Treatment for Bladder Cancer

A special issue of Cancers (ISSN 2072-6694). This special issue belongs to the section "Cancer Therapy".

Deadline for manuscript submissions: closed (30 April 2021) | Viewed by 13281

Special Issue Editor


E-Mail Website
Guest Editor
Department of Urology & Andrology, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria
Interests: non-muscle invasive bladder cancer; muscle invasive bladder cancer; robotic surgery
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues, 

After years of stable therapy approaches in non-muscle invasive bladder cancer (NMIBC) and muscle invasive bladder cancer (MIBC), uro-oncology dealing with this frequent tumour entity is finally facing several paradigmatic changes.

Nowadays, local and systemic immunotherapies are used for the management of bladder cancer (BC) in daily practice. BCG has been administered since 1976 in patients with NMIBC. Despite its efficacy, disease progression is observed in nearly 30% of patients. Given the antitumor activity of immune checkpoint inhibitors in metastatic setting, these therapies are currently investigated in NMIBC. Pembrolizumab is now approved by the Food and Drug Administration (FDA) for the treatment of patients with BCG-unresponsive, high-risk, NMIBC with carcinoma in situ with or without papillary tumors who are ineligible for or have not elected to undergo cystectomy. Several phase 2 and 3 trials are ongoing to investigate the efficacy of programmed cell death PD(L)1 inhibitors combined with BCG all over the world.

Although anti-programmed cell death 1 (PD-1) monoclonal antibody (mAb) therapy has been established as one of the standard therapies for BC many unknown factors remain to be resolved concerning the mechanism of anti–PD-1 mAb therapy. PD-1 is expressed on the cell surface of activated cytotoxic T lymphocytes (CTLs) that can recognize certain tumor antigens and acquire tumoricidal activity. Also, when PD-1 binds to programmed cell death ligand 1 (PD-L1) on tumor cells, a suppressive signal will be sent to CTLs, resulting in inactivation of CTLs and immune escape of the tumor cells. Treatment with anti–PD-1 or anti–PD-L1 mAbs inhibits the interaction of PD-1 and PD-L1 and revitalizes the tumoricidal CTLs, providing significant antitumor activity, even to highly advanced malignant tumors. Thus, PD-L1 expression in tumor tissue seemed to represent a promising prognostic factor for anti–PD-1 mAb therapy against BC.

However, as it became clear that anti–PD-1 mAb therapy might be effective in BC without PD-L1 expression, PD-L1 expression in tumor tissue as a biomarker for anti–PD-1 mAb therapy should be reconsidered. Thus, the mechanism of how anti–PD-1 mAb therapy is effective against PD-L1 BC warrants further investigation. Recently, it was suggested that blockade between PD-1 on T cells and PD-L1 on antigen-presenting cells (APCs) could induce antitumor immunity, even if tumor cells have no PD-L1 expression.

Although it is known that the blood levels of soluble programmed cell death ligand 1 (sPD-L1) are elevated in various malignancies, the nature of sPD-L1 has not been thoroughly elucidated especially for BC. In this special issue of CANCERS, we are focusing this potential novel biomarker.

Additionally in December 2019, the FDA granted accelerated approval to the novel nectin-4-targeting antibody-drug conjugate, enfortumab vedotin, for the treatment of platinum-refractory and immune checkpoint blockade-refractory locally advanced or metastatic urothelial carcinoma. Antibody-drug conjugates represent a new therapeutic modality in urothelial cancer this other appealing new therapies are merging.

Prof. Dr. Lukas Lusuardi
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. Cancers 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 2900 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

  • non-muscle invasive bladder cancer
  • muscle invasive bladder cancer
  • robotic surgery

Published Papers (3 papers)

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

Research

Jump to: Review, Other

14 pages, 3233 KiB  
Article
Ellagic Acid Resensitizes Gemcitabine-Resistant Bladder Cancer Cells by Inhibiting Epithelial-Mesenchymal Transition and Gemcitabine Transporters
by Ying-Si Wu, Jar-Yi Ho, Cheng-Ping Yu, Chun-Jung Cho, Chia-Lun Wu, Cheng-Shuo Huang, Hong-Wei Gao and Dah-Shyong Yu
Cancers 2021, 13(9), 2032; https://doi.org/10.3390/cancers13092032 - 22 Apr 2021
Cited by 13 | Viewed by 2806
Abstract
Gemcitabine (GCB) resistance is a major issue in bladder cancer chemoresistance, but its underlying mechanism has not been determined. Epithelial-mesenchymal transition (EMT) has been shown to be comprehensively involved in GCB resistance in several other cancer types, but the direct connection between EMT [...] Read more.
Gemcitabine (GCB) resistance is a major issue in bladder cancer chemoresistance, but its underlying mechanism has not been determined. Epithelial-mesenchymal transition (EMT) has been shown to be comprehensively involved in GCB resistance in several other cancer types, but the direct connection between EMT and GCB remains unclear. This study was designed to elucidate the mechanism of EMT-related GCB resistance in bladder cancer and identify a potential phytochemical to modulate drug sensitivity. The biological effects of ellagic acid (EA) or its combined effects with GCB were compared in GCB-resistant cells and the GCB-sensitive line in terms of cell viability, apoptosis, motility, and in vivo tumorigenicity. The molecular regulation of EMT-related GCB resistance was evaluated at both the mRNA and protein expression levels. Our results indicated that TGF-β/Smad induced the overactivation of EMT in GCB-resistant cells and reduced the expression of GCB influx transporters (hCNT1 and hENT1). Moreover, ellagic acid (EA) inhibited the TGF-β signaling pathway both in vitro and in vivo by reducing Smad2, Smad3, and Smad4 expression and thereby resensitized GCB sensitivity. In conclusion, our results demonstrate that TGF-β/Smad-induced EMT contributes to GCB resistance in bladder cancer by reducing GCB influx and also elucidate the novel mechanisms of EA-mediated inhibition of TGF-β/Smad-induced EMT to overcome GCB resistance. Our study warrants further investigation of EA as an effective therapeutic adjuvant agent for overcoming GCB resistance in bladder cancer. Full article
(This article belongs to the Special Issue Treatment for Bladder Cancer)
Show Figures

Figure 1

Review

Jump to: Research, Other

29 pages, 3012 KiB  
Review
Nanotechnology in Bladder Cancer: Diagnosis and Treatment
by Mahmood Barani, Seyedeh Maryam Hosseinikhah, Abbas Rahdar, Leila Farhoudi, Rabia Arshad, Magali Cucchiarini and Sadanand Pandey
Cancers 2021, 13(9), 2214; https://doi.org/10.3390/cancers13092214 - 5 May 2021
Cited by 58 | Viewed by 6817
Abstract
Bladder cancer (BC) is the second most common cancer of the urinary tract in men and the fourth most common cancer in women, and its incidence rises with age. There are many conventional methods for diagnosis and treatment of BC. There are some [...] Read more.
Bladder cancer (BC) is the second most common cancer of the urinary tract in men and the fourth most common cancer in women, and its incidence rises with age. There are many conventional methods for diagnosis and treatment of BC. There are some current biomarkers and clinical tests for the diagnosis and treatment of BC. For example, radiotherapy combined with chemotherapy and surgical, but residual tumor cells mostly cause tumor recurrence. In addition, chemotherapy after transurethral resection causes high side effects, and lack of selectivity, and low sensitivity in sensing. Therefore, it is essential to improve new procedures for the diagnosis and treatment of BC. Nanotechnology has recently sparked an interest in a variety of areas, including medicine, chemistry, physics, and biology. Nanoparticles (NP) have been used in tumor therapies as appropriate tools for enhancing drug delivery efficacy and enabling therapeutic performance. It is noteworthy, nanomaterial could be reduced the limitation of conventional cancer diagnosis and treatments. Since, the major disadvantages of therapeutic drugs are their insolubility in an aqueous solvent, for instance, paclitaxel (PTX) is one of the important therapeutic agents utilized to treating BC, due to its ability to prevent cancer cell growth. However, its major problem is the poor solubility, which has confirmed to be a challenge when improving stable formulations for BC treatment. In order to reduce this challenge, anti-cancer drugs can be loaded into NPs that can improve water solubility. In our review, we state several nanosystem, which can effective and useful for the diagnosis, treatment of BC. We investigate the function of metal NPs, polymeric NPs, liposomes, and exosomes accompanied therapeutic agents for BC Therapy, and then focused on the potential of nanotechnology to improve conventional approaches in sensing. Full article
(This article belongs to the Special Issue Treatment for Bladder Cancer)
Show Figures

Figure 1

Other

Jump to: Research, Review

22 pages, 679 KiB  
Systematic Review
Current Systemic Treatment Options in Metastatic Urothelial Carcinoma after Progression on Checkpoint Inhibition Therapy—A Systemic Review Combined with Single-Group Meta-Analysis of Three Studies Testing Enfortumab Vedotin
by Susanne Deininger, Peter Törzsök, David Oswald and Lukas Lusuardi
Cancers 2021, 13(13), 3206; https://doi.org/10.3390/cancers13133206 - 26 Jun 2021
Cited by 7 | Viewed by 2993
Abstract
Background: In the first and second-line therapy of metastatic urothelial carcinoma (mUC), checkpoint inhibitors (CPI) such as Pembrolizumab and Atezolizumab have been widely implemented. Little is currently known about what therapeutic options are effective after therapy with CPI. This article presents a systemic [...] Read more.
Background: In the first and second-line therapy of metastatic urothelial carcinoma (mUC), checkpoint inhibitors (CPI) such as Pembrolizumab and Atezolizumab have been widely implemented. Little is currently known about what therapeutic options are effective after therapy with CPI. This article presents a systemic review of current treatment options in this setting. Methods: From August 2020 to 15 April 2021, a literature search was performed through the PubMed/Medline. Subsequently, a single-group meta-analysis of three studies testing Enfortumab vedotin (EV) was conducted. Results: Five therapy regimens tested in the post-CPI setting with adequate data were identified: Chemotherapy (CT), Ramucirumab plus Docetaxel, Erdafitinib (Erd), EV, and Sacituzumab govitecan (SG). In n = 74 + 125 + 288 patients, the single-group meta-analysis showed an objective response rate of 42.1% for EV compared to 17.9% for CT in a similar setting. EV was also ahead in progression free survival (5.9 months with EV vs. 3.7 months with CT) and overall survival (12.8 months with EV vs. 9.0 months with CT). Conclusion: Most data are currently available for EV. Further research is needed on the question of which patients’ subcollectives particularly benefit from which therapeutic approach. Full article
(This article belongs to the Special Issue Treatment for Bladder Cancer)
Show Figures

Figure 1

Back to TopTop