1
|
Roy AM, George S. Emerging resistance vs. losing response to immune check point inhibitors in renal cell carcinoma: two differing phenomena. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:642-655. [PMID: 37842239 PMCID: PMC10571056 DOI: 10.20517/cdr.2023.47] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/27/2023] [Accepted: 09/16/2023] [Indexed: 10/17/2023]
Abstract
The introduction of immune checkpoint inhibitor (ICI) has revolutionized the treatment of metastatic renal cell carcinoma (mRCC) and has dramatically improved the outcomes of patients. The use of monotherapy or combinations of ICIs targeting PD-1/PD-L1 and CTLA-4, as well as the addition of ICIs with tyrosine kinase inhibitors, has significantly enhanced the overall survival of mRCC patients. Despite these promising results, there remains a subset of patients who either do not respond to treatment (primary resistance) or develop resistance to therapy over time (acquired resistance). Understanding the mechanisms underlying the development of resistance to ICI treatment is crucial in the management of mRCC, as they can be used to identify new targets for innovative therapeutic strategies. Currently, there is an unmet need to develop new predictive and prognostic biomarkers that can aid in the development of personalized treatment options for mRCC patients. In this review, we summarize several mechanisms of ICI resistance in RCC, including alterations in tumor microenvironment, upregulation of alternative immune checkpoint pathways, and genetic and epigenetic changes. Additionally, we highlight potential strategies that can be used to overcome resistance, such as combination therapy, targeted therapy, and immune modulation.
Collapse
Affiliation(s)
| | - Saby George
- Division of Hematology and Oncology, Department of Internal Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
| |
Collapse
|
2
|
CDCA3 Predicts Poor Prognosis and Affects CD8+ T Cell Infiltration in Renal Cell Carcinoma. JOURNAL OF ONCOLOGY 2022; 2022:6343760. [PMID: 36213833 PMCID: PMC9534638 DOI: 10.1155/2022/6343760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/05/2022] [Accepted: 09/07/2022] [Indexed: 12/03/2022]
Abstract
Background Cell division cycle associated 3 (CDCA3) mediates the ubiquitination WEE1 kinase at G2/M phase. However, its contribution to cancer immunity remains uncertain. Methods We first evaluated the effect of CDCA3 on the prognosis of patients with renal cell carcinoma (RCC). The results of bioinformatics analysis were verified by the tissue microarray, immunofluorescence (IF) staining, CCK-8 assay, colony formation, cell cycle, and Western blot. Results Bioinformatics analysis predicated CDCA3 was an independent predictor of poor prognosis in RCC and was associated with poor TNM stage and grade. CDCA3 was related to the infiltration of CD8+ T cells and Tregs. Tissue microarray demonstrated that CDCA3 was strongly associated with poor prognosis and positively relevant to CD8+ T infiltration. In vitro experiments showed that exgenomic interference of CDCA3 could attenuate cellular proliferation, arrest cell cycle, and blockade accumulation of CDK4, Bub3, and Cdc20 in mitosis process. Conclusion CDCA3 presents as a good biomarker candidate to predict the prognosis of RCC patients and potentiates the immune tumor microenvironment (TME) of RCC.
Collapse
|
3
|
Ged Y, Lee CH. Lenvatinib plus pembrolizumab combination therapy for adult patients with advanced renal cell carcinoma. Expert Rev Anticancer Ther 2022; 22:1049-1059. [PMID: 36154355 DOI: 10.1080/14737140.2022.2128336] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION : The treatment landscape of metastatic RCC has significantly evolved in recent years with the advent and approval of multiple combinations of anti-angiogenic agents with immune checkpoint inhibitors, of which the combination of lenvatinib plus pembrolizumab is the most recent to be incorporated into clinical practice. AREAS COVERED Herein, we provide an overview of the combination of lenvatinib plus pembrolizumab in metastatic RCC, including the mechanism of action, pharmacokinetics, efficacy, and safety profile. EXPERT OPINION Lenvatinib plus pembrolizumab has demonstrated substantial efficacy in patients with metastatic RCC in the first-line and refractory treatment setting with the highest reported results of radiological responses, complete responses, and progression free survival compared to all other RCC treatments. However, the field is currently still limited with the limited availability of biomarkers to inform on treatment selection and the lack of head-to-head studies across the effective RCC treatments. Ongoing and future studies are eagerly anticipated to uncover multiple unmet needs in RCC.
Collapse
Affiliation(s)
- Yasser Ged
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Chung-Han Lee
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
4
|
Gong Z, Wu X, Guo Q, Du H, Zhang F, Kong Y. Comprehensive Analysis of HMCN1 Somatic Mutation in Clear Cell Renal Cell Carcinoma. Genes (Basel) 2022; 13:genes13071282. [PMID: 35886066 PMCID: PMC9316380 DOI: 10.3390/genes13071282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/14/2022] [Accepted: 07/16/2022] [Indexed: 12/11/2022] Open
Abstract
Background: Renal cell carcinoma (RCC) is a common malignancy of the genitourinary system and clear cell renal cell carcinoma (ccRCC) is the most representative subtype. The morbidity and mortality of ccRCC have gradually risen during recent years; however, the pathogenesis and potential biomarkers remain unclear. The purpose of our study was to find out prognostic genes correlated with somatic mutation and the underlying mechanisms of HMCN1 mutation in ccRCC. Methods: Somatic mutation data of two ccRCC cohorts were acquired from TCGA and cBioPortal. Genes frequently mutated in both datasets were extracted, from which tumor mutation burden and survival analysis revealed three prognostic genes. Further comprehensive analysis of HMCN1 mutation was carried out to identify differentially expressed genes and apply functional annotations. The correlation of HMCN1 mutation and tumor immunity was also evaluated. Results: HMCN1, SYNE1, and BAP1 mutations were associated with both tumor mutation burden and clinical prognosis in ccRCC. Gene enrichment analysis suggested the effects of HMCN1 mutation on biological processes and pathways linked to energy metabolism. HMCN1 mutation was also correlated with anti-tumor immunity. There were several limitations in the sample size and cohort availability of the present computational study. Conclusions: The present results inferred that HMCN1 mutation might have an important clinical significance for ccRCC patients by regulating metabolism and the immune microenvironment.
Collapse
|
5
|
Ingrosso G, Becherini C, Francolini G, Lancia A, Alì E, Caini S, Teriaca MA, Marchionni A, Filippi AR, Livi L, Sanguineti G, Aristei C, Detti B. Stereotactic body radiotherapy (SBRT) in combination with drugs in metastatic kidney cancer: A systematic review. Crit Rev Oncol Hematol 2021; 159:103242. [PMID: 33545356 DOI: 10.1016/j.critrevonc.2021.103242] [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: 11/15/2020] [Revised: 01/15/2021] [Accepted: 01/20/2021] [Indexed: 10/22/2022] Open
Abstract
OBJECTIVE To conduct a systematic review and meta-analysis of the role of SBRTdrug combination in patients affected by mRCC and associated oncologic outcomes and toxicity profiles. EVIDENCE ACQUISITION We performed a critical review of the Pubmed, Medline, and Embase databases from January 1, 2000 through April 30, 2020 according to the Preferred Reporting Items and Meta-Analyses statement. To assess the overall quality of the literature reviewed, we used a modified Delphi tool. EVIDENCE SYNTHESIS A total of 6 studies were included, corresponding to a cohort of 216 patients. Tyrosine Kinases Inhibitors were the most widely used drugs in combination with SBRT, being administered in 93% patients. No study reported an increase of radiation-induced toxicity. CONCLUSIONS SBRT resulted to be safe, without increase in terms of drugs-related adverse events in this setting. Moreover, this approach showed promising clinical outcomes in terms of LC and OS.
Collapse
Affiliation(s)
- Gianluca Ingrosso
- Radiation Oncology Section, Department of Surgical and Biomedical Science, University of Perugia, Italy
| | - Carlotta Becherini
- Radiation Oncology, A.O.U. Careggi, University of Florence, Largo Brambilla, 3, 50134 Florence, Italy
| | - Giulio Francolini
- Radiation Oncology, A.O.U. Careggi, University of Florence, Largo Brambilla, 3, 50134 Florence, Italy
| | - Andrea Lancia
- Radiation Oncology, Fondazione IRCCS Policlinico S. Matteo, Pavia, Italy
| | - Emanuele Alì
- Radiation Oncology Section, Department of Surgical and Biomedical Science, University of Perugia, Italy
| | - Saverio Caini
- Cancer Risk Factors and Lifestyle Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Networking, Florence, Italy
| | - Maria Ausilia Teriaca
- Radiation Oncology, A.O.U. Careggi, University of Florence, Largo Brambilla, 3, 50134 Florence, Italy
| | - Alessandro Marchionni
- Radiation Oncology Section, Department of Surgical and Biomedical Science, University of Perugia, Italy
| | | | - Lorenzo Livi
- Radiation Oncology, A.O.U. Careggi, University of Florence, Largo Brambilla, 3, 50134 Florence, Italy
| | - Giuseppe Sanguineti
- Department of Radiation Oncology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Cynthia Aristei
- Radiation Oncology Section, Department of Surgical and Biomedical Science, University of Perugia, Italy
| | - Beatrice Detti
- Radiation Oncology, A.O.U. Careggi, University of Florence, Largo Brambilla, 3, 50134 Florence, Italy.
| |
Collapse
|
6
|
Adapala RKR, Prabhu GGL, Sanman KN, Yalla DR, Shetty R, Venugopal P. Is preoperative neutrophil-to-lymphocyte ratio a red flag which can predict high-risk pathological characteristics in renal cell carcinoma? Urol Ann 2021; 13:47-52. [PMID: 33897164 PMCID: PMC8052900 DOI: 10.4103/ua.ua_34_19] [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: 03/05/2019] [Accepted: 01/28/2020] [Indexed: 11/04/2022] Open
Abstract
Introduction Renal cell carcinoma (RCC) is known to invoke both immunological and inflammatory responses. While the neutrophils mediate the tumor-induced inflammatory response, the lymphocytes bring about the various immunological events associated with it. The neutrophil-to-lymphocyte ratio (NLR) is a simple indicator of this dual response. We investigated the association between preoperative NLR and histopathological prognostic variables of RCC intending to find out whether it can be of value as a red flag capable of alerting the clinician as to the biological character of the tumor under consideration. Methods Preoperative NLR and clinicopathological variables, namely histological subtype, nuclear grade, staging, lymphovascular invasion, capsular invasion, tumor necrosis, renal sinus invasion, and sarcomatoid differentiation of 60 patients who underwent radical or partial nephrectomy, were analyzed to detect the association between the two. Results We found that mean preoperative NLR was significantly higher in clear-cell carcinomas (3.25 ± 0.29) when compared with nonclear-cell carcinomas (2.25 ± 0.63). There was a linear trend of NLR rise as the stage of the disease advanced. A significant rise in preoperative NLR was noted in tumors with various high-risk histopathological features such as tumor size, capsular invasion, tumor necrosis, and sarcomatoid differentiation. Conclusion Preoperative measurement of NLR is a simple test which may provide an early clue of high-risk pathological features of renal cell cancer.
Collapse
Affiliation(s)
| | - G G Laxman Prabhu
- Department of Urology, Kasturba Medical College Hospital, Mangalore, Karnataka, India
| | - K N Sanman
- Department of Urology, Kasturba Medical College Hospital, Mangalore, Karnataka, India
| | - Durga Rao Yalla
- Department of Biochemistry, Kasturba Medical College Hospital, Mangalore, Karnataka, India
| | - Ranjit Shetty
- Department of Urology, Kasturba Medical College Hospital, Mangalore, Karnataka, India
| | - P Venugopal
- Department of Biochemistry, Kasturba Medical College Hospital, Mangalore, Karnataka, India
| |
Collapse
|
7
|
Chakravarty D, Huang L, Kahn M, Tewari AK. Immunotherapy for Metastatic Prostate Cancer: Current and Emerging Treatment Options. Urol Clin North Am 2020; 47:487-510. [PMID: 33008499 DOI: 10.1016/j.ucl.2020.07.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The advent of immunotherapy has revolutionized cancer treatment. Prostate cancer has an immunosuppressive microenvironment and a low tumor mutation burden, resulting in low neoantigen expression. The consensus was that immunotherapy would be less effective in prostate cancer. However, recent studies have reported that prostate cancer does have a high number of DNA damage and repair gene defects. Immunotherapies that have been tested in prostate cancer so far have been mainly vaccines and checkpoint inhibitors. A combination of genomically targeted therapies, with approaches to alleviate immune response and thereby make the tumor microenvironment immunologically hot, is promising.
Collapse
Affiliation(s)
- Dimple Chakravarty
- Department of Urology and the Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Li Huang
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Matthew Kahn
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ashutosh K Tewari
- Department of Urology and the Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
8
|
Attalla K, Weng S, Voss MH, Hakimi AA. Epidemiology, Risk Assessment, and Biomarkers for Patients with Advanced Renal Cell Carcinoma. Urol Clin North Am 2020; 47:293-303. [PMID: 32600532 DOI: 10.1016/j.ucl.2020.04.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In the preceding two decades, several milestones have been reached in the management of patients with metastatic renal cell carcinoma (mRCC), including the development of novel targeted agents paralleling an increased understanding of the molecular biology of this disease process. Recently, a renewed enthusiasm for immunotherapy in the form of immune checkpoint blockade has resulted in significant strides in the treatment of mRCC. Despite these advances, treatment remains challenging for clinicians, and only modest survival benefits are observed with current treatment paradigms. The risk-stratification tools and investigated predictive and prognostic biomarkers in patients with mRCC are detailed in this review.
Collapse
Affiliation(s)
- Kyrollis Attalla
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, 353 East 68th Street, 5th Floor, New York, NY 10065, USA
| | - Stanley Weng
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, 353 East 68th Street, 5th Floor, New York, NY 10065, USA
| | - Martin H Voss
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, 353 East 68th Street, 5th Floor, New York, NY 10065, USA
| | - A Ari Hakimi
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, 353 East 68th Street, 5th Floor, New York, NY 10065, USA.
| |
Collapse
|
9
|
Cervera-Carrascon V, Quixabeira DCA, Santos JM, Havunen R, Zafar S, Hemminki O, Heiniö C, Munaro E, Siurala M, Sorsa S, Mirtti T, Järvinen P, Mildh M, Nisen H, Rannikko A, Anttila M, Kanerva A, Hemminki A. Tumor microenvironment remodeling by an engineered oncolytic adenovirus results in improved outcome from PD-L1 inhibition. Oncoimmunology 2020; 9:1761229. [PMID: 32923123 PMCID: PMC7458667 DOI: 10.1080/2162402x.2020.1761229] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Checkpoint inhibitors have revolutionized cancer therapy and validated immunotherapy as an approach. Unfortunately, responses are seen in a minority of patients. Our objective is to use engineered adenoviruses designed to increase lymphocyte trafficking and cytokine production at the tumor, to assess if they increase the response rate to checkpoint inhibition, as these features have been regarded as predictive for the responses. When Ad5/3-E2F-d24-hTNFa-IRES-hIL2 (an oncolytic adenovirus coding for TNFa and IL-2, also known as TILT-123) and checkpoint inhibitors were used together in fresh urological tumor histocultures, a significant shift toward immune activity (not only tumor necrosis alpha and interleukin-2 but also interferon gamma and granzyme B) and increased T-cell trafficking signals (CXCL10) was observed. In vivo, our viruses enabled an anti-PD-L1 (a checkpoint inhibitor) delivering complete responses in all the treated animals (hazard ratios versus anti-PD-L1 alone 0.057 [0.007; 0.451] or virotherapy alone 0.067 [0.011; 0.415]). To conclude, when an engineered oncolytic adenovirus was utilized to modify the tumor microenvironment towards what meta-analyses have pointed as predictive markers for checkpoint inhibitory therapy, the response to them increased synergistically. Of note, key findings were confirmed in fresh patient-derived tumor explants.
Collapse
Affiliation(s)
- Victor Cervera-Carrascon
- Cancer Gene Therapy Group, Translational Immunology Research Program and Department of Oncology, University of Helsinki, Helsinki, Finland.,TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Dafne C A Quixabeira
- Cancer Gene Therapy Group, Translational Immunology Research Program and Department of Oncology, University of Helsinki, Helsinki, Finland
| | - Joao Manuel Santos
- Cancer Gene Therapy Group, Translational Immunology Research Program and Department of Oncology, University of Helsinki, Helsinki, Finland.,TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Riikka Havunen
- Cancer Gene Therapy Group, Translational Immunology Research Program and Department of Oncology, University of Helsinki, Helsinki, Finland.,TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Sadia Zafar
- Cancer Gene Therapy Group, Translational Immunology Research Program and Department of Oncology, University of Helsinki, Helsinki, Finland
| | - Otto Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program and Department of Oncology, University of Helsinki, Helsinki, Finland.,Department of Urology, Helsinki University Hospital, Helsinki, Finland
| | - Camilla Heiniö
- Cancer Gene Therapy Group, Translational Immunology Research Program and Department of Oncology, University of Helsinki, Helsinki, Finland
| | - Eleonora Munaro
- Cancer Gene Therapy Group, Translational Immunology Research Program and Department of Oncology, University of Helsinki, Helsinki, Finland
| | - Mikko Siurala
- Cancer Gene Therapy Group, Translational Immunology Research Program and Department of Oncology, University of Helsinki, Helsinki, Finland.,TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Suvi Sorsa
- Cancer Gene Therapy Group, Translational Immunology Research Program and Department of Oncology, University of Helsinki, Helsinki, Finland.,TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Tuomas Mirtti
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland.,Department of Pathology, Medicum, University of Helsinki, Helsinki, Finland.,Department of Pathology, HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Petrus Järvinen
- Department of Urology, Helsinki University Hospital, Helsinki, Finland
| | - Markus Mildh
- Department of Urology, Helsinki University Hospital, Helsinki, Finland
| | - Harry Nisen
- Department of Urology, Helsinki University Hospital, Helsinki, Finland
| | - Antti Rannikko
- Department of Urology, Helsinki University Hospital, Helsinki, Finland
| | - Marjukka Anttila
- Pathology Unit, Finnish Food Safety Authority (EVIRA), Helsinki, Finland
| | - Anna Kanerva
- Cancer Gene Therapy Group, Translational Immunology Research Program and Department of Oncology, University of Helsinki, Helsinki, Finland.,Department of Obstetrics and Gynecology, Helsinki University Central Hospital, Helsinki, Finland
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program and Department of Oncology, University of Helsinki, Helsinki, Finland.,TILT Biotherapeutics Ltd, Helsinki, Finland.,Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| |
Collapse
|
10
|
The Double-Faced Role of Nitric Oxide and Reactive Oxygen Species in Solid Tumors. Antioxidants (Basel) 2020; 9:antiox9050374. [PMID: 32365852 PMCID: PMC7278755 DOI: 10.3390/antiox9050374] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 02/08/2023] Open
Abstract
Disturbed redox homeostasis represents a hallmark of cancer phenotypes, affecting cellular metabolism and redox signaling. Since reactive oxygen and nitrogen species (ROS/RNS) are involved in regulation of proliferation and apoptosis, they may play a double-faced role in cancer, entailing protumorigenic and tumor-suppressing effects in early and later stages, respectively. In addition, ROS and RNS impact the activity and communication of all tumor constituents, mediating their reprogramming from anti- to protumorigenic phenotypes, and vice versa. An important role in this dichotomic action is played by the variable amounts of O2 in the tumor microenvironment, which dictates the ultimate outcome of the influence of ROS/RNS on carcinogenesis. Moreover, ROS/RNS levels remarkably influence the cancer response to therapy. The relevance of ROS/RNS signaling in solid tumors is witnessed by the emergence of novel targeted treatments of solid tumors with compounds that target ROS/RNS action and production, such as tyrosine kinase inhibitors and monoclonal antibodies, which might contribute to the complexity of redox regulation in cancer. Prospectively, the dual role of ROS/RNS in the different stages of tumorigenesis through different impact on oxidation and nitrosylation may also allow development of tailored diagnostic and therapeutic approaches.
Collapse
|
11
|
A 17-Gene Signature Predicted Prognosis in Renal Cell Carcinoma. DISEASE MARKERS 2020; 2020:8352809. [PMID: 32184905 PMCID: PMC7063218 DOI: 10.1155/2020/8352809] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 11/13/2019] [Indexed: 12/22/2022]
Abstract
Renal cell carcinoma (RCC), which was one of the most common malignant tumors in urinary system, had gradually increased incidence and mortality in recent years. Although significant advances had been made in molecular and biology research on the pathogenesis of RCC, effective treatments and prognostic indicators were still lacking. In order to predict the prognosis of RCC better, we identified 17 genes that were associated with the overall survival (OS) of RCC patients from The Cancer Genome Atlas (TCGA) dataset and a 17-gene signature was developed. Through SurvExpress, we analyzed the expression differences of the 17 genes and their correlation with the survival of RCC patients in five datasets (ZHAO, TCGA, KIPAN, KIRC, and KIRP), and then evaluated the survival prognostic significance of the 17-gene signature for RCC. Our results showed that the 17-gene signature had a predictive prognostic value not only in single pathologic RCC, but also in multiple pathologic types of RCC. In conclusion, the 17-gene signature model was related to the survival of RCC patients and could help predict the prognosis with significant clinical implications.
Collapse
|
12
|
Huang XQ, Hao S, Zhou ZQ, Huang B, Fang JY, Tang Y, Zhang JH, Xia JC. The Roles of Ubiquitination Factor E4B (UBE4B) in the Postoperative Prognosis of Patients with Renal Cell Carcinoma and in Renal Tumor Cells Growth and Metastasis. Onco Targets Ther 2020; 13:185-197. [PMID: 32021266 PMCID: PMC6956714 DOI: 10.2147/ott.s229577] [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: 09/02/2019] [Accepted: 12/24/2019] [Indexed: 12/22/2022] Open
Abstract
Object This study aimed at investigating the clinical significance and biological function of ubiquitination factor E4B (UBE4B) in human renal cell carcinoma (RCC). Methods 19 paired clear cell renal cell carcinoma (ccRCC) tumor samples and the matched neighboring non-tumor samples were used to detect the expression of UBE4B in RCC tumor by Western blotting and RT-qPCR. UBE4B expression was also detected in 151 ccRCC paraffin-embedded tumor samples by using immunohistochemistry. Overall survival (OS) in different UBE4B expression groups were compared with Log rank test. The prognostic value of UBE4B expression in OS was evaluated with the univariate and multivariate Cox regression models. UBE4B was knocked down by small interfering RNA (siRNA) technology, and the effect of UBE4B on cell proliferation, colony formation, metastasis, apoptosis and cell cycle of RCC cells were examined in vitro. Results Both protein and mRNA levels of UBE4B were up-regulated in ccRCC tumor tissues in contrast to the corresponding adjacent nontumor ones. UBE4B expression was positively associated with tumor-node-metastasis (TNM) stage and distant metastasis in ccRCC patients. Survival analyses indicated that low expression of UBE4B was associated with increased OS in ccRCC patients. Functional analyses demonstrated that siRNA silencing of UBE4B expression in SKRC39 and ACHN cells further reduced the growth, motility and invasiveness of RCC cells. Moreover, siRNA silencing of UBE4B in the RCC cell lines did not induce apoptosis, and an increase in the cell population was observed during the G0/G1 phase of the cell cycle. Conclusion UBE4B might act as an oncogene in regulating RCC development. Therefore it could be served as an effective indicator to predict OS and a potential biomarker for targeted therapy of RCC patients.
Collapse
Affiliation(s)
- Xu-Qiong Huang
- Huadu District People's Hospital of Guangzhou, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Shuai Hao
- Huadu District People's Hospital of Guangzhou, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Zi-Qi Zhou
- State Key Laboratory of Oncology in Southern China, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong Province, People's Republic of China
| | - Bin Huang
- Huadu District People's Hospital of Guangzhou, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Jia-Ying Fang
- Huadu District People's Hospital of Guangzhou, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Yan Tang
- State Key Laboratory of Oncology in Southern China, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong Province, People's Republic of China
| | - Jian-Hua Zhang
- Department of Health Service Management, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, People's Republic of China
| | - Jian-Chuan Xia
- State Key Laboratory of Oncology in Southern China, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong Province, People's Republic of China
| |
Collapse
|
13
|
Yeong J, Zhao Z, Lim JCT, Li H, Thike AA, Koh VCY, Teh BT, Kanesvaran R, Toh CK, Tan PH, Khor LY. PD-L1 expression is an unfavourable prognostic indicator in Asian renal cell carcinomas. J Clin Pathol 2020; 73:463-469. [PMID: 31980560 DOI: 10.1136/jclinpath-2019-206092] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/25/2019] [Accepted: 11/26/2019] [Indexed: 01/15/2023]
Abstract
BACKGROUND/AIMS The programmed cell death receptor 1 (PD-1) checkpoint inhibitor, nivolumab, has been approved for the treatment of metastatic renal cell carcinoma (RCC). However, the understanding of the expression and distribution of PD ligand 1 (PD-L1) in the tumour immune microenvironment and its prognostic role in an Asian cohort is limited. Our group investigated PD-L1 protein expression in a cohort of Asian patients with RCC of mixed ethnicity, using two commercially available antibody clones. METHODS E1L3N and SP263 anti-PD-L1 clones were used to categorise RCCs of various histological subtypes, diagnosed at our institution between 1995 and 2008, into PD-L1-positive or PD-L1-negative groups, based on a 1% Tumour Proportion Score (TPS) cut-off. RESULTS In total, 267 (83%) clear cell (cc)RCC and 55 (17%) non-ccRCC cases were studied. Overall PD-L1 protein expression rates for the entire cohort were 13% and 8% for the E1L3N and SP263 clones, respectively. Patients bearing PD-L1-positive tumours experienced significantly decreased disease-free survival (DFS; E1L3N: p=0.01; SP263: p=0.03) but not overall survival, compared with those with PD-L1-negative tumours. Multivariate survival analysis further confirmed the results of the E1L3N clone (HR 1.85, 95% CI 1.10 to 3.13, p=0.02), but not SP263, after adjusting for pathological stage, histological subtype and grade. The addition of PD-L1 (E1L3N) TPS to clinicopathological features significantly increased the prognostic value for DFS (∆LRχ2=5.25; p=0.022), compared with clinicopathological features alone. CONCLUSIONS PD-L1 protein expression was associated with an unfavourable prognosis in our study cohort. PD-L1 (E1L3N) expression was an independent prognostic indicator of clinical outcome in all RCCs when using a 1% cut-off.
Collapse
Affiliation(s)
- Joe Yeong
- Institute of Molecular Cell Biology (IMCB), Agency of Science, Technology and Research (A*STAR), Singapore.,Anatomical Pathology, Singapore General Hospital, Singapore
| | - Zitong Zhao
- Anatomical Pathology, Singapore General Hospital, Singapore
| | - Jeffrey Chun Tatt Lim
- Institute of Molecular Cell Biology (IMCB), Agency of Science, Technology and Research (A*STAR), Singapore.,Pathology, Singapore General Hospital, Singapore
| | - Huihua Li
- Center for Quantitative Medicine, Duke-NUS Medical School, Singapore.,Division of Medicine, Singapore General Hospital, Singapore
| | | | | | - Bin Tean Teh
- Laboratory of Cancer Epigenome, National Cancer Centre, Singapore.,Division of Cancer and Stem Cell Biology, Duke-NUS Graduate Medical School, Singapore
| | - Ravindran Kanesvaran
- Dean's Office, Duke-NUS Medical School, Singapore.,National Cancer Centre Singapore, Singapore
| | | | - Puay Hoon Tan
- Anatomical Pathology, Singapore General Hospital, Singapore.,Pathology, Singapore General Hospital, Singapore.,Dean's Office, Duke-NUS Medical School, Singapore
| | - Li Yan Khor
- Anatomical Pathology, Singapore General Hospital, Singapore .,Dean's Office, Duke-NUS Medical School, Singapore
| |
Collapse
|
14
|
Stenzel PJ, Schindeldecker M, Tagscherer KE, Foersch S, Herpel E, Hohenfellner M, Hatiboglu G, Alt J, Thomas C, Haferkamp A, Roth W, Macher-Goeppinger S. Prognostic and Predictive Value of Tumor-infiltrating Leukocytes and of Immune Checkpoint Molecules PD1 and PDL1 in Clear Cell Renal Cell Carcinoma. Transl Oncol 2019; 13:336-345. [PMID: 31881506 PMCID: PMC7031108 DOI: 10.1016/j.tranon.2019.11.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 11/06/2019] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION: Immune checkpoint inhibitors (ICI) have been approved for patients with clear cell renal cell carcinoma (ccRCC), but not all patients benefit from ICI. One reason is the tumor microenvironment (TME) that has substantial influence on patient's prognosis and therapy response. Thus, we comprehensively analyzed the TME of ccRCC regarding prognostic and predictive properties. METHODS: Tumor-infiltrating CD3-positive T-cells, CD8-positive cytotoxic T-lymphocytes (CTLs), regulatory T-cells, B-cells, plasma cells, macrophages, granulocytes, programmed cell death receptor 1 (PD-1), and its ligand PD-L1 were examined in a large hospital-based series of ccRCC with long-term follow-up information (n = 756) and in another patient collective with information on response to nivolumab therapy (n = 8). Tissue microarray technique and digital image analysis were used. Relationship between immune cell infiltration and tumor characteristics, cancer-specific survival (CSS), or response to ICI was examined. RESULTS: Univariate survival analysis revealed that increased tumor-infiltrating B-cells, T-cells, and PD-1-positive cells were significantly associated with favorable CSS and high levels of intratumoral granulocytes, macrophages, cytotoxic T-cells, and PD-L1 significantly with poor CSS. High CTL or B-cell infiltration and high PD-L1 expression of ccRCC tumor cells qualified as independent prognostic biomarkers for patients' CSS. Significantly higher densities of intratumoral T-cells, CTLs, and PD-1-positive immune cells were observed in ccRCC with response to ICI compared with patients with mixed or no response (CD3: p = 0.003; CD8: p = 0.006; PD-1: p = 0.01). DISCUSSION: This study shows that subsets of tumor-infiltrating leukocytes in the TME and also PD-1/PD-L1 provide prognostic and predictive information for patients with ccRCC.
Collapse
Affiliation(s)
- Philipp J Stenzel
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany.
| | - Mario Schindeldecker
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany; Tissue Biobank, University Medical Center Mainz, Mainz, Germany
| | | | - Sebastian Foersch
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Esther Herpel
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany; Tissue Bank of the National Center for Tumor Diseases, Heidelberg, Germany
| | | | - Gencay Hatiboglu
- Department of Urology, University Hospital Heidelberg, Heidelberg, Germany
| | - Juergen Alt
- Department of Hematology, Medical Oncology & Pneumology, University Medical Center Mainz, Mainz, Germany
| | - Christian Thomas
- Department of Urology, University Medical Center Mainz, Germany; Department of Urology, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Axel Haferkamp
- Department of Urology, University Medical Center Mainz, Germany
| | - Wilfried Roth
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Stephan Macher-Goeppinger
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany; Tissue Biobank, University Medical Center Mainz, Mainz, Germany
| |
Collapse
|
15
|
George S, Rini BI, Hammers HJ. Emerging Role of Combination Immunotherapy in the First-line Treatment of Advanced Renal Cell Carcinoma: A Review. JAMA Oncol 2019; 5:411-421. [PMID: 30476955 DOI: 10.1001/jamaoncol.2018.4604] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Importance Novel immunotherapies, notably the immune checkpoint inhibitors, have been shown to be efficacious in patients with advanced renal cell carcinoma, but innate or adaptive resistance is observed with single-agent immunotherapy. New combination treatment strategies are needed that can improve efficacy in a broader patient population, without exacerbating the toxic effects. Observations Numerous late-phase trials are ongoing to investigate (1) dual immune checkpoint inhibition or (2) combined inhibition of immune checkpoints and vascular endothelial growth factor. Initial results from studies of the nivolumab plus ipilimumab and atezolizumab plus bevacizumab combinations have demonstrated efficacy compared with sunitinib malate in treatment-naïve patients with advanced renal cell carcinoma; moreover, the safety profile of these combinations compare favorably with sunitinib. Nevertheless, immune checkpoint inhibition is associated with unique immune-related adverse events, and practicing physicians must be educated on how to best identify and manage these events. Conclusions and Relevance Evidence suggests that immunotherapy-based combination regimens will be an important addition to the treatment of advanced renal cell carcinoma in both the first- and later-line setting; however, clinical study data and clinical practice experience indicate that optimizing the management of the associated immune-related adverse events is essential to maximizing the advantages of these therapies.
Collapse
Affiliation(s)
- Saby George
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Brian I Rini
- Department of Hematology and Medical Oncology, Cleveland Clinic Taussig Cancer Institute, Cleveland, Ohio
| | - Hans J Hammers
- Department of Internal Medicine, UT Southwestern, Kidney Cancer Program, Dallas, Texas
| |
Collapse
|
16
|
Saeednejad Zanjani L, Madjd Z, Rasti A, Asgari M, Abolhasani M, Tam KJ, Roudi R, Mælandsmo GM, Fodstad Ø, Andersson Y. Spheroid-Derived Cells From Renal Adenocarcinoma Have Low Telomerase Activity and High Stem-Like and Invasive Characteristics. Front Oncol 2019; 9:1302. [PMID: 31921617 PMCID: PMC6915099 DOI: 10.3389/fonc.2019.01302] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Accepted: 11/11/2019] [Indexed: 12/12/2022] Open
Abstract
Cancer stem cells (CSCs) are a theorized small subpopulation of cells within tumors thought to be responsible for metastasis, tumor development, disease progression, treatment-resistance, and recurrence. The identification, isolation, and biological characterization of CSCs may therefore facilitate the development of efficient therapeutic strategies targeting CSCs. This study aims to compare the biology and telomerase activity of CSCs to parental cells (PCs) in renal cancer. Renal CSCs were enriched from the ACHN cell line using a sphere culture system. Spheroid-derived cells (SDCs) and their adherent counterparts were compared with respect to their colony and sphere formation, expression of putative CSC markers, tumorigenicity in non-obese diabetic/severe combined immunodeficiency (NOD/SCID) mice, and invasiveness. The expression of genes associated with CSCs, stemness, EMT, apoptosis, and ABC transporters was also compared between the two populations using quantitative real-time PCR (qRT-PCR). Finally, telomerase activity, hTERT expression, and sensitivity to MST-312, a telomerase inhibitor, was investigated between the two populations. We demonstrated that a subpopulation of ACHN cells was capable of growing as spheroids with many properties similar to CSCs, including higher clonogenicity, superior colony- and sphere-forming ability, and stronger tumorigenicity and invasiveness. In addition, SDCs demonstrated a higher expression of markers for CSCs, stemness, EMT, apoptosis, and ABC transporter genes compared to PCs. The expression of hTERT and telomerase activity in SDCs was significantly lower than PCs; however, the SDC population was more sensitive to MST-312 compared to PCs. These findings indicate that the SDC population exhibits stem-like potential and invasive characteristics. Moreover, the reduced expression of hTERT and telomerase activity in SDCs demonstrated that the expressions of hTERT and telomerase activity are not always higher in CSCs. Our results also showed that MST-312 treatment inhibited SDCs more strongly than PCs and may therefore be useful as a complementary targeted therapy against renal CSCs in the future.
Collapse
Affiliation(s)
- Leili Saeednejad Zanjani
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.,Department of Urologic Sciences, Vancouver Prostate Center, University of British Columbia, Vancouver, BC, Canada
| | - Arezoo Rasti
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Department of Basic Sciences/Medical Surgical Nursing, Faculty of Nursing and Midwifery, Tehran University of Medical Sciences, Tehran, Iran
| | - Mojgan Asgari
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Hasheminejad Kidney Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Maryam Abolhasani
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Hasheminejad Kidney Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Kevin J Tam
- Department of Urologic Sciences, Vancouver Prostate Center, University of British Columbia, Vancouver, BC, Canada
| | - Raheleh Roudi
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Gunhild Mari Mælandsmo
- Department of Tumor Biology, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Øystein Fodstad
- Department of Tumor Biology, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Yvonne Andersson
- Department of Tumor Biology, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| |
Collapse
|
17
|
Labadie BW, Liu P, Bao R, Crist M, Fernandes R, Ferreira L, Graupner S, Poklepovic AS, Duran I, Maleki Vareki S, Balar AV, Luke JJ. BMI, irAE, and gene expression signatures associate with resistance to immune-checkpoint inhibition and outcomes in renal cell carcinoma. J Transl Med 2019; 17:386. [PMID: 31767020 PMCID: PMC6878694 DOI: 10.1186/s12967-019-02144-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 11/18/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Clinical variables may correlate with lack of response to treatment (primary resistance) or clinical benefit in patients with clear cell renal cell carcinoma (ccRCC) treated with anti-programmed death 1/ligand one antibodies. METHODS In this multi-institutional collaboration, clinical characteristics of patients with primary resistance (defined as progression on initial computed tomography scan) were compared to patients with clinical benefit using Two sample t-test and Chi-square test (or Fisher's Exact test). The Kaplan-Meier method was used to estimate the distribution of progression-free survival (PFS) and overall survival (OS) in all patients and the subsets of patients with clinical benefit or primary resistance. Cox's regression model was used to evaluate the correlation between survival endpoints and variables of interest. To explore clinical factors in a larger, independent patient sample, The Cancer Genome Atlas (TCGA) was analyzed. RNAseq gene expression data as well as demographic and clinical information were downloaded for primary tumors of 517 patients included within TCGA-ccRCC. RESULTS Of 90 patients, 38 (42.2%) had primary resistance and 52 (57.8%) had clinical benefit. Compared with the cohort of patients with initial benefit, primary resistance was more likely to occur in patients with worse ECOG performance status (p = 0.03), earlier stage at diagnosis (p = 0.04), had no prior nephrectomy (p = 0.04) and no immune-related adverse events (irAE) (p = 0.02). In patients with primary resistance, improved OS was significantly correlated with lower International Metastatic RCC Database Consortium risk score (p = 0.02) and lower neutrophil:lymphocyte ratio (p = 0.04). In patients with clinical benefit, improved PFS was significantly associated with increased BMI (p = 0.007) and irAE occurrence (p = 0.02) while improved OS was significantly correlated with overweight BMI (BMI 25-30; p = 0.03) and no brain metastasis (p = 0.005). The cohort TCGA-ccRCC was examined for the correlations between gene expression patterns, clinical factors, and survival outcomes observing associations of T-cell inflammation and angiogenesis signatures with histologic grade, pathologic stage and OS. CONCLUSIONS Clinical characteristics including performance status, BMI and occurrence of an irAE associate with outcomes in patients with ccRCC treated with immunotherapy. The inverse association of angiogenesis gene signature with ccRCC histologic grade highlight opportunities for adjuvant combination VEGFR2 tyrosine kinase inhibitor and immune-checkpoint inhibition.
Collapse
Affiliation(s)
- Brian W Labadie
- Department of Internal Medicine, University of Chicago, Chicago, IL, USA
| | - Ping Liu
- Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
| | - Riyue Bao
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Division of Hematology/Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael Crist
- Division of Hematology/Oncology, New York University School of Medicine and Langone Medical Center, New York, NY, USA
| | - Ricardo Fernandes
- Division of Medical Oncology, Schulich School of Medicine & Dentistry, Western University and London Health Sciences Centre, London, ON, Canada
| | - Laura Ferreira
- Division of Medical Oncology, University Hospital Marques de Valdecilla, Cantabria, Spain
| | - Scott Graupner
- Division of Hematology/Oncology, Virginia Commonwealth University, Richmond, VA, USA
| | - Andrew S Poklepovic
- Division of Hematology/Oncology, Virginia Commonwealth University, Richmond, VA, USA
| | - Ignacio Duran
- Division of Medical Oncology, University Hospital Marques de Valdecilla, Cantabria, Spain
| | - Saman Maleki Vareki
- Division of Medical Oncology, Schulich School of Medicine & Dentistry, Western University and London Health Sciences Centre, London, ON, Canada
| | - Arjun V Balar
- Division of Hematology/Oncology, New York University School of Medicine and Langone Medical Center, New York, NY, USA
| | - Jason J Luke
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
- Division of Hematology/Oncology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
18
|
Mai AT, Tong WL, Tu YN, Blanck G. TcR-α recombinations in renal cell carcinoma exome files correlate with an intermediate level of T-cell exhaustion biomarkers. Int Immunol 2019; 30:35-40. [PMID: 29361059 DOI: 10.1093/intimm/dxx074] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Renal cell carcinoma exome-derived, V(D)J recombination reads had an elevated presence and variability, for both TcR-α and -β, when compared to marginal tissue, reflecting an opportunity to assess tumor immunogenicity by comparison with marginal tissue T cells. PD-1, PD-L2, CTLA4 and FOXP3, all of which are implicated in the evasion of an anti-tumor immune response, had a significantly higher expression for samples representing co-detection of productive TcR-α and -β recombination reads. Samples representing tumors with productive TcR-α recombination reads but no detectable, productive TcR-β recombination reads, reflected a 20% survival advantage, and RNASeq data indicated an intermediate level of immune checkpoint gene expression for those samples. These results raise the question of whether relatively high levels of detection of productive TcR-α recombination reads, in comparison with detection of reads representing the TcR-β gene, identify a microenvironment that has not yet entered a T-cell exhaustion phase and may thereby represent conditions for immune enhancements that do not require anti-immune checkpoint therapies.
Collapse
Affiliation(s)
- Anne T Mai
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, USA
| | - Wei Lue Tong
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, USA
| | - Yaping N Tu
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, USA
| | - George Blanck
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, USA.,Immunology Program, USF Magnolia Dr., H. Lee Moffitt Cancer Center and Research Institute, FL, USA
| |
Collapse
|
19
|
Wang T, Lu R, Kapur P, Jaiswal BS, Hannan R, Zhang Z, Pedrosa I, Luke JJ, Zhang H, Goldstein LD, Yousuf Q, Gu YF, McKenzie T, Joyce A, Kim MS, Wang X, Luo D, Onabolu O, Stevens C, Xie Z, Chen M, Filatenkov A, Torrealba J, Luo X, Guo W, He J, Stawiski E, Modrusan Z, Durinck S, Seshagiri S, Brugarolas J. An Empirical Approach Leveraging Tumorgrafts to Dissect the Tumor Microenvironment in Renal Cell Carcinoma Identifies Missing Link to Prognostic Inflammatory Factors. Cancer Discov 2018; 8:1142-1155. [PMID: 29884728 PMCID: PMC6125163 DOI: 10.1158/2159-8290.cd-17-1246] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 03/01/2018] [Accepted: 06/07/2018] [Indexed: 01/05/2023]
Abstract
By leveraging tumorgraft (patient-derived xenograft) RNA-sequencing data, we developed an empirical approach, DisHet, to dissect the tumor microenvironment (eTME). We found that 65% of previously defined immune signature genes are not abundantly expressed in renal cell carcinoma (RCC) and identified 610 novel immune/stromal transcripts. Using eTME, genomics, pathology, and medical record data involving >1,000 patients, we established an inflamed pan-RCC subtype (IS) enriched for regulatory T cells, natural killer cells, TH1 cells, neutrophils, macrophages, B cells, and CD8+ T cells. IS is enriched for aggressive RCCs, including BAP1-deficient clear-cell and type 2 papillary tumors. The IS subtype correlated with systemic manifestations of inflammation such as thrombocytosis and anemia, which are enigmatic predictors of poor prognosis. Furthermore, IS was a strong predictor of poor survival. Our analyses suggest that tumor cells drive the stromal immune response. These data provide a missing link between tumor cells, the TME, and systemic factors.Significance: We undertook a novel empirical approach to dissect the renal cell carcinoma TME by leveraging tumorgrafts. The dissection and downstream analyses uncovered missing links between tumor cells, the TME, systemic manifestations of inflammation, and poor prognosis. Cancer Discov; 8(9); 1142-55. ©2018 AACR.This article is highlighted in the In This Issue feature, p. 1047.
Collapse
Affiliation(s)
- Tao Wang
- Quantitative Biomedical Research Center, Department of Clinical Sciences, The University of Texas Southwestern Medical Center, Dallas, Texas.
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
- Center for the Genetics of Host Defense, The University of Texas Southwestern Medical Center, Texas
| | - Rong Lu
- Quantitative Biomedical Research Center, Department of Clinical Sciences, The University of Texas Southwestern Medical Center, Dallas, Texas
- Bioinformatics Core Facility, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Payal Kapur
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Bijay S Jaiswal
- Molecular Biology Department, Genentech, Inc., South San Francisco, California
| | - Raquibul Hannan
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ze Zhang
- Quantitative Biomedical Research Center, Department of Clinical Sciences, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ivan Pedrosa
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Radiology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jason J Luke
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - He Zhang
- Bioinformatics Core Facility, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Leonard D Goldstein
- Molecular Biology Department, Genentech, Inc., South San Francisco, California
| | - Qurratulain Yousuf
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Yi-Feng Gu
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Tiffani McKenzie
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Allison Joyce
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Min S Kim
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
- Bioinformatics Core Facility, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Xinlei Wang
- Department of Statistical Science, Southern Methodist University, Dallas, Texas
| | - Danni Luo
- Bioinformatics Core Facility, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Oreoluwa Onabolu
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Christina Stevens
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Zhiqun Xie
- Quantitative Biomedical Research Center, Department of Clinical Sciences, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Mingyi Chen
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Alexander Filatenkov
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jose Torrealba
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Xin Luo
- Bioinformatics Core Facility, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Wenbin Guo
- Quantitative Biomedical Research Center, Department of Clinical Sciences, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jingxuan He
- Quantitative Biomedical Research Center, Department of Clinical Sciences, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Eric Stawiski
- Molecular Biology Department, Genentech, Inc., South San Francisco, California
- Bioinformatics and Computational Biology Department, Genentech, Inc., South San Francisco, California
| | - Zora Modrusan
- Molecular Biology Department, Genentech, Inc., South San Francisco, California
| | - Steffen Durinck
- Molecular Biology Department, Genentech, Inc., South San Francisco, California
- Bioinformatics and Computational Biology Department, Genentech, Inc., South San Francisco, California
| | - Somasekar Seshagiri
- Molecular Biology Department, Genentech, Inc., South San Francisco, California.
| | - James Brugarolas
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas.
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
20
|
Lesueur P, Lequesne J, Barraux V, Kao W, Geffrelot J, Grellard JM, Habrand JL, Emery E, Marie B, Thariat J, Stefan D. Radiosurgery or hypofractionated stereotactic radiotherapy for brain metastases from radioresistant primaries (melanoma and renal cancer). Radiat Oncol 2018; 13:138. [PMID: 30055640 PMCID: PMC6064124 DOI: 10.1186/s13014-018-1083-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 07/20/2018] [Indexed: 01/08/2023] Open
Abstract
Background Until 50% of patients with renal cancer or melanoma, develop brain metastases during the course of their disease. Stereotactic radiotherapy has become a standard of care for patients with a limited number of brain metastases. Given the radioresistant nature of melanoma and renal cancer, optimization of the fractionation of stereotactic radiotherapy is needed. The purpose of this retrospective study was to elucidate if hypofractionated stereotactic radiotherapy (HFSRT) impacts local control of brain metastases from radioresistant tumors such as melanoma and renal cancer, in comparison with radiosurgery (SRS). Methods Between 2012 and 2016, 193 metastases, smaller than 3 cm, from patients suffering from radioresistant primaries (melanoma and renal cancer) were treated with HFSRT or SRS. The primary outcome was local progression free survival (LPFS) at 6, 12 and 18 months. Overall survival (OS) and cerebral progression free survival (CPFS) were secondary outcomes, and were evaluated per patient. Objective response rate and radionecrosis incidence were also reported. The statistical analysis included a supplementary propensity score analysis to deal with bias induced by non-randomized data. Results After a median follow-up of 7.4 months, LPFS rates at 6, 12 and 18 months for the whole population were 83, 74 and 70%, respectively. With respect to fractionation, LPFS rates at 6, 12 and 18 months were 89, 79 and 73% for the SRS group and 80, 72 and 68% for the HFSRT group. The fractionation schedule was not statistically associated with LPFS (HR = 1.39, CI95% [0.65–2.96], p = 0.38). Time from planning MRI to first irradiation session longer than 14 days was associated with a poorer local control rate. Over this time, LPFS at 12 months was reduced from 86 to 70% (p = 0.009). Radionecrosis occurred in 7.1% for HFSRT treated metastases to 9.6% to SRS treated metastases, without any difference according to fractionation (p = 0.55). The median OS was 9.6 months. Six, 12 and 18 months CPFS rates were 54, 24 and 17%, respectively. Conclusion Fractionation does not decrease LPFS. Even for small radioresistant brain metastases (< 3 cm), HFSRT, with 3 or 6 fractions, leads to an excellent local control rate of 72% at 1 year with a rate of 7.1% of radionecrosis. HFSRT is a safe and efficient alternative treatment to SRS. Electronic supplementary material The online version of this article (10.1186/s13014-018-1083-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Paul Lesueur
- Radiotherapy department, Centre François Baclesse, Caen, France. .,Laboratoire d'accueil et de recherche avec les ions accélérés, CEA-CIMAP, Caen, France. .,Medical university of Caen, Caen, France.
| | - Justine Lequesne
- Clinical research department, Centre François Baclesse, Caen, France
| | - Victor Barraux
- Medical physics department, Centre François Baclesse, Caen, France
| | - William Kao
- Radiotherapy department, Centre François Baclesse, Caen, France
| | | | | | - Jean-Louis Habrand
- Radiotherapy department, Centre François Baclesse, Caen, France.,Medical university of Caen, Caen, France
| | - Evelyne Emery
- Neurosurgery department, CHU Côte de Nacre, Caen, France.,Medical university of Caen, Caen, France
| | - Brigitte Marie
- Imaging department, Centre François Baclesse, Caen, France
| | - Juliette Thariat
- Radiotherapy department, Centre François Baclesse, Caen, France.,Medical university of Caen, Caen, France
| | - Dinu Stefan
- Radiotherapy department, Centre François Baclesse, Caen, France
| |
Collapse
|
21
|
Cimadamore A, Gasparrini S, Santoni M, Cheng L, Lopez-Beltran A, Battelli N, Massari F, Giunchi F, Fiorentino M, Scarpelli M, Montironi R. Biomarkers of aggressiveness in genitourinary tumors with emphasis on kidney, bladder, and prostate cancer. Expert Rev Mol Diagn 2018; 18:645-655. [PMID: 29912582 DOI: 10.1080/14737159.2018.1490179] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Over the last decade, the improvement in molecular techniques and the acquisition of genomic information has transformed and increased the quality of patient care and our knowledge of diseases. Areas covered: Protein expression levels in immunohistochemistry and molecular biomarkers are reported for their ability to predict recurrence, progression, development of metastases, or patient survival. In particular, for renal cell carcinoma, we take into consideration the biomarkers applicable to immunohistochemistry and with molecular and genetic analyses. In urothelial carcinoma, there is great interest in the possibility of distinguishing the basal vs. luminal subtypes and to acquire deeper insight into the tumor biology through examining exosomes in urine and biomarkers in the serum. In prostate cancer, single gene expression and multiple gene expression classifiers are reviewed as a tool to distinguish indolent vs. aggressive disease. Expert commentary: The genomic information along with the application of ancillary techniques allow the definition of a neoplasia not only by its morphology but also by its biological signature. This continuous increase in knowledge will result in a better comprehension of oncogenesis, development of targeted therapies and optimizing decision-making processes related to patient care.
Collapse
Affiliation(s)
- Alessia Cimadamore
- a Section of Pathological Anatomy, School of Medicine, United Hospitals , Polytechnic University of the Marche Region , Ancona , Italy
| | - Silvia Gasparrini
- a Section of Pathological Anatomy, School of Medicine, United Hospitals , Polytechnic University of the Marche Region , Ancona , Italy
| | | | - Liang Cheng
- c Department of Pathology and Laboratory Medicine , Indiana University School of Medicine , Indianapolis , IN , USA
| | | | | | - Francesco Massari
- e Division of Oncology , S. Orsola-Malpighi Hospital , Bologna , Italy
| | - Francesca Giunchi
- f Laboratory of Oncologic Molecular Pathology , S. Orsola-Malpighi Hospital , Bologna , Italy
| | - Michelangelo Fiorentino
- f Laboratory of Oncologic Molecular Pathology , S. Orsola-Malpighi Hospital , Bologna , Italy
| | - Marina Scarpelli
- a Section of Pathological Anatomy, School of Medicine, United Hospitals , Polytechnic University of the Marche Region , Ancona , Italy
| | - Rodolfo Montironi
- a Section of Pathological Anatomy, School of Medicine, United Hospitals , Polytechnic University of the Marche Region , Ancona , Italy
| |
Collapse
|
22
|
GV1001 Induces Apoptosis by Reducing Angiogenesis in Renal Cell Carcinoma Cells Both In Vitro and In Vivo. Urology 2018; 113:129-137. [PMID: 29154986 DOI: 10.1016/j.urology.2017.10.038] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 09/19/2017] [Accepted: 10/28/2017] [Indexed: 12/31/2022]
Abstract
OBJECTIVE To investigate the anticancer effects of GV1001 and its biological mechanism of action in renal cell carcinoma (RCC). METHODS The effects of GV1001 on cell survival and apoptosis in RCC cells were examined in vitro using cell viability assay, fluorescence-activated cell sorting, and terminal deoxynucleotidyl transferase dUTP nick end labeling assay. To evaluate the effect of GV1001 on migration, invasion, and angiogenesis, we used wound healing, invasion, endothelial cell tube formation assay, and western blot analysis. Furthermore, we used an RCC xenograft model with either phosphate buffered saline or GV1001 to confirm the anticancer effect of GV1001 in vivo. Tumor volume was monitored during treatment, and tumor weight was measured after animals were killed. Apoptosis and angiogenesis of the tumor tissue were assessed using hematoxylin and eosin staining, immunohistochemistry, and western blot analysis. RESULTS GV1001 reduced cell viability and induced apoptosis in RCC cells in vitro. Furthermore, GV1001 suppressed the migration and invasion of RCC cells through regulation of matrix metalloproteinases and tissue inhibitors of metalloproteinases. In addition, GV1001 reduced angiogenesis via regulation of hypoxia-inducible factor 1α. In xenograft mouse model experiment, GV1001 reduced tumor growth and induced apoptosis. As in the in vitro results, GV1001 significantly reduced angiogenesis through regulation of hypoxia-inducible factor 1α in vivo. CONCLUSION Our data demonstrated that GV1001 induced apoptosis through suppression of angiogenesis in RCCs both in vitro and in vivo, which suggests that GV1001 may be a potential therapeutic target for RCC.
Collapse
|
23
|
Abstract
Harnessing the host immune system to combat genitourinary cancers has key theoretical advantages over other anticancer strategies including specificity and memory which should translate to favorable tolerability and response durability in the clinic. Indeed, key examples of the potential for immunotherapeutic treatment of solid tumors are derived from data in genitourinary cancers including Bacillus Calmette-Guerin for urothelial cancer, sipuleucel-T for prostate cancer, and interleukin-2 for renal cancer. Despite these successes, developing effective immunotherapeutic strategies for the treatment of cancer has largely been hampered by an incomplete understanding of tumor immunobiology and mechanisms of immune resistance. In just a few years since entering the clinic, immune checkpoint blockade has dramatically changed the landscaped of treatment for genitourinary cancer and has secured a place as a standard pillar of treatment. Further iterative bench-bedside-bench research is anticipated to extend the benefits of immunotherapeutic-based approaches to additional patients.
Collapse
Affiliation(s)
- Kyrollis Attalla
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John P Sfakianos
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew D Galsky
- Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Genitourinary Medical Oncology, Tisch Cancer Institute, New York, NY, USA.
| |
Collapse
|
24
|
Ross K, Jones RJ. Immune checkpoint inhibitors in renal cell carcinoma. Clin Sci (Lond) 2017; 131:2627-2642. [PMID: 29079639 PMCID: PMC5869245 DOI: 10.1042/cs20160894] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 09/12/2017] [Accepted: 09/13/2017] [Indexed: 01/05/2023]
Abstract
The immune system has long been known to play a critical role in the body's defence against cancer, and there have been multiple attempts to harness it for therapeutic gain. Renal cancer was, historically, one of a small number of tumour types where immune manipulation had been shown to be effective. The current generation of immune checkpoint inhibitors are rapidly entering into routine clinical practice in the management of a number of tumour types, including renal cancer, where one drug, nivolumab, an anti-programmed death-1 (PD-1) monoclonal antibody (mAb), is licensed for patients who have progressed on prior systemic treatment. Ongoing trials aim to maximize the benefits that can be gained from this new class of drug by exploring optimal timing in the natural course of the disease as well as combinations with other checkpoint inhibitors and drugs from different classes.
Collapse
Affiliation(s)
- Kirsty Ross
- Department of Oncology, Beatson West of Scotland Cancer Centre, Glasgow G12 0YN, U.K
| | - Rob J Jones
- Institute of Cancer Sciences, University of Glasgow, Beatson West of Scotland Cancer Centre, University of Glasgow, Glasgow G12 0YN, U.K.
| |
Collapse
|
25
|
Reactive Oxygen Species-Mediated Mechanisms of Action of Targeted Cancer Therapy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017. [PMID: 28698765 DOI: 10.1155/2017/1485283,] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Targeted cancer therapies, involving tyrosine kinase inhibitors and monoclonal antibodies, for example, have recently led to substantial prolongation of survival in many metastatic cancers. Compared with traditional chemotherapy and radiotherapy, where reactive oxygen species (ROS) have been directly linked to the mediation of cytotoxic effects and adverse events, the field of oxidative stress regulation is still emerging in targeted cancer therapies. Here, we provide a comprehensive review regarding the current evidence of ROS-mediated effects of antibodies and tyrosine kinase inhibitors, use of which has been indicated in the treatment of solid malignancies and lymphomas. It can be concluded that there is rapidly emerging evidence of ROS-mediated effects of some of these compounds, which is also relevant in the context of drug resistance and how to overcome it.
Collapse
|
26
|
Teppo HR, Soini Y, Karihtala P. Reactive Oxygen Species-Mediated Mechanisms of Action of Targeted Cancer Therapy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:1485283. [PMID: 28698765 PMCID: PMC5494102 DOI: 10.1155/2017/1485283] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 05/15/2017] [Accepted: 05/21/2017] [Indexed: 01/21/2023]
Abstract
Targeted cancer therapies, involving tyrosine kinase inhibitors and monoclonal antibodies, for example, have recently led to substantial prolongation of survival in many metastatic cancers. Compared with traditional chemotherapy and radiotherapy, where reactive oxygen species (ROS) have been directly linked to the mediation of cytotoxic effects and adverse events, the field of oxidative stress regulation is still emerging in targeted cancer therapies. Here, we provide a comprehensive review regarding the current evidence of ROS-mediated effects of antibodies and tyrosine kinase inhibitors, use of which has been indicated in the treatment of solid malignancies and lymphomas. It can be concluded that there is rapidly emerging evidence of ROS-mediated effects of some of these compounds, which is also relevant in the context of drug resistance and how to overcome it.
Collapse
Affiliation(s)
- Hanna-Riikka Teppo
- Department of Pathology, Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Ylermi Soini
- Department of Pathology, Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Peeter Karihtala
- Department of Oncology and Radiotherapy, Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| |
Collapse
|
27
|
Hyper IgE Syndrome and Renal Cell Carcinoma. Case Rep Urol 2017; 2017:7083451. [PMID: 28607797 PMCID: PMC5451772 DOI: 10.1155/2017/7083451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 04/26/2017] [Indexed: 11/25/2022] Open
Abstract
Hyper IgE Syndrome (HIES) is an immunodeficiency disorder characterized by increased serum levels of IgE, eczema, and recurrent cutaneous and pulmonary infections. In this report, we present, to our knowledge, the first documented case of renal cell carcinoma (RCC) found in a patient with HIES. The patient received infectious disease clearance prior to obtaining a partial nephrectomy which revealed clear cell histology. Both HIES and RCC have an immunological basis for their pathophysiology and may involve common pathways. Further studies may provide insight into any possible link and clinicians should be mindful of immunocompromised patients who present with risk factors for genitourinary malignancy.
Collapse
|
28
|
Wang H, Luo F, Zhu Z, Xu Z, Huang X, Ma R, He H, Zhu Y, Shao K, Zhao J. ABCG2 is a potential prognostic marker of overall survival in patients with clear cell renal cell carcinoma. BMC Cancer 2017; 17:222. [PMID: 28347288 PMCID: PMC5368932 DOI: 10.1186/s12885-017-3224-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 03/23/2017] [Indexed: 12/18/2022] Open
Abstract
Background ATP-binding cassette sub-family G member 2 (ABCG2) is a semi-transport protein that plays a major role in multidrug resistance. We aimed to evaluate the prognostic significance of ABCG2 expression in patients with clear cell renal cell carcinoma. Methods From 2008 to 2013, 120 patients with clear cell kidney cancer underwent surgery with paraffin-embedded specimens and necessary clinical information available. Immunohistochemistry staining was performed to grade the expression of ABCG2 as ABCG2(−): less than 10% of tumor cells stained; ABCG2(+): weak membrane staining; and ABCG2(++): moderate or strong membrane staining. The overall survival was analyzed using Kaplan-Meier method. Multivariable Cox regression evaluated the independent predictors for overall survival. Results ABCG2(−) was diagnosed in 57 (48%) patients, ABCG2(+) in 52 (43%) patients, and ABCG2 (++) in 11(9.2%) patients. ABCG2 expression significantly correlated with the five-year survival (p < 0.001) and distant metastasis (p = 0.001). In the multivariable analysis, besides Fuhrman grade, the ABCG2 expression was an independent prognostic marker for overall survival (p < 0.001) when incorporating other relevant tumor and clinical parameters (HR = 3.84, 95% CI: 1.92–7.70). Conclusion The current data suggests that ABCG2 may serve as a prognostic marker for overall survival in patients with clear cell renal cell carcinoma. Further studies with large cohorts of patients will be essential for validating these findings and defining the clinical utility of ABCG2 in the patient population. Electronic supplementary material The online version of this article (doi:10.1186/s12885-017-3224-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Haofei Wang
- Department of Urology, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Building 6th, Floor 6th, 197# Ruijin 2nd road, Shanghai, 200025, China
| | - Fangxiu Luo
- Ruijin North Hospital, Department of Pathology, Shanghai JiaoTong University School of Medicine, Shanghai, 201801, China
| | - Zhe Zhu
- Department of Stem Cell Biology and Regenerative Medicine, Cleveland Clinic, Lerner Research Institute, Cleveland, OH, 44195, USA
| | - Zhaoping Xu
- Department of Urology, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Building 6th, Floor 6th, 197# Ruijin 2nd road, Shanghai, 200025, China
| | - Xin Huang
- Department of Urology, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Building 6th, Floor 6th, 197# Ruijin 2nd road, Shanghai, 200025, China
| | - Renyi Ma
- Ruijin North Hospital, Department of Pathology, Shanghai JiaoTong University School of Medicine, Shanghai, 201801, China
| | - Hongchao He
- Department of Urology, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Building 6th, Floor 6th, 197# Ruijin 2nd road, Shanghai, 200025, China
| | - Yu Zhu
- Department of Urology, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Building 6th, Floor 6th, 197# Ruijin 2nd road, Shanghai, 200025, China
| | - Kun Shao
- Department of Urology, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Building 6th, Floor 6th, 197# Ruijin 2nd road, Shanghai, 200025, China
| | - Juping Zhao
- Department of Urology, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Building 6th, Floor 6th, 197# Ruijin 2nd road, Shanghai, 200025, China.
| |
Collapse
|
29
|
Chaurasiya S, Warner S. Viroimmunotherapy for Colorectal Cancer: Clinical Studies. Biomedicines 2017; 5:E11. [PMID: 28536354 PMCID: PMC5423497 DOI: 10.3390/biomedicines5010011] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 03/02/2017] [Indexed: 12/31/2022] Open
Abstract
Colorectal cancer is a leading cause of cancer incidence and death. Therapies for those with unresectable or recurrent disease are not considered curative at present. More effective and less toxic therapies are desperately needed. Historically, the immune system was thought to be an enemy to oncolytic viral therapy. Thinking that oncolysis would be the only mechanism for cell death, oncolytic virologists theorized that immune clearance was a detriment to oncolysis. Recent advances in our understanding of the tumor microenvironment, and the interplay of tumor survival and a patient's immune system have called into question our understanding of both arenas. It remains unclear what combination of restrictions or enhancements of innate and/or cell-mediated immunity can yield the highest likelihood of viral efficacy. This article reviews the variety of mechanisms explored for viruses such as immunotherapy for colorectal cancer.
Collapse
Affiliation(s)
- Shyambabu Chaurasiya
- Beckman Research Institute, City of Hope National Medical Center, Duarte 91010, CA, USA.
| | - Susanne Warner
- Beckman Research Institute, City of Hope National Medical Center, Duarte 91010, CA, USA.
| |
Collapse
|
30
|
Mazza C, Escudier B, Albiges L. Nivolumab in renal cell carcinoma: latest evidence and clinical potential. Ther Adv Med Oncol 2017; 9:171-181. [PMID: 28344662 PMCID: PMC5349425 DOI: 10.1177/1758834016679942] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Similar to melanoma, renal cell carcinoma (RCC) has been historically considered as an immunogenic tumor, with interleukin 2 (IL-2) and interferon alpha (IFN-α) being the first approved treatments in the 1990s. However, these therapies were effective in only 10-20% of cases and were not well tolerated. Recently, new insights on the interaction between the immune system and tumor have identified the programmed death-1/programmed death-ligand-1 (PD-1/PD-L1) pathway to be a key player in evading host immune responses. The strategy of immune checkpoint blockade is to reduce inhibitory signaling and restore the patient's natural tumor-specific T-cell-mediated immune responses. Nivolumab is the first PD-1 inhibitor to have gained approval for the treatment of patients with metastatic melanoma, squamous and nonsquamous non-small cell lung cancer (NSCLC), Hodgkin disease and recently RCC. In this review, we discuss results from studies of nivolumab in RCC, clinical experience with this agent, and its future development.
Collapse
Affiliation(s)
- Camille Mazza
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Bernard Escudier
- Département de médecine Oncologique, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Laurence Albiges
- Département de médecine Oncologique, Gustave Roussy, Université Paris-Saclay, Villejuif, F-94805, 114 Rue Edouard Vaillant, 94800 Villejuif, France
| |
Collapse
|
31
|
Chinello C, L'imperio V, Stella M, Smith AJ, Bovo G, Grasso A, Grasso M, Raimondo F, Pitto M, Pagni F, Magni F. The proteomic landscape of renal tumors. Expert Rev Proteomics 2016; 13:1103-1120. [PMID: 27748142 DOI: 10.1080/14789450.2016.1248415] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Renal cell carcinoma (RCC) is the most fatal of the common urologic cancers, with approximately 35% of patients dying within 5 years following diagnosis. Therefore, there is a need for non-invasive markers that are capable of detecting and determining the severity of small renal masses at an early stage in order to tailor treatment and follow-up. Proteomic studies have proved to be very useful in the study of tumors. Areas covered: In this review, we will detail the current knowledge obtained by the different proteomic approaches, focusing on MS-based strategies, used to investigate RCC biology in order to identify diagnostic, prognostic and predictive biomarkers on tissue, cultured cells and biological fluids. Expert commentary: Currently, no reliable biomarkers or targets for RCC have been translated into the clinical setting. Moreover, despite the efforts of proteomics and other -omics disciplines, only a small number of them have been observed as shared targets between the different analytical platforms and biological specimens. The difficulty to define a specific molecular pattern for RCC and its subtypes highlights a peculiar profile and a heterogeneity that must be taken into account in future studies.
Collapse
Affiliation(s)
- Clizia Chinello
- a Department of Medicine and Surgery , University Milan Bicocca , Monza , Italy
| | - Vincenzo L'imperio
- a Department of Medicine and Surgery , University Milan Bicocca , Monza , Italy
| | - Martina Stella
- a Department of Medicine and Surgery , University Milan Bicocca , Monza , Italy
| | - Andrew James Smith
- a Department of Medicine and Surgery , University Milan Bicocca , Monza , Italy
| | - Giorgio Bovo
- b Pathology unit , San Gerardo Hospital , Monza , Italy
| | - Angelica Grasso
- c Department of Specialistic Surgical Sciences, Urology unit , Ospedale Maggiore Policlinico Foundation , Milano , Italy
| | - Marco Grasso
- d Department of Urology , San Gerardo Hospital , Monza , Italy
| | - Francesca Raimondo
- a Department of Medicine and Surgery , University Milan Bicocca , Monza , Italy
| | - Marina Pitto
- a Department of Medicine and Surgery , University Milan Bicocca , Monza , Italy
| | - Fabio Pagni
- a Department of Medicine and Surgery , University Milan Bicocca , Monza , Italy
| | - Fulvio Magni
- a Department of Medicine and Surgery , University Milan Bicocca , Monza , Italy
| |
Collapse
|
32
|
Ta L, Xuan C, Xing N, Zhu X. COP1 is downregulated in renal cell carcinoma (RCC) and inhibits the migration of RCC ACHN cells in vitro. Mol Med Rep 2016; 14:1371-8. [DOI: 10.3892/mmr.2016.5373] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 05/09/2016] [Indexed: 11/06/2022] Open
|
33
|
Fu H, Liu Y, Xu L, Chang Y, Zhou L, Zhang W, Yang Y, Xu J. Low Expression of Mucin-4 Predicts Poor Prognosis in Patients With Clear-Cell Renal Cell Carcinoma. Medicine (Baltimore) 2016; 95:e3225. [PMID: 27124015 PMCID: PMC4998678 DOI: 10.1097/md.0000000000003225] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Mucin-4 (MUC4), a member of membrane-bound mucins, has been reported to exert a large variety of distinctive roles in tumorigenesis of different cancers. MUC4 is aberrantly expressed in clear-cell renal cell carcinoma (ccRCC) but its prognostic value is still unveiled. This study aims to assess the clinical significance of MUC4 expression in patients with ccRCC.The expression of MUC4 was assessed by immunohistochemistry in 198 patients with ccRCC who underwent nephrectomy retrospectively in 2003 and 2004. Sixty-seven patients died before the last follow-up in the cohort. Kaplan-Meier method with log-rank test was applied to compare survival curves. Univariate and multivariate Cox regression models were applied to evaluate the prognostic value of MUC4 expression in overall survival (OS). The predictive nomogram was constructed based on the independent prognostic factors. The calibration was built to evaluate the predictive accuracy of nomogram.In patients with ccRCC, MUC4 expression, which was determined to be an independent prognostic indicator for OS (hazard ratio [HR] 3.891; P < 0.001), was negatively associated with tumor size (P = 0.036), Fuhrman grade (P = 0.044), and OS (P < 0.001). The prognostic accuracy of TNM stage, UCLA Integrated Scoring System (UISS), and Mayo clinic stage, size, grade, and necrosis score (SSIGN) prognostic models was improved when MUC4 expression was added. The independent prognostic factors, pT stage, distant metastases, Fuhrman grade, sarcomatoid, and MUC4 expression were integrated to establish a predictive nomogram with high predictive accuracy.MUC4 expression is an independent prognostic factor for OS in patients with ccRCC.
Collapse
Affiliation(s)
- Hangcheng Fu
- From the Department of Biochemistry and Molecular Biology (HF, YL, JX), Department of Immunology, School of Basic Medical Sciences (WZ), Department of Urology, Zhongshan Hospital, Fudan University (YC, LZ, YY), and Department of Urology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China (LX)
| | | | | | | | | | | | | | | |
Collapse
|
34
|
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: 422] [Impact Index Per Article: 46.9] [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
|
35
|
Fu H, Liu Y, Xu L, Liu W, Fu Q, Liu H, Zhang W, Xu J. Galectin-9 predicts postoperative recurrence and survival of patients with clear-cell renal cell carcinoma. Tumour Biol 2015; 36:5791-9. [PMID: 25716202 DOI: 10.1007/s13277-015-3248-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 02/10/2015] [Indexed: 12/30/2022] Open
Abstract
Galectin-9 (Gal-9), a member of animal lectin family with evolutionary conserved carbohydrate recognition domains, has been reported to exert a large variety of functional roles in tumorigenesis due to its β-galactoside-binding affinity. The aim of this study is to evaluate the expression and prognostic significance of Gal-9 in patients with clear-cell renal cell carcinoma (ccRCC). The expression of Gal-9 was assessed by immunohistochemistry in 196 patients with ccRCC who underwent nephrectomy. In the cohort, 48 patients died and 61 patients suffered recurrence. Kaplan-Meier method with log-rank test was applied to compare survival curves. The authors employed univariate and multivariate Cox regression models to evaluate the prognostic value of Gal-9 expression in overall survival (OS) and recurrence-free survival (RFS). In patients with ccRCC, Gal-9 expression, which was positively associated with tumor size (P = 0.014), Fuhrman grade (P = 0.010), and necrosis (P = 0.025), was determined to be an independent prognostic indicator for OS (hazard ratio [HR] 2.394; P = 0.005) and RFS (HR 2.096; P = 0.006). High expression of Gal-9 was associated with poor survival (P = 0.001) and early recurrence (P = 0.006). Furthermore, Gal-9 expression could significantly stratify the patients in early (grades I + II) tumor, node, and metastasis (TNM) stage (OS: P = 0.005; RFS: P = 0.041) and low (grades 1 + 2) Fuhrman grade (OS: P = 0.004; RFS: P = 0.006). The prognostic accuracy of TNM, SSIGN, and UISS prognostic models was improved when Gal-9 expression was added. Gal-9 expression is a potential independent prognostic factor for OS and RFS in patients with ccRCC.
Collapse
Affiliation(s)
- Hangcheng Fu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Chen B, Liu L, Xu H, Yang Y, Zhang L, Zhang F. Effectiveness of immune therapy combined with chemotherapy on the immune function and recurrence rate of cervical cancer. Exp Ther Med 2015; 9:1063-1067. [PMID: 25667679 PMCID: PMC4316956 DOI: 10.3892/etm.2015.2217] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 01/07/2015] [Indexed: 01/26/2023] Open
Abstract
The aim of this study was to compare the immune function of patients with cervical cancer and the cancer recurrence rate in patients treated with biological immune therapy combined with chemotherapy or with chemotherapy only. A total of 79 postoperative patients with cervical cancer participated in the present study. They were randomly divided into a control group and an experimental group. Patients in the control group were treated with cisplatin chemotherapy. Patients in the experimental group were treated with dendritic cell-cytokine-induced killer (DC-CIK) cells combined with cisplatin chemotherapy. The CD3+, CD4+, CD8+, CD16+, CD56+ and CD4+CD25+ cell ratios in peripheral blood, and the expression levels of perforin, granzyme B (GraB) and CD107a of peripheral blood mononuclear cells (PBMCs) in all patients prior to and following treatment were observed. The changes of immune function and recurrence rate between these two groups prior to and following treatment were compared. Prior to treatment, the lymphocyte ratio had no significant difference between the two groups (P>0.05). Following treatment, the lymphocyte ratio in the experimental group was significantly higher than that in the control group (P<0.05). The positive expression levels of perforin, GraB and CD107a of PBMCs in the experimental group following treatment were significantly higher than those prior to treatment and those of the control group (P<0.05). The cumulative recurrence rate in the experimental group was significantly lower than that in the control group (P<0.05). In conclusion, in postoperative patients with cervical cancer, treatment with DC-CIK cells combined with cisplatin chemotherapy significantly improved the immune function, reduced the recurrence rate and prolonged the survival time of the patients.
Collapse
Affiliation(s)
- Bin Chen
- Department of Oncology, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, Jiangsu 215000, P.R. China
| | - Lifen Liu
- Department of Gynecology and Obstetrics, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Haiyan Xu
- Department of Oncology, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, Jiangsu 215000, P.R. China
| | - Yijin Yang
- Department of Oncology, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, Jiangsu 215000, P.R. China
| | - Ling Zhang
- Department of Oncology, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, Jiangsu 215000, P.R. China
| | - Fengchun Zhang
- Department of Oncology, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, Jiangsu 215000, P.R. China
| |
Collapse
|
37
|
Sharma U, Pal D, Prasad R. A novel role of alkaline phosphatase in the ERK1/2 dephosphorylation in renal cell carcinoma cell lines: A new plausible therapeutic target. Biochimie 2014; 107 Pt B:406-9. [PMID: 25241253 DOI: 10.1016/j.biochi.2014.09.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2014] [Accepted: 09/10/2014] [Indexed: 01/11/2023]
|
38
|
Standard b-value versus low b-value diffusion-weighted MRI in renal cell carcinoma: a systematic review and meta-analysis. BMC Cancer 2014; 14:843. [PMID: 25406910 PMCID: PMC4289169 DOI: 10.1186/1471-2407-14-843] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 11/04/2014] [Indexed: 12/22/2022] Open
Abstract
Background We sought to determine the comparative diagnostic performance of standard b-value (800–1000 s/mm2) versus low b-value (400–500 s/mm2) diffusion-weighted magnetic resonance imaging (DW-MRI) in the detection of renal cell carcinoma (RCC). Method After a systematic review of the available literature, studies were included that reported b-values, used a histopathological reference standard, and allowed construction of 2 × 2 contingency tables for detection of RCC lesions using DW-MRI. In addition, a summary receiver operating characteristic (SROC) analysis was performed. Results Four articles that complied with all inclusion and exclusion criteria were selected for data extraction and analysis (n = 248 lesions in 266 patients). All four studies were high quality. Standard b-value DW-MRI displayed a pooled sensitivity of 0.59 (95% confidence interval (CI): 0.51-0.67) and a pooled specificity of 0.50 (95% CI: 0.30-0.70), while low b-value DW-MRI displayed a pooled sensitivity of 0.58 (95% CI: 0.48-0.63) and a pooled specificity of 0.23 (95% CI: 0.09-0.44). The SROC curve of standard b-value DW-MRI displayed an AUC of 0.61 and a Q*index of 0.59, while the SROC curve of low b-value DW-MRI displayed an AUC of 0.68 and a Q*index of 0.64. Conclusion Standard b-value DW-MRI showed a superior specificity but an approximately equivalent sensitivity to low b-value DW-MRI in detecting RCC lesions in the kidney. However, low b-value DW-MRI displayed an overall superior diagnostic accuracy over standard b-value DW-MRI.
Collapse
|
39
|
Gao H, Deng L. Sphingosine kinase-1 activation causes acquired resistance against Sunitinib in renal cell carcinoma cells. Cell Biochem Biophys 2014; 68:419-25. [PMID: 23975598 DOI: 10.1007/s12013-013-9723-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Multi-target tyrosine kinase inhibitor Sunitinib has been widely used in cancer treatment, including metastatic renal cell carcinoma. However, most patients who initially benefit from Sunitinib develop resistance with extended usage of Sunitinib, which is referred to as "acquired resistance". The molecular mechanisms contributing to this acquired resistance remain poorly understood. In this present study, we established Sunitinib-resistant cell lines from human renal cell lines (786-O, A498, ACHN and CAKI1) by continuous treatment with Sunitinib to explore the molecular mechanism leading to Sunitinib resistance. We found that PDGFR-β expression in cell seems to be a protective factor against Sunitinib resistance formation. In addition, we found that both SK1 and ERK were activated in Sunitinib-resistance cell lines and SK1 and ERK inhibitors could resensitize Sunitinib-resistant cell lines. In conclusion, our observations suggest that SK1 and ERK activation is a feature of resistant cell lines, which serves as an alternative pathway evading anti-tumor activity of Sunitinib.
Collapse
Affiliation(s)
- Hui Gao
- Medical College, Qingdao University, Qingdao, 266023, Shandong, China
| | | |
Collapse
|
40
|
Hispidulin Potentiates the Antitumor Effect of Sunitinib Against Human Renal Cell Carcinoma in Laboratory Models. Cell Biochem Biophys 2014; 71:757-64. [DOI: 10.1007/s12013-014-0260-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
41
|
Raimondo F, Corbetta S, Chinello C, Pitto M, Magni F. The urinary proteome and peptidome of renal cell carcinoma patients: a comparison of different techniques. Expert Rev Proteomics 2014; 11:503-14. [PMID: 24890767 DOI: 10.1586/14789450.2014.926222] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Renal cell carcinomas, originating from the renal cortex, account for about 80% of kidney primary malignancies. Small localized tumors rarely produce symptoms and diagnosis is often delayed until the disease is advanced. In contrast to other urological cancers, renal cell carcinomas are associated with a high degree of metastases and a low 5-year survival rate. The identification of diagnostic and prognostic markers, especially in the urine, remains an area of intense investigation. Different proteomic strategies have been applied so far to biomarker discovery in urine at the proteome or the peptidome level. Gel-based and gel-free strategies combined with mass spectrometry are the most-used strategies, have different success rates, and will be depicted here. We also prefigure a scenario in which the limitations of a single approach are overcome by applying new and complementary research strategies, relying on the excellent availability coupled to the intrinsic richness typical of urine samples.
Collapse
Affiliation(s)
- Francesca Raimondo
- Department of Health Sciences, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, Italy
| | | | | | | | | |
Collapse
|
42
|
Yu H, Lin X, Wang F, Zhang B, Wang W, Shi H, Zou B, Zhao J. Proliferation inhibition and the underlying molecular mechanisms of microRNA-30d in renal carcinoma cells. Oncol Lett 2013; 7:799-804. [PMID: 24520297 PMCID: PMC3919943 DOI: 10.3892/ol.2013.1754] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 12/06/2013] [Indexed: 12/26/2022] Open
Abstract
To investigate the inhibitory effects of microRNA-30d (miR-30d) on renal carcinoma cell proliferation and the underlying molecular mechanisms, miR-30d expression in renal cell carcinoma (RCC) specimens was analyzed by quantitative polymerase chain reaction (qPCR). The inhibition of the proliferation of miR-30d on renal carcinoma cells (ACHN cell line) was analyzed by MTT and colony formation assays. The effects of miR-30d on cyclin E2 expression were detected by the luciferase activity of the reporter gene. In addition, the effects of miR-30d on endogenous cyclin E2 expression at the RNA and protein levels were investigated by qPCR and western blot analysis, respectively. Cell cycles were analyzed by flow cytometry. The results showed the following: i) Expression of miR-30d was significantly downregulated in renal carcinoma tissues compared with paraneoplastic tissues; ii) overexpression of miR-30d inhibited renal carcinoma cell proliferation and colony formation; iii) miR-30d inhibited cyclin E2 3' untranslated region-mediated reporter gene expression; and iv) overexpression of miR-30d downregulated endogenous cyclin E2 expression and blocked the cell cycle at the G1 phase. In conclusion, miR-30d functions as a tumor suppressor gene in RCC and inhibits renal carcinoma cell proliferation. Cell cycle regulatory factor cyclin E2 is a target gene of miR-30d. miR-30d inhibits renal carcinoma cell proliferation via the regulation of cyclin E2 expression at the post-transcriptional level.
Collapse
Affiliation(s)
- Hongsheng Yu
- Medical School, Ningbo University, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Ningbo, Zhejiang 315211, P.R. China ; Affiliated Hospital, Ningbo University, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Ningbo, Zhejiang 315211, P.R. China
| | - Xialu Lin
- Medical School, Ningbo University, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Ningbo, Zhejiang 315211, P.R. China
| | - Fang Wang
- Affiliated Hospital, Ningbo University, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Ningbo, Zhejiang 315211, P.R. China
| | - Burong Zhang
- Affiliated Hospital, Ningbo University, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Ningbo, Zhejiang 315211, P.R. China
| | - Weihua Wang
- Affiliated Hospital, Ningbo University, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Ningbo, Zhejiang 315211, P.R. China
| | - Hongbo Shi
- Medical School, Ningbo University, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Ningbo, Zhejiang 315211, P.R. China
| | - Baobo Zou
- Medical School, Ningbo University, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Ningbo, Zhejiang 315211, P.R. China
| | - Jinshun Zhao
- Medical School, Ningbo University, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Ningbo, Zhejiang 315211, P.R. China
| |
Collapse
|
43
|
Dannenmann SR, Hermanns T, Bransi A, Matter C, von Boehmer L, Stevanovic S, Schraml P, Moch H, Knuth A, van den Broek M. Spontaneous peripheral T-cell responses toward the tumor-associated antigen cyclin D1 in patients with clear cell renal cell carcinoma. Cancer Immunol Res 2013; 1:288-95. [PMID: 24777966 DOI: 10.1158/2326-6066.cir-13-0113] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Renal cell carcinoma (RCC) is a heterogeneous group of kidney cancers with clear cell RCC (ccRCC) as the major subgroup. To expand the number of clinically relevant tumor-associated antigens (TAA) that can be targeted by immunotherapy, we analyzed samples from 23 patients with primary ccRCC for the expression and immunogenicity of various TAAs. We found high-frequency expression of MAGE-A9 and NY-ESO-1 in 36% and 55% of samples, respectively, and overexpression of PRAME, RAGE-1, CA-IX, Cyclin D1, ADFP, C-MET, and RGS-5 in many of the tumor samples. We analyzed the blood of patients with HLA-A2(+) ccRCC for the presence of CD8(+) T cells specific for TAA-derived HLA-A2-restricted peptides and found spontaneous responses to cyclin D1 in 5 of 6 patients with Cyclin D1-positive tumors. Cyclin D1-specific CD8(+) T cells secreted TNF-α, IFN-γ, and interleukin-2 (IL-2), and degranulated, indicating the presence of polyfunctional tumor-specific CD8(+) T cells in the blood of these patients with ccRCC. The high frequency (43%) of Cyclin D1 overexpression and the presence of functional cyclin D1-specific T cells in 83% of these patients with ccRCC suggest that cyclin D1 may be a target for immunotherapeutic strategies.
Collapse
Affiliation(s)
- Stefanie R Dannenmann
- Authors' Affiliations: Departments of Department of Immunology, Interfaculty Institute for Cell Biology, University of Tubingen, Tubingen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Graves A, Hessamodini H, Wong G, Lim WH. Metastatic renal cell carcinoma: update on epidemiology, genetics, and therapeutic modalities. Immunotargets Ther 2013; 2:73-90. [PMID: 27471690 PMCID: PMC4928369 DOI: 10.2147/itt.s31426] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The treatment of advanced renal cell carcinoma (RCC) remains a major therapeutic challenge for clinicians. Despite advances in the understanding of the immunobiology of RCC and the availability of several novel targeted agents, there has been little improvement in the survival of patients with metastatic RCC. This review will focus on the recent understanding of risk factors and treatment options and outcomes of metastatic RCC, in particular, targeted therapeutic agents that inhibit vascular endothelial growth factor and mammalian target of rapamycin pathways. Prospective studies are required to determine whether sequential targeted therapy will further improve progression-free survival in RCC. Ongoing research to develop novel agents with better tolerability and enhanced efficacy in the treatment of metastatic RCC is required.
Collapse
Affiliation(s)
- Angela Graves
- Department of Renal Medicine, Sir Charles Gairdner Hospital, Perth, WA, Australia
| | - Hannah Hessamodini
- Department of Renal Medicine, Sir Charles Gairdner Hospital, Perth, WA, Australia
| | - Germaine Wong
- Centre for Kidney Research, University of Sydney, Sydney, NSW, Australia
| | - Wai H Lim
- Department of Renal Medicine, Sir Charles Gairdner Hospital, Perth, WA, Australia; School of Medicine and Pharmacology, University of Western Australia, Perth, WA, Australia
| |
Collapse
|
45
|
Van Brussel I, Van Craenenbroeck AH, Schrijvers DM, Cools N. Does patient-tailored immunotherapy pave the way for new renal cell carcinoma treatment perspectives? Transl Androl Urol 2013; 2:85-8. [PMID: 26816729 PMCID: PMC4708222 DOI: 10.3978/j.issn.2223-4683.2012.10.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Affiliation(s)
- Ilse Van Brussel
- 1 Laboratory of Pharmacology, University of Antwerp, B-2610 Antwerp, Belgium ; 2 Division of Nephrology-Hypertension, Antwerp University Hospital, B-2650 Edegem, Antwerp, Belgium ; 3 Laboratory of Physiopharmacology, University of Antwerp, B-2610 Antwerp, Belgium ; 4 Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, B-2610 Antwerp, Belgium
| | - Amaryllis H Van Craenenbroeck
- 1 Laboratory of Pharmacology, University of Antwerp, B-2610 Antwerp, Belgium ; 2 Division of Nephrology-Hypertension, Antwerp University Hospital, B-2650 Edegem, Antwerp, Belgium ; 3 Laboratory of Physiopharmacology, University of Antwerp, B-2610 Antwerp, Belgium ; 4 Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, B-2610 Antwerp, Belgium
| | - Dorien M Schrijvers
- 1 Laboratory of Pharmacology, University of Antwerp, B-2610 Antwerp, Belgium ; 2 Division of Nephrology-Hypertension, Antwerp University Hospital, B-2650 Edegem, Antwerp, Belgium ; 3 Laboratory of Physiopharmacology, University of Antwerp, B-2610 Antwerp, Belgium ; 4 Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, B-2610 Antwerp, Belgium
| | - Nathalie Cools
- 1 Laboratory of Pharmacology, University of Antwerp, B-2610 Antwerp, Belgium ; 2 Division of Nephrology-Hypertension, Antwerp University Hospital, B-2650 Edegem, Antwerp, Belgium ; 3 Laboratory of Physiopharmacology, University of Antwerp, B-2610 Antwerp, Belgium ; 4 Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, B-2610 Antwerp, Belgium
| |
Collapse
|
46
|
Tang X, Liu T, Zang X, Liu H, Wang D, Chen H, Zhang B. Adoptive cellular immunotherapy in metastatic renal cell carcinoma: a systematic review and meta-analysis. PLoS One 2013; 8:e62847. [PMID: 23667530 PMCID: PMC3647060 DOI: 10.1371/journal.pone.0062847] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 03/26/2013] [Indexed: 12/31/2022] Open
Abstract
Purpose Metastatic renal cell carcinoma (mRCC), as one of the most immunogenic tumors has been the focus of adoptive cellular immunotherapy (ACI), but the effects of ACI on objective response and survival in patients with mRCC are still controversial. Therefore, a systematic review and meta-analysis was performed to address this issue. Methods A search was conducted in the PubMed database for randomized clinical trials (RCTs) with ACI in mRCC. All included articles in this study were assessed according to the selection criteria and were divided into two groups: ACI versus no ACI. Outcomes were toxicity, objective response, 1-, 3- and 5-year survival. Risk ratio (RR) and 95% confidence intervals (CI) were calculated using a fixed-effects meta-analysis. Heterogeneity was measured by value of I2 or P. Results 4 studies (469 patients) were included. Most of ACI-related adverse reactions were grade 1 or 2 and reversible. ACI provided significant benefit in terms of objective response (RR = 1.65; 95% CI, 1.15 to 2.38; P = 0.007, I2 = 49%), 1-year survival (RR = 1.30; 95% CI, 1.12 to 1.52; P = 0.0008, I2 = 0%), 3-year survival (RR = 2.76; 95% CI, 1.85 to 4.14; P<0.00001, I2 = 46%) and 5-year survival (RR = 2.42; 95% CI, 1.21 to 4.83; P = 0.01, I2 = 28%). Conclusions ACI may be a safe and effective treatment for improving objective response, 1-, 3- and 5-year survival in patients with mRCC. Besides, five obstacles for ACI, including high degree of personalization, unsuitable WHO/RECIST response criteria, inadequate identification of tumor-associated antigens (TAAs), lack of effective combination treatments and less attention paid to the quality of ACI products, should be overcome during the successful development of more potent ACI for cancer in the future.
Collapse
Affiliation(s)
- Xiaoyi Tang
- Department of Hematopoietic Stem Cell Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
- Cell and Gene Therapy Center, Academy of Military Medical Sciences, Beijing, China
| | - Ting Liu
- Department of Hematopoietic Stem Cell Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
- Cell and Gene Therapy Center, Academy of Military Medical Sciences, Beijing, China
| | - Xuefeng Zang
- Department of Hematopoietic Stem Cell Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
- Cell and Gene Therapy Center, Academy of Military Medical Sciences, Beijing, China
- Military Postgraduate Medical College, General Hospital of Chinese People’s Liberation Army, Beijing, China
| | - Hao Liu
- Department of Hematopoietic Stem Cell Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
- Cell and Gene Therapy Center, Academy of Military Medical Sciences, Beijing, China
| | - Danhong Wang
- Department of Hematopoietic Stem Cell Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
- Cell and Gene Therapy Center, Academy of Military Medical Sciences, Beijing, China
| | - Hu Chen
- Department of Hematopoietic Stem Cell Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
- Cell and Gene Therapy Center, Academy of Military Medical Sciences, Beijing, China
- * E-mail: (HC); (BZ)
| | - Bin Zhang
- Department of Hematopoietic Stem Cell Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
- Cell and Gene Therapy Center, Academy of Military Medical Sciences, Beijing, China
- * E-mail: (HC); (BZ)
| |
Collapse
|
47
|
Jawanda GG, Drachenberg D. Spontaneous regression of biopsy proven primary renal cell carcinoma: A case study. Can Urol Assoc J 2012; 6:E203-5. [PMID: 23093646 PMCID: PMC3478393 DOI: 10.5489/cuaj.11035] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Spontaneous regression of renal cell carcinoma (RCC) is a well-recognized and interesting phenomenon that is poorly understood and rarely documented. There are very few reported cases of spontaneously regressed primary RCC. We present a 63-year-old male with a biopsy-proven RCC that regressed with complete resolution of symptoms.
Collapse
|
48
|
Abstract
The inherent immunogenicity of melanoma and renal cell carcinoma (RCC) has made these tumors a focus of considerable research in vaccine development. Recent data from murine studies of immunosurveillance have highlighted the importance of both innate and adaptive immune responses in shaping a tumor's inherent susceptibility to immune surveillance and immunotherapy. Melanoma has been a useful model for the identification of tumor-associated antigens and a number of putative renal cell antigens have been described more recently. These antigens have been targeted using a variety of vaccine strategies, including protein- and peptide-based vaccines, recombinant antigen-expressing vectors, and whole cell vaccine approaches. While evidence for clinical benefit has been disappointing to date, several current phase III clinical trials are in progress based on promising results from phase II studies. Accumulating data suggest that the tumor microenvironment and mechanisms of immunological escape by established tumors are significant barriers that must be overcome before vaccine therapy can be fully realized. This review will discuss the basis for vaccine development, describe some of the more promising vaccine strategies in development, and mention some of the tumor escape mechanisms that block effective anti-tumor immunity for melanoma and RCC.
Collapse
|
49
|
IFNα2b augments immune responses of cisplatin+5-fluorouracil treated tongue squamous cell carcinoma patients--a preliminary study. Indian J Med Res 2012; 136:54-9. [PMID: 22885264 PMCID: PMC3461718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND & OBJECTIVES Interferon alpha 2b (IFNα2b) has been reported to regulate several immune functions efficiently to enhance the cytotoxic activity of NK and T cells towards various forms of tumours. The objective of the present study was to evaluate the efficacy of IFNα2b in overcoming disease induced and/or treatment associated imunosuppression of tongue squamous cell carcinoma (TSCC) patients undergoing chemotherapy for better clinical outcome. METHODS Seven TSCC patients under cisplatin + 5-fluorouracil chemotherapy in combination with IFNα2b were assessed for various immunohaematological parameters before treatment, after chemotherapy and after IFNα2b therapy. RESULTS Deterioration of the haematological and immune responses was detected in immunosuppressed TSCC patients after chemotherapy. IFNα2b treatment led to a recovery in these parameters in most of the patients. Greater number of T/NK cells and enhanced secretion of type 1 cytokines were also noted. Haematological complications were reduced after completion of the therapy. Immune- and haematostimulation were also observed in patients with partial response. No positive clinical response was detected in one patient. INTERPRETATION & CONCLUSIONS IFNα2b appears to be an effective immunostimulator having clinical impact to combat the immunosuppression in TSCC patients. Successful immunostimulation by IFNα2b may help TSCC patients in clinical improvement. The findings of this preliminary study need to be confirmed on a large number of patients with TSCC.
Collapse
|
50
|
Abstract
Although several cytokines have shown antitumor activity in renal cell carcinoma (RCC), the most consistent results have been reported with interleukin-2 (IL-2) and interferon (IFN). Recent insights into how the immune response to a tumor is regulated hold the promise of allowing patients to obtain a durable response to immunotherapy, perhaps without the significant toxicity associated with conventional approaches. This review describes how improvements in patient selection, combination therapy, and investigational agents might expand and better define the role of immunotherapy in metastatic RCC.
Collapse
|