1
|
Jackson-Spence F, Ackerman C, Jones R, Toms C, Jovaisaite A, Young M, Hussain S, Protheroe A, Birtle A, Chakraborti P, Huddart R, Jagdev S, Bahl A, Sundar S, Crabb S, Powles T, Szabados B. Biomarkers associated with survival in patients with platinum-refractory urothelial carcinoma treated with paclitaxel. Urol Oncol 2024; 42:372.e1-372.e10. [PMID: 39025719 DOI: 10.1016/j.urolonc.2024.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 05/18/2024] [Accepted: 05/19/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND Taxane- based chemotherapy is widely used in patients with platinum- and immunotherapy refractory, metastatic urothelial carcinoma (mUC). Outcomes are poor and biomarkers associated with outcome are lacking. We aim to identify cancer hallmarks associated with survival in patients receiving paclitaxel. METHODS Whole-transcriptome profiles were generated for a subset of patients enrolled in a randomised phase II study investigating paclitaxel and pazopanib in platinum refractory mUC (PLUTO, EudraCT 2011-001841-34). Estimates of gene expression were calculated and input into the Almac proprietary analysis pipeline and signature scores were calculated using ClaraT V3.0.0. Ten key gene signatures were assessed: Immuno-Oncology, Epithelial to Mesenchymal Transition, Angiogenesis, Proliferation, Cell Death, Genome Instability, Energetics, Inflammation, Immortality and Evading Growth. Hazard ratios were calculated using Cox regression model and Kaplan-Meier methods were used to estimate progression free survival (PFS) and overall survival (OS). RESULTS 38 and 45 patients treated with paclitaxel or pazopanib were included. Patients with high genome instability expression treated with paclitaxel had significantly improved survival with a HR of 0.29 (95% CI: 0.14-0.61, p=0.001) and HR 0.34 (95% CI: 0.17-0.69, p=0.003) for PFS and OS, respectively. Similarly, patients with high evading growth suppressor expression treated with paclitaxel had improved PFS and OS with a HR of 0.35 (95% CI: 0.19-0.77, p=0.007) and HR 0.46 (95% CI: 0.23-0.91, p=0.026), respectively. No other gene signatures had significant impact on outcome. In both paclitaxel and pazopanib cohorts, angiogenesis activation was associated with worse PFS and OS, and VEGF targeted therapy did not improve outcomes. CONCLUSION High Genome-instability and Evading-growth suppressor biologies are associated with improved survival in patients with platinum refractory mUC receiving paclitaxel. These may refine mUC risk stratification and guide treatment decision in the future.
Collapse
Affiliation(s)
| | - Charlotte Ackerman
- Department of Genitourinary Oncology, Barts Cancer Institute, QMUL, London, UK
| | - Robert Jones
- Department of Genitourinary Oncology, University of Glasgow, Glasgow, Scotland, UK
| | - Charlotte Toms
- Department of Genitourinary Oncology, Barts Cancer Institute, QMUL, London, UK
| | - Agne Jovaisaite
- Department of Genitourinary Oncology, Barts Cancer Institute, QMUL, London, UK
| | - Matthew Young
- Department of Genitourinary Oncology, Barts Cancer Institute, QMUL, London, UK
| | - Syed Hussain
- Department of Genitourinary Oncology, University of Liverpool, Liverpool, UK
| | - Andrew Protheroe
- Department of Genitourinary Oncology, Churchill Hospital, Oxford, UK
| | - Alison Birtle
- Department of Genitourinary Oncology, Preston Hospital, Preston, UK
| | - Prabir Chakraborti
- Department of Genitourinary Oncology, Derby Hospitals NHS Foundation trust, Derby, UK
| | - Robert Huddart
- Department of Genitourinary Oncology, Institute of Cancer Research, Sutton, UK
| | - Santinder Jagdev
- Department of Genitourinary Oncology, St James's University Hospital, Leeds, UK
| | - Amit Bahl
- Department of Genitourinary Oncology, Bristol Haematology and Oncology Centre, Bristol, UK
| | - Santhanam Sundar
- Department of Genitourinary Oncology, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Simon Crabb
- Department of Genitourinary Oncology, University of Southampton, Southampton UK
| | - Thomas Powles
- Department of Genitourinary Oncology, Barts Cancer Institute, QMUL, London, UK.
| | - Bernadett Szabados
- Department of Genitourinary Oncology, Barts Cancer Institute, QMUL, London, UK
| |
Collapse
|
2
|
Papadopoulou K, Koliou GA, Tsimiliotis D, Kotoula V, Foukas P, Goussia A, Tsiatas M, Visvikis A, Chatzopoulos K, Nifora M, Charchanti A, Koumarianou A, Christodoulou C, Pectasides D, Psyrri A, Fostira F, Fountzilas G, Samantas E. Investigation of prognostic biomarkers in patients with urothelial carcinoma treated with platinum-based regimens. Urol Oncol 2022; 40:538.e15-538.e24. [PMID: 36041976 DOI: 10.1016/j.urolonc.2022.07.007] [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: 05/11/2022] [Revised: 07/08/2022] [Accepted: 07/17/2022] [Indexed: 01/13/2023]
Abstract
BACKGROUND Bladder cancer (BC) is a heterogeneous malignancy with dismal outcome. PATIENTS AND METHODS Mutations in genes, altered or linked to platinum sensitivity in BC, were examined in 66 patients' tumors along with tumor infiltrating lymphocytes (TILs) density and MMR, PD-L1 and CD8 protein expression, as well as basal and luminal subtypes, defined by protein expression of markers, including CK5/6 and GATA3 or CK20, respectively. RESULTS 41 tumors harbored mutations, mainly in TP53 (38%), ARID1A (17%) and the DNA damage response and repair (DDR) genes ERCC2 (17%) and BRCA2 (15%). Mutations in other DDR relevant genes were also present. Age showed unfavorable prognosis for overall survival (HR=1.07, P = 0.026); no benefit was seen for patients with TP53, ARID1A, ERCC2 or BRCA2 mutations or mutations in 1 or more DDR genes. PD-L1 status positively correlated with stromal (rho=0.46, P < 0.001) and intratumoral (rho=0.53, P < 0.001) CD8 expression or TILs (rho=0.29, P = 0.018); none associated with overall survival (OS). A statistically significant difference was observed between PD-L1 status and immunohistochemistry (IHC)‑based subtypes, with tumors classified as luminal (GATA3+ and/or CK20+ and CK5/6-) showing lower PD-L1 expression relative to basal (CK5/6+ and GATA3- and/or CK20-) (median value 0 vs. 2.5, P = 0.029). Concerning OS, no statistically significant difference was seen among patients with basal or luminal tumors. CONCLUSION No association was seen herein between DDR mutations, TILs, PD-L1, CD8 expression or IHC-based subtypes and patient survival; these observations warrant validation within a larger cohort.
Collapse
Affiliation(s)
- Kyriaki Papadopoulou
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research/Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | | | | | - Vassiliki Kotoula
- Department of Pathology, Aristotle University of Thessaloniki, School of Health Sciences, Faculty of Medicine, Thessaloniki, Greece
| | - Periklis Foukas
- Second Department of Pathology, National and Kapodistrian University of Athens, School of Medicine, Attikon University Hospital, Haidari, Greece
| | - Anna Goussia
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Marinos Tsiatas
- Department of Oncology, Athens Medical Center, Marousi, Greece
| | - Anastasios Visvikis
- Third Department of Medical Oncology, Agii Anargiri Cancer Hospital, Athens, Greece
| | - Kyriakos Chatzopoulos
- Department of Pathology, Aristotle University of Thessaloniki, School of Health Sciences, Faculty of Medicine, Thessaloniki, Greece
| | - Martha Nifora
- Second Department of Pathology, National and Kapodistrian University of Athens, School of Medicine, Attikon University Hospital, Haidari, Greece
| | - Antonia Charchanti
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Anna Koumarianou
- Hematology Oncology Unit, Fourth Department of Internal Medicine, Attikon University Hospital, Athens, Greece
| | | | - Dimitrios Pectasides
- Oncology Section, Second Department of Internal Medicine, Hippokration Hospital, Section of Medical Oncology, Athens, Greece
| | - Amanda Psyrri
- Attikon University Hospital, Faculty of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Florentia Fostira
- Molecular Diagnostics Laboratory, InRASTES, National Centre for Scientific Research Demokritos, Athens, Greece
| | - George Fountzilas
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research/Aristotle University of Thessaloniki, Thessaloniki, Greece; Aristotle University of Thessaloniki, Thessaloniki, Greece; Department of Medical Oncology, German Oncology Center, Limassol, Cyprus
| | - Epaminontas Samantas
- Third Department of Medical Oncology, Agii Anargiri Cancer Hospital, Athens, Greece
| |
Collapse
|
3
|
HUS1 as a Potential Therapeutic Target in Urothelial Cancer. J Clin Med 2022; 11:jcm11082208. [PMID: 35456300 PMCID: PMC9031773 DOI: 10.3390/jcm11082208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 04/09/2022] [Accepted: 04/12/2022] [Indexed: 12/04/2022] Open
Abstract
Platinum-based chemotherapy is the standard of care with concern to first-line systemic therapy for metastatic disease in urothelial cancer (UC). Resistance to chemotherapy despite an initial response is linked with the ability to remove platinum-based DNA adducts and to repair chemotherapy-induced DNA lesions by various DNA repair proteins. The Rad9-Rad1-HUS1 complex that is loaded onto DNA at sites of damage is involved in checkpoint activation as well as DNA repair. Here, we addressed for the first time the potential influence of HUS1 expression in urothelial carcinogenesis (using two human basal urothelial cancer cell lines UM-UC-3 and HT1197) and its role as a potential therapeutic target for predicting responses to platinum-based chemotherapy. Specific inhibition of HUS1 expression in both cell lines was achieved by specific siRNA and validated by Western blot. In order to define the possible importance of HUS1 in the regulation of cellular proliferation, parental and resistant cells were treated with increasing concentrations of either control or HUS1 siRNA. HUS1 protein expression was observed in both human basal urothelial cancer cell lines UM-UC-3 and HT1197. In cisplatin-sensitive cells, knock-down of HUS1 inhibited cellular proliferation in the presence of cisplatin. On the contrary, knock-down of HUS1 in resistant cells did not result in a re-sensitization to cisplatin. Finally, RNAseq data from the Cancer Genome Atlas provided evidence that HUS1 expression is a significant prognostic factor for poor survival in UC patients. In summary, HUS1 may acts as an oncogene in UC and might be a key determinant of the cellular response to cisplatin-based chemotherapy.
Collapse
|
4
|
Liu S, Chen X, Lin T. Emerging strategies for the improvement of chemotherapy in bladder cancer: Current knowledge and future perspectives. J Adv Res 2021; 39:187-202. [PMID: 35777908 PMCID: PMC9263750 DOI: 10.1016/j.jare.2021.11.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/01/2021] [Accepted: 11/19/2021] [Indexed: 11/24/2022] Open
Abstract
The response of chemotherapy and prognosis in bladder cancer is unsatisfied. Immunotherapy, targeted therapy, and ADC improve the efficacy of chemotherapy. Emerging targets in cancer cells and TME spawned novel preclinical agents. Novel drug delivery, such as nanotechnology, enhances effects of chemotherapeutics. The organoid and PDX model are promising to screen and evaluate the target therapy.
Background Chemotherapy is a first-line treatment for advanced and metastatic bladder cancer, but the unsatisfactory objective response rate to this treatment yields poor 5-year patient survival. Only PD-1/PD-L1-based immune checkpoint inhibitors, FGFR3 inhibitors and antibody-drug conjugates are approved by the FDA to be used in bladder cancer, mainly for platinum-refractory or platinum-ineligible locally advanced or metastatic urothelial carcinoma. Emerging studies indicate that the combination of targeted therapy and chemotherapy shows better efficacy than targeted therapy or chemotherapy alone. Newly identified targets in cancer cells and various functions of the tumour microenvironment have spawned novel agents and regimens, which give impetus to sensitizing chemotherapy in the bladder cancer setting. Aim of Review This review aims to present the current evidence for potentiating the efficacy of chemotherapy in bladder cancer. We focus on combining chemotherapy with other treatments as follows: targeted therapy, including immunotherapy and antibody-drug conjugates in clinic; novel targeted drugs and nanoparticles in preclinical models and potential targets that may contribute to chemosensitivity in future clinical practice. The prospect of precision therapy is also discussed in bladder cancer. Key Scientific Concepts of Review Combining chemotherapy drugs with immune checkpoint inhibitors, antibody-drug conjugates and VEGF inhibitors potentially elevates the response rate and survival. Novel targets, including cancer stem cells, DNA damage repair, antiapoptosis, drug metabolism and the tumour microenvironment, contribute to chemosensitization. Gene alteration-based drug selection and patient-derived xenograft- and organoid-based drug validation are the future for precision therapy.
Collapse
|
5
|
Zheng X, Wezel F, Azoitei A, Meessen S, Wang W, Najjar G, Wang X, Kraus JM, Kestler HA, John A, Zengerling F, Bolenz C, Günes C. Shorter Leukocyte Telomere Length Is Associated with Worse Survival of Patients with Bladder Cancer and Renal Cell Carcinoma. Cancers (Basel) 2021; 13:3774. [PMID: 34359672 PMCID: PMC8345040 DOI: 10.3390/cancers13153774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/19/2021] [Accepted: 07/22/2021] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Telomeres are protein-DNA complexes at the tips of linear chromosomes. They protect the DNA from end-to-end fusion and exonucleolytic degradation. Shortening of telomeric DNA during aging can generate dysfunctional telomeres, promoting tumorigenesis. More recent data indicate that both short and long telomeres of peripheral blood leukocyte (PBL) cells can serve as prognostic biomarkers for cancer risk and may be associated with survival of patients with solid cancers. Telomere length in PBL cells could also be a potential prognostic biomarker for survival in bladder cancer (BC) or renal cell carcinoma (RCC). METHODS The relative telomere length (RTL) of PBL cells was assessed in patients with BC (n = 144) and RCC (n = 144) by using qPCR. A control population of patients without malignant disease (NC, n = 73) was included for comparison. The correlation and association of RTL with histopathological parameters and overall survival (OS) were evaluated. RESULTS Patients with BC and RCC had significantly shorter telomeres compared to patients without malignant disease. Within the cancer cohorts, multivariate analysis revealed that short RTL is an independent predictor of worse survival in BC (p = 0.039) and RCC (p = 0.041). CONCLUSION Patients with BC and RCC had significantly shorter telomeres compared to the normal population. Shorter RTL in BC and RCC was an independent predictor of reduced survival.
Collapse
Affiliation(s)
- Xi Zheng
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (X.Z.); (F.W.); (A.A.); (S.M.); (W.W.); (G.N.); (X.W.); (A.J.); (F.Z.); (C.B.)
| | - Felix Wezel
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (X.Z.); (F.W.); (A.A.); (S.M.); (W.W.); (G.N.); (X.W.); (A.J.); (F.Z.); (C.B.)
| | - Anca Azoitei
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (X.Z.); (F.W.); (A.A.); (S.M.); (W.W.); (G.N.); (X.W.); (A.J.); (F.Z.); (C.B.)
| | - Sabine Meessen
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (X.Z.); (F.W.); (A.A.); (S.M.); (W.W.); (G.N.); (X.W.); (A.J.); (F.Z.); (C.B.)
| | - Wenya Wang
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (X.Z.); (F.W.); (A.A.); (S.M.); (W.W.); (G.N.); (X.W.); (A.J.); (F.Z.); (C.B.)
| | - Gregoire Najjar
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (X.Z.); (F.W.); (A.A.); (S.M.); (W.W.); (G.N.); (X.W.); (A.J.); (F.Z.); (C.B.)
| | - Xue Wang
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (X.Z.); (F.W.); (A.A.); (S.M.); (W.W.); (G.N.); (X.W.); (A.J.); (F.Z.); (C.B.)
| | - Johann M. Kraus
- Institute of Medical Systems Biology, Ulm University, 89081 Ulm, Germany; (J.M.K.); (H.A.K.)
| | - Hans A. Kestler
- Institute of Medical Systems Biology, Ulm University, 89081 Ulm, Germany; (J.M.K.); (H.A.K.)
| | - Axel John
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (X.Z.); (F.W.); (A.A.); (S.M.); (W.W.); (G.N.); (X.W.); (A.J.); (F.Z.); (C.B.)
| | - Friedemann Zengerling
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (X.Z.); (F.W.); (A.A.); (S.M.); (W.W.); (G.N.); (X.W.); (A.J.); (F.Z.); (C.B.)
| | - Christian Bolenz
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (X.Z.); (F.W.); (A.A.); (S.M.); (W.W.); (G.N.); (X.W.); (A.J.); (F.Z.); (C.B.)
| | - Cagatay Günes
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (X.Z.); (F.W.); (A.A.); (S.M.); (W.W.); (G.N.); (X.W.); (A.J.); (F.Z.); (C.B.)
| |
Collapse
|
6
|
Morlacco A, Modonutti D, Motterle G, Martino F, Dal Moro F, Novara G. Nomograms in Urologic Oncology: Lights and Shadows. J Clin Med 2021; 10:jcm10050980. [PMID: 33801184 PMCID: PMC7957873 DOI: 10.3390/jcm10050980] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/08/2021] [Accepted: 02/20/2021] [Indexed: 12/29/2022] Open
Abstract
Decision-making in urologic oncology involves integrating multiple clinical data to provide an answer to the needs of a single patient. Although the practice of medicine has always been an “art” involving experience, clinical data, scientific evidence and judgment, the creation of specialties and subspecialties has multiplied the challenges faced every day by physicians. In the last decades, with the field of urologic oncology becoming more and more complex, there has been a rise in tools capable of compounding several pieces of information and supporting clinical judgment and experience when approaching a difficult decision. The vast majority of these tools provide a risk of a certain event based on various information integrated in a mathematical model. Specifically, most decision-making tools in the field of urologic focus on the preoperative or postoperative phase and provide a prognostic or predictive risk assessment based on the available clinical and pathological data. More recently, imaging and genomic features started to be incorporated in these models in order to improve their accuracy. Genomic classifiers, look-up tables, regression trees, risk-stratification tools and nomograms are all examples of this effort. Nomograms are by far the most frequently used in clinical practice, but are also among the most controversial of these tools. This critical, narrative review will focus on the use, diffusion and limitations of nomograms in the field of urologic oncology.
Collapse
Affiliation(s)
- Alessandro Morlacco
- Urology Unit, Department of Surgical, Oncological and Gastroenterological Sciences, University of Padua, 35128 Padua, Italy; (A.M.); (D.M.); (G.M.); (F.D.M.)
| | - Daniele Modonutti
- Urology Unit, Department of Surgical, Oncological and Gastroenterological Sciences, University of Padua, 35128 Padua, Italy; (A.M.); (D.M.); (G.M.); (F.D.M.)
| | - Giovanni Motterle
- Urology Unit, Department of Surgical, Oncological and Gastroenterological Sciences, University of Padua, 35128 Padua, Italy; (A.M.); (D.M.); (G.M.); (F.D.M.)
| | - Francesca Martino
- Department of Nephrology, Dialysis and Kidney Transplant, International Renal Research Institute, San Bortolo Hospital, 36100 Vicenza, Italy;
| | - Fabrizio Dal Moro
- Urology Unit, Department of Surgical, Oncological and Gastroenterological Sciences, University of Padua, 35128 Padua, Italy; (A.M.); (D.M.); (G.M.); (F.D.M.)
| | - Giacomo Novara
- Urology Unit, Department of Surgical, Oncological and Gastroenterological Sciences, University of Padua, 35128 Padua, Italy; (A.M.); (D.M.); (G.M.); (F.D.M.)
- Correspondence: or ; Tel.: +39-049-821-1250; Fax: +39-049-821-8757
| |
Collapse
|
7
|
Albisinni S, Aoun F, Diamand R, Mjaess G, Esperto F, Martinez Chanza N, Roumeguère T, De Nunzio C. Systematic review of neoadjuvant therapy by immune checkpoint inhibitors before radical cystectomy: where do we stand? MINERVA UROL NEFROL 2020; 72:663-672. [DOI: 10.23736/s0393-2249.20.03833-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
8
|
Abstract
Muscle-invasive bladder cancer (MIBC), a highly heterogeneous disease, shows genomic instability and a high mutation rate. Clinical outcomes are variable and responses to conventional chemotherapy differ among patients (due to inter-patient tumor heterogeneity and inter-tumor heterogeneity) and even within each individual tumor (intra-tumor heterogeneity). Emerging evidence indicates that tumor heterogeneity may play an important role in cancer progression, resistance to therapy, and metastasis. Comprehensive molecular subtyping classifies MIBC into distinct categories that have potential to guide prognosis, patient stratification, and treatment. Genomic characterization of time-series analyses at the single cell level, and of cell-free circulating tumor DNA or circulating tumor cells, are emerging technologies that enable dissection of the complex clonal architecture of MIBC. This review provides insight into the clinical significance of the molecular mechanisms underlying heterogeneity, focusing on inter- and intra-tumor heterogeneity, with special emphasis on molecular classification and methods used to analyze the complex patterns involved.
Collapse
Affiliation(s)
- Ho Won Kang
- Department of Urology, School of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Korea.,Department of Urology, Chungbuk National University Hospital, Cheongju, Korea
| | - Wun-Jae Kim
- Department of Urology, School of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Korea.,Department of Urology, Chungbuk National University Hospital, Cheongju, Korea
| | - Woonyoung Choi
- Johns Hopkins Greenberg Bladder Cancer Institute and Brady Urological Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Seok Joong Yun
- Department of Urology, School of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Korea.,Department of Urology, Chungbuk National University Hospital, Cheongju, Korea
| |
Collapse
|
9
|
Zucali PA, Cordua N, D'Antonio F, Borea F, Perrino M, De Vincenzo F, Santoro A. Current Perspectives on Immunotherapy in the Peri-Operative Setting of Muscle-Infiltrating Bladder Cancer. Front Oncol 2020; 10:568279. [PMID: 33194654 PMCID: PMC7609911 DOI: 10.3389/fonc.2020.568279] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 09/15/2020] [Indexed: 12/22/2022] Open
Abstract
Patients with muscle-infiltrating bladder cancer (MIBC) present a high risk of postoperative recurrence and death from metastatic urothelial cancer despite surgical resection. Before the use of peri-operative chemotherapy, about half (52%) of patients undergoing radical cystectomy had had a relapse of tumor disease within 5 years of surgery. However, when peri-operative cisplatin-based chemotherapy is added to radical cystectomy for patients with MIBC it provides limited benefit in terms of survival, disease recurrence and development of metastases, at the expense of toxic effects. In fact, a significant proportion of patients still recurs and die to metastatic disease. Given the success of immune-oncological drugs in metastatic urothelial cancer, several trials started to test them in patients with non-metastatic MIBC either in neo-adjuvant and adjuvant setting. The preliminary results of these studies in neo-adjuvant setting are showing great promise, confirming the potential benefits of immunotherapy also in patients with non-metastatic MIBC. The aim of this review is to present an overview of developments happening on the introduction of immunotherapy in peri-operative setting in non-metastatic urothelial cancer. Moreover, an analysis of the critical issues regarding how best customize the delivery of immunotherapy to optimize efficacy and minimize the adverse effects, with particular focus on potential prognostic and predictive molecular biomarkers, is done.
Collapse
Affiliation(s)
- Paolo Andrea Zucali
- Department of Medical Oncology and Hematology, Humanitas Clinical and Research Center-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Nadia Cordua
- Department of Medical Oncology and Hematology, Humanitas Clinical and Research Center-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rozzano, Italy
| | - Federica D'Antonio
- Department of Medical Oncology and Hematology, Humanitas Clinical and Research Center-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rozzano, Italy
| | - Federica Borea
- Department of Medical Oncology and Hematology, Humanitas Clinical and Research Center-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rozzano, Italy
| | - Matteo Perrino
- Department of Medical Oncology and Hematology, Humanitas Clinical and Research Center-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rozzano, Italy
| | - Fabio De Vincenzo
- Department of Medical Oncology and Hematology, Humanitas Clinical and Research Center-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rozzano, Italy
| | - Armando Santoro
- Department of Medical Oncology and Hematology, Humanitas Clinical and Research Center-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| |
Collapse
|
10
|
Doxorubicin metabolism moderately attributes to putative toxicity in prodigiosin/doxorubicin synergism in vitro cells. Mol Cell Biochem 2020; 475:119-126. [PMID: 32754875 PMCID: PMC7599147 DOI: 10.1007/s11010-020-03864-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 07/28/2020] [Indexed: 02/05/2023]
Abstract
Doxorubicin (Dox) is a widely neoplasm chemotherapeutic drug with high incidences of cardiotoxicity. Prodigiosin (PG), a red bacterial pigment from Serratia marcescens, has been demonstrated to potentiate Dox’s cytotoxicity against oral squamous cell carcinoma cells through elevating Dox influx and identified as a Dox enhancer via PG-induced autophagy; however, toxicity of normal cell remains unclear. This study is conducted to evaluate putative cytotoxicity features of PG/Dox synergism in the liver, kidney, and heart cells and further elucidate whether PG augmented Dox’s effect via modulating Dox metabolism in normal cells. Murine hepatocytes FL83B, cardio-myoblast h9c2, and human kidney epithelial cells HK-2 were sequentially treated with PG and Dox by measuring cell viability, cell death characteristics, oxidative stress, Dox flux, and Dox metabolism. PG could slightly significant increase Dox cytotoxicity in all tested normal cells whose toxic alteration was less than that of oral squamous carcinoma cells. The augmentation of Dox cytotoxicity might be attributed to the increase of Dox-mediated ROS accumulation that might cause slight reduction of Dox influx and reduction of Dox metabolism. It was noteworthy to notice that sustained cytotoxicity appeared in normal cells after PG and Dox were removed. Taken together, moderately metabolic reduction of Dox might be ascribed to the mechanism of increase Dox cytotoxicity in PG-induced normal cells; nevertheless, the determination of PG/Dox dose with sustained cytotoxicity in normal cells needs to be comprehensively considered.
Collapse
|
11
|
Pichler R, Lindner AK, Compérat E, Obrist P, Schäfer G, Todenhöfer T, Horninger W, Culig Z, Untergasser G. Amplification of 7p12 Is Associated with Pathologic Nonresponse to Neoadjuvant Chemotherapy in Muscle-Invasive Bladder Cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 190:442-452. [PMID: 31843500 DOI: 10.1016/j.ajpath.2019.10.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 09/12/2019] [Accepted: 10/08/2019] [Indexed: 12/25/2022]
Abstract
Pathologic downstaging (pDS) to neoadjuvant chemotherapy (NAC) is one of the most important predictors of survival in muscle-invasive bladder cancer (MIBC). The use of NAC is limited as pDS is only achieved in 30% to 40% of cases and predictive biomarkers are still lacking. We performed a comprehensive immunomolecular biomarker analysis to characterize the role of immune cells and inhibitory checkpoints, genome-wide frequencies of copy number alterations, mutational signatures in whole exome, and tumor mutational burden in predicting NAC response. Our retrospective study included 23 primary MIBC patients who underwent NAC, followed by radical cystectomy. pDS to NAC was a significant prognostic factor for better recurrence-free survival (P < 0.001), with a median time to recurrence of 41.2 versus 5.5 months in nonresponders. DNA damage repair alterations were noticed in 38.1% (n = 8), confirming a positive correlation with high tumor mutational burden (P = 0.007). Chromosomal 7p12 amplification, including the genes HUS1, EGFR, ABCA13, and IKZF1, predicted nonresponse in patients with a sensitivity, a negative predictive value, and a specificity of 71.4%, 87.5%, and 100%, respectively. Total count of CD3+ T cells/mm2 tumor was a significant predictor of NAC response. In conclusion, 7p12 amplification may predict nonresponse to NAC and worse survival in MIBC. Multicenter, prospective trials with sufficient statistical power may further fortify these findings.
Collapse
Affiliation(s)
- Renate Pichler
- Department of Urology, Medical University Innsbruck, Innsbruck, Austria.
| | - Andrea K Lindner
- Department of Urology, Medical University Innsbruck, Innsbruck, Austria
| | - Eva Compérat
- Department of Pathology, Hôspital Tenon, HUEP, Sorbonne University, Paris, France
| | - Peter Obrist
- Pathology Laboratory Obrist and Brunhuber, Zams, Austria
| | - Georg Schäfer
- Department of Pathology, Medical University Innsbruck, Innsbruck, Austria
| | | | | | - Zoran Culig
- Department of Urology, Medical University Innsbruck, Innsbruck, Austria.
| | - Gerold Untergasser
- Department of Internal Medicine V, Medical University Innsbruck, Innsbruck, Austria; Tyrolean Cancer Research Institute, Innsbruck, Austria
| |
Collapse
|
12
|
Abstract
The recent genomic characterization of urothelial carcinoma by the Cancer Genome Atlas Project, made possible by the introduction of high throughput, reduced cost, and sequence analysis, has shed new insights on the biology of advanced disease. In addition, studies on imaging of advanced urothelial carcinoma have widened the knowledge on disease presentation and on pattern of metastatic spread and their correlation with the underlying biology of urothelial carcinoma. The wide range of treatments for advanced urothelial cancer, including combined chemotherapy regimens and immune checkpoint inhibitors, each result in treatment class-specific patterns of response and adverse events. Results of studies point to the need for a reliable biomarker, perhaps with imaging, that predicts prognosis and treatment response to systemic treatment, and can be used to select the most effective treatment while minimizing toxicity. This review of advanced urothelial cancer introduces the latest advances in genetic profiling, the current role of imaging, the radiographic appearance of treatment response and their toxicities, and details potential future areas of imaging research.
Collapse
|
13
|
Intrinsic Molecular Subclassification of Urothelial Carcinoma of the Bladder: Are We Finally there? Adv Anat Pathol 2019; 26:251-256. [PMID: 31188799 DOI: 10.1097/pap.0000000000000235] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Bladder cancer is a highly prevalent disease throughout the world usually encountered in older patients, and associated with substantial morbidity, mortality, and cost. The treatment of bladder cancer has remained unchanged for the last several decades. However, in recent years the availability of comprehensive genomic data from The Cancer Genome Atlas and other large projects have considerably improved our understanding of the pathogenesis of these tumors. These studies demonstrated that bladder cancers can be grouped into 2 broad categories namely basal and luminal molecular subtypes with recognizable subgroups in each of these categories. Clinical data suggest that invasive basal cancers are more sensitive to neoadjuvant chemotherapy (NAC), such that most patients with basal cancers who are aggressively managed with NAC have excellent outcomes. Patients with luminal cancers do not appear to derive much clinical benefit from NAC, but some may appear to be sensitive to anti-programmed death-ligand 1 (PDL1) antibodies and possibly other immune checkpoint inhibitors. It is hoped that future studies will also identify biomarkers such as immunohistochemical markers which may be used to predict therapeutic response of these tumors. This will contribute substantially toward efficient and cost-effective diagnosis and management of these neoplasms.
Collapse
|
14
|
Houédé N, Milano G. [Care of advanced or metastatic bladder cancer in second line: A specific place for vinflunine]. Bull Cancer 2019; 106:431-435. [PMID: 30981463 DOI: 10.1016/j.bulcan.2019.02.007] [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: 11/12/2018] [Revised: 01/25/2019] [Accepted: 02/19/2019] [Indexed: 10/27/2022]
Abstract
Urothelial carcinoma of the bladder are rare but aggressive tumors with a high metastatic potential. The prognosis of these tumors has not drastically changed over the past 30 years, with an overall survival of less than two years in advanced or metastatic situations. Even though immune checkpoints inhibitors have changed this picture, it is beneficial for less than 30% of patients and there is no reliable biomarkers to identify this specific population of responders. Vinflunine is a vinca-alkaloid that was specifically developed as second line treatment post-platinum. As of today, it is the sole anticancer agent for which clinical trials have been pushed to phase III and that was approved for patients in good conditions. Unfortunately, it has been withdrawn from the list of reimbursed drugs, which impairs its prescription. Based on the results of phase III clinical trials with immunotherapies, this review provides the reader with argumentations in favor of patients' and clinicians' request to reimburse vinflunine for the treatment of advanced or metastatic urothelial carcinoma of the bladder.
Collapse
Affiliation(s)
- Nadine Houédé
- Montpellier university, centre hospitalier universitaire Carremeau Nîmes, place du Pr Robert-Debré, 30029 Nîmes, France.
| | | |
Collapse
|
15
|
Alessandrino F, Gujrathi R, Nassar AH, Alzaghal A, Ravi A, McGregor B, Sonpavde G, Shinagare AB. Predictive Role of Computed Tomography Texture Analysis in Patients with Metastatic Urothelial Cancer Treated with Programmed Death-1 and Programmed Death-ligand 1 Inhibitors. Eur Urol Oncol 2019; 3:680-686. [PMID: 31412003 DOI: 10.1016/j.euo.2019.02.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/31/2019] [Accepted: 02/14/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Reliable biomarkers to predict the response of metastatic urothelial cancer (mUC) to programmed death-1 and programmed death-ligand 1 (PD-1/PD-L1) inhibitors are being investigated. Texture analysis represents tumor heterogeneity and may serve as a predictor of response in mUC. OBJECTIVE To assess the predictive ability of computed tomography (CT) texture analysis for progression-free survival (PFS) in patients with mUC treated with PD-1/PD-L1 inhibitors. DESIGN, SETTING, AND PARTICIPANTS Forty-two postplatinum patients with mUC treated with PD-1/PD-L1 inhibitors from 2013 to 2018, including those with measurable disease per RECIST 1.1 who had contrast-enhanced baseline or first follow-up CT within 3mo after starting treatment, were included. PFS was calculated based on serial follow-up CT scans. Eleven patients with follow-up of <12mo without progression were excluded. Texture features of measurable lesions on baseline and first follow-up CT were extracted using commercially available software (TexRAD; Feedback Plc, Cambridge, UK) using different spatial scaling factors (0, 2-6). OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Stepwise logistic regression analysis was conducted to identify patients with PFS <12mo, and performance was assessed using receiver operator characteristic curves. RESULTS AND LIMITATIONS Of 31 included patients, 18 had PFS <12mo. Twenty-five baseline CT and 29 first follow-up CT scans met the inclusion criteria. In patients with PFS <12mo, entropy and mean were higher on first follow-up CT (p=0.02 and p=0.005, respectively). A predictive model including mean and entropy on first follow-up CT yielded 95% sensitivity, 80% specificity, 90% positive predictive value, 89% negative predictive value, and 90% accuracy (area under the curve=0.963) to identify patients with PFS <12mo. Limitations include retrospective nature and small sample size. CONCLUSIONS CT texture analysis can help predict early progression with high accuracy soon after starting PD-1/PD-L1 inhibitors. Studies investigating the correlation of texture analysis with survival endpoints may help validate texture analysis as a biomarker of PD-1/PD-L1 inhibitors' treatment response. PATIENT SUMMARY Computed tomography texture analysis can help predict durability of response in patients with metastatic urothelial cancer early during treatment with programmed death-1 and programmed death-ligand 1 (PD-1/PD-L1) inhibitors.
Collapse
Affiliation(s)
- Francesco Alessandrino
- Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rahul Gujrathi
- Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Amin H Nassar
- Lank Center for Genitourinary Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Arwa Alzaghal
- Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Arvind Ravi
- Lank Center for Genitourinary Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Broad Institute, Cambridge, MA, USA
| | - Bradley McGregor
- Lank Center for Genitourinary Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Guru Sonpavde
- Lank Center for Genitourinary Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Atul B Shinagare
- Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
16
|
Culine S, Allory Y, Pfister C. Refining the use of neoadjuvant chemotherapy in locally advanced bladder cancer: from conviction to optimization. Transl Androl Urol 2018; 7:757-759. [PMID: 30211071 PMCID: PMC6127548 DOI: 10.21037/tau.2018.06.11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Stéphane Culine
- Department of Medical Oncology, Hôpital Saint-Louis, AP-HP, and Paris-Diderot University, Paris, France
| | - Yves Allory
- Department of Pathology, Institut Curie, Saint-Cloud, France
| | - Christian Pfister
- Department of Urology, Centre Hospitalier Universitaire, Rouen University, Rouen, France
| |
Collapse
|
17
|
Rouanne M, Roumiguié M, Houédé N, Masson-Lecomte A, Colin P, Pignot G, Larré S, Xylinas E, Rouprêt M, Neuzillet Y. Development of immunotherapy in bladder cancer: present and future on targeting PD(L)1 and CTLA-4 pathways. World J Urol 2018; 36:1727-1740. [PMID: 29855698 DOI: 10.1007/s00345-018-2332-5] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 05/08/2018] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Over the past 3 decades, no major treatment breakthrough has been reported for advanced bladder cancer. Recent Food and Drug Administration (FDA) approval of five immune checkpoint inhibitors in the management of advanced bladder cancer represent new therapeutic opportunities. This review examines the available data of the clinical trials leading to the approval of ICIs in the management of metastatic bladder cancer and the ongoing trials in advanced and localized settings. METHODS A literature search was performed on PubMed and ClinicalTrials.gov combining the MeSH terms: 'urothelial carcinoma' OR 'bladder cancer', and 'immunotherapy' OR 'CTLA-4' OR 'PD-1' OR 'PD-L1' OR 'atezolizumab' OR 'nivolumab' OR 'ipilimumab' OR 'pembrolizumab' OR 'avelumab' OR 'durvalumab' OR 'tremelimumab'. Prospectives studies evaluating anti-PD(L)1 and anti-CTLA-4 monoclonal antibodies were included. RESULTS Evidence-data related to early phase and phase III trials evaluating the 5 ICIs in the advanced urothelial carcinoma are detailed in this review. Anti-tumour activity of the 5 ICIs supporting the FDA approval in the second-line setting are reported. The activity of PD(L)1 inhibitors in the first-line setting in cisplatin-ineligible patients are also presented. Ongoing trials in earlier disease-states including non-muscle-invasive and muscle-invasive bladder cancer are discussed. CONCLUSIONS Blocking the PD-1 negative immune receptor or its ligand, PD-L1, results in unprecedented rates of anti-tumour activity in patients with metastatic urothelial cancer. However, a large majority of patients do not respond to anti-PD(L)1 drugs monotherapy. Investigations exploring the potential value of predictive biomarkers, optimal combination and sequences are ongoing to improve such treatment strategies.
Collapse
Affiliation(s)
- Mathieu Rouanne
- Department of Urology, Hôpital Foch, Université Versailles-Saint-Quentin-en-Yvelines, Université Paris-Saclay, 40 Rue Worth, 92150, Suresnes, France. .,INSERM U1015, Gustave Roussy, Université Paris-Saclay, Villejuif, France.
| | - Mathieu Roumiguié
- Comité de Cancérologie de l'Association Française d'Urologie (ccAFU), Bladder Cancer Group, Maison de l'Urologie, Paris, France.,Department of Urology, Institut Universitaire du Cancer, Oncopole, Toulouse, France
| | - Nadine Houédé
- Comité de Cancérologie de l'Association Française d'Urologie (ccAFU), Bladder Cancer Group, Maison de l'Urologie, Paris, France.,Department of Medical Oncology, CHU de Nîmes, Nimes, France.,INSERM U1194, Montpellier Cancer Research Institute, Université de Montpellier, Montpellier, France
| | - Alexandra Masson-Lecomte
- Comité de Cancérologie de l'Association Française d'Urologie (ccAFU), Bladder Cancer Group, Maison de l'Urologie, Paris, France.,Department of Urology, Hôpital Saint-Louis, Université Paris-Diderot, Paris, France
| | - Pierre Colin
- Comité de Cancérologie de l'Association Française d'Urologie (ccAFU), Bladder Cancer Group, Maison de l'Urologie, Paris, France.,Department of Urology, Hôpital privé de la Louvière, Lille, France
| | - Géraldine Pignot
- Comité de Cancérologie de l'Association Française d'Urologie (ccAFU), Bladder Cancer Group, Maison de l'Urologie, Paris, France.,Department of Urology, Institut Paoli-Calmettes, Marseille, France
| | - Stéphane Larré
- Comité de Cancérologie de l'Association Française d'Urologie (ccAFU), Bladder Cancer Group, Maison de l'Urologie, Paris, France.,Department of Urology, CHU de Reims, Reims, France
| | - Evanguelos Xylinas
- Comité de Cancérologie de l'Association Française d'Urologie (ccAFU), Bladder Cancer Group, Maison de l'Urologie, Paris, France.,Department of Urology, CHU Bichat, Paris, France
| | - Morgan Rouprêt
- Comité de Cancérologie de l'Association Française d'Urologie (ccAFU), Bladder Cancer Group, Maison de l'Urologie, Paris, France.,Department of Urology, Hôpital La Pitié-Salpétrière, AP-HP, GRC n°5, ONCOTYPE-URO, Paris, France
| | - Yann Neuzillet
- Department of Urology, Hôpital Foch, Université Versailles-Saint-Quentin-en-Yvelines, Université Paris-Saclay, 40 Rue Worth, 92150, Suresnes, France.,Comité de Cancérologie de l'Association Française d'Urologie (ccAFU), Bladder Cancer Group, Maison de l'Urologie, Paris, France
| |
Collapse
|