Molecular and Cellular Mechanisms of Cancers: Prostate Cancer

A special issue of Cells (ISSN 2073-4409). This special issue belongs to the section "Cellular Pathology".

Deadline for manuscript submissions: closed (31 January 2024) | Viewed by 17659

Special Issue Editor


E-Mail Website
Guest Editor
Laboratory of Radiobiology, Department of Applied Clinical Sciences and Biotechnologies, University of l'Aquila, 67100 L'Aquila, Italy
Interests: prostate cancer
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Prostate cancer (PCa) incidence has been dramatically increasing in the last few years in industrialized Western countries. The androgen receptor (AR) plays a central role in the pathogenesis of this disease. Unfortunately, most PCa patients enter into an androgen-independent stage named castration-resistant prostate cancer (CRPC) in which AR signaling still is active. This more aggressive state of disease, in the vast majority of cases, leads to questions about the molecular mechanisms involved in tumor recurrence. Though localized PCa is usually treated by radical prostatectomy, androgen deprivation therapy (ADT) is preferred in locally advanced disease in combination with chemotherapy.

The inhibition of AR remains, however, a well-established promising drug target in CRPC. In spite of the improvements in the current treatment for CRPC by targeting the AR, the evolution of adaptive AR signaling leads to therapy-resistant CRPC. Treatment failure is mostly based on the inability to keep AR under long-term restraint due to adaptive responses of AR signaling. One underlying mechanism appears to be increased AR protein stability. Therefore, the regulation of AR protein stability and its degradation is another interesting path that could enhance our knowledge of carcinogenesis and tumor evolution, possibly leading to novel therapeutic targets.

The main focus of this Special Issue will be the evaluation of molecular pathways as pharmacological targets for treatment strategies that may improve the management of biologically aggressive and resistant CRPCs. This Special Issue will provide a platform for all pharmaceutical and translational scientists to research important breakthroughs in drug discovery and new therapeutics in this field.

Potential topics include, but are not limited to, those listed below.

  1. Molecular alterations associated with prostate cancer.
  • Androgen receptor—effectiveness and resistance to current pharmacological anti AR approaches;
  • Targeting mutated ARs and/or splicing variants;
  • Epigenetic regulation of AR expression—methylation and histone acetylation/deacetylation status, as well as AR protein stability, as targets for the therapy of CRPC and chemotherapy-resistant prostate cancers;
  • Intra-prostatic androgen synthesis as target for the treatment of aggressive/CRPCs (i.e., inhibitors of cytochrome P450 enzyme CYP17, 17α-hydroxylase and 17,20-lyase, as well as AKR1C3).
  1. Metabolic alterations and PCa progression—cellular constituents of the prostate stroma—key contributors to prostate cancer progression and therapy resistance.
  • Obesity and mesenchymal stem cells—key players in prostate cancer progression;
  • Lipid synthesis and metabolism, i.e., fatty acid synthase (FASN) and AMACR;
  • Glutathione metabolism—γ-glutamylcyclotransferase (GGCT);
  • Stroma–cancer cell interactions and hypoxia—stromal niche for epithelial stem cells;
  • Tumor-associated macrophage activation.
  1. Castration-resistant metastatic disease (mCRPC)
  • Epithelial-to-mesenchymal transition (EMT);
  • VEGF antagonists and VEGFR inhibitors;
  • Antagonists of integrins (avb3, avb5, a5b1, etc.) and bone metastases.
  1. Mechanisms and strategies to overcome resistance to radiotherapy or pharmacologic chemotherapy
  • Radio-sensitizing agents;
  • Modulators of intracellular trafficking of proteins and mRNA;
  • Antagonists of cell recruitment (monocytes and granulocytes, bone marrow myeloid cells, PCa stem-like cells);
  • CXCR4/CXCR7 antagonists;
  • Ephrin/ephrin receptor antagonists or inhibitors;
  • P21 activated kinases (PAKs).
  1. Inflammation and NF-κB signaling in prostate Cancer: mechanisms and clinical implications.
  2. Immunomodulators
    Immune checkpoint-mediated interactions between cancer and immune cells.

We are pleased to invite you to contribute original articles, reviews, communications, etc. We look forward to your contributions to this Special Issue.

Dr. Claudio Festuccia
Guest Editor

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. Cells 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 2700 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
  • PCa progression
  • molecular pathway
  • treatment strategies
  • immune checkpoint
  • radiotherapy
  • pharmacologic chemotherapy
  • signaling

Related Special Issues

Published Papers (9 papers)

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

Research

Jump to: Review

16 pages, 7919 KiB  
Article
Effects of Dickkopf-1 (DKK-1) on Prostate Cancer Growth and Bone Metastasis
by Shiyu Yuan, Nathan K. Hoggard, Noriko Kantake, Blake E. Hildreth III and Thomas J. Rosol
Cells 2023, 12(23), 2695; https://doi.org/10.3390/cells12232695 - 24 Nov 2023
Cited by 3 | Viewed by 1307
Abstract
Osteoblastic bone metastases are commonly detected in patients with advanced prostate cancer (PCa) and are associated with an increased mortality rate. Dickkopf-1 (DKK-1) antagonizes canonical WNT/β-catenin signaling and plays a complex role in bone metastases. We explored the function of cancer cell-specific DKK-1 [...] Read more.
Osteoblastic bone metastases are commonly detected in patients with advanced prostate cancer (PCa) and are associated with an increased mortality rate. Dickkopf-1 (DKK-1) antagonizes canonical WNT/β-catenin signaling and plays a complex role in bone metastases. We explored the function of cancer cell-specific DKK-1 in PCa growth, metastasis, and cancer–bone interactions using the osteoblastic canine PCa cell line, Probasco. Probasco or Probasco + DKK-1 (cells transduced with human DKK-1) were injected into the tibia or left cardiac ventricle of athymic nude mice. Bone metastases were detected by bioluminescent imaging in vivo and evaluated by micro-computed tomography and histopathology. Cancer cell proliferation, migration, gene/protein expression, and their impact on primary murine osteoblasts and osteoclasts, were evaluated in vitro. DKK-1 increased cancer growth and stimulated cell migration independent of canonical WNT signaling. Enhanced cancer progression by DKK-1 was associated with increased cell proliferation, up-regulation of NF-kB/p65 signaling, inhibition of caspase-dependent apoptosis by down-regulation of non-canonical WNT/JNK signaling, and increased expression of epithelial-to-mesenchymal transition genes. In addition, DKK-1 attenuated the osteoblastic activity of Probasco cells, and bone metastases had decreased cancer-induced intramedullary woven bone formation. Decreased bone formation might be due to the inhibition of osteoblast differentiation and stimulation of osteoclast activity through a decrease in the OPG/RANKL ratio in the bone microenvironment. The present study indicated that the cancer-promoting role of DKK-1 in PCa bone metastases was associated with increased growth of bone metastases, reduced bone induction, and altered signaling through the canonical WNT-independent pathway. DKK-1 could be a promising therapeutic target for PCa. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Cancers: Prostate Cancer)
Show Figures

Figure 1

18 pages, 2588 KiB  
Article
Targeting of AKT1 by miR-143-3p Suppresses Epithelial-to-Mesenchymal Transition in Prostate Cancer
by Lee Armstrong, Colin E. Willoughby and Declan J. McKenna
Cells 2023, 12(18), 2207; https://doi.org/10.3390/cells12182207 - 05 Sep 2023
Viewed by 1099
Abstract
An altered expression of miR-143-3p has been previously reported in prostate cancer where it is purported to play a tumor suppressor role. Evidence from other cancers suggests miR-143-3p acts as an inhibitor of epithelial-to-mesenchymal transition (EMT), a key biological process required for metastasis. [...] Read more.
An altered expression of miR-143-3p has been previously reported in prostate cancer where it is purported to play a tumor suppressor role. Evidence from other cancers suggests miR-143-3p acts as an inhibitor of epithelial-to-mesenchymal transition (EMT), a key biological process required for metastasis. However, in prostate cancer the interaction between miR-143-3p and EMT-associated mechanisms remains unclear. Therefore, this paper investigated the link between miR-143-3p and EMT in prostate cancer using in vitro and in silico analyses. PCR detected that miR-143-3p expression was significantly decreased in prostate cancer cell lines compared to normal prostate cells. Bioinformatic analysis of The Cancer Genome Atlas Prostate Adenocarcinoma (TCGA PRAD) data showed a significant downregulation of miR-143-3p in prostate cancer, correlating with pathological markers of advanced disease. Functional enrichment analysis confirmed the significant association of miR-143-3p and its target genes with EMT. The EMT-linked gene AKT1 was subsequently shown to be a novel target of miR-143-3p in prostate cancer cells. The in vitro manipulation of miR-143-3p levels significantly altered the cell proliferation, clonogenicity, migration and expression of EMT-associated markers. Further TCGA PRAD analysis suggested miR-143-3p tumor expression may be a useful predictor of disease recurrence. In summary, this is the first study to report that miR-143-3p overexpression in prostate cancer may inhibit EMT by targeting AKT1. The findings suggest miR-143-3p could be a useful diagnostic and prognostic biomarker for prostate cancer. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Cancers: Prostate Cancer)
Show Figures

Figure 1

17 pages, 3383 KiB  
Article
The SMARCD Family of SWI/SNF Accessory Proteins Is Involved in the Transcriptional Regulation of Androgen Receptor-Driven Genes and Plays a Role in Various Essential Processes of Prostate Cancer
by Iris E. Ertl, Robert Brettner, Hannah Kronabitter, Thomas Mohr, Sophia Derdak, Markus Jeitler, Martin Bilban, Nathalie Garstka and Shahrokh F. Shariat
Cells 2023, 12(1), 124; https://doi.org/10.3390/cells12010124 - 28 Dec 2022
Cited by 5 | Viewed by 1834
Abstract
Previous studies have demonstrated an involvement of chromatin-remodelling SWI/SNF complexes in the development of prostate cancer, suggesting both tumor suppressor and oncogenic activities. SMARCD1/BAF60A, SMARCD2/BAF60B, and SMARCD3/BAF60C are mutually exclusive accessory subunits that confer functional specificity and are components of all known SWI/SNF [...] Read more.
Previous studies have demonstrated an involvement of chromatin-remodelling SWI/SNF complexes in the development of prostate cancer, suggesting both tumor suppressor and oncogenic activities. SMARCD1/BAF60A, SMARCD2/BAF60B, and SMARCD3/BAF60C are mutually exclusive accessory subunits that confer functional specificity and are components of all known SWI/SNF subtypes. To assess the role of SWI/SNF in prostate tumorigenesis, we studied the functions and functional relations of the SMARCD family members. Performing RNA-seq in LnCAP cells grown in the presence or absence of dihydrotestosterone, we found that the SMARCD proteins are involved in the regulation of numerous hormone-dependent AR-driven genes. Moreover, we demonstrated that all SMARCD proteins can regulate AR-downstream targets in androgen-depleted cells, suggesting an involvement in the progression to castration-resistance. However, our approach also revealed a regulatory role for SMARCD proteins through antagonization of AR-signalling. We further demonstrated that the SMARCD proteins are involved in several important cellular processes such as the maintenance of cellular morphology and cytokinesis. Taken together, our findings suggest that the SMARCD proteins play an important, yet paradoxical, role in prostate carcinogenesis. Our approach also unmasked the complex interplay of paralogue SWI/SNF proteins that must be considered for the development of safe and efficient therapies targeting SWI/SNF. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Cancers: Prostate Cancer)
Show Figures

Figure 1

19 pages, 4785 KiB  
Article
SFRP2 Overexpression Induces an Osteoblast-like Phenotype in Prostate Cancer Cells
by Elif Akova Ölken, Attila Aszodi, Hanna Taipaleenmäki, Hiroaki Saito, Veronika Schönitzer, Michael Chaloupka, Maria Apfelbeck, Wolfgang Böcker and Maximilian Michael Saller
Cells 2022, 11(24), 4081; https://doi.org/10.3390/cells11244081 - 16 Dec 2022
Cited by 6 | Viewed by 1860
Abstract
Prostate cancer bone metastasis is still one of the most fatal cancer diagnoses for men. Survival of the circulating prostate tumor cells and their adaptation strategy to survive in the bone niche is the key point to determining metastasis in early cancer stages. [...] Read more.
Prostate cancer bone metastasis is still one of the most fatal cancer diagnoses for men. Survival of the circulating prostate tumor cells and their adaptation strategy to survive in the bone niche is the key point to determining metastasis in early cancer stages. The promoter of SFRP2 gene, encoding a WNT signaling modulator, is hypermethylated in many cancer types including prostate cancer. Moreover, SFRP2 can positively regulate osteogenic differentiation in vitro and in vivo. Here, we showed SFRP2 overexpression in the prostate cancer cell line PC3 induces an epithelial mesenchymal transition (EMT), increases the attachment, and modifies the transcriptome towards an osteoblast-like phenotype (osteomimicry) in a collagen 1-dependent manner. Our data reflect a novel molecular mechanism concerning how metastasizing prostate cancer cells might increase their chance to survive within bone tissue. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Cancers: Prostate Cancer)
Show Figures

Figure 1

24 pages, 10105 KiB  
Article
SCAND1 Reverses Epithelial-to-Mesenchymal Transition (EMT) and Suppresses Prostate Cancer Growth and Migration
by Takanori Eguchi, Eva Csizmadia, Hotaka Kawai, Mona Sheta, Kunihiro Yoshida, Thomas L. Prince, Barbara Wegiel and Stuart K. Calderwood
Cells 2022, 11(24), 3993; https://doi.org/10.3390/cells11243993 - 10 Dec 2022
Cited by 7 | Viewed by 1909
Abstract
Epithelial–mesenchymal transition (EMT) is a reversible cellular program that transiently places epithelial (E) cells into pseudo-mesenchymal (M) cell states. The malignant progression and resistance of many carcinomas depend on EMT activation, partial EMT, or hybrid E/M status in neoplastic cells. EMT is activated [...] Read more.
Epithelial–mesenchymal transition (EMT) is a reversible cellular program that transiently places epithelial (E) cells into pseudo-mesenchymal (M) cell states. The malignant progression and resistance of many carcinomas depend on EMT activation, partial EMT, or hybrid E/M status in neoplastic cells. EMT is activated by tumor microenvironmental TGFβ signal and EMT-inducing transcription factors, such as ZEB1/2, in tumor cells. However, reverse EMT factors are less studied. We demonstrate that prostate epithelial transcription factor SCAND1 can reverse the cancer cell mesenchymal and hybrid E/M phenotypes to a more epithelial, less invasive status and inhibit their proliferation and migration in DU-145 prostate cancer cells. SCAND1 is a SCAN domain-containing protein and hetero-oligomerizes with SCAN-zinc finger transcription factors, such as MZF1, for accessing DNA and the transcriptional co-repression of target genes. We found that SCAND1 expression correlated with maintaining epithelial features, whereas the loss of SCAND1 was associated with mesenchymal phenotypes of tumor cells. SCAND1 and MZF1 were mutually inducible and coordinately included in chromatin with hetero-chromatin protein HP1γ. The overexpression of SCAND1 reversed hybrid E/M status into an epithelial phenotype with E-cadherin and β-catenin relocation. Consistently, the co-expression analysis in TCGA PanCancer Atlas revealed that SCAND1 and MZF1 expression was negatively correlated with EMT driver genes, including CTNNB1, ZEB1, ZEB2 and TGFBRs, in prostate adenocarcinoma specimens. In addition, SCAND1 overexpression suppressed tumor cell proliferation by reducing the MAP3K-MEK-ERK signaling pathway. Of note, in a mouse tumor xenograft model, SCAND1 overexpression significantly reduced Ki-67(+) and Vimentin(+) tumor cells and inhibited migration and lymph node metastasis of prostate cancer. Kaplan–Meier analysis showed high expression of SCAND1 and MZF1 to correlate with better prognoses in pancreatic cancer and head and neck cancers, although with poorer prognosis in kidney cancer. Overall, these data suggest that SCAND1 induces expression and coordinated heterochromatin-binding of MZF1 to reverse the hybrid E/M status into an epithelial phenotype and, inhibits tumor cell proliferation, migration, and metastasis, potentially by repressing the gene expression of EMT drivers and the MAP3K-MEK-ERK signaling pathway. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Cancers: Prostate Cancer)
Show Figures

Figure 1

28 pages, 12899 KiB  
Article
LncRNA/miRNA/mRNA Network Introduces Novel Biomarkers in Prostate Cancer
by Mohammad Taheri, Arash Safarzadeh, Bashdar Mahmud Hussen, Soudeh Ghafouri-Fard and Aria Baniahmad
Cells 2022, 11(23), 3776; https://doi.org/10.3390/cells11233776 - 25 Nov 2022
Cited by 4 | Viewed by 1859
Abstract
The construction of a competing endogenous RNA (ceRNA) network is an important step in the identification of the role of differentially expressed genes in cancers. In the current research, we used a number of bioinformatics tools to construct the ceRNA network in prostate [...] Read more.
The construction of a competing endogenous RNA (ceRNA) network is an important step in the identification of the role of differentially expressed genes in cancers. In the current research, we used a number of bioinformatics tools to construct the ceRNA network in prostate cancer and identify the importance of these modules in predicting the survival of patients with this type of cancer. An assessment of microarray data of prostate cancer and normal samples using the Limma package led to the identification of differential expressed (DE) RNAs that we stratified into mRNA, lncRNA, and miRNAs, resulting in 684 DEmRNAs, including 437 downregulated DEmRNAs (such as TGM4 and SCGB1A1) and 241 upregulated DEmRNAs (such as TDRD1 and CRISP3); 6 DElncRNAs, including 1 downregulated DElncRNA (H19) and 5 upregulated DElncRNAs (such as PCA3 and PCGEM1); and 59 DEmiRNAs, including 30 downregulated DEmiRNAs (such as hsa-miR-1274a and hsa-miR-1274b) and 29 upregulated DEmiRNAs (such as hsa-miR-1268 and hsa-miR-1207-5p). The ceRNA network contained a total of 5 miRNAs, 5 lncRNAs, and 17 mRNAs. We identified hsa-miR-17, hsa-miR-93, hsa-miR-150, hsa-miR-25, PART1, hsa-miR-125b, PCA3, H19, RND3, and ITGB8 as the 10 hub genes in the ceRNA network. According to the ROC analysis, the expression levels of 19 hub genes showed a high diagnostic value. Taken together, we introduce a number of novel promising diagnostic biomarkers for prostate cancer. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Cancers: Prostate Cancer)
Show Figures

Figure 1

13 pages, 1531 KiB  
Article
Cell-Free DNA Sequencing Reveals Gene Variants in DNA Damage Repair Genes Associated with Prognosis of Prostate Cancer Patients
by Verena Lieb, Amer Abdulrahman, Katrin Weigelt, Siegfried Hauch, Michael Gombert, Juan Guzman, Laura Bellut, Peter J. Goebell, Robert Stöhr, Arndt Hartmann, Bernd Wullich, Helge Taubert and Sven Wach
Cells 2022, 11(22), 3618; https://doi.org/10.3390/cells11223618 - 15 Nov 2022
Cited by 1 | Viewed by 1488
Abstract
In the present study, we further analyzed the data obtained in our previous study, where we investigated the cell-free DNA (cfDNA) of 34 progressive prostate cancer patients via targeted sequencing. Here, we studied the occurrence and prognostic impact of sequence variants according to [...] Read more.
In the present study, we further analyzed the data obtained in our previous study, where we investigated the cell-free DNA (cfDNA) of 34 progressive prostate cancer patients via targeted sequencing. Here, we studied the occurrence and prognostic impact of sequence variants according to their clinical pathological significance (CPS) or their functional impact (FI) in 23 DNA damage repair (DDR) genes with a focus on the ATM serine/threonine kinase gene (ATM). All patients had at least one DDR gene with a CPS or FI variant. Kaplan-Meier analysis indicated that the group with a higher number of CPS variants in DDR genes had a shorter time to treatment change (TTC) compared to the group with a lower number of CPS variants (p = 0.038). Analysis of each DDR gene revealed that CPS variants in the ATM gene and FI variants in the nibrin (NBN) gene showed a shorter TTC (p = 0.034 and p = 0.042). In addition, patients with CPS variants in the ATM gene had shorter overall survival (OS; p = 0.022) and disease-specific survival (DSS; p = 0.010) than patients without these variants. Interestingly, patients with CPS variants in seven DDR genes possessed a better OS (p = 0.008) and DSS (p = 0.009), and patients with FI variants in four DDR genes showed a better OS (p = 0.007) and DSS (p = 0.008). Together, these findings demonstrated that the analysis of cfDNA for gene variants in DDR genes provides prognostic information that may be helpful for future temporal and targeted treatment decisions for advanced PCa patients. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Cancers: Prostate Cancer)
Show Figures

Figure 1

Review

Jump to: Research

15 pages, 1567 KiB  
Review
Cancer-Associated Fibroblast: Role in Prostate Cancer Progression to Metastatic Disease and Therapeutic Resistance
by Martina Bedeschi, Noemi Marino, Elena Cavassi, Filippo Piccinini and Anna Tesei
Cells 2023, 12(5), 802; https://doi.org/10.3390/cells12050802 - 04 Mar 2023
Cited by 9 | Viewed by 2288
Abstract
Prostate cancer (PCa) is one of the most common cancers in European males. Although therapeutic approaches have changed in recent years, and several new drugs have been approved by the Food and Drug Administration (FDA), androgen deprivation therapy (ADT) remains the standard of [...] Read more.
Prostate cancer (PCa) is one of the most common cancers in European males. Although therapeutic approaches have changed in recent years, and several new drugs have been approved by the Food and Drug Administration (FDA), androgen deprivation therapy (ADT) remains the standard of care. Currently, PCa represents a clinical and economic burden due to the development of resistance to ADT, paving the way to cancer progression, metastasis, and to long-term side effects induced by ADT and radio-chemotherapeutic regimens. In light of this, a growing number of studies are focusing on the tumor microenvironment (TME) because of its role in supporting tumor growth. Cancer-associated fibroblasts (CAFs) have a central function in the TME because they communicate with prostate cancer cells, altering their metabolism and sensitivity to drugs; hence, targeted therapy against the TME, and, in particular, CAFs, could represent an alternative therapeutic approach to defeat therapy resistance in PCa. In this review, we focus on different CAF origins, subsets, and functions to highlight their potential in future therapeutic strategies for prostate cancer. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Cancers: Prostate Cancer)
Show Figures

Figure 1

26 pages, 849 KiB  
Review
Boosting the Immune Response—Combining Local and Immune Therapy for Prostate Cancer Treatment
by Jakub Karwacki, Aleksander Kiełbik, Wojciech Szlasa, Natalia Sauer, Kamil Kowalczyk, Wojciech Krajewski, Jolanta Saczko, Julita Kulbacka, Tomasz Szydełko and Bartosz Małkiewicz
Cells 2022, 11(18), 2793; https://doi.org/10.3390/cells11182793 - 07 Sep 2022
Cited by 4 | Viewed by 3250
Abstract
Due to its slow progression and susceptibility to radical forms of treatment, low-grade PC is associated with high overall survival (OS). With the clinical progression of PC, the therapy is becoming more complex. The immunosuppressive tumor microenvironment (TME) makes PC a difficult target [...] Read more.
Due to its slow progression and susceptibility to radical forms of treatment, low-grade PC is associated with high overall survival (OS). With the clinical progression of PC, the therapy is becoming more complex. The immunosuppressive tumor microenvironment (TME) makes PC a difficult target for most immunotherapeutics. Its general immune resistance is established by e.g., immune evasion through Treg cells, synthesis of immunosuppressive mediators, and the defective expression of surface neoantigens. The success of sipuleucel-T in clinical trials initiated several other clinical studies that specifically target the immune escape of tumors and eliminate the immunosuppressive properties of the TME. In the settings of PC treatment, this can be commonly achieved with radiation therapy (RT). In addition, focal therapies usually applied for localized PC, such as high-intensity focused ultrasound (HIFU) therapy, cryotherapy, photodynamic therapy (PDT), and irreversible electroporation (IRE) were shown to boost the anti-cancer response. Nevertheless, the present guidelines restrict their application to the context of a clinical trial or a prospective cohort study. This review explains how RT and focal therapies enhance the immune response. We also provide data supporting the combination of RT and focal treatments with immune therapies. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Cancers: Prostate Cancer)
Show Figures

Figure 1

Back to TopTop