1
|
Maroof H, Paramore L, Ali A. Theories behind Bacillus Calmette-Guérin failure in high-risk non-muscle-invasive bladder cancer and update on current management. CANCER PATHOGENESIS AND THERAPY 2024; 2:74-80. [PMID: 38601486 PMCID: PMC11002745 DOI: 10.1016/j.cpt.2023.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 11/17/2023] [Accepted: 11/26/2023] [Indexed: 04/12/2024]
Abstract
Bladder cancer encapsulates a wide spectrum of disease severities, with non-muscle invasive bladder cancer (NMIBC) representing an entirely different entity from muscle-invasive disease. Bacillus Calmette-Guérin (BCG) is one of the most successful intravesical treatment methods for patients diagnosed. However, a considerable proportion of patients fail to respond to BCG treatment. Given the propensity for recurrence in patients with high-risk bladder cancer, these patients present with surgical dilemmas. There is currently no gold standard for salvage treatment post-BCG failure or unified definition as to what that means. In this review, we discuss the mechanisms of action and pathophysiology of BCG, potential theories behind BCG failure, and the scope of novel treatments for this surgical conundrum.
Collapse
Affiliation(s)
- Hanna Maroof
- Department of Urology, Frimley Park Hospital, Portsmouth Rd, Frimley, Camberley GU16 7UJ, United Kingdom
| | - Louise Paramore
- Department of Urology, Frimley Park Hospital, Portsmouth Rd, Frimley, Camberley GU16 7UJ, United Kingdom
| | - Ahmed Ali
- Department of Urology, Frimley Park Hospital, Portsmouth Rd, Frimley, Camberley GU16 7UJ, United Kingdom
| |
Collapse
|
2
|
Porreca A, Di Nicola M, Lucarelli G, Dorin VM, Soria F, Terracciano D, Mistretta FA, Luzzago S, Buonerba C, Cantiello F, Mari A, Minervini A, Veccia A, Antonelli A, Musi G, Hurle R, Busetto GM, Del Giudice F, Ferretti S, Perdonà S, Prete PD, Porreca A, Bove P, Crisan N, Russo GI, Damiano R, Amparore D, Porpiglia F, Autorino R, Piccinelli M, Brescia A, Tătaru SO, Crocetto F, Giudice AL, de Cobelli O, Schips L, Ferro M, Marchioni M. Time to progression is the main predictor of survival in patients with high-risk nonmuscle invasive bladder cancer: Results from a machine learning-based analysis of a large multi-institutional database. Urol Oncol 2024; 42:69.e17-69.e25. [PMID: 38302296 DOI: 10.1016/j.urolonc.2024.01.001] [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: 06/10/2023] [Revised: 11/13/2023] [Accepted: 01/01/2024] [Indexed: 02/03/2024]
Abstract
BACKGROUND In patients affected by high-risk nonmuscle invasive bladder cancer (HR-NMIBC) progression to muscle invasive status is considered as the main indicator of local treatment failure. We aimed to investigate the effect of progression and time to progression on overall survival (OS) and to investigate their validity as surrogate endpoints. METHODS A total of 1,510 patients from 18 different institutions treated for T1 high grade NMIBC, followed by a secondary transurethral resection and BCG intravesical instillation. We relied on random survival forest (RSF) to rank covariates based on OS prediction. Cox's regression models were used to quantify the effect of covariates on mortality. RESULTS During a median follow-up of 49.0 months, 485 (32.1%) patients progressed to MIBC, while 163 (10.8%) patients died. The median time to progression was 82 (95%CI: 78.0-93.0) months. In RSF time-to-progression and age were the most predictive covariates of OS. The survival tree defined 5 groups of risk. In multivariable Cox's regression models accounting for progression status as time-dependent covariate, shorter time to progression (as continuous covariate) was associated with longer OS (HR: 9.0, 95%CI: 3.0-6.7; P < 0.001). Virtually same results after time to progression stratification (time to progression ≥10.5 months as reference). CONCLUSION Time to progression is the main predictor of OS in patients with high risk NMIBC treated with BCG and might be considered a coprimary endpoint. In addition, models including time to progression could be considered for patients' stratification in clinical practice and at the time of clinical trials design.
Collapse
Affiliation(s)
- Annamaria Porreca
- Biostatistics Laboratory, Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti, Chieti, Italy
| | - Marta Di Nicola
- Biostatistics Laboratory, Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti, Chieti, Italy
| | - Giuseppe Lucarelli
- Urology, Andrology and Kidney Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Vartolomei Mihai Dorin
- Department of Cell and Molecular Biology, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology from Târgu Mureș, Târgu Mureș, Romania
| | - Francesco Soria
- Division of Urology, Department of Surgical Sciences, Torino School of Medicine, Torino, Italy
| | - Daniela Terracciano
- Department of Translational Medical Sciences, University Federico II, Napoli, Italy
| | | | - Stefano Luzzago
- Department of Urology, IEO European Institute of Oncology, IRCCS, Milan, Italy
| | - Carlo Buonerba
- CRTR Rare Tumors Reference Center, AOU Federico II, Napoli, Italy
| | - Francesco Cantiello
- Department of Urology, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Andrea Mari
- Unit of Oncologic Minimally-Invasive Urology and Andrology, Department of Urology, Careggi Hospital, University of Florence, Florence, Italy
| | - Andrea Minervini
- Unit of Oncologic Minimally-Invasive Urology and Andrology, Department of Urology, Careggi Hospital, University of Florence, Florence, Italy
| | | | | | - Gennaro Musi
- Department of Urology, IEO European Institute of Oncology, IRCCS, Milan, Italy
| | - Rodolfo Hurle
- Department of Urology, Humanitas Clinical and Research Hospital, Rozzano, Milan, Italy
| | | | | | - Simone Ferretti
- Urology Unit, Department of Medical, Oral and Biotechnological Sciences, "SS. Annunziata" Hospital, "G. d'Annunzio" University of Chieti, Chieti, Italy
| | - Sisto Perdonà
- Uro-Gynecological Department, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "G. Pascale"-IRCCS, Naples, Italy
| | - Paola Del Prete
- Scientific Directorate, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "G. Pascale"-IRCCS, Naples, Italy
| | - Angelo Porreca
- Department of Robotic Urologic Surgery, Abano Terme Hospital, Abano Terme, Italy
| | - Pierluigi Bove
- Division of Urology, Department of Experimental Medicine and Surgery, Tor Vergata University of Rome, Rome, Italy
| | - Nicolae Crisan
- Department of Urology, University of Medicine and Pharmacy Iuliu Haţieganu, Cluj-Napoca, Romania
| | | | - Rocco Damiano
- Department of Urology, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Daniele Amparore
- Division of Urology, Department of Oncology, School of Medicine, San Luigi Hospital, University of Turin, Orbassano, Turin, Italy
| | - Francesco Porpiglia
- Division of Urology, Department of Oncology, School of Medicine, San Luigi Hospital, University of Turin, Orbassano, Turin, Italy
| | | | - Mattia Piccinelli
- Department of Urology, IEO European Institute of Oncology, IRCCS, Milan, Italy
| | - Antonio Brescia
- Department of Urology, IEO European Institute of Oncology, IRCCS, Milan, Italy
| | - Sabin Octavian Tătaru
- I.O.S.U.D., George Emil Palade University of Medicine, Pharmacy, Sciences and Technology from Târgu Mureș, Târgu Mureș, Romania
| | - Felice Crocetto
- Department of Neurosciences, Science of Reproduction and Odontostomatology, University of Naples Federico II, 80131 Naples, Italy
| | | | - Ottavio de Cobelli
- Department of Urology, IEO European Institute of Oncology, IRCCS, Milan, Italy
| | - Luigi Schips
- Urology Unit, Department of Medical, Oral and Biotechnological Sciences, "SS. Annunziata" Hospital, "G. d'Annunzio" University of Chieti, Chieti, Italy
| | - Matteo Ferro
- Department of Urology, IEO European Institute of Oncology, IRCCS, Milan, Italy
| | - Michele Marchioni
- Urology Unit, Department of Medical, Oral and Biotechnological Sciences, "SS. Annunziata" Hospital, "G. d'Annunzio" University of Chieti, Chieti, Italy.
| |
Collapse
|
4
|
Chen M, Bie L, Ying J. Cancer cell-intrinsic PD-1: Its role in malignant progression and immunotherapy. Biomed Pharmacother 2023; 167:115514. [PMID: 37716115 DOI: 10.1016/j.biopha.2023.115514] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/08/2023] [Accepted: 09/12/2023] [Indexed: 09/18/2023] Open
Abstract
Programmed cell death protein-1 (PD-1), also called CD279, is coded by the PDCD1 gene and is constitutively expressed on the surface of immune cells. As a receptor and immune checkpoint, PD-1 can bind to programmed death ligand-1/programmed death ligand-2 (PD-L1/PD-L2) in tumor cells, leading to tumor immune evasion. Anti-PD-1 and anti-PD-L1 are important components in tumor immune therapy. PD-1 is also expressed as an intrinsic variant (iPD-1) in cancer cells where it plays important roles in malignant progression as proposed by recent studies. However, iPD-1 has received much less attention compared to PD-1 expressed on immune cells although there is an unmet medical need for fully elucidating the mechanisms of actions to achieve the best response in tumor immunotherapy. iPD-1 suppresses tumorigenesis in non-small cell lung cancer (NSCLC) and colon cancer, whereas it promotes tumorigenesis in melanoma, hepatocellular carcinoma (HCC), pancreatic ductal adenocarcinoma (PDAC), thyroid cancer (TC), glioblastoma (GBM), and triple-negative breast cancer (TNBC). In this review, we focus on the role of iPD-1 in tumorigenesis and development and its molecular mechanisms. We also deeply discuss nivolumab-based combined therapy in common tumor therapy. iPD-1 may explain the different therapeutic effects of anti-PD-1 treatment and provide critical information for use in combined anti-tumor approaches.
Collapse
Affiliation(s)
- Muhua Chen
- Department of Hepato-Pancreato-Biliary & Gastric Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| | - Lei Bie
- Department of Thoracic Surgery, Wuhan No.1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Jieer Ying
- Department of Hepato-Pancreato-Biliary & Gastric Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| |
Collapse
|
5
|
Freshour SL, Chen THP, Fisk B, Shen H, Mosior M, Skidmore ZL, Fronick C, Bolzenius JK, Griffith OL, Arora VK, Griffith M. Endothelial cells are a key target of IFN-g during response to combined PD-1/CTLA-4 ICB treatment in a mouse model of bladder cancer. iScience 2023; 26:107937. [PMID: 37810214 PMCID: PMC10558731 DOI: 10.1016/j.isci.2023.107937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 08/08/2023] [Accepted: 09/13/2023] [Indexed: 10/10/2023] Open
Abstract
To explore mechanisms of response to combined PD-1/CTLA-4 immune checkpoint blockade (ICB) treatment in individual cell types, we generated scRNA-seq using a mouse model of invasive urothelial carcinoma with three conditions: untreated tumor, treated tumor, and tumor treated after CD4+ T cell depletion. After classifying tumor cells based on detection of somatic variants and assigning non-tumor cell types using SingleR, we performed differential expression analysis, overrepresentation analysis, and gene set enrichment analysis (GSEA) within each cell type. GSEA revealed that endothelial cells were enriched for upregulated IFN-g response genes when comparing treated cells to both untreated cells and cells treated after CD4+ T cell depletion. Functional analysis showed that knocking out IFNgR1 in endothelial cells inhibited treatment response. Together, these results indicated that IFN-g signaling in endothelial cells is a key mediator of ICB induced anti-tumor activity.
Collapse
Affiliation(s)
- Sharon L. Freshour
- Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Timothy H.-P. Chen
- Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Bryan Fisk
- Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Haolin Shen
- Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Matthew Mosior
- Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Zachary L. Skidmore
- Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Catrina Fronick
- McDonnell Genome Institute, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Jennifer K. Bolzenius
- Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- Siteman Cancer Center, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Obi L. Griffith
- Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- Siteman Cancer Center, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- Department of Genetics, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Vivek K. Arora
- Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- Siteman Cancer Center, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Malachi Griffith
- Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- Siteman Cancer Center, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- Department of Genetics, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|