51
|
Huang JJ, Hsieh JJ. The Therapeutic Landscape of Renal Cell Carcinoma: From the Dark Age to the Golden Age. Semin Nephrol 2021; 40:28-41. [PMID: 32130964 DOI: 10.1016/j.semnephrol.2019.12.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Oncologic treatments for renal cell carcinoma (RCC) have undergone a major revolution in the past 2 decades, moving away from the pre-2004 Dark Age during which interleukin 2 and interferon-α were the only therapeutic options and induced treatment responses in only 5% to 10% of patients with metastatic disease. The development of anti-angiogenic tyrosine kinase inhibitors against vascular endothelial growth factor receptor 2 and inhibitors of mammalian target of rapamycin complex 1 in 2005 introduced the Modern Age with better overall and progression-free survival and a greater number of patients (30%-40%) responding to and (∼80%) benefiting from these targeted therapeutic agents. The coming of age of the immuno-oncology era with the use of immune checkpoint inhibitors (ICIs) have ushered us into the Golden Age of metastatic RCC care, in which combined administrations of two ICIs (anti-programmed cell death protein 1/programmed death-ligand 1 and anti-cytotoxic T-lymphocyte-associated protein 4 or one tyrosine kinase inhibitor plus one ICI (anti-programmed cell death protein 1/programmed death-ligand 1) have recast the treatment landscape of clear cell RCC, the most common RCC subtype, with an approximately 60% response rate and an approximately 90% disease control rate that further improves metastatic RCC survival. Exciting clinical trials are in the pipeline investigating complementary/synergistic molecular mechanisms, based on studies investigating the biology, pathology, and genomics of renal carcinoma and the respective treatment outcome. This will enable us to enter the Diamond Age of precision medicine in which a specific treatment can be tailored to the specific biological and pathologic circumstance of an individual kidney tumor to offer more effective yet less toxic therapy.
Collapse
Affiliation(s)
- Jennifer J Huang
- Molecular Oncology, Department of Medicine, Siteman Cancer Center, Washington University, St. Louis, MO
| | - James J Hsieh
- Molecular Oncology, Department of Medicine, Siteman Cancer Center, Washington University, St. Louis, MO.
| |
Collapse
|
52
|
Liu K, Gao R, Wu H, Wang Z, Han G. Single-cell analysis reveals metastatic cell heterogeneity in clear cell renal cell carcinoma. J Cell Mol Med 2021; 25:4260-4274. [PMID: 33759378 PMCID: PMC8093989 DOI: 10.1111/jcmm.16479] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 02/20/2021] [Accepted: 02/25/2021] [Indexed: 12/12/2022] Open
Abstract
Renal cell carcinoma (RCC) is one of the leading causes of cancer-related death worldwide. Tumour metastasis and heterogeneity lead to poor survival outcomes and drug resistance in patients with metastatic RCC (mRCC). In this study, we aimed to assess intratumoural heterogeneity (ITH) in mRCC cells by performing a combined analysis of bulk data and single-cell RNA-sequencing data, and develop novel biomarkers for prognosis prediction on the basis of the potential molecular mechanisms underlying tumorigenesis. Eligible single-cell cohorts related to mRCC were acquired using the Gene Expression Omnibus (GEO) dataset to identify potential mRCC subpopulations. We then performed gene set variation analysis to understand the differential function in primary RCC and mRCC samples. Subsequently, we applied weighted correlation network analysis to identify coexpressing gene modules that were related to the external trait of metastasis. Protein-protein interactions were used to screen hub subpopulation-difference (sub-dif) markers (ACTG1, IL6, CASP3, ACTB and RAP1B) that might be involved in the regulation of RCC metastasis and progression. Cox regression analysis revealed that ACTG1 was a protective factor (HR < 1), whereas the other four genes (IL6, CASP3, ACTB and RAP1B) were risk factors (HR > 1). Kaplan-Meier survival analysis suggested the potential prognostic value of these sub-dif markers. The expression of sub-dif markers in mRCC was further evaluated in clinical samples by immunohistochemistry (IHC). Additionally, the genetic features of sub-dif marker expression patterns, such as genetic variation profiles, correlations with tumour-infiltrating lymphocytes (TILs), and targeted signalling pathway activities, were assessed in bulk RNA-seq datasets. In conclusion, we established novel subpopulation markers as key prognostic factors affecting EMT-related signalling pathway activation in mRCC, which could facilitate the implementation of a treatment for mRCC patients.
Collapse
Affiliation(s)
- Kun Liu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Rui Gao
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hao Wu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhe Wang
- Department of Gastrointestinal Oncology, Cancer Hospital of China Medical University Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Guang Han
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
53
|
Lv D, Wu X, Wang M, Chen W, Yang S, Liu Y, Zeng G, Gu D. Functional Assessment of Four Novel Immune-Related Biomarkers in the Pathogenesis of Clear Cell Renal Cell Carcinoma. Front Cell Dev Biol 2021; 9:621618. [PMID: 33796525 PMCID: PMC8007883 DOI: 10.3389/fcell.2021.621618] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 02/26/2021] [Indexed: 12/24/2022] Open
Abstract
Background Clear cell renal cell carcinoma (ccRCC) is the most common subtype of renal cell carcinoma whose pathogenesis is not well understood. We aimed at identifying novel immune-related biomarkers that could be valuable in the diagnosis and prognosis of ccRCC. Methods The Robust Rank Aggregation (RRA) method was used to integrate differently expressed genes (DEGs) of 7 Gene Expression Omnibus (GEO) datasets and obtain robust DEGs. Weighted gene co-expression network analyses (WGCNA) were performed to identify hub genes associated with clinical traits in The Cancer Genome Atlas (TCGA) database. Comprehensive bioinformatic analyses were used to explore the role of hub genes in ccRCC. Results Four hub genes IFI16, LMNB1, RHBDF2 and TACC3 were screened by the RRA method and WGCNA. These genes were found to be up-regulated in ccRCC, an upregulation that could be due to their associations with late TNM stages and tumor grades. The Receiver Operating Characteristic (ROC) curve and Kaplan-Meier survival analysis showed that the four hub genes had great diagnostic and prognostic values for ccRCC, while Gene Set Enrichment Analysis (GSEA) showed that they were involved in immune signaling pathways. They were also found to be closely associated with multiple tumor-infiltrating lymphocytes and critical immune checkpoint expressions. The results of Quantitative Real-time PCR (qRT-PCR) and immunohistochemical staining (IHC) analysis were consistent with bioinformatics analysis results. Conclusion The four hub genes were shown to have great diagnostic and prognostic values and played key roles in the tumor microenvironment of ccRCC.
Collapse
Affiliation(s)
- Daojun Lv
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangdong Key Laboratory of Urology, Guangzhou Institute of Urology, Guangzhou, China
| | - Xiangkun Wu
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangdong Key Laboratory of Urology, Guangzhou Institute of Urology, Guangzhou, China
| | - Ming Wang
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangdong Key Laboratory of Urology, Guangzhou Institute of Urology, Guangzhou, China
| | - Wenzhe Chen
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangdong Key Laboratory of Urology, Guangzhou Institute of Urology, Guangzhou, China
| | - Shuxin Yang
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangdong Key Laboratory of Urology, Guangzhou Institute of Urology, Guangzhou, China
| | - Yongda Liu
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangdong Key Laboratory of Urology, Guangzhou Institute of Urology, Guangzhou, China
| | - Guohua Zeng
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangdong Key Laboratory of Urology, Guangzhou Institute of Urology, Guangzhou, China
| | - Di Gu
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangdong Key Laboratory of Urology, Guangzhou Institute of Urology, Guangzhou, China
| |
Collapse
|
54
|
Sun F, Chen Z, Yao P, Weng B, Liu Z, Cheng L. Meta-Analysis of ABCG2 and ABCB1 Polymorphisms With Sunitinib-Induced Toxicity and Efficacy in Renal Cell Carcinoma. Front Pharmacol 2021; 12:641075. [PMID: 33762959 PMCID: PMC7982400 DOI: 10.3389/fphar.2021.641075] [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: 12/13/2020] [Accepted: 01/19/2021] [Indexed: 11/13/2022] Open
Abstract
Background: ABCG2 and ABCB1 are genes related to the pharmacokinetics of sunitinib and have been associated with its toxicity and efficacy. However, the results have been controversial. This study aimed to evaluate the associations of ABCG2 and ABCB1 polymorphisms with sunitinib-induced toxicity and efficacy in renal cell carcinoma (RCC) by meta-analysis. Methods: PubMed, EMBASE, Cochrane Library, and Web of Science were systematically searched for studies investigating the associations of the ABCG2 rs2231142 polymorphism with sunitinib-induced toxicity and the associations of the ABCB1 rs1128503 and ABCB1 rs2032582 polymorphisms with sunitinib-induced toxicity and clinical outcomes. The associations were evaluated by effect size (ES) with 95% confidence intervals (CIs). Results: Eight and five studies were included in the toxicity and efficacy analysis, respectively, including a total of 1081 RCC patients. The ABCG2 rs2231142 A allele was associated with an increased risk of sunitinib-induced thrombocytopenia and hand-foot syndrome (HFS) in Asians (ES = 1.65, 95% CI = 1.15-2.36, p = 0.006; ES = 1.52, 95% CI = 1.02-2.27, p = 0.041). However, the ABCG2 rs2231142 polymorphism was not associated with sunitinib-induced hypertension or neutropenia (ES = 1.09, 95% CI = 0.69-1.73, p = 0.701; ES = 0.87, 95% CI = 0.57-1.31, p = 0.501). Compared with the C allele, the ABCB1 rs1128503 T allele was associated with a decreased risk of sunitinib-induced hypertension but worse progression-free survival (PFS) (ES = 0.44, 95% CI = 0.26-0.77, p = 0.004; ES = 1.36, 95% CI = 1.07-1.73, p = 0.011). There was no significant association between the T allele or C allele of ABCB1 rs1128503 and overall survival (OS) (ES = 0.82, 95% CI = 0.61-1.10, p = 0.184). The ABCB1 rs2032582 T allele was associated with worse PFS than the other alleles (ES = 1.46, 95% CI = 1.14-1.87, p = 0.003), while there was no significant association between the T allele or other alleles and sunitinib-induced hypertension, HFS, or OS (ES = 0.77, 95% CI = 0.46-1.29, p = 0.326; ES = 1.02, 95% CI = 0.65-1.62, p = 0.919; ES = 1.32, 95% CI = 0.85-2.05, p = 0.215). Conclusion: The results indicate that the ABCG2 rs2231142 polymorphism may serve as a predictor of sunitinib-induced thrombocytopenia and HFS in Asians, while the ABCB1 rs1128503 polymorphism may serve as a predictor of sunitinib-induced hypertension, and both the ABCB1 rs1128503 and rs2032582 polymorphisms may serve as predictors of PFS in RCC. These results suggest a possible application of individualized use of sunitinib according to the genetic background of patients.
Collapse
Affiliation(s)
- Fengjun Sun
- Department of Pharmacy, The First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing, China
| | - Zhuo Chen
- Department of Pharmacy, Chongqing Emergency Medical Center, Chongqing, China
| | - Pu Yao
- Department of Pharmacy, The First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing, China
| | - Bangbi Weng
- Department of Pharmacy, The First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing, China
| | - Zhirui Liu
- Department of Pharmacy, The First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing, China
| | - Lin Cheng
- Department of Pharmacy, The First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
55
|
Gkolfinopoulos S, Psyrri A, Bamias A. Clear-cell renal cell carcinoma - A comprehensive review of agents used in the contemporary management of advanced/metastatic disease. Oncol Rev 2021; 15:530. [PMID: 33747368 PMCID: PMC7967495 DOI: 10.4081/oncol.2021.530] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 12/28/2020] [Indexed: 01/05/2023] Open
Abstract
Renal cell carcinoma represents the most common malignancy of the kidney and the majority of cases are categorized as clear cell carcinomas. The elucidation of the specific alterations in key molecular and metabolic pathways responsible for cancer development and progression have prompted the rationalization of our classification of this disease and have provided specific targetable molecules implicated in carcinogenesis. Although immunotherapy has been an established option in the treatment of metastatic renal cell cancer for many years, its role has been renewed and upgraded with the implementation of anti-angiogenic agents and immune checkpoint inhibitors in our treatment armamentarium. The future holds promise, as newer agents become available and combination regimens of immunotherapy with anti-angiogenic agents have become the standard of care in the management of metastatic disease and are currently being evaluated in earlier settings. Proper patient selection and individualization of our treatment strategies are of utmost importance in order to provide optimal care to patients suffering from renal cell carcinoma.
Collapse
Affiliation(s)
- Stavros Gkolfinopoulos
- 2 Propaedeutic Dept. of Internal Medicine, National & Kapodistrian University of Athens, ATTIKON University Hospital, Athens, Greece
| | - Amanda Psyrri
- 2 Propaedeutic Dept. of Internal Medicine, National & Kapodistrian University of Athens, ATTIKON University Hospital, Athens, Greece
| | - Aristotelis Bamias
- 2 Propaedeutic Dept. of Internal Medicine, National & Kapodistrian University of Athens, ATTIKON University Hospital, Athens, Greece
| |
Collapse
|
56
|
Tolerability and outcome of sunitinib by giving 4/2 schedule versus 2/1 schedule in metastatic renal cell carcinoma patients: a prospective randomized multi-centric Egyptian study. Contemp Oncol (Pozn) 2021; 24:221-228. [PMID: 33531869 PMCID: PMC7836283 DOI: 10.5114/wo.2020.102802] [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: 08/29/2020] [Accepted: 11/02/2020] [Indexed: 11/17/2022] Open
Abstract
Introduction Sunitinib is a standard of care first line treatment for patients with metastatic renal cell carcinoma (RCC). Sunitinib standard dose is 50 mg once daily for 4 consecutive weeks followed by 2 weeks' off (4/2 schedule). Long-term and high exposure to this medication lead to severe adverse events (AEs); therefore, this trial was done to find the best schedule which gives the best outcome with minimal toxicity. Materials and methods Seventy patients were randomly assigned into 2 groups, then received 50 mg/day of sunitinib. Group 1 (40 patients) received sunitinib for 4 consecutive weeks followed by 2 weeks off (4/2 schedule) while 30 patients were admitted to group 2 with 2 weeks on and 1 week off (2/1 schedule). Results All patients (100%) had significantly higher AEs on schedule 4/2 vs. 73.3% on schedule 2/1 (p = 0.001). Furthermore, the grade 3 AEs on schedule 2/1 were significantly lower than those on schedule 4/2 (26.7% vs. 82.5%) respectively (p = 0.001), such as fatigue, diarrhea, hypertension, hand foot syndrome (HFS) and mucositis. Progression-free survival (PFS) rate was significantly higher in 2/1 schedule (60.9% vs. 38.6%) than in 4/2 schedule (p < 0.008). Multivariate analysis suggested that: age > 60 years, poor International Metastatic RCC Database Consortium (IMDC) risk category, tumor size > 10 cm and treatment schedule (group 1) were poor prognostic factors of PFS. Conclusions Our study supported the use of 2/1 schedule of sunitinib in patients with metastatic RCC because of lower toxicity profile and better efficacy with improved PFS in comparison to 4/2 schedule.
Collapse
|
57
|
Byrnes KG, Khan JSA, Haroon UM, McCawley N, Cheema IA. Management of colon-invading renal cell carcinoma: Operative technique and systematic review. Urol Ann 2021; 13:1-8. [PMID: 33897156 PMCID: PMC8052896 DOI: 10.4103/ua.ua_86_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/12/2020] [Indexed: 01/04/2023] Open
Abstract
Invasion into adjacent organs by non-metastatic renal cell carcinoma (RCC) occurs in 1% of patients suitable for resection. Colonic invasion is rare and presents technical challenges. No prospective data exists to guide management of these patients. We present the first reported case of a colon-invading RCC managed with simultaneous open right radical nephrectomy and extended right hemicolectomy. PubMed, Scopus and EMBASE databases were searched for relevant case reports reporting management of colon-invading renal cell carcinoma. Case reports, case series and cohort studies were eligible. A chart review was performed on a patient who presented with right-sided colon-invading RCC. Four previously reported cases were identified. The current case was managed with simultaneous open radical nephrectomy and extended right hemicolectomy. The patient remains well six months postoperatively with no evidence of disease recurrence. Histopathological evaluation of the resected specimen confirmed a T4 clear cell RCC with sarcomatoid differentiation. Colon-invading RCC is rare. This is the first reported case of right-sided, colon-invading RCC treated with radical resection. The current case confirms radical resection is a feasible management strategy for similar presentations. En bloc resection of involved organs remains the only potentially curative option for locally advanced disease.
Collapse
Affiliation(s)
| | | | | | - Niamh McCawley
- Department of Colorectal Surgery, Beaumont Hospital, Dublin, Ireland
| | | |
Collapse
|
58
|
Vedenko A, Panara K, Goldstein G, Ramasamy R, Arora H. Tumor Microenvironment and Nitric Oxide: Concepts and Mechanisms. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1277:143-158. [PMID: 33119871 DOI: 10.1007/978-3-030-50224-9_10] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The cancer tissue exists not as a single entity, but as a combination of different cellular phenotypes which, taken together, dramatically contribute to the entirety of their ecosystem, collectively termed as the tumor microenvironment (TME). The TME is composed of both immune and nonimmune cell types, stromal components, and vasculature-all of which cooperate to promote cancer progression. Not all immune cells, however, are immune-suppressive; some of them can promote the immune microenvironment to fight the invading and uncontrollably dividing cell populations at the initial stages of tumor growth. Yet, many of these processes and cellular phenotypes fall short, and the immune ecosystem more often than not ends up stabilizing in favor of the "resistant" resident cells that begin clonal expansion and may progress to metastatic forms. Stromal components, making up the extracellular matrix and basement membrane, are also not the most innocuous: CAFs embedded throughout secrete proteases that allow the onset of one of the most invasive processes-angiogenesis-through destruction of the ECM and the basement membrane. Vasculature formation, because of angiogenesis, is the largest invader of the TME and the reason metastasis happens. Vasculature is so sporadic and omnipresent in the TME that most drug therapies are mainly focused on stopping this uncontrollable process. As the tumor continues to grow, different processes are constantly supplying it with the ingredients favorable for tumor progression and eventual metastasis. For example, angiogenesis promotes blood vessel formation that will allow the bona fide escape of tumor cells to take place. Another process like hypoxia will present itself in several forms throughout the tumor (mild or acute, cycling or permanent), starting mechanisms such as epithelial to mesenchymal transitions (EMT) of resident cells and inadvertently placing the cells in such a stressful condition that production of ROS and DNA damage is unavoidable. DNA damage can induce mutagenicity while allowing resistant cells to survive. This is where drugs and treatments can subsequently suffer in effectiveness. Finally, another molecule has just surfaced as being a very important player in the TME: nitric oxide. Often overlooked and equated with ROS and initially assigned in the category of pathogenic molecules, nitric oxide can definitely do some damage by causing metabolic reprogramming and promotion of immunosuppressive phenotypes at low concentrations. However, its actions seem to be extremely dose-dependent, and this issue has become a hot target of current treatment goals. Shockingly, nitric oxide, although omnipresent in the TME, can have a positive effect on targeting the TME broadly. Thus, while the TME is a myriad of cellular phenotypes and a combination of different tumor-promoting processes, each process is interconnected into one whole: the tumor microenvironment.
Collapse
Affiliation(s)
- Anastasia Vedenko
- Department of Urology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Kush Panara
- The Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Gabriella Goldstein
- College of Health Professions and Sciences, University of Central Florida, Orlando, FL, USA
| | - Ranjith Ramasamy
- Department of Urology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Himanshu Arora
- Department of Urology, Miller School of Medicine, University of Miami, Miami, FL, USA.
- The Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
59
|
Tsai LL, Bhatt RS, Strob MF, Jegede OA, Sun MRM, Alsop DC, Catalano P, McDermott D, Robson PM, Atkins MB, Pedrosa I. Arterial Spin Labeled Perfusion MRI for the Evaluation of Response to Tyrosine Kinase Inhibition Therapy in Metastatic Renal Cell Carcinoma. Radiology 2020; 298:332-340. [PMID: 33258745 DOI: 10.1148/radiol.2020201763] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Background Tumor perfusion may inform therapeutic response and resistance in metastatic renal cell carcinoma (RCC) treated with antiangiogenic therapy. Purpose To determine if arterial spin labeled (ASL) MRI perfusion changes are associated with tumor response and disease progression in metastatic RCC treated with vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitors (TKIs). Materials and Methods In this prospective study (ClinicalTrials.gov identifier: NCT00749320), metastatic RCC perfusion was measured with ASL MRI before and during sunitinib or pazopanib therapy between October 2008 and March 2014. Objective response rate (ORR) and progression-free survival (PFS) were calculated. Perfusion was compared between responders and nonresponders at baseline, at week 2, after cycle 2 (12 weeks), after cycle 4 (24 weeks), and at disease progression and compared with the ORR by using the Wilcoxon rank sum test and with PFS by using the log-rank test. Results Seventeen participants received sunitinib (mean age, 59 years ± 7.0 [standard deviation]; 11 men); 11 participants received pazopanib (mean age, 63 years ± 6.6; eight men). Responders had higher baseline tumor perfusion than nonresponders (mean, 404 mL/100 g/min ± 213 vs 199 mL/100 g/min ± 136; P = .02). Perfusion decreased from baseline to week 2 (-53 mL/100 g/min ± 31; P < .001), after cycle 2 (-65 mL/100 g/min ± 25; P < .001), and after cycle 4 (-79 mL/100 g/min ± 15; P = .008). Interval reduction in perfusion at those three time points was not associated with ORR (P = .63, .29, and .27, respectively) or PFS (P = .28, .27, and .32). Perfusion increased from cycle 4 to disease progression (51% ± 11; P < .001). Conclusion Arterial spin labeled perfusion MRI may assist in identifying responders to vascular endothelial growth factor receptor tyrosine kinase inhibitors and may help detect early evidence of disease progression in patients with metastatic renal cell carcinoma. © RSNA, 2020 Online supplemental material is available for this article. See also the editorial by Goh and De Vita in this issue.
Collapse
Affiliation(s)
- Leo L Tsai
- From the Department of Radiology (L.L.T., M.F.S., D.C.A.) and Division of Hematology/Oncology (R.S.B., D.M.), Beth Israel Deaconess Medical Center, Boston, Mass; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Mass (O.A.J., P.C.); Department of Radiology, Lowell General Hospital, Lowell, Mass (M.R.M.S.); Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (P.M.R.); Division of Hematology/Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC (M.B.A.); and Department of Radiology, University of Texas Southwestern Medical School, 5323 Harry Hines Blvd, Dallas, TX 75390 (I.P.)
| | - Rupal S Bhatt
- From the Department of Radiology (L.L.T., M.F.S., D.C.A.) and Division of Hematology/Oncology (R.S.B., D.M.), Beth Israel Deaconess Medical Center, Boston, Mass; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Mass (O.A.J., P.C.); Department of Radiology, Lowell General Hospital, Lowell, Mass (M.R.M.S.); Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (P.M.R.); Division of Hematology/Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC (M.B.A.); and Department of Radiology, University of Texas Southwestern Medical School, 5323 Harry Hines Blvd, Dallas, TX 75390 (I.P.)
| | - Meaghan F Strob
- From the Department of Radiology (L.L.T., M.F.S., D.C.A.) and Division of Hematology/Oncology (R.S.B., D.M.), Beth Israel Deaconess Medical Center, Boston, Mass; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Mass (O.A.J., P.C.); Department of Radiology, Lowell General Hospital, Lowell, Mass (M.R.M.S.); Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (P.M.R.); Division of Hematology/Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC (M.B.A.); and Department of Radiology, University of Texas Southwestern Medical School, 5323 Harry Hines Blvd, Dallas, TX 75390 (I.P.)
| | - Opeyemi A Jegede
- From the Department of Radiology (L.L.T., M.F.S., D.C.A.) and Division of Hematology/Oncology (R.S.B., D.M.), Beth Israel Deaconess Medical Center, Boston, Mass; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Mass (O.A.J., P.C.); Department of Radiology, Lowell General Hospital, Lowell, Mass (M.R.M.S.); Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (P.M.R.); Division of Hematology/Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC (M.B.A.); and Department of Radiology, University of Texas Southwestern Medical School, 5323 Harry Hines Blvd, Dallas, TX 75390 (I.P.)
| | - Maryellen R M Sun
- From the Department of Radiology (L.L.T., M.F.S., D.C.A.) and Division of Hematology/Oncology (R.S.B., D.M.), Beth Israel Deaconess Medical Center, Boston, Mass; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Mass (O.A.J., P.C.); Department of Radiology, Lowell General Hospital, Lowell, Mass (M.R.M.S.); Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (P.M.R.); Division of Hematology/Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC (M.B.A.); and Department of Radiology, University of Texas Southwestern Medical School, 5323 Harry Hines Blvd, Dallas, TX 75390 (I.P.)
| | - David C Alsop
- From the Department of Radiology (L.L.T., M.F.S., D.C.A.) and Division of Hematology/Oncology (R.S.B., D.M.), Beth Israel Deaconess Medical Center, Boston, Mass; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Mass (O.A.J., P.C.); Department of Radiology, Lowell General Hospital, Lowell, Mass (M.R.M.S.); Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (P.M.R.); Division of Hematology/Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC (M.B.A.); and Department of Radiology, University of Texas Southwestern Medical School, 5323 Harry Hines Blvd, Dallas, TX 75390 (I.P.)
| | - Paul Catalano
- From the Department of Radiology (L.L.T., M.F.S., D.C.A.) and Division of Hematology/Oncology (R.S.B., D.M.), Beth Israel Deaconess Medical Center, Boston, Mass; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Mass (O.A.J., P.C.); Department of Radiology, Lowell General Hospital, Lowell, Mass (M.R.M.S.); Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (P.M.R.); Division of Hematology/Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC (M.B.A.); and Department of Radiology, University of Texas Southwestern Medical School, 5323 Harry Hines Blvd, Dallas, TX 75390 (I.P.)
| | - David McDermott
- From the Department of Radiology (L.L.T., M.F.S., D.C.A.) and Division of Hematology/Oncology (R.S.B., D.M.), Beth Israel Deaconess Medical Center, Boston, Mass; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Mass (O.A.J., P.C.); Department of Radiology, Lowell General Hospital, Lowell, Mass (M.R.M.S.); Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (P.M.R.); Division of Hematology/Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC (M.B.A.); and Department of Radiology, University of Texas Southwestern Medical School, 5323 Harry Hines Blvd, Dallas, TX 75390 (I.P.)
| | - Philip M Robson
- From the Department of Radiology (L.L.T., M.F.S., D.C.A.) and Division of Hematology/Oncology (R.S.B., D.M.), Beth Israel Deaconess Medical Center, Boston, Mass; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Mass (O.A.J., P.C.); Department of Radiology, Lowell General Hospital, Lowell, Mass (M.R.M.S.); Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (P.M.R.); Division of Hematology/Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC (M.B.A.); and Department of Radiology, University of Texas Southwestern Medical School, 5323 Harry Hines Blvd, Dallas, TX 75390 (I.P.)
| | - Michael B Atkins
- From the Department of Radiology (L.L.T., M.F.S., D.C.A.) and Division of Hematology/Oncology (R.S.B., D.M.), Beth Israel Deaconess Medical Center, Boston, Mass; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Mass (O.A.J., P.C.); Department of Radiology, Lowell General Hospital, Lowell, Mass (M.R.M.S.); Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (P.M.R.); Division of Hematology/Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC (M.B.A.); and Department of Radiology, University of Texas Southwestern Medical School, 5323 Harry Hines Blvd, Dallas, TX 75390 (I.P.)
| | - Ivan Pedrosa
- From the Department of Radiology (L.L.T., M.F.S., D.C.A.) and Division of Hematology/Oncology (R.S.B., D.M.), Beth Israel Deaconess Medical Center, Boston, Mass; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Mass (O.A.J., P.C.); Department of Radiology, Lowell General Hospital, Lowell, Mass (M.R.M.S.); Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (P.M.R.); Division of Hematology/Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC (M.B.A.); and Department of Radiology, University of Texas Southwestern Medical School, 5323 Harry Hines Blvd, Dallas, TX 75390 (I.P.)
| |
Collapse
|
60
|
Hemminki O, Perlis N, Bjorklund J, Finelli A, Zlotta AR, Hemminki A. Treatment of Advanced Renal Cell Carcinoma: Immunotherapies Have Demonstrated Overall Survival Benefits While Targeted Therapies Have Not. EUR UROL SUPPL 2020; 22:61-73. [PMID: 34337479 PMCID: PMC8317793 DOI: 10.1016/j.euros.2020.11.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2020] [Indexed: 12/16/2022] Open
Abstract
CONTEXT Current guidelines suggest several targeted therapies (TTs) and immunotherapies (ITs) in the treatment of advanced or metastatic renal cell carcinoma (mRCC). Ideal sequencing of these treatments is unclear. OBJECTIVE The primary objective was to evaluate the overall survival (OS) data of the treatments approved for mRCC. Secondary objectives included evaluating other signs of efficacy and adverse events. EVIDENCE ACQUISITION We reviewed the current Food and Drug Administration-approved treatments for mRCC. Trials associated with approval were reviewed. We also included pre- and postapproval publications when appropriate. EVIDENCE SYNTHESIS There is minimal evidence supporting OS benefit for the nine approved TTs. They result in adverse events and are a considerable economic burden. For these reasons, their future role in mRCC treatment should be re-evaluated, given the emergence of IT that have demonstrated OS benefits. Accumulating long-term survival data with high-dose interleukin-2 treatment suggests that this older treatment could still be considered for eligible patients. Checkpoint inhibitors have shown promising OS and durable responses; as such, the high cost of treatment might be justified. However, the available evidence does not suggest that adding TT to IT would increase efficacy over IT alone, but would add toxicity. CONCLUSIONS Trial data supporting OS benefit are much stronger for ITs than for TTs. Combining checkpoint inhibitors with TTs has not been shown to produce better OS than checkpoint inhibitors alone, while more adverse events are present. Granting drug approvals based on efficacy without demonstrated OS benefit should be revisited. PATIENT SUMMARY Approved treatments for metastatic kidney cancer include targeted and immune-based therapies. The former commonly produces temporary tumour shrinkage, but survival benefits are unclear. All approved immunotherapies have increased survival, and a proportion of patients appear cured.
Collapse
Affiliation(s)
- Otto Hemminki
- Division of Urologic Oncology, Department of Surgical Oncology, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, Ontario, Canada
- Helsinki University Hospital, Department of Urology, Helsinki, Finland
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Nathan Perlis
- Division of Urologic Oncology, Department of Surgical Oncology, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, Ontario, Canada
| | - Johan Bjorklund
- Division of Urologic Oncology, Department of Surgical Oncology, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, Ontario, Canada
- Institution for Molecular Medicine and Surgery, Urology, Karolinska Institutet, Stockholm, Sweden
| | - Antonio Finelli
- Division of Urologic Oncology, Department of Surgical Oncology, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, Ontario, Canada
| | - Alexandre R. Zlotta
- Division of Urologic Oncology, Department of Surgical Oncology, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, Ontario, Canada
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| |
Collapse
|
61
|
Campochiaro PA, Akhlaq A. Sustained suppression of VEGF for treatment of retinal/choroidal vascular diseases. Prog Retin Eye Res 2020; 83:100921. [PMID: 33248215 DOI: 10.1016/j.preteyeres.2020.100921] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/15/2020] [Accepted: 11/18/2020] [Indexed: 12/27/2022]
Abstract
Neovascular age-related macular degeneration (NVAMD) is the most prevalent choroidal vascular disease, and diabetic retinopathy (DR) and retinal vein occlusion (RVO) are the most prevalent retinal vascular diseases. In each of these, hypoxia plays a central role by stabilizing hypoxia-inducible factor-1 which increases production of vascular endothelial growth factor (VEGF) and other hypoxia-regulated gene products. High VEGF causes excessive vascular permeability, neovascularization, and in DR and RVO, promotes closure of retinal vessels exacerbating hypoxia and creating a positive feedback loop. Hence once VEGF expression is elevated it tends to remain elevated and drives disease progression. While other hypoxia-regulated gene products also contribute to pathology in these disease processes, it is remarkable how much pathology is reversed by selective inhibition of VEGF. Clinical trials have demonstrated outstanding visual outcomes in patients with NVAMD, DR, or RVO from frequent intraocular injections of VEGF-neutralizing proteins, but for a variety of reasons injection frequency has been substantially less in clinical practice and visual outcomes are disappointing. Herein we discuss the rationale, preclinical, and early clinical results of new approaches that provide sustained suppression of VEGF. These approaches will revolutionize the management of these prevalent retinal/choroidal vascular diseases.
Collapse
Affiliation(s)
- Peter A Campochiaro
- The Departments of Ophthalmology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| | - Anam Akhlaq
- The Departments of Ophthalmology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
62
|
Wood CG, Ferguson JE, Parker JS, Moore DT, Whisenant JG, Maygarden SJ, Wallen EM, Kim WY, Milowsky MI, Beckermann KE, Davis NB, Haake SM, Karam JA, Bortone DS, Vincent BG, Powles T, Rathmell WK. Neoadjuvant pazopanib and molecular analysis of tissue response in renal cell carcinoma. JCI Insight 2020; 5:132852. [PMID: 33208553 PMCID: PMC7710285 DOI: 10.1172/jci.insight.132852] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 10/08/2020] [Indexed: 01/12/2023] Open
Abstract
BACKGROUNDSurgery remains the frontline therapy for patients with localized clear cell renal cell carcinoma (ccRCC); however, 20%-40% recur. Angiogenesis inhibitors have improved survival in metastatic patients and may result in responses in the neoadjuvant setting. The impact of these agents on the tumor genetic heterogeneity or the immune milieu is largely unknown. This phase II study was designed to evaluate safety, response, and effect on tumor tissue of neoadjuvant pazopanib.METHODSccRCC patients with localized disease received pazopanib (800 mg daily; median 8 weeks), followed by nephrectomy. Five tumors were examined for mutations by whole exome sequencing from samples collected before therapy and at nephrectomy. These samples underwent RNA sequencing; 17 samples were available for posttreatment assessment.RESULTSTwenty-one patients were enrolled. The overall response rate was 8 of 21 (38%). No patients with progressive disease. At 1-year, response-free survival and overall survival was 83% and 89%, respectively. The most frequent grade 3 toxicity was hypertension (33%, 7 of 21). Sequencing revealed strong concordance between pre- and posttreatment samples within individual tumors, suggesting tumors harbor stable core profiles. However, a reduction in private mutations followed treatment, suggesting a selective process favoring enrichment of driver mutations.CONCLUSIONNeoadjuvant pazopanib is safe and active in ccRCC. Future genomic analyses may enable the segregation of driver and passenger mutations. Furthermore, tumor infiltrating immune cells persist during therapy, suggesting that pazopanib can be combined with immune checkpoint inhibitors without dampening the immune response.FUNDINGSupport was provided by Novartis and GlaxoSmithKline as part of an investigator-initiated study.
Collapse
Affiliation(s)
| | - James E. Ferguson
- Department of Urology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Joel S. Parker
- Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, USA
| | - Dominic T. Moore
- Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, USA
| | - Jennifer G. Whisenant
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Susan J. Maygarden
- Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, USA.,Department of Pathology
| | - Eric M. Wallen
- Department of Urology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.,Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, USA
| | - William Y. Kim
- Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, USA.,Department of Medicine, Division of Oncology, and
| | - Mathew I. Milowsky
- Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, USA.,Department of Medicine, Division of Oncology, and
| | - Kathryn E. Beckermann
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Nancy B. Davis
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Scott M. Haake
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jose A. Karam
- Department of Urology, MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Benjamin G. Vincent
- Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, USA.,Department of Medicine, Division of Hematology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - W. Kimryn Rathmell
- Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, USA.,Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
63
|
Larionova VB, Snegovoy AV. Possibilities of supportive therapy in patients with blood system tumors and malignant neoplasms. ONCOHEMATOLOGY 2020; 15:107-127. [DOI: 10.17650/1818-8346-2020-15-3-107-127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Finding opportunities to improve treatment outcomes of cancer patients remains a difficult and unresolved problem. Modern anticancer treatment due to the intensity and molecular biological orientation allows achieving higher efficiency and theoretically reducing the complications frequency. At the same time, the “increase in efficiency” in the modern oncology really exists, but a “decrease in the incidence of complications” is far from its solution. In many ways, the problems of diagnosis, treatment and monitoring of complications are associated with the impact on complex physiological processes in the body of an oncological patient. Timely implementation of modern and adequate programs for the prevention and treatment of these complications defines the concept of “supportive therapy”, which provides at least half of the effectiveness of anticancer treatment.The Multinational Association of Supportive Care in Cancer (MASCC) was formed in 1990. The main tasks of the association were the creation of supportive care system, its popularization and accumulation of scientific data. The MASCC was used not only oncologists experience, but also of specialists working in almost all areas of medicine. Supportive therapy provides prevention and treatment of complications from the moment of malignant disease develops, at all stages of anticancer treatment, during the rehabilitation period, and in patients in the terminal phase.An important stage in the development of maintenance care in Russia was the holding of annual conferences in Moscow with the support of MASCC. Russia is included in the European MASCC group and in working group on supportive therapy and palliative care of the Chemotherapists Society (ESMO). The Russian Society of Supportive care in Oncology (RASSC) was organized In Russia on June 1, 2017. In recent years, the main directions of supportive care have been developed in our country. Today, supportive therapy is an obligatory component of anticancer programs, which allows the patient to cope with severe but potentially reversible disorders of vital organs at all stages of treatment. This is a real way to increase the treatment efficacy and improve the quality of life of cancer patients.
Collapse
Affiliation(s)
- V. B. Larionova
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia
| | - A. V. Snegovoy
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia
| |
Collapse
|
64
|
Skeletal Muscle Loss during Multikinase Inhibitors Therapy: Molecular Pathways, Clinical Implications, and Nutritional Challenges. Nutrients 2020; 12:nu12103101. [PMID: 33053632 PMCID: PMC7601327 DOI: 10.3390/nu12103101] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/02/2020] [Accepted: 10/05/2020] [Indexed: 12/17/2022] Open
Abstract
In cancer patients, loss of muscle mass is significantly associated with low tolerability of chemotherapy and poor survival. Despite the great strides in the treatment of cancer, targeted therapies such as tyrosine kinase inhibitors (TKIs) could exacerbate muscle wasting. Over recent years, the impact of skeletal muscle loss during TKI therapy on clinical outcomes has been in the spotlight. In this review, we focus on the different molecular pathways of TKIs potentially involved in muscle wasting. Then, we report the results of the studies assessing the effects of different TKI therapies—such as sorafenib, regorafenib, sunitinib, and lenvatinib—on muscle mass, and highlight their potential clinical implications. Finally, we discuss an integrative nutritional approach to be adopted during TKI treatment. The assessment of muscle mass from computerized tomography imaging could be helpful in predicting toxicity and prognosis in patients treated with TKI such as sorafenib. Early recognition of low muscle mass and effective personalized nutritional support could prevent or attenuate muscle mass wasting. However, the role of nutrition is still overlooked, and future clinical trials are needed to find the optimal nutritional support to countermeasure muscle mass depletion during TKI therapy.
Collapse
|
65
|
Spisarová M, Melichar B, Vitásková D, Študentová H. Pembrolizumab plus axitinib combination and the paradigm change in the treatment of advanced renal cell carcinoma. Future Oncol 2020; 17:241-254. [PMID: 33016119 DOI: 10.2217/fon-2020-0079] [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] [Indexed: 01/17/2023] Open
Abstract
Sequential administration of single targeted agents has been challenged as the dominant treatment paradigm in patients with metastatic renal cell carcinoma by improved outcomes obtained with combination regimens based on immune checkpoint inhibitors. Most patients treated with sequential monotherapy eventually develop drug resistance and succumb to progressive disease, leading to the search for therapies that would overcome drug resistance and result in a more durable treatment response. Improved outcomes have been demonstrated in Phase III trials in comparison with sunitinib for the combinations of axitinib plus pembrolizumab, axitinib plus avelumab, bevacizumab plus atezolizumab and ipilimumab plus nivolumab. A statistically significant improvement of both progression-free and overall survival has been demonstrated for the axitinib plus pembrolizumab combination.
Collapse
Affiliation(s)
- Martina Spisarová
- Department of Oncology, Palacký University Medical School & Teaching Hospital, 77900 Olomouc, Czech Republic
| | - Bohuslav Melichar
- Department of Oncology, Palacký University Medical School & Teaching Hospital, 77900 Olomouc, Czech Republic.,Institute of Molecular & Translational Medicine, Palacký University Medical School Teaching Hospital, 77900 Olomouc, Czech Republic
| | - Denisa Vitásková
- Department of Oncology, Palacký University Medical School & Teaching Hospital, 77900 Olomouc, Czech Republic
| | - Hana Študentová
- Department of Oncology, Palacký University Medical School & Teaching Hospital, 77900 Olomouc, Czech Republic
| |
Collapse
|
66
|
Panian J, Lin X, Simantov R, Derweesh I, Choueiri TK, McKay RR. The Impact of Age and Gender on Outcomes of Patients With Advanced Renal Cell Carcinoma Treated With Targeted Therapy. Clin Genitourin Cancer 2020; 18:e598-e609. [PMID: 32280027 DOI: 10.1016/j.clgc.2020.03.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 03/06/2020] [Accepted: 03/10/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND A growing body of evidence suggests that age and gender play a role in cancer outcomes. The objective of this study was to investigate the effect of age and gender on survival of patients with metastatic renal cell carcinoma (RCC). METHODS We conducted a pooled analysis of patients with metastatic RCC treated on phase II and III clinical trials. Patients were stratified by age (young [<50 years], intermediate [50-70 years], versus elderly [>70 years]) and gender. Statistical analyses were performed using Cox regression adjusted for several risk factors and the Kaplan-Meier method. RESULTS We identified 4736 patients with metastatic RCC. Overall, there was no difference in overall survival (OS) when stratified by age (21.0 vs. 17.3 months for elderly vs. intermediate age groups, P = .382; 20.0 vs. 17.3 months for young vs. intermediate age groups, P = .155) or gender (19.8 vs. 19.0 for male vs. female, P = .510). Progression-free survival (PFS) was shorter in younger individuals compared with the intermediate age patients (6.0 vs. 7.1 months, P < .001), but similar across gender groups. Although all grade adverse events were more common in elderly patients (fatigue, diarrhea, decreased appetite, and weight), serious adverse events were similar between groups. CONCLUSIONS Although OS was similar between age groups, younger individuals had a shorter PFS. Gender was not an independent determinant of survival. Elderly patients experienced more adverse events than their younger counterparts. These findings are important to guide clinicians when counseling patients about expectations and toxicity associated with therapy.
Collapse
Affiliation(s)
- Justine Panian
- Department of Medicine, Division of Hematology/Oncology, University of California San Diego, Moores Cancer Center, La Jolla, CA
| | - Xun Lin
- Pfizer Oncology, New York, NY
| | | | - Ithaar Derweesh
- Department of Medicine, Division of Hematology/Oncology, University of California San Diego, Moores Cancer Center, La Jolla, CA
| | - Toni K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Rana R McKay
- Department of Medicine, Division of Hematology/Oncology, University of California San Diego, Moores Cancer Center, La Jolla, CA.
| |
Collapse
|
67
|
Xiong Z, Yuan C, shi J, Xiong W, Huang Y, Xiao W, Yang H, Chen K, Zhang X. Restoring the epigenetically silenced PCK2 suppresses renal cell carcinoma progression and increases sensitivity to sunitinib by promoting endoplasmic reticulum stress. Am J Cancer Res 2020; 10:11444-11461. [PMID: 33052225 PMCID: PMC7546001 DOI: 10.7150/thno.48469] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 09/03/2020] [Indexed: 12/16/2022] Open
Abstract
Rationale: Tumors have significant abnormalities in various biological properties. In renal cell carcinoma (RCC), metabolic abnormalities are characteristic biological dysfunction that cannot be ignored. Despite this, many aspects of this dysfunction have not been fully explained. The purpose of this study was to reveal a new mechanism of metabolic and energy-related biological abnormalities in RCC. Methods: Molecular screening and bioinformatics analysis were performed in RCC based on data from The Cancer Genome Atlas (TCGA) database. Regulated pathways were investigated by qRT-PCR, immunoblot analysis and immunohistochemistry. A series of functional analyses was performed in cell lines and xenograft models. Results: By screening the biological abnormality core dataset-mitochondria-related dataset and the metabolic abnormality core dataset-energy metabolism-related dataset in public RCC databases, PCK2 was found to be differentially expressed in RCC compared with normal tissue. Further analysis by the TCGA database showed that PCK2 was significantly downregulated in RCC and predicted a poor prognosis. Through additional studies, it was found that a low expression of PCK2 in RCC was caused by methylation of its promoter region. Restoration of PCK2 expression in RCC cells repressed tumor progression and increased their sensitivity to sunitinib. Finally, mechanistic investigations indicated that PCK2 mediated the above processes by promoting endoplasmic reticulum stress. Conclusions: Collectively, our results identify a specific mechanism by which PCK2 suppresses the progression of renal cell carcinoma (RCC) and increases sensitivity to sunitinib by promoting endoplasmic reticulum stress. This finding provides a new biomarker for RCC as well as novel targets and strategies for the treatment of RCC.
Collapse
|
68
|
Nishikawa R, Osaki M, Sasaki R, Ishikawa M, Yumioka T, Yamaguchi N, Iwamoto H, Honda M, Kabuta T, Takenaka A, Okada F. Splice variants of lysosome‑associated membrane proteins 2A and 2B are involved in sunitinib resistance in human renal cell carcinoma cells. Oncol Rep 2020; 44:1810-1820. [PMID: 32901843 PMCID: PMC7551029 DOI: 10.3892/or.2020.7752] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 07/20/2020] [Indexed: 12/13/2022] Open
Abstract
Sunitinib, a tyrosine kinase inhibitor, is among the first-line treatments for metastatic or advanced stage renal cell carcinoma (RCC). However, patients with RCC develop resistance to sunitinib. We have previously demonstrated that lysosome-associated membrane protein 2 (LAMP-2), which has three splice variants with different functions (LAMP-2A, LAMP-2B, and LAMP-2C), is involved in RCC. In the present study, we examined which splice variants of LAMP-2 contributed to sunitinib resistance in RCC cells. In vitro analysis using ACHN, human RCC cell line, revealed that the IC50 of sunitinib was significantly increased by overexpression of LAMP-2A and LAMP-2B, but not LAMP-2C (P<0.01). Kaplan-Meier survival analysis using clinical samples revealed an association between shorter survival and high expression of LAMP-2A and LAMP-2B, but not LAMP-2C, in patients with RCC treated with sunitinib (P=0.01). Furthermore, high expression of LAMP-2A and LAMP-2B in RCC revealed a weak to moderate inverse correlation with the tumor shrinkage rate and progression-free survival, respectively. Thus, high expression of LAMP-2A and LAMP-2B contributed to the acquisition of sunitinib resistance, indicating that the expression of these two variants can predict the efficacy of sunitinib treatment in patients with RCC.
Collapse
Affiliation(s)
- Ryoma Nishikawa
- Division of Urology, Faculty of Medicine, Tottori University, Yonago, Tottori 683‑8503, Japan
| | - Mitsuhiko Osaki
- Division of Experimental Pathology, Faculty of Medicine, Tottori University, Yonago, Tottori 683‑8503, Japan
| | - Ryo Sasaki
- Division of Experimental Pathology, Faculty of Medicine, Tottori University, Yonago, Tottori 683‑8503, Japan
| | - Mizuho Ishikawa
- Division of Experimental Pathology, Faculty of Medicine, Tottori University, Yonago, Tottori 683‑8503, Japan
| | - Tetsuya Yumioka
- Division of Urology, Faculty of Medicine, Tottori University, Yonago, Tottori 683‑8503, Japan
| | - Noriya Yamaguchi
- Division of Urology, Faculty of Medicine, Tottori University, Yonago, Tottori 683‑8503, Japan
| | - Hideto Iwamoto
- Division of Urology, Faculty of Medicine, Tottori University, Yonago, Tottori 683‑8503, Japan
| | - Masashi Honda
- Division of Urology, Faculty of Medicine, Tottori University, Yonago, Tottori 683‑8503, Japan
| | - Tomohiro Kabuta
- Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo 187‑8502, Japan
| | - Atsushi Takenaka
- Division of Urology, Faculty of Medicine, Tottori University, Yonago, Tottori 683‑8503, Japan
| | - Futoshi Okada
- Division of Experimental Pathology, Faculty of Medicine, Tottori University, Yonago, Tottori 683‑8503, Japan
| |
Collapse
|
69
|
Yao D, Xia S, Jin C, Zhao W, Lan W, Liu Z, Xiu Y. Feedback activation of GATA1/miR-885-5p/PLIN3 pathway decreases sunitinib sensitivity in clear cell renal cell carcinoma. Cell Cycle 2020; 19:2195-2206. [PMID: 32783497 DOI: 10.1080/15384101.2020.1801189] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Sunitinib is the most commonly used first-line therapy for the treatment of advanced renal cell carcinoma (RCC), but intrinsic and extrinsic resistance to targeted therapies dramatically compromise the benefit of clinical outcome. Dissecting the underlying mechanisms and discovering reliable predictive biomarkers are urgently needed in clinic. Here, we discovered miR-885-5p was notably decreased after sunitinib treatment and associated with poor disease progression in clear cell renal cell carcinoma (ccRCC). In vitro and in vivo studies identified miR-885-5p inhibition contributed to sunitinib resistance. Mechanistically, sunitinib treatment reduced GATA1 expression, which in turn reduced its binding to MIR885 promoter and resulted in miR-885-5p downregulation in transcriptional level. In addition, PLIN3 was confirmed to be directly targeted by miR-885-5p and its upregulation significantly increased lipid droplets formation to decrease sunitinib sensitivity. Therefore, GATA1/miR-885-5p/ PLIN3 pathway may serve as a potential therapeutic strategy and a biomarker for sunitinib treatment in ccRCC.
Collapse
Affiliation(s)
- Dayong Yao
- Department of Urology, The First Affiliated Hospital of Harbin Medical University , Harbin, China
| | - Shunyao Xia
- Department of Urology, The First Affiliated Hospital of Harbin Medical University , Harbin, China
| | - Chengjun Jin
- Department of Urology, The First Affiliated Hospital of Harbin Medical University , Harbin, China
| | - Weiming Zhao
- Department of Urology, The First Affiliated Hospital of Harbin Medical University , Harbin, China
| | - Wenjia Lan
- Central Laboratory of Hematology and Oncology, The First Affiliated Hospital of Harbin Medical University , Harbin, China
| | - Zan Liu
- Department of Urology, The First Affiliated Hospital of Harbin Medical University , Harbin, China
| | - Youcheng Xiu
- Department of Urology, The First Affiliated Hospital of Harbin Medical University , Harbin, China
| |
Collapse
|
70
|
Khan M, Zhao Z, Arooj S, Liao G. Impact of Tyrosine Kinase Inhibitors (TKIs) Combined With Radiation Therapy for the Management of Brain Metastases From Renal Cell Carcinoma. Front Oncol 2020; 10:1246. [PMID: 32793497 PMCID: PMC7390930 DOI: 10.3389/fonc.2020.01246] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 06/17/2020] [Indexed: 12/16/2022] Open
Abstract
Background: Targeted therapy has transformed the outcome for patients with metastatic renal cell carcinoma. Their efficacy and safety have also been demonstrated in brain metastatic RCC. Preclinical evidence suggests synergism of radiation and tyrosine kinase inhibitors. Consequently, several studies have compared their efficacy in the treatment of RCC brain metastases to the era of brain management with surgery/radiation only. Objectives: We seek to systematically review and meta-analyze the results of those studies that involved comparative intervention groups of brain management; TKIs, and never used TKIs. Methods and Materials: Online databases (PubMed, EMBASE, Cochrane library, and ClinicalTrials.gov) were searched for comparative studies. Overall survival as the primary outcome of interest, and local brain control, distant control, and adverse events as secondary outcomes of interest were recorded for meta-analysis. Hazard ratios were pooled together using Review Manager 5.3. Fixed effects or random effects model were adopted according to the level of heterogeneity. Subgroup analysis included studies that involved SRS as the local treatment of management. Results: Overall 7 studies (n = 897) were included for meta-analysis. TKI use was associated with better survival (HR 0.60 [0.52, 0.69], p < 0.00001) and local brain control (HR 0.34 [0.11, 0.98], p = 0.05). SRS subgroup also revealed significantly better survival (HR 0.61 [0.44, 0.83], p = 0.002) and local brain control (HR 0.19 [0.08, 0.45], p = 0.0002). Distant brain control (HR 0.95 [0.67, 1.35], p = 0.79) and brain progression free survival were unaffected (HR 0.94 [0.56, 1.56], p = 0.80). Only one study (n = 376) reported significantly greater 12-months cumulative incidence of radiation necrosis with TKI use within 30 days of SRS (10.9 vs. 6.4%, p = 0.04). Conclusions: TKIs use in combination with SRS is safe and effective for treating RCC brain metastases. Larger randomized controlled trials are warranted to validate the results.
Collapse
Affiliation(s)
- Muhammad Khan
- Department of Radiation Oncology, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China.,Department of Oncology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhihong Zhao
- Department of Nephrology, Shenzhen People's Hospital, Second Clinical Medicine Centre, Jinan University, Shenzhen, China
| | - Sumbal Arooj
- Department of Biochemistry, University of Sialkot, Sialkot, Pakistan
| | - Guixiang Liao
- Department of Radiation Oncology, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
71
|
Pauli FP, Martins JR, Paschoalin T, Ionta M, Barbosa MLC, Barreiro EJ. Novel VEGFR‐2 inhibitors with an
N
‐acylhydrazone scaffold. Arch Pharm (Weinheim) 2020; 353:e2000130. [DOI: 10.1002/ardp.202000130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Fernanda P. Pauli
- Laboratory of Evaluation and Synthesis of Bioactive Substances (LASSBio), Institute of Biomedical Sciences Federal University of Rio de Janeiro, CCS Rio de Janeiro RJ Brazil
- Graduate Program in Chemistry (PGQu) Federal University of Rio de Janeiro Rio de Janeiro Brazil
| | - Juliana R. Martins
- Department of Drugs and Medicines, Institute of Biomedical Sciences Federal University of Alfenas Alfenas Brazil
| | - Thaysa Paschoalin
- Department of Biophysics Federal University of São Paulo São Paulo Brazil
| | - Marisa Ionta
- Department of Drugs and Medicines, Institute of Biomedical Sciences Federal University of Alfenas Alfenas Brazil
| | - Maria Leticia C. Barbosa
- Laboratory of Evaluation and Synthesis of Bioactive Substances (LASSBio), Institute of Biomedical Sciences Federal University of Rio de Janeiro, CCS Rio de Janeiro RJ Brazil
- Faculty of Pharmacy Federal University of Rio de Janeiro Rio de Janeiro Brazil
| | - Eliezer J. Barreiro
- Laboratory of Evaluation and Synthesis of Bioactive Substances (LASSBio), Institute of Biomedical Sciences Federal University of Rio de Janeiro, CCS Rio de Janeiro RJ Brazil
| |
Collapse
|
72
|
Shen M, Chen G, Xie Q, Li X, Xu H, Wang H, Zhao S. Association between PD-L1 Expression and the Prognosis and Clinicopathologic Features of Renal Cell Carcinoma: A Systematic Review and Meta-Analysis. Urol Int 2020; 104:533-541. [PMID: 32623437 DOI: 10.1159/000506296] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 01/27/2020] [Indexed: 11/19/2022]
Abstract
The expression of programmed cell death-ligand 1 (PD-L1) and its correlation with the prognosis and clinicopathologic features of renal cell carcinoma (RCC) remain controversial to date. Concerning this issue, we had conducted a meta-analysis of relevant studies searched in the Web of Science, PubMed, EMBASE, and Cochrane Library databases. The Newcastle-Ottawa quality assessment scale was applied to assess the quality of the included studies. The hazard ratio (HR) and its corresponding 95% confidence intervals (CIs) were collected by Stata 12.0 and used for the results of overall survival (OS) and disease-free survival (DFS). A total of 1,644 patients in 8 studies were included in this meta-analysis. Results showed that PD-L1 expression significantly correlated with OS (HR = 1.98, 95% CI: 1.22-3.22, Z = 2.77, p = 0.006) and DFS (HR = 3.70, 95% CI: 2.07-6.62, Z = 4.40, p = 0.0001) in ccRCC. Subgroup analysis indicated that PD-L1 expression significantly correlated with the lymph-gland transfer ratio (HR = 2.45, 95% CI: 1.02-5.92, Z = 1.99, p = 0.05) and tumor necrosis (HR = 6.05, 95% CI: 3.78-9.67, Z = 7.51, p < 0.00001). This meta-analysis suggests that PD-L1 expression is a valuable prognostic tool for patients with ccRCC. Subgroup analyses demonstrated that it was helpful for screening patients with RCC who need anti-PD-1/PD-L1 treatment and support them to benefit from such immune-targeted therapy.
Collapse
Affiliation(s)
- Maolei Shen
- Department of Urology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Guang Chen
- Department of Pharmacology, School of Medicine, Taizhou University, Taizhou, China
| | - Qiang Xie
- Department of Reproduction, Southern Medical University Affiliate Dongguan People's Hospital, Dongguan, China
| | - Xin Li
- Department of Urology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Hao Xu
- Health Co., 69235 Army of PLA, Jiang Xin, China
| | - Hui Wang
- Department of Urology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Shankun Zhao
- Department of Urology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China,
| |
Collapse
|
73
|
Westerman ME, Shapiro DD, Wood CG, Karam JA. Neoadjuvant Therapy for Locally Advanced Renal Cell Carcinoma. Urol Clin North Am 2020; 47:329-343. [PMID: 32600535 DOI: 10.1016/j.ucl.2020.04.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There has been strong interest in using neoadjuvant therapy to decrease recurrence rates and facilitate surgical resection in locally advanced renal cell carcinoma. To date, no evidence exists to support improvement in oncologic outcomes with neoadjuvant therapy. Likewise, although targeted therapies have shown efficacy in tumor downsizing, this does not often translate to downstaging. Use of presurgical therapy for the purpose of downstaging inferior vena cava tumor thrombi is currently not supported. Future studies evaluating the benefit of newer immune checkpoint inhibitors will determine if there is a larger role for neoadjuvant therapy in locally advanced renal cell carcinoma.
Collapse
Affiliation(s)
- Mary E Westerman
- Department of Urology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1373, Houston, TX 77030, USA
| | - Daniel D Shapiro
- Department of Urology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1373, Houston, TX 77030, USA
| | - Christopher G Wood
- Department of Urology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1373, Houston, TX 77030, USA
| | - Jose A Karam
- Department of Urology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1373, Houston, TX 77030, USA.
| |
Collapse
|
74
|
Abstract
Patients with renal cell carcinoma may develop metastases after radical nephrectomy, and therefore monitoring with imaging for recurrent or metastatic disease is critical. Imaging varies with specific suspected site of disease. Computed tomography/MRI of the abdomen and pelvis are mainstay modalities. Osseous and central nervous system imaging is reserved for symptomatic patients. Radiologic reporting is evolving to reflect effects of systemic therapy on lesion morphology. Nuclear medicine studies compliment routine imaging as newer agents are evaluated for more accurate tumor staging. Imaging research aims to fill gaps in treatment selection and monitoring of treatment response in metastatic renal cell carcinoma.
Collapse
Affiliation(s)
- Soumya V L Vig
- Department of Radiology, NYU Langone Medical Center, New York, NY 10016, USA
| | - Elcin Zan
- Department of Radiology, NYU Langone Medical Center, New York, NY 10016, USA
| | - Stella K Kang
- Department of Radiology, NYU Langone Medical Center, New York, NY 10016, USA; Department of Population Health NYU Langone Medical Center, New York, NY 10016, USA.
| |
Collapse
|
75
|
McIntosh AG, Umbreit EC, Holland LC, Gu C, Tannir NM, Matin SF, Karam JA, Culp SH, Wood CG. Optimizing patient selection for cytoreductive nephrectomy based on outcomes in the contemporary era of systemic therapy. Cancer 2020; 126:3950-3960. [PMID: 32515845 DOI: 10.1002/cncr.32991] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 02/25/2020] [Accepted: 03/13/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND The management of metastatic renal cell carcinoma (mRCC) has evolved rapidly, and results from the Cancer du Rein Metastatique Nephrectomie et Antiangiogéniques (CARMENA) trial bring into question the utility of cytoreductive nephrectomy (CN). The objective of this study was to examine overall survival (OS) and identify risk factors associated with patients less likely to benefit from CN in the targeted therapy era. METHODS Patients with mRCC undergoing CN from 2005 to 2017 were identified. Kaplan-Meier methods and Cox proportional hazards regression analyses were used to assess OS and risk-stratify patients, respectively, on the basis of preoperative clinical and laboratory data. RESULTS Six hundred eight patients were eligible with a median follow-up of 29.4 months. Ninety-five percent of the patients had an Eastern Cooperative Oncology Group performance status less than or equal to 1, and 70% had a single site of metastatic disease. In a multivariable analysis, risk factors significantly associated with decreased OS included systemic symptoms at diagnosis, retroperitoneal and supradiaphragmatic lymphadenopathy, bone metastasis, clinical T4 disease, a hemoglobin level less than the lower limit of normal (LLN), a serum albumin level less than the LLN, a serum lactate dehydrogenase level greater than the upper limit of normal, and a neutrophil/lymphocyte ratio greater than or equal to 4. Patients were stratified into 3 risk groups: low (fewer than 2 risk factors), intermediate (2-3 risk factors), and high (more than 3 risk factors). These groups had median OS of 58.9 months (95% confidence interval [CI], 44.3-66.6 months), 30.6 months (95% CI, 27.0-35.0 months), and 19.2 months (95% CI, 13.9-22.6 months), respectively (P < .0001). The median time to postoperative systemic therapy was 45 days (interquartile range, 30-90 days). CONCLUSIONS Patients with more than 3 risk factors did not seem to benefit from CN. Importantly, OS in this group was equivalent to, if not higher than, OS for patients in the CN plus sunitinib arm of CARMENA, and this raises the possibility that a well-selected population might benefit from CN.
Collapse
Affiliation(s)
- Andrew G McIntosh
- Department of Urology, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Eric C Umbreit
- Department of Urology, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Levi C Holland
- Department of Urology, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Cindy Gu
- Department of Urology, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nizar M Tannir
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Surena F Matin
- Department of Urology, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jose A Karam
- Department of Urology, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Molecular and Translational Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Stephen H Culp
- Department of Urology, University of Virginia Health System, Charlottesville, Virginia, USA
| | - Christopher G Wood
- Department of Urology, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
76
|
Hypoxia: Turning vessels into vassals of cancer immunotolerance. Cancer Lett 2020; 487:74-84. [PMID: 32470491 DOI: 10.1016/j.canlet.2020.05.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 04/27/2020] [Accepted: 05/12/2020] [Indexed: 12/12/2022]
Abstract
Hypoxia is a universal feature of solid cancers caused by a mismatch between cellular oxygen supply and consumption. To meet the increased demand for oxygen, hypoxic cancer cells (CCs) induce a multifaceted process known as angiogenesis, wherein new vessels are formed by the sprouting of pre-existing ones. In addition to providing oxygen for growth and an exit route for dissemination, angiogenic vessels and factors are co-opted by CCs to enable the generation of an immunotolerant, hypoxic tumor microenvironment, leading to therapeutic failure and mortality. In this review, we discuss how hypoxia-inducible factors (HIFs), the mechanistic target of rapamycin (mTOR), and the unfolded protein response (UPR) control angiogenic factors serving both vascular and immunomodulatory functions in the tumor microenvironment. Possible therapeutic strategies, wherein targeting oxygen sensing might enhance anti-angiogenic and immunologically-mediated anti-cancer responses, are suggested.
Collapse
|
77
|
Moran M, Nickens D, Adcock K, Bennetts M, Desscan A, Charnley N, Fife K. Sunitinib for Metastatic Renal Cell Carcinoma: A Systematic Review and Meta-Analysis of Real-World and Clinical Trials Data. Target Oncol 2020; 14:405-416. [PMID: 31301015 PMCID: PMC6684538 DOI: 10.1007/s11523-019-00653-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Randomized controlled trials (RCTs) have stringent inclusion criteria and may not fully represent patients seen in everyday clinical practice. Real-world data (RWD) can provide supportive evidence for the effectiveness of medical interventions in more heterogeneous populations than RCTs. Sunitinib is a widely used first-line treatment for patients with metastatic renal cell carcinoma (mRCC). Objective This is the first comprehensive meta-analysis to evaluate the efficacy of sunitinib using the novel approach of combining RCTs and RWD. Methods RCTs and RWD studies published between 2000 and 2017 were identified from PubMed, Ovid, MEDLINE, and EMBASE. Eligible studies contained a cohort of ≥ 50 adult patients with mRCC receiving first-line sunitinib treatment. The meta-analysis combined RWD and RCT treatment groups, adjusting for data type (RCT or RWD). Recorded outcomes were median progression-free survival (mPFS), median overall survival (mOS), and objective response rate (ORR). Publication bias was assessed via review of funnel plots for each outcome measure. A random effects model to account for study heterogeneity was applied to each endpoint. Sensitivity analyses evaluated the robustness of the overall estimates. Results Of the 3611 studies identified through medical database searches, 22 (15 RWD studies, 7 RCTs) met eligibility criteria and were analyzed. mPFS (18 studies), mOS (19 studies), and ORR (15 studies) were reported for aggregate measures based on 4815, 5321, and 4183 patients, respectively. Reported mPFS (RWD, 7.5–11.0 months; RCTs, 5.6–15.1 months) and ORR data (RWD, 14.0–34.6%; RCTs, 18.8–46.9%) were consistent with the overall confidence estimates (95% confidence interval [CI]) of 9.3 (8.6–10.2) months and 27.9% (24.2–32.0), respectively. Reported mOS showed greater variation in RWD (6.8–33.2 months) compared with RCTs (21.8–31.5 months), with an overall confidence estimate (95% CI) of 23.0 (19.2–27.6) months. Inspection of funnel plots and sensitivity analyses indicated that there was no publication bias for any efficacy endpoint. Sensitivity analyses showed no evidence of lack of robustness for mPFS, mOS, or ORR. Interpretation of these results is limited by differences in trial design, cohort characteristics, and missing data. Conclusions This novel, comprehensive meta-analysis validates sunitinib as an effective first-line treatment for patients with mRCC in both RCTs and everyday clinical practice. The methodology provides a framework for future analyses combining data from RCTs and RWD. Electronic supplementary material The online version of this article (10.1007/s11523-019-00653-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Kate Fife
- Cambridge University Hospital, Cambridge, UK
| |
Collapse
|
78
|
Sustained treatment of retinal vascular diseases with self-aggregating sunitinib microparticles. Nat Commun 2020; 11:694. [PMID: 32019921 PMCID: PMC7000758 DOI: 10.1038/s41467-020-14340-x] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 12/27/2019] [Indexed: 11/16/2022] Open
Abstract
Neovascular age-related macular degeneration and diabetic retinopathy are prevalent causes of vision loss requiring frequent intravitreous injections of VEGF-neutralizing proteins, and under-treatment is common and problematic. Here we report incorporation of sunitinib, a tyrosine kinase inhibitor that blocks VEGF receptors, into a non-inflammatory biodegradable polymer to generate sunitinib microparticles specially formulated to self-aggregate into a depot. A single intravitreous injection of sunitinib microparticles potently suppresses choroidal neovascularization in mice for six months and in another model, blocks VEGF-induced leukostasis and retinal nonperfusion, which are associated with diabetic retinopathy progression. After intravitreous injection in rabbits, sunitinib microparticles self-aggregate into a depot that remains localized and maintains therapeutic levels of sunitinib in retinal pigmented epithelium/choroid and retina for more than six months. There is no intraocular inflammation or retinal toxicity. Intravitreous injection of sunitinib microparticles provides a promising approach to achieve sustained suppression of VEGF signaling and improve outcomes in patients with retinal vascular diseases. Neovascular age-related macular degeneration and diabetic retinopathy are currently treated with repeated intravitreous injections of VEGF neutralizing proteins. Here the authors develop a microparticle-loaded tyrosine kinase inhibitor therapy, which is effective for six months after a single injection in preclinical models.
Collapse
|
79
|
Survival benefit with extended lymphadenectomy for advanced renal malignancy: A population-based analysis. Asian J Urol 2020; 7:29-36. [PMID: 31970069 PMCID: PMC6962734 DOI: 10.1016/j.ajur.2019.06.004] [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: 06/14/2018] [Revised: 01/01/2019] [Accepted: 02/14/2019] [Indexed: 11/20/2022] Open
Abstract
Objective We used population-based data to examine the possible benefit of extended lymphadenectomy for patients with renal malignancy in the setting of more advanced disease. Methods The Surveillance, Epidemiology, and End Results (SEER) database was utilized to identify non-metastatic, T3-T4 renal cancer patients from 2004–2015 treated with removal of ≥1 lymph node at the time of nephrectomy. Non-parametric bivariate statistics were used to assess associations between covariates of interest and extended lymphadenectomy (≥10 lymph nodes removed). Cancer-specific survival (CSS) and overall survival (OS) benefit was evaluated using Kaplan–Meier analysis. Results Of the 4397 patients identified, 816 (18.6%) underwent extended lymphadenectomy. For patients with T3a disease, 5-year CSS and OS benefit with extended lymphadenectomy did not reach statistical significance (CSS: hazard ratio [HR] 0.98, 95% confidence interval [CI] 0.77–1.24; OS: HR 0.96, 95% CI 0.77–1.20). Conversely, for those with T3b-T3c disease, extended lymphadenectomy led to statistically significant improvements in both 5-year CSS and OS compared to non-extended lymphadenectomy (CSS: HR 0.78, 95% CI 0.61–0.99; OS: HR 0.72, 95% CI 0.58–0.90). Finally, for those with T4 disease, use of extended lymphadenectomy had OS benefit after 5 years (OS: HR 0.51, HR 0.29–0.90, p = 0.02). Conclusion Based on population-level data, extended lymphadenectomy was associated with improved survival in select patients with advanced renal malignancy treated with surgical nephrectomy. Understanding the basis of these real-world findings in the face of conflicting randomized trial results will be key, moving forward.
Collapse
|
80
|
Lim J, Lee A, Lee HG, Lim JS. Modulation of Immunosuppression by Oligonucleotide-Based Molecules and Small Molecules Targeting Myeloid-Derived Suppressor Cells. Biomol Ther (Seoul) 2020; 28:1-17. [PMID: 31431006 PMCID: PMC6939693 DOI: 10.4062/biomolther.2019.069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 07/02/2019] [Accepted: 07/04/2019] [Indexed: 12/12/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are immature myeloid cells that exert suppressive function on the immune response. MDSCs expand in tumor-bearing hosts or in the tumor microenvironment and suppress T cell responses via various mechanisms, whereas a reduction in their activities has been observed in autoimmune diseases or infections. It has been reported that the symptoms of various diseases, including malignant tumors, can be alleviated by targeting MDSCs. Moreover, MDSCs can contribute to patient resistance to therapy using immune checkpoint inhibitors. In line with these therapeutic approaches, diverse oligonucleotide-based molecules and small molecules have been evaluated for their therapeutic efficacy in several disease models via the modulation of MDSC activity. In the current review, MDSC-targeting oligonucleotides and small molecules are briefly summarized, and we highlight the immunomodulatory effects on MDSCs in a variety of disease models and the application of MDSC-targeting molecules for immuno-oncologic therapy.
Collapse
Affiliation(s)
- Jihyun Lim
- Department of Biological Science, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Aram Lee
- Department of Biological Science, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Hee Gu Lee
- Medical Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Jong-Seok Lim
- Department of Biological Science, Sookmyung Women's University, Seoul 04310, Republic of Korea.,Cellular Heterogeneity Research Center, Sookmyung Women's University, Seoul 04310, Republic of Korea
| |
Collapse
|
81
|
Juloori A, Miller JA, Parsai S, Kotecha R, Ahluwalia MS, Mohammadi AM, Murphy ES, Suh JH, Barnett GH, Yu JS, Vogelbaum MA, Rini B, Garcia J, Stevens GH, Angelov L, Chao ST. Overall survival and response to radiation and targeted therapies among patients with renal cell carcinoma brain metastases. J Neurosurg 2020; 132:188-196. [PMID: 30660120 DOI: 10.3171/2018.8.jns182100] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 08/14/2018] [Indexed: 11/12/2022]
Abstract
OBJECTIVE The object of this retrospective study was to investigate the impact of targeted therapies on overall survival (OS), distant intracranial failure, local failure, and radiation necrosis among patients treated with radiation therapy for renal cell carcinoma (RCC) metastases to the brain. METHODS All patients diagnosed with RCC brain metastasis (BM) between 1998 and 2015 at a single institution were included in this study. The primary outcome was OS, and secondary outcomes included local failure, distant intracranial failure, and radiation necrosis. The timing of targeted therapies was recorded. Multivariate Cox proportional-hazards regression was used to model OS, while multivariate competing-risks regression was used to model local failure, distant intracranial failure, and radiation necrosis, with death as a competing risk. RESULTS Three hundred seventy-six patients presented with 912 RCC BMs. Median OS was 9.7 months. Consistent with the previously validated diagnosis-specific graded prognostic assessment (DS-GPA) for RCC BM, Karnofsky Performance Status (KPS) and number of BMs were the only factors prognostic for OS. One hundred forty-seven patients (39%) received vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitors (TKIs). Median OS was significantly greater among patients receiving TKIs (16.8 vs 7.3 months, p < 0.001). Following multivariate analysis, KPS, number of metastases, and TKI use remained significantly associated with OS.The crude incidence of local failure was 14.9%, with a 12-month cumulative incidence of 13.4%. TKIs did not significantly decrease the 12-month cumulative incidence of local failure (11.4% vs 14.5%, p = 0.11). Following multivariate analysis, age, number of BMs, and lesion size remained associated with local failure. The 12-month cumulative incidence of radiation necrosis was 8.0%. Use of TKIs within 30 days of SRS was associated with a significantly increased 12-month cumulative incidence of radiation necrosis (10.9% vs 6.4%, p = 0.04). CONCLUSIONS Use of targeted therapies in patients with RCC BM treated with intracranial SRS was associated with improved OS. However, the use of TKIs within 30 days of SRS increases the rate of radiation necrosis without improving local control or reducing distant intracranial failure. Prospective studies are warranted to determine the optimal timing to reduce the rate of necrosis without detracting from survival.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Glen H Stevens
- 4Neurology, Neurological Institute, Cleveland Clinic, Cleveland, Ohio
| | | | | |
Collapse
|
82
|
|
83
|
Insulin-Like Growth Factor Binding Protein 5-A Probable Target of Kidney Renal Papillary Renal Cell Carcinoma. BIOMED RESEARCH INTERNATIONAL 2019; 2019:3210324. [PMID: 31886201 PMCID: PMC6925670 DOI: 10.1155/2019/3210324] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/30/2019] [Accepted: 08/06/2019] [Indexed: 12/17/2022]
Abstract
Kidney renal papillary renal cell carcinoma (KIRP) accounts for 10-15% of renal cell carcinoma (RCC). The need to find more therapeutic targets for KIRP is urgent because most targeted drugs have limited effects on advanced KIRP. Insulin-like growth factor (IGF) binding protein 5 (IGFBP5) is a secreted protein related to cell proliferation, cell adhesion, cell migration, the inflammatory response and fibrosis; these functions are independent of IGF. In our study, we determined the expression and functions of IGFBP5 with data from the database of The Cancer Genome Atlas (TCGA). We found that IGFBP5 is down regulated in KIRP kidney tissues compared to its expression in control tissues and that the expression of IGFBP5 is negatively related to patient survival. Bioinformatic analysis showed the probable processes and pathways involved in altered IGFBP5 expression, including blood vessel development, the cellular response to growth factor stimulus, the response to transforming growth factor β (TGF-β), and extracellular matrix organization. We proposed that VEGFA and TGF-β act as upstream regulatory factors of IGFBP5 and verified this in the Caki-2 cell line. Based on our results, we suggest that IGFBP5 might be a therapeutic target of KIRP.
Collapse
|
84
|
Huang H, Gao Y, Liu A, Yang X, Huang F, Xu L, Danfeng X, Chen L. EIF3D promotes sunitinib resistance of renal cell carcinoma by interacting with GRP78 and inhibiting its degradation. EBioMedicine 2019; 49:189-201. [PMID: 31669222 PMCID: PMC6945244 DOI: 10.1016/j.ebiom.2019.10.030] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/13/2019] [Accepted: 10/16/2019] [Indexed: 02/07/2023] Open
Abstract
Background Sunitinib is one of the multi-targeted tyrosine kinase inhibitors for the treatment of renal cell carcinoma (RCC) at present. However, its clinical efficacy is limited by chemoresistance of RCC. Our previous study found that eukaryotic translation initiation factor 3 subunit D (EIF3D) was an oncogene in the development and progression of RCC but little is known about whether EIF3D participated in sunitinib resistance of RCC. Methods The expression of EIF3D in the tumor tissue specimen was detected by immunohistochemistry. The effect of EIF3D on sunitinib-resistance of RCC cells was evaluated by colony formation, IC50 proliferation and in vivo tumor growth assays. The interaction between EIF3D and glucose regulated protein 78 (GRP78) was assessed by Co-IP and Western blot assays. Finding EIF3D expression was found higher in 786-OR and ACHN-R cells with acquired sunitinib resistance than that in 786-O and ACHN cells sunitinib to sensitive. The EIF3D level was also up-regulated in sunitinib-chemoresistant tumor tissues compared with chemosensitive tumor tissues. Functional study showed that EIF3D knockdown decreased cell viability with sunitinib treatment. Mechanistical study demonstrated that EIF3D interacted with GRP78 and enhanced protein stability through blocking the ubiquitin-mediated-proteasome degradation of GRP78. GRP78 overexpression induced sunitinib resistance of RCC cells by triggering the unfolded protein response, whereas GRP78 silencing inhibited cell viability. Forced expression of GRP78 eliminated the inhibitory effect of EIF3D silencing on cell growth in vitro and in vivo. Interpretation our results indicate that EIF3D played an important role in sunitinib resistance of RCC cells, suggesting that it may prove to be a potential therapeutic target for RCC.
Collapse
Affiliation(s)
- Hai Huang
- Department of Urinary Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Second Road, Shanghai, 200025, China
| | - Yi Gao
- Department of Urinary Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Second Road, Shanghai, 200025, China
| | - Ao Liu
- Department of Urinary Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Second Road, Shanghai, 200025, China
| | - Xiaoqun Yang
- Department of Pathology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Second Road, Shanghai, 200025, China
| | - Fang Huang
- Department of Urinary Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Second Road, Shanghai, 200025, China
| | - Le Xu
- Department of Urinary Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Second Road, Shanghai, 200025, China
| | - Xu Danfeng
- Department of Urinary Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Second Road, Shanghai, 200025, China.
| | - Lu Chen
- Department of Urinary Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Second Road, Shanghai, 200025, China.
| |
Collapse
|
85
|
Shen L, Li C, Zhang H, Qiu S, Fu T, Xu Y. Downregulation of miR-146a Contributes to Cardiac Dysfunction Induced by the Tyrosine Kinase Inhibitor Sunitinib. Front Pharmacol 2019; 10:914. [PMID: 31507414 PMCID: PMC6716347 DOI: 10.3389/fphar.2019.00914] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 07/19/2019] [Indexed: 12/15/2022] Open
Abstract
The main adverse effect of tyrosine kinase inhibitors, such as sunitinib, is cardiac contractile dysfunction; however, the molecular mechanisms of this effect remain largely obscure. MicroRNAs (miRNAs) are key regulatory factors in both cardiovascular diseases and the tyrosine kinase pathway. Therefore, we analyzed the differential expression of miRNAs in the myocardium in mice after exposure to sunitinib using miRNA microarray. A significant downregulation of miR-146a was observed in the myocardium of sunitinib-treated mice, along with a 20% decrease in left ventricle ejection fraction (LVEF). The downregulation of miR-146a was further validated by RT-qPCR. Among the potential targets of miR-146a, we focused on Pln and Ank2, which are closely related to cardiac contractile dysfunction. Results of luciferase reporter assay confirmed that miR-146a directly targeted the 3′ untranslated region of Pln and Ank2. Significant upregulation of PLN and ANK2 at the mRNA and protein levels was observed in the myocardium of sunitinib-treated mice. Cardiac-specific overexpression of miR-146a prevented the deteriorate effect of SNT on calcium transients, thereby alleviating the decreased contractility of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). SiRNA knockdown of PLN or ANK2 prevented sunitinib-induced suppression of contractility in hiPSC-CMs. Therefore, our in vivo and in vitro results showed that sunitinib downregulated miR-146a, which contributes to cardiac contractile dysfunction by regulating the downstream targets PLN and ANK2, and that upregulation of miR-146a alleviated the inhibitory effect of SNT on cardiac contractility. Thus, miR-146a could be a useful protective agent against sunitinib-induced cardiac dysfunction.
Collapse
Affiliation(s)
- Li Shen
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
| | - Congxin Li
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
| | - Hua Zhang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
| | - Suhua Qiu
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
| | - Tian Fu
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
| | - Yanfang Xu
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
| |
Collapse
|
86
|
London AJ, Kimmelman J. Clinical Trial Portfolios: A Critical Oversight in Human Research Ethics, Drug Regulation, and Policy. Hastings Cent Rep 2019; 49:31-41. [DOI: 10.1002/hast.1034] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
87
|
Wang YS, Shuang WB, Yin KQ, Tong XN, Xia MC, Yang HS. Analysis of the factors influencing the survival time of patients with sarcomatoid renal cell carcinoma. Mol Clin Oncol 2019; 11:405-410. [PMID: 31475069 DOI: 10.3892/mco.2019.1900] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 05/13/2019] [Indexed: 12/21/2022] Open
Abstract
The aim of the present study was to identify the factors influencing the survival time of patients with sarcomatoid renal cell carcinoma (SRCC). Between January 2000 and September 2017, a total of 21 patients were enrolled, all of whom were diagnosed with SRCC. In total, eight prognostic factors were analyzed using the Kaplan-Meier estimator, a log-rank test and Cox's proportional hazards model. The log-rank test results revealed that there was a significant association between the proportion of sarcoma elements and survival time of patients with SRCC (P<0.05). In addition, there was a significant association between post-operative drug treatment and SRCC survival time (P<0.05). The results of the Kaplan-Meier estimate demonstrated that the survival curve of post-operative drug treatment was significantly greater compared with the survival curve of patients who did not undergo drug treatment (P<0.05). The survival curve of patients with a proportion of sarcoma elements <50% was significantly greater compared with the survival curve of patients with a proportion of sarcoma elements ≥50% (P<0.05). Furthermore, the Cox's proportional hazards model revealed that the mortality risk in post-operative patients without drug treatment was 5.822 times greater compared with that of patients with drug treatment (P<0.05). Mortality risk in patients with a proportion of sarcoma elements ≥50% was 4.682 times higher compared with that of patients with sarcoma elements <50% (P<0.05). Finally, post-operative drug therapy was revealed to be a protective factor which significantly affected the survival time of patients with SRCC [risk ratio (RR)=0.172], in addition to the proportion of sarcoma elements ≥50% (RR=4.682). In conclusion, drug therapy should be promoted upon patient diagnosis with SRCC and attention should be given to the proportion of sarcomatoid components.
Collapse
Affiliation(s)
- Yu-Sheng Wang
- Department of Urology, The First Hospital of Shanxi Medical University, Yingze, Shanxi 030001, P.R. China
| | - Wei-Bing Shuang
- Department of Urology, The First Hospital of Shanxi Medical University, Yingze, Shanxi 030001, P.R. China
| | - Ke-Qiang Yin
- Department of Urology, The First Hospital of Shanxi Medical University, Yingze, Shanxi 030001, P.R. China
| | - Xu-Nan Tong
- Department of Urology, The First Hospital of Shanxi Medical University, Yingze, Shanxi 030001, P.R. China
| | - Man-Cheng Xia
- Department of Urology, The First Hospital of Shanxi Medical University, Yingze, Shanxi 030001, P.R. China
| | - Hao-Sen Yang
- Department of Urology, The First Hospital of Shanxi Medical University, Yingze, Shanxi 030001, P.R. China
| |
Collapse
|
88
|
Raczka AM, Reynolds PA. Glutaminase inhibition in renal cell carcinoma therapy. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:356-364. [PMID: 35582719 PMCID: PMC8992627 DOI: 10.20517/cdr.2018.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 04/01/2019] [Accepted: 04/08/2019] [Indexed: 11/26/2022]
Abstract
Receptor tyrosine kinase inhibitors have been a standard first-line therapy for renal cell carcinoma (RCC) for over a decade. Although they stabilize the disease, they are unable to remove all tumor cells, leading to relapse. Moreover, both intrinsic and acquired resistance to therapy are a significant health burden. In order to overcome resistance, several combination therapies have been recently approved by the FDA. Another approach takes advantage of altered metabolism in tumor cells, which switch to alternative metabolic pathways to sustain their rapid growth and proliferation. CB-839 is a small molecule inhibitor of kidney type glutaminase (GLS). GLS is often upregulated in glutamine addicted cancers, enhancing glutamine metabolism for the production of energy and the biosynthesis of various cellular building blocks. CB-839 is currently in clinical trials for several tumors, including clear cell (cc)RCC, both as monotherapy and in combination with the approved therapeutic agents everolimus, cabozantinib and nivolumab. Early results of Phase 1/2 clinical trials look promising, especially for CB-839 plus cabozantinib, and all combinations seem to be well tolerated. However, cancer cells can activate compensatory pathways to overcome glutaminolysis inhibition. Therefore, genetic and metabolomic studies are crucial for the successful implementation of CB-839 alone or in combination in subgroups of ccRCC patients.
Collapse
Affiliation(s)
- Aleksandra M. Raczka
- School of Medicine, University of St Andrews, St Andrews KY16 9TF, UK
- Biomedical Sciences Research Complex, University of St Andrews, St Andrews, UK
| | - Paul A. Reynolds
- School of Medicine, University of St Andrews, St Andrews KY16 9TF, UK
- Biomedical Sciences Research Complex, University of St Andrews, St Andrews, UK
| |
Collapse
|
89
|
Vaishampayan U. Personalized approach to systemic therapy of renal cancer. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2019. [DOI: 10.1080/23808993.2019.1612708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
90
|
Werter IM, Huijts CM, Lougheed SM, Hamberg P, Polee MB, Tascilar M, Los M, Haanen JBAG, Helgason HH, Verheul HM, de Gruijl TD, van der Vliet HJ. Metronomic cyclophosphamide attenuates mTOR-mediated expansion of regulatory T cells, but does not impact clinical outcome in patients with metastatic renal cell cancer treated with everolimus. Cancer Immunol Immunother 2019; 68:787-798. [PMID: 30756132 PMCID: PMC11028263 DOI: 10.1007/s00262-019-02313-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 02/02/2019] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Metastatic renal cell cancer (mRCC) patients have a median overall survival (mOS) of approximately 28 months. Until recently, mammalian target of rapamycin (mTOR) inhibition with everolimus was the standard second-line treatment regimen for mRCC patients, improving median progression-free survival (mPFS). Treatment with everolimus supports the expansion of immunosuppressive regulatory T cells (Tregs), which exert a negative effect on antitumor immune responses. In a phase 1 dose-escalation study, we have recently demonstrated that a low dose of 50 mg oral cyclophosphamide once daily can be safely combined with everolimus in mRCC patients and prevents the everolimus-induced increase in Tregs. MATERIALS AND METHODS In a multicenter phase 2 study, performed in patients with mRCC not amenable to or progressive on a vascular endothelial growth factor (VEGF)-receptor tyrosine kinase inhibitor (TKI) containing treatment regimen, we assessed whether the addition of this metronomic dosing schedule of cyclophosphamide to therapy with everolimus could result in an improvement of progression-free survival (PFS) after 4 months of treatment. RESULTS Though results from this study confirmed that combination treatment effectively lowered circulating levels of Tregs, addition of cyclophosphamide did not improve the PFS rate at 4 months. For this reason, the study was abrogated at the predefined interim analysis. CONCLUSION Although the comprehensive immunomonitoring analysis performed in this study provides relevant information for the design of future immunotherapeutic approaches, the addition of metronomic cyclophosphamide to mRCC patients receiving everolimus cannot be recommended.
Collapse
Affiliation(s)
- Inge M Werter
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centre, VU University Medical Centre, Vrije University, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| | - Charlotte M Huijts
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centre, VU University Medical Centre, Vrije University, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Sinéad M Lougheed
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centre, VU University Medical Centre, Vrije University, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Paul Hamberg
- Department of Medical Oncology, Franciscus Gasthuis and Vlietland, Rotterdam, The Netherlands
| | - Marco B Polee
- Department of Medical Oncology, Medical Center Leeuwarden, Leeuwarden, The Netherlands
| | - Metin Tascilar
- Department of Medical Oncology, Isala Clinics, Zwolle, The Netherlands
| | - Maartje Los
- Department of Medical Oncology, Saint Antonius Hospital, Nieuwegein, The Netherlands
| | - John B A G Haanen
- Department of Medical Oncology, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Helgi H Helgason
- Department of Medical Oncology, Haaglanden Medical Centre, The Hague, The Netherlands
| | - Henk M Verheul
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centre, VU University Medical Centre, Vrije University, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Tanja D de Gruijl
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centre, VU University Medical Centre, Vrije University, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Hans J van der Vliet
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centre, VU University Medical Centre, Vrije University, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
91
|
Should the Use of Biologic Agents in Patients With Renal and Lung Cancer Affect Our Surgical Management of Femoral Metastases? Clin Orthop Relat Res 2019; 477:707-714. [PMID: 30811363 PMCID: PMC6437361 DOI: 10.1097/corr.0000000000000434] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Biologic agents may prolong survival of patients with certain kidney and lung adenocarcinomas that have metastasized to bone, and patient response to these agents should be considered when choosing between an endoprosthesis and internal fixation for surgical treatment of femoral metastases. QUESTIONS/PURPOSES Among patients undergoing surgery for femoral metastases of lung or renal cell carcinoma, (1) Does survival differ between patients who receive only cytotoxic chemotherapy and those who either respond or do not respond to biologic therapy? (2) Does postsurgical incidence of local disease progression differ between groups stratified by systemic treatment and response? (3) Does implant survival differ among groups stratified by systemic treatment and response? METHODS From our institutional longitudinally maintained orthopaedic database, patients were identified by a query initially identifying all patients who carried a diagnosis of renal cell carcinoma or lung carcinoma. Patients who underwent internal fixation or prosthetic reconstruction between 2000 and 2016 for pathologic fracture of the femur and who survived ≥ 1 year after surgery were studied. Patients who received either traditional cytotoxic chemotherapy or a biologic agent were included. Patients were classified as responders or nonresponders to biologic agents based on whether they had clinical and imaging evidence of a response recorded on two consecutive office visits over ≥ 6 months. Endpoints were overall survival from the time of diagnosis, survival after the femoral operation, evidence of disease progression in the femoral operative site, and symptomatic local disease progression for which revision surgery was necessary. Our analysis included 148 patients with renal (n = 26) and lung (n = 122) adenocarcinoma. Fifty-one patients received traditional chemotherapy only. Of 97 patients who received a biologic agent, 41 achieved a response (stabilization/regression of visceral metastases), whereas 56 developed disease progression. We analyzed overall patient survival with the Kaplan-Meier method and used the log-rank test to identify significant differences (p < 0.05) between groups. RESULTS One-year survival after surgery among patients responsive to biologic therapy was 61% compared with 20% among patients nonresponsive to biologics (p < 0.001) and 10% among those who received chemotherapy only (p < 0.009). With the number of patients we had to study, we could not detect any difference in local progression of femoral disease associated with systemic treatment and response. Radiologic evidence of periimplant local disease progression developed in three (7%) of 41 patients who responded to biologic treatment, two (3%) of 56 patients nonresponsive to biologics, and one (2%) of 51 patients treated with traditional chemotherapy. With the numbers of patients we had, we could not detect a difference in patients who underwent revision. All three patients responsive to biologics who developed local recurrence underwent revision, whereas the two without a response to biologics did not. CONCLUSIONS Biologic therapy improves the overall longevity of some patients with lung and renal metastases to the femur in whom a visceral disease response occurred. In our limited cohort, we could not demonstrate an implant survival difference between such patients and those with shorter survival who may have had more aggressive disease. However, an increased life expectancy beyond 1 year among patients responsive to biologics may increase risk of mechanical failure of fixation constructs. LEVEL OF EVIDENCE Level III, therapeutic study.
Collapse
|
92
|
Schlemmer M, Spek A, Rodler S, Schott M, Casuscelli J, Staehler M. Sequential Treatment Based on Sunitinib and Sorafenib in Patients with Metastatic Renal Cell Carcinoma. Cureus 2019; 11:e4244. [PMID: 31131167 PMCID: PMC6516620 DOI: 10.7759/cureus.4244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Objectives: The aim of our study was to evaluate the outcome of alternative sequences of sunitinib followed by sorafenib versus sorafenib followed by sunitinib therapies in patients with metastatic renal cell carcinoma (mRCC). Materials and Methods: This single-center study analyzed patients with mRCC on systemic therapy between January 2005 and August 2011. Patients were treated with the recommended first-line therapy (sunitinib, sorafenib, pazopanib, or immunotherapy) until progression or intolerable toxicity and afterward switched to another guideline-recommended systemic therapy. Only patients starting first-line therapy on either sorafenib or sunitinib and switching to the other of these drugs were included in this analysis. Results: Out of 266 patients (females: 85, males: 181) with a median age of 57.1 years (30 - 76 years), 57 patients with a sequence of sunitinib and sorafenib were identified. First-line sorafenib therapy was followed by sunitinib (So-Su) in 32 patients; sunitinib was followed by sorafenib (Su-So) in 25 patients. Progression-free survival (PFS) for patients with first-line sorafenib was 11.6 months and was 8.7 months for sunitinib. Overall survival (OS) rates for Su-So was 118.8 months and 83.3 months with So-Su (p = 0.82). No new safety signals were detected. Conclusion: None of the therapeutic first-line approaches was superior to the other. Sequencing tyrosine kinase inhibitor (TKI) therapy seems to be effective in mRCC and superior to single-line therapy. Further studies should focus on the efficacy of single treatment lines rather than treatment sequences to estimate more potent drugs based on PFS rather than overall survival (OS).
Collapse
Affiliation(s)
- Marcus Schlemmer
- Department of Palliative Care, Barmherzige Brueder Hospital, Munich, DEU
| | - Annabel Spek
- Department of Urology, University Hospital, Ludwig Maximilian University of Munich, Munich, DEU
| | - Severin Rodler
- Department of Urology, University Hospital, Ludwig Maximilian University of Munich, Munich, DEU
| | - Melanie Schott
- Department of Urology, University Hospital, Ludwig Maximilian University of Munich, Munich, DEU
| | - Jozefina Casuscelli
- Department of Urology, University Hospital, Ludwig Maximilian University of Munich, Munich, DEU
| | - Michael Staehler
- Department of Urology, University Hospital, Ludwig Maximilian University of Munich, Munich, DEU
| |
Collapse
|
93
|
Antonaci G, Cossa LG, Muscella A, Vetrugno C, De Pascali SA, Fanizzi FP, Marsigliante S. [Pt( O,O'-acac)(γ-acac)(DMS)] Induces Autophagy in Caki-1 Renal Cancer Cells. Biomolecules 2019; 9:biom9030092. [PMID: 30845773 PMCID: PMC6468382 DOI: 10.3390/biom9030092] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 02/23/2019] [Accepted: 02/26/2019] [Indexed: 01/12/2023] Open
Abstract
We have demonstrated the cytotoxic effects of [Pt(O,O′-acac)(γ-acac)(dimethyl sulfide (DMS))] on various immortalized cell lines, in primary cultures, and in murine xenograft models in vivo. Recently, we also showed that [Pt(O,O′-acac)(γ-acac)(DMS)] is able to kill Caki-1 renal cells both in vivo and in vitro. In the present paper, apoptotic and autophagic effects of [Pt(O,O′-acac)(γ-acac)(DMS)] and cisplatin were studied and compared using Caki-1 cancerous renal cells. The effects of cisplatin include activation of caspases, proteolysis of enzyme poly ADP ribose polymerase (PARP), control of apoptosis modulators B-cell lymphoma 2 (Bcl-2), Bcl-2-associated X protein (Bax), and BH3-interacting domain death agonist (Bid), and cell cycle arrest in G2/M phase. Conversely, [Pt(O,O′-acac)(γ-acac)(DMS)] did not induce caspase activation, nor chromatin condensation or DNA fragmentation. The effects of [Pt(O,O′-acac)(γ-acac)(DMS)] include microtubule-associated proteins 1A/1B light chain 3B (LC3)-I to LC3-II conversion, Beclin-1 and Atg-3, -4, and -5 increase, Bcl-2 decrease, and monodansylcadaverine accumulation in autophagic vacuoles. [Pt(O,O′-acac)(γ-acac)(DMS)] also modulated various kinases involved in intracellular transduction regulating cell fate. [Pt(O,O′-acac)(γ-acac)(DMS)] inhibited the phosphorylation of mammalian target of rapmycin (mTOR), p70S6K, and AKT, and increased the phosphorylation of c-Jun N-terminal kinase (JNK1/2), a kinase activity pattern consistent with autophagy induction. In conclusion, while in past reports the high cytotoxicity of [Pt(O,O′-acac)(γ-acac)(DMS)] was always attributed to its ability to trigger an apoptotic process, in this paper we show that Caki-1 cells die as a result of the induction of a strong autophagic process.
Collapse
Affiliation(s)
- Giovanna Antonaci
- Laboratory of Cell Physiology, Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of Salento, 73100 Lecce, Italy.
| | - Luca Giulio Cossa
- Laboratory of Cell Physiology, Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of Salento, 73100 Lecce, Italy.
| | - Antonella Muscella
- Laboratory of Cell Pathology, Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of Salento, 73100 Lecce, Italy.
| | - Carla Vetrugno
- Laboratory of Cell Pathology, Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of Salento, 73100 Lecce, Italy.
| | - Sandra Angelica De Pascali
- Laboratory of General Inorganic Chemistry, Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of Salento, 73100 Lecce, Italy.
| | - Francesco Paolo Fanizzi
- Laboratory of General Inorganic Chemistry, Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of Salento, 73100 Lecce, Italy.
| | - Santo Marsigliante
- Laboratory of Cell Physiology, Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of Salento, 73100 Lecce, Italy.
| |
Collapse
|
94
|
Köstek O, Yılmaz E, Hacıoğlu MB, Demircan NC, Gökyer A, Uzunoğlu S, Tunçbilek N, Çiçin İ, Erdoğan B. Changes in skeletal muscle area and lean body mass during pazopanib vs sunitinib therapy for metastatic renal cancer. Cancer Chemother Pharmacol 2019; 83:735-742. [PMID: 30680522 DOI: 10.1007/s00280-019-03779-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Accepted: 01/13/2019] [Indexed: 01/06/2023]
Abstract
PURPOSE To evaluate whether sunitinib and pazopanib treatments are associated with change in skeletal muscle area (SMA) and total lean body mass (LBM) as well as to compare their efficacies and safety profiles in patients with metastatic renal cell cancer (mRCC). METHODS Thirty-six patients treated with a tyrosine kinase inhibitor were included. Eighteen of them received sunitinib and the rest/remaining received pazopanib in the first line of mRCC treatment. Baseline and follow-up computed tomography studies of the patients were performed to measure cross-sectional areas (cm2) of muscle tissues. RESULTS About 69% of patients were male and median age was 60 (49-68) years. Median time interval between two CT imagings was 6.1 (3.1-7.7) months and it was similar between the two groups (for sunitinib, 4.9 (2.5-6.9) months vs for pazopanib, 7.3 (3.2-9.5) months, p = 0.16, respectively). Disease control rate was 77.7% in all patients. Of these, 66.6% in sunitinib group was consisted of four partial responses and eight stable diseases. In addition, 88.8% in pazopanib group was consisted of three partial responses and 13 stable diseases. A significant decrease in SMA and LBM was observed after sunitinib therapy, whereas SMA and LBM values of pazopanib group did not change significantly (p = 0.02 and p = 0.70, respectively). No significant differences were observed between patients with sunitinib, and pazopanib group median PFS [11.9 (95% CI 6.1-17.6) vs 8.1 months (95% CI 7.2-9.1), respectively; p = 0.28] and median OS [28.6 (95% CI 24.3-32.9) vs 25.5 months (95% CI 18.9-52.7), respectively; p = 0.42]. Dose-limiting toxicities were significantly more frequent in sunitinib group than in pazopanib group (66.7% vs 22.2%, p = 0.02, respectively). CONCLUSIONS Loss of SMA and LBM with sunitinib was more substantial than with pazopanib. Treatment efficacies of both drugs were similar, but dose-limiting toxicity was more frequent in sunitinib group. Loss of SMA had no significant association with prognosis. Further studies are needed to clarify the possible association between SMA and prognosis in mRCC patients who receive sunitinib or pazopanib.
Collapse
Affiliation(s)
- Osman Köstek
- Division of Medical Oncology, Department of Internal Medicine, Trakya University School of Medicine, 22030, Edirne, Turkey.
| | - Erdem Yılmaz
- Department of Radiology, Trakya University School of Medicine, Edirne, Turkey
| | - Muhammet Bekir Hacıoğlu
- Division of Medical Oncology, Department of Internal Medicine, Trakya University School of Medicine, 22030, Edirne, Turkey
| | - Nazım Can Demircan
- Division of Medical Oncology, Department of Internal Medicine, Trakya University School of Medicine, 22030, Edirne, Turkey
| | - Ali Gökyer
- Division of Medical Oncology, Department of Internal Medicine, Trakya University School of Medicine, 22030, Edirne, Turkey
| | - Sernaz Uzunoğlu
- Division of Medical Oncology, Department of Internal Medicine, Trakya University School of Medicine, 22030, Edirne, Turkey
| | - Nermin Tunçbilek
- Department of Radiology, Trakya University School of Medicine, Edirne, Turkey
| | - İrfan Çiçin
- Division of Medical Oncology, Department of Internal Medicine, Trakya University School of Medicine, 22030, Edirne, Turkey
| | - Bülent Erdoğan
- Division of Medical Oncology, Department of Internal Medicine, Trakya University School of Medicine, 22030, Edirne, Turkey
| |
Collapse
|
95
|
Dosage adjustments in pivotal clinical trials with oral targeted therapies in solid tumors conducted in Europe. Eur J Clin Pharmacol 2019; 75:697-706. [PMID: 30617511 DOI: 10.1007/s00228-018-02621-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 12/26/2018] [Indexed: 02/01/2023]
Abstract
PURPOSE The aim of this study was to evaluate in what measure is dosage adjustment particularly prevalent in pivotal clinical trials of oral targeted therapy drugs approved by the European Medicine Agency as of July 31, 2018, for the treatment of solid tumors. METHODS We performed a search on the official EMA site on human medicines, using as Keyword Search the ATC Code L01X (other antineoplastic agents); from the list of drugs results, we subsequently excluded antineoplastic drugs for hematological diseases, as well as refused and withdrawn drugs. For all analyzed drugs, we recorded full dosages, dose adjustments with relative reduction percentage, reason for the adjustments, number of patients included in the trial, percentage of patients who reduced their dosage or temporarily discontinued therapy, cause of dose reduction, and presence or absence of reference to a clinical outcome in patients who reduced their dose or discontinued therapy. RESULTS We considered 74 pivotal trials on 29 target therapies, of which 56 (76%) provide information on dosage reduction, 41 (55%) on therapy suspension, and 29 (39%) on the dose taken by the sample. Trials that provide information on dosage adjustment include reduction and suspension data widely used to manage side effects; they concern, respectively, 32 and 44% of the samples considered. No trial results take account of the possible role of adjustment in clinical outcomes. CONCLUSION It would be advisable for pivotal clinical trials to give more relevance to dose management, which is a widely used tool for the management of adverse events in clinical practice. To date, such information is lacking.
Collapse
|
96
|
Chan HT, Chin YM, Low SK. The Roles of Common Variation and Somatic Mutation in Cancer Pharmacogenomics. Oncol Ther 2019; 7:1-32. [PMID: 32700193 PMCID: PMC7359987 DOI: 10.1007/s40487-018-0090-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Indexed: 02/07/2023] Open
Abstract
Cancer pharmacogenomics is the science concerned with understanding genetic alterations and its effects on the pharmacokinetics and pharmacodynamics of anti-cancer drugs, with the aim to provide cancer patients with the precise medication that will achieve a good response and cause low/no incidence of adverse events. Advances in biotechnology and bioinformatics have enabled genomic research to evolve from the evaluation of alterations at the single-gene level to studies on the whole-genome scale using large-scale genotyping and next generation sequencing techniques. International collaborative efforts have resulted in the construction of databases to curate the identified genetic alterations that are clinically significant, and these are currently utilized in clinical sequencing and liquid biopsy screening/monitoring. Furthermore, countless clinical studies have accumulated sufficient evidence to match cancer patients to therapies by utilizing the information of clinical-relevant alterations. In this review we summarize the importance of germline alterations that act as predictive biomarkers for drug-induced toxicity and drug response as well as somatic mutations in cancer cells that function as drug targets. The integration of genomics into the medical field has transformed the era of cancer therapy from one-size-fits-all to cancer precision medicine.
Collapse
Affiliation(s)
- Hiu Ting Chan
- Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yoon Ming Chin
- Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Siew-Kee Low
- Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan.
| |
Collapse
|
97
|
Ahmad S, Singh V, Sinha R, Srivastava A, Mandhani A. Role of MMP-2, MMP-9 and VEGF as serum biomarker in early prognosis of renal cell carcinoma. AFRICAN JOURNAL OF UROLOGY 2018. [DOI: 10.1016/j.afju.2018.09.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
98
|
Schuller Y, Biegstraaten M, Hollak CEM, Klümpen HJ, Gispen-de Wied CC, Stoyanova-Beninska V. Oncologic orphan drugs approved in the EU - do clinical trial data correspond with real-world effectiveness? Orphanet J Rare Dis 2018; 13:214. [PMID: 30486835 PMCID: PMC6263065 DOI: 10.1186/s13023-018-0900-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 08/29/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Evaluation of evidence for efficacy of orphan medicinal products (OMPs) for rare malignancies may be hampered by the use of tumor measurements instead of clinical endpoints. This may cause efficacy data to not always match effectiveness in the real-world. We investigated whether an efficacy-effectiveness gap exists for oncologic OMPs and aimed to identify which factors contribute to it. Also, the magnitude of the clinical efficacy of oncologic OMPs was evaluated. METHODS We included all oncologic OMPs authorized in the European Union from 2000 to 2017. Pivotal studies were evaluated by means of the European Society for Medical Oncology - Magnitude of Clinical Benefit Scale (ESMO-MCBS). To estimate real-world effectiveness, a literature search was performed to identify post-marketing studies, of which data on overall survival (OS) were extracted. OS of the new OMP was compared with OS data of standard of care. An OS gain of ≥3 months compared to pre-marketing data was considered clinically relevant. RESULTS Twenty OMPs were included, of which 5 were authorized based on OS as a primary endpoint. 10 OMPs had post-marketing data available, of which 40% did not show a clinically relevant OS gain in the real world. All OMPs that were studied with OS as primary endpoint in the pivotal study had a clinically relevant OS gain in the real world. Furthermore, all OMPs that had a high ESMO-MCBS score and post-marketing data available, resulted in a clinically relevant OS gain in the real world. CONCLUSIONS Although the sample size is small, our results indicate an efficacy-effectiveness gap for oncologic OMPs exists. Significant changes in PFS do not always lead to an increased OS. The use of PFS may be justified, but validation of surrogate endpoints is needed.
Collapse
Affiliation(s)
- Yvonne Schuller
- Department of Endocrinology and Metabolism, F5-165, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Marieke Biegstraaten
- Department of Endocrinology and Metabolism, F5-165, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Carla E. M. Hollak
- Department of Endocrinology and Metabolism, F5-165, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Heinz-Josef Klümpen
- Cancer Center Amsterdam, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | | | | |
Collapse
|
99
|
Amin A, Plimack ER, Ernstoff MS, Lewis LD, Bauer TM, McDermott DF, Carducci M, Kollmannsberger C, Rini BI, Heng DYC, Knox J, Voss MH, Spratlin J, Berghorn E, Yang L, Hammers HJ. Safety and efficacy of nivolumab in combination with sunitinib or pazopanib in advanced or metastatic renal cell carcinoma: the CheckMate 016 study. J Immunother Cancer 2018; 6:109. [PMID: 30348216 PMCID: PMC6196426 DOI: 10.1186/s40425-018-0420-0] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 09/26/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Combination treatment with immune checkpoint inhibitors and antiangiogenic drugs has shown encouraging preliminary antitumor activity across various tumor types including advanced or metastatic renal cell carcinoma (aRCC). The open-label, parallel-cohort, dose-escalation, phase I CheckMate 016 study evaluated the efficacy and safety of nivolumab in combination with antiangiogenic tyrosine kinase inhibitors or ipilimumab. Long-term outcomes from this study for the combination of nivolumab plus sunitinib or pazopanib in aRCC are presented. METHODS Patients with aRCC received nivolumab plus either sunitinib (50 mg/day, 4 weeks on/2 weeks off; N + S) or pazopanib (800 mg/day; N + P) until progression/unacceptable toxicity. The nivolumab starting dose was 2 mg/kg every 3 weeks, with planned escalation to 5 mg/kg every 3 weeks. Primary endpoints were safety and tolerability; antitumor activity was a secondary endpoint. RESULTS Arm N + S enrolled 33 patients, 19 of whom were treatment-naïve; this arm advanced to the expansion phase. Median follow-up was 50.0 months. Patients experienced high frequencies of adverse events (AEs) including treatment-related AEs (100%), grade 3/4 treatment-related AEs (82%), and treatment-related AEs leading to discontinuation (39%). Investigator-assessed objective response rate (ORR) was 55% (18/33) and median progression-free survival (PFS) was 12.7 months. Median overall survival (OS) was not reached. Arm N + P enrolled 20 patients, all had ≥1 prior systemic therapy; this arm was closed due to dose-limiting toxicities and did not proceed to expansion. Median follow-up was 27.1 months. Patients treated with N + P experienced high frequencies of AEs including treatment-related AEs (100%), grade 3/4 treatment-related AEs (70%), and treatment-related AEs leading to discontinuation (25%). Investigator-assessed ORR was 45% (9/20) and median PFS was 7.2 months. Median OS was 27.9 months. CONCLUSIONS The addition of standard doses of sunitinib or pazopanib to nivolumab resulted in a high incidence of high-grade toxicities limiting future development of either combination regimen. While there was no adverse impact on response and the OS outcome was notable, the findings suggest that the success of combination regimens based on immune checkpoint inhibitors and antiangiogenic drugs may be dependent on careful selection of the antiangiogenic component and dose. TRIAL REGISTRATION Clinicaltrials.gov identifier: NCT01472081 . Registered 16 November 2011.
Collapse
Affiliation(s)
- Asim Amin
- Immunotherapy program, Levine Cancer Institute, Carolinas HealthCare System, 1024 Morehead Medical Drive, Charlotte, NC 28204 USA
| | - Elizabeth R Plimack
- Division of Genitourinary Medical Oncology, Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA 19111 USA
| | - Marc S Ernstoff
- Division of Oncology, Department of Medicine, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14203 USA
| | - Lionel D Lewis
- Department of Medicine at The Geisel School of Medicine and The Norris Cotton Cancer Center at Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756 USA
| | - Todd M Bauer
- Sarah Cannon Research Institute/Tennessee Oncology, PLLC, Nashville, TN 37203 USA
| | - David F McDermott
- Department of Medicine, Beth Israel Deaconess Medical Center, Dana-Farber/Harvard Cancer Center, Boston, MA 02215 USA
| | - Michael Carducci
- Department of Oncology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287 USA
| | | | - Brian I Rini
- Lerner College of Medicine, Department of Hematology and Oncology, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH 44195 USA
| | - Daniel Y C Heng
- Department of Oncology, Tom Baker Cancer Center, University of Calgary, Calgary, AB T2N 4N2 Canada
| | - Jennifer Knox
- Cancer Clinical Research Unit (CCRU), Princess Margaret Cancer Centre, Toronto, ON M5G 1Z5 Canada
| | - Martin H Voss
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
| | - Jennifer Spratlin
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2 Canada
| | - Elmer Berghorn
- Oncology - Global Clinical Research, Bristol-Myers Squibb, Princeton, NJ 08541 USA
| | - Lingfeng Yang
- Oncology - Global Clinical Research, Bristol-Myers Squibb, Princeton, NJ 08541 USA
| | - Hans J Hammers
- Department of Internal Medicine, UT Southwestern – Kidney Cancer Program, Dallas, TX 75390 USA
| |
Collapse
|
100
|
Al-Daghmin A, Alhamss S, Al-Qasem K, Al-Najjar H, Al-Smadi K, Olaimat A, Al-Halbouni L. Patterns of management of translocation renal cell carcinoma. Turk J Urol 2018; 44:467-472. [PMID: 29975630 DOI: 10.5152/tud.2018.40460] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 02/26/2018] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Translocation renal cell carcinoma (TRCC) represents 1% to 5% of all cases of renal cell carcinoma (RCC), with the highest frequency among children and young adults. Management of these tumors is ill defined. We sought to characterize clinicopathological features of TRCC and patterns of medical and surgical management in a middle eastern health institute. MATERIAL AND METHODS Clinical and pathological data of 23 patients from a single institution diagnosed with TRCC between January 2005 and July 2017 were retrospectively reviewed. We dichotomized patients based on demographics, methods of surgical approach and pathologic tumor stage. We then evaluated the methods of medical management for metastatic disease and response to treatment based on cancer-specific survival (CSS) and progression-free survival (PFS). RESULTS The median age at diagnosis was 37 years. Fifteen (65%) patients were male. Most of the patients were diagnosed incidentally (65%) during abdominal imaging for other reasons. The mean tumor size was 9 cm, 47% of the patients had pathologic ≥ T3 stage. Eleven patients had lymph node dissection for clinically enlarged lymph nodes, 7 of which (64%) had lymph node metastasis. Partial nephrectomies were performed for three tumors. Eight patients had metastasis (34.7%), and 3 of them had metastasis at the time of diagnosis. Six patients received sunitinib for the treatment of metastatic disease, one patient had complete response, 4 patients had stable disease and one had disease progression. Three patients died during follow-up period because of development of metastasis at postoperative 4 (n=1), and 21 (n=1) months, and cerebral hemorrhage (n=1). The mean follow-up period was 35 months and 3-year disease-free survival was 75%. CONCLUSION TRCC is rarely seen but carries significant risk of disease progression with potential response to targeted therapy.
Collapse
Affiliation(s)
- Ali Al-Daghmin
- Department Of Surgery, King Hussein Cancer Center, Amman, Jordan
| | - Sohaib Alhamss
- Department Of Surgery, King Hussein Cancer Center, Amman, Jordan
| | - Khloud Al-Qasem
- Department Of Surgery, King Hussein Cancer Center, Amman, Jordan
| | - Hani Al-Najjar
- Department Of Surgery, King Hussein Cancer Center, Amman, Jordan
| | - Khaled Al-Smadi
- Department Of Surgery, King Hussein Cancer Center, Amman, Jordan
| | - Aseel Olaimat
- Department Of Surgery, King Hussein Cancer Center, Amman, Jordan
| | | |
Collapse
|