1
|
Kim GR, Kang JH, Kim HJ, Im E, Bae J, Kwon WS, Rha SY, Chung HC, Cho EY, Kim SY, Kim YC. Discovery of novel 1H-benzo[d]imidazole-4,7-dione based transglutaminase 2 inhibitors as p53 stabilizing anticancer agents in renal cell carcinoma. Bioorg Chem 2024; 143:107061. [PMID: 38154386 DOI: 10.1016/j.bioorg.2023.107061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/21/2023] [Accepted: 12/23/2023] [Indexed: 12/30/2023]
Abstract
Overexpression of transglutaminase 2 (TGase 2; TG2) has been implicated in the progression of renal cell carcinoma (RCC) through the inactivation of p53 by forming a protein complex. Because most p53 in RCC has no mutations, apoptosis can be increased by inhibiting the binding between TG2 and p53 to increase the stability of p53. In the present study, a novel TG2 inhibitor was discovered by investigating the structure of 1H-benzo[d]imidazole-4,7-dione as a simpler chemotype based on the amino-1,4-benzoquinone moiety of streptonigrin, a previously reported inhibitor. Through structure-activity relationship (SAR) studies, compound 8j (MD102) was discovered as a potent TG2 inhibitor with an IC50 value of 0.35 µM, p53 stabilization effect and anticancer effects in the ACHN and Caki-1 RCC cell lines with sulforhodamine B (SRB) GI50 values of 2.15 µM and 1.98 µM, respectively. The binding property of compound 8j (MD102) with TG2 was confirmed to be reversible in a competitive enzyme assay, and the binding interaction was expected to be formed at the β-sandwich domain, a p53 binding site, in the SPR binding assay with mutant proteins. The mode of binding of compound 8j (MD102) to the β-sandwich domain of TG2 was analyzed by molecular docking using the crystal structure of the active conformation of human TG2. Compound 8j (MD102) induced a decrease in the downstream signaling of p-AKT and p-mTOR through the stabilization of p53 by TG2 inhibition, resulting in tumor cell apoptosis. In a xenograft animal model using ACHN cancer cells, oral administration and intraperitoneal injection of compound 8j (MD102) showed an inhibitory effect on tumor growth, confirming increased levels of p53 and decreased levels of Ki-67 in tumor tissues through immunohistochemical (IHC) tissue staining. These results indicated that the inhibition of TG2 by compound 8j (MD102) could enhance p53 stabilization, thereby ultimately showing anticancer effects in RCC. Compound 8j (MD102), a novel TG2 inhibitor, can be further applied for the development of an anticancer candidate drug targeting RCC.
Collapse
Affiliation(s)
- Ga-Ram Kim
- School of Life Sciences, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Joon Hee Kang
- Graduate School of Cancer Science and Policy, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang 10408, Republic of Korea
| | - Hyeon Joo Kim
- MDbiopharm Corp., 114 Beobwon-ro, Songpa-gu, Seoul 05854, Republic of Korea
| | - Eunji Im
- MDbiopharm Corp., 114 Beobwon-ro, Songpa-gu, Seoul 05854, Republic of Korea
| | - Jinsu Bae
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Woo Sun Kwon
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Sun Young Rha
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea; Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Hyun Cheol Chung
- MDbiopharm Corp., 114 Beobwon-ro, Songpa-gu, Seoul 05854, Republic of Korea; Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Eun Yi Cho
- MDbiopharm Corp., 114 Beobwon-ro, Songpa-gu, Seoul 05854, Republic of Korea.
| | - Soo-Youl Kim
- Cancer Molecular Biology Branch, Division of Cancer Biology, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang 10408, Republic of Korea.
| | - Yong-Chul Kim
- School of Life Sciences, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea.
| |
Collapse
|
2
|
Lin L, Wu Q, Lu F, Lei J, Zhou Y, Liu Y, Zhu N, Yu Y, Ning Z, She T, Hu M. Nrf2 signaling pathway: current status and potential therapeutic targetable role in human cancers. Front Oncol 2023; 13:1184079. [PMID: 37810967 PMCID: PMC10559910 DOI: 10.3389/fonc.2023.1184079] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 08/18/2023] [Indexed: 10/10/2023] Open
Abstract
Cancer is a borderless global health challenge that continues to threaten human health. Studies have found that oxidative stress (OS) is often associated with the etiology of many diseases, especially the aging process and cancer. Involved in the OS reaction as a key transcription factor, Nrf2 is a pivotal regulator of cellular redox state and detoxification. Nrf2 can prevent oxidative damage by regulating gene expression with antioxidant response elements (ARE) to promote the antioxidant response process. OS is generated with an imbalance in the redox state and promotes the accumulation of mutations and genome instability, thus associated with the establishment and development of different cancers. Nrf2 activation regulates a plethora of processes inducing cellular proliferation, differentiation and death, and is strongly associated with OS-mediated cancer. What's more, Nrf2 activation is also involved in anti-inflammatory effects and metabolic disorders, neurodegenerative diseases, and multidrug resistance. Nrf2 is highly expressed in multiple human body parts of digestive system, respiratory system, reproductive system and nervous system. In oncology research, Nrf2 has emerged as a promising therapeutic target. Therefore, certain natural compounds and drugs can exert anti-cancer effects through the Nrf2 signaling pathway, and blocking the Nrf2 signaling pathway can reduce some types of tumor recurrence rates and increase sensitivity to chemotherapy. However, Nrf2's dual role and controversial impact in cancer are inevitable consideration factors when treating Nrf2 as a therapeutic target. In this review, we summarized the current state of biological characteristics of Nrf2 and its dual role and development mechanism in different tumor cells, discussed Keap1/Nrf2/ARE signaling pathway and its downstream genes, elaborated the expression of related signaling pathways such as AMPK/mTOR and NF-κB. Besides, the main mechanism of Nrf2 as a cancer therapeutic target and the therapeutic strategies using Nrf2 inhibitors or activators, as well as the possible positive and negative effects of Nrf2 activation were also reviewed. It can be concluded that Nrf2 is related to OS and serves as an important factor in cancer formation and development, thus provides a basis for targeted therapy in human cancers.
Collapse
Affiliation(s)
- Li Lin
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Qing Wu
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Feifei Lu
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Jiaming Lei
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Yanhong Zhou
- Department of Medical School of Facial Features, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Yifei Liu
- School of Biomedical Engineering, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Ni Zhu
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - You Yu
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Zhifeng Ning
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Tonghui She
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Meichun Hu
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| |
Collapse
|
3
|
Zhang HB, Hu Y, Deng JL, Fang GY, Zeng Y. Insights into the involvement of long non-coding RNAs in doxorubicin resistance of cancer. Front Pharmacol 2023; 14:1243934. [PMID: 37781691 PMCID: PMC10540237 DOI: 10.3389/fphar.2023.1243934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/31/2023] [Indexed: 10/03/2023] Open
Abstract
Doxorubicin is one of the most classical chemotherapeutic drugs for the treatment of cancer. However, resistance to the cytotoxic effects of doxorubicin in tumor cells remains a major obstacle. Aberrant expression of long non-coding RNAs (lncRNAs) has been associated with tumorigenesis and development via regulation of chromatin remodeling, transcription, and post-transcriptional processing. Emerging studies have also revealed that dysregulation of lncRNAs mediates the development of drug resistance through multiple molecules and pathways. In this review, we focus on the role and mechanism of lncRNAs in the progress of doxorubicin resistance in various cancers, which mainly include cellular drug transport, cell cycle disorder, anti-apoptosis, epithelial-mesenchymal transition, cancer stem cells, autophagy, tumor microenvironment, metabolic reprogramming and signaling pathways. This review is aimed to provide potential therapeutic targets for future cancer therapy, especially for the reversal of chemoresistance.
Collapse
Affiliation(s)
- Hai-Bo Zhang
- Department of Pharmacy, Hangzhou Women’s Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, China
| | - Yang Hu
- Guangzhou Institute of Respiratory Disease and China State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jun-Li Deng
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Guo-Ying Fang
- Department of Pharmacy, Hangzhou Women’s Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, China
| | - Ying Zeng
- Department of Pharmacy, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| |
Collapse
|
4
|
Identifying somatic changes in drug transporters using whole genome and transcriptome sequencing data of advanced tumors. Biomed Pharmacother 2023; 159:114210. [PMID: 36621142 DOI: 10.1016/j.biopha.2022.114210] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/23/2022] [Accepted: 12/30/2022] [Indexed: 01/09/2023] Open
Abstract
Drug resistance is a perpetual problem in cancer therapy with many underlying mechanisms. Alterations in drug transport over the cancer cell membrane can severely alter intratumoral drug exposure, contributing to resistance. Here, we present the somatic mutational landscape of 48 ATP-binding cassette and 416 solute carrier transporter genes in a cohort (CPCT-02; NCT01855477) of 3290 patients with different types of advanced and metastasized cancer through analysis of whole genome and transcriptome sequencing. In order to identify potential stressor mechanisms, we stratified patients based on previous systemic therapies and subsequently investigated the enrichment of mutations and copy-number alterations of transporter genes. In tumors from patients pretreated with protein kinase inhibitors (PKIs), genes encoding for specific copper (SLC31A1 and SLC31A2, χ2-test adjusted p-values: 6.9e-09 and 2.5e-09) and nucleoside transporters (SLC28A2 and SLC28A3, χ2-test adjusted p-values: 3.5e-06 and 6.8e-07) were deleted significantly more frequently than in patients pretreated with chemotherapy. Moreover, we detected 16 transporters that were differentially expressed at RNA level between these treatment groups. These findings contradict mechanisms of selective pressure, as they would be expected to originate during treatment with chemotherapy rather than with PKIs. Hence, they might constitute primary drug resistance mechanisms and, therefore, warrant further study.
Collapse
|
5
|
Seidel P, Rubarth A, Zodel K, Peighambari A, Neumann F, Federkiel Y, Huang H, Hoefflin R, Adlesic M, Witt C, Hoffmann DJ, Metzger P, Lindemann RK, Zenke FT, Schell C, Boerries M, von Elverfeldt D, Reichardt W, Follo M, Albers J, Frew IJ. ATR represents a therapeutic vulnerability in clear cell renal cell carcinoma. JCI Insight 2022; 7:156087. [PMID: 36413415 PMCID: PMC9869969 DOI: 10.1172/jci.insight.156087] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 11/10/2022] [Indexed: 11/24/2022] Open
Abstract
Metastatic clear cell renal cell carcinomas (ccRCCs) are resistant to DNA-damaging chemotherapies, limiting therapeutic options for patients whose tumors are resistant to tyrosine kinase inhibitors and/or immune checkpoint therapies. Here we show that mouse and human ccRCCs were frequently characterized by high levels of endogenous DNA damage and that cultured ccRCC cells exhibited intact cellular responses to chemotherapy-induced DNA damage. We identify that pharmacological inhibition of the DNA damage-sensing kinase ataxia telangiectasia and Rad3-related protein (ATR) with the orally administered, potent, and selective drug M4344 (gartisertib) induced antiproliferative effects in ccRCC cells. This effect was due to replication stress and accumulation of DNA damage in S phase. In some cells, DNA damage persisted into subsequent G2/M and G1 phases, leading to the frequent accumulation of micronuclei. Daily single-agent treatment with M4344 inhibited the growth of ccRCC xenograft tumors. M4344 synergized with chemotherapeutic drugs including cisplatin and carboplatin and the poly(ADP-ribose) polymerase inhibitor olaparib in mouse and human ccRCC cells. Weekly M4344 plus cisplatin treatment showed therapeutic synergy in ccRCC xenografts and was efficacious in an autochthonous mouse ccRCC model. These studies identify ATR inhibition as a potential novel therapeutic option for ccRCC.
Collapse
Affiliation(s)
- Philipp Seidel
- Department of Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany
| | - Anne Rubarth
- Department of Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany
| | - Kyra Zodel
- Department of Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany
| | - Asin Peighambari
- Department of Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany
| | - Felix Neumann
- Translational Innovation Platform Oncology and Immuno-Oncology, the Healthcare Business of Merck KGaA, Darmstadt, Germany
| | - Yannick Federkiel
- Department of Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany
| | - Hsin Huang
- Department of Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany
| | - Rouven Hoefflin
- Department of Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany
| | - Mojca Adlesic
- Department of Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany
| | - Christian Witt
- Department of Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany
| | - David J. Hoffmann
- Department of Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany
| | | | - Ralph K. Lindemann
- Translational Innovation Platform Oncology and Immuno-Oncology, the Healthcare Business of Merck KGaA, Darmstadt, Germany
| | - Frank T. Zenke
- Translational Innovation Platform Oncology and Immuno-Oncology, the Healthcare Business of Merck KGaA, Darmstadt, Germany
| | - Christoph Schell
- Institute for Surgical Pathology, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) Partner Site Freiburg, Freiburg, Germany.,Comprehensive Cancer Center Freiburg (CCCF) and
| | | | - Wilfried Reichardt
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) Partner Site Freiburg, Freiburg, Germany.,Medical Physics, Department of Radiology, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany
| | - Marie Follo
- Department of Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany
| | - Joachim Albers
- Translational Innovation Platform Oncology and Immuno-Oncology, the Healthcare Business of Merck KGaA, Darmstadt, Germany
| | - Ian J. Frew
- Department of Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) Partner Site Freiburg, Freiburg, Germany.,Comprehensive Cancer Center Freiburg (CCCF) and,Medical Physics, Department of Radiology, Faculty of Medicine, Medical Center – University of Freiburg, Freiburg, Germany
| |
Collapse
|
6
|
Neo SP, Alli-Shaik A, Wee S, Lim Z, Gunaratne J. Englerin A Rewires Phosphosignaling via Hsp27 Hyperphosphorylation to Induce Cytotoxicity in Renal Cancer Cells. J Proteome Res 2022; 21:1948-1960. [PMID: 35838755 DOI: 10.1021/acs.jproteome.2c00248] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Englerin A (EA) is a small-molecule natural product with selective cytotoxicity against renal cancer cells. EA has been shown to induce apoptosis and cell death through cell-cycle arrest and/or insulin signaling pathways. However, its biological mode of action or targets in renal cancer remains enigmatic. In this study, we employed advanced mass spectrometry-based phosphoproteomics approaches to identify EA's functional roles in renal cancer. We identified 10,940 phosphorylation sites, of which 706 sites exhibited EA-dependent phosphorylation changes. Integrated analysis of motifs and interaction networks suggested activation of stress-activated kinases including p38 upon EA treatment. Of note, a downstream target of p38, Hsp27, was found to be hyperphosphorylated on multiple sites upon EA treatment. Among these, a novel site Ser65 on Hsp27, which was further validated by targeted proteomics, was shown to be crucial for EA-induced cytotoxicity in renal cancer cells. Taken together, these data reveal the complex signaling cascade that is induced upon EA treatment and importantly provide insights into its effects on downstream molecular signaling.
Collapse
Affiliation(s)
- Suat Peng Neo
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Asfa Alli-Shaik
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Sheena Wee
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Zijie Lim
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Jayantha Gunaratne
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, 61 Biopolis Drive, Singapore 138673, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore 117597, Singapore
| |
Collapse
|
7
|
Simon AG, Esser LK, Ellinger J, Ritter M, Kristiansen G, Muders MH, Mayr T, Toma MI. RNA Sequencing Reveals Alterations and Similarities in Cell Metabolism, Hypoxia and Immune Evasion in Primary Cell Cultures of Clear Cell Renal Cell Carcinoma. Front Oncol 2022; 12:883195. [PMID: 35646693 PMCID: PMC9130782 DOI: 10.3389/fonc.2022.883195] [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: 02/24/2022] [Accepted: 04/08/2022] [Indexed: 11/17/2022] Open
Abstract
The treatment of advanced renal cell carcinoma remains a challenge. To develop novel therapeutic approaches, primary cell cultures as an in vitro model are considered more representative than commercial cell lines. In this study, we analyzed the gene expression of previously established primary cell cultures of clear cell renal cell carcinoma by bulk (3’m)RNA sequencing and compared it to the tissue of origin. The objectives were the identification of dysregulated pathways under cell culture conditions. Furthermore, we assessed the suitability of primary cell cultures for studying crucial biological pathways, including hypoxia, growth receptor signaling and immune evasion. RNA sequencing of primary cell cultures of renal cell carcinoma and a following Enrichr database analysis revealed multiple dysregulated pathways under cell culture conditions. 444 genes were significantly upregulated and 888 genes downregulated compared to the tissue of origin. The upregulated genes are crucial in DNA repair, cell cycle, hypoxia and metabolic shift towards aerobic glycolysis. A downregulation was observed for genes involved in pathways of immune cell differentiation and cell adhesion. We furthermore observed that 7275 genes have a similar mRNA expression in cell cultures and in tumor tissue, including genes involved in the immune checkpoint signaling or in pathways responsible for tyrosine kinase receptor resistance. Our findings confirm that primary cell cultures are a representative tool for specified experimental approaches. The results presented in this study give further valuable insights into the complex adaptation of patient-derived cells to a new microenvironment, hypoxia and other cell culture conditions, which are often neglected in daily research, and allow new translational and therapeutic approaches.
Collapse
Affiliation(s)
- Adrian Georg Simon
- Department of Pathology, University Hospital Bonn, Bonn, Germany.,Department of Pathology, University Hospital Cologne, Cologne, Germany
| | | | - Jörg Ellinger
- Department of Urology, University Hospital Bonn, Bonn, Germany
| | - Manuel Ritter
- Department of Urology, University Hospital Bonn, Bonn, Germany
| | - Glen Kristiansen
- Department of Pathology, University Hospital Bonn, Bonn, Germany
| | - Michael H Muders
- Department of Pathology, University Hospital Bonn, Bonn, Germany
| | - Thomas Mayr
- Department of Pathology, University Hospital Bonn, Bonn, Germany
| | | |
Collapse
|
8
|
Complementary roles of surgery and systemic treatment in clear cell renal cell carcinoma. Nat Rev Urol 2022; 19:391-418. [PMID: 35546184 DOI: 10.1038/s41585-022-00592-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2022] [Indexed: 12/12/2022]
Abstract
Standard-of-care management of renal cell carcinoma (RCC) indisputably relies on surgery for low-risk localized tumours and systemic treatment for poor-prognosis metastatic disease, but a grey area remains, encompassing high-risk localized tumours and patients with metastatic disease with a good-to-intermediate prognosis. Over the past few years, results of major practice-changing trials for the management of metastatic RCC have completely transformed the therapeutic options for this disease. Treatments targeting vascular endothelial growth factor (VEGF) have been the mainstay of therapy for metastatic RCC in the past decade, but the advent of immune checkpoint inhibitors has revolutionized the therapeutic landscape in the metastatic setting. Results from several pivotal trials have shown a substantial benefit from the combination of VEGF-directed therapy and immune checkpoint inhibition, raising new hopes for the treatment of high-risk localized RCC. The potential of these therapeutics to facilitate the surgical extirpation of the tumour in the neoadjuvant setting or to improve disease-free survival in the adjuvant setting has been investigated. The role of surgery for metastatic RCC has been redefined, with results of large trials bringing into question the paradigm of upfront cytoreductive nephrectomy, inherited from the era of cytokine therapy, when initial extirpation of the primary tumour did show clinical benefits. The potential benefits and risks of deferred surgery for residual primary tumours or metastases after partial response to checkpoint inhibitor treatment are also gaining interest, considering the long-lasting effects of these new drugs, which encourages the complete removal of residual masses.
Collapse
|
9
|
Amaro F, Carvalho M, Bastos MDL, Guedes de Pinho P, Pinto J. Pharmacometabolomics Applied to Personalized Medicine in Urological Cancers. Pharmaceuticals (Basel) 2022; 15:295. [PMID: 35337093 PMCID: PMC8952371 DOI: 10.3390/ph15030295] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 02/06/2023] Open
Abstract
Prostate cancer (PCa), bladder cancer (BCa), and renal cell carcinoma (RCC) are the most common urological cancers, and their incidence has been rising over time. Surgery is the standard treatment for these cancers, but this procedure is only effective when the disease is localized. For metastatic disease, PCa is typically treated with androgen deprivation therapy, while BCa is treated with chemotherapy, and RCC is managed primarily with targeted therapies. However, response rates to these therapeutic options remain unsatisfactory due to the development of resistance and treatment-related toxicity. Thus, the discovery of biomarkers with prognostic and predictive value is needed to stratify patients into different risk groups, minimizing overtreatment and the risk of drug resistance development. Pharmacometabolomics, a branch of metabolomics, is an attractive tool to predict drug response in an individual based on its own metabolic signature, which can be collected before, during, and after drug exposure. Hence, this review focuses on the application of pharmacometabolomic approaches to identify the metabolic responses to hormone therapy, targeted therapy, immunotherapy, and chemotherapy for the most prevalent urological cancers.
Collapse
Affiliation(s)
- Filipa Amaro
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Department of Biological Sciences, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (M.C.); (M.d.L.B.); (P.G.d.P.)
- UCIBIO/REQUIMTE, Department of Biological Sciences, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Márcia Carvalho
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Department of Biological Sciences, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (M.C.); (M.d.L.B.); (P.G.d.P.)
- UCIBIO/REQUIMTE, Department of Biological Sciences, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- FP-I3ID, FP-ENAS, CEBIMED, University Fernando Pessoa, 4200-150 Porto, Portugal
- Faculty of Health Sciences, University Fernando Pessoa, 4200-150 Porto, Portugal
| | - Maria de Lourdes Bastos
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Department of Biological Sciences, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (M.C.); (M.d.L.B.); (P.G.d.P.)
- UCIBIO/REQUIMTE, Department of Biological Sciences, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Paula Guedes de Pinho
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Department of Biological Sciences, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (M.C.); (M.d.L.B.); (P.G.d.P.)
- UCIBIO/REQUIMTE, Department of Biological Sciences, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Joana Pinto
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Department of Biological Sciences, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (M.C.); (M.d.L.B.); (P.G.d.P.)
- UCIBIO/REQUIMTE, Department of Biological Sciences, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
10
|
Buti S, Bersanelli M, Mazzaschi G, Cattrini C, Brunelli M, Di Maio M. Can we identify a preferred first-line strategy for sarcomatoid renal cell carcinoma? A network meta-analysis. Immunotherapy 2022; 14:145-153. [PMID: 34806404 DOI: 10.2217/imt-2021-0157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 11/04/2021] [Indexed: 11/21/2022] Open
Abstract
Background: Combinations based on immune checkpoint inhibitors are the new first-line standard treatment for metastatic renal cell carcinoma. Sarcomatoid renal cell carcinoma (sRCC) has a dismal prognosis but good immunogenicity. Methods: The authors performed a network meta-analysis of Phase III randomized trials of immune checkpoint inhibitor-based combinations versus standard tyrosine kinase inhibitor monotherapy reporting data for sRCC. The end points were overall survival, progression-free survival and objective response rate. Results: Five trials comprising 569 sRCC patients (out of a total of 4409 metastatic renal cell carcinoma patients) were included. Nivolumab-cabozantinib was the highest ranking treatment for overall survival (p-score = 88%) and progression-free survival (p-score = 81%). Atezolizumab-bevacizumab had the highest rank for objective response rate (p-score = 80%). Conclusion: Despite some limitations, nivolumab-cabozantinib might be the preferred first-line option for sRCC in terms of efficacy.
Collapse
Affiliation(s)
- Sebastiano Buti
- Medical Oncology Unit, University Hospital of Parma, Parma, 43126, Italy
| | - Melissa Bersanelli
- Medical Oncology Unit, University Hospital of Parma, Parma, 43126, Italy
- Department of Medicine and Surgery, University of Parma, Parma, 43126, Italy
| | - Giulia Mazzaschi
- Medical Oncology Unit, University Hospital of Parma, Parma, 43126, Italy
- Department of Medicine and Surgery, University of Parma, Parma, 43126, Italy
| | - Carlo Cattrini
- Division of Oncology, University Hospital 'Maggiore della Carità', Novara, Italy
| | - Matteo Brunelli
- Pathology Unit, Department of Diagnostics and Public Health, University and Hospital Trust of Verona, Verona, Italy
| | - Massimo Di Maio
- Department of Oncology, University of Turin at Ordine Mauriziano Hospital, Torino, 10128, Italy
| |
Collapse
|
11
|
A Mitochondrial Dysfunction and Oxidative Stress Pathway-Based Prognostic Signature for Clear Cell Renal Cell Carcinoma. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9939331. [PMID: 34868460 PMCID: PMC8635875 DOI: 10.1155/2021/9939331] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 09/07/2021] [Accepted: 11/02/2021] [Indexed: 11/17/2022]
Abstract
Mitochondria not only are the main source of ATP synthesis but also regulate cellular redox balance and calcium homeostasis. Its dysfunction can lead to a variety of diseases and promote cancer and metastasis. In this study, we aimed to explore the molecular characteristics and prognostic significance of mitochondrial genes (MTGs) related to oxidative stress in clear cell renal cell carcinoma (ccRCC). A total of 75 differentially expressed MTGs were analyzed from The Cancer Genome Atlas (TCGA) database, including 46 upregulated and 29 downregulated MTGs. Further analysis screened 6 prognostic-related MTGs (ACAD11, ACADSB, BID, PYCR1, SLC25A27, and STAR) and was used to develop a signature. Kaplan-Meier survival and receiver operating characteristic (ROC) curve analyses showed that the signature could accurately distinguish patients with poor prognosis and had good individual risk stratification and prognostic potential. Stratified analysis based on different clinical variables indicated that the signature could be used to evaluate tumor progression in ccRCC. Moreover, we found that there were significant differences in immune cell infiltration between the low- and high-risk groups based on the signature and that ccRCC patients in the low-risk group responded better to immunotherapy than those in the high-risk group (46.59% vs 35.34%, P = 0.008). We also found that the expression levels of these prognostic MTGs were significantly associated with drug sensitivity in multiple ccRCC cell lines. Our study for the first time elucidates the biological function and prognostic significance of mitochondrial molecules associated with oxidative stress and provides a new protocol for evaluating treatment strategies targeting mitochondria in ccRCC patients.
Collapse
|
12
|
Zhang H, Zhu G. Beyond Promoter: The Role of Macrophage in Invasion and Progression of Renal Cell Carcinoma. Curr Stem Cell Res Ther 2021; 15:588-596. [PMID: 32096752 DOI: 10.2174/1574888x15666200225093210] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/28/2019] [Accepted: 12/11/2019] [Indexed: 11/22/2022]
Abstract
Renal cell carcinoma (RCC) is one of the common urologic neoplasms, and its incidence has been increasing over the past several decades; however, its pathogenesis is still unknown up to now. Recent studies have found that in addition to tumor cells, other cells in the tumor microenvironment also affect the biological behavior of the tumor. Among them, macrophages exist in a large amount in tumor microenvironment, and they are generally considered to play a key role in promoting tumorigenesis. Therefore, we summarized the recent researches on macrophage in the invasiveness and progression of RCC in latest years, and we also introduced and discussed many studies about macrophage in RCC to promote angiogenesis by changing tumor microenvironment and inhibit immune response in order to activate tumor progression. Moreover, macrophage interactes with various cytokines to promote tumor proliferation, invasion and metastasis, and it also promotes tumor stem cell formation and induces drug resistance in the progression of RCC. The highlight of this review is to make a summary of the roles of macrophage in the invasion and progression of RCC; at the same time to raise some potential and possible targets for future RCC therapy.
Collapse
Affiliation(s)
- Haibao Zhang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Guodong Zhu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| |
Collapse
|
13
|
Rudzinska M, Czarnecka-Chrebelska KH, Kuznetsova EB, Maryanchik SV, Parodi A, Korolev DO, Potoldykova N, Svetikova Y, Vinarov AZ, Nemtsova MV, Zamyatnin AA. Long Non-Coding PROX1-AS1 Expression Correlates with Renal Cell Carcinoma Metastasis and Aggressiveness. Noncoding RNA 2021; 7:25. [PMID: 33920185 PMCID: PMC8167775 DOI: 10.3390/ncrna7020025] [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: 12/10/2020] [Revised: 04/02/2021] [Accepted: 04/08/2021] [Indexed: 11/16/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) can be specifically expressed in different tissues and cancers. By controlling the gene expression at the transcriptional and translational levels, lncRNAs have been reported to be involved in tumor growth and metastasis. Recent data demonstrated that multiple lncRNAs have a crucial role in renal cell carcinoma (RCC) progression-the most common malignant urogenital tumor. In the present study, we found a trend towards increased PROX1 antisense RNA 1 (PROX1-AS1) expression in RCC specimens compared to non-tumoral margins. Next, we found a positive correlation between PROX1-AS1 expression and the occurrence of distant and lymph node metastasis, higher tumor stage (pT1 vs. pT2 vs. pT3-T4) and high-grade (G1/G2 vs. G3/G4) clear RCC. Furthermore, global demethylation in RCC-derived cell lines (769-P and A498) and human embryonic kidney 293 (HEK293) cells induced a significant increase of PROX1-AS1 expression level, with the most remarkable change in HEK293 cells. In line with this evidence, bisulfite sequencing analysis confirmed the specific demethylation of bioinformatically selected CpG islands on the PROX1-AS1 promoter sequence in the HEK293 cell line but not in the tumor cells. Additionally, the human specimen analysis showed the hemimethylated state of CG dinucleotides in non-tumor kidney tissues, whereas the tumor samples presented the complete, partial, or no demethylation of CpG-islands. In conclusion, our study indicated that PROX1-AS1 could be associated with RCC progression, and further investigations may define its role as a new diagnostic marker and therapeutic target.
Collapse
Affiliation(s)
- Magdalena Rudzinska
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (E.B.K.); (S.V.M.); (A.P.); (M.V.N.)
| | | | - Ekaterina B. Kuznetsova
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (E.B.K.); (S.V.M.); (A.P.); (M.V.N.)
- Laboratory of Epigenetics, Research Centre for Medical Genetics, Moskvorechye str. 1, 115478 Moscow, Russia
| | - Sofya V. Maryanchik
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (E.B.K.); (S.V.M.); (A.P.); (M.V.N.)
| | - Alessandro Parodi
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (E.B.K.); (S.V.M.); (A.P.); (M.V.N.)
| | - Dmitry O. Korolev
- Institute for Urology and Reproductive Health, Sechenov University, 119992 Moscow, Russia; (D.O.K.); (N.P.); (Y.S.); (A.Z.V.)
| | - Nataliya Potoldykova
- Institute for Urology and Reproductive Health, Sechenov University, 119992 Moscow, Russia; (D.O.K.); (N.P.); (Y.S.); (A.Z.V.)
| | - Yulia Svetikova
- Institute for Urology and Reproductive Health, Sechenov University, 119992 Moscow, Russia; (D.O.K.); (N.P.); (Y.S.); (A.Z.V.)
| | - Andrey Z. Vinarov
- Institute for Urology and Reproductive Health, Sechenov University, 119992 Moscow, Russia; (D.O.K.); (N.P.); (Y.S.); (A.Z.V.)
| | - Marina V. Nemtsova
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (E.B.K.); (S.V.M.); (A.P.); (M.V.N.)
- Laboratory of Epigenetics, Research Centre for Medical Genetics, Moskvorechye str. 1, 115478 Moscow, Russia
| | - Andrey A. Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (E.B.K.); (S.V.M.); (A.P.); (M.V.N.)
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
- Department of Biotechnology, Sirius University of Science and Technology, 1 Olympic Ave, 354340 Sochi, Russia
| |
Collapse
|
14
|
Yu B, Zheng X, Sun Z, Cao P, Zhang J, Wang W. IFI16 Can Be Used as a Biomarker for Diagnosis of Renal Cell Carcinoma and Prediction of Patient Survival. Front Genet 2021; 12:599952. [PMID: 33659024 PMCID: PMC7917296 DOI: 10.3389/fgene.2021.599952] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 01/27/2021] [Indexed: 01/21/2023] Open
Abstract
The incidences of renal cell carcinoma (RCC) increase in number each year and account for about 2–3% of all malignant tumors. Many patients have metastasis by the time of diagnosis, and their prognosis is poor. Therefore, it is essential that new diagnostic and prognostic markers for kidney cancer are identified. In this study, we assessed the potential of IFI16 as a diagnostic and prognostic marker for RCC. We analyzed the TCGA and UALCAN databases and found IFI16 to be highly expressed in ccRCC. In addition, high IFI16 levels positively correlated with lymphatic metastasis, tumor stage, and histopathological grade. Kaplan-Meier curve analysis showed that IFI16 expression was related to the prognosis of patients, and high IFI16 expression indicated a worse overall survival (p = 5.1E–0.7). Receiver operating characteristic curve analysis showed that a combination of IFI16 expression and histopathological grade improved predictive accuracy (AUC = 0.697; 95%CI: 0.628–0.765, P < 0.001). Finally, the relative levels of IFI16 in ACHN and Caki-1 cells were higher than that of HK-2 cells by western blotting analysis and RT-PCR. Functional tests showed that knocking down IFI16 expression inhibited migration and invasion in vitro. Therefore, IFI16 is a potential biomarker for the diagnosis and prognosis of RCC patients.
Collapse
Affiliation(s)
- Baozhong Yu
- Department of Urology, Affiliated Beijing Chaoyang Hospital of Capital Medical University, Beijing, China
| | - Xiang Zheng
- Department of Clinical Medicine, Capital Medical University, Beijing, China
| | - Zejia Sun
- Department of Urology, Affiliated Beijing Chaoyang Hospital of Capital Medical University, Beijing, China
| | - Peng Cao
- Department of Clinical Medicine, Capital Medical University, Beijing, China
| | - Jiandong Zhang
- Department of Clinical Medicine, Capital Medical University, Beijing, China
| | - Wei Wang
- Department of Urology, Affiliated Beijing Chaoyang Hospital of Capital Medical University, Beijing, China
| |
Collapse
|
15
|
Zhu J, Liu Z, Zhang Z, Fan Y, Chen Y, He Z, Zhou L, Jin J, Shen C, Yu W. Development and internal validation of nomograms for the prediction of postoperative survival of patients with grade 4 renal cell carcinoma (RCC). Transl Androl Urol 2021; 9:2629-2639. [PMID: 33457235 PMCID: PMC7807345 DOI: 10.21037/tau-19-687] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Background To develop successful prognostic models for grade 4 renal cell carcinoma (RCC) following partial nephrectomy and radical nephrectomy. Methods The nomograms were established based on a retrospective study of 135 patients who underwent partial and radical nephrectomy for grade 4 RCC at the Department of Urology, Peking University First Hospital from January 2013 to October 2018. The predictive performance of the nomograms was assessed by the calibration plot and C-index. The results were validated using bootstrap resampling. Results Aspartate transaminase (AST), the maximum diameter of tumor (cutoff value =7 cm), lymph node metastasis, and the International Metastatic Renal Cell Carcinoma Database Consortium (IMDC) risk group were independent factors for determining the overall survival (OS) and cancer-specific survival (CSS) in multivariate analysis. AST, the maximum diameter of the tumor (cutoff value =7 cm), and lymph node metastasis were found to be independent variables for progression-free survival (PFS) in multivariate analysis. These variables were used for the studies to establish nomograms. All calibration plots revealed excellent predictive accuracy of the models. The C-indexes of the nomograms for predicting OS, CSS and PFS were 0.729 (95% CI, 0.659-0.799), 0.725 (95% CI, 0.654-0.796) and 0.702 (95% CI, 0.626-0.778), respectively. Moreover, the recurrence rate was not associated with open or laparoscopic radical nephrectomy in our cohort (P=0.126). Conclusions We have developed easy-to-use models that are internally validated to predict postoperative 1-, 3-, and 5-year OS, CSS, and PFS rates of grade 4 RCC patients. The new models could aid in identifying high-risk patients, making postoperative therapeutic and follow-up strategies as well as predicting patients' survival after externally validated. Besides, our study shows that the recurrence rate is not associated with open or laparoscopic radical nephrectomy.
Collapse
Affiliation(s)
- Jun Zhu
- Department of Urology, Peking University First Hospital, Beijing, China.,National Urological Cancer Center, Institute of Urology, Peking University, Beijing, China.,Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, China
| | - Zhifu Liu
- Department of Urology, Peking University First Hospital, Beijing, China.,National Urological Cancer Center, Institute of Urology, Peking University, Beijing, China.,Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, China
| | - Zhongyuan Zhang
- Department of Urology, Peking University First Hospital, Beijing, China.,National Urological Cancer Center, Institute of Urology, Peking University, Beijing, China.,Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, China
| | - Yu Fan
- Department of Urology, Peking University First Hospital, Beijing, China.,National Urological Cancer Center, Institute of Urology, Peking University, Beijing, China.,Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, China
| | - Yuke Chen
- Department of Urology, Peking University First Hospital, Beijing, China.,National Urological Cancer Center, Institute of Urology, Peking University, Beijing, China.,Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, China
| | - Zhisong He
- Department of Urology, Peking University First Hospital, Beijing, China.,National Urological Cancer Center, Institute of Urology, Peking University, Beijing, China.,Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, China
| | - Liqun Zhou
- Department of Urology, Peking University First Hospital, Beijing, China.,National Urological Cancer Center, Institute of Urology, Peking University, Beijing, China.,Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, China
| | - Jie Jin
- Department of Urology, Peking University First Hospital, Beijing, China.,National Urological Cancer Center, Institute of Urology, Peking University, Beijing, China.,Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, China
| | - Cheng Shen
- Department of Urology, Peking University First Hospital, Beijing, China.,National Urological Cancer Center, Institute of Urology, Peking University, Beijing, China.,Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, China
| | - Wei Yu
- Department of Urology, Peking University First Hospital, Beijing, China.,National Urological Cancer Center, Institute of Urology, Peking University, Beijing, China.,Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, China
| |
Collapse
|
16
|
Liu Q, Gu J, Zhang E, He L, Yuan ZX. Targeted Delivery of Therapeutics to Urological Cancer Stem Cells. Curr Pharm Des 2020; 26:2038-2056. [PMID: 32250210 DOI: 10.2174/1381612826666200403131514] [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: 11/18/2019] [Accepted: 02/27/2020] [Indexed: 02/07/2023]
Abstract
Urological cancer refers to cancer in organs of the urinary system and the male reproductive system. It mainly includes prostate cancer, bladder cancer, renal cancer, etc., seriously threatening patients' survival. Although there are many advances in the treatment of urological cancer, approved targeted therapies often result in tumor recurrence and therapy failure. An increasing amount of evidence indicated that cancer stem cells (CSCs) with tumor-initiating ability were the source of treatment failure in urological cancer. The development of CSCstargeted strategy can provide a possibility for the complete elimination of urological cancer. This review is based on a search of PubMed, Google scholar and NIH database (http://ClinicalTrials.gov/) for English language articles containing the terms: "biomarkers", "cancer stem cells", "targeting/targeted therapy", "prostate cancer", bladder cancer" and "kidney cancer". We summarized the biomarkers and stem cell features of the prostate, bladder and renal CSCs, outlined the targeted strategies for urological CSCs from signaling pathways, cytokines, angiogenesis, surface markers, elimination therapy, differentiation therapy, immunotherapy, microRNA, nanomedicine, etc., and highlighted the prospects and future challenges in this research field.
Collapse
Affiliation(s)
- Qiang Liu
- Yaopharma Co., Ltd. Chongqing, China
| | - Jian Gu
- College of Pharmacy, Southwest Minzu University, Chengdu, Sichuan, China
| | - E Zhang
- Officers college of PAP, Chengdu, Sichuan, China
| | - Lili He
- College of Pharmacy, Southwest Minzu University, Chengdu, Sichuan, China
| | - Zhi-Xiang Yuan
- College of Pharmacy, Southwest Minzu University, Chengdu, Sichuan, China
| |
Collapse
|
17
|
Bi R, Deng Y, Tang C, Xuan L, Xu B, Du Y, Wang C, Wei W. Andrographolide sensitizes human renal carcinoma cells to TRAIL‑induced apoptosis through upregulation of death receptor 4. Oncol Rep 2020; 44:1939-1948. [PMID: 33000263 PMCID: PMC7551412 DOI: 10.3892/or.2020.7737] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 07/23/2020] [Indexed: 12/11/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) selectively induces apoptosis in cancer cells, with minimal toxicity to normal tissues. However, accumulating evidence suggests that certain cancer types are insensitive to TRAIL signaling. The aim of this study was to identify an effective combination regimen, which can overcome TRAIL resistance in renal cancer cell. Herein, we found that human renal carcinoma cells (RCCs) are widely resistant to TRAIL-mediated growth inhibition and subsequently identified that andrographolide (Andro), a major constituent of Andrographis paniculate, an annual herbaceous plant in the family Acanthaceae, counteracts TRAIL resistance in RCCs. Combined treatment with TRAIL and Andro suppressed cell viability as determined by MTS and proliferation as determined by EdU in a dose-dependent manner and inactivated the clonogenic and migration ability of RCCs. Andro significantly enhances TRAIL-mediated cell cycle arrest at the G2/M phase as determined by flow cytometry and senescence. Moreover, Andro restored TRAIL signaling, which in turns activated pro-apoptosis caspases as determined by immunoblot assay. The TRAIL receptor, death receptor (DR)4, but not DR5, was found to be significantly upregulated in Andro-treated RCC cells, which contributed to the role of Andro as a TRAIL sensitizer. The present study demonstrated that the combined treatment of Andro and TRAIL has potential therapeutic value against renal cancer.
Collapse
Affiliation(s)
- Ran Bi
- Department of Urology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yuyou Deng
- Department of Urology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Chao Tang
- Department of Urology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Lei Xuan
- Department of Urology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Bo Xu
- Department of Urology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yujun Du
- Department of Nephrology, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Chunxi Wang
- Department of Urology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Wei Wei
- Key Laboratory of Organ Regeneration and Transplantation of The Ministry of Education, Institute of Translational Medicine, Institute of Virology and AIDS Research, The First Hospital of Jilin University, Changchun, Jilin 130061, P.R. China
| |
Collapse
|
18
|
Guo B, Liu M. Letter to the editor regarding: comparison of survival outcomes in patients with metastatic papillary vs. clear-cell renal cell carcinoma: a propensity-score analysis. World J Urol 2020; 39:4291-4292. [PMID: 32785763 DOI: 10.1007/s00345-020-03404-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 08/05/2020] [Indexed: 11/30/2022] Open
Affiliation(s)
- Boda Guo
- Department of Urology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China.,Graduate School of Peking Union Medical College, Beijing, 100730, China
| | - Ming Liu
- Department of Urology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China. .,Graduate School of Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
19
|
Li P, Hu J, Shi B, Tie J. Baicalein enhanced cisplatin sensitivity of gastric cancer cells by inducing cell apoptosis and autophagy via Akt/mTOR and Nrf2/Keap 1 pathway. Biochem Biophys Res Commun 2020; 531:320-327. [PMID: 32800561 DOI: 10.1016/j.bbrc.2020.07.045] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 07/09/2020] [Indexed: 01/02/2023]
Abstract
Baicalein is a natural flavonoid with various pharmacological activities including antitumor. The synergistic anti-cancer effect of the combination of baicalein and Cisplatin (DDP) on gastric cancer (GC) has not been reported. MTT assay and colony formation assay were used to determine the inhibitory effect of the combination of baicalein and DDP on cell survival. Invasive assay was performed to test the effects of baicalein and DDP on cell invasive capability. A flow cytometric analysis was conducted to determine the apoptosis-induced effects of baicalein on GC cells, especially SGC-7901/DDP (resistant to DDP). Confocal laser microscope and real-time PCR were used to test autophagy-induced effects of baicalein on SGC-7901 and SGC-7901/DDP cells. Western blotting was performed to investigate the molecular mechanisms of baicalein inducing apoptosis and autophagy. Our study showed that baicalein could inhibit cell proliferation of MGC-803, HGC-27, SGC-7901 and SGC-7901/DDP, and the inhibitory effect was extremely enhanced when combining with DDP. Additionally, combination of baicalein and DDP suppressed the invasive capability and induced apoptosis and autophagy in both SGC-7901 and SGC-7901/DDP, and the effect was stronger than that of DDP or baicalein alone. The further molecular mechanism analysis indicated that baicalein modulated the activities of Akt/mTOR and Nrf2/Keap 1 signaling. Our study demonstrated that baicalein enhanced DDP sensitivity of SGC-7901/DDP gastric cancer cells by inducing apoptosis and autophagy via Akt/mTOR and Nrf2/Keap 1 pathway.
Collapse
Affiliation(s)
- Ping Li
- Department of Biological Science and Technology, Changzhi University, Changzhi, 046011, China
| | - Jianran Hu
- Department of Biological Science and Technology, Changzhi University, Changzhi, 046011, China.
| | - Baozhong Shi
- Department of Biological Science and Technology, Changzhi University, Changzhi, 046011, China
| | - Jun Tie
- Department of Biological Science and Technology, Changzhi University, Changzhi, 046011, China
| |
Collapse
|
20
|
The long noncoding RNA EMBP1 inhibits the tumor suppressor miR-9-5p and promotes renal cell carcinoma tumorigenesis. Nefrologia 2020; 40:429-439. [DOI: 10.1016/j.nefro.2019.12.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 12/04/2019] [Accepted: 12/15/2019] [Indexed: 12/26/2022] Open
|
21
|
Simon AG, Esser LK, Ellinger J, Branchi V, Tolkach Y, Müller S, Ritter M, Kristiansen G, Muders MH, Mayr T, Toma MI. Targeting glycolysis with 2-deoxy-D-glucose sensitizes primary cell cultures of renal cell carcinoma to tyrosine kinase inhibitors. J Cancer Res Clin Oncol 2020; 146:2255-2265. [PMID: 32533404 DOI: 10.1007/s00432-020-03278-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 05/30/2020] [Indexed: 12/23/2022]
Abstract
PURPOSE To investigate the synergistic effect of glycolysis inhibition on therapy answer to tyrosine kinase inhibitors in renal carcinoma. METHODS Primary cell cultures from 33 renal tumors including clear cell RCC (ccRCC), papillary RCC and the rare subtype chromophobe RCC as well as two metastases of ccRCC were obtained and cultivated. The patient-derived cells were verified by immunohistochemistry. CcRCC cells were further examined by exon sequencing of the von Hippel-Lindau gene (VHL) and by RNA-sequencing. Next, cell cultures of all subtypes of RCC were exposed to increasing doses of various tyrosine kinase inhibitors (axitinib, cabozantinib and pazopanib) and the glycolysis inhibitor 2-deoxy-D-glucose, alone or combined. CellTiter-Glo® Luminescence assay and Crystal Violet staining were used to assess the inhibition of glycolysis and the viability of the cultured primary cells. RESULTS The cells expressed characteristic tissue markers and, in case of ccRCC cultures, the VHL status of the tumor they derived from. An upregulation of HK1, PFKP and SLC2A1 was observed, while components of the respiratory chain were downregulated, confirming a metabolic shift towards aerobic glycolysis. The tumors displayed variable individual responses for the therapeutics. All subtypes of RCC were susceptible to cabozantinib treatment indicated by decreased proliferation. Adding 2-deoxy-D-glucose to tyrosine kinase inhibitors decreased ATP production and increased the susceptibility of ccRCC to pazopanib treatment. CONCLUSION This study presents a valuable tool to cultivate even uncommon and rare renal cancer subtypes and allows testing of targeted therapies as a personalized approach as well as testing new therapies such as glycolysis inhibition in an in vitro model.
Collapse
Affiliation(s)
- Adrian Georg Simon
- Department of Pathology, Institute of Pathology, University Hospital Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Laura Kristin Esser
- Department of Pathology, Institute of Pathology, University Hospital Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Jörg Ellinger
- Department of Urology, University Hospital Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Vittorio Branchi
- Department of General, Visceral, Thoracic and Vascular Surgery, Institute of General, Visceral, Thoracic and Vascular Surgery, University Hospital Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Yuri Tolkach
- Department of Pathology, Institute of Pathology, University Hospital Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Stefan Müller
- Department of Urology, University Hospital Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Manuel Ritter
- Department of Urology, University Hospital Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Glen Kristiansen
- Department of Pathology, Institute of Pathology, University Hospital Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Michael Helmut Muders
- Department of Pathology, Institute of Pathology, University Hospital Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Thomas Mayr
- Department of Pathology, Institute of Pathology, University Hospital Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Marieta Ioana Toma
- Department of Pathology, Institute of Pathology, University Hospital Bonn, Venusberg Campus 1, 53127, Bonn, Germany.
| |
Collapse
|
22
|
Jin Y, Huang R, Xia Y, Huang C, Qiu F, Pu J, He X, Zhao X. Long Noncoding RNA KIF9-AS1 Regulates Transforming Growth Factor-β and Autophagy Signaling to Enhance Renal Cell Carcinoma Chemoresistance via microRNA-497-5p. DNA Cell Biol 2020; 39:1096-1103. [PMID: 32343913 DOI: 10.1089/dna.2020.5453] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Renal cell carcinoma (RCC) has been regarded as one of the most malignant tumor types. Chemotherapy (such as sorafenib) is used as common strategy for treating RCC. To date, whether long noncoding RNA KIF9-AS1 is involved in RCC progression and drug resistance remains unknown. In this investigation, we detected gene expression levels by western blot and RT-qPCR. MTT and TUNEL experiments were used to show cell viability and apoptosis, respectively. KIF9-AS1 overexpression led to enhanced cell viability, increased IC50 value of sorafenib, and decreased apoptosis. miR-497-5p acted as key interaction factor for KIF9-AS1 in RCC. More importantly, we found that transforming growth factor-β and autophagy signaling pathways were both critical effectors for mediating KIF9-AS1/miR-497-5p axis-induced drug resistance phenotypes (cell viability, IC50, apoptosis) of RCC. In conclusion, our study revealed that KIF9-AS1 played a positive role in drug resistance of RCC cells to sorafenib, potentially driving the development of targeted diagnostic and therapeutical approaches.
Collapse
Affiliation(s)
- Yichen Jin
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Ru Huang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Yanfu Xia
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Chen Huang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Feng Qiu
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Jinxian Pu
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Xuefeng He
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Xiaojun Zhao
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
23
|
Yu YP, Cai LC, Wang XY, Cheng SY, Zhang DM, Jian WG, Wang TD, Yang JK, Yang KB, Zhang C. BMP8A promotes survival and drug resistance via Nrf2/TRIM24 signaling pathway in clear cell renal cell carcinoma. Cancer Sci 2020; 111:1555-1566. [PMID: 32128917 PMCID: PMC7226287 DOI: 10.1111/cas.14376] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 12/14/2022] Open
Abstract
There is increasing evidence that bone morphogenetic proteins (BMP) are involved in the proliferation and drug tolerance of kidney cancer. However, the molecular mechanism of BMP8A in renal cell proliferation and drug tolerance is not clear. Here we showed that BMP8A was highly expressed in renal cell carcinoma, which suggests a poor prognosis of ccRCC. Promotion of cell proliferation and inhibition of apoptosis were detected by CCK‐8 assay, Trypan Blue staining, flow cytometry and bioluminescence. BMP8A promoted resistance of As2O3 by regulating Nrf2 and Wnt pathways in vitro and in vivo. Mechanistically, BMP8A enhanced phosphorylation of Nrf2, which, in turn, inhibited Keap1‐mediated Nrf2 ubiquitination and, ultimately, promoted nuclear translocation and transcriptional activity of Nrf2. Nrf2 regulates the transcription of TRIM24 detected by ChIP‐qPCR. BMP8A was highly expressed in ccRCC, which suggests a poor prognosis. BMP8A was expected to be an independent prognostic molecule for ccRCC. On the one hand, activated Nrf2 regulated reactive oxygen balance, and on the other hand, by regulating the transcription level of TRIM24, it was involved in the regulation of the Wnt pathway to promote the proliferation, invasion and metastasis of ccRCC and the resistance of As2O3. Taken together, our findings describe a regulatory axis where BMP8A promotes Nrf2 phosphorylation and activates TRIM24 to promote survival and drug resistance in ccRCC.
Collapse
Affiliation(s)
- Yi-Peng Yu
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Li-Cheng Cai
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xing-Yuan Wang
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Si-Yu Cheng
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Da-Ming Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wen-Gang Jian
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Teng-da Wang
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jian-Kun Yang
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Kong-Bin Yang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Cheng Zhang
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
24
|
Saeednejad Zanjani L, Madjd Z, Axcrona U, Abolhasani M, Rasti A, Asgari M, Fodstad Ø, Andersson Y. Cytoplasmic expression of B7-H3 and membranous EpCAM expression are associated with higher grade and survival outcomes in patients with clear cell renal cell carcinoma. Ann Diagn Pathol 2020; 46:151483. [PMID: 32143173 DOI: 10.1016/j.anndiagpath.2020.151483] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/04/2020] [Accepted: 02/15/2020] [Indexed: 12/14/2022]
Abstract
B7-H3 and EpCAM are overexpressed in cancer and play a role in tumorigenesis and metastasis. In this study, the membranous, cytoplasmic and nuclear expression levels of B7-H3 and EpCAM biomarkers were mapped in three major subtypes of renal cell carcinoma (RCC). Expression of B7-H3 and EpCAM were evaluated using immunohistochemistry in RCC samples on tissue microarrays (TMAs), including clear cell RCCs (ccRCCs), type I and II papillary RCCs (pRCCs), and chromophobe RCCs (chRCCs). The association between B7-H3 and EpCAM expression and clinicopathological features as well as survival outcomes was determined. There was a statistically significant difference between B7-H3 and EpCAM expression among the different RCC subtypes. In ccRCC, higher cytoplasmic expression of B7-H3 was significantly associated with increase in nucleolar grade, lymph node invasion (LNI), invasion of the Gerota's fascia, and tumor necrosis, while no association was found with the membranous and nuclear expression. Moreover tumors with cytoplasmic expression of B7-H3 tended to have a worse prognosis for disease-specific survival (DSS) than those with membranous expression. In case of EpCAM, increased membranous expression of EpCAM was associated with nucleolar grade and tumor necrosis in ccRCC. Additionally, membranous EpCAM expression added prognostic value in patients with ccRCC who had high nucleolar grade versus low nucleolar grade. Moreover, membranous EpCAM expression was found to be an independent favorable prognostic marker for progression-free survival (PFS) in ccRCC. Our results demonstrated that higher cytoplasmic B7-H3 and membranous EpCAM expression are clinically significant in ccRCC and are associated with more aggressiveness tumor behavior.
Collapse
Affiliation(s)
| | - 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
| | - Ulrika Axcrona
- Department of Pathology, The Norwegian Radium Hospital, Institute of Clinical Medicine, Oslo University Hospital, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Maryam Abolhasani
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran; Hasheminejad Kidney Center, Iran University of Medical Sciences, (IUMS), Tehran, Iran
| | - 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
| | - Øystein Fodstad
- Department of Tumor Biology, Norwegian Radium Hospital, Oslo University Hospital Radiumhospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Yvonne Andersson
- Department of Tumor Biology, Norwegian Radium Hospital, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| |
Collapse
|
25
|
Zhu R, Ge J, Ma J, Zheng J. Carcinoembryonic antigen related cell adhesion molecule 6 promotes the proliferation and migration of renal cancer cells through the ERK/AKT signaling pathway. Transl Androl Urol 2019; 8:457-466. [PMID: 31807423 DOI: 10.21037/tau.2019.09.02] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background Carcinoembryonic antigen related cell adhesion molecule 6 (CEACAM6) is a versatile glycoprotein and a member of the CEACAM family. Studies suggested that it served as a diagnostic and prognostic biomarker in some malignancies. In addition, it is involved in tumorigenesis by stimulating proliferation, suppressing apoptosis, facilitating migration and invasion, promoting angiogenesis, and inducing drug resistance. In the present study, we demonstrated the oncogenic effects of CEACAM6 in clear cell renal cell carcinoma (ccRCC). Methods CEACAM6 expression was detected by quantitative real-time PCR (qRT-PCR), immunohistochemical staining and western blot in ccRCC tumor tissues and cell lines. Survival analysis was performed using the data of TCGA database. Cell proliferation and migration were detected by CCK-8 and transwell assays with the overexpression or silencing of CEACAM6. LY294002 was used to block the activation of PI3K/AKT pathway. Associated pathway proteins were detected by western blot. Results CEACAM6 was upregulated in ccRCC cell lines and tumor tissues. Longer overall survival was observed in patients with relatively low CEACAM6 levels. Furthermore, overexpression of CEACAM6 promoted the proliferation and migration of ccRCC cells. Conversely, shRNA-mediated CEACAM6 depletion modulated those changes. Further investigation demonstrated that the ERK/AKT signaling pathway activation played a pivotal role. In addition, PI3K/AKT pathway blockade abrogated the effects of CEACAM6 overexpression. Conclusions Aberrantly high expression of CEACAM6 is a stimulus for the formation and progression of ccRCC.
Collapse
Affiliation(s)
- Rujian Zhu
- Department of Urology, The Affiliated Shanghai No.10 People's Hospital, Nanjing Medical University, Shanghai 200072, China.,Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Jiong Ge
- Department of Radiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Junjie Ma
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Junhua Zheng
- Department of Urology, The Affiliated Shanghai No.10 People's Hospital, Nanjing Medical University, Shanghai 200072, China.,Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| |
Collapse
|
26
|
Liu S, Wang Y, Li W, Yu S, Wen Z, Chen Z, Lin F. miR-221-5p acts as an oncogene and predicts worse survival in patients of renal cell cancer. Biomed Pharmacother 2019; 119:109406. [PMID: 31514066 DOI: 10.1016/j.biopha.2019.109406] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 08/24/2019] [Accepted: 08/28/2019] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND Renal cell carcinoma(RCC) is one of the most common malignancies in kidney, and usually leads to poor prognosis. Therefore, identifying novel biomarkers for predicting the progression and prognosis of RCC is essential. The purpose of this study is aimed to evaluate the function of miR-221-5p in RCC and the clinical value of miR-221-5p in RCC prognosis after surgery. MATERIALS AND METHODS In our study, RT-qPCR, wound scratch assay, cell proliferation assay, transwell assay, and flow cytometry assay were performed to explore miR-221-5p expression level and its proliferation, migration and apoptosis in clear cell RCC(ccRCC). Besides, we collected 196 formalin-fixed and paraffin-embedded (FFPE) tissue samples of patients who received partial or radical nephrectomy from May 2006 to October 2016 at Shenzhen Traditional Chinese Medicine Hospital and People's Liberation Army 303 Hospital. The relative levels of miR-221-5p from the FFPE tissue samples was detected by RT-qPCR. The Kaplan-Meier method, Cox regression analyses, and ROC curve analysis were performed to approve the effect of the miR-221-5p expression on patient survival. RESULTS In our study, we found that miR-221-5p is significantly upregulated in ccRCC tissues and ccRCC cell lines. Moreover, miR-221-5p promotes cell proliferation, mobility, and inhibits cell apoptosis in 786-O and ACHN cell lines. The Kaplan-Meier analysis indicated that patients with high expression of miR-221-5p had a significantly poor prognosis (P = 0.013). The Cox regression analyses showed that patients with high expression of miR-221-5p remained to have a shorter overall survival (P = 0.025). The ROC curve of miR-221-5p expression combined with tumor stage showed an area under the curve of 0.658 (P < 0.001). CONCLUSION Our results indicated that miR-221-5p might not only be an oncogene in ccRCC cells but also might be an independent prognosis factor of ccRCC.
Collapse
Affiliation(s)
- Shangwen Liu
- Department of Urology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518036, PR China
| | - Yadong Wang
- Department of Urology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518036, PR China
| | - Wengang Li
- Department of Urology, People's Liberation Army 303 Hospital, Nanning, Guangxi 530021, PR China
| | - Shaolong Yu
- Department of Urology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518036, PR China
| | - Zhipeng Wen
- Department of Urology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518036, PR China
| | - Zhifeng Chen
- Department of Urology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518036, PR China
| | - Feng Lin
- Department of Urology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518036, PR China.
| |
Collapse
|
27
|
Zhong B, Qin Z, Zhou H, Yang F, Wei K, Jiang X, Jia R. microRNA-505 negatively regulates HMGB1 to suppress cell proliferation in renal cell carcinoma. J Cell Physiol 2019; 234:15025-15034. [PMID: 30644098 PMCID: PMC6590343 DOI: 10.1002/jcp.28142] [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: 09/27/2018] [Accepted: 01/02/2019] [Indexed: 01/24/2023]
Abstract
microRNAs have been recognized to regulate a wide range of biology of renal cell carcinoma (RCC). Although miR-505 has been reported to play as a suppressor in several human tumors, the physiological function of miR-505 in RCC still remain unknown. Therefore, the role of miR-505 and relevant regulatory mechanisms were investigated in RCC in this study. Quantitative real-time polymerase chain reaction was conducted to detect the expression of miR-505 and high mobility group box 1 (HMGB1) in both RCC tissues and cell lines. Immunohistochemical staining was used to assess the correlation between HMGB1 expression and PCNA expression in RCC tissues. Subsequently, the effects of miR-505 on proliferation were determined in vitro using cell counting kit-8 proliferation assays and 5-ethynyl-2'-deoxyuridine incorporation. The molecular mechanism underlying the relevance between miR-505 and HMGB1 was confirmed by luciferase assay. Xenograft tumor formation was used to reflect the proliferative capacity of miR-505 in vivo experiments. Overall, a relatively lower miR-505 and higher HMGB1 expression in RCC specimens and cell lines were found. HMGB1 was verified as a direct target of miR-505 by luciferase assay. In vitro, overexpression of miR-505 negatively regulates HMGB1 to suppress the proliferation in Caki-1; meanwhile, knock-down of miR-505 negatively regulates HMGB1 to promote the proliferation in 769P. In addition, in vivo overexpression of miR-505 could inhibit tumor cell proliferation in RCC by xenograft tumor formation. Therefore, miR-505, as a tumor suppressor, negatively regulated HMGB1 to suppress the proliferation in RCC, and might serve as a novel therapeutic target for RCC clinical treatment.
Collapse
Affiliation(s)
- Bing Zhong
- Department of UrologyNanjing First Hospital, Nanjing Medical UniversityNanjingChina,Department of UrologyThe Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical UniversityHuai'anChina
| | - Zhiqiang Qin
- Department of UrologyNanjing First Hospital, Nanjing Medical UniversityNanjingChina
| | - Hui Zhou
- Department of UrologyHongze People's HospitalHuai'anChina
| | - Fengming Yang
- Department of OncologyFirst Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Ke Wei
- Department of Thoracic SurgeryFirst Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Xi Jiang
- Department of UrologyThe Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical UniversityHuai'anChina
| | - Ruipeng Jia
- Department of UrologyNanjing First Hospital, Nanjing Medical UniversityNanjingChina
| |
Collapse
|
28
|
Antioxidant Defenses: A Context-Specific Vulnerability of Cancer Cells. Cancers (Basel) 2019; 11:cancers11081208. [PMID: 31434226 PMCID: PMC6721511 DOI: 10.3390/cancers11081208] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/12/2019] [Accepted: 08/13/2019] [Indexed: 12/11/2022] Open
Abstract
Reactive oxygen species (ROS) are well known for their capacity to cause DNA damage, augment mutagenesis, and thereby promote oncogenic transformation. Similarly, agents that reduce ROS levels (antioxidants) are frequently thought to have anti-cancer properties given their propensity to minimize DNA damage and mutagenesis. However, numerous clinical studies focused on antioxidants suggest that this is a facile premise and that antioxidant capacity can be important for cancer cells in a similar fashion to normal cells. As a consequence of this realization, numerous laboratories have been motivated to investigate the biological underpinnings explaining how and when antioxidant activity can potentially be beneficial to cancer cells. Relatedly, it has become clear that the reliance of cancer cells on antioxidant activity in certain contexts represents a potential vulnerability that could be exploited for therapeutic gain. Here, we review some of the recent, exciting findings documenting how cancer cells utilized antioxidant activity and under what circumstances this activity could represent an opportunity for selective elimination of cancer cells.
Collapse
|
29
|
Zheng Y, Lan T, Wei D, Zhang G, Hou G, Yuan J, Yan F, Wang F, Meng P, Yang X, Chen G, Zhu Z, Lu Z, He W, Yuan J. Coupling the near-infrared fluorescent dye IR-780 with cabazitaxel makes renal cell carcinoma chemotherapy possible. Biomed Pharmacother 2019; 116:109001. [DOI: 10.1016/j.biopha.2019.109001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/12/2019] [Accepted: 05/14/2019] [Indexed: 01/22/2023] Open
|
30
|
Ha M, Jeong H, Roh JS, Lee B, Han ME, Oh SO, Sohn DH, Kim YH. DYSF expression in clear cell renal cell carcinoma: A retrospective study of 2 independent cohorts. Urol Oncol 2019; 37:735-741. [PMID: 31377166 DOI: 10.1016/j.urolonc.2019.07.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 06/28/2019] [Accepted: 07/11/2019] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Renal cell carcinoma (RCC) is the most typical type of kidney cancer in adults. Hypercalcemia is a well known paraneoplastic syndrome associated with RCC and recent studies have reported that hypercalcemia is closely related to the poor prognosis of RCC patients. Clear cell RCC (ccRCC) is the most common and aggressive subtype of RCC. Although the histological classification of RCC is important for determination of appropriate treatment strategies, effective biomarkers for predicting prognosis of ccRCC have not yet been identified. Since calcium levels affect the prognosis of RCC patients, we evaluated whether the calcium-sensing genes on the plasma membrane, including those encoding calcium channels, CaSR, GPRC6a, and DYSF, could be used as biomarkers to predict the prognosis of ccRCC patients. METHODS Information from 537 patients from The Cancer Genome Atlas (TCGA; n = 446) and International Cancer Genome Consortium (ICGC; n = 91) was used in this study. Among these genes, DYSF was the only gene whose expression correlated with overall survival of both TGCA and ICGC patients. RESULTS Although DYSF gene expression was higher in ccRCC tissue than in normal kidney tissue, Kaplan-Meier curves showed that the survival rate of ccRCC patients with high DYSF expression was significantly higher than that of patients with low DYSF expression (TCGA, P < 0.0001; ICGC, P = 0.0011). We also validated the potential of DYSF as a prognostic biomarker for ccRCC by conducting a time-dependent area under the curve (AUC) analysis and 5-years receiver operating characteristic curve analysis. Finally, multivariate regression analysis revealed that the expression of DYSF is independent of other prognostic parameters (TCGA, P = 0.017; ICGC, P = 0.006). CONCLUSIONS These results suggested that DYSF may play a suppressive role in the progression of ccRCC and could act as a promising prognostic biomarker for predicting the survival of ccRCC patients.
Collapse
Affiliation(s)
- Mihyang Ha
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Hoim Jeong
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Jong Seong Roh
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Beomgu Lee
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Myoung-Eun Han
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Sae-Ock Oh
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Dong Hyun Sohn
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Republic of Korea.
| | - Yun Hak Kim
- Department of Anatomy and Department of Biomedical Informatics, Pusan National University School of Medicine, Yangsan, Republic of Korea; Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea.
| |
Collapse
|
31
|
He J, He J, Min L, He Y, Guan H, Wang J, Peng X. Extracellular vesicles transmitted miR-31-5p promotes sorafenib resistance by targeting MLH1 in renal cell carcinoma. Int J Cancer 2019; 146:1052-1063. [PMID: 31259424 DOI: 10.1002/ijc.32543] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 06/18/2019] [Indexed: 02/05/2023]
Abstract
Sorafenib provides survival benefits in patients with advanced renal cell carcinoma (RCC), but its use is hampered by acquired drug resistance. It is important to fully clarify the molecular mechanisms of sorafenib resistance, which can help to avoid, delay or reverse drug resistance. Extracellular vesicles (EVs) can mediate intercellular communication by delivering effector molecules between cells. Here, we studied whether EVs are involved in sorafenib resistance of RCC and its possible molecular mechanisms. Using differential centrifugation, EVs were isolated from established sorafenib-resistant RCC cells (786-0 and ACHN), and EVs derived from sorafenib-resistant cells were uptaken by sensitive parental RCC cells and thus promoted drug resistance. Elevated exogenous miR-31-5p within EVs effectively downregulated MutL homolog 1 (MLH1) expression and thus promoted sorafenib resistance in vitro. Mice experiments also confirmed that miR-31-5p could mediate drug sensitivity in vivo. In addition, low expression of MLH1 was observed in sorafenib-resistant RCC cells and upregulation of MLH1 expression restored the sensitivity of resistant cell lines to sorafenib. Finally, miR-31-5p level in circulating EVs of RCC patients with progressive disease (PD) during sorafenib therapy was higher when compared to that in the pretherapy status. In conclusion, EVs shuttled miR-31-5p can transfer resistance information from sorafenib-resistant cells to sensitive cells by directly targeting MLH1, and thus magnify the drug resistance information to the whole tumor. Furthermore, miR-31-5p and MLH1 could be promising predictive biomarkers and therapeutic targets to prevent sorafenib resistance.
Collapse
Affiliation(s)
- Jinlan He
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,Department of Head and Neck Cancer, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jianxiong He
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Li Min
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yan He
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hui Guan
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jingjing Wang
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xingchen Peng
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
32
|
Buti S, Leonetti A, Lattanzi E, D’Abbiero N, Bersanelli M. Role of stereotactic body radiation therapy for the management of renal cell carcinoma: tailoring treatment in the era of the "embarrassment of riches". ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:S106. [PMID: 31576313 PMCID: PMC6685901 DOI: 10.21037/atm.2019.05.04] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 05/06/2019] [Indexed: 11/06/2022]
Affiliation(s)
- Sebastiano Buti
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | | | | | | | - Melissa Bersanelli
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
- Medicine and Surgery Department, University of Parma, Parma, Italy
| |
Collapse
|
33
|
Quan J, Pan X, Li Y, Hu Y, Tao L, Li Z, Zhao L, Wang J, Li H, Lai Y, Zhou L, Lin C, Gui Y, Ye J, Zhang F, Lai Y. MiR-23a-3p acts as an oncogene and potential prognostic biomarker by targeting PNRC2 in RCC. Biomed Pharmacother 2019; 110:656-666. [PMID: 30551118 DOI: 10.1016/j.biopha.2018.11.065] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 11/11/2018] [Accepted: 11/19/2018] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Renal cell carcinoma (RCC) is a most common kidney malignancy, with atypical symptoms in the early stage and poor outcome in the late stage. Recently, emerging evidence revealed that some miRNAs play an essential role in the tumorigenesis and progression of RCC. Therefore, the aim of this study is that understand the detailed molecular mechanism of miR-23a-3p in RCC and identify its potential clinical value. METHODS In this study, RT-qPCR, wound scratch assay, cell proliferation assay, transwell assay and flow cytometry assay were performed to detect miR-23a-3p expression and its proliferation, migration and apoptosis in RCC. The bioinformatics analysis, RT-qPCR, western blot and luciferase reporter assay were performed to discern and examine the relationship between miR-23a-3p and its potential targets. Moreover, we analyzed the relationship between miR-23a-3p expression and clinicopathological variables or overall survival (OS) from 118 formalin-fixed paraffin-embedded RCC samples. RESULTS miR-23a-3p is significantly up-regulated in RCC tissue samples, RCC cell lines and the TCGA database. Upregulating miR-23a-3p enhances, while silencing miR-23a-3p suppresses cell viability, proliferation and mobility in ACHN and 786-O cell lines. Besides, overexpression of miR-23a-3p inhibits the cell apoptosis. Then our study further reveals that miR-23a-3p regulates tumorigenesis by targeting Proline-Rich Nuclear Receptor Coactivator 2 (PNRC2). Also, the cox proportional hazard regression analysis indicates that low expression of miR-23a-3p patients has a remarkable longer OS. CONCLUSIONS Our results reveals that miR-23a-3p may not only serve as a new biomarker for prognosis but also serve as a new therapeutic strategy in the RCC treatment.
Collapse
Affiliation(s)
- Jing Quan
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China; Anhui Medical University, Hefei, Anhui, 230032, P.R. China; Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, Guangdong 518036, P.R. China.
| | - Xiang Pan
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China; Anhui Medical University, Hefei, Anhui, 230032, P.R. China; Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, Guangdong 518036, P.R. China.
| | - Yawen Li
- Anhui Medical University, Hefei, Anhui, 230032, P.R. China; Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, Guangdong 518036, P.R. China.
| | - Yimin Hu
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China.
| | - Lingzhi Tao
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China.
| | - Zuwei Li
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China.
| | - Liwen Zhao
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China; Anhui Medical University, Hefei, Anhui, 230032, P.R. China; Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, Guangdong 518036, P.R. China.
| | - Jingyao Wang
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China; Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, Guangdong 518036, P.R. China.
| | - Hang Li
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China.
| | - Yulin Lai
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China.
| | - Liang Zhou
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China.
| | - Canbin Lin
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China; Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, Guangdong 518036, P.R. China.
| | - Yaoting Gui
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, Guangdong 518036, P.R. China.
| | - Jing Ye
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, Guangdong 518036, P.R. China.
| | - Fangting Zhang
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, Guangdong 518036, P.R. China.
| | - Yongqing Lai
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China; Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, Guangdong 518036, P.R. China.
| |
Collapse
|
34
|
Zhai W, Li S, Zhang J, Chen Y, Ma J, Kong W, Gong D, Zheng J, Xue W, Xu Y. Sunitinib-suppressed miR-452-5p facilitates renal cancer cell invasion and metastasis through modulating SMAD4/SMAD7 signals. Mol Cancer 2018; 17:157. [PMID: 30419914 PMCID: PMC6231268 DOI: 10.1186/s12943-018-0906-x] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 10/15/2018] [Indexed: 12/11/2022] Open
Abstract
PURPOSE Although microRNAs (miRNAs) were revealed as crucial modulators in tumor metastasis and target therapy, our understanding of their roles in metastatic renal cell carcinoma (mRCC) and Sunitinib treatment was limited. Here we sought to identify human miRNAs that acted as key regulators in renal cancer metastasis and Sunitinib treatment. EXPERIMENTAL DESIGN We focused on 2 published microarray data to select out our anchored miRNA and then explored the roles of miR-452-5p both in vitro and in vivo, which was downregulated after Sunitinib treatment while upregulated in metastasis renal cell carcinoma (RCC) tissues. RESULTS Here, we discovered that treating with Sunitinib, the targeted receptor tyrosine kinase inhibitor (TKI), inhibited renal cancer cell migration and invasion via attenuating the expression of miR-452-5p. The novel identified miR-452-5p was upregulated and associated with poor prognosis in RCC. Preclinical studies using multiple RCC cells and xenografts model illustrated that miR-452-5p could promote RCC cell migration and invasion in vitro and in vivo. Mechanistically, P65 could directly bind to the miR-452-5p promoter and thus transcriptionally induce miR-452-5p expression, which led to post-transcriptionally abrogate SMAD4 expression, thus inhibition of its downstream gene SMAD7. CONCLUSION Our study presented a road map for targeting this newly identified miR-452-5p and its SMAD4/SMAD7 signals pathway, which imparted a new potential therapeutic strategy for mRCC treatment.
Collapse
Affiliation(s)
- Wei Zhai
- Department of Urology, Renji Hospital, School of Medicine in Shanghai Jiao Tong University, 160 Pujian Road, Pudong District, Shanghai, 200127, China.
| | - Saiyang Li
- Department of Urology, Shanghai Tenth People's Hospital, Nanjing Medical University, Nanjing, 211166, China
| | - Jin Zhang
- Department of Urology, Renji Hospital, School of Medicine in Shanghai Jiao Tong University, 160 Pujian Road, Pudong District, Shanghai, 200127, China
| | - Yonghui Chen
- Department of Urology, Renji Hospital, School of Medicine in Shanghai Jiao Tong University, 160 Pujian Road, Pudong District, Shanghai, 200127, China
| | - Junjie Ma
- Department of Urology, Shanghai Tenth People's Hospital, Nanjing Medical University, Nanjing, 211166, China
| | - Wen Kong
- Department of Urology, Renji Hospital, School of Medicine in Shanghai Jiao Tong University, 160 Pujian Road, Pudong District, Shanghai, 200127, China
| | - Dongkui Gong
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine in Tongji University, Shanghai, 200072, China
| | - Junhua Zheng
- Department of Urology, Shanghai First People's Hospital, School of Medicine in Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Wei Xue
- Department of Urology, Renji Hospital, School of Medicine in Shanghai Jiao Tong University, 160 Pujian Road, Pudong District, Shanghai, 200127, China.
| | - Yunfei Xu
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine in Tongji University, Shanghai, 200072, China.
| |
Collapse
|
35
|
Lu XW, Zhang HW, Yin BD, Wu JW. Apoptotic Effect of Arctigenin on Human Renal Cancer Cells by Arresting Cell Cycle and Down regulating P13k/Akt Pathway. INT J PHARMACOL 2018. [DOI: 10.3923/ijp.2018.929.935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
36
|
Abstract
BACKGROUND In patients with isolated meta- or synchronous pulmonary metastases from renal cell cancer, lung metastasectomy could be an appropriate treatment option after successful treatment of primary cancer. OBJECTIVES Presentation of lung metastasectomy as a treatment option in patients with pulmonary metastatic renal cell cancer and the postoperative outcome. Description of alternative treatment modalities focusing on "targeted therapies". MATERIALS AND METHODS Systematical literature research and qualitative analysis of studies on patients undergoing lung metastasectomy after primary nephrectomy published since 01 January 2000. We assessed operative findings, survival data, and prognostic factors. RESULTS Pulmonary metastasectomy results in a median postmetastasectomy survival of 26-94 months. The 5‑year survival rates vary between 33 and 58%. The patients' prognosis depends on a prolonged disease-free interval and complete resection of all suspected metastases. In particular, number and location of lung metastases should play a minor role for the indication for lung metastasectomy. CONCLUSIONS Pulmonary metastasectomy should be considered the treatment of choice in selected patients with successfully resected primary cancer showing no evidence of extrapulmonary metastases and having guaranteed operability and complete resection.
Collapse
|
37
|
Li H, Wang X, Zhang C, Cheng Y, Yu M, Zhao K, Ge W, Cai A, Zhang Y, Han F, Hu Y. HDAC1-induced epigenetic silencing of ASPP2 promotes cell motility, tumour growth and drug resistance in renal cell carcinoma. Cancer Lett 2018; 432:121-131. [DOI: 10.1016/j.canlet.2018.06.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 06/04/2018] [Accepted: 06/06/2018] [Indexed: 12/17/2022]
|
38
|
Xu J, Sun L, Sun W, Tian J, Guo H. Targeted Silencing of Kim-1 Inhibits the Growth of Clear Cell Renal Cell Carcinoma Cell Line 786-0 In Vitro and In Vivo. Oncol Res 2018; 26:997-1003. [PMID: 29295730 PMCID: PMC7844727 DOI: 10.3727/096504017x15140544654946] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
To investigate the effect of Kim-1 on 786-0 cells in vivo and in vitro, several experiments such as quantitative real-time PCR, Western blot, MTT, colony formation, and flow cytometry were performed to evaluate the biological behavior of 786-0 cells treated with Kim-1 siRNA. Furthermore, the tumor xenograft model was applied to BALB/c nude mice to assess the effect of Kim-1 silencing. Lentivirus-mediated RNAi effectively silenced Kim-1 in 786-0 cells. Kim-1 knockdown significantly inhibited the proliferation and colony formation ability of 786-0 cells (p < 0.01). The cell cycle of 786-0 cells was arrested in the G0/G1 phase (p < 0.01). Early and late apoptosis were significantly increased in the Kim-1 siRNA cells (p < 0.01). In addition, growth of 786-0 cells was significantly inhibited in the Kim-1-silenced mice. In conclusion, knockdown of Kim-1 inhibits the growth of 786-0 cells in vitro and in vivo, indicating that Kim-1 could be used as a potential target for clear cell renal cell carcinoma therapy.
Collapse
Affiliation(s)
- Jianping Xu
- Department of Urology, Tumor Hospital of Linyi City, Lanshan, Linyi, Shandong, P.R. China
| | - Liguo Sun
- Department of Urology, Tumor Hospital of Linyi City, Lanshan, Linyi, Shandong, P.R. China
| | - Wei Sun
- Department of Urology, Tumor Hospital of Linyi City, Lanshan, Linyi, Shandong, P.R. China
| | - Jianhai Tian
- Department of Urology, Tumor Hospital of Linyi City, Lanshan, Linyi, Shandong, P.R. China
| | - Huaiyuan Guo
- Department of Urology, Tumor Hospital of Linyi City, Lanshan, Linyi, Shandong, P.R. China
| |
Collapse
|
39
|
Kling SM, Tannouri S, Jiang W, Yeo CJ. Pancreatic Mass in a Patient with a History of Resected Renal Cell Carcinoma and Resected Adenocarcinoma of the Ampulla of Vater: A Case Report. J Pancreat Cancer 2018; 4:41-44. [PMID: 30631857 PMCID: PMC6145538 DOI: 10.1089/pancan.2018.0001] [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] [Indexed: 11/16/2022] Open
Abstract
Background: Metastases of renal cell carcinoma (RCC) to the pancreas are rare, whereas recurrence of pancreatic ductal adenocarcinoma (PDA) or a primary periampullary cancer is far more common. The time elapsed between a primary tumor and a new mass can aid in differentiation between the two. Presentation: A 70-year-old man with a history of RCC status after left nephrectomy and ampullary adenocarcinoma status after pancreaticoduodenectomy presents with an incidentally found mass in his remnant pancreas. Resection of the mass via completion pancreatectomy yielded pathology consistent with metastatic RCC. Conclusions: Metastases of RCC to the pancreas often present many years after a primary resection. Conversely, recurrent PDA often presents within 5 years of resection. Resection of RCC metastases yields better survival than resection of recurrent PDA, which is controversial. We recommend resection of suspected isolated pancreatic RCC metastases due to known favorable outcomes.
Collapse
Affiliation(s)
- Sarah M. Kling
- Department of Surgery, Sidney Kimmel Medical College at Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Sami Tannouri
- Department of Surgery, Sidney Kimmel Medical College at Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Wei Jiang
- Department of Surgery, Sidney Kimmel Medical College at Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Charles J. Yeo
- Department of Surgery, Sidney Kimmel Medical College at Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| |
Collapse
|
40
|
Thompson JA, Motzer RJ, Molina AM, Choueiri TK, Heath EI, Redman BG, Sangha RS, Ernst DS, Pili R, Kim SK, Reyno L, Wiseman A, Trave F, Anand B, Morrison K, Doñate F, Kollmannsberger CK. Phase I Trials of Anti-ENPP3 Antibody-Drug Conjugates in Advanced Refractory Renal Cell Carcinomas. Clin Cancer Res 2018; 24:4399-4406. [PMID: 29848572 DOI: 10.1158/1078-0432.ccr-18-0481] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/19/2018] [Accepted: 05/22/2018] [Indexed: 01/02/2023]
Abstract
Purpose: To determine the safety, pharmacokinetics, and recommended phase II dose of an antibody-drug conjugate (ADC) targeting ectonucleotide phosphodiesterases-pyrophosphatase 3 (ENPP3) conjugated to monomethyl auristatin F (MMAF) in subjects with advanced metastatic renal cell carcinoma (mRCC).Patients and Methods: Two phase I studies were conducted sequentially with 2 ADCs considered equivalent, hybridoma-derived AGS-16M8F and Chinese hamster ovary-derived AGS-16C3F. AGS-16M8F was administered intravenously every 3 weeks at 5 dose levels ranging from 0.6 to 4.8 mg/kg until unacceptable toxicity or progression. The study was terminated before reaching the MTD. A second study with AGS-16C3F started with the AGS-16M8F bridging dose of 4.8 mg/kg given every 3 weeks.Results: The AGS-16M8F study (n = 26) closed before reaching the MTD. The median duration of treatment was 12 weeks (1.7-83 weeks). One subject had durable partial response (PR; 83 weeks) and 1 subject had prolonged stable disease (48 weeks). In the AGS-16C3F study (n = 34), the protocol-defined MTD was 3.6 mg/kg, but this was not tolerated in multiple doses. Reversible keratopathy was dose limiting and required multiple dose deescalations. The 1.8 mg/kg dose was determined to be safe and was associated with clinically relevant signs of antitumor response. Three of 13 subjects at 1.8 mg/kg had durable PRs (range, 100-143 weeks). Eight subjects at 2.7 mg/kg and 1.8 mg/kg had disease control >37 weeks (37.5-141 weeks).Conclusions: AGS-16C3F was tolerated and had durable antitumor activity at 1.8 mg/kg every 3 weeks. Clin Cancer Res; 24(18); 4399-406. ©2018 AACR.
Collapse
Affiliation(s)
- John A Thompson
- Division of Medical Oncology, University of Washington, Seattle, Washington.
| | - Robert J Motzer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ana M Molina
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medical College, New York, New York
| | - Toni K Choueiri
- Harvard Medical School, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Elisabeth I Heath
- Division of Hematology/Oncology, Karmanos Cancer Center, Detroit, Michigan
| | - Bruce G Redman
- Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan
| | | | - D Scott Ernst
- London Health Sciences Centre, London, Ontario, Canada
| | - Roberto Pili
- Department of Oncology, Indiana University, Bloomington, Indiana
| | - Stella K Kim
- Department of Opthalmology and Visual Science, University of Texas McGovern Medical School, Houston, Texas
| | - Leonard Reyno
- Department of Translational Research, Agensys, Inc. Santa Monica, California
| | - Aya Wiseman
- Department of Translational Research, Agensys, Inc. Santa Monica, California
| | | | | | - Karen Morrison
- Department of Translational Research, Agensys, Inc. Santa Monica, California
| | - Fernando Doñate
- Department of Translational Research, Agensys, Inc. Santa Monica, California
| | | |
Collapse
|
41
|
Zhou KW, Jiang K, Zhu W, Weng G. Expression of cold-inducible RNA-binding protein (CIRP) in renal cell carcinoma and the effect of CIRP downregulation cell proliferation and chemosensitivity to gemcitabine. Oncol Lett 2018; 15:7611-7616. [PMID: 29849797 PMCID: PMC5962864 DOI: 10.3892/ol.2018.8338] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/21/2018] [Indexed: 01/16/2023] Open
Abstract
The aim of the present study was to investigate the expression of cold-inducible RNA-binding protein (CIRP) in renal cell carcinoma (RCC) and to determine the effects of downregulation of CIRP on cell proliferation and chemosensitivity to gemcitabine. The expression of CIRP was detected by western blot analysis, quantitative polymerase chain reaction and immunohistochemistry (IHC) in 17 RCC and peri-cancerous tissue samples. Subsequently, the RCC 786-0 cell line was selected in order to investigate the function of CIRP using RNA interference (RNAi) technology, which was able to inhibit the expression of CIRP in vitro. Furthermore, the chemosensitivity to gemcitabine of each group [CIRP small interfering RNA (siCIRP), negative control small interfering RNA (siNC) and blank control] was compared. There were marked differences between the RCC and peri-cancerous tissues. IHC demonstrated that the CIRP expression in 13/17 (76.50%) tumor samples was markedly positive compared with that in the peri-cancerous tissues and the most common pathological type was clear cell RCC (92.30%). This observation was further confirmed through western blot analysis of protein expression levels. CIRP downregulation by RNAi in the RCC 786-0 cell line significantly decreased RCC proliferation. Additionally, when RNAi was coupled with gemcitabine treatment, there was a significant increase in apoptosis in the siCIRP group. CIRP was overexpressed in RCC tissues and in the 786-0 cell line. Downregulation of CIRP by siRNA inhibited the proliferation of the 786-0 cell line and enhanced the chemosensitivity of the cells to gemcitabine. Therefore, CIRP downregulation may provide a novel pathway for the treatment of metastatic RCC.
Collapse
Affiliation(s)
- Ke-Wen Zhou
- Department of Urology, Ningbo Urology and Kidney Hospital, Ningbo, Zhejiang 315100, P.R. China
| | - Kun Jiang
- Department of Urology, Ren Min Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Weizhi Zhu
- Department of Urology, Ningbo Urology and Kidney Hospital, Ningbo, Zhejiang 315100, P.R. China
| | - Guobin Weng
- Department of Urology, Ningbo Urology and Kidney Hospital, Ningbo, Zhejiang 315100, P.R. China
| |
Collapse
|
42
|
Tang J, Wang F, Cheng G, Si S, Sun X, Han J, Yu H, Zhang W, Lv Q, Wei JF, Yang H. Wilms' tumor 1-associating protein promotes renal cell carcinoma proliferation by regulating CDK2 mRNA stability. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:40. [PMID: 29482572 PMCID: PMC5827993 DOI: 10.1186/s13046-018-0706-6] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 02/12/2018] [Indexed: 12/30/2022]
Abstract
Background Wilms’ tumor 1-associating protein (WTAP) plays an important role in physiological processes and the development of tumor such as cell cycle regulation. The regulation of cell cycle is mainly dependent on cyclins and cyclin-dependent protein kinases (CDKs). Recent studies have shown that CDKs are closely related to the tumor diagnosis, progression and response to treatment. However, their specific biological roles and related mechanism in renal cell carcinoma (RCC) remain unknown. Methods Quantitative real-time PCR, western blotting and immunohistochemistry were used to detect the expression of WTAP and CDK2. The survival analysis was adopted to explore the association between WTAP expression and the prognosis of RCC. Cells were stably transfected with lentivirus approach and cell proliferation and cell cycle, as well as tumorigenesis in nude mice were performed to assess the effect of WTAP in RCC. RNA immunoprecipitation, Luciferase reporter assay and siRNA were employed to identify the direct binding sites of WTAP with CDK2 transcript. Colony formation assay was conducted to confirm the function of CDK2 in WTAP-induced growth promoting. Results In RCC cell lines and tissues, WTAP was significantly over-expressed. Compared with patients with low expression of WTAP, patients with high expression of WTAP had lower overall survival rate. Additionally, cell function test indicated that cell proliferation abilities in WTAP over-expressed group were enhanced, while WTAP knockdown showed the opposite results. Subcutaneous xenograft tumor model displayed that knockdown of WTAP could impede tumorigenesis in vivo. Mechanism study exhibited that CDK2 expression was positively associated with the expression of WTAP. Moreover, WTAP stabilized CDK2 transcript to enhance CDK2 expression via binding to 3′-UTR of CDK2 transcript. Additionally, specific inhibitors of CDK2 activity and small interfering RNA (siRNA) of CDK2 expression inhibited WTAP-mediated promotion of proliferation. Conclusions These findings suggest that WTAP may have an oncogenic role in RCC through physically binding to CDK2 transcript and enhancing its transcript stability which might provide new insights into RCC therapy. Electronic supplementary material The online version of this article (10.1186/s13046-018-0706-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jingyuan Tang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.,Department of Urology, Jiangsu Province Hospital of TCM, Affiliated Hospital of Nanjing University of TCM, Nanjing, 210029, China
| | - Feng Wang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Gong Cheng
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Shuhui Si
- Research Division of Clinical Pharmacology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xi Sun
- Jiangsu Breast Disease Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Jie Han
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Hao Yu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Wei Zhang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Qiang Lv
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Ji-Fu Wei
- Research Division of Clinical Pharmacology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Haiwei Yang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
43
|
Bilen MA, Carlisle JW, Sonpavde G. The prospects for combination therapy with capecitabine in the rapidly evolving treatment landscape of renal cell carcinoma. Expert Opin Investig Drugs 2018; 27:163-170. [PMID: 29323560 DOI: 10.1080/13543784.2018.1427731] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Although significant advances have been made in the treatment of advanced renal cell carcinoma (RCC), patients still develop resistance to standard therapies and require the administration of subsequent lines of treatment. New therapeutic approaches are thus imperative to improve the prognosis for patients with RCC. AREAS COVERED Based on the current literature, we summarize the treatment of metastatic RCC, including the use of cytotoxic chemotherapy, in this review article. We also review the existing scientific literature regarding the role of capecitabine in the treatment of RCC. EXPERT OPINION Currently, targeted therapies including vascular endothelial growth factor (VEGF) and mammalian target of rapamycin (mTOR) inhibitors are widely used in the treatment of metastatic RCC. More recently, the role of immune checkpoint inhibitors has been established in the treatment of advanced RCC. Traditionally, the use of cytotoxic chemotherapy in the treatment of RCC is limited. However, cytotoxic chemotherapy may have benefit in different types of RCC, such as variant histology. Furthermore, new combinations of chemotherapy with immune checkpoint inhibitors may provide new treatment options for our patients.
Collapse
Affiliation(s)
- Mehmet Asim Bilen
- a Department of Hematology and Medical Oncology , Winship Cancer Institute of Emory University , Atlanta , GA , USA
| | - Jennifer W Carlisle
- a Department of Hematology and Medical Oncology , Winship Cancer Institute of Emory University , Atlanta , GA , USA
| | - Guru Sonpavde
- b Department of Medicine, Division of Hematology and Oncology , University of Alabama at Birmingham Comprehensive Cancer Center , Birmingham , AL , USA
| |
Collapse
|
44
|
Ge W, Zhao K, Wang X, Li H, Yu M, He M, Xue X, Zhu Y, Zhang C, Cheng Y, Jiang S, Hu Y. iASPP Is an Antioxidative Factor and Drives Cancer Growth and Drug Resistance by Competing with Nrf2 for Keap1 Binding. Cancer Cell 2017; 32:561-573.e6. [PMID: 29033244 DOI: 10.1016/j.ccell.2017.09.008] [Citation(s) in RCA: 136] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 06/15/2017] [Accepted: 09/15/2017] [Indexed: 12/14/2022]
Abstract
Reactive oxygen species (ROS) have emerged as important signaling molecules that play crucial roles in carcinogenesis and cytotoxic responses. Nrf2 is the master regulator of ROS balance. Thus, uncovering mechanisms of Nrf2 regulation is important for the development of alternative treatment strategies for cancers. Here, we demonstrate that iASPP, a known p53 inhibitor, lowers ROS independently of p53. Mechanistically, iASPP competes with Nrf2 for Keap1 binding via a DLT motif, leading to decreased Nrf2 ubiquitination and increased Nrf2 accumulation, nuclear translocation, and antioxidative transactivation. This iASPP-Keap1-Nrf2 axis promotes cancer growth and drug resistance both in vitro and in vivo. Thus, iASPP is an antioxidative factor and represents a promising target to improve cancer treatment, regardless of p53 status.
Collapse
Affiliation(s)
- Wenjie Ge
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province 150001, China
| | - Kunming Zhao
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province 150001, China
| | - Xingwen Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province 150001, China
| | - Huayi Li
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province 150001, China
| | - Miao Yu
- School of Chemistry, Harbin Institute of Technology, Harbin, Heilongjiang Province 150001, China
| | - Mengmeng He
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province 150001, China
| | - Xuting Xue
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province 150001, China
| | - Yifu Zhu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province 150001, China
| | - Cheng Zhang
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150006, China
| | - Yiwei Cheng
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150006, China
| | - Shijian Jiang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province 150001, China
| | - Ying Hu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province 150001, China; Shenzhen Graduate School of Harbin Institute of Technology, Shenzhen 518055, China.
| |
Collapse
|
45
|
Li X, Tang J, Huang W, Wang F, Li P, Qin C, Qin Z, Zou Q, Wei J, Hua L, Yang H, Wang Z. The M6A methyltransferase METTL3: acting as a tumor suppressor in renal cell carcinoma. Oncotarget 2017; 8:96103-96116. [PMID: 29221190 PMCID: PMC5707084 DOI: 10.18632/oncotarget.21726] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 07/25/2017] [Indexed: 12/30/2022] Open
Abstract
We aimed to study the role of METTL3 in renal cell carcinoma (RCC) carcinogenesis and development. Immunohistochemistry was performed in clinical tissue microarray. Expression level of METTL3 in RCC tissues and cell lines was evaluated by quantitative real-time PCR (qRT-PCR) and western blot. Then, the effects of METTL3 on proliferation, migration, invasion and cell cycle were studied in RCC cells. Additionally, in vivo study was carried out in nude mice. Negative METTL3 expression was associated with larger tumor size (P=0.010) and higher histological grade (P=0.021). Moreover, RCC patients with positive METTL3 expression had an obvious longer survival time (P=0.039). METTL3 mRNA and protein expression was lower in RCC samples compared with adjacent non-tumor samples, and lower in RCC cell lines (CAKI-1, CAKI-2 and ACHN) compared with HK-2. Afterwards, knockdown of METTL3 could obviously promote cell proliferation, migration and invasion function, and induce G0/G1 arrest. In contrast, up-regulation of METTL3 could inhibit such functions and reduce G0/G1 arrest. Additionally, up-regulation of METTL3 significantly suppressed tumor growth in vivo. Furthermore, significant changes in epithelial-to-mesenchymal transition (EMT) and PI3K-Akt-mTOR pathways were observed. Overall, our findings demonstrated that METTL3 might have a carcinostasis role in cell proliferation, migration, invasion function and cell cycle of RCC, indicating METTL3 may act as a novel marker for tumorigenesis, development and survival of RCC.
Collapse
Affiliation(s)
- Xiao Li
- State Key Laboratory of Reproductive Medicine, Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.,Department of Urology, Affiliated Cancer Hospital of Jiangsu Province of Nanjing Medical University, Nanjing 210009, China
| | - Jingyuan Tang
- State Key Laboratory of Reproductive Medicine, Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.,Department of Urology, Jiangsu Province Hospital of TCM, Affiliated Hospital of Nanjing University of TCM, Nanjing 210029,China
| | - Wen Huang
- Research Division of Clinical Pharmacology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Feng Wang
- State Key Laboratory of Reproductive Medicine, Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Pu Li
- State Key Laboratory of Reproductive Medicine, Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chao Qin
- State Key Laboratory of Reproductive Medicine, Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zhiqiang Qin
- State Key Laboratory of Reproductive Medicine, Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Qing Zou
- Department of Urology, Affiliated Cancer Hospital of Jiangsu Province of Nanjing Medical University, Nanjing 210009, China
| | - Jifu Wei
- Research Division of Clinical Pharmacology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Lixin Hua
- State Key Laboratory of Reproductive Medicine, Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Haiwei Yang
- State Key Laboratory of Reproductive Medicine, Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zengjun Wang
- State Key Laboratory of Reproductive Medicine, Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
46
|
Majuta LA, Guedon JMG, Mitchell SA, Kuskowski MA, Mantyh PW. Mice with cancer-induced bone pain show a marked decline in day/night activity. Pain Rep 2017; 2:e614. [PMID: 29392229 PMCID: PMC5777677 DOI: 10.1097/pr9.0000000000000614] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 06/15/2017] [Accepted: 06/17/2017] [Indexed: 01/01/2023] Open
Abstract
INTRODUCTION Cancer-induced bone pain (CIBP) is the most common type of pain with cancer. In humans, this pain can be difficult to control and highly disabling. A major problem with CIBP in humans is that it increases on weight-bearing and/or movement of a tumor-bearing bone limiting the activity and functional status of the patient. Currently, there is less data concerning whether similar negative changes in activity occur in rodent models of CIBP. OBJECTIVES To determine whether there are marked changes in activity in a rodent model of CIBP and compare this to changes in skin hypersensitivity. METHODS Osteosarcoma cells were injected and confined to 1 femur of the adult male mouse. Every 7 days, spontaneous horizontal and vertical activities were assessed over a 20-hour day and night period using automated activity boxes. Mechanical hypersensitivity of the hind paw skin was assessed using von Frey testing. RESULTS As the tumor cells grew within the femur, there was a significant decline in horizontal and vertical activity during the times of the day/night when the mice are normally most active. Mice also developed significant hypersensitivity in the skin of the hind paw in the tumor-bearing limb. CONCLUSION Even when the tumor is confined to a single load-bearing bone, CIBP drives a significant loss of activity, which increases with disease progression. Understanding the mechanisms that drive this reduction in activity may allow the development of therapies that allow CIBP patients to better maintain their activity and functional status.
Collapse
Affiliation(s)
- Lisa A. Majuta
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | | | | | | | - Patrick W. Mantyh
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
- Cancer Center, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
47
|
Wang X, Cheng Y, Zhu Y, Li H, Ge W, Wu X, Zhao K, Yuan J, Li Z, Jiang S, Han Z, Jiang Q, Wu Q, Liu T, Zhang C, Yu M, Hu Y. Epigenetic silencing of ASPP1 confers 5‐FU resistance in clear cell renal cell carcinoma by preventing p53 activation. Int J Cancer 2017; 141:1422-1433. [DOI: 10.1002/ijc.30852] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 05/07/2017] [Accepted: 06/21/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Xingwen Wang
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
- Shenzhen Graduate School of Harbin Institute of TechnologyXili University CityNanshanShenzhen Guangdong518055 China
| | - Yiwei Cheng
- The First Affiliated HospitalHarbin Medical UniversityHarbin Heilongjiang150081 China
| | - YiFu Zhu
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
| | - Huayi Li
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
| | - Wenjie Ge
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
- Shenzhen Graduate School of Harbin Institute of TechnologyXili University CityNanshanShenzhen Guangdong518055 China
| | - Xiaoliang Wu
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
| | - Kunming Zhao
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
| | - Jinyang Yuan
- The First Affiliated HospitalHarbin Medical UniversityHarbin Heilongjiang150081 China
| | - Zhenglin Li
- School of Chemical Engineering and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
| | - Shijian Jiang
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
| | - Zhengbin Han
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
| | - Qinghua Jiang
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
| | - Qiong Wu
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
| | - Tao Liu
- Shenzhen Luohu People's Hospital, Shenzhen Zhongxun Precision Medicine Research InstituteShenzhen Guangdong518001 China
| | - Cheng Zhang
- The First Affiliated HospitalHarbin Medical UniversityHarbin Heilongjiang150081 China
| | - Miao Yu
- School of Chemical Engineering and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
| | - Ying Hu
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
- Shenzhen Graduate School of Harbin Institute of TechnologyXili University CityNanshanShenzhen Guangdong518055 China
| |
Collapse
|
48
|
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
|
49
|
Zarrabi K, Fang C, Wu S. New treatment options for metastatic renal cell carcinoma with prior anti-angiogenesis therapy. J Hematol Oncol 2017; 10:38. [PMID: 28153029 PMCID: PMC5288948 DOI: 10.1186/s13045-016-0374-y] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 12/08/2016] [Indexed: 01/05/2023] Open
Abstract
Angiogenesis is a critical process in the progression of advanced renal cell carcinoma. Agents targeting angiogenesis have played a primary role in the treatment of metastatic renal cell carcinoma. However, resistance to anti-angiogenesis therapy almost always occurs, and major progress has been made in understanding its underlying molecular mechanism. Axitinib and everolimus have been used extensively in patients whom have had disease progression after prior anti-angiogenesis therapy. Recently, several new agents have been shown to improve overall survival in comparison with everolimus. This review provides an in-depth summary of drugs employable in the clinical setting, the rationale to their use, and the studies conducted leading to their approval for use and provides perspective on the paradigm shift in the treatment of renal cell carcinoma. Highlighted are the newly approved agents cabozantinib, nivolumab, and lenvatinib for advanced renal cell carcinoma patients treated with prior anti-angiogenesis therapy.
Collapse
Affiliation(s)
- Kevin Zarrabi
- Department of Medicine, Stony Brook University Hospital, 9447 Suny, Stony Brook, NY 11794-9447 USA
| | - Chunhui Fang
- Department of Medicine, Stony Brook University Hospital, 9447 Suny, Stony Brook, NY 11794-9447 USA
| | - Shenhong Wu
- Department of Medicine, Stony Brook University Hospital, 9447 Suny, Stony Brook, NY 11794-9447 USA
- Division of Hematology/Oncology, Department of Medicine, Northport VA Medical Center, Northport, NY USA
| |
Collapse
|
50
|
Massari F, Ciccarese C, Bria E, Porta C, La Russa F, Knuutila S, Artibani W, Porcaro AB, Bimbatti D, Modena A, Sava T, Tortora G, Cheng L, Eccher A, Cima L, Pedron S, Ghimenton C, Martignoni G, Brunelli M. Reprofiling Metastatic Samples for Chromosome 9p and 14q Aberrations as a Strategy to Overcome Tumor Heterogeneity in Clear-cell Renal Cell Carcinoma. Appl Immunohistochem Mol Morphol 2017; 25:39-43. [PMID: 26509904 DOI: 10.1097/pai.0000000000000257] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Losses of chromosomes 9p and 14q are associated with worse outcomes in patients affected by clear-cell renal cell carcinoma (RCC) and are helpful for prognostic risk stratification. Both chromosomal loci harbor several hot-spot molecular pathways suitable for targeted therapeutic interventions. Intratumor heterogeneity may foster tumor adaptation and therapeutic failure. We sought to investigate the presence of losses of the hot spots of chromosomal loci 9p and 14q in primary clear-cell RCC and matched metastatic tissues. CD10 and CD13 were performed on 7 cases of clear-cell RCC with hematogenous tissue metastases. Cytogenetic fluorescence in situ hybridization analysis was performed on primary and matched metastatic tissues using specific probes mapping the 9p and the 14q loci. The loss of chromosome 9p was observed in 85% of both primary clear-cell RCCs and in matched metastases; 14% showed discordance between primary and matched metastases showing gains. The loss of chromosome 14q was observed in 58% of both primary and matched metastases. Only 3/7 (42%) did show an equal status of loss of chromosome 14q. Heterogeneity of the cytogenetic status between metastatic and primary clear-cell RCCs is observed for the loss of chromosome 14q rather than chromosome 9p. The impact of chromosome 14q cytogenetic status, harboring the HIF1 gene, a major driver for the angiogenenic switch, may drive the efficacy of targeted inhibitors, whereas the loss of chromosome 9p, harboring other hot-spot genes, seems to be related to the metastatic behavior per se, without cytogenetic modulation. Reprofiling the metastatic tissue, as compared with the primary tumor, in patients affected by metastatic RCC could be a novel approach to overcome resistance to VEGF(Rs)-targeting agents.
Collapse
Affiliation(s)
- Francesco Massari
- *Department of Medical Oncology §Urologic Clinic ¶Department of Pathology and Diagnostic, University and Hospital Trust, Verona †Medical Oncology, I.R.C.C.S. San Matteo University Hospital Foundation, Pavia, Italy ‡Department of Pathology, Laboratory of Molecular Cytogenetic, University of Helsinki, Helsinki, Finland ∥Department of Pathology and Laboratory Medicine, Indiana University, Bloomington, IN
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|