1
|
Moussa MJ, Kovalenko I, Crupi E, Proskuriakova E, Geng Y, Fallara G, Benkhadra R, Raggi D, Campbell MT, Msaouel P, Alhalabi O. Antiangiogenic therapy combined with immune checkpoint blockade in urothelial cancer: Systematic review and meta-analysis. Bladder Cancer 2024; 10:300-312. [PMID: 40035076 PMCID: PMC11864237 DOI: 10.1177/23523735241296763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 10/09/2024] [Indexed: 03/05/2025]
Abstract
Background Antiangiogenic therapy had been tested in urothelial cancer (UC) without reaching the clinic. Objective We provide a systematic review and meta-analysis of trials to assess efficacy of immune checkpoint inhibitors (ICI) combined with antiangiogenic agents in UC. Methods Following PRISMA guidelines, we searched for trials with at least one arm of patients with UC treated with ICI plus antiangiogenics. Data were analyzed with the "meta" package from R using a one-staged frequentist meta-analysis. Results After screening 13,708 titles and abstracts, 9 studies were selected for analysis with 14 identified cohorts comprising 621 patients: 448 were ICI-naïve (ICI-N) and 173 were ICI-exposed (ICI-E). The estimated objective response rate (ORR) in all patients was 27% (21-35). In the ICI-N group, ORR was 34% (28-41). Conversely, the ICI-E group had a lower ORR of 16% (9-28). This difference was mainly driven by a higher partial response rate of 27% (23-31) in ICI-N group compared to 13% (8-20) in the ICI-E group. Disease control rate was 72% (66-77) ICI-N group vs. 71% (64-78) in ICI-E group. Median overall survival ranged from 6.4 to 24.9 months in the ICI-N group, and 8.2 to 10.4 months in ICI-E group. Median progression free survival ranged from 1.9 to 10.1 months and from 3 to 3.9 months in both groups, respectively. Conclusion ORR with ICI plus antiangiogenics was lower after prior ICI exposure, with substantial variability estimates among included trials, either due to differences among antiangiogenic agents used or trial-related factors. Future exploration of ICI combined with antiangiogenics in UC, especially in ICI-refractory setting, will benefit from better patient selection.
Collapse
Affiliation(s)
- Mohammad Jad Moussa
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Iuliia Kovalenko
- Department of Internal Medicine, Eastern Virginia Medical School, Old Dominion University, Norfolk, VA, USA
| | - Emanuele Crupi
- Department of Medical Oncology, IRCCS San Raffaele Hospital, Milan, Italy
- Department of Medical Oncology, Vita-Salute San Raffaele University, Milan, Italy
| | | | - Yimin Geng
- Research Medical Library, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Giuseppe Fallara
- Department of Urology and Division of Experimental Oncology, Urological Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
| | - Raed Benkhadra
- Department of Hematology and Oncology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Daniele Raggi
- Department of Medical Oncology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Matthew T. Campbell
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pavlos Msaouel
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Omar Alhalabi
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
2
|
Zhang C, Hu J, Liu Z, Deng H, Xiao J, Yi Z, He Y, Xiao Z, Huang J, Liang H, Fan B, Wang Z, Chen J, Zu X. Hsa_circ_0000520 suppresses vasculogenic mimicry formation and metastasis in bladder cancer through Lin28a/PTEN/PI3K signaling. Cell Mol Biol Lett 2024; 29:118. [PMID: 39237880 PMCID: PMC11378395 DOI: 10.1186/s11658-024-00627-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 08/01/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Vasculogenic mimicry (VM) is a potential cause of resistance to antiangiogenic therapy and is closely related to the malignant progression of tumors. It has been shown that noncoding RNAs play an important role in the formation of VM in malignant tumors. However, the role of circRNAs in VM of bladder cancer and the regulatory mechanisms are unclear. METHODS Firstly, hsa_circ_0000520 was identified to have circular character by Sanger sequencing and Rnase R assays. Secondly, the potential clinical value of hsa_circ_0000520 was explored by quantitative real-time polymerase chain reaction (qRT-PCR) and fluorescence in situ hybridization (FISH) of clinical specimens. Thirdly, the role of hsa_circ_0000520 in bladder cancer invasion, migration, and VM formation was examined by in vivo and in vitro experiments. Finally, the regulatory mechanisms of hsa_circ_0000520 in the malignant progression of bladder cancer were elucidated by RNA binding protein immunoprecipitation (RIP), RNA pulldown, co-immunoprecipitation (co-IP), qRT-PCR, Western blot (WB), and fluorescence co-localization. RESULTS Hsa_circ_0000520 was characterized as a circular RNA and was lowly expressed in bladder cancer compared with the paracancer. Bladder cancer patients with high expression of hsa_circ_0000520 had better survival prognosis. Functionally, hsa_circ_0000520 inhibited bladder cancer invasion, migration, and VM formation. Mechanistically, hsa_circ_0000520 acted as a scaffold to promote binding of UBE2V1/UBC13 to Lin28a, further promoting the ubiquitous degradation of Lin28a, improving PTEN mRNA stability, and inhibiting the phosphorylation of the PI3K/AKT pathway. The formation of hsa_circ_0000520 in bladder cancer was regulated by RNA binding protein QKI. CONCLUSIONS Hsa_circ_0000520 inhibits metastasis and VM formation in bladder cancer and is a potential target for bladder cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Chunyu Zhang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiao Hu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhi Liu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Hao Deng
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jiatong Xiao
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenglin Yi
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yunbo He
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zicheng Xiao
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jinliang Huang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Haisu Liang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Benyi Fan
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhihua Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jinbo Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Xiongbing Zu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
- Department of Urology, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, China.
| |
Collapse
|
3
|
Lin S, Chai Y, Zheng X, Xu X. The role of HIF in angiogenesis, lymphangiogenesis, and tumor microenvironment in urological cancers. Mol Biol Rep 2023; 51:14. [PMID: 38085375 PMCID: PMC10716070 DOI: 10.1007/s11033-023-08931-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 10/25/2023] [Indexed: 12/18/2023]
Abstract
Typically associated with solid tumors, hypoxia contributes to tumor angiogenesis and lymphangiogenesis through various molecular mechanisms. Accumulating studies indicate that hypoxia-inducible factor is the key transcription factor coordinating endothelial cells to respond to hypoxia in urological cancers, mainly renal cell carcinoma, prostate cancer, and bladder cancer. Moreover, it has been suggested that tumor hypoxia in tumor microenvironment simultaneously recruits stromal cells to suppress immune activities. This review summarizes the mechanisms by which HIF regulates tumorigenesis and elaborates on the associations between HIF and angiogenesis, lymphangiogenesis, and tumor microenvironment in urological cancers.
Collapse
Affiliation(s)
- Shen Lin
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yueyang Chai
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiangyi Zheng
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Xin Xu
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
4
|
Politis D, Konstantakou P, Bramis K, Alexandraki KI, Spyroglou A, Mastorakos G, Anastasiou I, Papaconstantinou I, Dimopoulos MA. Surgical Treatment of Solitary Metachronous Adrenal Metastasis from Urothelial Carcinoma of the Urinary Bladder. TOUCHREVIEWS IN ENDOCRINOLOGY 2023; 19:94-97. [PMID: 37313235 PMCID: PMC10258614 DOI: 10.17925/ee.2023.19.1.94] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 12/19/2022] [Indexed: 06/15/2023]
Abstract
Urothelial cancer is a common neoplasm and metastatic disease correlates with a poor prognosis. Isolated adrenal gland metastases of urothelial carcinoma are quite rare, and management options can decide a patient's prognosis. Herein we report the case of a 76-year-old man with a metachronous solitary adrenal metastasis from a bladder carcinoma, who underwent adrenalectomy as part of his treatment. Furthermore, we discuss the cases of solitary adrenal metastases of urothelial carcinoma available in the literature, to identify key features to direct appropriate treatment of this rare metastatic site of urothelial cancer and improve prognosis and survival. Still, further prospective studies are needed to design effective therapeutic strategies.
Collapse
Affiliation(s)
- Dimitrios Politis
- Second Department of Surgery, Aretaieion University Hospital, Medical School, National and Kapodistrian University of Athens, Greece
| | - Panagiota Konstantakou
- Endocrinology Department, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, Greece
| | - Konstantinos Bramis
- Second Department of Surgery, Aretaieion University Hospital, Medical School, National and Kapodistrian University of Athens, Greece
| | - Krystallenia I Alexandraki
- Endocrinology Department, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, Greece
| | - Ariadni Spyroglou
- Endocrinology Department, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, Greece
| | - George Mastorakos
- Endocrinology Department, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, Greece
| | - Ioannis Anastasiou
- First Department of Urology, National and Kapodistrian University of Athens, Laikon University Hospital, Athens, Greece
| | - Ioannis Papaconstantinou
- Second Department of Surgery, Aretaieion University Hospital, Medical School, National and Kapodistrian University of Athens, Greece
| | - Meletios A Dimopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Alexandra Hospital, Athens, Greece
| |
Collapse
|
5
|
Shen C, Li Z, Zhang Y, Zhang Z, Wu Z, Da L, Yang S, Wang Z, Zhang Y, Qie Y, Zhao G, Lin Y, Huang S, Zhou M, Hu H. Identification of a dysregulated CircRNA-associated gene signature for predicting prognosis, immune landscape, and drug candidates in bladder cancer. Front Oncol 2022; 12:1018285. [PMID: 36300085 PMCID: PMC9589509 DOI: 10.3389/fonc.2022.1018285] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Increasing evidences have demonstrated that circular RNA (circRNAs) plays a an essential regulatory role in initiation, progression and immunotherapy resistance of various cancers. However, circRNAs have rarely been studied in bladder cancer (BCa). The purpose of this research is to explore new circRNAs and their potential mechanisms in BCa. A novel ceRNA-regulated network, including 87 differentially expressed circRNAs (DE-circRNAs), 126 DE-miRNAs, and 217 DE-mRNAs was constructed to better understanding the biological processes using Cytoscape 3.7.1 based on our previously high-throughput circRNA sequencing and five GEO datasets. Subsequently, five randomly selected circRNAs (upregulated circ_0001681; downregulated circ_0000643, circ_0001798, circ_0006117 and circ_0067900) in 20 pairs of BCa and paracancerous tissues were confirmed using qRT-PCR. Functional analysis results determined that 772 GO functions and 32 KEGG pathways were enriched in the ceRNA network. Ten genes (PFKFB4, EDNRA, GSN, GAS1, PAPPA, DTL, TGFBI, PRSS8, RGS1 and TCF4) were selected for signature construction among the ceRNA network. The Human Protein Atlas (HPA) expression of these genes were consistent with the above sequencing data. Notably, the model was validated in multiple external datasets (GSE13507, GSE31684, GSE48075, IMvigor210 and GSE32894). The immune-infiltration was evaluated by 7 published algorithms (i.e., TIMER, CIBERSORT, CIBERSORT-ABS, QUANTISEQ, MCPCOUNTER, XCELL and EPIC). Next, Correlations between riskscore or risk groups and clinicopathological data, overall survival, recognized immunoregulatory cells or common chemotherapeutic agents of BCa patients were performed using wilcox rank test, chi-square test, cox regression and spearman’s correlation analysis; and, these results are significant. According to R package “GSVA” and “clusterProfiler”, the most significantly enriched HALLMARK and KEGG pathway was separately the ‘Epithelial Mesenchymal Transition’ and ‘Ecm Receptor Interaction’ in the high- vs. low-risk group. Additionally, the functional experiments in vitro also revealed that the overexpression of has_circ_0067900 significantly impaired the proliferation, migration, and invasion capacities of BCa cells. Collectively, the results of the current study provide a novel landscape of circRNA-associated ceRNA-regulated network in BCa. The ceRNA-associated gene model which was constructed presented a high predictive performance for the prognosis, immunotherapeutic responsiveness, and chemotherapeutic sensitivity of BCa. And, has_circ_0067900 was originally proposed as tumor suppressor for patients with BCa.
Collapse
Affiliation(s)
- Chong Shen
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhi Li
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yinglang Zhang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhe Zhang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhouliang Wu
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - La Da
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Shaobo Yang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zejin Wang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yu Zhang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yunkai Qie
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Gangjian Zhao
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yuda Lin
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Shiwang Huang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Mingli Zhou
- Department of Neuromuscular Diseases, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hailong Hu
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
- *Correspondence: Hailong Hu,
| |
Collapse
|
6
|
Hu X, Li G, Wu S. Advances in Diagnosis and Therapy for Bladder Cancer. Cancers (Basel) 2022; 14:3181. [PMID: 35804953 PMCID: PMC9265007 DOI: 10.3390/cancers14133181] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/19/2022] [Accepted: 06/24/2022] [Indexed: 02/04/2023] Open
Abstract
Bladder cancer (BCa) is one of the most common and expensive urinary system malignancies for its high recurrence and progression rate. In recent years, immense amounts of studies have been carried out to bring a more comprehensive cognition and numerous promising clinic approaches for BCa therapy. The development of innovative enhanced cystoscopy techniques (optical techniques, imaging systems) and tumor biomarkers-based non-invasive urine screening (DNA methylation-based urine test) would dramatically improve the accuracy of tumor detection, reducing the risk of recurrence and progression of BCa. Moreover, intravesical instillation and systemic therapeutic strategies (cocktail therapy, immunotherapy, vaccine therapy, targeted therapy) also provide plentiful measures to break the predicament of BCa. Several exploratory clinical studies, including novel surgical approaches, pharmaceutical compositions, and bladder preservation techniques, emerged continually, which are supposed to be promising candidates for BCa clinical treatment. Here, recent advances and prospects of diagnosis, intravesical or systemic treatment, and novel drug delivery systems for BCa therapy are reviewed in this paper.
Collapse
Affiliation(s)
- Xinzi Hu
- Institute of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, Shenzhen 518000, China; (X.H.); (G.L.)
- Department of Urology, South China Hospital, Health Science Center, Shenzhen University, Shenzhen 518116, China
| | - Guangzhi Li
- Institute of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, Shenzhen 518000, China; (X.H.); (G.L.)
- Department of Urology, South China Hospital, Health Science Center, Shenzhen University, Shenzhen 518116, China
| | - Song Wu
- Institute of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, Shenzhen 518000, China; (X.H.); (G.L.)
- Department of Urology, South China Hospital, Health Science Center, Shenzhen University, Shenzhen 518116, China
| |
Collapse
|
7
|
Cai C, Wang X, Fu Q, Chen A. The VEGF expression associated with prognosis in patients with intrahepatic cholangiocarcinoma: a systematic review and meta-analysis. World J Surg Oncol 2022; 20:40. [PMID: 35189920 PMCID: PMC8859901 DOI: 10.1186/s12957-022-02511-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 02/07/2022] [Indexed: 01/06/2023] Open
Abstract
Abstract
Objective
To systematically evaluate the relationship between vascular endothelial growth factor (VEGF) and prognosis of intrahepatic cholangiocarcinoma by meta-analysis.
Methods
We systematically searched relevant studies in the databases of PubMed, Embase, Cochrane Library, CNKI, Wangfang, and Web of Science, with search dates limited to September 1, 2021. We extracted relevant data, including prognosis and clinicopathological features of patients with different expressions of VEGF in intrahepatic cholangiocarcinoma. The combined hazard ratio (HR), odds ratio (OR), and 95% confidence interval (CI) were calculated to evaluate the link strength between VEGF and prognosis of cholangiocarcinoma patients.
Results
A total of 7 eligible studies with 495 patients were included in this meta-analysis. The results showed that the high expression of VEGF was significantly related to poor overall survival (OS) (HR = 1.93, 95% CI 1.52–2.46, P < 0.05) in patients with intrahepatic cholangiocarcinoma. Moreover, high expression of VEGF in tumor tissues associated with lymph node metastasis (LNM) (OR = 6.79, 95% CI 3.93–11.73, P < 0.05) and advanced TNM stage (OR = 4.35, 95% CI 2.34–8.07, P < 0.05) in intrahepatic cholangiocarcinoma. Sensitivity analysis shows that the meta-analysis results are stable and reliable.
Conclusion
The expression of VEGF is related to the OS of patients with intrahepatic cholangiocarcinoma, and the OS of patients with high expression of VEGF is shorter. VEGF may be a novel predictor of intrahepatic cholangiocarcinoma patients.
Trial registration
PROSPERO (CRD42022297443).
Collapse
|
8
|
Roviello G, Catalano M, Santi R, Palmieri VE, Vannini G, Galli IC, Buttitta E, Villari D, Rossi V, Nesi G. Immune Checkpoint Inhibitors in Urothelial Bladder Cancer: State of the Art and Future Perspectives. Cancers (Basel) 2021; 13:4411. [PMID: 34503220 PMCID: PMC8431680 DOI: 10.3390/cancers13174411] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/26/2021] [Accepted: 08/28/2021] [Indexed: 12/11/2022] Open
Abstract
Bladder cancer (BC) is the most common malignancy of the genitourinary tract, with high morbidity and mortality rates. Until recently, the treatment of locally advanced or metastatic urothelial BC was based on the use of chemotherapy alone. Since 2016, five immune checkpoint inhibitors (ICIs) have been approved by the Food and Drug Administration (FDA) in different settings, i.e., first-line, maintenance and second-line treatment, while several trials are still ongoing in the perioperative context. Lately, pembrolizumab, a programmed death-1 (PD-1) inhibitor, has been approved for Bacillus Calmette-Guérin (BCG)-unresponsive high-risk non-muscle invasive bladder cancer (NMIBC), using immunotherapy at an early stage of the disease. This review investigates the current state and future perspectives of immunotherapy in BC, focusing on the rationale and results of combining immunotherapy with other therapeutic strategies.
Collapse
Affiliation(s)
- Giandomenico Roviello
- Department of Health Sciences, University of Florence, 50139 Florence, Italy; (G.R.); (M.C.); (V.E.P.); (G.V.); (E.B.)
| | - Martina Catalano
- Department of Health Sciences, University of Florence, 50139 Florence, Italy; (G.R.); (M.C.); (V.E.P.); (G.V.); (E.B.)
| | - Raffaella Santi
- Histopathology and Molecular Diagnostics, Careggi Teaching Hospital, 50139 Florence, Italy; (R.S.); (I.C.G.)
| | - Valeria Emma Palmieri
- Department of Health Sciences, University of Florence, 50139 Florence, Italy; (G.R.); (M.C.); (V.E.P.); (G.V.); (E.B.)
| | - Gianmarco Vannini
- Department of Health Sciences, University of Florence, 50139 Florence, Italy; (G.R.); (M.C.); (V.E.P.); (G.V.); (E.B.)
| | - Ilaria Camilla Galli
- Histopathology and Molecular Diagnostics, Careggi Teaching Hospital, 50139 Florence, Italy; (R.S.); (I.C.G.)
| | - Eleonora Buttitta
- Department of Health Sciences, University of Florence, 50139 Florence, Italy; (G.R.); (M.C.); (V.E.P.); (G.V.); (E.B.)
| | - Donata Villari
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy;
| | - Virginia Rossi
- Clinical Oncology Unit, Careggi Teaching Hospital, 50139 Florence, Italy;
| | - Gabriella Nesi
- Department of Health Sciences, University of Florence, 50139 Florence, Italy; (G.R.); (M.C.); (V.E.P.); (G.V.); (E.B.)
| |
Collapse
|
9
|
Khalife N, Chahine C, Kordahi M, Felefly T, Kourie HR, Saleh K. Urothelial carcinoma in the era of immune checkpoint inhibitors. Immunotherapy 2021; 13:953-964. [PMID: 34184561 DOI: 10.2217/imt-2021-0042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Bladder cancer is the seventh most frequent cancer worldwide. The majority of patients present with nonmuscle invasive disease, while 20% of the patients are diagnosed with muscle-invasive bladder cancer. The treatment of nonmuscle invasive disease is endoscopic resection followed by intravesical adjuvant treatment for high risk patients. The standard treatment of localized muscle-invasive disease is neoadjuvant chemotherapy followed by radical cystectomy. Platinum-based chemotherapy is the first-line treatment in locally advanced or metastatic urothelial carcinoma. Immune checkpoint inhibitors have been approved for the treatment of metastatic urothelial carcinoma as second-line treatment or first-line in platinum-ineligible patients. Recently, pembrolizumab have been approved in bacillus Calmette-Guérin (BCG)-refractory nonmuscle invasive bladder cancer. This review summarizes the current evidence concerning immunotherapy in the treatment of urothelial carcinoma.
Collapse
Affiliation(s)
- Nadine Khalife
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, 94800, France
| | - Claude Chahine
- International Department, Gustave Roussy Cancer Campus, Villejuif, 94800, France
| | - Manal Kordahi
- National Institute of Pathology, Baabda, 1003, Lebanon
| | - Tony Felefly
- Department of Radiation Oncology, Hotel-Dieu de France University Hospital, School of Medicine, Saint-Joseph University, Beirut, 1100, Lebanon
| | - Hampig Raphael Kourie
- Hematology-Oncology Department, Faculty of Medicine, Saint Joseph University of Beirut, Beirut, 1100, Lebanon
| | - Khalil Saleh
- Department of Hematology, Gustave Roussy Cancer Campus, Villejuif, 94800, France
| |
Collapse
|
10
|
Ashrafizadeh M, Yaribeygi H, Sahebkar A. Therapeutic Effects of Curcumin against Bladder Cancer: A Review of Possible Molecular Pathways. Anticancer Agents Med Chem 2021; 20:667-677. [PMID: 32013836 DOI: 10.2174/1871520620666200203143803] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 01/03/2020] [Accepted: 01/09/2020] [Indexed: 12/22/2022]
Abstract
There are concerns about the increased incidence of cancer both in developing and developed countries. In spite of recent progress in cancer therapy, this disease is still one of the leading causes of death worldwide. Consequently, there have been rigorous attempts to improve cancer therapy by looking at nature as a rich source of naturally occurring anti-tumor drugs. Curcumin is a well-known plant-derived polyphenol found in turmeric. This compound has numerous pharmacological effects such as antioxidant, anti-inflammatory, antidiabetic and anti-tumor properties. Curcumin is capable of suppressing the growth of a variety of cancer cells including those of bladder cancer. Given the involvement of various signaling pathways such as PI3K, Akt, mTOR and VEGF in the progression and malignancy of bladder cancer, and considering the potential of curcumin in targeting signaling pathways, it seems that curcumin can be considered as a promising candidate in bladder cancer therapy. In the present review, we describe the molecular signaling pathways through which curcumin inhibits invasion and metastasis of bladder cancer cells.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Habib Yaribeygi
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran.,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
11
|
Dong Y, Hao L, Fang K, Han XX, Yu H, Zhang JJ, Cai LJ, Fan T, Zhang WD, Pang K, Ma WM, Wang XT, Han CH. A network pharmacology perspective for deciphering potential mechanisms of action of Solanum nigrum L. in bladder cancer. BMC Complement Med Ther 2021; 21:45. [PMID: 33494738 PMCID: PMC7836472 DOI: 10.1186/s12906-021-03215-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 01/11/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Solanum nigrum L. decoction has been used as a folklore medicine in China to prevent the postoperative recurrence of bladder cancer (BC). However, there are no previous pharmacological studies on the protective mechanisms of this activity of the plant. Thus, this study aimed to perform a systematic analysis and to predict the potential action mechanisms underlying S. nigrum activity in BC based on network pharmacology. METHODS Based on network pharmacology, the active ingredients of S. nigrum and the corresponding targets were identified using the Traditional Chinese Medicines for Systems Pharmacology Database and Analysis Platform database, and BC-related genes were screened using GeneCards and the Online Mendelian Inheritance in Man database. In addition, ingredient-target (I-T) and protein-protein interaction (PPI) networks were constructed using STRING and Cytoscape, Gene Ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were conducted, and then the pathways directly related to BC were integrated manually to reveal the pharmacological mechanism underlying S. nigrum-medicated therapeutic effects in BC. RESULTS Seven active herbal ingredients from 39 components of S. nigrum were identified, which shared 77 common target genes related to BC. I-T network analysis revealed that quercetin was associated with all targets and that NCOA2 was targeted by four ingredients. Besides, interleukin 6 had the highest degree value in the PPI network, indicating a hub role. A subsequent gene enrichment analysis yielded 86 significant GO terms and 89 significant pathways, implying that S. nigrum had therapeutic benefits in BC through multi-pathway effects, including the HIF-1, TNF, P53, MAPK, PI3K/Akt, apoptosis and bladder cancer pathway. CONCLUSIONS S. nigrum may mediate pharmacological effects in BC through multi-target and various signaling pathways. Further validation is required experimentally. Network pharmacology approach provides a predicative novel strategy to reveal the holistic mechanism of action of herbs.
Collapse
Affiliation(s)
- Yang Dong
- Department of Urology, XuZhou Central Hospital Affiliated to Nanjing University of Chinese Medicine, Jiefang South Road, No. 199, Jiangsu, Xuzhou, China.,Department of Clinical Medicine, Xuzhou Medical University, Xuzhou, China
| | - Lin Hao
- Department of Urology, XuZhou Central Hospital Affiliated to Nanjing University of Chinese Medicine, Jiefang South Road, No. 199, Jiangsu, Xuzhou, China.,Department of Clinical Medicine, Xuzhou Medical University, Xuzhou, China
| | - Kun Fang
- Xuzhou Clinical Medical College of Integrated Traditional Chinese and Western Medicine Affiliated to Nanjing University of Traditional Chinese Medicine, Xuzhou, China
| | - Xiao-Xiao Han
- Center of Reproductive Medicine, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hui Yu
- Yantai Hospital of Traditional Chinese Medicine, Yantai, China
| | - Jian-Jun Zhang
- Department of Urology, Suqian People's Hospital of Nanjing Drum-Tower Hospital Group, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, China
| | - Long-Jun Cai
- Department of Urology, Suqian People's Hospital of Nanjing Drum-Tower Hospital Group, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, China
| | - Tao Fan
- Department of Urology, XuZhou Central Hospital Affiliated to Nanjing University of Chinese Medicine, Jiefang South Road, No. 199, Jiangsu, Xuzhou, China
| | - Wen-da Zhang
- Department of Urology, XuZhou Central Hospital Affiliated to Nanjing University of Chinese Medicine, Jiefang South Road, No. 199, Jiangsu, Xuzhou, China
| | - Kun Pang
- Department of Urology, XuZhou Central Hospital Affiliated to Nanjing University of Chinese Medicine, Jiefang South Road, No. 199, Jiangsu, Xuzhou, China
| | - Wei-Ming Ma
- Department of Urology, XuZhou Central Hospital Affiliated to Nanjing University of Chinese Medicine, Jiefang South Road, No. 199, Jiangsu, Xuzhou, China
| | - Xi-Tao Wang
- Department of Urology, XuZhou Central Hospital Affiliated to Nanjing University of Chinese Medicine, Jiefang South Road, No. 199, Jiangsu, Xuzhou, China
| | - Cong-Hui Han
- Department of Urology, XuZhou Central Hospital Affiliated to Nanjing University of Chinese Medicine, Jiefang South Road, No. 199, Jiangsu, Xuzhou, China. .,Department of Clinical Medicine, Xuzhou Medical University, Xuzhou, China. .,Department of Biotechnology, College of Life Sciences, Jiangsu Normal University, Xuzhou, China.
| |
Collapse
|
12
|
Necchi A, Nishiyama H, Matsubara N, Lee JL, Petrylak DP, de Wit R, Drakaki A, Liepa AM, Mao H, Bell-McGuinn K, Powles T. Health-related quality of life in the randomized phase 3 study of ramucirumab plus docetaxel versus placebo plus docetaxel in platinum-refractory advanced urothelial carcinoma (RANGE). BMC Urol 2020; 20:181. [PMID: 33160359 PMCID: PMC7648381 DOI: 10.1186/s12894-020-00752-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/26/2020] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND To evaluate patient-reported outcomes with ramucirumab plus docetaxel, a regimen which improved progression-free survival in platinum-refractory advanced urothelial carcinoma (aUC). METHODS RANGE-a randomized, double-blinded, phase 3 trial in patients with platinum-refractory aUC. Ramucirumab (10 mg/kg) plus docetaxel (75 mg/m2) or placebo plus docetaxel were administered every 21 days until disease progression or unacceptable toxicity. Patients received maximum 10 cycles of docetaxel. European Organization for Research and Treatment of Cancer Quality of Life Questionnaire C30 (EORTC QLQ-C30) and EuroQoL five-dimensions (EQ-5D-5L) were administered at baseline, start of each cycle, and 30-day follow-up visit. A ≥ 10-point change in QLQ-C30 scores was considered meaningful. Rates of improved/stable scores were compared between treatment arms using Fisher's exact test. Time to deterioration (TtD) was estimated and compared using Kaplan-Meier estimation and log-rank test. RESULTS Of the 530 patients, ~ 97% patients in each arm provided baseline QLQ-C30 data. On-treatment compliance was ≥ 88% for first 8 cycles. Mean baseline QLQ-C30 scores were similar between arms, with global quality of life (QoL), fatigue, pain, and insomnia having greatest impairment. Postbaseline rates of improved/stable QLQ-C30 scores were similar between treatment arms except for greater improvement in pain score with ramucirumab. TtD of QLQ-C30 scales favored ramucirumab arm. Baseline EQ-5D-5L index and visual analogue scale scores were similar between arms, followed by relatively stable on-treatment scores. EQ-5D-5L scores worsened at post-discontinuation follow-up visit. CONCLUSIONS Ramucirumab plus docetaxel did not negatively impact QoL compared with docetaxel alone in platinum-refractory aUC. Improved TtD and tumor associated rates of pain favored ramucirumab treatment. CLINICAL TRAIL REGISTRATION NCT02426125. https://clinicaltrials.gov/ct2/show/NCT02426125 . Date of registration: April 24th 2015.
Collapse
Affiliation(s)
- Andrea Necchi
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Dei Tumori, 20133, Milan, Italy.
| | | | | | - Jae-Lyun Lee
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | - Huzhang Mao
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | | |
Collapse
|
13
|
Apolo AB, Nadal R, Girardi DM, Niglio SA, Ley L, Cordes LM, Steinberg SM, Sierra Ortiz O, Cadena J, Diaz C, Mallek M, Davarpanah NN, Costello R, Trepel JB, Lee MJ, Merino MJ, Bagheri MH, Monk P, Figg WD, Gulley JL, Agarwal PK, Valera V, Chalfin HJ, Jones J, Streicher H, Wright JJ, Ning YM, Parnes HL, Dahut WL, Bottaro DP, Lara PN, Saraiya B, Pal SK, Stein MN, Mortazavi A. Phase I Study of Cabozantinib and Nivolumab Alone or With Ipilimumab for Advanced or Metastatic Urothelial Carcinoma and Other Genitourinary Tumors. J Clin Oncol 2020; 38:3672-3684. [PMID: 32915679 PMCID: PMC7605393 DOI: 10.1200/jco.20.01652] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/18/2020] [Indexed: 12/22/2022] Open
Abstract
PURPOSE We assessed the safety and efficacy of cabozantinib and nivolumab (CaboNivo) and CaboNivo plus ipilimumab (CaboNivoIpi) in patients with metastatic urothelial carcinoma (mUC) and other genitourinary (GU) malignances. PATIENTS AND METHODS Patients received escalating doses of CaboNivo or CaboNivoIpi. The primary objective was to establish a recommended phase II dose (RP2D). Secondary objectives included objective response rate (ORR), progression-free survival (PFS), duration of response (DoR), and overall survival (OS). RESULTS Fifty-four patients were enrolled at eight dose levels with a median follow-up time of 44.6 months; data cutoff was January 20, 2020. Grade 3 or 4 treatment-related adverse events (AEs) occurred in 75% and 87% of patients treated with CaboNivo and CaboNivoIpi, respectively, and included fatigue (17% and 10%, respectively), diarrhea (4% and 7%, respectively), and hypertension (21% and 10%, respectively); grade 3 or 4 immune-related AEs included hepatitis (0% and 13%, respectively) and colitis (0% and 7%, respectively). The RP2D was cabozantinib 40 mg/d plus nivolumab 3 mg/kg for CaboNivo and cabozantinib 40 mg/d, nivolumab 3 mg/kg, and ipilimumab 1 mg/kg for CaboNivoIpi. ORR was 30.6% (95% CI, 20.0% to 47.5%) for all patients and 38.5% (95% CI, 13.9% to 68.4%) for patients with mUC. Median DoR was 21.0 months (95% CI, 5.4 to 24.1 months) for all patients and not reached for patients with mUC. Median PFS was 5.1 months (95% CI, 3.5 to 6.9 months) for all patients and 12.8 months (95% CI, 1.8 to 24.1 months) for patients with mUC. Median OS was 12.6 months (95% CI, 6.9 to 18.8 months) for all patients and 25.4 months (95% CI, 5.7 to 41.6 months) for patients with mUC. CONCLUSION CaboNivo and CaboNivoIpi demonstrated manageable toxicities with durable responses and encouraging survival in patients with mUC and other GU tumors. Multiple phase II and III trials are ongoing for these combinations.
Collapse
Affiliation(s)
- Andrea B. Apolo
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Rosa Nadal
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Daniel M. Girardi
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Scot A. Niglio
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Lisa Ley
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Lisa M. Cordes
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Seth M. Steinberg
- Biostatistics and Data Management Section, Office of the Clinical Director, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Olena Sierra Ortiz
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Jacqueline Cadena
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Carlos Diaz
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Marissa Mallek
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Nicole N. Davarpanah
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Rene Costello
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Jane B. Trepel
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Min-Jung Lee
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Maria J. Merino
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Mohammad Hadi Bagheri
- Clinical Image Processing Service, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Paul Monk
- Division of Medical Oncology, Department of Internal Medicine, College of Medicine, The Ohio State University, and the Comprehensive Cancer Center, Columbus, OH
| | - William D. Figg
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - James L. Gulley
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Piyush K. Agarwal
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Vladimir Valera
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Heather J. Chalfin
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Jennifer Jones
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Howard Streicher
- Investigational Drug Branch, Cancer Therapy Evaluation Program, National Cancer Institute, National Institutes of Health, Rockville, MD
| | - John J. Wright
- Investigational Drug Branch, Cancer Therapy Evaluation Program, National Cancer Institute, National Institutes of Health, Rockville, MD
| | - Yangmin M. Ning
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Howard L. Parnes
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - William L. Dahut
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Donald P. Bottaro
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Primo N. Lara
- University of California Davis Comprehensive Cancer Center, Sacramento, CA
| | - Biren Saraiya
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| | | | - Mark N. Stein
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| | - Amir Mortazavi
- Division of Medical Oncology, Department of Internal Medicine, College of Medicine, The Ohio State University, and the Comprehensive Cancer Center, Columbus, OH
| |
Collapse
|
14
|
Abstract
Bladder cancer accounts for nearly 170,000 deaths worldwide annually. For over 4 decades, the systemic management of muscle-invasive and advanced bladder cancer has primarily consisted of platinum-based chemotherapy. Over the past 10 years, innovations in sequencing technologies have led to rapid genomic characterization of bladder cancer, deepening our understanding of bladder cancer pathogenesis and exposing potential therapeutic vulnerabilities. On the basis of its high mutational burden, immune checkpoint inhibitors were investigated in advanced bladder cancer, revealing durable responses in a subset of patients. These agents are now approved for several indications and highlight the changing treatment landscape of advanced bladder cancer. In addition, commonly expressed molecular targets were leveraged to develop targeted therapies, such as fibroblast growth factor receptor inhibitors and antibody-drug conjugates. The molecular characterization of bladder cancer and the development of novel therapies also have stimulated investigations into optimizing treatment approaches for muscle-invasive bladder cancer. Herein, the authors review the history of muscle-invasive and advanced bladder cancer management, highlight the important molecular characteristics of bladder cancer, describe the major advances in treatment, and offer future directions for therapeutic development.
Collapse
Affiliation(s)
- Vaibhav G Patel
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - William K Oh
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Matthew D Galsky
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
15
|
Jones R, Crabb S, Chester J, Elliott T, Huddart R, Birtle A, Evans L, Lester J, Jagdev S, Casbard A, Huang C, Madden TA, Griffiths G. A randomised Phase II trial of carboplatin and gemcitabine ± vandetanib in first-line treatment of patients with advanced urothelial cell cancer not suitable to receive cisplatin. BJU Int 2020; 126:292-299. [PMID: 32336008 DOI: 10.1111/bju.15096] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
OBJECTIVES To assess the efficacy and tolerability of the dual epidermal growth factor receptor/vascular endothelial growth factor receptor inhibitor, vandetanib, in combination with carboplatin and gemcitabine in the first-line treatment of patients with advanced transitional cell carcinoma urothelial cancer (UC) who were unsuitable for cisplatin. PATIENTS AND METHODS From 2011 to 2014, 82 patients were randomised from 16 hospitals across the UK into the TOUCAN double-blind, placebo-controlled randomised Phase II trial, receiving six 21-day cycles of intravenous carboplatin (target area under the concentration versus time curve 4.5, day 1) and gemcitabine (1000 mg/m2 days 1 and 8) combined with either oral vandetanib 100 mg or placebo (once daily). Progression-free survival (PFS; primary endpoint), adverse events, tolerability and feasibility of use, objective response rate and overall survival (OS) were evaluated. Intention-to-treat and per-protocol analyses were used to analyse the primary endpoint. RESULTS The 82 patients were randomised 1:1 to vandetanib (n = 40) or placebo (n = 42), and 25 patients (30%) completed six cycles of all allocated treatment. Toxicity Grade ≥3 was experienced in 80% (n = 32) and 76% (n = 32) of patients in the vandetanib and placebo arms, respectively. The median PFS was 6.8 and 8.8 months for the vandetanib and placebo arms, respectively (hazard ratio [HR] 1.07, 95% confidence interval [CI] 0.65-1.76; P = 0.71); the median OS was 10.8 vs 13.8 months (HR 1.41, 95% CI 0.79-2.52; P = 0.88); and radiological response rates were 50% and 55%. CONCLUSION There is no evidence that vandetanib improves clinical outcome in this setting. Our present data do not support its adoption as the regimen of choice for first-line treatment in patients with UC who were unfit for cisplatin.
Collapse
Affiliation(s)
- Robert Jones
- Beatson West of Scotland Cancer Centre, University of Glasgow, Glasgow, UK
| | - Simon Crabb
- Southampton Clinical Trials Unit, University of Southampton, Southampton, UK
| | - John Chester
- Cardiff University, Cardiff, UK
- Velindre Cancer Centre, Cardiff, UK
- St. James's University Hospital, Leeds, UK
| | - Tony Elliott
- Christie Hospital NHS Foundation Trust, Manchester, UK
| | | | | | | | | | | | - Angela Casbard
- Centre for Trials Research, Cardiff University, Cardiff, UK
| | - Chao Huang
- Centre for Trials Research, Cardiff University, Cardiff, UK
- Hull York Medical School, University of Hull, Hull, UK
| | | | - Gareth Griffiths
- Southampton Clinical Trials Unit, University of Southampton, Southampton, UK
- Centre for Trials Research, Cardiff University, Cardiff, UK
| |
Collapse
|
16
|
Mukae Y, Miyata Y, Nakamura Y, Araki K, Otsubo A, Yuno T, Mitsunari K, Matsuo T, Ohba K, Sakai H. Pathological roles of c-Met in bladder cancer: Association with cyclooxygenase-2, heme oxygenase-1, vascular endothelial growth factor-A and programmed death ligand 1. Oncol Lett 2020; 20:135-144. [PMID: 32565941 PMCID: PMC7285828 DOI: 10.3892/ol.2020.11540] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 02/18/2020] [Indexed: 02/02/2023] Open
Abstract
c-Met is a receptor tyrosine kinase that binds a specific ligand, namely hepatocyte growth factor (HGF). The HGF/c-Met system is important for malignant aggressiveness in various types of cancer, including bladder cancer (BC). However, although phosphorylation is the essential step required for biological activation of c-Met, pathological roles of phosphorylated c-Met at the clinical and molecular levels in patients with BC are not fully understood. In the present study, the expression levels of c-Met and the phosphorylation of two of its tyrosine residues (pY1234/pY1235 and pY1349) were immunohistochemically examined in 185 BC tissues. The associations between these expression levels and cancer cell invasion, metastasis, and cyclooxygenase-2 (COX-2), heme oxygenase-1 (HO-1), VEGF-A and programmed death ligand 1 (PD-L1) levels were investigated. c-Met was associated with muscle invasion (P=0.021), as well as the expression levels of HO-1 (P=0.028) and PD-L1 (P<0.001), whereas pY1349 c-Met was associated with muscle invasion (P=0.003), metastasis (P=0.025), and COX-2 (P=0.017), HO-1 (P=0.031) and PD-L1 (P=0.001) expression. By contrast, pY1234/1235 c-Met was associated with muscle invasion and metastasis (P=0.006 and P=0.012, respectively), but not with the panel of cancer-associated molecules. Furthermore, COX-2 and PD-L1 expression were associated with muscle invasion and metastasis, respectively (P=0.045 and P=0.036, respectively). Hence, c-Met serves important roles in muscle invasion by regulating HO-1 and PD-L1, whereas its phosphorylation at Y1349 is associated with muscle invasion and metastasis via the regulation of COX-2, HO-1 and PD-L1 in patients with BC. Furthermore, phosphorylation at Y1234/1235 may lead to muscle invasion and metastasis via alternate mechanisms associated with c-Met and pY1349 c-Met.
Collapse
Affiliation(s)
- Yuta Mukae
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Yasuyoshi Miyata
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Yuichiro Nakamura
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Kyohei Araki
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Asato Otsubo
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Tsutomu Yuno
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Kensuke Mitsunari
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Tomohiro Matsuo
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Kojiro Ohba
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Hideki Sakai
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| |
Collapse
|
17
|
Mollica V, Rizzo A, Montironi R, Cheng L, Giunchi F, Schiavina R, Santoni M, Fiorentino M, Lopez-Beltran A, Brunocilla E, Brandi G, Massari F. Current Strategies and Novel Therapeutic Approaches for Metastatic Urothelial Carcinoma. Cancers (Basel) 2020; 12:E1449. [PMID: 32498352 PMCID: PMC7352972 DOI: 10.3390/cancers12061449] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 05/29/2020] [Accepted: 06/01/2020] [Indexed: 02/07/2023] Open
Abstract
Urothelial carcinoma (UC) is a frequent cause of cancer-related deaths worldwide. Metastatic UC has been historically associated with poor prognosis, with a median overall survival of approximately 15 months and a 5-year survival rate of 18%. Although platinum-based chemotherapy remains the mainstay of medical treatment for patients with metastatic UC, chemotherapy clinical trials produced modest benefit with short-lived, disappointing responses. In recent years, the better understanding of the role of immune system in cancer control has led to the development and approval of several immunotherapeutic approaches in UC therapy, where immune checkpoint inhibitors have been revolutionizing the treatment of metastatic UC. Because of a better tumor molecular profiling, FGFR inhibitors, PARP inhibitors, anti-HER2 agents, and antibody drug conjugates targeting Nectin-4 are also emerging as new therapeutic options. Moreover, a wide number of trials is ongoing with the aim to evaluate several other alterations and pathways as new potential targets in metastatic UC. In this review, we will discuss the recent advances and highlight future directions of the medical treatment of UC, with a particular focus on recently published data and ongoing active and recruiting trials.
Collapse
Affiliation(s)
- Veronica Mollica
- Division of Oncology, S.Orsola-Malpighi Hospital, 40138 Bologna, Italy; (V.M.); (A.R.); (G.B.)
- Department of Experimental, Diagnostic and Specialty Medicine, S.Orsola-Malpighi Hospital, 40138 Bologna, Italy
| | - Alessandro Rizzo
- Division of Oncology, S.Orsola-Malpighi Hospital, 40138 Bologna, Italy; (V.M.); (A.R.); (G.B.)
- Department of Experimental, Diagnostic and Specialty Medicine, S.Orsola-Malpighi Hospital, 40138 Bologna, Italy
| | - Rodolfo Montironi
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, 60121 Ancona, Italy;
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Francesca Giunchi
- Pathology Service, Addarii Institute of Oncology, S-Orsola-Malpighi Hospital, 40138 Bologna, Italy;
| | - Riccardo Schiavina
- Department of Urology, University of Bologna, S-Orsola-Malpighi Hospital, 40138 Bologna, Italy; (R.S.); (E.B.)
| | - Matteo Santoni
- Oncology Unit, Macerata Hospital, 62100 Macerata, Italy;
| | | | - Antonio Lopez-Beltran
- Unit of Anatomical Pathology, Faculty of Medicine, Cordoba University, 14071 Cordoba, Spain;
| | - Eugenio Brunocilla
- Department of Urology, University of Bologna, S-Orsola-Malpighi Hospital, 40138 Bologna, Italy; (R.S.); (E.B.)
| | - Giovanni Brandi
- Division of Oncology, S.Orsola-Malpighi Hospital, 40138 Bologna, Italy; (V.M.); (A.R.); (G.B.)
- Department of Experimental, Diagnostic and Specialty Medicine, S.Orsola-Malpighi Hospital, 40138 Bologna, Italy
| | - Francesco Massari
- Division of Oncology, S.Orsola-Malpighi Hospital, 40138 Bologna, Italy; (V.M.); (A.R.); (G.B.)
| |
Collapse
|
18
|
Abstract
In 2018 bladder cancer (urothelial carcinoma) was ranked twelfth concerning worldwide diagnosis of malignancies. At the time point of diagnosis of bladder cancer, approximately 75% of patients present with a nonmuscle-invasive disease (NMIBC), while the remaining 25% show invasion of tumor cells in the muscle layer of the bladder wall (MIBC). Among NMIBC tumors, flat, high-grade carcinoma in situ (CIS) is a therapeutic challenge. CIS shows a tendency to invade the muscle tissue of the bladder wall and thus become a MIBC. Standard therapy of NMIBC (including CIS) is done via intravesical instillation of BCG (bacillus Calmette Guerin) inducing a local immune reaction that finally promotes elimination of bladder cancer cells. However, BCG treatment of NMIBC proves to be ineffective in approximately 40% of patients. Therefore, new therapeutic approaches for the treatment of bladder cancer are urgently needed. Among promising new treatment options that are currently being investigated are the use of immune checkpoint inhibitors, and targeted approaches attacking (among others) long noncoding RNAs, micro RNAs, cancer stem cells, PARP1, and receptor signaling pathways. Moreover, the use of antibody-drug-conjugates (ADCs) is investigated also in bladder cancer therapy. Another approach that has been successfully established in preclinical studies uses the cytotoxic power of the alpha-emitter Bi-213 coupled to an antibody targeting EGFR. Overexpression of EGFR has been demonstrated in the majority of patients suffering from CIS. Feasibility, safety, toxicity and therapeutic efficacy of intravesical instillation of Bi-213-anti-EGFR have been evaluated in a pilot study. Since the results of the pilot study proved to be promising, a further optimization of alpha-emitter immunotherapy in bladder cancer seems mandatory.
Collapse
Affiliation(s)
- Christof Seidl
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München, München, Germany.
| |
Collapse
|
19
|
Grivas P, Yu EY. Role of Targeted Therapies in Management of Metastatic Urothelial Cancer in the Era of Immunotherapy. Curr Treat Options Oncol 2019; 20:67. [DOI: 10.1007/s11864-019-0665-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
20
|
Zhao Y, An M, Zhang H, Tan D, Chen X, Wu P, Qin W, Zhang C, Shi C. Patient-derived bladder cancer xenograft models reveal VEGF and CDK4 enhancing tumor metastasis behavior. RSC Adv 2019; 9:17877-17884. [PMID: 35520551 PMCID: PMC9064661 DOI: 10.1039/c9ra02362c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 06/01/2019] [Indexed: 11/21/2022] Open
Abstract
New strategies to treat advanced bladder cancer are urgently required. A patient-derived xenograft (PDX) model has become a useful tool to evaluate chemotherapeutics and investigate personalized cancer treatment options. In this study, fresh human bladder cancer specimens (C09303 and C21391) were transplanted subcutaneously into nude mice to establish PDX models. These models well retained pathological characteristics and molecular markers of the original tumor. Primary cells derived from C09303 were cultured and perfused into the bladders of nude mice to establish orthotopic xenograft models. Evident signals of lung and kidney metastases were detected by whole-body optical imaging. Gemcitabine displayed a significant therapeutic effect on the subcutaneous or orthotopic xenograft tumors of C09303. In vitro, matrigel invasion assays showed that C09303 primary culture cells are remarkably invasive. To study the metastatic properties of C09303, we sequenced the genomes of C09303 and C21391, and found that the expression of VEGF and CDK4 was significantly upregulated, and VEGF-knockdown or CDK4-knockdown C09303 cells showed attenuated invasiveness and migration. This PDX model revealed that VEGF and CDK4 enhanced tumor metastasis behavior, suggesting them as novel molecular therapeutic targets. This framework provides a tool for research on metastasis mechanism and individualized treatment for bladder cancer.
Collapse
Affiliation(s)
- Yong Zhao
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University Xi'an Shaanxi 710032 China
| | - Mingjie An
- MBBS Zhongshan School of Medicine, Sun Yat-sen University Guangzhou 510080 China
| | - He Zhang
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University Xi'an Shaanxi 710032 China
| | - Dengxu Tan
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University Xi'an Shaanxi 710032 China
| | - Xue Chen
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University Xi'an Shaanxi 710032 China
| | - Pengpeng Wu
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University Xi'an Shaanxi 710032 China
| | - Weijun Qin
- Department of Urology, Xijing Hospital, The Fourth Military Medical University Xi'an Shaanxi 710032 China
| | - Caiqin Zhang
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University Xi'an Shaanxi 710032 China
| | - Changhong Shi
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University Xi'an Shaanxi 710032 China
| |
Collapse
|
21
|
|
22
|
Peixoto A, Relvas-Santos M, Azevedo R, Santos LL, Ferreira JA. Protein Glycosylation and Tumor Microenvironment Alterations Driving Cancer Hallmarks. Front Oncol 2019; 9:380. [PMID: 31157165 PMCID: PMC6530332 DOI: 10.3389/fonc.2019.00380] [Citation(s) in RCA: 219] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 04/23/2019] [Indexed: 12/12/2022] Open
Abstract
Decades of research have disclosed a plethora of alterations in protein glycosylation that decisively impact in all stages of disease and ultimately contribute to more aggressive cell phenotypes. The biosynthesis of cancer-associated glycans and its reflection in the glycoproteome is driven by microenvironmental cues and these events act synergistically toward disease evolution. Such intricate crosstalk provides the molecular foundations for the activation of relevant oncogenic pathways and leads to functional alterations driving invasion and disease dissemination. However, it also provides an important source of relevant glyco(neo)epitopes holding tremendous potential for clinical intervention. Therefore, we highlight the transversal nature of glycans throughout the currently accepted cancer hallmarks, with emphasis on the crosstalk between glycans and the tumor microenvironment stromal components. Focus is also set on the pressing need to include glycans and glycoconjugates in comprehensive panomics models envisaging molecular-based precision medicine capable of improving patient care. We foresee that this may provide the necessary rationale for more comprehensive studies and molecular-based intervention.
Collapse
Affiliation(s)
- Andreia Peixoto
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal.,Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal.,Tumour and Microenvironment Interactions Group, INEB-Institute for Biomedical Engineering, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Marta Relvas-Santos
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal
| | - Rita Azevedo
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal.,Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Lúcio Lara Santos
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal.,Department of Surgical Oncology, Portuguese Institute of Oncology, Porto, Portugal
| | - José Alexandre Ferreira
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal.,Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal.,Porto Comprehensive Cancer Center, Porto, Portugal
| |
Collapse
|
23
|
Nadal R, Bellmunt J. Management of metastatic bladder cancer. Cancer Treat Rev 2019; 76:10-21. [PMID: 31030123 DOI: 10.1016/j.ctrv.2019.04.002] [Citation(s) in RCA: 182] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 04/09/2019] [Accepted: 04/11/2019] [Indexed: 02/06/2023]
Abstract
Important advances in the understanding of the biology and mechanisms of tumor progression of urothelial carcinoma (UC) have been achieved over the past decade. The treatment landscape for advanced-stage, unresectable or metastatic UC has shifted dramatically over a short period of time, with 6 new therapeutic agents available for clinical use. The use of traditional chemotherapy and new immune checkpoints inhibitors (ICIs) directed at programmed cell-death protein 1 (PD-1) or its ligand has led to unprecedented survival benefits in selected patients with metastatic UC. Data show that anti-PD-1 ICIs are not only improving long-term clinical benefit, but also quality of life for patients in the second-line setting. In the front-line setting, regulatory agencies have restricted the indications of atezolizumab and pembrolizumab (both ICIs) to patients with PD-L1positivity with advanced UC and who are platinum-ineligible. Very recently, erdafitinib, a pan-FGFR inhibitor, has been granted accelerated approval by FDA for platinum-pretreated advanced metastatic UC with susceptible FGFR3 or FGFR2 genetic alterations. Enfortumab vedotin, an antibody-drug conjugate, have been granted breakthrough designation by the FDA for the treatment of metastatic UC. Here we review the clinical trial data that have established standard-of-care treatment for advanced-stage UC. In addition, mechanisms of resistance and biomarkers of response to platinum-based chemotherapies and immunotherapies are also discussed, along with the clinical benefits and limitations of these therapies.
Collapse
Affiliation(s)
- Rosa Nadal
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Joaquim Bellmunt
- IMIM-Hospital del Mar Research Institute, Barcelona, Spain; Harvard Medical School, Boston, MD, USA.
| |
Collapse
|
24
|
Schiff JP, Barata PC, Yu EY, Grivas P. Precision therapy in advanced urothelial cancer. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2019. [DOI: 10.1080/23808993.2019.1582298] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Joshua P. Schiff
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA, USA
| | - Pedro C. Barata
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA, USA
| | - Evan Y. Yu
- Department of Medicine, Division of Oncology, University of Washington and Fred Hutchinson Cancer Research Center, Seattle Cancer Care Alliance, Seattle, WA, USA
| | - Petros Grivas
- Department of Medicine, Division of Oncology, University of Washington and Fred Hutchinson Cancer Research Center, Seattle Cancer Care Alliance, Seattle, WA, USA
| |
Collapse
|
25
|
Ciccarese C, Iacovelli R, Bria E, Mosillo C, Bimbatti D, Fantinel E, Bisogno I, Brunelli M, Tortora G. Second-line therapy for metastatic urothelial carcinoma: Defining the best treatment option among immunotherapy, chemotherapy, and antiangiogenic targeted therapies. A systematic review and meta-analysis. Semin Oncol 2019; 46:65-72. [PMID: 30665685 DOI: 10.1053/j.seminoncol.2019.01.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 11/29/2018] [Accepted: 01/07/2019] [Indexed: 11/11/2022]
Abstract
There is no second-line standard of care universally accepted for platinum-refractory metastatic urothelial carcinoma. Immunotherapy and anti-VEGF(R) targeted therapies are 2 emerging strategies with promising though inconclusive results. We perform a systematic meta-analysis to assess the available options. We searched MEDLINE/PubMed, the Cochrane Library, and American society of clinical oncology (ASCO) Meeting abstracts to identify prospective studies. Data extraction was conduced according to the preferred reporting items for systematic reviews and meta-analyses (PRISMA) statement. The measured outcomes were overall survival (OS) and progression free survival (PFS). Seven randomized controlled trials were selected for final analysis, with a total of 2,451 evaluable patients. Chemotherapy with vinflunine did not reduce the risk of progression (HR = 1.11; 95%CI 0.78-1.57; P = .56) or death (HR = 0.97; 95%CI 0.70-1.34; P = .87) compared to taxanes. Immunotherapy with anti-PD-1/PD-L1 mAb improved OS over chemotherapy (HR = 0.81; 95% CI 0.71-0.92; P<.0009). The OS benefit of immunotherapy was retained when compared to taxanes, but not compared to vinflunine, although without a significant difference between the 2 subgroups (P = .30). A lack of PFS (HR = 0.73; P = .08) and OS (HR = 1.0; P = .99) benefit was observed with an anti-VEGF(R) plus chemotherapy compared to chemotherapy alone. No PFS (P = .14) or OS (P = .13) differences were detected when comparing anti-VEGF(R) ± chemotherapy and immunotherapy. Immunotherapy significantly improved OS compared to chemotherapy in metastatic urothelial carcinoma unselected for PD-L1 status. The addition of anti-VEGF(R) to chemotherapy did not provide any statistically significant benefit in terms of PFS or OS. Single agent taxanes or vinflunine can be considered given their similar efficacy but different toxicity profiles.
Collapse
Affiliation(s)
- Chiara Ciccarese
- UOC di Oncologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Università Cattolica del Sacro Cuore, Rome, Italy
| | - Roberto Iacovelli
- UOC di Oncologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.
| | - Emilio Bria
- UOC di Oncologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Università Cattolica del Sacro Cuore, Rome, Italy
| | - Claudia Mosillo
- Department of Medical Oncology, Azienda Universitaria Integrata di Verona (AOUI), Verona, Italy
| | - Davide Bimbatti
- Department of Medical Oncology, Azienda Universitaria Integrata di Verona (AOUI), Verona, Italy
| | - Emanuela Fantinel
- Department of Medical Oncology, Azienda Universitaria Integrata di Verona (AOUI), Verona, Italy
| | - Iolanda Bisogno
- Department of Medical Oncology, Azienda Universitaria Integrata di Verona (AOUI), Verona, Italy
| | - Matteo Brunelli
- Department of Diagnostics and Public Health, Section of Anatomical Pathology, University of Verona, Verona, Italy
| | - Giampaolo Tortora
- UOC di Oncologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
26
|
Poyet C, Thomas L, Benoit TM, Delmo DA, Luberto L, Banzola I, Günthart MS, Sais G, Eberli D, Sulser T, Provenzano M. Implication of vascular endothelial growth factor A and C in revealing diagnostic lymphangiogenic markers in node-positive bladder cancer. Oncotarget 2017; 8:21871-21883. [PMID: 28423532 PMCID: PMC5400630 DOI: 10.18632/oncotarget.15669] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 01/11/2017] [Indexed: 11/25/2022] Open
Abstract
Several lymphangiogenic factors, such as vascular endothelial growth factors (VEGFs), have been found to drive the development of lymphatic metastasis in bladder cancer (BCa). Here, we have analyzed the gene expression of lymphangiogenic factors in tissue specimens from 12 non-muscle invasive bladder cancers (NMIBC) and 11 muscle invasive bladder cancers (MIBC), considering tumor and tumor-adjacent normal bladder areas obtained from the same organs. We then compared the results observed in patients with those obtained after treating human primary bladder microvascular endothelial cells (MEC) with either direct stimulation with VEGF-A or VEGF-C or by co-culturing (trans-well assay) MEC with bladder cancer cell lines varying in VEGF-A and VEGF-C production based on tumor grade. The genes of three markers of lymphatic endothelial commitment and development (PDPN, LYVE-1 and SLP-76) were significantly overexpressed in tissues of MIBC patients showing positive lymphovascular invasion (LVI+), lymph node metastasis (Ln+) and tumor progression. Their expression was also significantly enhanced either after direct stimulation of MEC by VEGF-A and VEGF-C or in the trans-well assay with each bladder cancer cell line. SLP-76 showed the highest gene expression. Both VEGF-A and VEGF-C also enhanced the expression of SLP-76 protein in MEC. However, a correlation between increase of SLP-76 gene expression and the ability of MEC to migrate could only be seen after induction by VEGF-C. The significant expression of SLP-76 in LVI+/Ln+ progressive MIBC and its overexpression in MEC after VEGF-A and VEGF-C stimulation suggest the need to develop this regulator of developmental lymphangiogenesis as a diagnostic tool in BCa.
Collapse
Affiliation(s)
- Cédric Poyet
- Oncology Research Unit, Department of Urology and Division of Surgical Research, University Hospital of Zurich, Switzerland
| | - Linto Thomas
- Oncology Research Unit, Department of Urology and Division of Surgical Research, University Hospital of Zurich, Switzerland
| | - Tobias M Benoit
- Oncology Research Unit, Department of Urology and Division of Surgical Research, University Hospital of Zurich, Switzerland
| | - David Aquino Delmo
- Oncology Research Unit, Department of Urology and Division of Surgical Research, University Hospital of Zurich, Switzerland
| | - Laura Luberto
- Oncology Research Unit, Department of Urology and Division of Surgical Research, University Hospital of Zurich, Switzerland
| | - Irina Banzola
- Oncology Research Unit, Department of Urology and Division of Surgical Research, University Hospital of Zurich, Switzerland
| | - Michèle S Günthart
- Oncology Research Unit, Department of Urology and Division of Surgical Research, University Hospital of Zurich, Switzerland
| | - Giovanni Sais
- Oncology Research Unit, Department of Urology and Division of Surgical Research, University Hospital of Zurich, Switzerland
| | - Daniel Eberli
- Oncology Research Unit, Department of Urology and Division of Surgical Research, University Hospital of Zurich, Switzerland
| | - Tullio Sulser
- Oncology Research Unit, Department of Urology and Division of Surgical Research, University Hospital of Zurich, Switzerland
| | - Maurizio Provenzano
- Oncology Research Unit, Department of Urology and Division of Surgical Research, University Hospital of Zurich, Switzerland
| |
Collapse
|
27
|
Jones RJ, Hussain SA, Protheroe AS, Birtle A, Chakraborti P, Huddart RA, Jagdev S, Bahl A, Stockdale A, Sundar S, Crabb SJ, Dixon-Hughes J, Alexander L, Morris A, Kelly C, Stobo J, Paul J, Powles T. Randomized Phase II Study Investigating Pazopanib Versus Weekly Paclitaxel in Relapsed or Progressive Urothelial Cancer. J Clin Oncol 2017; 35:1770-1777. [PMID: 28402747 DOI: 10.1200/jco.2016.70.7828] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Purpose Two previous single-arm trials have drawn conflicting conclusions regarding the activity of pazopanib in urothelial cancers after failure of platinum-based chemotherapy. Patients and Methods This randomized (1:1) open-label phase II trial compared the efficacy of pazopanib 800 mg orally with paclitaxel (80 mg/m2 days 1, 8, and 15 every 28 days) in the second-line setting. The primary end point was overall survival (OS). Results Between August 2012 and October 2014, 131 patients, out of 140 planned, were randomly assigned. The study was terminated early on the recommendation of the independent data monitoring committee because of futility. Final analysis after the preplanned number of deaths (n = 110) occurred after a median follow-up of 18 months. One hundred fifteen deaths had occurred at the final data extract presented here. Median OS was 8.0 months for paclitaxel (80% CI, 6.9 to 9.7 months) and 4.7 months for pazopanib (80% CI, 4.2 to 6.4 months). The hazard ratio (HR) adjusted for baseline stratification factors was 1.28 (80% CI, 0.99 to 1.67; one-sided P = .89). Median progression-free survival was 4.1 months for paclitaxel (80% CI, 3.0 to 5.6 months) and 3.1 months for pazopanib (80% CI, 2.7 to 4.6 months; HR, 1.09; 80% CI, 0.85 to 1.40; one-sided P = .67). Discontinuations for toxicity occurred in 7.8% and 23.1% for paclitaxel and pazopanib, respectively. Conclusion Pazopanib did not have greater efficacy than paclitaxel in the second-line treatment of urothelial cancers. There was a trend toward superior OS for paclitaxel.
Collapse
Affiliation(s)
- Robert J Jones
- Robert J. Jones, Judith Dixon-Hughes, Laura Alexander, Anna Morris, Caroline Kelly, Jon Stobo, and James Paul, University of Glasgow, Glasgow; Syed A. Hussain, University of Liverpool, Liverpool; Andrew S. Protheroe, Churchill Hospital, Oxford; Alison Birtle, Royal Preston Hospital, Preston; Prabir Chakraborti, Royal Derby Hospital, Derby; Robert A. Huddart, Institute of Cancer Research, Sutton; Satinder Jagdev, St James's University Hospital, Leeds; Amit Bahl, Bristol Haematology and Oncology Centre, Bristol; Andrew Stockdale, University Hospital, Coventry; Santhanam Sundar, Nottingham University Hospitals National Health Service Trust, Nottingham; Simon J. Crabb, University of Southampton, Southampton; and Thomas Powles, Queen Mary University of London, London, United Kingdom
| | - Syed A Hussain
- Robert J. Jones, Judith Dixon-Hughes, Laura Alexander, Anna Morris, Caroline Kelly, Jon Stobo, and James Paul, University of Glasgow, Glasgow; Syed A. Hussain, University of Liverpool, Liverpool; Andrew S. Protheroe, Churchill Hospital, Oxford; Alison Birtle, Royal Preston Hospital, Preston; Prabir Chakraborti, Royal Derby Hospital, Derby; Robert A. Huddart, Institute of Cancer Research, Sutton; Satinder Jagdev, St James's University Hospital, Leeds; Amit Bahl, Bristol Haematology and Oncology Centre, Bristol; Andrew Stockdale, University Hospital, Coventry; Santhanam Sundar, Nottingham University Hospitals National Health Service Trust, Nottingham; Simon J. Crabb, University of Southampton, Southampton; and Thomas Powles, Queen Mary University of London, London, United Kingdom
| | - Andrew S Protheroe
- Robert J. Jones, Judith Dixon-Hughes, Laura Alexander, Anna Morris, Caroline Kelly, Jon Stobo, and James Paul, University of Glasgow, Glasgow; Syed A. Hussain, University of Liverpool, Liverpool; Andrew S. Protheroe, Churchill Hospital, Oxford; Alison Birtle, Royal Preston Hospital, Preston; Prabir Chakraborti, Royal Derby Hospital, Derby; Robert A. Huddart, Institute of Cancer Research, Sutton; Satinder Jagdev, St James's University Hospital, Leeds; Amit Bahl, Bristol Haematology and Oncology Centre, Bristol; Andrew Stockdale, University Hospital, Coventry; Santhanam Sundar, Nottingham University Hospitals National Health Service Trust, Nottingham; Simon J. Crabb, University of Southampton, Southampton; and Thomas Powles, Queen Mary University of London, London, United Kingdom
| | - Alison Birtle
- Robert J. Jones, Judith Dixon-Hughes, Laura Alexander, Anna Morris, Caroline Kelly, Jon Stobo, and James Paul, University of Glasgow, Glasgow; Syed A. Hussain, University of Liverpool, Liverpool; Andrew S. Protheroe, Churchill Hospital, Oxford; Alison Birtle, Royal Preston Hospital, Preston; Prabir Chakraborti, Royal Derby Hospital, Derby; Robert A. Huddart, Institute of Cancer Research, Sutton; Satinder Jagdev, St James's University Hospital, Leeds; Amit Bahl, Bristol Haematology and Oncology Centre, Bristol; Andrew Stockdale, University Hospital, Coventry; Santhanam Sundar, Nottingham University Hospitals National Health Service Trust, Nottingham; Simon J. Crabb, University of Southampton, Southampton; and Thomas Powles, Queen Mary University of London, London, United Kingdom
| | - Prabir Chakraborti
- Robert J. Jones, Judith Dixon-Hughes, Laura Alexander, Anna Morris, Caroline Kelly, Jon Stobo, and James Paul, University of Glasgow, Glasgow; Syed A. Hussain, University of Liverpool, Liverpool; Andrew S. Protheroe, Churchill Hospital, Oxford; Alison Birtle, Royal Preston Hospital, Preston; Prabir Chakraborti, Royal Derby Hospital, Derby; Robert A. Huddart, Institute of Cancer Research, Sutton; Satinder Jagdev, St James's University Hospital, Leeds; Amit Bahl, Bristol Haematology and Oncology Centre, Bristol; Andrew Stockdale, University Hospital, Coventry; Santhanam Sundar, Nottingham University Hospitals National Health Service Trust, Nottingham; Simon J. Crabb, University of Southampton, Southampton; and Thomas Powles, Queen Mary University of London, London, United Kingdom
| | - Robert A Huddart
- Robert J. Jones, Judith Dixon-Hughes, Laura Alexander, Anna Morris, Caroline Kelly, Jon Stobo, and James Paul, University of Glasgow, Glasgow; Syed A. Hussain, University of Liverpool, Liverpool; Andrew S. Protheroe, Churchill Hospital, Oxford; Alison Birtle, Royal Preston Hospital, Preston; Prabir Chakraborti, Royal Derby Hospital, Derby; Robert A. Huddart, Institute of Cancer Research, Sutton; Satinder Jagdev, St James's University Hospital, Leeds; Amit Bahl, Bristol Haematology and Oncology Centre, Bristol; Andrew Stockdale, University Hospital, Coventry; Santhanam Sundar, Nottingham University Hospitals National Health Service Trust, Nottingham; Simon J. Crabb, University of Southampton, Southampton; and Thomas Powles, Queen Mary University of London, London, United Kingdom
| | - Satinder Jagdev
- Robert J. Jones, Judith Dixon-Hughes, Laura Alexander, Anna Morris, Caroline Kelly, Jon Stobo, and James Paul, University of Glasgow, Glasgow; Syed A. Hussain, University of Liverpool, Liverpool; Andrew S. Protheroe, Churchill Hospital, Oxford; Alison Birtle, Royal Preston Hospital, Preston; Prabir Chakraborti, Royal Derby Hospital, Derby; Robert A. Huddart, Institute of Cancer Research, Sutton; Satinder Jagdev, St James's University Hospital, Leeds; Amit Bahl, Bristol Haematology and Oncology Centre, Bristol; Andrew Stockdale, University Hospital, Coventry; Santhanam Sundar, Nottingham University Hospitals National Health Service Trust, Nottingham; Simon J. Crabb, University of Southampton, Southampton; and Thomas Powles, Queen Mary University of London, London, United Kingdom
| | - Amit Bahl
- Robert J. Jones, Judith Dixon-Hughes, Laura Alexander, Anna Morris, Caroline Kelly, Jon Stobo, and James Paul, University of Glasgow, Glasgow; Syed A. Hussain, University of Liverpool, Liverpool; Andrew S. Protheroe, Churchill Hospital, Oxford; Alison Birtle, Royal Preston Hospital, Preston; Prabir Chakraborti, Royal Derby Hospital, Derby; Robert A. Huddart, Institute of Cancer Research, Sutton; Satinder Jagdev, St James's University Hospital, Leeds; Amit Bahl, Bristol Haematology and Oncology Centre, Bristol; Andrew Stockdale, University Hospital, Coventry; Santhanam Sundar, Nottingham University Hospitals National Health Service Trust, Nottingham; Simon J. Crabb, University of Southampton, Southampton; and Thomas Powles, Queen Mary University of London, London, United Kingdom
| | - Andrew Stockdale
- Robert J. Jones, Judith Dixon-Hughes, Laura Alexander, Anna Morris, Caroline Kelly, Jon Stobo, and James Paul, University of Glasgow, Glasgow; Syed A. Hussain, University of Liverpool, Liverpool; Andrew S. Protheroe, Churchill Hospital, Oxford; Alison Birtle, Royal Preston Hospital, Preston; Prabir Chakraborti, Royal Derby Hospital, Derby; Robert A. Huddart, Institute of Cancer Research, Sutton; Satinder Jagdev, St James's University Hospital, Leeds; Amit Bahl, Bristol Haematology and Oncology Centre, Bristol; Andrew Stockdale, University Hospital, Coventry; Santhanam Sundar, Nottingham University Hospitals National Health Service Trust, Nottingham; Simon J. Crabb, University of Southampton, Southampton; and Thomas Powles, Queen Mary University of London, London, United Kingdom
| | - Santhanam Sundar
- Robert J. Jones, Judith Dixon-Hughes, Laura Alexander, Anna Morris, Caroline Kelly, Jon Stobo, and James Paul, University of Glasgow, Glasgow; Syed A. Hussain, University of Liverpool, Liverpool; Andrew S. Protheroe, Churchill Hospital, Oxford; Alison Birtle, Royal Preston Hospital, Preston; Prabir Chakraborti, Royal Derby Hospital, Derby; Robert A. Huddart, Institute of Cancer Research, Sutton; Satinder Jagdev, St James's University Hospital, Leeds; Amit Bahl, Bristol Haematology and Oncology Centre, Bristol; Andrew Stockdale, University Hospital, Coventry; Santhanam Sundar, Nottingham University Hospitals National Health Service Trust, Nottingham; Simon J. Crabb, University of Southampton, Southampton; and Thomas Powles, Queen Mary University of London, London, United Kingdom
| | - Simon J Crabb
- Robert J. Jones, Judith Dixon-Hughes, Laura Alexander, Anna Morris, Caroline Kelly, Jon Stobo, and James Paul, University of Glasgow, Glasgow; Syed A. Hussain, University of Liverpool, Liverpool; Andrew S. Protheroe, Churchill Hospital, Oxford; Alison Birtle, Royal Preston Hospital, Preston; Prabir Chakraborti, Royal Derby Hospital, Derby; Robert A. Huddart, Institute of Cancer Research, Sutton; Satinder Jagdev, St James's University Hospital, Leeds; Amit Bahl, Bristol Haematology and Oncology Centre, Bristol; Andrew Stockdale, University Hospital, Coventry; Santhanam Sundar, Nottingham University Hospitals National Health Service Trust, Nottingham; Simon J. Crabb, University of Southampton, Southampton; and Thomas Powles, Queen Mary University of London, London, United Kingdom
| | - Judith Dixon-Hughes
- Robert J. Jones, Judith Dixon-Hughes, Laura Alexander, Anna Morris, Caroline Kelly, Jon Stobo, and James Paul, University of Glasgow, Glasgow; Syed A. Hussain, University of Liverpool, Liverpool; Andrew S. Protheroe, Churchill Hospital, Oxford; Alison Birtle, Royal Preston Hospital, Preston; Prabir Chakraborti, Royal Derby Hospital, Derby; Robert A. Huddart, Institute of Cancer Research, Sutton; Satinder Jagdev, St James's University Hospital, Leeds; Amit Bahl, Bristol Haematology and Oncology Centre, Bristol; Andrew Stockdale, University Hospital, Coventry; Santhanam Sundar, Nottingham University Hospitals National Health Service Trust, Nottingham; Simon J. Crabb, University of Southampton, Southampton; and Thomas Powles, Queen Mary University of London, London, United Kingdom
| | - Laura Alexander
- Robert J. Jones, Judith Dixon-Hughes, Laura Alexander, Anna Morris, Caroline Kelly, Jon Stobo, and James Paul, University of Glasgow, Glasgow; Syed A. Hussain, University of Liverpool, Liverpool; Andrew S. Protheroe, Churchill Hospital, Oxford; Alison Birtle, Royal Preston Hospital, Preston; Prabir Chakraborti, Royal Derby Hospital, Derby; Robert A. Huddart, Institute of Cancer Research, Sutton; Satinder Jagdev, St James's University Hospital, Leeds; Amit Bahl, Bristol Haematology and Oncology Centre, Bristol; Andrew Stockdale, University Hospital, Coventry; Santhanam Sundar, Nottingham University Hospitals National Health Service Trust, Nottingham; Simon J. Crabb, University of Southampton, Southampton; and Thomas Powles, Queen Mary University of London, London, United Kingdom
| | - Anna Morris
- Robert J. Jones, Judith Dixon-Hughes, Laura Alexander, Anna Morris, Caroline Kelly, Jon Stobo, and James Paul, University of Glasgow, Glasgow; Syed A. Hussain, University of Liverpool, Liverpool; Andrew S. Protheroe, Churchill Hospital, Oxford; Alison Birtle, Royal Preston Hospital, Preston; Prabir Chakraborti, Royal Derby Hospital, Derby; Robert A. Huddart, Institute of Cancer Research, Sutton; Satinder Jagdev, St James's University Hospital, Leeds; Amit Bahl, Bristol Haematology and Oncology Centre, Bristol; Andrew Stockdale, University Hospital, Coventry; Santhanam Sundar, Nottingham University Hospitals National Health Service Trust, Nottingham; Simon J. Crabb, University of Southampton, Southampton; and Thomas Powles, Queen Mary University of London, London, United Kingdom
| | - Caroline Kelly
- Robert J. Jones, Judith Dixon-Hughes, Laura Alexander, Anna Morris, Caroline Kelly, Jon Stobo, and James Paul, University of Glasgow, Glasgow; Syed A. Hussain, University of Liverpool, Liverpool; Andrew S. Protheroe, Churchill Hospital, Oxford; Alison Birtle, Royal Preston Hospital, Preston; Prabir Chakraborti, Royal Derby Hospital, Derby; Robert A. Huddart, Institute of Cancer Research, Sutton; Satinder Jagdev, St James's University Hospital, Leeds; Amit Bahl, Bristol Haematology and Oncology Centre, Bristol; Andrew Stockdale, University Hospital, Coventry; Santhanam Sundar, Nottingham University Hospitals National Health Service Trust, Nottingham; Simon J. Crabb, University of Southampton, Southampton; and Thomas Powles, Queen Mary University of London, London, United Kingdom
| | - Jon Stobo
- Robert J. Jones, Judith Dixon-Hughes, Laura Alexander, Anna Morris, Caroline Kelly, Jon Stobo, and James Paul, University of Glasgow, Glasgow; Syed A. Hussain, University of Liverpool, Liverpool; Andrew S. Protheroe, Churchill Hospital, Oxford; Alison Birtle, Royal Preston Hospital, Preston; Prabir Chakraborti, Royal Derby Hospital, Derby; Robert A. Huddart, Institute of Cancer Research, Sutton; Satinder Jagdev, St James's University Hospital, Leeds; Amit Bahl, Bristol Haematology and Oncology Centre, Bristol; Andrew Stockdale, University Hospital, Coventry; Santhanam Sundar, Nottingham University Hospitals National Health Service Trust, Nottingham; Simon J. Crabb, University of Southampton, Southampton; and Thomas Powles, Queen Mary University of London, London, United Kingdom
| | - James Paul
- Robert J. Jones, Judith Dixon-Hughes, Laura Alexander, Anna Morris, Caroline Kelly, Jon Stobo, and James Paul, University of Glasgow, Glasgow; Syed A. Hussain, University of Liverpool, Liverpool; Andrew S. Protheroe, Churchill Hospital, Oxford; Alison Birtle, Royal Preston Hospital, Preston; Prabir Chakraborti, Royal Derby Hospital, Derby; Robert A. Huddart, Institute of Cancer Research, Sutton; Satinder Jagdev, St James's University Hospital, Leeds; Amit Bahl, Bristol Haematology and Oncology Centre, Bristol; Andrew Stockdale, University Hospital, Coventry; Santhanam Sundar, Nottingham University Hospitals National Health Service Trust, Nottingham; Simon J. Crabb, University of Southampton, Southampton; and Thomas Powles, Queen Mary University of London, London, United Kingdom
| | - Thomas Powles
- Robert J. Jones, Judith Dixon-Hughes, Laura Alexander, Anna Morris, Caroline Kelly, Jon Stobo, and James Paul, University of Glasgow, Glasgow; Syed A. Hussain, University of Liverpool, Liverpool; Andrew S. Protheroe, Churchill Hospital, Oxford; Alison Birtle, Royal Preston Hospital, Preston; Prabir Chakraborti, Royal Derby Hospital, Derby; Robert A. Huddart, Institute of Cancer Research, Sutton; Satinder Jagdev, St James's University Hospital, Leeds; Amit Bahl, Bristol Haematology and Oncology Centre, Bristol; Andrew Stockdale, University Hospital, Coventry; Santhanam Sundar, Nottingham University Hospitals National Health Service Trust, Nottingham; Simon J. Crabb, University of Southampton, Southampton; and Thomas Powles, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
28
|
Yoshida T, Matsuzaki T, Murota T, Kawa G, Matsuda T, Kinoshita H. Evaluation of the Clinical Utility of Renin-Angiotensin System Inhibitors in Patients Undergoing Radical Surgery for Urothelial Carcinoma of the Upper Urinary Tract. Clin Genitourin Cancer 2017; 15:e943-e954. [PMID: 28552573 DOI: 10.1016/j.clgc.2017.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 04/25/2017] [Accepted: 05/01/2017] [Indexed: 10/19/2022]
Abstract
BACKGROUND Renin-angiotensin system (RAS) inhibitors are effective for treating patients with cancer. The present study evaluated the impact of RAS inhibitors, including angiotensin-2 converting enzyme inhibitors and angiotensin 2 receptor blockers, after patients underwent radical surgery for upper urinary tract urothelial carcinoma (UTUC). METHODS This retrospective study included 312 patients with nonmetastatic UTUC who underwent radical surgery. The oncological outcomes of patients treated or not treated with RAS inhibitors following surgery were evaluated. Recurrence-free survival (RFS), cancer-specific survival (CSS), and overall survival (OS) were assessed using the Kaplan-Meier method and Cox regression analysis. RESULTS The median follow-up duration after radical surgery was 44.7 months. The 5-year RFS, CSS, and OS rates of patients who did or did not receive RAS inhibitors were 82.3% versus 68.9% (P = .018), 88.9% versus 71.8% (P = .0044), and 68.7% versus 61.8% (P = .047), respectively. Multivariable analyses revealed that the use of RAS inhibitors was an independent prognostic factor for RFS, CSS, and OS (hazard ratio [HR] 0.48, P = .013; HR 0.31, P = .002; and HR 0.52, P = .01, respectively). Moreover, patients treated with RAS inhibitors versus untreated patients had better 5-year RFS compared with those in the pT2 and < pN1 subgroups (pT2: 100.0% vs. 62.2%, P = .014 and < pN1: 87.2% vs. 74.7%, P = .034). CONCLUSIONS RAS inhibitors significantly improved RFS, CSS, and OS of patients with UTUC who underwent radical surgery. These agents may be particularly beneficial for patients with stage pT2 or < pN1 disease.
Collapse
Affiliation(s)
- Takashi Yoshida
- Department of Urology and Andrology, Kori Hospital, Kansai Medical University, Osaka, Japan
| | - Tomoaki Matsuzaki
- Department of Urology and Andrology, General Medical Hospital, Kansai Medical University, Osaka, Japan
| | - Takashi Murota
- Department of Urology and Andrology, General Medical Hospital, Kansai Medical University, Osaka, Japan
| | - Gen Kawa
- Department of Urology, Saisekai Noe Hospital, Osaka, Japan
| | - Tadashi Matsuda
- Department of Urology and Andrology, Kansai Medical University Hospital, Osaka, Japan
| | - Hidefumi Kinoshita
- Department of Urology and Andrology, Kansai Medical University Hospital, Osaka, Japan.
| |
Collapse
|
29
|
Ceci C, Tentori L, Atzori MG, Lacal PM, Bonanno E, Scimeca M, Cicconi R, Mattei M, de Martino MG, Vespasiani G, Miano R, Graziani G. Ellagic Acid Inhibits Bladder Cancer Invasiveness and In Vivo Tumor Growth. Nutrients 2016; 8:744. [PMID: 27879653 PMCID: PMC5133127 DOI: 10.3390/nu8110744] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 11/11/2016] [Accepted: 11/16/2016] [Indexed: 12/15/2022] Open
Abstract
Ellagic acid (EA) is a polyphenolic compound that can be found as a naturally occurring hydrolysis product of ellagitannins in pomegranates, berries, grapes, green tea and nuts. Previous studies have reported the antitumor properties of EA mainly using in vitro models. No data are available about EA influence on bladder cancer cell invasion of the extracellular matrix triggered by vascular endothelial growth factor-A (VEGF-A), an angiogenic factor associated with disease progression and recurrence, and tumor growth in vivo. In this study, we have investigated EA activity against four different human bladder cancer cell lines (i.e., T24, UM-UC-3, 5637 and HT-1376) by in vitro proliferation tests (measuring metabolic and foci forming activity), invasion and chemotactic assays in response to VEGF-A and in vivo preclinical models in nude mice. Results indicate that EA exerts anti-proliferative effects as a single agent and enhances the antitumor activity of mitomycin C, which is commonly used for the treatment of bladder cancer. EA also inhibits tumor invasion and chemotaxis, specifically induced by VEGF-A, and reduces VEGFR-2 expression. Moreover, EA down-regulates the expression of programmed cell death ligand 1 (PD-L1), an immune checkpoint involved in immune escape. EA in vitro activity was confirmed by the results of in vivo studies showing a significant reduction of the growth rate, infiltrative behavior and tumor-associated angiogenesis of human bladder cancer xenografts. In conclusion, these results suggest that EA may have a potential role as an adjunct therapy for bladder cancer.
Collapse
Affiliation(s)
- Claudia Ceci
- Department of Systems Medicine, University of Rome Tor Vergata, Rome 00173, Italy.
| | - Lucio Tentori
- Department of Systems Medicine, University of Rome Tor Vergata, Rome 00173, Italy.
| | - Maria Grazia Atzori
- Department of Systems Medicine, University of Rome Tor Vergata, Rome 00173, Italy.
| | - Pedro M Lacal
- Laboratory of Molecular Oncology, "Istituto Dermopatico dell'Immacolata"-IRCCS, Rome 00167, Italy.
| | - Elena Bonanno
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome 00173, Italy.
| | - Manuel Scimeca
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome 00173, Italy.
| | - Rosella Cicconi
- "Centro di Servizi Interdipartimentale, Stazione per la Tecnologia Animale", Department of Biology, University of Rome Tor Vergata, Rome 00173, Italy.
| | - Maurizio Mattei
- "Centro di Servizi Interdipartimentale, Stazione per la Tecnologia Animale", Department of Biology, University of Rome Tor Vergata, Rome 00173, Italy.
| | - Maria Gabriella de Martino
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome 00173, Italy.
| | - Giuseppe Vespasiani
- Urology Unit, Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome 00173, Italy.
| | - Roberto Miano
- Urology Unit, Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome 00173, Italy.
| | - Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Rome 00173, Italy.
| |
Collapse
|
30
|
Yoshida T, Kinoshita H, Fukui K, Matsuzaki T, Yoshida K, Mishima T, Yanishi M, Komai Y, Sugi M, Inoue T, Murota T, Matsuda T. Prognostic Impact of Renin-Angiotensin Inhibitors in Patients with Bladder Cancer Undergoing Radical Cystectomy. Ann Surg Oncol 2016; 24:823-831. [PMID: 27730369 DOI: 10.1245/s10434-016-5534-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Indexed: 01/17/2023]
Abstract
BACKGROUND Renin-angiotensin system blockade has been effective for the treatment of patients with several types of malignancy. This study evaluated the prognostic impact of renin-angiotensin system inhibitors, including angiotensin-2 converting enzyme inhibitors and angiotensin 2 receptor blockers, in patients with bladder cancer undergoing radical cystectomy. METHODS This retrospective study included 269 patients who had undergone radical cystectomy. The oncologic outcomes of patients treated or not treated with renin-angiotensin system inhibitors after surgery were evaluated. Overall survival and cancer-specific survival were assessed by the Kaplan-Meier method and by Cox regression analysis. RESULTS The median follow-up duration after radical cystectomy in survivors was 44.5 months. The 5-year, cancer-specific survival rates in patients who did and did not receive renin-angiotensin system inhibitors were 79.0 and 66.4 %, respectively (P = 0.011). Similarly, the 5-year overall survival rates were 76.1 and 61.4 %, respectively (P = 0.0097). Multivariable analyses showed that use of renin-angiotensin system inhibitors was an independent prognostic factor for cancer-specific survival (hazard ratio 0.47, P = 0.036) and for overall survival (hazard ratio 0.36, P = 0.022). CONCLUSIONS Renin-angiotensin system inhibitors significantly reduced the risks of cancer-specific and overall mortality after radical cystectomy in patients with bladder cancer. Renin-angiotensin system inhibitors may improve oncologic outcomes in high-risk patients with bladder cancer.
Collapse
Affiliation(s)
- Takashi Yoshida
- Department of Urology and Andrology, Kansai Medical University, Kori Hospital, Osaka, Japan.,Department of Urology and Andrology, Kansai Medical University Hospital, Osaka, Japan
| | - Hidefumi Kinoshita
- Department of Urology and Andrology, Kansai Medical University Hospital, Osaka, Japan.
| | - Katsuya Fukui
- Department of Urology and Andrology, Kansai Medical University, Kori Hospital, Osaka, Japan
| | - Tomoaki Matsuzaki
- Department of Urology and Andrology, Kansai Medical University, General Medical Center, Osaka, Japan
| | - Kenji Yoshida
- Department of Urology and Andrology, Kansai Medical University Hospital, Osaka, Japan
| | - Takao Mishima
- Department of Urology and Andrology, Kansai Medical University Hospital, Osaka, Japan
| | - Masaaki Yanishi
- Department of Urology and Andrology, Kansai Medical University Hospital, Osaka, Japan
| | - Yoshihiro Komai
- Department of Urology and Andrology, Kansai Medical University Hospital, Osaka, Japan
| | - Motohiko Sugi
- Department of Urology and Andrology, Kansai Medical University Hospital, Osaka, Japan
| | - Takaaki Inoue
- Department of Urology and Andrology, Kansai Medical University, General Medical Center, Osaka, Japan
| | - Takashi Murota
- Department of Urology and Andrology, Kansai Medical University, General Medical Center, Osaka, Japan
| | - Tadashi Matsuda
- Department of Urology and Andrology, Kansai Medical University Hospital, Osaka, Japan
| |
Collapse
|
31
|
Sonni I, Iagaru A. PET Imaging Toward Individualized Management of Urologic and Gynecologic Malignancies. PET Clin 2016; 11:261-72. [DOI: 10.1016/j.cpet.2016.02.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
32
|
Petrylak DP, Tagawa ST, Kohli M, Eisen A, Canil C, Sridhar SS, Spira A, Yu EY, Burke JM, Shaffer D, Pan CX, Kim JJ, Aragon-Ching JB, Quinn DI, Vogelzang NJ, Tang S, Zhang H, Cavanaugh CT, Gao L, Kauh JS, Walgren RA, Chi KN. Docetaxel As Monotherapy or Combined With Ramucirumab or Icrucumab in Second-Line Treatment for Locally Advanced or Metastatic Urothelial Carcinoma: An Open-Label, Three-Arm, Randomized Controlled Phase II Trial. J Clin Oncol 2016; 34:1500-9. [PMID: 26926681 DOI: 10.1200/jco.2015.65.0218] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
PURPOSE This trial assessed the efficacy and safety of docetaxel monotherapy or docetaxel in combination with ramucirumab (vascular endothelial growth factor receptor 2 antibody) or icrucumab (vascular endothelial growth factor receptor 1 antibody) after progression during or within 12 months of platinum-based regimens for patients with locally advanced or metastatic urothelial carcinoma. PATIENTS AND METHODS Patients were randomly assigned (1:1:1) to receive docetaxel 75 mg/m(2) intravenously (IV) on day 1 of a 3-week cycle (arm A), docetaxel 75 mg/m(2) IV plus ramucirumab 10 mg/kg IV on day 1 of a 3-week cycle (arm B), or docetaxel 75 mg/m(2) IV on day 1 plus icrucumab 12 mg/kg IV on days 1 and 8 of a 3-week cycle (arm C). Treatment continued until disease progression or unacceptable toxicity. The primary end point was investigator-assessed progression-free survival (PFS). RESULTS A total of 140 patients were randomly assigned and treated in arms A (n = 45), B (n = 46), or C (n = 49). PFS was significantly longer in arm B compared with arm A (median, 5.4 months; 95% CI, 3.1 to 6.9 months v 2.8 months; 95% CI, 1.9 to 3.6 months; stratified hazard ratio, 0.389; 95% CI, 0.235 to 0.643; P = .0002). Arm C did not experience improved PFS compared with arm A (1.6 months; 95% CI, 1.4 to 2.9; stratified hazard ratio, 0.863; 95% CI, 0.550 to 1.357; P = .5053). The most common grade 3 or worse adverse events (arms A, B, and C) were neutropenia (36%, 33%, and 39%), fatigue (13%, 30%, and 20%), febrile neutropenia (13%, 17%, and 6.1%), and anemia (6.7%, 13%, and 14%, respectively). CONCLUSION The addition of ramucirumab to docetaxel met the prespecified efficacy end point for prolonging PFS in patients with locally advanced or metastatic urothelial carcinoma receiving second-line treatment and warrants further investigation in the phase III setting.
Collapse
Affiliation(s)
- Daniel P Petrylak
- Daniel P. Petrylak, Yale University Cancer Center, New Haven, CT; Scott T. Tagawa, Weill Cornell Medical College, New York; David Shaffer, New York Oncology Hematology, Albany, NY; Manish Kohli, Mayo Clinic, Rochester, MN; Andrea Eisen, Sunnybrook Health Sciences Centre; Srikala S. Sridhar, Princess Margaret Hospital, Toronto; Christina Canil, Ottawa Hospital Cancer Centre, Ottawa, Ontario; Kim N. Chi, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; Alexander Spira, John M. Burke, David Shaffer, and Nicholas J. Vogelzang, US Oncology Research, The Woodlands, TX; Evan Y. Yu, University of Washington Medical Center and Fred Hutchinson Cancer Research Center, Seattle, WA; John M. Burke, Rocky Mountain Cancer Centers, Aurora, CO; Chong-Xian Pan, University of California-Davis Medical Center, Sacramento; David I. Quinn, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Jenny J. Kim, Johns Hopkins University, Baltimore, MD; Jeanny B. Aragon-Ching, George Washington University Medical Center, Washington, DC; Nicholas J. Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Shande Tang, Hui Zhang, Christopher T. Cavanaugh, Ling Gao, and John S. Kauh, Eli Lilly, Bridgewater, NJ; and Richard A. Walgren, Eli Lilly, Indianapolis, IN.
| | - Scott T Tagawa
- Daniel P. Petrylak, Yale University Cancer Center, New Haven, CT; Scott T. Tagawa, Weill Cornell Medical College, New York; David Shaffer, New York Oncology Hematology, Albany, NY; Manish Kohli, Mayo Clinic, Rochester, MN; Andrea Eisen, Sunnybrook Health Sciences Centre; Srikala S. Sridhar, Princess Margaret Hospital, Toronto; Christina Canil, Ottawa Hospital Cancer Centre, Ottawa, Ontario; Kim N. Chi, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; Alexander Spira, John M. Burke, David Shaffer, and Nicholas J. Vogelzang, US Oncology Research, The Woodlands, TX; Evan Y. Yu, University of Washington Medical Center and Fred Hutchinson Cancer Research Center, Seattle, WA; John M. Burke, Rocky Mountain Cancer Centers, Aurora, CO; Chong-Xian Pan, University of California-Davis Medical Center, Sacramento; David I. Quinn, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Jenny J. Kim, Johns Hopkins University, Baltimore, MD; Jeanny B. Aragon-Ching, George Washington University Medical Center, Washington, DC; Nicholas J. Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Shande Tang, Hui Zhang, Christopher T. Cavanaugh, Ling Gao, and John S. Kauh, Eli Lilly, Bridgewater, NJ; and Richard A. Walgren, Eli Lilly, Indianapolis, IN
| | - Manish Kohli
- Daniel P. Petrylak, Yale University Cancer Center, New Haven, CT; Scott T. Tagawa, Weill Cornell Medical College, New York; David Shaffer, New York Oncology Hematology, Albany, NY; Manish Kohli, Mayo Clinic, Rochester, MN; Andrea Eisen, Sunnybrook Health Sciences Centre; Srikala S. Sridhar, Princess Margaret Hospital, Toronto; Christina Canil, Ottawa Hospital Cancer Centre, Ottawa, Ontario; Kim N. Chi, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; Alexander Spira, John M. Burke, David Shaffer, and Nicholas J. Vogelzang, US Oncology Research, The Woodlands, TX; Evan Y. Yu, University of Washington Medical Center and Fred Hutchinson Cancer Research Center, Seattle, WA; John M. Burke, Rocky Mountain Cancer Centers, Aurora, CO; Chong-Xian Pan, University of California-Davis Medical Center, Sacramento; David I. Quinn, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Jenny J. Kim, Johns Hopkins University, Baltimore, MD; Jeanny B. Aragon-Ching, George Washington University Medical Center, Washington, DC; Nicholas J. Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Shande Tang, Hui Zhang, Christopher T. Cavanaugh, Ling Gao, and John S. Kauh, Eli Lilly, Bridgewater, NJ; and Richard A. Walgren, Eli Lilly, Indianapolis, IN
| | - Andrea Eisen
- Daniel P. Petrylak, Yale University Cancer Center, New Haven, CT; Scott T. Tagawa, Weill Cornell Medical College, New York; David Shaffer, New York Oncology Hematology, Albany, NY; Manish Kohli, Mayo Clinic, Rochester, MN; Andrea Eisen, Sunnybrook Health Sciences Centre; Srikala S. Sridhar, Princess Margaret Hospital, Toronto; Christina Canil, Ottawa Hospital Cancer Centre, Ottawa, Ontario; Kim N. Chi, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; Alexander Spira, John M. Burke, David Shaffer, and Nicholas J. Vogelzang, US Oncology Research, The Woodlands, TX; Evan Y. Yu, University of Washington Medical Center and Fred Hutchinson Cancer Research Center, Seattle, WA; John M. Burke, Rocky Mountain Cancer Centers, Aurora, CO; Chong-Xian Pan, University of California-Davis Medical Center, Sacramento; David I. Quinn, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Jenny J. Kim, Johns Hopkins University, Baltimore, MD; Jeanny B. Aragon-Ching, George Washington University Medical Center, Washington, DC; Nicholas J. Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Shande Tang, Hui Zhang, Christopher T. Cavanaugh, Ling Gao, and John S. Kauh, Eli Lilly, Bridgewater, NJ; and Richard A. Walgren, Eli Lilly, Indianapolis, IN
| | - Christina Canil
- Daniel P. Petrylak, Yale University Cancer Center, New Haven, CT; Scott T. Tagawa, Weill Cornell Medical College, New York; David Shaffer, New York Oncology Hematology, Albany, NY; Manish Kohli, Mayo Clinic, Rochester, MN; Andrea Eisen, Sunnybrook Health Sciences Centre; Srikala S. Sridhar, Princess Margaret Hospital, Toronto; Christina Canil, Ottawa Hospital Cancer Centre, Ottawa, Ontario; Kim N. Chi, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; Alexander Spira, John M. Burke, David Shaffer, and Nicholas J. Vogelzang, US Oncology Research, The Woodlands, TX; Evan Y. Yu, University of Washington Medical Center and Fred Hutchinson Cancer Research Center, Seattle, WA; John M. Burke, Rocky Mountain Cancer Centers, Aurora, CO; Chong-Xian Pan, University of California-Davis Medical Center, Sacramento; David I. Quinn, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Jenny J. Kim, Johns Hopkins University, Baltimore, MD; Jeanny B. Aragon-Ching, George Washington University Medical Center, Washington, DC; Nicholas J. Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Shande Tang, Hui Zhang, Christopher T. Cavanaugh, Ling Gao, and John S. Kauh, Eli Lilly, Bridgewater, NJ; and Richard A. Walgren, Eli Lilly, Indianapolis, IN
| | - Srikala S Sridhar
- Daniel P. Petrylak, Yale University Cancer Center, New Haven, CT; Scott T. Tagawa, Weill Cornell Medical College, New York; David Shaffer, New York Oncology Hematology, Albany, NY; Manish Kohli, Mayo Clinic, Rochester, MN; Andrea Eisen, Sunnybrook Health Sciences Centre; Srikala S. Sridhar, Princess Margaret Hospital, Toronto; Christina Canil, Ottawa Hospital Cancer Centre, Ottawa, Ontario; Kim N. Chi, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; Alexander Spira, John M. Burke, David Shaffer, and Nicholas J. Vogelzang, US Oncology Research, The Woodlands, TX; Evan Y. Yu, University of Washington Medical Center and Fred Hutchinson Cancer Research Center, Seattle, WA; John M. Burke, Rocky Mountain Cancer Centers, Aurora, CO; Chong-Xian Pan, University of California-Davis Medical Center, Sacramento; David I. Quinn, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Jenny J. Kim, Johns Hopkins University, Baltimore, MD; Jeanny B. Aragon-Ching, George Washington University Medical Center, Washington, DC; Nicholas J. Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Shande Tang, Hui Zhang, Christopher T. Cavanaugh, Ling Gao, and John S. Kauh, Eli Lilly, Bridgewater, NJ; and Richard A. Walgren, Eli Lilly, Indianapolis, IN
| | - Alexander Spira
- Daniel P. Petrylak, Yale University Cancer Center, New Haven, CT; Scott T. Tagawa, Weill Cornell Medical College, New York; David Shaffer, New York Oncology Hematology, Albany, NY; Manish Kohli, Mayo Clinic, Rochester, MN; Andrea Eisen, Sunnybrook Health Sciences Centre; Srikala S. Sridhar, Princess Margaret Hospital, Toronto; Christina Canil, Ottawa Hospital Cancer Centre, Ottawa, Ontario; Kim N. Chi, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; Alexander Spira, John M. Burke, David Shaffer, and Nicholas J. Vogelzang, US Oncology Research, The Woodlands, TX; Evan Y. Yu, University of Washington Medical Center and Fred Hutchinson Cancer Research Center, Seattle, WA; John M. Burke, Rocky Mountain Cancer Centers, Aurora, CO; Chong-Xian Pan, University of California-Davis Medical Center, Sacramento; David I. Quinn, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Jenny J. Kim, Johns Hopkins University, Baltimore, MD; Jeanny B. Aragon-Ching, George Washington University Medical Center, Washington, DC; Nicholas J. Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Shande Tang, Hui Zhang, Christopher T. Cavanaugh, Ling Gao, and John S. Kauh, Eli Lilly, Bridgewater, NJ; and Richard A. Walgren, Eli Lilly, Indianapolis, IN
| | - Evan Y Yu
- Daniel P. Petrylak, Yale University Cancer Center, New Haven, CT; Scott T. Tagawa, Weill Cornell Medical College, New York; David Shaffer, New York Oncology Hematology, Albany, NY; Manish Kohli, Mayo Clinic, Rochester, MN; Andrea Eisen, Sunnybrook Health Sciences Centre; Srikala S. Sridhar, Princess Margaret Hospital, Toronto; Christina Canil, Ottawa Hospital Cancer Centre, Ottawa, Ontario; Kim N. Chi, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; Alexander Spira, John M. Burke, David Shaffer, and Nicholas J. Vogelzang, US Oncology Research, The Woodlands, TX; Evan Y. Yu, University of Washington Medical Center and Fred Hutchinson Cancer Research Center, Seattle, WA; John M. Burke, Rocky Mountain Cancer Centers, Aurora, CO; Chong-Xian Pan, University of California-Davis Medical Center, Sacramento; David I. Quinn, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Jenny J. Kim, Johns Hopkins University, Baltimore, MD; Jeanny B. Aragon-Ching, George Washington University Medical Center, Washington, DC; Nicholas J. Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Shande Tang, Hui Zhang, Christopher T. Cavanaugh, Ling Gao, and John S. Kauh, Eli Lilly, Bridgewater, NJ; and Richard A. Walgren, Eli Lilly, Indianapolis, IN
| | - John M Burke
- Daniel P. Petrylak, Yale University Cancer Center, New Haven, CT; Scott T. Tagawa, Weill Cornell Medical College, New York; David Shaffer, New York Oncology Hematology, Albany, NY; Manish Kohli, Mayo Clinic, Rochester, MN; Andrea Eisen, Sunnybrook Health Sciences Centre; Srikala S. Sridhar, Princess Margaret Hospital, Toronto; Christina Canil, Ottawa Hospital Cancer Centre, Ottawa, Ontario; Kim N. Chi, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; Alexander Spira, John M. Burke, David Shaffer, and Nicholas J. Vogelzang, US Oncology Research, The Woodlands, TX; Evan Y. Yu, University of Washington Medical Center and Fred Hutchinson Cancer Research Center, Seattle, WA; John M. Burke, Rocky Mountain Cancer Centers, Aurora, CO; Chong-Xian Pan, University of California-Davis Medical Center, Sacramento; David I. Quinn, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Jenny J. Kim, Johns Hopkins University, Baltimore, MD; Jeanny B. Aragon-Ching, George Washington University Medical Center, Washington, DC; Nicholas J. Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Shande Tang, Hui Zhang, Christopher T. Cavanaugh, Ling Gao, and John S. Kauh, Eli Lilly, Bridgewater, NJ; and Richard A. Walgren, Eli Lilly, Indianapolis, IN
| | - David Shaffer
- Daniel P. Petrylak, Yale University Cancer Center, New Haven, CT; Scott T. Tagawa, Weill Cornell Medical College, New York; David Shaffer, New York Oncology Hematology, Albany, NY; Manish Kohli, Mayo Clinic, Rochester, MN; Andrea Eisen, Sunnybrook Health Sciences Centre; Srikala S. Sridhar, Princess Margaret Hospital, Toronto; Christina Canil, Ottawa Hospital Cancer Centre, Ottawa, Ontario; Kim N. Chi, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; Alexander Spira, John M. Burke, David Shaffer, and Nicholas J. Vogelzang, US Oncology Research, The Woodlands, TX; Evan Y. Yu, University of Washington Medical Center and Fred Hutchinson Cancer Research Center, Seattle, WA; John M. Burke, Rocky Mountain Cancer Centers, Aurora, CO; Chong-Xian Pan, University of California-Davis Medical Center, Sacramento; David I. Quinn, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Jenny J. Kim, Johns Hopkins University, Baltimore, MD; Jeanny B. Aragon-Ching, George Washington University Medical Center, Washington, DC; Nicholas J. Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Shande Tang, Hui Zhang, Christopher T. Cavanaugh, Ling Gao, and John S. Kauh, Eli Lilly, Bridgewater, NJ; and Richard A. Walgren, Eli Lilly, Indianapolis, IN
| | - Chong-Xian Pan
- Daniel P. Petrylak, Yale University Cancer Center, New Haven, CT; Scott T. Tagawa, Weill Cornell Medical College, New York; David Shaffer, New York Oncology Hematology, Albany, NY; Manish Kohli, Mayo Clinic, Rochester, MN; Andrea Eisen, Sunnybrook Health Sciences Centre; Srikala S. Sridhar, Princess Margaret Hospital, Toronto; Christina Canil, Ottawa Hospital Cancer Centre, Ottawa, Ontario; Kim N. Chi, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; Alexander Spira, John M. Burke, David Shaffer, and Nicholas J. Vogelzang, US Oncology Research, The Woodlands, TX; Evan Y. Yu, University of Washington Medical Center and Fred Hutchinson Cancer Research Center, Seattle, WA; John M. Burke, Rocky Mountain Cancer Centers, Aurora, CO; Chong-Xian Pan, University of California-Davis Medical Center, Sacramento; David I. Quinn, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Jenny J. Kim, Johns Hopkins University, Baltimore, MD; Jeanny B. Aragon-Ching, George Washington University Medical Center, Washington, DC; Nicholas J. Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Shande Tang, Hui Zhang, Christopher T. Cavanaugh, Ling Gao, and John S. Kauh, Eli Lilly, Bridgewater, NJ; and Richard A. Walgren, Eli Lilly, Indianapolis, IN
| | - Jenny J Kim
- Daniel P. Petrylak, Yale University Cancer Center, New Haven, CT; Scott T. Tagawa, Weill Cornell Medical College, New York; David Shaffer, New York Oncology Hematology, Albany, NY; Manish Kohli, Mayo Clinic, Rochester, MN; Andrea Eisen, Sunnybrook Health Sciences Centre; Srikala S. Sridhar, Princess Margaret Hospital, Toronto; Christina Canil, Ottawa Hospital Cancer Centre, Ottawa, Ontario; Kim N. Chi, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; Alexander Spira, John M. Burke, David Shaffer, and Nicholas J. Vogelzang, US Oncology Research, The Woodlands, TX; Evan Y. Yu, University of Washington Medical Center and Fred Hutchinson Cancer Research Center, Seattle, WA; John M. Burke, Rocky Mountain Cancer Centers, Aurora, CO; Chong-Xian Pan, University of California-Davis Medical Center, Sacramento; David I. Quinn, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Jenny J. Kim, Johns Hopkins University, Baltimore, MD; Jeanny B. Aragon-Ching, George Washington University Medical Center, Washington, DC; Nicholas J. Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Shande Tang, Hui Zhang, Christopher T. Cavanaugh, Ling Gao, and John S. Kauh, Eli Lilly, Bridgewater, NJ; and Richard A. Walgren, Eli Lilly, Indianapolis, IN
| | - Jeanny B Aragon-Ching
- Daniel P. Petrylak, Yale University Cancer Center, New Haven, CT; Scott T. Tagawa, Weill Cornell Medical College, New York; David Shaffer, New York Oncology Hematology, Albany, NY; Manish Kohli, Mayo Clinic, Rochester, MN; Andrea Eisen, Sunnybrook Health Sciences Centre; Srikala S. Sridhar, Princess Margaret Hospital, Toronto; Christina Canil, Ottawa Hospital Cancer Centre, Ottawa, Ontario; Kim N. Chi, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; Alexander Spira, John M. Burke, David Shaffer, and Nicholas J. Vogelzang, US Oncology Research, The Woodlands, TX; Evan Y. Yu, University of Washington Medical Center and Fred Hutchinson Cancer Research Center, Seattle, WA; John M. Burke, Rocky Mountain Cancer Centers, Aurora, CO; Chong-Xian Pan, University of California-Davis Medical Center, Sacramento; David I. Quinn, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Jenny J. Kim, Johns Hopkins University, Baltimore, MD; Jeanny B. Aragon-Ching, George Washington University Medical Center, Washington, DC; Nicholas J. Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Shande Tang, Hui Zhang, Christopher T. Cavanaugh, Ling Gao, and John S. Kauh, Eli Lilly, Bridgewater, NJ; and Richard A. Walgren, Eli Lilly, Indianapolis, IN
| | - David I Quinn
- Daniel P. Petrylak, Yale University Cancer Center, New Haven, CT; Scott T. Tagawa, Weill Cornell Medical College, New York; David Shaffer, New York Oncology Hematology, Albany, NY; Manish Kohli, Mayo Clinic, Rochester, MN; Andrea Eisen, Sunnybrook Health Sciences Centre; Srikala S. Sridhar, Princess Margaret Hospital, Toronto; Christina Canil, Ottawa Hospital Cancer Centre, Ottawa, Ontario; Kim N. Chi, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; Alexander Spira, John M. Burke, David Shaffer, and Nicholas J. Vogelzang, US Oncology Research, The Woodlands, TX; Evan Y. Yu, University of Washington Medical Center and Fred Hutchinson Cancer Research Center, Seattle, WA; John M. Burke, Rocky Mountain Cancer Centers, Aurora, CO; Chong-Xian Pan, University of California-Davis Medical Center, Sacramento; David I. Quinn, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Jenny J. Kim, Johns Hopkins University, Baltimore, MD; Jeanny B. Aragon-Ching, George Washington University Medical Center, Washington, DC; Nicholas J. Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Shande Tang, Hui Zhang, Christopher T. Cavanaugh, Ling Gao, and John S. Kauh, Eli Lilly, Bridgewater, NJ; and Richard A. Walgren, Eli Lilly, Indianapolis, IN
| | - Nicholas J Vogelzang
- Daniel P. Petrylak, Yale University Cancer Center, New Haven, CT; Scott T. Tagawa, Weill Cornell Medical College, New York; David Shaffer, New York Oncology Hematology, Albany, NY; Manish Kohli, Mayo Clinic, Rochester, MN; Andrea Eisen, Sunnybrook Health Sciences Centre; Srikala S. Sridhar, Princess Margaret Hospital, Toronto; Christina Canil, Ottawa Hospital Cancer Centre, Ottawa, Ontario; Kim N. Chi, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; Alexander Spira, John M. Burke, David Shaffer, and Nicholas J. Vogelzang, US Oncology Research, The Woodlands, TX; Evan Y. Yu, University of Washington Medical Center and Fred Hutchinson Cancer Research Center, Seattle, WA; John M. Burke, Rocky Mountain Cancer Centers, Aurora, CO; Chong-Xian Pan, University of California-Davis Medical Center, Sacramento; David I. Quinn, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Jenny J. Kim, Johns Hopkins University, Baltimore, MD; Jeanny B. Aragon-Ching, George Washington University Medical Center, Washington, DC; Nicholas J. Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Shande Tang, Hui Zhang, Christopher T. Cavanaugh, Ling Gao, and John S. Kauh, Eli Lilly, Bridgewater, NJ; and Richard A. Walgren, Eli Lilly, Indianapolis, IN
| | - Shande Tang
- Daniel P. Petrylak, Yale University Cancer Center, New Haven, CT; Scott T. Tagawa, Weill Cornell Medical College, New York; David Shaffer, New York Oncology Hematology, Albany, NY; Manish Kohli, Mayo Clinic, Rochester, MN; Andrea Eisen, Sunnybrook Health Sciences Centre; Srikala S. Sridhar, Princess Margaret Hospital, Toronto; Christina Canil, Ottawa Hospital Cancer Centre, Ottawa, Ontario; Kim N. Chi, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; Alexander Spira, John M. Burke, David Shaffer, and Nicholas J. Vogelzang, US Oncology Research, The Woodlands, TX; Evan Y. Yu, University of Washington Medical Center and Fred Hutchinson Cancer Research Center, Seattle, WA; John M. Burke, Rocky Mountain Cancer Centers, Aurora, CO; Chong-Xian Pan, University of California-Davis Medical Center, Sacramento; David I. Quinn, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Jenny J. Kim, Johns Hopkins University, Baltimore, MD; Jeanny B. Aragon-Ching, George Washington University Medical Center, Washington, DC; Nicholas J. Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Shande Tang, Hui Zhang, Christopher T. Cavanaugh, Ling Gao, and John S. Kauh, Eli Lilly, Bridgewater, NJ; and Richard A. Walgren, Eli Lilly, Indianapolis, IN
| | - Hui Zhang
- Daniel P. Petrylak, Yale University Cancer Center, New Haven, CT; Scott T. Tagawa, Weill Cornell Medical College, New York; David Shaffer, New York Oncology Hematology, Albany, NY; Manish Kohli, Mayo Clinic, Rochester, MN; Andrea Eisen, Sunnybrook Health Sciences Centre; Srikala S. Sridhar, Princess Margaret Hospital, Toronto; Christina Canil, Ottawa Hospital Cancer Centre, Ottawa, Ontario; Kim N. Chi, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; Alexander Spira, John M. Burke, David Shaffer, and Nicholas J. Vogelzang, US Oncology Research, The Woodlands, TX; Evan Y. Yu, University of Washington Medical Center and Fred Hutchinson Cancer Research Center, Seattle, WA; John M. Burke, Rocky Mountain Cancer Centers, Aurora, CO; Chong-Xian Pan, University of California-Davis Medical Center, Sacramento; David I. Quinn, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Jenny J. Kim, Johns Hopkins University, Baltimore, MD; Jeanny B. Aragon-Ching, George Washington University Medical Center, Washington, DC; Nicholas J. Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Shande Tang, Hui Zhang, Christopher T. Cavanaugh, Ling Gao, and John S. Kauh, Eli Lilly, Bridgewater, NJ; and Richard A. Walgren, Eli Lilly, Indianapolis, IN
| | - Christopher T Cavanaugh
- Daniel P. Petrylak, Yale University Cancer Center, New Haven, CT; Scott T. Tagawa, Weill Cornell Medical College, New York; David Shaffer, New York Oncology Hematology, Albany, NY; Manish Kohli, Mayo Clinic, Rochester, MN; Andrea Eisen, Sunnybrook Health Sciences Centre; Srikala S. Sridhar, Princess Margaret Hospital, Toronto; Christina Canil, Ottawa Hospital Cancer Centre, Ottawa, Ontario; Kim N. Chi, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; Alexander Spira, John M. Burke, David Shaffer, and Nicholas J. Vogelzang, US Oncology Research, The Woodlands, TX; Evan Y. Yu, University of Washington Medical Center and Fred Hutchinson Cancer Research Center, Seattle, WA; John M. Burke, Rocky Mountain Cancer Centers, Aurora, CO; Chong-Xian Pan, University of California-Davis Medical Center, Sacramento; David I. Quinn, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Jenny J. Kim, Johns Hopkins University, Baltimore, MD; Jeanny B. Aragon-Ching, George Washington University Medical Center, Washington, DC; Nicholas J. Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Shande Tang, Hui Zhang, Christopher T. Cavanaugh, Ling Gao, and John S. Kauh, Eli Lilly, Bridgewater, NJ; and Richard A. Walgren, Eli Lilly, Indianapolis, IN
| | - Ling Gao
- Daniel P. Petrylak, Yale University Cancer Center, New Haven, CT; Scott T. Tagawa, Weill Cornell Medical College, New York; David Shaffer, New York Oncology Hematology, Albany, NY; Manish Kohli, Mayo Clinic, Rochester, MN; Andrea Eisen, Sunnybrook Health Sciences Centre; Srikala S. Sridhar, Princess Margaret Hospital, Toronto; Christina Canil, Ottawa Hospital Cancer Centre, Ottawa, Ontario; Kim N. Chi, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; Alexander Spira, John M. Burke, David Shaffer, and Nicholas J. Vogelzang, US Oncology Research, The Woodlands, TX; Evan Y. Yu, University of Washington Medical Center and Fred Hutchinson Cancer Research Center, Seattle, WA; John M. Burke, Rocky Mountain Cancer Centers, Aurora, CO; Chong-Xian Pan, University of California-Davis Medical Center, Sacramento; David I. Quinn, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Jenny J. Kim, Johns Hopkins University, Baltimore, MD; Jeanny B. Aragon-Ching, George Washington University Medical Center, Washington, DC; Nicholas J. Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Shande Tang, Hui Zhang, Christopher T. Cavanaugh, Ling Gao, and John S. Kauh, Eli Lilly, Bridgewater, NJ; and Richard A. Walgren, Eli Lilly, Indianapolis, IN
| | - John S Kauh
- Daniel P. Petrylak, Yale University Cancer Center, New Haven, CT; Scott T. Tagawa, Weill Cornell Medical College, New York; David Shaffer, New York Oncology Hematology, Albany, NY; Manish Kohli, Mayo Clinic, Rochester, MN; Andrea Eisen, Sunnybrook Health Sciences Centre; Srikala S. Sridhar, Princess Margaret Hospital, Toronto; Christina Canil, Ottawa Hospital Cancer Centre, Ottawa, Ontario; Kim N. Chi, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; Alexander Spira, John M. Burke, David Shaffer, and Nicholas J. Vogelzang, US Oncology Research, The Woodlands, TX; Evan Y. Yu, University of Washington Medical Center and Fred Hutchinson Cancer Research Center, Seattle, WA; John M. Burke, Rocky Mountain Cancer Centers, Aurora, CO; Chong-Xian Pan, University of California-Davis Medical Center, Sacramento; David I. Quinn, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Jenny J. Kim, Johns Hopkins University, Baltimore, MD; Jeanny B. Aragon-Ching, George Washington University Medical Center, Washington, DC; Nicholas J. Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Shande Tang, Hui Zhang, Christopher T. Cavanaugh, Ling Gao, and John S. Kauh, Eli Lilly, Bridgewater, NJ; and Richard A. Walgren, Eli Lilly, Indianapolis, IN
| | - Richard A Walgren
- Daniel P. Petrylak, Yale University Cancer Center, New Haven, CT; Scott T. Tagawa, Weill Cornell Medical College, New York; David Shaffer, New York Oncology Hematology, Albany, NY; Manish Kohli, Mayo Clinic, Rochester, MN; Andrea Eisen, Sunnybrook Health Sciences Centre; Srikala S. Sridhar, Princess Margaret Hospital, Toronto; Christina Canil, Ottawa Hospital Cancer Centre, Ottawa, Ontario; Kim N. Chi, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; Alexander Spira, John M. Burke, David Shaffer, and Nicholas J. Vogelzang, US Oncology Research, The Woodlands, TX; Evan Y. Yu, University of Washington Medical Center and Fred Hutchinson Cancer Research Center, Seattle, WA; John M. Burke, Rocky Mountain Cancer Centers, Aurora, CO; Chong-Xian Pan, University of California-Davis Medical Center, Sacramento; David I. Quinn, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Jenny J. Kim, Johns Hopkins University, Baltimore, MD; Jeanny B. Aragon-Ching, George Washington University Medical Center, Washington, DC; Nicholas J. Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Shande Tang, Hui Zhang, Christopher T. Cavanaugh, Ling Gao, and John S. Kauh, Eli Lilly, Bridgewater, NJ; and Richard A. Walgren, Eli Lilly, Indianapolis, IN
| | - Kim N Chi
- Daniel P. Petrylak, Yale University Cancer Center, New Haven, CT; Scott T. Tagawa, Weill Cornell Medical College, New York; David Shaffer, New York Oncology Hematology, Albany, NY; Manish Kohli, Mayo Clinic, Rochester, MN; Andrea Eisen, Sunnybrook Health Sciences Centre; Srikala S. Sridhar, Princess Margaret Hospital, Toronto; Christina Canil, Ottawa Hospital Cancer Centre, Ottawa, Ontario; Kim N. Chi, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; Alexander Spira, John M. Burke, David Shaffer, and Nicholas J. Vogelzang, US Oncology Research, The Woodlands, TX; Evan Y. Yu, University of Washington Medical Center and Fred Hutchinson Cancer Research Center, Seattle, WA; John M. Burke, Rocky Mountain Cancer Centers, Aurora, CO; Chong-Xian Pan, University of California-Davis Medical Center, Sacramento; David I. Quinn, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA; Jenny J. Kim, Johns Hopkins University, Baltimore, MD; Jeanny B. Aragon-Ching, George Washington University Medical Center, Washington, DC; Nicholas J. Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Shande Tang, Hui Zhang, Christopher T. Cavanaugh, Ling Gao, and John S. Kauh, Eli Lilly, Bridgewater, NJ; and Richard A. Walgren, Eli Lilly, Indianapolis, IN
| |
Collapse
|
33
|
Preliminary Analysis of the Expression of Selected Proangiogenic and Antioxidant Genes and MicroRNAs in Patients with Non-Muscle-Invasive Bladder Cancer. J Clin Med 2016; 5:jcm5030029. [PMID: 26927195 PMCID: PMC4810100 DOI: 10.3390/jcm5030029] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 01/25/2016] [Accepted: 02/14/2016] [Indexed: 12/11/2022] Open
Abstract
Heme oxygenase-1 (HO-1) is an enzyme contributing to the development and progression of different cancer types. HO-1 plays a role in pathological angiogenesis in bladder cancer and contributes to the resistance of this cancer to therapy. It also regulates the expression of microRNAs in rhabdomyosarcoma and non-small cell lung cancer. The expression of HO-1 may be regulated by hypoxia inducible factors (HIFs) and Nrf2 transcription factor. The expression of HO-1 has not so far been examined in relation to Nrf2, HIF-1α, and potential mediators of angiogenesis in human bladder cancer. We measured the concentration of proinflammatory and proangiogenic cytokines and the expression of cytoprotective and proangiogenic mRNAs and miRNAs in healthy subjects and patients with bladder cancer. HO-1 expression was upregulated together with HIF-1α, HIF-2α, and Nrf2 in bladder cancer in comparison to healthy tissue. VEGF was elevated both at mRNA and protein level in the tumor and in sera, respectively. Additionally, IL-6 and IL-8 were increased in sera of patients affected with urothelial bladder cancer. Moreover, miR-155 was downregulated whereas miR-200c was elevated in cancer biopsies in comparison to healthy tissue. The results indicate that the increased expression of HO-1 in bladder cancer is paralleled by changes in the expression of other potentially interacting genes, like Nrf2, HIF-1α, HIF-2α, IL-6, IL-8, and VEGF. Further studies are necessary to also elucidate the potential links with miR-155 and miR-200c.
Collapse
|
34
|
Li Y, Lu J, Zhou S, Wang W, Tan G, Zhang Z, Dong Z, Kang T, Tang F. Clusterin induced by N,N'-Dinitrosopiperazine is involved in nasopharyngeal carcinoma metastasis. Oncotarget 2016; 7:5548-5563. [PMID: 26716898 PMCID: PMC4868705 DOI: 10.18632/oncotarget.6750] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 12/07/2015] [Indexed: 12/18/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) has a high metastatic clinicopathological feature. As a carcinogen factor, N,N'-Dinitrosopiperazine (DNP) is involved in NPC metastasis, but its precise mechanism has not been fully elucidated. Herein, we showed that DNP promotes NPC metastasis through up-regulating anterior clusterin (CLU). DNP was found to increase CLU, matrix metalloproteinases (MMP) 9 and vascular endothelial growth factor (VEGF) expression and activity, further DNP-increased MMP-9 and VEGF expression was through up-regulating CLU. We also found that DNP increased the binding of CLU with MMP-9 or VEGF. DNP induced the motility and invasion of NPC cell, which was inhibited by siRNA-CLU. The clinical investigation showed that CLU, MMP-9 and VEGF were positively correlated with the tumor-node -metastasis (TNM) classification. These results indicate that DNP may promote NPC tumor metastasis through up-regulating CLU, MMP-9 and VEGF expression. Therefore, DNP-increased CLU expression may be an important factor of NPC-high metastasis, and CLU may serve as a biomarker for NPC metastasis.
Collapse
Affiliation(s)
- Yuejin Li
- State Key Laboratory of Oncology in South China and Department of Experimental Research, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong, China
- Clinical Laboratory and Medical Research Center, Zhuhai Hospital, Jinan University, Zhuhai People's Hospital, Zhuhai 519000, Guangdong, China
| | - Jinping Lu
- Clinical Laboratory and Medical Research Center, Zhuhai Hospital, Jinan University, Zhuhai People's Hospital, Zhuhai 519000, Guangdong, China
| | - Shan Zhou
- Clinical Laboratory and Medical Research Center, Zhuhai Hospital, Jinan University, Zhuhai People's Hospital, Zhuhai 519000, Guangdong, China
| | - Weiwei Wang
- Clinical Laboratory and Medical Research Center, Zhuhai Hospital, Jinan University, Zhuhai People's Hospital, Zhuhai 519000, Guangdong, China
| | - Gongjun Tan
- Clinical Laboratory and Medical Research Center, Zhuhai Hospital, Jinan University, Zhuhai People's Hospital, Zhuhai 519000, Guangdong, China
| | - Zhenlin Zhang
- Clinical Laboratory and Medical Research Center, Zhuhai Hospital, Jinan University, Zhuhai People's Hospital, Zhuhai 519000, Guangdong, China
| | - Zigang Dong
- Hormel Institute, University of Minnesota, Austin, Minnesota 55912, USA
| | - Tiebang Kang
- State Key Laboratory of Oncology in South China and Department of Experimental Research, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong, China
| | - Faqing Tang
- Clinical Laboratory and Medical Research Center, Zhuhai Hospital, Jinan University, Zhuhai People's Hospital, Zhuhai 519000, Guangdong, China
| |
Collapse
|