1
|
Abstract
Currently available treatment options for patients with refractory metastatic prostate, bladder, or kidney cancers are limited with the prognosis remaining poor. Advances in the pathobiology of tumors has led to the discovery of cancer antigens that may be used as the target for cancer treatment. Antibody-drug conjugates (ADCs) are a relatively new concept in cancer treatment that broaden therapeutic landscape. ADCs are examples of a 'drug delivery into the tumor' system composed of an antigen-directed antibody linked to a cytotoxic drug that may release cytotoxic components after binding to the antigen located on the surface of tumor cells. The clinical properties of drugs are influenced by every component of ADCs. Regarding uro-oncology, enfortumab vedotin (EV) and sacituzumab govitecan (SG) are currently registered for patients with locally advanced or metastatic urothelial cancer following previous treatment with an immune checkpoint inhibitor (iCPI; programmed death receptor-1 [PD-1] or programmed death-ligand 1 [PD-L1]) inhibitor) and platinum-containing chemotherapy. The EV-301 trial showed that EV significantly prolonged the overall survival compared with classic chemotherapy. The TROPHY-U-01 trial conducted to evaluate SG demonstrated promising results as regards the objective response rate and duration of response. The safety and efficacy of ADCs in monotherapy and polytherapy (mainly with iCPIs) for different cancer stages and tumor types are assessed in numerous ongoing clinical trials. The aim of this review is to present new molecular biomarkers, specific mechanisms of action, and ongoing clinical trials of ADCs in genitourinary cancers. In the expert discussion, we assess the place of ADCs in uro-oncology and discuss their clinical value.
Collapse
|
2
|
Rampling R, Peoples S, Mulholland PJ, James A, Al-Salihi O, Twelves CJ, McBain C, Jefferies S, Jackson A, Stewart W, Lindner J, Kutscher S, Hilf N, McGuigan L, Peters J, Hill K, Schoor O, Singh-Jasuja H, Halford SE, Ritchie JWA. A Cancer Research UK First Time in Human Phase I Trial of IMA950 (Novel Multipeptide Therapeutic Vaccine) in Patients with Newly Diagnosed Glioblastoma. Clin Cancer Res 2016; 22:4776-4785. [PMID: 27225692 PMCID: PMC5026298 DOI: 10.1158/1078-0432.ccr-16-0506] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 05/11/2016] [Indexed: 12/31/2022]
Abstract
PURPOSE To perform a two-cohort, phase I safety and immunogenicity study of IMA950 in addition to standard chemoradiotherapy and adjuvant temozolomide in patients with newly diagnosed glioblastoma. IMA950 is a novel glioblastoma-specific therapeutic vaccine containing 11 tumor-associated peptides (TUMAP), identified on human leukocyte antigen (HLA) surface receptors in primary human glioblastoma tissue. EXPERIMENTAL DESIGN Patients were HLA-A*02-positive and had undergone tumor resection. Vaccination comprised 11 intradermal injections with IMA950 plus granulocyte macrophage colony-stimulating factor (GM-CSF) over a 24-week period, beginning 7 to 14 days prior to initiation of chemoradiotherapy (Cohort 1) or 7 days after chemoradiotherapy (Cohort 2). Safety was assessed according to NCI CTCAE Version 4.0 and TUMAP-specific T-cell immune responses determined. Secondary observations included progression-free survival (PFS), pretreatment regulatory T cell (Treg) levels, and the effect of steroids on T-cell responses. RESULTS Forty-five patients were recruited. Related adverse events included minor injection site reactions, rash, pruritus, fatigue, neutropenia and single cases of allergic reaction, anemia and anaphylaxis. Two patients experienced grade 3 dose-limiting toxicity of fatigue and anaphylaxis. Of 40 evaluable patients, 36 were TUMAP responders and 20 were multi-TUMAP responders, with no important differences between cohorts. No effect of pretreatment Treg levels on IMA950 immunogenicity was observed, and steroids did not affect TUMAP responses. PFS rates were 74% at 6 months and 31% at 9 months. CONCLUSIONS IMA950 plus GM-CSF was well-tolerated with the primary immunogenicity endpoint of observing multi-TUMAP responses in at least 30% of patients exceeded. Further development of IMA950 is encouraged. Clin Cancer Res; 22(19); 4776-85. ©2016 AACRSee related commentary by Lowenstein and Castro, p. 4760.
Collapse
Affiliation(s)
- Roy Rampling
- University of Glasgow, Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | - Sharon Peoples
- Edinburgh Centre for Neuro-Oncology, Western General Hospital, Edinburgh, United Kingdom
| | - Paul J Mulholland
- Department of Oncology, University College London Hospitals, London, United Kingdom
| | - Allan James
- University of Glasgow, Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | - Omar Al-Salihi
- Adult Neuro-Oncology, Southampton University Hospitals NHS Trust, Southampton, United Kingdom
| | - Christopher J Twelves
- Cancer Research UK Clinical Centre, St James's University Hospital, Leeds, United Kingdom
| | - Catherine McBain
- The Christie NHS Foundation Trust, Withington, Manchester, United Kingdom
| | - Sarah Jefferies
- Cambridge Cancer Trials Centre, Oncology Clinical Trials, Addensbrooke's Hospital, Cambridge, United Kingdom
| | - Alan Jackson
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, United Kingdom
| | - Willie Stewart
- Department of Neuropathology, The Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Juha Lindner
- Immatics Biotechnologies GmbH, Tübingen, Germany
| | | | - Norbert Hilf
- Immatics Biotechnologies GmbH, Tübingen, Germany
| | - Lesley McGuigan
- Cancer Research UK Centre for Drug Development, London, United Kingdom
| | - Jane Peters
- Cancer Research UK Centre for Drug Development, London, United Kingdom
| | - Karen Hill
- Cancer Research UK Centre for Drug Development, London, United Kingdom
| | | | | | - Sarah E Halford
- Cancer Research UK Centre for Drug Development, London, United Kingdom
| | - James W A Ritchie
- Cancer Research UK Centre for Drug Development, London, United Kingdom.
| |
Collapse
|
3
|
Aoun F, Kourie HR, Sideris S, Roumeguère T, Velthoven RV, Gil T. Checkpoint inhibitors in bladder and renal cancers: results and perspectives. Immunotherapy 2015; 7:1259-71. [DOI: 10.2217/imt.15.91] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The field of immunotherapy in urinary malignancy is expanding in several directions and checkpoint inhibitors are leading the way. The aim of this report is to highlight the efficacy and safety profile of the two classes of molecules, anti-cytotoxic T-lymphocyte antigen-4 and anti-programmed death receptor-1/programmed death ligand type 1, that are under investigation and represent potential candidates to be used in the near future for the management of bladder and renal cell cancer. The preliminary results as well as the future perspectives of this novel immunotherapy are analyzed. Novel immune checkpoint targets are reviewed as well.
Collapse
Affiliation(s)
- Fouad Aoun
- Department of Urology, Jules Bordet Institute, 1 Héger Bordet Street, 1000 Brussels, Belgium
| | - Hampig R Kourie
- Department of Oncology, Jules Bordet Institute, 1 Héger Bordet Street, 1000 Brussels, Belgium
| | - Spyridon Sideris
- Department of Oncology, Jules Bordet Institute, 1 Héger Bordet Street, 1000 Brussels, Belgium
| | - Thierry Roumeguère
- Department of Urology, Erasme Hospital, Route de Lennik 808, 1070 Brussels, Belgium
| | - Roland van Velthoven
- Department of Urology, Jules Bordet Institute, 1 Héger Bordet Street, 1000 Brussels, Belgium
| | - Thierry Gil
- Department of Oncology, Jules Bordet Institute, 1 Héger Bordet Street, 1000 Brussels, Belgium
| |
Collapse
|
4
|
Lerut E, Van Poppel H, Joniau S, Gruselle O, Coche T, Therasse P. Rates of MAGE-A3 and PRAME expressing tumors in FFPE tissue specimens from bladder cancer patients: potential targets for antigen-specific cancer immunotherapeutics. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:9522-9532. [PMID: 26464715 PMCID: PMC4583947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 07/24/2015] [Indexed: 06/05/2023]
Abstract
INTRODUCTION Antigen-specific active immunotherapy is an investigational therapeutic approach of potential interest for bladder cancer regardless of disease stage. Clinical development of antigen-specific immunotherapeutics against bladder cancer must be preceded by assessment of the expression of relevant genes in bladder tumors. The objectives of this study (NCT01706185) were to assess the rate of expression of the MAGE-A3 and PRAME genes in bladder tumors and to investigate the feasibility of using formalin-fixed paraffin-embedded (FFPE) tumor tissues for testing. MATERIALS AND METHODS Archived FFPE bladder tumor specimens (any stage) were tested for mRNA expression of MAGE-A3 and PRAME using antigen-specific quantitative reverse transcription polymerase chain reaction assays. Data on patients and tumor characteristics were obtained from hospital records to investigate these characteristics' possible association with the antigen expression. RESULTS Over 92% of the 156 tumors examined gave valid antigen test results. Of the tumors with a valid test, 46.5% were MAGE-A3-positive, 32.2% were PRAME-positive and 59.7% positive for at least one of them. Exploratory analyses of possible associations between antigen expression and patient or tumor characteristics did not identify clear associations between antigen expression and any of the variables investigated. CONCLUSIONS Assessment of tumor antigen mRNA expression by using FFPE bladder tissues was feasible. The rates of MAGE-A3-positive and PRAME-positive tumors indicate that both antigens may be interesting targets for immunotherapeutics against bladder cancer.
Collapse
Affiliation(s)
- Evelyne Lerut
- Department of Pathology, University Hospitals LeuvenLeuven, Belgium
| | | | - Steven Joniau
- Department of Urology, University Hospitals LeuvenLeuven, Belgium
| | | | | | | |
Collapse
|
5
|
Target expression of Staphylococcus enterotoxin A from an oncolytic adenovirus suppresses mouse bladder tumor growth and recruits CD3+ T cell. Tumour Biol 2013; 34:2863-9. [DOI: 10.1007/s13277-013-0847-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 05/07/2013] [Indexed: 10/26/2022] Open
|