1
|
Lv J, Zhang Y, Wu Q, Jiang P, Lin Y. Inhibition of SIRT4 promotes bladder cancer progression and immune escape via attenuating CD8 + T cells function. Int Immunopharmacol 2025; 147:113906. [PMID: 39756164 DOI: 10.1016/j.intimp.2024.113906] [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: 11/04/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND Bladder cancer (BCa) is one of the most common malignancies of the urinary system and is characterized by a high recurrence rate and significant mortality. Sirtuin 4 (SIRT4), a member of the NAD+-dependent deacetylase and ADP-ribosyltransferase family, is involved in regulating cellular metabolism, DNA repair, and longevity, potentially influencing tumor progression and immune escape. This study aimed to elucidate the role of SIRT4 in BCa. METHODS The correlation between the sirtuin family and immunotherapy sensitivity in BCa patients was analyzed via IMvigor210 data. The clinical significance and immunological role of SIRT4 across multiple cancer types were assessed by evaluating its associations with clinicopathologic features, prognosis, tumor mutation burden (TMB), microsatellite instability (MSI), immune cell infiltration, and immune response genes across 33 datasets from The Cancer Genome Atlas (TCGA). SIRT4 expression was confirmed in BCa tissues, and its functions were examined via proliferation and migration assays. CD8+ T cells were isolated from the peripheral blood of healthy individuals and activated with CD3 and CD28 antibodies and recombinant IL2. Coculture assays involving BCa cells and activated CD8+ T cells, alongside ELISA, were conducted to evaluate the immunological function of SIRT4. RESULTS SIRT4 was positively associated with the immunotherapy response of BCa patients on the basis of IMvigor210 data. Its expression was downregulated in 11 tumor types but upregulated in 3. SIRT4 was significantly correlated with tumor stage in 2 tumor types and showed varying associations with overall survival, progression-free survival, and disease-specific survival. Additionally, SIRT4 was correlated with TMB in 10 tumor types and with MSI in 8. GSEA indicated that SIRT4 was negatively associated with the immune response in 9 tumor types, excluding BCa. It was positively correlated with immune cell infiltration in 2 tumor types and negatively correlated in 6. The TCGA data revealed that SIRT4 was positively associated with activated NK cell infiltration but negatively associated with M1 macrophages, neutrophils, resting NK cells, and activated memory CD4 T cells. Enrichment analyses revealed positive correlations with various chemokines, immunoinhibitors, immunostimulators, lymphocytes, MHC molecules, and MHC receptors, suggesting that SIRT4 may enhance the immune response in BCa. Further experiments confirmed that SIRT4 was downregulated in BCa tissues compared with adjacent normal tissues. Inhibition of SIRT4 promoted BCa cell proliferation and migration, whereas knockdown of SIRT4 impaired the chemotaxis and tumor-killing ability of CD8+ T cells in the BCa tumor microenvironment. CONCLUSIONS In summary, SIRT4 inhibits the progression and immune escape of BCa, indicating its potential as a novel biomarker and immune checkpoint for immunotherapy.
Collapse
Affiliation(s)
- Jiancheng Lv
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yu Zhang
- Department of Urology, Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang, China
| | - Qikai Wu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Peng Jiang
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Yiwei Lin
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
2
|
Talukder R, Bakaloudi DR, Makrakis D, Diamantopoulos LN, Enright T, Leary JB, Raychaudhuri R, Tripathi N, Agarwal N, Jindal T, Brown JR, Zakharia Y, Rey-Cárdenas M, Castellano D, Nguyen CB, Alva A, Zakopoulou R, Bamias A, Barrera RM, Marmolejo D, Drakaki A, Pinato DJ, Korolewicz J, Buznego LA, Duran I, Carballeira CC, McKay RR, Stewart TF, Gupta S, Barata P, Yu EY, Koshkin VS, Khaki AR, Grivas P. Clinical Outcomes With Immune Checkpoint Inhibitors in Patients With FGFR2/3, MTAP or ERBB2 Genomic Alterations in Advanced Urothelial Carcinoma. Clin Genitourin Cancer 2025; 23:102284. [PMID: 39798390 DOI: 10.1016/j.clgc.2024.102284] [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: 09/12/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 01/15/2025]
Abstract
BACKGROUND FGFR2/3, MTAP and ERBB2 genomic alterations have treatment targets in advanced urothelial carcinoma (aUC). These alterations may affect tumor microenvironment and outcomes with immune checkpoint inhibitors (ICIs) in aUC. PATIENTS AND METHODS We identified patients with available genomic data in our multi-institution cohort of patients with aUC treated with ICI. Outcomes (observed response rate [ORR], progression-free and overall survival [PFS, OS]) with ICI were compared between patients with and without FGFR 2/3, MTAP, ERBB2 alterations. We compared ORR using logistic regression and PFS/OS using Cox proportional hazards. RESULTS Out of 1,514 patients, 276 (18%), 174 (11%) and 208 (14%) patients had known FGFR2/3, MTAP and ERBB2 alteration status, respectively. and were treated with ICI in 1L or 2 + L. In patients with (vs. without) FGFR2/3 alteration, ORR with ICI was 21% vs. 32% (OR 0.54; [95%CI 0.32-0.91]), PFS was significantly shorter in patients with FGFR2/3 alterations (HR = 1.36 [95%CI 1.03-1.80]; P=0.03); OS was not significantly different (HR = 1.22 [95%CI 0.86-1.47]). In patients with (vs. without) MTAP alteration, ORR with ICI was 25% versus 40% (OR 0.52 [95%CI 0.20-1.38]); PFS and OS were nonsignificantly different. In patients with (vs. without) ERBB2 alteration, ORR with ICI was similar (37% vs. 35%; OR 1.06; 95%CI 0.57-1.97); PFS and OS were significantly longer in patients with ERBB2 alteration [HR 0.63 (95%CI 0.41-0.95); P=0.03; HR 0.66, [95% CI 0.44-0.97]), respectively. CONCLUSION Our results support further evaluation of FGFR2/3, MTAP and ERBB2 alterations as putative biomarkers in patients with aUC treated with ICI.
Collapse
MESH Headings
- Humans
- Male
- Immune Checkpoint Inhibitors/therapeutic use
- Female
- Aged
- Receptor, Fibroblast Growth Factor, Type 3/genetics
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Receptor, Fibroblast Growth Factor, Type 2/genetics
- Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors
- Middle Aged
- Urinary Bladder Neoplasms/drug therapy
- Urinary Bladder Neoplasms/genetics
- Urinary Bladder Neoplasms/pathology
- Aged, 80 and over
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Transitional Cell/drug therapy
- Carcinoma, Transitional Cell/genetics
- Carcinoma, Transitional Cell/pathology
- Carcinoma, Transitional Cell/mortality
- Treatment Outcome
- Tumor Microenvironment
- Retrospective Studies
- Mutation
Collapse
Affiliation(s)
- Rafee Talukder
- Department of Medicine, University of Washington, Seattle, WA; Department of Medicine, Section of Hematology and Oncology, Baylor College of Medicine, Houston, TX
| | | | - Dimitrios Makrakis
- Department of Medicine, Jacobi Medical Center-Albert Einstein College of Medicine, Bronx, NY
| | | | - Thomas Enright
- Department of Medicine, University of Washington, Seattle, WA
| | - Jacob B Leary
- Department of Medicine, University of Washington, Seattle, WA
| | | | - Nishita Tripathi
- Division of Oncology, Department of Medicine, University of Utah, Salt Lake City, UT
| | - Neeraj Agarwal
- Division of Oncology, Department of Medicine, University of Utah, Salt Lake City, UT
| | - Tanya Jindal
- Division of Hematology/Oncology, Department of Medicine, Helen Diller Family Cancer Center, University of California San Francisco, San Francisco, CA
| | - Jason R Brown
- Division of Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH
| | - Yousef Zakharia
- Division of Oncology, Department of Medicine, University of Iowa, Iowa City, IA
| | | | - Daniel Castellano
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Charles B Nguyen
- Division of Oncology, Department of Medicine, University of Michigan, Ann Arbor, MI
| | - Ajjai Alva
- Division of Oncology, Department of Medicine, University of Michigan, Ann Arbor, MI
| | - Roubini Zakopoulou
- 2nd Propaedeutic Dept of Internal Medicine, School of Medicine, ATTIKON University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Aristotelis Bamias
- 2nd Propaedeutic Dept of Internal Medicine, School of Medicine, ATTIKON University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Rafael Morales Barrera
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology, Vall d' Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - David Marmolejo
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology, Vall d' Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alexandra Drakaki
- Division of Hematology/Oncology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA
| | - David J Pinato
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, London, UK; Division of Oncology, Department of Translational Medicine (DIMET), University of Piemonte Orientale, Novara, Italy
| | - James Korolewicz
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, London, UK
| | - Lucia Alonso Buznego
- Department of Oncology, University Hospital Marqués of Valdecilla, IDIVAL Santander, Cantabria, Spain
| | - Ignacio Duran
- Department of Oncology, University Hospital Marqués of Valdecilla, IDIVAL Santander, Cantabria, Spain
| | - Clara Castro Carballeira
- Department of Oncology, University Hospital Marqués of Valdecilla, IDIVAL Santander, Cantabria, Spain
| | - Rana R McKay
- Moores Cancer Center, University of California San Diego, La Jolla, CA
| | - Tyler F Stewart
- Moores Cancer Center, University of California San Diego, La Jolla, CA
| | - Shilpa Gupta
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH
| | - Pedro Barata
- Division of Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH
| | - Evan Y Yu
- Department of Medicine, University of Washington, Seattle, WA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Vadim S Koshkin
- Division of Hematology/Oncology, Department of Medicine, Helen Diller Family Cancer Center, University of California San Francisco, San Francisco, CA
| | - Ali Raza Khaki
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA
| | - Petros Grivas
- Department of Medicine, University of Washington, Seattle, WA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA.
| |
Collapse
|
3
|
Wang L, Hu Z, Zhang W, Wang Z, Cao M, Cao X. Promoting macrophage phagocytosis of cancer cells for effective cancer immunotherapy. Biochem Pharmacol 2025; 232:116712. [PMID: 39675588 DOI: 10.1016/j.bcp.2024.116712] [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: 10/02/2024] [Revised: 11/26/2024] [Accepted: 12/10/2024] [Indexed: 12/17/2024]
Abstract
Cancer therapy has been revolutionized by immunotherapeutic agents exploiting adaptive antitumor immunity in the past two decades. However, the overall response rate of these immunotherapies is limited, and patients also develop resistance upon treatment, promoting a rapidly growing exploration of anti-tumor innate immunity for effective cancer therapy. Among these, macrophage immunotherapy through harnessing macrophage phagocytosis has been thrust into the spotlight due to its potential for simultaneously inducing cancer cell killing effect and mobilizing adaptive antitumor responses. Here in this review, we summarize the current macrophage immunotherapy such as therapeutic antibodies, phagocytosis checkpoint blockades, and CAR-macrophages with a particular emphasis on the resistant mechanisms limiting their therapeutic effects. Moreover, we further survey the efforts being placed to seek synergistic mechanisms and combination strategies for promoting macrophage phagocytosis which might stand as next-generation cancer immunotherapy.
Collapse
Affiliation(s)
- Lei Wang
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, and the Engineering Research Center of Cell and Therapeutic Antibody of the Ministry of Education, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai, China
| | - Ziyi Hu
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, and the Engineering Research Center of Cell and Therapeutic Antibody of the Ministry of Education, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai, China
| | - Wencan Zhang
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Zhixin Wang
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, and the Engineering Research Center of Cell and Therapeutic Antibody of the Ministry of Education, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai, China
| | - Ming Cao
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xu Cao
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, and the Engineering Research Center of Cell and Therapeutic Antibody of the Ministry of Education, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
4
|
Klee M, Roesch MC, Eggers H, Ivanyi P, Merseburger AS, Kramer M. Enfortumab vedotin as a salvage option as 5th line therapy for metastatic urothelial bladder cancer. Aktuelle Urol 2025; 56:71-76. [PMID: 37963579 DOI: 10.1055/a-2148-5799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
Abstract
A 67-year-old female patient with a muscle-invasive, non-metastatic urothelial bladder cancer (UC) (pT2 G3 cN0 cM0) developed metachronous metastases within 6 months after radical cystectomy with ileal conduit urinary diversion. After a good primary response to platinum-based chemotherapy, treatment was switched to the immune checkpoint inhibitor (ICI) pembrolizumab due to progressive disease. Subsequently the patient underwent selective internal radiotherapy (SIRT) of the liver and received vinflunine as well as a re-challenge with pembrolizumab. Two years after the initial diagnosis, rapid disease progression ultimately led to a switch to 5th line therapy with enfortumab vedotin (EV), which had only been approved in the United States at that time. The antibody-drug conjugate was well tolerated by the patient after dose reduction to 1.0 mg/ kg body weight. Simultaneous irradiation of newly occurring precardiac, hepatic and cerebral metastases were necessary. After 10 months of therapy with EV, tumour regression was observed accompanied with good symptom control. The presented case illustrates the efficacy and tolerability of EV in a heavily pre-treated patient with metastatic UC (mUC).
Collapse
Affiliation(s)
- Melanie Klee
- Department of Urology, Universitätsklinikum Schleswig-Holstein - Campus Lübeck, Lubeck, Germany
| | - Marie Christine Roesch
- Department of Urology, Universitätsklinikum Schleswig-Holstein - Campus Lübeck, Lubeck, Germany
| | - Hendrik Eggers
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Medizinische Hochschule Hannover, Hannover, Germany
| | | | - Axel S Merseburger
- Department of Urology, Universitätsklinikum Schleswig-Holstein - Campus Lübeck, Lubeck, Germany
| | - Mario Kramer
- Department of Urology, Universitätsklinikum Schleswig-Holstein - Campus Lübeck, Lubeck, Germany
| |
Collapse
|
5
|
Wu Q, Lv J, Li X. Membrane palmitoylated protein MPP1 inhibits immune escape by regulating the USP12/ CCL5 axis in urothelial carcinoma. Int Immunopharmacol 2025; 146:113802. [PMID: 39700963 DOI: 10.1016/j.intimp.2024.113802] [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: 09/10/2024] [Revised: 11/27/2024] [Accepted: 12/04/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND The response rate to immunotherapy in patients with urothelial carcinoma remains limited. Studies have shown that membrane palmitoylated proteins (MPPs) play key roles in tumor progression. However, the mechanisms by which MPP1 regulates immune escape in urothelial carcinoma are not well understood. METHODS The TCGA and BEST databases were used to analyze the associations between the expression of members of the MPP family and the prognosis or immunotherapy sensitivity of urothelial carcinoma patients. MPP1 was identified due to its significant association with survival and immunotherapy sensitivity. MPP1 expression in urothelial carcinoma tissues and cell lines was examined. An MPP1 overexpression vector was used to transfect urothelial carcinoma cells. The functional assays included proliferation, migration, urothelial carcinoma cell-CD8+ T-cell coculture, CD8+ T-cell chemotaxis, and tumorigenesis in human immune reconstitution NOG mice (HuNOG). Bioinformatics, coimmunoprecipitation (CO-IP), mass spectrometry, quantitative real-time polymerase chain reaction (RT-qPCR), and western blotting were used to validate the activity of the MPP1/USP12/CCL5 cascade. RESULTS Analysis of the BEST data revealed that, compared with other MPP family genes, MPP1 was more strongly associated with urothelial carcinoma prognosis and immunotherapy response. Low MPP1 expression was observed in urothelial carcinoma patients and was positively associated with better survival. MPP1 inhibited the proliferation and migration of urothelial carcinoma cells. Bioinformatics, in vitro coculture assays, and in vivo tumorigenesis experiments demonstrated that MPP1 promotes CCL5 production and CD8+ T-cell chemotaxis in the urothelial carcinoma tumor microenvironment (TME). Mechanistically, bioinformatics, mass spectrometry, co-IP, RT-qPCR, and western blot analyses indicated that MPP1 increases CCL5 expression by binding to and promoting USP12. CONCLUSIONS MPP1 significantly inhibits urothelial carcinoma cell proliferation and immune escape via the MPP1/USP12/CCL5 cascade. MPP1 has the potential to serve as a biomarker for guiding immunotherapy in patients with urothelial carcinoma.
Collapse
Affiliation(s)
- Qikai Wu
- Laboratory of Urology and Andrology, Jiangsu Clinical Medicine Research Institution, Nanjing 210029, China
| | - Jiancheng Lv
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| | - Xiaojun Li
- Department of Urology, Taicang Affiliated Hospital of Soochow University, the First People's Hospital of Taicang, Taicang 215400, China.
| |
Collapse
|
6
|
Xu L, Lan T, Huang Y, Wang L, Lin J, Song X, Tang H, Cao H, Chai H. A generative deep neural network for pan-digestive tract cancer survival analysis. BioData Min 2025; 18:9. [PMID: 39871331 PMCID: PMC11771125 DOI: 10.1186/s13040-025-00426-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 01/20/2025] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND The accurate identification of molecular subtypes in digestive tract cancer (DTC) is crucial for making informed treatment decisions and selecting potential biomarkers. With the rapid advancement of artificial intelligence, various machine learning algorithms have been successfully applied in this field. However, the complexity and high dimensionality of the data features may lead to overlapping and ambiguous subtypes during clustering. RESULTS In this study, we propose GDEC, a multi-task generative deep neural network designed for precise digestive tract cancer subtyping. The network optimization process involves employing an integrated loss function consisting of two modules: the generative-adversarial module facilitates spatial data distribution understanding for extracting high-quality information, while the clustering module aids in identifying disease subtypes. The experiments conducted on digestive tract cancer datasets demonstrate that GDEC exhibits exceptional performance compared to other advanced methodologies and can separate different cancer molecular subtypes that possess both statistical and biological significance. Subsequently, 21 hub genes related to pan-DTC heterogeneity and prognosis were identified based on the subtypes clustered by GDEC. The following drug analysis suggested Dasatinib and YM155 as potential therapeutic agents for improving the prognosis of patients in pan-DTC immunotherapy, thereby contributing to the enhancement of cancer patient survival. CONCLUSIONS The experiment indicate that GDEC outperforms better than other deep-learning-based methods, and the interpretable algorithm can select biologically significant genes and potential drugs for DTC treatment.
Collapse
Affiliation(s)
- Lekai Xu
- School of Mathematics, Foshan University, Foshan, 528000, China
| | - Tianjun Lan
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, 510010, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Yiqian Huang
- School of Mathematics, Foshan University, Foshan, 528000, China
| | - Liansheng Wang
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, 510010, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Junqi Lin
- School of Mathematics, Foshan University, Foshan, 528000, China
| | - Xinpeng Song
- School of Mathematics, Foshan University, Foshan, 528000, China
| | - Hui Tang
- School of Mathematics, Foshan University, Foshan, 528000, China
| | - Haotian Cao
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, 510010, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Hua Chai
- School of Mathematics, Foshan University, Foshan, 528000, China.
| |
Collapse
|
7
|
Su H, Peng Y, Wu Y, Zeng X. Overcoming immune evasion with innovative multi-target approaches for glioblastoma. Front Immunol 2025; 16:1541467. [PMID: 39911397 PMCID: PMC11794508 DOI: 10.3389/fimmu.2025.1541467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 01/02/2025] [Indexed: 02/07/2025] Open
Abstract
Glioblastoma (GBM) cells leverage complex endogenous and environmental regulatory mechanisms to drive proliferation, invasion, and metastasis. Tumor immune evasion, facilitated by a multifactorial network, poses a significant challenge to effective therapy, as evidenced by the limited clinical benefits of monotherapies, highlighting the adaptive nature of immune evasion. This review explores glioblastoma's immune evasion mechanisms, the role of ICIs in the tumor microenvironment, and recent clinical advancements, offering theoretical insights and directions for monotherapy and combination therapy in glioblastoma management.
Collapse
Affiliation(s)
- Hai Su
- Department of Neurosurgery, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Yin Peng
- Department of Neurosurgery, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Yilong Wu
- Department of Neurosurgery, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoli Zeng
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Jiangxi “Flagship” Oncology Department of Synergy for Chinese and Western Medicine, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Oncology, Jiangxi Clinical Research Center for Cancer, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
8
|
Qin J, Hu S, Chen Y, Xu M, Xiao Q, Lou J, Ding M, Sun H, Xu T, Pan Y, Wang S. Hypoxia Promotes Malignant Progression of Colorectal Cancer by Inducing POSTN + Cancer-Associated Fibroblast Formation. Mol Carcinog 2025. [PMID: 39835715 DOI: 10.1002/mc.23882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 12/18/2024] [Accepted: 01/04/2025] [Indexed: 01/22/2025]
Abstract
Colorectal cancer (CRC) is one of the most common malignancies. Hypoxia can promote the occurrence and development of CRC. However, how hypoxia regulates the CRC immune microenvironment needs to be further explored. The bulk RNA sequencing data and clinicopathological information of CRC patients were enrolled from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) databases. The single-cell RNA sequencing (scRNA-seq) datasets of CRC were collected from and analyzed from the GEO database and the ArrayExpress database. The score of the hypoxia gene set was estimated using the "ssGSEA" algorithm in the "GSVA" R package. The functional characteristics of CAF subtypes were studied by bioinformatics analysis and in vitro experiments, and a prognostic model was constructed based on machine learning correlation. Hypoxia is associated with poor prognosis in CRC patients. Periostin (POSTN) + Fib is a cancer-associated fibroblast (CAF) closely associated with hypoxia, and high infiltration of POSTN + Fib is associated with adverse outcomes in overall survival (OS) and relapse-free survival (RFS) in CRC patients. Hypoxia can induce POSTN expression and secretion in CAFs. Hypoxia-induced increase of POSTN expression in CAFs can significantly promote the migration and proliferation of CRC cells. Hypoxia-induced increase of POSTN expression in CAFs can significantly promote the proliferation and migration of CRC cells. The POSTN+Fib Hypoxia-Related Risk Model (PFHRM) can predict the survival and immunotherapy response of CRC patients. Our study identified a POSTN+Fib cell subpopulation closely associated with hypoxia, which promotes the malignant progression of CRC. The development of PFHRM provides a theoretical basis for improving patient survival and prognosis.
Collapse
Affiliation(s)
- Jian Qin
- School of Medicine, Southeast University, Nanjing, China
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Shangshang Hu
- School of Medicine, Southeast University, Nanjing, China
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yuhan Chen
- School of Basic Medicine and Clinical Pharmacy, Nanjing First Hospital, China Pharmaceutical University, Nanjing, China
| | - Mu Xu
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Qianni Xiao
- School of Basic Medicine and Clinical Pharmacy, Nanjing First Hospital, China Pharmaceutical University, Nanjing, China
| | - Jinwei Lou
- School of Basic Medicine and Clinical Pharmacy, Nanjing First Hospital, China Pharmaceutical University, Nanjing, China
| | - Muzi Ding
- School of Basic Medicine and Clinical Pharmacy, Nanjing First Hospital, China Pharmaceutical University, Nanjing, China
| | - Huiling Sun
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Tao Xu
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yuqin Pan
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- School of Basic Medicine and Clinical Pharmacy, Nanjing First Hospital, China Pharmaceutical University, Nanjing, China
| | - Shukui Wang
- School of Medicine, Southeast University, Nanjing, China
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- School of Basic Medicine and Clinical Pharmacy, Nanjing First Hospital, China Pharmaceutical University, Nanjing, China
- Jiangsu Collaborative Innovation Center on Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
9
|
Sweis RF, Chatta GS, Jain RK, Moon H, Delacroix SE, Fang A, D’Amico L, Kask AS, Cheever MA, Fling S, Sharon E, Lacroix A, Kaiser JC, Pachynski RK, Yu EY. A Phase II Open-Label, Randomized Clinical Trial of Atezolizumab with or without Human Recombinant IL-7 (CYT107) in Advanced Urothelial Cancer. Clin Cancer Res 2025; 31:299-307. [PMID: 39576210 PMCID: PMC11747792 DOI: 10.1158/1078-0432.ccr-24-1728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/08/2024] [Accepted: 11/19/2024] [Indexed: 01/18/2025]
Abstract
PURPOSE Advanced urothelial cancer generally has high mortality despite modern anti-PD-1/L1 antibody-based combinations. Augmenting checkpoint inhibitor-mediated immune responses with lymphocyte growth factors may improve outcomes. We conducted a randomized phase II study (Cancer Immunotherapy Trials Network-14) in 47 patients to explore whether human recombinant IL-7 (CYT107) could be safely combined with PD-L1 inhibition to enhance responses. PATIENTS AND METHODS Patients with urothelial cancer after platinum chemotherapy were randomized to atezolizumab alone or with CYT107 weekly for four doses. The primary objective was clinical efficacy by the objective response rate (ORR). Secondary objectives included safety, toxicity, and other clinical outcomes. Correlative endpoints included peripheral immunophenotyping and quantification of cytokines. RESULTS CYT107 plus atezolizumab was well-tolerated, without dose-limiting toxicities and lower grade 3 to 4 treatment-related adverse events compared with atezolizumab monotherapy. The ORR was 26.3% for the combination therapy versus 23.8% for atezolizumab alone (P = 0.428). The complete response rate was 10.5% for the combination therapy versus 4.8% for monotherapy. Three patients on combination therapy had responses >21 months versus one with monotherapy. CD4+ and CD8+ T-lymphocyte expansion occurred in patients with response to combination therapy, with the greatest effect in T memory stem cells. Patients who responded to treatment exhibited elevated baseline levels of CCL4 and reduced levels of VEGFA and TNF. CONCLUSIONS Combining CYT107 with atezolizumab was safe and resulted in lymphocyte expansion, a doubling of the complete response rate, and durable responses exceeding 2 years. However, the ORR was similar to atezolizumab alone. Increased and sustained doses of CYT107 coupled with patient selection strategies should be further investigated.
Collapse
Affiliation(s)
- Randy F. Sweis
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL
| | | | | | | | - Scott Edward Delacroix
- Louisiana State University School of Medicine and Stanley S. Scott Cancer Ctr, New Orleans, LA
| | | | | | | | | | | | | | | | | | | | - Evan Y. Yu
- Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington
| |
Collapse
|
10
|
Khandakar H, Kaushal S, Seth A, Sahoo RK, Narwal A, Jangir H, Nayak B, Dinda AK. Comparative evaluation of PD-L1 expression and tumor immune microenvironment in molecular subtypes of muscle-invasive bladder cancer and its correlation with survival outcomes. Am J Clin Pathol 2025:aqae176. [PMID: 39805149 DOI: 10.1093/ajcp/aqae176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025] Open
Abstract
OBJECTIVES Immune checkpoint inhibitors have revolutionized treatment of platinum-refractory advanced bladder cancer, offering hope where options are limited. Response varies, however, influenced by factors such as the tumor's immune microenvironment and prior therapy. Muscle-invasive bladder cancer (MIBC) is stratified into molecular subtypes, with distinct clinicopathologic features affecting prognosis and treatment. This study assessed the expression of programmed cell death 1 ligand 1 (PD-L1) and other immune markers in MIBC, categorized by molecular phenotype. METHODS Using GATA3 and CK5/6 immunohistochemistry, 90 neoadjuvant chemotherapy-naive MIBC cases were classified into luminal and non-luminal subtypes. The immune microenvironment was characterized through immunostaining for PD-L1, CD4, and CD8. We applied PD-L1 positivity thresholds of 1% or greater for tumor cells and 5% or greater for immune cells. Tumors were examined for PD-L1 expression, histologic subtypes, and immune cell infiltration. RESULTS Varied expression of PD-L1 and T-cell subtype densities were observed among MIBC subtypes. The double-negative subtype displayed the highest PD-L1 immune cell expression and stromal CD4 and CD8 T-cell densities, indicating an active immune profile. The basal subtype exhibited the highest PD-L1 positivity in tumor cells. In contrast, the luminal type showed the lowest PD-L1 tumor and immune cell expression, with high intratumoral CD4 T-cell density. Although PD-L1 expression in tumor or immune cells did not independently affect survival, patients with basal and double-negative tumors had poorer overall survival. CONCLUSIONS This study highlighted the immune diversity of MIBC in the context of molecular subtypes. Distinct molecular and immune profiles could guide the development of predictive signatures for enhanced immunotherapy response in advanced bladder cancer.
Collapse
Affiliation(s)
- Hena Khandakar
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Seema Kaushal
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Amlesh Seth
- Department of Urology, All India Institute of Medical Sciences, New Delhi, India
| | - Ranjit K Sahoo
- Department of Medical Oncology (Dr B. R. Ambedkar Institute Rotary Cancer Hospital), All India Institute of Medical Sciences, New Delhi, India
| | - Anubhav Narwal
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Hemlata Jangir
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Brusabhanu Nayak
- Department of Urology, All India Institute of Medical Sciences, New Delhi, India
| | - Amit K Dinda
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
11
|
Ding X, Zhang L, Fan M, Li L. Network-based transfer of pan-cancer immunotherapy responses to guide breast cancer prognosis. NPJ Syst Biol Appl 2025; 11:4. [PMID: 39788975 PMCID: PMC11720706 DOI: 10.1038/s41540-024-00486-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 12/27/2024] [Indexed: 01/12/2025] Open
Abstract
Breast cancer prognosis is complicated by tumor heterogeneity. Traditional methods focus on cancer-specific gene signatures, but cross-cancer strategies that provide deeper insights into tumor homogeneity are rarely used. Immunotherapy, particularly immune checkpoint inhibitors, results from variable responses across cancers, offering valuable prognostic insights. We introduced a network-based transfer (NBT) of pan-cancer immunotherapy responses to enhance breast cancer prognosis using node embedding and heat diffusion algorithms, identifying gene signatures netNE and netHD. Our results showed that netHD and netNE outperformed seven established breast cancer signatures in prognostic metrics, with netHD excelling. All nine gene signatures were grouped into three clusters, with netHD and netNE enriching the immune-related interferon-gamma pathway. Stratifying TCGA patients into two groups based on netHD revealed significant immunological differences and variations in 20 of 50 cancer hallmarks, emphasizing immune-related markers. This approach leverages pan-cancer insights to enhance breast cancer prognosis, facilitating insight transfer and improving tumor homogeneity understanding.Abstract graph of network-based insights translating pan-cancer immunotherapy responses to breast cancer prognosis. This abstract graph illustrates the conceptual framework for transferring immunotherapy response insights from pan-cancer studies to breast cancer prognosis. It highlights the integration of PPI networks to bridge genetic data and clinical phenotypes. The network-based method facilitates the identification of prognostic gene signatures in breast cancer by leveraging immunotherapy response information, providing a novel perspective on tumor homogeneity and its implications for clinical outcomes.
Collapse
Affiliation(s)
- Xiaobao Ding
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou, China
- Institute of Big Data and Artificial Intelligence in Medicine, School of Electronics and Information Engineering, Taizhou University, Taizhou, China
- School of Computer Science and Technology, Hangzhou Dianzi University, Hangzhou, China
| | - Lin Zhang
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou, China
| | - Ming Fan
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou, China.
| | - Lihua Li
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou, China.
- School of Computer Science and Technology, Hangzhou Dianzi University, Hangzhou, China.
| |
Collapse
|
12
|
Fallara G, Belladelli F, Robesti D, Malavaud B, Tholomier C, Mokkapati S, Montorsi F, Dinney CP, Msaouel P, Martini A. Concomitant antihistamine administration is associated with improved survival outcomes in patients with locally advanced or metastatic urothelial carcinoma treated with atezolizumab. Analysis of individual participant data from IMvigor210 and IMvigor211. Urol Oncol 2025:S1078-1439(24)01047-0. [PMID: 39788823 DOI: 10.1016/j.urolonc.2024.12.267] [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: 03/03/2024] [Revised: 10/30/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025]
Abstract
OBJECTIVES Survival outcomes of patients with metastatic urothelial carcinoma (mUC) are still suboptimal and strategies to enhance response to immune-oncology (IO) compounds are under scrutiny. In preclinical studies, it has been demonstrated that antihistamines may reverse macrophage immunosuppression, reactivate T cell cytotoxicity, and enhance the immunotherapy response. We aimed to evaluate the role of concomitant antihistamines administration on oncological outcomes among patients with mUC. MATERIALS AND METHODS We relied on individual patient data from IMvigor210 (phase II single-arm trial on second line atezolizumab for mUC) and IMvigor211 trials (phase III randomized trial on second line atezolizumab vs chemotherapy for mUC). Among individuals treated with IO we identified patients who did and did not receive antihistamines. Multivariable Cox or competing-risks regression models were used to predict progression-free survival (PFS), overall survival (OS), and cancer-specific survival (CSS). The impact of antihistamines on the outcomes was assessed after adjusting for potential confounders. RESULTS Among 896 patients with locally advanced or metastatic urothelial cancer who had progressed after first-line chemotherapy, 155 (17 %) received antihistamines during the delivery of IO. Patients receiving antihistamines had longer OS (Hazard Ratio [HR]:0.59; 95 % Confidence interval [CI]: 0.47-0.74; P < 0.001), PFS (HR:0.70; 95 %CI: 0.57-0.87; P = 0.001) and CSS [sHR:0.58; 95 %CI:0.45-0.75; P < 0.001)] relative to those who had not used antihistamine drugs. A sensitivity analysis, after the exclusion of patients who experienced adverse events and received antihistamines, yielded similar findings of prolonged CSS (sHR 0.78; 95 %CI: 0.59-0.98, P = 0.031) and OS (HR 0.71; 95 %CI: 0.52-0.94, P = 0.021). CONCLUSIONS Concomitant antihistamines administration was associated with improved OS, CSS, and PFS in patients receiving atezolizumab as second line treatment for mUC. Further mechanistic and clinical investigation is warranted to elucidate the role of antihistamines in IO.
Collapse
Affiliation(s)
- Giuseppe Fallara
- Department of Urology, IRCCS European Institute of Oncology, Milan, Italy
| | - Federico Belladelli
- URI - Urological Research Institute, Department of Urology, Division of Experimental Oncology, IRCCS Ospedale San Raffaele, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Daniele Robesti
- URI - Urological Research Institute, Department of Urology, Division of Experimental Oncology, IRCCS Ospedale San Raffaele, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Bernard Malavaud
- Department of Urology, Institut Universitaire du Cancer Toulouse - Oncopôle, Toulouse, France
| | - Côme Tholomier
- Department of Urology, University of Texas, MD Anderson Cancer Center, Houston, TX
| | - Sharada Mokkapati
- Department of Genitourinary Medical Oncology, The University of Texas, MD Anderson Cancer Center, Houston, TX
| | - Francesco Montorsi
- URI - Urological Research Institute, Department of Urology, Division of Experimental Oncology, IRCCS Ospedale San Raffaele, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Colin P Dinney
- Department of Urology, University of Texas, MD Anderson Cancer Center, Houston, TX
| | - Pavlos Msaouel
- Department of Genitourinary Medical Oncology, The University of Texas, MD Anderson Cancer Center, Houston, TX; Department of Translational Molecular Pathology, The University of Texas, MD Anderson Cancer Center, Houston, TX; David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Alberto Martini
- Department of Urology, University of Texas, MD Anderson Cancer Center, Houston, TX; Department of Urology, University of Cincinnati, Cincinnati, Ohio.
| |
Collapse
|
13
|
Zhang K, Wang L, Chen H, Deng L, Hu M, Wang Z, Xie Y, Lian C, Wang X, Zhang J. Integration of single-cell transcriptomics and bulk transcriptomics to explore prognostic and immunotherapeutic characteristics of nucleotide metabolism in lung adenocarcinoma. Front Genet 2025; 15:1466249. [PMID: 39845190 PMCID: PMC11750784 DOI: 10.3389/fgene.2024.1466249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 12/11/2024] [Indexed: 01/24/2025] Open
Abstract
Background Lung adenocarcinoma (LUAD) is a highly aggressive tumor with one of the highest morbidity and mortality rates in the world. Nucleotide metabolic processes are critical for cancer development, progression, and alteration of the tumor microenvironment. However, the effect of nucleotide metabolism on LUAD remains to be thoroughly investigated. Methods Transcriptomic and clinical data of LUAD were downloaded and organized from TCGA and GEO databases. Genes related to nucleotide metabolism were downloaded from the Msigdb database. Genes associated with LUAD prognosis were identified using univariate COX analysis, and a prognostic risk model was constructed using the machine learning combination of Lasso + Stepcox. The model's predictive validity was evaluated using KM survival and timeROC curves. Based on the prognostic model, LUAD patients were classified into different nucleotide metabolism subtypes, and the differences between patients of different subtypes were explored in terms of genomic mutations, functional enrichment, tumor immune characteristics, and immunotherapy responses. Finally, the key gene SNRPA was screened, and a series of in vitro experiments were performed on LUAD cell lines to explore the role of SNRPA in LUAD. Result LUAD patients could be accurately categorized into subtypes based on the nucleotide metabolism-related prognostic risk score (NMBRS). There were significant differences in prognosis between patients of different subtypes, and the NMBRS showed high accuracy in predicting the prognosis of LUAD patients. In addition, patients of different subtypes showed significant differences in genomic mutation and functional enrichment and exhibited different anti-tumor immune profiles. Importantly, NMBRS can be used to predict the responsiveness of LUAD patients to immunotherapy. The results of in vitro cellular experiments indicate that SNRPA plays an important role in the development and progression of lung adenocarcinoma. Conclusion This study comprehensively reveals the prognostic value and clinical application of nucleotide metabolism in LUAD. A prognostic signature constructed based on genes related to nucleotide metabolism accurately predicted the prognosis of LUAD patients, and this signature can be used as a guide for LUAD immunotherapy.
Collapse
Affiliation(s)
- Kai Zhang
- Anhui Province Key Laboratory of Respiratory Tumor and Infectious Disease, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
- Department of Clinical Medicine, Bengbu Medical University, Bengbu, China
| | - Luyao Wang
- Department of Genetics, School of Life Sciences, Bengbu Medical University, Bengbu, China
| | - Huili Chen
- Research Center of Clinical Laboratory Science, Bengbu Medical University, Bengbu, China
| | - Lili Deng
- Anhui Province Key Laboratory of Respiratory Tumor and Infectious Disease, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Mengling Hu
- Department of Genetics, School of Life Sciences, Bengbu Medical University, Bengbu, China
| | - Ziqiang Wang
- Research Center of Clinical Laboratory Science, Bengbu Medical University, Bengbu, China
| | - Yiluo Xie
- Department of Clinical Medicine, Bengbu Medical University, Bengbu, China
| | - Chaoqun Lian
- Research Center of Clinical Laboratory Science, Bengbu Medical University, Bengbu, China
| | - Xiaojing Wang
- Anhui Province Key Laboratory of Respiratory Tumor and Infectious Disease, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
- Joint Research Center for Regional Diseases of IHM, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Jing Zhang
- Department of Genetics, School of Life Sciences, Bengbu Medical University, Bengbu, China
| |
Collapse
|
14
|
Retz M, Grimm MO, Leucht K, Zschäbitz S. [Enfortumab vedotin and pembrolizumab : Management of side effects during first line combined treatment for advanced or metastatic urothelial carcinoma]. UROLOGIE (HEIDELBERG, GERMANY) 2025; 64:60-74. [PMID: 39774681 DOI: 10.1007/s00120-024-02487-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/13/2024] [Indexed: 01/11/2025]
Abstract
The aim of this article is to raise awareness among healthcare providers about the adverse events (AEs) associated with the combined treatment with enfortumab vedotin and pembrolizumab. The differential diagnostic allocation of these AEs to the respective agents is discussed, overlaps between the side effect profiles of the two drugs are identified and strategies for an effective management of these AEs are presented. The recommendations are based on the currently valid prescription information for both drugs, the results of pivotal approval studies and the guidelines of recognized specialist organizations as well as the clinical experience of the authors.
Collapse
MESH Headings
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/administration & dosage
- Humans
- Antibodies, Monoclonal/adverse effects
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal/administration & dosage
- Carcinoma, Transitional Cell/drug therapy
- Carcinoma, Transitional Cell/pathology
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Antineoplastic Agents, Immunological/adverse effects
- Antineoplastic Agents, Immunological/therapeutic use
- Urinary Bladder Neoplasms/drug therapy
- Urinary Bladder Neoplasms/pathology
- Urologic Neoplasms/drug therapy
- Urologic Neoplasms/pathology
Collapse
Affiliation(s)
- Margitta Retz
- Urologische Klinik und Poliklinik, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, München, Deutschland.
| | - Marc-Oliver Grimm
- Klinik für Urologie, Universitätsklinikum Jena, Friedrich-Schiller-Universität, Jena, Deutschland
- Comprehensive Cancer Center Germany (CCCG), Jena, Deutschland
| | - Katharina Leucht
- Klinik für Urologie, Universitätsklinikum Jena, Friedrich-Schiller-Universität, Jena, Deutschland
- Comprehensive Cancer Center Germany (CCCG), Jena, Deutschland
| | - Stefanie Zschäbitz
- Medizinische Klinik VI, Nationales Zentrum für Tumorerkrankungen (NCT), Universitätsklinikum Heidelberg, Heidelberg, Deutschland
| |
Collapse
|
15
|
Bansal K, Chaudhary N, Bhati H, Singh V. Unveiling FDA-approved Drugs and Formulations in the Management of Bladder Cancer: A Review. Curr Pharm Biotechnol 2025; 26:48-62. [PMID: 38797905 DOI: 10.2174/0113892010314650240514053735] [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: 03/12/2024] [Revised: 04/22/2024] [Accepted: 04/29/2024] [Indexed: 05/29/2024]
Abstract
Urological cancers are one of the most prevalent malignancies around the globe. Specifically, bladder cancer severely threatens the health of humans because of its heterogeneous and aggressive nature. Extensive studies have been conducted for many years in order to address the limitations associated with the treatment of solid tumors with selective substances. This article aims to provide a summary of the therapeutic drugs that have received FDA approval or are presently in the testing phase for use in the prevention or treatment of bladder cancer. In this review, FDA-approved drugs for bladder cancer treatment have been listed along with their dose protocols, current status, pharmacokinetics, action mechanisms, and marketed products. The article also emphasizes the novel preparations of these drugs that are presently under clinical trials or are in the approval stage. Thus, this review will serve as a single point of reference for scientists involved in the formulation development of these drugs.
Collapse
Affiliation(s)
- Keshav Bansal
- Institute of Pharmaceutical Research, GLA University, Mathura-281406, Uttar Pradesh, India
| | - Neeraj Chaudhary
- Institute of Pharmaceutical Research, GLA University, Mathura-281406, Uttar Pradesh, India
| | - Hemant Bhati
- Institute of Pharmaceutical Research, GLA University, Mathura-281406, Uttar Pradesh, India
| | - Vanshita Singh
- Institute of Pharmaceutical Research, GLA University, Mathura-281406, Uttar Pradesh, India
| |
Collapse
|
16
|
Huang X, Sun C, Zhang P, Wang L. Programmed cell death 1 inhibitor alone or combined with chemotherapy for patients with locally advanced or metastatic urothelial carcinoma: a single-center experience. BMC Urol 2024; 24:289. [PMID: 39741257 DOI: 10.1186/s12894-024-01674-7] [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: 06/15/2024] [Accepted: 12/11/2024] [Indexed: 01/02/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) alone or in combination with standard chemotherapy for advanced urothelial carcinoma (UC) have been tested as first-line treatment in clinical trials. This study aimed to evaluate the clinical outcomes of programmed cell death 1 (PD-1) inhibitor alone or combined with chemotherapy for patients with locally advanced or metastatic UC in a real world clinical care setting, and sought to identify prognostic factors for overall survival (OS). METHODS A retrospective, real-world study involving 35 locally advanced or metastatic UC patients treated with PD-1 inhibitor alone or in combination with chemotherapy was conducted. Kaplan-Meier curves were used to assess progression-free survival (PFS) and OS. A Cox regression analysis was conducted to explore the association of baseline variables with OS. RESULTS In our cohort of 35 patients, 7 patients were treated with PD-1 inhibitor alone and 28 with PD-1 inhibitor plus platinum-based chemotherapy. The median OS was 16.0 months (95% CI: 11.9-20.1), and median PFS was 12.0 months (95% CI: 8.6-15.4) for all patients. PD-1 inhibitor combined with chemotherapy was associated with better PFS than PD-1 inhibitor monotherapy (HR: 0.19, p = 0.018). Treatment-related adverse events (AEs) of any grade occurred in 5 (71.4%) patients who received PD-1 inhibitor and 24 (85.7%) patients who received PD-1 inhibitor plus chemotherapy. Eastern Cooperative Oncology Group (ECOG) performance status (PS) and neutrophil-lymphocyte ratio (NLR) were identified as prognostic factors. CONCLUSION This study suggested that patients with locally advanced or metastatic UC could benefit from PD-1 inhibitor alone or combined with chemotherapy in daily clinical practice. ECOG PS and NLR can be used for prognostication of survival.
Collapse
Affiliation(s)
- Xing Huang
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chupeng Sun
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Peng Zhang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Lei Wang
- Department of Radiation Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| |
Collapse
|
17
|
Bai Z, Wang H, Han J, An J, Yang Z, Mo X. Multiomics integration and machine learning reveal prognostic programmed cell death signatures in gastric cancer. Sci Rep 2024; 14:31060. [PMID: 39730893 DOI: 10.1038/s41598-024-82233-w] [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: 07/13/2024] [Accepted: 12/03/2024] [Indexed: 12/29/2024] Open
Abstract
Gastric cancer (GC) is characterized by notable heterogeneity and the impact of molecular subtypes on treatment and prognosis. The role of programmed cell death (PCD) in cellular processes is critical, yet its specific function in GC is underexplored. This study applied multiomics approaches, integrating transcriptomic, epigenetic, and somatic mutation data, with consensus clustering algorithms to classify GC molecular subtypes and assess their biological and immunological features. A machine learning model was developed to create the Gastric Cancer Multi-Omics Programmed Cell Death Signature (GMPS), targeting PCD-related genes. We verified the expression of the GMPS hub genes using the RT-qPCR method. The prognostic influence of GMPS on GC was then evaluated. Single-cell analysis was performed to examine the heterogeneity of PCD characteristics in GC. Findings indicate that GMPS notably correlates with patient survival rates, tumor mutational burden (TMB), and copy number variations (CNV), demonstrating substantial prognostic predictive power. Moreover, GMPS is closely associated with the tumor microenvironment (TME) and immune therapy response. This research elucidates the molecular subtypes of GC, highlighting PCD's critical role in prognosis assessment. The relationship between GMPS and immune therapy response, alongside gastric cancer's microenvironmental features, provides insights for personalized treatment.
Collapse
Affiliation(s)
- Zihao Bai
- Clinical Teaching Hospital of Medical School, Nanjing Children's Hospital, Nanjing University, Nanjing, 210008, China
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Hao Wang
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Jingru Han
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, 510182, China
| | - Jia An
- Clinical Teaching Hospital of Medical School, Nanjing Children's Hospital, Nanjing University, Nanjing, 210008, China
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Zhaocong Yang
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China.
| | - Xuming Mo
- Clinical Teaching Hospital of Medical School, Nanjing Children's Hospital, Nanjing University, Nanjing, 210008, China.
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China.
| |
Collapse
|
18
|
Eshaq AM, Flanagan TW, Ba Abbad AA, Makarem ZAA, Bokir MS, Alasheq AK, Al Asheikh SA, Almashhor AM, Binyamani F, Al-Amoudi WA, Bawzir AS, Haikel Y, Megahed M, Hassan M. Immune Checkpoint Inhibitor-Associated Cutaneous Adverse Events: Mechanisms of Occurrence. Int J Mol Sci 2024; 26:88. [PMID: 39795946 PMCID: PMC11719825 DOI: 10.3390/ijms26010088] [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: 11/17/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 01/13/2025] Open
Abstract
Immunotherapy, particularly that based on blocking checkpoint proteins in many tumors, including melanoma, Merkel cell carcinoma, non-small cell lung cancer (NSCLC), triple-negative breast (TNB cancer), renal cancer, and gastrointestinal and endometrial neoplasms, is a therapeutic alternative to chemotherapy. Immune checkpoint inhibitor (ICI)-based therapies have the potential to target different pathways leading to the destruction of cancer cells. Although ICIs are an effective treatment strategy for patients with highly immune-infiltrated cancers, the development of different adverse effects including cutaneous adverse effects during and after the treatment with ICIs is common. ICI-associated cutaneous adverse effects include mostly inflammatory and bullous dermatoses, as well as severe cutaneous side reactions such as rash or inflammatory dermatitis encompassing erythema multiforme; lichenoid, eczematous, psoriasiform, and morbilliform lesions; and palmoplantar erythrodysesthesia. The development of immunotherapy-related adverse effects is a consequence of ICIs' unique molecular action that is mainly mediated by the activation of cytotoxic CD4+/CD8+ T cells. ICI-associated cutaneous disorders are the most prevalent effects induced in response to anti-programmed cell death 1 (PD-1), anti-cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4), and anti-programmed cell death ligand 1 (PD-L1) agents. Herein, we will elucidate the mechanisms regulating the occurrence of cutaneous adverse effects following treatment with ICIs.
Collapse
Affiliation(s)
- Abdulaziz M. Eshaq
- Department of Epidemiology and Biostatstics, Milken Institute School of Public Health, George Washington University Washington, Washington, DC 20052, USA;
- Research Laboratory of Surgery-Oncology, Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Thomas W. Flanagan
- Department of Pharmacology and Experimental Therapeutics, LSU Health Sciences Center, New Orleans, LA 70112, USA;
| | - Abdulqader A. Ba Abbad
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.A.B.A.); (Z.A.A.M.); (M.S.B.); (A.K.A.); (A.M.A.); (F.B.); (W.A.A.-A.)
| | - Zain Alabden A. Makarem
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.A.B.A.); (Z.A.A.M.); (M.S.B.); (A.K.A.); (A.M.A.); (F.B.); (W.A.A.-A.)
| | - Mohammed S. Bokir
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.A.B.A.); (Z.A.A.M.); (M.S.B.); (A.K.A.); (A.M.A.); (F.B.); (W.A.A.-A.)
| | - Ahmed K. Alasheq
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.A.B.A.); (Z.A.A.M.); (M.S.B.); (A.K.A.); (A.M.A.); (F.B.); (W.A.A.-A.)
| | - Sara A. Al Asheikh
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.A.B.A.); (Z.A.A.M.); (M.S.B.); (A.K.A.); (A.M.A.); (F.B.); (W.A.A.-A.)
| | - Abdullah M. Almashhor
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.A.B.A.); (Z.A.A.M.); (M.S.B.); (A.K.A.); (A.M.A.); (F.B.); (W.A.A.-A.)
| | - Faroq Binyamani
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.A.B.A.); (Z.A.A.M.); (M.S.B.); (A.K.A.); (A.M.A.); (F.B.); (W.A.A.-A.)
| | - Waleed A. Al-Amoudi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.A.B.A.); (Z.A.A.M.); (M.S.B.); (A.K.A.); (A.M.A.); (F.B.); (W.A.A.-A.)
| | - Abdulaziz S. Bawzir
- Department of Radiology, King Saud Medical City, Riyadh 11533, Saudi Arabia;
| | - Youssef Haikel
- Institut National de la Santé et de la Recherche Médicale, University of Strasbourg, 67000 Strasbourg, France;
- Department of Operative Dentistry and Endodontics, Dental Faculty, University of Strasbourg, 67000 Strasbourg, France
- Pôle de Médecine et Chirurgie Bucco-Dentaire, Hôpital Civil, Hôpitaux Universitaire de Strasbourg, 67000 Strasbourg, France
| | - Mossad Megahed
- Clinic of Dermatology, University Hospital of Aachen, 52074 Aachen, Germany;
| | - Mohamed Hassan
- Research Laboratory of Surgery-Oncology, Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Institut National de la Santé et de la Recherche Médicale, University of Strasbourg, 67000 Strasbourg, France;
- Department of Operative Dentistry and Endodontics, Dental Faculty, University of Strasbourg, 67000 Strasbourg, France
| |
Collapse
|
19
|
Mohamedali KA, Aguirre B, Lu CH, Chandla A, Kejriwal N, Liu L, Chan AM, Cheung LH, Kok S, Duarte S, Alvarez de Cienfuegos A, Casero D, Rosenblum MG, Wadehra M. GrB-Fc-KS49, an anti-EMP2 granzyme B fusion protein therapeutic alters immune cell infiltration and suppresses breast cancer growth. J Immunother Cancer 2024; 12:e008891. [PMID: 39794935 PMCID: PMC11667298 DOI: 10.1136/jitc-2024-008891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 11/25/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Granzyme B (GrB) is a key effector molecule, delivered by cytotoxic T lymphocytes and natural killer cells during immune surveillance to induce cell death. Fusion proteins and immunoconjugates represent an innovative therapeutic approach to specifically deliver a deadly payload to target cells. Epithelial membrane protein-2 (EMP2) is highly expressed in invasive breast cancer (BC), including triple-negative BC (TNBC), and represents an attractive therapeutic target. METHODS We designed a novel fusion protein (GrB-Fc-KS49) composed of an active GrB fused to an anti-EMP2 single-chain antibody tethered through the immunoglobulin G heavy chain (Fc) domain. We assessed the construct's GrB enzymatic activity, anti-EMP2 binding affinity, and cytotoxicity against a panel of BC cells. The construct's pharmacokinetics (PK), toxicity profile, and in vivo efficacy were also evaluated. RESULTS GrB-Fc-KS49 exhibited comparable GrB enzymatic activity to commercial GrB, as well as high affinity to an EMP2 peptide, with the dissociation constant in the picomolar range. The fusion protein rapidly internalized into EMP2+cancer cells and showed in vitro cytotoxicity to cell lines expressing surface EMP2, with half-maximal cytotoxicity (IC50) values below 100 nM for most positive lines. Ex vivo stability at 37°C indicated a half-life exceeding 96 hours while in vivo PK indicated a biexponential plasma clearance, with a moderate initial clearance (t1/2α=18.4 hours) and a much slower terminal clearance rate (t1/2β=73.1 hours). No toxicity was measured in a Chem16 panel between the control and the GrB-Fc-KS49. In vivo, the GrB-Fc-KS49 showed efficacy against a TNBC syngeneic (4T1/FLuc) mouse model, reducing tumor volume and cell proliferation and increasing cell death compared with controls. Treatment using an EMT6 mouse model confirmed these results. In addition to a significant impact on cell proliferation, GrB-Fc-KS49 treatment also resulted in a dramatic increase of tumor-infiltrating CD45+ cells and redistribution of tumor-associated macrophages. Transcriptomic analysis of tumors post-treatment confirmed the remodeling of the immune tumor microenvironment by the GrB-Fc-KS49 immunotoxin. CONCLUSIONS GrB-Fc-KS49 showed high specificity and cytotoxicity towards EMP2-positive cells. In vivo, it reduced tumor burden and increased the recruitment of immune cells into the tumor, suggesting that GrB-Fc-KS49 is a promising therapeutic candidate against BC.
Collapse
Affiliation(s)
- Khalid A Mohamedali
- Experimental Therapeutics, University of Texas MD Anderson Cancer Center Division of Cancer Medicine, Houston, Texas, USA
| | - Brian Aguirre
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Cheng-Hsiang Lu
- Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Anubhav Chandla
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Nidhi Kejriwal
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Lucia Liu
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Ann M Chan
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Lawrence H Cheung
- Experimental Therapeutics, University of Texas MD Anderson Cancer Center Division of Cancer Medicine, Houston, Texas, USA
| | - SuYin Kok
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Sergio Duarte
- Department of Surgery, University of California Los Angeles, Los Angeles, California, USA
| | - Ana Alvarez de Cienfuegos
- Experimental Therapeutics, University of Texas MD Anderson Cancer Center Division of Cancer Medicine, Houston, Texas, USA
| | - David Casero
- Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Michael G Rosenblum
- Experimental Therapeutics, University of Texas MD Anderson Cancer Center Division of Cancer Medicine, Houston, Texas, USA
| | - Madhuri Wadehra
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California, USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
20
|
Zhao Y, Wucherpfennig KW. Tissue-Resident T Cells in Clinical Response and Immune-Related Adverse Events of Immune Checkpoint Blockade. Clin Cancer Res 2024; 30:5527-5534. [PMID: 39404858 DOI: 10.1158/1078-0432.ccr-23-3296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/09/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024]
Abstract
T-cell surveillance of tissues is spatially organized: circulating memory T cells perform surveillance of secondary lymphoid organs, whereas tissue-resident memory T cells act as sentinels in barrier tissues. In the context of infection, tissue-resident memory T cells survive long term in barrier tissues and are poised to respond to re-encounter of infectious agents. The activity of such tissue-resident T cells is regulated by the PD-1 and cytotoxic T-lymphocyte-associated protein 4 inhibitory receptors targeted by cancer immunotherapies. This review investigates the hypothesis that T cells with a tissue residency program play an important role in both protective antitumor immunity and immune-related adverse events (irAE) of immune checkpoint blockade (ICB). A series of translational studies have demonstrated that a higher density of tissue-resident T cells within tumors is associated with favorable survival outcomes in a diverse range of cancer types. Tissue-resident T cells have also been implicated in clinical response to ICB, and dynamic tracking of T-cell populations in pre- and on-treatment tumor samples demonstrated that T cells with a tissue residency program responded early to ICB. Investigation of colitis and dermatitis as examples of irAEs demonstrated that tissue-resident memory T cells were reactivated at these epithelial sites, resulting in a highly cytotoxic state and secretion of inflammatory cytokines IFNγ and TNFα. It will therefore be important to consider how a tissue residency program can be enhanced to promote T-cell-mediated tumor immunity while preventing the development of irAEs.
Collapse
Affiliation(s)
- Ye Zhao
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kai W Wucherpfennig
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Immunology, Harvard Medical School, Boston, Massachusetts
- Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts
| |
Collapse
|
21
|
Cheng LY, Su PJ, Kuo MC, Lin CT, Luo HL, Chou CC, Huang SY, Wu CC, Chen CH, Huang CC, Tsai KL, Yu-Li Su H. Combining serum inflammatory markers and clinical factors to predict survival in metastatic urothelial carcinoma patients treated with immune checkpoint inhibitors. Ther Adv Med Oncol 2024; 16:17588359241305091. [PMID: 39687055 PMCID: PMC11648016 DOI: 10.1177/17588359241305091] [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: 07/20/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Background Despite the revolutionary impact of immune checkpoint inhibitors (ICIs) on the treatment of metastatic urothelial carcinoma (mUC), the clinical utility of reliable prognostic biomarkers to foresee survival outcomes remains underexplored. Objectives The purpose of this study was to ascertain the prognostic significance of serum inflammatory markers in mUC patients undergoing ICI therapy. Design This is a retrospective, multicenter study. Methods Data were collected from two independent medical centers in Taiwan, encompassing a validation and a training cohort (TC). Patients with histopathologically confirmed urothelial carcinoma who received at least one cycle of ICI monotherapy were included. Serum inflammatory markers such as neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and systemic immune-inflammation index (SII) were calculated prior to ICI therapy. Statistical analyses involved the use of receiver operating characteristic (ROC) curves to determine optimal biomarker cutoffs and Cox proportional hazards models to evaluate the independent predictive capability of these markers. Results A total of 192 patients were enrolled. In the univariate analysis, serum markers such as NLR, PLR, SII, and Hb were significantly associated with overall survival (OS) in both the training and validation cohorts (VC). White blood cells, NLR, and SII demonstrated a robust correlation with progression-free survival across both cohorts. Multivariate analysis revealed that Eastern Cooperative Oncology Group performance status ⩾2 (p < 0.001), visceral metastasis (p < 0.001), leukocytosis (p < 0.001), Hb levels ⩾10 mg/dL (p = 0.008), and NLR ⩾5 (p = 0.032) as independent predictors of OS. A prognostic nomogram integrating these independent factors yielded a C-index for a 3-year OS of 0.769 in the TC and 0.657 in the VC. Conclusion Serum inflammatory markers, combined with clinicopathologic factors, provide a practical prognostic tool in mUC treatment with ICIs.
Collapse
Affiliation(s)
- Liang-Yun Cheng
- Division of Hematology–Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Po-Jung Su
- Division of Hematology–Oncology, Chang Gung Memorial Hospital at Linkou and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Ming-Chun Kuo
- Division of Hematology–Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chang-Ting Lin
- Division of Hematology–Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hao-Lun Luo
- Department of Urology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chih-Chi Chou
- Department of Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Shih-Yu Huang
- Division of Hematology–Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chia-Che Wu
- Division of Hematology–Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chien-Hsu Chen
- Department of Urology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chun-Chieh Huang
- Department of Radiation Oncology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Kai-Lung Tsai
- Department of Colorectal Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Harvey Yu-Li Su
- Division of Hematology–Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, No. 123, Dapi Road, Niaosong District, Kaohsiung City 833, Taiwan
- Genomic and Proteomic Core Laboratory, Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Cancer Center, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
| |
Collapse
|
22
|
Liang W, Huang S, Huang Y, Huang M, Li C, Liang Y, Pang L. Efficacy and safety of first-line immunotherapy-containing regimens compared with chemotherapy for advanced or metastatic urothelial carcinoma: a network meta-analysis of randomized controlled trials. Front Oncol 2024; 14:1453338. [PMID: 39723374 PMCID: PMC11668658 DOI: 10.3389/fonc.2024.1453338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
Introduction To assess the efficacy and safety of first-line immunotherapy-containing regimens compared with chemotherapy for advanced or metastatic urothelial carcinoma (UC). Method A comprehensive search was performed in four databases (Pubmed, Embase, Web of Science, and the Cochrane Library) to identify randomized controlled trials (RCTs) assessing the efficacy of first-line immunotherapy-containing regimens for advanced or metastatic UC. The search encompassed the time span from the inception of the databases to April 23, 2024. A network meta-analysis (NMA) was conducted to assess the rates of progression-free survival (PFS), overall survival (OS), complete response (CR), objective response rate (ORR), and grade ≥ 3 adverse events (AEs). Results We conducted a comprehensive analysis of five randomized controlled trials (RCTs) that included a total of 4749 patients. Nine different treatment regimens included in the study were ranked statistically and intuitively using NMA. The top five effective regimens, ranked by OS, were EV + Pembro (1.000), Nivol + Chemo (0.724), Atezo + Chemo (0.610), Durva + Treme (0.558), and Pembro + Chemo (0.530). The top five effective regimens, ranked by PFS, were EV + Pembro (0.999), Nivol + Chemo (0.640), Pembro + Chemo (0.484), Atezo + Chemo (0.373) and Chemo (0.003). The top five effective regimens, ranked by CR, were EV + Pembro (0.969), Nivol + Chemo (0.803), Atezo + Chemo (0.772), Pembro + Chemo (0.472), Durva + Treme (0.449). The top five effective regimens, ranked by ORR, were EV + Pembro (0.995), Nivol + Chemo (0.852), Pembro + Chemo (0.761), Atezo + Chemo (0.623), and Chemo (0.519). Conclusion Our results indicated that EV + Pembro as first-line therapy resulted in considerably improved efficacy and safety compared to chemotherapy for advanced or metastatic UC. ICI plus chemotherapy as first-line treatment resulted in a longer PFS, a greater ORR, but no longer OS compared to chemotherapy alone, as well as higher toxicity. ICI alone as first-line therapy provided similar OS and lower toxicity compared to chemotherapy, but lower ORR. Systematic review registration https://www.crd.york.ac.uk/prospero, identifier CRD42024538546.
Collapse
Affiliation(s)
- Weiming Liang
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
| | - Shibo Huang
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
| | - Yanping Huang
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
| | - Miaoyan Huang
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
| | - Chunyan Li
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
| | - Yiwen Liang
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
| | - Li Pang
- Medicine Center, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
| |
Collapse
|
23
|
Hamidi H, Senbabaoglu Y, Beig N, Roels J, Manuel C, Guan X, Koeppen H, Assaf ZJ, Nabet BY, Waddell A, Yuen K, Maund S, Sokol E, Giltnane JM, Schedlbauer A, Fuentes E, Cowan JD, Kadel EE, Degaonkar V, Andreev-Drakhlin A, Williams P, Carter C, Gupta S, Steinberg E, Loriot Y, Bellmunt J, Grivas P, Rosenberg J, van der Heijden MS, Galsky MD, Powles T, Mariathasan S, Banchereau R. Molecular heterogeneity in urothelial carcinoma and determinants of clinical benefit to PD-L1 blockade. Cancer Cell 2024; 42:2098-2112.e4. [PMID: 39577421 DOI: 10.1016/j.ccell.2024.10.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 08/15/2024] [Accepted: 10/29/2024] [Indexed: 11/24/2024]
Abstract
Checkpoint inhibitors targeting programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) have revolutionized cancer therapy across many indications including urothelial carcinoma (UC). Because many patients do not benefit, a better understanding of the molecular mechanisms underlying response and resistance is needed to improve outcomes. We profiled tumors from 2,803 UC patients from four late-stage randomized clinical trials evaluating the PD-L1 inhibitor atezolizumab by RNA sequencing (RNA-seq), a targeted DNA panel, immunohistochemistry, and digital pathology. Machine learning identifies four transcriptional subtypes, representing luminal desert, stromal, immune, and basal tumors. Overall survival benefit from atezolizumab over standard-of-care is observed in immune and basal tumors, through different response mechanisms. A self-supervised digital pathology approach can classify molecular subtypes from H&E slides with high accuracy, which could accelerate tumor molecular profiling. This study represents a large integration of UC molecular and clinical data in randomized trials, paving the way for clinical studies tailoring treatment to specific molecular subtypes in UC and other indications.
Collapse
Affiliation(s)
| | | | - Niha Beig
- Genentech, South San Francisco CA, USA
| | | | | | | | | | | | | | | | - Kobe Yuen
- Genentech, South San Francisco CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Yohann Loriot
- Institut de Cancérologie Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Zhang T, Tan A, Shah AY, Iyer G, Morris V, Michaud S, Sridhar SS. Reevaluating the role of platinum-based chemotherapy in the evolving treatment landscape for patients with advanced urothelial carcinoma. Oncologist 2024; 29:1003-1013. [PMID: 39167703 PMCID: PMC11630754 DOI: 10.1093/oncolo/oyae215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 07/17/2024] [Indexed: 08/23/2024] Open
Abstract
Platinum-based chemotherapy has been the standard first-line (1L) treatment for advanced urothelial carcinoma (UC) for decades, based on the proven efficacy and established safety profiles of cisplatin- and carboplatin-based regimens. With the emergence of novel regimens, it is important to reevaluate and contextualize the role of 1L platinum-based chemotherapy. Platinum-based chemotherapy followed by avelumab 1L maintenance in patients without disease progression following platinum-based chemotherapy was established as a standard 1L regimen based on the JAVELIN Bladder 100 phase III trial. More recently, the EV-302 phase III trial showed the superiority of 1L enfortumab vedotin (EV) + pembrolizumab versus platinum-based chemotherapy, and the Checkmate 901 phase III trial showed the superiority of 1L nivolumab + cisplatin/gemcitabine versus cisplatin/gemcitabine alone. These 2 regimens have now been included as standard 1L options in treatment guidelines for advanced UC. EV + pembrolizumab is now the preferred 1L treatment, and in locations where EV + pembrolizumab is not available or individual patients are not considered suitable, recommended options are platinum-based chemotherapy followed by avelumab maintenance or nivolumab + cisplatin-based chemotherapy. In this review, we discuss current treatment options for advanced UC recommended in guidelines, practical considerations with platinum-based chemotherapy, the role of avelumab 1L maintenance, recent phase III trials of EV + pembrolizumab and nivolumab + cisplatin/gemcitabine, safety profiles of recommended 1L treatments, and second-line treatment options.
Collapse
Affiliation(s)
- Tian Zhang
- Department of Internal Medicine, Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, United States
| | - Alan Tan
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Amishi Y Shah
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Gopa Iyer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Valerie Morris
- EMD Serono, Inc., Rockland, MA, United States, an affiliate of Merck KGaA
| | - Sébastien Michaud
- EMD Serono, Inc., Rockland, MA, United States, an affiliate of Merck KGaA
| | - Srikala S Sridhar
- Princess Margaret Cancer Center, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
25
|
Kwon WA, Lee MK. Evolving Treatment Landscape of Frontline Therapy for Metastatic Urothelial Carcinoma: Current Insights and Future Perspectives. Cancers (Basel) 2024; 16:4078. [PMID: 39682263 DOI: 10.3390/cancers16234078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Cisplatin-based chemotherapy has long been the standard first-line (1L) treatment for metastatic urothelial carcinoma (mUC). However, up to 50% of patients with mUC may be ineligible for cisplatin owing to comorbidities, necessitating alternative primary treatment options. Immune checkpoint inhibitors (ICIs) have emerged as a vital alternative for those unable to receive cisplatin. Nevertheless, the prognosis of advanced UC remains dire and challenges persist in optimizing 1L therapy. Recent medical advancements have redirected attention towards innovative drug combinations for the primary treatment of mUC. The combination of enfortumab vedotin (EV) and pembrolizumab has shown significantly improved overall and progression-free survival rates compared to those with chemotherapy alone. This combination can be used as a 1L treatment for patients with mUC who are cisplatin-ineligible or require alternatives to standard chemotherapy. While platinum-based chemotherapy continues to be essential for many patients, the approval of EV and pembrolizumab as 1L treatments for cisplatin-ineligible patients signifies a major breakthrough in primary cancer care. These therapies offer enhanced outcomes in terms of survival and response rates and highlight the increasing relevance of ICI-containing regimens in frontline cancer care. This review provides an exhaustive overview of the current frontline treatment landscape of mUC and explores new therapeutic strategies, with the aim of facilitating clinical decision-making and guiding therapeutic strategies in patients with mUC.
Collapse
Affiliation(s)
- Whi-An Kwon
- Department of Urology, Myongji Hospital, Hanyang University College of Medicine, Goyang-si 10475, Republic of Korea
| | - Min-Kyung Lee
- Department of Internal Medicine, Myongji Hospital, Hanyang University College of Medicine, Goyang-si 10475, Republic of Korea
| |
Collapse
|
26
|
Fléchon A, Morales-Barrera R, Powles T, Alva A, Özgüroğlu M, Csöszi T, Loriot Y, Rodriguez-Vida A, Géczi L, Cheng SY, Fradet Y, Oudard S, Vulsteke C, Gunduz S, Mamtani R, Yu EY, Montesa Pino A, Anido U, Sendur MA, Gravis G, Révész J, Kostorov V, Huillard O, Ma J, Rajasagi M, Vajdi A, Lunceford J, Cristescu R, Imai K, Homet Moreno B, Matsubara N. Association of Tumor Mutational Burden and PD-L1 with the Efficacy of Pembrolizumab with or without Chemotherapy versus Chemotherapy in Advanced Urothelial Carcinoma. Clin Cancer Res 2024; 30:5353-5364. [PMID: 39475359 PMCID: PMC11609623 DOI: 10.1158/1078-0432.ccr-23-3518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/26/2024] [Accepted: 09/18/2024] [Indexed: 12/19/2024]
Abstract
PURPOSE The three-arm, phase III KEYNOTE-361 study did not meet its dual primary endpoints of progression-free survival (PFS) or overall survival (OS) with first-line pembrolizumab plus chemotherapy versus chemotherapy in advanced urothelial carcinoma. This prespecified exploratory analysis assessed the association of tumor mutational burden (TMB) and PD-L1 combined positive score (CPS) with clinical outcomes. PATIENTS AND METHODS TMB and PD-L1 CPS were determined via whole-exome sequencing and PD-L1 IHC 22C3 pharmDx, respectively. The association was evaluated in each treatment arm using logistic regression [objective response rate (ORR)] and Cox proportional hazards regression models (PFS and OS); one-sided (pembrolizumab monotherapy; pembrolizumab plus chemotherapy) and two-sided (chemotherapy) nominal P values were calculated. Significance was prespecified at α = 0.05 without multiplicity adjustment. Efficacy was evaluated by prespecified cutoffs of 175 mutations/exome (TMB) and CPS 10 (PD-L1). RESULTS Of the 993 treated patients, 820 (82.6%) and 993 (100%) had evaluable TMB and CPS data, respectively. Continuous TMB was positively associated with ORR, PFS, and OS for pembrolizumab monotherapy (one-sided P < 0.001, P < 0.001, and P = 0.007, respectively); PFS and OS for pembrolizumab plus chemotherapy (one-sided P = 0.007 and P = 0.010, respectively); and OS for chemotherapy alone (two-sided P = 0.040). Continuous PD-L1 CPS showed evidence of anticipated association with ORR and PFS for pembrolizumab monotherapy. The subgroup with TMB ≥175 mutations/exome and PD-L1 CPS ≥10 had the highest PFS and OS improvements with pembrolizumab alone or with chemotherapy versus chemotherapy alone. CONCLUSIONS These data suggest that TMB may be predictive of the response to pembrolizumab alone or with chemotherapy in advanced urothelial carcinoma.
Collapse
MESH Headings
- Humans
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/therapeutic use
- B7-H1 Antigen/genetics
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Mutation
- Female
- Male
- Aged
- Middle Aged
- Exome Sequencing
- Biomarkers, Tumor/genetics
- Carcinoma, Transitional Cell/drug therapy
- Carcinoma, Transitional Cell/genetics
- Carcinoma, Transitional Cell/pathology
- Carcinoma, Transitional Cell/mortality
- Urologic Neoplasms/drug therapy
- Urologic Neoplasms/genetics
- Urologic Neoplasms/pathology
- Urologic Neoplasms/mortality
- Urinary Bladder Neoplasms/drug therapy
- Urinary Bladder Neoplasms/genetics
- Urinary Bladder Neoplasms/pathology
- Urinary Bladder Neoplasms/mortality
- Antineoplastic Agents, Immunological/therapeutic use
- Aged, 80 and over
Collapse
Affiliation(s)
- Aude Fléchon
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | - Rafael Morales-Barrera
- Vall d’Hebron Institute of Oncology, Vall d´Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Thomas Powles
- Barts Cancer Centre, St Bartholomew’s Hospital, London, United Kingdom
- Barts Cancer Institute, Barts Health NHS Trust, Queen Mary University of London, London, United Kingdom
| | - Ajjai Alva
- University of Michigan Health System, Ann Arbor, Michigan
| | - Mustafa Özgüroğlu
- Division of Medical Oncology, Department of Internal Medicine, Cerrahpaşa School of Medicine, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Tibor Csöszi
- County Oncology Centre, Hetényi Géza Hospital, Szolnok, Hungary
| | - Yohann Loriot
- Institut Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Alejo Rodriguez-Vida
- Medical Oncology Department, Hospital del Mar Research Institute, Barcelona, Spain
| | - Lajos Géczi
- Medical Oncology Center and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, Hungary
| | - Susanna Y. Cheng
- Sunnybrook Health Sciences Centre, Odette Cancer Centre, Toronto, Canada
| | - Yves Fradet
- CHU de Québec-Université Laval, Quebec City, Canada
| | - Stéphane Oudard
- Georges Pompidou European Hospital, University Paris Cité, Paris, France
| | - Christof Vulsteke
- Department of Medical Oncology, Maria Middelares Hospital, Gent, Belgium
- Center for Oncological Research (CORE), University of Antwerp, Antwerp, Belgium
| | - Seyda Gunduz
- Istinye University Liv Hospital, Istanbul, Turkey
- Minimally Invasive Therapeutics Laboratory, Mayo Clinic, Scottsdale, Arizona
| | - Ronac Mamtani
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Evan Y. Yu
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, Washington
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Alvaro Montesa Pino
- UGC Medical Oncology, Hospital Regional Universitario de Málaga, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga, Málaga, Spain
| | - Urbano Anido
- Department of Medical Oncology, Complejo Hospitalario Universitario de Santiago, Santiago de Compostela, Spain
| | - Mehmet A.N. Sendur
- Department of Medical Oncology, Ankara Yıldırım Beyazıt University Faculty of Medicine, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Gwenaelle Gravis
- Department of Medical Oncology, Paoli-Calmettes Institute, Marseille, France
| | - János Révész
- Institute of Radiotherapy and Clinical Oncology, Borsod-Abaúj-Zemplén County Hospital and University Teaching Hospital, Miskolc, Hungary
| | - Vladimir Kostorov
- Leningrad Regional Oncology Dispensary, Ulitsa Savushkina, Saint Petersburg, Russia
| | - Olivier Huillard
- Department of Medical Oncology, Hôpital Cochin, Institut du Cancer Paris, Cancer Research for Personalized Medicine (CARPEM), AP-HP Centre, Université de Paris Cité, Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Rosenberg JE, Galsky MD, Powles T, Petrylak DP, Bellmunt J, Loriot Y, Necchi A, Hoffman-Censits J, Perez-Gracia JL, van der Heijden MS, Dreicer R, Durán I, Castellano D, Drakaki A, Retz M, Sridhar SS, Grivas P, Yu EY, O'Donnell PH, Burris HA, Mariathasan S, Shi Y, Goluboff E, Bajorin D. Atezolizumab monotherapy for metastatic urothelial carcinoma: final analysis from the phase II IMvigor210 trial. ESMO Open 2024; 9:103972. [PMID: 39642637 PMCID: PMC11667038 DOI: 10.1016/j.esmoop.2024.103972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 10/01/2024] [Indexed: 12/09/2024] Open
Abstract
BACKGROUND The IMvigor210 trial demonstrated clinical benefit and manageable toxicity with atezolizumab monotherapy [anti-programmed death-ligand 1 (PD-L1)] in patients with metastatic urothelial carcinoma (UC) in primary analyses. Final efficacy and safety results after long-term follow-up are reported. PATIENTS AND METHODS This phase II single-arm trial of atezolizumab monotherapy in patients with advanced UC included two cohorts: untreated patients ineligible for cisplatin-based chemotherapy (cohort 1; n = 119) and those previously treated with platinum-based chemotherapy (cohort 2; n = 310). Atezolizumab was administered i.v. (1200 mg every 21 days) until progression or unacceptable toxicity. Primary endpoints were independent review facility-assessed confirmed objective response rate (ORR) per RECIST 1.1 in cohort 1 and independent review facility-assessed ORR per RECIST 1.1 and investigator-assessed modified (m)RECIST in cohort 2. Overall survival (OS), efficacy by PD-L1 status, and safety were also assessed. RESULTS At data cut-off (1 June 2023), the median survival follow-up was 96.4 months (range, 0.2-103.4 months) in cohort 1 and 46.2 months [0.2 (censored)-54.9 months] in cohort 2. In cohort 1, the ORR [95% confidence interval (CI)] was 23.5% (16.2% to 32.2%) in all patients and 28.1% (13.8% to 46.8%) in the PD-L1 tumor-infiltrating immune cell (IC)2/3 subgroup. Median OS (95% CI) was 16.3 months (10.4-24.5 months) overall and 12.3 months (6.0-49.8 months) in the PD-L1 IC2/3 subgroup. In cohort 2, the ORR (95% CI) was 16.5% (12.5% to 21.1%) per RECIST 1.1 and 19.7% (95% CI 15.4% to 24.6%) per mRECIST in all patients and 27.0% (18.6% to 36.8%) and 28.0% (19.5% to 37.9%), respectively, in the PD-L1 IC2/3 subgroup. Median OS (95% CI) was 7.9 months (6.7-9.3 months) in all patients and 11.9 months (9.0-22.8 months) in the IC2/3 subgroup. Treatment-related grade 3/4 adverse events occurred in 21.8% (cohort 1) and 18.7% (cohort 2); one treatment-related death occurred in cohort 1. CONCLUSIONS With long-term follow-up, atezolizumab monotherapy demonstrated clinically meaningful efficacy with durable responses in a subset of patients with metastatic UC; there were no new safety signals.
Collapse
Affiliation(s)
- J E Rosenberg
- Memorial Sloan Kettering Cancer Center, New York, USA.
| | - M D Galsky
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - T Powles
- Barts Cancer Institute, Queen Mary University of London ECMC, Barts Health, London, UK
| | | | - J Bellmunt
- Dana-Farber Cancer Institute, PSMAR-IMIM Lab, Harvard Medical School, Boston, USA
| | - Y Loriot
- Université Paris-Sud, Université Paris-Saclay, Gustave Roussy, Villejuif, France
| | - A Necchi
- Vita-Salute San Raffaele University and IRCCS San Raffaele Hospital, Milan, Italy
| | - J Hoffman-Censits
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, USA
| | - J L Perez-Gracia
- Department of Oncology, Clinica Universidad de Navarra, Pamplona, Spain
| | | | - R Dreicer
- University of Virginia Cancer Center, Charlottesville, USA
| | - I Durán
- Hospital Universitario Virgen del Rocio, Seville, Spain
| | | | - A Drakaki
- University of California Los Angeles, Los Angeles, USA
| | - M Retz
- Technical University Munich, Munich, Germany
| | - S S Sridhar
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Center, Toronto, Canada
| | - P Grivas
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, USA
| | - E Y Yu
- University of Washington and Fred Hutchinson Cancer Center, Seattle, USA
| | | | - H A Burris
- Sarah Cannon Research Institute, Nashville, USA
| | | | - Y Shi
- Genentech, Inc., South San Francisco, CA, USA
| | - E Goluboff
- Genentech, Inc., South San Francisco, CA, USA
| | - D Bajorin
- Memorial Sloan Kettering Cancer Center, New York, USA
| |
Collapse
|
28
|
Bakaloudi DR, Talukder R, Makrakis D, Diamantopoulos L, Enright T, Leary JB, Patgunarajah U, Thomas VM, Swami U, Agarwal N, Jindal T, Koshkin VS, Brown JR, Barata P, Murgić J, Miletić M, Johnson J, Zakharia Y, Hui G, Drakaki A, Duran I, Buznego LA, Barrera RM, Castañeda DM, Rey-Cárdenas M, Castellano D, Nguyen CB, Park JJ, Alva A, McKay RR, Stewart TF, Epstein IB, Bellmunt J, Wright JL, Gupta S, Grivas P, Khaki AR. Association of Tumor Mutational Burden and Microsatellite Instability With Response and Outcomes in Patients With Urothelial Carcinoma Treated With Immune Checkpoint Inhibitor. Clin Genitourin Cancer 2024; 22:102198. [PMID: 39241315 DOI: 10.1016/j.clgc.2024.102198] [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: 04/08/2024] [Revised: 08/04/2024] [Accepted: 08/07/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND Microsatellite Instability (MSI) and Tumor Mutational Burden (TMB) are associated with immune checkpoint inhibitor (ICI) efficacy. We examined the association between TMB and MSI status with survival in patients with urothelial carcinoma (UC) treated with ICI. METHODS Patients from 15 institutions were treated with ICI monotherapy. Primary endpoint was overall survival and secondary endpoints included observed response rate (ORR), and progression-free (PFS) calculated from ICI initiation. TMB was analyzed as dichotomous (≥10 vs. <10 mut/Mb) and continuous variable. RESULTS We identified 411 patients: 203 were treated with ICI 1L/upfront; 104 with 2 + L. For the 1L/upfront: median [m] OS was numerically longer in patients with TMB ≥10 versus TMB <10: mOS 35 versus 26 months (HR = 0.6) and with MSI-H and MSI-S (mOS NR vs. 22 months), though neither association was statistically significant. A statistically significant association was found between TMB (continuous variable) and OS (HR = 0.96, P = .01). For 2 + L: mOS was numerically longer in patients with TMB ≥10 versus TMB <10: (20 vs. 12 months; HR = 0.9); mOS was 12 and 17 months for patients with MSI-H and MSI-S, respectively. Eighty-nine patients received maintenance avelumab (mAV): mOS was longer in patients with TMB ≥10 versus TMB <10: 61 versus 17 months; (HR = 0.2, P = .02) and with MSI-H and MSI-S (NR vs. 24 months). CONCLUSIONS Although not reaching statistical significance in several subsets, patients with high TMB and MSI-H had numerically longer OS with ICI, especially with mAV. Further validation is needed.
Collapse
Affiliation(s)
| | - Rafee Talukder
- Department of Medicine, Division of Hematology/Oncology, Baylor College of Medicine, Houston, TX
| | - Dimitrios Makrakis
- Department of Medicine, Jacobi Medical Center-Albert Einstein College of Medicine, Bronx, NY
| | | | - Thomas Enright
- Department of Medicine, University of Washington, Seattle, WA
| | - Jacob B Leary
- Department of Medicine, University of Washington, Seattle, WA
| | - Ubenthira Patgunarajah
- Department of Hematology and Oncology, Taussig cancer Institute, Cleveland Clinic Foundation, Cleveland, OH
| | - Vinay M Thomas
- Division of Oncology, Department of Medicine, University of Utah, Salt Lake City, UT
| | - Umang Swami
- Division of Oncology, Department of Medicine, University of Utah, Salt Lake City, UT
| | - Neeraj Agarwal
- Division of Oncology, Department of Medicine, University of Utah, Salt Lake City, UT
| | - Tanya Jindal
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, Helen Diller Family Cancer Center, San Francisco, CA
| | - Vadim S Koshkin
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, Helen Diller Family Cancer Center, San Francisco, CA
| | - Jason R Brown
- Division of Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH
| | - Pedro Barata
- Division of Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH
| | - Jure Murgić
- Department of Oncology and Nuclear Medicine, Sestre milosrdnice University Hospital Center, Zagreb, Croatia; Catholic University of Croatia School of Medicine, Zagreb, Croatia
| | - Marija Miletić
- Department of Oncology and Nuclear Medicine, Sestre milosrdnice University Hospital Center, Zagreb, Croatia
| | - Jeffrey Johnson
- Division of Oncology, Department of Medicine, University of Iowa, Iowa City, IA
| | - Yousef Zakharia
- Division of Oncology, Department of Medicine, University of Iowa, Iowa City, IA
| | - Gavin Hui
- Division of Hematology/Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Alexandra Drakaki
- Division of Hematology/Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Ignacio Duran
- Department of Oncology, University Hospital Marqués of Valdecilla, Cantabria, Spain
| | - Lucia A Buznego
- Department of Oncology, University Hospital Marqués of Valdecilla, Cantabria, Spain
| | - Rafael M Barrera
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology, Vall d' Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - David M Castañeda
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology, Vall d' Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Daniel Castellano
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Charles B Nguyen
- Division of Oncology, Department of Medicine, University of Michigan, Ann Arbor, MI
| | - Joseph J Park
- Division of Oncology, Department of Medicine, University of Michigan, Ann Arbor, MI
| | - Ajjai Alva
- Division of Oncology, Department of Medicine, University of Michigan, Ann Arbor, MI
| | - Rana R McKay
- Moores Cancer Center, University of California San Diego, La Jolla, CA
| | - Tyler F Stewart
- Moores Cancer Center, University of California San Diego, La Jolla, CA
| | - Ilana B Epstein
- Dana-Farber Cancer Institute/Brigham and Women's Hospital Harvard Medical School, Boston, MA
| | - Joaquim Bellmunt
- Dana-Farber Cancer Institute/Brigham and Women's Hospital Harvard Medical School, Boston, MA
| | | | - Shilpa Gupta
- Department of Hematology and Oncology, Taussig cancer Institute, Cleveland Clinic Foundation, Cleveland, OH.
| | - Petros Grivas
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA.
| | - Ali Raza Khaki
- Division of Oncology, Department of Medicine, Stanford University, Palo Alto, CA.
| |
Collapse
|
29
|
Stecca CE, Chowdhury D, Blais N, Alimohamed N, Wood L, Canil CM, Eigl B, Kulkarni GS, Black PC, Kassouf W, Chung P, North S, Jiang DM, Sridhar SS. 2024 CUA-GUMOC Expert Report: Management of unresectable locally advanced and metastatic urothelial carcinoma. Can Urol Assoc J 2024; 18:379-390. [PMID: 39641956 PMCID: PMC11623328 DOI: 10.5489/cuaj.8946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Affiliation(s)
- Carlos E. Stecca
- Division of Hematology and Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Deepro Chowdhury
- Division of Hematology and Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Normand Blais
- Division of Medical Oncology/Hematology, Centre Hospitalier de l’ Université de Montréal, Montreal, QC, Canada
| | - Nimira Alimohamed
- Department of Medicine, Division of Medical Oncology, Tom Baker Cancer Centre, University of Calgary, Calgary, AB, Canada
| | - Lori Wood
- Division of Medical Oncology, QEII Health Sciences Center, Halifax, NS, Canada
| | - Christina M. Canil
- The Ottawa Hospital Cancer Center, Ottawa, ON Canada; Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | | | - Girish S. Kulkarni
- Divisions of Urology and Surgical Oncology, Department of Surgery, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Peter C. Black
- Department of Urological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Wassim Kassouf
- Department of Surgery, McGill University Health Center, Montreal, QC, Canada
| | - Peter Chung
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Scott North
- Department of Medical Oncology, Cross Cancer Institute, Edmonton, AB, Canada
| | - Di Maria Jiang
- Division of Hematology and Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Srikala S. Sridhar
- Division of Hematology and Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
30
|
Fontes MS, de Almeida DVP, Cárcano F, Lages P, Dienstmann R. Precision medicine for urothelial carcinoma: An international perspective. Urol Oncol 2024; 42:402-410. [PMID: 38218630 DOI: 10.1016/j.urolonc.2023.11.008] [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: 09/04/2023] [Revised: 10/24/2023] [Accepted: 11/10/2023] [Indexed: 01/15/2024]
Abstract
The treatment landscape of urothelial cancers has evolved in the last decade with the approval of chemotherapy, immune checkpoint inhibitors, targeted therapies, and antibody drug conjugates. Although improvements in response and survival have been achieved with these strategies, in some scenarios their benefit is still questionable. Current efforts to identify prognostic and predictive biomarkers are crucial for better patient selection and treatment outcomes. In this paper we will review the most promising biomarkers under investigation, such as molecular classifiers, genomic alterations, programmed cell death ligand 1 expression, tumor mutational burden, circulating tumor DNA, urinary biomarkers among others, for muscle invasive bladder cancer and metastatic urothelial cancers. Deeper understanding of these biomarkers will aid clinical decision-making and help tailor treatment strategies.
Collapse
Affiliation(s)
- Mariane S Fontes
- Division of Genitourinary Medical Oncology, Oncoclínicas, Rio de Janeiro, Brazil; LAGOG-Latin American Cooperative Oncology Group, Porto Alegre, Brazil.
| | - Daniel Vargas Pivato de Almeida
- Division of Genitourinary Medical Oncology, Oncoclínicas, Brasília, Brazil; LAGOG-Latin American Cooperative Oncology Group, Porto Alegre, Brazil
| | - Flavio Cárcano
- Division of Genitourinary Medical Oncology, Oncoclínicas, Belo Horizonte, Brazil; Research and Teaching Institute, Barretos Cancer Hospital, Barretos, Brazil
| | - Paulo Lages
- Division of Genitourinary Medical Oncology, Oncoclínicas, Brasília, Brazil; LAGOG-Latin American Cooperative Oncology Group, Porto Alegre, Brazil
| | | |
Collapse
|
31
|
Gao H, Zhu J, Wu T, Long Q, Guan X, Chen Q, Yi W. Comprehensive pancancer analysis reveals that LPCAT1 is a novel predictive biomarker for prognosis and immunotherapy response. Apoptosis 2024; 29:2128-2146. [PMID: 39097858 DOI: 10.1007/s10495-024-02010-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2024] [Indexed: 08/05/2024]
Abstract
Lysophosphatidylcholine acyltransferase 1 (LPCAT1) is a crucial enzyme involved in phospholipid metabolism and is essential for maintaining the structure and functionality of biofilms. However, a comprehensive examination of the role of LPCAT1 across various cancer types is lacking. Multiple public databases have been utilized to examine LPCAT1 expression, genetic alterations, methylation, prognosis, biological function, and its relationship with antitumor immunity in different cancer types. The function of LPCAT1 in glioma, breast cancer and liver cancer cells was further verified using in vitro experiments. Our research indicated that LPCAT1 is upregulated in various cancers and is accompanied by a wide range of amplification mutations. Higher LPCAT1 expression was associated with poorer prognosis across multiple cancers. Further in vitro experiments demonstrated that interfering with LPCAT1 expression increased apoptosis in glioma, breast cancer and liver cancer cells and concurrently suppressed their proliferation and migration. Functional enrichment analysis revealed that LPCAT1-associated genes were primarily enriched in immune and cancer progression pathways, such as the JAK/STAT, MYC, and EMT, etc. Moreover, LPCAT1 expression was closely associated with immune cell infiltration and immune checkpoint-related gene expression. Interestingly, LPCAT1 expression levels were generally higher in patients in the immunotherapy response group. The combination of LPCAT1 and PDL1 serves as an effective predictor of immunotherapy response. In conclusion, LPCAT1 is involved in immune regulation and tumor progression and holds promise as a biomarker for predicting patient outcomes and immunotherapy efficacy.
Collapse
Affiliation(s)
- Hongyu Gao
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, Hunan, China
| | - Jinfeng Zhu
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, Hunan, China
| | - Tong Wu
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, Hunan, China
| | - Qian Long
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, Hunan, China
| | - Xinyu Guan
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, Hunan, China
| | - Qitong Chen
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, Hunan, China
| | - Wenjun Yi
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, Hunan, China.
| |
Collapse
|
32
|
Liu MZ, Chen JY, Lyu F, Gao XS, Ma MW, Li XY, Li HZ, Qin SB, Gao Y, Wang PY. Exploring Radiotherapy as a Promising Alternative for Managing Advanced Upper Tract Urothelial Carcinoma: Rescuing Chemotherapy-Intolerant Patients. Clin Genitourin Cancer 2024; 22:102203. [PMID: 39241310 DOI: 10.1016/j.clgc.2024.102203] [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: 05/13/2024] [Revised: 07/31/2024] [Accepted: 08/10/2024] [Indexed: 09/09/2024]
Abstract
PURPOSE To investigate the safety and effectiveness of radiotherapy for advanced upper tract urothelial carcinoma (UTUC) patients intolerant to chemotherapy. METHODS Data for 21 patients with advanced UTUC intolerant to chemotherapy were retrospectively collected. All patients were treated with conventionally fractionated radiotherapy (50-70 Gy/20-33 f) or partial-SABR boost to the lesions (50-60 Gy/20-25 f with tumor center boosted with 6-8 Gy/f, 3-5 f) for bulky tumors. RESULTS The median age was 75 years (range, 58-87 years). Primary tumor resection was performed for all patients and none underwent metastatic resection. Seventeen (81%) patients had oligometastasis (1-5 metastases) at diagnosis. Eighteen (85.7%) received irradiation to all tumor lesions. Lymph node metastasis was predominant in the whole group (17/21). Other lesions were distributed as local recurrence (7/21), bone metastases (2/21) and abdominal wall/muscle (2/21). The median follow-up time was 38.5 months (interquartile range, 15.2-48.7 months). Rate of local control (LC), progression-free survival (PFS) and overall survival (OS) of the whole group at 1 year were 90%, 46.6%, and 80.4%, respectively. At 3 years, LC, PFS and OS were 65.6%, 26.6%, and 40.9%, respectively. Fourteen patients developed acute mild gastrointestinal toxicity, generally of grade 1-2; 8 patients developed acute grade 1-2 hematological toxicity, consisting mainly of anemia and leukopenia. No grade 3 or higher acute or late toxicities were observed. CONCLUSION For patients with advanced UTUC who are not able to tolerate chemotherapy, radiotherapy is a safe treatment and can achieve good local tumor control.
Collapse
Affiliation(s)
- Ming-Zhu Liu
- Department of Radiation Oncology, Peking University First Hospital, Beijing, China; Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Jia-Yan Chen
- Department of Radiation Oncology, Peking University First Hospital, Beijing, China
| | - Feng Lyu
- Department of Radiation Oncology, Peking University First Hospital, Beijing, China
| | - Xian-Shu Gao
- Department of Radiation Oncology, Peking University First Hospital, Beijing, China.
| | - Ming-Wei Ma
- Department of Radiation Oncology, Peking University First Hospital, Beijing, China
| | - Xiao-Ying Li
- Department of Radiation Oncology, Peking University First Hospital, Beijing, China
| | - Hong-Zhen Li
- Department of Radiation Oncology, Peking University First Hospital, Beijing, China
| | - Shang-Bin Qin
- Department of Radiation Oncology, Peking University First Hospital, Beijing, China
| | - Yan Gao
- Department of Radiation Oncology, Peking University First Hospital, Beijing, China
| | - Pei-Yan Wang
- School of Information, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
33
|
Liu MZ, Li XY, Gao XS, Lyu F, Ma MW, Chen JY, Gao Y, Ren XY, Li XS. Outcomes of Radical Radiotherapy for the Treatment of Localized Renal Pelvic and Ureteral Carcinoma Intolerant to Surgery: A Real-World Study. Clin Genitourin Cancer 2024; 22:102216. [PMID: 39326156 DOI: 10.1016/j.clgc.2024.102216] [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: 03/13/2024] [Revised: 05/04/2024] [Accepted: 08/12/2024] [Indexed: 09/28/2024]
Abstract
PURPOSE To investigate the safety and efficacy of radical radiotherapy for localized inoperable renal pelvic and ureteral carcinoma. METHODS 23 patients who received radiotherapy were enrolled. The prescribed dose was 60 to 67.5 Gy in 25 fractions and for bulky tumors, SABR was used in the first 3 to 5 times with tumor center boosted synchronously with 6 to 8 Gy/f. The Kaplan-Meier method was used to calculate local control (LC), DMFS, CSS and OS. Univariate analysis was performed by the log-rank test. The change in the eGFR before and after radiotherapy was compared by paired t test. The side effects were graded by CTCAE, version 5.0. RESULTS The median follow-up time was 17 months. The LC rates at 2 years after radiotherapy were 85.0%; the DMFS rates were 52.2%; the CSS rates were 83.0%; and the OS rates were 77.8%. The main failure mode after radiotherapy was distant metastasis. Univariate analysis revealed that T3-4 stage (P = .001), N+ status (P < .001) and a tumor volume ≥ 20 cc (P = .005) were poor prognostic factors for DMFS. There was no significant difference in the mean eGFR before and after radiotherapy (47.0 mL/min/1.73m2 vs. 48.5 mL/min/1.73m2, P = .632). Only 1 patient developed acute grade 3 anemia. No patients developed grade 3 or higher late toxicities. CONCLUSION For localized inoperable renal pelvic and ureteral carcinoma, radiotherapy is well tolerable with high local control and expected to bring survival benefits. In such patients, radiotherapy may be an option when surgery is unsuitable.
Collapse
Affiliation(s)
- Ming-Zhu Liu
- Department of Radiation Oncology, Peking University First Hospital, Beijing, China; Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Xiao-Ying Li
- Department of Radiation Oncology, Peking University First Hospital, Beijing, China
| | - Xian-Shu Gao
- Department of Radiation Oncology, Peking University First Hospital, Beijing, China.
| | - Feng Lyu
- Department of Radiation Oncology, Peking University First Hospital, Beijing, China
| | - Ming-Wei Ma
- Department of Radiation Oncology, Peking University First Hospital, Beijing, China
| | - Jia-Yan Chen
- Department of Radiation Oncology, Peking University First Hospital, Beijing, China
| | - Yan Gao
- Department of Radiation Oncology, Peking University First Hospital, Beijing, China
| | - Xue-Ying Ren
- Department of Radiation Oncology, Peking University First Hospital, Beijing, China
| | - Xue-Song Li
- Department of Urology, Peking University First Hospital, Beijing, China.
| |
Collapse
|
34
|
Yilmaz F, Sagir S. Prognostic and predictive value of tertiary lymphoid structures in TURBT materials: Should it be seated in the routine pathological examination, and can it be used in deciding on the treatment method? Urol Oncol 2024; 42:450.e13-450.e22. [PMID: 39089974 DOI: 10.1016/j.urolonc.2024.06.010] [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: 02/24/2024] [Revised: 05/22/2024] [Accepted: 06/10/2024] [Indexed: 08/04/2024]
Abstract
OBJECTIVE This study aims to reveal the importance of tertiary lymphoid structures (TLS) in transurethral resection of bladder tumor (TURBT) materials with a practical and applicable method in which the effect of a certain threshold value on survival and treatment response can be implicated. METHODS TURBT materials that had not previously received any treatment (chemotherapy, radiotherapy, or immunotherapy) and were diagnosed for the first time at Mardin Training and Research Hospital between 2014 and 2022 were included in the study. The maximum TLS per 4× magnification field (field diameter: 4.5 mm) was recorded. Grouping and statistical analysis of the TLS number were performed using threshold values of "≥1", "≥2", and "≥3". RESULTS TLSs were more frequently found in high-grade tumors (P = 0.008) and showed a strong association with stage progression (P < 0.001). It was also significantly associated with many adverse histopathological parameters. Conversely, high TLS (≥1, ≥2, and ≥3) appeared to be associated with fewer recurrences (P = 0.032, P = 0.001, and P = 0.018, respectively), and cases with higher TLS showed longer recurrence-free survival (P = 0.089, P = 0.023, P = 0.037, respectively). TLS≥3 was found to be an independent parameter that was associated with favorable RFS (P = 0.019, HR = 0.401), and multifocality was found to be an independent risk factor for RFS (P = 0.023, HR = 2.302). CONCLUSION This study is the first to demonstrate the relationship between the presence and specific thresholds of TLS in TURBT materials with prognostic parameters. Including this information in the routine pathological examination of TURBT materials will allow a more accurate approach to treatment and follow-up, especially in patients with non-muscle invasive bladder cancer (NMIBC).
Collapse
Affiliation(s)
- Fatih Yilmaz
- Pathology Laboratory, Mardin Training and Research Hospital, Mardin, Turkey.
| | - Suleyman Sagir
- Department of Urology, Mardin Artuklu University, Mardin, Turkey.
| |
Collapse
|
35
|
Mavadia A, Choi S, Ismail A, Ghose A, Tan JK, Papadopoulos V, Sanchez E, Boussios S. An overview of immune checkpoint inhibitor toxicities in bladder cancer. Toxicol Rep 2024; 13:101732. [PMID: 39318722 PMCID: PMC11420502 DOI: 10.1016/j.toxrep.2024.101732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/04/2024] [Accepted: 09/09/2024] [Indexed: 09/26/2024] Open
Abstract
Bladder cancer is the tenth most prevalent malignancy worldwide, with a significant mortality burden. Urothelial carcinoma (UC) is the most common histological subtype, and treatment options are guided by whether the disease is muscle-invasive (MIBC) or non-muscle-invasive (NMIBC), with subsequent risk group stratification. The growing popularity of immune checkpoint inhibitors (ICIs) to treat MIBC and NMIBC as either monotherapy or combined with intravesical agents, may radically change the treatment paradigm of UC. Current treatments for NMBIC includes intravesical chemotherapy after trans-urethral resection of the bladder tumour, intravesical bacillus Calmette-Guerin (BCG) or radical cystectomy. Cisplatin-based chemotherapy is widely regarded as the first-line treatment for metastatic UC due to its beneficial response and survival rates when compared to alternative therapies. However, up to 70 % of metastatic UC patients are ineligible, and the prognosis of these patients remains poor, with a median survival of 13-16 months. For NMIBC and MIBC, ICIs provide a promising alternative for cisplatin-ineligible patients. In UC, ICIs including atezolizumab, nivolumab, avelumab, and pembrolizumab are Food and Drug Administration (FDA)-approved for monotherapy, and have demonstrated promising results, particularly in those who cannot receive cisplatin-based chemotherapy, and as a second-line treatment option for recurrent UC following platinum-based chemotherapy. It is important to consider that some patients may experience adverse events (AEs) with limited clinical benefit. Infusion-related reactions and immune-mediated AEs (imAEs) such as colitis, endocrinopathies, hepatitis, pneumonitis, interstitial lung disease, renal dysfunction, nephritis, cutaneous and neurological toxicities must be monitored for. Currently, there is no clear consensus on the role of a 'two-year stopping rule' in reducing the risk of imAEs, with further research on the optimal treatment duration of ICIs required. With increased ICI use, vigilance regarding their side effects is imperative. This review aims to provide an updated overview of ICI toxicities in bladder cancer, to assist clinicians in their therapeutic decision-making, with consideration of patient characteristics and the clinical context.
Collapse
Affiliation(s)
- Avenie Mavadia
- GKT School of Medicine, King's College London, London SE1 9RT, UK
| | - Sunyoung Choi
- Hampshire Hospitals NHS Foundation Trust, Aldermaston Road, Basingstoke, Hampshire RG24 9NA, UK
| | - Ayden Ismail
- GKT School of Medicine, King's College London, London SE1 9RT, UK
| | - Aruni Ghose
- Department of Medical Oncology, Barts Cancer Centre, St Bartholomew’s Hospital, Barts Heath NHS Trust, London EC1A 7BE, UK
- Department of Medical Oncology, Mount Vernon Cancer Centre, East and North Hertfordshire Trust, London HA6 2RN, UK
- Health Systems and Treatment Optimisation Network, European Cancer Organisation, Brussels 1040, Belgium
- Immuno-Oncology Clinical Network, Liverpool, UK
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, UK
| | - Joecelyn Kirani Tan
- School of Medicine, University of St. Andrews, North Haugh, Fife, Scotland KY16 9TF, UK
- St Andrews Oncology Society, St Mary's Place, Scotland KY16 9UZ, UK
| | | | - Elisabet Sanchez
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, UK
| | - Stergios Boussios
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, UK
- Faculty of Medicine, Health, and Social Care, Canterbury Christ Church University, Canterbury CT2 7PB, UK
- King's College London, Faculty of Life Sciences & Medicine, School of Cancer & Pharmaceutical Sciences, London SE1 9RT, UK
- Kent Medway Medical School, University of Kent, Canterbury CT2 7LX, UK
- AELIA Organization, 9th Km Thessaloniki – Thermi, Thessaloniki 57001, Greece
| |
Collapse
|
36
|
Zhu L, Xiao F, Hou Y, Huang S, Xu Y, Guo X, Dong X, Xu C, Zhang X, Gu H. Identification of anoikis-related molecular patterns and the novel risk model to predict prognosis, tumor microenvironment infiltration and immunotherapy response in bladder cancer. Front Immunol 2024; 15:1491808. [PMID: 39664392 PMCID: PMC11631915 DOI: 10.3389/fimmu.2024.1491808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 11/04/2024] [Indexed: 12/13/2024] Open
Abstract
Background Anoikis, a unique form of cell death, serves as a vital part of the organism's defense by preventing shedding cells from re-attaching to the incorrect positions, and plays pivotal role in cancer metastasis. Nonetheless, the specific mechanisms among anoikis, the clinical prognosis and tumor microenvironment (TME) of bladder cancer (BLCA) are insufficiently understood. Method BLCA patients were classified into different anoikis subtypes based on the expression of candidate anoikis-related genes (ARGs), and differences in the clinicopathological features, TME, immune cell infiltration, and immune checkpoints between two anoikis subtypes were analyzed. Next, patients in the TCGA cohort were randomized into the train and test groups in a 1:1 ratio. Subsequently, the anoikis-related model was constructed to predict the prognosis via utilizing the univariate Cox, LASSO and multivariate Cox analyses, and validated internally and externally. Moreover, the relationships between the risk score and clinicopathologic features, immune cell infiltration, immunotherapy response, and antitumor drug sensitivity were also analyzed. In addition, representative genes were evaluated using immunohistochemistry in clinical specimens, and in BLCA cell lines, functional experiments were performed to determine the biological behavior of hub gene PLOD1. Result Two definite anoikis subgroups were identified. Compared to ARGcluster A, patients assigned to ARGcluster B were characterized by an immunosuppressive microenvironment and worse prognosis. Then, the anoikis-related model, including PLOD1, EHBP1, and CSPG4, was constructed, and BLCA patients in the low-risk group were characterized by a better prognosis. Next, the accurate nomogram was built to improve the clinical applicability by combining the age, tumor stage and risk Score. Moreover, immune infiltration and clinical features differed significantly between high- and low-risk groups. We also found that the low-risk group exhibited a lower tumor immune dysfunction and exclusion score, a higher immunophenoscore (IPS), had more sensitivity to immunotherapy. Eventually, the expression levels of three genes were verified by our experiment, and knockdown of PLOD1 could inhibit invasion and migration abilities in BLCA cell lines. Conclusion These results demonstrated a new direction in precision therapy for BLCA, and indicated that the ARGs might be helpful to in predicting prognosis and as therapeutic targets in BLCA.
Collapse
Affiliation(s)
- Luochen Zhu
- Department of Pharmacy, Nantong Tumor Hospital (Tumor Hospital Affiliated to Nantong University), Nantong, China
| | - Feng Xiao
- Department of Pathology, Affiliated Nantong Hospital 3 of Nantong University (Nantong Third People’s Hospital), Nantong, China
| | - Yi Hou
- Department of Pharmacy, People’s Hospital of Zhongjiang, Deyang, China
| | - Shenjun Huang
- Department of Oncology, Nantong Tumor Hospital (Tumor Hospital Affiliated to Nantong University), Nantong, China
| | - Yanyan Xu
- Department of Pharmacy, Nantong Tumor Hospital (Tumor Hospital Affiliated to Nantong University), Nantong, China
| | - Xiaohong Guo
- Department of Pharmacy, Nantong Tumor Hospital (Tumor Hospital Affiliated to Nantong University), Nantong, China
| | - Xinwei Dong
- Department of Pharmacy, Nantong Tumor Hospital (Tumor Hospital Affiliated to Nantong University), Nantong, China
| | - Chunlu Xu
- Department of Andrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Xiaolei Zhang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Haijuan Gu
- Department of Pharmacy, Nantong Tumor Hospital (Tumor Hospital Affiliated to Nantong University), Nantong, China
| |
Collapse
|
37
|
Song DM, Feng K, Luo WF, Lv DS, Zhou LP, He YB, Jin Y. Predicting survival in bladder cancer with a novel apoptotic gene-related prognostic model. Discov Oncol 2024; 15:702. [PMID: 39580765 PMCID: PMC11586327 DOI: 10.1007/s12672-024-01575-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 11/12/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND Apoptosis and apoptotic genes play a critical role in the carcinogenesis and progression of bladder cancer. However, there is no prognostic model established by apoptotic genes. METHODS Messenger RNA (mRNA), Expression data, and related clinical data were obtained from The Cancer Genome Atlas (TCGA) database and Gene Expression Omnibus (GEO) database. After extracting the apoptosis-related genes, the survival-related apoptosis genes were screened by univariate Cox regression analysis in the TCGA cohort. Following the Least Absolute Shrinkage and Selection Operator (LASSO) regression method, these genes were modeled by multivariate Cox analysis. The predictive abilities of the Apoptosis-Related Gene Model (ARGM) for overall survival (OS) rate, disease-specific survival (DSS) measures, and progression-free survival (PFS) were verified by the Kaplan-Meier(K-M)survival analysis and time-dependent Receiver Operating Characteristic (ROC) curve. Functional enrichment analyses were performed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genome (KEGG). CIBERSORT and Single-Sample Gene Set Enrichment Analysis (ssGSEA) were used to calculate the type of immune cell infiltration and immune functions. The model's predictive ability for immunotherapy were evaluated using Tumor Immune Dysfunction and Exclusion (TIDE) and the Imvigor210 study.The single-cell sequencing was used to display the expression level of the ARGM.Finally,qRT-PCR was executed to validate the expression level of ARGM. RESULTS Several apoptosis genes were identified through the model, including ANXA1, CASP6, CD2, F2, PDGFRB, SATB1, and TSPO. The prognostic value of the model for OS, DSS, and PFS were verified using the TCGA and GEO cohort. The model can predict patient response to immunotherapy treatment as established through the model's score which was linked to different types of immune cell infiltration and identified significant differences in the signal pathways between high-risk and low-risk groups. Nomogram variables, prompted from ARGM and clinical parameters, also generate a high predictive value for patient survival. CONCLUSION Ourestablished apoptosis-related gene model (ARGM) has a substantial predictive value for prognosis and immunotherapy of bladder cancer. It may help with clinical consultation, clinical stratification, and treatment selection. The immune infiltration status and signal pathway of different risk groups also provide direction for further research.
Collapse
Affiliation(s)
- Ding-Ming Song
- Department of Urology, Jinzhou Medical University, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Kun Feng
- Department of Urology, Jinzhou Medical University, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Wen-Fei Luo
- Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Dong-Shan Lv
- Department of Urology, Jinzhou Medical University, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Li-Po Zhou
- Department of Urology, Jinzhou Medical University, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Yi-Bo He
- Department of Clinical Lab, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China.
| | - Yanyang Jin
- Department of Urology, Jinzhou Medical University, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China.
| |
Collapse
|
38
|
Wang Y, Hu M, Finn OJ, Wang XS. Tumor-Associated Antigen Burden Correlates with Immune Checkpoint Blockade Benefit in Tumors with Low Levels of T-cell Exhaustion. Cancer Immunol Res 2024; 12:1589-1602. [PMID: 39137006 PMCID: PMC11534523 DOI: 10.1158/2326-6066.cir-23-0932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 03/20/2024] [Accepted: 08/09/2024] [Indexed: 09/14/2024]
Abstract
Tumor-associated antigens (TAA) are important targets for cancer vaccines. However, TAA-based vaccines have not yet achieved their full potential in clinical trials. In contrast, immune checkpoint blockade (ICB) has emerged as an effective therapy, leading to durable responses in selected patients with cancer. To date, few generalizable associations between TAAs and ICB benefit have been reported, with most studies focusing on melanoma, which has the highest mutation rate in cancer. In this study, we developed a TAA burden (TAB) algorithm based on known and putative TAAs and investigated the association of TAB with ICB benefit. Analysis of the IMvigor210 patient cohort of urothelial carcinoma treated with anti-PDL1 revealed that high tumor mutation burden weakened the association of TAB with ICB benefit. Furthermore, TAB correlated with ICB efficacy in tumors characterized by negative PDL1 staining on immune cells; however, high levels of PDL1 staining on immune cells were linked to T-cell exhaustion. Validation across independent clinical datasets-including urothelial carcinoma cohorts treated with anti-PD1/PDL1 agents and neoadjuvant anti-PD1 trials for head and neck cancers-corroborated the finding that TAB correlates with ICB benefit in tumors with low T-cell exhaustion. Pan-cancer analyses revealed that in most cancer entities, tumors with higher T-cell exhaustion exhibited lower TAB levels, implying possible immunoediting of TAAs in tumors with established antitumor immunity. Our study challenges the prevailing notion of a lack of association between TAAs and ICB response. It also underscores the need for future investigations into the immunogenicity of TAAs and TAA-based vaccine strategies in tumors with low levels of T-cell exhaustion.
Collapse
Affiliation(s)
- Yue Wang
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15213, U.S.A
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15213, U.S.A
| | - Mengying Hu
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15213, U.S.A
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15213, U.S.A
| | - Olivera J Finn
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15213, U.S.A
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, U.S.A
| | - Xiao-Song Wang
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15213, U.S.A
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15213, U.S.A
| |
Collapse
|
39
|
Holland L, Bhanvadia R, Ibeziako N, Taylor J, Gerlt D, Chaplin I, Bagrodia A, Desai N, Gaston K, Lotan Y, Margulis V, Zhang T, Cole S, Woldu S. Socioeconomic and Demographic Disparities in Immunotherapy Utilization for Advanced Kidney and Bladder Cancer. Urol Oncol 2024; 42:374.e11-374.e20. [PMID: 39034222 DOI: 10.1016/j.urolonc.2024.06.012] [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: 01/17/2024] [Revised: 05/27/2024] [Accepted: 06/10/2024] [Indexed: 07/23/2024]
Abstract
OBJECTIVES Immunotherapy (IO) drugs have been increasingly utilized in locally advanced or metastatic clear cell renal cell carcinoma (ccRCC) and urothelial carcinoma of the bladder (UC). Multiple trials have demonstrated clear survival benefit, however, there are often barriers to access for these advanced therapies which has been demonstrated in other non-urologic malignancies. The goal of this study was to assess socioeconomic and demographic factors associated with the receipt of IO for advanced ccRCC and UC. MATERIALS AND METHODS We queried the National Cancer Database (NCDB) for patients with stage IV ccRCC and UC. The study period was 2015 to 2020 for ccRCC (FDA approval date of IO) and 2017 to 2020 for UC (FDA approval date of broadened indication for IO, initial limited approval in 2016). The primary outcome of interest was receipt of IO therapy using multivariable logistic regression, adjusting for relevant socioeconomic and demographic variables. RESULTS We identified 15,926 patients with stage IV ccRCC and 10,380 patients with stage IV UC of which 5,419 (34.0%) and 2,231 (21.5%) received IO therapy, respectively. IO utilization increased with each successive year. In both malignancies, treatment at a non-academic facility, education level, income, and insurance were independently associated with IO utilization. For ccRCC, black (OR = 0.77, 95% CI, 0.64-0.93, P = 0.009) and Hispanic race (OR = 0.73, 95% CI, 0.61-0.86, P = 0.006) were each associated with decreased IO utilization but there were no independent associations between race and receipt of IO in patients with UC. CONCLUSIONS In the era of FDA-approved IO therapy for advanced ccRCC and UC, this national cohort analysis suggests that IO utilization is increasing over time, but significant disparities exist based on income, education, and insurance status in both malignancies. Additionally, patients treated at non-academic facilities were less likely to receive IO therapy for these specific genitourinary malignancies. In ccRCC, additional disparities were seen black and Hispanic races which each were associated with lower odds of IO receipt. Identifying strategies to mitigate these differences and provide equitable access to IO therapy is of imperative need.
Collapse
Affiliation(s)
- Levi Holland
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Raj Bhanvadia
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Nathanaelle Ibeziako
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Jacob Taylor
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Deitrich Gerlt
- Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth, TX
| | - Iftach Chaplin
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Aditya Bagrodia
- Department of Urology, University of California San Diego, San Diego, CA
| | - Neil Desai
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Kris Gaston
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Yair Lotan
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Vitaly Margulis
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Tian Zhang
- Department of Internal Medicine, Division of Hematology and Oncology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Suzanne Cole
- Department of Internal Medicine, Division of Hematology and Oncology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Solomon Woldu
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX.
| |
Collapse
|
40
|
Cai C, Shen J. The roles of migrasomes in immunity, barriers, and diseases. Acta Biomater 2024; 189:88-102. [PMID: 39284502 DOI: 10.1016/j.actbio.2024.09.013] [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: 03/19/2024] [Revised: 09/01/2024] [Accepted: 09/10/2024] [Indexed: 10/14/2024]
Abstract
Migrasomes are recently identified extracellular vesicles and organelles formed in conjunction with cell migration. They are situated at the rear of migrating cells, exhibit a circular or elliptical membrane-enclosed structure, and function as a new organelle. Migrasomes selectively sort intercellular components, mediating a cell migration-dependent release mechanism known as migracytosis and modulating cell-cell communication. Accumulated evidence clarifies migrasome formation processes and indicates their diverse functional roles. Migrasomes may also be potentially correlated with the occurrence, progression, and prognosis of certain diseases. Migrasomes' involvement in physiological and pathological processes highlights their potential for expanding our understanding of biological procedures and as a target in clinical therapy. However, the precise mechanisms and full extent of their involvement in immunity, barriers, and diseases remain unclear. This review aimed to provide a comprehensive overview of the roles of migrasomes in human immunity and barriers, in addition to providing insights into their impact on human diseases. STATEMENT OF SIGNIFICANCE: Migrasomes, newly identified extracellular vesicles and organelles, form during cell migration and are located at the rear of migrating cells. These circular or elliptical structures mediate migracytosis, selectively sorting intercellular components and modulating cell-cell communication. Evidence suggests diverse functional roles for migrasomes, including potential links to disease occurrence, progression, and prognosis. Their involvement in physiological and pathological processes highlights their significance in understanding biological procedures and potential clinical therapies. However, their exact mechanisms in immunity, barriers, and diseases remain unclear. This review provides an overview of migrasomes' roles in human immunity and barriers, and their impact on diseases.
Collapse
Affiliation(s)
- Changsheng Cai
- Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Shanghai Institute of Digestive Disease, 160# Pu Jian Ave, Shanghai 200127, China
| | - Jun Shen
- Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Shanghai Institute of Digestive Disease, 160# Pu Jian Ave, Shanghai 200127, China.
| |
Collapse
|
41
|
Sweis RF, Gajate P, Morales-Barrera R, Lee JL, Necchi A, de Braud F, Penel N, Grünwald V, Maruzzo M, Meran J, Ishida TC, Bao W, Zhou Y, Ellinghaus P, Rosenberg JE. Rogaratinib Plus Atezolizumab in Cisplatin-Ineligible Patients With FGFR RNA-Overexpressing Urothelial Cancer: The FORT-2 Phase 1b Nonrandomized Clinical Trial. JAMA Oncol 2024; 10:1565-1570. [PMID: 39298147 PMCID: PMC11413753 DOI: 10.1001/jamaoncol.2024.3900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/14/2024] [Indexed: 09/25/2024]
Abstract
Importance The oral pan-fibroblast growth factor receptor inhibitor rogaratinib previously demonstrated encouraging safety and efficacy in a phase 1 study of patients with urothelial cancer (UC) overexpressing FGFR messenger RNA (mRNA). Objective To evaluate the safety, pharmacokinetics, and preliminary efficacy of rogaratinib in combination with the programmed cell death 1 ligand 1 (PD-L1) inhibitor atezolizumab in cisplatin-ineligible patients with FGFR mRNA-positive, locally advanced/metastatic UC. Design Setting and Participants The FORT-2 nonrandomized clinical trial was an open-label, single-arm, multicenter study conducted between May 15, 2018, and July 16, 2021, in 30 centers across Asia, Europe, and North America. Eligible patients had locally advanced/metastatic UC with FGFR1/3 mRNA overexpression and were ineligible for cisplatin-based chemotherapy. The data analysis was completed from July 2022 to September 2022. Interventions Patients received rogaratinib 600 mg or rogaratinib 800 mg twice daily in combination with intravenous atezolizumab 1200 mg every 21 days. Main Outcomes and Measures Primary end points included safety, tolerability, and the recommended phase 2 dose (RP2D) of rogaratinib in combination with atezolizumab. Results Among 153 patients screened, 73 (48%) had tumors with FGFR1/3 mRNA overexpression, and 37 patients were enrolled and treated (median [range] age, 75.0 [47.0-85.0] years; 32 [87%] male). The most common treatment-emergent adverse events (TEAEs) included diarrhea in 23 patients (62%), hyperphosphatemia in 19 (51%), and fatigue in 15 (41%). Grade 3 or higher TEAEs were reported in 27 patients (73%), and 4 grade 5 TEAEs were reported, though unrelated to treatment. The RP2D was rogaratinib 600 mg in combination with atezolizumab 1200 mg. At the RP2D, the overall response rate was 53.8% in the rogaratinib 600 mg group, including 4 patients (15%) with complete responses; 12 responders (86%) did not have an FGFR3 gene alteration, and 11 (79%) had low PD-L1 expression. Conclusions and Relevance In this phase 1b nonrandomized clinical trial, rogaratinib plus atezolizumab demonstrated a manageable safety profile, with no unexpected safety signals. Efficacy for this combination at the RP2D was observed in tumors with low PD-L1 and was not dependent on FGFR3 gene alterations, suggesting broad potential benefit for patients with locally advanced/metastatic UC and FGFR mRNA overexpression. Trial Registration ClinicalTrials.gov Identifier: NCT03473756.
Collapse
MESH Headings
- Humans
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/adverse effects
- Male
- Female
- Aged
- Middle Aged
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Cisplatin/administration & dosage
- Cisplatin/therapeutic use
- Cisplatin/adverse effects
- Receptor, Fibroblast Growth Factor, Type 3/genetics
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Aged, 80 and over
- Urologic Neoplasms/drug therapy
- Urologic Neoplasms/genetics
- Urologic Neoplasms/pathology
- Carcinoma, Transitional Cell/drug therapy
- Carcinoma, Transitional Cell/genetics
- RNA, Messenger/genetics
- Urinary Bladder Neoplasms/drug therapy
- Urinary Bladder Neoplasms/genetics
- Urinary Bladder Neoplasms/pathology
Collapse
Affiliation(s)
- Randy F. Sweis
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Pablo Gajate
- Medical Oncology Department, Ramón y Cajal University Hospital, Madrid, Spain
| | - Rafael Morales-Barrera
- Department of Medical Oncology, Vall d’Hebron Institute of Oncology, Vall d’Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jae-Lyun Lee
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Andrea Necchi
- Vita-Salute San Raffaele University, Milan, Italy
- Department of Medical Oncology, IRCCS San Raffaele Hospital, Milan, Italy
| | | | - Nicolas Penel
- Department of Medical Oncology, Centre Oscar Lambret, University of Lille, Lille, France
| | - Viktor Grünwald
- Departments of Urology and Medical Oncology, Universitätsklinikum Essen, Essen, Germany
| | - Marco Maruzzo
- Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Johannes Meran
- Department of Internal Medicine, Krankenhaus der Barmherzigen Brüder, Vienna, Austria
| | | | - Weichao Bao
- Bayer HealthCare Pharmaceuticals, Whippany, New Jersey
| | - Yinghui Zhou
- Bayer HealthCare Pharmaceuticals, Cambridge, Massachusetts
| | - Peter Ellinghaus
- Bayer AG, Wuppertal, Germany
- Now with Merck KGaA, Darmstadt, Germany
| | - Jonathan E. Rosenberg
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
42
|
Aydogdu C, Brinkmann I, Casuscelli J. [Novel systemic treatment options for advanced bladder cancer]. UROLOGIE (HEIDELBERG, GERMANY) 2024; 63:1162-1173. [PMID: 39207469 DOI: 10.1007/s00120-024-02413-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/11/2024] [Indexed: 09/04/2024]
Abstract
Systemic treatment of urothelial carcinoma of the bladder requires complex approaches and is constantly evolving. Neoadjuvant cisplatin-based chemotherapy followed by radical cystectomy remains the current standard of care for muscle-invasive advanced bladder cancer. For patients ineligible for cisplatin, adjuvant treatment with nivolumab is recommended. Innovative perioperative combinations could transform the treatment landscape in the future. First-line treatment for metastatic urothelial carcinoma has long been dominated by platinum-based combinations, recently followed by the immune checkpoint inhibitor avelumab as maintenance therapy; however, recent results on the use of enfortumab vedotin and pembrolizumab in the first-line setting are expected to fundamentally change the treatment options. In subsequent lines of treatment, the not yet approved erdafitinib, as the first targeted therapy for advanced urothelial carcinoma, offers an important alternative and underscores the need for molecular testing.
Collapse
Affiliation(s)
- C Aydogdu
- Urologische Klinik und Poliklinik, Klinikum der Universität München, Ludwig-Maximilians-Universität München, München, Deutschland
| | - I Brinkmann
- Urologische Klinik und Poliklinik, Klinikum der Universität München, Ludwig-Maximilians-Universität München, München, Deutschland
| | - J Casuscelli
- Urologische Klinik und Poliklinik, Klinikum der Universität München, Ludwig-Maximilians-Universität München, München, Deutschland.
- Uro-Onkologische Tagesklinik, Urologische Klinik und Poliklinik, LMU-Klinikum, Campus Großhadern, Marchioninistr. 15, 81377, München, Deutschland.
| |
Collapse
|
43
|
Gu Y, Feng Z, Xu X, Jin L, Jiang G. LINC01929 Is a Prognostic Biomarker for Multiple Tumours and Promotes Cell Proliferation in Breast Cancer Through the TNF/STAT3 Axis. J Cell Mol Med 2024; 28:e70227. [PMID: 39586790 PMCID: PMC11588430 DOI: 10.1111/jcmm.70227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 11/04/2024] [Accepted: 11/07/2024] [Indexed: 11/27/2024] Open
Abstract
The aim of this study was to investigate whether long intergenic non-coding RNA 1929 (LINC01929), a novel long non-coding RNA, could serve as a prognostic biomarker for various tumours and explore its function. The expression and prognosis of LINC01929 across 33 different tumour types in patients in the Cancer Genome Atlas (TCGA) database were analysed. Also, the correlation between LINC01929 expression, tumour mutational burden (TMB), microsatellite instability (MSI), immune checkpoint status and immune cell infiltration was examined. Moreover, the function of LINC01929 in the breast cancer cell lines was explored via CCK-8, colony formation and cell cycle assays. In addition, the downstream mechanisms of LINC01929 were analysed via transcriptome sequencing, RT-qPCR, and western blotting. Our analysis revealed that LINC01929 was weakly expressed in 3 tumour types and highly expressed in 14 tumour types, and low expression of LINC01929 was correlated with better clinical outcomes in 15 tumour types. Furthermore, LINC01929 expression was correlated significantly with the TMB, MSI, immune checkpoint and immune cell infiltration across multiple tumour types. The knockdown of LINC01929 inhibited cell cycle progression, cell proliferation, and tumorigenesis and downregulated the TNF pathway and STAT3 expression. The treatment with exogenous TNF-α partially reversed the cell cycle progression and proliferation inhibition caused by LINC01929 knockdown, and these effects were accompanied by changes in STAT3 expression. LINC01929 may serve as an effective biomarker affecting the TMB, MSI, immune cell infiltration and immune checkpoint status. Mechanistically, LINC01929 affects cell cycle progression and cell proliferation through the TNF/STAT3 axis. These findings offer valuable insights into the potential applications of LINC01929 in tumour therapy, which may yield novel targets and strategies for the diagnosis and treatment of patients.
Collapse
Affiliation(s)
- Yanlin Gu
- Department of Thyroid and Breast SurgeryThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Zhengyang Feng
- Department of OncologyThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Xiaoyan Xu
- Department of Operating RoomTraditional Chinese Medicine Hospital of KunshanKunshanChina
| | - Liyan Jin
- Department of Thyroid and Breast SurgeryThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Guoqin Jiang
- Department of Thyroid and Breast SurgeryThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
44
|
Miller SR, Schipper M, Fritsche LG, Jiang R, Strohbehn G, Ötleş E, McMahon BH, Crivelli S, Zamora‐Resendiz R, Ramnath N, Yoo S, Dai X, Sankar K, Edwards DM, Allen SG, Green MD, Bryant AK. Pan-Cancer Survival Impact of Immune Checkpoint Inhibitors in a National Healthcare System. Cancer Med 2024; 13:e70379. [PMID: 39508134 PMCID: PMC11541111 DOI: 10.1002/cam4.70379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 10/13/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND The cumulative, health system-wide survival benefit of immune checkpoint inhibitors (ICIs) is unclear, particularly among real-world patients with limited life expectancies and among subgroups poorly represented on clinical trials. We sought to determine the health system-wide survival impact of ICIs. METHODS We identified all patients receiving PD-1/PD-L1 or CTLA-4 inhibitors from 2010 to 2023 in the national Veterans Health Administration (VHA) system (ICI cohort) and all patients who received non-ICI systemic therapy in the years before ICI approval (historical control). ICI and historical control cohorts were matched on multiple cancer-related prognostic factors, comorbidities, and demographics. The effect of ICI on overall survival was quantified with Cox regression incorporating matching weights. Cumulative life-years gained system-wide were calculated from the difference in adjusted 5-year restricted mean survival times. RESULTS There were 27,322 patients in the ICI cohort and 69,801 patients in the historical control cohort. Among ICI patients, the most common cancer types were NSCLC (46%) and melanoma (10%). ICI demonstrated a large OS benefit in most cancer types with heterogeneity across cancer types (NSCLC: adjusted HR [aHR] 0.56, 95% confidence interval [CI] 0.54-0.58, p < 0.001; urothelial: aHR 0.91, 95% CI 0.83-1.01, p = 0.066). The relative benefit of ICI was stable across patient age, comorbidity, and self-reported race subgroups. Across VHA, 15,859 life-years gained were attributable to ICI within 5-years of treatment, with NSCLC contributing the most life-years gained. CONCLUSION We demonstrated substantial increase in survival due to ICIs across a national health system, including in patient subgroups poorly represented on clinical trials.
Collapse
Affiliation(s)
- Sean R. Miller
- Department of Radiation OncologyVeterans Affairs Ann Arbor Healthcare SystemAnn ArborMichiganUSA
- Department of Radiation OncologyUniversity of MichiganAnn ArborMichiganUSA
| | - Matthew Schipper
- Department of BiostatisticsUniversity of MichiganAnn ArborMichiganUSA
| | - Lars G. Fritsche
- Department of BiostatisticsUniversity of MichiganAnn ArborMichiganUSA
- Center for Statistical GeneticsUniversity of MichiganAnn ArborMichiganUSA
| | - Ralph Jiang
- Department of BiostatisticsUniversity of MichiganAnn ArborMichiganUSA
| | - Garth Strohbehn
- Veterans Affairs Center for Clinical Management ResearchAnn ArborMichiganUSA
- Division of Medical Oncology, Department of MedicineVeterans Affairs Ann Arbor Healthcare SystemAnn ArborMichiganUSA
- Division of Hematology/Oncology, Department of MedicineUniversity of MichiganAnn ArborMichiganUSA
- Rogel Cancer CenterUniversity of MichiganAnn ArborMichiganUSA
| | - Erkin Ötleş
- Medical Scientist Training ProgramUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
| | - Benjamin H. McMahon
- Theoretical Biology and BiophysicsLos Alamos National LaboratoryLos AlamosNew MexicoUSA
| | - Silvia Crivelli
- Applied Mathematics and Computational Research DivisionLawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| | - Rafael Zamora‐Resendiz
- Applied Mathematics and Computational Research DivisionLawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| | - Nithya Ramnath
- Division of Medical Oncology, Department of MedicineVeterans Affairs Ann Arbor Healthcare SystemAnn ArborMichiganUSA
- Division of Hematology/Oncology, Department of MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Shinjae Yoo
- Computational Science InitiativeBrookhaven National LaboratoryUptonNew YorkUSA
| | - Xin Dai
- Computational Science InitiativeBrookhaven National LaboratoryUptonNew YorkUSA
| | - Kamya Sankar
- Division of Medical Oncology, Department of MedicineSamuel Oschin Comprehensive Cancer Institute, Cedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Donna M. Edwards
- Department of Radiation OncologyVeterans Affairs Ann Arbor Healthcare SystemAnn ArborMichiganUSA
- Department of Radiation OncologyUniversity of MichiganAnn ArborMichiganUSA
| | - Steven G. Allen
- Department of Radiation OncologyVeterans Affairs Ann Arbor Healthcare SystemAnn ArborMichiganUSA
- Department of Radiation OncologyUniversity of MichiganAnn ArborMichiganUSA
| | - Michael D. Green
- Department of Radiation OncologyVeterans Affairs Ann Arbor Healthcare SystemAnn ArborMichiganUSA
- Department of Radiation OncologyUniversity of MichiganAnn ArborMichiganUSA
| | - Alex K. Bryant
- Department of Radiation OncologyVeterans Affairs Ann Arbor Healthcare SystemAnn ArborMichiganUSA
- Department of Radiation OncologyUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
45
|
Huang H, Dai Z, Jiang Z, Li X, Ma L, Ji Z, Fan X. Anti-PD-1 immunotherapy for the treatment of metastatic urothelial carcinoma in a kidney transplant recipient: a case report. BMC Nephrol 2024; 25:390. [PMID: 39482589 PMCID: PMC11529296 DOI: 10.1186/s12882-024-03825-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/16/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitor (ICI) therapy has been widely investigated in urothelial carcinoma; however, the utility of ICI therapy in the treatment of organ transplant recipients with metastatic urothelial carcinoma (mUC) is unclear. We herein report the first case of a first-line anti-programmed cell death-1 (anti-PD-1) monotherapy for a kidney transplant patient with mUC. CASE PRESENTATION A 71-year-old woman who received a kidney transplant in 2003 was diagnosed with urothelial carcinoma in 2018. After operation of the tumor, the patient developed local recurrence at the site of the right kidney and bladder and multiple distant metastases in May 2020. Considering the intolerance of chemotherapy and high tumor mutation burden, we administered the anti-PD-1 agent tislelizumab (200 mg every three weeks). Partial response was achieved after two cycles of therapy and sustained until 18th cycles. There were no signs of kidney graft rejection. The immunotherapy was temporarily stopped after the 18th course because of a suspicious immune-related pneumonitis and was continued in December 2021. CONCLUSIONS This case demonstrates the feasibility of safely achieving stable cancer control in a kidney transplant patient with mUC without encountering graft rejection by using single-agent anti-PD-1 treatment.
Collapse
Affiliation(s)
- Houfeng Huang
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ziyi Dai
- Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ziming Jiang
- Eight-Year MD Program, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoyuan Li
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Ma
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhigang Ji
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Xinrong Fan
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
46
|
Yamada C, Tone K, Gochi M, Kimura H, Takagi M, Araya J. Renal Pelvic Cancer with Multiple Lung Metastases in a Patient with Polycystic Kidney Disease, Initially Diagnosed as Non-small Cell Lung Cancer: An Autopsy Case Report. Intern Med 2024:4377-24. [PMID: 39462594 DOI: 10.2169/internalmedicine.4377-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/29/2024] Open
Abstract
A 64-year-old man with autosomal dominant polycystic kidney disease (ADPKD) on hemodialysis presented with multiple lung masses. A computed tomography (CT)-guided biopsy revealed non-small-cell lung cancer (NSCLC). A cavitary mass in the right lung indicated primary NSCLC (cT2N1M1a, stage IVA). Pembrolizumab was initiated because of a high programmed death-ligand 1 (PD-L1) expression (90%). On day 10 post-treatment, he developed acute respiratory failure with diffuse ground-glass opacities on chest CT, indicative of pembrolizumab-induced lung injury. Despite steroid pulse therapy, the patient died on day 13. An autopsy revealed left renal pelvic cancer with lung metastases, highlighting the diagnostic challenges in ADPKD.
Collapse
Affiliation(s)
- Chieri Yamada
- Department of Respiratory Medicine, The Jikei University School of Medicine Kashiwa Hospital, Japan
| | - Kazuya Tone
- Department of Respiratory Medicine, The Jikei University School of Medicine Kashiwa Hospital, Japan
| | - Mina Gochi
- Department of Respiratory Medicine, The Jikei University School of Medicine Kashiwa Hospital, Japan
| | - Hiroko Kimura
- Department of Pathology, The Jikei University School of Medicine Kashiwa Hospital, Japan
| | - Masamichi Takagi
- Department of Respiratory Medicine, The Jikei University School of Medicine Kashiwa Hospital, Japan
| | - Jun Araya
- Division of Respiratory Diseases, Department of Internal Medicine, The Jikei University School of Medicine, Japan
| |
Collapse
|
47
|
Wang B, Shangguan W, Li W, Xie M, Yu Y, Yang Q, Sun Q, Xue J, Zhu Z, Zhu Y, Wu P. Blautia coccoides and its metabolic products enhance the efficacy of bladder cancer immunotherapy by promoting CD8 + T cell infiltration. J Transl Med 2024; 22:964. [PMID: 39449013 PMCID: PMC11515615 DOI: 10.1186/s12967-024-05762-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 10/10/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have emerged as a novel and effective treatment strategy, yet their effectiveness is limited to a subset of patients. The gut microbiota, recognized as a promising anticancer adjuvant, is being increasingly suggested to augment the efficacy of ICIs. Despite this, the causal link between the gut microbiota and the success of immunotherapy is not well understood. This gap in knowledge has driven us to identify beneficial microbiota and explore the underlying molecular mechanisms. METHODS Through 16S rDNA sequencing, we identified distinct gut microbiota in patients undergoing treatment with ICIs. Following this, we assessed the impact of probiotics on anti-PD-1 therapy in bladder cancer using mouse models, employing a multi-omics strategy. Subsequently, we uncovered the mechanisms through which Blautia-produced metabolites enhance antitumor immunity, utilizing untargeted metabolomics and a range of molecular biology techniques. RESULTS In our research, the LEfSe analysis revealed a significant enrichment of the Blautia genus in the gut microbiota of patients who responded to immunotherapy. We discovered that the external addition of Blautia coccoides hampers tumor growth in a bladder cancer mouse model by enhancing the infiltration of CD8+ T cells within the tumor microenvironment (TME). Further investigations through untargeted metabolomics and molecular biology experiments showed that oral administration of Blautia coccoides elevated trigonelline levels. This, in turn, suppresses the β-catenin expression both in vitro and in vivo, thereby augmenting the cancer-killing activity of CD8+ T cells. CONCLUSIONS This research provided valuable insights into enhancing the efficacy of PD-1 inhibitors in clinical settings. It was suggested that applying Blautia coccoides and its metabolic product, trigonelline, could serve as a synergistic treatment method with PD-1 inhibitors in clinical applications.
Collapse
Affiliation(s)
- Benlin Wang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wentai Shangguan
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weijia Li
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ming Xie
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yao Yu
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qishen Yang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qi Sun
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jingwen Xue
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhangrui Zhu
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuexuan Zhu
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Peng Wu
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
48
|
Stockem CF, Einerhand SMH, Rodríguez IM, Salhi Y, Pérez E, Bakaloudi DR, Talukder R, Caramelo B, Morales-Barrera R, De Meulenaere A, Rametta A, Bottelli A, Lefort F, Giannatempo P, Vulsteke C, Carles J, Duran I, Grivas P, de Liaño AG, Robbrecht DGJ, Valderrama BP, van der Noort V, van der Heijden MS. Long-term survival following anti-PD-(L)1 monotherapy in advanced urothelial cancer and an assessment of potential prognostic clinical factors: a multicentre observational study. BJC REPORTS 2024; 2:84. [PMID: 39516359 PMCID: PMC11523962 DOI: 10.1038/s44276-024-00104-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Anti-PD-(L)1 agent are approved as first- and second-line treatment options in advanced urothelial cancer (UC), but information about long-term survival is scarce. There is a need for prognostic factors, as these may help in the decision-making concerning anti-PD-(L)1 in patients with UC. Here, we examined long-term survival following anti-PD-(L)1 in advanced UC and assessed clinical factors for their correlation with survival. METHODS We collected data from patients with advanced UC treated with anti-PD-(L)1 between 2013 and 2023. Overall- and progression-free survival (OS, PFS) were determined using the Kaplan-Meier method. Independent variables were analysed by uni- and multivariate Cox regression for their association with OS and PFS. RESULTS Survival analyses included 552 patients. Patient characteristics in our cohort were consistent with those of a typical advanced UC population. After median follow-up of 49 months, five-year OS and PFS rates were 16.0% and 6.9% respectively. The absence of visceral and/or bone metastases and elevated C-reactive protein level, gamma-glutamyltransferase level and neutrophil-to-lymphocyte ratio were identified as favourable prognostic factors for OS. CONCLUSIONS A selected subset of patients with advanced UC may experience long-term clinical benefit from anti-PD-(L)1 treatment. We identified prognostic factors that might be used for risk assessment and clinical trial stratification.
Collapse
Affiliation(s)
- Chantal F Stockem
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Sarah M H Einerhand
- Department of surgical oncology (urology), The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Youssra Salhi
- Department of Medical Oncology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Esther Pérez
- Department of Medical Oncology, Hospital Complex Insular-Materno Infantil, Las Palmas, Spain
| | - Dimitra R Bakaloudi
- Department of Medicine, Division of Hematology and Oncology, University of Washington, Seattle, USA
| | - Rafee Talukder
- Department of Medicine, Division of Hematology and Oncology, University of Washington, Seattle, USA
| | - Belen Caramelo
- Department of Medical Oncology, Hospital Universitario Marqués de Valdecilla, Santander, Cantabria, Spain
| | - Rafael Morales-Barrera
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
- Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Astrid De Meulenaere
- Department of Medical Oncology, Integrated Cancer Center, AZ Maria Middelares, Gent, Belgium
| | - Alessandro Rametta
- Oncologia Medica Genito-Urinaria, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Andrea Bottelli
- Oncologia Medica Genito-Urinaria, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Felix Lefort
- Department of Medical Oncology, Hôspital Saint André-CHU de Bordeaux, Bordeaux, France
| | - Patrizia Giannatempo
- Oncologia Medica Genito-Urinaria, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Christof Vulsteke
- Department of Medical Oncology, Integrated Cancer Center, AZ Maria Middelares, Gent, Belgium
- Centre for Oncological Research (CORE), Antwerp University, Antwerp, Belgium
| | - Joan Carles
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
- Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Ignacio Duran
- Department of Medical Oncology, Hospital Universitario Marqués de Valdecilla, Santander, Cantabria, Spain
| | - Petros Grivas
- Department of Medicine, Division of Hematology and Oncology, University of Washington, Seattle, USA
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Alfonso Gómez de Liaño
- Department of Medical Oncology, Hospital Complex Insular-Materno Infantil, Las Palmas, Spain
| | - Debbie G J Robbrecht
- Department of Medical Oncology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Begoña P Valderrama
- Department of Medical Oncology, Hospital Universitario Virgen del Rocio, Seville, Spain
| | - Vincent van der Noort
- Department of Biometrics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | |
Collapse
|
49
|
Tang B, Xiao J, Chi Z, Duan R, Cui C, Si L, Liu Y, Hu X, Liu Z, Xiang P, Li S, Yan X, Zhou L, Li J, Li Y, Yu X, Dai X, Li X, Guo J, Sheng X. Phase Ib study of anti-PD-L1 monoclonal antibody socazolimab in combination with nab-paclitaxel as first-line therapy for advanced urothelial carcinoma. Oncologist 2024:oyae260. [PMID: 39418340 DOI: 10.1093/oncolo/oyae260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 04/20/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND PD-1/PD-L1 immune checkpoint inhibitors (ICIs) have demonstrated activity in the post-platinum and platinum-ineligible settings for advanced urothelial carcinoma (aUC). As only around 50% of patients with aUC can tolerate platinum-containing treatment, treatments combining first-line ICIs with non-platinum drugs are urgently needed. Therefore, we assessed the safety and efficacy of the anti-PD-L1 monoclonal antibody Socazolimab in combination with nab-paclitaxel as first-line therapy in aUC (NCT04603846). METHODS This was a multi-center, single-arm, phase Ib study that enrolled patients with treatment-naive aUC. Patients received Socazolimab (5 mg/kg) and nab-paclitaxel (260 mg/m2) Q3w. The primary endpoint was safety and tolerability of the combination regimen. Second endpoints were the objective response rate (ORR) and progression-free survival. RESULTS Between September, 2020 and September, 2021, 20 patients with urothelial carcinoma were enrolled, arising from renal pelvis (5), bladder (8), and ureter (7). After a median follow-up of 17 months, the median number of treatment cycles was 12. No patients had dose limiting toxicity. All patients had treatment-related adverse events (TRAEs), most of which were grade 1 or 2. The common TRAEs (≥20%) were peripheral neurotoxicity, alopecia, rash, increased ALT, weight loss, weakness, pruritus, increased AST, increased γGT, increased ALP, neutropenia, emesis, and anorexia. Nine patients (45%) developed grade 3 TRAEs including peripheral neurotoxicity (30.0%), increased ALT (10.0%), and increased γGT (5.0%). Two patients (10%) discontinued treatment because of grade 3 mouth ulcer (n = 1) and grade 2 lung fibrosis (n = 1). No grade 4-5 TRAEs were observed. Among the 17 patients who had received at least one tumor assessment, ORR was 58.8% (95% CI, 32.9%-81.6%) and the median progression-free survival was 8.3 months (95% CI, 5.2-19.5). The median duration of response was 13.3 months (95% CI, 2.0-20.1), and the overall survival was 19.5 months (95% CI, 11.2-not reached). CONCLUSION Socazolimab combined with nab-paclitaxel has shown good safety and promising antitumor activity as first-line therapy in patients with advanced urothelial carcinoma.
Collapse
Affiliation(s)
- Bixia Tang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Melanoma and Soft Tissue Sarcoma, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Jun Xiao
- Department of Urology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, People's Republic of China
| | - Zhihong Chi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Melanoma and Soft Tissue Sarcoma, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Rong Duan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genitourinary Oncology, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Chuanliang Cui
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genitourinary Oncology, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Lu Si
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Melanoma and Soft Tissue Sarcoma, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Yixun Liu
- Department of Urology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, People's Republic of China
| | - Xuechun Hu
- Department of Urology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, People's Republic of China
| | - Zhi Liu
- Department of Urology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, People's Republic of China
| | - Ping Xiang
- Department of Urology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, People's Republic of China
| | - Siming Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genitourinary Oncology, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Xieqiao Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genitourinary Oncology, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Li Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genitourinary Oncology, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Juan Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genitourinary Oncology, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Yujie Li
- Zhaoke (Guangzhou) Oncology Pharmaceutical Limited, Guangzhou 511400, People's Republic of China
| | - Xiaohui Yu
- Zhaoke (Guangzhou) Oncology Pharmaceutical Limited, Guangzhou 511400, People's Republic of China
| | - Xiangrong Dai
- Zhaoke (Guangzhou) Oncology Pharmaceutical Limited, Guangzhou 511400, People's Republic of China
| | - Xiaoyi Li
- Zhaoke (Guangzhou) Oncology Pharmaceutical Limited, Guangzhou 511400, People's Republic of China
| | - Jun Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genitourinary Oncology, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Xinan Sheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genitourinary Oncology, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| |
Collapse
|
50
|
Bloom MW, Vo JB, Rodgers JE, Ferrari AM, Nohria A, Deswal A, Cheng RK, Kittleson MM, Upshaw JN, Palaskas N, Blaes A, Brown SA, Ky B, Lenihan D, Maurer MS, Fadol A, Skurka K, Cambareri C, Chauhan C, Barac A. Cardio-Oncology and Heart Failure: a Scientific Statement From the Heart Failure Society of America. J Card Fail 2024:S1071-9164(24)00363-4. [PMID: 39419165 DOI: 10.1016/j.cardfail.2024.08.045] [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: 04/16/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 10/19/2024]
Abstract
Heart failure and cancer remain 2 of the leading causes of morbidity and mortality, and the 2 disease entities are linked in a complex manner. Patients with cancer are at increased risk of cardiovascular complications related to the cancer therapies. The presence of cardiomyopathy or heart failure in a patient with new cancer diagnosis portends a high risk for adverse oncology and cardiovascular outcomes. With the rapid growth of cancer therapies, many of which interfere with cardiovascular homeostasis, heart failure practitioners need to be familiar with prevention, risk stratification, diagnosis, and management strategies in cardio-oncology. This Heart Failure Society of America statement addresses the complexities of heart failure care among patients with active cancer diagnoses and cancer survivors. Risk stratification, monitoring and management of cardiotoxicity are presented across stages A through D heart failure, with focused discussion on heart failure with preserved ejection fraction and special populations, such as survivors of childhood and young-adulthood cancers. We provide an overview of the shared risk factors between cancer and heart failure, highlighting heart failure as a form of cardiotoxicity associated with many different cancer therapeutics. Finally, we discuss disparities in the care of patients with cancer and cardiac disease and present a framework for a multidisciplinary-team approach and critical collaboration among heart failure, oncology, palliative care, pharmacy, and nursing teams in the management of these complex patients.
Collapse
Affiliation(s)
| | - Jacqueline B Vo
- Radiation Epidemiology Branch, Division of Cancer Epidemiology & Genetics, National Cancer Institute, Bethesda, MD
| | - Jo E Rodgers
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, Chapel Hill, NC
| | - Alana M Ferrari
- Division of Hematology/ Oncology, University of Virginia Health, Charlottesville, VA
| | - Anju Nohria
- Cardio-Oncology Program, Cardiovascular Division, Brigham and Women's Hospital, Boston, MA
| | - Anita Deswal
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Richard K Cheng
- Division of Cardiology, University of Washington, Seattle, WA
| | - Michelle M Kittleson
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | | | - Nicolas Palaskas
- Department of Cardiology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Anne Blaes
- Division of Hematology/Oncology/Transplantation, University of Minnesota, Minneapolis, MN
| | - Sherry-Ann Brown
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI; Research Collaborator, Mayo Clinic, Rochester, MN
| | - Bonnie Ky
- Division of Cardiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; Thalheimer Center for Cardio-Oncology, Abramson Cancer Center and Division of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Daniel Lenihan
- Saint Francis Healthcare, Cape Girardeau, MO and the International Cardio-Oncology Society, Tampa, FL
| | - Mathew S Maurer
- Division of Cardiology, Columbia University Irving Medical Center, New York, NY
| | | | | | - Christine Cambareri
- Clinical Oncology Pharmacist, Hospital of the University of Pennsylvania, Abramson Cancer Center, Philadelphia, PA
| | | | - Ana Barac
- Department of Cardiology, Inova Schar Heart and Vascular, Inova Schar Cancer, Falls Church, VA
| |
Collapse
|