1
|
Lou K, Cheng X. Prognostic value of the neutrophil‑to‑lymphocyte ratio in renal cell carcinoma: A systematic review and meta‑analysis. Oncol Lett 2025; 29:231. [PMID: 40114748 PMCID: PMC11925002 DOI: 10.3892/ol.2025.14977] [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: 11/01/2024] [Accepted: 02/11/2025] [Indexed: 03/22/2025] Open
Abstract
The neutrophil-to-lymphocyte ratio (NLR) not only indicates the inflammatory response within the tumor microenvironment but may also correlate with tumor biological behavior (such as aggressiveness). The present study aimed to systematically review and conduct a meta-analysis on the impact of the NLR on the prognosis of patients with renal cell carcinoma (RCC). To this aim, a comprehensive search of multiple relevant databases, including PubMed, Embase and the Cochrane Library, was conducted to identify literature related to NLR and RCC prognosis. Following rigorous literature screening and quality assessment, a systematic quantitative analysis was ultimately performed on several studies that met the inclusion criteria. The results indicated a significant association between elevated NLR levels and poor prognosis in patients with RCC, suggesting that high NLR levels may serve as an independent predictor of unfavorable outcomes. Therefore, the present study provides important evidence for clinical decision-making, further demonstrating that NLR can serve as an independent prognostic indicator for patients with RCC, aiding healthcare professionals in making more precise judgments in patient management and treatment strategy formulation.
Collapse
Affiliation(s)
- Kecheng Lou
- Department of Urology, Lanxi People's Hospital, Jinhua, Zhejiang 321100, P.R. China
| | - Xin Cheng
- Department of Urology, Ganzhou Cancer Hospital of Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| |
Collapse
|
2
|
Wang L, Zhu Y, Ren Z, Sun W, Wang Z, Zi T, Li H, Zhao Y, Qin X, Gao D, Zhang L, He Z, Le W, Wu Q, Wu G. An immunogenic cell death-related classification predicts prognosis and response to immunotherapy in kidney renal clear cell carcinoma. Front Oncol 2023; 13:1147805. [PMID: 37681027 PMCID: PMC10482408 DOI: 10.3389/fonc.2023.1147805] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 08/03/2023] [Indexed: 09/09/2023] Open
Abstract
Introduction Immunogenic cell death (ICD) is a form of regulated cell death that activates an adaptive immune response in an immunocompetent host and is particularly sensitive to antigens from tumor cells. Kidney clear cell carcinoma (KIRC) is an immunogenic tumor with extensive tumor heterogeneity. However, no reliable predictive biomarkers have been identified to reflect the immune microenvironment and therapeutic response of KIRC. Methods Therefore, we used the CIBERSORT and ESTIMATE algorithms to define three ICD clusters based on the expression of ICD-related genes in 661 KIRC patients. Subsequently, we identified three different ICD gene clusters based on the overlap of differentially expressed genes (DEGs) within the ICD clusters. In addition, principal component analysis (PCA) was performed to calculate the ICD scores. Results The results showed that patients with reduced ICD scores had a poorer prognosis and reduced transcript levels of immune checkpoint genes regulated with T cell differentiation. Furthermore, the ICD score was negatively correlated with the tumor mutation burden (TMB) value of KICD. patients with higher ICD scores showed clinical benefits and advantages of immunotherapy, indicating that the ICD score is an accurate and valid predictor to assess the effect of immunotherapy. Discussion Overall, our study presents a comprehensive KICD immune-related ICD landscape that can provide guidance for current immunotherapy and predict patient prognosis to help physicians make judgments about the patient's disease and treatment modalities, and can guide current research on immunotherapy strategies for KICD.
Collapse
Affiliation(s)
- Licheng Wang
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yaru Zhu
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhen Ren
- School of Biomedical Engineering, Dalian University of Technology, Dalian, China
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wenhuizi Sun
- Department of Obstetrics and Gynecology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhijing Wang
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Tong Zi
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Haopeng Li
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yan Zhao
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xin Qin
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Dacheng Gao
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Libo Zhang
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ziyang He
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wei Le
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qiang Wu
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Gang Wu
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
3
|
Kammerhofer G, Vegh D, Bányai D, Végh Á, Joob-Fancsaly A, Hermann P, Geczi Z, Hegedus T, Somogyi KS, Bencze B, Biczó Z, Juhász DH, Zaborszky P, Ujpál M, Vaszilkó MT, Németh Z. Association between Hyperglycemia and Medication-Related Osteonecrosis of the Jaw (MRONJ). J Clin Med 2023; 12:jcm12082976. [PMID: 37109314 PMCID: PMC10144577 DOI: 10.3390/jcm12082976] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/12/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND Medication-related osteonecrosis of the jaw (MRONJ) is a type of jawbone necrosis caused by the use of drugs for some types of cancer and osteoporosis. The current study aimed to evaluate the associations between hyperglycemia and the development of medication-related osteonecrosis of the jaw. METHODS Our research group investigated data collected between 1 January 2019 and 31 December 2020. A total of 260 patients were selected from the Inpatient Care Unit, Department of Oromaxillofacial Surgery and Stomatology, Semmelweis University. Fasting glucose data were used and included in the study. RESULTS Approximately 40% of the necrosis group and 21% of the control group presented with hyperglycemia. There was a significant association between hyperglycemia and MRONJ (p < 0.05, p = 0.003). Vascular anomaly and immune dysfunction caused by hyperglycemia can lead to necrosis after tooth extraction. Necrosis is more common in the mandible (75.0%) and in the case of parenteral antiresorptive treatment (intravenous Zoledronate and subcutaneous Denosumab). Hyperglycemia is a more relevant risk factor than bad oral habits (26.7%). CONCLUSIONS Ischemia is a complication of abnormal glucose levels, a possible risk factor for necrosis development. Hence, uncontrolled or poorly regulated plasma glucose levels can significantly increase the risk of jawbone necrosis after invasive dental or oral surgical interventions.
Collapse
Affiliation(s)
- Gabor Kammerhofer
- Department of Oromaxillofacial Surgery and Stomatology, Semmelweis University, 1088 Budapest, Hungary
- Diabetes-Dental Working Group, Semmelweis University, 1088 Budapest, Hungary
| | - Daniel Vegh
- Diabetes-Dental Working Group, Semmelweis University, 1088 Budapest, Hungary
- Department of Prosthodontics, Semmelweis University, 1088 Budapest, Hungary
| | - Dorottya Bányai
- Diabetes-Dental Working Group, Semmelweis University, 1088 Budapest, Hungary
- Department of Paediatric Dentistry and Orthodontics, Semmelweis University, 1088 Budapest, Hungary
| | - Ádám Végh
- Diabetes-Dental Working Group, Semmelweis University, 1088 Budapest, Hungary
- Department of Oral Diagnostics, Semmelweis University, 1088 Budapest, Hungary
| | - Arpad Joob-Fancsaly
- Department of Oromaxillofacial Surgery and Stomatology, Semmelweis University, 1088 Budapest, Hungary
- Diabetes-Dental Working Group, Semmelweis University, 1088 Budapest, Hungary
| | - Peter Hermann
- Diabetes-Dental Working Group, Semmelweis University, 1088 Budapest, Hungary
- Department of Prosthodontics, Semmelweis University, 1088 Budapest, Hungary
| | - Zoltan Geczi
- Diabetes-Dental Working Group, Semmelweis University, 1088 Budapest, Hungary
- Department of Prosthodontics, Semmelweis University, 1088 Budapest, Hungary
| | - Tamas Hegedus
- Diabetes-Dental Working Group, Semmelweis University, 1088 Budapest, Hungary
- Department of Prosthodontics, Semmelweis University, 1088 Budapest, Hungary
| | - Kata Sara Somogyi
- Diabetes-Dental Working Group, Semmelweis University, 1088 Budapest, Hungary
- Department of Prosthodontics, Semmelweis University, 1088 Budapest, Hungary
| | - Bulcsú Bencze
- Diabetes-Dental Working Group, Semmelweis University, 1088 Budapest, Hungary
- Department of Prosthodontics, Semmelweis University, 1088 Budapest, Hungary
| | - Zita Biczó
- Department of Oromaxillofacial Surgery and Stomatology, Semmelweis University, 1088 Budapest, Hungary
- Diabetes-Dental Working Group, Semmelweis University, 1088 Budapest, Hungary
| | - Donát Huba Juhász
- Diabetes-Dental Working Group, Semmelweis University, 1088 Budapest, Hungary
- Faculty of Dentistry, Semmelweis University, 1088 Budapest, Hungary
| | - Péter Zaborszky
- Diabetes-Dental Working Group, Semmelweis University, 1088 Budapest, Hungary
- Faculty of Dentistry, Semmelweis University, 1088 Budapest, Hungary
| | - Márta Ujpál
- Department of Oromaxillofacial Surgery and Stomatology, Semmelweis University, 1088 Budapest, Hungary
- Diabetes-Dental Working Group, Semmelweis University, 1088 Budapest, Hungary
| | - Mihály Tamás Vaszilkó
- Department of Oromaxillofacial Surgery and Stomatology, Semmelweis University, 1088 Budapest, Hungary
- Diabetes-Dental Working Group, Semmelweis University, 1088 Budapest, Hungary
| | - Zsolt Németh
- Department of Oromaxillofacial Surgery and Stomatology, Semmelweis University, 1088 Budapest, Hungary
- Diabetes-Dental Working Group, Semmelweis University, 1088 Budapest, Hungary
| |
Collapse
|
4
|
Matsuki M, Hirohashi Y, Nakatsugawa M, Murai A, Kubo T, Hashimoto S, Tokita S, Murata K, Kanaseki T, Tsukahara T, Nishida S, Tanaka T, Kitamura H, Masumori N, Torigoe T. Tumor-infiltrating CD8 + T cells recognize a heterogeneously expressed functional neoantigen in clear cell renal cell carcinoma. Cancer Immunol Immunother 2021; 71:905-918. [PMID: 34491407 DOI: 10.1007/s00262-021-03048-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 08/26/2021] [Indexed: 11/24/2022]
Abstract
Immune checkpoint inhibitors (ICIs) are used in cancer immunotherapy to block programmed death-1 and cytotoxic T-lymphocyte antigen 4, but the response rate for ICIs is still low and tumor cell heterogeneity is considered to be responsible for resistance to immunotherapy. Tumor-infiltrating lymphocytes (TILs) have an essential role in the anti-tumor effect of cancer immunotherapy; however, the specificity of TILs in renal cell carcinoma (RCC) is elusive. In this study, we analyzed a 58-year-old case with clear cell RCC (ccRCC) with the tumor showing macroscopic and microscopic heterogeneity. The tumor was composed of low-grade and high-grade ccRCC. A tumor cell line (1226 RCC cells) and TILs were isolated from the high-grade ccRCC lesion, and a TIL clone recognized a novel neoantigen peptide (YVVPGSPCL) encoded by a missense mutation of the tensin 1 (TNS1) gene in a human leukocyte antigen-C*03:03-restricted fashion. The TNS1 gene mutation was not detected in the low-grade ccRCC lesion and the TIL clone did not recognized low-grade ccRCC cells. The missense mutation of TNS1 encoding the S1309Y mutation was found to be related to cell migration by gene over-expression. These findings suggest that macroscopically and microscopically heterogenous tumors might show heterogenous gene mutations and reactivity to TILs.
Collapse
Affiliation(s)
- Masahiro Matsuki
- Department of Pathology, Sapporo Medical University School of Medicine, South-1 West-17, Chuo-Ku, Sapporo, 060-8556, Japan.,Department of Urology, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Japan
| | - Yoshihiko Hirohashi
- Department of Pathology, Sapporo Medical University School of Medicine, South-1 West-17, Chuo-Ku, Sapporo, 060-8556, Japan.
| | - Munehide Nakatsugawa
- Department of Pathology, Sapporo Medical University School of Medicine, South-1 West-17, Chuo-Ku, Sapporo, 060-8556, Japan.,Department of Diagnostic Pathology, Tokyo Medical University Hachioji Medical Center, Hachioji, Tokyo, 193-0998, Japan
| | - Aiko Murai
- Department of Pathology, Sapporo Medical University School of Medicine, South-1 West-17, Chuo-Ku, Sapporo, 060-8556, Japan
| | - Terufumi Kubo
- Department of Pathology, Sapporo Medical University School of Medicine, South-1 West-17, Chuo-Ku, Sapporo, 060-8556, Japan
| | - Shinichi Hashimoto
- Department of Molecular Pathophysiology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, 641-8509, Japan
| | - Serina Tokita
- Department of Pathology, Sapporo Medical University School of Medicine, South-1 West-17, Chuo-Ku, Sapporo, 060-8556, Japan
| | - Kenji Murata
- Department of Pathology, Sapporo Medical University School of Medicine, South-1 West-17, Chuo-Ku, Sapporo, 060-8556, Japan
| | - Takayuki Kanaseki
- Department of Pathology, Sapporo Medical University School of Medicine, South-1 West-17, Chuo-Ku, Sapporo, 060-8556, Japan
| | - Tomohide Tsukahara
- Department of Pathology, Sapporo Medical University School of Medicine, South-1 West-17, Chuo-Ku, Sapporo, 060-8556, Japan
| | - Sachiyo Nishida
- Department of Urology, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Japan
| | - Toshiaki Tanaka
- Department of Urology, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Japan
| | - Hiroshi Kitamura
- Department of Urology, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Naoya Masumori
- Department of Urology, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Japan
| | - Toshihiko Torigoe
- Department of Pathology, Sapporo Medical University School of Medicine, South-1 West-17, Chuo-Ku, Sapporo, 060-8556, Japan.
| |
Collapse
|
5
|
Singh A, Singh I, Singh N, Puzanov I. Optimal Management of First-Line Advanced Renal Cell Carcinoma: Focus on Pembrolizumab. Onco Targets Ther 2020; 13:4021-4034. [PMID: 32494157 PMCID: PMC7231754 DOI: 10.2147/ott.s215173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 04/03/2020] [Indexed: 01/05/2023] Open
Abstract
Renal cell carcinoma (RCC) is among the 10 most common cancers in the USA. One-third of the patients diagnosed with this cancer present with locally advanced or metastatic disease. In the past, advanced disease conferred poor survival outcomes; however, the treatment paradigm for RCC has been revolutionized twice since 2005. The initial wave of revolution came with the emergence of vascular endothelial growth factor (VEGF) inhibitors and a second wave arose more recently with the emergence and unprecedented success of checkpoint inhibitors in RCC. A third wave combining these two strategies is well underway and likely represents the new paradigm to improve survival outcomes for afflicted patients. In this review, we discuss the current treatment landscape for patients with advanced RCC, focusing on approved VEGF and checkpoint inhibitors in the first-line setting as well as highlighting landmark combination clinical trials.
Collapse
Affiliation(s)
- Abhay Singh
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Inderpreet Singh
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Namrata Singh
- Department of Medicine, Punjab Institute of Medical Sciences, Punjab, India
| | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
6
|
Hansen UK, Ramskov S, Bjerregaard AM, Borch A, Andersen R, Draghi A, Donia M, Bentzen AK, Marquard AM, Szallasi Z, Eklund AC, Svane IM, Hadrup SR. Tumor-Infiltrating T Cells From Clear Cell Renal Cell Carcinoma Patients Recognize Neoepitopes Derived From Point and Frameshift Mutations. Front Immunol 2020; 11:373. [PMID: 32226429 PMCID: PMC7080703 DOI: 10.3389/fimmu.2020.00373] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 02/17/2020] [Indexed: 12/14/2022] Open
Abstract
Mutation-derived neoantigens are important targets for T cell-mediated reactivity toward tumors and, due to their unique tumor expression, an attractive target for immunotherapy. Neoepitope-specific T cells have been detected across a number of solid cancers with high mutational burden tumors, but neoepitopes have been mostly selected from single nucleotide variations (SNVs), and little focus has been given to neoepitopes derived from in-frame and frameshift indels, which might be equally important and potentially highly immunogenic. Clear cell renal cell carcinomas (ccRCCs) are medium-range mutational burden tumors with a high pan-cancer proportion of frameshift mutations. In this study, the mutational landscape of tumors from six RCC patients was analyzed by whole-exome sequencing (WES) of DNA from tumor fragments (TFs), autologous tumor cell lines (TCLs), and tumor-infiltrating lymphocytes (TILs, germline reference). Neopeptides were predicted using MuPeXI, and patient-specific peptide–MHC (pMHC) libraries were created for all neopeptides with a rank score < 2 for binding to the patient's HLAs. T cell recognition toward neoepitopes in TILs was evaluated using the high-throughput technology of DNA barcode-labeled pMHC multimers. The patient-specific libraries consisted of, on average, 258 putative neopeptides (range, 103–397, n = 6). In four patients, WES was performed on two different sources (TF and TCL), whereas in two patients, WES was performed only on TF. Most of the peptides were predicted from both sources. However, a fraction was predicted from one source only. Among the total predicted neopeptides, 16% were derived from frameshift indels. T cell recognition of 52 neoepitopes was detected across all patients (range, 4–18, n = 6) and spanning two to five HLA restrictions per patient. On average, 21% of the recognized neoepitopes were derived from frameshift indels (range, 0–43%, n = 6). Thus, frameshift indels are equally represented in the pool of immunogenic neoepitopes as SNV-derived neoepitopes. This suggests the importance of a broad neopeptide prediction strategy covering multiple sources of tumor material, and including different genetic alterations. This study, for the first time, describes the T cell recognition of frameshift-derived neoepitopes in RCC and determines their immunogenic profile.
Collapse
Affiliation(s)
- Ulla Kring Hansen
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Sofie Ramskov
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | | | - Annie Borch
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Rikke Andersen
- Center for Cancer Immune Therapy, Copenhagen University Hospital, Copenhagen, Denmark
| | - Arianna Draghi
- Center for Cancer Immune Therapy, Copenhagen University Hospital, Copenhagen, Denmark
| | - Marco Donia
- Center for Cancer Immune Therapy, Copenhagen University Hospital, Copenhagen, Denmark
| | - Amalie Kai Bentzen
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | | | | | - Aron Charles Eklund
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark.,Clinical Microbiomics A/S, Copenhagen, Denmark
| | - Inge Marie Svane
- Center for Cancer Immune Therapy, Copenhagen University Hospital, Copenhagen, Denmark
| | - Sine Reker Hadrup
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| |
Collapse
|
7
|
Sorigue M, Prusila RE, Jauhiainen J, Mercadal S, Postila A, Salmi P, Tanhua T, Tikkanen S, Kakko S, Kuitunen H, Pollari M, Nystrand I, Kuusisto ME, Vasala K, Jantunen E, Korkeila E, Karihtala P, Sancho JM, Turpeenniemi-Hujanen T, Kuittinen O. Incidence of solid cancer in patients with follicular lymphoma. Acta Oncol 2019; 58:1564-1569. [PMID: 31368395 DOI: 10.1080/0284186x.2019.1643918] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Introduction: Patients with follicular lymphoma (FL) have classically had a higher risk of solid cancers than the general population, but there is little data available in patients diagnosed and treated with modern day regimens.Material and methods: We conducted a retrospective multicenter study assessing the cumulative incidence of solid cancers other than nonmelanoma skin cancer in patients with FL between 1997 and 2016 and determined the standardized incidence ratio (SIR) to compare the incidence of solid cancers with that of the general populationResults: Among 1002 FL patients with 7 years of median follow-up, we found 74 solid cancers (most common breast [n = 19], lung and colon [n = 9 each]). The cumulative incidence was 3.8% at 5 years (95%CI 2.6-5.2) from the time of diagnosis and 4.4% at 5 years (95%CI 3.1-5.9%) from the time of front-line treatment. Although a comparison of all front-line strategies did not reveal differences in the risk of solid cancers, patients treated with anthracycline-based regimens appeared to have a lower incidence than those treated with bendamustine-based strategies (2.8% vs. 6.9%). However, patients receiving the former regimen were younger than the latter. On multivariable analysis, older age was correlated with the incidence of solid cancer and bendamustine-based treatment was of borderline significance. SIR for any solid cancer was 1.22 (95%CI 0.91-1.64), indicating no increased risk of solid cancer in patients with FL over that of the general population. However, on subgroup analyses, female patients treated with bendamustine-based strategies appeared to have a greater risk (SIR 3.85 [95%CI 1.45-10.27])Discussion: The incidence of solid cancer in this cohort of patients with FL was low and not greater than in the general population. However, the risk may be greater in female patients treated with bendamustine.
Collapse
Affiliation(s)
- Marc Sorigue
- Department of Hematology, ICO-Hospital Germans Trias i Pujol, Institut de Recerca Josep Carreras, Barcelona, Spain
| | - Roosa E.I. Prusila
- Medical Research Center, Oulu University Hospital and Cancer and Translational Research Unit, University of Oulu, Oulu, Finland
- Department of Oncology and Radiotherapy, Oulu University Hospital, Oulu, Finland
| | - Jyrki Jauhiainen
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland
| | - Santiago Mercadal
- Department of Hematology, ICO-Hospital Duran i Reynals, Hospitalet de Llobregat, Spain
| | - Aleksi Postila
- Medical Research Center, Oulu University Hospital and Cancer and Translational Research Unit, University of Oulu, Oulu, Finland
- Department of Oncology and Radiotherapy, Oulu University Hospital, Oulu, Finland
| | - Petteri Salmi
- Medical Research Center, Oulu University Hospital and Cancer and Translational Research Unit, University of Oulu, Oulu, Finland
- Department of Oncology and Radiotherapy, Oulu University Hospital, Oulu, Finland
| | - Taru Tanhua
- Medical Research Center, Oulu University Hospital and Cancer and Translational Research Unit, University of Oulu, Oulu, Finland
- Department of Oncology and Radiotherapy, Oulu University Hospital, Oulu, Finland
| | - Susanna Tikkanen
- Medical Research Center, Oulu University Hospital and Cancer and Translational Research Unit, University of Oulu, Oulu, Finland
- Department of Oncology and Radiotherapy, Oulu University Hospital, Oulu, Finland
| | - Sakari Kakko
- Department of Oncology and Radiotherapy, Oulu University Hospital, Oulu, Finland
| | - Hanne Kuitunen
- Medical Research Center, Oulu University Hospital and Cancer and Translational Research Unit, University of Oulu, Oulu, Finland
- Department of Oncology and Radiotherapy, Oulu University Hospital, Oulu, Finland
| | - Marjukka Pollari
- Department of Oncology, Tampere University Hospital, Tampere, Finland
| | - Ilja Nystrand
- Department of Oncology, University of Turku and Turku University Hospital, Turku, Finland
| | - Milla E.L. Kuusisto
- Medical Research Center, Oulu University Hospital and Cancer and Translational Research Unit, University of Oulu, Oulu, Finland
- Siunsote – Hospital District of North Carelia, Joensuu, Finland
| | - Kaija Vasala
- Department of Oncology and Radiotherapy, Central Finland Central Hospital, Jyvaskyla, Finland
| | - Esa Jantunen
- Siunsote – Hospital District of North Carelia, Joensuu, Finland
- Department of Medicine, Kuopio University Hospital, Kuopio, Finland
- Institute of Clinical Medicine, Faculty of Health Medicine, University of Eastern Finland, Kuopio, Finland
| | - Eija Korkeila
- Department of Oncology, University of Turku and Turku University Hospital, Turku, Finland
| | - Peeter Karihtala
- Medical Research Center, Oulu University Hospital and Cancer and Translational Research Unit, University of Oulu, Oulu, Finland
- Department of Oncology and Radiotherapy, Oulu University Hospital, Oulu, Finland
| | - Juan-Manuel Sancho
- Department of Hematology, ICO-Hospital Germans Trias i Pujol, Institut de Recerca Josep Carreras, Barcelona, Spain
| | - Taina Turpeenniemi-Hujanen
- Medical Research Center, Oulu University Hospital and Cancer and Translational Research Unit, University of Oulu, Oulu, Finland
- Department of Oncology and Radiotherapy, Oulu University Hospital, Oulu, Finland
| | - Outi Kuittinen
- Department of Oncology and Radiotherapy, Oulu University Hospital, Oulu, Finland
- Institute of Clinical Medicine, Faculty of Health Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Oncology, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
8
|
Zhao W, Zhao F, Yang K, Lu Y, Zhang Y, Wang W, Xie H, Deng K, Yang C, Rong Z, Hou Y, Li K. An immunophenotyping of renal clear cell carcinoma with characteristics and a potential therapeutic target for patients insensitive to immune checkpoint blockade. J Cell Biochem 2019; 120:13330-13341. [PMID: 30916827 DOI: 10.1002/jcb.28607] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/07/2019] [Accepted: 01/14/2019] [Indexed: 12/11/2022]
Abstract
Renal clear cell carcinoma (RCC) patients who do not achieve optimal control of progression with immune checkpoint blockade (ICB) should be further studied. Unsupervised consensus clustering was used to group 525 RCC patients based on two typical ICB pathways, CTLA-4 and pogrammed death 1 (PD-1)/programmed death-ligand 1 (PD-L1), as well as two new discovered regulators, CMTM6 and CMTM4. Three immune molecular subtypes (IMMSs) with different clinical and immunological characteristics were identified (type I, II, and III), among which there were more stage I and low-grade tumors in type I RCC than in type II and III. The proportion of males was highest in type II RCC. Overall survival of type II and III was similar (5.2 and 6 years) and statistically shorter than that of type I (7.6 years) before and after adjusting for age and gender. When conducting stratified analysis, our IMMSs were able to identify high-risk patients among middle-aged patients, males, and stage IV patients. Among the differentially expressed genes, approximately 84% were highly expressed in type II and III RCC. Genes related to ICB (CTLA-4, CD274, and PDCD1LG2) and cytotoxic lymphocytes (CD8A, GZMA, and PRF1) were all highly expressed in type II and III RCC. These results documented that patients with type II and III cancer may be more sensitive to anti-CTLA-4 therapy, anti-PD-1/PD-L1 therapy, and a combination of immunotherapies. High expression of CMTM4 in type I RCC (69%) and a statistically significant interaction of CD274 and CMTM6 indicated that CMTM4/6 might be new therapy targets for type I, who are resistant to ICB.
Collapse
Affiliation(s)
- Weiwei Zhao
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, People's Republic of China
| | - Falin Zhao
- Department of Health Management, School of Medicine, Hangzhou Normal University, Hangzhou, People's Republic of China
| | - Kai Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, People's Republic of China
| | - Yaxin Lu
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, People's Republic of China
| | - Yuanyuan Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, People's Republic of China
| | - Wenjie Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, People's Republic of China
| | - Hongyu Xie
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, People's Republic of China
| | - Kui Deng
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, People's Republic of China
| | - Chunyan Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, People's Republic of China
| | - Zhiwei Rong
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, People's Republic of China
| | - Yan Hou
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, People's Republic of China
| | - Kang Li
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, People's Republic of China
| |
Collapse
|
9
|
Kwong DL, Lee VH, Nicholls JM. Cancer Immunotherapy for Nasopharyngeal Carcinoma. NASOPHARYNGEAL CARCINOMA 2019:337-351. [DOI: 10.1016/b978-0-12-814936-2.00015-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
10
|
Association between melanoma and renal-cell carcinoma for sequential diagnoses: A single-center retrospective study. Cancer Epidemiol 2018; 57:80-84. [PMID: 30347335 DOI: 10.1016/j.canep.2018.10.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 10/02/2018] [Accepted: 10/03/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND Melanoma and renal-cell carcinoma (RCC) are known to be immunological neoplasms. Previous studies have shown increased risks in patients with melanoma of developing RCC and in those with RCC of developing melanoma. However, data regarding immunocompromised status in these patients are lacking. METHODS We conducted a retrospective review of patients who had a diagnosis of melanoma and/or RCC. Using summary statistics, we calculated total person-years at risk for developing melanoma among patients with RCC and for developing RCC among patients with melanoma, and compared the results with the SEER data. We also assessed medical history related to immune status and the use of immunosuppressive drugs. RESULTS Among 13,879 patients with melanoma and 7597 patients with RCC, 89 had diagnoses of both melanoma and RCC (0.6% and 1.2% of melanoma and RCC patients, respectively): eight were diagnosed with both cancers concurrently, 54 were diagnosed with melanoma first, and 27 were diagnosed with RCC first. Standardized incidence ratios (SIRs) were 2.87 (95%CI 2.16-3.74) for developing RCC among the melanoma patients and 2.31 (95%CI 1.52-3.37) for developing melanoma among the RCC patients, compared to age-, sex-, race-, and calendar-specific adjusted incidence rates of each cancer in the SEER registry. None of the 81 patients with sequential diagnoses had a history of immunocompromised disease, nor did they receive chronic immunosuppressive drugs. Only two received chemotherapy and/or radiotherapy. CONCLUSION We demonstrated a strong association between the diagnoses of melanoma and RCC. These increased risks could not be attributed to either immune status or previous antineoplastic treatment.
Collapse
|
11
|
Nakanishi H, Miyata Y, Mochizuki Y, Yasuda T, Nakamura Y, Araki K, Sagara Y, Matsuo T, Ohba K, Sakai H. Pathological significance and prognostic roles of densities of CD57+ cells, CD68+ cells, and mast cells, and their ratios in clear cell renal cell carcinoma. Hum Pathol 2018; 79:102-108. [PMID: 29787818 DOI: 10.1016/j.humpath.2018.05.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 04/29/2018] [Accepted: 05/11/2018] [Indexed: 01/18/2023]
Abstract
The immune system is closely associated with malignant behavior in renal cell carcinoma (RCC). Therefore, understanding the pathological roles of immune cells in tumor stroma is essential to discuss the pathological characteristics of RCC. In this study, the clinical significance of densities of CD57+ cells, CD68+ cells, and mast cells, and their ratios were investigated in patients with clear cell RCC. The densities of CD57+, CD68+, and mast cells were evaluated by immunohistochemical techniques in 179 patients. Proliferation index, apoptotic index, and microvessel density were evaluated by using anti-Ki-67, anti-cleaved caspase-3, and anti-CD31 antibodies, respectively. The density of CD57+ cell was negatively correlated with grade, pT stage, and metastasis, although densities of CD68+ cell and mast cell were positively correlated. Ratios of CD68+ cell/CD57+ cell and mast cell/CD57+ cell were significantly correlated with grade, pT stage, and metastasis. Survival analyses showed that the CD68+ cell/CD57+ cell ratio was a significant predictor for cause-specific survival by multivariate analyses (hazard ratio = 1.41, 95% confidence interval = 1.03-1.93, P = .031) and was significantly correlated with proliferation index, apoptotic index, and microvessel density (r = .47, P <. 001; r = -.31, P < .001; and r = .40, P < .001, respectively). In conclusion, CD57+ cells, CD68+ cells, and mast cells played important roles in malignancy in clear cell RCC. The CD68+ cell/CD57+ cell ratio was strongly correlated with pathological features and prognosis in these patients because this ratio reflected the status of cancer cell proliferation, apoptosis, and angiogenesis.
Collapse
Affiliation(s)
- Hiromi Nakanishi
- Department of Nephro-Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Yasuyoshi Miyata
- Department of Nephro-Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan.
| | - Yasushi Mochizuki
- Department of Nephro-Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Takuji Yasuda
- Department of Nephro-Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Yuichiro Nakamura
- Department of Nephro-Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Kyohei Araki
- Department of Nephro-Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Yuji Sagara
- Department of Nephro-Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Tomohiro Matsuo
- Department of Nephro-Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Kojiro Ohba
- Department of Nephro-Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Hideki Sakai
- Department of Nephro-Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| |
Collapse
|
12
|
Wang Y, Fu D, Chen Y, Su J, Wang Y, Li X, Zhai W, Niu Y, Yue D, Geng H. G3BP1 promotes tumor progression and metastasis through IL-6/G3BP1/STAT3 signaling axis in renal cell carcinomas. Cell Death Dis 2018; 9:501. [PMID: 29717134 PMCID: PMC5931548 DOI: 10.1038/s41419-018-0504-2] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 03/18/2018] [Accepted: 03/20/2018] [Indexed: 12/12/2022]
Abstract
The chronic inflammatory microenvironment within or surrounding the primary renal cell carcinoma (RCC) site promotes oncogenic transformation as well as contributes to the development of metastasis. G3BP stress granule assembly factor 1 (G3BP1) was found to be involved in the regulation of multiple cellular functions. However, its functions in RCC have not been previously explored. Here, we first showed that the expression of G3BP1 is elevated in human RCC and correlates with RCC progression. In cultured RCC cells, knockdown of G3BP1 results in inhibition of tumor cell proliferation, migration, and invasion, consistently with the alteration of epithelial–mesenchymal transition (EMT) and cell proliferative markers, including Cadherins, Vimentin, Snail, Slug, c-Myc, and cyclin D1. Remarkably, knockdown of G3BP1 dramatically impaired the signaling connection of pro-inflammatory cytokine IL-6 stimulation and downstream STAT3 activation in RCC, thus eventually contributing to the disruption of IL-6-elicited RCC migration and metastasis. In addition, in vivo orthotopic tumor xenografts results confirmed that knockdown of G3BP1 suppressed RCC tumor growth and metastasis in mice. Collectively, our findings support the notion that G3BP1 promotes tumor progression and metastasis through IL-6/G3BP1/STAT3 signaling axis in RCC.
Collapse
Affiliation(s)
- Yong Wang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin Medical University, Tianjin, 300211, China
| | - Donghe Fu
- Department of Microbiology, School of Medical Laboratory, Tianjin Medical University, Tianjin, 300203, China
| | - Yajing Chen
- Research Center of Molecular Biology, Inner Mongolia Medical University, Hohhot, 010059, China
| | - Jing Su
- Department of Microbiology, School of Medical Laboratory, Tianjin Medical University, Tianjin, 300203, China.,Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Yiting Wang
- Department of Microbiology, School of Medical Laboratory, Tianjin Medical University, Tianjin, 300203, China.,Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Xin Li
- Department of Pharmacology, Tianjin Medical University, Tianjin, 300070, China
| | - Wei Zhai
- Department of Urology, Renji Hospital, School of Medicine in Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yuanjie Niu
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin Medical University, Tianjin, 300211, China
| | - Dan Yue
- Department of Microbiology, School of Medical Laboratory, Tianjin Medical University, Tianjin, 300203, China.
| | - Hua Geng
- Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
13
|
Liu W, Liu Y, Fu Q, Zhou L, Chang Y, Xu L, Zhang W, Xu J. Elevated expression of IFN-inducible CXCR3 ligands predicts poor prognosis in patients with non-metastatic clear-cell renal cell carcinoma. Oncotarget 2017; 7:13976-83. [PMID: 26910919 PMCID: PMC4924692 DOI: 10.18632/oncotarget.7468] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 02/05/2016] [Indexed: 02/06/2023] Open
Abstract
IFN-inducible CXCR3 ligands (ICL), namely CXCL9, CXCL10 and CXCL11, exhibit pleiotropic roles in orchestrating immunity and angiogenesis. However, the prognosis value of them in renal cell carcinoma (RCC) was still obscure. Thus, we retrospectively used immunohistochemistry approach to evaluate the impact of these ligands on recurrence and survival of non-metastatic clear cell RCC (ccRCC) patients after nephrectomy. We systemically built a prespecified ICL score based on these ligands, and found specimens with high ICL score were prone to possess high Fuhrman grade, necrosis, and high-risk level of SSIGN. Moreover, ICL score stratified patients into different risk subgroups, and remained an independent adverse prognosticator for overall survival (OS) and recurrence-free survival (RFS). Meanwhile, in TCGA database, the increasing ICL mRNA predicted poor survival and early recurrence. Furthermore, after adding ICL score into SSIGN, the C-index for OS and RFS increased from 0.705 to 0.746 and 0.712 to 0.765, respectively. In conclusion, the ICL score based on expression of CXCL9, CXCL10 and CXCL11 stratified non-metastatic ccRCC patients into different risk subgroups of recurrence and death, which might benefit preoperative risk stratification and guide immune therapy in the future.
Collapse
Affiliation(s)
- Weisi Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yidong Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qiang Fu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Lin Zhou
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuan Chang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Le Xu
- Department of Urology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Weijuan Zhang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jiejie Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
14
|
Li J, Sun J, Rong R, Li L, Shang W, Song D, Feng G, Luo F. HMGB1 promotes myeloid-derived suppressor cells and renal cell carcinoma immune escape. Oncotarget 2017; 8:63290-63298. [PMID: 28968989 PMCID: PMC5609921 DOI: 10.18632/oncotarget.18796] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 06/02/2017] [Indexed: 01/01/2023] Open
Abstract
Despite high immunogenicity and marked presence of immune cells in the RCC(renal cell carcinoma), immunotherapy fails to develop effective anti-tumor immune responses. This is due to the negative regulatory factors in the tumor microenvironment. As the main contributor of immunosuppression, myeloid-derived suppressor cells (MDSCs) inhibited anti-tumor immunity and promoted tumor progression. Meanwhile, it is confirmed that high mobility group box-1 protein (HMGB1) shows a high expression in many solid tumors and HMGB1 with high expression is involved in tumor immune escape. However, the mechanisms linking HMGB1 with tumor immune escape are unclear. The present study aimed to explore whether HMGB1 can promote RCC immune escape by inducing the generation of MDSCs. In this study, Renca mouse model was established and the influence of HMGB1 on MDSCs was investigated by using HMGB1 antibody to downregulate the expression of HMGB1 in tumor-bearing mice. The result showed that with the down-regulation of HMGB1, the tumor growth was inhibited significantly and the mice survival was prolonged greatly. Furthermore, the differentiation and proliferation of MDSCs were inhibited both in vitro and in vivo, and the inhibition rate showed a positive correlation with the degree of down-regulation of HMGB1. When MDSCs were eliminated with Gr-1 antibody in vivo, the ability of the HMGB1 to promote tumor growth was severely impaired. Thus, our findings indicated that HMGB1 might mediate tumor immune escape by promoting MDSCs cell proliferation, which provided a novel theoretical basis for preventing RCC using HMGB1 as the target.
Collapse
Affiliation(s)
- Jinfeng Li
- Kidney Transplantation Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiajia Sun
- Kidney Transplantation Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruiming Rong
- Department of Urology, Zhongshan Hospital and School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Long Li
- Department of Urology, Zhongshan Hospital and School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Wenjun Shang
- Kidney Transplantation Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dongkui Song
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guiwen Feng
- Kidney Transplantation Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Feifei Luo
- Department of Digestive Diseases, Huashan Hospital and Biotherapy Research Center, Fudan University, Shanghai, China
| |
Collapse
|
15
|
Goebel S, Kehlen A, Bluemke K, Altermann W, Schlaf G, Fischer K, Fornara P, Wullich B, Wach S, Taubert H. Differences in the frequencies of HLA-class I and II alleles between German patients with renal cell carcinoma and healthy controls. Cancer Immunol Immunother 2017; 66:565-571. [PMID: 28184970 PMCID: PMC11029131 DOI: 10.1007/s00262-017-1957-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 01/18/2017] [Indexed: 02/06/2023]
Abstract
The human leukocyte antigen (HLA) system is a major part of the human immune system and has an impact on tumor initiation, tumor progression, and immunosurveillance. Renal cell carcinoma tumors are considered to be immunogenic. Therefore, we studied the allele frequencies of four gene loci (HLA-A, -B, -C, and HLA-DR) in a cohort of German renal cell carcinoma (RCC) patients and in healthy controls. HLA-A-C were determined using serological methods, whereas HLA-C12, C14, C16, C18, and HLA-DR were characterized through the use of standard molecular biological methods. The occurrence of the HLA-C*12 allele was significantly increased in German RCC patients compared with healthy controls (P < 0.005; Fisher's exact test), whereas the occurrence of the HLA-DRB1*04 allele was significantly reduced in RCC patients compared with healthy controls (P < 0.05; Fisher's exact test). However, the presence of allele HLA-C*12 was not significantly associated with 10 year overall survival. We suggest that the frequency of HLA alleles can affect development of RCC and could add knowledge as predictive marker for future immunotherapies.
Collapse
Affiliation(s)
- Steffen Goebel
- Institute of Transfusion Medicine, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Astrid Kehlen
- Institute of Medical Microbiology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Karen Bluemke
- Institute of Legal Medicine, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Wolfgang Altermann
- HLA-Laboratory, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Gerald Schlaf
- HLA-Laboratory, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Kersten Fischer
- Clinic of Urology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Paolo Fornara
- Clinic of Urology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Bernd Wullich
- Division Molecular Urology, Department of Urology and Pediatric Urology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Hartmannstr. 14, 91054, Erlangen, Germany
| | - Sven Wach
- Division Molecular Urology, Department of Urology and Pediatric Urology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Hartmannstr. 14, 91054, Erlangen, Germany
| | - Helge Taubert
- Division Molecular Urology, Department of Urology and Pediatric Urology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Hartmannstr. 14, 91054, Erlangen, Germany.
| |
Collapse
|
16
|
|
17
|
Hirayama Y, Gi M, Yamano S, Tachibana H, Okuno T, Tamada S, Nakatani T, Wanibuchi H. Anti-PD-L1 treatment enhances antitumor effect of everolimus in a mouse model of renal cell carcinoma. Cancer Sci 2016; 107:1736-1744. [PMID: 27712020 PMCID: PMC5198964 DOI: 10.1111/cas.13099] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 10/01/2016] [Accepted: 10/04/2016] [Indexed: 02/06/2023] Open
Abstract
Immunotherapy based on blockade of the programmed death-1 (PD-1)/programmed death-ligand 1 (PD-L1) axis has shown promising clinical activity for renal cell carcinoma (RCC) patients; however, the most effective use of these agents in combination with conventional targeted therapy remains to be resolved. Here we evaluated the therapeutic efficacy of the combination of the mTOR inhibitor everolimus (EVE) and anti-PD-L1 using an immunocompetent mouse model of RCC. We first assessed the in vitro effect of EVE on PD-L1 expression in the human 786-O and mouse RENCA RCC cell lines and found that EVE upregulated PD-L1 expression in these RCC cell lines. We then treated RENCA tumor-bearing mice with EVE and found that PD-L1 expression was also increased in tumor cells after EVE treatment. To determine the antitumor effects of EVE alone, anti-PD-L1 alone, and EVE in combination with anti-PD-L1, we evaluated their antitumor effects on RENCA tumor-bearing mice. A significant decrease in the tumor burden was observed in the EVE alone but not in the anti-PD-L1 alone treatment group compared with the control group. Importantly, the combination of EVE with anti-PD-L1 significantly reduced tumor burden compared with the EVE alone treatment, increasing tumor infiltrating lymphocytes (TILs) and the ratio of cytotoxic CD8+ T cells to TILs. The results of the present study demonstrated that anti-PD-L1 treatment enhanced the antitumor effect of EVE in a mouse model, supporting a direct translation of this combination strategy to the clinic for the treatment of RCC.
Collapse
Affiliation(s)
- Yukiyoshi Hirayama
- Department of Molecular Pathology, Osaka City University Graduate School of Medicine, Osaka, Japan.,Department of Urology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Min Gi
- Department of Molecular Pathology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Shotaro Yamano
- Department of Molecular Pathology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Hirokazu Tachibana
- Department of Urology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Takahiro Okuno
- Department of Molecular Pathology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Satoshi Tamada
- Department of Urology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Tatsuya Nakatani
- Department of Urology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Hideki Wanibuchi
- Department of Molecular Pathology, Osaka City University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
18
|
Fu H, Zhu Y, Wang Y, Liu Z, Zhang J, Wang Z, Xie H, Dai B, Xu J, Ye D. Tumor Infiltrating Mast Cells (TIMs) Confers a Marked Survival Advantage in Nonmetastatic Clear-Cell Renal Cell Carcinoma. Ann Surg Oncol 2016; 24:1435-1442. [PMID: 27896514 DOI: 10.1245/s10434-016-5702-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Indexed: 11/18/2022]
Abstract
PURPOSE The role played by the innate immune system in determining the clinical outcome of clear-cell renal cell carcinoma (ccRCC) was still blurred. This study was designed to investigate the prognostic significance of tumor infiltrating mast cells (TIMs) in ccRCC. METHODS The study retrospectively enrolled a training set (474 patients) and a validation set (188 patients) with nonmetastasis (pT1-4N0M0) ccRCC from two institutional medical centers of China. TIMs was evaluated by immunohistochemical staining of tryptase and its association with clinicopathologic features and prognosis were evaluated. RESULTS In ccRCC tissues, TIMs ranged from 0 to 103 cells/mm2 and 0 to 113 cells/mm2 in the training set and validation set, respectively. TIMs was negatively correlated with tumor size (P < 0.001 and P < 0.001, respectively), pathological T stage (P = 0.005 and P = 0.007, respectively) and Fuhrman grade (P < 0.001 and P < 0.001, respectively). Patients with abundant TIMs infiltration showed significantly longer cancer-specific survival in the training cohort and the validation cohort (P < 0.001 and P < 0.001). Patients with abundant mast cell infiltration showed significantly longer overall survival in the TCGA cohort (P < 0.001). Moreover, multivariate analysis identified TIMs as an independent prognostic factor for cancer-specific survival (CSS) and relapse-free survival (RFS). Also, TIMs was significantly correlated with CSS and RFS of the mediate and high-risk patients in the training cohort and the validation cohort. CONCLUSIONS TIMs density is a powerful independent prognostic factor for CSS and RFS in patients with nonmetastasis (pT1-4N0M0) ccRCC.
Collapse
Affiliation(s)
- Hangcheng Fu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yiwei Wang
- Department of Urology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Liu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Junyu Zhang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zewei Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Huyang Xie
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bo Dai
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiejie Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
19
|
Farolfi A, Schepisi G, Conteduca V, Burgio SL, Lolli C, De Giorgi U. Pharmacokinetics, pharmacodynamics and clinical efficacy of nivolumab in the treatment of metastatic renal cell carcinoma. Expert Opin Drug Metab Toxicol 2016; 12:1089-96. [PMID: 27450183 DOI: 10.1080/17425255.2016.1214713] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Nivolumab is a recombinant, humanized monoclonal antibody that binds PD-1. The binding of PD-1 with PD-L1, expressed on antigen-presenting cells and tumor cells, suppresses the ability of T-lymphocytes to recognize and destroy tumor cells. Nivolumab reverts this inhibitory signal and has led to a significant prolongation of overall survival in patients with metastatic renal cell carcinoma (RCC). AREAS COVERED The rationale for immunotherapy in metastatic RCC, key immune checkpoint pathways, nivolumab pharmacodynamics, results from the main clinical trials, and predictors of response are discussed. EXPERT OPINION Nivolumab demonstrated a statistically significant advantage over everolimus in overall survival in metastatic RCC patients after first-line antiangiogenic therapy. Nevertheless, a number of issues remain to be resolved regarding the use of this drug in RCC. It is now imperative to identify which patients can benefit most from immunotherapy and studies are ongoing to define its role in other settings and/or in combinations with antiCTLA4 or antiangiogenic drugs.
Collapse
Affiliation(s)
- Alberto Farolfi
- a Department of Medical Oncology , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Giuseppe Schepisi
- a Department of Medical Oncology , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Vincenza Conteduca
- a Department of Medical Oncology , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Salvatore Luca Burgio
- a Department of Medical Oncology , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Cristian Lolli
- a Department of Medical Oncology , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Ugo De Giorgi
- a Department of Medical Oncology , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| |
Collapse
|
20
|
Ratta R, Zappasodi R, Raggi D, Grassi P, Verzoni E, Necchi A, Di Nicola M, Salvioni R, de Braud F, Procopio G. Immunotherapy advances in uro-genital malignancies. Crit Rev Oncol Hematol 2016; 105:52-64. [PMID: 27372200 DOI: 10.1016/j.critrevonc.2016.06.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 05/27/2016] [Accepted: 06/16/2016] [Indexed: 12/27/2022] Open
Abstract
Immunotherapy for the treatment of cancer has made significant progresses over the last 20 years. Multiple efforts have been attempted to restore immune-mediated tumor elimination, leading to the development of several targeted immunotherapies. Data from recent clinical trials suggest that these agents might improve the prognosis of patients with advanced genito-urinary (GU) malignancies. Nivolumab has been the first immune checkpoint-inhibitor approved for pre-treated patients with metastatic renal cell carcinoma. Pembrolizumab and atezolizumab have shown promising results in both phase I and II trials in urothelial carcinoma. Brentuximab vedotin has demonstrated early signals of clinical activity and immunomodulatory effects in highly pre-treated patients with testicular germ cell tumors. In this review, we have summarized the major clinical achievements of immunotherapy in GU cancers, focusing on immune checkpoint blockade as well as the new immunomodulatory monoclonal antibodies (mAbs) under clinical evaluation for these malignancies.
Collapse
Affiliation(s)
- Raffaele Ratta
- Department of Medical Oncology, Fondazione IRCSS Istituto Nazionale Tumori, Milan, Italy
| | - Roberta Zappasodi
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daniele Raggi
- Department of Medical Oncology, Fondazione IRCSS Istituto Nazionale Tumori, Milan, Italy
| | - Paolo Grassi
- Department of Medical Oncology, Fondazione IRCSS Istituto Nazionale Tumori, Milan, Italy
| | - Elena Verzoni
- Department of Medical Oncology, Fondazione IRCSS Istituto Nazionale Tumori, Milan, Italy
| | - Andrea Necchi
- Department of Medical Oncology, Fondazione IRCSS Istituto Nazionale Tumori, Milan, Italy
| | - Massimo Di Nicola
- Unit of Immunotherapy and Anticancer Innovative Therapeutics, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Roberto Salvioni
- Department of Surgery, Urology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo de Braud
- Department of Medical Oncology, Fondazione IRCSS Istituto Nazionale Tumori, Milan, Italy
| | - Giuseppe Procopio
- Department of Medical Oncology, Fondazione IRCSS Istituto Nazionale Tumori, Milan, Italy.
| |
Collapse
|
21
|
Kourie HR, Klastersky J. Immune checkpoint inhibitors side effects and management. Immunotherapy 2016; 8:799-807. [DOI: 10.2217/imt-2016-0029] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The next decade in cancer therapy will be marked by the expansion of immunotherapies, namely immune checkpoint inhibitors. The increasing number and combination of checkpoint inhibitors and the variety of their mechanisms of action and indications will most likely multiply the side effects associated with these therapies and make their management more complicated and diversified. Given the growing rate of approval of different checkpoint inhibitors in different cancers in multiple settings, a review summarizing the major side effects of the new agents in use today and their management seems to be appropriate. Highlighting these adverse events and their management in a single review might help the daily practice of the physicians and consequently contribute the patient's safety and quality of life.
Collapse
Affiliation(s)
- Hampig Raphael Kourie
- Institut Jules Bordet, Centre des Tumeurs de l'Université Libre de Bruxelles, Bruxelles, Belgium
| | - Jean Klastersky
- Institut Jules Bordet, Centre des Tumeurs de l'Université Libre de Bruxelles, Bruxelles, Belgium
| |
Collapse
|
22
|
Park YH, Kim YJ, Kang SH, Kim HH, Byun SS, Lee JY, Hong SH. Association between Perioperative Blood Transfusion and Oncologic Outcomes after Curative Surgery for Renal Cell Carcinoma. J Cancer 2016; 7:965-72. [PMID: 27313787 PMCID: PMC4910589 DOI: 10.7150/jca.15073] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 03/21/2016] [Indexed: 01/26/2023] Open
Abstract
Purpose: We aimed to elucidate the association between perioperative blood transfusion (PBT) and the prognosis of patients undergoing curative surgery for renal cell carcinoma (RCC). Methods: In all, 3,832 patients with RCC who had undergone curative surgery were included in this study from a multicenter database. PBT was defined as the transfusion of packed red blood cells within seven days before surgery, during surgery, or within the postoperative hospitalization period. The association of PBT with oncologic outcomes was evaluated using univariate and multivariate Cox regression analyses, and regression adjustment with propensity score matching. Results: Overall, 11.7% (447/3,832) of patients received PBT. Patients receiving PBT were significantly older at diagnosis, and had lower BMI, higher comorbidities, worse ECOG performance status, and more initial symptoms. Moreover, higher pathologic TNM stage, larger mass size, higher nuclear grade, more sarcomatoid differentiation, and more tumor necrosis were all observed more frequently in patients who received PBT. In univariate analysis, relapse-free survival, cancer-specific survival, and overall survival rates were worse in patients who received PBT; however, these factors became insignificant in the matched pairs after propensity score matching. On multivariate Cox regression analysis and regression adjustment with propensity score matching, significant prognostic effects of PBT on disease relapse, cancer-specific mortality, and all-cause mortality were not observed. Conclusions: This multicenter database analysis demonstrates no significant prognostic association between PBT and oncologic outcomes in patients with RCC.
Collapse
Affiliation(s)
- Yong Hyun Park
- 1. Department of Urology, Seoul St. Mary's Hospital, The Catholic University, Seoul, Korea
| | - Yong-June Kim
- 2. Department of Urology, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Seok Ho Kang
- 3. Department of Urology, Korea University School of Medicine, Seoul, Korea
| | - Hyeon Hoe Kim
- 4. Department of Urology, Seoul National University College of Medicine, Seoul, Korea
| | - Seok-Soo Byun
- 5. Department of Urology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Ji Youl Lee
- 1. Department of Urology, Seoul St. Mary's Hospital, The Catholic University, Seoul, Korea
| | - Sung-Hoo Hong
- 1. Department of Urology, Seoul St. Mary's Hospital, The Catholic University, Seoul, Korea
| |
Collapse
|
23
|
Matsushita H, Sato Y, Karasaki T, Nakagawa T, Kume H, Ogawa S, Homma Y, Kakimi K. Neoantigen Load, Antigen Presentation Machinery, and Immune Signatures Determine Prognosis in Clear Cell Renal Cell Carcinoma. Cancer Immunol Res 2016; 4:463-71. [PMID: 26980598 DOI: 10.1158/2326-6066.cir-15-0225] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 02/05/2016] [Indexed: 11/16/2022]
Abstract
Tumors commonly harbor multiple genetic alterations, some of which initiate tumorigenesis. Among these, some tumor-specific somatic mutations resulting in mutated protein have the potential to induce antitumor immune responses. To examine the relevance of the latter to immune responses in the tumor and to patient outcomes, we used datasets of whole-exome and RNA sequencing from 97 clear cell renal cell carcinoma (ccRCC) patients to identify neoepitopes predicted to be presented by each patient's autologous HLA molecules. We found that the number of nonsilent or missense mutations did not correlate with patient prognosis. However, combining the number of HLA-restricted neoepitopes with the cell surface expression of HLA or β2-microglobulin(β2M) revealed that an A-neo(hi)/HLA-A(hi) or ABC-neo(hi)/β2M(hi) phenotype correlated with better clinical outcomes. Higher expression of immune-related genes from CD8 T cells and their effector molecules [CD8A, perforin (PRF1) and granzyme A (GZMA)], however, did not correlate with prognosis. This may have been due to the observed correlation of these genes with the expression of other genes that were associated with immunosuppression in the tumor microenvironment (CTLA-4, PD-1, LAG-3, PD-L1, PD-L2, IDO1, and IL10). This suggested that abundant neoepitopes associated with greater antitumor effector immune responses were counterbalanced by a strongly immunosuppressive microenvironment. Therefore, immunosuppressive molecules should be considered high-priority targets for modulating immune responses in patients with ccRCC. Blockade of these molecular pathways could be combined with immunotherapies targeting neoantigens to achieve synergistic antitumor activity. Cancer Immunol Res; 4(5); 463-71. ©2016 AACR.
Collapse
Affiliation(s)
- Hirokazu Matsushita
- Department of Immunotherapeutics, The University of Tokyo Hospital, Tokyo, Japan
| | - Yusuke Sato
- Department of Urology, The University of Tokyo Hospital, Tokyo, Japan. Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takahiro Karasaki
- Department of Immunotherapeutics, The University of Tokyo Hospital, Tokyo, Japan
| | - Tohru Nakagawa
- Department of Urology, The University of Tokyo Hospital, Tokyo, Japan
| | - Haruki Kume
- Department of Urology, The University of Tokyo Hospital, Tokyo, Japan
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yukio Homma
- Department of Urology, The University of Tokyo Hospital, Tokyo, Japan
| | - Kazuhiro Kakimi
- Department of Immunotherapeutics, The University of Tokyo Hospital, Tokyo, Japan.
| |
Collapse
|
24
|
McDermott DF, Sosman JA, Sznol M, Massard C, Gordon MS, Hamid O, Powderly JD, Infante JR, Fassò M, Wang YV, Zou W, Hegde PS, Fine GD, Powles T. Atezolizumab, an Anti-Programmed Death-Ligand 1 Antibody, in Metastatic Renal Cell Carcinoma: Long-Term Safety, Clinical Activity, and Immune Correlates From a Phase Ia Study. J Clin Oncol 2016; 34:833-42. [PMID: 26755520 DOI: 10.1200/jco.2015.63.7421] [Citation(s) in RCA: 440] [Impact Index Per Article: 48.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
PURPOSE The objective was to determine the safety and clinical activity of atezolizumab (MPDL3280A), a humanized programmed death-ligand 1 (PD-L1) antibody, in renal cell carcinoma (RCC). Exploratory biomarkers were analyzed and associated with outcomes. PATIENTS AND METHODS Seventy patients with metastatic RCC, including clear cell (ccRCC; n = 63) and non-clear cell (ncc; n = 7) histologies, received atezolizumab intravenously every 3 weeks. PD-L1 expression was scored at four diagnostic levels (0/1/2/3) that were based on PD-L1 staining on tumor cells and tumor-infiltrating immune cells (IC) with the SP142 assay. Primary end points were safety and toxicity; secondary end points assessed clinical activity per Response Evaluation Criteria in Solid Tumors version 1.1 and immune-related response criteria. Plasma and tissue were analyzed for potential biomarkers of atezolizumab response. RESULTS Grade 3 treatment-related and immune-mediated adverse events occurred in 17% and 4% of patients, respectively, and there were no grade 4 or 5 events. Sixty-three patients with ccRCC were evaluable for overall survival (median, 28.9 months; 95% CI, 20.0 months to not reached) and progression-free survival (median, 5.6 months; 95% CI, 3.9 to 8.2 months), and 62 patients were evaluable for objective response rate (ORR; 15%; 95% CI, 7% to 26%). ORR was evaluated on the basis of PD-L1 IC expression (IC1/2/3: n = 33; 18%; 95% CI, 7% to 35%; and IC0: n = 22; 9%; 95% CI, 1% to 29%). The ORR for patients with Fuhrman grade 4 and/or sarcomatoid histology was 22% (n = 18; 95% CI, 6% to 48%). Decreases in circulating plasma markers and acute-phase proteins and an increased baseline effector T-cell-to-regulatory T-cell gene expression ratio correlated with response to atezolizumab. CONCLUSION Atezolizumab demonstrated a manageable safety profile and promising antitumor activity in patients with metastatic RCC. Correlative studies identified potential predictive and pharmacodynamic biomarkers. These results have guided ongoing studies and combinations with atezolizumab in RCC.
Collapse
Affiliation(s)
- David F McDermott
- David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Jeffrey A. Sosman, Vanderbilt University School of Medicine; Jeffrey R. Infante, Sarah Cannon Research Institute, Nashville, TN; Mario Sznol, Yale Cancer Center, New Haven, CT; Christophe Massard, Gustave Roussy, Villejuif Cedex, France; Michael S. Gordon, Pinnacle Oncology Hematology, Scottsdale, AZ; Omid Hamid, Angeles Clinic and Research Institute, Santa Monica; Marcella Fassò, Yan V. Wang, Wei Zou, Priti S. Hedge, and Gregg D. Fine, Genentech, South San Francisco, CA; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; and Thomas Powles, Barts Cancer Institute Centre for Experimental Cancer Medicine and the Royal Free National Health Service Trust, Queen Mary University of London, London, United Kingdom.
| | - Jeffrey A Sosman
- David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Jeffrey A. Sosman, Vanderbilt University School of Medicine; Jeffrey R. Infante, Sarah Cannon Research Institute, Nashville, TN; Mario Sznol, Yale Cancer Center, New Haven, CT; Christophe Massard, Gustave Roussy, Villejuif Cedex, France; Michael S. Gordon, Pinnacle Oncology Hematology, Scottsdale, AZ; Omid Hamid, Angeles Clinic and Research Institute, Santa Monica; Marcella Fassò, Yan V. Wang, Wei Zou, Priti S. Hedge, and Gregg D. Fine, Genentech, South San Francisco, CA; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; and Thomas Powles, Barts Cancer Institute Centre for Experimental Cancer Medicine and the Royal Free National Health Service Trust, Queen Mary University of London, London, United Kingdom
| | - Mario Sznol
- David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Jeffrey A. Sosman, Vanderbilt University School of Medicine; Jeffrey R. Infante, Sarah Cannon Research Institute, Nashville, TN; Mario Sznol, Yale Cancer Center, New Haven, CT; Christophe Massard, Gustave Roussy, Villejuif Cedex, France; Michael S. Gordon, Pinnacle Oncology Hematology, Scottsdale, AZ; Omid Hamid, Angeles Clinic and Research Institute, Santa Monica; Marcella Fassò, Yan V. Wang, Wei Zou, Priti S. Hedge, and Gregg D. Fine, Genentech, South San Francisco, CA; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; and Thomas Powles, Barts Cancer Institute Centre for Experimental Cancer Medicine and the Royal Free National Health Service Trust, Queen Mary University of London, London, United Kingdom
| | - Christophe Massard
- David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Jeffrey A. Sosman, Vanderbilt University School of Medicine; Jeffrey R. Infante, Sarah Cannon Research Institute, Nashville, TN; Mario Sznol, Yale Cancer Center, New Haven, CT; Christophe Massard, Gustave Roussy, Villejuif Cedex, France; Michael S. Gordon, Pinnacle Oncology Hematology, Scottsdale, AZ; Omid Hamid, Angeles Clinic and Research Institute, Santa Monica; Marcella Fassò, Yan V. Wang, Wei Zou, Priti S. Hedge, and Gregg D. Fine, Genentech, South San Francisco, CA; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; and Thomas Powles, Barts Cancer Institute Centre for Experimental Cancer Medicine and the Royal Free National Health Service Trust, Queen Mary University of London, London, United Kingdom
| | - Michael S Gordon
- David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Jeffrey A. Sosman, Vanderbilt University School of Medicine; Jeffrey R. Infante, Sarah Cannon Research Institute, Nashville, TN; Mario Sznol, Yale Cancer Center, New Haven, CT; Christophe Massard, Gustave Roussy, Villejuif Cedex, France; Michael S. Gordon, Pinnacle Oncology Hematology, Scottsdale, AZ; Omid Hamid, Angeles Clinic and Research Institute, Santa Monica; Marcella Fassò, Yan V. Wang, Wei Zou, Priti S. Hedge, and Gregg D. Fine, Genentech, South San Francisco, CA; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; and Thomas Powles, Barts Cancer Institute Centre for Experimental Cancer Medicine and the Royal Free National Health Service Trust, Queen Mary University of London, London, United Kingdom
| | - Omid Hamid
- David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Jeffrey A. Sosman, Vanderbilt University School of Medicine; Jeffrey R. Infante, Sarah Cannon Research Institute, Nashville, TN; Mario Sznol, Yale Cancer Center, New Haven, CT; Christophe Massard, Gustave Roussy, Villejuif Cedex, France; Michael S. Gordon, Pinnacle Oncology Hematology, Scottsdale, AZ; Omid Hamid, Angeles Clinic and Research Institute, Santa Monica; Marcella Fassò, Yan V. Wang, Wei Zou, Priti S. Hedge, and Gregg D. Fine, Genentech, South San Francisco, CA; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; and Thomas Powles, Barts Cancer Institute Centre for Experimental Cancer Medicine and the Royal Free National Health Service Trust, Queen Mary University of London, London, United Kingdom
| | - John D Powderly
- David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Jeffrey A. Sosman, Vanderbilt University School of Medicine; Jeffrey R. Infante, Sarah Cannon Research Institute, Nashville, TN; Mario Sznol, Yale Cancer Center, New Haven, CT; Christophe Massard, Gustave Roussy, Villejuif Cedex, France; Michael S. Gordon, Pinnacle Oncology Hematology, Scottsdale, AZ; Omid Hamid, Angeles Clinic and Research Institute, Santa Monica; Marcella Fassò, Yan V. Wang, Wei Zou, Priti S. Hedge, and Gregg D. Fine, Genentech, South San Francisco, CA; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; and Thomas Powles, Barts Cancer Institute Centre for Experimental Cancer Medicine and the Royal Free National Health Service Trust, Queen Mary University of London, London, United Kingdom
| | - Jeffrey R Infante
- David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Jeffrey A. Sosman, Vanderbilt University School of Medicine; Jeffrey R. Infante, Sarah Cannon Research Institute, Nashville, TN; Mario Sznol, Yale Cancer Center, New Haven, CT; Christophe Massard, Gustave Roussy, Villejuif Cedex, France; Michael S. Gordon, Pinnacle Oncology Hematology, Scottsdale, AZ; Omid Hamid, Angeles Clinic and Research Institute, Santa Monica; Marcella Fassò, Yan V. Wang, Wei Zou, Priti S. Hedge, and Gregg D. Fine, Genentech, South San Francisco, CA; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; and Thomas Powles, Barts Cancer Institute Centre for Experimental Cancer Medicine and the Royal Free National Health Service Trust, Queen Mary University of London, London, United Kingdom
| | - Marcella Fassò
- David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Jeffrey A. Sosman, Vanderbilt University School of Medicine; Jeffrey R. Infante, Sarah Cannon Research Institute, Nashville, TN; Mario Sznol, Yale Cancer Center, New Haven, CT; Christophe Massard, Gustave Roussy, Villejuif Cedex, France; Michael S. Gordon, Pinnacle Oncology Hematology, Scottsdale, AZ; Omid Hamid, Angeles Clinic and Research Institute, Santa Monica; Marcella Fassò, Yan V. Wang, Wei Zou, Priti S. Hedge, and Gregg D. Fine, Genentech, South San Francisco, CA; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; and Thomas Powles, Barts Cancer Institute Centre for Experimental Cancer Medicine and the Royal Free National Health Service Trust, Queen Mary University of London, London, United Kingdom
| | - Yan V Wang
- David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Jeffrey A. Sosman, Vanderbilt University School of Medicine; Jeffrey R. Infante, Sarah Cannon Research Institute, Nashville, TN; Mario Sznol, Yale Cancer Center, New Haven, CT; Christophe Massard, Gustave Roussy, Villejuif Cedex, France; Michael S. Gordon, Pinnacle Oncology Hematology, Scottsdale, AZ; Omid Hamid, Angeles Clinic and Research Institute, Santa Monica; Marcella Fassò, Yan V. Wang, Wei Zou, Priti S. Hedge, and Gregg D. Fine, Genentech, South San Francisco, CA; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; and Thomas Powles, Barts Cancer Institute Centre for Experimental Cancer Medicine and the Royal Free National Health Service Trust, Queen Mary University of London, London, United Kingdom
| | - Wei Zou
- David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Jeffrey A. Sosman, Vanderbilt University School of Medicine; Jeffrey R. Infante, Sarah Cannon Research Institute, Nashville, TN; Mario Sznol, Yale Cancer Center, New Haven, CT; Christophe Massard, Gustave Roussy, Villejuif Cedex, France; Michael S. Gordon, Pinnacle Oncology Hematology, Scottsdale, AZ; Omid Hamid, Angeles Clinic and Research Institute, Santa Monica; Marcella Fassò, Yan V. Wang, Wei Zou, Priti S. Hedge, and Gregg D. Fine, Genentech, South San Francisco, CA; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; and Thomas Powles, Barts Cancer Institute Centre for Experimental Cancer Medicine and the Royal Free National Health Service Trust, Queen Mary University of London, London, United Kingdom
| | - Priti S Hegde
- David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Jeffrey A. Sosman, Vanderbilt University School of Medicine; Jeffrey R. Infante, Sarah Cannon Research Institute, Nashville, TN; Mario Sznol, Yale Cancer Center, New Haven, CT; Christophe Massard, Gustave Roussy, Villejuif Cedex, France; Michael S. Gordon, Pinnacle Oncology Hematology, Scottsdale, AZ; Omid Hamid, Angeles Clinic and Research Institute, Santa Monica; Marcella Fassò, Yan V. Wang, Wei Zou, Priti S. Hedge, and Gregg D. Fine, Genentech, South San Francisco, CA; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; and Thomas Powles, Barts Cancer Institute Centre for Experimental Cancer Medicine and the Royal Free National Health Service Trust, Queen Mary University of London, London, United Kingdom
| | - Gregg D Fine
- David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Jeffrey A. Sosman, Vanderbilt University School of Medicine; Jeffrey R. Infante, Sarah Cannon Research Institute, Nashville, TN; Mario Sznol, Yale Cancer Center, New Haven, CT; Christophe Massard, Gustave Roussy, Villejuif Cedex, France; Michael S. Gordon, Pinnacle Oncology Hematology, Scottsdale, AZ; Omid Hamid, Angeles Clinic and Research Institute, Santa Monica; Marcella Fassò, Yan V. Wang, Wei Zou, Priti S. Hedge, and Gregg D. Fine, Genentech, South San Francisco, CA; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; and Thomas Powles, Barts Cancer Institute Centre for Experimental Cancer Medicine and the Royal Free National Health Service Trust, Queen Mary University of London, London, United Kingdom
| | - Thomas Powles
- David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Jeffrey A. Sosman, Vanderbilt University School of Medicine; Jeffrey R. Infante, Sarah Cannon Research Institute, Nashville, TN; Mario Sznol, Yale Cancer Center, New Haven, CT; Christophe Massard, Gustave Roussy, Villejuif Cedex, France; Michael S. Gordon, Pinnacle Oncology Hematology, Scottsdale, AZ; Omid Hamid, Angeles Clinic and Research Institute, Santa Monica; Marcella Fassò, Yan V. Wang, Wei Zou, Priti S. Hedge, and Gregg D. Fine, Genentech, South San Francisco, CA; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; and Thomas Powles, Barts Cancer Institute Centre for Experimental Cancer Medicine and the Royal Free National Health Service Trust, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
25
|
Chen S, Liang L, Wang Y, Diao J, Zhao C, Chen G, He Y, Luo C, Wu X, Zhang Y. Synergistic immunotherapeutic effects of Lycium barbarum polysaccharide and interferon-α2b on the murine Renca renal cell carcinoma cell line in vitro and in vivo. Mol Med Rep 2015; 12:6727-37. [PMID: 26300071 PMCID: PMC4626139 DOI: 10.3892/mmr.2015.4230] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 07/28/2015] [Indexed: 12/15/2022] Open
Abstract
Novel therapeutic strategies to improve clinical efficacy in patients with renal cell carcinoma (RCC) are required. The possibility of combination therapy with Lycium barbarum polysaccharides (LBP) and recombinant interferon (IFN)-α2b remains to be elucidated in RCC. The present study investigated the putative synergistic immunotherapeutic roles of LBP and IFN-α2b against RCC in vitro and in vivo. The mouse RCC cell line, Renca, was used for in vitro experiments. Treatment of the cells with a combination of LBP and IFN-α2b markedly inhibited cell proliferation, retarded cell cycle growth and promoted apoptosis in the Renca cells. Western blot analysis revealed that LBP and IFN-α2b synergistically downregulated the expression levels of cyclin D1, c-Myc and Bcl-2, and upregulated the expression of the antiapoptotic protein, Bax. Myeloid-derived suppressor cells (MDSCs) were markedly upregulated during tumour progression and promoted tumour growth by inhibiting the T-cell-mediated immune response. In vivo, a marked reduction in the MDSC ratio and tumour volume was observed in a group receiving combined treatment with LBP and IFN-α2b in a xenograft tumour model. In conclusion, the present study suggested that the combination of LBP and IFN-α2b is likely to be more effective in treating murine RCC compared with the less pronounced immunotherapeutic effects of administering LBP or IFN-α2b alone.
Collapse
Affiliation(s)
- Shiyou Chen
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Lunan Liang
- Department of Reproductive Medicine, The Affiliated Hospital of Jining Medical College, Jining, Shandong 272000, P.R. China
| | - Ying Wang
- Department of Oncology, The First People's Hospital of Jiulongpo District, Chongqing 400050, P.R. China
| | - Jianhun Diao
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Chunxiong Zhao
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Gang Chen
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yunfeng He
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Chunli Luo
- Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xiaohou Wu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yao Zhang
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
26
|
Kamińska K, Czarnecka AM, Escudier B, Lian F, Szczylik C. Interleukin-6 as an emerging regulator of renal cell cancer. Urol Oncol 2015; 33:476-85. [PMID: 26296264 DOI: 10.1016/j.urolonc.2015.07.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 07/10/2015] [Accepted: 07/11/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Our knowledge on the molecular basis of kidney cancer metastasisis still relatively low. About 25-30% of patients suffering from clear cell renal cell carcinoma (ccRCC)present metastatic disease at the time of primary diagnosis. Only 10% of patients diagnosed with stage IV disease survive 5 years and 20-50% of patients diagnosed with localized tumor develop metastases within 3 years. High mortality of patients with this cancer is associated with a large potential for metastasis and resistance to oncologic treatments such as chemo- and radiotherapy. Literature data based on studies conducted on other types of cancers suggest that in metastatic ccRCC, the complex of interleukin-6 (IL-6) and its soluble receptor (sIL-6R; complex IL-6/sIL-6R) and the signal transduction pathway (gp130/STAT3) might play a key role in this process. PURPOSE Therefore, in this review we focus on the role of IL-6 and its signaling pathways as a factor for development and spread of RCC. Analyzing the molecular basis of cancer spreading will enable the development of prognostic tests, evaluate individual predisposition for metastasis, and produce drugs that target metastases. As the development of effective systemic treatments evolve from advancements in molecular biology, continued studies directed at understanding the genetic and molecular complexities of this disease are critical to improve RCC treatment options.
Collapse
Affiliation(s)
| | - Anna M Czarnecka
- Department of Oncology, Military Institute of Medicine, Warsaw, Poland
| | - Bernard Escudier
- Medical Oncology Department, Institut Gustave Roussy, Villejuif, France
| | - Fei Lian
- Emory University School of Medicine, Atlanta, GA
| | - Cezary Szczylik
- Department of Oncology, Military Institute of Medicine, Warsaw, Poland
| |
Collapse
|
27
|
Zakharia Y, Zakharia K, Rixe O. Axitinib: from preclinical development to future clinical perspectives in renal cell carcinoma. Expert Opin Drug Discov 2015; 10:925-35. [DOI: 10.1517/17460441.2015.1045411] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Yousef Zakharia
- 1University of Iowa Division of Hematology/Oncology and Holden Comprehensive Cancer Center, Iowa City, Iowa, USA
| | - Kais Zakharia
- 2Mayo Clinic College of Medicine - Division of Gastroenterology and Hepatology, Rochester, Minnesota, USA
| | - Olivier Rixe
- 3University of New Mexico Cancer Center, Division of Hematology/Oncology, Albuquerque, New Mexico, USA
| |
Collapse
|
28
|
Bergmann L, Brugger W, Herr W, Mackensen A, Multhoff G. [What opportunities does Immuno-oncology indicate for overarching long-term survival?]. Oncol Res Treat 2015; 38 Suppl 3:6-11. [PMID: 25966812 DOI: 10.1159/000381363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Lothar Bergmann
- Medizinische Klinik II: Hämatologie/Onkologie, Universitätsklinikum Frankfurt, Frankfurt/M., Deutschland
| | | | | | | | | |
Collapse
|
29
|
Swaika A, Hammond WA, Joseph RW. Current state of anti-PD-L1 and anti-PD-1 agents in cancer therapy. Mol Immunol 2015; 67:4-17. [PMID: 25749122 DOI: 10.1016/j.molimm.2015.02.009] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 02/05/2015] [Accepted: 02/08/2015] [Indexed: 02/07/2023]
Abstract
Immunotherapy for the treatment of cancer is rapidly evolving from therapies that globally and non-specifically simulate the immune system to more targeted activation of individual components of the immune system. The net result of this targeted approach is decreased toxicity and increased efficacy of immunotherapy. More specifically, therapies that inhibit the interaction between programmed death ligand 1 (PD-L1), present on the surface of tumor or antigen-presenting cells, and programmed death 1 (PD-1), present on the surface of activated lymphocytes, are generating much excitement and enthusiasm, even in malignancies that are not traditionally considered to be immunogenic. Herein, we review the current landscape of anti-PD-1 and anti-PD-L1 therapies in the world of oncology. We have performed a comprehensive literature search on the data available through PubMed, Medline, Scopus, the ClinicalTrials.gov registry, and abstracts from major oncology meetings in order to summarize the clinical data of anti-PD-1/PD-L1 therapies.
Collapse
Affiliation(s)
- Abhisek Swaika
- Division of Hematology and Oncology, Mayo Clinic, 4500 San Pablo Rd., Jacksonville, FL, USA
| | - William A Hammond
- Division of Hematology and Oncology, Mayo Clinic, 4500 San Pablo Rd., Jacksonville, FL, USA
| | - Richard W Joseph
- Division of Hematology and Oncology, Mayo Clinic, 4500 San Pablo Rd., Jacksonville, FL, USA.
| |
Collapse
|
30
|
Massari F, Santoni M, Ciccarese C, Santini D, Alfieri S, Martignoni G, Brunelli M, Piva F, Berardi R, Montironi R, Porta C, Cascinu S, Tortora G. PD-1 blockade therapy in renal cell carcinoma: current studies and future promises. Cancer Treat Rev 2015; 41:114-121. [PMID: 25586601 DOI: 10.1016/j.ctrv.2014.12.013] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 12/30/2014] [Indexed: 10/24/2022]
Abstract
RCC is considered an immunogenic tumor with a prominent dysfunctional immune cell infiltrate, unable to control tumor growth. Evasion of immune surveillance, a process defined immune-editing, leads to malignant progression. The striking improvement of knowledge in immunology has led to the identification of immune checkpoints (such as CTLA-4 and PD-1), whose blockage enhances the antitumor immunity. The interaction between PD-1, an inducible inhibitory receptor expressed on lymphocytes and DCs, and PD-L1 ligand, expressed by tumor cells, results in a down-regulation of the T-cell response. Therefore, the PD-1/PD-L1 axis inhibition by targeted-antibodies, increasing the T-cell proliferation and cytotoxicity, represents a promising mechanism to stimulate the anti-tumor activity of the immune system, improving the outcomes of cancer patients. Several PD-1 and PD-L1 inhibitors have been evaluated in different tumor types, showing promising results. The interesting correlation between lymphocytes PD-1 expression and RCC advanced stage, grade and prognosis, as well as the selective PD-L1 expression by RCC tumor cells and its potential association with worse clinical outcomes, have led to the development of new anti PD-1/PD-L1 agents, alone or in combination with anti-angiogenic drugs or other immunotherapeutic approaches, for the treatment of RCC. In this review we discuss the role of PD-1/PD-L1 in RCC, focusing on the biological rationale, current clinical studies and promising therapeutic perspectives to target the PD-1 pathway.
Collapse
Affiliation(s)
- F Massari
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Piazzale L.A. Scuro 10, 37124 Verona, Italy
| | - M Santoni
- Clinica di Oncologia Medica, AOU "Ospedali Riuniti", Polytechnic University of the Marche Region, via Conca 71, 60126 Ancona, Italy.
| | - C Ciccarese
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Piazzale L.A. Scuro 10, 37124 Verona, Italy
| | - D Santini
- Department of Medical Oncology, University Campus Bio-Medico Roma, Oncologia Medica, Rome, Italy
| | - S Alfieri
- SSD Oncologia medica Tumori Testa e Collo, Fondazione IRCCS - Istituto Nazionale dei Tumori, Via Venezian, 1, 20133 Milano, Italy
| | - G Martignoni
- Department of Pathology and Diagnostic, Azienda Ospedaliera Universitaria Integrata, University of Verona, Piazzale L.A. Scuro 10, 37124 Verona, Italy
| | - M Brunelli
- Department of Pathology and Diagnostic, Azienda Ospedaliera Universitaria Integrata, University of Verona, Piazzale L.A. Scuro 10, 37124 Verona, Italy
| | - F Piva
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, 60131 Ancona, Italy
| | - R Berardi
- Clinica di Oncologia Medica, AOU "Ospedali Riuniti", Polytechnic University of the Marche Region, via Conca 71, 60126 Ancona, Italy
| | - R Montironi
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | - C Porta
- Medical Oncology, IRCCS San Matteo University Hospital Foundation, piazzale C. Golgi 19, 27100 Pavia, Italy
| | - S Cascinu
- Clinica di Oncologia Medica, AOU "Ospedali Riuniti", Polytechnic University of the Marche Region, via Conca 71, 60126 Ancona, Italy
| | - G Tortora
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Piazzale L.A. Scuro 10, 37124 Verona, Italy
| |
Collapse
|
31
|
Amenta V, Aschberger K. Carbon nanotubes: potential medical applications and safety concerns. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2014; 7:371-86. [PMID: 25429905 DOI: 10.1002/wnan.1317] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 09/29/2014] [Accepted: 10/11/2014] [Indexed: 11/06/2022]
Abstract
Carbon nanotubes (CNTs) have unique atomic structure, as well as outstanding thermal, mechanical, and electronic properties, making them extremely attractive materials for several different applications. Many research groups are focusing on biomedical applications of carbon-based nanomaterials, however the application of CNTs to the biomedical field is not developing as fast as in other areas. While CNTs-based products are already being used in textiles, polymer matrices to strengthen materials, sports articles, microelectronics, energy storage, etc., medicinal products and medical devices for in vivo application based on CNTs have not been commercialized yet. However, CNTs for biomedical application, i.e., CNTs conjugated to siRNA for cancer therapy, or CNTs for imaging of colorectal cancer and many other products may enter clinical trials in the next years. Concerns related to the toxicity of CNTs must be overcome in order to have these products commercialized in a near future. This article reviews emerging biomedical applications of CNTs, specifically for therapy. It also deals with challenges associated with possible medical applications of CNTs, such as their not fully understood toxicological profile in the human body.
Collapse
Affiliation(s)
- Valeria Amenta
- European Commission, Joint Research Centre, Institute for Health and Consumer Protection, Ispra, Italy
| | | |
Collapse
|
32
|
Tykodi SS. PD-1 as an emerging therapeutic target in renal cell carcinoma: current evidence. Onco Targets Ther 2014; 7:1349-59. [PMID: 25114573 PMCID: PMC4122552 DOI: 10.2147/ott.s48443] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Renal cell carcinoma (RCC) is the most common primary malignant tumor of the kidney in adults, representing approximately 4% of all adult cancers in the United States. Metastatic RCC is poorly responsive to conventional cytotoxic chemotherapies but can be sensitive to T-cell-directed immunotherapies such as interferon-α or interleukin-2. Despite recent progress in the application of antiangiogenic "targeted therapies" for metastatic RCC, high-dose interleukin-2 remains an appropriate first-line therapy for select patients and is associated with durable complete remissions in a small fraction of treated patients. Thus, advanced RCC provides a unique opportunity to investigate the requirements for effective antitumor immunotherapy. Accumulating evidence suggests that resistance mechanisms exploited by RCC and other tumor types may play a dominant role in limiting the effectiveness of tumor-reactive adaptive immune responses. Expression of the inhibitory coreceptor programmed cell death-1 (PD-1) on tumor-infiltrating lymphocytes within RCC tumors, as well as the expression of the PD-1 ligand (PD-L1) on RCC tumor cells, are strong negative prognostic markers for disease-specific death in RCC patients. Monoclonal antibodies targeting either PD-1 or PD-L1 have now entered clinic trials and have demonstrated promising antitumor effects for refractory metastatic RCC. This review summarizes the results of published and reported studies of PD-1- and PD-L1-targeted therapies enrolling patients with advanced RCC, focusing on key safety, toxicity, and efficacy end points. Prospects for advanced phase clinical testing and novel therapy combinations with PD-1- and PD-L1-targeted agents are discussed.
Collapse
Affiliation(s)
- Scott S Tykodi
- Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA, USA
| |
Collapse
|
33
|
Santoni M, Berardi R, Amantini C, Burattini L, Santini D, Santoni G, Cascinu S. Role of natural and adaptive immunity in renal cell carcinoma response to VEGFR-TKIs and mTOR inhibitor. Int J Cancer 2014; 134:2772-2777. [PMID: 24114790 DOI: 10.1002/ijc.28503] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 09/12/2013] [Indexed: 12/15/2022]
Abstract
Angiogenesis and immunosuppression work hand-in-hand in the renal cell carcinoma (RCC) microenvironment. Tumor growth is associated with impaired antitumor immune response in RCC, which involves T cells, natural killer cells, dendritic cells (DCs) and macrophages. Vascular endothelial growth factor receptor (VEGFR), such as sorafenib, sunitinib, pazopanib and axitinib, and mammalian target of rapamycin (mTOR) inhibitors, such as temsirolimus and everolimus, do exert both antiangiogenic and immunomodulatory functions. Indeed, these agents affect neutrophil migration, as well as T lymphocyte-DC cross-talk, DC maturation and immune cell metabolism and reactivity. In this review, we overview the essential role of innate and adaptive immune response in RCC proliferation, invasion and metastasis and the relationship between tumor-associated immune cells and the response to targeted agents approved for the treatment of metastatic RCC.
Collapse
Affiliation(s)
- Matteo Santoni
- Medical Oncology AOU Ospedali Riuniti, Polytechnic University of the Marche Region, Ancona, Italy
| | | | | | | | | | | | | |
Collapse
|
34
|
Cremona M, Espina V, Caccia D, Veneroni S, Colecchia M, Pierobon M, Deng J, Mueller C, Procopio G, Lanzi C, Daidone MG, Cho WCS, Petricoin EF, Liotta L, Bongarzone I. Stratification of clear cell renal cell carcinoma by signaling pathway analysis. Expert Rev Proteomics 2014; 11:237-249. [PMID: 24575852 DOI: 10.1586/14789450.2014.893193] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Investigation of cell signaling pathways in 16 clear cell renal cell carcinomas to identify groups based on commonly shared phosphorylation-driven signaling networks. Using laser capture microdissection and reverse-phase protein arrays, we profiled 75 key nodes spanning signaling pathways important in tumorigenesis. Analysis revealed significantly different (P < 0.05) signaling levels for 27 nodes between two groups of samples, designated A (4 samples; high EGFR, RET, and RASGFR1 levels, converging to activate AKT/mTOR) and B (12 samples; high ERK1/2 and STAT phosphorylation). Group B was further partitioned into groups C (7 samples; elevated expression of LC3B) and D (5 samples; activation of Src and STAT). Network analysis indicated that group A was characterized by signaling pathways related to cell cycle and proliferation, and group B by pathways related to cell death and survival. Homogeneous clear cell renal cell carcinomas could be stratified into at least two major functional groups.
Collapse
Affiliation(s)
- Mattia Cremona
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori , Milan , Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Coppin C. Immunotherapy for renal cell cancer in the era of targeted therapy. Expert Rev Anticancer Ther 2014; 8:907-19. [DOI: 10.1586/14737140.8.6.907] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
36
|
Santoni M, De Giorgi U, Iacovelli R, Conti A, Burattini L, Rossi L, Luca Burgio S, Berardi R, Muzzonigro G, Cortesi E, Amadori D, Cascinu S. Pre-treatment neutrophil-to-lymphocyte ratio may be associated with the outcome in patients treated with everolimus for metastatic renal cell carcinoma. Br J Cancer 2013; 109:1755-1759. [PMID: 24008663 PMCID: PMC3790174 DOI: 10.1038/bjc.2013.522] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Revised: 08/06/2013] [Accepted: 08/09/2013] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Everolimus is a mammalian target of rapamycin inhibitor approved for the treatment of metastatic renal cell carcinoma (mRCC). We aimed to assess the association between pre-treatment neutrophil-to-lymphocyte ratio (NLR) and the outcome of patients treated with everolimus for mRCC. METHODS Ninety-seven patients with mRCC were treated with everolimus till April 2013 in our institutions. Patients were stratified in two groups with NLR >3 (Group A) vs <3 (Group B). Progression-free survival (PFS) and overall survival (OS) were estimated using Kaplan-Meier method. Gender, age, Motzer prognostic group, PFS on first-line therapy, neutrophilia and NLR were included in the Cox analysis to investigate their prognostic relevance. RESULTS Median OS and PFS were 10.6 and 5.3 months, respectively. Median OS was 12.2 months in Group A and 24.4 months in Group B (P=0.001). Median PFS was 3.4 months in Group A and 9.9 months in Group B (P<0.001). At multivariate analysis, only Motzer prognostic group and NLR were independent prognostic factors for OS and PFS. CONCLUSION Pre-treatment NLR is an independent prognostic factor for patients with mRCC treated with second- or third-line everolimus. This should be investigated and validated in prospective studies.
Collapse
Affiliation(s)
- M Santoni
- Clinica di Oncologia Medica, AOU ‘Ospedali Riuniti', Università Politecnica delle Marche, Via Tronto 10/A, 60126 Ancona, Italy
| | - U De Giorgi
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori-IRST, Via Piero Maroncelli 40, 47014 Meldola (FC), Italy
| | - R Iacovelli
- Dipartimento di Scienze Radiologiche, Oncologiche e Anatomo-Patologiche, ‘Sapienza' Università di Roma, piazzale Aldo Moro 5, 00185 Rome, Italy
| | - A Conti
- Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche, Clinica di Urologia, AOU ‘Ospedali Riuniti', Università Politecnica delle Marche, Via Tronto 10/A, 60126 Ancona, Italy
| | - L Burattini
- Clinica di Oncologia Medica, AOU ‘Ospedali Riuniti', Università Politecnica delle Marche, Via Tronto 10/A, 60126 Ancona, Italy
| | - L Rossi
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori-IRST, Via Piero Maroncelli 40, 47014 Meldola (FC), Italy
| | - S Luca Burgio
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori-IRST, Via Piero Maroncelli 40, 47014 Meldola (FC), Italy
| | - R Berardi
- Clinica di Oncologia Medica, AOU ‘Ospedali Riuniti', Università Politecnica delle Marche, Via Tronto 10/A, 60126 Ancona, Italy
| | - G Muzzonigro
- Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche, Clinica di Urologia, AOU ‘Ospedali Riuniti', Università Politecnica delle Marche, Via Tronto 10/A, 60126 Ancona, Italy
| | - E Cortesi
- Dipartimento di Scienze Radiologiche, Oncologiche e Anatomo-Patologiche, ‘Sapienza' Università di Roma, piazzale Aldo Moro 5, 00185 Rome, Italy
| | - D Amadori
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori-IRST, Via Piero Maroncelli 40, 47014 Meldola (FC), Italy
| | - S Cascinu
- Clinica di Oncologia Medica, AOU ‘Ospedali Riuniti', Università Politecnica delle Marche, Via Tronto 10/A, 60126 Ancona, Italy
| |
Collapse
|
37
|
Said R, Amato RJ. Identification of Pre- and Post-Treatment Markers, Clinical, and Laboratory Parameters Associated with Outcome in Renal Cancer Patients Treated with MVA-5T4. Front Oncol 2013; 3:185. [PMID: 23875174 PMCID: PMC3711044 DOI: 10.3389/fonc.2013.00185] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 07/02/2013] [Indexed: 01/20/2023] Open
Abstract
The recent approvals of immunotherapeutic agents (Sipuleucel-T and Ipilimumab) for the treatment of different solid tumors gave a boost to the growing cancer immunotherapy field, even though few immunotherapy studies have demonstrated convincingly that there is a direct link between the predicted mode of action of an immunological compound and therapeutic benefit. MVA-5T4 (TroVax®) is a novel vaccine combining the tumor-associated antigen 5T4 to an engineered vector-modified vaccinia Ankara (MVA). MVA helps to express the oncofetal 5T4 antigen and subsequently trigger a tumor-directed immune reaction. The safety and clinical benefit reported in multiple phase I and II clinical trials using MVA-5T4 were encouraging; immune responses were induced in almost all treated patients, and associations between 5T4-specific cellular or humoral responses and clinical benefit were reported in most of the nine phase II trials. In particular, clinical studies conducted in renal cell carcinoma (RCC) patients have demonstrated an association between 5T4-specific (but not MVA) antibody responses and enhanced survival. This review describes the clinical studies using MVA-5T4 conducted in RCC that convincingly demonstrated that an antigen-specific immune response induced by vaccination is associated with enhanced patient survival and is not simply a function of the general “health” of patients. We will also provide our expert opinions on possible future better-designed clinical trials based on relevant biomarkers. In addition, various combinations of MVA-5T4 and different and newer immunomodulator agents with promising clinical benefit will be discussed.
Collapse
Affiliation(s)
- Rabih Said
- Division of Oncology, Department of Internal Medicine, Memorial Hermann Cancer Center, University of Texas Health Science Center at Houston (Medical School) , Houston, TX , USA
| | | |
Collapse
|
38
|
|
39
|
Daurkin I, Eruslanov E, Stoffs T, Perrin GQ, Algood C, Gilbert SM, Rosser CJ, Su LM, Vieweg J, Kusmartsev S. Tumor-associated macrophages mediate immunosuppression in the renal cancer microenvironment by activating the 15-lipoxygenase-2 pathway. Cancer Res 2011; 71:6400-9. [PMID: 21900394 DOI: 10.1158/0008-5472.can-11-1261] [Citation(s) in RCA: 176] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Renal cell carcinoma (RCC), the most common human kidney cancer, is frequently infiltrated with tumor-associated macrophages (TAM) that can promote malignant progression. Here, we show that TAMs isolated from human RCC produce substantial amounts of the proinflammatory chemokine CCL2 and immunosuppressive cytokine IL-10, in addition to enhanced eicosanoid production via an activated 15-lipoxygenase-2 (15-LOX2) pathway. TAMs isolated from RCC tumors had a high 15-LOX2 expression and secreted substantial amounts of 15(S)-hydroxyeicosatetraenoic acid, its major bioactive lipid product. Inhibition of lipoxygenase activity significantly reduced production of CCL2 and IL-10 by RCC TAMs. In addition, TAMs isolated from RCC were capable of inducing in T lymphocytes, the pivotal T regulatory cell transcription factor forkhead box P3 (FOXP3), and the inhibitory cytotoxic T-lymphocyte antigen 4 (CTLA-4) coreceptor. However, this TAM-mediated induction of FOXP3 and CTLA-4 in T cells was independent of lipoxygenase and could not be reversed by inhibiting lipoxygenase activity. Collectively, our results show that TAMs, often present in RCCs, display enhanced 15-LOX2 activity that contributes to RCC-related inflammation, immunosuppression, and malignant progression. Furthermore, we show that TAMs mediate the development of immune tolerance through both 15-LOX2-dependent and 15-LOX2-independent pathways. We propose that manipulating LOX-dependent arachidonic acid metabolism in the tumor microenvironment could offer new strategies to block cancer-related inflammation and immune escape in patients with RCC.
Collapse
Affiliation(s)
- Irina Daurkin
- Department of Urology, University of Florida, Gainesville, Florida, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Montagna D, Turin I, Schiavo R, Montini E, Zaffaroni N, Villa R, Secondino S, Schiavetto I, Caliogna L, Locatelli F, Libri V, Pession A, Tonelli R, Maccario R, Siena S, Pedrazzoli P. Feasibility and safety of adoptive immunotherapy with ex vivo-generated autologous, cytotoxic T lymphocytes in patients with solid tumor. Cytotherapy 2011; 14:80-90. [PMID: 21942841 DOI: 10.3109/14653249.2011.610303] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND AIMS Adoptive T-cell therapy with tumor-specific T cells has emerged as a potentially useful approach for treating patients with advanced malignancies. We have demonstrated previously the feasibility of obtaining large numbers of autologous anti-tumor-specific cytotoxic T lymphocytes (CTL) generated by stimulation of patients' peripheral blood mononuclear cells with dendritic cells pulsed with apoptotic tumor cells. Methods. Six patients with progressing metastatic solid tumors (one renal cell carcinoma, two ovarian cancers, two extraosseous peripheral neuroectodermal tumors, one soft tissue sarcoma) not eligible for conventional therapies were treated with adoptive immunotherapy. Anti-tumor CTL, proven to be reactive in vitro against patient tumor cells, but not against normal cells, were infused following lymphodepleting chemotherapy administered to favor T-cell proliferation in vivo. RESULTS Patients received a median of nine CTL infusions (range 2-19). The median number of CTL administered per infusion was 11 × 10(8) (range 1-55 × 10(8)). No patient experienced acute or late adverse events related to CTL infusion, even when large numbers of cells were given. Post-infusion laboratory investigations demonstrated an increase in the frequency of circulating anti-tumor T-cells and, in patients with a longer follow-up receiving two CTL infusions/year, a stabilization of these values. CONCLUSIONS Our study demonstrates that autologous ex vivo-generated anti-tumor CTL can be administered safely in patients with advanced solid tumors and can improve the immunologic reactivity of recipients against tumor. These preliminary results provide a rationale for evaluating the clinical efficacy of this immunotherapeutic approach in phase I/II studies.
Collapse
Affiliation(s)
- Daniela Montagna
- Dipartimento di Scienze Pediatriche, Università di Pavia, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Florescu A, Amir E, Bouganim N, Clemons M. Immune therapy for breast cancer in 2010-hype or hope? ACTA ACUST UNITED AC 2011; 18:e9-e18. [PMID: 21331271 DOI: 10.3747/co.v18i1.623] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The identification of numerous breast cancer antigens has generated increasing enthusiasm for the application of immune-based therapies in breast malignancies. Although the use of monoclonal antibodies has revolutionized the "targeted therapy" of breast cancer, and the immunomodulatory effects of bisphosphonates continue to be evaluated, few studies to date have demonstrated widespread utility for other forms of immunotherapy. The present review assesses modern research and explores whether the hopes for immunotherapy can overcome the hype.
Collapse
Affiliation(s)
- A Florescu
- Division of Medical Oncology and Hematology, Princess Margaret Hospital, and Department of Medicine, University of Toronto, Toronto, ON
| | | | | | | |
Collapse
|
42
|
A polymorphism in the splice donor site of ZNF419 results in the novel renal cell carcinoma-associated minor histocompatibility antigen ZAPHIR. PLoS One 2011; 6:e21699. [PMID: 21738768 PMCID: PMC3125305 DOI: 10.1371/journal.pone.0021699] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Accepted: 06/05/2011] [Indexed: 01/12/2023] Open
Abstract
Nonmyeloablative allogeneic stem cell transplantation (SCT) can induce remission in patients with renal cell carcinoma (RCC), but this graft-versus-tumor (GVT) effect is often accompanied by graft-versus-host disease (GVHD). Here, we evaluated minor histocompatibility antigen (MiHA)-specific T cell responses in two patients with metastatic RCC who were treated with reduced-intensity conditioning SCT followed by donor lymphocyte infusion (DLI). One patient had stable disease and emergence of SMCY.A2-specific CD8+ T cells was observed after DLI with the potential of targeting SMCY-expressing RCC tumor cells. The second patient experienced partial regression of lung metastases from whom we isolated a MiHA-specific CTL clone with the capability of targeting RCC cell lines. Whole genome association scanning revealed that this CTL recognizes a novel HLA-B7-restricted MiHA, designated ZAPHIR, resulting from a polymorphism in the splice donor site of the ZNF419 gene. Tetramer analysis showed that emergence of ZAPHIR-specific CD8+ T cells in peripheral blood occurred in the absence of GVHD. Furthermore, the expression of ZAPHIR in solid tumor cell lines indicates the involvement of ZAPHIR-specific CD8+ T cell responses in selective GVT immunity. These findings illustrate that the ZNF419-encoded MiHA ZAPHIR is an attractive target for specific immunotherapy after allogeneic SCT.
Collapse
|
43
|
Demiselle J, Lheureux S, Clarisse B, Sevin E, Joly F. Metastatic renal cancer: evolution of five complete response cases after the antiangiogenic discontinuation. Bull Cancer 2011; 98:bdc.2011.1368. [PMID: 21659064 DOI: 10.1684/bdc.2011.1368] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Antiangiogenic therapies have led to substantial progress in the management of kidney cancer, highly vascular tumor, and chemoresistant. These molecules have improved the prognosis of metastatic renal cancer. However, only a few isolated cases of complete response have been described and the evolution of these patients after treatment discontinuation remains unclear. From a series of patients treated for kidney cancer with antiangiogenic in first line, the purpose of this study was to identify patients in complete response in whom treatment had been interrupted. Complete response was defined according to RECIST criteria and data were collected retrospectively at the Centre François Baclesse - Caen. Five patients were identified in complete response with a treatment discontinuation among sixty-seven patients. These five patients of good or intermediate prognosis received an initial nephrectomy followed by a first-line treatment by Sunitinib (ten cycles on average). After one year of stopping treatment, two patients are still in complete response and three patients relapsed at three, 12 and 15 months. The treatment of relapsing disease was surgical followed by monitoring or resumption of sunitinib resulting in new complete response for the all three patients. The interruption of antiangiogenic treatment seems acceptable after a complete response.
Collapse
Affiliation(s)
- Julien Demiselle
- CLCC François-Baclesse, service de recherche clinique, avenue du Général-Harris, 14000 Caen, FranceCLCC François-Baclesse, service de recherche clinique, avenue du Général-Harris, 14000 Caen, France
| | | | | | | | | |
Collapse
|
44
|
Tykodi SS, Sandmaier BM, Warren EH, Thompson J. Allogeneic hematopoietic cell transplantation for renal cell carcinoma: ten years after. Expert Opin Biol Ther 2011; 11:763-73. [PMID: 21417772 PMCID: PMC3201801 DOI: 10.1517/14712598.2011.566855] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION The first series of patients with metastatic renal cell carcinoma (RCC) treated by non-myeloablative allogeneic hematopoietic cell transplantation (HCT) was reported in 2000 and demonstrated an allogeneic graft-versus-tumor (GVT) effect that encouraged further investigation of this approach. However, the past 10 years have also witnessed profound changes in the medical management of metastatic RCC with the introduction of targeted therapies directed against VEGF or mammalian target of rapamycin (mTOR) signaling pathways creating uncertainty about a continued role for allogeneic HCT in the treatment of RCC. AREAS COVERED A total of 21 published reports describing clinical outcomes for 398 patients with metastatic RCC treated by allogeneic HCT compiled herein provide a composite overview of the world wide experience for key efficacy and toxicity outcomes. Review of correlative studies that identify donor-derived T cells as mediators of RCC-specific GVT effects offers insight into both the potential as well as the technical barriers to the delivery of antigen-specific post-transplant cellular therapy or vaccination designed to augment the allogeneic GVT effect. EXPERT OPINION The future development of non-myeloablative allogeneic HCT for metastatic RCC will require novel treatment protocols designed to augment and sustain post-transplant GVT effects against RCC to generate renewed enthusiasm for this approach.
Collapse
Affiliation(s)
- Scott S Tykodi
- Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, MS D5-380, PO Box 19024, Seattle, 98109 United States
| | - Brenda M Sandmaier
- Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, MS D1-100, PO Box 19024, Seattle, 98109 United States,
| | - Edus H Warren
- Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, MS D3-100, PO Box 19024, Seattle, 98109 United States,
| | - J Thompson
- Seattle Cancer Care Alliance, 825 Eastlake Avenue E, MS G4-830, Seattle, 98109 United States,
| |
Collapse
|
45
|
Abstract
INTRODUCTION The efficacy of sequential everolimus (RAD001, Afinitor®), an orally administered inhibitor of mammalian target of rapamycin (mTOR), is one of the therapeutic options in patients with advanced renal cell cancer (RCC). The purpose of this review is to discuss the mTOR pathway and to update current knowledge of the role of everolimus in metastatic RCC. AREAS COVERED The function of mTOR and its inhibition, the early development trials of everolimus, the Phase II trial that lead to the Phase III study and ultimately to FDA approval are all discussed in this review. Literature utilized for this review consists of PUBMED for both description of the mTOR pathway and its role, in addition to publications using everolimus in RCC. EXPERT OPINION Everolimus is currently the only agent with a proven progression-free survival improvement for patients who progressed on a vascular endothelial growth factor receptor-tyrosine kinase inhibitor. Future studies may further shed light on the most optimal use of everolimus.
Collapse
Affiliation(s)
- Robert Amato
- UT Medical School at Houston -- Internal Medicine, Division of Oncology, Houston, TX 77030, USA.
| |
Collapse
|
46
|
Immunomodulatory effects of sorafenib on peripheral immune effector cells in metastatic renal cell carcinoma. Eur J Cancer 2011; 47:690-6. [DOI: 10.1016/j.ejca.2010.11.021] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2010] [Revised: 11/10/2010] [Accepted: 11/23/2010] [Indexed: 01/07/2023]
|
47
|
Abstract
Explorative knowledge of cellular and molecular mechanisms of immune function and regulation has provided optimism in developing cancer immunotherapy. However, three decades of experimental and clinical investigations to offer powerful immunotherapeutic strategies against solid tumors, with the possible exception of monoclonal antibody-targeted therapies, have not succeeded in significantly prolonging patient survival. Nonspecific immune approaches, including cytokine-based therapies and allogeneic hematopoietic stem cell transplantation, have so far produced consistent, although limited, results. In this review, we present the developments of cell transfer-based strategies that, in preclinical studies, have demonstrated potential efficacy, but have only established tumor regression in limited numbers of patients. The key to success demands creative combinations of tumor antigens, adjuvance, gene modification and various administration strategies in the development of cell-based therapies together with other cancer-treatment principles, often in a stepwise 'space-rocket-type' approach. Combined efforts of several scientific disciplines, such as tumor biology and immunology, as well as cell and gene research in transplantation, will open new venues. New regulation for clinical trials with advanced therapy medicine products to ensure patient safety will be highlighted.
Collapse
Affiliation(s)
- Lisbeth Barkholt
- Division of Clinical Immunology & Transfusion Medicine, Department of Laboratory Medicine, Karolinska University Hospital Huddinge F79, Stockholm, Sweden.
| | | |
Collapse
|
48
|
Immunotherapy for renal cell carcinoma. Clin Dev Immunol 2011; 2010:284581. [PMID: 21253521 PMCID: PMC3022170 DOI: 10.1155/2010/284581] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2010] [Accepted: 11/29/2010] [Indexed: 11/30/2022]
Abstract
Immunotherapy plays a significant role in the management of renal cell carcinoma (RCC) patients with metastatic disease because RCC is highly resistant to both chemotherapy and radiation therapy. Many reports illustrate various approaches to the treatment of RCC, such as cytokine-, antigen- or dendritic cell- (DC-) based immunotherapy, and the safety and effectiveness of immunotherapy have been highlighted by multiple clinical trials. Although antitumor immune responses and clinically significant outcomes have been achieved in these trials, the response rate is still low, and very few patients show long-term clinical improvement. Recently, the importance of immune regulation by antigen-presenting cells (APC) and regulatory T cells (Treg cells) has also been discussed. The authors outline the principles of cell-mediated tumor immunotherapy and discuss clinical trials of immunotherapy for RCC.
Collapse
|
49
|
Shablak A, O’Dwyer J, Hawkins R, Board R. Management of a New Isolated Metastasis during Sunitinib Treatment in Renal Cell Carcinoma Patients: A Lesson from Two Cases. Urol Int 2011; 86:245-8. [DOI: 10.1159/000321908] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Accepted: 10/03/2010] [Indexed: 12/24/2022]
|
50
|
Rixe O, Rini B. Renal cell carcinoma: ten years of significant advances. Target Oncol 2010; 5:73-4. [DOI: 10.1007/s11523-010-0150-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Accepted: 08/03/2010] [Indexed: 10/19/2022]
|