1
|
Tang H, Li YX, Lian JJ, Ng HY, Wang SSY. Personalized treatment using predictive biomarkers in solid organ malignancies: A review. TUMORI JOURNAL 2024; 110:386-404. [PMID: 39091157 DOI: 10.1177/03008916241261484] [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: 08/04/2024]
Abstract
In recent years, the influence of specific biomarkers in the diagnosis and prognosis of solid organ malignancies has been increasingly prominent. The relevance of the use of predictive biomarkers, which predict cancer response to specific forms of treatment provided, is playing a more significant role than ever before, as it affects diagnosis and initiation of treatment, monitoring for efficacy and side effects of treatment, and adjustment in treatment regimen in the long term. In the current review, we explored the use of predictive biomarkers in the treatment of solid organ malignancies, including common cancers such as colorectal cancer, breast cancer, lung cancer, prostate cancer, and cancers associated with high mortalities, such as pancreatic cancer, liver cancer, kidney cancer and cancers of the central nervous system. We additionally analyzed the goals and types of personalized treatment using predictive biomarkers, and the management of various types of solid organ malignancies using predictive biomarkers and their relative efficacies so far in the clinical settings.
Collapse
|
2
|
Omri L, Naigeon M, Flippot R, Gavira-Díaz J, Poveda-Ferriols J, Nguyen D, Abdi C, Arroyo-Salgado A, Chaput N, de Velasco G, Albigès L, Carril-Ajuria L. Blood-based circulating biomarkers for prediction of immune-checkpoint inhibitors efficacy in renal cell carcinoma. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:1199-1222. [PMID: 39465007 PMCID: PMC11502076 DOI: 10.37349/etat.2024.00271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/09/2024] [Indexed: 10/29/2024] Open
Abstract
Immune checkpoint inhibitors (ICI)-based combinations have become the standard first-line treatment for advanced clear cell renal cell carcinoma (ccRCC). Despite significant improvements in survival and the achievement of sustained long-term responses, a subset of patients remains refractory to ICI, and most will eventually develop resistance. Thus, identifying predictive biomarkers for ICI efficacy and resistance is essential for optimizing therapeutic strategies. Up to now, tissue-based biomarkers have not been successful as predictive biomarkers in RCC. Circulating blood-based biomarkers offer a promising alternative. These biomarkers, including circulating immune cells, soluble factors, tumor-derived markers, and those based on metabolomics, are less invasive, offer reproducibility over time, and provide a comprehensive assessment of tumor biology and patient immune status, as well as allow dynamic monitoring during treatment. This review aims to evaluate the current evidence on the different candidate circulating biomarkers being investigated for their potential to predict ICI efficacy in RCC patients.
Collapse
Affiliation(s)
- Loubna Omri
- Department of Medical Oncology, National Institute of Oncology, Rabat X4FH+66, Morocco
- Medical Oncology Department, Centre Hospitalier Universitaire Brugmann, 1020 Brussels, Belgium
| | - Marie Naigeon
- Laboratory of Immunomonitoring in Oncology, Gustave Roussy, 94805 Villejuif, France
- Paris-Saclay University, School of Pharmacy, 91190 Orsay, France
| | - Ronan Flippot
- Laboratory of Immunomonitoring in Oncology, Gustave Roussy, 94805 Villejuif, France
- Medical Oncology Department, Institut Gustave Roussy, 94805 Villejuif, France
| | - Javier Gavira-Díaz
- Medical Oncology Department, Institut Gustave Roussy, 94805 Villejuif, France
| | - Jesus Poveda-Ferriols
- Medical Oncology Department, Centre Hospitalier Universitaire Brugmann, 1020 Brussels, Belgium
- Medical Oncology Department, Centre Hospitalier Universitaire Saint-Pierre, 1000 Brussels, Belgium
| | - Dan Nguyen
- Medical Oncology Department, Centre Hospitalier Universitaire Brugmann, 1020 Brussels, Belgium
| | - Chaimae Abdi
- Department of Medical Oncology, National Institute of Oncology, Rabat X4FH+66, Morocco
| | - Alvaro Arroyo-Salgado
- Medical Oncology Department, Centre Hospitalier Universitaire Brugmann, 1020 Brussels, Belgium
| | - Nathalie Chaput
- Laboratory of Immunomonitoring in Oncology, Gustave Roussy, 94805 Villejuif, France
| | - Guillermo de Velasco
- Medical Oncology Department, University Hospital 12 de Octubre, 28041 Madrid, Spain
| | - Laurence Albigès
- Laboratory of Immunomonitoring in Oncology, Gustave Roussy, 94805 Villejuif, France
- Medical Oncology Department, Institut Gustave Roussy, 94805 Villejuif, France
| | - Lucía Carril-Ajuria
- Medical Oncology Department, Centre Hospitalier Universitaire Brugmann, 1020 Brussels, Belgium
- Medical Oncology Department, Centre Hospitalier Universitaire Saint-Pierre, 1000 Brussels, Belgium
| |
Collapse
|
3
|
Vargová D, Dargaj J, Dohál M, Fraňová S, Ľupták J, Škorňová I, Švihra J, Briš L, Slávik P, Šutovská M. Immune analysis of urine and plasma samples from patients with clear cell renal cell carcinoma. Oncol Lett 2024; 27:281. [PMID: 38736737 PMCID: PMC11082642 DOI: 10.3892/ol.2024.14414] [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: 10/31/2023] [Accepted: 03/25/2024] [Indexed: 05/14/2024] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the third most common type of urological malignancy worldwide, and it is associated with a silent progression and late manifestation. Patients with a metastatic form of ccRCC have a poor prognosis; however, when the disease is diagnosed early, it is largely curable. Currently, there are no biomarkers available in clinical practice for ccRCC. Thus, the aim of the present study was to measure 27 biologically relevant cytokines in preoperative and postoperative urine samples, and in preoperative plasma samples from 34 patients with ccRCC, and to evaluate their diagnostic significance. The concentrations of cytokines were assessed by multiplex immune assay. The results showed significantly higher levels of IL-1 receptor antagonist, IL-6, IL-15, chemokine (C-C motif) ligand (CCL)2, CCL3, CCL4, C-X-C motif ligand (CXCL)10, granulocyte-macrophage colony stimulating factor (GM-CSF) and platelet-derived growth factor-BB (PDGF-BB), and lower levels of granulocyte colony stimulating factor (G-CSF) in urine samples from patients prior to surgery compared with those in the controls. Notably, the urine levels of G-CSF, IL-5 and vascular endothelial growth factor differed following tumor removal compared with the preoperative urine levels. In addition, urinary G-CSF, GM-CSF, IL-6, CXCL10, CCL5 and PDGF-BB appeared to be potential markers of tumor grade. Plasma from patients with ccRCC contained significantly higher levels of IL-6 and lower levels of CCL2 than control plasma. In conclusion, the present findings indicated that urinary and circulating cytokines may represent a promising novel tool for the early diagnosis of ccRCC and/or prediction of tumor grade.
Collapse
Affiliation(s)
- Daniela Vargová
- Department of Pharmacology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Ján Dargaj
- Department of Urology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava and University Hospital Martin, 036 01 Martin, Slovakia
| | - Matúš Dohál
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Soňa Fraňová
- Department of Pharmacology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Ján Ľupták
- Department of Urology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava and University Hospital Martin, 036 01 Martin, Slovakia
| | - Ingrid Škorňová
- Department of Hematology and Transfusiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava and University Hospital Martin, 036 01 Martin, Slovakia
| | - Ján Švihra
- Department of Urology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava and University Hospital Martin, 036 01 Martin, Slovakia
| | - Lukáš Briš
- Department of Urology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava and University Hospital Martin, 036 01 Martin, Slovakia
| | - Pavol Slávik
- Department of Pathological Anatomy, Jessenius Faculty of Medicine in Martin, Comenius University, 036 01 Martin, Slovakia
| | - Martina Šutovská
- Department of Pharmacology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| |
Collapse
|
4
|
Choueiri TK, Donahue AC, Braun DA, Rini BI, Powles T, Haanen JB, Larkin J, Mu XJ, Pu J, Teresi RE, di Pietro A, Robbins PB, Motzer RJ. Integrative Analyses of Tumor and Peripheral Biomarkers in the Treatment of Advanced Renal Cell Carcinoma. Cancer Discov 2024; 14:406-423. [PMID: 38385846 PMCID: PMC10905671 DOI: 10.1158/2159-8290.cd-23-0680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/22/2023] [Accepted: 12/21/2023] [Indexed: 02/23/2024]
Abstract
The phase III JAVELIN Renal 101 trial demonstrated prolonged progression-free survival (PFS) in patients (N = 886) with advanced renal cell carcinoma treated with first-line avelumab + axitinib (A+Ax) versus sunitinib. We report novel findings from integrated analyses of longitudinal blood samples and baseline tumor tissue. PFS was associated with elevated lymphocyte levels in the sunitinib arm and an abundance of innate immune subsets in the A+Ax arm. Treatment with A+Ax led to greater T-cell repertoire modulation and less change in T-cell numbers versus sunitinib. In the A+Ax arm, patients with tumors harboring mutations in ≥2 of 10 previously identified PFS-associated genes (double mutants) had distinct circulating and tumor-infiltrating immunologic profiles versus those with wild-type or single-mutant tumors, suggesting a role for non-T-cell-mediated and non-natural killer cell-mediated mechanisms in double-mutant tumors. We provide evidence for different immunomodulatory mechanisms based on treatment (A+Ax vs. sunitinib) and tumor molecular subtypes. SIGNIFICANCE Our findings provide novel insights into the different immunomodulatory mechanisms governing responses in patients treated with avelumab (PD-L1 inhibitor) + axitinib or sunitinib (both VEGF inhibitors), highlighting the contribution of tumor biology to the complexity of the roles and interactions of infiltrating immune cells in response to these treatment regimens. This article is featured in Selected Articles from This Issue, p. 384.
Collapse
Affiliation(s)
- Toni K. Choueiri
- The Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - David A. Braun
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Brian I. Rini
- Hematology Oncology, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Thomas Powles
- Department of Genitourinary Oncology, Barts Cancer Institute, Experimental Cancer Medicine Centre, Queen Mary University of London, St Bartholomew's Hospital, London, United Kingdom
| | - John B.A.G. Haanen
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - James Larkin
- Department of Medical Oncology, Royal Marsden NHS Foundation Trust, London, United Kingdom
| | | | - Jie Pu
- Pfizer, La Jolla, California
| | | | | | | | - Robert J. Motzer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
5
|
Arévalo J, Campoy I, Durán M, Nemours S, Areny A, Vall-Palomar M, Martínez C, Cantero-Recasens G, Meseguer A. STAT3 phosphorylation at serine 727 activates specific genetic programs and promotes clear cell renal cell carcinoma (ccRCC) aggressiveness. Sci Rep 2023; 13:19552. [PMID: 37945711 PMCID: PMC10636117 DOI: 10.1038/s41598-023-46628-5] [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: 03/17/2023] [Accepted: 11/03/2023] [Indexed: 11/12/2023] Open
Abstract
The signal transducer and activator of transcription 3 (STAT3) is a transcription factor mainly activated by phosphorylation in either tyrosine 705 (Y705) or serine 727 (S727) residues that regulates essential processes such as cell differentiation, apoptosis inhibition, or cell survival. Aberrant activation of STAT3 has been related to development of nearly 50% of human cancers including clear cell renal cell carcinoma (ccRCC). In fact, phosho-S727 (pS727) levels correlate with overall survival of ccRCC patients. With the aim to elucidate the contribution of STAT3 phosphorylation in ccRCC development and progression, we have generated human-derived ccRCC cell lines carrying STAT3 Y705 and S727 phosphomutants. Our data show that the phosphomimetic substitution Ser727Asp facilitates a pro-tumoral phenotype in vitro, in a Y705-phosphorylation-independent manner. Moreover, we describe that STAT3 phosphorylation state determines the expression of different subsets of target genes associated with distinct biological processes, being pS727-dependent genes the most related to cellular hallmarks of cancer. In summary, the present study constitutes the first analysis on the role of overall STAT3 phosphorylation state in ccRCC and demonstrates that pS727 promotes the expression of a specific subset of target genes that might be clinically relevant as novel biomarkers and potential therapeutic targets for ccRCC.
Collapse
Affiliation(s)
- J Arévalo
- Renal Physiopathology Group, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain.
| | - I Campoy
- Renal Physiopathology Group, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - M Durán
- Renal Physiopathology Group, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - S Nemours
- Molecular Oncology Group, Biodonostia Health Research Institute, Paseo Dr. Begiristain, s/n, 20014, San Sebastián, Spain
| | - A Areny
- Renal Physiopathology Group, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - M Vall-Palomar
- Renal Physiopathology Group, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - C Martínez
- Vascular and Renal Translational Research Group, Lleida Institute for Biomedical Research Dr. Pifarré Foundation (IRBLleida), Av. Alcalde Rovira Roure, 80, 25198, Lleida, Spain
| | - G Cantero-Recasens
- Renal Physiopathology Group, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - A Meseguer
- Renal Physiopathology Group, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain.
- Unitat de Bioquímica de Medicina, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Bellaterra, Spain.
| |
Collapse
|
6
|
Marletta S, Caliò A, Bogina G, Rizzo M, Brunelli M, Pedron S, Marcolini L, Stefanizzi L, Gobbo S, Princiotta A, Porta C, Pecoraro A, Antonelli A, Martignoni G. STING is a prognostic factor related to tumor necrosis, sarcomatoid dedifferentiation, and distant metastasis in clear cell renal cell carcinoma. Virchows Arch 2023:10.1007/s00428-023-03549-y. [PMID: 37120444 DOI: 10.1007/s00428-023-03549-y] [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: 12/04/2022] [Revised: 02/21/2023] [Accepted: 04/21/2023] [Indexed: 05/01/2023]
Abstract
STING is a molecule involved in immune reactions against double-stranded DNA fragments, released in infective and neoplastic diseases, whose role in the interactions between immune and neoplastic cells in clear cell renal cell carcinoma has not been studied yet. We investigated the immunohistochemical expression of STING in a series of 146 clear-cell renal cell carcinomas and correlated it with the main pathological prognostic factors. Furthermore, tumoral inflammatory infiltrate was evaluated and studied for the subpopulations of lymphocytes. Expression of STING was observed in 36% (53/146) of the samples, more frequently in high-grade (G3-G4) tumors (48%,43/90) and recurrent/metastatic ones (75%, 24/32) than in low grade (G1-G2) and indolent neoplasms (16%, 9/55). STING staining correlated with parameters of aggressive behavior, including coagulative granular necrosis (p = 0.001), stage (p < 0.001), and development of metastases (p < 0.001). Among prognostic parameters, STING immune expression reached an independent statistical significance (p = 0.029) in multivariable analysis, along with the stage and the presence of coagulative granular necrosis. About tumor immune-environment, no significant statistical association has been demonstrated between tumor-infiltrating lymphocytes and STING. Our results provide novel insights regarding the role of STING in aggressive clear cell renal cell carcinomas, suggesting its adoption as a prognostic marker and a potentially targetable molecule for specific immunotherapies.
Collapse
Affiliation(s)
- Stefano Marletta
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Largo L. Scuro 10, 37134, Verona, Italy
- Department of Pathology, Pederzoli Hospital, Peschiera del Garda, Italy
| | - Anna Caliò
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Largo L. Scuro 10, 37134, Verona, Italy
| | - Giuseppe Bogina
- Department of Pathology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar, Italy
| | - Mimma Rizzo
- Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, Bari, Italy
| | - Matteo Brunelli
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Largo L. Scuro 10, 37134, Verona, Italy
| | - Serena Pedron
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Largo L. Scuro 10, 37134, Verona, Italy
| | - Lisa Marcolini
- Department of Pathology, Pederzoli Hospital, Peschiera del Garda, Italy
| | | | - Stefano Gobbo
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | | | - Camillo Porta
- Interdisciplinary Department of Medicine, University of Bari "A. Moro,", Bari, Italy
| | - Angela Pecoraro
- Department of Urology, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Turin, Italy
- Department of Urology, Pederzoli Hospital, Peschiera del Garda, Italy
| | | | - Guido Martignoni
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Largo L. Scuro 10, 37134, Verona, Italy.
- Department of Pathology, Pederzoli Hospital, Peschiera del Garda, Italy.
| |
Collapse
|
7
|
High Serum Levels of IL-6 Predict Poor Responses in Patients Treated with Pembrolizumab plus Axitinib for Advanced Renal Cell Carcinoma. Cancers (Basel) 2022; 14:cancers14235985. [PMID: 36497467 PMCID: PMC9738341 DOI: 10.3390/cancers14235985] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/21/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Renal cell carcinoma (RCC) is the most common type of kidney malignancy worldwide with Pembrolizumab and axitinib treatment (Pembro/Axi) amongst the most effective first-line immunotherapies for advanced RCC. However, it remains difficult to predict treatment response and early resistance. Therefore, we evaluated whether baseline serum interleukin-6 (IL-6) could be a predictive biomarker. Between November 2019 and December 2021, 58 patients with advanced RCC were enrolled, administered first-line Pembro/Axi, and baseline blood samples were analyzed using flow cytometry. The mean baseline serum IL-6 concentration was 8.6 pg/mL in responders and 84.1 pg/mL in patients with progressive disease. The IL-6 cut-off value was set at 6.5 pg/mL using time-dependent receiver operating characteristic curves, with 37.9% of patients having high baseline serum IL-6 levels and 62.1% having low levels. Objective response rates were 58.3% and 36.4% in low and high IL-6 groups, respectively. Overall survival and progression-free survival were longer in patients with low IL-6 levels than in those with high levels. High IL-6 levels were related to reduced interferon-γ and tumor necrosis factor-α production from CD8+ T cells. Overall, high baseline serum IL-6 levels were associated with worse survival outcomes and reduced T-cell responses in Pembro/Axi-treated advanced RCC patients.
Collapse
|
8
|
Rappold PM, Vuong L, Leibold J, Chakiryan NH, Curry M, Kuo F, Sabio E, Jiang H, Nixon BG, Liu M, Berglund AE, Silagy AW, Mascareno A, Golkaram M, Marker M, Reising A, Savchenko A, Millholland J, Chen YB, Russo P, Coleman J, Reznik E, Manley BJ, Ostrovnaya I, Makarov V, DiNatale RG, Blum KA, Ma X, Chowell D, Li MO, Solit DB, Lowe SW, Chan TA, Motzer RJ, Voss MH, Hakimi AA. A Targetable Myeloid Inflammatory State Governs Disease Recurrence in Clear-Cell Renal Cell Carcinoma. Cancer Discov 2022; 12:2308-2329. [PMID: 35758895 PMCID: PMC9720541 DOI: 10.1158/2159-8290.cd-21-0925] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 04/22/2022] [Accepted: 06/22/2022] [Indexed: 11/16/2022]
Abstract
It is poorly understood how the tumor immune microenvironment influences disease recurrence in localized clear-cell renal cell carcinoma (ccRCC). Here we performed whole-transcriptomic profiling of 236 tumors from patients assigned to the placebo-only arm of a randomized, adjuvant clinical trial for high-risk localized ccRCC. Unbiased pathway analysis identified myeloid-derived IL6 as a key mediator. Furthermore, a novel myeloid gene signature strongly correlated with disease recurrence and overall survival on uni- and multivariate analyses and is linked to TP53 inactivation across multiple data sets. Strikingly, effector T-cell gene signatures, infiltration patterns, and exhaustion markers were not associated with disease recurrence. Targeting immunosuppressive myeloid inflammation with an adenosine A2A receptor antagonist in a novel, immunocompetent, Tp53-inactivated mouse model significantly reduced metastatic development. Our findings suggest that myeloid inflammation promotes disease recurrence in ccRCC and is targetable as well as provide a potential biomarker-based framework for the design of future immuno-oncology trials in ccRCC. SIGNIFICANCE Improved understanding of factors that influence metastatic development in localized ccRCC is greatly needed to aid accurate prediction of disease recurrence, clinical decision-making, and future adjuvant clinical trial design. Our analysis implicates intratumoral myeloid inflammation as a key driver of metastasis in patients and a novel immunocompetent mouse model. This article is highlighted in the In This Issue feature, p. 2221.
Collapse
Affiliation(s)
- Phillip M. Rappold
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lynda Vuong
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, MSKCC, New York, NY, USA
| | - Josef Leibold
- Cancer Biology and Genetics Program, MSKCC, New York, NY, USA
- Department of Medical Oncology & Pneumology (Internal Medicine VIII), University Hospital Tuebingen, Tuebingen 72076, Germany
- DFG Cluster of Excellence 2180 Image-Guided and Functional Instructed Tumor Therapy (iFIT), University of Tuebingen, Tuebingen 72076, Germany
| | - Nicholas H. Chakiryan
- Department of Genitourinary Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Michael Curry
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fengshen Kuo
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, MSKCC, New York, NY, USA
| | - Erich Sabio
- Human Oncology and Pathogenesis Program, MSKCC, New York, NY, USA
| | - Hui Jiang
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, MSKCC, New York, NY, USA
| | - Briana G. Nixon
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ming Liu
- Legend Biotech USA Inc, NJ, USA
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anders E. Berglund
- Department of Biostatistics and Bioinformatics, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Andrew W. Silagy
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ankur Mascareno
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, MSKCC, New York, NY, USA
| | - Mahdi Golkaram
- Illumina, Inc., 5200 Illumina Way, San Diego, CA 92122, USA
| | | | | | | | | | | | - Paul Russo
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jonathan Coleman
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ed Reznik
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Brandon J. Manley
- Department of Genitourinary Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA Integrated Mathematical Oncology Department, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Irina Ostrovnaya
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vladimir Makarov
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
| | - Renzo G. DiNatale
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kyle A. Blum
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Xiaoxiao Ma
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
| | - Diego Chowell
- Department of Oncological Sciences, The Precision Immunology Institute, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ming O. Li
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David B. Solit
- Human Oncology and Pathogenesis Program, MSKCC, New York, NY, USA
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, MSKCC, New York, NY, USA
| | - Scott W. Lowe
- Cancer Biology and Genetics Program, MSKCC, New York, NY, USA
| | - Timothy A. Chan
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
| | - Robert J. Motzer
- Department of Medicine, Genitourinary Oncology, MSKCC, New York, NY, USA
| | - Martin H. Voss
- Department of Medicine, Genitourinary Oncology, MSKCC, New York, NY, USA
| | - A. Ari Hakimi
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
9
|
Construction of an Epithelial-Mesenchymal Transition-Related Model for Clear Cell Renal Cell Carcinoma Prognosis Prediction. DISEASE MARKERS 2022; 2022:3780391. [PMID: 35983409 PMCID: PMC9381281 DOI: 10.1155/2022/3780391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 07/06/2022] [Indexed: 12/24/2022]
Abstract
Background. A rising amount of data demonstrates that the epithelial-mesenchymal transition (EMT) in clear cell renal cell carcinomas (ccRCC) is connected with the advancement of the cancer. In order to understand the role of EMT in ccRCC, it is critical to integrate molecules involved in EMT into prognosis prediction. The objective of this project was to establish a prognosis prediction model using genes associated with EMT in ccRCC. Methods. We acquired the mRNA expression profiles and clinical information about ccRCC from TCGA database. In this study, we measured differentially expressed EMT-related genes (DEEGs) by two comparison groups (tumor versus normal tissues; “stages I-II” versus “stages III-IV” tumor tissues). Based on classification and regression random forest models, we identified the most important DEEGs in predicting prognosis. Afterwards, a risk-score model was created using the identified important DEEGs. The prediction ability of the risk-score model was calculated by the area under the curve (AUC). A nomogram for prognosis prediction was built using the risk-score in combination with clinical factors. Results. Among the 72 DEEGs, the classification and regression random forest models identified six hub genes (DKK1, DLX4, IL6, KCNN4, RPL22L1, and SPDEF), which exhibited the highest importance values in both models. Through the expression of these six hub genes, a novel risk-score was developed for the prognosis prediction of ccRCC. ROC curves showed the risk-score performed well in both the training (0.749) and testing (0.777) datasets. According to the survival analysis, individuals who were separated into high/low-risk groups had statistically different outcomes in terms of prognosis. Besides, the risk-score model also showed outstanding ability in assessing the progression of ccRCC after treatment. In terms of nomogram, the concordance index (C-index) was 0.79. Additionally, we predicted the differences in response to chemotherapy drugs among patients from low- and high-risk groups. Conclusion. Gene signatures related to EMT could be useful in predicting ccRCC prognosis.
Collapse
|
10
|
Yanagisawa T, Mori K, Katayama S, Mostafaei H, Quhal F, Laukhtina E, Rajwa P, Motlagh RS, Aydh A, König F, Grossmann NC, Pradere B, Miki J, Schmidinger M, Egawa S, Shariat SF. Hematological prognosticators in metastatic renal cell cancer treated with immune checkpoint inhibitors: a meta-analysis. Immunotherapy 2022; 14:709-725. [DOI: 10.2217/imt-2021-0207] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aim: We aimed to assess the prognostic value of pretreatment hematological biomarkers in patients with metastatic renal cell carcinoma (mRCC) treated with immune checkpoint inhibitors (ICIs). Methods: PubMed, Web of Science and Scopus databases were searched for articles according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses statement. Results: Fifteen studies comprising 1530 patients were eligible for meta-analysis. High levels of neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), C-reactive protein and lactate dehydrogenase were significantly associated with worse progression-free survival. High NLR and PLR were significantly associated with worse overall survival. Conclusion: High pretreatment NLR and PLR appear to be hematological prognostic factors of progression and overall mortality in mRCC patients treated with ICIs. These findings might help in the design of correlative biomarker studies to guide the clinical decision-making in the immune checkpoint inhibitor era.
Collapse
Affiliation(s)
- Takafumi Yanagisawa
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, 1090, Austria
- Department of Urology, The Jikei University School of Medicine, Tokyo105-8461, Japan
| | - Keiichiro Mori
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, 1090, Austria
- Department of Urology, The Jikei University School of Medicine, Tokyo105-8461, Japan
| | - Satoshi Katayama
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, 1090, Austria
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry & Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Hadi Mostafaei
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, 1090, Austria
- Research Center for Evidence Based Medicine, Tabriz University of Medical Sciences, Tabriz, 5166, Iran
| | - Fahad Quhal
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, 1090, Austria
- Department of Urology, King Fahad Specialist Hospital, Dammam, 32253, Saudi Arabia
| | - Ekaterina Laukhtina
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, 1090, Austria
- Institute for Urology & Reproductive Health, Sechenov University, Moscow, 119991, Russia
| | - Pawel Rajwa
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, 1090, Austria
- Department of Urology, Medical University of Silesia, Zabrze, 41-808, Poland
| | - Reza S Motlagh
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, 1090, Austria
- Men's Health & Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran , Iran
| | - Abdulmajeed Aydh
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, 1090, Austria
- Department of Urology, King Faisal Medical City, Abha, 614312, Saudi Arabia
| | - Frederik König
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, 1090, Austria
- Department of Urology, University Medical Centre Hamburg-Eppendorf, Hamburg, 20251, Germany
| | - Nico C Grossmann
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, 1090, Austria
- Department of Urology, University Hospital Zurich, Zurich, 8091, Switzerland
- Department of Urology, Luzerner Kantonsspital, Lucerne, 6004, Switzerland
| | - Benjamin Pradere
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, 1090, Austria
| | - Jun Miki
- Department of Urology, The Jikei University School of Medicine, Tokyo105-8461, Japan
| | - Manuela Schmidinger
- Department of Medicine I & Comprehensive Cancer Center, Clinical Division of Oncology, Medical University of Vienna, Vienna, 1090, Austria
| | - Shin Egawa
- Department of Urology, The Jikei University School of Medicine, Tokyo105-8461, Japan
| | - Shahrokh F Shariat
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, 1090, Austria
- Institute for Urology & Reproductive Health, Sechenov University, Moscow, 119991, Russia
- Division of Urology, Hourani Center for Applied Scientific Research, Al-Ahliyya Amman University, Amman, 19328, Jordan
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Urology, Second Faculty of Medicine, Charles University, Prague, 11638, Czech Republic
| |
Collapse
|
11
|
Lee E, Guan P, Lim AH, Loh JW, Tan GF, Loh T, Ng DYX, Lee JY, Goh S, Liu W, Ng CCY, Teh BT, Chan JY. Multiregion sequencing of sarcomatoid renal cell carcinoma arising from autosomal dominant polycystic kidney disease. Mol Genet Genomic Med 2022; 10:e1853. [PMID: 35122417 PMCID: PMC8922955 DOI: 10.1002/mgg3.1853] [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: 07/28/2021] [Revised: 11/26/2021] [Accepted: 12/14/2021] [Indexed: 11/24/2022] Open
Abstract
Background Autosomal dominant polycystic kidney disease (ADPKD) is an inherited cystic kidney disease associated with a spectrum of various renal and extrarenal manifestations, including increased risk of kidney cancers. Here, we present the initial molecular description of sarcomatoid renal cell carcinoma (sRCC) arising in the setting of ADPKD. Methods Multiregion whole‐exome sequencing and whole transcriptomic sequencing were used to examine intratumoral molecular heterogeneity among histologically‐distinct spindle (sarcomatoid), epithelioid, or biphasic compartments within the tumor and compared with the non‐malignant ADPKD component. Results Spindle and biphasic components harbored several overlapping driver gene mutations, but do not share any with the epithelioid component. Mutations in ATM, CTNNB1, and NF2 were present only in the biphasic and spindle components, while mutations in BID, FLT3, ARID1B, and SMARCA2 were present only in the epithelioid component. We observed dichotomous evolutionary pathways in the development of epithelioid and spindle compartments, involving early mutations in TP53 and ATM/CTNNB1/NF2 respectively. Wnt, PI3K‐mTOR, and MAPK signaling pathways, known key mechanisms involved in ADPKD development, featured prominently in the sarcomatoid component. Conclusion This highlights that common pro‐oncogenic signals are present between ADPKD and sRCC providing insights into their shared pathobiology.
Collapse
Affiliation(s)
- Elizabeth Lee
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore.,Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore, Singapore
| | - Peiyong Guan
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore, Singapore.,Laboratory of Biodiversity Genomics, Genome Institute of Singapore, Singapore, Singapore.,Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Abner Herbert Lim
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore, Singapore.,Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore
| | - Jui Wan Loh
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore, Singapore.,Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore
| | - Grace Fangmin Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Tracy Loh
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| | - Dave Yong Xiang Ng
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore, Singapore.,Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Jing Yi Lee
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore, Singapore
| | - Shane Goh
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore
| | - Wei Liu
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore
| | | | - Bin Tean Teh
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore, Singapore.,Laboratory of Biodiversity Genomics, Genome Institute of Singapore, Singapore, Singapore.,Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore.,Institute of Molecular and Cellular Biology, ASTAR, Singapore, Singapore.,Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
| | - Jason Yongsheng Chan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore.,Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore.,Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
12
|
Bassanelli M, Borro M, Roberto M, Giannarelli D, Giacinti S, Di Martino S, Ceribelli A, Russo A, Aschelter A, Scarpino S, Montori A, Pescarmona E, Tomao S, Simmaco M, Cognetti F, Milella M, Marchetti P. A 17-Gene Expression Signature for Early Identification of Poor Prognosis in Clear Cell Renal Cell Carcinoma. Cancers (Basel) 2021; 14:178. [PMID: 35008342 PMCID: PMC8750239 DOI: 10.3390/cancers14010178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/13/2021] [Accepted: 12/27/2021] [Indexed: 11/16/2022] Open
Abstract
The Identification of reliable Biomarkers able to predict the outcome after nephrectomy of patients with clear cell renal cell carcinoma (ccRCC) is an unmet need. The gene expression analysis in tumor tissues represents a promising tool for better stratification of ccRCC subtypes and patients' evaluation. METHODS In our study we retrospectively analyzed using Next-Generation expression analysis (NanoString), the expression of a gene panel in tumor tissue from 46 consecutive patients treated with nephrectomy for non-metastatic ccRCC at two Italian Oncological Centres. Significant differences in expression levels of selected genes was sought. Additionally, we performed a univariate and a multivariate analysis on overall survival according to Cox regression model. RESULTS A 17-gene expression signature of patients with a recurrence-free survival (RFS) < 1 year (unfavorable genomic signature (UGS)) and of patients with a RFS > 5 years (favorable genomic signature (FGS)) was identified and resulted in being significantly correlated with overall survival of the patients included in this analysis (HR 51.37, p < 0.0001). CONCLUSIONS The identified Genomic Signatures may serve as potential biomarkers for prognosis prediction of non-metastatic RCC and could drive both follow-up and treatment personalization in RCC management.
Collapse
Affiliation(s)
- Maria Bassanelli
- Department of Medical and Surgical Sciences and Translational Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00100 Rome, Italy;
| | - Marina Borro
- (DIMA) Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, 00187 Rome, Italy;
| | - Michela Roberto
- Department of Radiological, Oncological and Anatomo-Pathological Sciences, Medical Oncology Unit, Umberto I University Hospital, Sapienza University of Rome, 00185 Rome, Italy;
| | - Diana Giannarelli
- Clinical Trial Center, Biostatistics and Bioinformatics, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144 Rome, Italy;
| | - Silvana Giacinti
- Department of Oncology, Sant’Andrea Hospital, 00187 Rome, Italy; (S.G.); (A.A.)
| | - Simona Di Martino
- Department of Pathology, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144 Rome, Italy; (S.D.M.); (A.R.); (E.P.)
| | - Anna Ceribelli
- Department of Oncology, San Camillo de Lellis Hospital, Viale Kennedy, 12100 Rieti, Italy;
| | - Andrea Russo
- Department of Pathology, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144 Rome, Italy; (S.D.M.); (A.R.); (E.P.)
| | - Annamaria Aschelter
- Department of Oncology, Sant’Andrea Hospital, 00187 Rome, Italy; (S.G.); (A.A.)
| | - Stefania Scarpino
- Department of Clinical and Molecular Medicine, Pathology Unit, St. Andrea University Hospital, University of Rome La Sapienza, 00187 Rome, Italy; (S.S.); (A.M.)
| | - Andrea Montori
- Department of Clinical and Molecular Medicine, Pathology Unit, St. Andrea University Hospital, University of Rome La Sapienza, 00187 Rome, Italy; (S.S.); (A.M.)
| | - Edoardo Pescarmona
- Department of Pathology, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144 Rome, Italy; (S.D.M.); (A.R.); (E.P.)
| | - Silverio Tomao
- Department of Radiological, Oncological and Anatomo-Pathological Sciences, Medical Oncology Unit, Umberto I University Hospital, Sapienza University of Rome, 00185 Rome, Italy;
| | - Maurizio Simmaco
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Advanced Molecular Diagnostic Unit (Dima), Sapienza University, Sant’Andrea Hospital, 00187 Rome, Italy;
| | - Francesco Cognetti
- Medical Oncology 1, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144 Rome, Italy;
| | - Michele Milella
- Division of Oncology, Integrated University Hospital of Verona, Via S. Francesco 22, 37129 Verona, Italy;
| | - Paolo Marchetti
- Department of Clinical and Molecular Medicine, Oncology Unit, Sant’ Andrea Hospital, Sapienza University of Rome, 00187 Rome, Italy;
| |
Collapse
|
13
|
Roussel E, Kinget L, Verbiest A, Debruyne PR, Baldewijns M, Van Poppel H, Albersen M, Beuselinck B. C-reactive protein and neutrophil-lymphocyte ratio are prognostic in metastatic clear-cell renal cell carcinoma patients treated with nivolumab. Urol Oncol 2021; 39:239.e17-239.e25. [PMID: 33485762 DOI: 10.1016/j.urolonc.2020.12.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/14/2020] [Accepted: 12/19/2020] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To evaluate the impact of markers of systemic inflammation such as C-reactive protein (CRP) and neutrophil-lymphocyte ratio (NLR) on outcomes of metastatic clear-cell renal cell carcinoma (m-ccRCC) patients treated with nivolumab. PATIENTS AND METHODS We retrospectively evaluated m-ccRCC patients treated with nivolumab and collected known prognostic factors and survival data. We used Kaplan-Meier survival analysis and cox proportional hazards regression analysis to study prognostic factors for overall survival (OS) and progression-free survival (PFS) since start of nivolumab. Harrell's C-index was used to evaluate the models. RESULTS We included 113 patients. Median OS and PFS after initiation of nivolumab was 15 (interquartile range 7-28) and 4 months (interquartile range 3-11), respectively. Elevated baseline CRP was associated with worse OS (HR per 25 mg/l 1.35, 95% CI 1.16-1.52, P < 0.001) and PFS (HR per 25 mg/l 1.19, 95% CI 1.08-1.35, P = 0.001), independent from the international metastatic renal cell carcinoma database consortium (IMDC) prognostic criteria, increasing the model's C-index from 0.72 to 0.77 for OS and 0.59 to 0.62 for PFS. Elevated NLR was associated with worse OS (HR 1.10, 95% CI 1.04-1.17, P = 0.002) and PFS (HR 1.06, 95% CI 1.01-1.11, P = 0.03) independent from the other IMDC prognostic criteria. The model's C-index decreased from 0.72 to 0.70 for OS and increased from 0.59 to 0.60 for PFS. CONCLUSIONS Elevated baseline CRP and NLR predict worse OS and PFS on nivolumab in m-ccRCC patients. Including baseline CRP in the IMDC prognostic model improves its discriminatory power to predict OS and PFS since start of nivolumab.
Collapse
Affiliation(s)
- Eduard Roussel
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | - Lisa Kinget
- Department of General Medical Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Annelies Verbiest
- Department of General Medical Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Philip R Debruyne
- Department of General Medical Oncology, AZ Groeninge, Kortrijk, Belgium; Faculty of Health, Education, Medicine & Social Care, Anglia Ruskin University, Chelmsford, United Kingdom
| | | | | | - Maarten Albersen
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | - Benoit Beuselinck
- Department of General Medical Oncology, University Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|