers of the bladder, kidney, ureter, penis, prostate, and testicles. Last year, more than 444,000 people were diagnosed with urinary cancers in the United States. In this review, we talk about the complexity of prostate and kidney cancer. Full article

Research

Jump to: Editorial

12 pages, 1720 KiB  
Article
Prognostic Significance of Chromogranin A Expression in the Initial and Second Biopsies in Metastatic Prostate Cancer
by Zhuo Huang, Ying Tang, Yuyan Wei, Jingyu Qian, Yifan Kang, Duohao Wang, Miao Xu, Ling Nie, Xueqin Chen, Ni Chen and Qiao Zhou
J. Clin. Med. 2023, 12(10), 3362; https://doi.org/10.3390/jcm12103362 - 9 May 2023
Viewed by 1265
Abstract
Neuroendocrine differentiation (NED) characterized by the expression of neuroendocrine markers, such as chromogranin A (CgA), is frequently observed in advanced prostate cancer (PCa), the prognostic significance of which is still controversial. Here we specifically addressed the issue of the potential prognostic value of [...] Read more.
Neuroendocrine differentiation (NED) characterized by the expression of neuroendocrine markers, such as chromogranin A (CgA), is frequently observed in advanced prostate cancer (PCa), the prognostic significance of which is still controversial. Here we specifically addressed the issue of the potential prognostic value of CgA expression in advanced-stage PCa patients with distant metastases and its change over time from metastatic hormone-sensitive (mHSPC) to metastatic castration-resistant prostate cancer (mCRPC). CgA expression was assessed immunohistochemically in initial biopsies of mHSPC, as well as in second biopsies of mCRPC in sixty-eight patients, and its correlation with prognosis (together with conventional clinicopathologic parameters) was analyzed using the Kaplan–Meier method and Cox proportional hazard model. We found that CgA expression was an independent adverse prognostic factor for both mHSPC (CgA positivity ≥ 1%, HR = 2.16, 95% CI: 1.04–4.26, p = 0.031) and mCRPC (CgA ≥ 10%, HR = 20.19, 95% CI: 3.04–329.9, p = 0.008). CgA positivity generally increased from mHSPC to mCRPC and was a negative prognosticator. The assessment of CgA expression may help with the clinical evaluation of advanced-stage patients with distant metastases. Full article
Show Figures

Figure 1

11 pages, 1663 KiB  
Article
Delta Radiomics Model Predicts Lesion-Level Responses to Tyrosine Kinase Inhibitors in Patients with Advanced Renal Cell Carcinoma: A Preliminary Result
by Yuntian Chen, Enyu Yuan, Guangxi Sun, Bin Song and Jin Yao
J. Clin. Med. 2023, 12(4), 1301; https://doi.org/10.3390/jcm12041301 - 6 Feb 2023
Viewed by 1161
Abstract
Background: This study aimed to develop and internally validate computed tomography (CT)-based radiomic models to predict the lesion-level short-term response to tyrosine kinase inhibitors (TKIs) in patients with advanced renal cell carcinoma (RCC). Methods: This retrospective study included consecutive patients with RCC that [...] Read more.
Background: This study aimed to develop and internally validate computed tomography (CT)-based radiomic models to predict the lesion-level short-term response to tyrosine kinase inhibitors (TKIs) in patients with advanced renal cell carcinoma (RCC). Methods: This retrospective study included consecutive patients with RCC that were treated using TKIs as the first-line treatment. Radiomic features were extracted from noncontrast (NC) and arterial-phase (AP) CT images. The model performance was assessed using the area under the receiver operating characteristic curve (AUC), calibration curve, and decision curve analysis (DCA). Results: A total of 36 patients with 131 measurable lesions were enrolled (training: validation = 91: 40). The model with five delta features achieved the best discrimination capability with AUC values of 0.940 (95% CI, 0.890‒0.990) in the training cohort and 0.916 (95% CI, 0.828‒1.000) in the validation cohort. Only the delta model was well calibrated. The DCA showed that the net benefit of the delta model was greater than that of the other radiomic models, as well as that of the treat-all and treat-none criteria. Conclusions: Models based on CT delta radiomic features may help predict the short-term response to TKIs in patients with advanced RCC and aid in lesion stratification for potential treatments. Full article
Show Figures

Figure 1

12 pages, 2126 KiB  
Article
The Somatic Mutational Landscape of Mismatch Repair Deficient Prostate Cancer
by Bangwei Fang, Yu Wei, Jian Pan, Tingwei Zhang, Dingwei Ye and Yao Zhu
J. Clin. Med. 2023, 12(2), 623; https://doi.org/10.3390/jcm12020623 - 12 Jan 2023
Viewed by 1630
Abstract
Prostate cancers with mismatch repair deficiency (MMR-d) have aggressive clinical and histological features, and they are potentially responsive to immunotherapy. However, its rarity prevents the analysis of the underlying biology. Here, we collected the genomic data of 2664 primary prostate tumors and 1409 [...] Read more.
Prostate cancers with mismatch repair deficiency (MMR-d) have aggressive clinical and histological features, and they are potentially responsive to immunotherapy. However, its rarity prevents the analysis of the underlying biology. Here, we collected the genomic data of 2664 primary prostate tumors and 1409 metastatic prostate tumors from the GENIE and TCGA databases. A total of 69 (2.59%) primary and 60 (4.26%) metastatic MMR-d tumors were identified among these tumors. Single nucleotide variant (SNV) frequencies of 34 candidate genes (including KMT2D (46.4%), ZFHX3 (33.3%), JAK1 (31.9%), and RNF43 (27.5%)) and 16 candidate genes (including KMT2D (33.3%) and JAK1 (28.3%)) were higher in MMR-d primary tumors and MMR-d metastatic tumors, respectively. The tumor mutation burden (TMB) was higher in primary MMR-d tumors. Homozygous deletions of EPCAM and EPAS1 were enriched in MMR-d primary tumors, while EPCAM deletions were enriched in metastatic MMR-d tumors. For genomic rearrangement events, TMPRSS2-ETS fusions were less frequent in primary MMR-d tumors. Our study indicates MMR-d prostate cancers have unique genomic features. These may play an important role in providing therapeutic targets for the treatment of this subset of prostate cancer patients. Full article
Show Figures

Figure 1

Back to TopTop