1
|
Imyanitov EN, Preobrazhenskaya EV, Mitiushkina NV. Overview on biomarkers for immune oncology drugs. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002298. [PMID: 40135049 PMCID: PMC11933888 DOI: 10.37349/etat.2025.1002298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 02/24/2025] [Indexed: 03/27/2025] Open
Abstract
Although immune checkpoint inhibitors (ICIs) are widely used in clinical oncology, less than half of treated cancer patients derive benefit from this therapy. Both tumor- and host-related variables are implicated in response to ICIs. The predictive value of PD-L1 expression is confined only to several cancer types, so this molecule is not an agnostic biomarker. Highly elevated tumor mutation burden (TMB) caused either by excessive carcinogenic exposure or by a deficiency in DNA repair is a reliable indicator for ICI efficacy, as exemplified by tumors with high-level microsatellite instability (MSI-H). Other potentially relevant tumor-related characteristics include gene expression signatures, pattern of tumor infiltration by immune cells, and, perhaps, some immune-response modifying somatic mutations. Host-related factors have not yet been comprehensively considered in relevant clinical trials. Microbiome composition, markers of systemic inflammation [e.g., neutrophil-to-lymphocyte ratio (NLR)], and human leucocyte antigen (HLA) diversity may influence the efficacy of ICIs. Studies on ICI biomarkers are likely to reveal modifiable tumor or host characteristics, which can be utilized to direct the antitumor immune defense. Examples of the latter approach include tumor priming to immune therapy by cytotoxic drugs and elevation of ICI efficacy by microbiome modification.
Collapse
Affiliation(s)
- Evgeny N. Imyanitov
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St.-Petersburg, Russia
- Department of Medical Genetics, St.-Petersburg State Pediatric Medical University, 194100 St.-Petersburg, Russia
| | - Elena V. Preobrazhenskaya
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St.-Petersburg, Russia
- Department of Medical Genetics, St.-Petersburg State Pediatric Medical University, 194100 St.-Petersburg, Russia
| | - Natalia V. Mitiushkina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St.-Petersburg, Russia
| |
Collapse
|
2
|
White BS, de Reyniès A, Newman AM, Waterfall JJ, Lamb A, Petitprez F, Lin Y, Yu R, Guerrero-Gimenez ME, Domanskyi S, Monaco G, Chung V, Banerjee J, Derrick D, Valdeolivas A, Li H, Xiao X, Wang S, Zheng F, Yang W, Catania CA, Lang BJ, Bertus TJ, Piermarocchi C, Caruso FP, Ceccarelli M, Yu T, Guo X, Bletz J, Coller J, Maecker H, Duault C, Shokoohi V, Patel S, Liliental JE, Simon S, Saez-Rodriguez J, Heiser LM, Guinney J, Gentles AJ. Community assessment of methods to deconvolve cellular composition from bulk gene expression. Nat Commun 2024; 15:7362. [PMID: 39191725 PMCID: PMC11350143 DOI: 10.1038/s41467-024-50618-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 07/11/2024] [Indexed: 08/29/2024] Open
Abstract
We evaluate deconvolution methods, which infer levels of immune infiltration from bulk expression of tumor samples, through a community-wide DREAM Challenge. We assess six published and 22 community-contributed methods using in vitro and in silico transcriptional profiles of admixed cancer and healthy immune cells. Several published methods predict most cell types well, though they either were not trained to evaluate all functional CD8+ T cell states or do so with low accuracy. Several community-contributed methods address this gap, including a deep learning-based approach, whose strong performance establishes the applicability of this paradigm to deconvolution. Despite being developed largely using immune cells from healthy tissues, deconvolution methods predict levels of tumor-derived immune cells well. Our admixed and purified transcriptional profiles will be a valuable resource for developing deconvolution methods, including in response to common challenges we observe across methods, such as sensitive identification of functional CD4+ T cell states.
Collapse
Affiliation(s)
- Brian S White
- Sage Bionetworks, Seattle, WA, USA
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Aurélien de Reyniès
- Centre de Recherche des Cordeliers, INSERM U1138, Université Paris Cité, Paris, France
| | - Aaron M Newman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Joshua J Waterfall
- INSERM U830 and Translational Research Department, Institut Curie, PSL Research University, Paris, France
| | | | - Florent Petitprez
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre le Cancer, Paris, France
- MRC Centre for Reproductive Health, the Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Yating Lin
- Xiamen University, Xiamen, Fujian, China
| | | | - Martin E Guerrero-Gimenez
- Institute of Biochemistry and Biotechnology, School of Medicine, National University of Cuyo, Mendoza, Argentina
| | | | - Gianni Monaco
- BIOGEM Institute of Molecular Biology and Genetics, Ariano Irpino, AV, Italy
| | | | | | - Daniel Derrick
- Department of Biomedical Engineering, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Alberto Valdeolivas
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Haojun Li
- Xiamen University, Xiamen, Fujian, China
| | - Xu Xiao
- Xiamen University, Xiamen, Fujian, China
| | - Shun Wang
- Department of Pathology, Cancer Hospital, Chinese Aacdemy of Medical Science, Beijing, China
| | | | | | - Carlos A Catania
- Laboratory of Intelligent Systems (LABSIN), Engineering School, National University of Cuyo, Mendoza, Argentina
| | - Benjamin J Lang
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | | | - Francesca P Caruso
- BIOGEM Institute of Molecular Biology and Genetics, Ariano Irpino, AV, Italy
| | - Michele Ceccarelli
- BIOGEM Institute of Molecular Biology and Genetics, Ariano Irpino, AV, Italy
- Sylvester Comprehensive Cancer Center, Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, Florida, USA
| | | | | | | | - John Coller
- Stanford Functional Genomics Facility, Stanford University School of Medicine, Stanford, CA, USA
| | - Holden Maecker
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Caroline Duault
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Vida Shokoohi
- Stanford Functional Genomics Facility, Stanford University School of Medicine, Stanford, CA, USA
| | - Shailja Patel
- Translational Applications Service Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Joanna E Liliental
- Translational Applications Service Center, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Julio Saez-Rodriguez
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Laura M Heiser
- Department of Biomedical Engineering, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | | | - Andrew J Gentles
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA.
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Pathology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
3
|
Xu Z, Jiang W, Liu L, Qiu Y, Wang J, Dai S, Guo J, Xu J. Dual-loss of PBRM1 and RAD51 identifies hyper-sensitive subset patients to immunotherapy in clear cell renal cell carcinoma. Cancer Immunol Immunother 2024; 73:95. [PMID: 38607586 DOI: 10.1007/s00262-024-03681-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: 10/08/2023] [Accepted: 03/17/2024] [Indexed: 04/13/2024]
Abstract
BACKGROUND Homologous recombination deficiency (HRD), though largely uncharacterized in clear cell renal cell carcinoma (ccRCC), was found associated with RAD51 loss of expression. PBRM1 is the second most common mutated genes in ccRCC. Here, we introduce a HRD function-based PBRM1-RAD51 ccRCC classification endowed with diverse immune checkpoint blockade (ICB) responses. METHODS Totally 1542 patients from four independent cohorts were enrolled, including our localized Zhongshan hospital (ZSHS) cohort and Zhongshan hospital metastatic RCC (ZSHS-mRCC) cohort, The Cancer Genome Atlas (TCGA) cohort and CheckMate cohort. The genomic profile and immune microenvironment were depicted by genomic, transcriptome data and immunohistochemistry. RESULTS We observed that PBRM1-loss ccRCC harbored enriched HRD-associated mutational signature 3 and loss of RAD51. Dual-loss of PBRM1 and RAD51 identified patients hyper-sensitive to immunotherapy. This dual-loss subtype was featured by M1 macrophage infiltration. Dual-loss was, albeit homologous recombination defective, with high chromosomal stability. CONCLUSIONS PBRM1 and RAD51 dual-loss ccRCC indicates superior responses to immunotherapy. Dual-loss ccRCC harbors an immune-desert microenvironment but enriched with M1 macrophages. Dual-loss ccRCC is susceptible to defective homologous recombination but possesses high chromosomal stability.
Collapse
Affiliation(s)
- Ziyang Xu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wenbin Jiang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Li Liu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Youqi Qiu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jiahao Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Siyuan Dai
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jianming Guo
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Jiejie Xu
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
4
|
Reimold P, Tosev G, Kaczorowski A, Friedhoff J, Schwab C, Schütz V, Görtz M, Panzer N, Heller M, Aksoy C, Himmelsbach R, Walle T, Zschäbitz S, Jäger D, Duensing A, Stenzinger A, Hohenfellner M, Duensing S. PD-L1 as a Urine Biomarker in Renal Cell Carcinoma-A Case Series and Proof-of-Concept Study. Diagnostics (Basel) 2024; 14:741. [PMID: 38611655 PMCID: PMC11011373 DOI: 10.3390/diagnostics14070741] [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: 11/13/2023] [Revised: 03/15/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND Renal cell carcinoma (RCC) is among the most lethal urologic malignancies once metastatic. Current treatment approaches for metastatic RCC (mRCC) involve immune checkpoint inhibitors (ICIs) that target the PD-L1/PD-1 axis. High PD-L1 expression in tumor tissue has been identified as a negative prognostic factor in RCC. However, the role of PD-L1 as a liquid biomarker has not yet been fully explored. Herein, we analyze urine levels of PD-L1 in mRCC patients before and after either ICI therapy or surgical intervention, as well as in a series of patients with treatment-naïve RCC. PATIENTS AND METHODS The mid-stream urine of patients with mRCC (n = 4) or treatment-naïve RCC, i.e., prior to surgery from two centers (cohort I, n = 49: cohort II, n = 29) was analyzed for PD-L1 by ELISA. The results from cohort I were compared to a control group consisting of patients treated for non-malignant urologic diseases (n = 31). In the mRCC group, urine PD-L1 levels were measured before and after tumor nephrectomy (n = 1) or before and after ICI therapy (n = 3). Exosomal PD-L1 in the urine was analyzed in selected patients by immunoblotting. RESULTS A strong decrease in urine PD-L1 levels was found after tumor nephrectomy or following systemic treatment with ICIs. In patients with treatment-naïve RCC (cohort I), urine PD-L1 levels were significantly elevated in the RCC group in comparison to the control group (median 59 pg/mL vs. 25.7 pg/mL, p = 0.011). PD-L1 urine levels were found to be elevated, in particular, in low-grade RCCs in cohorts I and II. Exosomal PD-L1 was detected in the urine of a subset of patients. CONCLUSION In this proof-of-concept study, we show that PD-L1 can be detected in the urine of RCC patients. Urine PD-L1 levels were found to correlate with the treatment response in mRCC patients and were significantly elevated in treatment-naïve RCC patients.
Collapse
Affiliation(s)
- Philipp Reimold
- Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 420, D-69120 Heidelberg, Germany
| | - Georgi Tosev
- Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 420, D-69120 Heidelberg, Germany
| | - Adam Kaczorowski
- Molecular Urooncology, University Hospital Heidelberg, Im Neuenheimer Feld 517, D-69120 Heidelberg, Germany
| | - Jana Friedhoff
- Molecular Urooncology, University Hospital Heidelberg, Im Neuenheimer Feld 517, D-69120 Heidelberg, Germany
| | - Constantin Schwab
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, D-69120 Heidelberg, Germany
| | - Viktoria Schütz
- Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 420, D-69120 Heidelberg, Germany
| | - Magdalena Görtz
- Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 420, D-69120 Heidelberg, Germany
| | - Niklas Panzer
- Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 420, D-69120 Heidelberg, Germany
| | - Martina Heller
- Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 420, D-69120 Heidelberg, Germany
| | - Cem Aksoy
- Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 420, D-69120 Heidelberg, Germany
| | - Ruth Himmelsbach
- Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 420, D-69120 Heidelberg, Germany
| | - Thomas Walle
- Department of Medical Oncology, University Hospital Heidelberg and National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, D-69120 Heidelberg, Germany
| | - Stefanie Zschäbitz
- Department of Medical Oncology, University Hospital Heidelberg and National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, D-69120 Heidelberg, Germany
| | - Dirk Jäger
- Department of Medical Oncology, University Hospital Heidelberg and National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, D-69120 Heidelberg, Germany
| | - Anette Duensing
- Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 420, D-69120 Heidelberg, Germany
- Precision Oncology of Urological Malignancies, University Hospital Heidelberg, Im Neuenheimer Feld 517, D-69120 Heidelberg, Germany
| | - Albrecht Stenzinger
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, D-69120 Heidelberg, Germany
| | - Markus Hohenfellner
- Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 420, D-69120 Heidelberg, Germany
| | - Stefan Duensing
- Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 420, D-69120 Heidelberg, Germany
- Molecular Urooncology, University Hospital Heidelberg, Im Neuenheimer Feld 517, D-69120 Heidelberg, Germany
| |
Collapse
|
5
|
Huang T, Leung B, Huang Y, Price L, Gui J, Lau BW. A murine model to evaluate immunotherapy effectiveness for human Fanconi anemia-mutated acute myeloid leukemia. PLoS One 2024; 19:e0292375. [PMID: 38289944 PMCID: PMC10826936 DOI: 10.1371/journal.pone.0292375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 09/19/2023] [Indexed: 02/01/2024] Open
Abstract
Fanconi anemia (FA)-mutated acute myeloid leukemia (AML) is a secondary AML with very poor prognosis and limited therapeutic options due to increased sensitivity to DNA-damaging agents. PD-1 immune checkpoint inhibitors upregulate T-cell killing of cancer cells and is a class of promising treatment for FA-AML. Here, we developed a novel FA-AML murine model that allows the study of human AML with a humanized immune system in order to investigate immunotherapeutic treatments in vivo. FA-AML1 cells and non-FA-mutated Kasumi-1 cells were injected into 8-10 week old NSG mice. Once leukemic engraftment was confirmed by HLA-DR expression in the peripheral blood, human peripheral blood mononuclear cells (hPBMCs) were injected into the mice. One week post-hPBMCs injection, Nivolumab (PD-1 inhibitor) or PBS vehicle control was administered to the mice bi-weekly. In our Nivolumab treated mice, FA-AML1, but not Kasumi-1-engrafted mice, had significantly prolonged overall survival. Both FA-AML1 and Kasumi-1 engrafted mice had decreased spleen weights. Higher leukemic infiltration into vital organs was observed in FA-AML1 engrafted mice compared to Kasumi-1 engrafted mice. In conclusion, our novel humanized murine model of FA-mutated AML is an attractive tool for supporting further studies and clinical trials using PD-1 inhibitors to treat FA-mutated AML.
Collapse
Affiliation(s)
- Tingting Huang
- Dartmouth Health Cancer Center, Department of Pediatrics, Geisel School of Medicine at Dartmouth College, Lebanon, NH, United States of America
| | - Bernice Leung
- Dartmouth Health Cancer Center, Department of Pediatrics, Geisel School of Medicine at Dartmouth College, Lebanon, NH, United States of America
| | - Yuyang Huang
- Dartmouth Health Cancer Center, Department of Pediatrics, Geisel School of Medicine at Dartmouth College, Lebanon, NH, United States of America
| | - Laura Price
- Dartmouth Health Cancer Center, Department of Pediatrics, Geisel School of Medicine at Dartmouth College, Lebanon, NH, United States of America
| | - Jiang Gui
- Department of Biomedical Data Science, Geisel School of Medicine, Lebanon, NH, United States of America
| | - Bonnie W. Lau
- Dartmouth Health Cancer Center, Department of Pediatrics, Geisel School of Medicine at Dartmouth College, Lebanon, NH, United States of America
| |
Collapse
|
6
|
Qiu Y, Liu L, Jiang W, Xu Z, Wang J, Dai S, Guo J, Xu J. NKG2A +CD8 + T cells infiltration determines immunosuppressive contexture and inferior response to immunotherapy in clear cell renal cell carcinoma. J Immunother Cancer 2024; 12:e008368. [PMID: 38262706 PMCID: PMC10824007 DOI: 10.1136/jitc-2023-008368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/09/2024] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND Immunotherapy is gaining momentum, but current treatments have limitations in terms of beneficiaries. Clear cell renal cell carcinoma (ccRCC) harbors the highest expression of human leukocyte antigen E (HLA-E), ligand of NKG2A, among all solid tumors. In this study, we aim to investigate the role of NKG2A+CD8+ T cells in tumor microenvironment and its potential as a novel target in ccRCC. METHODS This study included four independent cohorts, including 234 patients from Zhongshan cohort (ZSHC) who underwent partial or radical nephrectomy at Zhongshan Hospital, and 117 metastatic patients from metastatic Zhongshan cohort (ZSHC-metastatic renal cell carcinoma) who were treated with immune checkpoint inhibitor or tyrosine kinase inhibitor alone. We also incorporated a cohort of 530 patients diagnosed with ccRCC from The Cancer Genome Atlas (referred to as TCGA-kidney renal clear cell carcinoma) and 311 patients from CheckMate cohort for bioinformatics exploration and hypothesis validation. Fresh surgical specimens from 15 patients who underwent ccRCC surgery at Zhongshan Hospital were collected for flow cytometry analysis. Another 10 fresh surgical specimens were used to investigate the therapeutic potential of NKG2A blockade after in vitro intervention. The infiltration of NKG2A+CD8+ T cells was assessed using immunohistochemical staining, flow cytometry, and immunofluorescence staining in ZSHC cohort. RESULTS Patients with higher infiltration of NKG2A+CD8+ T cells in ccRCC exhibited shorter overall survival and resistance to immunotherapy. NKG2A+CD8+ T cells expressed upregulated checkpoint molecules and displayed impaired effector functions, along with tissue-residency characteristics. Combination of programmed cell death protein-1 (PD-1) blockade and NKG2A blockade demonstrated an enhanced capability in reactivating CD8+ T cells effector functions. CONCLUSION Intense infiltration of NKG2A+CD8+ T cells were associated with poorer prognosis and response to immunotherapy. NKG2A blockade combined with current immunotherapy exhibited a robust ability to reactivate CD8+ T cells effector functions.
Collapse
Affiliation(s)
- Youqi Qiu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Li Liu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenbin Jiang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ziyang Xu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiahao Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Siyuan Dai
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jianming Guo
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiejie Xu
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
7
|
Grünwald V, Ivanyi P, Zschäbitz S, Wirth M, Staib P, Schostak M, Dargatz P, Müller L, Metz M, Bergmann L, Steiner T, Welslau M, Lorch A, Rafiyan R, Hellmis E, Darr C, Schütt P, Meiler J, Kretz T, Loidl W, Flörcken A, Mänz M, Hinke A, Hartmann A, Grüllich C. Nivolumab Switch Maintenance Therapy After Tyrosine Kinase Inhibitor Induction in Metastatic Renal Cell Carcinoma: A Randomized Clinical Trial by the Interdisciplinary Working Group on Renal Tumors of the German Cancer Society (NIVOSWITCH; AIO-NZK-0116ass). Eur Urol 2023; 84:571-578. [PMID: 37758574 DOI: 10.1016/j.eururo.2023.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 07/19/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND The role of immune checkpoint inhibitor (ICI) maintenance therapy in metastatic renal cell carcinoma (mRCC) is undefined. OBJECTIVE To determine whether switch maintenance therapy with nivolumab improves clinical outcomes in patients with mRCC with tyrosine kinase inhibitor (TKI) sensitivity. DESIGN, SETTING, AND PARTICIPANTS This open-label phase 2 trial randomized patients with a partial response or stable disease after 10-12-wk TKI induction therapy to either TKI or nivolumab maintenance. Key inclusion criteria were measurable disease, clear cell histology, Eastern Cooperative Oncology Group performance status (ECOG PS) 0-2, and adequate organ function. INTERVENTION Intravenous nivolumab 8 × 240 mg every 2 wk, followed by 480 mg every 4 wk or sunitinib 50 mg (4-2 regimen) or pazopanib 800 mg once daily orally. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSES The primary endpoint was overall survival (OS). Secondary endpoints were the objective response rate (ORR; Response Evaluation Criteria in Solid Tumors v1.1), progression-free survival (PFS), safety (Common Terminology Criteria for Adverse Events v4.03), and patient-reported outcomes (Functional Assessment of Cancer Therapy Kidney Symptom Index). The Kaplan-Meier method, two-sided log-rank tests, and Cox regression models were used for statistical analysis. RESULTS AND LIMITATIONS Maintenance therapy was nivolumab for 25 patients (51.0%) and TKI for 24 (48.9%). The median age was 65 yr (range 35-79). Nine patients (18.4%) were female, 31 (63.3%) had ECOG PS of 0, and 15 (30.6%) had favorable risk. OS data are immature (17 deaths, 34.7%). The ORR was 20.0% (n = 5) for nivolumab and 52.2% (n = 12) for TKI. PFS was worse with nivolumab (hazard ratio 2.57, 95% confidence interval 1.36-4.89; p = 0.003). Grade ≥3 adverse events occurred in 14 patients (56.0%) with nivolumab and 17 (70.8%) with TKI. A major limitation is early termination of our study. CONCLUSIONS TKI treatment achieved superior ORR and PFS in comparison to nivolumab maintenance therapy. Our data do not indicate a role for nivolumab switch maintenance in mRCC. PATIENT SUMMARY Patients with metastatic kidney cancer who experienced a tumor response or disease stabilization after a short period of targeted treatment with a tyrosine kinase inhibitor did not benefit from a switch to the immunotherapy drug nivolumab. Patients who continued their original treatment achieved better responses and a longer time without disease progression. This trial is registered on EudraCT as 2016-002170-13 and on ClinicalTrials.gov as NCT02959554.
Collapse
Affiliation(s)
- Viktor Grünwald
- Interdisciplinary Genitourinary Oncology, Internal Medicine (Tumor Research) and Urology Clinics, West-German Cancer Center, University Hospital Essen, Essen, Germany.
| | - Philipp Ivanyi
- Clinic for Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Claudia von Schilling Cancer Center, Medical School Hannover, Hannover, Germany
| | - Stefanie Zschäbitz
- Department of Medical Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Manfred Wirth
- Department of Urology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Peter Staib
- Clinic for Hematology and Oncology, St. Antonius Hospital, Eschweiler, Germany
| | - Martin Schostak
- Department of Urology, Uro-Oncology, Robot-Assisted and Focal Therapy, University of Magdeburg, Magdeburg, Germany
| | | | | | - Michael Metz
- Onkologische Schwerpunktpraxis Göttingen, Göttingen, Germany
| | - Lothar Bergmann
- Medical Clinic II, University Hospital Frankfurt, Frankfurt, Germany
| | | | - Martin Welslau
- Hemato-Oncology Practice, Aschaffenburg Hospital, Aschaffenburg, Germany
| | - Anja Lorch
- Department of Urology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Medical Oncology and Hematology, University Hospital Zürich, Zürich, Switzerland
| | - Reza Rafiyan
- Clinic for Oncology and Hematology, Krankenhaus Nordwest, Frankfurt, Germany
| | | | - Cristopher Darr
- Clinic for Urology, University Hospital Essen, Essen, Germany
| | | | | | | | - Wolfgang Loidl
- Department of Urology and Andrology, Ordensklinikum Linz, Linz, Austria
| | - Anne Flörcken
- Department of Hematology, Oncology and Tumorimmunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Axel Hinke
- Cancer Clinical Research Consulting, Düsseldorf, Germany
| | - Arndt Hartmann
- Institute for Pathology, University Hospital Erlangen, Erlangen, Germany
| | - Carsten Grüllich
- Department of Hematology and Oncology, Caritas-Hospital Lebach, Lebach, Germany
| |
Collapse
|
8
|
Du L, Wang B, Wu M, Chen W, Wang W, Diao W, Ding M, Chen W, Cao W, Guo H, Zhang G. LINC00926 promotes progression of renal cell carcinoma via regulating miR-30a-5p/SOX4 axis and activating IFNγ-JAK2-STAT1 pathway. Cancer Lett 2023; 578:216463. [PMID: 37866544 DOI: 10.1016/j.canlet.2023.216463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 10/24/2023]
Abstract
The role of long non-coding RNA (lncRNA) in the progression of renal cell carcinoma (RCC) remains further study. Whether lncRNA may be used to predict the immunotherapy efficacy of RCC is less studied. In this study, LINC00926 was found to be mainly located in cytoplasm by FISH and RNA nuclear-cytoplasmic fractionation. Downregulation of LINC00926 in RCC cell lines inhibited the progression and metastasis of RCC cells. RNA pull-down assay and dual-luciferase reporter assay demonstrated that LINC00926 functioned as miR-30a-5p sponge to facilitate SOX4 expression. LINC00926 overexpression in BALB/c mice enhanced PD-L1 surface expression and resulted in immune escape. Mechanistic investigations showed that LINC00926 competitively bound to Lyn, leading to the inhibition of CBL-mediated ubiquitination and degradation, and stabilized Lyn, contributing to the activation of IFNγ-JAK2-STAT1 signaling pathway. Moreover, LINC00926, together with PD-L1 or PD-1 expression, may predict the overall survival in RCC patients. Therefore, LINC00926 has the potential to be a novel therapeutic target and a biomarker to predict ICB immunotherapy response in RCC.
Collapse
Affiliation(s)
- Lin Du
- Department of Urology, Nanjing Drum Tower Hospital Clinical College of Southeast University, Nanjing, 210008, Jiangsu, China; Department of Urology, The First People's Hospital of Yancheng, Yancheng, 224006, Jiangsu, China
| | - Baojun Wang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230039, Anhui, China; Department of Urology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, Jiangsu, China
| | - Mengtong Wu
- Department of Urology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, Jiangsu, China
| | - Weixu Chen
- Department of Urology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, Jiangsu, China
| | - Wendi Wang
- Department of Urology, Nanjing Drum Tower Hospital Clinical College of Southeast University, Nanjing, 210008, Jiangsu, China
| | - Wenli Diao
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, Nanjing, 210008, Jiangsu, China
| | - Meng Ding
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, Nanjing, 210008, Jiangsu, China
| | - Wei Chen
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, Nanjing, 210008, Jiangsu, China
| | - Wenmin Cao
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, Nanjing, 210008, Jiangsu, China
| | - Hongqian Guo
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, Nanjing, 210008, Jiangsu, China.
| | - Gutian Zhang
- Department of Urology, Nanjing Drum Tower Hospital Clinical College of Southeast University, Nanjing, 210008, Jiangsu, China; Department of Urology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
9
|
Farha M, Nallandhighal S, Vince R, Cotta B, Stangl-Kremser J, Triner D, Morgan TM, Palapattu GS, Cieslik M, Vaishampayan U, Udager AM, Salami SS. Analysis of the Tumor Immune Microenvironment (TIME) in Clear Cell Renal Cell Carcinoma (ccRCC) Reveals an M0 Macrophage-Enriched Subtype: An Exploration of Prognostic and Biological Characteristics of This Immune Phenotype. Cancers (Basel) 2023; 15:5530. [PMID: 38067234 PMCID: PMC10705373 DOI: 10.3390/cancers15235530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 10/31/2023] [Accepted: 11/16/2023] [Indexed: 02/12/2024] Open
Abstract
There is a need to optimize the treatment of clear cell renal cell carcinoma (ccRCC) patients at high recurrence risk after nephrectomy. We sought to elucidate the tumor immune microenvironment (TIME) of localized ccRCC and understand the prognostic and predictive characteristics of certain features. The discovery cohort was clinically localized patients in the TCGA-Kidney Renal Clear Cell Carcinoma (KIRC) project (n = 382). We identified an M0 macrophage-enriched cluster (n = 25) in the TCGA-KIRC cohort. This cluster's median progression-free survival (PFS) and overall survival (OS) were 40.4 and 45.3 months, respectively, but this was not reached in the others (p = 0.0003 and <0.0001, respectively). Gene set enrichment (GSEA) analysis revealed an enrichment of epithelial to mesenchymal transition and cell cycle progression genes within this cluster, and these patients also had a lower predicted response to immune checkpoint blockade (ICB) (4% vs. 20-34%). An M0-enriched cluster (n = 9) with shorter PFS (p = 0.0006) was also identified in the Clinical Proteomics Tumor Analysis Consortium (CPTAC) cohort (n = 94). Through this characterization of the TIME in ccRCC, a cluster of patients defined by enrichment in M0 macrophages was identified that demonstrated poor prognosis and lower predicted ICB response. Pending further validation, this signature can identify localized ccRCC patients at high risk of recurrence after nephrectomy and who may require therapeutic approaches beyond ICB monotherapy.
Collapse
Affiliation(s)
- Mark Farha
- Department of Medical Education, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (M.F.); (U.V.)
| | | | - Randy Vince
- Department of Urology, Michigan Medicine, Ann Arbor, MI 48109, USA
| | - Brittney Cotta
- Department of Urology, Michigan Medicine, Ann Arbor, MI 48109, USA
| | - Judith Stangl-Kremser
- Department of Urology, Michigan Medicine, Ann Arbor, MI 48109, USA
- Department of Urology, Medical University of Vienna, 1090 Vienna, Austria
| | - Daniel Triner
- Department of Urology, Michigan Medicine, Ann Arbor, MI 48109, USA
| | - Todd M. Morgan
- Department of Urology, Michigan Medicine, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA; (M.C.); (A.M.U.)
| | - Ganesh S. Palapattu
- Department of Medical Education, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (M.F.); (U.V.)
- Department of Urology, Michigan Medicine, Ann Arbor, MI 48109, USA
- Department of Urology, Medical University of Vienna, 1090 Vienna, Austria
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA; (M.C.); (A.M.U.)
| | - Marcin Cieslik
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA; (M.C.); (A.M.U.)
- Department of Pathology, Michigan Medicine, Ann Arbor, MI 48109, USA
- Michigan Center for Translational Pathology, Michigan Medicine, Ann Arbor, MI 48109, USA
| | - Ulka Vaishampayan
- Department of Medical Education, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (M.F.); (U.V.)
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA; (M.C.); (A.M.U.)
- Department of Medicine, Michigan Medicine, Ann Arbor, MI 48109, USA
| | - Aaron M. Udager
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA; (M.C.); (A.M.U.)
- Department of Pathology, Michigan Medicine, Ann Arbor, MI 48109, USA
- Michigan Center for Translational Pathology, Michigan Medicine, Ann Arbor, MI 48109, USA
| | - Simpa S. Salami
- Department of Medical Education, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (M.F.); (U.V.)
- Department of Urology, Michigan Medicine, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA; (M.C.); (A.M.U.)
- Michigan Center for Translational Pathology, Michigan Medicine, Ann Arbor, MI 48109, USA
| |
Collapse
|
10
|
Matar S, El Ahmar N, Laimon YN, Ghandour F, Signoretti S. The Role of the Pathologist in Renal Cell Carcinoma Management. Hematol Oncol Clin North Am 2023; 37:849-862. [PMID: 37258353 DOI: 10.1016/j.hoc.2023.04.014] [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] [Indexed: 06/02/2023]
Abstract
Recent advances in our understanding of the molecular alterations underlying different types of renal cell carcinoma (RCC), as well as the implementation of immune checkpoint inhibitors in the treatment of patients with advanced disease, have significantly expanded the role of pathologists in the management of RCC patients and in the identification of predictive biomarkers that can guide patient treatment. In this chapter, we examine pathologists' evolving role in patient care and the development of precision medicine strategies for RCC.
Collapse
Affiliation(s)
- Sayed Matar
- Department of Pathology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA; Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Nourhan El Ahmar
- Department of Pathology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA; Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Yasmin Nabil Laimon
- Department of Pathology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA; Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Fatme Ghandour
- Department of Pathology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA; Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Sabina Signoretti
- Department of Pathology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA; Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Merkin Building, 415 Main Street, Cambridge, MA 02142, USA; Department of Oncologic Pathology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA.
| |
Collapse
|
11
|
Bersanelli M, Gnetti L, Pilato FP, Varotti E, Quaini F, Campanini N, Rapacchi E, Camisa R, Carbognani P, Silini EM, Rusca M, Leonardi F, Maestroni U, Rizzo M, Brunelli M, Buti S, Ampollini L. Retrospective immunophenotypical evaluation of MET, PD-1/PD-L1, and mTOR pathways in primary tumors and pulmonary metastases of renal cell carcinoma: the RIVELATOR study addresses the issue of biomarkers heterogeneity. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:743-756. [PMID: 37720351 PMCID: PMC10501858 DOI: 10.37349/etat.2023.00165] [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: 02/27/2023] [Accepted: 05/21/2023] [Indexed: 09/19/2023] Open
Abstract
Aim In renal cell carcinoma (RCC), tumor heterogeneity generated challenges to biomarker development and therapeutic management, often becoming responsible for primary and acquired drug resistance. This study aimed to assess the inter-tumoral, intra-tumoral, and intra-lesional heterogeneity of known druggable targets in metastatic RCC (mRCC). Methods The RIVELATOR study was a monocenter retrospective analysis of biological samples from 25 cases of primary RCC and their paired pulmonary metastases. The biomarkers analyzed included MET, mTOR, PD-1/PD-L1 pathways and the immune context. Results High multi-level heterogeneity was demonstrated. MET was the most reliable biomarker, with the lowest intratumor heterogeneity: the positive mutual correlation between MET expression in primary tumors and their metastases had a significantly proportional intensity (P = 0.038). The intratumor heterogeneity grade was significantly higher for the mTOR pathway proteins. Combined immunophenotypical expression patterns and their correlations with the immune context were uncovered [i.e., mTOR expression in the metastases positively correlated with PD-L1 expression in tumor-infiltrating lymphocytes (TILs), P = 0.019; MET expression was related to PD-1 expression on TILs (P = 0.041, ρ = 0.41) and peritumoral lymphocytes (RILs; P = 0.013, ρ = 0.49)], suggesting the possibility of predicting drug response or resistance to tyrosine kinase, mTOR, or immune checkpoint inhibitors. Conclusions In mRCC, multiple and multi-level assays of potentially predictive biomarkers are needed for their reliable translation into clinical practice. The easy-to-use immunohistochemical method of the present study allowed the identification of different combined expression patterns, providing cues for planning the management of systemic treatment combinations and sequences in an mRCC patient population. The quantitative heterogeneity of the investigated biomarkers suggests that multiple intralesional assays are needed to consider the assessment reliable for clinical considerations.
Collapse
Affiliation(s)
| | - Letizia Gnetti
- Pathologic Anatomy Unit, University Hospital of Parma, 43126 Parma, Italy
| | | | - Elena Varotti
- Pathologic Anatomy Unit, Azienda Socio-Sanitaria Territoriale di Cremona, 26100 Cremona, Italy
| | - Federico Quaini
- Medicine and Surgery Department, University of Parma, 43126 Parma, Italy
| | - Nicoletta Campanini
- Pathologic Anatomy Unit, University Hospital of Parma, 43126 Parma, Italy
- Medicine and Surgery Department, University of Parma, 43126 Parma, Italy
| | - Elena Rapacchi
- Medical Oncology Unit, University Hospital of Parma, 43126 Parma, Italy
| | - Roberta Camisa
- Medical Oncology Unit, University Hospital of Parma, 43126 Parma, Italy
| | - Paolo Carbognani
- Medicine and Surgery Department, University of Parma, 43126 Parma, Italy
- Thoracic Surgery Unit, University Hospital of Parma, 43126 Parma, Italy
| | - Enrico Maria Silini
- Pathologic Anatomy Unit, University Hospital of Parma, 43126 Parma, Italy
- Medicine and Surgery Department, University of Parma, 43126 Parma, Italy
| | - Michele Rusca
- Medicine and Surgery Department, University of Parma, 43126 Parma, Italy
| | | | | | - Mimma Rizzo
- Medical Oncology Unit, Azienda Ospedaliero Universitaria Consorziale Policlinico, 70124 Bari, Italy
| | - Matteo Brunelli
- Pathologic Anatomy Unit, University Hospital of Verona, 37126 Verona, Italy
- Medicine and Surgery Department, University of Verona, 37126 Verona, Italy
| | - Sebastiano Buti
- Medical Oncology Unit, University Hospital of Parma, 43126 Parma, Italy
- Medicine and Surgery Department, University of Parma, 43126 Parma, Italy
| | - Luca Ampollini
- Medicine and Surgery Department, University of Parma, 43126 Parma, Italy
- Thoracic Surgery Unit, University Hospital of Parma, 43126 Parma, Italy
| |
Collapse
|
12
|
Shen L, Brown JR, Johnston SA, Altan M, Sykes KF. Predicting response and toxicity to immune checkpoint inhibitors in lung cancer using antibodies to frameshift neoantigens. J Transl Med 2023; 21:338. [PMID: 37217961 DOI: 10.1186/s12967-023-04172-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 04/30/2023] [Indexed: 05/24/2023] Open
Abstract
PURPOSE To evaluate a new class of blood-based biomarkers, anti-frameshift peptide antibodies, for predicting both tumor responses and adverse immune events to immune checkpoint inhibitor (ICI) therapies in advanced lung cancer patients. EXPERIMENTAL DESIGN Serum samples were obtained from 74 lung cancer patients prior to palliative PD-(L)1 therapies with subsequently recorded tumor responses and immune adverse events (irAEs). Pretreatment samples were assayed on microarrays of frameshift peptides (FSPs), representing ~ 375,000 variant peptides that tumor cells can be informatically predicted to produce from translated mRNA processing errors. Serum-antibodies specifically recognizing these ligands were measured. Binding activities preferentially associated with best-response and adverse-event outcomes were determined. These antibody bound FSPs were used in iterative resampling analyses to develop predictive models of tumor response and immune toxicity. RESULTS Lung cancer serum samples were classified based on predictive models of ICI treatment outcomes. Disease progression was predicted pretreatment with ~ 98% accuracy in the full cohort of all response categories, though ~ 30% of the samples were indeterminate. This model was built with a heterogeneous sample cohort from patients that (i) would show either clear response or stable outcomes, (ii) would be administered either single or combination therapies and (iii) were diagnosed with different lung cancer subtypes. Removing the stable disease, combination therapy or SCLC groups from model building increased the proportion of samples classified while performance remained high. Informatic analyses showed that several of the FSPs in the all-response model mapped to translations of variant mRNAs from the same genes. In the predictive model for treatment toxicities, binding to irAE-associated FSPs provided 90% accuracy pretreatment, with no indeterminates. Several of the classifying FSPs displayed sequence similarity to self-proteins. CONCLUSIONS Anti-FSP antibodies may serve as biomarkers for predicting ICI outcomes when tested against ligands corresponding to mRNA-error derived FSPs. Model performances suggest this approach might provide a single test to predict treatment response to ICI and identify patients at high risk for immunotherapy toxicities.
Collapse
Affiliation(s)
- Luhui Shen
- Calviri, Inc, 850 N 5th St., Phoenix, AZ, 85004, USA
| | | | | | - Mehmet Altan
- MD Anderson Cancer Center, Department of Thoracic-Head & Neck Medical Oncology, Division of Cancer Medicine, Houston, TX, USA
| | | |
Collapse
|
13
|
Tian J, Cheng C, Gao J, Fu G, Xu Z, Chen X, Wu Y, Jin B. POLD1 as a Prognostic Biomarker Correlated with Cell Proliferation and Immune Infiltration in Clear Cell Renal Cell Carcinoma. Int J Mol Sci 2023; 24:ijms24076849. [PMID: 37047824 PMCID: PMC10095303 DOI: 10.3390/ijms24076849] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/25/2023] [Accepted: 03/31/2023] [Indexed: 04/08/2023] Open
Abstract
DNA polymerase delta 1 catalytic subunit (POLD1) plays a vital role in genomic copy with high fidelity and DNA damage repair processes. However, the prognostic value of POLD1 and its relationship with tumor immunity in clear cell renal cell carcinoma (ccRCC) remains to be further explored. Transcriptional data sets and clinical information were obtained from the TCGA, ICGC, and GEO databases. Differentially expressed genes (DEGs) were derived from the comparison between the low and high POLD1 expression groups in the TCGA–KIRC cohort. KEGG and gene ontology (GO) analyses were performed for those DEGs to explore the potential influence of POLD1 on the biological behaviors of ccRCC. The prognostic clinical value and mutational characteristics of patients were described and analyzed according to the POLD1 expression levels. TIMER and TISIDB databases were utilized to comprehensively investigate the potential relevance between the POLD1 levels and the status of the immune cells, as well as the tumor infiltration of immune cells. In addition, RT-qPCR, Western blot, immunohistochemistry and several functional and animal experiments were performed for clinical, in vitro and in vivo validation. POLD1 was highly expressed in a variety of tumors including ccRCC, and further verified in a validation cohort of 60 ccRCC samples and in vitro cell line experiments. POLD1 expression levels in the ccRCC samples were associated with various clinical characteristics including pathologic tumor stage and histologic grade. ccRCC patients with high POLD1 expression have poor clinical outcomes and exhibit a higher rate of somatic mutations than those with low POLD1 expression. Cox regression analysis also showed that POLD1 could act as a potential independent prognostic biomarker. The DEGs associated with POLD1 were significantly enriched in the immunity-related pathways. Moreover, further immune infiltration analysis indicated that high POLD1 expression was associated with high NK CD56bright cells, Treg cells, and myeloid-derived suppressor cells’ (MDSCs) infiltration scores, as well as their marker gene sets of immune cell status. Meanwhile, POLD1 exhibited resistance to various drugs when highly expressed. Finally, the knockdown of POLD1 inhibited the proliferation and migration, and promoted the apoptosis of ccRCC cells in vitro and in vivo, as well as influenced the activation of oncogenic signaling. Our current study demonstrated that POLD1 is a potential prognostic biomarker for ccRCC patients. It might create a tumor immunosuppressive microenvironment and inhibit the susceptibility to ferroptosis leading to a poor prognosis.
Collapse
Affiliation(s)
- Junjie Tian
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310024, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310024, China
| | - Cheng Cheng
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310024, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310024, China
| | - Jianguo Gao
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310024, China
| | - Guanghou Fu
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310024, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310024, China
| | - Zhijie Xu
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310024, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310024, China
| | - Xiaoyi Chen
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310024, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310024, China
| | - Yunfei Wu
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310024, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310024, China
| | - Baiye Jin
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310024, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310024, China
| |
Collapse
|
14
|
Zheng Z, Li X, Nie K, Wang X, Liang W, Yang F, Zheng K, Zheng Y. Identification of berberine as a potential therapeutic strategy for kidney clear cell carcinoma and COVID-19 based on analysis of large-scale datasets. Front Immunol 2023; 14:1038651. [PMID: 37033923 PMCID: PMC10076552 DOI: 10.3389/fimmu.2023.1038651] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 03/14/2023] [Indexed: 04/11/2023] Open
Abstract
Background Regarding the global coronavirus disease 2019 (COVID)-19 pandemic, kidney clear cell carcinoma (KIRC) has acquired a higher infection probability and may induce fatal complications and death following COVID-19 infection. However, effective treatment strategies remain unavailable. Berberine exhibits significant antiviral and antitumour effects. Thus, this study aimed to provide a promising and reliable therapeutic strategy for clinical decision-making by exploring the therapeutic mechanism of berberine against KIRC/COVID-19. Methods Based on large-scale data analysis, the target genes, clinical risk, and immune and pharmacological mechanisms of berberine against KIRC/COVID-19 were systematically investigated. Results In total, 1,038 and 12,992 differentially expressed genes (DEGs) of COVID-19 and KIRC, respectively, were verified from Gene Expression Omnibus and The Cancer Genome Atlas databases, respectively, and 489 berberine target genes were obtained from official websites. After intersecting, 26 genes were considered potential berberine therapeutic targets for KIRC/COVID-19. Berberine mechanism of action against KIRC/COVID-19 was revealed by protein-protein interaction, gene ontology, and Kyoto Encyclopedia of Genes and Genomes with terms including protein interaction, cell proliferation, viral carcinogenesis, and the PI3K/Akt signalling pathway. In COVID-19 patients, ACOX1, LRRK2, MMP8, SLC1A3, CPT1A, H2AC11, H4C8, and SLC1A3 were closely related to disease severity, and the general survival of KIRC patients was closely related to ACOX1, APP, CPT1A, PLK1, and TYMS. Additionally, the risk signature accurately and sensitively depicted the overall survival and patient survival status for KIRC. Numerous neutrophils were enriched in the immune system of COVID-19 patients, and the lives of KIRC patients were endangered due to significant immune cell infiltration. Molecular docking studies indicated that berberine binds strongly to target proteins. Conclusion This study demonstrated berberine as a potential treatment option in pharmacological, immunological, and clinical practice. Moreover, its therapeutic effects may provide potential and reliable treatment options for patients with KIRC/COVID-19.
Collapse
Affiliation(s)
- Zhihua Zheng
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Xiushen Li
- Shenzhen Key Laboratory, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| | - Kechao Nie
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xiaoyu Wang
- Department of Nephrology, Health College of Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Wencong Liang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Fuxia Yang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Kairi Zheng
- Traditional Chinese Medicine Department, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- *Correspondence: Kairi Zheng, ; Yihou Zheng,
| | - Yihou Zheng
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
- *Correspondence: Kairi Zheng, ; Yihou Zheng,
| |
Collapse
|
15
|
Yang H, Xue M, Su P, Zhou Y, Li X, Li Z, Xia Y, Zhang C, Fu M, Zheng X, Luo G, Wei T, Wang X, Ding Y, Zhu J, Zhuang T. RNF31 represses cell progression and immune evasion via YAP/PD-L1 suppression in triple negative breast Cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:364. [PMID: 36581998 PMCID: PMC9801641 DOI: 10.1186/s13046-022-02576-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 12/19/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND Recently genome-based studies revealed that the abnormality of Hippo signaling is pervasive in TNBC and played important role in cancer progression. RING finger protein 31 (RNF31) comes to RING family E3 ubiquitin ligase. Our previously published studies have revealed RNF31 is elevated in ER positive breast cancer via activating estrogen signaling and suppressing P53 pathway. METHODS We used several TNBC cell lines and xenograft models and performed immuno-blots, QPCR, in vivo studies to investigate the function of RNF31 in TNBC progression. RESULT Here, we demonstrate that RNF31 plays tumor suppressive function in triple negative breast cancer (TNBC). RNF31 depletion increased TNBC cell proliferation and migration in vitro and in vitro. RNF31 depletion in TNBC coupled with global genomic expression profiling indicated Hippo signaling could be the potential target for RNF31 to exert its function. Further data showed that RNF31 depletion could increase the level of YAP protein, and Hippo signaling target genes expression in several TNBC cell lines, while clinical data illustrated that RNF31 expression correlated with longer relapse-free survival in TNBC patients and reversely correlated with YAP protein level. The molecular biology assays implicated that RNF31 could associate with YAP protein, facilitate YAP poly-ubiquitination and degradation at YAP K76 sites. Interestingly, RNF31 could also repress PDL1 expression and sensitive TNBC immunotherapy via inhibiting Hippo/YAP/PDL1 axis. CONCLUSIONS Our study revealed the multi-faced function of RNF31 in different subtypes of breast malignancies, while activation RNF31 could be a plausible strategy for TNBC therapeutics.
Collapse
Affiliation(s)
- Huijie Yang
- grid.412990.70000 0004 1808 322XXinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003 Henan Province People’s Republic of China
| | - Min Xue
- grid.440265.10000 0004 6761 3768Molecular Biology Laboratory, First People’s Hospital of Shangqiu, Shangqiu, City, 476000 Henan Province People’s Republic of China
| | - Peng Su
- Department of Pathology, Shandong University Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan City, Shandong Province People’s Republic of China
| | - Yan Zhou
- grid.27255.370000 0004 1761 1174Department of General Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province 250033 People’s Republic of China
| | - Xin Li
- grid.412990.70000 0004 1808 322XXinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003 Henan Province People’s Republic of China
| | - Zhongbo Li
- grid.412990.70000 0004 1808 322XXinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003 Henan Province People’s Republic of China
| | - Yan Xia
- grid.412990.70000 0004 1808 322XXinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003 Henan Province People’s Republic of China
| | - Chenmiao Zhang
- grid.412990.70000 0004 1808 322XXinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003 Henan Province People’s Republic of China
| | - Mingxi Fu
- grid.412990.70000 0004 1808 322XXinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003 Henan Province People’s Republic of China
| | - Xiuxia Zheng
- grid.440265.10000 0004 6761 3768Molecular Biology Laboratory, First People’s Hospital of Shangqiu, Shangqiu, City, 476000 Henan Province People’s Republic of China
| | - Guosheng Luo
- grid.412990.70000 0004 1808 322XThe Affiliated people’s Hospital of Xinxiang Medical University, Xinxiang, 453003 Henan Province People’s Republic of China
| | - Tian Wei
- grid.27255.370000 0004 1761 1174Department of General Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province 250033 People’s Republic of China
| | - Xinxing Wang
- grid.412633.10000 0004 1799 0733Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450052 People’s Republic of China
| | - Yinlu Ding
- grid.27255.370000 0004 1761 1174Department of General Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province 250033 People’s Republic of China
| | - Jian Zhu
- grid.27255.370000 0004 1761 1174Department of General Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province 250033 People’s Republic of China
| | - Ting Zhuang
- grid.412990.70000 0004 1808 322XXinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003 Henan Province People’s Republic of China ,grid.412990.70000 0004 1808 322XThe Affiliated people’s Hospital of Xinxiang Medical University, Xinxiang, 453003 Henan Province People’s Republic of China
| |
Collapse
|
16
|
Bialek J, Yankulov S, Kawan F, Fornara P, Theil G. Role of Nivolumab in the Modulation of PD-1 and PD-L1 Expression in Papillary and Clear Cell Renal Carcinoma (RCC). Biomedicines 2022; 10:biomedicines10123244. [PMID: 36552000 PMCID: PMC9776360 DOI: 10.3390/biomedicines10123244] [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: 11/14/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
The expression and cellular mechanisms of programmed cell death-1 protein (PD-1) and its ligands (PD-L1 and PD-L2) in renal cancer cells are not well known. Here, we aimed to investigate the response of renal carcinoma subtypes to the immune checkpoint inhibitor nivolumab and its impact on related signaling pathways. All cell lines analyzed (clear cell (cc)RCC (Caki-1, RCC31) and papillary (p)RCC (ACHN, RCC30)) expressed PD-1 and both ccRCC cell lines, and RCC30 expressed PD-L1. Nivolumab treatment at increasing doses led to increased PD-1 levels in analyzed cells and resulted in aggressive behavior of pRCC but diminished this behavior in ccRCC. The analysis of PD-1/PD-L1-associated signaling pathways demonstrated increased AKT activity in Caki-1 and RCC30 cells but decreased activity in ACHN and RCC31 cells, while ribosomal protein S6 remained largely unchanged. Androgen receptors are related to RCC and were predominantly increased in RCC30 cells, which were the only cells that formed nivolumab-dependent spheroids. Finally, all cell lines exhibited a complex response to nivolumab treatment. Since the pRCC cells responded with increased tumorigenicity and PD-1/PD-L1 levels while ccRCC tumorigenicity was diminished, further studies are needed to improve nivolumab-based therapy for renal carcinoma subtypes, especially the identification of response-involved molecular pathways.
Collapse
|