1
|
Wekking D, Silva CAC, Viscò R, Denaro N, Lambertini M, Maccioni A, Loddo E, Willard-Gallo K, Scartozzi M, Derosa L, Solinas C. The interplay between gut microbiota, antibiotics, and immune checkpoint inhibitors in patients with cancer: A narrative review with biological and clinical aspects. Crit Rev Oncol Hematol 2025; 212:104767. [PMID: 40414545 DOI: 10.1016/j.critrevonc.2025.104767] [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: 04/05/2025] [Revised: 05/11/2025] [Accepted: 05/13/2025] [Indexed: 05/27/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) targeting the programmed cell death-1 (PD-1), programmed cell death ligand-1 (PD-L1), and cytotoxic T-lymphocyte antigen 4 (CTLA-4) pathways have revolutionized cancer therapy. However, primary and secondary resistance to ICI pose significant challenges. Recent studies underscore the critical role of gut microbiota (GM) in modulating ICI efficacy by shaping host immune responses and regulating the tumor microenvironment (TME). The composition of the GM has been linked to ICI treatment outcomes, with certain microbial taxa, such as Faecalibacterium spp., Bifidobacterium spp., Eubacterium spp., Roseburia spp., and Akkermansia muciniphila, associated with favorable responses. Mechanistically, the GM affects immune responses via multiple pathways, including induction of T cell differentiation, promotion of anti- or proinflammatory cytokine environments, and enhancement of T cell priming and effector functions. Moreover, microbial-derived metabolites play a role in shaping tumor immune responses and influencing ICI efficacy. Antibiotic treatment can disrupt GM diversity and composition (gut dysbiosis), potentially diminishing ICI effectiveness. A deeper understanding of the interplay between GM, antibiotic treatment, and ICI efficacy is crucial for developing personalized therapeutic strategies to improve patient outcomes. Herein, we review current evidence on the association between specific microbial taxa and tumor immunosurveillance, the impact of antibiotics on the GM composition and immune modulation, and its implications for ICI therapy efficacy.
Collapse
Affiliation(s)
- Demi Wekking
- Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.
| | - Carolina Alves Costa Silva
- Gustave Roussy Cancer Campus (GRCC), Clinicobiome, Villejuif, France; Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Roberto Viscò
- Ospedale Sant'Antonio Abate, Patologica Clinica, ASP Trapani, Italy
| | - Nerina Denaro
- Medical Oncology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Italy
| | - Matteo Lambertini
- Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genova, Genova, Italy; Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Antonio Maccioni
- Medical Oncology AOU Cagliari Policlinico Duilio Casula, Monserrato (CA) Italy
| | - Erica Loddo
- Gastroenterology University Hospital, Cagliari, Italy
| | | | - Mario Scartozzi
- Medical Oncology AOU Cagliari Policlinico Duilio Casula, Monserrato (CA) Italy; University Hospital of Cagliari, Italy
| | - Lisa Derosa
- Gustave Roussy Cancer Campus (GRCC), Clinicobiome, Villejuif, France; Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France; Faculté de Médecine, Université Paris-Saclay, Kremlin-Bicêtre, France
| | - Cinzia Solinas
- Medical Oncology AOU Cagliari Policlinico Duilio Casula, Monserrato (CA) Italy
| |
Collapse
|
2
|
Stouvenot M, Koch S, Frontzcak A, D'Engremont C, Boinette A, Doussot A, Maurina T, Vuitton L. Effectiveness and safety of endoscopic ultrasound-guided radiofrequency ablation for pancreatic metastases of renal cell carcinoma. Endosc Int Open 2025; 13:a25667350. [PMID: 40376026 PMCID: PMC12080524 DOI: 10.1055/a-2566-7350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 01/25/2025] [Indexed: 05/18/2025] Open
Abstract
Background and study aims Pancreatic metastases from renal cell carcinoma (RCC) are usually managed surgically but with significant morbidity. As an alternative, endoscopic ultrasound-guided radiofrequency ablation (EUS-RFA) has shown promising results in treatment of pancreatic neuroendocrine tumors. The aim of our study was to assess technical success, effectiveness, and safety of EUS-RFA in patients with pancreatic metastases of RCC. Patients and methods This retrospective, observational study included consecutive patients referred for EUS-RFA of pancreatic RCC metastases. EUS-RFA was performed through 18G or 19G dedicated RFA needles. Effectiveness of EUS-RFA treatment was defined by necrosis with no contrast enhancement or lesion disappearance, determined by contrast-enhanced computed tomography (CT) scan, at 2 to 5 months post procedure, 1 year, and at the end of follow-up. Safety was assessed per and post procedure. Results Between January 2015 and January 2021, eight patients with 11 lesions were treated and median time from RCC diagnosis to pancreatic metastases RFA was 8.5 years (1-15). Mean lesion size was 13.9 mm (± 3.9). Technical success assessed by immediate post procedure contrast-enhanced CT or Doppler was 100%. At the first CT scan follow-up, complete response was 45.4% and partial response was 27.3%. At 1 year, complete response was 45.4% and partial response was 27.3%. Three patients had multiple EUS-RFAs. Adverse events occurred in 3 patients (mild acute pancreatitis, abdominal pain, and pancreatic fistula with retro-gastric pseudocyst). Conclusions Our study demonstrated the feasibility and safety of EUS-RFA for patients with pancreatic metastases of RCC.
Collapse
Affiliation(s)
- Morgane Stouvenot
- Gastroenterology, Centre Hospitalier Régional Universitaire de Besançon, Besançon, France
| | - Stephane Koch
- Gastroenterology, Centre Hospitalier Régional Universitaire de Besançon, Besançon, France
| | - Alexandre Frontzcak
- Urology, Centre Hospitalier Régional Universitaire de Besançon, Besançon, France
| | | | - Aurélien Boinette
- Gastroenterology, Centre Hospitalier Régional Universitaire de Besançon, Besançon, France
| | - Alexandre Doussot
- Digestive Surgery, Centre Hospitalier Régional Universitaire de Besançon, Besançon, France
| | - Tristan Maurina
- Oncology, Centre Hospitalier Régional Universitaire de Besançon, Besançon, France
| | - Lucine Vuitton
- Gastroenterology, Centre Hospitalier Régional Universitaire de Besançon, Besançon, France
| |
Collapse
|
3
|
Pinto-Marín Á, Trilla-Fuertes L, Miranda Poma J, Vasudev NS, García-Fernández E, López-Vacas R, Miranda N, Wilson M, López-Camacho E, Pertejo A, Dittmann A, Kunz L, Brown J, Pedroche-Just Y, Zapater-Moros A, de Velasco G, Castellano D, González-Peramato P, Espinosa E, Banks RE, Fresno Vara JÁ, Gámez-Pozo A. A prognostic microRNA-based signature for localized clear cell renal cell carcinoma: the Bio-miR study. Br J Cancer 2025:10.1038/s41416-025-03008-2. [PMID: 40335662 DOI: 10.1038/s41416-025-03008-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 03/18/2025] [Accepted: 03/28/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Two thirds of renal cell carcinoma (RCC) patients have localized disease at diagnosis. A significant proportion of these patients will relapse. There is a need for prognostic biomarkers to improve risk-stratification and specific treatments for patients that relapse. The objective of this study is to determine the clinical utility of microRNA signatures as prognostic biomarkers in localized clear cell RCC (ccRCC) and propose new therapeutic targets in patients with a high-risk of relapse. PATIENTS AND METHODS The microRNA profiles from a discovery cohort of 71 T1-T2 ccRCC patients (n = 88) were analyzed using microarrays. MicroRNAs prognostic value was established, and a microRNAs signature predicting relapse for T1b-T3 disease was defined. Independent validation was carried out by qPCR in cohorts from UK (n = 75) and Spain (n = 180), and the TCGA cohort (n = 175). In the Spanish validation cohort, proteomics experiments were done. Proteins were extracted from FFPE tissue and analyzed using by data-independent acquisition mass spectrometry. Additionally, ccRCC TCGA RNA-seq data was also analyzed. Both protein and RNA-seq data was analyzed using Significance Analysis of Micorarrays (SAM) and probabilistic graphical models, which allow the identification of relevant biological processes between low and high-risk tumors. RESULTS A 9-microRNAs signature, Bio-miR, classified patients into low- and high-risk with disease-free survival (DFS) at 5 years of 87.12 vs. 54.17% respectively (p = 0.0086, HR = 3.58, 95%CI: 1.37-8.3). Results were confirmed in the validation cohorts with 5-year DFS rates of 94% vs. 62% in the UK cohort (HR = 7.14, p = 0.001), 82.9% vs. 58.7% in the Spanish cohort (HR = 2.46, p = 0.0013), and 5-year overall survival rates of 72.7% vs. 44.5% in the TCGA cohort (HR = 2.43, p = 0.0012). Among low-risk patients according to adjuvant immunotherapy clinical trial criteria, Bio-miR identified a high-risk group. Maybe those patients ought to be considered to receive adjuvant therapy. Proteins overexpressed in the high-risk group were mainly related to focal adhesion, serine and inositol metabolism, and angiogenesis. Probabilistic graphical models defined eight functional nodes related to specific biological processes. Differences between low- and high-risk tumors were detected in complement activation and translation functional nodes. In ccRCC TCGA cohort, 676 genes were differentially expressed between low and high-risk patients, mainly related to complement activation, adhesion, and chemokine and cytokine cascades. In this case, probabilistic graphical models defined ten functional nodes. Calcium binding, membrane, adhesion, extracellular matrix, blood microparticle, inflammatory response and immune response had higher functional node activity, and metabolism node, containing genes related to retinol and xenobiotic and CYP450 metabolism, had lower activity in the high-risk group. CONCLUSIONS Bio-miR dichotomizes ccRCC patients with non-metastatic disease into those with low- and high-risk of relapse. This has implications for treatment and follow-up, identifying patients most likely to benefit from adjuvant treatment in clinical trials, preventing unnecessary exposure to side-effects, and providing health economics benefits. Additionally, promising therapeutic targets, as angiogenesis, immune response, metabolism, or complement activation, were found deregulated in high-risk ccRCC patients defined by Bio-miR. These findings may be useful to select patients for tailored, molecularly-driven clinical trials. Identifying which patients with kidney cancer are most at risk of their cancer coming back after surgery is critical, so that they can be prioritized for early treatment. We have identified a combination of biomarkers present in the cancer tissue (called BiomiR) which can help to do this.
Collapse
Affiliation(s)
- Álvaro Pinto-Marín
- Medical Oncology Service, Hospital Universitario La Paz, IDIPAZ, Madrid, Spain.
| | | | - Jesús Miranda Poma
- Medical Oncology Service, Hospital Universitario La Paz, IDIPAZ, Madrid, Spain
| | - Naveen S Vasudev
- Leeds Institute of Medical Research at St James's, University of Leeds, St. James's University Hospital, Beckett Street, Leeds, LS9 7TF, UK
| | | | - Rocío López-Vacas
- Molecular Oncology Lab, Hospital Universitario La Paz, IDIPAZ, Madrid, Spain
| | - Natalia Miranda
- Urology Service, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Michelle Wilson
- Leeds Institute of Medical Research at St James's, University of Leeds, St. James's University Hospital, Beckett Street, Leeds, LS9 7TF, UK
| | | | - Ana Pertejo
- Medical Oncology Service, Hospital Universitario La Paz, IDIPAZ, Madrid, Spain
| | - Antje Dittmann
- Proteomics Group, Functional Genomics Center Zurich, Zurich, Switzerland
| | - Laura Kunz
- Proteomics Group, Functional Genomics Center Zurich, Zurich, Switzerland
| | - Joanne Brown
- Leeds Institute of Medical Research at St James's, University of Leeds, St. James's University Hospital, Beckett Street, Leeds, LS9 7TF, UK
| | | | | | | | - Daniel Castellano
- Medical Oncology Service, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | - Enrique Espinosa
- Medical Oncology Service, Hospital Universitario La Paz, IDIPAZ, Madrid, Spain
- Biomedical Research Networking Center on Oncology-CIBERONC, ISCIII, Madrid, Spain
| | - Rosamonde E Banks
- Leeds Institute of Medical Research at St James's, University of Leeds, St. James's University Hospital, Beckett Street, Leeds, LS9 7TF, UK
| | - Juan Ángel Fresno Vara
- Molecular Oncology Lab, Hospital Universitario La Paz, IDIPAZ, Madrid, Spain
- Biomedical Research Networking Center on Oncology-CIBERONC, ISCIII, Madrid, Spain
| | - Angelo Gámez-Pozo
- Molecular Oncology Lab, Hospital Universitario La Paz, IDIPAZ, Madrid, Spain.
- Biomedica Molecular Medicine SL, Madrid, Spain.
| |
Collapse
|
4
|
Hugaboom MB, Wirth LV, Street K, Ruthen N, Jegede OA, Schindler NR, Shah V, Zaemes JP, Ahmar NE, Matar S, Savla V, Choueiri TK, Denize T, West DJ, McDermott DF, Plimack ER, Sosman JA, Haas NB, Stein MN, Alter R, Bilen MA, Hurwitz ME, Hammers H, Signoretti S, Atkins MB, Wu CJ, Braun DA. Presence of Tertiary Lymphoid Structures and Exhausted Tissue-Resident T Cells Determines Clinical Response to PD-1 Blockade in Renal Cell Carcinoma. Cancer Discov 2025; 15:948-968. [PMID: 39992403 PMCID: PMC12048281 DOI: 10.1158/2159-8290.cd-24-0991] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 01/08/2025] [Accepted: 02/21/2025] [Indexed: 02/25/2025]
Abstract
SIGNIFICANCE We describe a paradigm wherein combined high TLS and low tissue-resident exhausted CD8+ T cells are required for optimal response to PD-1 blockade in RCC. This analysis identifies key determinants of response to PD-1 blockade in advanced RCC and suggests avenues for future immune modulation through rational combination therapy strategies.
Collapse
Affiliation(s)
- Miya B. Hugaboom
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Lena V. Wirth
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Kelly Street
- Division of Biostatistics, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Neil Ruthen
- Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Opeyemi A. Jegede
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Valisha Shah
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jacob P. Zaemes
- Georgetown Lombardi Comprehensive Cancer Center, Washington, DC, USA
| | - Nourhan El Ahmar
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Sayed Matar
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Varunika Savla
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Toni K. Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Thomas Denize
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Destiny J. West
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - David F. McDermott
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | - Jeffrey A. Sosman
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | - Naomi B. Haas
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Mark N. Stein
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Robert Alter
- Hackensack University Medical Center, Hackensack, NJ, USA
| | - Mehmet A. Bilen
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Michael E. Hurwitz
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Hans Hammers
- Department of Internal Medicine, Division of Hematology and Oncology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Sabina Signoretti
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Michael B. Atkins
- Georgetown Lombardi Comprehensive Cancer Center, Washington, DC, USA
| | - Catherine J. Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - David A. Braun
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
5
|
Bi Y, Wei H, Ma Q, Wang R, Jin J, Qu K, Liu Y, Zhai Z, Zhu L, Wang J. The fragility index of randomized controlled trials in advanced/metastatic renal cell cancer. Urol Oncol 2025; 43:333.e9-333.e15. [PMID: 40155257 DOI: 10.1016/j.urolonc.2025.03.003] [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: 09/13/2024] [Revised: 02/06/2025] [Accepted: 03/02/2025] [Indexed: 04/01/2025]
Abstract
PURPOSE The fragility index (FI) has been applied as a supplement to the noncomprehensive P-values to assess the robustness of randomized controlled trials (RCTs). The objective of this study is to evaluate the statistical robustness of RCTs of advanced/metastatic renal cell cancer (a/mRCC) using the FI. MATERIALS AND METHODS RCTs related to a/mRCC published in the 4 highest-impact general medical journals and the 25 highest-impact urological journals between January 1, 2000, and December 31, 2023, were identified from PubMed database. The FI was calculated by using Fisher's exact test. Spearman's correlation analysis was conducted to assess potential correlates regarding FI. RESULTS 16 eligible RCTs were screened with a median total sample size of 654.5 (IQR, 461-847) and a median patients lost to follow-up of 14 (IQR, 3-23). The median FI was 12.5 (IQR, 8.5-27), suggesting that a switch in outcomes in only 13 patients would have reversed the significance of the trials. The number of patients lost to follow-up exceeded or equaled to the FI in 7 (44%) RCTs. P-values were negatively associated with the FI, while the number of patients lost to follow-up and patients enrolled were not statistically significant. CONCLUSION Not all RCTs associated with a/mRCC are as statistically robust as previously considered and should therefore be construed carefully. We suggest that additional reporting of FI in urological RCTs as a supplement to the P-value to assist readers in concluding reliably by considering the fragility of the outcomes.
Collapse
Affiliation(s)
- Yingwei Bi
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Haotian Wei
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300202, China
| | - Qifeng Ma
- College of Basic Medicine, Dalian Medical University, Dalian 116041, China
| | - Rui Wang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Jiacheng Jin
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Kexin Qu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yuxin Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Ziwei Zhai
- College of Basic Medicine, Dalian Medical University, Dalian 116041, China
| | - Liang Zhu
- College of Basic Medicine, Dalian Medical University, Dalian 116041, China; College of Basic Medicine, Dalian University of Technology, Dalian 116081, China.
| | - Jianbo Wang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| |
Collapse
|
6
|
Osorio L, Grazioso TP, de Velasco G, Etxaniz O, Pérez-Gracia JL, Pinto Á, Durán I, Grande E, Garcia PB, Lázaro M, Rodriguez L, Villalobos ML, García L, Cuellar A, Solís-Hernández MP, Pernaut C, Rodríguez-Moreno JF, Rodriguez-Antona C, García-Donas J. Retrospective study assessing the role of the androgen receptor in clear cell renal cell cancer patients treated with VEGFR inhibitors in monotherapy. Clin Transl Oncol 2025; 27:2241-2255. [PMID: 39365364 DOI: 10.1007/s12094-024-03652-9] [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: 06/14/2024] [Accepted: 07/27/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND AND PURPOSE Despite that incorporating antiangiogenic in combination with immune-checkpoint inhibitors as the standard first-line treatment for advanced clear cell renal cell cancer (ccRCC) yields promising outcomes, these regimens often lead to significant toxicity. However, a subgroup of patients has shown responsiveness to VEGFR tyrosine-kinase inhibitors (TKIs) in monotherapy, leading to the question of whether employing combination therapies can significantly enhance overall survival in all patients over monotherapy. Thus, we aim to identify gene expression signatures that can predict TKI response within subpopulations that might benefit from single-agent therapies, to minimize unnecessary exposure to combination therapies and their associated toxicities, as well as to discover new potential therapeutic targets to improve ccRCC treatment. Based on prior data, the androgen receptor (AR) might meet both conditions. PATIENTS AND METHODS We evaluated the association between AR expression, assessed through NanoString® technology-derived mRNA counts, and the clinical outcomes of 98 ccRCC patients treated with first-line antiangiogenics and determined its association with other genes implicated in ccRCC tumorigenesis. RESULTS Higher AR-expression correlates significantly with better prognosis and survival based on the MSKCC risk score, and longer PFS. Furthermore, we have identified a gene set signature associated with AR-overexpression and several genes involved in angiogenesis and transcriptional targets of the hypoxia-inducible factor, a cornerstone of ccRCC. CONCLUSIONS AR-overexpression and its association with other genes could favor a transcriptomic signature set to aid in identifying patients suitable for TKI in monotherapy, rather than aggressive combinations, enhancing thus, precision and personalized therapeutic decisions.
Collapse
Affiliation(s)
- Lucia Osorio
- Servicio de Urología, Urología Hospitalaria, Hospital HM La Rosaleda, Santiago de Compostela, Spain
- Instituto de Investigación Sanitaria HM Hospitales (IISHM), Madrid, Spain
| | - Tatiana P Grazioso
- Instituto de Investigación Sanitaria HM Hospitales (IISHM), Madrid, Spain
- Laboratory of Innovation in Oncology, Gynecological, Genitourinary and Skin Cancer Unit, HM CIOCC, Centro Integral Oncológico Clara Campal, Hospital Universitario HM Sanchinarro, HM Hospitales, Madrid, Spain
- Institute of Applied Molecular Medicine (IMMA), Department of Basic Medical Sciences, Facultad de Medicina, Universidad San Pablo CEU, Madrid, Spain
| | | | - Olatz Etxaniz
- Grupo de Investigación Aplicada en Oncología de Badalona (B·ARGO), Hospital Germá Trials I Pujol, Barcelona, Spain
| | | | - Álvaro Pinto
- Medical Oncology Department, Hospital Universitario La Paz - IdiPAZ, Madrid, Spain
| | - Ignacio Durán
- Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - Enrique Grande
- Medical Oncology Department, MD Anderson Cancer Center Madrid, Madrid, Spain
| | | | | | | | | | | | | | | | | | - Juan Francisco Rodríguez-Moreno
- Instituto de Investigación Sanitaria HM Hospitales (IISHM), Madrid, Spain
- Laboratory of Innovation in Oncology, Gynecological, Genitourinary and Skin Cancer Unit, HM CIOCC, Centro Integral Oncológico Clara Campal, Hospital Universitario HM Sanchinarro, HM Hospitales, Madrid, Spain
- Institute of Applied Molecular Medicine (IMMA), Department of Basic Medical Sciences, Facultad de Medicina, Universidad San Pablo CEU, Madrid, Spain
| | - Cristina Rodriguez-Antona
- Pharmacogenomics and Tumor Biomarkers Group, Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM) CSIC/UAM, Madrid, Spain.
- Grupo de Cáncer Endocirno Hereditario, Centro Nacional de Investigaciones Oncológicas, CNIO, Madrid, Spain.
| | - Jesús García-Donas
- Instituto de Investigación Sanitaria HM Hospitales (IISHM), Madrid, Spain.
- Laboratory of Innovation in Oncology, Gynecological, Genitourinary and Skin Cancer Unit, HM CIOCC, Centro Integral Oncológico Clara Campal, Hospital Universitario HM Sanchinarro, HM Hospitales, Madrid, Spain.
- Institute of Applied Molecular Medicine (IMMA), Department of Basic Medical Sciences, Facultad de Medicina, Universidad San Pablo CEU, Madrid, Spain.
| |
Collapse
|
7
|
Liu M, Gao Y, Li L, Ge L, Yao L, Chu X, Wu F, Tian J. Anti-TIGIT inhibitors plus PD-1 or PD-L1 inhibitors versus PD-1 or PD-L1 inhibitors for first-line treatment of advanced non-small cell lung cancer. Cochrane Database Syst Rev 2025; 4:CD015888. [PMID: 40226904 PMCID: PMC11995688 DOI: 10.1002/14651858.cd015888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
OBJECTIVES This is a protocol for a Cochrane Review (intervention). The objectives are as follows: To evaluate the effectiveness and safety of anti-TIGIT inhibitors in combination with PD-1 inhibitors or PD-L1 inhibitors compared to treatment with PD-1 inhibitors or PD-L1 inhibitors in the first-line treatment of advanced NSCLC. To assess the effectiveness and safety of anti-TIGIT inhibitors in combination with chemotherapy and PD-1 inhibitors or PD-L1 inhibitors compared to treatment with chemotherapy plus PD-1 inhibitors or PD-L1 inhibitors in the first-line treatment of advanced NSCLC.
Collapse
Affiliation(s)
- Ming Liu
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou city, China
| | - Ya Gao
- Department of Medical Data, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lun Li
- Department of Breast Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Long Ge
- Evidence-Based Social Science Research Centre, School of Public Health, Lanzhou University, Lanzhou city, China
| | - Liang Yao
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
| | - Xiajing Chu
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada
| | - Fanqi Wu
- Department of Respirator, Lanzhou University Second Hospital, Lanzhou, China
| | - Jinhui Tian
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou city, China
| |
Collapse
|
8
|
Pan Z, Liu D, Cao J, Fu L, Zhang X, Zhu X, Pan Y, Liu J, Han C, Jin R, Shen S, Zhang X, Liu H, Yang X, Hu K, Shi X, Wang D, Zhao Y, Zhong J, Xiang B, Gu S, Li T, Zhang S, Zhou L, Zhao H, Zeng Y, Wen T, Kuang M, Liang X, Peng T, Wang K, Xu L, Li H, Song T, Sun H, Zhang W, China Liver Cancer Study Group Young Investigators (CLEAP). Comparison of Efficacy between Lenvatinib and Bevacizumab in Combination of Immune Checkpoint Inhibitor and Interventional Triple Therapy in Chinese Advanced Hepatocellular Carcinoma: The CLEAP 2302 Study. Liver Cancer 2025:1-19. [PMID: 40417458 PMCID: PMC12097796 DOI: 10.1159/000545545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 02/10/2025] [Indexed: 05/27/2025] Open
Abstract
Background The conversion therapy for advanced hepatocellular carcinoma (HCC) shows promise with a triple therapy approach that combines interventional therapy, immune checkpoint inhibitors, and molecular targeted therapy (primarily small-molecule TKIs and the large-molecule bevacizumab). This combination has achieved the highest objective response rates (ORR) along with acceptable safety profiles. The aim of this study was to compare the clinical efficacy of lenvatinib versus bevacizumab, when combined with immune checkpoint inhibitors and interventional triple therapy, as first-line treatments for Chinese patients with unresectable HCC (uHCC). Method This retrospective multicenter study involved 371 consecutive patients from 21 centers in China, observed between April 2017 and December 2023. The study focused on patients with uHCC at Chinese liver cancer stages IIb to IIIb (Barcelona Clinic Liver Cancer stage B or C) who received lenvatinib or bevacizumab combined with anti-PD-1/L1 and interventional therapy (including TACE and/or HAIC) as first-line treatment. Of the 371 patients, 258 received lenvatinib-based triple therapy, while 113 received bevacizumab-based triple therapy. The primary endpoints were overall survival (OS) and progression-free survival (PFS). To balance baseline clinical characteristics, propensity score matching (PSM) and inverse probability of treatment weighting (IPTW) were applied. Subgroup analysis was also performed based on different clinicopathological characteristics of the enrolled uHCC patients. Results The median OS in the lenvatinib group was significantly longer than in the bevacizumab group, both before (36.0 vs. 27.9 months; hazard ratio [HR]: 0.536; 95% confidence interval [CI]: 0.344-0.835; p = 0.0016) and after PSM (HR: 0.524; 95% CI: 0.305-0.900; p = 0.01), as well as after IPTW (HR: 0.549; 95% CI: 0.331-0.908; p = 0.01). Before adjustment, PFS in the lenvatinib group was also significantly longer than in the bevacizumab group (20.0 vs. 12.1 months; HR: 0.649; 95% CI: 0.457-0.922; p = 0.0078). However, after PSM (HR: 0.808; 95% CI: 0.535-1.222; p = 0.33) and IPTW, there was no significant difference in PFS between the two groups. Multivariate analysis showed that lenvatinib-based triple therapy was independently associated with improved OS compared to bevacizumab-based triple therapy. Subgroup analysis indicated that patients with age ≤65 years, no history of hepatitis B virus infection, Barcelona Clinic Liver Cancer stage C (BCLC-C), ALT levels ≤40 U/L, platelets ≥100 × 109/L, or log 10 AFP ≥1.40 benefited more from lenvatinib-based triple therapy. Conclusion Lenvatinib-based triple therapy tends to prolong OS compared to bevacizumab, although the PFS was similar between the two groups. Patients aged ≤65 years, without a history of hepatitis B virus infection, with BCLC-C stage, ALT ≤40 U/L, platelets ≥100 × 109/L, or log 10 AFP ≥1.40 are likely to benefit more from lenvatinib-based triple therapy.
Collapse
Affiliation(s)
- Zhaolong Pan
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Dongming Liu
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Junbo Cao
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Linlin Fu
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Xihao Zhang
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Xiaodong Zhu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yangxun Pan
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Jianwei Liu
- Department of Hepatic Surgery II, The Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Chuangye Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Renan Jin
- Department of General Surgery, Sir Run Run Shaw Hospital, College of Medicine, Institute of Minimally Invasive Surgery, Zhejiang University, Hangzhou, China
| | - Shunli Shen
- Department of Hepatic Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiaoyun Zhang
- Department of Liver Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Hongzhi Liu
- Department of Hepatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Xiaobo Yang
- Department of Hepatobiliary Surgery, Peking Union Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Kuan Hu
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoyi Shi
- Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dongxu Wang
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Yang Zhao
- Department of Interventional Therapy, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Jianhong Zhong
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Bangde Xiang
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Shanzhi Gu
- Department of Interventional Therapy, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Tao Li
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Shuijun Zhang
- Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ledu Zhou
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Haitao Zhao
- Department of Hepatobiliary Surgery, Peking Union Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yongyi Zeng
- Department of Hepatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Tianfu Wen
- Department of Liver Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Ming Kuang
- Department of Hepatic Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiao Liang
- Department of General Surgery, Sir Run Run Shaw Hospital, College of Medicine, Institute of Minimally Invasive Surgery, Zhejiang University, Hangzhou, China
| | - Tao Peng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Kui Wang
- Department of Hepatic Surgery II, The Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Li Xu
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Huikai Li
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Tianqiang Song
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Huichuan Sun
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wei Zhang
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - China Liver Cancer Study Group Young Investigators (CLEAP)
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Sun Yat-Sen University, Guangzhou, China
- Department of Hepatic Surgery II, The Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Department of General Surgery, Sir Run Run Shaw Hospital, College of Medicine, Institute of Minimally Invasive Surgery, Zhejiang University, Hangzhou, China
- Department of Hepatic Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Department of Liver Surgery, West China Hospital of Sichuan University, Chengdu, China
- Department of Hepatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
- Department of Hepatobiliary Surgery, Peking Union Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, China
- Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
- Department of Interventional Therapy, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
9
|
Jinushi K, Saito T, Kurose K, Suzuki S, Kojima T, Takahara T, Makino T, Ogawa T, Nishikawa H, Kakimi K, Iida S, Nakajima J, Doki Y, Oka M, Ueda R, Wada H. Phase I study on neoadjuvant combination immunotherapy with mogamulizumab and nivolumab for solid tumors. J Immunother Cancer 2025; 13:e010634. [PMID: 40180420 PMCID: PMC11966984 DOI: 10.1136/jitc-2024-010634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 03/14/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Effector regulatory T cells expressing C-C chemokine receptor 4 (CCR4) suppress antitumor immune responses. We conducted a phase I clinical trial to evaluate the safety and efficacy of preoperative combination therapy with mogamulizumab (an anti-CCR4 antibody) and nivolumab (an anti-programmed death-1 antibody) in patients with solid tumors. METHODS Patients with operable solid tumors were enrolled in a 3+3 design, with preoperative nivolumab (3.0 mg/kg) administered intravenously every 2 weeks three times and mogamulizumab at 0.1 mg/kg (cohort 1), 0.3 mg/kg (cohort 2), or 1.0 mg/kg (cohort 3) every week four times. The primary endpoints were safety and the effects of depleting Forkhead box P3+ (FoxP3+) T cells in the tumor. RESULTS 16 patients were enrolled between June 2016 and April 2020, including those with renal (n=7), lung (n=5), esophageal (n=3), and oral (n=1) cancers. Grade 3-4 treatment-related adverse events were observed in 6 of 16 patients, with lymphopenia (25%) and maculopapular rash (13%) being the most frequent. Grade 5 interstitial pneumonia was observed in one patient; however, the cause of death was disease progression. There were three partial responses (PRs) (one lung and two esophageal cancers) among clinical responses and one complete response (one lung cancer) and nine PRs (five kidney, two lung, and two esophageal cancers) among pathological responses. CCR4+FoxP3+ T cells were depleted in the tumors of all patients and increases in lymphocytes in tumor tissue according to the tumor immune microenvironment classification were observed in 50% of the patients, which correlated with a better prognosis. CONCLUSIONS The preoperative combination of mogamulizumab and nivolumab was safely managed, exerted antitumor effects, and may be an effective option in the preoperative setting. TRIAL REGISTRATION NUMBER The present study was registered with ClinicalTrials.gov as NCT02946671 (registration date 2016-10-05).
Collapse
Affiliation(s)
- Koichi Jinushi
- Department of Gastroenterological Surgery, Osaka University School of Medicine Graduate School of Medicine, Suita, Osaka Prefecture, Japan
- Department of Clinical Research in Tumor Immunology, Osaka University School of Medicine Graduate School of Medicine, Suita, Osaka Prefecture, Japan
| | - Takuro Saito
- Department of Gastroenterological Surgery, Osaka University School of Medicine Graduate School of Medicine, Suita, Osaka Prefecture, Japan
- Department of Clinical Research in Tumor Immunology, Osaka University School of Medicine Graduate School of Medicine, Suita, Osaka Prefecture, Japan
| | - Koji Kurose
- Department of Respiratory Medicine, Kawasaki Medical School, Kurashiki, Japan
| | - Susumu Suzuki
- Research Creation Support Center, Aichi Medical University, Nagakute, Aichi, Japan
- Tumor Immunology, Aichi Medical University, Nagakute, Aichi, Japan
| | - Takashi Kojima
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center-Hospital East, Kashiwa, Chiba, Japan
| | - Taishi Takahara
- Department of Surgical Pathology, Aichi Medical University Hospital, Nagakute, Aichi Prefecture, Japan
| | - Tomoki Makino
- Department of Gastroenterological Surgery, Osaka University School of Medicine Graduate School of Medicine, Suita, Osaka Prefecture, Japan
| | - Tetsuya Ogawa
- Department of Otorhinolaryngology-Head and Neck Surgery, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Hiroyoshi Nishikawa
- Division of Cancer Immunology, National Cancer Center Japan, Chuo, Tokyo, Japan
- Department of Immunology, Nagoya University Graduate School of Medicine Faculty of Medicine, Nagoya, Aichi Prefecture, Japan
- Division of Cancer Immune Multicellular System Regulation, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto, Kyoto, Japan
| | - Kazuhiro Kakimi
- Department of Immunology, Kindai University Faculty of Medicine Graduate School of Medical Sciences, sayama, Osaka, Japan
- Department of Immunotherapeutics, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Shinsuke Iida
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences and Medical School, Nagoya, Aichi Prefecture, Japan
| | - Jun Nakajima
- Department of Thoracic Surgery, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Osaka University School of Medicine Graduate School of Medicine, Suita, Osaka Prefecture, Japan
| | - Mikio Oka
- Department of Immuno-Oncology, Kawasaki Medical School, Kurashiki, Japan
| | - Ryuzo Ueda
- Department of Immunology, Nagoya University Graduate School of Medicine Faculty of Medicine, Nagoya, Aichi Prefecture, Japan
| | - Hisashi Wada
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
10
|
Park S, Park K, Kim C, Rhie SJ. Optimization of immunotherapy-based combinations for metastatic renal cell carcinoma: A network meta-analysis. Crit Rev Oncol Hematol 2025; 208:104630. [PMID: 39864536 DOI: 10.1016/j.critrevonc.2025.104630] [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: 12/10/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 01/28/2025] Open
Abstract
BACKGROUND Despite numerous meta-analyses comparing the efficacy and safety of immunotherapy-based combination therapies, the optimal therapeutic combinations remain unclear. This study aims to evaluate the optimal application of all immunotherapy-based combination therapy for advanced/metastatic renal cell carcinoma, focusing on efficacy and safety. METHODS We systemically searched the Web of Science, Cochrane Library, and PubMed for studies regarding the first-line immunotherapy-based combination therapy in patients with advanced or metastatic renal cell carcinoma until April 15, 2024. We used network meta-analysis using a random effect model to facilitate direct and indirect treatment comparisons across outcomes. RESULTS Seven clinical studies, including 5542 patients with metastatic renal cell carcinoma, were included in the network meta-analysis analysis. Regarding progression-free survival and overall survival, combined Toripalimab + Axitinib significantly outperformed other immunotherapy-based combination therapies. This regimen significantly improved progression-free survival in the intermediate/poor risk group when stratified by prognosis prediction risks compared to sunitinib alone. For the objective response rate, Avelumab + Axitinib was the most preferred strategy in the favorable-risk group, while Nivolumab + Cabozantinib was favored in the intermediate/poor-risk group compared to other immunotherapy-based combinations. The combinations of Nivolumab + Ipilimumab and Atezolizumab + Bevacizumab had favorable safety profiles. CONCLUSIONS Immunotherapy-based combination therapies significantly improved progression-free survival, overall survival and objective response rate in patients with metastatic renal cell carcinoma compared to sunitinib monotherapy. However, careful monitoring and personalized treatment strategies are required to balance efficacy and safety in patients with underlying conditions. Future research should focus on optimizing treatment protocols and elucidating the mechanisms of adverse events.
Collapse
Affiliation(s)
- Sohyeon Park
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea.
| | - Kalynn Park
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea.
| | - Chaeyoon Kim
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea.
| | - Sandy Jeong Rhie
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea.
| |
Collapse
|
11
|
Yan H, Wu X, Li H, Yu Z, Jin X. Pan-Cancer Analysis Identifies BCLAF1 as a Potential Biomarker for Renal Cell Carcinoma. Biochem Genet 2025; 63:1479-1508. [PMID: 38573525 DOI: 10.1007/s10528-024-10773-x] [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: 07/04/2023] [Accepted: 03/04/2024] [Indexed: 04/05/2024]
Abstract
B-cell lymphoma-2-associated transcription factor 1 (BCLAF1) is a versatile protein involved in the regulation of gene transcription and post-transcriptional processing. Although BCLAF1 exerts a broad tumor suppressor effect or tumor promoter effect in many cancer types, the specific roles concerning its expression levels, and its impact on tumorigenesis in Renal cell carcinoma (RCC) remain unclear. Here, we utilized the Cancer Genome Atlas (TCGA) and Genotype Tissue Expression (GTEx) datasets alongside R software and online tools to unravel the specific roles of BCLAF1 in 33 cancer types, including its expression levels, tumor immune and molecular subtypes, and its correlation with prognosis, diagnosis, DNA methylation, and immune microenvironment. Additionally, we carried out cell biology experiments to independently investigate the expression of BCLAF1 in RCC and its effects on tumor progression. BCLAF1 was differentially expressed in tumor tissues compared to normal tissues across various cancer types and was also differentially expressed in different immune and molecular subtypes. In RCC, patients with high BCLAF1 expression had a better prognosis and BCLAF1 was tightly correlated with the stage, gender, and histological grade of patients. Furthermore, BCLAF1 had higher DNA methylation levels and higher immune infiltration levels in tumor tissues. Additionally, cell functional experiments confirmed the low expression of BCLAF1 in RCC and that BCLAF1 significantly inhibited the proliferation, migration, and invasion, while inducing apoptosis and cell cycle arrest in RCC cells in vitro. Our study under-scored the potential of BCLAF1 as an important actor in tumorigenesis, especially concerning RCC where it may serve as an effective prognostic marker.
Collapse
Affiliation(s)
- Huan Yan
- Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xiang Wu
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Hong Li
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Zongdong Yu
- Department of Neurosurgery, Shangrao People's Hospital, Shangrao, 334099, China.
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China.
| | - Xiaofeng Jin
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
12
|
Ueda K, Ito N, Sakai Y, Ohnishi S, Hirano T, Kurose H, Chikui K, Uemura K, Nishihara K, Nakiri M, Suekane S, Igawa T. Therapeutic efficacy of immune-oncology combination therapy in advanced renal cell carcinoma without prior nephrectomy. Int J Clin Oncol 2025; 30:770-779. [PMID: 39899167 DOI: 10.1007/s10147-025-02710-8] [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: 08/30/2024] [Accepted: 01/20/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND Immuno-oncology (IO) combination therapies, including IO + IO or IO + vascular endothelial growth factor targeted therapies (VEGF-TT), have become the standard first-line treatment for advanced renal cell carcinoma (RCC). However, the optimal regimen for patients without prior nephrectomy remains unclear. METHODS Data from 99 patients with advanced RCC without nephrectomy, treated with VEGF-TT, IO + IO, or IO + VEGF-TT between May 2008 and May 2024, were retrospectively reviewed and analyzed. Patients were divided into VEGE-TT, IO + IO, and IO + VEGF-TT groups based on their first-line treatment, and survival and tumor response were compared. RESULTS All patients included in this study were categorized as either intermediate or poor risk according to the International Metastatic RCC Database Consortium risk classification. Among the 99 included patients, 41 initiated first-line therapy with VEGF-TT, 36 with IO + IO, and 22 with IO + VEGF-TT. The objective response rates were 17.5% for VEGF-TT, 38.9% for IO + IO, and 61.9% for IO + VEGF-TT. Notably, the IO + VEGF-TT group showed the greatest shrinkage of target kidney lesions (p = 0.0042). In multivariate analyses, bone metastasis (hazard ratio (HR) = 1.812, 95% confidence interval (CI) 1.017-3.228, p = 0.0436) and the first-line regimen (VEGF-TT vs IO + VEGF-TT: HR = 0.129, 95% CI 0.045-0.369, p = 0.0001) were independent prognostic factors for progression-free survival. The first-line regimen (VEGF-TT vs IO + VEGF-TT: HR = 0.303, 95% CI 0.104-0.879, p = 0.0279) independently affected overall survival. CONCLUSION IO combination therapy, especially IO + VEGF-TT, has demonstrated a higher anti-tumor response in patients with advanced RCC without nephrectomy and may also be highly effective against primary renal tumors. Therefore, further studies are needed to improve patient survival and validate efficacy of IO combination therapy.
Collapse
Affiliation(s)
- Kosuke Ueda
- Department of Urology, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan.
| | - Naoki Ito
- Department of Urology, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan
| | - Yuya Sakai
- Department of Urology, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan
| | - Satoshi Ohnishi
- Department of Urology, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan
| | - Taishi Hirano
- Department of Urology, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan
| | - Hirofumi Kurose
- Department of Urology, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan
| | - Katsuaki Chikui
- Department of Urology, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan
| | - Keiichiro Uemura
- Department of Urology, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan
| | - Kiyoaki Nishihara
- Department of Urology, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan
| | - Makoto Nakiri
- Department of Urology, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan
| | - Shigetaka Suekane
- Department of Urology, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan
| | - Tsukasa Igawa
- Department of Urology, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan
| |
Collapse
|
13
|
Zugman M, Wong M, Jaime-Casas S, Pal SK. The gut microbiome and dietary metabolites in the treatment of renal cell carcinoma. Urol Oncol 2025; 43:244-253. [PMID: 39095306 DOI: 10.1016/j.urolonc.2024.07.003] [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: 04/10/2024] [Revised: 06/13/2024] [Accepted: 07/02/2024] [Indexed: 08/04/2024]
Abstract
The gut microbiome is interlinked with renal cell carcinoma (RCC) and its response to systemic treatment. Mounting data suggests that certain elements of the gut microbiome may correlate with improved outcomes. New generation sequencing techniques and advanced bioinformatic data curation are accelerating the investigation of specific markers and metabolites that could predict treatment response. A variety of new therapeutic strategies, such as fecal microbiota transplantation, probiotic supplements, and dietary interventions, are currently being developed to modify the gut microbiome and improve anticancer therapies in patients with RCC. This review discusses the preliminary evidence indicating the role of the microbiome in cancer treatment, the techniques and tools necessary for its proper study and some of the current forms with which the microbiome can be modulated to improve patient outcomes.
Collapse
Affiliation(s)
- Miguel Zugman
- Department of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA; Centro de Oncologia e Hematologia Família Dayan-Daycoval Einstein, Hospital Israelita Albert, São Paulo, São Paulo, Brazil
| | - Megan Wong
- Department of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Salvador Jaime-Casas
- Department of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Sumanta K Pal
- Department of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA.
| |
Collapse
|
14
|
Zhu Y, Yu S, Yang D, Yu T, Liu Y, Du W. Integrated Multi-Omics Analysis Unveils Distinct Molecular Subtypes and a Robust Immune-Metabolic Prognostic Model in Clear Cell Renal Cell Carcinoma. Int J Mol Sci 2025; 26:3125. [PMID: 40243888 PMCID: PMC11988429 DOI: 10.3390/ijms26073125] [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: 03/03/2025] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is characterized by significant clinical and molecular heterogeneity, with immune and metabolic processes playing crucial roles in tumor progression and influencing patient outcomes. This study aims to elucidate the molecular subtypes of ccRCC by employing non-negative matrix factorization (NMF) clustering on differentially expressed genes (DEGs), thereby identifying distinct transcriptional profiles, immune cell infiltration patterns, and subsequent survival outcomes. Utilizing NMF clustering, we identified two molecular subtypes of ccRCC. We developed a prognostic model using LASSO-Cox regression, validated with multiple datasets and quantitative reverse transcription polymerase chain reaction (qRT-PCR), incorporating ten immunity- and metabolism-related genes (IMRGs) for overall survival (OS) prediction. Immune cell infiltration and tumor mutational burden (TMB) analyses were performed to explore differences between high- and low-risk groups, while Gene Set Enrichment Analysis (GSEA) provided insights into relevant biological pathways. The findings revealed that subtype C1, characterized by a "cold" tumor microenvironment, correlates with better prognostic outcomes compared to subtype C2, which exhibits an immunologically active environment and worse survival prospects. High-risk patients demonstrated poorer OS associated with alterations in immune and metabolic pathways. Immune checkpoint analysis indicated the upregulation of CTLA4, LAG3, and LGALS9 in high-risk patients, suggesting potential therapeutic targets. A nomogram integrating IMRG risk scores with clinical factors displayed high predictive accuracy for 1-, 3-, and 5-year OS. These findings provide novel insights into the molecular heterogeneity of ccRCC and emphasize the interconnected roles of immune dysregulation and metabolic alterations in tumor progression. By identifying key prognostic biomarkers and potential therapeutic targets, this study paves the way for innovative strategies aimed at harnessing immune and metabolic pathways for better clinical outcomes in ccRCC patients.
Collapse
Affiliation(s)
- Yilin Zhu
- Department of Bioinformatics, School of Life Sciences, Xuzhou Medical University, Xuzhou 221004, China
| | - Shihui Yu
- Department of Biophysics, School of Life Sciences, Xuzhou Medical University, Xuzhou 221004, China
| | - Dan Yang
- Department of Biophysics, School of Life Sciences, Xuzhou Medical University, Xuzhou 221004, China
| | - Tian Yu
- Department of Bioinformatics, School of Life Sciences, Xuzhou Medical University, Xuzhou 221004, China
| | - Yi Liu
- Department of Bioinformatics, School of Life Sciences, Xuzhou Medical University, Xuzhou 221004, China
- Department of Biophysics, School of Life Sciences, Xuzhou Medical University, Xuzhou 221004, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Wenlong Du
- Department of Bioinformatics, School of Life Sciences, Xuzhou Medical University, Xuzhou 221004, China
- Department of Biophysics, School of Life Sciences, Xuzhou Medical University, Xuzhou 221004, China
| |
Collapse
|
15
|
Kavran AJ, Bai Y, Rabe B, Kreshock A, Fisher A, Cheng Y, Lewin A, Dai C, Meyer MJ, Mavrakis KJ, Lyubetskaya A, Drokhlyansky E. Spatial genomics reveals cholesterol metabolism as a key factor in colorectal cancer immunotherapy resistance. Front Oncol 2025; 15:1549237. [PMID: 40171265 PMCID: PMC11959564 DOI: 10.3389/fonc.2025.1549237] [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: 12/20/2024] [Accepted: 02/24/2025] [Indexed: 04/03/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) have transformed the treatment landscape across multiple cancer types achieving durable responses for a significant number of patients. Despite their success, many patients still fail to respond to ICIs or develop resistance soon after treatment. We sought to identify early treatment features associated with ICI outcome. We leveraged the MC38 syngeneic tumor model because it has variable response to ICI therapy driven by tumor intrinsic heterogeneity. ICI response was assessed based on the level of immune cell infiltration into the tumor - a well-established clinical hallmark of ICI response. We generated a spatial atlas of 48,636 transcriptome-wide spots across 16 tumors using spatial transcriptomics; given the tumors were difficult to profile, we developed an enhanced transcriptome capture protocol yielding high quality spatial data. In total, we identified 8 tumor cell subsets (e.g., proliferative, inflamed, and vascularized) and 4 stroma subsets (e.g., immune and fibroblast). Each tumor had orthogonal histology and bulk-RNA sequencing data, which served to validate and benchmark observations from the spatial data. Our spatial atlas revealed that increased tumor cell cholesterol regulation, synthesis, and transport were associated with a lack of ICI response. Conversely, inflammation and T cell infiltration were associated with response. We further leveraged spatially aware gene expression analysis, to demonstrate that high cholesterol synthesis by tumor cells was associated with cytotoxic CD8 T cell exclusion. Finally, we demonstrate that bulk RNA-sequencing was able to detect immune correlates of response but lacked the sensitivity to detect cholesterol synthesis as a feature of resistance.
Collapse
Affiliation(s)
- Andrew J. Kavran
- Mechanisms of Cancer Resistance Thematic Research Center (TRC), Bristol Myers Squibb, Cambridge, MA, United States
| | - Yulong Bai
- Informatics and Predictive Sciences, Bristol Myers Squibb, Cambridge, MA, United States
| | - Brian Rabe
- Mechanisms of Cancer Resistance Thematic Research Center (TRC), Bristol Myers Squibb, Cambridge, MA, United States
| | - Anna Kreshock
- Mechanisms of Cancer Resistance Thematic Research Center (TRC), Bristol Myers Squibb, Cambridge, MA, United States
| | - Andrew Fisher
- Informatics and Predictive Sciences, Bristol Myers Squibb, Cambridge, MA, United States
| | - Yelena Cheng
- Mechanisms of Cancer Resistance Thematic Research Center (TRC), Bristol Myers Squibb, Cambridge, MA, United States
| | - Anne Lewin
- Translational Medicine, Bristol Myers Squibb, Cambridge, MA, United States
| | - Chao Dai
- Mechanisms of Cancer Resistance Thematic Research Center (TRC), Bristol Myers Squibb, Cambridge, MA, United States
| | - Matthew J. Meyer
- Mechanisms of Cancer Resistance Thematic Research Center (TRC), Bristol Myers Squibb, Cambridge, MA, United States
| | - Konstantinos J. Mavrakis
- Mechanisms of Cancer Resistance Thematic Research Center (TRC), Bristol Myers Squibb, Cambridge, MA, United States
| | - Anna Lyubetskaya
- Mechanisms of Cancer Resistance Thematic Research Center (TRC), Bristol Myers Squibb, Cambridge, MA, United States
| | - Eugene Drokhlyansky
- Mechanisms of Cancer Resistance Thematic Research Center (TRC), Bristol Myers Squibb, Cambridge, MA, United States
| |
Collapse
|
16
|
Choi SH, Chen YW, Panian J, Yuen K, McKay RR. Emerging innovative treatment strategies for advanced clear cell renal cell carcinoma. Oncologist 2025; 30:oyae276. [PMID: 39401004 PMCID: PMC11954509 DOI: 10.1093/oncolo/oyae276] [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: 06/16/2024] [Accepted: 09/10/2024] [Indexed: 10/15/2024] Open
Abstract
Dramatic advances in biological discoveries, since the 1990s, have continued to reshape the treatment paradigm of metastatic renal cell carcinoma (RCC). Von Hippel Lindau (VHL) gene alterations are associated with pro-angiogenic activity and are central to the pathogenesis of clear cell RCC (ccRCC), the most predominant histologic subtype of RCC. Antiangiogenic strategies revolving around this VHL/HIF/VEGF axis have been shown to improve survival in metastatic ccRCC. The discovery of immune checkpoints and agents that target their inhibition introduced a new treatment paradigm for patients with RCC. While initially approved as monotherapy, studies investigating immune checkpoint inhibitor combinations have led to their approval as the new standard of care, providing durable responses and unprecedented improvements in clinical outcome. Despite these advances, the projected 14 390 deaths in 2024 from RCC underscore the need to continue efforts in expanding and optimizing treatment options for patients with metastatic RCC. This article reviews key findings that have transformed the way we understand and treat metastatic RCC, in addition to highlighting novel treatment strategies that are currently under development.
Collapse
Affiliation(s)
- Sharon H Choi
- Division of Hematology Oncology, University of California San Diego, San Diego, CA, United States
| | - Yu-Wei Chen
- Division of Hematology Oncology, University of California San Diego, San Diego, CA, United States
| | - Justine Panian
- Division of Hematology Oncology, University of California San Diego, San Diego, CA, United States
| | - Kit Yuen
- Department of Urology, University of California San Diego, San Diego, CA, United States
| | - Rana R McKay
- Division of Hematology Oncology, University of California San Diego, San Diego, CA, United States
- Department of Urology, University of California San Diego, San Diego, CA, United States
| |
Collapse
|
17
|
Nassar AH, Abou Alaiwi S, El Zarif T, Denu R, Macaron W, Abdel-Wahab N, Freeman D, Vasbinder A, Hayeck S, Anderson E, Goodman RS, Johnson DB, Grynberg S, Shapira R, Kwan JM, Woodford R, Long GV, Haykal T, Dent S, Kojima Y, Yonemor K, Tandon A, Trevino A, Akhter N, Yang EH, Hui G, Drakaki A, El-Am E, Kozaily E, Al-Hader A, Bou Farhat E, Babu P, Mittra A, Li M, Jones N, Baena J, Juarez Herrera M, Foderaro S, Nana FA, Kim C, Sackstein P, Parikh K, Desai AP, Smith C, Cortellini A, Pinato DJ, Korolewicz J, Lopetegui-Lia N, Funchain P, Choudhary A, Asnani A, Navani V, Meyers D, Stukalin I, Ocejo Gallegos JA, Trent J, Nusrat S, Malvar C, McKay RR, Neilan TG, Choueiri TK, Naqash AR. Safety and efficacy of immune checkpoint therapy for the treatment of patients with cardiac metastasis: a multicenter international retrospective study. J Immunother Cancer 2025; 13:e009364. [PMID: 40032601 PMCID: PMC11877189 DOI: 10.1136/jitc-2024-009364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 01/03/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Data on the safety profiles and clinical outcomes of patients with solid tumors and cardiac metastasis treated with immune checkpoint inhibitors (ICIs) are limited. METHODS This is an international multicenter retrospective study of patients with cancer and cardiac metastasis at baseline. Patients who had received ≥1 dose of ICI were included. Treatment-related adverse events (trAEs) were graded per Common Terminology Criteria for Adverse Event V.5.0. Objective response rates (ORR) were evaluated by Response Evaluation Criteria in Solid Tumors V.1.1 when available. Overall survival (OS) and progression-free survival (PFS) were estimated by the Kaplan-Meier method. RESULTS Among 110 pts, median age at ICI initiation was 65 (IQR: 59-75). Median follow-up time since ICI initiation was 36 (95% CI: 26 to 51) months. Melanoma (38%, n=42) and non-small cell lung cancer (24%, n=26) were the most common. 68 (62%) patients received ICIs as first-line, and 29 (26%) patients were treated with combination anti-programmed death-1 and anti-cytotoxic T-lymphocyte antigen 4. The most common location of cardiac metastasis was in the atria (37%, n=41) and ventricles (35%, n=39). 15 patients (13.6%) had bilateral cardiac/pericardial metastasis, 44 (40%) had left-sided, and 43 (39.8%) had right-sided. At ICI initiation, 21% (n=23) had a cardiac thrombus. Cardiology referrals and cardiac MRIs at the time of cancer diagnosis were completed on 58 (53%) and 52 (47%) patients, respectively. Cardiac events occurred in 40 (36%) patients, including arrhythmias (n=14, 13%), arterial/venous emboli (n=4, 3.6%), and cardiac tamponade (n=3, 2.7%). 53 (47%) patients developed trAEs; most common were colitis/diarrhea (n=16, 15%), dermatitis (n=13, 12%), and hepatitis (n=9, 8.2%). ICI-related major cardiac trAEs occurred in 2 (1.8%) patients. 22 patients (20%) developed grade ≥3 trAE. Patients with multiple cardiac metastases had significantly lower responses to ICI-based regimens compared with patients with single cardiac metastasis (11% vs 63%, p=0.02). For melanoma, ORR, median PFS, and median OS were 38%, 9.0 months, and 28.9 months, respectively. 83% of patients with melanoma had concordant responses in overall disease burden and cardiac disease. 91 patients discontinued ICIs, and the main reason was progression or death in 55 (49%) patients. CONCLUSIONS Among patients with pre-existing cardiac metastasis, ICIs demonstrated meaningful clinical efficacy with no increase in safety signals. Most patients had concordant responses in the overall disease burden and cardiac mass. Multidisciplinary teams are crucial for the appropriate management of patients with cardiac metastasis.
Collapse
Affiliation(s)
- Amin H Nassar
- Yale University School of Medicine, New Haven, Connecticut, USA
| | | | - Talal El Zarif
- Yale University School of Medicine, New Haven, Connecticut, USA
| | - Ryan Denu
- Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Walid Macaron
- Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Noha Abdel-Wahab
- Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Dory Freeman
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Alexi Vasbinder
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Salim Hayeck
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Elizabeth Anderson
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Rachel S Goodman
- Department of Medicine, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, Tennessee, USA
| | - Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, Tennessee, USA
| | - Shirly Grynberg
- Ella Lemelbaum Institute of Immuno-Oncology, Sheba Medical Center, Ramat Gan, Israel
| | - Ronnie Shapira
- Ella Lemelbaum Institute of Immuno-Oncology, Sheba Medical Center, Ramat Gan, Israel
| | - Jennifer M Kwan
- Yale University School of Medicine, New Haven, Connecticut, USA
| | - Rachel Woodford
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
| | - Tarek Haykal
- Duke Cancer Institute, School of Medicine, Duke University, Durham, North Carolina, USA
- Department of Internal Medicine, Division of medical Oncology, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Susan Dent
- Duke Cancer Institute, School of Medicine, Duke University, Durham, North Carolina, USA
| | - Yuki Kojima
- National Cancer Center Hospital, Tokyo, Japan
| | - Kan Yonemor
- National Cancer Center Hospital, Tokyo, Japan
| | - Ankita Tandon
- Department of Medical Oncology, Loyola University Medical Center, Maywood, Illinois
| | - Alexandra Trevino
- Division of Cardiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Nausheen Akhter
- Division of Cardiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Eric H Yang
- UCLA Cardio-oncology Program, Division of Cardiology, Department of Medicine, University of California at Los Angeles, Los Angeles, California, USA
| | - Gavin Hui
- UCLA Cardio-oncology Program, Division of Cardiology, Department of Medicine, University of California at Los Angeles, Los Angeles, California, USA
- Hematology/Oncology Department, David Geffen School of Medicine, Los Angeles, California, USA
| | - Alexandra Drakaki
- UCLA Cardio-oncology Program, Division of Cardiology, Department of Medicine, University of California at Los Angeles, Los Angeles, California, USA
- Hematology/Oncology Department, David Geffen School of Medicine, Los Angeles, California, USA
| | - Edward El-Am
- Department of Medicine, Indiana University School of Medicine Eskenazi Hospital, Indianapolis, Indiana, USA
| | - Elie Kozaily
- Department of Medicine, Indiana University School of Medicine Eskenazi Hospital, Indianapolis, Indiana, USA
| | - Ahmad Al-Hader
- Department of Medicine, Indiana University School of Medicine Eskenazi Hospital, Indianapolis, Indiana, USA
| | | | - Priyanka Babu
- Yale University School of Medicine, New Haven, Connecticut, USA
| | - Arjun Mittra
- Division of Medical Oncology, The Ohio State University, Columbus, Ohio, USA
| | - Mingjia Li
- Division of Medical Oncology, The Ohio State University, Columbus, Ohio, USA
| | - Nicholas Jones
- Division of Medical Oncology, The Ohio State University, Columbus, Ohio, USA
| | - Javier Baena
- Department of Medical Oncology, Hospital 12 de Octubre, Madrid, Spain
| | | | - Simone Foderaro
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Rome, Italy
| | | | - Chul Kim
- Lombardi Comprehensive Cancer Center, MedStar Georgetown University Hospital, Washington, DC, Washington, DC, USA
| | - Paul Sackstein
- Lombardi Comprehensive Cancer Center, MedStar Georgetown University Hospital, Washington, DC, Washington, DC, USA
| | - Kaushal Parikh
- Division of Medical Oncology, Mayo Clinic, Rochester, New York, USA
| | - Aakash P Desai
- Division of Medical Oncology, Mayo Clinic, Rochester, New York, USA
| | - Caleb Smith
- Division of Medical Oncology, Mayo Clinic, Rochester, New York, USA
| | - Alessio Cortellini
- Operative Research Unit of Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
- Department of Medicine and Surgery, Universitá Campus Bio-Medico di Roma, Roma, Italy
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - David J Pinato
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
- Division of Oncology, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - James Korolewicz
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | | | - Pauline Funchain
- Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Arrush Choudhary
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Aarti Asnani
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Vishal Navani
- Tom Baker Cancer Centre, University of Calgary, Calgary, Alberta, Canada
| | - Daniel Meyers
- Tom Baker Cancer Centre, University of Calgary, Calgary, Alberta, Canada
| | - Igor Stukalin
- Tom Baker Cancer Centre, University of Calgary, Calgary, Alberta, Canada
| | | | - Jonathan Trent
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Sanober Nusrat
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, Oklahoma, USA
| | - Carmel Malvar
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA
| | - Rana R McKay
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA
| | - Tomas G Neilan
- Cardio-Oncology Program, Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Toni K Choueiri
- Division of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Abdul Rafeh Naqash
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, Oklahoma, USA
| |
Collapse
|
18
|
Chen J, Ma N, Chen B, Huang Y, Li J, Li J, Chen Z, Wang P, Ran B, Yang J, Bai J, Ning S, Ai J, Wei Q, Liu L, Cao D. Synergistic effects of immunotherapy and adjunctive therapies in prostate cancer management. Crit Rev Oncol Hematol 2025; 207:104604. [PMID: 39732304 DOI: 10.1016/j.critrevonc.2024.104604] [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: 11/15/2024] [Revised: 12/14/2024] [Accepted: 12/20/2024] [Indexed: 12/30/2024] Open
Abstract
In recent years, cancer immunotherapy has received widespread attention due to significant tumor clearance in some malignancies. Various immunotherapy approaches, including vaccines, immune checkpoint inhibitors, oncolytic virotherapy, bispecific T cell engagers, and adoptive T cell transfer, have completed or are undergoing clinical trials for prostate cancer. Despite immune checkpoint blockade's extraordinary effectiveness in treating a variety of cancers, targeted prostate cancer treatment using the immune system is still in its infancy. Multiple factors including the heterogeneity of prostate cancer, the cold tumor microenvironment, and a low level of neoantigens, contribute to the poor immunotherapy response. Significant effort is being devoted to improving immune-based prostate cancer therapy. Recently, several key discoveries demonstrate that prostate cancer immunotherapy agents may be used to promise better prognosis for patients as part of combination strategies with other agents targeting tumor-associated immune mechanism of resistance. Here, this review comprehensively examines the recent advancements in immunotherapy for prostate cancer, exploring its potential synergistic effects when combined with other treatment modalities to enhance clinical efficacy.
Collapse
Affiliation(s)
- Jie Chen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Na Ma
- Department of Pediatrics, West China Second University Hospital, Sichuan University, No. 20, 3rd section, South Renmin Road, Chengdu 610041, China
| | - Bo Chen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yin Huang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jinze Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jin Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zeyu Chen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Puze Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Biao Ran
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiahao Yang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jingxing Bai
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shu Ning
- Department of Urologic Surgery, University of California Davis, Davis, CA, USA
| | - Jianzhong Ai
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiang Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Liangren Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Dehong Cao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
19
|
Lauridsen KM, Møller HJ, Kristensen MW, Fristrup N, Donskov F, Hokland M, Andersen MN. Soluble CD206 in metastatic renal cell carcinoma: Relation to clinical-biochemical parameters and patient outcome. Int J Cancer 2025; 156:875-885. [PMID: 39319597 DOI: 10.1002/ijc.35194] [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: 04/13/2024] [Revised: 08/11/2024] [Accepted: 09/02/2024] [Indexed: 09/26/2024]
Abstract
The mannose receptor (MR/CD206) is a marker of M2-like tumor-associated macrophages. Membrane CD206 can be shed, releasing the receptor as a soluble protein (sCD206), which can be measured in serum. Here, we investigated the biomarker potential of sCD206 in patients with metastatic renal cell carcinoma (mRCC). Serum sCD206 was measured by an enzyme-linked immunosorbent assay in 88 mRCC patients and 20 healthy controls (HCs). At diagnosis, serum sCD206 was elevated in patients with intermediate-risk mRCC according to the Memorial Sloan Kettering Cancer Center (MSKCC) risk score, compared to both HCs and patients with favorable MSKCC risk score. Furthermore, sCD206 levels correlated with both sCD163 and C-reactive protein. Soluble CD206 levels decreased after treatment initiation (p < .0001 at 5 weeks) but with a tendency toward elevated levels at time of progression, compared to baseline (p = .06). In univariate survival analysis, high levels of serum sCD206 at baseline was a significant risk factor associated with reduced overall survival (hazard ratio [HR] = 1.37, 95% confidence interval: 1.12-1.67, p = .002). Stratified by clinical risk scores, increased sCD206 was still a statistically significant risk factor of overall mortality (p < .01) in the intermediate-risk group by both the MSKCC (HR = 1.48) and the newer International Metastatic RCC Database Consortium (IMDC) score (HR = 1.53). Furthermore, addition of sCD206 as a dichotomized variable to the IMDC risk score enabled separation of the intermediate-risk group into two groups with survival comparable to those with favorable and poor risk, respectively. Overall, sCD206 is a potential add-on biomarker for mRCC patients in the intermediate-risk group of the current clinical risk scores.
Collapse
Affiliation(s)
- Kasper Munch Lauridsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Holger Jon Møller
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Mie Wolff Kristensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Niels Fristrup
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Frede Donskov
- Department of Oncology, University Hospital of Southern Denmark, Esbjerg, Denmark
| | | | - Morten Nørgaard Andersen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
20
|
Pourmir I, Benhamouda N, Tran T, Roux H, Pineau J, Gey A, Munoz A, Mabrouk N, Epaillard N, Verkarre V, Vano YA, Tartour E, Oudard S. Soluble TIM-3, likely produced by myeloid cells, predicts resistance to immune checkpoint inhibitors in metastatic clear cell renal cell carcinoma. J Exp Clin Cancer Res 2025; 44:54. [PMID: 39953623 PMCID: PMC11827183 DOI: 10.1186/s13046-025-03293-y] [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] [Accepted: 01/15/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Immunotherapies targeting PD-1 and CTLA-4 are key components of the treatment of metastatic clear cell renal cell carcinoma (mccRCC). However, they have distinct safety profiles and resistance to treatment can occur. We assess soluble TIM-3 (sTIM-3) in the plasma of mccRCC patients as a potential theranostic biomarker, as well as its source and biological significance. METHODS We analyzed the association between sTIM-3 and overall survival (OS), tumor response, and common clinical and biological factors in two mccRCC cohorts treated with anti-PD-1 (nivolumab, n = 27), anti-PD-1 or anti-PD-1 + anti-CTLA-4 (nivolumab + ipilimumab - N + I, n = 124). The origin and role of sTIM-3 are studied on tumor and blood samples, using multiplex immunohistochemistry and flow cytometry, as well as analyses of publicly available single-cell transcriptomic (scRNAseq) and mass cytometry data. RESULTS sTIM-3 is significantly elevated in the plasma of treatment-naive mccRCC. It shows distinct associations with survival on anti-PD-1 vs anti-PD-1 + anti-CTLA-4: under nivolumab monotherapy, sTIM-3-high patients have a significantly reduced survival compared to sTIM-3-low patients, while they have similar survival probabilities under N + I. sTIM-3 is independent from other clinical and biological factors. Myeloid immune cells appear as the prominent source of sTIM-3, which may indicate their dysfunctional role in the antitumor immune response. CONCLUSIONS sTIM-3 appears to be a promising biomarker for optimizing treatment strategies in ccRCC as well as a potential therapeutic target, linked with to the immune myeloid compartment. Future investigations are warranted in patients treated with anti-PD-1 + antiangiogenic therapies.
Collapse
Affiliation(s)
- Ivan Pourmir
- Université Paris Cité, INSERM U970, PARCC, Paris, France.
- Medical Oncology Department, Georges Pompidou European Hospital, CARPEM Cancer Institute, AP-HP Centre, Université Paris Cité, Paris, France.
- Thoracic Oncology Department, Georges Pompidou European Hospital, CARPEM Cancer Institute, AP-HP Centre, Université Paris Cité, Paris, France.
- Paris - Cardiovascular Research Center, European Georges Pompidou Hospital, 56 Rue Leblanc, Paris, 75015, France.
| | - Nadine Benhamouda
- Université Paris Cité, INSERM U970, PARCC, Paris, France
- Immunology Department, Georges Pompidou European Hospital, AP-HP Centre, Université Paris Cité, Paris, France
| | - Thi Tran
- Université Paris Cité, INSERM U970, PARCC, Paris, France
| | - Hugo Roux
- Institut Curie, PSL University, Inserm U932, Immunity and Cancer, Paris, France
| | - Joséphine Pineau
- Université Paris Cité, INSERM U970, PARCC, Paris, France
- Immunology Department, Georges Pompidou European Hospital, AP-HP Centre, Université Paris Cité, Paris, France
| | - Alain Gey
- Université Paris Cité, INSERM U970, PARCC, Paris, France
- Immunology Department, Georges Pompidou European Hospital, AP-HP Centre, Université Paris Cité, Paris, France
| | - Andyara Munoz
- Immunology Department, Georges Pompidou European Hospital, AP-HP Centre, Université Paris Cité, Paris, France
| | - Nesrine Mabrouk
- Immunology Department, Georges Pompidou European Hospital, AP-HP Centre, Université Paris Cité, Paris, France
| | - Nicolas Epaillard
- Medical Oncology Department, Clinique Kuindo Magnin, Nouméa, Nouvelle Calédonie, France
| | - Virginie Verkarre
- Pathology Department, Georges Pompidou European Hospital, AP-HP Centre, Université Paris Cité, INSERM UMR970, Equipe Labellisée Ligue Contre Le Cancer, Paris, France
| | - Yann-Alexandre Vano
- Medical Oncology Department, Hôpital Foch, Suresnes, France
- Centre de Recherche Des Cordeliers, INSERM1138, Inflammation Complément Et Cancer, Université Paris Cité, Paris, France
| | - Eric Tartour
- Université Paris Cité, INSERM U970, PARCC, Paris, France.
- Immunology Department, Georges Pompidou European Hospital, AP-HP Centre, Université Paris Cité, Paris, France.
- Paris - Cardiovascular Research Center, European Georges Pompidou Hospital, 56 Rue Leblanc, Paris, 75015, France.
| | - Stéphane Oudard
- Université Paris Cité, INSERM U970, PARCC, Paris, France.
- Medical Oncology Department, Georges Pompidou European Hospital, CARPEM Cancer Institute, AP-HP Centre, Université Paris Cité, Paris, France.
- Paris - Cardiovascular Research Center, European Georges Pompidou Hospital, 56 Rue Leblanc, Paris, 75015, France.
| |
Collapse
|
21
|
Shani Shrem N, Beltran-Bless AA, Ghosh S, Tajzler C, Wood LA, Kollmannsberger C, Basappa NS, Graham J, Fallah-Rad N, Heng DY, Soulières D, Lalani AKA, Breau RH, Finelli A, Tanguay S, Bhindi B, Bjarnason G, Pouliot F, Canil C. Real-World Efficacy and Toxicity of Ipilimumab and Nivolumab as First-Line Treatment of Metastatic Renal Cell Carcinoma (mRCC) in a Subpopulation of Elderly and Poor Performance Status Patients. Cancers (Basel) 2025; 17:522. [PMID: 39941888 PMCID: PMC11816257 DOI: 10.3390/cancers17030522] [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: 12/11/2024] [Revised: 01/16/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Ipilimumab and nivolumab (ipi/nivo) improved overall survival (OS) compared to sunitinib in the pivotal Checkmate 214 trial of metastatic renal cell carcinoma (mRCC) with International Metastatic RCC Database Consortium (IMDC) intermediate/poor risk disease. We evaluated the efficacy and toxicity of ipi/nivo in older and frailer populations in a real-world mRCC cohort. METHODS Analysis was conducted on a real-world cohort with mRCC (N = 551) treated with first-line ipi/nivo from the Canadian Kidney Cancer information system (CKCis) database from January 2014 to December 2021. A comparison was made between outcomes and toxicity in patients 1. <70 versus (vs.) ≥70 yo, 2. <75 vs. ≥75 yo, and 3. KPS ≥70 vs. <70 yo. OS, progression-free survival (PFS), and time to treatment failure (TTF) were calculated by Kaplan-Meier analysis. Log-rank tests were used for comparison between groups. RESULTS Ipi/nivo treatment had no impact on survival outcomes or toxicity for patients >70 yo and >75 yo when controlled for IMDC. However, when comparing patients with KPS > 70 vs. KPS < 70, patients with a poor performance status had decreased median OS at 54.5 m vs. 10.8 m (p-value < 0.0001) and PFS at 11.6 vs. 3.1 m (p-value < 0.0001). CONCLUSIONS The use of ipi/nivo in mRCC demonstrated similar survival outcomes and toxicity in an older patient population. In patients with a poor performance status, it was associated with inferior OS and PFS. We believe that ipi/nivo is a reasonable treatment option for these patient populations, particularly in older patients.
Collapse
Affiliation(s)
- Noa Shani Shrem
- The Legacy Heritage Oncology Center & Dr. Larry Norton Institute, Soroka University Medical Center, Beer Sheva 84101, Israel
| | | | - Sunita Ghosh
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Camilla Tajzler
- Centre of Innovative Medicine, Research Institute—McGill University Health Centre, Montreal, QC H3H 2R9, Canada
| | - Lori A. Wood
- Capital Health Queen Elizabeth II Hospital, Halifax, NS B3H 2Y9, Canada
| | | | - Naveen S. Basappa
- Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | | | | | | | - Denis Soulières
- Centre Hospitalier de l’Université de Montréal, Montreal, QC H2X 0A9, Canada
| | - Aly-Khan A. Lalani
- Juravinski Cancer Centre, McMaster University, Hamilton, ON L8V 5C2, Canada
| | - Rodney H. Breau
- Division of Urology, Department of Surgery, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | | | - Simon Tanguay
- McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Bimal Bhindi
- Alberta Health Services, Calgary, AB T5J E34, Canada
| | - Georg Bjarnason
- Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
| | - Frederic Pouliot
- Centre Hospitalier Universitaire de Québec, Quebec, QC G1R 2J6, Canada
| | - Christina Canil
- Division of Medical Oncology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
22
|
Zou S, Cui L, Pai P, Lu Y, Li X, Wang G, Huang W, Wang D, Shaikh N, Peng Z, Peng Z, He H, Liao Z. Incidence and survival patterns of clear cell renal cell carcinoma from 2000 to 2017: A SEER Database Analysis. J Cancer 2025; 16:1591-1597. [PMID: 39991582 PMCID: PMC11843228 DOI: 10.7150/jca.105713] [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: 10/23/2024] [Accepted: 12/29/2024] [Indexed: 02/25/2025] Open
Abstract
Background: Clear cell renal cell carcinoma (ccRCC) incidence and death have considerably changed in recent years. Our study aimed to investigate the incidence, survival, and tumor characteristics of ccRCC in the year of diagnosis. Methods: Our study participants were selected from the SEER database (2000-2017). Age-standardized incidence rates were calculated to compare incidence rates across time. In addition, we used Kaplan-Meier curves to calculate overall survival (OS) and Cox proportional hazards models to explore risk factors associated with mortality outcomes in patients with ccRCC. Results: In the SEER analysis from 2000 to 2017, the increasing trend in age-adjusted incidence of ccRCC has remained relatively stable over the years, increasing from 2.63 per 100,000 in 2000 to 8.79 per 100,000 in 2017. The increase in the incidence of patients at a localized stage plays a decisive role in the overall increase in the incidence of ccRCC. Conclusions: In the general population, patients diagnosed between 2009-2017 had a higher survival rate than those diagnosed between 2000-2008, which is consistent with all stages of the tumor. The incidence of ccRCC increases steadily with the year of diagnosis, while overall survival has significantly improved.
Collapse
Affiliation(s)
- Sijue Zou
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Liwen Cui
- Department of Nephrology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Pearl Pai
- Department of Nephrology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Yiping Lu
- Department of Nephrology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - XiangYang Li
- Department of Nephrology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Gang Wang
- Department of Nephrology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Wen Huang
- Department of Nephrology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Dan Wang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Nikhat Shaikh
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Zhangzhe Peng
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhuoming Peng
- Department of Respiratory and Intensive Care Medicine, Union Shenzhen Hospital, Huazhong University of Science and Technology, Shenzhen 518000, Guangdong Province, China
| | - Haiyan He
- Department of Nephrology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Zhouning Liao
- Department of Nephrology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
23
|
Mahadevan A, Yazdanpanah O, Patel V, Benjamin DJ, Kalebasty AR. Ophthalmologic toxicities of antineoplastic agents in genitourinary cancers: Mechanisms, management, and clinical implications. Curr Probl Cancer 2025; 54:101171. [PMID: 39708456 DOI: 10.1016/j.currproblcancer.2024.101171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 11/19/2024] [Accepted: 11/29/2024] [Indexed: 12/23/2024]
Abstract
Genitourinary cancers affect over 480,000 patients in the United States annually. While promising therapeutic modalities continue to emerge, notably immune checkpoint inhibitors, molecular targeted therapies, antibody-drug conjugates, and radioligand therapies, these treatments are associated with a spectrum of adverse side-effects, including ophthalmologic toxicities. In this review, we cover the most commonly used antineoplastic agents for the kidneys, bladder, urinary tracts, prostate, testis, and penis, detailing mechanism, indication, and recent trials supporting their use. For each category of antineoplastic therapy, we describe the epidemiology, management, and clinical presentation, of common ophthalmologic toxicities stemming from these agents. This review serves to augment awareness and recognition of possible ophthalmologic manifestations resulting from the use of antineoplastic agents in genitourinary malignancy. Early identification of these side effects can hasten ophthalmology referral and ultimately improve visual outcomes in patients experiencing medication-induced ocular toxicities.
Collapse
Affiliation(s)
- Aditya Mahadevan
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA.
| | - Omid Yazdanpanah
- Division of Hematology/Oncology, University of California Irvine Health, Orange, CA, USA.
| | - Vivek Patel
- Department of Ophthalmology, University of California Irvine Health, Orange, CA, USA.
| | | | | |
Collapse
|
24
|
Bölek H, Sertesen E, Kuzu OF, Tural D, Sim S, Nahit Şendur MA, Uçar G, Işık S, Hacıoğlu B, Çiçin İ, Arslan Ç, Göksu SS, Sever ÖN, Karaçin C, Karadurmuş N, Özgüroğlu M, Yekedüz E, Ürün Y. Treatment Patterns and Attrition in Metastatic Renal Cell Carcinoma: Real-Life Experience from the Turkish Oncology Group Kidney Cancer Consortium (TKCC) Database. Clin Genitourin Cancer 2025; 23:102282. [PMID: 39709686 DOI: 10.1016/j.clgc.2024.102282] [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: 07/17/2024] [Revised: 11/24/2024] [Accepted: 11/25/2024] [Indexed: 12/24/2024]
Abstract
INTRODUCTION Despite the rapid evolution in management of metastatic renal cell carcinoma (mRCC) over the past decade, challenges remain in accessing new therapies in some parts of the world. Despite therapeutic advancements, attrition rates remain persistently high. This study aims to assess the treatment patterns and attrition rates of patients with mRCC in oncology clinics across Turkey. PATIENTS AND METHODS Patients diagnosed with mRCC between January 1, 2008, and December 31, 2022, with first-line systemic treatment data, were retrospectively evaluated using the Turkish Oncology Group Kidney Cancer Consortium (TKCC) Database. RESULTS The final analysis included a total of 1126 patients. The percentages of patients treated in the 2nd, 3rd, 4th, and 5th lines of therapy were 62.8%, 27.4%, 8.9%, and 2.1%, respectively. The drugs that were most commonly used in the groups were tyrosine kinase inhibitors (TKIs) (52.2%) and interferon (IFN)-alpha (43.3%) for the first line, TKIs (66.3%) and immunotherapy (IO) monotherapy (25.9%) for the second line, TKI (41.4%) and mTOR inhibitors (28.8%) for the third line, TKI (44.4%) and mTOR inhibitors (29%) for the fourth line, and IO monotherapy (37.5%) and TKI (25%) for the fifth line. For the first-line treatment, the primary cause of attrition was disease progression (66.4%), followed by toxicity (16.5%), death (11.2%), and patient preference (5.9%). The primary reason for attrition across all treatment lines was disease progression. Over time, the use of TKIs in first-line treatment increased, while IFN-alpha usage declined. IOs began to be utilized in earlier lines, predominantly in second-line treatment, though use of IO-based combination therapies remains limited. CONCLUSION This study underscores that despite significant progress in therapeutic options, the adoption of novel agents remains slow, and attrition rates are still high. These findings indicate a disparity in systemic therapy compared to developed countries.
Collapse
Affiliation(s)
- Hatice Bölek
- Ankara University School of Medicine, Department of Medical Oncology, Ankara, Turkey; Ankara University Cancer Institute, Ankara, Turkey
| | - Elif Sertesen
- University of Health Science, Dr Abdurrahman Yurtaslan Oncology Training and Research Hospital, Department of Medical Oncology, Ankara Turkey
| | - Omer Faruk Kuzu
- University of Health Science, Gülhane Training and Research Hospital, Department of Medical Oncology, Ankara Turkey
| | - Deniz Tural
- University of Health Science, Bakirkoy Dr. Sadi Konuk Training and Research Hospital, Department of Medical Oncology, Istanbul, Turkey
| | - Saadet Sim
- Ege University School of Medicine, Department of Medical Oncology, İzmir, Turkey
| | | | - Gökhan Uçar
- Bilkent City Hospital, Department of Medical Oncology, Ankara Turkey
| | - Selver Işık
- Marmara University School of Medicine, Department of Medical Oncology, Istanbul, Turkey
| | - Bekir Hacıoğlu
- Trakya University School of Medicine, Department of Medical Oncology, Edirne, Turkey
| | - İrfan Çiçin
- Istinye University, Department of Medical Oncology, Istanbul, Turkey
| | - Çağatay Arslan
- Izmir University of Economics, Medical Point Hospital, Izmir, Turkey
| | - Sema Sezgin Göksu
- Akdeniz University School of Medicine, Department of Medical Oncology, Antalya, Turkey
| | - Özlem Nuray Sever
- Kartal Dr Lutfi Kirdar City Hospital, Department of Medical Oncology, Istanbul, Turkey
| | - Cengiz Karaçin
- University of Health Science, Dr Abdurrahman Yurtaslan Oncology Training and Research Hospital, Department of Medical Oncology, Ankara Turkey
| | - Nuri Karadurmuş
- University of Health Science, Gülhane Training and Research Hospital, Department of Medical Oncology, Ankara Turkey
| | - Mustafa Özgüroğlu
- Cerrahpasa School of Medicine, Department of Medical Oncology, Istanbul, Turkey
| | - Emre Yekedüz
- Dana-Farber Cancer Institute, Department of Medical Oncology, Boston, MA
| | - Yüksel Ürün
- Ankara University School of Medicine, Department of Medical Oncology, Ankara, Turkey; Ankara University Cancer Institute, Ankara, Turkey.
| |
Collapse
|
25
|
Salimi Asl A, Davari M, Ghorbani A, Seddighi N, Arabi K, Saburi E. Neoadjuvant immunotherapy and oncolytic virotherapy in HPV positive and HPV negative skin cancer: A comprehensive review. Int Immunopharmacol 2025; 146:113790. [PMID: 39673996 DOI: 10.1016/j.intimp.2024.113790] [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: 10/07/2024] [Revised: 12/03/2024] [Accepted: 12/03/2024] [Indexed: 12/16/2024]
Abstract
Skin cancer is the most common new cancer among Caucasians. This cancer has different types, of which non-melanoma skin cancer is the most common type. Various factors affect this disease, one of which is viral infections, including HPV. This virus plays an important role in skin cancer, especially cSCCs. There are various options for the treatment of skin cancer, and today special attention has been paid to treatments based on therapeutic goals, immunotherapy and combination therapy. In this study, we have investigated treatments based on immunotherapy and virotherapy and the effect of HPV virus on the effectiveness of these treatments in skin cancer. Treatments based on virotherapy are performed for a long time in combination with other common treatments such as radiotherapy and chemotherapy in order to have a greater effect and lower its side effects, which include: shortness of breath, tachycardia, lowering blood pressure in the patient. Also, the most important axis of immunotherapy is to focus on PD1-PDL1, despite abundant evidence on the importance of immunotherapy, many studies investigate the use of immunotherapy inhibitors in the adjuvant and neoadjuvant setting in various cancers. Also, previous findings show conflicting evidence of the effect of HPV status on the response to immunotherapy.
Collapse
Affiliation(s)
- Ali Salimi Asl
- Student Research Committee, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| | - Mohsen Davari
- Student Research Committee, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| | - Atousa Ghorbani
- Department of Biology, North Tehran Branch, Islamic Azad University, Tehran, Iran; Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran.
| | - Narjes Seddighi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Kimia Arabi
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran.
| | - Ehsan Saburi
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Genetics and Molecular Medicine Department, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
26
|
Watanabe R, Kagimoto K, Chosei M, Sakaue T, Kurata M, Miura N, Kitazawa R, Kikugawa T, Higashiyama S, Saika T. Vesicles Secreted by Renal Cell Carcinoma Cells Cause Vascular Endothelial Cells to Express PSMA and Drive Tumor Progression. Cells 2025; 14:165. [PMID: 39936957 PMCID: PMC11817738 DOI: 10.3390/cells14030165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 02/13/2025] Open
Abstract
Prostate-specific membrane antigen (PSMA) protein expression is induced during prostate cancer progression and metastasis. Recently, we reported that PSMA-positive vesicles released by prostate cancer cell lines enhanced vascular endothelial cell angiogenesis and that PSMA may be involved in tumor angiogenesis. Similarly, it is known that PSMA is upregulated in peritumoral vessels in renal cell carcinoma (RCC). In this study, we investigated the significance and molecular function of PSMA in RCC. PSMA immunohistochemical staining confirmed PSMA presence only in perinephric tumor vessels, and PSMA intensity was strongly correlated with recurrence rate and venous invasion. Spatial gene expression analysis revealed that FOLH1 expression, which codes PSMA, was upregulated in tumor blood vessels around renal cancer, and that angiogenesis-related pathways were enhanced. The 10,000 g pellet fraction of the renal cancer cell lines Caki1- and ACHN-conditioned medium (CM) induced PSMA positivity in human umbilical vein endothelial cells (HUVECs) and enhanced tube formation. Mass spectrometry indicated that the 10,000 g pellet fraction contained various kinds of growth factors, like GDF15 and MYDGF. RNA sequencing showed that supplementing HUVECs with RCC cell CM-enhanced angiogenesis-related signaling pathways. Conclusively, microvesicle components secreted by RCC cells transform vascular endothelial cells into PSMA-positive cells, enhancing angiogenesis.
Collapse
MESH Headings
- Humans
- Carcinoma, Renal Cell/pathology
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/blood supply
- Kidney Neoplasms/pathology
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/blood supply
- Kidney Neoplasms/genetics
- Glutamate Carboxypeptidase II/metabolism
- Glutamate Carboxypeptidase II/genetics
- Cell Line, Tumor
- Disease Progression
- Human Umbilical Vein Endothelial Cells/metabolism
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/metabolism
- Antigens, Surface/metabolism
- Antigens, Surface/genetics
- Male
- Gene Expression Regulation, Neoplastic
- Endothelial Cells/metabolism
Collapse
Affiliation(s)
- Ryuta Watanabe
- Department of Urology, Ehime University Graduate School of Medicine, Toon 791-0295, Japan; (K.K.); (N.M.); (T.K.); (T.S.)
| | - Keito Kagimoto
- Department of Urology, Ehime University Graduate School of Medicine, Toon 791-0295, Japan; (K.K.); (N.M.); (T.K.); (T.S.)
| | - Mami Chosei
- Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Toon 791-0295, Japan; (M.C.); (S.H.)
| | - Tomohisa Sakaue
- Department of Cardiovascular and Thoracic Surgery, Ehime University Graduate School of Medicine, Toon 791-0295, Japan;
- Division of Cell Growth and Tumor Regulation Proteo-Science Center, Ehime University, Toon 791-0295, Japan
| | - Mie Kurata
- Department of Analytical Pathology, Ehime University Graduate School of Medicine, Toon 791-0295, Japan;
- Department of Pathology, Ehime University Graduate School of Medicine and Proteo-Science Center, Toon 791-0295, Japan
| | - Noriyoshi Miura
- Department of Urology, Ehime University Graduate School of Medicine, Toon 791-0295, Japan; (K.K.); (N.M.); (T.K.); (T.S.)
| | - Riko Kitazawa
- Division of Diagnostic Pathology, Ehime University Hospital, Toon 791-0295, Japan;
| | - Tadahiko Kikugawa
- Department of Urology, Ehime University Graduate School of Medicine, Toon 791-0295, Japan; (K.K.); (N.M.); (T.K.); (T.S.)
| | - Shigeki Higashiyama
- Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Toon 791-0295, Japan; (M.C.); (S.H.)
- Division of Cell Growth and Tumor Regulation Proteo-Science Center, Ehime University, Toon 791-0295, Japan
- Department of Oncogenesis and Growth Regulation, Osaka International Cancer Institute, Chuo-ku, Osaka 541-8567, Japan
| | - Takashi Saika
- Department of Urology, Ehime University Graduate School of Medicine, Toon 791-0295, Japan; (K.K.); (N.M.); (T.K.); (T.S.)
| |
Collapse
|
27
|
Moinard-Butot F, Thouvenin J, Bigot P, Martinez-Chanza N, Gaillard V, Cazzato RL, Boissier R, Margue G, Boudier P, Maillet D, Gross-Goupil M, Bernhard JC, Barthélémy P. Efficacy of immune checkpoint inhibitors in renal cell carcinoma venous tumour thrombus shrinkage (UroCCR 128). World J Urol 2025; 43:66. [PMID: 39792158 PMCID: PMC11723841 DOI: 10.1007/s00345-024-05428-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: 10/06/2024] [Accepted: 12/26/2024] [Indexed: 01/12/2025] Open
Abstract
PURPOSE Surgery remains the cornerstone of localized renal cell carcinoma (RCC) care. Pembrolizumab has recently been recommended as a standard of care for RCC patients who are at high risk of recurrence. Data regarding the efficacy of ICIs either alone or in combination with ICIs or VEGF TKIs for VTT shrinkage are scarce. METHODS In the framework of the French kidney cancer research network UroCCR (NCT03293563), we performed a retrospective multicentric European study to evaluate VTT shrinkage in patients treated with ICIs with metastatic or locally advanced renal cell carcinoma (RCC). The primary endpoint was the objective response rate (ORR) of patients with VTT to ICI-based therapy. Radiological assessment was performed by a treating physician according to the RECISTv1.1 criteria. RESULTS We included 44 patients. The median age was 69 years (range 37-88). All patients was intermediate or poor IMDC risk group. Twenty-three patients were treated with anti-PD-1 in combination with anti-CTLA-4 therapy, 13 patients with ICI monotherapy, and 8 patients with ICIs in combination with antiangiogenic TKI. At baseline, the median VTT diameter was 22 mm (range 7-93). After a median duration of treatment of 5.8 months (range 1.8-39.1), the ORR was 38% (n = 17), including 4 complete responses (CRs) and 13 partial responses (PRs). Ten patients had stable disease (SD), and 17 had progressive disease (PD) as the best response of the VTT. CONCLUSION These data highlight the potential efficacy of ICIs to shrink the VTT even if they seem to have little impact on the extent of VTT.
Collapse
Affiliation(s)
- Fabien Moinard-Butot
- Medical Oncology Department, Institut de Cancérologie Strasbourg Europe, Strasbourg, France
| | - Jonathan Thouvenin
- Medical Oncology Department, Hospices Civils de Lyon, Pierre-Bénite, France
| | - Pierre Bigot
- Department of Urology, Angers University Hospital, Angers, France
- Kidney Cancer Group of the French Association of Urology Cancer Committee, Paris, France
| | | | - Victor Gaillard
- Urology Department, Nouvel Hôpital Civil, Strasbourg, France
| | | | - Romain Boissier
- Department of Urology and Renal Transplantation, La Conception University Hospital, APHM, Marseille, France
- Kidney Cancer Group of the French Association of Urology Cancer Committee, Paris, France
| | - Gaëlle Margue
- Department of Urology, Bordeaux University Hospital, Bordeaux, France
- Kidney Cancer Group of the French Association of Urology Cancer Committee, Paris, France
| | - Philippe Boudier
- Medical Oncology Department, Institut de Cancérologie Strasbourg Europe, Strasbourg, France
| | - Denis Maillet
- Medical Oncology Department, Hospices Civils de Lyon, Pierre-Bénite, France
| | | | - Jean-Christophe Bernhard
- Department of Urology, Bordeaux University Hospital, Bordeaux, France
- Kidney Cancer Group of the French Association of Urology Cancer Committee, Paris, France
| | - Philippe Barthélémy
- Medical Oncology Department, Institut de Cancérologie Strasbourg Europe, Strasbourg, France.
- Kidney Cancer Group of the French Association of Urology Cancer Committee, Paris, France.
| |
Collapse
|
28
|
Dumbrava EE, Ben Haj Frej K, Sharon E, Tawbi H. Application and Expectations for Immune Checkpoint Blockade of LAG3 and TIGIT. Annu Rev Med 2025; 76:189-205. [PMID: 39656959 DOI: 10.1146/annurev-med-080222-100847] [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] [Indexed: 12/17/2024]
Abstract
Immune checkpoint blockade targeting the novel targets of the lymphocyte activation gene 3 (LAG3) and the T cell immunoreceptor with Ig and immunoreceptor tyrosine-based inhibition motif domains (TIGIT) has marked a significant advancement in oncology, offering new therapeutic opportunities to fight diverse malignancies. This review covers the biological basis and clinical application of LAG3 and TIGIT inhibitors, highlighting pivotal trials and therapeutic outcomes. We underscore the use of dual therapy immune checkpoint blockade in enhancing antitumor immunity, particularly in settings where monotherapy has shown limited efficacy. Additionally, we address the emerging challenges such as treatment resistance and adverse effects. We explore the strategic integration of LAG3 and TIGIT blockade within the broader immunotherapy landscape, emphasizing innovative combinations and the quest for predictive biomarkers to optimize patient selection and treatment efficacy.
Collapse
Affiliation(s)
- Ecaterina Elena Dumbrava
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA;
| | | | - Elad Sharon
- Immunotherapy Toxicity Service, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Hussein Tawbi
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
29
|
Failla CM, Carbone ML, Ramondino C, Bruni E, Orecchia A. Vascular Endothelial Growth Factor (VEGF) Family and the Immune System: Activators or Inhibitors? Biomedicines 2024; 13:6. [PMID: 39857591 PMCID: PMC11763294 DOI: 10.3390/biomedicines13010006] [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: 11/08/2024] [Revised: 11/29/2024] [Accepted: 12/20/2024] [Indexed: 01/27/2025] Open
Abstract
The vascular endothelial growth factor (VEGF) family includes key mediators of vasculogenesis and angiogenesis. VEGFs are secreted by various cells of epithelial and mesenchymal origin and by some immune cells in response to physiological and pathological stimuli. In addition, immune cells express VEGF receptors and/or co-receptors and can respond to VEGFs in an autocrine or paracrine manner. This immunological role of VEGFs has opened the possibility of using the VEGF inhibitors already developed to inhibit tumor angiogenesis also in combination approaches with different immunotherapies to enhance the action of effector T lymphocytes against tumor cells. This review pursues to examine the current understanding of the interplay between VEGFs and the immune system, while identifying key areas that require further evaluation.
Collapse
Affiliation(s)
- Cristina Maria Failla
- Experimental Immunology Laboratory, Istituto Dermopatico dell’Immacolata, IDI-IRCCS, 00167 Rome, Italy; (C.M.F.); (C.R.)
| | - Maria Luigia Carbone
- Clinical Trial Center, Istituto Dermopatico dell’Immacolata, IDI-IRCCS, 00167 Rome, Italy;
| | - Carmela Ramondino
- Experimental Immunology Laboratory, Istituto Dermopatico dell’Immacolata, IDI-IRCCS, 00167 Rome, Italy; (C.M.F.); (C.R.)
| | - Emanuele Bruni
- Departmental Faculty of Medicine and Surgery, UniCamillus-Saint Camillus International University of Health Sciences, 00131 Rome, Italy
| | | |
Collapse
|
30
|
Hosseininejad-Chafi M, Eftekhari Z, Oghalaie A, Behdani M, Sotoudeh N, Kazemi-Lomedasht F. Nanobodies as innovative immune checkpoint modulators: advancing cancer immunotherapy. Med Oncol 2024; 42:36. [PMID: 39719469 DOI: 10.1007/s12032-024-02588-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 12/14/2024] [Indexed: 12/26/2024]
Abstract
The immune system relies on a delicate balance between attacking harmful pathogens and preserving the body's own tissues, a balance maintained by immune checkpoints. These checkpoints play a critical role in preventing autoimmune diseases by restraining excessive immune responses while allowing the immune system to recognize and destroy abnormal cells, such as tumors. In recent years, immune checkpoint inhibitors (ICIs) have become central to cancer therapy, enabling the immune system to target and eliminate cancer cells that evade detection. Traditional antibodies, such as IgGs, have been widely used in immune therapies but are limited by their size and complexity. Nanobodies (Nbs), derived from camelid heavy-chain-only antibodies, offer a promising alternative. These small, stable antibody fragments retain the antigen-binding specificity of traditional antibodies but have enhanced solubility and the ability to target otherwise inaccessible epitopes. This review explores the use of Nbs as ICIs, emphasizing their potential in cancer immunotherapy and other immune-related treatments. Their unique structural properties and small size make Nbs highly effective tools for modulating immune responses, representing a novel approach in the evolving landscape of checkpoint inhibitor therapies.
Collapse
Affiliation(s)
- Mohammad Hosseininejad-Chafi
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Zohre Eftekhari
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Akbar Oghalaie
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Mahdi Behdani
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Nazli Sotoudeh
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Fatemeh Kazemi-Lomedasht
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran.
| |
Collapse
|
31
|
Figaroa O, Zondervan P, Kessels R, Berkhof J, Aarts M, Hamberg P, Los M, Piersma D, Rikhof B, Suelmann B, Tascilar M, van der Veldt A, Verhagen P, Westgeest H, Yildirim H, Bex A, Bins A. PrimerX: A Bayesian Multistage Cohort Embedded Randomised Trial to Evaluate the Role of Deferred Local Therapy of the Primary Tumour in Combination with Immune Checkpoint Inhibitor-based First-line Therapy in Metastatic Renal Cell Carcinoma Patients. EUR UROL SUPPL 2024; 70:28-35. [PMID: 39483517 PMCID: PMC11525453 DOI: 10.1016/j.euros.2024.09.002] [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] [Accepted: 09/19/2024] [Indexed: 11/03/2024] Open
Abstract
Background Historically, patients with metastatic renal cell carcinoma (mRCC) have been offered upfront cytoreductive nephrectomy (CN) followed by systemic therapy. Currently, CN is no longer the standard of care (SOC) based on the randomised phase 3 CARMENA study performed in the vascular endothelial growth factor receptor tyrosine kinase inhibitor era. With the advent of immune checkpoint inhibitor (ICI) combination therapy in first line, the role of CN needs to be reassessed. There is indirect evidence from small retrospective series that deferred CN after ICI combination therapy may lead to better outcomes. To reassess the role of CN, we designed PrimerX, a randomised controlled trial following the Trial within Cohorts (TwiCs) study design. The primary objective of this study is to re-evaluate the benefit of deferred local treatment in the current era of immunotherapy. Study design This PrimerX study has been designed as a TwiCs study within the Dutch Prospective Renal Cell Carcinoma (PRO-RCC) cohort. The PRO-RCC cohort includes patients with mRCC and nonmetastatic RCC, and has been set up for prospective collection of long-term clinical data and as an infrastructure for initiating TwiCs studies. The PrimerX TwiCs trial follows a Bayesian adaptive multistage design to allow for early discontinuation due to futility or efficacy. PrimerX has appropriate ethics approval and is registered at clinical.trials.gov (NCT05941169). End points The primary clinical endpoint is overall survival, defined as the time from randomisation to death from any cause. The secondary endpoint is the objective response rate within the primary tumour prior to local therapy, as assessed by a computed tomography scan. Patients and methods A maximum of 700 patients with synchronous mRCC and absence of progression at metastatic sites following at least 6 mo of standard first-line ICI combination therapy will be assigned randomly to receive local treatment of the primary tumour (experimental arm) or SOC (control arm). The experimental intervention consists of (partial) CN, any form of ablative local therapy, or magnetic resonance imaging guided ablative stereotactic radiotherapy, performed within 6 mo and 1.5 yr after the start of systemic treatment.
Collapse
Affiliation(s)
- Orlane Figaroa
- Department of Medical Oncology, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, The Netherlands
- Department of Urology, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam University Medical Centres, Amsterdam, The Netherlands
| | - Patricia Zondervan
- Department of Urology, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, The Netherlands
| | - Rob Kessels
- Julius Center for Health Sciences and Primary Care, Department of Data Science and Biostatistics, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Johannes Berkhof
- Department of Epidemiology and Data Science, Amsterdam UMC location Vrije Universiteit, Amsterdam, The Netherlands
| | - Maureen Aarts
- Department of Medical Oncology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Paul Hamberg
- Department of Internal Medicine, Franciscus Gasthuis, Vlietland, Rotterdam/Schiedam, The Netherlands
| | - Maartje Los
- Department of Medical Oncology, St. Antonius Ziekenhuis, Nieuwegein/Utrecht, The Netherlands
| | - Djura Piersma
- Department of Medical Oncology, Medical Spectrum Twente, Enschede, The Netherlands
| | - Bart Rikhof
- Department of Medical Oncology, Leeuwarden Medical Center, Leeuwarden, The Netherlands
| | - Britt Suelmann
- Department of Medical Oncology, Utrecht University Medical Centre, Utrecht, The Netherlands
| | - Metin Tascilar
- Department of Medical Oncology, Isala Medical Centre, Zwolle, The Netherlands
| | - Astrid van der Veldt
- Department of Medical Oncology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Paul Verhagen
- Department of Urology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Hans Westgeest
- Department of Medical Oncology, Amphia Medical Centre, Breda, The Netherlands
| | - Hilin Yildirim
- Department of Medical Oncology, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, The Netherlands
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation, Utrecht, The Netherlands
| | - Axel Bex
- Department of Urology, Antoni van Leeuwenhoek Hospital-The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Specialist Centre for Kidney Cancer, The Royal Free London NHS Foundation Trust, London, UK
| | - Adriaan Bins
- Department of Medical Oncology, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam University Medical Centres, Amsterdam, The Netherlands
| |
Collapse
|
32
|
Du EX, Betts KA, Wang T, Kitchen SA, He X, Yin X, Guttenplan SB, Beauchamp K, Delgado A, Rosenblatt L. Long-Term Temporal Trends of Real-World Healthcare Costs Associated with Nivolumab Plus Ipilimumab and Pembrolizumab Plus Axitinib as First-Line Treatment for Advanced or Metastatic Renal Cell Carcinoma. Oncol Ther 2024; 12:735-751. [PMID: 39127872 PMCID: PMC11573941 DOI: 10.1007/s40487-024-00297-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
INTRODUCTION Nivolumab plus ipilimumab (NIVO + IPI) and pembrolizumab plus axitinib (PEM + AXI) are first-line (1L) treatments for advanced or metastatic renal cell carcinoma (aRCC), although the long-term trends in their associated real-world healthcare costs are not well defined. We compared the real-world healthcare costs of patients with aRCC who received 1L NIVO + IPI or PEM + AXI over 24 months. METHODS Adults with RCC and secondary malignancy who initiated 1L NIVO + IPI or PEM + AXI were identified in the Merative MarketScan Commercial and Medicare Supplemental Databases (01/01/2004 to 09/30/2021). All-cause and RCC-related healthcare costs (unadjusted and adjusted) were assessed per patient per month (PPPM) at 6-month intervals post-treatment initiation (index date) up to 24 months, and differences between the NIVO + IPI and PEM + AXI cohorts were compared. RESULTS Of 325 patients with aRCC, 219 received NIVO + IPI and 106 received PEM + AXI as the 1L treatment. According to patients' follow-up length, the analyses for months 7-12 included 210 patients in the NIVO + IPI cohort and 103 in the PEM + AXI cohort; months 13-18 included 119 and 48 patients, respectively; and months 19-24 included 81 and 25 patients. PPPM unadjusted all-cause total costs were $46,348 for NIVO + IPI and $38,097 for PEM + AXI in months 1-6; $26,840 versus $27,983, respectively, in months 7-12; $22,899 versus $25,137 in months 13-18; and $22,279 versus $27,947 in months 19-24. PPPM unadjusted RCC-related costs were $44,059 for NIVO + IPI and $36,456 for PEM + AXI in months 1-6; $25,144 versus $26,692, respectively, in months 7-12; $21,645 versus $23,709 in months 13-18; and $20,486 versus $25,515 in months 19-24. PPPM costs declined more rapidly for patients receiving NIVO + IPI compared to those receiving PEM + AXI, resulting in significantly lower all-cause costs associated with NIVO + IPI during months 19-24 (difference - $10,914 [95% confidence interval - $21,436, - $1091]) and RCC-related costs during months 7-12 (- $4747 [(- $8929, - $512]) and 19-24 (- $10,261 [- $20,842, - $421]) after adjustment. Cost savings for NIVO + IPI versus PEM + AXI were driven by differences in drug costs which, after adjustment, were significantly lower in months 7-12 (difference - $5555 [all-cause], - $5689 [RCC-related]); 13-18 (- $7217 and - $6870, respectively); and 19-24 (- $16,682 and - $16,125). CONCLUSION Although the real-world PPPM healthcare costs of 1L NIVO + IPI were higher compared with PEM + AXI in the first 6 months of treatment, the costs associated with NIVO + IPI rapidly declined thereafter, resulting in significantly lower costs vs. PEM + AXI from months 7 to 24.
Collapse
Affiliation(s)
| | | | | | | | - Xuanhao He
- Analysis Group, Inc., Los Angeles, CA, USA
| | - Xin Yin
- Bristol Myers Squibb, Princeton, NJ, USA
| | | | | | | | | |
Collapse
|
33
|
Choueiri TK, Kuzel TM, Tykodi SS, Verzoni E, Kluger H, Nair S, Perets R, George S, Gurney H, Pachynski RK, Folefac E, Castonguay V, Lee CH, Vaishampayan U, Miller WH, Bhagavatheeswaran P, Wang Y, Gupta S, DeSilva H, Lee CW, Escudier B, Motzer RJ. Nivolumab plus relatlimab and nivolumab plus ipilimumab for patients with advanced renal cell carcinoma: results from the open-label, randomised, phase II FRACTION-RCC trial. ESMO Open 2024; 9:104073. [PMID: 39642635 PMCID: PMC11667034 DOI: 10.1016/j.esmoop.2024.104073] [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: 05/31/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 12/09/2024] Open
Abstract
BACKGROUND The Fast Real-time Assessment of Combination Therapies in Immuno-ONcology study in patients with aRCC (FRACTION-RCC) was designed to assess new immuno-oncology (IO) combinations in patients with advanced renal cell carcinoma (aRCC). We present results in IO-naive patients treated with nivolumab (NIVO) + relatlimab (RELA) or NIVO + ipilimumab (IPI) in track 1. METHODS The open-label, randomised, phase II FRACTION-RCC trial enrolled patients with aRCC from 32 hospitals and cancer centres across six countries. Patients were enrolled in track 1 (IO-naive) or track 2 (IO-experienced). IO-naive patients were stratified by previous tyrosine kinase inhibitor therapy and randomised to NIVO (240 mg) + RELA (80 mg) intravenously once every 2 weeks or NIVO (3 mg/kg) + IPI (1 mg/kg) intravenously once every 3 weeks for four doses, followed by NIVO (480 mg) once every 4 weeks, each up to ∼2 years. The primary endpoints were objective response by investigator (RECIST version 1.1), duration of response (DOR), and progression-free survival (PFS) rate at 24 weeks. Safety was a secondary endpoint; biomarker analyses were exploratory. RESULTS FRACTION-RCC enrolled patients between 2 February 2017 and 23 January 2020. In track 1, 30 patients each were treated with NIVO + RELA or NIVO + IPI (clinical database lock, 1 November 2021). With NIVO + RELA [median follow-up, 48.6 months; interquartile range (IQR) 46.9-51.7 months], objective response was 30% [95% confidence interval (CI) 15% to 49%], with 33 weeks (95% CI 16-53 weeks) median DOR. The PFS rate at 24 weeks was 43% (95% CI 25% to 60%). With NIVO + IPI (median follow-up, 48.7 months; IQR 47.1-52.0 months), the objective response was 20% (95% CI 8% to 39%), with the median DOR not reached (95% CI 33 weeks-not estimable). The PFS rate at 24 weeks was 49% (95% CI 29% to 66%). Higher baseline lymphocyte activation gene 3 (LAG-3) and programmed death-ligand 1 (PD-L1) expression levels were detected among track 1 NIVO + RELA responders. Grade 3-4 treatment-related adverse events were reported in 4/30 (13%) patients treated with NIVO + RELA and 10/30 (33%) patients treated with NIVO + IPI. No deaths were attributed to study treatments. CONCLUSIONS Results showed antitumour activity and manageable safety with NIVO + RELA. Findings also support NIVO + IPI as an effective combination regimen in IO-naive patients with aRCC.
Collapse
Affiliation(s)
- T K Choueiri
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - T M Kuzel
- Division of Hematology/Oncology/Cell Therapy, Rush University Medical Center, Chicago, IL, USA
| | - S S Tykodi
- Department of Medicine, Division of Hematology and Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - E Verzoni
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - H Kluger
- Section of Medical Oncology, Yale Cancer Center, New Haven, CT, USA
| | - S Nair
- Department of Hematology/Oncology, Lehigh Valley Topper Cancer Institute, Allentown, PA, USA
| | - R Perets
- Division of Oncology, Clinical Research Institute at Rambam, Rambam Health Care Campus, Technion - Israel Institute of Technology, Haifa, Israel
| | - S George
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - H Gurney
- Department of Clinical Medicine, Macquarie University, Sydney, NSW, Australia
| | - R K Pachynski
- Siteman Cancer Center, Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - E Folefac
- Department of Medical Oncology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - V Castonguay
- Department of Medicine, CHU de Quebec -Université Laval, Quebec City, Quebec, Canada
| | - C-H Lee
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - U Vaishampayan
- University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA; Department of Internal Medicine, Michigan Medicine, Ann Arbor, MI, USA
| | - W H Miller
- Segal Cancer Centre and Lady Davis Institute, Jewish General Hospital, Montréal, Quebec, Canada; Department of Oncology, McGill University, Montréal, Quebec, Canada
| | - P Bhagavatheeswaran
- Department of Biometrics and Data Sciences, Bristol Myers Squibb, Princeton, NJ, USA
| | - Y Wang
- Department of Biometrics and Data Sciences, Bristol Myers Squibb, Princeton, NJ, USA
| | - S Gupta
- Department of Translational Medicine Oncology, Bristol Myers Squibb, Princeton, NJ, USA
| | - H DeSilva
- Department of Global Drug Development, Bristol Myers Squibb, Princeton, NJ, USA
| | - C-W Lee
- Department of Clinical Trials, Bristol Myers Squibb, Princeton, NJ, USA
| | - B Escudier
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - R J Motzer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
34
|
Yan J, Yang L, Ren Q, Zhu C, Du H, Wang Z, Qi Y, Xian X, Chen D. Gut microbiota as a biomarker and modulator of anti-tumor immunotherapy outcomes. Front Immunol 2024; 15:1471273. [PMID: 39669573 PMCID: PMC11634861 DOI: 10.3389/fimmu.2024.1471273] [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: 07/27/2024] [Accepted: 10/25/2024] [Indexed: 12/14/2024] Open
Abstract
Although immune-checkpoint inhibitors (ICIs) have significantly improved cancer treatment, their effectiveness is limited by primary or acquired resistance in many patients. The gut microbiota, through its production of metabolites and regulation of immune cell functions, plays a vital role in maintaining immune balance and influencing the response to cancer immunotherapies. This review highlights evidence linking specific gut microbial characteristics to increased therapeutic efficacy in a variety of cancers, such as gastrointestinal cancers, melanoma, lung cancer, urinary system cancers, and reproductive system cancers, suggesting the gut microbiota's potential as a predictive biomarker for ICI responsiveness. It also explores the possibility of enhancing ICI effectiveness through fecal microbiota transplantation, probiotics, prebiotics, synbiotics, postbiotics, and dietary modifications. Moreover, the review underscores the need for extensive randomized controlled trials to confirm the gut microbiota's predictive value and to establish guidelines for microbiota-targeted interventions in immunotherapy. In summary, the article suggests that a balanced gut microbiota is key to maximizing immunotherapy benefits and calls for further research to optimize microbiota modulation strategies for cancer treatment. It advocates for a deeper comprehension of the complex interactions between gut microbiota, host immunity, and cancer therapy, aiming for more personalized and effective treatment options.
Collapse
Affiliation(s)
- Jiexi Yan
- The Precision Medicine Laboratory, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Lu Yang
- The State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, Jiangsu, China
| | - Qingmiao Ren
- The Precision Medicine Laboratory, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Chan Zhu
- The State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, Jiangsu, China
| | - Haiyun Du
- The Precision Medicine Laboratory, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Zhouyu Wang
- The State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, Jiangsu, China
| | - Yaya Qi
- The Precision Medicine Laboratory, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Xiaohong Xian
- The Precision Medicine Laboratory, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Dongsheng Chen
- The State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, Jiangsu, China
| |
Collapse
|
35
|
Pacholczak-Madej R, Drobniak A, Stokłosa Ł, Bidas A, Dobrzańska J, Grela-Wojewoda A, Roman A, Tusień-Małecka D, Walocha J, Blecharz P, Puskulluoglu M. Adverse events after nivolumab and ipilimumab combined immunotherapy in advanced renal cell carcinoma: a multicentre experience in Poland. BMC Cancer 2024; 24:1411. [PMID: 39548483 PMCID: PMC11566080 DOI: 10.1186/s12885-024-13192-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 11/12/2024] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have been employed in the adjuvant and metastatic setting of renal cell carcinoma (RCC) treatment. Among ICIs, combined immunotherapy has the highest risk for immune-related adverse events (irAEs). We aimed to document the incidence of irAEs in RCC patients treated with nivolumab and ipilimumab as data from the European population remain limited. MATERIALS AND METHODS We analysed data from 88 RCC patients treated with nivolumab + ipilimumab between May 2022 and June 2024 across six high-volume oncology units in Poland. We reviewed irAEs and estimated their impact on survival parameters via univariate and multivariate Cox proportional hazards regression models, along with log-rank tests. RESULTS With a median follow-up of 11.3 months, the median overall survival (OS) was not reached, whereas the median progression-free survival (PFS) was 12.8 months (6.3-19.3). A total of 74 irAEs were recorded in 50 patients. The most frequent events were endocrine (n = 20, 27%), hepatic (n = 15, 17%), general (n = 12, 13.6%), and cutaneous (n = 11, 12.5%). The occurrence of irAEs was associated with a 60% lower risk of disease progression (hazard ratio 0.44, 95% confidence interval 0.2-0.87, p = 0.018) without impacting OS and higher disease control rate (n = 45, 90% vs. n = 24, 63.2%, p = 0.004). In contrast, patients with hepatotoxicity had poorer outcomes, with a 2.6-fold greater risk of death (p = 0.05). CONCLUSIONS IrAEs may serve as a predictive factor for the efficacy of the nivolumab + ipilimumab regimen in RCC patients. Special attention is needed for hepatotoxicity, as it can significantly impact survival outcomes.
Collapse
Affiliation(s)
- Renata Pacholczak-Madej
- Department of Chemotherapy, The District Hospital, Sucha Beskidzka, Poland.
- Department of Gynecological Oncology, Krakow Branch, Maria Sklodowska-Curie National Research Institute of Oncology, Garncarska 11 Street, Krakow, 31-115, Poland.
- Department of Anatomy, Medical College, Jagiellonian University, Krakow, Poland.
| | - Artur Drobniak
- Department of Chemotherapy, The District Hospital, Sucha Beskidzka, Poland
| | - Łukasz Stokłosa
- Department of Chemotherapy, The District Hospital, Sucha Beskidzka, Poland
- Department of Chemotherapy, The Specialistic Hospital, Nowy Targ, Poland
| | - Anna Bidas
- Department of Clinical Oncology, Holy Cross Cancer Center, Kielce, Poland
| | - Jolanta Dobrzańska
- Department of Oncology, Jagiellonian University Medical College, Krakow, Poland
- Oncology Clinical Department, The University Hospital, Krakow, Poland
| | - Aleksandra Grela-Wojewoda
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Krakow Branch, Krakow, Poland
| | - Agnieszka Roman
- Department of Clinical Oncology, Ludwik Rydygier Hospital, Krakow, Poland
| | - Daria Tusień-Małecka
- Clinical and Experimental Oncology Clinic, Institute of Oncology, Karol Marcinkowski Medical University, Poznań, Poland
| | - Jerzy Walocha
- Department of Anatomy, Medical College, Jagiellonian University, Krakow, Poland
| | - Paweł Blecharz
- Department of Gynecological Oncology, Krakow Branch, Maria Sklodowska-Curie National Research Institute of Oncology, Garncarska 11 Street, Krakow, 31-115, Poland
| | - Mirosława Puskulluoglu
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Krakow Branch, Krakow, Poland
| |
Collapse
|
36
|
Chen L, Xing J, Lv J, Si S, Wang H, Yu W. Corynoxine suppresses lung adenocarcinoma proliferation and metastasis via inhibiting PI3K/AKT pathway and suppressing Cyclooxygenase-2 expression. Hereditas 2024; 161:41. [PMID: 39511658 PMCID: PMC11542349 DOI: 10.1186/s41065-024-00343-x] [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: 06/06/2024] [Accepted: 10/29/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is the most common lung cancer subtype, and the prognosis of affected patients is generally poor. The traditional Chinese medicine Uncaria rhychophaylla has been reported to exhibit anti-lung cancer properties. Accordingly, the main bioactive ingredient in Uncaria rhychophaylla, Corynoxine, may hold great value as a treatment for lung cancer. METHODS The impact of Corynoxine on the viability of LUAD cells was assessed using the Cell Counting Kit-8 (CCK-8) assay. Apoptosis in A549 cells was evaluated via flow cytometry. Migration and invasion capabilities were determined through wound healing and Transwell assays, respectively. The key pathways targeted by Corynoxine in LUAD were identified using a network pharmacology approach. Additionally, Western immunoblotting, quantitative real-time PCR (qRT-PCR), and ELISA assays were conducted to validate the underlying mechanisms. The in vivo anti-tumor efficacy of Corynoxine was assessed in xenograft nude mice. RESULTS In this study, Corynoxine treatment was found to markedly suppress in vitro LUAD cell proliferative, migratory, and invasive activity. It additionally downregulated Vimentin and promoted E-cadherin upregulation consistent with the disruption of epithelial-mesenchymal transition (EMT) induction while also accelerating apoptotic death. Furthermore, network pharmacology analysis revealed that the PI3K/AKT pathway is a potential target of Corynoxine in LUAD. In vitro assays demonstrated that treatment with Corynoxine resulted in the suppression of PI3K/AKT signaling and a consequent drop in cyclooxygenase-2 (COX-2) expression. These findings were further confirmed in vivo in mice harboring A549 tumor xenografts in which Corynoxine was able to interfere with the PI3K/AKT/COX-2 signaling axis. CONCLUSION This study elucidated the potential effects of Corynoxine in suppressing proliferation and metastasis in LUAD, along with investigating the underlying mechanisms. These data highlight the promise of Corynoxine as a novel therapeutic tool for the treatment of individuals diagnosed with LUAD.
Collapse
Affiliation(s)
- Liping Chen
- Department of Central Laboratory, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China.
- Department of Respiratory, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China.
| | - Jing Xing
- Department of Central Laboratory, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Jiapei Lv
- Department of Respiratory, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Sainv Si
- Department of Respiratory, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Huaying Wang
- Department of Respiratory, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Wanjun Yu
- Department of Respiratory, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China.
| |
Collapse
|
37
|
Antar RM, Fawaz C, Gonzalez D, Xu VE, Drouaud AP, Krastein J, Pio F, Murdock A, Youssef K, Sobol S, Whalen MJ. The Evolving Molecular Landscape and Actionable Alterations in Urologic Cancers. Curr Oncol 2024; 31:6909-6937. [PMID: 39590142 PMCID: PMC11593205 DOI: 10.3390/curroncol31110511] [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: 10/20/2024] [Revised: 11/03/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
The genetic landscape of urologic cancers has evolved with the identification of actionable mutations that impact diagnosis, prognosis, and therapeutic strategies. This narrative review consolidates existing literature on genetic mutations across key urologic cancers, including bladder, renal, prostate, upper tract urothelial, testicular, and penile. The review highlights mutations in DNA damage repair genes, such as BRCA1/2 and PTEN, as well as pathway alterations like FGFR and PD-L1 overexpression. These mutations influence tumor behavior and therapeutic outcomes, emphasizing the need for precision oncology approaches. Molecular profiling, through tools like next-generation sequencing, has revolutionized patient care by enabling targeted treatment strategies, especially in cancers with distinct molecular subtypes such as luminal or basal bladder cancer and clear cell renal carcinoma. Emerging therapies, including FGFR inhibitors and immune checkpoint blockade, offer new treatment avenues, although resistance mechanisms remain a challenge. We also emphasize the importance of biomarker identification for personalized management, especially in metastatic settings where treatment intensification is often required. Future research is needed to further elucidate our understanding of the genetics affecting urologic cancers, which will help develop novel, individualized therapies to enhance oncologic outcomes.
Collapse
Affiliation(s)
- Ryan Michael Antar
- Department of Urology, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA (M.J.W.)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Wu Y, Wu Z, Yao M, Liu L, Song Y, Ma L, Liu C. GABPA inhibits tumorigenesis in clear cell renal cell carcinoma by regulating ferroptosis through ACSL4. Sci Rep 2024; 14:26521. [PMID: 39489850 PMCID: PMC11532409 DOI: 10.1038/s41598-024-78441-z] [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: 03/11/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is a common genitourinary malignancy characterized by dysregulated cellular metabolism leading to aberrant glucose metabolism, fatty acid accumulation, and excessive reactive oxygen species production. ccRCC cells exhibit an augmented oxidative stress response. Complex interactions between iron metabolism and lipid homeostasis in ccRCC cells require a counteracting response that enables ferroptosis evasion and survival maintenance. Additionally, abnormal GA-binding protein transcription factor subunit alpha (GABPA) expression is associated with ccRCC occurrence and development, but its impact on ferroptosis-related molecular mechanisms remains unclear. Herein, we examined the impact of the GABPA-ACSL4 pathway on ferroptosis in ccRCC through bioinformatics analysis, as well as in vitro and in vivo experiments. In contrast to that in adjacent normal tissues, GABPA expression was significantly downregulated in ccRCC tissues, and this downregulation was linked to poor overall survival. Increased GABPA expression suppressed ccRCC cell proliferation, migration, and invasion. Moreover, GABPA overexpression increased the susceptibility of ccRCC cells to ferroptosis. Additionally, GABPA directly bound to the promoter region of ACSL4, promoting ferroptosis. Thus, inducing the GABPA-ACSL4 pathway activates ferroptosis, inhibits proliferation or metastasis, and exerts anticancer activity in ccRCC. These findings have important implications for regulating ccRCC occurrence and development.
Collapse
Affiliation(s)
- Yaqian Wu
- Department of Urology, Peking University Third Hospital, Beijing, 100191, People's Republic of China
- Department of Pathology, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, People's Republic of China
| | - Zonglong Wu
- Department of Urology, Peking University Third Hospital, Beijing, 100191, People's Republic of China
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Mengfei Yao
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China
| | - Li Liu
- School of Nursing, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Beijing, 100191, People's Republic of China
| | - Yimeng Song
- Department of Urology, Peking University Third Hospital, Beijing, 100191, People's Republic of China
| | - Lulin Ma
- Department of Urology, Peking University Third Hospital, Beijing, 100191, People's Republic of China.
| | - Cheng Liu
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China.
| |
Collapse
|
39
|
Tannir NM, Albigès L, McDermott DF, Burotto M, Choueiri TK, Hammers HJ, Barthélémy P, Plimack ER, Porta C, George S, Donskov F, Atkins MB, Gurney H, Kollmannsberger CK, Grimm MO, Barrios C, Tomita Y, Castellano D, Grünwald V, Rini BI, Jiang R, Desilva H, Fedorov V, Lee CW, Motzer RJ. Nivolumab plus ipilimumab versus sunitinib for first-line treatment of advanced renal cell carcinoma: extended 8-year follow-up results of efficacy and safety from the phase III CheckMate 214 trial. Ann Oncol 2024; 35:1026-1038. [PMID: 39098455 PMCID: PMC11907766 DOI: 10.1016/j.annonc.2024.07.727] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/10/2024] [Accepted: 07/25/2024] [Indexed: 08/06/2024] Open
Abstract
BACKGROUND Nivolumab plus ipilimumab (NIVO+IPI) has demonstrated superior overall survival (OS) and durable response benefits versus sunitinib (SUN) with long-term follow-up in patients with advanced renal cell carcinoma (aRCC). We report updated analyses with 8 years of median follow-up from CheckMate 214. PATIENTS AND METHODS Patients with aRCC (N = 1096) were randomized to NIVO 3 mg/kg plus IPI 1 mg/kg Q3W × four doses, followed by NIVO (3 mg/kg or 240 mg Q2W or 480 mg Q4W); or SUN (50 mg) once daily for 4 weeks on, 2 weeks off. The endpoints included OS, independent radiology review committee (IRRC)-assessed progression-free survival (PFS), and IRRC-assessed objective response rate (ORR) in intermediate/poor-risk (I/P; primary), intent-to-treat (ITT; secondary), and favorable-risk (FAV; exploratory) patients. RESULTS With 8 years (99.1 months) of median follow-up, the hazard ratio [HR; 95% confidence interval (CI)] for OS with NIVO+IPI versus SUN was 0.72 (0.62-0.83) in ITT patients, 0.69 (0.59-0.81) in I/P patients, and 0.82 (0.60-1.13) in FAV patients. PFS probabilities at 90 months were 22.8% versus 10.8% (ITT), 25.4% versus 8.5% (I/P), and 12.7% versus 17.0% (FAV), respectively. ORR with NIVO+IPI versus SUN was 39.5% versus 33.0% (ITT), 42.4% versus 27.5% (I/P), and 29.6% versus 51.6% (FAV). Rates of complete response were higher with NIVO+IPI versus SUN in all International Metastatic Renal Cell Carcinoma Database Consortium (IMDC) risk groups (ITT, 12.0% versus 3.5%; I/P, 11.8% versus 2.6%; FAV, 12.8% versus 6.5%). The median duration of response (95% CI) with NIVO+IPI versus SUN was 76.2 versus 25.1 months [59.1 months-not estimable (NE) versus 19.8-33.2 months] in ITT patients, 82.8 versus 19.8 months (54.1 months-NE versus 16.4-26.4 months) in I/P patients, and 61.5 versus 33.2 months (27.8 months-NE versus 24.8-51.4 months) in FAV patients. The incidence of treatment-related adverse events was consistent with previous reports. Exploratory post hoc analyses are reported for FAV patients, those receiving subsequent therapy based on their response status, clinical subpopulations, and adverse events over time. CONCLUSIONS Superior survival, durable response benefits, and a manageable safety profile were maintained with NIVO+IPI versus SUN at 8 years, the longest phase III follow-up for a first-line checkpoint inhibitor combination therapy in aRCC.
Collapse
Affiliation(s)
- N M Tannir
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, USA.
| | - L Albigès
- Department of Medical Oncology (Département de Médecine Oncologique), Gustave Roussy, Villejuif, France
| | - D F McDermott
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Dana-Farber/Harvard Cancer Center, Boston, USA
| | - M Burotto
- Department of Oncology, Bradford Hill Clinical Research Center, Santiago, Chile
| | - T K Choueiri
- Department of Medical Oncology, Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Boston; Harvard Medical School, Boston
| | - H J Hammers
- Department Internal Medicine, UT Southwestern Kidney Cancer Program, Dallas, USA
| | - P Barthélémy
- Department of Medical Oncology, Medical Oncology Unit, Institut de Cancérologie Strasbourg Europe, Strasbourg, France
| | - E R Plimack
- Department of Hematology and Oncology, Fox Chase Cancer Center, Philadelphia, USA
| | - C Porta
- Department of Internal Medicine, University of Pavia, Pavia, Italy
| | - S George
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, USA
| | - F Donskov
- Department of Oncology, Aarhus University Hospital, Aarhus; University Hospital of Southern Denmark, Esbjerg, Denmark
| | - M B Atkins
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Washington, DC, USA
| | - H Gurney
- Department of Medical Oncology, Westmead Hospital and Macquarie University, Sydney, Australia
| | - C K Kollmannsberger
- Department of Medical Oncology, British Columbia Cancer Agency, Vancouver, Canada
| | - M-O Grimm
- Department of Urology, Jena University Hospital and Comprehensive Cancer Center Central Germany (CCCG), Campus Jena, Jena, Germany
| | - C Barrios
- Department of Internal Medicine and Oncology Research Center, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Y Tomita
- Departments of Urology and Molecular Oncology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - D Castellano
- Medical Oncology Department, University Hospital 12 de Octubre, CIBER-ONC, Madrid, Spain
| | - V Grünwald
- Interdisciplinary Genitourinary Oncology, Clinic for Internal Medicine (Tumor Research) and Clinic for Urology, West-German Cancer Center Essen, University Hospital Essen, Essen, Germany
| | - B I Rini
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville
| | - R Jiang
- Global Biometrics and Data Sciences, Bristol Myers Squibb, Princeton
| | - H Desilva
- Late Clinical Development, Bristol Myers Squibb, Princeton
| | - V Fedorov
- Oncology Late Clinical Development, Bristol Myers Squibb, Princeton
| | - C-W Lee
- Department of Clinical Trials, Bristol Myers Squibb, Princeton
| | - R J Motzer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| |
Collapse
|
40
|
Bigot P, Boissier R, Khene ZE, Albigès L, Bernhard JC, Correas JM, De Vergie S, Doumerc N, Ferragu M, Ingels A, Margue G, Ouzaïd I, Pettenati C, Rioux-Leclercq N, Sargos P, Waeckel T, Barthelemy P, Rouprêt M. French AFU Cancer Committee Guidelines - Update 2024-2026: Management of kidney cancer. THE FRENCH JOURNAL OF UROLOGY 2024; 34:102735. [PMID: 39581661 DOI: 10.1016/j.fjurol.2024.102735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 09/01/2024] [Accepted: 09/02/2024] [Indexed: 11/26/2024]
Abstract
OBJECTIVE To update the French recommendations for the management of kidney cancer. METHODS A systematic review of the literature was conducted for the period from 2014 to 2024. The most relevant articles concerning the diagnosis, classification, surgical treatment, medical treatment, and follow-up of kidney cancer were selected and incorporated into the recommendations. The recommendations have been updated specifying the level of evidence (strong or weak). RESULTS Kidney cancer following prolonged occupational exposure to trichloroethylene should be considered an occupational disease. The reference examination for the diagnosis and staging of kidney cancer is the contrast-enhanced thoraco-abdominal CT scan. PET scans are not indicated in the staging of kidney cancer. Percutaneous biopsy is recommended in situations where its results will influence therapeutic decisions. It should be used to reduce the number of surgeries for benign tumors, particularly avoiding unnecessary radical nephrectomies. Kidney tumors should be classified according to the pTNM 2017 classification, the WHO 2022 classification, and the ISUP nucleolar grade. Metastatic kidney cancers should be classified according to IMDC criteria. Surveillance of tumors smaller than 2cm should be prioritized and can be offered regardless of patient age. Robot-assisted laparoscopic partial nephrectomy is the reference surgical treatment for T1 tumors. Ablative therapies and surveillance are options for elderly patients with comorbidities for tumors larger than 2cm. Stereotactic radiotherapy is an option to discuss for treating localized kidney tumors in patients not eligible for other treatments. Radical nephrectomy is the first-line treatment for locally advanced localized cancers. Pembrolizumab is recommended for patients at high risk of recurrence after surgery for localized kidney cancer. In metastatic patients, cytoreductive nephrectomy can be immediate in cases of good prognosis, delayed in cases of intermediate or poor prognosis for patients stabilized by medical treatment, or as "consolidation" in patients with complete or major partial response at metastatic sites after systemic treatment. Surgical or local treatment of metastases can be proposed for single lesions or oligometastases. Recommended first-line drugs for metastatic clear cell renal carcinoma are combinations of axitinib/pembrolizumab, nivolumab/ipilimumab, nivolumab/cabozantinib, and lenvatinib/pembrolizumab. Patients with non-clear cell metastatic kidney cancer should be presented to the CARARE Network and prioritized for inclusion in clinical trials. CONCLUSION These updated recommendations are a reference that will enable French and French-speaking practitioners to optimize their management of kidney cancer.
Collapse
Affiliation(s)
- Pierre Bigot
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology, Angers University Hospital, Angers, France.
| | - Romain Boissier
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology and Kidney Transplantation, Conception University Hospital, Aix-Marseille University, AP-HM, Marseille, France
| | - Zine-Eddine Khene
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology, Rennes University Hospital, Rennes, France
| | - Laurence Albigès
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Cancer Medicine, Gustave-Roussy, Paris-Saclay University, Villejuif, France
| | - Jean-Christophe Bernhard
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology, Hôpital Pellegrin, Bordeaux University Hospital, Bordeaux, France
| | - Jean-Michel Correas
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Adult Radiology, Hôpital Necker, University of Paris, AP-HP Centre, Paris, France
| | - Stéphane De Vergie
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology, Nantes University Hospital, Nantes, France
| | - Nicolas Doumerc
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology and Renal Transplantation, Toulouse University Hospital, Toulouse, France
| | - Matthieu Ferragu
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology, Angers University Hospital, Angers, France
| | - Alexandre Ingels
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology, UPEC, Hôpital Henri-Mondor, Créteil, France
| | - Gaëlle Margue
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology, Hôpital Pellegrin, Bordeaux University Hospital, Bordeaux, France
| | - Idir Ouzaïd
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology, Bichat University Hospital, AP-HP, Paris, France
| | - Caroline Pettenati
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology, Hôpital Foch, University of Versailles - Saint-Quentin-en-Yvelines, 40, rue Worth, 92150 Suresnes, France
| | - Nathalie Rioux-Leclercq
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Pathology, Rennes University Hospital, Rennes, France
| | - Paul Sargos
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Radiotherapy, Hôpital Pellegrin, Bordeaux University Hospital, Bordeaux, France
| | - Thibaut Waeckel
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology, Caen University Hospital, Caen, France
| | - Philippe Barthelemy
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Medical Oncology, Institut de Cancérologie Strasbourg Europe, Strasbourg, France
| | - Morgan Rouprêt
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Urology, Hôpital Pitié-Salpêtrière, Predictive Onco-Urology, GRC 5, Sorbonne University, AP-HP, 75013 Paris, France
| |
Collapse
|
41
|
Yanagisawa T, Mori K, Kawada T, Katayama S, Uchimoto T, Tsujino T, Nishimura K, Adachi T, Toyoda S, Nukaya T, Fukuokaya W, Urabe F, Murakami M, Yamanoi T, Bekku K, Komura K, Takahara K, Hashimoto T, Fujita K, Azuma H, Ohno Y, Shiroki R, Uemura H, Araki M, Kimura T. First-line therapy for metastatic renal cell carcinoma: A propensity score-matched comparison of efficacy and safety. Urol Oncol 2024; 42:374.e21-374.e29. [PMID: 39085019 DOI: 10.1016/j.urolonc.2024.06.013] [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: 03/12/2024] [Revised: 05/03/2024] [Accepted: 06/13/2024] [Indexed: 08/02/2024]
Abstract
PURPOSE Immune checkpoint inhibitor (ICI)-based combination therapy is a standard systemic treatment for metastatic renal cell carcinoma (mRCC). Although differential pharmacologic action between ICI+ICI and ICI+tyrosine kinase inhibitor (TKI) combinations may affect outcomes, comparative studies using real-world data are few. METHODS We retrospectively analyzed the records of 447 mRCC patients treated with 1st-line ICI-based combinations at multiple institutions between January 2018 and August 2023, and selected 320 patients diagnosed with clear cell RCC (ccRCC) for further study. Cohorts were matched using one-to-one propensity scores based on IMDC risk classification. Overall survival (OS), progression-free survival (PFS), objective response rates (ORRs), and treatment-related adverse events (TrAE) were compared. RESULTS The matching process yielded 228 metastatic ccRCC patients treated with ICI+ICI (n = 114) or ICI+TKI (n = 114). Median OS was 53 months (95%CI: 33-NA) in patients treated with ICI+ICI and was not reached (95%CI: 43-NA) with ICI+TKI (P = 0.24). Median PFS was significantly shorter for ICI+ICI (13 months, 95%CI: 7-25) than for ICI+TKI (25 months, 95%CI: 13-NA) (P = 0.047). There were no differences in second-line PFS for sequential therapy after 1st-line combinations of ICI+ICI or ICI+TKI (6 vs. 8 months, P = 0.6). There were no differences in ORR between the 2 groups (ICI+ICI: 51% vs. ICI+TKI: 55%, P = 0.8); the progressive disease (PD) rate was significantly higher in patients treated with the ICI+ICI combination (24% vs. 11%, P = 0.029). The rate of any grade TrAE was significantly higher in patients treated with ICI+TKI (71% vs. 85%, P = 0.016), but we found no differences in severe TrAE between the 2 groups (39% vs. 36%, P = 0.8). CONCLUSIONS In a matched cohort of real-world data, we confirmed comparable OS benefits between ICI+ICI and ICI+TKI combinations. However, differential clinical behaviors in terms of PFS, PD rates, and TrAE between ICI-based combinations may enrich clinical decision-making.
Collapse
Affiliation(s)
- Takafumi Yanagisawa
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan.
| | - Keiichiro Mori
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Tatsushi Kawada
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Satoshi Katayama
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Taizo Uchimoto
- Department of Urology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Takuya Tsujino
- Department of Urology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Kazuki Nishimura
- Department of Urology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Takahiro Adachi
- Department of Urology,Tokyo Medical University, Tokyo, Japan
| | - Shingo Toyoda
- Department of Urology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Takuhisa Nukaya
- Department of Urology, Fujita Health University School of Medicine, Aichi, Japan
| | - Wataru Fukuokaya
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Fumihiko Urabe
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Masaya Murakami
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Tomoaki Yamanoi
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kensuke Bekku
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kazumasa Komura
- Department of Urology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Kiyoshi Takahara
- Department of Urology, Fujita Health University School of Medicine, Aichi, Japan
| | | | - Kazutoshi Fujita
- Department of Urology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Haruhito Azuma
- Department of Urology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Yoshio Ohno
- Department of Urology,Tokyo Medical University, Tokyo, Japan
| | - Ryoichi Shiroki
- Department of Urology, Fujita Health University School of Medicine, Aichi, Japan
| | - Hirotsugu Uemura
- Department of Urology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Motoo Araki
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takahiro Kimura
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
42
|
Nayak RK, Aiello M, Maldonado LM, Clark TY, Buchwald ZS, Chang A. Impact of race, ethnicity, and social determinants on outcomes following immune checkpoint therapy. J Immunother Cancer 2024; 12:e010116. [PMID: 39461882 PMCID: PMC11529590 DOI: 10.1136/jitc-2024-010116] [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: 07/16/2024] [Accepted: 10/03/2024] [Indexed: 10/29/2024] Open
Abstract
Immune checkpoint blockade (ICB) therapies are one of the greatest advances in the history of cancer care and are now commonly used in the management of many different malignancies. However, much remains unknown about the factors that affect the efficacy and side effect profile of these agents. This review delves into the published literature that evaluates the intricate interplay between race, age, gender, and social determinants in shaping outcomes following ICB across solid tumors and hematologic malignancies. We examine the pivotal phase 2 and 3 trials to evaluate the demographics of participants and outcomes based on these variables, if reported. Most, but not all, trials reported some basic demographic information like age, sex, race, ethnicity, and/or geographic area for enrollment. Clinically relevant biological markers that could affect ICB outcomes such as obesity or markers of social determinants of health were largely not reported. Trials were generally representative for men and women based on expected prevalence for a given malignancy, but often under-represented non-white participants and rarely enrolled patients from the global south. Subgroup analyses were conducted in many ICB trials for solid malignancies, but rarely conducted for hematologic malignancies. These analyses largely showed similar qualitative benefit across subgroups, but adverse events were rarely reported by subgroup. This review adds to our understanding of the populations that these clinical trials have studied and highlight the urgent need to redouble our efforts at increasing the diversity of the population in future ICB trials.
Collapse
Affiliation(s)
- Rahul K Nayak
- Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia, USA
| | - Meili Aiello
- Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia, USA
| | | | - Tarralyn Y Clark
- Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia, USA
| | - Zachary S Buchwald
- Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia, USA
| | - Andres Chang
- Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia, USA
| |
Collapse
|
43
|
Chang JY, Xu X, Shroff GS, Comeaux NI, Li W, Rodon Ahnert J, Karp DD, Dumbrava EE, Verma V, Chen A, Welsh J, Hong DS. Phase I/II study of BMS-986156 with ipilimumab or nivolumab with or without stereotactic ablative radiotherapy in patients with advanced solid malignancies. J Immunother Cancer 2024; 12:e009975. [PMID: 39384194 PMCID: PMC11474930 DOI: 10.1136/jitc-2024-009975] [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: 07/01/2024] [Accepted: 09/16/2024] [Indexed: 10/11/2024] Open
Abstract
BACKGROUND BMS-986156 is an agonist of the glucocorticoid-induced tumor necrosis factor receptor (TNFR)-related protein (GITR) and promotes increased effector T-cell activation. Combined anti-GITR, anti-programmed death-1, anti-cytotoxic T-lymphocyte-associated protein 4 antibodies and radiotherapy improve tumor control in preclinical studies. Herein we describe the results of the safety and efficacy of BMS-986156+ipilimumab or nivolumab with/without stereotactic ablative radiotherapy (SABR) in patients with advanced solid cancers (NCT04021043). METHODS This open-label, multigroup, single-center phase I/II study enrolled patients with histologically-confirmed stage IV solid cancers resistant to standard treatments. Group 1 (G1, n=20) received four cycles of ipilimumab (3 mg/kg) plus BMS-986156 (30 mg as dose level 1 (L1) or 100 mg as dose level 2 (L2)), every 3 weeks (Q3W). Group 2 (G2, n=10) received four cycles of ipilimumab (3 mg/kg) plus BMS-986156 (dose as determined in G1, Q3W) with SABR (50 Gy/4 fx or 60-70 Gy/10 fx to liver/lung lesions. Group 3 (G3, n=20) received four cycles of nivolumab (480 mg) plus BMS-986156 (30 mg), every 4 weeks with SABR. Maintenance nivolumab could be given up to 2 years. Tumor responses were assessed every 1-3 months until progression, using immune-related response criteria. RESULTS 50 patients were enrolled between 10/2019 and 12/2021. Patients received a median of 3 (IQR 2-4.25) initial treatment cycles. 100 mg BMS-986156 with ipilimumab was tolerated well. Five discontinued BMS-986156 with ipilimumab due to treatment-related adverse events (TRAEs), with three in G1/L1, one in G1/L2 and one in G2, respectively. 22 patients (44%) experienced Grade 1-3 TRAEs (6, 4, 5, 7 patients for G1/L1, G1/L2, G2, G3). Six (12%) had Grade 3 TRAEs (2, 2, 1, 1 for G1/L1, G1/L2, G2, G3), with elevated alanine aminotransferase (n=3, in G1/L2, G2 and G3) and aspartate aminotransferase (n=2, in G2 and G3) being the most common. There was no Grade 4-5 TRAEs. Overall, 19/39 (48.7%) patients eligible for efficacy analysis had stable disease and 3 (7.7%) achieved a partial response. Out-of-field (abscopal) disease control rate (ACR) and out-of-field (abscopal) response rate (ARR) were 38.5% and 7.7%, respectively, with the highest ACR (50%, 9/18) and ARR (11.1%, 2/18) in G3. CONCLUSIONS BMS-986156 was well-tolerated with ipilimumab, nivolumab, with or without SABR. Outcomes were encouraging in this population, as more than half of patients had stable disease/partial response.
Collapse
Affiliation(s)
- Joe Y Chang
- Department of Thoracic Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Xinyan Xu
- Department of Thoracic Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Girish S Shroff
- Department of Thoracic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nathan I Comeaux
- Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Wei Li
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jordi Rodon Ahnert
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Daniel D Karp
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ecaterina E Dumbrava
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Vivek Verma
- Department of Thoracic Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Aileen Chen
- Department of Thoracic Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - James Welsh
- Department of Thoracic Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - David S Hong
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
44
|
Zhang S, Zhang X, Xiahou Z, Zuo S, Xue J, Zhang Y. Unraveling the ecological landscape of mast cells in esophageal cancer through single-cell RNA sequencing. Front Immunol 2024; 15:1470449. [PMID: 39430754 PMCID: PMC11486721 DOI: 10.3389/fimmu.2024.1470449] [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: 07/25/2024] [Accepted: 09/13/2024] [Indexed: 10/22/2024] Open
Abstract
Background Esophageal cancer (EC) is a major health issue, ranking seventh in incidence and sixth in mortality worldwide. Despite advancements in multidisciplinary treatment approaches, the 5-year survival rate for EC remains low at 21%. Challenges in EC treatment arise from late-stage diagnosis, high malignancy, and poor prognosis. Understanding the tumor microenvironment is critical, as it includes various cellular and extracellular components that influence tumor behavior and treatment response. Mast cells (MCs), as tissue-resident immune cells, play dual roles in tumor dynamics. High-throughput single-cell RNA sequencing offers a powerful tool for analyzing tumor heterogeneity and immune interactions, although its application in EC is limited. Methods In this study, we investigated the immune microenvironment of EC using single-cell RNA sequencing and established a comprehensive immune profile. We also performed analysis of upstream transcription factors and downstream pathway enrichment to further comprehensively decipher MCs in EC. Besides, we performed knockdown experiments to explore the role of epidermal growth factor receptor (EGFR) signaling pathway in MCs-tumor cell interactions, highlighting its potential as a prognostic marker. Finally, we constructed a prognostic model for EC, which provided valuable suggestions for the diagnosis and prognosis of EC. Results Our analysis identified 11 major cell types, of which MCs were particularly present in pericarcinoma tissues. Further grouping of the 5,001 MCs identified 8 distinct subtypes, including SRSF7-highly expressed MCs, which showed strong tumor preference and potential tumor-promoting properties. Moreover, we identified the key signaling receptor EGFR and validated it by in vitro knockdown experiments, demonstrating its cancer-promoting effects. In addition, we established an independent prognostic indicator, SRSF7+ MCs risk score (SMRS), which showed a correlation between high SMRS group and poor prognosis. Conclusion These findings illuminate the complex interactions within the tumor microenvironment of EC and suggest that targeting specific MCs subtypes, particularly via the EGFR signaling pathway, may present novel therapeutic strategies. This study establishes a comprehensive immune map of EC, offering insights for improved treatment approaches.
Collapse
Affiliation(s)
- Shengyi Zhang
- Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinyi Zhang
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Zhikai Xiahou
- China Institute of Sport and Health Science, Beijing Sport University, Beijing, China
| | - Shunqing Zuo
- Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jialong Xue
- Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Zhang
- Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
45
|
Osawa T, Sasaki K, Machida R, Matsumoto T, Matsui Y, Kitamura H, Nishiyama H. Real-world treatment trends for patients with advanced prostate cancer and renal cell carcinoma and their cost-a survey in Japan. Jpn J Clin Oncol 2024; 54:1062-1070. [PMID: 38843876 DOI: 10.1093/jjco/hyae045] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 03/28/2024] [Indexed: 10/08/2024] Open
Abstract
BACKGROUND Advanced (Stage IV) prostate and renal cancer have poor prognosis, and several therapies have been developed, but many are very costly. This study investigated drug regimens used in patients with untreated Stage IV prostate cancer and renal cell carcinoma and calculated the monthly cost of each. METHODS We surveyed first-line drugs administered to patients with untreated Stage IV prostate cancer and renal cancer at Japan Clinical Oncology Group affiliated centers from April 2022 to March 2023. Drug costs were calculated according to drug prices in September 2023. Individual drug costs were calculated or converted to 28-day costs. RESULTS A total of 700 patients with untreated Stage IV prostate cancer were surveyed. Androgen deprivation therapy + androgen receptor signaling inhibitor was the most common regimen (56%). The cost of androgen deprivation therapy + androgen receptor signaling inhibitor was 10.6-30.8-fold compared with conventional treatments. A total of 137 patients with Stage IV renal cancer were surveyed. Among them, 91% of patients received immune-oncology drug-based regimen. All patients received treatments with a monthly cost of ≥500 000 Japanese yen, and 80.4% of patients received treatments with a monthly cost of ≥1 million Japanese yen, of combination treatments. The cost of immune-oncology drug-based regimen was 1.2-3.1-fold that of TKI alone. CONCLUSION To the best of our knowledge, this is the first report of a survey of first-line drug therapy in untreated Stage IV prostate cancer and renal cell carcinoma stratified by age and treatment costs. Our results show that most Japanese patients received state-of-the-art, effective treatments with high financial burden.
Collapse
Affiliation(s)
- Takahiro Osawa
- Department of Urology, Hokkaido University Hospital, Sapporo, Japan
| | - Keita Sasaki
- Japan Clinical Oncology Group Data Center/Operations Office, National Cancer Center Hospital, Tokyo, Japan
| | - Ryunosuke Machida
- Japan Clinical Oncology Group Data Center/Operations Office, National Cancer Center Hospital, Tokyo, Japan
| | - Takashi Matsumoto
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshiyuki Matsui
- Department of Urology, National Cancer Center Hospital, Tokyo, Japan
| | - Hiroshi Kitamura
- Department of Urology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Hiroyuki Nishiyama
- Department of Urology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
46
|
Cerrato C, Crocerossa F, Marchioni M, Giannarini G, Gupta S, Albiges L, Brouwer O, Albersen M, Fankhauser C, Grimm MO, Gandaglia G, Roupret M, Mir MC. Effect of Sex on the Oncological Outcomes in Response to Immunotherapy and Antibody-drug Conjugates in Patients with Urothelial and Kidney Cancer: A Systematic Review and a Network Meta-analysis. Eur Urol Oncol 2024; 7:1005-1014. [PMID: 38644155 DOI: 10.1016/j.euo.2024.03.014] [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: 01/28/2024] [Revised: 03/03/2024] [Accepted: 03/26/2024] [Indexed: 04/23/2024]
Abstract
BACKGROUND AND OBJECTIVE Immune checkpoint inhibitors (ICIs) and antibody-drug conjugates (ADCs) herald a transformative era in metastatic renal cell carcinoma (RCC) and transitional cell carcinoma (TCC) treatment, amid acknowledged sex-based disparities in these cancers. We conducted a systematic review and network meta-analysis (NMA) to identify sex-specific differences in the efficacy of ICI/ADC monotherapy or combination therapies for RCC and TCC survival, in metastatic and adjuvant settings. METHODS A systematic search was conducted up to October 2023 for English articles on ICIs and ADCs as systemic therapies (ICIs in first-line and adjuvant treatment for RCC, ICIs and ADCs in first- and second-line treatment for TCC). Randomised clinical trials were considered. The primary objective was overall survival (OS) of ICIs and ADCs between males and females. The secondary outcomes included progression-free survival, overall response rate, disease-free survival, and recurrence-free survival. Treatment efficacy was evaluated by sex via odds ratios (ORs) and confidence intervals compared with controls. Log ORs were used for creating a frequentist NMA. This meta-analysis was registered on PROSPERO (CRD42023468632). KEY FINDINGS AND LIMITATIONS Eighteen articles met the inclusion criteria. Females had an advantage for RCC-adjuvant treatment for atezolizumab (log OR [SE] = -0.57 ± 0.25, p = 0.024) in OS. Males showed a survival advantage in TCC second-line treatment for ADC-Nectin 4 (log OR [SE] = 0.65 ± 0.28, p = 0.02). No other significant results were shown. CONCLUSIONS AND CLINICAL IMPLICATIONS The NMA revealed gender-specific variations in ICI and ADC responses for RCC and TCC, offering insights for personalised cancer care and addressing disparities in cancer care and outcomes. PATIENT SUMMARY In this systematic review, we looked at the sex differences for metastatic renal cell carcinoma (RCC) and transitional cell carcinoma (TCC) for antibody-drug conjugates and immune checkpoint inhibitors. In our analysis, female and male sex has better overall survival for adjuvant and second-line therapies for RCC and TCC, respectively. Urgent research on gender-specific cancer therapies is imperative.
Collapse
Affiliation(s)
- Clara Cerrato
- University Hospital Southampton NHS Trust, Southampton, UK
| | - Fabio Crocerossa
- Department of Urology, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | | | - Gianluca Giannarini
- Urology Unit, "Santa Maria della Misericordia" University Hospital, Udine, Italy
| | - Shilpa Gupta
- Cleveland Clinic Taussig Cancer Institute, Cleveland, OH, USA
| | - Laurence Albiges
- Department of Cancer Medicine, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Oscar Brouwer
- The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Maarten Albersen
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | - Christian Fankhauser
- Department of Urology, Luzerner Kantonsspital, Luzern, Switzerland; University of Lucerne, Lucerne, Switzerland; University of Zurich, Zurich, Switzerland
| | | | - Giorgio Gandaglia
- Department of Urology, Division of Experimental Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Morgan Roupret
- GRC 5 Predictive Onco-Uro, Department of Urology, AP-HP, Pitié Salpétrière Hospital, Sorbonne University, Paris, France
| | - Maria Carmen Mir
- Department of Urology, Hospital Universitario La Ribera, Valencia, Spain.
| |
Collapse
|
47
|
Moinard-Butot F, Oriel M, Tricard T, Cazzato RL, Pierard L, Gaillard V, Werle P, Lindner V, Martin S, Schuster C, Roy C, Burgy M, Anthony A, Bigot C, Boudier P, Fritsch A, Olland A, Malouf G, Lang H, Barthélémy P. Effect of Treatment of Residual Disease After Immunotherapy-Based Combinations on Complete Response Rate of Patients With Metastatic Renal Cell Carcinomas. Clin Genitourin Cancer 2024; 22:102134. [PMID: 38909529 DOI: 10.1016/j.clgc.2024.102134] [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: 02/02/2024] [Revised: 05/27/2024] [Accepted: 05/30/2024] [Indexed: 06/25/2024]
Abstract
INTRODUCTION Immune checkpoint inhibitor (ICI)-based combinations have revolutionized the management of first-line metastatic renal cell carcinoma (mRCC) by improving patient survival. Large phase 3 randomized trials assessing ICI-based combinations have reported complete response (CR) rates of 10% to 18% in the first-line setting. However, there is a scarcity of data about the effect of treatment of residual disease regarding CR rates improvement. MATERIALS AND METHODS We included retrospectively all consecutive mRCC patients treated in first-line setting at the Institut de Cancérologie Strasbourg Europe with an ICI-based combination involving ICI or TKI, either alone or with added local treatment of residual disease. Patients were characterized according to IMDC risk. Radiologic response was defined according to RECIST v1.1. RESULTS We enrolled 80 mRCC patients treated with ICI-based combinations between May 2015 and May 2022. The median age was 63 years. Regarding IMDC risk, there were 12 favourable (15%), 50 intermediate (63%), and 18 poor-risk (22%) patients. Forty-seven patients (59%) received ICI + ICI, 24 (30%) received ICI + TKI, and 9 (11%) received another ICI-based therapy. In total, 8 achieved CR (10%), 36 patients (45%) achieved partial response, 23 (29%) achieved stable disease and 12 achieved progressive disease (15%) as the best response with systemic therapy alone. By adding local treatment of residual disease, 11 additional patients (14%) achieved radiological NED. Residual disease resected sites included kidney (n = 6), lymph nodes (n = 5), lung metastases (n = 2) and liver metastases (n = 1). CONCLUSIONS The resection of residual disease after first-line ICI-based therapy is associated with improved CR rate (CR + NED) in patients with mRCC. These results need to be validated in prospective trial. PATIENT SUMMARY In recent years, the advent of immunotherapy has radically changed the management of patients with metastatic kidney cancer. Approximately 10% to 18% of these patients using immune checkpoint inhibitor (ICI)-based combinations no longer have detectable disease on CT scans (complete response). There are currently few data on the use of treatment of residual disease to increase the number of patients in complete response. In this retrospective study, the complete response rate with ICI-based treatment was 10%. When local treatment was added, the number of patients with a complete response increased to 24%. This strategy could increase the number of patients with a prolonged complete response in the future.
Collapse
Affiliation(s)
- F Moinard-Butot
- Department of medical oncology, Institut de Cancérologie Strasbourg Europe, Strasbourg, France.
| | - M Oriel
- Department of medical oncology, Institut de Cancérologie Strasbourg Europe, Strasbourg, France
| | - T Tricard
- Department of Urology, Strasbourg University Hospital, Strasbourg, France
| | - R L Cazzato
- Department of Interventional Radiology, Nouvel Hôpital Civil, Hopitaux Universitaires de Strasbourg, Strasbourg, France
| | - L Pierard
- Department of medical oncology, Institut de Cancérologie Strasbourg Europe, Strasbourg, France
| | - V Gaillard
- Department of Urology, Strasbourg University Hospital, Strasbourg, France
| | - P Werle
- Department of Urology, Strasbourg University Hospital, Strasbourg, France
| | - V Lindner
- Department of Pathology, Hôpitaux Universitaires de Strasbourg, Hôpital de Strasbourg-Hautepierre, Strasbourg, France
| | - S Martin
- Department of medical oncology, Institut de Cancérologie Strasbourg Europe, Strasbourg, France
| | - C Schuster
- Department of medical oncology, Institut de Cancérologie Strasbourg Europe, Strasbourg, France
| | - C Roy
- Department of Radiology, Strasbourg University Hospital-Nouvel Hopital Civil, Strasbourg, Cedex, France
| | - M Burgy
- Department of medical oncology, Institut de Cancérologie Strasbourg Europe, Strasbourg, France
| | - A Anthony
- Department of medical oncology, Institut de Cancérologie Strasbourg Europe, Strasbourg, France
| | - C Bigot
- Department of medical oncology, Institut de Cancérologie Strasbourg Europe, Strasbourg, France
| | - P Boudier
- Department of medical oncology, Institut de Cancérologie Strasbourg Europe, Strasbourg, France
| | - A Fritsch
- Department of medical oncology, Institut de Cancérologie Strasbourg Europe, Strasbourg, France
| | - A Olland
- Department of Thoracic Surgery, Strasbourg University Hospital-Nouvel Hopital Civil, Strasbourg, Cedex, France; Inserm 1260 "RNM", fédération de médecine translationnelle, Université de Strasbourg, Strasbourg, Cedex, France
| | - G Malouf
- Department of medical oncology, Institut de Cancérologie Strasbourg Europe, Strasbourg, France
| | - H Lang
- Department of Urology, Strasbourg University Hospital, Strasbourg, France
| | - P Barthélémy
- Department of medical oncology, Institut de Cancérologie Strasbourg Europe, Strasbourg, France
| |
Collapse
|
48
|
Bruckmann M, Brenet E, Boulagnon-Rombi C, Louvrier A, Mauprivez C. Effectiveness of immune checkpoint inhibitors in the treatment of kidney cancer oral metastasis. A case report. JOURNAL OF STOMATOLOGY, ORAL AND MAXILLOFACIAL SURGERY 2024; 125:101913. [PMID: 38719194 DOI: 10.1016/j.jormas.2024.101913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 05/06/2024] [Indexed: 05/21/2024]
Abstract
Oral cavity metastasis from renal cell carcinoma is rare with poor prognosis. Recently immune checkpoint inhibitors (ICIs) have shown promising outcomes in the treatment of advanced RCC. Herein, we report a case of palatal lesion mimicking vascular tumor in a 60-year-old woman with locally advanced kidney cancer (T3aN0M0). She underwent an excisional biopsy, and histopathological examinations revealed an oral metastasis from clear cell renal cell carcinoma (ccRCC). The patient was treated with a combination of two ICIs with nivolumab, a programmed death 1 (PD-1), and ipilimumab, a cytotoxic T-lymphocyte-associated antigen 4 (CTLA4). After 3 cycles of systemic immunotherapy, the palate was completely well healed and after 13 months of follow-up, there was no evidence of recurrence. Regarding treatments, radical surgery is often recommended due to a high local control in case of solitary lesions or oligo-metastases. However, this option therapy is associated with a poor quality of life. To the best of our knowledge, this is the first case to suggest the benefits of ICIs in the treatment of oral metastases from ccRCC. Combining ICIs with conservative surgery could be another treatment option for oral metastasis in patients with renal cell carcinoma.
Collapse
Affiliation(s)
- M Bruckmann
- Service d'Oto-Rhino-Laryngologie, CHU de Reims, F-51100 Reims, France
| | - E Brenet
- Service d'Oto-Rhino-Laryngologie, CHU de Reims, Université de Reims Champagne-Ardenne, Laboratoire EA4691 (BIOS), F-51100 Reims, France
| | - C Boulagnon-Rombi
- Université de Reims Champagne-Ardenne, UMR CNRS 7369, CHU Reims, Laboratoire de Pathologie, F-51100, Reims, France
| | - A Louvrier
- CHU Besançon, Chirurgie maxillo-faciale, stomatologie et odontologie hospitalière, Université de Franche-Comté, SINERGIES, F-25000 Besançon, France.
| | - C Mauprivez
- Service de Chirurgie Orale, Pôle de Médecine Bucco-Dentaire, CHU de Reims, Université de Reims Champagne-Ardenne, Laboratoire EA4691 (BIOS), F-51100 Reims, France
| |
Collapse
|
49
|
Chai YM, Zhou ZB, Liu RZ, Cui YS, Zhang Y. SNX4 Is Correlated With Immune Infiltration and Prognosis in Clear Cell Renal Cell Carcinoma. World J Oncol 2024; 15:809-824. [PMID: 39328330 PMCID: PMC11424112 DOI: 10.14740/wjon1868] [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: 04/06/2024] [Accepted: 06/18/2024] [Indexed: 09/28/2024] Open
Abstract
Background Clear cell renal cell carcinoma (ccRCC) is known as the most common and malignant histologic subtype of renal carcinoma. Sorting nexin 4 (SNX4) plays a regulatory role in recycling from endosomes to the plasma membrane and promotes autophagosome assembly and transport, which may exert the cancerous growth and progression. This study aimed to assess the biological role of SNX4 in ccRCC and their clinical association via public biological data platforms combined with experimental verification. Methods In our study, we analyzed the mRNA and protein expression of SNX4 in ccRCC under different clinicopathological characteristics through The Cancer Genome Atlas (TCGA), Human Protein Atlas (HPA) and Clinical Proteomic Tumor Analysis Consortium (CPTAC) databases. We used the Gene Expression Profiling Interactive Analysis (GEPIA) platform to conduct the survival analysis and figure out the immune cell infiltration level under different expression levels of SNX4 combined with Tumor Immune Estimation Resource (TIMER) database. Furthermore, we predicted competing endogenous RNA (ceRNA) regulatory network using TargetScan, miRDB, starBase and miRTarBase online databases. We totally collected six paired ccRCC tissues and adjacent tissues and applied quantitative real-time polymerase chain reaction (qRT-PCR) and western blot (WB) to detect the expression of SNX4 in the collected clinical specimens. Results The mRNA and protein expression level of SNX4 was significantly lower in ccRCC than those in normal tissues. The results proposed that lower SNX4 was expressed in patients with higher histologic grade and in male patients. Kaplan-Meier analysis demonstrated that lower mRNA expression level of SNX4 was correlated with poorer prognosis. SNX4 had positive correlation with immune cell infiltrating levels and programmed cell death-ligand 1 (PD-L1) expression. Furthermore, we constructed the SNX4/miR-221-3p/miR-222-3p/DHRS4-AS1 axis, which may be the underlying ceRNA interaction network. Finally, we verified the reduced expression of SNX4 in ccRCC by qRT-PCR and WB. Conclusion The expression of SNX4 in ccRCC was lower than adjacent tissues and its downregulated expression was associated with poor prognosis of ccRCC patients. SNX4 may exert critical roles in the tumorigenesis, development and migration of ccRCC via various mechanisms.
Collapse
Affiliation(s)
- Yu Meng Chai
- Department of Urology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- These authors contributed equally to this article
| | - Zhong Bao Zhou
- Department of Urology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- These authors contributed equally to this article
| | - Run Ze Liu
- Department of Urology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Yuan Shan Cui
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Yong Zhang
- Department of Urology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| |
Collapse
|
50
|
Xiong B, Liu W, Liu Y, Chen T, Lin A, Song J, Qu L, Luo P, Jiang A, Wang L. A Multi-Omics Prognostic Model Capturing Tumor Stemness and the Immune Microenvironment in Clear Cell Renal Cell Carcinoma. Biomedicines 2024; 12:2171. [PMID: 39457484 PMCID: PMC11504857 DOI: 10.3390/biomedicines12102171] [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: 08/06/2024] [Revised: 09/11/2024] [Accepted: 09/20/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Cancer stem-like cells (CSCs), a distinct subset recognized for their stem cell-like abilities, are intimately linked to the resistance to radiotherapy, metastatic behaviors, and self-renewal capacities in tumors. Despite their relevance, the definitive traits and importance of CSCs in the realm of oncology are still not fully comprehended, particularly in the context of clear cell renal cell carcinoma (ccRCC). A comprehensive understanding of these CSCs' properties in relation to stemness, and their impact on the efficacy of treatment and resistance to medication, is of paramount importance. Methods: In a meticulous research effort, we have identified new molecular categories designated as CRCS1 and CRCS2 through the application of an unsupervised clustering algorithm. The analysis of these subtypes included a comprehensive examination of the tumor immune environment, patterns of metabolic activity, progression of the disease, and its response to immunotherapy. In addition, we have delved into understanding these subtypes' distinctive clinical presentations, the landscape of their genomic alterations, and the likelihood of their response to various pharmacological interventions. Proceeding from these insights, prognostic models were developed that could potentially forecast the outcomes for patients with ccRCC, as well as inform strategies for the surveillance of recurrence after treatment and the handling of drug-resistant scenarios. Results: Compared with CRCS1, CRCS2 patients had a lower clinical stage/grading and a better prognosis. The CRCS2 subtype was in a hypoxic state and was characterized by suppression and exclusion of immune function, which was sensitive to gefitinib, erlotinib, and saracatinib. The constructed prognostic risk model performed well in both training and validation cohorts, helping to identify patients who may benefit from specific treatments or who are at risk of recurrence and drug resistance. A novel therapeutic target, SAA2, regulating neutrophil and fibroblast infiltration, and, thus promoting ccRCC progression, was identified. Conclusions: Our findings highlight the key role of CSCs in shaping the ccRCC tumor microenvironment, crucial for therapy research and clinical guidance. Recognizing tumor stemness helps to predict treatment efficacy, recurrence, and drug resistance, informing treatment strategies and enhancing ccRCC patient outcomes.
Collapse
Affiliation(s)
- Beibei Xiong
- Department of Oncology, The First People’s Hospital of Shuangliu District, Chengdu 610200, China;
| | - Wenqiang Liu
- Department of Urology, Changhai Hospital, Navel Medical University (Second Military Medical University), Shanghai 200433, China; (W.L.); (Y.L.); (T.C.); (J.S.)
| | - Ying Liu
- Department of Urology, Changhai Hospital, Navel Medical University (Second Military Medical University), Shanghai 200433, China; (W.L.); (Y.L.); (T.C.); (J.S.)
| | - Tong Chen
- Department of Urology, Changhai Hospital, Navel Medical University (Second Military Medical University), Shanghai 200433, China; (W.L.); (Y.L.); (T.C.); (J.S.)
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; (A.L.); (P.L.)
| | - Jiaao Song
- Department of Urology, Changhai Hospital, Navel Medical University (Second Military Medical University), Shanghai 200433, China; (W.L.); (Y.L.); (T.C.); (J.S.)
| | - Le Qu
- Department of Urology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China;
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; (A.L.); (P.L.)
| | - Aimin Jiang
- Department of Urology, Changhai Hospital, Navel Medical University (Second Military Medical University), Shanghai 200433, China; (W.L.); (Y.L.); (T.C.); (J.S.)
| | - Linhui Wang
- Department of Urology, Changhai Hospital, Navel Medical University (Second Military Medical University), Shanghai 200433, China; (W.L.); (Y.L.); (T.C.); (J.S.)
| |
Collapse
|