51
|
Zimmermannova O, Caiado I, Ferreira AG, Pereira CF. Cell Fate Reprogramming in the Era of Cancer Immunotherapy. Front Immunol 2021; 12:714822. [PMID: 34367185 PMCID: PMC8336566 DOI: 10.3389/fimmu.2021.714822] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022] Open
Abstract
Advances in understanding how cancer cells interact with the immune system allowed the development of immunotherapeutic strategies, harnessing patients' immune system to fight cancer. Dendritic cell-based vaccines are being explored to reactivate anti-tumor adaptive immunity. Immune checkpoint inhibitors and chimeric antigen receptor T-cells (CAR T) were however the main approaches that catapulted the therapeutic success of immunotherapy. Despite their success across a broad range of human cancers, many challenges remain for basic understanding and clinical progress as only a minority of patients benefit from immunotherapy. In addition, cellular immunotherapies face important limitations imposed by the availability and quality of immune cells isolated from donors. Cell fate reprogramming is offering interesting alternatives to meet these challenges. Induced pluripotent stem cell (iPSC) technology not only enables studying immune cell specification but also serves as a platform for the differentiation of a myriad of clinically useful immune cells including T-cells, NK cells, or monocytes at scale. Moreover, the utilization of iPSCs allows introduction of genetic modifications and generation of T/NK cells with enhanced anti-tumor properties. Immune cells, such as macrophages and dendritic cells, can also be generated by direct cellular reprogramming employing lineage-specific master regulators bypassing the pluripotent stage. Thus, the cellular reprogramming toolbox is now providing the means to address the potential of patient-tailored immune cell types for cancer immunotherapy. In parallel, development of viral vectors for gene delivery has opened the door for in vivo reprogramming in regenerative medicine, an elegant strategy circumventing the current limitations of in vitro cell manipulation. An analogous paradigm has been recently developed in cancer immunotherapy by the generation of CAR T-cells in vivo. These new ideas on endogenous reprogramming, cross-fertilized from the fields of regenerative medicine and gene therapy, are opening exciting avenues for direct modulation of immune or tumor cells in situ, widening our strategies to remove cancer immunotherapy roadblocks. Here, we review current strategies for cancer immunotherapy, summarize technologies for generation of immune cells by cell fate reprogramming as well as highlight the future potential of inducing these unique cell identities in vivo, providing new and exciting tools for the fast-paced field of cancer immunotherapy.
Collapse
Affiliation(s)
- Olga Zimmermannova
- Cell Reprogramming in Hematopoiesis and Immunity Laboratory, Lund Stem Cell Center, Department of Molecular Medicine and Gene Therapy, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Inês Caiado
- Cell Reprogramming in Hematopoiesis and Immunity Laboratory, Lund Stem Cell Center, Department of Molecular Medicine and Gene Therapy, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Doctoral Programme in Experimental Biology and Biomedicine, University of Coimbra, Coimbra, Portugal
| | - Alexandra G. Ferreira
- Cell Reprogramming in Hematopoiesis and Immunity Laboratory, Lund Stem Cell Center, Department of Molecular Medicine and Gene Therapy, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Doctoral Programme in Experimental Biology and Biomedicine, University of Coimbra, Coimbra, Portugal
| | - Carlos-Filipe Pereira
- Cell Reprogramming in Hematopoiesis and Immunity Laboratory, Lund Stem Cell Center, Department of Molecular Medicine and Gene Therapy, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
52
|
El Gharib K, Lilly E, Chebel R. Checkpoint inhibitors in BCG-unresponsive nonmuscle invasive bladder cancer: can they help spare the bladder? Immunotherapy 2021; 13:1105-1111. [PMID: 34184569 DOI: 10.2217/imt-2021-0030] [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: 11/21/2022] Open
Abstract
Intravesical BCG therapy has been for years, the standard of care in nonmuscle-invasive bladder cancer. But upon recurrence/relapse, radical cystectomy is imposed, due to the paucity of other therapeutic options. Immunotherapy has been revolutionizing cancer treatment, and its indications continue to broaden. It has been approved for the treatment of advanced urothelial cancer of the bladder, mainly as a second-line therapy. Its activity is being studied in nonmuscle-invasive bladder cancer that is not responsive to BCG; we herein report the trials investigating these checkpoint inhibitors (pembrolizumab, nivolumab, atezolizumab, durvalumab and avelumab) in this particular setting.
Collapse
Affiliation(s)
- Khalil El Gharib
- Department of Hematology-Oncology, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Eddy Lilly
- Department of Urology, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Roy Chebel
- Department of Urology, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| |
Collapse
|
53
|
Maio M, Blank C, Necchi A, Di Giacomo AM, Ibrahim R, Lahn M, Fox BA, Bell RB, Tortora G, Eggermont AMM. Neoadjuvant immunotherapy is reshaping cancer management across multiple tumour types: The future is now! Eur J Cancer 2021; 152:155-164. [PMID: 34107449 DOI: 10.1016/j.ejca.2021.04.035] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 04/12/2021] [Indexed: 12/30/2022]
Abstract
The Italian Network for Tumor Biotherapy (Network Italiano per la Bioterapia dei Tumori [NIBIT]) Foundation hosted its annual 2020 Think Tank meeting virtually, at which representatives from academic, clinical, industry, philanthropic, and regulatory organisations discussed the role of neoadjuvant immunotherapy for the treatment of cancer. Although the number of neoadjuvant immunotherapeutic trials is increasing across all malignancies, the Think Tank focused its discussion on the status of neoadjuvant trials in cutaneous melanoma (CM), muscle-invasive urothelial bladder cancer (MIBC), head and neck squamous cell carcinoma (HNSCC), and pancreatic adenocarcinoma (PDAC). Neoadjuvant developments in CM are nothing short of trailblazing. Pathologic Complete Response (pCR), pathologic near Complete Response, and partial Pathologic Responses reduce 90-100% of recurrences. This is in sharp contrast to targeted therapies in neoadjuvant CM trials, where only a pCR seems to reduce recurrence. The pCR rate after neoadjuvant immunotherapy varies among the different malignancies of CM, MIBC, HNSCC, and PDAC and may be associated with different reductions of recurrence rates. In CM, emerging evidence suggests that neoadjuvant immunotherapy with anti-CTLA-4 plus anti-PD1 is a game changer in patients with palpable nodal Stage III or resectable Stage IV disease by curing more patients, reducing recurrences and the need for surgical interventions, such as lymph node dissections and metastasectomies. The Think Tank panel discussed future approaches on how to optimise results across different tumour types. Future approaches should include reducing monocyte-mediated (tumour-associated macrophages) and fibroblast-mediated (cancer-associated fibroblasts) barriers in the tumour microenvironment to facilitate the recruitment of immune cells to the tumour site, to reduce immune-suppressive mediators, and to increase antigen presentation at the site of the tumour.
Collapse
Affiliation(s)
- Michele Maio
- Center for Immuno-Oncology, Department of Oncology, Medical Oncology and Immunotherapy, University Hospital of Siena, Viale Mario Bracci 16, Siena, Italy; Italian Network for Tumor Bio-Immunotherapy Foundation, Siena, Italy.
| | - Christian Blank
- Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands.
| | - Andrea Necchi
- Genitourinary Medical Oncology, Vita-Salute San Raffaele University, Via Olgettina 60, 20132 Milan, Italy.
| | - Anna Maria Di Giacomo
- Center for Immuno-Oncology, Department of Oncology, Medical Oncology and Immunotherapy, University Hospital of Siena, Viale Mario Bracci 16, Siena, Italy; Italian Network for Tumor Bio-Immunotherapy Foundation, Siena, Italy.
| | - Ramy Ibrahim
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA.
| | - Michael Lahn
- IOnctura SA, Avenue Secheron 15, Geneva, Switzerland.
| | - Bernard A Fox
- Earle A. Chiles Research Institute at the Robert W. Franz Cancer Center, Providence Cancer Institute, Providence Portland Medical Center, 4805 NE Glisan, Portland, OR 97213, USA.
| | - R Bryan Bell
- Earle A. Chiles Research Institute at the Robert W. Franz Cancer Center, Providence Cancer Institute, Providence Portland Medical Center, 4805 NE Glisan, Portland, OR 97213, USA.
| | - Giampaolo Tortora
- Medical Oncology, Fondazione Policlinico Universitario Gemelli IRCCS e Università Cattolica Del Sacro Cuore, Roma, Largo Agostino Gemelli 8, 00168 Roma, Italy.
| | - Alexander M M Eggermont
- Princess Máxima Center, University Medical Center Utrecht, Heidelberglaan 25, 3584 Utrecht, the Netherlands.
| |
Collapse
|
54
|
Zang J, Ye K, Fei Y, Zhang R, Chen H, Zhuang G. Immunotherapy in the Treatment of Urothelial Bladder Cancer: Insights From Single-Cell Analysis. Front Oncol 2021; 11:696716. [PMID: 34123863 PMCID: PMC8187798 DOI: 10.3389/fonc.2021.696716] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 05/11/2021] [Indexed: 12/25/2022] Open
Abstract
Urothelial bladder cancer (UBC) is a global challenge of public health with limited therapeutic options. Although the emergence of cancer immunotherapy, most notably immune checkpoint inhibitors, represents a major breakthrough in the past decade, many patients still suffer from unsatisfactory clinical outcome. A thorough understanding of the fundamental cellular and molecular mechanisms responsible for antitumor immunity may lead to optimized treatment guidelines and new immunotherapeutic strategies. With technological developments and protocol refinements, single-cell approaches have become powerful tools that provide unprecedented insights into the kaleidoscopic tumor microenvironment and intricate cell-cell communications. In this review, we summarize recent applications of single-cell analysis in characterizing the UBC multicellular ecosystem, and discuss how to leverage the high-resolution information for more effective immune-based therapies.
Collapse
Affiliation(s)
- Jingyu Zang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kaiyan Ye
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yang Fei
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ruiyun Zhang
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Haige Chen
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Guanglei Zhuang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
55
|
Hasanali SL, Morera DS, Racine RR, Hennig M, Ghosh S, Lopez LE, Hupe MC, Escudero DO, Wang J, Zhu H, Sarcan S, Azih I, Zhou M, Jordan AR, Terris MK, Kuczyk MA, Merseburger AS, Lokeshwar VB. HYAL4-V1/Chondroitinase (Chase) Drives Gemcitabine Resistance and Predicts Chemotherapy Failure in Patients with Bladder Cancer. Clin Cancer Res 2021; 27:4410-4421. [PMID: 34031055 DOI: 10.1158/1078-0432.ccr-21-0422] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 04/01/2021] [Accepted: 05/20/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Gemcitabine-based chemotherapy regimens are first-line for several advanced cancers. Because of better tolerability, gemcitabine + cisplatin is a preferred neoadjuvant, adjuvant, and/or palliative chemotherapy regimen for advanced bladder cancer. Nevertheless, predicting treatment failure and overcoming resistance remain unmet clinical needs. We discovered that splice variant (V1) of HYAL-4 is a first-in-class eukaryotic chondroitinase (Chase), and CD44 is its major substrate. V1 is upregulated in bladder cancer and drives a malignant phenotype. In this study, we investigated whether V1 drives chemotherapy resistance. EXPERIMENTAL DESIGN V1 expression was measured in muscle-invasive bladder cancer (MIBC) specimens by qRT-PCR and IHC. HYAL-4 wild-type (Wt) and V1 were stably expressed or silenced in normal urothelial and three bladder cancer cell lines. Transfectants were analyzed for chemoresistance and associated mechanism in preclinical models. RESULTS V1 levels in MIBC specimens of patients who developed metastasis, predicted response to gemcitabine + cisplatin adjuvant/salvage treatment and disease-specific mortality. V1-expressing bladder cells were resistant to gemcitabine but not to cisplatin. V1 expression neither affected gemcitabine influx nor the drug-efflux transporters. Instead, V1 increased gemcitabine metabolism and subsequent efflux of difluorodeoxyuridine, by upregulating cytidine deaminase (CDA) expression through increased CD44-JAK2/STAT3 signaling. CDA inhibitor tetrahydrouridine resensitized V1-expressing cells to gemcitabine. While gemcitabine (25-50 mg/kg) inhibited bladder cancer xenograft growth, V1-expressing tumors were resistant. Low-dose combination of gemcitabine and tetrahydrouridine abrogated the growth of V1 tumors with minimal toxicity. CONCLUSIONS V1/Chase drives gemcitabine resistance and potentially predicts gemcitabine + cisplatin failure. CDA inhibition resensitizes V1-expressing tumors to gemcitabine. Because several chemotherapy regimens include gemcitabine, our study could have broad significance.
Collapse
Affiliation(s)
- Sarrah L Hasanali
- Departments of Biochemistry and Molecular Biology, Augusta University, Augusta, Georgia
| | - Daley S Morera
- Departments of Biochemistry and Molecular Biology, Augusta University, Augusta, Georgia
| | - Ronny R Racine
- Department of Urology, University of Miami-Miller School of Medicine, Miami, Florida
| | - Martin Hennig
- Department of Urology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Santu Ghosh
- Department of Population Health Sciences, Augusta University, Augusta, Georgia
| | - Luis E Lopez
- Departments of Biochemistry and Molecular Biology, Augusta University, Augusta, Georgia
| | - Marie C Hupe
- Department of Urology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Diogo O Escudero
- Molecular Cell and Developmental Biology Graduate Program, University of Miami-Miller School of Medicine, Miami, Florida
| | - Jiaojiao Wang
- Departments of Biochemistry and Molecular Biology, Augusta University, Augusta, Georgia
| | - Huabin Zhu
- Departments of Biochemistry and Molecular Biology, Augusta University, Augusta, Georgia
| | - Semih Sarcan
- Department of Urology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Ijeoma Azih
- Clinical Trials Office, Augusta University, Augusta, Georgia
| | - Michael Zhou
- Departments of Biochemistry and Molecular Biology, Augusta University, Augusta, Georgia
| | - Andre R Jordan
- Departments of Biochemistry and Molecular Biology, Augusta University, Augusta, Georgia
| | - Martha K Terris
- Surgery, Division of Urology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Markus A Kuczyk
- Department of Urology and Urologic Oncology, Hannover Medical School, Hannover, Germany
| | - Axel S Merseburger
- Department of Urology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Vinata B Lokeshwar
- Departments of Biochemistry and Molecular Biology, Augusta University, Augusta, Georgia.
| |
Collapse
|
56
|
Sundar S, Symonds P. Re: Syed A. Hussain, Nuria Porta, Emma Hall, et al. Outcomes in Patients with Muscle-invasive Bladder Cancer Treated with Neoadjuvant Chemotherapy Followed by (Chemo)radiotherapy in the BC2001 Trial. Eur Urol 2021;79:307-15. Eur Urol 2021; 80:e49-e50. [PMID: 34024651 DOI: 10.1016/j.eururo.2021.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/11/2021] [Indexed: 11/26/2022]
Affiliation(s)
| | - Paul Symonds
- Leicester Cancer Research Centre, University of Leicester, Leicester Royal Infirmary, Leicester, UK
| |
Collapse
|
57
|
Wilkins A, Ost P, Sundahl N. Is There a Benefit of Combining Immunotherapy and Radiotherapy in Bladder Cancer? Clin Oncol (R Coll Radiol) 2021; 33:407-414. [PMID: 33726945 DOI: 10.1016/j.clon.2021.02.014] [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: 02/01/2021] [Revised: 02/10/2021] [Accepted: 02/25/2021] [Indexed: 01/13/2023]
Abstract
Immune checkpoint inhibitors have transformed the management of patients with metastatic urothelial cancer, by leading to long-term response and prolongation of survival in a subset of patients. Unfortunately, only one in five patients with metastatic urothelial cancer responds to anti-programmed death ligand-1 ([AQ1]anti-PD-1) monotherapy. Preclinical and early clinical evidence indicates that radiotherapy not only acts locally, but also exerts systemic anti-tumour effects by modulating the immune system. It is hypothesised that combining checkpoint inhibitors with radiotherapy might enhance an anti-tumour immune response and increase response rates. So far, a handful of early phase clinical trials have been performed seeking to answer this question in urothelial cancer patients. The current review summarises the available preclinical and clinical evidence on radiotherapy/immunotherapy combinations in locally advanced and metastatic bladder cancer and suggests future avenues worthy of exploration.
Collapse
Affiliation(s)
- A Wilkins
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London, UK
| | - P Ost
- Department of Radiation Oncology and Experimental Cancer Research, Ghent University Hospital, Ghent, Belgium
| | - N Sundahl
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London, UK; Department of Radiation Oncology and Experimental Cancer Research, Ghent University Hospital, Ghent, Belgium.
| |
Collapse
|
58
|
de Jong FC, Rutten VC, Zuiverloon TCM, Theodorescu D. Improving Anti-PD-1/PD-L1 Therapy for Localized Bladder Cancer. Int J Mol Sci 2021; 22:2800. [PMID: 33802033 PMCID: PMC7998260 DOI: 10.3390/ijms22062800] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 02/25/2021] [Accepted: 03/02/2021] [Indexed: 02/06/2023] Open
Abstract
In high-risk non-muscle invasive bladder cancer (HR-NMIBC), patient outcome is negatively affected by lack of response to Bacillus-Calmette Guérin (BCG) treatment. Lack of response to cisplatin-based neoadjuvant chemotherapy and cisplatin ineligibility reduces successful treatment outcomes in muscle-invasive bladder cancer (MIBC) patients. The effectiveness of PD-1/PD-L1 immune checkpoint inhibitors (ICI) in metastatic disease has stimulated its evaluation as a treatment option in HR-NMIBC and MIBC patients. However, the observed responses, immune-related adverse events and high costs associated with ICI have provided impetus for the development of methods to improve patient stratification, enhance anti-tumorigenic effects and reduce toxicity. Here, we review the challenges and opportunities offered by PD-1/PD-L1 inhibition in HR-NMIBC and MIBC. We highlight the gaps in the field that need to be addressed to improve patient outcome including biomarkers for response stratification and potentially synergistic combination therapy regimens with PD-1/PD-L1 blockade.
Collapse
Affiliation(s)
- Florus C. de Jong
- Department of Urology, Erasmus University Medical Center, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands; (F.C.d.J.); (V.C.R.); (T.C.M.Z.)
| | - Vera C. Rutten
- Department of Urology, Erasmus University Medical Center, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands; (F.C.d.J.); (V.C.R.); (T.C.M.Z.)
| | - Tahlita C. M. Zuiverloon
- Department of Urology, Erasmus University Medical Center, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands; (F.C.d.J.); (V.C.R.); (T.C.M.Z.)
| | - Dan Theodorescu
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Departments of Surgery (Urology) and Pathology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
59
|
Tripathi A, Khaki AR, Grivas P. Perioperative Immunotherapy in Muscle-invasive Bladder Cancer. Eur Urol Oncol 2021; 4:131-133. [PMID: 33642222 DOI: 10.1016/j.euo.2021.01.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 01/08/2021] [Indexed: 11/15/2022]
Abstract
Immune checkpoint inhibitors (ICIs) have already been approved for the treatment of metastatic urothelial carcinoma and are now being investigated for perioperative treatment of muscle-invasive bladder cancer (MIBC). Ongoing trials are assessing ICIs as monotherapy and in combination with other treatments. Early data are promising, and long-term survival data are awaited to confirm the potential of ICIs in MIBC.
Collapse
Affiliation(s)
- Abhishek Tripathi
- Department of Medicine, Section of Hematology Oncology, University of Oklahoma Stephenson Cancer Center, Oklahoma City, OK, USA
| | - Ali Raza Khaki
- Department of Medicine, Division of Medical Oncology, University of Washington, Fred Hutchinson Cancer Research Center, Seattle Cancer Care Alliance, Seattle, WA, USA
| | - Petros Grivas
- Department of Medicine, Division of Medical Oncology, University of Washington, Fred Hutchinson Cancer Research Center, Seattle Cancer Care Alliance, Seattle, WA, USA.
| |
Collapse
|
60
|
Montorsi F, Martini A. Re: Mathieu Rouanne, Dean F. Bajorin, Raquibul Hannan, et al. Rationale and Outcomes for Neoadjuvant Immunotherapy in Urothelial Carcinoma of the Bladder. Eur Urol Oncol 2020;3:728-38. Eur Urol Oncol 2021; 4:336. [PMID: 33541839 DOI: 10.1016/j.euo.2021.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 01/05/2021] [Accepted: 01/11/2021] [Indexed: 11/15/2022]
Affiliation(s)
- Francesco Montorsi
- Unit of Urology, University Vita-Salute, San Raffaele Scientific Institute, Milan, Italy; Division of Experimental Oncology, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Alberto Martini
- Unit of Urology, University Vita-Salute, San Raffaele Scientific Institute, Milan, Italy; Division of Experimental Oncology, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
61
|
Rey-Cárdenas M, Guerrero-Ramos F, Gómez de Liaño Lista A, Carretero-González A, Bote H, Herrera-Juárez M, Carril-Ajuria L, Martín-Soberón M, Sepulveda JM, Billalabeitia EG, Castellano D, de Velasco G. Recent advances in neoadjuvant immunotherapy for urothelial bladder cancer: What to expect in the near future. Cancer Treat Rev 2021; 93:102142. [PMID: 33453566 DOI: 10.1016/j.ctrv.2020.102142] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 12/13/2020] [Accepted: 12/16/2020] [Indexed: 11/18/2022]
Abstract
Urothelial bladder cancer (UC) is the most common malignancy involving the urinary system and represents a significant health problem. Immunotherapy has been used for decades for UC with intravesical bacillus Calmette-Guérin (BCG) set as the standard of care for non-muscle-invasive bladder cancer (NMIBC). The advent of immune checkpoint inhibitors (ICIs) has completely transformed the treatment landscape of bladder cancer enabling to expand the treatment strategies. Novel ICIs have successfully shown improved outcomes on metastatic disease to such an extent that the standard of care paradigm has changed leading to the development of different trials with the aim of determining whether ICIs may have a role in early disease. The localized muscle-invasive bladder cancer (MIBC) scenario remains challenging since the recurrence rate continues to be high despite all therapeutic efforts. This article will review the current experience of ICIs in the neoadjuvant setting of UC, the clinical trials landscape and finally, an insight of what to expect in the immediate and mid-term future.
Collapse
Affiliation(s)
- M Rey-Cárdenas
- Medical Oncology Department, University Hospital 12 de Octubre, Av. de Córdoba, s/n, 28041 Madrid, Spain.
| | - F Guerrero-Ramos
- Urology Department, University Hospital 12 de Octubre, Av. de Córdoba, s/n, 28041 Madrid, Spain.
| | - A Gómez de Liaño Lista
- Medical Oncology Department, Complejo Hospitalario Universitario Insular-Materno Infantil, Canary Islands, Avda. Marítima del Sur, s/n, 35016 Las Palmas de Gran Canaria, Spain.
| | - A Carretero-González
- Medical Oncology Department, University Hospital 12 de Octubre, Av. de Córdoba, s/n, 28041 Madrid, Spain.
| | - H Bote
- Medical Oncology Department, University Hospital 12 de Octubre, Av. de Córdoba, s/n, 28041 Madrid, Spain.
| | - M Herrera-Juárez
- Medical Oncology Department, University Hospital 12 de Octubre, Av. de Córdoba, s/n, 28041 Madrid, Spain.
| | - L Carril-Ajuria
- Medical Oncology Department, University Hospital 12 de Octubre, Av. de Córdoba, s/n, 28041 Madrid, Spain.
| | - M Martín-Soberón
- Medical Oncology Department, University Hospital 12 de Octubre, Av. de Córdoba, s/n, 28041 Madrid, Spain.
| | - J M Sepulveda
- Medical Oncology Department, University Hospital 12 de Octubre, Av. de Córdoba, s/n, 28041 Madrid, Spain.
| | - E G Billalabeitia
- Medical Oncology Department, University Hospital 12 de Octubre, Av. de Córdoba, s/n, 28041 Madrid, Spain.
| | - D Castellano
- Medical Oncology Department, University Hospital 12 de Octubre, Av. de Córdoba, s/n, 28041 Madrid, Spain.
| | - G de Velasco
- Medical Oncology Department, University Hospital 12 de Octubre, Av. de Córdoba, s/n, 28041 Madrid, Spain.
| |
Collapse
|
62
|
Lopez-Beltran A, Cimadamore A, Blanca A, Massari F, Vau N, Scarpelli M, Cheng L, Montironi R. Immune Checkpoint Inhibitors for the Treatment of Bladder Cancer. Cancers (Basel) 2021; 13:E131. [PMID: 33401585 PMCID: PMC7795541 DOI: 10.3390/cancers13010131] [Citation(s) in RCA: 183] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/28/2020] [Accepted: 12/30/2020] [Indexed: 12/11/2022] Open
Abstract
A number of immune checkpoint inhibitors (ICIs) have been approved as first-line therapy in case of cisplatin-ineligible patients or as second-line therapy for patients with metastatic urothelial carcinoma (mUC) of the bladder. About 30% of patients with mUC will respond to ICIs immunotherapy. Programmed death-ligand 1 (PD-L1) expression detected by immunohistochemistry seems to predict response to immune checkpoint inhibitors in patients with mUC as supported by the objective response rate (ORR) and overall survival (OS) associated with the response observed in most clinical trials. Pembrolizumab, an anti-PD-1 antibody, demonstrated better OS respective to chemotherapy in a randomized phase 3 study for second-line treatment of mUC. Nivolumab, a PD-1 antibody, also demonstrated an OS benefit when compared to controls. Atezolizumab, Durvalumab, and Avelumab antibodies targeting PD-L1 have also received approval as second-line treatments for mUC with durable response for more than 1 year in selected patients. Atezolizumab and Pembrolizumab also received approval for first-line treatment of patients that are ineligible for cisplatin. A focus on the utility of ICIs in the adjuvant or neoadjuvant setting, or as combination with chemotherapy, is the basis of some ongoing trials. The identification of a clinically useful biomarker, single or in association, to determine the optimal ICIs treatment for patients with mUC is very much needed as emphasized by the current literature. In this review, we examined relevant clinical trial results with ICIs in patients with mUC alone or as part of drug combinations; emphasis is also placed on the adjuvant and neoadjuvant setting. The current landscape of selected biomarkers of response to ICIs including anti-PD-L1 immunohistochemistry is also briefly reviewed.
Collapse
Affiliation(s)
- Antonio Lopez-Beltran
- Unit of Anatomic Pathology, Department of Morphological Sciences, Cordoba University Medical School, 14004 Cordoba, Spain
| | - Alessia Cimadamore
- Pathological Anatomy, School of Medicine, United Hospitals, Polytechnic University of the Marche Region, 60126 Ancona, Italy; (A.C.); (M.S.)
| | - Ana Blanca
- Maimonides Biomedical Research Institute of Cordoba, Department of Urology, University Hospital of Reina Sofia, 14004 Cordoba, Spain;
| | - Francesco Massari
- Division of Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Nuno Vau
- Medical Oncology, Champalimaud Clinical Center, 1400-038 Lisbon, Portugal;
| | - Marina Scarpelli
- Pathological Anatomy, School of Medicine, United Hospitals, Polytechnic University of the Marche Region, 60126 Ancona, Italy; (A.C.); (M.S.)
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, School of Medicine, Indiana University, Indianapolis, IN 46202, USA;
| | - Rodolfo Montironi
- Pathological Anatomy, School of Medicine, United Hospitals, Polytechnic University of the Marche Region, 60126 Ancona, Italy; (A.C.); (M.S.)
| |
Collapse
|
63
|
Olmos M, Glajzer J, Büntemeyer TO, Frohwitter G, Ries J, Eckstein M, Hecht M, Lutz R, Kesting MR, Weber M. Neoadjuvant Immunotherapy of Oral Squamous Cell Carcinoma: Case Report and Assessment of Histological Response. Front Oncol 2021; 11:720951. [PMID: 34368002 PMCID: PMC8334861 DOI: 10.3389/fonc.2021.720951] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 07/02/2021] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND The treatment of oral cancer remains challenging due to its infiltrative nature and a high tendency for tumour relapse leading to an overall poor prognosis. In the case of early recurrence, the patient's prognosis deteriorates dramatically, with survival rate dropping to below 30%. Minimal improvements in survival trends in recurrent and advanced stage tumours have been reported in recent decades. Neoadjuvant immunotherapy may represent a new therapeutic approach changing the standard of care in advanced oral cancer therapy. CASE PRESENTATION We describe the case of a woman in her late 30's who presented in mid-2019 with oral squamous cell carcinoma (OSCC) localized to the floor of the mouth. After initial R0 resection, selective neck dissection, and adjuvant brachytherapy, an early recurrence of OSCC located between the hyoid bone and the mandible was diagnosed at the end of 2019. An off-label treatment regimen was performed with neoadjuvant use of Pembrolizumab 19 days prior to salvage surgery. Radiological and histological assessment of T-cell and programmed cell death protein 1 ligand 1 (PD-L1) expression was performed before and after checkpoint inhibitor application. Neoadjuvant immunotherapy resulted in increased T-cell infiltration and PD-L1 expression, as well as a significant tumour necrosis rate. One cycle of Pembrolizumab led to significant regressive tumour changes with increases in immune infiltration, sclerosis, and necrosis of 75% of the tumour mass with only 25% vital tumour cells remaining. By June 2020, the patient remained without recurrence. CONCLUSIONS The case presented outlines the potential effects of neoadjuvant immunotherapy in recurrent or advanced OSCC prior to definitive surgical tumour treatment. The benefit of additional adjuvant treatment after histologic response will be discussed. The case is also analysed considering ongoing clinical trials of neoadjuvant immunotherapy for head and neck malignancies.
Collapse
Affiliation(s)
- Manuel Olmos
- Department of Oral and Maxillofacial Surgery, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany.,Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| | - Jacek Glajzer
- Department of Oral and Maxillofacial Surgery, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany.,Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| | - Tjark-Ole Büntemeyer
- Department of Oral and Maxillofacial Surgery, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany.,Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| | - Gesche Frohwitter
- Department of Oral and Maxillofacial Surgery, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany.,Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| | - Jutta Ries
- Department of Oral and Maxillofacial Surgery, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany.,Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| | - Markus Eckstein
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany.,Institute of Pathology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Markus Hecht
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany.,Department of Radiation Oncology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Rainer Lutz
- Department of Oral and Maxillofacial Surgery, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany.,Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| | - Marco Rainer Kesting
- Department of Oral and Maxillofacial Surgery, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany.,Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| | - Manuel Weber
- Department of Oral and Maxillofacial Surgery, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany.,Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| |
Collapse
|
64
|
Girard A, Vila Reyes H, Dercle L, Rouanne M. "Future role of [18F]-FDG PET/CT in patients with bladder cancer in the new era of neoadjuvant immunotherapy?". Urol Oncol 2020; 39:139-141. [PMID: 33353865 DOI: 10.1016/j.urolonc.2020.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 12/07/2020] [Indexed: 11/28/2022]
Affiliation(s)
- Antoine Girard
- Department of Nuclear Medicine, Centre Eugène Marquis, Université Rennes 1, Rennes, France.
| | - Helena Vila Reyes
- Department of Urology, New York Presbyterian Hospital - Columbia University Medical Center, New York, NY
| | - Laurent Dercle
- Department of Radiology, New York Presbyterian Hospital - Columbia University Medical Center, New York, NY
| | - Mathieu Rouanne
- Department of Urology, Hôpital Foch, Université Versailles-Saint-Quentin-en-Yvelines, Université Paris-Saclay, Suresnes, France
| |
Collapse
|