1
|
Heard JR, Ahdoot M, Theodorescu D, Mitra AP. Biomarkers of treatment response in bladder cancer. Expert Rev Mol Diagn 2024; 24:957-969. [PMID: 39535158 DOI: 10.1080/14737159.2024.2428747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION There have been many recent advancements in the treatment of bladder cancer including the approval of novel intravesical agents for non-muscle-invasive disease and systemic-targeted therapeutics for muscle-invasive and advanced disease. However, treatment strategies for bladder cancer are still largely based on clinicopathologic characteristics. AREAS COVERED Based on primary literature sourced from PubMed, Embase, and Cochrane Library, we review the current status of molecular markers and biomarker panels with respective to their value in predicting response to standard chemotherapeutics and novel agents in non-muscle-invasive, muscle-invasive, and advanced bladder cancer. EXPERT OPINION Several biomarkers based on molecular characterization of tumors and quantification of circulating tumor DNA have been associated with response or resistance to standard chemotherapeutics. More recent investigations have reported on predictive biomarkers for novel therapeutics in bladder cancer, although large-scale validation is still needed. Given the increasing therapeutic options for this disease, employment of such predictive biomarkers may help guide treatment selection and sequencing.
Collapse
Affiliation(s)
- John R Heard
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Michael Ahdoot
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Dan Theodorescu
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Anirban P Mitra
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
2
|
Zhu L, Liu X, Zhang W, Hu H, Wang Q, Xu K. MTHFD2 is a potential oncogene for its strong association with poor prognosis and high level of immune infiltrates in urothelial carcinomas of bladder. BMC Cancer 2022; 22:556. [PMID: 35581573 PMCID: PMC9112551 DOI: 10.1186/s12885-022-09606-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 04/25/2022] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND The bifunctional methylenetetrahydrofolate dehydrogenase (NADP+ dependent) 2, methenyltetrahydrofolate cyclohydrolase (MTHFD2) has been reported to play an oncogenic role in various types of cancers. However, the function of MTHFD2 in urothelial carcinomas of bladder (UCB) and its association with tumor immune infiltration remains unknown. We aim to examine the suitability of MTHFD2 to be a novel biomarker of bladder cancer and whether MTHFD2 is linked to immune infiltration. METHODS RNA sequencing data and clinical information (bladder cancer samples: normal samples = 414: 19) were downloaded from The Cancer Genome Atlas official website. Western blot analysis was performed to detect MTHFD2 expression in human bladder cancer (BLCA) cells and normal urothelial cell line SV-HUC-1. Associations between MTHFD2 expression and clinicopathological features were analyzed using Mann Whitney U test or Kruskal-Wallis H test. The "survival" and "survminer" packages were utilized to plot Kaplan-Meier survival curves. Moreover, the gene set enrichment analysis (GSEA) was conducted using a clusterProfiler package. The correlation of MTHFD2 expression with immune infiltration level was estimated using the single sample GSEA (ssGSEA) algorithm. Furthermore, associations between MTHFD2 and immune checkpoint genes were evaluated using the correlation analysis. RESULTS Transcriptome analysis manifested that MTHFD2 was highly expressed in UCB tissues than normal bladder tissues, which was further confirmed by western blot analysis in human BLCA cells and SV-HUC-1 cells. Moreover, MTHFD2 high expression was significantly associated with the advanced disease progression. Also, the high expression of MTHFD2 was correlated with poor prognosis, and MTHFD2 was considered as an independent prognostic factor for disease specific survival. Furthermore, a number of cancer-related pathways were enriched in MTHFD2 high group, including NF-κB activation, JAK/STAT, and cancer immunotherapy by PD1 blockade. Several immune checkpoint molecules were also strongly associated with MTHFD2 expression, including PDCD1, CD274, CTLA4, CD276, LAG3, HAVCR2, and TIGIT. CONCLUSIONS MTHFD2 expression was remarkably elevated in UCB, suggesting that MTHFD2 could be a promising biomarker for BLCA as well as novel target for anti-cancer immunotherapy since its close association with immune infiltration.
Collapse
Affiliation(s)
- Lin Zhu
- Department of Urology, Peking University People's Hospital, 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Xianhui Liu
- Department of Urology, Peking University People's Hospital, 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Weiyu Zhang
- Department of Urology, Peking University People's Hospital, 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China.,Peking University Applied Lithotripsy Institute, Peking University People's Hospital, Beijing, 100034, China
| | - Hao Hu
- Department of Urology, Peking University People's Hospital, 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Qi Wang
- Department of Urology, Peking University People's Hospital, 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Kexin Xu
- Department of Urology, Peking University People's Hospital, 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China.
| |
Collapse
|
3
|
Lyu ZJ, Wang Y, Huang JL, Chen M, Wu SY, Yan Q, Zhang Y, Tang Y, Jiang C, Li L, Jia YZ, Liu YC, Mei HB, Wang F, Li RH, Chen YC, Lin X, Cai ZM, Guan XY. Recurrent ZNF83-E293V Mutation Promotes Bladder Cancer Progression through the NF-κB Pathway via Transcriptional Dysregulation of S100A8. Mol Ther 2021; 29:275-290. [PMID: 33002420 PMCID: PMC7791007 DOI: 10.1016/j.ymthe.2020.09.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 08/07/2020] [Accepted: 09/01/2020] [Indexed: 02/05/2023] Open
Abstract
Urothelial carcinoma (UC) is the predominant form of bladder cancer. Significant molecular heterogeneity caused by diverse molecular alterations brings about large variations in the response to treatment in UC. An improved understanding of the genetic mechanisms underlying the development and progression of UC is essential. Through deep analysis of next-generation sequencing data of 99 UC patients, we found that 18% of cases had recurrent somatic mutations in zinc finger protein gene zinc finger protein 83 (ZNF83). ZNF83 mutations were correlated with poor prognosis of UC. We also found a hotspot mutation, p.E293V, in the evolutionarily well-conserved region of ZNF83. ZNF83-E293V increased tumor growth and reduced the apoptosis of UC cells compared to wild-type ZNF83 both in vitro and in mice xenografted tumors. ZNF83-E293V activated nuclear factor κB (NF-κB) more potently than did the wild-type protein owing to its decreased transcriptional repression for S100A8. The NF-κB inhibitors could pharmacologically block the tumor growth in mice engrafted with ZNF83-E293V-transfected UC cells. These findings provide a mechanistic insight and a potential therapeutic strategy for UC, which established a foundation for using the ZNF83-E293V mutation as a predictive biomarker of therapeutic response from NF-κB inhibitors.
Collapse
Affiliation(s)
- Zhao J Lyu
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518029, China; Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Ying Wang
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518029, China; Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China; Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510030, China; State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou 510030, China
| | - Jin L Huang
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China; Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou 510030, China
| | - Miao Chen
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Sha Y Wu
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Qian Yan
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Yu Zhang
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Ying Tang
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Chen Jiang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou 510030, China; Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou 510030, China
| | - Lei Li
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China; State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou 510030, China
| | - Yi Z Jia
- Core Laboratory, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518029, China
| | - Yu C Liu
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518029, China
| | - Hong B Mei
- Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518029, China
| | - Feng Wang
- Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518029, China
| | - Ren H Li
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Ya C Chen
- Department of Pathology, Shenzhen University General Hospital, Shenzhen 518055, China
| | - Xiang Lin
- School of Chinese Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Zhi M Cai
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518029, China; Department of Urology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China; Carson International Cancer Center, School of Medicine, Shenzhen University, Shenzhen 518061, China; BGI-Medicine, BGI, Shenzhen 518083, China.
| | - Xin Y Guan
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518029, China; Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China; State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou 510030, China.
| |
Collapse
|
4
|
Zolotovskaia M, Sorokin M, Garazha A, Borisov N, Buzdin A. Molecular Pathway Analysis of Mutation Data for Biomarkers Discovery and Scoring of Target Cancer Drugs. Methods Mol Biol 2020; 2063:207-234. [PMID: 31667773 DOI: 10.1007/978-1-0716-0138-9_16] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
DNA mutations govern cancer development. Cancer mutation profiles vary dramatically among the individuals. In some cases, they may serve as the predictors of disease progression and response to therapies. However, the biomarker potential of cancer mutations can be dramatically (several orders of magnitude) enhanced by applying molecular pathway-based approach. We developed Oncobox system for calculation of pathway instability (PI) values for the molecular pathways that are aggregated mutation frequencies of the pathway members normalized on gene lengths and on number of genes in the pathway. PI scores can be effective biomarkers in different types of comparisons, for example, as the cancer type biomarkers and as the predictors of tumor response to target therapies. The latter option is implemented using mutation drug score (MDS) values, which algorithmically rank the drugs capacity of interfering with the mutated molecular pathways. Here, describe the mathematical basis and algorithms for PI and MDS values calculation, validation and implementation. The example analysis is provided encompassing 5956 human tumor mutation profiles of 15 cancer types from The Cancer Genome Atlas (TCGA) project, that totally make 2,316,670 mutations in 19,872 genes and 1748 molecular pathways, thus enabling ranking of 128 clinically approved target drugs. Our results evidence that the Oncobox PI and MDS approaches are highly useful for basic and applied aspects of molecular oncology and pharmacology research.
Collapse
Affiliation(s)
- Marianna Zolotovskaia
- Omicsway Corp., Walnut, CA, USA
- Department of Oncology, Hematology and Radiotherapy of Pediatric Faculty, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Maxim Sorokin
- Omicsway Corp., Walnut, CA, USA
- Laboratory of Clinical Bioinformatics, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | | | - Nikolay Borisov
- Omicsway Corp., Walnut, CA, USA
- Laboratory of Clinical Bioinformatics, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Anton Buzdin
- Omicsway Corp., Walnut, CA, USA.
- Laboratory of Clinical Bioinformatics, I.M. Sechenov First Moscow State Medical University, Moscow, Russia.
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia.
| |
Collapse
|
5
|
Zolotovskaia MA, Sorokin MI, Roumiantsev SA, Borisov NM, Buzdin AA. Pathway Instability Is an Effective New Mutation-Based Type of Cancer Biomarkers. Front Oncol 2019; 8:658. [PMID: 30662873 PMCID: PMC6328788 DOI: 10.3389/fonc.2018.00658] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 12/12/2018] [Indexed: 01/20/2023] Open
Abstract
DNA mutations play a crucial role in cancer development and progression. Mutation profiles vary dramatically in different cancer types and between individual tumors. Mutations of several individual genes are known as reliable cancer biomarkers, although the number of such genes is tiny and does not enable differential diagnostics for most of the cancers. We report here a technique enabling dramatically increased efficiency of cancer biomarkers development using DNA mutations data. It includes a quantitative metric termed Pathway instability (PI) based on mutations enrichment of intracellular molecular pathways. This method was tested on 5,956 tumor mutation profiles of 15 cancer types from The Cancer Genome Atlas (TCGA) project. Totally, we screened 2,316,670 mutations in 19,872 genes and 1,748 molecular pathways. Our results demonstrated considerable advantage of pathway-based mutation biomarkers over individual gene mutation profiles, as reflected by more than two orders of magnitude greater numbers by high-quality [ROC area-under-curve (AUC)>0.75] biomarkers. For example, the number of such high-quality mutational biomarkers distinguishing between different cancer types was only six for the individual gene mutations, and already 660 for the pathway-based biomarkers. These results evidence that PI value can be used as a new generation of complex cancer biomarkers significantly outperforming the existing gene mutation biomarkers.
Collapse
Affiliation(s)
- Marianna A Zolotovskaia
- Department of Oncology, Hematology and Radiotherapy of Pediatric Faculty, Pirogov Russian National Research Medical University, Moscow, Russia.,Oncobox Ltd., Moscow, Russia
| | - Maxim I Sorokin
- The Laboratory of Clinical Bioinformatics, I.M. Sechenov First Moscow State Medical University, Moscow, Russia.,Omicsway Corp., Walnut, CA, United States
| | - Sergey A Roumiantsev
- Department of Oncology, Hematology and Radiotherapy of Pediatric Faculty, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Nikolay M Borisov
- Oncobox Ltd., Moscow, Russia.,The Laboratory of Clinical Bioinformatics, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Anton A Buzdin
- The Laboratory of Clinical Bioinformatics, I.M. Sechenov First Moscow State Medical University, Moscow, Russia.,Omicsway Corp., Walnut, CA, United States.,The Laboratory of Systems Biology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| |
Collapse
|
6
|
Abstract
Purpose Use of molecular markers in urine, tissue or blood offers potential opportunities to improve understanding of bladder cancer biology which may help identify disease earlier, risk stratify patients, improve prediction of outcomes or help target therapy. Methods A review of the published literature was performed, without restriction of time. Results Despite the fast-growing literature about the topic and the approval of several urinary biomarkers for use in clinical practice, they have not reached the level of evidence for widespread utilization. Biomarkers could be used in different clinical scenarios, mainly to overcome the limitations of current diagnostic, predictive, and prognostic tools. They have been evaluated to detect bladder cancer in asymptomatic populations or those with hematuria and in surveillance of disease as adjuncts to cystoscopy. There is also a potential role as prognosticators of disease recurrence, progression and survival both in patients with non-invasive cancers and in those with advanced disease. Finally, they promise to be helpful in predicting the response to local and/or systemic chemotherapy and/or immunotherapy. Conclusions To date, due to the lack of high-quality prospective trials, the level of evidence provided by the current literature remains low and, therefore, the potential of biomarkers exceeds utilization in clinical practice.
Collapse
|
7
|
Worst TS, Weis CA, Stöhr R, Bertz S, Eckstein M, Otto W, Breyer J, Hartmann A, Bolenz C, Wirtz RM, Erben P. CDKN2A as transcriptomic marker for muscle-invasive bladder cancer risk stratification and therapy decision-making. Sci Rep 2018; 8:14383. [PMID: 30258198 PMCID: PMC6158275 DOI: 10.1038/s41598-018-32569-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 09/10/2018] [Indexed: 12/22/2022] Open
Abstract
Deletions of the cell cycle control gene CDKN2A are described as progression markers of non-muscle invasive bladder cancer and to be associated with fibroblast growth factor 3 (FGFR3) mutations. The prognostic role of CDKN2A RNA expression in muscle invasive bladder cancer (MIBC) is under discussion. In 80 MIBC patients (m/f 60/20) who underwent radical cystectomy the expression of CDKN2A and FGFR3 was examined with qRT-PCR (test cohort). The MDA cohort (n = 57) and the TCGA cohort (n = 365) served for validation. The expression of drug target genes and TCGA molecular subtypes was correlated with CDKN2A expression. In the test cohort CDKN2Ahigh patients (n = 8; 10.0%) had a significantly shorter recurrence-free (p = 0.018) and disease-specific (p = 0.006) survival compared to the rest of the cohort. A similar stratification was seen in the validation cohorts (CDKN2Ahigh: n = 7, 12.3%, p = 0.001; n = 46, 12.6%, p = 0.011). In the TCGA cohort these patients had a comparably low expression of drug target genes. The expression of CDKN2A significantly differed among TGCA molecular subtypes. 71.7% of CDKN2Ahigh were TCGA basal squamous tumours but also show divergent molecular features compared to this group. In summary CDKN2A RNA expression-based risk stratification of MIBC allows the identification of a CDKN2Ahigh poor prognosis group with low expression of drug target genes.
Collapse
Affiliation(s)
- Thomas S Worst
- Department of Urology, University Medical Center Mannheim, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
| | - Cleo-Aron Weis
- Institute of Pathology, University Medical Center Mannheim, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Robert Stöhr
- Institute of Pathology, University of Erlangen-Nuremberg, Krankenhausstraße 8-10, 91054, Erlangen, Germany
| | - Simone Bertz
- Institute of Pathology, University of Erlangen-Nuremberg, Krankenhausstraße 8-10, 91054, Erlangen, Germany
| | - Markus Eckstein
- Institute of Pathology, University of Erlangen-Nuremberg, Krankenhausstraße 8-10, 91054, Erlangen, Germany
| | - Wolfgang Otto
- Department of Urology, University of Regensburg, Landshuter Straße 65, 93053, Regensburg, Germany
| | - Johannes Breyer
- Department of Urology, University of Regensburg, Landshuter Straße 65, 93053, Regensburg, Germany
| | - Arndt Hartmann
- Institute of Pathology, University of Erlangen-Nuremberg, Krankenhausstraße 8-10, 91054, Erlangen, Germany
| | - Christian Bolenz
- Department of Urology, University of Ulm, Prittwitzstraße 43, 89075, Ulm, Germany
| | - Ralph M Wirtz
- STRATIFYER Molecular Pathology GmbH, Werthmannstraße 1, 50935, Cologne, Germany
- Institute of Pathology at the St Elisabeth Hospital Köln-Hohenlind, Werthmannstraße 1, 50935, Cologne, Germany
| | - Philipp Erben
- Department of Urology, University Medical Center Mannheim, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| |
Collapse
|
8
|
Risk Factors and Molecular Features Associated with Bladder Cancer Development. MOLECULAR PATHOLOGY LIBRARY 2018. [DOI: 10.1007/978-3-319-64769-2_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
9
|
Abstract
Clinical outcomes for patients with bladder cancer have largely remained unchanged over the last three decades despite improvements in surgical techniques, perioperative therapies, and postoperative management. Current management still heavily relies on pathologic staging that does not always reflect an individual patient's risk. The genesis and progression of bladder cancer is now increasingly recognized as being a result of alterations in several pathways that affect the cell cycle, apoptosis, cellular signaling, gene regulation, immune modulation, angiogenesis, and tumor cell invasion. Multiplexed assessment of biomarkers associated with alterations in these pathways offers novel insights into tumor behavior while identifying panels that are capable of reproducibly predicting patient outcomes. Future management of bladder cancer will likely incorporate such prognostic molecular models for risk stratification and treatment personalization.
Collapse
Affiliation(s)
- Anirban P Mitra
- Institute of Urology, Keck School of Medicine of the University of Southern California, 1441 Eastlake Avenue, Suite 7416, MC 9178, Los Angeles, CA, 90033, USA.
| | - Siamak Daneshmand
- Institute of Urology, Keck School of Medicine of the University of Southern California, 1441 Eastlake Avenue, Suite 7416, MC 9178, Los Angeles, CA, 90033, USA
| |
Collapse
|
10
|
Soave A, Riethdorf S, Dahlem R, von Amsberg G, Minner S, Weisbach L, Engel O, Fisch M, Pantel K, Rink M. A nonrandomized, prospective, clinical study on the impact of circulating tumor cells on outcomes of urothelial carcinoma of the bladder patients treated with radical cystectomy with or without adjuvant chemotherapy. Int J Cancer 2016; 140:381-389. [DOI: 10.1002/ijc.30445] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 09/07/2016] [Indexed: 12/29/2022]
Affiliation(s)
- Armin Soave
- Department of Urology; University Medical Center Hamburg-Eppendorf; Hamburg Germany
| | - Sabine Riethdorf
- Institute of Tumor Biology; University Medical Center Hamburg-Eppendorf; Hamburg Germany
| | - Roland Dahlem
- Department of Urology; University Medical Center Hamburg-Eppendorf; Hamburg Germany
| | - Gunhild von Amsberg
- Department of Hematology and Oncology; University Medical Center Hamburg-Eppendorf; Hamburg Germany
| | - Sarah Minner
- Department of Pathology; University Medical Center Hamburg-Eppendorf; Hamburg Germany
| | - Lars Weisbach
- Department of Urology; University Medical Center Hamburg-Eppendorf; Hamburg Germany
| | - Oliver Engel
- Department of Urology; University Medical Center Hamburg-Eppendorf; Hamburg Germany
| | - Margit Fisch
- Department of Urology; University Medical Center Hamburg-Eppendorf; Hamburg Germany
| | - Klaus Pantel
- Institute of Tumor Biology; University Medical Center Hamburg-Eppendorf; Hamburg Germany
| | - Michael Rink
- Department of Urology; University Medical Center Hamburg-Eppendorf; Hamburg Germany
| |
Collapse
|
11
|
Knapp DW, Ruple-Czerniak A, Ramos-Vara JA, Naughton JF, Fulkerson CM, Honkisz SI. A Nonselective Cyclooxygenase Inhibitor Enhances the Activity of Vinblastine in a Naturally-Occurring Canine Model of Invasive Urothelial Carcinoma. Bladder Cancer 2016; 2:241-250. [PMID: 27376143 PMCID: PMC4927831 DOI: 10.3233/blc-150044] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background: Chemotherapy is expected to remain an important part of invasive urothelial carcinoma (UC) treatment. Strategies to enhance chemotherapy efficacy are needed. Objective: To determine the chemotherapy-enhancing effects of a nonselective cyclooxygenase (COX) inhibitor on vinblastine in a naturally-occurring canine model of invasive UC. Methods: With IACUC approval, privately-owned dogs with naturally-occurring histologically-diagnosed invasive UC, expected survival ≥6 weeks, and informed owner consent were randomly allocated to receive vinblastine (2.5 mg/m2 intravenously every 2 weeks) plus piroxicam (0.3 mg/kg daily per os) or vinblastine alone (same dose) with the option to receive piroxicam alone when vinblastine failed. Scheduled evaluations included physical exam, standard laboratory analyses, thoracic radiography, abdominal ultrasonography, and standardized measurement of urinary tract tumors. Results: Dogs receiving vinblastine alone (n = 27) and vinblastine-piroxicam (n = 24) were similar in age, sex, breed, tumor stage, and grade. Remission occurred more frequently (P < 0.02) with vinblastine-piroxicam (58.3%) than with vinblastine alone (22.2%). The median progression free interval was 143 days with vinblastine alone and 199 days with the combination. Interestingly, the overall median survival time was significantly longer (P < 0.03) in dogs receiving vinblastine alone followed by piroxicam alone (n = 20, 531 days) than in dogs receiving the combination (299 days). Treatment was well tolerated in both arms. Conclusions: Piroxicam significantly enhanced the activity of vinblastine in dogs with UC where the cancer closely mimics the human condition, clearly justifying further study. The study suggest the potential importance of tracking COX inhibitor use in patients in clinical trials as COX inhibitors could affect treatment response.
Collapse
Affiliation(s)
- Deborah W Knapp
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, IN, USA; Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, USA
| | | | - José A Ramos-Vara
- Department of Comparative Pathobiology, Purdue University , West Lafayette, IN, USA
| | | | | | - Sonia I Honkisz
- Department of Veterinary Clinical Sciences, Purdue University , West Lafayette, IN, USA
| |
Collapse
|
12
|
Mitra AP. Molecular substratification of bladder cancer: moving towards individualized patient management. Ther Adv Urol 2016; 8:215-33. [PMID: 27247631 DOI: 10.1177/1756287216638981] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Despite advances in surgical techniques, perioperative therapies and postoperative management, outcomes for patients with bladder cancer have largely remained unchanged. Current management of bladder cancer still relies on pathologic staging that does not always reflect the risk for an individual patient. Studies assessing molecular alterations in individual tumors are offering insights into the myriad of cellular pathways that are deregulated in bladder tumorigenesis and progression. Alterations in pathways involved in cell-cycle regulation, apoptosis, cell signaling, angiogenesis and tumor-cell invasion have been shown to influence disease behavior. High-throughput assays are now allowing multiplexed assessment of biomarker alterations, thereby enabling characterization of novel molecular subtypes of bladder cancer. Such approaches have also been used for discovery and validation of robust prognostic molecular signatures. The future of bladder cancer management will rely on the use of validated multimarker panels for risk stratification, optimal surgical management, and theranostic strategies to identify and target specific alterations in individual tumors.
Collapse
Affiliation(s)
- Anirban P Mitra
- Institute of Urology, University of Southern California, 1441 Eastlake Avenue, Suite 7416, MC 9178, Los Angeles, CA 90033, USA
| |
Collapse
|
13
|
Azevedo R, Ferreira JA, Peixoto A, Neves M, Sousa N, Lima A, Santos LL. Emerging antibody-based therapeutic strategies for bladder cancer: A systematic review. J Control Release 2015. [PMID: 26196222 DOI: 10.1016/j.jconrel.2015.07.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Bladder cancer is the most common malignancy of the urinary tract, presents the highest recurrence rate among solid tumors and is the second leading cause of death in genitourinary cancers. Despite recent advances in understanding of pathophysiology of the disease, the management of bladder cancer patients remains a clinically challenging problem. Particularly, bladder tumors invading the muscularis propria and disseminated disease are often not responsive to currently available therapeutic approaches, which include surgery and conventional chemotherapy. Antibody-based therapeutic strategies have become an established treatment option for over a decade in several types of cancer. However, bladder cancer has remained mostly an "orphan disease" regarding the introduction of these novel therapeutics, which has been translated in few improvements in patients overall survival. In order to shift this paradigm, several clinical studies involving antibody-based therapeutic strategies targeting the most prominent bladder cancer-related biomolecular pathways and immunological mediators are ongoing. This systematic review explores antibody-based therapeutics for bladder cancer undergoing clinical trial and discusses the future perspectives in this field, envisaging the development of more effective guided therapeutics.
Collapse
Affiliation(s)
- Rita Azevedo
- Experimental Pathology and Therapeutics Group - Research Center, Portuguese Institute of Oncology of Porto (IPO-Porto), Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal; Abel Salazar Institute of Biomedical Sciences (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - José Alexandre Ferreira
- Experimental Pathology and Therapeutics Group - Research Center, Portuguese Institute of Oncology of Porto (IPO-Porto), Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal; Mass Spectrometry Center, QOPNA, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| | - Andreia Peixoto
- Experimental Pathology and Therapeutics Group - Research Center, Portuguese Institute of Oncology of Porto (IPO-Porto), Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal
| | - Manuel Neves
- Experimental Pathology and Therapeutics Group - Research Center, Portuguese Institute of Oncology of Porto (IPO-Porto), Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal; Abel Salazar Institute of Biomedical Sciences (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Nuno Sousa
- Department of Medical Oncology, Portuguese Institute of Oncology of Porto (IPO-Porto), Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal
| | - Aurea Lima
- CESPU, Institute of Research and Advanced Training in Health Sciences and Technologies (Iinfacts), Department of Pharmaceutical Sciences, Rua Central de Gandra 1317, 4585-116, Gandra-PRD, Portugal; Molecular Oncology and Viral Pathology Group - Research Center, Portuguese Institute of Oncology of Porto (IPO-Porto), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Lucio Lara Santos
- Experimental Pathology and Therapeutics Group - Research Center, Portuguese Institute of Oncology of Porto (IPO-Porto), Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal; Health School of University of Fernando Pessoa, Praça 9 de Abril 349, 4249-004 Porto, Portugal; Department of Surgical Oncology, Portuguese Institute of Oncology of Porto (IPO-Porto), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal.
| |
Collapse
|